Skip to main content

Full text of "Review of NIAAA's neuroscience and behavioral research portfolio"

See other formats


National  Institute  on  Alcohol  Abuse  and  Alcoholism 

RESEARCH  MONOGRAPH  -  34 


Review  of  NIAAA's 

Neuroscience  and  Behavioral 

Research  Portfolio 


4 


U.S.  DEPARTMENT  OF  HEALTH  AND  HUMAN  SERVICES 

Public  Health  Service 
National  Institutes  of  Health 


NIAAA  Research  Monograph  No.  34 


REVIEW  OF  NIAAA'S 

NEUROSCIENCE 

AND  BEHAVIORAL 

RESEARCH  PORTFOLIO 


Edited  by: 

Antonio  Noronha,  Ph.D. 

Michael  Eckardt,  Ph.D. 

Kenneth  Warren,  Ph.D. 


NAT!  OMAL  I  NSTITf  ?7F5s  r 

U.S.  DEPARTMENT  OF  HEALTH  ANt)  HUMAN  SERVICES 

Public  Health  Service 

National  Institutes  of  Health 

National  Institute  on  Alcohol  Abu&e.and  Alcoholism 

6000  Executive  BouWrd    4  ZllOO 

Bethesda,  MD  20892 


Pi 


n   ■■-•? 


43 


About  the  Editors:  Antonio  Noronha,  Ph.D.,  is  chief  of  the  Neurosciences 
and  Behavioral  Research  Branch  of  the  National  Institute  on  Alcohol  Abuse 
and  Alcoholism  (NIAAA);  Michael  Eckardt,  Ph.D.,  is  senior  science  advisor  to 
the  Office  of  Scientific  Affairs,  NIAAA;  and  Kenneth  Warren,  Ph.D.,  is  director 
of  the  Office  of  Scientific  Affairs,  NIAAA. 

NIAAA  has  obtained  permission  from  the  copyright  holders  to  reproduce  fig- 
ures and  tables  throughout  this  monograph.  Further  reproduction  of  these 
materials  is  prohibited  without  specific  permission  from  the  copyright  holders. 
All  other  material  contained  in  this  monograph,  except  quoted  passages  from 
copyrighted  sources,  is  in  the  public  domain  and  may  be  reproduced  without 
permission  from  NIAAA  or  the  authors.  Citation  of  the  source  is  appreciated. 

The  U.S.  Government  does  not  endorse  or  favor  any  specific  commercial  prod- 
uct (or  commodity,  service,  or  company).  Trade  or  proprietary  names  (or  com- 
pany names)  that  appear  in  this  publication  are  used  only  because  they  are 
considered  essential  in  the  context  of  the  studies  reported  herein. 

The  opinions  expressed  herein  are  those  of  the  authors  and  do  not  necessarily 
reflect  the  official  position  of  NIAAA  or  any  other  part  of  the  National  Insti- 
tutes of  Health. 

Key  words  are  included  in  the  beginning  of  each  article.  These  descriptors  are 
drawn  from  The  Alcohol  and  Other  Drug  Thesaurus:  A  Guide  to  Concepts  and 
Terminology  in  Substance  Abuse  and  Addiction,  Second  Edition,  1995  and  may 
be  used  to  retrieve  this  monograph  in  the  Alcohol  and  Alcohol  Problems  Sci- 
ence Database  (commonly  referred  to  as  ETOH). 


NIH  Publication  No.  00-4520 
Printed  2000 


CONTENTS 


Foreword v 

Preface vii 

Abbreviations  and  Acronyms xi 

ACUTE  ETHANOL  ACTIONS  ON  SPECIFIC 
NEURAL  TARGETS 

1  Emerging  Areas  of  Research  on  Neural  Proteins  Involved  in  Acute 
Alcohol  Actions 

David  M.  Lovinger  3 

2  Lipid  Involvement  in  the  Acute  Actions  of  Alcohol  in  the  Nervous  System 
Steven  N.  Treistman 45 

3  Effects  of  Alcohol  on  the  Neuroendocrine  System 

Catherine  Rivier 61 


MOLECULAR  AND  CELLULAR  RESPONSES 
TO  CHRONIC  ETHANOL  EXPOSURE 

4  Neuroadaption  to  Ethanol  at  the  Molecular  and  Cellular  Levels 
Paula  L.  Hoffman,  A.  Leslie  Morrow,  TamaraJ.  Phillips, 

and  George  R.  Siggins    85 

5  Neurotoxicity  of  Alcohol:  Excitotoxicity,  Oxidative  Stress, 
Neurotrophic  Factors,  Apoptosis,  and  Cell  Adhesion  Molecules 

Pulton  T.  Crews  189 


ADDICTION  AND  OTHER  BEHAVIORS 
IN  ANIMAL  MODELS 

6      Basic  Behavioral  Effects  and  Underlying  Neurocircui  tries  of  Alcohol 

Kathleen  A.  Grant 209 


7  Neuroadaptive  Changes  in  Neurotransmitter  Systems  Mediating 
Ethanol-Induced  Behaviors 

Friedbert  Weiss 261 

8  Adolescent  Period:  Biological  Basis  of  Vulnerability  To  Develop 
Alcoholism  and  Other  Ethanol-Mediated  Behaviors 

Linda  Patia  Spear   315 

STUDIES  OF  ACUTE  AND  CHRONIC  EFFECTS 
OF  ALCOHOL  IN  HUMANS 

9  Acute  Effects  of  Alcohol  on  Cognition  and  Impulsive  - 
Disinhibited  Behavior 

Peter  R.  Finn   337 

10  Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcohol 
Consumption  in  Alcoholism 

John  H.  Krystal,  Ismene  L.  Petrakis,  Louis  Trevisan, 

and  Neill  Epperson 357 

1 1  The  Hypothalamic- Pituitary- Adrenal  Axis:  Changes  and  Risk 
for  Alcoholism 

Gary  Wand 397 

12  Alcohol  and  Sleep 

Cindy  L.  Ehlers 417 


STUDIES  OF  COGNITIVE/BEHAVIORAL/STRUCTURAL 
DEFICITS  IN  HUMANS 

13  Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 

Marlene  Oscar-Berman    437 

14  Human  Brain  Vulnerability  to  Alcoholism:  Evidence 
from  Neuroimaging  Studies 

Edith  V.  Sullivan  473 


15  Human  Brain  Dysfunction  Secondary  to  Alcohol  Abuse: 
Suggestions  for  New  Research  Initiatives 

George  Fein,  Daniel  Fletcher,  and  Victoria  Di  Sclafani    509 

SUBCOMMITTEE  REPORT 

16  Report  of  a  Subcommittee  of  the  National  Advisory  Council  on  Alcohol 
Abuse  and  Alcoholism  on  the  Review  of  the  Extramural  Research 
Portfolio  for  Neuroscience  and  Behavior    523 


IV 


FOREWORD 


The  National  Institute  on  Alcohol  Abuse  and  Alcoholism  (NIAAA)  is  charged 
with  the  important  mission  of  stimulating  research  on  the  causes,  consequences, 
prevention,  and  treatment  of  alcohol-related  problems.  One  aspect  of  this  mis- 
sion that  we  find  increasingly  satisfying  is  sharing  the  results  of  our  research 
efforts  with  the  scientific  community,  policymakers,  program  officials,  and  the 
general  public.  This  monograph  is  based  on  a  review  of  NIAAA's  neuroscience 
and  behavioral  research  portfolio  by  a  subcommittee  of  the  National  Advisory 
Council  on  Alcohol  Abuse  and  Alcoholism.  It  contains  reviews  of  the  breadth 
and  depth  of  our  current  neuroscience  and  behavioral  research  portfolio,  looks 
at  areas  that  are  ripe  for  research  stimulation,  and  serves  as  the  mechanism  by 
which  this  knowledge  can  be  shared  with  a  wider  audience. 

The  progress  made  in  the  neurosciences  over  the  last  two  decades  has  been 
nothing  short  of  spectacular.  NIAAA  has  taken  full  advantage  of  this  progress 
to  help  stimulate — and  provide  support  for — the  application  of  neuroscience 
techniques  to  the  study  of  alcohol  use  problems.  As  a  result,  our  understanding 
of  the  neural  processes  that  underlie  alcohol-seeking  behavior  and  of  how  alco- 
hol's actions  in  the  brain  are  related  to  the  phenomenon  of  addiction  has 
grown  dramatically.  Alcohol  neuroscience  has  led,  among  other  things,  to  the 
development  of  new  pharmacotherapies  for  alcoholism  treatment,  such  as  nal- 
trexone and  acamprosate,,  and  to  the  possibility  of  developing  "designer"  med- 
ications targeted  at  specific  alcohol  actions. 

Recently,  I  was  asked  to  predict  where  the  alcohol  research  field  would  be  by  the 
year  2020.  With  regard  to  neuroscience  research,  one  could  almost  say  the  sky's 
the  limit.  Often  prediction  turns  out  to  be  far  from  reality.  However,  based  on 
the  tremendous  progress  that  has  been  made  in  a  relatively  short  period  of  time 
in  the  neurosciences,  I  believe  that  by  2020  we  will  have  advanced  far  beyond 
our  current  grasp  of  individual  neural  connections  in  animals  and  in  humans  to 
an  understanding  of  how  circuits  in  the  brain  actually  operate  in  terms  of 
appetite,  affect,  and  cognition,  and  that  even  subjective  states,  such  as  volition 
and  consciousness,  will  yield  to  scientific  investigation.  Based  on  the  findings  of 
the  NIAAA  neuroscience  and  behavioral  research  portfolio  review,  I  am  certain 
that  we  will  be  prepared  for  the  challenges  and  opportunities  ahead. 

I  commend  the  members  of  the  Subcommittee  of  the  National  Advisory  Coun- 
cil on  Alcohol  Abuse  and  Alcoholism  on  the  Review  of  the  Extramural 


Research  Portfolio  for  Neuroscience  and  Behavior,  the  staff  of  the  NIAAA  Divi- 
sion of  Basic  Research  and  the  Neurosciences  and  Behavioral  Research  Branch, 
and  the  grantee  representatives  whose  work  is  reflected  in  this  compilation  for 
their  efforts  to  make  certain  that  NIAAA-supported  neuroscience  and  behav- 
ioral research  continues  to  represent  the  best  science  for  today  and  for  the 
future.  I  especially  commend  the  efforts  of  Dr.  Antonio  Noronha,  Chief,  Neu- 
rosciences and  Behavioral  Research  Branch,  Division  of  Basic  Research, 
NIAAA,  for  his  efforts  in  seeing  this  manuscript  to  completion. 

Enoch  Gordis,  M.D. 

Director 

National  Institute  on  Alcohol  Abuse  and  Alcoholism 


VI 


PREFACE 


The  actions  of  alcohol  that  cause  intoxication,  initiate  and  maintain  excessive 
drinking  behavior,  and  promote  relapse  during  abstinence  occur  primarily  in 
the  brain.  The  specific  mental  processes  thought  to  underlie  the  development 
of  alcoholism  involve  normal  brain  functions  such  as  learning,  attention,  emo- 
tion, and  cognition.  A  thorough  understanding  of  the  biochemical  mechanisms 
of  brain  function  and  their  response  to  alcohol  is  essential  to  develop  and 
improve  alcoholism  prevention  and  treatment  strategies.  Basic  neuroscience 
research  sponsored  by  the  National  Institute  on  Alcohol  Abuse  and  Alcoholism 
(NIAAA)  has  contributed  significantly  to  this  goal. 

NIAAA's  neuroscience  and  behavioral  research  portfolio  is  broad  and  diverse, 
reflecting  the  cooperative  efforts  of  the  Institute  and  the  alcohol  research  com- 
munity over  many  years.  Most  of  NIAAA's  research  portfolios  have  been 
recently  reviewed  by  subcommittees  of  the  National  Advisory  Council  on  Alco- 
hol Abuse  and  Alcoholism,  with  the  goal  of  evaluating  the  appropriateness, 
breadth,  coverage,  and  balance  of  each  portfolio  and  identifying  areas  that 
require  greater  attention.  Subcommittees  were  also  asked  to  provide  specific 
advice  and  guidance  on  the  scope  and  direction  of  the  Institute's  extramural 
research  activities. 

The  Subcommittee  for  the  Review  of  the  Extramural  Research  Portfolio  for 
Neuroscience  and  Behavior  consisted  of  an  advisory  group  of  individuals  with 
demonstrated  expertise  in  both  alcohol-and  non-alcohol-related  areas.  The 
review  process  was  initiated  by  having  experts  in  alcohol  neurosciences  and 
behavior  prepare  written  assessments  of  the  state  of  knowledge,  gaps  in  knowl- 
edge, and  research  opportunities  in  specific  areas.  NIAAA  program  staff 
presented  the  current  extramural  portfolio,  categorized  into  the  areas  of  basic 
neuroscience  and  behavioral  research,  and  also  included  training  and  career 
development  activities.  All  information  was  shared  with  experts,  selected 
NIAAA  staff,  and  the  subcommittee  before  the  meeting.  The  proceedings  and 
recommendations  of  the  subcommittee  were  conveyed  to  and  endorsed  by  the 
National  Advisory  Council.  NIAAA  has  begun  to  implement  these  recommen- 
dations. We  hope  that  this  monograph  will  serve  as  a  valuable  reference  as  well 
as  a  guide  to  future  research  questions. 

The  total  scope  of  NIAAA's  neuroscience  research  program  extends  beyond 
the  topics  covered  in  this  monograph.  For  example,  studies  on  the  neuroscience 


and  behavioral  aspects  of  fetal  alcohol  syndrome  (FAS),  neurogenetics,  and 
medications  development  were  reviewed  as  part  of  the  FAS,  genetics,  and 
prevention  and  treatment  research  portfolios,  respectively.  The  research 
covered  in  this  monograph  ranges  from  molecular  aspects  of  neuronal  commu- 
nication to  the  integrated  activity  of  multiple  brain  regions.  Brief  descriptions  of 
each  chapter  are  presented  below,  grouped  by  theme  rather  than  in  the  order  in 
which  they  appear.  These  themes  include  neurobiological  mechanisms  of 
alcoholism  development,  alcohol's  effects  on  brain  function,  and  factors  that 
influence  vulnerability  to  alcohol's  effects. 


MECHANISMS  OF  ADDICTION 

A  key  concept  in  alcoholism  research  is  neuroadaptation,  the  development 
of  persistent  alterations  in  brain  function  at  the  molecular,  cellular,  and  systems 
level  in  response  to  chronic  alcohol  exposure.  Chapter  4  explains  how 
neuroadaptation  associated  with  alcohol  dependence  can  induce  craving  and 
relapse  in  response  to  alcohol-related  cues  or  stress  even  after  long  periods 
of  abstinence. 

Alcohol  exposure  can  alter  the  structure  and  function  of  both  the  lipid  and 
protein  constituents  of  neuronal  membranes.  Chapter  1  discusses  alcohol's 
effects  on  proteins  at  the  molecular  level  and  their  influence  on  the  overall 
physiology  of  specific  brain  regions.  Chapter  2  emphasizes  the  importance 
of  lipid-protein  interactions  in  assessing  alcohol's  effects  on  the  function  of 
ion  channels. 

Interactions  among  multiple  neurotransmitter  systems  help  mediate  both  the 
acute  reinforcing  actions  of  alcohol  and  the  persistent  neuroadaptive  changes 
that  may  motivate  relapse.  Key  neurotransmitters  involved  in  these  processes 
include  dopamine,  serotonin,  and  gamma- aminobutyric  acid.  These  findings 
are  based  on  studies  using  laboratory  animals  (chapter  7)  as  well  as  clinical 
research  on  humans  (chapter  10). 

A  knowledge  of  the  integrated  activity  of  neural  circuits  is  required  to 
provide  the  link  between  molecular  events  and  behavior.  Chapter  6  explores 
mechanisms  by  which  ethanoPs  effects  on  cognitive  processes  such  as  learning 


vui 


and  memory  can  lead  to  reinforcement.  Exposure  to  stress  may  affect  the 
development  of  dependence  and  may  help  trigger  relapse  following  recovery. 
According  to  chapter  11,  alcohol  consumption  can  potentially  induce 
both  underactivity  and  overactivity  of  the  body's  primary  stress  response  system 
(i.e.,  the  hypothalamic-pituitary- adrenal  [HPA]  axis). 


ALCOHOL'S  EFFECTS  ON  BRAIN  FUNCTION 

Alcohol-induced  alteration  of  brain  function  can  itself  influence  alcohol  con- 
sumption patterns.  Alcohol's  effects  on  hormonal  balance  are  discussed  in  chap- 
ter 3.  For  example,  in  addition  to  inhibiting  the  release  of  reproductive 
hormones,  alcohol  administration  leads  to  increased  synthesis  of  a  key  compo- 
nent of  the  HPA  axis,  leading  to  dysregulation  of  the  stress  response.  Chapter  9 
reviews  alcohol-induced  impairment  of  cognition  and  impulse  control,  which 
can  lead  to  aggressive  behavior  and  decreased  caution  in  decisionmaking.  The 
prefrontal  cortex,  one  of  the  brain  regions  most  closely  associated  with  higher 
cognitive  functions,  such  as  decisionmaking,  is  particularly  vulnerable  to  alco- 
hol-induced dysfunction.  As  discussed  in  chapter  5,  this  dysfunction  may  be 
attributable  to  cell  death  caused  by  various  metabolic  mechanisms.  Finally, 
chapter  12  illustrates  that  both  acute  and  chronic  alcohol  consumption  alter  the 
normal  sleeping  pattern,  potentially  influencing  other  body  functions. 


VULNERABILITY  TO  ALCOHOL'S  EFFECTS 

Vulnerability  to  the  harmful  consequences  of  chronic  alcohol  consumption  is 
affected  by  factors  such  as  aging,  nutrition,  and  gender,  as  discussed  in  chapter 
13.  Adolescence  may  be  a  time  of  enhanced  vulnerability.  Research  presented  in 
chapter  8  suggests  that  adolescents  may  develop  tolerance  to  alcohol's  sedative 
effects  and  its  effects  on  coordination  more  readily  than  do  adults,  perhaps  con- 
tributing to  greater  levels  of  alcohol  use  later  in  life.  Chapter  14  describes  inno- 
vative neuroimaging  techniques  that  may  help  identify  structural  and  functional 
brain  abnormalities  associated  with  increased  vulnerability  to  alcohol's  psycho- 
logical and  behavioral  effects.  Finally,  chapter  15  suggests  multidisciplinary 
approaches  to  investigating  some  of  the  most  important  questions  regarding 
alcohol-related  brain  dysfunction. 


IX 


ACKNOWLEDGMENTS 

The  contributors  to  this  monograph  are  recognized  experts  in  their  respective 
disciplines.  Their  time  and  effort  in  preparing  their  presentations  for  the  review 
and  for  these  monograph  chapters  are  truly  appreciated.  We  thank  the  program 
staff  of  the  Neurosciences  and  Behavioral  Research  Branch,  especially  Drs.  Wal- 
ter Hunt,  Robert  Karp,  Yuan  Liu,  and  Ellen  Witt.  Their  efforts,  together  with 
those  of  the  advisory  group,  were  largely  responsible  for  the  success  of  the 
review  and  the  ensuing  recommendations.  We  also  wish  to  thank  Diana 
O'Donovan  of  the  Scientific  Communication  Branch,  Office  of  Scientific 
Affairs,  NIAAA,  and  Dianne  Welsh  and  her  staff  at  CSR,  Incorporated,  includ- 
ing John  Doria  and  Pat  Freedman,  for  their  valued  efforts  in  completing  this 
monograph. 


Antonio  Noronha,  Ph.D. 

Chief 

Neurosciences  and  Behavioral  Research  Branch 

Division  of  Basic  Research 

National  Institute  on  Alcohol  Abuse  and  Alcoholism 

Michael  Eckardt,  Ph.D. 

Senior  Science  Advisor 

Office  of  Scientific  Affairs 

National  Institute  on  Alcohol  Abuse  and  Alcoholism 

Kenneth  Warren,  Ph.D. 

Director 

Office  of  Scientific  Affairs 

National  Institute  on  Alcohol  Abuse  and  Alcoholism 


ABBREVIATIONS  AND  ACRONYMS 


A 

angstrom(s) 

AC 

adenylyl  cyclase 

ACh 

acetylcholine 

ACTH 

adrenocorticotropic  hormone  (Chapters  3,  8,  12,  16)  or 

adrenocorticotropin  (Chapter  11) 

AD 

Alzheimer's  disease 

ADH 

alcohol  dehydrogenase 

AIDS 

acquired  immunodeficiency  syndrome 

AMPA 

L-a-amino-3-hydroxy-5-methyl-4-isoxazole  propionate  (Chapter 

4)  or  a-amino-3-hydroxy-5-methyl-4-isoxazole  propionic  acid 

(Chapter  6) 

AOD 

alcohol  and  other  drug 

ATP 

adenosine  triphosphate 

AVP 

arginine  vasopressin 

BAL 

blood  alcohol  level 

BDNF 

brain-derived  neurotrophic  factor 

BOLD 

blood  oxygen  level  dependent 

BZD 

benzodiazepine 

cAMP 

cyclic  adenosine  monophosphate 

CCK 

cholecystokinin 

cDNA 

complementary  deoxyribonucleic  acid 

CeA 

central  nucleus  of  the  amygdala 

Cho 

choline 

CIE 

chronic  intermittent  ethanol 

CNS 

central  nervous  system 

CPT 

continuous  performance  task 

Cr 

creatine 

CRE 

3',5'-cyclic  adenosine  monophosphate  response  element 

CREB 

3',5'-cyclic  adenosine  monophosphate  response  element  binding 

CRP 

corticotropin-releasing  factor 

CRH 

corticotropin-releasing  hormone 

CSF 

cerebrospinal  fluid 

CT 

computed  tomography 

d 

day 

DA 

dopamine 

DHEA 

dehydroepiandrosterone 

DHT 

dihydrotestosterone 

XI 


dL 

deciliter(s) 

DNA 

deoxyribonucleic  acid 

DSM-IV 

Diagnostic  and  Statistical  Manual  of  Mental  Disorders,  4th  edition 

DTs 

delirium  tremens 

DZ 

dizygotic 

EC50 

median  effective  concentration 

ECF 

executive  cognitive  function 

ED50 

median  effective  dose 

EDE 

ethanol  delayed  effect 

EEG 

electroencephalographic 

EPR 

electron  paramagnetic  resonance 

EPSP 

excitatory  postsynaptic  potential 

ERP 

event-related  potential 

ES 

embryonic  stem  [cells] 

FAS 

fetal  alcohol  syndrome 

FDG 

fluorodeoxyglucose  (Chapter  6)  or  18fluorine- labeled 

deoxy glucose  (Chapter  14) 

FHP 

family  history  positive 

FHN 

family  history  negative 

fMRI 

functional  magnetic  resonance  imaging 

FRET 

fluorescence  resonance  energy  transfer 

FY97 

fiscal  year  1997 

g 

grams(s) 

GABA 

gamma-  aminobutyric  acid 

GABAA 

gamma-aminobutyric  acid  type  A 

GC 

glucocorticoids 

GHB 

y-hydroxybutyrate 

GnRH 

gonadotropin-releasing  hormone 

GRF 

growth  hormone-releasing  factor 

h 

hour(s) 

5-HIAA 

5-hydroxyindoleacetic  acid 

HIV 

human  immunodeficiency  virus 

HPA 

hypothalamic-pituitary- adrenal 

HPG 

hypothalamic-pituitary-gonadal 

HPS 

hypothalamic-pituitary-somatotropic 

5-HT 

5  -  hy droxytryptamine  ( serotonin ) 

Hz 

hertz 

Xll 


ICSS 

ntracranial  self- stimulation 

icv 

ntracerebroventricularly 

IEG 

mmediate  early  gene 

IGF-1 

msulin-like  growth  factor- 1 

IL-lp 

nterleukin-ip 

123jMp 

odoamphetamine  123 

ip 

ntraperitoneal 

IPSP 

nhibitory  postsynaptic  potential 

IQ 

ntelligence  quotient 

ISI 

nterstimulus  interval 

kd 

tilodalton(s) 

kg 

tilogram(s) 

KS 

Korsakoff  s  syndrome 

LD50 

median  lethal  dose 

LH 

uteinizing  hormone 

LS 

ong  sleep  [mice] 

LTD 

ong-term  depression 

LTP 

ong-term  potentiation 

MAP 

mitogen-activated  protein  [kinase] 

mCPP 

w-chlorophenylpiperazine 

MDMA 

3,4-methylenedioxymethamphetamine 

mg 

milligram(s) 

mg% 

milligrams  percent 

mL 

milliliter(s) 

mm 

millimeter(s) 

mM 

millimolar 

MRI 

magnetic  resonance  imaging 

mRNA 

messenger  ribonucleic  acid 

MRS 

magnetic  resonance  spectroscopy 

MRSI 

magnetic  resonance  spectroscopic  imag 

MT 

magnetization  transfer 

^g 

microgram(s) 

uL 

microliters ) 

[xm 

micrometer(s) 

MZ 

monozygotic 

Nac 

N-  acetyl  compounds 

nACh 

nicotinic  acetylcholine 

xm 


nAChR  nicotinic  acetylcholine  receptor 

NCAM  nerve  cell  adhesion  molecule 

NF-kB  nuclear  regulatory  factor-KB 

NGF  nerve  growth  factor 

NIAAA  National  Institute  on  Alcohol  Abuse  and  Alcoholism 

NIH  National  Institutes  of  Health 

NK  natural  killer  [cells] 

NMDA  N-methyl-D-aspartate 

NMR  nuclear  magnetic  resonance 

NO  nitric  oxide 

6-OHDA  6-hydroxydopamine 

8-OH-DPAT  8-hydroxy-2-(di-»-propylamino)tetralin 

PCP  phencyclidine 

PCR  polymerase  chain  reaction 

PE  phosphatidylethanolamine 

PEA  prenatal  exposure  to  alcohol 

PET  positron  emission  tomography 

PFC  prefrontal  cortex 

PKA  protein  kinase  A 

PKC  protein  kinase  C 

POMC  pro-opiomelanocortin 

PS  phosphatidylserine 

PTZ  pentylenetetrazol 

PVN  paraventricular  nucleus 

QTL  quantitative  trait  loci 

REM  rapid  eye  movement 

RIP  rapid  information  processing 

RNA  ribonucleic  acid 

RT  reaction  time 

RT-PCR  reverse  transcriptase-polymerase  chain  reaction  (Chapter  4)  or 

reverse  transcription-polymerase  chain  reaction  (Chapter  16) 

SCAM  substituted  cysteine  scanning  mutagenesis  [technique] 

SPECT  single  photon  emission  computed  tomography 

SS  short  sleep  [mice] 

T  testosterone 

TE  echo-time 

TFMPP  m-  trifluoromethylphenylpiperazine 


xiv 


THDOC  3a,5a-tetrahydrodeoxycorticosterone 

THIP  4,5,6,7-tetrahydroisoxazolo(5,4-c)pyridin-3-ol 

THP  tetrahydroprogesterone 

THDOC  tetrahydrodeoxycorticosterone 

VF  visual  field 

VGCC  voltage-gated  calcium  channel 

VTA  ventral  tegmental  area 

v/v  volume  to  volume  [ratio] 

WAIS-R  Wechsler  Adult  Intelligence  Scale — Revised 

WSP  withdrawal  seizure  prone 

WSR  withdrawal  seizure  resistant 

w/v  weight  per  volume 


xv 


ACUTE  ETHANOL 

ACTIONS  ON  SPECIFIC 

NEURAL  TARGETS 


Chapter  1 

Emerging  Areas  of  Research  on  Neural 
Proteins  Involved  in  Acute  Alcohol  Actions 

David  M.  Lovinger,  Ph.D. 


KEY  WORDS:  acute  AODE  (effects  ofAOD  [alcohol  or  other  drug]  use,  abuse, 
and  dependence);  proteins;  neurotransmission;  drug  binding;  molecular  struc- 
ture; spectroscopy;  brain  function;  research  and  evaluation  method;  AOD  use  be- 
havior; AOD  sensitivity; gene  expression;  animal  model;  literature  review 


Characterizing  the  primary  sites  of 
alcohol  action  and  the  mechanisms  of 
alcohol  effects  at  these  sites  is  an 
important  goal  of  basic  alcohol 
research.  The  understanding  gained 
from  this  research  will  lead  to  the 
development  of  pharmacological  and, 
perhaps,  genetic  treatments  aimed  at 
reducing  alcohol  abuse,  alcoholism, 
and  alcohol-related  brain  damage. 
This  research  should  make  it  possible 
to  target  therapeutic  approaches  not 
only  to  specific  molecules  within  the 
brain,  but  also  to  specific  molecular 
substructures  within  these  molecules.  In 
addition,  with  a  better  understanding 
of  alcohol  effects  in  brain  regions  that 
play  important  roles  in  alcohol-related 


behaviors,  appropriate  therapeutic 
treatments  can  be  focused  on  these 
brain  regions. 

This  chapter  begins  with  a  discus- 
sion of  research  examining  effects  of 
alcohol  at  the  molecular  level,  fol- 
lowed by  a  discussion  of  research 
examining  roles  of  different  molecules 
in  alcohol  effects  in  more  intact  sys- 
tems, up  to  and  including  the  intact 
organism.  The  chapter  outlines  exper- 
imental approaches  to  the  examina- 
tion of  acute  effects  of  alcohol  that  are 
likely  to  be  at  the  heart  of  the  most 
productive  research  in  this  area.  Some 
of  these  approaches  are  already  being 
applied  to  studies  of  acute  alcohol 
actions,  while  others  are  novel  and 


D.M.  Lovinger,  Ph.D.,  is  a  professor  in  the  Department  of  Molecular  Physiology  and  Biophysics  and  the 
Department  of  Pharmacology,  Vanderbilt  University  Medical  School,  702  Light  Hall,  Nashville,  TN 
37232-0615. 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


powerful  techniques  that  are  just 
emerging  in  the  field  of  neuroscience  as 
a  whole  and  should  be  applied  to 
alcohol  research  within  the  next  5  to  10 
years.  Each  section  in  the  chapter 
begins  with  a  statement  of  the  goals 
of  a  particular  research  area,  followed  by 
a  discussion  of  approaches  to  achieving 
these  goals  and,  finally,  a  brief  discus- 
sion of  the  significance  of  the  research 
to  understanding  and  treatment  of 
alcohol  abuse  and  alcoholism.  The 
chapter  concludes  with  a  discussion  of 
techniques  for  discovery  of  novel  targets 
of  alcohol  actions  and  a  special  note 
on  the  use  of  mouse  genetic  models 
in  future  alcohol  research. 

STRUCTURAL  BIOLOGICAL 
ANALYSIS  OF  ALCOHOL 
TARGETS 

Goals 

There  are  two  major  goals  of  this  line 
of  research.  The  first  goal  is  to 
describe  alcohol-induced  alterations 
in  the  molecular  structure  and 
dynamics  of  proteins  that  are  sensitive 
to  alcohol  and  are  believed  to  partici- 
pate in  the  neural  effects  of  acute 
alcohol.  This  type  of  information  will 
follow  from  studies  of  the  structures 
of  the  proteins  themselves.  Once 
some  information  about  protein 
structure  and  dynamics  has  been 
obtained,  then  analysis  of  alcohol 
effects  can  be  undertaken.  The  sec- 
ond goal  is  to  define  the  molecular 
structure  of  potential  sites  of  alcohol 
interactions  (alcohol  binding  sites?) 
within  these  proteins.  This  analysis 
will  take  the  field  to  the  next  stage  in 


understanding  the  molecular  basis  of 
alcohol  actions  by  allowing  investiga- 
tors to  truly  understand  the  sites  of 
alcohol  action  and  the  molecular 
changes  that  take  place  upon  acute 
exposure  to  alcohol. 

A  number  of  sophisticated  tech- 
niques for  examination  of  protein 
structure  have  been  devised  within  the 
last  25  years.  It  is  now  possible  to  gain 
information  about  protein  secondary 
and  tertiary  structure  as  well  as  pro- 
tein dynamics  using  an  array  of  bio- 
physical techniques.  These  techniques 
are  applicable  not  only  to  cytosolic 
proteins,  but  also  to  membrane  pro- 
teins. Several  strategies  to  employ 
these  structural  biology  techniques  in 
alcohol  research  are  described  in  the 
following  section.  This  discussion  will 
concentrate  mainly  on  techniques  that 
can  provide  information  about  struc- 
ture at  several  levels  (i.e.,  primary,  sec- 
ondary, and  tertiary  structure). 
Biochemical  techniques  that  provide 
information  about  molecular  topology 
and  solvent  exposure  of  amino  acid 
residues  (e.g.,  protease-based  assays, 
in  situ  phosphorylation,  and  antibody- 
based  assays)  will  not  be  reviewed  in 
this  chapter,  but  they  are  important 
additional  approaches  to  understand- 
ing protein  structure. 

Approaches 

The  most  definitive  information 
about  protein  structure  can  be 
obtained  from  x-ray  crystallography. 
Exact  molecular  coordinates  of  each 
amino  acid  residue  can  be  firmly 
established  with  this  approach. 
Furthermore,  crystallization  of  pro- 
teins with  small  interacting  molecules 


Neural  Proteins 


can  sometimes  be  achieved,  providing 
information  about  the  sites  of  interac- 
tion between  a  small  molecule  and 
the  protein  of  interest.  Thus,  the 
structure  of  a  protein  could  be  exam- 
ined in  the  presence  and  absence  of 
alcohol,  and  alcohol  binding  sites  on 
the  protein  could  be  localized  and 
understood  in  detail.  This  analysis  has 
been  performed  for  some  proteins, 
such  as  alcohol  dehydrogenase 
(Cedergren-Zeppezauer  et  al.  1982; 
Eklund  et  al.  1982).  Two  conditions 
must  be  met  in  order  to  apply  this 
technique:  (1)  purified  protein  must 
be  available  in  sufficient  quantity  to 
allow  for  growth  of  crystals,  and  (2) 
crystallization  must  be  feasible.  It  has 
proved  difficult  to  crystallize  integral 
membrane  proteins,  because  high 
concentrations  of  lipid  or  detergent 
are  needed  to  maintain  the  structure 
of  these  proteins.  Thus,  this  tech- 
nique has  proved  useful  mainly  for 
determining  the  structure  of  more 
hydrophilic  cytosolic  or  nuclear  pro- 
teins. However,  reports  of  crystalliza- 
tion of  Escherichia  coli  porins  (Cowan 
et  al.  1992)  and  a  more  recent  study 
of  a  prokaryotic  potassium  channel 
indicate  that  the  technology  necessary 
to  examine  some  membrane  proteins 
is  rapidly  evolving  (Doyle  et  al. 
1998).  In  addition,  it  may  be  possible 
to  obtain  crystals  of  the  non-mem- 
brane-spanning portions  of  some 
integral  membrane  proteins  (e.g.,  the 
ligand  binding  domains  of  ligand- 
gated  ion  channels).  However,  it  is 
not  clear  that  the  soluble  parts  of  the 
proteins  retain  their  native  configura- 
tion in  the  absence  of  the  membrane- 
spanning  domains.  It  must  also  be 


kept  in  mind  that  a  crystallized  pro- 
tein is  not  a  functional  protein.  X-ray 
crystallography  can  only  provide 
information  about  the  static  structure 
of  a  protein  in  one  particular  configu- 
ration. Information  about  protein 
dynamics  (e.g.,  during  acute  alcohol 
exposure)  must  be  obtained  with 
other  approaches. 

Techniques  that  can  be  used  to 
gain  structural  information  from 
intact  proteins  in  a  more  native  envi- 
ronment include  nuclear  magnetic 
resonance  (NMR)  spectroscopy,  elec- 
tron paramagnetic  resonance  (EPR) 
spectroscopy,  and  fluorescence  spec- 
troscopy. These  techniques  can  be 
applied  to  integral  membrane  proteins 
as  well  as  other  proteins. 

Nuclear  magnetic  resonance  spec- 
troscopy can  provide  information 
about  both  the  static  and  dynamic 
structures  of  proteins.  By  examining 
the  behavior  of  atomic  nuclei  under  a 
magnetic  field,  it  is  possible  to  gain 
information  about  the  relative  loca- 
tion and  motion  of  particular  amino 
acid  residues.  This  technique  has  the 
advantage  that  proteins  can  be  exam- 
ined in  solutions  or  lipid  environ- 
ments that  more  closely  mimic  the 
physiological  situation  than  a  crys- 
talline array.  Like  crystallography, 
NMR  can  potentially  provide  infor- 
mation about  the  entire  structure  of 
the  protein  at  one  time.  When  exam- 
ining small-  to  medium-sized  cytoso- 
lic proteins  (up  to  35  kd)  or  small 
stretches  of  membrane  proteins,  solu- 
tion NMR  can  be  used  to  provide  the 
entire  protein  structure  at  one  time. 
However,  limitations  on  this  tech- 
nique prevent  investigators  from 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


examining  particularly  large  proteins; 
these  limitations  mostly  result  from 
die  slower  motion  of  these  proteins, 
which  leads  to  broadening  of  spectral 
features.  Examination  of  integral 
membrane  protein  structure  with 
NMR  is  even  more  limited  (Opella 
1994,  1997).  One  can  use  solid-state 
NMR  to  examine  proteins  in  a  phos- 
pholipid environment,  but  the 
motion  of  these  proteins  is  limited, 
and  thus  limited  information  about 
the  movement  of  particular  amino 
acids  can  be  obtained.  This  technique 
can  be  combined  with  multidimen- 
sional solution  NMR  of  proteins  in 
nonpolar  solvent  mixtures,  but  the 
slow  motions  in  these  solutions  do 
not  lead  to  the  sorts  of  optimal  spec- 
tra that  one  obtains  from  more 
hydrophilic  proteins  in  a  polar  solvent 
environment.  The  purity  and  identity 
of  the  detergents  used  in  micelle  for- 
mation is  also  a  major  consideration 
in  multidimensional  solution  NMR 
studies.  Despite  these  limitations, 
investigators  have  been  able  to  deter- 
mine the  structures  of  small  mem- 
brane-spanning peptides  such  as  the 
M2  domain  of  the  nicotinic  acetyl- 
choline (ACh)  receptor  (Bechinger  et 
al.  1991)  and  to  gain  some  structural 
information  about  the  G  protein- 
coupled  family  of  receptors  by  exami- 
nation of  bacteriorhodopsin  (Keniry 
et  al.  1984;  Sobol  et  al.  1992). 

An  additional  limitation  of  this 
technique  is  the  large  amount  of  pure 
protein  needed  to  obtain  a  reasonable 
NMR  spectrum  (up  to  milligrams, 
rather  than  micrograms,  of  protein  are 
needed  in  some  cases).  The  clearest 
spectral  features  are  often  obtained 


with  isotopically  labeled  proteins,  and 
thus  it  is  best  to  be  able  to  incorpo- 
rate 15N  or  another  isotope  into  the 
protein  prior  to  purification.  This 
problem  may  be  solved  with  the  use 
of  techniques  that  allow  for  protein 
overexpression  in  cells  that  can  be 
grown  in  large  quantities.  Two  systems 
that  are  useful  in  this  regard  are  the 
baculovirus/SF9  insect  cell  system  (as 
in  Green  et  al.  1995)  and  the  Semliki 
Forest  Virus/BHK  cell  system  (as  in 
Hovius  et  al.  1998).  Both  of  these 
techniques  allow  for  infection  of  a  cell 
leading  to  overexpression  of  the 
desired  protein  in  cells  that  can  be 
grown  in  large-volume  suspension 
cultures.  Given  a  suitable  protein  sol- 
ubilization and  purification  strategy,  a 
large  amount  of  relatively  pure  protein 
can  be  obtained  from  such  cells. 
However,  it  is  still  a  daunting  task  to 
produce  sufficient  quantities  of  pro- 
tein for  NMR  analysis. 

The  theoretical  basis  of  EPR  is  sim- 
ilar to  that  of  NMR  with  the  exception 
that  spin  of  unpaired  electrons,  rather 
than  protons,  is  measured  in  a  mag- 
netic field.  The  unpaired  electrons  are 
part  of  a  "spin-labeled"  functional 
group  that  can  be  attached  to  a  mole- 
cule that  interacts  with  or  is  part  of  a 
protein.  Hubbell  and  colleagues  have 
pioneered  the  use  of  "site-directed 
spin-labeling"  in  which  the  spin  label  is 
covalently  attached  to  a  cysteine 
residue  within  a  protein  (Hubbell  and 
Altenbach  1994).  This  technique 
allows  the  investigator  to  gain  informa- 
tion about  the  molecular  environment 
of  a  particular  amino  acid  residue 
within  a  protein  as  well  as  information 
about  distances  between  different 


Neural  Proteins 


amino  acid  residues.  The  information 
can  be  used  to  gain  some  idea  about 
the  secondary  and  tertiary  structure  of 
a  protein.  This  technique  can  be 
applied  to  proteins  in  solution  or  in  a 
lipid  environment  such  that  functional 
proteins  can  be  analyzed.  This  allows 
the  investigator  to  examine  dynamic 
changes  in  protein  structure  when  a 
particular  protein  is  undergoing  a 
conformational  change,  such  as  upon 
activation  by  a  ligand  or  in  the  pres- 
ence of  alcohol.  For  example,  this 
technique  has  been  used  to  measure 
the  structural  changes  in  rhodopsin 
upon  exposure  to  light  (Altenbach  et 
al.  1996).  Application  of  this  tech- 
nique requires  a  lesser  amount  of 
purified  protein  than  is  required  for 
NMR  or  crystallization  (microgram 
quantities  are  needed  for  EPR  analysis 
of  most  proteins).  However,  examina- 
tion of  several  labeled  amino  acids  is 
rather  time-consuming.  Furthermore, 
the  mutations  needed  to  insert  the 
cysteine  residues  that  are  labeled  may 
disrupt  protein  function.  Still,  this  is  a 
promising  method  for  examining 
membrane  protein  structure  and 
dynamic  effects  of  alcohol. 

Several  techniques  have  been 
developed  for  measurement  of  protein 
structure  and  structural  dynamics 
using  fluorescence  spectroscopy. 
These  techniques  range  from  the  use 
of  intrinsically  fluorescent  amino  acids 
such  as  tryptophan  to  reactions  with 
fluorescently  labeled  ligands.  Amino 
acids  such  as  cysteine  can  also  be 
modified  to  incorporate  a  fluorescent 
label  (Wu  and  Kaback  1994;  Stratikos 
and  Gettins  1997;  Sahoo  et  al.  1998). 
Fluorescence  spectroscopy,  like  EPR, 


has  often  been  applied  to  functional 
proteins  in  a  more  or  less  native  envi- 
ronment, and  thus  information  about 
conformational  changes  in  relation  to 
protein  function  can  be  obtained 
using  fluorescence  techniques.  Fluo- 
rescence spectroscopy  can  have  quite 
favorable  signal/noise  characteristics 
with  the  proper  fluorophore,  and  this 
means  that  the  amounts  of  protein 
needed  to  apply  these  techniques  can 
often  be  smaller  than  those  needed  for 
EPR  analysis.  Through  the  use  of  time- 
resolved  laser  fluorescence  spectro- 
scopy, information  about  molecular 
conformational  changes  can  be 
acquired  in  the  picosecond  to  second 
temporal  domains  (Millar  1996; 
Beechem  1997).  This  technique 
allows  for  observation  of  very  fast 
molecular  transitions  within  a  protein. 
With  the  use  of  two  fluorophores  that 
can  emit  and  absorb  photons  in  an 
interactive  manner,  one  can  apply  flu- 
orescence resonance  energy  transfer 
(FRET)  techniques  to  determine  dis- 
tances between  fluorophores  (dos 
Remedios  and  Moens  1995).  This 
technique  can  be  used  to  gain  informa- 
tion about  molecular  distances  within 
a  protein.  Fluorescence  spectroscopy  has 
drawbacks  similar  to  EPR,  since  gener- 
ally only  one  or  two  fluorescent  groups 
can  be  examined  at  one  time.  However, 
investigators  using  fluorescence-based 
techniques  are  making  increasingly 
important  contributions  to  our  under- 
standing of  biomolecular  structure, 
and  these  techniques  should  be  quite 
useful  in  studying  alcohol  effects  on 
protein  structure  and  dynamics. 

Information  about  protein  structure 
can  also  be  obtained  using  photoaffinity 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


labeling  techniques.  If  a  suitable  probe 
molecule  with  a  photolabile  functional 
group  can  be  produced,  then  interac- 
tions between  this  molecule  and  spe- 
cific amino  acids  or  groups  of  amino 
acids  can  be  examined.  Using  photo- 
affinity  probes  that  act  via  different 
pathways  of  access  to  the  protein  (e.g., 
hydrophobic  access  vs.  access  through 
the  open  channel),  it  is  possible  to 
obtain  information  about  the  relative 
positions  of  different  residues  on  a 
membrane-bound  protein  such  as  an 
ion  channel  or  transporter.  Photo- 
affinity  techniques  have  already  been 
applied  to  examination  of  the  structure 
of  membrane  proteins  such  as  the 
nicotinic  ACh  receptor  (Galzi  et  al. 
1990;  Blanton  and  Cohen  1994; 
Czajkowski  and  Karlin  1995;  Hucho 
et  al.  1996).  Photoaffinity  labeling 
and  subsequent  determination  of  sites 
of  labeling  does  not  require  a  great 
deal  of  protein,  and  the  protein  does 
not  necessarily  have  to  be  completely 
pure  if  the  label  has  sufficiently  high 
affinity;  thus,  this  technique  is  applica- 
ble to  many  purified  and  even  partially 
purified  proteins.  Examination  of 
alcohol-induced  alterations  in  photo- 
affinity  labeling  may  help  to  elucidate 
structural  changes  in  proteins  in  the 
presence  of  alcohol. 

Another  approach  that  uses  muta- 
genesis techniques  to  gain  information 
about  protein  structure  is  the  substi- 
tuted cysteine  scanning  mutagenesis 
(SCAM)  technique.  In  this  approach, 
cysteines  are  inserted  into  different 
amino  acid  positions  within  a  protein, 
and  effects  of  modification  by  thiol 
compounds  on  protein  function  are 
examined.  Thiol-modifying  agents 


with  different  relative  hydro-  and 
lipophilicities  can  be  used  to  determine 
the  molecular  mode  of  access  to  dif- 
ferent sites  of  cysteine  modification 
within  the  protein.  This  technique  has 
been  used  to  gain  information  about 
which  residues  are  exposed  to  the  ion- 
conducting  pore  within  ion  channels 
(Akabas  et  al.  1994,  1995).  The  tech- 
nique has  several  advantages  over  the 
techniques  discussed  previously,  since 
it  does  not  require  protein  purifica- 
tion. Functional  studies  can  be  per- 
formed on  proteins  expressed  at  the 
sorts  of  levels  usually  achieved  with 
standard  heterologous  expression  sys- 
tems. Furthermore,  the  technique 
involves  assaying  protein  function  in 
ways  that  are  normally  used  by  investi- 
gators to  examine  their  proteins  of 
interest.  The  major  drawbacks  of  the 
approach  include  (1)  the  possibility 
that  cysteine  substitution  may  greatly 
alter  protein  structure  or  function  and 
(2)  direct  effects  of  the  thiol  reagents 
on  the  unmodified  protein  that  may 
preclude  the  use  of  this  technique.  It 
may  be  possible  to  use  this  technique 
to  examine  alcohol  effects  on  accessi- 
bility of  particular  amino  acid  residues 
within  proteins. 

All  of  the  data  pertaining  to  protein 
structure  and  structural  dynamics  are 
difficult  to  comprehend  without 
proper  models  of  protein  structure  and 
possible  conformational  changes  within 
proteins.  This  type  of  modeling  can  be 
achieved  using  many  molecular  mod- 
eling programs,  such  as  INSIGHT. 
Using  software  that  allows  molecular 
dynamics  simulations  can  greatly  aid 
investigators  in  understanding  changes 
in  molecular  structure  and  can  help 


Neural  Proteins 


describe  conformational  changes  in 
proteins.  Hypotheses  about  the  effects 
of  particular  alterations  in  a  protein 
and  molecular  interactions  can  also  be 
generated  using  these  approaches. 
This  sort  of  molecular  modeling  will 
also  be  an  integral  part  of  structural 
biological  analysis  of  alcohol  targets. 

The  most  likely  impact  of  this 
research  will  be  on  the  development 
of  pharmacotherapeutic  approaches  to 
the  treatment  of  alcohol  abuse  and 
alcoholism.  A  more  exact  definition  of 
the  sites  of  action  and  allosteric  effects 
of  alcohol  at  the  molecular  level  will 
aid  in  application  of  rational  drug 
design  to  the  development  of  such 
pharmacotherapies.  If  particular  alco- 
hol binding  sites  can  be  identified,  it 
may  lead  to  development  of  drugs 
that  have  very  specific  actions  at  dif- 
ferent molecular  sites  of  alcohol  inter- 
action. This  could  lead  to  selective 
alterations  of  some,  but  not  all,  of  the 
effects  of  alcohol,  while  minimizing 
the  side  effects  of  the  treatment. 

MOLECULAR  BIOLOGICAL 
ANALYSIS  OF  ALCOHOL 
TARGETS 

Goals 

After  discovery  of  protein  targets  of 
acute  alcohol  actions,  it  becomes  pos- 
sible to  examine  in  more  detail  the 
molecular  basis  of  alcohol  actions  on 
these  proteins.  This  analysis  should 
proceed  at  several  levels.  Many  of  the 
holoproteins  that  are  alcohol  sensitive 
are  made  up  of  multiple  subunits. 
Examination  of  the  relationship 
between  the  subunit  composition  of 
these  holoproteins  and  their  alcohol 


sensitivity  is  thus  an  important  area  for 
future  research.  In-depth  analysis  of 
the  secondary  structure  of  proteins  has 
revealed  several  different  domain  struc- 
tures or  local  regions  that  can  exist 
within  a  protein.  This  domain  structure 
can  be  important  in  forming  hydro- 
phobic pockets  that  could  be  sites  of 
direct  alcohol-protein  interactions. 
Thus,  analysis  of  the  relationship 
between  alcohol  sensitivity  and  protein 
domain  structure  will  make  up  an 
important  facet  of  future  research 
designed  to  characterize  the  molecu- 
lar sites  of  alcohol  actions.  Of  course, 
all  of  these  structural  properties  of 
proteins  are  conferred  by  the  amino 
acid  sequence,  or  primary  structure, 
of  the  protein.  A  complete  under- 
standing of  the  molecular  makeup  of 
an  alcohol-sensitive  site  on  a  protein 
will  require  an  understanding  of  the 
role  of  individual  amino  acids  in 
determining  protein  domain  structure 
and  in  conferring  alcohol  sensitivity. 
Individual  amino  acids  can  also  be  sites 
for  posttranslational  protein  modifica- 
tion, and  analysis  of  this  aspect  of  the 
relationship  between  primary  struc- 
ture and  alcohol  sensitivity  will  also  be 
a  part  of  future  alcohol  research. 

Protein-protein  interactions  are 
emerging  as  a  major  theme  in  molecular 
biology  and  will  no  doubt  be  important 
determinants  of  the  effects  of  alcohol 
on  molecules  within  a  given  neuron. 
Interactions  between  two  proteins  can 
determine  subcellular  localization  of  a 
given  protein  and  can  affect  protein 
function  and  enzyme-substrate  inter- 
actions. Recent  studies  have  provided 
an  enormous  body  of  information  on 
interactions  between  proteins  that  can 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


be  explored  in  relation  to  alcohol 
actions.  Some  of  the  questions  that 
need  to  be  addressed  are  as  follows: 
Does  alcohol  alter  subcellular  localiza- 
tion of  proteins  by  altering  protein- 
protein  interactions?  Does  alcohol 
alter  function  of  particular  proteins  via 
indirect  effects  on  partner  proteins? 
Does  the  alcohol  sensitivity  of  a  single 
molecule  differ  in  different  subcellular 
regions  due  to  differences  in  protein- 
protein  interactions?  Acute  alcohol 
actions  on  some  target  proteins  are 
known  to  vary  depending  on  the  cellular 
and  experimental  context  in  which  the 
alcohol  effects  are  examined.  Some  of 
this  variability  might  arise  from  differ- 
ent interactions  of  these  targets  with 
other  proteins  in  different  cellular 
contexts  or  under  different  experi- 
mental conditions.  These  possibilities 
need  to  be  explored  to  fully  under- 
stand the  molecular  determinants  of 
acute  alcohol  actions. 

Approaches 

Molecular  biological  analysis  of  alcohol- 
sensitive  proteins  has  already  been 
undertaken  in  several  laboratories. 
Two  approaches  that  have  proved 
quite  powerful  are  the  use  of  chimeric 
and  point-mutated  receptors  to  exam- 
ine the  relationship  between  receptor 
secondary/primary  structure  and  alco- 
hol sensitivity.  Experiments  examining 
both  voltage-gated  and  ligand-gated 
ion  channels  have  revealed  particular 
regions  of  channels  that  confer  alcohol 
sensitivity,  or  in  some  cases  account 
for  small  differences  in  alcohol  sensi- 
tivity (Covarrubias  et  al.  1995;  Mascia 
et  al.  1996;  Yu  et  al.  1996;  Mihic  et  al. 
1997).  The  construction  of  chimeric 


proteins  is  a  good  starting  point  for 
this  sort  of  analysis.  Using  recombi- 
nant DNA  technology,  researchers 
can  create  proteins  in  which  large  or 
small  stretches  of  amino  acids  from 
one  protein  are  combined  with  those 
from  another  protein  (Mihic  et  al. 
1997).  If  the  two  proteins  differ  in 
some  important  characteristic,  such  as 
alcohol  sensitivity,  this  approach  can 
help  the  investigator  to  determine  the 
importance  of  protein  domains  in 
conferring  this  characteristic,  and  it 
can  also  help  to  pinpoint  regions  of  the 
protein  where  amino  acids  reside  that 
have  important  roles  in  the  function 
of  interest.  Amino  acids  that  differ 
within  the  important  regions  of  the 
two  proteins  can  then  be  altered  by 
point  mutation,  and  the  effects  of  these 
alterations  on  protein  function  and 
alcohol  sensitivity  can  be  examined. 
Single  amino  acids  that  play  important 
roles  in  the  action  of  alcohol  can  be 
identified  in  this  way.  By  examining 
mutations  of  a  single  site  to  several 
different  amino  acids,  one  can  begin  to 
appreciate  what  molecular  attributes  are 
needed  at  that  site  (e.g.,  side  chain  bulk, 
hydrophobicity)  for  alcohol  sensitivity. 
For  particular  alcohol  targets  it  may 
not  be  possible  to  use  the  chimeric 
protein  approach,  since  a  highly 
homologous  protein  that  differs 
greatly  in  alcohol  sensitivity  has  not 
been  identified.  In  this  case,  the  muta- 
genesis approach  should  be  consid- 
ered, but  it  must  proceed  in  a  manner 
that  is  as  logical  as  possible  based  on 
existing  information.  For  example,  we 
know  that  most  molecular  target  sites 
for  alcohol  actions  are  hydrophobic  in 
character.  Thus,  it  would  make  sense 


10 


Neural  Proteins 


to  target  mutations  to  hydrophobic 
regions  of  proteins.  Investigators  can 
also  use  information  from  studies  of 
structurally  related  proteins,  if  available, 
to  help  pinpoint  regions  conferring 
alcohol  sensitivity.  With  the  advent  of 
software  for  prediction  of  protein  sec- 
ondary structure  based  on  primary 
sequence  information  and  analysis  of 
related  proteins,  it  may  become  possi- 
ble for  investigators  to  compare  the 
structure  of  an  alcohol-sensitive  pro- 
tein with  that  of  a  protein  that  has 
already  been  characterized  with 
respect  to  molecular  determinants  of 
alcohol  sensitivity.  This  might  allow 
investigators  to  identify  "alcohol- 
responsive"  domains  within  proteins 
and  proceed  to  examine  effects  of  muta- 
genesis in  these  domains  on  alcohol 
sensitivity.  More  will  be  said  about 
this  possible  approach  later  in  this 
chapter  (see  the  section  Searching  for 
Molecular  Targets  With  High  Sensi- 
tivity to  Acute  Alcohol). 

The  chimera/mutagenesis  approach 
can  also  help  to  localize  sites  of  poten- 
tial alcohol-protein  interactions  that 
can  be  examined  more  closely  with 
direct  physical  measurements.  This 
molecular  biological  approach  is 
already  being  applied  to  alcohol 
research  in  several  laboratories,  and  it 
should  continue  to  be  a  major  focus 
of  research.  In  particular,  extending 
this  analysis  to  newly  identified  alcohol- 
sensitive  proteins  should  be  a  priority 
for  the  coming  years. 

In  light  of  newly  reported  evidence 
for  roles  of  protein  phosphorylation  in 
modulating  alcohol  sensitivity  of  par- 
ticular proteins,  it  will  be  important  to 
use  site-directed  mutagenesis  to  alter 


potential  sites  of  protein  phosphoryla- 
tion on  alcohol  target  molecules  (as  in 
Coultrap  and  Machu  1997).  This  will 
allow  investigators  to  determine  if 
phosphorylation  at  a  particular  site  on 
a  protein  plays  a  role  in  determining 
alcohol  effects  on  the  function  of  that 
protein.  Mutations  can  be  made  that 
prevent  and/or  mimic  the  addition  of 
a  phosphate  group  to  a  particular 
amino  acid  residue,  thus  allowing 
investigators  to  look  at  both  gain  and 
loss  of  function.  This  technique  may 
also  lead  investigators  to  examine 
alcohol  effects  on  the  activity  of  pro- 
tein kinases  that  phosphorylate  impor- 
tant amino  acid  residues,  since  these 
enzymes  may  be  actual  primary  targets 
of  alcohol  actions.  The  effects  of 
mutagenesis  on  phosphorylation  of 
the  substrate  protein  can  be  directly 
assessed,  and  this  sort  of  analysis  will 
be  facilitated  by  development  of  anti- 
bodies to  target  proteins  that  can  be 
used  to  identify  particular  phosphory- 
lated  proteins.  This  line  of  research 
should  benefit  greatly  from  the  recent 
initiative  to  produce  antibodies  spe- 
cific for  alcohol  target  proteins. 

Use  of  genetically  engineered  mice 
is  also  likely  to  play  a  big  role  in  the 
future  of  molecular  analysis  of  alcohol 
effects  on  target  proteins.  Homologous 
recombination  procedures  are  now  in 
use  that  make  it  possible  to  create  mice 
in  which  point  mutations  have  been 
introduced  in  one  key  protein  (Askew 
et  al.  1993;  MacMillan  et  al.  1996).  If 
successful,  the  protein  is  expressed  as  in 
the  wild-type  animal  but  contains  the 
point  mutation.  This  approach  has  the 
potential  to  allow  an  investigator  to 
examine  the  importance  of  a  particular 


11 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


amino  acid  in  a  particular  protein 
within  the  context  of  a  whole  animal 
or  an  individual  neuron.  For  studies  of 
acute  neural  actions  of  alcohol,  these 
animals  can  be  examined  for  alcohol- 
related  behavior.  In  addition,  more 
reduced  neuronal  preparations  can  be 
used  to  examine  alcohol  effects  on 
neuronal  function  and  protein  function. 
In  this  way,  information  gathered  in 
molecular  biological  studies  of  recom- 
binant proteins  can  be  used  to  test 
predictions  about  the  importance  of 
particular  alcohol-sensitive  sites  on  a 
protein  in  intact  neurons  and  the 
intact  brain. 

The  alcohol  sensitivity  of  a  protein 
may  also  differ  as  a  function  of  the 
subunit  composition  of  the  holoprotein, 
or  it  may  depend  on  crucial  interac- 
tions with  other  proteins.  Indeed,  dif- 
ferences in  protein  assembly  and 
interactions  might  underlie  some  of 
the  variability  in  alcohol  actions.  In 
addition,  closely  related  proteins,  such 
as  different  kinase  isoforms,  may  differ 
in  alcohol  sensitivity,  and  it  will  be 
important  to  determine  which  protein 
subtypes  are  most  sensitive  to  alcohol 
and  contribute  to  acute  alcohol  effects 
on  the  brain. 

I  will  begin  with  a  discussion  of 
multimeric  proteins.  The  first  step  in 
this  line  of  research  is  to  determine 
the  subunits  that  can  contribute  to  the 
formation  of  multimeric  proteins  by 
cloning  homologous  proteins  and  to 
examine  subunit  expression  in  neurons 
as  well  as  co-assembly  in  heterologous 
expression  systems.  This  has  already 
been  done  for  a  number  of  potential 
alcohol  targets,  including  ligand-  and 
voltage-gated  ion  channels  (for  reviews, 


see  Sanna  and  Harris  1993;  Seeburg 
et  al.  1995;  Diamond  and  Gordon 
1997;  Lovinger  1997).  Determining 
subunit  composition  and  stoichiometry 
of  assembled,  functional  proteins  is 
more  difficult.  Techniques  such  as  co- 
immunoprecipitation  can  be  used  to 
provide  evidence  for  subunit  co-assembly 
(Khan  et  al.  1994;  Ruano  et  al.  1994; 
Sheng  et  al.  1994),  and  this  line  of 
research  will  benefit  from  development 
of  subunit-specific  antibodies.  Pharma- 
cological approaches  that  can  identify 
certain  subclasses  of  heteromeric  pro- 
teins might  also  be  of  use. 

Single-cell  polymerase  chain  reac- 
tion (PCR)  identification  of  subunit 
expression  in  neurons,  combined  with 
analysis  of  alcohol  effects  and  pharma- 
cological analysis,  is  likely  to  be  useful 
(Criswell  et  al.  1996;  Sapp  and  Yeh 
1997).  However,  this  technique  cannot 
provide  any  information  about  protein 
expression  or  co-assembly.  The  develop- 
ment of  techniques  for  more  rapid 
and  complete  identification  of  protein 
expression  in  a  single  neuron  would 
be  a  significant  advance,  but  no  suitable 
approach  has  been  developed  as  yet. 

Co-expression  of  protein  subunits  in 
a  heterologous  expression  system  can 
also  yield  valuable  information  about 
the  relationship  between  protein  subunit 
composition  and  acute  alcohol  sensi- 
tivity. This  approach  has  now  been 
widely  used  to  examine  alcohol  effects 
on  ion  channels  and  other  proteins 
expressed  in  Xenopus  oocytes  and 
mammalian  cells  ( Wafford  and  Whit- 
ing 1992;  Kuner  et  al.  1993;  Lovinger 
1993;  Sigel  et  al.  1993;  Masood  et  al. 
1994;  Dildy-Mayfield  and  Harris 
1995;  Lovinger  1995;  Mihic  et  al. 


12 


Neural  Proteins 


1997).  It  is  best  to  use  this  approach 
in  conjunction  with  techniques  that 
can  allow  the  investigator  to  be  sure 
that  all  of  the  desired  subunits  are 
expressed.  This  sort  of  analysis  might 
include  pharmacological  characteriza- 
tion of  receptors  and  immunological 
detection  of  subunit  protein  expression. 
Alcohol  effects  on  proteins  in  heterol- 
ogous expression  systems  may  not 
always  agree  with  effects  observed  in 
neurons  expressing  native  receptors 
(e.g.,  Lovinger  1993,  1995).  Likewise, 
results  obtained  using  different  heterol- 
ogous systems  and  methods  of  assaying 
receptor  function  may  not  always  agree 
( Wafford  and  Whiting  1992;  Kuner  et 
al.  1993;  Sigel  et  al.  1993;  Marszalec 
et  al.  1994;  Masood  et  al.  1994; 
Lovinger  1995;  Mihic  et  al.  1997).  It 
will  be  important  to  document  these 
differences,  since  differences  in  alco- 
hol sensitivity  of  a  protein  in  different 
cellular  contexts  may  provide  informa- 
tion that  will  be  useful  in  identifying 
the  cellular  constituents  that  deter- 
mine differential  alcohol  sensitivity. 

Preventing  expression  of  a  particular 
subunit  protein  followed  by  analysis 
of  changes  in  protein  function,  phar- 
macology, and  alcohol  sensitivity  is  an 
experimental  approach  that  could 
yield  a  wealth  of  information  about 
subunit  structure  of  proteins  and  the 
importance  of  particular  subunits  in 
conferring  alcohol  sensitivity.  Anti- 
sense  RNA  technology  can  be  used  to 
reduce  expression  of  a  particular  pro- 
tein subunit  (for  reviews,  see  Baertschi 
1994;  Bennett  1998).  However,  this 
technique  is  not  always  applicable  to 
proteins  with  a  slow  turnover  rate  in 
cells,  and  antisense  knockout  is  often 


incomplete.  Still,  there  may  be  specific 
cases  where  antisense  knockout  of  a 
particular  protein  subunit  is  feasible  and 
can  yield  information  about  the  impor- 
tance of  that  subunit  in  alcohol  actions. 

Production  of  subunit  knockout 
mice  is  likely  to  be  a  more  fruitful 
approach  to  the  problem.  Indeed,  sev- 
eral animals  with  receptor  subunit 
knockouts  have  already  been  produced, 
and  some  of  these  animals  have  been 
tested  for  acute  alcohol  sensitivity 
(Homanics  et  al.  1997).  Other  knock- 
out mouse  lines  have  been  made  that 
would  be  quite  useful  in  alcohol 
research,  but  are  not  presently  avail- 
able to  alcohol  researchers.  A  crucial 
direction  for  future  research  on  acute 
alcohol  actions  will  be  to  foster  the 
creation  and  use  in  alcohol  studies  of 
knockout  mice  that  lack  proteins 
thought  to  be  critical  for  acute  alco- 
hol actions  in  the  brain.  The  standard 
mouse  gene  knockout  approach  is  not 
without  problems,  however.  The  gene 
is  usually  engineered  such  that  no 
expression  of  the  protein  occurs 
throughout  the  lifespan  of  the  mouse. 
This  can  lead  to  problems  of  develop- 
ment and  compensation  that  may 
affect  analysis  of  acute  alcohol  actions 
in  the  mature  animal.  Thus,  it  will  be 
important  to  use  alternative  knockout 
strategies,  such  as  inducible  knockouts, 
that  allow  for  removal  of  protein  expres- 
sion at  defined  times  in  mouse  devel- 
opment. More  will  be  said  about  these 
powerful  alternative  approaches,  and  the 
need  to  use  them  in  alcohol  research, 
in  the  last  section  of  this  chapter. 

Many  of  the  experimental  strategies 
discussed  in  the  last  few  paragraphs 
can  also  be  applied  to  the  study  of 


13 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


different  protein  subtypes  even  in 
homomeric  proteins.  For  example,  in 
examining  alcohol  effects  on  protein 
kinase  activity  it  will  be  important  to 
identify  kinase  subtypes  within  neu- 
rons that  are  expressed  and  are  alco- 
hol sensitive.  Likewise,  examination  of 
acute  alcohol  effects  in  kinase  knock- 
out mice  will  be  an  important  step  in 
determining  the  role  of  these  enzyme 
subtypes  in  alcohol  sensitivity.  A  few 
kinase  knockout  mice  have  already 
been  found  to  exhibit  differences  in 
alcohol  sensitivity  and  acute  tolerance 
(Harris  et  al.  1995;  Miyakawa  et  al. 
1997).  One  additional  strategy  that 
should  be  valuable  in  the  study  of 
alcohol  effects  on  enzyme  function  is 
the  examination  of  the  activity  of 
purified  enzyme  subtypes.  Many  of 
the  enzymes  that  are  of  interest  to 
alcohol  researchers  can  be  purified  to 
homogeneity,  and  their  activity  can  be 
assayed  directly.  This  approach  is  use- 
ful in  examining  acute  alcohol  effects 
on  particular  enzyme  subtypes. 

As  discussed  earlier,  interactions 
between  proteins  can  regulate  the 
subcellular  distribution  and  function 
of  alcohol  target  proteins.  Analysis  of 
the  importance  of  such  interactions  in 
acute  alcohol  sensitivity  is  thus  a  key 
area  for  future  research.  The  first  step 
in  this  line  of  research  is  the  identifica- 
tion of  proteins  that  interact  with 
alcohol  target  proteins.  This  identifi- 
cation can  proceed  in  a  number  of 
ways.  Interacting  proteins  can  be 
identified  using  gel  overlay  assays 
(Carr  and  Scott  1992),  solution  bind- 
ing assays  such  as  surface  plasmon  res- 
onance (Faux  and  Scott  1997),  and 
genetic  screens  for  binding  such  as  the 


yeast  two-hybrid  screen  (Fields  and 
Stern glanz  1994).  Most  of  the  binding 
assays  require  having  at  least  one  puri- 
fied protein,  such  as  a  GST-fusion 
protein,  that  can  be  used  to  assay 
binding  to  the  unknown  interacting 
protein.  The  yeast  two-hybrid  screen 
requires  only  that  DNA  for  the  pro- 
tein of  interest  can  be  successfully 
transfected  into  yeast  and  will  yield 
protein.  This  protein  is  used  as  "bait" 
to  catch  other  proteins  from  brain 
DNA  libraries  that  are  expressed  in 
yeast  (Fields  and  Sternglanz  1994). 
The  DNA  sequences  for  these  proteins 
can  then  be  determined  and  the  pro- 
teins, if  unknown,  can  be  cloned  and 
characterized.  All  of  these  techniques 
have  been  used  to  identify  interacting 
proteins  with  great  success  in  recent 
years.  One  other  way  to  search  for 
potential  protein-protein  interactions 
is  to  examine  the  sequences  of  puta- 
tive alcohol  target  proteins  for  motifs 
known  to  be  involved  in  protein  bind- 
ing (e.g.,  PDZ  domains  [Kornau  et  al. 
1997;  Ranganathan  and  Ross  1997]). 
This  approach  can  help  direct  the 
search  toward  proteins  that  are  known 
to  interact  with  such  motifs. 

Once  interacting  proteins  are  iden- 
tified, then  the  impact  of  expressing 
or  removing  these  proteins  on  alcohol 
sensitivity  of  the  putative  alcohol  tar- 
get protein  can  be  examined.  These 
sorts  of  analyses  can  be  performed  in 
neurons  and  in  heterologous  expres- 
sion systems.  Knockout  animals  that 
lack  an  interacting  protein  of  interest 
can  also  be  produced,  and  their  alco- 
hol sensitivity  can  then  be  examined. 
This  work  should  include  examination 
of  alcohol  effects  in  the  intact  animal 


14 


Neural  Proteins 


using  behavioral-pharmacological 
approaches,  as  well  as  examination  of 
changes  in  alcohol  effects  on  the  recep- 
tor, channel,  transporter,  or  signaling 
enzyme  of  interest  in  cells  or  brain 
slices.  Strategies  aimed  at  preventing 
specific  protein-protein  interactions  in 
intact  cells  can  also  be  employed.  Pep- 
tides that  disrupt  interactions  by  bind- 
ing to  the  interaction  domain  can  be 
overexpressed  in  cells  by  cDNA  trans - 
fection  (Hundle  et  al.  1997)  or  injected 
into  cells  via  a  patch-clamp  pipette 
(Rosenmund  et  al.  1994).  These  strate- 
gies allow  investigators  to  disrupt 
interactions  and  examine  the  alcohol 
sensitivity  of  the  target  protein  with 
and  without  this  protein  interaction. 

In  some  cases  direct  alcohol  effects 
on  protein -protein  interactions  can  be 
examined.  If  the  proteins  are  sufficiently 
pure  to  allow  performance  of  a  solution 
binding  assay,  then  alcohol  effects  on 
binding  can  be  quantified  using  this 
approach.  Semiquantitative  measure- 
ments of  binding  and  alcohol  effects 
can  be  obtained  using  a  gel  overlay 
assay.  A  less  direct  approach,  but  one 
that  can  yield  important  preliminary  data, 
is  immunocytochemical  examination  of 
alcohol  effects  on  target  protein  subcel- 
lular localization  (Gordon  et  al.  1997). 
This  technique  allows  researchers  to 
determine  if  alcohol  alters  interaction  of 
one  protein  with  a  target  protein  that 
participates  in  localization.  Powerful 
techniques  for  subcellular  imaging,  such 
as  confocal  and  multiphoton  excitation 
laser  microscopy,  will  be  needed  to  carry 
out  these  studies.  This  sort  of  experiment 
might  be  a  good  first  step  in  identifying 
protein-protein  interactions  that  are 
affected  by  alcohol  in  the  intact  cell. 


The  significance  of  this  research  will 
be  mosdy  in  the  area  of  development  of 
pharmacotherapies  for  treatment  of  alco- 
hol abuse  and  alcoholism.  More  exact 
definition  of  the  sites  of  action  and 
allosteric  effects  of  alcohol  at  the  level  of 
single  molecules,  molecular  interactions, 
and  subcellular  compartments  should 
aid  in  application  of  rational  drug  design 
to  the  development  of  such  pharma- 
cotherapies. However,  understanding 
targets  of  alcohol  action  at  the  molecular 
level  may  also  lead  to  clinical  applica- 
tions such  as  enhanced  diagnosis  of 
susceptibility  to  alcoholism. 

ANALYSIS  OF  PROTEIN 
FUNCTION 

Goals 

An  important  part  of  understanding 
acute  alcohol  effects  on  key  neuromol- 
ecular  targets  is  determining  the  changes 
in  molecular  function  produced  by 
alcohol.  However,  describing  functional 
effects  should  not  be  the  ultimate  goal 
of  investigators  examining  functional 
effects  of  alcohol.  Alcohol  effects  on 
protein  function  should  be  undertaken 
with  an  eye  to  identifying  molecular 
characteristics  that  impart  alcohol  sen- 
sitivity. With  a  wealth  of  information 
emerging  about  the  relationship 
between  protein  structure  and  func- 
tion, functional  information  can  be 
used  to  home  in  on  regions  of  proteins 
that  impart  alcohol  sensitivity. 

Approaches 

Different  subtypes  of  ion  channels 
appear  to  be  sensitive  to  pharmaco- 
logically relevant  concentrations  of 


15 


NIAAA's  Ncuroscicncc  and  Behavioral  Research  Portfolio 


alcohol  (Sanna  and  Harris  1993;  Dia- 
mond and  Gordon  1997;  Lovinger 
1997).  A  great  deal  of  information 
about  the  perturbation  of  ion  channel 
function  can  be  gained  by  kinetic 
analyses  using  data  collected  at  the 
whole-cell,  and  particularly  at  the  sin- 
gle-channel, electrophysiological 
recording  level.  These  techniques 
have  already  been  applied  to  research- 
ing alcohol  effects  on  several  ion 
channel  subtypes  (Mullikin-Kilpatrick 
and  Treistman  1995;  Nagata  et  al. 
1996;  Wright  et  al.  1996;  Zhou  et  al. 
1998).  Studies  have  revealed  effects 
on  probability  of  channel  opening, 
ligand  dissociation  at  ligand-gated 
channels,  and  interactions  with  G  pro- 
tein modulation  of  voltage-gated 
channels  (Mullikin-Kilpatrick  and 
Treistman  1995;  Mullikin-Kilpatrick 
et  al.  1995;  Nagata  et  al.  1996; 
Wright  et  al.  1996;  Zhou  et  al.  1998). 
This  sort  of  information,  along  with 
information  about  alcohol  interactions 
with  agonists,  antagonists,  and 
allosteric  modulators,  can  help  focus 
further  studies  designed  to  identify 
molecular  sites  of  alcohol  action. 
Since  these  approaches  are  well  known 
in  the  alcohol  field,  there  is  no  need 
for  further  description  of  this  research 
area.  However,  it  will  be  important  to 
apply  these  kinetic  analysis  techniques 
to  examination  of  alcohol  effects  on 
recombinant  receptors  of  known 
structure,  since  this  might  help  to 
avoid  problems  with  mixed  alcohol 
effects  observed  in  neurons  containing 
many  channel  subtypes. 

Kinetic  analysis  can  also  be  per- 
formed on  alcohol-sensitive  nonchannel 
receptors  as  well  as  alcohol-sensitive 


enzymes  and  even  nuclear  factors. 
This  information  can  be  combined  with 
thermodynamic  analysis  to  provide 
some  idea  of  the  energetic  changes 
taking  place  within  target  proteins 
during  exposure  to  alcohol. 

Transporter  proteins  for  neuro- 
transmitters and  other  molecules  can 
function  as  ion  channels  (Parent  and 
Wright  1993;  DeFelice  and  Galli 
1998).  The  channel  mode  of  activity 
may  or  may  not  play  a  role  in  trans- 
port, but  it  is  likely  to  have  an  impact 
on  neuronal  physiology  (Bruns  et  al. 
1993).  It  will  be  important  for  investi- 
gators to  examine  alcohol  effects  on 
channel  activity  of  these  neuromole- 
cules.  This  will  be  particularly  impor- 
tant in  examination  of  transporters 
that  are  found  to  display  high  alcohol 
sensitivity  when  assayed  for  transport 
activity.  Transporters  for  neurotrans- 
mitters suspected  to  play  a  role  in 
acute  alcohol  actions  (adenosine, 
dopamine,  gamma-aminobutyric  acid 
[GABA],  glutamate,  serotonin)  might 
also  be  examined  for  changes  in  trans- 
porter/channel function. 

The  body  of  evidence  implicating 
protein  phosphorylation/dephosphory- 
lation  in  the  neural  effects  of  alcohol 
continues  to  grow,  although  examina- 
tion of  phosphorylation  and  kinases 
has  greatly  outpaced  study  of  dephos- 
phorylation  and  phosphatases.  In  par- 
allel, our  understanding  of  the 
mechanisms  regulating  protein  kinases 
and  phosphatases  has  also  greatly 
expanded.  For  example,  it  is  now  well 
known  that  these  enzymes  are  regu- 
lated by  intracellular  targeting,  and 
that  this  targeting  involves  interac- 
tions with  "anchoring"  proteins  or 


16 


Neural  Proteins 


scaffolding  proteins  (Mochly- Rosen 
1995;  Dell'Aqua  and  Scott  1997). 
This  is  an  area  that  is  just  beginning 
to  receive  attention  in  the  alcohol 
research  field,  and  was  discussed  in 
more  detail  earlier  in  this  chapter.  In 
the  context  of  differential  kinase/ 
phosphatase  localization,  it  is  also 
important  to  examine  enzyme  activity  at 
different  substrates,  since  differential 
enzyme  localization  will  bring 
kinases/phosphatases  into  contact 
with  different  substrates.  Future  studies 
of  kinase  and  phosphatase  function 
should  include  examination  of  phos- 
phorylation and  dephosphorylation  of 
different  substrates  and  activation  of 
the  kinase  by  different  cofactors.  Sub- 
strate and  cofactor  identity  has  already 
proved  important  in  studies  of  protein 
kinase  C  (PKC),  since  alcohol  and 
anesthetic  effects  on  this  kinase  appear 
to  be  different  under  different  sub- 
strate and  cofactor  conditions  (Slater 
et  al.  1993,  1997).  Examination  of 
activation  of  enzymes  by  different 
activators  and  cofactors  will  also  be 
helpful  in  identifying  potential  sites  of 
alcohol  action  on  the  enzyme  molecule. 
Likewise,  examination  of  holoen- 
zymes  and  enzyme  activity  in  the 
presence  of  interacting  proteins  will 
be  important,  since  these  studies 
might  reveal  actions  of  alcohol  that 
involve  other  proteins  in  addition  to 
the  enzyme  itself. 

The  importance  of  this  line  of 
research  is  mainly  to  help  guide  analysis 
of  molecular  structure.  Understanding 
the  functional  effects  of  alcohol  on  a 
protein  should  help  investigators  to 
discover  the  parts  of  the  protein  that 
are  involved  in  the  actions  of  alcohol. 


Understanding  the  functional  effects 
of  alcohol  on  individual  proteins  can  also 
aid  in  the  design  and  evaluation  of 
potential  pharmacotherapies.  Preclini- 
cal screens  for  the  efficacy  of  potential 
therapeutic  agents  can  be  designed 
using  assays  of  protein  function. 

ALCOHOL  EFFECTS 
ON  TRANSMISSION 
AT  INTACT  SYNAPSES 

Goals 

Another  important  goal  of  alcohol 
research  is  to  understand  the  way  in 
which  alcohol  alters  communication 
between  neurons  at  intact  synapses.  It 
has  now  become  apparent  that  alcohol 
has  potent  actions  on  synaptic  transmis- 
sion. To  some  extent,  these  actions 
can  be  accounted  for  by  alterations  in 
synaptic  proteins  that  are  known  to  be 
alcohol  sensitive  (e.g.,  neurotransmitter 
receptors  and  ion  channels).  However,  it 
is  not  clear  that  all  of  the  actions  of  alco- 
hol at  intact  synapses  can  be  explained 
by  effects  on  these  proteins.  For  exam- 
ple, there  is  emerging  evidence  that 
alcohol  enhances  inhibitory  transmission 
and  inhibits  excitatory  neurotransmission 
by  altering  presynaptic  mechanisms  of 
neurotransmitter  secretion  (Thomas 
and  Morrisett  1997#;  Weiner  et  al. 
1997  a).  Furthermore,  past  studies  have 
suggested  alcohol  effects  on  release  of 
neuromodulators  (Wang  et  al.  1991; 
Wozniak  et  al.  1991).  Many  of  these 
actions  cannot  be  fully  explained  by 
our  current  knowledge  of  alcohol 
effects  on  synaptic  targets.  The  extent 
to  which  known  alcohol-sensitive  target 
proteins  contribute  to  alcohol  actions 


17 


NIAAA's  Ncurosciencc  and  Behavioral  Research  Portfolio 


on  synaptic  transmission  also  needs  to 
be  examined  in  greater  detail.  For  these 
reasons,  it  is  important  to  examine 
alcohol  effects  on  synaptic  transmission 
using  techniques  that  allow  the  inves- 
tigator to  infer  the  pre-  or  postsynap- 
tic locus  of  the  effects  and  to  assay  the 
involvement  of  particular  synaptic 
proteins  in  the  actions  of  alcohol. 

Approaches 

The  quantal  nature  of  neurotransmit- 
ter release  has  long  been  known  from 
studies  of  the  neuromuscular  junction. 
However,  it  had  been  quite  difficult 
to  apply  analyses  based  on  quantal 
theory  to  examination  of  central 
synapses  (Redman  1990;  Korn  and 
Faber  1991).  Recent  innovations, 
including  development  of  tight-seal 
whole-cell  recording  from  neurons 
visualized  in  brain  slices,  have  made  it 
possible  to  analyze  quantal  transmis- 
sion at  central  nervous  system  (CNS) 
synapses  with  greater  accuracy 
(Clements  1990;  Bekkers  and  Stevens 
1994;  von  Kitzing  et  al.  1994;  Isaac 
et  al.  1996).  The  most  promising 
techniques  are  those  that  allow  the 
investigator  to  examine  spontaneous 
synaptic  responses  at  excitatory  and 
inhibitory  synapses.  Information 
about  the  pre-  versus  postsynaptic 
locus  of  changes  in  transmission  can 
be  gained  from  analysis  of  the  fre- 
quency, amplitude,  and  time  course  of 
these  responses.  Furthermore,  one  can 
directly  examine  quantal  responses  by 
examining  spontaneous  "miniature" 
synaptic  currents  under  conditions  in 
which  calcium-dependent  secretion 
has  been  blocked.  This  technique  pro- 
vides even  more  powerful  determina- 


tion of  the  locus  of  changes  at  individ- 
ual synapses,  and  allows  the  investiga- 
tor to  separate  effects  on  presynaptic 
neuronal  firing  from  effects  on  the 
presynaptic  terminal  itself.  Using  vari- 
ations on  this  basic  experimental 
approach,  the  involvement  of  presy- 
naptic calcium  entry  and  specific  post- 
synaptic receptors  and  signaling 
enzymes  in  effects  on  transmission  can 
also  be  examined.  These  techniques  are 
just  beginning  to  be  applied  to  exami- 
nation of  alcohol  effects  at  glutamater- 
gic  and  GABAergic  synapses,  and 
preliminary  studies  indicate  that 
important  information  about  hereto- 
fore-overlooked presynaptic  effects  of 
alcohol  will  be  forthcoming  from  this 
line  of  research  (Thomas  and  Mor- 
risett  1997^;  Weiner  et  al.  1997^). 

More  sophisticated  methods  for 
analysis  of  transmission  at  single  CNS 
synapses  have  been  developed  in 
recent  years.  Investigators  can  use 
modifications  of  basic  techniques  for 
examination  of  stimulus-evoked 
transmission  to  investigate  changes  in 
quantal  release  when  afferent  fibers 
are  stimulated  (e.g.,  Sr2+-induced 
asynchronous  transmitter  release 
[Oliet  et  al.  1996;  Choi  and  Lovinger 
1997]).  In  addition,  the  possibility 
that  CNS  synapses  deviate  from 
quantal  behavior  observed  at  periph- 
eral synapses  can  now  be  assessed 
using  techniques  to  examine  "silent 
synapses"  (Malenka  and  Nicoll  1997) 
and  variations  in  neurotransmitter 
release  at  single  synapses  (Liu  and 
Tsien  1997).  Thus,  detailed  investiga- 
tion of  alcohol  effects  on  quantal 
synaptic  transmission  can  now  be  car- 
ried out  at  CNS  synapses. 


18 


Neural  Proteins 


If,  indeed,  presynaptic  mechanisms 
contribute  to  the  actions  of  alcohol  at 
central  synapses,  then  it  will  be  neces- 
sary to  more  closely  examine  alcohol 
effects  on  physiological  and  neuro- 
chemical events  taking  place  within 
axon  terminals.  Calcium  has  a  key  role 
in  neurotransmitter  secretion,  and 
examination  of  axon  terminal  calcium 
dynamics  will  likely  be  a  prominent 
area  of  future  investigation.  Although 
alcohol  effects  on  calcium  flux  have 
been  examined  in  numerous  studies, 
most  of  these  studies  have  measured 
calcium  flux  into  synaptosomes  or 
other  reduced  preparations,  and  cal- 
cium flux  has  been  evoked  by  stimuli 
of  quite  long  duration  (seconds)  in 
comparison  with  the  timing  of  influx 
during  excitation/secretion  coupling 
at  CNS  synapses  (a  few  milliseconds). 
Thus,  it  cannot  be  determined  from 
these  studies  if  alcohol  predominantly 
affects  calcium  influx  related  to  neu- 
rotransmitter secretion  or  calcium 
increases  that  take  place  at  later  times 
and  may  not  be  so  crucial  for  release.  A 
large  body  of  literature  on  alcohol 
effects  on  voltage -gated  calcium  chan- 
nels also  exists,  and  some  of  these  stud- 
ies have  focused  on  channels,  such  as 
the  N-channel,  that  play  a  role  in  exci- 
tation/secretion coupling  (Solem  et  al. 
1997).  However,  these  examinations 
have  been  limited  to  measuring  chan- 
nel function  in  neuronal  somata  and 
not  at  synaptic  terminals,  and  it  is  quite 
possible  that  channels  within  terminals 
are  regulated  differently  than  channels 
in  the  cell  body.  Thus,  further  investi- 
gation of  alcohol  effects  on  calcium 
transients  and  calcium  channel  func- 
tion in  axon  terminals  is  warranted. 


The  most  direct  way  to  examine 
calcium  channel  function  in  presynaptic 
terminals  is  to  use  voltage-clamp  tech- 
niques to  measure  pharmacologically 
isolated  calcium  currents.  Studies  indi- 
cate that  simultaneous  voltage-clamp 
recording  from  both  pre-  and  postsy- 
naptic elements  of  the  synapse  can  be 
carried  out  at  brainstem  calyx  synapses 
such  as  the  synapse  of  Held  (Borst 
and  Sakmann  1996).  This  approach 
will  allow  investigators  to  examine 
alcohol  effects  on  presynaptic  physiol- 
ogy, including  calcium  channel  func- 
tion, and  to  relate  those  effects  to 
alcohol-induced  changes  in  synaptic 
transmission  at  an  excitatory  synapse. 

However,  most  presynaptic  terminals 
are  too  small  to  be  examined  with  patch- 
clamp  recording.  Thus,  presynaptic 
terminal  calcium  dynamics  must  be 
examined  in  other  ways  at  these 
synapses.  Newer  and  more  sophisti- 
cated techniques  for  measurement 
and  imaging  of  presynaptic  calcium 
dynamics  have  been  developed  in 
recent  years.  Most  of  these  techniques 
involve  the  use  of  calcium-sensitive 
fluorescent  dyes,  such  as  fura-2,  that 
are  reliable  indicators  of  changes  in 
intracellular  calcium.  Using  a  photo- 
multiplier  tube,  it  is  possible  to  detect 
photon  emission  excitation  from  a  flu- 
orescent dye  in  real  time  and  thus 
measure  intracellular  calcium  dynamics 
without  the  need  for  sophisticated 
imaging  hardware.  Saggau  and  col- 
leagues have  elegantly  demonstrated 
that  one  can  use  such  an  approach  to 
measure  pre-  and  postsynaptic  calcium 
transients  during  synaptic  transmission 
evoked  by  a  single  presynaptic  stimu- 
lus at  a  population  of  CA3-CA1 


19 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


synapses  in  die  hippocampal  slice  (Wu 
and  Saggau  1997).  Through  the  use 
of  fura-2  loading  only  in  the  synaptic 
region  and  pharmacological  separation 
of  pre-  and  postsynaptic  transients, 
these  investigators  have  demonstrated 
modulation  of  presynaptic  calcium 
transients  by  G  protein-coupled 
receptors.  This  method  for  analysis 
could  easily  be  applied  to  examination 
of  alcohol  effects  on  synapses  in  the 
hippocampus  and  other  brain  regions. 
Direct  visualization  of  presynaptic 
terminal  calcium  transients  can  be  car- 
ried out  using  confocal  and  two-pho- 
ton excitation  laser  microscopy.  These 
techniques  allow  one  to  examine 
regions  as  small  as  0.5  um,  making 
presynaptic  terminal  visualization  pos- 
sible. Indeed,  these  techniques  have 
already  been  used  to  image  calcium 
transients  in  single  dendritic  spines 
(Denk  et  al.  1996),  and  this  technol- 
ogy should  be  applicable  to  axon  ter- 
minals as  well.  Using  techniques 
designed  to  load  dyes  into  terminals 
in  combination  with  vital  dyes  that 
can  label  terminals  (such  as  FM-143), 
it  should  be  possible  to  unequivocally 
identify  terminals  that  are  loaded  with 
calcium  indicator  dye  and  examine 
calcium  transients  and  the  effects  of 
alcohol  on  these  transients  in  single 
axon  terminals.  Improvements  in 
microscopy  hardware  should  make  it 
possible  to  measure  such  transients  at 
high  scan  and  digitization  rates,  and 
thus  it  should  be  possible  to  measure 
the  transients  that  are  directly  related 
to  neurotransmitter  release.  Combining 
this  analysis  with  pharmacological 
approaches  to  assess  the  involvement 
of  calcium  channels,  intracellular  calcium 


release  processes,  and  calcium  buffering/ 
extrusion  mechanisms  will  allow  inves- 
tigators to  pinpoint  particular  aspects 
of  the  calcium/secretion  relationship 
that  are  altered  by  alcohol.  This  will 
stimulate  further  analysis  of  alcohol 
effects  on  potential  target  proteins 
involved  in  these  processes.  Combining 
calcium  measurement  techniques  with 
sophisticated  electrophysiological 
analysis  of  synaptic  transmission  will 
provide  detailed  information  about  the 
role  of  the  physiological  consequences 
of  alterations  in  calcium  dynamics. 

Imaging  techniques  can  also  be 
applied  to  obtain  real-time  measure- 
ments of  neurotransmitter  release 
processes  in  addition  to  calcium 
dynamics.  For  example,  one  can  mea- 
sure vesicle  release  and  recycling  using 
the  fluorescent  membrane  probe  FM- 
143  (Ryan  et  al.  1993,  1997).  This 
dye  incorporates  into  vesicular  mem- 
branes and  will  remain  incorporated 
until  vesicle  fusion  is  stimulated.  Mea- 
surements of  secretion  can  be  made 
using  this  technique  in  combination 
with  confocal  microscopy.  Similar 
approaches  can  also  be  taken  using 
fluorescent  antibody  detection  of 
synaptic  vesicle-associated  proteins 
(Malgaroli  et  al.  1995).  This  approach 
can  provide  information  about  alcohol 
effects  on  secretion  that  can  be  com- 
pared with  measurement  of  calcium 
dynamics  in  order  to  help  investiga- 
tors determine  the  most  probable 
locus  of  alcohol  effects  on  neurotrans- 
mitter secretion. 

Monoamines,  such  as  dopamine,  and 
neuropeptides,  such  as  opioids,  may 
have  significant  roles  in  acute  alcohol 
actions  related  to  the  reinforcing 


20 


Neural  Proteins 


actions  of  the  drug.  Studies  performed 
in  vivo  indicate  that  alcohol  can  alter 
extracellular  dopamine  concentrations 
in  the  nucleus  accumbens  ( Wozniak  et 
al.  1991;  Samson  and  Hodge  1993; 
Weiss  et  al.  1993).  These  studies  have 
been  performed  using  neurotransmitter 
turnover,  microdialysis,  and  in  vivo 
voltammetry  techniques.  However,  it 
appears  that  alcohol  does  not  act  directly 
on  dopaminergic  terminals  and  may 
increase  extracellular  dopamine  in  vivo 
by  actions  in  the  ventral  tegmental  area 
(VTA)  (Samson  et  al.  1997;  Yim  et  al. 
1997).  Thus,  it  is  not  clear  that  further 
examination  of  alcohol  effects  on 
dopaminergic  axon  terminals  is  war- 
ranted at  this  time.  However,  exami- 
nation of  mechanisms  of  release  of 
other  monoaminergic  transmitters 
might  be  warranted. 

Little  emphasis  has  been  placed  on 
examining  alcohol  effects  on  presy- 
naptic proteins  involved  in  the  secretion 
process.  However,  many  of  the  pro- 
teins that  have  been  implicated  in 
alcohol  actions  reside  in  presynaptic 
terminals  as  well  as  in  postsynaptic 
elements.  Notable  among  these  pro- 
teins are  modulatory  neurotransmitter 
receptors  (e.g.,  adenosine  and  opiate 
receptors),  neurotransmitter  trans- 
porters, voltage-gated  calcium  chan- 
nels, and  protein  kinases,  such  as 
PKC,  that  have  been  implicated  in 
altering  the  secretion  process.  In  addi- 
tion, there  has  been  substantial 
progress  in  recent  years  in  the  identifi- 
cation of  proteins  that  make  up  the 
neurotransmitter  release  machinery 
(see  Sudhof  1995  for  review),  and  as 
yet  there  has  been  little  effort  to 
examine  these  proteins  in  relation  to 


alcohol  effects  on  synaptic  transmission. 
This  is  obviously  another  area  that 
needs  to  be  emphasized  more  strongly 
in  future  studies  of  alcohol  effects  on 
synaptic  transmission. 

Plastic  changes  in  the  efficacy  of  syn- 
aptic transmission  have  been  suggested 
as  a  major  mechanism  of  information 
storage  in  the  nervous  system  (Bliss  and 
Collingridge  1993;  Goda  and  Stevens 
1996).  Alcohol  is  known  to  have 
amnestic  effects  and  to  disrupt  other 
aspects  of  cognitive  and  motor  function 
that  may  involve  such  plastic  changes. 
However,  studies  of  alcohol  effects  on 
synaptic  plasticity  have  been  limited 
mainly  to  examination  of  long-term 
potentiation  (LTP)  in  the  hippocampal 
formation  (Blitzer  et  al.  1990;  Mor- 
risett  and  Swartzwelder  1993;  Criado  et 
al.  1996;  Schummers  et  al.  1997).  Long- 
term  potentiation  and  long-term  depres- 
sion (LTD),  as  well  as  shorter  lasting 
changes  in  synaptic  efficacy,  have  been 
observed  at  a  number  of  synapses  in 
the  CNS  (Bear  and  Malenka  1994; 
Linden  and  Connor  1995).  There  is  no 
compelling  reason  to  examine  the  alcohol 
sensitivity  of  every  one  of  these  forms  of 
plasticity.  It  is  known,  for  example,  that 
LTP  in  the  cortex  involves  mechanisms 
similar  to  those  involved  in  LTP  at  the 
hippocampal  Schaffer  collateral-CAl 
synapses.  Thus,  it  may  not  be  necessary 
to  examine  alcohol  effects  on  cortical  LTP 
in  too  much  depth.  However,  LTP  at 
mossy  fiber-CA3  synapses  in  hippocam- 
pus and  LTP  at  parallel  fiber-Purkinje 
neurons  in  cerebellum  are  forms  of 
plasticity  that  involve  mechanisms  dif- 
ferent from  "classical"  NMDA  receptor- 
dependent  LTP  (Nicoll  and  Malenka 
1995;  Salin  et  al.  1996).  It  may  well  be 


21 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


important  to  examine  alcohol  effects 
on  these  forms  of  plasticity.  In  particular, 
cerebellar  LTD  should  be  examined 
since  alcohol  produces  ataxic  effects 
and  cerebellar  LTD  has  been  impli- 
cated in  motor  learning  and  is  related 
to  ataxic  phenotypes  in  mutant  mice 
(Linden  1994).  Examination  of  these 
forms  of  plasticity  may  help  investigators 
to  pinpoint  molecular  mechanisms  by 
which  alcohol  disrupts  plasticity  and  to 
identify  new  molecular  targets  of  alcohol 
action.  It  will  also  be  important  to 
examine  forms  of  plasticity  that  involve 
molecules  identified  as  alcohol  sensitive 
since  these  forms  of  plasticity  should 
be  altered  by  alcohol.  Ultimately,  infor- 
mation about  alcohol  effects  on  trans- 
mission and  synaptic  plasticity  may  be 
combined  at  certain  synapses  to  provide 
more  detailed  information  about  alcohol 
effects  on  neuronal  communication. 

The  use  of  mutant,  knockout,  and 
transgenic  mouse  models  will  also  be 
an  important  component  of  future 
studies  of  alcohol  effects  on  synaptic 
transmission.  Brain  slices  as  well  as  cul- 
tured and  acutely  isolated  neurons  and 
subneuronal  preparations  (e.g., 
microsacs  and  synaptoneurosomes)  can 
easily  be  prepared  from  mouse  brain. 
Studies  of  alcohol  effects  on  synaptic 
transmission  and  ligand-gated  ion 
channel  function  have  already  been  per- 
formed in  preparations  from  mutant  mice, 
highlighting  the  utility  of  this  pre- 
paration (Harris  et  al.  1995;  Miyakawa 
et  al.  1997).  Combining  analysis  of  neu- 
rophysiology and  synaptic  transmission 
with  the  new  generation  of  mouse 
genetic  alterations,  as  described  later  in 
this  chapter,  is  a  crucial  future  direction 
for  research  on  alcohol  mechanisms. 


It  is  known  that  alcohol  has  potent 
effects  on  synaptic  transmission. 
Understanding  the  mechanisms  under- 
lying these  effects  will  require  exami- 
nation of  intact  synapses  to  determine 
the  relative  contribution  of  pre-  and 
postsynaptic  mechanisms  in  the  effects 
of  alcohol.  This  will  lead  to  identifica- 
tion of  additional  important  molecular 
targets  of  alcohol  actions  and  will  pro- 
vide assays  for  preclinical  tests  of  drugs 
that  may  be  developed  for  treatment  of 
alcohol  abuse  and  alcoholism. 

EXAMINATION  OF 
ALCOHOL  EFFECTS 
ON  NEUROPHYSIOLOGY 
IN  KEY  BRAIN  REGIONS 

Goal 

The  aim  of  this  line  of  research  is  to 
determine  the  relationship  between 
alcohol  effects  on  neuronal  activity  and 
behavior  in  brain  regions,  such  as  the 
VTA,  amygdala,  hypothalamus,  cere- 
bellum, and  prefrontal  cortex,  thought 
to  be  important  in  alcohol-related 
behaviors.  This  research  will  help  inves- 
tigators to  evaluate  the  potential  role  of 
particular  alcohol  targets  in  alcohol 
effects  on  key  brain  regions;  it  will  also 
aid  in  the  search  for  previously  undis- 
covered targets  of  alcohol  action  that 
play  key  roles  in  alcohol  effects  in  these 
brain  regions. 

Approaches 

Relationship  of  Neuronal 
Activity  to  Behavior 

Brain  regions  that  appear  to  play 
important  roles  in  different  aspects  of 


22 


Neural  Proteins 


drug- seeking  behavior  and  different 
behavioral  consequences  of  alcohol 
intake  have  been  defined  using  experi- 
mental approaches  such  as  behavioral 
pharmacology  (see  Koob  and  Nestler 
1997  for  review)  and  gross  measures 
of  neuronal  activity  in  defined  brain 
regions  (2-deoxyglucose,  c-Fos) 
(Ryabinin  et  al.  1997;  Williams- 
Hemby  and  Porrino  1997).  In  vivo 
neurophysiological  approaches  have 
also  been  widely  used  (Gessa  et  al. 
1985;  Mereu  and  Gessa  1985;  Criado 
et  al.  1995;  Lee  et  al.  1995;  Ludvig  et 
al.  1995;  Givens  1996;  Matthews  et 
al.  1996;  Wang  et  al.  1996;  Wood- 
ward 1996).  Studies  designed  to 
examine  the  relationship  between  the 
activity  of  identified  neurons  within 
these  regions  and  the  behavioral 
effects  of  acute  alcohol  have  begun  to 
appear  in  the  last  few  years  (Givens 
1996;  Woodward  1996).  These  stud- 
ies are  of  particular  importance 
because  understanding  the  pattern  of 
changes  in  the  activity  of  these  neu- 
rons will  help  to  guide  examination  of 
the  specific  alcohol-induced  alter- 
ations in  ion  channel  function  and 
membrane  properties  that  underlie 
the  in  vivo  activity  changes.  Put 
another  way,  it  will  be  easier  to  search 
for  the  most  relevant  cellular  and  mol- 
ecular targets  of  alcohol  in  these  neu- 
rons once  we  know  how  their  activity 
is  altered  in  vivo  by  alcohol. 

While  it  is  true  that  a  number  of 
laboratories  have  examined  alcohol 
effects  on  the  activity  of  single  neu- 
rons in  vivo  over  the  years,  many  of 
these  investigators  have  not  been  able 
to  take  advantage  of  newly  developed 
techniques  for  neurophysiological 


recording  and  simultaneous  behavioral 
analysis.  These  techniques  can  also  be 
combined  with  pharmacological  analy- 
sis to  provide  an  even  stronger  experi- 
mental approach.  Several  techniques 
should  be  applied  to  this  analysis.  Multi- 
unit  recording  allows  the  investigator 
to  sample  several  neurons  within  a 
brain  region  and  gain  a  more  complete 
picture  of  activity  in  a  given  brain 
region  in  a  shorter  time  period  (Wilson 
and  McNaughton  1994;  Woodward 
1996).  Identification  of  neuronal  sub- 
types, while  often  performed  in  studies 
of  in  vivo  alcohol  effects,  is  not  uni- 
versally performed.  This  technique  is 
a  necessity,  given  the  idea  that  this 
analysis  will  guide  future  studies  of 
alcohol  effects  in  vitro,  and  investigators 
will  need  to  know  which  neurons  to 
target  for  in  vitro  analysis.  Video  analy- 
sis of  animal  behavior  correlated  in  time 
with  the  activity  of  single  neurons 
allows  investigators  to  determine  the 
temporal  relationship  between  neu- 
ronal activity  and  different  aspects  of  a 
complex  behavior  pattern.  This  tech- 
nique is  beginning  to  be  applied  to 
studies  of  alcohol  actions  in  the  nucleus 
accumbens  and  striatum  (Woodward 
1996).  Patterns  of  activity  of  neurons 
that  have  well-known  behavioral  cor- 
relates, such  as  place  cell  activity  and 
activity  during  learning  and  memory 
tasks  of  hippocampal  formation  neu- 
rons, are  also  being  examined  in  rela- 
tion to  alcohol  effects  (Givens  1996; 
Ludvig  et  al.  1995;  Matthews  et  al. 
1996).  Investigators  can  now  combine 
the  physiological/behavioral  analysis 
with  pharmacological  manipulation  of 
the  local  neurons  by  in  vivo  microdial- 
ysis  and  other  techniques  (Ludvig  et 


23 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


al.  1995;  Yang  et  al.  1996).  These 
techniques  will  allow  investigators  to 
examine  localized  actions  of  alcohol 
and  interactions  between  alcohol  and 
pharmacological  agents  acting  at  sus- 
pected alcohol  target  sites.  This  analy- 
sis will  help  to  guide  future  studies  by 
providing  information  about  which 
brain  regions  are  directly  affected  by 
alcohol  and  what  targets  may  be 
important  in  these  alcohol  actions. 

Brain  regions  that  need  to  be 
examined  include  the  VTA,  nucleus 
accumbens,  amygdala,  and  prefrontal 
cortex,  which  are  part  of  the  brain 
"reward"  circuitry  (Koob  and  Nestler 
1997).  The  amygdala  also  has  impor- 
tant roles  in  anxiety  and  may  play  an 
important  role  in  the  anxiolytic 
actions  of  alcohol.  The  cerebellum  is 
known  to  participate  in  alcohol- 
induced  ataxia,  and  thus  it  is  impor- 
tant to  examine  this  brain  region  in 
more  detail.  Continuing  examination 
of  the  hippocampal  formation  is  war- 
ranted, but  this  brain  region  need  not 
receive  the  majority  of  attention  as  it 
has  in  the  past.  Finally,  the  prefrontal 
cortex  has  important  cognitive  roles 
that  may  also  be  affected  by  alcohol. 
Thus,  the  different  types  of  neurons  in 
this  brain  region  require  examination 
as  well. 

Cellular/Molecular 
Mechanisms  That  Underlie 
In  Vivo  Alcohol  Effects 

Neurons  suspected  to  be  alcohol  sen- 
sitive from  in  vivo  studies  need  to  be 
studied  in  vitro  to  determine  alcohol 
effects  on  intrinsic  membrane  proper- 
ties or  synaptic  transmission  that 
underlie  the  in  vivo  alcohol  effects. 


These  studies  should  not  take  the 
form  of  surveys  of  all  neuronal 
responses,  but  should  instead  be  tar- 
geted at  mechanisms  that  are  likely  to 
underlie  the  types  of  changes  in  activ- 
ity observed  in  vivo.  For  example,  if  a 
rhythmically  firing  neuron  shows 
decreased  activity  in  the  presence  of 
alcohol,  then  ion  conductances 
known  to  contribute  to  that  rhythmic 
activity  should  be  examined.  If  an 
alcohol-sensitive  neuron  is  generally 
quiescent  in  the  absence  of  synaptic 
input,  then  synaptic  transmission 
might  be  the  logical  target  for  exami- 
nation. Where  possible,  these  studies 
should  utilize  information  from  in 
vivo  pharmacological  studies  in 
designing  experiments. 

The  techniques  that  can  be  used 
for  examination  of  alcohol  effects  on 
intrinsic  neuronal  responses  and 
synaptic  transmission  are  numerous. 
Neuronal  isolation  techniques  used  in 
combination  with  patch-clamp  elec- 
trophysiological approaches  have 
advanced  significantly  in  recent  years, 
such  that  healthy  neurons  can  be 
acutely  dissociated  from  many  brain 
regions  in  relatively  mature  rodents. 
This  is  the  preferred  preparation  for 
studying  ion  channel  modulation  by 
alcohol,  because  isolated  neurons  are 
free  of  the  influences  of  neighboring 
cells,  and  voltage-clamp  is  easily 
achieved  in  these  preparations.  Ion 
currents  of  different  types  can  also  be 
easily  isolated  and  pharmacologically 
manipulated  in  this  preparation. 

Brain  slices  and  primary  neuronal 
cultures  allow  investigators  to  exam- 
ine synaptic  transmission  in  reduced, 
semi-intact  preparations.  This  allows 


24 


Neural  Proteins 


for  sophisticated  analysis  of  the  pre-  or 
postsynaptic  locus  of  alcohol  effects  as 
described  earlier  in  this  chapter.  Phar- 
macological analysis  of  ligand- gated  ion 
channels  under  voltage -clamp  can  also 
be  performed  in  cultured  neurons. 
This  can  best  be  achieved  by  blocking 
action  potential  firing  and  using  local, 
rapid  application  of  receptor  agonists 
and  modulatory  drugs  with  a  system 
that  can  completely  superfuse  neurons 
with  a  known  concentration  of  drug 
(Lovinger  1995).  Detailed  analysis  of 
postsynaptic  receptors  in  neurons  in 
brain  slices  is  not  recommended  since 
it  is  difficult  to  achieve  application  of 
known  concentrations  of  drugs  to  the 
entire  cell  in  this  preparation.  Further- 
more, it  is  not  possible  to  definitively 
rule  out  presynaptic  effects  in  slice 
preparations.  However,  local  pressure 
or  iontophoretic  application  of  drugs 
to  these  cells  may  be  applied  to  neurons 
in  culture  to  examine  alcohol  effects  on 
receptors  or  transporters  in  different 
parts  of  a  given  neuron.  In  addition, 
techniques  for  laser  uncaging  of  agonists 
in  spatially  defined  extracellular  regions 
might  help  investigators  to  pinpoint 
regions  of  neurons  where  receptors  and 
transporters  are  particularly  sensitive 
to  alcohol  (Pettit  et  al.  1997). 

Examination  of  voltage-gated  ion 
channel  function  in  brain  slices  and  cul- 
tured neurons  is  unlikely  to  be  produc- 
tive given  the  space-clamp  problems 
encountered  in  these  studies.  However, 
some  channels  with  slow  kinetics,  such 
as  the  M-current  and  inwardly  rectify- 
ing K+  channels  activated  by  G  protein- 
coupled  receptors,  can  be  studied  with 
single-electrode  voltage-clamp  or 
patch-clamp  techniques  in  brain  slices 


(Dutar  and  Nicoll  1988;  Moore  et 
al.  1990). 

Given  the  emerging  evidence  for 
modulation  of  alcohol  effects  on  recep- 
tors involving  protein  phosphorylation 
(Mirshahi  and  Woodward  1997;  Weiner 
1997&),  it  will  be  especially  important 
for  investigators  to  take  precautions  to 
prevent  alteration  of  phosphoryla- 
tion/dephosphorylation  pathways 
during  whole-cell  recording  experi- 
ments. This  can  be  accomplished 
using  the  perforated  patch  technique, 
in  which  macroscopic  current  record- 
ing can  be  performed  without  dialysis 
of  intracellular  molecules  larger  than 
small  ions. 

One  problem  that  may  be  encoun- 
tered when  comparing  alcohol  effects 
in  vitro  with  those  observed  in  the 
same  neurons  in  vivo  is  that  synaptic 
connections  that  participate  in  alcohol 
effects  might  be  severed  during  brain 
slice  preparation.  Indeed,  recent  stud- 
ies in  dopaminergic  VTA  neurons 
suggest  that  addition  of  neurotrans- 
mitters in  vitro  can  enhance  alcohol 
sensitivity  of  neuronal  activity  (Brodie 
et  al.  1995).  This  sort  of  result  sug- 
gests the  possibility  that  a  neurotrans- 
mitter that  is  known  to  have  strong 
effects  on  a  neuron  in  vivo  might  not 
be  active  at  the  neuron  in  the  slice 
preparation.  If  this  neurotransmitter  is 
a  key  component  in  conferring  alco- 
hol sensitivity  on  the  neuron,  then  its 
absence  may  reduce  or  eliminate  alco- 
hol effects. 

One  way  to  attempt  to  overcome 
such  problems  in  vitro  will  be  through 
the  use  of  organotypic  brain  slice  cul- 
ture preparations  such  as  those  cur- 
rently in  use  for  studies  of  chronic 


25 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


alcohol  actions  (Thomas  and  Mor- 
risett  1997b).  Complex  brain  circuitry 
can  be  reconstituted  in  such  a  prepa- 
ration (Plenz  and  Kitai  1998).  The 
advantages  of  in  vitro  preparations, 
such  as  the  ability  to  examine  synaptic 
transmission  in  detail  and  apply  drugs 
to  defined  regions  of  a  neuron,  are 
retained  using  such  techniques. 

One  additional  advantage  of  in 
vitro  slice  and  slice  culture  prepara- 
tions is  the  ability  to  perform  experi- 
ments using  intracellular  imaging 
techniques.  These  approaches  allow 
the  investigator  to  examine  membrane 
potential  or  intracellular  calcium 
changes  in  large  arrays  of  neurons 
within  a  given  brain  region.  Examina- 
tion of  intracellular  changes  within 
small  subregions  of  neurons  (e.g., 
dendritic  spines  or  axon  terminals)  is 
also  possible  with  these  techniques. 
The  use  of  such  techniques  will  also 
allow  investigators  to  examine  physio- 
logical changes  and  effects  on  intracel- 
lular signaling  in  different  subcellular 
compartments. 

Examination  of  alcohol  effects  in 
knockout,  knockin,  transgenic,  and 
other  mutant  mice  will  be  an  impor- 
tant component  of  future  studies  of 
alcohol  actions  in  defined  brain 
regions.  Information  from  combined 
physiological  and  pharmacological 
studies  can  be  used  to  determine  what 
specific  neuronal  proteins  should  be 
overexpressed,  knocked  out,  or  subtiy 
altered  in  the  intact  animal.  These  ani- 
mals can  then  be  examined  using  in 
vivo  electrophysiological  and  behav- 
ioral approaches,  as  well  as  in  vitro 
electrophysiological  techniques.  This 
will  allow  investigators  to  determine 


the  importance  of  particular  proteins 
in  the  effects  of  alcohol  on  more  inte- 
grated neural  systems. 

These  studies  will  guide  preclinical 
investigation  of  potential  pharmaco- 
therapeutic  agents  by  providing  infor- 
mation about  which  molecular  targets 
are  affected  by  alcohol  in  a  given  brain 
region.  Understanding  alcohol  effects 
in  a  given  brain  region  will  aid  in  the 
development  of  diagnostic  tools  for 
use  in  humans  examined  with  nonin- 
vasive techniques.  Development  of 
brain  region-specific  therapeutic 
approaches  might  also  be  a  useful 
product  of  this  research. 

SEARCHING  FOR 
MOLECULAR  TARGETS 
WITH  HIGH  SENSITIVITY 
TO  ACUTE  ALCOHOL 

A  number  of  neurotransmitter  recep- 
tors, ion  channels,  and  signaling  mol- 
ecules have  been  identified  that  are 
sensitive  to  effects  of  alcohol  in  the 
25-100  mM  concentration  range. 
Certainly,  effects  of  these  concentra- 
tions are  relevant  to  in  vivo  actions 
after  ingestion  of  moderate  to  high 
doses  of  alcohol.  However,  alcohol 
has  effects  on  neural  function  and 
behavior  at  much  lower  concentra- 
tions (5-10  mM  and  lower),  and  these 
effects  may  be  especially  relevant  to 
the  behavioral  activation  and  rein- 
forcement produced  by  acute  alcohol. 
To  date,  very  few  neuromolecules 
have  been  identified  that  show  consistent 
functional  alterations  in  the  presence 
of  such  low  alcohol  concentrations. 
Identification  of  molecules  that  are 
extremely  sensitive  to  alcohol  actions 


26 


Neural  Proteins 


should  thus  be  a  priority  in  future 
alcohol  research. 

The  search  for  such  molecular  targets 
can  proceed  in  a  number  of  ways.  The 
first  approach  is  essentially  the  "top- 
down"  experimental  strategy 
described  in  the  preceding  section  of 
this  chapter.  This  approach  involves 
identification  of  changes  in  neuronal 
activity  in  vivo  that  are  produced  by 
low  concentrations  of  alcohol.  For 
example,  it  is  known  that  the  firing  of 
dopaminergic  VTA  and  substantia 
nigra  reticulata  neurons  is  altered  after 
low-dose  alcohol  administration 
(Gessa  et  al.  1985;  Mereu  and  Gessa 
1985).  Strategies  for  identifying  the 
molecular  targets  that  underlie  these 
effects  using  a  reductionist  approach 
have  already  been  discussed  in  this 
chapter.  However,  it  is  possible  that 
these  techniques  will  not  meet  with 
success  if  in  vivo  actions  are  secondary 
to  actions  on  other  neurons  or  involve 
alcohol  interactions  with  agents  or 
neuronal  pathways  that  are  not  pre- 
served in  a  particular  brain  slice  prepa- 
ration. If  this  is  the  case,  then  other 
screening  strategies  may  be  needed. 

The  use  of  genetic  screening  meth- 
ods is  already  established  in  the  alco- 
hol field.  Animals  with  differential 
responses  to  low-dose  alcohol  and  dif- 
ferential alcohol-drinking  behavior 
have  been  identified  and  selectively 
bred.  The  use  of  F2  intercross  breed- 
ing strategies  combined  with  genetic 
analyses  such  as  quantitative  trait  loci 
(QTL)  screening  has  the  potential  to 
provide  information  about  which 
gene  products  are  likely  to  be 
involved  in  these  differential  responses 
to  alcohol  (Belknap  et  al.  1997;  Buck 


et  al.  1997).  The  proteins  identified  in 
this  manner  can  then  be  tested  for 
alcohol  sensitivity,  and  many  of  the 
molecular  and  genetic  strategies  for 
examination  of  these  proteins  that  are 
outlined  in  this  chapter  can  be  applied 
to  these  gene  products.  The  major 
drawbacks  to  this  approach  are  the 
time  and  expense  needed  to  identify 
the  genes  within  QTL  that  are  impor- 
tant, and  the  possibility  that  the  prod- 
ucts of  these  genes  are  secondarily 
influencing  alcohol  sensitivity  and  are 
not  primary  targets  of  alcohol  action. 
However,  this  experimental  approach 
is  likely  to  provide  information  that  is 
relevant  to  understanding  the  neural 
basis  of  differentially  acute  alcohol 
effects  in  humans. 

An  alternative  genetic  screening  strat- 
egy involves  the  generation  of  mutant 
animals  that  can  then  be  screened  for 
altered  alcohol  sensitivity.  This 
approach  can  most  readily  be  applied 
to  invertebrates  such  as  Drosophila 
melanogaster  and  Caenorhabditis  ele- 
gans  at  present,  because  single-gene 
mutations  and  propagation  of  mutant 
animals  can  be  rapidly  achieved  in  these 
organisms.  Of  the  two  models, 
Drosophila  may  hold  more  promise 
since  these  animals  have  a  somewhat 
larger  behavioral  repertoire  than  C.  ele- 
gans.  Studies  by  the  Heberlein  and 
Nash  laboratories  are  aimed  at  reveal- 
ing mutations  in  Drosophila  that  alter 
effects  of  alcohol  and  general  anesthet- 
ics (Krishnan  and  Nash  1990;  Lei- 
bovitch  et  al.  1995;  Lin  and  Nash 
1996;  Scholz  et  al.  1997;  Moore  et  al. 
1998).  However,  most  of  the  alcohol 
effects  being  examined  in  Drosophila 
at  present  are  responses  to  fairly  high 


27 


NIAAA's  Neurosciencc  and  Behavioral  Research  Portfolio 


doses  of  alcohol,  and  one  challenge  in 
future  studies  will  be  to  develop  behav- 
ioral assays  of  lower  dose  alcohol  effects 
and  alcohol  preference  in  Drosophila. 
The  differences  in  molecular  complexity 
of  potential  alcohol  targets  in 
Drosophila  and  mammals  are  also  a 
concern.  The  diversity  of  subtypes/ 
subunits  of  a  particular  molecule  may 
be  much  greater  in  mammals  than  in 
Drosophila,  and  the  alcohol  sensitivity 
of  these  different  molecules  may  differ 
as  well.  However,  there  is  great  poten- 
tial for  initial  identification  of  classes 
of  molecules  that  contribute  to  acute 
alcohol  sensitivity  using  this  approach. 
Techniques  for  production  of  "ran- 
dom" mutations  in  rodents  are  also 
being  developed,  and  this  approach 
could  potentially  be  used  to  screen  for 
mutations  that  would  alter  acute 
behavioral  and  neuronal  sensitivity  to 
alcohol.  One  such  approach  is  called 
gene  trapping  (Hicks  et  al.  1997). 
This  involves  insertion  of  a  gene  trap 
retroviral  shuttle  vector  into  embry- 
onic stem  (ES)  cells.  Clonal  lines  of 
these  cells  are  then  grown  to  generate 
a  library  of  ES  clones,  each  of  which 
contains  a  single  gene  that  has  incor- 
porated the  retroviral  DNA.  The 
incorporation  of  this  DNA  will  usually 
disrupt  proper  expression  of  that 
gene.  The  expression  vector  codes  for 
a  Neo  resistance  gene  that  can  be 
used  to  select  for  expressed  genes,  and 
it  also  contains  a  PST  sequence  tag 
that  allows  the  disrupted  gene  to  be 
identified  in  the  ES  cells  prior  to 
insertion  into  an  animal.  The  ES  cells 
can  be  inserted  into  a  blastocyst  and 
implanted  into  mice;  if  germ  line  trans- 
mission is  achieved,  then  mice  that 


lack  the  disrupted  gene  can  be  gener- 
ated. This  approach  has  already  been 
used  to  identify  a  number  of  genes 
that  are  disrupted  in  the  ES  cell 
libraries.  In  addition,  several  mutations 
have  been  introduced  into  the  germ  line, 
and  many  of  these  have  identifiable 
phenotypes.  This  approach  is  much 
more  costly  and  time-consuming  than 
mutagenesis  and  screening  of  Dro- 
sophila. Furthermore,  not  too  many 
laboratories  are  currently  using  this 
technique.  An  additional  consideration 
is  that  the  genes  targeted  for  disrup- 
tion using  this  technique  are  those 
expressed  in  ES  cells,  and  thus  the  tech- 
nique may  select  for  genes  involved  in 
early  development.  This  may  lead  to 
several  embryonic  lethal  phenotypes 
and  may  not  allow  disruption  of  genes 
that  play  key  roles  in  alcohol  effects  in 
the  adult  animal.  However,  the  use  of 
rodents  would  allow  investigators  to 
examine  a  wide  variety  of  behavioral 
effects  of  acute  alcohol  in  mutant 
animals  and  to  concentrate  on 
subtle  alterations  in  responses  to  low 
alcohol  doses. 

Screening  of  available  mutant,  trans- 
genic, and  knockout  mice  for  alter- 
ations in  low- dose  alcohol  sensitivity  is 
also  a  possibility.  However,  a  survey  of 
alcohol  effects  on  genetically  altered 
animals  is  not  advisable.  Ultimately, 
this  research  will  best  proceed  from 
hypotheses  generated  from  pharmaco- 
logical and  physiological  studies. 

One  alternative  possibility  would 
be  to  search  for  alcohol- sensitive  protein 
motifs  within  known  proteins  with 
high  alcohol  sensitivity.  Such  proteins 
might  include  alcohol  dehydrogenase 
(Cedergren-Zeppezauer  et  al.  1982; 


Neural  Proteins 


Eklund  et  al.  1982;  Xie  et  al.  1997) 
and  olfactory  receptor  molecules  (Buck 
and  Axel  1991;  Raming  et  al.  1993), 
some  of  which  respond  to  relatively  low 
concentrations  of  airborne  alcohol  (Sato 
et  al.  1994).  A  combination  of  struc- 
tural biological,  molecular  biological 
(e.g.,  chimera  production,  site-directed 
mutagenesis),  and  three-dimensional 
molecular  modeling  approaches  can  be 
used  by  investigators  to  examine  the 
molecular  features  of  the  regions  of 
these  proteins  that  interact  with  alco- 
hol. This  analysis  can  lead  to  a  search 
for  similar  motifs  in  CNS  proteins 
that  may  help  to  identify  proteins  that 
are  particularly  sensitive  to  alcohol. 
This  approach  may  fail  because  of  the 
weak  interactions  of  alcohol  with  even 
the  most  alcohol-sensitive  protein 
sites.  There  may  be  several  hydropho- 
bic molecular  sites  that  interact  with 
alcohol  with  approximately  equal 
"affinity,"  and  thus  it  may  not  be  pos- 
sible to  identify  a  single  alcohol-sensitive 
protein  motif.  However,  searching 
highly  alcohol-sensitive  molecules  has 
the  potential  to  yield  information 
about  the  general  features  of  alcohol 
target  sites. 

SPECIAL  NOTE  ON  MOUSE 
GENETIC  MODELS 

As  can  be  seen  from  the  foregoing 
discussion,  mouse  genetic  models  are 
likely  to  play  an  increasingly  impor- 
tant role  in  studies  of  acute  alcohol 
actions  over  the  next  few  years.  It  will 
be  important  for  alcohol  researchers 
to  have  access  to  mice  with  desired 
genetic  alterations,  to  be  able  to  take 
advantage  of  significant  advances  in 


the  development  of  these  mouse 
models.  In  addition,  researchers  need 
to  be  cognizant  of  problems  associated 
with  the  use  of  genetically  engineered 
mice.  In  this  section  I  briefly  discuss 
these  issues  and  present  some  ideas 
about  how  the  National  Institute  on 
Alcohol  Abuse  and  Alcoholism  can 
encourage  and  support  the  use  of  these 
powerful  techniques  by  investigators 
interested  in  acute  alcohol  actions. 

Several  techniques  are  now  widely 
in  use  for  the  production  of  mice  with 
altered  expression  of  particular  genes. 
Transgenic  mice  are  usually  engi- 
neered to  overexpress  a  particular  gene 
and  its  gene  product  through  insertion 
of  a  genetic  sequence  into  a  mouse 
blastocyst,  which  is  then  implanted  into 
a  pseudopregnant  female  (Faerman  and 
Shard  1997).  The  gene  of  interest  is 
generally  linked  to  a  mammalian  pro- 
moter of  some  type  that  controls  gene 
expression.  Levels  of  expression  of  the 
desired  gene  as  well  as  the  cellular 
locus  of  expression  will  be  controlled 
by  factors  including  the  number  of 
copies  of  the  transgene  expressed  in  a 
particular  mouse  line  and  the  size  and 
identity  of  the  promoter  chosen  to  drive 
expression.  Thus,  two  lines  of  mice 
designed  to  overexpress  the  same  gene 
may  differ  considerably  in  locus  and 
amount  of  actual  protein  expression  and 
hence  in  behavioral  and  cellular  pheno- 
types.  These  factors  must  be  taken 
into  consideration  when  embarking 
on  production  of  transgenic  mice, 
since  the  investigator  most  often 
wishes  to  produce  overexpression  in 
brain  regions  and  at  times  when  the 
gene  is  normally  expressed.  Temporal 
control  of  gene  expression  can  be 


29 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


gained  by  fusing  the  gene  of  interest 
to  a  promoter  that  is  inducible  (e.g., 
by  steroids  or  antibiotics).  One  advan- 
tage of  the  transgenic  approach  is  that 
the  investigator  can  choose  to  produce 
transgenic  lines  on  any  one  of  a  variety 
of  mouse  strain  genetic  backgrounds. 
Thus,  a  strain  with  a  known  phenotype 
can  be  chosen,  and  differences  in  that 
phenotype  produced  by  transgene 
expression  can  be  examined. 

A  powerful  technique  for  examina- 
tion of  the  role  of  a  particular  protein 
in  a  set  of  cellular  and  behavioral  pro- 
cesses is  to  produce  a  line  of  knockout 
mice  that  do  not  express  the  gene  cod- 
ing for  that  protein  (see  Tonegawa  et 
al.  1995  for  a  review).  Production  of 
such  a  line  is  achieved  by  the  gene- 
targeting  technique  using  ES  cells, 
and  takes  advantage  of  homologous 
recombination  events  that  take  place 
during  cell  division.  This  technique 
allows  an  altered  gene  to  be  inserted 
into  the  ES  cell  genome  in  place  of 
the  wild-type  gene  (Capecchi  1989). 
This  substitute  gene  is  engineered  to 
contain  a  marker  driven  by  a  promoter 
that  can  help  to  select  for  the  knockout 
in  the  cells.  In  some  cases  the  wild-type 
gene  is  replaced  with  a  part  of  that  gene 
that  lacks  a  region  needed  for  proper 
expression.  The  stem  cell  containing 
this  altered  gene  is  then  inserted  into 
a  blastocyst  and  implanted  into  a 
pseudopregnant  female  to  give  rise  to 
production  of  genetically  altered  off- 
spring. If  the  altered  gene  makes  its 
way  into  the  germ  line  of  animals  after 
breeding  the  progeny  of  the  female, 
then  the  replacement  gene  can  be 
propagated  through  breeding  and 
eventually  mice  that  are  homo-  and 


heterozygotic  for  the  gene  knockout 
can  be  produced.  This  technique  has 
already  been  used  by  several  investigators 
in  the  alcohol  research  field  (Harris  et 
al.  1995;  Crabbe  et  al.  1996;  Homanics 
et  al.  1997;  Miyakawa  et  al.  1997; 
Rubinstein  et  al.  1997).  For  more 
information  on  these  techniques  and 
their  application  to  neuroscience 
research,  I  refer  the  reader  to  a  review 
by  Chen  and  Tonegawa  (1997). 

Having  an  animal  that  does  not 
express  a  particular  protein  is,  of  course, 
valuable  to  a  researcher  with  an  interest 
in  determining  the  role  of  that  protein 
in  a  behavior  such  as  alcohol  intoxica- 
tion or  a  cellular  response  such  as 
synaptic  inhibition  by  alcohol.  How- 
ever, the  standard  mouse  gene  knock- 
out approach  is  not  without  its 
drawbacks.  The  fact  that  most  homo- 
2ygotic  knockout  animals  never  express 
the  gene  of  interest  at  any  point  dur- 
ing development  can  lead  to  develop- 
mental abnormalities.  Thus,  altered 
phenotypes  may  be  a  secondary  or  even 
tertiary  consequence  of  the  absence  of 
the  gene  of  interest.  In  addition,  homo- 
logous proteins  that  can  compensate 
for  the  missing  protein  may  become 
overexpressed  in  the  animal  during 
development.  This  can  lead  to  a 
"false-negative"  lack  of  phenotype 
that  can  be  too  easily  interpreted  as 
showing  no  important  role  for  the 
protein  of  interest.  These  problems 
have  been  discussed  in  reviews  of  the 
subject  (e.g.,  Joyner  1994). 

To  avoid  these  problems  and  the 
large  numbers  of  control  experiments 
necessitated  by  them,  investigators  have 
begun  to  develop  approaches  that  will 
allow  gene  knockout  at  any  time  during 


30 


Neural  Proteins 


development.  This  inducible  knockout 
technique  potentially  allows  the  animals 
to  develop  normally  until  the  time  at 
which  the  investigator  decides  to  remove 
the  gene  of  interest.  The  gene  can  then  be 
eliminated  and  the  animals  can  be  exam- 
ined shortly  thereafter  to  determine  the 
effects  of  losing  gene  expression  prior 
to  any  substantial  compensation.  The 
most  popular  current  technique  for  pro- 
ducing such  mice  is  the  cre/lox  tech- 
nique (Marth  1996).  This  technique 
takes  advantage  of  the  fact  that  the  ere 
recombinase  will  excise  genes  that  are 
flanked  by  the  loxV  signal  sequence.  Thus, 
one  can  create  mice  with  the  loxV 
sequences  surrounding  a  gene  by  homo- 
logous recombination,  and  then  breed 
these  animals  with  transgenic  mice  that 
express  the  ere  gene  driven  by  a  suitable 
promoter.  If  the  ere  gene  is  fused  with  a 
hormone  or  antibiotic  binding  domain 
such  the  human  estrogen  receptor  (Feil 
et  al.  1996),  then  ere  expression  can  be 
induced  in  the  cre/lox  animals,  leading 
to  temporally  controlled  gene  knockout. 
This  technique  can  also  help  to  elimi- 
nate another  problem  with  standard 
knockout  technology,  namely,  the  fact 
that  gene  expression  is  eliminated 
throughout  the  brain  and  maybe  even 
throughout  the  entire  body.  By  driving 
ere  expression  with  a  tissue-  or  cell-specific 
promoter,  the  cre/lox  technique  can  be 
adapted  to  generate  tissue-  or  brain 
region-specific  knockouts  by  breeding 
"floxed"  mice  with  those  expressing 
the  ere  transgene  only  in  a  specific  set 
of  neurons  (Tsien  et  al.  1996). 

An  additional  problem  that  arises 
in  examining  knockout  mice  is  the 
problem  of  strain  differences  in  pheno- 
type  (Gerlai  1996;  Crawley  et  al.  1997). 


As  we  in  the  alcohol  research  field  know 
all  too  well,  different  mouse  strains 
exhibit  different  behavioral  pheno- 
types,  and  these  differences  can  also 
extend  to  physiological  processes. 
This  factor  must  be  taken  into  account 
when  designing  experiments  to  examine 
phenotypes  of  knockout  mice.  The  stem 
cells  currently  available  for  creation  of 
knockouts  are  derived  from  the  129/ 
Sv  mouse  line,  and  thus  the  initial 
knockout  animals  will  necessarily  have 
this  genetic  background.  These  animals 
are  known  to  drink  alcohol  and  to 
show  normal  sensitivity  to  acute  alco- 
hol actions  (Crabbe  et  al.  1996),  and 
these  behaviors  can  be  altered  by 
knockouts  on  this  genetic  background. 
However,  the  factors  that  control 
alcohol  consumption  in  this  strain  of 
animals  in  comparison  with  better  char- 
acterized strains  are  relatively  unclear. 
An  additional  problem  with  this  mouse 
strain  is  the  relatively  small  average  lit- 
ter size,  which  may  make  it  difficult  to 
perform  within-litter  comparisons.  A 
strategy  that  can  be  used  to  overcome 
this  problem  is  to  outbreed  the  initial 
knockout  mice  with  another  strain  of 
mouse  with  a  more  desirable  back- 
ground phenotype  until  mice  are 
obtained  that  are  congenic  with  the 
desired  strain  and  still  lack  the  wild-type 
gene.  These  animals  can  then  be  com- 
pared with  wild-type  animals  of  the 
same  strain.  However,  this  can  take  sev- 
eral generations  of  breeding.  Misinter- 
pretation of  phenotypic  characteristics 
is  quite  possible  if  the  knockout  animals 
are  not  genetically  homogeneous,  in 
that  a  wild-type  strain  with  suitable 
genetic  background  for  comparison 
with  the  knockout  mice  may  not  be 


31 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


available.  More  attention  needs  to  be 
paid  to  the  genetic  background  of  the 
wild-type  and  mutant  mice  in  future 
studies  of  alcohol  effects  using  mouse 
genetic  models.  In  the  future  it  may 
be  possible  to  use  stem  cells  that  can  be 
implanted  into  a  more  commonly  used 
mouse  strain.  However,  these  cells  are 
not  yet  widely  available.  A  recent 
report  suggests  that  interactions 
between  genetics  and  laboratory  envi- 
ronment can  also  influence  alcohol's 
effects  in  mice  (Crabbe  et  al.  1999). 

An  alternative  to  production  of  true 
knockout  mice  is  production  of  mice 
that  express  a  particular  protein  that 
has  been  subtly  mutated  in  a  region 
that  is  thought  to  be  important  for  a 
particular  function.  For  example,  an 
amino  acid  residue  thought  to  be 
important  for  alcohol  sensitivity  of  a 
particular  protein  might  be  mutated, 
and  a  receptor  containing  this  mutation 
could  be  expressed  in  a  genetically 
engineered  mouse.  This  is  possible 
using  techniques  that  take  advantage 
of  homologous  recombination,  such 
as  the  "tag-and-exchange"  (Askew  et 
al.  1993)  and  "hit-and-run" 
(Ramirez-Solis  et  al.  1993,  MacMillan 
et  al.  1996)  techniques.  Since  the 
mutated  receptor  coding  region  can  be 
substituted  for  that  of  the  wild-type 
receptor,  this  technique  theoretically 
allows  the  investigator  to  produce 
mice  that  retain  expression  of  a  protein, 
but  with  a  single-point  mutation.  This 
technique  should  allow  researchers  to 
determine  the  importance  of  a  region 
of  a  protein  or  a  single  amino  acid  in 
an  alcohol  response  within  the  context 
of  an  intact  neuron  or  an  intact  animal. 
This  is  a  potentially  powerful  approach 


that  may  be  free  of  many  of  the  unde- 
sirable consequences  of  true  knockout 
approaches.  For  example,  removing 
expression  of  some  genes  and  inser- 
tion of  a  novel  promoter  in  the  stan- 
dard knockout  strategy  can  affect 
expression  of  other  genes,  and  this 
can  be  avoided  by  substitution  of  a 
gene  that  has  only  a  subtie  mutation. 
Furthermore,  the  tag-and-exchange 
approach  allows  the  mutated  gene  to 
be  substituted  into  a  traditional 
knockout  animal.  This  allows  for 
investigation  of  both  the  knockout 
and  subtle  mutation  phenotypes,  and 
may  allow  for  gene  rescue  in  knock- 
outs. However,  care  must  be  taken  to 
ensure  that  the  mutation  of  the  pro- 
tein does  not  disrupt  its  expression 
such  that  the  animal  inadvertently 
becomes  a  "functional  knockout" 
(e.g.,  as  in  Lakhlani  et  al.  1997). 

It  will  also  be  important  for  investi- 
gators using  these  techniques  to 
attempt  to  use  approaches  to  rescue 
the  knockout  phenotype  by  reintro- 
ducing the  gene  of  interest  in  the 
knockout  mice.  This  could  be  accom- 
plished by  the  tag-and-exchange  tech- 
nique or  by  overexpression  with  a 
transgene  (Iwasato  et  al.  1997).  Tech- 
niques for  inducing  expression  of  a 
rescue  gene  should  also  become  avail- 
able in  the  not- too -distant  future. 

Researchers  interested  in  acute 
actions  of  alcohol  must  be  poised  not 
only  to  use  mouse  genetic  models  but 
also  to  take  advantage  of  the  next 
generation  of  approaches  to  creation 
of  these  mouse  models.  Developing 
research  strategies  for  the  use  of  these 
mice  that  avoid  the  confounds  dis- 
cussed earlier  will  help  prevent  the 


32 


Neural  Proteins 


field  from  becoming  bogged  down 
with  issues  of  compensation  and 
genetic  background  when  interpreting 
data  from  knockout  mice. 

An  additional  problem  faced  by 
alcohol  researchers  anxious  to  use 
genetically  altered  mice  is  the  limited 
availability  of  mice  that  were  originally 
created  for  other  research  purposes. 
These  mice  often  cannot  be  obtained 
by  researchers  with  an  interest  in 
acute  alcohol  effects.  One  important 
step  in  solving  this  problem  would  be 
to  encourage  more  investigators  with 
experience  in  the  creation  of  geneti- 
cally engineered  mice  to  enter  the 
alcohol  research  field.  This  could  be 
done  by  a  request  for  applications 
(RFA)  or  other  grant  submission 
process  that  would  call  for  proposals 
aimed  at  the  creation  of  mouse  lines 
for  alcohol  research,  or  the  use  of 
existing  mouse  lines  for  alcohol 
research.  One  possible  structure  of 
such  a  program  would  be  to  encour- 
age investigators  with  the  skills  to  cre- 
ate the  mouse  lines  to  team  up  with 
investigators  who  can  assess  behav- 
ioral and  cellular  actions  of  alcohol  in 
the  mice.  Encouraging  the  use  of 
genetically  engineered  mice  in  alcohol 
research  should  be  a  high  priority  in 
coming  years. 

In  addition  to  mice  in  which  the 
genome  is  explicitly  altered  to  express  or 
not  express  a  particular  protein,  alcohol 
researchers  can  take  advantage  of 
recombinant  inbreeding  approaches 
to  create  animals  in  which  small  areas 
of  the  genome  differ  that  give  rise  to 
differences  in  alcohol -related  behav- 
ioral phenotypes  as  discussed  earlier. 
These  animals  can  be  examined  at  the 


cellular  and  molecular  level  to  gain 
more  information  about  the  differ- 
ences that  underlie  the  differing 
behavioral  phenotypes.  Proteins  that 
are  coded  for  by  candidate  genes 
identified  by  genetic  analyses,  such  as 
QTL  screening,  may  be  targets  for 
examination  in  these  analyses. 

REFERENCES 

Akabas,  M.H.;  Kaufmann,  C;  Archdeacon, 
P.;  and  Karlin,  A.  Identification  of  acetyl- 
choline receptor  channel -lining  residues 
in  the  entire  M2  segment  of  the  alpha 
subunit.  Neuron  13(4):919-927,  1994. 

Akabas,  M.H.;  StaufFer,  D.A.;  Xu,  M.;  and 
Karlin,  A.  Acetylcholine  receptor  channel 
structure  probed  in  cysteine  substitution 
mutants.  Science 258:307-310,  1995. 

Altenbach,  C;  Yang,  K.;  Farrens,  D.L.; 
Farahbakhsh,  Z.T.;  Khorana,  H.G.;  and 
Hubbell,  W.L.  Structural  features  and 
light-dependent  changes  in  the  cytoplasmic 
interhelical  E-F  loop  region  of  rhodopsin: 
A  site-directed  spin-labeling  study. 
Biochemistry  35:12470-12478,  1996. 

Askew,  G.R.;  Doetschman,  T.;  and  Lingrel, 
J.B.  Site-directed  point  mutations  in 
emrbryonic  stem  cells:  A  gene -targeting 
tag-and-exchange  strategy.  Mol  Cell  Biol 
13:4115^124,1993. 

Baertschi,  A.J.  Antisense  oligonucleotide 
strategies  in  physiology.  Mol  Cell 
Endocrinol  101(1-2):R15-R24,  1994. 

Bear,  M.F.,  and  Malenka,  R.C.  Synaptic 
plasticity:  LTP  and  LTD.  Curr  Opin 
Neurobiol  4(3):3S9-399,  1994. 

Bechinger,  B.;  Kim,  Y.;  Chirlian,  L.E.; 
Gesell,  J.;  Neumann,  J.;  Montal,  M.; 
Tomich,  J.;  ZaslofF,  M.;  and  Opella,  SJ. 
Orientation  of  amphipathic  helical  peptides 


33 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  membrane  bilayers  determined  by 
solid-state  NMR  spectroscopy.  J  Biomol 
NMR  1:  167-173,  1991. 

Beechem,  J.M.  Picosecond  fluorescence 
decay  curves  collected  on  millisecond  time 
scale:  Direct  measurement  of  hydrodynamic 
radii,  local/global  mobility,  and  intramole- 
cular distances  during  protein-folding  reac- 
tions. Methods  Ewzymo/ 278:24-49,  1997. 

Bekkers,  J.M.,  and  Stevens,  C.F.  The 
nature  of  quantal  transmission  at  central 
excitatory  synapses.  Adv  Second  Messenger 
Phosphoprotein  Res  29:261-273,  1994. 

Belknap,  J.K.;  Richards,  S.P.;  O'Toole, 
L.A.;  Helms,  M.L.;  and  Phillips,  T.J.  Short- 
term  selective  breeding  as  a  tool  for  QTL 
mapping:  Ethanol  preference  drinking  in 
mice.  Behav  Genet  27(l):55-66,  1997. 

Bennett,  C.F.  Antisense  oligonucleotides: 
Is  the  glass  half  full  or  half  empty?  Biochem 
Pharmacol  55(l):9-l9,  1998. 

Blanton,  M.P.,  and  Cohen,  J.B.  Identifying 
the  lipid-protein  interface  of  the  Torpedo 
nicotinic  acetylcholine  receptor:  Secondary 
structure  implications.  Biochemistry  33(10): 
2859-2872,  1994. 

Bliss,  T.V.,  and  Collingridge,  G.L.  A 
synaptic  model  of  memory:  Long-term 
potentiation  in  the  hippocampus.  Nature 
361(6407):31-39,  1993. 

Blitzer,  R.D.;  Gil,  O.;  and  Landau,  E.M. 
Long-term  potentiation  in  rat  hippocampus 
is  inhibited  by  low  concentrations  of  ethanol. 
Brain  Res  537 (l-2):203-208,  1990. 

Borst,  J.G.,  and  Sakmann,  B.  Calcium 
influx  and  transmitter  release  in  a  fast 
CNS  synapse.  Nature  383(6599): 
431^34,  1996. 

Brodie,  M.S.;  Trifunovic,  R.D.;  and 
Shefner,  S.A.  Serotonin  potentiates 
ethanol-induced  excitation  of  ventral 


tegmental  area  neurons  in  brain  slices 
from  three  different  rat  strains.  J  Pharmacol 
Exp  Ther273(3):l  139-1 146,  1995. 

Bruns,  D.;  Engert,  F.;  and  Lux,  H.D.  A 
fast  activating  presynaptic  reuptake  current 
during  serotonergic  transmission  in  iden- 
tified neurons  of  Hirudo.  Neuron  10(4): 
559-572,  1993. 

Buck,  K.J.;  Metten,  P.;  Belknap,  J.K.;  and 
Crabbe,  J.C.  Quantitative  trait  loci  involved 
in  genetic  predisposition  to  acute  alcohol 
withdrawal  in  mice.  /  Neurosci  17(10): 
3946-3955,  1997. 

Buck,  L.,  and  Axel,  R.  A  novel  multigene 
family  may  encode  odorant  receptor:  A 
molecular  basis  for  odor  recognition.  Cell 
65:175-187,  1991. 

Capecchi,  M.R.  Altering  the  genome  by 
homologous  recombination.  Science  244: 
1288-1292,  1989. 

Carr,  D.W.,  and  Scott,  J.D.  Blotting  and 
band-shifting:  Techniques  for  studying 
protein-protein  interactions.  Trends 
Biochem  Sci  17(7):246-249,  1992. 

Cedergren-Zeppezauer,  E.;  Samama,  J. P.; 
and  Eklund,  H.  Crystal  structure  determina- 
tions of  coenzyme  analogue  and  substrate 
complexes  of  liver  alcohol  dehydrogenase: 
Binding  of  1,4,5,6-tetrahydronicotinamide 
adenine  dinucleotide  and  trans-4-(N,N- 
dimethylamino)cinnamaldehyde  to  the 
enzyme.  Biochemistry  21(20):4895- 
4908, 1982. 

Chen,  C,  and  Tonegawa,  S.  Molecular 
genetic  analysis  of  synaptic  plasticity,  activity- 
dependent  neural  development,  learning, 
and  memory  in  the  mammalian  brain. 
Annu  Rev  Neurosci  20:157-184,  1997. 

Choi,  S.,  and  Lovinger,  D.M.  Decreased 
frequency  but  not  amplitude  of  quantal 
synaptic  responses  associated  with  expression 


34 


Neural  Proteins 


of  corticostriatal  long-term  depression.  / 
Neurosci  17(21):8613-8620,  1997. 

Clements,  J.D.  A  statistical  test  for 
demonstrating  a  presynaptic  site  of  action 
for  a  modulator  of  synaptic  amplitude.  / 
Neurosci  Methods  31(  1  ):75-88,  1990. 

Coultrap,  S.J.,  and  Machu,  T.K. 
Mutation  of  putative  phosphorylation 
sites  on  the  5-HT3  receptor  does  not 
eliminate  its  modulation  by  ethanol. 
Alcohol  Clin  Exp  Res 21(3):7A,  1997. 

Covarrubias,  M.;  Vyas,  T.B.;  Escobar,  L.; 
and  Wei,  A.  Alcohols  inhibit  a  cloned 
potassium  channel  at  a  discrete  saturable 
site.  J  Biol  Chem  270(33):19408-19416, 
1995. 

Cowan,  S.W.;  Schirmer,  T.;  Rummel,  G.; 
Seifert,  M.;  Ghosh,  R.;  Pauptit,  RA.; 
Jansonius,  J.N.;  and  Rosenbusch,  J. P. 
Crystal  structures  explain  functional  prop- 
erties of  two  E.  coli  porins.  Nature 
358:727-733,  1992. 

Crabbe,  J.C.;  Phillips,  T.J.;  Feller,  D.J.; 
Hen,  R;  Wenger,  CD.;  Lessov,  C.N.; 
and  Schafer,  G.L.  Elevated  alcohol  con- 
sumption in  null  mutant  mice  lacking  5- 
HT1B  serotonin  receptors.  Nat  Genet 
14(1):98-101,  1996. 

Crabbe,  J.C.;  Wahlsten,  D.;  and  Dudek, 
B.C.  Genetics  of  mouse  behavior: 
Interactions  with  laboratory  environment. 
Science  284(5420):1670-1672,  1999. 

Crawley,  J.N.;  Belknap,  J.K.;  Collins,  A.; 
Crabbe,  J.C.;  Frankel,  W.;  Henderson, 
N.;  Hitzemann,  R.J.;  Maxson,  S.C.; 
Miner,  L.L.;  Silva,  A.J.;  Wehner,  J.M.; 
Wynshaw- Boris,  A.;  and  Paylor,  R. 
Behavioral  phenotypes  of  inbred  mouse 
strains:  Implications  and  recommenda- 
tions for  molecular  studies.  Psychopharma- 
cology  (Berl)  132(2):107-124,  1997. 


Criado,  J.R;  Lee,  R.S.;  Berg,  G.L;  and 
Henriksen,  S.J.  Sensitivity  of  nucleus 
accumbens  neurons  in  vivo  to  intoxicating 
doses  of  ethanol.  Alcohol  Clin  Exp  Res 
19(1):164-169,  1995. 

Criado,  J.R.;  Steffensen,  S.C.;  and 
Henriksen,  S.J.  Microelectrophoretic 
application  of  SCH-23390  into  the 
lateral  septal  nucleus  blocks  ethanol- 
induced  suppression  of  LTP,  in  vivo,  in 
the  adult  rodent  hippocampus.  Brain 
Res  7\6{l-2):\92-\96,  1996. 

Criswell,  H.E.;  McCown,  T.J.;  Morrow, 
A.L.;  and  Breese,  G.R  GABA  receptors  on 
SNR  neurons  which  respond  to  low  concen- 
trations of  Zolpidem  also  respond  to  ethanol 
and  contain  mRNA  for  the  al  subunit. 
Alcohol  Clin  Exp  ito20(2):10A,  1996. 

Czajkowski,  C,  and  Karlin,  A.  Structure 
of  the  nicotinic  receptor  acetylcholine 
binding  site — identification  of  acidic 
residues  in  the  d  subunit  within  0.9  nm 
of  the  a  subunit  binding  site  disulfide.  / 
Biol  Chem  270:3160-3164,  1995. 

DeFelice,  L.J.,  and  Galli,  A  Electrophysio- 
logical analysis  of  transporter  function. 
Adv  Pharmacol  42:186-190,  1998. 

Dell'Aqua,  MX.,  and  Scott,  J.D.  Protein 
kinase  A  anchoring.  J  Biol  Chem  272: 
12881-12884,  1997. 

Denk,  W.;  Yuste,  R;  Svoboda,  K.;  and 
Tank,  D.W.  Imaging  calcium  dynamics  in 
dendritic  spines.  Curr  Opin  Neurobiol 
6(3):372-378,  1996. 

Diamond,  I.,  and  Gordon,  A.S.  Cellular 
and  molecular  neuroscience  of  alcoholism. 
Physiol  Rev  77(1):  1-20,  1997. 

Dildy-Mayfield,  J.E.,  and  Harris,  RA. 
Ethanol  inhibits  kainate  responses  of 
glutamate  receptors  expressed  in  Xenopus 
oocytes:  Role  of  calcium  and  protein  kinase 
C.J  Neurosci  15(4):3162-3171,  1995. 


35 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


dos  Remedios,  C.G.,  and  Moens,  P.D. 
Fluorescence  resonance  energy  transfer 
spectroscopy  is  a  reliable  "ruler"  for  mea- 
suring structural  changes  in  proteins. 
Dispelling  the  problem  of  the  unknown 
orientation  factor.  J  Struct  Biol  115(2): 
175-185,  1995. 

Doyle,  D.A.;  Morais  Cabral,  J.;  Pfuetzner, 
R.A.;  Kuo,  A.;  Gulbis,  J.M.;  Cohen,  S.L.; 
Chait,  B.T.;  and  MacKinnon,  R  The  struc- 
ture of  the  potassium  channel:  Molecular 
basis  of  K+  conduction  and  selectivity. 
Science  280(5360):69-77,  1998. 

Dutar,  P.,  and  Nicoll,  R.A.  Pre-  and  post- 
synaptic GABAB  receptors  in  the  hippo- 
campus have  different  pharmacological 
properties.  Neuron  1(7):585-591,  1988. 

Eklund,  H.;  Plapp,  B.V.;  Samama,  J.P.; 
and  Branden,  C.I.  Binding  of  substrate  in 
a  ternary  complex  of  horse  liver  alcohol 
dehydrogenase.  J  Biol  Chem  257(23): 
14349-14358,  1982. 

Faerman,  A.,  and  Shani,  M.  Transgenic 
mice:  Production  and  analysis  of  expression. 
Methods  Cell  Biol  52:373-403,  1997. 

Faux,  M.D.,  and  Scott,  J.D.  Regulation  of 
the  AKAP79 -protein  kinase  C  interaction 
by  Ca2+/calmodulin.  J  Biol  Chem  272: 
17038-17044,  1997. 

Feil,  R.;  Brocard,  J.;  Mascrez,  B.;  LeMeur, 
M.;  Metzger,  D.;  and  Chambon,  P. 
Ligand-activated  site-specific  recombina- 
tion in  mice.  Proc  Natl  Acad  Sci  USA 
93:10887-10890,  1996. 

Fields,  S.,  and  Sternglanz,  R.  The  two- 
hybrid  system:  An  assay  for  protein-protein 
interactions.  Trends  Genet  10(8):286- 
292, 1994. 

Galzi,  J.L.;  Revah,  F.;  Black,  D.;  Goeldner, 
M.;  Hirth,  C;  and  Changeux,  J. -P.  Identi- 
fication of  a  novel  amino  acid  a-Tyr93 
within  the  active  site  of  the  acetylcholine 


receptor  by  photoaffinity  labelling: 
Additional  evidence  for  a  three  loop 
model  of  the  acetylcholine  binding  site.  / 
Biol  Chem  265:10430-10437,  1990. 

Gerlai,  R.  Gene-targeting  studies  of  mam- 
malian behavior:  Is  it  the  mutation  or  the 
background  genotype?  Trends  Neurosci 
19:177-181,  1996. 

Gessa,  G.L.;  Muntoni,  F.;  Collu,  M.; 
Vargiu,  L.;  and  Mereu,  G.  Low  doses  of 
ethanol  activate  dopaminergic  neurons  in 
the  ventral  tegmental  area.  Brain  Res 
348(l):201-203,  1985. 

Givens,  B.  Behavioral  correlates  of  single 
units  in  the  medial  septal  area:  The  effect 
of  ethanol.  Neuroscience  71(2):417-427, 
1996. 

Goda,  Y.,  and  Stevens,  C.F.  Synaptic 
plasticity:  The  basis  of  particular  types  of 
learning.  Curr  Biol6(4):375-S7S,  1996. 

Gordon,  A.S.;  Yao,  L.;  Wu,  Z.L.;  Coe,  I.R.; 
and  Diamond,  I.  Ethanol  alters  the  sub- 
cellular localization  of  delta-  and  epsilon 
protein  kinase  C  in  NG108-15  cells.  Mol 
Pharmacol  52(4):554-559,  1997. 

Green,  T.;  Stauffer,  K.A.;  and  Lummis, 
S.C.R.  Expression  of  recombinant  homo- 
oligomeric  5-hydroxytryptamine3  recep- 
tors provides  new  insights  into  their 
maturation  and  structure.  J  Biol  Chem 
270(11):6056-6061,  1995. 

Harris,  R.A.;  McQuilkin,  S.J.;  Paylor,  R; 
Abeliovich,  A.;  Tonegawa,  S.;  and 
Wehner,  J.M.  Mutant  mice  lacking  the 
gamma  isoform  of  protein  kinase  C  show 
decreased  behavioral  actions  of  ethanol 
and  altered  function  of  gamma-aminobu- 
tyrate  type  A  receptors.  Proc  Natl  Acad 
Sci  USA  92:3658-3662,  1995. 

Hicks,  G.G.;  Shi,  E.;  Li,  X.-M.;  Li, 
C.-H.;  Pawlak,  M.;  and  Ruley,  H.E. 
Functional  genomics  in  mice  by  tagged 


36 


Neural  Proteins 


sequence  mutagenesis.  Nat  Genet 
16:338-344,  1997. 

Homanics,  G.E.;  Ferguson,  C;  Quinlan, 
J.J.;  Daggett,  J.;  Snyder,  K.;  Lagenaur, 
C;  Mi,  Z.P.;  Wang,  X.H.;  Grayson,  D.R.; 
and  Firestone,  L.L.  Gene  knockout  of  the 
alpha6  subunit  of  the  gamma- aminobu- 
tyric  acid  type  A  receptor:  Lack  of  effect 
on  responses  to  ethanol,  pentobarbital, 
and  general  anesthetics.  Mol  Pharmacol 
51(4):588-596,  1997. 

Hovius,  R.;  Tairi,  A.-P.;  Blasey,  H.; 
Bernard,  A.;  Lundstrom,  K.;  and  Vogel, 
H.  Characterization  of  a  mouse  serotonin 
5-HT3  receptor  purified  from  mammalian 
cells.  J  Neurocbem  70(2):824-834,  1998. 

Hubbell,  W.L.,  and  Altenbach,  C.  Site 
directed  spin-labeling  of  membrane  proteins. 
In:  White,  S.H.,  ed.  Membrane  Protein 
Structure:  Experimental  Approaches.  Oxford, 
England:  Oxford  University  Press,  1994. 
pp.  224-248. 

Hucho,  F.;  Tsetlin,  V.I.;  and  Machold,  J. 
The  emerging  three-dimensional  struc- 
ture of  a  receptor.  The  nicotinic  acetyl- 
choline receptor.  Eur  J  Biochem 
239(3):539-557,  1996. 

Hundle,  B.;  McMahon,  T.;  Dadgar,  J.; 
Chen,  C.-H.;  Mochly-Rosen,  D.;  and 
Messing,  R.O.  An  inhibitory  fragment 
derived  from  protein  kinase  C  prevents 
enhancement  of  nerve  growth  factor 
responses  by  ethanol  and  phorbol  esters.  / 
Biol  Chem  272(23):15028-15035,  1997. 

Isaac,  J.T.;  Oliet,  S.H.;  Hjelmstad,  G.O.; 
Nicoll,  R.A.;  and  Malenka,  RC. 
Expression  mechanisms  of  long-term 
potentiation  in  the  hippocampus.  J  Physiol 
Paris  90(5-6):299-303,  1996. 

Iwasato,  T.;  Erzurumlu,  RS.;  Huerta,  P.T.; 
Chen,  D.F.;  Sasaoka,  T.;  Ulupinar,  E.;  and 
Tonegawa,  S.  NMDA  receptor-dependent 


refinement  of  somatotopic  maps.  Neuron 
19(6):1201-1210,  1997. 

Joyner,  A.L.  Gene  targeting  and  develop- 
ment of  the  nervous  system.  Curr  Opin 
Neurobiol  4:37-42,  1994. 

Keniry,  M.;  Gutowsky,  J.;  and  Oldfield, 
E.  Surface  dynamics  of  the  integral  mem- 
brane protein  bacteriorhodopsin.  Nature 
307:383-386,  1984. 

Khan,  Z.U.;  Gutierrez,  A.;  and  De  Bias, 
A.L.  The  subunit  composition  of  a 
GABAA/benzodiazepine  receptor  from 
rat  cerebellum.  /  Neurochem 
63(l):371-374,  1994. 

Koob,  G.F.,  and  Nestler,  E.J.  The  neuro- 
biology of  drug  addiction.  J  Neuropsy- 
chiatry Clin  Neurosci  9(3)  A82-497,  1997. 

Korn,  H.,  and  Faber,  D.S.  Quantal  analy- 
sis and  synaptic  efficacy  in  the  CNS. 
Trends  Neurosci  14(10):439-445, 1991. 

Kornau,  H.C.;  Seeburg,  P.H.;  and  Kennedy, 
M.B.  Interaction  of  ion  channels  and 
receptors  with  PDZ  domain  proteins.  Curr 
Opin  Neurobiol7(3):368-373,  1997. 

Krishnan,  K.S.,  and  Nash,  HA.  A  genetic 
study  of  the  anesthetic  response:  Mutants 
of  Drosophila  melanogaster  altered  in 
sensitivity  to  halothane.  Proc  Natl  Acad 
Sci  USA  87(21  ):8632-8636,  1990. 

Kuner,  T.;  Schoepfer,  R;  and  Korpi,  E.R 
Ethanol  inhibits  glutamate-induced  currents 
in  heteromeric  NMDA  receptor  subtypes. 
Neuroreport  5(3):297-300, 1993. 

Lakhlani,  P.P.;  MacMillan,  L.B.;  Guo,  T.Z.; 
McCool,  B.A.;  Lovinger,  D.M.;  Maze,  M.; 
and  Limbird,  L.E.  Substitution  of  a  mutant 
alphaZa-adrenergjc  receptor  via  "hit  and 
run"  gene  targeting  reveals  the  role  of  this 
subtype  in  sedative,  analgesic,  and  anes- 
thetic-sparing responses  in  vivo.  Proc  Natl 
Acad  Sci  U&4  94(18):9950-9955, 1997. 


37 


NIAAA's  Neurosciencc  and  Behavioral  Research  Portfolio 


Lee,  R.S.;  Smith,  S.S.;  Chapin,  J.K.; 
Shimizu,  N.;  Waterhouse,  B.D.;  Maddus, 
B.N.;  and  Woodward,  D.J.  Effects  of 
systemic  and  local  ethanol  on  responses  of 
rat  cerebellar  Purkinje  neurons  to  ion- 
tophoretically  applied  norepinephrine  and 
gamma- aminobutyric  acid.  Brain  Res 
687(1-2):12-21,  1995. 

Leibovitch,  B  A.;  Campbell,  D.B.;  Krishnan, 
K.S.;  and  Nash,  HA.  Mutations  that  affect 
ion  channels  change  the  sensitivity  of 
Drosophila  melanogaster  to  volatile  anes- 
thetics. J  Neurogenet  10(1):1-13,  1995. 

Lin,  M.,  and  Nash,  HA.  Influence  of 
general  anesthetics  on  a  specific  neural 
pathway  in  Drosophila  melanog aster.  Proc 
Natl  Acad  Sci  USA  93(19):  10446- 
10451,  1996. 

Linden,  DJ.  Long-term  synaptic  depres- 
sion in  the  mammalian  brain.  Neuron  12(3): 
457-472, 1994. 

Linden,  D.J.,  and  Connor,  JA.  Long-term 
synaptic  depression.  Annu  Rev  Neurosci 
18:319-357,  1995. 

Liu,  G.,  and  Tsien,  R.W.  Variable  size  of 
synaptic  events  at  single  hippocampal 
synapses  is  due  to  variation  of  transmitter 
concentration  in  the  synaptic  cleft.  Soc 
Neurosci  Abstr23(2):2286,  1997. 

Lovinger,  D.M.  High  ethanol  sensitivity 
of  recombinant  AMPA-type  glutamate 
receptors  expressed  in  mammalian  cells. 
Neurosci  Lett  159(l-2):83-87,  1993. 

Lovinger,  D.M.  Developmental  decrease 
in  ethanol  sensitivity  of  NMDA  receptors 
on  cortical  neurons:  Relation  to  the  ac- 
tions of  ifenprodil.  J  Pharmacol  Exp  Ther 
274(1):164-172,  1995. 

Lovinger,  D.M.  Alcohols  and  neurotrans- 
mitter gated  ion  channels:  Past,  present 
and  future.  Naunyn  Schmiedebergs  Arch 
Pharmacol  356:267-282,  1997. 


Ludvig,  N.;  Altura,  B.T.;  Fox,  S.E.;  and 
Altura,  B.M.  The  suppressant  effect  of 
ethanol,  delivered  via  intrahippocampal 
microdialysis,  on  the  firing  of  local  pyra- 
midal cells  in  freely  behaving  rats.  Alcohol 
12(5):417^L21,  1995. 

MacKinnon,  R,  and  Doyle,  DA. 
Prokaryotes  offer  hope  for  potassium 
channel  structural  studies.  Nat  Struct 
£w/4(ll):877-879,  1997. 

MacMillan,  L.B.;  Hein,  L.;  Smith,  M.S.; 
Piascik,  M.T.;  and  Limbird,  L.E.  Central 
hypotensive  effects  of  the  alpha2a-adren- 
ergic  receptor  subtype.  Science  273(5276): 
801-803,1996. 

Malenka,  R.C.,  and  Nicoll,  R.A.  Silent 
synapses  speak  up.  Neuron  19(3):473- 
476, 1997. 

Malgaroli,  A.;  Ting,  A.E.;  Wendland,  B.; 
Bergamaschi,  A.;  Villa,  A.;  Tsien,  R.W.; 
and  Scheller,  R.H.  Presynaptic  component 
of  long-term  potentiation  visualized  at 
individual  hippocampal  synapses.  Science 
268(5217):1624-1628,  1995. 

Marszalec,  W.;  Kurata,  Y.;  Hamilton,  B.J.; 
Carter,  D.B.;  and  Narahashi,  T.  Selective 
effects  of  alcohols  on  gamma-aminobutyric 
acid  A  receptor  subunits  expressed  in  hu- 
man embryonic  kidney  cells.  J  Pharmacol 
Exp  Ther  269:157-163,  1994. 

Marth,  J.D.  Molecular  medicine  in  genet- 
ically engineered  animals:  Recent  advances 
in  gene  mutagenesis  by  site-directed 
recombination.  /  Clin  Invest  97(9) -.1999- 
2002,  1996. 

Mascia,  M.P.;  Mihic,  S.J.;  Valenzuela, 
C.F.;  Schofield,  P.R.;  and  Harris,  R.A.  A 
single  amino  acid  determines  differences 
in  ethanol  actions  on  strychnine-sensitive 
glycine  receptors.  Mol  Pharmacol  50(2): 
402-406,  1996. 


38 


Neural  Proteins 


Masood,  K.;  Wu,  C;  Brauneis,  U.;  and 
Weight,  F.F.  Differential  ethanol  sensitivity 
of  recombinant  N-methyl-D-aspartate 
receptor  subunits.  Mol  Pharmacol  45:324- 
329,  1994. 

Matthews,  D.B.;  Simson,  P.E.;  and  Best, 
P.J.  Ethanol  alters  spatial  processing  of 
hippocampal  place  cells:  A  mechanism  for 
impaired  navigation  when  intoxicated. 
Alcohol  Clin  Exp  ite20(2):404-407, 1996. 

Mereu,  G.,  and  Gessa,  G.L.  Low  doses  of 
ethanol  inhibit  the  firing  of  neurons  in  the 
substantia  nigra,  pars  reticulata:  A  GABAergic 
effect?  5ramlto360(l-2):325-330, 1985. 

Mihic,  S.J.;  Ye,  Q.;  Wick,  M.J.;  Koltchine, 
V.V.;  Krasowski,  M.D.;  Finn,  S.E.; 
Mascia,  M.P.;  Valenzuela,  C.F.;  Hanson, 
K.K.;  Greenblatt,  E.P.;  Harris,  R.A.;  and 
Harrison,  N.L.  Sites  of  alcohol  and 
volatile  anaesthetic  action  on  GABAA  and 
glycine  receptors.  Nature  389:385-389, 
1997. 

Millar,  D.P.  Time-resolved  fluorescence 
spectroscopy.  Curr  Opin  Struct  Biol 
6(5):637-642,  1996. 

Mirshahi,  T.,  and  Woodward,  J.J.  C- 
terminus  truncation  reduces  the  ethanol 
sensitivity  of  NMDA  receptors  under 
elevated  calcium  conditions.  Alcohol  Clin 
Exp  Res  2l(3):72A,  1997. 

Miyakawa,  T.;  Yagi,  T.;  Kitazawa,  H.; 
Yasuda,  M.;  Kawai,  N.;  Tsuboi,  K.;  and 
Niki,  H.  Fyn  kinase  as  a  determinant  of 
ethanol  sensitivity:  Relation  to  NMDA- 
receptor  function.  Science  278:698-701, 
1997. 

Mochly-Rosen,  D.  Compartmentalization 
of  protein  kinases  by  intracellular  receptor 
proteins:  A  theme  in  signal  transduction. 
Science  268:247-251,  1995. 

Moore,  M.S.;  DeZazzo,  J.;  Luk,  AY.; 
Tully,  T.;  Singh,  C.M.;  and  Heberlein,  U. 


Ethanol  intoxication  in  Drosophila: 
Genetic  and  pharmacological  evidence  for 
regulation  by  the  cAMP  signaling  path- 
way. O//93(6):997-1007, 1998. 

Moore,  S.D.;  Madamba,  S.G.;  and 
Siggins,  G.R.  Ethanol  diminishes  a  volt- 
age-dependent K+  current,  the  M-cur- 
rent,  in  CA1  hippocampal  pyramidal 
neurons  in  vitro.  Brain  Res 
516(2):222-228,  1990. 

Morrisett,  R.A.,  and  Swartzwelder,  H.S. 
Attenuation  of  hippocampal  long-term 
potentiation  by  ethanol:  A  patch-clamp 
analysis  of  glutamatergic  and  GABAergic 
mechanisms.  J Neurosci  13(5):2264- 
2272, 1993. 

Mullikin-Kilpatrick,  D.,  and  Treistman, 
S.N.  Inhibition  of  dihydropyridine-sensi- 
tive  Ca++  channels  by  ethanol  in  undiffer- 
entiated and  nerve  growth  factor-treated 
PC  12  cells:  Interaction  with  the  inacti- 
vated state.  J  Pharmacol  Exp  Ther 
272(2):489^97,  1995. 

Mullikin-Kilpatrick,  D.;  Mehta,  N.D.; 
Hildebrandt,  J.D.;  and  Treistman,  S.N.  G; 
is  involved  in  ethanol  inhibition  of  L- type 
calcium  channels  in  undifferentiated  but 
not  differentiated  PC  12  cells.  Mol 
Pharmacol  47  (5):997-l005,  1995. 

Nagata,  K.;  Aistrup,  G.L.;  Huang,  C.S.; 
Marszalec,  W.;  Song,  J.H.;  Yeh,  J.Z.;  and 
Narahashi,  T.  Potent  modulation  of  neu- 
ronal nicotinic  acetylcholine  receptor- 
channel  by  ethanol.  Neurosci  Lett 
217(2-3):189-193,  1996. 

Nicoll,  R.A.,  and  Malenka,  RC. 
Contrasting  properties  of  two  forms  of 
long-term  potentiation  in  the  hippocam- 
pus, mzwr  377(6545):  11 5-1 18,  1995. 

Oliet,  S.H.;  Malenka,  RC;  and  Nicoll, 
RA.  Bidirectional  control  of  quantal  size 


39 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


by  synaptic  activity  in  the  hippocampus. 
Science  271(5253):1294-1297,  1996. 

Opella,  S.J.  Nuclear  magnetic  resonance 
approaches  to  membrane  protein  struc- 
ture. In:  White,  S.H.,  ed.  Membrane 
Protein  Structure:  Experimental 
Approaches.  Oxford,  England:  Oxford 
University  Press,  1994.  pp.  249-267. 

Opella,  S.J.  NMR  and  membrane  proteins. 
Nat  Struct  Biol  4(suppl):84S-858,  1997. 

Parent,  L.,  and  Wright,  E.M.  Electrophy- 
siology  of  the  Na+/glucose  cotransporter. 
Soc  Gen  Physiol  Ser  48:263-281,  1993. 

Pettit,  D.L.;  Wang,  S.S.;  Gee,  K.R.;  and 
Augustine,  G.J.  Chemical  two-photon 
uncaging:  A  novel  approach  to  mapping 
glutamate  receptors.  Neuron  19(3): 
465-471, 1997. 

Plenz,  D.,  and  Kitai,  S.T.  Up  and  down 
states  in  striatal  medium  spiny  neurons 
simultaneously  recorded  with  spontaneous 
activity  in  fast-spiking  interneurons  studied 
in  cortex-striatum-substantia  nigra  organ- 
otypic cultures.  J  Neurosci  18(1):266- 
283,  1998. 

Raming,  K.;  Krieger,  J.;  Strotmann,  J.; 
Boekhoff,  I.;  Kubick,  S.;  Baumstark,  C;  and 
Breer,  J.  Cloning  and  expression  of  odorant 
receptors.  Nature  361:353-356,  1993. 

Ramirez-Solis,  R;  Zheng,  H.;  Whiting, 
J.;  Krumlauf,  R;  and  Bradley,  A.  Hoxb-4 
(Hox-2.6)  mutant  mice  show  homeotic 
transformation  of  a  cervical  vertebra  and 
defects  in  the  closure  of  the  sternal  rudi- 
ments. Cell  73:279-294,  1993. 

Ranganathan,  R.,  and  Ross,  E.M.  PDZ 
domain  proteins:  Scaffolds  for  signaling  com- 
plexes. Curr  Biol7(l2):K770-K773, 1997. 

Redman,  S.  Quantal  analysis  of  synaptic 
potentials  in  neurons  of  the  central  nervous 
system.  Physiol  Rev  70(1):  165-198,  1990. 


Rosenmund,  C;  Carr,  D.W.;  Bergeson, 
S.E.;  Nilaver,  G.;  Scott,  J.D.;  and 
Westbrook,  G.L.  Anchoring  of  protein 
kinase  A  is  required  for  modulation  of 
AMPA/kainate  receptors  on  hippocampal 
neurons.  Nature  368(6474):853-856, 
1994. 

Ruano,  D.;  Araujo,  F.;  Machado,  A.;  de 
Bias,  A.L.;  and  Vitorica,  J.  Molecular 
characterization  of  type  I  GABAA  recep- 
tor complex  from  rat  cerebral  cortex  and 
hippocampus.  Brain  Res  Mol  Brain  Res 
25(3-4):225-233,  1994. 

Rubinstein,  M.;  Phillips,  T.J.;  Bunzow, 
J.R.;  Falzone,  T.L.;  Dziewczapolski,  G.; 
Zhang,  G.;  Fang,  Y.;  Larson,  J.L.; 
McDougall,  J.A.;  Chester,  J.A.;  Saez,  C; 
Pugsley,  T.A.;  Gershanik,  O.;  Low,  M.J.; 
and  Grandy,  D.K.  Mice  lacking  dopamine 
D4  receptors  are  supersensitive  to 
ethanol,  cocaine,  and  methamphetamine. 
O//90(6):991-1001,  1997. 

Ryabinin,  A.E.;  Criado,  J.R;  Henriksen, 
S.J.;  Bloom,  F.E.;  and  Wilson,  M.C. 
Differential  sensitivity  of  c-Fos  expression 
in  hippocampus  and  other  brain  regions 
to  moderate  and  low  doses  of  alcohol. 
Mol  Psychiatry  2(  1  ):32-43,  1997. 

Ryan,  T.A.;  Reuter,  PL;  Wendland,  B.; 
Schweizer,  F.E.;  Tsien,  R.W.;  and  Smith, 
S.J.  The  kinetics  of  synaptic  vesicle  recy- 
cling measured  at  single  presynaptic  bou- 
tons.  Neuron  ll(4):713-724,  1993. 

Ryan,  T.A.;  Reuter,  H.;  and  Smith,  S.J. 
Optical  detection  of  a  quantal  presynaptic 
membrane  turnover.  Nature 
388(6641  ):478-482,  1997. 

Sahoo,  D.;  Narayanaswami,  V.;  Kay,  CM.; 
and  Ryan,  RO.  Fluorescence  studies  of 
exchangeable  apolipoprotein-lipid  interac- 
tions. Superficial  association  of  apoli- 
pophorin  III  with  lipoprotein  surfaces.  / 
Biol  Chem  27  3(3):U03-U08,  1998. 


40 


Neural  Proteins 


Salin,  P. A.;  Malenka,  R.C.;  and  Nicoll, 
R.A.  Cyclic  AMP  mediates  a  presynaptic 
form  of  LTP  at  cerebellar  parallel  fiber. 
Neuron  16(4):797-803,  1996. 

Samson,  H.H.,  and  Hodge,  C.W.  The 
role  of  the  mesoaccumbens  dopamine 
system  in  ethanol  reinforcement:  Studies 
using  the  techniques  of  microinjection 
and  voltammetry.  Alcohol  Alcohol  Suppl 
2:469^74,  1993. 

Samson,  H.H.;  Hodge,  C.W.;  Erickson, 
H.L.;  Niehus,  J.S.;  Gerhardt,  G.A.;  Kalivas, 
P.W.;  and  Floyd,  EA.  The  effects  of  local 
application  of  ethanol  in  the  nucleus  ac- 
cumbens  on  dopamine  overflow  and  clear- 
ance. ^/co^/14(5):485-492,  1997. 

Sanna,  E.,  and  Harris,  KA.  Recent  develop- 
ments in  alcoholism:  Neuronal  ion  channels. 
Recent  Dev  Alcohol  11:169-186,  1993. 

Sapp,  D.W.,  and  Yeh,  H.H.  Effect  of 
chronic  ethanol  on  GABA  response  pro- 
files and  GABAA  receptor  subunit  expres- 
sion in  cultured  cerebellar  neurons.  Alcohol 
Clin  Exp  Res 20(2):74A,  1997. 

Sato,  T.;  Hirono,  J.;  Tonoike,  M.;  and 
Takebayashi,  M.  Tuning  specificities  to 
aliphatic  odorants  in  mouse  olfactory 
receptor  neurons  and  their  local  distribution. 
J  Neurophysiol  72(6):2980-2989,  1994. 

Scholz,  J.;  Singh,  C.;  Luk,  A.;  and  Heberlein, 
U.  The  development  of  ethanol  tolerance 
in  Drosophila.  Alcohol  Clin  Exp  Res  21(3): 
16A,  1997. 

Schummers,  J.;  Bentz,  S.;  and  Browning, 
M.D.  EthanoPs  inhibition  of  LTP  may 
not  be  mediated  solely  via  direct  effects 
on  the  NMD  A  receptor.  Alcohol  Clin  Exp 
ito21(3):404-408,  1997. 

Seeburg,  P.H.;  Burnashev,  N.;  Kohr,  G.; 
Kuner,  T.;  Sprengel,  R.;  and  Monyer,  H. 
The  NMDA  receptor  channel:  Molecular 


design  of  a  coincidence  detector.  Recent 
Prog  Horm  Res  50:19-34,  1995. 

Sheng,  M.;  Cummings,  J.;  Roldan,  L.A.; 
Jan,  Y.N.;  and  Jan,  L.Y.  Changing  sub- 
unit  composition  of  heteromeric  NMDA 
receptors  during  development  of  rat  cor- 
tex. Nature  368(6467 ):144-147,  1994. 

Sigel,  E.;  Baur,  R;  and  Malherbe,  P. 
Recombinant  GABAA  receptor  function 
and  ethanol.  FEBSLett  324:140-142, 1993. 

Slater,  S.J.;  Cox,  K.J.;  Lombardi,  J.V.; 
Ho,  C;  Kelly,  M.B.;  Rubin,  E.;  and 
Stubbs,  CD.  Inhibition  of  protein  kinase 
C  by  alcohols  and  anaesthetics.  Nature 
364(6432):82-84,  1993. 

Slater,  S.J.;  Kelly,  M.B.;  Larkin,  J.D.;  Ho, 
C;  Mazurek,  A.;  Taddeo,  F.J.;  Yeager, 
M.D.;  and  Stubbs,  CD.  Interaction  of 
alcohols  and  anesthetics  with  protein 
kinase  Calpha.  J  Biol  Chem  272(10): 
6167-6173,  1997. 

Sobol,  A.;  Arseniau,  A.;  Abdulaeva,  G.; 
Musina,  L.;  and  Bystrov,  V.  Sequence- 
specific  resonance  assignment  and  sec- 
ondary structure  of  (1-71)  bacterioopsin. 
JBiomolNMR  2:161-171,  1992. 

Solem,  M.;  McMahon,  T.;  and  Messing, 
RO.  Protein  kinase  A  regulates  inhibition 
of  N-  and  P/Q-type  calcium  channels  by 
ethanol  in  PC12  cells.  / Pharmacol  Exp 
7^r282(3):1487-1495,  1997. 

Stratikos,  E.,  and  Gettins,  P.G.  Major 
proteinase  movement  upon  stable  serpin- 
proteinase  complex  formation.  Proc  Natl 
AcadSci  USA  94(2):453-458,  1997. 

Sudhof,  T.  The  synaptic  vesicle  cycle:  A 
cascade  of  protein-protein  interactions. 
Nature  37 5(6533):64S-653,  1995. 

Thomas,  M.P.,  and  Morrisett,  RA. 
Modulation  of  synaptic  transmission  by 


41 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol:  Role  of  L-type  calcium  channels. 
Soc  Neurosci  Abstr23(2):17 56,  1997a. 

Thomas,  M.P.,  and  Morrisett,  RA. 
Neuronal  viability  and  NMDA-induced 
neurotoxicity  following  ethanol  exposure 
and  withdrawal  in  hippocampal  explants. 
Alcohol  Clin  Exp  Res  21(3):  71A,  19976. 

Tonegawa,  S.;  Li,  Y.;  Erzurumlu,  R.S.; 
Jhaveri,  S.;  Chen,  C;  Goda,  Y.;  Paylor, 
R;  Silva,  A.J.;  Kim,  J. J.;  and  Wehner, 
J.M.  The  gene  knockout  technology  for 
the  analysis  of  learning  and  memory,  and 
neural  development.  Prog  Brain  Res 
105:3-14,  1995. 

Tsien,  J.Z.;  Chen,  D.F.;  Gerber,  D.;  Tom, 
C;  Mercer,  E.H.;  Anderson,  D.J.;  Mayford, 
M.;  Kandel,  E.R.;  and  Tonegawa,  S. 
Subregion-  and  cell  type-restricted  gene 
knockout  in  mouse  brain.  Cell  87(7): 
1317-1326,  1996. 

von  Kitzing,  E.;  Jonas,  P.;  and  Sakmann, 
B.  Quantal  analysis  of  excitatory  postsy- 
naptic currents  at  the  hippocampal  mossy 
fiber- CA3  pyramidal  cell  synapse.  Adv 
Second  Messenger  Phosphoprotein  Res 
29:235-260,  1994. 

Wafford,  K.A.,  and  Whiting,  P.J.  Ethanol 
potentiation  of  GABAA  receptors  requires 
phosphorylation  of  the  alternatively 
spliced  variant  of  the  gamma  2  subunit. 
i«EBSLrt*313(2):113-117,  1992. 

Wang,  X.M.;  Dayanithi,  G.;  Lemos,  J.R.; 
Nordmann,  J.J.;  and  Treistman,  S.N. 
Calcium  currents  and  peptide  release  from 
neurohypophysial  terminals  are  inhibited 
by  ethanol.  J  Pharmacol  Exp  Ther 
259(2):705-711, 1991. 

Wang,  Y.;  Jeng,  C.H.;  Lin,  J.C.;  and 
Wang,  J.Y.  Serotonin  modulates  ethanol- 
induced  depression  in  cerebellar  Purkinje 
neurons.  Alcohol  Clin  Exp  Res  20(7 ):1229- 
1236,  1996. 


Weiner,  J.L.;  Svoboda,  K.R.;  Lupica, 
C.R;  and  Dunwiddie,  T.V.  Pre-  and  post- 
synaptic actions  of  ethanol  on  GABAA 
receptor-mediated  synaptic  transmission 
in  rat  hippocampal  CA1  pyramidal  neu- 
rons. Soc  NeurosciAbstr  23(l):959,  1997a. 

Weiner,  J.L.;  Valenzuela,  C.F.;  Watson, 
P.L.;  Frazier,  C.J.;  and  Dunwiddie,  T.V. 
Elevation  of  basal  protein  kinase  C  activity 
increases  ethanol  sensitivity  of  GABA( A) 
receptors  in  rat  hippocampal  CA1  pyrami- 
dal neurons.  J  Neurochem  68(5):1949- 
1959,  1997b. 

Weiss,  F.;  Lorang,  M.T.;  Bloom,  F.E.; 
and  Koob,  G.F.  Oral  alcohol  self- adminis- 
tration stimulates  dopamine  release  in  the 
rat  nucleus  accumbens:  Genetic  and  moti- 
vational determinants.  J  Pharmacol  Exp 
77kt267(1):250-258,  1993. 

Williams-Hemby,  L.,  and  Porrino,  L.J. 
Functional  consequences  of  intragastri- 
cally  administered  ethanol  in  rats  as  mea- 
sured by  the  2-[14C]deoxyglucose  method: 
The  contribution  of  dopamine.  Alcohol 
Clin  Exp  Res  21(9):  158 1-1 591,  1997. 

Wilson,  M.A.,  and  McNaughton,  B.L. 
Reactivation  of  hippocampal  ensemble 
memories  during  sleep.  Science  265(5172): 
676-679,  1994. 

Woodward,  D.J.  Behavioral  neurophysiol- 
ogy: Neuronal  spike  train  activity  in  alco- 
hol research.  Alcohol  Clin  Exp  Res2§(% 
suppl):101A-105A,  1996. 

Wozniak,  K.M.;  Pert,  A.;  Mele,  A.;  and 
Linnoila,  M.  Focal  application  of  alcohols 
elevates  extracellular  dopamine  in  rat 
brain:  A  microdialysis  study.  Brain  Res 
540(l-2):31-40,  1991. 

Wright,  J.M.;  Peoples,  R.W.;  and  Weight, 
F.F.  Single-channel  and  whole-cell  analy- 
sis of  ethanol  inhibition  of  NMD  A- acti- 
vated currents  in  cultured  mouse  cortical 


42 


Neural  Proteins 


and  hippocampal  neurons.  Brain  Res 
738(2):249-256,  1996. 

Wu,  J.,  and  Kaback,  H.R.  Cysteine  148  in 
the  lactose  permease  of  Escherichia  coli  is 
a  component  of  a  substrate  binding  site. 
2.  Site-directed  fluorescence  studies. 
Biochemistry  33(40):12166-12171,  1994. 

Wu,  L.G.,  and  Saggau,  P.  Presynaptic  inhi- 
bition of  elicited  neurotransmitter  release. 
Trends  Neurosci  20(5):204-212,  1997. 

Xie,  P.;  Parsons,  S.H.;  Speckhard,  D.C.; 
Bosron,  W.F.;  and  Hurley,  T.D.  X-ray 
structure  of  human  class  IV  sigmasigma 
alcohol  dehydrogenase.  J  Biol  Chem 
272(30):18558-18563,  1997. 

Yang,  X.;  Criswell,  H.E.;  Simson,  P.; 
Moy,  S.;  and  Breese,  G.R.  Evidence  for  a 
selective  effect  of  ethanol  on  N-methyl-D- 
aspartate  responses:  Ethanol  affects  a 
subtype  of  the  ifenprodil-sensitive  N- 


methyl-D-aspartate  receptors.  J  Pharmacol 
Exp  Ther  278(1):  114-124,  1996. 

Yim,  HJ.;  Schallert,  T.;  Randall,  P.K.; 
Bungay,  P.M.;  and  Gonzales,  RA.  Effect 
of  ethanol  on  extracellular  dopamine  in 
rat  striatum  by  direct  perfusion  with  mi- 
crodialysis.  J  Neurochem  68(4):1527- 
1533, 1997. 

Yu,  D.;  Zhang,  L.;  Eisele,  J.L.;  Bertrand, 
D.;  Changeux,  J. P.;  and  Weight,  F.F. 
Ethanol  inhibition  of  nicotinic  acetyl- 
choline type  alpha  7  receptors  involves  the 
amino -terminal  domain  of  the  receptor. 
Mol  Pharmacol  50(4):1010-1016,  1996. 

Zhou,  Q.;  Verdoorn,  T.A.;  Lovinger, 
D.M.  Alcohols  potentiate  the  function  of 
5-HT3  receptor-channels  on  NCB-20 
neuroblastoma  cells  by  favouring  and 
stabilizing  the  open  channel  state.  / 
Physiol  (Lond)  507(Part  2):335-352,  1998. 


43 


Chapter  2 

Lipid  Involvement  in  the  Acute  Actions 
of  Alcohol  in  the  Nervous  System 

Steven  N.  Treistman,  Ph.D. 


KEY  WORDS:  acute  AODE  (effects  ofAOD  [alcohol  or  other  drug]  use,  abuse, 
and  dependence);  lipid  metabolism;  nervous  system;  lipids;  membrane  proteins; 
calcium;  potassium;  voltage  gated  channel;  biochemical  mechanism;  cholesterol; 
literature  review 


In  this  chapter  I  address  the  effects  of 
alcohol  (ethanol)  on  membrane  lipids 
(in  particular  in  the  nervous  system) 
and  the  influence  of  the  lipid  environ- 
ment on  the  functioning  of  membrane 
proteins,  and  I  propose  some  recom- 
mendations for  the  conduct  of  research 
in  this  area.  Although  many  of  the 
studies  of  lipids  and  alcohol  have  been 
framed  and  interpreted  to  determine 
whether  the  primary  target  of  alcohol 
action  is  the  lipid  or  the  protein,  I 
would  argue  that  this  is  not  the  most 
productive  approach.  Rather,  it  is  nec- 
essary to  consider  functioning  mem- 
brane proteins  and  their  lipid 
environment  (as  well  as  the  various 
interfaces,  such  as  lipid-protein,  lipid- 


protein-water,  protein-lipid-protein, 
etc.)  as  a  dynamic  system,  in  which  the 
small  amphiphilic  alcohol  molecule  will 
interact  with  a  number  of  targets. 
Although  alcohol  is  sometimes  referred 
to  as  a  lipophilic  molecule,  it  is  impor- 
tant to  keep  in  mind  that  it  is  about  10 
times  more  "comfortable"  in  the  aque- 
ous compartment  (Goldstein  1983). 

The  volume  of  literature  on  lipids 
and  alcohol  is  so  great  that  an  exhaus- 
tive summary  is  impossible  within  the 
space  limitations  of  this  chapter. 
Moreover,  a  summary  of  this  research 
would  produce  a  bewildering  mass  of 
data,  with  little  indication  of  the 
import  of  each  of  the  pieces  on  our 
ultimate  understanding  of  alcohol 


S.N.  Treistman,  Ph.D.,  is  professor  of  pharmacology  and  director  of  the  Neuroscience  Program  at  the 
Department  of  Pharmacology,  University  of  Massachusetts  Medical  Center,  55  Lake  Ave.  North, 
Worcester,  MA  01655. 


45 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


action.  In  fact,  this  observation  sug- 
gests one  of  the  problems  that  have 
accompanied  much  of  the  work  on 
lipids  and  alcohol.  It  is  certainly  fair 
to  say  that,  driven  by  the  results  of 
classic  studies  dating  from  the  turn  of 
the  century,  membrane  lipids  were 
the  primary  focus  of  biochemical  and 
biophysical  research  on  alcohol's 
actions  until  recently.  However,  the 
need  to  use  very  high  alcohol  concen- 
trations to  get  reliably  measurable 
perturbations,  frequently  coupled 
with  an  absence  of  the  physiological 
consequence  (e.g.,  the  functioning  of 
membrane  channels  and  receptors)  of 
the  reported  perturbations,  makes  it 
extremely  difficult  to  assess  the 
importance  of  each  of  the  reports.  In 
this  chapter,  therefore,  I  have  chosen 
only  a  small  subset  of  the  experiments 
reported  in  the  literature,  focusing  on 
some  of  those  that  offer  the  greatest 
promise  for  pinpointing  those  aspects 
of  lipids  and  alcohol  action  that  are 
physiologically  relevant.  The  data  dis- 
cussed are  chosen  to  provide  some 
background  for  the  approaches  that 
will  be  advocated  at  the  conclusion  of 
this  chapter.  Unavoidably,  many 
exciting  results  from  the  literature 
have  been  ignored,  because  they  did 
not  fit  this  criterion. 

I  will  explore  the  role  of  lipids  in 
alcohol  action  from  three  primary 
perspectives.  First,  I  will  look  at  some 
of  the  lipid  perturbations  produced 
by  acute  alcohol,  highlighting  a  shift 
in  emphasis  from  bulk  lipid  effects  to 
more  subtle  effects  on  different  lipid 
compartments.  Second,  I  will  exam- 
ine the  manner  in  which  lipid  envi- 
ronment affects  the  functioning  of 


ligand-  and  voltage-gated  channels 
and  might  affect  the  manner  in  which 
the  channels  respond  to  alcohol. 
Finally,  I  will  discuss  some  approaches 
that  combine  our  knowledge  of  lipid 
effects  on  channel  function  with  our 
knowledge  of  alcohol's  actions  on 
membrane  lipids.  Because  so  much  of 
the  impetus  for  questioning  the  role 
of  lipids  in  alcohol  action  derives 
from  a  large  body  of  data  document- 
ing changes  in  membrane  lipid  com- 
position as  a  function  of  chronic 
exposure  of  the  cells  or  the  animal  to 
the  drug,  some  discussion  will  be 
devoted  to  these  effects,  even  though 
the  primary  focus  of  this  paper  is  on 
the  acute  effects  of  alcohol. 

The  remarkable  shift  in  emphasis 
from  lipids  to  proteins  as  the  targets 
of  acute  alcohol  action  is  well 
founded.  A  significant  contributor  to 
this  trend  has  been  the  development 
of  cloned  channels  and  receptors,  as 
well  as  the  use  of  mutagenesis  to 
relate  protein  sequence  to  physiology 
and  pharmacology.  It  is  arguable, 
based  upon  data  already  collected, 
that  alcohol  can  interact  directly  with 
membrane  proteins  to  produce  alter- 
ations in  function.  However,  this 
should  not  distract  us  from  the 
important  role  that  lipid  environment 
may  play  in  the  interaction  between 
the  drug  and  the  protein.  That  lipids 
may  play  an  important  role  is  sug- 
gested by  the  great  diversity  of  lipids 
in  the  membrane,  by  the  strong  influ- 
ence of  lipid  composition  on  channel 
protein  function,  and  by  the  fact  that 
apparent  compensatory  changes  in 
lipids  occur  as  a  function  of  chronic 
drug  exposure. 


46 


Lipid  Involvement  in  Acute  Alcohol  Actions 


EVIDENCE  THAT 
ALCOHOL  AFFECTS  THE 
LIPID  ENVIRONMENT 

Much  of  the  extensive  literature  on 
alcohol  and  lipids  represents  a  body 
of  sophisticated  biophysical  measure- 
ments documenting  the  alteration  of 
various  parameters  of  the  lipid  phase 
of  the  membrane.  Although  many  of 
these  studies  provide  potential  mech- 
anisms for  the  actions  of  alcohol  in 
the  nervous  system,  it  is  important  to 
point  out  that  many  of  these  studies 
use  exceedingly  high  concentrations 
of  alcohol  and  that  there  remains  a 
question  of  whether  the  perturbations 
seen  translate  into  altered  protein 
function.  In  recent  years,  it  has 
become  apparent  that  it  is  necessary 
to  think  of  the  membrane  as  com- 
posed of  various  compartments,  such 
as  the  separate  leaflets,  the  annular 
versus  the  bulk  lipid,  the  acyl  chain 
region  versus  the  headgroup  region, 
interfacial  regions,  and  a  number  of 
others.  The  effects  of  alcohol  may  be 
significantly  more  potent  on  individual 
components  than  on  the  bulk  proper- 
ties of  the  membrane  taken  as  a  whole. 
Various  functions  of  proteins  such  as 
membrane  channels  are  far  more  sen- 
sitive to  influences  from  some  of  these 
compartments  than  others. 

Biological  membranes  are  highly 
organized  structures  with  nonrandom 
distribution  of  lipids  (Gennis  1989). 
For  example,  some  lipid  species  pref- 
erentially distribute  into  the  extracel- 
lular leaflet  of  the  membrane,  while 
others  are  found  predominantly  in 
the  intracellular  leaflet,  forming  verti- 
cal "transbilayer"  domains  (Gennis 


1989;  Devaux  and  Zachowski  1994). 
Lipids  also  preferentially  cluster 
within  a  bilayer  leaflet  to  form  lateral 
domains  (Welti  and  Glaser  1994). 
Formation  of  lateral  domains  can 
result  from  the  juxtaposition  of  coex- 
isting areas  of  gel  and  fluid  phase 
lipids,  the  nonrandom  mixing 
between  different  lipid  species,  or  the 
presence  of  cholesterol,  Ca2+,  or  pro- 
teins. A  number  of  studies  have 
demonstrated  that  alcohols  have 
selective  actions  on  vertical  and  lateral 
domains.  For  example,  ethanol  selec- 
tively increases  the  fluidity  of  the 
extracellular  leaflet  in  synaptic  plasma 
membranes,  an  effect  attributable  to 
differences  in  transbilayer  cholesterol 
distribution.  Using  fluorescence  pho- 
tobleaching  recovery  techniques  in 
Aplysia  neurons,  ethanol  was  shown 
to  increase  the  diffusion  of  the  probe 
rhodamine-phosphatidyl-ethanolamine 
more  than  the  probe  l-acyl-2-(6-[?vr- 
(7-nitrobenz-2-oxa-l,3-diazol-4-yl)] 
aminohexanoyl )  phosphatidylcholine , 
suggesting  that  ethanol's  actions  on 
membrane  proteins,  such  as  gated  ion 
channels,  might  be  dependent  upon  the 
existence  of  dissimilar  lateral  domains 
(Treistman  et  al.  1987). 

More  detailed  studies  with  respect 
to  alcohol  action  on  lateral  domains 
have  been  conducted  in  model  mem- 
branes. For  example,  alcohol's  ability 
to  disorder  model  membranes  is 
enhanced  by  gangliosides  (Harris  et  al. 
1984)  and  phospholipid  polyunsatura- 
tion  (Ho  et  al.  1994)  but  is  antagonized 
by  cholesterol  (Chin  and  Goldstein 
1981).  Consequently,  native  mem- 
brane domains  rich  in  gangliosides 
and  polyunsaturated  phospholipids,  but 


47 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


low  in  cholesterol,  would  presumably 
be  particularly  sensitive  to  perturbation 
by  alcohol  (Deitrich  et  al.  1989). 
Computer  modeling  studies  also  sug- 
gest that  ethanol  preferentially  accu- 
mulates in  domains  with  special 
packing  properties  favoring  the  inter- 
calation of  alcohols  (Jorgensen  et  al. 
1993).  Membrane  proteins  in  these 
domains  would  be  exposed  to  concen- 
trations of  ethanol  higher  than  that  in 
the  bulk  membrane  (Goldstein  1984). 

One  argument  that  has  received 
significant  attention  is  that  lipids  are 
unlikely  to  be  a  primary  player  in  the 
actions  of  alcohol,  since  a  small  tem- 
perature change  will  produce  changes 
in,  for  example,  probe  mobility, 
greater  than  the  changes  measured  in 
the  presence  of  alcohol.  However, 
this  argument  must  be  taken  with 
caution,  given  the  existence  of  lipid 
compartments  within  the  membrane. 
For  example,  in  our  Aplysia  studies, 
temperature  and  alcohols  had  decid- 
edly different  and  selective  effects  on 
each  of  the  probes  used,  as  well  as  on 
the  kinetics  of  channel  function 
(Treistman  and  Wilson  1987&, 
1987b;  Treistman  et  al.  1987),  and 
similar  discrepancies  between  temper- 
ature and  alcohol  effects  have  been 
reported  for  a  number  of  other  pro- 
teins (Wood  et  al.  1996). 

One  of  the  earlier  attempts  to 
assign  alcohol's  effects  on  lipids  to 
membrane  compartments  examined 
the  role  of  gangliosides  in  alcohol's 
actions.  Harris,  Hitzemann,  and  col- 
leagues (Harris  et  al.  1984)  noted  that 
the  signal  obtained  from  a  fluorescent 
probe  intercalated  into  most  artificial 
vesicles  was  not  affected  by  alcohol, 


whereas  the  fluorescence  signal  from 
natural  membranes  was.  They  examined 
whether  the  presence  of  gangliosides 
might  be  contributing  to  this  differ- 
ence, and  they  examined  three  probes, 
thought  to  selectively  sample  different 
depths  and  environments  of  the 
bilayer.  They  concluded  that  the  outer 
leaflet  was  most  sensitive  to  alcohol, 
in  the  presence  of  the  gangliosides, 
and  that  phosphatidylcholine  was  par- 
ticularly important  for  the  effect. 

Interfacial  surfaces  are  proving  par- 
ticularly attractive  as  sites  of  alcohol 
action.  Gawrisch  and  Barry  have  ques- 
tioned the  prevailing  belief  that  protein 
hydrophobic  pockets  are  the  site  of 
alcohol  action,  noting  that  the  ampho- 
philic nature  of  alcohols  favors  an  inter- 
facial location,  and  interactions  are 
driven  by  both  the  opportunity  for 
hydrogen  bonding  and  hydrophobic 
interactions  (Holte  and  Gawrisch 
1997).  Using  nuclear  magnetic  reso- 
nance (NMR)  spectroscopy  of  artifi- 
cial phosphatidylcholine  membranes, 
they  probed  the  lipid-water  interface 
and  found  evidence  for  alcohol  acting 
in  this  region,  noting  that  the  disor- 
dering influence  of  the  drug  was 
enhanced  by  gangliosides  and  inhib- 
ited by  cholesterol. 

Wood  and  colleagues  (1996)  made 
the  argument  that  the  membrane 
must  be  treated  as  a  complex  system, 
in  which  properties  such  as  dielectric 
constant,  interdigitation,  lipid  domains, 
and  lipid-protein  interactions  are  con- 
sidered in  discussions  of  alcohol  action. 
The  dielectric  constant  can  be  viewed 
as  a  measure  of  the  access  of  water  to 
the  membrane  interior.  At  20  °C  the 
dielectric  constant  of  water  is  80, 


48 


Lipid  Involvement  in  Acute  Alcohol  Actions 


while  that  of  oleic  acid  is  2.5.  Alcohol 
appears  to  increase  the  dielectric  con- 
stant of  membranes,  weakening  hydro- 
gen bonding  and  allowing  water  to 
infiltrate  the  hydrocarbon  core  (Orme 
et  al.  1988;  Rottenberg  1992;  Wood 
et  al.  1996).  Increased  interdigitation 
of  the  leaflets  induced  by  alcohol  may 
have  consequences  for  protein  function 
by,  for  example,  changing  the  protein 
conformation,  allowing  more  hydro- 
phobic portions  to  become  available 
to  the  aqueous  medium  (Wood  et  al. 
1996).  Wood  and  colleagues  stressed 
the  fact  that  measurements  of  bulk 
lipid  properties  may  be  misleading  in 
the  determination  of  lipid  involve- 
ment in  alcohol  actions.  For  example, 
cholesterol  is  not  distributed  evenly 
between  the  two  membrane  leaflets, 
and  a  change  in  the  concentration 
ratio  between  the  exo-  and  cytofacial 
leaflets  could  have  significant  conse- 
quences for  protein  function,  even  if 
the  change  in  overall  cholesterol  con- 
tent is  minimal. 

Rubin,  Janes,  Taraschi,  and  collea- 
gues (Janes  et  al.  1992;  Channareddy 
et  al.  1996)  have  put  forth  the  idea 
that  configurational  entropy  is  the 
driving  force  for  alcohol  action  on 
membrane  architecture.  Because  the 
membrane  is  quite  different  at  different 
levels,  particular  characteristics  of  indi- 
vidual drugs,  such  as  charge,  result  in 
actions  specific  to  particular  regions  of 
the  membrane.  These  authors  used 
data  obtained  with  NMR  techniques 
in  artificial  membranes  and  thermody- 
namic analysis  to  demonstrate  that 
partitioned  alcohols  perturb  the  proper- 
ties of  the  entire  membrane,  both  sur- 
face and  core,  by  altering  the  energetic 


balance  among  membrane  structures. 
Their  analysis  precludes  the  need  to 
provide  a  specific  mechanism  of  surface 
adsorption  to  account  for  alcohol- 
induced  alterations  at  the  membrane 
surface.  Partitioning  differences 
between  membrane  structures  are  a 
prerequisite  for  alcohol  action  via  con- 
figurational entropy.  Barry  and  Gawrisch 
(1994)  presented  data  using  NMR 
spectroscopy  that  suggest  that  alcohol 
binds  at  the  lipid-water  interface  of 
phospholipid  bilayers,  disorienting  the 
headgroups  and,  through  interfacial 
interaction,  causing  significant  disor- 
dering along  the  entire  hydrocarbon 
acyl  chain. 

Although  the  focus  of  this  chapter  is 
to  assess  lipid  involvement  in  the  acute 
actions  of  alcohol,  it  is  difficult  to 
completely  ignore  the  vast  body  of 
work  addressing  the  influence  of  chronic 
alcohol  exposure  on  membrane  lipid 
composition  and  function.  As  pointed 
out  by  Salem  (1989),  the  number  of 
variables  that  enter  into  these  studies 
makes  it  unsurprising  that  results  from 
different  laboratories,  such  as  reports 
of  alterations  in  membrane  cholesterol 
levels,  are  often  at  odds.  However,  as 
indicated  later  in  this  chapter,  the 
influence  of  cholesterol  on  the  function- 
ing of  some  membrane  receptors  and 
channels  is  well  established,  making 
such  alterations  particularly  important. 
Some  reported  changes,  such  as  a 
decrease  in  22:6u)3  fatty  acids  in 
retina  (Pawlosky  and  Salem  1995), 
may  be  associated  with  functional  con- 
sequences, such  as  visual  pathologies. 
Some  of  the  chronic  studies  suggest 
that  bulk  lipid  properties,  such  as 
membrane  disordering  induced  by 


49 


NIAAA's  Ncurosciencc  and  Behavioral  Research  Portfolio 


alcohol,  are  significantly  reduced  in 
animals  that  have  been  chronically 
exposed  to  the  drug  (Ellingson  et  al. 
1988;  Rubin  1990). 

HOW  DOES  THE  LIPID 
ENVIRONMENT  AFFECT 
THE  FUNCTIONING  OF 
LIGAND-  AND  VOLTAGE- 
GATED  CHANNELS? 

As  already  discussed,  there  is  abundant 
evidence  that  exposure  to  alcohol  has 
measurable  effects  on  the  lipid  matrix 
of  the  cell  membrane.  However,  the 
mechanisms  by  which  these  effects 
translate  into  altered  functioning  of 
the  ion  channels  and  receptors  that 
are  the  ultimate  arbiters  of  neuronal 
activity  have  been  difficult  to  determine. 
It  is  especially  difficult  to  make  sense 
of  this  relationship  when  studying 
intact  cells,  with  their  complex  intracell- 
ular milieu  and  membrane.  However, 
I  believe  that  it  is  imperative  that  we 
understand  the  manner  in  which  lipids 
modulate  protein  activity  in  the  absence 
of  alcohol,  before  we  can  reasonably 
expect  to  understand  the  basis  for  the 
influence  of  lipid  environment  on 
alcohol's  actions  on  those  proteins. 

In  this  section,  I  will  first  describe 
data  that  focus  on  the  large  conductance 
calcium-activated  potassium  channel 
(BK  channel).  This  is  a  widely  present 
channel  in  the  nervous  system  and, 
because  it  is  activated  by  intracellular  cal- 
cium as  well  as  transmembrane  voltage, 
it  serves  to  link  different  channel  types, 
in  addition  to  intracellular  metabolic 
processes.  Functionally,  it  serves  impor- 
tant functions  both  in  shaping  individual 
action  potentials  and  in  controlling 


complex  firing  patterns  in  neurons.  It 
has  been  studied  in  situ  and  in  a  very 
reduced  preparation,  the  planar  lipid 
bilayer,  over  the  last  12  years.  The  effects 
of  the  lipid  microenvironment  on  chan- 
nel activity  have  been  described,  and 
some  hypotheses  for  the  biophysical  basis 
of  these  effects  have  been  put  forward. 

In  their  elucidation  of  lipid  effects 
on  this  channel,  Moczydlowski  and 
colleagues  (1985)  noted  that  one  of  the 
first  suggestions  that  the  lipid  micro- 
environment  has  significant  effects  on 
the  functioning  of  ion  channels  was 
put  forward  by  Frankenhauser  and 
Hodgkin  (1957),  who  invoked  the 
presence  of  membrane  surface  charge 
to  explain  the  shift  in  activation  of  Na 
and  K  currents  in  squid  axon  to  more 
depolarized  values  (i.e.,  less  excitable) 
when  the  external  Ca  concentration  was 
increased.  Frankenhauser  and  Hodgkin 
suggested  that  Ca  effectively  screened 
the  negative  charge  associated  with 
membrane  lipids.  However,  this 
hypothesis  is  difficult  to  prove  in  a 
complex  system.  In  the  experiments 
conducted  by  Moczydlowski  and  col- 
leagues, the  BK  channel  was  studied 
in  a  very  simplified  system,  in  which  the 
isolated  channel  protein  was  reconsti- 
tuted into  an  artificial  planar  lipid 
bilayer.  This  technique  has  proved  to 
be  a  very  powerful  method  to  elucidate 
the  role  of  lipids  in  channel  modulation. 
The  functioning  of  an  ion  channel  can 
be  broken  down  into  a  number  of 
separate  components,  including  the 
gating  component,  which  shifts  the 
channel  confirmation  between  con- 
ducting and  nonconducting  states;  the 
permeation  component,  which  is 
determined  by  the  ion-passing  pore;  and 


50 


Lipid  Involvement  in  Acute  Alcohol  Actions 


a  number  of  other  components,  such 
as  kinetic  aspects  of  function,  including 
the  inactivation  of  the  channel  after  it 
activates.  Single-channel  recording 
techniques  allow  the  examination  of 
each  of  these  parameters.  Considerable 
power  is  added  to  this  technique  by  the 
emerging  database  from  mutagenesis 
studies  relating  protein  sequence  to 
function.  In  relating  these  functional 
parameters  to  lipid  environment,  single- 
channel  recording  is  coupled  with  the 
planar  bilayer  technique,  in  which  the 
protein  of  interest  is  biochemically 
removed  from  the  native  membrane  and 
reconstituted  into  an  artificial  bilayer. 
Using  single -channel  recording  and 
planar  bilayer  techniques,  Moczydlowski 
and  colleagues  (1985)  reexamined  the 
effect  of  membrane  lipid  surface  charge 
on  ion  channel  function.  By  manipu- 
lating the  composition  of  the  bilayer, 
they  were  able  to  use  Gouy-Chapman- 
Stern  double  layer  theory  to  assess  the 
effect  of  lipid  surface  charge  on  channel 
conduction  properties.  Bilayers  were 
composed  of  either  phosphatidyleth- 
anolamine  (PE)  or  phosphatidylserine 
(PS),  allowing  comparison  of  a  neutral 
versus  a  negatively  charged  lipid  envi- 
ronment. The  conductance  of  the  chan- 
nel for  K  was  significantly  increased  in 
the  PS  bilayer,  compared  with  the  PE 
bilayer.  The  simplest  interpretation  of 
this  finding  is  that  the  K  and  Ca  concen- 
trations near  the  surface  of  a  negatively 
charged  membrane  are  greater  than 
the  concentration  of  these  ions  in  the 
bulk  solution.  This  would  both  shift 
the  gating  of  the  channel  toward  the 
open  state  (because  of  the  higher  con- 
centrations of  the  "agonist,"  Ca)  and 
provide  for  a  greater  K  flux  through 


the  open  channel  (because  of  the 
increased  driving  force  resulting  from 
the  increased  concentration  of  K).  A 
series  of  calculations  determined  that 
the  mouth  of  the  channel  did  not  "see" 
the  negative  charge  in  the  PS  bilayer 
immediately  adjacent  to  the  channel, 
but  rather  at  a  distance  of  approxi- 
mately 8-10  A.  They  interpreted  this 
to  represent  the  "insulation,"  presum- 
ably part  of  the  channel  protein,  of  the 
mouth  of  the  channel  from  the  lipid 
charge.  This  is  consistent  with  the  size 
of  channel  proteins,  which  are  as  large 
as  85  A  for  the  nicotinic  acetylcholine 
receptor  (nAChR),  whereas  the  thick- 
ness of  the  membrane  hydrocarbon 
layer  is  40  A.  The  gating  of  the  chan- 
nel was  also  "potentiated"  by  the 
presence  of  the  negative  charge,  with 
the  current- voltage  relationship  indi- 
cating that  less  depolarization  was 
necessary  to  shift  the  channel  to  the 
open  state  in  the  PS  bilayer,  compared 
with  the  PE  bilayer.  Once  again,  the 
protein  structure  appeared  to  insulate 
the  voltage -sensing  site  from  the  sur- 
rounding charge  by  a  value  <  10  A. 

The  interpretations  of  these  results 
are  predicated  upon  a  number  of 
assumptions.  First,  the  native  lipid  car- 
ried with  the  protein  must  exchange 
with  the  bilayer  lipid.  The  authors 
present  convincing  arguments  that  this 
is  the  case  (Moczydlowski  et  al.  1985). 
Less  easy  to  show  is  that  the  differ- 
ences are  due  solely  to  charge  and  do 
not  reflect  either  lipid-specific  lipid- 
protein  interactions  or  differences  in 
dipole  potential  between  the  two  lipids, 
which  cannot  be  definitively  ruled  out 
on  the  basis  of  the  experiments  per- 
formed. The  results  obtained  in  these 


51 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


experiments  demonstrate  "indirect" 
effects  of  the  lipid  environment  on 
protein  function,  as  contrasted  with 
other  effects  of  lipids,  in  which  they 
influence  the  conformation  or  integral 
properties  of  the  protein. 

I  will  next  describe  in  situ  studies  of 
the  BK  channel,  which  provide  further 
insights  into  the  lipid  modulation  of 
this  channel,  and  I  will  follow  that 
with  a  description  of  subsequent  planar 
bilayer  studies  that  help  to  illuminate 
the  results  obtained  in  the  natural 
membranes.  In  addition  to  their  role  in 
nerve  cells,  BK  channels  are  critical 
players  in  vascular  tissue.  Bregestovski 
and  colleagues  studied  the  role  of 
membrane  cholesterol  and  membrane 
fluidity  on  the  kinetic  properties  of  BK 
channels  in  cultured  vascular  smooth 
muscle  cells,  using  a  combination  of 
fluorescence  techniques  and  patch- 
clamping  (Bolotina  et  al.  1989).  They 
manipulated  the  cholesterol  content 
of  the  plasma  membrane  of  these  cells 
and  found  that  depletion  of  cholesterol 
caused  an  increase  in  D,  the  rotational 
diffusion  coefficient  of  a  fluorescent 
probe,  concurrent  with  a  ninefold 
increase  in  P0,  the  probability  of  the 
channel  being  in  the  open  state.  Con- 
versely, treatments  that  led  to  increased 
membrane  cholesterol  produced  a 
twofold  decrease  in  D  and  a  twofold 
decrease  in  P0.  Alterations  in  P0  could 
be  explained  by  a  redistribution  of  open 
and  closed  times  of  the  channel,  reflect- 
ing the  thermodynamic  stability  of  the 
channel  in  each  of  these  states.  These 
changes  in  channel  gating  were  unac- 
companied by  any  change  in  the  unitary 
conductance  of  the  channel,  high- 
lighting the  fact  that  all  regions  of  the 


protein  are  not  similarly  responsive  to 
the  putative  change  in  membrane  fluid- 
ity. Since  these  experiments  were  per- 
formed with  "ripped-off  patches  in 
the  inside-out  configuration,  the 
influence  of  intracellular  milieu  and 
cytoskeleton  is  minimized.  There  is 
reasonable  evidence  that  the  influence 
of  cholesterol  on  channel  gating  does 
not  result  from  direct  interaction  of 
the  lipid  with  the  channel,  or  with  a 
lipid  annulus  associated  with  the 
channel,  but  rather  reflects  changes  in 
the  bulk  (or  some  subbulk  compart- 
ment) membrane  lipid  properties. 

Work  from  Gruener  and  colleagues 
has  provided  a  mechanistic  framework 
for  the  actions  of  cholesterol  on  the 
gating  of  the  BK  channel,  and  a  model 
for  a  class  of  lipid-protein  interaction 
(Chang  et  al.  1995).  This  group,  using 
brain  BK  channels  reconstituted  in 
PE/PS  lipid  bilayers,  confirmed  the 
finding  made  in  natural  membranes, 
that  increased  membrane  cholesterol 
decreased  the  open  probability  of  the 
channel.  They  also  tested  the  idea  that 
since  cholesterol  is  known  to  change 
the  order  state  and  the  modulus  of 
compressibility  of  bilayers,  altered  pro- 
tein conformation  may  result  from  an 
overall  increase  in  the  lateral  stress  in 
the  bilayer,  transmitted  to  the  channel 
and  biasing  it  into  the  closed  state.  It 
was  found  that  there  was  a  relatively 
sharp  transition  of  the  P0,  with  short- 
ened openings,  at  cholesterol  concen- 
trations around  10  percent.  Below 
and  above  this  concentration  of  choles- 
terol, there  was  little  concentration- 
dependent  effect  on  channel  function. 
Data  obtained  from  small  angle  x-ray 
diffraction  of  the  bilayers  indicated 


52 


Lipid  Involvement  in  Acute  Alcohol  Actions 


that  at  cholesterol  concentrations  that 
alter  channel  activity  the  liquid  crys- 
talline/gel phase  composition  of  the 
membrane  was  altered,  and  the  gel  phase 
was  no  longer  present.  The  authors  also 
constructed  Arrhenius  plots,  using  temp- 
erature as  a  modulator  of  channel 
activity,  and  determined  the  thermo- 
dynamic properties  of  the  BK  channel 
open-to-closed  transition  as  a  function 
of  temperature.  They  concluded  that 
the  calculated  reduction  in  activation 
energy  required  to  move  the  channel 
from  the  open  to  the  closed  state  is 
consistent  with  the  hypothesis  that 
cholesterol  destabilizes  the  open  state 
of  the  channel,  causing  it  to  close 
sooner  than  in  the  absence  of  choles- 
terol. Estimates  of  the  lateral  elastic 
stress  energy  produced  by  cholesterol 
are  higher  than  estimates  of  the  activa- 
tion energy  required  to  move  the 
channel  from  the  open  to  the  closed 
state,  consistent  with  the  bias  into  the 
closed  state  by  lateral  elastic  stress.  In 
other  words,  the  conformational  change 
from  the  closed  to  the  open  state 
involves  an  increase  in  protein  volume, 
generating  lateral  stress  force,  gener- 
ating, in  turn,  a  counterforce  deflected 
back  on  the  channel.  The  magnitude  of 
this  force  would  depend  on  the  mod- 
ulus of  compressibility  of  the  mem- 
brane, and  would  facilitate  the  return 
of  the  channel  to  the  closed  state  at 
different  rates.  The  authors  go  on  to 
describe  values  of  enthalpy,  entropy, 
and  free  energy  calculated  from  their 
data,  consistent  with  this  interpretation 
(Chang  et  al.  1995).  While  this  type 
of  analysis  is  far  from  foolproof,  the 
composite  work  done  with  this  channel 
and  lipid  modulation  provides  some 


insights  into  approaches  that  will 
likely  be  the  most  productive. 

The  previous  discussion  focused  on 
the  Ca-activated  potassium  channel, 
but  a  similar  type  of  analysis  of  lipid 
influence  on  channel  function  has  also 
been  done  for  other  channels,  includ- 
ing ligand-gated  channels,  such  as  the 
nAChR.  Barrantes  (1993)  explored 
the  role  of  the  motionally  restricted 
shell  of  lipids  typically  referred  to  as  the 
"annulus  lipids,"  immediately  adjacent 
to  the  nAChR  protein  and  other  pro- 
teins. These  lipids  do  exchange  with 
adjacent  membrane  lipids,  but  at  a 
slower  rate  than  those  lipid  molecules 
that  are  not  part  of  the  annulus.  Bar- 
rantes outlined  a  number  of  influences 
of  the  lipid  environment  on  the  func- 
tioning of  the  nAChR  The  findings 
include  the  fact  that  cholesterol  and 
negatively  charged  phospholipids  are 
necessary  for  receptor  function,  and 
that  fatty  acids  can  block  the  activity 
of  the  receptor.  Although  most  head- 
group  modifications  were  without  sig- 
nificant effect,  exposure  of  the  inner 
face  of  the  membrane  to  PE  (in 
ripped-off  patches)  affected  gating  by 
reducing  the  channel  open  time. 
Other  phospholipids  did  not  have  this 
effect.  Barrantes  suggested  that  this 
PE  effect  may  reflect  the  perturbation 
of  the  normal  membrane  asymmetry, 
in  which  PE  is  predominant  in  the 
exofacial  leaflet. 

Miller  and  colleagues  have  also 
provided  information  on  the  influence 
of  membrane  lipids  in  nAChR  func- 
tion. They  have  used  ethidium  to 
measure  nACh  channel  activity,  allow- 
ing fast  kinetic  analysis,  necessary  to 
avoid  complications  from  receptor 


53 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


desensitization  (Rankin  et  al.  1997). 
Both  in  natural  membrane  preparations 
and  in  artificial  bilayers,  these  investi- 
gators confirmed  that  cholesterol  is 
necessary  for  receptor  function,  but 
that  the  action  of  cholesterol  is  specific 
to  particular  aspects  of  receptor  func- 
tion. While  the  baseline  activity  of  the 
channel  was  relatively  similar  in  differ- 
ent lipid  environments,  the  ability  to 
reach  the  open  state  from  the  resting 
state  was  cholesterol  dependent,  but 
the  transition  from  the  open  to  the  fast 
desensitized  state  was  not.  Miller  and 
colleagues  have  proposed  a  new  form 
of  posttranslational  processing,  stating 
that  nAChR  channels  are  not  primed  by 
cholesterol  until  they  are  inserted  into 
the  membrane  (Rankin  et  al.  1997). 
Work  is  proceeding  in  Miller's  and 
collaborators'  laboratories  to  determine 
the  nature  of  the  cholesterol  influence, 
although  they  present  evidence  sug- 
gesting that  a  simple  alteration  of 
membrane  fluidity  is  unlikely  to  be 
the  determinant.  In  addition,  work  is 
ongoing  to  determine  the  influence  of 
lipids  such  as  cholesterol  on  alcohol 
sensitivity  of  the  receptor  channel. 

In  my  laboratory,  we  have  been 
focusing  on  the  function  and  alcohol 
modulation  of  proteins  involved  in  the 
secretion  of  vasopressin  from  neuro- 
hypophysial terminals.  One  of  these  is 
the  BK  channel  described  in  detail 
above,  and  for  which  a  significant 
body  of  data  describing  the  influence 
of  membrane  lipids  exists.  We  have 
amassed  quite  a  bit  of  understanding 
of  the  actions  of  alcohol  on  the  BK 
channel  in  situ,  using  both  macroscopic 
and  single-channel  recording  tech- 
niques (Dopico  et  al.  1996,  1998)  in 


nerve  terminals  of  hypothalamic  neu- 
rons. Reasonable  concentrations  of 
alcohol  potentiate  channel  activity, 
primarily  by  altering  the  gating  of  the 
channel.  Other  parameters,  such  as 
voltage  dependency  and  ion  selectiv- 
ity, appear  unaffected  by  the  drug.  In 
addition,  we  are  able  to  monitor  the 
activity  of  cloned  BK  channels  in 
expression  systems  and  to  assess  alco- 
hol action  in  this  situation.  These 
studies  have  allowed  us  to  observe  the 
manner  in  which  alcohol  interacts 
with  the  channel,  leading  to  some 
specific  hypotheses,  such  as  that  alco- 
hol acts  as  a  partial  agonist  at  the  Ca- 
binding  site  within  the  channel  protein 
(Dopico  et  al.  1998).  The  large  body 
of  data  that  we  have  collected  on  this 
channel  in  situ  and  in  expression  systems 
allows  us  to  now  explore  the  role  of 
lipid  environment  on  alcohol's  actions 
on  the  channel.  To  this  end,  we  have 
begun  studies  on  the  BK  channel  recon- 
stituted into  planar  lipid  bilayers  (Chu 
et  al.  1998).  These  studies  are  still  at  an 
early  stage,  but  we  have  found  that 
the  reconstituted  channel  is  potentiated 
by  alcohol  in  a  manner  qualitatively 
similar  to  the  channel  in  situ  and  in 
expression  systems.  The  potentiation  is 
not  significantly  different  in  PE  versus 
PE/PS  bilayers,  although  baseline  char- 
acteristics of  the  channel  are  altered  in  a 
manner  consistent  with  exchange  of  the 
native  lipid  with  the  lipid  constituents 
of  the  artificial  bilayer.  We  are  encour- 
aged by  these  early  results,  and  hopeful 
that  this  approach  will  yield  insights 
not  possible  without  integrating  results 
focused  on  a  single  protein. 

In  addition,  we  have  discovered  that 
acute  alcohol  suppression  of  vasopressin 


54 


Lipid  Involvement  in  Acute  Alcohol  Actions 


release  from  the  terminals  taken  from 
rats  that  have  been  chronically  exposed 
to  alcohol  is  lessened  in  comparison  to 
the  suppression  seen  in  terminals  from 
alcohol-naive  rats.  Of  course,  the  ter- 
minal Ca  and  BK  channels  discussed 
above  are  an  important  part  of  the 
release  machinery.  The  ability  to 
explore  the  activity  of  the  BK  channel 
both  in  situ  and  in  artificial  bilayers 
will  allow  a  full  examination  of  both 
protein  and  lipid  alterations  in  the 
mechanisms  of  this  tolerance. 

CONCLUSIONS  AND 
RECOMMENDATIONS 

The  complexity  of  natural  membranes 
and  the  numerous  and  interlinked 
lipid  metabolism  pathways  make  a  rea- 
sonable analysis  of  lipid  involvement 
in  alcohol's  actions  in  intact  animals 
and  tissue  problematic.  Contrasting  the 
complexity  of  the  situation  with  lipids, 
recent  developments  in  the  study  of 
ligand-  and  voltage-gated  channels 
have  made  the  study  of  alcohol  effects 
on  proteins  significantly  more  direct. 
The  advent  of  cloning,  expression, 
and  mutagenesis  of  putative  target 
proteins  allows  the  testing  of  specific 
hypotheses  regarding  the  relationship 
between  protein  structure  and  alcohol 
action.  However,  we  should  not  for- 
get that  these  expressed  proteins  are 
operating  in  a  lipid  environment,  and 
as  already  shown,  protein  function  is 
strongly  influenced  by  this  environ- 
ment. Since,  ultimately,  our  under- 
standing of  alcohol's  effects  on  nervous 
system  function  will  derive  from  alter- 
ations of  protein  function,  our  explo- 
ration of  the  role  of  lipids  in  alcohol's 


actions  must  not  be  done  in  isolation 
from  the  impact  on  protein  function. 
Once  again,  I  would  stress  a  basic  tenet 
of  my  commentary,  which  is  to  say  that 
phrasing  the  question  as  whether  alco- 
hol acts  primarily  on  the  lipid  or  the 
protein  is  unproductive;  rather,  we  must 
ask  how  the  interactions  between  pro- 
tein subunits,  lipids,  and  water  are 
affected  by  alcohol.  A  few  suggestions 
for  how  to  accomplish  this  follow: 

1 .  The  function  of  a  protein  should 
serve  as  the  readout  of  the  lipid 
perturbations.  Measurements  of 
lipid  perturbations  without  a  pro- 
tein "sensor,"  even  when  employ- 
ing elegant  biophysics,  are  not 
going  to  be  as  productive  as  those 
studies  that  approach  the  system  as 
a  dynamic  interaction  between  the 
various  components. 

2.  The  operation  of  the  protein 
should  be  well  understood  before 
attempts  are  made  to  assess  the 
perturbation  of  function  by  alco- 
hol. Ideally,  this  understanding  will 
include  information  in  the  native 
membrane,  as  well  as  in  artificial 
bilayers.  The  functioning  of  mem- 
brane proteins  is  becoming  under- 
stood at  a  very  sophisticated  level. 
For  ligand-  and  voltage-gated 
channels,  this  knowledge  includes 
an  understanding  of  the  modular 
nature  of  individual  proteins,  with 
conserved  stretches  of  amino  acids 
controlling  discrete  functions,  such 
as  voltage  dependency,  permeation, 
inactivation,  desensitization,  and  so 
on.  This  type  of  knowledge  will 
greatly  facilitate  the  testing  of 


55 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


hypotheses  regarding  the  interac- 
tion of  alcohol,  lipids,  and  proteins. 
Similar  structure -function  relation- 
ships are  also  becoming  available 
for  nonchannel  proteins  important 
in  nervous  system  function. 

3.  Reasonable  concentrations  of  alcohol 
should  be  used.  This  is  a  difficult 
problem,  since  an  argument  is  often 
made  that  our  physical  measurement 
techniques  are  not  sophisticated 
enough  (e.g.,  may  not  be  sampling  a 
subcompartment  where  the  effects 
are  larger)  to  detect  important 
changes  occurring  at  relevant  con- 
centrations. Although  this  argument 
may  be  reasonable,  there  are  many 
processes  that  can  be  successfully 
studied  at  reasonable  concentrations 
of  alcohol  with  current  technology. 
The  alcohol  field  should  wait  for 
the  appropriate  technology  before 
attacking  problems  that  require 
very  high  drug  concentrations  to  see 
measurable  effects.  Parenthetically, 
a  case  can  be  made  that  when  tech- 
nology development  is  critical  for  the 
testing  of  an  important  hypothesis 
regarding  alcohol  perturbation  of 
the  lipid  environment,  such  studies 
should  be  supported  enthusiastically, 
even  if  they  will  not  involve  alcohol 
at  the  early  stages  of  the  research. 

4.  Studies  should  be  integrated  when- 
ever possible.  The  ideal  system  will 
have  certain  attributes  and  will 
allow  data  collection  at  multiple 
levels:  (a)  a  target  protein  of 
known  behavioral  or  physiological 
relevance  to  alcohol  action;  (b)  the 
effects  of  lipid  perturbation  and 


possibly  modulation  of  lipid  com- 
position on  the  protein  in  its  native 
environments;  (c)  a  protein  that  can 
be  reconstituted  into  a  simplified 
lipid  environment  for  examining 
alcohol's  effects;  and  (d)  a  protein 
that  has  been  cloned,  allowing  for 
expression  in  a  variety  of  native 
membranes,  and  for  mutagenesis 
studies.  Also,  as  it  is  becoming 
apparent  that  the  function  of  most 
membrane  channels  and  receptors 
actually  reflects  communities  of 
coupled  proteins,  ideally  we  will  be 
able  to  co-express  combinations  of, 
for  example,  channel  subunits,  to 
explore  the  role  of  lipids  in  their 
interactions  and  the  actions  of  alco- 
hol on  these  interactions,  resulting 
in  altered  function. 

5.  We  should  not  lose  sight  of  the  fact 
that  lipids  appear  to  play  a  major  role 
in  the  compensatory  responses  of  the 
cell  to  chronic  exposure  to  the  drug, 
and  these  responses  should  continue 
to  be  explored.  However,  whenever 
possible,  these  studies  should  also  be 
performed  within  the  framework 
described  in  the  preceding  sugges- 
tion. The  response  to  alcohol  is  likely 
to  be  both  short  term  and  long  term. 
In  the  short  term,  we  might  expect 
posttranslational  changes  in  protein, 
such  as  phosphorylation  and/or 
changes  in  acyl  chains  of  phospho- 
lipids. In  the  longer  term,  we  might 
predict  shifts  in  the  subunit  com- 
position of  the  channel  protein  or  in 
the  phospholipid  headgroup  popu- 
lation, with  changes  in  (e.g.) 
charge  characteristics  or  the  com- 
partmentalization  of  cholesterol. 


56 


Lipid  Involvement  in  Acute  Alcohol  Actions 


Of  course,  these  are  just  a  few  of 
very  many  possibilities. 

Each  of  the  separate  approaches, 
taken  alone,  has  shortcomings.  For 
example,  for  the  reductionist  approach, 
I  realize  that  proteins  do  not  exist  in 
one-  or  two-lipid  environments  (prob- 
ably). However,  the  information 
obtained  from  these  simple  experiments 
can  be  used  to  derive  hypotheses 
testable  in  more  complicated  environ- 
ments. Of  course,  information  will 
flow  the  other  way  as  well,  with  infor- 
mation (some  already  present  in  the 
literature)  obtained  in  complex  mem- 
brane environments  leading  to  experi- 
ments performed  in  the  simplified 
bilayer.  There  is  reasonable  evidence 
that  lipids  play  a  role  in  the  actions  of 
alcohol  on  proteins.  The  explosion  of 
information  on  the  functioning  of 
neural  membrane  channels,  as  well  as 
other  proteins,  presents  a  wonderful 
opportunity  to  obtain  significant 
insights  into  this  role. 

ACKNOWLEDGMENT 

I  would  like  to  thank  the  National 
Institute  on  Alcohol  Abuse  and  Alco- 
holism for  financial  support. 

REFERENCES 

Barrantes,  F.J.  Structural -functional  corre- 
lates of  the  nicotinic  acetylcholine  receptor 
and  its  lipid  microenvironment.  FASEBJ 
7:1460-1467,  1993. 

Barry,  J.A.,  and  Gawrisch,  K.  Direct 
NMR  evidence  for  ethanol  binding  to 
the  lipid- water  interface  of  phospholipid 


bilayers.  Biochemistry  33:8082-8088, 
1994. 

Bolotina,  V.;  Omelyanenko,  V.;  Heyes,  B.; 
Ryan,  U.;  and  Bregestovski,  P.  Variations 
of  membrane  cholesterol  alter  the  kinetics 
of  Ca2+-dependent  K+  channels  and  mem- 
brane fluidity  in  vascular  smooth  muscle 
cells.  PflugersArch  415:262-268,  1989. 

Chang,  H.M.;  Reitstetter,  R;  Mason,  RP.; 
and  Gruener,  R.  Attenuation  of  channel 
kinetics  and  conductance  by  cholesterol: 
An  interpretation  using  structural  stress  as 
a  unifying  concept.  J Membr  Biol  143: 
51-63,  1995. 

Channareddy,  S.;  Jose,  S.S.;  Eryomin,  V.A.; 
Rubin,  E.;  Taraschi,  T.F.;  and  Janes,  N. 
Saturable  ethanol  binding  in  rat  liver 
microsomes.  J  Biol  Chem  271:17625- 
17628,  1996. 

Chin,  J.H.,  and  Goldstein,  D.B. 
Membrane-disordering  action  of  ethanol: 
Variation  with  membrane  cholesterol 
content  and  depth  of  the  spin  labeled 
probe.  Mol Pharmacol  19:425^31,  1981. 

Chu,  B.;  Dopico,  A.M.;  and  Treistman, 
S.N.  Ethanol  potentiation  of  calcium- 
activated  potassium  channels  reconstituted 
into  planar  lipid  bilayers.  Mol  Pharmacol 
54:397-406,  1998. 

Deitrich,  R.A.;  Dunwiddie,  TV.;  Harris, 
RA.;  and  Erwin,  V.G.  Mechanism  of  action 
of  ethanol:  Initial  central  nervous  system 
actions.  Pharmacol  Rev  41:489-537,  1989. 

Devaux,  P.F.,  and  Zachowski,  A.  Main- 
tenance and  consequences  of  membrane 
phospholipid  asymmetry:  Lipid  domains 
in  model  and  biological  membranes. 
Chem  Phys  Lipids  73:107-120,  1994. 

Dopico,  A.M.;  Lemos,  J.R;  and  Treistman, 
S.N.  Ethanol  increases  the  activity  of  large 
conductance,  Ca2+-activated  K+  channels 


57 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  isolated  neurohypophysial  terminals. 
Mol  Pharmacol  49:40-48,  1996. 

Dopico,  A.M.;  Anantharam,  V.;  and 
Treistman,  S.N.  Ethanol  increases  the 
activity  of  Ca++-dependent  K+  ( mslo) 
channels:  Functional  interaction  with 
cytosolic  Ca++ .  /  Pharmacol  Exp  Ther 
284:258-268,  1998. 

Ellingson,  J.S.;  Taraschi,  T.F.;  Wu,  A.; 
Zimmerman,  R.;  and  Rubin,  E.  Cardio- 
lipin  from  ethanol-fed  rats  confers  tolerance 
to  ethanol  in  liver  mitochondrial  mem- 
branes. Proc  Natl  Acad  Sci  USA  85:3353- 
3357,  1988. 

Frankenhauser,  B.,  and  Hodgkin,  A.L.  The 
action  of  calcium  on  the  electrical  proper- 
ties of  squid  axons.  J  Physiol  137:218- 
244,  1957. 

Gennis,  R.B.  Lateral  and  transverse  asym- 
metry in  membranes.  In:  Cantor,  C.R., 
ed.  Springer  Advanced  Texts  in  Chemistry: 
Biomembranes.  Molecular  Structure  and 
Function.  New  York:  Springer- Verlag, 
1989. pp. 138-165. 

Goldstein,  D.B.  Pharmacology  of  Alcohol. 
New  York:  Oxford,  1983. 

Goldstein,  D.B.  The  effects  of  drugs  on 
membrane  fluidity.  Annu  Rev  Pharmacol 
Toxicol  24:43-64,  1984. 

Harris,  RA.;  Groh,  G.I.;  Baxter,  D.M.;  and 
Hitzemann,  RJ.  Gangliosides  enhance  the 
membrane  actions  of  ethanol  and  pentobar- 
bital. Mol  Pharmacol  2SA10-417,  1984. 

Ho,  C.;  Kelly,  M.B.;  and  Stubbs,  CD. 
The  effects  of  phospholipid  unsaturation 
and  alcohol  perturbation  at  the  protein/ 
lipid  interface  probed  using  fluorophore 
lifetime  heterogeneity.  Biochim  Biophys 
Acta  1193:307-315,  1994. 

Hoke,  L.L.,  and  Gawrisch,  K.  Determining 
ethanol  distribution  in  phospholipid 


multilayers  with  MAS-NOESY. 
Biochemistry  36:4669^*674,  1997. 

Janes,  N.;  Hsu,  J.W.;  Rubin,  E.;  and 
Taraschi,  T.F.  Nature  of  alcohol  and 
anesthetic  action  on  cooperative  mem- 
brane equilibria.  Biochemistry 
31:9467-9472,  1992. 

Jorgensen,  K.;  Ipsen,  J.H.;  Mouritsen, 
O.G.;  and  Zuckermann,  M.J.  The  effect 
of  anesthetics  on  the  dynamic  heterogene- 
ity of  lipid  membranes.  Chem  Phys  Lipids 
65:205-216,  1993. 

Moczydlowski,  E.;  Alvarez,  O.;  Vergara, 
C.;  and  Latorre,  R  Effect  of  phospholipid 
surface  charge  on  the  conductance  and 
gating  of  a  Ca2+-activated  K+  channel  in 
planar  lipid  bilayers.  J  Membr  Biol 
83:273-282,  1985. 

Orme,  F.W.;  Moronne,  M.M.;  and 
Macey,  R.I.  Modification  of  erythrocyte 
membrane  dielectric  constant  by  alcohols. 
J  Membr  Biol  104:57-68,  1988. 

Pawlosky,  R.J.,  and  Salem,  N.,  Jr.  Ethanol 
exposure  causes  a  decrease  in  docosahex- 
aenoic  acid  and  an  increase  in  docosapen- 
taenoic  acid  in  feline  brains  and  retinas. 
Am  J  Clin  Nutr  61:1284-1289,  1995. 

Rankin,  S.E.;  Addona,  G.H.;  Kloczewiak, 
M.A.;  Bugge,  B.;  and  Miller,  K.W.  The 
cholesterol  dependence  of  activation  and 
fast  desensitization  of  the  nicotinic  acetyl- 
choline receptor.  Biophys  J  73:2446- 
2455,  1997. 

Rottenberg,  H.  Probing  the  interactions 
of  alcohols  with  biological  membranes 
with  the  fluorescent  probe  Prodan. 
Biochemistry  31:9473-9481,  1992. 

Rubin,  H.  On  the  nature  of  enduring 
modifications  induced  in  cells  and  organ- 
isms. Am  J  Physiol  25&±19-L24,  1990. 


58 


Lipid  Involvement  in  Acute  Alcohol  Actions 


Salem,  N.,  Jr.  Alcohol,  fatty  acids,  and 
diet.  Alcohol  Health  Res  World 
13:211-218,  1989. 

Treistman,  S.N.,  and  Wilson,  A.  Alkanol 
effects  on  early  potassium  currents  in  Aplysia 
neurons  depend  on  chain  length.  Proc  Natl 
Acad  Set  USA  84:9299-9303,  1987a. 

Treistman,  S.N.,  and  Wilson,  A.  Effects  of 
ethanol  on  early  potassium  currents  in 
Aplysia:  Cell  specificity  and  influence  of 
channel  state.  J  Neurosci  7:3207-3214, 
1987b. 

Treistman,  S.N.;  Moynihan,  M.;  and 
Wolf,  D.E.  Influence  of  alcohols,  temper- 


ature, and  region  on  the  mobility  of  lipids 
in  neuronal  membrane.  Biochim  Biophys 
Acta  898:109-120,  1987. 

Welti,  R,  and  Glaser,  M.  Lipid  domains 
in  model  and  biological  membranes. 
Chem  Phys  Lipids 73:121-137 ',  1994. 

Wood,  W.G.;  Schroeder,  F.;  Rao,  A.M.; 
Igbavboa,  U.;  and  Avdulov,  NA. 
Membranes  and  ethanol:  Lipid  domains 
and  lipid-protein  interactions.  In: 
Deitrich,  R.A.,  and  Erwin,  V.G.,  eds. 
Pharmacological  Effects  of  Ethanol  on  the 
Nervous  System.  Boca  Raton:  CRC  Press, 
1996.  pp.  13-27. 


59 


Chapter  3 

Effects  of  Alcohol  on  the 
Neuroendocrine  System 

Catherine  Rivier,  Ph.D. 


KEY  WORDS:  AODE  (effects  of  AOD  [alcohol  or  other  drug]  use,  abuse,  and 
dependence);  endocrine  system;  hypothalamus-pituitary  axis;  pituitary-adrenal 
axis;  corticotropin  RH  (releasing  hormone);  glucocorticoids;  cytokines;  brain 
function;  adrenocorticotropic  hormone;  homeostasis;  neurotransmitters;  biological 
regulation;  gonad  function;  gonadotropin  RH;  luteinizing  hormone;  animal 
study;  literature  review 


Research  on  the  effects  of  alcohol  as 
they  pertain  to  neuroscience  can  be 
broadly  divided  into  three  parts:  (1) 
the  search  for  the  mechanisms  that 
lead  to  excessive  alcohol  consumption 
and  addiction  and  the  associated  syn- 
dromes of  withdrawal  and  relapse;  (2) 
the  investigation  of  the  pharmacologi- 
cal influence  of  alcohol  on  molecular, 
cellular,  and  system  biology,  which  is 
a  consequence  of  alcohol  exposure/ 
consumption;  and  (3)  the  study  of  the 
possible  contributing  effect  of  specific 
hormones — in  particular,  those  of  the 
hypothalamic -pituitary- adrenal  (HPA) 
axis — on  alcohol  consumption. 


Alcohol  exerts  a  wide  spectrum  of 
effects,  which  affect  virtually  every  cell 
in  the  body.  It  is  not  entirely  clear 
whether  alcohol  exerts  similar  effects 
on  most  signaling  pathways  (e.g.,  by 
similarly  altering  cyclic  adenosine 
monophosphate  (cAMP)-dependent 
processes,  or  gene  transcription,  or 
binding  of  ligands  to  their  receptors), 
or  whether  these  effects  are  system 
specific.  Indeed,  the  ability  of  the 
drug  to  indiscriminately  distribute 
itself  throughout  the  body  (including 
the  brain)  renders  studies  of  its  specific 
influence  within  a  particular  system 
difficult.  Also,  because  alcohol  does 


C.  Rivier,  Ph.D.,  is  a  professor  at  The  Salk  Institute,  The  Clayton  Foundation  Laboratories  for 
Peptide  Biology,  10010  North  Torrey  Pines  Rd.,  La  folia,  CA  92037-1099. 


61 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


not  have  a  receptor,  the  mechanisms 
through  which  it  alters  cellular  function 
are  not  easy  to  elucidate.  It  is  important 
to  keep  these  points  in  mind  when 
reviewing  what  is  known  and  suggest- 
ing topics  for  future  emphasis. 

This  chapter  focuses  on  the  neuro- 
endocrine influence  of  alcohol,  a  topic 
that  was  the  focus  of  a  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
(NIAAA)  workshop  and  monograph 
(Zakhari  1993).  I  will  therefore  present 
only  a  brief  overview  of  the  alcohol - 
related  literature  and  will  list  selected 
topics  of  interest  for  NIAAA  that 
either  represent  a  logical  extension  of 
present  NIAAA-supported  programs 
or  are  not  represented  in  the  portfolio. 

SCIENTIFIC  REVIEW 

Study  of  the  molecular  and  cellular 
aspects  of  the  effects  of  alcohol,  which  is 
routinely  done  in  isolated  cell  systems, 
presents  the  challenge  of  determining 
the  molar  concentrations  of  the  drug 
within  which  results  can  be  interpreted 
as  being  relevant  for  the  whole  organ- 
ism. Study  of  the  influence  of  alcohol 
in  the  intact  animal,  on  the  other  hand, 
presents  the  challenge  of  determining 
whether  results  obtained  with  forced 
exposure  to  alcohol  are  relevant  for 
conditions  associated  with  spontaneous 
alcohol  consumption,  or  whether  one 
should  dissociate  between  the  two. 
Most  of  the  results  in  the  published 
literature  were  obtained  in  laboratory 
animals  exposed  to  alcohol  through 
an  experimenter- controlled  procedure. 
This  is  primarily  because  unselected 
animals  do  not  spontaneously  drink 
alcohol  or,  if  they  are  forced  to  do  so 


(e.g.,  when  presented  with  a  chocolate- 
based  alcohol  diet  as  die  sole  source  of 
nutrients),  only  consume  it  in  limited 
amounts.  There  is  no  doubt  that  results 
obtained  with  this  experimental  approach 
have  been  very  interesting  and  useful. 
Nevertheless,  it  is  important  to  remain 
cognizant  of  the  possibility  that  at  least 
some  of  the  biological  effects  of  alcohol 
may  be  different  in  animals  forcibly 
exposed  to  alcohol  from  those  that 
self- administer  the  drug  (Moolten  and 
Kornetsky  1990).  If  there  are  differ- 
ences between  the  two  paradigms,  they 
may  represent  the  fact  that  animals 
that  are  not  selected  for  alcohol  self- 
administration  usually  consume  small 
amounts  of  alcohol  and  therefore  only 
reach  low  blood  alcohol  levels  (BALs); 
that  forced  exposure  to  alcohol  may 
induce  aversiveness;  and  whether  or  not 
there  are  rewarding  efFects.  It  is  extremely 
important  to  determine  the  origin  of 
these  differences  in  alcohol's  effects 
because  investigators  may  pursue  mech- 
anisms that  are  not  those  that  underlie 
the  effect  of  alcohol  during  spontaneous 
consumption.  Although  I  do  not  advo- 
cate abandoning  models  based  on 
experimenter-induced  alcohol  exposure, 
the  field  would  greatly  benefit  from 
the  increased  availability  of  and  ease  of 
access  to  animals  selected  for  the 
spontaneous  consumption  of  mean- 
ingful amounts  of  alcohol. 

HPAAxis 

Many  studies  have  shown  that  alcohol 
administration  to  laboratory  rodents 
causes  a  rapid  and  significant  activation 
of  the  HPA  axis  (Rivier  1996). 
Increased  levels  of  corticotropin- 
releasing  factor  (CRT)  and  possibly 


62 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


vasopressin  in  the  brain  are  important 
in  modulating  the  effect  of  the  drug 
on  this  axis  (Rivier  et  al.  1984;  Car- 
mona-Calero  et  al.  1995;  Rivier  and 
Lee  1996).  There  is  some  controversy 
regarding  the  acute  effect  of  alcohol 
in  humans;  some  investigators  claim 
that  increased  HPA  axis  function  in 
human  volunteers  consuming  an  alco- 
holic beverage  is  only  present  in  the 
subjects  who  experience  gastrointesti- 
nal discomfort  (Inder  et  al.  1995 b). 
Nevertheless,  the  consensus  appears 
to  be  that  humans  who  consume  large 
amounts  of  alcohol  exhibit  increased 
HPA  axis  activity  (though  with  a  great 
deal  of  variability  [Wand  1993]),  as 
indicated  by  the  fact  that  enhanced 
basal  Cortisol  production  is  found  in 
some  alcoholics  (Wand  and  Dobs 
1991)  and  can  lead  to  a  pseudo-Cush- 
ing's  syndrome  (Veldman  and  Mein- 
ders  1996).  To  state  that  the  HPA 
axis  of  alcoholics  is  activated,  however, 
is  simplistic.  Indeed,  possibly  as  a  con- 
sequence of  increased  corticosteroid 
feedback  and/or  down- regulated  pitu- 
itary CRF  receptors,  many  alcoholics 
show  a  blunted  response  to  exoge- 
nous CRF  injection  or  exposure  to  a 
non-alcohol-related  stress  (Wand  and 
Dobs  1991).  Furthermore,  the  HPA 
axis  remains  pathologically  altered  in 
short-term  abstinent  alcoholics,  who 
also  show  blunted  responses  to  CRF 
or  exposure  to  non-alcohol  stresses 
(Heuser  et  al.  1988;  Adinoff  et  al. 
1990;  Inder  et  al.  1995a;  Ehrenreich 
et  al.  1997).  Therefore,  while  there 
appears  to  be  reasonable  evidence  for 
alcohol-induced  changes  in  the  HPA 
axis  of  humans  who  abuse  this  drug, 
much  remains  to  be  investigated. 


The  importance  of  studies  of  the 
effect  of  alcohol  on  the  HPA  axis  of 
any  mammalian  species  extends 
beyond  the  mechanisms  that  they  will 
uncover.  Because  of  its  pivotal  influ- 
ence as  a  general  regulator  and  coor- 
dinator of  the  stress  responses,  and 
because  its  hormones  exert  such  a 
wide  range  of  effects,  changes  in  the 
activity  of  this  axis  are  likely  to  con- 
tribute to  many  of  the  effects  of  alcohol. 
Although  CRF  is  the  primary  regula- 
tor of  the  HPA  axis  (Rivier  and  Plot- 
sky  1986),  this  peptide  also  exerts 
many  other  effects.  Thanks  to  studies 
of  the  distribution  of  CRF  through 
the  brain,  its  pharmacological  effects 
on  a  wide  array  of  parameters,  and  the 
consequence  of  immunoneutralizing 
it  or  blocking  its  receptors,  we  now 
know  that  this  peptide  also  controls  or 
participates  in  the  regulation  of  the 
hypo  thalamic -pituitary -gonadal 
(HPG)  axis,  growth  hormone  release, 
gastrointestinal  functions,  and  natural 
killer  cell  activity,  which  it  inhibits 
(Rivier  and  Vale  1984;  Irwin  et  al. 
1990;  Tache  et  al.  1993;  Rivest  and  Riv- 
ier 1995);  opioids  and  catecholamines, 
which  it  stimulates  (Brown  and  Fisher 
1985;  Boyadjieva  et  al.  1997);  depres- 
sion, which  it  appears  to  induce 
(Nemeroff  1996);  and  anxiety,  which 
it  promotes  (Koob  et  al.  1993).  Any 
alterations  in  CRF  production  and  the 
activity  of  CRF -dependent  circuitries 
(such  as  those  seen  after  alcohol)  will 
therefore  have  profound  conse- 
quences for  the  organism,  both  under 
basal  conditions  and  during  attempts 
to  restore  homeostasis.  A  full  and 
complete  knowledge  of  what  alcohol 
does  and  how  is  therefore  crucial. 


63 


NlAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Another  important  point  is  that  in 
experimental  animals,  CRF  has  been 
shown  to  induce  or  participate  in  many 
responses  that  are  very  similar  to  those 
associated  with  fetal  alcohol  exposure 
in  humans,  such  as  hyperactivity, 
decreased  attentiveness,  aggressive- 
ness, increased  incidence  of  infections, 
augmented  activity  of  the  HPA  axis, 
abnormal  sexual  behavior,  and  prema- 
ture aging.  Many  of  these  pathologies 
might  therefore  be  directly  or  indi- 
rectly caused  by  elevated  CRF  levels. 
However,  while  our  understanding  of 
the  consequences  of  prenatal  alcohol  on 
CRF-dependent  circuitries  is  increas- 
ing (Rivier  1996),  there  is  a  paucity  of 
studies  testing  the  hypothesis  that  this 
peptide  participates  in  fetal  alcohol  syn- 
drome (FAS)-related  disorders.  Finally, 
there  is  the  very  interesting  finding 
that  animals  displaying  increased  vol- 
untary alcohol  consumption  have  ele- 
vated corticosterone  levels  under  basal 
conditions  (Prasad  and  Prasad  1995) — 
although  this  observation  is  not  uni- 
versal (Tuominen  and  Korpi  1991).  If 
true,  this  finding  suggests  the  intrigu- 
ing possibility  that  increased  brain 
CRF  levels  may  be  associated  with 
increased  drinking. 

CRF  also  exerts  direct  effects  in  many 
systems  outside  of  the  brain.  In  view 
of  the  ability  of  alcohol  to  up-regulate 
the  CRF  gene,  it  seems  reasonable  to 
propose  that  it  would  be  of  great 
interest  to  probe  the  hypothesis  that 
cardiovascular  and  immune  effects  (for 
example)  of  the  drug  might  be  modu- 
lated through  this  peptide.  Although 
it  is  outside  the  scope  of  this  chapter 
to  review  this  field,  it  may  be  useful  to 
remember  that  CRF  is  reported  to  be 


manufactured  by  and  to  have  receptors 
in  macrophages  (Webster  et  al.  1990) 
and  other  immune  cells  (Aird  et  al. 
1993;  Kravchenco  and  Furalev  1994), 
and  is  also  reported  to  be  present  in 
arthritic  (Crofford  et  al.  1992)  and 
inflamed  tissues  (Hargreaves  et  al. 
1989),  where  it  is  believed  to  partici- 
pate in  the  inflammatory  process  (Kar- 
alis  et  al.  1991;  Theoharides  et  al. 
1997).  Indeed  the  concept  of  a  "tis- 
sue CRF"  that  is  released  in  response 
to  immune  challenges  and  plays  a  local 
regulatory  role  (Hargreaves  et  al.  1989) 
has  long  been  recognized.  Although 
the  effect  of  alcohol  on  this  CRF  has 
not  been  extensively  studied,  it  has 
been  described  (Dave  and  Eskay  1986). 
CRF  is  also  present  in  steroid-produc- 
ing cells  (Audhya  et  al.  1989;  Ulisse 
et  al.  1989;  Tortorella  et  al.  1993), 
where  it  is  reported  to  play  a  (mostly 
inhibitory)  physiological  role  in  regu- 
lating sex  steroid  production  (Fabbri 
et  al.  1990;  Eskeland  et  al.  1992; 
Dufau  et  al.  1993;  Calogero  et  al. 
1996;  Gnessi  et  al.  1997).  Finally, 
CRF  and/or  its  receptors  are  found  in 
the  gastrointestinal  tract  and  in  the 
heart  (Chalmers  et  al.  1996).  It  is  not 
known  if  alcohol  influences  CRF  and 
CRF  receptors  in  these  tissues,  and 
whether  this  might  play  a  role  in  the 
gastrointestinal  and  cardiovascular 
effects  of  the  drug. 

In  addition  to  exerting  effects  by 
itself,  CRF  alters  homeostasis  by  stim- 
ulating the  release  of  glucocorticoids 
(GC).  These  steroids  influence  immune 
functions:  if  their  levels  are  too  high, 
infection  can  develop  because  the 
activity  of  immune  cells  is  inhibited 
(Black  1994;  Kusnecov  and  Rabin 


64 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


1994;  McEwen  et  al.  1997);  if  their 
levels  are  too  low,  inflammation  can 
take  place  because  of  increased  reac- 
tivity (Sternberg  1992;  Chrousos 
1995;  Sternberg  1997).  Glucocorti- 
coids also  play  a  critical  role  in  the 
general  metabolism  by  regulating  car- 
bohydrate levels  (Dallman  et  al. 
1993)  and  by  influencing  the  tone  of 
blood  vessels  (Munck  and  Guyre 
1986).  Within  the  brain,  GC  maintain 
the  integrity  of  neuronal  networks 
(Meyer  1985)  and  chronic  elevations 
of  its  levels  can  enhance  susceptibility 
to  neurodegeneration  (Joels  and  de 
Kloet  1995)  and  premature  aging 
(Sapolsky  1992;  Seckl  and  Olsson 
1995).  There,  too,  the  potential  for 
an  influence  exerted  by  alcohol 
through  GC  is  enormous,  and  it 
should  be  investigated. 

Hormones  of  the  HPA  Axis  in 
Drug- Seeking  Behavior 

There  is  good  evidence  that  CRF  is 
involved  in  many  aspects  of  drug- 
seeking  behavior.  First,  individual  vul- 
nerability appears  to  correlate  well 
with  various  responses  to  stress, 
including  GC  release  (Deminiere  et 
al.  1989;  Deroche  et  al.  1995).  The 
role  of  GC  is  further  supported  by  the 
findings  that  adrenalectomy  prevents 
the  development  of  alcohol  preference 
in  rats  (Lamblin  and  De  Witte  1996) 
and  that  corticosterone  stimulates 
alcohol  intake  and  the  consumption 
of  other  drugs  (Piazza  et  al.  1993; 
Fahlke  et  al.  1995).  This  may  explain, 
at  least  in  part,  why  stress  reinstates 
drug-seeking  behavior  (Erb  et  al. 
1996).  It  must  be  pointed  out,  how- 
ever, that  the  respective  role  of  CRF 


and  GC  in  drug-seeking  behavior 
remains  unclear  (Deroche  et  al.  1993; 
Shaham  et  al.  1997).  In  addition, 
there  is  a  great  deal  of  controversy  as 
to  whether  brain  levels  of  CRF  are 
elevated  (George  et  al.  1990)  or 
decreased  (Ehlers  et  al.  1992)  in  ani- 
mals with  high  preference  for  alcohol, 
and  whether  alcohol-sensitive  mice 
exhibit  a  blunted  (Tuominen  and 
Korpi  1991)  or  enhanced  (Ehlers  et 
al.  1992)  HPA  axis  response  upon 
exposure  to  stimuli. 

Interactions  Between  the  HPA 
Axis  and  Cytokines:  An  Example  of 
How  Alcohol  Can  Indirectly 
Compromise  Homeostasis 

The  appropriate  release  of  CRF, 
adrenocorticotropic  hormone  (ACTH), 
and  GC  in  response  to  threats  to 
homeostasis  is  essential  for  the  health  of 
the  organism.  This  is  true,  for  exam- 
ple, during  immune  stimulation,  and 
we  know  that  hypo-  or  hypersecretion 
of  CRF  and  GC  leads  to  inflammation 
or  infection,  respectively.  In  dis- 
cussing the  potential  influence  of  alco- 
hol in  altering  the  HPA  axis's  ability 
to  restore  homeostasis,  a  few  intro- 
ductory remarks  may  be  helpful. 

Stress  consists  of  a  stimulus  input,  a 
central  processing  system,  and  a 
response  output  (Levine  and  Ursin 
1991).  Therefore,  the  systems  that  are 
responsible  for  restoring  homeostasis 
in  the  face  of  stress,  such  as  the  HPA 
axis,  need  a  set  of  components  such  as 
sensors  to  monitor  stimuli  or  pertur- 
bations, afferent  transducers  of  infor- 
mation, a  brain  region  that  can  receive 
and  be  responsive  to  the  afferent 
inputs,  and  efferent  transducers  to 


65 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


command  appropriate  responses  to 
the  effector  system.  The  brain  center 
that  controls  the  activity  of  the  HPA 
axis  is  the  paraventricular  nucleus 
(PVN)  of  the  hypothalamus  (Swanson 
1987),  which  contains  CRF  neurons 
(Swanson  1986),  and  the  efferent  sys- 
tem includes  ACTH  and  GC.  Follow- 
ing exposure  to  antigens  in  the 
periphery,  activated  immune  cells  pro- 
duce and  release  cytokines.  These  pro- 
teins convey  to  the  brain  in  general, 
and  the  PVN  in  particular,  the  occur- 
rence of  immune  stimulation  by 
releasing  intermediates  (prostaglandins, 
catecholamines,  etc.),  by  inducing 
production  of  cytokines  in  the  brain 
itself,  by  stimulating  vagal  afferents  to 
the  brain,  or  possibly  by  entering  the 
brain  themselves  (Rivier  1995&;  Bese- 
dovsky  and  Del  Rey  1996).  In 
response,  the  PVN  up-regulates  CRF 
synthesis,  and  ACTH  secretion 
increases.  The  subsequent  release  of 
GC  subserves  many  purposes  in  the 
metabolic  adjustments  that  are  neces- 
sary during  the  acute-phase  response. 
Through  increased  feedback,  these 
steroids  also  ensure  that  PVN  activa- 
tion remains  within  limits  that  are  not 
themselves  threatening  to  homeostasis. 
In  particular,  they  prevent  overpro- 
duction of  cytokines.  The  ability  of 
the  organism  to  restore  homeostasis 
therefore  depends  on  complex  func- 
tional interactions  (a  "checks  and  bal- 
ances" process)  between  cytokines 
and  the  HPA  axis,  and  on  the  appro- 
priate response  of  this  axis.  If  the 
activity  of  the  HPA  axis  has  been  pre- 
viously altered  by  other  stimuli  (e.g., 
alcohol),  PVN  CRF  synthesis  may  be 
enhanced  or  inhibited,  CRF  receptors 


may  be  up-  or  down-regulated,  and  the 
HPA  axis  will  respond  inappropriately. 

Our  laboratory  has  shown  that  in 
adult  rats  exposed  to  alcohol,  subsequent 
exposure  to  exogenous  cytokines  or 
to  a  cytokine-releasing  inflammatory 
process  results  in  a  significandy  blunted 
response  of  the  HPA  axis  (Lee  and 
Rivier  1994^,  1994£,  1995).  This  may 
be  at  least  in  part  due  to  increased 
steroid  feedback,  but  many  other 
mechanisms  probably  play  a  role.  For 
example,  increased  nitric  oxide  (NO) 
production  may  be  important  (Rivier 
1995#).  In  view  of  the  relevance  of 
this  gas  in  brain  function  (see,  e.g., 
Vincent  1994),  and  possible  functional 
relationships  between  alcohol  and  NO 
(Khanna  et  al.  1993;  Fitzgerald  et  al. 
1995;  Lancaster  1995;  Calapai  et  al. 
1996;  Zou  et  al.  1996;  Naassila  et  al. 
1997),  we  need  studies  investigating 
the  effect  of  NO  and  the  other  gaseous 
neurotransmitter,  carbon  monoxide, 
in  regulating  the  response  of  CRF  cir- 
cuitries to  alcohol. 

Another  second  messenger-type 
factor  that  deserves  attention  is 
nuclear  regulatory  factor-KB  (NF-kB) 
(O'Neill  and  Kaltschmidt  1997), 
which  is  emerging  as  a  crucial  regula- 
tor of  brain  function.  The  NF-kB 
family  of  transcription  factors  is  a  pri- 
mary regulatory  component  of  the 
intracellular  signal  pathways  in  cells  of 
the  immune  system,  and  endotoxin 
and  interleukin-1  represent  its  key 
activators.  An  emerging  concept  is 
that  NF-kB  is  an  important  stress  sensor. 
If  this  is  true,  it  may  interact  with 
CRF  in  the  maintenance  of  homeostasis, 
for  example,  by  disrupting  functional 
relationships  between  CRF  and  cytokine - 


66 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


dependent  pathways.  The  observation 
that  physiologically  relevant  concen- 
trations of  alcohol  may  alter  cytokine 
production  by  disrupting  NF-kB  sig- 
naling (see,  e.g.,  Mandrekar  et  al. 
1997)  lends  support  to  this  concept. 

In  contrast  to  the  inhibitory  effect 
of  alcohol  postnatally,  exposure  to  the 
drug  during  embryonic  development 
up-regulates  PVN  CRF  gene  transcrip- 
tion (Lee  et  al.  1990),  which  may  par- 
ticipate in  the  enhanced  HPA  axis 
response  of  the  adult  offspring  (Taylor 
et  al.  1984;  Weinberg  1988;  Lee  and 
Rivier  1996)  to  immune  challenges 
(Lee  and  Rivier  1996)  and  other 
stresses  (Taylor  et  al.  1986;  Weinberg 
1989).  As  a  result,  these  offspring  per- 
manently secrete  too  much  CRF  and 
GC  in  response  to  any  stress,  with  the 
potential  consequences  outlined 
above.  The  mechanisms  through 
which  alcohol  exerts  this  effect  remain 
to  be  fully  elucidated. 

These  experiments  exemplify  the 
type  of  effects  exerted  by  alcohol 
when  it  disrupts  pathways  that  are 
essential  for  health,  and  the  impor- 
tance of  understanding  the  mecha- 
nisms responsible  for  these  effects. 

Stress  Hormones 

Broadly  speaking,  stress  hormones 
include  hormones  of  the  HPA  axis 
(CRF,  ACTH,  GC,  opiates)  and  cate- 
cholamines. The  opioid  peptide  (3- 
endorphin  has  important  functions  in 
the  brain  as  a  neurotransmitter  and  is 
believed  to  play  a  role  in  positive  rein- 
forcement, adaptive  processes,  mood, 
and  the  development  of  alcoholism 
(Gianoulakis  1996;  Boyadjieva  et  al. 
1997).  Both  (3-endorphin  and  another 


opioid  peptide,  pro-opiomelanocortin 
(POMC),  are  regulated  in  part  by  CRF, 
which  may  explain  why  their  levels  are 
elevated  in  both  the  pituitary  and  the 
hypothalamus  following  alcohol  expo- 
sure (Angelogianni  and  Gianoulakis 
1993;  Fickel  et  al.  1994).  On  the 
other  hand,  hypothalamic  POMC  levels 
are  decreased  in  rats  made  dependent 
on  alcohol  (Scanlon  et  al.  1993),  which 
could  be  due  to  decreased  testosterone 
levels.  Although  there  are  differences 
between  the  brain  opioid  levels  of 
rodents  with  different  voluntary  alcohol 
consumption,  these  differences  are 
region  specific  (Gianoulakis  et  al. 
1992),  and  their  importance  in  modulat- 
ing alcohol  consumption  remains  to  be 
fully  understood  (George  et  al.  1991). 

HPG  Axis 

The  fact  that  alcohol  inhibits  reproduc- 
tive functions  in  experimental  animals 
as  well  as  in  humans  abusing  the  drug 
is  well  known  (see,  e.g.,  Bannister  and 
Lowosky  1987;  Purohit  1993).  How- 
ever, understanding  of  the  mecha- 
nisms responsible  for  this  influence 
has  remained  surprisingly  elusive  and 
controversial.  A  careful  evaluation  of 
the  published  data  indicates  significant 
alcohol-induced  decreases  in  luteiniz- 
ing hormone  (LH)  and  sex  steroid 
release.  Alcohol  could  act  at  one  or 
several  levels  of  the  HPG  axis:  the 
gonadotropin-releasing  hormone 
(GnRH)  neurons,  the  afferent  circuits 
to  these  neurons,  pituitary  responsive- 
ness (including  GnRH  receptors, 
GnRH  signaling  pathways,  and  LH 
synthesis),  and  steroidogenesis 
(including  LH  receptors,  postreceptor 
events  such  as  steroidogenic  acute 


67 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


regulatory  [StAR]  protein-mediated 
cholesterol  transport  to  the  inner 
mitochondrial  membrane,  and  the 
activity  of  steroidogenic  enzymes). 

Alcohol  can  inhibit  GnRH  release 
(Ching  et  al.  1988;  Ogilvie  and  Rivier 
1997),  and  the  fact  that  this  effect  is 
usually  only  seen  in  the  intact  animal 
(Uddin  et  al.  1996)  suggests  that  the 
drug  may  primarily  act  on  afferent  cir- 
cuits to  GnRH  neurons,  rather  than 
on  GnRH  production  itself.  Indeed,  a 
decrease  in  catecholamine-induced 
prostaglandins  secretion  has  been  sug- 
gested to  play  a  role  (Hiney  and  Dees 
1991).  Decreases  in  LH  levels  often 
take  place  at  least  1  hour  after  alcohol 
treatment  (Dees  et  al.  1985),  and  they 
probably  reflect  blunted  GnRH  secre- 
tion in  the  hypothalamus  and/or 
release  from  nerve  terminals  (Canteros 
et  al.  1995)  rather  than  changes  in 
pituitary  responsiveness  (Rivier  et  al. 
1992).  Alcohol  may  also  decrease  LH 
mRNA  stability  (Emanuele  et  al. 
1991;  Halloran  et  al.  1995).  Overall, 
it  appears  that  the  influence  of  alcohol 
on  gonadotropin  production  may  be 
modest  following  acute  exposure  to  the 
drug,  but  probably  contributes  to  hypo- 
gonadism during  chronic  treatment. 

The  inhibitory  effect  of  alcohol  on 
the  activity  of  both  male  and  female 
gonads  is  very  strong,  but  it  is  inter- 
esting to  note  that  decreases  in  sex 
steroid  levels  (particularly  testosterone 
[T])  often  precede  measurable 
changes  in  LH  release.  This  suggests 
that  alcohol  can  directly  inhibit 
steroidogenesis,  a  phenomenon  that  is 
now  well  recognized  (Adams  et  al. 
1997).  In  addition,  alcohol  can  act  on 
Sertoli  cells  (Zhu  et  al.  1997).  The 


gonadal  influence  of  alcohol  includes  a 
decrease  in  the  number  of  LH  recep- 
tors (particularly  after  long-term  drug 
treatment  [Salonen  and  Huhtaniemi 
1990]),  decreased  availability  of  the 
metabolites  that  are  necessary  for 
mitochondrial  activity  (Orpana  et  al. 
1990),  and  impaired  synthesis/activ- 
ity of  steroidogenic  enzymes  (Akane 
et  al.  1988).  Alcohol  may  impair 
steroidogenesis  through  increased 
production  of  testicular  opioids 
(Adams  et  al.  1997),  but  the  possibil- 
ity that  the  drug  can  increase  cytokine 
production  in  the  liver,  coupled  with 
the  known  inhibitory  influence  of 
these  proteins  on  gonadal  function 
(Rivier  and  Vale  1989;  Adashi  1990), 
suggests  that  a  similar  mechanism  might 
also  deserve  attention  in  the  gonads. 

One  important  aspect  of  the  effect  of 
alcohol  on  the  gonads  is  its  extreme 
rapidity.  Indeed,  in  our  laboratory,  its 
intraperitoneal  or  intragastric  injection 
significantly  lowered  basal  T  levels  and 
impaired  gonadotropin-induced  T 
response  in  less  than  15  minutes  (Rivier 
1999).  The  mechanisms  responsible 
for  this  rapid  effect  have  not  been 
explored,  but  they  may  be  important 
for  even  the  casual  drinker.  Consistent 
with  results  obtained  in  laboratory 
animals,  chronic  alcohol  has  been 
shown  to  impair  the  activity  of  the 
HPG  axis  in  nonhuman  primates 
(Mello  et  al.  1989)  and  in  humans 
(Bell  et  al.  1995;  Villalta  et  al.  1997). 
It  is  intriguing  to  note  that  although 
acute  alcohol  usually  induces  a  rapid 
and  significant  drop  in  T  levels,  it  not 
only  increases  concentrations  of  the 
steroid  in  women  (Eriksson  et  al. 
1994)  but  also  augments  estradiol 


68 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


production  (Mendelson  et  al.  1989). 
Finally,  alcohol  interferes  with  the 
normal  appearance  of  puberty  (Cicero 
et  al.  1990). 

FUTURE  DIRECTIONS 

General 

Despite  the  pivotal  role  of  CRF  in  so 
many  alcohol-related  disorders  and  as 
a  reinforcer  of  drug  abuse,  the  number 
of  studies  supported  by  NIAAA  that 
investigate  its  synthesis,  release,  and 
effects  on  endocrine  functions  in  gen- 
eral, and  the  HPA  axis  in  particular,  is 
quite  low.  There  may  be  many  rea- 
sons for  this.  Experiments  dealing 
with  endocrine  axes  are  difficult, 
expensive,  and  technically  challenging, 
and  they  require  investigators  who  are 
very  familiar  with  the  biology  of  stress. 
In  particular,  extensive  expertise  with 
in  vivo  work  of  the  quality  required 
for  a  valid  assessment  of  HPA  axis 
activity  is  required,  but  is  a  fast- disap- 
pearing knowledge.  Many  of  the  tools 
necessary  to  carry  out  these  studies  are 
not  widely  available;  these  include 
CRF  antagonists  as  well  as  standardized, 
easy-to-use,  and  affordable  reagents  to 
measure  ACTH  and  corticosterone 
levels.  Proposals  describing  studies 
pertaining  to  the  HPA  axis  should  be, 
but  are  not  always,  reviewed  by  inves- 
tigators who  are  keenly  aware  of  both 
the  potential  and  limitations  of  these 
studies,  which  results  in  a  dwindling 
number  of  laboratories  that  are  funded 
for  this  work.  Along  the  same  lines, 
the  investigation  of  hypotheses  related 
to  the  possible  role  of  CRF  in  FAS 
pathologies  may  require  experimental 


approaches  that  do  not  immediately 
yield  hard  data,  that  require  many 
false  starts,  or  that  may  even  fail. 
Investigators  who  understand  these 
difficulties  are  often  the  same  ones 
who  would  be  best  suited  to  conduct 
these  studies,  but  few  may  spend  time 
and  effort  writing  proposals  under 
these  circumstances. 

Animal  Studies 

Functional  interactions  have  been 
shown  between  CRF  and  the  neuro- 
transmitters involved  in  drug-seeking 
behavior,  reinforcement,  and  relapse. 
There  are  undoubtedly  many  neuro- 
transmitters that  play  a  role  in  the 
development  of  alcohol  abuse.  For 
example,  serotonin  has  been  impli- 
cated in  drug-seeking  behavior,  and 
animals  or  humans  with  low  serotonin 
levels  in  their  brains  are  described  as 
more  aggressive  (Brown  et  al.  1982) 
and  prone  to  depression  (van  Praag 
1996).  Mice  lacking  serotonin  or  its 
receptors  show  less  evidence  of  intoxi- 
cation and  tolerance  (Crabbe  et  al. 
1996),  which  supports  possible  corre- 
lations between  low  serotonin  levels 
and  the  development  of  alcoholism 
(Roy  et  al.  1987;  Schulz  et  al.  1998). 
Although  it  is  known  that  serotonin 
does  not  act  alone  in  the  brain,  its 
potential  functional  connection  with 
CRF  in  the  context  of  alcohol  con- 
sumption has  not  been  given  much 
attention. 

Role  of  CRF  in  Adult  Animals 

A  powerful  tool  to  investigate  the  role 
of  a  secretagogue  is  to  study  biologi- 
cal responses  in  mice  lacking  the  gene 
for  this  secretagogue  or  its  receptors. 


69 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


One  must,  however,  be  aware  of  the 
limitation  of  this  approach.  A  devel- 
oping system  that  is  deprived  of  a  par- 
ticular component  will  often,  if  it  is 
viable,  rely  on  alternate  components 
to  replace  the  missing  entity,  or  on 
redundant  systems  that  are  normally 
not  active  in  the  intact  animal.  Conse- 
quently, one  needs  to  be  very  careful 
when  interpreting  results  obtained 
with  mutant  animals.  For  example, 
NO  is  presently  considered  an  essen- 
tial neurotransmitter,  and  its  acute 
removal  in  the  intact  animal  has  pro- 
found consequences  for  brain  func- 
tion. However,  mice  with  null 
mutation  of  the  gene  for  NO  synthase 
(NOS)  (the  enzyme  that  is  responsi- 
ble for  NO  formation)  display  surpris- 
ingly normal  brain  function,  including 
long-term  potentiation,  an  activity 
thought  to  be  extremely  dependent 
on  NO. 

In  the  future,  the  availability  of 
conditional  mutants  (i.e.,  animals  in 
which  a  gene  can  be  deleted  in  adult- 
hood) will  probably  represent  a  much 
better  method  to  evaluate  the  role  of 
a  particular  secretagogue.  In  the 
meantime,  however,  mutant  ("knock- 
out") rodents  provide  valuable  insight 
into  both  the  role  of  a  secretagogue 
and  the  pathways  that  come  into  play 
when  it  is  absent.  Mice  with  null 
mutation  for  the  CRF  or  CRF-receptor 
gene  are  available,  they  are  capable 
of  sustaining  many  experimental  pro- 
cedures, and  they  are  fertile.  Valuable 
information  would  be  gained  from 
studies  investigating  the  effect 
of  exposing  rodents  with  null  muta- 
tion of  the  CRF  or  CRF-receptor  gene 
to  alcohol. 


Like  most  other  peptides,  CRF  exerts 
its  effect  through  receptors.  Two  CRF 
receptor  families  have  been  identified 
(reviewed  in  DeSouza  1995).  CRF- 
Rl  in  the  pituitary  (Pozzoli  et  al. 
1996;  Sakai  et  al.  1996)  mediate  the 
stimulatory  effect  of  CRF  on  ACTH 
release.  CRF-R1  in  the  PVN  of  the 
hypothalamus  (Sawchenko  et  al. 
1995)  probably  also  play  a  role  for  the 
activity  of  the  HPA  axis,  though  this 
remains  to  be  fully  elucidated.  Certainly, 
these  receptors  could  participate  in 
the  effect  of  CRF  on  non-HPA  axis- 
related  events,  and  as  such  represent 
important  mediators  of  the  influence 
of  this  peptide.  CRF-R1  are  also 
found  in  extrahypothalamic  areas  such 
as  the  amygdala  (Makino  et  al.  1995), 
where  they  may  regulate  emotions. 

CRF-R2,  which  come  in  two 
forms — 2a  and  2(3  (Chalmers  et  al. 
1996) — are  present  in  fewer  brain 
structures  than  CRF-R1  (Lovenberg 
et  al.  1995).  The  structures  in  which 
CRF-R2  are  present  include  the  limbic 
system  (type  2a)  and  the  choroid  plexus 
(type  2|3)  (Lacroix  and  Rivest  1996). 
In  the  hypothalamic  ventromedial 
nucleus  (Chalmers  et  al.  1995),  CRF- 
R2  might  regulate  the  anorexic  effect 
of  CRF  (Choi  et  al.  1996).  Some 
investigators  have  reported  the  pres- 
ence of  the  CRF-R2  gene  in  the  PVN 
(Chalmers  et  al.  1995;  Makino  et  al. 
1997),  but  this  remains  somewhat 
controversial.  CRF-R2  are  also  found  in 
the  periphery,  particularly  in  the  heart 
(Lovenberg  et  al.  1995;  Heldwein  et 
al.  1997).  Overall,  the  role  of  CRF- 
R2  remains  poorly  understood. 

Studies  using  CRF -Rl -deficient 
rodents  will  test  hypotheses  related  to 


70 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


those  tested  with  animals  lacking  the 
gene  for  CRF  itself.  In  addition,  they 
will  provide  information  regarding  the 
type  of  CRF  receptors  that  mediate  a 
particular  effect  of  alcohol,  which  is 
important  for  the  future  development 
of  therapies  aimed  at  alleviating  or 
preventing  unwanted  consequences  of 
alcohol  exposure. 

Role  of  CRF  in  Adult  Offspring  of 
Dams  Exposed  to  Alcohol 

If  we  postulate  the  hypothesis  that  an 
up-regulated  CRF  system  in  the  brain 
is  key  for  many  of  the  adult  endocrine, 
behavioral,  autonomic,  and  immune 
pathologies  observed  in  animals  and 
humans  exposed  to  alcohol  during  fetal 
development,  there  is  a  dire  need  for 
studies  investigating  the  consequences 
of  exposing  pregnant  dams  lacking 
the  CRF  or  CRF-R1  gene  to  alcohol. 
These  studies  should  include  not  only 
alterations  in  HPA  axis  activity  but 
also  changes  in  behavior,  reproductive 
capacity,  immune  functions,  and  drug- 
seeking  behavior. 

Functional  Interactions  Between 
Alcohol  and  Nitric  Oxide,  Carbon 
Monoxide,  and  NF-kB 

Nitric  oxide,  carbon  monoxide,  and 
NF-kB  are  essential  for  normal  brain 
activity.  They  are  likely  to  be  influenced 
by  alcohol  and  probably  participate  in 
pathologies  due  to  this  drug.  Studies 
of  functional  interactions  between 
alcohol  and  these  entities  are  essential. 

In  Vitro  Systems 

The  study  of  the  effect  of  alcohol  on 
CRF  systems  depends  on  the  availabil- 
ity of  reliable  systems  in  which  to 


study  CRF  gene  transcription.  Such 
studies  are  seriously  hampered  by  the 
lack  of  a  good  model  of  isolated  cells 
(either  primary  culture  or  immortal- 
ized cells)  that  produce  CRF.  Fetal 
hypothalami,  which  can  be  main- 
tained in  culture,  produce  little  CRF 
in  comparison  with  other  peptides, 
and  adult  hypothalami  are  very  diffi- 
cult to  maintain  in  culture.  Models  in 
which  to  study  CRF  signaling  path- 
ways and  gene  transcription  are  there- 
fore greatly  needed. 

Reagents  and  Animal  Models 

CRF  Antagonists 

The  investigation  of  the  physiological 
role  of  CRF  can  be  addressed  via  a 
number  of  avenues.  One  is  the  use  of 
potent  CRF  antagonists.  The  devel- 
opment of  these  analogs  has  been 
surprisingly  slow  and  difficult.  If  they 
are  to  be  of  experimental,  and  even- 
tually therapeutic,  benefit,  CRF 
antagonists  (whether  peptidic  or  non- 
peptidic)  will  need  to  be  not  only 
potent  and  long-lasting  but  also 
receptor  specific.  Stability,  the  ability 
to  penetrate  the  brain  following  sys- 
temic administration,  and  cost  of 
manufacture  are  also  factors  in  the 
ultimate  usefulness  of  these  analogs. 
However,  these  factors  cannot  be 
determined  until  we  have  an  array  of 
CRF  antagonists  to  choose  from. 
Interactions  between  various  agencies 
of  the  National  Institutes  of  Health 
(NIH) — for  example,  the  National 
Institute  of  Diabetes  and  Digestive 
and  Kidney  Diseases,  the  National 
Institute  of  Mental  Health,  the 
National  Institute  on  Drug  Abuse, 


71 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  NIAAA — might  be  of  great  benefit 
in  the  development  of  these  antagonists. 

ACTH  and  Corticosterone  Assays 

The  cost  of  measuring  ACTH  levels 
in  unextracted  plasmas  with  assays 
that  can  reliably  handle  hundreds  of 
samples  is  escalating  at  an  alarming 
rate  and  may  soon  threaten  studies 
focused  on  the  HPA  axis.  Similarly, 
the  availability  of  reliable  antibodies 
for  corticosterone  is  dwindling.  NIH- 
distributed  reagents  for  the  measure- 
ment of  plasma  ACTH  and 
corticosterone  levels  in  laboratory 
rodents  are  urgently  needed.  Here 
also,  collaborative  arrangements  with 
other  NIH  agencies  may  be  beneficial. 

Alcohol -Preferring  Animals 

As  mentioned  earlier,  results  obtained 
in  experimental  animals  forcefully 
administered  alcohol  and  results 
obtained  in  those  that  spontaneously 
consume  the  drug  may  be  different.  It 
would  be  extremely  useful  to  have 
access  to  different  strains  of  alcohol- 
preferring  rats  and  mice.  These  strains 
have  been  created,  but  a  mechanism 
making  them  readily  available  to 
appropriate  investigators  would 
greatiy  facilitate  comparative  studies. 

REFERENCES 

Adams,  M.L.;  Meyer,  E.R.;  and  Cicero, 
T.J.  Interactions  between  alcohol-  and 
opioid-induced  suppression  of  rat  testicu- 
lar steroidogenesis  in  vivo.  Alcohol  Clin 
Exp  Res  2 1:684-690,  1997. 

Adashi,  E.Y.  The  potential  relevance  of 
cytokines  to  ovarian  physiology:  The 
emerging  role  of  resident  ovarian  cells  of 


the  white  blood  cell  series.  Endocr  Rev 
11:454-464,  1990. 

Adinoff,  B.;  Martin,  P.R;  Bone,  G.H.A.; 
Eckardt,  M.J.;  Roehrich,  L.;  George,  D.T.; 
Moss,  H.B.;  Eskay,  R;  Linnoila,  M.;  and 
Gold,  P.W.  Hypothalamic-pituitary- 
adrenal  axis  functioning  and  cerebrospinal 
fluid  corticotropin  releasing  hormone  and 
corticotropin  levels  in  alcoholics  after 
recent  and  long-term  abstinence.  Arch 
Gen  Psychiatry  47 :325-330,  1990. 

Aird,  F.;  Clevenger,  C.V.;  Prystowsky, 
M.B.;  and  Redei,  E.  Corticotropin-releas- 
ing  factor  messenger  RNA  in  rat  thymus 
and  spleen.  Proc  Natl  Acad  Sci  USA 
90:7104-7108,  1993. 

Akane,  A.;  Fukushima,  S.;  Shiono,  H.; 
and  Fukui,  Y.  Effects  of  ethanol  on  testic- 
ular steroidogenesis  in  the  rat.  Alcohol 
Alcohol  23 :203-209,  1988. 

Angelogianni,  P.,  and  Gianoulakis,  C. 
Chronic  ethanol  increases  proopiome- 
lanocortin gene  expression  in  the  rat 
hypothalamus.  Neuroendocrinolojjy 
57:106-114,  1993. 

Audhya,  T.;  Hollander,  C.S.;  Schlesinger, 
D.H.;  and  Hutchinson,  B.  Structural 
characterization  and  localization  of  corti- 
cotropin-releasing  factor  in  testis.  Biochim 
Biophys  Acta  995:10-16,  1989. 

Bannister,  P.,  and  Lowosky,  M.S.  Ethanol 
and  hypogonadism.  Alcohol  Alcohol  22:213- 
217,  1987. 

Bell,  H.;  Raknerud,  N.;  Falch,  J.A.;  and 
Haug,  E.  Inappropriately  low  levels  of 
gonadotrophins  in  amenorrhoeic  women 
with  alcoholic  and  non-alcoholic  cirrhosis. 
Eur  J  Endocrinol  132:444^49,  1995. 

Besedovsky,  H.O.,  and  Del  Rey,  A. 
Immune-neuro-endocrine  interactions: 
Facts  and  hypotheses.  Endocr  Rev 
17:64-102,  1996. 


72 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


Black,  P.H.  Central  nervous  system- 
immune  system  interactions:  Psychoneuro- 
endocrinology  of  stress  and  its  immune 
consequences.  Antimicrob  Agents 
Chemother  38:1-6,  1994. 

Boyadjieva,  N.;  Reddy,  B.;  and  Sarkar,  D. 
Forskolin  delays  the  ethanol-induced 
desensitization  of  hypothalamic  (3 -endor- 
phin neurons  in  primary  culture.  Alcohol 
Clin  Exp  Res  21 :477-482,  1997. 

Brown,  G.L.;  Ebert,  M.H.;  Goyer,  P.F.; 
Jimerson,  D.C.;  Klein,  W.J.;  Bunney, 
W.E.;  and  Goodwin,  F.K.  Aggression, 
suicide,  and  serotonin:  Relationships  to 
CSF  amine  metabolites.  Am  J  Psychiatry 
139:741-746,  1982. 

Brown,  M.R.,  and  Fisher,  L.A.  Cortico- 
tropin releasing  factor:  Effects  on  the 
autonomic  nervous  system  and  visceral 
systems.  Federation  Proc 44:243-248, 1985. 

Calapai,  G.;  Mazzaglia,  G.;  Sautebin,  L.; 
Costantino,  G.;  Marciano,  M.C.; 
Cuzzocrea,  S.;  Di  Rosa,  M.;  and  Caputi, 
A.P.  Inhibition  of  nitric  oxide  formation 
reduces  voluntary  ethanol  consumption  in 
the  rat.  Psychopharmacology  (Berl) 
125:398-401, 1996. 

Calogero,  A.E.;  Burrello,  N.;  Negri-Cesi, 
P.;  Papale,  L.;  Palumbo,  M.A.;  Cianci,  A.; 
Sanfilippo,  S.;  and  D'Agata,  R.  Effects  of 
corticotropin-releasing  hormone  on  ovar- 
ian estrogen  production  in  vitro. 
Endocrinology  137:4161-4166,  1996. 

Canteros,  G.;  Rettori,  V.;  Franchi,  A.; 
Genaro,  A.;  Cebral,  E.;  Faletti,  A.; 
Gimeno,  M.;  and  McCann,  S.M.  Ethanol 
inhibits  luteinizing  hormone-releasing 
hormone  (LHRH)  secretion  by  blocking 
the  response  of  LHRH  neuronal  termi- 
nals to  nitric  oxide.  Proc  Natl  Acad  Sci 
USA  92:3416-3420, 1995. 


Carmona-Calero,  E.;  del  Mar  Perez- 
Delgado,  M.;  Banuelos-Pineda,  J.; 
Marrero-Gordillo,  N.;  Ferres-Torres,  R; 
and  Castaneyra-Perdomo,  A.  Effects  of 
chronic  alcohol  intake  on  the  vasopressin 
content  in  the  hypothalamic  paraventricu- 
lar and  supraoptic  nuclei  of  the  mouse. 
An  immunohistochemical  and  morpho- 
metric  study.  Drug  Alcohol  Depend 
38:19-24,  1995. 

Chalmers,  D.T.;  Lovenberg,  T.W.;  and 
De  Souza,  E.B.  Localization  of  novel 
corticotropin-releasing  factor  receptor 
(CRF,)  mRNA  expression  to  specific 
subcortical  nuclei  in  rat  brain:  Comparison 
with  CRF,  receptor  mRNA  expression.  / 
Neurosci  15:6340-6350,  1995. 

Chalmers,  D.T.;  Lovenberg,  T.W.; 
Grigoriadis,  D.E.;  Behan,  D.P.;  and 
DeSouza,  E.B.  Corticotrophin-releasing 
factor  receptors:  From  molecular  biology 
to  drug  design.  Trends  Pharmacol  Sci 
17:166-172,  1996. 

Ching,  M.;  Valenca,  M.;  and  Negro-Vilar, 
A.  Acute  ethanol  treatment  lowers  hypo- 
physeal portal  plasma  luteinizing  hormone- 
releasing  hormone  (LH-RH)  and  systemic 
plasma  LH  levels  in  orchidectomized  rats. 
Brain  Res 443:325-328,  1988. 

Choi,  S.;  Horsley,  C;  Aguila,  S.;  and 
Dallman,  M.F.  The  hypothalamic  ventro- 
medial nuclei  couple  activity  in  the  hypo- 
thalamo-pituitary- adrenal  axis  to  the 
morning  fed  or  fasted  state.  /  Neurosci 
16:8170-8180,  1996. 

Chrousos,  G.P.  The  hypothalamic-pituitary- 
adrenal  axis  and  immune-mediated  in- 
flammation. N  Engl  J  Med  332:1351- 
1362, 1995. 

Cicero,  T.J.;  Adams,  M.L.;  O'Connor,  L.; 
Nock,  B.;  Meyer,  E.R;  and  Wozniak,  D. 
Influence  of  chronic  alcohol  administra- 
tion on  representative  indices  of  puberty 


73 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  sexual  maturation  in  male  rats  and 
the  development  of  their  progeny.  / 
Pharmacol  Exp  Ther  255:707-715,  1990. 

Crabbe,  J.;  Phillips,  T.;  Feller,  D.;  Hen, 
R.;  Wenger,  C;  Lessov,  C;  and  Schafer, 
G.  Elevated  alcohol  consumption  in  null 
mutant  mice  lacking  5-HT1B  serotonin. 
Nat  Genet  14:90-101,  1996. 

Crofford,  L.J.;  Sano,  H.;  Karalis,  K.; 
Webster,  E.L.;  Goldmuntz,  E.A.; 
Chrousos,  G.P.;  and  Wilder,  R.L.  Local 
secretion  of  corticotropin-releasing  hor- 
mone in  the  joints  of  Lewis  rats  with 
inflammatory  arthritis.  /  Clin  Invest 
90:2555-2564,  1992. 

Dallman,  M.F.;  Strack,  A.M.;  Akana,  S.F.; 
Bradbury,  M.J.;  Hanson,  E.S.;  Scribner, 
K.A.;  and  Smith,  M.  Feast  and  famine: 
Critical  role  of  glucocorticoids  with  in- 
sulin in  daily  energy  flow.  Front 
Neuroendocrinal  14:303-347,  1993. 

Dave,  J.R,  and  Eskay,  RL.  Demonstration 
of  corticotropin-releasing  factor  binding 
sites  on  human  and  rat  erythrocyte  mem- 
branes and  their  modulation  by  chronic 
ethanol  treatment  in  rats.  Biochem  Biopbys 
ResCommun  136:137-144,  1986. 

Dees,  W.L.;  Rettori,  V.;  Kozlowski,  G.P.; 
and  McCann,  S.M.  Ethanol  and  the  pul- 
satile release  of  luteinizing  hormone, 
follicle  stimulating  hormone  and  prolactin 
in  ovariectomized  rats.  Alcohol 2:641  - 
646,  1985. 

Deminiere,  J.M.;  Piazza,  P.V.;  Le  Moal, 
M.;  and  Simon,  H.  Experimental 
approach  to  individual  vulnerability  to 
psychostimulant  addiction.  Neurosci 
BiobehavRev  13:141-147,  1989. 

Deroche,  V.;  Piazza,  P.V.;  Deminiere,  J.- 
M.;  Le  Moal,  M.;  and  Simon,  H.  Rats 
orally  self- administer  corticosterone.  Brain 
Res  622:315-320,  1993. 


Deroche,  V.;  Marinelli,  M.;  Maccari,  S.; 
Le  Moal,  M.;  Simon,  H.;  and  Piazza,  P.V. 
Stress-induced  sensitization  and  glucocor- 
ticoids. I.  Sensitization  of  dopamine- 
dependent  locomotor  effects  of 
amphetamine  and  morphine  depends  on 
stress-induced  corticosterone  secretion.  / 
Neurosci  15:7181-7188, 1995. 

DeSouza,  E.B.  Corticotropin-releasing 
factor  receptors:  Physiology,  pharmacology, 
biochemistry  and  role  in  central  nervous 
system  and  immune  disorders.  Psycho- 
neuroendocrinology  20:789-819,  1995. 

Dufau,  M.L.;  Tinajero,  J.C.;  and  Fabbri, 
A.  Corticotropin-releasing  factor:  An 
antireproductive  hormone  of  the  testis. 
FASEB  J 7:299-307,  1993. 

Ehlers,  C.L.;  Chaplin,  RL;  Wall,  T.L.; 
Lumeng,  L.;  Li,  T.K.;  Owens,  M.J.;  and 
Nemeroff,  C.B.  Corticotropin  releasing 
factor  (CRF):  Studies  in  alcohol  prefer- 
ring and  non-preferring  rats.  Psycho- 
pharmacokgy  106:359-364,  1992. 

Ehrenreich,  H.;  Schuck,  J.;  Stender,  N.; 
Pilz,  J.;  Gefeller,  O.;  Schilling,  L.;  Poser, 
W.;  and  Kaw,  S.  Endocrine  and  hemody- 
namic effects  of  stress  versus  systemic 
CRF  in  alcoholics  during  early  and 
medium  term  abstinence.  Alcohol  Clin 
Exp  Res  21:1285-1293,  1997. 

Emanuele,  M.A.;  Tentler,  J.;  Emanuele, 
N.V.;  and  Kelley,  M.R.  In  vivo  effects  of 
acute  EtOH  on  rat  alpha  and  beta 
luteinizing  hormone  gene  expression. 
Afcofco/ 8:345-348,  1991. 

Erb,  S.;  Shaham,  Y.;  and  Stewart,  J.  Stress 
reinstates  cocaine-seeking  behavior  after 
prolonged  extinction  and  a  drug-free 
period.  Psychopharmacology  (Berl) 
128:408^12,  1996. 


74 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


Eriksson,  C;  Fukunaga,  T.;  and 
Lindman,  R.  Sex  hormone  response  to 
alcohol.  Nature  369:711,  1994. 

Eskeland,  N.L.;  Molineaux,  C.J.;  and 
Schachter,  B.S.  Regulation  of  |3-endor- 
phin  secretion  by  corticotropin-releasing 
factor  in  the  intact  rat  testis. 
Endocrinology  130:1173-1179,  1992. 

Fabbri,  A.;  Tinajero,  J.C.;  and  DuFau, 
M.L.  Corticotropin-releasing  factor  is 
produced  by  rat  Leydig  cells  and  has  a 
major  local  antireproductive  role  in  the 
testis.  Endocrinology  127:1541-1543, 
1990. 

Fahlke,  C;  Hard,  E.;  Eriksson,  C.J. P.; 
Engel,  J.A.;  and  Hansen,  S.  Consequence 
of  long-term  exposure  to  corticosterone 
or  dexamethasone  on  ethanol  consump- 
tion in  the  adrenalectomized  rat,  and  the 
effect  of  type  I  and  type  II  corticosteroid 
receptor  antagonists.  Psychopharmacology 
(Berl)  117:216-224, 1995. 

Fickel,  W.;  Roske,  J.;  Frenzel,  R;  and 
Melzig,  M.  Effects  of  chronic  ethanol 
treatment  on  the  POMC  mRNA  synthesis 
in  the  rat  pituitary.  Regul  Pept  Suppl  1 : 
S231-S232,  1994. 

Fitzgerald,  L.W.;  Charlton,  M.E.; 
Duman,  R.S.;  and  Nestler,  E.J. 
Regulation  of  neuronal  nitric  oxide  syn- 
thase by  chronic  ethanol  ingestion. 
Synapse  21:93-95,  1995. 

George,  S.;  Fan,  T.;  Roldan,  L.;  and 
Naranjo,  C.  Corticotropin-releasing  factor 
is  altered  in  brains  of  animals  with  high 
preference  for  ethanol.  Alcohol  Clin  Exp 
Res  14:425-429,  1990. 

George,  S.R.;  Roldan,  L.;  Lui,  A.;  and 
Naranjo,  C.A.  Endogenous  opioids  are 
involved  in  the  genetically  determined 
high  preference  for  ethanol  consumption. 
Alcohol  Clin  Exp  Res  15:668-672,  1991. 


Gianoulakis,  C.  Implications  of  endoge- 
nous opioids  and  dopamine  in  alcoholism: 
Human  and  basic  science  studies.  Alcohol 
Alcohol  31:33-^2,  1996. 

Gianoulakis,  C;  de  Waele,  J.;  and 
Kiianmaa,  K.  Differences  in  the  brain  and 
pituitary  b-endorphin  system  between  the 
alcohol-preferring  AA  and  alcohol-avoid- 
ing ANA  rats.  Alcohol  Clin  Exp  Res 
16:453-459,  1992. 

Gnessi,  L.;  Fabbri,  A.;  and  Spera,  G. 
Gonadal  peptides  as  mediators  of  devel- 
opment and  functional  control  of  the 
testis:  An  integrated  system  with  hor- 
mones and  local  environment.  Endocr  Rev 
18:541-609,  1997. 

Halloran,  M.M.;  Emanuele,  M.A.; 
Emanuele,  N.V.;  Tender,  J.J.;  and  Kelley, 
M.R.  Further  characterization  of  the 
impact  of  ethanol  on  (3LH:  Alterations  in 
polyribosome  association  of  |3LH  mRNA. 
Endocrine  3:469-473,  1995. 

Hargreaves,  K.M.;  Costello,  A.H.;  and 
Joris,  J.L.  Release  from  inflamed  tissue  of 
a  substance  with  properties  similar  to 
corticotropin-releasing  factor.  Neuro- 
endocrinology  49:476-482,  1989. 

Heldwein,  K.A.;  Duncan,  J.E.;  Stenzel, 
P.;  Rittenberg,  M.B.;  and  Stenzel-Poore, 
M.P.  Endotoxin  regulates  corticotropin- 
releasing  hormone  receptor  2  in  heart  and 
skeletal  muscle.  Mol  Cell  Endocrinol 
131:167-172,  1997. 

Heuser,  I.;  von  Bardeleben,  U.;  Boll,  E.; 
and  Holsboer,  F.  Response  of  ACTH  and 
Cortisol  to  human  corticotropin-releasing 
hormone  after  short-term  abstention  from 
alcohol  abuse.  Biol  Psychiatry  24:316- 
321,  1988. 

Hiney,  J.K.,  and  Dees,  W.L.  Ethanol  inhibits 
luteinizing  hormone-releasing  hormone 
release  from  the  median  eminence  of 


75 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


prepubertal  female  rats  in  vitro: 
Investigation  of  its  actions  on  norepi- 
nephrine and  prostaglandin-E2. 
Endocrinology  128:1404-1408,  1991. 

Inder,  W.J.;  Joyce,  P.R;  Ellis,  M.J.;  Evans, 
M.J.;  Livesey,  J.H.;  and  Donald,  R.A. 
The  effects  of  alcoholism  on  the  hypothal- 
amic-pituitary-adrenal  axis:  Interaction 
with  endogenous  opioid  peptides.  Clin 
Endocrinol  43:283-290,  1995a. 

Inder,  W.J.;  Joyce,  P.R;  Wells,  J.E.; 
Evans,  M.J.;  Ellis,  M.J.;  Mattioli,  L.;  and 
Donald,  RA.  The  acute  effects  of  oral 
ethanol  on  the  hypothalamic -pituitary- 
adrenal  axis  in  normal  human  subjects. 
Clin  Endocrinol  42:65-71,  19956. 

Irwin,  M.;  Vale,  W.;  and  Rivier,  C.  Central 
corticotropin-releasing  factor  mediates  the 
suppressive  effect  of  stress  on  natural  killer 
cytotoxicity.  Endocrinology  126:2837- 
2844,  1990. 

Joels,  M.;  and  de  Kloet,  E.  Corticosteroid 
hormones:  Endocrine  messengers  in  the 
brain.  News  in  Psychological  Sciences  10:71- 
76,  1995. 

Karalis,  K.;  Sano,  H.;  Redwine,  J.; 
Listwak,  S.;  Wilder,  R.L.;  and  Chrousos, 
G.P.  Autocrine  or  paracrine  inflammatory 
actions  of  corticotropin-releasing  hormone 
in  vivo.  Science  254:421^23,  1991. 

Khanna,  J.M.;  Morato,  G.S.;  Shah,  G.; 
Chau,  A.;  and  Kalant,  H.  Inhibition  of 
nitric  oxide  synthesis  impairs  rapid  tolerance 
to  ethanol.  Brain  Res  Bull  32:43^:7,  1993. 

Koob,  G.F.;  Heinrichs,  S.C.;  Pich,  E.M.; 
Menzaghi,  F.;  Baldwin,  H.;  Miczek,  K.; 
and  Britton,  K.T.  The  role  of 
corticotropin-releasing  factor  in  behav- 
ioural responses  to  stress.  In:  Chadwick, 
D.J.;.  Marsh,  J.;  and  Ackrill,  K.,  eds.  Ciba 
Foundation  Symposium  No.  172  on 
Corticotropin-Releasing  Factor.  New 


York:  John  Wiley  &  Sons,  1993.  pp. 

277-295. 

Kravchenco,  I.V.,  and  Furalev,  VA. 
Secretion  of  immunoreactive 
corticotropin  releasing  factor  and  adreno- 
corticotropic hormone  by  T-  and  B- 
lymphocytes  in  response  to  cellular  stress 
factors.  Biochem  Biophys  Res  Commun 
204:828-834,  1994. 

Kusnecov,  A.W.,  and  Rabin,  B.S.  Stressor- 
induced  alterations  of  immune  function: 
Mechanisms  and  issues.  Int  Arch  Allergy 
Immunol  105:107-121,  1994. 

Lacroix,  S.,  and  Rivest,  S.  Role  of  cyclo- 
oxygenase  pathways  in  the  stimulatory 
influence  of  immune  challenge  on  the 
transcription  of  a  specific  CRF  receptor 
subtype  in  the  rat  brain.  /  Chem  Neuroanat 
10:53-71,  1996. 

Lamblin,  F.,  and  De  Witte,  P.  Adrenal- 
ectomy prevents  the  development  of 
alcohol  preference  in  male  rats.  Alcohol 
13:233-238,  1996. 

Lancaster,  F.E.  Alcohol  and  the  brain: 
What's  NO  got  to  do  with  it?  Metab 
Brain  Dis  10:125-133,  1995. 

Lee,  S.,  and  Rivier,  C.  Effect  of  postnatal 
exposure  of  female  rats  to  an  alcohol  diet: 
Influence  of  age  and  circulating  sex 
steroids.  Alcohol  Clin  Exp  Res  18:998- 
1003,  1994a. 

Lee,  S.,  and  Rivier,  C.  Interaction  between 
alcohol  and  interleukin-ip  on  ACTH 
secretion  and  the  expression  of  immediate 
early  genes  in  the  hypothalamus.  Mol  Cell 
NeurosciS  :442^50,  1994b. 

Lee,  S.,  and  Rivier,  C.  Altered  ACTH  and 
corticosterone  responses  to  interleukin-ip 
in  male  rats  exposed  to  an  alcohol  diet:  Pos- 
sible role  of  vasopressin  and  testosterone. 
Alcohol  Clin  Exp  Res  19:200-208,  1995. 


76 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


Lee,  S.,  and  Rivier,  C.  Gender  differences 
in  the  effect  of  prenatal  alcohol  exposure 
on  the  hypothalamic-pituitary-adrenal  axis 
response  to  immune  signals.  Psychoneuro- 
endocrinology  21:145-156,  1996. 

Lee,  S.Y.;  Imaki,  T.;  Vale,  W.;  and  Rivier, 
C.L.  Effect  of  prenatal  exposure  to  ethanol 
on  the  activity  of  the  hypothalamic-pituitary- 
adrenal  axis  of  the  offspring:  Importance 
of  the  time  of  exposure  to  ethanol  and 
possible  modulating  mechanisms.  Mol 
CellNeurosci  1:168-177,  1990. 

Levine,  S.,  and  Ursin,  H.  What  is  stress? 
In:  Brown,  M.R.;  Koob,  G.;  and  Rivier, 
C,  eds.  Stress:  Neurobiology  and  Neuro- 
endocrinology.  New  York:  Marcel  Dekker, 
1991.  pp.  3-21. 

Lovenberg,  T.W.;  Chalmers,  D.T.;  Liu, 
C.;  and  DeSouza,  E.B.  CRF,a  and  CRF2(3 
receptor  mRNAs  are  differentially  distrib- 
uted between  the  rat  central  nervous  system 
and  peripheral  tissues.  Endocrinology  136: 
4139^142,  1995. 

Makino,  S.;  Schulkin,  J.;  Smith,  M.A.; 
Pacak,  K.;  Palkovits,  M.;  and  Gold,  P.W. 
Regulation  of  corticotropin-releasing 
hormone  receptor  messenger  ribonucleic 
acid  in  the  rat  brain  and  pituitary  by  glu- 
cocorticoids and  stress.  Endocrinology 
136:4517-4525,  1995. 

Makino,  S.;  Takemura,  T.;  Asaba,  K.; 
Nishiyama,  M.;  Takao,  T.;  and  Hashimoto, 
K.  Differential  regulation  of  type- 1  and 
type-2  a  corticotropin-releasing  hormone 
receptor  mRNA  in  the  hypothalamic 
paraventricular  nucleus  of  the  rat.  Brain 
Res  Mol  Brain  Res  47:170-176,  1997. 

Mandrekar,  P.;  Catalano,  D.;  and  Szabo, 
G.  Alcohol-induced  regulation  of  nuclear 
regulatory  factor-K(3  in  human  monocytes. 
Alcohol  Clin  Exp  Res  21:988-994,  1997. 


McEwen,  B.;  Biron,  C;  Brunson,  K.; 
Bulloch,  K.;  Chambers,  W.;  Dhabhar,  P.; 
Goldfarb,  R.;  Kitson,  R.;  Miller,  A.; 
Spencer,  R.;  and  Weiss,  J.  The  role  of 
adrenocorticoids  as  modulators  of  im- 
mune function  in  health  and  disease: 
Neural,  endocrine  and  immune  interac- 
tions. Brain  Res  Rev  23:79-133,  1997. 

Mello,  N.K.;  Mendelson,  J.H.;  Bree, 
M.P.;  and  Skupny,  A.  Alcohol  effects  on 
LH  and  FSH  in  ovariectomized  female 
monkeys.  Alcohol  6:147-159,  1989. 

Mendelson,  J.H.;  Mello,  N.K.;  Teoh, 
S.K.;  and  Ellingboe,  J.  Alcohol  effects  on 
luteinizing  hormone  releasing  hormone- 
stimulated  anterior  pituitary  and  gonadal 
hormones  in  women.  J  Pharmacol  Exp 
Tfor  250:902-909,  1989. 

Meyer,  J.S.  Biochemical  effects  of  corti- 
costeroids on  neural  tissues.  Physiol  Rev 
65:946-1020,  1985. 

Moolten,  M.,  and  Kornetsky,  C.  Oral  self- 
administration  of  ethanol  and  not  experi- 
menter-administered ethanol  facilitates 
rewarding  electrical  brain  stimulation. 
Alcohol  7:221-225,  1990. 

Munck,  A.,  and  Guyre,  P.M.  Glucocorticoid 
physiology,  pharmacology  and  stress.  Adv 
Exp  Med  Biol  196:81-96,  1986. 

Naassila,  M.;  Beauge,  F.;  and  Daoust,  M. 
Regulation  of  rat  neuronal  nitric  oxide 
synthase  activity  by  chronic  alcoholiza- 
tion. Alcohol  Alcohol  32:13-17,  1997. 

Nemeroff,  C.B.  The  corticotropin-releasing 
factor  (CRF)  hypothesis  of  depression: 
New  findings  and  new  directions.  Mol 
Psychiatry  1:336-342,  1996. 

Ogilvie,  K.,  and  Rivier,  C.  Effect  of  alcohol 
on  the  proestrous  surge  of  luteinizing 
hormone  (LH)  and  the  activation  of  LH- 
releasing  hormone  (LHRH)  neurons  in  the 
female  rat.  /  Neurosci  17:2595-2604, 1997. 


77 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


O'Neill,  L.A.J. ,  and  Kaltschmidt,  C.  NF- 
kB:  A  crucial  transcription  factor  for  glial 
and  neuronal  cell  function.  Trends 
Neurosci  20:252-258,  1997. 

Orpana,  A.K.;  Harkonen,  M.;  and  Eriksson, 
C.J. P.  Ethanol-induced  inhibition  of 
testosterone  biosynthesis  in  rat  Leydig 
cells:  Role  of  mitochondrial  substrate 
shuttles  and  citrate.  Alcohol  Alcohol 
25:499-507,  1990. 

Piazza,  P.V.;  Deroche,  V.;  Deminiere,  ].- 
M.;  Maccari,  S.;  Le  Moal,  M.;  and  Simon, 
H.  Corticosterone  in  the  range  of  stress- 
induced  levels  possesses  reinforcing  prop- 
erties: Implications  for  sensation-seeking 
behaviors.  Proc  Natl  Acad  Sci  USA 
90:11738-11742,1993. 

Pozzoli,  G.;  Bilezikjian,  L.M.;  Perrin, 
M.H.;  Blount,  A.L.;  and  Vale,  W.W. 
Corticotropin-releasing  factor  (CRF)  and 
glucocorticoids  modulate  the  expression 
of  type  1  CRF  receptor  messenger  ri- 
bonucleic acid  in  rat  anterior  pituitary  cell 
cultures.  Endocrinology  137:65-71,  1996. 

Prasad,  C,  and  Prasad,  A.  A  relationship 
between  increased  voluntary  alcohol 
preference  and  basal  hypercorticosterone- 
mia  associated  with  an  attenuated  rise  in 
corticosterone  output  during  stress. 
Alcohol  12:59-63, 1995. 

Purohit,  V.  Effects  of  alcohol  on  the 
hypothalamic-pituitary-gonadal  axis.  In: 
Zakhari,  S.  ed.  Alcohol  and  the  Endocrine 
System.  National  Institute  on  Alcohol 
Abuse  and  Alcoholism  Research 
Monograph  No.  23.  NIH  Pub.  No. 
93-3533.  Bethesda,  MD:  the  Institute, 
1993.  pp.  189-203. 

Rivest,  S.,  and  Rivier,  C.  The  role  of 
corticotropin-releasing  factor  and  inter- 
leukin-1  in  the  regulation  of  neurons 
controlling  reproductive  functions. 
EndocrRev  16:177-199,  1995. 


Rivier,  C.  Adult  male  rats  exposed  to  an 
alcohol  diet  exhibit  a  blunted  ACTH 
response  to  immune  or  physical  stress: 
Possible  role  of  nitric  oxide.  Alcohol  Clin 
Exp  Res  19:1474-1479,  1995a. 

Rivier,  C.  Influence  of  immune  signals  on 
the  hypothalamic-pituitary  axis  of  the 
rodent.  Front  Neuroendocrinal  16:151- 
182,  19956. 

Rivier,  C.  Alcohol  stimulates  ACTH 
secretion  in  the  rat:  Mechanisms  of  action 
and  interactions  with  other  stimuli. 
Alcohol  Clin  Exp  Res  20:240-254,  1996. 

Rivier,  C.  Alcohol  rapidly  lowers  plasma 
testosterone  levels  in  the  rat:  Evidence 
that  a  neural  brain-gonadal  pathway  may 
be  important  for  decreased  testicular 
responsiveness  to  gonadotropin.  Alcohol 
Clin  Exp  Res  23:38-45,  1999. 

Rivier,  C,  and  Lee,  S.  Acute  alcohol 
administration  stimulates  the  activity  of 
hypothalamic  neurons  that  express  corti- 
cotropin-releasing factor  and  vasopressin. 
Brain  Res  726:1-10,  1996. 

Rivier,  C.L.,  and  Plotsky,  P.M.  Mediation 
by  corticotropin-releasing  factor  (CRF)  of 
adenohypophysial  hormone  secretion. 
Annu  Rev  Physiol 48:475^94,  1986. 

Rivier,  C,  and  Vale,  W.  Corticotropin- 
releasing  factor  (CRF)  acts  centrally  to 
inhibit  growth  hormone  secretion  in  the 
rat.  Endocrinology  114:2409-2411,  1984. 

Rivier,  C,  and  Vale,  W.  In  the  rat,  inter- 
leukin-la  acts  at  the  level  of  the  brain  and 
the  gonads  to  interfere  with  gonadotropin 
and  sex  steroid  secretion.  Endocrinology 
124:2105-2109,  1989. 

Rivier,  C;  Bruhn,  T.;  and  Vale,  W.  Effect 
of  ethanol  on  the  hypothalamic-pituitary- 
adrenal  axis  in  the  rat:  Role  of 
corticotropin-releasing  factor  (CRF).  / 
Pharmacol  Exp  Ther  229:127-131,  1984. 


78 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


Rivier,  C;  Rivest,  S.;  and  Vale,  W. 
Alcohol-induced  inhibition  of  LH  secre- 
tion in  intact  and  gonadectomized  male 
and  female  rats:  Possible  mechanisms. 
Alcohol  Clin  Exp  Res  16:928-934,  1992. 

Roy,  A.;  Virkkunen,  M.;  and  Linnoila,  M. 
Reduced  central  serotonin  turnover  in  a 
subgroup  of  alcoholics?  Frog  Neuropsycho- 
pharmacol  Biol  Psychiatry  1 1 :  173-177, 1987. 

Sakai,  K\;  Horiba,  N.;  Sakai,  Y.;  Tozawa,  F.; 
Demura,  H.;  and  Suda,  T.  Regulation  of 
corticotropin-releasing  factor  receptor  mes- 
senger ribonucleic  acid  in  rat  anterior  pitu- 
itary. Endocrinology  137:1758-1763, 1996. 

Salonen,  I.,  and  Huhtaniemi,  I.  Effects  of 
chronic  ethanol  diet  on  pituitary- testicular 
function  of  the  rat.  Biol  Reprod  42:55-62, 
1990. 

Sapolsky,  R.  Stress,  the  Aging  Brain  and 
the  Mechanisms  of  Neuron  Death. 
Cambridge,  MA:  MIT  Press,  1992. 

Sawchenko,  P.E.;  Arias,  C;  Van  Pett,  K.; 
Viau,  V.;  Chen,  R.;  Donaldson,  C; 
Blount,  A.;  Bilezikjian,  L.;  Perrin,  M.; 
and  Vale,  W.  Distribution  of  mRNAs 
encoding  two  distinct  CRF  receptors  in 
brain  and  pituitary  of  rat  and  mouse.  The 
Endocrine  Society  77th  Annual  Meeting: 
Program  and  Abstracts.  Bethesda,  MD: 
Endocrine  Society  Press,  1995.  p.  252, 
Abstract  Pl-558. 

Scanlon,  M.N.;  Lazar-Wesley,  E.;  Grant, 
K.A.;  and  Kunos,  G.  Proopiomelanocortin 
messenger  RNA  is  decreased  in  the 
mediobasal  hypothalamus  of  rats  made 
dependent  on  ethanol.  Alcohol  Clin  Exp 
Res  16:1147-1151,  1993. 

Schulz,  K.P.;  McKay,  K.E.;  Newcorn,  J.H.; 
Sharma,  V.;  Gabriel,  S.;  and  Halperin, 
J.M.  Serotonin  function  and  risk  for 
alcoholism  in  boys  with  attention-deficit 


hyperactivity  disorder.  Neuropsycho- 
pharmacology  18:10-17,  1998. 

Seckl,  J.R.,  and  Olsson,  T.  Glucocorticoid 
hypersecretion  and  the  age-impaired 
hippocampus:  Cause  or  effect?  / 
Endocrinol  145:201-211,  1995. 

Shaham,  Y.;  Funk,  D.;  Erb,  S.;  Brown, 
T.J.;  Walker,  C.-D.;  and  Stewart,  J. 
Corticotropin-releasing  factor,  but  not 
corticosterone,  is  involved  in  stress-in- 
duced relapse  to  heroin-seeking  in  rats.  / 
Neurosci  17:2605-2614, 1997. 

Sternberg,  E.M.  The  stress  response  and 
the  regulation  of  inflammatory  disease. 
Ann  Intern  Med  117:854-866,  1992. 

Sternberg,  E.M.  Neural-immune  interac- 
tions in  health  and  disease.  J  Clin  Invest 
100:2641-2647,  1997. 

Swanson,  L.W.  Organization  of  mam- 
malian neuroendocrine  system.  In: 
Mountcastle,  V.B.;  Bloom,  F.E.;  and 
Geiger,  S.R.,  eds.  Handbook  of  Physiology, 
a  Critical,  Comprehensive  Presentation  of 
Physiological  Knowledge  and  Concepts. 
Section  I:  The  Nervous  System, 
Neurophysiology.  Vol.  IV,  Chapter  6. 
Bethesda,  MD:  American  Physiological 
Society,  1986.  [Distributed  by  Williams 
&Wilkins.]pp.  317-363. 

Swanson,  L.W.  The  hypothalamus.  In: 
Bjorklund,  A.;  Hokfelt,  P.;  and  Swanson, 
L.W.,  eds.  Handbook  of  Chemical 
Neuroanatomy.  Vol.  5.  Amsterdam: 
Elsevier,  1987.  pp.  1-124. 

Tache,  Y.;  Monnikes,  H.;  Bonaz,  B.; 
and  Rivier,  J.  Role  of  CRF  in  stress- 
related  alterations  of  gastric  and  colonic 
motor  function.  In:  Tache,  Y.,  and 
Rivier,  C,  eds.  Corticotropin-Releasing 
Factor  and  Cytokines:  Role  in  the  Stress 
Response.  Vol.  697.  New  York:  The  New 


79 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


York  Academy  of  Sciences,  1993.  pp. 
233-243. 

Taylor,  A.N.;  Nelson,  L.R.;  Branch,  B.J.; 
Kokka,  N.;  and  Poland,  R.E.  Altered 
stress  responsiveness  in  adult  rats  exposed 
to  ethanol  in  utero:  Neuroendocrine 
mechanisms.  Ciba  Found  Symp 
105:47-65,  1984. 

Taylor,  A.N.;  Branch,  B.J.;  Nelson,  L.R; 
Lane,  LA.;  and  Poland,  RE.  Prenatal 
ethanol  and  ontogeny  of  pituitary-adrenal 
responses  to  ethanol  and  morphine. 
Alcohol  3:255-259,  1986. 

Theoharides,  T.C.;  Singh,  L.K.;  Boucher, 
W.;  Pang,  X.;  Letourneau,  R.;  Webster, 
E.;  and  Chrousos,  G.  Corticotropin- 
releasing  hormone  induces  skin  mast  cell 
degranulation  and  increased  vascular 
permeability,  a  possible  explanation  for  its 
proinflammatory  effects.  Endocrinology 
139:403-^13,  1997. 

Tortorella,  C;  Malendowicz,  L.K.; 
Andreis,  P.G.;  Markowska,  A.;  Neri,  G.; 
Mazzocchi,  G.;  and  Nussdorfer,  G.G. 
Effects  of  interleukin-l(3  on  steroidogene- 
sis in  Leydig  cells  of  the  rat  testis:  In  vivo 
and  in  vitro  studies.  Biomed  Res 
14:209-215,  1993. 

Tuominen,  K.,  and  Korpi,  E.R. 
Diminished  stress  responses  in  the  alco- 
hol-sensitive ANT  rat  line.  Pharmacol 
Biochem  Behav  40:409-415,  1991. 

Uddin,  S.;  Wilson,  T.;  Emanuele,  M.A.; 
Williams,  D.;  Kelley,  M.R;  and 
Emanuele,  N.  Ethanol-induced  alterations 
in  the  posttranslational  processing,  but 
not  secretion  of  luteinizing  hormone- 
releasing  hormone  in  vitro.  Alcohol  Clin 
Exp  Res  20:556-560,  1996. 

Ulisse,  S.;  Fabbri,  A.;  and  Dufau,  M.L. 
Corticotropin-releasing  factor  receptors 


and  actions  in  rat  Leydig  cells.  J  Biol 
Chem  264:2156-2163,  1989. 

van  Praag,  H.M.  Faulty  Cortisol/ 
serotonin  interplay.  Psychopathological 
and  biological  charaterisation  of  a 
new,  hypothetical  depression  subtype 
(SeCA  depression).  Psychiatry  Res 
65:143-157,  1996. 

Veldman,  R.G.,  and  Meinders,  A.E.  On 
the  mechanism  of  alcohol-induced 
pseudo-Cushing's  syndrome.  Endocr  Rev 
17:262-268,  1996. 

Villalta,  J.;  Ballesca,  J.L.;  Nicolas,  J.M.; 
Martinez  de  Osaba,  M.J.;  Antunez,  E.; 
and  Pimentel,  C.  Testicular  function  in 
asymptomatic  chronic  alcoholics:  Relation 
to  ethanol  intake.  Alcohol  Clin  Exp  Res 
21:128-133,  1997. 

Vincent,  S.R.  Nitric  oxide:  A  radical  neu- 
rotransmitter in  the  central  nervous  sys- 
tem. Prog  Neurobiol  42:129-160,  1994. 

Wand,  G.S.  Alcohol,  the  hypothalamic- 
pituitary-adrenal  axis,  and  hormonal 
tolerance.  In:  Zakhari,  S.,  ed.  Alcohol  and 
the  Endocrine  System.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  23.  NIH  Pub. 
No.  93-3533.  Bethesda,  MD:  the 
Institute,  1993.  pp.  251-270. 

Wand,  G.S.,  and  Dobs,  A.S.  Alterations  in 
the  hypothalamic-pituitary-adrenal  axis  in 
actively  drinking  alcoholics.  J  Clin 
Endocrinol  Metab  72:1290-1295,  1991. 

Webster,  E.L.;  Tracey,  D.E.;  Jutila,  M.A.; 
Wolfe,  S.A.,  Jr.;  and  DeSouza,  E.B. 
Corticotropin-releasing  factor  receptors  in 
mouse  spleen:  Identification  of  receptor- 
bearing  cells  as  resident  macrophages. 
Endocrinology  127:440^52,  1990. 

Weinberg,  J.  Hyperresponsiveness 

to  stress:  Differential  effects  of  prenatal 


80 


Effects  of  Alcohol  on  the  Neuroendocrine  System 


ethanol  on  males  and  females.  Alcohol  Clin 
Exp  Res  12:647-652,  1988. 

Weinberg,  J.  Prenatal  ethanol  exposure 
alters  adrenocortical  development  of  off- 
spring. Alcohol  Clin  Exp  Res  13:73-83, 1989. 

Zakhari,  S.  ed.  Alcohol  and  the  Endocrine 
System.  National  Institute  on  Alcohol  Abuse 
and  Alcoholism  Research  Monograph  No. 
23.  NIH  Pub.  No.  93-3533.  Bethesda, 
MD:  the  Institute,  1993. 


Zhu,  Q.;  Van  Thiel,  D.H.;  and  Gavaler, 
J.S.  Effects  of  ethanol  on  rat  Sertoli  cell 
function:  Studies  in  vitro  and  in  vivo. 
Alcohol  Clin  Exp  Res  21 :1409-1417, 
1997. 

Zou,  J.-Y.;  Martinez,  D.B.;  Neafsey,  E.J.; 
and  Collins,  MA.  Binge  ethanol-induced 
brain  damage  in  rats:  Effect  of  inhibitors 
of  nitric  oxide  synthase.  Alcohol  Clin  Exp 
Res  20: 1406-1411,  1996. 


81 


MOLECULAR  AND  CELLULAR 

RESPONSES  TO  CHRONIC 

ETHANOL  EXPOSURE 


Chapter  4 

Neuroadaptation  to  Ethanol  at  the 
Molecular  and  Cellular  Levels 


Paula  L.  Hoffman,  Ph.D.,  A.  Leslie  Morrow,  Ph.D., 
Tamara  J.  Phillips,  Ph.D.,  and  George  R.  Siggins,  Ph.D. 

KEY  WORDS:  AOD  (alcohol  or  other  drug)  tolerance;  biological  adaptation; 
chronic  AODE  (effects  of  AOD  use,  abuse,  and  dependence);  brain  function; 
AOD  dependence;  homeostasis;  AOD  withdrawal  syndrome;  AOD  sensitivity; 
memory;  learning;  central  nervous  system;  membrane  channel;  neurotransmitter 
receptors;  dopamine;  serotonin;  opioid  receptors;  adenylate  cyclase;  protein  kinases; 
signal  transduction;  gene  regulation;  neuropeptides;  arginine;  vasopressin;  anti 
alcohol  craving  agents; gender  differences; genetics  and  heredity;  literature  review 


DEFINITIONS  drinking  has  ceased  or  been  reduced; 

the  intake  of  alcohol  to  relieve  signs 
Alcohol  dependence  currently  has        c    ■  ,    ,         ,         ,    ,.rr-     u- 

..    .  ,,      ,r..  .       .      .    ,,£„,,       or  withdrawal;  and  difficulties  in  con- 

well-  denned  diagnostic  cntena  (DSM-  ...        ....  .  ,  ,    . 

Tr7  rA        •         t^     i  •  .  •     *         ■    ■  trolling  drinking,  with  a  strong  desire 

IV  [American  Psychiatric  Association  &  &'  & 

1994]  and  ICD-10  [World  Health  or  compulsion  to  drink  (Tabakoff  and 

Organization] ),  including  the  presence  Hoffman  1996a).  All  of  these  symptoms 

of  alcohol  tolerance;  the  presence  of  may  be  considered  to  arise  as  a  result 

an  alcohol  withdrawal  syndrome  when  of  changes  in  brain  function  that 

P.L.  Hoffman,  Ph.D.,  is  a  professor  in  the  Department  of  Pharmacology,  University  of  Colorado 
Health  Sciences  Center,  4200  East  9th  Ave.,  Box  C-236,  Denver,  CO  80262-0001.  A.  Leslie  Morrow, 
Ph.D.,  is  associate  director  of  the  Bowles  Center  for  Alcohol  Studies  and  associate  professor  of  psychiatry 
and  pharmacology  at  the  University  of  North  Carolina  School  of  Medicine,  3027  Thurston  Bowles 
Bldg.,  CB  7178,  Chapel  Hill,  NC  27599-7178.  T.J.  Phillips,  Ph.D.,  is  a  professor  in  the  Department 
of  Behavioral  Neuroscience,  School  of  Medicine,  Oregon  Health  Sciences  University  and  a  research 
geneticist  at  Veterans  Affairs  Medical  Center,  Research  Division,  R&D-32,  3710  SW  US  Veterans 
Hospital  Rd.,  Portland,  OR  97201.  G.R.  Siggins,  Ph.D.,  is  a  professor  in  the  Department  of 
Neuropharmacology,  CVN-12,  The  Scripps  Research  Institute,  10550  North  Torrey  Pines  Rd.,  La 
folia,  CA  92037. 


85 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


occur  following  chronic  exposure  to 
alcohol  (ethanol).  It  is  generally  believed 
that  these  changes  represent  adaptations 
of  the  brain  to  ethanol:  acutely,  ethanol 
produces  changes  in  the  function  of  a 
number  of  neuronal  systems,  and  the 
consequence  of  homeostatic  responses 
to  these  changes,  induced  by  the 
chronic  presence  of  ethanol  in  the 
brain,  is  the  production  of  the  alcohol 
dependence  syndrome  (Tabakoff  and 
Hoffman  1996#).  These  homeostatic 
responses  may  involve  positive  or  neg- 
ative feedback  mechanisms  or  may 
reflect  more  permanent,  qualitative 
changes  in  synaptic  connections,  which 
may  be  either  beneficial  or  harmful  to 
the  organism  as  a  whole  (Hyman  and 
Nestler  1996).  One  of  the  challenges 
of  alcohol  and  other  drug  research  is 
to  identify  the  changes  in  central  ner- 
vous system  (CNS)  function  that 
reflect  adaptation  to  the  chronic  pres- 
ence of  the  drug,  and  to  define  the 
relationship  of  those  cellular,  bio- 
chemical, and  molecular  changes  to 
various  aspects  of  the  dependence 
syndrome.  In  this  review,  we  will  out- 
line certain  CNS  changes  produced 
by  chronic  ethanol  treatment  that  are 
thought  to  be  neuroadaptive,  and  we 
will  attempt  to  determine  how  these 
changes  lead  to  or  reflect  the  behav- 
ioral aspects  of  neuroadaptation  to 
ethanol,  which  are  defined  in  the  fol- 
lowing sections. 

Ethanol  Tolerance 

Tolerance  to  ethanol  is  defined  as 
acquired  resistance  to  the  effects  of 
the  drug,  but  it  is  a  more  complex 
phenomenon  than  is  suggested  by  this 
definition  (Tabakoff  and  Rothstein 


1983;  Kalant  1998).  Tolerance  may  be 
metabolic  or  dispositional,  meaning  that 
previous  exposure  to  ethanol  results  in  a 
change  in  the  metabolism,  distribution, 
or  excretion  of  the  drug  such  that  the 
organism  is  exposed  to  lower  blood  or 
brain  ethanol  levels  after  ethanol  inges- 
tion. Functional  tolerance,  which  is  the 
focus  of  this  review,  refers  to  an  increase 
in  cellular  resistance  to  the  effects  of 
ethanol  in  the  CNS.  Tolerance  can  occur 
within  the  time  that  a  single  dose  of 
ethanol  is  ingested  (acute  or  within  - 
session  tolerance)  or  after  repeated 
exposure  to  ethanol  (chronic  tolerance). 
A  form  of  tolerance  known  as  "rapid 
tolerance"  has  also  been  described,  in 
which  exposure  of  an  animal  to  a  single 
dose  of  ethanol  generates  tolerance 
when  a  second  dose  is  administered 
8-24  hours  after  the  first  dose.  It  is 
not  known  whether  similar  or  identical 
mechanisms  underlie  the  development 
of  these  different  forms  of  tolerance. 

Ethanol  tolerance  can  also  be  influ- 
enced by  environmental  variables.  For 
example,  it  has  been  shown  that  if  a 
task  is  practiced  under  the  influence  of 
ethanol,  tolerance  to  the  effect  of 
ethanol  on  the  performance  of  that  task 
develops  more  rapidly  than  if  no  prac- 
tice occurs  ("behaviorally  augmented 
tolerance").  In  this  case,  it  appears  to 
be  the  rate  of  tolerance  development 
that  is  affected,  as  the  same  maximal 
degree  of  tolerance  develops  eventu- 
ally in  both  situations  (practice  or  no 
practice).  There  is  also  evidence  for  a 
role  of  classical  conditioning  in  the 
development  of  ethanol  tolerance  (as 
for  tolerance  to  other  drugs  [e.g., 
Siegel  1976]),  and  it  has  even  been 
suggested  that  the  occurrence  of 


86 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


ethanol  tolerance  is  entirely  dependent 
on  learned  responses  (Tabakoff  and 
Hoffman  1992).  It  has  been  demon- 
strated that  when  ethanol  administra- 
tion is  repeatedly  paired  with  a  distinct 
environment,  tolerance  to  various  effects 
of  ethanol  can  be  demonstrated  in  that 
environment  but  is  absent  if  the  animal 
is  tested  in  a  different  environment. 
Tolerance  in  this  case  results  from  a 
conditioned  response,  which  is  associ- 
ated with  cues  in  the  environment  in 
which  ethanol  was  administered  and 
which  is  opposite  to  the  initial  effect  of 
ethanol.  For  example,  animals  that  have 
been  treated  with  ethanol  in  a  particular 
environment,  and  are  then  tested  with 
saline  in  that  same  environment,  show 
hyperthermia  in  response  to  the  saline 
treatment.  This  hyperthermia  counters 
the  hypothermic  effect  of  ethanol  and 
produces  tolerance  in  the  "cued"  envi- 
ronment, but  it  does  not  occur  if  the 
animal  is  tested  in  an  environment  dis- 
tinct from  that  in  which  ethanol  was 
administered  (Le  et  al.  1979;  Mansfield 
and  Cunningham  1980;  Crowell  et  al. 
1981;  Melchior  and  Tabakoff  1981, 
1985).This  type  of  tolerance  has  been 
called  conditional  or  environment- 
dependent  tolerance.  Its  characteristics 
are  different  from  those  of  environ- 
ment-independent tolerance^  which 
can  be  demonstrated  regardless  of  the 
environment  in  which  the  animals  are 
treated  with  ethanol  and  tested.  Envi- 
ronment-dependent tolerance  is 
induced  by  lower  doses  of  ethanol, 
administered  as  repeated  doses  (e.g., 
by  injection),  and  persists  for  a  longer 
time  than  environment-independent 
tolerance,  which  can  be  induced  by 
giving  higher  doses  of  ethanol  in  a 


continuous  manner,  such  as  in  a  liquid 
diet  or  by  vapor  inhalation  (Melchior 
and  Tabakoff  1981).  It  is  not  known 
whether  environment-dependent  and 
-independent  forms  of  ethanol  toler- 
ance result  from  different  underlying 
mechanisms;  however,  studies  in  the 
area  of  learning  and  memory  may  pro- 
vide some  clues. 

For  considering  the  cellular,  neuro- 
chemical, and  molecular  changes  that 
may  underlie  ethanol  tolerance,  we  have 
previously  found  it  useful  to  consider 
tolerance  within  a  framework  that  had 
been  used  to  discuss  the  neurobiology 
of  learning:  that  is,  intrinsic  and  extrin- 
sic neuronal  systems  (Tabakoff  and 
Hoffman  1992).  Extrinsic  systems  are 
those  that  influence  the  development, 
maintenance,  or  expression  of  tolerance 
or  other  neuroadaptive  phenomena,  but 
do  not  encode  tolerance  within  them- 
selves. Intrinsic  systems,  on  the  other 
hand,  do  encode  specific  information, 
such  as  tolerance  to  a  specific  effect  of 
ethanol,  presumably  by  changes  in 
synaptic  efficacy  in  a  particular  neu- 
ronal pathway.  There  are  a  number  of 
behaviors  and  physiological  functions 
that  are  affected  by  ethanol  and  that  are 
commonly  used  to  assess  ethanol  toler- 
ance. These  include  ethanol-induced 
incoordination,  loss  of  righting  reflex 
("sleep  time"),  changes  in  body  temper- 
ature, and  anxiolytic  effects.  Identifi- 
cation of  intrinsic  systems  would  be 
facilitated  by  knowledge  of  the  neuronal 
systems  that  mediate  behaviors  or 
physiological  functions  that  become  tol- 
erant to  ethanol,  but  in  many  cases  (e.g., 
sleep  time,  body  temperature  changes) 
these  systems  are  not  well  characterized 
or  are  very  complex.  Another  means 


87 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  identifying  possible  intrinsic  sys- 
tems is  by  analyzing  the  acute  effects 
of  ethanol.  It  is  assumed  that  adapta- 
tions will  occur  in  neurochemical 
and/or  neurophysiological  systems 
that  are  initially  perturbed  by  ethanol. 
Whether  the  same  or  different  neu- 
ronal systems  mediate  the  occurrence 
of  acute,  rapid,  and  chronic  tolerance, 
as  well  as  the  environment-dependent 
and  -independent  forms  of  tolerance, 
is  a  question  that  has  only  begun  to 
be  addressed. 

Ethanol  Dependence 

As  described  above,  the  alcohol 
dependence  syndrome  comprises  not 
only  tolerance,  but  also  physical 
dependence  and  the  compulsion  to 
use  alcohol  ("psychological  depen- 
dence"). Physical  dependence  on 
alcohol  is  defined  primarily  by  a  char- 
acteristic set  of  symptoms  and  signs 
that  appear  when  the  chronic  adminis- 
tration or  consumption  of  relatively 
high  doses  of  alcohol  is  abruptly  ter- 
minated. The  signs  and  symptoms  of 
withdrawal  are,  in  most  instances, 
opposite  in  nature  to  the  signs  of 
acute  intoxication,  and  follow  a  char- 
acteristic time  course  after  the  cessa- 
tion of  alcohol  intake  (see  Tabakoff 
and  Rothstein  1983).  In  humans, 
alcohol  withdrawal  signs  and  symp- 
toms can  be  divided  into  early  and 
late  stages;  the  early  stages  (first  36 
hours)  are  characterized  by  tremors, 
convulsions,  mild  diaphoresis,  and 
hallucinations,  and  the  later  stages 
include  severe  autonomic  dysfunction 
and  delirium.  Many  of  the  early  signs 
of  withdrawal,  such  as  tremors,  con- 
vulsions, and  temperature  aberrations, 


can  also  be  observed  in  animal  models 
of  physical  dependence  on  alcohol 
(Tabakoff  and  Rothstein  1983). 

The  biological  mechanisms  that 
underlie  the  signs  and  symptoms  of 
alcohol  withdrawal  are,  by  definition, 
the  factors  that  are  responsible  for 
physical  dependence.  As  discussed  for 
tolerance,  one  can  attempt  to  identify 
the  neuronal  systems  involved  in  phys- 
ical dependence  and  withdrawal  based 
on  the  systems  that  mediate  the  physi- 
ological functions  that  are  disrupted 
during  withdrawal.  However,  in  most 
cases,  the  systems  underlying  the  signs 
that  are  used  to  assess  alcohol  with- 
drawal in  animals  (e.g.,  spontaneous 
and  "handling-induced"  convulsions, 
temperature  aberrations)  are  complex 
and  poorly  understood.  Nevertheless, 
it  is  believed  that  the  chronic  exposure 
of  the  susceptible  neuronal  systems  to 
alcohol  results  in  an  adaptation  that 
generates  exaggerated  and  maladap- 
tive responses  to  normal  neuronal 
input  after  the  cessation  of  alcohol 
intake. 

It  has  been  postulated  that  the 
same  adaptations  that  lead  to  toler- 
ance to  certain  effects  of  alcohol  will, 
when  alcohol  intake  or  exposure 
is  terminated,  produce  the  signs  of 
withdrawal  (e.g.,  Goldstein  and  Gold- 
stein 1968).  For  example,  a  change 
that  produced  tolerance  to  the 
hypothermic  effect  of  alcohol  could, 
in  the  absence  of  alcohol,  result 
in  hyperthermia.  However,  differences 
in  the  time  course  of  development 
of  physical  dependence  and  functional 
tolerance,  as  well  as  the  demonstra- 
tion of  treatments  that  can  block  the 
development  of  tolerance  but  not 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


withdrawal  signs,  and  vice  versa  (e.g., 
Tabakoff  and  Ritzmann  1977;  Snell 
et  al.  1996^),  suggest  that  different 
neuroadaptations  may  underlie 
physical  dependence  and  tolerance  to 
various  effects  of  alcohol. 

The  degree  to  which  environmen- 
tal variables  affect  physical  depen- 
dence on  alcohol  is  not  clear. 
Although  it  has  been  hypothesized 
that  conditioned  withdrawal  signs 
may  occur  when  abstinent  individuals 
are  exposed  to  the  environment  previ- 
ously associated  with  opiate  with- 
drawal (O'Brien  et  al.  1986),  there  is 
very  little  evidence  for  conditioning 
associated  with  alcohol  withdrawal. 
Such  "conditioned  withdrawal" 
would  be  important  in  that  it  could 
trigger  increased  alcohol  consump- 
tion, to  alleviate  the  perceived  with- 
drawal signs  or  symptoms.  However, 
another  controversial  issue  is  whether 
individuals  will  continue  to  consume 
alcohol  in  order  to  relieve  the  symp- 
toms of  withdrawal  associated  with 
physical  dependence.  There  have  been 
several  studies  (Woods  and  Winger 
1971;  Mello  and  Mendelson  1977; 
Roehrs  and  Samson  1981;  Tang  et  al. 
1982)  showing  that  neither  humans 
nor  animals  will  continue  alcohol  con- 
sumption in  order  to  avoid  with- 
drawal signs  and  symptoms. 

On  the  other  hand,  the  depen- 
dence syndrome  also  includes  the 
compulsion  to  consume  alcohol.  The 
concepts  of  craving  and  need  have  been 
described  as  psychological  depen- 
dence, but  they  are  rooted  in  neuro- 
logical processes.  Many  investigators 
have  operationally  defined  craving  by 
measuring  drug-seeking  behaviors 


(Schuster  1986).  Drug- seeking  behav- 
iors are  responses  that  have  previously 
been  associated  with  the  administra- 
tion of  a  drug  and  are  believed  to 
reflect  the  reinforcing  efficacy  of  a 
drug  (Schuster  and  Johanson  1981). 
Of  particular  interest  for  this  review  is 
the  role  that  neuroadaptive  processes 
may  play  in  altering  the  reinforcing 
properties  of  alcohol,  and  thus  the 
degree  of  alcohol-seeking  behavior. 

In  animals,  alcohol  is  not  an  effica- 
cious reinforcer,  and,  in  almost  all 
studies  of  alcohol  self- administration, 
the  animal  must  be  induced  to  drink 
alcohol  by  procedures  such  as  food 
deprivation,  adulteration  of  the  taste 
of  alcohol,  or  acclimatization  to  grad- 
ually increasing  concentrations  of 
alcohol  (e.g.,  Meisch  1984;  Samson 
1987).  The  difficulty  in  demonstrat- 
ing alcohol  self- administration  has 
been  attributed  to  the  aversive  effects 
of  alcohol,  which  can  overshadow  its 
reinforcing  effects.  Thus,  one  could 
speculate  that  if  tolerance  to  the  aver- 
sive properties  of  alcohol  were  devel- 
oped, the  reinforcing  properties  might 
become  more  prominent.  This  possi- 
bility is  supported  by  studies  showing 
that  repeated  exposure  of  animals  to 
alcohol  can  result  in  conditioned  place 
preference  and  conditioned  taste  pref- 
erence for  alcohol  (e.g.,  Crawford  and 
Baker  1982;  Reid  et  al.  1985).  How- 
ever, this  hypothesis  has  yet  to  be 
tested  directly,  for  example,  by  block- 
ing the  development  of  tolerance  to 
aversive  properties  of  alcohol  and 
measuring  alcohol  self- administration. 
It  is  also  not  clear  whether  tolerance 
to  the  reinforcing  effects  of  alcohol 
can  develop.  The  occurrence  of  such 


89 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


tolerance  could  lead  to  continued 
alcohol  intake  (see  chapter  7).  Thus, 
neuroadaptive  changes  in  the  neu- 
ronal systems  that  mediate  either  the 
aversive  or  reinforcing  properties  of 
alcohol  could  conceivably  contribute 
to  alterations  in  the  reinforcing  effects 
of  alcohol  that  would  lead  to  craving, 
or  a  compulsion  to  drink  alcohol. 
Another  possible  mechanism  that  may 
contribute  to  increased  alcohol  intake 
is  the  development  of  sensitization  to 
the  effects  of  alcohol. 

Ethanol  Sensitization 

Drug-induced  behavioral  sensitization 
may  be  defined  as  the  augmentation 
of  a  response  to  a  drug  with  repeated 
exposures.  The  first  demonstration  of 
behavioral  ethanol  sensitization  was 
that  of  Masur  and  Boerngen  (1980), 
who  treated  mice  for  up  to  60  days, 
once  daily,  with  1-3.5  g/kg  ethanol 
and  showed  increases  in  the  initial 
locomotor  responses  to  some  ethanol 
doses.  Ethanol  sensitization  has  been 
little  investigated  since  publication  of 
those  results,  but  some  studies  do 
exist  (Masur  et  al.  1986;  Newlin  and 
Thomson  1991;  Phillips  et  al.  1991; 
Cunningham  and  Noble  1992; 
Phillips  et  al.  1994;  Cunningham 
1995;  Phillips  et  al.  1995). 

There  is  speculation  as  to  whether 
ethanol  sensitization  is  a  determinant 
factor  in  addictive  behavior  (Wise  and 
Leeb  1993).  Hunt  and  Lands  (1992) 
suggested  that  sensitization  may 
increase  the  probability  of  the  devel- 
opment of  uncontrolled  ethanol 
intake,  and  Newlin  and  Thomson 
(1991)  asserted  that  sensitization  to 
ethanol  might  reflect  greater  reward 


value  of  the  drug.  It  is  our  view  that 
this  has  yet  to  be  conclusively  demon- 
strated. Several  key  questions  remain 
unanswered.  Does  sensitization 
develop  to  the  reinforcing  effects  of 
ethanol?  If  so,  does  it  contribute  to 
increased  alcohol  consumption?  Are 
alcoholics  sensitized?  These  are  all 
important  questions  that  have  yet  to 
be  answered. 

It  is  possible  that  locomotor  sensi- 
tization reflects  intensified  reinforce- 
ment, or  at  least  reflects  alteration  in  a 
system  that  results  in  increased  sensi- 
tivity to  reinforcing  stimuli  (Stewart 
and  Badiani  1993;  Wise  and  Leeb 
1993).  A  recent  study  by  Lessov  and 
Phillips  (1998)  demonstrated  that 
ethanol  sensitization  can  be  relatively 
long-lasting,  suggesting  that  lasting 
neuroadaptive  mechanisms  may  be 
engaged.  The  behavioral  significance 
of  sensitization  may  be  that  it  results  in 
increased  efficacy  of  ethanol  reinforce- 
ment and  thus  increases  the  likelihood 
that  ethanol  will  be  self- administered. 
In  other  words,  molecular  changes 
accompanying  sensitization  might  be 
viewed  as  adaptive  phenomena,  per- 
mitting facilitation  within  a  system, 
and  making  responses  controlled  by 
that  system  easier  to  elicit  on  future 
encounters  (see  Stewart  and  Badiani 
1993).  If  the  sensitized  system  should 
happen  to  be  one  contributing  to  the 
reward  experienced  with  ethanol  intake, 
then  it  is  easy  to  see  how  sensitization 
might  result  in  increased  drinking.  More 
plainly,  ethanol  would  come  to  more 
easily  elicit  its  reinforcing  effects. 

An  example  of  increased  drug  reward 
with  repeated  administration  is  seen  in 
a  study  by  Lett  (1989),  who  measured 


90 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


conditioned  place  preference  to  amphet- 
amine, morphine,  or  cocaine  and 
found  that  prior  exposure  to  these 
drugs  enhanced  conditioned  place 
preference.  Perhaps  more  direct  evi- 
dence is  provided  by  studies  like  that 
of  Horger  and  colleagues  (1990), 
who  showed  that  rats  sensitized  to 
cocaine  acquired  cocaine  self- adminis- 
tration at  lower  doses.  Such  studies 
have  not  been  performed  with  ethanol. 
Sparse  data  using  rats  and  chronic 
treatment  with  drugs  other  than 
ethanol  show  an  increase  in  ethanol 
consumption  or  preference  following 
amphetamine,  nicotine,  or  morphine 
treatment  (Potthoff  et  al.  1983;  Levy 
and  Ellison  1985;  Hubbell  et  al. 
1988;  McMillan  and  Snodgras  1991; 
Fahlke  et  al.  1994).  Such  results  may 
be  interpretable  as  increased  sensitivity 
to  ethanol  reinforcement  due  to  chronic 
drug  treatment.  However,  develop- 
ment of  behavioral  sensitization  due 
to  the  long-term  drug  administration 
was  not  measured  in  any  of  these 
studies.  We  know  of  no  studies  that 
have  measured  ethanol  sensitization 
and  then  looked  at  its  influence  on 
ethanol  drinking,  or  vice  versa.  A 
negative  genetic  correlation  between 
ethanol  sensitization  and  ethanol  con- 
sumption was  found  in  BXD  recombi- 
nant inbred  strains  (Phillips  et  al. 
1995).  Those  strains  more  prone  to 
sensitization  consumed  less  ethanol. 
However,  independent  groups  of  ani- 
mals were  tested  for  the  two  traits. 
There  is  a  need  for  more  research  to 
establish  the  importance  of  the  sensiti- 
zation phenomenon  to  addiction. 

One  reason  for  the  relative  paucity 
of  ethanol  sensitization  reports  in  the 


literature  may  be  that  there  appears  to 
be  an  important  species  difference  in 
its  occurrence:  it  can  be  demonstrated 
in  the  mouse  but  may  be  difficult  to 
demonstrate  in  the  rat  (Masur  et  al. 
1986),  the  research  animal  that  has 
been  most  commonly  used  in  studies 
of  sensitization  to  other  drugs.  How- 
ever, as  already  mentioned,  there  are 
genotype-dependent  differences 
among  mouse  strains  in  propensity 
toward  the  development  of  ethanol 
sensitization.  This  has  also  proven  to 
be  important  in  some  rat  studies  of 
acute  ethanol  stimulant  effects  (Waller 
et  al.  1986;  Krimmer  1991).  How- 
ever, we  do  not  know  of  any  strain 
surveys  or  other  genetic  investigations 
of  ethanol  sensitization  susceptibility 
in  rats. 

Memory  as  Neuroadaptation: 
Cellular,  Biochemical,  and 
Molecular  Models 

Although  the  focus  of  this  review  is 
ethanol-induced  neuroadaptation,  it 
should  be  recognized  that  the  physio- 
logical processes  of  learning — the 
process  by  which  new  information 
about  the  environment  is  acquired — 
and  memory — the  process  by  which 
that  knowledge  is  retained — also 
reflect  adaptations  at  the  cellular,  bio- 
chemical, and  molecular  levels  in  the 
CNS.  In  other  words,  learning  and 
memory,  like  tolerance  and  depen- 
dence, can  be  viewed  as  adaptive 
responses  of  the  CNS  to  external 
stimuli.  Therefore,  studies  of  learning 
and  memory  have  the  potential  to 
provide  clues  to  the  mechanisms 
underlying  neuronal  adaptation  to 
alcohol  and  other  drugs,  as  well  as 


91 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


approaches  to  investigate  such  adapta- 
tion at  the  molecular  level. 

There  is  no  question  that  learning 
and  memory  are  complex  processes. 
Memory  has  been  classified,  for  exam- 
ple, as  explicit  or  implicit  (Bailey  et  al. 
1996).  Explicit  memory  is  conscious 
recall,  while  implicit  memory  is  non- 
conscious  recall  of  motor  skills  and 
other  tasks.  Implicit  memory  includes 
associative  forms  (e.g.,  classical  condi- 
tioning) and  nonassociative  forms, 
and  it  is  believed  to  involve  changes  in 
the  same  pathways  that  are  used  in  the 
learning  process.  These  aspects  of 
implicit  memory  are  reminiscent  of 
the  characteristics  of  neuroadaptation 
to  alcohol  and  other  drugs,  such  as 
tolerance,  and  the  pathways  that  are 
altered  in  implicit  memory  are  those 
previously  defined  as  intrinsic  systems 
in  the  Ethanol  Tolerance  section  of 
this  chapter. 

Memory  is  also  often  divided  into 
short-term  and  long-term  compo- 
nents. It  is  perhaps  too  simplistic  to 
compare  these  processes,  for  example, 
to  the  acute  and  chronic  forms  of 
ethanol  tolerance;  however,  it  is  of 
interest,  in  terms  of  the  mechanisms 
of  neuroadaptation  to  ethanol,  to 
note  that  short-term  memory  is 
believed  to  involve  covalent  modifica- 
tion of  existing  proteins,  whereas 
long-term  memory  is  more  stable  and 
requires  transcription,  translation,  and 
the  growth  of  new  synaptic  connec- 
tions (Bailey  et  al.  1996).  Similar 
processes  may  be  involved  in  neuroad- 
aptations  to  ethanol,  as  discussed  in 
more  detail  below. 

The  studies  of  Kandel  and  his  col- 
leagues provide  a  model  for  investigating 


the  mechanisms  that  underlie  neuro- 
adaptation. One  of  the  examples  of 
implicit  memory  that  has  been  studied 
in  detail  by  this  group  is  sensitization 
of  the  gill/siphon  withdrawal  reflex  in 
the  marine  snail,  Aplysia.  This  animal 
learns  to  respond  to  a  variety  of  previ- 
ously neutral  stimuli  once  it  has  been 
exposed  to  a  potentially  threatening 
stimulus.  The  neural  pathway  for  sen- 
sitization of  the  reflex  has  been  deter- 
mined, and  it  involves  activation  of 
facilitatory  interneurons  that  synapse 
on  sensory  neurons,  to  strengthen  the 
connection  between  the  sensory  neu- 
rons and  their  central  target  neurons 
(Kandel  1991;  Bailey  et  al.  1996). 
The  Kandel  group  has  been  able  to 
study  the  neuronal  mechanisms  of 
sensitization  in  this  system  by  isolating 
a  component  of  the  reflex,  a  monosy- 
naptic connection  between  sensory 
neurons  and  their  target  cells.  This 
monosynaptic  connection  can  be  studied 
in  cell  culture,  where  serotonin,  which 
is  released  upon  stimulation,  can  sub- 
stitute for  the  training  stimulus.  Bio- 
physical studies  established  that  short- 
term  (minutes  to  hours)  changes  in 
synaptic  effectiveness  in  this  pathway 
are  attributable  to  enhanced  release  of 
neurotransmitter,  due  to  a  change  in 
activity  of  a  neuronal  potassium  channel 
(Bailey  et  al.  1996).  This  change  also 
occurs  in  the  longer  term  sensitization 
(days  to  weeks),  but  studies  using 
inhibitors  of  transcription  and  transla- 
tion both  in  the  intact  animal  and  in 
cell  culture  demonstrated  that  long-term 
sensitization  depends  on  new  protein 
and  RNA  synthesis,  and  is  also  associated 
with  structural  changes — that  is,  the 
growth  of  new  synaptic  connections 


92 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


(Kandel  1991;  Bailey  et  al.  1996).  The 
idea  that  memory  processes  and  neuro- 
adaptation to  ethanol  may  have  certain 
common  mechanisms  is  not  new,  and 
it  is  interesting  to  note  that  studies 
have  also  showed  that  inhibitors  of 
protein  synthesis  could  block  the 
development  of  chronic  ethanol  toler- 
ance in  animals  (LeBlanc  et  al.  1976; 
Bitran  and  Kalant  1993). 

Kandel  and  his  colleagues  have 
defined  the  molecular  substrates  of 
memory  in  the  Aplysia  system  in  some 
detail.  Using  a  variety  of  biochemical 
and  molecular  biological  techniques, 
they  have  provided  evidence  that  the 
serotonin-induced  activation  of  adenylyl 
cyclase  (AC)  and  subsequent  activation 
of  protein  kinase  A  (PKA)  are  critical 
factors  in  short-  and  long-term  sensiti- 
zation. Initially,  phosphorylation  of  the 
potassium  channel  or  related  proteins 
results  in  decreased  activity  of  the  channel, 
producing  the  enhanced  neurotrans- 
mitter release  that  underlies  short-term 
sensitization.  In  the  cell  nucleus,  PKA- 
induced  phosphorylation  of  a  3',5'-cyclic 
adenosine  monophosphate  response 
element  binding  (CREB)-like  protein 
that  binds  to  the  3 ',5 '-cyclic  adenosine 
monophosphate  response  element  (CRE) 
is  necessary  for  long-term  sensitization 
(Kandel  1991;  Bailey  et  al.  1996). 
These  investigators  have  also  identified 
an  immediate  early  gene  (IEG),  a  3,5'- 
cyclic  adenosine  monophosphate 
(cAMP)-regulated  transcription  factor 
that  was  demonstrated  to  be  involved  in 
the  development  of  long-term  sensitiza- 
tion by  the  use  of  antisense  oligonucleo- 
tides (Alberini  et  al.  1994).  In  addition, 
the  structural  alterations  associated  with 
long-term  sensitization  in  Aplysia  have 


been  suggested  to  be  related  to  down- 
regulation  of  cell  adhesion  molecules  that 
are  related  to  nerve  cell  adhesion  mole- 
cule (NCAM)  (Mayford  et  al.  1992). 

These  studies  help  to  define  the 
molecular  and  neurochemical  pathways 
required  for  an  elementary  form  of 
memory.  It  is  interesting  to  note  that  the 
cAMP  system  has  also  been  implicated 
in  learning  and  memory  in  Drosophila, 
where  mutants  that  produce  defects  in 
various  portions  of  the  cAMP  signaling 
pathway  are  deficient  in  the  ability  to 
learn  a  classical  conditioning  task  (Byers 
et  al.  1981;  Levin  et  al.  1992).  Inter- 
estingly, recent  studies  with  Drosophila 
also  demonstrated  the  importance  of 
proteins  in  the  cAMP/PKA  cascade  for 
ethanol  sensitivity  and  tolerance  (Moore 
et  al.  1998).  Cyclic  AMP  is  also  impor- 
tant for  the  maintenance  of  long-term 
potentiation  in  certain  areas  of  mammalian 
brain  (e.g.,  Hopkins  and  Johnston  1988), 
and  deficiencies  in  learning  and  long-term 
memory  have  been  demonstrated  in  mice 
with  mutations  in  AC  (Wu  et  al.  1995)  or 
CREB  (Bourtchuladze  et  al.  1994).  It 
could  prove  very  informative  to  study 
ethanol  tolerance  and  dependence  in 
these  mice,  especially  in  view  of  the 
effects  of  ethanol  on  the  cAMP  signal- 
ing system  that  are  described  later  in 
this  chapter. 

This  discussion  is  not  meant  to  sug- 
gest, however,  that  the  cAMP  signal 
transduction  system  is  the  only  system 
that  should  be  investigated  with  regard 
to  neuroadaptation  to  ethanol.  In 
another  invertebrate  system,  for  exam- 
ple, protein  kinase  C  (PKC)  and  cal- 
cium/calmodulin-dependent  protein 
kinases,  as  well  as  various  IEGs,  have 
been  implicated  in  associative  learning 


93 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


(Alkon  and  Nelson  1990).  The  advan- 
tage of  using  these  invertebrate  models  is 
the  well-characterized  nature  of  the 
neuronal  systems  involved  in  the  behav- 
iors being  measured.  The  relative  sim- 
plicity of  the  studied  pathways  allows 
for  the  construction  of  testable  models 
of  the  biochemical  and  molecular 
changes  that  underlie  learning  and 
memory.  Furthermore,  there  is  substan- 
tial evidence  that  the  signaling  pathways 
that  are  implicated  in  adaptive  processes 
in  these  simple  models  may  also  be  applic- 
able to  neuroadaptation  in  the  vertebrate 
CNS.  Consideration  should  therefore 
be  given  both  to  using  simpler  models, 
but  those  involving  the  whole  organism 
(such  as  Aplysia),  to  study  neuroadapta- 
tion to  ethanol,  and  to  using  the  studies 
of  learning  and  memory  that  have  been 
carried  out  with  these  models  to  guide 
research  into  the  cellular,  neurochemical, 
and  molecular  pathways  that  mediate 
ethanol-induced  neuroadaptation. 

WHAT  IS  KNOWN  ABOUT 
ETHANOL-INDUCED 
CHANGE  AND 
NEUROADAPTATION 
AT  THE  CELLULAR, 
BIOCHEMICAL,  AND 
MOLECULAR  LEVELS 
IN  THE  CNS? 

Systems  That  Show 
Changes  After  Chronic 
Ethanol  Exposure 

Ligand-Gated  Ion  Channels 

Within  the  past  10  years  it  has  become 
increasingly  apparent  that  a  major  site 
of  action  of  ethanol  is  ion  channels. 


Ion  channels  are  multimeric  struc- 
tures, comprising  different  subunits, 
that  gate  ions  following  subtle  changes 
in  tertiary  structure.  Ethanol  is  more 
hydrophobic  than  water  and  in  some 
instances  can  replace  water  within  mol- 
ecular structures,  but  it  does  not  have 
the  hydrogen- bonding  capacity.  Ethanol 
readily  enters  molecular  sites  within 
multimeric  ion  channels  modifying 
intermolecular  forces  and  bonds  that  are 
important  for  the  open-close-inactivation 
kinetic  properties  of  channels.  The 
diversity  of  channel  composition  due 
to  the  multimeric  structures  results  in 
subtypes  of  channels  that  are  differen- 
tially distributed  across  brain  regions. 
There  are  also  regional  differences  in 
the  sensitivity  of  ion  channels  to  the 
action  of  ethanol. 

The  acute  intoxicating  and  incoor- 
dinating  effects  of  ethanol  may  be 
related  to  inhibition  of  subtypes  of  N- 
methyl-D-aspartate  (NMDA)-glutamate 
receptors  and  potentiation  of  certain 
subtypes  of  GABAA  receptor  ion 
channels.  The  effects  of  ethanol  on 
glycinergic,  nicotinic  cholinergic,  and 
serotonergic  receptors,  and  voltage- 
gated  calcium  and  potassium  channels, 
are  also  considered  in  this  chapter.  A 
considerable  amount  of  data  suggests 
that  alterations  in  NMDA  and  GABAA 
receptors,  and  voltage-gated  calcium 
channels,  contribute  to  the  development 
of  ethanol  tolerance,  dependence,  and 
withdrawal.  Many  of  the  other  chan- 
nels that  are  sensitive  to  ethanol  at  rel- 
evant concentrations  have  not  been 
studied  in  this  context.  In  the  following 
sections  we  attempt  to  summarize  the 
existing  data  and  better  understand  the 
relationships  between  the  effects  of 


94 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


ethanol  on  ion  channels  and  the 
chronic  behavioral  effects  of  ethanol. 

Glutamate  Receptor  Ion  Channels. 
There  are  three  classes  of  ionotropic 
glutamate  receptors,  including  the 
NMD  A,  the  kainate,  and  the  AMPA 
(L-a-amino-3-hydroxy-5-methyl-4- 
isoxazole  propionate)  receptor  subtypes 
(Sommer  and  Seeburg  1992;  Sprengel 
and  Seeburg  1993;  Hollmann  and 
Heinemann  1994;  McBain  and  Mayer 
1994;  Bettler  and  Mulle  1995).  The 
kainate  and  AMPA  receptors  mediate 
fast  excitatory  neurotransmission.  The 
NMDA  receptor  is  coupled  to  an  ion 
channel  that,  when  activated,  is  per- 
meable to  calcium  as  well  as  monovalent 
cations.  The  pharmacology  of  the 
NMDA  receptor  is  well  defined 
(Collingridge  and  Lester  1989; 
McBain  and  Mayer  1994).  The  func- 
tion of  the  NMDA  receptor  is  voltage 
dependent,  meaning  that  the  response 
to  NMDA  is  increased  as  the  cell  is 
depolarized.  The  voltage  dependence 
is  the  result  of  Mg2+  binding  within 
the  ion  channel.  Mg2+  blocks  the 
channel  but  is  released  upon  cellular 
depolarization.  The  NMDA  receptor- 
channel  complex  also  contains  bind- 
ing sites  for  several  other  agents  that 
influence  receptor  activity.  Glycine  is  a 
co- agonist  at  the  receptor,  and  both 
glutamate  and  glycine  are  required  for 
activation  of  the  receptor.  NMDA 
receptor  activity  is  also  affected  by 
phencyclidine,  which  binds  within  the 
ion  channel,  as  does  the  uncompeti- 
tive inhibitor,  dizocilpine  (MK-801). 
The  complex  also  contains  binding 
sites  for  Zn2+  and  polyamines. 

Both  the  non-NMDA  glutamate 
receptors  and  the  NMDA  receptors 


consist  of  a  number  of  subunits. 
GluRl-GluR4  are  believed  to  form 
AMPA  receptors,  while  GluR5-GluR7 
appear  to  form  low-affinity  kainate 
receptors.  There  are  also  proteins 
called  KA-1  and  KA-2  that  can  bind 
kainate  with  high  affinity  and  can 
form  functional  receptors  when 
expressed  with  members  of  the  GluR 
family  (Hollmann  and  Heinemann 
1994).  The  NMDA  receptor  consists 
of  (a)  an  NR1  subunit,  which  has 
eight  splice  variants  and  is  ubiqui- 
tously localized  in  the  brain,  and  (b)  a 
family  of  NR2  subunits  (NR2A-D) 
(Monyer  et  al.  1992;  Nakanishi  1992; 
Sugihara  et  al.  1992;  McBain  and 
Mayer  1994).  Receptors  composed  of 
NR1  and  NR2  subunits  show 
responses  to  NMDA  that  are  charac- 
teristic of  native  receptors,  and  the 
NR2  subunits  significantly  influence 
the  pharmacological  properties  of  the 
NMDA  receptor  (Ishii  et  al.  1993; 
Scheetz  and  Constantine-Paton 
1994).  There  is  evidence  that  both 
the  non-NMDA  and  the  NMDA 
receptor  subunits  can  be  phosphory- 
lated  by  serine-threonine  and  tyrosine 
kinases,  and  phosphorylation  may 
influence  activity  and/or  localization 
of  the  receptors  in  the  cell  (Tabakoff 
and  Hoffman  1996£). 

The  characteristics  of  the  NMDA 
receptor-gated  channel,  including  its 
slow  activation  and  permeability  to 
calcium,  contribute  to  its  involvement 
in  learning  and  memory  processes 
(long-term  potentiation  [LTP])  and 
in  neuronal  development  (Collingridge 
and  Lester  1989).  When  overstimulated, 
the  NMDA  receptor  plays  a  role  in  gen- 
erating seizure  activity  and  excitotoxic 


95 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


(and  possibly  apoptotic)  neuronal 
death  (Choi  1988,  1992).  These  prop- 
erties of  the  NMDA  receptor,  as  well 
as  the  fact  that  its  function  is  potently 
inhibited  by  acute  ethanol  exposure, 
suggest  that  it  might  play  a  role  in 
ethanol-induced  neuroadaptation. 

Hyperexcitability  of  the  CNS  is  a 
key  component  of  ethanol  withdrawal 
(Tabakoff  and  Rothstein  1983).  Both 
a  reduction  in  GABA-mediated  inhi- 
bition and  a  supersensitive  NMDA 
response  may  be  involved.  One  of  the 
earliest  findings  suggesting  that  chronic 
ethanol  exposure  produces  up-regula- 
tion  of  glutamate  receptors  was  an 
increase  in  [3H]glutamate  binding 
reported  in  hippocampus  of  human 
alcoholics  (Michaelis  et  al.  1990).  A 
more  recent  postmortem  study  of 
human  alcoholics  found  an  increase  in 
NMDA-sensitive  glutamate  binding, 
and  in  binding  of  an  NMDA  receptor 
antagonist,  in  frontal  cortex  (Freund 
and  Anderson  1996).  Hoffman's  labora- 
tory has  reported  increases  in  the  den- 
sity of  NMDA  receptors  in  C57BL/6 
mice  treated  chronically  with  a  7  per- 
cent ethanol  liquid  diet.  Seven  days  of 
chronic  ethanol  ingestion,  leading  to 
functional  tolerance  to  and  physical 
dependence  on  ethanol,  led  to  signifi- 
cantly increased  [3H]MK-801  binding 
in  hippocampal  membranes  (Grant  et 
al.  1990).  These  animals  were  depen- 
dent on  ethanol,  as  indicated  by  mea- 
surement of  withdrawal  seizures.  An 
autoradiographic  study  using  the  same 
ethanol  administration  paradigm  also 
reported  increased  [3H]MK-801 
binding  in  cortex,  hippocampus,  and 
striatum  (Gulya  et  al.  1991).  Extensions 
of  these  experiments  with  membrane 


binding  techniques  found  significant 
increases  in  MK-801  binding  only  in 
hippocampus,  but  not  cerebral  cortex 
(Snell  et  al.  1993).  These  studies 
found  that  both  [3H]MK-801  and 
NMDA- specific  [3H] glutamate  binding 
were  significantly  increased  in  hip- 
pocampus by  chronic  ethanol  treatment, 
but  there  were  no  changes  in  binding 
of  [3H]glycine  or  [3H]CGS19755,  a 
competitive  NMDA  receptor  antago- 
nist. Sanna  and  colleagues  (1993)  also 
found  a  significant  increase  in  MK- 
801  binding  in  hippocampal  tissue  of 
rats  given  ethanol  for  6  days;  the  rats 
showed  withdrawal  signs  upon  cessa- 
tion of  ethanol  treatment. 

In  contrast,  there  have  been  a  few 
studies  in  which  increases  in  NMDA 
receptor  binding  were  not  observed 
after  chronic  ethanol  exposure.  Carter 
and  colleagues  (1995)  saw  no  changes 
in  MK-801  binding  in  brains  of  mice 
bred  selectively  for  differences  in  sus- 
ceptibility to  ethanol  withdrawal  seizures 
(withdrawal  seizure  prone  [WSP]  and 
withdrawal  seizure  resistant  [WSR] 
mice)  following  24  hours  of  ethanol 
exposure,  although  handling-induced 
withdrawal  seizures  could  be  observed 
in  these  animals.  This  study  contrasts 
with  earlier  work  using  WSP  and  WSR 
mice,  in  which  the  WSP  mice  were 
reported  to  have  a  higher  density  of 
MK-801  binding  sites  in  hippocampus 
than  WSR  mice,  and  in  which  chronic 
(7  days)  ingestion  of  ethanol  in  a  liq- 
uid diet  increased  MK-801  binding  in 
hippocampus  of  both  lines  of  mice 
(Valverius  et  al.  1990).  Differences 
between  these  studies  include  differ- 
ent durations  and  methods  of  ethanol 
exposure.  In  particular,  in  the  study  of 


96 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Carter  and  colleagues,  mice  were 
exposed  to  ethanol  vapor  by  inhalation 
and  were  given  pyrazole  to  retard  ethanol 
metabolism.  Pyrazole  interacts  with 
the  NMDA  receptor  (Pereira  et  al. 
1992)  and  may,  therefore,  confound 
the  results.  Another  difference  is  that, 
with  the  method  used  in  the  study  by 
Valverius  and  colleagues,  both  spon- 
taneous and  handling-induced  with- 
drawal seizures  are  observed;  that  is, 
withdrawal  symptomatology  is  more 
severe  in  this  paradigm. 

Rudolph  and  colleagues  (1997) 
reported  no  changes  or  only  very  small, 
but  significant,  increases  in  MK-801 
binding  or  binding  of  other  ligands  to 
the  NMDA  receptor  in  brains  of  rats 
that  were  treated  chronically  with 
ethanol  by  a  number  of  different  meth- 
ods. Although  some  of  these  methods 
have  previously  been  reported  to  pro- 
duce physical  dependence  in  the  rats, 
no  measures  of  withdrawal  signs  or 
symptoms  were  included  in  this  study. 

Alterations  in  ligand  binding  may 
reflect  changes  in  NMDA  receptor 
subunit  composition,  and  there  have 
also  been  several  investigations  of 
NMDA  receptor  subunit  expression  in 
brain  following  chronic  ethanol  expo- 
sure. Trevisan  and  colleagues  (1994) 
found  that  12  weeks  of  ingestion  of 
an  ethanol-containing  liquid  diet  by 
rats  resulted  in  an  increase  in  the  level 
of  NR1  immunoreactivity  in  the  hip- 
pocampus, but  not  in  the  cortex,  stria- 
tum, or  nucleus  accumbens.  Long-term 
treatment  of  rats  with  ethanol  (12 
weeks)  was  also  found  to  be  required 
to  increase  NRI  immunoreactivity  in 
the  ventral  tegmental  area  (VTA), 
whereas  1  and  6  weeks  of  chronic  5 


percent  ethanol  liquid  diet  were  not 
sufficient  (Ortiz  et  al.  1995).  Interest- 
ingly, studies  of  the  levels  of  mRNA 
for  NMDA  receptor  subunits  have 
indicated  that  chronic  ethanol  treat- 
ment of  rats  (by  repeated  gavage  for 
several  days)  does  not  change  NRI 
mRNA,  but  increases  NR2A  and 
NR2B  mRNA  levels  in  hippocampus 
and  cortex  (Follesa  and  Ticku  1995).  In 
contrast,  Snell  and  colleagues  (1996#) 
found  that  ingestion  of  an  ethanol- 
containing  liquid  diet  by  C57BL/6 
mice  for  7  days  resulted  in  an  increase 
in  NRI  and  NR2A  proteins  in  several 
brain  areas,  with  no  change  in  mRNA 
levels.  They  suggested  that  the  increase 
in  these  two  receptor  subunits  in  hippo- 
campus was  consistent  with  their  pre- 
vious finding  of  an  increase  in  MK-801 
binding  in  this  brain  region.  Since 
MK-801  binding  involves  both  an 
NRI  and  NR2  subunit,  an  increase  in 
binding  could  be  due  to  changes  in 
the  subunit  expression  (presumably 
receptor  subunit  stoichiometry),  with- 
out necessarily  an  increase  in  the  den- 
sity of  receptors. 

A  factor  that  may  influence  changes 
in  NMDA  receptor  properties  following 
chronic  ethanol  treatment  is  suggested 
by  the  fact  that  both  stress  and  treat- 
ment with  glucocorticoids  have  been 
shown  to  increase  NMDA  receptor 
binding  in  a  manner  similar  to  ethanol 
treatment  (Yoneda  et  al.  1994;  Tabakoff 
and  Hoffman  1996&).  Since  ethanol 
increases  glucocorticoids  and  is  a  stres- 
sor, it  is  possible  that  stress-induced 
glucocorticoids  may  play  a  role  in 
chronic  ethanol-induced  increases  in 
NMDA  receptor  binding  in  brain.  The 
degree  of  stress  induced  by  different 


97 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol  administration  paradigms 
could  then  influence  the  results. 

For  the  most  part,  the  literature 
supports  the  view  that  chronic  ethanol 
administration  resulting  in  physical 
dependence  and  withdrawal  convulsions 
is  accompanied  by  an  up-regulation  of 
NMDA  receptors  and/or  increased 
expression  of  NMDA  receptor  subunit 
proteins  in  various  brain  areas.  Little 
or  no  work  has  yet  been  done  to  deter- 
mine if  these  changes  in  NMDA 
receptor  properties  are  reflected  in 
receptor  function  in  the  adult  brain. 
However,  a  number  of  studies  of  chronic 
ethanol  effects  on  NMDA  receptor 
properties  in  neuronal  culture  have 
been  performed.  Chronic  exposure  of 
primary  cultures  of  cerebellar  granule 
neurons  or  cerebral  cortical  neurons 
to  ethanol  (e.g.,  100  mM  ethanol  for 
3  days)  resulted  in  enhanced  NMDA- 
stimulated  increases  in  intracellular 
Ca2+  (Iorio  et  al.  1992;  Ahern  et  al. 
1994),  as  well  as  increased  NMDA- 
stimulated  nitric  oxide  formation 
(Chandler  et  al.  1997).  Dizocilpine 
binding  was  also  increased  in  intact 
cerebellar  granule  neurons  that  had 
been  treated  chronically  with  ethanol, 
indicating  an  increase  in  NMDA 
receptor  number  after  this  treatment 
(Hoffman  et  al.  1995).  Increases  in 
expression  of  NMDA  receptor  subunits 
have  also  been  reported.  In  cerebellar 
granule  neurons,  chronic  ethanol  treat- 
ment produced  a  small  increase  in  NRI 
protein  and  a  decrease  in  NR2A  pro- 
tein, with  no  change  in  mRNA  for 
either  subunit.  The  same  treatment 
produced  larger  increases  in  the  gluta- 
mate  binding  protein  (mRNA  and 
protein  levels),  which  has  been  sug- 


gested to  be  a  component  of  a  complex 
of  proteins  that  has  ligand  binding 
sites  characteristic  of  NMDA  receptors 
(Hoffman  et  al.  1996).  In  primary  cul- 
tures of  cerebral  cortical  cells,  chronic 
ethanol  exposure  (50  mM,  5  days) 
increased  the  mRNA  level  for  the  NR2B 
subunit  and  increased  the  expression 
of  NRI  and  NR2B  proteins  (Follesa 
and  Ticku  1996;  Hu  et  al.  1996).  In 
HEK  293  cells  transfected  with 
NMDA  receptor  subunits,  chronic 
ethanol  treatment  (50  mM  or  greater 
for  24  hours)  did  not  alter  the  expres- 
sion of  any  receptor  subunit  but 
changed  the  sensitivity  of  the  recep- 
tors to  ifenprodil,  a  ligand  that  is 
selective  for  receptors  containing  the 
NR2B  subunit  (Blevins  et  al.  1997). 

Caution  must  be  used  in  extrapolat- 
ing the  results  seen  in  primary  cultures 
of  neurons  to  the  situation  in  the 
adult  brain,  since  the  neurons  in  culture 
are  undergoing  development.  The 
mechanism  by  which  ethanol  induces 
NMDA  receptor  changes  in  the  cultures 
may  be  different  from  the  mechanism 
in  the  adult  brain,  even  though  the 
receptor  changes  themselves  may  appear 
to  be  similar.  The  results  obtained 
with  recombinant  receptors  are  even 
more  problematic,  because  the  receptor 
subunits  are  overexpressed,  and  both 
regulation  of  receptor  expression  and 
receptor  stoichiometry  are  likely  to  be 
very  different  from  native  receptors. 

Nonetheless,  the  consequences  of 
NMDA  receptor  up-regulation  caused 
by  chronic  ethanol  exposure  can  be 
readily  measured  in  the  cell  culture 
models.  Ethanol-exposed  cerebellar 
granule  neurons  and  cerebral  cortical 
neurons  display  enhanced  sensitivity 


98 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


to  glutamate- induced  excitotoxicity 
(Chandler  et  al.  1993#;  Crews  and 
Chandler  1993;  Iorio  et  al.  1993; 
Ahern  et  al.  1994).  It  is  likely  that  this 
increased  sensitivity  to  neurotoxic  dam- 
age is  a  consequence  of  ethanol  with- 
drawal, because  ethanol,  while  present 
in  the  cellular  milieu,  inhibits  NMDA 
receptor  function  and  blocks  neuro- 
toxicity (Takadera  et  al.  1990;  Chandler 
et  al.  1993^).  Enhanced  susceptibility 
to  glutamate  receptor-mediated  neu- 
rotoxicity has  also  been  reported  in 
rats  that  were  exposed  chronically  to 
ethanol  by  inhalation  and  then 
injected  intrahippocampally  with 
NMDA  (Davidson  et  al.  1993).  This 
ethanol  withdrawal-induced  increase  in 
susceptibility  to  glutamate  excitotoxi- 
city may  represent  the  basis  for  the 
observation  of  neuronal  damage  in 
alcoholics  (Charness  1993). 

In  the  intact  animal,  another  con- 
sequence of  NMDA  receptor  up- 
regulation  may  be  the  generation  of 
alcohol  withdrawal  seizures  and/or 
convulsions.  Competitive  and  non- 
competitive antagonists  of  the  NMDA 
receptor  can  reduce  ethanol  withdrawal 
convulsions  in  mice  and  rats  (Grant  et 
al.  1990;  Morrisett  et  al.  1990;  Liljequist 
1991;  Kotlinska  and  Liljequist  1996). 
Furthermore,  the  time  course  for  the 
increase  in  hippocampal  dizocilpine 
binding  sites  in  mice  treated  chroni- 
cally with  ethanol  paralleled  the  time 
course  for  appearance  of  ethanol  with- 
drawal convulsions  (Gulya  et  al. 
1991).  Tremwel  and  colleagues 
(1994&)  reported  no  change  in  dizo- 
cilpine binding  at  48  hours  after  ethanol 
withdrawal,  a  time  when  overt  with- 
drawal signs  have  dissipated,  consistent 


with  the  hypothesis  that  NMDA  recep- 
tor up-regulation  is  a  transient  phe- 
nomenon that  contributes  to  withdrawal 
seizures  and  convulsions.  Snell  and 
colleagues  (1996&)  reported  that  gan- 
glioside  treatment  of  mice  during 
chronic  ethanol  exposure  resulted  in  a 
significant  attenuation  of  withdrawal 
seizures  and  blocked  the  up-regula- 
tion of  NMDA  receptors  (MK-801 
binding)  in  the  hippocampus.  The 
interpretation  of  these  findings  was 
that  ganglioside  treatment  may  have 
prevented  the  development  of  physi- 
cal dependence  in  the  mice.  All  of 
these  studies  are  consistent  with  the 
hypothesis  that  an  increase  in  NMDA 
receptor  number  and/or  function 
plays  a  role  in  the  generation  of  ethanol 
withdrawal  signs,  specifically  seizures 
and  convulsions  (i.e.,  ethanol  with- 
drawal hyperexcitability).  Electrophys- 
iological studies  with  hippocampal  slices 
obtained  from  mice  treated  chronically 
with  ethanol  also  indicated  the  pres- 
ence of  an  enhanced  NMDA  recep- 
tor-mediated component  of  synaptic 
excitation  during  ethanol  withdrawal 
(Whittington  et  al.  1995). 

NMDA  receptor  up-regulation  may 
also  contribute  to  changes  in  dopamine 
release  following  chronic  ethanol 
administration,  which  could,  in  turn, 
be  associated  with  altered  reinforcing 
effects  of  ethanol.  During  ethanol 
withdrawal,  the  firing  rates  and  number 
of  spontaneously  firing  dopaminergic 
neurons  in  the  VTA  are  significantly 
reduced,  and  the  release  of  dopamine 
in  the  nucleus  accumbens  of  rats 
undergoing  withdrawal  is  diminished 
(Diana  et  al.  1993).  This  decreased 
release  of  dopamine  could  be  reversed 


99 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


by  administration  of  dizocilpine  (MK- 
801).  That  is,  the  up-regulation  of 
NMDA  receptors  during  ethanol  with- 
drawal may  increase  the  level  of  tonic 
inhibition  of  dopamine  release  by  glu- 
tamate  (Imperato  et  al.  1990;  Tabakoff 
and  Hoffman  1996#),  leading  to 
changes  in  dopaminergic  function  that 
could  influence  ethanol  intake. 

The  data  reviewed  suggest  that 
changes  in  NMDA  receptor  function 
induced  by  chronic  ethanol  exposure 
can  contribute  to  ethanol  withdrawal 
hyperexcitability  and  withdrawal- 
induced  neurotoxicity.  These  findings 
suggest  the  possibility  of  developing 
therapeutic  agents  that  would  not 
only  reduce  ethanol  withdrawal  signs 
but  could  also  reduce  the  neuronal 
damage  seen  in  chronic  alcoholics. 

An  investigation  by  Miyakawa  and 
colleagues  (1997)  also  suggests  the  pos- 
sibility that  acute  ethanol  tolerance  may 
be  related  to  NMDA  receptor  function. 
Mice  that  lacked  Fyn,  a  tyrosine  kinase, 
were  found  to  be  more  sensitive  to  the 
hypnotic  effect  of  ethanol  than  wild- 
type  mice.  Furthermore,  "acute  toler- 
ance" to  ethanol  inhibition  of  NMDA 
receptor  responses  in  hippocampal  slices 
developed  in  control  mice,  but  not  in 
the  Fyn-deficient  mice.  These  findings 
suggest  that  posttranslational  modifi- 
cation of  the  NMDA  receptor  could 
contribute  to  short-term  tolerance  to 
ethanol  effects  (i.e.,  the  NMDA  recep- 
tor might  represent  an  "intrinsic  system" 
for  some  aspect  of  acute  tolerance  to 
ethanol),  but  behavioral  measures  of 
acute  tolerance  to  ethanol  were  not 
included  in  this  study. 

Several  studies  have  suggested  that 
chronic  ethanol  exposure  may  also 


modify  AMPA  receptor  properties. 
Although  12  weeks  of  ethanol  liquid 
diet  ingestion  did  not  increase  GluPvl  or 
GluR2  in  hippocampus  (Trevisan  et 
al.  1994),  this  treatment  did  increase 
GluRl  immunoreactivity  in  the  VTA 
and  substantia  nigra  (Ortiz  et  al. 
1995).  Chronic  oral  ethanol  exposure 
(20  percent  v/v,  28  weeks)  increased 
GluR3  subunit  mRNA  in  hippocampus 
by  15  to  30  percent,  while  GluRl  and 
GluR2  subunit  mRNAs  were  unaltered 
(Buckner  et  al.  1997).  Furthermore, 
Breese  and  colleagues  (1995)  found 
significant  increases  in  GluR2  and 
GluR3  subunit  immunoreactivity  in 
human  postmortem  hippocampal  tis- 
sue of  patients  with  alcohol  abuse  his- 
tories. Thus,  there  is  accumulating 
evidence  that  changes  in  AMPA  recep- 
tors may  also  occur  following  chronic 
ethanol  exposure. 

GABAA  Receptors.  GABA  is  the 
most  ubiquitous  inhibitory  neuro- 
transmitter in  the  brain.  It  interacts 
with  a  family  of  receptors  containing 
recognition  sites  for  the  anxiolytic 
and  sedative  benzodiazepines,  barbi- 
turates, and  endogenous  neuro- 
steroids.  These  binding  sites  are  linked 
allosterically  to  a  GABA  recognition 
site,  and  each  site  is  involved  directly 
or  indirectly  in  the  gating  properties 
of  integral  CI"  channels.  GABA 
receptor-mediated  activation  of  CI" 
conductance  results  in  membrane 
hyperpolarization  and  decreased  neu- 
ronal excitability  (Skolnick  and  Paul 
1982).  Ethanol  acutely  alters  the  gating 
properties  of  this  receptor  complex; 
however,  ethanol  binds  with  little  or 
no  affinity  to  recognition  sites  for 
GABA,  benzodiazepines,  barbiturates, 


100 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


and  cage  convulsants  on  GABAA 
receptors  (Davis  and  Ticku  1981). 

Chronic  ethanol  treatment,  using 
paradigms  that  are  known  to  produce 
tolerance  and  physical  dependence,  alters 
many  of  the  properties  of  GABAA  recep- 
tors in  brain  (table  1).  Chronic  ethanol 
exposure  is  associated  with  a  decrease 
in  the  sensitivity  of  GABAA  receptor- 
mediated  responses  in  cerebral  cortex 
(Morrow  et  al.  1988;  Sanna  et  al.  1993), 
nucleus  accumbens  (Szmigielski  et  al. 
1992),  spinal  cord  cultured  neurons 
(Mehta  and  Ticku  1988;  Ticku  1989), 
and  medial  septal  nucleus  (Criswell  et 
al.  1993).  In  cerebral  cortex,  muscimol- 
or  phenobarbital-stimulated  CI"  uptake 
is  decreased  following  chronic  ethanol 
inhalation  (Morrow  et  al.  1988).  The 
ability  of  ethanol  to  potentiate  GABA 
or  muscimol-stimulated  Cl~  uptake  is 
also  lost  following  chronic  ethanol 
administration  in  both  cortex  and 
cerebellum  (Allan  and  Harris  1987; 
Morrow  et  al.  1988;  Sanna  et  al. 
1993).  Benzodiazepine  enhancement 
of  muscimol-stimulated  chloride  flux 
is  reduced  in  the  cerebral  cortex  of 
mouse  microsacs,  while  the  functional 
efficacy  of  inverse  agonists  is  enhanced 
(Mehta  and  Ticku  1989;  Buck  and 
Harris  1990).  In  contrast,  potentia- 
tion of  Cl~  uptake  by  the  neuroactive 
steroids  3a,5a-tetrahydroproges- 
terone  (THP)  and  tetrahydrodeoxy- 
corticosterone  (THDOC)  is  enhanced 
in  ethanol-dependent  rats  (Devaud  et 
al.  1996). 

While  behavioral  and  functional  data 
clearly  suggest  that  chronic  ethanol 
administration  alters  GABAA  receptor 
function,  data  from  radioligand  binding 
studies  do  not  provide  an  explanation 


for  these  effects.  No  consistent  alter- 
ations in  GABAA  receptor  recognition 
sites  have  been  observed  (see  table  1). 
Therefore,  chronic  ethanol  administra- 
tion induces  functional  tolerance  of 
GABAA  receptors  without  reducing 
the  total  number  of  GABA-gated 
chloride  channels.  This  apparent  paradox 
has  driven  researchers  to  identify  alter- 
native mechanistic  explanations  for  these 
phenomena.  Many  studies  have  shown 
that  chronic  ethanol  administration  alters 
the  expression  of  various  GABAA 
receptor  subunits,  suggesting  that  alter- 
ations in  GABAA  receptor  expression 
may  account  for  alterations  in  GABAA 
receptor  function  (Morrow  et  al.  1990; 
Buck  et  al.  1991;  Montpied  et  al. 
1991;  Mhatre  and  Ticku  1992;  Mor- 
row et  al.  1992;  Mhatre  et  al.  1993; 
Devaud  et  al.  1995^,  1996,  1997). 

Chronic  ethanol  administration  dif- 
ferentially alters  the  expression  of  distinct 
GABAA  receptor  subunit  mRNAs  in 
the  cerebral  cortex  (Morrow  et  al.  1990; 
Montpied  et  al.  1991;  Devaud  et  al. 
1995^)  and  cerebellum  (Mhatre  and 
Ticku  1992;  Morrow  et  al.  1992).  The 
levels  of  GABAA  receptor  al  subunit 
mRNAs  and  peptides  are  reduced, 
whereas  a4  subunit  mRNAs  and  pep- 
tides are  increased  by  approximately 
equal  amounts  in  cerebral  cortex 
(Devaud  et  al.  1995&,  1997).  In  the 
cerebellum,  decreases  in  GABAA 
receptor  al  subunit  mRNA  and  increases 
in  a6  subunit  mRNA  levels  are  found 
(Mhatre  and  Ticku  1992;  Morrow  et 
al.  1992).  These  changes  in  subunit 
expression  suggest  that  alterations  in  the 
assembly  of  GABAA  receptors  could 
account  for  the  observed  changes  in 
receptor  function  and  binding.  For 


101 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


example,  the  increases  in  a4  and  ct6 
subunit  expression  could  explain  the 
increases  in  [3H]Ro  15-4513  binding 
(Mhatre  et  al.  1988)  and  inverse  agonist 


sensitivity  (Mhatre  et  al.  1988;  Buck 
and  Harris  1990)  following  chronic 
ethanol  administration.  The  increased 
expression  of  ct4  subunits  may  underlie 


Table  1.  Effects  of  Chronic  Ethanol  Administration  on 

GABAA  Receptor  Function,  Recognition 

Receptor  Property 

Alteration 

GAB  A- mediated  CI"  channel  function3 

Decreased 

GABA-mediated  CI"  channel  function0 

No  change 

Phenobarbital- mediated  CI"  flux3 

Decreased 

Ethanol-enhanced  CI"  flux0 

Abolished 

Benzodiazepine -enhanced  CI"  fluxc 

Decreased 

Inverse  agonist  modulation0 

Increased 

Neuroactive  steroid  modulation3 

Increased 

High -affinity  [3H]  muscimol  binding*-' 

No  change 

Low-affinity  [3H]muscimol  bindingd 

Decreased 

[3H]flunitrazepam  (flu)  binding0 

No  change 

[35S]TBPS  binding0 

No  change  or  increased 

GABA  enhancement  of  [3H]flu  binding01 

Decreased 

[3H]zolpidem  binding0 

Increased  or  no  change 

[3H]Ro  15-4513  binding0 

Increased 

al  Subunit  mRNA  and  peptides0 

Decreased 

a2  Subunit  mRNA  and  peptides3 

Decreased 

a3  Subunit  mRNA  and  peptides3 

No  change  or  decreased 

a4  Subunit  mRNA  and  peptides3 

Increased 

a5  Subunit  mRNAs3 

No  change 

a6  Subunit  mRNA  and  peptidesb 

Increased 

61  Subunit  mRNA  and  peptides3 

No  change  or  increased 

62  Subunit  mRNA  and  peptides0 

No  change  or  increased 

63  Subunit  mRNA  and  peptides3 

No  change  or  increased 

yl  Subunit  mRNA  and  peptides3 

Increased 

y2S  Subunit  mRNA3 

Increased 

y2L  Subunit  mRNA3 

No  change 

y2  Subunit  peptides3 

No  change 

y3  Subunit  mRNA3 

No  change 

8  Subunit  mRNA3 

No  change 

Note:  TBPS  =  tert-butyl-bicyclophosphorothionate. 

"Cerebral  cortex. 

bCerebellum. 

cCerebral  cortex  and  cerebellum. 

"Whole  brain. 

102 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


the  reduced  sensitivity  to  GABA  receptors  with  a4^2y2  subunits  are 

(Morrow  et  al.  1988)  and  benzodi-  less  sensitive  to  GABA  agonists  and 

azepine  agonists  (Buck  and  Harris  benzodiazepines  than  al(32y2  recep- 

1990),  since  recombinant  GABA  tors   (Whittemore   et  al.    1996). 


Sites,  and  Subunit  Expression  in  Cerebral  Cortex  and  Cerebellum. 


Source 


Martz  et  al.  1983;  Gonzalez  and  Czachura  1989; 

Criswell  et  al.  1993;  Sanna  et  al.  1993;  Devaud  et  al.  1996 

Frye  et  al.  1983;  Allan  and  Harris  1987;  Buck  and  Harris  1990 

Morrow  et  al.  1988 

Allan  and  Harris  1987;  Morrow  et  al.  1988 

Buck  and  Harris  1990 

Mehta  and  Ticku  1989;  Buck  and  Harris  1990 

Devaud  et  al.  1996 

Volicer  1980;  Volicer  and  Biagioni  1982 

Ticku  and  Burch  1980;  Unwin  and  Taberner  1980 

Karobath  et  al.  1980;  Volicer  and  Biagioni  1982;  Rastogi  et  al.  1986 

Thyagarajan  and  Ticku  1985;  Rastogi  et  al.  1986;  Sanna  et  al.  1993 

DeVries  et  al.  1987 

Devaud  et  al.  1995a,  19956 

Mhatre  et  al.  1988 

Morrow  et  al.  1990;  Montpied  et  al.  1991;  Mhatre  and  Ticku  1992; 

Morrow  et  al.  1992;  Mhatre  and  Ticku  1993;  Mhatre  et  al.  1993; 

Devaud  et  al.  19956,  1997 

Morrow  et  al.  1990;  Montpied  et  al.  1991;  Mhatre  et  al.  1993; 

Mhatre  and  Ticku  1994# 

Morrow  et  al.  1990;  Montpied  et  al.  1991;  Mhatre  et  al.  1993; 

Mhatre  and  Ticku  1994« 

Devaud  et  al.  19956,  1997 

Devaud  et  al.  19956 

Mhatre  and  Ticku  1992;  Morrow  et  al.  1992;  Mhatre  and  Ticku  19946 

Mhatre  and  Ticku  19946;  Devaud  et  al.  19956 

Morrow  et  al.  1992;  Devaud  et  al.  19956,  1997 

Devaud  et  al.  19956, 1997 

Mhatre  and  Ticku  19946;  Devaud  et  al.  19956,  1997 

Devaud  et  al.  19956 

Devaud  et  al.  19956 

Devaud  et  al.  1997 

Devaud  et  al.  19956 

Devaud  et  al.  19956 


103 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Ethanol-dependent  and  -withdrawn 
rats  are  also  sensitized  to  the  anticon- 
vulsant effects  of  the  neurosteroid 
3a,5a-THP  (Devaud  et  al.  1995a, 
1996).  This  effect  may  be  related  to 
the  increase  in  |3  and  yl  subunit  mRNAs 
and  peptides  following  chronic  ethanol 
exposure  (Mhatre  et  al.  1993;  Mhatre 
and  Ticku  1993;  Devaud  et  al.  1995£, 
1996,  1997),  because  homologous  (3 
subunit  expression  is  sufficient  for 
neurosteroid  potentiation  of  GABA 
responses  (Puia  et  al.  1990)  and  yl 
subunits  enhance  neurosteroid  sensitiv- 
ity in  recombinant  expression  studies 
(Puia  et  al.  1993). 

At  the  peak  of  ethanol  withdrawal, 
GABAA  receptor  subunit  mRNA  levels 
appear  to  be  in  a  state  of  flux,  whereas 
GABAA  receptor  subunit  peptide  levels 
exhibit  more  stable  changes.  GABAA 
receptor  al,  a4,  and  yl  subunit  mRNAs 
return  nearly  to  control  levels,  but  (32 
and  (33  subunit  mRNA  levels  increase, 
compared  with  both  control  and 
dependent  rats  (Devaud  et  al.  1996). 
At  this  time  point,  6-8  hours  after 
removal  of  ethanol,  GABAA  receptor 
protein  expression  remains  similar  to 
that  found  in  ethanol-dependent  rats; 
withdrawn  animals  show  reduced  levels 
of  al  protein  and  elevated  levels  of  a4, 
(32/3,  and  yl  protein  relative  to  pair-fed 
controls  (Devaud  et  al.  1997).  Thus, 
changes  in  GABAA  receptor  subunit 
mRNA  expression  are  dynamic  and 
reflect  the  rapidly  changing  state  of  CNS 
excitability.  In  contrast  to  the  changes 
in  mRNA  levels,  changes  in  GABAA 
receptor  subunit  peptides  may  reflect 
the  long-term  changes  associated  with 
ethanol  dependence  and  addiction. 
Clearly,  the  measurement  of  peptide 


expression  is  important  for  this  reason 
and  should  represent  an  important 
mechanism  of  adaptation  to  chronic 
ethanol  consumption. 

Recent  evidence  suggests  that  ethanol 
modulates  promoter  activity  for  GABAA 
receptor  a  and  (3  subunits  in  vitro 
(Russek  et  al.  1997;  S.J.  Russek  and 
D.H.  Farb,  personal  communication, 
June  1998).  Human  GABAA  receptor 
promoter  regions  were  cloned  and 
transfected  in  cultured  embryonic 
neurons  that  were  exposed  to  ethanol. 
Although  the  data  are  still  preliminary, 
it  appears  that  chronic  ethanol  exposure 
decreases  human  GABAA  receptor  al 
subunit  promoter  activity  and  increases 
GABAA  receptor  (3  subunit  promoter 
activity  (Russek  et  al.  1997).  This  work 
may  identify  mechanisms  of  regulation 
of  GABAA  receptor  genes  by  ethanol. 

Alterations  in  GABAA  receptor  func- 
tion and  gene  expression  are  regionally 
as  well  as  temporally  dependent.  Chronic 
ethanol  exposure  differentially  alters 
GABAA  receptor  subunit  expression 
in  the  hippocampus  compared  with 
the  cerebral  cortex.  For  example, 
chronic  ethanol  consumption  for  40 
days  (Matthews  et  al.  1998)  or  60 
bouts  of  chronic,  intermittent  exposure 
(Mahmoudi  et  al.  1997)  increase  a4 
subunit  expression  in  hippocampus. 
However,  shorter  duration  treatments 
with  ethanol  (14  days)  do  not  alter  a4 
subunit  peptides.  In  contrast,  a4  sub- 
unit  peptide  levels  are  significantly 
increased  in  the  cerebral  cortex  fol- 
lowing both  40  and  14  days  of  ethanol 
consumption  (Devaud  et  al.  1997). 
The  level  of  hippocampal  al  subunit 
peptide  is  not  altered  following  14 
days,  28  days  (Charlton  et  al.  1997), 


104 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


or  40  days  (Matthews  et  al.  1998)  of 
chronic  ethanol  consumption.  How- 
ever, a  significant  decrease  in  the  al 
subunit  peptide  level  is  found  in  the 
cerebral  cortex  of  the  same  animals 
following  both  14  (Devaud  et  al. 

1997)  and  40  days  (Matthews  et  al. 

1998)  of  chronic  ethanol  consumption. 
Finally,  GABAA  receptor  (32/3  subunit 
peptide  expression  is  not  altered  in  the 
hippocampus  following  either  period 
of  ethanol  consumption,  although 
GABAA  receptor  (32/3  subunit  expres- 
sion was  previously  shown  to  increase 
in  the  cerebral  cortex  of  the  same  ani- 
mals (Devaud  et  al.  1997;  Matthews 
et  al.  1998).  These  findings  suggest  that 
GABAA  receptor  gene  expression  is 
differentially  regulated  by  ethanol  in  the 
hippocampus  compared  with  the  cerebral 
cortex.  Therefore,  ethanol  regulation 
of  GABAA  receptor  gene  expression 
varies  across  brain  regions. 

Alterations  in  native  GABAA  recep- 
tor subunit  assembly  in  vivo  may  be 
an  adaptation  elicited  by  chronic  drug 
exposure  (Morrow  et  al.  1992;  Devaud 
et  al.  1995 b).  An  alteration  in  subunit 
assembly  could  confer  alterations  of 
functional  properties  of  receptors, 
with  no  change  in  the  total  number  of 
receptors  expressed.  This  mechanism 
may  play  a  role  in  neuroadaptations  to 
chronic  exposure  of  drugs  other  than 
ethanol  that  modulate  GABAA  receptors 
(Morrow  1995).  There  is  ample  evidence 
for  alterations  in  GABAA  receptor  sub- 
unit  composition  during  development 
(see  Morrow  1995  for  review),  suggest- 
ing that  changes  in  subunit  expression 
may  be  an  endogenous  regulatory 
mechanism  controlling  the  activity  of 
GABAA  receptors.  Similarly,  alterations 


in  subunit  expression  of  nicotinic 
cholinergic  (Mishina  et  al.  1986),  gluta- 
mate  (Sheng  et  al.  1994),  and  glycine 
receptors  (Malosio  et  al.  1991)  are 
also  observed  during  neuronal  devel- 
opment. Therefore,  we  propose  that 
ligand-gated  ion  channels  may  be  sub- 
ject to  alterations  in  subunit  assembly 
that  serve  to  modulate  receptor  func- 
tion in  vivo. 

Other  evidence  suggests  that  chronic 
ethanol  administration  may  result  in 
the  functional  uncoupling  of  GABA 
and  benzodiazepine  recognition  sites 
(DeVries  et  al.  1987;  Klein  et  al., 
1995#),  independent  of  alterations  in 
GABAA  receptor  gene  expression 
(Klein  et  al.  1995&).  Another  possible 
mechanism  mediating  neuroadaptations 
of  GABAA  receptors  following  chronic 
ethanol  exposure  involves  internalization 
of  the  receptor  complex.  Although  there 
is  some  evidence  that  GABA  receptors 
can  be  internalized  (Calkin  and  Barnes 
1994),  this  would  explain  the  loss  of 
GABAA  receptor  function  without 
altering  receptor  number  only  if  the 
receptors  remain  attached  to  the 
membrane  and  remain  detectable  in 
radioligand  binding  studies.  It  is  also 
possible  that  ethanol  affects  the  stoi- 
chiometry  of  GABAA  receptors.  How- 
ever, the  stoichiometry  of  native  GABAA 
receptors  is  undetermined.  Likewise,  a 
dissociation  of  subunits  could  account 
for  decrements  in  GABAA  receptor 
function  while  preserving  receptor 
number,  yet  be  nearly  impossible  to 
detect  using  currently  available  tech- 
niques. Conformational  changes  in 
receptor  structure  are  another  potential 
adaptation.  Although  this  mechanism 
is  more  suited  to  explain  rapid  tolerance, 


105 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


recent  data  narrowing  the  site  of  ethanol 
action  to  the  second  transmembrane 
domain  of  GABAA  receptors  (Mihic  et 
al.  1997)  suggest  that  ethanol  may  affect 
the  conformation  of  GABAA  receptors 
as  they  reside  in  the  membrane. 

The  tertiary  structure  of  proteins  is 
profoundly  affected  by  posttranslational 
modifications,  such  as  phosphorylation 
and  glycosylation.  Several  lines  of  con- 
vergent data  support  the  suggestion 
that  phosphorylation  or  dephosphory- 
lation  of  GABAA  receptors  may  play  a 
role  in  the  development  of  ethanol 
tolerance  and  dependence.  Most 
major  known  kinases  can  phosphorylate 
GABAA  receptor  subunits  (most  often 
the  (31)  in  vitro  (Browning  et  al.  1990; 
Porter  et  al.  1990;  Moss  et  al.  1995). 
Furthermore,  phosphorylation  of  GABAA 
receptors  has  been  shown  to  modulate 
receptor  function.  PKC  and  PKA  phos- 
phorylation of  GABAA  receptors  reduces 
receptor  activation  (Kellenberger  et  al. 
1992;  Leidenheimer  et  al.  1992; 
Krishek  et  al.  1994),  whereas  phos- 
phorylation by  Ca++/calmodulin- 
dependent  protein  kinase  II  or 
tyrosine  kinase  enhances  GABAA 
receptor  function  (Valenzuela  et  al. 
1995;  Wang  et  al.  1995). 

Several  studies  have  provided  evi- 
dence for  the  suggestion  that  post- 
translational modifications  may  underlie 
changes  in  GABAA  receptor  function 
following  ethanol  administration.  As 
discussed  previously,  acute  ethanol 
administration  induces  changes  in 
GABAA  receptor  function  that  may  be 
dependent  on  phosphorylation  of  par- 
ticular proteins.  Prolonged  ethanol 
exposure  might  be  expected  to  result  in 
long-term  changes  in  second  messenger 


systems,  including  kinase  activity.  The 
heterogeneity  of  GABAA  receptors 
expressed  in  vivo  has  precluded  defini- 
tively answering  this  question.  Expres- 
sion systems  with  stably  expressed, 
functional  GABAA  receptors  under 
the  control  of  an  inducible  promoter 
are  promising  model  systems.  When 
chronically  treated  with  ethanol,  Ltk- 
cells  show  changes  in  GABAA  receptor 
function  that  cannot  be  explained  by 
changes  in  gene  expression,  but  could 
be  explained  by  posttranslational  reg- 
ulation such  as  phosphorylation  (Klein 
et  al.  19956;  Harris  et  al.  1997).  How- 
ever, none  of  these  studies  has  directly 
demonstrated  that  phosphorylation  is 
involved  in  ethanol  modulation  of 
GABAA  receptor  function. 

Chronic  ethanol  administration  alters 
PKC  and  PKA  activity  in  neuronal  cells 
(for  a  review,  see  Diamond  and  Gordon 
1997),  and  these  alterations  have  been 
linked  to  the  development  of  cellular 
tolerance  to  the  effects  of  ethanol  on 
adenosine  uptake  (Coe  et  al.  1996). 
The  proposed  mechanism  for  this 
effect  involves  compartmentalization 
of  the  kinase  to  specific  regions  of  the 
cell  (Dohrman  et  al.  1996).  Interest- 
ingly, the  PKC -activating  effects  of 
ethanol  on  GABAA  receptors  are 
found  only  in  desensitized  receptors 
(Leidenheimer  et  al.  1992),  suggesting 
that  the  involvement  of  phosphorylation 
in  ethanol  modulation  of  GABAA 
receptors  may  be  state  dependent.  Alter- 
natively, the  functional  consequences 
of  phosphorylation  may  be  dependent 
on  the  subunit  composition  (Krishek 
et  al.  1994)  and  fine-tuning  of  func- 
tion may  be  achieved  by  selective  com- 
partmentalization of  either  GABAA 


106 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


receptors  or  kinases.  Kinase  activation/ 
inactivation  may  be  involved  in  the 
changes  in  gene  expression  observed 
after  chronic  ethanol  exposure.  As 
mentioned,  preliminary  evidence  sug- 
gests that  ethanol  modulates  promoter 
activity  for  GABAA  receptor  a  and  (3 
subunits  (Russek  et  al.  1997;  S.J. 
Russek  and  D.H.  Farb,  personal  com- 
munication, June  1998).  In  addition, 
altered  subunit  assembly  of  GABAA 
receptors  may  involve  induction  of  chap- 
erone  proteins,  whose  putative  cellular 
role  is  trafficking  of  proteins  between 
organelles,  thereby  controlling  receptor 
assembly  (Haas  1994).  In  support  of 
this  possibility,  chronic  ethanol  expo- 
sure up -regulates  expression  of  certain 
chaperones  in  neural  cell  cultures 
(Hsieh  et  al.  1996).  Thus,  it  is  con- 
ceivable that  certain  ethanol-sensitive 


chaperones  play  a  role  in  altering 
GABAA  receptor  subunit  assembly  fol- 
lowing chronic  ethanol  exposure.  The 
exact  mechanisms  that  account  for 
alterations  in  GABAA  receptor  func- 
tion following  chronic  ethanol  admin- 
istration remain  under  avid 
investigation.  These  mechanisms  may 
ultimately  encompass  several  of  the 
possibilities  described  in  figure  1 . 

As  a  caveat  to  the  proposed  postsy- 
naptic mechanisms,  it  is  also  possible 
that  ethanol  tolerance  and  depen- 
dence involve  alterations  in  presynap- 
tic processes.  Early  studies  suggested 
that  chronic  ethanol  administration 
did  not  affect  GAB  A  turnover  (Hunt 
and  Majchrowicz  1983).  However, 
more  recent  studies  using  in  vivo  micro- 
dialysis  sampling  suggest  that  chronic 
ethanol  treatment  increases  basal  GABA 


Altered  receptor  assembly 


Post-translational 
modifications 


Altered  stoichiometry 


Dissociation  of  subunits 


Figure  1.  Possible  mechanisms  of  GABAA  receptor  regulation.  ER  =  endoplasmic  reticulum; 
EtOH  =  ethanol;  P04  =  phosphate.  Adapted  from  Grobin,  A.C.;  Matthews,  O.B.;  Devaud, 
L.L.;  and  Morrow,  A.L.  The  role  of  GABAA  receptors  in  the  acute  and  chronic  effects  of 
ethanol.  Psycho-pharmacology  (Berl)  139:2-19,  1998. 


107 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


levels  (Dahchour  et  al.  1996).  Since 
multiple  GABA  transporters  have  now 
been  cloned,  the  hypothesis  that 
ethanol  modulates  GABA  neurotrans- 
mission via  altering  GABA  release  or 
uptake  merits  reinvestigation  with 
modern  molecular  techniques. 

Genetic  models  have  also  implicated 
GABAA  receptors  in  ethanol  tolerance 
and  dependence.  Animals  selected  for 
differences  in  ethanol  sensitivity  or 
withdrawal  show  differences  in  GABAA 
receptor  function.  Mice  selected  for 
differential  sensitivity  to  the  hypnotic 
effects  of  ethanol  display  corresponding 
differences  in  the  ataxic  effects  of  GABAA 
receptor  agonists  (Martz  et  al.  1983). 
Cerebral  microsacs  prepared  from  mice 
selected  for  high  sensitivity  to  the  hyp- 
notic effects  of  ethanol  show  ethanol 
potentiation  of  muscimol  stimulated 
Cl~  flux,  whereas  mice  insensitive  to 
the  hypnotic  effects  of  ethanol  do  not 
(Allan  and  Harris  1986).  WSP  and 
WSR  mice  show  differential  expression 
of  GABAA  receptor  subunit  mRNAs 
(Keir  and  Morrow  1994)  and  diver- 
gent changes  in  GABAA  receptor  sub- 
unit  mRNA  levels  after  chronic  ethanol 
treatment  (Buck  et  al.  1991).  Thus, 
genetic  models  of  ethanol  dependence 
selected  for  divergent  behavioral  respon- 
ses to  ethanol  support  the  hypothesis 
that  GABAA  receptors  underlie  the  neu- 
roadaptations  promulgated  by  ethanol 
(see  Crabbe  et  al.  1994  for  review). 

Inbred  mouse  strains  (such  as  the 
BXD  recombinant  inbreds)  have  been 
used  to  correlate  the  magnitude  of  acute 
ethanol  withdrawal  severity  with  bar- 
biturate withdrawal,  suggesting  that 
common  genes  may  influence  with- 
drawal from  these  two  GABAergic  drugs 


(Finn  and  Crabbe  1997).  Recent  stud- 
ies, using  a  powerful  two-step  approach 
to  quantitative  trait  loci  (QTL)  map- 
ping, have  identified  loci  on  murine 
chromosomes  1,  2,  4,  and  11  that 
influence  alcohol  withdrawal  severity 
(Buck  et  al.  1997).  Twenty  percent  of 
the  candidate  genes  put  forth  in  this 
study  are  related  to  GABA  neuro- 
transmission, including  the  three  genes 
that  encode  GABAA  receptor  al,  a6, 
and  y2  subunits.  QTL  mapping,  along 
with  our  rapidly  expanding  knowl- 
edge of  mammalian  genomes,  will 
allow  future  researchers  to  identify  and 
test  candidate  genes  in  genetic  linkage 
studies  of  human  and  animal  popula- 
tions susceptible  to  the  effects  of  ethanol. 
Taken  together,  findings  from  genetic 
animal  models  add  support  to  the 
suggestion  that  GABAA  receptor 
modulation  underlies  many  of  the 
behavioral  manifestations  of  ethanol 
dependence  and  withdrawal. 

Glycine,  5-Hydroxytryptamine3,  and 
Neuronal  Nicotinic  Acetylcholine  Ion 
Channel  Receptors.  At  pharmacologically 
relevant  concentrations,  ethanol  alters 
the  function  of  glycine,  5-hydroxy- 
tryptamine3  (5-HT3),  and  nicotinic 
acetylcholine  (nACh)  receptors, 
although  the  physiological  consequences 
of  these  actions  are  largely  unknown. 
Ethanol  potentiates  glycine  receptor- 
mediated  CI"  currents  in  spinal  cord 
neurons  (Celentano  et  al.  1988). 
Williams  and  colleagues  (1995)  showed 
that  intraventricularly  administered 
glycine  enhances  the  loss  of  righting 
reflex  produced  by  ethanol.  A  similar 
effect  could  be  produced  by  microin- 
jection of  the  glycine  precursor  D-serine. 
Strychnine  attenuated  the  effects  of 


108 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


these  agents  on  the  loss  of  righting,  indi- 
cating that  a  strychnine-sensitive  glycine 
site  was  involved.  The  effect  of  chronic 
ethanol  exposure  on  glycine  receptors 
has  not  been  studied  directly.  Gonzalez 
(1993)  demonstrated  there  was  no 
change  in  sensitivity  to  strychnine- 
induced  seizures  during  ethanol  with- 
drawal, but  the  sensitivity  of  glycine 
receptors  was  not  investigated.  Further 
studies  are  needed  to  determine  the 
role  of  glycine  receptors  in  ethanol- 
induced  hypnosis  and  to  determine 
whether  adaptations  in  these  receptors 
are  related  to  tolerance  to  the  hypnotic 
effects  of  ethanol. 

Ethanol  potentiates  5-HT3  receptors 
in  cell  culture  by  increasing  the  initial 
amplitude  of  the  ion  current  induced  by 
stimulation  with  serotonin  (Lovinger 
and  Peoples  1991;  Lovinger  and  White 
1991).  This  action  is  dependent  on 
the  open  channel  state  of  the  recep- 
tor; if,  for  example,  the  channel  is  in  a 
desensitized  state,  ethanol  has  no 
effect  on  ion  currents  (Sellers  et  al. 
1992).  Little  is  known  about  the 
action  of  ethanol  on  5-HT3  receptors 
in  vivo.  However,  there  is  increasing 
evidence  that  5-HT3  receptors  con- 
tribute to  the  activating  and  reinforcing 
effect  of  ethanol  (see  Sellers  et  al. 
1992  for  a  review).  The  effects  of 
chronic  ethanol  administration  on  5- 
HT3  receptors  have  not  been  studied, 
but  it  is  likely  that  studies  of  this  area 
would  provide  insight  into  the  role  of 
5-HT3  receptors  in  ethanol  reinforce- 
ment and  perhaps  craving. 

Electrophysiological  studies  have 
demonstrated  that  ethanol  has  effects 
on  nicotine-evoked  responses  in  brain. 
Mancillas  and  colleagues  (1986) 


reported  that  ethanol  enhanced  the 
effect  of  acetylcholine  in  hippocampal 
pyramidal  cells  without  affecting 
GABA-mediated  inhibition.  Criswell 
and  colleagues  showed  that  ethanol 
affects  responses  to  nicotine  from 
some,  but  not  all,  neurons  in  the  sub- 
stantia nigra  reticulata  and  blocks  the 
inhibitory  response  to  nicotine  on 
some,  but  not  all,  nACh  receptors  in 
the  medial  septum.  Nicotine  was 
demonstrated  to  act  on  presynaptic 
nACh  receptors  on  medial  septal  neu- 
rons to  facilitate  the  release  of  GABA 
(Yang  et  al.  1996&),  an  interpretation 
congruous  with  other  data  indicating 
that  nicotine  can  have  a  presynaptic 
action  (Schwartz  et  al.  1984;  Wonna- 
cott  et  al.  1989;  McGehee  et  al.  1995; 
Sershen  et  al.  1995;  Vizi  et  al.  1995). 
It  is  known  that  nicotine-induced 
dopamine  release  from  striatal  synap- 
tosomes  is  blocked  by  ethanol  (Con- 
nolly et  al.  1996),  whereas  ethanol 
does  not  affect  86Rb+  efflux  induced 
by  nicotine  from  thalamic  synapto- 
somes  (Collins  1996).  In  addition  to 
its  action  on  presynaptic  nACh  recep- 
tors, ethanol  has  been  demonstrated 
to  have  an  action  on  postsynaptic 
responses  to  nicotine  (i.e.,  the  nico- 
tine response  not  blocked  by  Mg2+). 
In  the  cerebellum,  ethanol  blocks  the 
nicotine-induced  inhibitor)'  response 
and  enhances  the  excitatory  response 
to  nicotine  (Yang  et  al.  1996#). 

The  chronic  effects  of  ethanol  on 
nACh  receptors  have  not  been  evalu- 
ated. Cross-tolerance  between  certain 
physiological  effects  of  chronic  nicotine 
and  ethanol  administration  have  been 
observed  (Collins  et  al.  1987;  Marks 
et  al.  1987;  Collins  et  al.  1990;  Welzl 


109 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


et  al.  1990;  deFiebre  and  Collins  1993). 
Additionally,  the  degree  of  nicotine 
tolerance  was  found  to  differ  between 
long  sleep  (LS)  and  short  sleep  (SS) 
mice  bred  for  differing  responses  to 
ethanol  (Collins  et  al.  1993;  deFiebre 
and  Collins  1993).  These  studies  sug- 
gest a  change  in  nACh  receptor  func- 
tion after  chronic  ethanol  treatment. 
However,  further  studies  are  clearly 
needed  to  establish  these  effects  and 
their  physiological  significance. 

Voltage-Sensitive  Ion  Channels 

Voltage-Gated  Calcium  Channels. 
Voltage-gated  calcium  channels  are 
classified  into  L-,  N-,  P-,  Q-,  and  T- 
types  based  on  their  electrophysiological 
and  pharmacological  properties  (for  a 
review,  see  Stea  et  al.  1995).  T-type 
calcium  channels  consist  of  a  variety  of 
low  voltage-activated  channels  that 
activate  transientiy  and  are  very  sensitive 
to  changes  in  resting  potential.  The  L-, 
N-,  P-,  and  Q-types  are  high  volt- 
age-activated channels,  which  overlap 
considerably  in  their  electrophysiolog- 
ical characteristics. 

Immunoprecipitation  studies  indi- 
cate that  L-type  calcium  channels  are 
heteroligomeric  complexes  consisting 
of  five  distinct  protein  subunits,  al, 
a2,  (3,  y,  and  6  (Campbell  et  al.  1988; 
Catterall  et  al.  1988).  The  al  subunit 
is  the  major  voltage -sensitive  and  pore- 
forming  subunit;  the  other  four  sub- 
units  are  believed  to  be  ancillary, 
modulatory  molecules  (Ellinor  et  al. 
1993).  In  addition,  different  classes  of 
al  subunits  have  been  isolated  and 
cloned.  Three  known  al  subunits,  alS, 
alC,  and  alD,  form  L-type  channels 
(reviewed  in  Stea  et  al.  1995).  The 


alC  and  alD  subunits  are  expressed 
in  rat  brain,  heart,  and  PC  12  cells. 

Ethanol  has  been  shown  to  inhibit 
depolarization-induced  calcium  influx 
through  voltage -gated  calcium  channels 
in  synaptosomes  and  presynaptic  nerve 
terminals  without  altering  basal  uptake 
(Leslie  et  al.  1983;  Skattebol  and 
Rabin  1987;  Dildy-Mayfield  and  Harris 
1995).  Ethanol  also  reduces  voltage- 
dependent  calcium  influx  in  cultured 
neuronal  and  PC  12  cells  (Messing  et  al. 
1986;  Skattebol  and  Rabin  1987).  Most 
studies  have  focused  on  L-type  voltage  - 
gated  calcium  channels.  Twombly  and 
colleagues  (1990)  found  that  both  T- 
type  and  L-type  voltage-gated  calcium 
channels  were  inhibited  by  ethanol, 
with  the  L-type  voltage-gated  calcium 
channels  showing  greater  inhibition  at 
the  same  concentration  of  ethanol. 
Further  support  for  the  involvement 
of  L-type  voltage-gated  calcium  chan- 
nels in  ethanol's  response  comes  from 
studies  showing  that  dihydropyridines 
potentiate  the  acute  pharmacological 
effects  of  ethanol,  including  ethanol- 
induced  hypothermia,  motor  incoordi- 
nation, and  sedation  (see  Leslie  et  al. 
1990  for  a  review).  Inhibition  of  L- 
type  voltage-gated  calcium  channels 
has  also  been  related  to  ethanol-induced 
decreases  in  arginine  vasopressin  (AVP) 
release  that  contribute  to  the  diuretic 
effect  of  ethanol  (Wang  et  al.  1991). 

A  variety  of  studies  suggest  a  role  for 
voltage -sensitive  calcium  channel  reg- 
ulation in  ethanol  dependence.  The  L- 
type  calcium  channel  antagonists, 
nitrendipine,  nimodipine,  and  PN200- 
110,  prevent  the  behavioral  signs  of 
ethanol  withdrawal  (handling-induced 
or  audiogenic  seizures)  (Little  et  al. 


110 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


1986;  Littleton  et  al.  1990;  Whittington 
and  Little  1991;  Watson  et  al.  1994). 
Extracellular  recordings  made  from 
mouse  hippocampal  slices  prepared  after 
chronic  treatment  with  ethanol  or 
ethanol  and  nitrendipine  suggest  that 
the  electrophysiological  changes  that 
occur  during  ethanol  withdrawal  are 
also  prevented  by  L-type  calcium 
channel  blockers  (Whittington  and 
Little  1991).  Furthermore,  Bay  K  8644, 
an  L-type  calcium  channel  activator, 
increases  withdrawal-induced  hyperex- 
citability  in  mouse  hippocampal  slices 
prepared  after  chronic  ethanol  treat- 
ment (Whittington  and  Little  1993), 
whereas  PN200-110,  an  antagonist, 
decreases  withdrawal-induced  hyper- 
excitability  (Whittington  and  Little 
1991).  Several  studies  have  indicated 
that  chronic  ethanol  treatment  increases 
dihydropyridine  binding  sites  in  mem- 
branes prepared  from  whole  brain  and 
cerebral  cortex  from  mouse  and  rats 
(Dolin  et  al.  1987;  Dolin  and  Little 
1989;  Whittington  et  al.  1991). 
Chronic  ethanol  administration  has  also 
been  shown  to  increase  the  functional 
effects  of  Bay  K  8644 — inositol  lipid 
breakdown  and  neurotransmitter  release 
(Dolin  et  al.  1987).  These  effects  on 
dihydropyridine  binding  and  function 
are  blocked  by  calcium  channel  antag- 
onists (Dolin  et  al.  1987;  Dolin  and 
Little  1989;  Whittington  et  al.  1991). 
Clearly,  regulation  of  dihydropyridine 
binding  sites  is  important  in  the  devel- 
opment of  ethanol  dependence  and  the 
manifestations  of  ethanol  withdrawal. 
Chronic  ethanol  exposure  has  been 
found  to  produce  tolerance  to  the  in 
vitro  inhibitory  effects  of  ethanol  on 
calcium  uptake  (Harris  and  Wood  1980; 


Leslie  et  al.  1983),  but  the  development 
of  tolerance  is  pronounced  in  some 
brain  areas  (e.g.,  hypothalamus)  and 
does  not  occur  in  other  areas  (e.g., 
cerebellum)  (Daniell  and  Leslie  1986). 
Thus,  the  adaptive  changes  in  voltage- 
gated  calcium  channels  may  vary  with 
the  brain  region. 

Interestingly,  striking  increases  in 
dihydropyridine  binding  are  seen  in 
heart  tissue  of  ethanol-dependent  rats 
(Guppy  and  Littleton  1994).  This  may 
be  related  to  the  cardiovascular  effects 
of  alcohol  reported  in  humans.  There 
is  considerable  evidence  that  chronic 
heavy  alcohol  consumption  is  associated 
with  cardiovascular  disease  such  as 
cardiomyopathy,  hypertension,  and 
arrhythmia  (e.g.,  Lands  and  Zakhari 
1990).  Any  or  all  of  these  afflictions 
could  be  related  to  calcium  channel 
deregulation  caused  by  alcohol  depen- 
dence. Since  different  calcium  channel 
subunits  may  be  expressed  in  distinct 
tissues  (e.g.,  the  heart  vs.  the  brain), 
the  differential  regulation  of  various  L- 
type  channel  subunits  may  have  broader 
implications  for  ethanol  pathology 
than  just  the  CNS. 

Cells  in  culture  have  been  used  as  in 
vitro  models  for  the  study  of  chronic 
ethanol  effects  on  voltage-sensitive  cal- 
cium channels.  Increases  in  L-type  cal- 
cium channel  binding  sites  have  been 
reported  in  both  adrenal  chromaffin 
cells  and  PC  12  cells  (a  clonal  line  of 
neural  crest  origin)  after  growing  cells 
in  200  mM  ethanol  for  6  days  (Messing 
et  al.  1986;  Harper  et  al.  1989;  Brennan 
and  Littleton  1990, 1991).  It  is  believed 
that  such  changes  in  calcium  channel 
binding  following  chronic  ethanol  expo- 
sure constitute  an  adaptive  response  to 


111 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol,  since  acute  alcohol  decreases 
voltage -dependent  calcium  influx  (e.g., 
Harris  and  Wood  1980;  Leslie  et  al. 
1983;  Messing  et  al.  1986).  Although 
the  biochemical  mechanisms  involved 
in  this  up-regulation  of  dihydropyridine 
binding  sites  are  as  yet  unclear,  several 
studies  implicate  the  involvement  of  a 
PKC  (Brennan  and  Littleton  1990; 
Messing  et  al.  1990,  1991).  The 
increase  in  dihydropyridine  binding  sites 
can  be  prevented  by  inhibitors  of  pro- 
tein and  mRNA  synthesis,  suggesting 
that  transcription  of  some  gene(s)  may 
be  involved  in  the  response  (Harper  et 
al.  1989).  It  has  yet  to  be  shown,  how- 
ever, that  regulation  of  the  genes  encod- 
ing calcium  channel  subunits,  per  se,  is 
a  component  of  the  adaptive  process. 
Dihydropyridines  appear  to  bind  to 
the  al  subunit  of  the  L-type  calcium 
channel.  However,  it  is  unknown  how 
changes  in  dihydropyridine  binding 
relate  to  changes  in  a  1  or  other  subunit 
protein  or  mRNA.  Also,  since  no  lig- 
ands  exist  that  specifically  bind  to  other 
L-type  channel  subunits,  it  is  unknown 
whether  other  subunits  are  changed 
following  chronic  ethanol  exposure. 

Innate  differences  in  susceptibility 
to  dependence  and  withdrawal  have 
been  demonstrated  (e.g.,  WSP  and 
WSR  mouse  strains  [Crabbe  et  al. 
1983]).  Differences  in  the  effect  of 
chronic  ethanol  treatment  on  dihy- 
dropyridine binding  sites  in  WSP  and 
WSR  mouse  strains  have  been  observed 
(Brennan  et  al.  1990),  implying  that 
these  innate  genetic  differences  in  the 
susceptibility  to  dependence  and 
withdrawal  may  be  explained,  in  part, 
by  the  differential  regulation  of  volt- 
age-sensitive calcium  channels.  Thus, 


voltage-gated  calcium  channels  likely 
contribute  to  the  acute  actions  of 
ethanol  as  well  as  the  development  of 
tolerance  and  dependence  to  ethanol. 
Innate  differences  in  these  channels 
may  contribute  to  innate  differences 
in  sensitivity.  Significantly  more 
research  will  be  required  to  under- 
stand the  role  of  various  subtypes  of 
voltage-gated  calcium  channels  in 
ethanol  responses. 

~Purinergic  P2x  I°n  Channel 
Receptors.  Adenosine  triphosphate 
(ATP)  has  been  recognized  as  an 
excitatory  transmitter  in  both  the 
CNS  and  the  peripheral  nervous  system, 
acting  through  purinergic  receptors 
that  include  G  protein-coupled  recep- 
tors (e.g.,  P2y  and  P2u  receptors)  and 
ligand-gated  ion  channel  receptors 
(e.g.,  P2X)  (Kennedy  and  Leff  1995). 
Weight's  laboratory  has  investigated 
the  effects  of  ethanol  on  ATP-gated 
channels  using  whole  cell  patch-clamp 
on  neurons  from  bullfrog  dorsal  root 
ganglion  neurons.  The  amplitude  of 
ATP-activated  currents  was  decreased 
by  ethanol  (EC50  =  68  mM).  Unlike 
NMDA  receptors  where  ethanol  is 
noncompetitive,  inhibition  of  ATP 
followed  competitive  kinetics  by 
right-shifting  the  dose-response  curve 
for  ATP  (Li  et  al.  1993).  Studies  of  a 
series  of  alcohols  indicated  that  alco- 
hol potency  correlated  with  lipid  sol- 
ubility from  one  to  three  carbons;  for 
example,  methanol  <  ethanol  <  pro- 
panol.  For  alcohols  of  four  or  more 
carbons,  no  effect  was  found.  These 
studies  suggest  that  a  hydrophobic 
pocket  exists  within  the  receptor  ion 
channel  complex  that  has  a  specific 
molecular  volume  (Li  et  al.  1994). 


112 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Additional  studies  are  needed  to 
determine  if  ethanol  alters  mammalian 
P2x  receptors  and  to  understand  the 
role  of  this  family  of  ion  channel 
receptors  in  ethanol  adaptations. 

Voltage-Gated  Potassium  Channels. 
Potassium  ion  channels  dominate  the 
resting  membrane  potentials  of  almost 
all  cell  types  and  bring  stimulated, 
depolarized  cells  back  to  their  resting 
potential.  Potassium  ion  channels  are 
diverse  and  generally  divided  into  two 
broad  categories:  delayed  rectifiers 
and  inward-rectifiers.  Osmanovic  and 
Shefner  (1987,  1994)  studied  the 
effect  of  ethanol  on  locus  coeruleus 
neuronal  inward-rectifier  currents  and 
found  that  ethanol  shifted  the  inward 
rectification  in  the  depolarizing  direc- 
tion, essentially  increasing  current. 
Ethanol-induced  hyperpolarization  of 
hippocampal  neurons  has  also  been 
suggested  to  be  due  to  increased  potas- 
sium conductance  (Carlen  et  al.  1985). 
Similarly,  single -channel  currents  stud- 
ied using  the  patch-clamp  cell-attached 
technique  found  that  ethanol  increased 
the  open  probability  of  potassium 
channels  in  human  T  cells  at  concen- 
trations of  35-50  mM  ethanol  (Oleson 
et  al.  1993).  Ethanol  selectively  inhibits 
a  voltage-dependent  potassium  current, 
known  as  the  muscarinic  cholinergic- 
responsive  M-current,  in  hippocampal 
CA1  neurons  (Madamba  et  al.  1995). 
Ethanol  had  little  or  no  effect  on  other 
K+  conductances  in  CA1  pyramidal 
cells  (Moore  et  al.  1990).  The  regula- 
tory role  of  these  channels  makes  them 
likely  to  be  sites  of  adaptation  involved 
in  the  development  of  tolerance  to  and 
dependence  on  ethanol.  Additional  stud- 
ies are  needed  to  determine  if  adaptive 


changes  in  potassium  channels  occur 
during  chronic  ethanol  treatment. 

Neurotransmitter  and 
Neuromodulator  Systems: 
Dopamine,  Serotonin,  and  Opiates 

In  the  past  two  decades,  a  series  of 
behavioral  and  pharmacological  reports 
from  several  laboratories  have  reinforced 
the  idea  that  the  nucleus  accumbens 
and  particularly  the  VTA- accumbens 
(mesolimbic)  system  may  play  a  central 
role  in  reinforcement  for,  and  depen- 
dence on,  an  assortment  of  addictive 
drugs  (Koob  and  Bloom  1988;  see  also 
chapter  7  in  this  monograph).  Because 
considerable  data  support  synaptic 
transmission  as  the  most  ethanol-sensitive 
central  site,  it  is  instructive  for  the 
purpose  of  this  review  to  evaluate  the 
types  of  transmitters  and  neuromodula- 
tors, and  their  interactions  with  ethanol, 
in  these  two  brain  regions.  There  is  an 
abundance  of  opioid  peptides  and  their 
receptors  in  both  brain  regions  (see, 
e.g.,  Herkenham  et  al.  1984;  Mansour 
et  al.  1988),  and  5-HT  and  associated 
receptors  are  also  abundant  in  these 
regions  (see,  e.g.,  Li  et  al.  1989; 
Lavoie  and  Parent  1990;  Matsuzaki  et 
al.  1993;  Van  Bockstaele  et  al.  1993), 
with  convergence  of  DA-  and  5-HT- 
containing  fibers  in  accumbens  (Phe- 
lix  and  Broderick  1995)  suggesting  an 
interaction  of  these  two  transmitters. 
Of  course,  the  major  projection  neu- 
rons from  VTA  to  accumbens  contain 
dopamine,  and  these  neurons  also 
receive  5-HT-containing  afferent 
inputs  (Van  Bockstaele  et  al.  1994).  A 
significant  number  of  ethanol  studies 
have  reported  changes  in,  or  a  significant 
role  for,  other  nucleus  accumbens 


113 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


transmitters,  such  as  GABA  (Hodge 
et  al.  1995)  and  glutamate  (Nie  et  al. 
1993;  Moghaddam  and  Bolinao  1994; 
Nie  et  al.  1994). 

The  fact  that  the  accumbens  is  an 
integral  part  of  the  extended  amygdala 
adds  more  significance  to  the  role  of  the 
nucleus  accumbens  in  alcohol-seeking 
behavior  and  suggests  the  need  to 
evaluate  the  role  of  various  known 
transmitters  in  the  amygdala  as  well. 
In  the  final  analysis,  the  multifaceted 
nature  of  alcohol  abuse  and  alcoholism 
will  likely  involve  a  complex  interplay 
or  interaction  among  the  several  neu- 
rotransmitters and  neuromodulators 
found  in  these  and  other  (e.g.,  inferior 
colHcular  cortex  [McCown  and  Breese 
1993])  brain  regions  thought  to  be 
involved  in  these  phenomena.  The 
reader  is  directed  to  the  excellent  review 
by  Weiss  in  chapter  7  for  a  more  detailed 
analysis  of  the  changes  in,  and  the  role 
for,  the  dopamine,  5-HT,  opioid,  and 
corticotropin-releasing  factor  systems 
in  the  various  aspects  of  alcohol  prefer- 
ence, reinforcement,  dependence,  crav- 
ing, and  sensitization.  The  following 
sections  present  cellular/biochemical/ 
molecular  data  obtained  from  some  of 
these  models,  but  also  briefly  highlight 
some  aspects  of  acute  ethanol  effects  as 
a  baseline  to  aid  in  understanding  the 
changes  (e.g.,  whether  tolerance  devel- 
ops) occurring  in  the  chronic  or  prefer- 
ence models,  or  as  hints  for  what  to 
examine  in  such  models  in  future  studies. 

Dopamine.  A  dopamine  link  is  seen 
in  a  large  percentage  of  literature  cita- 
tions on  neurochemical  and  behavioral 
effects  of  ethanol  (see,  e.g.,  Weiss  et 
al.  1993).  Acute  systemic  ethanol 
increases  extracellular  dopamine  levels 


in  accumbens  (see  Weiss  et  al.  1993 
and  chapter  7  in  this  monograph), 
consistent  with  early  electrophysiolog- 
ical studies  showing  enhanced  VTA 
neuron  firing  (Gessa  et  al.  1985;  Brodie 
et  al.  1990)  and  perhaps  predictive  of 
a  possible  dopamine  link  in  ethanol 
reinforcement.  Unfortunately,  charac- 
terizations of  dopamine  effects  in 
accumbens  slices  have  shown  that  the 
effects  of  this  transmitter  per  se,  and 
its  interaction  with  acute  ethanol,  are 
rather  complex  (i.e.,  both  state  and 
cell  dependent)  (Cepeda  et  al.  1993; 
Surmeier  and  Kitai  1993,  1994; 
Surmeier  et  al.  1995;  Levine  et  al. 
1996;  Surmeier  and  Kitai  1997;  Yan 
and  Surmeier  1997;  Yan  et  al.  1997). 
The  cellular  mechanisms  of  action  of 
acute  ethanol  (let  alone  chronic 
ethanol)  on  the  dopamine  system 
have  not  been  well  defined.  Nonethe- 
less, the  Henriksen  group  has  found 
an  interesting  role  for  dopamine  in 
several  actions  of  acute  ethanol  tested 
in  vivo,  with  respect  to  a  modulating 
role  in  hippocampal  function  (Criado 
et  al.  1994). 

The  importance  of  these  acute  effects 
of  ethanol  on  the  dopamine  system  is 
underscored  by  the  burgeoning  litera- 
ture on  this  system  in  various  models 
of  alcoholism.  Of  the  alcohol-preferring 
rat  genetic  models,  the  Indiana  P  and 
HAD  rat  lines  show  abnormalities  in 
forebrain  dopamine  function,  and 
there  is  an  increased  sensitivity  to  the 
dopamine-releasing  and  locomotor 
effects  of  ethanol  in  the  Indiana  lines 
as  well  as  in  the  Sardinian  alcohol-pre- 
ferring (sP)  lines  (Fadda  et  al.  1980; 
Waller  et  al.  1986;  Gongwer  et  al. 
1989;  McBride  et  al.  1990;  Weiss  et 


114 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


al.  1993).  However,  the  Finnish  AA/ 
ANA  rats  show  dopamine  changes 
opposite  to  those  in  the  Indiana  P  and 
HAD  lines  (see,  e.g.,  Sinclair  et  al. 
1989;  Kiianmaa  et  al.  1991).  In  addi- 
tion, self- administration  of  ethanol  is 
accompanied  by  dopamine  release  in 
the  accumbens  (Weiss  et  al.  1993), 
and  rats  will  self- administer  ethanol 
directly  into  the  VTA  (Gatto  et  al. 
1994).  Other  pharmacological  data  also 
show  that  ethanol  preference  or  rein- 
forcement can  be  altered  by  dopamine - 
related  drugs.  Furthermore,  in  contrast 
to  acute  ethanol,  ethanol  withdrawal 
is  accompanied  by  reduced  firing  of 
dopamine  neurons  (Chiodo  and  Berger 
1986;  Diana  et  al.  1992,  1993)  and 
reduced  dopamine  release  in  accumbens 
(Rossetti  et  al.  1992&;  Weiss  et  al. 
1996).  Interestingly,  chronic  ethanol 
treatment  also  decreases  acute  ethanol- 
evoked  dopamine  release,  suggesting 
tolerance  to  the  dopamine-releasing 
effect.  Weiss  (see  chapter  7)  proposes 
that  these  and  other  related  data  sup- 
port a  role  for  dopamine  in  accumbens 
in  continued  ethanol  abuse  and  depen- 
dence, in  relapse  after  protracted  absti- 
nence, and  perhaps  in  the  motivational 
aspects  of  these  states;  however,  a  link 
between  dopaminergic  systems  and 
ethanol  sensitization  has  been  more 
difficult  to  verify. 

The  mechanism  of  the  "tolerance" 
to  the  dopamine-releasing  ethanol  effect 
seen  after  chronic  ethanol  exposure  is 
still  under  investigation,  but  it  may 
involve  reduced  presynaptic  Ca++ 
influx  (Kim  et  al.  1994),  uncoupled 
Ca++  entry  for  dopamine  release  (Leslie 
et  al.  1986),  or  depolarization  inactiva- 
tion  of  the  dopamine  cells  (Shen  and 


Chiodo  1993).  Despite  such  evidence 
for  reduced  dopamine  release,  recent 
biochemical  studies  showing  that 
chronic  ethanol  increases  tyrosine 
hydroxylase  expression  (and  other 
measures  of  activation)  in  VTA  suggest 
that  VTA  neurons  are  actually  acti- 
vated by  such  treatment  (Ortiz  et  al. 
1995).  The  apparent  discrepancy 
between  these  findings  and  the  obser- 
vation of  reduced  numbers  of  sponta- 
neously firing  neurons  in  VTA  under 
these  conditions  (Shen  and  Chiodo 
1993)  has  not  been  clarified. 

Unfortunately,  other  than  this  one 
extracellular  in  vivo  study  by  Shen  and 
Chiodo  (1993),  few  studies  have 
investigated  the  effect  of  chronic 
ethanol  treatment  or  other  alcoholism 
models  on  the  cellular  (electrophysio- 
logical) aspects  of  dopamine  function, 
and  to  our  knowledge  no  reports  using 
intracellular  or  patch-clamp  analyses 
of  dopamine  function  in  such  models 
have  appeared.  Only  recently  has  an 
intracellular  study  of  acute  ethanol 
effects  on  VTA  neurons  appeared 
(Brodie  and  Appel  1998),  to  reveal  a 
few  mechanisms  (depolarization, 
increased  "h"  current,  reduced  spike 
afterhyperpolarization)  that  may  cause 
the  increased  VTA  firing  previously 
reported.  The  lack  of  such  studies  in 
chronic  ethanol  models  may  arise 
because  of  the  complexity  of  the  models 
and  the  dopamine  receptor  system  and 
the  often  multiple,  covert  actions  of 
dopamine  in  electrophysiological  studies 
(see,  e.g.,  Surmeier  and  Kitai  1994; 
Surmeier  et  al.  1995;  Surmeier  and 
Kitai  1997;  Yan  and  Surmeier  1997; 
Yan  et  al.  1997).  For  example,  little  is 
known  about  cellular  mechanisms  of 


115 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


possible  changes  in  dopamine  autore- 
ceptor  function  under  these  conditions. 
Furthermore,  it  should  be  emphasized 
for  future  work  that  dopamine  recep- 
tors, as  G  protein-linked  receptors, 
would  fall  into  the  generic  "metabotro- 
pic"  category  that,  according  to  one 
hypothesis  (see  the  Neurotransmitter 
Systems  section,  p.  138),  could  regu- 
late the  ongoing  sensitivity  of  ligand- 
and  voltage-gated  ion  channels  to 
ethanol  (Siggins  et  al.  1999). 

5-Hydroxytryptamine.  Serotonin  (5- 
HT)  has  also  been  strongly  implicated 
in  ethanol-seeking  behavior.  To  summa- 
rize, as  with  dopamine,  there  are  clear 
differences  in  5-HT  levels  of  the  Indi- 
ana P  and  HAD  lines  (see,  e.g.,  McBride 
et  al.  1990,  1995);  unfortunately,  no 
such  differences  have  been  seen  in  the 
Finnish  AA  alcohol -preferring  rat  lines 
(Sinclair  et  al.  1989;  Kiianmaa  et  al. 
1991).  Also,  acute  ethanol  increases  5- 
HT  release  in  accumbens  after  passive 
administration  or  self- administration 
(Yoshimoto  et  al.  1991;  Yoshimoto 
and  McBride  1992;  Weiss  et  al. 
1996).  Furthermore,  pharmacological 
manipulations  that  should  alter  5-HT 
levels  or  receptors  alter  ethanol-seeking 
behavior  in  animals  (Sellers  et  al.  1992; 
LeMarquand  et  al.  \99Aa)  and  in 
humans  (see,  e.g.,  Monti  and  Alterwain 
1991;  Naranjo  and  Bremner  1993; 
LeMarquand  et  al.  1994&),  and  drugs 
related  to  both  the  5-HT1A  and  the  5- 
HT3  receptors  alter  ethanol's  discrimi- 
native stimulus  properties  (Signs  and 
Schechter  1988;  Grant  and  Barrett  1991; 
Grant  and  Colombo  1993;  Krystal  et 
al.  1994).  The  latter  findings  are  consis- 
tent with  clinical  data  showing  that  5- 
HT  receptor-related  drugs  can  produce 


ethanol-like  feelings  (Benkelfat  et  al. 
1991;  Lee  and  Meltzer  1991;  Krystal 
et  al.  1994)  and  alter  alcohol  craving 
and  relapse  rates  in  alcoholics  (Lud- 
wig  et  al.  1974;  Modell  et  al.  1993; 
Make  et  al.  1996;  Buydens-Branchey 
etal.  1997). 

Neuroadaptation  with  chronic  ethanol 
treatment  in  animals  also  seems  to 
involve  central  5-HT  systems,  as  indi- 
cated by  the  following  findings: 

1.  Withdrawal  from  chronic  ethanol 
suppresses  5-HT  release,  levels,  and 
metabolism  in  brain  (Kahn  and 
Scudder  1976;  Tabakoff  et  al. 
1977;  Badawy  and  Evans  1983; 
Yamamura  et  al.  1992),  including 
nucleus  accumbens  (Yoshimoto 
and  McBride  1992;  Yoshimoto  et 
al.  1992).  As  with  dopamine,  this 
suppression  can  be  reversed  by 
ethanol  self- administration  (Weiss 
et  al.  1996). 

2.  Pharmacological  manipulation  of 
5-HT  receptors  (and  especially  5- 
HT1C  or  5-HT1A  receptors)  can 
alter  the  anxiogenic  effects  of  such 
withdrawal  for  up  to  a  week  (Lai  et 
al.  1993;  Rezazadeh  et  al.  1993;  see 
also  Lai  et  al.  1991;  Kleven  et  al. 
1995;  and  chapter  7).  These  findings 
have  strong  implications  for  the 
clinical  treatment  of  alcohol  depen- 
dence and  withdrawal  phenomena,  as 
discussed  later  in  this  chapter. 

Unfortunately,  despite  such  strong 
evidence  for  a  role  of  5-HT  in  ethanol- 
related  phenomena,  little  has  been  done 
to  reveal  the  molecular  or  cellular  mech- 
anisms behind  these  ethanol-induced 


116 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


changes  in  the  5-HT  system.  Acute 
ethanol  has  been  shown  to  inhibit 
responses  to  activation  of  5-HTlc 
receptors  in  an  oocyte  expression  system, 
probably  by  interfering  with  (uncou- 
pling) a  G  protein-PKC  linkage  and 
involving  PKC-mediated  receptor  phos- 
phorylation (Sanna  et  al.  1994).  In 
another  acute  study,  5-HT  and  two 
other  5-HT  receptor  agonists  potenti- 
ated the  excitatory  effects  of  acute 
ethanol  on  VTA  dopamine  neurons  in 
three  different  brain  slice  preparations 
(Brodie  et  al.  1995),  further  supporting 
the  likely  interplay  of  dopamine  and  5- 
HT  systems  in  ethanol's  central  actions. 
These  researchers  have  additionally 
explored  this  interaction  by  showing  that 
a  5-HT  uptake  inhibitor  (clomipra- 
mine, but  not  zimelidine),  such  as 
those  used  to  treat  alcoholism  clinically, 
can  markedly  enhance  the  excitatory 
effect  of  acute  ethanol  on  dopamine 
neurons  in  the  VTA  in  vitro  (Trifunovic 
and  Brodie  1996). 

Despite  these  interesting  findings 
on  the  interaction  of  acute  ethanol 
with  5-HT  receptors,  as  far  as  we  are 
aware  there  are  no  such  mechanistic 
studies  on  these  interactions  in  any 
chronic  ethanol  or  ethanol-seeking 
model.  As  with  dopamine,  this  lack 
may  be  in  part  due  to  the  complexity 
of  5-HT  neuronal  anatomy  and  5- 
HT  receptors.  And  again,  for  future 
work  on  the  interaction  between 
ethanol  and  5-HT  receptors,  it 
should  be  emphasized  that  most  5- 
HT  receptors  (all  except  5-HT3 
receptors)  fall  into  the  G  protein- 
linked  metabotropic  category  that 
may  regulate  the  sensitivity  of  ligand- 
gated  ion  channels  to  ethanol  (see  the 


Neurotransmitter  Systems  section,  p. 
138;  see  also  Siggins  et  al.  1999). 

Opiates.  Opiate  receptors  constitute 
another  group  of  such  generic  meta- 
botropic receptors.  Therefore,  it  may 
not  be  surprising  that  the  alcohol  lit- 
erature is  strewn  with  references  to 
the  opiate -like  effects  of  ethanol  (cf. 
Rossetti  et  al.  1992&),  including  early 
data  showing  cross-tolerance  between 
ethanol  and  opiates  (Mayer  et  al.  1980) 
and  showing  that  opiate  antagonists 
could  alter  ethanol  self- administration 
in  animals  (Altshuler  et  al.  1980). 
Although  the  Koob  group  and  others 
found  that  the  opiate  antagonist 
naloxone  does  block  responding  for 
ethanol  in  rats,  responding  for  water 
was  also  reduced  (Weiss  et  al.  1990). 
There  is  also  evidence  that  a  lack  of 
transsynaptic  opioid  peptides  may  be 
linked  to  a  genetically  determined 
preference  for  ethanol  consumption  in 
mice  (George  et  al.  1991),  and  a  later 
study  by  the  Koob  group  showed  that 
naloxone  could  alter  some  aspects  of 
ethanol  withdrawal  (Schulteis  et  al. 
1994).  It  has  been  suggested  that 
ethanol's  ability  to  activate  the  dopa- 
mine system  may  involve  the  opiate  sys- 
tem as  an  intermediary  (Badawy  and 
Evans  1983;  Widdowson  and  Holman 
1992;  Acquas  et  al.  1993;  Di  Chiara 
et  al.  1996;  Gonzales  and  Weiss 
1998).  Recent  clinical  data  (see  chapter 
7),  particularly  data  showing  a  benefi- 
cial therapeutic  effect  of  naltrexone  in 
reducing  craving  and  relapse  in  abstain- 
ing alcoholics  (O'Malley  et  al.  1992; 
Volpicelli  et  al.  1992;  see  also  the  section 
on  craving  later  in  this  chapter),  further 
reinforce  the  relationship  of  the  endoge- 
nous opioid  systems  to  alcohol  abuse 


117 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  alcoholism.  Interestingly,  animal 
studies  have  shown  that  both  naltrexone 
and  the  6  opiate  receptor  antagonist 
naltrindol  can  reduce  ethanol-stimulated 
dopamine  release  in  accumbens  (Acquas 
et  al.  1993;  Benjamin  et  al.  1993), 
and  a  recent  study  has  shown  that  this 
effect  of  naltrexone  correlates  directly 
with  decreases  in  ethanol  self- adminis- 
tration (Gonzales  and  Weiss  1998). 

Furthermore,  at  the  functional,  cellu- 
lar level,  there  are  often  striking  simi- 
larities between  the  electrophysiological 
actions  of  ethanol,  dopamine,  and 
opiates  in  the  nucleus  accumbens.  In 
fact,  an  early  in  vivo  extracellular  study 
found  that  condensation  products  of 
ethanol  and  dopamine  evoked  a  pattern 
of  effects  across  several  brain  regions 
very  similar  to  the  effects  elicited  by 
ethanol  and  opiates  (Siggins  et  al. 
1982).  In  hippocampus,  both  ethanol 
and  6  opiate  receptor  agonists  can  excite 
pyramidal  neurons  by  reducing  the  M- 
current,  a  voltage -sensitive  K+  conduc- 
tance (Moore  et  al.  1990,  1994;  Siggins 
et  al.  1995).  Analysis  of  several  studies 
in  nucleus  accumbens  shows  that 
ethanol,  dopamine,  and  opiates  all 
predominantly  reduce  spontaneous 
discharge  in  vivo  (Hakan  and  Henrik- 
sen  1987,  1989)  and  evoked  discharge 
in  vitro  (Uchimura  et  al.  1986;  Yuan 
et  al.  1992),  and  all  three  agents  in 
the  accumbens  slice  preparation  can 
markedly  reduce  glutamatergic  excita- 
tory postsynaptic  potential  (EPSP) 
amplitudes,  with  little  or  no  effect  on 
membrane  potential  or  resistance  (Yuan 
et  al.  1992;  Nie  et  al.  1993).  Naloxone 
significantly  reverses  ethanol-induced 
reduction  of  glutamatergic  EPSPs  in 
accumbens  core  neurons,  especially 


when  these  EPSPs  are  evoked  by  lower 
stimulus  strengths  applied  to  the  peri- 
tubercle  region  (Nie  et  al.  1993).  This 
ethanol-naloxone  interaction  does  not 
apply  to  effects  of  exogenously  applied 
NMDA  or  kainate  (Nie  et  al.  1994) 
and  thus  is  likely  to  be  a  presynaptic 
effect.  These  data  support  a  role  for 
endogenous  opioids  or  opiate  receptors 
in  ethanol  actions  in  reducing  gluta- 
mate  release  at  presynaptic  sites.  This 
mechanism  could  provide  the  physio- 
logical, cellular  underpinnings  (see 
Siggins  et  al.  1995)  for  the  reported 
efficacy  of  another  opiate  antagonist, 
naltrexone,  in  reducing  relapse  in 
abstaining  alcoholics  (O'Malley  et  al. 
1992;  Volpicelli  et  al.  1992).  However, 
it  also  should  be  noted  that  in  vivo 
studies  using  different  stimulus  sites 
(subiculum,  amygdala)  found  no 
influence  of  systemic  naloxone  on 
ethanol  inhibition  of  these  inputs 
(Siggins  et  al.  1995;  Criado  et  al. 
1997),  showing  that  there  are  sites  of 
ethanol  action  that  clearly  do  not 
involve  opioid  systems. 

As  noted  above,  recent  evidence 
suggests  that  ethanol  effects  in  certain 
mesolimbic  brain  regions  may  involve  a 
more  selective  opiate  receptor  action  on 
6  receptors  (Acquas  et  al.  1993;  see 
also  chapter  7).  It  is  therefore  of  some 
interest  that  chronic  treatment  with 
ethanol  and/or  naloxone  up -regulates 
delta  opioid  receptor  gene  expression  in 
neuroblastoma  hybrid  NG108-15  cells 
(Jenab  and  Inturrisi  1994),  an  effect 
that  appears  to  be  mediated  by  a  reduc- 
tion of  PKA  activity  (Jenab  and  Inturrisi 
1997).  To  our  knowledge,  similar 
molecular  studies  performed  in  vivo 
have  not  been  reported,  but  such  studies 


118 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


would  be  very  useful  for  comparison 
with  the  effects  of  chronic  treatment 
with  other  abused  drugs  (see,  e.g., 
J.Q.  Wang  et  al.  1994;  McGinty  and 
Wang  1998).  As  yet  there  appear  to 
be  no  cellular  studies  of  the  opioid 
systems  in  any  chronic  ethanol  or 
preference  model. 

Synaptic  Transmission; 
Presynaptic  Mechanisms 

The  foregoing  discussion  implies  that 
synaptic  transmission  would  be  highly 
affected  by  acute  and  chronic  ethanol 
exposure,  at  least  by  virtue  of  the  fact 
that  ethanol  affects  most  of  the  post- 
synaptic transmitter  receptors  discussed 
so  far.  In  fact,  the  idea  that  the  synapse 
might  be  the  most  sensitive  substrate 
for  ethanol  action  originated  in  part 
from  early  electrophysiological  findings 
showing  a  greater  ethanol  effect  in 
multisynaptic  than  monosynaptic  path- 
ways (Berry  and  Pentreath  1980). 
Subsequent  cellular  studies  on  cerebel- 
lar, hippocampal,  accumbens,  and  other 
neurons  have  helped  confirm  this  idea 
(see,  e.g.,  Carlen  et  al.  1982;  Mancillas 
et  al.  1986;  Siggins  et  al.  1987; 
Lovinger  et  al.  1990;  Lin  et  al.  1991; 
Proctor  et  al.  1992;  Nie  et  al.  1993, 
1994).  Because  synapses  are  storehouses 
of  messenger  agents,  neurochemical 
studies  have  been  able  to  elucidate 
ethanol  effects  on  transmitter  release 
and  metabolic  pathways  in  brain. 
Early  studies  showed  reproducible 
effects  of  ethanol  on  release  of  some 
transmitters.  For  example,  acetylcho- 
line release  evoked  from  brain  slices  is 
reduced  by  ethanol  (Erickson  and 
Grahm  1973),  and  the  newer  micro- 
dialysis  and  electrochemical  methods 


have  shown  increased  extracellular 
dopamine  and  5-HT  levels  in  nucleus 
accumbens  with  systemic  ethanol 
treatment  (see  chapter  7);  although 
more  difficult  to  measure,  release  of 
corticotropin-releasing  factor  has  also 
been  found. 

Much  attention  has  focused  on  the 
ionic  consequences  of  transmitter  and 
ethanol  interactions — for  example, 
biochemical  findings  that  GABA- 
induced  chloride  fluxes  are  increased 
by  ethanol  in  cultured  neurons  (Mehta 
and  Ticku  1988)  and  synaptoneuro- 
somes  (Suzdak  et  al.  1988;  Allan  et  al. 
1991).  It  has  also  been  found  that 
ethanol  reduces  calcium  influx  into 
cultured  cerebellar  granule  cells 
evoked  by  the  NMDA  glutamate 
receptors  (Hoffman  et  al.  1989£).  Still, 
electrophysiological  methods  have  been 
particularly  useful  for  finding  trans- 
mitter effects  most  sensitive  to  ethanol, 
with  the  implication  that  the  most 
sensitive  systems  are  involved  in  ethanol 
intoxication  and  perhaps  in  alcohol 
dependence  and  abuse  as  well.  The  area 
of  ethanol  effects  on  electrophysiological 
membrane  and  synaptic  properties  and 
transmitter  responses  has  been  well 
reviewed  (Deitrich  et  al.  1989;  Shefher 
1990;  Weight  1992);  therefore,  we 
will  not  provide  an  exhaustive  review 
here.  However,  it  should  be  noted  that 
the  finding  of  potent  ethanol  inhibition 
of  NMDA  receptors  (Hoffman  et  al. 
1989/*,  1989£;  Lovinger  et  al.  1989, 
1990;  White  et  al.  1990)  not  only  has 
greatly  boosted  interest  in  the  study 
of  ethanol  and  synaptic  transmission 
but  also  has  provided  an  underlying 
explanation  for  the  important  finding 
that  ethanol  reduces  LTP  (Morrisett 


119 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  Swartzwelder  1993;  Tremwel  and 
Hunter  1994;  Swartzwelder  et  al. 
1995),  the  cellular  model  of  learning 
and  memory. 

Although  electrophysiological  stud- 
ies would  seem  suited  for  examining 
ethanol  effects  on  synaptic  transmission, 
the  results  from  such  studies  have  not 
always  been  consistent  with  the  behav- 
ioral and  biochemical  findings,  as  best 
exemplified  by  GAB  A  studies.  A  well- 
known  example  of  this  problem  is  that, 
in  contrast  to  the  CI"  flux  studies,  many 
past  studies  in  hippocampal  pyramidal 
and  other  neurons  either  in  vivo 
(Mancillas  et  al.  1986)  or  in  vitro 
(e.g.,  Siggins  et  al.  1987)  have  had 
difficulty  showing  an  influence  of 
acute  ethanol  on  responses  to  exogenous 
GABA  or  evoked  GABAergic  inhibitory 
postsynaptic  potentials  (IPSPs)  (GABA- 
IPSPs).  A  more  recent  revaluation  of 
this  subject  found  that  ethanol  sensi- 
tivity of  CA1  hippocampal  and 
accumbal  IPSPs  was  conditional:  that 
is,  their  augmentation  by  acute  ethanol 
in  vitro  depended  upon  activation  of 
PKC  (Weiner  et  al.  1994,  1997)  or 
inhibition  of  GABAB  receptors  (Wan 
et  al.  1996;  Siggins  et  al.  1999).  How- 
ever, responses  to  locally  applied 
exogenous  GABA  (in  the  presence  of 
tetrodotoxin  to  minimize  presynaptic 
effects)  were  still  not  altered  by  ethanol, 
even  after  block  of  GABAB  receptors 
(Wan  et  al.  1996;  Siggins  et  al.  1999), 
suggesting  that  the  ethanol  and  GABAB 
receptor  effects  may  be  exerted  presy- 
naptically,  to  enhance  GABA  release. 
This  example  illustrates  the  critical 
need  to  closely  examine  the  synaptic 
effects  of  ethanol,  as  well  as  ethanol's 
effects  on  responses  to  exogenous 


transmitter,  for  a  complete  under- 
standing of  mechanisms  of  chronic 
ethanol  action  relevant  to  behavior  and 
clinical  phenomena. 

This  conditional  effect  of  acute  ethanol 
on  synaptic  transmission  has  also  been 
evaluated  in  accumbens  neurons,  as  a 
prelude  to  studies  of  chronic  ethanol. 
Here,  as  in  most  other  brain  regions 
(see  Siggins  et  al.  1987),  ethanol  (like 
opioid  peptides  [Yuan  et  al.  1992]) 
clearly  reduces  excitatory  glutamatergic 
transmission  evoked  by  either  local  or 
distal  stimulation  (Nie  et  al.  1993, 
1994).  Detailed  evaluation  of  the  sensi- 
tivity of  ethanol's  effects  in  this  region, 
using  pharmacological  isolation  of 
synaptic  components,  has  shown  that 
ethanol  has  both  pre-  and  postsynaptic 
inhibitory  effects  on  the  glutamatergic 
components  of  EPSPs,  with  the  former 
being  somewhat  more  potent  and 
involving  an  opiate  receptor  link  (Nie 
et  al.  1993,  1994).  Interestingly,  inhi- 
bition of  apparently  presynaptic  GABAB 
receptors  blocks  the  depressant  effect 
of  ethanol  on  the  NMDA  receptor- 
mediated  component  of  the  EPSPs 
(NMDA-EPSPs)  (Nie  et  al.  1996; 
Siggins  et  al.  1999),  by  an  as  yet 
unknown  mechanism.  Findings  such  as 
these  have  led  to  a  metabotropic  hypo- 
thesis of  ethanol  sensitivity  for  neuro- 
transmission mediated  by  ligand- gated 
ion  channels  (Siggins  et  al.  1999). 

Several  groups  have  specifically 
examined  the  effects  of  chronic 
ethanol  treatment  on  synaptic  activity. 
Little  and  colleagues  focused  on  the 
effects  of  withdrawal  from  chronic 
ethanol  on  CA1  hippocampal  function, 
using  extracellular  field  recordings  in 
a  slice  preparation.  They  showed  that 


120 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


the  hyperexcitability  (lowered  stimu- 
lus thresholds  for  population  EPSPs, 
increased  paired-pulse  facilitation, 
epileptiform  activity)  seen  in  such 
slices  is  not  mediated  by  changes  in 
IPSP  function  (Whittington  et  al. 
1992),  but  rather  by  an  up-regulation 
of  Ca++  currents  and  NMDA  receptor 
activation  (Whittington  et  al.  1995; 
Ripley  et  al.  1996).  Similar  electro- 
physiological findings  of  up-regulation 
of  NMDA-mediated  synaptic  hyperex- 
citability after  withdrawal  from  chronic 
ethanol  treatment  have  been  seen  in 
the  dentate  gyrus,  again  in  a  slice  prepa- 
ration. Here,  in  vitro  ethanol  exposure 
with  subsequent  withdrawal  was  asso- 
ciated with  an  enhancement  and  pro- 
longation of  evoked  NMDA  receptor- 
dependent  afterdischarges  (Morrisett 
1994).  Again,  these  data  were  consid- 
ered to  be  consistent  with  the  involve- 
ment of  NMDA  receptors  in  ethanol 
withdrawal  hyperexcitability.  More 
recent  studies  of  organotypic  explant 
cultures  of  the  hippocampal  CA1 
region  have  shown  that  long-term  in 
vitro  ethanol  exposure  with  subse- 
quent withdrawal  causes  a  specific 
enhancement  of  NMDA  receptor- 
mediated  synaptic  responses  preceding 
the  expression  of  frank  epileptiform 
events  (Thomas  et  al.  1998).  A  similar 
enhancement  of  NMDA  receptor  func- 
tion, leading  to  neurotoxicity  and  loss 
of  neurons,  has  been  seen  in  primary 
hippocampal  cultures  (Smothers  et  al. 
1997).  It  is  of  some  relevance  to  alcohol 
dependence  and  alcohol-seeking  behav- 
ior that  a  similar  significant  up-regulation 
of  NMDA  receptor  function  (assessed 
by  application  of  exogenous  NMDA) 
has  been  seen  in  nucleus  accumbens 


neurons  after  chronic  ethanol  treatment 
in  vivo  (via  the  vapor  chamber  method) 
and  subsequent  withdrawal  in  vitro 
(Nie  et  al.  1995). 

A  long,  elegant  series  of  studies 
highly  relevant  to  the  subject  of  neu- 
roadaptation has  come  from  the  Walker 
and  Hunter  group.  This  team  used 
extracellular  recording  in  vivo  and  in 
vitro  to  examine  the  effects  of  very 
long-term  (20-28  weeks)  exposure  to 
ethanol  via  liquid  diet,  followed  by 
long-term  withdrawal  (at  least  8-28 
weeks).  This  protocol  resulted  in  (1) 
neuronal  loss  and  synaptic  reorganiza- 
tion in  both  CA1  and  dentate  (Walker 
et  al.  1980;  Abraham  and  Hunter  1982; 
Abraham  et  al.  1982;  King  et  al. 
1988;  Orona  et  al.  1988);  (2)  reduction 
of  recurrent  paired-pulse  inhibition 
(Rogers  and  Hunter  1992);  (3)  reduc- 
tion in  muscarinic  cholinergic  function 
(Rothberg  and  Hunter  1991;  Rothberg 
et  al.  1993,  1996);  (4)  an  enhance- 
ment of  GABA  release  without  change 
in  GABAA  receptor  function  (Tremwel 
et  al.  1994£;  Peris  et  al.  1997);  and 
(5)  a  persistent  reduction  of  LTP 
(Tremwel  and  Hunter  1994).  The  find- 
ing of  item  1  may  be  conceived  of  as  a 
relatively  "permanent"  type  of  neuroad- 
aptation to  chronic  ethanol,  similar 
to  the  neurotoxicity  of  the  NMDA 
receptor-mediated  type  described  by 
Crews  and  colleagues  (Chandler  et  al. 
1997).  The  finding  of  item  5  is  of 
particular  relevance  as  a  synaptic  locus 
of  the  type  of  neuroadaptation  that 
might  be  involved  in  alcoholism  and 
could  provide  the  basis  for  the 
reduced  memory  function  seen  in 
alcoholics  and  chronically  treated 
animals.  It  was  thought  that  the 


121 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


enhancement  of  GABA  release  (item 
4)  could  underlie  the  persistent  reduc- 
tion in  LTP  (Peris  et  al.  1997);  how- 
ever, the  possible  involvement  of 
persistent  changes  in  NMDA  receptor 
structure  or  function  has  not  been 
ruled  out. 

Enhanced  GABA  release  again  sup- 
ports a  role  for  presynaptic  influences  in 
chronic  ethanol  effects.  The  mechanisms 
for  such  presynaptic  effects  (e.g., 
enhancing  GABA  release  or  reducing 
glutamate  release)  are  not  completely 
known,  although  the  several  studies 
showing  acute  ethanol  inhibition  of 
voltage-activated  Ca++  currents  in  several 
neuron  types  (e.g.,  Camacho-Nasi  and 
Treistman  1987;  Twombly  et  al.  1990; 
Jahromi  and  Carlen  1991;  Mullikin- 
Kilpatrick  and  Treistman  1994;  see 
also  the  Voltage-Sensitive  Ion  Channels 
section  earlier  in  this  chapter)  are  sug- 
gestive of  an  underlying  mechanism. 
Interestingly,  chronic  ethanol  treatment 
of  Aplysia  neurons  caused  no  toler- 
ance effect:  that  is,  Ca++  currents 
appeared  normal  and  the  inhibitory 
response  to  a  test  dose  of  ethanol  was 
not  changed  (Treistman  and  Wilson 
1991),  in  contrast  to  undifferentiated 
PC  12  cells  where  chronic  ethanol  led 
to  significantly  larger  voltage-gated 
Ca++  currents  (Grant  et  al.  1993)  and 
tolerance  to  a  standard  test  dose  of 
ethanol  (Mullikin-Kilpatrick  and 
Treistman  1994). 

The  cellular  mechanisms  behind 
the  presynaptic  effects  of  both  acute 
and  chroruc  ethanol  exposure  have 
been  addressed  by  a  series  of  elegant 
studies  on  a  model  of  transmitter 
release:  presynaptic  vasopressinergic 
nerve  endings  isolated  from  the  rat 


neurohypophysis  and  studied  by  patch- 
clamp  electrophysiological  methods 
(including  single -channel  recording) 
in  vitro  (X.  Wang  et  al.  1994;  Dopico 
et  al.  1996,  1998).  These  studies  have 
determined  that  acute  ethanol  reduces 
vasopressin  release  in  this  model  by 
acting  on  two  ionic  conductances:  (1) 
enhancing  the  open-duration  of  voltage- 
sensitive,  dihydropyridine-sensitive 
L-type  Ca++  channels,  in  a  manner  con- 
sistent with  the  interaction  of  a  single 
drug  molecule  with  a  single  target 
site,  possibly  the  L-channel  itself  (X. 
Wang  et  al.  1994);  and  (2)  enhancing 
a  Ca++-dependent  K+  conductance 
(probably  BK  channels:  mslo,  alpha 
subunit)  via  a  direct  interaction  of 
ethanol  with  the  channel  alpha  sub- 
unit  protein,  resulting  in  a  modifica- 
tion of  channel  gating  properties  to 
increased  open  state  durations  (Dopico 
et  al.  1996,  1998).  These  two  effects 
of  ethanol  have  been  correlated  with 
the  reduced  release  of  vasopressin 
after  ethanol  ingestion  in  the  intact 
animal.  More  recent  studies  of  chronic 
ethanol  effects  in  this  model  indicate 
that  rats  chronically  exposed  to 
ethanol  show  significantly  less  inhibi- 
tion of  release  from  their  terminals 
when  acutely  challenged  with  ethanol. 
The  Treistman  group  is  currently 
examining  the  acute  effects  of  ethanol 
on  the  ion  channels  in  terminals  iso- 
lated from  these  chronically  treated 
animals,  to  determine  if  they  change 
in  a  manner  that  would  explain  the 
shift  in  sensitivity  of  release  (S.  Treist- 
man, personal  communication,  April 
1998).  These  results  promise  to  pro- 
vide exciting  new  information  on  the 
molecular  and  cellular  mechanisms 


122 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


behind  the  apparent  tolerance  seen  in 
this  model. 

In  summary,  these  findings  have 
profound  implications  for  the  mecha- 
nisms underlying  the  effects  of  acute 
and  chronic  ethanol  on  presynaptic 
release  mechanisms  in  central  neurons 
in  general.  Such  mechanisms  in  turn 
may  provide  important  clues  as,  to 
critical  cellular  and  molecular  sites  of 
neuroadaptation  in  alcoholism  that 
may  lead  to  new  treatment  strategies. 

Signal  Transduction  Systems: 
Adenylyl  Cyclase.and  Protein  Kinases 

AC  and  PICA.  Cyclic  AMP  is  a  ubiq- 
uitous intracellular  second  messenger 
formed  when  a  hormone  or  neuro- 
transmitter acts  at  the  cell  surface  to 
activate  AC.  The  receptor  is  coupled  to 
AC  through  the  heterotrimeric  guanine 
nucleotide  binding  proteins  (G  pro- 
teins), which  can  be  either  stimulatory 
(Gs)  or  inhibitory  (Gi)  to  AC.  To  date, 
nine  isoforms  of  AC  have  been  identi- 
fied, and  each  has  distinct  regulatory 
characteristics  and  localization  (Sunahara 
et  al.  1996).  The  membrane -bound  AC 
signaling  system,  is  sensitive  to  acute 
perturbation  by  pharmacologically 
relevant  concentrations  of  ethanol, 
and  changes  in  this  system  after 
chronic  exposure  of  cells  or  animals  to 
ethanol  have  also  been  noted  (Hoff- 
man and  Tabakoff  1990;  Tabakoff 
and  Hoffman  1998).  As  discussed  ear- 
lier in  this  chapter,  the  AC  signaling 
system,  which  also  involves  the 
cAMP-stimulated  protein  kinase 
(PKA),  has  been  implicated  in  learn- 
ing and  memory  in  both  invertebrate 
systems  and  mammalian  brain,  and 
thus  also  represents  a  key  candidate 


for  adaptations  induced  by  alcohol  and 
other  drugs. 

In  general,  after  chronic  ethanol 
exposure,  a  decreased  response  of  AC 
to  various  stimulatory  agents 
is  observed;  this  is  the  opposite  of 
the  acute  effect  of  ethanol  to  potenti- 
ate agonist-stimulated  AC  activity 
(Hoffman  and  Tabakoff  1990; 
Tabakoff  and  Hoffman  1998).  Early 
studies  with  brain  tissue  from  mice 
and  rats  that  had  been  chronically 
treated  with  ethanol  revealed  decreased 
neurotransmitt-er-stimulated  AC  activ- 
ity, compared  with  controls  (Hoffman 
and  Tabakoff  1990;  Tabakoff  and 
Hoffman  1998).  Later  investigations 
confirmed  these  findings,  but  also 
showed  that  stimulation  of  AC  activity 
by  agents  that  acted  at  the  level  of  the 
G  protein  and/or  directly  on  the  cat- 
alytic unit  of  AC  (e.g.,  forskolin,  fluo- 
ride, Mn2+)  was  reduced  (e.g.,  Saito  et 
al.  1987;  Tabakoff  et  al.  1995).  These 
results  suggested  that  the  change  in 
AC  activity  following  chronic  ethanol 
exposure  had  the  characteristics  of 
heterologous  desensitization.  Heterol- 
ogous desensitization  is  defined  by 
the  refractoriness  of  AC  to  stimulation 
by  multiple  activators,  acting  through 
various  receptors,  following  prolonged 
exposure  of  the  system  to  a  particular 
agonist.  It  is  distinguished  from 
homologous  desensitization,  where 
only  the  response  to  the  agonist  to 
which  the  cells  were  exposed  is  reduced 
(Clark  1986). 

Decreased  agonist  and  guanine 
nucleotide-stimulated  AC  activity  has 
been  reported  in  striatal,  cortical, 
and  hippocampal  tissue  of  chron- 
ically ethanol-treated  mice  and  rats 


123 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


(Tabakoff  and  Hoffman  1979;  Saito 
et  al.  1987;  Valverius  et  al.  1989).  In 
some,  but  not  all,  studies,  a  decrease 
in  stimulation  of  cerebellar  AC  has 
also  been  reported  (Valverius  et  al. 
1989;  Wand  and  Levine  1991).  Dif- 
ferences among  studies  may  reflect 
differences  in  the  duration  of  ethanol 
exposure,  or  in  the  development  of 
tolerance  and/or  physical  dependence 
on  ethanol,  which  is  not  always  mea- 
sured. The  chronic  effect  of  ethanol 
on  AC  activity  may  also  vary  among 
cells  and  brain  areas,  due  to  differen- 
tial localization  of  various  forms  of  G 
proteins  or  isoforms  of  AC.  In  this 
regard,  it  is  important  to  note  that  the 
various  isoforms  of  AC  show  differen- 
tial sensitivity  to  the  acute  effects  of 
ethanol  (Yoshimura  and  Tabakoff 
1995),  which  could  have  an  impact 
on  adaptations  induced  by  chronic 
ethanol  exposure. 

"Desensitization"  of  AC  activity  after 
chronic  ethanol  exposure  has  also  been 
a  frequent  finding  when  activity  is 
measured  in  cultured  neuronal  (and 
nonneuronal)  cells,  although  there  are 
some  exceptions  (see  Tabakoff  and 
Hoffman  1998).  Chronic  in  vitro  expo- 
sure of  N1E-115  or  NG108-15  neu- 
roblastoma cells,  S49  lymphoma  cells, 
or  primary  cultures  of  cerebellar  granule 
neurons  to  concentrations  of  ethanol 
ranging  from  25  raM  to  200  mM  for 
several  days  resulted  in  reduced  respon- 
siveness of  AC  to  stimulation  by  various 
agonists.  Most  of  these  studies  were 
carried  out  using  membrane  prepara- 
tions of  the  cells  to  assay  AC  activity, 
but  in  PC  12  cells  the  reduced  response 
to  stimulation  was  only  observed  in  an 
assay  where  cAMP  production  in  intact 


cells  was  measured  (Rabin  1993, 
1988).  With  certain  cell  culture  prepara- 
tions and  brain  areas,  it  has  been 
reported  that  chronic  ethanol  exposure 
does  not  alter  AC  activity  (e.g.,  Charness 
et  al.  1988),  and  these  exceptions  may 
be  important  in  understanding  the 
mechanism  of  the  chronic  ethanol 
effect.  For  example,  it  would  be  of 
interest  to  compare  the  elements  of 
the  AC  system  (G  proteins,  isoforms  of 
AC)  and  the  modulators  of  this  system, 
such  as  protein  kinases  or  phosphatases, 
that  could  contribute  to  the  differing 
responses  to  chronic  ethanol  exposure. 
The  cell  culture  systems  have,  in  fact, 
been  utilized  for  investigations  of  the 
mechanism  by  which  chronic  ethanol 
exposure  leads  to  desensitization  of 
AC  activity.  In  NG108-15  cells,  a  30 
percent  decrease  in  the  amount  of 
mRNA  for  Gsa,  and  a  corresponding 
decrease  in  Gsa  protein,  was  reported 
(Mochly-Rosen  et  al.  1988).  This 
decrease  was  accompanied  by  a  decreased 
response  of  AC  to  stimulation  by  ago- 
nist (adenosine),  but  not  forskolin.  In 
these  cells,  the  mechanism  of  the 
change  induced  by  ethanol  was  sug- 
gested to  be  a  result  of  agonist-induced 
heterologous  desensitization.  Evidence 
was  presented  that  ethanol  inhibited 
adenosine  transport  into  the  NG108- 
1 5  cells  by  inhibiting  a  particular  form 
of  the  adenosine  transporter,  resulting 
in  accumulation  of  extracellular  adeno- 
sine. Prolonged  exposure  of  the  cells 
to  adenosine  was  proposed  to  result  in 
the  observed  decrease  in  Gsa  and 
agonist- stimulated  AC  activity  (Nagy 
et  al.  1989,  1990;  Krauss  et  al.  1993). 
Later  studies  indicated  that  PKA  is 
necessary  for  the  ethanol-induced 


124 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


"heterologous  desensitization"  in  S49 
cells,  supporting  the  hypothesis  that 
adenosine-stimulated  increases  in 
cAMP  led  to  the  observed  desensiti- 
zation of  the  AC  system  (Nagy  et  al. 
1991;  Coe  et  al.  1995). 

Somewhat  similar  results  were 
reported  for  experiments  with  PC  12 
cells,  where  chronic  ethanol  exposure 
resulted  in  a  decrease  in  agonist-stim- 
ulated AC  activity  and  a  decrease  in 
Gsa  (Rabin  1993).  PKA  was  also  sug- 
gested to  be  involved  in  this  desensiti- 
zation (Rabin  1993).  However,  in 
PC  12  cells  ethanol  treatment  did  not 
result  in  an  accumulation  of  extracel- 
lular adenosine  (Rabin  et  al.  1993). 
Furthermore,  in  more  recent  studies 
of  NG108-15  cells,  although  chronic 
ethanol  exposure  was  found  to  reduce 
the  level  of  Gsa  (as  well  as  Gia),  AC 
activity  was  increased,  and  these  changes 
were  reported  not  to  be  due  to  accu- 
mulation of  extracellular  adenosine 
(Williams  et  al.  1995).  Therefore,  the 
mechanism  by  which  chronic  ethanol 
treatment  produces  desensitization  of 
the  AC  system  is  still  controversial.  In 
some  cell  culture  systems,  for  example, 
decreases  in  stimulated  AC  activity 
have  been  found  to  be  accompanied  by 
increases  in  Gia,  although  this  is  by 
no  means  universal  (Charness  et  al. 
1988;  Rabin  1993;  Wand  et  al. 
1993).  In  addition,  the  relationship 
between  changes  in  G  protein  levels 
and  activity  of  AC  is  not  clear.  Increases 
in  Gia  levels  (for  example)  have  been 
reported  to  be  associated  with  both 
higher  and  lower  AC  activity  (Reithmann 
et  al.  1991).  Furthermore,  studies  of 
the  stoichiometry  of  proteins  involved 
in  regulation  of  AC  activity  suggest 


that  Gsa  is  not  the  rate -limiting  element 
in  activation  of  AC  activity.  It  has  been 
reported  that  large  reductions  (90  per- 
cent or  greater)  in  Gsa  are  necessary 
to  lower  agonist-stimulated  AC  activity 
(Milligan  1996),  much  greater  than 
the  changes  observed  with  chronic 
ethanol  exposure  of  cells. 

Nevertheless,  changes  in  G  protein 
levels  have  also  been  investigated  in 
brains  of  animals  treated  chronically 
with  ethanol,  where  desensitization  of 
AC  activity  also  occurs.  A  30  percent 
decrease  in  Gsa  was  found  in  pituitary 
tissue  of  LS  mice,  and  a  two-  to-fourfold 
increase  in  Gia  was  observed  in  cere- 
bellar tissue  of  LS  and  SS  mice,  along 
with  decreased  agonist-stimulated  AC 
activity  (Wand  and  Levine  1991;  Wand 
et  al.  1993).  However,  there  was  no 
significant  change  in  the  content  of  a 
number  of  G  protein  subunits  in  various 
brain  areas  of  C57BL/6  mice  treated 
chronically  with  ethanol  using  a  regi- 
men known  to  produce  alcohol  tolerance 
and  physical  dependence,  although 
stimulation  of  AC  by  various  receptor 
agonists,  guanine  nucleotides,  and 
forskolin  was  reduced  (Tabakoff  et  al. 
1995).  A  similar  lack  of  change  in  G 
protein  subunits  in  several  brain  areas 
was  reported  for  rats  treated  chronically 
with  ethanol  using  a  paradigm  that  gen- 
erates tolerance  and  physical  depen- 
dence (Pellegrino  et  al.  1993). 

The  overall  conclusion  that  can  be 
drawn  from  the  literature  is  that  changes 
in  the  total  quantity  of  G  proteins 
cannot  well  account  for  the  desensiti- 
zation of  AC  activity  that  is  produced 
by  chronic  ethanol  exposure.  It  has 
been  argued  that  the  heterogeneity  of 
brain  tissue  preparations  precludes  an 


125 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


accurate  determination  of  the  relation- 
ship of  G  protein  levels  to  AC  activity 
in  particular  cells;  however,  even  in 
more  homogeneous  cell  culture 
preparations,  the  evidence  linking 
changes  in  total  G  protein  content  to 
the  decreased  AC  activity  is  not 
strong.  It  is  still  possible  that  changes 
in  G  protein  levels  do  play  a  role  in 
ethanol-induced  AC  desensitization, 
however.  There  is  evidence,  for  example, 
that  G  proteins  can  interact  with  tubu- 
lin, and  that  microtubule-disrupting 
agents  can  increase  stimulated  AC  activ- 
ity, suggesting  enhanced  accessibility 
of  Gsa  to  AC  (Popova  et  al.  1994; 
Yan  et  al.  1996).  It  has  also  been  sug- 
gested that  the  level  of  Gsa-AC  com- 
plexes is  considerably  lower  than  the 
total  amount  of  Gsa  in  the  cell  (Milligan 
1996).  Thus,  if  ethanol  treatment  dis- 
rupted the  cellular  cytoskeleton,  result- 
ing in  changes  in  accessibility  of  G 
protein  subunits  to  AC  and/or  in  the 
number  of  Gsa-AC  complexes,  alter- 
ations in  AC  activity  might  be  observed 
in  the  absence  of  changes  in  the  total 
content  of  G  proteins. 

Another  possible  basis  for  the 
desensitization  of  AC  activity  after 
chronic  ethanol  treatment  is  a  quanti- 
tative or  qualitative  change  in  the  cat- 
alytic unit  of  AC.  In  some  instances, 
changes  in  stimulation  of  AC  by  Mn2+ 
and  forskolin,  both  of  which  can 
interact  directly  with  the  AC  catalytic 
unit,  have  been  observed  in  brain  tis- 
sue of  animals  treated  chronically  with 
ethanol  (Wand  et  al.  1993;  Tabakoff 
et  al.  1995).  Chronic  morphine  treat- 
ment of  rats  has  been  reported  to  alter 
the  expression  of  a  particular  isoform 
of  AC  in  the  brain  (Matsuoka  et  al. 


1994),  but  this  possibility  has  not  been 
addressed  in  animals  or  cells  treated 
chronically  with  ethanol. 

Although  the  molecular  mechanism 
of  the  chronic  ethanol-induced  change 
in  AC  activity  and  cAMP  production 
is  not  yet  clear,  there  is  evidence  for  a 
role  of  this  system  in  the  development 
of  alcohol  tolerance.  In  mice,  depletion 
of  brain  norepinephrine  by  treatment 
with  6-hydroxydopamine  (6-OHDA) 
prevents  the  development  of  functional 
tolerance  to  ethanol  (Tabakoff  and 
Ritzmann  1977).  However,  when  the 
lesioned  mice  were  treated  repeatedly 
with  forskolin  during  chronic  ethanol 
exposure,  tolerance  developed  normally 
(Szabo  et  al.  1988#).  These  findings 
can  be  interpreted  to  indicate  that 
ethanol  potentiation  of  norepinephrine - 
mediated  increases  in  cAMP  production 
may  be  necessary  for  tolerance  to 
develop.  Chronic  activation  of  AC  by 
ethanol  in  the  presence  of  an  agonist, 
or  chronic  exposure  to  forskolin,  may 
be  necessary  to  produce  the  desensiti- 
zation of  the  AC  system  that  is  associ- 
ated with  ethanol  tolerance.  This 
possibility  is  supported  by  a  study 
showing  that,  while  acute  ethanol 
treatment  of  rats  results  in  an  increase 
in  PKA  activity  and  an  increase  in  the 
phosphorylated  form  of  the  CREB 
protein  in  brain,  these  increases  are  no 
longer  observed  in  animals  that  have 
been  chronically  treated  with  ethanol 
(Yang  et  al.  1996£,  1998).  This 
change  could  result  from  a  reduction 
of  PKA  activity,  due  to  desensitization 
of  the  AC  system,  in  cells  exposed 
chronically  to  ethanol.  However, 
decreased  PKA  activity  has  been  sug- 
gested to  occur,  at  least  in  part,  from 


126 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


chronic  ethanol-induced  translocation 
of  the  catalytic  unit  of  PKA  to  the 
nucleus  (Dohrman  et  al.  1996),  which 
might  be  expected  to  enhance  CREB 
phosphorylation.  It  is  difficult  to  com- 
pare the  studies  of  CREB  phosphory- 
lation in  animals  with  the  investigations 
of  PKA  in  cultured  cells,  and  measures 
of  PKA  activity  and  translocation  in 
brains  of  alcohol-tolerant  or  -dependent 
animals  would  be  of  interest.  In  partic- 
ular, the  studies  of  Palmer  and  colleagues 
have  shown  that  ethanol  potentiation 
of  GABAA  receptor-mediated  responses 
in  cerebellar  Purkinje  cells  is  greatly 
enhanced  in  the  presence  of  a  |3- 
adrenergic  agonist  (Lin  et  al.  1993) 
and  that  this  enhancement  of  the  effect 
of  ethanol  involves  the  cAMP/PKA 
signal  transduction  system  (Freund 
and  Palmer  1997).  Desensitization  of 
the  AC/PKA  system  would  be  expected 
to  reduce  the  ability  of  ethanol  to 
potentiate  the  effect  of  GABA  at  the 
GABAA  receptor  on  Purkinje  cells 
("tolerance"  to  ethanol).  Since  ethanol 
enhancement  of  GABA  responses  in 
Purkinje  cells  has  been  related  to  the 
hypnotic  effect  of  ethanol  (e.g., 
Sorensen  et  al.  1980),  one  might 
speculate  that  the  desensitization  of 
the  AC  system  would  play  a  role  in 
tolerance  to  this  effect  of  ethanol.  In 
this  model,  the  extrinsic  system  (nor- 
epinephrine-stimulated  AC  activity) 
would  impinge  on  the  intrinsic  system 
(GABAA  receptors)  to  produce  toler- 
ance to  a  behavioral  effect  of  ethanol. 
PKC.  There  is  also  some  evidence 
for  changes  in  PKC  following  chronic 
ethanol  exposure  in  cells  of  cultures. 
The  levels  of  two  novel  PKC  isoforms, 
6  and  e,  are  increased  in  PC  12  cells 


and  NG108-15  cells  after  chronic 
ethanol  treatment  (Diamond  and 
Gordon  1997).  The  change  in  PKC- 8 
has  been  found  to  mediate  the  effect 
of  chronic  ethanol  treatment  to 
increase  neurite  outgrowth  in  PC  12 
cells  (Messing  et  al.  1991;  Hundle  et 
al.  1995;  Roivainen  et  al.  1995). 

Changes  in  Signal  Transduction 
and  Neuroadaptation.  These  studies 
raise  an  important  question  regarding 
the  applicability  of  investigations 
using  cultured  neurons  to  adaptation 
to  ethanol  in  the  intact  animal.  In  a 
number  of  instances,  "tolerance"  to 
the  effect  of  ethanol  can  be  observed 
in  cultured  cells.  For  example,  ethanol 
inhibition  of  the  adenosine  transporter 
in  NG108-15  cells  is  reduced  in  cells 
that  have  been  exposed  chronically  to 
ethanol,  and  this  change  has  been 
cited  as  an  example  of  tolerance  to 
ethanol  at  the  cellular  level  (Nagy  et 
al.  1990;  Diamond  and  Gordon  1997). 
However,  in  the  absence  of  behavioral 
measures  of  tolerance  in  the  intact 
organism,  one  cannot  determine  the 
relationship  between  such  cellular 
resistance  to  the  effect  of  ethanol  on  a 
biochemical  system  and  functional  tol- 
erance (or  physical  dependence)  in  the 
animal.  Similarly,  changes  in  neurite 
outgrowth  can  be  suggested  to  be 
related  to  synaptic  plasticity  and  remod- 
eling of  synapses,  but  this  hypothesis 
cannot  be  tested  in  the  absence  of  mea- 
surable behavioral  changes.  Although 
cell  culture  models  provide  simpler 
systems  to  study  adaptation  to  ethanol, 
the  results  of  these  studies  must  be 
integrated  with  behavioral  or  physio- 
logical responses,  as  has  been  done  in 
the  studies  of  learning  and  memory 


127 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


described  earlier  in  this  chapter,  in 
order  to  relate  adaptation  at  die  mole- 
cular level  to  adaptations  in  ethanol- 
affected  behaviors. 

IEG/Transcription  Factor 
Expression  and  Expression 
of  Chaperone  Proteins 

IEGs.  Immediate  early  genes  such  as  c- 
fosarc  a  class  of  genes  whose  expression 
is  rapidly  and  transiently  stimulated  in 
response  to  a  wide  variety  of  extracellular 
factors.  These  genes  encode  transcrip- 
tion factors  that  regulate  the  expression 
of  other  genes  ("late  genes"),  and 
these  factors  were  originally  thought 
to  be  involved  in  growth  and  differen- 
tiation. However,  the  induction  of  IEGs 
by  neurotransmitters  in  adult  brain  led 
to  the  suggestion  that  these  genes  may 
also  play  a  role  in  neuronal  plasticity, 
that  is,  in  the  processes  of  adaptation 
such  as  memory  (Robertson  1992).  It 
has  been  hypothesized  that  the  same  or 
similar  molecular  events  that  regulate 
growth  and  development  may  also 
regulate  long-term  synaptic  changes  in 
the  adult  brain  (Robertson  1992).  The 
IEGs  may  in  this  sense  represent  third 
messengers  that  mediate  communication 
between  neurotransmitters  acting  at 
cell  surface  receptors  and  gene  expres- 
sion leading  to  long-term  changes  in 
neuronal  function. 

Most  studies  of  changes  in  IEG 
expression  after  chronic  ethanol  expo- 
sure have  investigated  genes  of  the  fos 
and  jun  families.  Protein  products  of 
these  two  genes  can  interact  to  form 
heterodimeric  transcription  factor 
complexes.  For  example,  the  c-Fos  and 
c-Jun  proteins  dimerize  to  form  a 
transcription  factor  that  binds  to  the 


AP-1  consensus  sequence  on  DNA 
(Curran  and  Franza  1988).  There  are 
a  large  number  of  Fos  and  Jun  family 
proteins,  and  different  homo-  and 
heterodimers  can  influence  transcrip- 
tion in  different  ways  (Robertson 
1992).  It  is  not  yet  clear  which  genes 
are  regulated  by  AP-1,  although  there 
is  some  evidence  that  this  transcrip- 
tion factor  can  influence  the  expres- 
sion of  proenkephalin  and  nerve 
growth  factor  (NGF)  (Sonnenberg  et 
al.  1989;  Hengerer  et  al.  1990). 

The  expression  of  c-fos  was  found 
to  be  induced  in  brains  (hippocampus, 
cortex,  and  cerebellum)  of  C57BL/6 
mice  that  displayed  withdrawal 
seizures  following  the  induction  of 
physical  dependence  on  ethanol  by 
ingestion  of  a  liquid  diet  containing 
ethanol  for  7  days  (Dave  et  al.  1990). 
In  these  mice,  no  increase  in  c-fos 
mRNA  was  seen  if  the  mice  did  not 
undergo  ethanol  withdrawal  seizures. 
A  later  study  of  IEG  expression  in  rat 
brain  showed  increases  not  only  in 
expression  of  c-fos  but  also  of  c-jun 
and  another  IEG,  zif/268  (also  called 
Egr-T),  during  the  period  that  overt 
withdrawal  signs  were  evident  follow- 
ing cessation  of  chronic  exposure  to 
ethanol  by  vapor  inhalation  (Mat- 
sumoto  et  al.  1993).  As  in  the  first 
study,  changes  in  c-fos  expression  in 
the  hippocampus  were  observed  only 
in  rats  that  demonstrated  withdrawal 
seizures,  although  the  other  IEGs 
were  induced  even  in  the  absence  of 
such  convulsions.  In  a  third  study, 
increases  in  c-fos  mRNA  were  observed 
in  the  dentate  gyrus  and  piriform  cortex 
of  rats  undergoing  withdrawal  after 
cessation  of  7  days  of  exposure  to 


128 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


ethanol  by  vapor  inhalation.  The 
increase  in  c-fos  mRNA  peaked  at  8 
hours  after  withdrawal.  Interestingly, 
this  increase  could  be  prevented  by 
administration  of  an  NMDA  receptor 
antagonist,  suggesting  that  the  up- 
regulation  of  NMDA  receptors  observed 
during  ethanol  withdrawal  contributes 
to  the  increased  c-fos  expression. 
However,  in  this  study,  increased  c-fos 
mRNA  was  observed  regardless  of 
whether  the  animals  displayed  with- 
drawal convulsions  (Morgan  et  al. 
1992).  Knapp  and  colleagues  (1995) 
also  reported  an  increase  in  Fos-like 
immunoreactivity  in  several  brain 
regions  of  ethanol-withdrawn  rats,  in 
the  absence  of  withdrawal  seizures. 

The  functional  significance  of  the 
changes  in  IEG  expression  was  con- 
firmed by  the  finding  of  an  increase  in 
DNA  binding  activities  of  AP-1  and  Egr 
proteins  in  brains  of  ethanol-withdrawn 
rats  (Beckmann  et  al.  1997).  This 
increased  DNA  binding  was  observed 
at  16  hours  after  ethanol  withdrawal 
(i.e.,  when  withdrawal  signs  were  evi- 
dent). It  was  suggested  that  the  increase 
in  IEG  expression  and  transcription 
factor  binding  might  be  related  to 
long-term  neuroadaptive  changes 
associated  with  ethanol  physical 
dependence  and/or  withdrawal.  One 
specific  possibility  derives  from  the 
finding  that  ethanol  withdrawal 
seizures  become  more  severe  upon 
repeated  episodes  of  chronic  ethanol 
exposure  and  withdrawal.  This  change 
has  been  likened  to  kindling  of 
seizures  (Ballenger  and  Post  1978; 
Becker  and  Hale  1993),  which  is  also 
associated  with  changes  in  IEG 
expression  (Dragunow  and  Robertson 


1987).  However,  the  hypothesis  that 
changes  in  expression  of  transcription 
factors  play  a  role  in  neuronal  plasticity, 
possibly  in  generating  the  structural 
changes  in  synaptic  connections  asso- 
ciated with  long-term  adaptations,  is 
consistent  with  the  IEGs  playing  a 
role  in  the  broader  aspects  of  adapta- 
tion to  ethanol. 

There  have  also  been  studies  of  the 
chronic  effects  of  ethanol  on  IEG 
expression  in  cultured  cells.  In  SH- 
SY5Y  neuroblastoma  cells,  ethanol 
exposure  for  2-A  days  resulted  in  an 
increase  in  mRNA  for  c-jun  and  junD 
and  an  increase  in  AP- 1  binding  activity 
(Ding  et  al.  1996).  Chronic  ethanol 
exposure  (50  mM,  3  days)  also  selec- 
tively enhanced  NMDA-induced  AP-1 
transcription  factor  binding  activity  in 
primary  cultures  of  rat  cerebellar  gran- 
ule cells  (Cebers  et  al.  1996).  The  pro- 
tein composition  of  the  AP- 1  complex 
was  not  altered  by  ethanol  exposure. 
Although  these  findings  are  somewhat 
similar  to  those  in  animal  brain,  it  is  not 
clear  whether  these  particular  changes 
would  be  related  to  neuroadaptation 
to  ethanol  in  the  whole  animal. 

Chaperone  Protein  Expression. 
Another  mechanism  of  ethanol  adap- 
tation may  involve  the  induction  of 
chaperone  protein  transcription  (Miles 
et  al.  1991,  1994;  Hsieh  et  al.  1996). 
At  pharmacologically  relevant  concen- 
trations, ethanol  induces  Hsc70  mRNA 
and  protein  expression  in  NG108-15 
neuroblastoma  x  glioma  cells  (Miles  et 
al.  1991).  In  addition,  two  other  mol- 
ecular chaperones,  GRP94  and  GRP78, 
are  ethanol-responsive  genes  that  are 
induced  more  than  threefold  by 
ethanol  exposure  (Miles  et  al.  1994). 


129 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


These  genes  encode  proteins  that  could 
serve  protein -trafficking  roles,  produc- 
ing widespread  changes  in  cellular 
membrane  functioning.  Chaperone 
proteins  are  required  for  the  assembly 
of  multimeric  ion  channels  (Haas 
1994)  and  may  therefore  play  a  role  in 
adaptations  of  membrane  receptors  that 
are  sensitive  to  ethanol.  The  relation- 
ship of  alterations  in  the  expression  of 
these  genes  to  tolerance  and  depen- 
dence is  completely  unknown. 

Systems  That  Influence 
the  Development, 
Maintenance,  and  Loss 
of  Tolerance,  Dependence, 
and  Sensitization 

Tolerance 

In  the  studies  described  thus  far,  ani- 
mals or  cells  have  been  treated  chroni- 
cally with  ethanol,  and  changes  in 
various  neurochemical  systems  have 
been  investigated.  As  has  been 
pointed  out,  however,  changes  in 
neuronal  function  in  one  area  of  brain 
will  have  many  downstream  effects, 
such  that  it  becomes  very  difficult  to 
determine  whether  an  observed 
change  is  really  an  underlying  mecha- 
nism for  neuroadaptation  to  alcohol 
(Kalant  1998).  Another  strategy  to 
investigate  the  cellular/molecular 
basis  for  neuroadaptation  is  to  alter 
the  activity  of  a  neural  system  in  a  spe- 
cific manner  and  then  evaluate  the 
effects  of  the  alteration  on  neuroadap- 
tation. This  approach  has  been  partic- 
ularly effective  for  investigations  of 
ethanol  tolerance.  The  paradigm  bor- 
rowed from  studies  of  learning  and 
memory — the  definition  of  intrinsic 


and  extrinsic  systems — is  useful  in  dis- 
cussing the  systems  that  modify  the 
development,  expression,  and  mainte- 
nance of  ethanol  tolerance  (extrinsic 
systems).  The  systems  described  in  the 
following  sections  represent  such 
extrinsic  systems;  one  characteristic  of 
these  systems  is  that  they  affect  toler- 
ance to  a  number  of  behavioral  effects 
of  ethanol. 

Neurotransmitters.  In  mice,  the 
development  (but  not  expression)  of 
functional  tolerance  to  the  hypnotic 
and  hypothermic  effects  of  ethanol 
was  blocked  by  partial  destruction  of 
noradrenergic  neurons  with  6-OHDA 
before  the  ingestion  of  ethanol  by  the 
animals  (Tabakoff  and  Ritzmann 
1977).  As  already  discussed,  stimulation 
of  AC  activity  by  norepinephrine  in 
the  presence  of  ethanol  seems  to  be 
important  for  tolerance  development 
(Szabo  et  al.  1988a).  The  noradrenergic 
system  in  the  rat  brain,  on  the  other 
hand,  does  not  appear  to  play  a  primary 
role  in  tolerance  development,  since 
6-OHDA  lesions  did  not  block  the 
development  of  tolerance  to  the  hyp- 
notic effect  of  ethanol,  although  this 
tolerance  could  be  blocked  by  treatment 
of  rats  with  the  noradrenergic  toxin 
DSP-4  (Le  et  al.  1981a;  Trzaskowska 
et  al.  1986).  Development  of  tolerance 
in  the  rat  was  also  reported  to  be 
blocked  by  combined  destruction  of 
noradrenergic  and  serotonergic  systems 
in  brain  (Le  et  al.  198 la).  Depletion 
of  serotonin  alone  delayed  the  develop- 
ment of  chronic  tolerance  to  the  motor- 
impairing  and  hypothermic  effects  of 
ethanol  in  rats  (Le  et  al.  1981  £),  and 
also  delayed  the  development  of  acute 
ethanol  tolerance  (Campanelli  et  al. 


130 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


1988).  It  was  shown  in  these  studies 
that  a  specific  lesion  of  the  serotonergic 
pathway  connecting  the  raphe  nucleus 
and  the  forebrain  was  important  in 
modulating  tolerance  development. 
The  development  of  rapid  tolerance  in 
rats  also  appears  to  involve  the  sero- 
tonergic system,  since  rapid  tolerance 
development  is  facilitated  if  mice  are 
treated  with  the  serotonin  precursor, 
tryptophan  (Khanna  et  al.  1994).  In 
mice,  the  development  of  environment- 
dependent  tolerance  to  ethanol  was  also 
shown  to  be  slowed  by  lesions  of  the 
serotonin  system  (Melchior  and 
Tabakoff  1981).  These  studies  suggest 
that  both  the  catecholaminergic  and 
serotonergic  systems,  and  possibly 
interactions  between  these  systems,  are 
important  for  the  normal  development 
of  tolerance  to  several  effects  of  ethanol. 
An  important  conclusion  from  these 
studies  is  that  the  presence  of  ethanol 
in  the  brain  is  a  necessary,  but  not  suf- 
ficient, condition  for  the  development 
of  tolerance;  concomitant  activity  of 
certain  neurochemical  pathways, 
including  postsynaptic  effects  on  the 
AC  system,  is  also  required. 

The  key  role  of  the  NMDA  receptor 
in  the  development  of  LTP  (Collingridge 
and  Lester  1989) — that  is,  its  putative 
role  in  learning  and  memory  processes — 
led  investigators  to  investigate  whether 
NMDA  receptor  activity  also  plays  a 
role  in  ethanol  tolerance.  NMDA  receptor 
antagonists,  including  ketamine  and 
dizocilpine  (MK-801),  were  reported 
to  prevent  the  development  of  rapid 
and  chronic  tolerance  to  the  motor- 
impairing  and  hypothermic  effects  of 
ethanol  (Khanna  et  al.  1992;  Wu  et 
al.  1993).  Because,  acutely,  ethanol  is 


a  potent  inhibitor  of  NMDA  receptor 
function,  it  seems  paradoxical  that 
NMDA  receptor  antagonists  should 
block  the  development  of  tolerance  to 
alcohol.  However,  it  is  important  to 
note  that  when  the  effects  of  dizocilpine 
on  environment-dependent  ethanol 
tolerance  (produced  by  ethanol  injec- 
tions) and  environment-independent 
ethanol  tolerance  (produced  by  liquid 
diet  ingestion)  were  directly  compared, 
dizocilpine  only  blocked  the  development 
of  environment-dependent  tolerance 
to  the  hypothermic  and  ^coordinating 
effects  of  ethanol.  The  same  or  higher 
doses  of  dizocilpine  did  not  block  the 
acquisition  of  environment-independent 
tolerance  to  the  hypothermic,  incoor- 
dinating,  or  hypnotic  effects  of  ethanol 
(Szabo  et  al.  1994).  The  explanation 
for  this  difference  was  postulated  to  be 
the  different  contributions  of  learning 
or  conditioning  to  the  two  forms  of 
tolerance.  Thus,  it  was  suggested  that 
dizocilpine  did  not  block  the  develop- 
ment of  ethanol  tolerance  per  se,  but 
blocked  learning  or  conditioning  that 
is  necessary  for  the  development  of  envi- 
ronment-dependent tolerance.  Simi- 
larly, dizocilpine  had  a  much  greater 
effect  on  the  development  of  behav- 
iorally  augmented  tolerance  than  on 
tolerance  that  did  not  involve  intoxi- 
cated practice  (learning)  (Khanna  et 
al.  1994). 

Neuropeptides:  AVP  and  Neuro- 
trophins.  Arginine  vasopressin  is  a 
mammalian  antidiuretic  hormone  that 
is  synthesized  primarily  in  the  hypo- 
thalamus, as  well  as  in  some  extrahy- 
pothalamic  brain  areas.  The  studies  of  de 
Wied  and  colleagues  (1997)  demon- 
strated that  AVP  could  influence  learning 


131 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  memory  and  led  to  investigations 
of  the  effect  of  the  peptide  on  tolerance. 
In  both  mice  and  rats,  the  administra- 
tion of  AVP  has  been  shown  to  main- 
tain (reduce  the  rate  of  loss  of) 
functional  tolerance  to  a  number  of  dif- 
ferent effects  of  ethanol  (hypnosis, 
hypothermia,  incoordination)  (Hoffman 
1994).  The  action  of  AVP  on  tolerance 
was  shown  to  be  mediated  by  CNS 
receptors,  since  analogs  without  periph- 
eral activity  could  maintain  tolerance. 
Furthermore,  administration  of  AVP 
intracerebroventricularly  (icv),  at  doses 
with  no  discernible  peripheral  effect, 
also  maintained  tolerance  (Hung  et  al. 
1984).  The  action  of  vasopressin  on 
alcohol  tolerance  is  mediated  by  the 
Vx  subtype  of  vasopressin  receptor,  as 
determined  by  studies  with  selective 
agonists  and  antagonists  (Szabo  et  al. 
1988^).  These  studies  also  showed 
that  a  Vi  receptor  antagonist  could 
increase  the  rate  of  loss  of  tolerance, 
indicating  a  role  of  the  endogenous 
peptide  in  maintenance  of  tolerance. 

It  is  important  to  note  that  vasopressin 
does  not  appear  to  facilitate  the  induc- 
tion of  chronic  tolerance  to  ethanol, 
but  in  fact  was  found  to  retard  the  devel- 
opment of  tolerance  when  given  in  con- 
junction with  chronic  ethanol  treatment 
(Mannix  et  al.  1986).  On  the  other 
hand,  it  has  been  reported  that  a  single 
dose  of  AVP,  given  together  with  a 
low  dose  of  ethanol,  resulted  in  the 
production  of  long-lasting  tolerance 
to  the  motor-incoordinating  effect  of 
ethanol,  whereas  the  same  dose  of 
ethanol  alone  did  not  produce  tolerance. 
These  findings  were  taken  to  indicate 
a  role  for  AVP  in  facilitating  the 
acquisition  of  acute  tolerance  to  ethanol 


(Wu  et  al.  1996).  Such  disparate  findings 
could  reflect  differences  in  the  mecha- 
nisms underlying  acute  and  chronic 
tolerance  or  in  the  role  of  learning  and 
memory  in  the  two  forms  of  tolerance. 

The  neurochemical  and  molecular 
actions  of  AVP  that  may  influence  tol- 
erance may  be  pre-  or  postsynaptic.  In  mice, 
depletion  of  norepinephrine  after  toler- 
ance had  developed  did  not  block  the 
expression  of  tolerance  to  the  hyp- 
notic effect  of  ethanol,  but  did  block 
the  ability  of  AVP  to  maintain  toler- 
ance (Hoffman  et  al.  1983).  It  was 
also  shown  that  the  6-OHDA  lesions 
used  to  deplete  norepinephrine  reduced 
the  number  of  vasopressin  receptors  in 
the  lateral  septum,  an  area  of  high  con- 
centration of  Vl  receptors  (Ishizawa  et 
al.  1990).  These  data  suggested  that  a 
portion  of  the  vasopressin  receptors 
involved  in  maintaining  ethanol  toler- 
ance may  be  localized  to  the  terminals 
of  catecholaminergic  neurons  in  mouse 
brain,  and  that  vasopressin's  actions 
could  be  at  least  partially  explained  by 
an  effect  of  the  peptide  on  neurotrans- 
mitter release.  In  the  rat,  serotonergic 
systems  have  been  reported  to  play  a 
key  role  in  the  action  of  AVP  on 
ethanol  tolerance.  The  peptide  could 
no  longer  maintain  tolerance  in  animals 
in  which  the  forebrain  serotonin  ter- 
minals had  been  destroyed  (Speisky  and 
Kalant  1985).  In  such  animals,  infusion 
of  a  5-HT2  receptor  agonist  restored 
the  ability  of  AVP  to  maintain  toler- 
ance (Wu  et  al.  1996). 

Kalant  has  postulated,  and  pro- 
vided evidence  for,  a  circuit  involving 
serotonin-,  glutamate-,  vasopressin-,  and 
GABA-containing  neurons  in  the  septum 
and  hippocampus,  and  inputs  to  these 


132 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


regions,  that  are  required  for  the 
development  and  retention  of  ethanol 
tolerance  in  the  rat  (Kalant  1998). 
Slightly  different  pathways  may  be 
involved  in  the  mouse,  where  cate- 
cholaminergic  systems  play  a  more  pri- 
mary role.  In  these  pathways,  both  pre- 
and  postsynaptic  effects  of  vasopressin 
may  be  important  for  maintenance  of 
tolerance.  When  AVP  was  administered 
icv  to  mice,  it  was  found  to  increase  the 
mRNA  levels  for  the  IEG  c-fos  in  the 
lateral  septum,  and  later  studies  showed 
that  AVP  administration  also  led  to  an 
increase  in  Fos  protein  (Giri  et  al.  1990; 
Andreae  and  Herbert  1993).  The  effect 
of  AVP  on  c-fos  expression  was  medi- 
ated by  Vj  receptors,  and  a  positive 
correlation  was  found  between  the  abil- 
ity of  several  AVP-related  peptides  to 
maintain  tolerance  and  to  increase  c- 
fos  expression  in  the  septum  (Giri  et 
al.  1990;  Szabo  et  al.  1991).  Further- 
more, icv  administration  of  an  antisense 
oligonucleotide  to  c-fos  blocked  the 
ability  of  vasopressin  to  increase  expres- 
sion of  this  IEG  in  the  septum  and 
also  blocked  the  ability  of  AVP  to 
maintain  tolerance  (Szabo  et  al.  1996). 
Fos  can  form  transcription  factors  that 
may  generate  long-term  changes  in 
CNS  function  in  response  to  neuro- 
transmitters or  neuropeptides  that  act 
at  the  cell  surface.  In  particular,  Fos 
family  proteins  can  dimerize  with  pro- 
teins of  the  Jun  family  to  form  the 
transcription  factor  AP-1.  The  genes 
whose  expression  is  affected  by  AP-1 
have  not  been  identified,  although 
NGF  has  been  suggested  to  be  regu- 
lated by  this  transcription  factor. 

To  determine  whether  AVP  might 
be  acting  by  inducing  the  expression  of 


NGF  or  other  neurotrophins,  the  ability 
of  these  agents  to  maintain  ethanol  tol- 
erance in  mice  was  tested.  It  was  found 
that,  whereas  NGF  was  relatively  inef- 
fective in  maintaining  tolerance  to  the 
hypnotic  effect  of  ethanol  (Szabo  et  al. 
1991),  brain-derived  neurotrophic 
factor  (BDNF),  as  well  as  neurotrophins 
3  and  4/5,  could  maintain  tolerance 
(Szabo  and  Hoffman  1995).  It  has 
been  reported  that  a  vasopressin  analog 
that  can  maintain  ethanol  tolerance  can 
also  induce  the  expression  of  BDNF 
in  rat  brain  (Zhou  et  al.  1997).  Whether 
or  not  the  effect  of  vasopressin  on  tol- 
erance is  mediated  through  neurotro- 
phins, the  ability  of  neurotrophins  to 
maintain  tolerance  to  ethanol  indicates 
that  these  compounds,  believed  to 
play  a  role  in  growth  and  differentiation 
in  the  developing  brain,  can  also  affect 
neuroadaptation  to  ethanol  in  the  adult 
brain.  As  previously  postulated,  changes 
in  synaptic  efficacy  in  the  adult  brain, 
including  those  that  underlie  cognitive 
function  and  adaptations  to  drugs,  may 
arise  from  the  same  mechanisms  that 
influence  growth  in  the  developing 
brain.  In  this  sense,  while  studies  of 
learning  and  memory  may  provide 
guidelines  for  investigation  of  neuro- 
adaptive  processes  involved  in  alcohol 
tolerance  and  dependence,  studies  of 
tolerance  may  also  provide  insight  into 
the  mechanisms  of  cognitive  function 
in  the  adult  brain. 

Dependence 

The  foregoing  discussion  provides  sev- 
eral clues  as  to  the  factors  or  systems 
that  cause  or  modulate  alcohol  depen- 
dence, defined  operationally  here  in 
part  as  a  compensatory  rebound  to 


133 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


chronic  ethanol  exposure  following  ces- 
sation of  that  exposure  (alcohol  with- 
drawal syndrome,  as  described  earlier  in 
this  chapter),  combined  with  a  strong 
desire  to  drink  (craving).  It  should  be 
noted  at  the  outset  that,  at  the  cellular 
level,  the  expected  neuronal  hyperex- 
citability  following  withdrawal  from 
chronic  ethanol  does  not  occur  in  all 
brain  regions  (e.g.,  in  accumbens  core 
neurons  [Nie  et  al.  unpublished  man- 
uscript]), despite  satisfaction  of  behav- 
ioral criteria  for  dependence.  However, 
data  from  other  brain  regions  suggest 
that  several  candidate  systems  could 
be  involved  in,  or  modulate,  the  with- 
drawal or  craving  aspects  following 
chronic  ethanol  exposure,  including 
several  ligand- gated  transmitter  systems 
(e.g.,  those  for  NMDA,  GABA,  5- 
HT,  acetylcholine,  and  ATP),  Ca++ 
channels,  and  the  G  protein-linked 
(metabotropic)  transmitter  or  modula- 
tors. Detailed  understanding  of  these 
factors  could  lead  to  powerful  therapeu- 
tic treatments  for  alcohol  dependence 
in  the  future.  In  this  regard,  at  least 
two  different  strategies  may  be  used 
to  develop  therapeutic  approaches  to 
dependence:  (1)  developing  new  types 
of  drugs  based  on  an  understanding  of 
the  mechanisms  of  the  underlying  per- 
turbation^) involved  and  (2)  developing 
new  drugs  or  improving  old  drugs  based 
on  a  newly  discovered  site  or  mecha- 
nism of  action  of  an  existing  effective 
but  empirically  discovered  drug. 

Pharmacotherapy  of  Withdrawal.  It 
might  be  assumed  from  the  previous  dis- 
cussion of  the  effects  of  chronic  ethanol 
on  transmitters  and  synaptic  transmis- 
sion that  the  best  strategy  for  treating 
withdrawal  phenomena  would  be  to 


compensate  in  some  way  for  changes  in 
those  channel  and  transmitter  systems 
most  affected  by  ethanol  and/or  its 
withdrawal.  Therefore,  likely  candidates 
for  such  therapeutic  approaches  would 
include  various  ion  channels,  the  GABA, 
NMDA,  ATP,  cholinergic,  5-HT, 
dopamine,  and  opioid  systems,  and  per- 
haps their  associated  second  messengers. 
To  date,  the  most  obvious  and  frequently 
discussed  treatments  include  the  ben- 
zodiazepines, for  enhancement  of 
GABAAergic  systems  to  overcome  per- 
ceived hyperexcitability  of  central  neu- 
rons; dopamine  receptor-related  drugs 
(perhaps  useful  primarily  as  anxiolytics); 
and,  more  recently,  5-HT  receptor- 
related  drugs,  such  as  the  5-HT 
uptake  inhibitors. 

Recent  research  findings  on  the  role 
of  GABAergic  systems  in  withdrawal, 
and  especially  after  chronic  intermit- 
tent ethanol  (CIE)  treatment,  are  very 
instructive  and  relevant  to  the  devel- 
opment of  long-lasting  dependence. 
The  CIE  models  of  the  Becker  and 
Olsen  groups  were  developed  largely 
to  mimic  the  human  condition  of 
alcoholism  more  closely  than  the  stan- 
dard continuous  long-term  treatment 
models,  and  they  indeed  lead  to  a  kin- 
dling of  greater  susceptibility  to  with- 
drawal hyperexcitability  and  seizure 
than  do  standard  models  (Becker  and 
Hale  1993;  Kokka  et  al.  1993).  The 
Olsen  group  has  used  the  CIE  rat  model 
for  study  of  hippocampal  GABAergic 
function  in  withdrawal  and  has  found 
prolonged  changes  in  some  indices  of 
GABAergic  inhibition,  such  as  decreased 
muscimol-induced  Cl~  flux  and 
paired-pulse  inhibition  (Kang  et  al. 
1996)  and  increased  expression  of  the 


134 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


GAB  A  receptor  a4  subunit  (Mahmoudi 
et  al.  1997).  By  contrast,  more  recent 
intracellular  studies  of  the  same  hip- 
pocampal  model  after  CIE  (Rang  et  al. 
1998)  showed  no  change  in  the  overall 
size  of  GABAergic  IPSPs.  Nonetheless, 
after  pretreatment  of  hippocampal 
slices  with  a  GABAB  receptor  antagonist 
to  unmask  ethanol  interactions  with 
GABAA-IPSPs  (see  Wan  et  al.  1996), 
a  significantly  greater  ethanol  enhance- 
ment of  GABAA-IPSP  areas  occurred 
in  hippocampal  slices  from  CIE  animals 
than  in  control  animals  (Kang  et  al. 
1998).  This  finding  shows  that  toler- 
ance to  this  ethanol  effect  does  not 
occur  in  the  CIE  model;  rather,  a  sensi- 
tization develops  that  could  play  some 
role  in  dependence  (Kang  et  al.  1998). 
However,  there  is  still  a  need  for  clarifi- 
cation as  to  whether  the  ethanol  effect 
on  IPSPs,  and  therefore  its  sensitization 
in  the  CIE-treated  hippocampus,  is 
exerted  pre-  or  postsynaptically  (Wan 
et  al.  1996;  Siggins  et  al.  1999)— that 
is,  indirectly  via  GABA  release  or 
uptake  or  directly  at  the  GABAA  recep- 
tor. A  further  major  finding  in  the 
Kang  et  al.  study  of  considerable  rele- 
vance to  treatment  strategies  was  a 
possible  sensitization  of  the  GABAergic 
system  to  benzodiazepine  inverse  ago- 
nists and  neurosteroids  in  the  hip- 
pocampus of  CIE  rats.  This  finding 
supports  the  possibility  raised  by  Suzdak 
and  colleagues  (1986)  that  binding 
sites  on  the  GABAA  receptor  for  benzo- 
diazepine inverse  agonists  may  be  linked 
to  changes  associated  with  tolerance 
and  dependence,  and  it  also  supports 
the  possibility  that  neurosteroids  might 
be  a  useful  avenue  for  alcohol  depen- 
dence therapy. 


Based  on  the  more  recent  cellular  and 
neurochemical  findings  (see,  e.g.,  the 
Synaptic  Transmission;  Presynaptic 
Mechanisms  section  earlier  in  this  chap- 
ter), it  would  seem  wise  also  to  focus  on 
the  newer  agents  acting  on  NMDA 
receptors  and  Ca++  channels,  or  perhaps 
on  systems  downstream  from  these  ele- 
ments (e.g.,  the  nitric  oxide  or  eicosanoid 
systems).  Thus,  agents  like  the  NMDA 
receptor  antagonists  MK-801,  meman- 
tine,  or  one  of  the  D-CPPene  com- 
pounds (NMDA  receptor  antagonists 
developed  by  Novartis  Pharma  that  can 
be  injected  peripherally),  Ca++  channel 
antagonists  like  nifedipine,  nitrendipine, 
or  nimodipine,  or  nitric  oxide  antago- 
nists such  as  nitroglycerin  should  come 
under  scrutiny  as  therapeutic  agents  for 
early  withdrawal  hyperexcitability  and 
associated  neurotoxicities.  These  com- 
pounds might  be  used  early  in  the 
detoxification  stage. 

Pharmacotherapy  of  Craving 
(Acamprosate,  Naltrexone).  Recently, 
distinctions  have  made  between  the 
mechanisms  underlying  the  withdrawal 
syndrome  and  craving  or  relapse.  There- 
fore, it  is  interesting  that  two  of  the 
newest  therapeutic  approaches  for 
alcoholism  involve  treating  the  more 
long-term  effects  of  alcohol  dependence 
(i.e.,  neuroadaptation  to  chronic  abuse, 
leading  to  craving).  These  approaches 
are  based  on  a  different  rationale  than 
that  for  treating  withdrawal  and  thus 
are  often  prescribed  several  weeks  after 
detoxification  or  initiation  of  abstinence. 
Initial  positive  clinical  trials  in  Europe 
for  acamprosate  and  in  the  United 
States  for  naltrexone  have  led  to  the  use 
of  these  two  drugs  found  to  be  effec- 
tive in  reducing  relapse  in  abstaining 


135 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


alcoholics  (Lhuintre  et  al.  1985; 
O'Malley  et  al.  1992;  Volpicelli  et  al. 
1992).  Obviously,  the  rationale  behind 
the  clinical  use  of  naltrexone  may  be 
found  in  the  lands  of  opiate  studies  in 
animals  and  humans  described  earlier 
in  this  chapter  and  in  chapter  7.  By  con- 
trast, the  original  rationale  for  acam- 
prosate  was  to  develop  a  congener  of 
GABA  (a  homotaurinate  derivative) 
that  might  counteract  ethanol's  pre- 
sumed GABAA  receptor-enhancing 
effects  (Lhuintre  et  al.  1985).  Behavioral 
studies  in  animals  have  shown  that 
acamprosate  can  reduce  the  ethanol 
deprivation  effect  (in  a  forced  absti- 
nence paradigm)  that  produces 
rebound  enhanced  responding  for 
ethanol  (Heyser  et  al.  1996;  Spanagel 
et  al.  1996). 

Despite  the  fact  that  acamprosate 
was  developed  as  a  GABA  receptor 
agonist,  initial  in  vivo  and  in  vitro 
electrophysiological  studies  of  cortical 
neurons  found  that  high  concentrations 
of  acamprosate  had  no  discernible 
GABAergic  effect  but  instead  acted  to 
reduce  glutamatergic  responses  (Zeise 
et  al.  1990,  1993).  Subsequent  studies 
in  two  different  slice  preparations,  hip- 
pocampus and  nucleus  accumbens, 
showed  that  acamprosate  again  had 
no  GABAAergic  action,  but  instead 
significantly  augmented  NMDA- 
EPSPs  and  responses  to  exogenous 
NMDA  (Madamba  et  al.  1996; 
Berton  et  al.  1998).  However,  in  con- 
trast to  the  studies  of  Zeise  and  col- 
leagues (1990,  1993),  acamprosate  had 
no  significant  effect  on  non-NMDA 
glutamatergic  EPSPs  in  these  brain 
regions.  Interestingly,  in  accumbens 
neurons  acamprosate  also  acted  like  a 


GABAB  antagonist  in  blocking  paired- 
pulse  inhibition  (a  presynaptic  site  of 
action)  of  IPSPs,  suggesting  that  the 
drug  might  act  on  GABAB  rather  than 
GABAA  receptors  (Berton  et  al.  1998). 
This  finding  also  seems  consistent 
with  the  metabotropic  hypothesis  of 
ethanol  sensitivity  (Siggins  et  al. 
1999)  and  suggests  that  GABAB 
receptors  would  be  an  interesting  tar- 
get for  alcoholism  therapy. 

This  idea  is  strengthened  by  pre- 
liminary electrophysiological  findings 
with  another  drug,  y-hydroxybutyrate 
(GHB),  shown  to  have  some  efficacy 
against  alcohol  dependence  in  humans 
and  in  animal  alcohol  preference  mod- 
els (Biggio  et  al.  1992;  Gallimberti  et 
al.  1992;  Gessa  and  Gallimberti  1992). 
Intracellular  recording  of  CA1  pyramidal 
neurons  in  a  hippocampal  slice  prepa- 
ration has  shown  that  GHB,  like  the 
GABAB  agonist  baclofen,  hyperpolarizes 
these  neurons  and  augments  the  inward- 
rectifying  Q  or  h  current  (Madamba 
et  al.  1997).  The  effects  of  both  GHB 
and  baclofen  were  blocked  by  the 
GABAB  antagonist  CGP  35348,  sug- 
gesting that  the  clinical  efficacy  of  GHB 
may  be  due  to  its  action  on  GABAB 
receptors,  and  providing  additional 
support  for  study  of  these  metabotropic 
receptors  as  therapeutic  targets  against 
alcoholism.  However,  it  should  be 
noted  that  GHB  is  also  being  used  in 
the  detoxification  stage,  probably 
because  it  has  alcohol-like  effects. 

Sensitization 

Evidence  for  a  direct  role  of  sensitization 
in  alcoholism  might  come  from  the 
demonstration  that  similar  biological 
systems  underlie  both  sensitization  and 


136 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


vulnerability  to  drug  self-a<iministration 
or  reinforcement.  Dopamine  systems 
have  been  widely  investigated  with 
regard  to  sensitization  to  other  drugs 
of  abuse,  and  research  results  have 
supported  their  involvement  in  drug 
reward.  Evidence  for  this  involvement 
has  been  found,  most  commonly,  by 
analysis  of  the  role  of  dopamine  systems 
in  drug  self- administration  (Koob  et 
al.  1987;  Britton  et  al.  1991;  Hubner 
and  Moreton  1991;  Caine  and  Koob 
1993;  Maldonado  et  al.  1993; 
Richardson  et  al.  1993;  Ng  and  George 
1994)  or  by  measuring  dopaminergic 
changes  associated  with  drug  self- 
administration  (Goeders  and  Smith 
1993;  Weiss  et  al.  1993;  Laurier  et  al. 
1994).  Enduring  changes  in  mesoac- 
cumbens  dopamine  transmission  have 
also  been  postulated  to  be  involved  in 
drug  sensitization  (Trulson  et  al. 
1987;  Robinson  et  al.  1988;  Peris  et 
al.  1990;  Segal  and  Kuczenski  1992; 
Kalivas  et  al.  1993;  Parsons  and  Jus- 
tice 1993;  Burger  and  Martin -Iverson 
1994;  Self  and  Nestler  1995).  How- 
ever, some  studies  support  involve- 
ment of  projections  involving  other 
pathways  and  neurotransmitters  inter- 
acting with  the  dopamine  system 
(Kalivas  and  Alesdatter  1993;  Kalivas 
et  al.  1993;  White  et  al.  1995),  as  well 
as  effects  independent  of  the  dopamine 
system  (Koob  and  Cador  1993).  This 
work  has  not  been  accomplished  for 
ethanol  sensitization.  Nestby  and  col- 
leagues (1997)  did  show  neurochemi- 
cal changes  associated  with  repeated 
ethanol  administration  that  paralleled 
those  associated  with  repeated  amphet- 
amine, morphine,  and  cocaine  admin- 
istration; there  was  increased  dopamine 


and  acetylcholine  release  from  nucleus 
accumbens  slices.  However,  they  pro- 
vided no  evidence  of  ethanol  sensiti- 
zation in  that  study. 

A  growing  body  of  literature  sup- 
ports hypothalamic-pituitary-adrenal 
axis  involvement  in  cocaine,  ampheta- 
mine, and  morphine  sensitization.  A 
series  of  studies  addressed  the  involve- 
ment of  this  axis  in  ethanol  sensitiza- 
tion (Roberts  et  al.  1995).  The  principal 
findings  were  as  follows:  (a)  repeated 
exposure  to  restraint  stress  sensitized 
mice  to  the  locomotor  stimulant  effects 
of  ethanol,  (b)  stress-induced  sensitiza- 
tion of  ethanol's  locomotor  stimulant 
effects  was  attenuated  by  a  glucocorti- 
coid receptor  antagonist,  and  (c)  the 
glucocorticoid  receptor  antagonist  was 
also  capable  of  preventing  sensitization 
to  ethanol  produced  by  repeated  ethanol 
injections.  The  only  other  study  we 
know  of  that  has  directly  assessed  any 
neuropharmacological  mechanism 
associated  with  ethanol  sensitization  is 
that  of  Broadbent  and  colleagues 
(1995),  who  reported  no  effect  of  a 
dopamine  antagonist  on  ethanol  sen- 
sitization. This  mechanistic  work 
should  be  expanded. 

Critical  Gaps 

in  Our  Knowledge 

Ligand- Gated  Ion  Channels 

Mechanisms  of  Receptor  Adaptations. 
Ethanol  alters  both  GABAA  and 
NMDA  receptor  function  and  results 
in  alterations  in  the  expression  of  vari- 
ous subunits  that  comprise  these 
receptor  channels.  Numerous  investi- 
gators have  hypothesized  that  alterations 
in  gene  expression  for  the  subunit 


137 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


proteins  could  result  in  alterations  in 
receptor  assembly  that  could  explain  the 
functional  alterations  in  these  receptors. 
Although  this  hypothesis  has  clear 
heuristic  value,  alterations  in  subunit 
assembly  have  not  been  directly  demon- 
strated in  response  to  ethanol  admin- 
istration. We  have  very  little  knowledge 
of  the  regulation  of  receptor  assembly 
for  these  receptors.  It  is  clear  that  chap- 
erone  and  other  proteins  are  involved 
in  these  processes.  This  is  an  important 
area  for  future  research. 

Most  ion  channel  proteins  are  subject 
to  posttranslational  modifications  that 
modify  the  functional  properties  of 
these  receptors.  Ethanol  has  been  shown 
to  activate  various  kinases  that  are 
capable  of  modifying  ion  channel  recep- 
tors. Does  this  process  underlie  ethanol  - 
induced  alterations  in  ion  channel 
receptor  function?  Is  this  process 
involved  in  ethanol  regulation  of  ion 
channel  gene  expression?  There  is  a  great 
deal  of  indirect  evidence  suggesting 
that  posttranslational  receptor  modifi- 
cations may  be  an  important  mechanism 
involved  in  ethanol  adaptations,  but 
there  has  never  been  a  direct  demon- 
stration that  ethanol  induces  such  an 
alteration  or  that  such  alterations  can 
explain  any  of  the  physiological  effects 
of  chronic  ethanol  administration.  Such 
investigations  are  of  critical  impor- 
tance and  interest. 

Another  possible  mechanism  medi- 
ating neuroadaptations  of  ion  channel 
receptors  following  chronic  ethanol 
exposure  involves  internalization  of 
the  receptor  complex.  There  is  substan- 
tial evidence  that  various  receptors  can 
be  internalized  (Calkin  and  Barnes 
1994),  and  this  mechanism  could 


explain  alterations  in  receptor  function. 
It  is  also  possible  that  ethanol  affects 
the  stoichiometry  of  ion  channel 
receptors.  However,  the  stoichiometry 
of  most  native  receptors  that  are  sensitive 
to  ethanol  remains  undetermined. 
Likewise,  a  dissociation  of  subunits  could 
account  for  decrements  in  ion  channel 
function  while  preserving  receptor  num- 
ber, yet  be  nearly  impossible  to  detect 
using  currently  available  techniques. 
Conformational  changes  in  receptor 
structure  are  another  potential  adap- 
tation that  may  explain  rapid  changes 
in  receptor  function.  Recent  studies 
demonstrating  ethanol  action  at  the 
second  transmembrane  domain  of 
glycine/GABAA  receptor  chimeras 
(Mihic  et  al.  1997)  suggest  that 
ethanol  may  affect  the  conformation  of 
ion  channel  receptors.  Finally,  although 
we  have  focused  on  postsynaptic  regu- 
lation by  ethanol,  physiological  adap- 
tations may  also  involve  presynaptic 
mechanisms.  This  possibility  should 
be  reexamined  in  the  light  of  identifi- 
cation of  multiple  novel  neurotrans- 
mitter transporters  and  more  sensitive 
molecular  techniques  to  investigate 
their  function. 

Significance  of  Regional  Differences  in 
Gene  Regulation.  Both  glutamate  and 
GABAA  receptor  subunits  are  differentially 
regulated  by  ethanol  in  hippocampus 
versus  cortex  and  other  brain  regions. 
Since  ethanol  mediates  distinct  physio- 
logical effects  in  distinct  brain  regions, 
this  differential  regulation  may  have 
significant  physiological  implications. 
Few  studies  have  addressed  the  role  of 
gene  regulation  in  specific  neuronal 
circuits  that  control  specific  ethanol- 
mediated  physiological/behavioral 


138 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


responses.  Such  studies  could  lead  to  a 
better  understanding  of  the  relation- 
ship of  molecular  adaptations  to  the 
behavioral  manifestations  of  tolerance, 
dependence,  and  sensitization. 

What  are  the  signal  transduction 
pathways  involved  in  ethanol  regulation 
of  ion  channel  genes?  Do  regional  dif- 
ferences in  the  regulation  of  a  single 
gene  suggest  multiple  mechanisms  of 
gene  regulation?  Is  ion  channel  gene 
regulation  dependent  on  stimulation  of 
the  ion  channel  independent  of  action 
on  the  membrane  receptors?  If  ethanol 
acts  directly  on  gene  promoters,  what 
are  the  signal  transduction  pathways 
involved  in  this  activity?  Will  this 
action  differ  in  various  brain  regions? 

Ethanol-Nicotine  Interactions.  Con- 
verging clinical  data  have  shown  that 
alcoholism  and  excessive  drinking  are 
several  times  more  prevalent  in  smok- 
ers than  in  nonsmokers  (Deher  and 
Fraser  1976;  Bien  and  Burge  1990). 
It  is  also  known  that  the  ability  to 
treat  alcoholism  is  enhanced  if  indi- 
viduals stop  smoking  and  vice  versa 
(Griffiths  et  al.  1976;  Johnson  and 
Jennison  1992;  Joseph  1993;  Gulliver 
et  al.  1995;  Murray  et  al.  1995). 
Because  of  the  interactive  association 
of  these  abused  substances,  the  chal- 
lenge will  be  to  resolve  the  basis  of 
this  strong  interaction  between  the 
actions  of  chronic  ethanol  and  chronic 
nicotine  exposure.  Consequently,  it  is 
extremely  important  that  we  have  a 
complete  understanding  of  the  direct 
interactions  these  chronically  abused 
substances  have  on  various  nACh 
isoreceptors  in  brain,  alone  and  when 
combined.  The  critical  question  to  be 
answered  is  how  changes  in  nACh 


receptor  function,  induced  by  the 
presence  of  chronic  nicotine  and 
ethanol,  contribute  to  and  perpetuate 
alcoholism  and  the  desire  to  smoke. 

Neurotransmitter  Systems 

With  respect  to  the  effect  of  chronic 
ethanol  on  transmitter  systems,  and 
given  the  important  role  the  dopamine 
system  is  likely  to  play,  one  of  the  first 
questions  that  might  be  asked  is:  what 
happens  to  the  membrane  and  synaptic 
properties  of  VTA  neurons  in  any  of 
the  alcohol-preferring,  chronic  ethanol, 
ethanol -withdrawn,  or  relapse  models? 
It  would  also  be  useful  to  know  if  there 
are  changes  in  dopamine  autoreceptor 
function  in  these  models,  given  the 
importance  many  researchers  place  on 
this  interesting  regulatory  property  of 
dopaminergic  neurons.  It  would  also 
be  helpful  to  know  if  there  are  changes 
in  possible  dopamine -ethanol  interac- 
tions in  target  areas  of  the  VTA  dopa- 
mine projection. 

There  are  even  more  gaps  in  our 
knowledge  of  the  5-HT  system  in  these 
alcohol  models.  First  and  foremost, 
we  have  no  information  on  what  hap- 
pens to  the  neuronal  or  molecular  prop- 
erties of  5-HT-containing  neurons 
(e.g.,  in  the  raphe  nuclei)  even  with 
acute  ethanol,  as  we  have  for  the  effects 
of  opiates  (see,  e.g.,  Pan  et  al.  1990). 
Understanding  this  aspect  in  chronic 
models  would  be  even  more  useful.  In 
addition,  it  would  be  helpful  to  know 
whether  there  are  ethanol-5-HT  inter- 
actions at  the  level  of  the  various  5  - 
HT  receptors  in  the  chronic  models, 
and  whether  there  are  interactions 
between  5-HT  and  dopamine  cells  in 
these  models  (i.e.,  the  studies  of  the 


139 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Brodie  group  need  to  be  repeated  in 
the  chronic  models). 

Similar  questions  apply  to  the  role  of 
the  opioid  systems  in  the  chronic  ethanol 
or  preference  models.  Fortunately,  there 
is  a  great  background  of  data  from  mul- 
tiple brain  regions  on  opiate  effects  or 
functions  of  opioid  systems  that  can 
be  used  for  comparison  in  the  chronic 
or  preference  models.  As  for  the 
dopamine  and  5-HT  systems,  under- 
standing possible  changes  in  the  opi- 
oid-ethanol  interactions  in  the 
chronic/preference  models  would  also 
be  very  instructive. 

As  all  three  of  these  transmitters  act 
mostly  on  the  G  protein-linked,  7- 
transmembrane -spanning- domain 
superfamily  of  receptors,  they  fall  into 
the  generic  category  of  metabotropic 
receptors  capable  of  altering,  through 
various  kinases  and  phosphatases,  the 
phosphorylation  state  of  other  recep- 
tors, ion  channels,  and  other  (intracel- 
lular) regulatory  proteins.  Therefore,  some 
effort  should  be  made  toward  under- 
standing how  these  three  transmitter 
systems  might  regulate  the  ethanol 
sensitivity  of,  for  example,  ligand-gated 
ion  channels,  especially  in  the  depen- 
dence or  preference  models  (see  Sig- 
gins  et  al.  1999).  Such  studies  of 
metabotropic,  posttranslational  systems, 
and  their  related  second  messengers 
and  kinases/phosphatases,  have  the 
potential  to  lead  to  new  and  exciting 
treatments  for  alcoholism  or  with- 
drawal phenomena. 

Since  the  synapse  seems  to  be  the  most 
ethanol-sensitive  neuronal  element, 
the  study  of  synapses  either  in  isolation 
or  under  strong  experimental  control 
would  seem  to  be  paramount.  In  fact, 


at  the  cellular  and  molecular  levels, 
there  is  a  need  especially  for  electrophys- 
iological studies  of  the  role  of  all 
ethanol-sensitive  transmitter  systems 
in  protracted  abstinence,  relapse,  or  crav- 
ing models.  We  encourage  the  use  in 
these  chronic  models  of  new  cutting- 
edge  techniques  to  isolate  pre-  versus 
postsynaptic  sites  of  action:  for  example, 
statistical  analyses  of  spontaneous  and 
miniature  synaptic  potentials  or  cur- 
rents, analysis  of  paired-pulse  facilitation, 
and  study  of  single-fiber  synaptic  units 
or  synaptic  pairs  in  slices,  or  autaptic 
synapses  in  cultures  (Malinow  1991; 
Dobrunz  et  al.  1997;  Goda  and  Stevens 
1998).  It  appears  that  some  aspects  of 
these  exciting  new  avenues  in  alcohol 
research  are  now  getting  under  way; 
for  example,  analysis  of  miniature 
synaptic  currents  for  elucidation  of 
presynaptic  ethanol  actions  (R.  Mor- 
risett,  personal  communication,  July 
1998;  J.  Weiner,  personal  communi- 
cation, July  1998). 

There  also  is  a  need  for  additional 
cellular  studies,  but  now  in  the  depen- 
dence, relapse,  and  preference  models, 
of  drugs  known  to  be  efficacious  in 
treating  alcoholics.  For  example,  it 
would  be  very  interesting  to  know  what 
membrane  or  synaptic  effects  acam- 
prosate,  naltrexone,  or  GHB  would 
have  at  various  time  points  in  a  relapse 
or  protracted  abstinence  model,  or 
how  long-term  treatment  with  these 
drugs  could  alter  the  neuroadaptations 
seen  in  these  models.  This  informa- 
tion could  then  be  used  strategically 
to  develop  additional  new  therapies. 

In  terms  of  finding  the  neuroadap- 
tion(s)  most  important  for  alcoholism, 
we  also  need  to  examine  the  role  of 


140 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


neurotoxicity,  as,  for  example,  that 
produced  by  NMD  A  receptor  actions, 
activation  of  excessive  Ca++  conduc- 
tances, or  eicosanoids.  Here  we  might 
benefit  from  review  of  the  findings  of 
the  Walker/Hunter,  Morrisett,  Little, 
and  Crews  groups.  The  recent  studies 
of  the  Collins  group  (see  Corso  et  al. 
1998)  represent  a  significant  step  in 
this  direction.  It  would  be  important  to 
know  if  the  newer  antagonists  of  NMDA 
receptors  and  Ca++  currents  block 
ethanol  preference,  relapse,  or  craving. 
In  a  related  area,  there  is  an  urgent 
need  for  understanding  possible  changes 
in  the  anatomical  distribution  of  trans- 
mitter receptors  or  their  subunits  in 
the  dependence  and  preference  models 
by  using,  for  example,  quantitative 
immunocytochemical  confocal  imaging 
of  receptor  subunits  tagged  with  mon- 
oclonal antibodies,  as  carried  out  by  the 
Morrison  group  (Siegel  et  al.  1995; 
Gazzaley  et  al.  1996;  Nimchinsky  et 
al.  1996).  Such  studies  could  tell  us  if 
neuroadaptation  involves  subtle  trans- 
locations of  receptors  (e.g.,  from  den- 
dritic spines  to  somata  or  dendritic 
shafts)  or  frank  loss  of  receptors.  The 
use  of  patch-clamp  recording  com- 
bined with  quantitative  single-cell 
reverse  transcriptase-polymerase  chain 
reaction  (RT-PCR)  in  chronic  ethanol, 
relapse,  or  preference  models,  similar 
to  that  currently  being  performed  in 
acute  ethanol  paradigms  by  the  Yeh 
group  (Eberwine  et  al.  1992;  Grigorenko 
and  Yeh  1994),  could  tell  us  if  neu- 
roadaptation involves  changes  in  sub- 
unit  composition  (stoichiometry)  in 
single  cells.  However,  as  with  all  such 
studies,  there  will  be  variability  con- 
cerns and  problems  of  quantification 


of  measures  across  subjects,  neuron 
types,  or  treatment  groups.  Therefore, 
there  will  be  a  great  need  in  the  future 
to  develop  new  methods  (e.g.,  statistical 
designs,  quantification  of  immunocy- 
tochemical and  RT-PCR  data,  stan- 
dardization of  synaptic  stimulation, 
and  dose-response  analyses)  to  deal 
with  these  sources  of  variability. 

Finally,  there  is  a  gap  in  our  knowl- 
edge of  the  more  long-term  molecular 
changes  that  might  occur  in  specific 
brain  regions  (e.g.,  the  VTA,  accumbens, 
or  amygdala)  in  the  chronic  ethanol, 
relapse,  or  preference  models.  For  this 
we  need  studies  of  the  expression  of 
IEGs  (e.g.,  c-fos  and  c-jun),  the  RNA 
for  important  transmitter  receptors 
(e.g.,  dopamine,  5-HT,  and  opiate 
receptors)  and  their  proteins.  For 
example,  it  would  be  important  to 
know  the  time  course  of  possible 
changes  in  opiate  receptor,  IEG,  or 
CREB  expression  with  chronic 
ethanol,  relapse,  or  preference,  as  has 
been  seen  with  chronic  psychostimu- 
lant treatment  (J.Q.  Wang  et  al. 
1994;  McGinty  and  Wang  1998). 

The  Influence  of  Gender 
on  Ethanol  Dependence 
and  Withdrawal 

Many  neuroactive  steroids  are  derived 
from  progesterone  and  so  occur  at  dif- 
ferent levels  in  males  and  females. 
Female  rats  show  higher  3a,5a-THP 
levels  in  plasma  and  brain  than  male 
rats,  and  these  levels  fluctuate  across 
the  estrous  cycle  (Purdy  et  al.  1990; 
Paul  and  Purdy  1992;  Corpechot  et 
al.  1993).  The  levels  of  3ct,5ct-THP 
in  brain  during  estrus  in  female  rats,  or 
acute  stress  in  male  rats,  are  sufficient 


141 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


to  modulate  GABAA  receptor  function 
in  brain  (Purdy  et  al.  1990,  1991; 
Paul  and  Purdy  1992).  Functional 
changes  in  GABAA  receptor  stimulation 
and  responses  are  observed  during  the 
estrous  cycle  and  differ  between 
female  and  male  rats  (Westerling  et  al. 
1991;  Wilson  1992;  Finn  and  Gee 
1993).  In  addition,  levels  of  the  excita- 
tory neuroactive  steroid  dehy- 
droepiandrosterone  sulfate  (DHEAS), 
are  significantly  higher  in  men  than  in 
women  (Orentreich  et  al.  1984). 
Therefore,  males  and  females  operate 
with  a  differing  hormonal  regulation  at 
the  receptor  level,  in  addition  to  gen- 
der differences  in  genomic  regulation. 
The  differences  in  the  steroid  hormone 
environment  between  males  and 
females  could  have  an  effect  on  the 
influence  of  ethanol  or  other  drugs  in 
the  brain. 

Studies  have  shown  gender  differences 
in  measures  of  ethanol  dependence  and 
withdrawal  and  the  response  to  3a,5a- 
THP.  Ethanol- with  drawn  female  rats 
show  greater  sensitization  to  the  anti- 
convulsant effect  of  3a,5a-THP  than 
ethanol- with  drawn  male  rats  (Devaud 
et  al.  1995a).  Female  rats  also  exhibit 
increased  sensitization  to  the  anticonvul- 
sant effects  of  THDOC  compared  with 
male  rats  (Devaud  et  al.  1998).  Further- 
more, we  recently  observed  differences 
in  the  effects  of  ethanol  dependence 
on  both  GABAA  receptor  and  NMDA 
receptor  subunit  gene  expression 
(Devaud  and  Morrow  1998;  Devaud 
et  al.  1998).  Cerebral  cortical  levels  of 
GABAA  receptor  al  subunit  peptide 
are  not  decreased  in  dependent  female 
rats,  although  a  decrease  is  consistently 
observed  in  male  rats  (Devaud  et  al. 


1997).  Furthermore,  chronic  ethanol 
administration  increases  NMDA  recep- 
tor NR1  subunit  peptide  expression  in 
female  cerebral  cortical  homogenates, 
but  not  in  male  cortex  (Devaud  and 
Morrow  1998).  NMDA  receptor  NR2B 
subunit  peptide  expression  is  increased 
in  both  male  and  female  cortical  homo- 
genates (Devaud  and  Morrow  1998). 
Since  both  males  and  females  exhibit 
comparable  signs  of  ethanol  depen- 
dence, including  equivalent  seizure 
thresholds  during  ethanol  withdrawal 
(Devaud  et  al.  1995#),  what  is  the 
significance  of  gender  differences  in 
the  effects  of  ethanol  dependence  on 
GABAA  and  NMDA  receptor  subunit 
expression?  How  do  such  divergent 
alterations  in  these  receptors  produce 
similar  behavioral  adaptations?  What 
are  the  modulatory  mechanisms  that 
control  these  responses? 

Studies  in  Human  Alcoholics 

Are  the  alterations  in  receptor  expression 
observed  in  animal  models  of  ethanol 
dependence  predictive  of  changes  in 
human  alcoholics?  Like  ethanol- 
dependent  rodents,  human  alcoholics 
exhibit  behavioral  adaptations  to  pro- 
longed alcohol  intake,  including  alco- 
hol tolerance  and  benzodiazepine  and 
barbiturate  cross-tolerance  (Woo  and 
Greenblatt  1979).  Unfortunately,  there 
is  a  paucity  of  data  in  human  alcoholics, 
and  the  data  that  exist  often  are  quite 
different  from  the  effects  of  chronic 
ethanol  exposure  in  animal  models. 
There  are  examples  of  this  dichotomy 
with  respect  to  both  GABA  and  NMDA 
receptor  expression. 

Positron  emission  tomography 
(PET)  studies  show  a  decrease  in 


142 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


benzodiazepine  binding  in  the  frontal 
lobes  of  human  alcoholics  relative  to 
normal  control  subjects  (Gilman  et  al. 
1996).  Preliminary  single  photon  emis- 
sion computed  tomography  (SPECT) 
studies  suggest  a  decrease  in  benzodi- 
azepine binding  in  the  inferior  medial 
frontal  cortex  and  in  the  temporal  and 
parietal  cortices  (Abi-Dargham  et  al. 
1995;  Lingford-Hughes  et  al.  1998). 
PET  studies  of  the  metabolic  (2- 
deoxyglucose)  responses  to  a  benzo- 
diazepine challenge  show  a  reduction 
in  the  inhibitory  effects  of  benzodi- 
azepines and  further  support  the 
decrement  of  benzodiazepine  recep- 
tors in  specific  brain  regions  of  alco- 
holics (Volkow  et  al.  1997).  These 
decrements  in  benzodiazepine  receptor 
density  are  inconsistent  with  studies  in 
animal  models  of  ethanol  dependence, 
where  no  change  in  benzodiazepine 
receptor  binding  is  observed  (see  table 
1).  However,  there  is  a  decrease  in  the 
sensitivity  of  GABAA  receptors  to  ben- 
zodiazepines in  animal  studies  (see 
table  1).  In  addition,  the  decrease  in 
the  density  of  low- affinity  [3H]musci- 
mol  sites  and  the  decline  in  GABAA 
receptor  al  subunit  expression 
observed  in  the  cerebral  cortex  of 
ethanol-dependent  rodents  are  not 
observed  in  human  studies. 

In  postmortem  human  brain,  the 
GABAergic  correlates  of  ethanol  depen- 
dence also  appear  to  differ  from  rat 
models  of  ethanol  dependence. 
[3H]muscimol  binding  density  is 
greater  in  alcoholic  cerebral  cortex 
(Tran  1981)  and  in  the  superior 
frontal  gyrus  of  noncirrhotic  alcoholics 
(Dodd  et  al.  1992)  compared  with 
normal  control  subjects.  Significant 


increases,  decreases,  and  no  change  in 
benzodiazepine  binding  have  been 
reported  (Freund  and  Ballinger  1988; 
Dodd  et  al.  1992;  Dodd  1995).  GABAA 
receptor  al  subunit  mRNA  levels 
were  reported  to  increase  in  the  frontal 
cortex  of  noncirrhotic  alcoholics  (Lewohl 
et  al.  1997),  although  we  recently 
found  no  change  in  GABAA  receptor 
al  or  a4  subunit  mRNA  or  peptide 
levels  in  frontal  cortex  of  alcoholics 
(Mitsuyama  et  al.  1998).  These  results 
do  not  correlate  with  data  from  ani- 
mal models  of  ethanol  dependence, 
which  have  shown  reproducible 
changes  in  GABAA  receptor  gene 
expression.  The  most  parsimonious 
explanation  for  these  discrepancies, 
supported  in  part  by  the  distinct 
regional  differences  in  benzodiazepine 
binding  observed  with  in  vivo  imaging 
techniques  (Abi-Dargham  et  al. 
1995),  is  that  changes  in  GABAA 
receptor  expression  are  localized  to 
particular  cortical  regions.  In  addition, 
differences  between  the  longevity  of 
human  alcoholism  and  animal  models 
of  ethanol  dependence  probably  con- 
tribute to  these  discrepancies. 

There  are  also  discrepancies  between 
human  studies  and  animal  studies  with 
respect  to  changes  in  glutamate  recep- 
tors. The  kainate  receptors  GluR2  and 
GluR3  appear  to  be  elevated  in  human 
postmortem  hippocampus  of  alcohol 
abusers  (Breese  et  al.  1995),  but  not  in 
ethanol-dependent  rats  (Trevisan  et  al. 
1994;  Buckner  et  al.  1997).  Clearly, 
further  studies  of  human  alcoholics  are 
critically  needed.  Human  imaging 
studies  will  be  particularly  useful  to  our 
understanding  of  the  adaptations 
induced  by  ethanol  in  human  alcoholics. 


143 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


The  fact  that  ion  channels  represent 
a  clear  and  prominent  site  of  action  of 
ethanol  prompts  speculation  regarding 
the  role  of  these  channels  in  individual 
responses  to  ethanol.  The  findings  that 
ethanol  can  act  directly  on  specific  ion 
channel  subtypes  suggests  that  allelic 
differences  in  channel  subunit  expression 
and/or  amino  acid  composition  could 
have  dramatic  effects  of  responses  to 
ethanol.  Individuals  with  a  family  history 
of  alcoholism  are  known  to  have  an 
innately  greater  tolerance  (or  lesser 
sensitivity)  to  ethanol  than  those  with- 
out a  family  history  (Schuckit  1994). 
Since  even  a  minor  mutation  in  subunit 
structure  has  been  shown  to  change  the 
sensitivity  of  an  ion  channel  to  ethanol 
(Mihic  et  al.  1997),  it  is  possible  that 
allelic  differences  in  ion  channel  sub- 
units  underlie  genetic  differences  in 
ethanol  responses.  These  speculations 
require  additional  research;  however,  the 
continuously  accumulating  evidence 
that  ethanol  acts  on  ion  channels  pro- 
vides the  impetus  to  determine  if  these 
sites  of  ethanol  action  also  contribute 
to  the  development  of  alcoholism. 

Interactions  Among 
Neurotransmitter  Systems 

GABAA-NMDA  receptor  interactions 
have  been  documented.  Grayson  and 
colleagues  (Memo  et  al.  1991;  Zhu  et 
al.  1995)  have  demonstrated  NMDA- 
mediated  alterations  in  GABAA  recep- 
tor function  and  gene  expression  in 
cultured  neurons.  Since  many  neurons 
contain  both  GABAA  and  NMDA 
receptors  and  ethanol  acts  directly  on 
subtypes  of  both  GABAA  and  NMDA 
receptors,  this  type  of  modulatory 
activity  may  play  an  important  role  in 


adaptations  to  ethanol.  This  type  of 
interaction  may  contribute  to  the 
complexity  of  factors  that  regulate  ion 
channel  function  and  expression  and 
may  help  explain  regional  differences  in 
the  effects  of  chronic  ethanol  adminis- 
tration on  ion  channel  function  and 
gene  regulation. 

Integration  of  Ethanol's  Effects 
at  the  Level  of  Signal  Transduction 

The  AC/cAMP  system  in  neurons  has 
been  the  most  extensively  studied  signal 
transduction  pathway  with  respect  to 
chronic  effects  of  ethanol.  However, 
the  mechanism  of  the  desensitization 
of  this  system  following  chronic 
ethanol  treatment  is  not  yet  clear. 
New  knowledge  regarding  the  isoforms 
of  AC,  their  differential  sensitivity  to 
the  acute  effect  of  ethanol,  and  their 
differential  modes  of  regulation,  sug- 
gests that  investigations  of  quantitative 
or  qualitative  changes  induced  by  chronic 
ethanol  exposure  in  these  enzymes  may 
prove  fruitful.  In  addition,  more 
detailed  analysis  of  G  protein  quantity — 
in  particular,  in  neurons — may  provide 
a  better  understanding  of  the  role  of 
these  proteins  in  AC  desensitization. 
It  is  important  to  realize  that  G  proteins 
affect  certain  isoforms  of  AC  through 
their  |3y  subunits,  as  well  as  the  a  sub- 
units,  and  therefore  G  proteins  other 
than  Gs  or  Gi  could  be  involved  in 
changes  in  AC  activity  produced  by 
chronic  ethanol  treatment. 

In  addition  to  AC,  some  studies  of 
the  effects  of  chronic  ethanol  expo- 
sure on  PKA  and  PKC  have  been 
performed.  There  is  significant  cross- 
talk between  these  systems;  for  exam- 
ple, PKC  can  modulate  the  activity  of 


144 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


certain  isoforms  of  AC,  leading  to 
changes  in  cAMP  generation  and 
PKA  activity.  Although  chronic  ethanol 
treatment  has  been  shown  to  increase 
neurite  outgrowth  mediated  by  PKC 
in  vitro,  the  significance  of  such  effects 
in  vivo  (e.g.,  new  synaptic  connec- 
tions) and  the  interaction  of  the  PKC 
and  PKA  pathways  to  produce  such 
changes  have  not  been  investigated. 

Other  signal  transduction  system 
interactions  that  have  not  yet  been 
addressed  are  those  between  the  het- 
erotrimeric  G  proteins  and  the  "small" 
GTPases  of  the  Ras  superfamily 
(Bokoch  1996).  There  is  substantial 
evidence  that  G  protein  (3y  subunits 
mediate  activation  of  the  mitogen- 
activated  protein  (MAP)  kinase  path- 
way by  G  protein-coupled  receptors, 
and  do  so  through  activation  of  Ras. 
Because  the  classical  MAP  kinase 
pathway  (i.e.,  ERK1  and  ERK2)  is 
mitogenic,  changes  in  the  activity  of 
this  pathway  could  conceivably  affect 
neuronal  survival  by  opposing  apoptotic 
effects  and  could  also  be  involved  in 
the  generation  of  new  synaptic  con- 
nections, that  is,  the  structural  changes 
associated  with  neuroadaptation.  A  study 
by  Brambilla  and  colleagues  (1997) 
demonstrated  a  role  for  the  Ras  signaling 
pathway  in  a  process  of  memory  consol- 
idation in  mice,  suggesting  that  this 
pathway  may  play  a  more  general  role 
in  neuroadaptation. 

The  myriad  interconnections  among 
various  signal  transduction  systems 
(Hop kin  1997)  that  influence  cell  sur- 
vival, differentiation,  and  responses  to 
stress  and  other  external  stimuli  provide 
an  important  area  for  investigation  of 
the  chronic  effects  of  ethanol  that  lead 


to  neuroadaptation.  The  studies  of  the 
AC  signaling  system  have  already  pro- 
vided evidence  for  a  role  of  this  system 
in  ethanol  neuroadaptation.  Investiga- 
tions of  the  interactions  of  this  system 
with  other  cellular  signaling  pathways, 
and  modification  of  these  interactions  by 
ethanol,  are  promising  areas  of  research. 

Integration  of  Ethanol's  Effects  at 
the  Level  of  Neural  Circuitry 

Systems  (neural  circuitry)  analyses  of 
ethanol  actions  and  preference  are  among 
the  most  laborious  and  time-consum- 
ing, perhaps  accounting  for  the  relative 
paucity  of  such  studies  in  the  alcohol 
field.  The  two  most  popular  approaches 
for  such  investigations  are  ( 1 )  the  use 
of  multiple  recording  and  stimulus 
sites  in  anesthetized  animals  and  (2) 
the  use  of  single-  or  multi-unit  record- 
ings in  freely  moving  animals  that  may 
be  self- administering  ethanol  in  an  oper- 
ant situation.  Several  recent  and  prelim- 
inary studies  highlight  the  possibilities 
these  approaches  offer  for  understand- 
ing the  neural  circuit  correlates  of 
ethanol  preference. 

Hints  of  the  first  type  of  study,  using 
long-term  ethanol  treatment  and  with- 
drawal followed  by  electrophysiological 
studies  in  anesthetized  animals,  appeared 
in  the  early  reports  from  the  Walker/ 
Hunter  group  (cited  in  the  Synaptic 
Transmission;  Presynaptic  Mechanisms 
section  earlier  in  this  chapter).  How- 
ever, these  were  studies  on  relatively 
local  circuits  (e.g.,  Schaffer  collaterals 
to  CA1  pyramidal  neurons)  within  the 
hippocampal  CA1  and  within  the  den- 
tate. Similar  but  more  systems-ori- 
ented studies  are  now  emerging:  for 
example,  those  based  on  earlier  findings 


145 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


that  the  function  of  (local)  recurrent 
inhibitory  circuits  (e.g.,  as  expressed 
in  paired-pulse  inhibition)  recorded  in 
CA1  and  dentate  was  markedly  regu- 
lated by  extrinsic  sources  from  VTA 
and  lateral  septum,  and  that  acute 
ethanoPs  effect  in  enhancing  this 
inhibitory  function  was  exerted  at 
these  extrinsic  sites  (Criado  et  al. 
1994).  Two  weeks  of  chronic  ethanol 
vapor  treatment  followed  by  withdrawal 
for  2-8  hours  (under  halothane  anes- 
thesia) reduced  paired-pulse  inhibition 
(only  in  dentate)  with  a  return  to  nor- 
mal within  24  hours.  Interestingly, 
acute  ethanol  injected  at  this  point  pro- 
duced a  paradoxical  decrease  in  paired- 
pulse  inhibition,  in  contrast  to  ethanol's 
effect  in  controls,  suggesting  complex 
adaptive  circuit  responses  between 
hippocampus  and  VTA/lateral  septum 
(Steffensen  and  Henriksen  1997). 
Similar  studies  now  planned  for  the 
hippocampal/accumbens/amygdala 
complex  could  reveal  critical  informa- 
tion regarding  the  adaptation  to  chronic 
ethanol  in  the  mesolimbic,  extended 
amygdala  reward  pathways. 

The  second  favored  approach,  study- 
ing ethanol  preference  in  awake  ani- 
mals, is  best  exemplified  by  a  recent 
publication  from  the  Woodward  group 
(Woodward  et  al.  1998;  see  also  Givens 
et  al.  1998).  This  work  elegantly  illus- 
trates what  can  be  learned  about  the 
function  of  temporal-spatial  functioning 
of  distributed  neural  elements  during 
operant  responding  for  drugs  of  abuse. 
Here,  simultaneous  groups  of  many 
individual  neurons  in  one  or  more  brain 
regions  are  recorded  during  specific, 
controlled  behavioral  events.  Performing 
such  recordings  from  accumbens  and 


amygdala  neurons  of  the  rat  meso- 
corticolimbic  circuit  during  operant 
responding  for  ethanol  has  revealed 
that  accumbens  neurons  show  multiple 
activity  patterns  within  each  cell  just 
prior  to  and  during  ethanol  responding, 
with  slowing  of  firing  predominating. 
Recording  of  neuronal  ensembles  (e.g., 
consisting  of  25  neurons)  has  begun 
to  reveal  what  appears  to  be  patterns 
of  response  activity  across  multiple 
neurons  that,  with  the  use  of  sophisti- 
cated statistical  pattern  recognition 
techniques,  may  ultimately  be  used  to 
reveal  how  networks  of  neurons  con- 
trol behavior  (e.g.,  via  propagation  of 
complex  conditioned  cues  within  the 
mesolimbic  system  to  initiate  ethanol - 
seeking  behavior). 

As  pointed  out  by  these  authors, 
the  power  of  this  method  for  under- 
standing the  systems  complexities  of 
ethanol  actions  and  alcohol  addiction 
is  great.  For  example,  in  this  study 
neuronal  recordings  began  only  after 
the  rats  were  trained  to  self- administer 
ethanol;  it  would  be  highly  desirable 
to  observe  neuronal  ensemble  activity 
during  learning  of  this  behavior,  to  bet- 
ter understand  the  events  leading  up 
to  the  addiction  process.  Alternatively, 
one  could  apply  this  method  to  a  pro- 
tracted abstinence  model  to  follow 
changes  in  ensemble  activity  just  prior 
to  relapse,  when  craving  might  be  at 
its  highest.  In  addition,  similar  recording 
methods  may  be  used  in  conjunction 
with  microdialysis  (Ludvig  et  al.  1998) 
or  voltammetric  (Rebec  1998)  methods, 
to  assess  neurochemical  correlates  (e.g., 
transmitter  release)  of  the  electrophys- 
iological and  behavioral  events  in  these 
alcohol  models. 


146 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Finally,  the  use  of  organotypic 
(explant)  brain  slice  cultures  is  a  relatively 
new  approach  for  the  future  study  of 
adaptation  in  neural  circuits  with 
chronic  ethanol  (Thomas  et  al.  1998). 
These  cultures  can  be  prepared  from 
horizontal  slices  of  whole  forebrain  to 
include,  for  example,  a  functional  corti- 
costriatal  pathway  that  can  be  stimulated 
selectively  and  recorded  at  multiple  sites 
with  extracellular  field  and  patch-clamp 
techniques.  These  cultures  may  then 
be  examined  before  and  after  chronic 
ethanol  treatment  (by  bath  application), 
followed  by  ethanol  withdrawal.  It  also 
may  be  of  interest  to  apply  an  in  vitro 
sort  of  "protracted  abstinence"  protocol 
to  such  cultures.  Major  advantages  of 
such  a  model  system  include  the  possi- 
bility of  using  a  within-subjects  design 
and  allowing  cutting-edge  recording  and 
pharmacological  techniques  for  analysis 
of  synaptic  events  across  neural  circuits 
or  neuronal  ensembles  during  various 
stages  of  the  ethanol  protocol. 

STRATEGIES 

FOR  FUTURE  WORK 

Paradigms  for  Ethanol 
Administration  and  Choice 
of  Neural  Systems  for  Study 

The  most  difficult  issue  to  address  in 
studies  of  cellular  and  molecular  changes 
induced  by  chronic  ethanol  exposure  is 
whether  these  observed  changes  are  in 
fact  related  to  neuroadaptation  to 
ethanol.  The  previous  sections  have 
focused  on  neurochemical  and  molec- 
ular alterations  induced  by  chronic 
ethanol  ingestion  or  exposure,  and  in 
some  cases  the  relationship  of  changes 


in  the  function  of  a  particular  neuro- 
chemical system  to  ethanol-induced 
neuroadaptation  has  been  discussed. 
However,  more  work  is  needed  to 
determine  the  importance  of  each  of 
these  "candidate  systems"  for  ethanol- 
induced  neuroadaptations.  As  pointed 
out  earlier,  any  change  in  neuronal 
function  is  likely  to  have  many  down- 
stream effects.  Therefore,  one  will  find 
effects  of  chronic  ethanol  treatment  on 
any  number  of  neuronal  systems,  but  the 
question  of  which  is  the  primary  effect 
that  mediates  behavioral  aspects  of 
neuroadaptation  remains.  Added  to 
this  complexity  is  the  fact  that  different 
investigators  use  differing  paradigms 
of  ethanol  administration,  often  not 
measuring  blood  and  brain  ethanol 
levels,  let  alone  the  development  of 
tolerance,  physical  dependence,  or 
sensitization.  Either  it  is  assumed  that 
the  ethanol  treatment  has  produced  a 
neuroadaptation,  or  this  aspect  is 
ignored,  and  cellular  or  neurochemical 
changes  are  simply  ascribed  to  the 
chronic  ethanol  exposure.  A  more  prof- 
itable approach  is  to  use  or  develop 
ethanol  administration  paradigms  that 
lead  to  the  desired,  measurable  behaviors 
or  physiological  changes  characteristic 
of  tolerance  or  physical  dependence,  for 
example,  and  to  correlate  candidate 
cellular  and  molecular  changes  tempo- 
rally and  quantitatively  with  the  partic- 
ular behavioral  aspect  of  neuroadaptation 
that  is  under  investigation. 

There  are  a  number  of  other 
approaches  that  can  lead  to  a  better 
understanding  of  the  mechanisms  of 
adaptation  to  ethanol.  One  of  these 
approaches  has  been  used  with  some 
success  in  the  studies  of  tolerance 


147 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


already  described — alteration,  prior  to 
alcohol  exposure,  of  brain  systems  that 
are  believed  to  play  a  role  in  neuroad- 
aptation  (extrinsic  systems).  Studies 
can  then  be  performed  to  determine 
whether  these  molecular  or  neurochem- 
ical interventions  affect  the  develop- 
ment, expression,  or  loss  of  tolerance, 
physical  dependence,  or  sensitization. 
This  approach  includes  newer  methods 
such  as  generation  of  transgenic  or 
knockout  mice.  It  should  be  kept  in 
mind  that  the  extrinsic  systems  do  not 
necessarily  undergo  changes  during 
the  course  of  chronic  ethanol  adminis- 
tration (although  they  can).  However,  in 
the  absence  of  these  systems,  particular 
neuroadaptive  processes  cannot  occur. 
More  consideration  also  needs  to  be 
given  to  the  adaptation  that  is  to  be 
studied.  For  example,  the  complex 
nature  of  tolerance  means  that  one 
cannot  simply  administer  ethanol  to 
animals  and  generate  a  generic  form  of 
"tolerance."  First,  the  form  of  func- 
tional tolerance  that  is  being  studied 
must  be  considered,  including  the  role 
of  learning  and  memory.  The  develop- 
ment of  environment-dependent  tol- 
erance, for  example,  may  entail  changes 
that  are  different  from  those  occurring 
in  association  with  environment- 
independent  tolerance  (Melchior  and 
Tabakoff  1985).  Second,  it  is  impor- 
tant, when  assessing  changes  at  the 
cellular  level,  to  consider  which  effect 
of  ethanol  is  demonstrating  tolerance. 
For  instance,  one  might  examine 
changes  in  the  GABAA  receptor  in 
relation  to  the  development  of  toler- 
ance to  the  anxiolytic  and/or  incoor- 
dinating  effects  of  ethanol — but  is  this 
system  as  important  in  tolerance  to 


the  hypothermic  effect  of  ethanol? 
This  question  bears  on  the  notion  of 
"intrinsic"  systems — that  is,  the  systems 
that  encode  tolerance  to  particular 
effects  of  ethanol.  If  we  know  which 
neurochemical  systems  are  involved  in 
a  particular  behavior,  we  can  devise 
experiments  to  determine  whether 
changes  in  those  systems  are  important 
for  tolerance  to  the  effects  of  ethanol 
on  that  behavior.  It  is  often  assumed 
that  those  systems  most  sensitive  to 
acute  perturbations  by  ethanol  will 
adapt  to  the  chronic  presence  of 
ethanol.  While  this  is  undoubtedly 
true,  these  systems  may  or  may  not  be 
important  for  a  particular  neuroadaptive 
response  in  the  whole  organism. 

Similar  considerations  can  apply  to 
studies  of  physical  dependence;  that  is, 
it  is  important  to  consider  the  under- 
lying mechanisms  of  withdrawal  signs 
and  symptoms  when  investigating  cel- 
lular or  neurochemical  changes  that 
may  be  responsible  for  these  signs. 
The  glutamatergic  and  GABA  systems 
may  well  play  important  roles  in  gen- 
erating alcohol  withdrawal  hyperex- 
citability  and  convulsions,  but  not  in 
other  withdrawal  signs  or  symptoms 
such  as  autonomic  disturbances. 

These  considerations  raise  the  issue  of 
the  relevance  of  studies  of  the  chronic 
effects  of  ethanol  on  neurons  in  culture. 
In  such  systems,  no  behavioral  correlates 
can  be  assessed,  and  "adaptation"  is 
defined  entirely  on  the  basis  of 
changes  in  response  to  ethanol  of  an 
in  vitro  system,  or  changes  that  occur 
after  alcohol  has  been  removed  from  the 
system  (withdrawal).  While  these  sys- 
tems can  be  extremely  useful  for  assay- 
ing biochemical  and  molecular  effects 


148 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


of  ethanol,  a  careful  analysis  of  the 
relationship  of  the  in  vitro  system  to 
the  in  vivo  situation  is  required.  For 
example,  primary  neuronal  cultures 
are  often  used  to  assess  ethanol 
effects,  but  these  neurons  are  in  the 
developmental  stage,  and  results 
obtained  with  these  systems  may  not 
reflect  effects  of  ethanol  in  the  adult 
brain.  Similarly,  transformed  neurons 
may  have  different  characteristics  from 
native  neurons.  In  all  in  vitro  systems, 
many  of  the  interconnections  among 
neurons  may  be  lost,  resulting  in  mis- 
leading conclusions.  On  the  other 
hand,  if  the  cell  culture  system  can  be 
shown  to  have  characteristics  of  a 
neuronal  system  in  the  adult  brain, 
the  neuronal  culture  may  provide  an 
excellent  model  to  investigate  adaptive 
responses  to  ethanol  that  may  be 
extrapolatable  to  the  behaving  animal. 

Application  of  Simple 
Models  That  Have  Provided 
Information  About  Learning 
and  Memory 

As  discussed  earlier,  invertebrate  systems 
have  been  used  extensively  to  study  learn- 
ing and  memory  processes,  and  these 
animals  could  also  be  used  to  advantage 
to  study  adaptation  to  alcohol  and  other 
drugs.  The  advantage  of  the  invertebrate 
systems  is  their  relative  simplicity;  that  is, 
only  a  limited  number  of  neurons  may 
be  involved  in  the  measured  behaviors. 
These  systems  can  be  investigated  both  in 
vivo  and  in  vitro,  but  in  contrast  to  the 
cell  cultures  alluded  to  in  the  previous 
section,  it  may  be  easier  to  demonstrate 
the  similarity  of  the  cell  culture  system  to 
the  intact  animal.  As  illustrated  by  the 
work  of  Kandel  and  his  colleagues,  the 


relative  simplicity  of  the  Aplysia  system 
has  allowed  for  a  detailed  analysis  of 
the  cellular  and  molecular  mechanisms 
involved  in  the  adaptations  that  underlie 
learning  and  memory,  and  these  mecha- 
nisms also  appear  to  play  a  role  in  adap- 
tation in  the  mammalian  brain.  The 
introduction  of  ethanol  into  such  systems 
would  provide  a  model  in  which  toler- 
ance and  physical  dependence,  as  well  as 
sensitization,  might  be  induced,  and  the 
pathways  leading  to  these  adaptive 
changes  could  be  determined  in  the  man- 
ner described  for  the  studies  of  learning 
and  memory.  It  is  interesting  to  note  that 
ethanol  has  been  shown  to  accelerate 
the  rate  of  decay  of  posttetanic  potentia- 
tion at  an  identified  synapse  in  a  ganglion 
preparation  from  Aplysia,  and  that  resis- 
tance to  this  effect  of  ethanol  occurs 
rapidly  after  ethanol  exposure  (Barondes 
et  al.  1979).  Although  high  concentra- 
tions of  ethanol  were  used  in  these  par- 
ticular studies,  this  type  of  system  has  the 
potential  to  provide  significant  infor- 
mation about  the  pathways  involved  in 
ethanol-induced  neuroadaptation.  More 
recently,  studies  of  ethanol  sensitivity 
and  tolerance  in  Drosophila  have  begun 
to  illustrate  the  use  of  simpler,  inverte- 
brate models  in  alcohol  research 
(Moore  et  al.  1998). 

More  Complex  Cellular 
Models  for  Correlations  of 
Physiology  and  Behavior 

Other,  less  simple  models  also  offer 
advantages  in  the  search  for  behaviorally 
meaningful  neuroadaptations  to 
ethanol.  Earlier  in  this  chapter  we  dis- 
cussed the  effects  of  chronic  ethanol 
on  hippocampal  LTP,  which  has  been 
much  discussed  as  a  cellular  correlate  of 


149 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


spatial  memory  and  learning  (see,  e.g., 
Stevens  1998).  In  fact,  this  is  just  one 
form  of  synaptic  plasticity  that  may  be 
altered  by  chronic  ethanol  and  could 
provide  a  mechanism  underlying  or 
related  to  tolerance.  Other  forms 
include  posttetanic  potentiation,  short- 
term  potentiation,  and  long-term 
depression  (LTD)  of  synaptic  transmis- 
sion. These  measures  may  be  extracellu- 
larly  or  intracellularly  recorded,  both  in 
vivo  as  well  as  in  vitro  in  a  slice  prepara- 
tion, and  correlated  with  behavioral 
changes  occuring  in  the  source  animal 
just  prior  to  or  during  the  recordings. 
The  major  considerations  and 
advantages  of  the  brain  slice  plasticity 
models  are  as  follows: 

1.  Mammalian  tissue  may  be  used  for 
closer  modeling  of  human  disor- 
ders. 

2.  Generally  low,  known  concentra- 
tions of  ethanol  and  other  drugs 
may  be  applied. 

3.  Anesthetics  are  not  required. 

4.  Relatively  small  numbers  of  neurons 
can  be  included. 

5.  Ethanol  can  be  chronically  admin- 
istered to  the  animal  followed  by 
later  slice  preparation  and  continued 
incubation  in  ethanol  in  vitro,  for 
subsequent  controlled  withdrawal. 

6.  The  several  mechanisms  (e.g., 
NMD  A  receptor  and  AMPA  recep- 
tor activation,  GABA  receptors, 
Ca++  channels,  kinases)  involved  in 
LTP  and  the  other  forms  of  synaptic 


plasticity  are  precisely  the  ones  most 
sensitive  to  acute  and  chronic  ethanol. 

7.  The  genetically  defined  or  altered 
animals  described  in  the  next  section, 
as  well  as  those  subjected  to  the  newer 
protracted  abstinence  protocols,  can 
be  subjects  for  these  models. 

These  forms  of  synaptic  plasticity  could 
serve  as  ideal  candidates  for  sites  or  mech- 
anisms underlying  some  behavioral  neu- 
roadaptations  occuring  with  ethanol 
preference,  alcohol-seeking  behavior,  or 
alcohol  craving.  Such  multiple  plasticity 
measures  in  transgenic  mice  have  pro- 
vided a  considerable  depth  of  correlates 
for  behavioral  and  neuropathologies 
changes  (see,  e.g.,  Krucker  et  al.  1998). 
Given  the  possible  role  of  the  hippo- 
campus in  spatial  memory,  such  plastic- 
ity changes  could  also  be  highly  relevant 
for  sites  of  adaptations  responsible  for 
spatial  conditioned  cues  related  to  drug- 
seeking  behavior.  Interestingly,  our  pre- 
liminary studies  have  shown  that  the 
anticraving  agent  acamprosate  enhances 
LTP  in  rat  hippocampal  slices  (Madamba 
and  Siggins  manuscript  in  preparation). 
It  also  may  be  relevant  that  several  forms 
of  hippocampal  LTP  and  LTD  have  been 
shown  to  involve  opiate  mechanisms 
(see,  e.g.,  Francesconi  et  al.  1997). 

Brain  slices  of  other  brain  regions  (e.g., 
accumbens,  VTA,  amygdala)  known  to 
be  involved  in  ethanol  preference  and 
ethanol-seeking  behavior,  taken  from 
genetically  manipulated  animals  or  those 
previously  subjected  to  chronic  ethanol 
protocols  (including  forced  abstinence), 
may  also  be  excellent  candidates  for 
elucidation  of  cellular  sites  or  molecular 
mechanisms  underlying  behavioral 


150 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


neuroadaptations  occuring  in  ethanol 
dependence  or  craving.  Ideally,  data  from 
these  slices  should  be  correlated  with 
the  behaviors  expressed  by  the  animals 
from  which  the  slices  were  taken. 

Two  other  quite  different  but  poten- 
tially very  powerful  cellular  models  for 
correlations  to  ethanol-induced  behav- 
ioral neuroadaptations,  discussed  earlier 
in  this  chapter,  are  worth  highlighting 
here:  organotypic  forebrain  cultures 
(Thomas  et  al.  1998)  and  multineuron 
recording  in  freely  moving  animals 
(Woodward  et  al.  1998).  In  both  these 
cases  neural  circuits  could  be  studied 
under  a  variety  of  conditions,  including 
elicitation  of  synaptic  plasticity  and  the 
various  treatment  protocols  for  chronic 
ethanol,  self- administration,  and  pro- 
tracted abstinence.  In  the  case  of  organ- 
otypic cultures,  tissue  from  transgenic  and 
null-mutant  mice  could  be  investigated. 
In  the  case  of  multineuron  recording  from 
freely  moving  animals,  generation  of  the 
various  forms  of  synaptic  plasticity  could 
be  obtained  via  stimulation  electrodes, 
together  with  measures  of  precisely  con- 
trolled and  monitored  behavioral  cor- 
relates (e.g.,  using  learning  and 
memory  paradigms  such  as  the  Y 
maze).  Superimposition  of  drug  treat- 
ments for  the  various  target  systems 
(e.g.,  the  therapeutic  agents  mentioned 
earlier  in  this  chapter,  in  the  section 
Systems  That  Influence  the  Development, 
Maintenance,  and  Loss  of  Tolerance, 
Dependence,  and  Sensitization)  could 
provide  several  more  fruitful  avenues 
of  research  in  both  of  these  models. 

Genetics 

There  is  substantial  evidence  that 
genetic  variation  is  one  determinant  of 


individual  differences  in  neuroadapta- 
tion to  ethanol.  Environmental  variation 
and  genotype-environment  interaction 
likely  also  bear  some  responsibility  for 
individual  variation.  Investigations  of 
genetically  heterogeneous  populations 
are  limited  in  their  ability  to  separate 
genetic  from  environmental  sources  of 
variation.  However,  there  are  a  number 
of  methods  that  can  be  used  to  ascer- 
tain genetic  influence  on  neuroadaptive 
processes.  Much  of  the  work  that  has 
been  done  in  animals  has  relied  on 
inbred  strains,  which  provide  well- 
defined  genetics.  Differences  between 
inbred  strains,  when  environmental 
factors  have  been  carefully  controlled, 
can  be  interpreted  as  genetically  deter- 
mined differences.  Differences  between 
individuals  within  a  strain  must  be  due 
to  nongenetic  factors. 

Selected  lines  provide  another  pow- 
erful tool  to  assess  genetic  influences  on 
ethanol-induced  neuroadaptation.  Work 
done  with  animals  that  display  differ- 
ential sensitivity  to  alcohol  withdrawal 
seizures  (WSP  and  WSR  mice), 
described  earlier  in  this  chapter,  as  well 
as  work  with  a  second  selection,  high 
alcohol  withdrawal  (HW)  and  low  alco- 
hol withdrawal  (LW)  mice  (Hoffman 
et  al.  unpublished  data),  has  suggested 
certain  mechanisms  that  may  underlie 
alcohol  withdrawal  seizures.  In  addition, 
mice  have  been  selectively  bred  for 
differences  in  acute  functional  tolerance 
to  an  incoordinating  effect  of  ethanol 
(high  acute  functional  tolerance  and 
low  acute  functional  tolerance  mice), 
and  these  lines  are  currently  being 
used  for  a  variety  of  studies.  Selected 
lines  do  not  currently  exist  that  have  been 
bred  for  differences  in  ethanol-induced 


151 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


sensitization,  but  FAST  and  SLOW 
mouse  lines,  bred  for  differences  in 
sensitivity  to  the  acute  activating  effects 
of  ethanol,  have  been  found  to  differ 
in  susceptibility  to  ethanol  sensitization 
as  well.  In  theory,  biochemical  differ- 
ences between  selected  lines  of  animals 
are  related  to  the  trait  for  which  they 
have  been  selectively  bred,  and  contin- 
uation of  studies  with  the  selected  lines, 
as  well  as  the  generation  of  lines  selected 
for  differences  in  other  aspects  of  neu- 
roadaptation,  should  be  encouraged. 

Recombinant  inbred  strains  com- 
prise a  powerful  genetic  animal  model 
appropriate  for  gene  mapping  of  alco- 
hol neuroadaptation.  For  example,  the 
BXD/Ty  recombinant  inbred  strains 
are  a  set  of  26  inbred  mouse  strains 
produced  from  the  F2  cross  of  the 
C57BL/6J  and  DBA/2J  strains.  They 
are  particularly  useful  for  examination 
of  genetic  correlations  and  for  mapping 
of  genes  affecting  the  measured  pheno- 
type.  The  observation  that  two  traits 
differ  between  two  inbred  strains  does 
not  provide  evidence  of  a  genetic  cor- 
relation. However,  if  many  strains  are 
tested  and  sensitivity  for  trait  one  is 
predictive  of  sensitivity  for  trait  two, 
this  is  suggestive  of  common  genetic 
mediation  of  the  two  traits. 

In  large  part,  the  traits  in  question 
are  under  polygenic  control:  more  than 
one  gene  contributes  to  the  magnitude 
of  the  trait.  Quantitative  trait  loci  are 
genes  whose  collective  effects  con- 
tribute to  the  determination  of  such 
traits.  The  goal  in  behavioral  genetic 
studies  of  the  constituents  of  alcohol 
dependence  is  gene  identification  and, 
ultimately,  gene  cloning,  to  provide  a 
specific  target  for  disease  intervention. 


QTL  analysis  represents  one  step  in  a 
multistep  approach  to  gene  identifica- 
tion. Some  QTL  mapping  work  has 
been  performed  for  phenotypes  like 
tolerance,  withdrawal,  and  sensitization, 
but  additional  work  is  needed  to  convert 
provisional  markers  from  recombinant 
inbred  studies  to  confirmed  linkages. 

Certain  sophisticated  genetic  prepa- 
rations are  beginning  to  be  utilized  in 
alcohol  research  and  should  be  applied 
to  studies  of  neuroadaptation.  Congenic 
strain  production  through  repetitive 
backcrossing  permits  the  creation  of 
animals  that  are  genetically  segregating 
at  only  one  genetic  locus  that  has  been 
targeted.  Such  congenic  strains  can  be 
produced  by  direct  genotyping  to 
provide  further  verification  of  QTLs 
and  to  facilitate  the  identification  of 
important  functional  genes. 

New  or  altered  genes  can  be  stably 
introduced  into  the  mouse  genome  by 
the  use  of  transgenic  technology, 
which  is  now  readily  available.  These 
genes  can  be  expressed  in  all  tissues  or 
targeted  to  particular  organs  or  tissues, 
such  as  brain.  The  extension  of  this 
technology  to  the  study  of  behavior  is 
growing,  and  it  can  certainly  be  used  to 
study  the  role  of  specific  genes,  sug- 
gested by  past  investigations,  in  neuro- 
adaptation to  ethanol.  The  production 
of  knockout  mice  involves  homologous 
recombination  in  embryonic  stem 
cells  combined  with  the  generation  of 
chimeric  mice.  These  chimeric  mice 
can  then  be  bred  to  an  inbred  strain, 
and  the  offspring  genotyped  for  the 
presence  of  the  mutation.  In  some  cases, 
knockout  of  a  gene  critical  to  develop- 
ment can  mean  death  to  the  recipient. 
However,  techniques  have  now  been 


152 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


devised  to  permit  creation  of  condi- 
tional knockouts  in  which  a  gene  can  be 
eliminated  in  specific  classes  of  cells. 
Inducible  knockout  procedures  are  also 
being  developed  to  circumvent  prob- 
lems associated  with  knockout  of  a  gene 
during  development.  There  is  infinite 
opportunity  to  develop  transgenic  and 
knockout  mice  targeting  any  number 
of  neurochemical  processes  that  might 
be  expected  to  be  involved  in  neuroad- 
aptation to  ethanol. 

Application  of  Findings 
at  the  Cellular,  Biochemical, 
and  Molecular  Levels  to  the 
Development  of  Effective 
Intervention  Strategies 

In  order  to  achieve  a  level  of  knowledge  to 
develop  rational  strategies  for  inter- 
vention, it  is  important  to  understand 
mechanisms  that  underlie  neuroadaptive 
responses  to  ethanol.  Once  mechanisms 
are  understood,  potential  intervention 
strategies  include  pharmacotherapy  and 
genetic  therapies.  Gene  therapy  has 
emerged  as  both  a  novel  treatment 
modality  and  a  powerful  tool  for  basic 
science  investigations.  Viral  vectors 
can  transfer  and  express  foreign  genes 
in  a  wide  variety  of  nondividing  mam- 
malian cells,  and  in  the  case  of  adeno- 
associated  virus  (AAV)  vectors,  studies 
have  demonstrated  stable,  long-term, 
nontoxic  gene  expression  in  brain 
(McCown  et  al.  1996;  Xiao  et  al.  1997). 
Studies  are  needed  that  will  utilize 
gene  transfer  techniques  to  probe  ques- 
tions central  to  ethanol's  pathological 
actions.  However,  before  this  goal 
becomes  a  reality,  many  studies  are  needed 
to  refine  the  technology  of  gene  delivery. 
Vectors  must  be  designed  that  allow 


long-term  expression  throughout  brain 
without  producing  toxic  side  effects. 
Inducible  vectors — for  example,  vec- 
tors that  can  be  turned  on  by  adminis- 
tration of  exogenous  agents — could 
be  highly  advantageous  for  therapeu- 
tic applications.  These  studies  are  very 
promising  and  well  worth  the  invest- 
ment for  the  promise  of  future  thera- 
peutic applications. 

ACKNOWLEDGMENTS 

Portions  of  this  chapter  were  adapted 
from  the  following  reviews:  Tabakoff 
and  Hoffman  1992,  Morrow  1995, 
Tabakoff  and  Hoffman  1996£, 
Grobin  et  al.  1998,  and  Tabakoff  and 
Hoffman  1998. 

REFERENCES 

Abi-Dargham,  A.;  Gandleman,  M.;  Zoghbi, 
S.S.;  Laruelle,  M.;  Baldwin,  KM.;  Randall, 
P.;  Zea-Ponce,  Y.;  Charney,  D.S.;  Hoofer, 
P.B.;  and  Innis,  KB.  Reproducibility  of 
SPECT  measurement  of  benzodiazepine 
receptors  in  human  brain  with  iodine-123- 
lomazcnti.  J  Nucl  Med  36:167-175,  1995. 

Abraham,  W.C.,  and  Hunter,  B.E.  An 
electrophysiological  analysis  of  chronic 
ethanol  neurotoxicity  in  the  dentate 
gyrus:  Distribution  of  entorhinal  afferents. 
Exp  Brain  Res  47:61-68,  1982. 

Abraham,  W.C.;  Manis,  P.B.;  Hunter, 
B.E.;  Zornetzer,  S.F.;  and  Walker,  D.W. 
Electrophysiological  analysis  of  synaptic 
distribution  in  CA1  of  rat  hippocampus  after 
chronic  ethanol  exposure.  Brain  Res 
237:91-105,  1982. 

Acquas,  E.;  Meloni,  M.;  and  Di  Chiara, 
G.  Blockade  of  6-opioid  receptors  in  the 
nucleus  accumbens  prevents  ethanol- 


153 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


induced  stimulation  of  dopamine  release. 
Eur  J  Pharmacol  230:239-241,  1993. 

Ahern,  K.;  Lustig,  U.S.;  and  Greenberg, 
D.A.  Enhancement  of  NMDA  toxicity 
and  calcium  responses  by  chronic  expo- 
sure of  cultured  cortical  neurons  to 
ethanol.  Neurosci Lett  165:211-214, 
1994. 

Alberini,  CM.;  Ghirardi,  M.;  Metz,  R.; 
and  Kandel,  E.R  C/EBP  is  an  immediate- 
early  gene  required  for  the  consolidation 
of  long-term  facilitation  in  Aplysia.  Cell 
76:1099-1114, 1994. 

Alkon,  D.L.,  and  Nelson,  T.J.  Specificity 
of  molecular  changes  in  neurons  involved 
in  memory  storage.  FASEB  J  4:1567- 
1576,  1990. 

Allan,  A.M.,  and  Harris,  R.A.  Gamma- 
aminobutyric  acid  and  alcohol  actions:  Neu- 
rochemical studies  of  long  sleep  and  short 
sleep  mice.  Life  Sci  39:2005-2015,  1986. 

Allan,  A.M.,  and  Harris,  RA.  Acute  and 
chronic  ethanol  treatments  alter  GABA 
receptor-operated  chloride  channels.  Pharm- 
acol Biochem  Behav  27:665-670,  1987. 

Allan,  A.M.;  Burnett,  D.;  and  Harris, 
R.A.  Ethanol-induced  changes  in  chloride 
flux  are  mediated  by  both  GABAA  and 
GABAB  receptors.  Alcohol  Clin  Exp  Res 
15:233-237,  1991. 

Altshuler,  H.L.;  Philips,  P.E.;  and 
Feinhandler,  D.E.  Alteration  of  ethanol 
self- administration  by  naltrexone.  Life  Sci 
26:679-688,  1980. 

American  Psychiatric  Association.  Diagnostic 
and  Statistical  Manual  of  Mental 
Disorders.  4th  ed.  Washington,  DC:  the 
Association,  1994. 

Andreae,  L.C.,  and  Herbert,  J.  Expression 
of  c-fos'm  restricted  areas  of  the  basal 
forebrain  and  brainstem  following  single 


or  combined  intraventricular  infusions  of 
vasopressin  and  corticotropin-releasing 
factor.  Neuroscience  53:735-748,  1993. 

Badawy,  A.B.,  and  Evans,  M.  Opposite 
effects  of  chronic  administration  and  sub- 
sequent withdrawal  of  drugs  of  dependence 
on  the  metabolism  and  disposition  of  endo- 
genous and  exogenous  tryptophan  in  the 
rat.  Alcohol  Alcohol  18:369-382,  1983. 

Bailey,  C.H.;  Bartsch,  D.;  and  Kandel, 
E.R.  Toward  a  molecular  definition  of 
long-term  memory  storage.  Proc  Natl 
Acad  Sci  U&4  93:13445-13452,  1996. 

Ballenger,  J.C.,  and  Post,  R.M.  Kindling 
as  a  model  for  alcohol  withdrawal  syn- 
dromes. Br  J  Psychiatry  133:1-14, 1978. 

Barondes,  S.H.;  Traynor,  M.E.;  Schlapfer, 
W.T.;  and  Woodson,  P.B.J.  Rapid  adapta- 
tion to  neuronal  membrane  effects  of 
ethanol  and  low  temperature:  Some  spec- 
ulations on  mechanism.  Drug  Alcohol 
Depend  4:155-166,  1979. 

Becker,  H.C.,  and  Hale,  R.L.  Repeated 
episodes  of  ethanol  withdrawal  potentiate 
the  severity  of  subsequent  withdrawal 
seizures:  An  animal  model  of  alcohol 
withdrawal  "kindling."  Alcohol  Clin  Exp 
Res  17:94-98,  1993. 

Beckmann,  A.M.;  Matsumoto,  I.;  and 
Wilce,  P.A.  AP-1  and  Egr  DNA- binding 
activities  are  increased  in  rat  brain  during 
ethanol  withdrawal.  J  Neurochem  69:306- 
314,  1997. 

Benjamin,  D.;  Grant,  E.R;  and 
Pohorecky,  L.A.  Naltrexone  reverses  the 
ethanol-induced  dopamine  release  in  the 
nucleus  accumbens  in  awake,  freely  mov- 
ing rats.  Brain  Res  621: 137-140,  1993. 

Benkelfat,  C.;  Murphy,  D.L.;  Hill,  J.L.; 
George,  D.T.;  Nutt,  D.;  and  Linnoila,  M. 
Ethanollike  properties  of  the  serotonergic 
partial  agonist  m-chlorophenylpiperazine 


154 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


in  chronic  alcoholic  patients.  Arch  Gen 
Psychiatry  48:383,  1991. 

Berry,  M.S.,  and  Pentreath,  V.W.  The 
neurophysiology  of  alcohol.  In:  Sandler, 
M.,  ed.  Psychopharmacology  of  Alcohol. 
New  York:  Raven  Press,  1980.  p.  43. 

Berton,  F.;  Francesconi,  W.;  Madamba, 
S.M.;  Zieglgansberger,  W.;  and  Siggins, 
G.K  Acamprosate  enhances  AT-methyl-D- 
aspartate  receptor-mediated  neurotrans- 
mission but  inhibits  presynaptic  GABAB 
receptors  in  nucleus  accumbens  neurons. 
Alcohol  Clin  Exp  £^22:183-191,  1998. 

Bettler,  B.,  and  Mulle,  C.  Neurotransmitter 
receptors.  II.  AMPA  and  kainate  receptors. 
Neuropharmacology  34:123-139,  1995. 

Bien,  T.H.,  and  Burge,  K  Smoking  and 
drinking:  A  review  of  the  literature.  IntJ 
Addict  25:1429-1454,  1990. 

Biggio,  G.;  Cibin,  M.;  Diana,  M.;  Fadda, 
F.;  Ferrara,  S.D.;  Gallimberti,  L.;  Gessa, 
G.L.;  Mereu,  G.P.;  Rossetti,  Z.L.;  and 
Serra,  M.  Suppression  of  voluntary  alcohol 
intake  in  rats  and  alcoholics  by  gamma- 
hydroxybutyric  acid:  A  non-GABAergic 
mechanism.  Adv  Biochem 
Psychopharmacol  47:281-288,  1992. 

Bitran,  M.,  and  Kalant,  H.  Effect  of  ani- 
somycin  on  the  development  of  rapid 
tolerance  to  ethanol-induced  motor  im- 
pairment. Pharmacol  Biochem  Behav 
45:225-228,  1993. 

Blevins,  T.;  Mirshahi,  T.;  Chandler,  L.J.; 
and  Woodward,  J.J.  Effects  of  acute  and 
chronic  ethanol  exposure  on  heteromeric 
N-methyl-D- aspartate  receptors  expressed 
in  HEK  293  cells.  /  Neurochem  69:2345- 
2354,  1997. 

Bokoch,  GM.  Interplay  between  Ras-related 
and  heterotrimeric  GTP  binding  proteins: 
Lifestyles  of  the  BIG  and  little.  FASEBJ 
10:1290-1295,  1996. 


Bourtchuladze,  R.;  Frenguelli,  B.;  Blendy, 
J.;  Cioffi,  D.;  Schultz,  G.;  and  Silva,  A.J. 
Deficient  long-term  memory  in  mice  with 
a  targeted  mutation  of  the  cAMP-respon- 
sive  element- binding  protein.  Cell  79:59- 
68,  1994. 

Brambilla,  R.;  Gnesutta,  N.;  Minischiello, 
L.;  White,  G.;  Roylance,  A. J.;  Herron, 
C.E.;  Ramsey,  M.;  Wolfer,  D.P.;  Cestari, 
V.;  Rossi-Arnaud,  C;  Grant,  S.G.N.; 
Chapman,  P.F.;  Lipp,  H.-P.;  Sturani,  E.; 
and  Klein,  R.  A  role  for  the  Ras  signalling 
pathway  in  synaptic  transmission  and 
long-term  memory.  Nature  390:281- 
286,  1997. 

Breese,  C.R.;  Freedman,  R.;  and  Leonard, 
S.S.  Glutamate  receptor  subtype  expression 
in  human  postmortem  brain  tissue  from 
schizophrenics  and  alcohol  abusers.  Brain 
Res  674:82-90,1995. 

Brennan,  C.H.,  and  Littleton,  J.M.  Second 
messengers  involved  in  genetic  regulation 
of  the  number  of  calcium  channels  in 
bovine  adrenal  chromaffin  cells  in  culture. 
Neuropharmacology  29:689-693,  1990. 

Brennan,  C.H.,  and  Littleton,  J.M. 
Chronic  exposure  to  anxiolytic  drugs, 
working  by  different  mechanisms 
causes  up-regulation  of  dihydropyridine 
binding  sites  on  cultured  bovine  adrenal 
chromaffin  cells.  Neuropharmacology 
30:199-205,1991. 

Brennan,  C.H.;  Crabbe,  J.;  and  Littleton, 
J.M.  Genetic  regulation  of  dihydropyridine - 
sensitive  calcium  channels  in  brain  may 
determine  susceptibility  to  physical  depen- 
dence on  alcohol.  Neuropharmacology 
29:429^32,  1990. 

Britton,  D.K;  Curzon,  P.;  Mackinzie,  KG.; 
Kebabian,  J.W.;  Williams,  J. E.G.;  and 
Kerkman,  D.  Evidence  for  involvement  of 
both  Dl  and  D2  receptors  in  maintaining 


155 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


cocaine  self- administration.  Pharmacol 
Biochem  Behav  39:911-915,  1991. 

Broadbent,  J.;  Grahame,  N.J.;  and 
Cunningham,  C.L.  Haloperidol  prevents 
ethanol-stimulated  locomotor  activity  but 
fails  to  block  sensitization.  Psychopharma- 
cology  (Berl)  120:475-482,  1995. 

Brodie,  M.,  and  Appel,  S.  The  effects  of 
ethanol  on  dopaminergic  neurons  of  the 
ventral  tegmental  area  studied  with  intra- 
cellular recording  in  brain  slices.  Alcohol 
Clin  Exp  Res 22:236-244,  1998. 

Brodie,  M.S.;  Shefher,  S.A.;  and  Dunwiddie, 
T.V.  Ethanol  increases  the  firing  rate  of 
dopamine  neurons  of  the  rat  ventral  tegmen- 
tal area  in  vitro.  Brain  Res  508:65-69, 1990. 

Brodie,  M.S.;  Trifunovic,  R.D.;  and 
Shefner,  S.A.  Serotonin  potentiates 
ethanol-induced  excitation  of  ventral 
tegmental  area  neurons  in  brain  slices 
from  three  different  rat  strains.  / 
Pharmacol  Exp  Ther  273:1139-1146, 1995. 

Browning,  M.D.;  Bureau,  M.;  Dudek, 
E.M.;  and  Olsen,  R.W.  Protein  kinase  C 
and  cAMP-dependent  protein  kinase 
phosphorylate  the  f3  subunit  of  the  puri- 
fied Y-aminobutyric  acid  A  receptor.  Proc 
Natl  Acad  Sci  USA  87:1315-1318,  1990. 

Buck,  KJ.,  and  Harris,  RA.  Benzodiazepine 
agonist  and  inverse  agonist  actions  on  GABAA 
receptor-operated  chloride  channels.  / 
Pharmacol  Exp  Ther  253:713-719,  1990. 

Buck,  K.J.;  Hahner,  L.;  Sikela,  J.;  and 
Harris,  R.A.  Chronic  ethanol  treatment 
alters  brain  levels  of  gamma-aminobutyric 
acidA  receptor  subunit  mRNAs:  Relationship 
to  genetic  differences  in  ethanol  withdrawal 
seizure  severity.  J  Neurochem  57:1452- 
1455,  1991. 

Buck,  K.J.;  Metten,  P.;  Belknap,  J.K.;  and 
Crabbe,  J.C.  Quantitative  trait  loci  involved 
in  genetic  predisposition  to  acute  alcohol 


withdrawal  in  mice.  J  Neurosci  17:3946- 
3955,  1997. 

Buckner,  M.K.;  Rossner,  S.;  and  Arendt, 
T.  Differential  changes  in  the  expression 
of  AMPA  receptors  genes  in  rat  brain 
after  chronic  exposure  to  ethanol:  An  in 
situ  hybridization  study.  J  Hirnforsch 
38:369-376,  1997. 

Burger,  L.Y.,  and  Martin- Iverson,  M.T. 
Increased  occupation  of  Dl  and  D2 
dopamine  receptors  accompanies  cocaine- 
induced  behavioral  sensitization.  Brain 
Res  639:228-232,  1994. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin, 
C.  Hormonal,  psychological,  and  alcohol 
craving  changes  after  m-chlorophenyl- 
piperazine  administration  in  alcoholics. 
Alcohol  Clin  Exp  Res  21 :220-226,  1997. 

Byers,  D.;  Davis,  R.L.;  and  Kiger,  J.A.,  Jr. 
Defect  in  cyclic  AMP  phosphodiesterase 
due  to  the  dunce  mutation  of  learning  in 
Drosophila  melanog aster.  Nature  289:79- 
81,  1981. 

Caine,  S.B.,  and  Koob,  G.F.  Modulation 
of  cocaine  self- administered  in  the  rat 
through  D-3  dopamine  receptors.  Science 
260:1814-1816,  1993. 

Calkin,  P.A.,  and  Barnes,  E.M.,  Jr. 
Gamma-aminobutyric  acid-A  (GABAA) 
agonists  down-regulate  GABAA/benzodi- 
azepine  receptor  polypeptides  from  the 
surface  of  chick  cortical  neurons.  ]  Biol 
Chem  269:1548-1553,  1994. 

Camacho-Nasi,  P.,  and  Treistman,  S.N. 
Ethanol-induced  reduction  of  neuronal 
calcium  currents  in  Aplysia:  An  examina- 
tion of  possible  mechanisms.  Cell  Mol 
Neurobiol  7:191-207,  1987. 

Campanelli,  C;  Le,  A.D.;  Khanna,  J.M.; 
and  Kalant,  H.  Effect  of  raphe  lesions  on 
the  development  of  acute  tolerance  to 


156 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


ethanol  and  pentobarbital.  Psychopharma- 
cology  (Berl)  96A54-A57,  1988. 

Campbell,  K.P.;  Leung,  A.T.;  and  Sharp, 
A.H.  The  biochemistry  and  molecular 
biology  of  the  dihydropyridine- sensitive 
calcium  channel.  Trends  Neurosci  11:425- 
430, 1988. 

Carlen,  P.L.;  Gurevich,  N.;  and  Durand, 
D.  Ethanol  in  low  doses  augments  calcium 
mediated  mechanisms  measured  intracel- 
lularly  in  hippocampal  neurons.  Science 
215:306-309,  1982. 

Carlen,  P.L.;  Gurevich,  N.;  Davies,  M.F.; 
Blaxter,  T.J.;  and  O'Beirne,  M.  Enhanced 
neuronal  K+  conductance:  A  possible  common 
mechanism  for  sedative-hypnotic  drug 
action .  Can  J  Physiol  Pharmacol  63:831- 
837,  1985. 

Carter,  L.A.;  Belknap,  J.K.;  Crabbe,  J.C.; 
and  Janowsky,  A.  Allosteric  regulation  of 
the  i^-methyl-D-aspartate  receptor-linked 
ion  channel  complex  and  effects  of 
ethanol  in  ethanol-withdrawal  seizure- 
prone  and  -resistant  mice.  J  Neurochem 
64:213-219,  1995. 

Catterall,  W.A.;  Seagar,  M.J.;  and  Takahashi, 
M.  Molecular  properties  of  the  dihydropy- 
ridine-sensitive  calcium  channel.  J  Biol 
Chem  263:3535-3538,  1988. 

Cebers,  G.;  Hou,  Y.-N.;  Cebere,  A;  Terenius, 
L.;  and  Liljequist,  S.  Chronic  ethanol 
enhances  NMDA-induced  AP- 1  activity 
in  cultured  rat  cerebellar  granule  cells. 
Neuroreport  8:217-220,  1996. 

Celentano,  J.J.;  Gibbs,  T.T.;  and  Farb, 
D.H.  Ethanol  potentiates  GABA-  and 
glycine -induced  chloride  currents  in  chick 
spinal  cord  neurons.  Brain  Res 455: 
377-380, 1988. 

Cepeda,  C;  Buchwald,  N.A.;  and  Levine, 
M.S.  Neuromodulatory  actions  of  dopamine 
in  the  neostriatum  are  dependent  upon 


the  excitatory  amino  acid  receptor  sub- 
types activated.  Proc  Natl  Acad  Sci  USA 
90:9576-9580,  1993. 

Chandler,  L.J.;  Newsom,  H.;  Sumners, 
C;  and  Crews,  F.  Chronic  ethanol  expo- 
sure potentiates  NMDA  excitotoxicity  in 
cerebral  cortical  neurons.  J  Neurochem 
60:1578-1581,  1993^. 

Chandler,  L.J.;  Sumners,  C;  and  Crews, 
F.T.  Ethanol  inhibits  NMDA  receptor- 
mediated  excitoxicity  in  rat  primary  neu- 
ronal cultures.  Alcohol  Clin  Exp  Res 
17:54-60,  \99Zb. 

Chandler,  L.J.;  Sutton,  G.;  Norwood,  D.; 
Sumners,  C;  and  Crews,  F.  Chronic 
ethanol  increases  N-methyl-D-aspartate- 
stimulated  nitric  oxide  formation  but  not 
receptor  density  in  cultured  cortical  neu- 
rons. Mol  Pharmacol  51:733-740,  1997. 

Charlton,  M.E.;  Sweetnam,  P.M.;  Fitzgerald, 
L.W.;  Terwilliger,  R.Z.;  Nestler,  E.J.;  and 
Duman,  KS.  Chronic  ethanol  administra- 
tion regulates  the  expression  of  GABAA 
receptor  al  and  a5  subunits  in  the  ven- 
tral tegmental  area  and  hippocampus.  / 
Neurochem  68:121-127,  1997. 

Charness,  M.E.  Brain  lesions  in  alcoholics. 
Alcohol  Clin  Exp  Res  17:2-1 1,  1993. 

Charness,  M.E.;  Querimit,  L.A.;  and 
Henteleff,  M.  Ethanol  differentially  regu- 
lates G  proteins  in  neural  cells.  Biochem 
Biophys Res  Commun  155:138-143,  1988. 

Chiodo,  L.A.,  and  Berger,  T. W.  Interactions 
between  dopamine  and  amino  acid-induced 
excitation  and  inhibition  in  the  striatum. 
Brain  Res  375:198-203,  1986. 

Choi,  D.W.  Calcium-mediated  neurotoxi- 
city: Relationship  to  specific  channel  types 
and  role  in  ischemic  damage.  Trends 
Neurosci  11:465-469,  1988. 


157 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Choi,  D.W.  Excitotoxic  cell  death.  / 
Neurobiol  23:1261-1276,  1992. 

Clark,  R.B.  Desensitization  of  hormonal 
stimuli  coupled  to  regulation  of  cyclic 
AMP  levels.  Adv  Cyclic  Nucleotide  Protein 
Phosphorylation  Res  20:155-209,  1986. 

Coe,  I.R.;  Dohrman,  D.P.;  Diamond,  I.; 
and  Gordon,  A.S.  Activation  of  cAMP- 
dependent  protein  kinase  reverses  tolerance 
of  the  nucleoside  transporter  to  ethanol.  / 
Pharmacol  Exp  Ther  276:365-369,  1995. 

Coe,  I.R.  Yao,  L.N.;  Diamond,  L;  and 
Gordon,  A.S.  The  role  of  protein  kinase  C 
in  cellular  tolerance  to  ethanol.  J  Biol 
Chem  271:29468-29472,  1996. 

Collingridge,  G.L.,  and  Lester,  RAJ. 
Excitatory  amino  acid  receptors  in  the 
vertebrate  central  nervous  system. 
Pharmacol  Rev  40:143-210,  1989. 

Collins,  A.C.  The  nicotinic  cholinergic 
receptor  as  a  potential  site  of  ethanol 
action.  In:  Deitrich,  R.A.,  and  Erwin, 
V.G.,  eds.  Pharmacological  Effects  of 
Ethanol  on  the  Nervous  System.  Boca 
Raton,  FL:  CRC  Press,  1996.  p.  95. 

Collins,  A.C;  Burch,  J.B.;  deFiebre, 
CM.;  and  Marks,  M.J.  Tolerance  to  and 
cross  tolerance  between  ethanol  and 
nicotine.  Pharmacol  Biochem  Behav 
29:365-373,  1987. 

Collins,  A.C;  Romm,  E.;  and  Wehner, 
J.M.  Dissociation  of  the  apparent  rela- 
tionship between  nicotine  tolerance  and 
upregulation  of  nicotinic  receptors.  Brain 
Res  Bull  25 :37 '3-379,  1990. 

Collins,  A.C;  Romm,  E.;  Selvaag,  S.; 
Turner,  S.;  and  Marks,  M.J.  A  comparison 
of  the  effects  of  chronic  nicotine  infusion 
on  tolerance  to  nicotine  and  cross-toler- 
ance to  ethanol  in  long-  and  short-sleep 
mice.  ] Pharmacol  Exp  Ther  266:1390- 
1397,  1993. 


Connolly,  C.N.;  Drishek,  B.J.;  McDonald, 
B.J.;  Smart,  T.G.;  and  Moss,  S.J.  Assembly 
and  cell  surface  expression  of  heteromeric 
and  homomeric  Y-aminobutyric  acid  type 
A  receptors.  J  Biol  Chem  271:89-96,  1996. 

Corpechot,  C;  Young,  J.;  Calvel,  M.; 
Wehrey,  C;  Veltz,  J.N.;  Touyer,  C; 
Mouren,  M.;  Prasad,  V.V.K.;  Banner,  C; 
Sjovall,  J.;  Baulieu,  E.E.;  and  Robel,  P. 
Neurosteroids:  3a-Hydroxy-5a-pregnant- 
20-one  and  its  precursors  in  the  brain, 
plasma,  and  steroidogenic  glands  of  male 
and  female  rats.  Endocrinology  133:1003- 
1009,  1993. 

Corso,  T.;  Mostafa,  H.;  Collins,  M.;  and 
Neafsey,  E.  Brain  neuronal  degeneration 
caused  by  episodic  alcohol  intoxication  in 
rats:  Effects  of  nimodipine,  6,7-dinitro- 
quinoxaline-2,3-dione,  and  MK-801. 
Alcohol  Clin  Exp  Res  22:217-224,  1998. 

Crabbe,  J.C.;  Kosobud,  A.;  and  Young, 
E.R.  Genetic  selection  for  ethanol  with- 
drawal severity:  Differences  in  replicate 
mouse  lines.  Life  Sci  3:955-962,  1983. 

Crabbe,  J.C;  Belknap,  J.K.;  and  Buck, 
K.J.  Genetic  animal  models  of  alcohol  and 
drug  abuse.  Science  264:1715-1723,  1994. 

Crawford,  D.,  and  Baker,  T.B.  Alcohol 
dependence  and  taste -mediated  learning 
in  the  rat.  Pharmacol  Biochem  Behav 
16:253-261,  1982. 

Crews,  FT.,  and  Chandler,  L.J.  Excitotoxicity 
and  the  neuropathology  of  ethanol.  In: 
Hunt,  W.A.,  and  Nixon,  S.J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  22.  NIH  Pub. 
No.  93-3549.  Rockville,  MD:  the  Institute, 
1993.  p.  355-371. 

Criado,  J.R.;  Steffensen,  S.C.;  and 
Henriksen,  S.J.  Ethanol  acts  via  the  ventral 


158 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


tegmental  area  to  influence  hippocampal 
physiology.  Synapse  17:84-91,  1994. 

Criado,  J.;  Lee,  R;  Berg,  G.;  and  Henriksen, 
S.  Ethanol  inhibits  single-unit  responses 
in  the  nucleus  accumbens  evoked  by 
stimulation  of  the  basolateral  nucleus  of 
the  amygdala.  Alcohol  Clin  Exp  Res 
21:368-374,  1997. 

Criswell,  H.E.;  Simson,  P.E.;  Johnson, 
K.B.;  and  Breese,  G.R.  Chronic  ethanol 
decreases  the  ability  of  GABA  to  inhibit 
ethanol-sensitive  neurons  in  the  medial 
septum.  Alcohol  Clin  Exp  Res  17:477, 1993. 

Crowell,  C.R;  Hinson,  R.E.;  and  Siegel, 
S.  The  role  of  conditional  drug  responses 
in  tolerance  to  the  hypothermic  effects  of 
ethanol.  Psychopharmacolojyy  (Berl)  73:51- 
54,1981." 

Cunningham,  C.L.  Localization  of  genes 
influencing  ethanol-induced  conditioned 
place  preference  and  locomotor  activity  in 
BXD  recombinant  inbred  mice.  Psycho- 
pharmacology  (Berl)  120:  28-41,  1995. 

Cunningham,  C.L.,  and  Noble,  D. 
Conditioned  activation  induced  by 
ethanol:  Role  in  sensitization  and  condi- 
tioned place  preference.  Pharmacol 
Biochem  Behav  43:307-313,  1992. 

Curran,  T.,  and  Franza,  B.R.,  Jr.  Fos  and 
Jun:  The  API  connection.  Cell  55:395- 
397,  1988. 

Dahchour,  A.;  Quertemont,  E.;  and  De 
Witte,  P.  Taurine  increases  in  the  nucleus 
accumbens  microdialysate  after  acute 
ethanol  administration  to  naive  and 
chronically  alcoholised  rats.  Brain  Res 
735:9-16,  1996. 

Daniell,  L.C.,  and  Leslie,  S.W.  Inhibition 
of  fast  phase  calcium  uptake  and  endoge- 
nous norepinephrine  release  in  rat  brain 
region  synaptosomes  by  ethanol.  Brain 
Res  377:18-28, 1986. 


Dave,  J.R.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Ethanol  withdrawal  seizures  produce 
increased  c-fos  mRNA  in  mouse  brain. 
Mol  Pharmacol  37:367-371,  1990. 

Davidson,  M.D.;  Wilce,  P.;  and  Shanley, 
B.C.  Increased  sensitivity  of  the  hippo- 
campus in  ethanol-dependent  rats  to  toxic 
effect  of  N-methyl-D-aspartic  acid  in  vivo. 
Brain  Res  606(l):5-9,  1993. 

Davis,  W.C.,  and  Ticku,  M.K.  Ethanol 
enhances  [3H]  diazepam  binding  at  the 
benzodiazepine- GABA  receptor  ionophore 
complex.  Mol  Pharmacol  20:287,  1981. 

deFiebre,  CM.,  and  Collins,  A.C.  A 
comparison  of  the  development  of  toler- 
ance to  ethanol  and  cross-tolerance  to 
nicotine  after  chronic  ethanol  treatment 
in  long-  and  short-sleep  mice.  J  Pharmacol 
Exp  Ther  266: 1398-1406,  1993. 

Deher,  K.F.,  and  Fraser,  J.G.  Smoking 
habits  of  alcohol  outpatients.  Int  J  Addict 
2:259-268,  1976. 

Deitrich,  R.A.;  Dunwiddie,  T.V.;  Harris, 
R.A.;  and  Erwin,  V.G.  Mechanism  of 
action  of  ethanol:  Initial  central  nervous 
system  actions.  Pharmacol  Rev  41:489- 
537,  1989. 

Devaud,  L.L.,  and  Morrow,  A.L.  Gender 
influences  the  effects  of  ethanol  depen- 
dence on  GABAA  and  NMDA  receptor 
subunit  peptide  expression  in  rat  cortex 
and  hypothalamus.  Alcohol  Clin  Exp  Res 
2L88A,  1998. 

Devaud,  L.L.;  Purdy,  R.H.;  and  Morrow, 
A.L.  The  neurosteroid,  3a-hydroxy-5ct- 
pregnan-20-one,  protects  against  bicu- 
culline-induced  seizures  during  ethanol 
withdrawal  in  rats.  Alcohol  Clin  Exp  Res 
19:350-355,  1995a. 

Devaud,  L.L.;  Smith,  F.D.;  Grayson,  D.R.; 
and  Morrow,  A.L.  Chronic  ethanol  con- 
sumption differentially  alters  the  expression 


159 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  Y-aminobutyric  acidA  receptor  subunit 
mRNAs  in  rat  cerebral  cortex:  Competitive, 
quantitative  reverse  transcriptase -polymerase 
chain  reaction  analysis.  Mol  Pharmacol 
48:861-868,  1995£. 

Devaud,  L.L.;  Purdy,  R.H.;  Finn,  DA.; 
and  Morrow,  A.L.  Sensitization  of  y- 
aminobutyric  acidA  receptors  to  neuroactive 
steroids  in  rats  during  ethanol  withdrawal. 
]  Pharmacol  Exp  TTkr  278:510-517,  1996. 

Devaud,  L.L.;  Fritschy,  J.-M.;  Sieghart, 
W.;  and  Morrow,  A.L.  Bidirectional  alter- 
ations of  GABAA  receptor  subunit  peptide 
levels  in  rat  cortex  during  chronic  ethanol 
consumption  and  withdrawal.  /  Neurochem 
69:126-130,  1997. 

Devaud,  L.L.;  Fritschy,  J.M.;  and  Morrow, 
A.L.  Influence  of  gender  on  chronic  ethanol- 
induced  alterations  in  GABAA  receptors  in 
rats.  Brain  Res  796:222-230,  1998. 

DeVries,  D.J.;  Johnston,  G.A.R.;  Ward, 
L.C.;  Wilce,  P.A.;  and  Shanley,  B.C.  Effects 
of  chronic  ethanol  inhalation  on  the  en- 
hancement of  benzodiazepine  binding  to 
mouse  brain  membranes  by  GABA. 
Neurochem  Int  10:231-235,  1987. 

de  Wied,  D.  Neuropeptides  in  learning 
and  memory  processes.  Behav  Brain  Res 
83:83-90, 1997. 

Diamond,  I.,  and  Gordon,  A.S.  Cellular 
and  molecular  neuroscience  of  alcoholism. 
Physiol  Rev  77:1-20,  1997. 

Diana,  M.;  Pistis,  M.;  Muntoni,  A.;  Rossetti, 
Z.L.;  and  Gessa,  G.  Marked  decrease  of 
A10  dopamine  neuronal  firing  during 
ethanol  withdrawal  syndrome  in  rats.  Eur 
J  Pharmacol  221:403-404,  1992. 

Diana,  M.;  Pistis,  M.;  Carboni,  S.;  Gessa, 
G.L.;  and  Rossetti,  Z.L.  Profound  decre- 
ment of  mesolimbic  dopaminergic  neu- 
ronal activity  during  ethanol  withdrawal 
syndrome  in  rats:  Electrophysiological 


and  biochemical  evidence.  Proc  Natl 
Acad  Sci  USA  90:7966-7969,  1993. 

Di  Chiara,  G.;  Acquas,  E.;  and  Tanda,  G. 
Ethanol  as  a  neurochemical  surrogate  of 
conventional  reinforcers:  The  dopamine - 
opioid  link.  Alcohol  13:13-17,  1996. 

Dildy-Mayfield,  J.E.,  and  Harris,  RA. 
Ethanol  inhibits  kainate  responses  of 
glutamate  receptors  expressed  in  Xenopus 
oocytes:  Role  of  calcium  and  protein 
kinase  C. / Neurosci  15:3162-3171,  1995. 

Ding,  W.-Q.;  Larsson,  C;  and  Ailing,  C. 
Ethanol  exposure  increases  expression  of 
c-jun  and  junD  in  human  neuroblastoma 
cells.  Neuroreport  7:2191-2195,  1996. 

Dobrunz,  L.E.;  Huang,  E.P.;  and  Stevens, 
C.F.  Very  short-term  plasticity  in  hippocam- 
pal  synapses.  Proc  Natl  Acad  Sci  USA 
94:14843-14847,  1997. 

Dodd,  P.R  Benzodiazepine  binding  sites 
in  alcoholic  cirrhotics:  Evidence  for 
gender  differences.  Metab  Brain  Dis 
10:93-104,  1995. 

Dodd,  P.R.;  Thomas,  G.J.;  Harper,  C.G.; 
and  Kril,  J.J.  Amino  acid  neurotransmitter 
receptor  changes  in  cerebral  cortex  in 
alcoholism:  Effect  of  cirrhosis  of  the  liver. 
J  Neurochem  59:1506-1515,  1992. 

Dohrman,  D.P.;  Diamond,  L;  and  Gordon, 
A.S.  Ethanol  causes  translocation  of 
cAMP-dependent  protein  kinase  catalytic 
subunit  to  the  nucleus.  Proc  Natl  Acad 
Sci  USA  93:10217-10221,  1996. 

Dolin,  S.J.,  and  Little,  H.J.  Are  changes 
in  neuronal  calcium  channels  involved  in 
ethanol  tolerance?  J  Pharmacol  Exp  Ther 
250:985-991,  1989. 

Dolin,  S.J.;  Little,  H.J.;  Hudsith,  M.; 
Pagonis,  C;  and  Littleton,  J.M.  Increased 
dihydropyridine-sensitive  calcium  chan- 
nels in  rat  brain  may  underlie  ethanol 


160 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


physical  dependence.  Neuropharmacology 
26:275-279,  1987. 

Dopico,  A.M.;  Lemos,  J.R;  and  Treistman, 
S.N.  Ethanol  increases  the  activity  of  large 
conductance,  Ca(2+)-activated  K+  chan- 
nels in  isolated  neurohypophysial  termi- 
nals. Mol  Pharmacol  49  AO-48,  1996. 

Dopico,  A.M.;  Anantharam,  V.;  and 
Treistman,  S.N.  Ethanol  increases  the 
activity  of  Ca(++)-dependent  K+  (mslo) 
channels:  Functional  interaction  with 
cytosolic  Ca++.  /  Pharmacol  Exp  Ther 
284:258-268,  1998. 

Dragunow,  M.,  and  Robertson,  HA. 
Kindling  stimulation  induces  c-fos  pro- 
tein(s)  in  granule  cells  of  the  rat  dentate 
gyrus.  Nature  329:441-442,  1987. 

Eberwine,  J.;  Yeh,  H.;  Miyashiro,  K.; 
Cao,  Y.;  Nair,  S.;  Finnell,  R.;  Zettel,  M.; 
and  Coleman,  P.  Analysis  of  gene  expres- 
sion in  single  live  neurons.  Proc  Natl 
AcadSci  USA  89:3010-3014,  1992. 

Ellinor,  P.T.;  Zhang,  J.-F.;  Randall,  A.D.; 
Zhou,  M.;  Schwarz,  T.L.;  Tsien,  RW.;  and 
Home,  WA.  Functional  expression  of  a 
novel  neuronal  voltage-dependent  calcium 
channel.  Nature  363A55-4S8,  1993. 

Erickson,  C.K.,  and  Grahm,  D.T. 
Alteration  of  cortical  and  reticular  acetyl- 
choline release  by  ethanol  in  vivo.  / 
Pharmacol  Exp  Ther  185:583-593,  1973. 

Fadda,  F.;  Argiolas,  A.;  Melis,  M.R.; 
Serra,  G.;  and  Gessa,  G.L.  Differential 
effect  of  acute  and  chronic  ethanol  on 
dopamine  metabolism  in  frontal  cortex, 
caudate  nucleus  and  substantia  nigra.  Life 
Sci  27 ':979-986,  1980. 

Fahlke,  C;  Hansen,  S.;  Engel,  J.A.;  and 
Hard,  E.  Effects  of  ventral  striatal  6-OHDA 
lesions  or  amphetamine  sensitization  on 
ethanol  consumption  in  the  rat.  Pharmacol 
Biochem  Behav  47:345-349,  1994. 


Finn,  DA.,  and  Crabbe,  J.C.  Exploring 
alcohol  withdrawal  syndrome.  Alcohol 
Health  Research  World  21:149-156,  1997. 

Finn,  DA.,  and  Gee,  K.W.  The  influence 
of  estrus  cycle  on  neurosteroid  potency  at 
the  gamma-  aminobutyric  acidA  receptor 
complex.  J  Pharmacol  Exp  Ther  265 : 1 374— 
1379,  1993. 

Follesa,  P.,  and  Ticku,  M.K.  Chronic 
ethanol  treatment  differentially  regulates 
NMDA  receptor  subunit  mRNA  expres- 
sion in  rat  brain.  Mol  Brain  Res  29:99- 
106,  1995. 

Follesa,  P.,  and  Ticku,  M.K.  Chronic 
ethanol-mediated  up-regulation  of  the  N- 
methyl-D- aspartate  receptor  polypeptide 
subunits  in  mouse  cortical  neurons  in  culture. 
J  Biol  Chem  271:13297-13299,  1996. 

Francesconi,  W.;  Berton,  F.;  Demuro,  A.; 
Madamba,  S.G.;  and  Siggins,  G.R. 
Naloxone  blocks  long-term  depression  of 
excitatory  transmission  in  rat  CA1  hip- 
pocampus in  vitro.  Arch  Ital  Biol 
135:274-286,  1997. 

Freund,  G.,  and  Anderson,  K.J.  Glutamate 
receptors  in  the  frontal  cortex  of  alcoholics. 
Alcohol  Clin  Exp  Res  20:1 165-1 172, 1996. 

Freund,  G.,  and  Ballinger,  W.E.,  Jr. 
Decrease  of  benzodiazepine  receptors  in 
frontal  cortex  of  alcoholics.  Alcohol 
5:275-282,  1988. 

Freund,  R.K.,  and  Palmer,  M.R  Beta 
adrenergic  sensitization  of  gamma- 
aminobutyric  acid  receptors  to  ethanol 
involves  a  cyclic  AMP/protein  kinase  A 
second-messenger  mechanism.  J  Pharmacol 
Exp  Ther  280:1 192-1200,  1997. 

Gallimberti,  L.;  Ferri,  M.;  Ferrara,  S.D.; 
Fadda,  F.;  and  Gessa,  G.L.  Gamma- 
hydroxybutyric  acid  in  the  treatment  of 
alcohol  dependence:  A  double-blind  study. 
Alcohol  Clin  Exp  Res  16:673-676,  1992. 


161 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Gatto,  G.J.;  McBride,  W.J.;  Murphy, 
J.M.;  Lumeng,  L.;  and  Li,  T.-K.  Ethanol 
self-infusion  into  the  ventral  tegmental 
area  by  alcohol-preferring  (P)  rats. 
Alcohol  11:557-564,  1994. 

Gazzaley,  A.H.;  Siegel,  S.J.;  Kordower, 
J.H.;  Mufson,  E.J.;  and  Morrison,  J.H. 
Circuit-specific  alterations  of  N-methyl- 
D- aspartate  receptor  subunit  1  in  the 
dentate  gyrus  of  aged  monkeys.  Proc  Natl 
AcadSci  USA  93:3121-3125,  1996. 

George,  S.R.;  Roldan,  L.;  Lui,  A.;  and 
Naranjo,  C.A.  Endogenous  opioids  are 
involved  in  the  genetically  determined 
high  preference  for  ethanol  comsumption. 
Alcohol  Clin  Exp  Res  15:668-672,  1991. 

Gessa,  G.L.,  and  Gallimberti,  L.  Gamma- 
hydroxybutyric  acid  in  the  treatment  of 
alcohol  dependence.  Clin 
Neuropharmacol  15:303A-304A,  1992. 

Gessa,  G.L.;  Muntoni,  F.;  Collu,  M.; 
Vargiu,  L.;  and  Mereu,  G.  Low  doses  of 
ethanol  activate  dopaminergic  neurons  of 
the  ventral  tegmental  area.  Brain  Res 
348:201-204,  1985. 

Gilman,  S.;  Koeppe,  R.A.;  Adams,  K.; 
Johnson-Greene,  D.;  Junck,  L.;  Kluin, 
K.J.;  Brunberg,  J.;  Martorello,  S.;  and 
Lohman,  M.  Positron  emission  tomo- 
graphic studies  of  cerebral  benzodiazepine  - 
receptor  binding  in  chronic  alcoholics. 
Ann  Neurol 40:163-171,  1996. 

Giri,  P.R.;  Dave,  J.R;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Arginine  vasopressin  in- 
duces the  expression  of  c- fos  in  the  mouse 
septum  and  hippocampus.  Mol  Brain  Res 
7:131,  1990. 

Givens,  B.;  Williams,  J.;  and  Gill,  T. 
Cognitive  correlates  of  single  neuron 
activity  in  task-performing  animals: 
Application  to  ethanol  research.  Alcohol 
Clin  Exp  Res  22:23-31,  1998. 


Goda,  Y.,  and  Stevens,  C.F.  Readily  re- 
leasable  pool  size  changes  associated  with 
long  term  depression.  Proc  Natl  Acad  Sci 
USA  95:1283-1288,  1998. 

Goeders,  N.E.,  and  Smith,  J.E.  Intracranial 
cocaine  self- administration  into  the  medial 
prefrontal  cortex  increases  dopamine 
turnover  in  the  nucleus  accumbens.  / 
Pharmacol  Exp  Ther 265:592-600,  1993. 

Goldstein,  A.,  and  Goldstein,  D.B.  Enzyme 
expansion  theory  of  drug  tolerance  and 
physical  dependence.  Res  Publications  Assoc 
Res  NervMentDis  46:265-267,  1968. 

Gongwer,  M.A.;  Murphy,  J.M.;  McBride, 
W.J.;  Lumeng,  L.;  and  Li,  T.K.  Regional 
brain  contents  of  serotonin,  dopamine 
and  their  metabolites  in  the  selectively 
bred  high-  and  low-alcohol  drinking  lines 
of  rats.  Alcohol  6:317-320,  1989. 

Gonzales,  R.A.,  and  Weiss,  F.  Suppression 
of  ethanol-reinforced  behavior  by  naltrex- 
one is  associated  with  attenuation  of  the 
ethanol-induced  increase  in  dialysate 
dopamine  levels  in  the  nucleus  accumbens. 
JNeurosci  18:10663-10671,  1998. 

Gonzalez,  L.P.  Sensitivity  to  strychnine 
seizures  is  unaltered  during  ethanol  with- 
drawal. Alcohol  Clin  Exp  Res  17:1029- 
1034,  1993. 

Gonzalez,  L.P.,  and  Czachura,  J.F. 
Reduced  behavioral  responses  to  intrani- 
gral  muscimol  following  chronic  ethanol. 
Physiol  Behav  46:473-477,  1989. 

Grant,  A.J.;  Koski,  G.;  and  Treistman, 
S.N.  Effect  of  chronic  ethanol  on  calcium 
currents  and  calcium  uptake  in  undiffer- 
entiated PC12  cells.  Brain  Res  600:280- 
284,  1993. 

Grant,  K.A.,  and  Barrett,  J.E.  Blockade  of 
the  discriminative  stimulus  and  anxiolytic 
effects  of  ethanol  with  5-HT3-receptor 
antagonists.  In:  Racagni,  G.;  Brunells,  N.; 


162 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


and  Fukuyda,  T.,  eds.  Biological  Psychiatry. 
Vol.  2.  New  York:  Elsevier,  1991.  pp.  11-13. 

Grant,  K.A.,  and  Colombo,  G.  Substitution 
of  the  5-HT1  agonist  trifluoromethyl- 
phenylpiperazine  (TFMPP)  for  the  dis- 
criminative stimulus  effects  of  ethanol: 
Effect  of  training  dose.  Psychopharmacology 
(Berl)  113:26-30,  1993. 

Grant,  K.A.;  Valverius,  P.;  Hudspith,  M.; 
and  Tabakoff,  B.  Ethanol  withdrawal 
seizures  and  the  NMDA  receptor  complex. 
Eur  J  Pharmacol  176:289-296,  1990. 

Griffiths,  RR;  Bigelow,  G.E.;  and 
Leibson,  I.  Facilitation  of  human  tobacco 
self- administration  by  ethanol.  J  Exp 
Anal  Behav  25:279-292,  1976. 

Grigorenko,  E.V.,  and  Yeh,  H.H. 
Expression  profiling  of  GABAA  receptor 
beta-subunits  in  the  rat  retina.  Vis 
Neurosci  11:379-387,  1994. 

Grobin,  A.C.;  Matthews,  O.B.;  Devaud, 
L.L.;  and  Morrow,  A.L.  The  role  of 
GABAA  receptors  in  the  acute  and  chronic 
effects  of  ethanol.  Psychopharmacology 
(Berl)  139:2-19,  1998. 

Gulliver,  S.B.;  Rohsenow,  D.J.;  Colby, 
S.M.;  Dey,  A.M.;  Abrams,  D.B.;  Niaura, 
R.S.;  and  Monti,  P.M.  Interrelationship 
of  smoking  and  alcohol  dependence,  use 
and  urges  to  use.  J  Stud  Alcohol  56:202- 
206,  1995. 

Gulya,  K.;  Grant,  K.A.;  Valverius,  P.; 
Hoffman,  P.L.;  and  Tabakoff,  B.  Brain 
regional  specificity  and  time  course  of 
changes  in  the  NMDA  receptor-ionophore 
complex  during  ethanol  withdrawal. 
Brain  Res  547:129-134, 1991. 

Guppy,  L.J.,  and  Littleton,  J.M.  Binding 
characteristics  of  the  calcium  channel 
antagonist  [3H] -nitrendipine  in  tissues 
from  ethanol- dependent  rats.  Alcohol 
Alcohol  29:283-293,  1994. 


Haas,  I.G.  BiP  (GRP78),  an  essential  hsp70 
resident  protein  in  the  endoplasmic  reticu- 
lum. Experientia  50:1012-1020,  1994. 

Hakan,  R.L.,  and  Henriksen,  S.J. 
Systemic  opiate  administration  has  hetero- 
geneous effects  on  activity  recorded  from 
nucleus  neurons  in  vivo.  Neurosci  Lett 
83:307-312,  1987. 

Hakan,  RL.,  and  Henriksen,  S.J.  Opiate 
influences  on  nucleus  accumbens  neu- 
ronal electrophysiology:  Dopamine  and 
non-dopamine  mechanisms.  J  Neurosci 
9:3538-3546,  1989. 

Harper,  J.C.;  Brennan,  C.H.;  and 
Littleton,  J.M.  Genetic  up-regulation  of 
calcium  channels  in  a  cellular  model  of 
ethanol  dependence.  Neuropharmacology 
28:1299-1302,  1989. 

Harris,  RA.,  and  Wood,  W.F.  Inhibition 
of  synaptosomal  calcium  uptake  by 
ethanol.  J  Pharmacol  Exp  Ther  213:562- 
568,  1980. 

Harris,  R.A.;  Mihic,  S.J.;  Brozowski,  S.; 
Hadingham,  K.;  and  Whiting,  P.J. 
Ethanol,  flunitrazepam,  and  pentobarbital 
modulation  of  GABAA  receptors 
expressed  in  mammalian  cells  and 
Xenopus  oocytes.  Alcohol  Clin  Exp  Res 
21:444-451,  1997. 

Hengerer,  B.;  Lindholm,  D.;  Heumann, 
R;  Ruther,  U.;  Wagner,  E.F.;  and 
Thoenen,  H.  Lesion-induced  increase  in 
nerve  growth  factor  mRNA  is  mediated 
by  c-fos.  Proc  Natl  Acad  Sci  USA 
87:3899-3903,  1990. 

Herkenham,  M.;  Edley,  S.M.;  and  Stuart, 
J.  Cell  clusters  in  the  nucleus  accumbens 
of  the  rat,  and  the  mosaic  relationship  of 
opiate  receptors,  acetylcholinesterase  and 
subcortical  afferent  terminations. 
Neuroscience  11:561-593,  1984. 


163 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Heyser,  C;  Schulteis,  G.;  Durbin,  P.;  and 
Koob,  G.  Chronic  acamprosate  decreases 
deprivation  induced  ethanol  self- adminis- 
tration in  rats.  Alcohol  Clin  Exp  Res 
20:15A,  1996. 

Hodge,  C.W.;  Chappelle,  A.M.;  and 
Samson,  H.H.  GABAergic  transmission  in 
the  nucleus  accumbens  is  involved  in  the 
termination  of  ethanol  self- administration 
in  rats.  Alcohol  Clin  Exp  Res  19:1486- 
1493,  1995. 

Hoffman,  P.L.  Neuroadaptive  functions 
of  the  neuropeptide  arginine  vasopressin. 
Ann  NT  Acad  Sci  739:168-175,  1994. 

Hoffman,  P.L.,  and  Tabakoff,  B.  Ethanol 
and  guanine  nucleotide  binding  proteins: 
A  selective  interaction.  FASEB  J  4:2612- 
2622,  1990. 

Hoffman,  P.L.;  Melchior,  C.L.;  and 
Tabakoff,  B.  Vasopressin  maintenance  of 
ethanol  tolerance  requires  intact  brain 
noradrenergic  systems.  Life  Sci  32:1065- 
1071,  1983. 

Hoffman,  P.L.;  Moses,  F.;  Hudspith,  M.; 
and  Tabakoff,  B.  Ethanol:  Potent  and 
selective  inhibitor  of  NMDA-stimulated 
cyclic  GMP  production.  Alcohol  Clin  Exp 
Res  13:310,  1989«. 

Hoffman,  P.L.;  Rabe,  C.;  Moses,  F.;  and 
Tabakoff,  B.  N-methyl-D-aspartate  recep- 
tors and  ethanol:  Inhibition  of  calcium 
flux  and  cyclic  GMP  production.  / 
Neurochem  52:1937-1940,  1989&. 

Hoffman,  P.L.;  Iorio,  K.R.;  Snell,  L.D.; 
and  Tabakoff,  B.  Attenuation  of  gluta- 
mate -induced  neurotoxicity  in  chronically 
ethanol-exposed  cerebellar  granule  cells 
by  NMDA  receptor  antagonists  and  gan- 
glioside  GMX.  Alcohol  Clin  Exp  Res 
19:721-726,  1995. 

Hoffman,  P.L.;  Bhave,  S.V.;  Kumar, 
K.N.;  lorio,  K.R.;  Snell,  L.D.;  Tabakoff, 


B.;  and  Michaelis,  E.K.  The  71  kDa  glu- 
tamate-binding  protein  is  increased  in 
cerebellar  granule  cells  after  chronic 
ethanol  treatment.  Mol  Brain  Res 
39:167-176,  1996. 

Hollmann,  M.,  and  Heinemann,  S. 
Cloned  glutamate  receptors.  Annu  Rev 
Neurosci  17:31-108,  1994. 

Hopkin,  K.  How  and  where  might  signal- 
transduction  cascades  intersect?  /  NIH 
Res 9:  25-28,  1997. 

Hopkins,  W.F.,  and  Johnston,  D.  Norad- 
renergic enhancement  of  long-term  po- 
tentiation at  mossy  fiber  synapses  in  the 
hippocampus.  /  Neurophysiol  59:667- 
687, 1988. 

Horger,  B.A.;  Shelton,  K.;  and  Schenk,  S. 
Preexposure  sensitizes  rats  to  the  reward- 
ing effects  of  cocaine.  Pharmacol  Biochem 
Behav  37:707-711,  1990. 

Hsieh,  K.-P.;  Wilke,  N.;  Harris,  R.A.;  and 
Miles,  M.F.  Interaction  of  ethanol  with 
inducers  of  glucose-regulated  stress  pro- 
teins. J  Biol  Chem  271:2709-2716,  1996. 

Hu,  X.-J.;  Follesa,  P.;  and  Ticku,  M.K. 
Chronic  ethanol  treatment  produces  a 
selective  upregulation  of  the  NMDA 
receptor  subunit  gene  expression  in  mam- 
malian cultured  cortical  neurons.  Mol 
Brain  Res  36:211-218,  1996. 

Hubbell,  C.L.;  Abelson,  M.L.;  Burkhardt, 
C.A.;  Herlands,  S.E.;  and  Reid,  L.D. 
Constant  infusions  of  morphine  and 
intakes  of  sweetened  ethanol  solution 
among  rats.  Alcohol  5:409^15,  1988. 

Hubner,  C.B.,  and  Moreton,  J.E.  Effects 
of  selective  Dl  and  D2  dopamine  antago- 
nists on  cocaine  self- administration  in  the 
rat.  Psychopharmacolojuy  (Berl)  105: 
151-156,  1991. 


164 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Hundle,  B.;  McMahon,  T.;  Dadgar,  J.; 
and  Messing,  RO.  Overexpression  of  e- 
protein  kinase  C  enhances  nerve  growth 
factor- induced  phosphorylation  of  mito- 
gen-activated  protein  kinases  and  neurite 
outgrowth.  J  Biol  Chem  270:30134- 
30140, 1995. 

Hung,  C.-R.;  Tabakoff,  B.H.;  Melchior, 
C.L.;  and  Hoffman,  P.L.  Intraventricular 
arginine  vasopressin  maintains  ethanol  toler- 
ance. Eur  J  Pharmacol  106:645-648, 1984. 

Hunt,  W.A.,  and  Lands,  W.E.M.  A  role 
for  behavioral  sensitization  in  uncontrolled 
ethanol  intake.  Alcohol 9:327-328,  1992. 

Hunt,  W.A.,  and  Majchrowicz,  E.  Studies 
of  neurotransmitter  interactions  after 
acute  and  chronic  ethanol  administration. 
Pharmacol  Biochem  Behav  18  (Suppl 
l):371-374,  1983. 

Hyman,  S.E.  ,  and  Nestler,  E.J.  Initiation 
and  adaptation:  A  paradigm  for  under- 
standing psychotropic  drug  action.  Am  J 
Psychiatry  153:151-162,  1996. 

Imperato,  A.;  Scrocco,  M.G.;  Bacchi,  S.; 
and  Angelucci,  L.  NMDA  receptors  and 
in  vivo  dopamine  release  in  the  nucleus 
accumbens  and  caudatus.  Eur  J 
Pharmacol  187:555-556,  1990. 

Iorio,  K.R.;  Reinlib,  L.;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Chronic  exposure  of  cere- 
bellar granule  cells  to  ethanol  results  in 
increased  N-methyl-D-aspartate  receptor 
function.  Mol  Pharmacol  41:1 142-1 148, 
1992. 

Iorio,  K.R.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Glutamate-induced  neurotoxicity  is 
increased  in  cerebellar  granule  cells  ex- 
posed chronically  to  ethanol.  Eur  J 
Pharmacol  248:209-212,  1993. 

Ishii,  T.;  Moriyoshi,  K.;  Sugihara,  H.; 
Sakurada,  K.;  Kadotani,  H.;  Yokoi,  M.; 
Akazawa,  C;  Shigemoto,  R.;  Mizuno,  N.; 


Masu,  M.;  and  Nakanishi,  S.  Molecular 
characterization  of  the  family  of  the  N- 
methyl-D-aspartate  receptor  subunits.  / 
Biol Chem  268:2836-2843,  1993. 

Ishizawa,  H.;  Mefford,  I.N.;  Tabakoff,  B.; 
and  Hoffman,  P.L.  Reduction  of  arginine 
vasopressin  binding  sites  in  mouse  lateral 
septum  by  treatment  with  6-hydroxydopa- 
mine.  Brain  Res  507:189-194,  1990. 

Jahromi,  S.S.,  and  Carlen,  P.L. 
Electrophysiological  effects  of  low  dose 
ethanol  on  calcium  currents  in  dentate 
granule  neurons  at  physiological  termper- 
ature.  Soc  Neurosci  Abstr  17:64,  1991. 

Jenab,  S.,  and  Inturrisi,  C.  Ethanol  and 
naloxone  differentially  upregulate  delta 
opioid  receptor  gene  expression  in  neu- 
roblastoma hybrid  (NG108-15)  cells.  Mol 
Brain  Res  27:95-102,  1994. 

Jenab,  S.,  and  Inturrisi,  C.  Activation  of 
protein  kinase  A  prevents  the  ethanol- 
induced  up-regulation  of  delta- opioid 
receptor  mRNA  in  NG108-15  cells.  Mol 
Brain  Res 47:44-48,  1997. 

Johnson,  K.A.,  and  Jennison,  K.M.  The 
drinking-smoking  syndrome  and  social 
context.  Int J Addict 27:749-792,  1992. 

Joseph,  A.M.  Nicotine  treatment  at  the 
drug  dependency  program  of  the 
Minneapolis  VA  medical  center.  /  Subst 
Abuse  Treat  10:147,  1993. 

Kahn,  A.J.,  and  Scudder,  C.L.  Alcohol 
withdrawal  effects  on  brain  serotonin  in 
mice  behaviorally  attracted  to  alcohol.  / 
Stud  Alcohol  37:1572-1580,  1976. 

Kalant,  H.  Research  on  tolerance:  What 
can  we  learn  from  history?  Alcohol  Clin 
Exp  Res  22:67-76,  1998. 

Kalivas,  P.W.,  and  Alesdatter,  J.E. 
Involvement  of  2V-methyl-D-aspartate 
receptor  stimulation  in  the  ventral 


165 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


tegmental  area  and  amygdala  in  behav- 
ioral sensitization  to  cocaine.  J  Pharmacol 
Exp  Ther  267:486-495,  1993. 

Kalivas,  P.W.;  Sorg,  B.A.;  and  Hooks, 
M.S.  The  pharmacology  and  neural  circuitry 
of  sensitization  to  psychostimulants.  Behav 
Pharmacol  4:315,  1993. 

Kandel,  E.R.  Cellular  mechanisms  of 
learning  and  the  biological  basis  of  indi- 
viduality. In:  Kandel,  E.R.;  Schwartz, 
J.A.;  and  Jessell,  T.M.,  eds.  Principles  of 
Neural  Science.  3d  ed.  New  York: 
Elsevier,  1991.  pp.  1009-1031. 

Kang,  M.;  Spigelman,  I.;  Sapp,  D.W.;  and 
Olsen,  R.W.  Persistent  reduction  of 
GABAA  receptor-mediated  inhibition  in 
rat  hippocampus  after  chronic  intermit- 
tent ethanol  treatment.  Brain  Res 
709:221-228,  1996. 

Kang,  M.H.;  Spigelman,  I.;  and  Olsen, 
R.W.  Alteration  in  the  sensitivity  of 
GABAA  receptors  to  allosteric  modulatory 
drugs  in  rat  hippocampus  following 
chronic  intermittent  ethanol  treatment. 
Alcohol  Clin  Exp  Res 22:2165-217 '3, 1988. 

Karobath,  M.;  Rogers,  J.;  and  Bloom,  F.E. 
Benzodiazepine  receptors  remain  unchanged 
after  chronic  ethanol  administration. 
Neuropharmacology  19:125-128,  1980. 

Keir,  W.J.,  and  Morrow,  A.L.  Differential 
expression  of  GABAA  receptor  subunit 
mRNAs  in  ethanol-naive  withdrawal 
seizure  resistant  (WSR)  vs.  withdrawal 
seizure  prone  (WSP)  mouse  brain.  Mol 
Brain  Res  25:200-208,  1994. 

Kellenberger,  S.;  Malherbe,  P.;  and  Sigel, 
E.  Function  of  the  ci1|32y2s  y-aminobu- 
tyric  acid  type  A  receptor  is  modulated  by 
protein  kinase  C  via  multiple  phosphory- 
lation sites.  J  Biol  Chem  267:25660- 
25663, 1992. 


Kennedy,  C,  and  Leff,  P.  How  should 
P2X  purinoceptors  be  classified  pharmaco- 
logically? Trends  Pharmacol  Sci  16:168- 
174, 1995. 

Khanna,  J.M.;  Kalant,  H.;  Shah,  G.;  and 
Chau,  A.  Effect  of  (+)MK-801  and  keta- 
mine  on  rapid  tolerance  to  ethanol.  Brain 
Res  Bull  28:311-314,  1992. 

Khanna,  J.M.;  Morato,  G.S.;  Chau,  A.; 
Shah,  G.;  and  Kalant,  H.  Effect  of 
NMDA  antagonists  on  rapid  and  chronic 
tolerance  to  ethanol:  Importance  of  intox- 
icated practice.  Pharmacol  Biochem  Behav 
48:755-763,  1994. 

Kiianmaa,  K.;  Stenius,  K.;  and  Sinclair, 
J.D.  Determinants  of  alcohol  preference 
in  the  AA  and  ANA  rat  lines  selected  for 
differential  ethanol  intake.  Alcohol  Alcohol 
Suppl  1:115-120,  1991. 

Kim,  W.-K.;  Johnson,  R.G.;  Izu,  L.T.; 
and  Rabin,  RA.  Chronic  ethanol  expo- 
sure inhibits  ATP -stimulated  but  not 
KC1- stimulated  dopamine  release  in  PC 
12  cells.  J  Pharmacol  Exp  Ther  270:336- 
341,  1994. 

King,  M.A.;  Hunter,  B.E.;  and  Walker, 
D.W.  Alterations  and  recovery  of  dendritic 
spine  density  in  rat  hippocampus  following 
long-term  ethanol  ingestion.  Brain  Res 
459:381-385,  1988. 

Klein,  R.L.;  Mascia,  M.P.;  Harkness, 
P.C.;  Hadingham,  K.L.;  Whiting,  P.J.; 
and  Harris,  RA.  Regulation  of  allosteric 
coupling  and  function  of  stably  expressed 
gamma-aminobutyric  acid  (GABA)A 
receptors  by  chronic  treatment  with  GABAA 
and  benzodiazepine  agonists.  / 
Pharmacol  Exp  Ther  274:1484-1492, 
19950. 

Klein,  R.L.;  Mascia,  M.P.;  Whiting,  P.J.; 
and  Harris,  RA.  GABAA  receptor  func- 
tion and  binding  in  stably  transfected 


166 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


cells:  Chronic  ethanol  treatment.  Alcohol 
ClinExpRes  19:1338-1344,  1995&. 

Kleven,  M.;  Ybema,  C;  Carilla,  E.; 
Hamon,  M.;  and  Koek,  W.  Modification 
of  behavioral  effects  of  8-hydroxy-2-(di- 
n-propylamino)tetralin  following  chronic 
ethanol  consumption  in  the  rat:  Evidence 
for  the  involvement  of  5-HT1A  receptors 
in  ethanol  dependence.  Eur  J  Pharmacol 
281:219-228,  1995. 

Knapp,  D.J.;  Criswell,  H.E.;  and  Breese, 
G.R.  Regional  effects  of  isoproterenol  on 
ethanol  enhancement  of  GABA  responses 
in  vivo.  Soc  Neurosci  Abstr  21:354,  1995. 

Kokka,  N.;  Sapp,  D.W.;  Taylor,  A.M.;  and 
Olsen,  RW.  The  kindling  model  of  alcohol 
dependence:  Similar  persistent  reduction 
in  seizure  threshold  to  pentylenetetrazol 
in  animals  receiving  chronic  ethanol  or 
chronic  pentylenetetrazol.  Alcohol  Clin 
Exp  Res  17:525-531,  1993. 

Koob,  G.F.,  and  Bloom,  F.E.  Cellular  and 
molecular  mechanisms  of  drug  depen- 
dence. Science  242:715-722,  1988. 

Koob,  G.F.,  and  Cador,  M.  Psychomotor 
stimulant  sensitization:  The  corticotropin  - 
releasing  factor-steroid  connection. 
Commentary  on  Wise  and  Leeb 
"Psychomotor-stimulant  sensitization:  a 
unitary  phenomenon?"  Behav  Pharmacol 
4:351-354,  1993. 

Koob,  G.F.;  Le,  H.T.;  and  Creese,  I.  The 
Dl  dopamine  receptor  antagonist  SCH 
23390  increases  cocaine  self- administration 
in  the  rat.  Neurosci  Lett  79:315-320,  1987. 

Kotlinska,  J.,  and  Liljequist,  S.  Oral  ad- 
ministration of  glycine  and  polyamine 
receptor  antagonists  blocks  ethanol  with- 
drawal seizures.  Psycho-pharmacology  (Berl) 
127:238-244,  1996. 

Krauss,  S.W.;  Ghirnikar,  R.B.;  Diamond,  I.; 
and  Gordon,  A.S.  Inhibition  of  adenosine 


uptake  by  ethanol  is  specific  for  one  class 
of  nucleoside  transporter.  Mol  Pharmacol 
44:1021-1026,  1993. 

Krimmer,  E.C.  HAS  and  LAS  rats  respond 
differentially  to  behavioral  effects  of  ethanol, 
pentobarbital,  chlorpromazine  and  chlor- 
diazepoxide.  Pharmacol  Biochem  Behav 
39:5-13, 1991. 

Krishek,  B.J.;  Xie,  X.;  Blackstone,  C; 
Huganir,  R.L.;  Moss,  S.J.;  and  Smart,  T.G. 
Regulation  of  GABAA  receptor  function 
by  protein  kinase  C  phosphorylation. 
Neuron  12:1081-1095,  1994. 

Krucker,  T.;  Toggas,  S.M.;  Mucke,  L.; 
and  Siggins,  G.R.  Transgenic  mice  with 
cerebral  expression  of  human  immunode- 
ficiency virus  type-1  coat  protein  gpl20 
show  divergent  changes  in  short-  and 
long-term  potentiation  in  CA1  hippocam- 
pus. Neuroscience  83:691-700,  1998. 

Krystal,  J.H.;  Webb,  E.;  Cooney,  N.; 
Kranzler,  H.R;  and  Charney,  D.S. 
Specificity  of  ethanollike  effects  elicited  by 
serotonergic  and  noradrenergic  mechanisms. 
Arch  Gen  Psychiatry  51:898-911,  1994. 

Lai,  H.;  Prather,  P.L.;  and  Rezazadeh, 
S.M.  Anxiogenic  behavior  in  rats  during 
ethanol  withdrawal:  Reversal  by  buspirone. 
Alcohol  8:467-471,  1991. 

Lai,  H.;  Prather,  P.L.;  and  Rezazadeh, 
S.M.  Potential  role  of  5-HT1C  and/or  5- 
HT2  receptors  in  the  mianserin -induced 
prevention  of  anxiogenic  behaviors  occur- 
ring during  ethanol  withdrawal.  Alcohol 
Clin  Exp  Res  17:411^17,  1993. 

Lands,  W.E.M.,  and  Zakhari,  S.  Alcohol 
and  cardiovascular  disease.  Alcohol  Health 
Res  World  14:304-312,  1990. 

Laurier,  L.G.;  Corrigall,  W.A.;  and 
George,  S.R.  Dopamine  receptor  density, 
sensitivity  and  mRNA  levels  are  altered 


167 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


following  self-administration  of  cocaine  in 
the  rat.  Brain  Res  634:31-40,  1994. 

Lavoie,  B.,  and  Parent,  A.  Immunohisto- 
chemical  study  of  the  serotoninergic 
innervation  of  the  basal  ganglia  in  the 
squirrel  monkey.  /  Comp  Neurol  299:1- 
16,  1990. 

Le,  A.D.;  Poulos,  C.X.;  and  Cappell,  H. 
Conditioned  tolerance  to  the  hypothermic 
effect  of  ethyl  alcohol.  Science  206: 1109- 
1110,1979. 

Le,  A.D.;  Khanna,  J.M.;  Kalant,  H.;  and 
LeBlanc,  A.E.  Effect  of  modification  of 
brain  serotonin  (5-HT),  norepinephrine 
(NE)  and  dopamine  (DA)  on  ethanol 
tolerance.  Psychopharmacology  (Berl) 
75:231-235,  1981*. 

Le,  A.D.;  Khanna,  J.M.;  Kalant,  H.;  and 
LeBlanc,  A.E.  The  effect  of  lesions  in  the 
dorsal,  median  and  magnus  raphe  nuclei 
on  the  development  of  tolerance  to  ethanol. 
J  Pharmacol  Exp  Ther  218:525-529, 1981*. 

LeBlanc,  A.E.;  Matsunaga,  M.;  and 
Kalant,  H.  Effects  of  frontal  polar  cortical 
ablation  and  cycloheximide  on  ethanol 
tolerance  in  rats.  Pharmacol  Biochem 
Behav  4:175-179,  1976. 

Lee,  M.A.,  and  Meltzer,  H.Y.  Neuroendo- 
crine responses  to  serotonergic  agents 
in  alcoholics.  Biol  Psychiatry  30:1017- 
1030,  1991. 

Leidenheimer,  N.J.;  McQuilkin,  S.J.; 
Harner,  L.D.;  Whiting,  P.;  and  Harris, 
RA.  Activation  of  protein  kinase  C  selec- 
tively inhibits  the  y-aminobutyric  acidA 
receptor:  Role  of  desensitization.  Mol 
Pharmacol  41:1116-1123,  1992. 

LeMarquand,  D.;  Pihl,  RO.;  and  Benkelfat, 
C.  Serotonin  and  alcohol  intake,  abuse, 
and  dependence:  Clinical  evidence.  Biol 
Psychiatry  36:326-337,  1994a. 


LeMarquand,  D.;  Pihl,  RO.;  and  Benkelfat, 
C.  Serotonin  and  alcohol  intake,  abuse,  and 
dependence:  Findings  of  animal  studies. 
Biol  Psychiatry  36:395^21,  1994*. 

Leslie,  S.W.;  Barl,  E.;  Chandler,  J.;  and 
Farrar,  RP.  Inhibition  of  fast  and  slow 
phase  depolarisation  dependent  synapto- 
somal calcium  uptake  by  ethanol.  / 
Pharmacol  Exp  Ther  225:571-575,  1983. 

Leslie,  S.W.;  Woodward,  J.J.;  Wilcox, 
R.E.;  and  Farrar,  R.P.  Chronic  ethanol 
treatment  uncouples  striatal  calcium  entry 
and  endogenous  dopamine  release.  Brain 
Res  368:174-177,  1986. 

Leslie,  S.W.;  Brown,  L.M.;  Dildy,  J.E.;  and 
Sims,  J.S.  Ethanol  and  neuronal  calcium 
channels.  Alcohol  7:233-236,  1990. 

Lessov,  C.N.,  and  Phillips,  T.J.  Duration 
of  sensitization  to  the  locomotor  stimu- 
lant effects  of  ethanol  in  mice.  Psycho- 
pharmacology  (Berl)  135:374-382,  1998. 

Lett,  B.T.  Repeated  exposures  intensify 
rather  than  diminish  the  rewarding  effects  of 
amphetamine,  morphine,  and  cocaine.  Psycho- 
pharmacology  (Berl)  98:357-362,  1989. 

Levin,  L.R.;  Han,  P.L.;  Hwang,  P.M.; 
Feinstein,  P.G.;  and  Davis,  R.L.  The 
Drosophila  learning  and  memory  gene 
rutabaga  encodes  a  Ca2+/calmodulin- 
responsive  adenylyl  cyclase.  Cell  68:479- 
489, 1992. 

Levine,  M.S.;  Li,  Z.;  Cepeda,  C;  Cromwell, 
H.C.;  and  Altemus,  K.L.  Neuromodula- 
tory  actions  of  dopamine  on  synaptically- 
evoked  neostriatal  responses  in  slices.  Synapse 
24:65-78,  1996. 

Levy,  A.D.,  and  Ellison,  G. 
Amphetamine -induced  enhancement  of 
ethanol  consumption:  Role  of  central 
catecholamines .  Psychopharmacology 
(Berl)  86:233-236,  1985. 


168 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Lewohl,  J.M.;  Crane,  D.I.;  and  Dodd,  P.R 
Expression  of  the  a^o^,  and  a3  isoforms 
of  the  GABAA  receptor  in  human  alcoholic 
brain.  Brain  Res  75 1:102-1 12,  1997. 

Lhuintre,  J. P.;  Daoust,  M.;  Moore,  N.; 
Chretien,  P.;  Saligaut,  C;  Tran,  G.; 
Boismare,  F.;  and  Hillemand,  B.  Ability 
of  calcium  bis  acetyl  homo  taurine,  a  GAB  A 
agonist,  to  prevent  relapse  in  weaned 
alcoholics.  Lancet i:1014-1016,  1985. 

Li,  C;  Aguayo,  L.;  Peoples,  R.W.;  and 
Weight,  F.F.  Ethanol  inhibits  neuronal 
ATP-gated  ion  channels.  Mol  Pharmacol 
44:871-875,  1993. 

Li,  C;  Peoples,  R.W.;  and  Weight,  F.F. 
Alcohol  action  on  a  neuronal  membrane 
receptor:  Evidence  for  a  direct  interaction 
with  the  receptor  protein.  Proc  Natl  Acad 
Set  [75^4  91:8200-8204,  1994. 

Li,  Y.Q.;  Rao,  Z.R;  and  Shi,  J.W. 
Serotoninergic  projections  from  the  mid- 
brain periaqueductal  gray  to  the  nucleus 
accumbens  in  the  rat.  Neurosci  Lett 
98:276-279,  1989. 

Liljequist,  S.  The  competitive  NMDA 
receptor  antagonist,  CGP  39551,  inhibits 
ethanol  withdrawal  seizures.  Eur  J 
Pharmacol  192:197 '-198,  1991. 

Lin,  A.;  Freund,  R.;  and  Palmer,  M. 
Ethanol  potentiation  of  GABA  induced 
electrophysiological  responses  in  cerebel- 
lum. Neurosci  Lett  122:154-158,  1991. 

Lin,  A.M.Y.;  Freund,  R.K.;  and  Palmer, 
M.R.  Sensitization  of  Y-aminobutyric 
acid-induced  depressions  of  cerebellar 
Purkinje  neurons  to  the  potentiative 
effects  of  ethanol  by  beta  adrenergic 
mechanisms  in  rat  brain.  J  Pharmacol  Exp 
Ther  265:426^32,  1993. 

Lingford- Hughes,  A.R.;  Acton,  P.D.; 
Gacinovic,  S.;  Suckling,  J.;  Busatto,  G.F.; 
Boddington,  S.J.;  Bullmore,  E.; 


Woodruff,  P.W.;  Costa,  D.C.;  Pilowsky, 
L.S.;  Ell,  P.J.;  Marshall,  E.J.;  and  Kerwin, 
R.W.  Reduced  levels  of  GABA- benzodi- 
azepine receptor  in  alcohol  dependency  in 
the  absence  of  grey  matter  atrophy.  Br  J 
Psychiatry  173:116-122,  1998. 

Little,  H.J.;  Dolin,  S.J.;  and  Halsey,  M.J. 
Calcium  channel  antagonists  decrease  the 
ethanol  withdrawal  syndrome.  Life  Sci 
39:2059-2065,  1986. 

Littleton,  J.M.;  Little,  H.J.;  and  Whittington, 
MA.  Effects  of  dihydropyridine  calcium 
channel  antagonists  in  ethanol  withdrawal; 
doses  required,  stereospecificity  and  actions 
of  Bay  K8644.  Psycho-pharmacology  (Berl) 
100:387-392,  1990. 

Lovinger,  D.M.,  and  Peoples,  R.W. 
Ethanol  (EtOH)  potentiation  of  5-HT3 
receptor  mediated  ion  current:  Comparison 
to  actions  of  other  sedative/hypnotic 
agents.  Alcohol  Clin  Exp  Res  15:325,  1991. 

Lovinger,  D.M.,  and  White,  G.  Ethanol 
potentiation  of  5-hydroxytryptamine3 
receptor-mediated  ion  current  in  neuro- 
blastoma cells  and  isolated  adult  mam- 
malian neurons.  Mol  Pharmacol  40:263- 
270,  1991. 

Lovinger,  D.M.;  White,  G.;  and  Weight, 
F.F.  Ethanol  inhibits  NMDA- activated 
ion  currents  in  hippocampal  neurons. 
Science  243:1721-1724,  1989. 

Lovinger,  D.M.;  White,  G.;  and  Weight, 
F.F.  NMDA  receptor- mediated  synaptic 
excitation  selectively  inhibited  by  ethanol 
in  hippocampal  slices  from  adult  rat.  / 
Neurosci  10:1372-1379,  1990. 

Ludvig,  N.;  Fox,  S.;  Kubie,  J.;  Altura,  B.; 
and  Altura,  B.  Application  of  the  com- 
bined single-cell  recording  intracerebral 
microdialysis  method  to  alcohol  research 
in  freely  behaving  animals.  Alcohol  Clin 
Exp  Res  22:41-50,  1998. 


169 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Ludwig,  A.M.;  Wilder,  A.;  and  Stark, 
L.H.  The  first  drink:  Psychobiological 
aspects  of  craving.  Arch  Gen  Psychiatry 
30:539-547,  1974. 

Madamba,  S.G.;  Hsu,  M.;  Schweitzer,  P.; 
and  Siggins,  G.R.  Ethanol  enhances  mus- 
carinic cholinergic  neurotransmission  in 
rat  hippocampus  in  vitro.  Brain  Res 
685:21-32,  1995. 

Madamba,  S.;  Schweitzer,  P.;  Ziegl- 
gansberger,  W.;  and  Siggins,  G.R. 
Acamprosate  (calcium  acetylhomotauri- 
nate)  enhances  the  NMD  A  component  of 
excitatory  neurotransmission  in  rat  hip- 
pocampal  CA1  neurons  in  vitro.  Alcohol 
Clin  Exp  Res  20:651-658, 1996. 

Madamba,  S.;  Schweitzer,  P.;  and  Siggins, 
G.  Gamma-hydroxybutyrate  decreases  the 
Q-current  in  rat  hippocampal  CA1  neu- 
rons. Alcohol  Clin  Exp  Res  21:73 A,  1997. 

Mahmoudi,  M.;  Kang,  M.H.;  Tillakaratne, 
N.;  Tobin,  A.J.;  and  Olsen,  RW.  Chronic 
intermittent  ethanol  treatment  in  rats 
increases  GABAA  receptor  a4-subunit 
expression:  Possible  relevance  to  alcohol 
dependence.  /  Neurochem  68:2485- 
2492,  1997. 

Maldonado,  R;  Robledo,  P.;  Chover, 
A.J.;  Caine,  S.B.;  and  Koob,  G.F.  Dl 
dopamine  receptors  in  the  nucleus  accum- 
bens  modulate  cocaine  self- administration 
in  the  rat.  Pharmacol  Biochem  Behav  45:239- 
242,  1993. 

Malec,  T.S.;  Malec,  E.A.;  and  Dongier, 
M.  Efficacy  of  buspirone  in  alcohol  de- 
pendence: A  review.  Alcohol  Clin  Exp  Res 
20:853-858,  1996. 

Malinow,  R  Transmission  between  pairs 
of  hippocampal  slice  neurons:  Quantal 
levels,  oscillations,  and  LTP.  Science 
252:722-724,  1991. 


Malosio,  M.;  Marqueze-Pouey,  B.; 
Kuhse,  J.;  and  Betz,  H.  Widespread  ex- 
pression of  glycine  receptor  subunit 
mRNAs  in  the  adult  and  developing  rat 
brain.  EMBOJ  10:2401-2409,  1991. 

Mancillas,  J.R;  Siggins,  J.R.;  and  Bloom, 
F.E.  Systemic  ethanol:  Selective  enhance- 
ment of  responses  to  acetylcholine  and 
somatostatin  in  hippocampus.  Science 
231:161-163,  1986. 

Mannix,  S.A.;  Hoffman,  P.L.;  and 
Melchior,  C.L.  Intraventricular  arginine 
vasopressin  blocks  the  acquisition  of 
ethanol  tolerance  in  mice.  Eur  J 
Pharmacol  128:137-141,  1986. 

Mansfield,  J.G.,  and  Cunningham,  C.L. 
Conditioning  and  extinction  of  tolerance  to 
the  hypothermic  effect  of  ethanol  in  rats. 
J  Comp  Physiol  Psychol  94:962-969,  1980. 

Mansour,  A.;  Kachaturian,  H.;  Lewis, 
M.E.;  Akil,  H.;  and  Watson,  S.J.  Anatomy 
of  CNS  opioid  receptors.  Trends  Neurosci 
11:308-314,  1988. 

Marks,  M.J.;  Stitzel,  J.A.;  and  Collins, 
A.C.  Influence  of  kinetics  of  nicotine 
administration  on  tolerance  development 
and  receptor  levels.  Pharmacol  Biochem 
Behav  27:505-512,  1987. 

Martz,  A.;  Dietrich,  R.A.;  and  Harris,  RA. 
Behavioral  evidence  for  the  involvement 
of  Y-aminobutyric  acid  in  the  actions  of 
ethanol.  Eur  J  Pharmacol  89:53-62,  1983. 

Masur,  J.,  and  Boerngen,  R  The  excitatory 
component  of  ethanol  in  mice:  A  chronic 
study.  Pharmacol  Biochem  Behav  13(6): 
777-780,  1980. 

Masur,  J.;  Oliveira  De  Souza,  M.L.;  and 
Zwicker,  A. P.  The  excitatory  effect  of 
ethanol:  Absence  in  rats,  no  tolerance  and 
increased  sensitivity  in  mice.  Pharmacol 
Biochem  Behav  24:1225-1228,  1986. 


170 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Matsumoto,  I.;  Leah,  J.;  Shanley,  B.;  and 
Wilce,  P.  Immediate  early  gene  expression 
in  the  rat  brain  during  ethanol  withdrawal. 
Mol  Cell  Neurosci  4:485-491, 1993. 

Matsuoka,  I.;  Maldonado,  R.;  Defer,  N.; 
Noel,  F.;  Hanoune,  J.;  and  Roques,  B.P. 
Chronic  morphine  administration  causes 
region-specific  increase  of  brain  type  VIII 
adenylyl  cyclase  mRNA.  Eur  J  Pharmacol 
268:215-221,  1994. 

Matsuzaki,  S.;  Takada,  M.;  Li,  Y.Q.; 
Tokuno,  H.;  and  Mizuno,  N.  Serotoninergic 
projections  from  the  dorsal  raphe  nucleus 
to  the  nucleus  submedius  in  the  rat  and 
cat.  Neuroscience  55:403-16,  1993. 

Matthews,  D.B.;  Devaud,  L.L.;  Fritschy, 
J.M.;  Sieghart,  W.;  and  Morrow,  A.L. 
Differential  regulation  of  GABAA  receptor 
gene  expression  by  ethanol  in  the  rat 
hippocampus  vs.  cerebral  cortex.  / 
Neurochem  70:1160-1166,  1998. 

Mayer,  J.;  Khanna,  J.;  Kalant,  H.;  and 
Spero,  L.  Cross-tolerance  between 
ethanol  and  morphine  in  guinea-pig 
ileum  longitudinal-muscle/myenteric- 
plexus  preparation.  Eur  J  Pharmacol 
63:223-227,  1980. 

Mayford,  M.;  Barzilai,  A.;  Keller,  F.; 
Schacher,  S.;  and  Kandel,  E.R  Modulation 
of  an  NCAM-related  adhesion  molecule 
with  long-term  synaptic  plasticity  in 
Aplysia.  Science  256:638-644,  1992. 

McBain,  C.J.,  and  Mayer,  M.L.  iV-methyl- 
D-aspartic  acid  receptor  structure  and 
function.  Physiol  Rev  74:723-760,  1994. 

McBride,  W.J.;  Guan,  X.-M.;  Chernet, 
E.;  Lumeng,  L.;  and  Li,  T.-K.  Regional 
differences  in  the  densities  of  serotonin- 
1A  receptors  between  P  and  NP  rats. 
Alcohol  Clin  Exp  Res  14:316,  1990. 

McBride,  W.J.;  Bodart,  B.;  Lumeng,  L.; 
and  Li,  T.K.  Association  between  low 


contents  of  dopamine  and  serotonin  in 
the  nucleus  accumbens  and  high  alcohol 
preference.  Alcohol  Clin  Exp  Res  19:1420- 
1422, 1995. 

McCown,  T.,  and  Breese,  G.  A  potential 
contribution  to  ethanol  withdrawal  kin- 
dling: Reduced  GABA  function  in  the 
inferior  collicular  cortex.  Alcohol  Clin  Exp 
Res  17:1290-1294,  1993. 

McCown,  T.J.;  Xiao,  X.;  Breese,  G.R.;  and 
Samulski,  R.J.  Differential  and  persistent 
expression  patterns  of  CNS  gene  transfer 
by  an  adeno-associated  virus  (AAV)  vec- 
tor. Brain  Res  713:99-107,  1996. 

McGehee,  D.S.;  Heath,  M.J.S.;  Gelber, 
S.;  Devay,  P.;  and  Role,  L.W.  Nicotine 
enhancement  of  fast  excitatory  synaptic 
transmission  in  CNS  by  presynaptic  recep- 
tors. Science  269:1692-1696,  1995. 

McGinty,  J.F.,  and  Wang,  J.Q.  Drugs  of 
abuse  and  striatal  gene  expression.  Adv 
Pharmacol  42:1017-1019,  1998. 

McMillan,  D.E.,  and  Snodgras,  S.H. 
Effects  of  acute  and  chronic  administra- 
tion of  delta-9-tetrahydrocannabinol  or 
cocaine  on  ethanol  intake  in  a  rat  model. 
Drug  Alcohol  Depend  27:263-274,  1991. 

Mehta,  A.K.,  and  Ticku,  M.K.  Ethanol 
potentiation  of  GABAergic  transmission 
in  cultured  spinal  cord  neurons  involves  7- 
aminobutyric  acid- gated  chloride  channels. 
J  Pharmacol  Exp  Ther  246:558-564, 1988. 

Mehta,  A.K.,  and  Ticku,  M.K.  Chronic 
ethanol  treatment  alters  the  behavioral 
effects  of  R015-4513,  a  partially  negative 
ligand  for  benzodiazepine  binding  sites. 
Brain  Res  489:93-100,  1989. 

Meisch,  RA.  Alcohol  self- administration 
by  experimental  animals.  In:  Smart,  R.G.; 
Cappell,  H.D.;  Glaser,  F.B.;  Israel,  Y.; 
Kalant,  H.;  Popham,  R.E.;  Schmidt,  W.; 
and  Sellers,  E.M.,  eds.  Research  Advances 


171 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  Alcohol  and  Drug  Problems.  New  York: 
Plenum  Press,  1984.  pp.  23-45. 

Melchior,  C.L.,  and  Tabakoff,  B.  Mod- 
ification of  environmentally  cued  toler- 
ance to  ethanol  in  mice.  J  Pharmacol  Exp 
Ther 219 ':175-180,  1981. 

Melchior,  C.L.,  and  Tabakoff,  B.  Features 
of  environment- dependent  tolerance  to 
ethanol.  Psychopharmacology  (Berl) 
87:94-100,  1985. 

Mello,  N.K.,  and  Mendelson,  H.J.  Clinical 
aspects  of  alcohol  dependence.  In:  Martin, 
W.R.,  ed.  Drug  Addiction  I:  Handbook  of 
Experimental  Pharmacology.  New  York: 
Springer- Verlag,  1977.  pp.  613-666. 

Memo,  M.;  Bovolin,  P.;  Costa,  E.;  and 
Grayson,  D.R.  Regulation  of  Y-aminobu- 
tyric  acid  receptor  subunit  expression  by 
activation  of  N-methyl-D-aspartate-selec- 
tive  glutamate  receptors.  Mol  Pharmacol 
39:599-603,  1991. 

Messing,  R.O.;  Carpenter,  C.L.;  Diamond, 
I.;  and  Greenberg,  D.A.  Ethanol  regulates 
calcium  channels  in  clonal  neural  cells.  Proc 
Natl  Acad  Sci  USA  83:6213-6215,  1986. 

Messing,  R.O.;  Sneade,  A.B.;  and  Savidge, 
B.  Protein  kinase  C  participates  in  upreg- 
ulation  of  dihydropyridine-sensitive  cal- 
cium channels  by  ethanol.  J  Neurochem 
55:1383-1389,  1990. 

Messing,  R.O.;  Petersen,  P.J.;  and 
Henrich,  C.J.  Chronic  ethanol  exposure 
increases  levels  of  protein  kinase  CS  and  £ 
and  protein  kinase  C-mediated  phospho- 
rylation in  cultured  neural  cells.  J  Biol 
Chem  266:23428-23432,  1991. 

Mhatre,  M.C.,  and  Ticku,  M.K.  Chronic 
ethanol  administration  alters  y-aminobu- 
tyric  acidA  receptor  gene  expression.  Mol 
Pharmacol  42:4 15-422,  1992. 


Mhatre,  M.C.,  and  Ticku,  M.K.  Chronic 
ethanol  treatment  downregulates  GABAA 
receptor  a  subunit  polypeptide  expression 
in  rat  cerebral  cortex  and  cerebellum. 
Alcohol  Clin  Exp  Res  17:442,  1993. 

Mhatre,  M.C.,  and  Ticku,  M.K.  Chronic 
ethanol  treatment  upregulates  the  GABA 
receptor  (3  subunit  expression.  Mol  Brain 
Res  23:246-252,  1994*. 

Mhatre,  M.C.,  and  Ticku,  M.K.  Chronic 
GABA  treatment  downregulates  the 
GABAA  receptor  alpha  2  and  alpha  3 
subunit  mRNAS  as  well  as  polypeptide 
expression  in  primary  cultured  cerebral 
cortical  neurons.  Mol  Brain  Res  24: 
159-165,  1994b. 

Mhatre,  M.;  Mehta,  A.K.;  and  Ticku, 
M.K.  Chronic  ethanol  administration 
increases  the  binding  of  the  benzodi- 
azepine inverse  agonist  and  alcohol  antag- 
onist [3H]R015-4513  in  rat  brain.  Eur  J 
Pharmacol  153:141-145,  1988. 

Mhatre,  M.C.;  Pena,  G.;  Sieghart,  W.; 
and  Ticku,  M.K.  Antibodies  specific  for 
GABAA  receptor  alpha  subunits  reveal 
that  chronic  alcohol  treatment  down- 
regulates alpha-subunit  expression  in  rat 
brain  regions.  /  Neurochem  61:1620- 
1625,  1993. 

Michaelis,  E.K;  Freed,  W.J.;  Galton,  N.; 
Foye,  J.;  Michaelis,  M.L.;  Phillips,  I.;  and 
Kleinman,  J.E.  Glutamate  receptor 
changes  in  brain  synaptic  membranes 
from  human  alcoholics.  Neurochem  Res 
15:1055-1063,  1990. 

Mihic,  S.J.;  Ye,  Q.;  Wick,  M.J.;  Koltchine, 
V.V.;  Krasowski,  M.A.;  Finn,  S.E.;  Mascia, 
M.P.;  Valenzuela,  C.F.;  Hanson,  K.K.; 
Greenblatt,  E.P.;  Harris,  R.A.;  and 
Harrison,  N.L.  Sites  of  alcohol  and  volatile 
anaesthetic  action  on  GABAA  and  glycine 
receptors.  Nature  389:334-335,  1997. 


172 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Miles,  M.F.;  Diaz,  J.E.;  and  DeGuzman, 
V.S.  Mechanisms  of  neuronal  adaptation 
to  ethanol.  Ethanol  induces  Hsc70  gene 
transcription  in  NG108-15  neuroblastoma 
x  glioma  cells.  J  Biol  Chem  266:2409- 
2414,  1991. 

Miles,  M.F.;  Wilke,  N.;  Elliot,  M.;  Tanner, 
W.;  and  Shah,  S.  Ethanol  responsive  genes 
in  neural  cells  include  the  78-kilodalton 
glucose-regulation  protein  (GRP78)  and 
94-kilodalton  glucose  regulated  protein 
(GRP94)  molecular  chaperones.  Mol 
Pharmacol  46:873-879,  1994. 

Milligan,  G.  The  stoichiometry  of  expression 
of  protein  components  of  the  stimulatory 
adenyl  cyclase  cascade  and  the  regulation 
of  information  transfer.  Cell  Signal  8:87- 
96,  1996. 

Mishina,  M.;  Toshiyuke,  T.;  Imoto,  K.; 
Noda,  M.;  Takahashi,  T.;  Numa,  S.; 
Methfessel,  C;  and  Sakmann,  B.  Molecular 
distinction  between  fetal  and  adult  forms 
of  muscle  acetylcholine  receptor.  Nature 
321:406-411,1986. 

Mitsuyama,  H.;  Little,  K.Y.;  Sieghart,  W.; 
Devaud,  L.L.;  and  Morrow,  A.L.  GABAA 
receptor  al,  a4  and  (33  subunit  mRNA 
and  protein  expression  in  frontal  cortex  of 
human  alcoholics.  Alcohol  Clin  Exp  Res 
22:815-822,  1998. 

Miyakawa,  T.;  Yagi,  T.;  Kitazawa,  H.; 
Yasuda,  M.;  Kawai,  N.;  Tsuboi,  K.;  and 
Niki,  H.  Fyn-kinase  as  a  determinant  of 
ethanol  sensitivity:  Relation  to  NMDA- 
receptor  function.  Science  278:698- 
701,  1997. 

Mochly-Rosen,  D.;  Chang,  F.-H.;  Cheever, 
L.;  Kim,  M.;  Diamond,  I.;  and  Gordon, 
A.S.  Chronic  ethanol  causes  heterologous 
desensitization  of  receptors  by  reducing 
as  messenger  RNA.  Nature  333:848- 
850,  1988. 


Modell,  J.G.;  Mountz,  J.M.;  Glaser,  F.B.; 
and  Lee,  JY.  Effect  of  haloperidol  on 
measures  of  craving  and  impaired  control 
in  alcoholic  subjects.  Alcohol  Clin  Exp  Res 
17:234-240,  1993. 

Moghaddam,  B.,  and  Bolinao,  M.L. 
Biphasic  effect  of  ethanol  on  extracellular 
accumulation  of  glutamate  in  the  hippo- 
campus and  the  nucleus  accumbens. 
Neurosci  Lett 178:99-102,  1994. 

Monti,  J.M.,  and  Alterwain,  P.  Ritanserin 
decreases  alcohol  intake  in  chronic  alcoholics. 
Lancet  337:  60,  1991. 

Montpied,  P.;  Morrow,  A.L.;  Karanian, 
J.W.;  Ginns,  E.I.;  Martin,  B.M.;  and  Paul, 
S.M.  Prolonged  ethanol  inhibition  de- 
creases gamma-aminobutyric  acidA  recep- 
tor a  subunit  mRNAs  in  the  rat  cerebral 
cortex.  Mol  Pharmacol  39:157-163,  1991. 

Monyer,  H.;  Sprengel,  R.;  Schoepfer,  R; 
Herb,  A.;  Higuchi,  M.;  Lomeli,  H.; 
Burnashev,  N.;  Sakmann,  B.;  and  Seeburg, 
P.H.  Heteromeric  NMDA  receptors: 
Molecular  and  functional  distinction  of 
subtypes.  Science  256:1217-1221,  1992. 

Moore,  M.S.;  DeZazzo,  J.;  Luk,  AY.; 
Tully,  T.;  Singh,  CM.;  and  Heberlein,  U. 
Ethanol  intoxication  in  Drosophila: 
Genetic  and  pharmacological  evidence  for 
regulation  by  the  cAMP  signaling  path- 
way [see  comments].  Cell 93(6):997- 
1007,  1998;  comment  on  pp.  909-912. 

Moore,  S.D.;  Madamba,  S.G.;  and 
Siggins,  G.R.  Ethanol  diminishes  a  volt- 
age-dependent K+  current,  the  M-current, 
in  CA1  hippocampal  pyramidal  neurons 
in  vitro.  Brain  Res  516:222-228,  1990. 

Moore,  S.D.;  Madamba,  S.G.;  Schweitzer, 
P.;  and  Siggins,  G.R.  Voltage-dependent 
effects  of  opioid  peptides  on  hippocampal 
CA3  pyramidal  neurons  in  vitro.  /  Neurosci 
14:809-820,  1994. 


173 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Morgan,  P.F.;  Nadi,  N.S.;  Karanian,  J.; 
and  Linnoila,  M.  Mapping  rat  brain  struc- 
tures activated  during  ethanol  withdrawal: 
Role  of  glutamate  and  NMDA  receptors. 
Eur  J  Pharmacol  225:217-223,  1992. 

Morrisett,  RA.  Potentiation  of  N-methyl- 
D-aspartate  receptor-dependent  afterdis- 
charges  in  rat  dentate  gyrus  following  in 
vitro  ethanol  withdrawal.  Neurosci  Lett 
167:175-178,  1994. 

Morrisett,  R.A.,  and  Swartzwelder,  H.S. 
Attenuation  of  hippocampal  long-term 
potentiation  by  ethanol:  A  patch-clamp 
analysis  of  glutamatergic  and  GABAergic 
mechanisms.  J  Neurosci  13:2264- 
2272,  1993. 

Morrisett,  R.A.;  Rezvani,  A.H.;  Overstreet, 
D.;  Janowsky,  D.S.;  Wilson,  W.A.;  and 
Swartzwelder,  H.S.  MK-801  potently 
inhibits  alcohol  withdrawal  seizures  in  rats. 
Eur  J  Pharmacol  176:103-105,  1990. 

Morrow,  A.L.  Regulation  of  GABAA 
receptor  function  and  gene  expression  in 
the  central  nervous  system.  In:  Bradley, 
R.J.,  and  Harris,  R.A.,  eds.  International 
Review  of  Neurobiology.  Vol.  38.  New 
York:  Academic  Press,  1995.  p.  1. 

Morrow,  A.L.;  Suzdak,  P.D.;  Karanian, 
J.W.;  and  Paul,  S.M.  Chronic  ethanol 
administration  alters  Y-aminobutyric  acid, 
pentobarbital  and  ethanol-mediated  36C1~ 
uptake  in  cerebral  cortical  synaptoneuro- 
somes.  J  Pharmacol  Exp  Ther  246:158- 
164,  1988. 

Morrow,  A.L.;  Montpied,  P.;  Lingford- 
Hughes,  A.;  and  Paul,  S.M.  Chronic 
ethanol  and  pentobarbital  administration 
in  the  rat:  Effects  on  GABAA  receptor 
function  and  expression  in  brain.  Alcohol 
7:237-244,  1990. 

Morrow,  A.L.;  Herbert,  J.S.;  and  Montpied, 
P.  Differential  effects  of  chronic  ethanol 


administration  on  GABAA  receptor  ctl  and 
a6  subunit  mRNA  levels  in  rat  cerebel- 
lum. Mol  Cell  Neurosci  3:251-258,  1992. 

Moss,  S.J.;  Gorrie,  G.H.;  Amato,  A.;  and 
Smart,  T.G.  Modulation  of  GABAA  recep- 
tors by  tyrosine  phosphorylation.  Nature 
377:344-348,  1995. 

Mullikin-Kilpatrick,  D.,  and  Treistman, 
S.N.  Ethanol  inhibition  of  L-type  Ca2+ 
channels  in  PC12  cells:  Role  of  permeant 
ions.  Eur  J  Pharmacol  270:17-25,  1994. 

Murray,  R.P.;  Istvan,  J.A.;  Voelker,  H.T.; 
Rigdon,  M.A.;  and  Wallace,  M.D.  Level 
of  involvement  with  alcohol  and  success 
at  smoking  cessation  in  the  lung  health 
study.  J  Stud  Alcohol  56:74-82,  1995. 

Nagy,  L.E.;  Diamond,  I.;  Collier,  K.; 
Lopez,  L.;  Ullman,  B.;  and  Gordon,  A.S. 
Adenosine  is  required  for  ethanol-induced 
heterologous  desensitization.  Mol 
Pharmacol  36:744-748,  1989. 

Nagy,  L.E.;  Diamond,  I.;  Casso,  D.J.; 
Franklin,  C;  and  Gordon,  A.S.  Ethanol 
increases  extracellular  adenosine  by  in- 
hibiting adenosine  uptake  via  the  nucleo- 
side transporter.  J  Biol  Chem  265:1946- 
1951,1990. 

Nagy,  L.;  Diamond,  I.;  and  Gordon,  A.S. 
cAMP-dependent  protein  kinase  regulates 
inhibition  of  adenosine  transport  by 
ethanol.  Mol  Pharmacol  40:8 12-8 17, 1991. 

Nakanishi,  S.  Molecular  diversity  of  gluta- 
mate receptors  and  implications  for  brain 
function.  Science  258:597-603,  1992. 

Naranjo,  C.A.,  and  Bremner,  K.E. 
Clinical  pharmacology  of  serotonin-alter- 
ing medications  for  decreasing  alcohol 
consumption.  Alcohol  Alcohol  Suppl 
2:221-229,  1993. 

Nestby,  P.;  Vanderschuren,  L.J.M.J.;  De 
Vries,  T.J.;  Hogenboom,  F.;  Wardeh,  G.; 


174 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Mulder,  A.H.;  and  Schoffelmeer,  A.N.M. 
Ethanol,  like  psychostimulants  and  mor- 
phine, causes  long-lasting  hyperreactivity 
of  dopamine  and  acetylcholine  neurons  of 
rat  nucleus  accumbens:  Possible  role  in 
behavioral  sensitization.  Psychopharma- 
cology  (Berl)  133:69-76,  1997. 

Newlin,  D.B.,  and  Thomson,  J.B.  Chronic 
tolerance  and  sensitization  to  alcohol  in 
sons  of  alcoholics.  Alcohol  Clin  Exp  Res 
15:399^05, 1991. 

Ng,  G.Y.K.,  and  George,  S.R.  Dopamine 
receptor  agonist  reduces  ethanol  self- 
administration  in  the  ethanol-preferring 
C57BL/6J  inbred  mouse.  Eur  J 
Pharmacol  269:365-374,  1994. 

Nie,  Z.;  Yuan,  X.;  Madamba,  S.G.;  and 
Siggins,  G.R.  Ethanol  decreases  gluta- 
matergic  synaptic  transmission  in  rat 
nucleus  accumbens  in  vitro:  Naloxone 
reversal.  J  Pharmacol  Exp  Ther  266: 
1705-1712,  1993. 

Nie,  Z.;  Madamba,  S.G.;  and  Siggins, 
G.R.  Ethanol  inhibits  glutamatergic  neuro- 
transmission in  nucleus  accumbens  neurons 
by  multiple  mechanisms.  J  Pharmacol  Exp 
Ther  271:1566-1573,  1994. 

Nie,  Z.;  Madamba,  S.G.;  and  Siggins, 
G.R.  Withdrawal  from  chronic  ethanol 
exposure  increases  sensitivity  to  N-methyl- 
D-aspartate  (NMDA)  in  rat  nucleus  ac- 
cumbens neurons.  Soc  Neurosci  Abstr 
21:2101,1995. 

Nie,  Z.;  Steffensen,  S.C.;  Criado,  J.R.; 
Henriksen,  S.J.;  and  Siggins,  G.R.  Ethanol 
inhibition  of  NMDA  responses  by  activa- 
tion of  presynaptic  GABAB  receptors.  Soc 
Neurosci  Abstr 22:207 '4,  1996. 

Nimchinsky,  E.A.;  Hof,  P.R.;  Young, 
W.G.;  and  Morrison,  J.H.  Neurochemical, 
morphologic  and  laminar  characterization 
of  cortical  projection  neurons  in  the  cin- 


gulate  motor  areas  of  the  macaque  mon- 
key. /  Comp  Neurol  374:136-160,  1996. 

O'Brien,  C.P.;  Ehrman,  R.N.;  and 
Ternes,  J.W.  Classical  conditioning  in 
human  opioid  dependence.  In:  Goldberg, 
S.R.,  and  Stolerman,  LP.,  eds.  Behavioral 
Analysis  of  Drug  Dependence.  Orlando, 
FL:  Academic  Press,  1986.  pp.  329-356. 

Oleson,  D.R.;  DeFelice,  L.J.;  and 
Donahoe,  RM.  Ethanol  increases  K+ 
conductance  in  human  T- cells.  Alcohol 
Clin  Exp  Res  17:604-609,  1993. 

O'Malley,  S.S.;  Jaffe,  A.;  Chang,  G.; 
Schottenfeld,  R.S.;  Meyer,  RE.;  and 
Rounsaville,  B.  Naltrexone  and  coping 
skills  therapy  for  alcohol  dependence:  A 
controlled  study.  Arch  Gen  Psychiatry 
49:881-887,  1992. 

Orentreich,  N.;  Brind,  J.L.;  Rizer,  R.L.; 
and  Vogelman,  J.H.  Age  changes  and  sex 
differences  in  serum  dehydroepiandos- 
terone  sulfate  concentrations  throughout 
adulthood.  /  Clin  Endocrinol  Metab 
59:551-555,  1984. 

Orona,  E.;  Hunter,  B.E.;  and  Walker, 
D.W.  Ethanol  exposure  following  unilat- 
eral entorhinal  deafferentation  alters 
synaptic  reorganization  in  the  rat  dentate 
gyrus:  A  quantitative  analysis  of  acetyl- 
cholinesterase histochemistry.  Exp  Neurol 
101:114-131,1988. 

Ortiz,  J.;  Fitzgerald,  L.W.;  Charlton,  M.; 
Lane,  S.;  Trevisan,  L.;  Guitart,  X.; 
Shoemaker,  W.;  Duman,  R.S.;  and  Nestier, 
E.J.  Biochemical  actions  of  chronic  ethanol 
exposure  in  the  mesolimbic  dopamine 
system.  Synapse 21:289-298,  1995. 

Osmanovic,  S.S.,  and  Shefner,  S.A. 
Anomalous  rectification  in  rat  locus 
coeruleus  neurons.  Brain  Res  417: 
161-166,  1987. 


175 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Osmanovic,  S.S.,  and  Shefner,  S.A. 
Ethanol  enhances  inward  rectification  in 
rat  locus  coeruleus  neurons  by  increasing 
the  extracellular  potassium  concentration. 
J  Pharmacol  Exp  Ther  271:334-342,  1994. 

Pan,  Z.Z.;  Williams,  J.T.;  and  Osborne, 
P.B.  Opioid  actions  on  single  nucleus 
raphe  magnus  neurons  from  rat  and 
guinea-pig  in  vitro.  J  Physiol  (Lond) 
427:519-532,  1990. 

Parsons,  L.H.,  and  Justice,  J.B.,  Jr. 
Serotonin  and  dopamine  sensitization  in 
the  nucleus  accumbens,  ventral  tegmental 
area,  and  dorsal  raphe  nucleus  following 
repeated  cocaine  administration.  / 
Neurochem  61:1611-1619,  1993. 

Paul,  S.M.,  and  Purdy,  R.H.  Neuroactive 
steroids.  FASEB J 6:231 1-2322,  1992. 

Pellegrino,  S.M.;  Woods,  J.M.;  and 
Druse,  M.J.  Effects  of  chronic  ethanol 
consumption  on  G  proteins  in  brain  areas 
associated  with  the  nigrostriatal  and 
mesolimbic  dopamine  systems.  Alcohol 
Clin  Exp  Res  17:1247-1253,  1993. 

Pereira,  E.F.;  Aracava,  Y.;  Aronstam, 
R.A.;  Barreiro,  E.J.;  and  Albuquerque, 
E.X.  Pyrazole,  an  alcohol  dehydrogenase 
inhibitor,  has  dual  effects  on  N-methyl- 
D-aspartate  receptors  of  hippocampal 
pyramidal  cells:  Agonist  and  noncompeti- 
tive antagonist.  J  Pharmacol  Exp  Ther 
261:331-340,  1992. 

Peris,  J.;  Boyson,  S.J.;  Cass,  W.A.; 
Curella,  P.;  Dwoskin,  L.P.;  Larson,  G.; 
Lin,  L.-H.;  Yasuda,  R.P.;  and  Zahniser, 
N.R.  Persistence  of  neurochemical  changes 
in  dopamine  systems  after  repeated  co- 
caine administration.  J  Pharmacol  Exp 
7^253:38^4,1990. 

Peris,  J.;  Eppler,  B.;  Hu,  M.;  Walker, 
D.W.;  Hunter,  B.E.;  Mason,  K.;  and 
Anderson,  K.J.  Effects  of  chronic  ethanol 


exposure  on  GABA  receptors  and  GABAB 
receptor  modulation  of  3H-GABA  release 
in  the  hippocampus.  Alcohol  Clin  Exp  Res 
21:1047-1052,  1997. 

Phelix,  C.F.,  and  Broderick,  P.A.  Light 
microscopic  immunocytochemical  evi- 
dence of  converging  serotonin  and 
dopamine  terminals  in  ventrolateral  nu- 
cleus accumbens.  Brain  Res  Bull  37: 
37-40,  1995. 

Phillips,  T.J.;  Burkhart-Kasch,  S.;  and 
Crabbe,  J.C.  Locomotor  activity  response 
to  chronic  ethanol  treatment  in  selectively 
bred  FAST  and  SLOW  mice.  Alcohol 
Alcohol  Suppl  1:109-113,  1991. 

Phillips,  T.J.;  Dickinson,  S.;  and  Burkhart- 
Kasch,  S.  Behavioral  sensitization  to  drug 
stimulant  effects  in  C57BL/6J  and 
DBA/2  J  inbred  mice.  Behav  Neurosci 
108:789-803,  1994. 

Phillips,  T.J.;  Huson,  M.;  Gwiazdon,  C.; 
Burkhart-Kasch,  S.;  and  Shen,  E.H. 
Effects  of  acute  and  repeated  ethanol 
exposures  on  the  locomotor  activity  of 
BXD  recombinant  inbred  mice.  Alcohol 
Clin  Exp  Res  19:269-278,  1995. 

Popova,  J.S.;  Johnson,  G.L.;  and  Rasenick, 
M.M.  Chimeric  Gas/G,^  proteins  define 
domains  on  Gas  which  interact  with  tubu- 
lin for  the  p  adrenergic  activation  of 
adenylyl  cyclase.  J  Biol  Chem  269: 
21748-21754,  1994. 

Porter,  N.M.;  Twyman,  R.E.;  Uhler, 
M.D.;  and  Macdonald,  RL.  Cyclic  AMP- 
dependent  protein  kinase  decreases 
GABAA  receptor  current  in  mouse  spinal 
neurons.  Neuron  5:789-796,  1990. 

Potthoff,  A.D.;  Ellison,  G.;  and  Nelson,  L. 
Ethanol  intake  increases  during  continuous 
administration  of  amphetamine  and  nicotine, 
but  not  several  other  drugs.  Pharmacol 
Biochem  Behav  18:909-918,  1983. 


176 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Proctor,  W.R.;  Soldo,  B.L.;  Allan,  A.M.; 
and  Dunwiddie,  T.V.  Ethanol  enhances 
synaptically  evoked  GABAA  receptor- 
mediated  responses  in  cerebral  cortical 
neurons  in  rat  brain  slices.  Brain  Res 
595:220-227,  1992. 

Puia,  G.;  Santi,  M.;  Vicini,  S.;  Pritchett, 
D.B.;  Purdy,  R.H.;  Paul,  S.M.;  Seeburg, 
P.H.;  and  Costa,  E.  Neurosteroids  act  on 
recombinant  human  GABAA  receptors. 
Neuron  4:  759-765,  1990. 

Puia,  G.;  Duck,  I.;  Vicini,  S.;  and  Costa, 
E.  Does  neurosteroid  modulatory  efficacy 
depend  on  GABAA  receptor  subunit 
composition?  Receptors  Channels  1:135— 
142,  1993. 

Purdy,  R.H.;  Moore,  P.H.;  Rao,  P.N.; 
Hagino,  N.;  Yamaguchi,  T.;  Schmidt,  P.; 
Rubinow,  D.;  Morrow,  A.L.;  and  Paul, 
S.M.  Radioimmunoassay  of  3a-hydroxy- 
5a-pregnan-20-one  in  rat  and  human 
plasma.  Steroids  55:290-296,  1990. 

Purdy,  R.H.;  Morrow,  A.L.;  Moore, 
P.H.,  Jr.;  and  Paul,  S.M.  Stress-induced 
elevations  of  gamma-aminobutyric  acid 
type  A  receptor-active  steroids  in  the  rat 
brain.  Proc  Natl  Acad  Sci  USA  88:4553- 
4557,  1991. 

Rabin,  RA.  Differential  response  of 
adenylate  cyclase  and  ATPase  activities 
after  chronic  ethanol  exposure  of  PC12 
ceOs.JNeurocbem  51:1148-1155, 1988. 

Rabin,  RA.  Ethanol-induced  desensitiza- 
tion  of  adenylate  cyclase:  Role  of  the 
adenosine  receptor  and  GTP-binding 
proteins.  J  Pharmacol  Exp  Ther  264:977- 
983, 1993. 

Rabin,  R.A.;  Fiorella,  D.;  and  Van  Wylen, 
D.G.L.  Role  of  extracellular  adenosine  in 
ethanol-induced  desensitization  of  cyclic 
AMP  production.  J  Neurochem  60:1012- 
1017,  1993. 


Rastogi,  S.K.;  Thyagarajan,  R;  Clothier,  J.; 
and  Ticku,  M.K.  Effect  of  chronic  treatment 
of  ethanol  on  benzodiazepine  and  picrotoxin 
sites  on  the  GABA  receptor  complex  in 
regions  of  the  brain  of  the  rat.  Neuro- 
pharmacology 25:1179-1184,  1986. 

Rebec,  G.  Real-time  assessments  of 
dopamine  function  during  behavior: 
Single-unit  recording,  iontophoresis,  and 
fast-scan  cyclic  voltammetry  in  awake, 
unrestrained  rats.  Alcohol  Clin  Exp  Res 
22:32^0,  1998. 

Reid,  L.D.;  Hunter,  G.A.;  Beaman,  CM.; 
and  Hubbell,  C.L.  Toward  understanding 
ethanol's  capacity  to  be  reinforcing:  A 
conditioned  place  preference  following 
injections  of  ethanol.  Pharmacol  Biochem 
Behav  22:483^87,  1985. 

Reithmann,  C;  Geirschik,  P.;  Werdan,  K.; 
and  Jakobs,  K.H.  Role  of  inhibitory  G 
protein  a-subunits  in  adenylyl  cyclase 
desensitization.  Mol  Cell  Endocrinol 
82:C215-C221,  1991. 

Rezazadeh,  S.M.;  Prather,  P.L.;  and  Lai, 
H.  Sensitization  to  5-HT1C  receptor 
agonist  in  rats  observed  following  with- 
drawal from  chronic  ethanol.  Alcohol 
10:281-283,  1993. 

Richardson,  N.R.;  Piercey,  M.F.; 
Svensson,  K.;  Collins,  R.J.;  Myers,  J.E.; 
and  Roberts,  D.C.S.  Antagonism  of  co- 
caine self-administration  by  the  preferen- 
tial dopamine  autoreceptor  antagonist, 
(+)-AJ  76.  Brain  Res  619:15-21,  1993. 

Ripley,  T.L.;  Whittington,  M.A.; 
Butterworth,  A.R;  and  Little,  H.J. 
Ethanol  withdrawal  hyperexcitability  in 
vivo  and  in  isolated  mouse  hippocampal 
slices.  Alcohol  Alcohol  31:347-357,  1996. 

Roberts,  A.J.;  Lessov,  C.N.;  and  Phillips, 
T.J.  Critical  role  of  glucocorticoid  recep- 
tors in  stress-  and  ethanol-induced  loco- 


177 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


motor  sensitization.  J  Pharmacol  Exp 
Ther  275:790-797,  1995. 

Robertson,  H.A.  Immediate-early  genes, 
neuronal  plasticity,  and  memory.  Biochem 
Cell  Biol  70:729-7 37,  1992. 

Robinson,  T.E.;  Jurson,  P.A.;  Bennett, 
J.A.;  and  Bentgen,  K.M.  Persistent  sensi- 
tization of  dopamine  neurotransmission 
in  ventral  striatum  (nucleus  accumbens) 
produced  by  prior  experience  with  (+)- 
amphetamine:  A  microdialysis  study  in 
freely  moving  rats.  Brain  Res  540:159- 
163,  1988. 

Roehrs,  T.A.,  and  Samson,  H.H.  Ethanol 
reinforced  behavior:  Effect  of  chronic 
ethanol  overdrinking.  Alcohol  Clin  Exp 
2t«  5:165, 1981. 

Rogers,  C.J.,  and  Hunter,  B.E.  Chronic 
ethanol  treatment  reduces  inhibition  in 
CA1  of  the  rat  hippocampus.  Brain  Res 
Bull  28:587-592,  1992. 

Roivainen,  R.;  Hundle,  B.;  and  Messing, 
R.O.  Ethanol  enhances  growth  factor 
activation  of  mitogen-activation  protein 
kinases  by  a  protein  kinase  C-dependent 
mechanism.  Proc  Natl  Acad  Sci  USA 
92:1891-1895,  1995. 

Rossetti,  Z.L.;  Hmaidan,  Y.;  and  Gessa, 
G.L.  Marked  inhibition  of  mesolimbic 
dopamine  release:  A  common  feature  of 
ethanol,  morphine,  cocaine  and  ampheta- 
mine abstinence  in  rats.  Eur  J  Pharmacol 
221:227-234,  1992/?. 

Rossetti,  Z.L.;  Melis,  F.;  Carboni,  S.; 
Diana,  M.;  and  Gessa,  G.L.  Alcohol  with- 
drawal in  rats  is  associated  with  a  marked 
fall  in  extraneuronal  dopamine.  Alcohol 
Clin  Exp  Res  16:529-532,  1992/;. 

Rothberg,  B.S.,  and  Hunter,  B.E. 
Chronic  ethanol  treatment  differentially 
affects  muscarinic  receptor  responses  in 


rat  hippocampus.  Neurosci  Lett  132: 
243-246, 1991. 

Rothberg,  B.S.;  Yasuda,  R.P.;  Satkus, 
S.A.;  Wolfe,  B.B.;  and  Hunter,  B.E. 
Effects  of  chronic  ethanol  on  cholinergic 
actions  in  rat  hippocampus:  Electro- 
physiological studies  and  quantification  of 
ml-m5  muscarinic  receptor  subtypes. 
Brain  Res  631:227-234,  1993. 

Rothberg,  B.S.;  Hunter,  B.E.;  Walker, 
D.W.;  Anderson,  J.F.;  and  Anderson,  K.J. 
Long-term  effects  of  chronic  ethanol  on 
muscarinic  receptor  binding  in  rat  brain. 
Alcohol  Clin  Exp  Res  20:1613-1617, 1996. 

Rudolph,  J.G.;  Walker,  D.W.;  Iimuro,  Y.; 
Thurman,  R.G.;  and  Crews,  F.T.  NMDA 
receptor  binding  in  adult  rat  brain  after 
several  chronic  ethanol  treatment  protocols. 
Alcohol  Clin  Exp  Res  21:1508-1519, 1997. 

Russek,  S.J.;  Bandyopadhyay,  S.;  and 
Farb,  D.H.  Structural  analysis  of  the 
human  GAB  RBI  promotor  reveals  critical 
regulatory  regions  responsible  for  the 
neural  specific  and  autologous  regulation 
of  gene  transcription.  Soc  Neurosci  Abstr 
23:1,  1997. 

Saito,  T.;  Lee,  J.M.;  Hoffman,  P.L.;  and 
Tabakoff,  B .  Effects  of  chronic  ethanol 
treatment  on  the  (3-adrenergic  receptor- 
coupled  adenylate  cyclase  system  of 
mouse  cerebral  cortex.  /  Neurochem 
48:1817-1822,  1987. 

Samson,  H.H.  Initiation  of  ethanol- 
maintained  behavior:  A  comparison  of 
animal  models  and  their  implication  to 
human  drinking.  In:  Thompson,  T.; 
Dews,  P.B.;  and  Barrett,  J.E.,  eds. 
Advances  in  Behavioral  Pharmacology,  Vol. 
VI:  Neurobehavioral  Pharmacology. 
Hillsdale,  NJ:  Lawrence  Erlbaum 
Associates,  1987.  pp.  221-248. 


178 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Sanna,  E.;  Serra,  M.;  Cossu,  A.;  Colombo, 
G.;  Follesa,  P.;  Cuccheddu,  T.;  Concas, 
A.;  and  Biggio,  G.  Chronic  ethanol  intox- 
ication induces  differential  effects  on 
GABAA  and  NMDA  receptor  function  in 
the  rat  brain.  Alcohol  Clin  Exp  Res 
17:115-123,1993. 

Sanna,  E.;  Dildy-Mayfield,  J.E.;  and  Harris, 
R.A.  Ethanol  inhibits  the  function  of  5- 
hydroxytryptamine  type  lc  and  muscarinic 
Ml  G  protein-linked  receptors  in  Xenopus 
oocytes  expressing  brain  mRNA:  Role  of 
protein  kinase  C.  Mol  Pharmacol  45:1004- 
1012, 1994. 

Scheetz,  A.J.,  and  Constantine-Paton,  M. 
Modulation  of  NMDA  receptor  function: 
Implications  for  vertebrate  neural  devel- 
opment. FASEB  J 8:745-752,  1994. 

Schuckit,  MA.  Low  level  of  response  to 
alcohol  as  a  predictor  of  future  alcoholism. 
Am  J  Psychiatry  151:184-189,  1994. 

Schulteis,  G.;  Markou,  A.;  Gold,  L.H.; 
Stinus,  L.;  and  Koob,  G.F.  Relative  sensi- 
tivity to  naloxone  of  multiple  indices  of 
opiate  withdrawal:  A  quantitative  dose- 
response  analysis.  J  Pharmacol  Exp  Ther 
271:1391-1398,  1994. 

Schuster,  C.R  Implications  of  laboratory 
research  for  the  treatment  of  drug  depen- 
dence. In:  Goldberg,  S.R,  and  Stolerman, 
LP.,  eds.  Behavioral  Analysis  of  Drug 
Dependence.  Orlando,  FL:  Academic 
Press,  1986.  pp.  357-385. 

Schuster,  C.R.,  and  Johanson,  C.E.  An 
analysis  of  drug-seeking  behavior  in  animals. 
Neurosci  Biobehav  Rev  5:315-323,  1981. 

Schwartz,  RD.;  Lehmann,  J.;  and  Kellar, 
K.J.  Presynaptic  nicotinic  cholinergic 
receptors  labeled  by  [3H]  acetylcholine  on 
catecholamine  and  serotonin  axons  in 
brain.  /  Neurochem  42:1495-1498,  1984. 


Segal,  D.S.,  and  Kuczenski,  R  Repeated 
cocaine  administration  induces  behavioral 
sensitization  and  corresponding  decreas- 
ing extracellular  dopamine  responses  in 
caudate  and  accumbens.  Brain  Res  577: 
351-355, 1992. 

Self,  D.W.,  and  Nestler,  E.J.  Molecular 
mechanisms  of  drug  reinforcement  and 
addiction.  Annu  Rev  Neurosci  18:463- 
495,  1995. 

Sellers,  E.M.;  Higgins,  G.A.;  and  Sobell, 
M.B.  5HT  and  alcohol  abuse.  Trends 
Pharmacol  Sci  13:69-75,  1992. 

Sershen,  LL;  Toth,  E.;  Lajtha,  A.;  and 
Vizi,  E.S.  Nicotine  effects  on  presynaptic 
receptor  interactions.  Ann  NT  Acad  Sci 
757:238-252,  1995. 

Shefner,  SA.  Electrophysiological  effects 
of  ethanol  on  brain  neurons.  In:  Watson, 
R.R.,  ed.  Biochemistry  and  Physiology  of 
Substance  Abuse.  Vol.  2.  Boca  Raton,  FL: 
CRC  Press,  1990.  p.  25. 

Shen,  R.Y.,  and  Chiodo,  LA.  Acute 
withdrawal  after  repeated  ethanol  treat- 
ment reduces  the  number  of  spontaneously 
active  dopaminergic  neurons  in  the  ven- 
tral tegmental  area.  Brain  Res  622:289- 
293,  1993. 

Sheng,  M.;  Cummings,  J.;  Roldan,  L.A.; 
Jan,  Y.N.;  and  Jan,  L.Y.  Changing  sub- 
unit  composition  of  heteromeric  NMDA 
receptors  during  development  of  rat 
cortex.  Nature  368:144-147,  1994. 

Siegel,  S.  Morphine  analgesic  tolerance: 
Its  situation  specificity  supports  a 
Pavlovian  conditioning  model.  Science 
193:323-325,  1976. 

Siegel,  S.J.;  Janssen,  W.G.;  Tullai,  J.W.; 
Rogers,  S.W.;  Moran,  T.;  Heinemann, 
S.F.;  and  Morrison,  J.H.  Distribution  of 
the  excitatory  amino  acid  receptor  subunits 
GluR2(4)  in  monkey  hippocampus  and 


179 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


colocalization  with  subunits  GluR5-7  and 
NMDARl.JNeurosci  15:2707-2719, 1995. 

Siggins,  G.R.;  Berger,  T.;  French,  E.D.; 
Shier,  T.;  and  Bloom,  F.  Ethanol,  salsoli- 
nol  and  tetrahydropapaveroline  alter  the 
discharge  of  neurons  in  several  brain 
regions:  Comparisons  to  opioid  effects. 
Prog  Clin  Biol  Res  90:275-287,  1982. 

Siggins,  G.R.;  Pittman,  Q.;  and  French, 
E.  Effects  of  ethanol  on  CM  and  CA3 
pyramidal  cells  in  the  hippocampal  slice 
preparation:  An  intracellular  study.  Brain 
Res.  414:22-34,  1987. 

Siggins,  G.R.;  Nie,  Z.;  Schweitzer,  P.; 
Madamba,  S.;  and  Henriksen,  S.J.  Electro- 
physiological interactions  of  ethanol  with 
central  neuropeptide  systems:  Opioids 
and  somatostatins.  In:  Deitrich,  R.,  and 
Erwin,  V.  eds.  Pharmacological  Effects 
of  Ethanol  on  the  Nervous  System.  Boca 
Raton,  FL:  CRC  Press,  1995.  p.  175. 

Siggins,  G.;  Nie,  Z.;  and  Madamba  S.  A 
metabotropic  hypothesis  for  ethanol 
sensitivity  of  GABAergic  and  glutamater- 
gic  central  synapses.  In:  Liu,  Y.,  and 
Hunt,  W. A.,  eds.  The  "Drunken"  Synapse: 
Studies  of  Alcohol  Related  Disorders.  New 
York:  Kluwer  Academic/Plenum 
Publishers,  1999.  pp.  135-143. 

Signs,  S.A.,  and  Schechter,  M.D.  The  role 
of  dopamine  and  serotonin  receptors  in  the 
mediation  of  the  ethanol  interoceptive  cue. 
Pharmacol  Biochem  Behav  30:55-64,  1988. 

Sinclair,  J.D.;  Le,  A.D.;  and  Kiianmaa,  K. 
The  AA  and  ANA  rat  lines,  selected  for 
differences  in  voluntary  alcohol  consump- 
tion. Experientia  45:798-805,  1989. 

Skattebol,  A.,  and  Rabin,  R.A.  Effects  of 
ethanol  on  45Ca2+  uptake  in  synaptosomes 
and  in  PC  12  cells.  Biochem  Pharmacol 
36:2227,  1987. 


Skolnick,  P.,  and  Paul,  S.M.  Molecular 
pharmacology  of  the  benzodiazepines. 
IntRev  Neurobiol  23:103-140,  1982. 

Smothers,  C;  Mrotek,  J.;  and  Lovinger, 
D.  Chronic  ethanol  exposure  leads  to  a 
selective  enhancement  of  N-  methyl-  D- 
aspartate  receptor  function  in  cultured 
hippocampal  neurons.  J  Pharmacol  Exp 
Ther 283:1214-1222,  1997. 

Snell,  L.D.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Radioligand  binding  to  the  N-methyl- 
D-aspartate  receptor/ionophore  complex: 
Alterations  by  ethanol  in  vitro  and  by 
chronic  in  vivo  ethanol  ingestion.  Brain 
Res  602:91-98,  1993. 

Snell,  L.D.;  Nunley,  K.R.;  Lickteig,  R.L.; 
Browning,  M.D.;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Regional  and  subunit 
specific  changes  in  NMDA  receptor 
mRNA  and  immunoreactivity  in  mouse 
brain  following  chronic  ethanol  ingestion. 
Mol  Brain  Res  40:71-78,  1996a. 

Snell,  L.D.;  Szabo,  G.;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Gangliosides  reduce  the 
development  of  ethanol  dependence 
without  affecting  ethanol  tolerance.  / 
Pharmacol  Exp  Ther  279:128-136, 
1996&. 

Sommer,  B.,  and  Seeburg,  P.H.  Glutamate 
receptor  channels:  Novel  properties  and 
new  clones.  Trends  Pharmacol  Sci  13:291- 
296,  1992. 

Sonnenberg,  J.L.;  Rauscher,  F.J.,  3d; 
Morgan,  J.I.;  and  Curran,  T.  Regulation 
of  proenkephalin  by  Fos  and  Jun.  Science 
246:1622-1625,  1989. 

Sorensen,  S.;  Palmer,  M.;  Dunwiddie,  T.; 
and  Hoffer,  B.  Electrophysiological  corre- 
lates of  ethanol-induced  sedation  in  dif- 
ferentially sensitive  lines  of  mice.  Science 
210:1143-1145,1980. 


180 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Spanagel,  R.;  Holter,  S.;  Allingham,  K.; 
Landgraf,  R.;  and  Zieglgansberger,  W. 
Acamprosate  and  alcohol:  I.  Effects  on 
alcohol  intake  following  alcohol  depriva- 
tion in  the  rat.  Eur  J  Pharmacol 
305:39^4,  1996. 

Speisky,  M.B.,  and  Kalant,  H.  Site  of 
interaction  of  serotonin  and  desglyci- 
namide-arginine  vasopressin  in  mainte- 
nance of  ethanol  tolerance.  Brain  Res 
326:281-290,  1985. 

Sprengel,  R,  and  Seeburg,  P.H.  The 
unique  properties  of  glutamate  receptor 
channels.  FEBS  Lett  325:90-94,  1993. 

Stea,  A.;  Soong,  T.W.;  and  Snutch,  T.P. 
Determinants  of  PKC-dependent  modu- 
lation of  a  family  of  neuronal  calcium 
channels.  Neuron  15:929-940,  1995. 

Steffensen,  S.,  and  Henriksen,  S.  Acute 
ethanol-induced  paradoxical  decrease  in 
recurrent  inhibition  in  the  dentate  gyrus 
of  chronically- treated,  ethanol-withdrawn 
rats.  Alcohol  Clin  Exp  Res  21:7 '5 A,  1997. 

Stevens,  C.F.  A  million  dollar  question:  Does 
LTP  =  memory?  Neuron  20:1-2,  1998. 

Stewart,  J.,  and  Badiani,  A.  Tolerance  and 
sensitization  to  the  behavioral  effects  of 
drugs.  Behav  Pharmacol 4:289-312,  1993. 

Sugihara,  H.;  Moriyoshi,  K;  Ishii,  T.; 
Masu,  M.;  and  Nakanishi,  S.  Structures 
and  properties  of  seven  isoforms  of  the 
NMDA  receptor  generated  by  alternative 
splicing.  Biochem  Biophys  Res  Commun 
185:826-832,  1992. 

Sunahara,  R.K.;  Dessauer,  C.W.;  and 
Gilman,  A.G.  Complexity  and  diversity  of 
mammalian  adenyl  cyclases.  Annu  Rev 
Pharmacol  Toxicol  36:461-480,  1996. 

Surmeier,  D.J.,  and  Kitai,  S.T.  Dl  and 
D2  dopamine  receptor  modulation  of 
sodium  and  potassium  currents  in  rat 


in  Res 


neostriatal  neurons.  Prog 
99:309-324,  1993. 

Surmeier,  D.J.,  and  Kitai,  S.T.  Dopa- 
minergic regulation  of  striatal  efferent 
pathways.  Curr  Opin  Neurobiol  4:915- 
919,  1994. 

Surmeier,  D.J.,  and  Kitai,  S.T.  State- 
dependent  regulation  of  neuronal  excitabil- 
ity by  dopamine.  Nihon  Shinkei  Seishin 
Takurigaku  Zasshi  17:105-110,  1997. 

Surmeier,  D.J.;  Bargas,  J.;  Hemmings, 
H.C.,  Jr.;  Nairn,  A.C.;  and  Greengard,  P. 
Modulation  of  calcium  currents  by  a  Dl 
dopaminergic  protein  kinase/phosphatase 
cascade  in  rat  neostriatal  neurons.  Neuron 
14:385-397,  1995. 

Suzdak,  P.D.;  Glowa,  J.R;  Crawley,  J.N.; 
Schwartz,  R.D.;  Skolnick,  P.;  and  Paul, 
S.M.  A  selective  imidazobenzodiazepine 
antagonist  of  ethanol  in  the  rat.  Science 
234:1243-1247,  1986. 

Suzdak,  P.S.;  Schwartz,  R.D.;  Skolnick, 
P.;  and  Paul,  S.M.  Alcohols  stimulate  g- 
aminobutyric  acid  receptor-mediated 
chloride  uptake  in  brain  vesicles:  Correlation 
with  intoxication  potency.  Brain  Res 
444:340-345,  1988. 

Swartzwelder,  H.S.;  Wilson,  W.A.;  and 
Tayyeb,  M.I.  Age -dependent  inhibition  of 
long-term  potentiation  by  ethanol  in  imma- 
ture versus  mature  hippocampus.  Alcohol 
Clin  Exp  Res  19:1480-1485,  1995. 

Szabo,  G.,  and  Hoffman,  P.L.  Brain- 
derived  neurotrophic  factor,  neuro- 
trophin-3  and  neurotrophin-4/5  maintain 
functional  tolerance  to  ethanol.  Eur  J 
Pharmacol  287 :35^1,  1995. 

Szabo,  G.;  Hoffman,  P.L.;  and  Tabakoff, 
B.  Forskolin  promotes  the  development  of 
ethanol  tolerance  in  6-hydroxydopamine- 
treated  mice.  Life  S«'42(6):615-621, 1988a. 


181 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Szabo,  G.;  TabakofF,  B.;  and  Hoffman, 
P.L.  Receptors  with  Vj  characteristics 
mediate  the  maintenance  of  ethanol  toler- 
ance by  vasopressin.  J  Pharmacol  Exp 
Ther  247:536-541,  19886. 

Szabo,  G.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Comparative  effects  of  arginine  vaso- 
pressin, [pGlu4,Cyt6]  arginine  vasopressin^ 
and  nerve  growth  factor  on  maintenance 
of  functional  tolerance  to  ethanol  in  mice. 
Eur  J  Pharmacol  199:131-134,  1991. 

Szabo,  G.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  The  NMD  A  receptor  antagonist 
dizocilpine  differentially  affects  environment- 
dependent  and  environment-independent 
ethanol  tolerance.  Psychopharmacology 
(Berl)  113:511-517, 1994. 

Szabo,  G.;  Nunley,  K.R.;  and  Hoffman, 
P.L.  Antisense  oligonucleotide  to  c-fos 
blocks  the  ability  of  arginine  vasopressin 
to  maintain  ethanol  tolerance.  Eur  J 
Pharmacol  306:67-72,  1996. 

Szmigielski,  A.;  Szmigielska,  H.;  and 
Wejman,  I.  The  effect  of  single  and  pro- 
longed ethanol  administration  on  the 
sensitivity  of  central  GABA-A  and  benzo- 
diazepine receptors  in  vivo.  Pol  J 
Pharmacol  Pharm  44:271-280,  1992. 

Tabakoff,  B.,  and  Hoffman,  P.L. 
Development  of  functional  dependence 
on  ethanol  in  dopaminergic  systems.  / 
Pharmacol  Exp  Ther  208:216-222,  1979. 

Tabakoff,  B.,  and  Hoffman,  P.L.  Alcohol: 
Neurobiology.  In:  Lowinson,  J.H.;  Ruiz, 
P.;  and  Miilman,  R.B.,  eds.  Substance 
Abuse:  A  Comprehensive  Textbook. 
Baltimore,  MD:  Williams  &  Wilkins, 
1992. p. 152. 

Tabakoff,  B.,  and  Hoffman,  P.L.  Alcohol 
addiction:  An  enigma  among  us.  Neuron 
16:909-912,  1996a. 


Tabakoff,  B.,  and  Hoffman,  P.L.  Ethanol 
and  glutamate  receptors.  In:  Deitrich, 
R.A.,  and  Erwin,  V.G.,  eds.  Pharma- 
cological Effects  of  Ethanol  on  the  Nervous 
System.  Boca  Raton,  FL:  CRC  Press, 
19966.  p.  73. 

Tabakoff,  B.,  and  Hoffman,  P.L.  Adenylyl 
cyclases  and  alcohol.  In:  Cooper,  D.M.F.,  ed. 
Advances  in  Second  Messenger  and  Phospho- 
protein  Research.  Vol.  32.  Philadelphia: 
Lippincott-Raven,  1998.  p.  173. 

Tabakoff,  B.,  and  Ritzmann,  RF.  The 
effects  of  6-hydroxydopamine  on  tolerance 
to  and  dependence  on  ethanol.  / 
Pharmacol  Exp  Ther  203:319-332,  1977. 

Tabakoff,  B.,  and  Rothstein,  J.D.  Biology 
of  tolerance  and  dependence.  In:  Tabakoff, 
B.;  Sutker,  P.B.;  and  Randall,  C.L.,  eds. 
Medical  and  Social  Aspects  of  Alcohol 
Abuse.  New  York:  Plenum  Press,  1983. 
pp. 187-220. 

Tabakoff,  B.;  Hoffman,  P.;  and  Moses,  F. 
Neurochemical  correlates  of  ethanol 
withdrawal:  Alterations  in  serotonergic 
function.  J  Pharm  Pharmacol  29:471- 
476,  1977. 

Tabakoff,  B.;  Whelan,  J. P.;  Ovchinnikova, 
L.;  Nhamburo,  P.;  Yoshimura,  M.;  and 
Hoffman,  P.L.  Quantitative  changes  in  G 
proteins  do  not  mediate  ethanol-induced 
downregulation  of  adenylyl  cyclase  in 
mouse  cerebral  cortex.  Alcohol  Clin  Exp 
Res  19:187-194, 1995. 

Takadera,  T.;  Suzuki,  R.;  and  Mohri,  T. 
Protection  by  ethanol  of  cortical  neurons 
from  N-methyl-D-aspartate-induced 
neurotoxicity  is  associated  with  blocking 
calcium  influx.  Brain  Res  537:109-115, 
1990. 

Tang,  M.;  Brown,  C;  and  Falk,  J.L.  Com- 
plete reversal  of  chronic  ethanol  polydipsia 


182 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


by  schedule  withdrawal.  Pharmacol 
BiochemBehav  16:155-158,  1982. 

Thomas,  M.P.;  Davis,  M.I.;  Monaghan, 
D.T.;  and  Morrisett,  RA.  Organotypic 
brain  slice  cultures  for  functional  analysis 
of  alcohol-related  disorders:  Novel  versus 
conventional  preparations.  Alcohol  Clin 
Exp  Res  22: 5 1-59,  1998. 

Thyagarajan,  R,  and  Ticku,  M.K.  The  effect 
of  in  vitro  and  in  vivo  ethanol  administra- 
tion on  [35S]t-butylbicyclophosphorothi- 
onate  binding  in  C57  mice.  Brain  Res 
Bull  15:343-345,  1985. 

Ticku,  M.K.  Ethanol  and  the  benzodi- 
azepine-GABA  receptor-ionophore  com- 
plex. Experientia  45:413^118,  1989. 

Ticku,  M.K.,  and  Burch,  T.  Alterations  in 
GABA  receptor  sensitivity  following  acute 
and  chronic  ethanol  treatments.  J 
Neurochem  34:417-423,  1980. 

Tran,  V.T.;  Snyder,  S.H.;  Major,  L.F.; 
and  Hawley,  R.J.  GABA  receptors  are 
increased  in  brains  of  alcoholics.  Ann 
Neurol  9:289-292,  1981. 

Treistman,  S.N.,  and  Wilson,  A.  Effects  of 
chronic  ethanol  on  currents  carried 
through  calcium  channels  in  Aplysia. 
Alcohol  Clin  Exp  Res  15:489^93,  1991 . 

Tremwel,  M.F.,  and  Hunter,  B.E.  Effects 
of  chronic  ethanol  ingestion  on  long-term 
potentiation  remain  even  after  a  prolonged 
recovery  from  ethanol  exposure.  Synapse 
17:141-148,  1994. 

Tremwel,  M.F.;  Anderson,  K.J.;  and 
Hunter,  B.E.  Stability  of  [3H]MK-801 
binding  sites  following  chronic  ethanol 
consumption.  Alcohol  Clin  Exp  Res 
18:1004-1008, 1994a. 

Tremwel,  M.F.;  Hunter,  B.E.;  and  Peris, 
J.  Chronic  ethanol  exposure  enhances 
[3H]GABA  release  and  does  not  affect 


GABAA  receptor  mediated  36C1  uptake. 
Synapse  17:149-154,  1994&. 

Trevisan,  L.;  Fitzgerald,  L.W.;  Brose,  N.; 
Gasic,  G.P.;  Heinemann,  S.F.;  Duman,  R.S.; 
and  Nestler,  E.J.  Chronic  ingestion  of 
ethanol  up-regulates  NMD  ART  receptor 
subunit  immunoreactivity  in  rat  hippo- 
campus. JNeurochem  62:1635-1638, 1994. 

Trifunovic,  R.D.,  and  Brodie,  M.S.  The 
effects  of  clomipramine  on  the  excitatory 
action  of  ethanol  on  dopaminergic  neu- 
rons of  the  ventral  tegmental  area  in  vitro. 
J  Pharmacol  Exp  Ther  276:34-40,  1996. 

Trulson,  M.E.;  Joe,  J.C.;  Babb,  S.;  and 
Raese,  J.D.  Chronic  cocaine  administration 
depletes  tyrosine  hydroxylase  immunore- 
activity in  the  mesolimbic  dopamine 
system  in  rat  brain:  Quantitative  light 
microscopic  studies.  Brain  Res  Rev 
3:141-152,  1987. 

Trzaskowska,  E.;  Pucilowski,  O.;  Dyr,  W.; 
Kostowski,  W.;  and  Hauptmann,  M. 
Suppression  of  ethanol  tolerance  and 
dependence  in  rats  treated  with  DSP-4,  a 
noradrenergic  neurotoxin.  Drug  Alcohol 
Depend  18:349-353,  1986. 

Twombly,  D.A.;  Herman,  M.D.;  Kye, 
C.H.;  and  Narahashi,  T.  Ethanol  effects 
on  two  types  of  voltage-activated  calcium 
channels.  J  Pharmacol  Exp  Ther  254:1029- 
1037,  1990. 

Uchimura,  N.;  Higashi,  H.;  and  Nishi,  S. 
Hyperpolarizing  and  depolarizing  actions 
of  dopamine  actions  of  dopamine  via  D-l 
and  D-2  receptors  on  nucleus  accumbens 
neurons.  Brain  Res  375:368-372,  1986. 

Unwin,  J.W.,  and  Taberner,  P.V.  Sex  and 
strain  differences  in  GABA  receptor  bind- 
ing after  chronic  ethanol  drinking  in  mice. 
Neuropharmacology  19:1257-1259,  1980. 

Valenzuela,  C.F.;  Machu,  T.K.;  McKernan, 
R.M.;  Whiting,  P.;  VanRenterghem,  B.B.; 


183 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


McManaman,  J.L.;  Brozowski,  S.J.; 
Smith,  G.B.;  Olsen,  R.W.;  and  Harris, 
R.A.  Tyrosine  kinase  phosphorylation  of 
GABAA  receptors.  Mol  Brain  Res 
31:165-172,1995. 

Valverius,  P.;  Hoffman,  P.L.;  and 
Tabakoff,  B.  Hippocampal  and  cerebellar 
(3-adrenergic  receptors  and  adenylate 
cyclase  are  differentially  altered  by  chronic 
ethanol  ingestion.  J  Neurochem  52:492- 
497,  1989. 

Valverius,  P.;  Crabbe,  J.C.;  Hoffman, 
P.L.;  and  Tabakoff,  B.  NMDA  receptors 
in  mice  bred  to  be  prone  or  resistant  to 
ethanol  withdrawal  seizures.  Eur  J 
Pharmacol  184:185-189,  1990. 

Van  Bockstaele,  E.J.;  Biswas,  A.;  and 
Pickel,  V.M.  Topography  of  serotonin 
neurons  in  the  dorsal  raphe  nucleus  that 
send  axon  collaterals  to  the  rat  prefrontal 
cortex  and  nucleus  accumbens.  Brain  Res 
624:188-198,  1993. 

Van  Bockstaele,  E.J.;  Cestari,  D.M.;  and 
Pickel,  V.M.  Synaptic  structure  and  con- 
nectivity of  serotonin  terminals  in  the 
ventral  tegmental  area:  Potential  sites  for 
modulation  of  mesolimbic  dopamine 
neurons.  Brain  Res  647:307-322,  1994. 

Vizi,  E.S.;  Sershen,  H.;  Balla,  A.; 
Windisch,  K.;  Jurany,  Z.;  and  Lajtha,  A. 
Neurochemical  evidence  of  heterogeneity 
of  presynaptic  and  somatodendritic  nico- 
tinic cholinergic  receptors.  Ann  NT  Acad 
Sci  757:84-99,  1995. 

Volicer,  L.  GABA  levels  and  receptor 
binding  after  acute  and  chronic  ethanol 
administration.  Brain  Res  Bull  5(Suppl 
2):809-813,  1980. 

Volicer,  L.,  and  Biagioni,  T.M.  Effect  of 
ethanol  administration  and  withdrawal  on 
GABA  receptor  binding  in  rat  cerebral 


cortex.  Subst  Alcohol  Actions  Misuse 
3:31-39,  1982. 

Volkow,  N.D.;  Wang,  G.J.;  Overall,  J.E.; 
Hitzemann,  R;  Fowler,  J.S.;  Pappas,  N.; 
Frecska,  E.;  and  Piscani,  K.  Regional 
brain  metabolic  response  to  lorazepam  in 
alcoholics  during  early  and  late  alcohol 
detoxification.  Alcohol  Clin  Exp  Res 
21:1278-1284,  1997. 

Volpicelli,  J.R.;  Alterman,  A.I.;  Hayashida, 
M.;  and  O'Brien,  C.P.  Naltrexone  in  the 
treatment  of  alcohol  dependence.  Arch 
Gen  Psychiatry  49:876-880,  1992. 

Walker,  D.W.;  Barnes,  D.E.;  Zornetzer, 
S.F.;  Hunter,  B.E.;  and  Kubanis,  P. 
Neuronal  loss  in  hippocampus  induced  by 
prolonged  ethanol  consumption  in  rats. 
Science  209:711-713,  1980. 

Waller,  M.B.;  Murphy,  J.M.;  McBride, 
W.J.;  Lumeng,  L.;  and  Li,  T.-K.  Effect  of 
low  dose  ethanol  on  spontaneous  motor 
activity  in  alcohol-preferring  and  -nonpre- 
ferring  lines  of  rats.  Pharmacol  Biochem 
Behav  24:617-623,  1986. 

Wan,  F.J.;  Berton,  F.;  Madamba,  S.G.; 
Francesconi,  W.;  and  Siggins,  G.R.  Low 
ethanol  concentrations  enhance  GABAergic 
inhibitory  postsynaptic  potentials  in  hip- 
pocampal pyramidal  neurons  only  after 
block  of  GABAB  receptors.  Proc  Natl 
Acad  Sci  USA  93:5049-5054,  1996. 

Wand,  G.S.,  and  Levine,  MA.  Hormonal 
tolerance  to  ethanol  is  associated  with 
decreased  expression  of  the  GTP-binding 
protein,  Gsa,  and  adenylyl  cyclase  activity 
in  ethanol-treated  mice.  Alcohol  Clin  Exp 
Res  15:705-710,  1991. 

Wand,  G.S.;  Diehl,  A.M.;  Levine,  M.A; 
Wolfgang,  D.;  and  Samy,  S.  Chronic 
ethanol  treatment  increases  expression  of 
inhibitory  G  proteins  and  reduces  adeny- 
lylcyclase  activity  in  the  central  nervous 


184 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


system  of  two  lines  of  ethanol-sensitive 
mice.  J  Biol  Chem  268:2595-2601,  1993. 

Wang,  J.Q.;  Daunais,  J.B.;  and  McGinty, 
J.F.  NMDA  receptors  mediate  ampheta- 
mine-induced upregulation  of  zif/268  and 
preprodynorphin  mRNA  expression  in  rat 
striatum.  Synapse  18:343-353,  1994. 

Wang,  R.A.;  Cheng,  G.;  Kolaj,  M.;  and 
Randic,  M.  ci-Subunit  of  calcium/ 
calmodulin-dependent  protein  kinase  II 
enhances  Y-aminobutyric  acid  and  inhibitory 
synaptic  responses  of  rat  neurons  in  vitro. 
J  Neurophysiol  73:2099-2106,  1995. 

Wang,  X.;  Dayanithi,  G.;  Lemos,  J.R.; 
Nordmann,  J.J.;  and  Treistman,  S.N. 
Calcium  currents  and  peptide  release  from 
neurohypophysial  terminals  are  inhibited 
by  ethanol.  J  Pharmacol  Exp  Ther  259:705- 
711,1991. 

Wang,  X.;  Wang,  G.;  Lemos,  J.R.;  and 
Treistman,  S.N.  Ethanol  directly  modu- 
lates gating  of  a  dihydropyridine- sensitive 
Ca2+  channel  in  neurohypophysial  termi- 
nals. JNeurosci  14:5453-5460,  1994. 

Watson,  W.P.;  Misra,  A.;  Cross,  A.J.; 
Green,  A.R.;  and  Little,  H.J.  The  differ- 
ential effects  of  felodipine  and  nitrendip- 
ine on  cerebral  dihydopyridine  binding  ex 
vivo  and  the  ethanol  withdrawal  syndrome. 
Br  J  Pharmacol  112:1017-1024,  1994. 

Weight,  F.F.  Cellular  and  molecular 
physiology  of  alcohol  actions  in  the 
nervous  system.  Int  Rev  Neurobiol 
33:289-348,  1992. 

Weiner,  J.L.;  Zhang,  L.;  and  Carlen,  P.L. 
Potentiation  of  GABAA-mediated  synap- 
tic current  by  ethanol  in  hippocampal 
CA1  neurons:  Possible  role  of  protein 
kinase  C.  J  Pharmacol  Exp  Ther 
268:1388-1395,  1994. 

Weiner,  J.L.;  Valenzuela,  C.F.;  Watson,  P.L.; 
Frazier,  C.J.;  and  Dunwiddie,  T.V.  Elevation 


of  basal  protein  kinase  C  activity  increases 
ethanol  sensitivity  of  GABA(A)  receptors 
in  rat  hippocampal  CA1  pyramidal  neu- 
rons. JNeurochem  68:1949-1959,  1997. 

Weiss,  F.;  Mitchiner,  M.;  Bloom,  F.E.; 
and  Koob,  G.F.  Free  choice  responding 
for  ethanol  versus  water  in  alcohol  prefer- 
ring (P)  and  unselected  Wistar  rats  is 
differentially  modified  by  naloxone, 
bromocriptine  and  methysergide. 
Psychopharmacology  1101:178-186,  1990. 

Weiss,  F.;  Lorang,  M.T.;  Bloom,  F.E.; 
and  Koob,  G.F.  Oral  alcohol  self-adminis- 
tration stimulates  dopamine  release  in  the 
rat  nucleus  accumbens:  Genetic  and  moti- 
vational determinants.  J  Pharmacol  Exp 
Ther  267:250-258,  1993. 

Weiss,  F.;  Parsons,  L.H.;  Schulteis,  G.; 
Lorang,  M.T.;  Hyytia,  P.;  Bloom,  F.E.; 
and  Koob,  G.F.  Ethanol  self- administra- 
tion restores  withdrawal-associated  defi- 
ciencies in  accumbal  dopamine  and 
serotonin  release  in  dependent  rats.  / 
Neurosci  16:3474-3485,  1996. 

Welzl,  H.;  Batting,  K;  and  Berz,  S.  Acute 
effects  of  nicotine  injection  into  the  nucleus 
accumbens  on  locomotor  activity  in  nicotine- 
naive  and  nicotine-tolerant  rats.  Pharma- 
col Biochem  Behav  37:743-746,  1990. 

Westerling,  P.;  Lindgren,  S.;  and  Meyerson, 
B.  Functional  changes  in  GABAA  receptor 
stimulation  during  the  oestrous  cycle  of  the 
rat.  Br  J  Pharmacol  103:1580-1584, 1991. 

White,  F.J.;  Hu,  X.-T.;  Zhang,  X.-F.;  and 
Wolf,  M.E.  Repeated  administration  of 
cocaine  or  amphetamine  alters  neuronal 
responses  to  glutamate  in  the  mesoac- 
cumbens  dopamine  system.  J  Pharmacol 
Exp  Ther  273:445^54, 1995. 

White,  G.;  Lovinger,  D.;  and  Weight,  F. 
Ethanol  inhibits  NMDA- activated  current 
but  does  not  alter  GAB  A- activated  current 


185 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  an  isolated  adult  mammalian  neuron. 
Brain  Res  507:332-336,  1990. 

Whittemore,  E.R.;  Yang,  W.;  Drewe,  J. A.; 
and  Woodward,  R.M.  Functional  and 
pharmacological  characterization  of  the 
human  GABAA  receptor  ct4  subunit  ex- 
pressed in  Xenopus  oocytes.  Soc  Neurosci 
Abstr  21:850,  1996. 

Whittington,  M.A.,  and  Little,  H.J.  A 
calcium  channel  antagonist  stereoselectively 
decreases  ethanol  withdrawal  hyperex- 
citability  but  not  that  due  to  bicuculline, 
in  hippocampal  slices.  Br  J  Pharmacol 
103:1313-1320,  1991. 

Whittington,  M.A.,  and  Litde,  H.J. 
Changes  in  voltage-operated  calcium 
channels  modify  ethanol  withdrawal  hy- 
perexcitability  in  mouse  hippocampal 
slices.  Exp  Physiol  78:347-370,  1993. 

Whittington,  M.A.;  Dolin,  S.J.;  Patch, 
T.L.;  Siarey,  R.J.;  Butterworth,  A.R.;  and 
Little,  H.J.  Chronic  dihydopyridine  treat- 
ment can  reverse  the  behavioral  conse- 
quences of  and  prevent  adaptations  to 
chronic  ethanol  treatment.  Br  J 
Pharmacol  103:1669-1676,  1991. 

Whittington,  M.A.;  Little,  H.J.;  and 
Lambert,  J.D.  Changes  in  intrinsic  inhibi- 
tion in  isolated  hippocampal  slices  during 
ethanol  withdrawal;  lack  of  correlation 
with  withdrawal  hyperexcitability.  Br  J 
Pharmacol  107 :521-527,  1992. 

Whittington,  M.A.;  Lambert,  J.D.;  and 
Little,  H.J.  Increased  NMDA  receptor 
and  calcium  channel  activity  underlying 
ethanol  withdrawal  hyperexcitability. 
Alcohol  Alcohol  30:105-114,  1995. 

Widdowson,  P.S.,  and  Holman,  R.B. 
Ethanol -induced  increase  in  endogenous 
dopamine  release  may  involve  endogenous 
opiates.  J  Neurochem  59:157-163,  1992. 


Williams,  K.L.;  Ferko,  A.P.;  Barbieri,  E.J; 
and  Digregorio,  G.J.  Glycine  enhances 
the  central  depressant  properties  of  ethanol 
in  mice.  Pharmacol  Biochem  Behav  50:199- 
205,  1995. 

Wilson,  M.A.  Influences  of  gender,  go- 
nadectomy,  and  estrous  cycle  on  GABA/ 
BZ  receptors  and  benzodiazepine  responses 
in  rats.  Brain  Res  Bull  29:165-172,  1992. 

Wise,  R.A.,  and  Leeb,  K.  Psychomotor- 
stimulant  sensitization:  A  unitary  phenom- 
enon? Behav  Pharmacol 4:339-349,  1993. 

Wonnacott,  S.;  Drasdo,  A.;  Sanderson,  E.; 
and  Rowell,  P.  Presynaptic  nicotinic  re- 
ceptors and  the  modulation  of  transmitter 
release.  In:  Bock,  G.,  and  Marsh,  J.,  eds. 
The  Biology  of  Nicotine  Dependence.  Ciba 
Foundation  Symposium  No.  152. 
Chichester,  England:  John  Wiley  &  Sons, 
1989.  p.  87. 

Woo,  E.,  and  Greenblatt,  D.J.  Massive 
benzodiazepine  requirements  during 
acute  alcohol  withdrawal.  Am  J  Psychiatry 
136:821-823,  1979. 

Woods,  J.H.,  and  Winger,  G.D.  A  critique 
of  methods  for  inducing  ethanol  self- 
intoxication  in  animals.  In:  Mello,  N.K., 
and  Mendelson,  J.H.,  eds.  Recent  Advances 
in  Studies  of  Alcoholism;  an  Interdisciplinary 
Symposium.  DHEW  Pub.  No.  (HSM)71- 
9045.  Washington,  DC:  U.S.  Government 
Printing  Office,  1971.  p.  413. 

Woodward,  D.;  Janak,  P.;  and  Chang,  J.- 
Y.  Ethanol  action  on  neural  networks 
studied  with  multineuron  recording  in 
freely  moving  animals.  Alcohol  Clin  Exp 
Res  22:10-22,  1988. 

World  Health  Organization.  The  ICD-10 

Classification  of  Mental  and  Behavioural 
Disorders:  Clinical  Descriptions  and 
Diagnostic  Guidelines.  Geneva:  the 
Organization,  1992. 


186 


Neuroadaptation  to  Ethanol  at  the  Molecular  and  Cellular  Levels 


Wu,  P.H.;  Mihic,  S.J.;  Liu,  J.F.;  Le,  A.D.; 
and  Kalant,  H.  Blockade  of  chronic  tolerance 
to  ethanol  by  the  NMD  A  antagonist,  (+)MK- 
801.  Eur  J  Pharmacol  231:157-164, 1993. 

Wu,  P.H.;  Liu,  J.F.;  Wu,  W.L.;  Lanca, 
A.J.;  and  Kalant,  H.  Development  of 
alcohol  tolerance  in  the  rat  after  a  single 
exposure  to  combined  treatment  with 
arginine8 -vasopressin  and  ethanol.  J 
PharmacolExp  Ther 276:1283-1291, 1996. 

Wu,  Z.-L.;  Thomas,  S.A.;  Villachres, 
E.C.;  Zia,  Z.;  Simmons,  M.L.;  Chavkin, 
C;  Palmiter,  R.D.;  and  Storm,  D.R. 
Altered  behavior  and  long-term  potentia- 
tion in  type  I  adenylyl  cyclase  mutant 
mice.  Proc  Natl  Acad  Sci  USA  92:220- 
224,  1995. 

Xiao,  X.;  McCown,  T.J.;  Li,  J.;  Breese, 
G.R.;  Morrow,  A.L.;  and  Samulski,  RJ. 
Adeno-  associated  virus  (AAV)  vector 
antisense  gene  transfer  in  vivo  decreases 
GABAA  cii  containing  receptors  and 
increases  inferior  collicular  seizure  sensi- 
tivity. Brain  Res  756:76-83,  1997. 

Yamamura,  T.;  Hishida,  S.;  Hatake,  K.; 
Taniguchi,  T.;  and  Ouchi,  H.  Effects  of 
methamphetamine  and  ethanol  on  learning 
and  brain  neurotransmitters  in  rats.  Pharma- 
col Biochem  Behav  42:389^00,  1992. 

Yan,  K.;  Greene,  E.;  Belga,  F.;  and 
Rasenick,  M.M.  Synaptic  membrane  G 
proteins  are  complexed  with  tubulin  in 
situ.  J  Neurochem  66:1489-1495,  1996. 

Yan,  Z.,  and  Surmeier,  D.J.  D5  dopamine 
receptors  enhance  Zn2 -(--sensitive  GABA(A) 
currents  in  striatal  cholinergic  interneurons 
through  a  PKA/PP1  cascade.  Neuron 
19:1115-1126,  1997. 

Yan,  Z.;  Song,  W.J.;  and  Surmeier,  J.  D2 
dopamine  receptors  reduce  N-type  Ca2+ 
currents  in  rat  neostriatal  cholinergic 
interneurons  through  a  membrane-delim- 


ited, protein-kinase-C-insensitive  path- 
way. /  Neurophysiol  77:1003-1015,  1997. 

Yang,  X.;  Criswell,  H.E.;  and  Breese, 
G.R  Nicotine-induced  inhibition  in 
medial  septum  involves  activation  of 
presynaptic  nicotinic  cholinergic  receptors 
on  gamma-  aminobutyric  acid-containing 
neurons.  J  Pharmacol  Exp  Ther  276: 
482^89,  1996*. 

Yang,  X.;  Diehl,  A.M.;  and  Wand,  G.S. 
Ethanol  exposure  alters  the  phosphoryla- 
tion of  cyclic  AMP  responsive  element 
binding  protein  and  cyclic  AMP  respon- 
sive element  binding  activity  in  rat  cere- 
bellum. J  Pharmacol  Exp  Ther  278:338- 
346,  19966. 

Yang,  X.;  Horn,  K.;  Baraban,  J.M.;  and 
Wand,  G.S.  Chronic  ethanol  administra- 
tion decreases  phosphorylation  of  cyclic 
AMP  response  element- binding  protein  in 
granule  cells  of  rat  cerebellum.  J 
Neurochem  70:224-232,  1998. 

Yoneda,  Y.;  Enomoto,  R;  and  Kiyokazu, 
O.  Supporting  evidence  for  negative  modu- 
lation by  protons  of  an  ion  channel  associ- 
ated with  the  N-methyl-D- aspartate 
receptor  complex  in  rat  brain  using  ligand 
binding  techniques.  Brain  Res  636: 
298-307,  1994. 

Yoshimoto,  K.,  and  McBride,  W.J. 
Regulation  of  nucleus  accumbens 
dopamine  release  by  the  dorsal  raphe 
nucleus  in  the  rat.  Neurochem  Res 
17:401^07,  1992. 

Yoshimoto,  K.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Alcohol  stimulates  the 
release  of  dopamine  and  serotonin  in  the 
nucleus  accumbens.  Alcohol  9:17 -22,  1991. 

Yoshimoto,  K.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Ethanol  enhances  the 
release  of  dopamine  and  serotonin  in  the 
nucleus  accumbens  of  ELAD  and  LAD 


187 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


lines  of  rats.  Alcohol  Clin  Exp  Res 
16:781-785,  1992. 

Yoshimura,  M.,  and  Tabakoff,  B.  Selective 
effects  of  ethanol  on  the  generation  of 
cAMP  by  particular  members  of  the 
adenylyl  cyclase  family.  Alcohol  Clin  Exp 
Res  19:1435-1440,  1995. 

Yuan,  X.;  Madamba,  S.G.;  and  Siggins, 
G.R.  Opioid  peptides  reduce  synaptic 
transmission  in  the  nucleus  accumbens. 
NeurosciLett  134:223-228,  1992. 

Zeise,  M.L.;  Kasparow,  S.;  Capogna,  M.; 
and  Zieglgansberger,  W.  Calcium  di- 
acetylhomotaurinate  (Ca-AOTA)  de- 
creases the  action  of  excitatory  amino 
acids  in  the  rat  neocortex  in  vitro.  Prog 
Clin  Biol  Res  351:237-242,  1990. 


Zeise,  M.L.;  Kasparow,  S.;  Capogna,  M.; 
and  Zieglgansberger,  W.  Acamprosate 
(calcium  acetylhomotaurinate)  decreases 
postsynaptic  potentials  in  the  rat  neocor- 
tex: Possible  involvement  of  excitatory 
amino  acid  receptors.  Eur  J  Pharmacol 
231:47-52,1993. 

Zhou,  A.-W.;  Li,  W.-X.;  Guo,  J.;  and  Du, 
Y.-C.  Facilitation  of  AVP(4-8)  on  gene 
expression  of  BDNF  and  NGF  in  rat 
brain.  Peptides  18:1179-1187,  1997. 

Zhu,  W.J.;  Vicini,  S.;  Harris,  B.T.;  and 
Grayson,  D.R.  NMDA-mediated  modula- 
tion of  gamma-  aminobutyric  acid  type  A 
receptor  function  in  cerebellar  granule 
neurons.  J  Neurosci  15:7692-7701,  1995. 


Chapter  5 

Neurotoxicity  of  Alcohol:  Excitotoxicity, 
Oxidative  Stress,  Neurotrophic  Factors, 
Apoptosis,  and  Cell  Adhesion  Molecules 

Fulton  T.  Crews,  Ph.D. 


KEY  WORDS:  toxic  drug  effect;  AODR  (AOD  [alcohol  or  other  drug]  related) 
disorder;  neuron;  chronic  AODE  (effects  of  AOD  use,  abuse,  and  dependence); 
brain  damage;  NMD  A  receptors;  physiological  stress;  oxidation-reduction;  growth 
promoting  factors;  cytolysis;  literature  review 


Studies  of  alcohol-induced  brain  dam- 
age have  clearly  indicated  that  alcohol 
is  neurotoxic.  Alcoholics  are  at 
increased  risk  for  brain  damage  from  a 
variety  of  causes,  including  poor 
nutrition,  liver  disease,  and  head 
trauma.  Furthermore,  alcoholic 
dementia  is  the  second  leading  cause 
of  adult  dementia  in  the  United 
States,  accounting  for  approximately 
10  percent  of  the  cases  (Alzheimer's 
disease  is  the  leading  cause,  account- 
ing for  40  to  60  percent  of  cases).  A 
variety  of  studies  report  that  50  to  75 
percent  of  sober,  detoxified,  long- 
term  alcohol-dependent  individuals 
suffer  from  some  degree  of  detectable 


cognitive  impairment,  with  approxi- 
mately 10  percent  suffering  from  serious 
dementia  (Martin  et  al.  1986;  Char- 
ness  1993;  Dufour  1993).  Although 
more  research  is  required  to  precisely 
delineate  the  effects  of  alcohol  on  var- 
ious types  of  brain  function,  there 
appears  to  be  a  continuum  of  moderate 
deficits  in  the  majority  of  long-term 
alcoholics,  progressing  to  much  more 
severe  deficits  of  Wernicke's  disease 
and  Wernicke's  encephalopathy  with 
Korsakoff's  amnestic  syndrome  (But- 
terworth  1995;  Pfefferbaum  et  al. 
1996).  A  variety  of  lifestyle  factors, 
including  nutrition,  are  implicated  in 
the  more  severe  cases.  However,  all  of 


F.T.  Crews,  Ph.D.,  is  director  of  the  Center  for  Alcohol  Studies  and  professor  of  psychiatry  and 
pharmacology  at  the  University  of  North  Carolina  at  Chapel  Hill,  CB#7178,  Thurston  Bowles  Bldg., 
Chapel  Hill,  NC  27599-7178. 


189 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


these  cases  on  the  continuum  appear  to 
be  related  to  alcohol  consumption  and 
to  amount  of  alcohol  consumed.  That 
is,  the  more  severe  cases  are  associated 
with  more  severe  and  chronic  long- 
term  alcoholism  (Butterworth  1995; 
Pfefferbaum  et  al.  1996). 

Alcohol-induced  changes  in  the 
structure  of  adult  brain  have  been 
studied  in  both  humans  and  rodents 
(Charness  1993).  A  variety  of  post- 
mortem histological  analyses  as  well  as 
supporting  imaging  analyses  suggest 
that  chronic  alcohol  changes  brain 
structure.  Computed  tomography 
(CT)  and  magnetic  resonance  imag- 
ing (MR1)  studies  of  human  brain 
have  repeatedly  shown  enlargement 
of  the  cerebral  ventricles  and  sulci  in 
most  alcoholics.  The  enlargement  of 
the  ventricles  and  sulci  essentially 
reflect  a  shrinking  of  the  brain  mass. 
This  is  consistent  with  studies  on 
postmortem  brain  tissue  showing  that 
alcoholics  have  a  reduction  in  total 
brain  weight.  In  particularly  severe 
alcoholics,  global  cerebral  hemisphere 
and  cerebellar  brain  weights  are  sig- 
nificantly reduced  compared  with 
control  subjects  and  moderate 
drinkers  (Harper  and  Kril  1993). 
Some  of  this  loss  of  brain  mass  is 
likely  due  to  actual  loss  of  neurons 
and  resulting  loss  of  myelin  sheath 
white  matter,  which  normally  envelops 
neuronal  extensions.  However,  a  por- 
tion of  the  loss  in  brain  mass  is  also 
likely  to  be  due  to  a  reduction  in  the 
brain  parenchyma — that  is,  the  size  of 
the  cells  and  their  processes — during 
chronic  alcohol  abuse.  Studies  have 
indicated  that  within  1-5  months  of 
recovery  from  alcoholism,  and  with 


sustained  abstinence,  the  size  of  the 
brain  returns  toward  normal  levels 
(Muuronen  et  al.  1989;  Pfefferbaum 
and  Rosenbloom  1993;  Pfefferbaum 
et  al.  1998).  It  is  likely  that  this  return 
involves  an  increase  in  neuronal  cell 
size,  arborization,  and  density  of  the 
neuronal  processes  that  make  up  cel- 
lular brain  mass,  as  well  as  increases  in 
the  number  and  size  of  glial  cells 
(Franke  et  al.  1997).  Although  it  is  not 
clear  exactly  how  alcoholism  leads  to  a 
reduction  in  brain  weight  and  volume,  it 
is  clear  that  this  does  occur  during  active 
alcohol  abuse,  and  that  some  recovery 
of  brain  mass  does  occur  during  absti- 
nence. More  studies  are  needed  to  more 
clearly  understand  the  mechanisms 
underlying  these  events. 

Some  studies  have  focused  on  the 
frontal  lobes  as  being  particularly  sen- 
sitive to  alcohol-induced  changes 
(Jernigan  et  al.  1991).  Quantitative 
morphometry  suggests  that  the  frontal 
lobes  of  the  human  brain  show  the 
greatest  loss  and  account  for  much  of 
the  associated  ventricular  enlarge- 
ment. Specific  types  of  brain  cells 
appear  to  be  disrupted.  Both  gray 
matter,  which  is  composed  largely  of 
neurons,  and  white  matter,  which 
involves  neuronal  tracks  surrounded 
by  myelin  sheaths,  appear  to  be 
decreased.  Harper  and  colleagues 
(1987)  found  that  neuronal  density  in 
the  superior  frontal  cortex  was 
reduced  by  22  percent  in  alcoholics 
compared  with  nonalcoholic  control 
subjects,  in  contrast  to  other  areas  of 
the  cortex,  which  were  not  different 
between  the  groups.  Furthermore,  the 
complexity  of  the  basal  dendritic 
arborization  of  layer  III  pyramidal 


190 


Neurotoxicity  of  Alcohol 


cells  in  both  superior  frontal  and  motor 
cortices  was  significantly  reduced  in 
alcoholics  compared  with  control  sub- 
jects. A  reduction  in  dendritic  arboriza- 
tion of  Purkinje  cells  in  the  anterior 
superior  vermis  of  the  cerebellum 
was  also  found  in  alcoholics.  Taken 
together,  the  data  demonstrate  a 
selective  neuronal  loss,  dendritic  sim- 
plification, and  reduction  of  synaptic 
complexity  in  specific  brain  regions  of 
alcoholics.  It  is  uncertain  how  these 
cellular  lesions  relate  to  selective  loss 
of  white  matter  that  appears  to  occur 
particularly  in  frontal  lobes.  One  rea- 
son these  frontal  lobe  changes  are 
more  evident  is  the  greater  proportion 
of  white  matter  to  cortical  gray  matter 
in  the  frontal  regions.  Frontal  lobe 
shrinkage  has  been  reported  with  or 
without  seizures,  with  some  studies 
suggesting  that  temporal  lobe  shrink- 
age occurs  particularly  in  individuals 
with  alcohol  withdrawal  seizure  his- 
tory (Sullivan  et  al.  1996).  Decreases 
in  the  amounts  of  N-acetylaspartate  in 
the  frontal  lobe,  a  measure  of  neuron 
levels,  also  illustrate  frontal  lobe 
degeneration  in  alcoholics  (Jagan- 
nathan  et  al.  1996).  Alcoholics  with 
more  severe  brain  disorders,  such  as 
Wernicke's  and/or  Korsakoff's  syn- 
drome, show  more  significant  reduc- 
tion in  white  matter  and  more 
extensive  brain  region  degeneration, 
which  is  consistent  with  the  greater 
alcohol  consumption  associated  with 
more  severely  damaged  individuals. 

Studies  have  found  that  in  addition 
to  the  global  shrinkage  of  brain 
regions,  certain  key  neuronal  nuclei 
that  have  broad-ranging  effects  on 
brain  activity  are  selectively  lost  with 


chronic  alcohol  abuse.  Perhaps  the 
most  extensively  studied  are  the 
cholinergic  basal  forebrain  nuclei, 
which  are  also  lost  in  Alzheimer's  dis- 
ease. Animal  studies  and  some  human 
studies  have  suggested  that  this  region 
is  particularly  damaged  in  alcoholic 
subjects.  Arendt  (1993)  found  a  sig- 
nificant loss  of  neurons  in  this  region 
in  alcoholic  Korsakoff's  psychosis 
patients.  Additional  brain  nuclei  that 
appear  to  be  particularly  sensitive  are 
the  locus  coeruleus  and  raphe  nuclei. 
These  two  nuclei  contain  many  of  the 
noradrenergic  and  serotonergic  neurons 
within  the  brain,  respectively.  Although 
these  nuclei  are  small  in  size,  they  are 
particularly  important  because  their 
neuronal  processes  project  throughout 
the  brain  and  modulate  global  aspects 
of  brain  activity.  Chemical  studies 
have  shown  abnormally  low  levels  of 
serotonergic  metabolites  in  the  cerebro- 
spinal fluid  of  alcoholics  with  Wernicke - 
Korsakoff  syndrome,  and  more  recent 
morphological  studies  have  found  a 
significant  reduction  (e.g.,  50  per- 
cent) in  the  number  of  serotonergic 
neurons  from  the  raphe  nuclei  of 
severe  alcoholic  cases  studied  com- 
pared with  control  subjects  (McEntee 
and  Mair  1990;  Halliday  et  al.  1995; 
Baker  et  al.  1996;  Higley  and  Bennett 
1999).  Thus  the  serotonergic  system 
appears  to  be  disrupted  in  alcoholics, 
especially  in  severe  alcoholics.  Several 
investigators  have  also  reported  signif- 
icant noradrenergic  cell  loss  in  the 
locus  coeruleus  (Arendt  et  al.  1995; 
Arango  et  al.  1996;  Lu  et  al.  1997), 
although  not  all  have  found  this  loss 
(Harper  and  Kril  1993).  Certain  neurons 
that  contain  the  peptide  vasopressin 


191 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


may  be  sensitive  to  chronic  ethanol- 
induced  neurotoxicity  in  both  rats  and 
humans  (Harding  et  al.  1996;  Madeira 
et  al.  1997).  Damage  to  hypothalamic 
vasopressin  and  other  peptide-con- 
taining  neurons  could  disrupt  a  vari- 
ety of  hormone  functions  as  well  as 
daily  rhythms  that  are  important  for 
healthy  living.  Additional  studies  are 
needed  to  determine  which  specific 
cell  groups  within  the  brain  might  be 
particularly  damaged.  The  findings  of 
specific  neuronal  loss  in  small  but 
functionally  significant  brain  areas 
could  result  in  global  changes  in 
attention,  mood,  and  personality  that 
are  difficult  to  quantify  but  have  a 
great  impact  on  brain  function  and 
overall  behavior. 

Long-term  ethanol  intoxication  is 
not  necessary  to  cause  brain  damage. 
Studies  have  shown  that  as  little  as  a 
few  days  of  intoxication  can  lead  to 
neuronal  loss  in  several  brain  areas, 
including  dentate  gyrus;  entorhinal, 
piriform,  insular,  orbital,  and  perirhi- 
nal cortices;  and  the  olfactory  bulb 
(Collins  et  al.  1996).  These  structures 
are  involved  in  frontal  cortical  neu- 
ronal circuits,  including  the  limbic 
and  association  cortex.  These  findings 
are  consistent  with  human  studies 
reporting  damage  to  entorhinal  cortex 
(Ibanez  et  al.  1995)  and  significant 
hippocampal  shrinkage  in  alcoholics 
(Harding  et  al.  1997).  Hippocampal 
damage  during  chronic  ethanol  treat- 
ment has  been  correlated  with  deficits 
in  spatial  learning  and  memory 
(Franke  et  al.  1997).  Thus,  cortical 
and  hippocampal  damage  also  occurs 
with  chronic  ethanol  treatment,  and 
relatively  short  durations  of  alcohol 


abuse  may  cause  some  form  of  damage. 
Additional  studies  are  needed  to  under- 
stand the  molecular  mechanisms  in- 
volved in  selective  neuronal  death  and 
the  factors  that  regulate  brain  regional 
sensitivity  to  ethanol  neurotoxicity. 

Some  exciting  studies  have  begun  to 
address  the  effects  of  gender  on  brain 
damage.  Interestingly,  alcoholic  women 
appear  to  have  an  increased  sensitivity 
for  brain  damage,  when  compared 
with  alcoholic  men  (Hommer  et  al. 
1996).  This  appears  to  be  true  for  liver 
disease  as  well.  Although  there  are 
more  men  diagnosed  as  alcoholic,  the 
number  of  alcoholic  women  is  increas- 
ing. The  increased  susceptibility  of 
women  to  alcoholic  pathology  is  an 
area  that  needs  further  research. 

Alcoholics  who  do  not  have  Kor- 
sakoffs  syndrome  problems  show 
decreased  neuropsychological  perfor- 
mance compared  with  peer  nonalco- 
holics  on  tests  of  learning,  memory, 
abstracting,  problem  solving,  visu- 
ospatial  and  perceptual  motor  func- 
tioning, and  information  processing 
(Parsons  1993).  Alcoholics  are  less 
accurate  and  take  considerably  longer 
to  complete  tasks.  Alcoholics  are  dif- 
ferentially vulnerable  to  these  deficits, 
and  many  of  the  deficits  appear  to 
recover  to  age -appropriate  levels  of  per- 
formance over  a  4-  to  5 -year  period  of 
abstinence  (Parsons  1993).  Although 
global  cerebral  atrophy  returns  to  nor- 
mal levels  with  extended  abstinence, 
not  all  cognitive  functions  return.  Some 
abstinent  alcoholics  appear  to  have 
permanent  cognitive  impairments, 
particularly  in  memory  and  visual-spatial- 
motor  skills  (Di  Sclafani  et  al.  1995). 
Other  studies  support  a  loss  of  logical 


192 


Neurotoxicity  of  Alcohol 


memory  and  paired  association  learn- 
ing tasks  in  alcoholics  that  may  be 
long-lasting  (Eckardt  et  al.  1996). 

Cognitive  deficits  are  not  the  only 
factors  that  suggest  long-term  changes 
in  brain  function  following  chronic 
ethanol  treatment.  Electrophysiological 
studies  using  brain  electroencephalo- 
grams and  event-related  potentials 
have  suggested  that  alcoholics  have 
difficulty  differentiating  relevant  and 
irrelevant,  easy  and  difficult,  and 
familiar  and  unfamiliar  stimuli  (Porjesz 
and  Begleiter  1993).  These  deficits 
appear  to  be  consistent  for  alcoholics 
and  may  be  related  to  frontal  cortical 
function.  Both  clinical  and  experimen- 
tal studies  support  a  role  of  frontal 
cortical  involvement  in  neuropsycho- 
logical dysfunction  in  alcoholics,  par- 
ticularly those  with  Korsakoff's 
syndrome  ( Oscar-  Berman  and  Hutner 
1993).  Areas  affected  include  emotional 
abilities,  disinhibition,  perseverative 
responding,  reduced  problem- solving 
abilities,  and  poor  attention.  Prefrontal 
damage  typically  is  associated  with 
changes  in  personality  and  elusive 
cognitive  abnormalities.  In  the  last  few 
years  studies  have  emphasized  the  role 
of  the  prefrontal  cortex  in  executive 
cognitive  function  (ECF)  (Giancola 
and  Moss  1998).  Executive  cognitive 
function  is  the  ability  to  use  higher 
mental  functions  such  as  attention,  plan- 
ning, organization,  sequencing,  abstract 
reasoning,  and  the  ability  to  use  external 
and  internal  feedback  to  adaptively  mod- 
ulate future  behavior  (Foster  et  al. 
1994).  ECF  is  dysfunctional  in  alco- 
holics and  in  individuals  with  other 
diseases  showing  prefrontal  damage 
(Boiler  et  al.  1995),  and  disruption  of 


abilities  has  been  implicated  in  the 
underlying  aggression  associated  with 
substance  abuse  (Hoaken  et  al.  1998). 
Although  these  types  of  changes  in 
brain  function  are  more  difficult  to 
assess,  they  are  consistent  with  the 
morphological  changes  found  in 
frontal  cortex  and  with  the  findings  of 
studies  on  damage  to  association  cortex 
in  animals  (Hunt  and  Nixon  1993; 
Giancola  and  Moss  1998). 

EXCITOTOXICITY 

The  mechanisms  of  ethanol  neurotox- 
icity have  only  recently  begun  to  be 
elucidated.  There  are  several  reports  that 
N-methyl-D-aspartate  (NMDA)- 
glutamate  receptors  may  be  involved 
in  tolerance  to  and  dependence  on 
ethanol  as  well  as  ethanol-induced 
brain  damage.  When  MK-801  (dizo- 
cilpine),  an  antagonist  to  NMDA- 
glutamate  receptors,  was  coadminis- 
tered with  ethanol,  the  tolerance  to 
ethanol  was  eliminated  (Khanna  et  al. 
1992;  Szabo  et  al.  1994).  Khanna  and 
colleagues  (1993)  found  that  inhibition 
of  nitric  oxide  also  inhibited  the  devel- 
opment of  tolerance  to  ethanol,  and 
NMDA  receptors  are  closely  coupled 
to  nitric  oxide  formation  (Chandler  et 
al.  1994).  Thus,  NMDA  receptors 
appear  to  be  involved  in  the  develop- 
ment of  tolerance  to  ethanol. 

Hyperexcitability  of  the  central 
nervous  system  is  a  key  component  of 
ethanol  withdrawal,  and  a  supersensi- 
tive NMDA-glutamate  response 
appears  to  be  involved,  although  a 
reduction  in  gamma-aminobutyric 
acid  (GABA)-mediated  inhibition 
may  also  contribute  (Crews  et  al. 


193 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


1996).  One  of  the  earliest  findings 
suggesting  glutamate  involvement  was 
that  [3H]  glutamate  binding  is  increased 
in  human  hippocampus  of  alcoholics 
(Michaelis  et  al.  1990).  Although  the 
subtype  of  glutamate  receptor  involved 
is  not  clear,  this  finding  is  consistent 
with  increased  glutamate  receptor 
density  and  sensitivity.  It  has.  been  dis- 
covered that  NMD  A- glutamate  recep- 
tors have  a  unique  property,  in  that 
excessive  stimulation  of  these  recep- 
tors triggers  a  process  in  neurons  that 
leads  to  neuronal  death.  This  appears 
to  play  a  key  role  in  neurodegenera- 
tive diseases  in  general  as  well  as  in 
stroke,  brain  trauma,  and  other  types 
of  brain  damage  (Crews  et  al.  1996). 
Basic  studies  on  alcohol  have  con- 
tributed significantly  to  the  under- 
standing of  this  process. 

Several  studies  in  isolated  neuronal 
cells  have  indicated  that  a  few  days  of 
chronic  ethanol  treatment  leads  to  super- 
sensitive NMDA-stimulated  calcium 
flux  (Iorio  et  al.  1992;  Ahern  et  al. 
1994),  as  well  as  NMDA-stimulated 
excitotoxicity  (Crews  and  Chandler 
1993;  Crews  et  al.  1993;  Iorio  et  al. 
1993)  and  NMDA-stimulated  nitric 
oxide  formation  (Chandler  et  al. 
1995).  Nitric  oxide  has  been  impli- 
cated in  neuronal  toxicity  because  of 
the  formation  of  highly  oxidative 
metabolites  (Crews  and  Chandler 
1993).  Although  NMD  A  supersensi- 
tivity was  found  in  all  of  these 
responses,  changes  in[3H]MK-801 
binding  or  the  amounts  of  NMDAR1 
or  NR2A  or  NR2B  immunoreactivity 
were  not  found  in  all  cases,  suggest- 
ing that  posttranslational  changes  in 
the  NMD  A  receptor  structure,  not 


density  or  subunit  composition,  may 
be  responsible  for  the  supersensitivity. 
Administration  of  ethanol  has  been 
shown  to  enhance  tyrosine  phospho- 
rylation of  the  NMDA  receptor,  and 
this  has  been  associated  with  acute 
tolerance  to  ethanol's  inhibition  of 
NMDA-mediated  excitatory  postsy- 
naptic potentials  (Miyakawa  et  al. 
1997).  Mice  lacking  a  Fyn  nonreceptor 
tyrosine  kinase  do  not  show  acute  tol- 
erance and  are  hypersensitive  to  ethanol 
(Miyakawa  et  al.  1997).  The  Fyn 
kinase  appears  to  be  involved  in  both 
NMDA  and  GABAA  function  and 
thus  could  play  a  role  in  ethanol  toler- 
ance and  dependence  (Miyakawa  et  al. 
1997).  Although  the  mechanisms  are 
not  totally  resolved,  it  is  clear  that 
chronic  ethanol  can  induce  NMDA 
supersensitivity.  Supersensitive 
NMDA  responses  likely  contribute  to 
the  hyperexcitability  and  seizures  asso- 
ciated with  ethanol  withdrawal,  as  well 
as  causing  neurotoxicity. 

Hoffman's  laboratory  has  reported 
increases  in  the  density  of  NMDA 
receptors  in  C57BL  mice  treated 
chronically  with  a  7  percent  ethanol 
liquid  diet.  Seven  days  of  chronic 
ethanol  increased  [3H]MK-801  bind- 
ing in  hippocampal  membranes  by 
approximately  16  percent  (Grant  et  al. 
1990).  These  animals  were  dependent 
upon  ethanol,  as  indicated  by  with- 
drawal seizures.  An  autoradiographic 
study  also  reported  increased 
[3H]MK-801  binding  in  cortex,  hip- 
pocampus, and  striatum  (Gulya  et  al. 
1991),  which  returned  to  control  val- 
ues in  approximately  24  hours,  a  time 
course  similar  to  the  return  of  seizure 
scores  to  control  levels.  Extensions  of 


194 


Neurotoxicity  of  Alcohol 


these  experiments  with  membrane 
binding  found  only  changes  in  hippo- 
campus, not  cerebral  cortex  (Snell  et 
al.  1993).  These  studies  found  that 
[3H]MK-801-  and  NMDA-specific 
[3H]glutamate  binding  slightly 
increased  in  hippocampus  during 
chronic  ethanol  treatment,  but  there 
were  no  changes  in  [3H]glycine  or 
[3H]CGS19755,  a  competitive  NMDA 
antagonist,  in  hippocampus.  No 
changes  in  any  ligand  binding  were 
found  in  cerebral  cortex  (Snell  et  al. 
1993).  Changes  in  some  NMDA  lig- 
and binding  sites  may  be  involved  in 
subunit  composition  changes  but  not 
in  an  increased  density  of  channels. 

Trevisan  and  colleagues  (1994) 
found  that  12  weeks  of  ethanol  liquid 
diet  to  rats  increased  the  levels  of 
NMDAR1  immunore activity  in  the 
hippocampus,  but  not  in  cortex,  stria- 
tum, or  nucleus  accumbens.  Other 
studies  of  levels  of  NMDAR1  mRNA 
have  indicated  that  chronic  ethanol 
does  not  change  NMDAR1  mRNA 
but  increases  NMDAR2A  and 
NMDAR2B  mRNA  levels  in  hip- 
pocampus and  cortex  (Follesa  and 
Ticku  1995).  Binding  changes  were 
not  reported  in  this  study.  Since  MK- 
801  apparently  requires  both  an 
NMDAR1  subunit  and  an  NMDAR2 
subunit  for  binding,  an  increase  in 
binding  could  be  due  to  changes  in 
channel  subunits  without  necessarily 
an  increase  in  the  density  of  channels. 
Other  studies  have  not  found 
increases  in  MK-801  binding  follow- 
ing chronic  ethanol  treatment  of  mice 
(Carter  et  al.  1995)  or  rats  (Rudolph 
and  Crews  1996).  These  differences 
could  be  due  to  different  ethanol 


treatment  protocols  or  the  responses 
of  different  strains  of  animals.  Long-term 
treatment  of  rats  with  ethanol  (12 
weeks)  was  found  to  be  required  to 
increase  NMDAR1  immunoreactivity 
in  the  ventral  tegmental  area,  whereas 
1  and  6  weeks  of  chronic  5  percent 
ethanol  liquid  diet  were  not  sufficient 
(Ortiz  et  al.  1995).  Although  the 
exact  molecular  processes  require 
additional  experimentation,  a  number 
of  studies  support  the  hypothesis  that 
chronic  ethanol  results  in  supersensitive 
NMDA  receptors  and  that  this  could 
be  a  significant  factor  in  ethanol- 
induced  brain  damage. 

The  mechanism  of  the  neurode- 
generation  in  alcoholic  Wernicke's 
syndrome  also  appears  to  involve  excito- 
toxicity  from  glutamate  in  the  neural 
destructive  process  similar  to  the  less 
severe  alcoholic  brain  damage  (Langlais 
and  Zhang  1993).  In  animal  studies, 
extracellular  concentrations  of  gluta- 
mate in  brain  increase  severalfold  dur- 
ing seizures  in  thiamine-deficient 
animals  (Langlais  and  Zhang  1993). 
Furthermore,  MK-801,  an  NMDA 
antagonist,  reduces  experimental  neuro- 
biological  symptoms  and  severity  of 
neural  lesioning  in  a  thiamine-deficient 
rat  model  (Langlais  and  Mair  1990). 
It  is  not  known  whether  coadministra- 
tion of  ethanol  and  thiamine  deficiency 
would  enhance  the  degree  of  neuro- 
degeneration  seen  in  experimental 
Wernicke's  encephalopathy  (i.e.,  thi- 
amine deficiency).  A  complicating  fac- 
tor of  Wernicke's  encephalopathy  is 
Korsakoff's  amnestic  syndrome  (Victor 
et  al.  1989),  in  which  there  is  a  major 
loss  of  memory.  The  memory  dysfunc- 
tion correlates  best  with  the  presence 


195 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  lesions  in  the  thalamus  (Victor  et 
al.  1989).  It  must  be  recognized  that 
KorsakofPs  amnestic  syndrome  can 
occur  in  the  absence  of  ethanol, 
although  it  is  extremely  rare.  How- 
ever, in  susceptible  individuals, 
chronic  ethanol  clearly  facilitates  the 
course  of  this  disease.  In  any  case,  there 
is  strong  evidence  that  ethanol  with- 
drawal hyperexcitability  is  related  at 
least  in  part  to  NMDA  supersensitivity 
and  that  this  supersensitivity  could 
underlie  ethanol-induced  brain  dam- 
age. In  summary,  chronic  ethanol 
appears  to  cause  NMDA  receptor 
supersensitivity  in  a  variety  of  systems. 
NMDA  supersensitivity  is  likely 
involved  in  ethanol  tolerance,  depen- 
dence, withdrawal,  and  neurotoxicity. 

OXIDATIVE  STRESS 

Another  likely  mechanism  of  ethanol- 
induced  brain  damage  involves  increased 
oxidative  stress  of  neurons.  Cells  use 
oxygen  for  energy  metabolism,  but  they 
normally  have  protective  mechanisms 
against  oxidative  damage.  Studies  exam- 
ining the  effects  of  both  acute  and  chronic 
ethanol  administration  upon  cellular 
oxidation  have  primarily  focused  on  either 
ethanol's  effects  on  intracellular  antiox- 
idant mechanisms  such  as  a-tocopherol, 
ascorbate,  glutathione,  catalase,  and 
superoxide  dismutase  activity  (Ledig  et 
al.  1981;  Nordmann  1987;  Rouach  et 
al.  1987;  Montoliu  et  al.  1994)  or 
potential  sources  of  oxidative  radicals 
such  as  CYP2E1,  an  ethanol-inducible 
form  of  cytochrome  P-450  and  a 
potent  generator  of  oxidative  radicals 
(Montoliu  et  al.  1994,  1995).  Chronic 
ethanol-induced  increases  in  CYP2E1 


and  other  oxidases  have  been  related  to 
increased  lipid  peroxidation  and  reactive 
oxygen  radicals  in  brain  (Montoliu  et 
al.  1994).  However,  levels  of  antioxi- 
dant enzymes  such  as  catalase  and 
superoxide  dismutase  appear  to  increase 
as  a  compensatory  mechanism  to 
ethanol-induced  oxidant  enzyme  levels 
(Montoliu  et  al.  1994).  The  brain  is 
particularly  susceptible  to  lipid  peroxi- 
dation, because  it  consumes  a  large 
amount  of  oxygen  and  is  rich  in  poly- 
unsaturated fatty  acids,  which  are  espe- 
cially prone  to  reactive  oxygen  injury.  It 
has  been  demonstrated  that  a  single  dose 
of  ethanol  results  in  both  the  elevation  of 
lipid  hydroperoxide  levels  and  decreases 
in  glutathione  levels  in  rat  brain  homo- 
genates  (Uysal  et  al.  1986,  1989; 
Nordmann  et  al.  1990,  1992).  How- 
ever, it  is  not  clear  how  this  increased 
oxidation  translates  to  increased  brain 
damage,  if  it  does  at  all.  Although  most 
studies  have  focused  on  the  whole  brain, 
a  recent  study  of  ethanol-induced 
depression  of  glutathione  and  gluta- 
mine  synthetase  levels,  two  indices  in 
increased  oxidative  radical  formation, 
found  changes  only  in  striatum,  but 
not  in  cerebral  cortex  or  cerebellum 
(Bondy  and  Guo  1995). 

Oxidative  stress  has  been  implicated 
in  a  variety  of  conditions,  particularly 
aging,  Alzheimer's  disease,  parkinson- 
ism, stroke,  and  other  neurodegenera- 
tive diseases.  Much  more  research  is 
needed  to  completely  understand  how 
oxidation  damages  neurons  and  how 
other  brain  cells  respond  to  increased 
oxidative  stress.  Ethanol-induced 
neurodegeneration  may  be  related  to 
an  induction  of  oxidative  enzymes, 
and  alcohol  research  provides  an 


196 


Neurotoxicity  of  Alcohol 


opportunity  to  clearly  address  this 
aspect  of  neurodegeneration. 

NEUROTROPHIC  FACTORS 

Growth  factors  are  specific  protein  ele- 
ments of  brain  that  stimulate  growth  and 
extensions  of  neurons  and  are  essential 
for  the  survival  of  certain  neurons.  Fur- 
thermore, growth  factors  are  known  to 
increase  neuronal  antioxidant  and  exci- 
totoxic  protective  mechanisms.  Ethanol 
has  been  found  to  alter  brain  levels  of 
growth  factors  (Arendt  et  al.  1995; 
MacLennan  et  al.  1995;  Baek  et  al. 
1996;  Nakano  et  al.  1996).  Chronic 
ethanol  does  not  lead  to  a  loss  of  all 
growth  factor  activity,  but  appears  to  be 
somewhat  selective.  Chronic  ethanol 
reduces  brain-derived  neurotrophic  factor 
but  does  not  alter  nerve  growth  factor, 
neurotrophin  3  factor,  or  fibroblast 
growth  factor  levels  (MacLennan  et  al. 
1995;  Baek  et  al.  1996).  Receptors  for 
the  growth  factors  remain  intact  after 
chronic  ethanol  abuse  (Arendt  et  al. 
1995;  MacLennan  et  al.  1995)  and 
present  the  promising  possibility  that 
growth  factors  may  be  used  to  treat 
ethanol-induced  brain  damage  as  well  as 
other  neurodegenerative  conditions. 
Studies  of  growth  factor  action  and  their 
role  in  ethanol-induced  brain  damage 
represent  an  exciting  area  of  discovery 
with  the  tremendous  potential  to  pro- 
vide a  variety  of  new  approaches  to 
treating  neurodegeneration. 

APOPTOSIS 

Apoptosis  is  a  physiological  form  of 
cell  death  with  characteristic  morpho- 
logical and  biochemical  features  that 


include  nuclear  pyknosis,  DNA  frag- 
mentation, and  dependence  on  new 
protein  synthesis.  A  cascade  leading 
to  apoptotic  cell  death  includes 
induction  of  p53  protein;  recruitment 
of  other  transcription  factor  proteins, 
such  as  bax,  bcl,  and  bad;  and  activa- 
tion of  a  series  of  caspase  proteases 
and  tyrosine  kinases.  Few  of  these 
markers  of  apoptosis  have  been  stud- 
ied in  intact  brain.  Although  some 
reports  have  suggested  a  role  in  exci- 
totoxic  cell  death,  most  studies  have 
found  excitotoxicity  to  be  primarily 
necrotic  (Cheung  et  al.  1998;  Martin 
et  al.  1998).  A  few  studies  have 
reported  that  adrenalectomy-induced 
loss  of  dentate  granule  cells 
(Schreiber  et  al.  1994)  or  kainate- 
induced  cell  death  (Sakhi  et  al.  1994) 
is  associated  with  induction  of  p53, 
suggesting  that  apoptosis  may  occur 
in  intact  adult  brain.  However,  few 
other  studies  have  extended  these 
findings,  and  most  studies  have  been 
done  in  cell  culture  with  neonatal 
neurons.  Recent  studies  have  sug- 
gested that  apoptosis  is  common  dur- 
ing development,  but  that  necrosis 
predominates  in  adult  brain  (Portera- 
Cailliau  et  al.  1997). 

Few  studies  of  ethanol  and  apopto- 
sis have  been  done.  Ethanol  has  been 
found  to  inhibit  NMDA  inhibition  of 
apoptosis  in  cerebellar  granule  cell 
cultures  (Hoffman  et  al.  1989).  In 
astroglial  cultures,  ethanol-induced 
death  was  found  to  be  due  to 
necrotic,  but  not  apoptotic,  mecha- 
nisms (Holownia  et  al.  1997).  Thus, 
few  studies  support  a  major  role  of 
apoptotic  death  in  ethanol-induced 
brain  damage  in  adults,  although 


197 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


important  actions  may  occur  as  part  of 
ethanol's  teratogenic  actions. 

CELL  ADHESION 
MOLECULES 

Cell  adhesion  molecules  clearly  play  a 
role  in  neuronal  structure  and  func- 
tion. Chronic  treatment  with  NMDA 
antagonists  results  in  neuronal  dam- 
age and  increased  NMDA  receptor 
clustering  (Rao  and  Craig  1997).  Since 
ethanol  is  an  NMDA  antagonist,  it  is 
possible  that  this  results  in  structural 
changes  in  cell  adhesion  proteins  or 
cytoskeletal  elements.  Charness's  lab- 
oratory found  that  ethanol  potently 
and  completely  inhibits  LI -mediated 
cell  adhesion  in  transfected  cells,  but 
has  no  effect  on  other  adhesion  mole- 
cules such  as  NCAM  140  (Ramanathan 
et  al.  1996).  Studies  in  chick  develop- 
ment have  suggested  that  ethanol's 
teratogenic  effects  are  due  to  a  disrup- 
tion of  cell  adhesion  molecule  synthesis 
and  function  (Kentroti  et  al.  1995). 
This  is  a  new  and  exciting  area,  but 
there  is  not  enough  data  relating  adhe- 
sion proteins  to  ethanol's  neurotoxic 
actions  to  prompt  a  significant  invest- 
ment in  this  field  at  this  time. 

BRAIN  DAMAGE 
AND  ALCOHOLISM: 
IMPLICATIONS  FOR 
THE  PROGRESSION  TO 
ALCOHOL  DEPENDENCE 

Alcoholism  is  a  progressive  disease  that 
starts  with  experimentation  and  pro- 
gresses to  addiction,  usually  over  the 
course  of  several  years.  Addiction 
involves  the  loss  of  control  over  the 


ability  to  abstain  from  the  drug  and  per- 
severative  preoccupation  with  obtaining 
and  using  the  drug.  Although  earlier 
studies  focused  on  alcohol-induced 
changes  in  cognition,  more  recent 
studies  have  focused  on  the  frontal 
cortex  and  the  role  of  this  brain 
region  in  behavior.  Researchers  have 
begun  to  investigate  ECF  as  an 
important  function  of  the  prefrontal 
cortex.  A  variety  of  evidence  has 
focused  attention  on  the  prefrontal 
cortex  as  an  area  of  brain  that  is  partic- 
ularly sensitive  to  alcohol-induced 
brain  damage.  At  the  same  time  scien- 
tists have  developed  ways  to  investi- 
gate the  role  of  ECF  in  behavior. 
ECF/prefrontal  cortical  characteristics 
are  associated  with  decreased  regulation 
of  human  social  behavior,  including 
disinhibition  syndrome  characterized 
by  impulsivity,  socially  inappropriate 
behavior,  and  aggression  (Giancola 
and  Moss  1998).  Neuroimaging  stud- 
ies have  indicated  that  hypofunction 
of  the  frontal  lobes  is  associated  with 
violence  (Raine  et  al.  1994).  Experi- 
mental subjects  with  poor  prefrontal 
functioning  appear  unable  to  inhibit 
impulsive  behavior  (Lau  and  Pihl 
1996),  particularly  violence  (Lau  et  al. 
1995).  Taken  together,  these  studies 
suggest  that  some  of  the  greatest 
sociopathic  problems  of  alcoholism, 
such  as  violence  and  loss  of  control 
over  the  drug,  may  be  directly  related  to 
the  neurotoxic  effects  of  ethanol  on  pre- 
frontal cortical  function.  Animal  stud- 
ies have  shown  that  chronic  exposure 
to  ethanol  and  repeated  withdrawal 
episodes  increase  self- administration 
of  ethanol  if  particularly  high  blood 
levels  are  reached  (Schulteis  et  al. 


198 


Neurotoxicity  of  Alcohol 


1996).  More  studies  are  needed  to 
directly  determine  the  relationship  of 
prefrontal  cortical  function  to  alcohol- 
induced  brain  damage  and  addiction. 

SUMMARY 

Alcohol  can  be  neurotoxic.  Recent  stud- 
ies have  indicated  that  the  prefrontal 
cortex  is  particularly  sensitive  to  the 
neurotoxic  actions  of  alcohol,  as  are 
specific  groups  of  neurons  that  project 
throughout  the  brain,  including  the 
frontal  cortex,  biogenic  amine,  and 
peptidergic  neurons.  Areas  that  need 
increased  emphasis  include  the  specific 
types  of  neurons  that  are  most  sensi- 
tive to  ethanol  neurotoxicity  and  the 
mechanisms  of  neurotoxicity,  particu- 
larly NMDA  excitotoxicity,  oxidative 
mechanisms  of  neuronal  stress,  and 
protein  induction  during  chronic 
ethanol  consumption. 

GAPS  IN  KNOWLEDGE  AND 
RECOMMENDATIONS  FOR 
FUTURE  DIRECTIONS 

Neurotoxicity 

Neurons.  Human  data  supports  loss  of 
neurons,  particularly  in  frontal  cortex. 
Animal  data  indicates  specific  limbic 
cortical  neuronal  damage  after  4  days 
of  intoxication.  The  role  of  ethanol 
withdrawal  and  extended  intoxication, 
as  well  as  other  factors  in  alcoholic 
neurotoxicity,  is  a  significant  gap  in 
our  knowledge. 

White  Matter  Loss.  Human  studies 
indicate  loss  of  white  matter  (e.g., 
myelin).  This  loss  could  be  secondary 
to  the  loss  of  neurons  or  due  to  specific 


toxicity  to  various  types  of  glia.  This  is 
a  crucial  question  that  needs  to  be 
addressed  to  clearly  understand  the  pri- 
mary site  of  alcoholic  insults  to  the  brain. 

U.S.  Brain  Bank.  Most  of  the 
human  postmortem  data  comes  from 
one  group  in  Australia.  Although  this 
is  an  excellent  group,  there  is  a  critical 
need  to  establish  resources  within  the 
United  States  to  provide  opportunities 
for  U.S.  investigators.  It  is  recom- 
mended that  a  consortium — through 
the  Research  Society  on  Alcoholism, 
existing  brain  bank  facilities  sponsored 
for  other  diseases,  or  a  specific  U.S. 
institution — be  established  to  begin 
forming  a  human  alcoholic  brain  bank 
that  U.S.  investigators  will  be  able  to 
use  for  experimentation. 

Gender.  Although  women  represent 
approximately  25  percent  of  alcoholics 
(Grant  et  al.  1991),  they  may  suffer 
greater  pathology.  Data  regarding  the 
role  of  gender  could  provide  important 
fundamental  insights  into  the  mecha- 
nisms of  brain  damage  as  well  as 
important  new  information.  There  is  a 
significant  gap  in  our  knowledge  in 
this  area  that  needs  to  be  addressed. 

Mechanisms  of  Brain  Damage 

Mechanisms  of  NMDA  Excitotoxicity. 
Both  in  vitro  and  in  vivo  data  suggest 
that  NMDA  excitotoxicity  contributes 
to  neurodegeneration  in  a  variety  of 
pathologies,  including  alcoholic  brain 
damage,  Alzheimer's  disease,  stroke,  and 
Parkinson's  disease.  A  significant  gap 
in  knowledge  exists  with  regard  to  the 
processes  between  the  acute  excessive 
stimulation  of  neurons  by  glutamate 
and  the  processes  that  lead  to  delayed 
neuronal  death.  Understanding  these 


199 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


events  will  greatly  enhance  our  under- 
standing of  the  mechanisms  of  neu- 
ronal death. 

Selective  Neuronal  Death.  Studies 
have  indicated  that  frontal  cortex  neu- 
rons as  well  as  specific  neuronal  popu- 
lations, such  as  cholinergic  forebrain 
nuclei,  raphe  nuclei,  locus  coeruleus, 
and  vasopressin  neurons,  may  be  lost 
during  chronic  ethanol  consumption. 
Why  these  neuronal  nuclei  are  specifi- 
cally sensitive  is  an  important  question 
that  needs  to  be  answered. 

Role  of  Oxidation.  A  variety  of  stud- 
ies have  suggested  that  oxidative  stress 
is  increased  in  brain  by  alcohol.  It  is 
often  hypothesized  that  this  contributes 
to  brain  damage;  however,  there  is  a 
gap  in  our  knowledge,  with  littie  data 
for  or  against  this  hypothesis. 

Consequences 
of  Brain  Damage 

Cortical  Function  and  Alcoholism.  A 
variety  of  studies  have  indicated  loss 
of  specific  memory  tasks  and  other 
cognitive  abilities  with  alcoholic  brain 
damage.  An  important  future  direction 
will  be  to  understand  the  role  alcoholic 
brain  damage  plays  in  the  progression 
to  alcoholism,  recovery  from  alcoholism, 
and  other  behaviors  associated  with 
alcoholism  (e.g.,  violence,  relapse,  and 
trauma  injury).  There  is  a  significant 
gap  in  our  knowledge  in  this  area. 
Understanding  the  relationship  of 
neuropathology  to  behavioral  pathology 
is  essential  and  fundamental  to  improv- 
ing prevention  and  treatment. 

Adolescent  Factors.  Researchers 
need  to  investigate  the  effects  of  alcohol 
on  the  adolescent  brain.  Recent  stud- 
ies (e.g.,  Grant  and  Dawson  1998) 


have  suggested  that  teenagers  who  start 
drinking  earlier  are  more  likely  to 
develop  alcoholism.  Adolescents 
respond  differentiy  to  alcohol,  but  little 
or  no  biological  data  address  this  issue. 
Relationship  to  Other  Pathology. 
Alcoholics  are  at  greater  risk  of 
trauma,  seizures,  and  stroke-induced 
brain  damage.  The  interaction  of 
these  pathologies  and  the  behavioral 
and  physiological  factors  that  con- 
tribute to  these  pathologies  are  poorly 
understood. 

ACKNOWLEDGMENT 

The  preparation  of  this  chapter  was 
supported  by  the  National  Institute 
on  Alcohol  Abuse  and  Alcoholism. 


REFERENCES 

Ahern,  K.B.;  Lustig,  H.S.;  and  Greenberg, 
D.A.  Enhancement  of  NMD  A  toxicity  and 
calcium  responses  by  chronic  exposure  of 
cultured  cortical  neurons  to  ethanol. 
NeurosciLett  165:211-214,  1994. 

Arango,  V.;  Underwood,  M.D.;  Pauler, 
D.K.;  Kass,  R.E.;  and  Mann,  J.J. 
Differential  age-related  loss  of  pigmented 
locus  coeruleus  neurons  in  suicides,  alco- 
holics, and  alcoholic  suicides.  Alcohol  Clin 
Exp  Res  20(7):1141-1147 ',  1996. 

Arendt,  T.  The  cholinergic  differentiation 
of  the  cerebral  cortex  induced  by  chronic 
consumption  of  alcohol:  Reversal  by 
cholinergic  drugs  and  transplantation.  In: 
Hunt,  W.A.,  and  Nixon,  S.J.,  eds. 
Alcohol-Induced  Brain  Damage.  National 
Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  431-460. 


200 


Neurotoxicity  of  Alcohol 


Arendt,  T.;  Bruckner,  M.K.;  Magliusi, 
S.;  and  Krell,  T.  Degeneration  of  rat 
cholinergic  basal  forebrain  neurons  and 
reactive  changes  in  nerve  growth  factor 
expression  after  chronic  neurotoxic  injury. 
I.  Degeneration  and  plastic  response  of 
basal  forebrain  neurons.  Neuroscience 
65(3):633-645,  1995. 

Baek,  J.K.;  Heaton,  M.B.;  and  Walker, 
D.W.  Up-regulation  of  high -affinity 
neurotrophin  receptor,  trk  B-like 
protein  on  western  blots  of  rat  cortex 
after  chronic  ethanol.  Brain  Res 
40(1):161-164,  1996. 

Baker,  K.G.;  Halliday,  G.M.;  Kril,  J.J.; 
and  Harper,  C.G.  Chronic  alcoholism  in 
the  absence  of  Wernicke -Korsakoff  syn- 
drome and  cirrhosis  does  not  result  in  the 
loss  of  serotonergic  neurons  from  the 
median  raphe  nucleus.  Metab  Brain  Dis 
ll(3):217-227,  1996. 

Boiler,  F.;  Traykov,  L.;  Dao-Castellana, 
M.H.;  and  Fontaine-Dabernard,  A. 
Cognitive  functioning  in  "diffuse  pathol- 
ogy": Role  of  prefrontal  and  limbic  struc- 
tures. Ann  NT  Acad  Sci  769:23-39,  1995. 

Bondy,  S.C.,  and  Guo,  S.X.  Regional 
selectivity  in  ethanol-induced  pro-oxidant 
events  within  the  brain.  Biochem  Pharmacol 
49(l):69-72,  1995. 

Butterworth,  R.F.  Pathophysiology  of 
alcoholic  brain  damage:  Synergistic  effects 
of  ethanol,  thiamine  deficiency  and  alco- 
holic liver  disease.  Metab  Brain  Dis  10(1): 
1-8,  1995. 

Carter,  LA.;  Belknap,  J.K.;  Crabbe,  J.C.; 
and  Janowsky,  A.  Allosteric  regulation  of 
the  N-methyl-D-aspartate  receptor-linked 
ion  channel  complex  and  effects  of 
ethanol  in  ethanol  withdrawal  seizure - 
prone  and  -resistant  mice.  J  Neurochem 
64:213-219,  1995. 


Chandler,  L.J.;  Guzman,  N.;  Sumners, 
C;  and  Crews,  F.T.  Induction  of  nitric 
oxide  synthase  in  brain  glial  cells:  Possible 
interaction  with  ethanol  in  reactive  neuronal 
injury.  In:  Lancaster,  F.E.,  ed.  Alcohol  and 
Glial  Cells.  National  Institute  on  Alcohol 
Abuse  and  Alcoholism  Research 
Monograph  No.  27.  NIH  Pub.  No. 
94-3742.  Bethesda,  MD:  the  Institute, 
1994.  pp.  195-214. 

Chandler,  L.J.;  Sumners,  C;  and  Crews, 
F.T.  Chronic  ethanol  increases  NMDA- 
stimulated  NO  formation  but  not  MK- 
801  binding  or  NMDAR1  subunits  in 
primary  neuronal  cultures.  Mo  I  Pharmacol 
19:6A-14,  1995. 

Charness,  M.E.  Brain  lesions  in  alcoholics. 
Alcohol  Clin  Exp  Res  17(1):2-11,  1993. 

Cheung,  N.S.;  Pascoe,  C.J.;  Giardina,  S.F.; 
John,  C.A.,  and  Beart,  P.M.  Micromolar 
L-glutamate  induces  extensive  apoptosis 
in  an  apoptotic-necrotic  continuum  of 
insult-dependent,  excitotoxic  injury  in 
cultured  cortical  neurones.  Neuropharm- 
acology 37:1419-1429,  1998. 

Collins,  M.;  Corso,  T.;  and  Neafsey,  E. 
Neuronal  degeneration  in  rat  cerebrocor- 
tical  olfactory  regions  during  subchronic 
"binge"  intoxication  with  ethanol:  Possible 
explanation  for  olfactory  deficits  in  alco- 
holics. Alcohol  Clin  Exp  Res  20(2): 
284-292,  1996. 

Crews,  F.T.,  and  Chandler,  L.J.  Excito- 
toxicity  and  the  neuropathology  of 
ethanol.  In:  Hunt,  W.A.,  and  Nixon,  S.J., 
eds.  Alcohol-Induced  Brain  Damage. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  355-371. 

Crews,  F.T.;  Newsom,  H.;  Gerber,  M.; 
Sumners,  C;  Chandler,  L.J.;  and  Freund, 
G.  Molecular  mechanisms  of  alcohol 


201 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


neurotoxicity.  In:  Ailing,  C;  Diamond  I.; 
Leslie,  S.W.;  Sun,  G.Y.;  and  Wood,  W.G., 
eds.  Alcohol,  Cell  Membranes,  and  Signal 
Transduction  in  Brain.  New  York:  Plenum 
Press,  1993.  pp.  123-138. 

Crews,  F.T.;  Morrow,  A.L.;  Criswell,  H.;  and 
Breese,  G.  Effects  of  ethanol  on  ion  chan- 
nels. Int  Rev  Neurobiol  39 -.283-367,  1996. 

Di  Sclafani,  V.;  Ezekiel,  F.;  Meyerhoff, 
D.J.;  MacKay,  S.;  Dillon,  W.P.;  Weiner, 
M.W.;  and  Fein,  G.  Brain  atrophy  and 
cognitive  function  in  older  abstinent 
alcoholic  men.  Alcohol  Clin  Exp  Res 
19(5):1121-1126,  1995. 

Dufour,  M.C.  The  epidemiology  of  alcohol- 
induced  brain  damage.  In:  Hunt,  W.A.,  and 
Nixon,  S.J.,  eds.  Alcohol-Induced  Brain 
Damage.  National  Institute  on  Alcohol 
Abuse  and  Alcoholism  Research  Monograph 
No.  22.  NIH  Pub.  No.  93-3549.  Rockville, 
MD:  the  Institute,  1993.  pp.  3-14. 

Eckardt,  M.J.;  Rohrbaugh,  J.W.;  Stapleton, 
J.M.;  Davis,  E.Z.;  Martin,  P.R.;  and 
Weingartner,  H.J.  Attention-related  brain 
potential  and  cognition  in  alcoholism- 
associated  organic  brain  disorders.  Biol 
Psychiatry  39(2):143-146,  1996. 

Follesa,  P.,  and  Ticku,  M.  Chronic  ethanol 
treatment  differentially  regulates  NMDA 
receptor  subunit  mRNA  expression  in  rat 
brain.  Brain  Res  Mol  Brain  Res  29:99- 
106, 1995. 

Foster,  J.;  Eskes,  G.;  and  Stuss,  D.  The 
cognitive  neuropsychology  of  attention:  A 
frontal  lobe  perspective.  Cogn  Neuropsychol 
11:133-147,1994. 

Franke,  H;  Kittner,  H.;  Berger,  P.;  Wirkner, 
K.;  and  Schramek,  J.  The  reaction  of 
astrocytes  and  neurons  in  the  hippocam- 
pus of  adult  rats  during  chronic  ethanol 
treatment  and  correlations  to  behavioral 
impairments.  Alcohol  14(5):445^54,  1997. 


Giancola,  P.R.,  and  Moss,  H.B.  Executive 
cognitive  functioning  in  alcohol  use  disor- 
der. Recent  Dev  Alcohol  14:227-251,  1998. 

Grant,  B.F.,  and  Dawson,  DA.  Age  of 
onset  of  drug  use  and  its  association  with 
DSM-rV  drug  abuse  and  dependence: 
Results  from  the  National  Longitudinal 
Alcohol  Epidemiologic  Survey.  J  Subst 
Abuse  10(2):163-173,  1998. 

Grant,  B.F.;  Harford,  T.C.;  Chou,  P.; 
Pickering,  R.;  Dawson,  D.;  Stinson,  F., 
and  Noble,  J.  Epidemiologic  Bulletin  No. 
28:  Prevalence  of  DSM-III-R  alcohol 
abuse  and  dependence:  United  States, 
1988.  Alcohol  Health  Res  World  15(2): 
15-20,1991. 

Grant,  K.A.;  Valverius,  P.;  Hudspith,  M.; 
and  Tabakoff,  B.  Ethanol  withdrawal 
seizures  and  the  NMDA  receptor  com- 
plex. Eur  J  Pharmacol  176:289-296, 1990. 

Gulya,  K.;  Grant,  K.A.;  Valverius,  P.; 
Hoffman,  P.L.;  and  Tabakoff,  B.  Brain 
regional  specificity  and  time-course  of 
changes  in  the  NMDA  receptor- 
ionophore  complex  during  ethanol  with- 
drawal. Brain  Res  547:129-134,  1991. 

Halliday,  G.;  Baker,  K.;  and  Harper,  C. 
Serotonin  and  alcohol-related  brain  dam- 
age. Metab  Brain  Dis  10(l):25-30,  1995. 

Harding,  A.J.;  Halliday,  G.M.;  Ng,  J.L.; 
Harper,  C.G.;  and  Kril,  J.J.  Loss  of  vaso- 
pressin-immunoreactive  neurons  in  alco- 
holics is  dose-related  and  time-dependent. 
J  Neurosci  72(3):699-708,  1996. 

Harding,  A.J.;  Wong,  A.;  Svoboda,  M.; 
Kril,  J.J.;  and  Halliday,  G.M.  Chronic 
alcohol  consumption  does  not  cause 
hippocampal  neuron  loss  in  humans. 
Hippocampus  7(l):78-87,  1997. 

Harper,  C.G.,  and  Kril,  J.J.  Neuropatho- 
logical  changes  in  alcoholics.  In:  Hunt, 
W.A.,  and  Nixon,  S.J.,  eds.  Alcohol-Induced 


202 


Neurotoxicity  of  Alcohol 


Brain  Damage.  National  Institute  on 
Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Pub.  No. 
93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  39-70. 

Harper,  C.G.;  Kril,  J.J.;  and  Daly,  J.  Are 
we  drinking  our  neurons  away?  Br  Med  J 
294:534-536,  1987. 

Higley,  J.D.,  and  Bennett,  A.J.  Central 
nervous  system  serotonin  and  personality 
as  variables  contributing  to  excessive  alco- 
hol consumption  in  non-human  primates. 
Alcohol  Alcohol  34(3):402-418,  1999. 

Hoaken,  P.N.S.;  Giancola,  P.R.;  and  Pihl, 
R.O.  Executive  cognitive  functions  as 
mediators  of  alcohol-related  aggression. 
Alcohol  Alcohol  33(1  ):47-54,  1998. 

Hoffman,  P.L.;  Moses,  F.;  and  Tabakoff, 
B.  Selective  inhibition  by  ethanol  of  glu- 
tamate-stimulated  cyclic  GMP  production 
in  primary  cultures  of  cerebellar  granule  cells. 
Neuropharmacology  28:1239-1243,  1989. 

Holownia,  A.;  Ledig,  M.;  and  Menez,  J.F. 
Ethanol- induced  cell  death  in  cultured  rat 
astroglia.  Neurotoxicol  Teratol  19(2):141- 
146,  1997. 

Hommer,  D.;  Momenan,  R;  Rawlings, 
R;  Ragan,  P.;  Williams,  W.;  Rio,  D.;  and 
Eckardt,  M.  Decreased  corpus  callosum 
size  among  alcoholic  women.  Arch  Neurol 
53(4):359-363,  1996. 

Hunt,  W.A.,  and  Nixon,  S.J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Pub.  No.  93- 
3549.  Rockville,  MD:  the  Institute,  1993. 

Ibanez,  J.;  Herrero,  M.T.;  Insausti,  R; 
Balzunegui,  T.;  Tunon,  T.;  Garcia- 
Bragado,  F.;  and  Gonzalo,  L.M.  Chronic 
alcoholism  decreases  neuronal  nuclear  size 
in  the  human  entorhinal  cortex.  Neurosci 
Lett  183(l-2):71-74,  1995. 


Iorio,  K.R.;  Reinlib,  L.;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Chronic  exposure  of  cere- 
bellar granule  cells  to  ethanol  results  in 
increased  N-methyl-D-aspartate  receptor 
function.  Mol  Pharmacol  41:1142- 
1148,  1992. 

Iorio,  K.R.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Glutamate -induced  neurotoxicity  is 
increased  in  cerebellar  granule  cells  exposed 
chronically  to  ethanol.  Eur  J  Pharmacol 
248:209-212,  1993. 

Jagannathan,  N.R.;  Desai,  M.G.;  and 
Raghunathan,  P.  Brain  metabolite 
changes  in  alcoholism:  An  in  vivo  proton 
magnetic  resonance  spectroscopy.  Magn 
Reson  Imaging  14(5):553-557,  1996. 

Jernigan,  T.L.;  Butters,  N.;  DiTraglia,  G.; 
Schafer,  K.;  Smith,  T.;  Irwin,  M.;  Grant, 
I.;  Schuckit,  M.;  and  Cermak,  L.S.  Reduced 
cerebral  gray  matter  observed  in  alcoholics 
using  magnetic  resonance  imaging.  Alcohol 
Clin  Exp  Res  15(3):418^27,  1991. 

Kentroti,  S.;  Rahman,  H.;  Grove,  J.;  and 
Vernadakis,  A.  Ethanol  neurotoxicity  in 
the  embryonic  chick  brain  in  ovo  and  in 
culture:  Interaction  of  the  neural  cell 
adhesion  molecule  (NCAM).  IntJDev 
Neurosci  13(8):859-870,  1995. 

Khanna,  J.M.;  Weiner,  J.;  Kalant,  H.; 
Chau,  A.;  and  Shah,  G.  Ketamine  blocks 
chronic,  but  not  acute  tolerance  to 
ethanol.  Pharmacol  Biochem  Behav 
42:347-350,  1992. 

Khanna,  J.M.;  Morato,  G.S.;  Shah,  G.; 
Chau,  A.;  and  Kalant,  H.  Inhibition  of 
nitric  oxide  synthesis  impairs  rapid  toler- 
ance to  ethanol.  Brain  Res  Bull  32(1): 
43^7,  1993. 

Langlais,  P.J.,  and  Mair,  R.G.  Protective 
effects  of  the  glutamate  antagonist  MK- 
801  on  pyrithiamine-induced  lesions  and 


203 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


amino  acid  changes  in  rat  brain.  / 
Neurosci  10(5):1664-1674,  1990. 

Langlais,  P.J.,  and  Zhang,  S.X.  Extracellular 
glutamate  is  increased  in  thalamus  during 
thiamine  deficiency-induced  lesions  and  is 
blocked  by  MK-801.  J  Neurochem  61(6): 
2175-2182,  1993. 

Lau,  M.A.,  and  Pihl,  R.O.  Cognitive  per- 
formance, monetary  incentive,  and  aggres- 
sion. Aggressive  Behav  22:150-155,  1996. 

Lau,  M.A.;  Pihl,  R.O.;  and  Peterson,  J.B. 
Provocation,  acute  alcohol  intoxication, 
cognitive  performance,  and  aggression.  / 
Abnorm  Psychol  104(1):150-155,  1995. 

Ledig,  M.;  M'Paria,  J.;  and  Mandel,  P. 
Superoxide  dismutase  activity  in  rat  brain 
during  acute  and  chronic  alcohol  intoxica- 
tion. Neurochem  Res  6(4):385-390,  1981. 

Lu,  W.;  Jaatinen,  P.;  Rintala,  J.;  Sarviharju, 
M.;  Kiianmaa,  K.;  and  Hervonen,  A.  Effects 
of  life -long  ethanol  consumption  on  rat 
locus.  Alcohol  Alcohol  32(4):463-470, 1997. 

MacLennan,  A.J.;  Lee,  N.;  and  Walker, 
D.W.  Chronic  ethanol  administration 
decreases  brain-derived  neurotrophic  factor 
gene  expression  in  the  rat  hippocampus. 
Neurosci  Lett  197(2):105-108,  1995. 

Madeira,  M.D.;  Andrade,  J. P.;  Lieberman, 
A.R;  Sousa,  N.;  Almeida,  O.F.;  and  Paula- 
Barbosa,  M.M.  Chronic  alcohol  consump- 
tion and  withdrawal  do  not  induce  cell 
death  in  the  suprachiasmatic  nucleus,  but 
lead  to  irreversible  depression  of  peptide 
immunoreactivity  and  mRNA  levels.  / 
Neurosci  17(4):1302-1319,  1997. 

Martin,  L.J.;  Al-Abdulla,  N.A.;  Brambrink, 
A.M.;  Kirsch,  J.R.;  Sieber,  F.E.,  and 
Porte ra-Cailliau,  C.  Neurodegeneration  in 
excitotoxicity,  global  cerebral  ischemia, 
and  target  deprivation:  A  perspective  on 
the  contributions  of  apoptosis  and  necro- 
sis. Brain  Res  Bull  46(4):28l-309,  1998. 


Martin,  P.R.;  Adinoff,  B.;  Weingartner, 
H.;  Mukherjee,  A.B.,  and  Edhardt,  M.J. 
Alcoholic  organic  brain  disease:  Nosology 
and  pathophysiologic  mechanisms.  Prog 
Neuropsychopharmacol  Biol  Psychiatry 
10:147-164,  1986. 

McEntee,  W.J.,  and  Mair,  R.G.  The 
Korsakoff  syndrome:  A  neurochemical 
perspective.  Trends  Neurosci  13:340- 
344,  1990. 

Michaelis,  E.K.;  Freed,  W.J.;  Galton,  N.; 
Foye,  J.;  Michaelis,  M.L.;  Phillips,  I.;  and 
Kleinman,  J.E.  Glutamate  receptor 
changes  in  brain  synaptic  membranes 
from  human  alcoholics.  Neurochem  Res 
15(11):1055-1063,  1990. 

Miyakawa,  T.;  Yagi,  T.;  Kitazawa,  PL; 
Yasuda,  M.;  Kawai,  N.;  Tsuboi,  K.;  and 
Niki,  H.  Fyn-kinase  as  a  determinant  of 
ethanol  sensitivity:  Relation  to  NMDA- 
receptor  function.  Science  278(Oct 
24):698-701,  1997. 

Montoliu,  C;  Valles,  S.;  Renau-Piqueras, 
J.;  and  Guerri,  C.  Ethanol-induced  oxy- 
gen radical  formation  and  lipid  peroxida- 
tion in  rat  brain:  Effect  of  chronic  alcohol 
consumption.  J  Neurochem  63(5): 
1855-1862,  1994. 

Montoliu,  C;  Sancho-Tello,  M.;  Azorin, 
L;  Burgal,  M.;  Valles,  S.;  Renau-Piqueras, 
J.;  and  Guerri,  C.  Ethanol  increases  cy- 
tochrome P4502E1  and  induces  oxidative 
stress  in  astrocytes.  J  Neurochem 
65:2561-2570, 1995. 

Muuronen,  A.;  Bergman,  PL;  Hindmarsh, 
T.,  and  Telakivi,  T.  Influence  of  improved 
drinking  habits  on  brain  atrophy  and  cog- 
nitive performance  in  alcoholic  patients:  A 
5 -year  follow-up  study.  Alcohol  Clin  Exp 
Res  13:137-141,  1989. 

Nakano,  T.;  Fujimoto,  T.;  Shimooki,  S.; 
Fukudome,  T.;  Uchida,  T.;  Tsuji,  T.; 


204 


Neurotoxicity  of  Alcohol 


Mitsuyama,  Y.;  Akimoto,  H.;  and 
Furukawa,  S.  Transient  elevation  of 
nerve  growth  factor  content  in  the  rat 
hippocampus  and  frontal  cortex  by 
chronic  ethanol  treatment.  Psychiatry 
Clin  Neurosci  50(3):157-160,  1996. 

Nordmann,  R.  Oxidative  stress  from 
alcohol  in  the  brain.  Alcohol  Alcohol 
Suppl  1:75-82,  1987. 

Nordmann,  R.;  Ribiere,  C;  and  Rouach, 
H.  Ethanol-induced  lipid  peroxidation 
and  oxidative  stress  in  extrahepatic  tissues. 
Alcohol  Alcohol  25(2-Z):23I-237,  1990. 

Nordmann,  R.;  Ribiere,  C;  and  Rouach, 
H.  Implication  of  free  radical  mechanisms 
in  ethanol  induced  cellular  injury.  Free 
Radic  Biol  Med  12(3):219-240,  1992. 

Ortiz,  J.;  Fitzgerald,  L.W.;  Charlton,  M.; 
Lane,  S.;  Trevisan,  L.;  Guitart,  X.; 
Shoemaker,  W.;  Duman,  R.S.;  and  Nestler, 
E.J.  Biochemical  actions  of  chronic  ethanol 
exposure  in  the  mesolimbic  dopamine 
system.  Synapse  21:289-298,  1995. 

Oscar-Berman,  M.,  and  Hutner,  N. 
Frontal  lobe  changes  after  chronic  alcohol 
ingestion.  In:  Hunt,  W.A.,  and  Nixon, 
S.J.,  eds.  Alcohol-Induced  Brain  Damage. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  121-156. 

Parsons,  O.A.  Impaired  neuropsychologi- 
cal cognitive  functioning  in  sober  alco- 
holics. In:  Hunt,  W.A.,  and  Nixon,  S.J., 
eds.  Alcohol-Induced  Brain  Damage. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  173-194. 

Pfefferbaum,  A.,  and  Rosenbloom,  M.  In 
vivo  imaging  of  morphological  brain 
alterations  associated  with  alcoholism.  In: 


Hunt,  W.A.,  and  Nixon,  S.J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  22.  NIH  Pub. 
No.  93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  71-87. 

Pfefferbaum,  A.;  Lim,  K.O.;  Desmond, 
J.E.;  and  Sullivan,  E.V.  Thinning  of  the 
corpus  callosum  in  older  alcoholic  men:  A 
magnetic  resonance  imaging  study.  Alcohol 
Clin  Exp  Res  20:752-757,  1996. 

Pfefferbaum,  A.;  Sullivan,  E.V.; 
Rosenbloom,  M.J.;  Mathalon,  D.H., 
and  Lim,  K.O.  A  controlled  study  of 
cortical  gray  matter  and  ventricular 
changes  in  alcoholic  men  over  a  5 -year 
interval.  Arch  Gen  Psychiatry 
55(10):905-912,  1998. 

Porjesz,  B.,  and  Begleiter,  H.  Neuro- 
physiological  factors  associated  with  alco- 
holism. In:  Hunt,  W.A.,  and  Nixon,  S.J., 
eds.  Alcohol-Induced  Brain  Damage. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  89-120. 

Portera-Cailliau,  C;  Price,  D.L.;  and 
Martin,  L.J.  Non-NMDA  and  NMDA 
receptor-mediated  excitotoxic  neuronal 
deaths  in  adult  brain  are  morphologically 
distinct:  Further  evidence  for  an  apopto- 
sis- necrosis  continuum.  /  Comp  Neurol 
378(1):88-104,  1997. 

Raine,  A.;  Buchsbaum,  M.S.;  Stanley,  J.; 
Lottenberg,  S.;  Abel,  L.;  and  Stoddard,  J. 
Selective  reductions  in  prefrontal  glucose 
metabolism  in  murderers.  Biol  Psychiatry 
36(6):365-373,  1994. 

Ramanathan,  K;  Wilkemeyer,  M.F.;  Mitral, 
B.;  Perides,  G.;  and  Charness,  M.E.  Alcohol 
inhibits  cell-cell  adhesion  mediated  by 
Human  LI. /C*/Z£w/133(22):381-390, 


205 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


1996.  [Erratum  appears  in  / Cell  Biol 
133(5):  1139-1140,  1996.] 

Rao,  A.,  and  Craig,  A.M.  Activity  regulates 
the  synaptic  localization  of  the  NMDA 
receptor  in  hippocampal  neurons.  Neuron 
19(4):801-812,  1997. 

Rouach,  H.;  Park,  M.K.;  Orfanelli,  M.T.; 
Janvier,  B.;  and  Nordman,  R.  Ethanol  - 
induced  oxidative  stress  in  the  rat  cerebel- 
lum. Alcohol  Alcohol  22(Suppl  1):207- 
211,1987. 

Rudolph,  J.G.,  and  Crews,  F.T.  No  change 
in  NMDA  receptor  binding  in  rat  brain 
following  chronic  ethanol  treatment. 
Alcohol:  Clin  Exp  ito20(2):8A,  1996. 

Sakhi,  S.;  Bruce,  A.;  Sun,  N.;  Tocco,  G.; 
Baudry,  M.;  and  Schreiber,  S.S.  P53 
induction  is  associated  with  neuronal 
damage  in  the  central  nervous  system.  Proc 
Natl  Acad  Sci  USA  92:7525-7529,  1994. 

Schreiber,  S.S.;  Sakhi,  S.;  Dugich- 
Djordjevic,  M.M.;  and  Nichols,  N.R. 
Tumor  suppressor  p53  induction  and 
DNA  damage  in  hippocampal  granule 
cells  after  adrenalectomy.  Exp  Neurol 
130:368-376,  1994. 

Schulteis,  G.;  Hyytia,  P.;  Heinrichs,  S.C.; 
and  Koob,  G.F.  Effects  of  chronic  ethanol 
exposure  on  oral  self-administration  of 
ethanol  or  saccharin  by  Wistar  rats.  Alcohol 
Clin  Exp  Res 20(l):l64-l7l,  1996. 

Snell,  L.D.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  Radioligand  binding  to  the  N- 
methyl - D- aspartate  receptor/ionophore 


complex:  Alterations  by  ethanol  in  vitro 
and  by  chronic  in  vivo  ethanol  ingestion. 
Brain  Res  602(l):9 1-98,  1993. 

Sullivan,  E.V.;  Marsh,  L.;  Mathalon,  D.H.; 
Lim,  K.O.;  and  Pfefferbaum,  A.  Relationship 
between  alcohol  withdrawal  seizures  and 
temporal  lobe  white  matter  volume  deficits. 
Alcohol  Clin  Exp  Res  20(2):348-354, 1996. 

Szabo,  G.;  Tabakoff,  B.;  and  Hoffman, 
P.L.  The  NMDA  receptor  antagonist 
dizocilpine  differentially  affects  environment- 
dependent  and  environment-independent 
ethanol  tolerance.  Psychopharmacology 
(Berl)  113:511-517,  1994. 

Trevisan,  L.;  Fitzgerald,  L.W.;  Brose,  N.; 
Gasic,  G.P.;  Heinemann,  S.F.;  Duman, 
R.S.;  and  Nestler,  E.J.  Chronic  ingestion 
of  ethanol  up-regulates  NMDAR1  receptor 
subunit  immunoreactivity  in  rat  hippocam- 
pus. J  Neurochem  62:1635-1638,  1994. 

Uysal,  M.;  Keyer-Uysal,  M.;  Kocak-Toker, 
N.;  and  Aykac,  G.  Effect  of  chronic 
ethanol  ingestion  on  brain  lipid  peroxide 
and  glutathione  levels  in  rats.  Drug 
Alcohol  Depend  18(l):73-75,  1986. 

Uysal,  M.;  Kutalp,  G.;  Ozdemirler,  G.; 
and  Aykac,  G.  Ethanol-induced  changes 
in  lipid  peroxidation  and  glutathione 
content  in  rat  brain.  Drug  Alcohol  Depend 
23(3):227-230,  1989. 

Victor,  M.;  Adams,  R.D.;  and  Collins, 
G.H.  The  Wernicke-Korsakoff  Syndrome 
and  Related  Disorders  Due  to  Alcoholism 
and  Malnutrition.  Philadelphia,  FA. 
Davis,  1989. 


206 


ADDICTION 

AND  OTHER  BEHAVIORS 

IN  ANIMAL  MODELS 


Chapter  6 

Basic  Behavioral  Effects  and  Underlying 
Neurocircuitries  of  Alcohol 

Kathleen  A.  Grant,  Ph.D. 


KEY  WORDS:  AODE  (effects  of  AOD  [alcohol  or  other  drug]  use,  abuse,  and 
dependence);  neuron;  brain  function;  animal  behavior;  locomotion;  uncondi- 
tioned response;  anxiety  state;  conditioned  response;  dose  effect  relationship;  drug 
discrimination;  neurotransmitter  receptors;  taste  perception;  reinforcement; 
AOD  preference;  animal  model;  self  administration  of  drugs;  learning;  memory; 
cognition;  aggressive  behavior;  risk  factors;  literature  review 


Higher  organisms  interact  with  the 
environment  primarily  through  condi- 
tioned behavioral  processes,  and  the 
nervous  system  is  where  environmen- 
tal information  is  integrated  and 
behavioral  responses  are  generated. 
Thus,  the  integrity  of  behavior  reflects 
the  integrity  of  the  nervous  system 
(Weiss  and  Cory-Slechta  1994).  Alco- 
holism is  mediated  through  the  ner- 
vous system,  but  its  primary  diagnosis 
is  behavioral,  not  neurological.  Thus, 
to  understand  the  behavioral  disorder 
of  alcoholism  we  must  integrate  what 
we  know  about  the  determinants  of 
behavioral  responses  with  our  knowl- 
edge of  alcohol's  action  on  the  ner- 


vous system.  The  neurobiological 
basis  of  alcohol's  behavioral  effects 
can  be  characterized  by  co-application 
of  modern  approaches  to  measuring 
behavior  and  brain  function. 

NEUROBIOLOGICAL 
CHARACTERIZATIONS 
OF  NEURAL  CIRCUITRY 
IN  BEHAVIOR 

The  vertebrate  brain  appears  to 
process  information  through  anatomical 
specialization  of  function.  For  nearly  a 
century  it  has  been  clear  that  specific 
brain  areas  are  involved  in  the  process- 
ing of  sensory,  motor,  or  cognitive 


K.A.  Grant,  Ph.D.,  is  a  professor  in  the  Department  of  Physiology  and  Pharmacology,  Wake  Forest 
University  School  of  Medicine,  Medical  Center  Blvd.,  Winston-Salem,  NC  27157-1083. 


209 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


information,  in  an  arrangement  com- 
monly referred  to  as  localization  of 
function  (Cohen  and  Bookheimer 
1994).  However,  localization  does 
not  imply  isolation,  and  it  is  also  clear 
that  there  is  an  orchestration  of  infor- 
mation from  all  brain  areas  that 
results  in  appropriate  and  predictable 
behavior.  There  are  a  number  of  tech- 
niques currently  available  that  have 
been  used  to  study  the  integration  of 
information  within  the  brain  as  it 
relates  to  behavior.  These  techniques 
have  differing  degrees  of  resolution 
along  several  dimensions,  suggesting 
that  complementary  and  simultaneous 
use  of  two  or  more  techniques  will 
ultimately  provide  the  extensive  infor- 
mation necessary  to  understand  the 
neural  basis  of  behavior.  No  technique 
to  date  has  completely  identified  the 
neural  circuitry  associated  with  a  com- 
plex behavior  such  as  drinking  alcohol 
to  intoxication.  However,  we  do  have 
a  basis  for  beginning  to  speak  in 
terms  of  neural  function,  brain  cir- 
cuitry, and  behavior. 

The  major  modern  techniques  for 
understanding  neural  function  in  a 
behaving  organism  can  be  divided  into 
two  categories,  based  upon  whether  the 
technique  measures  changes  within 
only  a  single  locus  of  a  neural  network 
or  within  the  entire  neural  network. 
Those  techniques  that  gather  measures 
only  from  a  discrete  locus  are  single  - 
unit  recordings  of  neurons;  in  vivo 
voltammetry  of  extracellular  ions  and 
neurotransmitters;  in  vivo  microdialy- 
sis  of  extracellular  ions  and  neuro- 
transmitters; and  site -specific  injections 
of  receptor  ligands  or  neurotoxins. 
Those  techniques  that  gather  measures 


from  entire  neural  networks  or  trans- 
mitter systems  have  the  potential 
advantage  in  identifying  the  conglom- 
eration of  relatively  small  effects  that 
sum  together  and  correlate  with 
behavior.  For  example,  relatively  small 
effects  noted  with  individual  neurons 
may  be  overlooked  when  using  serial 
sampling  of  single-unit  recording,  but 
simultaneous  occurrence  of  many 
small  effects  may  show  much  larger 
effects  when  measured  by  ensemble 
recording  (multi-unit  recording). 
Important  procedures  that  address 
network  circuitry  are  ensemble 
recording  of  multiple  neurons  with 
microwire  arrays;  site-specific  injec- 
tions of  multiple  sites;  quantitative  2- 
deoxyglucose  sequestration  reflecting 
energy  utilization;  neurotransmitter 
turnover  rates  reflecting  activity  of 
neurotransmitter  systems;  functional 
magnetic  resonance  imaging  (MRI)  of 
blood  flow;  positron  emission  tomog- 
raphy (PET)  of  glucose  utilization, 
blood  flow,  or  ligand  binding;  trans- 
genic knockouts,  knockins,  and 
knockdowns;  and  event-related  poten- 
tials (ERPs). 

Each  of  these  techniques  has  strengths 
and  weaknesses  in  measuring  neuronal 
activity  associated  with  behavioral  events. 
These  strengths  and  weaknesses  can  be 
summarized  by  examining  the  resolution 
of  brain  activity  along  spatial  and  tem- 
poral parameters,  as  well  as  the  inva- 
siveness and  longevity  of  the  preparation 
(Cohen  and  Bookheimer  1994).  Since 
it  is  the  spatiotemporal  pattern  of 
neuronal  firing,  within  and  between 
neural  networks,  that  is  believed  to 
underlie  the  actual  orchestration  of  a 
behavioral  pattern,  the  resolution  of 


210 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


information  along  these  dimensions  is 
critical  to  understanding  the  neural 
control  of  behavior.  The  invasiveness 
and  longevity  of  a  preparation  will 
limit  either  the  behaviors  that  can  be 
studied  or  the  repeated  analysis  of  a 
behavior. 

Along  the  spatial  dimension,  the 
best  resolution  is  at  the  level  of  the 
neuron,  which  can  be  achieved  with 
the  genetic  techniques  of  gene  inser- 
tion and  deletion.  Electrophysiologi- 
cal recordings  such  as  single-unit  and 
ensemble  recordings  can  discriminate 
at  the  level  of  individual  neurons  (10- 
50  \im).  The  technique  of  voltammetry 
has  a  similar  resolution,  within  the 
30-50  ^im  range.  Quantitative  2- 
deoxyglucose  and  functional  MRI 
(fMRI)  have  spatial  resolutions  in  the 
50-100  n-m  range  and  can  sample 
from  neuronal  layers  or  at  the  subnuclei 
level.  Similar  resolution  is  achieved 
with  neurotransmitter  turnover  stud- 
ies that  use  micropunches  of  tissue, 
depending  on  the  transmitter  being 
studied.  Microdialysis  requires  a  probe 
that  is  in  the  100-250  |am  range;  how- 
ever, this  technique  is  still  able  to  detect 
columnar  or  subnucleic  resolution.  Site- 
specific  injections  of  ligands  or  toxins 
result  in  a  spread  of  material  generally 
agreed  to  be  in  the  200-500  \im 
range,  depending  on  the  brain  area. 
The  resolution  of  PET  depends  on 
the  scanner  but  is  generally  at  the 
level  of  brain  nuclei,  2-8  mm,  and  can 
be  increased  if  images  are  co-registered 
with  nuclear  magnetic  resonance 
(NMR).  Finally,  the  resolution  of 
ERPs  is  at  the  level  of  cortical  lobes 
and  in  the  range  of  centimeters.  The 
most  important  point  of  spatial  reso- 


lution is  the  size  of  the  brain  under 
study.  For  most  animal  models,  the 
useful  cutoff  is  below  the  resolution  of 
PET  and  ERPs  unless  large  species  of 
nonhuman  primates,  such  as  macaques 
or  baboons,  are  used  as  subjects. 

When  choosing  a  technique  to  cor- 
relate neural  function  with  behavior,  a 
temporal  dimension  is  another  very 
important  consideration.  The  fastest 
resolution  is  direct  neural  recording, 
with  the  ability  to  reliably  separate 
spike  trains  in  the  range  of  10  mil- 
liseconds. This  resolution  holds 
equally  well  for  single -unit  or  ensem- 
ble recording.  Event-related  potentials 
have  a  resolution  in  the  range  of  mil- 
liseconds to  seconds  from  stimulus 
onset  to  potential  recording.  Voltam- 
metry also  has  a  resolution  in  the 
200-500  milliseconds  range,  depend- 
ing on  the  length  of  the  applied 
oxidative  voltage  and  the  rate  of  sam- 
pling (commonly  5  Hz).  The  tempo- 
ral resolution  of  fMRI  is  100 
milliseconds,  depending  on  vascular 
response  mechanisms.  Microdialysis 
has  a  relatively  long  resolution  time  of 
2-20  minutes,  primarily  depending  on 
the  sensitivity  of  the  chromatography. 
As  the  sensitivity  of  high-pressure  liq- 
uid chromatography  increases,  the 
quantity  of  dialysate  necessary  for 
analysis  decreases,  resulting  in  less 
time  needed  to  gather  the  sample.  Site- 
specific  injections  of  ligands  require 
several  minutes,  primarily  due  to 
actual  handling  of  the  animal,  but  also 
to  the  kinetics  of  the  drug.  Quantitative 
2-deoxyglucose  typically  requires  an 
incorporation  time  of  45  minutes,  but 
a  majority  of  the  radioactivity  is  taken 
up  within  a  20-minute  period.  Fluo- 


211 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


rodeoxy glucose  (FDG)  incorporation 
is  30  minutes.  Using  a  repetitive  task, 
one  can  measure  circuits  because  each 
unit  of  the  circuit  has  been  repeatedly 
assessed  and  cumulated  over  time.  In 
essence,  2-deoxyglucose  and  FDG 
procedures  provide  an  integrated  his- 
tory of  neural  activation  associated 
with  a  behavior.  PET  analysis  of  lig- 
and  binding  reflects  receptor  changes 
in  number  or  affinity  and,  therefore, 
reflects  neural  changes  that  occur  over 
the  course  of  hours  to  days,  depend- 
ing on  the  stimulus  presentation  (e.g., 
chronic  ethanol  exposure). 

Thus,  if  the  interest  is  in  neural  cir- 
cuitry related  to  emission  of  a  fast 
response,  for  example,  an  approach 
behavior  or  reaction  time,  the  electro- 
physiological techniques  are  most  appro- 
priate. In  addition,  if  one  is  interested  in 
a  component  analysis  of  neuronal  activity 
in  relation  to  aspects  of  the  behavioral 
chain,  single-unit  or  ensemble  record- 
ing is  appropriate.  However,  if  repeti- 
tive tasks  are  used,  microdialysis  and 
turnover  studies  can  help  identify  the 
actual  neurotransmission  pathways 
involved  in  a  response.  Likewise,  glu- 
cose utilization  procedures  and  fMRI 
reflect  the  integrated  history  of  the 
circuitry  involved  in  the  response. 

Another  dimension  to  consider  is  that 
of  invasiveness,  in  terms  of  both  tissue 
damage  and  restricting  behavior.  The 
techniques  of  single-unit  recordings, 
ensemble  recordings,  voltammetry, 
microdialysis,  and  site-specific  injections 
require  placement  of  electrodes,  probes, 
or  cannulas  into  neural  tissue,  resulting 
in  direct  tissue  damage  along  the  probe  or 
electrode  track.  In  addition,  the  electrode 
leads  and  probe  tubing  need  to  be 


physically  attached  to  additional  equip- 
ment, often  restricting  movement  of  the 
animals,  or  at  the  very  least  requiring 
that  a  tether  be  attached  to  the  animal 
during  data  gathering.  Lesion  studies 
are  by  definition  invasive.  Quantitative 
2-deoxyglucose  is  not  invasive  to  brain 
tissue  because  the  radiotracer  is  injected 
into  die  peripheral  circulatory  system, 
leaving  the  central  nervous  system 
(CNS)  intact.  However,  quantitative 
2-deoxyglucose  requires  surgical  place- 
ment of  arterial  catheters,  which  can 
restrict  movement.  Although  the  tech- 
niques of  fMRI  and  PET  are  considered 
noninvasive,  the  measurements  are  very 
sensitive  to  head  motion,  and  misreg- 
istrations due  to  movement  can  create 
significant  artifacts.  Thus,  animals  need 
to  be  anesthetized  or  severely  restricted 
in  movement  during  the  scanning  pro- 
cedure. The  technique  of  FDG  imag- 
ing with  PET  is  somewhat  immune  to 
these  criticisms,  since  the  incorpora- 
tion of  FDG  can  occur  during  an  active 
behavioral  task  and  the  scanning  can 
be  done  under  anesthesia  without 
complicating  the  analysis  of  what  brain 
areas  were  active  during  the  behavior. 
Finally,  although  the  ERP  technique 
is  noninvasive,  the  procedure  requires 
the  animals  to  sit  quietly.  Although 
small  head  movements  are  not  nearly  as 
problematic  as  with  fMRI  or  PET, 
extensive  training  of  nonhuman  sub- 
jects may  be  required. 

A  final  dimension  to  consider  when 
linking  a  technique  to  behavior  is  the 
patency  of  the  preparation  and 
repeated  measures.  For  example,  to 
measure  the  incorporation  of  2- 
deoxyglucose  with  autoradiographic 
techniques  requires  decapitation  and 


212 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


histology.  Likewise,  neurotransmitter 
turnover  studies  requires  rapid  removal 
by  decapitation  and  freezing  of  neural 
tissue.  Therefore,  only  a  single  experi- 
mental manipulation  can  be  measured 
in  an  animal,  and  group  designs  are 
necessary  for  parametric  examinations, 
such  as  dose-response  determinations, 
with  these  techniques.  The  techniques 
of  microdialysis  and  voltammetry  have 
experimental  half-lives  in  terms  of 
days,  and  probably  less  in  terms  of 
voltammetry  specificity.  Site-specific 
injections  are  patent  for  5-7  injections 
of  1-2  [iL  before  direct  tissue  damage 
can  confound  interpretation  of  the 
data.  Single-unit  recording  is  patent 
for  a  number  of  days.  Ensemble 
recording  is  patent  for  an  average  of  2 
weeks,  but  clear  identification  of  the 
same  neuron  across  days  is  problematic. 
Thus  many,  if  not  all,  of  the  techniques 
are  limited  in  longitudinal  designs.  On 
the  other  hand,  fMRI,  PET,  and  ERP 
are  techniques  that  can  be  repeatedly 
administered,  with  caution  provided 
for  the  effect  of  repeated  exposure  to 
anesthetics  in  the  use  of  fMRI  and  PET. 
From  the  descriptions  given  here,  it 
is  clear  that  there  are  many  approaches 
to  studying  the  neural  circuitry  of 
behavior.  It  is  equally  clear  that  each 
technique  has  particular  strengths  and 
weaknesses.  The  complementary  use 
of  techniques  can  increase  the  resolution 
in  defining  the  neural  networks  and 
help  investigators  gain  a  more  com- 
plete understanding  of  the  circuitry  of 
behavior.  For  example,  single-unit 
recordings  can  be  measured  in  con- 
junction with  microdialysis  to  identify 
neural  activity  and  the  neurotransmit- 
ters released  during  this  activity.  This 


information  allows  the  investigator  to 
identify  an  area  of  the  brain  as  well  as 
a  receptor  system,  which  can  then  be 
used  to  test  hypotheses  addressing  the 
neural  basis  of  behavior. 

BEHAVIORAL 
CHARACTERIZATIONS 
IN  ANIMAL  MODELS 

To  understand  the  behavioral  mecha- 
nisms of  alcohol  (ethanol),  we  must 
first  understand  the  variables  that  con- 
trol behavior  and  then  understand 
how  alcohol  interacts  with  these  vari- 
ables. Several  techniques  have  been 
developed  to  model  aspects  of  behavior 
associated  with  the  administration  of 
alcohol  and  other  drugs.  Similar  to  the 
techniques  to  study  neural  circuitry,  the 
behavioral  procedures  focus  on  different 
aspects  of  an  overall  constellation  of 
effects  when  alcohol  is  administered 
to  an  awake  animal.  Thus,  any  single 
behavioral  procedure  in  isolation  will 
not  provide  information  about  all  the 
behavioral  effects  of  alcohol.  In  addi- 
tion, each  procedure  has  particular 
strengths  and  weaknesses,  including 
the  amount  of  training,  the  ability  to 
conduct  parallel  studies  across  different 
species,  the  requirement  for  specialized 
equipment  and  behavioral  expertise, 
and  the  ability  to  incorporate  the  neuro- 
biological  techniques  listed  in  the  pre- 
ceding section.  A  description  of  the 
most  commonly  used  procedures  is 
provided  in  the  following  sections, 
arranged  by  the  class  of  behavior 
being  modeled. 

Psychoactive  drugs  produce  a  num- 
ber of  effects  on  behavior.  Those  effects 
of  a  drug  that  uniquely  covary  with  an 


213 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


aspect  of  behavior  are  called  stimuli. 
Unconditioned  stimuli  elicit  reflex 
responses  without  prior  co-occurrence 
of  stimulus  and  response.  The  uncon- 
ditioned effects  of  ethanol  are  respon- 
sible for  unconditioned  behaviors 
such  as  loss  of  righting  reflex  and  for 
unconditioned  physiological  responses 
such  as  hypothermia.  Conditioning 
involves  a  process  whereby  a  new  asso- 
ciation is  formed  between  the  stimulus 
presentation  and  the  occurrence  of 
behavior.  For  example,  new  behaviors 
can  be  established  in  relation  to  the 
presentation  of  reinforcing  stimulus 
effects,  with  either  increases  or 
decreases  in  behaviors  that  were  instru- 
mental in  producing  the  stimulus. 

The  study  of  the  neurobiological 
bases  of  ethanoPs  behavioral  effects 
necessitates  using  an  ethanol  concentra- 
tion range  compatible  with  behavior. 
Hypotheses  concerning  the  neu- 
ropharmacological  mechanisms  of 
ethanol's  activity  and  the  transference 
of  these  concepts  to  the  realm  of  psy- 
chopharmacology  have  changed  sig- 
nificantly over  the  past  two  decades. 
Ethanol,  along  with  other  anesthetics, 
was  considered  an  exception  to  the 
receptor  theory  of  drug  action  (Smith 
and  Reynard  1995),  in  which  there  is 
a  physical  combination  of  a  drug  with 
a  specific  macromolecule  (Goldstein 
et  al.  1968).  The  effects  of  ethanol  were 
viewed  as  nonspecific,  disrupting  the 
electrochemical  communication  between 
neurons  by  generally  disordering  all 
membrane  activity  through  a  lipid  par- 
titioning effect.  However,  advancements 
in  the  application  of  electrophysiological, 
biochemical,  and  molecular  techniques 
provide  conclusive  data  to  show  that 


ethanol  acts  as  a  modulator  at  particu- 
lar receptor  complexes,  selectively 
altering  neurochemical  processes  in 
discrete  regions  of  the  CNS  (see  chap- 
ter 1).  The  specificity  in  the  mecha- 
nisms of  ethanol's  activity,  in  some 
studies  localized  to  a  handful  of  amino 
acids  (Mihic  et  al.  1997),  reflects 
tremendous  advances  in  defining  the 
basic  neuropharmacology  of  ethanol. 
These  advances  have  also  provided 
rational  avenues  of  research  for  identi- 
fying the  receptor  mechanisms 
involved  in  mediating  ethanol's  stimu- 
lus effects. 

Unconditioned  Behavior 

Unconditioned  behavior  is  often 
referred  to  as  "innate"  or  "reflexive," 
and  does  not  require  learning  on  the 
part  of  the  organism.  Generally  speak- 
ing, organisms  on  the  lower  end  of  the 
phylogenetic  scale  predominanriy  dis- 
play unlearned  behaviors;  however, 
learning  has  been  demonstrated  in  most 
multicellular  organisms.  Although 
these  behaviors  do  not  require  associa- 
tive processes  to  be  established,  uncon- 
ditioned behaviors  such  as  a  motor 
reflex  (or  salivation)  can  be  the  basis 
of  a  conditioning  paradigm.  Thus, 
through  learning  paradigms,  uncondi- 
tioned behaviors  are  associated  with 
stimuli  and  produce  conditioned 
behaviors,  and  these  will  be  discussed 
at  greater  length  later  in  this  chapter. 
In  alcohol  research,  unconditioned 
behaviors  are  often  measured  only  in 
the  context  of  single -dose  exposures. 

Motor  Responses 

With  ethanol  administration,  most 
reflexive  responses  studied  have  been 


214 


Behavioral  Effects  and  Underlying  Neurocircui tries  of  Alcohol 


locomotion  responses,  but  there  are 
notable  exceptions.1  Three  responses — 
open  field  activity,  righting  reflex,  and 
ability  to  remain  on  a  moving 
rod/track — have  been  heavily  used  in 
alcohol  research  addressing  neurobio- 
logies correlates,  and  these  responses 
are  discussed  in  this  section.  Other 
examples  of  unconditioned  motor 
responses  include  exploration  in  mazes, 
orientation  of  an  inclined  plane,  and 
grip  strength  on  an  inverted  screen. 
With  the  exception  of  elevated  plus 
mazes  to  measure  anxiety  (discussed  in 
the  next  section),  these  responses  have 
not  been  used  in  alcohol  research  to  any 
great  extent  and  will  not  be  described 
in  detail  here. 

Open  field  activity,  in  which  hori- 
zontal and  vertical  motor  activity  is 
detected  by  photobeams,  is  a  common 
measure  of  an  unconditioned  locomo- 
tor response  in  alcohol  research.  Open 
field  locomotion  implies  motor  coor- 
dination, but  animals  can  move  though 
the  open  field  in  uncoordinated  activity. 
Thus,  an  intoxicated  animal  that  could 
not  stay  on  a  moving  belt  could  exhibit 
increases  in  open  field  activity.  In 
addition,  patterns  of  behavior,  such  as 
intense  locomotion  followed  by 
immobility  versus  constant  activity,  are 
often  not  measured.  Open  field  loco- 
motion can  detect  changes  induced  by 
ethanol  in  the  1.0-3.0  g/kg  dose  range 
and  has  been  used  to  measure  both 
the  stimulatory  and  sedative  effects  of 
ethanol.  The  animals  of  choice  for 
these  procedures  have  been  the  mouse 
or  the  rat,  and  neurochemical  measures 
associated  with  open  field  activity 
have  focused  on  the  dopaminergic  or 
GABAergic  system.  A  sophisticated 


neuroscience  technique,  the  use  of 
genetically  selected  mice,  has  been 
applied  to  this  analysis.  In  addition, 
quantitative  trait  loci  (QTL)  analyses 
have  been  performed  and  have  pro- 
vided provisional  loci  that  may  contain 
genes  important  for  the  expression  of 
ethanol-induced  unconditioned  stim- 
ulatory locomotion. 

Another  common  measure  is  the 
loss  of  a  righting  reflex  following  a 
high  dose  of  ethanol.  The  primary 
measures  of  behavior  are  the  latency 
to  lose  the  reflex  and  the  duration  the 
reflex  is  lost.  Loss  of  righting  reflex  is 
associated  with  high  doses  of  ethanol 
and  is  incompatible  with  most  other 
alcohol-related  behaviors  in  the  non- 
tolerant  animal.  Mice  and  rats  are  the 
most  common  subjects,  and  the  neu- 
rotransmitter system  studied  most 
extensively  has  been  the  GABAergic 
system,  although  N-methyl-D-aspar- 
tate  (NMDA),  serotonin,  and  neu- 
ropeptidergic  receptor  systems  have 
also  been  explored.  Most  of  the  neu- 
rotransmitter systems  have  been  inves- 
tigated in  isolation,  primarily  through 
the  use  of  receptor  binding,  with  little 
information  on  circuitry.  As  with  the 
locomotor  response,  the  most  sophis- 
ticated technique  applied  to  this 
analysis  has  been  the  use  of  genetically 
selected  rodents.  QTL  analyses  using 
this  measure  have  provided  provi- 
sional loci  for  genes  important  for  the 
expression  of  ethanol-induced  uncon- 
ditioned  impairment  of  motor 
reflexes.  This  measure  has  also  been 
used  extensively  to  study  the  develop- 
ment of  tolerance  to  ethanol. 

Direct  measures  of  motor  coordi- 
nation are  best  represented  in  alcohol 


215 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


research  by  rotorod  or  moving  belt 
performance.  In  these  procedures  the 
animals  are  required  to  stay  on  a  dowel 
or  a  track  that  is  rotating  at  either  a 
constant  or  an  accelerating  speed. 
Although  the  response  of  staying  on 
the  rotorod  or  track  is  unconditioned, 
animals  are  often  given  a  few  training 
sessions  to  have  baseline  performances 
matched  in  group  designs.  Therefore, 
there  is  the  opportunity  for  condition- 
ing if  several  pre-ethanol  trials  are 
given.  Latency  to  fall  off  the  rotorod 
or  step  off  the  track  following  the 
administration  of  ethanol  is  generally 
the  measure  of  interest.  This  response 
is  sensitive  to  lower  doses  of  ethanol 
compared  with  doses  associated  with 
the  loss  of  righting  reflex.  To  date, 
the  response  has  been  used  most  com- 
monly to  assess  sensitivity  to  ethanol 
and  the  development  of  tolerance  in 
mice  and  rats.  Neurochemical  mea- 
sures associated  with  this  response 
have  also  focused  on  the  GABAergic 
system,  although  the  serotonin  system 
has  been  examined  in  the  develop- 
ment of  tolerance. 

Effects  of  ethanol  on  these  uncon- 
ditioned behaviors  have  been  the  basis 
for  selective  breeding  using  alcohol- 
related  phenotypes.  Because  selective 
breeding  requires  that  the  same  pheno- 
type  be  accurately  characterized  each 
generation,  it  is  easy  to  understand  why 
these  unconditioned  motor  responses 
were  chosen  to  demonstrate  a  genetic 
basis  to  an  ethanol-related  behavior. 
However,  it  is  clear  that  drugs  from  a 
wide  variety  of  pharmacological  classes 
can  disrupt  performance  on  these 
tasks.  Thus,  using  these  uncondi- 
tioned responses  to  investigate  genetic 


or  neural  bases  for  ethanol-induced 
behavioral  responses  appears  to  be  based 
more  on  replicating  the  response  than 
on  specificity  or  sensitivity  to  ethanol. 
For  example,  GABAergic  mechanisms 
may  represent  a  focal  point  of  the 
effects  of  ethanol  on  the  nervous  sys- 
tem, or  they  may  be  secondary  to  the 
functional  integrity  of  another  system 
disrupted  by  ethanol.  Motor  reflexive 
behaviors  are  rather  robust  and  gener- 
ally require  high  doses  of  ethanol  for 
alterations  in  response  generation.  In 
turn,  the  high  doses  of  ethanol  are 
likely  to  alter  the  integrity  of  multiple 
neural  circuits,  and  pinpointing  which 
neural  mechanism(s)  is(are)  involved 
may  be  impossible.  To  date,  the  use 
of  specific  receptor  ligands,  such  as 
inverse  agonists  at  GABAA  receptors, 
has  attenuated  but  not  completely 
antagonized  the  effects  of  ethanol  on 
these  tasks. 

With  the  technology  available  today 
to  selectively  knock  out  genes,  it  seems 
unnecessary  to  continue  to  extrapolate 
from  these  unconditioned  responses  to 
mechanisms  associated  with  the  behav- 
ioral basis  of  alcohol  abuse  and  alcohol- 
ism. Possible  exceptions  to  this  conclusion 
could  be  the  study  of  unconditioned 
motor  responses  to  ethanol  in  testing 
specific  hypotheses  such  as  the  con- 
cordance of  tolerance  to  the  motor 
effects  of  ethanol  and  increases  in 
ethanol  self- administration.  However, 
such  tolerance  to  ethanol  is  easily  gath- 
ered within  self- administration  or  place 
preference  procedures,  in  the  context  of 
appropriate  dose  ranges.  Thus,  although 
unconditioned  motor  behaviors  are  very 
useful  for  determining  dose  ranges  of 
activity  and  can  serve  as  initial  screens  for 


216 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


receptor-mediated  activity,  they  are 
not  particularly  useful  for  understanding 
the  neural  circuitry  involved  in  the 
process  of  alcohol  addiction. 

Anxiolytic  Responses 

A  constellation  of  unconditioned 
responses  that  has  implications  for  the 
abuse  potential  of  alcohol  is  derived 
from  unfamiliar  and  extreme  environ- 
mental settings.  Unconditioned  behav- 
iors exhibited  in  extreme  heights,  in 
the  presence  of  an  unfamiliar  individual, 
or  in  the  presence  of  bright  lighting  have 
all  been  used  as  measures  of  anxiety  in 
mice  and  rats  (Lister  1990).  The  ele- 
vated plus  maze  measures  the  uncon- 
ditioned responses  of  rodents  to  stay  in 
enclosed  spaces  and  to  avoid  cliffs.  In 
this  procedure,  rats  or  mice  are  placed 
in  a  maze  with  a  central  post  that  elevates 
the  maze.  Radiating  out  from  the  central 
post  are  four  arms  that  are  perpendicular 
to  each  other.  Two  of  the  maze  arms, 
opposite  to  one  another,  have  sides 
(closed  arms),  and  the  other  two  arms 
have  no  sides  (open  arms).  When 
placed  in  the  maze,  undrugged  animals 
spend  most  of  their  time  in  the  closed 
arms  of  the  maze;  anxiolytics  and  alcohol 
increase  the  amount  of  time  spent  in  the 
open  arms  of  the  maze  (Lister  1990; 
but  see  Dawson  and  Tricklebank  1995). 
The  social  interaction  test  measures  the 
unconditioned  responses  of  rodents  to 
engage  in  social  investigation.  The 
social  interaction  test  capitalizes  on  the 
observation  that  social  interactions 
decrease  when  animals  are  placed  in 
unfamiliar  or  brightly  illuminated  envi- 
ronments. Typically,  anxiolytics  increase 
the  social  interactions  of  rats  in  these 
environments,  and  alcohol  has  also 


been  shown  to  increase  social  interac- 
tions under  these  conditions.  The  light- 
dark  box  test  measures  the  tendency 
of  rats  and  mice  to  avoid  bright  light  by 
placing  the  animal  in  a  two-compartment 
shuttle  box  in  which  one  side  is  brightly 
illuminated  and  the  other  is  darkened. 
Anxiolytics  typically  increase  time  spent 
in  the  illuminated  side  of  the  chamber. 
In  the  mouse,  ethanoPs  anxiolytic 
effects  are  in  the  dose  range  of  1.5-2.5 
g/kg  ethanol  (intraperitoneal  [ip]), 
corresponding  to  blood  ethanol  con- 
centrations of  150  mg/dL.  Lower 
doses  in  the  range  of  0.5-1.0  g/kg 
(ip),  corresponding  to  100  mg/dL 
ethanol,  produce  anxiolytic-like  effects 
in  rats.  The  receptor  system  most 
extensively  studied  with  regard  to 
unconditioned  anxiolytic  behaviors  is 
the  GABAA  receptor  system,  and  this  is 
the  system  that  has  been  most  consis- 
tently implicated  in  the  effects  of  ethanol 
on  these  unconditioned  responses.  No 
specific  information  is  available  con- 
cerning the  neural  circuitry  of  ethanoPs 
anxiolytic  responses  using  these  mea- 
sures. Most  often,  GABAA  inverse 
agonists  have  been  tested  in  combina- 
tion with  ethanol  to  block  ethanoPs 
anxiolytic  responses.  However,  GABAA 
inverse  agonists  are  anxiogenic,  and 
the  attenuation  of  ethanoPs  effects 
could  be  due  to  two  separate  mecha- 
nisms, canceling  each  other's  effects. 
Flumazenil  does  not  appear  to  block 
the  anxiolytic  effects  of  acute  ethanol 
in  conditioned  conflict  procedures. 
Finally,  correlations  have  been  found 
between  preference  for  alcohol  solutions 
and  basal  expression  of  these  responses 
in  some  (P/NP  and  SP/SNP  rats), 
but  not  all,  genetic  lines  selectively 


217 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


bred  for  alcohol  preference  (AA/ANA) 
(reviewed  in  Eckhardt  et  al.  1998). 

Vocalization  is  another  type  of 
unconditioned  response  that  is  elicited 
under  circumstances  associated  with 
changes  in  extreme  changes  in  the 
environment,  including  maternal  or 
conspecific  separation.  Although 
ethanol  administration  will  increase 
auditory  vocalizations,  particularly  in 
nonhuman  and  human  primates,  these 
vocalizations  are  more  affiliative  and 
associated  with  increased  social  inter- 
action. In  contrast,  many  laboratory 
species,  including  monkeys,  have  a 
range  of  auditory  information  that  is 
in  the  ultrasonic  range  of  human  per- 
ception. These  ultrasonic  vocalizations 
are  often  associated  with  distress,  either 
social  separation  or  hypothermia.  Low 
doses  of  ethanol  tend  to  suppress 
stress-induced  ultrasonic  vocalizations; 
however,  investigation  of  the  neuro- 
circuitry  involved  in  ethanol-associated 
changes  in  vocalizations  has  been  rudi- 
mentary. The  GABAergic  and  5-ETTi 
receptor  systems  have  been  investigated 
through  the  use  of  specific  ligands, 
but  pathway-specific  delineations  have 
not  been  made. 

In  general,  the  neurobiological 
investigation  of  unconditioned  anxi- 
olytic responses  to  ethanol  has  not 
extended  beyond  correlational  studies 
with  selected  or  inbred  lines  of  rodents 
or  use  of  specific  receptor  ligands.  The 
unconditioned  responses  associated  with 
anxiety  appear  to  provide  relatively  more 
construct  validity  than  unconditioned 
motor  responses  in  efforts  to  under- 
stand the  neurobiological  basis  of 
ethanol  related  to  its  abuse.  However, 
very  few  studies  have  actually  linked 


initial  anxiolytic  effects  of  ethanol  in 
these  measures  and  subsequent  ethanol 
self- administration  or  preference  in  a 
within-subjects  design.  An  important 
caveat  to  remember  is  that  the  "anxi- 
olytic" effects  of  ethanol  measured  in 
these  procedures  involve  the  response 
to  the  first  injection  of  ethanol  in  a  naive 
animal.  Under  these  circumstances,  it 
has  clearly  been  shown  that  ethanol 
dramatically  heightens  activity  of  the 
hypothalamic-pituitary-adrenal  (HPA) 
axis,  an  effect  that  is  normally  correlated 
with  increased  stress  and  anxiety  (Rivier 
1996).  Clearly,  there  is  a  complicated 
interaction  between  the  expression  of 
the  unconditioned  responses  associated 
with  "fear"  and  the  effects  of  ethanol 
(see  the  discussion  of  conditioned 
anxiolytic  stimulus  effects  later  in  this 
chapter).  Important  factors  include  the 
motoric  responses  to  ethanol,  including 
both  stimulatory  and  sedative  effects, 
either  of  which  could  alter  the  mea- 
surement and  interpretation  of  these 
responses.  These  confounds  are  not 
unique  to  ethanol,  prompting  leading 
investigators  to  conclude  that  "it  is 
difficult  to  justify  [use  of  the  elevated 
plus  maze]  as  anything  other  than  a 
preliminary  screen  as  a  prelude  to  test- 
ing more  robust  animal  models  of 
anxiety"  (Dawson  and  Tricklebank 
1995,  p.  36). 

On  the  other  hand,  the  use  of 
unconditioned  responses  to  demonstrate 
anxiogenic  states  associated  with  ethanol 
withdrawal  may  be  useful  in  identifying 
mechanisms  underlying  these  states. 
For  example,  the  GABAA  benzodi- 
azepine site  antagonist  flumazenil  can 
block  the  anxiogenic  effects  of  ethanol 
withdrawal  in  an  elevated  plus  maze 


218 


Behavioral  Effects  and  Underlying  Neurocircui tries  of  Alcohol 


(Moy  et  al.  1997).  This  is  an  unusual 
finding,  since  ethanol  is  not  believed 
to  interact  directly  with  the  benzodi- 
azepine site,  and  flumazenil  does  not 
block  the  anxiolytic  effects  of  ethanol. 
It  has  been  suggested  that  ethanol 
withdrawal  increases  the  level  of 
diazepam  binding  inhibitor  (DBI) 
protein  (see  Moy  et  al.  1997).  Another 
study  found  that  a  corticotropin- 
releasing  factor  (CRF)  antagonist  can 
block  the  effects  of  ethanol  withdrawal 
on  the  elevated  plus  maze,  suggesting 
that  ethanol  withdrawal  increases  CRF 
(Baldwin  et  al.  1991).  The  promise  of 
these  unconditioned  responses  in 
studies  of  ethanol  withdrawal  lies  in 
the  need  to  characterize  the  short 
time  course  of  the  ethanol  withdrawal 
in  rodents. 

First  Dose  Effects 

Unconditioned  responses  to  ethanol 
have  been  studied  primarily  in  the 
context  of  first  dose  effects.  Although 
easy  to  implement,  first  dose  effects 
need  to  be  evaluated  in  terms  of  the 
potential  to  help  understand  the  alco- 
hol addiction  process.  A  vast  majority 
of  people  have  encountered  a  "first 
dose"  of  alcohol,  yet  only  5  to  10  per- 
cent of  the  population  establish  a  pat- 
tern of  behavior  that  results  in  alcohol 
abuse  or  addiction.  The  neurophar- 
macologies basis  of  these  responses 
has  been  difficult  to  discern,  often 
because  false-negative  data  can  be 
extensive  (Dawson  and  Tricklebank 
1995)  or  because  the  response  can  be 
produced  (and  attenuated)  by  many 
different  pharmacological  agents. 
More  sophisticated  apparent  pA2 
analyses  of  antagonists  could  help 


identify  receptor  heterogeneity  in 
behavioral  assays  (Kenakin  1993),  but 
these  analyses  have  not  been  applied. 
Thus,  responses  associated  with  the 
first  dose  effects  have  not  been,  and 
may  never  be,  useful  in  determining 
the  underlying  neuropharmacology  of 
ethanol  associated  with  abuse  and 
addiction.  However,  these  uncondi- 
tioned responses  to  ethanol  have  been 
successfully  used  in  selectively  breeding 
animals  for  response  to  ethanol.  The 
selective  breeding  approaches  have 
shown  beyond  a  doubt  that  there  is  a 
genetic  basis  of  these  responses  to 
ethanol.  However,  this  no  longer  seems 
an  appropriate  goal  for  behavioral 
genetic  studies,  because  techniques 
are  now  available  to  target  specific 
genes  in  relation  to  more  complex 
behaviors  associated  with  ethanol. 
The  characterizations  of  first  dose 
effects  are  also  necessary  in  following 
the  development  of  tolerance  or  sensi- 
tization to  ethanol.  However,  toler- 
ance and  sensitization  are  processes 
that  primarily  use  a  repeated  dosing 
design.  Therefore,  it  would  seem  that 
studying  the  effects  of  only  an  initial 
single  dose  of  alcohol  is  not  sufficient 
to  understand  the  neurobiological 
basis  of  behavior  associated  with  alco- 
hol's use  and  abuse. 

Conditioned  Behavior 

Behaviors  that  are  classified  as  learned, 
acquired,  or  conditioned  can  be  mod- 
ified by  environmental  events,  and  the 
resultant  behaviors  can,  in  turn,  alter  the 
immediate  environment.  The  dynamic 
interaction  between  the  environment 
and  behavioral  responses  serves  to  pro- 
duce, refine,  and  eliminate  conditioned 


219 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


behaviors  (Weiss  and  Cory-Slechta 
1994).  This  constant  redefining  of 
behavioral  responses  within  environ- 
mental contexts  provides  a  rich  behav- 
ioral repertoire,  emanating  from  a 
complex  set  of  neurobiological 
processes.  Alcohol  can  interact  to  dif- 
ferent degrees  with  any  of  these 
neurobiological  processes  and  alter 
the  behavioral  response  to  the  envi- 
ronment. It  is  incumbent  upon  behav- 
ioral science  to  direct  research  in 
animal  models  that  is  accessible  to 
modern  neurobiological  techniques 
and  to  translate  the  results  of  this 
research  into  meaningful  principles  to 
guide  research  in  humans.  In  alcohol 
research,  conditioned  behaviors  have 
primarily  focused  on  alcohol's  stimu- 
lus effects  associated  with  discrimina- 
tion, anxiety,  reinforcement,  learning 
and  memory,  and  aggression.  In  addi- 
tion, the  processes  of  tolerance  and 
the  roles  of  acute  withdrawal  and 
stress  on  these  stimulus  effects  have 
been  the  subject  of  intensive  studies 
and  theoretical  debates. 

Discriminative  Stimulus  Effects 

Discriminative  stimuli  specifically 
covary  with  the  availability  of  rein- 
forcement. Most  early  investigations 
of  discriminative  stimuli  used  external 
environmental  stimuli  perceived 
through  sensory  mechanisms.  However, 
it  is  clear  that  the  internally  produced 
(interoceptive)  stimulus  effects  of  a 
psychoactive  drug  exert  robust  stimulus 
control.  In  simple  drug  discrimination 
studies,  the  animal  is  trained,  through 
differential  reinforcement,  to  engage 
in  a  particular  behavior  in  the  presence 
of  the  internal  effects  of  the  drug,  and 


to  engage  in  a  different  behavior  in 
the  absence  of  the  internal  effects  of 
the  drug.  Thus,  the  discrimination 
paradigm  provides  a  measure  of  the 
association  between  interoceptive  sen- 
sations and  observable  behaviors,  and 
is  often  interpreted  to  be  an  animal 
model  of  the  subjective  effects  of 
drugs  (Preston  and  Bigelow  1991). 

Drug  discrimination  procedures  have 
been  refined  to  provide  one  of  the  most 
powerful  avenues  of  research  for  char- 
acterizing the  pharmacological  aspects 
of  the  drug  in  relation  to  behavior.  As 
with  other  procedures  in  the  analysis  of 
behavior,  the  reliable  baseline  of  behavior 
produced  by  drug  discrimination  pro- 
cedures allows  a  systematic  approach  to 
characterizing  the  influence  of  pharma- 
cological variables.  Over  the  past  half- 
century,  these  procedures  have  been  used 
extensively  to  characterize  the  pharmaco- 
logical effects  of  many  drugs  of  abuse 
(Colpaert  1986).  Data  accumulated  over 
these  years  have  demonstrated  that  the 
discriminative  stimulus  effects  of  drugs 
vary  along  quantitative  and  qualitative 
dimensions  and  have  characteristics 
indicative  of  receptor- mediated  activity 
(Holtzman  1990).  Drug  discrimina- 
tion procedures  have  proved  to  be  a 
reliable  and  valuable  tool  for  screening 
substances  for  abuse  potential,  character- 
izing potential  antagonists,  identifying 
active  metabolites,  and  establishing 
structure-activity  relationships  of  psy- 
choactive substances.  Because  of  their 
usefulness  in  these  areas,  drug  discrim- 
ination procedures  have  become  an 
important  method  for  categorizing  and 
adding  information  regarding  the  phar- 
macological action  of  drugs  with  simi- 
lar behavioral  profiles. 


220 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


The  specificity  of  drug  discrimination 
procedures,  as  they  apply  to  ethanol, 
is  perhaps  best  illustrated  by  the  abil- 
ity of  these  procedures  to  separate  the 
effects  of  ligands  at  the  various  sites 
on  the  GABAA  receptor  complex. 
Specifically,  muscimol  and  4,5,6,7- 
tetrahydroisoxazolo(  5 ,4-c  )pyridin-  3  - 
ol  (THIP)  do  not  substitute  for 
midazolam,  diazepam,  pentobarbital, 
or  ethanol  (Ator  and  Griffiths  1989; 
Grech  and  Balster  1993;  Shelton  and 
Balster  1994).  Pentobarbital  and 
midazolam  result  in  partial  substitution 
for  muscimol  and  THIP  (Grech  and 
Balster  1997).  These  data  imply  that 
positive  modulators  of  GABAA  recep- 
tors, including  ethanol,  produce  stim- 
ulus effects  that  are  fundamentally 
different  than  those  produced  by 
direct  GAB  A  agonists.2  In  contrast  to 
the  GAB  A- site  agonists,  GABAA  posi- 
tive modulators  consistently  produce 
ethanol-like  discriminative  stimulus 
effects.  Specifically,  barbiturates  that 
produce  ethanol  discriminative  stimu- 
lus effects  are  pentobarbital,  pheno- 
barbital,  or  barbital  in  rats  and 
pentobarbital  in  monkeys  (Grant  et  al. 
1996).  Ligands  at  the  benzodiazepine 
site  that  produce  ethanol-like  effects 
include  chlordiazepoxide,  lorazepam, 
and  midazolam  in  rats;  chlordiazepox- 
ide in  gerbils;  diazepam  in  pigeons; 
and  oxazepam  in  mice  (Sanger  1997). 
In  addition  to  positive  modulators  at 
the  barbiturate  and  benzodiazepine 
site  on  GABAA  receptors,  positive 
modulators  at  the  neurosteroid  site  on 
the  GABAA  receptors  also  produce 
ethanol-like  discriminative  stimulus 
effects  in  rats  (Ator  et  al.  1993)  and 
monkeys  (Grant  et  al.  1996).  Thus, 


positive  modulation  of  the  GABAA 
receptor  system  appears  to  be  a  robust 
component  of  the  ethanol  cue. 

Diazepam-sensitive  receptors  can 
be  divided  into  BZ1  receptors,  with 
high  affinity  for  Zolpidem,  and  BZ2 
receptors,  with  low  affinity  for  Zolpi- 
dem. There  is  mounting  evidence  that 
drug  discrimination  procedures  differ- 
entiate BZ1  ligands  from  benzodi- 
azepines that  have  activity  at  both 
BZ1  and  BZ2  receptor  subtypes. 
Zolpidem  results  in  only  partial  sub- 
stitution in  chlordiazepoxide  discrimi- 
nation, and  benzodiazepines  result  in 
partial  substitutions  for  Zolpidem 
(Sanger  et  al.  1987).  Similar  to  benzo- 
diazepines, the  BZ1  selective  agonists 
Zolpidem,  zaleplon  (CL  284,846),  and 
SX  3228  produce  only  partial  substi- 
tution in  rats  trained  to  discriminate  1.0 
g/kg  ethanol  (Sanger  1997).  These 
data  suggest  that  activity  only  at  BZ1 
receptors  is  insufficient  to  produce  an 
ethanol-like  effect.  Although  these 
data  fit  well  with  ethanol  potentiating 
activity  at  both  BZ1  and  BZ2  receptor 
subtypes,  they  are  in  contrast  to  the  in 
vitro  data  of  Criswell  and  colleagues 
(1995),  who  suggested  that  selective 
BZ1  activity  predicts  sensitivity  to 
ethanol.  It  should  be  noted  that  the  dis- 
criminative stimulus  effects  of  ethanol 
are  not  antagonized  by  flumazenil 
(Hiltunen  and  Jarbe  1986).  These 
data  suggest  that  ethanol  does  not 
interact  directly  with  the  benzodi- 
azepine binding  site. 

Functional  antagonism  of  ethanol- 
enhanced  activity  at  GABAA  receptors 
using  negative  modulators  of  CI"  flux 
active  at  the  receptor  (inverse  agonists) 
has  been  extensively  characterized 


221 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


with  the  partial  inverse  agonist  Ro  15- 
4513  (see  Grant  and  Lovinger  1995). 
Numerous  in  vitro  assays  demonstrate 
that  Ro  15-4513  blocks  the  effects  of 
ethanol.  In  contrast,  the  benzodiazepine 
inverse  agonists  block  some,  but  not 
all,  of  the  behavioral  effects  of  ethanol. 
In  general,  the  behavioral  effects  of 
ethanol  that  are  resistant  to  the  effects 
of  benzodiazepine  inverse  agonists 
often  are  associated  with  higher  ethanol 
doses.  The  mechanisms  underlying  the 
specificity  of  Ro  15-4513  in  attenuating 
some  of  ethanofs  actions  are  unknown. 
Since  Ro  15-4513  has  intrinsic  activity 
at  the  GABAA  receptor  and  has  anxio- 
genic  and  proconvulsant  activity,  it 
has  been  suggested  that  the  ability  to 
block  some  of  the  behavioral  effects  of 
ethanol  is  due  to  an  additive  interac- 
tion rather  than  a  pharmacological 
antagonism  at  a  single  receptor  mech- 
anism (Mihic  and  Harris  1996). 

There  are  mixed  reports  about  the 
efficacy  of  Ro  15-4513  to  block  the 
discriminative  stimulus  effects  of 
ethanol.  The  discriminative  stimulus 
effects  of  1.0  g/kg  ethanol  were 
antagonized  in  mice  and  rats.  However, 
other  studies  using  rats  have  failed  to 
report  any  substantial  antagonism  of  Ro 
15-4513  on  the  discriminative  stimulus 
effects  of  ethanol.  Gatto  and  Grant 
(1997)  reported  a  wide  range  of  sensi- 
tivities to  the  ethanol-attenuating  effects 
of  Ro  15-4513,  and  the  potency  of  Ro 
15-4513  to  block  ethanol's  discrimina- 
tive stimulus  effects  generally  decreased 
as  the  substitution  dose  of  ethanol 
increased.  Overall,  it  appears  that  the 
ethanol-blocking  effects  of  Ro  15-4513 
are  surmountable  with  higher  doses  of 
ethanol,  indicating  a  competitive  antag- 


onism. Current  studies  using  cynomolgus 
monkeys  trained  to  discriminate  ethanol 
are  replicating  these  general  findings, 
showing  a  blockade  of  the  cUscriminative 
stimulus  effects  of  1 .0  g/kg  ethanol  but 
not  2.0  g/kg  ethanol  (Grant  unpub- 
lished data). 

An  intriguing  hypothesis  to  account 
for  the  attenuation  of  ethanol's  actions  is 
the  presence  of  benzodiazepine-insensitive 
GABAA  receptors  that  contain  a  binding 
site  for  Ro  15-4513  on  a6  subunits 
(Grobin  et  al.  1998).  However,  it  is 
clear  that  Ro  15-4513  has  activity  at 
benzodiazepine -sensitive  receptors  and 
can  block  the  discriminative  stimulus 
effects  of  benzodiazepines  (Hiltunen 
and  Jarbe  1988;  Rees  and  Balster 
1988;  Hiltunen  and  Jarbe  1989).  In 
addition,  the  in  vitro  data  showing  that 
a6  subunits  correlate  with  ethanol  sen- 
sitivity of  chronic  effects  are  not  com- 
pelling (Grobin  et  al.  1998). 
Alternative  to  a  GABAA  hypothesis, 
overcoming  the  attenuating  effects  of 
Ro  15-4513  in  an  ethanol  discrimina- 
tion with  increased  doses  of  ethanol 
may  be  due  to  other  neural  targets 
(i.e.,  NMDA  and/or  serotonin  recep- 
tors) that  serve  as  the  basis  of  ethanol 
discrimination.  Thus,  by  increasing  the 
dose  of  ethanol,  discriminative  stimulus 
effects  of  ethanol  not  mediated  by  the 
GABAA  receptor  can  serve  as  the  basis 
for  the  discrimination.  Since  rats  can 
be  trained  to  discriminate  ethanol  from 
pentobarbital  using  a  two-choice  or  a 
three-choice  method  (Bowen  et  al. 
1997),  there  is  evidence  that  these 
other  receptor  systems  can  play  an 
important  role  in  ethanol  discrimina- 
tion. One  of  these  systems  is  the 
NMDA  receptor  system. 


222 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


Shortly  after  the  in  vitro  biochemical 
and  electrophysiological  findings  that 
ethanol  attenuated  NMDA  receptor- 
mediated  ion  flux,  the  NMDA  channel 
blockers  ketamine,  phencyclidine 
(PCP),  and  dizocilpine,  or  (  +  )MK- 
801,  were  shown  to  substitute  for 
ethanol  in  mice  and  pigeons  (Grant  et 
al.  1991).  This  work  was  quickly 
extended  to  rats,  and  NMDA  channel 
blockers  have  been  shown  to  substitute 
for  ethanol  in  a  number  of  laboratories 
(Gauvin  et  al.  1994;  Grant  1994; 
Harrison  et  al.  1998;  Hundt  et  al. 
1998).  A  recent  study  found  that  five 
channel  blockers — dizocilpine, 
memantine,  PCP,  (  +  )  pentazocine, 
and  (  +  )NAMN  ( TV-allynormeta- 
zocine) — substituted  for  a  1.0  g/kg 
(ip)  training  dose  of  ethanol  in  rats 
(Hundt  et  al.  1998).  This  study  also 
found  no  substitution  of  the  sigma- 
opioid  selective  antagonists  rimcazole 
and  FH  510,  indicating  that  the  sub- 
stitution of  the  NMDA  channel 
blockers  was  due  to  their  activity  at 
the  NMDA  channel  and  not  the 
sigma  site,  where  they  also  bind.  The 
substitution  of  competitive  NMDA 
antagonists  for  ethanol  is  less  reliable, 
with  one  study  showing  full  substitution 
of  the  competitive  antagonist  CGS 
19755  and  other  studies  showing  par- 
tial substitution  of  the  competitive 
antagonists  CGS  19755,  NPC  17742, 
and  CPPene  (cited  in  Hundt  et  al. 
1998).  No  study  has  found  substantial 
substitution  of  glycine  site  antagonists 
for  ethanol,  including  L-701,324, 
MRZ-2/504,  ACEA  1021  (cited  in 
Hundt  et  al.  1998),  and  (+)  HA-966 
(Grant  unpublished  data).  Finally,  the 
polyamine  site  antagonists  eliprodil, 


arcaine,  and  spermidine  all  failed  to 
produce  even  partial  ethanol-like 
responding  (cited  in  Hundt  et  al. 
1998).  In  addition,  the  ct-amino-3- 
hydroxy-5-methyl-4-isoxazole  propi- 
onic acid  (AMPA)  antagonist  GYKI 
52466  did  not  substitute  for  ethanol, 
suggesting  that  the  NMDA  antagonism 
effects  of  ethanol  are  more  prominent 
than  the  AMPA  antagonist  effects. 
However,  these  results  need  to  be 
interpreted  cautiously,  because  only  a 
single  AMPA  antagonist  has  been 
tested  in  an  ethanol  discrimination. 

The  composite  picture  from  the 
NMDA  complex  ligands  suggests  that 
ethanoPs  discriminative  stimulus 
effects  that  are  mediated  by  NMDA 
antagonism  are  generated  by  channel 
blockade  and  not  interaction  with  the 
NMDA,  glycine,  or  polyamine  sites. 
The  molecular  composition  of  the 
NMDA  channel  complex  is  still  being 
characterized,  but  there  is  now  con- 
siderable evidence  that  the  subunit 
composition  confers  pharmacological 
sensitivity.  The  competitive  NMDA 
antagonists  are  more  potent  at 
NR1/NR2A  heteromeric  receptors, 
the  glycine  site  antagonists  are  more 
potent  at  NR1/NR2C  heteromeric 
receptors,  the  polyamine  site  antago- 
nists are  more  potent  at  NR1/NR2B 
heteromeric  receptors,  and  the  chan- 
nel blockers  and  ethanol  are  equipo- 
tent  at  NR1/NR2A  and  NR1/NR2B 
combinations  (Sucher  et  al.  1996). 
Interestingly,  detoxified  alcoholics 
report  that  the  subjective  effects  of 
ketamine  are  similar  to  the  effects  of 
high  doses  of  ethanol  (Krystal  et  al. 
1998).  A  recent  study  by  Hodge  and 
Cox  (1998)  found  that  dizocilpine, 


223 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


but  not  the  competitive  antagonist 
CPP,  administered  directly  into  the 
core  of  the  nucleus  accumbens  or  the 
CA1  region  of  the  hippocampus  was 
sufficient  to  produce  ethanol-like 
effects  in  rats.  In  the  same  study, 
dizocilpine  administration  into  the 
amygdala  or  the  prelimbic  cortex  did 
not  engender  ethanol  substitution. 

GABAA  receptors  may  be  present  on 
a  majority  of  CNS  neurons,  and  there  is 
considerable  evidence  that  GABAergic 
activity  can  regulate  glutamatergic, 
particularly  NMDA-mediated,  neuro- 
transmission (Lovinger  1993).  Specifi- 
cally, when  GABAergic  transmission  is 
decreased,  neuronal  depolarization  is 
more  likely  to  occur,  releasing  the 
Mg++  blockage  of  the  NMDA  receptor 
and  leading  to  increased  functional 
activity  of  the  NMDA  channel.  Con- 
versely, GABAA  receptor  activation 
decreases  the  probability  of  the  NMDA 
receptor  being  activated  due  to  the 
greater  Mg++  blockage  of  the  NMDA 
channel  at  more  hyperpolarized 
potentials.  In  addition,  the  inhibitory 
postsynaptic  potentials  mediated  by 
GABAergic  transmission  and  the  excita- 
tory postsynaptic  potentials  have  a  similar 
time  course  (Lovinger  1993).  Thus,  it 
is  believed  that  NMDA-mediated  glu- 
tamatergic transmission  is  normally 
regulated  by  GABAergic  transmission. 
The  evidence  to  suggest  a  functional 
interaction  of  ethanoPs  effects  at  GABAA 
and  NMDA  receptors  largely  comes 
from  studies  aimed  at  characterizing 
the  hyperexcitability  following  chronic 
ethanol  exposure  (see  Grant  and 
Lovinger  1995).  Nevertheless,  the  acute 
action  of  ethanol  to  potentiate  the 
effects  of  GABAA  activity  may  result  in 


an  enhancement  of  NMDA  channel 
attenuation,  in  addition  to  ethanol's 
direct  actions  in  antagonizing  the 
NMDA  channel.  This  hypothesis  pre- 
dicts a  greater  effect  of  ethanol  on 
inhibition  of  localized  neuronal  activity 
than  the  effects  of  either  a  GABAA 
positive  modulator  or  an  NMDA 
antagonist  alone.  It  appears  reasonable 
to  suggest  that  the  interaction  of 
ethanol's  simultaneous  effects  at  GABAA 
and  NMDA  channels  is  an  important 
aspect  of  ethanol's  stimulus  effects. 

Several  laboratories  have  begun  to 
address  the  simultaneous  activity  of 
ethanol  at  NMDA  and  GABAA  receptor 
systems  in  producing  discriminative 
stimulus  effects.  One  demonstration 
administered  combinations  of  dizo- 
cilpine or  CPP  and  muscimol  in  specific 
brain  regions.  Concentrations  of  mus- 
cimol and  dizocilpine  (or  CPP)  that  did 
not  engender  ethanol  substitution  when 
administered  separately  into  the  nucleus 
accumbens  produced  robust  ethanol  sub- 
stitution in  this  demonstration  (Hodge 
and  Cox  1998).  Using  a  different 
strategy,  a  combination  of  diazepam 
and  ketamine  was  used  as  the  training 
condition,  and  ethanol  produced  sub- 
stitution for  the  mixture  (Harrison  et 
al.  1998).  In  ethanol-trained  rats,  this 
combination  of  ketamine  and  diazepam 
substituted  for  ethanol.  This  is  a 
demonstration  of  cross -generalization 
between  ethanol  and  an  NMDA 
antagonist/GABAA  positive  modulator 
mixture.  Given  the  overwhelming  data 
showing  asymmetrical  generalizations, 
the  data  suggest  that  co-occurrence  of 
GABA^NMDA  activity  is  an  impor- 
tant aspect  of  the  receptor  mediation 
of  ethanol's  discriminative  stimulus 


224 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


effects.  Finally,  in  humans  the  benzo- 
diazepine lorazepam  can  attenuate 
ketamine-induced  emotional  distress 
and  perceptual  alterations  while  exacer- 
bating the  sedative,  attention-impairing, 
and  amnestic  effects  of  ketamine  and 
having  no  effect  on  the  subjective  report 
of  high  (Krystal  et  al.  1998).  Notably, 
subjects  described  the  high  as  ethanol- 
like  ("like  when  I'm  drinking,  but 
hazier  than  when  I'm  drinking"). 

Studies  have  shown  that  5-HT1B 
receptors  are  up-regulated  following 
chronic  ethanol  exposure  and  are  lower 
in  the  CNS  of  rats  bred  to  prefer  10 
percent  ethanol  (Grant  and  Lovinger 
1995).  In  drug  discrimination  studies, 
it  was  initially  reported  that  the  rela- 
tively nonselective  5-HT\  receptor 
agonist  m-trifluoromethylphenylpiper- 
azine  (TFMPP)  substituted  for  the 
discriminative  stimulus  effects  of  ethanol 
in  rats  (Signs  and  Schechter  1988). 
Subsequent  studies  replicated  this  find- 
ing in  rats  trained  to  discriminate  1.0 
and  1.5  g/kg  ethanol  but  not  in  rats 
trained  to  discriminate  2.0  g/kg  ethanol 
(Grant  and  Colombo  1993;  Green  and 
Grant  1998).  The  finding  that  a  5-HT 
receptor  agonist  was  similar  to  the  dis- 
criminative effects  of  ethanol  is  intrigu- 
ing because  another  5-HT  agonist, 
w-chlorophenylpiperazine  (mCPP), 
has  been  reported  to  produce  ethanol- 
like  subjective  effects  and  alcohol  crav- 
ing in  recently  detoxified  alcoholics 
(Benkelfat  et  al.  1991;  Krystal  et  al. 
1994;  Buydens-Branchey  et  al.  1997). 

5-HT1B  receptors  are  negatively 
coupled  to  cyclic  adenosine  monophos- 
phate through  G:  proteins  and  act  as 
presynaptic  autoreceptors  and  hetero- 
receptors  to  decrease  neurotransmitter 


release.  There  is  accumulating  evidence 
that  5-HT1B  receptors  are  important 
in  the  control  of  striatal  dopaminergic 
release  and  that  5-HT1B  agonists 
increase  striatal  dopamine  levels.  These 
effects  are  consistent  with  the  ability  of 
5-HT1B  agonists  to  increase  locomotor 
activity,  disrupt  prepulse  inhibition, 
and  substitute  for,  or  enhance,  the 
discriminative  stimulus  effects  of 
cocaine.  The  interaction  between  5- 
HT1B  receptors  and  dopaminergic 
release  in  the  striatum  is  not  clear,  but 
one  hypothesis  is  that  5-HT1B  receptors 
function  as  heteroreceptors  on  inter- 
neurons.  Data  gathered  with  drug  dis- 
crimination suggest  a  functional  link 
between  the  actions  of  ethanol  at 
GABAA  receptors  and  5-HT1B  media- 
tion of  ethanol-like  activity  (Green 
and  Grant  1998). 

There  are  only  a  few  investigations 
of  5-HT2  receptor  ligands  and  the  dis- 
criminative stimulus  effects  of  ethanol. 
These  studies  have  shown  that  the  5- 
HT2a  antagonists  ketanserin  in 
pigeons  (Grant  and  Barrett  1991)  and 
cinanserin  in  rats  (Winter  1977)  do  not 
block  the  discriminative  stimulus 
effects  of  ethanol.  Likewise,  the  5- 
HT2A  agonist  5-MeODMT  (5- 
methoxy-N,Ar-dimethyltryptamine) 
does  not  appreciably  substitute  for 
ethanol  in  rats  (Signs  and  Schechter 
1988).  In  contrast,  the  5-HT  transport 
inhibitor  fluoxetine  produces  substitu- 
tion in  an  ethanol  discrimination,  and 
this  substitution  can  be  selectively 
blocked  by  the  5-HT2A  antagonist 
MDL  100,907  (Maurel  et  al.  1997). 
These  results  suggest  that  the  5-HT2A 
receptor  mediates  ethanol-like  dis- 
criminative stimulus  effects.  However, 


225 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


a  recent  study  found  the  5-HT2a/2c 
agonist  DOI  did  not  engender  or 
potentiate  the  discriminative  stimulus 
effects  of  ethanol  (Szeliga  and  Grant 
1998).  Likewise,  the  5 -HT2A  antago- 
nist ketanserin  failed  to  attenuate  the 
discriminative  stimulus  effects  of 
ethanol  (Szeliga  and  Grant  1998). 
The  data  collectively  suggest  that  the 
5-HT2  receptor  ligands  DOI  and 
ketanserin  interact  with  receptors  that 
are  not  substantially  involved  in  medi- 
ating the  discriminative  stimulus 
effects  of  ethanol  associated  with 
approximately  40-120  mg/dL  blood 
ethanol  concentrations. 

To  date,  only  a  limited  number  of 
studies  have  investigated  the 
pharmacological  basis  of  ethanoPs  dis- 
criminative stimulus  effects  in  selectively 
bred  ethanol-preferring  (P)  and  ethanol  - 
nonpreferring  (NP)  rats.  Cholinergic, 
serotonergic,  and  GABAergic  receptor 
systems  are  preliminarily  implicated  in 
mediating  differences  in  the  stimulus 
effects  of  rats  differentially  bred  to 
prefer  ethanol.  Data  implicating  the 
nicotinic  cholinergic  system  is  derived 
from  nicotine's  partial  substitution  for 
1.0  g/kg  ethanol  in  P  rats,  with  no 
substitution  in  NP  rats  (Gordon  et  al. 
1993).  However,  ascribing  receptor 
mechanisms  based  on  partial  substitu- 
tion in  drug  discrimination  procedures 
is  controversial  (see  Colpaert  1986). 
The  serotonergic  system  is  implicated 
by  the  substitution  of  the  hallucinogenic 
amphetamine  MDMA  (3,4-methyl- 
enedioxymethamphetamine),  a  com- 
pound whose  discriminative  stimulus 
effects  are  believed  to  be  primarily 
serotonergic  in  nature  (Schechter 
1989).  In  an  ethanol  discrimination 


using  a  training  dose  of  0.6  g/kg 
ethanol,  MDMA  substituted  in  HAD 
but  not  in  LAD  rats,  suggesting  a 
greater  relevance  of  serotonergic  acti- 
vation in  the  effects  of  ethanol  in  the 
preferred  line  (Meehan  et  al.  1995). 
Finally,  GABAergic  system  differences 
in  rat  lines  selected  for  ethanol  prefer- 
ence are  suggested  by  data  showing  a 
greater  sensitivity  to  the  ethanol-like 
effects  of  pentobarbital  HAD  rats 
compared  with  LAD  rats,  as  measured 
by  lower  ED50  value  for  pentobarbital 
substitution  (Krimmer  1991).  How- 
ever, line  differences  were  only  noted 
between  HAD  and  LAD  rats  trained 
to  discriminate  the  stimulus  effects  of 
0.75  g/kg  ethanol  30  minutes  after 
injection.  There  were  no  line  differ- 
ences in  rats  trained  to  discriminate 
0.75  g/kg  ethanol  2  minutes  postin- 
fection (Krimmer  1992).  Overall, 
there  is  a  scarcity  of  data  available  to 
address  the  basis  for  ethanol's  discrim- 
inative stimulus  effects  in  rat  lines 
selected  for  ethanol  preference. 

There  are  a  host  of  other  receptor 
systems  that  have  been  only  superfi- 
cially addressed  in  terms  of  discrimina- 
tive stimulus  effects.  Most  notable  are 
the  opiate,  dopaminergic,  and  voltage- 
gated  calcium  channel  (VGCC)  systems. 
In  general,  the  opiate  agonist  morphine 
does  not  produce  ethanol-like  discrim- 
inative stimulus  effects.  However,  nalox- 
one has  been  reported  to  block  the 
discriminative  stimulus  effects  of  ethanol 
associated  with  the  rising  phase  of  the 
blood  ethanol  curve  (Spanagel  1996). 
Only  partial  antagonism  by  naloxone  (50 
percent  ethanol- appropriate  responding) 
was  found  with  a  1.0  g/kg  (ip)  ethanol 
training  dose.  In  the  same  study,  delta 


226 


Behavioral  Effects  and  Underlying  Neurocircui tries  of  Alcohol 


and  kappa  opioid  antagonists  had  no 
effect  (Spanagel  1996).  Dopaminergic 
agonists  do  not  substitute  and  dopa- 
minergic antagonists  do  not  block  the 
discriminative  stimulus  effects  of  ethanol. 
However,  apomorphine  and  ampheta- 
mine potentiate  the  discriminative 
stimulus  effects  of  ethanol  (Schechter 
1985).  Similar  findings  have  been 
reported  with  nicotine,  where  substi- 
tution is  not  found  but  potentiation 
of  ethanol's  stimulus  effects  is  evident. 
Finally,  VGCC  antagonists  have  been 
reported  to  partially  substitute  (De  Beun 
et  al.  1996),  to  have  no  effect  (Schechter 
1994),  or  to  block  (Colombo  et  al. 
1994)  the  discriminative  stimulus 
effects  of  ethanol.  Recent  data  have 
shown  that  VGCC  agonists  can  antag- 
onize and  VGCC  antagonists  can 
potentiate  ethanol's  discriminative 
stimulus  effects  (Green  and  Grant 
unpublished  data).  These  findings 
illustrate  an  important  strategy  in  drug 
discrimination:  investigating  the  modu- 
latory effects  of  candidate  receptor  sys- 
tems on  discriminative  stimulus  effects. 
The  drug  discrimination  procedure 
is  one  technique  where  the  multiple 
stimulus  effects  of  ethanol  have  been 
extensively  studied  and  can  be  readily 
compared.  An  important  issue  in 
using  the  discriminative  stimulus 
effects  of  ethanol  to  help  identify  can- 
didate receptor  systems  to  explore  in 
other  behavioral  paradigms  is  the 
importance  of  dose.  The  training  dose 
of  a  drug  that  acts  with  a  high  degree 
of  specificity  at  a  single  receptor  system 
may  be  analogous  to  stimulus  intensity. 
In  contrast,  the  evidence  reviewed 
above  clearly  shows  that  ethanol  has 
interactions  at  multiple  receptors. 


Thus,  the  training  dose  of  ethanol 
may  determine  both  the  intensity  and 
the  qualitative  effects  of  the  drug 
stimulus.  That  is,  ethanol's  functional 
activity  at  the  various  receptor  systems 
may  not  amplify  in  equal  proportion 
as  the  dose  of  ethanol  is  increased, 
resulting  in  ethanol  having  qualita- 
tively different  discriminative  stimulus 
effects  at  different  doses.  Ethanol  has 
a  well-known  biphasic  profile,  with 
low  doses  resulting  in  activation  and 
high  doses  resulting  in  sedation.  Some 
of  these  biphasic  effects  may  reflect 
differential  sensitivity  of  the  receptor- 
linked  ionophores  to  a  given  dose  of 
ethanol.  The  relative  contribution  of 
receptor  systems  to  the  qualitative 
effects  of  ethanol  as  a  function  of  dose 
can  be  addressed  by  examining  the 
discriminative  stimulus  effects  of  dif- 
ferent ethanol  training  doses. 

In  summary,  drug  discrimination 
procedures  can  be  used  in  alcohol 
research  to  provide  candidate  receptor 
systems  and  receptor  mechanisms  to 
target  for  altering  the  behavioral  effects 
of  ethanol.  For  example,  in  vitro  data 
suggest  that  ethanol  interacts  with 
GABAA  receptors  to  increase  chloride 
flux.  However,  there  are  several  sites 
on  this  receptor  system  that  interact 
with  ligands  to  alter  chloride  flux. 
Drug  discrimination  procedures  can 
be  used  to  differentiate  these  sites  on 
a  behavioral  level.  By  acting  as  a  dis- 
criminative stimulus,  factual  evidence  is 
provided  that  these  effects  of  the  drug 
can  be  perceived  and  control  behavior. 
Thus,  the  discriminative  stimulus 
effects  of  a  drug  provide  information  on 
the  realm  of  possible  stimulus  effects 
that  can  serve  other  functions,  for 


227 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


example,  reinforcing  stimulus  effects. 
After  identifying  the  possible  receptor 
effects  that  can  act  as  stimuli,  addi- 
tional studies  must  characterize  the 
role  of  particular  receptor  mechanisms 
in  mediating  other  stimulus  effects  of 
ethanol.  Although  these  additional 
stimulus  effects  of  ethanol  can  be  (and 
have  been)  studied  without  first  being 
characterized  in  discrimination  studies, 
it  is  not  always  possible  to  separate 
ethanol -specific  from  task- specific  recep- 
tor mechanisms  underlying  behavior. 
Drug  discrimination  studies  are  also 
providing  hypotheses  of  simultaneous, 
but  independent,  receptor  activity  by 
ethanol  that  may  alter  in  prominence  as 
the  dose  of  ethanol  changes.  Thus,  the 
candidate  receptor  systems  that  may 
mediate  the  behavioral  effects  associ- 
ated with  specific  doses  of  ethanol  can 
be  identified  and  then  tested  within 
more  appropriate  paradigms. 

Anxiolytic  and  Anxiogenic 
Stimulus  Effects 

It  has  been  hypothesized  for  many  years 
that  the  ability  of  alcohol  to  reduce 
stress  underlies  its  ability  to  serve  as  a 
reinforcer  (Williams  1966;  Pohorecky 
1981).  Indeed,  accumulating  clinical 
evidence  indicates  a  high  degree  of 
co-occurrence  of  anxiety  and  alcohol 
dependence  (see  Langenbucher  and 
Nathan  1990;  Crum  et  al  1995).  Two 
mechanisms  are  likely  responsible  for 
this  comorbidity,  both  involving  condi- 
tioning. First,  excessive  alcohol  intakes 
may  be  due  to  the  anxiolytic  properties 
of  this  drug  alleviating  a  constant 
"basal"  state  of  anxiety  in  some  individ- 
uals. Second,  abstinence  subsequent  to 
excessive  and  prolonged  consumption 


of  alcoholic  beverages  gives  rise  to 
dysphoric  effects,  including  anxiety, 
that  may  be  relieved  by  alcohol  con- 
sumption. Evidence  for  an  association 
of  alcohol's  anxiolytic  effects  with  the 
use  of  alcohol  as  a  basis  for  promoting 
future  alcohol  consumption  is  derived 
from  both  human  and  animal  studies. 
When  social  drinkers  believe  that  they 
have  consumed  alcohol,  imbibing  a 
nonalcoholic  drink  results  in  decreased 
levels  of  anxiety  (Abrams  and  Wilson 
1979).  Thus,  humans  can  associate 
the  consumption  of  alcohol  with  its 
anxiolytic  effects.  However,  caution  is 
necessary  in  extrapolating  from  the 
anxiolytic  effects  of  alcohol  and  sub- 
sequent reinforcing  effects  of  alcohol 
in  humans.  First,  an  important  consid- 
eration is  the  generally  weak  anxiolytic 
effects  of  alcohol  noted  in  humans.  A 
possible  exception  may  be  people  with 
signs  of  anxiety  disorder  who  report 
anxiolytic  effects  of  alcohol  (Chutuape 
and  deWit  1995).  However,  these 
individuals  did  not  choose  to  drink 
alcohol,  again  suggesting  a  dichotomy 
between  the  weak  anxiolytic  effects 
and  the  reinforcing  effects  of  alcohol. 
In  animal  models,  anxiety  has  been 
associated  with  increases  in  ethanol  self- 
administration  and  consumption.  For 
example,  early  stressful  rearing  condi- 
tions and  social  separations  are  associ- 
ated with  behavioral  and  physiological 
reactions  associated  with  stress,  and 
these  conditions  are  sufficient  to  initi- 
ate and  maintain  ethanol  consump- 
tion (Kraemer  and  McKinney  1985; 
Blanchard  et  al.  1987;  Higley  et  al 
1991).  Consistent  with  these  findings, 
higher  innate  levels  of  anxiety  have 
also  been  detected  in  selectively  bred, 


228 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


ethanol-preferring  P  (Stewart  et  al. 
1993),  SP  (Colombo  et  al.  1995), 
and  HARF  (Le  1996)  rats,  compared 
with  ethaiiol-nonpreferring  NP,  SNP, 
and  LARF  rats,  respectively.  Thus, 
genetic  models  appear  to  have  both 
increased  baseline  levels  of  anxiety  and 
increased  ethanol  consumption,  suggest- 
ing a  genetic  basis  for  a  functional  link 
between  the  anxiolytic  effects  of  ethanol 
and  increased  ethanol  consumption. 
This  hypothetical  association  is  similar 
to  findings  that  alcohol  attenuates  stress 
reactions  in  young  adults  at  risk  for 
alcohol  dependence  (Sher  and  Leven- 
son  1982). 

The  evidence  for  anxiolytic  effects 
of  ethanol  in  animal  models  has  been 
reviewed  (Pohorecky  1990).  Condition- 
ing procedures  frequently  use  conflict, 
in  which  responding  maintained  by 
food  or  water  presentation  in  a  deprived 
animal  is  occasionally  punished,  usually 
by  the  delivery  of  shock  (Pohorecky 
1981).  These  procedures  result  in  a  sup- 
pression of  behavior  that  can  be  rein- 
stated with  typical  anxiolytics  such  as 
benzodiazepines.  These  effects  of 
ethanol  are  fairly  robust,  although 
ethanol  is  somewhat  less  efficacious  as 
an  anxiolytic  compared  with  benzodi- 
azepines. The  anxiolytic  effects  of 
ethanol  occur  at  low  to  moderate 
dosages  and  are  not  always  separable 
from  other  behavioral  effects  of 
ethanol  (Koob  and  Britton  1996). 

The  receptor  mechanisms  involved 
in  the  anxiolytic  effects  of  ethanol 
have  investigated  primarily  the  GABAA 
and  5-HT  receptor  systems.  The  sci- 
entific literature  is  replete  with  evidence 
showing  the  involvement  of  GABAA 
receptor  systems  in  regulating  anxiety. 


As  a  class  of  compounds,  benzodi- 
azepines that  act  as  positive  modulators 
of  GABAA  receptors  produce  the  most 
robust  anxiolytic  effects  in  experimental 
models  (e.g.,  Pellow  et  al.  1985;  Jones 
et  al.  1994;  Rex  et  al.  1996)  and  con- 
stitute the  first-line  therapy  for  the 
treatment  of  anxiety  in  general  practice 
(e.g.,  Ballinger  1990;  Ashton  1994). 
Recent  attention  has  been  focused  on 
the  interactions  of  neurosteroids  with 
the  GABAA  receptor  complex.  Similar 
to  the  benzodiazepines,  neuroactive 
steroids  that  positively  modulate 
GABAA  receptors  produce  anxiolytic 
activity  (Crawley  et  al.  1986;  Britan  et  al. 
1991;  Wieland  et  al.  1991;  Fernandez- 
Guasti  and  Picazo  1995).  The  intrigu- 
ing aspect  of  the  neurosteroids  is  that 
these  are  endogenous  compounds, 
synthesized  within  the  CNS  as  well  as 
being  derived  from  adrenal  and  gonadal 
steroids.  Furthermore,  endogenous 
levels  of  neurosteroids  are  associated 
with  stressful  events  (see  Paul  and 
Purdy  1992).  Anticonflict  effects  of 
ethanol  can  be  blocked  by  GABAA 
ligands  to  the  convulsant  site  and 
GABAA  inverse  agonists,  but  not  by 
benzodiazepine  site  antagonists  (Koob 
and  Britton  1996).  Interestingly, 
naloxone  has  been  reported  to  reverse 
the  anticonflict  effects  of  ethanol  and 
benzodiazepines  (Koob  and  Britton 
1996).  Corticotropin-releasing  factor 
also  reverses  the  anticonflict  effect  of 
ethanol,  although  it  has  proconflict 
activity  so  the  effect  may  not  be  spe- 
cific to  ethanol.  CRF  appears  to  inter- 
act through  extrahypothalamic  sites 
within  the  mesolimbic  dopaminergic 
system,  notably  the  amygdala,  in 
response  to  stressful  events,  including 


229 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol  withdrawal  (Pich  et  al.  1995). 
Only  a  few  studies  have  used  site-specific 
injections  to  investigate  neural  cir- 
cuitry of  conflict  behavior.  These 
studies  suggest  that  the  basolateral 
and  central  nuclei  of  the  amygdala, 
areas  that  are  rich  in  GABAA  recep- 
tors, are  important  in  the  mediation 
of  anticonflict  activity  by  benzodi- 
azepines and  barbiturates. 

Over  the  last  two  decades,  the  poten- 
tial role  of  serotonin  in  the  modulation 
of  emotional  states,  including  anxiety, 
has  been  addressed  by  several  animal 
studies  (reviewed  in  Barrett  and 
Vanover  1993;  Griebel  1995).  It  has 
been  proposed  that  a  reduction  in 
central  serotonin  results  in  anxiolysis 
(Griebel  1995).  Several  studies  have 
focused  on  the  involvement  of  the  5- 
HT1A  subtype  of  the  serotonin  receptor 
in  modulating  anxiety  (Lucki  1992; 
Barrett  and  Vanover  1993;  Barrett  et 
al.  1994;  Griebel  1995),  although 
inconsistent  results  have  been  reported 
(Dawson  and  Tricklebank  1995).  The 
anxiolytic  effects  of  5-HT1A  receptor 
agonists  have  been  suggested  to  be 
related  to  activation  of  5-HT1A  autore- 
ceptors,  which  results  in  a  reduced 
serotonin  neuronal  function  (Dourish 
et  al.  1986).  Specifically,  5-HT1A soma- 
todendritic autoreceptors  in  the  raphe 
nuclei  result  in  a  reduction  in  5-HT 
neuronal  firing  rate  and  subsequent 
decreases  in  5-HT  release  in  terminal 
areas  of  the  limbic  system,  such  as  the 
hippocampus.  Postsynaptic  5-HT1A 
receptor  activation  in  the  amygdala  and 
dorsal  hippocampus  has  been  reported 
to  be  anxiogenic,  suggesting  that  an 
overall  reduction  in  5-HT  neurotrans- 
mission to  the  dorsal  hippocampus  is 


anxiolytic  (File  et  al.  1996).  Direct 
administration  of  5-HT1A  agonist  8- 
OH-DPAT  into  the  raphe  nucleus 
produces  anxiolytic  activity  (see  File  et 
al.  1996),  conditioned  place  preference 
(Fletcher  et  al.  1994),  and  selective 
increases  in  ethanol  consumption 
(Tomkins  et  al.  1994#).  Interestingly, 
when  given  peripherally,  very  low 
doses  of  8-OH-DPAT  (30-60 
^xg/kg)  increase  ethanol  consumption 
(Tomkins  et  al.  1994&),  whereas 
higher  doses  of  8-OH-DPAT,  as  well 
as  buspirone  and  ipsapirone,  signifi- 
cantly reduce  voluntary  ethanol  intake 
in  rats,  mice,  and  monkeys  (DeVry 
1995).  The  reduction  in  ethanol 
intake  following  peripheral  injections 
of  higher  doses  of  5-HT1A  agonists 
possibly  reflects  anxiogenic  activity  at 
postsynaptic  5-HT1A  receptors  (File  et 
al.  1996).  In  a  social  confrontation 
procedure  using  consecutive  "phases" 
of  increasing  threat,  ethanol  was  simi- 
lar to  benzodiazepines  and  the  5-HTi 
agonist  gepirone  in  decreasing  signs 
of  anxiety  associated  with  future 
antagonistic  encounters  (e.g.,  tachy- 
cardia, hyperthermia,  vocalizations) 
(Tornatzky  and  Miczek  1995).  Addi- 
tionally, buspirone  has  been  success- 
fully tested  in  clinical  trials  of  anxiety 
associated  with  abstinence  from  alco- 
hol in  alcoholics  (Litten  et  al.  1996). 

Pentylenetetrazol  (PTZ)  binds  to  the 
picrotoxin  site  of  the  GABAA  receptor 
complex,  resulting  in  a  reduction  of 
the  chloride  ion  flux,  an  increase  in 
CNS  excitability,  and  convulsions  at 
high  doses  (Simmonds  1982).  PTZ 
increases  the  subjective  reports  of  anxiety 
in  humans  (Rodin  1958)  and  has  been 
reported  to  be  anxiogenic  in  laboratory 


230 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


animals  in  several  experimental  ap- 
proaches (e.g.,  Buczek  et  al.  1994; 
Rodgers  et  al.  1995).  The  discriminative 
stimulus  effects  produced  by  PTZ  have 
been  promoted  as  an  animal  model  of 
anxiety  (Lai  and  Emmett-Oglesby 
1983).  Features  of  this  discrimination 
that  address  the  validity  of  an  animal 
model  of  anxiety  include  the  genera- 
tion of  PTZ- appropriate  responding 
following  (a)  exposure  to  stressful 
exteroceptive  stimuli,  such  as  a  novel 
environment  (Carey  et  al.  1990),  a  cage 
intruder  (Carey  et  al.  1990),  and  a 
predator-prey  interaction  (Gauvin  and 
Holloway  1991);  (b)  withdrawal  from 
chronic  benzodiazepines  (Emmett- 
Oglesby  et  al.  1990)  and  ethanol  (Lai 
et  al.  1988);  and  (c)  administration  of 
anxiogenic  drugs,  such  as  FG  7142 
(Leidenheimer  and  Schechter  1990), 
Ro  5-3663,  Ro  15-4513,  and  (3-car- 
bolines  (Emmett-Oglesby  et  al. 
1990).  Furthermore,  anxiolytic  com- 
pounds, mainly  those  acting  at  the 
GABAA  receptor,  have  been  reported  to 
completely  block  the  interoceptive  cues 
of  PTZ  (Andrews  et  al.  1989).  Ethanol 
is  effective  in  blocking  a  training  dose 
of  10  mg/kg  PTZ,  indicating  that  the 
anxiolytic  effects  of  ethanol  are  assess- 
able in  a  PTZ  discrimination  (Emmett- 
Oglesby  et  al.  1990). 

In  addition  to  serving  as  an  animal 
model  of  anxiety,  PTZ  discriminations 
have  also  been  used  to  assess  anxio- 
genic aspects  of  ethanol  withdrawal 
(Lai  and  Emmett-Oglesby  1983;  Lai 
et  al.  1988;  Emmett-Oglesby  et  al. 
1990).  It  is  important  to  note  that 
acute  ethanol  withdrawal,  several 
hours  following  administration  of 
moderate  to  large  (2-4  g/kg)  doses 


of  ethanol,  resulted  in  PTZ  generaliza- 
tion (Gauvin  et  al.  1989,  1992).  An 
important  process  in  the  high  ethanol 
intakes  may  be  the  onset  of  with- 
drawal- or  hangover- associated  anxiety 
several  hours  following  the  consumption 
of  similar  doses  of  ethanol,  initiating 
further  consumption  of  ethanol.  PTZ 
discriminations  also  show  considerable 
promise  for  studying  the  anxiety  asso- 
ciated with  alcohol  withdrawal  (Lai  et 
al.  1988;  Emmett-Oglesby  et  al.  1990). 
Following  termination  of  chronic 
ethanol  treatment  rats  respond  on  the 
PTZ-appropriate  lever,  indicating 
withdrawal  has  effects  similar  to  the 
PTZ  stimulus.  This  state  lasts  from  12 
to  48  hours,  at  which  time  the  per- 
centages of  rats  choosing  the  PTZ  lever 
are  80  and  30,  respectively.  The  length 
of  chronic  ethanol  treatment  necessary 
to  show  this  withdrawal  effect  is 
apparently  3  days  at  12.5  g/kg/d 
(Emmett-Oglesby  et  al.  1990).  How- 
ever, in  a  more  recent  analysis  of  the 
ethanol  withdrawal  state,  acute  admin- 
istration of  moderate  to  large  (2-4 
g/kg)  doses  of  ethanol  resulted  in  PTZ 
generalization  (Gauvin  et  al.  1989, 
1992).  Thus,  PTZ  discrimination 
appears  to  be  sensitive  to  ethanol  with- 
drawal effects  following  acute  (i.e., 
hangover)  or  chronic  (i.e.,  withdrawal) 
ethanol  treatment.  It  may  be  possible  to 
use  this  model  to  quantify  and  char- 
acterize the  severity  and  time  course 
of  interoceptive  stimuli  associated 
with  ethanol  withdrawal. 

The  neurocircuitry  involved  in  con- 
ditioned anxiolytic  effects  of  fear- 
potentiated  startle  responses  has  been 
extensively  studied  (Davis  1992).  In  this 
procedure,  the  startle  response  to  an 


231 


NIAAA's  Ncuroscience  and  Behavioral  Research  Portfolio 


acoustic  stimulus  is  augmented  by 
presenting  the  eliciting  acoustic  stimulus 
with  a  cue  that  has  previously  been 
paired  with  shock.  The  conditioned 
fear  is  defined  by  the  increase  in  startle 
amplitude  in  the  presence  of  the  cue 
previously  paired  with  shock  compared 
with  the  startle  amplitude  in  the 
absence  of  the  cue.  This  procedure  can 
be  used  in  both  humans  and  laboratory 
animals  (Krystal  et  al.  1997).  In 
humans,  startle  amplitude  has  been 
used  to  assess  posttraumatic  stress  dis- 
order and  sensory  gating  in  schizo- 
phrenia. Ethanol  attenuates  startle  in 
rodents  and  humans.  Benzodiazepines 
reduce  fear-potentiated  startle,  as  do 
5-HTj  agonists  and  morphine.  On 
the  other  hand,  drugs  that  increase 
reports  of  anxiety  in  humans,  namely 
yohimbine  and  (3-carbolines,  increase 
this  response  in  rats. 

An  important  finding  with  this  pro- 
cedure is  the  synergistic  effect  of  5- 
HTj  agonists  and  D}  receptor 
antagonists.  The  D2  receptor  antagonist 
raclopride  also  decreases  potentiated 
startle  at  doses  that  do  not  alter  baseline 
startle  levels.  It  is  known  that  dopa- 
minergic tone  in  the  mesolimbic  and 
mesocortical  pathways  increases  follow- 
ing stressful  events,  and  the  effect  of 
dopamine  antagonists  suggests  that 
one  effect  of  this  increase  in  dopamine 
is  the  potentiation  of  response  to  fearful 
events.  Dopaminergic  tone  in  these 
pathways  has  also  been  suggested  to 
potentiate  response  to  pleasurable 
stimuli,  and  has  led  to  speculation  that 
ventral  tegmental  area  (VTA) 
dopaminergic  projections  are  not  sig- 
naling pleasure  but  accentuating  con- 
ditioned responses  to  significant 


stimuli  (Salamone  1994;  Wickelgren 
1997).  In  potentiated  startle,  informa- 
tion from  the  conditioned  stimulus 
(light  cue)  and  information  from  the 
unconditioned  stimulus  (shock)  con- 
verge in  the  lateral  and  basolateral 
amygdala.  After  pairing  with  shock, 
the  conditioned  stimulus  can  activate 
the  lateral  and  basal  amygdala,  which 
projects  to  the  central  amygdala.  Acti- 
vation of  the  central  amygdala  then 
facilitates  startle  through  the  reticu- 
laris pontis  caudalis,  which  mediates 
the  startle  response  from  acoustic 
information  through  the  lateral  lem- 
niscus to  the  spinal  cord  neurons 
(Davis  1992). 

There  are  several  compelling  hypo- 
theses that  have  yet  to  be  addressed 
with  ethanol.  Namely,  local  infusion  of 
NMDA  antagonists  into  the  accum- 
bens  attenuates  the  acquisition  of  fear- 
potentiated  startle,  suggesting  that 
this  may  also  be  an  action  of  ethanol, 
an  outcome  that  could  be  extrapolated 
to  increases  in  risk-taking  behaviors  due 
to  decreased  conditioned  fear.  In  this 
light  it  is  interesting  that  the  amygdala 
is  one  area  where  NMDA  antagonist 
application  produced  ethanol-like 
stimulus  effects  (Hodge  and  Cox 
1998).  Postconditioning  anticonfiict 
effects  in  punished  responding  can  be 
shown  by  direct  infusion  of  benzodi- 
azepines, GABA,  or  muscimol  into 
the  amygdala,  as  discussed  earlier  in 
this  section.  Thus,  it  is  possible  that 
ethanol  could  act  in  the  amygdala  to 
inhibit  learning  about  fearful  stimuli 
through  NMDA  antagonism  and  also 
to  inhibit  response  to  fearful  stimuli 
even  after  it  has  been  learned.  In  addi- 
tion to  NMDA  and  GABAA,  it  has 


232 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


been  estimated  that  25  percent  of  the 
neurons  in  the  central  amygdaloid 
nucleus  contain  CRF,  somatostatin, 
and  neurotensin,  as  well  as  5-HT3 
receptors  (Davis  1992).  Thus,  this 
pathway  and  the  potentiated  startle 
response  would  appear  to  be  an 
important  area  to  characterize  for 
alcohol  responsiveness  in  future 
behavioral  studies. 

In  summary,  ethanol  appears  to 
have  attenuating  effects  on  condi- 
tioned responses  associated  with  anxi- 
ety. A  few  studies  have  addressed  the 
correlation  between  anxiolytic 
responses  and  ethanol  consumption. 
Other  studies  have  investigated  the 
neurotransmitter  systems  involved  in 
the  anxiolytic  responses,  primarily  the 
GABAA  and  5-HT  systems.  One  area 
of  study  that  appears  promising  is  the 
potentiated  startle  response,  because 
the  response  can  be  studied  in  both 
humans  and  laboratory  animals  and 
because  the  neurocircuitry  has  been 
established.  Notably,  in  the  startle 
procedures,  NMDA  antagonists 
decrease  fear  responding,  allowing  the 
incorporation  of  this  neurotransmitter 
system  into  hypotheses  of  ethanol's 
activity  on  conditioned  anxiolytic 
effects.  Other  neurotransmitter  recep- 
tors, such  as  5-HT3  and  neuropep- 
tides, that  have  previously  been 
implicated  in  other  behavioral  effects 
of  ethanol  have  yet  to  be  extensively 
studied  in  mediating  the  acute  anxi- 
olytic effects  of  ethanol. 

Place/Taste  Conditioning  Effects 

Ethanol  reinforcement  is  a  conditioned 
process  that  is  pivotal  in  animal  models 
of  alcohol  addiction.  Two  behaviors, 


found  under  different  experimental 
conditions,  are  representative  of  ethanol 
reinforcement.  These  behaviors  are 
ethanol  self- administration  and  ethanol- 
conditioned  preferences.  Other  responses 
may  reflect  the  ability  of  ethanol  to  serve 
as  a  reinforcer.  For  example,  effects 
such  as  anxiolytic,  aversive,  discrimi- 
native, motor,  or  amnestic  effects  each 
address  important  aspects  of  ethanol's 
ability  to  serve  as  a  reinforcer.  How- 
ever, these  behavioral  outcomes  are 
not  direct  measures  of  ethanol  rein- 
forcement. Likewise,  the  effects  of 
ethanol  on  the  threshold  for  intracra- 
nial self-stimulation  are  believed  to 
reflect  the  reinforcing  effects  of 
ethanol  (Wise  et  al.  1992);  however, 
this  is  also  an  indirect  measure  of 
ethanol  reinforcement. 

Conditioned  preference  refers  to  the 
process  of  a  laboratory  animal  becom- 
ing attracted  to  the  place  associated 
with  the  delivery  of  ethanol  (Mucha 
et  al.  1982;  Hoffman  1989).  The  ani- 
mal is  given  a  specific  dose  of  ethanol 
and  placed  in  a  distinct  environment. 
Through  the  association  of  ethanol's 
effects  with  the  specific  environmental 
stimuli,  these  environmental  stimuli 
come  to  serve  as  incentive  stimuli  and 
elicit  approach  behaviors.  When  given 
a  choice  between  two  environments, 
only  one  of  which  is  associated  with 
the  effects  of  ethanol,  a  preference  is 
apparent  if  the  animal  spends  a  greater 
proportion  of  time  in  the  environment 
associated  with  ethanol.  An  aversion 
would  be  apparent  if  a  smaller  propor- 
tion of  time  was  spent  in  the  environ- 
ment associated  with  ethanol.  These 
procedures  emphasize  environmental 
stimuli  as  generating  motivational 


233 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


states,  reflected  in  approach  or  avoid- 
ance behaviors  that  are  indicative  of 
the  reinforcing  effects  of  ethanol. 

These  procedures  also  provide 
important  alternatives  to  oral  self- 
administration  paradigms  for  studying 
the  motivational  effects  of  ethanol. 
The  strengths  of  the  preference  proce- 
dures include  the  ability  to  detect  both 
positive  and  negative  motivational 
effects,  the  ability  to  test  for  motiva- 
tional impact  in  the  absence  of 
ethanol's  direct  sensory-motor  effects, 
the  ability  to  assess  ethanol  dose 
effects  in  the  absence  of  confounding 
influence  of  taste/palatability  factors, 
the  ability  to  assess  potential  pharma- 
cotherapies without  the  need  to  assess 
nonspecific  effects  on  ingestive  behav- 
ior, the  ability  to  separately  assess 
manipulations  that  influence  acquisition 
versus  expression  of  ethanol-induced 
motivational  effects,  and  the  ability  to 
implement  procedures  without 
surgery  or  lengthy  periods  of  training. 

Both  place  and  taste  preference 
procedures  serve  as  models  for  studying 
mechanisms  underlying  the  acquisition 
and  extinction  of  associations  between 
taste  or  other  environmental  cues  and 
ethanol.  The  understanding  of  these 
mechanisms  is  important  because  con- 
ditioned learning  involving  exterocep- 
tive stimuli  is  thought  to  contribute  to 
craving  and  relapse  to  ethanol-seeking 
behavior  after  withdrawal  and  long 
periods  of  abstinence.  For  example, 
Schuster  and  Woods  (1968)  found  that 
response-contingent  presentation  of 
stimuli  previously  associated  with 
morphine  self- administration  increased 
responding  under  extinction  condi- 
tions. Similarly,  rats  withdrawn  from 


morphine  drank  more  vehicle  when 
placed  in  environments  where  mor- 
phine self-administration  had  been 
acquired  than  did  rats  placed  in  envi- 
ronments not  associated  with  mor- 
phine self- administration  (Thompson 
and  Ostland  1965;  Hinson  et  al.  1986). 
Finally,  noncontingent  amphetamine 
given  to  monkeys  under  extinction 
conditions  reinstated  responding  pre- 
viously maintained  by  amphetamine 
only  if  a  masking  noise,  present  dur- 
ing the  self-administration  sessions, 
was  also  present  (Stretch  et  al.  1971). 
Thus,  contact  with  the  drug  may  not 
be  sufficient  to  elicit  drug-seeking 
behavior  outside  an  environment  in 
which  the  drug  was  normally  taken.  A 
related  area  of  research  is  the  modu- 
lating role  of  tolerance  or  sensitization 
in  the  ability  of  conditioned  stimuli  to 
affect  motivation  for  and  self- adminis- 
tration of  ethanol. 

An  overwhelming  amount  of  evidence 
shows  that  place  conditioning  and  taste 
conditioning  are  sensitive  to  ethanol 
dose,  number  of  trials,  trial  duration 
(place  conditioning),  the  temporal  rela- 
tionship between  the  paired  stimulus 
and  ethanol,  and  environmental  condi- 
tions such  as  ambient  temperature 
(Bormann  and  Cunningham  1997; 
Cunningham  et  al.  1997;  Bormann 
and  Cunningham  1998;  Dickinson  and 
Cunningham  1998).  Thus,  the  condi- 
tioning aspects  of  the  tasks  reflect  a 
learning  process.  Motor  activation  can 
also  be  measured  within  the  context  of 
place  preference  procedures  (Cunning- 
ham and  Noble  1992;  Risinger  et  al. 
1992#,  1992&).  A  study  investigating 
both  the  stimulant  and  rewarding 
effects  of  ethanol  in  a  place  preference 


234 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


procedure  reported  blockade  of  the 
motor- activating  effects  of  ethanol  with 
a  dopaminergic  antagonist  without 
blocking  the  expression  of  place  pref- 
erence (Risinger  et  al.  1992#).  In  addi- 
tion, the  locomotor  effects  of  2.0  g/kg 
ethanol  were  enhanced  by  fluoxetine 
pretreatment  within  the  preference 
apparatus;  however,  there  was  no  effect 
on  place  preference  (Risinger  1997). 
Thus,  motor-activating  effects  of 
ethanol  are  neither  necessary  nor  suffi- 
cient for  the  development  of  ethanol 
reinforcement. 

The  neurochemical  basis  of  these 
conditioned  effects  has  been  examined 
with  genetic  and  pharmacological 
tools.  Genetic  differences  in  ethanol's 
rewarding  and  aversive  effects  and 
genetic  correlations  with  other  ethanol 
effects  have  been  studied  using  inbred 
lines  and  selectively  bred  lines  of  mice 
and  rats  (e.g.,  Froehlich  et  al.  1988; 
Cunningham  et  al.  1991;  Krimmer 
1991;  Crabbe  et  al.  1992;  Cunningham 
et  al.  1992;  Krimmer  1992;  Cunning- 
ham 1995;  Risinger  and  Cunningham 
1995;  Broadbent  et  al.  1996;  Risinger 
et  al.  1996;  Stewart  et  al.  1996; 
Chester  et  al.  1998).  For  example, 
inbred  strains  (C57BL/6J)  or  selec- 
tively bred  lines  (COLD,  FAST)  that 
drink  elevated  amounts  of  ethanol  show 
reduced  sensitivity  to  an  ethanol- 
induced  taste  aversion  (Cunningham 
et  al.  1991;  Risinger  et  al.  1994).  In 
addition,  place  conditioning  and  taste 
conditioning  have  been  used  success- 
fully to  identify  several  provisional 
QTL  that  may  contain  genes  influenc- 
ing ethanol's  rewarding  and  aversive 
motivational  effects  (Cunningham 
1995).  Finally,  the  5-HT1B  knockout 


mouse  has  been  assessed  for  develop- 
ment of  ethanol  place  and  taste  condi- 
tioning (Risinger  et  al.  1996)  because 
these  5-HT1B  deficient  mice  drink  sig- 
nificantly larger  amounts  of  ethanol 
compared  with  wild-type  mice  (Crabbe 
et  al.  1996).  The  knockout  mice  were 
more  sensitive  to  ethanol-induced 
conditioned  place  preference,  but 
equally  sensitive  to  ethanol-induced 
taste  aversion.  These  results  show  that 
taste  aversion  and  place  preference  are 
separable  and  that  oral  ethanol  con- 
sumption correlated  with  place  prefer- 
ence but  not  taste  aversion  outcomes. 

Pharmacological  investigation  of 
ethanol-induced  conditioned  place 
preference  has  examined  opioid,  5-HT, 
GABAA,  and  dopamine  involvements. 
Specifically,  the  5-HT2  antagonist 
mianserin  enhances  (Risinger  and 
Oakes  1996)  but  the  5-HT  reuptake 
blocker  fluoxetine  has  no  effect 
(Risinger  1997)  on  ethanol-induced 
conditioned  place  preference.  However, 
fluoxetine  enhances  ethanol-induced 
taste  aversion  (Risinger  1997).  Inter- 
estingly, fluoxetine  has  been  reported 
to  enhance  (Risinger  1997)  or  substi- 
tute (Maurel  et  al.  1997)  for  the  dis- 
criminative stimulus  effects  of  ethanol. 
Neither  the  inverse  agonist  Ro  15-4513 
nor  the  dopaminergic  antagonist 
haloperidol  inhibits  ethanol-induced 
place  preference  (Cunningham  et  al. 
1992;  Risinger  et  al.  1992^,  1992&). 
However,  additional  GABAA  studies 
using  a  wider  range  of  ligands  at  each 
receptor  system  are  clearly  needed. 

Pharmacological  studies  of  ethanol's 
place  preference  using  the  opiate  antag- 
onist naloxone  have  clearly  differentiated 
the  acquisition  from  the  expression  of 


235 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol-induced  conditioning  (e.g., 
Cunningham  et  al.  1995;  Broadbent 
et  al.  1996;  Risinger  and  Oakes  1996; 
Risinger  1997;  Cunningham  et  al. 
1998).  Of  particular  interest  are  studies 
showing  that  pretreatment  with  the 
opiate  antagonist  naloxone  at  the  time  of 
testing  facilitates  extinction  of  ethanol- 
induced  conditioned  place  preference, 
but  retards  extinction  of  conditioned 
place  aversion  (Cunningham  et  al. 
1995,  1998).  These  outcomes  suggest 
that  the  opioid  system  maintains  the 
learned  association  between  appetitive 
events  and  responses,  while  also  retard- 
ing the  maintenance  of  learned  associa- 
tion of  aversive  events.  Thus,  naloxone 
appears  to  have  a  detrimental  impact 
on  conditioned  rewarding  effects  of 
ethanol  (weakening  approach  behav- 
iors), while  enhancing  a  conditioned 
aversive  effect  of  ethanol  (enhancing 
avoidance  behaviors)  (Cunningham  et 
al.  1998). 

In  addition  to  measuring  preferences 
associated  with  ethanol  administration, 
conditioned  place/taste  procedures  can 
assess  the  aversive  effects  of  ethanol. 
The  dose  range  for  the  aversive  effects 
of  ethanol  are  1.0-2.0  g/kg  for  rats 
(Davies  and  Parker  1990;  Gauvin  and 
Holloway  1992;  Holloway  et  al. 
1992;  Schechter  1992;  Schechter  and 
Krimmer  1992)  and  2.0-3.0  g/kg  for 
mice  (Cunningham  et  al.  1991; 
Risinger  and  Cunningham  1992). 
Selectively  bred  NP  rats,  but  not  P 
rats,  develop  conditioned  taste  aversion 
associated  with  1.0  g/kg  ethanol 
(Froehlich  et  al.  1988).  However,  at 
higher  doses  of  ethanol,  P  rats  also 
show  conditioned  taste  aversions 
(Froehlich  et  al.  1988).  Using  place 


preference  procedures,  rather  than 
conditioned  taste  procedures,  both  the 
P  and  NP  selected  lines  developed  place 
aversions  following  1.0  g/kg  ethanol 
(Schechter  1992).  Where  conditioned 
preferences  are  found  using  rats, 
repeated  exposure  to  ethanol  is  neces- 
sary (Grant  et  al.  1990).  The  necessity 
of  multiple  exposures  to  ethanol 
implies  that  tolerance  to  the  aversive 
effects  may  expose  the  positive  rein- 
forcing effects  of  ethanol.  However,  it 
is  worth  noting  that  at  least  some 
strains  and  lines  of  mice  do  not  show 
the  initial  aversive  effects  of  ethanol 
when  tested  in  place  preference  para- 
digms. Both  DBA/2J  mice  and  mice 
selected  for  hyperthermia  in  response 
to  ethanol  develop  conditioned  place 
preferences  associated  with  2-A  g/kg 
ethanol  (Cunningham  et  al.  1991, 
1992;  Risinger  et  al.  1992&).  Future 
studies  will  need  to  continue  to  delin- 
eate the  experimental  determinants  of 
ethanol-induced  conditioned  prefer- 
ence and  conditioned  aversion  and 
why  ethanol  induces  conditioned  pref- 
erences in  some  circumstances  but 
conditioned  aversion  in  others. 

In  summary,  conditioned  place  and 
taste  preference  are  reliable  procedures 
that  allow  the  investigation  of  receptor 
mechanisms  mediating  either  the  posi- 
tive or  negative  hedonic  effects  of 
ethanol.  These  procedures  are  particu- 
larly useful  in  studying  the  condi- 
tioned effects  of  ethanol  in  mice.  Thus, 
they  appear  well  suited  to  characterize 
the  genetic  bases  of  ethanol-related 
effects.  Indeed,  several  provisional 
QTL  have  been  identified  for  place 
preference  by  typing  recombinant 
inbred  strains.  In  addition,  knockout 


236 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


mice  have  been  characterized  in  pref- 
erence procedures  and  have  generated 
hypotheses  concerning  a  common 
genetic  basis  for  ethanol's  effects  across 
selected  lines  of  mice.  These  proce- 
dures appear  to  provide  an  important 
window  into  the  genetic  basis  of 
ethanol-related  effects.  Combined  with 
procedures  such  as  drug  discrimina- 
tion, specific  receptor  mechanisms  can 
be  identified  and  verified  as  important 
in  mediating  ethanol-associated 
behaviors  in  the  mouse  and  provide 
potential  mechanisms  for  study  in 
other  species.  To  date,  there  is  no 
information  available  on  the  specific 
circuitry  involved  in  conditioned  taste 
or  place  preferences. 

Self- Administration 
and  Reinforcing  Effects 

Self- administration  refers  to  the  process 
of  a  laboratory  animal  or  human  engag- 
ing in  a  behavior  that  results  in  the 
administration  of  alcohol.  The  most 
common  self- administration  procedure 
involves  the  presentation  of  alcohol 
following  a  set  number  of  specific 
responses  in  a  distinct  environment. 
The  presentation  of  alcohol  in  self- 
administration  procedures  has  been 
accomplished  by  several  routes, 
including  oral,  intravenous,  intragastric, 
and  intracranial  delivery  (see  Carroll 
et  al.  1990).  In  short,  self- administra- 
tion procedures  emphasize  the  conse- 
quences of  behavior  in  the  role  of 
alcohol  seeking. 

While  it  is  clear  that  alcohol  has 
reinforcing  effects  that  maintain  the 
consumption  of  alcoholic  beverages, 
ethanol  is  not  an  efficacious  reinforcer 
to  drug-naive  laboratory  animals 


(Meisch  1977;  Grant  et  al.  1990).  In 
a  vast  majority  of  the  studies  of  oral 
ethanol  self- administration  in  labora- 
tory animals,  simply  allowing  access  to 
ethanol  is  not  sufficient  to  result  in 
repeated  consumption  of  intoxicating 
quantities.  The  low  levels  of  ethanol 
intake  of  uninitiated  animals  have  been 
attributed  to  the  taste  of  ethanol,  the 
delay  between  the  consumption  of 
ethanol  and  its  pharmacological 
effects,  the  volume  of  ethanol  needed 
for  a  pharmacological  effect,  and  the 
particular  pharmacological  effects  of 
ethanol  (including  positively  reinforc- 
ing and  aversive  effects).  To  circumvent 
these  difficulties,  it  is  now  standard  to 
use  an  induction  procedure  to  estab- 
lish ethanol  drinking  in  animals  that 
have  not  been  specifically  bred  to  drink 
large  amounts  of  an  ethanol  solution. 
Induction  procedures  include  food 
deprivation,  adulterating  the  taste  of 
ethanol,  associating  the  consumption 
of  ethanol  with  the  presentation  or 
removal  of  other  reinforcers,  acclimating 
the  animal  to  gradually  increasing  con- 
centrations of  ethanol,  and  restricting 
access  to  the  ethanol  solution  (see 
Meisch  1977;  Samson  1987). 

Although  a  wide  variety  of  species 
have  been  studied  in  ethanol  self- 
administration  procedures  (see  Caroll 
et  al.  1990),  most  of  the  extensive 
investigations  of  the  neural  basis  of 
ethanol  self- administration  have  been 
in  rats.  Species  differences  have  not 
been  extensively  studied  with  self- 
administration  procedures,  primarily 
because  the  rat  has  been  the  overwhelm- 
ing animal  of  choice.  Monkeys  were 
initially  investigated  in  the  1970s  using 
oral,  intravenous,  and  intragastric 


237 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


procedures.  However,  not  all  primate 
species  are  alike  in  a  predisposition  to 
drink  ethanol.  Similar  to  rodents,  female 
vervet  monkeys  {Cercopithecus  aethiops) 
drink  more  ethanol  than  males  (Juarez 
et  al.  1993).  In  contrast,  female  rhesus 
monkeys  {Macaco  mulatto)  are  less 
likely  to  initiate  and  maintain  ethanol 
consumption  compared  with  males 
(Grant  and  Johanson  1988). 
Although  monkeys  can  acquire  ethanol 
self- administration  when  investigators 
provide  access  without  an  induction 
procedure  (Macenski  and  Meisch  1992; 
Stewart  et  al.  1996)  the  average  intakes 
are  low  (0.2-1.0  g/kg/3  h).  When 
induction  procedures  or  food  depriva- 
tion is  imposed,  ethanol  intakes  increase 
to  more  than  1.0  g/kg/h  (Carroll  et 
al.  1990;  Macenski  and  Meisch  1992; 
Williams  et  al.  1998).  Preliminary 
results  following  a  schedule  induction 
procedure  to  establish  ethanol  self- 
administration  and  then  16  h/d  access 
to  4  percent  (w/v)  ethanol  and  water 
show  that  cynomolgus  monkeys  drank 
1-4  g/kg  ethanol  per  day  and  developed 
signs  of  fatty  liver  after  6  months  of 
drinking  these  quantities  (Grant  et  al. 
1998).  Interestingly,  the  female  mon- 
keys were  in  the  lower  50th  percentile 
of  ethanol  intake  and  still  developed 
signs  of  fatty  liver.  These  results  suggest 
that  increasing  the  access  to  16  h/d 
can  result  in  excessive  intakes  with 
biomedical  consequences. 

Some  induction  procedures  use 
stressful  conditions.  In  one  study,  mon- 
keys were  either  continuously  housed  in 
individual  cages  or  housed  socially  and 
subjected  to  social  disruption  (Krae- 
mer  and  McKinney  1985).  Monkeys 
in  the  socially  disrupted  group  were 


intermittently  separated  from  and 
reunited  with  their  cage  mates  for  1- 
week  periods.  Overall,  the  socially  dis- 
rupted monkeys  drank  more  ethanol 
than  the  individually  caged  monkeys 
and  drank  more  ethanol  when  they 
were  isolated  than  when  they  were 
socially  housed.  The  authors  interpreted 
these  findings  as  evidence  that  the  stress 
of  intermittent  social  isolation  pro- 
moted ethanol  consumption  (Kraemer 
and  McKinney  1985).  Social  stress 
was  also  invoked  as  an  explanation  of 
induced  drinking  in  socially  subordi- 
nate male  rats  housed  in  social  groups 
(Blanchard  et  al.  1987).  Subordinate 
social  status  is  thought  to  be  stressful 
in  mammals  because  subordinates  of 
many  species  receive  more  aggression, 
spend  more  time  alone,  and  hyper- 
secrete  stress  hormone  relative  to  their 
dominant  counterparts  (Shively  et  al. 
1986,  1990).  However,  in  the  only 
available  study  of  social  status  effects 
on  ethanol  consumption  in  macaques, 
dominant  monkeys  drank  more  than 
subordinates  (Crowley  et  al.  1990). 
The  authors'  explanation  was  that  only 
one  source  of  ethanol  was  available  to 
all  social  group  members,  and  domi- 
nants exercised  their  status  by  control- 
ling access  to  the  drinking  station. 
With  access  to  the  ethanol  source 
controlled  by  dominants,  investigators 
could  not  detect  social  stress  effects 
on  ethanol  consumption  among  sub- 
ordinates (Crowley  et  al.  1990). 

An  important  aspect  of  self- 
administration  procedures  is  the  ability 
to  use  schedules  of  reinforcement  that 
maintain  drug- seeking  behavior  in  the 
absence  of  drug  delivery.  These  are 
termed  "complex  schedules,"  and 


238 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


they  have  been  noticeably  absent  from 
the  characterization  of  ethanol  self- 
administration.  With  such  schedules 
(chain  schedules,  second  order  sched- 
ules), in  which  responses  are  reinforced 
with  the  presentation  of  conditioned 
stimuli  associated  with  the  presentation 
of  ethanol,  but  not  directly  with 
ethanol,  the  neurocircuitry  involved  in 
conditioned  reinforcement  can  be 
addressed  in  self- administration  proce- 
dures. A  series  of  studies  have  shown 
that  limbic  innervation  of  the  nucleus 
accumbens  from  the  hippocampus 
and  basolateral  amygdala  is  essential 
to  behavior  controlled  by  drug-associ- 
ated stimuli  (Hitchcott  and  Phillips 
1997;  Hitchcott  et  al.  1997a,  1997*). 
These  authors  suggest  that  the  basolat- 
eral projection  to  the  nucleus  accumbens 
determines  the  degree  of  discriminative 
control  over  conditioned  responses, 
while  hippocampal/subiculum  input 
determines  the  efficacy  of  the  condi- 
tioned stimulus.  The  input  to  the 
accumbens  from  the  subiculum  is  glu- 
tamatergic,  and  from  the  amygdala  is 
dopaminergic,  possibly  D3  (Hitchcott 
et  al.  1997  a).  These  data  fit  well  with 
the  data  from  Samson  and  Hodge 
(1996)  showing  that  intra-accumbens 
injections  of  dopaminergic  agonists 
increase  ethanol  self- administration, 
possibly  through  increasing  the  discrim- 
inative control  exerted  over  behavior  by 
drug-associated  stimuli.  However,  these 
data  also  have  important  implications 
for  the  hippocampus,  amygdala,  and 
nucleus  accumbens  as  target  sites  when 
studying  the  discriminative  stimulus 
effects  of  ethanol.  Enhanced  discrimi- 
nation with  glutamatergic,  GABAergic, 
or  dopaminergic  compounds  may  affect 


discriminated  responses  without  con- 
veying information  concerning  ethanol- 
specific  activity. 

An  emerging  principle  in  studying 
the  neurochemical  basis  of  self- admin- 
istration is  that  the  ability  of  ethanol 
to  function  as  a  positive  reinforcer  is 
not  due  to  an  immutable  neurochem- 
istry  that  mediates  positive  affect.  Ani- 
mal models  involving  several  drug 
classes  support  the  dissociation  between 
drug  presentation  (and  presumably 
pharmacological  action  in  the  CNS), 
positive  affect,  and  subsequent  self- 
administration.  For  example,  several 
studies  have  shown  that  the  responding 
of  morphine-dependent  monkeys  can 
be  maintained  by  the  administration 
of  naloxone  (Goldberg  et  al.  1971; 
Woods  et  al.  1975;  Goldberg  et  al. 
1978)  at  doses  that  produce  avoidance 
responding  in  the  same  (Goldberg  et 
al.  1971)  or  other  (Kandel  and  Schuster 
1977)  morphine-dependent  monkeys. 
Furthermore,  within-subject  designs 
have  shown  that  the  same  doses  of 
cocaine  can  be  either  actively  avoided 
or  self- administered  (Spealman  1979). 
Thus,  any  other  intrinsic  effect  of  a 
drug — for  example,  dopamine  release 
in  the  nucleus  accumbens — is  likely 
only  to  influence,  rather  than  determine, 
drug  seeking.  Alcohol's  ability  to  serve 
as  a  reinforcer  at  any  given  time  will  be 
due  to  a  combination  of  antecedent 
events,  current  environmental  contin- 
gencies, and  the  pharmacological  basis 
of  alcohol's  stimulus  effects.  Clearly, 
alcohol-seeking  behavior  can  be  viewed 
as  malleable  and  not  wholly  determined 
by  intrinsic  pharmacological  effects. 

An  additional  finding  with  self- 
administration  procedures  is  that  the 


239 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


neurochemical  effects  of  the  drug  are 
determined,  in  part,  by  the  act  of  self- 
administration.  That  is,  the  neuro- 
chemical effects  of  self-administered 
drugs  are  different  from  the  neuro- 
chemical effects  of  the  same  doses  when 
administered  irrespective  of  ongoing 
behavior  (e.g.,  yoked  administrations). 
The  interaction  of  self- administration 
behavior  and  neurochemical  conse- 
quences of  drugs  has  parallels  in  the 
tolerance  literature.  According  to  toler- 
ance models,  the  initial  effects  of  a  drug 
or  the  environment  in  self- administration 
can  serve  as  conditioned  stimuli  to 
compensate  for  effects  of  the  drug. 
Indeed,  conditioned  drug  effects  and 
conditioned  stimuli  altering  drug 
effects  have  now  become  central  tenets 
in  drug  abuse  research.  Investigating 
the  neurocircuitry  of  ethanol  within 
a  self-administration  paradigm  is 
central  to  our  understanding  of 
ethanol's  effects  that  mediate  alcohol 
abuse  and  alcoholism. 

The  neurochemical  basis  of  the 
reinforcing  effects  of  ethanol  within 
self-administration  or  consumption 
procedures  has  been  studied  for 
decades  and  has  filled  volumes  of  sci- 
entific literature.  Practically  every 
neurotransmitter,  neuromodulator,  or 
neuropeptide  system  has  been  investi- 
gated for  effects  on  ethanol  intakes. 
Most  of  these  studies  use  receptor  lig- 
ands,  but  there  are  also  genetic,  elec- 
trophysiological, neurochemical,  and 
imaging  techniques,  as  well  as  lesion 
techniques,  that  have  been  applied  to 
the  quest  for  understanding  ethanol's 
reinforcing  effects  within  the  drinking 
context.  However,  making  a  list  of 
these  findings  may  not  be  as  useful  as 


examining  the  functional  role  of  the 
various  neurotransmitter  systems  and 
the  circuitry  in  which  they  reside.  For 
example,  the  mesolimbic  dopamine 
system  has  been  extensively  studied, 
and  we  now  have  evidence  that 
mesolimbic  dopamine  is  released  within 
a  self- administration  context  in  antici- 
pation of  and  response  to  ethanol  con- 
sumption (Weiss  et  al.  1993).  We  also 
know  that  we  can  inject  dopamine 
agonists  and  antagonists  directly  into 
different  sites  of  this  circuit  (VTA, 
accumbens,  amygdala,  frontal  cortex) 
and  alter  ethanol  self- administration 
(see  Samson  and  Hodge  1996). 
Finally,  there  is  evidence  that  ethanol 
injected  directly  into  the  VTA  will 
support  self- administration  (Gatto  et 
al.  1994).  However,  it  is  clear  that  the 
dopamine  pathway  is  involved  in  medi- 
ating reinforced  responses  in  general 
(Salamone  1994;  Hitchcott  et  al. 
1997#,  1997b).  In  short,  activity  in 
this  complex  system  appears  to  be 
related  to  the  more  complex  organiza- 
tion of  appetitive-consummatory  behav- 
ior, in  which  the  actions  of  reinforcing 
stimuli  integrate  with  other  stimuli  to 
direct  and  then  maintain  goal-ori- 
ented behaviors  (Wilner  and  Scheel- 
Kruger  1991).  Therefore,  just  knowing 
that  this  pathway  is  involved  in  ethanol 
reinforcement  is  insufficient  to  under- 
stand the  mechanisms  unique  to 
ethanol  and  potential  targets  for 
ethanol -specific  pharmacotherapies. 

An  example  of  modifying  circuitry 
involved  in  appetitive  behaviors  rather 
than  ethanol-specific  interactions  is  the 
reduction  in  alcohol  self- administration 
in  the  presence  of  opioid  antagonists. 
Activation  of  the  endogenous  opioid 


240 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


system  has  been  postulated  to  mediate 
the  reinforcing  effects  of  low  to  mod- 
erate doses  of  ethanol.  This  hypothesis 
is  supported  by  data  in  rats  showing 
that  mu  and  delta  opioid  antagonists 
selectively  decrease  ethanol  intake 
when  water  is  available  (Froehlich  and 
Li  1993).  Data  from  macaque  mon- 
keys show  that  naltrexone  decreases 
oral  ethanol  consumption  in  a  dose- 
dependent  manner  but  also  has  effects 
on  concurrently  available  water  (Myers 
et  al.  1986;  Kornet  et  al.  1991).  When 
a  sweetened  solution  is  the  alternative 
reinforcer,  mu  and  delta  opioid  antag- 
onists decrease  both  ethanol  and 
sucrose  consumption  in  rats  (Samson 
and  Doyle  1985;  Krishnan-Sarin  et  al. 
1995)  and  monkeys  (Williams  et  al. 
1998).  Thus,  the  opioid  system  most 
likely  does  not  selectively  mediate 
ethanol's  effects.  In  fact,  endogenous 
opioid  peptides  mediate  a  variety  of 
ingestive  behaviors,  and  naloxone  can 
suppress  intake  of  water,  fats,  and 
sucrose  in  rats,  monkeys,  and  humans 
(Brown  and  Holtzman  1981;  Locke 
et  al.  1982;  Krishnan-Sarin  et  al. 
1995).  It  is  important  to  note  that 
naltrexone  decreases  ethanol  intake 
via  the  intravenous  route  in  monkeys 
(Altshuler  et  al.  1980;  Williams  et  al. 
1998)  and  the  intragastric  route  in  rats 
(Sinden  et  al.  1983),  suggesting  that 
the  effect  of  the  opiate  attenuation  is 
through  a  centrally  mediated  pathway. 
There  is  evidence  that  both  alcohol  and 
palatable  substances  increase  endoge- 
nous opioid  peptide  synthesis  and  release 
(Wand  1989;  Gianoulakis  1990;  De 
Waele  et  al.  1992;  Froelich  and  Li 
1993),  and  preferences  for  alcohol  and 
sweet  solutions  are  correlated  in  some 


outbred  rat  strains  (Overstreet  et  al. 
1993)  and  lines  selectively  bred  for 
ethanol  preference  (Sinclair  et  al. 
1992;  Stewart  et  al.  1994).  However, 
it  is  also  clear  that  there  is  not  a  com- 
plete correspondence  between  sweet 
preference  and  alcohol  preference 
(Phillips  et  al.  1994).  It  appears  that 
the  opiate  system  mediates  information 
about  ingesta,  including  ethanol's 
effects.  However,  ethanol  is  not  unique 
in  its  ability  to  activate  this  pathway, 
and  ligands  of  this  pathway  should 
not,  in  turn,  be  expected  to  selectively 
attenuate  the  self- administration  of 
alcohol.  Nevertheless,  naltrexone  has 
some  efficacy  in  preventing  relapse,  and 
recent  animal  studies  have  focused  on 
the  endogenous  opioid  system  in 
mediating  aversive  effects  of  ethanol 
(Cunningham  et  al.  1998;  Froehlich 
etal.  1998). 

Similarly,  many  of  the  neuropeptides 
alter  ethanol  intake  when  administered 
peripherally  or  centrally.  These  include 
CRF  (Bell  et  al.  1998),  cholecystokinin 
(CCK),  and  bombesin  (Kulkosky 
1985),  which  decrease  oral  ethanol 
consumption,  and  neuropeptide  Y, 
which  increases  ethanol  intake 
(Kulkosky  et  al.  1988).  However,  in 
the  same  studies  CRF,  CCK,  and 
bombesin  also  decreased  food  intake, 
whereas  neuropeptide  Y  increased 
food  intake.  Thus,  the  manipulation 
of  ethanol  self- administration  using 
these  peptides  appears  to  be  through 
circuitry  that  regulates  food  intake 
and  consummatory  behaviors  in  gen- 
eral, rather  than  information  about 
ethanol  specifically. 

The  overriding  theme  is  that  "con- 
summatory/regulatory"  systems  are 


241 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


active  when  ethanol  is  consumed. 
Through  conditioning  processes,  the 
unique  pharmacological  effects  of 
ethanol  interact  with  these  regulatory 
systems  to  establish  a  pattern  of  con- 
sumption that  is  regulated  by  both 
ethanol's  stimulus  effects  and  the 
functioning  state  of  the  regulatory  sys- 
tem. If  ethanol  has  deleterious  effects 
on  consummatory  systems,  dysregula- 
tion  will  result.  If,  however,  other  events 
in  the  environment  produce  dysregu- 
lation  in  the  consummatory  system, 
elevated  ethanol  intake  could  be  an 
outcome  (as  could  overeating,  gam- 
bling, and  some  other  behaviors),  which 
in  turn,  can  further  alter  regulatory 
control.  The  challenge  for  behavioral 
neuroscience  is  to  define  the  consum- 
matory/regulatory  systems  and  inte- 
grate mechanisms  of  how  ethanol 
self- administration  is  both  an  antecedent 
and  a  consequence  to  dysregulation. 

Learning,  Memory, 
and  Cognitive  Effects 

As  reviewed  thus  far  the  acute  actions 
of  alcohol  produce  a  constellation  of 
physiological  and  behavioral  effects  in 
humans  and  laboratory  animals.  The 
subset  of  these  actions  that  affect  cog- 
nitive function  can  be  subjective  in 
nature,  such  as  how  a  drink  of  alcohol 
makes  a  person  feel,  or  they  can  be 
objective  in  nature,  such  as  the  effects 
of  alcohol  on  the  recall  of  recently 
learned  material.  Since  the  1940s  lit- 
erally thousands  of  scientific  studies 
have  been  conducted  to  classify  and 
measure  both  the  objective  and  sub- 
jective stimulus  effects  of  alcohol  that 
alter  cognitive  function  and  subse- 
quent reaction  to  alcohol  (see  NIAAA 


1995).  However,  these  studies  have 
largely  been  conducted  in  human  sub- 
jects, and  the  application  of  animal 
models  to  study  the  underlying  neu- 
rocircuitry  has  been  limited. 

The  classification  of  the  cognitive 
effects  of  alcohol  depends  on  the  tasks 
used  to  measure  these  effects.  Most  of 
these  tasks  focus  on  characterizing  three 
broad  areas  of  interactive  processes,  usu- 
ally referred  to  as  information  processing, 
psychomotor  skills,  and  subjective  effects. 
Briefly,  information  processing  involves 
the  ability  to  perceive,  learn,  and  remem- 
ber information.  Psychomotor  skills 
predominantly  include  measures  of 
reaction  time,  proprioceptive  ability  (such 
as  tracking),  and  vigilance  (attending 
to  a  particular  stimulus  when  there  are 
distracting  stimuli  presented).  Subjective 
reactions  to  alcohol  are  most  often  mea- 
sured by  the  perceived  degree  of  intox- 
ication, pleasant  affect,  dysphoria,  or 
sedation,  or  by  other  mood  descriptors. 
Clearly,  the  cognitive  processes  that 
determine  the  outcome  of  these  tasks  are 
not  mutually  exclusive,  and  the  alteration 
of  one  of  these  processes  can  change 
the  outcome  of  the  others.  Sophisti- 
cated techniques  have  been  developed 
in  research  with  human  subjects  to  sep- 
arate and  quantify  different  aspects  of 
cognitive  ability,  including  measures  of 
overall  mental  ability,  verbal/visuospa- 
tial  learning,  conceptual  learning,  and 
perceptual/motor  abilities.  Human 
studies  have  also  correlated  physiolog- 
ical measures  of  brain  function  with 
the  cognitive  effects  of  alcohol  (e.g., 
electroencephalographic,  hormonal, 
and  functional  imaging  measures). 

Research  in  humans  has  suggested 
areas  for  study  in  animal  models, 


242 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


including  increasing  the  complexity  of 
the  task.  Performance  tasks  that  are 
complex  are  very  sensitive  to  the  effects 
of  alcohol  and  can  be  disrupted  by 
one  to  two  drinks,  with  corresponding 
blood  alcohol  concentrations  in  the 
range  of  40  mg/dL.  In  contrast,  simple 
reaction  time  tasks  may  require  three 
drinks  before  disruption.  Another 
finding  from  the  human  literature  is 
the  relatively  high  threshold  dose  for 
amnestic  effects,  which  usually  occurs 
at  blood  alcohol  levels  that  are  reported 
as  blatantly  intoxicating  (e.g.,  stu- 
porous). This  finding  is  in  contrast  to 
the  benzodiazepines,  which  can  dis- 
rupt memory  at  doses  that  are  not 
subjectively  perceived.  Another  vari- 
ables that  is  known  to  influence  both 
performance  measures  and  subjective 
effects  of  alcohol  is  family  history  of 
alcoholism.  A  family  history  may 
dampen  the  effects  on  performance 
measures  and  enhance  the  subjective 
effects  of  the  rising  phase  of  the  blood 
alcohol  curve. 

Compared  with  human  studies, 
animal  studies  lag  far  behind.  A 
notable  recent  addition  to  the  literature 
is  the  use  of  eyeblink  conditioning  to 
study  the  effects  of  neonatal  ethanol 
exposure  in  rats  (Stanton  and 
Goodlett  1998).  The  eyeblink  condi- 
tioning procedure  uses  classical  condi- 
tioning and  is  mediated  through  an 
identified  and  characterized  circuit 
from  the  brainstem  to  the  cerebellum 
(Harvey  1987).  Cerebellar  damage 
noted  with  neonatal  ethanol  exposure 
was  found  to  have  functional  conse- 
quences. However,  acute  effects  of 
ethanol  in  this  exposure  have  not  been 
explored.  Two  procedures  used  in 


animals,  particularly  nonhuman  pri- 
mates, that  are  sensitive  to  impairments 
produced  by  GABAA  positive  modula- 
tors and  NMDA  antagonists  are  a 
delayed  nonmatching  to  sample  pro- 
cedure (Ogura  and  Aigner  1993)  and 
a  repeated  acquisition  procedure 
(Baron  and  Moerschbaecher  1996). 
However,  ethanol  has  not  been  stud- 
ied in  either  procedure.  One  procedure 
that  has  been  used  in  animals  exposed 
to  ethanol  is  a  spatial  working  memory 
task,  which  is  disrupted  by  0.75  and 
1.0  g/kg  ethanol,  an  effect  that  has 
been  correlated  with  ethanol's  impair- 
ment of  hippocampal  theta  activity 
(Givens  1995).  There  have  also  been 
some  studies  involving  maze  perfor- 
mance and  working  memory  in  mice 
(Melchior  et  al.  1993).  Givens  (1995) 
has  described  a  circuitry  involving 
ethanol-induced  disruption  of  medial 
septal  area  activity  via  a  GABAergic 
mechanism,  thereby  disrupting  septo- 
hippocampal  input  and  interfering 
with  information  processing  in  the 
hippocampal  circuitry.  Recent  work 
has  used  single-neuron  recording 
techniques  to  measure  cognitive 
processes  affected  by  ethanol  (Givens 
etal.  1998). 

Aggression 

Aggression  has  been  studied  in  animal 
models  to  only  a  limited  extent.  There 
are  two  distinct  types  of  aggressive 
behavior,  predatory  and  affective. 
Predatory  aggression  is  interspecific, 
normally  related  to  feeding  and  not 
associated  with  increased  irritability. 
This  type  of  aggression  is  accompa- 
nied by  minimal  vocalization,  stalking 
posture,  and  lethally  directed  attacks 


243 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


(e.g.,  at  the  back  of  the  prey's  neck). 
Affective  agression  is  intraspecific  and 
involves  intense  autonomic  arousal, 
vocalizations,  and  threatening  and 
defensive  postures. 

By  far,  the  receptor  system  most 
widely  implicated  in  both  predatory 
and  affective  aggression  is  serotonin 
(Olivier  et  al.  1990).  Increases  in  preda- 
tory behavior  are  associated  with  low 
serotonin  levels  produced  by  serotonin 
depletion  with  neurotoxins.  Stimula- 
tion of  serotonin  systems  by  electrical 
stimulation  of  dorsal  raphe  nucleus, 
serotonin  precursors,  or  5-HT  reuptake 
blockers  decreases  aggressive  acts.  Affec- 
tive  aggression  has  also  been  closely 
linked  to  serotonergic  function,  but 
the  data  are  not  as  clear  as  with  preda- 
tory aggression.  Nearly  all  pharmaco- 
logical manipulations  that  either 
increase  or  decrease  5-HT  neurotrans- 
mission can  inhibit  offensive  aggres- 
sion (Miczek  et  al.  1989;  Olivier  et  al. 
1990).  These  mixed  results  probably 
reflect  differential  effect  on  neural  cir- 
cuitry, in  that  stimulation  of  hypothal- 
amic serotonergic  system  increases 
aggression,  whereas  ablation  of  amyg- 
dala serotonin  decreases  aggression 
(Fileetal.  1981). 

The  serotonin  subtype  most  impli- 
cated in  animal  models  of  affective 
aggression  is  the  5-HT1B  receptor.  A 
class  of  substituted  phenlypiperazine 
analogs  that  display  remarkable  anti- 
aggressive  activity  in  animal  models 
has  been  termed  "serenics"  (Olivier  et 
al.  1990).  Serenics  specifically  reduce 
offensive  behavior  without  resulting  in 
sedation,  muscle  relaxation,  or  motor 
stimulation.  The  two  serenics  with  the 
greatest  specificity  are  TFMPP  and  RU 


24969,  phenlypiperazines  with  modest 
selectivity  and  high  affinity  for  the  5- 
HT1B  receptor.  As  already  mentioned, 
there  is  a  5-HT1B  knockout  mouse, 
and  this  animal  is  highly  aggressive 
(Saudou  et  al.  1994). 

The  effect  of  ethanol  on  aggression  is 
dose  dependent,  with  low  doses  increas- 
ing aggressive  acts  and  higher  doses 
decreasing  aggression,  probably  due  to 
sedation  (Blanchard  et  al.  1987;  Miczek 
et  al.  1993).  Increased  aggression  with 
low  doses  of  ethanol  has  been  found 
with  both  experimenter-administered 
and  self- administered  ethanol  studies 
(van  Erp  and  Miczek  1997).  However, 
increased  aggression  is  not  found  in 
every  animal  tested,  and  a  proportion  of 
the  population  shows  reduced  aggres- 
sion after  consuming  similar  doses  of 
ethanol  as  animals  showing  aggression. 
There  is  evidence  that  the  social  context 
can  determine  the  direction  and  mag- 
nitude of  ethanol's  effects  on  aggres- 
sion, particularly  in  monkeys  (Weerts 
and  Miczek  1996).  The  neurotrans- 
mitter systems  studied  in  relation  to 
ethanol-induced  aggression  in  animals 
models  have  been  the  5-HT  and  the 
GABAA  receptor  systems  (Blanchard 
et  al.  1993;  Miczek  et  al.  1993;  Weerts 
and  Miczek  1996).  The  5-HT1B 
receptor  system  has  not  been  specifi- 
cally studied  in  the  context  of  ethanol- 
induced  aggression  or  antiaggression, 
but  drug  discrimination  data  show 
that  the  5-HT1B  receptor  activation  is 
a  component  of  the  discriminative 
stimulus  effects  of  low  doses  of  ethanol 
(Grant  et  al.  1997).  5-HT1B  receptors 
are  autoreceptors  that  result  in  decreased 
5-HT  release,  and  low  5-HT  is  associ- 
ated with  increased  levels  of  aggression. 


244 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


However,  the  neuroanatomies  path- 
ways that  mediate  these  effects  are 
largely  unknown.  Very  recent  micro- 
dialysis  data  show  decreased  prefrontal 
cortex  5-HT  levels  in  aggressive  rats 
(van  Erp  and  Miczek  1997).  Since  the 
5-HT1B  receptor  system  is  predomi- 
nantly expressed  in  rodents,  studies  in 
primates  will  need  to  focus  on  the  5- 
HT1D  receptor  system,  using  the 
appropriate  ligands. 

The  interaction  of  alcohol  and 
aggression  is  a  major  public  health  con- 
cern. The  genetic,  neurochemical,  and 
neuroanatomical  bases  of  this  interac- 
tion are  largely  unknown.  There  are 
promising  data  implicating  both  the 
5-HT  and  the  GABAA  receptor  sys- 
tems, but  there  is  a  large  degree  of 
individual  variability  in  response  to 
ethanol  that  is  highly  dependent  on 
environmental  circumstances. 

FUTURE  RESEARCH 
BASED  ON  CURRENT 
APPROACHES 

A  priority  for  future  research  efforts 
should  be  transferring  information 
between  various  levels  of  analysis: 
molecular,  cellular,  physiological,  animal 
behavioral,  human  behavioral,  and 
epidemiologic.  The  transfer  from  cel- 
lular data  to  behavioral  models  appears 
to  be  well  in  hand,  and  the  use  of 
conditioning  procedures  such  as  place 
preference  and  drug  discrimination 
should  be  emphasized  in  mouse  mod- 
els. The  transfer  from  animal  models 
to  human  studies  is  less  apparent. 
Behavioral  neuroscience  can  address 
the  transfer  from  animals  to  humans 
by  applying  the  same  noninvasive 


technologies  in  animal  models  that  are 
used  in  human  studies.  These  tech- 
nologies include  brain  imaging  tech- 
niques of  PET,  single-photon 
emission  computed  tomography,  and 
functional  NMR  and  electrophysio- 
logical techniques  such  as  ERP. 

RISK  FACTORS 

Perhaps  one  of  the  greatest  challenges 
to  our  future  research  is  to  address  the 
neuroscientific  basis  of  risk  factors 
know  to  affect  alcohol  dependence  in 
humans.  Risk  factors  can  be  viewed  as 
an  additional  independent  variable  in 
the  design  of  our  animal  models,  but 
their  characterization  will  undoubt- 
edly require  that  data  be  obtained 
using  several  approaches.  Notable  risk 
factors  from  human  epidemiologic 
data  include  genetics,  gender  differ- 
ences, stress,  depression,  age  of  onset 
of  drinking,  and  the  concurrent  use  of 
other  drugs. 

The  most  progress  has  been  made  in 
addressing  the  role  of  genetics.  Alco- 
hol research  clearly  leads  the  way  in 
studying  the  influence  of  genes  on 
behavior.  Our  approaches  in  this  area 
are  strong,  but  the  behaviors  under 
study  are  often  of  questionable  extrap- 
olation to  the  human  condition  and 
the  animal  models  are  currently  lim- 
ited to  the  use  of  rodents.  Cloning 
techniques  of  nuclear  transplantation 
from  adult  monkeys  cells  should  at 
least  be  given  thoughtful  considera- 
tion. Such  approaches  could  be  used 
to  address  the  genetic  basis  of  com- 
plex behavioral  responses  associated 
with  the  development  of  alcohol 
abuse  and  alcoholism. 


245 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Gender  differences  in  the  behav- 
ioral neuroscience  of  alcohol  using 
animal  models  are  understudied.  The 
hypothalamic-pituitary-gonadal  axis 
hormones  have  been  implicated  in 
behavioral  outcomes,  primarily  cognitive 
function,  aggression,  and  stress.  Drug 
discrimination  studies  have  shown 
that  sensitivity  to  alcohol  is  altered  by 
menstrual  cycle  phase.  Gender  differ- 
ences in  self- administration  are  under- 
studied, as  are  the  anxiolytic  effects  of 
ethanol.  That  progesterone  derivatives 
have  been  shown  to  produce  ethanol- 
like  subjective  effects  and  alter  self- 
administration  is  an  important  clue  to 
follow  in  future  studies.  These  find- 
ings suggest  that  gender  differences 
must  take  into  account  the  menstrual 
cycle  phase. 

The  role  of  stress  in  ethanol's  behav- 
ioral effects  requires  more  sophisti- 
cated approaches.  It  is  important  to 
note  that  ethanol  has  both  anxiolytic 
and  anxiogenic  effects.  This  dual 
nature  of  ethanol  can  initiate  a  cycle 
of  behavior  centered  around  the  issue 
of  stress,  but  reflecting  different 
aspects  (circuitry?)  of  ethanol's  pharma- 
cology and  environmental  interaction. 
Studies  of  the  HPA  axis  hormones, 
particularly  the  central  action,  but  also 
the  extrahypothalamic  action,  of  hor- 
mones such  as  CRT  and  glucocorti- 
coids, are  beginning  to  yield  promising 
avenues  of  research. 

Depression  is  a  risk  factor  that  ani- 
mal models  of  alcohol  abuse  have  not 
addressed  extensively.  The  basic  ques- 
tions of  cause  and  effects  are  still 
unanswered  for  depression  and  its 
relationship  to  alcohol  abuse.  Animal 
models  are  best  suited  to  address  these 


questions,  since  ethical  and  economic 
considerations  limit  experimental  designs 
using  human  beings.  Links  between 
the  5-HT  and  dopamine  receptor  sys- 
tems with  depression  are  encouraging 
and  should  be  pursued. 

The  age  at  which  individuals  start 
regular,  heavy  use  of  alcohol  has 
recently  been  reported  to  predict  the 
occurrence  of  alcohol  dependence.  In 
particular,  individuals  who  begin  to 
drink  heavily  as  adolescents  have 
increased  risk  to  develop  alcohol 
dependence.  The  adolescent  period 
has  been  well  documented  in  nonhu- 
man  primates,  but  its  occurrence  in 
rodents  is  debatable.  If  solely  defined 
as  hypothalamic-pituitary-gonadal 
maturation,  the  adolescent  period  in 
rats  would  be  very  limited  and  studies 
of  complex  or  conditioned  behaviors 
might  not  be  possible.  The  macaque 
monkey  has  at  least  a  12-month  ado- 
lescent phase,  which  allows  a  window 
of  opportunity  to  design  appropriate 
experimental  manipulations.  In  addi- 
tion, the  social  behavior  of  macaques, 
particularly  in  response  to  aggressive 
behavior,  provides  a  rich  data  set  to 
test  hypotheses  of  predisposition  (trait) 
versus  reactivity  (state)  in  antisocial 
outcomes  following  alcohol  consump- 
tion within  a  social  context. 

The  concurrent  use  of  alcohol  and 
other  drugs  of  abuse  has  also  received 
limited  attention.  Heavy  alcohol  use  is 
correlated  with  benzodiazepine,  cocaine, 
opiate,  marijuana,  and  tobacco  abuse. 
Given  this  wide  pharmacological  diver- 
sity, explicit  receptor  interactions  are 
not  likely  to  explain  the  patterns  of 
abuse.  Behavioral  patterns  of  drug 
abuse  appear  to  be  robust  enough  to 


246 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


incorporate  a  wide  variety  of  psy- 
choactive substances  once  those 
behavioral  patterns  are  entrenched. 
How  these  codependent  use  patterns 
are  related  to  ethanol- specific  interac- 
tion in  the  CNS  remains  unclear. 

NOTES 

1 .  A  measure  of  coordinated  muscle 
movement  not  involved  in  locomotion 
or  anxiolytic  responses,  per  se,  is  taste 
reactivity.  This  measure  involves  the 
reflexive  ingestive  or  expulsive  oral 
movements  in  response  to  stimuli 
affecting  taste  sensory  pathways.  The 
procedure  involves  placing  small 
amounts  of  a  tastant,  such  as  ethanol, 
on  the  caudal  portion  of  the  tongue 
and  measuring  coordinated  tongue 
movements  that  result  in  either  the 
ingestion  or  the  expulsion  of  material 
from  the  oral  cavity.  Preference  for 
ethanol  has  been  assessed  with  this 
measure,  as  well  as  tolerance  to  expulsive 
response  and  sensitivity  to  ingestive 
response  given  repeated  exposure  to 
ethanol.  Since  the  taste  of  ethanol 
could  serve  as  a  conditioned  stimulus 
for  postingestional  effects  of  ethanol, 
changes  in  response  to  ethanol  exposure 
could  be  an  important  indicator  of 
reinforcement  development.  However, 
taste  reactivity  needs  to  be  investigated 
in  conjunction  with  reinforcement  and 
not  simply  ethanol  exposure  to  address 
these  possibilities.  Very  few  data  are 
available  on  the  underlying  neural  cir- 
cuitry involved  in  these  unconditioned 
responses,  and  no  data  address  the  cir- 
cuitry in  response  to  ethanol. 

2.  An  exception  to  this  generaliza- 
tion is  recent  evidence  suggesting  that 
muscimol  can  produce  ethanol-like 


discriminative  stimulus  effects  if 
injected  into  core  of  the  nucleus 
accumbens  or  amygdala  in  the  brain 
(Hodge  and  Aiken  1996;  Hodge  and 
Cox  1998). 

ACKNOWLEDGMENT 

Financial  support  for  the  preparation  of 
this  chapter  was  provided,  in  part,  by 
P50AA11997  from  the  National  Insti- 
tute on  Alcohol  Abuse  and  Alcoholism. 

REFERENCES 

Abrams,  D.G.,  and  Wilson,  G.T.  Effects 
of  alcohol  on  social  anxiety  in  women: 
Cognitive  versus  physiological  processes.  J 
Abnorm  Psychol  88:161-173,  1979. 

Altshuler,  H.L.;  Phillips,  P.E.;  and 
Feinhandler,  D.A.  Alteration  of  ethanol 
self-administration  by  naltrexone.  Life  Sci 
26:679-688,  1980. 

Andrews,  J.;  Turski,  L.;  and  Stephens, 
D.N.  Does  the  pentylenetetrazole  (PTZ) 
cue  reflect  PTZ-induced  kindling  of  PTZ- 
induced  angiogenesis?  Drug  Dev  Res 
16:247-256,  1989. 

Ashton,  H.  Guidelines  for  the  rational  use 
of  benzodiazepines:  When  and  what  to  use. 
Drugs  48-.25-A0,  1994. 

Ator,  N.A.,  and  Griffiths,  R.R. 
Asymmetrical  cross-generalization  in  drug 
discrimination  with  lorazepam  and  pento- 
barbital training  conditions.  Drug  Dev  Res 
16:355-364,  1989. 

Ator,  N.A.;  Grant,  K.A.;  Purdy,  R.H.; 
Paul,  S.M.;  and  Griffiths,  R.R.  Drug 
discrimination  analysis  of  endogenous 
neuroactive  steroids  in  rats.  Eur  J 
Pharmacol  241:237-244,  1993. 


247 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Baldwin,  H.A.;  Rassnick,  S.;  Rivier,  J.; 
Koob,  G.F.;  and  Britton,  K.T.  CRF  an- 
tagonist reverses  the  "anxiogenic"  re- 
sponse to  ethanol  withdrawal  in  the  rat. 
Psychopharmacology  103:227-232,  1991. 

Ballinger,  B.R.  Hypnotics  and  anxiolytics. 
5^/300:456^58,  1990. 

Baron,  S.,  and  Moerschbaecher,  J.M. 
Disruption  of  learning  by  excitatory 
amino  acid  receptor  antagonists.  Behav 
Pharmacol  6:573-584,  1996. 

Barrett,  J.E.,  and  Vanover,  K.E.  5-HT 
receptors  as  targets  for  the  development 
of  novel  anxiolytic  drugs:  Models,  mecha- 
nisms and  future  directions.  Psycho- 
pharmacology  112:1-12,  1993. 

Barrett,  J.E.;  Zhang,  L.;  Gleeson,  S.;  and 
Gamble,  E.H.  Anxiolytic  and  antidepressant 
mechanisms  of  5-HT1A  drugs  in  the  pigeon: 
Contributions  from  behavioral  studies. 
Neurosci  Biobehav  Rev  18:73-83,  1994. 

Bell,  S.M.;  Reynolds,  J.G.;  Thiele,  T.E.; 
Gan,  J.;  Figlewicz,  D.P.;  and  Woods,  S.C. 
Effects  of  third  intracerebroventricular 
injections  of  corticotropin-releasing  factor 
(CRF)  on  ethanol  drinking  and  food 
intake.  Psychopharmacology  (Berl) 
139:128-135,  1998. 

Benkelfat,  C;  Murphy,  D.L.;  Hill,  J.I.; 
George,  D.T.;  Nutt,  D.;  and  Linnoila,  M. 
Ethanolike  properties  of  the  serotonergic 
partial  agonist  m-chlorophenylpiperazine 
in  chronic  alcoholic  patients.  Arch  Gen 
Psychiatry  48:383,  1991. 

Blanchard,  D.C.;  Veniegas,  R;  Elloran,  I.; 
and  Blanchard,  RJ.  Alcohol  and  anxiety: 
Effects  on  offensive  and  defensive  aggres- 
sion. J  Stud  Alcohol  Suppl  11:9-19,  1993. 

Blanchard,  R.J.;  Hori,  K.;  Blanchard, 
D.C.;  and  Hall.  J.  Ethanol  effects  on 
aggression  of  rats  selected  for  different 


levels  of  aggressiveness.  Pharmacol 
Biochem  Behav  27:641-644,  1987. 

Bormann,  N.M.,  and  Cunningham,  C.L. 
The  effects  of  naloxone  on  expression  and 
acquisition  of  ethanol  place  conditioning 
in  rats.  Pharmacol  Biochem  Behav  58:975- 
982,  1997. 

Bormann,  N.M.,  and  Cunningham,  C.L. 
Ethanol-induced  conditioned  place  aver- 
sion in  rats:  Effect  of  interstimulus  inter- 
val. Pharmacol  Biochem  Behav  59:427- 
432,  1998. 

Bowen,  C.A.;  Gatto,  G.J.;  and  Grant,  KA. 
Assessment  of  the  mixed  discriminative 
stimulus  effects  of  ethanol  using  an  ethanol  - 
pentobarbital-water  discrimination  in  rats. 
Behav  Pharmacol  8:339-352,  1997. 

Britan,  D.;  Shiekh,  M.;  and  McLeod,  M. 
Anxiolytic  effect  of  progesterone  is  medi- 
ated by  the  neurosteroid  allopregnanolone 
at  brain  GABAA  receptors.  /  Neuroendo- 
crinal 7:17 '1-177 ',  1991. 

Broadbent,  J.;  Linder,  H.V.;  and 
Cunningham,  C.L.  Genetic  differences  in 
naloxone  enhancement  of  ethanol-induced 
conditioned  taste  aversion.  Psychopharma- 
cology  (Berl)  126:147-155,  1996. 

Brown,  D.,  and  Holtzman,  S.  Narcotic 
antagonists  attenuate  drinking  induced  by 
water  deprivation  in  a  primate.  Life  Sci 
28:1287-1294,1981. 

Buczek,  Y.;  Tomkins,  D.M.;  Higins,  G.A.; 
and  Sellers,  E.M.  Dissociation  of  seroton- 
ergic regulation  of  anxiety  and  ethanol 
self- administration:  A  study  with  mCPP. 
Behav  Pharmacol  5:470-484,  1994. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin,  C. 
Hormonal,  psychological  and  alcohol  crav- 
ing changes  after  m-cholorophenylpiperazine 
administration  in  alcoholics.  Alcohol  Clin 
Exp  Res  21:220-227,  1997. 


248 


Behavioral  Effects  and  Underlying  Neurocircui tries  of  Alcohol 


Carey,  M.P.;  Fry,  J.P.;  and  White,  D.G. 
Anxiety  and  the  discriminative  stimulus 
induced  by  pentylenetetrazole  in  the  pig. 
Psychopharmacology  101:S66,  1990. 

Carroll,  M.E.;  Stitzer,  M.L.;  Strain,  E.;  and 
Meisch,  RA.  The  behavioral  pharmacology 
of  alcohol  and  other  drugs.  In:  Galanter, 
M.,  ed.  Recent  Developments  in  Alcoholism. 
Vol.  8.  New  York,  Plenum,  1990.  pp.  5^6. 

Chester,  J. A.;  Risinger,  F.O.;  and 
Cunningham,  C.L.  Ethanol  reward  and 
aversion  in  mice  bred  for  sensitivity  to 
ethanol  withdrawal.  Alcohol  Clin  Exp  Res 
22:468^173,  1998. 

Chutuape,  M.A.D.,  and  deWit,  H. 
Preferences  for  ethanol  and  diazepam  in 
anxious  individuals:  An  evaluation  of  the 
self-medication  hypothesis.  Psychopharma- 
cology  (Berl)  121:91-103,  1995. 

Cohen,  M.S.,  and  Bookheimer,  S.Y. 
Localization  of  brain  function  using  mag- 
netic resonance  imaging.  Trends  Neurosci 
17:268-277,  1994. 

Colombo,  G.;  Agabio,  R.;  Lobina,  C; 
Reali,  R.;  Fadda,  F.;  and  Gessa,  G.L. 
Blockade  of  ethanol  discrimination  by 
isradipine.  Eur  J  Pharmacol  265:167- 
170, 1994. 

Colombo,  G.;  Agabio,  R.;  Lobina,  C; 
Reali,  R.;  Zocchi,  A.;  Fadda,  F.;  and 
Gessa,  G.L.  Sardinian  alcohol-preferring 
rats:  A  genetic  model  of  anxiety.  Physiol 
.foW  57:1181-1185,  1995. 

Colpaert,  F.C.  Drug  discrimination: 
Behavioral,  pharmacological,  and  molecular 
mechanisms  of  discriminative  drug  effects. 
In:  Goldberg,  S.R,  and  Stolerman,  LP., 
eds.  Behavioral  Analysis  of  Drug  Dependence. 
Orlando,  FL:  Academic  Press,  1986.  pp 
161-193. 

Crabbe,  J.C.;  Phillips,  T.J.;  Cunningham, 
C.L.;  and  Belknap,  J.K.  Genetic  determi- 


nants of  ethanol  reinforcement.  Ann  NT 
AcadSci  654:302-310,  1992. 

Crabbe,  J.C.;  Phillips,  T.J.;  Feller,  D.J.; 
Hen,  R;  Wenger,  CD.;  Lessov,  C.N.; 
and  Schafer,  G.L.  Elevated  alcohol  con- 
sumption in  null  mutant  mice  lacking  5- 
HT1B  serotonin  receptors.  Nat  Genet 
14:98-102,  1996. 

Crawley,  J.N.;  Glowa,  J.R.;  Majewska, 
M.D.;  and  Paul,  A.M.  Anxiolytic  activity 
of  an  endogenous  adrenal  steroid.  Brain 
Res  398:382-385,  1986. 

Criswell,  H.E.;  Simson,  P.E.;  Knapp, 
D.J.;  Devaus,  L.L.;  McCown,  T.J.; 
Duncan,  G.E.;  Morrow,  A.L.;  and  Breese, 
G.R.  Effect  of  Zolpidem  of  gamma- 
aminobutyric  acid  (GAB A) -induced  inhi- 
bition predicts  the  interaction  of  ethanol 
with  GABA  on  individual  neurons  in 
several  rat  brain  regions.  J  Pharmacol  Exp 
Ther  273:526-536,1995. 

Crowley,  T.J.;  Williams,  E.A.;  and  Jones, 
RH.  Initiating  ethanol  drinking  in  a  simian 
social  group  in  a  naturalistic  setting. 
Alcohol  Clin  Exp  Res  14:444-^45,  1990. 

Crum,  R.M.;  Muntraner,  C;  Eaton, 
W.W.;  and  Anthony,  J.C.  Occupational 
stress  and  the  risk  of  alcohol  abuse  and 
dependence.  Alcohol  Clin  Exp  Res 
19:647-655,  1995. 

Cunningham,  C.L.  Localization  of  genes 
influencing  ethanol-induced  conditioned 
place  preference  and  locomotor  activity  in 
BXD  recombinant  inbred  mice.  Psycho- 
pharmacology  (Berl)  120:28-^1,  1995. 

Cunningham,  C.L.,  and  Noble,  D. 
Conditioned  activation  induced  by 
ethanol:  Role  in  sensitization  and  condi- 
tioned place  preference.  Pharmacol 
Biochem  Behav  43:307-313,  1992. 

Cunningham,  C.L.;  Hallett,  C.L.; 
Niehus,  D.R.;  Hunter,  J.S.;  Nouth,  L.; 


249 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  Risinger,  F.O.  Assessment  of  ethanol's 
hedonic  effects  in  mice  selectively  bred  for 
sensitivity  to  ethanol-induced  hypothermia. 
Psychopharmacology  105:84-92,  1991. 

Cunningham,  C.L.;  Niehus,  D.R.;  Malott, 
D.H.;  and  Prather,  L.K.  Genetic  differences 
in  the  rewarding  and  activating  effects  of 
morphine  and  ethanol.  Psychopharma- 
cology  107:385-393,  1992. 

Cunningham,  C.L.;  Dickinson,  S.D.;  and 
Okorn,  D.M.  Naloxone  facilitates  extinction 
but  does  not  affect  acquisition  or  expres- 
sion of  ethanol-induced  conditioned  place 
preference.  Exp  Clin  Psychopbarmacol 
3:330-343,  1995. 

Cunningham,  C.L.;  Okorn,  D.M.;  and 
Howard,  C.E.  Interstimulus  interval  deter- 
mines whether  ethanol  produces  condi- 
tioned place  preference  or  aversion  in  mice. 
Anim  Learning  Behav  25:31^i2,  1997. 

Cunningham,  C.L.;  Henderson,  CM.; 
and  Bormann,  N.M.  Extinction  of 
ethanol-induced  conditioned  place  prefer- 
ence and  conditioned  place  aversion: 
Effects  of  naloxone.  Psychopharmacology 
(Berl)  139:62-70,  1998. 

Davies,  B.T.,  and  Parker,  LA.  Novel  versus 
familiar  ethanol:  A  comparison  of  aversive 
and  rewarding  properties.  Alcohol  7:523- 
529,  1990. 

Davis,  M.  The  role  of  the  amygdala  in 
fear-potentiated  startle:  Implications  for 
animal  models  of  anxiety.  Trends 
Pharmacol  Sci  13(1):35-41,  1992. 

Dawson,  G.R.,  and  Tricklebank,  M.D. 
Use  of  elevated  plus  maze  in  the  search 
for  novel  anxiolytic  agents.  Trends 
Pharmacol  Sci  16:33-36,  1995. 

De  Beun,  R.;  Lohmann,  A.;  Schneider, 
R.;  and  De  Vry,  J.  Comparison  of  the 
stimulus  properties  of  ethanol  and  the 


Ca2+  channel  antagonist  nimodipine  in 
rats.  Eur  J  Pharmacol  306:5-13,  1996. 

De  Waele,  J. -P.;  Papachristou,  D.N.;  and 
Gianoulakis,  C.  The  alcohol-preferring 
C57BL/6  mice  present  an  enhanced 
sensitivity  of  the  hypothalamic  (3-endor- 
phin  system  to  ethanol  than  the  alcohol- 
avoiding  DBA/2  mice.  J  Pharmacol  Exp 
Ther  261:788-794,  1992. 

De  Vry,  J.  5-HT1A  receptor  agonists: 
Recent  developments  and  controversial 
issues.  Psychopharmacology  (Berl) 
121:1-26,  1995. 

Dickinson,  S.D.,  and  Cunningham,  C.L. 
Altered  ambient  temperature  and  ethanol- 
induced  conditioned  place  preference  in 
mice.  Alcohol  16:13-18,  1998. 

Dourish,  C.T.;  Hutson,  P.H.;  and  Curzon, 
G.  Putative  anxiolytics  8-OH-DPAT, 
buspirone  and  TVXQ  7821  are  agonists  at 
5-HT1A  autoreceptors  in  the  raphe  nuclei. 
Trends  Pharmacol  Sci  7:212-214,  1986. 

Eckhardt,  M.J.;  File,  S.E.;  Gessa,  G.L.; 
Grant,  K.A.;  Guerri,  C;  Hoffman,  P.L.; 
Kalant,  H.;  Koob,  G.F.;  Li,  T.-K.;  and 
Tabakoff,  B.  Effects  of  moderate  alcohol 
consumption  on  the  central  nervous  system. 
Alcohol  Clin  Exp  Res  22:998-1040,  1998. 

Emmett-Ogelsby,  M.W.;  Mathis,  D.A.; 
Moon,  R.T.Y.;  and  Lai,  H.  Animal  mod- 
els of  drug  withdrawal  symptoms.  Psycho- 
pharmacology  101:292-309,  1990. 

Fernandez-Guasti,  A.,  and  Picazo,  O. 
Flumazenil  blocks  the  anxiolytic  action  of 
allopregnanolone.  Eur  J  Pharmacol  281: 
113-115, 1995. 

File,  S.E.;  James,  R.A.;  and  MacLeod, 
N.K.  Depletion  in  amygdaloid  5-hydrox- 
ytryptamine  concentration  and  changes  in 
social  and  aggressive  behaviour.  /  Neural 
Transm  50:1-12,  1981. 


250 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


File,  S.E.;  Gonzalez,  L.E.;  and  Andrews, 
N.  Comparative  study  of  pre-  and  post- 
synaptic 5-HT1A  receptor  modulation  of 
anxiety  in  two  ethological  animal  tests.  / 
Neurosci  16:4810-^815,  1996. 

Fletcher,  P.J.;  Ming,  Z.H.;  and  Higgins, 
GA.  Conditioned  place  preference  induced 
by  microinjection  of  8-OH  DPAT  into 
the  dorsal  or  median  raphe  nucleus. 
Psychopharmacology  113:31-36,  1994. 

Froelich,  J.,  and  Li,  T.-K.  Opioid  pep- 
tides. In:  Galanter,  M.,  ed.  Recent 
Developments  in  Alcoholism,  Vol.  11:  Ten 
Tears  of  Progress.  New  York:  Plenum 
Press,  1993.  pp.  187-205. 

Froehlich,  J.C.;  Harts,  J.;  Lumeng,  L.; 
and  Li,  T.-K.  Differences  in  response 
to  the  aversive  properties  of  ethanol  in 
rats  selectively  bred  for  oral  ethanol 
preference.  Pharmacol  Biochem  Behav 
31:215-222,  1988. 

Froehlich,  J.C.;  Badia-Elder,  N.E.;  Zink, 
R.W.;  McCullough,  D.E.;  and  Portoghese, 
P.S.  Contribution  of  the  opioid  system  to 
alcohol  aversion  and  alcohol  drinking 
behavior.  J  Pharmacol  Exp  Ther  287:284- 
292, 1998. 

Gatto,  G.J.,  and  Grant,  KA.  Attenuation 
of  the  discriminative  stimulus  effects  of 
ethanol  by  the  benzodiazepine  partial 
inverse  agonist  Ro  15-4513.  Behav 
Pharmacol  8:139-146,  1997. 

Gatto,  G.J.;  McBride,  W.J.;  Murphy, 
J.M.;  Lumeng,  L.;  and  Li,  T.K.  Ethanol 
self-infusion  into  the  ventral  tegmental 
area  by  alcohol -preferring  rats.  Alcohol 
11:557-564,  1994. 

Gauvin,  D.V.,  and  Holloway,  FA.  Cross- 
generalization  between  an  ecologically 
relevant  stimulus  and  a  pentylenetetrazole- 
discriminative  cue.  Pharmacol  Biochem 
Behav  39:521-523,  1991. 


Gauvin,  D.V.,  and  Holloway,  FA. 
Ethanol  tolerance  developed  during  in- 
toxicated operant  performance  in  rats 
prevents  subsequent  ethanol-induced 
conditioned  taste  aversion.  Alcohol 
9:167-170,  1992. 

Gauvin,  D.V.;  Hardland,  R.D.;  Criado, 
J.R.;  Michaelis,  R.C.;  and  Holloway,  FA. 
The  discriminative  stimulus  properties  of 
ethanol  and  acute  ethanol  withdrawal 
states  in  rats.  Drug  Alcohol  Depend 
24:103-113,  1989. 

Gauvin,  D.V.;  Youngblood,  B.D.;  and 
Holloway,  F.A.  The  discriminative  stimu- 
lus properties  of  acute  ethanol  withdrawal 
(hangover)  in  rats.  Alcohol  Clin  Exp  Res 
16:336-341,  1992. 

Gauvin,  D.V.;  Youngblood,  B.D.; 
Goulden,  K.L.;  Briscoe,  R.J.;  and 
Holloway,  FA.  Multidimensional  analyses 
of  an  ethanol  discriminative  cue.  Exp  Clin 
Psychopharmacol  4:299-309,  1994. 

Gianoulakis,  C.  Characterization  of  the 
effects  of  acute  ethanol  administration  on 
the  release  of  B-E  peptides  by  rat  hypothal- 
amus. Eur  J  Pharmacol  180:21-29,  1990. 

Givens,  B.  Low  doses  of  ethanol  impair 
spatial  working  memory  and  reduce  hip- 
pocampal  theta  activity.  Alcohol  Clin  Exp 
Ther  19:763-767,  1995. 

Givens,  B.;  Williams,  J.;  Gill,  T.M. 
Cognitive  correlates  of  single  neuron 
activity  in  task-performing  animals: 
Application  to  ethanol  research.  Alcohol 
Clin  Exp  Res  22:23-31,  1998. 

Goldberg,  S.R.;  Hoffmeister,  F.;  and 
Schlichting,  U.U.  Morphine  antagonists: 
Modification  of  behavioral  effects  by 
morphine  dependence.  In:  Singh,  J.M.; 
Miller,  L.;  and  Lai,  H.,  eds.  Experimental 
Pharmacology.  New  York:  Futura,  1978. 
pp.  31-48. 


251 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Goldberg,  S.R;  Hoffmeister,  F.;  Schlichting, 
U.U.;  and  Wuttke,  W.  Aversive  properties 
of  nalorphine  and  naloxone  in  morphine- 
dependent  monkeys.  J  Pharmacol  Exp 
Ther  179:268-276,  1971. 

Goldstein,  A.;  Aronow,  L.;  and  Kalman, 
S.M.  Principles  of  Drug  Action.  New 
York:  Harper  &  Row,  1968. 

Gordon,  T.L.;  Meehan,  S.M.;  and 
Schechter,  M.D.  P  and  NP  rats  respond 
differentially  to  the  discriminative  stimu- 
lus effects  of  nicotine.  Pharmacol  Biochem 
5^  45:305-308,  1993. 

Grant,  K.A.  Emerging  neurochemical 
concepts  in  the  actions  of  ethanol  at  lig- 
and-gated  ion  channels.  Behav  Pharmacol 
5:383-404,  1994. 

Grant,  K.A.,  and  Barrett,  J.E.  Blockade  of 
the  discriminative  stimulus  effects  of 
ethanol  with  5-HT3  receptor  antagonists. 
Psychopharmacology  104:451-456,  1991. 

Grant,  K.A.,  and  Colombo,  G.  Substitution 
of  the  5-HT!  agonist  trifluoromethyl- 
phenylpiperazine  (TFMPP)  for  the  dis- 
criminative stimulus  effects  of  ethanol: 
Effect  of  training  dose.  Psychopharmacology 
113:26-30,1993. 

Grant,  K.A.,  and  Johanson,  C.E.  Oral 
ethanol  self- administration  in  free-feeding 
rhesus  monkeys,  Alcohol  Clin  Exp  Res 
12:780-784,  1988. 

Grant,  K.A.,  and  Lovinger,  D.L.  Cellular 
and  behavioral  neurobiology:  Acute  and 
chronic  effects  of  alcohol  on  receptor- 
mediated  neuronal  processes  and  their 
impact  on  animal  behavior.  Clin  Neurosci 
3:155-164,  1995. 

Grant,  K.A.;  Hoffman,  P.L.;  and  Tabakoff, 
B.  Neurobiological  and  behavioral  ap- 
proaches to  tolerance  and  dependence. 
In:  Edwards,  G.,  and  Lader,  M.,  eds.  The 


Nature  of  Dependence.  New  York:  Oxford 
University  Press,  1990.  pp.  135-169. 

Grant,  K.A.;  Kinsley,  J.S.;  Tabakoff,  B.; 
Barrett,  J.E.;  and  Balster,  RL.  Ethanol-like 
discrimination  stimulus  effects  of  non- 
competitive N-methyl-D-aspartate  antag- 
onists. Behav  Pharmacol  2:87-95,  1991. 

Grant,  K.A.;  Azarov,  A.;  Bowen,  C.A.; 
Mirkis,  S.;  and  Purdy,  R.H.  Ethanol-like 
discriminative  stimulus  effects  of  the  neuro- 
steroid  3a-hydroxy-5a-pregnan-20-one  in 
female  Macaca  fascicularis  monkeys.  Psycho- 
pharmacology  (Berl)  124:340-346,  1996. 

Grant,  K.A.;  Colombo,  G.;  and  Gatto, 
GJ.  Characterization  of  the  ethanol-like 
discriminative  stimulus  effects  of  5-HT 
receptor  agonists  as  a  function  of  ethanol 
training  dose.  Psychopharmacology  (Berl) 
133:133-141,  1997. 

Grant,  K.A.;  Young,  J.E.;  Kautz,  M.A.; 
Green,  H.L.;  Sharpe,  A.L.;  and  Shiely, 
C.A.  Induction  and  maintenance  of  oral 
ethanol  self- administration  in  male  and 
female  monkeys.  Alcohol  Clin  Exp  Res 
22:12A,  1998. 

Grech,  D.M.,  and  Balster,  RL. 
Pentobarbital-like  discriminative  stimulus 
effects  of  direct  GAB  A  agonists  in  rats. 
Psychopharmacology  (Berl)  110:295- 
301,  1993. 

Grech,  D.M.,  and  Balster,  RL.  The  dis- 
criminative stimulus  effects  of  muscimol 
in  rats.  Psychopharmacology  (Berl)  129:339- 
347,  1997. 

Green,  K.L.,  and  Grant,  K.A.  Evidence 
for  overshadowing  by  components  of  the 
heterogeneous  discriminative  stimulus 
effects  of  ethanol.  Drug  Alcohol  Depend 
52:149-159,  1998. 

Griebel,  G.  5-Hydroxytryptamine -interacting 
drugs  in  animal  models  of  anxiety  disorders: 


252 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


More  than  30  years  of  research.  Pharmacol 
Ther  65:319-395,  1995. 

Harrison,  Y.E.;  Jenkins,  J.A.;  Rocha,  B.A.; 
Lytle,  DA.;  Jung,  M.E.;  and  Ogelsby, 
M.E.  Discriminative  stimulus  effects  of 
diazepam,  ketamine,  and  their  mixture: 
Ethanol  substitution  patterns.  Behav 
Pharmacol  9:31-40,  1998. 

Harvey,  J  A.  Effects  of  drugs  on  associa- 
tive learning.  In:  Meltzer,  H.,  ed.  Psycho- 
pharmacology:  The  Third  Generation  of 
Progress.  New  York:  Raven  Press,  1987. 
pp. 1485-1492. 

Higley,  J.D.;  Hasert,  M.F.;  Suomi,  S.J.; 
and  Linnoila,  M.  Nonhuman  primate 
model  of  alcohol  abuse:  Effects  of  early 
experience,  personality,  and  stress  on 
alcohol  consumption.  Proc  Natl  Acad  Sci 
USA  88:7261-7265,  1991. 

Hiltunen,  A.J.,  and  Jarbe,  T.U.C.  Discrim- 
ination of  Ro  11-6896,  chlordiazepoxide 
and  ethanol  in  gerbils:  Generalization  and 
antagonism  tests.  Psychopharmacology 
89:284-290,  1986. 

Hiltunen,  A.J.,  and  Jarbe,  T.U.C.  Ro  15- 
4513  does  not  antagonize  the  discrimina- 
tive stimulus-  or  rate-depressant  effects  of 
ethanol  in  rats.  Alcohol  5:203-207,  1988. 

Hiltunen,  A.J.,  and  Jarbe,  T.U.C.  Discrim- 
inative stimulus  properties  of  ethanol: 
Effects  of  cumulative  dosing  and  Ro  15- 
4513.  Behav  Pharmacol  1:133-140,  1989. 

Hinson,  R.E.;  Poulos,  C.X.;  Thomas,  W.; 
and  Cappell,  H.  Pavlovian  conditioning 
and  addictive  behavior:  Relapse  to  oral 
self- administration  of  morphine.  Behav 
Neurosci  100:368-375,  1986. 

Hitchcott,  P.K.,  and  Phillips,  G.D.  Amy- 
gdala and  hippocampus  control  dissocia- 
ble aspects  of  drug-associated  conditioned 
rewards.  Psycho-pharmacology  (Berl)  131: 
187-195,  1997. 


Hitchcott,  P.K.;  Bonardi,  C.M.T.;  and 
Phillips,  G.D.  Enhanced  stimulus-reward 
learning  by  intra- amygdala  administration 
of  a  D3  dopamine  receptor  agonist.  Psycho- 
pharmacology  (Berl)  133:240-248,  1997a;. 

Hitchcott,  P.K.;  Harmer,  C.J.;  and 
Phillips,  G.D.  Enhanced  acquisition  of 
discriminative  approach  following  intra- 
amygdala  d-amphetamine.  Psychopharma- 
cology  (Berl)  132:237-246,  1997b. 

Hodge,  C.W.,  and  Aiken,  A.S.  Discrim- 
inative stimulus  function  of  ethanol:  Role 
of  GABAA  receptors  in  the  nucleus  accum- 
bens.  Alcohol  Clin  Exp  Res  20:1221- 
1228, 1996. 

Hodge,  C.W.,  and  Cox,  AA.  The  discrim- 
inative stimulus  effects  of  ethanol  are  medi- 
ated by  NMDA  and  GABAA  receptors  in 
specific  limbic  brain  regions.  Psychopharma- 
cology  (Berl)  139:95-107,  1998. 

Hoffman,  D.C.  The  use  of  place  condition- 
ing in  studying  the  neuropharmacology  of 
drug  reinforcement.  Brain  Res  Bull  23: 
373-397,  1989. 

Holloway,  FA.;  King,  DA.;  Bedingfield, 
J.B.;  and  Gauvin,  D.V.  Role  of  context  in 
ethanol  tolerance  and  subsequent  hedonic 
effects.  ^/c<^o/9:109-116,  1992. 

Holtzman,  S.G.  Discriminative  stimulus 
effects  of  drugs:  Relationship  to  potential 
for  abuse.  In:  Adler,  M. W.,  and  Cowan,  A., 
eds.  Modern  Methods  in  Pharmacology,  Vol. 
6:  Testing  and  Evaluation  of  Drugs  of  Abuse. 
New  York:  Wiley-Liss,  1990.  pp.  193-210. 

Hundt,  W.;  Danysz,  W.;  Holter,  S.M.;  and 
Spanagel,  R.  Ethanol  and  N-methyl-D- 
aspartate  receptor  complex  interactions:  A 
detailed  drug  discrimination  analysis.  Psycho- 
pharmacology  (Berl)  135:44-51,  1998. 

Jones,  G.H.;  Schneider,  C;  Schneider, 
H.H.;  Seidler,  J.;  Cole,  B.J.;  and 
Stephens,  D.N.  Comparison  of  several 


253 


NIAAA's  Neuroscience  and  Beha\rioral  Research  Portfolio 


benzodiazepine  receptor  ligands  in  two 
models  of  anxiolytic  activity  in  the  mouse: 
An  analysis  based  on  fractional  receptor 
occupancies.  Psychopharmacology  (Berl) 
114:191-199,  1994. 

Juarez,  J.;  Guzman-Flores,  C;  Ervin,  F.R.; 
and  Palmour,  R.M.  Voluntary  alcohol 
consumption  in  vervet  monkeys:  Individual, 
sex,  and  age  differences.  Pharmacol 
BiocbemBehav  46:985-988,  1993. 

Kandel,  D.A.,  and  Schuster,  C.R  An  inves- 
tigation of  nalorphine  and  perphenazine 
as  negative  reinforcers  in  an  escape  paradigm. 
Pharmacol  Biochem  Behav  6:61-71,  1977. 

Kenakin,  T.  Pharmacological  Analysis  of 
Drug-Receptor  Interactions.  New  York: 
Raven  Press,  1993. 

Koob,  G.F.,  and  Britton,  K.T.  Neurobio- 
logical  substrates  for  the  anti-anxiety  effects 
of  ethanol.  In:  Begleiter,  H.,  and  Kissin, 
B.,  eds.  Pharmacology  of  Alcohol  and 
Alcohol  Dependence.  New  York:  Oxford 
University  Press,  1996.  pp.  477-506. 

Kornet,  M.;  Goosen,  C.;  and  Van  Ree,  J. 
Effect  of  naltrexone  on  alcohol  consump- 
tion during  chronic  alcohol  drinking  and 
after  a  period  of  imposed  abstinence  in 
free-choice  drinking  rhesus  monkeys. 
Psychopharmacology  104:367-376,  1991. 

Kraemer,  G. W.,  and  McKinney,  W.T. 
Social  separation  increases  alcohol  con- 
sumption in  rhesus  monkeys. 
Psychopharmcology  86:182-189,  1985. 

Krimmer,  E.C.  LLAS  and  LAS  rats  re- 
spond differentially  to  behavioral  effects 
of  ethanol,  pentobarbital,  chlorpromazine 
and  chlordiazepoxide.  Pharmacol  Biochem 
Behav  39:5-13,  1991. 

Krimmer,  E.C.  Biphasic  effects  of  ethanol 
tested  with  drug  discrimination  in  EIAD 
and  LAD  rats.  Pharmacol  Biochem  Behav 
43:1233-1240,  1992. 


Krishnan-Sarin,  S.;  Jing,  S.-L.;  Kurtz,  D.L.; 
Zweifel,  M.;  Portoghese,  P.S.;  Li,  T.-K;  and 
Froehlich,  J.C.  The  delta  opioid  receptor 
antagonist  naltrindole  attenuates  both  alco- 
hol and  saccharin  intake  in  rats  selectively 
bred  for  alcohol  preference.  Psychopharma- 
cology (Berl)  120:177-185,  1995. 

Krystal,  J.H.;  Webb,  E.;  Cooney,  N.; 
Kranzler,  H.R.;  and  Charney,  D.S. 
Specificity  of  ethanol-like  effects  elicited 
by  serotonergic  and  noradrenergic 
mechanisms.  Arch  Gen  Psychiatry 
51:898-911,1994. 

Krystal,  J.LL;  Webb,  E.;  Grillon,  C.; 
Cooney,  N.;  Casal,  L.;  Morgan,  C.A.,  III; 
Southwick,  S.M.;  Davis,  M.;  and  Charney, 
D.S.  Evidence  of  acoustic  startle  hyperflexia 
in  recently  detoxified  early  onset  male 
alcoholics:  Modulation  by  yohimbine  and 
m-chlorophenlypiperazine  (mCPP)  Psycho- 
pharmacology  (Berl)  131:207-215,  1997. 

Krystal,  J.H.;  Karper,  L.P.;  Bennett,  A.; 
D'Souza,  D.C.;  Abi-Dargham,  A.; 
Morrissey,  K.;  Abi-Saab,  D.;  Bremner, 
J.D.;  Bowers,  M.B.;  Suckow,  RF.;  Stetson, 
P.;  Heninger,  G.R.;  and  Charney,  D.  Inter- 
active effects  of  subanesthetic  ketamine  and 
subhypnotic  lorazepam  in  humans.  Psycho- 
pharmacology  (Berl)  135:213-229,  1998. 

Kulkosky,  P.J.  Brain-gut  neuropeptides 
and  the  limitation  of  ethanol  consumption. 
Neurosci  Biobehav  Rev  9:179-190,  1985. 

Kulkosky,  P.J.;  Glazner,  G.W.;  Moore, 
H.D.;  Low,  C.A.;  and  Woods,  S.C.  Neuro- 
peptide Y:  Behavioral  effects  in  the  golden 
hamster.  Peptides 9:1389-1393,  1988. 

Lai,  PL,  and  Emmett-Oglesby,  MW. 
Behavioral  analogues  of  anxiety:  Animal 
models.  Neuropharmacology  22:1423- 
1441,  1983. 

Lai,  PL;  Harris,  CM.;  Benjamin,  D.; 
Springfield,  A.C.;  Bhadra,  S.;  and 


254 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


Emmett-Oglesby,  M.W.  Characterization 
of  a  pentylenetetrazol-like  interoceptive 
stimulus  produced  by  ethanol  withdrawal. 
J  Pharmacol  Exp  Ther  247:508-518, 
1988. 

Langenbucher,  J.W.,  and  Nathan,  P.E.  The 
tension-reduction  hypothesis:  A  reanalysis 
of  some  early  crucial  data.  In:  Cox,  W.M., 
ed.  Why  People  Drink:  Parameters  of  Alcohol 
as  a  Reinforcer.  New  York,  Gardner  Press, 
1990.  pp.  131-168. 

Le,  A.D.  Selective  breeding  for  rats  which 
voluntarily  consume  high  and  low 
amounts  of  alcohol  in  a  limited  access 
paradigm.  Oral  communication  presented 
at  the  International  Society  for 
Biomedical  Research  on  Alcohol  (IS- 
BRA)-Research  Society  on  Alcoholism 
(RSA)  joint  meeting.  Washington,  DC, 
June  22-27,  1996. 

Leidenheimer,  N.J.,  and  Schechter,  M.D. 
Inverse  agonist  properties  of  FG  7142 
discriminative  stimulus.  Pharmacol 
Biochem  Behav  38:99-104,  1990. 

Lister,  R.G.  Ethologically- based  animal 
models  of  anxiety  disorders.  Pharmacol 
Ther  46:321-340,  1990. 

Litten,  R.Z.;  Allen,  J.;  and  Fertig,  J. 
Pharmacotherapies  for  alcohol  problems: 
A  review  of  research  with  focus  on  devel- 
opments since  1991.  Alcohol  Clin  Exp  Res 
20:859-876,  1996. 

Locke,  K.;  Brown,  D.;  and  Holtzman,  S. 
Effects  of  opiate  antagonist  and  putative 
mu-  and  kappa-agonist  on  milk  intake  in 
rat  and  squirrel  monkey.  Pharmacol 
Biochem  Behav  17:1276-1279,  1982. 

Lovinger,  D.M.  Excitotoxicity  and  alco- 
hol-related brain  damage.  Alcohol  Clin 
Exp  Res  17 A9-27,  1993. 

Lucki,  I.  5-HT1  receptors  and  behavior. 
Neurosci  Biobehav  Rev  16:83-93,  1992. 


Macenski,  M.J.,  and  Meisch,  RA.  Ethanol- 
reinforced  responding  of  naive  rhesus 
monkeys:  Acquisition  without  induction 
procedures.  Alcohol 9:547-554,  1992. 

Maurel,  S.;  Schreiber,  R.;  and  De  Vry,  J. 
Substitution  of  the  selective  serotonin 
reuptake  inhibitors  fluoxetine  and  paroxe- 
tine for  the  discriminative  stimulus  effects 
of  ethanol  in  rats.  Psychopharmacology 
(Berl)  130:404^06,  1997. 

Meehan,  S.M.;  Gordon,  T.L.;  and 
Schechter,  M.D.  MDMA  (ecstasy)  substi- 
tutes for  the  ethanol  discriminative  cue  in 
LIAD  but  not  LAD  rats.  Alcohol 
12:569-572,  1995. 

Meisch,  RA.  Ethanol  self- administration: 
Infrahuman  studies.  In:  Thompson,  T., 
and  Dews,  P.B.,  eds.  Advances  in 
Behavioral  Pharmacology.  Vol.  1.  New 
York:  Academic  Press,  1977.  pp.  35-84. 

Melchior,  C.L.;  Glasky,  A.J.;  and 
Ritzmann,  R.F.  A  low  dose  of  ethanol 
impairs  working  memory  in  mice  in  a 
win-shift  foraging  paradigm.  Alcohol 
10:491^93,  1993. 

Miczek,  K.A.;  Mos,  J.;  and  Olivier,  B. 
Brain  5-HT  and  inhibition  of  aggressive 
behavior  in  animals:  5-HIAA  and  receptor 
subtypes.  Psychopharmacol  Bull 
25:399-403,  1989. 

Miczek,  K.A.;  Weerts,  E.M.;  and  DeBold, 
J.F.  Alcohol,  benzodiazepine-GABA, 
receptor  complex  and  aggression: 
Ethological  analysis  of  individual  differ- 
ences in  rodents  and  primates.  J  Stud 
Alcohol  Suppl  11:170-178,  1993. 

Mihic,  S.J.,  and  Harris,  RA.  Alcohol 
actions  at  the  GABAA  receptor/chloride 
channel  complex.  In:  Deitrich,  R.A.,  and 
Erwin,  V.G.,  eds.  Pharmacological  Effects 
of  Ethanol  on  the  Nervous  System.  Boca 
Raton,  FL:  CRC  Press,  1996.  pp.  51-72. 


255 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Mihic,  S.J.;  Ye,  Q.;  Wick,  M.J.;  Koltchine, 
V.V.;  Krasowski,  M.D.;  Finn,  S.E.; 
Mascia,  M.P.;  Valenzuela,  C.F.;  Hanson, 
K.K.;  Greenblatt,  E.P.;  Harris,  R.A.;  and 
Harrison,  N.L.  Sites  of  alcohol  and  volatile 
anaesthetic  action  on  GABAA  and  glycine 
receptors.  Nature  389:385-389,  1997. 

Moy,  S.S.;  Knapp,  D.J.;  Criswell,  H.E.; 
and  Breese,  G.R.  Flumazenil  blockade  of 
anxiety  following  ethanol  withdrawal  in 
rats.  Psychopharmacology  (Berl)  131:354- 
360,  1997. 

Mucha,  R.F.;  van  der  Kooy,  D.; 
O'Shaughnessy,  M.;  and  Bucenieks,  P. 
Drug  reinforcement  studies  by  the  use  of 
place  conditioning  in  rat.  Brain  Res 
243:91-105,  1982. 

Myers,  R.;  Borg,  S.;  and  Mossberg,  R 
Antagonism  of  naltrexone  of  voluntary 
alcohol  selection  in  the  chronically  drinking 
macaque  monkey.  Alcohol 3:383-388, 1986. 

National  Institute  on  Alcohol  Abuse  and 
Alcoholism  (NIAAA).  Alcohol's  Effect  on 
Cognition.  Alcohol  Health  Res  World 
19(2),  1995. 

Ogura,  H.,  and  Aigner,  T.G.  MK-801 
impairs  recognition  memory  in  rhesus  mon- 
keys: Comparisons  with  cholinergic  drugs. 
J  Pharmacol  Exp  Ther  266:60-64,  1993. 

Olivier,  B.;  Mos,  J.;  Tulp,  M.;  Schipper, 
J.;  den  Daas,  S.;  and  van  Oortmerssen,  G. 
Serotonergic  involvement  in  aggressive 
behavior  in  animals.  In:  van  Pragg,  H.M.; 
Plutchik,  R;  and  Apter,  A.,  eds.  Violence 
and  Suicidality.  New  York:  Brunner/ 
Mazel,  1990.  pp.  79-137. 

Overstreet,  D.H.;  Kampov-Polevoy,  A.B.; 
Rezvani,  A.H.;  Murrelle,  L.;  Halikas,  J.A.; 
and  Janowsky,  D.S.  Saccharin  intake 
predicts  ethanol  intake  in  genetically 


heterogeneous  rats  as  well  as  different 
rat  strains.  Alcohol  Clin  Exp  Res  17:366- 
369,  1993. 

Paul,  S.M.,  and  Purdy,  RH.  Neuroactive 
steroids.  FASEB  J 6:231 1-2322,  1992. 

Pellow,  S.;  Chopin,  P.;  File,  S.E.;  and  Briley, 
M.  Validation  of  openxlosed  entries  in  an 
elevated  plus-maze  as  a  measure  of  anxiety 
in  the  rat.  J  Neurosci  Methods  14:149- 
167,  1985. 

Phillips,  A.J.;  Crabbe,  J.C.;  Metten,  P.; 
and  Belknap,  J.K.  Localization  of  genes 
affecting  alcohol  drinking  in  mice.  Alcohol 
Clin  Exp  Res  18:931-941,  1994. 

Pich,  E.M.;  Lorang,  M.;  Yeganeh,  M.;  de 
Fonseca,  F.G.;  Raber,  J.;  Koob,  G.F.;  and 
Weiss,  F.  Increase  of  extracellular  corti- 
cotropin-releasing  factor-like  immunore- 
activity  levels  in  the  amygdala  of  awake 
rats  during  restraint  stress  and  ethanol 
withdrawal  as  measured  by  microdialysis. 
J  Neurosci  15:5439-5447,  1995. 

Pohorecky,  LA.  The  interaction  of  ethanol 
and  stress.  Neurosci  Biobehav  Rev  5:209- 
229,  1981. 

Pohorecky,  L.  A.  Interaction  of  ethanol 
and  stress:  Research  with  experimental 
animals — an  update.  Alcohol  Alcohol 
25:263-276,  1990. 

Preston,  K.L.,  and  Bigelow,  G.E.  Subjective 
and  discriminative  effects  of  drugs.  Behav 
Pharmacol  2:293-313,  1991. 

Rees,  D.C.,  and  Balster,  R.L.  Attenuation 
of  the  discriminative  stimulus  properties 
of  ethanol  and  oxazepam,  but  not  of 
pentobarbital,  by  Ro  15-4513  in  mice./ 
Pharmacol  Exp  Ther  244:592-598,  1988. 

Rex,  A.;  Stephens,  D.N.;  and  Fink,  H. 
"Anxiolytic"  action  of  diazepam  and  abe- 
carnil  in  a  modified  open  field  test.  Pharmacol 
Biochem  Behav  53:1005-1011,  1996. 


256 


Behavioral  Effects  and  Underlying  Neurocircui tries  of  Alcohol 


Risinger,  F.O.  Fluoxetine's  effects  on 
ethanol's  rewarding,  aversive  and  stimulus 
properties.  Life  Sci  61:235-242,  1997. 

Risinger,  F.O.,  and  Cunningham,  C.L.  Eth- 
anol  produces  rapid  biphasic  hedonic  effects. 
Ann  NT  Acad  Sci  654:506-508,  1992. 

Risinger,  F.O.,  and  Cunningham,  C.L. 
Genetic  differences  in  ethanol-induced 
conditioned  taste  aversion  after  ethanol 
pre-exposure.  Alcohol  12:535-539,  1995. 

Risinger,  F.O.,  and  Oakes,  RA.  Mianserin 
enhancement  of  ethanol-induced  condi- 
tioned place  preference.  Behav  Pharmacol 
7:294-298,  1996. 

Risinger,  F.O.;  Dickinson,  S.D.;  and 
Cunningham,  C.L.  Haloperidol  reduces 
ethanol-induced  motor  activity  stimulation 
but  not  conditioned  place  preference. 
Psychopharmacology  107:453-456,  1992«. 

Risinger,  F.O.;  Malott,  D.H.;  Riley,  A.L.; 
and  Cunningham,  C.L.  Effect  of  RO  15- 
4513  on  ethanol-induced  conditioned  place 
preference.  Pharmacol  Biochem  Behav 
43:97-102,  1992£. 

Risinger,  F.O.;  Malott,  D.H.;  Prather, 
L.K.;  Niehus,  D.R.;  and  Cunningham, 
C.L.  Motivational  properties  of  ethanol  in 
mice  selectively  bred  for  ethanol-induced 
locomotor  differences.  Psychopharmacology 
116:207-216,  1994. 

Risinger,  F.O.;  Bormann,  N.M.;  and  Oakes, 
RA.  Reduced  sensitivity  to  ethanol  reward, 
but  not  ethanol  aversion,  in  mice  lacking 
5-HT1B  receptors.  Alcohol  Clin  Exp  Res 
20:1401-1405,  1996. 

Rivier,  C.  Alcohol  stimulates  ACTH 
secretion  in  the  rat:  Mechanisms  of  action 
and  interactions  with  other  stimuli.  Alcohol 
Clin  Exp  Res  20:240-254,  1996. 

Rodgers,  R.J.;  Cole,  J.C.;  Aboualafa,  K.; 
and  Stephenson,  L.H.  Ethopharmacological 


analysis  of  the  effects  of  putative  "anxio- 
genic"  agents  in  the  mouse  elevated  plus 
maze.  Pharmacol  Biochem  Behav  32: 
805-813,  1995. 

Rodin,  E.  Metrazol  tolerance  in  a  "normal" 
volunteer  population.  Electroencephalogr 
Clin  Neurophysiol  10:433-146,  1958. 

Salamone,  J.D.  The  involvement  of  nu- 
cleus accumbens  dopamine  in  appetitive 
and  aversive  motivation.  Behav  Brain  Res 
61:117-133,  1994. 

Samson,  H.H.  Initiation  of  ethanol- 
maintained  behavior:  A  comparison  of 
animal  models  and  their  implication  to 
human  drinking.  In:  Thompson,  T.; 
Dews,  P.B.;  and  Barrett,  J.E.,  eds. 
Neurobehavioral  Pharmacology.  Vol.  6. 
Hillsdale,  NJ:  Lawrence  Erlbaum 
Associates,  1987.  pp.  221-248. 

Samson,  H.H.,  and  Doyle,  T.F.  Oral 
ethanol  self- administration  in  the  rat: 
Effect  of  naloxone.  Pharmacol  Biochem 
Behav  22:9-99,  1985. 

Samson,  H.H.,  and  Hodge,  C.W.  Neuro- 
behavioral regulation  of  ethanol  intake. 
In:  Deitrich,  R.A.,  and  Erwin,  V.G.,  eds. 
Pharmacological  Effects  of  Ethanol  on  the 
Nervous  System.  Boca  Raton,  FL:  CRC 
Press,  1996.  pp.  203-226. 

Sanger,  D.J.  The  effects  of  new  hypnotic 
drugs  in  rats  trained  to  discriminate  ethanol. 
Behav  Pharmacol  8:287-292,  1997. 

Sanger,  D.J.;  Perrault,  G.;  Morel,  E.;  Joly, 
D.;  and  Zivkovic,  B.  The  behavioral  pro- 
file of  Zolpidem,  a  novel  hyponic  drug  of 
imidazopyridine  structure.  Physiol  Behav 
41:235-240,  1987. 

Saudou,  F.;  Amara,  D.A.;  Dierich,  A.; 
LeMeur,  M.;  Ramboz,  S.;  Segu,  L.; 
Buhot,  M.-C;  and  Hen,  R.  Enhanced 
aggressive  behavior  in  mice  lacking  5-HTlb 
receptor.  Science  265:1875-1878,  1994. 


257 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Schechter,  M.D.  Apomorphine  increases 
ethanol  discrimination.  Pharmacol 
Biochem  Behav  22:179-182,  1985. 

Schechter,  M.D.  Locomotor  activity  but 
not  conditioned  place  preference  is  differ- 
entially affected  by  a  moderate  dose  of 
ethanol  administered  to  P  and  NP  rats. 
Alcohol  9:185-188, 1992. 

Schechter,  M.D.  Discriminative  stimulus 
properties  of  isradipine:  Effect  of  other 
calcium  channel  blockers.  Pharmacol 
Biochem  Behav  50:539-543,  1994. 

Schechter,  M.D.,  and  Krimmer,  E.C. 
Differences  in  response  to  the  aversive 
properties  and  activity  of  low  dose  ethanol 
in  LAS  and  HAS  selectively  bred  rats. 
Psychopharmacology  107:564-568,  1992. 

Schuster,  C.R.,  and  Woods,  J.H.  The 
conditioned  reinforcing  effects  of  stimuli 
associated  with  morphine  reinforcement. 
Int  J  Addict  3:223-230,  1968. 

Shelton,  K.L.,  and  Balster,  R.L.  Ethanol 
drug  discrimination  in  rats:  Substitution 
with  GABA  agonists  and  NMDA 
antagonists.  Behav  Pharmacol  5:441- 
450, 1994. 

Sher,  K.J.,  and  Levenson,  R.W.  Risk 
for  alcoholism  and  individual  differences 
in  the  stress-response  dampening  effect 
of  alcohol.  J  Abnorm  Psychol  19:350- 
367,  1982. 

Shively,  C.A.;  Kaplan,  J.R.;  and  Adams, 
M.R.  Effects  of  ovariectomy,  social  insta- 
bility and  social  status  on  female  Macaca 
fascicularis  social  behavior.  Physiol  Behav 
36:1147-1153,  1986. 

Shively,  C.A.;  Manuck,  S.B.;  Kaplan,  J.R.; 
and  Koritnik,  D.R.  Oral  contraceptive 
administration,  interfemale  relationships, 
and  sexual  behavior  in  Macaca  fascicularis. 
Arch  Sex  Behav  19:101-117,  1990. 


Signs,  S.A.,  and  Schechter,  M.D.  The  role 
of  dopamine  and  serotonin  receptors  in  the 
mediation  of  the  ethanol  interoceptive  cue. 
Pharmacol  Biochem  Behav  30:55-64,  1988. 

Simmonds,  MA.  Classification  of  some 
GABA  antagonists  with  regard  to  site  of 
action  and  potency  in  slices  of  cuneate  nu- 
cleus. Eur  J  Pharmacol  80:347-358,  1982. 

Sinclair,  J.D.;  Kampov-Polevoy,  A.;  Stewart, 
R.;  and  Li,  T.-K.  Taste  preferences  in  rat 
lines  selected  for  low  and  high  alcohol 
consumption.  Alcohol  9:155-160,  1992. 

Sinden,  J.D.;  Marfaing-Jallet,  P.;  and 
Magnen,  J.  The  effects  of  naloxone  on  intra- 
gastric ethanol  self- administration.  Pharmacol 
Biochem  Behav  19:1045-1048,  1983. 

Smith,  CM.,  and  Reynard,  A.M.,  eds. 
Essentials  of  Pharmacology.  Philadelphia: 
W.B.  Saunders,  1995. 

Spanagel,  R.  The  influence  of  opioid 
antagonists  on  the  discriminative  stimulus 
effects  of  ethanol.  Pharmacol  Biochem 
Behav  54:645-649, 1996. 

Spealman,  R.D.  Behavior  maintained  by 
termination  of  a  schedule  of  self- adminis- 
tration of  cocaine.  Science  204:1231- 
1233,  1979. 

Stanton,  M.E.,  and  Goodlett,  C.R. 
Neonatal  ethanol  exposure  impairs  eye- 
blink  conditioning  in  weanling  rats. 
Alcohol  Clin  Exp  Res  22:21 '0-275,  1998. 

Stewart,  R.B.;  Gatto,  G.J.;  Lumeng,  L.; 
Li,  T.-K.;  and  Murphy,  J.M.  Comparison 
of  alcohol-preferring  (P)  and  nonprefer- 
ring  (NP)  rats  on  tests  of  anxiety  and  for 
the  anxiolytic  effects  of  alcohol.  Alcohol 
10:1-10,  1993. 

Stewart,  R.B.;  Russell,  R.N.;  Lumeng,  L.; 
Li,  T.-K.;  and  Murphy,  J.M.  Consumption 
of  sweet,  salty,  sour,  bitter  solutions  by 
selectively  bred  alcohol-preferring  P  and 


258 


Behavioral  Effects  and  Underlying  Neurocircuitries  of  Alcohol 


nonpreferring  NP  lines  of  rats.  Alcohol 
Clin  Exp  Res  18:375-381,  1994. 

Stewart,  R.B.;  Bass,  A.A.;  Wang,  N.S.; 
and  Meisch,  R.A.  Ethanol  as  an  oral  rein- 
forcer  in  normal  weight  rhesus  monkeys: 
Dose-response  functions.  Alcohol  13:341- 
346,  1996. 

Stretch,  R;  Gerber,  G.J.;  and  Wood,  S.M. 
Factors  affecting  behavior  maintained  by 
response-contingent  intravenous  infusions 
of  amphetamine  by  squirrel  monkeys.  Can 
J  Physiol  Pharmacol  49:581-589,  1971. 

Sucher,  N.J.;  Awobuluyi,  M.;  Choi,  Y.-B.; 
and  Lip  ton,  S.A.  NMD  A  receptors:  From 
genes  to  channels.  Trends  Pharmacol  Sci 
17:348-355,  1996. 

Szeliga,  K.,  and  Grant,  K.A.  An  analysis  of 
the  5-HT2  receptor  ligands  DOI  and 
ketanserin  in  ethanol  discriminations. 
Alcohol  Clin  Exp  Res  22:646-651,  1998. 

Thompson,  T.,  and  Ostland,  W. 
Susceptibility  to  readdiction  as  a  function 
of  the  addiction  and  withdrawal  environ- 
ments. J  Comp  Physiol  Psychol  60:388- 
392, 1965. 

Tomkins,  D.M.;  Fliggins,  G.A.;  and  Sellers, 
E.M.  Low  doses  of  5-HT1A  agonist  8- 
hydroxy-2-(di-n-propylamino)-tetralin  (8- 
OH  DPAT)  increase  ethanol  intake. 
Psychopharmacology  115:173-179,  1994&. 

Tomkins,  D.M.;  Sellers,  E.M.;  and  Fletcher, 
P.J.  Median  and  dorsal  raphe  injections  of 
the  5-HT1A  agonist  8-OH  DPAT,  and 
the  GABAA  agonist,  muscimol,  increase 
voluntary  ethanol  intake  in  Wistar  rats. 
Neuropharmacology  33:349-358,  1994b. 

Tornatzky,  W.,  and  Miczek,  KA.  Alcohol, 
anxiolytics  and  social  stress  in  rats.  Psycho- 
pharmacology  (Berl)  121:135-144,  1995. 

van  Erp,  A.M.M.,  and  Miczek,  K.A. 
Increased  aggression  after  ethanol  self- 


administration  in  male  resident  rats.  Psycho - 
pharmacology  (Berl)  131:287-295,  1997. 

Wand,  G.S.  Ethanol  differentially  regu- 
lates proadrenocorticotropin/endorphin 
production  and  corticosterone  secretion 
in  LS  and  SS  lines  of  mice.  Endocrinology 
124:518-525,  1989. 

Weerts,  E.M.,  and  Miczek,  K.A.  Primate 
vocalizations  during  social  separation  and 
aggression:  Effects  of  alcohol  and  benzo- 
diazepines. Psychopharmacology  (Berl)  127: 
255-264,  1996. 

Wieland,  S.;  Lan,  N.C.;  Mirasedeghi,  S.; 
and  Gee,  K.W.  Anxiolytic  activity  of  the 
progesterone  metabolite  5a-pregnan-3a- 
ol-20-one.  Brain  Res  565:263-268,  1991. 

Weiss,  B.,  and  Cory-Slechta,  DA. 
Assessment  of  behavioral  toxicity.  In: 
Hayes,  A.W.,  ed.  Principles  and  Methods 
of  Toxicology.  3d  ed.  New  York:  Raven 
Press,  1994.  pp.  1091-1155. 

Weiss,  F.;  Lorang,  M.T.;  Blom,  F.E.;  and 
Koob,  G.F.  Oral  alcohol  self- administra- 
tion stimulates  dopamine  release  in  the  rat 
nucleus  accumbens:  Genetic  and  motiva- 
tional determinants.  J  Pharmacol  Exp  Ther 
267:250-258,  1993. 

Wickelgren,  I.  Getting  the  brain's  atten- 
tion. Science  278:35-37,  1997. 

Williams,  A.F.  Social  drinking,  anxiety 
and  depression.  J Pers  Soc  Psychol  3:689- 
693,  1966. 

Williams,  K.L.;  Winger,  G.;  Pakarinen,  E.P.; 
and  Woods,  J.H.  Naltrexone  reduces 
ethanol  and  sucrose  reinforced  responding 
in  rhesus  monkeys.  Psychopharmacology 
(Berl)  139:53-61,  1998. 

Wilner,  P.,  and  Scheel-Kruger,  J.,  eds. 
The  Mesolimbic  Dopamine  System:  From 
Motivation  to  Action.  New  York:  John 
Wiley  &  Sons,  1991. 


259 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Winter,  J.C.  Morphine  and  ethanol  as 
discriminative  stimuli:  Absence  of  antago- 
nism by  p-chloro  phenylalanine  methyl 
ester,  cinanserin,  or  BC-105. 
Psychopharmacolqgy  53:159-163,  1977. 

Wise,  R.A.;  Bauco,  P.;  Carlezon,  W.A.; 
and  Trojniar,  W.  Self-stimulation  and 
drug  reward  mechanisms.  In:  Kalivas, 


P.W.,  and  Samson,  H.H.,  eds.  The 
Neurobiology  of  Drug  and  Alcohol 
Addiction.  [Special  Issue.]  Ann  NT  Acad 
Set  654:192-198,  1992. 

Woods,  J.H.;  Downs,  D.A.;  and  Carney, 
J.  Behavioral  functions  of  narcotic  antago- 
nists: Response-drug  contingencies. 
Federation  Proc  34:1777-1784,  1975. 


260 


Chapter  7 

Neuroadaptive  Changes  in 

Neurotransmitter  Systems  Mediating 

Ethanol-Induced  Behaviors 

Friedbert  Weiss,  Ph.D. 


KEY  WORDS:  AOD  (alcohol  or  other  drug)  dependence;  AOD  withdrawal  syn- 
drome; animal  model;  neurobiological  theory;  neurochemistry;  AODD  (AOD  use 
disorder)  relapse;  AOD  abstinence;  reinforcement;  chronic  AODE  (effects  of  AOD 
use,  abuse,  and  dependence);  AOD  use  behavior;  neurotransmitters;  AOD  sensi- 
tivity; compulsion;  literature  review 


This  review  will  highlight  current  under- 
standing of  the  neurobiologies  basis  of 
ethanol  dependence,  compulsive  ethanol- 
seeking  behavior  associated  with  depen- 
dence, and  persistent  neuroadaptive 
changes  in  animals  with  a  history  of 
dependence  that  may  motivate  relapse 
and  perpetuate  alcohol  abuse. 

MOTIVATIONAL  SIGNIFI- 
CANCE OF  ETHANOL 
DEPENDENCE  AND 
WITHDRAWAL 

Theoretical  Perspectives  on 
Addiction  and  Compulsive 
Ethanol- Seeking  Behavior 

Recent  views  in  addiction  theory 


involve  the  recognition  of  adaptations 
within  the  central  nervous  system 
(CNS).  These  adaptations  induced  by 
chronic  drug  use  are  thought  to  result 
in  a  disruption  or  desensitization  of 
the  neural  mechanisms  that  mediate 
reward  (Koob  and  Bloom  1988;  Koob 
et  al.  1993;  Koob  1996;  Wise  1996). 
This  view  is  concerned  not  so  much 
with  physical  withdrawal  symptoms  as  a 
motivating  factor  in  continued  drug  tak- 
ing but  rather  with  symptoms  that  result 
from  a  compromised  state  of  the  reward 
system,  which  leads  to  affective  states 
(e.g.,  dysphoria,  depression,  and  anxi- 
ety) that  are  opposite  to  the  initial 
mood-elevating  effects  of  drugs.  The 
significance  of  this  emerging  view  of 
dependence  is  that  it  identifies  a  single 


F.  Weiss,  Ph.D.,  is  an  associate  professor  in  the  Department  of  Neuropharmacology,  CVN-15,  The 
Scripps  Research  Institute,  10550  North  Torrey  Pines  Rd.,  Lajolla,  CA  92037. 


261 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


motivational  withdrawal  effect,  negative 
affect,  rather  than  a  constellation  of  aver- 
sive,  mostly  physical  withdrawal  events 
in  continued  and  escalating  drug-seeking 
behavior,  and  it  suggests  a  common 
basis  for  withdrawal  from  many,  if  not 
all,  classes  of  drugs  of  abuse. 

Another  form  of  neuroadaptation, 
sensitization,  has  received  growing  atten- 
tion as  a  possible  mechanism  in  compul- 
sive drug  use.  Sensitization  involves  a 
dramatic  augmentation  of  behavioral 
and  neurochemical  responses  associated 
predominantly  with  meso-accumbens 
dopamine  (DA)  transmission  that  often 
develops  with  chronic,  intermittent  expo- 
sure to  drugs  of  abuse  (e.g.,  Robinson 
and  Becker  1986;  Kalivas  and  Stewart 
1991;  Robinson  and  Berridge  1993). 
One  of  the  most  prominent  theoretical 
positions  on  the  significance  of  drug- 
induced  sensitization  holds  that  compul- 
sive drug-seeking  behavior  is  the  result  of 
a  progressive  and  persistent  hypersen- 
sitivity of  neural  systems  that  mediate 
"incentive  salience,"  resulting  in  a 
transformation  of  ordinary  "wanting" 
into  excessive  craving  (Robinson  and 
Berridge  1993).  According  to  this  view, 
this  process  can  occur  independently 
of  changes  in  neural  systems  that  medi- 
ate the  subjective  pleasurable  effects  of 
drugs  or  systems  that  mediate  with- 
drawal. Thus,  this  view  does  not  neces- 
sarily rule  out  the  possibility  that  other 
elements  of  the  drug  response,  such  as 
mood  elevation  or  anxiolysis,  undergo 
tolerance  or  become  desensitized. 

Ethanol  Dependence  and 
Withdrawal 

The  physical  symptoms  that  accom- 
pany ethanol  withdrawal  have  been 


well  characterized  and  include  auto- 
nomic hyperactivity  such  as  hyperten- 
sion and  increased  heart  rate,  neuronal 
hyperexcitability  with  tremor  and 
seizures,  and  perceptual  distortions 
such  as  hallucinations  and  delirium 
(e.g.,  Rosenblatt  et  al.  1972;  Cas- 
taneda  and  Cushman  1989;  Turner  et 
al.  1989;  Benzer  1994;  O'Brien 
1996).  Animal  models  exist  for  these 
different  ethanol  withdrawal  symp- 
toms, and  physical  signs  have  been 
used  to  study  the  neural  basis  of  the 
alcohol  withdrawal  syndrome  (Meert 
and  Huysmans  1994).  The  neuro bio- 
logical basis  for  these  physical  signs  of 
ethanol  withdrawal  involves  CNS 
rebound  hyperexcitability,  such  as  a 
decrease  in  the  function  of  the 
inhibitory  amino  acid  transmitter 
gamma-aminobutyric  acid  (GABA) 
receptors  or  increases  in  the  function 
of  the  excitatory  transmitter  glutamate 
and  its  receptors  (Grant  et  al.  1990; 
Morrisett  et  al.  1990;  Hoffman  and 
Tabakoff  1994).  More  recently  other 
neurotransmitter  and  neuromodula- 
tory  systems,  including  serotonin,  DA, 
norepinephrine,  adenosine,  ganglio- 
sides,  and  neurosteroids,  have  been 
implicated  in  neural  hyperexcitability 
associated  with  ethanol  withdrawal 
(Crabbe  1992;  Concas  et  al.  1994; 
Hoffman  and  Tabakoff  1994;  Meert 
1994;  Adams  et  al.  1995;  Finn  et  al. 
1995;  Kotlinska  and  Liljequist  1996; 
Snell  et  al.  1996;  Riihioja  et  al.  1997). 
In  addition  to  these  physiological 
symptoms,  ethanol  withdrawal  has  a 
variety  of  behavioral  and  affective  con- 
sequences that  are  more  "motiva- 
tional" in  nature.  Acute  withdrawal 
from  ethanol  is  associated  with  a  neg- 


262 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


ative  affective  state  consisting  of  dys- 
phoria, depression,  irritability,  and 
anxiety  (e.g.,  Bjorkqvist  1975;  Moss- 
berg  et  al.  1985;  Turner  et  al.  1989; 
Bockstrom  and  Balldin  1992;  O'Brien 
1996),  which  appear  independent  of 
physical  signs  of  withdrawal.  These 
affective  withdrawal  symptoms  may 
motivate  resumption  of  drinking 
(Koob  and  Bloom  1988;  Koob  et  al. 
1993;  Wise  1996),  and  by  ameliorat- 
ing these  symptoms  ethanol  could 
then  serve  as  a  negative  reinforcer  for 
continued  alcohol  use  and  abuse.  Two 
major  categories  of  withdrawal 
responses  reflecting  the  motivational 
aspects  of  ethanol  withdrawal  will  be 
the  focus  of  this  review:  changes  in 
reward  function  and  anxiogenic  con- 
sequences. 

Behavioral  and  Reward  Functions 
in  Dependence  and  Withdrawal 

Upon  acute  administration,  ethanol 
has  anxiolytic  and  mild  euphorigenic 
effects,  which  are  thought  to  be  cen- 
tral to  its  reinforcing  properties  and 
abuse  potential.  These  actions  have 
been  well  documented  in  animal 
behavioral  models.  For  example,  like 
other  drugs  of  abuse,  ethanol  stimu- 
lates locomotor  activity  (Waller  et  al. 
1986;  Lewis  and  June  1990;  Wolff- 
gramm  1991;  Broadbent  et  al.  1995; 
Cohen  et  al.  1997)  and  can  lower  the 
thresholds  for  intracranial  self-stimula- 
tion (ICSS)  under  relevant  dosing  and 
treatment  conditions  (Bain  and  Kor- 
netsky  1989;  Lewis  and  June  1990; 
Moolten  and  Kornetsky  1990;  Lewis 
1991).  Both  stimulation  of  locomotor 
activity  and  facilitation  of  brain  stimu- 
lation reward  are  generally  thought  to 


reflect  activation  of  the  mesolimbic 
DA  system  and  are  good  predictors  of 
rewarding  properties  and  abuse  liabil- 
ity. Similarly,  like  classical  anxiolytic 
drugs,  ethanol  exerts  antianxiety 
effects  in  several  behavioral  models. 
Ethanol  attenuates  suppression  of 
exploratory  activity  in  the  elevated 
plus  maze  (Pellow  and  File  1986;  Lis- 
ter 1987)  and  social  interaction  test 
(Lister  1988;  File  1980)  and  can 
effectively  reverse  behavioral  suppres- 
sion in  conflict  tests  (Koob  and  Brit- 
ton  1996).  Although  these  positive 
affective  responses  are  considered  a 
critical  factor  in  the  positive  reinforc- 
ing effects  of  ethanol,  evidence  is 
accumulating  to  suggest  that,  with 
chronic  exposure  and  dependence, 
adaptive  processes  within  the  CNS 
develop  that  oppose  the  acute  rein- 
forcing actions  of  drugs,  leading  to 
the  emergence  of  affective  changes  in 
the  absence  of  the  drug. 

Negative  Affect:  Anxiogenic 
Responses  and  Reward  Deficits  During 
Withdrawal.  In  humans,  chronic  alco- 
hol use  and  alcohol  withdrawal  pro- 
duce anxiety,  and  these  symptoms  can 
persist  long  after  physical  withdrawal 
and  detoxification  (Bjorkqvist  1975; 
Mossberg  et  al.  1985;  Roelofs  1985; 
Bockstrom  and  Balldin  1992).  In  ani- 
mals, anxiogenic-like  consequences  of 
withdrawal  have  been  extensively  docu- 
mented by  suppression  of  exploratory 
activity  on  the  unprotected  arms  of 
the  elevated  plus  maze  (e.g.,  Baldwin 
et  al.  1991;  File  et  al.  1991;  Prather  et 
al.  1991;  File  et  al.  1993;  Rassnick  et 
al.  19936;  Moy  et  al.  1997;  Watson  et 
al.  1997).  Ethanol  withdrawal  anxiety 
has  also  been  demonstrated  in  drug 


263 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


discrimination  studies  where  rats 
undergoing  ethanol  withdrawal 
selected  a  lever  previously  associated 
with  the  interoceptive  effects  of  the 
anxiogenic  compound  pentylenetetra- 
zol (PTZ),  and  this  effect  was  reversed 
by  administration  of  ethanol  (Lai  et  al. 
1988).  Interestingly,  the  generaliza- 
tion to  PTZ  during  ethanol  with- 
drawal occurred  before  the  onset  of 
overt  signs  of  withdrawal.  This  obser- 
vation is  indicative  of  a  dissociation 
between  physical  and  affective-motiva- 
tional withdrawal  effects,  and  suggests 
that  the  anxiogenic  consequences  of 
ethanol  withdrawal  may  play  an 
important  role  in  the  maintenance  of 
ethanol  abuse  independent  of  physical 
withdrawal  distress. 

Evidence  is  also  accumulating  that 
chronic  use  of  ethanol  compromises 
neural  mechanisms  that  mediate  posi- 
tive reinforcement.  This  is  illustrated 
by  the  finding  that  in  contrast  to  the 
acute  effects  of  ethanol  on  ICSS 
thresholds,  withdrawal  in  dependent 
animals  leads  to  a  significant  impair- 
ment in  the  rewarding  efficacy  of  elec- 
trical brain  stimulation  that  lasts  up  to 
48  hours  after  termination  of  expo- 
sure to  ethanol  (Schulteis  et  al.  1995). 
This  withdrawal-associated  reward 
deficit  is  similar  to  that  induced  by  all 
other  major  drugs  of  abuse,  including 
opiates,  tetrahydrocannabinol,  psy- 
chostimulants, and  nicotine  (Markou 
and  Koob  1991;  Legault  and  Wise 
1994;  Schulteis  et  al.  1995),  and  may 
reflect  both  adaptations  within  the 
mesolimbic  DA  system,  which  has 
been  implicated  in  mediating  the  posi- 
tive reinforcing  actions  of  alcohol  and 
other  drugs  (AODs)  (Leith  and  Bar- 


rett 1976;  Markou  and  Koob  1991; 
Parsons  et  al.  1995;  Pich  et  al.  1997), 
and  the  recruitment  of  brain  stress 
systems.  Like  the  anxiogenic  effects  of 
withdrawal,  ICSS  reward  deficits  are 
observed  well  before  the  onset  of 
overt  physical  signs  of  ethanol  with- 
drawal (Schulteis  et  al.  1995),  con- 
firming a  dissociation  between 
affective -motivational  and  physical 
withdrawal  symptoms. 

Together,  these  data  suggest  that 
while  physical  withdrawal  symptoms 
have  a  role  in  the  aversive  aspects  of 
ethanol  withdrawal,  withdrawal- 
induced  anxiety  and  reward  deficits 
may  be  critical  in  the  maintenance  of 
alcoholism.  Since  these  consequences 
of  withdrawal  accrue  well  before  the 
emergence  of  physical  withdrawal 
symptoms,  they  may  motivate  contin- 
ued or  increased  ethanol  consumption 
to  avoid  their  occurrence  and  thereby 
contribute  to  the  negative  reinforcing 
properties  of  ethanol. 

Sleep.  Ethanol  withdrawal  is  also 
associated  with  marked  sleep  and  cir- 
cadian  disturbances.  Alcoholics  show 
less  total  sleep  time  during  acute  with- 
drawal, with  reductions  in  both  rapid 
eye  movement  (REM)  and  non-REM 
sleep  (Gillin  et  al.  1990;  Thompson  et 
al.  1995),  and  these  sleep  disturbances 
seem  coupled  to  a  suppression  of 
melatonin  secretion  (Schmitz  et  al. 
1996).  Disruptions  in  circadian  vigi- 
lance states,  in  particular  reduced 
REM  sleep,  have  also  been  reported 
during  withdrawal  in  ethanol-depen- 
dent  rats  (Rouhani  et  al.  1990).  With- 
drawal-related circadian  and  sleep 
abnormalities  may  have  an  important 
role  in  the  susceptibility  to  relapse 


264 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


during  withdrawal  and  the  protracted 
abstinence  phase.  However,  little  is 
known  about  the  persistence  and 
long-term  consequences  of  these  dys- 
functions. This  is  an  area  of  active 
research  (Viglinskaya  1992;  Peter  et 
al.  1995;  Brower  et  al.  1998;  Clark  et 
al.  1998;  Drummond  et  al.  1998; 
Mackenzie  et  al.  1999)  that  promises 
to  provide  a  better  understanding  of 
homeostatic  disturbances  in  the  main- 
tenance of  ethanol  abuse  habits. 

ANIMAL  MODELS  OF 
EXCESSIVE  DRINKING 
AND  DEPENDENCE 

Alcoholism,  by  definition,  involves 
compulsive  and  excessive  use  of  alcohol 
(ethanol).  Thus,  the  concepts  of  rein- 
forcement and  motivation  are  crucial 
to  the  understanding  of  this  syn- 
drome, and  ethanol  self- administra- 
tion and  ethanol-seeking  behavior 
have  emerged  as  the  primary  behav- 
ioral measure  of  interest  in  contempo- 
rary research  on  alcohol  abuse  and 
addiction  (Grant  1995).  Two  princi- 
pal categories  of  motivated  behavior 
supported  by  ethanol  can  be  distin- 
guished: (1)  a  consummatory  aspect 
where  drinking  is  reinforced  and 
maintained  by  the  rewarding  conse- 
quences resulting  from  the  consumption 
of  ethanol  and  (2)  an  incentive-motiva- 
tional aspect  which  elicits  and  maintains 
behavior  that  brings  the  organism  into 
contact  with  the  reinforcing  stimulus 
(i.e.,  ethanol).  The  immediate,  pharma- 
cological effects  of  ethanol  are  thought 
to  maintain  consumption  through 
positive  or  negative  reinforcement 
of  the  drinking  habit,  where  negative 


reinforcement  would  involve  self- 
medication  of  an  existing  aversive  state 
or  self- medication  of  a  drug-generated 
aversive  state  such  as  withdrawal 
(Wilder  1973).  The  incentive-motiva- 
tional aspects  of  ethanol-seeking 
behavior,  on  the  other  hand,  involve 
association  of  previously  neutral  stim- 
uli with  either  the  pleasurable  subjec- 
tive effects  of  ethanol  or  relief  from 
the  aversive  effects  of  withdrawal,  and 
these  aspects  are  thought  to  be 
involved  in  the  initiation  of  alcohol- 
seeking  behavior,  craving,  and  relapse. 
Reliable  self- administration  models 
have  been  established  for  measuring 
the  positive  reinforcing  effects  of 
ethanol  (Samson  1986,  1987)  and, 
more  recently,  the  negative  reinforc- 
ing effects  of  ethanol  self- administra- 
tion (Roberts  et  al.  1996;  Schulteis  et 
al.  1996).  However,  few  effective 
models  suited  for  the  study  of  incen- 
tive-motivational effects  of  ethanol 
and  their  role  in  relapse  are  currently 
available.  Efforts  to  develop  such 
models  are  under  way  (Heyser  and 
Koob  1997;  Katner  et  al.  1999)  and 
should  eventually  aid  greatly  in  the 
investigation  of  the  neurobiological 
basis  of  ethanol  craving  and  relapse. 

Ethanol  Self-Administration 
During  Withdrawal 

As  already  pointed  out,  the  consump- 
tion of  ethanol,  not  simply  for  its 
euphorigenic  effects  but  to  avoid  or 
reverse  symptoms  of  withdrawal,  may 
be  an  important  factor  in  the  perpetu- 
ation of  ethanol  dependence  (for 
reviews,  see  Cappell  1981;  Edwards 
1990).  Indeed,  withdrawal  symptoms, 
in  particular  depression  and  anxiety, 


265 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


were  found  to  provoke  drinking  in  83 
out  of  100  male  alcoholics  (Hershon 
1977).  However,  it  has  traditionally 
been  difficult  to  demonstrate  that 
ethanol  withdrawal  motivates  ethanol- 
seeking  behavior  in  animals.  Numer- 
ous studies  indicate  that  the  mere 
induction  of  physical  dependence  is 
not  sufficient  to  promote  ethanol 
intake,  because  ethanol-dependent 
rats  and  monkeys  subjected  to  with- 
drawal often  refuse  to  consume 
ethanol  even  though  ethanol  con- 
sumption would  alleviate  withdrawal 
distress  (Veale  and  Myers  1969;  Myers 
et  al.  1972;  Begleiter  1975;  Cicero 
1980;  Meisch  1984;  Winger  1988; 
Samson  and  Harris  1992;  Meisch  and 
Stewart  1994).  More  positive  results 
have,  however,  been  obtained  in  rats 
with  procedures  that  either  bypass  the 
aversive  taste  effects  of  ethanol  or  pro- 
vide for  repeated  opportunities  to 
associate  ethanol  consumption  with 
the  alleviation  of  withdrawal  symp- 
toms (Deutsch  and  Koopmans  1973; 
Hunter  et  al.  1974;  Samson  and  Falk 
1974;  Deutsch  and  Walton  1977; 
Trapold  and  Sullivan  1979). 

It  has  been  more  conclusively  estab- 
lished that  ethanol  can  serve  as  a  potent 
negative  reinforcer  in  dependent  ani- 
mals undergoing  withdrawal  (Roberts 
et  al.  1996;  Schulteis  et  al.  1996;  Weiss 
et  al.  1996).  Specifically,  rats  made 
dependent  on  ethanol  via  a  liquid-diet 
procedure  were  shown  to  operantly 
respond  for  oral  ethanol  during  with- 
drawal, and  these  animals  consumed 
significantly  more  ethanol  than  non- 
dependent  rats  (Schulteis  et  al.  1996; 
Weiss  et  al.  1996).  An  even  more  strik- 
ing demonstration  of  the  motivational 


effects  of  ethanol  dependence  is  the 
finding  that  rats  given  access  to  oral 
ethanol  in  an  operant  self- administra- 
tion task  immediately  after  removal 
from  chronic  ethanol  vapor  exposure 
responded  for  ethanol  in  a  manner 
that  maintained  blood  alcohol  levels 
(BALs)  in  excess  of  100  mg%  over  a 
12-hour  "withdrawal"  period  and  pre- 
vented withdrawal  symptoms  present 
in  dependent  rats  not  given  access  to 
ethanol  during  the  withdrawal  phase 
(Roberts  et  al.  1996).  Interestingly, 
responding  for  ethanol  became  more 
stable  over  the  course  of  four  repeated 
withdrawal  episodes,  suggesting  not 
only  that  ethanol  became  more  firmly 
established  as  a  negative  reinforcer  but 
also  that  rats  learn  to  regulate  ethanol 
intake  in  a  manner  that  stabilizes 
BALs  and  minimizes  or  prevents  with- 
drawal discomfort. 

In  comparing  the  positive  reports 
with  studies  that  have  failed  to 
observe  ethanol  intake  during  with- 
drawal, it  appears  that  procedures 
designed  to  overcome  the  aversive 
taste  cues  of  ethanol  and  the  opportu- 
nity to  learn  that  ethanol  consumption 
can  alleviate  withdrawal  discomfort 
are  essential  for  the  demonstration  of 
reinforcing  effects  of  ethanol  during 
withdrawal.  An  additional  factor  in 
contributing  to  the  differences 
between  these  studies  may  be  with- 
drawal severity.  Severe  tremors  and 
seizures  such  as  those  reported  in  ear- 
lier work  with  intragastric  intubation 
or  forced  consumption  of  high 
ethanol  concentrations  (Veale  and 
Myers  1969;  Myers  et  al.  1972; 
Begleiter  1975;  Winger  1988)  are 
rarely  observed  with  the  liquid-diet 


266 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


procedure  or  moderate  ethanol  vapor 
exposure  in  the  more  recent  studies 
(Majchrowicz  1975;  Lai  et  al.  1988; 
Emmett-Oglesby  et  al.  1990;  Baldwin 
et  al.  1991;  Rassnick  et  al.  1992«; 
Merlo  Pich  et  al.  1995;  Macey  et  al. 
1996).  It  is,  therefore,  possible  that 
milder  forms  of  withdrawal  may  more 
readily  support  ethanol  self- adminis- 
tration, whereas  severe  withdrawal 
distress  may  have  general  inhibitory 
effects  on  behavior,  including  self- 
administration  (Winger  1988;  Meisch 
and  Stewart  1994),  and  thereby  retard 
learning  that  consumption  of  ethanol 
can  alleviate  withdrawal. 

Ethanol  Self- Administration 
After  Periods  of  Abstinence 

Early  experiments  revealed  that  rats 
show  marked  increases  in  voluntary 
ethanol  consumption  after  periods  of 
forced  abstinence  (LeMagen  1960; 
Sinclair  and  Senter  1967,  1968;  Sinclair 
1972,  1979).  This  so-called  alcohol 
deprivation  effect  has  since  been  con- 
firmed in  mice  (Salimov  and  Salimova 
1993),  rats  (Wolffgramm  and  Heyne 
1995;  Spanagel  et  al.  1996;  Heyser  et 
al.  1997;  Holter  et  al.  1997),  mon- 
keys (Kornet  et  al.  1990,  1991),  and 
human  social  drinkers  (Burish  et  al. 
1981),  and  it  is  well  established  as  a 
robust  and  reliable  phenomenon  in 
animal  models  of  alcohol  drinking. 

The  alcohol  deprivation  effect  can 
be  readily  demonstrated  in  nondepen- 
dent  animals  and  may  provide  a 
potentially  valuable  model  for  under- 
standing changes  in  the  reinforcing 
efficacy  of  ethanol  that  occur  with 
abstinence  (e.g.,  Heyser  et  al.  1996, 
1997;  Holter  et  al.  1997).  More 


importantly,  however,  under  appro- 
priate conditions,  this  phenomenon 
appears  to  become  resistant  to  manip- 
ulations of  ethanol  concentration, 
taste,  and  environmental  factors 
(Wolffgramm  and  Heyne  1995; 
Spanagel  et  al.  1996)  and,  therefore, 
may  prove  useful  as  a  model  for  com- 
pulsive ethanol-seeking  behavior  and 
loss  of  control  that  characterize  sub- 
stance dependence  on  alcohol  (per 
DSM-IV  [American  Psychiatric  Asso- 
ciation 1994]).  Studies  that  have  char- 
acterized the  alcohol  deprivation 
effect  in  rats  given  long-term  (8  to  24 
months)  continuous  free  access  to  dif- 
ferent concentrations  of  ethanol  and 
water,  interspersed  with  deprivation 
periods  of  varying  lengths,  indicate 
that  ethanol  consumption  increases 
significantly  over  baseline  as  a  result  of 
deprivation  (Wolffgramm  and  Heyne 
1995;  Spanagel  et  al.  1996),  reaching 
levels  of  intake  similar  to  those  in  rats 
selectively  bred  for  alcohol  preference 
(Li  et  al.  1979).  The  increase  in 
ethanol  intake  associated  with  alcohol 
deprivation  was  characterized  not  only 
by  enhanced  preference  for  ethanol 
over  water  but  also  by  preference  for 
higher  ethanol  concentrations  (>  10 
percent  v/v)  and  resistance  to  change 
by  altering  the  palatability  of  the 
ethanol  solution  (by  either  quinine  or 
sucrose)  or  by  manipulating  environ- 
mental and  social  conditions  (such  as 
isolation  housing  or  changing  domi- 
nance hierarchies)  (Wolffgramm  and 
Heyne  1991,  1995;  Spanaget  et  al. 
1996).  Moreover,  ethanol  deprivation 
under  these  exposure  conditions  revealed 
a  behavioral  withdrawal  syndrome,  as 
measured  by  lowered  thresholds  of 


267 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


footshock  reactivity,  which  reached 
maximum  on  the  2d  day  of  abstinence 
and  persisted  for  up  to  5  days  post- 
ethanol  (Heyne  et  al.  1991). 

Finally,  the  alcohol  deprivation 
effect  appears  to  outlast  long  abstinence 
phases  (Spanagel  et  al.  1996).  Indeed, 
it  has  been  suggested  that  this  effect 
is  irreversible  since  it  remained  unaltered 
after  9  months  of  abstinence  (Wolff- 
gramm  1991;  Wolffgramm  and  Heyne 
1991,  1995).  In  particular,  the  loss  of 
reversibility  of  this  effect  and  the 
reduced  adaptability  of  ethanol-seek- 
ing  behavior  in  response  to  environ- 
mental or  taste  manipulations  suggest 
that  these  procedures  may  provide  an 
effective  model  to  study  mechanisms 
underlying  specific  aspects  of  ethanol- 
maintained  addictive  behavior  and  loss 
of  control.  As  discussed  later  in  this 
chapter,  measures  of  the  alcohol 
deprivation  effect  may  also  offer 
promise  as  a  tool  to  study  aspects  of 
the  relapse  process  in  dependent  and 
postdependent  animals. 

Selective  Breeding  for  High 
Ethanol  Intake 

Several  lines  of  rats  genetically  selected 
for  traits  of  ethanol  aversion  or  self- 
selection  have  been  developed  to 
model  voluntary  excessive  drinking 
and  alcohol  abuse  (Li  et  al.  1986; 
Kiianmaa  et  al.  1992;  Li  and  McBride 
1995);  these  lines  fulfill  many  of  the 
established  criteria  as  animal  models 
of  alcohol  abuse  (Lester  and  Freed 
1973).  Lines  that  have  been  best  char- 
acterized behaviorally  and  neuro- 
chemically  include  the  Indiana  P/NP, 
HAD/LAD,  and  Alko  AA/ANA  rats 
(Murphy  et  al.  1982;  Li  et  al.  1986; 


Murphy  et  al.  1987;  Gongwer  et  al. 
1989;  McBride  et  al.  19906;  Kiianmaa 
and  Saito  1991;  Kiianmaa  et  al.  1991; 
Gianoulakis  et  al.  1992;  Kiianmaa  et 
al.  1992;  McBride  et  al.  1992).  Simi- 
larities exist  among  these  lines  in 
ethanol-related  behavioral  and  physi- 
ological characteristics,  including  the 
development  of  rapid  tolerance,  volun- 
tary 24-hour  ethanol  intake,  and  acqui- 
sition of  ethanol-reinforced  operant 
behavior.  Common  neurochemical 
markers  of  preference  that  have  been 
identified  to  date  include  abnormali- 
ties in  the  function  of  forebrain  DA 
and  5-hydroxytryptamine  (serotonin 
or  5-HT)  neurotransmission  in  the 
Indiana  P  and  FIAD  lines  (Murphy  et 
al.  1982,  1987;  Gongwer  et  al.  1989; 
McBride  et  al.  1990#),  and  heightened 
sensitivity  to  the  DA  release-enhancing 
and  locomotor  activating  effects  of 
ethanol  in  the  P  and  Sardinian  alcohol- 
preferring  (sP)  lines  (Fadda  et  al. 
1980;  Waller  et  al.  1986;  Cloninger 
1987;  Engel  et  al.  1992;  Weiss  et  al. 
1993).  These  neurochemical  abnor- 
malities involve  the  same  neural  sys- 
tems that  have  been  implicated  in 
neuroadaptive  changes  associated  with 
chronic  ethanol  consumption  (as  dis- 
cussed later  in  this  chapter),  suggest- 
ing that  both  environmental  and 
genetic  factors  can  converge  to  drive 
excessive  drinking. 

In  spite  of  these  neurochemical 
commonalities  and  the  existence  of 
certain  behavioral  similarities  among 
lines  of  alcohol-preferring  rats,  the 
neurochemical  mechanisms  underlying 
ethanol  preference  remain  unclear  in 
that  it  has  been  difficult  to  demonstrate 
common  neurochemical  markers  of 


268 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


ethanol  preference  across  multiple 
selected  lines  (e.g.,  Murphy  et  al.  1982, 
1987;  Gongwer  et  al.  1989  vs.  Sinclair 
et  al.  1989;  Kiianmaa  et  al.  1991).  In 
contrast  to  the  P  and  HAD  lines,  no 
reductions  and  even  elevated  levels  of 
DA  and  5-HT  have  been  found  in 
limbic  and  cortical  forebrain  regions 
of  alcohol-preferring  AA  compared 
with  nonp referring  ANA  rats  (Sinclair 
et  al.  1989;  Kiianmaa  et  al.  1991). 
Similarly,  AA  rats  show  lower  con- 
tents of  (3 -endorphin  in  the  periaque- 
ductal gray  and  amygdala  than  ANA 
rats  (Gianoulakis  et  al.  1992),  and  P 
rats  have  increased  levels  of  met- 
enkephalin  in  the  hypothalamus  and 
striatum  relative  to  NP  rats  (Froehlich 
etal.  1987). 

Additionally,  though  there  is  good 
consistency  across  alcohol-preferring 
lines  in  24-hour  home  cage  ethanol 
intake,  differences  exist  among  these 
animals  in  other  ethanol-related 
behaviors.  These  include  differences 
in  the  magnitude  of  the  alcohol  depri- 
vation effect  (Sinclair  and  Tiihonen 
1988;  Sinclair  and  Li  1989),  the 
degree  to  which  availability  of  a  palat- 
able alternative  fluid  attenuates 
ethanol  intake  (Lankford  et  al.  1991; 
Lankford  and  Myers  1994)  and,  in 
particular,  ethanol-reinforced  operant 
responding.  Specifically,  in  operant 
self- administration  tasks  ethanol  can 
serve  as  a  reinforcer  in  NP  rats,  which 
avoid  ethanol  in  a  24 -hour  preference 
test  (Files  et  al.  1993;  Rassnick  et  al. 
1993#;  Ritz  et  al.  1994).  Moreover, 
when  the  response  requirements  for 
ethanol  are  increased,  NP  rats  are  will- 
ing to  "work  harder"  than  HAD  rats, 
a  line  that  shows  high  ethanol  intake 


in  preference  tests  (Ritz  et  al.  1994). 
A  reduced  "willingness"  to  work  for 
ethanol  under  higher  response  require- 
ments has  also  been  reported  in  the 
alcohol-preferring  AA  rats  (Ritz  et  al. 
1989).  Only  in  P  rats  was  ethanol  intake 
in  operant  tests  consistent  with  their 
high  ethanol  consumption  in  24-hour 
preference  tests.  These  studies  suggest 
that  inherited  factors  that  determine 
whether  ethanol  will  come  to  serve  as 
a  reinforcer  per  se  (i.e.,  in  preference 
tests)  differ  from  those  that  mediate 
the  motivating  value  of  ethanol,  as 
inferred  from  the  amount  of  work 
that  rats  will  expend  to  gain  access  to 
the  drug.  Findings  such  as  these  indicate 
that  it  will  be  important  to  incorporate 
animals'  "motivation"  or  degree  of 
intensity  and  persistence  in  the  effort 
to  obtain  ethanol  at  a  particular  time 
or  set  of  circumstances  in  pharmaco- 
genetic  models  of  high  ethanol  intake. 

MECHANISMS  OF 
REINFORCEMENT 
ASSOCIATED  WITH 
CHRONIC  DRINKING 

Compromised  Reward  Systems 
and  Negative  Reinforcement 

As  discussed  earlier  in  this  chapter, 
measures  of  ICSS  reward  thresholds 
indicate  that,  in  contrast  to  the  acute 
effects  of  ethanol,  withdrawal  is 
accompanied  by  a  decrease  in  brain 
stimulation  reward.  It  is  becoming 
increasingly  clear  that  at  the  neuro- 
chemical level,  as  well,  the  same  sys- 
tems that  have  been  implicated  in  the 
acute  reinforcing  effects  of  ethanol 
show  adaptive  changes  after  chronic 


269 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


exposure,  and  may  have  an  important 
role  both  in  the  affective  changes 
associated  with  abstinence  and  the 
reinforcing  actions  of  ethanol  in  the 
dependent  state. 

Dopamine 

The  role  of  DA  neurotransmission  in  die 
acute  reinforcing  actions  of  ethanol  is 
well  established.  Electrophysiological 
(Gessa  et  al.  1985;  Brodie  et  al.  1990; 
Diana  et  al.  1992#),  neurochemical 
(Imperato  and  DiChiara  1986;  Wozniak 
et  al.  1991;  Yoshimoto  et  al.  1992«; 
Engel  et  al.  1992;  Rossetti  et  al. 
1993;  Kiianmaa  et  al.  1995),  and 
behavioral  (Imperato  and  DiChiara 
1986;  Waller  et  al.  1986;  Pecins- 
Thompson  and  Peris  1993)  data  indi- 
cate that  behaviorally  relevant  doses  of 
ethanol  activate  the  mesolimbic  DA 
reward  pathway.  Direct  evidence  of  a 
role  for  DA  in  the  acute  reinforcing 
actions  of  ethanol  comes  from  find- 
ings that  operantly  self-administered 
ethanol  stimulates  DA  release  in  the 
nucleus  accumbens  (Weiss  et  al.  1993), 
that  rats  will  self- administer  ethanol 
directly  into  the  ventral  tegmental  cell 
body  region  of  the  meso-accumbens 
DA  reward  pathway  (Gatto  et  al. 
1994),  and  that  ethanol  preference  or 
ethanol-maintained  reinforcement  is 
modified  by  pharmacological  agents 
that  interact  with  DA  neurotransmis- 
sion (e.g.,  Weiss  et  al.  1990;  Samson 
et  al.  1992,  1993;  George  et  al.  1995; 
Panockaetal.  1995). 

Although  ethanol  acutely  activates 
mesolimbic  DA  neurotransmission, 
this  effect  shows  tolerance  in  the  depen- 
dent state.  Extraneuronal  DA  concen- 
trations in  the  nucleus  accumbens  after 


consumption  of  ethanol  liquid-diet  in 
dependent  rats  are  indistinguishable 
from  those  in  ethanol-naive  rats 
(Weiss  et  al.  1996),  and  stimulation  of 
DA  synthesis,  commonly  observed  with 
acute  ethanol,  is  attenuated  after 
chronic  ethanol  (Tabakoff  and  Hoff- 
man 1978;  Fadda  et  al.  1980).  These 
observations  suggest  that  long-term 
exposure  to  ethanol  suppresses  meso- 
accumbens  DA  activity  to  "balance" 
chronic  stimulation  by  ethanol.  This 
hypothesis  is  supported  further  by 
findings  that  ethanol  withdrawal  is 
associated  with  deficient  DA  release  in 
the  nucleus  accumbens  (Rossetti  et  al. 
1992&;  Weiss  et  al.  1996)  and  a  pro- 
found decrement  of  mesolimbic  neu- 
ronal activity  (Diana  et  al.  1992&, 
1993;  Shen  and  Chiodo  1993).  Inter- 
estingly, the  suppression  of  DA  release 
during  withdrawal  can  be  reversed  by 
systemic  injection  of  ethanol  (Rossetti 
et  al.  1992/7),  and  rats  given  the 
opportunity  to  self- administer  ethanol 
during  withdrawal  regulate  their 
ethanol  intake  in  a  manner  that 
restores  accumbal  DA  release  to  pre- 
withdrawal  levels  (Weiss  et  al.  1996). 
The  reversal  of  this  neurochemical 
deficit  by  systemic  ethanol  and,  more 
importantly,  the  apparent  behavioral 
"titration"  of  ethanol  intake  in  self- 
administering  rats  to  regain  prewith- 
drawal  conditions  implicate  accumbal 
DA  release  in  ethanol-maintained  neg- 
ative reinforcement  and,  by  extension, 
in  continued  abuse  and  dependence. 

Although  there  is  clear  evidence  for  a 
role  of  neuroadaptive  changes  in  forebrain 
DA  transmission  in  the  motivational 
effects  of  ethanol  dependence  and  with- 
drawal, the  mechanisms  underlying 


270 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


these  changes  are  presently  not  well 
understood.  Acute  ethanol  administra- 
tion stimulates  DA  synthesis,  but  this 
effect  is  blunted  in  chronically  ethanol- 
treated  animals  (Tabakoff  and  Hoff- 
man 1978;  Fadda  et  al.  1980).  Also, 
chronic  ethanol  exposure  suppresses 
K+-stimulated  DA  release  (Darden  and 
Hunt  1977),  possibly  via  inhibition  of 
Ca++  influx  (Kim  et  al.  1994)  or  by 
uncoupling  of  calcium  entry  and  DA 
release  (Leslie  et  al.  1986).  In  addi- 
tion, depolarization  inactivation  has 
been  proposed  as  a  possible  mechanism 
(Shen  and  Chiodo  1993).  These  findings 
point  toward  changes  at  the  biochem- 
ical and  cellular  level.  Future  research 
will  need  to  more  precisely  characterize 
these  potential  mechanisms,  including 
molecular  changes,  to  provide  a  basis 
for  the  pharmacotherapeutic  reversal 
of  ethanol-induced  neuroadaptive 
alterations  in  DA  transmission. 

5  -  Hydroxytryptamine 

Ample  evidence  exists  for  an  involvement 
of  5-HT  in  ethanol-seeking  behavior  as 
well.  Ethanol  increases  5-HT  release  in 
the  nucleus  accumbens  after  local,  sys- 
temic, and  self- administration  (Yoshi- 
moto  and  McBride  1992;  Yoshimoto  et 
al.  1992&;  Weiss  et  al.  1996;  Yoshimoto  et 
al.  1996).  Pharmacological  treatments 
that  increase  the  synaptic  availability  of 
5-HT,  or  direct  activation  of  5-HT 
transmission  by  receptor  agonists,  sup- 
press voluntary  ethanol  intake  in  animals 
(for  reviews,  see  Sellers  et  al.  1992; 
LeMarquand  et  al.  1994&)  and  can 
reduce  alcohol  consumption  in  humans 
(Naranjo  et  al.  1987,  1990;  Monti  and 
Alterwain  1991;  Naranjo  and  Bremner 
1993;  LeMarquand  et  al.   1994^; 


Naranjo  et  al.  1995).  A  serotonergic  role 
in  ethanol  abuse  is  supported  also  by 
findings  that  the  subjective  effects  of 
ethanol  depend,  at  least  partially,  on  5- 
HT  neurotransmission,  since  agonists 
of  the  5-HT1A  receptor  substitute  for 
the  discriminative  stimulus  properties 
of  ethanol  (Signs  and  Schechter  1988; 
Grant  and  Colombo  1993  b\  Krystal  et 
al.  1994),  whereas  5-HT3  antagonists 
block  these  properties  (Grant  and  Bar- 
rett 1991  b). 

Neuroadaptive  changes  similar  to 
those  observed  with  DA  have  been 
reported  in  serotonergic  systems  after 
chronic  ethanol  exposure.  Ethanol 
acutely  activates  central  5-HT  trans- 
mission (for  a  review,  see  LeMar- 
quand et  al.  1994&).  In  contrast, 
ethanol  withdrawal  after  induction  of 
dependence  leads  to  reductions  in  5-HT 
metabolism  and  content  of  5-HT  or 
its  metabolite,  5-hydroxyindoleacetic 
acid  (5-HIAA),  in  whole  brain,  lim- 
bic, and  striatal  tissues  (Kahn  and 
Scudder  1976;  Tabakoff  et  al.  1977; 
Badawy  and  Evans  1983;  Kempf  et  al. 
1990;  Wahlstrom  et  al.  1991;  Yama- 
mura  et  al.  1992).  More  recently  it 
was  shown  that  whereas  ethanol 
acutely  enhances  the  release  of  5-HT 
from  the  nucleus  accumbens  (Yoshi- 
moto and  McBride  1992;  Yoshimoto 
et  al.  1992£,  1996),  ethanol  with- 
drawal in  dependent  rats  is  associated 
with  a  progressive  suppression  of  5- 
HT  release  in  this  brain  region  (Weiss 
et  al.  1996).  These  findings  of  seroton- 
ergic deficiencies  during  withdrawal 
are  consistent  with  clinical  studies  that 
have  revealed  deficits  in  5-HT  synthesis, 
turnover,  or  receptor  function  in  alco- 
holics (Ballenger  et  al.  1979;  Linnoila 


271 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


et  al.  1983;  Thomson  and  McMillen 
1987;  Lee  and  Meltzer  1991)  and 
implicate  impaired  5-HT  function  as 
an  important  neurochemical  factor  in 
alcohol  abuse  and  dependence 
(LeMarquand  et  al.  1994#). 

Studies  in  ethanol-dependent  rats 
implicate  adaptations  in  5-HT  receptors 
in  addition  to  presynaptic  changes  in 
the  development  and  maintenance  of 
alcoholism.  Blockade  of  5-HTlc  and 
5-HT2  receptors  by  a  single  large  dose 
of  a  pharmacological  antagonist  pre- 
vented the  anxiogenic  effects  of 
ethanol  withdrawal  in  rats  for  up  to  7 
days  after  treatment  as  measured  on 
the  elevated  plus  maze  (Lai  et  al. 
1993).  It  appears  that  chronic  ethanol 
exposure  may  up-regulate  or  enhance 
the  sensitivity  of  5-HTlc  receptors  in 
particular,  since  chronic  ethanol-treated 
rats  show  enhanced  susceptibility  to  the 
anxiogenic-like  effects  of  a  selective  5- 
HT1C  agonist  (Rezazadeh  et  al.  1993). 
Adaptive  changes  in  the  function  of  5- 
HT1C  receptors  may,  therefore,  play  a 
significant  role  in  ethanol  withdrawal 
anxiety.  In  addition,  changes  in  the 
sensitivity  of  5-HT1A  receptors  may  have 
a  role  in  the  anxiogenic  consequences  of 
ethanol  withdrawal,  as  indicated  by  find- 
ings that  chronic  ethanol  exposure  alters 
the  sensitivity  to  several  pharmacological 
effects  of  the  5-HT1A  receptor  agonist 
8-OH-DPAT  (Kleven  et  al.  1995)  and 
that  5-HT1A  agonists  can  reverse  ethanol 
withdrawal-induced  suppression  of 
exploratory  activity  in  the  open  arms  of 
the  elevated  plus  maze  (Lai  et  al.  1991). 

Gamma- Aminobutyric  Acid 

The  anxiolytic  effects  of  ethanol,  in  addi- 
tion to  its  mood-elevating  actions,  are 


thought  to  be  an  important  mecha- 
nism promoting  its  abuse.  Studies 
exploring  the  mechanisms  by  which 
ethanol  exerts  its  anti-anxiety  effects 
have  implicated  interactions  with  the 
GABA-benzodiazepine  (BZD)  receptor 
complex.  In  general,  GABA-BZD 
antagonists  and  inverse  agonists  reverse 
the  anxiolytic  effects  of  ethanol  in 
conflict  tests  (Liljequist  and  Engel 
1984;  Koob  et  al.  1986)  and  the  ele- 
vated plus  maze  (Lister  1988;  Criswell 
et  al.  1994;  Prunell  et  al.  1994).  In 
addition,  studies  examining  the  effects 
of  ligands  interacting  with  the  GABA- 
BZD  receptor  complex  on  ethanol 
self-administration  have  provided  evi- 
dence for  a  role  of  GABA  in  ethanol- 
maintained  reinforcement.  Partial 
inverse  BZD  agonists  such  as  Ro  15- 
4513  or  Ro  19-4603  dose-depen- 
dently  suppress  ethanol  intake  in  both 
free-drinking  and  operant  self-admin- 
istration models  without  concomitant 
reduction  in  the  consumption  of 
water  or  saccharin  (McBride  et  al. 
1988;  Samson  et  al.  1989;  June  et  al. 
1991;  Rassnick  et  al.  1993#;  June  et 
al.  1994b).  Moreover,  decreases  in 
responding  for  ethanol  induced  by 
BZD  inverse  agonists  are  reversed  by 
coadministration  of  the  BZD  antago- 
nist flumazenil,  confirming  that  the 
effect  of  the  partial  agonist  on  self- 
administration  is  specific  to  the  BZD 
site  of  the  GABA-BZD  receptor 
(Samson  et  al.  1989;  Rassnick  et  al. 
1993#;  June  et  al.  1994a,  1994&). 

Evidence  supporting  a  role  for  neu- 
roadaptive  processes  in  behavioral 
changes  associated  with  chronic 
ethanol  treatment  and  dependence  is 
emerging  also  in  the  case  of  GABA.  It 


272 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


is  well  documented  that  in  contrast  to 
acute  ethanol,  which  potentiates 
GAB  A- stimulated  Cl~  flux,  chronic 
ethanol  administration  decreases 
GAB  A- dependent  CI"  influx  (Morrow 
et  al.  1988;  Kuriyama  et  al.  1993). 
This  inhibition  of  GABAergic  func- 
tion persists  for  some  time  during 
withdrawal  from  ethanol  (Kuriyama  et 
al.  1993;  Kang  et  al.  1996)  and  is 
thought  to  contribute  to  ethanol 
withdrawal  symptoms  such  as  seizure 
susceptibility,  anxiety,  and  negative 
affect,  which  may  motivate  continued 
ethanol  consumption.  Consistent  with 
this  view,  microinjections  of  the 
GABA  agonist  muscimol  into  the  cen- 
tral nucleus  of  the  amygdala  (CeA)  of 
ethanol  self-administering  rats 
decreased  enhanced  responding  for 
ethanol  associated  with  ethanol  with- 
drawal, at  doses  that  did  not  alter 
ethanol  intake  in  nondependent  rats 
(Roberts  et  al.  1996).  Counteradapta- 
tions  in  GABAergic  mechanisms  after 
chronic  ethanol  are  also  suggested  by 
a  finding  that  a  single  dose  of  the 
BZD  antagonist  flumazenil,  adminis- 
tered 14  hours  before  withdrawal, 
reversed  behavioral  manifestations 
of  ethanol  withdrawal  in  mice  (Buck 
et  al.  1991).  This  observation  sug- 
gests that  brief  occupation  of  BZD 
receptors  by  an  antagonist  may  per- 
haps "reset"  adaptive  cellular  mecha- 
nisms responsible  for  the  development 
of  dependence. 

Overall,  these  findings  are  suggestive 
of  the  development  of  counteradap- 
tive  responses  within  dopaminergic, 
serotonergic,  and  GABAergic  systems 
that  oppose  the  acute  pharmacologi- 
cal actions  of  ethanol  such  that  these 


systems  exhibit  functional  deficiencies 
in  the  absence  of  continued  stimula- 
tion by  ethanol  (Koob  and  Bloom 
1988).  These  changes,  in  conjunction 
with  adaptive  responses  that  develop 
in  systems  that  are  not  involved  in  the 
acute  reinforcing  effects  of  ethanol 
but  that  when  engaged  counter  the 
positive,  mood-elevating  effects  of 
ethanol  (see  the  next  section),  may 
provide  a  neurobiological  basis  for 
aspects  of  the  ethanol  withdrawal 
symptomatology — in  particular,  affec- 
tive changes  opposite  to  those  pro- 
duced by  ethanol  acutely. 

Disruption  of  Brain 
Stress  Systems  After 
Chronic  Ethanol 

Chronic  alcohol  abuse  has  profound 
effects  on  the  hypothalamic -pituitary- 
adrenal  (HPA)  axis.  Alcoholics  exhibit 
a  blunted  adrenocorticotropic  hormone 
(ACTH)  response  to  corticotropin- 
releasing  factor  (CRT)  administration 
(Wand  and  Dobs  1991),  suggesting 
that  chronic  ethanol  induces  HPA  axis 
injury,  which  results  in  impaired 
responsiveness  to  non-ethanol-induced 
stress.  A  blunted  stress  response  as  well 
as  abnormal  circadian  Cortisol  secretion, 
perturbations  of  the  noradrenergic 
system,  and  changes  in  CRT-norepi- 
nephrine  interactions  are  also  observed 
during  ethanol  withdrawal  (Risher- 
Flowers  et  al.  1988;  von  Bardeleben 
et  al.  1989;  Adinoffet  al.  1991;  Hawley 
et  al.  1994;  Inder  et  al.  1995;  Costa  et 
al.  1996;  Ehrenreich  et  al.  1997).  These 
changes  can  persist  beyond  the  acute 
withdrawal  phase  and  thus  may  con- 
tribute to  the  physiological  and  behav- 
ioral complications  of  chronic  alcoholism. 


273 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


In  animals,  ethanol  withdrawal  is 
associated  with  HPA  axis  activation  at 
both  the  adrenal  and  hypothalamic/ 
pituitary  levels  (Freund  1969; 
Tabakoff  et  al.  1978;  Rivier  et  al. 
1984;  De  Soto  et  al.  1985;  Ehlers  and 
Chaplin  1987;  Redei  et  al.  1988; 
Kleven  et  al.  1995;  Lamblin  et  al. 
1996).  These  neuroendocrine  effects 
are  accompanied  by  increases  in  emo- 
tionality that  resemble  the  effects  of 
stress  (Freund  1969;  Tabakoff  et  al. 
1978;  De  Soto  et  al.  1985)  and, 
therefore,  may  contribute  to  the  moti- 
vational effects  of  ethanol  withdrawal. 
A  role  for  neuroendocrine  stress  sys- 
tems in  excessive  alcohol  drinking  is 
suggested  also  by  findings  that  P  rats 
show  enhanced  electrophysiological 
responses  to  administration  of  CRF,  a 
finding  that  has  been  attributed  to  up- 
regulation  of  CRF  receptors  in  these 
animals  (Ehlers  et  al.  1992).  These 
observations  are  consistent  with  find- 
ings that  P  rats  are  more  "anxious"  in 
behavioral  tests  of  anxiety  (Stewart  et 
al.  1993)  and  suggest  that  a  relation- 
ship may  exist  between  responsivity  to 
stress,  the  status  of  the  HPA  axis 
including  the  CRF  system,  and  alco- 
hol preference. 

Much  attention  has  been  directed 
recently  at  understanding  the  role  of 
the  nonneuroendocrine  CRF  system 
in  the  CeA  in  the  affective  conse- 
quences of  ethanol  withdrawal.  Grow- 
ing evidence  suggests  that  the  CRF 
neuropeptide  system  in  the  CeA  has 
an  essential  role  in  the  mediation  of 
emotional  responses  to  stress  such  as 
anxiety  (Dunn  and  Berridge  1990; 
Heinrichs  et  al.  1992;  Swiergiel  et  al. 
1993;  Koob  et  al.  1994),  symptoms 


which  are  also  an  integral  part  of  the 
alcohol  withdrawal  syndrome  and, 
therefore,  may  involve  activation  of 
CRF  mechanisms  in  the  CeA.  In  sup- 
port of  this  hypothesis,  marked 
increases  in  CRF  release  in  the  CeA 
have  been  observed  during  withdrawal 
in  ethanol-dependent  rats  (Merlo  Pich 
et  al.  1995),  and  microinjection  of  the 
CRF  receptor  antagonist  a-helical 
CRF(9-41)  into  the  CeA  selectively 
reversed  the  anxiogenic  effects  of 
ethanol  withdrawal  as  measured  in  the 
elevated  plus  maze  (Baldwin  et  al. 
1991;  Rassnick  et  al.  1993£).  These 
results  suggest  that  in  addition  to  the 
classic  HPA  axis  activation,  discrete 
extrahypothalamic  CRF  systems  may 
be  affected  by  chronic  exposure  to 
ethanol,  and  this  may  be  reflected  in 
an  overactivity  of  these  systems  during 
withdrawal.  As  discussed  later  in  this 
chapter,  disruptions  in  both  HPA 
function  and  the  CRF  system  in  the 
CeA  may  be  an  important  factor  in 
protracted  withdrawal  and  vulnerabil- 
ity to  relapse. 

Neurosteroids 

A  comparatively  novel  area  of  research 
concerned  with  the  biological  basis  of 
alcohol  addiction  focuses  on  a  group 
of  endogenous  steroids,  termed  "neu- 
roactive  steroids,"  because  these  com- 
pounds are  synthesized  from  cholesterol 
in  the  brain  or  can  be  formed  in  the 
brain  as  metabolites  of  the  gonadal 
steroids  and  mineralocorticoids.  In 
contrast  to  the  slow,  delayed  intracel- 
lular effects  of  traditional  steroids, 
neuroactive  steroids  have  a  fast  action 
on  neuronal  membranes.  Neuroactive 
steroids  bind  to  a  specific  recognition 


274 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


site  at  the  GABA-BZD  receptor  com- 
plex, the  neurosteroid  site,  where  they 
can  modulate  the  activity  of  GABA 
(Majewska  et  al.  1986;  Morrow  et  al. 
1987).  Some  of  these  compounds, 
such  as  allopregnanolone  and  3a, Sa- 
te trahydrodeoxy  cor  ticoster  one 
(THDOC),  act  as  agonists  at  the 
GABA-BZD  receptor  complex  pro- 
ducing neuronal  inhibition  and, 
behaviorally,  exert  hypnotic  and  anxi- 
olytic-like  effects  (Crawley  et  al.  1986; 
Bitran  et  al.  1991;  Wieland  et  al. 
1991);  others,  such  as  pregnenolone 
sulfate  and  dehydroepiandrosterone 
(DHEA),  act  as  antagonists  at  the 
GABA-BZD  receptor,  increase  neu- 
ronal excitability,  and  can  produce 
anxiogenic-like  and  proconvulsant 
effects  (Majewska  and  Schwartz  1987; 
Majewska  et  al.  1990;  Melchior  and 
Ritzmann  1994&). 

Since  both  ethanol  and  neuroactive 
steroids  produce  many  of  their  biolog- 
ical and  behavioral  effects  by  interacting 
with  the  GABA-BZD  receptor,  it  is 
possible  that  neurosteroids  modulate 
the  actions  of  ethanol  and  play  a  role 
both  in  the  acute  intoxicating  effects  of 
ethanol  and  in  neuroadaptive  changes 
associated  with  chronic  ethanol  con- 
sumption. Indeed,  allopregnanolone 
and  THDOC  share  sedative  and  anxi- 
olytic discriminative  stimulus  properties 
with  ethanol  (Ator  et  al.  1993).  Neuro- 
active steroids  can  also  potentiate  (or 
reverse  [Melchior  and  Ritzmann 
1994^])  the  anxiolytic  actions  of  ethanol 
in  the  elevated  plus  maze  (Melchior 
and  Ritzmann  1994#)  and  can  enhance 
the  hypnotic  effects  of  ethanol  by 
increasing  ethanol-induced  sleep  time 
(Melchior  and  Ritzmann  1992). 


These  findings  confirm  the  exis- 
tence of  interactions  between  neuro- 
active steroids  and  the  acute  behavioral 
effects  of  ethanol,  but  there  is  also 
growing  evidence  for  a  modulatory 
role  of  neurosteroids  in  dependence 
and  withdrawal.  Plasma  levels  of  allo- 
pregnanolone and  THDOC,  the  most 
potent  endogenous  positive  modula- 
tors of  GABA-BZD  receptors,  were 
markedly  reduced  in  alcoholic  subjects 
during  the  early  withdrawal  phase, 
when  anxiety  and  depression  scores 
were  elevated.  Allopregnanolone  and 
THDOC  levels  recovered  during  the 
late  withdrawal  phase  at  a  time  when 
anxiety  and  depression  scores  returned 
to  normal  (Romeo  et  al.  1996),  and 
these  observations  suggest  that  chronic 
ethanol-induced  decrease  in  neuroactive 
steroid  biosynthesis  may  contribute  to 
ethanol  withdrawal  symptoms.  Inter- 
estingly, these  deficiencies  in  neurosteroid 
synthesis  appear  to  be  accompanied 
by  a  sensitization  of  the  GABA-BZD 
receptor  to  neuroactive  steroids  dur- 
ing withdrawal.  In  both  rats  and  mice, 
administration  of  the  anxiolytic  neu- 
rosteroid 3a,5a-tetrahydroproges- 
terone  abolished  anxiety  and  seizure 
susceptibility  during  ethanol  with- 
drawal (Devaud  et  al.  1995;  Finn  et 
al.  1995),  although  this  effect  was 
strain  dependent  in  mice  (Finn  et  al. 
1995).  These  data  not  only  provide 
further  evidence  for  an  interaction 
between  ethanol  and  neuroactive 
steroids  at  the  GABA-BZD  receptor 
but  also  suggest  that  genetic  factors  in 
neuroactive  steroid  sensitivity  and 
biosynthesis  may  contribute  to 
ethanol  withdrawal  severity,  and  that 
neurosteroids  may  be  an  important 


275 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


target  for  the  development  of  pharma- 
cological agents  capable  of  antagoniz- 
ing acute  or  chronic  effects  of  ethanol. 
The  precise  interactions  of  neu- 
roactive  steroids  with  ethanol  are  only 
beginning  to  be  understood. 
Nonetheless,  the  discovery  of  these 
endogenous  modulators  of  GABA 
function  and  their  unique  binding  site 
at  the  GABAA  receptor  complex  pro- 
vides new  perspectives  and  tools  for 
the  investigation  of  the  mechanism  by 
which  ethanol  exerts  its  behavioral 
actions,  and  for  the  development  of 
novel  compounds  that  selectively 
block  the  intoxicating  effects  of 
ethanol. 

Neurotransmitter 
Interactions  in  the 
Control  of  Ethanol- 
Seeking  Behavior 

Research  on  the  neuropharmacological 
basis  of  ethanol  reinforcement  over  the 
past  decade  has  led  to  the  recognition 
that  the  acute  reinforcing  effects  of 
ethanol  depend  on  multiple  transmit- 
ter systems  and  their  interactions 
(e.g.,  Engel  et  al.  1992).  For  example, 
evidence  is  now  accumulating  to  sug- 
gest that  one  mechanism  by  which 
ethanol  activates  the  dopaminergic 
"reward  circuitry"  involves  an  action 
on  endogenous  opioid  systems  (Wid- 
dowson  and  Holman  1992;  Acquas  et 
al.  1993;  Benjamin  et  al.  1993;  Di 
Chiara  et  al.  1996;  Gonzales  and 
Weiss  1998).  There  is  also  growing 
evidence  for  interactions  between  5- 
HT  and  DA  in  the  control  of  ethanol- 
seeking  behavior.  Serotonin  potentiates 
ethanol-induced  excitation  of  meso- 
limbic  DA  neurons  (Brodie  et  al. 


1995).  In  contrast,  5-HT3  antagonists 
suppress  ethanol-induced  DA  release 
in  the  nucleus  accumbens  and  ventral 
tegmental  area  (VTA)  (Yoshimoto  et 
al.  1992a;  Campbell  and  McBride  1995; 
Campbell  et  al.  1996),  reduce  ethanol 
intake  (Fadda  et  al.  1991;  Knapp  and 
Pohorecky  1992;  Hodge  et  al.  1993; 
lohnson  et  al.  1993),  block  the  anxi- 
olytic effects  of  ethanol  (Grant  and 
Barrett  1991a),  and  attenuate  the  dis- 
criminative stimulus  properties  of 
ethanol  (Grant  and  Barrett  1991a, 
1991/7).  These  findings  suggest  that 
5-HT3  antagonists  block  these  behav- 
ioral effects  of  ethanol  by  interfering 
with  ethanol-induced  DA  release. 
Interactions  relevant  to  the  reinforc- 
ing actions  of  ethanol  may  also  exist 
between  DA  and  glutamate  neuro- 
transmission (Rassnick  et  al.  1992&). 
The  nucleus  accumbens  receives  neu- 
ronal projections  using  glutamate, 
serotonin,  and  endogenous  opioids  as 
their  transmitters  from  limbic  and 
midbrain  regions  that  play  a  role  in 
motivational  and  emotional  processes. 
Interactions  among  these  transmitters 
in  the  nucleus  accumbens  can,  per- 
haps, be  viewed  as  "orchestrating"  the 
rewarding  effects  of  ethanol  by  orga- 
nizing the  functional  output  from  this 
structure  (Engel  et  al.  1992).  How- 
ever, the  investigation  of  such  interac- 
tions has  yet  to  be  expanded  to  their 
role  in  addiction  and  withdrawal. 

Isolated  findings  have,  in  fact, 
begun  to  suggest  that  neurotransmit- 
ter interactions  may  be  important  in 
the  development  of  dependence  and 
regulation  of  ethanol- seeking  behavior 
in  dependent  animals.  For  example, 
alterations  in  the  modulation  of  DA 


276 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


release  in  the  nucleus  accumbens  by 
5-HT  develop  over  the  course  of 
chronic  ethanol  exposure.  These 
involve,  in  particular,  changes  in  the 
regulation  of  DA  release  by  5-HT3 
receptors,  which  appear  to  compen- 
sate for  deficient  serotonergic  activity 
associated  with  chronic  ethanol  expo- 
sure and  maintain  or  even  enhance 
responsiveness  of  the  dopaminergic 
system  to  ethanol  (Yoshimoto  et  al. 
1996).  Interactions  between  neuro- 
chemical systems  in  the  development 
of  dependence  can  perhaps  also  be 
inferred  from  the  regionally  specific 
effects  of  chronic  ethanol  on  the 
expression  of  GABAA  receptor  sub- 
units.  In  particular,  a  finding  that  pro- 
longed 12-week,  but  not  4-week, 
exposure  to  ethanol  decreased  alpha- 1 
subunit  immunoreactivity  in  the  VTA 
and  hippocampus  (Charlton  et  al. 
1997)  may  suggest  that  interactions 
exist  between  GABA  and  other  trans- 
mitter systems  involved  in  reward  and 
cognitive  functions  in  these  respective 
brain  regions.  As  discussed  later  in 
this  chapter,  there  are  also  putative 
interactions  between  endogenous  opi- 
oids and  mesolimbic  DA  transmission 
that  may  be  relevant  to  mediation  of 
ethanol-seeking  behavior  in  depen- 
dent subjects. 

Sensitization 

Ethanol -Induced  Sensitization: 
Determinant  of  Heightened  Drug- 
Seeking  Behavior? 

Repeated  administration  of  drugs  can 
result  in  an  enhancement  of  their 
behavioral  and  other  pharmacological 
effects,  particularly  if  the  treatment 


regimen  involves  intermittent,  non- 
continuous  administration  (Robinson 
and  Becker  1986;  Kalivas  and  Stewart 
1991),  and  it  has  been  suggested  that 
drug-induced  sensitization  may  play 
an  important  role  in  the  development 
of  compulsive  drug- seeking  behavior, 
craving,  and  perhaps  relapse  (Hunt 
and  Lands  1992;  Robinson  and 
Berridge  1993). 

Ethanol-induced  sensitization  has 
predominantly  been  studied  in  mice,  a 
species  that  shows  augmented  loco- 
motor stimulant  responses  to  ethanol 
after  repeated  treatment  (Phillips  et  al. 
1994;  Broadbent  et  al.  1995;  Phillips 
et  al.  1995;  Roberts  et  al.  1995; 
Phillips  et  al.  1996,  1997).  This  effect 
seems  to  be  highly  strain  dependent  in 
that  it  is  limited  largely  to  DBA/2J 
mice,  implicating  genetic  factors  in 
ethanol  sensitization  (Phillips  et  al. 
1994,  1995).  Since  the  psychomotor 
stimulant  effects  of  ethanol  (Broad- 
bent  et  al.  1995;  Cohen  et  al.  1997) 
and  other  drugs  of  abuse  involve  acti- 
vation of  mesolimbic  DA  transmission 
and,  consequently,  are  thought  to 
reflect  reinforcing  properties,  such 
strain  differences  in  locomotor  sensiti- 
zation may  provide  important  clues 
with  regard  to  genetic  factors  in 
ethanol  preference.  However,  the 
interpretation  of  a  link  between  sensi- 
tization and  ethanol  preference  has 
been  complicated  by  several  issues. 

Quantitative  trait  loci  analysis 
revealed  a  negative  association  between 
ethanol  preference  and  both  the  acute 
locomotor  response  to  ethanol  and 
ethanol  sensitization  (Phillips  et  al. 
1995),  a  finding  that  supports  a  rela- 
tionship between  ethanol  preference 


277 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  reduced  susceptibility  to  the  sensi- 
tizing effects  of  ethanol  but  is  inconsis- 
tent with  the  notion  that  sensitization 
is  associated  with  enhanced  drug- 
seeking  behavior  (Hunt  and  Lands 
1992;  Robinson  and  Berridge  1993; 
Piazza  and  Le  Moal  1996).  The 
understanding  of  the  significance  of 
sensitization  in  ethanol-seeking 
behavior  and  genetic  ethanol  prefer- 
ence is  further  complicated  by  the 
question  of  whether  the  enhanced 
locomotor  activity  associated  with 
repeated  ethanol  treatments  reflects 
"true"  sensitization  or  rather  toler- 
ance to  the  sedative  effects  of  ethanol. 
This  remains  an  open  issue.  If  ethanol 
sensitization  is  the  result  of  sedative 
tolerance,  then  a  positive  relationship 
should  exist  between  sensitization  and 
tolerance  in  strains  of  mice  exhibiting 
greatest  susceptibility  to  sensitization. 
Using  ataxia  as  a  measure  of  tolerance, 
genetic  correlations  did  not  support 
the  existence  of  such  relationships  in 
recombinant  inbred  mouse  strains 
(Phillips  et  al.  1996).  In  contrast, 
complex  dose-response  analyses  of 
ethanoPs  motor  effects  in  DBA/2J 
mice  confirmed  that  apparent  sensiti- 
zation to  ethanol  was  accounted  for 
by  selective  tolerance  in  the  sedative 
limb  of  the  dose-response  function 
(Tritto  and  Dudek  1997).  A  similar 
effect  has  been  reported  in  Wistar  rats 
given  free  access  to  ethanol  for  8  weeks. 
In  these  animals  the  sedative  effects  of 
higher  ethanol  challenge  doses  were 
abolished,  and  the  animals  showed 
greatly  increased  motor  activation 
over  the  range  of  ethanol  doses  that 
produced  sedation  in  naive  animals 
(Wolffgramm  and  Heyne  1995). 


Thus,  interpretation  of  this  rather 
complex  phenomenon  seems  to  hinge 
on  the  particular  dependent  measure  of 
tolerance  used.  Clear  tolerance  to  motor 
activation  by  ethanol  was  also  observed 
in  rats  after  2  weeks  of  24-hour  access 
to  ethanol  (Pecins-Thompson  and 
Peris  1993),  suggesting  that  the  devel- 
opment of  sensitization  or  tolerance 
may  also  be  a  function  of  the  mode  of 
administration  (i.e.,  self  vs.  forced). 

If  it  is  presumed  that  ethanol  sensi- 
tization can  develop  independently  of 
sedative  tolerance,  an  important  issue 
that  arises  is  whether  sensitization  has 
a  role  in  altered  reward  or  heightened 
susceptibility  to  the  addictive  effects  of 
ethanol.  The  genetic  analyses  dis- 
cussed above  suggest  that  ethanol 
preference  is  negatively  correlated  with 
ethanol  sensitization  (Phillips  et  al. 
1995).  On  the  other  hand,  female  rats 
previously  sensitized  to  the  locomotor 
stimulant  effects  of  amphetamine 
show  enhanced  ethanol  intake  (Fahlke 
et  al.  1994),  a  finding  that  seems  con- 
sistent with  a  role  of  sensitized  dopa- 
minergic transmission  in  heightened 
ethanol-seeking  behavior  or  prefer- 
ence. Whether  such  increases  in  ethanol 
intake  reflect  heightened  ethanol- 
seeking  behavior  or  tolerance  to  the 
psychoactive  effects  of  ethanol 
remains  a  matter  of  debate.  Moreover, 
robust  changes  in  ethanol  intake  were 
not  observed  in  amphetamine-sensi- 
tized male  rats  (Samson  1995),  so 
gender  differences  may  be  another 
variable  that  must  be  considered  when 
evaluating  the  relationship  between 
ethanol  sensitization  and  preference. 

It  also  remains  unclear  on  the  basis 
of  available  data  whether  or  to  what 


278 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


extent  ethanol-induced  behavioral 
sensitization  has  a  dopaminergic  basis. 
Rats  made  dependent  on  ethanol  by  a 
chronic  liquid-diet  procedure  showed 
an  enhanced  propensity  to  develop 
sensitized  locomotor  responses  to 
cocaine  and  amphetamine  (Manley 
and  Little  1997),  suggesting  that  a 
history  of  ethanol  dependence  may, 
indeed,  lead  to  a  sensitization  of  the 
meso-accumbens  DA  system.  In  con- 
trast, repeated  ethanol  pretreatment  in 
rats  enhanced  the  locomotor  effect  of 
morphine  but  not  amphetamine,  raising 
doubt  as  to  whether  ethanol-induced 
sensitization  has  a  dopaminergic  basis 
(Nestby  et  al.  1997).  Similarly,  findings 
that  the  DA  antagonist  haloperidol 
prevented  ethanol-stimulated  locomotor 
activity  in  DBA/2J  mice  but  failed  to 
block  ethanol-induced  sensitization 
suggest  that  there  is  a  dissociation  of  the 
neurobiological  mechanisms  that  medi- 
ate the  acute  stimulant  versus  sensitiza- 
tion effects  of  ethanol  (Broadbent  et 
al.  1995)  and  that  ethanol  sensitization 
may  not  involve  alterations  in  dopami- 
nergic function  in  mice.  This  view  is 
further  supported  by  the  failure  of 
haloperidol  to  reverse  ethanol  prefer- 
ence in  ethanol -sensitized  C57  mice, 
a  finding  that,  in  fact,  suggests  that 
the  mechanisms  mediating  the  acute 
reinforcing  actions  of  ethanol  are  distinct 
from  those  mediating  ethanol-drinking 
behavior  after  the  development  of  sensi- 
tization (Ng  and  George  1994).  Thus, 
the  understanding  of  the  neurobiological 
basis  of  sensitization,  the  question  as 
to  how  the  neurobiological  changes 
observed  with  intermittent  ethanol 
treatment  relate  to  psychostimulant- 
induced  sensitization,  and  elucidation 


of  the  significance  of  sensitization  in 
ethanol-seeking  behavior  remain  great 
challenges  for  future  studies. 

Sensitization  to  stimulant  drugs  can 
also  be  induced  by  prior  exposure  to 
stress  (Kalivas  and  Duffy  1989),  and 
there  appears  to  be  a  role  for  both  the 
HPA  axis  and  extrahypothalamic  CRF 
in  stress-induced  sensitization  (Koob 
and  Cador  1993;  see  also  Richter  et 
al.  1995).  Both  stress  and  repeated 
administration  of  glucocorticoids  can 
increase  the  behavioral  effects  of  psycho- 
stimulants, and  it  has  been  hypothe- 
sized that  circulating  glucocorticoids 
may  convey  susceptibility  to  sensitiza- 
tion (and  thus  impart  heightened  sus- 
ceptibility to  psychostimulant  abuse) 
and  may  maintain  the  sensitized  state 
once  induced  (Piazza  and  Le  Moal 
1996,  1997).  Several  findings  indicate 
that,  similar  to  previous  findings  with 
psychostimulants,  repeated  exposure 
to  stress  sensitizes  DBA/2J  mice  to  the 
locomotor  activating  effects  of  ethanol. 
Moreover,  both  stress  and  ethanol- 
induced  sensitization  were  attenuated 
by  the  glucocorticoid  receptor  antagonist 
RU  38486  (Roberts  et  al.  1995;  Phillips 
et  al.  1997).  These  findings  implicate  a 
role  for  the  HPA  axis  in  ethanol  sensiti- 
zation and  cross-sensitization  with  stress 
consistent  with  the  mechanisms  proposed 
to  contribute  to  sensitization  to  other 
drugs  of  abuse.  Whether  a  link  exists 
between  ethanol  reward  and  sensitiza- 
tion involving  HPA  neuroendocrine 
mechanisms  remains  to  be  deter- 
mined, however,  in  future  research. 

Repeated  Withdrawal  and  Kindling 

Another  form  of  sensitization  that  may 
contribute  to  excessive  drinking  and 


279 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


vulnerability  to  relapse  is  the  enhanced 
withdrawal  response  after  repeated 
intoxication  and  withdrawal.  Initial 
observations  in  humans  indicated  that 
the  severity  of  withdrawal  symptoms 
increases  with  increased  duration  of 
alcohol  abuse,  leading  to  the  hypothe- 
sis that  a  "kindling"  process  similar  to 
limbic  kindling  stimulation  (Goddard 
et  al.  1969)  was  responsible  for  the 
exacerbation  of  withdrawal  severity 
(Ballenger  and  Post  1978).  It  was 
later  found  that  the  number  of  with- 
drawal episodes  was  more  predictive 
of  the  likelihood  and  severity  of  alco- 
hol withdrawal  seizures  than  the 
absolute  amount  of  alcohol  consumed 
over  the  course  of  the  addicts'  history 
of  alcohol  abuse  (Brown  et  al.  1988). 
The  kindling  hypothesis  has  received 
support  from  numerous  studies 
employing  animal  models  of  repeated 
intoxication  and  withdrawal  (Branchey 
et  al.  1971;  Carrington  et  al.  1984; 
McCown  and  Breese  1990;  Becker 
and  Hale  1993;  Kokka  et  al.  1993; 
Ulrichsen  et  al.  1995;  Becker  et  al. 
1997^,  1997b).  Mice  exposed  to 
chronic  ethanol  vapor  and  then  sub- 
jected to  repeated  withdrawal  episodes 
show  progressive  increases  in  the 
intensity  of  withdrawal  seizures 
(Becker  and  Hale  1993;  Becker  et  al. 
1997a,  1997b).  Similarly,  rats  repeat- 
edly withdrawn  from  chronic  ethanol 
treatment  exhibit  "kindling"  effects  in 
seizure  activity  (McCown  and  Breese 
1990;  Ulrichsen  et  al.  1995).  Studies 
aimed  at  examining  the  brain  sites  and 
mechanisms  responsible  for  this  phe- 
nomenon have  implicated  the  inferior 
collicular  cortex  (McCown  and  Breese 
1990;  see  also  Kang  et  al.  1996; 


N'Gouemo  et  al.  1996)  rather  than 
the  traditional  limbic  foci  in  the  kindling 
action  of  repeated  ethanol  withdrawal. 
These  studies  also  suggest  that  the  pro- 
gressive increase  in  seizure  activity 
may  be  linked  to  impaired  GABAA- 
mediated  inhibitory  input  (Kang  et  al. 
1996),  because  this  effect  was  associ- 
ated with  deregulated  flunitrazepam 
binding  (Ulrichsen  et  al.  1996)  and 
was  blocked  by  diazepam  (Ulrichsen  et 
al.  1995).  An  important  question  for 
future  research  will  be  to  examine  the 
possible  relationship  between  repeated 
withdrawal  and  kindling  phenomena 
and  changes  in  the  reinforcing  and 
motivational  effects  of  ethanol. 

PROTRACTED 
ABSTINENCE  AND 
RELAPSE 

Theoretical  Considerations 
on  the  Relapse  Process 

Alcoholism  is  a  chronic  relapsing  dis- 
order, and  the  resumption  of  alcohol 
abuse  after  periods  of  abstinence  is 
one  of  the  principal  characteristics  of 
substance  dependence  on  alcohol.  Two 
major  theoretical  positions  exist  to 
explain  the  persistence  of  addictive 
behavior  and  the  high  risk  of  relapse 
long  after  withdrawal:  conditioning 
and  homeostatic  hypotheses. 

Conditioning  hypotheses  are  based  on 
observations  that  relapse  is  often  asso- 
ciated with  exposure  to  ethanol-related 
environmental  stimuli.  This  view  holds 
that  environmental  stimuli  that  have 
become  associated  with  the  subjective 
actions  of  ethanol  by  means  of  classical 
conditioning,  or  act  as  discriminative 


280 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


stimuli  signaling  the  availability  of 
ethanol  and  "setting  the  occasion"  to 
engage  in  drug-taking  behavior,  elicit 
subjective  states  that  trigger  resumption 
of  drug  use.  Homeostatic  hypotheses 
relate  relapse  to  the  persistence  of  resid- 
ual neuroadaptive  changes  and  disrup- 
tion of  neuroendocrine  homeostasis, 
which  are  thought  to  underlie  mood 
dysregulation  and  somatic  symptoms 
such  as  insomnia  that  are  often  observed 
during  the  "protracted  abstinence" 
phase.  This  view,  therefore,  implicates 
alleviation  of  discomfort  and  negative 
affect  and,  consequently,  negative 
reinforcement  as  a  motivational  basis 
in  relapse.  The  homeostatic  and  con- 
ditioning hypotheses  are  not  mutually 
exclusive  but  may,  in  fact,  be  additive 
in  that  exposure  to  ethanol-associated 
environmental  stimuli  may  augment 
vulnerability  to  relapse  imparted  by 
homeostatic  disturbances  alone  (Koob 
and  Le  Moal  1997). 

Homeostatic  Factors:  Protracted 
Abstinence 

The  persistence  of  affective  changes  and 
heightened  susceptibility  to  relapse  after 
withdrawal  suggests  that  a  history  of 
alcohol  dependence  leads  to  long-last- 
ing changes  in  cellular  or  molecular 
mechanisms  associated  with  the  con- 
trol of  drug- seeking  behavior.  Evidence 
of  such  persistent  neurobiological 
alterations  is  beginning  to  accrue, 
although  research  directed  at  the 
identification  and  understanding  of 
the  nature  of  these  changes  is  still  in 
its  infancy. 

Clinical  studies  have  identified  retar- 
dation in  the  recovery  of  dopaminergic 
transmission  after  detoxification  and 


withdrawal  as  a  possible  factor  in  relapse 
(Heinz  et  al.  1995a,  1995&).  In 
patients  with  good  subsequent  treat- 
ment outcome,  apomorphine-stimulated 
plasma  growth  hormone  levels  (as  a 
measure  of  DA  receptor  sensitivity)  and 
G  protein-induced  inhibition  of  adeny- 
lyl  cyclase  activity  in  platelet  mem- 
branes (as  an  index  of  DA  D2-receptor 
coupled  second  messenger  mecha- 
nisms) returned  to  normal  within  the 
first  24  hours  of  withdrawal.  In  con- 
trast, in  patients  who  subsequently 
relapsed,  normalization  of  dopaminer- 
gic function  was  delayed  and  still 
showed  signs  of  disruptions  after  8 
days  of  abstinence  (Heinz  et  al.  1995  &). 
These  data  suggest  that  blunting  of  the 
growth  hormone  response  to  DA  ago- 
nists and,  by  inference,  an  impairment 
in  DA  receptor  function  or  DA- 
dependent  signal  transduction  mecha- 
nisms are  associated  with  the  risk  of 
early  relapse  in  alcoholics  (Heinz  et  al. 
1995  a)  and  poor  treatment  outcome 
(Heinz  et  al.  1995^).  The  possibility 
of  a  sustained  dopaminergic  dysfunction 
during  protracted  abstinence  has  also 
received  some  support  from  animal 
studies.  Electrophysiological  measures 
suggest  that  mesolimbic  DA  function 
in  dependent  rats  is  still  reduced  72 
hours  after  termination  of  chronic 
ethanol  treatment,  although  behav- 
ioral manifestations  of  the  alcohol 
withdrawal  syndrome  recede  within 
48  hours  (Diana  et  al.  1996). 

Other  possible  mechanisms  in  pro- 
tracted abstinence  symptoms  are  changes 
in  neuroendocrine  function.  A  history 
of  alcohol  dependence  can  change  the 
responsiveness  of  the  HPA  axis  to  stres- 
sors during  early  abstinence  (Muller  et 


281 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


al.  1989;  Costa  et  al.  1996),  and  some 
HPA  axis  dysfunction  was  still  evident 
up  to  12  weeks  postwithdrawal  in  alco- 
holics (von  Bardeleben  et  al.  1989; 
Ehrenreich  et  al.  1997).  In  addition, 
alcoholics  show  abnormalities  in  the 
hypothalamic-pituitary- thyroid  axis 
(Baumgartner  et  al.  1994),  sleep  distur- 
bances (Gillin  et  al.  1990;  Thompson 
et  al.  1995),  and  circadian  dysregula- 
tion  (Schmitz  et  al.  1996)  during  and 
after  withdrawal.  The  persistence  of 
such  changes  beyond  the  acute  with- 
drawal phase  may  convey  enhanced 
vulnerability  to  relapse.  Indeed,  mea- 
sures of  HPA  axis  dysfunction  have 
been  proposed  as  a  useful  clinical 
measure  of  susceptibility  to  relapse 
during  protracted  abstinence  (Wilkins 
etal.  1992). 

Other  neuroendocrine  mechanisms 
may  be  affected  as  well.  Withdrawal 
from  chronic  ethanol  consumption  in 
humans  causes  a  significant  increase  in 
plasma  nerve  growth  factor  (Aloe  et 
al.  1996).  Although  the  functional 
significance  of  this  phenomenon  is  not 
clear,  it  is  possible  that  the  increased 
levels  of  circulating  nerve  growth  fac- 
tor might  be  involved  in  homeostatic 
adaptive  or  reparative  processes. 
Finally,  the  activation  of  the  CRT  sys- 
tem in  the  CeA  by  ethanol  withdrawal 
(Rassnick  et  al.  1993 b\  Merlo  Pich  et  al. 
1995)  implicates  changes  in  the  func- 
tional activity  of  this  extrahypothalamic 
CRT  system  as  a  possible  mediator  of 
protracted  withdrawal  effects. 

The  long-lasting  nature  of  protracted 
abstinence  symptoms  after  withdrawal 
and  detoxification  has  naturally  begun  to 
draw  attention  to  the  molecular  mech- 
anisms and  regulation  of  gene  expression 


that  may  underlie  homeostatic  or  neuro- 
adaptive  changes  within  brain  circuit- 
ries mediating  AOD  reinforcement. 
Although  not  the  focus  of  this  review, 
research  on  the  molecular  basis  of  pro- 
tracted abstinence  has  identified  tran- 
scription factors  including  CREB  (cyclic 
adenosine  monophosphate  response 
element  binding)  and  novel  Fos-like 
proteins  (chronic  FRAs  or  Fos-related 
antigens)  as  possible  mediators  of  per- 
sistent drug  effects  (Hope  et  al.  1992; 
Hyman  1996;  Widnell  et  al.  1996) 
that  may  also  underlie  long-term 
changes  at  the  molecular  level  induced 
by  chronic  ethanol. 

Together,  these  observations  indicate 
that  neuroadaptive  or  homeostatic 
changes  induced  by  chronic  ethanol 
can  outlast  physical  withdrawal  and 
detoxification.  However,  little  is 
known  about  the  persistence,  time 
course,  and  reversibility  of  these 
changes  and  the  relationship  of  these 
parameters  to  vulnerability  to  relapse. 
Thus,  a  challenge  for  future  research 
will  be  to  better  characterize  the  func- 
tional and  behavioral  consequences  of 
protracted  abstinence  in  animal  mod- 
els, and  to  relate  alterations  in  the 
HPA  activity,  regulation  of  specific 
transcription  factors,  or  receptor  sys- 
tems to  specific  aspects  of  drug  rein- 
forcement in  animals  with  different 
histories  of  ethanol  exposure  (e.g., 
sensitization  to  acute  challenges  vs. 
changes  in  set-point  associated  with 
protracted  abstinence). 

Conditioning  Factors:  Ethanol  - 
Associated  Environmental  Stimuli 

Environmental  stimuli  associated  with 
the  availability  or  the  subjective  effects 


282 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


of  ethanol  can  induce  alcohol  craving 
and  reinstate  alcohol -seeking  behavior 
in  humans  (Heather  and  Stallard 
1989;  Rohsenow  et  al.  1994;  Stormark 
et  al.  1995;  Cooney  et  al.  1997).  In 
alcoholics  the  sight  and  smell  of  a  pre- 
ferred alcoholic  beverage  elicits  large 
changes  in  measures  of  heart  rate, 
desire  to  drink,  and  self-reported 
withdrawal  symptoms  (Staiger  and 
White  1991).  Similarly,  cue  reactivity 
such  as  salivation  and  the  urge  to 
drink  is  significantly  enhanced  in  alco- 
holics after  exposure  to  the  odor  of 
their  favorite  alcoholic  beverage,  but 
not  after  exposure  to  water  (Monti  et 
al.  1993).  The  motivational  signifi- 
cance of  ethanol-associated  environ- 
mental cues  is  also  illustrated  by  the 
finding  that  abstinent  alcoholic 
patients  worked  harder  for  alcohol 
and  experienced  greater  subjective 
and  physiological  responses  in  a  "bar- 
like" environment  than  in  a  neutral 
setting  (Ludwig  and  Stark  1974). 

Significance  of  Stress  in  the 
Relapse  Process 

Stress  has  an  established  role  in  drug 
abuse  and  dependence.  Stress  is  a  major 
determinant  of  relapse  in  humans  and 
has  been  implicated  in  the  resumption 
of  drug  abuse  habits  for  ethanol  as  well 
as  other  drugs  of  abuse  (Marlatt  1985; 
Wallace  1989;  Brown  et  al.  1995; 
McKay  et  al.  1995).  The  significance 
of  stress  as  a  factor  in  AOD-seeking 
behavior  has  also  been  amply  docu- 
mented in  animal  studies.  In  both 
rodents  and  nonhuman  primates, 
physical,  social,  and  emotional  stress 
facilitates  acquisition  or  increases  self- 
administration  of  ethanol  (Kraemer 


and  McKinney  1985;  Blanchard  et  al. 
1987;  Nash  and  Maickel  1988;  Schenk 
et  al.  1990;  Higley  et  al.  1991; 
Hilakivi- Clarke  and  Lister  1992;  Mol- 
lenauer  et  al.  1993;  Roske  et  al.  1994) 
and  other  drugs  of  abuse  (e.g.,  Ram- 
sey and  Van  Ree  1993;  Shaham  1993; 
Goeders  and  Guerin  1994;  Shaham 
and  Stewart  1994;  Haney  et  al. 
1995).  In  addition,  studies  showing 
that  exposure  to  stress  can  reinstate 
cocaine,  heroin,  and  ethanol-seeking 
behavior  in  drug-free  animals  have 
provided  direct  experimental  evidence 
that  stress  has  a  role  in  relapse  (e.g., 
Shaham  and  Stewart  1995;  Ahmed 
and  Koob  1997).  However,  with  the 
exception  of  a  single  recent  study  in 
which  footshock  stress  reinstated 
extinguished  ethanol-seeking  behavior 
(Le  et  al.  1998),  the  effects  of  stress 
have  not  yet  been  systematically  exam- 
ined in  animal  models  of  relapse. 

Animal  Models  of  Relapse 

In  spite  of  the  significance  of  stress  and 
environmental  stimuli  in  relapse  to 
alcohol  abuse,  studies  examining  these 
factors  and  their  neurobiological  basis 
in  the  relapse  process  have  been 
scarce,  and  appropriate  animal  models 
suited  for  such  investigations  are  still 
under  development. 

Ethanol-Associated 
Environmental  Stimuli 

One  recent  study  examined  the  effects 
of  olfactory  discriminative  stimuli  pre- 
dictive of  alcohol  availability  (ethanol 
odor)  or  nonavailability  (water  odor) 
on  ethanol-seeking  behavior  (Katner  et 
al.  1999).  Rats  were  given  the  opportu- 
nity to  self-administer  10  percent 


283 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol  or  water  and  then  subjected  to 
extinction  procedures  where  lever 
presses  had  no  scheduled  consequences. 
After  extinction  of  ethanol-seeking 
behavior,  the  animals  were  again  pre- 
sented with  the  respective  discrimina- 
tive stimuli,  but  without  the 
availability  of  ethanol  or  water.  Pre- 
sentation of  the  ethanol-associated 
cue  but  not  the  water- associated  cue 
elicited  and  maintained  significant 
responding  despite  the  continued 
unavailability  of  ethanol,  and  this  effect 
was  selectively  attenuated  by  the  opiate 
antagonist  naltrexone  (Katner  et  al. 
1999).  Similar  effects  were  obtained 
when  rats  were  presented  with  a  cue 
light  that  had  been  contiguously  paired 
with  lever  responses  for  ethanol  during 
self-adrninistration  training  (Heyser  and 
Koob  1997).  Moreover,  the  ethanol- 
associated  cue  light  increased  the  resis- 
tance to  extinction  when  the  cue  light 
was  presented  in  the  absence  of  the 
ethanol  reinforcer  (Heyser  and  Koob 
1997).  These  results  suggest  that 
ethanol-associated  cues  can  both 
maintain  ongoing  ethanol-seeking 
behavior  and  reinstate  extinguished 
ethanol-seeking  behavior  in  rats.  A 
further  striking  demonstration  of  the 
control  of  ethanol-seeking  behavior 
by  ethanol-related  environmental  cues 
is  the  finding  that  exposure  to  such 
cues  during  a  period  of  extinction,  but 
not  abstinence  alone,  drastically 
reduces  subsequent  reacquisition  of 
ethanol  self- administration  (Krank  and 
Wall  1990). 

Alcohol  Deprivation  Effect 

The  increase  in  voluntary  ethanol 
consumption  after  forced  deprivation 


(alcohol  deprivation  effect)  discussed 
earlier  in  this  chapter  is  receiving 
attention  as  a  tool  to  study  the  neuro- 
biology of  alcohol  craving  and  relapse 
(Heyser  et  al.  1996;  Holter  et  al.  1996; 
Spanagel  et  al.  1996;  Heyser  et  al. 
1997;  Holter  et  al.  1997).  Similarities 
exist  between  the  alcohol  deprivation 
effect  in  animals  and  certain  character- 
istics of  human  alcohol  abuse,  includ- 
ing enhanced  alcohol  consumption  after 
abstinence  in  social  drinkers  (Burish  et 
al.  1981);  binge  drinking,  in  which 
alcohol  consumption  is  followed  by  a 
period  of  abstinence  (Mendelson  and 
Mello  1966;  Woods  and  Winger 
1971);  and  aspects  of  the  "loss  of 
control"  phenomenon  surrounding 
the  first  drink  after  abstinence  in  alco- 
holics (Ludwig  and  Wikler  1974; 
Ludwig  et  al.  1974;  O'Donnell  1984). 
In  view  of  these  similarities,  this  pro- 
cedure appears  to  have  appropriate 
face  validity  as  a  model  for  certain 
aspects  of  the  relapse  process.  More- 
over, findings  that  pharmacological 
agents  that  suppress  ethanol  intake 
and  reduce  the  likelihood  of  relapse  in 
humans  effectively  attenuate  the  alco- 
hol deprivation  effect  in  animals  (see 
the  section  Excitatory  and  Inhibitory 
Amino  Acids  later  in  this  chapter) 
have  added  support  for  the  predictive 
validity  of  this  procedure  as  a  model 
of  relapse. 

The  alcohol  deprivation  effect  can 
be  demonstrated  under  both  limited- 
access  and  unlimited-access  conditions 
and  with  both  home -cage  free  drink- 
ing and  operant  self-administration 
(e.g.,  Sinclair  and  Senter  1967;  Kor- 
net  et  al.  1990;  Wolffgramm  and 
Heyne  1995;  Spanagel  et  al.  1996; 


284 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Heyser  et  al.  1997;  Holter  et  al.  1997). 
Given  the  stability  and  robustness  of 
the  alcohol  deprivation  effect  under  a 
variety  of  experimental  conditions, 
this  phenomenon  may  have  great  utility 
for  the  exploration  of  diverse  variables 
in  the  relapse  process  with  a  variety  of 
experimental  approaches,  including 
motivational  measures  such  as  pro- 
gressive ratio  performance  and  analyses 
involving  behavioral  economics. 

Neurochemical  Basis 
of  Relapse 

Available  information  on  the  possible 
neuropharmacological  basis  of  relapse 
processes  is  limited.  In  the  case  of  psy- 
chostimulants and  opiates,  pharmaco- 
logical manipulations  that  activate 
meso-accumbens  DA  transmission,  as 
well  as  footshock  stress,  have  been 
shown  to  reinstate  drug  self- adminis- 
tration in  animals  trained  and  then 
extinguished  on  intravenous  drug  self- 
administration  (deWit  and  Stewart 
1981;  Stewart  and  deWit  1987;  Shaham 
1993;  Shaham  and  Stewart  1994, 
1995;  Weissenborn  et  al.  1995;  Self  et 
al.  1996).  This  suggests  that  conditions 
that  activate  the  mesolimbic  DA  system 
may  exert  a  "priming  effect"  and  thereby 
elicit  drug-seeking  behavior. 

Dopamine 

There  is,  indeed,  evidence  that  the 
anticipation  of  ethanol  availability 
stimulates  DA  release.  Rats  placed  in 
an  environment  previously  associated 
with  ethanol  self- administration  consis- 
tently show  a  transient  rise  in  extracel- 
lular DA  levels  in  the  nucleus  accumbens 
(Weiss  et  al.  1993;  Gonzales  and  Weiss 
1998;  see  also  Vavrousek-Jakuba  et  al. 


1992).  There  is  also  evidence  that 
self- administration  of  saccharin,  which 
is  not  normally  associated  with  signifi- 
cant dopaminergic  activation,  will 
produce  stimulation  of  DA  release 
when  ethanol,  rather  than  saccharin,  is 
the  expected  reinforcer  (Katner  et  al. 
1996).  These  effects  presumably  are  a 
consequence  of  the  presence  of  incen- 
tive motivational  stimuli,  which  are 
predictive  of  impending  availability  of 
ethanol.  One  may  speculate,  therefore, 
that  ethanol-related  cues  may  have  a 
role  in  relapse  by  exerting  a  priming 
action  (Kaplan  et  al.  1983,  1984, 
1985;  Laberg  1986;  Laberg  and 
Ellertsen  1987;  Heather  and  Stallard 
1989),  because,  like  ethanol,  these 
stimuli  increase  the  release  of  DA  in 
the  nucleus  accumbens.  Consistent 
with  this  possibility,  the  DA  antago- 
nist haloperidol  reversed  craving  and 
difficulty  in  resisting  additional  alcohol 
consumption  induced  by  a  "priming 
dose"  of  alcohol  in  detoxified  alco- 
holic patients  (Modell  et  al.  1993). 
However,  "anticipatory"  increases  in 
DA  release  have  also  been  observed 
with  saccharin  and  food  reinforcers 
(Weiss  et  al.  1993;  Wilson  et  al. 
1995),  and  expectation  of  a  nondrug 
reinforcer  has  been  associated  with  a 
discrete  increase  in  the  firing  rate  of 
mesolimbic  DA  neurons  (Schultz  et 
al.  1997).  Stimulation  of  DA  neuronal 
activity  and  release  appears  to  be  asso- 
ciated, therefore,  with  the  anticipation 
of  reinforcing  stimuli  in  general  and  is 
not  restricted  to  drug  reinforcers. 

Thus,  a  better  understanding  of  the 
neurochemical  basis  of  relapse  elicited  by 
ethanol-related  cues  is  urgently  needed. 
Moreover,  the  priming  hypothesis 


285 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


remains  to  be  confirmed  by  demon- 
strations that  pharmacological  interfer- 
ence with  the  dopaminergic  activation 
produced  by  ethanol  cues  reduces  their 
efficacy  to  reinstate  ethanol-seeking 
behavior.  Finally,  DA  release  associated 
with  the  expectation  of  ethanol  and  the 
effects  of  ethanol-related  cues  on  the 
reinstatement  of  ethanol-seeking  behav- 
ior have,  to  date,  only  been  demon- 
strated in  nondependent  animals.  It 
will  be  important  to  confirm  and  char- 
acterize these  effects  in  animals  with  a 
history  of  alcohol  dependence  in 
appropriate  "reinstatement"  models. 

Endogenous  Opioids 

A  growing  number  of  clinical  studies 
suggest  that  naltrexone  is  an  effective 
pharmacological  adjunct  for  reducing 
ethanol  craving  and  relapse  in  human 
alcoholics  (O'Malley  et  al.  1992; 
Volpicelli  et  al.  1992,  1995#,  1995 b, 
1995 r,  O'Brien  et  al.  1996;  Oslin  et  al. 
1997),  and  recent  preliminary  work 
indicates  that  naltrexone  can  prevent  the 
reinstatement  of  extinguished  respond- 
ing by  ethanol-associated  environmen- 
tal cues  in  rats  (Katner  et  al.  1999). 
The  mechanisms  underlying  the  atten- 
uation of  volitional  ethanol  intake  by 
opiate  receptor  antagonists  are  not 
well  understood  but  may  involve  an 
interaction  with  mesolimbic  DA  neu- 
rotransmission in  addition  to  DA- 
independent  effects.  Both  the  nucleus 
accumbens  and  VTA  are  rich  in  opi- 
oid peptides  and  receptors  (Wamsley 
et  al.  1980;  Lewis  et  al.  1983;  Dilts 
and  Kalivas  1989,  1990).  Afferent 
projections  to  these  areas  as  well  as 
local  interneurons  (Khachaturian  et  al. 
1993;  de  Waele  et  al.  1995)  provide  a 


potential  anatomical  substrate  by  which 
endogenous  opioids  may  modulate 
the  dopaminergic  and,  ultimately,  the 
rewarding  effects  of  ethanol.  In  effect, 
both  naltrexone  and  the  delta-selec- 
tive opiate  antagonist  naltrindole  have 
been  shown  to  blunt  ethanol -induced 
increases  in  DA  release  from  the 
nucleus  accumbens  after  systemic  or 
local  administration  of  ethanol 
(Acquas  et  al.  1993;  Benjamin  et  al. 
1993).  Thus,  it  is  possible  that  interfer- 
ence with  ethanol-induced  stimulation 
of  DA  release  may  be  a  mechanism  by 
which  opiate  antagonists  suppress 
ethanol-seeking  behavior.  This  hypoth- 
esis has  received  strong  support  by  the 
recent  demonstration  that  decreases  in 
ethanol-reinforced  operant  respond- 
ing produced  by  naltrexone  are 
directly  coupled  to  naltrexone- 
induced  decreases  in  the  efficacy  of 
ethanol  to  increase  DA  release  in  the 
nucleus  accumbens  (Gonzales  and 
Weiss  1998). 

Excitatory  and  Inhibitory 
Amino  Acids 

A  potential  role  for  GABAergic  and 
glutamatergic  mechanisms  in  relapse  has 
emerged  on  the  basis  of  both  clinical  and 
basic  research  involving  the  drug  acam- 
prosate  (calcium  N^cetylhomotaurine). 
Acamprosate  has  been  used  successfully 
for  relapse  prevention  in  detoxified 
alcoholics  in  Europe  (Lhuintre  et  al. 
1985,  1990;  Soyka  and  Sass  1994; 
Paille  et  al.  1995;  Sass  et  al.  1996; 
Whitworth  et  al.  1996;  Pelc  et  al. 
1997;  for  reviews,  see  Chick  1995; 
Wilde  and  Wagstaff  1997).  In  behav- 
ioral studies  that  have  used  the  alcohol 
deprivation  procedure  as  a  model  of 


286 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


relapse  in  rats,  acamprosate  effectively 
reversed  the  increase  in  ethanol  intake 
associated  with  forced  abstinence 
(Heyser  et  al.  1996;  Spanagel  et  al. 
1996;  Holter  et  al.  1997;  Spanagel 
and  Zieglgansberger  1997). 

Although  the  neurobiological  mech- 
anisms by  which  acamprosate  acts  to 
exert  its  putative  anticraving  and 
antirelapse  action  are  not  well  under- 
stood, there  is  some  evidence  that  this 
agent  reduces  ethanol  withdrawal- 
induced  neuronal  hyperexcitability  by 
interacting  with  glutamatergic/NMDA 
(N-methyl-D-aspartate)  and,  perhaps, 
GABAergic  transmission  (Zeise  et  al. 
1993;  Spanagel  et  al.  1996;  Holter  et 
al.  1997).  Withdrawal-associated 
hyperexcitability  is  predominantiy  medi- 
ated by  changes  in  GABA  systems, 
voltage-gated  Ca2+  channels,  and  glu- 
tamate/NMDA  systems,  and  evidence 
from  functional  studies  confirms  that 
acamprosate  interacts  with  these  sys- 
tems (Littleton  1995;  Spanagel  and 
Zieglgansberger  1997).  In  particular, 
acamprosate  may  have  a  modulatory 
action  on  glutamatergic  mechanisms 
by  enhancing  glutamate  transmission 
under  some  conditions  (Madamba  et 
al.  1996)  but  inhibiting  it  under  other 
conditions  (Zeise  et  al.  1993).  Acam- 
prosate also  suppresses  elevated  c-fos 
expression  in  rats  undergoing  with- 
drawal within  brain  reward  regions 
(Putzke  et  al.  1996),  a  finding  that  is 
consistent  with  the  putative  antire- 
lapse properties  of  this  agent. 

Overall,  there  is  growing,  albeit  still 
tentative,  evidence  that  acamprosate 
can  suppress  neuronal  excitability  and 
associated  motivational  effects  during 
ethanol  withdrawal,  and  that  these 


effects  involve,  in  particular,  interactions 
with  excitatory  (NMD A)  amino  acid 
transmission.  A  possible  role  for  NMDA 
receptors  in  ethanol-seeking  behavior 
and  relapse  has  also  recently  been  impli- 
cated by  the  finding  that  memantine 
(l-amino-3,5-dimethyl-adamantane), 
an  uncompetitive  NMDA  antagonist, 
suppressed  ethanol  deprivation- 
induced  increases  in  ethanol  consump- 
tion (Holter  et  al.  1996). 

5  -Hydroxytryptamine 

Clinical  studies  implicate  a  possible 
involvement  of  serotonergic  mechanisms 
in  alcohol  craving  and  relapse.  Several 
pharmacological  agents  that  interact 
with  5-HT  receptors,  including  the 
nonselective  partial  5-HT  agonist  m- 
chlorophenylpiperazine  (mCPP) 
(Malec  et  al.  1996)  and  the  partial  5- 
HT1A  agonist  buspirone  (Buydens- 
Branchey  et  al.  1997),  reduce  alcohol 
craving  and  rates  of  relapse  in  alco- 
holic patients.  In  the  case  of  bus- 
pirone, this  effect  has  been  attributed 
to  the  established  anxiolytic  efficacy  of 
this  drug,  which  is  consistent  with  the 
hypothesis  that  anxiety  associated  with 
ethanol  withdrawal  and  protracted 
abstinence  is  a  major  motivational  fac- 
tor in  relapse.  The  anticraving  effect 
of  mCPP  was  observed  with  oral 
administration  (Malec  et  al.  1996); 
this  is  an  interesting  finding  because, 
when  administered  intravenously,  par- 
tial 5-HT  receptor  agonists  have  been 
shown  to  produce  an  ethanol-like 
feeling  of  "high"  and  craving  for  alco- 
hol in  detoxified  alcoholics  (Benkelfat 
et  al.  1991;  Lee  and  Meltzer  1991; 
Krystal  et  al.  1994).  These  observa- 
tions are  consistent  with  data  from 


287 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


animal  studies  that  have  implicated  5- 
HT  in  die  discriminative  stimulus  prop- 
erties of  ethanol  (Grant  and  Barrett 
.19916;  Grant  and  Colombo  1993^, 
19936)  and  suggest  that,  depending 
on  the  rate  of  rise  in  their  blood  and 
brain  concentrations,  these  agents 
may  either  have  priming  effects  lead- 
ing to  enhanced  craving  (e.g.,  Ludwig 
et  al.  1974;  Jaffe  et  al.  1989;  Modell 
et  al.  1993)  or  substitute  for  aspects 
of  ethanoPs  psychoactive  effects  and, 
thereby,  reduce  craving. 

Stress  and  Neuroendocrine 
Mechanisms 

As  discussed  earlier,  the  literature  iden- 
tifies stress  as  a  major  factor  in  relapse. 
There  are  currently  no  effective  animal 
models  to  study  the  role  of  stress  in 
relapse  to  ethanol-seeking  behavior, 
and  understanding  of  the  neurobiol- 
ogy of  stress-induced  relapse  in  the 
case  of  both  alcohol  and  other  drugs 
of  abuse  is  limited.  Stressful  stimuli 
are  known  to  activate  the  HPA  axis, 
and  stress-induced  increases  in  psy- 
chostimulant (Haney  et  al.  1995)  and 
ethanol  (Nash  and  Maickel  1988) 
intake  have  been  directly  linked  to  HPA 
activation.  Stress  can  also  increase  the 
release  of  DA  in  the  nucleus  accum- 
bens  (Imperato  et  al.  1992;  Shaham 
and  Stewart  1995;  Weiss  et  al.  1997), 
which  may,  in  turn,  perhaps  serve  as  a 
discriminative  or  "priming  stimulus" 
for  the  initiation  of  AOD-seeking 
behavior.  Other  studies  suggest  that 
certain  forms  of  stress  may  exacerbate 
dopaminergic  deficits  associated  with 
psychostimulant  withdrawal  (Rossetti 
et  al.  1992&)  and,  thereby,  perhaps 
contribute  to  the  increased  likelihood 


of  relapse  associated  with  stress  (Weiss 
et  al.  1997). 

In  addition  to  HPA  activation,  dis- 
turbances in  the  amygdaloid  CRF  sys- 
tem by  chronic  drug  use  may  have  a 
role  in  stress-induced  drug-seeking 
behavior.  Amygdaloid  CRF  neurons 
project  to  brain  regions  involved  in 
autonomic  and  neuroendocrine  func- 
tions such  as  hypothalamic  nuclei,  but 
also  innervate  midbrain  monoaminer- 
gic  neurons  that  regulate  behavioral 
and  reward  functions.  It  has,  therefore, 
been  suggested  that  amygdaloid  CRF 
neurons  are  a  component  of  an  intrin- 
sic CRF  brain  circuitry  that  activates 
other,  more  classical,  central  neuro- 
transmitter systems  which,  in  turn, 
initiate  or  control  components  of 
behavioral  and  autonomic  responses 
to  stress  (Gray  1993).  Thus,  by  acti- 
vating these  autonomic  and  behavioral 
centers,  CRF  neurons  in  the  amygdala 
may  not  only  contribute  to  withdrawal 
distress  and  behavioral  withdrawal 
responses  but  also  play  a  role  in  the 
initiation  of  AOD-seeking  behavior 
and  relapse. 

GAPS  IN  SCIENTIFIC 
KNOWLEDGE  AND 
FUTURE  RESEARCH 
PRIORITIES 

Although  substantial  advances  in  the 
understanding  of  the  neurobiology  of 
alcohol  addiction  have  been  made,  there 
are  numerous  areas  in  which  current 
knowledge  is  limited.  The  recommen- 
dations below  represent  those  research 
needs  that  are  considered  most  critical 
for  the  advancement  of  present  under- 
standing of  neurobiological,  genetic, 


288 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


and  environmental  factors  in  alco- 
holism, and  for  the  development  of 
more  effective  pharmacotherapeutic 
tools  to  treat  this  condition. 

Protracted  Abstinence 
and  Relapse 

Alcoholism  is  a  chronic  relapsing  dis- 
order. Yet,  the  precise  conditions  that 
lead  to  relapse,  whether  environmentally 
determined  or  a  result  of  persistent 
homeostatic-neuroadaptive  disruptions, 
are  poorly  understood.  A  systematic 
research  effort  at  the  behavioral,  neuro- 
chemical, cellular,  and  molecular  level 
will  be  needed  to  identify  neuroadaptive 
changes  and  homeostatic  disturbances 
during  protracted  withdrawal,  and  to 
determine  their  motivational  significance 
in  appropriate  models  of  ethanol-seeking 
behavior  and  relapse.  At  the  same  time, 
it  will  be  important  to  better  under- 
stand the  role  and  neurobiological  basis 
of  conditioning  factors,  external  stres- 
sors, and  alterations  in  the  reinforcing 
qualities  of  ethanol,  as  well  as  interac- 
tions between  these  variables  and  neu- 
roadaptive changes  associated  with  a 
history  of  ethanol  dependence. 

Mechanisms  of  Reinforcement 
in  Dependent  Preparations 

There  is  a  need  to  better  model  various 
aspects  of  alcoholism  in  laboratory  ani- 
mals. This  includes,  in  particular,  volun- 
tary drinking  models  that  promote 
spontaneous  and  persistent  intake  of 
high  ethanol  concentrations  or  volumes 
without  prior  need  to  induce  depen- 
dence. Such  models  will  represent  an 
important  step  toward  the  need  for 
studying  critical  issues  such  as  the  mech- 
anisms underlying  the  switch  from 


nondependence  to  dependence  and 
mechanisms  that  maintain  alcohol 
consumption  in  dependent  individuals. 

Neurocircui  tries 
and  Transmitter  Interactions 
Mediating  Ethanol  Reinforcement 
in  Dependent  Subjects 

There  is  a  need  to  study  neurotrans- 
mitter circuitries  and  interactions 
mediating  ethanol  reward  in  the 
dependent  and  postdependent  state. 
Although  there  is  increasing  evidence 
that  the  acute  reinforcing  actions 
of  ethanol  depend  on  multiple  neuro- 
chemical systems  and  their  interac- 
tions, little,  if  anything,  is  known 
about  these  mechanisms  in  dependent 
subjects.  In  this  context,  it  will  also 
be  beneficial  to  incorporate  multiple 
systems  approaches  in  medication 
development  efforts  and  to  examine 
the  therapeutic  efficacy  of  combina- 
tions of  relevant  pharmacological 
agents. 

The  DA  Hypothesis  of  Ethanol 
Reward:  Revisited 

It  will  be  important  to  clarify  the  role 
of  DA  in  ethanol  reinforcement.  This 
need  involves  both  a  better  under- 
standing of  mechanistic  questions 
(e.g.,  how  ethanol  activates  mesolim- 
bic  DA  transmission)  and  a  better 
understanding  of  the  precise  role  of 
DA  in  various  aspects  of  ethanol-seek- 
ing behavior,  such  as  craving,  relapse, 
and  loss  of  control. 

Ethanol-Induced 
Sensitization 

An  important  emerging  issue  is  the  role 
of  sensitization  in  ethanol  reinforcement, 


289 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


genetic  preference,  and  dependence.  In 
particular,  the  following  questions  will 
require  clarification:  (1)  Does  ethanol 
sensitization  augment  the  reinforcing 
efficacy  or  potency  of  ethanol?  (2) 
Does  ethanol  sensitization  promote  a 
heightened  motivational  state  with 
increased  ethanol-seeking  behavior 
("craving"),  without  necessarily  alter- 
ing the  reinforcing  efficacy  of  ethanol? 
(3)  Is  ethanol  sensitization  a  correlate 
of  aversive  or  side  effects  of  repeated 
ethanol  intoxication,  and,  if  so,  is 
ethanol  sensitization  negatively  linked 
with  ethanol  preference  or  vulnerabil- 
ity to  abuse? 

Significance  of  Repeated 
Intoxication  and  Withdrawal 

The  kindling  phenomenon  associated 
with  repeated  intoxication  and  with- 
drawal may  have  important  implications 
with  regard  to  changes  in  motiva- 
tional and  reinforcement  processes.  It 
is  important  to  better  understand  the 
mechanisms  of  kindling  or  sensitiza- 
tion of  withdrawal  severity  at  the 
molecular,  cellular,  and  biochemical 
levels.  This  includes  efforts  to  define 
sensitization  of  psychological  compo- 
nents of  withdrawal  (e.g.,  anxiety, 
affective  changes),  to  characterize 
possible  changes  in  the  subjective  per- 
ception of  ethanol's  intoxicating 
actions  (i.e.,  ethanol's  discriminative 
stimulus  effects),  to  determine 
whether  multiple  ethanol  withdrawal 
experiences  alter  the  reinforcing  prop- 
erties of  ethanol,  and  to  examine 
potential  changes  in  susceptibility  to 
ethanol  neurotoxicity  and  associated 
cognitive  impairments.  Finally,  it  is 
important  to  examine  whether  condi- 


tioning factors  contribute  to  the  kin- 
dling phenomenon. 

Development  of  New 
Pharmacogenetic  Models 

Motivational  measures  of  ethanol  rein- 
forcement have  revealed  that  there  can 
be  overlap  between  aspects  of  ethanol- 
seeking  behavior  in  alcohol-preferring 
animals  of  certain  lines  with  the  behavior 
of  nonpreferring  animals  of  other  lines. 
This  situation  makes  it  difficult  to  con- 
duct comparative  neurobiologies  and 
behavioral  investigations  among  the 
present  rat  models.  Thus,  some  consen- 
sus will  be  required  of  what  constitutes 
a  valid  animal  model  of  ethanol  drink- 
ing. Free-choice  drinking  preference  is 
one  criterion,  but  it  may  be  insufficient 
by  itself  for  defining  a  good  animal 
model.  Human  alcoholics  will  expend 
considerable  time  and  effort  to  secure 
an  adequate  supply  of  their  preferred 
beverage,  so  motivational  characteris- 
tics such  as  persistence  of  ethanol-seek- 
ing behavior  and  "willingness"  to 
expend  effort  in  obtaining  ethanol 
would  seem  to  be  essential  criteria  of 
animal  models  of  alcoholism.  Such 
models,  which  would  incorporate  ani- 
mals that  show  robust  ethanol-seeking 
behavior  when  subjected  to  motiva- 
tional tests  (e.g.,  second-order  and 
progressive-ratio  schedules),  are  likely 
to  provide  an  invaluable  means  for  the 
study  of  genetic  and  neurobiologies 
factors  underlying  compulsive  ethanol- 
seeking  behavior  and  dependence. 

ACKNOWLEDGMENTS 

The  author  gratefully  acknowledges 
financial  support  by  the  National 


290 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Institute  on  Alcohol  Abuse  and 
Alcoholism  Extramural  Advisory 
Board,  and  thanks  Mike  Arends  for 
excellent  assistance  in  the  preparation 
of  the  manuscript. 

REFERENCES 

Acquas,  E.;  Meloni,  M.;  and  Di  Chiara, 
G.  Blockade  of  6-opioid  receptors  in  the 
nucleus  accumbens  prevents  ethanol- 
induced  stimulation  of  dopamine  release. 
Eur  J  Pharmacol  230:239-241,  1993. 

Adams,  M.L.;  Sewing,  B.N.;  Chen,  J.; 
Meyer,  E.R.;  and  Cicero,  T.J.  Nitric  oxide 
related  agents  alter  alcohol  withdrawal  in 
male  rats.  Alcohol  Clin  Exp  Res  19:195- 
199, 1995. 

Adinoff,  B.;  Risher-Flowers,  D.;  De  Jong, 
J.;  Ravitz,  B.;  Bone,  G.H.;  Nutt,  D.J.; 
Roehrich,  L.;  Martin,  P.R.;  and  Linnoila, 
M.  Disturbances  of  hypothalamic-pituitary- 
adrenal  axis  functioning  during  ethanol 
withdrawal  in  six  men.  Am  J  Psychiatry 
148:1023-1025,  1991. 

Ahmed,  S.H.,  and  Koob,  G.F.  Cocaine  - 
but  not  food-seeking  behavior  is  reinstated 
by  stress  after  extinction.  Psychopharmacology 
(Berl)  132:289-295,  1997. 

Aloe,  L.;  Tuveri,  M.A.;  Guerra,  G.;  Pinna, 
L.;  Tirassa,  P.;  Micera,  A.;  and  Alleva,  E. 
Changes  in  human  plasma  nerve  growth 
factor  level  after  chronic  alcohol  consump- 
tion and  withdrawal.  Alcohol  Clin  Exp  Res 
20:462^165,  1996. 

American  Psychiatric  Association.  Diagnostic 
and  Statistical  Manual  of  Mental 
Disorders.  4th  ed.  Washington,  DC:  the 
Association,  1994. 

Ator,  N.A.;  Grant,  K.A.;  Purdy,  R.H.; 
Paul,  S.M.;  and  Griffiths,  RR  Drug 
discrimination  analysis  of  endogenous 


neuroactive  steroids  in  rats.  Eur  J 
Pharmacol  241:237-243,  1993. 

Badawy,  A.A.,  and  Evans,  M.  Opposite 
effects  of  chronic  administration  and  subse- 
quent withdrawal  of  drugs  of  dependence 
on  the  metabolism  and  disposition  of 
endogenous  and  exogenous  tryptophan  in 
the  rat.  Alcohol  Alcohol  18:369-382,  1983. 

Bain,  G.T.,  and  Kornetsky,  C.  Ethanol 
oral  self- administration  and  rewarding  brain 
stimulation.  Alcohol  6:499-503,  1989. 

Baldwin,  H.A.;  Rassnick,  S.;  Rivier,  J.; 
Koob,  G.F.;  and  Britton,  K.T.  CRF  antag- 
onist reverses  the  "anxiogenic"  response 
to  ethanol  withdrawal  in  the  rat.  Psycho- 
pharmacology  103:227-232,  1991. 

Ballenger,  J.C.,  and  Post,  RM.  Kindling 
as  a  model  for  alcohol  withdrawal  syn- 
dromes. Br  J  Psychiatry  133:1-14,  1978. 

Ballenger,  J.;  Goodwin,  F.;  Major,  L.;  and 
Brown,  G.  Alcohol  and  central  serotonin 
metabolites  in  man.  Arch  Gen  Psychiatry 
36:224-227,  1979. 

Baumgartner,  A.;  Rommelspacher,  H.; 
Otto,  M.;  Schmidt,  L.G.;  Kurten,  I.;  Graf, 
K.J.;  Campos-Barros,  A.;  and  Platz,  W. 
Hypothalamic-pituitary- thyroid  (HPT) 
axis  in  chronic  alcoholism.  I.  HPT  axis  in 
chronic  alcoholics  during  withdrawal  and 
after  3  weeks  of  abstinence.  Alcohol  Clin 
Exp  Res  18:284-294,  1994. 

Becker,  H.C.,  and  Hale,  RL.  Repeated 
episodes  of  ethanol  withdrawal  potentiate 
the  severity  of  subsequent  withdrawal 
seizures:  An  animal  model  of  alcohol 
withdrawal  "kindling. "Alcohol  Clin  Exp 
Res  17:94-98,  1993. 

Becker,  H.C.;  Diaz-Granados,  J.L.;  and 
Hale,  RL.  Exacerbation  of  ethanol  with- 
drawal seizures  in  mice  with  a  history  of 
multiple  withdrawal  experience.  Pharmacol 
Biochem  Behav  57:179-183,  1997a. 


291 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Becker,  H.C.;  Diaz-Granados,  J.L.;  and 
Weathersby,  R.T.  Repeated  ethanol  with- 
drawal experience  increases  the  severity  and 
duration  of  subsequent  withdrawal  seizures 
in  mice.  Alcohol  14:319-326,  1997b. 

Begleiter,  H.  Ethanol  consumption  subse- 
quent to  physical  dependence.  In:  Gross, 
M.M.,  ed.  Advances  in  Experimental 
Medicine  and  Biology,  Vol.  59:  Alcohol 
Intoxication  and  Withdrawal,  Experimental 
Studies  II.  New  York:  Plenum  Press, 
1975. pp.  373-378. 

Benjamin,  D.;  Grant,  E.R.;  and 
Pohorecky,  L.A.  Naltrexone  reverses  the 
ethanol-induced  dopamine  release  in  the 
nucleus  accumbens  in  awake,  freely  mov- 
ing rats.  Brain  Res  621:1 37-140,  1993. 

Benkelfat,  C.;  Murphy,  D.L.;  Hill,  J.L.; 
George,  D.T.;  Nutt,  D.;  and  Linnoila,  M. 
Ethanollike  properties  of  the  serotonergic 
partial  agonist  m-chlorophenylpiperazine 
in  chronic  alcoholic  patients.  Arch  Gen 
Psychiatry  48:383,  1991. 

Benzer,  D.G.  Management  of  alcohol 
intoxication  and  withdrawal.  In:  Miller, 
N.S.,  ed.  Principles  of  Addiction  Medicine. 
Chevy  Chase,  MD:  American  Society  of 
Addiction  Medicine,  1994. 

Bitran,  D.;  Hilvers,  R.J.;  and  Kellogg, 
C.K.  Anxiolytic  effects  of  3-alpha  hydroxy- 
5-alpha  [B]-pregnan-20-one:  Endogenous 
metabolites  of  progesterone  that  are 
active  at  the  GABA-A  receptor.  Brain  Res 
561:157-161,1991. 

Bjorkqvist,  S.E.  Clonidine  in  alcohol 
withdrawal.  Acta  Psychiatr  Scand  52:256- 
263,  1975. 

Blanchard,  R.J.;  Hori,  K.;  Tom,  P.;  and 
Blanchard,  D.C.  Social  structure  and 
ethanol  consumption  in  laboratory  rat. 
Pharmacol  Biochem  Behav  28:437-442, 
1987. 


Bokstrom,  K.,  and  Balldin,  J.  A  rating 
scale  for  assessment  of  alcohol  withdrawal 
paychopathology.  Alcohol  Clin  Exp  Res 
16:241-249,  1992. 

Branchey,  M.;  Rauscher,  G.;  and  Kissin, 
B.  Modifications  in  the  response  to  alcohol 
following  the  establishment  of  physical 
dependence.  Psychopharmacology  22:314- 
322,  1971. 

Broadbent,  J.;  Grahame,  N.J.;  and 
Cunningham,  C.L.  Haloperidol  prevents 
ethanol-stimulated  locomotor  activity  but 
fails  to  block  sensitization.  Psychopharma- 
cology  120:475-482,  1995. 

Brodie,  M.S.;  Shefner,  S.A.;  and  Dunwiddie, 
T.V.  Ethanol  increases  the  firing  rate  of 
dopamine  neurons  of  the  rat  ventral  tegmen- 
tal area.  Brain  Res  508:65-69,  1990. 

Brodie,  M.S.;  Trifunovic,  RD.;  and  Shefner, 
S.A.  Serotonin  potentiates  ethanol-in- 
duced excitation  of  ventral  tegmental  area 
neurons  in  brain  slices  from  three  differ- 
ent rat  strains.  J  Pharmacol  Exp  Ther 
273:1139-1146,1995. 

Brower,  K.J.;  Aldrich,  M.S.;  and  Hall, 
J.M.  Polysomnographic  and  subjective 
sleep  predictors  of  alcoholic  relapse.  Alcohol 
Clin  Exp  Res 22{8):\864-\87 1,  1998. 

Brown,  M.E.;  Anton,  R.F.;  Malcolm,  R.; 
and  Ballenger,  J.C.  Alcohol  detoxification 
and  withdrawal  seizures:  Clinical  support 
for  a  kindling  hypothesis.  Biol  Psychiatry 
23:507-514,  1988. 

Brown,  S.A.;  Vik,  P.W.;  Patterson,  T.L.; 
Grant,  I.;  and  Schuckit,  M.A.  Stress, 
vulnerability  and  adult  alcohol  relapse.  / 
Stud  Alcohol  56:538-545,  1995. 

Buck,  K.J.;  Heim,  H;  and  Harris,  R.A. 
Reversal  of  alcohol  dependence  and  toler- 
ance by  a  single  administration  of 
Qumzzenil.  J  Pharmacol  Exp  Ther 
257:984-989,  1991. 


292 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Burish,  T.G.;  Maisto,  S.A.;  Cooper,  A.M.; 
and  Sobell,  M.B.  Effects  of  voluntary  short- 
term  abstinence  from  alcohol  on  subsequent 
drinking  patterns  of  college  students./ 
Stud  Alcohol  42: 101 3-1020,  1981. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin, 
C.  Hormonal,  psychological,  and  alcohol 
craving  changes  after  m-chlorophenyl- 
piperazine  administration  in  alcoholics. 
Alcohol  Clin  Exp  Res  21:220-226,  1997. 

Campbell,  A.D.,  and  McBride,  W.J. 
Serotonin- 3  receptor  and  ethanol-stimu- 
lated  dopamine  release  in  the  nucleus 
accumbens.  Pharmacol  Biochem  Behav 
51:835-842,  1995. 

Campbell,  A.D.;  Kohl,  RR;  and  McBride, 
W.J.  Serotonin- 3  receptor  and  ethanol- 
stimulated  somatodendritic  dopamine 
release.  Alcohol  13:569-574,  1996. 

Cappell,  H.  Tolerance  to  ethanol  and 
treatment  of  its  abuse,  some  fundamental 
issues.  Addict  Behav  6:197-204,  1981. 

Carrington,  CD.;  Ellinwood,  E.H.;  and 
Krishnan,  R.R  Effects  of  single  and  re- 
peated alcohol  withdrawal  on  kindling. 
Biol  Psychiatry  19:525-537,  1984. 

Castaneda,  R,  and  Cushman,  P.  Alcohol 
withdrawal:  A  review  of  clinical  manage- 
ment. /  Clin  Psychiatry  50:278-284, 1989. 

Charlton,  M.E.;  Sweetnam,  P.M.;  Fitzgerald, 
L.W.;  Terwilliger,  R.Z.;  Nestler,  E.J.;  and 
Duman,  RS.  Chronic  ethanol  administra- 
tion regulates  the  expression  of  GABAA 
receptor  aY  and  a5  subunits  in  the  ventral 
tegmental  area  and  hippocampus.  / 
Neurochem  68:121-127,  1997. 

Chick,  J.  Acamprosate  as  an  aid  in  the 
treatment  of  alcoholism.  Alcohol  Alcohol 
30:785-787,  1995. 


Cicero,  T.J.  Alcohol  self-administration, 
tolerance  and  withdrawal  in  humans  and 
animals.  Theoretical  and  methodological 
issues.  In:  Rigter,  H.,  and  Crabbe,  J.C., 
eds.  Alcohol  Tolerance  and  Dependence. 
New  York:  Elsevier,  1980.  pp.  1-51. 

Clark,  C.P.;  Gillin,  J.C.;  Golshan,  S.; 
Demodena,  A.;  Smith,  T.L.;  Danowski, 
S.;  Irwin,  M.;  and  Schuckit,  M.  Increased 
REM  sleep  density  at  admission  predicts 
relapse  by  three  months  in  primary  alco- 
holics with  a  lifetime  diagnosis  of  secondary 
depression.  Biol  Psychiatry  43(8):601- 
607,  1998. 

Cloninger,  C.R  Neurogenetic  adaptive 
mechanisms  in  alcoholism.  Science  236:410- 
416,  1987. 

Cohen,  C;  Perrault,  G.;  and  Sanger,  D.J. 
Evidence  for  the  involvement  of  dopamine 
receptors  in  ethanol-induced  hyperactivity 
in  mice.  Neuropharmacology  36:1099- 
1108,1997. 

Concas,  A.;  Cuccheddu,  T.;  Floris,  S.; 
Mascia,  M.P.;  and  Biggio,  G.  2-Chloro- 
N6-cyclopentyl-adenosine  (CCPA),  an 
adenosine  A!  receptor  agonist,  suppresses 
ethanol  withdrawal  syndrome  in  rats. 
Alcohol  Alcohol  29:261-264,  1994. 

Cooney,  N.L.;  Litt,  M.D.;  Morse,  PA.; 
Bauer,  L.O.;  and  Gaupp,  L.  Alcohol  cue 
reactivity,  negative-mood  reactivity,  and 
relapse  in  treated  alcoholic  men.  / 
Abnorm  Psychol  106:243-250,  1997. 

Costa,  A.;  Bono,  G;  Martignoni,  E.;  Merlo, 
P.;  Sances,  G.;  and  Nappi,  G.  An  assessment 
of  hypothalamo-pituitary- adrenal  axis 
functioning  in  non-depressed,  early  absti- 
nent alcoholics.  Psychoneuroendocrinology 
21:263-275,  1996. 

Crabbe,  J.C.  Antagonism  of  ethanol  with- 
drawal convulsions  in  withdrawal  seizure 


293 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


prone  mice  by  diazepam  and  abecarnil. 
Eur  J  Pharmacol  221:85-90,  1992. 

Crawley,  J.N.;  Glowa,  J.R.;  Majewska, 
M.D.;  and  Paul,  S.M.  Anxiolytic  activity 
of  an  endogenous  adrenal  steroid.  Brain 
Res  398:382-385,  1986. 

Criswell,  H.E.;  Knapp,  D.J.;  Overstreet, 
D.H.;  and  Breese,  G.R.  Effects  of 
ethanol,  chlordiazepoxide,  and  MK-801 
on  performance  in  the  elevated  plus  maze 
and  on  locomotor  activity.  Alcohol  Clin 
Exp  Res  18:596-601,  1994. 

Darden,  J.H.,  and  Hunt,  W.A.  Reduction 
of  striatal  dopamine  release  during  an 
ethanol  withdrawal  syndrome.  / 
Neurochem  29:1143-1145,  1977. 

De  Soto,  C.B.;  O'Donnell,  W.E.;  Allred, 
L.J.;  and  Lopez,  C.E.  Symptomatology  in 
alcoholics  at  various  stages  of  abstinence. 
Alcohol  Clin  Exp  Res  9:505-5 12,  1985. 

Deutsch,  J. A.,  and  Koopmans,  H.S.  Prefer- 
ence enhancement  for  ethanol  by  passive 
exposure.  Science  179:1242-1243,  1973. 

Deutsch,  J.A.,  and  Walton,  N.Y.  A  rat 
alcoholism  model  in  a  free  choice  situa- 
tion. Behav  Biol  19:349-360,  1977. 

Devaud,  L.L.;  Purdy,  R.H.;  and  Morrow, 
A.L.  The  neurosteroid,  3a-hydroxy-5a- 
pregnan-20-one,  protects  against  bicu- 
culline-induced  seizures  during  ethanol 
withdrawal  in  rats.  Alcohol  Clin  Exp  Res 
19:350-355,  1995. 

de  Waele,  J. P.;  Kiianmaa,  K.;  and 
Gianoulakis,  C.  Distribution  of  the  mu 
and  delta  opioid  binding  sites  in  the  brain 
of  the  alcohol-preferring  AA  and  alcohol- 
avoiding  ANA  lines  of  rats.  /  Pharmacol 
Exp  Ther27S:  518-527,  1995. 

deWit,  H.,  and  Stewart,  J.  Reinstatement 
of  cocaine-reinforced  responding  in  the 
rat.  Psychopharmacology  75 : 1 34-143,  198 1 . 


Diana,  M.;  Gessa,  G.L.;  and  Rossetti, 
Z.L.  Lack  of  tolerance  to  ethanol-induced 
stimulation  of  mesolimbic  dopamine 
system.  Alcohol  Alcohol  27:329-333,  1992a. 

Diana,  M.;  Pistis,  M.;  Carboni,  S.;  Gessa, 
G.L.;  and  Rossetti,  Z.L.  Profound 
decrement  of  mesolimbic  neuronal  activ- 
ity during  ethanol  withdrawal  syndrome 
in  rats:  Electrophysiological  and  biochem- 
ical evidence.  Proc  Natl  Acad  Sci  USA 
90:7966-7969,  1992/;. 

Diana,  M.;  Pistis,  M.;  Muntoni,  A.; 
Rossetti,  Z.L.;  and  Gessa,  G.  Marked 
decrease  of  A10  dopamine  neuronal  firing 
during  ethanol  withdrawal  syndrome  in 
rats.  Eur  J  Pharmacol  221:403^04,  1993. 

Diana,  M.;  Pistis,  M.;  Muntoni,  A.;  and 
Gessa,  G.  Mesolimbic  dopaminergic 
reduction  outlasts  ethanol  withdrawal 
syndrome:  Evidence  of  protracted  absti- 
nence. Neuroscience  71:411-415,  1996. 

Di  Chiara,  G.;  Acquas,  E.;  and  Tanda,  G. 
Ethanol  as  a  neurochemical  surrogate  of 
conventional  reinforcers:  The  dopamine- 
opioid  link.  Alcohol  13:13-17,  1996. 

Dilts,  R.P.,  and  Kalivas,  P.W.  Autora- 
diographic localization  of  mu-opioid  and 
neurotensin  receptors  within  the  mesolim- 
bic dopamine  system.  Brain  Res 488: 
311-327, 1989. 

Dilts,  R.P.,  and  Kalivas,  P.W.  Autoradio- 
graphic localization  of  delta  opioid  receptors 
within  the  mesocorticolimbic  dopamine 
system  using  radioiodonated  [2-D-peni- 
cillamine ,  5  -  D  -penicillamine  ]  enkephalin 
(125I-DPDPE).  Synapse  6:121-132, 1990. 

Drummond,  S.P.;  Gillin,  J.C.;  Smith,  T.L.; 
and  DeModena,  A.  The  sleep  of  abstinent 
pure  primary  alcoholic  patients:  Natural 
course  and  relationship  to  relapse.  Alcohol 
Clin  Exp  Res  22(8):1796-1802,  1998. 


294 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Dunn,  A.}.,  and  Berridge,  C.W.  Physiological 
and  behavioral  responses  to  corticotropin- 
releasing  factor  administration:  Is  CRF  a 
mediator  of  anxiety  or  stress  responses? 
Brain  Res  Rev  15:71-100, 1990. 

Edwards,  G.  Withdrawal  symptoms  and 
alcohol  dependence:  Fruitful  mysteries. 
Br  J  Addict  85:447-461,  1990. 

Ehlers,  C.L.,  and  Chaplin,  R.I.  Chronic 
ethanol  exposure  potentiates  the  locomo- 
tor-activating  effects  of  corticotropin- 
releasing  factor  (CRF)  in  rats.  RegulPept 
19:345-353,  1987. 

Ehlers,  C.L.;  Chaplin,  R.I.;  Wall,  T.L.; 
Lumeng,  L.;  Li,  T.-K.;  Owens,  M.J.;  and 
Nemeroff,  C.B.  Corticotropin  releasing 
factor  (CRF)  studies  in  alcohol  preferring 
and  non-preferring  rats.  Psychopharmacology 
106:359-364,  1992. 

Ehrenreich,  H.;  Schuck,  J.;  Stender,  N.; 
Pilz,  J.;  Gefeller,  O.;  Schilling,  L.;  Poser, 
W.;  and  Kaw,  S.  Endocrine  and  hemody- 
namic effects  of  stress  versus  systemic 
CRF  in  alcoholics  during  early  and 
medium  term  abstinence.  Alcohol  Clin 
E^ito21:1285-1293,  1997. 

Emmett-Oglesby,  M.W.;  Mathis,  D.A.; 
Moon,  R.T.Y.;  and  Lai,  H.  Animal  mod- 
els of  drug  withdrawal  symptoms. 
Psychopharmacology  101:292-309,  1990. 

Engel,  J.A.;  Enerback,  C;  Fahlke,  C; 
Hulthe,  P.;  Hard,  E.;  Johannessen,  K.; 
Svensson,  L.;  and  Soderpalm,  B. 
Serotonergic  and  dopaminergic  involve- 
ment in  ethanol  intake.  In:  Naranjo,  C.A., 
and  Sellers,  E.M.,  eds.  Novel  Pharmaco- 
logical Interventions  for  Alcoholism.  New 
York:  Springer,  1992.  pp.  68-82. 

Fadda,  F.;  Argiolas,  A.;  Melis,  M.R; 
Serra,  G.;  and  Gessa,  G.L.  Differential 
effect  of  acute  and  chronic  ethanol  on 
dopamine  metabolism  in  frontal  cortex, 


caudate  nucleus  and  substantia  nigra.  Life 
Sci  27:979-986,  1980. 

Fadda,  F.;  Garau,  B.;  Marchei,  F.; 
Colombo,  G.;  and  Gessa,  G.L.  MDL 
72222,  a  selective  5 -HT3  receptor  antago- 
nist, suppresses  voluntary  ethanol  con- 
sumption in  alcohol-preferring  rats. 
Alcohol  Alcohol  26:107-110,  1991. 

Fahlke,  C;  Hansen,  S.;  Engel,  J.A.;  and 
Hard,  E.  Effects  of  ventral  striatal  6-OHDA 
lesions  or  amphetamine  sensitization  on 
ethanol  consumption  in  the  rat.  Pharmacol 
Biochem  Behav  47:345-349,  1994. 

File,  S.E.  The  use  of  social  interaction  as  a 
method  for  detecting  anxiolytic  activity  of 
chlordiazepoxide-like  drugs.  /  Neurosci 
Methods  2:219-238,  1980. 

File,  S.E.;  Zharkovsky,  A.;  and  Gulati,  K. 
Effects  of  baclofen  and  nitrendipine  on 
ethanol  withdrawal  responses  in  the  rat. 
Neuropharmacology  30:183-190,  1991. 

File,  S.E.;  Andrews,  N.;  and  Al-Farhan, 
M.  Anxiogenic  responses  of  rats  on  with- 
drawal from  chronic  ethanol  treatment: 
Effects  of  tianeptine.  Alcohol  Alcohol 
28:281-286,  1993. 

Files,  F.J.;  Lewis,  R.S.;  and  Samson,  H.H. 
Ethanol  self- administration  by  alcohol- 
nonp referring  (NP)  rats  in  a  continuous 
access  situation.  Alcohol  10:139-144,  1993. 

Finn,  D.A.;  Roberts,  A.J.;  and  Crabbe,  J.C.. 
Neuroactive  steroid  sensitivity  in  with- 
drawal seizure-prone  and  -resistant  mice. 
Alcohol  Clin  Exp  Res  19:410-415,  1995. 

Freund,  G.  Alcohol  withdrawal  syndrome 
in  mice.  Arch  Neurol  21:315-320,  1969. 

Froehlich,  J.C.;  Harts,  J.;  Lumeng,  L.; 
and  Li,  T.K.  Naloxone  attenuation  of 
voluntary  alcohol  consumption.  Alcohol 
Alcohol  Suppl  1:333-337,  1987. 


295 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Gatto,  G.J.;  McBride,  W.J.;  Murphy, 
J.M.;  Lumeng,  L.;  and  Li,  T.-K.  Ethanol 
self- infusion  into  the  ventral  tegmental 
area  by  alcohol-preferring  rats.  Alcohol 
11:557-564,  1994. 

George,  S.R.;  Fan,  T.;  Ng,  G.Y.;  Jung, 
S.Y.;  O'Dowd,  B.F.;  and  Naranjo,  C.A. 
Low  endogenous  dopamine  function  in 
brain  predisposes  to  high  alcohol  prefer- 
ence and  consumption:  Reversal  by  in- 
creasing synaptic  dopamine.  J  Pharmacol 
Exp  Ther  273:373-379,  1995. 

Gessa,  G.L.;  Muntoni,  F.;  Collu,  M.; 
Vargiu,  L.;  and  Mereu,  G.  Low  doses  of 
ethanol  activate  dopaminergic  neurons  of 
the  ventral  tegmental  area.  Brain  Res 
348:201-203,  1985. 

Gianoulakis,  C;  de  Waele,  J. P.;  and 
Kiianmaa,  K.  Differences  in  the  brain  and 
pituitary  (3-endorphin  system  between  the 
alcohol-preferring  AA  and  alcohol-avoid- 
ing ANA  rats.  Alcohol  Clin  Exp  Res 
16:453-459,  1992. 

Gillin,  J.C.;  Smith,  T.L.;  Irwin,  M.;  Kripke, 
D.F.;  and  Schuckit,  M.  EEG  sleep  studies 
in  "pure"  primary  alcoholism  during  suba- 
cute withdrawal:  Relationships  to  normal 
controls,  age,  and  other  clinical  variables. 
Biol  Psychiatry  27:477^88,  1990. 

Goddard,  G.V.;  Mclntyre,  D.C.;  and 
Leech,  C.K.  A  permanent  change  in  brain 
function  resulting  from  daily  electrical 
stimulation.  Exp  Neurol 25:295-330,  1969. 

Goeders,  N.E.,  and  Guerin,  G.F.  Non- 
contingent  electric  footshock  facilitates 
the  acquisition  of  intravenous  cocaine  self- 
administration  in  rats.  Psychopharmacology 
114:63-70,1994. 

Gongwer,  M.A.;  Murphy,  J.M.;  McBride, 
W.J.;  Lumeng,  L.;  and  Li,  T.K.  Regional 
brain  contents  of  serotonin,  dopamine 


and  their  metabolites  in  the  selectively 
bred  high-  and  low- alcohol  drinking  lines 
of  rats.  Alcohol  6:317-320,  1989. 

Gonzales,  R.A.,  and  Weiss,  F.  Suppression 
of  ethanol -reinforced  behavior  by  naltrex- 
one is  associated  with  attenuation  of  the 
ethanol-induced  increase  in  dialysate 
dopamine  levels  in  the  nucleus  accum- 
bens.  / Neurosci  18:10663-10671,  1998. 

Grant,  K.A.  Animal  models  of  the  alcohol 
addiction  process.  In:  Kranzler,  H.R.,  ed. 
Handbook  of  Experimental  Pharmacology, 
Vol.  114:  The  Pharmacology  of  Alcohol 
Abuse.  Berlin:  Springer  Verlag,  1995.  pp. 
185-229. 

Grant,  K.A.,  and  Barrett,  J.E.  Blockade  of 
the  discriminative  stimulus  and  anxiolytic 
effects  of  ethanol  with  5-HT-3  receptor 
antagonists.  In:  Racagni,  G.;  Brunello, 
N.;  and  Fukuda,  T.,  eds.  International 
Congress  Series,  Vol.  968:  Biological 
Psychiatry.  New  York:  Excerpta  Medica, 
\99la.  pp.  11-13. 

Grant,  K.A.,  and  Barrett,  J.E.  Blockade  of 
the  discriminative  stimulus  effects  of 
ethanol  with  5-HT3-receptor  antagonists. 
Psychopharmacology  104:451^-56,  1991  b. 

Grant,  K.A.,  and  Colombo,  G.  Pharma- 
cological analysis  of  the  mixed  discriminative 
stimulus  effects  of  ethanol.  Alcohol  Alcohol 
Suppl  2:445-449,  1993^. 

Grant,  K.A.,  and  Colombo,  G.  Substitution 
of  the  5-HTj  agonist  trifluoromethylphenyl- 
piperazine  (TFMPP)  for  the  discrimina- 
tive stimulus  effects  of  ethanol:  Effect 
of  training  dose.  Psychopharmacology 
113:26-30,  19936. 

Grant,  K.A.;  Valverius,  P.;  Hudspith,  M.; 
and  Tabakoff,  B.  Ethanol  withdrawal 
seizures  and  the  NMD  A  receptor  complex. 
Eur  J  Pharmacol  176:289-296,  1990. 


296 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Gray,  T.S.  Amygdaloid  CRF  pathways. 
Role  in  autonomic,  neuroendocrine,  and 
behavioral  responses  to  stress.  Ann  NY 
Acad  Sci  697:53-60,  1993. 

Haney,  M.;  Maccari,  S.;  Le  Moal,  M.; 
Simon,  H.;  and  Piazza,  P.V.  Social  stress 
increases  the  acquisition  of  cocaine  self- 
administration  in  male  and  female  rats. 
Brain  Res  698:46-52,  1995. 

Hawley,  R.J.;  Nemeroff,  C.B.;  Bissette, 
G.;  Guidotti,  A.;  Rawlings,  R;  and  Linnoila, 
M.  Neurochemical  correlates  of  sympathetic 
activation  during  severe  alcohol  withdrawal. 
Alcohol  Clin  Exp  Res  18:1312-1316,  1994. 

Heather,  N.,  and  Stallard,  A.  Does  the 
Marlatt  model  underestimate  the  impor- 
tance of  conditioned  craving  in  the  relapse 
process?  In:  Gossop,  M.,  ed.  Relapse  and 
Addictive  Behaviour.  London:  Tavistock/ 
Routledge,  1989. 

Heinrichs,  S.C.;  Merlo  Pich,  E.;  Miczek, 
K.A.;  Britton,  K.T.;  and  Koob,  G.F. 
Corticotropin-releasing  factor  reduces 
emotionality  in  socially  defeated  rats  via 
direct  neurotropic  action.  Brain  Res 
581:190-197,  1992. 

Heinz,  A.;  Dettiing,  M.;  Kuhn,  S.;  Dufeu, 
P.;  Graf,  K.J.;  Kurten,  I.;  Rommelspacher, 
H.;  and  Schmidt,  I.G.  Blunted  growth 
hormone  response  is  associated  with  early 
relapse  in  alcohol-dependent  patients. 
Alcohol  Clin  Exp  Res  19:62-65,  \99Sa. 

Heinz,  A.;  Lichtenberg-Kraag,  B.;  Baum, 
S.S.;  Graf,  K.;  Kruger,  R;  Dettiing,  M.; 
and  Rommelspacher,  H.  Evidence  for  pro- 
longed recovery  of  dopaminergic  trans- 
mission after  detoxification  in  alcoholics 
with  poor  treatment  outcome.  J  Neural 
Transm  Gen  Sect  102:149-157,  1995&. 


Hershon,  H.I.  Alcohol  withdrawal  symp- 
toms and  drinking  behavior.  J  Stud  Alcohol 
38:953-971,  1977. 

Heyne,  A.;  Swodlowska,  G.;  and 
Wolffgramm,  J.  Physical  dependence  on 
ethanol  vs.  rebound  phenomena  in  an 
animal  model  of  alcoholism.  Naunyn 
Schmiedebergs  Arch  Pharmacol  Suppl 
344:R70,  1991. 

Heyser,  C.J.,  and  Koob,  G.F.  Cues  associ- 
ated with  ethanol  self- administration 
function  as  conditioned  reinforcers  in  rats. 
Alcohol  Clin  Exp  Res2\:9K,  1997. 

Heyser,  C.J.;  Schulteis,  G.;  Durbin,  P.;  and 
Koob,  G.F.  Chronic  acamprosate  elimi- 
nates the  alcohol  deprivation  effect  while 
having  limited  effects  of  baseline  respond- 
ing for  ethanol  in  rats.  Neuropsychopharma- 
cology  18:125-133, 1996. 

Heyser,  C.J.;  Schulteis,  G.;  and  Koob, 
G.F.  Increased  ethanol  self- administration 
after  a  period  of  imposed  ethanol  deprivation 
in  rats  trained  in  a  limited  access  paradigm. 
Alcohol  Clin  Exp  Res  2 1:784-791,  1997. 

Higley,  J.D.;  Hasert,  M.F.;  Suomi,  S.J.; 
and  Linnoila,  M.  Nonhuman  primate 
model  of  alcohol  abuse:  Effect  of  early 
experience,  personality,  and  stress  on 
alcohol  consumption.  Proc  Natl  Acad  Sci 
USA  88:7261-7265,  1991. 

Hilakivi-Clarke,  L.,  and  Lister,  R  Social 
status  and  voluntary  alcohol  consumption 
in  mice:  Interaction  with  stress.  Psycho- 
pharmacology  108:276-282,  1992. 

Hodge,  C.W.;  Samson,  H.H.;  Lewis, 
R.S.;  and  Erickson,  H.L.  Specific  decreases 
in  ethanol-  but  not  water-reinforced  respond- 
ing produced  by  the  5  -HT3  antagonist 
ICS  205-930.  Alcohol  10:191-196,  1993. 

Hoffman,  P.L.,  and  Tabakoff,  B.  The  role 
of  the  NMDA  receptor  in  ethanol  with- 


297 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


drawal.  In:  Jansson,  B.;  Hornvall,  H.; 
Rydberg,  U.;  Terenius,  L.;  and  Vallee, 
B.L.,  eds.  Experientia  Supplementum,  Vol. 
71:  Toward  a  Molecular  Basis  of  Alcohol 
Use  and  Abuse.  Basel,  Switzerland: 
Birkhauser  Verlag,  1994.  pp.  61-70. 

Holter,  S.M.;  Danysz,  W.;  and  Spanagel, 
R  Evidence  for  alcohol  anti-craving  prop- 
erties of  memantine.  Eur  J  Pharmacol 
314:R1-R2,  1996. 

Holter,  S.M.;  Landgr>f,  R;  Zieglgansberger, 
W.;  and  Spanagel,  R  i^me  course  of 
acamprosate  action  on  operant  ethanol 
self- administration  after  ethanol  deprivation. 
Alcohol  Clin  Exp  Res  21:862-868,  1997. 

Hope,  B.;  Kosofsky,  B.;  Hyman,  S.E.;  and 
Nestler,  E.J.  Regulation  of  immediate  early 
gene  expression  and  AP-1  binding  in  the 
rat  nucleus  accumbens  by  chronic  cocaine. 
Proc  Natl  Acad  Sci  USA  89:5764-5768, 
1992. 

Hunt,  W.A.,  and  Lands,  W.E.M.  A  role 
for  behavioral  sensitization  in  uncontrolled 
ethanol  intake.  Alcohol 9:327-328,  1992. 

Hunter,  B.E.;  Walker,  D.W.;  and  Filey, 
J.N.  Dissociation  between  physical  depen- 
dence and  volitional  ethanol  consumption: 
Role  of  multiple  withdrawal  episodes.  Pharm- 
acol Biochem  Behav  2:523-529,  1974. 

Hyman,  S.E.  Addiction  to  cocaine  and 
amphetamine.  Neuron  16:901-904,  1996. 

Imperato,  A.,  and  DiChiara,  G.  Preferential 
stimulation  of  dopamine  release  in  the 
nucleus  accumbens  of  freely  moving  rats 
by  ethanol.  /  Pharmacol  Exp  Ther  239:219- 
228,  1986. 

Imperato,  A.;  Angelucci,  L.;  Casolini,  P.; 
Zocchi,  A.;  and  Puglisi-Allegra,  S.  Repeated 
stressful  experiences  differently  affect 
limbic  dopamine  release  during  and  follow- 
ing stress.  Brain  Res  577:194-199,  1992. 


Inder,  W.J.;  Joyce,  P.R;  Ellis,  M.J.;  Evans, 
M.J.;  Livesey,  J.H.;  and  Donald,  RA. 
The  effects  of  alcoholism  on  the  hypothal- 
amic-pituitary- adrenal  axis:  Interaction 
with  endogenous  opioid  peptides.  Clin 
Endocrinol  (Chef)  43:283-290,  1995. 

Jaffe,  J.H.;  Cascella,  N.G.;  Kumor,  K.M.; 
and  Sherer,  MA.  Cocaine -induced  cocaine 
craving.  Psychopharmacolqjjy  97:59-64, 1989. 

Johnson,  B.A.;  Campling,  G.M.;  Griffiths, 
P.;  and  Cowen,  P.J.  Attenuation  of  some 
alcohol-induced  mood  changes  and  the 
desire  to  drink  by  5 -HT3  receptor  block- 
ade: A  preliminary  study  in  healthy  male 
volunteers.  Psychopharmacology  112: 
142-144,  1993. 

June,  H.L.;  Lummis,  G.H.;  Colker,  R.E.; 
Moore,  T.O.;  and  Lewis,  M.J.  Ro  15- 
4513  attenuates  the  consumption  of 
ethanol  in  deprived  rats.  Alcohol  Clin  Exp 
Res  15:406-411,  1991. 

June,  H.L.;  Murphy,  J.M.;  Mellor-Burke, 
J.J.;  Lumeng,  L.;  and  Li,  T.-K.  The  benzo- 
diazepine inverse  agonist  RO  19-4603  exerts 
prolonged  and  selective  supression  of  ethanol 
intake  in  alcohol-preferring  (P)  rats.  Psycho- 
pharmacology  115:325-331,  1994a. 

June,  H.L.;  Hughes,  R.W.;  Spurlock,  H.L.; 
and  Lewis,  M.J.  Ethanol  self- administra- 
tion in  freely  feeding  and  drinking  rats: 
Effects  of  Rol5-4513  alone,  and  in  com- 
bination with  Rol5-1788  (flumazenil). 
Psychopharmacology  115:332-339,  1994b. 

Kahn,  A.J.,  and  Scudder,  C.L.  Alcohol 
withdrawal  effects  on  brain  serotonin  in 
mice  behaviorally  attracted  to  alcohol.  / 
Stud  Alcohol  37:1572-1580,  1976. 

Kalivas,  P.W.,  and  Duffy,  P.  Similar  effects 
of  daily  cocaine  and  stress  on  mesocorti- 
colimbic  dopamine  neurotransmission  in 
the  rat.  Biol  Psychiatry  25:913-928,  1989. 


298 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Kalivas,  P.W.,  and  Stewart,  J.  Dopamine 
transmission  in  the  initiation  and  expression 
of  drug-  and  stress-induced  sensitization 
of  motor  activity.  Brain  Res  Rev  16:223- 
244,1991. 

Kang,  M.;  Spigelman,  I.;  Sapp,  D.W.;  and 
Olsen,  R.W.  Persistent  reduction  of 
GABA(A)  receptor-mediated  inhibition  in 
rat  hippocampus  after  chronic  intermittent 
ethanol  treatment.  Brain  Res  709:221- 
228,  1996. 

Kaplan,  R.F.;  Meyer,  R.E.;  and  Stroebel, 
C.F.  Alcohol  dependence  and  responsivity 
to  an  ethanol  stimulus  as  predictors  of 
alcohol  consumption.  Br  J  Addict  7 '8:259- 
267,  1983. 

Kaplan,  R.F.;  Meyer,  R.E.;  and  Virgilio, 
L.M.  Physiological  reactivity  to  alcohol 
cues  and  the  awareness  of  an  alcohol 
effect  in  a  double-blind  placebo  design. 
Br  J  Addict  79:439-442,  1984. 

Kaplan,  R.F.;  Cooney,  N.L.;  Baker,  L.H.; 
Gillespie,  RA.;  Meyer,  RE.;  and  Pomerleau, 
O.F.  Reactivity  to  alcohol-related  cues: 
Physiological  and  subjective  responses  in 
alcoholics  and  nonproblem  drinkers.  / 
Stud  Alcohol  46:267-272,  1985. 

Katner,  S.N.;  Kerr,  T.M.;  and  Weiss,  F. 
Ethanol  anticipation  enhances  dopamine 
efflux  in  the  nucleus  accumbens  of  alco- 
hol preferring  (P)  but  not  Wistar  rats. 
Behav  Pharmacol  7:669-674,  1996. 

Katner,  S.N.;  Magalong,  J.G.;  and  Weiss, 
F.  Reinstatement  of  alcohol-seeking  be- 
havior by  drug- associated  discriminative 
stimuli  after  prolonged  extinction  in  the 
rat.  Neuropsychopharmacology  20:471- 
479, 1999. 

Kempf,  E.;  Kempf,  J.;  and  Ebel,  A. 
Alcohol  withdrawal-induced  changes  in 
brain  biogenic  amines  in  mice:  Influence 


of  the  genotype.  Neurochem  Res  15:69- 
75,  1990. 

Khachaturian,  H.;  Schaefer,  M.K.H.;  and 
Lewis,  M.E.  Anatomy  and  function  of  the 
endogenous  opioid  system.  In:  Akil,  H.,  and 
Simon,  E.J.,  eds.  Handbook  of  Experimental 
Pharmacology,  Vol.  104:  Opioids.  New  York: 
Springer  Verlag,  1993.  pp.  471^79. 

Kiianmaa,  K,  and  Saito,  T.  The  AA  and 
ANA  rat  lines  as  a  tool  for  identifying  the 
biological  mechanisms  of  alcohol  abuse. 
In:  Racagni,  G.,  Brunello,  N.,  and  Fukuda, 
T.,  eds.  International  Congress  Series,  Vol. 
968:  Biological  Psychiatry.  New  York: 
Excerpta  Medica,  1991.  pp.  7-9. 

Kiianmaa,  K;  Stenius,  K;  and  Sinclair, 
J.D.  Determinants  of  alcohol  preference 
in  the  AA  and  ANA  rat  lines  selected  for 
differential  ethanol  intake.  Alcohol  Alcohol 
Suppl  1:115-120,  1991. 

Kiianmaa,  K;  Hyytia,  P.;  and  Sinclair,  J.D. 
Development  of  an  animal  model  of  ethanol 
abuse:  Genetic  approach.  In:  Boulton,  A.; 
Baker,  G.;  and  Wu,  P.H.,  eds.  Animal 
Models  of  Drug  Addiction.  Totowa,  NJ: 
Humana  Press,  1992.  pp.  29-63. 

Kiianmaa,  K;  Nurmi,  M.;  Nykanen,  I.; 
and  Sinclair,  J.D.  Effect  of  ethanol  on 
extracellular  dopamine  in  the  nucleus 
accumbens  of  alcohol-preferring  AA  and 
alcohol-avoiding  ANA  rats.  Pharmacol 
Biochem  Behav  52:29-34,  1995. 

Kim,  W.-K;  Johnson,  R.G.;  Izu,  L.T.;  and 
Rabin,  RA.  Chronic  ethanol  exposure 
inhibits  ATP-stimulated  but  not  KC1- 
stimulated  dopamine  release  in  PC  12  cells. 
J  Pharmacol  Exp  Ther  270:336-341,  1994. 

Kleven,  M.;  Ybema,  C;  Carilla,  E.;  Hamon, 
M.;  and  Koek,  W.  Modification  of  behav- 
ioral effects  of  8-hydroxy-2-(di-n-propyl- 
amino)tetralin  following  chronic  ethanol 
consumption  in  the  rat:  Evidence  for  the 


299 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


involvement  of  5-HT1A  receptors  in 
ethanol  dependence.  Eur  J  Pharmacol 
281:219-228,  1995. 

Knapp,  D.J.,  and  Pohorecky,  LA. 
Zacopride,  a  5-HT3  receptor  antagonist, 
reduces  voluntary  ethanol  consumption  in 
rats.  Pharmacol  Biochem  Behav  41 :847- 
850,  1992. 

Kokka,  N.;  Sapp,  D.W.;  Taylor,  A.M.; 
and  Olsen,  R.W.  The  kindling  model  of 
alcohol  dependence:  Similar  persistent 
reduction  in  seizure  threshold  to  pentylenete- 
trazol in  animals  receiving  chronic  ethanol 
or  chronic  pentylenetetrazol.  Alcohol  Clin 
Exp  Res  17:525-531,  1993. 

Koob,  G.F.  Drug  addiction:  The  yin  and 
yang  of  hedonic  homeostasis.  Neuron 
16:893-896,  1996. 

Koob,  G.F.,  and  Bloom,  F.E.  Cellular  and 
molecular  mechanisms  of  drug  depen- 
dence. Science  242:715-723,  1988. 

Koob,  G.  F.,  and  Britton,  K.T. 
Neurobiological  substrates  for  the  anti- 
anxiety effects  of  ethanol.  In:  Begleiter, 
PL,  and  Kissin,  B.,  eds.  The  Pharmacology 
of  Alcohol  and  Alcohol  Dependence.  New 
York:  Oxford  University  Press,  1996.  pp. 
477-506. 

Koob,  G.F.,  and  Cador,  M.  Psychomotor 
stimulant  sensitization:  The  corticotropin- 
releasing  factor- steroid  connection.  Behav 
Pharmacol  4:351-354,  1993. 

Koob,  G.F.,  and  Le  Moal,  M.  Drug 
abuse:  Hedonic  homeostatic  dysregula- 
tion.  Science  278:52-58,  1997. 

Koob,  G.  F.;  Braestrup,  C.;  and  Thatcher- 
Britton,  K.  The  effects  of  FG  7142  and 
RO  15-1788  on  the  release  of  punished 
responding  produced  by  chlordiazepoxide 
and  ethanol  in  the  rat.  Psychopharmacology 
90:173-178,  1986. 


Koob,  G.F.;  Markou,  A.;  Weiss,  F.;  and 
Schulteis,  G.  Opponent  process  and  drug 
dependence:  Neurobiological  mecha- 
nisms. Semin  Neurosci  5:351-358,  1993. 

Koob,  G.F.;  Heinrichs,  S.C.;  Menzaghi, 
F.;  Pich,  E.M.;  and  Britton  K.T. 
Corticotropin  releasing  factor,  stress 
and  behavior.  Seminars  in  the 
Neurosciences  6:  221-229,  1994. 

Kornet,  M.;  Goosen,  C;  and  Van  Ree, 
J.M.  The  effect  of  interrupted  alcohol 
supply  on  spontaneous  alcohol  consump- 
tion by  rhesus  monkeys.  Alcohol  Alcohol 
25:407-412,  1990. 

Kornet,  M.;  Goosen,  O;  and  Van  Ree, 
J.M.  Effect  of  naltrexone  on  alcohol 
consumption  during  chronic  alcohol 
drinking  and  after  a  period  of  imposed 
abstinence  in  free-choice  drinking  rhesus 
monkeys.  Psychopharmacology  104: 
367-376,  1991. 

Kotlinska,  J.,  and  Liljequist,  S.  Oral  ad- 
ministration of  glycine  and  polyamine 
receptor  antagonists  blocks  ethanol  with- 
drawal seizures.  Psychopharmacology 
127:238-244,  1996. 

Kraemer,  G.W.,  and  McKinney,  W.T. 
Social  separation  increases  alcohol  con- 
sumption in  rhesus  monkeys.  Psycho- 
pharmacology  86:182-189,  1985. 

Krank,  M.D.,  and  Wall,  A.M.  Cue  expo- 
sure during  a  period  of  abstinence  reduces 
the  resumption  of  operant  behavior  for 
oral  ethanol  reinforcement.  Behav 
Neurosci  104:725-733,  1990. 

Krystal,  J.H.;  Webb,  E.;  Cooney,  N.; 
Rranzler,  H.R.;  and  Charney,  D.S. 
Specificity  of  ethanollike  effects  elicited  by 
serotonergic  and  noradrenergic  mechanisms. 
Arch  Gen  Psychiatry  51:898-911,  1994. 

Kuriyama,  K;  Ueha,  T.;  Hirouchi,  M.; 
Hashimoto,  T.;  and  Ohkuma,  S. 


300 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Functional  alterations  in  GABAA  receptor 
complex  induced  by  ethanol.  Alcohol 
Alcohol  Suppl  2:321-325,  1993. 

Laberg,  J.C.  Alcohol  and  expectancy: 
Subjective,  psychophysiological  and  be- 
havioral responses  to  alcohol  stimuli  in 
severely,  moderately  and  non-dependent 
drinkers.  Br  J  Addict  81:797-808,  1986. 

Laberg,  J.C,  and  Ellertsen,  B.  Psycho- 
physiological indicators  of  craving  in 
alcoholics:  Effects  of  cue  exposure.  Br  J 
Addict  82:1341-134%,  1987. 

Lai,  H.;  Harris,  CM.;  Benjamin,  D.; 
Springfield,  A.C.;  Bhadra,  S.;  and  Emmett- 
Oglesby,  M.W.  Characterization  of  a 
pentylenetetrazol-like  interoceptive  stimu- 
lus produced  by  ethanol  withdrawal.  / 
Pharmacol  Exp  Ther  247:508-518,  1988. 

Lai,  H.;  Prather,  P.L.;  and  Rezazadeh,  S.M. 
Anxiogenic  behavior  in  rats  during  acute 
and  protracted  ethanol  withdrawal:  Reversal 
by  buspirone.  Alcohol  8:467-471,  1991. 

Lai,  PL;  Prather,  P.L.;  and  Rezazadeh, 
S.M.  Potential  role  of  5-HTlc  and/or  5- 
HT2  receptors  in  the  mianserin-induced 
prevention  of  anxiogenic  behaviors  occur- 
ring during  ethanol  withdrawal.  Alcohol 
Clin  Exp  Res  17 AU-417,  1993. 

Lamblin,  F.;  Meert,  T.F.;  and  de  Witte, 
P.  Adrenalectomy  protects  ethanol-with- 
drawn  rats  from  harmine-induced  tremor. 
Alcohol  Alcohol  31:175-181,  1996. 

Lankford,  M.F.,  and  Myers,  R.D. 
Genetics  of  alcoholism:  Simultaneous 
presentation  of  a  chocolate  drink  dimin- 
ishes alcohol  preference  in  high  drinking 
FLAD  rats.  Pharmacol  Biochem  Behav 
49:417-425,  1994. 

Lankford,  M.F.;  Roscoe,  A.K.; 
Pennington,  S.N.;  and  Myers,  R.D. 
Drinking  of  high  concentrations  of 
ethanol  versus  palatable  fluids  in  alcohol- 


preferring  (P)  rats:  Valid  animal  model  of 
alcoholism.  Alcohol  8:293-299,  1991. 

Le,  A.D.;  Quan,  B.;  Juzytch,  W.; 
Fletcher,  P.J.;  Joharchi,  N.;  and  Shaham, 
Y.  Reinstatement  of  alcohol-seeking  by 
priming  injections  of  alcohol  and  expo- 
sure to  stress  in  rats.  Psychopharmacology 
(Berl)  135:169-174,  1998. 

Lee,  M.A.,  and  Meltzer,  H.Y.  Neuro- 
endocrine responses  to  serotonergic 
agents  in  alcoholics.  Biol  Psychiatry 
30:1017-1030,  1991. 

Legault,  M.,  and  Wise,  RA.  Effects  of 
withdrawal  from  nicotine  on  intracranial 
self-stimulation.  Soc  Ncurosci  Abstr 
20:1032,  1994. 

Leith,  N.J.,  and  Barrett,  R.J. 
Amphetamine  and  the  reward  system: 
Evidence  for  tolerance  and  post-drug 
depression.  Psychopharmacologia 
46:19-25,  1976. 

LeMagen,  J.  Etude  de  quelques  facteurs 
associe  a  des  modifications  de  la  consom- 
mation  spontanee  dalcool  ethylique  par 
le  rat.  ] Physiol  Paris  52:873-884,  1960. 

LeMarquand,  D.;  Pihl,  R.O.;  and  Benkelfat, 
C.  Serotonin  and  alcohol  intake,  abuse, 
and  dependence:  Clinical  evidence.  Biol 
Psychiatry  36:326-337,  1994^. 

LeMarquand,  D.;  Pihl,  R.O.;  and  Benkelfat, 
C.  Serotonin  and  alcohol  intake,  abuse, 
and  dependence:  Findings  of  animal  stud- 
ies. Biol  Psychiatry  36:395-421,  1994b. 

Leslie,  S.W.;  Woodward,  J.J.;  Wilcox, 
RE.;  and  Farrar,  RP.  Chronic  ethanol 
treatment  uncouples  striatal  calcium  entry 
and  endogenous  dopamine  release.  Brain 
Res  368:174-177,  1986. 

Lester,  D.,  and  Freed,  E.X.  Criteria  for  an 
animal  model  of  alcoholism.  Pharmacol 
Biochem  Behav  1:103-107,  1973. 


301 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Lewis,  M.E.;  Khachaturian,  H.;  and 
Watson,  S.J.  Comparative  distribution  of 
opiate  receptors  and  three  opioid  peptide 
neuronal  systems  in  rhesus  monkey  central 
nervous  system.  Life  Sci  1:239-242,  1983. 

Lewis,  M.J.  Alcohol  effects  on  brain- 
stimulation  reward:  Blood  alcohol  con- 
centration and  site  specificity.  In:  Meyer, 
R.;  Koob,  G.F.;  Lewis,  M.;  and  Paul,  S., 
eds.  The  Neuropharmacology  ofEthanol: 
New  Approaches.  Boston:  Birkhauser,  1991. 
pp.  163-178. 

Lewis,  M.J.,  and  June,  H.L.  Neuro- 
behavioral  studies  of  ethanol  reward  and 
activation.  Alcohol  7:213-219,  1990. 

Lhuintre,  J. P.;  Daoust,  M.;  Moore,  N.D.; 
Chretien,  P.;  Saligaut,  C;  Tran,  G.; 
Boismare,  F.;  and  Hillemand,  B.  Ability  of 
calcium  bis  acetyl  homotaurine,  a  GAB  A 
agonist,  to  prevent  relapse  in  weaned 
alcoholics.  Lancet  1:1014-1016,  1985. 

Lhuintre,  J. P.;  Moore,  N.;  Tran,  G.; 
Steru,  L.;  Langrenon,  S.;  Daoust,  M.; 
Parot,  P.;  Ladure,  P.;  Libert,  C;  Boismare, 
F.;  et  al.  Acamprosate  appears  to  decrease 
alcohol  intake  in  weaned  alcoholics. 
Alcohol  Alcohol  25:613-622,  1990. 

Li,  T.K.,  and  McBride,  W.J.  Pharma- 
cogenetic  models  of  alcoholism.  Clin 
Neurosci  3:182-188,  1995. 

Li,  T.K.;  Lumeng,  L.;  McBride,  W.J.;  and 
Waller,  M.B.  Progress  towards  a  voluntary 
oral  consumption  model  of  alcoholism. 
Drug  Alcohol  Depend  4:45-60,  1979. 

Li,  T.K.;  Lumeng,  L.;  McBride,  W.J.; 
Waller,  M.B.;  and  Murphy,  J.M.  Studies 
on  an  animal  model  of  alcoholism.  NIDA 
ResMonogr  66:41-49,  1986. 

Liljequist,  S.,  and  Engel,  JA.  Reversal  of 
the  anti  conflict  action  of  valproate  by 
various  GABA  and  benzodiazepine  antag- 
onists. Life  Sci  34:2525-2533,  1984. 


Linnoila,  M.;  Virkkunen,  M.;  Scheinin,  M.; 
Nuutila,  A.;  Rimon,  R.;  and  Goodwin,  F. 
Low  cerebrospinal  fluid  5-hydroxyin- 
doleacetic  acid  concentration  differenti- 
ates impulsive  from  nonimpulsive  violent 
behavior.  Life  Sci  33:2609-2614,  1983. 

Lister,  R.G.  The  use  of  a  plus-maze  to 
measure  anxiety  in  the  mouse.  Psycho- 
pharmacology  92:180-185,  1987. 

Lister,  RG.  Interaction  of  three  benzodiaze- 
pine receptor  inverse  agonists  with  ethanol 
in  a  plus-maze  test  of  anxiety.  Pharmacol 
Biochem  Behav  30:701-706,  1988. 

Littleton,  J.  Acamprosate  in  alcohol  de- 
pendence: How  does  it  work?  Addiction 
90:1179-1188,  1995. 

Ludwig,  A.M.,  and  Stark,  L.H.  Alcohol 
craving.  Subjective  and  situational  aspects. 
QJ  Stud  Alcohol  35:899-905,  1974. 

Ludwig,  A.M.,  and  Wilder,  A.  "Craving" 
and  relapse  to  drink.  QJ  Stud  Alcohol 
35:108-130,  1974. 

Ludwig,  A.M.;  Wilder,  A.;  and  Stark, 
L.H.  The  first  drink:  Psychobiological 
aspects  of  craving.  Arch  Gen  Psychiatry 
30:539-547,  1974. 

Macey,  D.J.;  Schulteis,  G.;  Heinrichs, 
S.C.;  and  Koob,  G.F.  Time-dependent 
quantifiable  withdrawal  from  ethanol  in 
the  rat:  Effect  of  method  of  dependence 
induction.  Alcohol  13:163-170,  1996. 

Madamba,  S.G.;  Schweitzer,  P.; 
Zieglgansberger,  W.;  and  Siggins,  G.R. 
Acamprosate  (calcium  acetylhomotaurinate) 
enhances  the  N-methyl-D-aspartate  com- 
ponent of  excitatory  neurotransmission  in 
rat  hippocampal  CA1  neurons  in  vitro. 
Alcohol  Clin  Exp  Res  20:651-658,  1996. 

Majchrowicz,  E.  Induction  of  physical 
dependence  upon  ethanol  and  the  associ- 


502 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


ated  behavioral  changes  in  rats.  Psycho- 
pharmacologia  43:245-254,  1975. 

Majewska,  M.D.,  and  Schwartz,  R.D. 
Pregnenolone  sulfate:  An  endogenous 
antagonist  of  the  gamma-aminobutyric 
acid  receptor  complex  in  brain?  Brain  Res 
404:355-360,  1987. 

Majewska,  M.D.;  Harrison,  N.L.; 
Schwartz,  R.D.;  Barker,  J.L.;  and  Paul, 
S.M.  Steroid  hormone  metabolites  are 
barbiturate-like  modulators  of  the  GABA 
receptor.  Science  232:1004-1007,  1986. 

Majewska,  M.D.;  Demirgoren,  S.;  Spivak, 
C.E.;  and  London,  E.D.  The  neurosteroid 
dehydroepiandrosterone  sulfate  is  an 
allosteric  antagonist  of  the  GABAA  receptor. 
Brain  Res  526:143-146,  1990. 

Make,  T.S.;  Make,  E.A.;  and  Dongkr, 
M.  Efficacy  of  buspirone  in  alcohol  de- 
pendence: A  review.  Alcohol  Clin  Exp  Res 
20:853-858,  1996. 

Manley,  S.J.,  and  Little,  H.J.  Enhancement 
of  amphetamine-  and  cocaine-induced 
locomotor  activity  after  chronic  ethanol 
administration.  J  Pharmacol  Exp  Ther 
281:1330-1339,1997. 

Markou,  A.,  and  Koob,  G.F.  Post  cocaine 
anhedonia:  An  animal  model  of  cocaine 
withdrawal.  Neuropsychopharmacology 
4:17-26,  1991. 

Marlatt,  GA.  Relapse  prevention: 
Introduction  and  overview  of  the  model. 
In:  Marlatt,  G.A.,  and  Gordon,  J.R,  eds. 

Relapse  Prevention:  Maintenance  Strategies 
in  the  Treatment  of  Addictive  Behaviors. 
London:  Guilford,  1985.  pp.  3-70. 

McBride,  W.J.;  Murphy,  J.M.;  Lumeng,  L.; 
and  Li,  T.-K  Effects  of  Ro-15-4513,  fluox- 
etine and  desipramine  on  the  intake  of 
ethanol,  water  and  food  by  the  alcohol- 
preferring  (P)  and  non-preferring  (NP)  lines 


of  rats.  Pharmacol  Biochem  Behav  30: 
1045-1050,  1988. 

McBride,  W.J.;  Guan,  X.M.;  Chernet,  E.; 
Lumeng,  L.;  and  Li,  T.-K.  Regional  dif- 
ferences in  the  densities  of  serotonin- 1 A 
receptors  between  P  and  NP  rats.  Alcohol 
Clin  Exp  Res  14:316,  1990a. 

McBride,  W.J.;  Murphy,  J.M.;  Lumeng, 
L.;  and  Li,  T.-K.  Serotonin,  dopamine, 
and  GABA  involvement  in  alcohol  drink- 
ing of  selectively  bred  rats.  Alcohol  7:199- 
205,  1990&. 

McBride,  W.J.;  Murphy,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Serotonin  and  alcohol 
consumption.  In:  Naranjo,  C.A.,  and 
Sellers,  E.M.,  eds.  Novel  Pharmacological 
Interventions  for  Alcoholism.  New  York: 
Springer,  1992. 

McCown,  T.J.,  and  Breese,  G.R.  Multiple 
withdrawals  from  chronic  ethanol  "kin- 
dles" inferior  collicular  seizure  activity: 
Evidence  for  kindling  seizures  associated 
with  alcoholism.  Alcohol  Clin  Exp  Res 
14:394-399,  1990. 

McKay,  J.R;  Rutherford,  M.J.;  Alterman, 
A.I.;  Cacciola,  J.S.;  and  Kaplan,  M.R.  An 
examination  of  the  cocaine  relapse  process. 
Drug  Alcohol  Depend  38:35-43,  1995. 

Meert,  T.F.  Pharmacological  evaluation  of 
alcohol  withdrawal-induced  inhibition  of 
exploratory  behaviour  and  supersensitivity 
to  harmine-induced  tremor.  Alcohol  Alcohol 
29:91-102,  1994. 

Meert,  T.F.,  and  Huysmans,  H.  Repeated 
characterization  of  alcohol  withdrawal 
reactions  in  rats  chronically  exposed  to  an 
alcohol  liquid  diet.  Prog  Neuropsychopharm- 
acol  Biol  Psychiatry  18:947-960,  1994. 

Meisch,  RA.  Alcohol  self- administration 
by  experimental  animals.  In:  Smart,  R.G.; 
Cappell,  H.D.;  Glaser,  F.B.;  Israel,  Y.; 
Kalant,  H.;  Popham,  R.E.;  Schmidt,  W.; 


303 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  Sellers,  E.M.,  eds.  Research  Advances 
in  Alcohol  and  Drug  Problems.  New  York: 
Plenum  Press,  1984.  pp.  23-45. 

Meisch,  R.A.,  and  Stewart,  R.B.  Ethanol 
as  a  reinforcer:  A  review  of  laboratory 
studies  of  nonhuman  primates.  Behav 
Pharmacol  5:425-440,  1994. 

Melchior,  C.L.,  and  Ritzmann,  R.F. 
Dehydroepiandrosterone  enhances  the 
hypnotic  and  hypothermic  effects  of 
ethanol  and  pentobarbital.  Pharmacol 
Biochem  Behav  43:223-227,  1992. 

Melchior,  C.L.,  and  Ritzmann,  R.F. 
Dehydroepiandrosterone  is  an  anxiolytic 
in  mice  on  the  plus  maze.  Pharmacol 
Biochem  Behav  47:437-441,  1994a. 

Melchior,  C.L.,  and  Ritzmann,  R.F. 
Pregnenolone  and  pregnenolone  sulfate, 
alone  and  with  ethanol,  in  mice  on  the 
plus  maze.  Pharmacol  Biochem  Behav 
48:893-897,  19946. 

Mendelson,  J.H.,  and  Mello,  N.K. 
Experimental  analysis  of  drinking  behav- 
ior of  chronic  alcoholics.  Ann  NT  Acad 
Sci  133:828-845,  1966. 

Merlo  Pich,  E.;  Lorang,  M.T.;  Yeganeh, 
M.;  Rodriguez  De  Fonseca,  F.;  Raber,  J.; 
Koob,  G.F.;  and  Weiss,  F.  Increase  of 
extracellular  corticotropin-releasing  fac- 
tor-like immunoreactivity  levels  in  the 
amygdala  of  awake  rats  during  restraint 
stress  and  ethanol  withdrawal  as  measured 
by  microdialysis.  J  Neurosci  15:5439- 
5447,  1995. 

Modell,  J.G.;  Mountz,  J.M.;  Glaser,  F.B.; 
and  Lee,  J.Y.  Effect  of  haloperidol  on 
measures  of  craving  and  impaired  control 
in  alcoholic  subjects.  Alcohol  Clin  Exp  Res 
17:234-240,  1993. 

Mollenauer,  S.;  Bryson,  R;  Robinson, 
M.;  Sardo,  J.;  and  Coleman,  C.  EtOH 
self-administration  in  anticipation  of  noise 


stress  in  C57BL/6J  mice.  Pharmacol 
Biochem  Behav  46:35-38,  1993. 

Monti,  J.M.,  and  Alterwain,  P.  Ritanserin 
decreases  alcohol  intake  in  chronic  alco- 
holics. Lancet  337:60,  1991. 

Monti,  P.M.;  Rohsenow,  D.J.;  Rubonis,  A.V.; 
Niaura,  R.S.;  Sirota,  A.D.;  Colby,  S.M.; 
and  Abrams,  D.B.  Alcohol  cue  reactivity: 
Effects  of  detoxification  and  extended 
exposure.  J  Stud  Alcohol  54:235-245, 1993. 

Moolten,  M.,  and  Kornetsky,  C.  Oral  self- 
administration  of  ethanol  and  not  experi- 
menter-administered ethanol  facilitates 
rewarding  electrical  brain  stimulation. 
Alcohol  7:221-225,  1990. 

Morrisett,  RA.;  Rezvani,  A.H.;  Overstreet, 
D.;  Janowsky,  D.S.;  Wilson,  W.A.;  and 
Swartzwelder,  H.S.  MK-801  potently 
inhibits  alcohol  withdrawal  seizures  in 
rats.  Eur  J  Pharmacol  176:103-105,  1990. 

Morrow,  A.L.;  Suzdak,  P.D.;  and  Paul, 
S.M.  Steroid  hormone  metabolites  poten- 
tiate GABA  receptor-mediated  chloride, 
ion  flux  with  nanomolar  potency.  Eur  J 
Pharmacol  142:483-485,  1987. 

Morrow,  A.L.;  Suzdak,  P.D.;  Karanian, 
J.W.;  and  Paul,  S.M.  Chronic  ethanol 
administration  alters  Y-aminobutyric  acid, 
pentobarbital  and  ethanol-mediated  36C1~ 
uptake  in  cerebral  cortical  synaptoneuro- 
somes.  J  Pharmacol  Exp  Ther  246:158- 
164, 1988. 

Mossberg,  D.;  Liljeberg,  P.;  and  Borg,  S. 
Clinical  conditions  in  alcoholics  during 
long-term  abstinence:  A  descriptive,  lon- 
gitudinal treatment  study.  Alcohol 2:551- 
553,1985. 

Moy,  S.S.;  Knapp,  D.J.;  Criswell,  H.E.; 
and  Breese,  G.R.  Flumazenil  blockade  of 
anxiety  following  ethanol  withdrawal  in 
rats.  Psychopharmacology  (Berl)  131:354- 
360,  1997. 


304 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Muller,  N.;  Hoehe,  M.;  Klein,  H.E.; 
Nieberle,  G.;  Kapfhammer,  H.P.;  May, 
F.;  Muller,  O.A.;  and  Fichter,  M.  Endo- 
crinological studies  in  alcoholics  during 
withdrawal  and  after  abstinence.  Psycho- 
neuroendocrinology  14:113-123,  1989. 

Murphy,  J.M.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Regional  brain  levels  of 
monoamines  in  alcohol-preferring  and 
non-preferring  lines  of  rats.  Pharmacol 
Biochem  Behav  16:145-149,  1982. 

Murphy,  J.M.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Contents  of  monoamines 
in  forebrain  regions  of  alcohol-preferring 
(P)  and  non-preferring  (NP)  lines  of  rats. 
Pharmacol  Biochem  Behav  26:389-392, 1987. 

Myers,  R.D.;  Stoltman,  W.P.;  and  Martin, 
G.E.  Effects  of  ethanol  dependence  induced 
artificially  in  the  rhesus  monkey  on  the 
subsequent  preference  for  ethyl  alcohol. 
Physiol  Behav  9:43^8,  1972. 

Naranjo,  C.A.,  and  Bremner,  K.E.  Clinical 
pharmacology  of  serotonin-altering  medica- 
tions for  decreasing  alcohol  consumption. 
Alcohol  Alcohol  Supp I 2:221-229,  1993. 

Naranjo,  C.A.;  Sellers,  E.M.;  Sullivan,  J.T.; 
Woodley,  D.V.;  Kadlec,  K.;  and  Sykora, 
K.  The  serotonin  inhibitor  citalopram 
attenuates  ethanol  intake.  Clin  Pharmacol 
Ther  41:266-274, 1987. 

Naranjo,  C;  Kadlec,  K.;  Sanhueza,  P.; 
Woodley-Remus,  D.;  and  Sellers,  E.M. 
Fluoxetine  differentially  alters  alcohol 
intake  and  other  consummatory  behaviors 
in  problem  drinkers.  Clin  Pharmacol  Ther 
47:490-498,  1990. 

Naranjo,  C.A.;  Poulos,  C.X.;  Lanctot, 
K.L;  Bremner,  K.E.;  Kwok,  M.;  and 
Umana,  M.  Ritanserin,  a  central  5-HT2 
antagonist,  in  heavy  social  drinkers: 
Desire  to  drink,  alcohol  intake  and  related 
effects.  Addiction 90:893-905,  1995. 


Nash,  J.,  Jr.,  and  Maickel,  R.P.  The  role 
of  the  hypothalamic-pituitary-adrenocortical 
axis  in  post-  stress  induced  ethanol  consump- 
tion by  rats.  Prog  Neuropsychopharmacol 
Biol  Psychiatry  12:653-671,  1988. 

Nestby,  P.;  Vanderschuren,  L.J.;  De  Vries, 
T.J.;  Hogenboom,  F.;  Wardeh,  G.;  Mulder, 
A.H.;  and  Schoffelmeer,  A.N.  Ethanol, 
like  psychostimulants  and  morphine,  causes 
long-lasting  hyperreactivity  of  dopamine 
and  acetylcholine  neurons  of  rat  nucleus 
accumbens:  Possible  role  in  behavioural 
sensitization.  Psychopharmacology  (Berl) 
133:69-76,  1997. 

Ng,  G.Y.,  and  George,  S.R.  Dopamine 
receptor  agonist  reduces  ethanol  self- 
administration  in  the  ethanol-preferring 
C57BL/6J  inbred  mouse.  Eur  J 
Pharmacol  269:365-374,  1994. 

N'Gouemo,  P.;  Caspary,  D.M.;  and 
Faingold,  C.L.  Decreased  GABA  effec- 
tiveness in  the  inferior  colliculus  neurons 
during  ethanol  withdrawal  in  rats  suscep- 
tible to  audiogenic  seizures.  Brain  Res 
724:200-204,  1996. 

O'Brien,  C.P.  Drug  addiction  and  drug 
abuse.  In:  Hardman,  J.G.;  Limbird,  L.E.; 
Molinoff,  P.B.;  Ruddon,  RW.;  and  Gilman, 
A.G.,  eds.  Goodman  and  Gilman's  The 
Pharmacological  Basis  of  Therapeutics.  9th 
ed.  New  York:  McGraw-Hill,  1996.  pp. 
557-577. 

O'Brien,  C.P.;  Volpicelli,  LA.;  and 
Volpicelli,  J.R  Naltrexone  in  the  treatment 
of  alcoholism:  A  clinical  review.  Alcohol 
13:35-39,  1996. 

O'Donnell,  P.J.  The  abstinence  violation 
effect  and  circumstances  surrounding 
relapse  as  a  predictor  of  outcome  status  in 
male  alcoholic  outpatients.  J  Psychol 
117:257-262,  1984. 


305 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


O'Malley,  S.S.;  JafFe,  A.J.;  Chang,  G.; 
Schottenfeld,  R.S.;  Meyer,  R.E.;  and 
Rounsaville,  B.  Naltrexone  and  coping 
skills  therapy  for  alcohol  dependence:  A 
controlled  study.  Arch  Gen  Psychiatry 
49:881-887,  1992. 

Oslin,  D.;  Liberto,  J.G.;  O'Brien,  J.; 
Krois,  S.;  and  Norbeck,  J.  Naltrexone  as 
an  adjunctive  treatment  for  older  patients 
with  alcohol  dependence.  Am  J  Geriatr 
Psychiatry  5:  324-332,  1997. 

Paille,  F.M.;  Guelfi,  J.D.;  Perkins,  A.C.; 
Royer,  R.J.;  Steru,  L.;  and  Parot,  P. 
Double-blind  randomized  multicentre 
trial  of  acamprosate  in  maintaining  absti- 
nence from  alcohol.  Alcohol  Alcohol 
30:239-247,  1995. 

Panocka,  I.;  Ciccocioppo,  R;  Mosca,  M.; 
Palidori,  C;  and  Massi,  M.  Effects  of  the 
dopamine  Dx  receptor  antagonist  SCH 
39166  on  the  ingestive  behaviour  of  alco- 
hol-preferring rats.  Psychopharmacolqgy 
120:227-235,  1995. 

Parsons,  L.H.;  Koob,  G.F.;  and  Weiss,  F. 
Serotonin  dysfunction  in  the  nucleus 
accumbens  of  rats  during  withdrawal  after 
unlimited  access  to  intravenous  cocaine.  J 
Pharmacol  Exp  Ther  274:1182-1191, 1995. 

Pecins-Thompson,  M.,  and  Peris,  J. 
Behavioral  and  neurochemical  changes 
caused  by  repeated  ethanol  and  cocaine 
administration.  Psychopharmacology  110: 
443^50, 1993. 

Pelc,  I.;  Verbanck,  P.;  Le  Bon,  O.; 
Gavrilovic,  M.;  Lion,  K.;  and  Lehert,  P. 
Efficacy  and  safety  of  acamprosate  in  the 
treatment  of  detoxified  alcohol-dependent 
patients.  A  90-day  placebo-controlled 
dose-finding  study.  Br  J Psychiatry  171: 
73-77,  1997. 

Pellow,  S.,  and  File,  S.E.  Anxiolytic  and 
anxiogenic  drug  effects  on  exploratory 


activity  in  an  elevated  plus-maze:  A  novel 
test  of  anxiety  in  the  rat.  Pharmacol 
Biochem  Behav  24:525-529,  1986. 

Peter,  K.;  Jauch,  M.;  and  Bauer,  U.  [Study 
of  psychopathologic-biorhythm  aspects  of 
alcoholic  patients  in  the  withdrawal  phase — 
initial  results  of  a  pilot  study].  Wien  Med 
Wochenschr  145:454-455,  1995. 

Phillips,  T.J.;  Dickinson,  S.;  and  Burkhart- 
Kasch,  S.  Behavioral  sensitization  to  drug 
stimulant  effects  in  C57BL/6J  and  DBA/ 
2 J  inbred  mice.  Behav  Neurosci  108:789- 
803, 1994. 

Phillips,  T.J.;  Huson,  M.;  Gwiazdon,  C; 
Burkhart-Kasch,  S.;  and  Shen,  E.H. 
Effects  of  acute  and  repeated  ethanol 
exposures  on  the  locomotor  activity  of 
BXD  recombinant  inbred  mice.  Alcohol 
Clin  Exp  Res  19:269-278,  1995. 

Phillips,  T.J.;  Lessov,  C.N.;  Harland,  R.D.; 
and  Mitchell,  S.R  Evaluation  of  potential 
genetic  associations  between  ethanol 
tolerance  and  sensitization  in  BXD/Ty 
recombinant  inbred  mice.  / Pharmacol 
Exp  Ther  277 r:6 13-623,  1996. 

Phillips,  T.J.;  Roberts,  A.J.;  and  Lessov, 
C.N.  Behavioral  sensitization  to  ethanol: 
Genetics  and  the  effects  of  stress.  Pharmacol 
Biochem  Behav  57:487-493,  1997. 

Piazza,  P.V.,  and  Le  Moal,  M.  Gluco- 
corticoids as  a  biological  substrate  of 
reward:  Physiological  and  pathophysio- 
logical implications.  Brain  Res  Rev  25: 
359-372,  1997. 

Piazza,  P.V.,  and  Le  Moal,  M.  Patho- 
physiological basis  of  vulnerability  to  drug 
abuse:  Role  of  an  interaction  between 
stress,  glucocorticoids,  and  dopaminergic 
neurons.  Annu  Rev  Pharmacol  Toxicol 
36:359-378,  1996. 

Pich,  E.M.;  Pagliusi,  S.R;  Tessari,  M.; 
Talabot-Ayer,  D.;  Hooftvan  Huijsduijnen, 


306 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


R;  and  Chiamulera,  C.  Common  neural 
substrates  for  the  addictive  properties  of 
nicotine  and  cocaine.  Science  275:83-86, 
1997. 

Prather,  P.L.;  Rezazadeh,  S.M.;  and  Lai, 
H.  Mianserin  in  the  treatment  of  ethanol 
withdrawal  in  the  rat:  Prevention  of  be- 
haviors indicative  of  anxiety.  Psychopharmacol 
Bull  27 -.285-289,  1991. 

Prunell,  M.;  Escorihuela,  KM.;  Fernandez- 
Teruel,  A.;  Nunez,  J.F.;  and  Tobena,  A. 
Differential  interactions  between  ethanol 
and  Ro  15-4513  on  two  anxiety  tests  in 
rats.  Pharmacol  Biochem  Behav  47:147- 
151,  1994. 

Putzke,  J.;  Spanagel,  R.;  Tolle,  T.R.;  and 
Zieglgansberger,  W.  The  anti-craving 
drug  acamprosate  reduces  c-fos  expression 
in  rats  undergoing  ethanol  withdrawal. 
Eur  J  Pharmacol  317:39-48,  1996. 

Ramsey,  N.  R,  and  Van  Ree,  J.  M.  Emotional 
but  not  physical  stress  enhances  intravenous 
cocaine  self- administration  in  drug  naive 
rats.  Brain  Res  608:216-222,  1993. 

Rassnick,  S.;  Koob,  G.F.;  and  Geyer,  MA. 
Responding  to  acoustic  startle  during  chronic 
ethanol  intoxication  and  withdrawal. 
Psychopharmacology  106:351-358,  1992#. 

Rassnick,  S.;  Pulvirenti,  L.;  and  Koob,  G.F. 
Oral  ethanol  self- administration  in  rats  is 
reduced  by  the  administration  of  dopamine 
and  glutamate  receptor  antagonists  into 
the  nucleus  accumbens.  Psycho-pharmacology 
109:92-98,  1992£. 

Rassnick,  S.;  D'Amico,  E.;  Riley,  E.;  and 
Koob,  G.F.  GAB  A  antagonist  and  benzo- 
diazepine partial  inverse  agonist  reduce 
motivated  responding  for  ethanol.  Alcohol 
Clin  Exp  Res  17:124-130,  1993^. 

Rassnick,  S.;  Heinrichs,  S.C.;  Britton, 
K.T.;  and  Koob,  G.F.  Microinjection  of  a 
corticotropin-releasing  factor  antagonist 


into  the  central  nucleus  of  the  amygdala 
reverses  anxiogenic-like  effects  of  ethanol 
withdrawal.  Brain  2Ur  605:25-32,  1993&. 

Redei,  E.;  Branch,  B.J.;  Gholami,  S.;  Lin, 
E.Y.R;  and  Taylor,  A.N.  Effects  of  ethanol 
on  CRF  release  in  vitro.  Endocrinology 
123:2736-2743,  1988. 

Rezazadeh,  S.M.;  Prather,  P.L.;  and  Lai, 
H.  Sensitization  to  5-HT1C  receptor 
agonist  in  rats  observed  following  with- 
drawal from  chronic  ethanol.  Alcohol 
10:281-283,  1993. 

Richter,  R.M.;  Merlo  Pich,  E.;  Koob, 
G.F.;  and  Weiss,  F.  Sensitization  of  co- 
caine-stimulated increase  in  extracellular 
levels  of  corticotropin-releasing  factor 
from  the  rat  amygdala  after  repeated 
administration  as  determined  by  microdial- 
ysis.  Neurosci Lett  187 -.169-172,  1995. 

Riihioja,  P.;  Jaatinen,  P.;  Oksanen,  PL; 
Haapalinna,  A.;  Heinonen,  E.;  and 
Hervonen,  A.  Dexmedetomidine,  diazepam, 
and  propranolol  in  the  treatment  of 
ethanol  withdrawal  symptoms  in  the  rat. 
Alcohol  Clin  Exp  Res  21:804-808,  1997. 

Risher-Flowers,  D.;  Adinoff,  B.;  Ravitz, 
B.;  Bone,  G.H.;  Martin,  P.R;  Nutt,  D.; 
and  Linnoila,  M.  Orcadian  rhythms  of 
Cortisol  during  alcohol  withdrawal.  Adv 
Alcohol  Subst  Abuse  7:37-41,  1988. 

Ritz,  M.C.;  George,  F.R.;  and  Meisch, 
RA.  Ethanol  self- administration  in  ALKO 
rats:  II  Effects  of  selection  and  fixed-ratio 
size.  Alcohol  6:235-239, 1989. 

Ritz,  M.C.;  Garcia,  J.M.;  Protz,  D.;  Rael, 
A.-M.;  and  George,  F.R  Ethanol-reinforced 
behavior  in  P,  NP,  HAD  and  LAD  rats: 
Differential  genetic  regulation  of  reinforce- 
ment and  motivation.  Behav  Pharmacol 
5:521-531,  1994. 

Rivier,  C;  Bruhn,  T.;  and  Vale,  W.  Effect 
of  ethanol  on  the  hypothalamic-pituitary- 


307 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


adrenal  axis  in  the  rat:  Role  of  corticotropin  - 
releasing  factor  (CRF).  J  Pharmacol  Exp 
7fc*r  229:127-131,  1984. 

Roberts,  A.J.;  Lessov,  C.N.;  and  Phillips, 
TJ.  Critical  role  for  glucocorticoid  recep- 
tors in  stress-  and  ethanol-induced  loco- 
motor sensitization.  J  Pharmacol  Exp  Ther 
275:790-797,  1995. 

Roberts,  A.J.;  Cole,  M.;  and  Koob,  G.F. 
Intra- amygdala  muscimol  decreases  oper- 
ant ethanol  self-administration  in  depen- 
dent rats.  Alcohol  Clin  Exp  Res 20:1289- 
1298,  1996. 

Robinson,  T.E.,  and  Becker,  J.B.  Enduring 
changes  in  brain  and  behavior  produced 
by  chronic  amphetamine  administration: 
A  review  and  evaluation  of  animal  models 
of  amphetamine  psychosis.  Brain  Res  Rev 
11:157-198,1986. 

Robinson,  T.E.,  and  Berridge,  K.C.  The 
neural  basis  of  drug  craving:  An  incentive  - 
sensitization  theory  of  addiction.  Brain 
Res  Rev  18:247-291,  1993. 

Roelofs,  S.M.  Hyperventilation,  anxiety, 
craving  for  alcohol:  A  subcute  alcohol  with- 
drawal syndrome.  Alcohol 2:5Ql-5Q5,  1985. 

Rohsenow,  D.J.;  Monti,  P.M.;  Rubonis, 
A. V.;  Sirota,  A.D.;  Niaura,  R.S.;  Colby,  S. 
M.;  Wunschel,  S.M.;  and  Abrams,  D.B. 
Cue  reactivity  as  a  predictor  of  drinking 
among  male  alcoholics.  J  Consult  Clin 
Psychol  62:620-626,  1994. 

Romeo,  E.;  Brancati,  A.;  De  Lorenzo,  A.; 
Fucci,  P.;  Furnari,  C;  Pompili,  E.;  Sasso, 
G.F.;  Spalletta,  G.;  Troisi,  A.;  and  Pasini, 
A.  Marked  decrease  of  plasma  neuroactive 
steroids  during  alcohol  withdrawal.  Clin 
Neuropharmacol  19:366-369,  1996. 

Rosenblatt,  S.M.;  Gross,  M.M;  Malenowski, 
B.;  Browman,  M.;  and  Lewis,  E..  Factor 
analysis  of  the  daily  clinical  course  rating 


scale  of  the  acute  alcoholic  psychosis.  QJ 
Stud  Alcohol  33:1060-1064,  1972. 

Roske,  I.;  Baeger,  I.;  Frenzel,  R;  and 
Oehme,  P.  Does  a  relationship  exist  be- 
tween the  quality  of  stress  and  the  moti- 
vation to  ingest  alcohol?  Alcohol  1 1 : 
113-124,  1994. 

Rossetti,  Z.L.;  Hmaidan,  Y.;  and  Gessa, 
G.L.  Marked  inhibition  of  mesolimbic 
dopamine  release:  A  common  feature  of 
ethanol,  morphine,  cocaine  and  ampheta- 
mine abstinence  in  rats.  Eur ]  Pharmacol 
221:227-234,  1992a. 

Rossetti,  Z.L.;  Melis,  F.;  Carboni,  S.; 
Diana,  M.;  and  Gessa,  G.L.  Alcohol  with- 
drawal in  rats  is  associated  with  a  marked 
fall  in  extraneuronal  dopamine.  Alcohol 
Clin  Exp  Res  16:529-532,  \992b. 

Rossetti,  Z.L.;  Hmaidan,  Y.;  Diana,  M.; 
and  Gessa,  G.L.  Lack  of  tolerance  to 
ethanol-induced  dopamine  release  in  the 
rat  ventral  striatum.  Eur  J  Pharmacol 
231:203-207,  1993. 

Rouhani,  S.;  Emmanouilidis,  E.;  Tran,  G.; 
Durlach,  J.;  Payan,  C;  Fermanian,  J.; 
Manicom,  R.;  Soulairac,  A.;  and  Poenaru, 
S.  Orcadian  variations  in  vigilance  states 
in  the  alcohol-dependent  rat.  Physiol 
Behav  48:637-640,  1990. 

Salimov,  R.M.,  and  Salimova,  N.B.  The 
alcohol  deprivation  effect  in  hybrid  mice. 
Dru£  Alcohol  Depend  32:187-191,  1993. 

Samson,  H.H.  Initiation  of  ethanol  rein- 
forcement using  a  sucrose -substitution 
procedure  in  food-  and  water-sated  rats. 
Alcohol  Clin  Exp  Res  10:436-442,  1986. 

Samson,  H.H.  Initiation  of  ethanol-main- 
tained  behavior:  A  comparison  of  animal 
models  and  their  implication  to  human 
drinking.  In:  Thompson,  T.;  Dews,  P.; 
and  Barrett,  J.,  eds.  Advances  in  Behavioral 
Pharmacology,  Vol.  6:  Neurobehavioral 


308 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Pharmacology.  Hillsdale,  NJ:  Erlbaum 
Associates,  1987.  pp.  221-248. 

Samson,  H.H.  Environmental  effects  on 
drinking  patterns:  Stress  and  sensitization. 
In:  Hunt,  W.A.,  and  Zakhari,  S.,  eds. 
Stress,  Gender,  and  Alcohol-Seeking  Behavior. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  29. 
NIH  Pub.  No.  95-3893.  Bethesda,  MD: 
the  Institute,  1995.  pp.  277-291. 

Samson,  H.H.,  and  Falk,  J.L.  Alteration 
of  fluid  preference  in  ethanol-dependent 
animals.  J  Pharmacol  Exp  Ther  190:365- 
376,  1974. 

Samson,  H.H.,  and  Harris,  R.A. 
Neurobiology  of  alcohol  abuse.  Trends 
Pharmacol  Set  13:206-211,  1992. 

Samson,  H.H.;  Haraguchi,  M.;  Tolliver, 
G.A.;  and  Sadegi,  K.G.  Antagonism  of 
ethanol-reinforced  behavior  by  the  ben- 
zodiazepine inverse  agonists  Rol5-4513 
and  FG7142:  Relation  to  sucrose  rein- 
forcement. Pharmacol  Biochem  Behav 
33:601-608,  1989. 

Samson,  H.H.;  Tolliver,  G.A.;  Haraguchi, 
M.;  and  Hodge,  C.W.  Alcohol  self- admin- 
istration: Role  of  mesolimbic  dopamine. 
Ann  NT  Acad  Sci  654:242-253,  1992. 

Samson,  H.H.;  Hodge,  C.W.;  Tolliver, 
G.A.;  and  Haraguchi,  M.  Effect  of 
dopamine  agonists  and  antagonists  on 
ethanol  reinforced  behavior:  The  involve- 
ment of  the  nucleus  accumbens.  Brain 
Res  Bull  30:133-141,  1993. 

Sass,  H.;  Soyka,  M.;  Mann,  K.;  and 
Zieglgansberger,  W.  Relapse  prevention 
by  acamprosate.  Results  from  a  placebo- 
controlled  study  on  alcohol  dependence. 
Arch  Gen  Psychiatry  53:673-680,  1996. 

Schenk,  S.;  Gorman,  K.;  and  Amit,  Z. 
Age-dependent  effects  of  isolation  hous- 


ing on  the  self- administration  of  ethanol 
in  laboratory  rats.  Alcohol  7:321-326, 1990. 

Schmitz,  M.M.;  Sepandj,  A.;  Pichler, 
P.M.;  and  Rudas,  S.  Disrupted  melatonin- 
secretion  during  alcohol  withdrawal.  Prog 
Neuropsychopharmacol  Biol  Psychiatry 
20:983-995,  1996. 

Schulteis,  G.;  Markou,  A.;  Cole,  M.;  and 
Koob,  G.F.  Decreased  brain  reward  pro- 
duced by  ethanol  withdrawal.  Proc  Natl 
Acad  Sci  USA  92:5880-5884,  1995. 

Schulteis,  G.;  Hyytia,  P.;  Heinrichs,  S.C.; 
and  Koob,  G.F.  Effects  of  chronic  ethanol 
exposure  on  oral  self- administration  of 
ethanol  or  saccharin  by  Wistar  rats. 
Alcohol  Clin  Exp  Res  20:164-171,  1996. 

Schultz,  W.;  Dayan,  P.;  and  Montague, 
P.R.  A  neural  substrate  of  prediction  and 
reward.  Science  275:1593-1599,  1997. 

Self,  D.W.;  Barnhart,  W.J.;  Lehrman, 
D.A.;  and  Nestier,  E.J.  Opposite  modula- 
tion of  cocaine -seeking  behavior  by  Dl- 
and  D2-like  dopamine  receptor  agonists. 
Science  271:1586-1589,  1996. 

Sellers,  E.M.;  Higgins,  G.A.;  and  Sobell, 
M.B.  5-HT  and  alcohol  abuse.  Trends 
Pharmacol  Sci  13:69-75,  1992. 

Shaham,  Y.  Immobilization  stress-induced 
oral  opioid  self- administration  and  with- 
drawal in  rats:  Role  of  conditioning  factors 
and  the  effect  of  stress  on  "relapse"  to 
opioid  drugs.  Psychopharmacology  111: 
477-485,  1993. 

Shaham,  Y.,  and  Stewart,  J.  Exposure  to 
mild  stress  enhances  the  reinforcing  effi- 
cacy of  intravenous  heroin  self- administra- 
tion in  rats.  Psychopharmacolqgy  111:523- 
527,  1994. 

Shaham,  Y.,  and  Stewart,  J.  Stress  reinstates 
heroin-seeking  in  drug-free  animals:  An 


309 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


effect  mimicking  heroin,  not  withdrawal. 
Psychopharmacology  119:334-341,  1995. 

Shen,  R.  Y.,  and  Chiodo,  L.A.  Acute 
withdrawal  after  repeated  ethanol  treatment 
reduces  the  number  of  spontaneously  active 
dopaminergic  neurons  in  the  ventral  tegmen- 
tal area.  Brain  Res  622:289-293,  1993. 

Signs,  S.A.,  and  Schechter,  M.D.  The  role 
of  dopamine  and  serotonin  receptors  in  the 
mediation  of  the  ethanol  interoceptive  cue. 
Pharmacol  Biochem  Bebav  30:55-64,  1988. 

Sinclair,  J.D.  The  alcohol-deprivation 
effect:  Influence  of  various  factors.  QJ 
Stud  Alcohol  33:769-782,  1972. 

Sinclair,  J.D.  Alcohol -deprivation  effect  in 
rats  genetically  selected  for  their  ethanol 
preference.  Pharmacol  Biochem  Behav  10: 
597-602,  1979. 

Sinclair,  J.D.,  and  Li,  T.-K.  Long  and 
short  alcohol  deprivation:  Effects  on  AA 
and  P  alcohol-preferring  rats.  Alcohol 
6:505-509,  1989. 

Sinclair,  J.D.,  and  Senter,  RJ.  Increased 
preference  for  ethanol  in  rats  following 
alcohol  deprivation.  Psychonomic  Sci  8:11- 
12,  1967. 

Sinclair,  J.D.,  and  Senter,  RJ.  Development 
of  an  alcohol  deprivation  effect  in  rats.  Q 
J  Stud  Alcohol  29:863-867,  1968. 

Sinclair,  J.D.,  and  Tiihonen,  K.  Lack  of 
alcohol-deprivation  effect  in  AA  rats.  Alcohol 
5:85-87,  1988. 

Sinclair,  J.D.;  Le,  A.D.;  and  Kiianmaa,  K. 
The  AA  and  ANA  rat  lines,  selected  for 
differences  in  voluntary  alcohol  consump- 
tion. Experientia  45:798-805,  1989. 

Snell,  L.D.;  Szabo,  G.;  Tabakoff,  B.;  and 
Hoffman,  P.L.  Gangliosides  reduce  the 
development  of  ethanol  dependence 
without  affecting  ethanol  tolerance.  / 
Pharmacol  Exp  Ther  279:128-136,  1996. 


Soyka,  M.,  and  Sass,  H.  Acamprosate:  A  new 
pharmacotherapeutic  approach  to  relapse 
prevention  in  alcoholism-preliminary  data. 
Alcohol  Alcohol  Suppl  2:531-536,  1994. 

Spanagel,  R.,  and  Zieglgansberger,  W. 
Anti-craving  compounds  for  ethanol: 
New  pharmacological  tools  to  study  ad- 
dictive processes.  Trends  Pharmacol  Sci 
18:54-59,  1997. 

Spanagel,  R;  Holter,  S.M.;  Allingham,  K.; 
Landgraf,  R;  and  Zieglgansberger,  W.  Acam- 
prosate and  alcohol:  I.  Effects  on  alcohol 
intake  following  alcohol  deprivation  in 
the  rat.  Eur  J  Pharmacol  305:39^4,  1996. 

Staiger,  P.  K.,  and  White,  J.M.  Cue  reactiv- 
ity in  alcohol  abusers:  Stimulus  specificity 
and  extinction  of  the  responses.  Addict 
Behav  16:211-221,  1991. 

Stewart,  J.,  and  deWit,  H.  Reinstatement 
of  drug- taking  behavior  as  a  method  of 
assessing  incentive  motivational  properties 
of  drugs.  In:  Bozarth,  M.A.,  ed.  Methods 
of  Assessing  the  Reinforcing  Properties  of 
Abused  Drugs.  New  York:  Springer-Verlag, 
1987.  pp.  211-227. 

Stewart,  R.B.;  Gatto,  G.J.;  Lumeng,  L.; 
Li,  T.-K.;  and  Murphy,  J.M.  Comparison 
of  alcohol-preferring  (P)  and  nonprefer- 
ring  (NP)  rats  on  test  of  anxiety  and  for 
the  anxiolytic  effects  of  ethanol.  Alcohol 
10:1-10,  1993. 

Stormark,  K.M.;  Laberg,  J.C.;  Bjerland, 
T.;  Nordby,  H.;  and  Hugdahl,  K.  Auto- 
nomic cued  reactivity  in  alcoholics:  The 
effect  of  olfactory  stimuli.  Addict  Behav 
20:571-584,  1995. 

Swiergiel,  A.H.;  Takahashi,  L.K.;  and 
Kalin,  N.H.  Attenuation  of  stress-induced 
behavior  by  antagonism  of  corticotropin - 
releasing  factor  receptors  in  the  central 
amygdala  in  the  rat.  Brain  Res  623:229- 
234,  1993. 


310 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


Tabakoff,  B.,  and  Hoffman,  P.L.  Alterations 
in  receptors  controlling  dopamine  synthe- 
sis after  chronic  ethanol  ingestion.  / 
Neurochem  31:1223-1229,  1978. 

Tabakoff,  B.;  Hoffman,  P.;  and  Moses,  F. 
Neurochemical  correlates  of  ethanol  with- 
drawal: Alterations  in  serotonergic  function. 
J  Ph arm  Pharmacol  29:471^176,  1977. 

Tabakoff,  B.;  Jafee,  R.C.;  and  Ritzmann,  R.F. 
Corticosterone  concentrations  in  mice 
during  ethanol  drinking  and  withdrawal.  / 
Pharm  Pharmacol  30:371-374,  1978. 

Thompson,  P.M.;  Gillin,  J.C.;  Golshan, 
S.;  and  Irwin,  M.  Polygraphic  sleep  mea- 
sures differentiate  alcoholics  and  stimu- 
lant abusers  during  short-term  abstinence. 
Biol  Psychiatry  38:831-836,1995. 

Thomson,  S.M.,  Jr.,  and  McMillen,  B.A. 
Test  for  decreased  serotonin/tryptophan 
metabolite  ratios  in  abstinent  alcoholics. 
Alcohol  4:1-5,  1987. 

Trapold,  M.A.,  and  Sullivan,  H.L.  A 
withdrawal-related  reinforcing  effect  of 
alcohol.  Bull  Psychonomic  Soc  13:327- 
329,  1979. 

Tritto,  T.,  and  Dudek,  B.C.  Chronic 
ethanol  effects  in  B  DA/2  J  mice:  True 
sensitization,  or  sedative  tolerance?  Alcohol 
Clin  Exp  Res  2l(suppl)A2A,  1997. 

Turner,  KC;  Lichstein,  P.R.;  Peden, 
J.G.,  Jr.;  Busher,  J.T.;  and  Waivers,  L.E. 
Alcohol  withdrawal  syndromes:  A  review 
of  pathophysiology,  clinical  presentation, 
and  treatment.  /  Gen  Intern  Med 
4:432^44,  1989. 

Ulrichsen,  J.;  Bech,  B.;  AUerup,  P.;  and 
Hemmingsen,  R.  Diazepam  prevents 
progression  of  kindled  alcohol  withdrawal 
behaviour.  Psychopharmacology  (Berl) 
121:451-460,  1995. 


Ulrichsen,  J.;  Bech,  B.;  Ebert,  B.;  Diemer, 
N.H.;  Allerup,  P.;  and  Hemmingsen,  R. 
Glutamate  and  benzodiazepine  receptor 
autoradiography  in  rat  brain  after  repetition 
of  alcohol  dependence.  Psychopharmacology 
(Berl)  126:31-41,  1996. 

Vavrousek-Jakuba,  E.;  Cohen,  C.A.;  and 
Shoemaker,  W.J.  Ethanol  effects  on  dopamine 
receptors:  In  vivo  binding  following  vol- 
untary ethanol  (ETOH)  intake  in  rats.  In: 
Naranjo,  C.A.,  and  Sellers,  E.M.,  eds.  Novel 
Pharmacological  Interventions  for  Alcoholism. 
New  York:  Springer,  1992.  pp.  372-374. 

Veale,  W.L.,  and  Myers,  R.D.  Increased 
alcohol  preference  in  rats  following  repeated 
exposures  to  alcohol.  Psychopharmacologia 
15:361-372,  1969. 

Viglinskaya,  I.V.  Experimental  approaches 
to  the  evaluation  of  alcohol  abuse  and 
dependence  development.  Drug  Alcohol 
Depend  30:11 1-1 15,  1992. 

Volpicelli,  J.R.;  Alterman,  A.I.;  Hayashida, 
M.;  and  O'Brien,  C.P.  Naltrexone  in  the 
treatment  of  alcohol  dependence.  Arch 
Gen  Psychiatry  49:876-880,  1992. 

Volpicelli,  J.R.;  Clay,  K.L.;  Watson,  N.T.; 
and  O'Brien,  C.P.  Naltrexone  in  the 
treatment  of  alcoholism:  Predicting  re- 
sponse to  naltrexone.  J  Clin  Psychiatry  56 
(Suppl.  7):39^4,  1995^. 

Volpicelli,  J.R.;  Volpicelli,  LA.;  and  O'Brien, 
C.P.  Medical  management  of  alcohol 
dependence:  Clinical  use  and  limitations 
of  naltrexone  treatment.  Alcohol  Alcohol 
30:789-798,  19956. 

Volpicelli,  J.R.;  Watson,  N.T.;  King,  A.C.; 
Sherman,  C.E.;  and  O'Brien,  C.P.  Effect 
of  naltrexone  on  alcohol  "high"  in  alcoholics. 
Am  J  Psychiatry  152:61 3-6 15, 1995  c. 

von  Bardeleben,  U.;  Heuser,  I.;  and  Holsboer, 
F.  Human  CRH  stimulation  response 
during  acute  withdrawal  and  after  medium- 


311 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


term  abstention  from  alcohol  abuse.  Psycho- 
neuroendocrinology  14:441-449,  1989. 

Wahlstrom,  G.;  Magnusson,  O.;  Nordberg, 
A.;  Stenstrom,  A.;  and  Tiger,  G.  Temporal 
studies  of  the  inhibition  of  voluntary 
ethanol  intake  in  the  rat  induced  by  inter- 
mittent ethanol  treatment  and  some  long 
term  neurochemical  consequences.  Drug 
Alcohol  Depend  28:129-144,  199.1. 

Wallace,  B.C.  Psychological  and  environ- 
mental determinants  of  relapse  in  crack 
cocaine  smokers.  J  Subst  Abuse  Treat 
6:95-106,  1989. 

Waller,  M.B.;  Murphy,  J.M.;  McBride, 
W.J.;  Lumeng,  L.;  and  Li,  T.-K.  Effect  of 
low-dose  ethanol  on  spontaneous  motor 
activity  in  alcohol-preferring  and  nonpre- 
ferring  rats.  Pharmacol  Bio ch em  Behav 
24:617-623,  1986. 

Wamsley,  J.K.;  Young,  W.S.D.;  and  Kuhar, 
M.J.  Anatomical  localization  of  enkephalin 
immunoreactive  sites  in  rat  forebrain.  Adv 
Biochem  Psychopharmacol  22:257-270, 1980. 

Wand,  G.  S.,  and  Dobs,  A.S.  Alterations 
in  the  hypothalamic-pituitary-adrenal  axis 
in  actively  drinking  alcoholics.  /  Clin 
Endocrinol  Metab  72:1290-1295,  1991. 

Watson,  W.P.;  Robinson,  E.;  and  Little,  H.J. 
The  novel  anticonvulsant,  gabapentin,  protects 
against  both  convulsant  and  anxiogenic 
aspects  of  the  ethanol  withdrawal  syndrome. 
Neuropharmacology  36:1369-1375,  1997. 

Weiss,  R;  Mitchiner,  M.;  Bloom,  F.E.;  and 
Koob,  G.F.  Free-choice  responding  for 
ethanol  versus  water  in  alcohol-preferring 
(P)  and  unselected  Wistar  rats  is  differen- 
tially altered  by  naloxone,  bromocriptine 
and  methysergide.  Psychopharmacology 
101:178-186,  1990. 

Weiss,  F.;  Lorang,  M.T.;  Bloom,  F.E.; 
and  Koob,  G.F.  Oral  alcohol  self-adminis- 
tration stimulates  dopamine  release  in  the 


rat  nucleus  accumbens:  Genetic  and  moti- 
vational determinants.  J  Pharmacol  Exp 
Ther  267:250-258,  1993. 

Weiss,  F.;  Parsons,  L.H.;  Schulteis,  G.; 
Lorang,  M.T.;  Hyytia,  P.;  Bloom,  F.E.; 
and  Koob,  G.F.  Ethanol  self- administra- 
tion restores  withdrawal-associated  defi- 
ciencies in  accumbal  dopamine  and 
serotonin  release  in  dependent  rats.  / 
Neurosci  16:3474-3485,  1996. 

Weiss,  F.;  Imperato,  A.;  Casu,  M.A.; 
Mascia,  M.S.;  and  Gessa,  G.L.  Opposite 
effects  of  stress  on  dopamine  release  in 
the  limbic  system  of  drug-naive  and 
chronically  amphetamine -treated  rats.  Eur 
J Pharmacol  337 ':219-222,  1997. 

Weissenborn,  R;  Yackey,  M.;  Koob,  G.F.; 
and  Weiss,  F.  Measures  of  cocaine-seeking 
behavior  using  a  multiple  schedule  of 
food  and  drug  self- administration  in  rats. 
Drug  Alcohol  Depend  38:237-246,  1995. 

Whitworth,  A.B.;  Fischer,  F.;  Lesch, 
O.M.;  Nimmerrichter,  A.;  Oberbauer,  PL; 
Platz,  T.;  Potgieter,  A.;  Walter,  PL;  and 
Fleischhacker,  WW.  Comparison  of 
acamprosate  and  placebo  in  long-term 
treatment  of  alcohol  dependence.  Lancet 
347:1438-1442,  1996. 

Widdowson,  P.S.,  and  Holman,  R.B. 
Ethanol-induced  increase  in  endogenous 
dopamine  release  may  involve  endogenous 
opiates.  J Neurochem  59:157-163,  1992. 

Widnell,  K.L.;  Self,  D.W.;  Lane,  S.B.; 
Russell,  D.S.;  Vaidya,  V.A.;  Miserendino, 
M.J.;  Rubin,  C.S.;  Duman,  RS.;  and  Nestler, 
E.J.  Regulation  of  CREB  expression:  In 
vivo  evidence  for  a  functional  role  in  mor- 
phine action  in  the  nucleus  accumbens.  / 
Pharmacol  Exp  Ther  276:306-315,  1996. 

Wieland,  S.;  Lan,  N.C.;  Mirasedeghi,  S.; 
and  Gee,  K.W.  Anxiolytic  activity  of  the 


312 


Neuroadaptive  Changes  in  Neurotransmitter  Systems 


progesterone  metabolite  5a-pregan-3ct- 
ol-20-one.  Brain  Res  565:263-268,  1991. 

Wilder,  A.  Dynamics  of  drug  dependence: 
Implications  of  a  conditioning  theory  for 
research  and  treatment.  Arch  Gen  Psychiatry 
28:611-616,1973. 

Wilde,  M.I.,  and  WagstafF,  A.J.  Acamprosate. 
A  review  of  its  pharmacology  and  clinical 
potential  in  the  management  of  alcohol 
dependence  after  detoxification.  Drugs 
53:1038-1053,  1997. 

Wilkins,  J.N.;  Gorelick,  D.A.;  Nademanee, 
K.;  Taylor,  A.;  and  Herzberg,  D.S. 
Hypothalamic-pituitary  function  during 
alcohol  exposure  and  withdrawal  and 
cocaine  exposure.  Recent  Dev  Alcohol 
10:57-71,  1992. 

Wilson,  C;  Nomikos,  G.G.;  Collu,  M.; 
and  Fibiger,  H.C.  Dopaminergic  correlates 
of  motivated  behavior:  Importance  of 
drive.  JNeurosci  15:5169-5178,  1995. 

Winger,  G.  Effects  of  ethanol  withdrawal 
on  ethanol-reinforced  responding  in 
rhesus  monkeys.  Drug  Alcohol  Depend 
22:235-240,  1988. 

Wise,  R.A.  Neurobiology  of  addiction. 
Curr  Opin  Neurobiol  6:243-251,  1996. 

Wolffgramm,  J.  An  ethopharmacological 
approach  to  the  development  of  drug  addic- 
tion. Neurosci  Biobehav  Rev  15:515-519, 1991. 

Wolffgramm,  J.,  and  Heyne,  A.  Social 
behavior,  dominance  and  social  deprivation 
of  rat  determine  drug  choice.  Pharmacol 
Biochem  Behav  38:389-399,  1991. 

Wolffgramm,  J.,  and  Heyne,  A.  From 
controlled  drug  intake  to  loss  of  control: 
The  irreversible  development  of  drug 
addiction  in  the  rat.  Behav  Brain  Res 
70:77-94,  1995. 

Woods,  J.H.,  and  Winger,  G.D.  A  critique 
of  methods  for  inducing  ethanol  self- 


intoxication  in  animals.  In:  Mello,  N.K., 
and  Mendelson,  J.H.,  eds.  Recent  Advances 
in  Studies  of  Alcoholism.  Rockville,  MD: 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism,  1971.  pp.  413-436. 

Wozniak,  K.M.;  Pert,  A.;  Mele,  A.;  and 
Linnoila,  M.  Focal  application  of  alcohols 
elevates  extracellular  dopamine  in  rat  brain: 
A  microdialysis  study.  Brain  Res  540:31- 
40,  1991. 

Yamamura,  T.;  Hishida,  S.;  Hatake,  K.; 
Taniguchi,  T.;  and  Ouchi,  H.  Effects  of 
methamphetamine  and  ethanol  on  learning 
and  brain  neurotransmitters  in  rats. 
Pharmacol  Biochem  Behav  42:389-400, 1992. 

Yoshimoto,  K,  and  McBride,  W.J.  Regulation 
of  nucleus  accumbens  dopamine  release 
by  the  dorsal  raphe  nucleus  in  the  rat. 
Neurochem  Res  17:401-407,  1992. 

Yoshimoto,  K.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Alcohol  stimulates  the 
release  of  dopamine  and  serotonin  in  the 
nucleus  accumbens.  Alcohol 9 :\7 -22,  \992a. 

Yoshimoto,  K.;  McBride,  W.J.;  Lumeng, 
L.;  and  Li,  T.-K.  Ethanol  enhances  the 
release  of  dopamine  and  serotonin  in  the 
nucleus  accumbens  of  FIAD  and  LAD  lines  of 
rats.  Alcohol  Clin  Exp  Res  16:781-785, 1992& 

Yoshimoto,  K;  Yayama,  K.;  Sorimachi,  Y.; 
Tani,  J.;  Ogata,  M.;  Nishimura,  A.; 
Yoshida,  T.;  Ueda,  S.;  and  Komura,  S. 
Possibility  of  5-HT3  receptor  involvement 
in  alcohol  dependence:  A  microdialysis 
study  of  nucleus  accumbens  dopamine  and 
serotonin  release  in  rats  with  chronic 
alcohol  consumption.  Alcohol  Clin  Exp 
ito20:311A-319A,  1996. 

Zeise,  M.L.;  Kasparov,  S.;  Capogna,  M.; 
and  Zieglgansberger,  W.  Acamprosate 
(calcium  acetylhomotaurinate)  decreases 
postsynaptic  potentials  in  the  rat  neocor- 
tex. Eur  J  Pharmacol  231:47-52,  1993. 


313 


Chapter  8 

Adolescent  Period:  Biological  Basis  of 

Vulnerability  To  Develop  Alcoholism 

and  Other  Ethanol-Mediated  Behaviors 

Linda  Patia  Spear,  Ph.D. 


KEY  WORDS:  AOD  (alcohol  or  other  drug)  dependence;  adolescence;  biological 
AOD  use  disorder  theory;  psychological  AODC  (causes  of  AOD  use,  abuse,  and 
dependence);  AOD  use  susceptibility;  AOD  use  behavior;  growth  and  develop- 
ment; brain  function;  physiological  stress;  dopamine;  AOD  sensitivity;  AOD  tol- 
erance; hormones;  AOD  use  initiation;  prevalence;  predictive  factor;  animal 
study;  etiology;  literature  review 


THE  IMPORTANCE  OF 
ADOLESCENCE  IN 
ALCOHOL  STUDIES 

Adolescents  across  a  variety  of  species 
are  faced  with  the  similar  developmental 
challenge  of  acquiring  the  necessary  skills 
to  permit  survival  away  from  parental 
caretakers.  At  this  critical  juncture 
between  childhood  and  adulthood, 
adolescents  undergo  marked  hormonal 
and  neural  alterations.  Among  the  brain 
regions  showing  prominent  alterations 
during  adolescence  are  the  prefrontal 
cortex  (PFC)  and  other  forebrain  dopa- 
mine (DA)  projection  regions,  areas 
implicated  in  mediating  the  reinforcing 
effects  of  alcohol  and  other  drugs  of 


abuse  (see  Koob  1992;  Goeders  1997 
for  review). 

It  is  in  this  unique  neurobehavioral 
state  of  adolescence  that  most  humans 
begin  alcohol  use,  yet  little  is  known 
about  alcohol  in  adolescence.  This  age 
is  critical  for  study  for  three  reasons. 
First,  brain  function  in  regions  modu- 
lating drug  reinforcement  is  altered 
during  adolescence,  and  it  cannot  be 
assumed  that  factors  precipitating 
alcohol  use  or  abuse  would  be  the 
same  in  adolescence  as  in  adulthood. 
Second,  rapidly  changing  systems  are 
particularly  vulnerable  to  disruption,  so 
there  may  be  long-term  consequences 
of  alcohol  exposure  during  this  time  of 
rapid  neural  and  endocrine  maturation. 


L.P.  Spear,  Ph.D.,  is  a  Distinguished  Professor  in  the  Department  of  Psychology  and  director  of  the 
Center  for  Developmental  Psychobiology  at  Binghamton  University,  Binghamton,  NY  13902-6000. 


315 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Third,  early  onset  of  alcohol  use  is 
currently  one  of  the  most  powerful 
predictors  of  later  alcohol  abuse. 
Before  considering  these  issues  in 
more  detail,  a  few  pertinent  age -spe- 
cific characteristics  of  adolescence  will 
be  briefly  summarized. 

CHARACTERISTICS  OF 
ADOLESCENCE  IN 
HUMANS  AND  OTHER 
ANIMALS 

The  process  of  adolescence  is  not  syn- 
onymous with  puberty.  Whereas  ado- 
lescence subsumes  the  entire  process  of 
transition  from  childhood  to  adulthood, 
puberty  is  a  more  temporally  restricted 
phase  during  which  the  physiological 
and  neuroendocrine  alterations  associ- 
ated with  sexual  maturation  occur. 
Puberty  is  but  one  of  the  numerous 
ontogenetic  alterations  occurring  during 
adolescence,  with  the  timing  of  this 
maturational  event  within  the  broader 
framework  of  adolescence  varying 
noticeably  among  human  adolescents 
(see,  e.g.,  Dubas  1991).  In  humans, 
adolescence  spans  the  age  range  from  as 
early  as  9  years  to  approximately  18 
years  (see,  e.g.,  Buchanan  et  al.  1992). 
In  rats,  commonly  cited  times  for  ado- 
lescence onset  are  postnatal  days  28-32 
(P28-32),  with  offsets  between  P38 
and  P55  (see,  e.g.,  Ojeda  and  Urban- 
ski  1994),  although  this  timing  is 
somewhat  disputed  (Odell  1990)  and 
may  depend  on  the  growth  rate  of  the 
animals  (Kennedy  and  Mitra  1963)  and 
the  maturational  index  used.  Spear  and 
Brake  (1983)  operationally  defined 
"periadolescence"  as  the  age  period 
around  the  time  of  sexual  maturation 


when  age-specific  behavioral  and  psycho- 
pharmacological  discontinuities  are  evi- 
dent; using  this  criteria  the  age  period  of 
approximately  P30-42  was  conserva- 
tively designated  as  periadolescence, 
with  animals  of  this  age  showing 
numerous  neurobehavioral  alterations 
from  significantly  younger  (pre-  or 
postweanlings)  as  well  as  more  mature 
(P60  and  older)  animals.  Adolescence 
in  monkeys  typically  occurs  in  the  age 
range  of  2-\  years  (Lewis  1997). 

Hormonal  Concomitants  of 
Adolescence 

Puberty  represents  a  reactivation,  after 
a  prolonged  period  of  suppression  during 
the  childhood/juvenile  period,  of  pul- 
satile release  of  gonadotropin-releasing 
hormone  (GnRH)  that  was  evident 
perinatally.  This  reinstatement  of  pulsatile 
GnRH  release  induces  pulsed  release  of 
follicle-stimulating  hormone  and  luteiniz- 
ing hormone,  which  in  turn  stimulate 
release  of  gonadal  hormones  (e.g.,  testos- 
terone in  males  and  estrogen  in  females) 
(see,  e.g.,  Brooks-Gunn  and  Reiter 
1990).  Pulsatile  release  of  growth  hor- 
mone also  increases  more  than  tenfold 
during  the  growth  spurt  of  adolescence 
(Gabriel  et  al.  1992).  It  remains  to  be 
determined  which  neural  and  behavioral 
features  of  adolescence  are  driven  by 
maturational  changes  in  gonadal  hor- 
mones, and  which  instead  may  emerge 
independently  from  (or  in  the  case  of 
neural  alterations,  might  even  contribute 
to)  processes  of  sexual  maturation  per  se. 

Behavioral  Characteristics 
of  Adolescence 

Periadolescents  differ  behaviorally  from 
younger  and  older  individuals  on  a 


316 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


number  of  dimensions  consistent  with 
a  developmental  trajectory  toward  the 
goal  of  independence.  Rats  in  the  age 
range  from  approximately  P30  to  P42 
are  often  hyperactive  and  explore  more 
relative  to  rats  of  other  ages  (see,  e.g., 
Spear  et  al.  1980).  They  also  spend  more 
time  in  social  interactions  with  conspecifics 
(Primus  and  Kellogg  1989)  and  exhibit 
peak  levels  of  play  behavior  (see,  e.g., 
Fassino  and  Campbell  1981).  Sex  dif- 
ferences in  behavior  also  begin  to 
emerge  in  adolescence,  with  some  of 
these  differences  being  driven  in  part  by 
organizational  influences  of  pubertal 
hormones  (see,  e.g.,  Beatty  and  Fessler 
1977;  Brand  and  Slob  1988).  Human 
adolescents  likewise  exhibit  increases  in 
social  behavior,  as  well  as  a  dispropor- 
tionate amount  of  reckless  behavior, 
sensation  seeking,  and  risk  taking  rela- 
tive to  individuals  at  other  ages  (Arnett 
1992).  Such  age-related  modifications 
in  behavior  are  consistent  with  the  need 
of  the  adolescent  to  explore  novel 
domains  and  establish  new  social  rela- 
tionships during  the  process  of  achieving 
independence  from  their  parents. 

In  addition  to  the  continuing  devel- 
opment of  cognitive  function  during 
adolescence  (see,  e.g.,  Levin  et  al.  1991), 
age-specific  discontinuities  are  seen  in 
some  learning  tasks.  Adolescent  rats 
sometimes  exhibit  enhanced  perfor- 
mance on  tasks  in  which  increases  in 
activity/exploration  could  facilitate 
performance  (e.g.,  radial  arm  maze 
[Chambers  et  al.  1996]  and  active 
avoidance  [Bauer  1980]).  However, 
relative  to  younger  or  older  rats  ado- 
lescents tend  toward  impaired  perfor- 
mance on  more  complex  avoidance 
tasks  (such  as  discriminated  escape 


and  Sidman  avoidance  tasks),  perhaps 
as  a  function  of  increased  distractibility 
and  difficulties  in  focusing  attention 
on  salient  cues  and  reward  contingen- 
cies (see  Spear  and  Brake  1983  for 
review  and  references). 

Adolescents  also  exhibit  characteris- 
tic alterations  in  psychopharmacological 
sensitivity.  For  instance,  adolescent  rats 
are  less  sensitive  than  their  younger  or 
older  counterparts  to  the  stimulatory 
effects  of  catecholaminergic  agonists 
such  as  amphetamine  and  cocaine,  but 
conversely  are  more  sensitive  to  the 
DA  antagonist  haloperidol,  a  psy- 
chopharmacological pattern  sugges- 
tive of  a  temporary  hyposensitivity  of 
one  or  more  DA  systems  during  ado- 
lescence (see  Spear  and  Brake  1983 
for  references  and  discussion).  Adult- 
typical  suppressant  effects  of  low  doses 
of  D2/D3  DA  agonists  also  emerge 
during  early  adolescence  (see,  e.g., 
Shalaby  and  Spear  1980;  Arnt  1983; 
Van  Hartesveldt  et  al.  1994),  an  effect 
formerly  thought  to  be  associated 
with  development  of  DA  autorecep- 
tors  (Shalaby  et  al.  1981;  Hedner  and 
Lundborg  1985)  but  that  instead  may 
be  mediated  by  maturation  of  a  sub- 
population  of  postsynaptic  DA  recep- 
tors (see  Andersen  et  al  1997a  for 
evidence  refuting  an  autoreceptor 
explanation;  see  also  Stahle  1992). 

Neural  Alterations  During 
Adolescence 

The  adolescent  brain  is  unique  and  in 
a  state  of  transition  as  it  undergoes 
both  progressive  and  regressive 
changes.  One  brain  region  prominently 
altered  during  adolescence  across  a 
variety  of  species  is  the  PFC,  an  area 


317 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


thought  to  subserve  higher  cognitive 
abilities  such  as  the  bridging  of  tem- 
poral delays  in  memory  (see,  e.g.,  Dia- 
mond 1991).  For  instance,  absolute 
PFC  volume  declines  in  adolescence 
in  humans  (Jernigan  et  al.  1991)  and 
in  rats  (van  Eden  et  al.  1990).  Sub- 
stantial synapse  elimination  occurs 
during  adolescence  in  the  PFC  and 
other  cortical  regions  in  humans 
(Huttenlocher  1984)  and  nonhuman 
primates  (Zecevic  et  al.  1989).  At 
least  a  portion  of  this  synapse  elimina- 
tion in  the  PFC  appears  to  be  associ- 
ated with  the  marked  developmental 
loss  of  presumed  glutaminergic  excita- 
tory input  (Zecevic  et  al.  1989).  In 
contrast,  DA  input  to  the  PFC  in 
nonhuman  primates  increases  during 
adolescence  to  peak  at  levels  well 
above  those  seen  earlier  or  later  in  life 
(Rosenberg  and  Lewis  1994;  see  Lewis 
1997  for  a  review);  increases  in  PFC 
DA  input  through  adolescence  are  also 
evident  in  rats  (Kalsbeek  et  al.  1988). 
Cholinergic  innervation  of  the  PFC 
likewise  increases  in  adolescence  to 
reach  mature  levels  in  rats  (Gould  et  al. 
1991)  and  humans  (Kostovic  1990). 

Maturational  changes  during  ado- 
lescence are  also  evident  in  other  brain 
regions  such  as  the  hippocampus  of 
rodents  (Wolfer  and  Lipp  1995;  Dumas 
and  Foster  1998)  and  humans  (Benes 
1989).  Alterations  evident  in  the  hypo- 
thalamus include  qualitative  differences 
in  norepinephrine  release  in  adolescents 
relative  to  younger  or  older  rats,  along 
with  pharmacological  alterations  consis- 
tent with  the  suggested  emergence  in  ado- 
lescence of  inhibitory  a2  norepinephrine 
autoreceptors  (Choi  and  Kellogg  1992; 
Choietal.  1997). 


Dopaminergic  systems  undergo 
substantial  reorganization  during  ado- 
lescence. Over  one-third  to  one-half  of 
the  Dl  and  D2  receptors  present  in 
the  striatum  of  juveniles  are  lost  by 
adulthood  in  both  humans  (Seeman 
et  al.  1987)  and  rats  (Gelbard  et  al. 
1989;  Teicher  et  al.  1995).  This  peak 
in  Dx  and  D2  binding  during  adoles- 
cence and  subsequent  decline  is  much 
more  pronounced  in  striatum  than  in 
nucleus  accumbens  (Teicher  et  al. 
1995),  and  in  male  than  in  female  rats 
(Andersen  et  al.  1997b).  Not  all  DA 
receptors  show  this  overproduction 
and  pruning,  with  juveniles  having 
only  40  percent  of  adult-typical  D3 
receptor  levels  in  striatal  and  accum- 
bens regions  (Stanwood  et  al.  1997). 
The  DA  transporter  likewise  under- 
goes a  protracted  period  of  develop- 
ment in  mesolimbic  and  mesocortical 
brain  regions,  with  only  about  70  per- 
cent of  adult  uptake  levels  being  seen 
prior  to  adolescence  onset  in  rats 
(Coulter  et  al.  1996). 

Developmental  events  during  adoles- 
cence may  alter  the  relative  balance  of 
DA  activity  between  the  PFC  and  stri- 
atal or  mesolimbic  terminal  regions. 
Basal  DA  synthesis  increases  during 
adolescence  in  the  nucleus  accumbens 
and  the  striatum  of  rats,  while  the  rate 
of  DA  synthesis  peaks  in  the  PFC  at 
P30  before  declining  to  much  lower 
levels  by  late  adolescence  (Andersen  et 
al  1997  a).  Similar  data  are  obtained 
from  estimates  of  DA  turnover,  with 
basal  turnover  increasing  during  ado- 
lescence in  the  nucleus  accumbens  and 
the  striatum  and  decreasing  in  the 
PFC  (Teicher  et  al.  1993).  Interestingly, 
although  the  PFC  is  seemingly  devoid 


318 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


of  synthesis-modulating  autoreceptors 
in  adulthood  (see,  e.g.,  Galloway  et 
al.  1986),  convincing  evidence  has 
been  obtained  for  a  transient  expres- 
sion of  DA  autoreceptor-like  modula- 
tion of  DA  synthesis  in  the  PFC  early 
in  life  that  disappears  during  adoles- 
cence (Teicher  et  al.  1991;  Andersen 
etal.  1997  a). 

The  brain  of  the  adolescent  is  clearly 
in  transition.  Neural  regions  showing 
prominent  alterations  during  adoles- 
cence include  the  PFC  as  well  as  other 
forebrain  DA  projection  regions.  Given 
the  important  role  of  these  brain  areas 
in  modulating  reward  efficacy  of  rein- 
forcing drugs  (Koob  1992),  sensitiv- 
ity to  the  environment  and  stressors 
(see,  e.g.,  Dunn  and  Kramarcy  1984), 
and  the  association  between  the  two  (see, 
e.g.,  Piazza  et  al.  1991;  Goeders  1997), 
it  is  not  surprising  that  adolescents  vary 
notably  from  more  mature  animals  in 
their  responsivity  to  ethanol,  stressors, 
and  their  interaction. 

ONTOGENY  OF 
RESPONSIVITY  TO 
ETHANOL 

Prevalence  of  Alcohol  Use 
in  Adolescents 

In  a  1996  National  Institute  on  Drug 
Abuse  survey,  26  percent  of  8th 
graders,  40  percent  of  10th  graders, 
and  51  percent  of  12th  graders 
reported  that  they  had  used  alcohol  in 
the  past  month.  Ten  percent  of  8th 
graders,  21  percent  of  10th  graders, 
and  31  percent  of  12th  graders 
(Mathias  1997)  also  reported  getting 
drunk  on  one  or  more  occasions  during 


the  past  month.  Clearly,  many  adoles- 
cents use  alcohol,  with  evidence  of  exces- 
sive use  emerging  in  some  individuals. 

Adolescent  rats  display  two  to  three 
times  higher  levels  of  ethanol  intake 
relative  to  their  body  weights  than  do 
more  mature  animals  (Lancaster  et  al. 
1996;  Bannoura  et  al.  unpublished 
manuscript),  although  ethanol  prefer- 
ence per  se  does  not  peak  until  well  into 
adulthood  (around  5  months  of  age 
[Parisella  and  Pritham  1964;  Goodrick 
1967]).  The  notably  different  ontoge- 
netic conclusions  reached  when  using 
grams  per  kilogram  intake  versus  per- 
cent total  fluid  to  index  ethanol  con- 
sumption seemingly  reflect  ontogenetic 
differences  in  total  fluid  consumption, 
with  adolescent  rats  exhibiting  greater 
overall  fluid  (and  food)  consumption 
than  adults.  Indeed,  during  the  ado- 
lescent growth  spurt,  adolescent  rats 
consume  the  greatest  caloric  intake 
relative  to  their  body  weight  of  any 
time  in  the  lifespan  (see,  e.g.,  Nance 
1983).  Adolescent  humans  also 
exhibit  elevated  metabolic  activity  and 
developmental  hyperphagia  (see,  e.g., 
Post  and  Kemper  1993;  Ganji  and 
Betts  1995),  with  heavy  alcohol  use 
often  being  "adolescence-limited" 
(see,  e.g.,  Bates  and  Labouvie  1997). 

The  elevated  consummatory  behav- 
iors of  adolescence  could  contribute  to 
high  levels  of  ethanol  intake  by  these 
growing  individuals  relative  to  their 
body  weight.  As  discussed  below, 
adolescents  might  be  able  to  sustain 
comparatively  large  ethanol  intakes 
due  to  their  relative  insensitivity  to  the 
sedative  and  locomotor  incoordinating 
effects  of  ethanol,  which  may  be  in  part 
related  to  their  greater  propensity  to 


319 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


develop  acute  and  functional  tolerance 
relative  to  more  mature  organisms. 

Acute  Responsivity 
to  Ethanol 

Studies  using  a  variety  of  measures  in 
laboratory  animals  have  observed  increases 
in  ethanol  sensitivity  from  infancy,  with 
further  increases  in  sensitivity  during  the 
aging  process  (see,  e.g.,  York  and  Chan 
1993).  This  early  attenuation  in  ethanol 
sensitivity  is  evident  despite  slower  rates 
of  ethanol  metabolism  in  younger  ani- 
mals (see,  e.g.,  Zorzano  and  Herrera 
1989;  Silveri  and  Spear  2000)  and  is 
evident  using  measures  such  as  LD50 
(Hollstedt  and  Rydberg  1985),  tilting- 
plane  performance  (Hollstedt  et  al.  1980), 
ethanol-induced  hypnosis  (Ernst  et  al. 
1976;  Little  et  al.  1996;  Silveri  and 
Spear  1998),  and  hypothermia  (Spiers 
and  Fusco  1991;  Silveri  and  Spear 
2000).  However,  this  finding  of  early 
attenuation  in  ethanol  sensitivity  is  not 
universal — see,  for  example,  Keir  and 
Deitrich(1990). 

Tolerance  Development 

The  resistance  of  young  organisms  to 
ethanol  may  be  in  part  attributable  to  the 
development  of  pronounced  acute  toler- 
ance early  in  life,  with  the  propensity  for 
acute  tolerance  to  ethanol  gradually 
declining  to  reach  adult  levels  only  fol- 
lowing puberty  (Silveri  and  Spear  1998). 
This  ontogenetic  decline  may  be  specific 
to  within- session  tolerance,  with  other 
forms  of  tolerance  such  as  rapid  toler- 
ance showing  ontogenetic  increases 
(Silveri  and  Spear  1999). 

Grieve  and  Littleton  (1979)  reported 
that  preweanling  mice  showed  no 
evidence  of  functional  tolerance  to 


ethanol-induced  sleep,  whereas  adoles- 
cents showed  more  pronounced  tolerance 
development  than  adults.  Adolescents 
also  have  been  reported  to  exhibit  more 
chronic  tolerance  to  ethanol-induced 
hypothermia  than  adult  rats  (Swartz- 
welder  et  al.  1998).  This  greater  propen- 
sity for  adolescents  to  develop  acute  and 
chronic  tolerance  may  contribute  to  their 
relative  resistance  to  the  motor-impair- 
ing and  sedative  effects  of  ethanol  relative 
to  their  more  mature  counterparts. 

Adolescent  Vulnerabilities 
to  Ethanol  Disruption 

In  some  respects,  young  rats  may  be  un- 
usually sensitive  to  ethanol.  Swartzwelder 
and  his  group  found  that  hippocampal 
slices  from  preadolescent  (PI 5-25)  rats 
were  more  sensitive  than  adult  slices  to 
ethanol  disruption  of  both  NMDA- 
mediated  excitation  and  stimulus-induced 
long-term  potentiation  (Swartzwelder 
et  al.  1995^,  1995£).  Behaviorally,  P30 
adolescents  were  found  to  be  more 
impaired  than  adult  rats  by  ethanol  in  a 
spatial  memory  task  in  the  Morris  maze, 
while  nonspatial  performance  was  unaf- 
fected by  ethanol  at  either  age  (Markwiese 
et  al.  1998).  Although  reduced  sensi- 
tivity to  motor-impairing  and  sedative 
consequences  of  ethanol  may  permit 
adolescents  to  consume  greater 
amounts  of  ethanol,  this  exposure  might 
have  more  adverse  effects  on  hip- 
pocampally-related  memory  processing 
than  later  in  life. 

STRESS,  ADOLESCENCE, 
AND  ALCOHOL  ABUSE 

Navigating  the  developmental  transition 
toward  independence  may  be  stressful 


320 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


for  adolescents.  In  humans,  for 
instance,  levels  of  anxiety  have  been 
reported  to  peak  at  around  13-15 
years  of  age  (see  Buchanan  et  al.  1992 
for  discussion  and  references).  This 
presumed  increase  in  stress  during 
adolescence  has  been  postulated  to 
contribute  to  the  frequent  initiation  of 
alcohol  and  other  drug  use  in  adoles- 
cence (see,  e.g.,  Pohorecky  1991; 
Wagner  1993),  as  well  as  to  the  fre- 
quent emergence  in  adolescence  of 
schizophrenic  symptomology  in  vul- 
nerable individuals  (Walker  and 
Diforio  1997).  In  addition  to  the 
actual  frequency  of  life  stressors  possi- 
bly being  greater  in  adolescence  than 
at  other  ages,  adolescents  may  also 
respond  differently  to  stress  than  indi- 
viduals at  other  ages. 

Hormonal  Response  to 
Stressors  in  Adolescents 

Exposure  to  a  stressor  activates  the 
hypothalamic-pituitary- adrenal  (HPA) 
axis,  resulting  in  a  cascading  sequence 
of  hormone  release  from  the  hypo- 
thalamus (corticotropin-releasing  fac- 
tor), pituitary  (adrenocorticotropic 
hormone  [ACTH]),  and  adrenals 
(corticosterone  in  rats;  Cortisol  in 
humans).  Ontogenetic  increases  in 
stress-induced  activation  of  the  HPA 
system  have  been  explored  systemati- 
cally in  animal  studies.  Peak  ACTH 
and  corticosterone  responses  to  stress 
generally  increase  during  ontogeny  to 
reach  an  asymptote  in  rats  around 
adolescence,  at  least  in  males  (Ramaley 
and  Olson  1974;  Meaney  et  al.  1985&; 
Walker  et  al.  1986;  Bailey  and  Kitchen 
1987;  Rivier  1989).  Gender  differences 
in  the  corticosterone  response  to  stress 


begin  to  emerge  late  in  adolescence, 
with  elevated  levels  in  female  rats 
compared  with  males  and  with  prepu- 
bescent  females  (Ramaley  1972;  Cir- 
ulli  et  al.  1996). 

Adolescent  rats  sometimes  exhibit 
more  prolonged  stress-induced  increases 
in  corticosterone  than  adults  (Gold- 
man et  al.  1973;  Sapolsky  et  al.  1985; 
Choi  and  Kellogg  1996).  This  delayed 
poststress  recovery  presumably  reflects 
immature  feedback  regulation  mediated 
in  part  by  glucocorticoid  receptors  in 
hippocampus  (see,  e.g.,  Meaney  et  al. 
1985«,  1985&).  Thus,  adolescence 
may  be  associated  with  a  greater  over- 
all corticoid  response  to  stress,  with 
this  stress-induced  increase  being  ele- 
vated relative  to  younger  animals  and 
prolonged  relative  to  adults. 

Behavioral  and 
Physiological  Stress 
Responses  of  Adolescents 

Although  limited  in  number,  studies 
in  laboratory  animals  have  shown  that 
adolescents  are  sometimes  more  dis- 
rupted behaviorally  by  stressors  than 
are  adults.  Compared  with  adults, 
adolescent  rats  show  more  stress- 
induced  immobility  during  forced  swim 
testing  (Walker  et  al.  1995)  or  in  the 
presence  of  intermittent  footshock 
(Campbell  et  al.  unpublished  manu- 
script). Tail-pinch-induced  feeding 
also  has  been  reported  to  peak  in 
"juvenile"  rats  (Heinrichs  et  al. 
1992),  although  the  precise  ontogeny 
of  this  response  has  apparently  not 
been  well  characterized.  Digging  in 
novel  or  other  mildly  stressful  situations 
is  another  response  that  appears  to  peak 
in  adolescence  in  gerbils  (Wiedenmayer 


321 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


1997)  and  rats  (S.  Barron,  personal 
communication,  June  1998).  Stone 
and  Quartermain  (1998)  reported 
that  chronic  social  stress  (placement  in 
the  cage  of  an  isolated  adult  male  for 
5  minutes  daily  for  5  days)  had 
a  greater  impact  on  adolescent 
(P28-32)  than  adult  male  mice,  sup- 
pressing food  intake,  body  weight 
gain,  and  time  spent  on  open  arms  of 
a  plus  maze  in  adolescents  but  not 
adults.  Chronic  restraint  stress  was 
also  found  to  suppress  body  weight 
gain  in  adolescents  but  not  adults. 

Adult-typical  environmental  inhibi- 
tion of  social  behavior  emerges  in 
adolescence,  with  an  unfamiliar  envi- 
ronment decreasing  social  interactions 
in  adult  (P60)  and  midadolescent 
(P35),  but  not  early  adolescent  (P28), 
male  rats  (Primus  and  Kellogg  1989). 
Rats  at  P28  were  similarly  reported  to 
be  insensitive  to  effects  of  the  environ- 
ment on  the  benzodiazepine/gamma- 
aminobutyric  acid  receptor  complex 
(Primus  and  Kellogg  1991)  and  to  show 
less  stress-induced  Fos  immunoreac- 
tivity  than  adult  rats  in  brain  regions 
such  as  the  anterior  olfactory  nucleus, 
anterior  cingulate  cortex,  and  medial 
and  cortical  amygdaloid  nuclei  (Kellogg 
etal.  1998). 

Choi  and  Kellogg  (1996)  observed 
a  blunted  hypothalamic  norepineph- 
rine response  to  stress  in  late  adolescent 
rats  (P42),  a  transition  between  the 
increased  stress-related  norepinephrine 
utilization  seen  in  early  adolescence 
(P28)  and  the  decreased  utilization 
seen  in  adulthood.  A  similar  adolescent 
transitional  period  was  seen  in  terms 
of  autonomic  reactivity  to  stressor 
stimuli;  whereas  preweanling  rat  pups 


exhibit  heart  rate  bradycardia  to  an 
aversive  stimulus,  heart  rate  tachycardia 
emerges  by  adolescence,  with  this 
increased  heart  rate  mediated  by  para- 
sympathetic withdrawal  in  adolescents 
but  primarily  by  sympathetic  activation 
in  adults  (Kurtz  and  Campbell  1994). 
Taken  together,  these  data  suggest 
that  adolescents  may  differ  hormonally, 
behaviorally,  and  neurally  in  the  way 
they  respond  to  stressors  when  com- 
pared with  animals  of  other  ages. 

Stress  and  Alcohol 
Consumption  in  Adolescents 

Corticosterone  levels  in  rats  generally 
have  been  positively  related  to  rates  of 
self- administration  of  ethanol  or  other 
drugs,  with  adrenalectomy  suppress- 
ing ethanol  consumption  (Fahlke  et 
al.  1994)  and  stress-induced  eleva- 
tions in  corticosterone  increasing 
ethanol  consumption  (see,  e.g.,  Bowers 
et  al.  1997),  although  the  interaction 
of  stress  and  ethanol  intake  is  complex 
(see  Pohorecky  1990  for  a  review). 
Stressors  may  also  enhance  the  rate  of 
development  of  tolerance  to  ethanol 
(Maier  and  Pohorecky  1986),  which 
could  indirectly  contribute  to  the 
capacity  for  increased  consumption. 
The  overall  greater  corticoid  response 
to  stress  that  adolescents  seem  to 
exhibit  relative  to  individuals  at  other 
ages  may  increase  their  propensity  for 
self- administration  of  ethanol. 

Indeed,  perceived  levels  of  stress  may 
be  one  of  a  number  of  factors  exacer- 
bating the  already  elevated  propensity 
of  human  adolescents  to  exhibit  alco- 
hol use  and  other  drug-taking  behav- 
ior (Wills  1986;  Baer  et  al.  1987; 
Deykin  et  al.  1987;  Tschann  et  al. 


322 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


1994;  but  see  also  Hansell  and  White 
1991).  In  her  review  of  the  literature 
on  stress  effects  on  alcohol  consumption 
in  humans,  Pohorecky  (1991)  con- 
cluded that  stress  is  most  convincingly 
associated  with  alcohol  consumption 
in  adolescence,  with  more  mixed  findings 
evident  in  studies  conducted  in  adults. 
Indeed,  after  peer  substance  use,  the  next 
most  powerful  predictor  of  adolescent 
alcohol  and  drug  use  was  found  by 
Wagner  (1993)  to  be  levels  of  perceived 
stress,  with  the  appraisal  of  events  as 
being  stressful  of  more  importance  than 
the  absolute  number  of  such  events. 

Much  remains  to  be  explored  about 
the  relationship  between  adolescent  stress 
and  alcohol  use.  Such  investigations  may 
anticipate  intriguing  complexities.  For 
instance,  lower  cardiovascular  stress 
responses  were  correlated  with  high-risk 
behavior  in  adolescent  boys  (Liang  et  al. 
1995),  and  low  heart  rate  reactivity  to 
alcohol  was  associated  with  familial  risk 
for  developing  alcoholism  (Peterson  et 
al.  1993).  King  and  colleagues  (1990) 
reported  that  abstinent  adult  drug  abusers 
exhibited  lower  basal  levels  of  Cortisol  than 
control  subjects,  with  impulsivity  being 
inversely  correlated  with  basal  Cortisol  lev- 
els in  the  control  group.  It  is  unclear  how 
these  findings  will  ultimately  relate  to 
evidence  discussed  previously  that 
enhanced  physiological  responses  to  stress 
may  exacerbate  drug-seeking  behavior. 

THE  EARLY  EXPOSURE 
EFFECT:  EARLY  ALCOHOL 
USE  AS  A  PREDICTOR 
OF  LATER  ABUSE 

Early  onset  of  alcohol  use  has  been  shown 
in  both  prospective  and  retrospective 


studies  to  be  a  powerful  predictor  of 
later  alcohol  abuse  and  dependence 
(Rachal  et  al.  1982;  Friedman  and 
Humphrey  1985;  Deykin  et  al.  1987; 
Fergusson  et  al.  1994;  Grant  and  Dawson 
1997;  Hawkins  et  al.  1997).  In  a  study 
of  27,616  current  and  former  drinkers 
interviewed  for  the  1992  National 
Longitudinal  Alcohol  Epidemiologic 
Survey,  the  rate  of  lifetime  alcohol 
dependence  was  found  to  be  40  percent 
when  individuals  started  drinking  at  or 
before  14  years  of  age,  but  only  10 
percent  when  drinking  was  not  initiated 
until  20  years  or  later  (Grant  and  Dawson 
1997).  Overall,  with  each  year  of  delay  in 
onset  of  alcohol  use,  the  odds  of  depen- 
dence decreased  by  14  percent  and  the 
odds  of  abuse  decreased  by  8  percent. 
Effects  of  early  ethanol  experience  were 
evident  with  exposures  occurring  as 
early  as  6  years  of  age  (Fergusson  et 
al.  1994). 

This  early  exposure  effect  has  been 
suggested  to  be  one  of  the  strongest 
predictors  of  subsequent  alcohol  abuse 
(Robins  and  Przybeck  1985;  Barnes 
and  Welte  1986;  Hawkins  et  al.  1997) 
and  is  seen  in  relation  to  other  drugs  as 
well.  Early  alcohol  exposure  is  correlated 
with  increased  later  use  and  abuse  of 
other  drugs  (Yamaguchi  and  Kandel 
1984;  Robins  and  Przybeck  1985; 
Deykin  et  al.  1987;  Robins  and  McEvoy 
1990),  and  early  exposure  to  illicit 
drugs  is  associated  with  increased  later 
abuse  of  alcohol  (Robins  and  Przybeck 
1985)  as  well  as  other  drugs  of  abuse 
(Yamaguchi  and  Kandel  1984;  Robins 
and  Przybeck  1985;  Kandel  and 
Davies  1992). 

There  are  at  least  two  possible  expla- 
nations of  this  powerful  effect.  First, 


323 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


traits  associated  with  early  alcohol  use 
might  independently  predict  later 
problem  use,  regardless  of  prior  drug 
history;  according  to  this  view,  early 
alcohol  use  serves  as  a  marker,  not  a 
precursor,  of  a  later  abuse  disorder.  For 
instance,  high  novelty  seeking  in  pre- 
teens  was  predictive  of  alcohol  abuse 
at  27  years  of  age  (Cloninger  et  al. 
1988);  high  novelty  seeking  is  one  of  a 
number  of  traits  that  seem  to  facilitate 
initiation  of  alcohol  and  other  drug 
use  (Baumrind  1987).  Second,  early 
exposure  to  alcohol  may  alter  develop- 
mental processes  during  adolescence, 
with  long-term  effects  on  neurobehav- 
ioral  function  that  increase  the  propen- 
sity for  later  abuse.  Indirect  support 
for  this  possibility  was  obtained  via 
path  analysis  of  data  from  10-  to  11- 
year-old  children  collected  prospec- 
tively for  7  years;  in  this  study,  effects 
of  all  significant  risk  factors  for  alcohol 
misuse  were  found  to  be  mediated 
through  age  of  alcohol  initiation,  other 
than  a  modest  independent  influence 
of  gender  (Hawkins  et  al.  1997). 

Animal  models  could  help  determine 
whether  there  is  a  causal  relationship 
between  early  exposure  and  later  alcohol- 
related  problems,  and  could  help  in 
exploring  the  mechanisms  underlying 
this  association.  Although  chronic 
exposure  of  adolescent  rats  to  ethanol 
has  been  reported  to  induce  long-last- 
ing alterations  in  cognitive  function- 
ing (Osborne  and  Butler  1983)  and 
to  disrupt  puberty-associated  increases 
in  reproductive  endocrinology  in  both 
males  (Cicero  et  al.  1990)  and  females 
(Dees  et  al.  1990),  no  functional  animal 
models  of  the  early  exposure  effect  per 
se  have  yet  been  developed.  Although 


there  are  reports  that  pre-  (Hayashi 
and  Tadokoro  1985)  or  postweaning 
(Ho  et  al.  1989)  exposure  to  ethanol 
can  increase  later  ethanol  preference, 
several  groups  have  reported  no  increase 
in  later  consumption  following  periods 
of  ethanol  exposure  that  include  ado- 
lescence (Kakihana  and  McClearn  1963; 
Parisella  and  Pritham  1964;  Tolliver 
and  Samson  1991).  In  the  develop- 
ment of  animal  models  of  the  early 
exposure  effect,  it  may  prove  useful  to 
consider  the  intriguing  suggestion  of 
Tolliver  and  Samson  (1991)  that 
stress  may  serve  to  unmask  effects  of 
early  exposure  on  later  intake. 

SUMMARY  AND 
RECOMMENDATIONS 

Study  of  the  relationship  between  alco- 
hol use  and  adolescence  is  still  in  its 
infancy,  despite  the  frequent  initiation 
of  alcohol  use  by  adolescents  and  the 
implications  that  this  early  use  has  for 
later  problem  use.  A  number  of  impor- 
tant goals  for  future  research  in  this 
critical  but  underinvestigated  area  are 
suggested  in  the  following  paragraphs. 
Although  studies  in  laboratory 
animals  have  shown  that  adolescents 
are  relatively  resistant  to  the  motor- 
impairing  and  sedative  effects  of  alco- 
hol, they  conversely  appear  to  be 
more  sensitive  to  ethanol-induced  dis- 
ruptions in  hippocampally-related  spa- 
tial memory.  Further  research  is  needed 
to  specify  the  circumstances  under 
which  ethanol  responsivity  is  exacer- 
bated or  attenuated  in  adolescents,  and 
to  determine  the  neural  mechanisms 
underlying  these  differential  effects.  This 
work  should  consider  the  ontogeny  of 


324 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


ethanol  tolerance  and  sensitization, 
given  that  reported  differences  in  the 
ontogeny  of  within-  and  between- 
session  tolerances  could  contribute  to 
the  insensitivity  to  ethanol  often 
observed  in  adolescents,  and  could 
potentially  contribute  to  greater  levels 
of  use  later  in  life. 

We  also  need  to  know  more  about 
adolescence  and  the  ontogeny  of  stress 
responsivity,  particularly  given  clinical 
evidence  that  stress  is  more  strongly 
associated  with  alcohol  consumption 
in  adolescents  than  in  adults.  In  addi- 
tion to  elucidating  the  relationship 
among  hormonal,  behavioral,  and 
neural  responses  to  stressors  during 
ontogeny,  stress  effects  on  alcohol 
self-administration  should  also  be 
considered  within  a  developmental 
timeframe.  Conclusions  about  the 
relationship  between  stress  and  adults' 
self-administration  of  alcohol  and 
other  drugs  may  bear  little  resem- 
blance to  the  relationship  between 
these  variables  in  adolescence,  the  age 
period  when  most  alcohol  and  drug 
use  is  initiated. 

Additional  research  is  needed  to 
examine  normal  brain  function  in 
adolescence.  The  limited  work  to 
date  has  revealed  various  neural  alter- 
ations during  adolescence,  but  these 
rather  piecemeal  observations  remain 
to  be  confirmed  and  more  completely 
characterized  and  integrated.  What 
are  the  functional  implications  of 
potential  alterations  in  the  balance 
among  different  forebrain  DA  regions 
during  adolescence?  How  widespread 
are  adolescent-associated  changes 
among  other  neural  systems  and 
brain  regions? 


Despite  evidence  for  alterations 
during  adolescence  in  forebrain 
regions  (such  as  the  PFC  and  meso- 
corticolimbic  DA  systems)  modulat- 
ing the  reward  efficacy  of  reinforcing 
drugs,  little  is  yet  known  regarding 
the  reinforcing  efficacy  of  alcohol  and 
other  reinforcers  during  adolescence. 
Such  knowledge  is  obviously  critical. 

Work  is  also  needed  to  determine 
the  factors  that  trigger  these  critical 
developmental  changes  in  brain  func- 
tion as  well  as  in  ethanol  sensitivity 
and  responsivity  to  stressors  during 
adolescence.  Do  puberty-associated 
increases  in  gonadal  hormones  or 
adrenal  hormones  play  significant  acti- 
vational  roles? 

It  is  critical  that  future  efforts  also  be 
directed  to  the  question  of  why  early 
exposure  to  alcohol  is  seemingly  so  much 
more  dangerous  than  later  use,  and  to 
determine  whether  early  exposure  indeed 
increases  the  propensity  for  later  alcohol 
problems.  This  issue  is  highly  relevant 
for  prevention  efforts,  with  numerous 
clinicians  suggesting  that  prevention 
be  directed  toward  "just  say  later"  efforts 
of  postponing  first  use  (see,  e.g.,  Robins 
and  McEvoy  1990). 

Multiple  research  approaches  will 
be  needed  to  explore  the  neurobehav- 
ioral  and  environmental  antecedents 
influencing  alcohol  sensitivity  and 
alcohol  use  during  adolescence,  as  well 
as  lasting  consequences  of  this  use. 
While  some  aspects  of  adolescence  can 
be  properly  and  productively  modeled 
in  laboratory  animals,  others  clearly  can- 
not and  will  require  studies  in  human 
adolescents.  Rodent  studies  can  be 
used  to  rapidly  and  cost-effectively 
characterize  neural,  hormonal,  and 


325 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


behavioral  features  of  adolescence  as 
well  as  the  interrelationships  among 
these  factors,  environmental  stressors, 
and  their  association  with  alcohol  use 
and  later  abuse.  For  some  research 
questions,  use  of  nonhuman  primates 
may  prove  preferable.  The  protracted 
ontogeny  generally  evident  in  primates 
relative  to  other  laboratory  animals 
might  potentially  prove  beneficial  for 
long-term  studies  of  the  pharmacology 
of  adolescent  drug  self- administration, 
although  more  needs  to  be  understood 
regarding  the  time  course  of  adolescence 
in  nonhuman  primates — particularly 
that  of  seasonal  breeders  where  pubertal 
changes  unfold  in  a  compressed  time- 
frame temporally  synchronized  with 
the  mating  season  (see,  e.g.,  Coe  et  al. 
1981;  Plant  1996).  In  all  of  this  work  it 
will  be  critical  for  researchers  to  remem- 
ber that  adolescents  cannot  be  treated 
just  as  immature  adults.  The  distinctive 
characteristics  and  proclivities  of  adoles- 
cents must  be  considered  when  devel- 
oping appropriate  animal  models, 
approaches,  and  techniques  to  study 
this  unique  developmental  stage. 

ACKNOWLEDGMENT 

Preparation  of  this  manuscript  was  sup- 
ported in  part  by  National  Institute  on 
Alcohol  Abuse  and  Alcoholism  grant 
R01  AA10288. 


REFERENCES 

Andersen,  S.L.;  Dumont,  N.L.;  and 
Teicher,  M.H.  Developmental  differences 
in  dopamine  synthesis  inhibition  by  (±)-7- 
OH-DPAT.  Naunyn  Schmiedeberjjs  Arch 
Pharmacol  356:173-181,  1997a. 


Andersen,  S.L.;  Rutstein,  M.;  Benzo, 
J.M.;  Hostetter,  J.C.;  and  Teicher,  M.H. 
Sex  differences  in  dopamine  receptor 
overproduction  and  elimination. 
Neuroreport  8:1495-1498,  19976. 

Arnett,  J.  Reckless  behavior  in  adolescence: 
A  developmental  perspective.  Dev  Rev 
12:339-373,  1992. 

Arnt,  J.  Differential  behavioural  effects  of 
dopamine  agonists  in  developing  rats:  A 
study  of  3-PPP  enantiomers.  Eur  J 
Pharmacol  91:273-278,  1983. 

Baer,  P.E.;  Garmezy,  L.B.;  McLaughlin, 
RJ.;  Pokorny,  A.D.;  and  Wernick,  MJ. 
Stress,  coping,  family  conflict,  and  adoles- 
cent alcohol  use.  J Behav  Med  10:449- 
466,  1987. 

Bailey,  C.C.,  and  Kitchen,  I.  Developmental 
responses  to  opioids  reveals  a  lack  of 
effect  on  stress-induced  corticosterone 
levels  in  neonatal  rats.  Br  J  Pharmacol 
91:119-125,1987. 

Barnes,  G.M.,  and  Welte,  J.W.  Patterns 
and  predictors  of  alcohol  use  among  7th- 
12th  grade  students  in  New  York  State.  / 
Stud  Alcohol  47 :53-62, 1986. 

Bates,  M.E.,  and  Labouvie,  E.W.  Adolescent 
risk  factors  and  the  prediction  of  persis- 
tent alcohol  and  drug  use  into  adulthood. 
Alcohol  Clin  Exp  Res  21:944-950, 1997. 

Bauer,  R.H.  Shuttiebox  avoidance  in 
Norway  rats  from  infancy  to  maturity. 
Bull  Psychonomic  Soc  15:15-17,  1980. 

Baumrind,  D.  A  developmental  perspective 
on  adolescent  risk  taking  in  contemporary 
America.  In:  Irwin,  C.E.,  Ir.,  ed.  Adolescent 
Social  Behavior  and  Health.  San  Francisco: 
lossey-Bass,  1987.  pp.  93-125. 

Beatty,  W.W.,  and  Fessler,  RG.  Gonad- 
ectomy  and  sensitivity  to  electric  shock  in 
the  rat.  Physiol  Behav  19:1-6,  1977. 


326 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


Benes,  F.M.  Myelination  of  cortical  - 
hippocampal  relays  during  late  adolescence. 
Schizophr  Bull  15:585-593,  1989. 

Bowers,  W.J.;  Sabongui,  A.G.;  and  Amit, 
Z.  The  role  of  ethanol  availability  on  stress- 
induced  increases  in  ethanol  consump- 
tion. Alcohol  14:551-556,  1997. 

Brand,  T.,  and  Slob,  A.K.  Peripubertal 
castration  of  male  rats,  adult  open  field 
ambulation  and  partner  preference  behav- 
ior. Behav  Brain  Res  30:111-117,  1988. 

Brooks-Gunn,  J.,  and  Reiter,  E.O.  The 
role  of  pubertal  processes.  In:  Feldman, 
S.S.,  and  Elliott,  G.R.,  eds.  At  the 
Threshold:  The  Developing  Adolescent. 
Cambridge,  MA:  Harvard  University 
Press,  1990.  pp.  16-53. 

Buchanan,  C.M.;  Eccles,  J.S.;  and  Becker, 
J.B.  Are  adolescents  the  victims  of  raging 
hormones:  Evidence  for  activational  effects 
of  hormones  on  moods  and  behavior  at 
adolescence.  Psychol  Bull  111:62-107, 1992. 

Chambers,  R.A.;  Moore,  J.;  McEvoy, 
J.P.;  and  Levin,  E.D.  Cognitive  effects  of 
neonatal  hippocampal  lesions  in  a  rat 
model  of  schizophrenia.  Neuropsycho- 
pharmacology  15:587-594,  1996. 

Choi,  S.,  and  Kellogg,  C.K  Norepinephrine 
utilization  in  the  hypothalamus  of  the 
male  rat  during  adolescent  development. 
Dev  Neurosci  14:369-376,  1992. 

Choi,  S.,  and  Kellogg,  C.K.  Adolescent 
development  influences  functional  respon- 
siveness of  noradrenergic  projections  to 
the  hypothalamus  in  male  rats.  Dev  Brain 
Res  94:144-151,  1996. 

Choi,  S.;  Weisberg,  S.N.;  and  Kellogg,  C.K. 
Control  of  endogenous  norepinephrine 
release  in  the  hypothalamus  of  male  rats 
changes  over  adolescent  development.  Dev 
Brain  Res 98: 134-141,  1997. 


Cicero,  T.J.;  Adams,  M.L.;  O'Connor,  L.; 
Nock,  B.;  Meyer,  E.R;  and  Wozniak,  D. 
Influence  of  chronic  alcohol  administration 
on  representative  indices  of  puberty  and 
sexual  maturation  in  male  rats  and  the 
development  of  their  progeny.  J  Pharmacol 
Exp  Ther  255:707-715,  1990. 

Cirulli,  F.;  Terranova,  M.L.;  and  Laviola, 
G.  Affiliation  in  periadolescent  rats: 
Behavioral  and  corticosterone  response  to 
social  reunion  with  familiar  or  unfamiliar 
partners.  Pharmacol  Biochem  Behav  54:99- 
105,  1996. 

Cloninger,  C.R;  Sigvardsson,  S.;  and 
Bohman,  M.  Childhood  personality  pre- 
dicts alcohol  abuse  in  young  adults.  Alcohol 
Clin  Exp  Res  12:494-505,  1988. 

Coe,  C.L.;  Chen,  J.;  Lowe,  E.L.;  Davidson, 
J.M.;  and  Levine,  S.  Hormonal  and  be- 
havioral changes  at  puberty  in  the  squirrel 
monkey.  Horm  Behav  15:36-53,  1981. 

Coulter,  C.L.;  Happe,  H.K.;  and  Murrin, 
L.C.  Postnatal  development  of  the  dopa- 
mine transporter:  A  quantitative  autoradi- 
ographic study.  Dev  Brain  Res  92:172- 
181,  1996. 

Dees,  W.L.;  Skelley,  C.W.;  Hiney,  J.K;  and 
Johnston,  CA.  Actions  of  ethanol  on  hypo- 
thalamic and  pituitary  hormones  in  prepu- 
bertal female  rats.  Alcohol  7:21-25,  1990. 

Deykin,  E.Y.;  Levy,  J.C.;  and  Wells,  V.  Ado- 
lescent depression,  alcohol  and  drug  abuse. 
Am  J  Public  Health  77:178-182,  1987. 

Diamond,  A.  Guidelines  for  the  study  of 
brain -behavior  relationships  during  devel- 
opment. In:  Levin,  H.S.;  Eisenberg,  H.M.; 
and  Benton,  A.L.,  eds.  Frontal  Lobe  Func- 
tion and  Dysfunction.  New  York:  Oxford 
University  Press,  1991.  pp.  339-378. 

Dubas,  J.S.  Cognitive  abilities  and  physical 
maturation.  In:  Lerner,  R.M.;  Petersen,  A.C.; 
and  Brooks-Gunn,  J.,  eds.  Encyclopedia  of 


327 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Adolescence.  Vol.  I.  New  York:  Garland 
Publishing,  1991.  pp.  133-138. 

Dumas,  T.C.,  and  Foster,  T.C.  Late  devel- 
opmental changes  in  the  ability  of  adeno- 
sine Al  receptors  to  regulate  synaptic 
transmission  in  the  hippocampus.  Dev  Brain 
Res  105:137-139,  1998. 

Dunn,  A.J.,  and  Kramarcy,  N.R.  Neuro- 
chemical responses  in  stress:  Relationships 
between  the  hypothalamic-pituitary-adrenal 
and  catecholamine  systems.  In:  Iversen, 
L.L.;  Iversen,  S.D.;  and  Snyder,  S.H.,  eds. 
Handbook  of  Psycho-pharmacology.  Vol.  18. 
New  York:  Plenum  Publishing,  1984.  pp. 
455-515. 

Ernst,  A.J.;  Dempster,  J. P.;  Yee,  R;  St. 
Dennis,  C;  and  Nakano,  L.  Alcohol 
toxicity,  blood  alcohol  concentration  and 
body  water  in  young  and  adult  rats.  J  Stud 
Alcohol  37:347-356,  1976. 

Fahlke,  C;  Engel,  J.A.;  Eriksson,  C.J. P.; 
Hard,  E.;  and  Soderpalm,  B.  Involvement 
of  corticosterone  in  the  modulation  of 
ethanol  consumption  in  the  rat.  Alcohol 
11:195-202,1994. 

Fassino,  M.J.,  and  Campbell,  BA.  The 
ontogeny  of  play  in  rats.  Paper  presented 
at  the  meeting  of  the  Eastern  Psychological 
Association,  New  York,  NY,  1981. 

Fergusson,  D.M.;  Lynskey,  M.T.;  and 
Horwood,  L.J.  Childhood  exposure  to 
alcohol  and  adolescent  drinking  patterns. 
Addiction  89:1007-1016,  1994. 

Friedman,  J.,  and  Humphrey,  J. A. 
Antecedents  of  collegiate  drinking.  / 
Youth  Adolesc  14:11-21,  1985. 

Gabriel,  S.M.;  Roncancio,  J.R.;  and  Ruiz, 
N.S.  Growth  hormone  pulsatility  and  the 
endocrine  milieu  during  sexual  maturation 
in  male  and  female  rats.  Neuroendocrinolojjy 
56:619-628,  1992. 


Galloway,  M.P.;  Wolf,  M.E.;  and  Roth, 
R.H.  Regulation  of  dopamine  synthesis  in 
the  medial  prefrontal  cortex  is  mediated 
by  release  modulating  autoreceptors: 
Studies  in  vivo.  J  Pharmacol  Exp  Ther 
236:689-698,  1986. 

Ganji,  V.,  and  Betts,  N.  Fat,  cholesterol, 
fiber  and  sodium  intakes  of  US  population: 
Evaluation  of  diets  reported  in  1987-88 
Nationwide  Food  Consumption  Survey. 
Eur  J  Clin  Nutr  49:915-920,  1995. 

Gelbard,  H.A.;  Teicher,  M.H.;  Faedda, 
G.;  and  Baldessarini,  RJ.  Postnatal  devel- 
opment of  dopamine  Dj  and  D2  receptor 
sites  in  rat  striatum.  Brain  Res  49:123- 
130,  1989. 

Goeders,  N.E.  A  neuroendocrine  role  in 
cocaine  reinforcement.  Psychoneuro- 
endocrinology  22:237-259,  1997. 

Goldman,  L.;  Winget,  C;  Hollingshead, 
G.W.;  and  Levin,  S.  Postweaning  develop- 
ment of  negative  feedback  in  the  pituitary- 
adrenal  system  of  the  rat.  Neuroendocrinolojjy 
12:199-211,  1973. 

Goodrick,  C.L.  Alcohol  preference  of  the 
male  Sprague-Dawley  albino  rat  as  a  func- 
tion of  age.  J  Gerontol  22:369-371,  1967. 

Gould,  E.;  Woolf,  N.J.;  and  Butcher,  L.L. 
Postnatal  development  of  cholinergic 
neurons  in  the  rat:  I.  Forebrain.  Brain  Res 
Bull  27:767-789,  1991. 

Grant,  B.F.,  and  Dawson,  DA.  Age  at 
onset  of  alcohol  use  and  its  association  with 
DSM-rV  alcohol  abuse  and  dependence: 
Results  from  the  National  Longitudinal 
Alcohol  Epidemiologic  Survey.  /  Subst 
Abuse  9:103-110, 1997. 

Grieve,  S.J.,  and  Littleton,  J.M.  Age  and 
strain  differences  in  the  rate  of  development 
of  functional  tolerance  to  ethanol  by  mice. 
/ Pharm  Pharmacol  31:696-700,  1979. 


328 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


Hansell,  S.,  and  White,  H.R.  Adolescent 
drug  use,  psychological  distress,  and 
physical  symptoms.  J  Health  Soc  Behav 
32:288-301,  1991. 

Hawkins,  J.D.;  Graham,  J.W.;  Maguin, 
E.;  Abbott,  R.;  Hill,  K.G.;  and  Catalano, 
R.F.  Exploring  the  effects  of  age  of  alco- 
hol use  initiation  and  psychosocial  risk 
factors  on  subsequent  alcohol  misuse.  / 
Stud  Alcohol  58:280-290,  1997. 

Hayashi,  T.,  and  Tadokoro,  S.  Learning 
retardation  and  enhanced  ethanol  prefer- 
ence produced  by  postnatal  pretreatments 
with  ethanol  in  adult  rats.  Jpn  J  Pharmacol 
37:269-276,  1985. 

Hedner,  T.,  and  Lundborg,  P.  Development 
of  dopamine  autoreceptors  in  the  postnatal 
rat  brain.  J  Neural  Transm  62:53-63, 1985. 

Heinrichs,  S.C.;  Cole,  B.J.;  Pich,  E.M.; 
Menzaghi,  F.;  Koob,  G.F.;  and  Hauger, 
R.L.  Endogenous  corticotropin-releasing 
factor  modulates  feeding  induced  by 
neuropeptide  Y  or  a  tail-pinch  stressor. 
Peptides  13:879-884,  1992. 

Ho,  A.;  Chin,  A.J.;  and  Dole,  V.P.  Early 
experience  and  the  consumption  of  alco- 
hol by  adult  C57BL/6J  mice.  Alcohol 
6:511-515,  1989. 

Hollstedt,  C,  and  Rydberg,  U.  Postnatal 
effects  of  alcohol  on  the  developing  rat.  In: 
Rydberg,  U.;  Ailing,  C;  and  Engel,  J.A., 
eds.  Alcohol  and  the  Developing  Brain. 
New  York:  Raven  Press,  1985.  pp.  69-84. 

Hollstedt,  C;  Olsson,  O.;  and  Rydberg,  U. 
Effects  of  ethanol  on  the  developing  rat.  II. 
Coordination  as  measured  by  the  tilting- 
plane  test.  Med  Biol  58:164-168,  1980. 

Huttenlocher,  P.R  Synapse  elimination 
and  plasticity  in  developing  human  cerebral 
cortex.  Am  J  Mental  Deficiency  88:488- 
496,  1984. 


Jernigan,  T.L.;  Trauner,  DA.;  Hesselink, 
J.R.;and  Tallal,  PA.  Maturation  of  human 
cerebrum  observed  in  vivo  during  adoles- 
cence. Brain  114:2037-2049,  1991. 

Kakihana,  R,  and  McClearn,  G.E.  Develop- 
ment of  alcohol  preference  in  BALB/c 
mice.  Nature  199:511-512,  1963. 

Kalsbeek,  A.;  Voorn,  P.;  Buijs,  KM.;  Pool, 
C.W.;  and  Uylings,  H.B.M.  Development 
of  the  dopaminergic  innervation  in  the 
prefrontal  cortex  of  the  rat.  J  Comp  Neurol 
269:58-72,  1988. 

Kandel,  D.B.,  and  Davies,  M.  Progression 
to  regular  marijuana  involvement:  Pheno- 
menology and  risk  factors  for  near-daily 
use.  In:  Glantz,  M.D.,  and  Pickens,  R.W., 
eds.  Vulnerability  to  Drug  Abuse. 
Washington,  DC:  American  Psychological 
Association  Press,  1992.  pp.  211-253. 

Keir,  W.J.,  and  Deitrich,  RA.  Development 
of  central  nervous  system  sensitivity  to 
ethanol  and  pentobarbital  in  short-  and 
long-sleep  mice.  /  Pharmacol  Exp  Ther 
254:831-835,  1990. 

Kellogg,  C.K.;  Awatramani,  G.B.;  and 
Piekut,  D.T.  Adolescent  development  alters 
stressor-induced  Fos  immunoreactivity  in 
rat  brain.  Neuroscience  83:681-689,  1998. 

Kennedy,  G.C.,  and  Mitra,  J.  Body 
weight  and  food  intake  as  initiating  fac- 
tors for  puberty  in  the  rat.  /  Physiol 
166:408-418,  1963. 

King,  R.J.;  Jones,  J.;  Scheuer,  J.W.;  Curtis, 
D;  and  Zarcone,  V.P.  Plasma  Cortisol  corre- 
lates of  impulsivity  and  substance  abuse.  Pers 
Individual  Differences  11:287-291,  1990. 

Koob,  G.F.  Neural  mechanisms  of  drug 
reinforcement.  Ann  NT  Acad  Sci  654:171- 
191,  1992. 

Kostovic,  I.  Structural  and  histochemical 
reorganization  of  the  human  prefrontal 


329 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


cortex  during  perinatal  and  postnatal  life. 
In:  Uylings,  H.B.M.;  van  Eden,  C.G.;  De 
Bruin,  J.P.C.;  Corner,  M.A.;  and 
Feenstra,  M.G.P.,  eds.  Progress  in  Brain 
Research,  Vol.  85:  The  Prefrontal  Cortex: 
Its  Structure,  Function  and  Pathology. 
Amsterdam:  Elsevier  Science,  1990.  pp. 
223-240. 

Kurtz,  M.M.,  and  Campbell,  BA.  Para- 
doxical autonomic  responses  to  aversive 
stimuli  in  the  developing  rat.  Behav  Neurosci 
108:962-971,  1994. 

Lancaster,  F.E.;  Brown,  T.D.;  Coker,  K.L.; 
Elliott,  J.A.;  and  Wren,  S.B.  Sex  differences 
in  alcohol  preference  and  drinking  patterns 
emerge  during  the  early  postpuberal  period 
in  Sprague-Dawley  rats.  Alcohol  Clin  Exp 
Res  20:1043-1049,  1996. 

Levin,  H.S.;  Culhane,  K.A.;  Hartmann,  J.; 
Evankovich,  K.;  Mattson,  A.J.;  Harward, 
H.;  Ringholz,  G.;  Ewing-Cobbs,  L.;  and 
Fletcher,  J.M.  Developmental  changes  in 
performance  on  tests  of  purported  frontal 
lobe  functioning.  Dev  Neuropsychol  7:377- 
395,  1991. 

Lewis,  DA.  Development  of  the  prefrontal 
cortex  during  adolescence:  Insights  into 
vulnerable  neural  circuits  in  schizophrenia. 
Neuropsychopharmacology  16:385-398, 1997. 

Liang,  S.-W.;  Jemerin,  J.M.;  Tschann, 
J.M.;  Irwin,  C.E.,  Jr.;  Wara,  D.W.;  and 
Boyce,  W.T.  Life  events,  cardiovascular 
reactivity,  and  risk  behavior  in  adolescent 
boys.  Pediatrics 96:1 101-1 105,  1995. 

Little,  P.J.;  Kuhn,  CM.;  Wilson,  W.A.;  and 
Swartzwelder,  H.S.  Differential  effects  of 
ethanol  in  adolescent  and  adult  rats.  Alcohol 
Clin  Exp  Res  20: 1346-1 351,  1996. 

Maier,  D.M.,  and  Pohorecky,  LA.  The 
effect  of  stress  on  tolerance  to  ethanol  in 
rats.  Alcohol  Drug  Res  6:387-A0l,  1986. 


Markwiese,  B.J.;  Acheson,  S.K.;  Levin, 
E.D.;  Wilson,  W.A.;  and  Swartzwelder, 
H.S.  Differential  effects  of  ethanol  on 
memory  in  adolescent  and  adult  rats. 
Alcohol  Clin  Exp  Res  22:416-421,  1998. 

Mathias,  R.  Marijuana  and  tobacco  use  up 
again  among  8th  and  10th  graders.  In: 
NIDA  Notes.  NIH  Pub.  No.  97-3478. 
Rockville,  MD:  U.S.  Department  of  Health 
and  Human  Services,  1997.  pp.  12-13. 

Meaney,  M.J.;  Sapolsky,  R.M.;  and  McEwen, 
B.S.  The  development  of  the  glucocorticoid 
receptor  system  in  the  rat  limbic  brain.  I. 
Ontogeny  and  auto  regulation.  Dev  Brain 
Res  18:159-164,  1985a. 

Meaney,  M.J.;  Sapolsky,  R.M.;  and 
McEwen,  B.S.  The  development  of  the 
glucocorticoid  receptor  system  in  the  rat 
limbic  brain.  II.  An  autoradiographic 
study.  Dev  Brain  Res  18:165-168,  1985£. 

Nance,  D.M.  The  developmental  and  neural 
determinants  of  the  effects  of  estrogen  on 
feeding  behavior  in  the  rat:  A  theoretical 
perspective.  Neurosci  Biobehav  Rev  7:189- 
211,1983. 

Odell,  W.D.  Sexual  maturation  in  the  rat. 
In:  Grumbach,  M.M.;  Sizonenko,  P.C.;  and 
Aubert,  M.L.,  eds.  Control  of  the  Onset  of 
Puberty.  Baltimore,  MD:  Williams  & 
Wilkins,  1990.  pp.  183-210. 

Ojeda,  S.R.,  and  Urbanski,  H.F.  Puberty 
in  the  rat.  In:  Knobil,  E.,  and  Neill,  J.D., 
eds.  The  Physiology  of  Reproduction.  2d  ed. 
New  York:  Raven  Press,  1994.  pp.  363^09. 

Osborne,  G.L.,  and  Butler,  A.C.  Enduring 
effects  of  periadolescent  alcohol  exposure 
on  passive  avoidance  performance  in  rats. 
Physiol  Psychol  11:205-208,  1983. 

Parisella,  KM,  and  Pritham,  G.H.  Effect  of 
age  on  alcohol  preference  by  rats.  QJ  Stud 
Alcohol  Parts  A  &  B  25:248-252,  1964. 


330 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


Peterson,  J.B.;  Pihl,  R.O.;  Seguin,  J.R., 
Finn,  P.R.;  and  Stewart,  S.H.  Heart-rate 
reactivity  and  alcohol  consumption  among 
sons  of  male  alcoholics  and  sons  of  non- 
alcoholics.  J  Psychiatry  Neurosci  18:190- 
198, 1993. 

Piazza,  P.V.;  Rouge-Pont,  F.;  Deminiere, 
J.M.;  Kharoubi,  M.;  LeMoal,  M.;  and 
Simon,  H.  Dopaminergic  activity  is  re- 
duced in  the  prefrontal  cortex  and  increased 
in  the  nucleus  accumbens  of  rats  predisposed 
to  develop  amphetamine  self- administration. 
Brain  Res  567:169-174,  1991. 

Plant,  T.M.  Environmental  factors  and 
puberty  in  non-human  primates.  Acta 
Paediatr  Suppl  417 ':89-91,  1996. 

Pohorecky,  LA.  Interaction  of  ethanol 
and  stress:  Research  with  experimental 
animals — an  update.  Alcohol  Alcohol 
25:263-276,  1990. 

Pohorecky,  LA.  Stress  and  alcohol  inter- 
action: An  update  of  human  research. 
Alcohol  Clin  Exp  Res  15:438-459,  1991. 

Post,  G.B.,  and  Kemper,  H.C.G.  Nutrient 
intake  and  biological  maturation  during 
adolescence.  The  Amsterdam  growth  and 
health  longitudinal  study.  Eur  J  Clin 
Nutr47A00-A08,  1993. 

Primus,  R.J.,  and  Kellogg,  C.K.  Pubertal- 
related  changes  influence  the  development 
of  environment-related  social  interaction 
in  the  male  rat.  Dev  Psychobiol  22:633- 
643,  1989. 

Primus,  R.J.,  and  Kellogg,  C.K.  Gonadal 
status  and  pubertal  age  influence  the 
responsiveness  of  the  benzodiazepine/ 
GABA  receptor  complex  to  environmen- 
tal challenge  in  male  rats.  Brain  Res 
561:299-306,1991. 

Rachal,  J.V.;  Guess,  L.L.;  Hubbard,  R.L.; 
and  Maisto,  SA.  Alcohol  misuse  by 


adolescents.  Alcohol  Health  Res  World 
6:61-68,  1982. 

Ramaley,  J. A.  Changes  in  daily  serum 
corticosterone  values  in  maturing  male 
and  female  rats.  Steroids 20:185-197, 1972. 

Ramaley,  J.A.,  and  Olson,  J.  Adrenal 
function  in  rats  given  PMS  before  puberty: 
Response  to  ether  stress.  Neuroendo- 
crinology  14:1-13,  1974. 

Rivier,  C.  Effect  of  the  age  of  the  rat  and 
the  duration  of  the  stimulus  on  stress- 
induced  ACTH  secretion.  In  Weiner,  H.; 
Florin,  I.;  Murison,  R;  and  Hellhammer, 
D.,  eds.  Frontiers  of  Stress  Research. 
Toronto:  Hans  Huber  Publishers,  1989. 
pp. 223-232. 

Robins,  L.N.,  and  McEvoy,  L.  Conduct 
problems  as  predictors  of  substance  abuse. 
In:  Robins,  L.N.,  and  Rutter,  M.,  eds. 
Straight  and  Devious  Pathways  from 
Childhood  to  Advdthood.  New  York:  Oxford 
University  Press,  1990.  pp.  182-204. 

Robins,  L.N.,  and  Przybeck,  T.R.  Age  of 
onset  of  drug  use  as  a  factor  in  drug  and 
other  disorders.  In:  Jones,  C.L.,  and 
Battjes,  R.,  eds.  Etiology  of  Drug  Abuse: 
Implications  for  Prevention.  Research 
Monograph  Series,  No.  56.  Rockville, 
MD:  National  Institute  on  Drug  Abuse, 
1985. pp.  178-192. 

Rosenberg,  D.R.,  and  Lewis,  DA. 
Changes  in  the  dopaminergic  innervation 
of  monkey  prefrontal  cortex  during  late 
postnatal  development:  A  tyrosine  hy- 
droxylase immunohistochemical  study. 
Biol  Psychiatry  36:272-277,  1994. 

Sapolsky,  R.M.;  Meaney,  M.J.;  and 
McEwen,  B.S.  The  development  of  the 
glucocorticoid  receptor  system  in  the  rat 
limbic  brain.  III.  Negative-feedback  regu- 
lation. Dev  Brain  Res  18:169-173,  1985. 


331 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Seeman,  P.;  Bzowej,  N.H.;  Guan,  FLO; 
Bergeron,  C;  Becker,  L.E.;  Reynolds, 
G.P.;  Bird,  E.D.;  Riederer,  P.;  Jellinger, 
K.;  Watanabe,  S.;  and  Tourtellotte,  W.W. 
Human  brain  dopamine  receptors  in  chil- 
dren and  aging  adults.  Synapse  1:399- 
404,  1987. 

Shalaby,  I. A.,  and  Spear,  L.P.  Psycho- 
pharmacological  effects  of  low  and  high 
doses  of  apomorphine  during  ontogeny. 
Eur  J  Pharmacol  67 :451^59,  1980. 

Shalaby,  I  .A.;  Dendel,  P.S.;  and  Spear, 
L.P.  Differential  functional  ontogeny  of 
dopamine  presynaptic  receptor  regulation. 
Dev  Brain  Res  1:434^39,  1981. 

Silveri,  M.M.,  and  Spear,  L.P.  Decreased 
sensitivity  to  the  hypnotic  effects  of 
ethanol  early  in  ontogeny.  Alcohol  Clin 
Exp  Res  22:670-676,  1998. 

Silveri,  M.M.,  and  Spear,  L.P.  Ontogeny 
of  rapid  tolerance  to  the  hypnotic  effects 
of  ethanol.  Alcohol  Clin  Exp  Res  23: 1180- 
1184,1999. 

Silveri,  M.M.,  and  Spear,  L.P.  Ontogeny 
of  ethanol  elimination  and  ethanol-induced 
hypothermia.  Alcohol  20(l)A5-53,  2000. 

Spear,  L.P.,  and  Brake,  S.C.  Periadolescence: 
Age -dependent  behavior  and  psychophar- 
macological  responsivity  in  rats.  Dev 
Psychobiol  16:83-109,  1983. 

Spear,  L.P.;  Shalaby,  LA.;  and  Brick,  J. 
Chronic  administration  of  haloperidol 
during  development:  Behavioral  and  psycho- 
pharmacological  effects.  Psychopharmacology 
70:47-58,  1980. 

Spiers,  D.E.,  and  Fusco,  L.E.  Age-depen- 
dent differences  in  the  thermoregulatory 
response  of  the  immature  rat  to  ethanol. 
Alcohol  Clin  Exp  Res  15:23-28,  1991. 

Stahle,  L.  Do  autoreceptors  mediate 
dopamine  agonist-induced  yawning  and 


suppression  of  exploration?  A  critical 
review.  Psychopharmacology  106:1-13, 1992. 

Stanwood,  G.D.;  McElligot,  S.;  Lu,  L.; 
and  McGonigle,  P.  Ontogeny  of  dopamine 
D3  receptors  in  the  nucleus  accumbens  of 
the  rat.  Neurosci  Lett 223:13-16,  1997. 

Stone,  E.A.,  and  Quartermain,  D.  Greater 
behavioral  effects  of  stress  in  immature  as 
compared  to  mature  male  mice.  Physiol 
fl*W  63:143-145,  1998. 

Swartzwelder,  H.S.;  Wilson,  W.A.;  and 
Tayyeb,  M.I.  Age-dependent  inhibition  of 
long-term  potentiation  by  ethanol  in 
immature  versus  mature  hippocampus. 
Alcohol  Clin  Exp  Res  19:1480-1485, 1995a. 

Swartzwelder,  H.S.;  Wilson,  W.A.;  and 
Tayyeb,  M.I.  Differential  sensitivity  of 
NMDA  receptor- mediated  synaptic  po- 
tentials to  ethanol  in  immature  versus 
mature  hippocampus.  Alcohol  Clin  Exp 
Res  19:320-323,  19956. 

Swartzwelder,  H.S.;  Richardson,  R.C.; 
Markwiese-Foerch,  B.;  Wilson,  W.A.;  and 
Little,  P.J.  Developmental  differences  in 
the  acquisition  of  tolerance  to  ethanol. 
Alcohol  15:1-4,  1998. 

Teicher,  M.H.;  Gallitano,  A.L.;  Gelbard, 
H.A.;  Evans,  H.K.;  Marsh,  E.R.;  Booth, 
R.G.;  and  Baldessarini,  R.J.  Dopamine  D1 
autoreceptor  function:  Possible  expression 
in  developing  rat  prefrontal  cortex  and 
striatum.  Dev  Brain  Res  63:229-235,  1991. 

Teicher,  M.H.;  Barber,  N.I.;  Gelbard, 
H.A.;  Gallitano,  A.L.;  Campbell,  A.; 
Marsh,  E.;  and  Baldessarini,  RJ. 
Developmental  differences  in  acute  ni- 
grostriatal  and  mesocorticolimbic  system 
response  to  haloperidol.  Neuropsycho- 
pharmacology  9:147-156,  1993. 

Teicher,  M.H.;  Andersen,  S.L.;  and 
Hostetter,  J.C.,  Jr.  Evidence  for  dopamine 
receptor  pruning  between  adolescence 


332 


Adolescence:  Biological  Basis  of  Vulnerability  to  Alcohol  Abuse 


and  adulthood  in  striatum  but  not  nu- 
cleus accumbens.  Dev  Brain  Res 
89:167-172,  1995. 

Tolliver,  GA.,  and  Samson,  H.H.  The 
influence  of  early  postweaning  ethanol 
exposure  on  oral  self- administration  be- 
havior in  the  rat.  Pharmacol  Biochem 
Baton;  38:575-580,  1991. 

Tschann,  J.M.;  Adler,  N.E.;  Irwin,  C.E., 
Jr.;  Millstein,  S.G.;  Turner,  R.A.;  and 
Kegeles,  S.M.  Initiation  of  substance  use 
in  early  adolescence:  The  roles  of  pubertal 
timing  and  emotional  distress.  Health 
Psychol  12:326-333,  1994. 

van  Eden,  C.G.;  Kros,  J.M.;  and  Uylings, 
H.B.M.  The  development  of  the  rat  pre- 
frontal cortex:  Its  size  and  development 
of  connections  with  thalamus,  spinal  cord 
and  other  cortical  areas.  In:  Uylings,  H.B.M.; 
van  Eden,  C.G.;  De  Bruin,  J.P.C.;  Corner, 
M.A.;  and  Feenstra,  M.G.P.,  eds.  Progress 
in  Brain  Research,  Vol.  85:  The  Prefrontal 
Cortex:  Its  Structure,  Function  and 
Pathology.  Amsterdam:  Elsevier  Science, 
1990.  pp.  169-183. 

Van  Hartesveldt,  C.;  Meyer,  M.E.;  and 
Potter,  T.J.  Ontogeny  of  biphasic  loco- 
motor effects  of  quinpirole.  Pharmacol 
Biochem  Behav  48:781-786,  1994. 

Wagner,  E.F.  Delay  of  gratification,  coping 
with  stress,  and  substance  use  in  adolescence. 
Exp  Clin  Psychopharmacol  1:27-4:3,  1993. 

Walker,  C.-D.;  Perrin,  M.;  Vale,  W.;  and 
Rivier,  C.  Ontogeny  of  the  stress  response 
in  the  rat:  Role  of  the  pituitary  and  the 
hypothalamus.  Endocrinology  118:1445- 
1451,1986. 


Walker,  C.-D.;  Trottier,  G.;  Rochford,  J.; 
and  Lavallee,  D.  Dissociation  between 
behavioral  and  hormonal  responses  to  the 
forced  swim  stress  in  lactating  rats.  J 
Neuroendocrinal  7 -.615-622,  1995. 

Walker,  E.F.,  and  Diforio,  D. 
Schizophrenia:  A  neural  diathesis-stress 
model.  Psychol  Rev  104:667-685,  1997. 

Wiedenmayer,  C.  Causation  of  the  onto- 
genetic development  of  stereotypic  digging 
in  gerbils.  Animal  Behav  53:461^170, 1997. 

Wills,  TA.  Stress  and  coping  in  early 
adolescence:  Relationships  to  substance 
use  in  urban  school  samples.  Health 
Psychol  5:503-529,  1986. 

Wolfer,  D.P.,  and  Lipp,  H.-P.  Evidence 
for  physiological  growth  of  hippocampal 
mossy  fiber  collaterals  in  the  guinea  pig 
during  puberty  and  adulthood. 
Hippocampus  5:329-340,  1995. 

Yamaguchi,  K.,  and  Kandel,  D.B.  Patterns 
of  drug  use  from  adolescence  to  young 
adulthood:  III.  Predictors  of  progression. 
Am  J  Public  Health  74:673-681,  1984. 

York,  J.L.,  and  Chan,  A.W.K.  Age-related 
differences  in  sensitivity  to  alcohol  in  the 
rat.  Alcohol  Clin  Exp  Res  17:864-869, 1993. 

Zecevic,  N.;  Bourgeois,  J. -P.;  and  Rakic,  P. 
Changes  in  synaptic  density  in  motor  cortex 
of  rhesus  monkey  during  fetal  and  postna- 
tal life.  Dev  Brain  Res  50:1 1-32,  1989. 

Zorzano,  A.,  and  Herrera,  E.  Decreased 
in  vivo  rate  of  ethanol  metabolism  in  the 
suckling  rat.  Alcohol  Clin  Exp  Res 
13:527-532,  1989. 


333 


STUDIES  OF  ACUTE  AND 
CHRONIC  EFFECTS  OF 
ALCOHOL  IN  HUMANS 


Chapter  9 

Acute  Effects  of  Alcohol  on  Cognition 
and  Impulsive-Disinhibited  Behavior 

Peter  R.  Finn,  Ph.D. 

KEY  WORDS:  cognition;  disinhibition;  CNS  (central  nervous  system)  information  pro- 
cessing; acute  AODE  (effects  ofAOD  [alcohol  or  other  drug]  use,  abuse,  and  dependence); 
self  control;  memory;  learning;  attention;  AOD  impairment;  biological  inhibition;  psy- 
chomotor impairment;  risk-taking  behavior;  etiology;  psychoactive  substances;  aggressive 
behavior;  literature  review 


The  majority  of  studies  of  the  acute 
effects  of  alcohol  on  cognition  and 
behavior  have  been  conducted  with 
the  general  purpose  of  understanding 
the  short-term  adverse  consequences 
of  alcohol  on  aspects  of  information 
processing  or  on  factors  that  influ- 
ence the  expression  of  aggressive  or 
risky  behavior.  Such  studies  address 
larger  societal  concerns,  such  as  how 
alcohol  impairs  driving,  work  perfor- 
mance, and  cognitive  functioning;  the 
long-term  and  potentially  permanent 
effects  of  alcohol  on  cognitive  function 
(e.g.,  Korsakoff's  syndrome);  the  asso- 
ciation between  alcohol  consumption 
and  increased  physical  and  sexual  aggres- 
sion; and,  more  recently,  the  associa- 


tion between  alcohol  consumption 
and  engaging  in  behaviors  that  elevate 
risk  for  contracting  sexually  transmit- 
ted diseases,  especially  AIDS. 

Very  few  studies  have  directly 
investigated  the  relevance  of  the  acute 
cognitive  and  behavioral  effects  of 
alcohol  for  the  etiology  of  alcoholism. 
However,  many  studies  have  investi- 
gated the  potential  relevance  for  the 
etiology  of  alcoholism  of  the  acute 
effects  of  alcohol  on  mood,  subjective 
intoxication,  and  autonomic  or  elec- 
troencephalographic  activity  (e.g., 
Finn  et  al.  1992;  Cohen  et  al.  1993; 
Schuckit  1995).  Lacking  are  studies 
of  the  acute  impairing  effect  of  alco- 
hol on  factors  that  affect  the  self-reg- 


P.R.  Finn,  Ph.D.,  is  director  of  the  Clinical  Science  Training  Program  and  an  associate  professor  in 
the  Department  of  Psychology,  Indiana  University,  1101  East  10th  St.,  Bloomington,  IN  47405-7007. 


337 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ulation  of  drinking  and  studies  of  the 
acute  effect  of  alcohol  on  impulsivity, 
which  in  turn  could  contribute  to 
an  increased  long-term  risk  of  devel- 
oping alcohol  problems.  In  addition, 
there  is  a  general  scarcity  of  studies 
that  attempt  to  control  for  or  assess 
the  effect  of  individual  differences 
in  response  to  alcohol.  As  is  the 
case  for  studies  of  long-term  effects 
of  alcohol  on  cognition  (cf.  Parsons 
and  Nixon  1998),  only  a  few  studies 
have  assessed  the  influence  of  impor- 
tant individual  differences,  such  as  a 
positive  family  history  of  alcoholism 
(e.g.,  Schuckit  et  al.  1988;  Lex  et  al. 
1994),  personality  traits  (e.g., 
Nagoshi  et  al.  1991),  level  and 
history  of  alcohol  consumption  (e.g., 
Salame  1991;  Schandler  et  al. 
1988«),  and  limb  (rising  or  falling) 
of  the  blood  alcohol  curve  (e.g., 
Nicholson  et  al.  1992;  Wang  et 
al.  1992). 

In  this  chapter  I  review  the  key 
findings  of  studies  of  the  acute  effects 
of  alcohol  on  cognition  and  on 
factors  associated  with  impulsive- 
disinhibited  behavior,  identify  impor- 
tant gaps  in  knowledge,  and  suggest 
some  potentially  valuable  avenues  for 
future  research.  The  studies  reviewed 
here  can  be  divided  into  two  areas: 
( 1 )  studies  of  the  effects  of  alcohol 
on  basic  aspects  of  information  pro- 
cessing and  (2)  studies  of  the  effects 
of  alcohol  on  cognitive  function  asso- 
ciated with  poor  impulse  control, 
such  as  risky  decision  making, 
response  bias,  and  cognitive  inhibi- 
tion assessed  with  the  Stroop  test. 
The  review  is  restricted  to  studies 
that  include  some  form  of  alcohol 


administration,  either  experimental  or 
self- administered. 


ACUTE  EFFECTS 
OF  ALCOHOL  ON 
INFORMATION 
PROCESSING 

Studies  documenting  the  presence  of 
serious  cognitive  deficits  in  chronic 
alcoholics  (e.g.,  Fitzhugh  et  al.  1965; 
Kleinknecht  and  Goldstein  1972;  Parsons 
1977)  and  studies  suggesting  that 
impaired  information  processing  is  an 
important  factor  underlying  alcohol's 
effect  on  driving  performance  (e.g., 
Brewer  and  Sandow  1980;  Linnoila  et  al. 
1986)  have  provided  the  basic  impetus 
for  an  extensive  literature  documenting 
the  acute  effects  of  alcohol  on  aspects 
of  information  processing,  including 
speed,  capacity,  attention,  memory,  visual- 
spatial  ability,  problem  solving,  and  learn- 
ing. The  great  majority  of  these  studies 
have  assessed  alcohol's  effect  on  active, 
controlled  (effortful)  processing,  as 
opposed  to  passive,  or  more  automatic 
(preconscious),  processing  of  informa- 
tion (such  as  implicit  memory). 

Most  of  the  studies  of  the  acute 
effects  of  alcohol  on  information  pro- 
cessing have  focused  on  the  effects  of 
alcohol  on  aspects  of  processing  capac- 
ity and  resources,  such  as  processing 
speed,  explicit  memory,  and  attention. 
Overall,  the  literature  indicates  that 
moderate  to  high  doses  of  alcohol 
slow  the  speed  of  information  process- 
ing and  impair  attention  and  memory, 
and  that  such  impairments  are  exacer- 
bated at  higher  alcohol  doses  and  with 
more  complex  tasks  (Rohrbaugh  et  al. 
1987, 1988;  Maylor  and  Rabbitt  1993). 


338 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive-Disinhibited  Behavior 


The  general  interpretation  offered  for 
these  effects  is  that  alcohol  reduces 
the  available  processing  capacity  and 
resources,  contributing  to  a  general 
slowing  of  information  processing 
(Rohrbaugh  et  al.  1987;  Maylor  and 
Rabbitt  1993;  Post  et  al.  1996)  with 
its  concomitant  impairment  in  atten- 
tion and  explicit  memory. 

Processing  Speed 
and  Capacity 

Alcohol's  slowing  effect  on  information 
processing  is  apparent  from  studies  of 
reaction  time  (RT)  (Maylor  and  Rabbitt 
1993),  backward  masking  (Moskowitz 
and  Murray  1976),  self-paced  contin- 
uous performance  tasks  (CPTs)  (e.g., 
Hasenfratz  et  al.  1993),  and  the  latency 
of  event-related  potential  (ERP) 
components  such  as  the  P300  (e.g., 
Rohrbaugh  et  al.  1987).  With  few 
exceptions,  studies  consistently  report 
that  alcohol  increases  RT  on  a  range 
of  choice  RT-type  tasks  (e.g.,  Maylor 
et  al.  1987,  1992;  Hasenfratz  et  al. 
1993;  Post  et  al.  1996).  In  addition, 
impaired  RT  is  greater  on  the  rising 
compared  with  the  falling  limb  of  the 
blood  alcohol  curve  (Beirness  and  Vogel- 
Sprott  1984;  Nicholson  et  al.  1992; 
Wang  et  al.  1992).  Most  of  those 
studies  reporting  no  effect  of  alcohol 
on  RT  have  used  between-subjects 
designs  (Sommer  et  al.  1993)  or  simple 
tasks  (Pfefferbaum  et  al.  1980)  that 
may  not  be  sensitive  enough  to  detect 
alcohol-related  impairments. 

In  their  selective  meta-analysis, 
Maylor  and  Rabbitt  (1993)  presented 
convincing  evidence  of  a  linear  effect 
of  alcohol  on  RT  (RT^^^  1.12RTno 
alcohol  ~  17.85)  accounting  for  99  per- 


cent of  the  variance.  Their  analysis 
revealed  that  the  absolute  effect  of 
alcohol  increased  linearly  with  task 
duration  and  complexity,  or  as  a  func- 
tion of  the  requirements  for  informa- 
tion-processing resources.  Other 
studies  have  also  reported  that  the 
effects  of  alcohol  are  more  pro- 
nounced in  tasks  requiring  more  pro- 
cessing resources,  such  as 
divided-attention  tasks,  tasks  using 
larger  arrays  or  displays  of  stimuli 
(e.g.,  Moskowitz  and  DePry  1968; 
Maylor  et  al.  1990;  Post  et  al.  1996), 
or  vigilance  tasks  with  extended  duration 
(Rorhbaugh  et  al.  1988).  An  analysis 
of  Maylor  and  colleagues'  studies  of 
recognition  memory  (see  Maylor  and 
Rabbitt  1993)  also  suggests  that  alco- 
hol's effect  on  recognition  memory 
follows  a  linear  function  (log  propor- 
tion correct  aiCohoi=  1-31  log  propor- 
tion correct  no  alcohol  -  0.004,  ex- 
plaining 96  percent  of  the  variance), 
with  greater  impairment  on  tasks 
requiring  more  processing  resources. 

Tasks  that  involve  backward  mask- 
ing also  indirectly  demonstrate  the 
cognitive  slowing  effect  of  alcohol. 
Backward  masking  involves  the  pre- 
sentation of  a  masking  stimulus  soon 
after  a  target  stimulus,  where  relatively 
short  interstimulus  intervals  (ISIs) 
between  target  and  masking  stimulus 
impede  the  accurate  detection  of  the 
target.  Alcohol's  cognitive  slowing  effect 
is  inferred  from  the  relatively  longer 
ISIs  required  for  accurately  identifying 
the  target  stimulus  (Moskowitz  and 
Murray  1976;  Moskowitz  et  al.  1985). 
Cognitive  slowing  after  alcohol  is  also 
apparent  on  tasks  that  directly  assess 
speed  of  information  processing,  such 


339 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


as  the  self-paced  rapid  information 
processing  (RIP)  task  (Battig  and 
Buzzi  1986).  The  RIP  task  is  a  com- 
plex CPT  involving  the  consecutive 
presentation  of  individual  numbers  on 
a  screen  and  requiring  a  response  after 
the  detection  of  three  consecutive 
even  or  odd  digits  (see  Fillmore  et  al. 
1998).  Correct  responses  increase  the 
rate  of  presentation  of  stimuli.  False 
alarms  or  misses  result  in  a  slowing  of 
the  stimulus  presentation  rate.  Alco- 
hol results  in  an  overall  slower  rate  of 
stimulus  presentation  due  to  poorer 
performance  at  higher  rates  (Michel 
and  Battig  1989;  Hasenfratz  et  al. 
1993;  Fillmore  et  al.  1998).  Interest- 
ingly, the  slowing  effect  of  alcohol  on 
the  RIP  task  has  been  shown  to  be 
partly  attenuated  by  cigarette  smoking 
(Michel  and  Battig  1989),  caffeine 
(Hasenfratz  et  al.  1993),  and  the 
expectation  that  alcohol  would  mini- 
mally impair  performance  (Fillmore  et 
al.  1998). 

Finally,  results  of  studies  of  the  effect 
of  alcohol  on  the  ERP  provide  some 
indirect  evidence  that  alcohol  slows  the 
speed  of  information  processing  and 
reduces  overall  central  processing  capacity 
(Oscar-Berman  1987;  Jaaskelainen  et 
al.  1996).  The  majority  of  studies  report- 
ing on  the  effects  of  alcohol  on  the 
latency  of  components  of  the  ERP  show 
that  alcohol  increases  latency.  A  number 
of  studies  report  that  alcohol  increases 
the  latency  of  the  P3  ERP  component 
(Teo  and  Ferguson  1986;  Campbell 
and  Lowick  1987;  Krein  et  al.  1987; 
Rohrbaugh  et  al.  1987;  Schuckit  et  al. 
1988;  Lukas  et  al.  1990).  Alcohol  has 
also  been  found  to  increase  the  latencies 
of  the  N2  (Teo  and  Ferguson  1986; 


Rohrbaugh  et  al.  1987;  Jaaskelainen  et 
al.  1995)  and  P2  ERP  components  (Teo 
and  Ferguson  1986).  Reductions  in  P3 
amplitudes  after  alcohol,  which  are 
thought  to  reflect  decreases  in  available 
processing  resources,  have  also  frequently 
been  reported  (Teo  and  Ferguson  1986; 
Campbell  and  Lowick  1987;  Krein  et 
al.  1987;  Rohrbaugh  et  al.  1987). 
Although  alcohol-induced  increases  in 
ERP  latencies  and  reductions  in  P3 
amplitudes  frequently  do  not  parallel 
behavioral  measures  of  alcohol  effects 
(Oscar-Berman  1987),  increased  latencies 
and  reduced  P3s  after  alcohol  have  been 
associated  with  slower  RTs  and  poorer 
signal  detection  performance  (decreased 
sensitivity)  with  more  demanding  tasks 
(Campbell  and  Lowick  1987;  Rohrbaugh 
et  al.  1987).  The  lack  of  a  parallel 
between  alcohol-induced  changes  in  ERP 
and  performance  measures  could  be 
because,  though  alcohol  may  reduce  the 
relative  availability  of  processing 
resources,  some  tasks  do  not  supersede 
the  actual  available  processing  resources 
(e.g.,  working  memory  space). 

What  is  generally  lacking  in  this  liter- 
ature are  studies  designed  to  identify  the 
neurophysiological  basis  (both  struc- 
turally and  at  the  receptor  level)  for  the 
acute  effects  of  alcohol  on  processing 
capacity.  Such  studies  could  use  a  variety 
(combination)  of  brain  imaging  tech- 
niques such  as  functional  magnetic  res- 
onance imaging  (fMRI)  and  positron 
emission  tomography  (PET)  along  with 
specific  cognitive  tasks  (CPT,  recognition 
memory,  choice  RT,  or  word  categori- 
zation) under  alcohol  and  no  alcohol  (and 
placebo)  conditions.  Some  candidate  sys- 
tems suggested  by  Jaaskelainen  and 
colleagues  (1996)  in  their  review  of 


340 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive-Disinhibited  Behavior 


alcohol-ERP  studies  are  the  gamma- 
aminobutyric  acid  (GAB A)  system  (alco- 
hol augmentation)  and  the  N-methyl- 
D-aspartate  (NMDA)  system  (alcohol 
attenuation).  Such  studies  may  reveal 
how  the  acute  effects  of  alcohol  on 
such  systems  may  translate  into  long- 
term  deficits  in  cognitive  functioning. 

Attention 

Studies  have  consistently  indicated  that 
alcohol  impairs  attention  (Koelega 
1995)  and  that  alcohol's  disrupting 
effect  on  attention  is  most  apparent  in 
tasks  involving  divided  or  sustained 
attention  or  high  processing  demands 
(Rohrbaugh  et  al.  1987,  1988;  Maylor 
et  al.  1990;  Lex  et  al.  1994;  Koelega 
1995;  Post  et  al.  1996).  Alcohol  appears 
to  impair  attention  on  divided-attention 
tasks  even  at  very  low  (0.02-0.03  mg%) 
dose  levels  (Koelega  1995).  Rorhbaugh 
and  colleagues  (1987, 1988)  also  found 
that  decrements  in  sustained  attention 
increased  with  higher  doses,  with  a  rapid 
decrement  in  performance  at  high 
doses.  They  suggested  that  alcohol- 
induced  performance  deficits  on  signal 
detection-type  tasks  are  due  to  reduced 
central  processing  capacity  and  reduced 
availability  of  processing  resources  over 
time,  an  interpretation  shared  by  oth- 
ers (Maylor  and  Rabbitt  1993; 
Koelega  1995;  Post  et  al.  1996). 
A  report  of  alcohol's  suppression  of 
mismatch  negativity  of  the  ERP  sug- 
gests that  alcohol  may  also  disrupt 
preconscious  detection  of  stimulus 
events  (Jaaskelainen  et  al.  1995). 

Memory 

Moderate  to  high  doses  of  alcohol 
have  consistently  been  found  to 


impair  both  recognition  and  recall 
memory  (e.g.,  Birnbaum  and  Parker 
1977;  Peterson  et  al.  1990;  Maylor 
and  Rabbitt  1993).  Explanations  for 
the  acute  impairing  effects  of  alcohol 
on  memory  include  impaired  consolida- 
tion and  storage/encoding  (Jones  and 
Jones  1977),  a  slowing  of  the  transfer 
process  from  sensory  storage  to  short- 
term  memory  (Moskowitz  and  Murray 
1977),  a  curtailment  in  deep  (elabo- 
rated) processing  (Craik  1977)  or 
rehearsal  (Landauer  1977),  increased 
forgetting  (Maylor  and  Rabbitt  1987), 
and  a  general  reduction  in  processing 
capacity  (cognitive  slowing)  (Maylor 
and  Rabbitt  1993).  Although  the 
debate  continues,  strong  arguments  can 
be  made  that  the  overall  capacity  for 
processing  information  is  reduced  by 
alcohol,  leading  to  deficits  in  memory 
processes — especially  those  processes 
with  large  demands,  such  as  encoding 
large  stimulus  arrays  or  retaining  items 
in  short-term  memory  sequentially  over 
time  (Maylor  et  al.  1987;  Peterson  et 
al.  1990;  Maylor  and  Rabbitt  1993). 
Although  not  directly  assessed,  these 
data  suggest  that  alcohol  impairs  work- 
ing memory  at  higher  processing  loads. 
Although  it  is  clear  that  alcohol 
impairs  explicit  memory  processes, 
alcohol  does  not  appear  to  impair 
implicit  memory  processes  (Lister  et 
al.  1991).  The  differential  effect  of 
acute  intoxication  on  explicit  versus 
implicit  memory  parallels  the  selective 
deficits  in  explicit,  but  not  implicit, 
memory  observed  in  Korsakoff's 
syndrome  (Lister  et  al.  1991).  Not 
only  do  these  data  provide  credence 
to  the  distinction  between  these  two 
types  of  memory,  but  they  also  sug- 


341 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


gest  a  common  neurophysiological 
basis  (perhaps  involving  cortical 
circuitry)  for  acute  and  long-term 
alcohol-induced  impairments  in 
memory.  Comparing  the  acute  effects 
of  alcohol  on  explicit  and  implicit 
learning  skills  (e.g.,  memory  and 
motor)  in  combination  with  brain 
imaging  (fMRI  and  PET)  could  be  a 
very  fruitful  line  of  work  and  might 
serve  clinical  (chronic  alcohol  effects) 
and  basic  (cognitive-neuroscience) 
research  objectives. 

In  addition  to  more  work  on  the 
assessment  of  the  effects  of  alcohol 
on  implicit  memory  and  learning 
processes,  systematic  research  is 
needed  on  the  effect  of  alcohol  on 
working  memory.  As  noted  above, 
the  fact  that  alcohol  has  a  greater 
memory  impairment  effect  with 
higher  processing  demands  suggests 
that  alcohol  impairs  working  mem- 
ory. This  is  directly  corroborated  in 
animal  research  designed  to  directly 
assess  the  effects  of  alcohol  on  work- 
ing memory.  Studies  clearly  indicate 
that  alcohol  impairs  spatial  and  non- 
spatial  working  memory  in  mice  and 
rats  (Melchior  et  al.  1993;  Givens  and 
McMahon  1997;  White  et  al.  1997). 
Since  working  memory  is  critical  for 
complex  problem  solving  and  the 
self- regulation  of  behavior  (Barkley 
1997),  both  of  which  are  thought  to 
be  associated  with  risk  for  alcoholism 
(Peterson  and  Pihl  1990;  Finn  et  al. 
1994),  it  is  quite  important  that  the 
effects  of  alcohol  on  working  memory 
be  systematically  studied  in  humans. 
Surprisingly,  a  literature  search  turned 
up  only  one  study  since  1983  that 
specifically  assessed  the  acute  effects  of 


a  moderate  dose  of  alcohol  on  work- 
ing memory  (Stokes  et.  al.  1991),  and 
this  study  reported  no  significant 
effects  of  alcohol  on  working  memory. 
This  finding  seems  unusual  given  the 
results  of  memory  studies  reported 
above.  In  addition,  divided  attention, 
which  is  significantly  disrupted  by 
even  low  doses  of  alcohol,  is  thought 
to  be  a  component  of  working  memory 
(Greene  et  al.  1995).  Interestingly, 
while  divided-attention  tasks  might 
tap  working  memory,  some  human 
and  animal  work  suggests  that  sus- 
tained attention  is  separate  from 
working  memory  processes  (Elliott  et 
al.  1997;  Givens  and  McMahon  1997). 
Studies  of  the  acute  effects  of  alcohol 
on  working  memory  and  performance 
in  sustained  and  divided-attention 
tasks  may  help  facilitate  the  identifica- 
tion of  the  neurophysiological  basis 
for  the  effect  of  alcohol  on  information 
processing.  Such  studies  may  also  reveal 
valuable  information  about  the  acute 
disruptive  effects  of  alcohol  on  the  self- 
regulation  of  behavior,  especially  the  self- 
regulation  of  alcohol  consumption. 

Other  Aspects  of 
Information  Processing 

Given  that  the  disruptive  effects  of 
acute  alcohol  intoxication  are  espe- 
cially apparent  on  more  complex  tasks 
that  demand  more  processing 
resources,  it  is  not  surprising  that 
alcohol  disrupts  basic  learning  and 
problem-solving  ability  (Carpenter  et 
al.  1961;  Peterson  et  al.  1990;  Salame 
1991).  Peterson  and  colleagues 
(1990)  reported  that  a  high  dose  of 
alcohol  impaired  performance  on  a 
paired-associates  learning  task,  a  maze 


342 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive -Disinhibited  Behavior 


task,  and  measures  of  word  fluency.  A 
high  dose  of  alcohol  has  also  been 
associated  with  deficits  in  lexical 
access  (Maylor  et  al.  1988),  word  cat- 
egorization even  when  combined 
with  semantic  priming  (Maylor  et  al. 
1987),  and  visual-spatial  ability  (Lex 
et  al.  1988;  Maylor  and  Rabbin 
1988;  Wang  et  al.  1992;  Pearson  and 
Timney  1998).  Interestingly,  acute 
intoxication  has  been  reported  to 
ameliorate  visual-spatial  learning 
deficits  in  alcoholics  (Schandler  et  al. 
1988&).  Schandler  and  colleagues 
(1988*,  1988&)  found  that  the  ame- 
lioration of  these  deficits  paralleled 
alcohol-induced  increases  in  right- 
hemisphere  ERP  components  (P3, 
P2,  Nl)  and  autonomic  activation 
after  alcohol,  suggesting  a  cognitive 
compensatory  effect  for  continued 
drinking  in  alcoholics  with  alcohol- 
induced  chronic  cognitive  deficits. 

Summary 

The  literature  on  the  acute  effects  of 
alcohol  on  information  processing  is 
extensive  and  quite  strong,  with  only 
a  relatively  few  gaps.  Studies  of  the 
acute  effects  of  alcohol  indicate  rather 
widespread  disruption  of  a  range  of 
facets  of  controlled  information  pro- 
cessing. A  compelling  argument  can 
be  offered  that  alcohol's  effect  on 
most,  if  not  all,  aspects  of  controlled 
processing  is  due  to  a  decrement 
in  available  processing  resources 
(Rohrbaugh  et  al.  1987;  Maylor  and 
Rabbitt  1993).  There  are  a  number 
of  gaps  in  this  literature  that  leave 
questions  about  (1)  the  influence  of 
individual  differences  and  develop- 
mental processes  on  acute  responses 


to  alcohol,  (2)  the  specific  effects  of 
alcohol  on  working  memory,  (3)  the 
basic  neurophysiological  mechanisms 
responsible  for  the  acute  effects  of 
alcohol  on  information  processing, 
and  (4)  the  mechanisms  by  which 
the  acute  effects  of  alcohol  are  trans- 
lated to  the  long-term,  more  chronic 
effects  of  high  levels  of  alcohol  con- 
sumption. Needed  are  studies  that 
combine  a  variety  of  central  nervous 
system  imaging  techniques  (e.g., 
fJVlRI  and  PET)  with  electrophysio- 
logical measures  and  behavioral  and 
psychological  measures  of  cognitive 
function,  using  creative  paradigms 
such  as  implicit  and  explicit  memory 
and  learning  tasks  and  working  mem- 
ory tasks.  Also  needed  are  studies 
that  include  subject  groups  with  low, 
moderate,  and  high  levels  of  alcohol 
consumption  with  varying  durations 
of  overall  alcohol  exposure  and 
studies  that  examine  the  influence  of 
the  limb  of  the  blood  alcohol  curve 
on  cognitive  performance. 

In  general,  studies  of  the  acute  effects 
of  alcohol  have  failed  to  adequately  mea- 
sure and  control  for  important  sources 
in  individual  differences  in  response  to 
alcohol,  such  as  drinking  level,  history, 
mood,  a  family  history  of  alcoholism, 
or  the  presence  of  other  traits  such  as 
behavior  problems  and  impulsivity  (cf. 
Parsons  and  Nixon  1998).  Also,  we 
know  absolutely  nothing  about  the  acute 
effects  of  alcohol  on  cognition  in  young 
persons  (preadolescent  and  adolescent 
samples).  Ethical  considerations  preclude 
the  administration  of  alcohol  to  such 
populations;  however,  it  is  possible  to 
use  a  longitudinal  study  design  to  track 
younger  persons  and  estimate  the 


343 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


effects  of  alcohol  exposure  on  various 
aspects  of  cognition  and  behavior. 

ACUTE  EFFECTS  OF 
ALCOHOL  ON  IMPULSIVE- 
DISINHIBITED  BEHAVIOR 

Although  studies  consistently  indicate 
that  impulsive -disinhibited  behavior  is 
associated  with  increased  drinking  and 
a  greater  risk  of  alcoholism  (e.g.,  Stewart 
and  Wilcox  1985;  Nagoshi  et  al.  1991; 
Sher  et  al.  1991;  Finn  et  al.  1994),  apart 
from  the  extensive  literature  on  alcohol - 
related  aggression  there  have  been  rela- 
tively few  studies  of  the  acute  effects  of 
alcohol  on  other  types  of  disinhibited 
behavior  or  factors  that  might  influence 
disinhibited  behavior. 

Disinhibited  behavior  includes  a 
number  of  specific,  but  highly  correlated, 
behaviors  labeled  as  impulsive,  aggressive, 
risk-taking,  undercontrolled,  external- 
izing, conduct-disordered,  sociopathic, 
hyperactive,  and  antisocial  (Gorenstein 
and  Newman  1980;  Finn  et  al.  1994). 
The  basic  construct  of  disinhibition  is 
defined  as  a  failure  to  inhibit  a  behavior 
that  is  likely  to  be  punished  (i.e.,  a 
behavioral  and  contingency  focus); 
however,  disinhibited  behavior  is  multi- 
faceted  and  multidetermined,  involving 
the  interaction  of  different  traits  (e.g., 
impulsivity  and  sensation  seeking),  motor 
processes,  and  a  range  of  cognitive  pro- 
cesses, conditioning  processes,  and  situa- 
tional determinants  (Barratt  and  Patton 
1983;  Finn  et  al.  1994;  Lykken  1995). 
For  instance,  impulsivity  (a  trait  com- 
ponent of  disinhibition)  is  considered  to 
have  both  cognitive  and  behavioral 
dimensions  (Barratt  and  Patton  1983; 
White  et  al.  1994).  Cognitive  dimensions 


include  fast  and  unreflective  (risky) 
decision  making,  a  lack  of  attention  paid 
to  future  events  (poor  planning),  poor 
time  perception,  and  poor  cognitive  inhi- 
bition assessed  using  Stroop  conflict 
errors.  Behavioral  dimensions  include 
poor  inhibitory  motor  control  (Logan 
et  al.  1997),  acting  without  thinking 
(Barratt  and  Patton  1983),  and  motor 
restlessness  and  impatience  (White  et  al. 
1994).  Individual  differences  in  impul- 
sivity, sensation  seeking,  and  neuro- 
psychological function  also  strongly 
influence  the  likelihood  for  disinhibited 
behavior  (e.g.,  Earleywine  and  Finn 
1991;  Lau  et  al.  1995;  Lykken  1995). 
Therefore,  to  fully  understand  the 
acute  effects  of  alcohol  on  disinhibited 
behavior  it  is  essential  that  research  sys- 
tematically address  the  multidimen- 
sional nature  of  disinhibited  behavior 
and  carefully  assess  the  role  of  individ- 
ual differences. 

The  vast  majority  of  studies  of  the 
acute  effects  of  alcohol  on  disinhibited- 
type  behavior  have  examined  the  alcohol- 
aggression  relationship.  A  handful  of 
studies  have  addressed  the  effects  of 
alcohol  on  other  facets  of  impulsive - 
disinhibited  behavior,  such  as  cognitive 
processes  (decision  making  and  bias, 
cognitive  inhibition  on  the  Stroop), 
attitudes,  and  motor  inhibition.  This 
section  of  the  chapter  is  divided  into 
two  parts:  (1)  a  review  of  studies  of 
the  acute  effects  of  alcohol  on  aggres- 
sion and  (2)  a  review  of  studies  of  the 
acute  effects  of  alcohol  on  other  facets 
of  disinhibited  behavior. 

Aggression 

The  association  between  alcohol 
intoxication  and  aggression  is  well 


344 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive-Disinhibited  Behavior 


documented  (Pernanen  1976;  Brain 
1986;  Bushman  and  Cooper  1990), 
extensively  researched  (cf.  reviews: 
Pihl  1983;  Bushman  and  Cooper 
1990;  Gustafson  1993),  and  appears 
to  be  common  knowledge  in  Western 
society.  Aggressive  behavior  in  child- 
hood is  a  strong  predictor  of  adolescent 
and  adult  alcohol  problems  (Farring- 
ton  1991),  and  alcohol  intoxication 
appears  to  increase  the  likelihood  for 
aggressive  behavior  (Gustafson  1993). 
The  alcohol- aggression  relationship  has 
been  studied  from  various  perspectives, 
including  epidemiologic  (e.g.,  Perna- 
nen 1976;  Roizen  1997),  longitudinal 
(Farrington  1991;  White  et  al.  1993), 
biological  (Virkkunen  and  Linnoila 
1990),  social  information  processing 
(Sayette  et  al.  1993),  psychological  or 
individual  differences  (Gustafson 
1993;  Lau  et  al.  1995),  and  interper- 
sonal (Leonard  and  Senchak  1993). 

There  is  an  extensive  literature  on 
laboratory  studies  of  the  acute  effects 
of  alcohol  on  aggressive  behavior  (cf. 
Gustafson  1993).  A  considerable  amount 
is  known  about  the  factors  that  might 
mediate,  or  moderate,  the  relationship 
between  alcohol  intoxication  and  aggres- 
sion. For  instance,  the  level  of  provoca- 
tion appears  to  be  critical  in  determining 
whether  intoxicated  subjects  will  respond 
aggressively  in  laboratory  settings. 
Without  provocation,  alcohol  does 
not  appear  to  increase  aggression 
(Gustafson  1993).  Other  factors  that 
appear  to  increase  the  likelihood  that 
alcohol  intoxication  will  lead  to 
aggression  in  laboratory  and  field  set- 
tings are  the  type  of  beverage  (spirits 
but  not  beer  or  wine)  (Gustafson 
1988;  Murdoch  and  Pihl  1988; 


Gustafson  1990),  alcohol  dose 
(Sayette  et  al.  1993),  frustration  level 
(Gustafson  1993;  Sayette  et  al.  1993), 
schedule  of  provocation  (Kelly  and 
Cerek  1993),  availability  of  nonag- 
gressive  alternatives  (Gustafson  1993), 
and  the  saliency  of  threat  or  frustra- 
tion cues  (Gustafson  1986;  Gantner 
and  Taylor  1992). 

Although  most  of  the  factors  that 
influence  the  likelihood  of  intoxication 
leading  to  violence  in  laboratory  studies 
are  contextual  in  nature,  support  for 
the  compelling  argument  that  only 
some  individuals  become  violent  when 
drunk  is  offered  by  a  longitudinal 
study.  White  and  colleagues  (1993) 
studied  a  large  sample  (N  =  1,380)  of 
adolescents  at  3 -year  intervals  (12,  15, 
and  18  years  of  age)  and  modeled  the 
influence  of  parental  education,  alcohol 
use  at  the  three  time  periods,  and 
alcohol-related  aggression  at  ages  15 
and  18  years.  Their  analysis  revealed 
that  early  aggression  led  to  later  alcohol 
use,  but  early  alcohol  use  did  not  predict 
later  aggression  (when  controlling  for 
initial  levels  of  aggression).  Further- 
more, the  only  predictors  of  alcohol- 
related  aggressive  behavior  were  both 
early  and  contemporary  levels  of  aggres- 
sive behavior  (non-alcohol  related).  In 
other  words,  aggressive  people  were 
the  ones  who  were  aggressive  when 
drinking  (White  et  al.  1993).  Interest- 
ingly, these  researchers  observed  that 
lower  levels  of  parental  education 
were  also  directly  predictive  of  alco- 
hol-related aggression  at  age  15,  sug- 
gesting a  role  for  familial  intellectual 
functioning  in  alcohol-related  aggres- 
sion. Their  data  are  consistent  with  the 
finding  by  Lau  and  colleagues  (1995) 


345 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


that  subjects  with  evidence  of  poor 
executive  cognitive  function  (frontal 
lobe  function)  showed  the  highest 
level  of  provocation-induced  aggressive 
behavior  when  intoxicated.  Research 
and  theory  suggest  that  deficits  in 
frontal  lobe  function  are  associated 
with  disinhibited,  aggressive  personal- 
ity traits  (Giancola  and  Zeichner 
1994;  Lau  et  al.  1995)  and  may  form, 
in  part,  the  neuropsychological  basis 
for  early-onset  aggressive  behavior. 

The  findings  of  White  and  colleagues 
(1993)  and  Lau  and  colleagues  (1995) 
are  also  consistent  with  research  associat- 
ing deficits  in  central  serotonergic 
function  with  alcohol-related  violence 
(Virkkunen  and  Linnoila  1990,  1993). 
Reduced  brain  serotonin  function  and 
frontal  lobe  deficits  both  have  been 
associated  with  disinhibited,  impulsive 
behavior  (Gorenstein  and  Newman  1980; 
Miller  1987;  Virkkunen  and  Linnoila 
1993).  Research  on  the  interrelationships 
between  alcohol-related  aggression, 
impulsive  or  aggressive  traits,  serotonin 
function,  and  executive  cognitive  function 
is  a  promising  avenue  for  research  on  the 
biobehavioral  basis  for  the  acute  effects 
of  alcohol  on  aggression.  However,  much 
remains  to  be  understood  about  ( 1 )  the 
origins  (biological  and  environmental) 
of  reduced  brain  serotonin  function,  poor 
executive  cognitive  function,  and  early- 
onset  aggression  and  (2)  the  mechanisms 
by  which  reduced  serotonin  function 
and  poor  executive  function  lead  to 
aggressive  behavior,  alcohol  problems, 
and  alcohol-related  aggression. 

The  literature  on  the  acute  effects  of 
alcohol  on  aggression  is  extensive  and 
strong.  The  primary  strength  of  the  liter- 
ature is  the  delineation  of  the  important 


role  that  a  range  of  contextual  factors 
might  play  in  influencing  the  likelihood  of 
alcohol-related  aggression.  Less  exten- 
sively researched  are  the  biobehavioral 
bases  (origins  and  mechanisms)  of  the 
individual  differences  that  influence  the 
alcohol- aggression  relationship.  Research 
combining  brain  imaging  (fMRI,  PET, 
dipole  ERP  source),  behavioral  and 
cognitive  paradigms,  and  careful  assess- 
ments of  environmental  context 
(parental  influence,  stress,  peer  factors) 
using  longitudinal  designs  (to  assess  the 
developmental  origins  and  processes  and 
the  effects  of  early  exposure  to  alcohol) 
and  cross-sectional  alcohol  challenge 
designs  (with  high-risk  [impulsive  or 
aggressive]  and  low- risk  subjects)  should 
substantially  increase  our  knowledge 
of  the  biobehavioral  bases  of  important 
individual  differences  affecting  alcohol - 
related  aggression. 

Other  Facets 

of  Disinhibited  Behavior 

Alcohol  might  increase  the  likelihood 
for  disinhibited  behavior  by  increasing 
risky- type  decision  making  (decreasing 
caution);  affecting  attitudes  about  engag- 
ing in  risky  behaviors  (e.g.,  attitudes 
about  condom  use  or  drinking  and  dri- 
ving); affecting  skills  and  self-efficacy 
for  engaging  in  safe  behaviors;  compro- 
mising planning  or  future-oriented  think- 
ing; decreasing  attention  to,  or  salience  of, 
stimuli  signaling  potential  punishment; 
and  decreasing  general  impulse  control 
(poor  inhibitory  motor  control). 

Attitudes  and  Decisions  About 
Risky  Behaviors 

There  is  some  evidence  that  alcohol 
intoxication  affects  specific  attitudes 


346 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive-Disinhibited  Behavior 


that,  in  turn,  may  increase  the  likeli- 
hood of  choosing  to  engage  in  risky 
behaviors.  In  a  field  study,  MacDon- 
ald  and  colleagues  (1995)  found  that 
intoxicated  subjects  appeared  to  be 
more  willing  to  consider  driving  a 
short  distance  after  drinking  than 
nonintoxicated  subjects,  suggesting 
that  alcohol  may  result  in  somewhat 
more  risky  decision  making.  In  two 
studies  of  the  effects  of  a  moderate 
dose  of  alcohol  on  attitudes  about 
condom  use  and  self- efficacy  toward 
negotiating  condom  use,  acute  intoxi- 
cation was  associated  with  more  nega- 
tive attitudes  about  condom  use 
(Gordon  and  Carey  1996;  Gordon  et 
al.  1997),  lower  perceived  self-efficacy 
to  initiate  or  negotiate  condom  use 
(Gordon  and  Carey  1996),  and 
poorer  actual  behavioral  skills  (role 
play)  to  negotiate  condom  use  (Gor- 
don et  al.  1997),  when  compared 
with  control  subjects. 

This  literature  is  still  in  its  infancy 
and  is  weak.  The  dependent  measures 
and  experimental  paradigms  are  more 
primitive  and  not  abreast  with  current 
paradigms  for  attitude  measurement 
and  perceptual  organization  used  in 
social  and  cognitive  psychology  (for 
discussions  of  paradigms,  see  Smith  et 
al.  1996;  McFall  et  al.  1998).  Apart 
from  a  role  play  in  the  study  by 
Gordon  and  colleagues  (1997),  the 
dependent  measures  used  in  these 
studies  were  paper-and-pencil  self- 
report  scales  of  attitudes  or  ratings  of 
self-efficacy.  These  measures  do  not 
employ  the  more  sophisticated  and 
objective  approaches  to  attitude 
measurement  that  have  been  routinely 
employed  in  social  psychological 


research,  such  as  categorization  tasks 
involving  similarity  judgments  (Smith 
et  al.  1996;  Fazio  and  Dunton  1997). 
Such  tasks  are  firmly  grounded  in  well- 
researched  theories  and  methodologies 
in  cognitive  science  (Nosofsky  and 
Palmeri  1996,  1997)  and  provide  a 
more  objective  assessment  of  attitude 
strength  (Smith  et  al.  1996;  Fazio  and 
Dunton  1997).  Finally,  the  studies  by 
MacDonald  and  colleagues  (1995) 
and  Gordon  and  colleagues  (1997) 
also  purport  to  have  some  relevance 
for  decision  making;  however,  the 
effects  of  alcohol  on  decision-making 
processes  are  not  directly  assessed. 

In  a  more  direct  assessment  of  the 
effects  of  alcohol  on  risky  decisions, 
Meier  and  colleagues  (1996)  studied 
the  acute  effect  of  alcohol  using  a  risky 
decision-type  task  situation  where  the 
magnitude  and  probability  to  punish- 
ments (failure  to  win  money)  were 
varied.  These  investigators  found,  to 
their  surprise,  that  alcohol  did  not 
suppress  the  effects  of  punishment 
(loss  of  money).  Although  this  study's 
design  represents  a  step  in  the  right 
direction  for  the  literature  (its  design 
is  systematic  and  the  dependent  mea- 
sures are  objective),  it  is  the  only  study 
directly  assessing  the  effects  of  alcohol 
on  decision-making  processes.  Although 
it  appears  surprising  that  alcohol  failed 
to  suppress  the  effects  of  punishment 
on  risky  decisions,  the  lack  of  punish- 
ment suppression  by  alcohol  may  have 
been  due  to  the  type  of  punishment 
(loss  of  money),  whose  salience  may 
have  remained  high  for  this  popula- 
tion (college  students)  in  spite  of 
being  intoxicated.  Models  of  behav- 
ioral disinhibition  focus  on  a  failure  to 


347 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


inhibit  behavior  in  contexts  of 
impending  aversive  stimulation,  rather 
than  loss  of  reward  (Gray  1975;  Finn 
et  al.  1994).  For  instance,  other 
research  suggests  that  disinhibited 
individuals  (psychopaths)  learn  to 
inhibit  behavior  when  that  behavior 
leads  to  loss  of  money,  but  fail  to 
learn  to  inhibit  when  the  behavior 
leads  to  an  aversive  outcome,  such  as 
electric  shock  (Schmauk  1970). 

There  is  some  evidence  suggesting 
that  alcohol  results  in  an  overall  bias 
to  respond  (decreases  the  beta 
[caution]  parameter)  and  decreases 
caution  in  perceptual  decision  making 
on  CPT  tasks  (Rohrbaugh  et  al. 
1987;  Mongrain  and  Standing  1989). 
However,  in  CPT  tasks  assessing 
recognition  memory,  alcohol  has  been 
reported  to  increase  the  beta  para- 
meter (Maylor  and  Rabbitt  1987). 
The  beta  parameter  may  be  a  useful 
dependent  measure  for  assessing  cau- 
tious versus  risky  shifts  in  decision 
bias  after  alcohol;  however,  it  is  prob- 
ably more  useful  in  signal  detection 
tasks  where  a  risk  parameter  is  more 
directly  manipulated. 

Research  on  the  effects  of  alcohol 
on  decision  making  would  be 
greatly  enhanced  by  looking  to  theory 
and  research  on  dynamic,  stochastic 
decision-making  processes  within  the 
cognitive  science  research  literature 
(Busemeyer  and  Townsend  1993; 
Diederich  1997).  In  dynamic  decision- 
making experimental  paradigms, 
the  influence  of  time  constraints, 
uncertainty,  risk,  and  preferences 
(as  they  may  change  over  time) 
on  sequential  decision-making  pro- 
cesses can  be  assessed  over  time  and 


mathematically  predicted  (Busemeyer 
and  Townsend  1993). 

Motor  Inhibition 

Although  the  data  are  scant,  there  is 
evidence  that  alcohol  may  decrease 
inhibitory  motor  control.  A  recent 
study  using  Logan  and  Cowan's  (1984) 
stop -signal  task  indicates  that  alcohol 
can  impair  inhibitory  motor  control 
(Mulvihill  et  al.  1997).  The  stop-signal 
task  is  a  two-choice  CPT,  where  the 
subject  responds  on  each  trial  using 
one  of  two  keys  depending  on  the 
stimulus  (X  or  O  =  go  signal).  On  25 
percent  of  trials  an  acoustic  stop-signal 
(a  tone)  is  presented  at  some  latency 
after  the  go  signal.  The  subject  is 
required  to  inhibit  his  or  her  response 
after  detecting  the  stop-signal  (see 
Logan  et  al.  1997  for  a  description). 
Impulsive  traits  in  adults  and  child- 
hood behavioral  problems  have  been 
associated  with  relatively  poor  inhibition 
on  this  task  (Logan  et  al.  1997). 
Compared  with  placebo  and  no  alco- 
hol conditions,  alcohol  was  associated 
with  a  sevenfold  decrease  in  inhibitory 
control  (Mulvihill  et  al.  1997).  This 
effect  has  been  replicated  by  the  same 
laboratory  (Fillmore  and  Vogel-Sprott 
unpublished  data). 

The  stop -signal  task  has  been  used 
extensively  in  the  study  of  inhibitory 
control  in  patient  populations  (see,  e.g., 
Schachar  and  Logan  1990;  Schachar 
et  al.  1995),  and  it  is  associated  with  a 
well-tested  and  quantifiable  information- 
processing  theoretical  framework  (Logan 
and  Cowan  1984;  Logan  et  al.  1997). 
The  stop-signal  paradigm  is  a  very 
promising  paradigm  to  systematically 
apply  to  the  study  of  the  acute  effects 


348 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive -Disinhibited  Behavior 


of  alcohol;  however,  researchers  also 
need  to  study  other  paradigms  assessing 
other  facets  of  disinhibited  behavior 
and  the  influence  of  preexisting  disin- 
hibited traits,  such  as  impulsive,  aggres- 
sive, and  sensation- seeking  traits. 

Other  evidence  that  acute  intoxica- 
tion may  compromise  inhibitory  motor 
control  comes  from  Peterson  and  col- 
leagues (1990),  who  found  that  alcohol 
had  a  detrimental  effect  on  Porteus 
maze  performance.  Although  the  Por- 
teus maze  does  not  directly  assess  motor 
inhibition,  poor  performance  on  the 
task  is  associated  with  disinhibited 
behavior  after  prefrontal  lobotomy 
(Crown  1952).  Another  promising 
paradigm  for  investigating  the  effects 
of  alcohol  on  behavioral  inhibition  is 
the  GO/NO  GO  learning  task,  where 
passive  avoidance  in  response  to  differ- 
ent types  of  punishment  contingencies 
can  be  assessed  (Newman  et  al.  1985; 
Iaboni  et  al.  1995).  Performance  on 
GO/NO  GO  tasks  can  be  used  to 
assess  relative  roles  of  the  behavioral 
inhibition  and  activation  systems  (Gray 
1975),  which  have  been  associated 
with  activity  in  central  serotonin  and 
dopamine  systems  (Gray  1975). 

Cognitive  Inhibition 

Interference  on  the  Stroop  task  is 
thought  to  tap  the  ability  to  inhibit  an 
overlearned  response  (reading  a  word) 
in  favor  of  a  competing  novel  response 
(White  et  al.  1994).  This  measure  of 
cognitive  inhibition  has  been  modestly 
associated  with  other  measures  of 
impulsivity  (White  et  al.  1994). 
Gustafson  and  Kallmen  (1990#, 
1990&)  reported  that  alcohol  had  no 
effect  on  Stroop  performance  (i.e.,  no 


apparent  interference  effect).  A  poten- 
tial problem  with  these  two  studies 
was  the  lack  of  a  specific  and  sensitive 
measure  of  interference. 

Summary 

There  is  a  general  dearth  of  studies  of 
the  effect  of  alcohol  on  the  cognitive 
and  behavioral  elements  of  impulsivity 
and  disinhibition.  Many  of  the  studies 
that  have  been  done  are  not  well 
informed  by  research  in  other  areas, 
such  as  cognitive  science.  This  is  an 
extremely  important  area  of  research 
because  of  its  relevance  for  alcohol's 
effect  on  impulsive  decision  making, 
motor  control,  and  response  persevera- 
tion on  the  self- regulation  of  drinking. 
For  instance,  research  in  this  area  could 
shed  some  light  on  the  mechanisms 
by  which  some  individuals  might 
impulsively  extend  social  drinking  into 
binge  drinking.  In  addition,  more 
research  in  this  area  would  increase 
knowledge  about  how  alcohol  might 
influence  decisions  to  engage  in  risky 
and/or  illegal  behaviors  (drinking  and 
driving,  participating  in  dangerous 
activities  while  intoxicated,  engaging 
in  unprotected  sex)  and  how  early 
exposure  to  alcohol  might  affect  the 
development  of  impulsive  traits  in 
younger  individuals,  which,  in  turn, 
would  contribute  to  their  overall  risk 
for  alcoholism. 

Clearly,  a  substantial  amount  of 
research  is  needed  in  this  area.  Two 
major  avenues  of  research  should  be 
undertaken:  (1)  longitudinal  studies 
of  the  effects  of  early  exposure  to 
alcohol  (preadolescent  and  adolescent) 
on  the  development  of  disinhibited 
traits  and  (2)  cross-sectional  studies  of 


349 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


the  effects  of  alcohol  challenge  on  dis- 
inhibited/impulsive  behavior  (and  fac- 
tors associated  with  such  behavior), 
using  multimethod  measurements  to 
capture  the  multidimensional  nature 
of  behavioral  disinhibition  and  control- 
ling for  important  sources  of  individ- 
ual differences,  such  as  preexisting 
disinhibited  traits,  a  family  history  of 
alcoholism  (and  antisocial  behavior), 
drinking  history  and  level,  and  limb  of 
the  blood  alcohol  curve.  Longitudinal 
research  is  needed  where  the  effects  of 
early  exposure  to  alcohol  on  behavioral 
disinhibition  are  assessed,  because  it  is 
likely  that  early  exposure  to  alcohol  itself 
may  contribute  to  increased  impulsivity 
in  childhood,  leading  to  increased  risk 
for  serious  and  long-term  problems 
with  alcohol  and  other  drugs. 

Finally,  literature  in  the  areas  of 
stochastic  decision- making  dynamics 
(Busemeyer  and  Townsend  1993), 
the  measurement  of  impulsivity 
(Barratt  and  Patton  1983;  White  et 
al.  1994;  Logan  et  al.  1997),  condi- 
tioning and  disinhibition  (Newman  et 
al.  1985;  Finn  et  al.  1994;  Iaboni  et 
al.  1995),  and  the  cognitive  correlates 
of  disinhibited  traits  (Kosson  and 
Newman  1986;  Howland  et  al.  1993) 
presents  potentially  valuable  theoreti- 
cal models,  experimental  paradigms, 
and  measurement  methods  that  can 
be  used  in  the  study  of  the  effects  of 
alcohol  on  cognitive  aspects  of  poor 
impulse  control. 

ACKNOWLEDGMENTS 

Support  for  this  work  was  provided 
by  National  Institutes  of  Health/ 
National  Institute  on  Alcohol  Abuse 


and  Alcoholism  grants  R01-AA10120 
andP50-AA07611. 


REFERENCES 

Barkley,  RA.  Behavioral  inhibition,  sustained 
attention,  and  executive  cognitive  function: 
Constructing  a  unifying  theory  of  ADHD. 
Psychol  Bull  121:65-94,  1997. 

Barratt,  E.S.,  and  Patton,  J.H.  Impulsivity: 
Cognitive,  behavioral,  and  psychophysio- 
logical correlates.  In:  Zuckerman,  M.,  ed. 
The  Biological  Basis  of  Sensation  Seeking, 
Impulsivity,  and  Anxiety.  Hillsdale,  NJ: 
Erlbaum,  1983.  pp.  77-116. 

Battig,  K.,  and  Buzzi,  R.  Effect  of  coffee 
on  the  speed  of  subject-paced  information 
processing.  Neuropsychology  16:126- 
130, 1986. 

Beirness,  DJ.,  and  Vogel-Sprott,  M.  The 
development  of  acute  tolerance:  Acute 
recovery  as  a  predictor.  Psychopharmacology 
84:398-401,  1984. 

Birnbaum,  I.M.,  and  Parker,  E.S.  Acute 
effects  of  alcohol  on  storage  and  retrieval. 
In:  Birnbaum,  I.M.,  and  Parker,  E.S.,  eds. 
Alcohol  and  Human  Memory.  Hillsdale, 
NJ:  Erlbaum,  1977. 

Brain,  P.F.  Alcohol  and  Agression.  Dover, 
NH:  Croom  Helm,  1986. 

Brewer,  N.,  and  Sandow,  B.  Alcohol  effects 
on  driver  performance  under  conditions 
of  divided  attention.  Ergonomics  23:185- 
190, 1980. 

Busemeyer,  J.R.,  and  Townsend,  J.T. 
Decision  field  theory:  A  dynamic-cognitive 
approach  to  decision  making  in  an  uncer- 
tain environment.  Psychol  Rev  100:432- 
459, 1993. 

Bushman,  BJ.,  and  Cooper,  H.M.  Effects 
of  alcohol  on  human  aggression:  An 


350 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive- Disinhibited  Behavior 


integrative  research  review.  Psychol  Bull 
107:341-354,  1990. 

Campbell,  K.B.,  and  Lowick,  B.M. 
Ethanol  and  event-related  potentials:  The 
influence  of  distractor  stimuli.  Alcohol 
4:257-263,  1987. 

Carpenter,  J. A.;  Moore,  O.K.;  Snyder, 
C.R.;  and  Lisansky,  E.S.  Alcohol  and 
higher-order  problem  solving.  QJ  Stud 
Alcohol 22:182-222,  1961. 

Cohen,  H.L.;  Porjesz,  B.;  and  Begleiter, 
H.  The  effects  of  ethanol  on  EEG  activity 
in  males  at  risk  for  alcoholism.  Electro- 
encephalogr  Clin  Neurophysiol  86:368- 
376,  1993. 

Craik,  F.I.M.  Similarities  between  the 
effects  of  aging  and  alcoholic  intoxication 
on  memory,  performance,  construed 
within  a  "levels  of  processing"  framework. 
In:  Birnbaum,  I.M.,  and  Parker,  E.S.,  eds. 
Alcohol  and  Human  Memory.  Hillsdale, 
NJ:  Erlbaum,  1977.  pp.  9-21. 

Crown,  S.  An  experimental  study  of  psy- 
chological changes  following  prefrontal 
lobotomy.  J  Gen  Psychol  47:3-41,  1952. 

Diederich,  A.  Dynamic  stochastic  models 
for  decision  making  under  time  constraints. 
J  Math  Psychol  41:260-274,  1997. 

Earleywine,  M.,  and  Finn,  P.R.  Sensation 
seeking  explains  the  relation  between 
behavioral  disinhibition  and  alcohol  con- 
sumption. Addict  Behav  16:123-128,  1991. 

Elliott,  R.;  Sahakian,  B.J.;  and  Mathew, 
K.  Effects  of  methylphenidate  on  spatial 
working  memory  and  planning  in  healthy 
young  adults.  Psychopharmacology  (Berl) 
131:196-206,  1997. 

Farrington,  D.P.  Childhood  aggression 
and  adult  violence:  Early  precursors  and 
later  life  outcomes.  In:  Pepler,  D.J.,  and 
Rubin,  K.H.,  eds.  The  Development  and 


Treatment  of  Childhood  Aggression.  New 
York:  Plenum,  1991.  pp.  5-29. 

Fazio,  R.H.,  and  Dunton,  B.C.  Categoriza- 
tion by  race:  The  impact  of  automatic  and 
controlled  components  of  racial  prejudice. 
J  Exp  Soc  Psychol  33:451-470,  1997. 

Fillmore,  M.T.;  Carscadden,  J.L.;  and 
Vogel-Sprott,  M.  Alcohol,  cognitive 
impairment  and  expectancies.  J  Stud 
Alcohol  59:174-179,  1998. 

Finn,  P.R;  Earleywine,  M.;  and  Pihl, 
R.O.  Sensation  seeking,  stress  reactivity, 
and  alcohol  dampening  discriminate  the 
density  of  a  family  history  of  alcoholism. 
Alcohol  Clin  Exp  Res.  16:585-590,  1992. 

Finn,  P.R;  Kessler,  D.N.;  and  Hussong,  AM. 
Risk  for  alcoholism  and  classical  condition- 
ing to  signals  for  punishment:  Evidence 
for  a  weak  behavioral  inhibition  system.  / 
Abnorm  Psychol  103:293-301,  1994. 

Fitzhugh,  L.;  Fitzhugh,  K.;  and  Reitan, 
R.  Adaptive  abilities  and  intellectual  func- 
tioning of  hospitalized  alcoholics:  Further 
considerations.  QJ  Stud  Alcohol  26:402- 
411,  1965. 

Gantner,  A.B.,  and  Taylor,  S.P.  Human 
physical  aggression  as  a  function  of  alcohol 
threat  and  harm.  Aggressive  Behav  18:29- 
36,  1992. 

Giancola,  P.R,  and  Zeichner,  A.  Neuro- 
psychological performance  on  tests  of 
frontal-lobe  functioning  and  aggressive 
behavior  in  men.  ]  Abnorm  Psychol 
103:832-835, 1994. 

Givens,  B.,  and  McMahon,  K.  Effects  of 
ethanol  on  nonspatial  working  memory 
and  attention  in  rats.  Behav  Neurosci 
111:275-282,1997. 

Gordon,  CM.,  and  Carey,  M.P.  Alcohol's 
effects  on  requisites  for  sexual  risk  reduction 


351 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  men:  An  initial  experimental  investigation. 
Health  Psychol  15:56-60,  1996. 

Gordon,  CM.;  Carey,  M.P.;  and  Carey, 
K.B.  Effects  of  drinking  on  behavioral  skills 
and  condom  attitudes  in  men:  Implications 
for  HIV  risk  from  a  controlled  experiment. 
Health  Psychol  16:490^195,  1997. 

Gorenstein,  E.E.,  and  Newman,  J. P. 
Disinhibitory  psychopathology:  A  new 
perspective  and  a  model  for  research. 
Psychol  Rev  87:301-315,  1980. 

Gray,  J. A.  Elements  in  a  Two-Process  Theory 
of  Learning.  San  Diego,  CA:  Academic 
Press,  1975. 

Greene,  J.D.W.;  Hodges,  J.R.;  and 
Baddeley,  A.D.  Autobiographical  memory 
and  executive  function  in  early  dementia 
of  the  Alzheimer  type.  Neuropsychologia 
33:1647-1670,  1995. 

Gustafson,  R.  Alcohol,  frustration,  and 
aggression.  Psychol  Rep  59:207-218,  1986. 

Gustafson,  R.  Beer  intoxication  and  physi- 
cal aggression  in  males.  Drug  Alcohol  Depend 
21:237-242,  1988. 

Gustafson,  R.  Wine  and  male  physical 
aggression.  J  Drug  Issues  20:75-86,  1990. 

Gustafson,  R.  What  do  experimental 
paradigms  tell  us  about  alcohol-related 
aggressive  responding?  /  Stud  Alcohol 
Suppl  11:20-29, 1993. 

Gustafson,  R,  and  Kallmen,  H.  Effects  of 
alcohol  on  cognitive  performance  measured 
with  Stroop's  Color  Word  Test.  Percept 
Mot  Skills  71:99-105,  1990*. 

Gustafson,  R.,  and  Kallmen,  H.  Effects  of 
alcohol  on  prolonged  cognitive  performance 
measured  with  Stroop's  Color  Word  Test. 
Psych  Rep  67:643-650,  1990£. 

Hasenfratz,  M.;  Bunge,  A.;  Dal-Pra,  G.; 
and  Baettig,  K.  Antagonistic  effects  of 


caffeine  and  alcohol  on  mental  perfor- 
mance. Pharmacol  Biochem  Behav 
46:463-465,  1993. 

Howland,  E.W.;  Kosson,  D.S.;  Patterson, 
M.;  and  Newman,  J. P.  Altering  a  dominant 
response:  Performance  of  psychopaths 
and  low-socialization  college  students  on 
a  cued  reaction  time  task.  J  Abnorm 
Psychol  102:379-387 ,  1993. 

Iaboni,  F.;  Douglas,  V.I.;  and  Baker,  A.G. 
Effects  of  reward  and  response  costs  on 
inhibition  in  ADHD  children.  J  Abnorm 
Psychol  104:232-240,  1995. 

Jaaskelainen,  LP.;  Lehtokoski,  A.;  Kujala, 
T.;  Pekkonen,  E.;  Sinclair,  J.D.;  Naatanen, 
R;  and  Sillanaukee,  P.  Low  dose  of  ethanol 
suppresses  mismatch  negativity  of  auditory 
event-related  potential.  Alcohol  Clin  Exp 
Res  19:607-610,  1995. 

Jaaskelainen,  LP.;  Naatanen,  R.;  and 
Sillanaukee,  P.  Effect  of  acute  ethanol  on 
auditory  and  visual  event-related  poten- 
tials: A  review  and  reinterpretation.  Biol 
Psychiatry  40:284-291,  1996. 

Jones,  B.M.,  and  Jones,  M.K.  Alcohol 
and  memory  impairment  in  male  and 
female  social  drinkers.  In:  Birnbaum, 
I.M.,  and  Parker,  E.S.,  eds.  Alcohol  and 
Human  Memory.  Hillsdale,  NJ:  Erlbaum, 
1977.  pp.  127-138. 

Kelly,  T.H.,  and  Cerek,  D.R  The  effects 
of  alcohol  on  free-operant  aggressive  be- 
havior. /  Stud  Alcohol  Suppl  1 1 :40-52, 1993. 

Kleinknecht,  R.,  and  Goldstein,  S.  Neuro- 
psychological deficits  associated  with 
alcoholism.  QJ  Stud  Alcohol  33:999- 
1019, 1972. 

Koelega,  H.S.  Alcohol  and  vigilance 
performance:  A  review.  Psychopharmacology 
(Berl)  118:233-249,  1995. 


352 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive -Disinhibited  Behavior 


Kosson,  D.S.,  and  Newman,  J. P.  Psycho- 
pathy and  the  allocation  of  attentional 
capacity  in  a  divided- attention  situation.  J 
Abnorm  Psychol  5:257-263,  1986. 

Krein,  S.;  Overton,  S.;  Young,  M.;  Spreier, 
K.;  and  Yolton,  R.L.  Effects  of  alcohol  on 
event- related  brain  potentials  produced 
by  viewing  a  simulated  traffic  signal.  J  Am 
Optom  Assoc  58:474-477,  1987. 

Landauer,  T.K.  Remarks  on  the  detection 
and  analysis  of  memory  deficits.  In: 
Birnbaum,  I.M.,  and  Parker,  E.S.,  eds. 
Alcohol  and  Human  Memory.  Hillsdale, 
NJ:  Erlbaum,  1977.  pp.  23-41. 

Lau,  M.A.;  Pihl,  R.O.;  and  Peterson,  J.B. 
Provation,  acute  alcohol  intoxication, 
cognitive  performance,  and  aggression.  / 
Abnorm  Psychol  104:150-155,  1995. 

Leonard,  K.E.,  and  Senchak,  M.  Alcohol 
and  premarital  aggression  among  newly- 
wed  couples.  J  Stud  Alcohol  Suppl 
11:96-108,  1993. 

Lex,  B.W.;  Greenwald,  N.E.;  Lukas,  S.E.; 
Slater,  J. P.;  and  Mendelson,  J.H.  Blood 
ethanol  levels,  self-rated  ethanol  effects 
and  cognitive-perceptual  tasks.  Pharmacol 
Biochem  Behav  29:509-515,  1988. 

Lex,  B.W.;  Rhoades,  E.M.;  Teoh,  S.K.; 
Mendelson,  J.H.;  and  Greenwald,  N.E. 
Divided  attention  task  performance  and 
subjective  effects  following  alcohol  and 
placebo:  Differences  between  women  with 
and  without  a  family  history  of  alcoholism. 
Drug  Alcohol  Depend  35:95-105,  1994. 

Linnoila,  M.;  Stapleton,  J.M.;  Lister,  R; 
Guthrie,  S.;  and  Eckardt,  M.  Effects  of 
alcohol  on  accident  risk:  Epidemiology 
and  laboratory  studies.  Pathologist 
40:36-41,  1986. 

Lister,  RG;  Gorenstein,  C;  Risher-Flowers, 
D.;  Weingartner,  H.J.;  and  Eckardt,  M. 
Dissociation  of  the  acute  effects  of  alcohol 


on  implicit  and  explicit  memory  processes. 
Neuropsycholo£fia  29:1205-1212,  1991. 

Logan,  G.D.,  and  Cowan,  W.B.  On  the 
ability  to  inhibit  thought  and  action:  A 
theory  of  act  control.  Psychol  Rev  91 :295- 
327,  1984. 

Logan,  G.D.;  Schachar,  R.J.;  and 
Tannock,  R.  Impulsivity  and  inhibitory 
control.  Psychol  Sci  8:60-64,  1997. 

Lukas,  S.A.;  Mendelson,  J.H.;  Kouri,  E.; 
Bolduc,  M.;  and  Amass,  L.  Ethanol-induced 
alterations  in  EEG  alpha  activity  and 
apparent  source  of  the  auditory  P300 
evoked  response  potential.  Alcohol 
7:471-477,  1990. 

Lykken,  D.T.  The  Antisocial  Personalities. 
Hillsdale,  NJ:  Erlbaum,  1995. 

MacDonald,  T.K.;  Zanna,  M.P.;  and 
Fong,  G.T.  Decision  making  in  altered 
states:  Effects  of  alcohol  on  attitudes 
toward  drinking  and  driving.  J  Pers  Soc 
Psychol  68:973-985,  1995. 

Maylor,  E.A,  and  Rabbitt,  P.M.A  Effect 
of  alcohol  on  rate  of  forgetting. 
Psychopharmacology  91:230-235,  1987. 

Maylor,  E.A.,  and  Rabbitt,  P.M.A. 
Amount  of  practice  and  degree  of  atten- 
tional  control  have  no  influence  on  the 
adverse  effect  of  alcohol  in  word  catego- 
rization and  visual  search  tasks.  Percept 
Psychophysics  44:117-126,  1988. 

Maylor,  E.A.,  and  Rabbitt,  P.MA.  Alcohol, 
reaction  time  and  memory:  A  meta-analysis. 
Br  J  Psychol  84:301-317,  1993. 

Maylor,  E.A.;  Rabbitt,  P.M.A.;  and 
Kingstone,  A.F.  Effects  of  alcohol  on 
word  categorization  and  recognition 
memory.  Br  J  Psychol  78:233-239,  1987. 

Maylor,  E.A.;  Rabbitt,  P.M.A.;  and 
Kingstone,  A.F.  Effects  of  alcohol  on 


353 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


lexical  access.  Psychopharmacology 
95:119-123,1988. 

Maylor,  E.A.;  Rabbitt,  P.M.A.;  James, 
G.LL;  and  Kerr,  SA.  Effects  of  alcohol 
and  extended  practice  on  divided  atten- 
tion performance.  Percept  Psychophysics 
48:445-452,  1990. 

Maylor,  E.A.;  Rabbitt,  P.M.A.;  James, 
G.LL;  and  Kerr,  S.A.  Effects  of  alcohol, 
practice,  and  task  complexity  on  reaction 
time  distribution.  QJ  Exp  Psychol  44A-.119- 
139, 1992. 

McFall,  KM.;  Treat,  T.A.;  and  Viken,  RT. 
Contribution  of  cognitive  approaches  to 
studying  clinical  problems.  In:  Routh, 
D.K,  and  Derubeis,  R.J.,  eds.  Clinical 
Psychology  for  the  Next  Century.  Washington, 
DC:  APA  Books,  1998.  pp.  168-197. 

Meier,  S.E.;  Brigham,  T.A.;  Ward,  DA.; 
Myers,  F.;  and  Warren,  L.  Effects  of 
blood  alcohol  concentrations  on  negative 
punishment:  Implications  for  decision 
making.  J  Stud  Alcohol  57:85-96,  1996. 

Melchior,  C.L.;  Glasky,  A.J.;  and  Rtzmann, 
RF.  A  low  dose  of  ethanol  impairs  working 
memory  in  mice  in  a  win-shift  foraging 
paradigm.  Alcohol  10:491-493,  1993. 

Michel,  C.H.,  and  Battig,  K.  Separate  and 
combined  psychophysiological  effects  of 
cigarette  smoking  and  alcohol  consumption. 
Psychopharmacology  97:65-73,  1989. 

Miller,  L.  Neuropsychology  of  the  aggres- 
sive psychopath:  An  integrative  review. 
Aggressive  Behav  13:119-140,  1987. 

Mongrain,  S.,  and  Standing,  L.  Impairment 
of  cognition,  risk-taking,  and  self-perception 
by  alcohol.  Percept  Mot  Skills  69 \\99- 
210,  1989. 


Moskowitz,  H.,  and  DePry,  D.  Differential 
effect  of  alcohol  on  auditory  vigilance  and 
divided  attention  tasks.  QJ  Stud  Alcohol 
29:54-63,  1968. 

Moskowitz,  PL,  and  Murray,  J.T.  Alcohol 
and  backward  masking  of  visual  informa- 
tion. J  Stud  Alcohol  37:40-45,  1976. 

Moskowitz,  PL;  Burns,  M.M.;  and 
Williams,  A.F.  Skills  performance  at  low 
blood  alcohol  levels.  J  Stud  Alcohol 
46:482^85,  1985. 

Mulvihill,  L.E.;  Skilling,  T.A.;  and  Vogel- 
Sprott,  M.  Alcohol  and  the  ability  to 
inhibit  behavior  in  men  and  women.  / 
Stud  Alcohol  58:600-605,  1997. 

Murdoch,  D.D.,  and  Pihl,  RO.  The 
influence  of  beverage  type  on  aggression 
in  males  in  natural  settings.  Aggressive 
Behav  14:325-335,  1988. 

Nagoshi,  C.T.;  Wilson,  J.R;  and 
Rodriguez,  LA.  Impulsivity,  sensation 
seeking,  and  behavioral  and  emotional 
responses  to  alcohol.  Alcohol  Clin  Exp  Res 
15:661-667,  1991. 

Newman,  J. P.;  Widom,  C.S.;  and  Nathan, 
S.  Passive  avoidance  learning  in  syndromes 
of  disinhibition:  Psychopathy  and  extrover- 
sion. /  PersSoc  Psychol  48:1316-1327, 1985. 

Nicholson,  M.E.;  Wang,  M.Q.; 
Airhihenbuwa,  CO.;  Mahoney,  B.S.; 
Christina,  R;  and  Maney,  D.W. 
Variability  in  behavioral  impairment  in- 
volved in  the  rising  and  falling  BAC 
curve.  J  Stud  Alcohol  53:349-365,  1992. 

Nosofsky,  R.M.,  and  Palmeri,  T.J.  Learning 
to  classify  integral-dimension  stimuli. 
Psychonomic  Bull  Rev  3:222-226,  1996. 

Nosofsky,  R.M.,  and  Palmeri,  T.J.  An 
exemplar- based  random  walk  model  of 
speeded  classification.  Psychol  Rev 
104:266-300,  1997. 


354 


Acute  Alcohol  Effects  on  Cognition  and  Impulsive -Disinhibi ted  Behavior 


Oscar-Berman,  M.  Alcohol-related  ERP 
changes  in  cognition.  Alcohol  4:289-292, 
1987. 

Parsons,  O.A.  Neuropsychological  deficits 
in  alcoholics:  Facts  and  fancies.  Alcohol 
Clin  Exp  Res  1:51-56,  1977. 

Parsons,  O.A.,  and  Nixon,  S.J.  Cognitive 
functioning  in  sober  social  drinkers:  A 
review  of  research  since  1986.  J  Stud 
Alcohol  59:180-190,  1998. 

Pearson,  P.,  and  Timney,  B.  Effects  of 
moderate  blood  alcohol  concentrations 
on  spatial  and  temporal  contrast  sensitiv- 
ity. J  Stud  Alcohol  59:163-173,  1998. 

Pernanen,  K.  Alcohol  and  crimes  of  vio- 
lence. In:  Kissin,  B.,  and  Begleiter,  H., 
eds.  The  Biology  of  Alcoholism.  Vol.  4.  New 
York:  Plenum,  1976.  pp.  351^44. 

Peterson,  J.B.,  and  Pihl,  RO.  Information 
processing,  neuropsychological  function, 
and  the  inherited  predisposition  to  alcohol- 
ism. Neuropsychol  Rev  1:343-369,  1990. 

Peterson,  J.B.;  Rothfleisch,  J.;  Zelazo, 
P.D.;  and  Pihl,  R.O.  Acute  alcohol  intoxi- 
cation and  cognitive  functioning.  /  Stud 
Alcohol  51:114-120,  1990. 

Pfefferbaum,  A.;  Horvath,  T.B.;  Roth, 
W.T.;  Clifford,  S.T.;  and  Kopell,  B.S. 
Acute  and  chronic  effects  of  ethanol  on 
event-related  potentials.  In:  Begleiter,  H., 
ed.  Biological  Effects  of  Alcohol.  New  York: 
Plenum,  1980.  pp.  579-587. 

Pihl,  RO.  Alcohol  and  aggression:  A 
psychological  perspective.  In:  Gottheil, 
E.;  Druley,  K.A;  Skoloda,  T.E.;  and 
Waxman,  H.M.,  eds.  Alcohol,  Drug  Abuse 
and  Aggression.  Springfield,  IL:  Charles 
C.  Thomas,  1983.  pp.  292-313. 

Post,  R.B.;  Lott,  L.;  Maddock,  RJ.;  and 
Beede,  J.I.  An  effect  of  alcohol  on  the 


distribution  of  spatial  attention.  J  Stud 
Alcohol  57:260-266,  1996. 

Rohrbaugh,  J.;  Stapleton,  J.M.; 
Parasuraman,  R;  Zubovic,  E.A.;  Frowein, 
H.;  Varner,  J.L.;  Adinoff,  B.;  Lane,  E.A.; 
Eckardt,  M.J.;  and  Linnoila,  M.  Dose- 
related  effects  of  ethanol  on  visual  sus- 
tained attention  and  event-related 
potentials.  Alcohol 4:293-300,  1987. 

Rohrbaugh,  J.;  Stapleton,  J.M.; 
Parasuraman,  R;  Frowein,  PL;  Adinoff, 
B.;  Varner,  J.L.;  Zubovic,  E.A.;  Lane, 
E.A.;  Eckardt,  M.J.;  and  Linnoila,  M. 
Alcohol  intoxication  reduces  visual  sus- 
tained attention.  Psychopharmacology 
96:442^46,  1988. 

Roizen,  J.  Epidemiological  issues  in  alco- 
hol-related violence.  In:  Galanter,  M.,  ed. 
Recent  Developments  in  Alcoholism.  Vol. 
13.  New  York:  Plenum,  1997.  pp.  7-40. 

Salame,  P.  The  effects  of  alcohol  on  learn- 
ing as  a  function  of  drinking  habits. 
Ergonomics  34:1231-1241,  1991. 

Sayette,  M.A.;  Wilson,  G.T.;  and  Elias, 
M.J.  Alcohol  and  aggression:  A  social 
information  processing  analysis.  J  Stud 
Alcohol  54:399-407,  1993. 

Schachar,  RJ.,  and  Logan,  G.D.  Impulsivity 
and  inhibitory  control  in  normal  develop- 
ment and  childhood  psychopathology. 
Dev  Psychol  26:710-720,  1990. 

Schachar,  RJ.;  Tannock,  R;  Marriott, 
M.;  and  Logan,  G.D.  Control  of  inhibitory 
response  processes  in  attention  deficit 
hyperactivity  disorder.  J  Abnorm  Child 
Psychol  23 :411^t37,  1995. 

Schandler,  S.L.;  Cohen,  M.J.;  and  McAithur, 
D.L.  Event-related  brain  potentials  in 
intoxicated  and  detoxified  alcoholics 
during  visuospatial  learning.  Psychopharma- 
cology 94:275-283,  1988a. 


355 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Schandler,  S.L.;  Cohen,  M.J.;  McArthur, 
D.L.;  Naliboff,  B.D.;  and  Hassell,  A. 
Activation  peaking  in  intoxicated  and 
detoxified  alcoholics  during  visuospatial 
learning.  /  Stud  Alcohol  49: 126-1 30, 
1988£. 

Schmauk,  F.J.  Punishment,  arousal,  and 
avoidance  learning  in  sociopaths.  / 
Abnorm  Psychol  76:325-335,  1970. 

Schuckit,  MA.  A  longterm  study  of  sons 
of  alcoholics.  Alcohol  Health  Res  World 
19:172-175,  1995. 

Schuckit,  M.A.;  Gold,  E.O.;  Croot,  K.;  Finn, 
P.;  and  Polich,  J.  P300  latency  after  ethanol 
ingestion  in  sons  of  alcoholics  and  in  con- 
trols. Biol  Psychiatry  24:310-315,  1988. 

Sher,  K.J.;  Walitizer,  K.S.;  Wood,  P.K.; 
and  Brent,  E.E.  Characteristics  of  children 
of  alcoholics:  Putative  risk  factors,  substance 
use  and  abuse  and  psychopathology.  / 
Abnorm  Psychol  100:427-448,  1991. 

Smith,  E.R.;  Fazio,  R.H.;  and  Cejk,  M.D. 
Accessible  attitudes  influence  categorization 
of  multiply  categorizable  objects.  J  Pers 
Soc  Psychol  71:888-898,  1996. 

Sommer,  W.;  Leuthold,  PL;  and  Hermanutz, 
M.  Covert  effects  of  alcohol  revealed  by 
event-related  potentials.  Percept  Psycho- 
physics  54:127-135,  1993. 

Stewart,  M.A.,  and  Wilcox,  J.A.  Childhood 
antecedents  of  alcoholism.  Adv  Dev  Behav 
Pedtatr  6:151-175,  1985. 

Stokes,  A.F.;  Belger,  A.;  Banich,  M.T.; 
and  Taylor,  H.  Effects  of  acute  aspartame 


and  acute  alcohol  ingestion  upon  the 
cognitive  performance  of  pilots.  Aviat 
Space  Environ  Med  62:648-653,  1991. 

Teo,  RK.C,  and  Ferguson,  DA.  The  acute 
effects  of  ethanol  on  auditory  event-related 
potentials.  Psychopharmacology  90:179- 
184,1986. 

Virkkunen,  M.,  and  Linnoila,  M.  Serotonin 
in  early  onset,  male  alcoholics  with  violent 
behavior.  Ann  Med  22:327-331,  1990. 

Virkkunen,  M.,  and  Linnoila,  M.  Brain 
serotonin,  Type  II  alcoholism  and  impulsive 
violence.  /  Stud  Alcohol  Suppl  1 1 :  163- 
169,  1993. 

Wang,  M.Q.;  Nicholson,  M.E.; 
Airhihenbuwa,  CO.;  and  Mahoney,  B.S. 
Psychomotor  and  visual  performance 
under  the  time-course  effect  of  alcohol. 
Percept  Mot  Skills  75:1095-1106,  1992. 

White,  A.H.;  Simson,  P.E.;  and  Best,  P.J. 
Comparison  between  the  effects  of  ethanol 
and  diazepam  in  spatial  working  memory 
in  the  rat.  Psychopharmacology  (Berl) 
133:256-261,  1997. 

White,  H.R.;  Brick,  J.;  and  Hansell,  S.  A 
longitudinal  investigation  of  alcohol  use 
and  aggression  in  adolescence.  J  Stud  Alcohol 
Suppl  11:62-77,  1993. 

White,  J.L.;  Moffitt,  T.E.;  Caspi,  A.; 
Bartusch,  D.J.;  Needles,  D.J.;  and 
Stouthamer-Loeber,  M.  Measuring  impul- 
sivity  and  examining  its  relationship  to 
delinquency.  J  Abnorm  Psychol  103:192- 
205,  1994. 


356 


Chapter  10 

Clinical  Neuroscience  Studies  of 

Behaviors  Associated  With  Alcohol 

Consumption  in  Alcoholism 


John  H.  Krystal,  M.D.,  Ismene  L.  Petrakis,  M.D., 
Louis  Trevisan,  M.D.,  and  Neill  Epperson,  M.D. 

KEY  WORDS:  AOD  (alcohol  or  other  drug)  use  behavior;  AODE  (effects  ofAOD 
use,  abuse,  and  dependence);  psycho-pharmacological  therapy;  serotonin  receptors; 
agonists;  serotonin  uptake  inhibitors;  antagonists;  GABA  receptors;  NMDA  re- 
ceptors; glutamate;  opiates;  dopaminergic  receptors;  catecholamines;  calcium 
acetylhomotaurinate;  steroid  hormones;  tryptophan;  ketamine;  literature  review 

The  past  decade  has  been  an  era  of  and  craving.  In  most  cases,  preclinical 
important  advances  in  clinical  neuro-  research  has  influenced  the  design  and 
science  and  psychopharmacological  interpretation  of  subsequent  clinical 
studies  of  alcoholism.  These  studies  have  research.  However,  clinical  research 
developed  along  parallel  tracks,  with  data  have  often  provided  novel  insights 
groups  studying  the  contributions  of  that  reshape  the  expectations  based  on 
many  neurotransmitter  systems  to  data  collected  in  other  species, 
clinical  phenomena  associated  with  The  purpose  of  this  chapter  is  to  pro- 
alcoholism,  intoxication,  consumption,  vide  an  overview  of  clinical  neuroscience 

J.H.  Krystal,  M.D.,  is  a  professor  and  director  of  the  Division  of  Cognitive  and  Clinical  Neuroscience, 
Department  of  Psychiatry,  Tale  University  School  of  Medicine,  New  Haven,  CT 06510;  deputy 
director  of  the  Abraham  Ribicoff  Research  Facilities,  Connecticut  Mental  Health  Center,  34  Park  St., 
New  Haven,  CT  06519;  and  director  of  the  VA-Tale  Alcoholism  Research  Center,  Psychiatry  Service 
(116-A),  VA  Connecticut  Healthcare  System,  950  Campbell  Ave.,  West  Haven,  CT  06516.  I.L. 
Petrakis,  M.D.,  is  an  associate  professor  in  the  Department  of  Psychiatry,  Tale  University  School  of 
Medicine,  and  director  of  substance  abuse  treatment  at  the  Psychiatry  Service  of  the  VA  Connecticut 
Healthcare  System  and  the  VA-Tale  Alcoholism  Research  Center,  Psychiatry  Service.  L.  Trevisan, 
M.D.,  is  an  assistant  clinical  professor  in  the  Department  of  Psychiatry,  Tale  University  School  of 
Medicine;  staff  psychiatrist  at  the  Psychiatry  Service  of  the  VA  Connecticut  Healthcare  System;  and  an 
investigator  in  the  VA-Tale  Alcoholism  Research  Center.  N.  Epperson,  M.D.,  is  an  assistant  professor 
in  the  Department  of  Psychiatry,  Tale  University  School  of  Medicine,  and  director  of  the  Behavioral 
Gynecology  Program,  Abraham  Ribicoff  Research  Facilities. 

357 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


approaches  to  understanding  behaviors 
associated  with  alcoholism.  We  will  focus 
on  several  neurotransmitter  systems  that 
have  been  implicated  in  alcoholism 
and  its  treatment:  serotonin  (5-HT), 
glutamate,  gamma-aminobutyric  acid 
(GABA),  endogenous  opiate,  and 
dopamine.  However,  there  are  a  number 
of  limitations  of  this  review  and  the  field 
of  research  that  it  considers.  These  neu- 
rotransmitter systems  have  a  compli- 
cated matrix  of  interactions  at  multiple 
sites  in  the  brain.  The  diversity  of  mech- 
anisms to  be  considered  interacts  with 
the  clinical  complexity  of  alcoholism. 
Because  of  space  limitations,  the  inter- 
face of  neurotransmitter  systems  in 
contributing  to  alcoholism-related 
behaviors  will  receive  relatively  superficial 
attention  in  this  chapter. 

SEROTONERGIC  SYSTEMS 

Serotonergic  systems  have  received  the 
greatest  extent  of  psychopharmaco- 
logical  study  in  humans.  As  a  result, 
this  section  is  organized  by  type  of 
agent  studied  rather  than  by  the 
behavior  of  interest. 

Serotonin  Agonists 

Serotonergic  systems  appear  to  contri- 
bute to  the  discriminative  properties  of 
ethanol  in  humans.  Ethanol  appears  to 
facilitate  that  activity  of  a  number  of  5- 
HT  receptors,  including  the  5-HT1B, 
5-HT2c,  and  5-HT3  receptors,  and  to 
share  discriminative  stimulus  properties 
with  drugs  acting  at  these  sites  (Lovinger 
1991;  Grant  et  al.  1997b).  Although 
these  studies  have  guided  clinical 
research,  they  may  not  be  directly 
applicable  to  humans  because  the 


human  5-HT1B  receptor,  previously 
called  the  5-HT1D  receptor,  has  very  dif- 
ferent properties  from  the  rodent  5- 
HT1B  receptor  (Pauwels  et  al.  1996). 
At  first  glance,  the  clinical  findings 
appear  to  be  quite  similar  to  the  pre- 
clinical data.  However,  the  human 
data  may  differ  from  the  preclinical 
data  in  the  receptor  sites  mediating 
the  actions  of  agents  that  have  been 
studied  in  both  animals  and  humans. 
In  particular,  5-HT2  receptor  subtypes 
may  contribute  to  the  ethanol-like 
effects  of  m-chlorophenylpiperazine 
(mCPP)  in  humans  (Krystal  et  al. 
1994).  In  contrast,  5-HT2  receptors 
do  not  appear  to  figure  prominently  in 
the  ethanol-like  effects  of  piperazine 
5-HT  agonists,  including  mCPP,  in 
animals  (Grant  and  Barrett  1991). 

The  administration  of  piperazine  5- 
HT  partial  agonists  produces  a  euphoric 
effect  that  is  perceived  as  ethanol-like  in 
early-onset  alcoholic  patients  (Buydens- 
Branchey  et  al.  1989;  Benkelfat  et  al. 
1991;  Lee  and  Meltzer  1991;  Krystal 
et  al.  1994;  Buydens-Branchey  et  al. 
19976;  George  et  al.  1997).  How- 
ever, the  effects  of  the  partial  5-HT 
agonist  mCPP  were  not  specifically 
similar  to  ethanol;  that  is,  the  effects 
were  similar  to  several  substances  of 
abuse  (figure  1). 

The  two  mCPP  studies  that  adminis- 
tered mCPP  intravenously  also  reported 
the  induction  of  craving  (Krystal  et  al. 
1994;  George  et  al.  1997),  whereas 
the  study  administering  this  drug 
orally  found  the  opposite  (Buydens- 
Branchey  et  al.  1997 b).  The  induction 
of  craving  by  intravenous  mCPP  sug- 
gests that  the  similarity  between  the 
subjective  effects  of  mCPP  and 


358 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


30    SO    »   120   150   180   210   240 


Time  (min) 


fc   ?4, 

E 

o 

V  21  • 

A 

C   18  J 

« 

** 

O   15  - 

r1 

.** 

£"  12  - 

i 
/ 

\ 

e   9 

1 

\ 

(0 

S     6- 

1 1 

\ 

** 

a 

/i\    "\ 

K 

;">\              \ 

/           \                   T              \ 

■5    J, 

J/' 

i    \ 

0 

-jL 

5-0=^=5=* 

^-r-f^L-P  ,    ;■ 

90       120       150      180      210      240 


Time  (min) 


Time  (min) 


Figure  1.  Similarity  of  m-chlorophenylpiperazine  (mCPP)  and  yohimbine  effects  to  ethanol 
(left;  n  =  21),  marijuana  (middle;  n  =  18),  and  cocaine  (right;  n  =  14).  Placebo  (open  circles), 
mCPP  (filled  circles),  and  yohimbine  (open  squares)  effects  are  presented.  Each  value  repre- 
sents a  mean  ±  SEM.  Increases  from  baseline  were  evaluated  using  the  Dunnett  t  test,  *  p  < 
0.05,  **  p  <  0.01.  The  drug  x  drug  of  abuse  x  time  interaction  was  not  significant.  However, 
the  drug  x  time  interactions  were  significant  for  ethanol  (p  =  0.01)  and  marijuana  (p  =  0.04). 
The  drug  x  time  interaction  for  similarity  to  cocaine  was  not  significant  (p  =  0.1).  Adjusting 
for  placebo  day  responses,  all  significant  increases  from  baseline  retained  their  level  of  signifi- 
cance except  one  value,  #,  p  <  0.05  on  the  yohimbine  day,  nonsignificant  after  adjusting  for 
placebo  response.  Reprinted  with  permission  from  Krystal,  J.H.;  Webb,  E.;  Cooney,  N.; 
Kranzler,  H.R.;  and  Charney,  D.S.  Specificity  of  ethanollike  effects  elicited  by  serotonergic  and 
noradrenergic  mechanisms.  Archives  of  General  Psychiatry  51:898-911,  November  1994. 
Copyright  1994,  American  Medical  Association. 


359 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ethanol  is  significant  clinically.  In  addi- 
tion to  the  elicitation  of  craving,  mCPP 
also  produced  anxiety  and  irritability 
(Krystal  et  al.  1994;  Handelsman  et  al. 
1996).  Thus,  the  induction  of  dysphoria 
by  this  drug  may  have  contributed  to 
the  elicitation  of  craving. 

The  response  to  mCPP  and  other 
direct  and  indirect  5 -HT  agonists  dis- 
tinguishes early-onset  alcoholic  patients 
(type  II)  from  late-onset  alcoholic 
patients  and  healthy  control  subjects. 
Several  studies  report  that  the  Cortisol  or 
prolactin  response  to  mCPP,  MK- 
212,  5 -hydroxy try tophan,  or  fenflu- 
ramine is  reduced  in  early-onset 
alcoholic  patients  (Lee  and  Meltzer 
1991;  Balldin  et  al.  1992;  Coccaro  et 
al.  1994;  Farren  et  al.  1995 £;  Han- 
delsman et  al.  1996;  Krystal  et  al. 
1996;  Buydens-Branchey  et  al.  1997b; 
George  et  al.  1997).  Furthermore,  the 
cerebral  metabolic  response  to  mCPP, 
assessed  using  the  positron  emission 
tomography  (PET)  flurodeoxy glucose 
technique,  was  reduced  in  early-onset 
alcoholics  (Hommer  et  al.  1997).  In 
contrast,  the  euphoric  responses  to 
mCPP  appeared  to  be  particularly 
enhanced  in  early-onset  alcoholic 
patients  (George  et  al.  1997). 

The  capacity  to  distinguish  subtypes 
of  alcoholic  patients  by  their  response 
to  5-HT  agonists  may  have  important 
clinical  implications.  First,  mCPP  may 
provide  a  tool  for  studying  the  constel- 
lation of  problems  associated  with  type 
II  alcoholism,  including  familial  addic- 
tion, early  onset,  severe  course,  and 
sociopathy  (Cloninger  1987;  Babor  et 
al.  1992).  Early-onset  alcoholic 
patients  appear  to  have  other  seroton- 
ergic abnormalities,  particularly  lower 


central  5-HT  turnover,  that  may  contri- 
bute to  their  expression  of  sociopathic 
or  impulsive  behaviors  (Virkkunen  et 
al.  1995;  Virkkunen  and  Linnoila 
1997).  If  related  to  reduced  central  5- 
HT  turnover  in  these  patients,  phar- 
macological challenge  with  mCPP  may 
provide  a  phenotypic  marker  of  trypto- 
phan hydroxylase  alleles  associated  with 
risk  for  impulsive  behavior  (Nielsen  et 
al.  1994).  Second,  worsening  of  crav- 
ing in  response  to  mCPP  may  predict 
poor  response  to  particular  antidipso- 
tropic  medications,  such  as  fluoxetine, 
that  may  stimulate  5-HT  receptors 
(Kranzler  et  al.  1996).  A  corollary  of 
this  hypothesis  is  that  mCPP  stimulation 
of  craving  may  help  to  predict  beneficial 
therapeutic  effects  of  drugs  that  block 
mCPP  effects.  Third,  the  actions  of 
mCPP  may  be  particularly  relevant  to 
early-onset  poly  drug  abuse.  Supporting 
this  view,  mCPP  effects  are  similar  to 
several  drugs  of  abuse  (Krystal  et  al. 
1994)  and  mCPP  effects  in  patients 
with  an  early  onset  of  cocaine  addic- 
tion parallel  the  findings  in  early-onset 
alcoholism  (Buydens-Branchey  et  al. 
19970,  1997c). 

However,  the  site  of  action  of  the 
ethanol-like  effects  of  mCPP  is  not 
clear.  The  piperazine  serotonin  agonists 
are  notoriously  nonselective  for  5-HT 
receptor  subtypes.  The  most  potent 
actions  of  mCPP  are  partial  agonism 
of  the  5-HT2c  site  and  antagonism  of 
the  5-HT3  site  (Conn  and  Sanders- 
Bush  1987;  Robertson  et  al.  1992).  Of 
these  effects,  the  5-HT2c  agonist  action 
appears  to  figure  most  prominently  in 
its  general  discriminative  stimulus  effects 
(Callahan  and  Cunningham  1994). 
mCPP  also  has  activity  at  the  5-HT7 


360 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


receptor  (To  et  al.  1995).  Preliminary 
data  suggested  that  ritanserin,  a  drug 
that  blocks  several  serotonin  receptors 
(5-HT2A,  5-HT2C,  5-HT6,  5-HT7,  5- 
HT1D)  and  the  dopamine-2  (D2)  recep- 
tor, reduced  some  effects  of  mCPP  in 
healthy  humans  (Conn  and  Sanders- 
Bush  1987;  Seibyl  et  al.  1991;  Shi  et 
al.  1995;  To  et  al.  1995;  Pauwels  et 
al.  1996;  Boess  et  al.  1997).  The  lack 
of  specificity  regarding  the  site  of  action 
of  both  mCPP  and  ritanserin  limits 
the  interpretation  of  the  mechanisms 
underlying  the  ethanol-like  and  craving 
effects  of  mCPP. 

The  lack  of  specificity  of  both  5-HT 
agonists  and  antagonists  available  for 
clinical  research  has  had  important 
consequences.  Despite  preliminary 
promise  (Meert  1994),  subsequent 
larger  trials  failed  to  find  that  ritanserin 
was  efficacious  for  alcoholism  or  heavy 
social  drinking  (Naranjo  et  al.  1995; 
Johnson  et  al.  1996).  Similarly,  ritan- 
serin did  not  substantially  modulate 
ethanol  intoxication  in  the  laboratory 
(Estevez  et  al.  1995).  Given  the  limi- 
tations in  the  specificity  of  ritanserin, 
it  is  difficult  to  know  whether  this  lack 
of  efficacy  arises  from  shortcomings  in 
the  selection  of  early-onset  alcoholism 
patients,  the  dose  of  ritanserin  employed, 
the  selection  of  ritanserin  as  a  5-HT2c 
antagonist,  or  the  expectation  that  5- 
HT2C  antagonists  would  be  effective. 

Other  drugs  that  stimulate  receptor 
subtypes  from  the  5-HTi  receptor 
family  have  been  studied  in  alcoholic 
patients.  One  study  reported  a  reduc- 
tion in  the  human  growth  hormone 
response  to  sumatriptan,  a  human  5- 
HT1B  agonist,  in  recently  detoxified 
alcoholic  patients  (Vescovi  and  Coiro 


1997).  Buspirone  is  a  drug  with  5-HT1A 
agonist  and  D2  antagonist  activity.  Its 
major  metabolite,  l-(2-pyrimidinyl)- 
piperazine  (1-PP),  is  also  a  potent  a2 
adrenoceptor  antagonist.  In  animals, 
5-HT1A  agonists  have  had  mixed 
effects  on  ethanol  self- administration 
(Tomkins  et  al.  1994).  Acutely,  bus- 
pirone  appears  to  have  no  ethanol-like 
effects  (Griffith  et  al.  1986).  With 
chronic  administration,  it  appears  to 
produce  modest  beneficial  effects  on 
anxiety,  alcohol  craving,  or  alcohol 
consumption  in  anxious  alcoholic 
patients  (Bruno  1989;  Malcolm  et  al. 
1992;  Tollefson  et  al.  1992;  Kranzler 
et  al.  1994;  Make  et  al.  1996). 

5-HT  Transporter 
Antagonists 

Insights  into  alcoholism  have  been  pro- 
vided by  5-HT  transporter  antagonists 
(serotonin  reuptake  blockers).  One 
line  of  interest  has  built  on  evidence 
that  deficits  in  central  5-HT  turnover 
may  contribute  to  ethanol  consump- 
tion in  animals  and  humans.  This  line 
of  research  has  hypothesized  that  the 
serotonergic  antidepressants  would 
increase  synaptic  availability  of  5-HT 
and  result  in  reduced  ethanol  con- 
sumption. This  view  builds  on  the 
parallels  between  reductions  in  cere- 
brospinal fluid  (CSF)  5-hydroxyin- 
doleacetic  acid  (5-HIAA)  in  depression 
and  early-onset  alcoholism.  Molecular 
genetic  studies  further  increased  interest 
in  genetic  variation  associated  with  the 
function  of  the  5-HT  transporter.  Al- 
though the  findings  have  not  been  repli- 
cated (Nielsen  et  al.  1996;  Gelernter 
et  al.  1997),  two  groups  have  associated 
5-HT  transporter  alleles  with  reduced 


361 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


central  5-HT  function  or  poor  impulse 
control  in  alcoholic  individuals 
(Nielsen  et  al.  1994;  Sander  et  al. 
1995).  However,  one  study  failed  to 
find  that  alleles  of  the  5-HT  trans- 
porter were  associated  with  alcoholism 
(Gelernter  et  al.  1997). 

One  hypothesis  guiding  these  studies 
was  that  reduction  in  the  efficacy  or 
availability  of  synaptic  5-HT  resulting 
from  enhanced  density  or  function  of 
the  5-HT  transporter  would  contribute 
to  the  constellation  of  behaviors  asso- 
ciated with  early-onset  alcoholism 
(Daoust  et  al.  1991;  Rausch  et  al. 
1991;  Mellerup  et  al.  1992;  Faraj  et 
al.  1997).  Alternatively,  reduced  den- 
sity of  5-HT  transporter  binding  in  the 
brain  might  reflect  reductions  in  the 
density  of  5-HT  terminals  that  might 
contribute  to  reduced  central  5-HT 
function  (Chen  et  al.  1991;  Tiihonen 
et  al.  1997).  Reductions  in  5-HT 
nerve  terminal  density  might  also  par- 
allel immunohistochemical  evidence 
of  reductions  in  the  number  of  pontine 
serotonergic  cell  bodies  (Halliday  et 
al.  1993). 

5-HT  transporter  antagonists  have 
shown  utility  in  the  treatment  of 
alcoholics  with  comorbid  psychiatric 
disorders.  Acute  administration  of 
clomipramine  did  not  have  ethanol- 
like  effects  in  alcoholic  patients  with 
comorbid  panic  disorder,  although 
this  drug  made  these  patients  acutely 
anxious  (George  et  al.  1995).  Other 
studies  have  suggested  that  5-HT 
transporter  antagonists  have  limited 
efficacy  for  alcoholism  and  may  make 
early-onset  patients  worse  (Naranjo 
and  Bremner  1993;  Naranjo  et  al. 
1994;  Kranzler  et  al.  1996;  Pettinati 


1996).  However,  subgroups  of  patients 
with  comorbid  depression  may  benefit 
from  treatment  with  5-HT  transporter 
antagonists  (Griffith  et  al.  1986; 
Bruno  1989;  Tollefson  et  al.  1992). 

5-HT3  Antagonists 

Preclinical  research  suggests  that  ethanol 
stimulates  5-HT3  receptors  and  that  5- 
HT3  antagonists  may  attenuate  the 
discriminative  stimulus  properties  of 
ethanol  and  ethanol  self- administration 
in  animals  (Grant  and  Barrett  1991; 
Tomkins  et  al.  1995).  The  capacity  of 
5-HT3  antagonists  to  reduce  the  stim- 
ulation of  limbic  dopamine  systems  by 
drugs  of  abuse,  including  ethanol,  may 
contribute  to  this  effect  (Grant  1995). 
In  humans,  the  5-HT3  antagonist 
ondansetron  did  not  robustly  attenuate 
the  discriminative  stimulus  effects  of 
ethanol  (Swift  et  al.  1996).  A  clinical 
trial  employing  ondansetron  found 
some  evidence  of  potential  usefulness 
of  this  drug  (Sellers  et  al.  1994). 

Modulation  of  5-HT 
Precursor  Availability 

There  is  a  long  history  of  interest  in 
the  impact  of  5-HT  precursor  availabil- 
ity on  clinical  phenomena  related  to 
alcoholism.  This  interest  arises  from  the 
fact  that  a  5-HT  precursor,  tryptophan, 
is  an  essential  amino  acid  and  that  the 
hydroxylation  of  tryptophan  is  the  rate- 
limiting  step  in  5-HT  synthesis  (Fern- 
strom  1983;  Fernstrom  and  Fernstrom 
1995).  As  a  result  there  is  a  direct  rela- 
tionship between  profound  acute 
changes  in  tryptophan  availability  and 
reductions  in  brain  5-HT  synthesis. 
Under  chronic  conditions,  this  relation- 
ship is  altered  as  protein  catabolism  and 


362 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


other  adaptive  changes  can  compensate 
to  varying  degrees  for  reductions  in 
plasma  free  tryptophan. 

There  are  conflicting  data  regarding 
tryptophan  levels  in  alcoholism.  Reports 
suggest  that  central  and  peripheral  levels 
of  tryptophan  are  increased  in  recently 
detoxified  alcoholics  (Beck  et  al.  1980, 
1983;  Farren  and  Dinan  1996).  Other 
studies  suggest  that  plasma  tryptophan, 
its  ratio  to  other  neutral  amino  acids  that 
compete  with  it  for  transport  into  the 
brain,  or  other  indices  of  its  transport 
into  the  central  nervous  system  are 
reduced  in  recently  detoxified  alco- 
holics (Branchey  et  al.  1981;  Branchey 
et  al.  1985;  Friedman  et  al.  1988; 
Buydens-Branchey  et  al.  1989;  Roy  et 
al.  1990).  Although  a  component  of 
acute  tryptophan  reductions  may  be 
related  to  malnutrition,  the  primary 
effect  thought  to  account  for  reductions 
in  plasma  tryptophan  in  recovering 
alcoholics  is  enhancement  of  hepatic 
tryptophan  pyrrolase  (Badawy  1996). 

The  clinical  significance  of  changes 
in  plasma  tryptophan  is  not  clear.  Two 
studies  suggest  that  reductions  in  tryp- 
tophan availability  to  the  brain  might 
be  related  to  memory  impairments  or 
clinical  characteristics  associated  with 
early-onset  alcoholism  (Branchey  et 
al.  1985;  Buydens-Branchey  et  al. 
1989).  However,  it  is  possible  that 
this  relationship  is  mediated  by  the 
association  of  greater  ethanol  consump- 
tion with  both  memory  impairments 
and  early- onset  alcoholism. 

Clinically  significant  reductions  in 
plasma  tryptophan  may  be  achieved 
using  a  tryptophan-free  amino  acid  drink. 
This  technique  was  developed  after  it 
became  evident  that  substantial 


dietary  tryptophan  restrictions  had  no 
discernible  behavioral  effects  in  humans 
(Delgado  et  al.  1989).  The  amino 
acid  drink  stimulates  the  use  of  plasma 
free  tryptophan  for  protein  synthesis 
and  can  reduce  levels  by  over  80  percent 
(Young  et  al.  1985,  1988;  Delgado  et 
al.  1989,  1990).  The  amino  acid  drink 
reduces  CSF  levels  of  tryptophan  and 
5-HIAA  in  healthy  individuals 
(Carpenter  et  al.  1998).  In  contrast  to 
dietary  tryptophan  restriction,  this  level 
of  tryptophan  depletion  appears  to 
modulate  mood — most  notably,  tran- 
siently reversing  the  antidepressant 
effects  of  serotonergic  antidepressant 
treatment  in  depressed  patients.  Tryp- 
tophan depletion  also  reduced  cue- 
induced  cocaine  craving  (Satel  et  al. 
1995).  However,  it  is  not  yet  clear 
whether  tryptophan  depletion  produces 
a  clinically  significant  reduction  in 
alcohol  cue-induced  craving  in  sober 
alcoholic  patients  (Petrakiset  al.  1995). 
Previous  studies  also  failed  to  show 
effects  of  modulation  of  plasma  trypto- 
phan levels  on  ethanol  intoxication  or 
ethanol  self- administration  in  popula- 
tions other  than  early- onset  alcoholics 
(Pihl  et  al.  1987;  Zacchia  et  al.  1987). 
Tryptophan  depletion  has  also  been 
reported  to  disinhibit  or  even  promote 
aggression  in  normal  males  (Pihl  et  al. 
1995).  However,  this  effect  has  not  been 
replicated  in  patients  with  aggression- 
related  problems  (Young  et  al.  1988; 
Salomon  et  al.  1994).  Together,  these 
data  suggest  that  profound  alterations 
in  the  availability  may  have  some 
influence  on  mood  and  behaviors 
associated  with  alcoholism.  However, 
these  studies  question  the  clinical  signif- 
icance of  the  relatively  modest  changes 


363 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


in  the  levels  of  plasma  free  tryptophan 
in  recovering  alcoholic  patients. 

5-HT  Overview 

A  number  of  clinical  research  strategies 
have  been  used  to  study  the  contribu- 
tions of  5-HT  systems  to  behaviors 
associated  with  alcoholism.  As  has 
been  suggested  previously  (Virkkunen 
and  Linnoila  1997),  clinical  neuro- 
science research  studies  link  alterations 
in  5-HT  function  to  problems  with 
impulse  control  that  are  most  closely 
associated  with  the  early-onset  subtype 
of  alcoholism.  Regarding  addictive 
behaviors,  the  strongest  finding  is  that 
stimulation  of  population(s)  of  5-HT 
receptors,  as  of  yet  not  conclusively 
identified,  by  mCPP  results  in  ethanol- 
like  interceptive  cues  that  may  contribute 
to  craving.  The  genetic  underpinnings 
of  mCPP  response  may  provide  insights 
into  early-onset  alcoholism.  Further- 
more, drugs  that  effectively  antagonize 
the  ethanol-like  components  of  mCPP  in 
humans  should  be  evaluated  as  potential 
pharmacotherapies  for  alcoholism. 

GABA  SYSTEMS 

Contribution  of  GABAa 
Receptors  to  the 
Discriminative  Stimulus 
Effects  of  Ethanol 

GABA  systems,  particularly  in  limbic 
and  cortical  structures,  have  been 
implicated  in  the  rewarding  effects  of 
ethanol  administration  (Koob  et  al. 
1994).  Ethanol  facilitates  GABAA 
receptor  function  (Suzdak  et  al.  1988b). 
Consistent  with  this  view,  the  discrim- 
inative stimulus  effects  of  ethanol  in 


animals  are  shared  among  drugs  that 
facilitate  GABAA  receptor  function, 
particularly  the  barbiturates  and  benzo- 
diazepines (Ticku  et  al.  1992;  Grant 
and  Colombo  1993b).  In  particular, 
these  GABAA  receptor-facilitating 
drugs  appear  to  substitute  particularly 
well  for  the  discriminative  stimulus 
effects  of  low  to  moderate  amounts  of 
ethanol  consumption  (Grant  and 
Colombo  1993a,  1993b). 

The  five  GABAA  receptor  subunits 
may  combine  in  multiple  configura- 
tions. Particular  attention  has  focused 
on  GABAA  receptors  bearing  the  al5 
a6,  and  y2  subunits.  The  ax  subunit  is 
distinctive  in  that  it  is  selective  for 
sites  above  the  spinal  cord  (Luddens 
and  Korpi  1995).  As  a  result,  drugs 
that  are  selective  for  GABAA  receptors 
bearing  the  aj  subunit,  such  as  the 
hypnotic  Zolpidem,  do  not  possess  the 
full  range  of  motor,  muscle-relaxing, 
and  perhaps  autonomic  effects  associated 
with  drugs  that  nonselectively  facilitate 
GABAA  receptor  function.  Limitations 
in  the  ability  of  Zolpidem  to  substitute 
for  ethanol  in  drug  discrimination  para- 
digms suggest  that  ethanol  actions  at 
the  GABAA  receptors  that  do  not  bear 
the  a:  subunit  contribute  to  the  dis- 
criminative stimulus  effects  of  ethanol 
(Bienkowski  et  al.  1997).  However,  it 
is  not  yet  clear  whether  the  non-o^ 
actions  contribute  significantly  to 
ethanol  self- administration  in  animals 
with  inherited  propensities  for  ethanol 
consumption.  The  a6  subunit  is 
expressed  in  the  cerebellum,  where  it 
may  contribute  to  the  ataxic  effects  of 
ethanol  (Luddens  and  Korpi  1995). 
This  site  received  significant  attention 
when  it  was  found  that  a  (3-carboline 


364 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


drug  with  benzodiazepine  inverse  ago- 
nist properties,  Ro  15-4513,  reduced 
some  ethanol  effects  and  ethanol  self- 
administration  via  this  site  (Suzdak  et 
al.  1988a;  June  et  al.  1994). 

GABA  Systems 

and  Alcoholism  Vulnerability 

In  humans,  GABAA  receptors  have 
been  implicated  in  the  vulnerability  to 
alcohol  consumption.  In  particular, 
reduced  sensitivity  to  the  facilitatory 
effects  of  ethanol  on  GABAA  receptor 
function,  particularly  in  neural  systems 
mediating  adverse  cognitive  or  motor 
effects,  may  predispose  vulnerable 
individuals  to  consume  more  alcohol. 
Individuals  at  risk  for  alcoholism  may 
not  receive  the  expected  dysphoric 
interoceptive  feedback  signals  to  stop 
drinking  until  they  achieve  higher 
blood  ethanol  levels  than  do  individuals 
at  lower  risk  for  alcoholism  (Schuckit 
and  Smith  1997). 

To  date,  no  specific  gene  has  been 
clearly  implicated  in  a  GABA-related 
vulnerability  for  alcoholism.  A  haplotype 
relative-risk  study  did  not  find  evidence 
associating  either  the  a:  or  a3  subunit 
genes  with  alcoholism,  although  sug- 
gestive data  supporting  further  study 
were  obtained  for  the  latter  gene  in  an 
association  study  (Parsian  and  Cloninger 
1997).  The  strongest  tie  between 
GABAA  receptor  involvement  and  the 
vulnerability  to  alcoholism  has  come 
from  studies  evaluating  ethanol  and 
benzodiazepine  effects  in  populations 
at  high  risk  for  developing  alcoholism. 
Most  (Cowley  et  al.  1994;  Schuckit 
1994;  Schuckit  and  Smith  1996; 
Schuckit  et  al.  1996,  1997),  but  not 
all  (de  Wit  and  McCracken  1990),  of 


these  studies  found  that  male  offspring 
of  alcoholics  have  reduced  sensitivity  to 
the  cognitive,  behavioral,  and  motor 
effects  of  both  benzodiazepines  and 
ethanol.  Similarly,  individuals  at  risk 
for  alcoholism  exhibited  blunted  cere- 
bellar metabolic  inhibition,  but  not 
cortical  metabolic  inhibition,  follow- 
ing a  dose  of  lorazepam  (Volkow  et  al. 
1995).  However,  there  may  be  greater 
sensitivity  to  or  preference  for  the 
euphoric  effects  of  benzodiazepines  in 
this  population  (Cowley  et  al.  1992, 
1994),  although  these  effects  are  not 
uniformly  replicated  (de  Wit  1991). 
The  rewarding  effects  of  ethanol  and 
benzodiazepines  in  humans  may  be 
increased  in  anxious  individuals,  who 
tend  to  experience  more  pronounced 
anxiolytic  effects  of  these  drugs 
(Chutuape  and  de  Wit  1995). 

Clinical  Evidence  of  GABA 
Dysregulation  in  Alcoholism 

Several  clinical  studies  suggest  that  the 
regulation  of  brain  GABA  systems 
may  differ  in  alcoholic  and  nonalcoholic 
populations.  To  date  there  has  been 
very  little  direct  study  of  the  potential 
contributions  of  dysregulation  of  cor- 
tical GABA  systems  to  behaviors  asso- 
ciated with  alcohol  consumption  in 
alcoholic  patients.  Thus,  a  relatively 
large  group  of  hypotheses  remain  to 
be  tested  from  a  clinical  perspective: 

( 1 )  that  reductions  in  GABAA  receptor 
density  (related  to  changes  in  the 
expression  of  particular  GABAA  receptor 
subunits)  or  function  convey  a  vulner- 
ability to  greater  alcohol  consumption, 

(2)  that  reductions  in  GABA  release 
combined  with  reductions  in  GABAA 
receptor  density  convey  a  vulnerability 


365 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


to  acute  and  protracted  ethanol  with- 
drawal symptoms  that  might  contribute 
to  alcohol  craving  and  relapse,  and  (3) 
that  normalization  (i.e.,  treatment)  of 
abnormalities  in  GABA  function  in 
alcoholic  patients  would  reduce  the 
drive  to  consume  alcohol.  By  implica- 
tion, the  genes  that  might  modulate 
each  component  of  these  pathophysi- 
ological hypotheses  would  be  impli- 
cated in  the  vulnerability  to  alcoholism 
and  its  treatment.  The  current  research 
database  provides  some  support  for  these 
hypotheses.  However,  it  is  quite  limited 
in  its  capacity  to  determine  the  extent  to 
which  current  findings  are  explained 
by  genetic  factors,  specific  pharmacolog- 
ical effects  of  ethanol  on  brain  GABA 
systems,  and  other  effects  of  ethanol 
on  the  brain,  including  neurotoxicity. 
There  are  no  studies  that  directly 
evaluate  abnormalities  in  brain  GABA 
turnover  in  alcoholism  or  in  vulnerable 
populations.  Indirect  evidence  of 
GABAergic  abnormalities  comes  from 
measurements  of  GABA  levels  in  the 
plasma,  CSF,  and  brain.  In  offspring 
of  alcoholics  there  are  conflicting 
reports  that  suggest  that  plasma  GABA 
may  be  low  (Moss  et  al.  1990),  elevated 
(Garbutt  et  al.  1995),  or  unchanged 
(Cowley  et  al.  1996).  Ethanol  has  been 
reported  to  elevate  plasma  GABA  levels 
in  high-risk  populations  with  low 
baseline  levels  (Moss  et  al.  1990).  In 
contrast,  in  a  group  of  high-risk  offspring 
who  did  not  show  baseline  reductions, 
benzodiazepines  had  no  effects  (Cowley 
et  al.  1996).  Plasma  GABA  in  this  high- 
risk  population  may  correlate  with  behav- 
ior traits,  such  as  novelty  seeking,  that 
are  also  associated  with  vulnerability 
to  alcoholism  (Cowley  et  al.  1996). 


GABA  levels  appear  to  be  reduced 
in  plasma,  CSF,  and  brain  in  recently 
detoxified  alcoholics.  In  patients,  plasma 
and  CSF  studies  found  reductions  in 
GABA  levels  in  alcoholics  over  the 
first  month  of  detoxification  (Coffman 
and  Petty  1985;  Petty  et  al.  1993; 
Adinoff  et  al.  1995).  In  these  patients, 
low  plasma  GABA  levels  predicted 
relapse  (Petty  et  al.  1997),  perhaps 
because  low  GABA  levels  were  associ- 
ated with  protracted  abstinence  symp- 
toms. Although  there  are  some 
questions  regarding  the  origin  and 
implications  of  plasma  GABA,  CSF 
and  brain  GABA  levels  show  some 
similar  patterns  of  regulation  (Petty  et 
al.  1987).  The  decline  in  CSF  GABA 
levels  may  not  drop  significantly 
below  control  levels  (G.D.  Goldman 
et  al.  1981;  Hawley  et  al.  1981;  Petty 
et  al.  1987;  Roy  et  al.  1990).  CSF 
GABA  levels  in  alcoholics  are  not 
influenced  by  levels  of  comorbid 
depression  (Roy  et  al.  1991).  How- 
ever, CSF  studies  are  prone  to  error 
because  of  a  gradient  in  GABA  levels 
across  CSF  samples  and  because  CSF 
GABA  levels  are  influenced  by  age 
(Grove  et  al.  1982,  1983).  Also,  brain 
and  CSF  levels  of  GABA  may  corre- 
late (Bohlen  et  al.  1979).  However,  it 
is  possible  that  significant,  perhaps 
regionally  localized,  differences  in 
brain  GABA  levels  between  patients 
and  control  subjects  may  not  be 
reflected  in  CSF  studies.  To  date,  a 
single  report  suggests  that  occipital 
cortex  GABA  levels  are  reduced  in 
recently  detoxified  alcoholic  patients 
assessed  in  vivo  using  [:H]  magnetic 
resonance  spectroscopy  (MRS)  (Behar 
etal.  1999). 


366 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Preclinical  studies  provide  insights 
into  mechanisms  that  may  underlie 
alterations  in  brain  GAB  A  levels.  With 
long-term  administration,  GABA  lev- 
els decline  and  drop  below  baseline 
(Sytinsky  et  al.  1975;  Kulonen  1983). 
Reduced  GABA  release  and  stimula- 
tion of  glutamic  acid  dehydrogenase 
have  been  implicated  in  the  rise  of 
brain  GABA  levels  produced  by  acute 
ethanol  administration  (Hunt  1983; 
Sherif  et  al.  1997).  In  contrast,  enhan- 
ced activity  of  the  catabolic  enzyme 
GABA  transaminase  (GABA-T)  has 
been  implicated  in  subcortical  but  not 
cortical  reductions  in  GABA  levels 
(Sherif  etal.  1994,  1997). 

To  date,  insights  into  GABAA  recep- 
tor changes  in  alcoholism  come  pri- 
marily from  receptor  binding  studies. 
This  reflects  both  the  development  of 
in  vivo  neuroreceptor  imaging  strate- 
gies and  the  dearth  of  postmortem 
studies  of  GABA-related  gene  expres- 
sion in  brain  tissue  from  alcoholic 
patients  or  high-risk  populations 
(Buck  1996).  Preclinical  research 
describing  alterations  in  GABAA 
receptor  subunit  gene  expression, 
binding,  and  function  has  produced  a 
complicated  picture.  It  appears  that  the 
expression  of  some  subunits,  such  as  the 
ax,  a2,  and  a3  subunits,  are  decreased, 
while  others  are  unchanged  or  increased 
(Devaud  et  al.  1995b).  Similarly, 
there  is  evidence  of  both  reductions 
and  increases  in  binding  to  GABAA 
and  benzodiazepine  sites  (Ticku  and 
Burch  1980;  Hemmingsen  et  al.  1982; 
Ticku  et  al.  1983;  Tamborska  and 
Marangos  1986;  Mhatre  and  Ticku 
1989;  Buck  et  al.  1991;  Montpied  et 
al.  1991;  Montpied  and  Paul  1992; 


Ticku  and  Mhatre  1992;  Mhatre  and 
Ticku  1993).  Some  postmortem  stud- 
ies have  found  reductions  in  GABAa/ 
benzodiazepine  binding,  but  several 
studies  had  conflicting  results  (Tran  et 
al.  1981;  Freund  and  Ballinger  1988; 
Kril  et  al.  1988;  Freund  and  Ballinger 
1991;  Korpi  1994;  Lewohl  et  al.  1996, 
1997;  Tanda  et  al.  1997).  Some  of 
the  variability  between  studies  may 
reflect  a  compensatory  up-regulation 
in  GABAA/benzodiazepine  binding 
that  may  follow  alcoholism-related 
neurotoxicity  (Dodd  et  al.  1996). 

Three  studies  have  now  reported  ben- 
zodiazepine binding  data  using  PET 
and  single  photon  emission  computed 
tomography  (SPECT).  One  small  study 
(five  patients,  five  control  subjects) 
failed  to  find  differences  in  [nC]flum- 
azenil  binding  (Litton  et  al.  1993). 
Two  larger  studies  (Gilman  et  al. 
1996;  Abi-Dargham  et  al.  1998) 
reported  reductions  in  frontal  and 
anterior  cingulate  gyrus  benzodi- 
azepine binding  using  [nC]flumazenil/ 
PET  and  [123I]iomazenil/SPECT, 
respectively.  There  is  evidence  of 
reduced  metabolic  sensitivity  of  these 
regions  in  alcoholics  in  response  to 
lorazepam  (Volkow  et  al.  1993).  This 
blunting  did  not  rapidly  recover  with 
sobriety  (Volkow  et  al.  1997),  raising 
the  possibility  that  this  deficit 
reflected  genetic  vulnerability  or  irre- 
versible toxicity. 

A  number  of  technical  strategies  are 
emerging  to  facilitate  the  evaluation 
of  the  role  of  neurotoxicity  in  GABA- 
related  functional  and  biochemical 
neuroimaging.  First,  across  imaging 
modality,  assessments  should  adjust 
for  atrophic  changes  in  cortical  gray 


367 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


matter  based  on  structural  magnetic 
resonance  imaging  techniques  for  sep- 
arating gray  matter,  white  matter,  and 
CSF  (Hetherington  et  al.  1996).  Sec- 
ond, proton  MRS  may  play  a  role  in 
providing  insights  into  neuronal  viabil- 
ity, through  assessments  of  metabolite 
levels  in  gray  matter  N-acetylaspartate 
and  creatine  (Tsai  et  al.  1995;  Jagan- 
nathan  et  al.  1996;  Behar  et  al.  1999). 
Phosphorus  MRS  may  also  provide 
insights  to  phospholipid  synthesis  and 
catabolism  through  the  measurement 
of  phosphomonoesters  and  phospho- 
diesters  (Meyerhoffet  al.  1995). 

Psychopharmacological  studies  have 
provided  some  insights  into  the  involve- 
ment of  GABA  systems  in  withdrawal 
and  relapse  in  alcoholic  patients.  Clearly, 
drugs  facilitating  GABAA  receptor 
function,  including  benzodiazepines, 
barbiturates,  and  anticonvulsants,  are 
effective  clinically  in  suppressing  acute 
ethanol  withdrawal  symptoms  (Nutt 
et  al.  1989;  Stuppaeck  et  al.  1996). 
Preclinical  studies  suggest  that  the 
benzodiazepine  antagonist  flumazenil 
may  reduce  the  ataxic  effects  of  ethanol 
and  reduce  the  vulnerability  to  with- 
drawal seizures  in  dependent  animals 
(Buck  et  al.  1991).  In  humans,  flumaze- 
nil also  has  been  reported  to  reduce 
intoxication  associated  with  ethanol  at 
doses  greater  than  what  is  required  to 
antagonize  benzodiazepine  effects 
(Lheureux  and  Askenasi  1991).  How- 
ever, this  drug  does  not  produce  ethanol 
withdrawal  symptoms  in  alcohol- 
dependent  individuals  (Potokar  et  al. 
1997).  Ethanol  dependence  contrasts 
with  benzodiazepine  dependence,  where 
flumazenil  response  is  variable  but  more 
likely  to  precipitate  withdrawal  in  animals 


and  humans  (Griffiths  et  al.  1993; 
Gerra  et  al.  1996;  Saxon  et  al.  1997; 
Wala  et  al.  1997).  These  findings  may 
be  consistent  with  preclinical  studies 
that  suggest  that  adaptations  to  chronic 
ethanol  administration  include  changes 
in  the  expression  of  GABAA  receptor 
subunits  that  do  not  possess  the  ben- 
zodiazepine binding  site  (Bosio  et  al. 
1982;  De  Bias  1996).  Lastly,  the 
homotaurine  derivative  acamprosate  is 
structurally  related  to  GABA,  and  its 
efficacy  in  reducing  ethanol  consump- 
tion in  patients  may  be  related  to  its 
modulatory  effects  on  brain  GABA 
function  (Chick  1995). 

Neurosteroids 
and  Alcoholism 

There  is  growing  interest  in  the  role  of 
neuroactive  intermediates  in  sex 
steroid  synthesis  and  metabolism  in 
alcoholism.  Among  these  compounds, 
allopregnanolone  has  received  the 
greatest  amount  of  study.  However, 
other  compounds,  such  as  pregenolone 
sulfate  and  dihydroepiandrosterone,  also 
are  of  interest  (Melchior  and  Allen 
1992;  Melchior  and  Ritzmann  1992). 
Allopregnanolone  compound  binds 
to  the  steroid  anesthetic  site  of  the 
GABAA  receptor,  where  it  acts  as  a  co- 
agonist  (Luddens  and  Korpi  1995). 
Subsequent  studies  indicate  that  allo- 
pregnanolone shares  discriminative 
stimulus  properties  with  ethanol  and 
suppresses  the  ethanol  abstinence  syn- 
drome in  animals  (Ator  et  al.  1993; 
Devaud  et  al.  1995&;  K.  Grant  et  al. 
1996#).  Allopregnanolone  also  has 
anxiolytic  and  sedative  properties,  and 
circulating  levels  may  potentiate  the 
effects  of  ethanol  (Brot  et  al.  1995, 


368 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


1997).  Allopregnanolone  levels,  like 
those  of  its  precursor,  progesterone, 
show  marked  variation  during  the 
menstrual  cycle.  In  late  luteal  phase 
allopregnanolone  levels  drop,  and 
these  reductions  have  been  related  to 
mood  disturbances  during  this  phase 
of  the  menstrual  cycle  (Rapkin  et  al. 
1997).  Similarly,  there  has  been  signif- 
icant interest  in  the  possibility  that 
reductions  in  neurosteroid  levels  in 
the  late  luteal  phase  might  contribute 
to  increased  intensity  of  the  discrimi- 
native stimulus  properties  of  ethanol 
(Grant  et  al.  1997a)  and  increased 
alcohol  consumption  in  women  dur- 
ing this  phase  of  the  menstrual  cycle 
(Charette  et  al.  1990;  Allen  1996). 
Supporting  this  view,  higher  allopreg- 
nanolone levels  in  the  late  luteal  phase 
are  associated  with  reduced  triazolam 
self- administration  in  women  (de  Wit 
and  Rukstalis  1997).  Thus,  cyclical 
variation  in  neurosteroid  levels  or 
their  consequences  could  be  an 
important  focus  for  pharmacotherapy 
development  for  female  alcoholics. 

GABA  Overview 

GABAA  receptors  are  clearly  an  impor- 
tant target  for  the  rewarding  effects  of 
ethanol  in  the  brain.  There  is  growing 
evidence  that  reduced  sensitivity  to 
ethanol  effects  on  GABA  systems  may 
contribute  to  a  familial  vulnerability 
to  alcoholism.  This  vulnerability  is 
hypothesized  to  be  mediated,  in  part, 
by  reduced  sensitivity  to  the  dysphoric 
consequences  of  ethanol  actions  at 
GABAA  receptors  and  preservation  of 
its  rewarding  effects.  It  is  not  yet  clear 
whether,  as  with  5-HT  systems, 
reduced  sensitivity  to  the  GABAergic 


component  of  ethanol  response  is  lim- 
ited to  a  subtype  of  familial  alcoholism. 
The  involvement  of  GABA  systems  in 
the  rewarding  effects  of  ethanol  sug- 
gests that  GABAergic  systems  may  be 
involved  in  the  component  of  ethanol 
craving  involving  the  pairing  of  envi- 
ronmental cues  with  ethanol  effects  in 
the  brain.  However,  this  has  not 
received  sufficient  clinical  study  to 
draw  direct  inferences. 

Chronic  ethanol  administration 
produces  adaptations  within  GABA 
systems  that  contribute  to  tolerance 
and  withdrawal.  GABA  agonists 
clearly  are  effective  in  the  treatment  of 
acute  withdrawal  symptoms.  Because 
a  component  of  craving  may  be  driven 
by  acute  or  protracted  withdrawal 
symptoms,  disturbances  in  GABA  sys- 
tems may  also  contribute  to  craving 
and  relapse.  Furthermore,  neurosteroids 
may  provide  a  novel  and  important 
target  for  medication  development  for 
female  alcoholics. 

GLUTAMATE  SYSTEMS 

Contribution  of  the  NMDA 
Glutamate  Receptor 
Antagonist  Properties 
of  Ethanol  to  the 
Discriminative  Stimulus 
Effects  of  Ethanol 

Glutamate  is  the  most  prevalent  exci- 
tatory neurotransmitter  in  the  brain, 
and  it  mediates  most  cortico-cortical 
communication.  Glutamate  receptors 
are  divided  into  three  classes  of  recep- 
tors that  bear  ion  channels  and  one 
family  of  metabotropic  receptors  cou- 
pled by  G  proteins  to  intracellular  sec- 


369 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


ond  messenger  systems.  Through  the 
range  of  doses  associated  with  human 
intoxication,  ethanol  preferentially 
and  dose-dependently  blocks  the  N- 
methyl-D-aspartate  (NMDA)  subclass 
of  glutamate  receptors  (Lovinger 
1997).  This  action  is  not  competitive 
with  glutamate  and  may  be  mediated 
by  ethanol  binding  to  a  hydrophobic 
pocket  on  the  receptor  (Peoples  and 
Weight  1995)  and  inhibitory  effects 
on  the  strychnine-insensitive  glycine 
co-agonist  site  of  these  receptors 
(Michaelis  et  al.  1996). 

A  growing  body  of  research  sug- 
gests that  NMDA  receptor  antago- 
nism contributes  to  the  discriminative 
stimulus  effects  of  ethanol  in  animals 
and  humans.  In  drug  discrimination 
paradigms,  animals  identify  NMDA 
antagonists  as  being  similar  to  ethanol 
(Grant  and  Colombo  1993«;  K.  Grant 
et  al.  1996^).  The  highest  degree  of 


similarity  between  NMDA  antagonist 
effects  and  ethanol  effects  is  observed 
when  animals  are  trained  to  recognize 
ethanol  doses  comparable  to  several 
drinks  of  alcohol  in  humans. 

The  NMDA  antagonist  ketamine 
produced  dose-related  ethanol-like 
effects  in  recently  detoxified  alcoholic 
patients  (Krystal  et  al.  1998&). 
Ethanol-like  effects,  measured  by  the 
Sensation  Scale,  a  visual  analog  scale  of 
euphoria,  and  the  Biphasic  Alcohol 
Effects  Scale  were  more  prominent  at 
0.5  mg/kg  than  at  0.1  mg/kg.  The 
response  to  ketamine  was  rated  signif- 
icantly more  similar  to  the  effects  of 
ethanol  than  it  was  to  the  effects  of 
marijuana  or  cocaine  (figure  2). 

At  0.1  mg/kg,  ketamine  effects 
were  rated  as  similar  to  one  to  two 
standard  ethanol  drinks  (-15  mL 
absolute  ethanol);  at  0.5  mg/kg,  its 
effects  were  rated  similar  to  eight  to 


Base  10    20   30    40   50   60    70   80 

Time  (min) 


i  10    20   30   40   50    60   70   80 

Time  (min) 


i  10   20   30   40   50   60   70   80 

Time  (min) 


Figure  2.  Similarity  of  the  effects  of  placebo  (open  circles),  ketamine  0.1  mg/kg  (filled  circles), 
and  ketamine  0.5  mg/kg  (filled  squares)  to  ethanol  (left),  marijuana  (middle),  and  cocaine  (right) 
in  alcoholic  patients  (n  =  20).  Values  are  expressed  as  mean  ±  SEM.  Ketamine  effects  were  signifi- 
cantly more  similar  to  ethanol  than  both  marijuana  and  cocaine  by  post  hoc  contrast  (Fl  =  6.7, 
p  =  0.02).  Reprinted  with  permission  from  Krystal,  J.H.;  Petrakis,  I.L.;  Webb,  E.;  Cooney,  N.L.; 
Karper,  L.P.;  Namanworth,  S.;  Trevisan,  L.A.;  and  Charney,  D.S.  Dose-related  ethanol-like  effects 
of  the  NMDA  antagonist,  ketamine,  in  recently  detoxified  alcoholics.  Archives  of  General  Psychiatry 
55:354-360,  April  1998.  Copyright  1998,  American  Medical  Association. 


370 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


nine  standard  ethanol  drinks.  Keta- 
mine  effects  were  rated  as  more  simi- 
lar to  the  sedative-descending  limb 
than  the  stimulant-ascending  limb 
effects  of  ethanol.  Ketamine  did  not 
stimulate  ethanol  craving  in  patients. 
Perhaps  contributing  to  this  effect, 
patients  anecdotally  reported  that  the 
low  dose  of  ketamine  was  not  suffi- 
ciently ethanol-like  and  that  the  high 
dose  of  ketamine  sated  their  desire  for 
ethanol.  Recent  data  from  our  labora- 
tory also  suggest  that  a  high  dose  of 
the  partial  agonist  of  the  strychnine - 
insensitive  glycine  co-agonist  site  of 
the  NMDA  receptor,  D-cycloserine 
(1,000  mg),  produces  ethanol-like 
subjective  effects  in  healthy  subjects 
and  recently  detoxified  male  early- 
onset  alcoholics;  glycine  (0.3  g/kg),  a 
full  agonist  of  this  site,  does  not  pro- 
duce these  effects  (Krystal  et  al. 
1998a).  Like  ketamine,  D-cycloserine 
did  not  produce  alcohol  craving  in 
patients.  These  data  further  implicate 
NMDA  receptors  in  ethanol  effects. 

Modulation  of  Human 
Ethanol  Intoxication  by 
Glutamatergic  Agents 

D-cycloserine  modulates  ethanol 
intoxication  in  healthy  human  subjects 
(Trevisan  et  al.  1995).  In  animals, 
ethanol  potentiates  the  action  of  NMDA 
antagonists  (Toropainen  et  al.  1997). 
Also,  ethanol  effects  in  neurochemical 
and  behavioral  paradigms  are  potenti- 
ated by  high-dose  glycine  partial  agonists 
or  antagonists  and  attenuated  by 
glycine  agonists  and  low- dose  glycine 
partial  agonists  (Khanna  et  al.  1995; 
Moraes  Ferreira  and  Morato  1997). 
Complementing  these  studies,  prelim- 


inary data  suggest  that  D-cycloserine, 
500  mg,  potentiated  ethanol  intoxica- 
tion (Trevisan  et  al.  1995).  Also,  there 
was  evidence  that  cycloserine  and 
ethanol  had  additive  amnestic  effects. 
However,  D-cycloserine  reduced  ethanol 
intoxication  beginning  approximately 
6  hours  after  D-cycloserine  administra- 
tion, when  its  levels  declined.  D- 
cycloserine  did  not  alter  ethanol  levels 
at  any  time  point.  These  data  are  con- 
sistent with  the  view  that  D-cycloser- 
ine has  glycine/NMDA  antagonist-like 
effects  at  oral  doses  of  500  mg  or 
greater.  At  lower  doses,  D-cycloserine 
primarily  has  agonist-like  effects 
(D'Souza  et  al.  1995).  These  data 
suggest  a  potential  role  for  NMDA 
antagonists  in  reducing  drinking  in 
recovering  alcoholics  by  potentiating 
the  adverse  effects  of  ethanol  intoxica- 
tion. They  may  also  suggest  that 
glycine  agonists  have  therapeutic 
potential  arising  from  their  capacity  to 
attenuate  ethanol  intoxication. 

Association  of  Human 
Ethanol  Dependence 
With  Protracted  Up- 
Regulation  of  NMDA 
Receptor  Function 
and  Altered  Responses 
to  NMDA  Antagonists 

In  humans,  as  in  animals,  acute 
ethanol  withdrawal  activates  glutamate 
systems.  Human  ethanol  withdrawal  is 
associated  with  increases  in  CSF  gluta- 
mate  levels  (Tsai  et  al.  1995). 
Increases  in  extracellular  glutamate 
levels  may  be  consistent  with  regional 
reductions  in  binding  to  the  glutamate 
transporter  (Cummins  et  al.  1990)  or 
increased  release  (Keller  et  al.  1983). 


371 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Increased  glutamate  levels  might  be 
expected  to  down-regulate  NMDA 
receptors.  However,  the  predominant 
effect  appears  to  be  NMDA  receptor 
up-regulation  in  animal  studies  (Gulya 
et  al.  1991).  A  postmortem  study 
found  that  the  Bmax  of  NMDA,  but 
not  a-amino-3-hydroxy-5-methylisox- 
azole  propionic  acid  (AMPA),  receptors 
was  increased  in  the  frontal  cortex  of 
alcoholics  (Freund  and  Anderson  1996). 
In  the  hippocampus,  NMDA  receptor 
affinity,  but  not  density,  was  increased 
(Michaelis  et  al.  1990).  The  persistent 
alterations  in  NMDA  receptor  function 
may  potentiate  the  neurotoxic  and 
proconvulsant  effects  of  increased  glu- 
tamate release  during  withdrawal.  How- 
ever, the  etiology  of  persistent  NMDA 
receptor  up-regulation  in  alcoholics  is 
not  clear  and  may  also  reflect  a 
genetic  predisposition  to  alcoholism 
or  alcoholism-related  neurotoxicity. 
Regardless  of  etiology,  long-lasting 
facilitation  of  NMDA  receptor  func- 
tion in  alcoholics  may  have  ongoing 
neurological  implications,  including 
increased  seizure  risk  (Brown  et  al. 
1988;  Lechtenberg  and  Worner 
1991)  and  startle  hyperreflexia  (Krys- 
tal  et  al.  1997). 

Alterations  in  NMDA  receptor  func- 
tion in  recovering  alcohol-dependent 
patients  may  be  associated  with  a  shift 
in  the  reward  valence  of  the  NMDA 
antagonist  component  of  ethanol 
response.  Recently  detoxified  early- 
onset  male  alcoholic  patients  show 
marked  reductions  in  their  sensitivity  to 
the  perceptual  and  psychotogenic  effects 
of  ketamine  (Krystal  et  al.  1995).  In 
contrast,  these  data  suggest  that  these 
patients  exhibited  a  trend  for  increased 


euphoric  responses  to  this  drug.  Data 
from  another  study  suggest  that  these 
patients  also  showed  blunted  responses 
to  the  effects  of  high-dose  (1,000  mg) 
D-cycloserine  (Krystal  et  al.  1998&). 
Together,  these  data  suggest  that 
recently  detoxified  alcoholics  are  less 
sensitive  to  the  dysphoric  effects  of 
NMDA  antagonists,  while  the  euphoric 
effects  of  these  drugs  are  preserved. 
Shifts  in  the  reward  valence  of  other 
NMDA  antagonists  in  early-onset 
alcoholic  patients  may  also  apply  to 
the  NMDA  antagonist  component  of 
ethanol  response.  If  so,  then  the  blunted 
cognitive  and  behavioral  effects  of 
ethanol  in  alcoholic  and  high-risk 
populations  described  earlier  in  the  con- 
text of  GABA  systems  may,  in  part,  also 
reflect  a  glutamatergic  disturbance. 

However,  the  current  clinical  data- 
base does  not  yet  allow  one  to  choose 
from  among  the  potential  pathophysi- 
ological processes  that  might  result  in 
altered  NMDA  antagonist  response  in 
alcoholic  patients.  One  possibility  is 
that  reduced  NMDA  receptor  sensi- 
tivity reflects  cross -tolerance  between 
ethanol  and  other  NMDA  antagonists. 
Although  this  cross-tolerance  has 
been  described  in  preclinical  research 
(Fitzgerald  and  Nestler  1995;  Grant 
and  Lovinger  1995),  it  is  not  yet  clear 
whether  this  concept  is  applicable  to 
the  clinical  studies.  A  second  possibility 
is  that  reduced  ketamine  sensitivity 
reflects  neurotoxic  changes  associated 
with  alcoholism.  A  third  explanation 
could  be  that  alterations  in  glutamatergic 
regulation  reflect  a  pathophysiological 
process  predisposing  individuals  to 
alcoholism.  Additional  research  will  be 
needed  to  untangle  these  possibilities. 


372 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Acamprosate:  A 
Pharmacotherapy  for 
Alcoholism  With  Both 
NMDA  Antagonist 
and  Agonist  Properties 

Acamprosate  is  a  homotaurine  deriva- 
tive without  ethanol-like  behavioral 
effects  that  reduces  ethanol  consump- 
tion in  animals  (Spanagel  et  al. 
1996/j,  1996^).  Several  European 
studies  suggest  that  this  drug  is  a 
highly  promising  pharmacotherapeu- 
tic  support  for  reducing  alcohol  con- 
sumption in  alcoholics  (Paille  et  al. 
1995;  Sass  et  al.  1996;  Whitworth  et 
al.  1996;  Pelc  et  al.  1997). 

Acamprosate  has  been  reported  to 
have  NMDA  antagonist  activity  and  to 
suppress  ethanol  withdrawal  (Zeise  et  al. 
1993;  Putzke  et  al.  1996).  Based  on 
these  findings,  some  researchers  have 
hypothesized  that  acamprosate  may 
reduce  alcohol  consumption,  in  part, 
by  reducing  alcohol  withdrawal  symp- 
toms that  contribute  to  craving  (Lit- 
tleton 1995).  In  contrast,  acamprosate 
may  also  reduce  alcohol  consumption 
by  potentiating  the  dysphoric  effects 
of  ethanol.  If  reduced  ketamine  sensi- 
tivity in  alcoholics  reflects  a  reduction 
in  the  sensitivity  to  the  dysphoric 
effects  of  the  NMDA  antagonist  com- 
ponent of  ethanol,  then  maintenance 
treatment  with  acamprosate  could 
potentiate  those  very  effects  of 
ethanol  that  limit  alcohol  consump- 
tion in  nonalcoholic  populations. 
However,  acamprosate  also  appears  to 
have  NMDA  agonist  effects  in  pre- 
clinical research  paradigms  (Madamba 
et  al.  1996).  If  so,  then  acamprosate 
may  reduce  the  behavioral  effects  of 
ethanol  via  this  mechanism. 


Glutamate  Overview 

The  clinical  literature  now  appears  to 
support  the  basic  literature  in  suggest- 
ing that  NMDA  antagonism  is  relevant 
to  the  discriminative  and  perhaps  the 
rewarding  effects  of  ethanol.  Although 
drugs  that  selectively  block  NMDA 
receptors  are  unlikely  to  have  therapeu- 
tic potential  in  the  treatment  of  alcohol- 
ism due  to  their  potential  to  produce 
psychosis,  other  glutamatergic  agents 
may  be  acceptable  for  this  purpose. 
One  potential  target  for  these  drugs  is 
the  strychnine-insensitive  glycine  site 
of  the  NMDA  receptor  complex. 
Acamprosate,  a  drug  with  GAB  A  ago- 
nist, NMDA  antagonist,  and  NMDA 
agonist  effects,  does  not  have  ethanol- 
like  discriminative  effects.  Its  mecha- 
nism of  action  is  currently  unclear. 

OPIATES 

Preclinical  studies  suggest  that  the 
behavioral  effects  of  ethanol  are  mod- 
ulated by  opiate  antagonists  (Herz 
1997).  The  opiate -facilitating  actions 
of  ethanol  may  contribute  to  its 
inhibitory  effects  on  nucleus  accum- 
bens  glutamatergic  function  (Nie  et 
al.  1994).  Opiate  antagonists  reduced 
ethanol  self- administration  in  rodents 
and  primates  (Myers  et  al.  1986; 
Volpicelli  et  al.  1986;  Honkanen  et  al. 
1996).  Similarly,  most  studies  suggest 
that  naltrexone  appears  to  reduce  the 
rewarding  effects  of  ethanol  and  alco- 
hol consumption  in  social  drinkers 
(Swift  et  al.  1994;  Davidson  et  al. 
1996;  Doty  et  al.  1997).  Also,  naltrex- 
one maintenance  appears  to  reduce 
the  pleasurable  aspects  of  alcohol  con- 
sumed during  treatment  for  alco- 


373 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


holism  (Volpicelli  et  al.  1995;  O'Malley 
et  al.  1996).  This  property  appears  to 
contribute  to  the  capacity  of  opiate 
antagonists  to  reduce  alcohol  con- 
sumption in  alcoholic  patients 
(O'Malley  et  al.  1992;  Volpicelli  et  al. 
1992;  Mason  et  al.  1994). 

The  contributions  of  endogenous 
opiate  systems  to  the  rewarding  effects 
of  ethanol  are  further  supported  by 
evidence  of  abnormalities  in  these  sys- 
tems in  alcoholic  patients.  However, 
the  current  data  do  not  yield  a  clear 
picture  of  these  abnormalities.  Post- 
mortem studies  have  described  both 
increased  u.  receptor  density  (Ritchie 
and  Noble  1996)  and  decreased  [i 
receptor  affinity  (Tabakoff  et  al. 
1985).  CSF  and  plasma  studies  have 
also  suggested  the  existence  of  reduc- 
tions in  (3-endorphin  levels  and 
ethanol -stimulated  increases  in  plasma 
(3-endorphin  levels  in  some  (Borg  et 
al.  1982;  Genazzani  et  al.  1982; 
Gianoulakis  et  al.  1996),  but  not  all, 
studies  (Petrakis  et  al.  1997). 
Naltrexone  reductions  in  ethanol 
effects  appear  to  be  particularly  evi- 
dent in  individuals  at  high  risk  for 
developing  alcoholism  (King  et  al. 
1997).  These  data  suggest  a  genetic 
component  underlying  the  efficacy  of 
naltrexone  treatment  for  alcoholism. 
To  date,  there  have  been  negative 
results  from  studies  evaluating  the 
association  of  the  proenkephalin 
A  gene  and  [i  opiate  receptor  and 
alcoholism  (Chan  et  al.  1994;  Bergen 
et  al.  1997). 

Opiate  Overview 

Unlike  the  other  systems  reviewed  here, 
it  remains  controversial  as  to  whether 


opiate  receptors  are  a  direct  target  for 
ethanol  in  the  brain.  However,  opiate 
receptor  systems  modulate  most  other 
systems  implicated  in  the  rewarding 
effects  of  ethanol.  Perhaps  through 
these  mechanisms,  opiate  antagonists 
are  effective  in  reducing  the  rewarding 
effects  of  ethanol  and  reducing  con- 
sumption. A  growing  number  of  studies 
suggest  that  a  genetic  factor  associated 
with  the  vulnerability  for  alcoholism 
may  particularly  distinguish  those 
individuals  for  whom  naltrexone 
reduces  the  rewarding  effects  of 
ethanol.  However,  the  gene  underly- 
ing this  interactive  effect  has  not  yet 
been  identified. 

CATECHOLAMINES 

To  date,  catecholamine  systems  have 
received  relatively  little  human  study  in 
relation  to  behaviors  associated  with 
alcoholism,  despite  extensive  preclinical 
data  implicating  dopamine  systems,  in 
particular,  in  the  rewarding  effects  of 
ethanol  (Koob  1998).  Both  D1  and 
D2  receptors  are  implicated  in  the 
rewarding  effects  of  ethanol  and  in 
animal  models  for  drug  craving.  Fur- 
thermore, ethanol  withdrawal  is  associ- 
ated with  reductions  in  limbic  dopamine 
function  (Hunt  and  Majchrowicz 
1983)  in  animal  studies  and  reduced 
CSF  homovanillic  acid  in  some,  but 
not  all,  clinical  studies  (Major  et  al. 
1977).  Noradrenergic  systems  are  less 
clearly  implicated  in  the  rewarding 
effects  of  ethanol  or  craving.  How- 
ever, a2  adrenergic  antagonists  have 
been  reported  to  reduce  some  effects 
of  ethanol  in  rats  (Lister  et  al.  1989; 
Durcanetal.  1991). 


374 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


There  have  been  relatively  few 
human  studies  evaluating  the  contribu- 
tions of  catecholamine  systems  to 
ethanol  intoxication,  craving,  or  self- 
administration.  Some  studies  used 
strategies  that  did  not  distinguish  among 
catecholamines.  For  example,  one 
study  found  that  catecholamine  syn- 
thesis inhibition,  produced  by  a- 
methyl-para-tyrosine,  modestly  reduced 
ethanol  intoxication  in  healthy 
humans  (Ahlenius  et  al.  1973).  Simi- 
larly, dexamphetamine  and  metham- 
phetamine  pretreatment  either  had  no 
effect  or  modestly  potentiated  ethanol 
intoxication  in  humans  (Perez- Reyes 
et  al.  1992;  Mendelson  et  al.  1995). 
Building  on  preclinical  findings,  clinical 
studies  suggest  that  the  a2  adrenergic 
antagonist  yohimbine  potentiated  the 
intoxicating  effects  of  ethanol  but  did 
not  substantially  alter  the  subjective 
sense  of  euphoria  associated  with 
intoxication  (McDougle  et  al.  1995). 
In  recently  detoxified  early-onset  alco- 
hol-dependent patients,  yohimbine 
effects  have  a  low  degree  of  similarity 
to  ethanol  effects  (Krystal  et  al.  1994). 
In  this  study,  yohimbine  also  reduced 
craving  relative  to  placebo  and  mCPP. 
Conversely,  clonidine  does  not  appear 
to  be  effective  in  suppressing  ethanol 
cue-induced  craving  (Petrakis  and 
Krystal  unpublished  data). 

Several  studies  document  reduced 
sensitivity  of  both  dopamine  and  nora- 
drenergic receptors  in  recently  detoxified 
patients.  Reduced  sensitivity  of  dopa- 
mine receptors  is  suggested  by  blunted 
growth  hormone  responses  to  apomor- 
phine  or  bromocriptine  (Balldin  et  al. 
1985;  Balldin  et  al.  1992;  Farren  et  al. 
1995«;  Heinz  et  al.  1995).  These 


data  are  consistent  with  neuroimaging 
data  suggesting  that  the  density  of 
dopamine  transporter  binding  is  pre- 
served but  striatal  D2  receptor  density 
is  decreased  in  alcoholic  patients 
(Volkow  et  al.  1996;  Tiihonen,  et  al. 
1997).  Down-regulation  of  postsy- 
naptic cc2  adrenergic  receptors  is  sug- 
gested by  blunted  growth  hormone 
responses  to  clonidine  and  increased 
Cortisol  responses  to  yohimbine  (Glue 
et  al.  1989;  Krystal  et  al.  1996).  In 
contrast,  presynaptic  noradrenergic 
activity  appears  to  normalize  rapidly 
following  withdrawal  (Borg  et  al. 
1983).  Similarly,  the  presynaptic  com- 
ponent of  the  noradrenergic  response 
to  yohimbine  is  normal  in  patients 
sober  for  approximately  1  month 
(Krystal  et  al.  1996). 

Genetic  studies  relating  catechol- 
amine receptor  alleles  to  the  vulnera- 
bility to  alcoholism  have  been  a 
promising  but  controversial  research 
strategy  that  has  not  yet  born  fruit. 
Initial  studies  suggested  that  alleles  of 
the  D2  receptor  were  associated  with 
alcoholism,  particularly  severe  alco- 
holism (Blum  et  al.  1990,  1991; 
Parsian  et  al.  1991;  Higuchi  et  al. 
1994).  However,  subsequent  studies 
using  more  definitive  techniques  were 
negative  (Bolos  et  al.  1990;  Gelernter 
et  al.  1991;  Geijer  et  al.  1994;  Gejman 
et  al.  1994;  D.  Goldman  et  al.  1997). 
Reanalysis  of  the  published  studies  also 
suggests  that  the  positive  finding  may 
have  arisen  due  to  ethnic  differences 
between  the  patient  and  control  pop- 
ulations (Gelernter  et  al.  1993).  A 
subsequent  study  also  suggested  that 
alleles  of  the  D2  receptor  might  predict 
an  anticraving  response  to  bromocrip- 


375 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


tine  (Lawford  et  al.  1995).  However, 
studies  using  dopamine  agonists  to 
treat  alcoholism  have  so  far  had  limited 
promise  (Penick  et  al.  1996;  Naranjo  et 
al.  1997).  There  are  also  interesting 
potential  associations  between  versions 
of  the  D4  receptor  and  novelty-seeking 
(Sander  et  al.  1997),  with  some  negative 
reports  (Sullivan  et  al.  1998).  However, 
so  far  there  is  little  evidence  linking  the 
Dl5  D3,  or  D4  receptor  genes  to  alco- 
holism (Adamson  et  al.  1995;  Gorwood 
et  al.  1995;  Sander  et  al.  1995;  Malhotra 
et  al.  1996;  Muramatsu  et  al.  1996; 
Chang  et  al.  1997;  Parsian  et  al.  1997). 

Catecholamine  Overview 

Of  all  systems  in  the  brain,  dopamine 
systems  are  perhaps  most  strongly 
linked  to  the  rewarding  properties  of 
drugs  of  abuse.  However,  dopaminer- 
gic agents  have  not  yet  shown  signifi- 
cant promise  in  modulating  the 
behavioral  effects  of  ethanol,  reducing 
ethanol  craving,  or  preventing 
ethanol  consumption.  One  factor  that 
may  contribute  to  this  problem  is  the 
absence  of  clinical  studies  focused  on 
the  T)1  receptor.  However,  overall 
dopaminergic  contributions  have 
received  relatively  little  attention. 
Noradrenergic  systems  have  been 
studied  in  the  clinical  laboratory,  but 
a  tentative  finding  suggesting  that  a2 
adrenergic  antagonists  might  potenti- 
ate ethanol  effects  in  healthy  subjects 
and  reduce  craving  in  patients  has  not 
yet  been  followed  by  a  clinical  trial. 

DISCUSSION 

There  is  good  news  and  bad  news 
regarding  achievements  in  the  study 


of  behaviors  related  to  alcohol  abuse. 
The  good  news  is  that  human  laboratory 
studies  support  the  efficacy  of  naltrex- 
one. Although  these  studies  did  not  lead 
to  the  development  of  naltrexone,  it  is 
at  least  reassuring  that  these  paradigms 
may  have  the  sensitivity  to  uncover 
other  agents  that  might  be  of  therapeu- 
tic use  in  the  treatment  of  alcoholism. 
Mostly,  however,  the  clinical  studies 
have  produced  unanswered  questions. 
These  questions,  as  a  group,  pertain  to 
the  importance  of  mechanisms  under- 
lying the  subjective  effects  of  ethanol 
to  the  treatment  of  alcoholism.  In  this 
regard,  the  therapeutic  potential  of 
ethanol  effects  on  brain  5-HT, 
GABA,  NMDA,  and  catecholamine 
systems  does  not  appear  to  be  fully 
realized.  Although  the  field  of  alco- 
holism research  seemingly  should  be 
interested  in  drugs  that  blocked  the 
euphoric  effects  of  mCPP,  ketamine, 
benzodiazepines,  and  ethanol,  this 
line  of  research  (with  the  possible 
exception  of  attempts  to  block  ethanol 
effects)  has  received  vanishingly  little 
clinical  study.  A  major  obstacle  to  this 
line  of  research  is  the  limited  availability 
of  selective  agonists  and  antagonists. 
These  agents  are  often  in  Phase  II  or 
III  of  testing,  but  are  unavailable  to 
clinical  researchers.  It  would  be  useful 
for  the  National  Institute  on  Alcohol 
Abuse  and  Alcoholism  (NIAAA)  to 
consider  establishing  a  program  that 
parallels  programs  in  the  National 
Institute  of  Mental  Health  and  the 
National  Institute  on  Drug  Abuse 
designed  to  target  particular  agents  of 
high  interest  and  facilitate  their  avail- 
ability to  clinical  investigators  to  pursue 
aims  consistent  with  those  of  NIAAA. 


376 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


A  second  line  of  research  that  is 
beginning  to  be  tapped  is  the  use  of 
the  laboratory  to  study  the  neurobiology 
of  triggers  for  relapse  to  alcohol  use, 
particularly  alcohol-related  cues,  the 
priming  effects  of  alcohol  consumption 
on  drinking,  and  the  interactive  effects 
of  stress  (negative  mood  induction) 
and  alcohol  cues.  If,  as  for  naltrexone, 
clinical  research  can  successfully  move 
from  preclinical  research  to  full-scale 
clinical  trials,  then  deficits  in  the  clinical 
research  portfolio  may  not  be  an 
immovable  obstacle  to  medications 
development  for  alcoholism.  However, 
if  the  differences  between  animal  and 
human  pharmacology  are  important, 
as  illustrated  by  the  difference  between 
the  animal  and  human  5-HT1B,  then 
the  deficits  in  the  clinical  research 
arena  may  be  reflected  in  the  fact  that 
there  are  relatively  few  effective  phar- 
macotherapies for  alcoholism. 

Alcoholism  research  has  also  not 
fully  integrated  functional  neuroimaging 
techniques  to  facilitate  the  study  of  the 
neural  circuitry  underlying  craving  or 
factors  related  to  alcohol  consumption. 
Some  progress  has  been  made  in  study- 
ing the  circuitry  of  ethanol  effects  on 
the  human  brain  (de  Wit  et  al.  1990; 
Volkow  et  al.  1990).  However,  alco- 
holism studies  lag  behind  studies 
attempting  to  identify  the  circuitry  of 
cocaine  craving  (Grant  et  al.  1996)  A 
critical  step  in  alcoholism  research  will 
be  the  integration  of  evolving  studies 
of  the  neural  circuitry  of  behavior  and 
psychopharmacology. 

The  field  of  alcoholism  research, 
however,  may  be  leading  psychiatric 
research  in  its  sensitivity  to  subtypes 
in  bridging  clinical  molecular  genetics 


and  other  domains  of  clinical  neuro- 
science. From  the  descriptive  perspec- 
tive, there  is  great  interest  in  the 
relationship  between  the  vulnerability 
to  early- onset  alcoholism  and  altered 
function  within  several  systems  in  the 
brain.  The  diversity  of  systems  impli- 
cated in  the  vulnerability  to  alcoholism 
could  very  likely  reflect  the  worst  of  all 
possible  worlds  with  respect  to  a  quick 
answer  to  the  etiology  and  treatment  of 
alcoholism:  multiple  genes  modulating 
multiple  neurotransmitter  systems. 
Regardless  of  the  intrinsic  complexity, 
the  bridging  of  pharmacology,  behav- 
ioral science,  neuroimaging,  and  mole- 
cular genetics  remains  the  brightest 
hope  for  a  "home  run"  in  the  treat- 
ment of  alcoholism. 

ACKNOWLEDGMENTS 

This  work  was  supported  by  research 
funds  from  the  Department  of  Veter- 
ans Affairs  and  NIAAA  (ROl 
AA10121-01). 

REFERENCES 

Abi-Dargham,  A.;  Krystal,  J.H.;  Anjivel, 
S.;  Scanley,  E.;  Zoghbi,  S.;  Baldwin, 
R.M.;  Rajeevan,  N.;  Seibyl,  J. P.;  Charney, 
D.S.;  Laruelle,  M.;  and  Innis,  R.B. 
Alterations  of  benzodiazepine  receptors  in 
type  II  alcoholics  measured  with  SPECT 
and  [123I]iomazenil.  Am  J  Psychiatry 
155:1550-1555,  1998. 

Adamson,  M.D.;  Kennedy,  I.;  Petronis, 
A.;  Dean,  M.;  Virkkunen,  M.;  Linnoila, 
M.;  and  Goldman,  D.  DRD4  dopamine 
receptor  genotype  and  CSF  monoamine 
metabolites  in  Finnish  alcoholics  and 


377 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


controls.  Am  J  Med  Genet  60(3): 
199-205, 1995. 

Adinoff,  B.;  Kramer,  G.L.;  and  Petty,  F. 
Levels  of  gamma-aminobutyric  acid  in 
cerebrospinal  fluid  and  plasma  during 
alcohol  withdrawal.  Psychiatry  Res 
59:137-144,  1995. 

Ahlenius,  S.;  Carlsson,  A.;  Engel,  J.; 
Svensson,  T.;  and  Sodersten,  P. 
Antagonism  by  alpha  methyltyrosine  of 
the  ethanol-induced  stimulation  and 
euphoria  in  man.  Clin  Pharmacol  Ther 
14:586-591,  1973. 

Allen,  D.  Are  alcoholic  women  more 
likely  to  drink  premenstrually?  Alcohol 
Alcohol  31:145-147 ,  1996. 

Ator,  N.A.;  Grant,  K.A.;  Purdy,  R.H.; 
Paul,  S.M.;  and  Griffiths,  R.R.  Drug 
discrimination  analysis  of  endogenous 
neuroactive  steroids  in  rats.  Eur  J 
Pharmacol 241:237 '-243,  1993. 

Babor,  T.F.;  Hofmann,  M.;  DelBoca, 
F.K.;  Hesselbrock,  V.;  Meyer,  R.E.; 
Dolinsky,  Z.S.;  and  Rounsaville,  B.  Types 
of  alcoholics,  I.  Evidence  for  an  empiri- 
cally derived  typology  based  on  indicators 
of  vulnerability  and  severity.  Arch  Gen 
Psychiatry  49 :  599-608 ,  1 992 . 

Badawy,  A.A.  Tryptophan  metabolism  and 
disposition  in  relation  to  alcohol  and  alco- 
holism. Adv  Exp  Med  Biol  398:75-82, 1996. 

Balldin,  J.;  Ailing,  C.;  Gottfries,  C.G.; 
Lindstedt,  G.;  and  Langstrom,  G. 
Changes  in  dopamine  receptor  sensitivity 
in  humans  after  heavy  alcohol  intake. 
Psychopharmacology  86:142-146,  1985. 

Balldin,  J. I.;  Berggren,  U.C.;  and 
Lindstedt,  G.  Neuroendocrine  evidence 
for  reduced  dopamine  receptor  sensitivity 


in  alcoholism.  Alcohol  Clin  Exp  Res 
16:71-74,  1992. 

Beck,  O.;  Borg,  S.;  Holmstedt,  B.;  Kvande, 
PL;  and  Shroder,  R.  Concentration  of 
serotonin  metabolites  in  the  cerebrospinal 
fluid  from  alcoholics  before  and  during 
disulfiram  therapy.  Acta  Pharmacol 
Toxicol  47:305-307,  1980. 

Beck,  O.;  Borg,  S.;  and  Sedvall,  G. 
Tryptophan  levels  in  human  cerebrospinal 
fluid  after  acute  and  chronic  ethanol 
consumption.  Drug  Alcohol  Depend 
12:217-222,  1983. 

Behar,  K.L.;  Rothman,  D.L.;  Petersen, 
K.F.;  Hooten,  M.;  Delaney,  R.;  Petroff, 
O.A.C.;  Shulman,  G.I.;  Navarro,  V.; 
Petrakis,  I.L.;  Charney,  D.S.;  and  Krystal, 
J.H.  Preliminary  evidence  of  reduced 
cortical  GABA  levels  in  localized  1H 
NMR  spectra  of  alcohol  dependent  and 
hepatic  encephalopathy  patients.  Am  J 
Psychiatry  156:952-955,  1999. 

Benkelfat,  C;  Murphy,  D.L.;  Hill,  J.L.; 
George,  D.T.;  Nutt,  D.;  and  Linnoila,  M. 
Ethanollike  properties  of  the  serotonergic 
partial  agonist  m-chlorophenylpiperazine 
in  chronic  alcoholic  patients  [letter].  Arch 
Gen  Psychiatry  48:383,1991. 

Bergen,  A.W.;  Kokoszka,  J.;  Peterson,  R; 
Long,  J.C.;  Virkkunen,  M.;  Linnoila,  M.; 
and  Goldman,  D.  Mu  opioid  receptor 
gene  variants:  Lack  of  association  with 
alcohol  dependence.  Mol  Psychiatry 
2:490^94,  1997. 

Bienkowski,  P.;  Iwinska,  K.;  Stefanski,  R; 
and  Kostowski,  W.  Discriminative  stimulus 
properties  of  ethanol  in  the  rat:  Differential 
effects  of  selective  and  nonselective  ben- 
zodiazepine receptor  agonists.  Pharmacol 
Biochem  Behav  58:969-973,  1997. 


378 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Blum,  K.;  Noble,  E.P.;  Sheridan,  P.J.; 
Montgomery,  A.;  Ritchie,  T.; 
Jagadeeswaran,  P.;  Nogami,  H.;  Briggs, 
A.H.;  and  Cohn,  J.B.  Allelic  association 
of  human  dopamine  D2  receptor  gene  in 
alcoholism  [see  comments].  JAMA 
263:2055-2060,  1990. 

Blum,  K;  Noble,  E.P.;  Sheridan,  P.J.; 
Finley,  O.;  Montgomery,  A.;  Ritchie,  T.; 
Ozkaragoz,  T.;  Fitch,  R.J.;  Sadlack,  F.; 
Sheffield,  D.;  et  al.  Association  of  the  Al 
allele  of  the  D2  dopamine  receptor  gene 
with  severe  alcoholism.  Alcohol  8:409- 
416,1991. 

Boess,  F.G.;  Monsma,  F.J.,  Jr.;  Carolo,  C; 
Meyer,  V.;  Rudler,  A.;  Zwingelstein,  C;  and 
Sleight,  A.J.  Functional  and  radioligand 
binding  characterization  of  rat  5-HT6 
receptors  stably  expressed  in  HEK293  cells. 
Neuropharmacology  36:713-720,  1997. 

Bohlen,  P.;  Huot,  S.;  and  Palfreyman, 
M.G.  The  relationship  between  GABA 
concentrations  in  brain  and  cerebrospinal 
fluid.  Brain  Res  167:297-305,  1979. 

Bolos,  A.M.;  Dean,  M.;  Lucas-Derse,  S.; 
Ramsburg,  M.;  Brown,  G.L.;  and  Goldman, 
D.  Population  and  pedigree  studies  reveal 
a  lack  of  association  between  the  dopamine 
D2  receptor  gene  and  alcoholism  [see  com- 
ments]. /AM4  264(24):3 156-3160, 1990. 

Borg,  S.;  Kvande,  H.;  Rydberg,  U.; 
Terenius,  L.;  and  Wahlstrom,  A. 
Endorphin  levels  in  human  cerebrospinal 
fluid  during  alcohol  intoxication  and 
withdrawal.  Psychopharmacology 
78:101-103,  1982. 

Borg,  S.;  Czarnecka,  A.;  Kvande,  FL; 
Mossberg,  D.;  and  Sedvall,  G.  Clinical 
conditions  and  concentrations  of  MOPEG 
in  the  cerebrospinal  fluid  and  urine  of 


male  alcoholic  patients  during  withdrawal. 
Alcohol  Clin  Exp  Res  7:411-415,  1983. 

Bosio,  A.;  Lucchi,  L.;  Spano,  P.F.;  and 
Trabucchi,  M.  Central  toxic  effects  of 
chronic  ethanol  treatment:  Actions  on 
GABA  and  benzodiazepine  recognition 
sites.  Toxicol  Lett  13:99-103,  1982. 

Branchey,  L.;  Shaw,  S.;  and  Lieber,  C.S. 
Ethanol  impairs  tryptophan  transport  into 
the  brain  and  depresses  serotonin.  Life  Sci 
29:2751-2755,  1981. 

Branchey,  L.;  Branchey,  M.;  Zucker,  D.; 
Shaw,  S.;  and  Lieber,  C.S.  Association 
between  low  plasma  tryptophan  and 
blackouts  in  male  alcoholic  patients. 
Alcohol  Clin  Exp  Res9:393-395,  1985. 

Brot,  M.D.;  Koob,  G.F.;  and  Britton, 
K.T.  Anxiolytic  effects  of  steroid  hor- 
mones during  the  estrous  cycle. 
Interactions  with  ethanol.  Recent  Dev 
Alcohol  12:243-259,  1995. 

Brot,  M.D.;  Akwa,  Y.;  Purdy,  R.H.; 
Koob,  G.F.;  and  Britton,  K.T.  The  anxi- 
olytic-like  effects  of  the  neurosteroid 
allopregnanolone:  Interactions  with 
GABA(A)  receptors.  Eur  J  Pharmacol 
325:1-7,  1997. 

Brown,  M.E.;  Anton,  R.F.;  Malcolm,  K; 
and  Ballenger,  J.C.  Alcohol  detoxification 
and  withdrawal  seizures:  Clinical  support 
for  a  kindling  hypothesis.  Biol  Psychiatry 
23:507-514,  1988. 

Bruno,  F.  Buspirone  in  the  treatment  of 
alcoholic  patients.  Psychopathology 22:49- 
59,  1989. 

Buck,  KJ.  New  insight  into  the  mechanisms 
of  ethanol  effects  on  GABAA  receptor 
function  and  expression,  and  their  relevance 


379 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


to  behavior.  Alcohol  Clin  Exp  il«20:198A- 
202A,  1996. 

Buck,  K.J.;  Hahner,  L.;  Sikela,  J.;  and 
Harris,  R.A.  Chronic  ethanol  treatment 
alters  brain  levels  of  gamma-aminobutyric 
acidA  receptor  subunit  mRNAs: 
Relationship  to  genetic  differences  in 
ethanol  withdrawal  seizure  severity.  / 
Neurochem  57:1452-1455,  1991. 

Buydens-Branchey,  L.;  Branchey,  M.H.; 
Noumair,  D.;  and  Lieber,  C.S.  Age  of 
alcoholism  onset.  II.  Relationship  to  sus- 
ceptibility to  serotonin  precursor  availabil- 
ity. Arch  Gen  Psychiatry  46:231-236, 1989. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin, 
C.  Craving  for  cocaine  in  addicted  users. 
Role  of  serotonergic  mechanisms.  Am  J 
Addict  6:65-73,  1997  a. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin, 
C.  Hormonal,  psychological,  and  alcohol 
craving  changes  after  m-chlorophenylpiper- 
azine  administration  in  alcoholics.  Alcohol 
Clin  Exp  Res  21:220-226,  19976. 

Buydens-Branchey,  L.;  Branchey,  M.; 
Fergeson,  P.;  Hudson,  J.;  and  McKernin, 
C.  The  meta-chlorophenylpiperazine 
challenge  test  in  cocaine  addicts:  Hormonal 
and  psychological  responses.  Biol 
Psychiatry  41:1071-1086,  1997c. 

Callahan,  P.M.,  and  Cunningham,  KA. 
Involvement  of  5-HT2C  receptors  in  medi- 
ating the  discriminative  stimulus  properties 
of  m-chlorophenylpiperazine  (mCPP). 
Eur  J  Pharmacol  257:27-38,  1994. 

Carpenter,  L.;  Anderson,  G.;  Pelton,  G.; 
Gudin,  J.;  Kirwin,  P.;  Price,  L.;  Heninger, 
G.;  and  McDougle,  C.  Tryptophan  deple- 


tion during  continuous  CSF  sampling  in 
healthy  human  subjects.  Neuropsycho- 
pharmacology  19:26-35,  1998. 

Chan,  R.J.;  McBride,  A.W.;  Thomasson, 
H.R.;  Ykenney,  A.;  and  Crabb,  D.W. 
Allele  frequencies  of  the  preproenkephalin 
A  (PENK)  gene  CA  repeat  in  Asians, 
African-Americans,  and  Caucasians:  Lack 
of  evidence  for  different  allele  frequencies 
in  alcoholics.  Alcohol  Clin  Exp  Res 
18(3):533-535,  1994. 

Chang,  F.M.;  Ko,  H.C.;  Lu,  R.B.;  Pakstis, 
A.J.;  and  Kidd,  K.K.  The  dopamine  D4 
receptor  gene  (DRD4)  is  not  associated 
with  alcoholism  in  three  Taiwanese  popu- 
lations: Six  polymorphisms  tested  sepa- 
rately and  as  haplotypes.  Biol  Psychiatry 
41(4):394^405,  1997. 

Charette,  L.;  Tate,  D.L.;  and  Wilson,  A. 
Alcohol  consumption  and  menstrual 
distress  in  women  at  higher  and  lower  risk 
for  alcoholism.  Alcohol  Clin  Exp  Res 
14:152-157,  1990. 

Chen,  H.T.;  Casanova,  M.F.;  Kleinman, 
J.E.;  Zito,  M.;  Goldman,  D.;  and 
Linnoila,  M.  3H-paroxetine  binding  in 
brains  of  alcoholics.  Psychiatry  Res 
38:293-299,  1991. 

Chick,  J.  Acamprosate  as  an  aid  in  the 
treatment  of  alcoholism.  Alcohol  Alcohol 
30:785-7,  1995. 

Chutuape,  M.A.,  and  de  Wit,  H.  Preferences 
for  ethanol  and  diazepam  in  anxious  indi- 
viduals: An  evaluation  of  the  self-medication 
hypothesis.  Psychopharmacology  (Berl) 
121:91-103,  1995. 

Cloninger,  C.R  Neurogenetic  adaptive 
mechanisms  in  alcoholism.  Science  236: 
410-416,  1987. 


380 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Coccaro,  E.F.;  Silverman,  J.M.;  Klar, 
H.M.;  Horvath,  T.B.;  and  Siever,  L.J. 
Familial  correlates  of  reduced  central 
serotonergic  system  function  in  patients 
with  personality  disorders.  Arch  Gen 
Psychiatry  51:318-324,  1994. 

Coffman,  J.A.,  and  Petty,  F.  Plasma 
GAB  A  levels  in  chronic  alcoholics.  Am  J 
Psychiatry  142:1204-1205,  1985. 

Conn,  P.J.,  and  Sanders-Bush,  E.  Relative 
efficacies  of  piperazines  at  the  phospho- 
inositide  hydrolysis-linked  serotonergic 
(5-HT-2  and  5-HT-lc)  receptors.  / 
Pharmacol  Exp  Ther  242:552-557 ',  1987. 

Cowley,  D.S.;  Roy-Byrne,  P.P.;  Godon, 
C;  Greenblatt,  D.J.;  Ries,  R;  Walker,  R.D.; 
Samson,  H.H.;  and  Hommer,  DW. 
Response  to  diazepam  in  sons  of  alcoholics. 
Alcohol  Clin  Exp  Res  16:1057-1063,  1992. 

Cowley,  D.S.;  Roy- Byrne,  P.P.;  Radant, 
A.;  Hommer,  D.W.;  Greenblatt,  D.J.; 
Vitaliano,  P.P.;  and  Godon,  C.  Eye  move- 
ment effects  of  diazepam  in  sons  of  alco- 
holic fathers  and  male  control  subjects. 
Alcohol  Clin  Exp  Res  18:324-332,  1994. 

Cowley,  D.S.;  Roy- Byrne,  P.P.;  Greenblatt, 
D.J.;  Kramer,  G.L.;  and  Petty,  F.  Effect 
of  diazepam  on  plasma  gamma-aminobu- 
tyric  acid  in  sons  of  alcoholic  fathers. 
Alcohol  Clin  Exp  Res  20:343-347,  1996. 

Cummins,  J.T.;  Sack,  M.;  and  von  Hungen, 
K.  The  effect  of  chronic  ethanol  on  gluta- 
mate  binding  in  human  and  rat  brain.  Life 
Sci  47:877-882,  1990. 

Daoust,  M.;  Lhuintre,  J. P.;  Ernouf,  D.; 
Legrand,  E.;  Breton,  P.;  and  Boucly,  P. 
Ethanol  intake  and  3H-serotonin  uptake. 
II:  A  study  in  alcoholic  patients  using 


platelets  3H-paroxetine  binding.  Life  Sci 
48:1977-1983,  1991. 

Davidson,  D.;  Swift,  R.;  and  Fitz,  E. 
Naltrexone  increases  the  latency  to  drink 
alcohol  in  social  drinkers.  Alcohol  Clin 
Exp  Res  20:732-739,  1996. 

De  Bias,  A.L.  Brain  GABAA  receptors 
studied  with  subunit-specific  antibodies. 
Molecular  Neurobiology  12:55-71,  1996. 

Delgado,  P.L.;  Charney,  D.S.;  Price, 
L.H.;  Landis,  H.;  and  Heninger,  G.R. 
Neuroendocrine  and  behavioral  effects  of 
dietary  tryptophan  restriction  in  healthy 
subjects.  Life  Sci  45:2323-2332,  1989. 

Delgado,  P.L.;  Charney,  D.S.;  Price, 
L.H.;  Aghajanian,  G.K.;  Landis,  H.;  and 
Heninger,  G.R.  Serotonin  function  and 
the  mechanism  of  antidepressant  action. 
Reversal  of  antidepressant-induced  remis- 
sion by  rapid  depletion  of  plasma  trypto- 
phan. Arch  Gen  Psychiatry  47:411^118, 
1990. 

Devaud,  L.L.;  Purdy,  R.H.;  and  Morrow, 
A.L.  The  neurosteroid,  3  alpha-hydroxy-5 
alpha-pregnan-20-one,  protects  against 
bicuculline-induced  seizures  during 
ethanol  withdrawal  in  rats.  Alcohol  Clin 
£*#  21*19:350-355,  1995^. 

Devaud,  L.L.;  Smith,  F.D.;  Grayson, 
D.R;  and  Morrow,  A.L.  Chronic  ethanol 
consumption  differentially  alters  the  ex- 
pression of  gamma-  aminobutyric  acidA 
receptor  subunit  mRNAs  in  rat  cerebral 
cortex:  Competitive,  quantitative  reverse 
transcriptase-polymerase  chain  reaction 
analysis.  Mol Pharmacol 48:861-868, 
19956. 

de  Wit,  H.  Diazepam  preference  in  males 
with  and  without  an  alcoholic  first-degree 


381 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


relative.  Alcohol  Clin  Exp  Res  15:593- 
600,1991. 

de  Wit,  H.,  and  McCracken,  S.G.  Ethanol 
self- administration  in  males  with  and 
without  an  alcoholic  first-degree  relative. 
Alcohol  Clin  Exp  Res  14:63-70,  1990. 

de  Wit,  H.,  and  Rukstalis,  M.  Acute  ef- 
fects of  triazolam  in  women:  Relationships 
with  progesterone,  estradiol  and  allopreg- 
nanolone.  Psychopharmacology  (Berl) 
130:69-78,  1997. 

de  Wit,  H.;  Metz,  J.;  Wagner,  N.;  and 
Cooper,  M.  Behavioral  and  subjective 
effects  of  ethanol:  Relationship  to  cerebral 
metabolism  using  PET.  Alcohol  Clin  Exp 
Res  14:482-489,  1990. 

Dodd,  P.R.;  Kril,  J.J.;  Thomas,  G.J.; 
Watson,  W.E.;  Johnston,  G.A.;  and 
Harper,  C.G.  Receptor  binding  sites  and 
uptake  activities  mediating  GABA  neuro- 
transmission in  chronic  alcoholics  with 
Wernicke  encephalopathy.  Brain  Res 
710:215-228,  1996. 

Doty,  P.;  Kirk,  J.M.;  Cramblett,  M.J.;  and 
de  Wit,  H.  Behavioral  responses  to  ethanol 
in  light  and  moderate  social  drinkers 
following  naltrexone  pretreatment.  Drug 
Alcohol  Depend  47:109-116,  1997. 

D'Souza,  D.C.;  Charney,  D.S.;  and 
Krystal,  J.H.  Glycine  site  agonists  of  the 
NMDA  receptor:  A  review.  CNS  Drug 
Rev  1:227-260,  1995. 

Durcan,  M.J.;  Lister,  R.G.;  and  Linnoila, 
M.  Evidence  for  central  alpha-2  adreno- 
ceptors, not  imidazoline  binding  sites, 
mediating  the  ethanol-attenuating  proper- 
ties of  alpha-2  adrenoceptor  antagonists.  / 
Pharmacol  Exp  Ther  258:576-582,  1991. 


Estevez,  F.;  Parrillo,  S.;  Giusti,  M.;  and 
Monti,  J.M.  Single-dose  ritanserin  and 
alcohol  in  healthy  volunteers:  A  placebo- 
controlled  trial.  Alcohol  12:541-545,  1995. 

Faraj,  B.A.;  Olkowski,  Z.L.;  and  Jackson, 
R.T.  Prevalence  of  high  serotonin  uptake 
in  lymphocytes  of  abstinent  alcoholics. 
Biochem  Pharmacol  53:53-57,  1997. 

Farren,  C.K.,  and  Dinan,  T.G.  Elevated 
tryptophan  levels  in  post-withdrawal 
alcoholics.  Acta  Psychiatr  Scand 
94:465-170,  1996. 

Farren,  C.K.;  Clare,  A.W.;  Ziedonis,  D.; 
Hammeedi,  F.A.;  and  Dinan,  T.G. 
Evidence  for  reduced  dopamine  D2  re- 
ceptor sensitivity  in  postwithdrawal  alco- 
holics. Alcohol  Clin  Exp  Res  19:1520- 
1524,  1995*. 

Farren,  C.K.;  Ziedonis,  D.;  Clare,  A.W.; 
Hammeedi,  F.A.;  and  Dinan,  T.G.  D- 
fenfluramine-induced  prolactin  responses 
in  postwithdrawal  alcoholics  and  controls. 
Alcohol  Clin  Exp  Res  19:1578-1582, 19956. 

Fernstrom,  J.D.  Role  of  precursor  availabil- 
ity in  control  of  monoamine  biosynthesis 
in  brain.  Physiol  Rev  63:484-546,  1983. 

Fernstrom,  M.H.,  and  Fernstrom,  J.D. 
Acute  tyrosine  depletion  reduces  tyrosine 
hydroxylation  rate  in  rat  central  nervous 
system.  Life  Sci  57:PL97-PL102,  1995. 

Fitzgerald,  L.W.,  and  Nestler,  E.J.  Mole- 
cular and  cellular  adaptations  in  signal 
transduction  pathways  following  ethanol 
exposure.  Clin  Neurosci  3:165-173,  1995. 

Freund,  G.,  and  Anderson,  K.J.  Glutamate 
receptors  in  the  frontal  cortex  of  alcoholics. 
Alcohol  Clin  Exp  Res  20:1165-1172,  1996. 


382 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Freund,  G.,  and  Ballinger,  W.E.,  Jr. 
Decrease  of  benzodiazepine  receptors  in 
frontal  cortex  of  alcoholics.  Alcohol 
5:275-282,  1988. 

Freund,  G.,  and  Ballinger,  W.E.  Loss  of 
synaptic  receptors  can  precede  morpho- 
logic changes  induced  by  alcoholism. 
Alcohol  Alcohol  Suppl  1:385-391,  1991. 

Friedman,  M.J.;  Krstulovic,  A.M.; 
Severinghaus,  J.M.;  and  Brown,  S.J. 
Altered  conversion  of  tryptophan  to 
kynurenine  in  newly  abstinent  alcoholics. 
Biol  Psychiatry  23:89-93,  1988. 

Garbutt,  J.C.;  Miller,  L.P.;  Kramer,  G.; 
Davis,  L.L.;  Mason,  G.A.;  Prange,  A.J., 
Jr.;  and  Petty,  F.  Increased  serum 
gamma-aminobutyric  acid  (GABA)  levels 
in  young  men  at  high-risk  for  alcoholism. 
Biol  Psychiatry  38:704-706, 1995. 

Geijer,  T.;  Neiman,  J.;  Rydberg,  U.; 
Gyllander,  A.;  Jonsson,  E.;  Sedvall,  G.; 
Valverius,  P.;  and  Terenius,  L.  Dopamine 
D2-receptor  gene  polymorphisms  in 
Scandinavian  chronic  alcoholics.  Eur  Arch 
Psychiatry  Clin  Neurosci  244(1) :26-32, 1994. 

Gejman,  P.V.;  Ram,  A.;  Gelernter,  J.; 
Friedman,  E.;  Cao,  Q.;  Pickar,  D.;  Blum,  K; 
Noble,  E.P.;  Kranzler,  H.R.;  O'Malley,  S.; 
et  al.  No  structural  mutation  in  the  dopa- 
mine D2  receptor  gene  in  alcoholism  or 
schizophrenia.  Analysis  using  denaturing 
gradient  gel  electrophoresis.  JAMA 
271(3):204-208,  1994. 

Gelernter,  J.;  O'Malley,  S.;  Risch,  N; 
Kranzler,  H.R.;  Krystal,  J.;  Merikangas, 
K;  Kennedy,  J.L.;  and  Kidd,  K.K.  No 
association  between  an  allele  at  the  D2 
dopamine  receptor  gene  (DRD2)  and 
alcoholism  [see  comments].  JAMA 
266(13):1801-1807,  1991. 


Gelernter,  J.;  Goldman,  D.;  and  Risch,  N. 
The  Al  allele  at  the  D2  dopamine  receptor 
gene  and  alcoholism.  A  reappraisal  [see 
comments].  JAMA  269:1673-1677, 1993. 

Gelernter,  J.;  Kranzler,  H.;  and  Cubells, 
J.F.  Serotonin  transporter  protein 
(SLC6A4)  allele  and  haplotype  frequen- 
cies and  linkage  disequilibria  in  African- 
and  European-American  and  Japanese 
populations  and  in  alcohol- dependent 
subjects.  Hum  Genet  101:243-246,  1997. 

Genazzani,  A.R.;  Nappi,  G.;  Facchinetti, 
F.;  Mazzella,  G.L.;  Parrini,  D.;  Sinforiani, 
E.;  Petraglia,  F.;  and  Savoldi,  F.  Central 
deficiency  of  beta-endorphin  in  alcohol 
addicts.  /  Clin  Endocrinol  Metab  55:583- 
586,  1982. 

George,  D.T.;  Nutt,  D.J.;  Rawlings,  RR; 
Phillips,  M.J.;  Eckardt,  M.J.;  Potter,  W.Z.; 
and  Linnoila,  M.  Behavioral  and  endocrine 
responses  to  clomipramine  in  panic  disor- 
der patients  with  or  without  alcoholism. 
Biol  Psychiatry  37:112-119,  1995. 

George,  D.T.;  Benkelfat,  C;  Rawlings, 
RR;  Eckardt,  M.J.;  Phillips,  M.J.;  Nutt, 
D.J.;  Wynne,  D.;  Murphy,  D.L.;  and 
Linnoila,  M.  Behavioral  and  neuroen- 
docrine responses  to  m-chlorophenyl- 
piperazine  in  subtypes  of  alcoholics  and  in 
healthy  comparison  subjects.  Am  J 
Psychiatry  154:81-87,  1997. 

Gerra,  G.;  Giucasto,  G.;  Zaimovic,  A.; 
Fertonani,  G.;  Chittolini,  B.;  Avanzini,  P.; 
Caccavari,  R;  and  Delsignore,  R 
Intravenous  flumazenil  following  pro- 
longed exposure  to  lormetazepam  in 
humans:  Lack  of  precipitated  withdrawal. 
Int  Clin  Psychopharmacol  11:81-88, 
1996. 


383 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Gianoulakis,  C;  de  Waele,  J. P.;  and 
Thavundayil,  J.  Implication  of  the  endo- 
genous opioid  system  in  excessive  ethanol 
consumption.  Alcohol  13:19-23,  1996. 

Gilman,  S.;  Koeppe,  R.A.;  Adams,  K.; 
Johnson-Greene,  D.;  Junck,  L.;  Kluin, 
K.J.;  Brunberg,  J.;  Martorello,  S.;  and 
Lohman,  M.  Positron  emission  tomo- 
graphic studies  of  cerebral  benzodiazepine- 
receptor  binding  in  chronic  alcoholics. 
Ann  Neurol  40: 163-171,  1996. 

Glue,  P.;  Sellman,  J.D.;  Nicholls,  M.G.; 
Abbott,  R;  Joyce,  P.R.;  and  Nutt,  D.J. 
Studies  of  alpha-2-adrenoceptor  function 
in  abstinent  alcoholics.  Br  J  Addict 
84:97-102,  1989. 

Goldman,  D.;  Urbanek,  M.;  Guenther, 
D.;  Robin,  R.;  and  Long,  J.C.  Linkage 
and  association  of  a  functional  DRD2 
variant  [Ser311Cys]  and  DRD2  markers 
to  alcoholism;  substance  abuse  and  schiz- 
ophrenia in  Southwestern  American  Indians. 
Am  J  Med  Genet  74(4):386-394,  1997. 

Goldman,  G.D.;  Volicer,  L.;  Gold,  B.I.; 
and  Roth,  R.H.  Cerebrospinal  fluid 
GABA  and  cyclic  nucleotides  in  alcoholics 
with  and  without  seizures.  Alcohol  Clin 
Exp  Res  5:431-434,  1981. 

Gorwood,  P.;  Martres,  M.P.;  Ades,  J.; 
Sokoloff,  P.;  Noble,  E.P.;  Geijer,  T.;  Blum, 
K.;  Neiman,  J.;  Jonsson,  E.;  Feingold,  J.; 
et  al.  Lack  of  association  between  alcohol- 
dependence  and  D3  dopamine  receptor 
gene  in  three  independent  samples.  Am  J 
Med  G^r60(6):529-531,  1995. 

Grant,  K.A.  The  role  of  5-HT3  receptors 
in  drug  dependence.  Drug  Alcohol  Depend 
38:155-171,1995. 


Grant,  K.A.,  and  Barrett,  J.E.  Blockade  of 
the  discriminative  stimulus  effects  of  ethanol 
with  5-HT3  receptor  antagonists.  Psycho  - 
pharmacology  104:451-456,  1991. 

Grant,  K.A.,  and  Colombo,  G.  Discrim- 
inative stimulus  effects  of  ethanol:  Effect 
of  training  dose  on  the  substitution  of  N- 
methyl-D -aspartate  antagonists.  J  Pharmacol 
Exp  Ther  264:1241-1247,  1993a. 

Grant,  K.A.,  and  Colombo,  G.  Pharma- 
cological analysis  of  the  mixed  discrimina- 
tive stimulus  effects  of  ethanol.  Alcohol 
Alcohol  Suppl  2 :445-449,  1993£. 

Grant,  K.A.,  and  Lovinger,  D.M.  Cellular 
and  behavioral  neurobiology  of  alcohol: 
Receptor-mediated  neuronal  processes. 
Clin  Neurosci  3:155-164,  1995. 

Grant,  K.A.;  Azarov,  A.;  Bowen,  C.A.; 
Mirkis,  S.;  and  Purdy,  RH.  Ethanol-like 
discriminative  stimulus  effects  of  the 
neurosteroid  3  alpha-hydroxy-5  alpha- 
pregnan-20-one  in  female  Macaca  fascicu- 
laris  monkeys.  Psychopharmacology  (Berl) 
124:340-346,  1996a. 

Grant,  K.A.;  Colombo,  G.;  Grant,  J.;  and 
Rogawski,  M.A.  Dizocilpine-like  discrimi- 
native stimulus  effects  of  low-affinity 
uncompetitive  NMDA  antagonists. 
Neuropharmacology  35:1709-1719,  \996b. 

Grant,  K.A.;  Azarov,  A.;  Shively,  C.A.; 
and  Purdy,  R.H.  Discriminative  stimulus 
effects  of  ethanol  and  3  alpha-hydroxy-5 
alpha-pregnan-20-one  in  relation  to  men- 
strual cycle  phase  in  cynomolgus  monkeys 
(Macaca  fascicularis) .  Psychopharmacolqgy 
(Berl)  130:59-68,  1997a. 

Grant,  K.A.;  Colombo,  G.;  and  Gatto, 
G.J.  Characterization  of  the  ethanol-like 
discriminative  stimulus  effects  of  5-HT 


384 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


receptor  agonists  as  a  function  of  ethanol 
training  dose.  Psychopharmacology  (Berl) 
133:133-141,  1997b. 

Grant,  S.;  London,  E.D.;  Newlin,  D.B.; 
Villemagne,  V.L.;  Liu,  X.;  Contoreggi, 
C;  Phillips,  R.L.;  Kimes,  A.S.;  and  Margolin, 
A.  Activation  of  memory  circuits  during 
cue-elicited  cocaine  craving.  Proc  Natl 
AcadSci  USA  93:12040-120405,  1996. 

Griffith,  J.D.;  Jasinski,  D.R.;  Casten, 
G.P.;  and  McKinney,  G.R.  Investigation 
of  the  abuse  liability  of  buspirone  in  alco- 
hol-dependent patients.  Am  J  Med 
80:30-35,  1986. 

Griffiths,  R.R.;  Evans,  S.M.;  Guarino,  J.J.; 
Roache,  J.D.;  Furman,  W.R;  Liebson,  I.; 
and  Schwam,  E.M.  Intravenous  flumaze- 
nil  following  acute  and  repeated  exposure 
to  lorazepam  in  healthy  volunteers:  Anta- 
gonism and  precipitated  withdrawal.  / 
Pharmacol  Exp  Ther  265:1 163-1174,  1993. 

Grove,  J.;  Palfreyman,  M.G.;  and 
Schechter,  P.J.  Cerebrospinal  fluid  GABA 
as  an  index  of  brain  GABA  activity.  Clin 
Neuropharmacol  6:223-229,  1983. 

Gulya,  K.;  Grant,  K.A.;  Valverius,  P.; 
Hoffman,  P.L.;  and  Tabakoff,  B.  Brain 
regional  specificity  and  time -course  of 
changes  in  the  NMDA  receptor-ionophore 
complex  during  ethanol  withdrawal.  Brain 
Res  547:129-134,  1991. 

Halliday,  G.;  Ellis,  J.;  Heard,  R;  Caine, 
D.;  and  Harper,  C.  Brainstem  serotoner- 
gic neurons  in  chronic  alcoholics  with  and 
without  the  memory  impairment  of 
Korsakoff's  psychosis.  /  Neuropathol  Exp 
Neurol  52:567-579,  1993. 

Handelsman,  L.;  Holloway,  K;  Kahn,  RS.; 
Sturiano,  C;  Rinaldi,  P.J.;  Bernstein,  D.P.; 


Siever,  L.;  Gabriel,  S.;  and  Cooper,  T.B. 
Hostility  is  associated  with  a  low  prolactin 
response  to  meta-chlorophenylpiperazine 
in  abstinent  alcoholics.  Alcohol  Clin  Exp 
Res  20:824-829,  1996. 

Hawley,  R.J.;  Major,  L.F.;  Schulman,  E.; 
Trocha,  P.J.;  Takenaga,  J.K.;  and  Catravas, 
G.N.  Cerebrospinal  fluid  cyclic  nucleotides 
and  GABA  do  not  change  in  alcohol 
withdrawal.  Life  Sci  28:295-299,  1981. 

Heinz,  A.;  Lichtenberg-Kraag,  B.;  Baum, 
S.S.;  Graf,  R;  Kruger,  R;  Dettling,  M.;  and 
Rommelspacher,  H.  Evidence  for  prolonged 
recovery  of  dopaminergic  transmission 
after  detoxification  in  alcoholics  with  poor 
treatment  outcome.  /  Neural  Transm  Gen 
Section  102:149-157,  1995. 

Hemmingsen,  R;  Braestrup,  C;  Nielsen, 
M.;  and  Barry,  D.I.  The  benzodiazepine/ 
GABA  receptor  complex  during  severe 
ethanol  intoxication  and  withdrawal  in  the 
rat.  Acta  Psychiatr  Scand  65:120-126, 1982. 

Herz,  A.  Endogenous  opioid  systems  and 
alcohol  addiction.  Psychopharmacology 
(Berl)  129:99-111,  1997. 

Hetherington,  H.P.;  Pan,  J.W.;  Mason, 
G.F.;  Adams,  D.;  Vaughn,  M.J.;  Twieg, 
D.B.;  and  Pohost,  G.M.  Quantitative  1H 
spectroscopic  imaging  of  human  brain  at 
4.1  T  using  image  segmentation.  Magn 
Reson  Med  36:21-29,  1996. 

Higuchi,  S.;  Muramatsu,  T.;  Murayama, 
M.;  and  Hayashida,  M.  Association  of 
structural  polymorphism  of  the  dopamine 
D2  receptor  gene  and  alcoholism.  Biochem 
BiophysRes  Commun  204:1199-1205, 1994. 

Hommer,  D.;  Andreasen,  P.;  Rio,  D.; 
Williams,  W.;  Ruttimann,  U.;  Momenan,  R; 
Zametkin,  A.;  Rawlings,  R;  and  Linnoila, 


385 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


M.  Effects  of  m-chlorophenylpiperazine 
on  regional  brain  glucose  utilization:  A 
positron  emission  tomographic  compari- 
son of  alcoholic  and  control  subjects.  J 
Neurosci  17:2796-2806,  1997. 

Honkanen,  A.;  Vilamo,  L.;  Wegelius,  K.; 
Sarviharju,  M.;  Hyytia,  P.;  and  Korpi, 
E.R.  Alcohol  drinking  is  reduced  by  a  mu 
1-  but  not  by  a  delta- opioid  receptor 
antagonist  in  alcohol-preferring  rats.  Eur 
J  Pharmacol  304:7-13 ,  1996. 

Hunt,  W.A.  The  effect  of  ethanol  on 
GABAergic  transmission.  Neurosci  Biobehav 
Rev  7:87-95,  1983. 

Hunt,  W.A.,  and  Majchrowicz,  E.  Studies 
of  neurotransmitter  interactions  after  acute 
and  chronic  ethanol  administration.  Pharm- 
acol Biochem  Behav  18:371-374,  1983. 

Jagannathan,  N.R.;  Desai,  N.G.;  and 
Raghunathan,  P.  Brain  metabolite  changes 
in  alcoholism:  An  in  vivo  proton  magnetic 
resonance  spectroscopy  (MRS)  study. 
Magn  Reson  Imaging  14:553-557,  1996. 

Johnson,  B.A.;  Jasinski,  D.R.;  Galloway, 
G.P.;  Kranzler,  H.;  Weinreib,  R;  Anton, 
R.F.;  Mason,  B.J.;  Bohn,  M.J.;  Pettinati, 
H.M.;  Rawson,  R.;  and  Clyde,  C. 
Ritanserin  in  the  treatment  of  alcohol 
dependence — a  multi-center  clinical  trial. 
Ritanserin  Study  Group.  Psychopharmacology 
(Berl)  128:206-215,  1996. 

June,  H.L.;  Hughes,  R.W.;  Spurlock, 
H.L.;  and  Lewis,  M.J.  Ethanol  self-ad- 
ministration in  freely  feeding  and  drinking 
rats:  Effects  of  Rol5-4513  alone,  and  in 
combination  with  Rol5-1788  (flumazenil). 
Psychopharmacology  115:332-339,  1994. 

Keller,  E.;  Cummins,  J.T.;  and  von 
Hungen,  K.  Regional  effects  of  ethanol 


on  glutamate  levels,  uptake  and  release  in 
slice  and  synaptosome  preparations  from 
rat  brain.  Subst  Alcohol  Actions  Misuse 
4:383-392,  1983. 

Khanna,  J.M.;  Morato,  G.S.;  Chau,  A.; 
and  Shah,  G.  D-cycloserine  enhances  rapid 
tolerance  to  ethanol  motor  incoordina- 
tion. Pharmacol  Biochem  Behav 
52:609-614,  1995. 

King,  A.C.;  Volpicelli,  J.R.;  Frazer,  A.; 
and  O'Brien,  C.P.  Effect  of  naltrexone  on 
subjective  alcohol  response  in  subjects  at 
high  and  low  risk  for  future  alcohol  de- 
pendence. Psychopharmacology  (Berl) 
129:15-22,  1997. 

Koob,  G.F.  Drug  abuse  and  alcoholism. 
Overview.  Adv  Pharmacol  42:969- 
977,  1998. 

Koob,  G.F.;  Rassnick,  S.;  Heinrichs,  S.; 
and  Weiss,  F.  Alcohol,  the  reward  system 
and  dependence.  EXS  71:103-114,  1994. 

Korpi,  E.R.  Role  of  GABAA  receptors  in 
the  actions  of  alcohol  and  in  alcoholism: 
Recent  advances.  Alcohol  Alcohol  29:115- 
129,  1994. 

Kranzler,  H.R.;  Burleson,  J.A.;  Del  Boca, 
F.K.;  Babor,  T.F.;  Korner,  P.;  Brown,  J.; 
and  Bohn,  M.J.  Buspirone  treatment  of 
anxious  alcoholics.  A  placebo-controlled 
trial.  Arch  Gen  Psychiatry  51:720-731,  1994. 

Kranzler,  H.R.;  Burleson,  J.A.;  Brown,  J.; 
and  Babor,  T.F.  Fluoxetine  treatment 
seems  to  reduce  the  beneficial  effects  of 
cognitive-behavioral  therapy  in  type  B 
alcoholics.  Alcohol  Clin  Exp  Res 20: 1534- 
1541,  1996. 

Kril,  J.J.;  Dodd,  P.R.;  Gundlach,  A.L.; 
Davies,  N.;  Watson,  W.E.;  Johnston,  G.A.; 
and  Harper,  C.G.  Necropsy  study  of  GABA/ 


386 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


benzodiazepine  receptor  binding  sites  in 
brain  tissue  from  chronic  alcoholic  patients. 
Clin  Exp  Neurol 25:135-141,  1988. 

Krystal,  J.H.;  Webb,  E.;  Cooney,  N.;  Kranzler, 
H.R.;  and  Charney,  D.S.  Specificity  of 
ethanollike  effects  elicited  by  serotonergic 
and  noradrenergic  mechanisms.  Arch  Gen 
Psychiatry  51:898-911,  1994. 

Krystal,  J.;  Karper,  L.;  D'Souza,  D.C.; 
Webb,  E.;  Bennett,  A.;  Abi-Dargham,  A.; 
Petrakis,  I.L.;  Brenner,  L.;  Morrissey,  K.; 
Abi-Saab,  D.;  and  Charney,  D.S. 
Differentiating  NMDA  dysregulation  in 
schizophrenia  and  alcoholism  using  keta- 
mine .  Schizophr  Res  15:156,  1995. 

Krystal,  J.H.;  Webb,  E.;  Cooney,  N.L.; 
Kranzler,  H.R;  Southwick,  S.W.;  Heninger, 
G.R.;  and  Charney,  D.S.  Serotonergic 
and  noradrenergic  dysregulation  in  alco- 
holism: m-Chlorophenylpiperazine  and 
yohimbine  effects  in  recently  detoxified 
alcoholics  and  healthy  comparison  sub- 
jects. Am  J  Psychiatry  153:83-92,  1996. 

Krystal,  J.H.;  Webb,  E.;  Grillon,  C; 
Cooney,  N.;  Casal,  L.;  Morgan,  C.A., 
3rd;  Southwick,  S.M.;  Davis,  M.;  and 
Charney,  D.S.  Evidence  of  acoustic  startle 
hyperreflexia  in  recently  detoxified  early 
onset  male  alcoholics:  Modulation  by 
yohimbine  and  m-chlorophenylpiperazine 
(mCPP).  Psychopharmacology  (Berl) 
131(3):207-215,  1997. 

Krystal,  J.H.;  Petrakis,  I.L.;  Webb,  E.; 
Cooney,  N.L.;  Karper,  L.P.;  Namanworth, 
S.;  Trevisan,  L.A.;  and  Charney,  D.S. 
Dose-related  ethanol-like  effects  of  the 
NMDA  antagonist,  ketamine,  in  recently 
detoxified  alcoholics.  Arch  Gen  Psychiatry 
55:354-360,  1998a. 


Krystal,  J.H.;  Petrakis,  I.L.;  Krasnicki,  S.; 
Trevisan,  L.;  Boutros,  N.;  and  D'Souza, 
D.C.  Altered  responses  to  agonists  of  the 
strychnine -insensitive  glycine  NMDA 
coagonist  (SIGLY)  site  in  recently  detoxi- 
fied alcoholics.  Alcohol  Clin  Exp  Res 
22:44A  (Abstract  245),  19986. 

Kulonen,  E.  Ethanol  and  GABA.  Med 
Biol  61:147-167 ,1983. 

Lawford,  B.R.;  Young,  R.M.;  Rowell, 
J.A.;  Qualichefski,  J.;  Fletcher,  B.H.; 
Syndulko,  K.;  Ritchie,  T.;  and  Noble, 
E.P.  Bromocriptine  in  the  treatment  of 
alcoholics  with  the  D2  dopamine  receptor 
Al  allele  [see  comments].  Nat  Med 
1:337-341,  1995. 

Lechtenberg,  R,  and  Worner,  T.M.  Relative 
kindling  effect  of  detoxification  and  non- 
detoxification  admissions  in  alcoholics. 
Alcohol  Alcohol  26:221-225,  1991. 

Lee,  M.A.,  and  Meltzer,  H.Y.  Neuro- 
endocrine responses  to  serotonergic 
agents  in  alcoholics.  Biol  Psychiatry 
30:1017-1030,  1991. 

Lewohl,  J.M.;  Crane,  D.I.;  and  Dodd, 
P.R  Alcohol,  alcoholic  brain  damage,  and 
GABAA  receptor  isoform  gene  expres- 
sion. Neurochem  Int  29:677-684,  1996. 

Lewohl,  J.M.;  Crane,  D.I.;  and  Dodd, 
P.R.  Zolpidem  binding  sites  on  the 
GABA(A)  receptor  in  brain  from  human 
cirrhotic  and  non-cirrhotic  alcoholics.  Eur 
J  Pharmacol  326:265-272,  1997. 

Lheureux,  P.,  and  Askenasi,  R.  Efficacy  of 
flumazenil  in  acute  alcohol  intoxication: 
Double  blind  placebo-controlled  evalua- 
tion. Hum  Exp  Toxicol  10:235-239,  1991. 

Lister,  R.G.;  Durcan,  M.J.;  Nutt,  D.J.; 
and  Linnoila,  M.  Attenuation  of  ethanol 


387 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


intoxication  by  alpha-2  adrenoceptor 
antagonists.  Life  Set  44: 1 1 1-1 19,  1989. 

Littleton,  J.  Acamprosate  in  alcohol  de- 
pendence: How  does  it  work?  Addiction 
90:1179-1188,  1995. 

Litton,  J.E.;  Neiman,  J.;  Pauli,  S.;  Farde, 
L.;  Hindmarsh,  T.;  Halldin,  C;  and  Sedvall, 
G.  PET  analysis  of  [llC]flumazenil  bind- 
ing to  benzodiazepine  receptors  in  chronic 
alcohol-dependent  men  and  healthy  con- 
trols .  Psychiatry  Res  50:1-13,  1993. 

Lovinger,  D.M.  Ethanol  potentiates  ion 
current  mediated  by  5-HT3  receptors  on 
neuroblastoma  cells  and  isolated  neurons. 
Alcohol  Alcohol  Suppl  1:181-185,  1991. 

Lovinger,  D.M.  Alcohols  and  neurotrans- 
mitter gated  ion  channels:  Past,  present 
and  future.  Naunyn  Schmiedebergs  Arch 
Pharmacol  356:267-282,  1997. 

Luddens,  H.,  and  Korpi,  E.R  Biological 
function  of  GABAA/benzodiazepine 
receptor  heterogeneity.  J  Psychiatr  Res 
29:77-94,  1995. 

Madamba,  S.G.;  Schweitzer,  P.; 
Zieglgansberger,  W.;  and  Siggins,  G.R. 
Acamprosate  (calcium  acetylhomotaurinate) 
enhances  the  N-methyl-D-aspartate  com- 
ponent of  excitatory  neurotransmission  in 
rat  hippocampal  CA1  neurons  in  vitro. 
Alcohol  Clin  Exp  Res  20:65 1-658,  1996. 

Major,  L.F.;  Ballenger,  J.C.;  Goodwin, 
F.K.;  and  Brown,  G.L.  Cerebrospinal 
fluid  homovanillic  acid  in  male  alcoholics: 
Effects  of  disulfiram.  Biol  Psychiatry 
12:635-642,  1977. 

Malcolm,  R.;  Anton,  R.F.;  Randall,  C.L.; 
Johnston,  A.;  Brady,  K.;  and  Thevos,  A.  A 
placebo-controlled  trial  of  buspirone  in 


anxious  inpatient  alcoholics.  Alcohol  Clin 
Exp  Res  16:1007-1013,  1992. 

Malec,  E.;  Malec,  T.;  Gagne,  M.A.;  and 
Dongier,  M.  Buspirone  in  the  treatment 
of  alcohol  dependence:  A  placebo-con- 
trolled trial.  Alcohol  Clin  Exp  Res  20:307- 
312,  1996. 

Malhotra,  A.K.;  Virkkunen,  M.;  Rooney, 
W.;  Eggert,  M.;  Linnoila,  M.;  and 
Goldman,  D.  The  association  between  the 
dopamine  D4  receptor  (D4DR)  16  amino 
acid  repeat  polymorphism  and  novelty 
seeking.  Mol  Psychiatry  1:388-391,  1996. 

Mason,  B.J.;  Ritvo,  E.C.;  Morgan,  R.O.; 
Salvato,  F.R.;  Goldberg,  G.;  Welch,  B.; 
and  Mantero-Atienza,  E.  A  double-blind, 
placebo -controlled  pilot  study  to  evaluate 
the  efficacy  and  safety  of  oral  nalmefene 
HC1  for  alcohol  dependence.  Alcohol  Clin 
Exp  Res  18:1162-1167,  1994. 

McDougle,  C.J.;  Krystal,  J.H.;  Price,  L.H.; 
Heninger,  G.R.;  and  Charney,  D.S.  Nora- 
drenergic response  to  acute  ethanol  admin- 
istration in  healthy  subjects,  comparison 
with  intravenous  yohimbine.  Psycho- 
pharmacology  (Berl)  118:127-135,  1995. 

Meert,  T.F.  Ritanserin  and  alcohol  abuse 
and  dependence.  Alcohol  Alcohol  Suppl 
2:523-530,  1994. 

Melchior,  C.L.,  and  Allen,  P.M.  Interaction 
of  pregnanolone  and  pregnenolone  sulfate 
with  ethanol  and  pentobarbital.  Pharmacol 
Biochem  Behav  42:605-611 ,  1992. 

Melchior,  C.L.,  and  Ritzmann,  RF. 
Dehydroepiandrosterone  enhances  the 
hypnotic  and  hypothermic  effects  of 
ethanol  and  pentobarbital.  Pharmacol 
Biochem  Behav  43:223-227,  1992. 


388 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Mellerup,  E.T.;  Bech,  P.;  Lauritzen,  L.; 
Lunde,  M.;  and  Plenge,  P.  Platelet  parox- 
etine binding  in  alcoholics.  Alcohol 
Alcohol  27:603-606,  1992. 

Mendelson,  J.;  Jones,  R.T.;  Upton,  R.; 
and  Jacob,  P.R.  Methamphetamine  and 
ethanol  interactions  in  humans.  Clin 
Pharmacol  Ther  57:559-568,  1995. 

Meyerhoff,  D.J.;  MacKay,  S.;  Sappey- 
Marinier,  D.;  Deicken,  R.;  Calabrese,  G.; 
Dillon,  W.P.;  Weiner,  M.W.;  and  Fein,  G. 
Effects  of  chronic  alcohol  abuse  and  HrV 
infection  on  brain  phosphorus  metabolites. 
Alcohol  Clin  Exp  Res  19:685-692,  1995. 

Mhatre,  M.,  and  Ticku,  M.K.  Chronic 
ethanol  treatment  selectively  increases  the 
binding  of  inverse  agonists  for  benzodi- 
azepine binding  sites  in  cultured  spinal 
cord  neurons.  J  Pharmacol  Exp  Ther 
251:164-168,  1989. 

Mhatre,  M.C.,  and  Ticku,  M.K.  Alcohol: 
Effects  on  GABAA  receptor  function  and 
gene  expression.  Alcohol  Alcohol  2:331- 
335,  1993. 

Michaelis,  E.K.;  Freed,  W.J.;  Galton,  N.; 
Foye,  J.;  Michaelis,  M.L.;  Phillips,  I.;  and 
Kleinman,  J.E.  Glutamate  receptor 
changes  in  brain  synaptic  membranes 
from  human  alcoholics.  Neurochem  Res 
15:1055-1063,  1990. 

Michaelis,  E.K.;  Chen,  X.;  Joseph,  D.B.; 
Hurlbert,  M.;  Kumar,  K.N.;  and 
Michaelis,  M.L.  Ethanol-induced  inhibi- 
tion of  [3H]thienylcyclohexylpiperidine 
(TCP)  binding  to  NMDA  receptors  in 
brain  synaptic  membranes  and  to  a  puri- 
fied protein  complex.  J  Neurochem 
67:201-211,  1996. 


Montpied,  P.,  and  Paul,  S.M.  Studies  on 
the  expression  of  GABAA  receptor  alpha 
subunit  mRNAs.  Adv  Biochem 
Psychopharmacol  47 :301-309,  1992. 

Montpied,  P.;  Morrow,  A.L.;  Karanian, 
J.W.;  Ginns,  E.I.;  Martin,  B.M.;  and  Paul, 
S.M.  Prolonged  ethanol  inhalation  decreases 
gamma- aminobutyric  acidA  receptor 
alpha  subunit  mRNAs  in  the  rat  cerebral 
cortex.  Mol  Pharmacol  39:157-163,  1991. 

Moraes  Ferreira,  V.M.,  and  Morato,  G.S. 
D-cycloserine  blocks  the  effects  of  ethanol 
and  FtA-966  in  rats  tested  in  the  elevated 
plus-maze.  Alcohol  Clin  Exp  Res  21 :1638- 
1642, 1997. 

Moss,  H.B.;  Yao,  J.K.;  Burns,  M.; 
Maddock,  J.;  and  Tarter,  RE.  Plasma 
GABA-like  activity  in  response  to  ethanol 
challenge  in  men  at  high  risk  for  alco- 
holism. Biol  Psychiatry  27:617-625,  1990. 

Muramatsu,  T.;  Higuchi,  S.;  Murayama, 
M.;  Matsushita,  S.;  and  Hayashida,  M. 
Association  between  alcoholism  and  the 
dopamine  D4  receptor  gene.  J  Med  Genet 
33(2):113-115,  1996. 

Myers,  R.D.;  Borg,  S.;  and  Mossberg,  R 
Antagonism  by  naltrexone  of  voluntary 
alcohol  selection  in  the  chronically  drinking 
macaque  monkey.  Alcohol 3:383-388, 1986. 

Naranjo,  C.A.,  and  Bremner,  K.E. 
Clinical  pharmacology  of  serotonin- 
altering  medications  for  decreasing  alco- 
hol consumption.  Alcohol  Alcohol  Suppl 
2:221-229,  1993. 

Naranjo,  C.A.;  Poulos,  C.X.;  Bremner, 
K.E.;  and  Lanctot,  K.L.  Fluoxetine  atten- 
uates alcohol  intake  and  desire  to  drink. 
Int  Clin  Psychopharmacol  9:163-72,  1994. 


389 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Naranjo,  C.A.;  Poulos,  C.X.;  Lanctot, 
K.L.;  Bremner,  K.E.;  Kwok,  M.;  and 
Umana,  M.  Ritanserin,  a  central  5-HT2 
antagonist,  in  heavy  social  drinkers: 
Desire  to  drink,  alcohol  intake  and  related 
effects.  Addiction  90:893-905,  1995. 

Naranjo,  C.A.;  Dongier,  M.;  and  Bremner, 
K.E.  Long-acting  injectable  bromocrip- 
tine does  not  reduce  relapse  in  alcoholics. 
Addiction  92(8):969-978,  1997. 

Nie,  Z.;  Madamba,  S.G.;  and  Siggins, 
G.R.  Ethanol  inhibits  glutamatergic  neu- 
rotransmission in  nucleus  accumbens 
neurons  by  multiple  mechanisms.  / 
Pharmacol  Exp  T&«-  271:1 566-1 573, 1994. 

Nielsen,  D.A.;  Goldman,  D.;  Virkkunen, 
M.;  Tokola,  R;  Rawlings,  R;  and  Linnoila, 
M.  Suicidality  and  5-hydroxyindoleacetic 
acid  concentration  associated  with  a  tryp- 
tophan hydroxylase  polymorphism.  Arch 
Gen  Psychiatry  51:34-38,  1994. 

Nielsen,  D.A.;  Goldman,  D.;  Virkkunen, 
M.;  Tokola,  R;  Rawlings,  R;  and  Linnoila, 
M.  TPH  replication  study:  Not.  [letter: 
comment].  Arch  Gen  Psychiatry  53:964- 
965,  1996. 

Nutt,  D.;  Adinoff,  B.;  and  Linnoila,  M. 
Benzodiazepines  in  the  treatment  of 
alcoholism.  Recent  Dev  Alcohol  7:283- 
313,  1989. 

O'Malley,  S.S.;  Jaffe,  A.J.;  Chang,  G.; 
Schottenfeld,  R.S.;  Meyer,  RE.;  and 
Rounsaville,  B.  Naltrexone  and  coping 
skills  therapy  for  alcohol  dependence.  A 
controlled  study.  Arch  Gen  Psychiatry 
49:881-887,  1992. 

O'Malley,  S.S.;  Jaffe,  A.J.;  Rode,  S.;  and 
Rounsaville,  B.J.  Experience  of  a  "slip" 
among  alcoholics  treated  with  naltrexone 


or  placebo.  Am  J  Psychiatry  153:281- 
283, 1996. 

Paille,  F.M.;  Guelfi,  J.D.;  Perkins,  A.C.; 
Royer,  R.J.;  Steru,  L.;  and  Parot,  P. 
Double -blind  randomized  multicentre 
trial  of  acamprosate  in  maintaining  absti- 
nence from  alcohol.  Alcohol  Alcohol 
30:239-247,  1995. 

Parsian,  A.,  and  Cloninger,  C.R  Human 
GABAA  receptor  alpha  1  and  alpha  3 
subunits  genes  and  alcoholism.  Alcohol 
Clin  Exp  Res  21:430-433,  1997. 

Parsian,  A.;  Todd,  R.D.;  Devor,  E.J.; 
O'Malley,  K.L.;  Suarez,  B.K.;  Reich,  T.; 
and  Cloninger,  C.R.  Alcoholism  and 
alleles  of  the  human  D2  dopamine  recep- 
tor locus.  Studies  of  association  and  link- 
age. Arch  Gen  Psychiatry  48:655-663, 1991. 

Parsian,  A.;  Chakraverty,  S.;  Fisher,  L.; 
and  Cloninger,  C.R.  No  association  be- 
tween polymorphisms  in  the  human 
dopamine  D3  and  D4  receptors  genes 
and  alcoholism.  Am  J  Med  Genet 
74(3):281-285,  1997. 

Pauwels,  P. J.;  Palmier,  C;  Wurch,  T.;  and 
Colpaert,  F.C.  Pharmacology  of  cloned 
human  5-HT1D  receptor- mediated  func- 
tional responses  in  stably  transfected  rat 
C6- glial  cell  lines:  Further  evidence  differ- 
entiating human  5-HT1D  and  5-HT1B 
receptors.  Naunyn  Schmiedebergs  Arch 
Pharmacol  353:144-156,  1996. 

Pelc,  I.;  Verbanck,  P.;  Le  Bon,  O.; 
Gavrilovic,  M.;  Lion,  K.;  and  Lehert,  P. 
Efficacy  and  safety  of  acamprosate  in  the 
treatment  of  detoxified  alcohol-depen- 
dent patients.  A  90-day  placebo-con- 
trolled dose-finding  study.  Br  J  Psychiatry 
171:73-77,  1997. 


390 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Penick,  E.C.;  Powell,  B.J.;  Campbell,  J.; 
Liskow,  B.I.;  Nickel,  E.J.;  Dale,  T.M.; 
Thomas,  H.M.;  Laster,  L.J.;  and  Noble, 
E.  Pharmacological  treatment  for  antiso- 
cial personality  disorder  alcoholics:  A 
preliminary  study.  Alcohol  Clin  Exp  Res 
20:477^84,  1996. 

Peoples,  R.W.,  and  Weight,  F.F.  Cutoff 
inpotency  implicates  alcohol  inhibition  of 
N-methyl-D-aspartate  receptors  in  alco- 
hol intoxication.  Proc  Natl  Acad  Set  USA 
92:2825-2829,  1995. 

Perez-Reyes,  M.;  White,  W.R.;  McDonald, 
S.A.;  and  Hicks,  RE.  Interaction  between 
ethanol  and  dextroamphetamine:  Effects 
on  psychomotor  performance.  Alcohol 
Clin  Exp  Res  16:75-81,  1992. 

Petrakis,  I.L.;  Webb,  E.;  Cooney,  N.; 
Nammanworth,  S.;  Randall,  P.;  Charney, 
D.S.;  and  Krystal,  J.H.  Tryptophan  deple- 
tion blunts  cue-induced  craving  for 
ethanol  in  sober  alcoholics.  Alcohol  Clin 
Exp  Res  19:38A(Abstract  210),  1995. 

Petrakis,  I.L.;  Heninger,  G.R;  Trevisan, 
L.;  D'Souza;  D.C.;  and  Krystal,  J.H.  CSF 
measures  of  monoamine  metabolites  and 
neural-immune  activation  in  alcohol 
dependent  subjects  compared  to  healthy 
volunteers.  Alcohol  Clin  Exp  Res 
21(suppl):77A  (Abstract  441),  1997. 

Pettinati,  H.M.  Use  of  serotonin  selective 
pharmacotherapy  in  the  treatment  of 
alcohol  dependence.  Alcoholism  Clin  Exp 
ito20:23A-29A,  1996. 

Petty,  F.;  Kramer,  G.;  and  Feldman,  M.  Is 
plasma  GABA  of  peripheral  origin?  Biol 
Psychiatry  22:725-732,  1987. 

Petty,  F.;  Fulton,  M.;  Moeller,  F.G.; 
Kramer,  G.;  Wilson,  L.;  Fraser,  K.;  and 


Isbell,  P.  Plasma  gamma-aminobutyric 
acid  (GABA)  is  low  in  alcoholics. 
Psychopharmacol  Bull  29:277-281,  1993. 

Petty,  F.;  Kramer,  G.L.;  Davis,  L.L.; 
Fulton,  M.;  and  Adinoff,  B.  Plasma 
gamma-aminobutyric  acid  (GABA)  pre- 
dicts outcome  in  patients  with  alcohol 
dependence.  Prog  Neuropsychopharmacol 
Biol  Psychiatry  21:809-816,  1997. 

Pihl,  R.O.;  Young,  S.N.;  Ervin,  F.R;  and 
Plotnick,  S.  Influence  of  tryptophan  avail- 
ability on  selection  of  alcohol  and  water 
by  men.  J  Stud  Alcohol  48:260-264,  1987. 

Pihl,  R.O.;  Young,  S.N.;  Harden,  P.; 
Plotnick,  S.;  Chamberlain,  B.;  and  Ervin, 
F.R.  Acute  effect  of  altered  tryptophan 
levels  and  alcohol  on  aggression  in  normal 
human  males.  Psychopharmacology  (Berl) 
119:353-360,  1995. 

Potokar,  J.;  Coupland,  N.;  Glue,  P.; 
Groves,  S.;  Malizia,  A.;  Bailey,  J.;  Wilson, 
S.;  and  Nutt,  D.  Flumazenil  in  alcohol 
withdrawal:  A  double-blind  placebo- 
controlled  study.  Alcohol  Alcohol  32:605- 
611,1997. 

Putzke,  J.;  Spanagel,  R;  Tolle,  T.R.;  and 
Zieglgansberger,  W.  The  anti-craving 
drug  acamprosate  reduces  c-fos  expression 
in  rats  undergoing  ethanol  withdrawal. 
Eur  J  Pharmacol  317:39-48,  1996. 

Rapkin,  A.J.;  Morgan,  M.;  Goldman,  L.; 
Brann,  D.W.;  Simone,  D.;  and  Mahesh, 
V.B.  Progesterone  metabolite  allopregnano- 
lone  in  women  with  premenstrual  syndrome. 
Obstet  Gynecol  90:709-714.  1997. 

Rausch,  J.L.;  Monteiro,  M.G.;  and 
Schuckit,  MA.  Platelet  serotonin  uptake 
in  men  with  family  histories  of  alcoholism. 
Neuropsychopharmacolo£ry  4:83-86,  1991 . 


391 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Ritchie,  T.;  and  Noble,  E.P.  [3H]naloxone 
binding  in  die  human  brain:  Alcoholism 
and  the  TaqI  A  D2  dopamine  receptor  poly- 
morphism. Brain  Res  718:193-197,  1996. 

Robertson,  D. W.;  Bloomquist,  W.;  Wong, 
D.T.;  and  Cohen,  M.L.  mCPP  but  not 
TFMPP  is  an  antagonist  at  cardiac  5HT3 
receptors.  Life  Sci  50:599-605,  1992. 

Roy,  A.;  Dejong,  J.;  Ferraro,  T.;  Adinoff, 
B.;  Ravitz,  B.;  and  Linnoila,  M.  CSF 
gamma-aminobutyric  acid  in  alcoholics 
and  control  subjects.  Am  J  Psychiatry 
147:1294-1296,  1990. 

Roy,  A.;  Dejong,  J.;  Lamparski,  D.; 
George,  T.;  and  Linnoila,  M.  Depression 
among  alcoholics.  Relationship  to  clinical 
and  cerebrospinal  fluid  variables.  Arch 
Gen  Psychiatry  48:428-432,  1991. 

Salomon,  R.M.;  Mazure,  CM.;  Delgado, 
P.L.;  Mendia,  P.;  and  Charney,  D.S. 
Serotonin  function  in  aggression:  The 
effect  of  acute  plasma  tryptophan  deple- 
tion in  aggressive  patients.  Biol  Psychiatry 
35:570-572,  1994. 

Sander,  T.;  Harms,  H.;  Podschus,  J.; 
Finckh,  U.;  Nickel,  B.;  Rolfs,  A.; 
Rommelspacher,  PL;  and  Schmidt,  L.G. 
Dopamine  Dl,  D2  and  D3  receptor 
genes  in  alcohol  dependence.  Psychiatr 
Genet  5:171-176,  1995. 

Sander,  T.;  Harms,  H.;  Dufeu,  P.;  Kuhn, 
S.;  Rommelspacher,  H.;  and  Schmidt,  L.G. 
Dopamine  D4  receptor  exon  III  alleles  and 
variation  of  novelty  seeking  in  alcoholics. 
Am  J  Med  Genet  74:483-487,  1997. 

Sass,  H.;  Soyka,  M.;  Mann,  K.;  and 
Zieglgansberger,  W.  Relapse  prevention 
by  acamprosate.  Results  from  a  placebo- 
controlled  study  on  alcohol  dependence 


[published  erratum  appears  in  Arch  Gen 
Psychiatry  53(12):1097,  1996].  Arch  Gen 
Psychiatry  53:673-680,  1996. 

Satel,  S.L.;  Krystal,  J.H.;  Delgado,  P.L.; 
Kosten,  T.R.;  and  Charney,  D.S. 
Tryptophan  depletion  and  attenuation  of 
cue-induced  craving  for  cocaine.  Am  J 
Psychiatry  152:778-783,  1995. 

Saxon,  L.;  Hjemdahl,  P.;  Hiltunen,  A.J.; 
and  Borg,  S.  Effects  of  flumazenil  in  the 
treatment  of  benzodiazepine  withdrawal — 
a  double-blind  pilot  study.  Psychopharma- 
cology  (Berl)  131:153-160,  1997. 

Schuckit,  M.A.  Low  level  of  response  to 
alcohol  as  a  predictor  of  future  alcoholism. 
Am  J  Psychiatry  151:184-189,  1994. 

Schuckit,  M.A.,  and  Smith,  T.L.  An  8- 
year  follow-up  of  450  sons  of  alcoholic 
and  control  subjects.  Arch  Gen  Psychiatry 
53:202-10,  1996. 

Schuckit,  M.A.,  and  Smith,  T.L.  Assessing 
the  risk  for  alcoholism  among  sons  of 
alcoholics.  /  Stud  Alcohol  58 :  141-145, 1997. 

Schuckit,  M.A.;  Tsuang,  J.W.;  Anthenelli, 
R.M.;  Tipp,  J.E.;  and  Nurnberger,  J. I.,  Jr. 
Alcohol  challenges  in  young  men  from 
alcoholic  pedigrees  and  control  families:  A 
report  from  the  COGA  project.  J  Stud 
Alcohol  57:368-377,  1996. 

Schuckit,  M.A.;  Tipp,  J.E.;  Smith,  T.L.; 
Wiesbeck,  G.A.;  and  Kalmijn,  J.  The 
relationship  between  self-rating  of  the 
effects  of  alcohol  and  alcohol  challenge 
results  in  ninety-eight  young  men.  /  Stud 
Alcohol  58:397-404,  1997. 

Seibyl,  J.P.;  Krystal,  J.H.;  Price,  L.H.; 
Woods,  S.W.;  D'Amico,  C;  Heninger, 
G.R.;  and  Charney,  D.S.  Effects  of  ri- 


392 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


tanserin  on  the  behavioral,  neuroendocrine, 
and  cardiovascular  responses  to  meta- 
chlorophenylpiperazine  in  healthy  human 
subjects.  Psychiatry  Res  38:227-236,  1991. 

Sellers,  E.M.;  Toneatto,  T.;  Romach, 
M.K.;  Somer,  G.R.;  Sobell,  L.C.;  and 
Sobell,  M.B.  Clinical  efficacy  of  the  5- 
HT3  antagonist  ondansetron  in  alcohol 
abuse  and  dependence .  Alcohol  Clin  Exp 
Res  18:879-885,  1994. 

Sherif,  F.;  Wahlstrom,  G.;  and  Oreland, 
L.  Increase  in  brain  GABA-transaminase 
activity  after  chronic  ethanol  treatment  in 
rats.  J  Neural  Transm  Gen  Section  98:69- 
79,  1994. 

Sherif,  F.M.;  Tawati,  A.M.;  Ahmed,  S.S.; 
and  Sharif,  S.I.  Basic  aspects  of  GABA- 
transmission  in  alcoholism,  with  particular 
reference  to  GABA-transaminase.  Eur 
Neuropsychopharmacol  7:1-7 ',  1997. 

Shi,  W.X.;  Nathaniel,  P.;  and  Bunney, 
B.S.  Ri tanserin,  a  5-HT2A/2C  antago- 
nist, reverses  direct  dopamine  agonist- 
induced  inhibition  of  midbrain  dopamine 
neurons.  J  Pharmacol  Exp  Ther  274:735- 
740, 1995. 

Spanagel,  R;  Holter,  S.M.;  Allingham,  K.; 
Landgraf,  R;  and  Zieglgansberger,  W. 
Acamprosate  and  alcohol:  I.  Effects  on 
alcohol  intake  following  alcohol  depriva- 
tion in  the  rat.  Eur  J  Pharmacol  305:39- 
44,  19960. 

Spanagel,  R;  Zieglgansberger,  W.;  and 
Hundt,  W.  Acamprosate  and  alcohol:  III. 
Effects  on  alcohol  discrimination  in  the 
rat.  Eur  J  Pharmacol  305:51-56,  1996&. 

Stuppaeck,  C.H.;  Deisenhammer,  E.A.; 
Kurz,  M.;  Whitworth,  A.B.;  and 
Hinterhuber,  H.  The  irreversible  gamma- 


aminobutyrate  transaminase  inhibitor 
vigabatrin  in  the  treatment  of  the  alcohol 
withdrawal  syndrome  [letter].  Alcohol 
Alcohon\:l09-lll,l996. 

Sullivan,  P.F.;  Fifield,  W.J.;  Kennedy,  M.A.; 
Mulder,  R.T.;  Sellman,  J.D.;  and  Joyce, 
P.R  No  association  between  novelty 
seeking  and  the  type  4  dopamine  receptor 
gene  (DRD4)  in  two  New  Zealand  sam- 
ples. Am  J  Psychiatry  155:98-101,  1998. 

Suzdak,  P.D.;  Paul,  S.M.;  and  Crawley,  J.N. 
Effects  of  Rol5-4513  and  other  benzodi- 
azepine receptor  inverse  agonists  on  alcohol- 
induced  intoxication  in  the  rat.  J  Pharmacol 
Exp  Ther  245:880-886,  1988#. 

Suzdak,  P.D.;  Schwartz,  R.D.;  Skolnick, 
P.;  and  Paul,  S.M.  Alcohols  stimulate 
gamma-  aminobutyric  acid  receptor- medi- 
ated chloride  uptake  in  brain  vesicles: 
Correlation  with  intoxication  potency. 
Brain  Res  444:340-345,  1988&. 

Swift,  R.M.;  Whelihan,  W.;  Kuznetsov, 
O.;  Buongiorno,  G.;  and  Hsuing,  H. 
Naltrexone-induced  alterations  in  human 
ethanol  intoxication.  Am  J  Psychiatry 
151:1463-1467,  1994. 

Swift,  R.M.;  Davidson,  D.;  Whelihan,  W.; 
and  Kuznetsov,  O.  Ondansetron  alters 
human  alcohol  intoxication.  Biol  Psychiatry 
40:514-521,  1996. 

Sytinsky,  LA.;  Guzikov,  B.M.;  Gomanko, 
M.V.;  Eremin,  V.P.;  and  Konovalova, 
N.N.  The  gamma- aminobutyric  acid 
(GAB A)  system  in  brain  during  acute  and 
chronic  ethanol  intoxication.  /  Neurochem 
25:43^8,  1975. 

Tabakoff,  B.;  Hoffman,  P.L.;  Valverius, 
P.;  Borg,  S.;  Lee,  J.M.;  Jaffe,  R; 
U'Prichard,  D.;  and  DeLeon-Jones,  F. 


393 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Characteristics  of  receptors  and  enzymes 
in  brains  of  human  alcoholics.  Alcohol 
2:419-423,  1985. 

Tamborska,  E.,  and  Marangos,  P.J.  Brain 
benzodiazepine  binding  sites  in  ethanol 
dependent  and  withdrawal  states.  Life  Sci 
38:  465-472,  1986. 

Tanda,  G.;  Pontieri,  F.E.;  and  Di  Chiara,  G. 
Cannabinoid  and  heroin  activation  of  meso- 
limbic  dopamine  transmission  by  a  common 
mul  opioid  receptor  mechanism  [see  com- 
ments]. Science  276:2048-2050,  1997. 

Ticku,  M.K.,  and  Burch,  T.  Alterations  in 
gamma-aminobutyric  acid  receptor  sensitiv- 
ity following  acute  and  chronic  ethanol 
treatments.  J  Neurochem  34:417-423,  1980. 

Ticku,  M.K.,  and  Mhatre,  M.C.  Alcohol 
modulation  of  GABAA  receptor  function 
and  gene  expression.  Clin  Neuropharmacol 
15:299A-300A,  1992. 

Ticku,  M.K.;  Burch,  T.P.;  and  Davis,  W.C. 
The  interactions  of  ethanol  with  the  ben- 
zodiazepine -  GAB  A  receptor-ionophore 
complex.  Pharmacol  Biocbem  Behav  18:15- 
18,  1983. 

Ticku,  M.K.;  Mhatre,  M.;  and  Mehta,  A.K. 
Modulation  of  GABAergic  transmission 
by  ethanol.  Adv  Biochem  Psychopharmacol 
47:255-268,  1992. 

Tiihonen,  J.;  Kuikka,  J.T.;  Bergstrom, 
K.A.;  Karhu,  J.;  Viinamaki,  H.;  Lehtonen, 
J.;  Hallikainen,  T.;  Yang,  J.;  and  Hakola, 
P.  Single-photon  emission  tomography 
imaging  of  monoamine  transporters  in 
impulsive  violent  behaviour.  EurJNucl 
Med  24:1253-1260,  1997. 

To,  Z.P.;  Bonhaus,  D.W.;  Eglen,  R.M.; 
and  Jakeman,  L.B.  Characterization  and 
distribution  of  putative  5-HT7  receptors 


in  guinea-pig  brain.  Br  J  Pharmacol 
115:107-116,  1995. 

Tollefson,  G.D.;  Montague-Clouse,  J.; 
and  Tollefson,  S.L.  Treatment  of  comor- 
bid  generalized  anxiety  in  a  recently 
detoxified  alcoholic  population  with  a 
selective  serotonergic  drug  (buspirone)./ 
Clin  Psychopharmacol  12:19-26,  1992. 

Tomkins,  D.M.;  Higgins,  G.A.;  and  Sellers, 
E.M.  Low  doses  of  the  5-HT1A  agonist 
8-hydroxy-2-(di-n-propylamino)-tetralin 
(8-OH  DPAT)  increase  ethanol  intake. 
Psychopharmacology  115:1 73-1 79,  1994. 

Tomkins,  D.M.;  Le,  A.D.;  and  Sellers, 
E.M.  Effect  of  the  5-HT3  antagonist 
ondansetron  on  voluntary  ethanol  intake 
in  rats  and  mice  maintained  on  a  limited 
access  procedure.  Psychopharmacology 
117:479-485,1995. 

Toropainen,  M.;  Nakki,  R.;  Honkanen, 
A.;  Rosenberg,  P.H.;  Laurie,  D.J.;  Pelto- 
Huikko,  M.;  Koistinaho,  J.;  Eriksson, 
C.J.;  and  Korpi,  E.R.  Behavioral  sensitiv- 
ity and  ethanol  potentiation  of  the  N- 
methyl-D-aspartate  receptor  antagonist 
MK-801  in  a  rat  line  selected  for  high 
ethanol  sensitivity.  Alcohol  Clin  Exp  Res 
21:666-671,  1997. 

Tran,  V.T.;  Snyder,  S.H.;  Major,  L.F.; 
and  Hawley,  RJ.  GAB  A  receptors  are 
increased  in  brains  of  alcoholics.  Ann 
Neurol  9 :289-292,  1981. 

Trevisan,  L.;  Nammanworth,  S.;  Petrakis, 
I.;  Randall,  P.;  Charney,  D.S.;  and  Krystal, 
J.H.  Cycloserine  modulation  of  human 
ethanol  intoxication:  Implications  for 
NMDA  receptor  contributions  to  human 
ethanol  intoxication.  Alcohol  Clin  Exp  Res 
19:4A(  Abstract  4),  1995. 


394 


Clinical  Neuroscience  Studies  of  Behaviors  Associated  With  Alcoholism 


Tsai,  G.;  Gastfriend,  D.R.;  and  Coyle, 
J.T.  The  glutamatergic  basis  of  human 
alcoholism  [see  comments].  Am  J 
Psychiatry  152:332-340,  1995. 

Vescovi,  P.P.,  and  Coiro,  V.  Persistence 
of  defective  serotonergic  and  GABAergic 
controls  of  growth  hormone  secretion  in 
long-term  abstinent  alcoholics.  Alcohol 
Alcohol  32:85-90,  1997. 

Virkkunen,  M.,  and  Linnoila,  M. 
Serotonin  in  early-onset  alcoholism. 
Recent  Dev  Alcohol  13:173-189,  1997. 

Virkkunen,  M.;  Goldman,  D.;  Nielsen, 
D.A.;  and  Linnoila,  M.  Low  brain  sero- 
tonin turnover  rate  (low  CSF  5-HIAA) 
and  impulsive  violence.  J  Psychiatry 
Neurosci  20:271-275,  1995. 

Volkow,  N.D.;  Hitzemann,  R.;  Wolf, 
A.P.;  Logan,  J.;  Fowler,  J.S.;  Christman, 
D.;  Dewey,  S.L.;  Schlyer,  D.;  Burr,  G.; 
Vitkun,  S.;  et  al.  Acute  effects  of  ethanol 
on  regional  brain  glucose  metabolism  and 
transport.  Psychiatry  Res  35:39-48,  1990. 

Volkow,  N.D.;  Wang,  G.J.;  Hitzemann, 
R.;  Fowler,  J.S.;  Wolf,  A.P.;  Pappas,  N.; 
Biegon,  A.;  and  Dewey,  S.L.  Decreased 
cerebral  response  to  inhibitory  neuro- 
transmission in  alcoholics.  Am  J  Psychiatry 
150:417-422,  1993. 

Volkow,  N.D.;  Wang,  G.J.;  Begleiter,  PL; 
Hitzemann,  R;  Pappas,  N.;  Burr,  G.; 
Pascani,  K.;  Wong,  C.;  Fowler,  J.S.;  and 
Wolf,  A. P.  Regional  brain  metabolic 
response  to  lorazepam  in  subjects  at  risk 
for  alcoholism.  Alcohol  Clin  Exp  Res 
19:510-516,  1995. 

Volkow,  N.D.;  Wang,  G.J.;  Fowler,  J.S.; 
Logan,  J.;  Hitzemann,  R;  Ding,  Y.S.;  Pappas, 
N.;  Shea,  C.;  and  Piscani,  K.  Decreases  in 
dopamine  receptors  but  not  in  dopamine 


transporters  in  alcoholics.  Alcohol  Clin 
Exp  Res  20:1594-1598,  1996. 

Volkow,  N.D.;  Wang,  G.J.;  Overall,  J.E.; 
Hitzemann,  R.;  Fowler,  J.S.;  Pappas,  N.; 
Frecska,  E.;  and  Piscani,  K.  Regional  brain 
metabolic  response  to  lorazepam  in  alco- 
holics during  early  and  late  alcohol  detox- 
ification. Alcohol  Clin  Exp  Res  21: 
1278-1284,  1997. 

Volpicelli,  J.R.;  Davis,  M.A.;  and  Olgin, 
J.E.  Naltrexone  blocks  the  post-shock 
increase  of  ethanol  consumption.  Life  Sci 
38:841-847,  1986. 

Volpicelli,  J.R.;  Alterman,  A.I.; 
Hayashida,  M.;  and  O'Brien,  C.P. 
Naltrexone  in  the  treatment  of  alcohol 
dependence  [see  comments].  Arch  Gen 
Psychiatry  49:876-880,  1992. 

Volpicelli,  J.R;  Watson,  N.T.;  King,  A.C.; 
Sherman,  C.E.;  and  O'Brien,  C.P.  Effect 
of  naltrexone  on  alcohol  "high"  in  alcoholics. 
Am  J  Psychiatry  152:613-615,  1995. 

Wala,  E.P.;  Sloan,  J.W.;  and  Jing,  X.  Com- 
parison of  abstinence  syndromes  precipitated 
by  flumazenil  and  PK  11195  in  female 
diazepam-dependent  rats.  Psychopharmacofojjy 
(Berl)  133:214-223,  1997. 

Whitworth,  A.B.;  Fischer,  F.;  Lesch,  O.M.; 
Nimmerrichter,  A.;  Oberbauer,  H.;  Platz, 
T.;  Potgieter,  A.;  Walter,  H.;  and  Fleisch- 
hacker,  W.W.  Comparison  of  acamprosate 
and  placebo  in  long-term  treatment  of 
alcohol  dependence  [see  comments]. 
Lancet  347:1438-1442,  1996. 

Young,  S.N.;  Smith,  S.E.;  Pihl,  R.O.;  and 
Ervin,  F.R.  Tryptophan  depletion  causes  a 
rapid  lowering  of  mood  in  normal  males. 
Psychopharmacology  87:173-177,  1985. 


395 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Young,  S.N.;  Pihl,  R.O.;  and  Ervin,  F.R. 
The  effect  of  altered  tryptophan  levels  on 
mood  and  behavior  in  normal  human 
males.  Clin  Neuropharmacol  11:S207- 
S215,  1988. 

Zacchia,  C;  Pihl,  R.O.;  Young,  S.;  and 
Ervin,  F.R.  The  influence  of  altered  tryp- 
tophan availability  on  ethanol  intoxication 


in  normal  men.  J  Stud  Alcohol  48:535- 
540,  1987. 

Zeise,  M.L.;  Kasparov,  S.;  Capogna,  M.; 
and  Zieglgansberger,  W.  Acamprosate 
(calcium  acetylhomotaurinate)  decreases 
postsynaptic  potentials  in  the  rat  neocor- 
tex: Possible  involvement  of  excitatory 
amino  acid  receptors.  Eur  J  Pharmacol 
231:47-52,  1993. 


396 


Chapter  11 

The  Hypothalamic-Pituitary- Adrenal 
Axis:  Changes  and  Risk  for  Alcoholism 

Gary  Wand,  M.D. 


KEY  WORDS:  hypothalamus-pituitary  axis;  pituitary-adrenal  axis;  AODE  (effects 
ofAOD  [alcohol  or  other  drug]  use,  abuse,  and  dependence);  psychological  AODC 
(causes  of  AOD  use,  abuse,  and  dependence);  physiological  stress;  corticotropin 
RH;  adrenocorticotropic  hormone;  genetic  marker  of  potential  AODD  (AOD  use 
disorder);  Cortisol;  endocrine  system;  AOD  use  susceptibility;  dopamine;  opioids; 
naloxone;  literature  review 


Stress  threatens  homeostasis  and  is 
counteracted  by  a  series  of  physiolog- 
ical and  behavioral  responses  that 
improve  the  chances  for  survival 
(Chrousos  and  Gold  1992).  A  success- 
ful response  to  stress  plays  an  important 
role  in  maintaining  good  health  and 
well-being.  The  unique  characteristics 
of  an  individual's  stress  response  are 
the  product  of  genetic  and  environ- 
mental determinants.  A  hyperactive 
stress  response  as  well  as  an  attenu- 
ated response  to  stress  is  maladaptive. 
Mounting  evidence  suggests  that 
abnormalities  in  the  stress  response 
heighten  an  individual's  vulnerability  to 
certain  endocrine,  metabolic,  psychiatric, 


and  immunological  disorders.  There  is 
also  growing  evidence  that  nonalco- 
holic persons  at  increased  risk  for  the 
future  development  of  alcoholism 
have  abnormalities  in  the  hypothalamic - 
pituitary- adrenal  (HPA)  component 
of  the  stress  response.  Moreover,  the 
performance  of  the  HPA  axis  becomes 
impaired  as  a  result  of  chronic,  heavy 
ethanol  ingestion.  Animal  studies 
have  demonstrated  that  stress  alters 
mesolimbic  dopamine  accumulation, 
resulting  in  aberrant  responses  to 
ethanol  and  certain  other  drugs  of 
abuse.  For  these  reasons  it  is  important 
to  more  fully  understand  the  nature 
of  the  HPA  axis  in  alcohol-dependent 


G.  Wand,  M.D.,  is  professor  of  medicine  and  psychiatry,  Departments  of  Medicine  and  Psychiatry, 
Division  of  Endocrinology,  Ross  863,  The  Johns  Hopkins  University  School  of  Medicine,  720  Rutland 
Ave.,  Baltimore,  MD  21205. 


397 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


persons  and  in  their  nonalcoholic 
family  members. 

PHYSIOLOGY 
OF  THE  HPA  AXIS 

The  response  to  stress  is  generated 
within  two  well-defined  brain  regions, 
the  hypothalamus  and  the  brainstem. 
The  hypothalamic  center  includes  cor- 
ticotropin-releasing  hormone  (CRH) 
and  arginine  vasopressin  (AVP)  neu- 
rons in  the  paraventricular  nucleus. 
The  brainstem  center  includes  CRH 
neurons  of  the  paragigantocellular 
and  parabranchial  nuclei  of  the 
medulla,  as  well  as  the  locus  coeruleus 
and  other  catecholaminergic  neurons 
of  the  medulla  and  pons  (Tsigos  and 
Chrousos  1995).  The  HPA  axis,  the 
efferent  sympathetic/adrenomedullary 
system,  and  components  of  the  para- 
sympathetic system  constitute  the 
peripheral  branches  of  the  stress  system 
(Tsigos  and  Chrousos  1995). 

Following  perception  of  stress  the 
hypothalamus  releases  CRH,  AVP, 
and  other  adrenocorticotropin  (ACTH) 
secretagogues  into  the  pituitary  portal 
circulation,  resulting  in  release  of  ACTH 
from  the  pituitary  gland.  In  turn, 
ACTH  initiates  the  release  of  the  adrenal 
androgens  and  glucocorticoids.  Glu- 
cocorticoids prepare  the  organism  for 
stress  by  modulating  fuel  metabolism 
and  thereby  increasing  glucose  and 
fatty  acid  availability  for  the  central 
nervous  system  (CNS).  Another 
important  function  of  the  HPA  axis  is 
its  bidirectional  interactions  with  the 
immune  system  (Rivier  1993). 
Inflammatory  cytokines  (e.g.,  tumor 
necrosis  factor  alpha,  interleukin-1, 


and  interleukin-6)  and  other  humoral 
mediators  of  inflammation  stimulate 
the  HPA  axis.  In  response,  glucocorti- 
coids inhibit  cytokine  production  and 
virtually  all  components  of  the  immune 
reaction,  thus  preventing  an  over- 
whelming and  potentially  harmful 
immune  response  to  stressors.  Finally, 
glucocorticoids  participate  in  their 
own  production  through  binding  to 
glucocorticoid  receptors  in  the  pitu- 
itary, hypothalamus,  and  hippocam- 
pus, inhibiting  the  synthesis  and 
release  of  CRH  and  ACTH. 

PATHOPHYSIOLOGY 
OF  THE  HPA  AXIS 

A  healthy  HPA  axis  response  to  stress 
is  characterized  by  rapid  onset  and  lim- 
ited duration.  The  time-limited  nature 
of  the  event  ensures  that  the  accom- 
panying behavioral,  metabolic,  and 
immunosuppressive  effects  induced  by 
CRH  and  Cortisol  are  beneficial  and 
have  no  adverse  sequelae  (Tsigos  and 
Chrousos  1995).  Excessive  HPA  axis 
responses  to  stress  are  maladaptive. 
Sustained  hypercortisolism  initiates  a 
spiral-like  cycle  of  HPA  hyperactivity 
(e.g.,  hypercortisolism  begets  hyper- 
cortisolism). This  is  because  pro- 
tracted exposure  to  Cortisol  is  toxic 
and  injures  the  glucocorticoid  nega- 
tive feedback  set-point,  resulting  in 
worsening  hyperactivity  over  time. 

A  well-studied  example  of  inappro- 
priate hypercortisolism  is  melancholic 
depression  characterized  by  dysphoria, 
sustained  activation  of  the  HPA  axis, 
immunosuppression,  and  osteoporosis 
(Gold  et  al.  1988^,  1988b).  Cortisol 
production  is  elevated,  CRH  levels  in 


398 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


cerebrospinal  fluid  are  increased,  and 
there  are  more  CRH  neurons  found  in 
depressed  patients  on  autopsy,  all  con- 
sistent with  overactivity  of  CRH  neu- 
rons (Raadsheer  et  al.  1994).  Other 
disorders  associated  with  sustained 
activation  of  the  HPA  axis  include 
anorexia  nervosa  (Chrousos  and  Gold 
1992),  obsessive -compulsive  disorder 
(Insel  et  al.  1982),  panic  and  anxiety 
disorders  (Gold  et  al.  1988c),  excessive 
exercising  (Luger  et  al.  1987),  visceral 
obesity  (Pasquali  et  al.  1993),  the  pre- 
menstrual tension  syndrome  (Robin  et 
al.  1990),  and  chronic  active  alco- 
holism (Wand  and  Dobs  1991). 
Moreover,  elevated  Cortisol  levels  have 
been  found  in  preschool  children  with 
poor  inhibitory  control  and  in  associa- 
tion with  boredom,  impatience,  irrita- 
tion, fear,  and  anxiety  (Stansbury  and 
Gunnar  1994). 

Why  should  abnormal  states  of  CRH 
and/or  Cortisol  secretion  compromise 
health?  First,  sustained  exposure  to 
elevated  levels  of  glucocorticoids  can 
have  detrimental  effects  on  bone  and 
muscle  mass  as  well  as  on  growth, 
reproductive  status,  and  immune  func- 
tion. There  is  evidence  indicating  that 
hypercortisolism  seen  in  Cushing's 
syndrome  and  depression  decreases 
immune  function  and  bone  mass 
(Chrousos  and  Gold  1992).  Neuro- 
toxicity is  another  important  conse- 
quence of  increased  glucocorticoid 
production.  Elevated  levels  of  gluco- 
corticoids impair  CNS  energy  metab- 
olism, making  the  brain  vulnerable  to 
injury.  Hippocampal  damage  is 
observed  in  conditions  linked  with 
hypercortisolism,  an  event  that  may 
alter  learning,  memory,  and  regulation 


of  the  HPA  axis  (Sapolsky  1996).  In 
fact,  studies  have  shown  a  negative  cor- 
relation between  hippocampal  volume 
and  ( 1 )  plasma  Cortisol  levels  in  Cush- 
ing's syndrome,  (2)  duration  of  depres- 
sion in  patients  with  affective  illness, 
and  (3)  months  of  combat  exposure 
in  patients  with  posttraumatic  stress 
disorder  (Sapolsky  1996).  Since  CRH 
modulates  behavioral,  neuroendocrino- 
logical, and  immunological  responses, 
altered  production  of  CRH  could  also 
explain  certain  manifestations  in  the 
syndromes  noted  above. 

Pathological  consequences  resulting 
from  altered  HPA  axis  dynamics  are 
not  limited  to  states  of  hyperfunction. 
An  attenuated  HPA  axis  response  to 
stress  can  also  be  problematic.  For 
example,  patients  with  atypical,  seasonal 
depression  and  chronic  fatigue  syn- 
drome have  decreased  activity  of  the 
HPA  axis  (Chrousos  and  Gold  1992). 
Patients  diagnosed  with  fibromyalgia 
and  fatigue  have  decreased  urinary  free 
Cortisol  values  (Griep  et  al.  1993).  Ani- 
mal models  have  shown  that  a  weak 
HPA  axis  response  to  stress  results  in 
excessive  cytokine  activation  and  a 
predisposition  for  certain  autoimmune 
disorders  (Sternberg  et  al.  1992). 

ETHANOL 

AND  THE  HPA  AXIS 

Ethanol  has  several  contrasting  effects 
on  the  HPA  axis.  In  social  drinkers, 
acute  ethanol  ingestion  activates  the 
HPA  axis,  especially  when  intoxication 
is  accompanied  by  gastrointestinal 
symptoms.  Chronic  alcohol  abuse  is 
associated  with  activation  of  the  HPA 
axis,  most  profoundly  observed  during 


399 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


periods  of  acute  withdrawal.  However, 
during  early  and  even  sustained  absti- 
nence, the  HPA  axis  is  dampened.  In 
the  alcohol-dependent  person,  cyclical 
episodes  of  intoxication  and  withdrawal 
provoke  cycles  of  hyper-  and  hypoac- 
tivity,  contributing  to  tissue  injury, 
craving,  and  relapse. 

Acute  Effects  of  Ethanol 
on  the  HPA  Axis 
in  Social  Drinkers 

In  1966,  Mendelson  and  Stein  demon- 
strated that  ingestion  of  ethanol 
increased  plasma  Cortisol  concentration. 
Subsequently,  investigators  more  fully 
characterized  the  biological  mecha- 
nisms of  ethanol-induced  HPA  axis 
activation  in  rodents  (Rivier  et  al. 
1984;  Thiagarajan  et  al.  1989).  In  the 
rat,  ethanol  activates  the  HPA  axis  by 
inducing  release  of  CRH  from  the 
hypothalamus  (Rivier  et  al.  1984). 
Although  never  directly  proved  in 
humans,  ethanol  is  thought  to  induce 
ACTH  and  Cortisol  release  through 
similar  mechanisms. 

Oral  administration  of  ethanol  to 
humans  stimulates  the  HPA  axis  as  it 
does  in  rodents.  However,  the  response 
is  not  clearly  dose  dependent,  and  is 
most  likely  observed  when  blood  levels 
are  greater  than  100  mg%.  More  recent 
studies  have  shown  that  intoxicating 
doses  of  ethanol  per  se  do  not  result  in 
activation  of  the  HPA  axis.  However, 
subjects  experiencing  nausea  or  vomit- 
ing as  a  result  of  the  ethanol  exposure 
are  much  more  likely  to  develop  sig- 
nificant rises  in  ACTH  and  Cortisol 
(Waltman  et  al.  1993;  Aguirre  et  al. 
1995;  Inder  et  al.  1995/;).  Interestingly, 
mild  levels  of  intoxication  (blood 


ethanol  level  75  mg%)  result  in  blunted 
ACTH  and  Cortisol  responses  to 
exogenously  administered  CRH,  sug- 
gesting that  even  acute  ethanol  expo- 
sure impairs  the  ability  of  the  HPA 
axis  to  respond  to  physiological  stressors 
(Waltman  et  al.  1993).  We  have  pro- 
posed that  the  impairment  results 
from  ethanol-induced  inhibition  in 
AVP,  a  secretagogue  that  potentiates 
the  action  of  CRH  on  ACTH  release 
(Waltman  et  al.  1993). 

The  HPA  Axis  in  Alcohol- 
Dependent  Persons 

Ethanol  alters  the  integrity  of  the 
HPA  axis  in  alcohol-dependent  per- 
sons. Paradoxically,  chronic  ethanol 
exposure  can  result  in  episodes  of 
increased  as  well  as  decreased  HPA 
axis  activity.  The  direct  effects  of 
chronic  ethanol  ingestion  on  the 
HPA  axis  have  been  difficult  to  ascer- 
tain because  of  comorbid  states  such 
as  depression,  liver  disease,  malnutri- 
tion, and  other  stressors,  as  well  as 
the  underlying  fluctuations  of  intoxi- 
cation and  withdrawal.  Depression,  in 
particular,  is  common  in  alcoholics 
seeking  treatment  (Weissman  et  al. 
1977).  Although  hypercortisolemia  is 
frequent  in  depression,  the  alterations 
of  the  HPA  axis  seen  in  alcoholics 
can  be  attributed,  at  least  in  part, 
directly  to  the  development  of  alco- 
hol dependence. 

A  dramatic  alteration  in  the  HPA 
axis  observed  in  alcohol-dependent 
subjects  is  pseudo-Cushing's 
syndrome,  which  was  described  by 
Smals  and  colleagues  in  1976  and 
later  also  reported  by  others  (Paton 
1976;  Frajria  and  Angeli  1977;  Rees 


400 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


et  at.  1977).  The  syndrome  is  charac- 
terized by  an  elevated  plasma  or  urine 
Cortisol  level,  which  is  not  suppressed 
following  dexamethasone,  accompa- 
nied by  the  clinical  stigmata  of  hyper- 
cortisolism,  which  resolve  with 
abstinence  from  ethanol.  Although 
chronic  ethanol  exposure  stimulates 
Cortisol  secretion,  few  actively  drink- 
ing alcoholics  develop  frank  pseudo- 
Cushing's  syndrome.  The  mechanism 
of  tolerance  that  prevents  the  devel- 
opment of  symptomatic  hypercorti- 
solemia  in  alcoholics  is  not  known. 
However,  based  on  our  animal  mod- 
els, we  have  proposed  that  ethanol- 
induced  dampening  in  adenylyl 
cyclase  signal  transduction  is  a  factor 
(Wand  1989,  1990;  Wand  and 
Levine  1991). 

Although  most  alcoholics  do  not 
develop  pseudo-Cushing's  syndrome, 
almost  all  have  abnormalities  in  HPA 
axis  function.  Most  studies  have 
examined  the  HPA  axis  during  acute 
withdrawal;  few  studies  have  exam- 
ined the  HPA  axis  during  periods  of 
active  drinking.  A  study  performed  in 
our  laboratory  evaluated  the  HPA 
axis  in  a  group  of  actively  drinking, 
nondepressed,  alcohol-dependent 
men  (Wand  and  Dobs  1991).  Subjects 
were  outpatients  who  continued  their 
normal  drinking  routine  throughout 
the  study.  Alcoholic  subjects  had 
twofold  higher  24-hour  urinary  free 
Cortisol  values  compared  with  matched 
control  subjects.  Paradoxically,  despite 
elevations  in  integrated  Cortisol  mea- 
surements, the  alcoholic  subjects 
demonstrated  blunted  ACTH  and 
Cortisol  responses  to  CRH  as  well  as  a 
blunted  adrenocortical  response  to 


the  ACTH  analog  cosyntropin 
(Cortrosyn).  In  fact,  25  percent  of 
alcoholic  subjects  met  criteria  for 
secondary  adrenal  insufficiency  as 
assessed  by  metyrapone  blockade.  It 
is  clear  that  actively  drinking,  alcohol- 
dependent  persons  have  a  deranged 
HPA  axis. 

The  HPA  Axis  During  Acute 
Withdrawal 

The  abnormalities  in  HPA  axis  function 
during  acute  withdrawal  are  similar  to 
the  abnormalities  found  during  active 
drinking.  There  is  clearly  an  increase 
in  HPA  axis  activation  during  ethanol 
withdrawal  (Willenbring  et  al.  1984). 
Cortisol  concentrations  are  elevated  as 
a  result  of  increases  in  Cortisol  burst 
amplitude  and  Cortisol  mass  secreted 
per  burst  (Iranmanesh  et  al.  1989). 
There  is  also  disruption  of  the  circadian 
pattern  of  Cortisol  release  (Iranmanesh 
et  al.  1989).  Moreover,  there  is  nonsup- 
pressibility  of  the  HPA  axis  to  low 
doses  of  dexamethasone  in  alcoholics 
during  acute  withdrawal  (Swartz  and 
Dunner  1982;  LaFuente  et  al.  1983; 
Del  Porto  et  al.  1985;  Burrov  et  al. 
1986;  Kirkman  and  Nelson  1988; 
Iranmanesh  et  al.  1989).  Longitudinal 
Cortisol  studies  on  alcoholics  admitted 
to  inpatient  services  for  acute  with- 
drawal have  shown  that  the  hypercor- 
tisolism  subsides  over  time.  Cortisol 
levels  normalize  following  7-10  days 
of  abstinence  (Adinoff  et  al.  1991). 

The  HPA  Axis  During 
Abstinence 

As  the  alcohol-dependent  person  pro- 
gresses from  acute  withdrawal  to  early 
abstinence,  another  impairment  in 


401 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


HPA  axis  function  become  manifest, 
namely,  hyporesponsivity.  A  series  of 
HPA  axis  challenge  studies  with 
CPvH,  opioid  receptor  antagonists, 
insulin-induced  hypoglycemia,  and 
ACTH  has  documented  dampened 
hormonal  responsiveness  at  each  level 
of  the  axis  (Costa  et  al.  1996).  Hypo- 
responsiveness  persists  as  far  out  as  6 
months  into  abstinence. 

Summary  of  Abnormalities 
in  the  HPA  Axis 

These  findings  demonstrate  that  alco- 
hol-dependent persons  have  altered 
HPA  axis  dynamics.  The  nature  of 
the  injury  is  twofold.  First,  alcohol- 
dependent  subjects  who  are  actively 
drinking  or  in  acute  withdrawal  gen- 
erate more  Cortisol  per  24-hour  time 
period  relative  to  non-alcohol-depen- 
dent subjects.  Second,  despite  this 
elevation  in  Cortisol  production,  the 
HPA  axis  is  hyporesponsive  to  most 
forms  of  stress.  The  following  sce- 
nario may  explain  this  paradox:  Daily 
cycles  of  intoxication  and  withdrawal 
are  stressful  and  result  in  a  net  eleva- 
tion in  Cortisol  production;  however, 
ethanol-induced  injury  to  the  axis 
results  in  dampening  responsiveness 
to  stressors.  Therefore,  alcohol- 
dependent  subjects  respond  to  stress 
in  a  suboptimal  manner.  We  speculate 
that  non-alcohol-dependent  subjects 
put  under  a  comparable  level  of  stress 
would  demonstrate  much  higher  lev- 
els of  ACTH  and  Cortisol.  Abstinent 
alcohol-dependent  subjects  who  have 
fully  completed  the  withdrawal 
process  only  manifest  HPA  axis 
hyporesponsivity  and  no  longer  have 
elevated  Cortisol  production.  The 


period  of  hyporesponsiveness  can  last 
at  least  6  months  and  perhaps  longer. 

Possible  Consequences 
of  HPA  Axis  Disturbances 
in  Alcohol  Dependence 

As  alcohol-dependent  persons  cycle 
through  episodes  of  intoxication  and 
withdrawal  alternating  with  periods  of 
abstinence,  the  HPA  axis  cycles  through 
periods  of  hyper-  and  hypoactivity. 
This  cyclical  pattern  of  CRH  and  Cor- 
tisol secretion  may  induce  various 
pathological  states  associated  with 
alcoholism.  For  example,  periods  of 
sustained  hypercortisolism  may  exac- 
erbate osteoporosis,  diabetes  mellitus, 
and  hypertension  and  may  alter 
growth,  reproductive  status,  and 
immune  function.  Moreover,  it  has 
been  proposed  that  withdrawal-induced 
hypercortisolism  increases  accumula- 
tion of  excitatory  amino  acids  within 
the  CNS,  resulting  in  neurotoxicity 
(Adinoff  1994).  Several  studies  have 
suggested  that  excess  CRH  and  Corti- 
sol production  triggered  by  with- 
drawal actually  enhance  the  magnitude 
of  withdrawal  symptoms,  including 
seizure  activity  (Menzagh  et  al.  1994; 
Schuikin  et  al.  1994).  Rodent  studies 
demonstrate  that  glucocorticoids  have 
reinforcing  properties  (Deroche  et  al. 
1995)  and  will  increase  mesolimbic 
dopamine,  a  process  that  may  accelerate 
addiction  to  drugs  of  abuse.  Other 
studies  have  shown  that  stress-induced 
increases  in  corticosterone  result  in 
increased  ethanol  intake,  whereas 
medical  or  surgical  adrenalectomy 
reduces  ethanol  consumption  (Samson 
1995).  Furthermore,  CRH  antago- 
nists have  been  shown  to  block  the 


402 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


stress-induced  relapse  to  drug-seeking 
behavior,  indicating  a  significant  role  of 
CRH  in  the  consumption  of  drugs 
of  abuse,  including  alcohol  (Erb  et 
al.  1998). 

Whereas  HPA  axis  hyperactivity 
induced  by  chronic  alcohol  dependence 
may  increase  the  risk  for  alcohol- 
induced  injury  and  for  altered  neuro- 
chemistry  within  the  mesolimbic  system, 
hyporesponsivity  of  the  HPA  axis 
observed  during  abstinence  may  gener- 
ate another  set  of  consequences.  Low 
levels  of  CRH  and  Cortisol  have  been 
correlated  with  atypical  depression, 
impulsivity,  lack  of  behavioral  inhibi- 
tions, and  low  motivation  (Stansbury 
and  Gunnar  1994).  Extrapolating  from 
these  observations,  it  has  been  proposed 
that  suppressed  HPA  axis  responsiveness 
experienced  during  early  abstinence 
may  provoke  dysphoria,  craving,  and 
ultimately  the  high  rate  of  relapse 
associated  with  acute  abstinence  (Sher 
and  Trull  1994).  It  is  provocative  to 
speculate  that  part  of  the  mechanism 
through  which  opioid  antagonists 
reduce  relapse  is  their  ability  to  alter 
responsivity  of  the  HPA  axis. 

There  are  additional  serious  implica- 
tions for  HPA  axis  hyporesponsiveness 
in  alcohol-dependent  persons.  For 
example,  a  subset  of  alcohol -dependent 
patients  have  some  degree  of  adrenal 
insufficiency.  Alcohol-dependent  persons 
are  at  increased  risk  for  severe  infections 
and  have  a  compromised  immune 
system.  Once  ill,  alcohol-dependent 
persons  have  increased  morbidity  and 
mortality  compared  with  non-alcohol - 
dependent  persons.  It  is  plausible  that 
altered  HPA  axis  dynamics  coupled  with 
an  injured  immune  function  in  this 


patient  population  are  the  reasons  for 
increased  susceptibility  to  illness  and 
for  poor  treatment  outcomes.  Since 
ethanol  can  injure  the  HPA  axis  and 
immune  function  (Rivier  1993),  the 
increased  risk  of  infection  may  be  the 
result  of  chronic  alcoholism. 

THE  HPA  AXIS  AND  THE 
RISK  FOR  ALCOHOLISM 

There  is  strong  evidence  that  certain 
genetic  determinants  increase  the  risk 
for  alcoholism  (Kaig  1960;  Goodwin 
et  al.  1973;  Cotton  1979;  Hrubec 
and  Omenn  1981;  Cloninger  1988; 
Merikangas  1990;  Pickens  et  al.  1991; 
Kendler  et  al.  1992).  Such  findings 
have  led  to  investigations  of  the  non- 
alcoholic offspring  of  alcohol-dependent 
persons  for  the  purpose  of  uncovering 
markers  that  identify  those  at  increased 
risk  for  alcohol  dependence.  The  family 
history  of  alcoholism  research  strategy 
compares  nonalcoholic  offspring  from 
families  with  a  high  density  of  alcoholism 
(referred  to  as  family  history  positive 
[FHP])  with  offspring  from  families 
with  no  history  of  alcoholism  (family 
history  negative  [FHN]).  Using  this 
research  design,  ethanol  challenge  stud- 
ies have  compared  FHPs  and  FHNs 
on  their  reaction  to  ethanol  (Begleiter 
et  al.  1984;  Hill  et  al.  1988;  Schuckit 
et  al.  1987a,  1987&;  Finn  et  al.  1990; 
Pfefferbaum  et  al.  1991;  Gianoulakis 
and  De  Waele  1994;  Wand  et  al. 
1994;  Gianoulakis  et  al.  1996).  The  San 
Diego  group  has  reported  the  largest 
series  employing  this  design.  In  this 
longitudinal  investigation,  over  400 
men  were  originally  studied  at  about 
age  20  (Schuckit  and  Smith  1996). 


403 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Subjects  were  evaluated  for  responses 
to  ethanol  based  on  changes  in  sub- 
jective feelings,  hormonal  responses, 
motor  performance,  and  physiological 
measures.  Interestingly,  a  low-level 
Cortisol  response  to  ethanol  character- 
ized those  individuals  who  were  most 
likely  to  develop  alcohol  abuse  or 
dependence  almost  a  decade  later. 

Other  studies  have  also  shown 
differences  in  HPA  axis  dynamics  as  a 
function  of  family  history.  Women 
with  alcoholic  fathers  had  higher 
Cortisol  levels  after  ingesting  ethanol, 
compared  with  women  whose  fathers 
were  not  alcoholics  (Lex  et  al.  1988). 
Gianoulakis  and  colleagues  (1996) 
investigated  plasma  ^-endorphin 
responses  in  a  group  of  nonalcoholic 
men  and  women  with  a  three-genera- 
tion family  history  of  alcoholism. 
Mild  intoxication  significantly 
increased  opioid  release  in  FHP  com- 
pared with  FHN  subjects.  It  has  been 
proposed  that  increased  responsive- 
ness of  the  opioid  system  to  ethanol 
may  create  vulnerability  for  alcohol 
dependence  (Gianoulakis  et  al. 
1996).  The  mechanisms  by  which 
high-  and  low-risk  persons  have  dif- 
ferential plasma  ACTH,  (3-endorphin, 
and  Cortisol  responses  to  ethanol 
challenges  have  not  been  elucidated. 

Ethanol  challenges  have  only  a 
modest  stimulatory  effect  on  the 
HPA  axis.  And,  as  noted  earlier  in 
this  chapter,  nausea  is  often  a  requi- 
site condition  for  ethanol-induced 
activation  of  the  HPA  axis.  However, 
important  information  concerning 
family  history  differences  in  HPA  axis 
dynamics  can  also  be  obtained  by 
stimulating  the  HPA  axis  with 


other  agents.  As  discussed  later  in  this 
chapter,  we  have  demonstrated  that 
HPA  axis  responses  to  CRH  and 
naloxone  differ  as  a  function  of  family 
history  of  alcoholism  (Waltman  et  al. 
1994;  Wand  et  al.  1998). 

Ethanol- Seeking  Behavior 
and  the  HPA  Axis 

In  the  preceding  section  I  reviewed 
evidence  that  the  HPA  axis  may  serve  as 
a  marker  for  individuals  at  increased 
risk  for  alcoholism.  It  appears  that 
Cortisol  responses  to  ethanol  have 
predictive  ability.  It  is  also  possible 
that  the  unique  HPA  hormone  profile 
in  high-risk  individuals  has  a  causal 
relationship  to  vulnerability  for  alco- 
holism. To  understand  why  this  might 
be  true,  it  is  important  to  understand 
the  anatomical  and  functional  relation- 
ships between  the  HPA  axis  and  the 
mesolimbic  reward  center. 

The  mesolimbic  dopaminergic  sys- 
tem mediates  the  reinforcing  effects 
of  many  drugs  of  abuse,  including 
ethanol.  This  system  consists  mainly 
of  A10  dopaminergic  neurons  whose 
cell  bodies  are  located  in  the  ventral 
tegmental  area  and  project  terminals 
to  the  nucleus  accumbens,  frontal  cor- 
tex, amygdala,  and  septal  area  (Koob 
et  al.  1988).  Experimental  evidence 
suggests  that  dopamine  released  within 
the  nucleus  accumbens  is  responsible,  at 
least  in  part,  for  mediating  the  reward- 
ing effects  of  many  drugs  of  abuse. 
For  example,  ethanol  administration 
increases  synaptic  dopamine  accumula- 
tion within  this  important  brain  region 
and  is  thought  to  contribute  to  its 
rewarding  properties.  The  ethanol- 
induced  release  of  nucleus  accumbens 


404 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


dopamine  is  blocked  by  opioid  antag- 
onists, implicating  opioidergic  activity 
as  an  intermediary  between  ethanol 
exposure  and  the  release  of  dopamine 
(Benjamin  et  al.  1992).  Interestingly, 
the  HPA  axis  is  also  modulated  by  the 
same  opioid  systems,  which  regulate 
dopamine-induced  reward  within  the 
mesolimbic  system.  Following  per- 
ception of  stress,  efferent  fibers  from 
several  brain  regions  innervate  hypo- 
thalamic CRH  neurons  in  the  para- 
ventricular nucleus.  This  input 
regulates  the  glucocorticoid  compo- 
nent of  the  stress  response.  The  CRH 
neurons  receive  this  signal  through 
four  major  neurotransmitter  systems 
(Tsigos  and  Chrousos  1995):  stimu- 
latory input  from  serotonergic  and 
adrenergic  neurons  and  inhibitory 
input  from  GABAergic  and  opioider- 
gic neurons.  These  four  neurotrans- 
mitters, which  are  responsible  for 
modulating  the  activity  of  CRH  neu- 
rons, are  also  "candidate"  neurotrans- 
mitters implicated  in  conveying 
biological  vulnerability  for  alcoholism 
(Chick  et  al.  1996). 

Once  CRH  is  released  into  the 
hypophyseal-portal  circulation,  it,  in 
turn,  stimulates  ACTH  secretion  from 
the  pituitary.  ACTH  stimulates  Corti- 
sol release  from  the  adrenal  gland. 
Because  of  the  powerful  inhibitory 
effect  of  opioid  neurons  ((3-endorphin 
and  enkephalin)  on  CRH  activity,  an 
acquired  or  inborn  abnormality  in 
opioid  activity  in  this  region  would 
alter  inhibitory  tone  on  CRH  neu- 
rons. A  subset  of  these  opioidergic 
neurons  responsible  for  providing  the 
inhibitory  effect  on  the  CRH  neurons 
simultaneously  provides  stimulatory 


input  to  presynaptic  dopamine  termi- 
nals within  the  nucleus  accumbens.  If 
the  opioid  system  is  a  biological  sub- 
strate that  modulates  craving,  then  this 
group  of  opioidergic  neurons  is  posi- 
tioned to  play  an  important  role  in 
mesolimbic  contributions  to  craving 
and  ethanol-seeking  behaviors.  There- 
fore, ethanol  ingestion,  through  its 
effects  on  opioidergic  neurons,  can 
activate  mesolimbic  dopamine  genera- 
tion and  the  HPA  axis  simultaneously. 
In  turn,  glucocorticoids,  released  fol- 
lowing ethanol  ingestion,  modulate 
the  activities  of  the  opioidergic,  CRH, 
and  mesolimbic  dopamine  systems; 
chronic  glucocorticoid  exposure 
decreases  CRH  and  opioid  expression, 
whereas  it  enhances  mesolimbic 
dopamine  production.  In  this  manner, 
a  primary  brain  reward  pathway — the 
mesolimbic  dopamine  system — and  a 
primary  stress  pathway — the  HPA 
axis — modulate  one  another. 

Studying  the  release  of  CRH  pro- 
vides a  window  on  CNS  function  and 
can  uncover  differences  in  neuro- 
transmitter systems  as  a  function  of 
both  alcoholism  and  family  history  of 
alcoholism.  It  is  plausible  that  if  a 
neurotransmitter  system  is  altered  in 
persons  at  increased  risk  for  alcoholism, 
then  a  neuroendocrine  system  (e.g., 
HPA  axis)  regulated  by  the  deranged 
neurotransmitter  system  will  also  be 
altered.  Thus,  we  have  hypothesized 
that  differences  in  hypothalamic  opi- 
oid activity  between  high-risk  and 
low-risk  non-alcohol-dependent  sub- 
jects may  be  the  basis  for  differences 
in  the  dynamics  of  the  HPA  axis  in 
the  two  groups  (Wand  et  al.  1998). 
The  next  section  describes  how  we 


405 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


designed  a  research  strategy  to  test 
this  hypothesis. 

Hypothalamic  Opioid 
Activity  and  HPA  Dynamics 
in  Persons  at  Increased  Risk 
for  Alcoholism 

Several  lines  of  evidence  suggest  that 
the  brain  opioid  system  is  part  of  a 
neurocircuitry  involved  in  alcohol  rein- 
forcement and  heavy  alcohol  drinking 
(Gianoulakis  and  De  Waele  1994; 
Chick  et  al.  1996).  Initially,  pharma- 
cological studies  indicated  that  opioid 
receptor  antagonists,  such  as  naloxone 
and  naltrexone,  decreased  ethanol 
self- administration  in  animal  models 
(Altshuler  et  al.  1980;  Marfaing-Jallat 
et  al.  1983;  Samson  and  Doyle  1985; 
Froehlich  et  al.  1990;  Weiss  et  al. 
1990;  Hubbell  et  al.  1991,  1996).  Sub- 
sequent clinical  trials  demonstrated 
that  the  opioid  antagonist  naltrexone 
reduced  alcohol  drinking,  alcohol 
craving,  and  relapse  rates  in  detoxified, 
outpatient  alcoholics  (O'Malley  et  al. 
1992;  Volpicelli  et  al.  1992).  In  fact, 
naltrexone  was  approved  by  the  Food 
and  Drug  Administration  as  a  pharma- 
cotherapeutic  agent  for  the  treatment 
of  alcohol  dependence.  Several  studies 
have  shown  that  a  genetic  predisposi- 
tion toward  alcohol  drinking  is  associ- 
ated with  increased  responsiveness  of 
the  opioid  system  to  ethanol  (De 
Waele  et  al.  1992;  De  Waele  and 
Gianoulakis  1994;  Gianoulakis  et  al. 
1996),  and  several  "opioid  hypotheses" 
have  been  proposed  to  explain  the 
role  of  the  endogenous  opioid  system 
in  vulnerability  to  and  maintenance  of 
alcoholism  (Reid  and  Hunter  1984; 
Reid  1990;  Reid  et  al.  1991;  Gianoulakis 


and  De  Waele  1994).  To  determine 
the  validity  of  an  opioid  model  for 
alcoholism,  it  would  be  necessary  to 
generate  accurate  measurements  of 
endogenous  opioid  tone  in  human 
subjects.  One  technique  that  can  mea- 
sure innate  differences  in  endogenous 
opioid  activity  is  the  induction  of  opi- 
oid receptor  blockade  with  an  opioid 
receptor  antagonist. 

Naloxone,  a  nonselective  opioid 
receptor  antagonist,  induces  a  rise  in 
ACTH  and  Cortisol  by  blocking  the 
opioid  component  of  the  inhibitory 
activity  directed  at  the  CRH  neurons. 
As  a  result  of  opioidergic  modulation 
of  CRH  neurons,  the  naloxone  chal- 
lenge test  can  identify  inborn  and 
acquired  alterations  in  endogenous 
opioid  activity  (Torpy  et  al.  1993;  Del 
Campo  et  al.  1994;  Delitala  et  al. 
1994;  Facchinetti  et  al.  1994;  Inder  et 
al.  1995#;  Kassimos  et  al.  1996).  For 
example,  persons  with  less  opioid 
activity  (less  inhibitory  tone  directed 
at  CRH  neurons)  would  be  maximally 
blocked  by  a  lower  dose  of  naloxone 
compared  with  persons  with  greater 
opioid  activity  (more  inhibitory  tone), 
who  require  higher  doses  of  naloxone 
to  induce  blockade.  In  other  words, 
persons  with  less  opioid  activity  would 
be  more  sensitive  to  opioid  receptor 
blockade  than  persons  with  greater 
opioid  activity.  Thus,  opioid  receptor 
blockade  by  naloxone  provides  a  non- 
invasive, functional  assessment  of 
hypothalamic  opioid  activity. 

To  explore  the  hypothesis  that  off- 
spring from  families  with  a  high  density 
of  alcohol-dependent  persons  have 
altered  hypothalamic  opioid  activity 
compared  with  offspring  from  families 


406 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


with  no  history  of  alcoholism,  we 
employed  opioid  receptor  blockade 
with  naloxone  (Wand  et  al.  1998). 
The  impact  of  opioid  blockade  by 
naloxone  was  evaluated  by  measuring 
Cortisol  response  curves  over  2  hours 
following  administration  of  different 
naloxone  doses.  To  date,  we  have 
conducted  this  analysis  in  over  70 
nonalcoholic  subjects  ages  18-25. 
Half  of  the  subjects  were  offspring 
from  families  with  a  high  density  of 
alcohol  dependence  and  were  desig- 
nated as  FHP  subjects;  the  other  half 
of  the  sample  population  were  the  off- 
spring of  non-alcohol -dependent  par- 
ents and  were  designated  as  FHN 
subjects.  Subjects  received  graded 
amounts  of  naloxone  in  double- blind, 
randomized  order,  and  serum  Cortisol 
was  monitored.  FHN  subjects  demon- 
strated a  graded  Cortisol  response  to 
each  dose  of  naloxone.  In  contrast, 
FHP  subjects  were  maximally  blocked 
by  the  lowest  dose  of  naloxone.  These 
results  show  that  FHP  subjects  are 
more  sensitive  to  naloxone  relative  to 
FHN  subjects. 

Although  there  are  several  potential 
neurochemical  explanations  for  this 
finding  (Wand  et  al.  1998),  we  specu- 
late that  FHP  subjects  have  dimin- 
ished hypothalamic  opioid  activity 
compared  with  FHN  subjects.  Fur- 
thermore, we  speculate  that  chronic 
ethanol  ingestion  may  exacerbate  this 
underlying  opioid  defect.  In  our  model, 
offspring  of  alcoholics  are  born  with, 
or  acquire,  a  deficit  in  opioid  activity, 
compared  with  offspring  of  nonalco- 
holics  ("trait").  Moreover,  alcohol- 
dependent  persons,  regardless  of  family 
history,  will  evidence  an  opioid  deficit 


as  a  consequence  of  chronic  alcohol 
abuse  ("state").  Lastly,  FHP  alcohol- 
dependent  persons  will  have  even  less 
opioid  activity  then  FHN  alcohol- 
dependent  persons  due  to  additive  effects 
of  trait  and  state  factors.  These  trait  and 
state  differences  in  opioid  activity  may 
simultaneously  account  for  abnormal 
mesolimbic  dopamine  generation  and 
for  abnormal  HPA  axis  dynamics  in 
alcoholics  and  in  persons  at  increased 
risk  for  alcoholism.  We  think  that  this 
alteration  in  endogenous  opioid  activ- 
ity modulates  the  vulnerability  to  and 
the  maintenance  of  alcoholism  for  at 
least  three  reasons: 

1.  Abnormally  low  amounts  of 
endogenous  opioid  activity  in  the 
offspring  of  alcoholics  may  create  a 
neurochemical  milieu,  which  pre- 
disposes them  to  drug-seeking 
behaviors  (trait). 

2.  Abnormal  reductions  of  opioid  activ- 
ity in  the  alcoholic,  as  a  result  of 
chronic  alcohol  exposure,  may  inten- 
sify craving  for  alcohol  and  may  alter 
mood  and  affect  in  a  manner  that 
makes  relapse  more  likely  (state). 

3.  Low  opioid  activity  contributes  to 
a  labile  HPA  axis;  the  ensuing  hyper- 
cortisolism  may  sensitize  the  meso- 
limbic system  to  drugs  of  abuse, 
including  alcohol. 


AREAS  FOR  FUTURE 
INVESTIGATION 

Studies  of  the  HPA  axis  can  inform  us 
about  genetic  and  neurobiological  risk 


407 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


for  alcohol  dependence.  This  is  not 
surprising  when  one  considers  that  the 
CRH  neuron,  which  is  responsible  for 
initiating  the  HPA  axis  response,  is 
modulated  by  many  of  the  candidate 
neurotransmitter  systems  implicated  in 
genetic  vulnerability  for  alcoholism. 
CRH  neurons  are  under  inhibitory 
control  by  opioidergic  and  GABAergic 
neurons  and  under  stimulatory  control 
by  serotonergic  and  catecholaminergic 
systems.  Studying  the  release  of  CRH 
provides  a  window  on  CNS  function 
and  can  uncover  potential  group  differ- 
ences in  neurotransmitter  systems  as  a 
function  of  alcoholism  and  family  his- 
tory of  alcoholism.  It  is  difficult  in  liv- 
ing human  subjects  to  make  direct 
measurements  of  these  important  neuro- 
transmitter systems  in  order  to  compare 
them  as  a  function  of  family  history  of 
alcoholism  and  then  estimate  their  rel- 
evance for  vulnerability  for  alcoholism. 
However,  neuroendocrine  strategies 
allow  noninvasive  assessment  of  these 
crucial  pathways  in  living  human  subjects. 
To  this  end,  neuropharmacological 
studies  designed  to  activate  or  block  5- 
hydroxytryptamine  (5-HT),  GABA, 
and  opioidergic  input  on  CRH  neurons 
may  provide  crucial  information.  It  is 
important  that  such  studies  generate 
dose-response  curves  to  provide  mean- 
ingful interpretations.  In  this  manner, 
important  statements  about  opioid,  5- 
HT,  GABA,  and  catecholaminergic 
tone  directed  at  the  CRH  neuron  can 
be  established.  It  is  also  important  that 
nonhormonal  measures  be  included, 
such  as  physiological  and  behavioral 
responses  to  the  neuropharmacological 
challenges.  Such  studies  could  be  per- 
formed during  various  phases  of  alcohol 


dependence:  active  drinking,  acute  with- 
drawal, early  and  late  abstinence.  How- 
ever, because  chronic  ethanol  exposure 
can  injure  the  HPA  axis,  studies 
would  be  more  powerful  using  non- 
alcohol-dependent  subjects  in  a  family 
history  design.  Moreover,  the  use  of 
positron  emission  tomography  or 
other  imaging  measurements  of  5-HT, 
GABA,  and  opioids  in  combination 
with  neuroendocrine  challenges  may 
provide  powerful  supplemental  confir- 
mation of  alterations  in  neurotrans- 
mitter systems  as  a  function  of  alcoholism 
and  family  history  of  alcoholism. 

The  San  Diego  group  has  provided 
important  evidence  that  ACTH  and 
Cortisol  responses  following  an  ethanol 
challenge  have  predictive  value  vis-a-vis 
the  future  development  of  alcoholism. 
It  is  now  important  to  establish  the 
mechanism  through  which  high-  and 
low-risk  persons  show  a  differential 
HPA  axis  response  to  ethanol.  Is  it 
possible  that  such  differences  are  not 
only  predictive  but  have  causal  impli- 
cations as  well?  Moreover,  as  the  num- 
ber of  markers  for  the  risk  of  alcohol 
dependence  grows,  it  would  be  benefi- 
cial to  know  if  altered  HPA  axis 
dynamics  identified  in  high-risk  nonal- 
coholics  co-segregates  with  other  mark- 
ers of  risk  (e.g.,  reduced  P300  amplitude 
or  low  adenylyl  cyclase  activity). 

Differences  in  the  dynamics  of  the 
HPA  axis  may  distinguish  animals 
with  ethanol-seeking  behavior  from 
animals  that  will  not  voluntarily  drink 
ethanol.  As  noted  earlier  in  this  chap- 
ter, there  are  genetic  as  well  as  envi- 
ronmental factors  that  determine  the 
dynamics  of  the  HPA  axis.  In  otherwise 
healthy  people,  there  is  a  continuum 


408 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


of  HPA  axis  activity  (e.g.,  from  low  to 
high  responders).  On  either  end  of 
the  HPA  axis  spectrum,  low  or  high 
producers  of  CRH,  ACTH,  and  Corti- 
sol may  be  at  risk  for  certain  behav- 
ioral disturbances.  Drug-reinforcing 
properties  may  be  related  to  the  sensi- 
tivity level  of  the  mesolimbic  system 
(Samson  1995).  The  sensitivity  of  the 
mesolimbic  system  to  drugs  of  abuse 
may  be  modulated  by  glucocorticoids. 
In  this  manner,  the  onset  and  magni- 
tude of  sensitization  to  drug  actions 
may  be  modulated  by  stress.  Address- 
ing this  issue,  Samson  hypothesized 
that  "stress,  via  its  ability  to  result  in 
sensitization  and  altered  mesolimbic 
dopamine  system  function,  results  in  an 
increase  in  the  reinforcing  efficacy  of 
alcohoF  (pp.  284-285). 

Since  the  degree  of  HPA  axis  lability 
may  influence  alcohol-seeking  behav- 
iors, it  behooves  us  to  systematically 
analyze  and  compare  HPA  axis  dynam- 
ics as  a  function  of  family  history  of 
alcoholism.  To  this  end,  there  should 
be  a  head-to-head  comparison  of  the 
entire  HPA  axis  in  alcohol-dependent 
and  non-alcohol- dependent  people  as 
a  function  of  family  history  of  alco- 
holism. A  series  of  studies  could  compare 
integrated  measurements  of  24-hour 
ACTH  and  Cortisol  production  in 
stressed  and  nonstressed  persons. 
Studies  could  also  measure  hypothala- 
mic CRH/AVP  responses  to  nalox- 
one or  hypoglycemia,  pituitary  ACTH 
responses  to  CRH,  and  adrenocortical 
responses  to  ACTH.  Additionally, 
much  may  be  gained  by  measuring 
the  glucocorticoid  negative  feedback 
set-point  as  a  function  of  family  history 
of  alcoholism.  Moreover,  the  behavioral 


(e.g.,  reinforcing)  effects  of  CRH, 
ACTH,  and  glucocorticoids  should  be 
studied  in  a  family  history  design  in 
both  alcoholics  and  nonalcoholics. 

One  potential  confounding  variable 
in  family  history  designs  is  that  the 
subjects,  although  not  alcohol  abusers, 
have  been  exposed  to  alcohol.  This 
means  a  certain  degree  of  tolerance  to 
ethanol  may  already  be  established.  In 
addition,  concerns  for  multiple  blood 
drawing  and  administering  pharmaco- 
logical agents  in  children  have  pre- 
vented serious  hormonal  assessments 
in  preteenage,  ethanol-naive  offspring 
of  alcohol-dependent  persons.  The  use 
of  salivary  Cortisol  measurements  in 
preteenagers  may  help  in  this  regard. 
Multiple  salivary  Cortisol  determina- 
tions in  a  24-hour  period  can  be  per- 
formed with  little  or  no  distress  for 
the  subject. 

Studies  using  humans  and  rodents 
have  indicated  that  anxiety  and  stress 
are  positively  correlated  with  high 
alcohol  consumption  as  well  as  relapse 
to  heavy  drinking  by  abstinent  alco- 
holics (Hore  1971;  Kushner  et  al. 
1990).  The  reason  for  this  relationship 
between  stress  and  alcohol  drinking  is 
not  well  understood.  It  has  been  sug- 
gested that  alcohol  consumption 
relieves  anxiety  and  as  a  result  helps 
the  individual  to  cope  with  the  stress- 
ful situation  (Pohorecky  1991).  The 
possible  relationship  between  stress 
and  alcohol  consumption  has  led 
some  investigators  to  propose  a  "tension 
reduction  hypothesis"  to  explain  a 
subset  of  alcoholism  and  the  ability  of 
alcohol  to  significantly  dampen  this 
response  (Pohorecky  1991).  Self- 
medication  with  alcohol  for  anxiolytic 


409 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


purposes  may  be  one  route  through 
which  high-risk  persons  begin  to 
become  alcohol  dependent.  The 
effects  of  stress  dampening  by  ethanol 
should  be  studied  in  a  family  history 
design  using  the  HPA  axis  as  the  end 
point  measures. 

Although  understanding  the  interac- 
tions of  the  HPA  axis  and  alcoholism 
may  help  in  developing  better  methods 
of  prevention  and  treatment  and  under- 
standing neuromechanisms  that  con- 
tribute to  alcohol  dependence,  it  is  also 
important  to  understand  why  alcohol- 
dependent  persons  are  at  increased  risk 
for  severe  infections.  Since  the  HPA  axis 
and  immune  system  modulate  one 
another,  the  increased  risk  of  infection 
may  be  the  result  of  both  direct  and  indi- 
rect sequelae  on  both  systems.  Future 
studies  should  more  clearly  define  the 
relationship  between  ethanol -induced 
injury  to  the  HPA  axis  and  immune  sys- 
tem. It  is  also  important  to  determine  if 
alcohol-dependent  persons  would  ben- 
efit from  stress  glucocorticoid  cover- 
age during  acute  illness. 

ACKNOWLEDGMENT 

Preparation  of  this  chapter  was  sup- 
ported by  grant  ROl  10158  from  the 
National  Institute  on  Alcohol  Abuse 
and  Alcoholism. 


REFERENCES 

Adinoff,  B.  Double  blind  study  of  alpra- 
zolam, diazepam,  clonidine  and  placebo 
in  the  alcohol  withdrawal  syndrome. 
Alcohol  Clin  Exp  Res  18:873-878,  1994. 

Adinoff,  B.;  Risher-Flowers,  D.;  De  long 
I;  Ravitz,  B.;  Bone,  G.;  Nutt,  D.; 


Roehrich,  L.;  Martin,  P.;  and  Linnoila,  M. 
Disturbances  of  hypothalamic -pituitary- 
adrenal  axis  functioning  during  ethanol 
withdrawal  in  six  men.  Am  J  Psychiatry 
148(8):1023-1025,  1991. 

Aguirre,  J.;  Del  Arbol,  J.;  Rico,  J.;  Raya, 
J.;  and  Ruiz-Requena,  M.  Effect  of  acute 
alcohol  intoxication  on  the  opioid  system 
in  humans.  Alcohol T2(6):559-562,  1995. 

Altshuler,  H.;  Phillips,  P.;  and  Feinhandler, 
D.  Alteration  of  ethanol  self- administration 
by  naltrexone.  Life  Sci  26:679-688,  1980. 

Begleiter,  H.;  Porjesz,  B.;  Bihari,  B.;  and 
Kissin,  B.  Event-related  potentials  in  boys 
at  risk  for  alcoholism.  Science  225:1493- 
1496,  1984. 

Benjamin,  D.;  Grant,  E.;  and  Pohorecky, 
L.  Naltrexone  reverses  ethanol  stimulated 
dopamine  release  in  the  nucleus  accum- 
bens  of  awake  freely  moving  rats.  Alcohol 
Clin  Exp  Res  16:617-626,  1992. 

Burrov,  Y.;  Treskov,  V.;  Vedernikova,  N.; 
and  Shevelyova,  O.  Types  of  alcohol 
withdrawal  syndrome  and  dexamethasone 
suppression  test.   Drug  Alcohol  Depend 
17:81,  1986. 

Chick,  J.;  Erickson,  C.;  and  the  Amsterdam 
Consensus  Conference  Participants. 
Conference  summary:  Consensus  conference 
on  alcohol  dependence  and  the  role  of 
pharmacotherapy  in  its  treatment.  Alcohol 
Clin  Exp  Res  20:391^102,  1996. 

Chrousos,  G.,  and  Gold,  P.  The  concepts 
of  stress  and  stress  system  disorders:  Overview 
of  behavioral  and  physical  homeostasis. 
JAMA  267:1244-1252,  1992. 

Cloninger,  C.  Etiologic  factors  in  sub- 
stance abuse:  An  adoption  study  perspec- 
tive. NIDA  ResMonogr  89:52-72,1988. 

Costa,  A.;  Buno,  G.;  Martignoni,  E.; 
Merlo,  P.;  Sances,  G.;  and  Nappi,  G.  An 


410 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


assessment  of  HPA  axis  functioning  in  non- 
depressed,  early  abstinent  alcoholics.  Psycho- 
neuroendocrinology  21(3):263-275,  1996. 

Cotton,  N.  The  familial  incidence  of  alco- 
holism./S*w/*A/«wfo/  40:89-1 16,  1979. 

De  Waele,  J.,  and  Gianoulakis,  C.  Enhanced 
activity  of  the  brain  (3-endorphin  system 
by  free-choice  ethanol  drinking  in  C57BL/ 
6  but  not  DBA/2  mice.  Eur  J  Pharmacol 
258:119-129,  1994. 

De  Waele,  J.;  Papachristou,  D.;  and 
Gianoulakis,  C.  The  alcohol-preferring 
C57BL/6  mice  present  an  enhanced 
sensitivity  of  the  hypothalamic  (3-endor- 
phin system  to  ethanol  than  the  alcohol 
avoiding  DBA/2  mice.  /  Pharmacol  Exp 
Ther  261:788-794,  1992. 

Del  Campo,  M.;  Dowson,  J.;  Herbert,  J.; 
and  Paykel,  E.  Effects  of  naloxone  on 
diurnal  rhythms  in  mood  and  endocrine 
function:  A  dose-response  study  in  man. 
Psychopharmacology  144:583-590,  1994. 

Del  Porto,  J.;  Monteiro,  M.;  Laranjeiro, 
R.;  Jorge,  M.;  and  Masur,  J.  Reversal  of 
abnormal  dexamethasone  suppression  rest 
in  alcoholics  abstinent  for  4  weeks.  Biol 
Pzychol  20:1 156,  1985. 

Delitala,  G.;  Trainer,  P.;  Oliva,  O.; 
Fanciulli,  G.;  and  Grossman,  A.  Opioid 
peptide  and  a- adrenoceptor  pathways  in 
the  regulation  of  the  pituitary- adrenal  axis 
in  man.  ]  Endocrinol  141:163-168,  1994. 

Deroche,  V.;  Marinelli,  M.;  and  Maccar, 
S.  Stress-induced  sensitization  and  gluco- 
corticoids. JNeurosci  15:181-188,  1995. 

Erb,  S.;  Shaham,  Y.,  and  Stewart,  J.  The 
role  of  corticotropin -releasing  factor  and 
corticosterone  in  stress-cocaine-induced 
relapse  to  cocaine  seeking  in  rats.  / 
Neurosci  18:5529-5536,  1998. 


Facchinetti,  F.;  Fioroni,  L.;  Martignoni, 
E.;  Sances,  G.;  Costa,  A.;  and  Genazzani, 
A.  Changes  of  opioid  modulation  of  the 
hypothalamo-pituitary- adrenal  axis  in 
patients  with  severe  premenstrual  syndrome. 
Psychosom  Med  56:418-422,  1994. 

Finn,  P.;  Zeitouni,  N.;  and  Pihl,  R. 
Effects  of  alcohol  on  psychophysiological 
hyperactivity  to  nonaversive  and  aversive 
stimuli  in  men  at  high  risk  for  alcoholism. 
J  Abnorm  Psychol  99:79-85,  1990. 

Frajria,  R,  and  Angeli,  A.  Alcohol-induced 
pseudo-Cushing's  syndrome.  Lancet 
1:1050-1051,  1977. 

Froehlich,  J.;  Harts,  J.;  Lumeng,  L.;  and 
Li,  T.  Naloxone  attenuates  voluntary 
ethanol  intake  in  rats  selectively  bred  for 
high  ethanol  preference.  Pharmacol 
Biochem  Behav  35:385-390,  1990. 

Gianoulakis,  C;  and  De  Waele,  J. 
Genetics  of  alcoholism:  Role  of  endoge- 
nous opioid  system.  Metabol  Brain  Dis 
9:105-131,  1994. 

Gianoulakis,  C;  Krishman,  B.;  and 
Thayundayil,  J.  Enhanced  sensitivity  of 
pituitary  (3-endorphin  to  ethanol  in  sub- 
jects at  high  risk  of  alcoholism.  Arch  Gen 
Psychiatry  53:250-257,  1996. 

Gold,  P.;  Goodwin,  F.;  and  Chrousos,  G. 
Clinical  and  biochemical  manifestations  of 
depression:  Relationship  to  the  neurobiol- 
ogy of  stress,  part  1.  N  Engl  J  Med  319: 
348-353,  1988a. 

Gold,  P.;  Goodwin,  F.;  and  Chrousos,  G. 
Clinical  and  biochemical  manifestations  of 
depression:  Relationship  to  the  neurobiol- 
ogy of  stress,  part  2.  N Engl  J Med  319: 
413^120,  19886. 

Gold,  P.;  Pigott,  T.;  Kling,  M.;  Kalogeras, 
K.;  and  Chrousos,  G.  Basic  and  clinical 
studies  with  corticotropin  releasing  hor- 
mone: Implications  for  a  possible  role  in 


411 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


panic  disorder.  Psychiatr  Clin  North  Am 
11:327-334,  1988c. 

Goodwin,  D.;  Schulsinger,  F.;  Hermansen, 
L.;  Guze,  S.;  and  Winokur,  G.  Alcohol 
problems  in  adoptees  raised  apart  from 
alcoholic  biological  parents.  Arch  Gen 
Psychiatry  28:238-243,  1973. 

Griep,  E.;  Boerdma,  J.;  and  de  Kloet,  E. 
Altered  reactivity  of  the  hypothalamic  - 
pituitary-adrenal  axis  in  the  primary 
fibromyalgia  syndrome.  J  Rheumatol 
20:469-474,  1993. 

Hill,  S.;  Steinhauer,  S.;  Zubin,  J.;  and 
Baughman,  T.  Event-related  potentials  as 
markers  for  alcoholism  risk  in  high  density 
families.  Alcohol  Clin  Exp  Res  12:545- 
554, 1988. 

Hore,  B.  Factors  in  alcohol  relapse.  Br  J 
Addict  66:89-96,  1971. 

Hrubec,  S.,  and  Omenn,  G.  Evidence  of 
genetic  predisposition  to  alcoholic  cirrho- 
sis and  psychosis:  Twin  concordances  for 
alcoholism  and  its  biological  end  points 
by  zygosity  among  male  veterans.  Alcohol 
Clin  Exp  Res  5 :207-2 15,1981. 

Hubbell,  C.;  Marglin,  S.;  Spitalnic,  S.; 
Abelson,  J.;  Wild,  K.;  and  Reid,  L. 
Opioidergic,  serotonergic,  and  dopami- 
nergic manipulations  and  rats'  intake  of  a 
sweetened  alcoholic  beverage.  Alcohol 
8:355-367,  1991. 

Hubbell,  C.;  Czirr,  S.;  Hunter,  G.; 
Beaman,  C;  LeCann,  N.;  and  Reid,  L. 
Consumption  of  ethanol  solution  is  po- 
tentiated by  morphine  and  attenuated  by 
naloxone  persistently  across  repeated  daily 
administrations.  Alcohol  3:39-54,  1996. 

Inder,  W.;  Hellemans,  J.;  Ellis,  M.;  Evans, 
M.;  Livesey,  J.;  and  Donald,  R.  Elevated 
basal  adrenocorticotropin  and  evidence 
for  increased  central  opioid  tone  in  highly 


trained  male  athletes.  J  Clin  Endocrinol 
Metab  80(l):244-248,  1995«. 

Inder,  W.;  Joyce,  P.;  Ellis,  M.;  Evans,  M.; 
Livesey,  J.;  and  Donald,  R.  The  effects  of 
alcoholism  on  the  hypothalamic -pituitary- 
adrenal  axis:  Interaction  with  endogenous 
opioid  peptides.  Clin  Endocrinol  (Oxf) 
43:283-290,  19956. 

Insel,  T.;  Kalin,  N.;  Guttmacher,  L.; 
Cohen,  R;  and  Murphy,  D.  The  dexa- 
methasone  suppression  test  in  patients 
with  primary  obsessive-compulsive  disor- 
der. Psychiatry  Res  6:153,  1982. 

Iranmanesh,  A.;  Veldhuis,  J.;  Johnson, 
M.;  and  Lizarraide,  G.  24-hr  pulsatile  and 
circadian  pattern  of  Cortisol  secretion  in 
alcoholic  men.  /  Androl  10:54,  1989. 

Kaig,  L.  Alcoholism  in  Twins.  Stockholm, 
Sweden:  Almqvist  and  Wiksell,  1960. 

Kassimos,  D.;  Choy,  E.;  Grossman,  A.; 
Chikanza,  I.;  and  Panayi,  G.  Endogenous 
opioid  tone  in  patients  with  rheumatoid 
arthritis.  Br  J  Rheumatol  35:436-440, 1996. 

Kendler,  K.;  Heath,  A.;  Neale,  M.;  Kessler, 
R.;  and  Eaves,  L.  A  population-based  twin 
study  of  alcoholism  in  women.  JAMA 
268(14):1877-1882,  1992. 

Kirkman,  S.,  and  Nelson,  D.  Alcohol- 
induced  pseudo-Cushing's  disease:  A 
study  of  prevalence  with  review  of  the 
literature.  Metabolism  37:390,  1988. 

Koob,  G.;  Roberts,  A.;  Schulteis,  G.; 
Parsons,  L.;  Charles,  J.;  Hyytia,  P.; 
Merlo-Pich,  E.;  and  Weiss,  F.  Neuro- 
circuitry  targets  in  ethanol  reward  and 
dependence  (review).  Alcohol  Clin  Exp 
Res  22:3-9,  1988. 

Kushner,  M.;  Sher,  K.;  and  Beitman,  B. 
The  relation  between  alcohol  problems 
and  anxiety  disorders.  Am  J  Psychiatry 
147:685-695,  1990. 


412 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


LaFuente,  J.;  Rosenbaum,  A.;  Morse  R.; 
Niven,  R.;  Abboud,  C;  Jiang,  N.;  and 
Schatzberg,  A.  The  hypothalamic-pituitary- 
adrenal  axis  in  alcoholics.  Alcohol  Clin 
Exp  Res  7:35,  1983. 

Lex,  B.;  Lukas,  S.;  Greenwald,  N.;  and 
Mendelson,  J.  Alcohol-induced  changes 
in  body  sway  in  women  at  risk  for  alco- 
holism: A  pilot  study.  /  Stud  Alcohol 
49:346-356,  1988. 

Luger,  A.;  Deuster,  P.;  Kyle,  S.;  Gallucci, 
W.;  Montgomery  L.;  Gold,  P.;  Loriaux, 
D.;  and  Chrousos,  G.  Acute  hypothala- 
mic-pituitary- adrenal  responses  to  the 
stress  of  treadmill  exercise:  Physiologic 
adaptations  to  physical  training.  N  Engl  J 
Med  316:1309-1315, 1987. 

Marfaing-Jallat,  P.;  Miceli,  D.;  and 
LeMagnen,  J.  Decrease  in  ethanol  con- 
sumption by  naloxone  in  naive  and  de- 
pendent rats.  Pharmacol  Biochem  Behav 
18(Suppl  l):537-539,  1983. 

Mendelson,  J.,  and  Stein,  S.  The  definition 
of  alcoholism.  Int  Psych  Clin  3:3-16,  1966. 

Menzagh,  F.;  Rassnick,  S.;  and  Heinrichs, 
S.  Role  of  CRF  in  the  anxiogenic  effects 
of  ethanol  withdrawal.  Ann  NT  Acad  Sci 
739:176-184,  1994. 

Merikangas,  K.  The  genetic  epidemiology 
of  alcoholism.  Psychol  Med  20:11-22,  1990. 

O'Malley,  S.;  Jaffe,  A;  Chang,  G.; 
Schottenfeld,  R.;  Meyer,  R.;  and 
Rounsaville,  B.  Naltrexone  and  coping 
skills  therapy  for  alcohol  dependence.  A 
controlled  study.  Arch  Gen  Psychiatry 
49:881-887,  1992. 

Pasquali,  R.;  Cantobelli,  S.;  Casimiri,  F.; 
Capelli,  M.;  Bortoluzzi,  L.;  Flamia,  R.; 
Labate,  A.;  and  Barbara,  L.  The  hypothal- 
amic-pituitary- adrenal  axis  in  obese 
women  with  different  patterns  of  body  fat 


distribution.  /  Clin  Endocrinol  Metab 
77:341-346,  1993. 

Paton,  A.  Alcohol-induced  cushingoid 
syndrome.  Br  Med  J  2:1504,  1976. 

Pfefferbaum,  A.;  Ford  ,  J.;  White,  P.;  and 
Mathalon,  D.  Event-related  potentials  in 
alcoholic  men:  P3  amplitude  reflects  fam- 
ily history  but  not  alcohol  consumption. 
Alcohol  Clin  Exp  Res  15(5):839-850, 1991. 

Pickens,  R.;  Svikis,  D.;  McGue,  M.; 
Lykken,  D.;  Heston,  L.;  and  Clayton,  P. 
Heterogeneity  in  the  inheritance  of  alcohol- 
ism. Arch  Gen  Psychiatry  48:19-28,  1991. 

Pohorecky,  L.  Stress  and  alcohol  interac- 
tions: An  update  of  human  research.  Alcohol 
Clin  Exp  Res  15:438-458,  1991. 

Raadsheer,  F.;  Hoogendijk,  W.;  Stam,  F.; 
Tilders,  F.;  and  Swaab,  D.  Increased  num- 
bers of  corticotropin-releasing  hormone 
expressing  neurons  in  the  hypothalamic 
paraventricular  nucleus  of  depressed  patients. 
Neuroendocrinolqgy  60:436^44,  1994. 

Rees,  L.;  Besser,  G.;  Jeffcoate,  W.;  Goldie, 
D.;  and  Marks,  V.  Alcohol-induced 
pseudo-Cushing's  syndrome.  Lancet 
1:726,  1977. 

Reid,  L.  Summary.  In:  Reid,  L.D.,  ed. 
Opioids,  Bulimia,  Alcohol  Abuse  and 
Alcoholism.  New  York:  Springer- Verlag, 
1990. pp.  289-304. 

Reid,  L.,  and  Hunter,  G.  Morphine  and 
naloxone  modulate  intake  of  ethanol. 
Alcohol  1:33-37,  1984. 

Reid,  L.;  Delconte,  J.;  Nichols,  M.;  Bilsky, 
E.;  and  Hubbell,  C.  Tests  of  opioid  defi- 
ciency hypothesis  of  alcoholism.  Alcohol 
8:247-257,  1991. 

Rivier,  C.  Acute  interactions  between 
cytokine  and  alcohol  on  ACTH  and  corti- 
costerone  secretion  in  rats.  Alcohol  Clin 
Exp  Res  17:946-950,  1993. 


413 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Rivier,  C;  Bruhn,  T.;  and  Vale,  W.  Effect 
of  ethanol  on  the  hypothalamic -pituitary- 
adrenal  axis  in  the  rat:  Role  of  corticotropin- 
releasing  factor  (CRF).  J  Pharmacol  Exp 
T^r  229: 127-1 3 1,1984. 

Robin,  D.;  Schmidt,  P.;  Campbell,  G; 
Gold,  P.;  Jensvold,  M.;  Rubinow,  D.;  and 
Chrousos,  G.  Hypothalamic -pituitary- adrenal 
function  in  patients  with  the  premenstrual 
syndrome.  /  Clin  Endocrinol  Metab  71 : 
1158-1162,1990. 

Samson,  H.  Environmental  effects  on 
drinking  patterns:  Stress  and  sensitization. 
In:  Hunt,  W.A.,  and  Zakhari,  S.,  eds. 
Stress,  Gender,  and  Alcohol-Seeking  Behavior. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  29. 
NIH  Pub.  No.  95-3893.  Bethesda,  MD: 
the  Institute,  1995.  pp.  277-291. 

Samson,  H.,  and  Doyle,  T.  Oral  ethanol 
self- administration  in  the  rat:  Effect  of 
naloxone.  Pharmacol  Biochem  Behav 
22:91-100,  1985. 

Sapolsky,  R  Why  stress  is  bad  for  your 
brain.  Science  273:74-75,  1996. 

Schuckit,  M.,  and  Smith,  T.  An  8 -year 
follow-up  of  450  sons  of  alcoholic  and 
control  subjects.  Arch  Gen  Psychiatry 
53:202-210,  1996. 

Schuckit,  M.;  Gold,  E.;  and  Risch,  C. 
Plasma  Cortisol  levels  following  ethanol  in 
sons  of  alcoholics  and  controls.  Arch  Gen 
Psychiatry  44:942-945,  1987. 

Schuckit,  M.;  Gold,  E.;  and  Risch,  C. 
Serum  prolactin  levels  following  ethanol 
in  sons  of  alcoholics  and  controls.  Am  J 
Psychiatry  144:854-859,  1987. 

Schuikin,  J.;  McEwen,  B.;  and  Gold,  P. 
Allostasis,  amygdala  and  anticipatory  angst. 
Neurosci  Behav  Rev  18:385-396,  1994. 


Sher,  K.,  and  Trull,  T.  Personality  and 
disinhibitory  psychopathology.  J  Abnorm 
Psychol  103:92-102,  1994. 

Smals,  A.;  Kloppenborg,  P.;  Njo,  K.; 
Knoben,  J.;  and  Ruland,  C.  Alcohol- 
induced  cushingoid  syndrome.  Br  Med  J 
2:129,  1976. 

Stansbury,  K.,  and  Gunnar,  M.  Adreno- 
cortical activity  and  emotion  regulation. 
Monogr  Soc  Res  Child  Dev  59(2-3): 
108-134, 1994. 

Sternberg,  E.;  Glowa,  J.;  and  Smith,  M. 
Corticotropin  releasing  hormone  related 
behavioral  and  neuroendocrine  responses 
to  stress  in  Lewis  and  Fischer  rats.  Brain 
Res  570:54-60,  1992. 

Swartz,  C,  and  Dunner,  F.  Dexamethasone 
suppression  testing  of  alcoholics.  Arch 
Gen  Psychiatry  39:1309,  1982. 

Thiagarajan,  A.;  Mefford,  I.;  and  Eskay, 
R.  Single-dose  ethanol  administration 
activates  the  hypothalamic-pituitary-adrenal 
axis:  Exploration  of  the  mechanism  of  action. 
Neuroendocrinology  50:427-432,  1989. 

Torpy,  D.;  Grice,  J.;  Hockings,  G.; 
Walters,  M.;  Crosbie,  G.;  and  Jackson,  R 
Alprazolam  blocks  the  naloxone-stimu- 
lated  hypothalamo-pituitary-adrenal  axis 
in  man.  /  Clin  Endocrinol  Metab  76(2): 
388-391,  1993 

Tsigos,  C,  and  Chrousos,  G.  The  neuro- 
endocrinology  of  the  stress  response.  In: 
Hunt,  W.A.,  and  Zakhari,  S.,  eds.  Stress, 
Gender,  and  Alcohol-Seeking  Behavior. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  29. 
NIH  Pub.  No.  95-3893.  Bethesda,  MD: 
the  Institute,  1995.  pp.  103-124. 

Volpicelli,  J.;  Alterman,  A.;  Hayashida, 
M.;  and  O'Brien,  C.  Naltrexone  in  the 
treatment  of  alcohol  dependence.  Arch 
Gen  Psychiatry  49:876-888,  1992. 


414 


The  HPA  Axis:  Changes  and  Risk  for  Alcoholism 


Waltman,  C;  Blevins,  L.,  Jr.;  Boyd,  G.; 
and  Wand,  G.  The  effects  of  mild  ethanol 
intoxication  on  the  hypothalamic-pituitary- 
adrenal  axis  in  nonalcoholic  men.  /  Clin 
Endocrinol  Metab  77(2):518-522,  1993. 

Waltman,  C.;  McCaul,  M.;  and  Wand,  G. 
Adrenocorticotropin  responses  following 
administration  of  ethanol  and  ovine  corti- 
cotropin-releasing  hormone  in  the  sons  of 
alcoholics  and  control  subjects.  Alcohol 
Clin  Exp  Res  18(4):826-830,  1994. 

Wand,  G.  Ethanol  differentially  regulates 
proadrenocorticotropin-endorphin  produc- 
tion and  corticosterone  secretion  in  LS  and 
SS  lines  of  mice.  Endocrinology  124:518- 
526,  1989. 

Wand,  G.  Differential  regulation  of  ante- 
rior pituitary  corticotrope  function  is 
observed  in  vivo  but  not  in  vitro  in  two 
lines  of  ethanol-sensitive  mice.  Alcohol 
Clin  Exp  Res  14:100-106,  1990. 

Wand,  G.,  and  Dobs,  A.  Alterations  in 
the  hypothalamic-pituitary-adrenal  axis  in 
actively  drinking  alcoholics.  /  Clin 
Endocrinol  Metab  72:1290-1295,  1991. 

Wand,  G.,  and  Levine,  M.  Hormonal 
tolerance  to  ethanol  is  associated  with 
decreased  expression  of  the  GTP-  binding 
protein,  Gsct,  and  adenylyl  cyclase  activity 
in  ethanol-treated  LS  mice.  Alcohol  Clin 
Exp  Res  15:705-710,  1991. 


Wand,  G.;  Waltman,  C;  McCaul.  M.; 
Levine,  M.;  and  Wolfgang,  D.  Differential 
expression  of  GTP- binding  proteins  in 
individuals  with  high  and  low  risk  for  the 
future  development  of  alcoholism.  J  Clin 
Invest  94: 1004-1011,  1994. 

Wand,  G.;  Mangold,  D.;  El  Deiry,  S.; 
McCaul,  M.;  and  Hoover,  D.  Family 
history  of  alcoholism  and  hypothalamic 
opioidergic  activity.  Arch  Gen  Psychiatry 
55:1114-1119,1998. 

Weiss,  F.;  Mitchiner,  M.;  Bloom,  F.;  and 
Koob,  G.  Free-choice  responding  for 
ethanol  versus  water  in  alcohol  preferring 
and  unselected  Wistar  rats  is  differentially 
modified  by  naloxone,  bromocriptine  and 
methysergide.  Psychopharmacology 
101:178-186,  1990. 

Weissman,  M.;  Pottenger,  M.;  Kleber,  H.; 
Ruben,  H.;  Williams  D.;  and  Thompson, 
W.  Symptom  patterns  in  primary  and 
secondary  depression:  A  comparison  of 
primary  depressives  with  depressed  opiate 
addicts,  alcoholics,  and  schizophrenics. 
Arch  Gen  Psychiatry  34:854-862,  1977. 

Willenbring,  M.;  Morley,  J.;  Niewoehner, 
C;  Heilman,  R.;  Carlson,  C;  and  Shafer, 
R  Adrenocortical  hyperactivity  in  newly 
admitted  alcoholics:  Prevalence,  course 
and  associated  variables.  Psychoneuro- 
endocrinology  9:415-422,  1984. 


415 


Chapter  12 
Alcohol  and  Sleep 


Cindy  L.  Ehlers,  Ph.D. 

KEY  WORDS:  Sleep  disorder;  respiratory  disorder;  acute  AODE  (effect  ofAOD 
[alcohol  or  other  drug  effects]  use,  abuse,  and  dependence);  respiratory  system 
function;  electroencephalography;  REM  sleep;  circadian  rhythm;  polypeptide  hor- 
mones; risk  factors;  relapse  prevention;  muscle  function;  literature  review 


EFFECTS  OF  ALCOHOL  ON 
ELECTROENCEPHALO- 
GRAPHIC  (EEG)  SLEEP 
AND  BREATHING 
DURING  SLEEP 

Acute  doses  of  alcohol  (ethanol)  sig- 
nificantly alter  the  sleep  of  healthy 
subjects,  producing  changes  in  the 
electroencephalogram,  eye  movements 
during  sleep,  muscle  activity,  and 
breathing  patterns.  Changes  in  sleep 
patterns  produced  by  acute  alcohol  in 
normal  subjects  include  increases  in 
delta  (slow  wave,  stages  3  and  4)  sleep 
(Yules  et  al.  1967;  MacLean  and 
Cairns  1982)  and  delta  EEG  power 
(Landolt  et  al.  1996),  reductions  in 
rapid  eye  movement  (REM)  sleep 


(Gresham  et  al.  1963;  Knowles  et  al. 
1968;  Rundell  et  al.  1972;  Landolt  et 
al.  1996;  Lobo  and  Tufik  1997),  and 
increases  in  stage  2  sleep  (Yules  et  al. 
1966).  Subjects  given  alcohol  in  exper- 
imental procedures  reported  sleeping 
more  superficially  and  experiencing 
lighter  sleep,  especially  during  the  sec- 
ond half  of  the  night  (Landolt  et  al. 
1996).  Periodic  limb  movements  were 
also  increased  in  some  subjects  and 
may  contribute  to  sleep  disturbance 
following  moderate  alcohol  consump- 
tion (Aldrich  and  Shipley  1993). 

Alcohol  consumption  also  influences 
breathing  during  sleep.  Moderate  doses 
of  alcohol  (0.7-1.05  mL/kg)  produce 
respiratory  depression  (Johnstone  and 
Reier  1973),  vasodilation  and  swelling 


C.L.  Ehlers,  Ph.D.,  is  an  associate  professor  in  the  Department  of  Neuropharmacology,  The  Scripps 
Research  Institute,  CVN-14,  10550  North  Torrey  Pines  Rd.,  Lajolla,  CA  92037. 


417 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  the  respiratory  mucosa  (Rains  et  al. 
1991).  Alcohol  increases  inspiratory 
resistance  during  stage  2  sleep  in  both 
snoring  and  nonsnoring  men  (Dawson 
et  al.  1993,  1997).  The  mechanism 
whereby  alcohol  increases  inspiratory 
resistance  appears  to  be  through  a 
selective  reduction  in  respiratory  motor 
activity  of  the  hypoglossal  and  laryngeal 
nerves.  (Bonora  et  al.  1984;  Krol  et  al. 
1984).  This  reduced  muscle  tone  pre- 
sumably makes  the  upper  airway  more 
likely  to  collapse.  Thus,  it  has  been  sug- 
gested that  alcohol  consumption  can 
lead  to  an  aggravation  of  obstructive 
sleep  apnea  (Taasan  et  al.  1981;  Issa 
and  Sullivan  1982;  Scrima  et  al.  1982; 
Block  et  al.  1986;  Mitler  et  al.  1988; 
Scrima  et  al.  1989),  although  the  data 
are  still  somewhat  controversial 
(Teschler  et  al.  1996). 

Sleep  Apnea  in  Alcoholics 

Sleep  apnea  and  sleep-related  breathing 
and  movement  disturbances  are  not 
uncommon  in  abstinent  alcoholics 
(Tan  et  al.  1985;  Vitiello  et  al.  1987; 
Mamdani  et  al.  1989;  Vitiello  et  al. 
1990«,  1990&;  Aldrich  et  al.  1993;  Le 
Bon  et  al.  1997).  Although  there  is 
evidence  that  sleep-related  breathing 
disorders  are  increased  in  alcoholics, 
there  are  several  mediating  factors  (see 
Aldrich  et  al.  1993).  For  instance, 
Vitiello  and  colleagues  (1987)  found 
increased  nocturnal  hypoxemia  in 
abstinent  male  alcoholics,  but  more 
than  half  of  those  alcoholics  (vs.  none 
of  the  control  subjects)  were  smokers. 
It  has  been  suggested  that  the  increase 
in  sleep  apnea  seen  in  some  alcoholics 
is  secondary  to  either  chronic  lung 
disease  or  cardiovascular  morbidity 


associated  with  smoking  combined  with 
alcohol  use.  However,  an  alternative 
hypothesis  has  been  put  forth  by  Tan 
and  colleagues  (1985),  who  found  that 
central  apneas  and  hypopneas  were 
associated  with  the  presence  of  nervous 
system  damage  in  alcoholics.  It  appears 
that  apneas  in  alcoholics  may  be  of 
mixed  origin  and  may  include  both 
central  nervous  system  effects  and  car- 
diopulmonary factors.  Of  interest  is 
the  fact  that  sleep-disordered  breath- 
ing is  apparently  not  seen  in  alcoholic 
women  (Aldrich  et  al.  1993).  This 
suggests  that  women  may  be  protected 
from  the  effects  of  alcohol  on  the  mech- 
anisms that  generate  apnea. 

EEG  Sleep  in  Alcoholics 

Few  studies  have  used  polysomno- 
graphic  measures  to  evaluate  the  sleep- 
ing electroencephalogram  in  alcoholics 
(see  Zarcone  1978).  This  stands  in  sharp 
contrast  to  the  frequency  of  com- 
plaints of  sleep  disturbance  in  alco- 
hol-dependent individuals  observed 
in  general  medical  practice.  Various 
forms  of  hypersomnia,  parasomnia, 
disrupted  circadian  and  ultradian 
rhythms,  and  insomnia  have  all  been 
described  in  alcoholic  patients  (Wagman 
and  Allen  1975;  Gross  and  Hastey 
1976;  Wagman  et  al.  1977;  Zarcone 
1979;  Snyder  et  al.  1981;  Williams 
and  Rundell  1981;  Snyder  and  Karacan 
1985;  Ishibashi  et  al.  1987;  Gillin  et 
al.  1990).  In  1992,  a  meta-analysis 
evaluated  the  published  polygraphic 
sleep  studies  of  psychiatric  and  substance 
abuse  disorders  (Benca  et  al.  1992). 
In  that  meta-analysis  only  eight  stud- 
ies on  alcoholism  were  available  that 
met  criteria  for  inclusion  and  only 


418 


Alcohol  and  Sleep 


four  of  those  were  controlled  with 
normal  volunteers,  for  a  total  of  only 
123  subjects.  In  the  smaller  comparison 
with  matched  normal  control  sub- 
jects, sleep  latency  was  significantly 
prolonged,  stages  3  and  4  (delta) 
sleep  percentage  of  total  sleep  time 
(Delta%)  was  reduced,  and  REM% 
was  increased.  In  the  larger  compari- 
son with  normal  subjects  (20-50 
years  old),  alcoholics  had  significantiy 
reduced  total  sleep  time,  total  non- 
REM  sleep,  delta  sleep,  and  Delta% 
compared  with  normal  control  sub- 
jects; sleep  latency,  REM  latency  (the 
elapsed  time  from  sleep  onset  to  the 
first  REM  period),  and  REM  times 
were  within  normal  limits. 

In  comparison  with  studies  of 
affective  disorder  patients,  alcoholics 
were  found  to  have  significantly 
shorter  sleep  latency  and  longer  REM 
latency  but  did  not  differ  statistically 
on  total  sleep  time,  delta  sleep,  non- 
REM  sleep,  or  REM  sleep  time  (table 
1).  The  sleep  of  alcoholics  can  also  be 
differentiated  from  that  of  abstinent 
stimulant  abusers  (Thompson  et  al. 
1995).  Thus,  the  sleep  disturbance  in 


alcoholism  appears  to  be  somewhat 
distinct  from  the  syndrome  described 
in  affective  disorder  (Moeller  et  al. 
1993),  or  other  psychiatric  disorders 
and  may  arise  from  disruption  of  dif- 
ferent brain  mechanisms. 

This  typology  must  be  regarded  as 
tentative  until  it  is  replicated  in  addi- 
tional prospective  studies.  Further- 
more, formal  comparison  of  the 
alcoholic  groups  with  matched  normal 
control  subjects  would  be  highly 
desirable  to  establish  the  normal  limits 
for  each  of  the  measures. 

SLEEP  DISTURBANCE 
DURING  ALCOHOL 
WITHDRAWAL, 
ABSTINENCE, 
AND  RELAPSE 

Insomnia  is  perhaps  the  most  common 
complaint  in  the  alcoholic  patient  during 
acute  and  subacute  withdrawal  and 
may  be  a  key  variable  in  predicting 
relapse.  Sleep  is  often  markedly  disturbed 
during  the  first  days  of  abstinence  (Mello 
and  Mendelson  1970;  Allen  et  al. 
1971;  Kotorii  et  al.  1980),  even  in  the 


Table  1.  Sleep  Disturbance  in  Alcoholics  and  Depressed  Patients  Compared  With 
Normal  Subjects,  as  formulated  by  Gillin  et  al.  1990. 


Diagnosis 


RL 


REM 


RD 


Delta 


TST 


Major  depressive  disorder 
Alcohol  dependence 

Good  prognosis 

Poor  prognosis 
Alcohol  dependence  with 
alcohol-induced  mood  disorder 

Good  prognosis 

Poor  prognosis 


t 


t 


t 


Note:  RL  =  REM  latency;  REM  =  rapid  eye  movement  sleep;  RD  =  REM  density;  Delta  =  delta  sleep;  TST  =  total  sleep  time. 


419 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


absence  of  delirium  tremens  (DTs),  con- 
vulsions, or  other  major  withdrawal 
syndromes,  but  may  improve  about 
the  3d  or  4th  week  of  abstinence  (Gillin 
et  al.  1990).  Nevertheless,  some  recov- 
ered alcoholic  patients  show  reduced 
total  sleep  time,  loss  of  stages  3  and  4 
(delta)  sleep,  and  fragmented  sleep 
despite  prolonged  abstinence  (Johnson 
et  al.  1970;  Allen  et  al.  1971;  Williams 
and  Rundell  1981;  Snyder  and  Karacan 
1985;  Ishibashi  et  al.  1987).  Subjec- 
tively unrefreshing  sleep  or  specific 
sleep  abnormalities  (such  as  reduced 
delta  sleep)  during  chronic  abstinence 
have  been  associated  with  protracted  tol- 
erance or  increased  risk  of  renewed  drink- 
ing (Allen  and  Wagman  1975;  Wagman 
etal.  1978;  Allen  et  al.  1980). 

A  well-controlled  study  by  Gillin  and 
colleagues  (1994)  sheds  more  light  on 
the  mechanisms  underlying  disturbed 
sleep  in  withdrawal  and  abstinence  and 
its  relation  to  relapse.  Poly  graphic  sleep 
recordings  were  obtained  at  the  time  of 
admission  to  an  inpatient  alcohol  treat- 
ment program  and  evaluated  for  their 
predictive  value  in  determining  absti- 
nence and  relapse  3  months  following 
hospital  discharge.  Abstinence  and 
relapse  were  not  consistently  related  to 
any  clinical  measures  at  the  time  of 
hospital  admission,  including  age, 
duration  and  severity  of  alcoholism, 
marital  status,  employment,  hepatic 
enzyme  levels,  cognitive  performance, 
or  depression  ratings.  However,  short 
REM  latency,  increased  REM%,  and, 
possibly,  increased  REM  density  at 
the  time  of  admission  were  successful 
in  predicting  relapse  in  nondepressed 
alcohol-dependent  men  at  3  months 
following  hospital  discharge  (Gillin  et 


al.  1994).  Further  investigation  of  these 
patients  demonstrated  that  some  facets 
of  their  sleep  remained  abnormal  even 
after  27  months  of  abstinence  and  that 
insomnia  and  sleep  fragmentation  after 
approximately  5  months  of  abstinence 
may  be  related  to  relapse  by  14  months 
(Drummond  et  al  1998).  In  a  recent 
study  of  10  abstinent  and  11  relapsing 
depressed  alcoholics,  increased  REM 
density  at  admission  also  predicted 
relapse  at  3  months  (Clark  et  al.  1998). 
Other  studies  have  confirmed  that  sleep 
disturbance  is  an  important  predictor 
of  relapse.  In  a  study  of  74  alcoholic 
patients,  Brower  and  colleagues  (1998) 
found  that  relapsed  patients  did  not 
differ  from  abstinent  patients  at  baseline 
in  demographics  or  psychiatric  comor- 
bidity but  had  more  difficulty  falling 
asleep,  more  complaints  of  abnormal 
sleep,  and  longer  sleep  latencies  and 
less  stage  4  sleep  percentage  than 
abstinent  patients. 

POTENTIAL 
NEUROBIOLOGICAL 
MECHANISMS 
UNDERLYING  SLEEP 
DISTURBANCE 
IN  ALCOHOLICS 

The  neurobiological  mechanisms  under- 
lying disturbed  sleep  in  alcoholics 
remain  largely  unknown.  Based  on 
concepts  derived  from  the  neurobiology 
of  sleep  and  clinical  studies  (Hobson 
et  al.  1986),  considerable  evidence 
implicates  neurochemical  factors,  such 
as  a  relative  increase  in  cholinergic 
neurotransmission  (Gillin  et  al.  1979), 
which  may  underlie  short  REM 
latency  and  reduced  stages  3  and  4 


420 


Alcohol  and  Sleep 


sleep  seen  in  some  alcoholics.  With 
respect  to  short  REM  latency,  increased 
REM%,  and  increased  REM  density 
found  in  alcoholic  patients,  these  find- 
ings are  consistent  with  current  findings 
suggesting  that  serotonergic  neurons 
within  the  dorsal  raphe  suppress  REM 
sleep  by  inhibiting  cholinergic  neurons 
in  the  lateral  dorsal  tegmentum  and 
pedunculopontine  tegmental  nucleus 
(Luebke  et  al.  1992).  Data  from  normal 
volunteers  also  support  this  hypothesis, 
where  using  a  tryptophan-free  amino 
acid  drink  challenge,  which  depletes  the 
brain  of  serotonin,  has  been  found  to 
significantly  reduce  REM  latency  and 
increase  REM%  and  REM  density 
(Bhattietal.  1995). 

Sleep  disturbances  in  abstinent  alco- 
holics may  also  have  a  neurohormonal 
basis.  It  has  been  clearly  demonstrated 
in  animal  models  that  hypothalamic 
peptides  can  profoundly  affect  sleep 
quality.  Growth  hormone-releasing  fac- 
tor (GRF)  has  been  shown  to  increase 
slow  wave  sleep  in  several  species 
(Ehlers  et  al.  1986;  Wehrenberg  and 
Ehlers  1986;  Obal  et  al.  1988),  whereas 
corticotropin-releasing  factor  (CRF) 
produces  hyperarousal,  reductions  in 
slow  wave  sleep,  insomnia,  and  delays 
in  sleep  onset  (Ehlers  et  al.  1986, 
1997).  There  are  some  data  to  suggest 
similar  effects  of  these  peptides  on 
sleep  in  human  subjects  (Steiger  et  al. 
1992,  1994;  Friess  et  al.  1995). 
Whether  disruption  in  these  peptides 
leads  to  sleep  disturbance  in  alcoholics 
is  not  clear. 

There  are  also  data  to  suggest  that  the 
hypothalamic -pituitary- adrenal  system 
(HPA)  is  altered  in  some  alcoholics. 
Chronic  alcohol  ingestion  has  been 


demonstrated  to  produce  an  activating 
effect  on  the  HPA  system  in  both  clinical 
and  animal  studies  (see  Wand  1993  for 
a  review;  see  also  chapter  11  in  this 
monograph).  In  certain  chronic  alco- 
holics, hypercortisolemia  can  be  severe 
enough  so  that  several  features  of 
"pseudo-Cushing's  syndrome"  are 
present,  although  this  is  not  typical 
(Smals  et  al.  1976;  Rees  et  al.  1977). 
During  early  withdrawal,  nonsuppression 
of  plasma  Cortisol  to  dexamethasone 
has  been  reported  (Swartz  and  Dunner 
1982;  Kroll  et  al.  1983;  Newsom  and 
Murray  1983;  Porto  et  al.  1985),  as 
well  as  blunted  plasma  adrenocorti- 
cotropic hormone  (ACTH)  response  to 
metyrapone  blockade  (Wand  and  Dobs 
1991)  and  CRF  challenge  (Inder  et  al. 
1995).  Reduced  concentrations  of 
CRF  in  cerebrospinal  fluid  have  also 
been  reported  in  some  alcoholic  patients 
(Geracioti  et  al.  1994).  Cortisol  levels 
generally  return  to  normal  over  pro- 
longed abstinence,  but  stress  responses 
may  not  (Errico  et  al.  1993;  George 
et  al.  1994). 

Although  the  locus  of  disturbed 
HPA  functioning  in  alcoholics  is  not 
clear,  studies  by  Rivier  and  colleagues 
(1984,  1990)  suggest  that  regulation 
of  CRF  activity  may  be  important.  For 
instance,  in  animal  studies  chronic 
exposure  to  ethanol  vapors  decreased 
the  hypothalamic  content  and  increased 
the  synthesis  of  CRF.  Immunoneu- 
tralization  of  endogenous  CRF  has 
also  been  demonstrated  to  abolish 
ethanol-induced  release  of  ACTH  in 
rodents  (Rivier  et  al.  1984;  Zgombick 
and  Erwin  1987).  Whether  the 
changes  in  the  HPA  system  seen  in 
alcoholics  are  associated  with  specific 


421 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


sleep  difficulties  is  unclear,  because 
evaluation  of  the  relationship  between 
the  functioning  of  the  HPA  system 
and  polysomnographically  recorded 
sleep  in  alcoholic  patients  has  received 
little  attention. 

The  hypothalamic-pituitary-soma- 
totropic  system  (HPS)  may  also  be 
important  in  understanding  disturbed 
sleep  in  alcoholism.  It  has  been  suggested 
that  alteration  in  growth  hormone  secre- 
tion may  be  one  of  the  major  endocrine 
characteristics  of  alcoholism  (see  Badger 
et  al.  1993  for  a  review).  Growth  hor- 
mone is  normally  secreted  in  associa- 
tion with  delta  sleep  in  normal  control 
subjects,  at  least  up  until  middle  to 
old  age  (Carlson  et  al.  1972;  Holl  et 
al.  1991;  Van  Cauter  et  al.  1992).  Few 
sleep  studies  have  examined  nocturnal 
growth  hormone  secretion  during 
poly  graphically  recorded  sleep  in  alco- 
holics. Othmer  and  colleagues  (1982) 
found  that  growth  hormone  secretion 
was  generally  low  throughout  the 
night  and  was  dissociated  from  both 
sleep  onset  and  delta  sleep  in  chronic 
sober  alcoholics  (n  -  8)  compared 
with  normal  control  subjects  (n  =  3). 
Interestingly,  in  those  studies,  acute 
alcoholic  intoxication  increased  delta 
sleep  after  sleep  onset  but  did  not 
alter  the  pattern  of  growth  hormone 
secretion  in  alcoholic  patients. 

Prinz  and  colleagues  (1980)  previously 
reported  that  alcohol  reduced  nocturnal 
growth  hormone  secretion  in  normal 
volunteers.  GRF  secretion  has  also  been 
examined  in  early  abstinent  alcoholic 
patients  (De  Marinis  et  al.  1993).  Blunted 
growth  hormone  responses  to  clonidine 
but  not  GRF  were  found,  suggesting 
that  the  pituitary  response  to  GRF  is 


intact  in  abstinent  alcoholics;  however, 
hypothalamic  regulation  of  the  HPS 
system  may  be  disturbed.  In  another 
study,  growth  hormone  response  to  GRF 
was  intact  in  alcoholics;  growth  hormone 
response  to  sumatriptan  (5-HT1D  sero- 
tonergic receptor  agonist),  however, 
was  blunted  in  alcoholics  (Coiro  and 
Vescove  1995).  Blunted  growth  hor- 
mone responses  to  apomorphine  have 
also  been  associated  with  early  relapse 
(Heinz  et  al.  1995). 

Taken  together,  these  studies  suggest 
that  dysregulation  of  both  the  HPA  and 
HPS  systems  at  the  level  of  the  hypothal- 
amus may  occur  in  alcoholics  during 
abstinence.  Whether  this  dysregulation 
contributes  to  the  sleep  disturbance  that 
can  commonly  occur  at  that  time  is  still 
unknown,  because  of  the  paucity  of  stud- 
ies in  both  human  and  animal  models.  It 
is  possible  that  sleep  and  hormonal  dis- 
turbances in  alcoholism  are  linked  and 
that  a  common  mechanism  (serotoner- 
gic) may  be  responsible  for  dysregulation 
in  both.  Alternatively,  sleep  disturbance 
and  hormonal  disturbance  may  be  causally 
related  to  each  other  in  either  direction. 
Clearly,  further  studies  are  necessary  to 
describe  the  phenomenology  of  sleep  and 
endocrine  secretion  within  individuals 
in  order  to  begin  to  form  hypotheses 
regarding  causal  relationships. 

RELATIONSHIP  BETWEEN 
CIRCADIAN  RHYTHMS 
AND  SLEEP  DISTURBANCE 
IN  ALCOHOL  DEPENDENCE 
AND  ABSTINENCE 

Sleep  research  and  circadian  rhythm 
research  are  inextricably  linked,  because 
any  phenomenon  that  disrupts  circadian 


422 


Alcohol  and  Sleep 


functioning  (e.g.,  jet  lag)  will  disturb 
sleep.  Several  studies  have  demon- 
strated that  alcohol  can  inhibit  or 
alter  melatonin  secretion  (Ekman  et 
al.  1993;  Steindl  et  al.  1995),  a 
marker  of  the  circadian  systems  in 
humans.  Low  levels  of  melatonin 
have  also  been  reported  in  alcohol 
withdrawal  (Schmitz  et  al.  1996)  and 
abstinence  (Wetterberg  et  al.  1992). 
If  alcohol  disturbs  sleep  through  a 
circadian  rhythm  disturbance,  this 
suggests  that  specific  brain  mecha- 
nisms may  be  responsible.  Some  evi- 
dence that  this  may  be  true  is 
provided  in  a  study  by  Madeira  and 
colleagues  (1997).  They  reported 
that  rats  experiencing  long-term 
ethanol  treatment  (6  and  12  months) 
followed  by  long-term  abstinence  (6 
months)  had  significant,  irreversible 
depression  of  peptide  immunoreactivity 
and  mRNA  levels  in  the  suprachias- 
matic  nucleus.  Since  the  suprachias- 
matic  nucleus  is  considered  the  site  of 
the  biological  clock  in  rodents,  these 
data  suggest  that  disturbance  of  the 
biological  clock  is  a  potential  mecha- 
nism for  disturbed  sleep  in  long-term 
abstinent  alcoholics. 

GAPS  IN  KNOWLEDGE 
AND  PROMISING 
RESEARCH  AREAS 

Sleep  and  alcohol  is  clearly  an  under- 
studied area.  Although  progress  was 
made  in  the  1970s  defining  the  field, 
in  every  decade  since  fewer  investiga- 
tors remain  in  the  field.  Clearly,  an 
effort  needs  to  be  made  to  rekindle 
this  area,  especially  considering  that 
sleep  complaints  may  be  a  core  issue 


in  alcohol  relapse.  Can  newer  method- 
ologies shed  further  light  on  the 
hypothesis  that  chronic  alcohol  intake 
produces  specific  and  persistent 
changes  in  brain  function  that  lead  to 
prolonged  sleep  disturbance  and 
increased  risk  for  relapse? 

Although  previous  clinical  sleep 
studies  have  been  invaluable  in  defining 
the  sleep  disturbance  in  alcoholism,  they 
are  somewhat  limited.  Aside  from  the 
small  number  of  subjects  enrolled  in  the 
studies,  all  of  these  studies  relied  on 
evaluating  the  results  of  manual  scor- 
ing of  EEG  sleep  records.  None  of 
these  studies  used  any  quantitative 
measures  of  the  electroencephalogram, 
such  as  spectral  analysis  or  period/ 
amplitude  analysis.  Additional  studies 
using  quantitative  techniques  in  a 
larger  sample  should  be  able  to  better 
characterize  the  features  of  the  sleep 
electroencephalogram  that  distinctly 
characterize  alcohol  dependence  and 
potentially  link  that  disturbance  to 
underlying  neural  activity. 

Some  clinical  studies  support  the 
notion  that  chronic  alcohol  exposure 
causes  direct  toxic  or  neuroadaptive 
changes  in  the  brain  areas  that  are 
involved  in  the  regulation  of  sleep. 
For  instance,  cerebral  atrophy  and 
slow  wave  sleep  have  been  correlated 
in  abstinent  chronic  alcoholics 
(Ishibashi  et  al.  1987),  as  have  REM 
sleep  time  and  digit  span  impairment 
(Benson  et  al.  1978).  Further  studies 
using  magnetic  resonance  imaging  or 
single  photon  emission  computed 
tomography  imaging  techniques  in 
abstinent  alcoholics  in  combination 
with  better  quantification  of  waking 
and  sleeping  electroencephalograms 


423 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


are  likely  to  allow  for  a  better  testing 
of  this  hypothesis. 

Brain  Mechanisms  Underlying 
Alcohol- Induced 
Sleep  Disturbance 

There  have  been  very  few  animal  studies 
that  have  attempted  to  link  brain 
mechanisms  important  in  the  regulation 
of  sleep  and  the  long-term  effects  of 
alcohol.  Studies  need  to  be  conducted 
in  animals  that  are  exposed  to  alcohol  in 
clinically  relevant  amounts  over  long 
periods  of  time  and  then  studied  in 
acute  and  chronic  withdrawal,  as  well 
as  following  long-term  abstinence.  The 
specific  target  neurochemical  systems 
could  include  but  are  not  limited  to 
serotonin,  acetylcholine,  and  specific 
neuropeptides  such  as  GRF,  CRT,  and 
neuropeptide  Y.  Molecular  techniques 
using  such  markers  as  COX  (cyclo- 
oxygenase)  and  c-Fos  may  also  shed 
light  on  target  brain  areas. 

Risk  Factors  for  Sleep 
Disturbance  and  Alcohol  Use 

Evidence  from  family,  twin,  and  adop- 
tion studies  suggest  that  alcoholism  is  a 
genetically  influenced  disorder.  Poten- 
tially important  neurophysiological 
markers  of  alcoholism,  as  well  as  risk  for 
alcohol  dependence,  are  event-related 
potentials  and  EEG  measures.  Many, 
although  not  all  (Emmerson  et  al. 
1987),  studies  in  alcoholics  have  found 
that  their  resting,  waking  electroen- 
cephalogram was  deficient  in  alpha 
activity,  was  of  lower  voltage,  and 
tended  to  contain  theta  and  excessive 
fast  activity  (Varga  and  Nagy  1960; 
Begleiter  and  Platz  1972;  Naitoh  1973; 
Jones  and  Holmes  1976;  Coger  et  al. 


1978;  Propping  et  al.  1981;  Kaplan  et 
al.  1985;  Spehr  and  Stemmler  1985; 
Krauss  and  Niedermeyer  1991;  Pollock 
et  al.  1992).  More  recently  it  has  been 
suggested  that  EEG  fast  frequency 
activity  may  also  be  related  to  risk  of 
relapse  in  abstinent  alcoholics,  with 
abstinence -prone  patients  not  differing 
on  this  variable  from  control  subjects 
(Bauer  1994). 

Low-voltage  fast  EEG  patterns  are 
also  known  to  be  genetically  influenced 
and  appear  to  be  transmitted  by  an 
autosomal-dominant  mode  of  inheri- 
tance. Low- voltage  EEG  variants  have 
also  been  linked  to  risk  for  the  devel- 
opment of  alcoholism  (see  Enoch  et  al. 
1995;  Ehlers  et  al.  1999).  However,  no 
studies  have  determined  whether  certain 
EEG  sleep  patterns  (e.g.,  sleep  pheno- 
types)  actually  pre-date  the  development 
of  alcohol  dependence  and  thus  rep- 
resent preexisting  risk  factors  rather 
than  the  consequence  of  chronic  alco- 
hol exposure.  It  is  also  not  known 
whether  waking  EEG  phenotypes  are 
reflected  in  the  sleep  electroencepha- 
logram in  alcoholics,  and  specifically 
whether  waking  EEG  patterns  may  be 
predictive  of  sleep  disturbance.  If  so, 
this  would  suggest  that  a  common  set 
of  brain  mechanisms  may  influence 
the  patterns  of  both  the  waking  and 
sleeping  electroencephalograms  in 
alcohol-dependent  patients  and  those 
at  risk  for  alcohol  dependence. 

To  answer  this  set  of  questions, 
studies  need  to  be  conducted  in  subjects 
at  differing  genetic  risk  for  alcoholism. 
Sleep  studies  should  be  carried  out  in 
children  of  alcoholics  and  appropriate 
control  subjects.  In  addition,  prospective 
studies  should  be  conducted  addressing 


424 


Alcohol  and  Sleep 


the  question  of  whether  sleep  disturbance 
per  se  is  a  risk  factor  for  developing 
alcoholism.  In  this  light,  a  recent  study 
(Roehrs  et  al.  1999)  showed  that  in  the 
controlled  conditions  of  the  sleep  labor- 
atory insomniacs  were  more  likely  to 
select  a  disguised  alcohol  drink  before 
bedtime  than  control  subjects.  Adminis- 
tration of  alcohol  to  both  control  sub- 
jects and  insomniacs  was  found  to 
decrease  REM  sleep  in  both  groups  but 
to  increase  delta  sleep  selectively  in 
insomniacs.  These  data  suggest  that 
alcohol  may  be  more  reinforcing  for 
insomniacs  and  thus  may  be  potentially 
more  addicting. 

To  What  Extent  Is  Alcohol- 
Induced  Sleep  Disturbance 
Gender  Related? 

There  are  virtually  no  studies  that  have 
specifically  addressed  gender  issues  in 
sleep  and  alcohol.  The  studies  of  Aldrich 
and  colleagues  (1993)  are  provocative  in 
demonstrating  that  alcohol-related  sleep 
apnea  may  be  exclusively  a  male  phenom- 
enon. Recent  studies  in  large,  healthy 
subject  populations  have  demonstrated 
that  the  sleep  of  young  men  (30-40 
years  old)  deteriorates  at  a  much  faster 
rate  than  that  of  age-matched  women 
(see  Ehlers  and  Kupfer  1997).  These 
data  are  intriguing  in  view  of  the  later  age 
of  onset  of  alcoholism  in  many  women. 
Studies  evaluating  gender  issues  in 
sleep  and  alcohol  need  to  be  accom- 
plished in  both  humans  and  animals. 

Should  New  Therapies  Target 
Sleep  as  a  Core  Symptom 
in  Preventing  Relapse? 

If  the  neuroadaptive  changes  associated 
with  the  protracted  abstinence  syndrome 


include  sleep  and  circadian  rhythm  dis- 
turbance, then  some  novel  treatment 
modalities  may  be  useful  in  preventing 
relapse.  In  an  animal  study,  stimulation 
of  GABAA  receptors  by  muscimol  or 
homotaurine  administration  during 
alcohol  withdrawal  was  found  to  sig- 
nificantly improve  the  disturbances  in 
the  sleep-wake  states  in  the  alcohol- 
dependent  rats,  in  a  time-related  manner 
(Rouhani  et  al.  1998).  Thus,  GAJBAergic 
drugs  might  be  specifically  targeted  to 
treat  sleep  disturbance  during  acute 
withdrawal.  One  study  investigated 
sleep  in  abstinent  alcoholics  during 
ritanserin  treatment  (Monti  et  al.  1993). 
In  that  study  mood  and  sleep  were 
improved  in  the  treated  group;  however, 
no  information  was  given  on  long-term 
abstinence,  drug  response,  and  sleep 
quality.  In  another  study  the  efficacy 
of  L-tryptophan  was  evaluated  and 
found  to  be  useful  in  improving  sleep 
and  mood  in  alcoholics  (Asheychik  et 
al.  1989). 

In  addition  to  pharmacotherapy, 
interventions  aimed  at  regulating  cir- 
cadian functioning  may  also  be  useful 
in  treating  sleep  disturbance  during 
abstinence.  In  one  study,  bright  light 
exposure  was  evaluated  in  patients  dur- 
ing alcohol  withdrawal.  Subjective  mea- 
sures of  sleep  quality,  sleep  maintenance, 
and  sleep  architecture  were  all  found 
to  be  improved  after  bright  light  therapy 
(Schmitz  et  al.  1997),  but  the  study  was 
not  placebo  controlled.  Dawn  simula- 
tion (early  morning  light  treatment 
simulating  dawn)  has  also  been  used 
to  treat  abstinent  alcoholics  with  winter 
depression,  and  has  been  found  useful 
(Avery  et  al.  1998).  Further  therapeutic 
trials  that  include  sleep  measurements, 


425 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


relapse  rates,  and  psychiatric  states  are 
clearly  needed. 

REFERENCES 

Aldrich,  M.S.,  and  Shipley,  J.E.  Alcohol 
use  and  periodic  limb  movements  of  sleep. 
Alcohol  Clin  Exp  Res  17:192-196,  1993. 

Aldrich,  M.S.;  Shipley,  J.E.;  Tandon,  R; 
Kroll,  P.D.;  and  Brower,  K.J.  Sleep-disor- 
dered breathing  in  alcoholics:  Association 
with  age.  Alcohol  Clin  Exp  Res  17:1179- 
1183,  1993. 

Allen,  R.P.,  and  Wagman,  A.M.  Do  sleep 
patterns  relate  to  the  desire  for  alcohol? 
Adv  Exp  Med  Biol  59:495-508,  1975. 

Allen,  R.P.;  Wagman,  A.;  Faillace,  LA.; 
and  Macintosh,  M.  Electroencephalographic 
(EEG)  sleep  recovery  following  prolonged 
alcohol  intoxication  in  alcoholics.  J  Nerv 
MentDis  153 :424^33,  1971. 

Allen,  R.P.;  Wagman,  A.M.;  and 
Funderburk,  F.R.  Slow  wave  sleep 
changes:  Alcohol  tolerance  and  treatment 
implications.  Adv  Exp  Med  Biol 
85A:629-640,  1977. 

Allen,  R.P.;  Wagman,  A.M.;  Funderburk, 
F.R.;  and  Wells,  D.T.  Slow  wave  sleep:  A 
predictor  of  individual  differences  in 
response  to  drinking?  Biol  Psychiatry 
15:345-348,  1980. 

Asheychik,  R.;  Jackson,  T.;  Baker,  PL; 
Ferraro,  R;  Ashton,  T.;  and  Kilgore,  J. 
The  efficacy  of  L- tryptophan  in  the  reduc- 
tion of  sleep  disturbance  and  depressive 
state  in  alcoholic  patients.  J  Stud  Alcohol 
50:525-532,  1989. 

Avery,  D.H.;  Bolte,  MA.;  and  Ries,  R 
Dawn  simulation  treatment  of  abstinent 
alcoholics  with  winter  depression.  /  Clin 
Psychiatry  59:36-42,  1998. 


Badger,  T.M.;  Ronis,  M.J.;  and  Lumpkin, 
C.K.,  Jr.  The  effects  of  alcohol  on  growth 
hormone  and  growth  hormone-regulated 
systems.  In:  Zakhari,  S.,  ed.  Alcohol  and 
the  Endocrine  System.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  23.  NIH  Pub. 
No.  93-3533.  Bethesda,  MD:  the 
Institute,  1993.  pp.  339-361. 

Bauer,  L.O.  Electroencephalographic  and 
autonomic  predictors  of  relapse  in  alco- 
hol-dependent patients.  Alcohol  Clin  Exp 
Res  18:755-760,  1994. 

Begleiter,  PL,  and  Platz,  A.  The  effects  of 
alcohol  on  the  central  nervous  system  in 
humans.  In:  Kissin,  B.,  and  Begleiter,  PL, 
eds.  The  Biology  of  Alcoholism.  Vol.  11. 
New  York:  Plenum  Press,  1972.  pp. 
293-343. 

Benca,  R.M.;  Obermeyer,  W.H.;  Thisted, 
R.A.;  and  Gillin,  J.C.  Sleep  and  psychi- 
atric disorders:  A  meta-analysis.  Arch  Gen 
Psychiatry  49:651-668,  1992. 

Benson,  K.;  Cohen,  M.;  and  Zarcone,  V., 
Jr.  REM  sleep  time  and  digit  span  impair- 
ment in  alcoholics.  /  Stud  Alcohol 
39:1488-1498,  1978. 

Bhatti,  T.;  Gillin,  J.C.;  Golshan,  S.;  Clark, 
C;  Demodena,  A.;  Scholosser,  A.;  et  al. 
The  effect  of  a  tryptophan-free  amino  acid 
drink  on  sleep  and  mood  in  normal  con- 
trols. Sleep  Res  24:153  (abstract),  1995. 

Block,  A. J.;  Hellard,  D.W.;  and  Slayton, 
P.C.  Effect  of  alcohol  ingestion  on 
breathing  and  oxygenation  during  sleep. 
Analysis  of  the  influence  of  age  and  sex. 
Am  J  Med  80:595-600,  1986. 

Bonora,  M.;  Shields,  G.I.;  Knuth,  S.L.; 
Bartlett,  D.,  Jr.;  and  St.  John,  W.M. 
Selective  depression  by  ethanol  of  upper 
airway  respiratory  motor  activity  in  cats. 
Am  Rev  Respir  Dis  130:156-161,  1984. 


426 


Alcohol  and  Sleep 


Brower,  K.J.;  Aldrich,  M.S.;  and  Hall, 
J.M.  Polysomnographic  and  subjective 
sleep  predictors  of  alcoholic  relapse.  Alcohol 
Clin  Exp  Res  22{8):I864-I87l,  1998. 

Carlson,  H.E.;  Gillin,  J.C.;  Gorden,  P.; 
and  Snyder,  F.  Absence  of  sleep-related 
growth  hormone  peaks  in  aged  normal 
subjects  and  in  acromegaly.  /  Clin 
Endocrinol  Metab  34: 1 1 02-1 1 05 ,  1972 . 

Clark,  C.P.;  Gillin,  J.C.;  Golshan,  S.; 
Demodena,  A.;  Smith,  T.L.;  Danowski, 
S.;  Irwin,  M.;  and  Schuckit,  M.  Increased 
REM  sleep  density  at  admission  predicts 
relapse  by  three  months  in  primary  alco- 
holics with  a  lifetime  diagnosis  of  sec 
ondary  depression.  Biol  Psychiatry  43:601- 
607,  Apr  15,  1998. 

Coger,  R.W.;  Dymond,  A.M.;  Serafetinides, 
E.A.;  Lowenstam,  I.;  and  Pearson,  D.  EEG 
signs  of  brain  impairment  in  alcoholism. 
Biol  Psychiatry  13:729-739,  1978. 

Coiro,  V.,  and  Vescov,  P.P.  Alcoholism 
abolishes  the  growth  hormone  response 
to  sumatriptan  administration  in  man. 
Metabolism  44:1577-1580,  1995. 

Dawson,  A.;  Lehr,  P.;  Bigby,  B.G.;  and 
Mitler,  M.M.  Effect  of  bedtime  ethanol 
on  total  inspiratory  resistance  and  respira- 
tory drive  in  normal  nonsnoring  men. 
Alcohol  Clin  Exp  Res  17:257-262,  1993. 

Dawson,  A.;  Bigby,  B.G.;  Poceta,  J.S.;  and 
Mitler,  M.M.  Effect  of  bedtime  alcohol 
on  inspiratory  resistance  and  respiratory 
drive  in  snoring  and  nonsnoring  men. 
Alcohol  Clin  Exp  Res  21:183-190,  1997. 

De  Marinis,  L.;  Mancini,  A.;  Fiumara,  C; 
Conte,  G.;  Iacona,  T.;  Ianiri,  L.;  Zeppetelli, 
E.;  Gobbi,  G.;  and  Tempesta,  E.  Growth 
hormone  response  to  growth  hormone- 
releasing  hormone  in  early  abstinent  alco- 
holic patients.  Psychoneuroendocrinology 
18:475-483,  1993. 


Drummond,  S.P.A.;  Gillin,  J.C.;  Smith, 
T.L.;  and  Demodena,  A.  The  sleep  of 
abstinent  pure  primary  alcoholic  patients: 
Natural  course  and  relationship  to  relapse. 
Alcohol  Clin  Exp  Res 22:1796-1802,  1998. 

Ehlers,  C.L.,  and  Kupfer,  D.J.  Slow  wave 
sleep:  Do  young  adult  men  and  women  age 
differently?  / Sleep  Res  6:2 1 1-2 1 5,  1997. 

Ehlers,  C.L.;  Reed,  T.K.;  and  Henriksen, 
S.J.  Effects  of  corticotropin  releasing 
factor  and  growth  hormone-releasing 
factor  on  sleep  and  activity  in  rats. 
Neuroendocrinology  42:467^174,  1986. 

Ehlers,  C.L.;  Somes,  C;  Seifritz,  E.;  and 
Rivier,  J.E.  CRF/NPY  interactions:  A 
potential  role  in  sleep  dysregulation  in 
depression  and  anxiety.  Depress  Anxiety 
6:1-9,  1997. 

Ehlers,  C.L.;  Garcia- Andrade,  C;  Wall, 
T.L.;  Cloutier,  C;  and  Phillips,  E. 
Encephalographic  responses  to  alcohol 
challenge  in  Native  American  Mission 
Indians.  Biol  Psychiatry  45:776-787, 1999. 

Ekman,  A.C.;  Leppaluoto,  J.;  Huttunen, 
P.;  Aranko,  K.;  and  Vakkuri,  O.  Ethanol 
inhibits  melatonin  secretion  in  healthy 
volunteers  in  a  dose-dependent  random- 
ized double  blind  cross-over  study.  /  Clin 
Endocrinol  Metab  77:780-783,  1993. 

Emmerson,  R.Y.;  Dustman,  R.E.; 
Shearer,  D.E.;  and  Chamberlin,  H.M. 
EEG,  visually  evoked  and  event  related 
potentials  in  young  abstinent  alcoholics. 
Alcohol  4:241-248,  1987. 

Enoch,  M.A.;  Rohrbaugh,  J.W.;  Davis, 
E.Z.;  Harris,  C.R.;  Ellingson,  R.J.; 
Andreason,  P.;  Moore,  V.;  Varner,  J.L.; 
Brown,  G.L.;  Eckardt,  M.J.;  et  al.  Relation- 
ship of  genetically  transmitted  alpha  EEG 
traits  to  anxiety  disorders  and  alcoholism. 
Am  J  Med  Genet  60:400-408,  1995. 


427 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Errico,  A.L.;  Parsons,  O  A.;  King,  A.C.; 
and  Lovallo,  W.R.  Attenuated  Cortisol 
response  to  biobehavioral  stressors  in  sober 
alcoholics.  J  Stud  Alcohol  54:393-398, 1993. 

Friess,  E.;  Wiedemann,  K.;  Steiger,  A.;  and 
Holsboer,  F.  The  hypothalamic-pituitary- 
adrenocortical  system  and  sleep  in  man. 
Adv  Neuroimmunol  5:111-125,  1995. 

George,  D.T.;  Lindquist,  T.;  Alim,  T.;  Flood, 
M.;  Eckardt,  M.J.;  and  Linnoila,  M. 
Abstinent  alcoholics  exhibit  an  exaggerated 
stress  response  to  2-deoxy-D-glucose 
challenge.  Alcohol  Clin  Exp  Res 
18:685-691,  1994. 

Geracioti,  T.D.,  Jr.;  Loosen,  P.T.;  Ebert, 
M.H.;  Ekhator,  N.N.;  Burns,  D.; 
Nicholson,  W.E.;  and  Orth,  D.N. 
Concentrations  of  corticotropin-releasing 
hormone,  norepinephrine,  MHPG,  5- 
hydroxyindoleacetic  acid,  and  tryptophan 
in  the  cerebrospinal  fluid  of  alcoholic 
patients:  Serial  sampling  studies. 
Neuroendocrinology  60:635-642,  1994. 

Gillin,  J.C.;  Sitaram,  N.;  and  Duncan,  W.C. 
Muscarinic  supersensitivity:  A  possible  model 
for  the  sleep  disturbance  of  primary  depres- 
sion? Psychiatry  Res  1:17-22,  July  1979. 

Gillin,  J.C.;  Smith,  T.L.;  Irwin,  M.;  Kripke, 
D.F.;  and  Schuckit,  M.  EEG  sleep  studies 
in  "pure"  primary  alcoholism  during  suba- 
cute withdrawal:  Relationships  to  normal 
controls,  age,  and  other  clinical  variables. 
Biol  Psychiatry  27:447-^88,  1990. 

Gillin,  J.C.;  Smith,  T.L.;  Irwin,  M.;  Butters, 
N.;  Demodena,  A.;  and  Schuckit,  M. 
Increased  pressure  for  rapid  eye  movement 
sleep  at  time  of  hospital  admission  predicts 
relapse  in  nondepressed  patients  with 
primary  alcoholism  at  3-month  follow-up. 
Arch  Gen  Psychiatry  51:189-197, 1994. 

Gresham,  S.C.;  Webb,  W.B.;  and  Williams, 
RL.  Alcohol  and  caffeine:  Effect  on  in- 


ferred visual  dreaming.  Science 
140:1226-1227,  1963. 

Gross,  M.M.,  and  Hastey,  J.M.  Sleep 
disturbances  in  alcoholism.  In:  Tarter, 
RE.,  and  Sugarman,  A.,  eds.  Alcoholism: 
Interdisciplinary  Approaches  to  an 
Enduring  Problem.  Addison-Wesley, 
1976.  pp.  257-309. 

Heinz,  A.;  Dettling,  M.;  Kuhn,  S.;  Dufeu, 
P.;  Graf,  K.J.;  Kurten,  I.;  Rommelspacher, 
H.;  and  Schmidt,  I.G.  Blunted  growth 
hormone  response  is  associated  with  early 
relapse  in  alcohol-dependent  patients. 
Alcohol  Clin  Exp  Res  19:62-65,  1995. 

Hobson,  J.A.;  Lydic,  R;  and  Baghdoyan, 
HA.  Evolving  concepts  of  sleep  cycle 
generation:  From  brain  centers  to  neu- 
ronal populations.  Behav  Brain  Sci 
9:371-448,  1986. 

Holl,  R.W.;  Harman,  M.L.;  Veldhuis, 
J.D;  Taylor,  W.M.;  and  Thorner,  M.O. 
Thirty- second  sampling  of  plasma  growth 
hormone  in  man:  Correlation  with  sleep 
stages.  /  Clin  Endocrinol  Metab  72:854- 
861,  1991. 

Inder,  W.J.;  Joyce,  P.R;  Ellis,  M.J.; 
Evans,  M.H.;  Livesey,  J.H.;  and  Donald, 
R.  A.  The  effects  of  alcoholism  on  the 
hypothalamic-pituitary-adrenal  axis.  Clin 
Endocrinol  43:283-290,  1995. 

Ishibashi,  M.;  Nakazawa,  Y.;  Yokoyama, 
T.;  Koga,  Y.;  Miyahara,  Y.;  Hayashida, 
N.;  and  Ohse,  K.  Cerebral  atrophy  and 
slow  wave  sleep  of  abstinent  chronic 
alcoholics.  Drug  Alcohol  Depend 
19:325-332,  1987. 

Issa,  F.Q.,  and  Sullivan,  C.E.  Alcohol, 
snoring  and  sleep  apnoea.  /  Neurol 
Neurosurgr  Psychiatry  45:93-99,  1982. 

Johnson,  L.C.;  Burdick,  J.A.;  and  Smith,  J. 
Sleep  during  alcohol  intake  and  withdrawal 


428 


Alcohol  and  Sleep 


in  the  chronic  alcoholic.  Arch  Gen  Psychiatry 
22:406-418,  1970. 

Johnstone,  R.E.,  and  Reier,  C.E.  Acute 
respiratory  effects  of  ethanol  in  man.  Clin 
Pharmacol  Ther  14:501-508,  1973. 

Jones,  F.W.,  and  Holmes,  D.S.  Alcoholism, 
alpha  production  and  biofeedback.  / 
Consult  Clin  Psychol 44:224-228,  1976. 

Kaplan,  D.T.;  Glueck,  B.C.;  Hesselbrock, 
M.N.;  and  Reed,  H.B.,  Jr.  Power  and 
coherence  analysis  of  the  EEG  in  hospital- 
ized alcoholics  and  nonalcoholic  controls. 
J  Stud  Alcohol  46: 122-127,  1985. 

Knowles,  J.B.;  Laverty,  S.G.;  and  Kuechler, 
HA.  Effect  of  alcohol  on  REM  sleep.  QJ 
Stud  Alcohol  29 :342-349,  1968. 

Kotorii,  T.;  Nakazawa,  Y.;  Yokoyama,  T.; 
Kurauchi,  H.;  Sakurada,  H.;  Ohkawa,  T.; 
Nonaka,  K.;  Hasuzawa,  H.;  Dainoson,  K.; 
and  Inanaga,  K.  The  sleep  pattern  of 
chronic  alcoholics  during  the  alcohol 
withdrawal  period.  Folia  Psychiatr  Neurol 
7^34:89-95,1980. 

Krauss,  G.L.,  and  Niedermeyer,  E. 
Electroencephalogram  and  seizures  in 
chronic  alcoholism.  Electroencephalogr 
Clin  Neurophysiol  78:97-104,  1991. 

Krol,  R.C.;  Knuth,  S.L.;  and  Bartlett,  D., 
Jr.  Selective  reduction  of  genioglossal 
muscle  activity  by  alcohol  in  norman 
human  subjects.  Am  Rev  Respir  Dis 
129:247-250,  1984. 

Kroll,  P.;  Palmer,  C;  and  Greden,  J.F. 
The  dexamethasone  suppression  testing  in 
patients  with  alcoholism.  Biol  Psychiatry 
18:441-^50,  1983. 

Landolt,  H.P.;  Roth,  C.;  Dijk,  D.J.;  and 
Borbely,  AA.  Late -afternoon  ethanol 
intake  affects  nocturnal  sleep  and  the 
sleep  EEG  in  middle-aged  men.  J  Clin 
Psychopharmacol  16:428^136,  1996. 


Le  Bon,  O.;  Verbanck,  P.;  Hoffmann,  G.; 
Murphy,  J.R.;  Staner,  L.;  De  Groote,  D.; 
Mampunza,  S.;  Den  Dulk,  A.;  Vacher,  C; 
Kornreich,  C.;  et  al.  Sleep  in  detoxified 
alcoholics:  Impairment  of  most  standard 
sleep  parameters  and  increased  risk  for  sleep 
apnea,  but  not  for  myoclonias — a  controlled 
study.  J  Stud  Alcohol  58:30-36,  1997. 

Lobo,  L.L.,  and  Tufik,  S.  Effects  of  alco- 
hol on  sleep  parameters  of  sleep-deprived 
healthy  volunteers.  Sleep  20:52-59,  1997. 

Luebke,  J.I.;  Greene,  R.W.;  Semba,  K.; 
Kamondi,  A.;  McCarley,  R.W.;  and  Reiner, 
P.B.  Serotonin  hyperpolarizes  cholinergic 
low-threshold  burst  neurons  in  the  rat  lat- 
erodorsal  tegmental  nucleus  in  vitro.  Proc 
Natl  Acad  Sci  USA  89:743-747,  1992. 

MacLean,  A.W.,  and  Cairns,  J.  Dose-response 
effects  of  ethanol  on  the  sleep  of  young 
men.  J  Stud  Alcohol  43:434^44, 1982. 

Madeira,  M.D.;  Andrade,  J. P.;  Lieberman, 
A.R;  Sousa,  N.;  Almeida,  O.F.;  and  Paula- 
Barbosa,  M.M.  Chronic  alcohol  consump- 
tion and  withdrawal  do  not  induce  cell 
death  in  the  suprachiasmatic  nucleus,  but 
lead  to  irreversible  depression  of  peptide 
immunoreactivity  and  mRNA  levels.  / 
Neurosci  17:1302-1319,  1997. 

Mamdani,  M.;  Hollyfield,  R;  Ravi,  S.D.; 
Dorun,  W.;  and  Borge,  G.F.  Prevalence 
of  sleep  apnea  among  abstinent  chronic 
alcoholic  men.  Sleep  Res  18:349,  1989. 

Mello,  N.K.,  and  Mendelson,  J.H. 
Behavioral  studies  of  sleep  patterns  in 
alcoholics  during  intoxication  and  with- 
drawal. J  Pharmacol  Exp  Ther  175:94- 
112,1970. 

Mitler,  M.M.;  Dawson,  A.;  Henriksen, 
S.J.;  Sobers,  M.;  and  Bloom,  F.E.  Bedtime 
ethanol  increases  resistance  of  upper 
airways  and  produces  sleep  apneas  in 


429 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


asymptomatic  snore rs.  Alcohol  Clin  Exp 
Res  12:801-805,  1988. 

Moeller,  F.G.;  Gillin,  J.C.;  Irwin,  M.; 
Golshan,  S.;  Kripke,  D.F.;  and  Schuckit, 
M.A.  A  comparison  of  sleep  EEGs  in 
patients  with  primary  major  depression 
and  major  depression  secondary  to  alco- 
holism. J  Affect  Disord  27:39^2,  1993. 

Monti,  J.M.;  Alterwain,  P.;  Estevez,  F.; 
Alvarino,  F.;  Giusti,  M.;  Olivera,  S.;  and 
Labraga,  P.  The  effects  of  ritanserin  on 
mood  and  sleep  in  abstinent  alcoholic 
patients.  Sleep  16:647-654,  1993. 

Naitoh,  P.  The  value  of  electroencephalo- 
graphy in  alcoholism.  Ann  NT  Acad  Sci 
215:303-320,  1973. 

Newsom,  G.,  and  Murray,  N.  Reversal  of 
dexamethasone  suppression  test: 
Nonsuppression  in  alcohol  abusers.  Am  J 
Psychiatry  140:353-354,  1983. 

Obal,  F.,  Jr.;  Alfoldi,  P.;  Cady,  A.B.; 
Johannsen,  L.;  Sary,  G.;  and  Krueger, 
J.M.  Growth  hormone-releasing  factor 
enhances  sleep  in  rats  and  rabbits.  Am  J 
P^«o/255:R310-R316,  1988. 

Othmer,  E.;  Daughaday,  W.H.;  Goodwin, 
D.W.;  Levine,  W.R.;  Malarkey,  W.B.; 
Freemon,  F.;  et  al.  Sleep  and  growth 
hormone  secretion  in  alcoholics.  /  Clin 
Psychiatry  43:411-414,  1982. 

Pollock,  V.;  Schneider,  L.S.;  Zemansky, 
M.F.;  Gleason,  R.P.;  and  Pawluczyk,  S. 
Topographic  quantitative  EEG  amplitude 
in  recovered  alcoholics.  Psychiatry  Res 
~Neuroimagin£j  45:25-32,  1992. 

Porto,  J.A.;  Monteiro,  M.G.;  Laranjeira, 
R.R.;  Jorge,  M.R.;  and  Masur,  J.  Reversal 
of  abnormal  dexamethasone  suppression 
test  in  alcoholics  abstinent  for  4  weeks. 
Biol  Psychiatry  20: 1 156-1 160,  1985 . 


Prinz,  P.N.;  Roehrs,  T.A.;  Vitaliano,  P.P.; 
Linnoila,  M.;  and  Weitzmann,  E.D.  Effect 
of  alcohol  on  sleep  and  nighttime  plasma 
growth  hormone  and  Cortisol  concentra- 
tions. /  Clin  Endocrinol  Metab  5 1 :759- 
764,  1980. 

Propping,  P.;  Kurger,  J.;  and  Mark,  N. 
Genetic  disposition  to  alcoholism:  An  EEG 
study  in  alcoholics  and  their  relatives.  Hum 
Genet  59:51-59, 1981. 

Rains,  V.S.;  Ditzler,  T.F.;  Newsom,  R.D.; 
Lee-Gushi,  S.;  and  Morgan,  E.J.  Alcohol 
and  sleep  apnea.  Hawaii  Med  J  50:282- 
287,  1991. 

Rees,  L.H.;  Besser,  G.M.;  Jeffcoate,  W.F.; 
Goldie,  D.J.;  and  Marks,  V.  Alcohol-induced 
pseudo-Cushing's  syndrome.  Lancet 
1:726-728,1977. 

Rivier,  C.;  Bruhn,  T.;  and  Vale,  W.  Effect 
of  ethanol  on  the  hypothalamic-pituitary- 
adrenal  axis  in  the  rat:  Role  of 
corticotropin-releasing  factor  (CRF).  / 
Pharmacol  Exp  Ther  229:127-131,  1984. 

Rivier,  C.;  Imaki,  T.;  and  Vale,  W. 
Prolonged  exposure  to  alcohol:  Effect  on 
CRF  mRNA  levels,  and  CRF-  and  stress- 
induced  ACTH  secretion  in  the  rat.  Brain 
Res  520:1-5,  1990. 

Roehrs,  T.;  Papineau,  K;  Rosenthal,  L.;  and 
Roth,  T.  Ethanol  as  a  hypnotic  in  insom- 
niacs: Self  administration  and  effects  on 
sleep  and  mood.  Neuropsychopharmacolojuy 
20:279-286,  1999. 

Rouhani,  S.;  DalFAva-Santucci,  J.;  Bajenaru, 
O.;  Emmanouilidis,  E.;  Tran,  G.;  Manicom, 
R;  Dinh-Xuan,  A.T.;  and  Poenaru,  S. 
Effects  of  muscimol  or  homotaurine  on 
sleep-wake  states  in  alcohol-dependent 
rats  during  withdrawal.  Pharmacol 
Biochem  Behav  59:955-960,  Apr  1998. 

Rundell,  O.H.;  Lester,  B.K.;  Griffiths, 
W.S.;  and  Williams,  H.L.  Alcohol  and 


430 


Alcohol  and  Sleep 


sleep  in  young  adults.  Psychopharmacolqgia 
26:201-218,  1972. 

Schmitz,  M.M.;  Sepandj,  A.;  Pichler, 
P.M.;  and  Rudas,  S.  Disrupted  melatonin- 
secretion  during  alcohol  withdrawal.  Prog 
Neuropsychopharmacol  Biol  Psychiatry 
20:983-985,  1996. 

Schmitz,  M.;  Frey,  R.;  Pichler,  P.;  Ropke, 
H.;  Anderer,  P.;  Saletu,  B.;  and  Rudas,  S. 
Sleep  quality  during  alcohol  withdrawal 
with  bright  light  therapy.  Prog  'Neuro- 
psychopharmacol Biol  Psychiatry 
21:965-977,  1997. 

Scrima,  L.;  Broudy,  M.;  Nay,  K.N.;  and 
Cohn,  MA.  Increased  severity  of  obstruc- 
tive sleep  apnea  after  bedtime  alcohol 
ingestion:  Diagnostic  potential  and  pro- 
posed mechanism  of  action.  Sleep 
5:318-328,  1982. 

Scrima,  L.;  Hartman,  P.G.;  and  Hiller, 
F.C.  Effect  of  three  alcohol  doses  on 
breathing  during  sleep  in  30-49  year  old 
nonobese  snorers  and  nonsnorers.  Alcohol 
Clin  Exp  Res  13:420^27,  1989. 

Smals,  A.G.;  Kloppenborg,  P.W.;  Njo, 
K.T.;  Knoben,  J.M.;  and  Ruland,  CM. 
Alcohol-induced  Cushingoid  syndrome. 
Br  Med  J  2:1298,  1976. 

Snyder,  S.,  and  Karacan,  I.  Sleep  patterns 
of  sober  chronic  alcoholics.  Neuro- 
psychology 13:97-100,  1985. 

Snyder,  S.;  Karacan,  I.;  and  Salis,  P.J. 
Effects  of  disulfiram  on  the  sleep  of 
chronic  alcoholics.  In:  Galanter,  M.,  ed. 
Currents  in  Alcoholism:  Recent  Advances 
in  Research  and  Treatment.  Vol.  VIII. 
New  York:  Grune  &  Stratton,  1981.  pp. 
159-166. 

Spehr,  W.,  and  Stemmler,  G. 
Postalcoholic  diseases:  Diagnostic  rele- 
vance of  computerized  EEG. 


Electroencepbalogr  Clin  Neurophysiol 
60:106-114,1985. 

Steiger,  A.;  Guldner,  J.;  Hemmeter,  U.; 
Rothe,  B.;  Wiedemann,  K.;  and  Holsboer, 
F.  Effects  of  growth  hormone-releasing 
hormone  and  somatostatin  on  sleep  EEG 
and  nocturnal  hormone  secretion  in  male 
controls.  Neuroendocrinology  56:566- 
573,  1992. 

Steiger,  A.;  Guldner,  J.;  Colla-Muller,  M.; 
Friess,  E.;  Sonntag,  A.;  and  Schier,  T. 
Growth  hormone -releasing  hormone 
(GHRH)-induced  effects  on  sleep  EEG 
and  nocturnal  secretion  of  growth  hor- 
mone, Cortisol  and  ACTH  in  patients 
with  major  depression.  J  Psychiatr  Res 
28:225-238,  1994. 

Steindl,  P.E.;  Finn,  B.;  Bendok,  B.;  Rothke, 
S.;  Zee,  P.C.;  and  Blei,  A.T.  Disruption  of 
the  diurnal  rhythm  of  plasma  melatonin  in 
cirrhosis.  [See  comments.]  Ann  Intern  Med 
123:274-277,  1995. 

Swartz,  CM.,  and  Dunner,  F.J.  Dexameth- 
asone  suppression  test:  Nonsuppression  in 
alcohol  abusers.  Am  J  Psychiatry  39:1309- 
1312, 1982. 

Taasan,  V.C;  Block,  A.J.;  Boysen,  P.G.; 
and  Wynne,  J.W.  Alcohol  increases  sleep 
apnea  and  oxygen  desaturation  in  asymp- 
tomatic men.  Am  J  Med  71:240-245, 1981. 

Tan,  E.T.;  Lambie,  D.G.;  Johnson,  R.H.; 
Robinson,  B.J.;  and  Whiteside,  EA.  Sleep 
apnoea  in  alcoholic  patients  after  with- 
drawal. ClinSci  69:655-661,  1985. 

Teschler,  H.;  Berthon-Jones,  M.; 
Wessendorf,  T.;  Meyer,  H.J.;  and 
Konietzko,  N.  Influence  of  moderate 
alcohol  consumption  on  obstructive  sleep 
apnoea  with  and  without  AutoSet  nasal 
CPAP  therapy.  Eur  Respir  J  9:2371- 
2377,  1996. 


431 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Thompson,  P.M.;  Gillin,  J.C.;  Golshan,  S.; 
and  Irwin,  M.  Polygraphic  sleep  measures 
differentiate  alcoholics  and  stimulant  abusers 
during  short-term  abstinence.  Biol  Psychiatry 
38:831-836, 1995. 

Van  Cauter,  E.;  Kerkhos,  M.;  Caufriez, 
A.;  Van  Onderbergen,  A.;  Thorner, 
M.O.;  and  Copinschi,  G.  A  quantitative 
estimation  of  growth  hormone  secretion 
in  normal  man:  Reproducibility  and 
relation  to  sleep  and  time  of  day. 
/  Clin  Endocrinol  Metab  74: 1441- 
1450, 1992. 

Varga,  B.,  and  Nagy,  T.  Analysis  of  alpha 
rhythms  in  the  electroencephalogram  of 
alcoholics.  Electroencephalogr  Clin 
Neurophysiol  12:933,  1960. 

Vitiello,  M.V.;  Prinz,  P.N.;  Personius, 
J. P.;  Vitaliano,  P.P.;  Nuccio,  M.A.; 
Ries,  R.K.;  and  Koerker,  R  History 
of  chronic  alcohol  abuse  is  associated 
with  increased  nighttime  hypoxemia 
in  older  men.  Alcohol  Clin  Exp  Res 
11:368-371,1987. 

Vitiello,  M.V.;  Prinz,  P.N.;  Personius, 
J. P.;  Vitaliano,  P.P.;  Nuccio,  M.A.;  and 
Koerker,  R.  Relationship  of  alcohol 
abuse  history  to  nighttime  hypoxemia  in 
abstaining  chronic  alcoholic  men.  J  Stud 
Alcohol  51:29-33,  1990a. 

Vitiello,  M.V.;  Prinz,  P.N.;  Personius, 
J. P.;  Vitaliano,  P.P.;  Nuccio,  M.A.; 
Koerker,  R;  and  Scurfield,  R  Nigh  time 
hypoxemia  is  increased  in  abstaining 
chronic  alcoholic  men.  Alcohol  Clin  Exp 
Res  14:38^1,  1990£. 

Wagman,  A.M.,  and  Allen,  RP.  Effects  of 
alcohol  ingestion  and  abstinence  on  slow 
wave  sleep  of  alcoholics.  Adv  Exp  Med 
5^/59:453^66,1975. 

Wagman,  A.M.;  Allen,  R.P.;  and  Upright, 
D.  Effects  of  alcohol  consumption  upon 


parameters  of  ultradian  sleep  rhythms  in 
alcoholics.  Adv  Exp  Med  Biol  8 5 A: 601- 
616,  1977. 

Wagman,  A.M.;  Allen,  R.P.;  Funderburk, 
F.;  and  Upright,  D.  EEG  measures  of 
functional  tolerance  to  alcohol.  Biol 
Psychiatry  13:719-778,  1978. 

Wand,  G.S.  Alcohol,  the  hypothalamic- 
pituitary-adrenal  axis,  and  hormonal 
tolerance.  In:  Zakhari,  S.,  ed.  Alcohol  and 
the  Endocrine  System.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  23.  NIH  Pub. 
No.  93-3533.  Bethesda,  MD:  the 
Institute,  1993.  pp.  251-270. 

Wand,  G.S.,  and  Dobs,  A.S.  Alterations 
in  the  hypothalamic-pituitary-adrenal  axis 
in  actively  drinking  alcoholics.  /  Clin 
Endocrinol  Metab  72:1290-1295,  1991. 

Wehrenberg,  W.,  and  Ehlers,  C.L.  The 
effects  of  growth  hormone  releasing  fac- 
tor on  the  brain.  Science  232:1271- 
1272,  1986. 

Wetterberg,  L.;  Aperia,  B.;  Gorelick, 
DA.;  Gwirtzman,  H.E.;  McGuire, 
M.T.;  Serafetinides,  E.A.;  and  Yuwiler, 
A.  Age,  alcoholism  and  depression  are 
associated  with  low  levels  of  urinary 
melatonin.  J  Psychiatry  Neurosci 
17:215-224,  1992. 

Williams,  H.L.,  and  Rundell,  O.H.,  Jr. 
Altered  sleep  physiology  in  chronic  alco- 
hol: Reversal  with  abstinence.  Alcohol 
Clin  Exp  it«  5:318-325,  1981. 

Yules,  R.B.;  Freedman,  D.X.;  and 
Chandler,  K.A.  The  effect  of  ethyl 
alcohol  on  man's  electroencephalo- 
graphic  sleep  cycle.  Electroencephalogr 
Clin  Neurophysiol  20:109-111,  1966. 

Yules,  R.B.;  Lippman,  M.E.;  and  Freedman, 
D.X.  Alcohol  administration  prior  to 


432 


Alcohol  and  Sleep 


sleep:  The  effect  on  EEG  sleep  stages.  Pharmacology  of  the  State  of  Alertness. 

Arch  Gen  Psychiatry  16:94-97,  1967.  Oxford,  England:  Pergamon  Press,  1979. 

pp.  9-38. 
Zarcone,  V.  Alcoholism  and  sleep.  Adv 
Biosci  21:29-38,  1978.  Zgombick,  J.,  and  Erwin,  V.G.  Central 

mechanisms  of  ethanol-induced  adrenocor- 
Zarcone,  V.  Alcoholism  and  sleep.  In:  deal  response  in  selectively  bred  lines  of  mice. 

Passonant,  P.,  and  Oswald,  I.,  eds.  Neuroendocrinolojjy  46:324-332,  1987. 


433 


STUDIES  OF 

COGNITIVE/BEHAVIORAL/ 

STRUCTURAL  DEFICITS 

IN  HUMANS 


Chapter  13 

Neuropsychological  Vulnerabilities 
in  Chronic  Alcoholism 

Marlene  Oscar-Berman,  Ph.D. 


KEY  WORDS:  brain  damage;  neuropsychological  assessment;  AODR  (alcohol  or 
other  drug-related)  disorder;  chronic  AODE  (effects  ofAOD  use,  abuse,  and  de- 
pendence); aging;  gender  differences;  nutrition;  elderly;  malnutrition;  thiamine 
deficiency;  Korsakoff  s  syndrome;  cerebral  cortex;  cerebral  hemisphere;  neurobio- 
logical  theory;  emotion;  memory;  risk  factors;  literature  review 


Alcohol-related  brain  damage  has  been 
associated  with  a  variety  of  neuropsy- 
chological changes,  among  which  are 
deficits  in  cognitive,  emotional,  and 
behavioral  functioning.  This  chapter 
reviews  accomplishments  in  neuropsy- 
chological investigations  of  the  residual 
consequences  of  long-term  chronic  alco- 
holism. (There  is  minimal  coverage  of 
imaging  and  electrophysiological  studies 
in  this  chapter  because  these  topics  are 
discussed  in  chapters  14  and  15,  respec- 
tively.) Residual  effects — in  contrast  to 
acute  effects — can  be  studied  only  in 
people  who  have  abused  alcohol  for  a 
lengthy  period  of  time  but  who  have 
been  sober  for  at  least  4  weeks  before 
testing.  In  addition,  they  must  be  free  of 
comorbid  medical  complications  that 
can  affect  brain  functioning.  Comorbid 


conditions  (e.g.,  head  injury,  history  of 
abuse  of  other  drugs,  concurrent  psy- 
chiatric diagnoses,  liver  disease,  HIV 
infection)  usually  are  exclusionary  criteria 
in  the  study  of  alcohol-related  brain 
damage  because  comorbidities  may 
contribute  to  artifactual  results  that 
complicate  interpretations  about  the 
neurobehavioral  sequelae  of  alcoholism 
(Oscar-Berman  et  al.  1997;  Neiman 
1998).  The  exception  is  when  researchers 
are  specifically  interested  in  understand- 
ing the  combined  consequences  of  a 
comorbid  condition  with  alcoholism 
(see  Galanter  1998  and  chapter  14  in 
this  monograph). 

The  locus  and  extent  of  alcoholism- 
related  brain  damage,  as  well  as  the 
type  and  degree  of  impairment,  differ 
across  individuals.  These  differences 


M.  Oscar-Berman,  Ph.D.,  is  professor  of  neurology  and  professor  of  psychiatry,  Division  of  Psychiatry, 
M-902,  Boston  University  School  of  Medicine,  715  Albany  St.,  Boston,  MA  02118;  she  is  also  a  research 
scientist  at  the  Department  of  Veterans  Affairs  New  England  Healthcare  Systems,  Boston, 
Massachusetts  Division. 


437 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


suggest  vulnerability  of  certain  sub- 
groups of  alcoholics  (e.g.,  the  elderly 
and  women),  and  susceptibility  of  par- 
ticular areas  of  the  brain  to  alcoholism- 
related  damage  (e.g.,  cerebral  cortex 
and  limbic  and  diencephalic  structures). 
To  account  for  the  divergent  findings, 
researchers  have  proposed  different 
models  (or  hypotheses).  For  context, 
table  1  lists  common  current  theoretical 
frameworks  proposed  to  explain  various 
residual  neuropsychological  effects  of 
alcoholism.  Although  these  models 
tend  to  focus  on  vulnerable  subject 
groups,  brain  regions,  or  mental  pro- 
cesses, they  are  not  mutually  exclusive; 
some  are  interrelated.  Supporting 
data  for  these  models  come  from 
neurobehavioral  studies,  brain  scans,  elec- 
trophysiological studies,  and  postmor- 
tem neuropathology. 


In  this  chapter  I  discuss  three 
significant  subject  characteristics  that 
contribute  to  increased  vulnerability 
to  the  effects  of  alcoholism:  aging, 
nutrition,  and  gender.  Another  focus 
of  the  discussion  is  on  vulnerable 
cortical  brain  systems  that  are  pur- 
ported to  mediate  specific  functional 
deficits:  the  frontal  lobes  and  the 
right  hemisphere.  A  primary  goal  of 
this  review  is  to  help  establish  future 
priorities  in  research  on  alcohol  abuse 
and  alcoholism. 

VULNERABILITY 
RELATED  TO  SUBJECT 
CHARACTERISTICS 

Not  all  alcoholics  are  equally  at  risk 
for  brain  changes  and  neuropsycho- 
logical deficits,  nor  are  all  alcoholics 


Table  1.   Models  Proposed  To  Explain  the  Neuropsychological  Consequences  of  Alcohol- 
Related  Brain  Damage. 

Models  Emphasizing  Vulnerability  in  Terms  of  Subject  Variables 

•  Premature  aging:  Alcoholism  accelerates  aging.  Brains  of  alcoholics  resemble  brains  of 
chronologically  older  nonalcoholics. 

•  Gender:  Alcoholism  affects  women  more  than  men. 

•  Vitamin  deficiency:  Thiamine  deficiency  contributes  to  brain  lesions,  especially  in 
Wernicke  -  Korsakoff  syndrome . 

Models  Emphasizing  Vulnerability  of  Brain  Regions/Systems  or  Functional  Processes 

•  Diffuse  cortical  atrophy:  Cerebral  atrophy  occurs  throughout  the  brain. 

•  Right  hemisphere  functions  are  more  vulnerable  to  the  effects  of  alcoholism  than  are  left 
hemisphere  functions.  (Similar  observations  have  been  made  with  normal  chronological 
aging,  hence  support  for  the  premature  aging  hypothesis.) 

•  Frontal  lobe  systems  are  more  vulnerable  to  the  effects  of  alcoholism  than  other  cortical 
systems. 

•  Limbic  and  diencephalic  brain  regions  are  vulnerable  to  the  effects  of  alcoholism  in 
Korsakoff's  syndrome. 

•  Neurotransmitter  systems:  Emphasis  is  placed  on  disruption  of  neurotransmitter  systems, 
e.g.,  acetylcholine,  dopamine,  gamma-aminobutyric  acid,  glutamate,  and  serotonin 
systems. 

•  Reduction  in  efficiency  of  mental  operations:  Emphasis  is  placed  on  impairments  of 
underlying  cognitive  processes. 


438 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


affected  similarly.  Therefore,  when 
trying  to  understand  the  effects 
of  alcoholism  on  brain  and  behavior, 
researchers  must  be  aware  of  the 
influence  of  a  wide  range  of  interre- 
lated variables  (figure  1).  These 
variables  include  the  following  indi- 
vidual differences: 

•  Age:  The  very  young  and  the 
elderly  are  especially  vulnerable 
(Dufour  1996;  Oscar-Berman  and 
Schendan  in  press). 

•  Comorbid  medical,  drug  abuse, 
and  psychiatric  conditions  (Sher 
and  Trull  1994;  Lieber  1995; 
Galanter  1998;  Bates  and  Convit 
1999;  see  also  chapters  14  and  15 
in  this  monograph). 

•  Nutrition  (Cook  et  al.  1998; 
Shimomura  et  al.  1998). 

•  Education,  intelligence,  and  socio- 
economic background  (Lezak 
1995;  Rourke  and  Loberg  1996). 

•  Ethnicity:  In  addition  to  the 
contribution  of  gene  variants, 
ethnicity  influences  environmental 
factors  such  as  drinking  habits 
and  nutrition  (Keefe  and  New- 
comb  1996). 

•  Gender:  Deleterious  effects  of  alco- 
holism may  differ  for  men  and 
women  (Glenn  1993;  Nixon  1993). 

•  Genetics  and  family  history: 
Alcoholism  is  genetically  heteroge- 
neous. Its  expression  is  influenced 
by  gene  variants,  which  also  con- 
tribute to  ethnicity  (Wall  et  al. 
1996)  and  to  personality  traits 
(e.g.,  antisocial  personality  disor- 
der and  attention -deficit/hyperac - 
tivity  disorder  [Schuckit  1997]). 
In  addition,  nonalcoholic  offspring 


of  alcoholics  show  abnormal 
neuropsychological  signs  (Pihl  and 
Bruce  1995;  Porjesz  and  Begleiter 
1995),  and  patients  with  alcoholic 
dementia  may  be  genotypically  dis- 
tinct from  patients  with  alcoholic 
Korsakoff's  syndrome  (Muramatsu 
et  al.  1997). 
•  Other  factors:  Cranial  circumference 
(see  chapter  15);  experimental 
methodologies  (e.g.,  specific  tests 
and  procedures);  size  and  homogene- 
ity of  samples;  comparability  of 
human  and  nonhuman  animal 
models  (Lancaster  1995;  Rourke 
and  Loberg  1996;  Gallagher  and 
Rapp  1997;  Oscar-Berman  and 
Bardenhagen  1998). 

Although  the  contributions  of 
many  of  these  variables  are  not  spe- 
cific to  the  effects  of  alcoholism,  some 
have  direct  relevance.  Among  the 
variables  that  have  been  assessed 
recently,  and  that  have  potentially 
informative  consequences  for  eluci- 
dating the  neuropsychological  seque- 
lae of  alcoholism,  are  the  subject 
characteristics  of  aging,  nutrition,  and 
gender.  However,  it  is  important  to 
note  that  comorbid  medical,  neuro- 
logical, and  psychiatric  conditions 
occur  in  a  large  number  of  alcoholics. 
In  an  attempt  to  clarify  the  nature  of 
residual  effects  of  alcoholism,  many 
alcohol  researchers  cautiously  exclude 
alcoholic  subjects  with  common  con- 
ditions that  may  contribute  to  arti- 
facts in  interpreting  the  results.  Such 
a  strategy  ignores  a  large  population 
of  alcoholics  who  have  co-occurring 
risk  factors.  For  progress  to  be 
made  on  this  front,  future  research 


439 


NLAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


strategies  must  include  samples  of  alco- 
holics who  have  comorbid  conditions. 
For  consistency  in  research  protocols 
across  laboratories,  study  groups  can 
be  homogeneous  with  respect  to  many 
subject  characteristics  (e.g.,  age,  gender, 
family  history),  and  each  co-occurring 
risk  factor  can  be  studied  systematically. 
Some  laboratories  are  already  taking 
this  approach  (e.g.,  Nixon  et  al.  1998; 
see  reviews  by  Rourke  and  Loberg  1996; 


Galanter  1998;  Bates  and  Convit 
1999;  see  also  chapters  14  and  15  in 
this  monograph). 

Aging 

Background 

Neuropathological  analyses  provided 
some  of  the  earliest  insights  into  the 
relationship  between  alcoholism  and 
aging.  In  postmortem  specimens  of 


Neuropsychiatric  Risk  Factors 

Pre-abuse:  FAS/FAE 

Systemic  illnesses  and  general  health 

Head  injury 

Psychiatric  comorbidity 

Use  of  other  drugs 


Age 

Genetics 

Temperament 

FH+ 


Alcohol  Abuse 

Amount  per  occasion 
Duration  of  abusive  drinkin 
Pattern  over  lifetime 
Recent  amount/duration 
Length  of  abstinence 


Test 

Characteristics 

&  Sample 


I 


Education 
&SES 


Brain 
Structure 

& 
Function 


Motivation 
Expectancies 


Neuropsychological 
Performance 


Figure  1.  Variables  to  consider  in  models  of  alcohol -associated  neuropsychological  abnormali- 
ties. FAS  =  fetal  alcohol  syndrome;  FAE  =  fetal  alcohol  effects;  FH  =  family  history;  SES  = 
socioeconomic  status.  Reprinted  with  permission  from  Rourke,  S.B.,  and  Loberg,  T.  The 
neurobehavioral  correlates  of  alcoholism.  In:  Grant,  I.,  and  Adams,  K.M.,  eds. 
Neuropsychological  Assessment  of  Neuropsychiatric  Disorders.  2d  ed.  New  York:  Oxford 
University  Press,  1996.  p.  443. 


440 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


brains  of  alcoholics,  cerebral  atrophy 
was  noted  to  resemble  the  brain  shrink- 
age that  occurs  with  normal  chronologi- 
cal aging  (Courville  1966).  The  atrophy 
was  most  prominent  in  the  frontal 
lobes,  and  it  extended  backward  to  the 
parietal  lobes.  This  finding  was  replicated 
by  others,  who  reported  abnormal 
ventricular  enlargement  and  widening 
of  the  cerebral  sulci  of  alcoholics  in 
relation  to  increasing  age  (Pfefferbaum 
et  al.  1993).  From  the  observed  simi- 
larities in  the  brains  of  alcoholic  and 
aging  individuals  came  a  search  for 
parallels  in  functional  decline  associated 
with  alcoholism  and  aging. 

The  premature  aging  hypothesis  has 
been  put  forth  in  two  versions  (reviewed 
by  Ellis  and  Oscar-Berman  1989). 
According  to  the  first  version,  the 
"accelerated  aging"  (or  "cumulative 
effects")  model,  alcoholism  is  accom- 
panied by  the  precocious  onset  of 
neuroanatomical  and  behavioral  changes 
typically  associated  with  advancing 
age.  Cognitively,  or  neuropsychologi- 
cally,  alcoholics  become  old  before 
their  time.  This  version  proposes  that 
alcoholics  at  all  ages  are  impaired 
compared  with  age-matched  nonalco- 
holic control  subjects.  The  second  ver- 
sion places  the  timing  of  the  changes 
somewhat  differently.  In  this  view, 
which  has  been  labeled  the  "increased 
vulnerability"  interpretation,  the  aging 
brain  is  more  vulnerable  to  the  delete- 
rious influences  of  toxic  substances, 
including  ethanol,  than  is  the  brain  of 
a  younger  person.  Therefore,  the  cog- 
nitive decline  associated  with  normal 
chronological  aging  (beginning  at 
around  age  50)  receives  added  momen- 
tum when  combined  with  alcoholism. 


This  version  proposes  that  older 
alcoholics  are  impaired  compared  with 
age-matched  nonalcoholics;  however, 
this  would  not  be  the  case  for 
younger  alcoholics. 

Research  Review 

Taken  together,  most  of  the  evidence 
from  neuropathological  and  neurora- 
diological  investigations  supports  the 
view  of  a  link  between  alcoholism  and 
premature  aging.  Furthermore,  studies 
favoring  the  increased  vulnerability 
model  are  more  common  than  those 
supporting  the  accelerated  aging 
model,  although  results  of  a  study  by 
Belzunegui  and  colleagues  (1995) 
favor  the  latter.  These  investigators 
examined  neuronal  nuclear  size  and 
neuronal  population  of  the  mammillary 
bodies  and  anterior  thalamic  complex 
in  alcoholics  and  control  subjects  ages 
30-75.  These  subcortical  structures  are 
of  particular  interest  because  lesions  to 
them  are  thought  to  be  critical  for 
establishing  the  memory  impairment 
in  alcoholic  Korsakoff's  syndrome  (for 
reviews,  see  Kopelman  1995;  Oscar- 
Berman  and  Evert  1997).  Belzunegui 
and  colleagues  found  significant 
reductions  in  neuronal  size  and  number 
in  the  alcoholics,  but  the  youngest  alco- 
holics showed  the  greatest  differences. 
Most  data,  however,  support  the 
idea  that  older  alcoholics  are  especially 
vulnerable  to  the  effects  of  alcoholism. 
Elderly  alcoholics  have  an  increased 
risk  of  accidents,  side  effects,  and  toxi- 
city resulting  from  alcohol  intake.  In 
part  this  is  because  older  people  have 
a  decreased  ability  to  metabolize  alco- 
hol and  may  have  concomitant  med- 
ical problems  (National  Institute  on 


441 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Alcohol  Abuse  and  Alcoholism  [NIAAA] 
1997).  As  predicted  by  the  increased  vul- 
nerability model,  certain  brain  structures 
show  greater  reduction  in  size  in  older 
alcoholics  than  in  younger  alcoholics. 
These  are  the  cerebral  cortex  (Nicolas 
et  al.  1997;  Pfefferbaum  et  al.  1997), 
the  corpus  callosum  (Pfefferbaum  et 
al.  1996),  the  hippocampus  (Sullivan 
et  al.  1995#;  but  see  Harding  et  al. 
1997),  and  the  cerebellum  (Sullivan  et 
al.  1995^,  1996;  Harris  et  al.  1999;  see 
also  chapter  14  in  this  monograph). 
Alcoholics  also  have  shown  a  stronger 
association  between  age  and  ventricular 
dilation  compared  with  nonalcoholic 
control  subjects  (Di  Sclafani  et  al. 
1995;  Pfefferbaum  et  al.  1997). 

Results  of  neurobehavioral  investi- 
gations tend  to  support  the  view  that 
aging  increases  one's  vulnerability  to 
alcohol-related  decline,  but  controversy 
prevails  because  the  association  between 
alcoholism  and  aging  is  less  reliable 
with  behavioral  measures  than  with 
imaging  techniques  (see  reviews  by 
Evert  and  Oscar-  Berman  [1995]  and  by 
Oscar- Berman  and  Schendan  [in  press]). 
More  important,  when  neuroradio- 
logical  and  behavioral  changes  are 
examined  together  in  the  same  study, 
concomitant  brain  damage  with  per- 
formance impairments  is  not  always 
found.  For  example,  Sullivan  and  col- 
leagues (1995#)  reported  clear  evidence 
of  brain  abnormalities  but  no  differ- 
ences on  explicit  memory  tests  in  alco- 
holics relative  to  nonalcoholic  control 
subjects  ages  21-70  years.  Explicit 
memory  refers  to  the  ability  to  con- 
sciously remember  facts  and  events,  is 
assessed  by  recognition  and  recall  tests, 
and  is  impaired  in  alcoholic  Korsakoff 


and  other  amnesic  patients  (for  reviews, 
see  Kopelman  1995;  Mayes  1995).  The 
hippocampus  and  adjacent  cortical 
areas  have  been  strongly  implicated  in 
explicit  memory  by  a  host  of  neuropsy- 
chological, brain  imaging,  and  nonhu- 
man  animal  studies  (Eichenbaum  1997). 
Sullivan  and  colleagues  (1995#)  eval- 
uated whether  there  were  correlations 
between  magnetic  resonance  imaging 
(MRI)  measures  of  hippocampal  volume 
and  behavioral  measures  of  verbal  and 
nonverbal  delayed  recall,  as  defined  by 
the  Wechsler  Memory  Scale  (Wechsler 
and  Stone  1945).  Anterior  portions  of 
the  hippocampus  were  found  to  be 
smaller  in  the  alcoholics  than  in  the 
control  subjects,  and  this  difference  was 
even  greater  in  older  than  in  younger 
alcoholics.  Although  the  MRI  findings 
supported  the  increased  vulnerability 
model,  the  behavioral  significance  of 
the  neuroanatomical  reduction  (i.e., 
increased  hippocampal  vulnerability  in 
aging  and  alcoholism)  was  unclear 
because  the  explicit  memory  scores  did 
not  correlate  with  hippocampal  volumes. 
Indeed,  the  alcoholics  were  not  impaired 
on  the  memory  tests.  Furthermore, 
although  Sullivan  and  colleagues 
observed  visuospatial  impairments  on 
an  IQ  subtest  with  a  memory  compo- 
nent (Digit  Symbol),  there  were  no 
significant  correlations  with  hippo- 
campal volume. 

In  contrast  to  the  findings  of  Sullivan 
and  colleagues  (1995#)  were  those 
from  another  study,  which  found  no 
evidence  of  cortical  atrophy  but  did 
show  aging-related  cognitive  deficits 
(Di  Sclafani  et  al.  1995).  In  that  study, 
older  alcoholics  and  control  subjects 
were  given  MRI  scans  and  numerous 


442 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


age-normed  neuropsychological  tests 
to  compare  brain  measures  and  cognitive 
abilities.  The  alcoholics  displayed  clear 
impairments  on  memory  and  visuospa- 
tial  tasks,  but  there  were  no  group  dif- 
ferences in  global  cerebral  atrophy 
(although  two  alcoholics  had  significant 
atrophy).  One  reason  for  cognitive 
dysfunction  in  the  absence  of  changes 
in  gross  brain  morphology  is  that  there 
can  be  synaptic  neuronal  alterations 
that  affect  processing  but  are  unde- 
tectable at  a  macroscopic  anatomical 
scale  (see  Harper  1998  for  review). 
However,  Di  Sclafani  and  colleagues 
did  find  a  stronger  association  between 
age  and  ventricular  dilation  in  the 
alcoholics  than  in  the  control  subjects. 

In  a  neurobehavioral  study,  Ellis 
(1990)  found  that  alcoholics  ages  48-74 
were  impaired  compared  with  nonalco- 
holic peers  and  younger  alcoholics  (ages 
25-47)  on  Performance  IQ  subtests 
of  the  Wechsler  Adult  Intelligence 
Scale— Revised  (WAIS-R)  (Wechsler 
1981).  Although  that  finding  was  sup- 
portive of  the  increased  vulnerability 
model,  another  observation  in  the  same 
study  was  not  supportive.  The  older 
alcoholics  did  not  show  deficits  out  of 
proportion  to  their  age  on  dichotic 
listening  tasks  of  right  hemisphere 
functioning  (a  string  of  piano  notes). 

In  another  study  assessing  the  cog- 
nitive domain  of  selective  attention 
(Evert  and  Oscar-Berman  unpublished 
manuscript),  male  and  female  alcoholics 
and  nonalcoholic  control  subjects  ages 
29-76  were  given  a  cued  detection  task 
in  which  the  pre-cue  provided  infor- 
mation about  the  most  probable  visual 
field  (VF)  location  of  a  subsequent 
target  letter.  The  nonalcoholic  controls 


showed  a  VE  asymmetry,  reflected  as  a 
specific  disruption  when  the  cue  was 
presented  in  the  Right  VF  and  the  target 
was  presented  in  the  Left  VF.  Unlike 
the  younger  control  subjects,  the  alco- 
holics in  both  age  groups  (and  the 
older  nonalcoholic  control  subjects) 
showed  no  evidence  of  a  right  hemi- 
sphere (Left  VF)  advantage  on  our  task; 
they  were  influenced  by  the  validity  of 
the  cue  regardless  of  the  VF  in  which 
it  was  presented.  Gender  differences 
were  absent.  In  sum,  within  the  cog- 
nitive domain  of  selective  attentional 
processing,  the  results  from  this 
experiment  provided  support  for  the 
accelerated  aging  model  of  premature 
aging  (and  also  for  the  right  hemi- 
sphere hypothesis  discussed  later  in 
this  chapter). 

Results  of  numerous  additional  stud- 
ies examining  right  hemisphere  func- 
tional decline  in  relation  to  alcoholism 
and  aging  have  not  been  sufficienriy 
consistent  to  resolve  the  premature  aging 
issue  (for  reviews,  see  Oscar-Berman 
1992;  Oscar-Berman  and  Schendan  in 
press;  see  also  Coffey  et  al.  1998). 
Consequentiy,  the  concept  of  aging  as 
a  model  for  the  study  of  alcoholism 
remains  unresolved. 

It  should  be  noted  that  patients  with 
alcoholic  Korsakoff's  syndrome  (see  the 
next  section)  typically  display  deficits 
that  are  more  severe  than  those  of 
age-equivalent  non- Korsakoff  alcoholics 
(Evert  and  Oscar-Berman  1995;  Oscar- 
Berman  and  Pulaski  1997).  Interestingly, 
Wilkinson  and  Carlen  (1982)  indi- 
cated that  the  cortical  brain  morphology 
scores  of  patients  with  Korsakoff's 
syndrome  were  not  age  related,  as  they 
were  in  alcoholics  without  Korsakoff's 


443 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


syndrome.  This  may  be  because  patients 
with  KorsakofPs  syndrome  already 
have  suffered  maximal  alcohol-related 
brain  damage,  such  that  age-related 
cortical  cell  loss  becomes  irrelevant.  In 
any  case,  it  is  important  in  considering 
alcoholic  populations  (with  and  without 
KorsakofPs  syndrome)  to  be  able  to 
differentiate,  describe,  and  quantify 
the  separate  contributions  of  aging  and 
alcoholism  to  cognitive  decline. 

Nutrition 

Background 

Malnutrition  is  not  a  common  conse- 
quence in  alcoholics  who  have  adequate 
nourishment,  but  it  is  in  those  who 
eat  poorly  or  who  have  medical  condi- 
tions that  affect  the  utilization  of 
nutrients.  Groups  at  risk  include  the 
poor,  elderly  people  living  alone,  people 
with  liver  disease,  and  schizophrenics 
(Butterworth  1995;  Casanova  1996; 
NIAAA  1997;  Nussbaum  1997). 
Dietary  factors  such  as  ingestion  of 
vitamins,  protein,  and  saturated  fats 
may  modulate  the  severity  of  alcohol- 
related  organ  damage,  and  secondary 
malnutrition  occurs  from  malabsorption 
due  to  gastrointestinal  complications 
(e.g.,  pancreatic  insufficiency,  gastrec- 
tomy, and  changes  in  hepatic  metabo- 
lism) (Lieber  1995;  Shimomura  et  al. 
1998).  Thus,  malnutrition  can  influ- 
ence brain  structure  and  function  in 
many  ways  (Witt  1985;  Marsano 
1993;  Tarter  et  al.  1993;  Lieber  1995). 
One  area  of  growing  interest  is 
damage  to  the  cerebellum,  which  may 
be  induced  by  thiamine  deficiency 
(Welch  et  al.  1997).  Cerebellar  damage 
produces  problems  with  movement 


(mainly  gait,  balance,  and  coordina- 
tion). However,  cerebellar  atrophy  can 
be  present  in  alcoholics  without  sig- 
nificant movement  abnormalities  (Sul- 
livan et  al.  1995&).  The  role  of  the 
cerebellum  in  cognition  is  controversial 
and,  therefore,  so  is  the  connection  of 
cerebellar  damage  to  cognitive  changes 
in  alcoholism  (McGlinchy-Berroth  et 
al.  1995;  Sullivan  et  al.  1995 b\  Shear  et 
al.  1996;  Schmahmann  1997;  Ivry 
1998;  Woodruff-Pak  1998;  Harris  et 
al.  1999;  see  also  chapter  14  in  this 
monograph).  Some  investigators  have 
suggested  that  alcoholic  cerebellar 
degeneration  represents  the  same  dis- 
ease as  Wernicke's  encephalopathy,  an 
acute,  transient  stage  of  alcohol- 
related  neurological  problems  which 
include  confusion  and  abnormalities 
in  oculomotor  and  gross  muscle  con- 
trol (Reuler  et  al  1985;  Victor  1992; 
Cooketal.  1998). 

Thiamine  deficiency  also  has  long 
been  associated  with  KorsakofPs  syn- 
drome (see  reviews  by  Bowden  1990; 
Kopelman  1995;  Langlais  1995; 
Oscar-Berman  and  Evert  1997;  Cook 
et  al.  1998).  The  syndrome  is  character- 
ized most  notably  by  a  severe  antero- 
grade memory  loss,  which  coexists 
with  an  IQ  within  normal  limits,  but 
other  cognitive,  emotional,  and  moti- 
vational deficits  exist  (Oscar-Berman 
and  Evert  1997).  Memory  is  better 
for  semantic,  procedural,  implicit,  and 
incidental  information  than  for  explicit 
factual  knowledge  (Beauregard  et  al. 
1997;  Oscar-Berman  and  Evert  1997; 
Seger  et  al.  1997;  Oscar-Berman  and 
Bardenhagen  1998).  Alcoholic 
KorsakofPs  syndrome  is  usually  pre- 
ceded by  Wernicke's  encephalopathy; 


444 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


this  acute  phase  typically  disappears 
with  abstinence  and  good  nutrition. 
In  the  following  section,  I  discuss 
research  findings  relevant  to  purported 
relationships  among  nutritional 
considerations,  brain  damage,  and 
neurobehavioral  impairments  in 
Wernicke -Korsakoff  patients. 

Research  Review 

The  etiology  of  Wernicke-Korsakoff 
syndrome  is  controversial  (Butterworth 
1995;  Kopelman  1995;  Ma  and 
Truswell  1995;  Cook  et  al.  1998; 
Lishman  1998).  Some  investigators 
favor  the  view  that  thiamine  deficiency 
is  the  primary  cause  (Bowden  1990; 
Joyce  1994;  Shimomura  et  al.  1998); 
others  believe  that  the  direct  neuro- 
toxic effects  of  ethanol  are  causal 
(Laas  and  Hagel  1994);  still  others 
have  suggested  that  acute  Wernicke's 
encephalopathy  is  attributable  to  the 
effects  of  thiamine  deficiency,  whereas 
chronic  Korsakoff  s  syndrome  is  due  to 
the  interaction  of  thiamine  deficiency  and 
ethanol  neurotoxicity  (Lishman  1998). 
Joyce  (1994)  has  stated  that  while 
ethanol  neurotoxicity  can  damage  cor- 
tical neurons,  thiamine  malnutrition 
affecting  the  diencephalon  can  account 
for  neuropsychological  deficits  in  all 
brain-damaged  alcoholics.  Cullen  and 
Halliday's  evidence  (1995)  placed  the 
locus  of  brain  damage  from  thiamine 
deficiency  in  the  cholinergic  nucleus 
basalis  of  alcoholics,  but  in  a  later  study 
(Cullen  et  al.  1997)  the  investigators 
provided  evidence  that  cell  loss  in  that 
region  was  unrelated  to  memory  loss  in 
alcoholic  amnesia.  In  contrast,  Lishman 
(1998)  suggested  distinct  subgroups  or 
clinical  forms  of  alcoholics  according 


to  the  brain's  vulnerability  to  alcoholism. 
The  vulnerability  derives  from  two 
distinct  pathological  influences,  which 
may  operate  independently  in  some 
people  and  may  interact  in  others.  The 
first,  characterized  as  shrinkage  of  the 
cerebral  cortex,  as  well  as  possible  atro- 
phy of  basal  forebrain  regions,  is  thought 
to  result  from  the  direct  neurotoxic 
effects  of  ethanol  or  its  metabolites.  The 
second,  characterized  by  damage  to 
the  diencephalon,  is  attributed  to  thi- 
amine deficiency.  According  to  Lishman, 
alcoholics  who  are  susceptible  to  ethanol 
toxicity  alone  may  develop  permanent 
or  transient  cognitive  deficits  associated 
with  cortical  shrinkage.  Those  who  are 
susceptible  to  thiamine  deficiency 
alone  will  develop  a  mild  or  transient 
Korsakoff  state,  with  anterograde 
amnesia  as  a  salient  feature.  Individuals 
with  dual  vulnerability,  suffering  from 
a  combination  of  ethanol  neurotoxicity 
and  thiamine  deficiency,  will  have  wide- 
spread damage  to  large  regions  of  the 
cerebral  cortex,  as  well  as  to  deep  brain 
structures.  These  people  will  exhibit 
severe  anterograde  amnesia  as  well  as 
other  cognitive  impairments. 

Evidence  to  support  the  various 
models  is  contradictory  (Langlais 
1995).  In  Australia,  an  investigation 
was  launched  of  the  cost- effectiveness 
of  thiamine  supplementation  alterna- 
tives, on  the  assumption  that  thiamine 
deficiency  causes  Wernicke -Korsakoff 
syndrome  (Connolly  and  Price  1996), 
and  a  policy  of  thiamine  enrichment  of 
bread  flour  was  introduced  in  1991.  Ma 
and  Truswell  (1995)  conducted  a  ret- 
rospective survey  of  Australian  hospital 
records  indicating  diagnoses  of 
Wernicke's     encephalopathy     or 


445 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Korsakoff's  syndrome  over  a  16-year 
period  (1978-1993).  The  goal  of  the 
study  was  to  determine  whether  the 
introduction  of  the  thiamine  enrichment 
program  was  associated  with  a  decline 
in  incidence  of  the  disorders.  The  num- 
bers of  acute  cases  began  to  fall  before 
1991,  but  the  numbers  for  years  1992 
and  1993  were  significantiy  lower  than 
in  all  previous  years.  Ma  and  Truswell 
concluded  that  although  their  results  are 
consistent  with  a  preventive  effect  of 
mandatory  thiamine  enrichment  of 
bread,  the  evidence  is  not  conclusive.  It 
may  be  that  response  to  treatment  is  a 
function  of  age  of  onset  of  the  symp- 
toms and  the  rapidity  with  which  treat- 
ment is  instituted  (Tallaksen  et  al.  1993). 
Other  important  factors  include  the 
synergistic  effects  of  thiamine  deficiency, 
ethanol  toxicity,  and  liver  disease 
(Butterworth  1995;  KrU  1995). 

Not  all  alcoholics  develop  Korsakoff  s 
syndrome.  Therefore,  some  investiga- 
tors have  suggested  a  possible  genetic 
component,  or  inborn  predisposition 
for  its  occurrence  (reviewed  by  Oscar  - 
Berman  and  Evert  1997).  In  addition, 
patients  with  alcoholic  Korsakoff's 
syndrome  have  been  described  as  being 
genotypically  distinct  from  patients 
with  alcoholic  dementia  (Muramatsu  et 
al.  1997).  Evidence  in  favor  of  a  genetic 
predisposition  for  Korsakoff's  syn- 
drome revolves  around  the  demonstra- 
tion of  deficient  transketolase  activity 
in  Korsakoff  patients,  thought  to  be 
inherited,  such  that  thiamine  metabo- 
lism is  compromised  (Blass  and  Gibson 
1977;  Butterworth  et  al.  1993;  Wang 
et  al.  1997).  Indeed,  Butterworth  and 
colleagues  (1993)  found  significant 
reductions  of  thiamine-dependent 


enzymes  in  autopsied  cerebellar  samples 
from  alcoholic  patients  with  a  diagnosis 
of  Wernicke  -  Korsakoff  syndrome .  How- 
ever, whereas  the  enzyme  activities  in 
brain  samples  from  non-Korsakoff  alco- 
holics were  within  normal  limits  in 
one  study  (Butterworth  et  al.  1993),  in 
another  study  (Lavoie  and  Butterworth 
1995)  thiamine  deficiency  and  reduced 
enzyme  activity  were  observed.  Also, 
McCool  and  colleagues  (1993)  pro- 
vided evidence  against  genetic  predis- 
position as  a  viable  mechanism,  based 
on  their  own  work  with  genetic  sensi- 
tivity to  thiamine  deficiency  and  on 
other  studies  of  the  development  of 
alcoholic  Korsakoff  s  syndrome. 

Gender 

Background 

Until  recently,  gender  differences  in  the 
neurobiological  effects  of  alcoholism 
have  focused  mainly  on  the  reproductive 
system  and  hepatic  injury.  Evidence 
suggests  that  female  alcoholics  have 
increased  menstrual  disturbances,  spon- 
taneous abortions,  and  miscarriages,  and 
women  are  more  susceptible  to  alcoholic 
liver  disease  than  are  men  (NIAAA 
1997).  Only  in  the  last  decade  have 
women  (and  female  nonhuman  animals) 
been  the  focus  of  research  on  alcohol- 
related  brain  damage  (Lancaster  1994, 
1995).  Still,  sample  sizes  often  have 
been  small,  and  insufficient  attention 
has  been  paid  to  gender  differences  in 
body  size  and  drinking  habits.  Because 
humans  and  rodents  have  opposite 
gender  differences  in  drinking  patterns 
and  responses  to  alcohol  (Lancaster 
1995),  some  animal  models  may  have 
limited  generality  with  respect  to  people. 


446 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


In  any  event,  controversy  remains 
about  whether  and  to  what  extent 
alcoholism  affects  the  brain  and  neuro- 
psychological vulnerability  differently 
in  females  and  males  (reviewed  by 
Glenn  1993  and  Nixon  1993). 

Research  Review 

Parsons  (1994)  reported  that  although 
male  and  female  alcoholic  subjects 
showed  impaired  performance  on  neu- 
ropsychological tests  relative  to  same-sex 
control  subjects,  only  the  male  alcohol- 
ics differed  from  their  control  subjects 
on  measures  of  the  brain's  electrical 
activity.  However,  other  investigators 
found  that  female  alcoholics  displayed 
electrophysiological  abnormalities 
similar  to  those  reported  in  male  alco- 
holics (Hill  and  Steinhauer  1993).  In 
still  other  studies,  using  brain  scans  to 
measure  gender  differences  in  brain  size, 
evidence  is  contradictory.  For  example, 
Lishman  and  colleagues  ( 1987)  reported 
that  female  alcoholics  had  larger  ven- 
tricles compared  with  female  control 
subjects  (as  measured  by  computed 
tomography  [CT]  scans)  than  did  male 
alcoholics  compared  with  male  control 
subjects.  However,  Kroft  and  colleagues 
(1991)  found  that  the  average  ventric- 
ular volume  in  female  alcoholics  was 
within  the  normal  range  of  ventricular 
volumes  found  in  MRI  studies  of 
nonalcoholic  females  of  similar  ages. 
In  another  study  using  CT  brain  scans 
to  measure  atrophy  (Mann  et  al.  1992), 
there  was  evidence  of  a  similar  degree 
of  brain  shrinkage  (and  reexpansion 
after  6  weeks  of  abstinence)  in  men 
and  women,  despite  significantly  shorter 
drinking  histories  in  the  women.  The 
findings  were  obtained  after  controlling 


for  moderating  variables  such  as  age, 
daily  alcohol  consumption  (based  on 
body  weight),  and  liver  dysfunction. 

Using  MRIs,  Hommer  and  col- 
leagues (1996)  measured  the  size  of  the 
corpus  callosum  in  hospitalized  male 
and  female  alcoholics,  and  they  reported 
that  alcoholic  women  had  smaller  cal- 
losal  areas  than  alcoholic  men  and 
nonalcoholic  control  subjects.  The 
reduction  in  size  of  the  corpus  callosum 
was  equally  distributed  along  its  length, 
as  revealed  by  comparisons  among  four 
equal  segments  of  the  callosal  images. 
Alcoholic  men  did  not  differ  from 
nonalcoholic  male  control  subjects  in 
the  size  of  the  corpus  callosum.  Inter- 
estingly, callosal  area  is  notably 
reduced  with  age  in  alcoholics  (Pfef- 
ferbaum  et  al.  1996). 

The  corpus  callosum  enables  com- 
munication between  the  cerebral  hemi- 
spheres. In  both  genders,  the  left  and 
right  sides  of  the  brain  subsume  asym- 
metrical abilities  in  linguistic  (e.g., 
words  or  phrases)  and  nonverbal  (e.g., 
visuospatial  or  musical)  domains.  Specif- 
ically, the  left  hemisphere  usually  is  more 
efficient  than  the  right  for  verbal  signals, 
and  the  right  hemisphere  is  more  efficient 
than  the  left  for  nonverbal  signals.  The 
two  hemispheres  also  are  complementary 
in  their  processing  modes,  depending 
on  context  and  task  demands.  The  left 
hemisphere  plays  a  special  role  in  pro- 
cessing piecemeal  information  analyti- 
cally and  sequentially,  and  the  right 
hemisphere  plays  a  special  role  in  inte- 
grating information  holistically. 

Abnormalities  in  the  structure  of  the 
corpus  callosum  can  occur  as  a  conse- 
quence of  diffuse  cortical  damage  and 
subsequent  degeneration  of  cortical 


447 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


axons.  Diffuse  cortical  damage,  in 
turn,  would  be  interpreted  in  conven- 
tional neuropsychological  testing  as  a 
selective  right  hemisphere  functional 
deficit  either  because  right  hemisphere 
functions  have  less  cortical  representa- 
tion than  left  hemisphere  functions  or 
because  nonverbal  tasks  tend  to  be 
more  difficult  or  less  familiar  than  ver- 
bal tasks  (Ellis  and  Oscar-Berman 
1989).  Alternatively,  diffuse  cortical 
and/or  callosal  atrophy  could  interfere 
with  cross-callosal  transfer  of  informa- 
tion, causing  some  of  the  cognitive 
deficits  observed  in  alcoholics  and 
aging  people  (Hutner  and  Oscar- 
Berman  1996;  Rourke  and  Loberg 
1996).  For  example,  if  the  left  hemi- 
sphere's contribution  to  nonverbal 
tasks  normally  is  greater  than  the  right 
hemisphere's  contribution  to  verbal 
tasks,  an  interhemispheric  transfer  dys- 
function would  affect  visuospatial 
functions  more  than  verbal  functions. 
Because  the  characteristics  of  hemi- 
spheric dominance  are  specific  to  each 
functional  brain  area,  the  altered  pat- 
terns of  hemispheric  dominance  from 
alcoholism  (and  aging)  may  be  idio- 
syncratic to  those  brain  areas  and 
behavioral  functions.  It  is,  therefore, 
important  to  appreciate  which  brain 
regions  are  affected  by  chronic  alco- 
holism (and  aging). 

Results  of  neuropsychological  stud- 
ies, like  the  evidence  from  imaging 
and  electrophysiological  studies,  are 
inconclusive  regarding  the  connection 
between  gender  and  susceptibility  to 
alcohol-related  brain  damage.  For 
example,  Parsons  (1994)  and  Lishman 
and  colleagues  (1987)  reported  no  gen- 
der differences  in  neuropsychological 


functioning,  despite  the  presence  of 
gender  differences  in  alcoholics'  brain 
electrical  activity  and  ventricular  size. 
However,  women  often  have  shorter 
drinking  histories,  consume  less  alcohol, 
and  have  been  abstinent  for  a  longer 
period  of  time  than  men.  Because  other 
individual  differences  in  brain  structure 
and  function  tend  to  obscure  gender 
differences  (Lezak  1995),  one  fruitful 
way  of  studying  possible  gender- related 
neuropsychological  effects  of  alco- 
holism is  to  capitalize  on  known  sexually 
dimorphic  brain  and  behavioral  char- 
acteristics. This  can  be  achieved  in 
conjunction  with  neuropsychological 
tests  having  special  sensitivity  to  the 
functions  of  particular  brain  regions 
or  systems,  especially  in  view  of  differ- 
ences between  men  and  women  in  the 
organization  and  functioning  of  the 
two  cerebral  hemispheres.  Results  of 
meta-analytic  studies  of  functional 
brain  asymmetries  have  shown  that 
men  tend  to  be  more  lateralized  than 
women  (Hiscock  et  al.  1994,  1995); 
women  also  have  a  larger  corpus  callo- 
sum  (Voyer  1996).  Such  differences 
suggest  that  bilateral  representation  of 
verbal  and  nonverbal  functions,  along 
with  greater  interhemispheric  commu- 
nication, is  more  prevalent  in  women 
than  men.  Furthermore,  gender  dif- 
ferences in  brain  size  have  been 
reported  as  a  function  of  aging:  cere- 
brospinal fluid  (CSF)  volume  overall 
was  found  to  be  greater  in  elderly  men 
than  in  elderly  women,  as  were  CSF 
volumes  in  the  sylvian  fissure  and  parieto- 
occipital regions  (Coffey  et  al.  1998). 
However,  aging-related  structural 
cerebral  asymmetries  were  indepen- 
dent of  gender.  In  any  case,  Coffey  et 


448 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


al.'s  normative  measures  of  brain 
aging  (as  well  as  results  of  a  meta-analy- 
sis by  Meinz  and  Salthouse  [1998]) 
appear  to  show  greater  decline  in  men 
than  in  women,  in  distinct  contrast  to 
the  gender  effects  of  alcoholism. 

These  differences  suggest  an  impor- 
tant area  for  future  research,  because 
studies  of  alcoholics  have  consistently 
revealed  a  pattern  of  poor  performance 
on  visuospatial  tasks.  Visuospatial  tasks 
are  also  difficult  for  patients  with  dam- 
age to  the  right  hemisphere.  The  simi- 
larity in  performance  between  alcoholics 
and  right  hemisphere  patients  led 
researchers  to  hypothesize  that  right 
brain  functions  are  more  vulnerable  to 
the  effects  of  alcoholism  than  left 
brain  functions  (see  the  section  on  right 
hemisphere  vulnerability  later  in  this 
chapter).  Of  interest  in  this  regard  is  a 
study  by  Errico  and  colleagues  (1992) 
that  examined  hormone  levels  and 
cognitive  performance  in  male  alco- 
holics. (Alcoholic  women  were  not 
included  in  the  study.)  The  investiga- 
tors measured  testosterone  levels  in 
alcoholics  and  nonalcoholic  control 
subjects  and  related  the  hormone  levels 
to  performance  on  visuospatial  and 
verbal  tasks.  Errico  and  colleagues 
replicated,  in  the  control  group,  the 
findings  of  Christiansen  and  Knussmann 
(1987),  who  observed  a  positive  rela- 
tionship between  testosterone  levels 
and  performance  on  visuospatial  tasks 
in  healthy  nonalcoholic  men.  However, 
in  the  alcoholics,  Errico  and  colleagues 
found  that  the  expected  relationship 
between  testosterone  levels  and  perfor- 
mance on  visuospatial  tasks  was  absent 
compared  with  nonalcoholic  peer  con- 
trol subjects.  Instead,  the  alcoholics' 


testosterone  levels  were  significantly 
correlated  with  performance  on  verbal 
tests.  As  an  explanation,  Parsons  sug- 
gested that  the  alcoholics'  impaired 
visuospatial  abilities  might  have  led 
them  to  rely  on  their  intact  verbal 
functioning,  perhaps  indicating  a  shift 
in  cerebral  dominance  (OA.  Parsons, 
personal  communication,  May  1998). 
Drake  and  colleagues  (1990)  mea- 
sured gender  differences  in  hemi- 
spheric asymmetries  using  dichotic 
listening  procedures  sensitive  to  left 
and  right  hemisphere  functioning.  They 
presented  a  series  of  two  different 
words  or  two  different  melodies  to  male 
and  female  alcoholics  and  nonalcoholic 
control  subjects  to  measure  their  abili- 
ties at  identifying  the  competing  verbal 
or  nonverbal  signals.  Male  alcoholics 
showed  atypical  laterality  patterns. 
Compared  with  control  subjects,  the 
male  alcoholics  showed  a  larger  left 
hemisphere  advantage  for  identifying 
words  and  a  smaller  right  hemisphere 
advantage  for  identifying  melodies.  In 
contrast,  female  alcoholics'  laterality 
patterns  did  not  differ  from  those  of 
control  subjects  on  either  of  the  dichotic 
listening  tasks.  The  authors  concluded 
that  male  alcoholics  showed  evidence  of 
right  hemisphere  dysfunction,  but  female 
alcoholics  did  not.  Unfortunately,  in 
the  alcoholism  literature,  other  studies 
comparing  the  separate  functions  of 
the  left  and  right  cerebral  hemispheres 
(based  on  visual,  tactual,  and  auditory 
signals)  have  relied  mainly  on  male 
research  participants.  Results  of  these 
studies  have  been  inconsistent  in 
showing  atypical  asymmetries,  and  no 
consistent  pattern  of  abnormalities  has 
emerged.  However,  gender  differences 


449 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


typically  have  not  been  examined  (see 
review  by  Oscar-Berman  1992).  In 
addition,  the  question  of  aging-related 
gender  difference  in  hemispheric 
asymmetries  has  not  been  addressed  in 
most  studies  of  alcoholics  (but  Evert 
and  Oscar-Berman  [unpublished  man- 
uscript] found  no  gender  differences). 
This  issue  is  ripe  for  future  research. 

Research  Opportunities 

As  was  indicated  early  on,  numerous 
individual  differences  contribute  to 
the  lack  of  consensus  about  the  effects 
of  alcoholism  on  brain  and  behavior. 
However,  interesting  trends  have 
emerged  when  subject  characteristics 
have  been  studied  systematically 
(aging,  nutrition,  and  gender).  For 
example,  the  role  of  malnutrition 
(especially  thiamine  deficiency)  seems 
to  contribute  to  severe  cognitive 
deficits  (e.g.,  as  in  Wernicke -Korsakoff 
syndrome),  but  the  critical  lesions  and 
the  underlying  mechanisms  remain 
unknown.  New  research  is  needed  to 
clarify  the  separate  and  combined  roles 
of  malnutrition  and  ethanol  neurotox- 
icity to  brain  damage  and  functional 
impairments.  In  addition,  older  alco- 
holics and  women  alcoholics  are  espe- 
cially vulnerable  to  the  deleterious  effects 
of  alcoholism,  as  evidenced  by  a  conver- 
gence of  findings  on  these  subgroups. 
Typically,  older  alcoholic  women  have 
not  been  the  focus  of  investigation. 
Future  studies  can  capitalize  on  normal 
gender  differences  in  brain  organization 
to  address  questions  such  as  whether 
and  how  alcoholism  affects  perceptual 
asymmetries  and  other  neurobehavioral 
measures,  and  how  the  effects  are  bound 


to  aging  (Hiscock  et  al.  1994,  1995; 
Voyer  1996;  Seeman  1997). 

Future  research  should  concentrate 
on  the  promising  trends,  and  investi- 
gations should  be  intensified  of  the 
less-studied  subject  characteristics. 
Expermiental  methods  should  be 
improved  by  encouraging  consistency 
in  research  protocols  across  laborato- 
ries, employing  study  groups  that  are 
homogeneous  with  respect  to  specific 
individual  differences  (e.g.,  age,  gen- 
der, family  history),  and  systematically 
evaluating  each  co-occurring  risk  factor 
(including  the  use  of  statistical  covari- 
ance  procedures  for  confounding  factors, 
when  necessary).  Because  there  may  be 
differing  degrees  of  liability  that  each 
relevant  subject  characteristic  contributes 
to  brain- behavior  impairment,  longi- 
tudinal or  retrospective  methods  could 
be  used  to  evaluate  subgroup  vulnera- 
bility. In  addition,  topics  related  to 
vulnerability  to  cognitive  deficits  may 
be  approached  by  using  nonhuman 
animal  models  (Gallagher  and  Rapp 
1997;  Oscar-Berman  and  Bardenhagen 
1998).  It  is  important  to  assess  the 
comparability  of  human  and  nonhuman 
models  in  alcohol-related  brain  damage; 
similarities  and  differences  among  study 
species  need  to  be  clearly  established 
in  future  work  on  aging,  nutrition,  and 
gender.  Table  2  outlines  the  ideal  meth- 
odological strategies  for  future  research. 

VULNERABILITY  OF 
SPECIFIC  BRAIN  REGIONS 

The  Cerebral  Cortex 

Cortical  changes  have  been  reported 
throughout  the  brain,  but  there  is 


450 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


evidence  that  some  cortical  regions,  (Jernigan  et  al.  1991&;  Pfefferbaum 

especially  the  frontal  and  parietal  and  Rosenbloom  1993;  Wang  et  al. 

lobes,  are  more  consistendy  vulnerable  1993;  Davila  et  al.  1994;  Sullivan  et 

to  the  effects  of  chronic  alcoholism  (as  al.  1995#;  Shear  et  al.  1996;  Estruch 

well  as  aging)  than  other  regions  et  al.  1997;  Kril  et  al.  1997;  Adams  et 


Table  2.  Ideal  Methodological  Strategies  for  Future  Research. 


Make  Subject  Sample  Characteristics  Clear  and  Explicit 

Age,  gender,  race,  ethnicity 

Education;  IQ 

Drinking  history,  family  history  of  alcoholism 

Medical  and  psychiatric  comorbidity 

Develop  Uniform  Definitions  of  Subgroups  of  Alcoholics  With  Brain  Impairments 

Mild  cognitive  deficits 
Wernicke's  encephalopathy 
Korsakoff's  syndrome 
Alcohol-induced  dementia 
Marchiafava-Bignami  disease 
Cerebellar  degeneration 

Develop  Operational  Definitions  of  Functions  To  Be  Measured 

Perceptual  processing/information  processing 
Attention  (e.g.,  sustained  attention,  divided  attention) 
Memory  (e.g.,  working  memory,  explicit/implicit  memory) 
Disinhibition/response  perseveration 
Executive  dysfunction 

Use  Multiple  Methodologies  for  Convergence  of  Evidence 

Neuropsychology 

Standardized  psychiatric  and  neuropsychological  assessment  procedures  (e.g.,  IQ,  memory  tests) 

Paradigms  from  behavioral  neuroscience  (e.g.,  perceptual  processing,  attention,  working 

memory,  disinhibition/response  perseveration) 
Neuroimaging 

Magnetic  resonance  spectroscopy  and  magnetic  resonance  spectroscopic  imaging,  positron 

emission  tomography,  single  photon  emission  computed  tomography,  structural  and 

functional  magnetic  resonance  imaging,  diffuse  tensor  imaging,  magnetoencephalography 
Electrophysiology 

Electroencephalography 
Event- related  potentials 
Neuropathology/Neurochemistry 

Gross  structural  morphology 

Cellular  structure 

Receptor  level  (neurotransmitters) 

Establish  Comparability  Between  Human  and  Nonhuman  Animal  Models 


451 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


al.  1998).  Frontal,  cingulate,  parietal, 
parietooccipital,  and  mesial  temporal 
cortices  in  alcoholics  show  reduced 
metabolic  activity  with  positron  emis- 
sion tomography  (PET)  and  single 
photon  emission  computed  tomography 
(SPECT)  (e.g.,  Adams  et  al.  1993; 
Volkow  et  al.  1995;  Gilman  et  al. 
1996;  Volkow  et  al.  1997;  Gansler  et 
al.  2000)  and  significantly  smaller  gray 
and/or  white  matter  volume  with 
structural  MRI  (e.g.,  Jernigan  et  al. 
1991**;  Sullivan  et  al.  1998). 

Kril  and  colleagues  (1997)  described 
selective  neuronal  loss  at  a  microscopic 
level  in  the  frontal  lobes  of  alcoholics. 
Pfefferbaum  and  colleagues  (1997) 
conducted  regional  MRI  analyses  of 
cortical  integrity  and  found  evidence  that 
the  frontal  lobes  were  especially  vul- 
nerable to  chronic  alcoholism  across  a 
wide  age  range,  but  the  effects  were 
exacerbated  in  elderly  people.  Of  inter- 
est, Pfefferbaum  and  colleagues  also 
noted  that  temporal-parietal  loss 
occurred  mainly  in  older  alcoholics. 
Thus,  their  findings  favor  the 
increased  vulnerability  model  of  pre- 
mature aging  in  alcoholism.  Another 
recent  study  by  Sullivan  and  colleagues 
(1998)  showed  fewer  regional  abnor- 
malities than  those  of  Pfefferbaum  and 
colleagues,  but  both  studies  reported 
a  relative  sparing  of  gray  matter  (but 
loss  of  white  matter)  in  the  posterior 
superior  temporal  region. 

In  a  study  that  compared  MRI 
measures  of  the  brains  of  Korsakoff 
and  non-Korsakoff  alcoholics,  Jernigan 
and  colleagues  (1991^)  observed  that 
Korsakoff  patients  had  greater  ventric- 
ular size,  and  smaller  gray  matter  vol- 
umes in  the  septal  nuclei,  anterior 


hypothalamus,  mesial  temporal  cortex, 
and  orbitofrontal  cortex.  However,  in 
a  later  study  by  Emsley  and  colleagues 
(1996),  comparing  MRI  measures  of 
cortical  and  subcortical  regions  in  alco- 
holics with  and  without  Korsakoff's 
syndrome,  the  extent  of  cortical  gray 
matter  was  similar  in  Korsakoff  patients 
and  control  subjects,  even  though  sub- 
cortical findings  were  similar  to  those  of 
Jernigan  and  colleagues.  Hence,  there 
is  controversy  about  the  locus  and 
extent  of  brain  involvement  among 
alcoholic  subtypes. 

Neurobehavioral  deficits  suggestive 
of  cortical  atrophy  in  alcoholics  have 
been  reported.  Various  studies  have 
disclosed  difficulty  with  tests  of  problem 
solving  and  conceptualization  (Beatty 
et  al.  1993;  Sullivan  et  al.  1993;  Pollux 
et  al.  1995;  Brunfaut  and  d'Ydewalle 
1996;  Sullivan  et  al.  1997),  selective 
attention  (Evert  and  Oscar-Berman 
unpublished  manuscript),  spatial  memory 
(Joyce  and  Robbins  1991;  Verfaellie 
et  al.  1992;  Bowden  and  McCarter 
1993),  working  memory  (Oscar-Berman 
and  Hutner  1993;  Sullivan  et  al. 
1997),  visual  association  learning  and 
recognition  memory  (Bowden  et  al. 
1992;  Oscar-Berman  and  Pulaski  1997), 
and  tactual  learning  (Oscar-Berman  et 
al.  1990).  In  most  studies,  neurobehav- 
ioral deficits  have  been  assessed  indepen- 
dently of  brain  changes.  The  importance 
of  using  both  approaches  (neurobehav- 
ioral and  structural/neuroradiological) 
is  to  evaluate  ideas  related  to  the  con- 
nection between  the  locus  of  damage 
and  the  nature  of  the  decline  in  func- 
tioning. In  studies  where  both  types  of 
measures  have  been  obtained,  results 
have  not  revealed  consistent  convergence 


452 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


between  brain  changes  and  presumed 
functional  loss. 

For  example,  in  a  study  by  Jernigan 
and  colleagues  (1991«),  although  several 
significant  correlations  were  found 
between  MRI  measures  and  perfor- 
mance on  cognitive  tests,  there  was  little 
evidence  of  a  relationship  between  gray 
matter  measures  and  cognitive  test 
scores.  In  another  study,  Wang  and  col- 
leagues (1993)  used  MRI  and  func- 
tional imaging  techniques  in  conjunction 
with  behavioral  measures  for  comparing 
long-term  alcoholic  patients  with  non- 
alcoholic control  subjects.  The  inves- 
tigators noted  that  the  degree  of 
cortical  atrophy  on  MRI  was  associ- 
ated witii  decreased  brain  metabolism, 
perhaps  indicative  of  loss  of  brain  tissue. 
Although  they  did  report  a  relationship 
between  certain  neuropsychological 
test  scores  and  measures  of  frontal 
brain  metabolism  in  the  alcoholics, 
neuropsychological  performance  did 
not  correlate  with  MRI  structural 
changes.  The  lack  of  correlation  was 
interpreted  as  reflecting  either  the 
preservation  of  cognitive  abilities  with 
mild  brain  structural  changes  or  insen- 
sitivity  of  the  tests  employed  for 
detecting  mild  brain  structural  changes. 
In  a  different  study,  CT  scans  and 
cerebral  blood  flow  measures  were 
obtained  in  a  sample  of  40  chronic 
alcoholic  patients,  and  the  findings 
were  related  to  results  of  neuropsycho- 
logical testing  (Nicolas  et  al.  1993). 
The  researchers  found  evidence  of  sig- 
nificant brain  hypoperfusion  in  65 
percent  of  the  alcoholics  (26  out  of 
the  40),  but  only  about  25  percent  of 
them  (11  of  the  40)  had  CT  evidence 
of  cerebral  atrophy  (mainly  in  the 


frontal  lobes).  In  this  study,  the  alco- 
holics exhibited  significant  impairments 
on  tests  of  frontal  lobe  functioning 
and  visuospatial  skills.  Frontal  lobe 
test  performance  was  independently 
related  both  to  frontal  atrophy  and  to 
frontal  hypoperfusion. 

Frontal  Brain  Systems 

Results  of  neuropsychological,  neuro- 
physiological,  and  functional  neuroimag- 
ing  studies  generally  are  compatible 
with  deficits  in  brain  systems  involving 
the  frontal  lobes  (Erbas  et  al.  1992; 
Nicolas  et  al.  1993;  Wang  et  al.  1993; 
Cohen  et  al.  1997;  Pfefferbaum  et  al. 
1997;  Zhang  et  al.  1997;  Adams  et  al. 
1998;  Hoaken  et  al.  1998;  Gansler  et 
al.  2000).  Melgaard  and  colleagues 
(1990)  measured  regional  cerebral  blood 
flow  in  alcoholic  and  nonalcoholic  men 
and  found  that  the  alcoholics  showed 
significant  flow  reduction  in  the  antero- 
mesial  frontal  region,  in  a  small  area 
of  the  left  parietal  region,  and  in  the 
mesial  part  of  the  right  occipital  lobe. 
Severity  of  alcoholism  was  related  to 
greater  flow  reduction  in  frontal  cortex 
and  in  periventricular  regions.  When 
alcoholics  were  compared  on  the  basis 
of  severity  of  intellectual  impairment, 
the  more  impaired  group  showed  a 
greater  flow  reduction  in  frontal  cortical 
and  periventricular  regions  as  well  as  in 
cortical  regions  of  the  temporal  lobes. 
Thus,  greater  flow  reduction  was  asso- 
ciated with  poorer  test  performance. 
Adams  and  colleagues  (1993)  obtained 
similar  results:  Alcoholics  showed 
hypometabolism  in  the  medial  frontal 
region.  Significant  correlations  were 
found  ( 1 )  between  frontal  lobe  metab- 
olism and  errors  on  a  test  sensitive  to 


453 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


frontal  lobe  damage  and  (2)  between 
cerebral  metabolism  and  atrophy  of 
medial  frontal  cortex  on  CT  or  MRI 
scans.  Other  studies  have  confirmed  the 
reports  of  a  correlation  between  impaired 
neuropsychological  performance  on  tests 
of  frontal  functioning  and  decreased 
frontal  lobe  perfusion  or  metabolism  in 
alcoholics  (Nicolas  et  al.  1993;  Wang 
et  al.  1993;  Adams  et  al.  1995,  1998). 
Thus,  investigators  have  observed 
expected  relationships  between  reduced 
frontal  brain  activity  and  abnormalities 
in  abilities  such  as  executive  control 
skills.  The  findings  collectively  support 
the  view  that  alcoholism  results  in 
impaired  metabolic  and  neurobehavioral 
functions  of  the  frontal  cortex  (Hoaken 
et  al.  1998).  In  addition,  frontal  lobe 
hypometabolism  may  reflect  loss  of  tis- 
sue. In  a  study  by  Erbas  and  colleagues 
(1992),  decreased  blood  flow  (mostly  in 
frontal  regions)  was  found  in  85  percent 
of  the  alcoholics  measured,  but  only 
60  percent  of  the  alcoholics  showed 
structural  changes  on  CT  scans. 

Of  interest  also  are  studies  demon- 
strating recovery  of  brain  functioning 
with  abstinence.  During  die  first  1-2 
months  of  abstinence  from  alcohol,  a 
partial  recovery  of  metabolic  or  perfu- 
sion deficits  has  been  observed,  with  the 
greatest  improvement  in  the  frontal 
lobes  (Nicolas  et  al.  1993;  Volkow  et 
al.  1994).  In  a  pilot  SPECT  study  of 
10  alcoholics  with  long  and  short 
periods  of  abstinence,  Gansler  and 
colleagues  (2000)  observed  a  positive 
relationship  between  perfusion  levels 
in  the  left  inferior  frontal  brain  region 
and  years  of  sobriety.  Alcoholics  with 
less  than  4  years  of  sobriety  had  signif- 
icantly reduced  left  inferior  frontal 


perfusion  compared  with  nonalcoholic 
control  subjects  and  alcoholics  having 
longer  periods  of  sobriety.  However, 
despite  a  significant  correlation  between 
left  inferior  frontal  perfusion  and 
memory  scores,  the  short-  and  long- 
term  sobriety  groups  did  not  differ  on 
measures  of  IQ  and  memory.  It  is  pos- 
sible that  SPECT  measures  are  indepen- 
dent of  overt  neurobehavioral  measures 
(e.g.,  see  Dupont  et  al.  1996).  In  any 
case,  the  effects  of  long-term  absti- 
nence on  frontal  lobe  perfusion  and 
corresponding  cognitive  deficits  have 
been  insufficiently  investigated. 

Hunter  and  colleagues  (1989)  mea- 
sured cerebral  blood  flow  in  Korsakoff 
patients.  Compared  with  nonalcoholic 
control  subjects,  Korsakoff  patients 
showed  a  trend  toward  reduced  blood 
flow  in  frontal  areas.  The  Korsakoff 
patients  showed  several  significant 
correlations  between  the  degree  of  flow 
reduction  in  frontal  areas  and  the  degree 
of  impairment  on  memory  and  orienta- 
tion tests  (decreased  flow  corresponded 
to  increased  impairments).  Hunter 
(1990)  noted  that  frontal  metabolic 
deficits  could  mean  that  a  normal  tissue 
mass  has  reduced  neuronal  activity,  or 
that  a  reduced  tissue  mass  has  normal 
activity  levels — or  some  of  both.  Hunter 
further  noted  that  since  some  CT  and 
neuropathological  studies  point  to 
structural  loss  of  gray  and  white  matter 
in  the  frontal  lobes  of  Korsakoff  patients, 
the  metabolic  impairment  in  this  region 
probably  at  least  in  part  reflects  reduced 
tissue  mass.  Paller  and  colleagues  (1997) 
used  PET  to  measure  regional  cerebral 
metabolism  in  Korsakoff  patients  per- 
forming a  continuous  recognition  test. 
The  investigators  found  that  Korsakoff 


454 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


patients  demonstrated  a  severe  memory 
impairment  in  delayed  recognition  and 
concomitant  widespread  decline  in 
glucose  metabolism  in  frontal,  parietal, 
and  cingulate  regions  (but  not  in  the 
hippocampus),  suggesting  that  cortical 
and  neuropsychological  abnormalities 
are  related.  Neurobehavioral  studies 
have  shown  that  Korsakoff  patients 
exhibit  clinical  signs  associated  with 
damage  to  the  frontal  cortex,  for  exam- 
ple, emotional  apathy,  disinhibition, 
and  abnormal  response  perseveration 
(Kopelman  1995;  Oscar- Berman  and 
Evert  1997).  Research  adapting  tests 
highly  sensitive  to  frontal  lobe  damage 
in  monkeys  also  supports  the  view  of 
frontal  system  dysfunction  in  alcoholic 
Korsakoff's  syndrome  (for  a  review,  see 
Oscar-Berman  and  Bardenhagen  1998). 
It  is  important  to  point  out  that  new 
discoveries  concerning  the  functioning 
of  healthy  and  diseased  frontal  brain 
systems  are  appearing  in  neuroscience 
literature  unrelated  to  alcoholism  (see 
Wickelgren  1997).  These  discoveries 
reveal  an  exquisite  cooperation  among 
various  brain  areas,  partly  under  control 
of  the  frontal  cortex;  they  provide  excit- 
ing insights  that  can  be  applied  to  the 
design  of  studies  aimed  at  under- 
standing abnormal  frontal  system 
functioning  in  alcoholism  (e.g.,  Tracy 
et  al.  1995;  Sullivan  et  al.  1997; 
Freedman  et  al.  1998). 

Right  Hemisphere 

Clinical  reports  and  experimental 
studies  have  provided  evidence  that 
each  hemisphere  of  the  human  brain 
is  important  for  mediating  different 
functions.  The  left  hemisphere  has  a 
dominant  role  in  communication  and 


in  understanding  the  spoken  and  written 
word,  and  the  right  hemisphere  plays 
a  dominant  role  in  coordinating  inter- 
actions with  the  three-dimensional 
world  around  us  (e.g.,  spatial  cognition). 
Processing  modes  of  the  two  hemi- 
spheres complement  each  other. 
Depending  on  context  and  task  demands 
(Banich  and  Heller  1998),  the  left 
hemisphere  processes  information 
analytically  and  sequentially,  whereas 
the  right  hemisphere  integrates  infor- 
mation holistically.  Differences  between 
the  two  cerebral  hemispheres  can  be 
seen  easily  in  cases  of  unilateral  brain 
damage,  and  standard  clinical  neuro- 
psychological tests  are  helpful  for  educ- 
ing the  dichotomy  (e.g.,  see  Lezak 
1995).  Patients  with  left  hemisphere 
damage  often  have  problems  with  lan- 
guage, and  patients  with  right  hemi- 
sphere damage  often  have  difficulty 
with  maps,  designs,  music,  and  other 
nonlinguistic  materials.  Of  interest  to 
this  discussion  is  the  fact  that  alcoholics 
(and  elderly  people)  have  difficulty  on 
tasks  that  resemble  those  on  which 
patients  with  damage  to  the  right 
hemisphere  also  encounter  problems. 
In  particular,  patients  with  right  hemi- 
sphere lesions,  as  well  as  alcoholic  and 
elderly  individuals,  are  disproportion- 
ately impaired  on  nonverbal  visuospa- 
tial  tasks,  as  assessed  by  Performance  IQ 
subtests  (compared  with  Verbal  IQ 
subtests)  of  the  WAIS-R  These  obser- 
vations led,  independently,  to  the  right 
hemisphere  hypothesis  in  both  the  alco- 
holism literature  and  the  aging  literature 
(Oscar-Berman  and  Schendan  in  press). 
The  right  hemisphere  hypothesis 
states  simply  that  the  right  half  of  the 
brain  is  more  vulnerable  to  the  effects 


455 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  alcoholism  (or  to  the  effects  of  aging) 
than  the  left  half  of  the  brain.  In  other 
words,  a  disproportionate  decline  in 
nonverbal,  visuospatial  functions  is 
attributed  to  a  greater  sensitivity  of 
the  right  hemisphere  to  the  neurobio- 
logical  consequences  of  alcoholism  or 
aging.  Studies  of  right  hemisphere 
contributions  to  cognitive  functions  in 
alcoholism  and  aging  have  been  reviewed 
extensively  (e.g.,  Ellis  and  Oscar- 
Berman  1989;  Oscar-Berman  1992; 
Parsons  and  Nixon  1993;  Gerhardstein 
et  al.  1998;  Oscar-Berman  and  Schendan 
in  press)  and  will  not  be  summarized 
here.  In  general,  the  results  of  most  of 
the  research  in  this  area — as  was  seen 
earlier  in  reviewing  gender  differences 
and  the  right  hemisphere — have 
provided  only  equivocal  support  for 
the  hypothesis  that  alcoholism  and 
aging  (alone  or  together)  differentially 
affect  the  functioning  of  the  two  cere- 
bral hemispheres. 

The  topic  of  right  hemisphere 
integrity  is  important  not  just  from  the 
standpoint  of  contributions  to  cognitive 
functions  (such  as  visuospatial  skills),  but 
also  because  of  the  role  of  the  right 
hemisphere  in  emotion.  Emotional 
abnormalities  such  as  those  known  to 
accompany  alcoholism  (Oscar-Berman 
et  al.  1990;  Oscar-Berman  1992; 
McGue  et  al.  1997)  result  from  lesions 
in  multiple  brain  systems  (Borod  1993; 
Gainotti  et  al.  1993;  Heller  et  al.  1998). 
Modular  models  of  emotion  combine 
concepts  from  lateral  dominance  with 
ideas  about  cortical-subcortical  inter- 
actions (Borod  1993).  Although  many 
brain  centers  act  to  take  in  emotional 
cues,  evaluate  them,  and  execute  appro- 
priate reactions,  the  right  hemisphere's 


role  in  emotional  functions  generally  is 
more  relevant  than  the  left  hemisphere's 
role,  and  the  right  hemisphere  is  more 
relevant  for  processing  negative  emo- 
tions than  for  positive  emotions  (Borod 
1993;  Gainotti  et  al.  1993;  Heller  et 
al.  1998).  Therefore,  emotional  percep- 
tion tasks  (especially  those  that  include 
stimuli  with  a  negative  valence)  can  be 
used  to  tap  right  hemisphere  abilities. 
If  alcoholics  have  deficient  right  hemi- 
sphere function,  they  should  have  more 
difficulty  perceiving  emotional  materials 
than  nonemotional  materials,  and  they 
should  have  more  difficulty  perceiving 
negative  than  positive  emotional  stimuli. 
Likewise,  if  alcoholism  interacts  with 
aging,  then  deficits  should  be  most 
pronounced  in  older  alcoholics  (com- 
pared with  their  nonalcoholic  peers). 

In  a  recent  study  (Covall  and  Oscar- 
Berman  unpublished  data),  we  used 
variations  of  the  Stroop  color-word 
interference  task  to  assess  emotional 
abilities  in  alcoholics  and  nonalcoholic 
control  subjects  across  a  wide  age 
span  (29-83  years).  We  presented 
positive  and  negative  emotional  words, 
and  neutral  words,  in  a  color-naming 
paradigm — that  is,  the  participants 
were  asked  to  report  only  the  colors 
of  the  words,  which  were  written  in 
different  colored  inks.  We  hypothesized 
that  if  there  are  changes  in  emotional 
perception  as  a  function  of  alcoholism 
and/or  aging,  then  alcoholics  (especially 
those  over  age  50)  would  show  abnor- 
mal interference  effects  for  emotional 
as  compared  with  neutral  words,  and 
negative  words  might  be  less  disruptive 
than  positive  words.  Results  showed 
expected  emotional  interference  effects 
for  all  subjects,  and  differences  in 


456 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


emotional  valence  processing  were 
found  among  older  subgroups:  older 
alcoholics  were  more  susceptible  than 
older  nonalcoholic  control  subjects  to 
the  interference  effects,  and  positive 
emotional  words  produced  a  greater 
interference  effect  than  negative  and 
neutral  words.  Thus,  the  findings  pro- 
vided some  support  for  the  right 
hemisphere  hypothesis  (as  well  as  for 
the  premature  aging  hypothesis). 

In  another  study,  Hutner  and 
Oscar-Berman  (1996)  used  a  visual 
backward -masking  perceptual  laterality 
paradigm  to  evaluate  emotional  process- 
ing abilities  in  detoxified  alcoholics 
compared  with  nonalcoholic  control 
subjects  ages  30-69.  Emotional  and 
nonemotional  words  were  presented 
tachistoscopically  to  the  Left  or  Right 
VFs,  followed  by  a  visual  masking 
stimulus.  The  research  participants 
were  asked  to  judge  the  emotional 
valence  of  each  word  (positive,  negative, 
or  neutral),  and  to  respond  verbally  or 
manually  (button  presses).  The  depen- 
dent measure  was  the  critical  inter- 
stimulus  interval  needed  to  escape  the 
backward-masking  effect.  The  alco- 
holics showed  a  significant  Right  VF 
advantage  in  both  response  mode 
conditions,  whereas  the  control  subjects 
did  not.  In  addition,  older  alcoholics 
showed  a  selective  impairment  in  pro- 
cessing negative  words.  Therefore,  once 
again  the  findings  were  in  support  of 
suggestions  that  alcoholics  (especially 
older  alcoholics)  may  have  deficient 
right  hemisphere  functions  when 
emotional  materials  are  used. 

Since  distinct  affective/cognitive 
domains,  as  well  as  multiple  brain  areas, 
can  underlie  different  emotional  tasks, 


broadly  defined  neurobehavioral  alter- 
ations can  occur  from  injury  to  only  a 
subset  of  functions.  Cognitive  neuro- 
scientific  research  has  made  notable 
progress  in  recent  years  with  the  advent 
of  new  functional  brain  imaging  tech- 
niques and  improvements  to  tradi- 
tional approaches  toward  defining  the 
functional  contribution  of  specific  brain 
systems  to  human  behavior.  These  find- 
ings with  populations  of  neurologkally 
intact  and  impaired  nonalcoholics, 
coupled  with  the  neuroanatomical, 
neuropsychological,  and  cognitive 
neuroscientific  studies  of  alcoholics, 
have  provided  additional  means  for 
more  precisely  testing  the  right  hemi- 
sphere hypothesis  (and  the  premature 
aging  hypothesis).  With  the  new  tech- 
niques available,  we  anticipate  a  suc- 
cessful future  in  understanding  the 
ways  the  cerebral  hemispheres  act  to 
integrate  and  complement  their  func- 
tions, and  a  similar  leap  in  progress  in 
pinpointing  precisely  the  neurobehav- 
ioral consequences  of  long-term 
chronic  alcoholism,  as  well  as  the  syn- 
ergism of  alcoholism  and  aging. 

Other  Brain  Systems 

Memory  loss  similar  to  the  amnesia  in 
Korsakoff  patients  has  long  been  associ- 
ated with  surgical  lesions  of  the  temporal 
lobes,  which  include  the  hippocampus 
and  the  amygdala  (Zola-Morgan  and 
Squire  1993;  Petri  and  Mishkin  1994). 
Memory  impairment  in  Korsakoff 
patients  has  also  been  associated  with 
lesions  in  basal  forebrain  and  diencephalic 
areas  (including  the  mammillary  bodies 
of  the  hypothalamus,  the  dorsomedial 
thalamic  nucleus,  and  the  fibers  connect- 
ing these  two  structures)  (Victor  1992). 


457 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


In  addition  to  memory  impairments, 
anomalies  most  probably  related  to 
limbic  system  damage  that  have  been 
studied  in  alcoholic  Korsakoff  patients 
include  emotional  changes  (Douglas 
and  Wilkinson  1993;  Hutner  and  Oscar- 
Berman  1996;  Oscar-Berman  and  Evert 
1997)  and  a  reduced  ability  to  integrate 
information  coming  in  from  more  than 
one  sense  modality  (e.g.,  cross-modal 
functions  [Oscar-Berman  et  al.  1990; 
Shaw  et  al.  1990]).  Emotional  changes 
and  multimodal  deficits  in  Korsakoff 
patients  have  been  inadequately 
researched,  however,  and  they  remain 
poorly  understood. 

In  studies  by  Jernigan  and  colleagues 
(reviewed  by  Butters  and  Jernigan  1995), 
MRI  scans  from  Korsakoff  and  non- 
Korsakoff  alcoholics  showed  that 
Korsakoff  patients  had — in  addition  to 
widespread  reductions  in  cortical  gray 
matter  volumes  and  increases  in  CSF — 
significant  reductions  in  diencephalic 
structures.  Volume  losses  in  anterior 
portions  of  the  diencephalon,  medial 
temporal  lobe  structures,  and  the  orbito- 
frontal  cortex  were  found  to  differentiate 
best  between  the  Korsakoff  and  control 
groups.  These  findings  were  used  to 
support  the  view  that  damage  to  dien- 
cephalic structures  is  involved  in  antero- 
grade amnesia  and  that  other  regions, 
such  as  the  hippocampus,  may  also  con- 
tribute to  Korsakoff  patients'  amnesic 
symptoms.  However,  in  a  different  study 
comparing  MRI  measures  of  dien- 
cephalic damage  in  alcoholics  with  and 
without  the  amnesia  of  Korsakoff  s  syn- 
drome, Blansjaar  and  colleagues  (1992) 
found  that  diencephalic  atrophy  was  of 
similar  frequency  in  both  groups. 
Based  on  these  results,  the  investigators 


suggested  that  diencephalic  lesions 
develop  regardless  of  whether  patients 
acquire  the  amnesia  of  Korsakoff  s  syn- 
drome, and  are  not  so  much  typical  of 
Korsakoff  s  syndrome  as  they  are  of 
chronic  alcoholism  and  malnutrition. 

In  regard  to  the  nature  and  location 
of  brain  damage  in  Korsakoff  s  syn- 
drome, it  is  important  to  note  that 
Korsakoff  described  neuropathology 
in  the  cerebral  cortex,  but  he  did  not 
specify  a  subcortical  locus  of  brain 
damage  for  the  syndrome  bearing  his 
name.  Rather,  he  described  a  set  of 
neuropsychological  characteristics  in 
association  with  various  etiologies 
(Victor  and  Yakovlev  1955).  The  brain 
regions  that  have  been  implicated  in 
the  syndrome  were  described  much 
later  by  researchers  using  inconsistent 
neuropsychological  criteria  to  classify 
the  patients  whose  brains  were  subse- 
quently examined  at  autopsy  (Victor 
1992;  Kopelman  1995;  Oscar-Berman 
and  Evert  1997).  Controversy  still  exists 
with  regard  to  the  critical  lesion  site(s). 
Therefore,  the  idea  that  alcoholic 
Korsakoff  s  syndrome  can  be  "neuro- 
pathologically  confirmed"  (e.g.,  Martin 
et  al.  1993;  Butterworth  1995),  without 
knowledge  and  standardization  of  ante- 
mortem  neuropsychological  symptoma- 
tology, may  be  misleading.  The  best 
approach  will  be  one  in  which  multiple 
methodologies  are  used,  namely,  neuro- 
behavioral,  neuroimaging,  and  neu- 
ropathological  approaches. 

Research  Opportunities 

Convergent  results  indicate  that  much  of 
the  brain  is  vulnerable  to  alcohol-related 
damage,  but  cortical  changes  have 
been  reported  most  consistently  across 


458 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


subgroups  of  alcoholics.  Of  the  corti- 
cal systems  that  have  been  implicated, 
frontal  and  right  hemisphere  systems 
have  received  considerable  research 
attention.  Nonetheless,  many  topics 
remain  poorly  explored  and  are  in 
need  of  future  research.  Some  of  these 
topics  are  (1)  the  manner  in  which 
disruption  of  specific  subsystems  of 
frontal  brain  regions  contributes  to 
impairments  in  attention,  perception, 
emotional  functioning  (and  regulation 
of  drinking),  and  executive  functioning; 
(2)  the  ways  in  which  atrophy  of  the 
corpus  callosum  interferes  with  inter- 
hemispheric  communication  and/or 
normal  cerebral  asymmetries;  (3)  char- 
acterization of  deficits  in  memory  and 
emotional  functioning  resulting  from 
damage  to  limbic  and  diencephalic 
structures  (and  how  they  differ  from 
frontal-induced  deficits);  (4)  the  dif- 
ferential contributions  of  cerebellar 
damage  to  motor  versus  cognitive 
impairments;  and  (5)  the  nature  of 
abnormalities  in  specific  neurotrans- 
mitter systems. 

To  understand  the  etiologies  and 
mechanisms  of  brain  damage  across 
subgroups  of  brain-impaired  alcoholics, 
future  studies  should  use  multiple 
methodologies  such  as  structural  and 
functional  imaging  techniques,  neuro- 
pathological  studies,  electrophysiolog- 
ical studies,  and  neurobehavioral 
studies  (Rabbitt  1997;  Wickelgren 
1997;  Liu  1998;  Oscar-Berman  and 
Bardenhagen  1998).  In  many  instances, 
diagnostic  procedures  for  establishing 
alcoholic  functional  impairments  vary 
across  studies.  Alcohol-induced  demen- 
tia, for  example,  is  a  controversial  dis- 
order because  investigators  differ  with 


respect  to  the  deficits  that  should  be 
included  when  defining  its  clinical  pre- 
sentation (Smith  and  Atkinson  1995). 
Likewise,  there  is  lack  of  consensus 
regarding  criteria  for  classifying 
patients  with  Wernicke -Korsakoff  syn- 
drome. At  the  outset,  therefore,  it  is 
important  to  establish  uniform  positive 
diagnostic  criteria  for  subtypes  of 
brain-impaired  alcoholics;  this  can  be 
approached  with  the  aid  of  sophisti- 
cated neurological  and  neurobehav- 
ioral techniques. 

SUMMARY  AND 
CONCLUSIONS 

Neuropsychological  impairments  result 
from  prolonged  ingestion  of  alcohol 
in  vulnerable  populations.  I  reviewed 
research  findings  on  three  factors  that 
make  certain  alcoholics  susceptible  to 
behavioral  and  brain  changes:  chrono- 
logical aging,  gender,  and  nutritional 
deficiencies.  A  convergence  of  the  evi- 
dence supports  the  view  that  older 
alcoholics  are  especially  at  risk  for  the 
deleterious  effects  of  alcohol  on  the 
brain,  but  women  are  not  consistentiy 
found  to  be  vulnerable.  However, 
older  alcoholic  women  have  not  been 
studied  sufficiently.  Future  research  can 
capitalize  on  normal  gender- dimorphic 
differences  in  brain  organization  (e.g., 
gender  differences  in  hemispheric  asym- 
metries) to  address  questions  such  as 
whether  and  how  alcoholism  affects 
perception,  attention,  emotion,  and 
other  neurobehavioral  functions,  as 
well  as  gender  effects  in  association 
with  aging.  Malnutrition — especially 
thiamine  deficiency — seems  to  con- 
tribute to  limbic  and  basal  forebrain 


459 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


lesions  leading  to  severe  cognitive  deficits  unknown.  Similarly,  it  is  important  to 

in  Wernicke -Korsakoff  syndrome,  but  clarify  the  etiology  and  consequences 

the  critical  lesions  and  the  underlying  of  cerebellar  degeneration.  Research  is 

neuropathological  mechanisms  remain  needed  to  explicate  the  separate  and 


Table  3.  Important  Research  Questions  and  Topics. 


How  do  individual  differences  (subject  characteristics)  contribute  to  alcohol's  effects  on 
neuropsychological  functioning? 

Age 

Investigate  adolescents 

Investigate  elderly  (including  women) 
Gender 

Use  sexually  dimorphic  human  characteristics  (e.g.,  hemispheric  differences) 

Compare  human  and  nonhuman  animal  models 
Nutrition  vs.  toxicity 

Clarify  roles  in  Wernicke's  encephalopathy,  KorsakofFs  syndrome,  cerebellar  degeneration, 

and  Marchiafava-Bignami  disease 

Evaluate  nutritional  therapies  (e.g.,  thiamine  fortification) 
Drinking  history 

Study  amount  and  type  of  alcoholic  beverages  consumed,  and  duration  of  alcoholism 

Consider  duration  of  abstinence 
Family  history 

Investigate  genetic  factors 

Investigate  drinking  habits 
Comorbidity:  Evaluate  the  combined  effects  of  alcoholism  and 

Polysubstance  abuse 

Psychiatric  comorbidities 

Medical  problems  (e.g.,  HIV  and  liver  disease) 

Head  injury 

Which  individual  differences  (subject  characteristics)  influence  recovery  of  structural 
and/or  functional  integrity? 

Age  (when  tested  and  at  onset  of  drinking) 

Gender 

Nutrition 

Drinking  history 

Family  history 

Comorbidity 

What  are  the  similarities  and  differences  between  the  residual  and  the  acute  effects  of 
alcoholism? 

How  do  they  parallel  each  other  (e.g.,  brain  dysfunction  and  brain  damage)? 
How  do  they  contribute  to  abusive  drinking  (e.g.,  risky  behaviors,  disinhibitions)? 

Are  animal  models  sensitive  to  problems  associated  with  human  alcoholism? 

Are  the  behavioral  paradigms  comparable  across  species  for  measuring  alcohol's  effects? 
Are  "lesion  effects"  comparable  across  species? 
Are  gender  effects  comparable  across  species? 


460 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


combined  roles  of  malnutrition  and 
ethanol  neurotoxicity  to  brain  damage 
and  functional  impairments  in  these 
devastating  disorders,  as  well  as  in 
alcoholic  dementia.  A  prerequisite  to 
research  on  severely  impaired  alcoholic 
patients,  however,  is  the  adoption  of 
uniform  terminology  for  defining  the 
disorders  and  syndromes  under  study. 
Methodological  standardization  also 
is  critical. 

Although  alcoholics  have  diffuse 
cortical  damage  that  affects  functioning 
of  the  two  cerebral  hemispheres,  no  def- 
inite relationships  have  been  established 
between  alcoholism-related  damage  to 
specific  cortical  areas  and  concurrent 
cognitive  impairments.  However,  find- 
ings from  neuroimaging  and  neuro- 
pathological  studies  point  to  increased 
susceptibility  of  frontal  brain  systems. 
In  addition,  while  alcoholics'  deficits 
in  visuospatial  functions  persistently 
have  implicated  right  hemisphere  vul- 
nerability, neuropsychological  and 
neuroanatomies  evidence  for  unilateral 
damage  is  not  convincing.  Research 
on  cognitive  capabilities  and  differences 
between  the  hemispheres  in  alcoholics 
has  provided  only  limited  support  for 
the  premature  aging  hypothesis,  and 
even  less  consistent  support  for  the 
right  hemisphere  hypothesis. 

Future  research  would  benefit  greatly 
from  cooperation  among  disciplines. 
Behavioral  neuroscience  offers  excel- 
lent techniques  for  sensitively  accessing 
distinct  cognitive  and  emotional  func- 
tions. Electrophysiological  and  neu- 
roimaging techniques  provide  a 
window  on  the  active  brain,  as  well  as 
a  glimpse  at  regions  with  structural 
damage.  Likewise,  followup  post- 


mortem examination  of  brains  of  well - 
studied  alcoholic  individuals  should 
be  encouraged  for  clues  about  neuro- 
transmitter abnormalities,  as  well  as 
analyses  of  injury  at  the  cellular  level. 
Specific  questions  and  topics  in  need 
of  research  attention  are  outlined  in 
table  3.  They  include  the  following: 
neurobehavioral  and  brain  func- 
tional/structural recovery  with  absti- 
nence, the  extent  of  multimodal 
sensory  and  perceptual  deficits,  the 
nature  of  the  loss  of  emotional  and 
motivational  functioning  among  sub- 
groups of  alcoholics,  the  synergistic 
influence  of  common  comorbid  con- 
ditions and  alcoholism,  and  the  direct 
contribution  of  frontal  system  dys- 
function to  alcohol  consumption  and 
alcoholic  symptomatology. 

ACKNOWLEDGMENTS 

The  writing  of  this  report  was  sup- 
ported by  NIAAA  grants  R37- 
AA07112  and  K05-AA00219,  and  by 
funds  from  the  Medical  Research 
Service  of  the  U.S.  Department  of 
Veterans  Affairs. 


REFERENCES 

Adams,  K.M.;  Gilman,  S.;  Koeppe,  R.A.; 
Kluin,  K.J.;  Brunberg,  J.A.;  Dede,  D.; 
Berent,  S.;  and  Kroll,  P.D.  Neuro- 
psychological deficits  are  correlated 
with  frontal  hypometabolism  in  positron 
emission  tomography  studies  in  older 
alcoholic  patients.  Alcohol  Clin  Exp  Res 
17(2):205-210,  1993. 

Adams,  K.M.;  Gilman,  S.;  Koeppe,  R.A.; 
Kluin,  K.J.;  Junck,  L.;  Lohman,  M.; 
Johnson-Greene,  D.;  Berent,  S.;  Dede, 


461 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


D.;  and  Kroll,  P.D.  Correlation  of  neu- 
ropsychological function  with  cerebral 
metabolic  rate  in  subdivisions  of  the 
frontal  lobes  of  older  alcoholic  patients 
measured  with  [18F]fluorodeoxyglucose 
and  positron  emission  tomography. 
Neuropsychology  9:275-280, 1995. 

Adams,  K.M.;  Gilman,  S.;  Johnson- 
Greene,  D.;  Koeppe,  R.A.;  Junck,  L.; 
Kluin,  K.J.;  Martorello,  S.;  Johnson,  M.J.; 
Heumann,  M.;  and  Hill,  E.  The  signifi- 
cance of  family  history  status  in  relation  to 
neuropsychological  test  performance  and 
cerebral  glucose  metabolism  studied  with 
positron  emission  tomography  in  older 
alcoholic  patients.  Alcohol  Clin  Exp  Res 
22(1):105-210,  1998. 

Banich,  M.T.,  and  Heller,  W.,  eds.  Evolving 
Perspectives  on  Lateralization  of  Function. 
[Special  Issue.]  Curr  Dir  Psychol  Sci,  Vol. 
7,  No.  1,1998. 

Bates,  M.E.,  and  Convit,  A.  Neuropsychology 
and  neuroimaging  of  alcohol  and  illicit 
drug  abuse.  In:  Calev,  A.,  ed.  The  Assessment 
of  Neuropsychological  Functions  in  Psychiatric 
Disorders.  Washington,  DC:  American 
Psychiatric  Press,  1999.  pp.  373-445. 

Beatty,  W.W.;  Katzung,  V.M.;  Nixon, 
S.J.;  and  Moreland,  V.J.  Problem-solving 
deficits  in  alcoholics:  Evidence  from  the 
California  Card  Sorting  Test.  /  Stud 
Alcohol  54(6):687-692,  1993. 

Beauregard,  M.;  Chertkow,  H.;  Gold,  D.; 
Karama,  S.;  Benhamou,  J.;  Babins,  L.; 
and  Faucher,  A.  Word  priming  with  brief 
multiple  presentation  technique:  Preservation 
in  amnesia.  Neuropsychologia  35(5):611- 
621,  1997. 

Belzunegui,  T.;  Insausti,  R.;  Ibanez,  J.; 
and  Gonzalo,  L.M.  Effect  of  chronic 
alcoholism  on  neuronal  nuclear  size  and 
neuronal  population  in  the  mammillary 
body  and  the  anterior  thalamic  complex 


of  man.  Histol  Histopathol  10(3):633- 
638,  1995. 

Blansjaar,  B.A.;  Vielvoye,  G.J.;  van  Dijk, 
J.G.;  and  Rijnders,  RJ.  Similar  brain 
lesions  in  alcoholics  and  Korsakoff 
patients:  MRI,  psychometric,  and  clinical 
findings.  Clin  Neurol  Neurosurg 
94(3):197-203,  1992. 

Blass,  J. P.,  and  Gibson,  G.E.  Abnormality 
of  a  thiamine-requiring  enzyme  in  patients 
with  Wernicke-Korsakoff  syndrome.  N 
Engl  J  Med  297(25):  1367-1 370,  1977. 

Borod,  J.C.  Cerebral  mechanisms  under- 
lying facial,  prosodic,  and  lexical  emotional 
expression:  A  review  of  neuropsychologi- 
cal studies  and  methodological  issues. 
Neuropsychology  7:445^63,  1993. 

Bowden,  S.C.  Separating  cognitive  im- 
pairment in  neurologically  asymptomatic 
alcoholism  from  Wernicke-Korsakoff 
syndrome:  Is  the  neuropsychological 
distinction  justified?  Psychol  Bull  107: 
355-366,  1990. 

Bowden,  S.C,  and  McCarter,  RJ.  Spatial 
memory  in  alcohol-dependent  subjects: 
Using  a  push-button  maze  to  test  the 
principle  of  equiavailability.  Brain  Cogn 
22(l):51-62,  1993. 

Bowden,  S.C;  Benedikt,  R.;  and  Ritter, 
A.J.  Delayed  matching  to  sample  and 
concurrent  learning  in  nonamnesic 
humans  with  alcohol  dependence. 
Neuropsychologia  30(5):427^35,  1992. 

Brunfaut,  E.,  and  d'Ydewalle,  G.  A  compar- 
ison of  implicit  memory  tasks  in  Korsakoff 
and  alcoholic  patients.  Neuropsychologia 
34(12):1143-1150,  1996. 

Butters,  N.,  and  Jernigan,  T.  Cognitive 
and  morphometric  features  of  long-term 
alcoholics  with  and  without  Korsakoff's 
syndrome.  Paper  presented  at  the  Annual 


462 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


Scientific  Meeting,  Research  Society  on 
Alcoholism,  Steamboat  Springs,  CO,  1995. 

Butterworth,  R.F.  Pathophysiology  of 
alcoholic  brain  damage:  Synergistic  effect 
of  ethanol,  thiamine  deficiency  and  alco- 
holic liver  disease.  Metab  Brain  Dis 
10:1-8,  1995. 

Butterworth,  R.F.;  Kril,  J.J.;  and  Harper, 
C.G.  Thiamine-dependent  enzyme  changes 
in  the  brains  of  alcoholics:  Relationship  to 
the  Wernicke-Korsakoff  syndrome.  Alcohol 
Clin  Exp  Res  17:1084-1088,  1993. 

Casanova,  M.F.  Wernicke's  disease  and 
schizophrenia:  A  case  report  and  review  of 
the  literature.  Int  J  Psychiatry  Med  26(3): 
319-328,  1996. 

Christiansen,  K.,  and  Knussmann,  R.  Sex 
hormones  and  cognitive  functioning  in 
men.  Neuropsycholobiology  18:27-36,  1987. 

Coffey,  C.E.;  Lucke,  J.F.;  Saxton,  J.A.; 
Ratcliff,  G.;  Unitas,  L.J.;  Billig,  B.;  and 
Bryan,  R.N.  Sex  differences  in  brain  ag- 
ing. A  quantitative  magnetic  resonance 
imaging  study.  Arch  Neurol  55(2):169- 
179,  1998. 

Cohen,  H.L.;  Porjesz,  B.;  Begleiter,  H.; 
and  Wang,  W.  Neurophysiologies  corre- 
late of  response  production  and  inhibition 
in  alcoholics.  Alcohol  Clin  Exp  Res  2l(%): 
1398-1406,  1997. 

Connolly,  L.,  and  Price,  J.  Preventing  the 
Wernicke-Korsakoff  syndrome  in  Australia: 
Cost-effectiveness  of  thiamin- supplemen- 
tation alternatives.  Aust  N Z J Public 
Health  20:181-187,  1996. 

Cook,  C.C.H.;  Hallwood,  P.M.;  and 
Thomson,  A.D.  B  vitamin  deficiency  and 
neuropsychiatric  syndromes  in  alcohol  misuse. 
Alcohol  Alcohol  33(4):317-336,  1998. 


Courville,  C.B.  Effects  of  Alcohol  on  the 
Nervous  System  of  Man.  2d  ed.  Los 
Angeles:  San  Lucas  Press,  1996. 

Cullen,  KM.,  and  Halliday,  G.M.  Neuro- 
fibrillary tangles  in  chronic  alcoholics. 
Neuropathol  Appl  Neurobiol  2 1  (4 ):  3 1 2- 
318,1995. 

Cullen,  KM.;  Halliday,  G.M.;  Caine,  D.; 
and  Kril,  J.J.  The  nucleus  basalis  (Ch4)  in 
the  alcoholic  Wernicke -Korsakoff  syndrome: 
Reduced  cell  number  in  both  amnesic  and 
non-amnesic  patients.  J  Neurol  Neurosurg 
Psychiatry  63(3):315-320,  1997. 

Davila,  M.D.;  Shear,  P.K;  Lane,  B.;  Sullivan, 
E.V.;  and  Pfefferbaum,  A.  Mammillary  body 
and  cerebellar  shrinkage  in  chronic  alco- 
holics. Neuropsychology  8:433^44,  1994. 

Di  Sclafani,  V.;  Ezekiel,  F.;  Meyerhoff, 
D.J.;  MacKay,  S.;  Dillon,  W.P.;  Weiner, 
M.W.;  and  Fein,  G.  Brain  atrophy  and 
cognitive  function  in  older  abstinent 
alcoholic  men.  Alcohol  Clin  Exp  Res  19(5): 
1121-1126,  1995. 

Douglas,  J.J.,  and  Wilkinson,  DA.  Evidence 
of  normal  emotional  responsiveness  in 
alcoholic  Korsakoff  s  syndrome  in  the 
presence  of  profound  memory  impair- 
ment. Addiction  88:1637-1645,  1993. 

Drake,  A. I.;  Hannay,  H.  J.;  and  Gam,  J. 
Effects  of  chronic  alcoholism  on  hemispheric 
functioning:  An  examination  of  gender 
differences  for  cognitive  and  dichotic 
listening  tasks.  /  Clin  Exp  Neuropsychol 
12(5):781-797,  1990. 

Dufour,  M.C.  Risks  and  benefits  of  alcohol 
use  over  the  life  span.  Alcohol  Health  Res 
Wor/tf  20(3):145-151,  1996. 

Dupont,  R.M.;  Rourke,  S.B.;  Grant,  I.; 
Lehr,  P.P.;  Reed,  R.J.;  Challakere,  K; 
Lamoureux,  G.;  and  Halpern,  S. 
Single  photon  emission  computed 
tomography  with  iodoamphetamine-123 


463 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  neuropsychological  studies  in  long- 
term  abstinent  alcoholics.  Psychiatry  Res 
Neuroimaging  67:99-1 1 1 ,  1996. 

Eichenbaum,  H.  Declarative  memory: 
Insights  from  cognitive  neurobiology. 
A nnu  Rev  Psychol 48: 547-5 72,  1997. 

Ellis,  R.J.  Dichotic  asymmetries  in  aging 
and  alcoholic  subjects.  Alcohol  Clin  Exp 
it«14(6):863-871,1990. 

Ellis,  R.J.,  and  Oscar-Berman,  M.  Alcohol- 
ism, aging,  and  functional  cerebral  asym- 
metries. Psychol  Bull  106:128-147,  1989. 

Emsley,  R;  Smith,  R.;  Roberts,  M.; 
Kapnias,  S.;  Pieters,  H.;  and  Maritz,  S. 
Magnetic  resonance  imaging  in  alcoholic 
Korsakoff  syndrome:  Evidence  for  an 
association  with  alcoholic  dementia. 
Alcohol  Alcohol  3l(5)A79-486,  1996. 

Erbas,  B.;  Bekdik,  C;  Erbengi,  G.;  Eniinlii, 
T.;  Aytac,  S.;  Kummbasar,  H.;  and  Dogan, 
Y.  Regional  cerebral  blood  flow  changes 
in  chronic  alcoholism  using  Tc-99m 
HMPAO  SPECT.  Comparison  with  CT 
parameters.  Clin  Nucl Med  17(2):123- 
127,  1992. 

Errico,  A.L.;  Parsons,  O.A.;  Kling,  O.R.; 
and  King,  A.C.  Investigation  of  the  role 
of  sex  hormones  in  alcoholics'  visuospatial 
deficits.  Neuropsychologia  30(5):417- 
426,  1992. 

Estruch,  R;  Nicolas,  J.M.;  Salamero,  M.; 
Aragon,  C;  Sacanella,  E.;  Fernandez-Sola, 
J.;  and  Urbano-Marquez,  A.  Atrophy  of 
the  corpus  callosum  in  chronic  alcoholism. 
J  Neurol  Sci  146(2):145-151,  1997. 

Evert,  D.L.,  and  Oscar-Berman,  M.  Alcohol- 
related  cognitive  impairments:  An  overview 
of  how  alcoholism  may  affect  the  workings 
of  the  brain.  Alcohol  Health  Res  World 
19(2):89-96,  1995. 


Freedman,  M.;  Black,  S.;  Ebert,  P.;  and 
Binns,  M.  Orbitofrontal  function,  object 
alternation  and  perseveration.  Cereb 
Cortex  8:18-27,  1998. 

Gainotti,  G.;  Caltagirone,  C.;  and 
Zocolotti,  P.  Left/ right  and  cortical/sub- 
cortical  dichotomies  in  the  neuropsycho- 
logical study  of  human  emotions.  Cogn 
Emotion  7:71-93,  1993. 

Galanter,  M.,  ed.  Recent  Developments  in 
Alcoholism,  Vol.  14:  The  Consequences  of 
Alcoholism:  Medical,  Neuropsychiatry 
Economic,  Cross-Cultural.  New  York: 
Plenum,  1998. 

Gallagher,  M.,  and  Rapp,  P.R.  The  use  of 
animal  models  to  study  the  effects  of 
aging  on  cognition.  Annu  Rev  Psychol 
48:339-370,  1997. 

Gansler,  DA.;  Harris,  G.J.;  Oscar-Berman, 
M.;  Streeter,  C.;  Lewis,  R.F.;  Ahmed,  I.; 
and  Achong,  D.  Hypoperfusion  of  infe- 
rior frontal  brain  regions  in  abstinent 
alcoholics:  A  pilot  SPECT  study.  J  Stud 
Alcohol 6l(l):32-37,  2000. 

Gerhardstein,  P.;  Peterson,  M.A.;  and 
Rapcsak,  S.Z.  Age-related  hemispheric 
asymmetry  in  object  discrimination.  J  Clin 
Exp  Neuropsychol 20(2):  174-1 85,  1998. 

Gilman,  S.;  Adams,  KM.;  Johnson-Greene, 
D.;  Koeppe,  R.A.;  Junck,  L.;  Kluin,  K.J.; 
Martorello,  S.;  Heumann,  N.;  and  Hill,  E. 
Effects  of  disulfiram  on  positron  emission 
tomography  and  neuropsychological  stud- 
ies in  severe  chronic  alcoholism.  Alcohol 
Clin  Exp  it«20(8):1456-1461,  1996. 

Glenn,  S.W.  Sex  differences  in  alcohol- 
induced  brain  damage.  In:  Hunt,  W.A., 
and  Nixon,  S.J.,  eds.  Alcohol-Induced 
Brain  Damage.  National  Institute  on 
Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Publication 


464 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


No.  93-3549.  Rockville,  MD:  the  Institute, 
1993. pp.  195-212. 

Harding,  A.J.;  Wong,  A.;  Svoboda,  M.;  Kril, 
J.J.;  and  Halliday,  G.M.  Chronic  alcohol 
consumption  does  not  cause  hippocampal 
neuron  loss  in  humans.  Hippocampus 
7(l):78-87,  1997. 

Harper,  C.  The  neuropathology  of  alcohol- 
specific  brain  damage,  or  does  alcohol 
damage  the  brain?  /  Neuropathol  Exp 
Neurol  57 '(2):101-110,  1998. 

Harris,  G.;  Oscar-Berman,  M.;  Gansler, 
D.A.;  Streeter,  C;  Lewis,  R.F.;  Ahmed, 
I.;  and  Achong,  D.  Hypoperfusion  of  the 
cerebellum  and  aging  effects  on  cerebral 
cortex  blood  flow  in  abstinent  alcoholics: 
A  SPECT  study.  Alcohol  Clin  Exp  Res 
23(7):1219-1227,  1999. 

Heller,  W.;  Nitschke,  J.B.;  and  Miller,  GA. 
Lateralization  in  emotion  and  emotional 
disorders.  Curr  Dir  Psychol  Sci  7(  1  ):26- 
32, 1998. 

Hill,  S.Y.,  and  Steinhauer,  S.K  Event-related 
potentials  in  women  at  risk  for  alcoholism. 
Alcohol  10:349-354,  1993. 

Hiscock,  M.;  Inch,  R;  Jacek,  C;  and 
Hiscock-Kalil,  C.  Is  there  a  sex  difference 
in  human  laterality?  I.  An  exhaustive 
survey  of  auditory  laterality  studies  from 
six  neuropsychology  journals.  /  Clin  Exp 
Neuropsychol  16(3):423^35,  1994. 

Hiscock,  M.;  Israelian,  M.;  Inch,  R.;  Jacek, 
C;  and  Hiscock-Kalil,  C.  Is  there  a  differ- 
ence in  human  laterality?  II.  An  exhaus- 
tive survey  of  visual  laterality  studies  from 
six  neuropsychology  journals.  /  Clin  Exp 
Neuropsychol  17(4):590-610,  1995. 

Hoaken,  P.N.S.;  Giancola,  P.R.;  and  Phil, 
RO.  Executive  cognitive  functions  as 
mediators  of  alcohol-related  aggression. 
Alcohol  Alcohol  SS(1)A7-S4,  1998. 


Hommer,  D.;  Momenan,  R;  Rawlings, 
R;  Ragan,  P.;  Williams,  W.;  Rio,  D.;  and 
Eckardt,  M.  Decreased  corpus  callosum 
size  among  alcoholic  women.  Arch  Neurol 
53(4):359-363,  1996. 

Hunter,  R  Frontal  metabolic  deficits  in 
Korsakoff  syndrome .  Br  J  Psychiatry 
157(9):454^55,  1990. 

Hunter,  R;  McLuskie,  R;  Wyper,  D.; 
Patterson,  J.;  Christie,  J.E.;  Brooks,  D.N.; 
McCulloch,  J.;  Fink,  G.;  and  Goodwin, 
G.M.  The  pattern  of  function-related 
regional  cerebral  blood  flow  investigated 
by  single  photon  emission  tomography 
with  99mTc-HMPAO  in  patients  with 
presenile  Alzheimer's  disease  and 
Korsakoff's  psychosis.  Psychol  Med 
19(4):847-855,  1989. 

Hutner,  N.,  and  Oscar-Berman,  M.  Visual 
laterality  patterns  for  the  perception  of 
emotional  words  in  alcholic  and  aging 
individuals .  J  Stud  Alcohol  5  7  ( 2 ) :  1 44- 
154,  1996. 

Ivry,  R.  Functional  hypotheses  regarding 
cerebellar  contributions  to  cognition. 
Paper  presented  at  the  Tenth  Annual 
Meeting  of  the  Memory  Disorders 
Research  Society,  Cambridge,  MA,  1998. 

Jernigan,  T.L.;  Butters,  N.;  DiTraglia,  G.; 
Schafer,  K;  Smith,  T.;  Irwin,  M.;  Grant, 
I.;  Schuckit,  M.;  and  Cermak,  L.S.  Reduced 
cerebral  grey  matter  observed  in  alcoholics 
using  magnetic  resonance  imaging.  Alcohol 
Clin  Exp  Res  15(3):418-427,  1991a. 

Jernigan,  T.L.;  Schafer,  K;  Butters,  N.;  and 
Cermak,  L.S.  Magnetic  resonance  imaging 
of  alcoholic  Korsakoff  patients.  Neuropsycho- 
pharmacology  4(3):175-186,  1991&. 

Joyce,  E.M.  Aetiology  of  alcoholic  brain 
damage:  Alcoholic  neurotoxicity  or 
thiamine  malnutrition?  Br  Med  Bull 
50(1):99-114,  1994. 


465 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Joyce,  E.M.,  and  Robbins,  T.W.  Frontal 
lobe  function  in  Korsakoff  and  non-Korsakoff 
alcoholics:  Planning  and  spatial  working 
memory.  Neuropsycholqgia  29(8):709- 
723,  1991. 

Keefe,  K.,  and  Newcomb,  M.D.  Demo- 
graphic and  psychosocial  risk  for  alcohol 
use:  Ethnic  differences.  J  Stud  Alcohol 
57(5):521-530,  1996. 

Kopelman,  M.D.  The  Korsakoff" syndrome. 
Br  J  Psychiatry  166(2):154-173,  1995. 

Kril,  J.J.  The  contribution  of  alcohol, 
thiamine  deficiency  and  cirrhosis  of  the 
liver  to  cerebral  cortical  damage  in  alcoholics. 
Metab  Brain  Dis  10(1):9-16,  1995. 

Kril,  J.J.;  Halliday,  G.M.;  Svoboda,  M.D.; 
and  Cartwright,  H.  The  cerebral  cortex  is 
damaged  in  chronic  alcoholics.  Neuroscience 
79(4):993-998,  1997. 

Kroft,  C.L.;  Gescuk,  B.;  Woods,  B.T.; 
Mello,  N.K.;  Weiss,  R.D.;  and  Mendelson, 
J.H.  Brain  ventricular  size  in  female  alco- 
holics: An  MRI  study.  Alcohol  Clin  Exp 
ite8(l):31-34, 1991. 

Laas,  R.,  and  Hagel,  C.  Hirano  bodies 
and  chronic  alcoholism.  Neuropathol  Appl 
Neurobiol  20(l):l2-21,  1994. 

Lancaster,  F.E.  Gender  differences  in  the 
brain.  Implications  for  the  study  of  human 
alcoholism.  Alcohol  Clin  Exp  Res  18(3):740- 
746,  1994. 

Lancaster,  F.E.  Gender  differences  in 
animal  studies.  Implications  for  the  study 
of  human  alcoholism.  Recent  Dev  Alcohol 
12:209-215,  1995. 

Langlais,  P.J.  Alcohol-related  thiamine 
deficiency:  Impact  on  cognitive  and  mem- 
ory functioning.  Alcohol  Health  Res  World 
19(2):113-121, 1995. 

Lavoie,  J.,  and  Butterworth,  R.F.  Reduced 
activities  of  thiamine -dependent  enzymes 


in  brains  of  alcoholics  in  the  absence  of 
Wernicke's  encephalopathy.  Alcohol  Clin 
£j^jR«19(4):1073-1077,  1995. 

Lezak,  M.D.  Neuropsychological  Assessment. 
3d  ed.  New  York:  Oxford  University 
Press,  1995. 

Lieber,  C.S.  Seminars  in  medicine  of  the 
Beth  Israel  Hospital,  Boston:  Medical 
disorders  of  alcoholism.  N  Engl  J  Med 
333(16):1058-1065,  1995. 

Lishman,  WA.  Organic  Psychiatry.  3d  ed. 
Oxford,  England:  Blackwell  Scientific 
Publications,  1998. 

Lishman,  W.A.;  Jacobson,  R.R.;  and 
Acker,  C.  Brain  damage  in  alcoholism: 
Current  concepts.  Acta  Med  Scand  Suppl 
717:5-17,  1987. 

Liu,  Y.  Approaches  for  studying  neural 
circuits:  Application  to  alcohol  research. 
Alcohol  Clin  Exp  Res  22(1):  1-2,  1998. 

Ma,  J.J.,  and  Truswell,  A.S.  Wernicke- 
Korsakoff  syndrome  in  Sydney  hospitals: 
Before  and  after  thiamin  enrichment  of 
flour.  MedJAustl63(l0):53l-534, 1995. 

Mann,  K;  Batra,  A.;  Giinthner,  A.;  and 
Schroth,  G.  Do  women  develop  alcoholic 
brain  damage  more  readily  than  men?  Alcohol 
Clin  Exp  Res  16(6):1052-1056,  1992. 

Marsano,  L.  Alcohol  and  malnutrition. 
Alcohol  Health  Res  World  17(4):284- 
291,  1993. 

Martin,  P.R;  McCool,  B  A.;  and  Singleton, 
C.K  Genetic  sensitivity  to  thiamine  defi- 
ciency and  development  of  alcoholic  organic 
brain  disease.  Alcohol  Clin  Exp  Res  17:31- 
37,  1993. 

Mayes,  A.R.  Memory  and  amnesia.  Behav 
Brain  Res 66(l-2):29-36,  1995. 

McCool,  B.A.;  Plonk,  S.G.;  Martin,  P.R.; 
and  Singleton,  C.K.  Cloning  of  human 


466 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


transketolase  cDNAs  and  comparison  of 
the  nucleotide  sequence  of  the  coding 
region  in  Wernicke -Korsakoff  and  non- 
Wernicke- Korsakoff  individuals.  /  Biol 
Chem  268:1397-1404,  1993. 

McGlinchy-Berroth,  R.;  Cermak,  L.S.; 
Carrillo,  M.C.;  Armfield,  S.;  Gabrieli, 
J.D.;  and  Disterhoft,  J.F.  Impaired  delay 
eyeblink  conditioning  in  amnesic  Korsakoff 
patients  and  recovered  alcoholics.  Alcohol 
Clin  Exp  Res  19(5):1127-1132,  1995. 

McGue,  M.;  Slutske,  W.;  Taylor,  J.;  and 
Iacono,  W.G.  Personality  and  substance 
use  disorders:  Effects  of  gender  and  alco- 
holism subtype.  Alcohol  Clin  Exp  Res 
21(3):513-520,  1997. 

Meinz,  E.J.,  and  Salthouse,  T.A.  Is  age 
kinder  to  females  than  to  males? 
Psychonomic  Bull  Rev  5(l):56-70,  1998. 

Melgaard,  B.;  Henriksen,  L.;  Ahlgren,  P.; 
Danielsen,  U.T.;  Sorensen,  H.;  and 
Paulson,  O.B.  Regional  cerebral  blood 
flow  in  chronic  alcoholics  measured  by 
single  photon  emission  computerized 
tomography.  Acta  Neurol  Scand  82(  1  ):87- 
93,  1990. 

Muramatsu,  T.;  Kato,  M.;  Matsui,  T.; 
Yoshimasu,  H.;  Yoshino,  A.;  Matsushita, 
S.;  Higuchi,  S.;  and  Kashima,  H.  Apolipo- 
protein  E  epsilon  4  allele  distribution  in 
Wernicke-Korsakoff  syndrome  with  or 
without  global  intellectual  deficits.  J 
Neural  Transm  1 04( 8-9 ):9 13-920,  1997. 

National  Institute  on  Alcohol  Abuse  and 
Alcoholism.  Ninth  Special  Report  to  the 
U.S.  Congress  on  Alcohol  and  Health.  NIH 
Pub.  No.  97^017.  Bethesda,  MD: 
National  Institutes  of  Health,  1997. 

Neiman,  J.  Alcohol  as  a  risk  factor  for 
brain  damage:  Neurologic  aspects.  Alcohol 
Clin  Exp  it«22(7):346S-351S,  1998. 


Nicolas,  J.M.;  Catafau,  A.M.;  Estruch,  R; 
Lomeha,  F.J.;  Salamero,  M.;  Herranz,  R; 
Monforte,  R.;  Cardenal,  C.;  and  Urbano- 
Marquez,  A.  Regional  cerebral  blood 
flow-SPECT  in  chronic  alcoholism: 
Relation  to  neuropsychological  testing.  / 
Nucl  Med  34(9):1452-1459,  1993. 

Nicolas,  J.M.;  Estruch,  R.;  Salamero,  M.; 
Orteu,  N.;  Fernandez-Sola,  J.;  Sacanella, 
E.;  and  Urbano-Marquez,  A.  Brain  im- 
pairment in  well-nourished  chronic  alcoholic 
is  related  to  ethanol  intake.  Ann  Neurol 
41(5):590-598,  1997. 

Nixon,  S.J.  Typologies  in  women.  Recent 
Dev  Alcohol  11:305-323,  1993. 

Nixon,  S.J.;  Paul,  R;  and  Phillips,  M. 
Cognitive  efficiency  in  alcoholics  and 
polysubstance  abusers.  Alcohol  Clin  Exp 
it«22(7):1414-1420,  1998. 

Nussbaum,  P.D.,  ed.  Handbook  of 
Neuropsychology  and  Aging.  New  York: 
Plenum,  1997. 

Oscar-Berman,  M.  Alcoholism  and  asym- 
metries of  brain  function.  Alcohol  Health 
Res  World  16:273-279,  1992. 

Oscar-Berman,  M.,  and  Bardenhagen,  F. 
Nonhuman  animal  models  of  memory 
dysfunction  in  neurodegenerative  disease.  In: 
Troster,  A.,  ed.  Memory  in  Neurodegenerative 
Disease.  New  York:  Cambridge  University 
Press,  1998.  pp.  3-20. 

Oscar-Berman,  M.,  and  Evert,  D.L. 
Alcoholic  Korsakoff's  syndrome.  In: 
Nussbaum,  P.D.,  ed.  Handbook  of 
Neuropsychology  and  Aging.  New  York: 
Plenum,  1997.  pp.  201-215. 

Oscar-Berman,  M.,  and  Hutner,  N.  Frontal 
lobe  changes  after  chronic  alcohol  ingestion. 
In:  Hunt,  W.A.,  and  Nixon,  S.J.,  eds. 
Alcohol-Induced  Brain  Damage.  National 
Institute  on  Alcohol  Abuse  and  Alcoholism 
Research  Monograph  No.  22.  NIH 


467 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Publication  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  121-156. 

Oscar- Berman,  M.,  and  Pulaski,  J.L. 
Association  learning  and  recognition 
memory  in  alcoholic  Korsakoff  patients. 
Neuropsychology  ll(2):282-289,  1997. 

Oscar-Berman,  M.,  and  Schendan,  H.E. 
Asymmetries  of  brain  function  in  alcoholism: 
Relationship  to  aging.  In:  Obler,  L.,  and 
Connor,  L.T.,  eds.  Neurobehavior  of 
Language  and  Cognition:  Studies  of  Normal 
Aging  and  Brain  Damage.  Norwell,  MA: 
Kluwer  Academic  Publishers,  in  press. 

Oscar-Berman,  M.;  Pulaski,  J.L.;  Hutner, 
N.;  Weber,  D.A.;  and  Freedman,  M.  Cross 
modal  functions  in  alcoholism  and  aging. 
Neuropsychologia  28(8):85 1-869,  1990. 

Oscar-Berman,  M.;  Shagrin,  B.;  Evert, 
D.L.;  and  Epstein,  C.  Impairments  of 
brain  and  behavior.  The  neurological 
effects  of  alcohol.  Alcohol  Health  Res 
World  2l(l):6S-7S,  1997. 

Paller,  K.A.;  Acharya,  A.;  Richardson, 
B.C.;  Plaisant,  O.;  Shimamura,  A. P.; 
Reed,  B.R.;  and  Jagust,  W.J.  Functional 
neuroimaging  of  cortical  dysfunction  in 
alcoholic  Korsakoff's  syndrome.  /  Cogn 
Neurosci9{2):277-293,l997. 

Parsons,  OA.  Neuropsychological  measures 
and  event-related  potentials  in  alcoholics: 
Interrelationships,  long-term  reliabilities, 
and  prediction  of  resumption  of  drinking. 
J  Clin  Psychol  50(1  ):37-46,  1994. 

Parsons,  O.A.,  and  Nixon,  S.J.  Neuro- 
behavioral  sequelae  of  alcoholism.  Neurol 
Clin  11:205-218,  1993. 

Petri,  H.L.,  and  Mishkin,  M.  Behaviorism, 
cognitivism  and  the  neuropsychology  of 
memory.  Am  Scientist  82:30-37 ,  1994. 

Pfefferbaum,  A.,  and  Rosenbloom,  M.J. 
In  vivo  imaging  of  morphological  brain 


alterations  associated  with  alcoholism.  In: 
Hunt,  W.A.,  and  Nixon,  S.J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Publication  No. 
93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  71-87. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Rosenbloom, 
M.J.;  Shear,  P.K;  Mathalon,  D.H.;  and 
Lim,  K.O.  Increase  in  brain  cerebrospinal 
fluid  volume  is  greater  in  older  than  in 
younger  alcoholic  patients:  A  replication 
study  and  CT/MRI  comparison. 
Psychiatry  Res  50(4):257-274,  1993. 

Pfefferbaum,  A.;  Lim,  K.O.;  Desmond, 
J.E.;  and  Sullivan,  E.V.  Thinning  of  the 
corpus  callosum  in  older  alcoholic  men:  A 
magnetic  resonance  imaging  study.  Alcohol 
Clin  Exp  Res  20:752-757,  1996. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Mathalon, 
D.H.;  and  Lim,  K.O.  Frontal  lobe  volume 
loss  observed  with  magnetic  resonance 
imaging  in  older  chronic  alcoholics.  Alcohol 
Clin  Exp  Res  2l(3):S2l-S29,  1997. 

Pihl,  R.O.,  and  Bruce,  K.R.  Cognitive 
impairment  in  children  of  alcoholics.  Alcohol 
Health  Res  World  19(2):142-147,  1995. 

Pollux,  P.M.;  Webster,  A.;  and  De  Hann, 
E.H.  Random  generation  deficit  in  alcoholic 
Korsakoff  patients.  Neuropsychologia  33(1): 
125-129,  1995 

Porjesz,  B.,  and  Begleiter,  H.  Event  re- 
lated brain  potentials  and  cognitive  func- 
tion in  alcoholism.  Alcohol  Health  Res 
World  19(2):108-ll2,  1995. 

Rabbitt,  P.  Methodology  of  Frontal  and 
Executive  Function.  East  Sussex,  England: 
Psychology  Press,  1997. 

Reuler,  J.B.;  Girard,  D.E.;  and  Cooney, 
T.G.  Wernicke's  encephalopathy.  N  Engl 
J  Med  312:1035-1039,  1985. 


468 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


Rourke,  S.B.,  and  Loberg,  T.  The  neu- 
robehavioral  correlates  of  alcoholism.  In: 
Grant,  I.,  and  Adams,  K.M.,  eds.  Neuro- 
psychological Assessment  of  Neuropsychiatry 
Disorders.  2d  ed.  New  York:  Oxford 
University  Press,  1996.  pp.  423^85. 

Schmahmann,  J.D.  The  Cerebellum  and 
Cognition.  New  York:  Academic  Press,  1997. 

Schuckit,  M.A.  Biologic  phenotypes 
associated  with  individuals  at  high  risk  for 
developing  alcohol-related  disorders. 
Paper  prepared  for  the  Second  NIAAA 
Portfolio  Review,  Bethesda,  MD,  1997. 

Seeman,  M.V.  Psychopathology  in  women 
and  men:  Focus  on  female  hormones.  Am 
J  Psychiatry  154(12):  1641-1647,  1997. 

Seger,  C.A.;  Rabin,  L.A.;  Zarella,  M.;  and 
Gabrieli,  J.D.  Preserved  verb  generation 
priming  in  global  amnesia.  Neuropsychologia 
35(8):1069-1074,  1997. 

Shaw,  C;  Kentridge,  R. W.;  and  Aggleton, 
J. P.  Cross-modal  matching  by  amnesic 
subjects.  Neuropsychologia  28:665-671, 1990. 

Shear,  P.K.;  Sullivan,  E.V.;  Lane,  B.;  and 
Pfefferbaum,  A.  Mammillary  body  and 
cerebellar  shrinkage  in  chronic  alcoholics 
with  and  without  amnesia.  Alcohol  Clin 
Ex#itw20(8):1489-1495,  1996. 

Sher,  K.J.,  and  Trull,  T.J.  Personality  and 
disinhibitory  psychopathology:  Alcoholism 
and  antisocial  personality  disorder.  / 
Abnorm  Psychol  103:92-102,  1994. 

Shimomura,  T.;  Mori,  E.;  Hirono,  N.; 
Imamura,  T.;  and  Yamashita,  H.  Develop- 
ment of  Wernicke -Korsakoff  syndrome 
after  long  intervals  following  gastrectomy. 
Arch  Neurol  55:1242-1245,  1998. 

Smith,  D.M.,  and  Atkinson,  RM. 
Alcoholism  and  dementia.  Int  J  Addict 
30(13-14):1843-1869,  1995. 


Sullivan,  E.V.;  Mathalon,  D.H.;  Zipursky, 
RB.;  Kersteen-Tucker,  Z.;  Knight,  RT.; 
and  Pfefferbaum,  A.  Factors  of  the 
Wisconsin  Card  Sorting  Test  as  measures 
of  frontal-lobe  function  in  schizophrenia 
and  in  chronic  alcoholism.  Psychiatry  Res 
46(2):175-199,  1993. 

Sullivan,  E.V.;  Marsh,  L.;  Mathalon,  D.H.; 
Lim,  K.O.;  and  Pfefferbaum,  A.  Anterior 
hippocampal  volume  deficits  in  nonamnesic, 
aging  chronic  alcoholics.  Alcohol  Clin  Exp 
itol9(l):110-122,  1995a. 

Sullivan,  E.V.;  Rosenbloom,  M.J.; 
Deshmukh,  A.;  Desmond,  J.E.;  and 
Pfefferbaum,  A.  Alcohol  and  the  cerebellum: 
Effects  on  balance,  motor  coordination, 
and  cognition.  Alcohol  Health  Res  World 
19(2):138-141,  19956. 

Sullivan,  E.V.;  Deshmukh,  A.;  Desmond, 
J.E.;  Lane,  B.,  Jr.;  Shear,  P.K.;  and 
Pfefferbaum,  A.  Volumetric  MRI  analysis 
of  cerebellar  hemispheres  and  vermis  in 
chronic  alcoholics:  Relationship  to  ataxia. 
J  Int  Neuropsychol  Soc  2:34  (abstract),  1996. 

Sullivan,  E.V.;  Zipursky,  R.B.;  Shear, 
P.K.;  Sagar,  H.J.;  and  Pfefferbaum,  A. 
Patterns  of  content,  contextual,  and 
working  memory  impairments  in  schizo- 
phrenia and  nonamnesic  alcoholism. 
Neuropsychology  11:195-206,  1997. 

Sullivan,  E.V.;  Mathalon,  D.H.;  Lim, 
K.O.;  Marsh,  L.;  and  Pfefferbaum,  A. 
Patterns  of  regional  cortical  dysmorphology 
distinguishing  schizophrenia  and  chronic 
alcoholism.  Biol  Psychiatry  43(2):  1 18— 
131,1998. 

Tallaksen,  C.M.E.;  Bell,  H.;  and  Bohmer, 
T.  Thiamin  and  thiamin  phosphate  ester 
deficiency  assessed  by  high  performance 
liquid  chromatography  in  four  clinical 
cases  of  Wernicke  encephalopathy.  Alcohol 
Clin  Exp  Res  17:712-716,  1993. 


469 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Tarter,  RE.;  Arria,  A.;  and  Van  Thiel,  D.H. 
Liver- brain  interactions  in  alcoholism.  In: 
Hunt,  W.A.,  and  Nixon,  S.J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Publication  No. 
93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  415-430. 

Tracy,  J. I.;  Josiassen,  R.C.;  and  Bellack, 
A.S.  Neuropsychology  of  dual  diagnosis: 
Understanding  the  combined  effects  of 
schizophrenia  and  substance  use  disor- 
ders. Clin  Psychol  Rev  15:67-97,  1995. 

Verfaellie,  M.;  Milberg,  W.P.;  Cermak, 
L.S.;  and  Letourneau,  L.L.  Priming  of 
spatial  configurations  in  alcoholic  Korsakoff's 
amnesia.  Brain  Cogn  18(l):34-45,  1992. 

Victor,  M.  The  effects  of  alcohol  on  the 
nervous  system.  In:  Lieber,  C.S.,  ed.  Medical 
and  Nutritional  Complications  of  Alcoholism: 
Mechanisms  and  Management.  New  York: 
Plenum  Press,  1992.  pp.  413^57. 

Victor,  M.,  and  Yakovlev,  P.I.  S.S.  Korsakoff's 
psychic  disorder  in  conjunction  with  periph- 
eral neuritis.  A  translation  of  Korsakoff  s 
original  article  with  brief  comments  on 
the  author  and  his  contribution  to  clinical 
medicine.  Neurology  5:394-406,  1955. 

Volkow,  N.D.;  Wang,  G.J.;  Hitzemann, 
R;  Fowler,  J.S.;  Overall,  J.E.;  Burr,  G.; 
and  Wolf,  A.P.  Recovery  of  brain  glucose 
metabolism  in  detoxified  alcoholics.  Am  J 
Psychiatry  151:178-183,  1994. 

Volkow,  N.;  Wang,  G.-J.;  and  Doria,  J.J. 
Monitoring  the  brain's  response  to  alcohol 
with  positron  emission  tomography.  Alcohol 
Health  Res  World  19(4):296-299,  1995. 

Volkow,  N.D.;  Wang,  G.J.;  Overall,  J.E.; 
Hitzemann,  R;  Fowler,  J.S.;  Pappas,  N.; 
Frecska,  E.;  and  Piscani,  K.  Regional 
brain  metabolic  response  to  lorazepam  in 
alcoholics  during  early  and  late  alcohol 


detoxification.  Alcohol  Clin  Exp  Res 
21(7):1278-1284,  1997. 

Voyer,  D.  On  the  magnitude  of  laterality 
effects  and  sex  differences  in  functional 
lateralities.  Laterality  l(l):51-83,  1996. 

Wall,  T.L.;  Thomasson,  H.R.;  and  Ehlers, 
C.L.  Investigator-observed  alcohol-induced 
flushing  but  not  self- report  of  flushing  is  a 
valid  predictor  of  ALDH2  genotype./ 
Stud  Alcohol  57(3):267-272,  1996. 

Wang,  G.-J.;  Volkow,  N.D.;  Roque,  C.T.; 
Cestaro,  V.L.;  Hitzemann,  R.J.;  Cantos, 
E.L.;  Levy,  A.V.;  and  Dhawan,  A.P. 
Functional  importance  of  ventricular 
enlargement  and  cortical  atrophy  in 
healthy  subjects  and  alcoholics  as  assessed 
with  PET,  MR  imaging,  and  neuropsycho- 
logic testing.  Radiology  186(1  ):59-65, 1993. 

Wang,  J. J.;  Martin,  P.R;  and  Singleton, 
C.K.  A  transketolase  assembly  defect  in  a 
Wernicke-Korsakoff  patient.  Alcohol  Clin 
£xpit«21(4):576-580,  1997. 

Wechsler,  D.  Wechsler  Adult  Intelligence 
Scale-Revised:  Manual.  New  York: 
Psychological  Corporation,  1981. 

Wechsler,  D.,  and  Stone,  C.P.  Wechsler 
Memory  Scale.  New  York:  Psychological 
Corporation,  1945. 

Welch,  L.W.;  Cunningham,  A.T.;  Eckardt, 
M.J.;  and  Martin,  P.R.  Fine  motor  speed 
deficits  in  alcoholic  Korsakoff  syndrome. 
Alcohol  Clin  Exp  ite21(l):134-139, 1997. 

Wickelgren,  I.  Getting  a  grasp  on  working 
memory.  Science  275:1580-1582, 1997. 

Wilkinson,  DA.,  and  Carlen,  P.L. 
Morphological  abnormalities  in  the  brains 
of  alcoholics:  Relationship  to  age,  psycho- 
logical test  scores  and  patient  type.  In: 
Wood,  W.G.,  and  Elias,  M.F.,  eds. 
Alcoholism  and  Aging:  Advances  in 


470 


Neuropsychological  Vulnerabilities  in  Chronic  Alcoholism 


Research.  Boca  Raton,  FL:  CRC  Press, 
1982.  pp.  61-77. 

Witt,  E.D.  Neuroanatomical  consequences 
of  thiamine  deficiency:  A  comparative 
analysis.  Alcohol  Alcohol  20(2): 
201-221,  1985. 

Woodruff-Pak,  D.  Cerebellar  role  in 
learning  and  timing.  Paper  presented  at 
the  Tenth  Annual  Meeting  of  the 


Memory  Disorders  Research  Society, 
Cambridge,  MA,  1998. 

Zhang,  X.L.;  Begleiter,  H.;  Porjesz,  B.; 
and  Litke,  A.  Electrophysiological  evi- 
dence of  memory  impairment  in  alcoholic 
patients .  Biol  Psychiatry  42(12): 
1157-1171,  1997. 

Zola-Morgan,  S.M.,  and  Squire,  L. 
Neuroanatomy  of  memory.  Anna  Rev 
Neurosci  16:547-563,  1993. 


471 


Chapter  14 

Human  Brain  Vulnerability  to  Alcoholism: 
Evidence  From  Neuroimaging  Studies 

Edith  V.  Sullivan,  Ph.D. 


KEY  WORDS:  AOD  (alcohol  or  other  drug)  dependence;  neuroimaging;  alcoholic 
brain  syndrome;  brain  damage;  chronic  AODE  (effects  of  AOD  use,  abuse,  and 
dependence);  risk  factors;  nutritional  disorder;  computed  x-ray  tomography;  magnetic 
resonance  imaging;  single  photon  emission  computed  tomography;  magnetic  resonance 
spectroscopy;  Korsakoff's  syndrome;  gender  differences;  biological  repair;  ethanol 
metabolite; glucose  metabolism;  comorbidity; gender  differences;  aging;  literature  review 


It  has  been  suspected  for  centuries  and 
known  for  decades  that  excessive,  chronic 
alcohol  consumption  damages  brain  struc- 
ture and  impairs  brain  function.  Before 
the  introduction  of  in  vivo  neuroimaging 
techniques,  the  establishment  of  brain 
structural  compromise  relied  on  post- 
mortem data,  which  by  definition,  are 
cross-sectional,  biased  to  cases  that  come 
to  autopsy  (in  years  gone  by,  samples 
may  have  been  typically  indigent  and 
malnourished  [cf.  Sullivan  1899]),  and 
susceptible  to  fixation  artifacts.  In  vivo 
imaging  methods  have  the  advantage  of 
capturing  the  dynamic  course  of  alcohol- 
ism, and  some  methods  are  amenable 
to  experimental  challenges  and  exami- 
nation of  neurochemistry,  thus  providing 
opportunities  to  go  beyond  description 


of  abnormality  and  to  identify  mecha- 
nisms of  neuronal  disruption  and 
vulnerability.  Collection  of  three- 
dimensional  data  sets  of  neuroimages 
provides  snapshots  of  an  individual's 
brain  at  a  specific  time  point,  can  be 
repeated  to  follow  change,  and,  unlike 
the  autopsied  brain,  can  be  resliced  on 
later  occasions  to  address  new  ques- 
tions. Despite  their  unparalleled  worth, 
in  vivo  studies  provide  relatively  coarse 
data  and  are  unlikely  to  fully  replace 
postmortem  studies,  which  at  least  cur- 
rently are  essential  for  verifying  actual 
pathology  and  for  providing  microscopic 
analyses  of  cytoarchi tectonic  and  cellu- 
lar structure. 

The  initial  optimism  accompanying 
the  emergence  of  in  vivo  imaging 


E.V.  Sullivan,  Ph.D.,  is  an  associate  professor  in  the  Department  of  Psychiatry  and  Behavioral  Sciences 
and  the  Neurosciences  Program,  Stanford  University  School  of  Medicine,  401  Quarry  Rd.,  Stanford, 
CA  94305-5717. 


473 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


methods  was  somewhat  damped  when 
the  predicted  brain  "pathology"  was 
not  always  clearly  visible  and  when  the 
predicted  relationships  between  brain 
integrity  and  cognition  or  alcohol 
consumption  variables  were  not  readily 
forthcoming.  Brain-behavior  discoveries 
based  on  patient  groups  with  gross, 
focal  brain  lesions  may  have  been  pos- 
sible and  the  application  of  the  double 
dissociation  model  may  have  been 
successful  because  the  "signal"  from 
the  lesion  was  powerful  enough  to 
override  the  "noise"  from  other  factors, 
such  as  age  or  comorbid  disease,  which 
may  have  had  an  influence  in  cases  where 
the  lesion  was  less  prominent,  as  holds 
for  alcoholism.  Additional  factors  that 
may  attenuate  obvious  relationships,  such 
as  dose-response  relationships  (greater 
alcohol  intake  and  greater  brain  dam- 
age), include  age,  gender,  nutrition, 
genetics,  and  comorbidities,  each  of 
which  may  exert  its  own  contribution 
to  brain  dysmorphology  and  may  dif- 
ferentially dispose  the  alcoholic  indi- 
vidual to  relatively  greater  or  less  insult. 
Thus  there  is  a  multiplicity  of  factors 
that  may  influence  the  brain  and  its 
functions  in  alcoholism,  and  each 
must  be  controlled  and  will  be  consid- 
ered in  this  review. 

A  growing  number  of  in  vivo  brain 
imaging  technologies  are  available. 
Methods  already  applied  to  study 
alcoholism  include  those  for  structural 
imaging  (computed  tomography  [CT], 
magnetic  resonance  imaging  [MRI], 
and  magnetization  transfer  [MT])  and 
functional  imaging  (single  photon  emis- 
sion computed  tomography  [SPECT], 
positron  emission  tomography  [PET], 
and  magnetic  resonance  spectroscopy 


[MRS]  and  spectroscopic  imaging 
[MRSI]).  Findings  from  a  sample  of 
these  studies  will  be  summarized  here 
(see  also  reviews  by  Pfefferbaum  and 
Rosenbloom  1993;  Jernigan  and  Cermak 
1994;  Fein  et  al.  1995;  Volkow  et  al. 
1995);  this  review  will  reflect  the  fact 
that  most  imaging  studies  in  alcohol- 
ism have  examined  brain  structure. 

PATTERNS  OF  STRUCTURAL 
BRAIN  ABNORMALITIES 

Early  imaging  studies  in  chronic  alcohol- 
ics were  based  on  CT  and  revealed  a 
significantly  dilated  ventricular  system 
and  enlarged  cortical  sulci  throughout 
the  cortex  (Haubek  and  Lee  1979; 
Ron  et  al.  1982;  Wilkinson  1985; 
Pfefferbaum  et  al.  1988;  Lishman 
1990).  The  resolution  of  CT,  how- 
ever, did  not  permit  reliable  separation 
of  tissue  types.  With  the  appropriate 
image  acquisition  sequences,  MRI  can 
distinguish  gray  matter  and  white  mat- 
ter (Wehrli  et  al.  1984).  Application 
of  such  MRI  sequences  in  studies  of 
detoxified  alcoholics  has  provided 
convincing  evidence  that  the  increases 
in  cortical  cerebrospinal  fluid  (CSF)- 
filled  spaces,  previously  observed  with 
CT  (Pfefferbaum  et  al.  1988,  1993), 
occur  at  the  expense  of  both  gray  mat- 
ter and  white  matter  (Jernigan  et  al. 
1991  a;  Pfefferbaum  et  al.  1992).  This 
pattern  of  gray  and  white  matter  tissue 
involvement  differs  from  that  seen  in 
other  neuropsychiatric  conditions  such 
as  schizophrenia  (Zipursky  et  al. 
1992;  Sullivan  et  al.  1998&)  and 
Alzheimer's  disease  (Jernigan  et  al. 
1991  b;  Fama  et  al.  1997;  Tanabe  et 
al.  1997;  Pfefferbaum  et  al.  1999&), 


474 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


both  of  which  have  cortical  gray  but 
little,  if  any,  white  matter  volume  loss. 
The  tissue  shrinkage  visually  detectable 
on  MRI  can  be  as  extensive  in  "uncom- 
plicated" alcoholism  (figure  1)  as  it  is 
in  Alzheimer's  disease,  yet  the  behav- 
ioral consequences,  such  as  memory 
impairment,  are  not  so  clinically  appar- 
ent in  alcoholism. 

Cortical  volume  deficits  are  present 
throughout  the  cortex  and  subjacent 
white  matter  (Jernigan  et  al.  199 la; 
Pfefferbaum  et  al.  1992,  1995;  but 
see  Di  Sclafani  et  al.  1995).  A  regional 
profile  of  volume  abnormalities,  how- 
ever, was  not  apparent  in  alcoholics 
until  age  was  considered.  Identifying 
any  regional  pattern  of  abnormality 
first  requires  standardizing  structures 
of  widely  different  fundamental  sizes 
(e.g.,  lateral  and  third  ventricles,  frontal 
lobes  and  temporal  lobes)  to  a  common 
metric.  If  such  a  metric  also  takes  the 
effects  of  normal  aging  and  variations 
associated  with  head  size  difference  into 


account,  regional  patterns  of  volume 
abnormalities  can  be  seen. 

Age 

The  brain  morphology  of  older  alco- 
holics (i.e.,  from  about  age  45  years 
onward)  is  especially  vulnerable  to  the 
adverse  effects  of  alcohol  consumption 
(Wilkinson  and  Carlen  1980;  Ron  et  al. 
1982;  Pfefferbaum  et  al.  1988,  1992; 
Sullivan  et  al.  1995«;  Pfefferbaum  et  al. 
1996#).  To  legitimately  reach  such  a  con- 
clusion, however,  the  effects  of  normal 
aging,  as  well  as  normal  variation  in  head 
size,  on  the  brain  must  be  measured  and 
statistically  taken  into  account;  this 
can  be  done  with  regression  analysis 
(Pfefferbaum  et  al.  1986,  1992; 
Mathalon  et  al.  1993).  The  use  of  age - 
corrected  standardized  brain  volumes 
permits  a  cross-sectional  approach  for 
estimating  the  dual  effect  of  age  and 
alcohol  on  different  regions  of  the  brain. 
Our  studies  based  on  this  method  have 
consistently  revealed  an  age-alcohol 


Figure  1.  Surface-rendered  brains  of  a  57-year-old  healthy  man  (left),  who  consumed  about 
60  kg  of  ethanol  in  his  lifetime,  and  a  57-year-old  alcoholic  man  (right),  who  consumed  about 
1,866  kg  of  ethanol  in  his  lifetime.  Note  the  thinner  gyri  and  wider  sulci  and  interhemispheric 
fissure  in  the  alcoholic  man's  brain  compared  with  the  healthy  man's  brain. 


475 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


interaction,  where  older  alcoholics 
have  greater  tissue  volume  loss  and 
CSF  volume  expansion  for  their  ages 
(and  for  a  given  head  size)  than  do 
younger  alcoholics. 

The  age-alcohol  interaction  holds 
for  a  number  of  brain  regions.  Specifi- 
cally, although  gray  matter  and  white 
matter  volume  loss  in  alcoholics  is  wide- 
spread throughout  the  cortex,  the  vol- 
ume loss  of  both  tissue  types  in  the 
frontal  lobes  is  relatively  greater  than  that 
detected  in  the  more  posterior  reaches 
of  the  cortex  in  older  (>  45  years  of  age) 
than  younger  alcoholics  (Pfefferbaum 
et  al.  1997).  When  regional  volumes 
were  compared,  the  older  groups  exhib- 
ited substantial  deficits  in  prefrontal 
cortical  volume  of  gray  matter  and  in 
prefrontal  and  frontal  white  matter  vol- 
ume. This  observation  supports  post- 
mortem studies,  which  identified  the 
frontal  lobes  as  being  particularly 
damaged  in  alcoholics  (Courville  1955; 
Harper  and  Kril  1990).  Similar  age- 
related  declines  occur  in  cerebellar 
hemisphere  gray  matter  volumes  in 
alcoholic  men  (Sullivan  et  al.  in  press). 

Other  selective  brain  structures  are 
also  deleteriously  affected  by  chronic, 
excessive  alcohol  consumption.  Mea- 
surement of  these  structures  entails 
careful  tracing  of  the  anatomical  bor- 
ders across  multiple,  contiguous  MRI 
slices  of  the  target  brain  structure. 
These  analyses  have  shown  that  non- 
amnesic  alcoholics  have  significant, 
age-related  bilateral  volume  deficits  in 
the  anterior  but  not  posterior  hippocam- 
pus (Sullivan  et  al.  1995#).  The  volume 
deficits  were  even  more  severe  (by  1 
standard  deviation)  in  alcoholics  with 
global  amnesia  and  likely  Korsakoff's 


syndrome  (KS)  (Sullivan  et  al.  1996; 
see  also  Jernigan  et  al.  1991c). 

Prenatal  exposure  to  alcohol  (PEA) 
disrupts  the  development  and  integrity 
of  the  corpus  callosum  (human  [Peiffer 
et  al.  1979;  Clarren  1986;  Mattson  et 
al.  1994];  rat  [Zimmerberg  and  Scalzi 
1989;  Zimmerberg  and  Mickus  1990; 
Coulter  et  al.  1993]).  The  adult  aging 
corpus  callosum  is  also  affected  by  exces- 
sive alcohol  consumption.  Hommer  and 
colleagues  (1996)  reported  smaller  cal- 
losal  areas  in  alcoholic  women  but  not 
men  who  ranged  in  age  from  30  to  50 
years.  In  a  sample  of  alcoholic  men 
ages  28-67,  we  observed  that  the  corpus 
callosum  is  thinner  throughout  its 
extent,  from  rostrum  and  genu  to 
splenium  (Pfefferbaum  et  al.  1996&) 
and  that  smaller  callosal  areas  signifi- 
cantly correlated  with  older  age  (r  = 
-0.56,  p<  0.01). 

Alcohol  abuse  and  dependence  are 
prevalent  in  adolescents  as  well  as  older 
individuals,  yet  alcohol's  adverse  effects 
on  the  adolescent  brain  is  unknown. 
Alcohol's  teratological  effects  in  humans 
are  under  study  in  children  and  adults 
with  fetal  alcohol  syndrome  (FAS)  or 
PEA.  To  date,  quantitative  MRI  studies 
of  FAS  and  PEA  patients  have  revealed 
abnormally  small  volumes  of  the  cranial 
vault,  basal  ganglia  (in  particular,  the 
caudate  nucleus),  and  diencephalon  (a 
brain  region  including  thalamic  and 
septal  nuclei)  (Mattson  et  al.  1996). 
Additional  sites  of  dysmorphology 
included  the  anterior  superior  vermis 
(Sowell  et  al.  1996)  and  corpus  callo- 
sum (Mattson  et  al.  1992;  Riley  et  al. 
1995),  which  are  consistent  with  post- 
mortem studies  and  animal  models  of 
FAS  (Clarren  1986;  West  et  al.  1990; 


476 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Goodlett  and  Lundahl  1996).  These 
teratological  studies  suggest  that  even 
in  adolescents  free  of  PEA  the  still- 
developing  brain  could  be  particularly 
vulnerable  to  alcohol's  neurotoxicity, 
which  could  result  in  aberrant  neural 
connectivity  and  arrested  neuronal 
growth,  and  consequently  irreversible 
brain  insult. 

Nutrition  and  Other 
Factors 

Nutritional  status  is  known  to  contri- 
bute to  alcohol-related  brain  dysfunction 
and  dysmorphology.  Korsakoff's  syn- 
drome, whether  or  not  accompanied  by 
alcoholism,  is  associated  with  lesions  of 
the  diencephalon  and  thalamic  nuclei 
resulting  from  thiamine  deficiency 
(Witt  and  Goldman-Rakic  1983;  Victor 
et  al.  1989;  Langlais  and  Zhang  1997). 
Thiamine  replacement  therapy  is  reg- 
ularly used  clinically  and  prophylactically 
to  reduce  the  chances  of  developing 
KS  in  alcoholics  who  undergo  with- 
drawal. The  dramatic  syndrome  of 
Marchiafava-Bignami  disease,  often 
associated  with  alcoholism  and  possi- 
bly caused  by  deficiency  in  dietary 
nicotinic  acid  or  its  precursor,  trypto- 
phan (Victor  et  al.  1989),  pathogno- 
monically  involves  demyelination  of 
the  corpus  callosum  and  the  discon- 
nection syndrome.  Early  studies  of 
severe  alcoholism  with  Wernicke's 
encephalopathy  reported  no  cases  of 
corpus  callosum  degeneration  (Victor  et 
al.  1989)  and  suggested  that  the  two 
conditions  have  different  etiologies.  It 
is  not  known  whether  adequate  nutri- 
tional supplements  improve  the  struc- 
tural condition  of  the  brain  as  they  do 
cognition  and  motor  abilities. 


Subjects  in  our  neuroimaging  stud- 
ies who  were  scanned  after  a  2 8 -day 
inpatient  rehabilitation  program  were 
well  nourished  at  the  time,  yet  they 
still  manifested  relationships  between 
nutritionally  dependent  variables,  such 
as  indices  of  macrocytic  anemia  and 
brain  volumes  (Pfefferbaum  et  al.  1988, 
1992;  Davila  et  al.  1994).  Specifically, 
body  mass  index,  hemoglobin,  and 
mean  corpuscular  volume  were  corre- 
lated with  CT  measures  of  ventricular 
volume  (Pfefferbaum  et  al.  1988).  In  a 
different  sample,  also  abstinent  about  1 
month,  mean  corpuscular  volume, 
hemoglobin,  and  hematocrit  were  cor- 
related with  MRI  measures  of  ventricu- 
lar volumes  (Pfefferbaum  et  al.  1992). 

The  landmark  neuropathology  stud- 
ies of  Victor,  Adams,  and  Collins  (Victor 
et  al.  1971, 1989)  suggested  that  severe 
cell  damage  in  the  anterior  superior 
vermis,  dorsomedial  nucleus  of  the  thal- 
amus, and  in  many  cases  the  mammillary 
bodies  is  pathognomonic  for  alcoholic 
KS  and  is  probably  caused  by  thiamine 
deficiency  (Butterworth  et  al.  1993; 
Langlais  and  Savage  1995).  The  KS- 
specific  cerebral  damage  to  periven- 
tricular and  thalamic  nuclei  is  thought 
to  cause  the  prominent  global  amne- 
sia defining  KS  (Butters  1985;  Butters 
and  Stuss  1989;  Victor  et  al.  1989). 
Some  pathology  studies  conclude  that 
autopsied  cases  with  significant  atrophy 
in  these  regions  together  with  the  vermis 
have  KS  even  in  the  absence  of  the 
diagnostic  constellation  of  aberrant 
behavior  in  life  (Torvik  1991). 

With  in  vivo  imaging,  we  observed 
that  the  cerebellar  vermis  can  be  sub- 
stantially affected  in  alcoholics  who  do 
not  show  signs  of  KS,  and  the  region 


477 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  greatest  loss  is  the  anterior  superior 
lobules  (Sullivan  et  al.  1995  b),  which  is 
consistent  with  postmortem  studies  of 
alcoholic  KS  (Victor  et  al.  1989).  The 
mammillary  bodies  are  also  abnormally 
small  in  volume  in  nonamnesic  alcohol- 
ics (Sullivan  et  al.  1999).  Mammillary 
body  dysmorphology  was  evident  even 
in  a  series  of  studies  based  on  a  3 -point 
rating  system  of  MRI  films  in  older 
alcoholics  (younger  alcoholics  were 
comparable  to  controls  [Charness 
1993]).  In  those  studies,  we  observed  a 
significant  incidence  of  mammillary 
body  shrinkage  in  alcoholics  with  or 
without  symptoms  of  KS  (Davila  et  al. 
1994),  and  instances  of  normal-sized 
mammillary  bodies  in  alcoholics  with  KS 
amnesia  (Shear  et  al.  1996).  Mammil- 
lary body  volume  deficits,  however,  were 
not  predictive  of  performance  on  tests 
of  explicit  memory,  such  as  delayed 
recognition  or  recall. 

Using  anatomically  determined  MRI 
measurement  of  brain  structures,  Squire 
and  colleagues  (1990)  observed  signif- 
icantly smaller  area  measures  of  the 
mammillary  nuclei  but  not  of  the  hip- 
pocampal  formation  in  four  alcoholic 
KS  patients  relative  to  four  non-KS 
amnesic  patients,  whereas  the  non-KS 
amnesics  showed  the  opposite  pattern 
of  regional  sparing  and  loss.  These 
authors  further  concluded  that  the 
amnesia  in  the  KS  patients  is  more  likely 
associated  with  the  shrinkage  observed 
in  the  mammillary  nuclei,  possibly 
combined  with  thalamic  abnormalities 
observed  with  CT  in  these  same 
patients  by  Shimamura  and  colleagues 
(1988).  An  MRI  study  by  Jernigan 
and  colleagues  (1991c),  using  geomet- 
rically determined  volume  measurements, 


observed  significant  cortical  sulcal 
enlargement,  which  occurred  at  the 
expense  of  cortical  gray  matter  loss,  in 
nonamnesic  alcoholics;  these  volume 
abnormalities  were  even  greater  in  a  KS 
group.  Consistent  with  this  observation, 
another  MRI  study  (Sullivan  et  al. 
1996&)  reported  that  relative  to  an 
age-range-matched  control  group,  KS 
patients  and  nonamnesic  alcoholics 
had  significantly  and  comparably  smaller 
gray  matter  volumes  throughout  the 
cortex;  in  addition,  an  observed  white 
matter  volume  deficit  was  confined  to 
the  prefrontal  region.  Although  both 
alcoholic  groups  had  significantly  smaller 
anterior  hippocampi  than  the  control 
group,  these  structures  were  smaller  by 
1  standard  deviation  in  the  KS  relative 
to  the  nonamnesic  alcoholics.  Together 
with  the  observation  that  amnesic  and 
nonamnesic  alcoholics  share  a  similar 
degree  of  clinically  detectable  mammil- 
lary body  shrinkage,  these  results  suggest 
that  extensive  hippocampal  volume 
deficits  probably  contribute  to  alcohol- 
related  amnesia  of  KS. 

The  extent  and  nature  of  brain  dys- 
morphology found  in  chronic  alcoholics 
may  be  exacerbated  by  conditions  fre- 
quently associated  with  chronic  alco- 
holism: liver  disease  (Patek  1979; 
Acker  et  al.  1982;  Rao  and  Butterworth 
1995),  use  of  hepatotoxic  drugs  (Miitzell 
1992),  withdrawal  seizures  (Sullivan 
et  al.  1996#),  or  withdrawal  delirium 
tremens  (Daryanani  et  al.  1994).  Signif- 
icant liver  disease  is  a  standard  criterion 
for  exclusion  from  brain  imaging  stud- 
ies of  alcoholism.  Withdrawal-related 
neurological  symptoms  may  explain 
differential  vulnerability;  this  is  particularly 
relevant  given  reports  that  withdrawal 


478 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


symptoms  are  more  severe  in  older 
than  in  younger  alcoholics  (Brower  et 
al.  1994). 

As  many  as  2  to  15  percent  of  chronic 
alcoholics  experience  seizures,  typi- 
cally grand  mal  in  type  (Victor  and 
Brausch  1967),  when  they  withdraw 
from  drinking  alcohol  (Chan  1985; 
Booth  and  Blow  1993;  Anton  and 
Becker  1995).  Alcohol-related  seizures 
constitute  about  50  percent  of  hospital- 
izations for  seizure  in  the  United  States 
(Anton  and  Becker  1995).  Several 
attempts  made  to  identify  brain  correlates 
of  withdrawal  seizures  have  been  largely 
unsuccessful  (Meyer- Wahl  and  Braun 
1982;  Daryanani  et  al.  1994;  see  also 
Fox  et  al.  1976).  Given  the  low  seizure 
threshold  of  the  hippocampus,  we  asked 
specifically  whether  alcoholics  with  a 
positive  history  of  withdrawal  seizures, 
established  by  retrospective  chart 
review,  would  have  greater  hippocampal 
volume  deficits  than  those  without 
such  a  history.  Alcoholics  with  and 
without  withdrawal  seizure  history 
demonstrated  comparable  tissue  volume 
deficits  in  hippocampus,  as  well  as 
frontoparietal  gray  and  white  matter 
and  temporal  lobe  gray  matter.  Tempo- 
ral lobe  white  matter,  however,  showed 
greater  volume  deficits  in  the  patients 
with  histories  of  withdrawal  seizures 
than  in  those  without  (Sullivan  et  al. 
1996#).  Although  a  history  of  seizures 
puts  alcoholics  at  a  particularly  high 
risk  for  future  seizures  (e.g.,  Victor 
and  Brausch  1967;  Ballenger  and  Post 
1978;  Brown  et  al.  1988;  Lechtenberg 
and  Worner  1990;  Lechtenberg  and 
Worner  1991;  Booth  and  Blow  1993), 
we  do  not  know  whether  the  seizure - 
prone  alcoholics  already  had  less  white 


matter,  or  whether  repeated  seizures 
induced  the  structural  change.  Nonethe- 
less, it  appears  that  reduced  white 
matter  volume  in  the  temporal  lobes 
is  either  a  risk  factor  for  or  sequela  of 
alcohol  withdrawal  seizures. 

Gender  Differences 

There  is  a  paucity  of  data  on  the  effects 
of  alcoholism  on  the  brains  of  women. 
Anecdotal  reports  suggest  that  women 
with  alcoholism  are  more  difficult  to 
recruit  for  such  study  than  are  their  male 
counterparts  and  that  those  women 
who  do  volunteer  for  study  are  more 
likely  than  men  to  carry  comorbidities, 
such  as  depression  or  eating  disorders, 
that  would  typically  exclude  them 
from  study.  Another  problem  is  that 
female  alcoholic  subjects  often  consume 
less  alcohol  than  male  alcoholic  subjects. 
Consequently,  the  samples  of  women 
and  men  currently  studied  may  not  be 
comparable.  It  is  predicted,  however, 
that,  as  was  observed  with  cardiovascular 
disease  and  smoking,  alcohol  consump- 
tion rates  in  women  may  approach  those 
of  men  as  more  women  enter  the  work- 
force (Parker  and  Harford  1992;  Roman 
and  Blum  1992).  Indeed,  gender  dif- 
ferences in  hormones  and  neuro- 
steroids  and  the  presence  of  estrous 
and  menstrual  cycles  in  women  pro- 
vide additional  factors  for  interaction 
with  alcohol  that  may  either  be  pro- 
tective to  alcohol-consuming  women 
or  render  them  more  vulnerable  (Lan- 
caster et  al.  1984;  Harper  et  al.  1990; 
Lancaster  1994^,  1994b).  Further 
complicating  direct  gender  comparisons 
are  fundamental  gender  differences  in 
the  normal  population  in  intracranial 
volume  (Dorst  1971;  Pfefferbaum  et 


479 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


al.  1994),  size  of  different  brain  struc- 
tures, and  patterns  of  aging  (Gur  et  al. 
1991;  Blatter  et  al.  1995;  Raz  et  al. 
1997;  Coffey  et  al.  1998). 

Studies  of  alcoholic  or  problem  drink- 
ing in  women  have  suggested  that 
women  can  be  more  vulnerable  not  only 
to  adverse  medical  consequences  (Ashley 
et  al.  1977)  but  also  to  structural  brain 
deficits  (Jacobson  1986;  Mann  et  al. 
\992a).  The  higher  average  percentage 
of  body  fat  in  women  than  men  means 
that  equivalent  doses  of  ingested  alco- 
hol per  pound  of  body  weight  are  less 
diluted  in  total  body  water  in  women 
than  men.  In  addition,  women  exhibit 
lower  levels  of  first-pass  metabolism 
than  men  (Frezza  et  al.  1990),  result- 
ing in  higher  levels  of  acetaldehyde  to 
affect  the  liver  and  other  organs.  Con- 
versely, hormonal  differences  may 
contribute  to  higher  metabolic  rates 
for  ethanol  in  women  than  men 
(Thomasson  1995).  However,  an  analy- 
sis of  cross-national  epidemiologic  data 
showed  that  after  controlling  for  drink- 
ing levels  (quantity  and  frequency),  male 
and  female  alcoholics  were  equally  at 
risk  for  self-reported  health  problems 
(Fillmore  et  al.  1995).  Epidemiologic 
studies  of  the  population  as  a  whole 
show  that  compared  with  men,  women 
start  to  drink  later  in  life,  drink  less  per 
occasion,  are  more  likely  to  be  abstinent 
(York  and  Welte  1994),  and  have  lower 
rates  of  alcoholism  (Kessler  et  al.  1994). 
Some  studies  report  that  drinking  levels 
in  alcoholic  women  decline  with  old  age 
(Wilsnack  et  al.  1984;  Hilton  1991). 
Although  epidemiologic  data  suggest 
that  alcoholism  is  less  likely  to  affect 
women  than  men,  for  those  afflicted 
the  consequences  pose  a  major  public 


health  problem.  All  told,  however,  the 
biological  consequences  of  alcoholism 
in  women  have  been  studied  less  thor- 
oughly than  in  men. 

There  are  few  investigations  of 
morphological  brain  changes  in  alcoholic 
women.  To  date,  none  has  been  opti- 
mally designed  to  determine  whether 
the  female  brain  is  more  vulnerable  than 
the  male  brain  to  the  chronic  effects  of 
alcohol.  A  neuropathological  study 
(Harper  et  al.  1990)  showed  similar 
changes  in  alcoholic  women  compared 
with  control  women  as  were  found 
comparing  alcoholic  and  control  men.  A 
CT  study  comparing  recently  detoxified 
alcoholic  men  and  women,  long-sober 
(Alcoholics  Anonymous)  men  and 
women,  and  control  men  and  women 
found  that  alcoholic  women,  despite 
shorter  histories  of  alcoholic  drinking, 
had  CT  indices  of  atrophy  similar  to 
those  of  the  men  (Jacobson  1986),  sug- 
gesting greater  vulnerability  of  the  female 
brain.  Like  Jacobson,  Mann  and  col- 
leagues (\992a)  did  not  detect  differ- 
ences between  alcoholic  men  and  women 
on  a  number  of  indices  of  brain  shrink- 
age on  CT.  Although  the  women  were 
most  recently  drinking  at  a  comparable 
level  to  the  men,  they  had  shorter  drink- 
ing histories.  With  MRI,  Kroft  and 
colleagues  (1991)  found  no  structural 
abnormalities  in  alcoholic  women  relative 
to  controls,  whereas  Hommer  and  col- 
leagues (1996)  found  corpus  callosum 
reduction  in  alcoholic  women  but  not 
in  men. 

We  compared  MRI  data  from  36 
alcoholic  women  (ages  28-63)  and  48 
healthy  community  control  women 
(ages  20-85).  As  with  men,  control 
women  had  predicted  age  effects  for 


480 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


head  size-corrected  volumes  of  cortical 
gray  matter,  cortical  sulci,  lateral  and 
third  ventricles,  and  no  age  effect  on 
white  matter.  Alcoholic  women  also 
showed  age-related  declines  in  gray 
but  not  white  matter  volumes  and 
increases  in  volumes  of  CSF-filled 
spaces.  These  alcoholic  women,  how- 
ever, showed  less  structural  brain  deficit 
relative  to  age  and  gender  norms  than 
alcoholic  men  (Pfefferbaum  et  al. 
1996^).  Although  the  results  do  not 
directly  support  earlier  reports  ( Jacobson 
1986;  Mann  et  al.  1992&)  that  brains 
of  alcoholic  women  are  more  vulnerable 
to  the  effects  of  alcohol  than  those  of 
men,  it  must  be  noted  that  the  recent 
alcoholic  drinking  levels  in  our  sample 
of  women  were  lower  than  those 
reported  for  women  in  other  studies. 
Further  to  this  point,  the  alcoholic 
women  in  the  sample  who  were  scanned 
following  at  least  2  months  of  sobriety 
had  smaller  third  ventricles  and 
tended  to  have  more  cortical  white 
matter  than  women  scanned  at  shorter 
intervals  of  sobriety. 

Reversibility 

A  CT  study  by  Carlen  and  colleagues 
(1978)  was  the  first  neuroradiological 
report  to  provide  evidence  of  at  least 
partial  reversal  of  alcohol-related  cortical 
sulcal  and  ventricular  dilatation  in  a  small 
sample  of  alcoholics  who  remained 
abstinent  for  about  1  year.  Since  that 
time,  a  number  of  CT  (Artmann  et  al. 
1981;  Ron  et  al.  1982;  Carlen  et  al. 
1984;  Schroth  et  al.  1985;  Muuronen 
et  al.  1989)  and  MRI  (Schroth  et  al. 
1988;  Zipursky  et  al.  1989;  Shear  et 
al.  1994;  Pfefferbaum  et  al.  1995, 1998) 
studies  have  reported  that  alcoholics 


who  remain  abstinent  can  show  signif- 
icant reversal  of  cortical  sulcal  and  ven- 
tricular enlargement.  Of  considerable 
interest  is  the  time  course  of  the  changes, 
the  nature  and  locus  of  the  presumed 
brain  tissue  increases  that  are  reflected 
in  sulcal  and  ventricular  size  reduc- 
tions, and  possible  mechanisms  for 
this  recovery. 

The  first  study  to  employ  a  healthy 
cohort  to  control  for  test-retest  mea- 
surement changes  (Zipursky  et  al. 
1989)  used  MRI  and  reported  that  a 
small  but  significant  decrease  in  ventric- 
ular volume  occurs  early  (10-14  days) 
in  the  course  of  abstinence.  Another 
MRI  study  (Shear  et  al.  1994)  examined 
alcoholics  1  month  after  sobriety  and 
again  3.5  months  later,  when  some 
remained  sober  and  others  had  relapsed. 
White  matter  volume  increased  and 
ventricular  volume  decreased  in  the 
abstainers,  whereas  the  relapsers  showed 
a  trend  toward  further  white  matter 
volume  loss  and  ventricular  dilatation. 
A  reexamination  of  this  sample  about 
1  year  after  the  first  MRI  revealed  that 
relapsers  exhibited  little  change  in  CSF 
volume  during  the  interscan  interval, 
whereas  the  abstainers  showed  a  signif- 
icant decrease  in  cortical  and  subcorti- 
cal CSF  volumes  and  a  trend  toward 
an  increase  in  white  matter  volume 
(Drake  et  al.  1994). 

Differences  in  head  placement  typi- 
cally occur  when  the  same  individual 
is  rescanned,  even  when  great  care  is 
taken  to  standardize  placement.  To 
address  this  methodological  confound, 
we  developed  a  rescan  correction  fac- 
tor to  statistically  remove  the  interscan 
difference  in  intracranial  volume  from  the 
interscan  difference  in  regional  volumes. 


481 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Applying  this  method  in  a  longitudinal 
study  (Pfefferbaum  et  al.  1995),  we 
observed  that  enlargement  in  cortical 
gray  matter  and  reduction  in  sulcal 
and  lateral  ventricular  volumes  occur 
early  in  the  course  of  abstinence  (about 
1  month),  and  that  reduction  in  third 
ventricular  volume  appears  later  with 
continued  abstinence  (about  3  to  12 
months).  Resumption  of  drinking  after 
a  short  period  of  abstinence  arrests 
third  ventricular  volume  improvement 
and  produces  white  matter  volume 
loss.  The  shrinkage  of  the  third  ventricle 
with  further  abstinence  is  consistent 
with  the  5 -year  longitudinal  CT  study 
of  Muuronen  and  colleagues  (1989), 
who  reported  that  third  ventricular 
shrinkage  was  related  to  improvement  in 
performance  on  cognitive  tests,  thus 
providing  evidence  for  functional  rele- 
vance of  the  brain  structural  change. 

In  a  5 -year  longitudinal  MRI  study 
of  control  and  alcoholic  men,  cortical 
gray  matter  diminished  over  time  in 
the  control  subjects,  most  prominently 
in  the  prefrontal  cortex,  whereas  lat- 
eral and  third  ventricles  enlarged 
(Pfefferbaum  et  al.  1998).  The  alco- 
holics showed  similar  age-related 
changes  with  a  greater  rate  of  cortical 
gray  matter  volume  loss  than  the  control 
subjects  in  the  anterior  superior  tempo- 
ral lobe.  Amount  of  alcohol  consumed 
during  followup  predicted  rate  of  corti- 
cal gray  matter  volume  loss,  particularly 
in  the  frontal  regions,  as  well  as  corti- 
cal sulcal  expansion. 

Until  recently,  alcohol  history  factors, 
such  as  total  lifetime  consumption  of 
alcohol,  have  generally  proved  to  be  dis- 
appointing predictors  of  alcohol -related 
conditions,  including  brain  volume 


abnormalities  (for  a  review,  see  Parsons 
19876).  Estruch  and  colleagues  (1993) 
reported  a  dose-response  relationship 
with  medical  and  neurological  compli- 
cations of  alcoholism,  such  as  cirrhosis 
and  peripheral  neuropathy.  Likewise, 
Pfefferbaum  and  colleagues  (1995) 
observed  that  within  abstainers,  lifetime 
alcohol  consumption,  shorter  disease 
duration,  and  less  cortical  white  matter 
volume  after  1  month  of  sobriety  were 
significant  and  independent  predictors 
of  cortical  white  matter  volume  increase 
with  prolonged  abstinence.  Within  the 
relapsers,  greater  lifetime  consumption 
of  alcohol  was  associated  with  greater 
decrease  in  anterior  white  matter  and 
increase  in  third  ventricular  volume. 
This  dose  relationship  persisted  even 
after  accounting  for  age,  disease  dura- 
tion, and  brain  volume  at  the  start  of 
the  comparison  interval.  In  addition, 
amount  of  alcohol  intake  between  MRIs 
in  the  relapsed  alcoholics  was  related  to 
greater  enlargement  of  the  lateral  ventri- 
cles. These  relationships  provide  support 
for  the  reasonable  speculation  of  dose- 
related,  adverse  influences  of  alcohol  on 
brain  structure. 

The  mechanisms  underlying  morpho- 
logical recovery  are  still  unknown.  A 
well-controlled  study  (Mann  et  al.  1994) 
tested  the  hypothesis  that  reduction  in 
ventricular  size  with  abstinence  reflects 
rehydration  of  brain  tissue  and  demon- 
strated that  tissue  and  CSF  volume 
changes  during  withdrawal  occur  in 
the  absence  of  significant  changes  in 
magnetic  resonance  relaxation  times 
(Tl  and  T2),  measures  derived  from 
magnetic  resonance  images  that  provide 
information  about  the  amount  and  dis- 
tribution of  free  versus  bound  water 


482 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


in  brain  tissue.  CT  studies  also  refuted 
the  rehydration  hypothesis  of  recovery 
by  showing  an  increase  in  CT  density 
measures  with  abstinence,  suggesting 
a  decrease  in  the  water  content  of  brain 
tissue  (reviewed  in  the  study  by  Mann 
et  al.  1992  b).  Neuropathological  reports 
also  support  the  presence  of  increased 
tissue  density  with  abstinence  (Harper 
and  Corbett  1990). 

PATTERNS  OF  FUNCTIONAL 
BRAIN  ABNORMALITIES 

MRS  and  MRSI  Studies 

MRSI  permits  in  vivo  observation  of 
several  metabolites,  some  of  which  have 
distinct  profiles  in  different  brain  tissue 
types.  Studied  in  the  context  of  struc- 
tural anatomy,  the  concentration  of 
different  metabolites  should  help  to 
delineate  metabolite  changes  underlying 
age-  and  disease-related  deterioration 
in  tissue  integrity.  In  vivo  metabolite 
imaging  may  help  to  define  disease 
progress,  assess  efficacy  of  treatments, 
and  track  alcohol-related  changes  across 
the  lifespan.  Most  MRS  alcohol  studies 


to  date  have  measured  metabolites  on 
the  proton  spectrum,  which  provides 
information  about  four  major  brain 
chemicals:  the  N-acetyl  compounds 
(NAc),  creatine  (Cr),  choline  (Cho) 
and  wzyo- inositol  (figure  2).  Further- 
more, MRS  or  MRSI  can  be  used  to 
determine  the  in  vivo  concentration 
and  distribution  of  ethanol  per  se  in 
the  brain. 

Magnetic  resonance-visible  NAc  is 
primarily,  but  not  uniquely,  N-  acetyl  - 
aspartate,  which  is  found  almost 
exclusively  in  neurons  (Urenjak  et  al. 
1992,  1993)  and  thus  may  serve  as  an 
in  vivo  measure  of  neuronal  integrity. 
The  Cr  signal,  generated  by  creatine 
and  phosphocreatine,  is  an  indicator 
of  high-energy  phosphate  metabolism 
(Tedeschi  et  al.  1995).  The  in  vivo 
visible  Cho  peak  is  generated  primar- 
ily by  water-soluble  Cho-containing 
compounds — free  choline,  phospho- 
choline,  and  glycerophosphocholine 
(Barker  et  al.  1994) — and  is  associated 
with  cell  membrane  synthesis  and 
turnover.  Myo-\nosito\  is  present  in  glial 
but  not  neuronal  cell  cultures  (Brand 
et  al.  1993)  and  is  a  precursor  of  the 


Figure  2.  Axial  brain  images  of  a  30-year-old  healthy  man:  (a)  fast  spin  echo  magnetic  reso- 
nance structural  image  segmented  into  compartments  of  cerebrospinal  fluid  (dark  gray),  gray 
matter  (medium  gray),  and  white  matter  (light  gray);  (b)  low-pass  tissue  image  to  match  the 
spatial  frequency  characteristics  of  the  metabolite  images;  (c)  ^-acetyl  image;  (d)  creatine  im- 
age; (e)  choline  image. 


483 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


inositol-triphosphate  second  messen- 
ger pathway,  where  it  functions  to 
maintain  cell  volume  (Lien  et  al. 
1990;  Ernst  et  al.  1997).  Also  spectro- 
scopically  visible  but  more  challenging 
to  measure  are  gamma- aminobutyric 
acid  (GABA),  the  primary  inhibitory 
neurotransmitter  in  the  brain,  and  glu- 
tamate,  a  principal  excitatory  neuro- 
transmitter (Paul  1995). 

Metabolite  Studies 

An  MRSI  study  of  older  alcoholic  men, 
abstinent  for  3-24  months,  revealed 
lower  NAc/Cr  ratios  in  frontal  lobes 
and  somewhat  higher  ratios  in  parietal 
lobes  than  in  control  subjects.  The  rel- 
atively lower  frontal  than  parietal  meta- 
bolite ratio  was  interpreted  as  reflecting 
neuronal  loss  and  glial  hyperplasia  in 
the  alcoholics  (Fein  et  al.  1994).  As 
noted  by  the  authors,  use  of  ratios  does 
not  permit  firm  conclusions  to  be  drawn 
regarding  the  independent  rise  or  fall 
of  either  or  both  metabolites  expressing 
the  ratio. 

Martin  and  colleagues  (1995)  used 
proton  MRS  to  examine  vermian 
integrity  and  potential  changes  in 
recently  detoxified  alcoholics  scanned 
twice:  0-41  days  abstinent  and  then 
10-55  days  abstinent.  Control  subjects 
were  scanned  at  comparable  intervals. 
NAc/Cho  substantially  increased  with 
longer  abstinence,  suggesting  an 
improvement  due  to  either  increases 
in  NAc  levels  or  decreases  in  Cho  levels, 
or  both.  In  a  naturalistic  experiment, 
the  NAc/Cho  ratio  increased  with  absti- 
nence and  decreased  with  relapse  in 
one  alcoholic  who  was  scanned  on  three 
occasions.  Another  study  of  vermian 
integrity  observed  lower  NAc/Cr  and 


Cho/Cr  ratios  in  the  alcoholics  than 
in  the  control  subjects  and  no  group 
differences  in  glutamate/glutamine  or 
wyo-inositol  (Seitz  et  al.  1999).  If  the 
lower  ratios  involving  NAc  are  due  to 
the  NAc  rather  than  the  Cr  component, 
then  this  study  provides  some  evidence 
for  alcohol-related  reduction  in  neuronal 
size,  number,  or  dendritic  arbors  in 
the  absence  of  glial  proliferation  in  the 
cerebellar  vermis.  Consistent  with  this 
finding,  Jagannathan  and  colleagues 
(1996)  detected  lower  NAc/Cr  and 
NAc/Cho  ratios  in  the  thalamus,  frontal 
lobe,  and  cerebellum  of  alcoholics 
abstinent  for  at  least  1  month  relative 
to  control  subjects. 

The  only  study  considering  the 
potential  compounded  effects  of  alcohol 
comorbidity  used  3 1  -phosphorus  MRS 
in  alcoholics  infected  with  the  human 
immunodeficiency  virus  (HIV+),  alco- 
holics HIV-,  low- drinking  HIV+,  and 
low- drinking  HIV-  (Meyerhoff  et  al. 
1995).  The  results  showed  a  cumulative 
rather  than  an  interactive  effect  of  the 
two  diseases,  where  the  alcoholic  HIV+ 
group  had  the  lowest  concentrations 
of  white  matter  and  subcortical  gray 
matter  phosphodiester  and  of  white 
matter  phosphocreatine.  This  pattern  is 
reflective  of  cell  membrane  degeneration, 
which  was  greatest  in  the  comorbid 
group  with  greater  alcohol  use. 

In  an  animal  model  (rat),  injections 
of  the  antagonist  pyrithiamine  resulted 
in  reduced  Cho/NAc  ratios.  Thiamine 
treatment  resulted  in  increased  Cho/ 
NAc  ratios  and  no  change  in  Cr/NAc 
ratios  (Lee  et  al.  1995).  These  results 
suggest  that  the  brain  may  be  resilient 
enough  to  reverse  biochemical  evidence 
of  KS-like  pathology  and,  furthermore, 


484 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


if  treated  early  enough  may  escape 
structural  damage. 

A  major  criticism  of  MRS  studies 
of  the  chronic  effects  of  alcohol  as 
well  as  other  diseases  has  been  the 
expression  of  one  metabolite  as  a  ratio 
of  the  other  to  generate  standardized 
measures.  Furthermore,  few  studies 
have  distinguished  spectroscopic  signals 
arising  from  gray  matter  and  white 
matter,  even  though  metabolites  have 
different  concentrations  in  the  two  tis- 
sue types.  Recent  studies  from  our 
laboratory,  using  absolute  measures, 
showed  disease -related  decrease  in  NAc 
concentration  in  gray  and  white  matter, 
age-related  increase  in  Cr  concentration 
in  gray  matter,  and  age-  and  disease - 
related  increase  in  Cho  concentration 
in  gray  matter  (Pfefferbaum  et  al.  1999 a, 
19996),  pointing  to  the  limitation  of 
ratio  measures. 

The  recently  developed  techniques 
for  estimating  in  vivo  absolute  measures 
of  NAc,  Cho,  and  Cr  separately  for 
white  and  gray  matter,  and  confirmed 
in  vitro  studies  indicating  different 
concentrations  of  these  metabolites  in 
different  tissue  types,  complement  stud- 
ies by  Petroff  and  colleagues  (1995) 
demonstrating  that  spectroscopically 
visible  metabolites  differ  in  concentra- 
tion between  neurons  and  glia.  In 
vitro  studies  show  that  neurosomes 
are  enriched  in  glutamate,  GAB  A,  and 
NAc,  whereas  gliasomes  are  enriched 
in  lactate,  glutamine,  and  myoinositol. 
This  opens  the  possibility  of  designing 
studies  in  which  MRSI  techniques  are 
used  to  identify  the  contribution  of  neu- 
ronal replacement  by  glia  to  document 
mechanisms  of  volume  loss  or  shrinkage 
that  occur  through  withdrawal,  longer 


term  abstinence,  and  relapse.  Separate 
analyses  of  white  and  gray  matter  can 
also  document  the  relative  involvement 
of  white  and  gray  matter  in  these 
processes.  Such  functional  studies  may 
be  more  likely  to  yield  predictable 
brain -behavior  relationships  than  have 
structural  studies. 

Ethanol  Detection  Studies 

MRS  provides  a  powerful  method  for 
tracking  presence  and  distribution  of 
ethanol  in  the  brain  and  the  rate  of  its 
metabolism.  Ethanol  is  acutely  detectable 
with  MRS  and  has  a  characteristic  triplet 
profile  on  the  proton  spectrum.  The 
ethanol  peak  is  clearly  visible  when  blood 
alcohol  levels  are  0.10  percent  (100 
mg/dL),  the  legal  level  of  intoxication 
in  many  States  (Hanstock  et  al.  1990). 
One  study  reported  a  dose-response 
effect,  where  regional  brain  alcohol  levels 
correlated  with  ascending  blood  alcohol 
levels  and  also  with  subjective  estimates 
of  euphoria  and  dysphoria  (Mendelson 
et  al.  1990).  Another  study  showed  that 
acute  ethanol  has  a  heterogeneous  distri- 
bution across  tissue  types  (figure  3).  When 
blood  alcohol  levels  were  between  0.08 
and  0.12  percent,  about  1  hour  after 
ingesting  alcohol,  relative  ethanol  signal 
intensities  were  highest  in  CSF,  30  percent 
less  in  gray  matter,  and  60  percent  less 
in  white  matter  (Spielman  et  al.  1993). 
MRS  of  acute  ethanol  concentration 
and  distribution  may  also  provide  an 
in  vivo  method  for  studying  tolerance, 
behavioral  indices  of  which  are  used  to 
define  alcohol  dependence  (American 
Psychiatric  Association  1994).  One  study 
reported  higher  MRS-measured  brain 
ethanol  concentrations  in  heavy  (non- 
alcoholic) drinkers  than  in  occasional 


485 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


drinkers  (Chiu  et  al.  1994)  following 
alcohol  consumption  yielding  blood 
alcohol  levels  of  0.08  percent.  These 
results  were  interpreted  as  evidence  of 
tolerance  in  the  heavy  drinkers  resulting 
in  membrane  lipid  rigidity  and  decreased 
membrane  binding  of  ethanol,  leading 
to  more  spectroscopically  visible  ethanol. 
To  examine  the  extent  that  tolerance 
may  be  genetically  determined,  it  would 
be  enlightening  to  compare  estimates 
of  nonabsorbed  ethanol  in  nonalcoholic, 
low-alcohol-consuming  persons  who 
have  a  positive  family  history  for  alco- 
holism versus  those  with  a  negative 
family  history. 

PET  Studies 

Functional  imaging  with  PET  requires 
the  use  of  positron-emitting  radioactive 
isotopes  (ligands),  which  are  injected 


into  study  subjects  and  detected  by 
the  scanner.  Depending  on  the  isotope 
used,  PET  can  track  regional  cerebral 
metabolism  in  resting  or  behaviorally 
activated  states  or  can  identify  brain 
regions  and  quantify  the  binding  sites  of 
a  specific  neurotransmitter  ligand  (e.g., 
the  dopa  receptor  with  spiperone). 

Studies  of  brain  metabolism  in 
alcoholism  have  primarily  relied  on 
three  different  positron  emitters. 
18Fluorine  (half-life  of  110  minutes )- 
labeled  deoxyglucose  (FDG)  is  used 
to  image  the  regional  distribution  of 
glucose  in  the  brain  and  provides  an 
index  of  metabolism  rate.  nCarbon 
(half-life  of  20  minutes)  can  also  be  used 
to  label  deoxyglucose.  15Oxygen,  with 
its  short  half-life  (2  minutes),  can  be 
injected  serially  while  subjects  perform 
cognitive,  sensory,  and  motor  tasks 


\  before  EtOH  ingestion 


residual  water 


NAc 


creatine 
choline    \ 


EtOH 


60  min  post-EtOH  ingestion 


i      i      i      r 


5  0 

Chemical  Shift  (ppm) 

Figure  3.  Proton  spectra  acquired  before  alcohol  (EtOH)  ingestion  and  60  minutes  after 
EtOH  ingestion.  The  notable  difference  in  the  spectra  is  the  appearance  of  the  ethanol  triplet  fol- 
lowing alcohol  consumption.  NAc  =  N-acetyl  compounds;  ppm  =  parts  per  million.  Adapted 
from  Spielman,  D.M.;  Glover,  G.H.;  Macovski,  A.;  and  Pfefferbaum,  A.  Magnetic  resonance 
spectroscopic  imaging  of  ethanol  in  the  human  brain:  A  feasibility  study.  Alcohol  Clin  Exp  Res 
17:1072-1077,  1993. 


486 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


under  different  conditions;  differences 
in  regional  brain  activity  from  task  to 
task  can  serve  to  map  neural  networks 
of  regions  invoked  during  specific, 
controlled  tasks.  In  addition,  150  is 
capable  of  tracking  temporal  and 
regional  blood  flow  effects  of  acute 
alcohol  uptake.  Brain  glucose  metabo- 
lism detectable  with  PET  occurs  pri- 
marily in  gray  matter,  that  is,  in  cell 
bodies.  Thus  it  is  a  critical  control  in 
PET  studies  to  express  metabolism  in 
terms  of  the  amount  of  gray  matter 
present  in  a  target  region  as  deter- 
mined by  structural  imaging.  In  the 
case  of  alcoholism,  low  FDG  signal 
may  occur  because  of  abnormally  low 
cerebral  metabolism  or  because  alco- 
holics as  a  group  have  smaller  gray 
matter  volumes,  and  thus  appear  to 
have  lower  metabolism  rates,  than 
their  nonalcoholic  cohorts. 

PET  studies  have  consistently  revealed 
lower  whole  brain  glucose  metabolism 
in  alcoholics  than  in  control  subjects 
(Volkow  et  al.  1992;  Wang  et  al. 
1993).  Wang  and  colleagues  (1993) 
showed  reduced  PET  brain  metabolic 
activity  in  alcoholics  in  the  absence  of 
structural  volume  deficits.  Regionally,  the 
pattern  of  differences  is  greatest  in  frontal, 
cingulate,  parietal,  and  cerebellar  cortices 
(Wik  et  al.  1988;  Gilman  et  al.  1990; 
Martin  et  al.  1992;  Volkow  et  al.  1992, 
1995).  Presence  of  alcoholic  cerebellar 
degeneration  determined  from  signs,  such 
as  gait  ataxia,  was  associated  with  abnor- 
mally low  metabolic  rates  in  the  anterior 
superior  vermis  of  cerebellum  compared 
with  that  observed  in  healthy  control 
subjects  and  alcoholics  free  of  cerebellar 
signs;  however,  alcoholics,  regardless  of 
cerebellar  signs,  had  abnormally  low 


glucose  metabolism  in  the  frontal  lobes 
(Gilman  et  al.  1990,  19966).  Medial 
frontal  hypometabolism  was  associated 
with  poor  performance  on  the  Wisconsin 
Card  Sorting  Test,  which  is  sensitive  to 
dysfunction  or  lesions  of  the  dorsolateral 
prefrontal  cortex  (Milner  1963;  Sullivan 
et  al.  1993).  KS  alcoholics  have  especially 
low  glucose  utilization  in  the  cingulate 
and  precuneal  regions  (Joyce  et  al.  1994). 
Older  alcoholics  may  show  a  further 
age-related  decrease  in  cerebral  metab- 
olism (Sachs  et  al.  1987),  but  rigorous 
studies  have  yet  to  be  conducted  that 
control  for  the  effect  of  normal  aging 
on  cortical  gray  matter  volume  as  well 
as  potentially  normal  age-related 
decline  in  cerebral  metabolism  that  may 
occur  even  after  age-related  volume 
declines  are  considered. 

Family  history  of  alcoholism  may  not 
be  predictive  of  glucose  metabolism 
differences  in  the  cerebrum  (Adams  et 
al.  1998)  but  may  be  predictive  of  such 
differences  in  the  cerebellum  (Volkow 
et  al.  1995).  Volkow  and  colleagues 
used  lorazepam  in  a  PET  FDG  study 
to  seek  mechanisms  underlying  the 
often-reported  blunted  response  of 
alcoholics  to  alcohol.  Brain  regions 
consistently  showing  decreased  glucose 
metabolism  are  those  with  substantial 
density  of  GABAA  and  benzodiazepine 
receptors  (Gilman  et  al  1996a).  Lora- 
zepam is  a  compound  with  behavioral 
and  pharmacological  effects  that,  like 
other  benzodiazepines,  mimic  alcohol, 
including  sleepiness  and  decreased 
motor  and  cognitive  functions.  The  par- 
ticipants in  the  Volkow  et  al.  (1995) 
study  were  nonalcoholics,  but  one  group 
had  a  family  history  of  alcoholism 
whereas  the  other  group  did  not.  Those 


487 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


with  a  positive  family  history  showed  a 
blunted  effect  following  acute  alcohol 
administration,  especially  in  cerebellar 
metabolism.  This  response  suggests 
that  the  GAB  A- benzodiazepine  receptor 
complex  may  have  different  sensitivi- 
ties depending  on  genetic  disposition 
to  alcoholism. 

Regarding  metabolic  changes  with 
alcohol  abstinence,  the  increase  in 
cortical  gray  matter  volume  with  alco- 
hol abstinence  observed  with  MRI 
(Pfefferbaum  et  al.  1995)  is  consistent 
with  the  findings  of  a  PET  FDG  study 
of  recovery  of  brain  glucose  metabolism 
in  detoxified  alcoholics.  An  increase  in 
cerebral  metabolism,  especially  in  the 
frontal  region,  occurred  predominantly 
16-30  days  after  alcohol  withdrawal, 
compared  with  PET  studies  done  in 
alcoholics  8-5  days  and  31-60  days 
after  withdrawal  (Volkow  et  al.  1994). 

lsO  studies  have  focused  on  KS 
because  identification  of  the  neural  sites 
underlying  its  characteristic  global  amne- 
sia for  declarative  events  remains  an 
enigma.  One  study  reported  low  cerebral 
blood  flow  prominent  in  the  frontal- 
temporal  regions  bilaterally  and  the  left 
thalamus,  brain  regions  known  to  con- 
tribute to  memory  impairments  (Matsuda 
et  al.  1997).  Paller  and  colleagues  (1997) 
used  high-resolution  PET  and  observed 
an  anterior  to  posterior  gradient  in 
cerebral  metabolic  rates  for  glucose  uti- 
lization that  were  especially  marked  in 
frontal,  anterior,  and  posterior  cingulate 
regions,  less  so  in  parietal  regions  and 
even  less  so  in  occipital  and  temporal 
regions.  Using  a  cognitive  activation 
paradigm,  these  authors  noted  widespread 
declines  in  glucose  metabolism  in  frontal, 
parietal,  and  cingulate  areas  in  KS  relative 


to  nonamnesic  alcoholics  in  a  delayed 
recognition  task.  These  two  alcoholic 
groups  did  not  differ,  however,  in  glucose 
utilization  in  hippocampus,  a  structure 
in  which  lesions  can  result  in  profound 
amnesia  (Squire  and  Zola  1996). 

SPECT  Studies 

SPECT  studies  with  ligands  that  detect 
regional  cerebral  blood  flow  provide 
evidence  of  hypoperfusion  in  frontal 
lobes  of  detoxified  alcoholics.  The  extent 
of  reduced  perfusion  correlated  with 
performance  on  tests  of  frontal  lobe 
function,  such  as  the  Wisconsin  Card 
Sorting  Test  and  the  Trail  Making  Test 
(Nicolas  et  al.  1993).  Frontal  hypoper- 
fusion was  also  noted  to  be  especially 
prominent  in  alcoholics  with  antisocial 
personality  compared  with  alcoholics  with 
dependent  personality  disorder  (Kuruoglu 
et  al.  1996).  Cerebral  blood  flow  showed 
different  patterns  of  hypoperfusion  in  two 
different  diseases:  frontal  regions  were 
notably  affected  in  KS,  whereas  posterior 
temporal  and  parietal  regions  were  most 
affected  in  Alzheimer's  disease  (Hunter 
et  al.  1989).  In  a  study  (Dupont  et  al. 
1996)  of  recovery  after  abstinence  from 
alcohol,  alcoholics,  whether  recently 
detoxified  or  long-term  abstinent,  showed 
persistently  low  cerebral  blood  flow  as 
measured  by  uptake  of  the  blood  flow 
tracer  iodoamphetamine  (123IMP).  A 
neuropsychological  test  of  nonverbal 
reasoning,  however,  did  distinguish  the 
two  alcohol  groups:  the  long-term  absti- 
nent group  performed  better  than  the 
short-term  abstinent  group  on  the  Raven's 
Progressive  Matrices  (Dupont  et  al  1996). 
Interestingly,  smokers  had  the  lowest 
123IMP  uptake  in  any  group,  thus  identi- 
fying smoking  as  a  significant  confounding 


488 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


factor  in  functional  studies  of  alco- 
holics, who  typically  are  smokers. 

New  Imaging  Directions 

Functional  MRI 

The  functional  significance  of  brain 
volume  loss  in  terms  of  the  commonly 
observed  cognitive  behavioral  deficits 
has  been  difficult  to  demonstrate  (Parsons 
et  al.  1987).  Obstacles  to  observation 
of  selective  structure-function  relationships 
may  include  imprecise  measures  of  com- 
plex, multicomponent  processes,  poor 
understanding  of  the  neural  networks 
underlying  them,  and  a  limited  under- 
standing of  the  role  of  compensatory 
mechanisms.  Functional  MRI  (fMRI) 
techniques  have  the  potential  to  identify 
areas  throughout  the  brain  that  are  acti- 
vated during  performance  of  specific 
components  of  cognitive  operations. 
These  activated  regions  can  then  be 
mapped  onto  structural  images  to  locate 
the  regions  precisely  and  determine 
their  structural  integrity.  The  consistent 
deficits  reported  in  executive  functions 
in  alcoholics  together  with  the  relatively 
selective  cortical  gray  matter  volume  deficit 
observed  in  older  alcoholics  provide  a 
strong  rationale  for  using  functional  imag- 
ing techniques  to  examine,  in  real  time, 
potential  behavioral  deficits  associated 
with  circumscribed  brain  regions. 

One  of  the  few  fMRI  studies  to  date 
involving  alcoholics  examined  repetition 
priming,  a  form  of  implicit  memory 
thought  to  be  relatively  preserved  in 
global  amnesia.  Amnesic  alcoholics, 
nonamnesic  alcoholics,  and  normal 
(nonalcoholic)  control  subjects  each 
showed  the  predicted  reduction  in 
BOLD  (blood  oxygen  level  dependent) 


signal  of  the  left  inferior  prefrontal  cor- 
tex in  conditions  comparing  repeated 
stimuli  with  novel  stimuli  relative  to 
conditions  comparing  only  novel  stim- 
uli (Gabrieli  et  al.  1996).  These  data 
support  the  contention  that  repetition 
priming  is  preserved  even  in  patients 
with  amnesia  for  declarative,  or  explicit, 
events.  The  apparently  normal  pattern 
of  BOLD  response  to  primed  and 
unprimed  stimuli  in  this  experiment 
also  suggests  that  despite  the  potential 
structural  damage  in  frontal  cortex  of 
alcoholics,  at  least  some  cognitive  func- 
tion remains  intact.  This  again  high- 
lights the  importance  of  incorporating 
indices  of  brain  function  as  well  as  struc- 
ture in  studies  investigating  brain- 
behavior  relationships  in  alcoholism. 

Diffusion  Tensor  Imaging 

The  neuropathological  literature,  espe- 
cially that  describing  alcoholics  in 
Australia,  consistently  reports  volumet- 
ric loss  of  white  matter  far  in  excess  of 
that  seen  in  gray  matter  (Harper  and 
Kril  1988,  1990,  1991).  In  vivo  neu- 
roimaging studies  consistently  observe 
volumetric  decrements  in  both  white 
and  gray  matter  volumes,  and  both 
tissue  types  show  some,  albeit  modest, 
recovery  with  abstinence.  There  are 
also  the  dramatic  subsyndromes  of 
alcoholism,  such  as  Marchiafava-Bignami 
disease  and  central  pontine  myelinosis, 
which  are  characterized  by  specific  white 
matter  pathology.  Diffusion  tensor 
imaging  is  a  newly  developed  MRI 
technique  that  provides  a  measure  of 
the  anisotropy  of  water  diffusion  in 
every  voxel  imaged  (Belzunegui  et  al. 
1995).  Diffusion  of  water  molecules  is 
essentially  isotropic;  that  is,  the  diffusion 


489 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


of  water  molecules  exhibits  brownian 
motion  with  diffusion  in  all  directions 
equally  as  occurs  in  CSF.  In  white 
matter,  where  there  are  closely  packed 
bundles  of  parallel  axons,  diffusion  of 
water  molecules  occurs  along  the  axons 
and  is  thus  "anisotropic";  that  is,  the 
diffusion  proceeds  in  a  specific  direction. 
Diffusion  of  molecules  in  gray  matter 
falls  between  that  of  CSF  and  white  mat- 
ter, as  can  be  seen  in  figure  4.  Diffusion 
tensor  imaging  provides  a  number  of 
measures  of  diffusion  anisotropy,  includ- 
ing its  direction  and  magnitude,  and 
thus  offers  a  potential  metric  for 
assessing  the  integrity  of  white  matter 
throughout  the  course  of  alcoholism 
(Pfefferbaum  et  al.  1999c). 

DIFFERENCES  IN 
VULNERABILITY: 
SPECIAL  POPULATIONS 

Psychiatric  Comorbidities 

Alcohol  use  disorders  are  commonly 
present  in  major  psychiatric  diseases 
(Regier  et  al.  1990;  Kendler  et  al. 


1996;  Kessler  et  al.  1996)  (figure  5) 
and  thus  dispose  patients  comorbid  for 
both  diagnoses  to  be  susceptible  to  brain 
volume  abnormalities  from  both  sources. 
Because  most  studies  have  sought  to 
include  only  "clean"  patients  meeting 
strict  criteria  for  a  particular  disease, 
patients  comorbid  for  a  psychiatric 
disease  plus  alcoholism  have  been 
carefully  excluded.  Only  recently  have 
these  combined  effects  been  investi- 
gated rigorously. 

We  compared  regional  cortical  and 
cerebellar  volumes  in  samples  of  schiz- 
ophrenics, alcoholics,  patients  comor- 
bid for  both  schizophrenia  and 
alcoholism,  and  control  subjects.  The 
first  analysis  (Mathalon  et  al.  1995) 
quantified  gray  matter  volumes  across 
six  regions,  from  prefrontal  to  occipi- 
tal cortex.  The  resulting  profile  of  vol- 
ume deficits  was  similar  for  the 
schizophrenic  and  comorbid  groups, 
with  the  greatest  deficits  present  in 
the  prefrontal  and  anterior  temporal 
cortex,  and  different  from  that  of  the 
alcoholic  group,  which  showed  a  flatter 
deficit  profile.  The  prefrontal  cortex  was 


Figure  4.  (a)  Early  (TE  =  20)  and  (b)  late  (TE  =  80)  fast  spin  echo  axial  magnetic  resonance 
images  at  the  level  of  the  lateral  ventricles;  (c)  fractional  anisotropy  image  at  the  same  anatomi- 
cal location  (bright  signal  =  greater  anisotropy  primarily  seen  in  the  white  matter). 


490 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


particularly  vulnerable,  showing  greater 
deficits  in  the  comorbid  group  than 
either  single -diagnosis  group.  Further- 
more, when  the  comorbid  patients  were 
divided  according  to  which  disease 
developed  first,  the  "schizophrenia-first" 
group  had  a  regional  volume  profile 
more  like  the  schizophrenics,  and  the 
"alcoholism-first"  group  was  more 
like  the  alcoholics.  Thus,  alcohol- 
schizophrenia  comorbidity  may  represent 
a  biologically  heterogeneous  group  of 
clinical  entities.  For  the  cerebellar  hemi- 
spheres, the  alcoholic  and  comorbid 
groups  had  volume  deficits  in  gray 
matter.  The  alcoholics  and  comorbid 
patients,  but  not  schizophrenics,  had 
deficits  in  the  anterior  superior  vermis. 
Conversely,  the  schizophrenic  and  the 


Figure  5.  Lifetime  prevalence  of  comor- 
bidity of  psychiatric  and  addictive  disorders, 
including  alcoholism,  in  the  United  States. 
Adapted  from  Regier,  D.A.;  Farmer,  M.E.; 
Rae,  D.S.;  Locke,  B.Z.;  Keith,  S.J.;  Judd, 
L.L.;  and  Goodwin,  F.K.  Comorbidity  of 
mental  disorders  with  alcohol  and  other 
drug  abuse — results  from  the  Epidemiologic 
Catchment  Area  (ECA)  Study.  JAMA 
264:2511-2518,1990. 


comorbid  groups  but  not  the  alcoholic 
group  both  had  severely  enlarged  fourth 
ventricles  relative  to  control  subjects 
(Sullivan  et  al.  in  press).  Thus,  fourth 
ventricular  neuropathology  is  common 
to  schizophrenia  and  anterior  superior 
vermian  neuropathology  is  common 
to  alcoholism.  The  vermian  volume 
deficits  may  underlie  the  residual 
ataxia  of  detoxified  alcoholics  and  eye- 
tracking  impairment  of  schizophrenics. 
The  presence  of  both  sites  of  vermian 
dysmorphology  in  the  comorbid  group 
puts  them  at  risk  for  both  types  of 
motor  compromise. 

Other  diseases  with  a  high  incidence 
of  alcohol  abuse  and  dependence  are 
bipolar  disorder  (Regier  et  al.  1990)  and 
HIV  infection  (Schleifer  et  al.  1996). 
Anonymous  HIV  testing  at  an  inner- 
city  outpatient  alcohol  treatment  center 
revealed  that  16.7  percent  of  patients 
exclusively  abusing  alcohol  were  HIV 
seropositive.  HIV  infection  can  result  in 
progressive  ventricular  volume  enlarge- 
ment, reduced  volume  of  white  matter, 
and  caudate  atrophy  as  the  disease  pro- 
gresses (Stout  et  al.  1998).  Some  (Lim 
et  al.  1999)  but  not  all  (Schlaepfer  et 
al.  1994;  Zipursky  et  al.  1997)  MRI 
studies  have  detected  cortical  gray  mat- 
ter volume  deficits  in  patients  with  bipo- 
lar disorder  without  alcohol  dependence. 
Although  both  HIV  infection  and 
bipolar  disease  may  have  at  least  some 
degree  of  associated  brain  pathology, 
few  studies  have  rigorously  examined 
the  added  effect  of  alcoholism.  Those 
that  have  typically  report  a  combined 
effect  of  HIV  and  excessive  alcohol 
consumption.  These  studies  are  usually 
based  on  infected  individuals  who  are 
still  drinking  heavily  and  thus  reflect 


491 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


the  combined  acute  and  chronic 
effects  of  alcohol  consumption. 

Polydrug  Abuse 

Studies  of  structural  brain  abnormalities 
in  polydrug  abusers  tend  to  have  neg- 
ative results  (Liu  et  al.  1995),  unless 
alcohol  is  a  significant  ingredient  in 
the  mix  of  drugs  abused  (Cascella  et 
al.  1991;  Aasly  et  al.  1993).  In  one 
study,  statistical  analysis  of  the  role  of 
severity  of  use  of  different  drugs 
abused  found  that,  after  taking  age  into 
account,  only  alcohol  contributed  addi- 
tionally to  ventricular  enlargement 
(Cascella  et  al.  1991).  In  another  study, 
the  authors  attributed  findings  of 
reduced  cerebellar  vermis  and  increased 
incidence  of  clinically  defined  white 
matter  abnormalities  in  polydrug  abusers 
to  alcohol  consumption  in  their  sam- 
ple (Aasly  et  al.  1993).  Thus,  abuse  of 
other  drugs  provides  no  protection 
against  the  known  effects  of  alcohol 
on  brain  structure.  Whether  the  use 
of  drugs  such  as  cocaine,  marijuana, 
or  barbiturates  potentiates  the  brain's 
vulnerability  to  alcohol  has  not  been 
adequately  studied. 

HOW  DO  STRUCTURAL 
AND  FUNCTIONAL  BRAIN 
ABNORMALITIES 
PREDISPOSE  ALCOHOLICS 
TO  BEHAVIORAL  AND 
NEUROPSYCHOLOGICAL 
PROBLEMS? 

Neuronal  Shrinkage 
Versus  Loss 

Gray  matter  volume  reduction  could 
be  indicative  of  shrinkage  of  neuronal 


cell  bodies  or  dendrites  or  actual  neu- 
ronal loss.  When  postmortem  studies 
have  documented  neuronal  loss,  it 
usually  affects  large  cells,  such  as 
pyramidal  cells  found  in  the  frontal 
lobes  and  in  cerebellum  (Courville 
1955;  Harper  and  Kril  1990,  1993; 
Pentney  1993;  but  see  Badsberg- 
Jensen  and  Pakkenberg  1993).  In 
animal  models  (mouse),  long-term 
ethanol  consumption  produces  loss  of 
dendritic  spines  on  hippocampal  pyra- 
midal cells  and  dentate  granule  cells 
(Riley  and  Walker  1978;  Davies  and 
Smith  1981)  and  significant  loss  of 
pyramidal  and  dentate  gyral  cells  even 
in  the  absence  of  malnutrition  (Walker 
et  al.  1980).  Conversely,  one  study 
showed  that  abstinence  following  a 
period  of  ethanol  intake  was  accompa- 
nied by  increases  in  dendritic  arboriza- 
tion of  hippocampal  pyramidal  cells 
(McMullen  et  al.  1984).  In  humans, 
decreased  basal  dendritic  arborization 
of  layer  III  pyramidal  neurons  was 
observed  in  alcoholic  compared  with 
normal,  nonalcoholic  cases  (Harper 
and  Corbett  1990).  The  critical  dif- 
ference between  these  mechanisms  of 
volume  reduction  is  that  cell  loss  is 
presumably  permanent,  whereas  cell 
size  reduction  may  well  be  transient 
and  amenable  to  reversal  with  absti- 
nence from  alcohol  or  with  adequate 
nutrition  or  other  treatment. 

Postmortem  studies  have  shown 
white  matter  volume  reduction  and 
could  reflect  demyelination,  axonal 
shrinkage,  or  loss  of  fibers  due  to  cell 
degeneration  (Harper  and  Kril  1990). 
Longitudinal  in  vivo  studies  following 
the  course  of  alcoholism  show 
that  white  matter  volume  may  be 


492 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


recoverable,  at  least  in  part,  with  alco- 
hol abstinence  (Shear  et  al.  1994; 
Pfefferbaum  et  al.  1995). 

Regrowth  of  neuronal  processes 
does  not  ensure  normal  "rewiring" 
and  restitution  of  function  (see,  e.g., 
Hill  and  Mikhael  1979).  Thus,  neuro- 
psychological studies  are  essential  for 
testing  whether,  with  abstinence, 
alcoholics  whose  brains  show  recov- 
ery of  tissue  volume  are  the  individu- 
als who  also  show  improvement  in 
their  cognitive  and  motor  abilities 
(see,  e.g.,  Eckardt  et  al.  1979;  Brandt 
et  al.  1983;  Parsons  1987^).  A  recent 
naturalistic,  longitudinal  study,  which 
used  quantitative  neuroimaging  and 
neuropsychological  methods,  exam- 
ined alcoholics  who  returned  to  the 
community  following  a  2 8 -day  reha- 
bilitation program.  Alcoholics  who 
maintained  sobriety  sustained  greater 
improvement  than  relapsers  on  tests 
of  nonverbal  memory,  visuospatial 
functions,  attention,  gait,  and  balance 
(Sullivan  et  al.  in  press).  In  general, 
improvement  in  cognitive  and  motor 
functions  was  related  to  improvement 
in  the  condition  of  the  brain;  in  partic- 
ular, increases  in  scores  on  short-term 
memory  tests  correlated  with  shrink- 
age of  the  third  ventricle.  Neuronal 
recovery  could  also  be  inferred  from 
proton  MRSI  studies  if  increases  in 
tissue  volumes  were  accompanied  by 
further  increases  in  gray  matter  or 
white  matter  NAc  concentrations, 
which  would  occur  with  sobriety. 
Such  recovery  would  suggest  that  the 
remaining  cell  bodies,  although  once 
shrunken,  were  resilient  and  viable  (cf. 
Badsberg-Jensen  and  Pakkenberg 
1993).  Whether  such  plasticity  is 


curtailed  by  the  aging  process  (Ron  et 
al.  1982;  Carlen  et  al.  1984)  also 
remains  to  be  established. 

Discontinuous  Syndromes 
or  Continuous  Brain 
Abnormalities 

In  vivo  neuroimaging  has  enabled  us 
to  identify  brain  structural  abnormalities 
in  chronic  alcoholics  without  clinical 
symptoms  of  severe  liver  disease,  amne- 
sia, or  alcoholic  dementia  that  previ- 
ously had  been  considered  hallmarks 
of  specific  subsyndromes,  such  as 
Wernicke -Korsakoff  syndrome  (mam- 
millary  body  and  possibly  hippocampal 
shrinkage),  Marchiafava-Bignami  disease 
(thinning  of  corpus  callosum),  and 
alcoholic  cerebellar  degeneration  with 
ataxia  (figure  6).  These  observations 
were  made  possible  through  survey, 


Figure  6.  Alcoholism  and  its  nutritional 
and  genetic  attendant  and  disposing  factors 
appear  central  to  specific  neuropsychologi- 
cal and  neuropathological  syndromes. 
However,  many  of  the  same  brain  abnor- 
malities characterizing  these  syndromes  are 
also  found  with  in  vivo  neuroimaging  in 
"uncomplicated"  alcoholism. 


493 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


controlled  quantification,  and  com- 
parison of  large  regions  and  multiple 
brain  structures  that  yielded  patterns 
of  normality  and  abnormality.  Rather 
than  there  being  classical  alcoholic 
subsyndromes,  the  findings  suggest  a 
continuum  of  graded  brain  dysmor- 
phology,  whereby  lesions  in  specific 
brain  regions  usually  leading  to  syn- 
dromes of  selective  cognitive  or  motor 
deficits  occur  in  clinically  unsymptomatic 
alcoholics  with  far  greater  frequency 
than  previously  thought. 

What  needs  to  be  established  is 
whether  these  brain  volume  deficits 
are  accompanied  by  predictable  neuro- 
psychological symptoms.  Furthermore, 
although  alcoholic  dementia  has  been 
seldom  studied  with  imaging  tech- 
niques, one  hypothesis  would  be  that 
alcoholic  dementia  results  from  the 
cumulative  effect  of  volume  deficits  in 
multiple  brain  structures,  each  of  which 
traditionally  define  only  selective  syn- 
dromes. The  question  of  gene-envi- 
ronment interaction  with  respect  to 
apolipoprotein  genotype  (as  seen  in 
Alzheimer's  disease  [Strittmatter  and 
Roses  1996])  and  alcohol  on  the  devel- 
opment of  dementia  and  accelerated 
brain  volume  loss  in  alcoholics  has  yet 
to  be  addressed. 

Functional  Outcome  of 
Frontal- Cerebellar  Circuit 
Disruption 

Converging  evidence  from  every 
imaging  modality,  neuropsychological 
examination,  and  neuropathological 
study  identifies  the  frontal  lobes  and 
the  cerebellum  as  principal  targets  of 
alcohol-related  corruption  of  brain 
structure  and  function.  The  extent  to 


which  these  regions  recover  in  structure 
or  function  remains  unknown  despite 
their  importance  to  problem  solving, 
contextual  memory,  and  execution 
and  learning  of  procedures.  A  recent 
brain- behavior  analysis  revealed  that 
lower  scores  on  test  composites  assessing 
executive  function  and  motor  function, 
gait,  and  balance  showed  independent 
relationships  with  white  matter  volumes 
of  the  anterior  superior  vermis  of  the 
cerebellum  (Sullivan  et  al.  1998#). 
Thus,  vermian  volume  deficits  in  alco- 
holics appear  to  have  functional  rami- 
fications for  cognitive  executive  function 
as  well  as  motoric  functions,  raising 
the  possibility  of  disruption  of  frontal  - 
cerebellar  circuitry  in  alcoholism.  The 
presence  of  cerebellar  structural  abnor- 
malities might  also  set  limits  on  the  type 
of  procedural  retraining  and  learning 
of  new  habits  required  to  maintain 
sobriety  in  alcoholics  (Finn  et  al. 
1990)  and  could  contribute  to  problem 
solving  and  working  memory  deficits 
known  to  persistent  in  long-detoxified 
alcoholics  (Butters  and  Cermak  1980; 
Becker  et  al.  1983;  Brandt  et  al.  1983; 
Nixon  etal.  1992). 

Models  of  Aging 

Despite  the  usefulness  of  the  cross- 
sectional  age-regression  method  for  esti- 
mating the  effects  of  age,  it  remains  essen- 
tial to  verify  cross-sectional  observations 
with  those  from  controlled,  longitudinal 
study.  Although  longitudinal  neuroimag- 
ing  studies  of  alcoholics  have  been 
reported,  they  typically  focus  on  issues  of 
relapse  and  abstinence.  Still  needed  are 
controlled  studies  of  alcoholics  who 
remain  abstinent  for  years  in  comparison 
to  those  who  drink  for  years.  It  would 


494 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


be  of  great  importance  to  know,  for 
example,  whether  treatment  of  and 
recovery  from  early-onset  alcoholism  at 
a  young  age  can  adequately  protect  the 
individual  from  the  usual  alcohol-related 
accelerated  brain  aging,  or  whether  the 
early  bout  with  alcoholism  reduces  the 
margin  of  safety  thought  to  be  available 
to  pathology- free  individuals.  Other 
conditions,  such  as  polio  (Cosgrove  et  al. 
1987)  and  head  injury  (Corkin  et  al. 
1984,  1989),  do  increase  the  risk  of 
exacerbated  functional  declines  with  age. 
Early  alcoholism  may  be  an  added  factor 
of  vulnerability  and  may  interact  with 
genetic  underpinnings  to  accelerate  aging. 
For  instance,  in  a  study  of  monozygotic 
(MZ)  and  dizygotic  (DZ)  elderly  normal 
(nonalcoholic)  twin  pairs,  intracranial 
volume  and  age-corrected  cortical  sulcal 
volume,  but  not  ventricular  volume,  were 
highly  heritable,  with  evidence  for 
additive  genetic  variance.  However, 
highly  significant  MZ  but  not  DZ  intra- 
class  correlation  for  ventricular  volumes 
suggested  a  smaller  role  for  additive 
genetic  effects  and  a  larger  role  for  gene- 
gene  interaction  and/or  shared  environ- 
mental effects  on  this  morphometric 
measure  (Pfefferbaum  et  al  in  press). 
To  date,  no  quantitative  in  vivo  studies 
have  been  conducted  to  examine,  even 
cross-sectionally,  potential  alcohol-gene 
interactions  on  brain  morphology. 
These  studies  are  essential  to  deter- 
mine the  pattern  and  extent  of  poten- 
tially preexisting  differences  in 
individuals  genetically  predisposed  to 
alcohol  addiction  compared  with  those 
not  so  predisposed. 

The  severity  of  the  brain  dysmorphol- 
ogy  observed  in  chronic  alcoholics,  and 
possibly  the  degree  of  recovery  with 


abstinence,  may  also  be  related  to 
demographic  and  drinking  factors, 
including  age,  duration  of  alcoholism, 
and  lifetime  alcohol  consumption. 
Although  far  more  longitudinal  studies 
using  structural  than  functional  imag- 
ing techniques  have  been  conducted, 
cohorts  studied  to  date  have  not  been 
large  and  diverse  enough  to  adequately 
address  these  possibilities. 

CONCLUSION 

Human  in  vivo  neuroimaging  studies 
have  characterized  gross  dysmorphol- 
ogy  associated  with  alcoholism  and  in 
excess  of  what  is  expected  in  normal 
aging.  Even  in  "uncomplicated  alco- 
holism," significant  brain  tissue  volume 
deficits  are  present  in  brain  structures 
previously  thought  to  be  affected  only  in 
certain  alcohol-induced  subsyndromes. 
Regional  brain  metabolism  and  selec- 
tive neuroreceptor  systems  are  also 
affected  in  alcoholism.  These  struc- 
tural and  functional  abnormalities 
wax  and  wane  with  abstinence  and 
relapse,  but  exactly  how  they  pro- 
mote cognitive,  sensory,  and  motor 
dysfunction  and  whether  the  degree 
of  recovery  has  diminishing  returns 
with  advancing  age  and  recurrent 
bouts  of  alcoholism  remain  unknown. 
Furthering  knowledge  of  these  neural 
functions  and  mechanisms  may  lead 
to  rational  approaches  for  behavioral 
and  pharmacological  treatment  of 
alcoholism  and  for  its  prevention. 
Combining  structural  and  functional 
imaging  modalities  provides  the 
opportunity  to  identify  both  mecha- 
nisms of  vulnerability  and  points 
of  damage,  some  of  which  may  be 


495 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


uniquely  human  and  present  only      Mental  Disorders.  4th  ed.  Washington, 
in  life.  DC:  the  Association,  1994. 


ACKNOWLEDGMENTS 

This  work  was  supported  by  National 
Institute  on  Alcohol  Abuse  and  Alcohol- 
ism grants  AA10723  and  AA05965.  I 
would  like  to  thank  Adolf  Pfefferbaum, 
M.D.,  my  collaborator  and  mentor  in 
imaging  and  alcoholism,  for  the  mag- 
netic resonance  images  and  other  incal- 
culable contributions  to  this  chapter.  I 
also  thank  Daniel  Spielman,  Ph.D., 
for  the  proton  spectra  and  Margaret  J. 
Rosenbloom,  M.A.,  for  helpful  com- 
ments on  the  manuscript. 

REFERENCES 

Aasly,  J.;  Storsaeter,  O.;  Nilsen,  G.;  Smevik, 
O.;  and  Rinck,  P.  Minor  structural  brain 
changes  in  young  drug  abusers.  A  magnetic 
resonance  study.  Acta  Neurol  Scand 
87:210-214,  1993. 

Acker,  W.;  Aps,  E.J.;  Majumdar,  S.K.; 
Shaw,  G.K.;  and  Thomson,  A.D.  The 
relationship  between  brain  and  liver 
damage  in  chronic  alcoholic  patients. 
/  Neurol  Neurosurg  Psychiatry  45 : 
984-987, 1982. 

Adams,  K.M.;  Gilman,  S.;  Johnson- 
Greene,  D.;  Koeppe,  R.A.;  Junck,  L.; 
Kluin,  K.J.;  Martorello,  S.;  Johnson,  M.J.; 
Heumann,  M.;  and  Hill,  E.  The  signifi- 
cance of  family  history  status  in  relation  to 
neuropsychological  test  performance  and 
cerebral  glucose  metabolism  studied  with 
positron  emission  tomography  in  older 
alcoholic  patients.  Alcohol  Clin  Exp  Res 
22:105-110,  1998. 

American  Psychiatric  Association. 
Diagnostic  and  Statistical  Manual  of 


Anton,  R.F.,  and  Becker,  H.C.  Pharma- 
cology and  pathophysiology  of  alcohol 
withdrawal.  In:  Kranzler,  H.R.,  ed.  The 
Pharmacology  of  Alcohol  Abuse.  Berlin: 
Springer- Verlag,  1995.  pp.  315-367. 

Artmann,  H.;  Gall,  M.V.;  Hacker,  H.;  and 
Herrlich,  J.  Reversible  enlargement  of  cere- 
bral spinal  fluid  spaces  in  chronic  alcoholics. 

Am  J  Neuroradiol  2:23-27 ',  1981. 

Ashley,  M.J.;  Olin,  J.S.;  and  le  Riche, 
W.H.  Morbidity  in  alcoholics:  Evidence 
for  accelerated  development  of  physical 
disease  in  women.  Arch  Intern  Med 
137:883-887,  1977. 

BadsbergTensen,  G.,  and  Pakkenberg,  B. 
Do  alcoholics  drink  their  neurons  away? 
Lancet  342:1201-1204,  1993. 

Ballenger,  J.C.,  and  Post,  R.M.  Kindling 
as  a  model  for  alcohol  withdrawal  syn- 
dromes. Br  J  Psychiatry  133:1-14,  1978. 

Barker,  P.B.;  Breiter,  S.N.;  and  Soher, 
B.J.  Quantitative  proton  spectroscopy  of 
canine  brain:  In  vivo  and  in  vitro  correla- 
tions. Magn  Reson  Med  32:157-163, 1994. 


Becker,  J.T.;  Butters,  N.;  Hermann,  A.; 
and  D'Angelo,  N.  A  comparison  of  the 
effects  of  long-term  alcohol  abuse  and 
aging  on  the  performance  of  verbal  and 
nonverbal  divided  attention  tasks.  Alcohol 
Clin  Exp  Res  7 -.213-219,  1983. 

Belzunegui,  T.;  Insausti,  R;  Ibanez,  J.;  and 
Gonzalo,  L.M.  Effect  of  chronic  alcoholism 
on  neuronal  nuclear  size  and  neuronal 
population  in  the  mammillary  body  and 
the  anterior  thalamic  complex  of  man. 
Histol  Histopathol  10:633-638,  1995. 

Blatter,  D.D.;  Bigler,  E.D.;  Gale,  S.D.; 
lohnson,  S.C.;  Anderson,  C;  Burnett, 
B.M.;  Parker,  N.;  Kurth,  S.;  and  Horn,  S. 


496 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Quantitative  volumetric  analysis  of  brain 
MRI:  Normative  database  spanning  five 
decades  of  life.  Am  J  Neuroradiol 
16:241-245,  1995. 

Booth,  B.M.,  and  Blow,  F.C.  The  kindling 
hypothesis:  Further  evidence  from  a  U.S. 
national  study  of  alcoholic  men.  Alcohol 
Alcohol  28:593-598, 1993. 

Brand,  A.;  Richter-Landsberg,  C;  and 
Leibfritz,  D.  NMR  studies  on  the  energy 
metabolism  of  glial  and  neuronal  cells. 
Dev  Neurosci  15:289-298,  1993. 

Brandt,  J.;  Butters,  N.;  Ryan,  C;  and 
Bayog,  R.  Cognitive  loss  and  recovery  in 
long-term  alcohol  abusers.  Arch  Gen 
Psychiatry  40:435-442,  1983. 

Brower,  K.;  Mudd,  S.;  Blow,  F.;  Young, 
J.;  and  Hill,  E.  Severity  and  treatment  of 
alcohol  withdrawal  in  elderly  versus 
younger  patients.  Alcohol  Clin  Exp  Res 
18:196-201,  1994. 

Brown,  M.E.;  Anton,  R.F.;  Malcolm,  R.; 
and  Ballenger,  J.C.  Alcohol  detoxification 
and  withdrawal  seizures:  Clinical  support 
for  a  kindling  hypothesis.  Biol  Psychiatry 
23:507-514,  1988. 

Butters,  N.  Alcoholic  Korsakoff's  syndrome: 
Some  unresolved  issues  concerning  etiology, 
neuropathology,  and  cognitive  deficits.  / 
Clin  Exp  Neuropsychol  7:181-210,  1985. 

Butters,  N.,  and  Cermak,  L.S.  Alcoholic 
Korsakoff's  Syndrome.  New  York:  Academic 
Press,  1980. 

Butters,  N.,  and  Stuss,  D.T.  Diencephalic 
amnesia.  In:  Boiler,  F.,  and  Grafman,  J., 
eds.  Handbook  of  Neuropsychology.  Vol.  3. 
Amsterdam:  Elsevier,  1989.  pp.  107-148. 

Butterworth,  R.F.;  Kril,  J.J.;  and  Harper, 
C.G.  Thiamine-dependent  enzyme 
changes  in  the  brains  of  alcoholics — 
relationship  to  the  Wernicke -Korsakoff 


syndrome.  Alcohol  Clin  Exp  Res 
17:1084-1088,  1993. 

Carlen,  P.L.;  Wortzman,  G.;  Holgate, 
R.C.;  Wilkinson,  D.A.;  and  Rankin,  J.G. 
Reversible  cerebral  atrophy  in  recently 
abstinent  chronic  alcoholics  measured  by 
computed  tomography  scans.  Science 
200:1076-1078,  1978. 

Carlen,  P.L.;  Wilkinson,  D.A.;  Wortzman, 
G.;  and  Holgate,  R.  Partially  reversible 
cerebral  atrophy  and  functional  improve- 
ment in  recently  abstinent  alcoholics.  Can 
J  Neurol  Sci  11:441-446,  1984. 

Cascella,  N.G.;  Pearlson,  G.;  Wong,  D.F.; 
Broussolle,  E.;  Nagoshi,  C;  Margolin, 
R.A.;  and  London,  E.D.  Effects  of  substance 
abuse  on  ventricular  and  sulcal  measures 
assessed  by  computerised  tomography.  Br 
J  Psychiatry  159:217-221,  1991. 

Chan,  A.W.K.  Alcoholism  and  epilepsy. 
Epilepsia  26:323-333,  1985. 

Charness,  M.E.  Brain  lesions  in  alcoholics. 
Alcohol  Clin  Exp  Res  17:2-11,  1993. 

Chiu,  T.;  Mendelson,  J.;  Woods,  B.;  Teoh, 
S.;  Levisohn,  L.;  and  Mello,  N.  In  vivo 
proton  magnetic  resonance  spectroscopy 
detection  of  human  alcohol  tolerance. 
Magn  Reson  Med  32:511-516,  1994. 

Clarren,  S.K.  Neuropathology  in  fetal  alco- 
hol syndrome.  In:  West,  J.R.,  ed.  Alcohol 
and  Brain  Development.  New  York:  Oxford 
University  Press,  1986.  pp.  158-166. 

Coffey,  C.E.;  Lucke,  J.E.;  Saxton,  J.A.; 
Ratcliff,  G.;  Unitas,  L.J.;  Billig,  B.;  and 
Bryan,  R.N.  Sex  differences  in  brain  aging: 
A  quantitative  magnetic  resonance  imaging 
study.  Arch  Neurol  55:169-179,  1998. 

Corkin,  S.;  Sullivan,  E.V.;  and  Carr,  F.A. 
Prognostic  factors  for  life  expectancy  after 
penetrating  head  injury.  Arch  Neurol 
41:975-977,  1984. 


497 


-^ 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Corkin,  S.;  Rosen,  J.;  Sullivan,  E.V.;  and 
Clegg,  RA.  Penetrating  head  injury  in 
young  adulthood  exacerbates  cognitive 
decline  in  later  years.  /  Neurosci  9:3876- 
3883, 1989. 

Cosgrove,  J.;  Alexander,  M.;  Kitts,  E.; 
Swan,  B.;  Klein,  M.;  and  Bauer,  R  Late 
effects  of  poliomyelitis.  Arch  Phys  Med 
Rehabil  68:4-7, 1987. 

Coulter,  C;  Leech,  R.W.;  Schaefer,  B.; 
Scheithauer,  B.W.;  and  Brumback,  R.A. 
Midline  cerebral  dysgenesis,  disfunction 
of  the  hypothalamic-pituitary  axis  and 
fetal  alcohol  effects.  Arch  Neurol 
50:771-775,  1993. 

Courville,  C.B.  Effects  of  Alcohol  on  the 
Nervous  System  of  Man.  Los  Angeles:  San 
Lucas  Press,  1955. 

Daryanani,  H.E.;  Santolaria,  F.J.;  Reimers, 
E.G.;  Jorge,  J.A.;  Lopez,  N.B.;  Hernandez, 
F.M.;  Riera,  A.M.;  and  Rodriguez,  E.R. 
Alcohol  withdrawal  syndrome  and  seizures. 
Alcohol  Alcohol  29:323-328,  1994. 

Davies,  A.R,  and  Smith,  D.E.  A  golgi  study 
of  mouse  hippocampal  CA1  pyramidal 
neurons  following  perinatal  ethanol  expo- 
sure. Neurosci  Lett  26:49-54,  1981. 

Davila,  M.D.;  Shear,  P.K;  Lane,  B.;  Sullivan, 
E.V.;  and  Pfefferbaum,  A  Mammillary  body 
and  cerebellar  shrinkage  in  chronic  alco- 
holics: An  MRI  and  neuropsychological 
study.  Neuropsychology  8:433^1:4:4:,  1994. 

Di  Sclafani,  V.;  Ezekiel,  F.;  Meyerhoff, 
D.J.;  Mackay,  S.;  Dillon,  W.P.;  Weiner, 
M.W.;  and  Fein,  G.  Brain  atrophy  and 
cognitive  function  in  older  abstinent 
alcoholic  men.  Alcohol  Clin  Exp  Res 
19:1121-1126,1995. 

Dorst,  J. P.  Changes  of  the  skull  during 
childhood.  In:  Newton,  T.H.,  and  Potts, 
D.G.,  eds.  Radiology  of  the  Skull  and 


Brain:  The  Skull.  St.  Louis,  MO:  C.V. 
Mosby,  1971.  pp.  118-131. 

Drake,  A.I.;  Jernigan,  T.L.;  Butters,  N.; 
Shear,  P.K.;  and  Archibald,  S.L.  Volumetric 
changes  on  magnetic  resonance  imaging 
in  chronic  alcoholics:  A  one  year  followup 
(abstract).  Paper  presented  at  the  Twenty- 
Second  Annual  Meeting  of  the  International 
Neuropsychological  Society,  Cincinnati, 
Ohio,  1994. 

Dupont,  KM.;  Rourke,  S.B.;  Grant,  L; 
Lehr,  P.P.;  Reed,  R.J.;  Challakere,  K.; 
Lamoureux,  G.;  and  Halpern,  S.  Single 
photon  emission  computed  tomography 
with  iodoamphetamine-123  and  neu- 
ropsychological studies  in  long-term 
abstinent  alcoholics.  Psychiatry  Res 
Neuroimaging  67:99-111,  1996. 

Eckardt,  M.J.;  Parker,  E.S.;  Noble,  E.P.; 
Pautler,  C.P.;  and  Gottschalk,  L.A.  Changes 
in  neuropsychological  performance  during 
treatment  for  alcoholism.  Biol  Psychiatry 
14:943-954,  1979. 

Ernst,  T.;  Chang,  L.;  Melchor,  R;  and 
Mehringer,  CM.  Frontotemporal  dementia 
and  early  Alzheimer  disease:  Differentiation 
with  frontal  lobe  H-l  NTR  spectroscopy. 
Radiology  203:829-836,  1997. 

Estruch,  R;  Nicolas,  J.M.;  Villegas,  E.; 
Junque,  A.;  and  Urbano-Marquez,  A. 
Relationship  between  ethanol-related 
diseases  and  nutritional  status  in  chronically 
alcoholic  men.  Alcohol  Alcohol  28:543- 
550, 1993. 

Fama,  R;  Sullivan,  E.V.;  Shear,  P.K.; 
Marsh,  L.;  Yesavage,  J.A.;  Tinklenberg, 
J.R.;  Lim,  K.O.;  and  Pfefferbaum,  A. 
Selective  cortical  and  hippocampal  volume 
correlates  of  Mattis  Dementia  Rating 
Scale  in  Alzheimer  disease.  Arch  Neurol 
54:719-728,  1997. 


498 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Fein,  G.;  Meyerhoff,  D.J.;  Di  Sclafani,  V.; 
Ezekiel,  F.;  Poole,  N.;  MacKay,  S.;  Dillon, 
W.P.;  Constans,  J.-M.;  and  Weiner,  M.W. 
1H  magnetic  resonance  spectroscopic 
imaging  separates  neuronal  from  glial 
changes  in  alcohol-related  brain  atrophy. 
In:  Lancaster,  F.E.,  ed.  Alcohol  and  Glial 
Cells.  National  Institute  on  Alcohol  Abuse 
and  Alcoholism  Research  Monograph  No. 
27.  NIH  Pub.  No.  94-3742.  Bethesda, 
MD:  the  Institute,  1994.  pp.  227-241. 

Fein,  G.;  Meyerhoff,  D.J.;  and  Weiner, 
M.W.  Magnetic  resonance  spectroscopy 
of  the  brain  in  alcohol  abuse.  Alcohol 
Health  Res  World  19:306-314,  1995. 

Fillmore,  KM.;  Golding,  J.M.;  Kneip,  S.; 
Leino,  E.V.;  Shoemaker,  C.;  Ager,  C.R.; 
and  Ferrer,  H.P.  Gender  differences  for 
the  risk  of  alcohol-related  problems  in 
multiple  national  contexts.  In:  Galanter, 
M.,  ed.  Recent  Developments  in  Alcoholism, 
Vol  12:  Alcoholism  and  Women.  New 
York:  Plenum  Press,  1995.  pp.  410^40. 

Finn,  P.R.;  Earleywine,  M.;  and  Ramsey, 
S.  Response  to  relevant,  irrelevant  and  novel 
stimuli:  The  effects  of  alcohol  and  risk 
status.  Alcohol  Clin  Exp  Res  14:288,  1990. 

Fox,  J.H.;  Ramsey,  R.G.;  Huckman,  M.S.; 
and  Proske,  A.E.  Cerebral  ventricular 
enlargement,  chronic  alcoholics  examined 
by  computerized  tomography.  JAMA 
236:365-368,  1976. 

Frezza,  M.;  di  Padova,  C.;  Pozzato,  G.; 
Terpin,  M.;  Baraona,  E.;  and  Lieber,  C.S. 
High  blood  alcohol  levels  in  women.  N 
Engl  J  Med  322:95-99,  1990. 

Gabrieli,  J.D.;  Sullivan,  E.V.;  Desmond, 
J.E.;  Stebbins,  G.T.;  Daidya,  C.J.;  Keane, 
M.M.;  Wagner,  A.D.;  Sarella,  M.M.; 
Glover,  G.H.;  and  Pfefferbaum,  A. 
Behavioral  and  functional  neuroimaging 
evidence  for  preserved  conceptual  implicit 
memory  in  global  amnesia.  In:  Society  for 


Neuroscience  Abstracts,  Vol.  22,  Part  2. 
Washington,  DC:  Society  for  Neuroscience, 
1996.  Abstract  575.6,  p.  1449. 

Gilman,  S.;  Adams,  K;  Koeppe,  R.A.; 
Berent,  S.;  Khun,  K.J.;  Modell,  J.G.; 
Kroll,  P.;  and  Brunberg,  JA.  Cerebellar 
and  frontal  hypometabolism  in  alcoholic 
cerebellar  degeneration  studied  with 
positron  emission  tomography.  Ann 
Neurol  28:775-785,  1990. 

Gilman,  S.;  Adams,  KM.;  Johnson- 
Greene,  D.;  Koeppe,  R.A.;  Junck,  L.; 
Kluin,  K.J.;  Martorello,  S.;  Heumann,  N.; 
and  Hill,  E.  Effects  of  disulfiram  on 
positron  emission  tomography  and  neu- 
ropsychological studies  in  severe  chronic 
alcoholism.  Alcohol  Clin  Exp  Res 
20:1456-1461,  1996^. 

Gilman,  S.;  Koeppe,  R.A.;  Adams,  K; 
Johnson-Greene,  D.;  Junck,  L.;  Kluin, 
K.J.;  Brunberg,  J.;  Martorello,  S.;  and 
Lohman,  M.  Positron  emission  tomographic 
studies  of  cerebral  benzodiazepine -receptor 
binding  in  chronic  alcoholics.  Ann  Neurol 
40:163-171,  1996&. 

Goodlett,  C.R,  and  Lundahl,  KR  Temporal 
determinants  of  neonatal  alcohol-induced 
cerebellar  damage  and  motor  performance 
deficits.  Pharmacol  Biochem  Behav  55: 
531-540, 1996. 

Gur,  R.C.;  Mozley,  P.D.;  Resnick,  S.M.; 
Gottlieb,  G.L.;  Kohn,  M.;  Zimmerman, 
R;  Herman,  G.;  Atlas,  S.;  Grossman,  R; 
Berretta,  D.;  Erwin,  R.;  and  Gur,  R.E. 
Gender  differences  in  age  effect  on  brain 
atrophy  measured  by  magnetic  resonance 
imaging.  Proc  Natl  Acad  Sci  USA  88: 
2845-2849,  1991. 

Hanstock,  C.C.;  Rothman,  D.L.;  Shulman, 
R.G.;  Novotny,  E.J.;  Petroff,  O.A.C.;  and 
Prichard,  J.W.  Measurement  of  ethanol  in 
the  human  brain  using  NMR  spectroscopy. 
J  Stud  Alcohol  5 1:104-107,  1990. 


499 


NIAAAs  Neuroscience  and  Behavioral  Research  Portfolio 


Harper,  C,  and  Corbett,  D.  Changes  in 
the  basal  dendrites  of  cortical  pyramidal 
cells  from  alcoholic  patients — a  quantitative 
golgi  study.  J  Neurol  Neurosurg  Psychiatry 
53:856-861,  1990. 

Harper,  C.G.,  and  Kril,  J.J.  Corpus  cal- 
losal  thickness  in  alcoholics.  Br  J  Addict 
83:577-580,  1988. 

Harper,  C.G.,  and  Kril,  J.J.  Neuropathology 
of  alcoholism .  Alcohol  Alcohol  25:207- 
216,  1990. 

Harper,  C,  and  Kril,  J.  If  you  drink  your 
brain  will  shrink.  Neuropathological 
considerations.  Alcohol  Alcohol  Suppl 
1:375-380,  1991. 

Harper,  C.G.,  and  Kril,  J.J.  Neuropatho- 
logical changes  in  alcoholics.  In:  Hunt, 
W.A.,  and  Nixon,  S.J.,  eds.  Alcohol-Induced 
Brain  Damage.  National  Institute  on 
Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Pub.  No. 
93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  39-69. 

Harper,  C.G.;  Smith,  N.A.;  and  Kril,  J.J. 
The  effects  of  alcohol  on  the  female 
brain — a  neuropathological  study.  Alcohol 
Alcohol  25:445-448,  1990. 

Haubek,  A.,  and  Lee,  K.  Computed  to- 
mography in  alcoholic  cerebellar  atrophy. 
Neuroradiology  18:77-79,  1979. 

Hill,  S.Y.,  and  Mikhael,  M.  Computed 
tomography  scans  of  alcoholics:  Cerebral 
atrophy?  Science 204:1237-1238,  1979. 

Hilton,  M.E.  The  demographic  distribu- 
tion of  drinking  patterns  in  1984.  In:  Clark, 
W.B.,  and  Hilton,  M.E.,  eds.  Alcohol  in 
America:  Drinking  Practices  and  Problems. 
Albany,  NY:  State  University  of  New  York 
Press,  1991.  pp.  73-86. 

Hommer,  D.;  Momenan,  R;  Rawlings, 
R;  Ragan,  P.;  Williams,  W.;  Rio,  D.;  and 


Eckardt,  M.  Decreased  corpus  callosum 
size  among  alcoholic  women.  Arch  Neurol 
53:359-363,  1996. 

Hunter,  R;  McLuskie,  R;  Wyper,  D.; 
Patterson,  J.;  Christie,  J.E.;  Brooks,  D.N.; 
McCulloch,  J.;  Fink,  G.;  and  Goodwin, 
G.M.  The  pattern  of  function-related 
regional  cerebral  blood  flow  investigated 
by  single  photon  emission  tomography 
with  99mTc-HMPAO  in  patients  with 
presenile  Alzheimer's  disease  and  Korsakoff's 
psychosis.  Psychol  Med  19:847-855,  1989. 

Jacobson,  R.  The  contributions  of  sex  and 
drinking  history  to  the  CT  brain  scan 
changes  in  alcoholics.  Psychol  Med 
16:547-549,  1986. 

Jagannathan,  N.R.;  Desai,  N.G.;  and 
Raghunathan,  P.  Brain  metabolite  changes 
in  alcoholism:  An  in  vivo  proton  magnetic 
resonance  spectroscopy  (MRS)  study. 
Magn  Reson  Imaging  14:553-557,  1996. 

Jernigan,  T.L.,  and  Cermak,  L.S. 
Structural  basis  of  memory.  In:  Kertesz, 
A.,  ed.  Localization  and  Neuroimaging  in 
Neuropsychology.  New  York:  Academic 
Press,  1994.  pp.  599-620. 

Jernigan,  T.L.;  Butters,  N.;  DiTraglia,  G.; 
Schafer,  K.;  Smith,  T.;  Irwin,  M.;  Grant, 
I.;  Schuckit,  M.;  and  Cermak,  L.  Reduced 
cerebral  grey  matter  observed  in  alcoholics 
using  magnetic  resonance  imaging.  Alcohol 
Clin  Exp  Res  15:418-427,  1991a. 

Jernigan,  T.L.;  Salmon,  D.P.;  Butters,  N.; 
and  Hesselink,  J.R.  Cerebral  structure  on 
MRI.  2.  Specific  changes  in  Alzheimer's 
and  Huntington's  diseases.  Biol  Psychiatry 
29:68-81,  199U. 

Jernigan,  T.L.;  Schafer,  K.;  Butters,  N.; 
and  Cermak,  L.S.  Magnetic  resonance 
imaging  of  alcoholic  Korsakoff  patients. 
Neuropsychopharmaco  logy  4:175-186, 1991c. 


500 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Joyce,  E.M.;  Rio,  D.E.;  Ruttimann,  U.E.; 
Rohrbaugh,  J.W.;  Martin,  P.R;  Rawlings, 
R.R.;  and  Eckardt,  M.J.  Decreased  cingu- 
late  and  precuneate  glucose  utilization  in 
alcoholic  Korsakoff's  syndrome.  Psychiatry 
Res  54:225-239, 1994. 

Kendler,  K.S.;  Gallagher,  T.J.;  Abelson,  J.M.; 
and  Kessler,  RC.  Lifetime  prevalence, 
demographic  risk  factors,  and  diagnostic 
validity  of  nonaffective  psychosis  as  assessed 
in  a  US  community  sample — the  national 
comorbidity  survey.  Arch  Gen  Psychiatry 
53:1022-1031,  1996. 

Kessler,  RC;  McGonagle,  K.A.;  Zhao, 
S.Y.;  Nelson,  C.H.;  Hughes,  M.;  Eshleman, 
S.;  Wittchen,  H.U.;  and  Kendler,  K.S. 
Lifetime  and  12 -month  prevalence  of 
DSM-III-R  psychiatric  disorders  in  the 
United  States:  Results  from  the  National 
Comorbidity  Survey.  Arch  Gen  Psychiatry 
51:8-19,  1994. 

Kessler,  RC;  Nelson,  C.B.;  McGonagle, 
K.A.;  Edlund,  M.J.;  Frank,  RC;  and  Leaf, 
P.J.  The  epidemiology  of  co-occurring 
addictive  and  mental  disorders:  Implications 
for  prevention  and  service  utilization.  Am 
J  Orthopsychiatry  66:1 7-3 1,1996. 

Kraft,  C.L.;  Gescuk,  B.;  Woods,  B.T.;  Mello, 
N.K.;  Weiss,  R.D.;  and  Mendelson,  J.H. 
Brain  ventricular  size  in  female  alcoholics: 
An  MRI  study.  Alcohol  8:31-34,  1991. 

Kuruoglu,  A.C.;  Arikan,  Z.;  Vural,  C; 
Karatas,  M.;  Arac,  M.;  and  Isik,  E.  Single 
photon  emission  computerised  tomogra- 
phy in  chronic  alcoholism — antisocial 
personality  disorder  may  be  associated 
with  decreased  frontal  perfusion.  Br  J 
Psychiatry  169:348-354,  1996. 

Lancaster,  F.E.  Alcohol  and  white  matter 
development — a  review.  Alcohol  Clin  Exp 
Res  18:644-647,  1994a. 


Lancaster,  F.E.  Gender  differences  in  the 
brain:  Implications  for  the  study  of  human 
alcoholism.  Alcohol  Clin  Exp  Res  18:740- 
746,  1994£. 

Lancaster,  F.E.;  Philips,  S.M.;  Patsalos,  P.N.; 
and  Wiggins,  RC.  Brain  myelination  in 
the  offspring  of  ethanol-treated  rats:  In 
utero  versus  lactational  exposure  by  cross- 
fostering  offspring  of  control,  pairfed,  and 
ethanol  treated  dams.  Brain  Res  309: 
209-216,  1984. 

Langlais,  P.J.,  and  Savage,  L.M.  Thiamine 
deficiency  in  rats  produces  cognitive  and 
memory  deficits  on  spatial  tasks  that 
correlate  with  tissue  loss  in  diencephalon, 
cortex  and  white  matter.  Behav  Brain  Res 
68:75-89,  1995. 

Langlais,  P. J.,  and  Zhang,  S.X.  Cortical 
and  subcortical  white  matter  damage 
without  Wernicke's  encephalopathy  after 
recovery  from  thiamine  deficiency  in  the 
rat.  Alcohol  Clin  Exp  ito  21:434443, 1997. 

Lechtenberg,  R.,  and  Worner,  T.M. 
Seizure  risk  with  recurrent  alcohol  detoxi- 
fication. Arch  Neurol 47:535-538,  1990. 

Lechtenberg,  R.,  and  Worner,  T.M. 
Relative  kindling  effect  of  detoxification 
and  non-detoxification  admission  in  alco- 
holics. Alcohol  Alcohol  26:221-225,  1991. 

Lee,  PL;  Tarter,  J.;  Holbourn,  G.E.; 
Price,  RR;  Weinstein,  D.D.;  and  Martin, 
P.R  In  vivo  localized  proton  NMR  spec- 
troscopy of  thiamine -deficient  rat  brain. 
Magn  Reson  Med  34:313-318,  1995. 

Lien,  Y.;  Shapiro,  J.;  and  Chan,  L.  Effects 
of  hypernatremia  on  organic  brain  osmoles. 
J  Clin  Invest  85:1427-1435,  1990. 

Lim,  K.O.;  Rosenbloom,  M.J.;  Faustman, 
W.O.;  Sullivan,  E.V.;  and  Pfefferbaum,  A. 
Cortical  gray  matter  deficit  in  patients  with 
bipolar  disorder.  Schizophr  Res  40(3)  :2 19- 
227,  1999. 


501 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Lishman,  WA.  Alcohol  and  the  brain.  Br 
J  Psychiatry  156:635-644,  1990. 

Liu,  X.;  Phillips,  R.L.;  Resnick,  S.M.; 
Villemagne,  V.L.;  Stapleton,  J.M.;  and 
London,  E.D.  Magnetic  resonance  imag- 
ing reveals  no  ventriculomegaly  in  poly- 
drug  abusers.  Acta  Neurol  Scand 
92:83-90,  1995. 

Mann,  K.;  Batra,  A.;  Gunthner,  A.;  and 
Schroth,  G.  Do  women  develop  alcoholic 
brain  damage  more  readily  than  men? 
Alcohol  Clin  Exp  Res  16:1052-1056, 1992a. 

Mann,  K.;  Mundle,  G.;  Strayle,  M.;  and 
Schroth,  G.  The  reversibility  of  alcoholic 
brain  damage  is  not  due  to  rehydration:  A 
controlled  MRI  and  CT  study.  Alcohol 
Clin  Exp  Res  16:80  (abstract),  19926. 

Mann,  K.;  Dengler,  W.;  Klose,  U.;  Neagiele, 
T.;  and  Mundle,  G.  MR- spectroscopy  in 
alcoholic  brain  damage:  No  evidence  for 
water  shift  in  abstinence.  Alcohol  Clin  Exp 
Res  18:430  (abstract),  1994. 

Martin,  P.R;  Rio,  D.;  Adinoff,  B.; 
Johnson,  J.L.;  Bisserbe,  J.-C.;  Rawlings, 
R.R.;  Rohrbaugh,  J.W.;  Stapleton,  J.M.; 
and  Eckardt,  M.J.  Regional  cerebral  glu- 
cose utilization  in  chronic  organic  mental 
disorders  associated  with  alcoholism.  / 
Neuropsychiatry  Clin  Neurosci  4(2):  159- 
167,  1992. 

Martin,  P.R.;  Gibbs,  S.J.;  Nimmerrichter, 
A.A.;  Riddle,  W.R.;  Welch,  L.W.;  and 
Willcott,  M.R.  Brain  proton  magnetic 
resonance  spectroscopy  studies  in  recently 
abstinent  alcoholics.  Alcohol  Clin  Exp  Res 
19:1078-1082,  1995. 

Mathalon,  D.H.;  Sullivan,  E.V.;  Rawles, 
J.M.;  and  Pfefferbaum,  A.  Correction  for 
head  size  in  brain-imaging  measurements. 
Psychiatry  Res  Neuroimaging  50:121- 
139,  1993. 


Mathalon,  D.H.;  Sullivan,  E.V.;  Lim, 
K.O.;  and  Pfefferbaum,  A.  Gray  matter 
deficits  in  schizophrenia-alcoholism  co- 
morbidity. Biol  Psychiatry  37 :629  (ab- 
stract), 1995. 

Matsuda,  K.;  Yamaji,  S.;  Ishii,  K.;  Sasaki, 
M.;  Sakamoto,  S.;  Kitagaki,  H.;  Imamura, 
T.;  and  Mori,  E.  Regional  cerebral  blood 
flow  and  oxygen  metabolism  in  a  patient 
with  Korsakoff  syndrome.  Ann  Nucl  Med 
11:33-35,1997. 

Mattson,  S.;  Riley,  E.;  Jernigan,  T.; 
Ehlers,  C.;  Delis,  D.;  Jones,  K.;  Stern,  C.; 
Johnson,  K.;  Hesselink,  J.;  and  Bellugi, 
U.  Fetal  alcohol  syndrome:  A  case  report 
of  neuropsychological,  MRI  and  EEG 
assessment  of  two  children.  Alcohol  Clin 
Exp  Res  16:1001-1003,  1992. 

Mattson,  S.N.;  Jernigan,  T.L.;  and  Riley, 
E.P.  MRI  and  prenatal  alcohol  exposure. 
Alcohol  Health  Res  World  18:49-52, 1994. 

Mattson,  S.N.;  Riley,  E.P.;  Sowell,  E.R; 
Jernigan,  T.L.;  Sobel,  D.F.;  and  Jones, 
K.L.  A  decrease  in  the  size  of  the  basal 
ganglia  in  children  with  fetal  alcohol 
syndrome.  Alcohol  Clin  Exp  Res  20:1088- 
1093,  1996. 

McMullen,  PA.;  Saint-Cyr,  J.A.;  and 
Carlen,  P.L.  Morphological  alterations  in 
the  rat  CA1  hippocampal  pyramidal  cell 
dendrites  resulting  from  chronic  ethanol 
consumption  and  withdrawal.  J  Comp 
Neurol  225:111-118,  1984. 

Mendelson,  J.H.;  Woods,  B.T.;  Chiu,  T.- 
M.;  Mello,  N.K.;  Lukas,  S.E.;  Teoh,  S.K.; 
Sintavanarong,  P.;  Cochin,  J.;  Hopkins, 
M.A.;  and  Dobrosielski,  M.  In  vivo  proton 
magnetic  resonance  spectroscopy  of  alcohol 
in  human  brain.  Alcohol  7:443-448,  1990. 

Meyerhoff,  D.J.;  MacKay,  S.;  Sappey- 
Marinier,  D.;  Deicken,  R;  Calabrese,  G.; 
Dillon,  W.P.;  Weiner,  M.W.;  and  Fein,  G. 


502 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Effects  of  chronic  alcohol  abuse  and  HIV 
infection  on  brain  phosphorus  metabolites. 
Alcohol  Clin  Exp  Res  19:685-692,  1995. 

Meyer-Wahl,  J.G.,  and  Braun,  J.  Epileptic 
seizures  and  cerebral  atrophy  in  alcoholics. 
Neurology  228:17-23,  1982. 

Milner,  B.  Effects  of  different  brain 
lesions  on  card  sorting.  Arch  Neurol 
9:90-100,  1963. 

Miitzell,  S.  Computed  tomography  of  the 
brain,  hepatotoxic  drugs  and  high  alcohol 
consumption  in  male  alcoholic  patients  and 
a  random  sample  from  the  general  male  pop- 
ulation. UpsJMedSci  97:183-194,  1992. 

Muuronen,  A.;  Bergman,  H.;  Hindmarsh, 
T.;  and  Telakivi,  T.  Influence  of  improved 
drinking  habits  on  brain  atrophy  and 
cognitive  performance  in  alcoholic  patients: 
A  5 -year  follow-up  study.  Alcohol  Clin  Exp 
ito  13:137-141,  1989. 

Nicolas,  J.M.;  Catafau,  A.M.;  Estruch,  R.; 
Lomena,  F.J.;  Salamero,  M.;  Herranz,  R.; 
Monforte,  R.;  Cardenal,  C;  and  Urbano- 
Marquez,  A.  Regional  cerebral  blood  flow- 
SPECT  in  chronic  alcoholism:  Relation  to 
neuropsychological  testing.  /  Nucl  Med 
34:1452-1459, 1993. 

Nixon,  S.J.;  Errico,  A.L.;  Parsons,  O.A.; 
Leber,  W.R;  and  Kelley,  C.J.  The  role  of 
instructional  set  on  alcoholic  performance. 
Alcohol  Clin  Exp  Res  16:949-954,  1992. 

Paller,  K.A.;  Acharya,  A.;  Richardson, 
B.C.;  Plaisant,  O.;  Shimamura,  A.P.; 
Reed,  B.R;  and  Jagust,  W.J.  Functional 
neuroimaging  of  cortical  dysfunction  in 
alcoholic  Korsakoff's  syndrome.  J  Cogn 
Neurosci  9:277-293,  1997. 

Parker,  D.,  and  Harford,  T.  Gender-role 
attitudes,  job  competition  and  alcohol 
consumption  among  women  and  men. 
Alcohol  Clin  Exp  Res  16:159-165,  1992. 


Parsons,  O.A.  Do  neuropsychological 
deficits  predict  alcoholics  treatment  course 
and  recovery?  In:  Parsons,  O.A.;  Butters, 
N.;  and  Nathan,  P.E.,  eds.  Neuropsycho- 
logy of  Alcoholism:  Implications  for 
Diagnosis  and  Treatment.  New  York: 
Guilford  Press,  1987a.  pp.  273-290. 

Parsons,  O.A.  Neuropsychological  conse- 
quences of  alcohol  abuse:  Many  ques- 
tions— some  answers.  In:  Parsons,  O.A.; 
Butters,  N.;  and  Nathan,  P.E.,  eds. 
Neuropsychology  of  Alcoholism:  Implications 
for  Diagnosis  and  Treatment.  New  York: 
Guilford  Press,  1987b.  pp.  153-175. 

Parsons,  O.A.;  Butters,  N.;  and  Nathan, 
P.E.,  eds.  Neuropsychology  of  Alcoholism: 
Implications  for  Diagnosis  and  Treatment. 
New  York:  Guilford  Press,  1987. 

Patek,  A.J.  Alcohol,  malnutrition  and 
alcoholic  cirrhosis.  Am  J  Clin  Nutr 
32:1304-1312,  1979. 

Paul,  S.M.  GABA  and  glycine.  In:  Bloom, 
F.E.,  and  Kupfer,  D.J.,  eds.  Psych  o- 
pharmacology:  The  Fourth  Generation  of 
Progress.  New  York:  Raven  Press,  1995. 
pp.  87-94. 

Peiffer,  J.;  Majewski,  F.;  and  Fischbach, 
H.  Alcohol  embryo-  and  fetopathy  neu- 
ropathology of  children  and  3  fetuses.  / 
Neurol  Sci  41:125-137,  1979. 

Pentney,  RJ.  Alterations  in  the  structure 
of  the  cerebellum  after  long-term  ethanol 
consumption.  In:  Hunt,  W.A.,  and  Nixon, 
S.J.,  eds.  Alcohol-Induced  Brain  Damage. 
National  Institute  on  Alcohol  Abuse  and 
Alcoholism  Research  Monograph  No.  22. 
NIH  Pub.  No.  93-3549.  Rockville,  MD: 
the  Institute,  1993.  pp.  249-276. 

Petroff,  O.A.C.;  Pleban,  LA.;  and 
Spencer,  D.D.  Symbiosis  between  in  vivo 
and  in  vitro  NMR  spectroscopy:  The 
creatine,  isf-acetylaspartate,  glutamate  and 


503 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


GABA  content  of  the  epileptic  human 
brain.  Magn  Reson  Imaging  13:1197- 
1211,1995. 

Pfefferbaum,  A.,  and  Rosenbloom,  M.  In 
vivo  imaging  of  morphological  brain 
alterations  associated  with  alcoholism.  In: 
Hunt,  W.A.,  and  Nixon,  S  J.,  eds.  Alcohol- 
Induced  Brain  Damage.  National  Institute 
on  Alcohol  Abuse  and  Alcoholism  Research 
Monograph  No.  22.  NIH  Pub.  No. 
93-3549.  Rockville,  MD:  the  Institute, 
1993.  pp.  71-87. 

Pfefferbaum,  A.;  Zatz,  L.;  and  Jernigan, 
T.L.  Computer-interactive  method  for 
quantifying  cerebrospinal  fluid  and  tissue 
in  brain  CT  scans:  Effects  of  aging.  / 
Comput  Assist  Tomogr  10:571-578,  1986. 

Pfefferbaum,  A.;  Rosenbloom,  M.J.; 
Crusan,  K.;  and  Jernigan,  T.L.  Brain  CT 
changes  in  alcoholics.  The  effects  of  age 
and  alcohol  consumption.  Alcohol  Clin 
Exp  Res  12:81-87, 1988. 

Pfefferbaum,  A.;  Lim,  K.O.;  Zipursky, 
R.B.;  Mathalon,  D.H.;  Lane,  B.;  Ha, 
C.N.;  Rosenbloom,  M.J.;  and  Sullivan, 
E.V.  Brain  gray  and  white  matter  volume 
loss  accelerates  with  aging  in  chronic 
alcoholics:  A  quantitative  MRI  study. 
Alcohol  Clin  Exp  Res  16:1078-1089, 1992. 

Pfefferbaum,  A.;  Sullivan,  E.V.; 
Rosenbloom,  M.J.;  Shear,  P.K.;  Mathalon, 
D.H.;  and  Lim,  K.O.  Increase  in  brain 
cerebrospinal  fluid  volume  is  greater  in 
older  than  in  younger  alcoholic  patients: 
A  replication  study  and  CT/MRI  com- 
parison. Psychiatry  Res  Neuroimaging 
50:257-274,  1993. 

Pfefferbaum,  A.;  Mathalon,  D.H.;  Sullivan, 
E.V.;  Rawles,  J.M.;  Zipursky,  R.B.;  and 
Lim,  K.O.  A  quantitative  magnetic  reso- 
nance imaging  study  of  changes  in  brain 
morphology  from  infancy  to  late  adult- 
hood. Arch  Neurol  51:874-887,  1994. 


Pfefferbaum,  A.;  Sullivan,  E.V.;  Mathalon, 
D.H.;  Shear,  P.K.;  Rosenbloom,  M.J.; 
and  Lim,  K.O.  Longitudinal  changes  in 
magnetic  resonance  imaging  brain  volumes 
in  abstinent  and  relapsed  alcoholics.  Alcohol 
Clin  Exp  Res  19:1177-1191,  1995. 

Pfefferbaum,  A.;  Lim,  K.O.;  Desmond,  J.; 
and  Sullivan,  E.V.  Thinning  of  the  corpus 
callosum  in  older  alcoholic  men:  A  mag- 
netic resonance  imaging  study.  Alcohol 
Clin  Exp  Res  20:752-757,  1996a. 

Pfefferbaum,  A;  Rosenbloom,  M.J.;  O'Neil, 
L.O.;  Jackson,  J.;  Lutz,  A.;  Deshmukh, 
A.;  Sullivan,  E.V.;  and  Lim,  K.O.  An  MRI 
study  of  brain  volume  changes  in  alcoholic 
women.  Alcohol  Clin  Exp  Res  20:33A 
(abstract),  1996&. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Swan, 
G.E.;  and  Carmelli,  D.  Brain  structure 
in  men  remains  highly  heritable  in  the 
seventh  and  eighth  decades  of  life. 
Neurobiology  of  Aging,  in  press. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Mathalon, 
D.H.;  and  Lim,  K.O.  Frontal  lobe  volume 
loss  observed  with  magnetic  resonance 
imaging  in  older  chronic  alcoholics. 
Alcohol  Clin  Exp  ^^21:521-529,  1997. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Rosenbloom, 
M.J.;  Mathalon,  D.H.;  and  Lim,  K.O.  A 
controlled  study  of  cortical  gray  matter 
and  ventricular  changes  in  alcoholic  men 
over  a  five  year  interval.  Arch  Gen 
Psychiatry  55:905-912,  1998. 

Pfefferbaum,  A.;  Sullivan,  E.;  Adalsteinsson, 
A.;  Spielman,  D.;  and  Lim,  K.O.  In 
vivo  spectroscopic  quantification  of  the 
N- acetyl  moiety,  creatine  and  choline 
from  large  volumes  of  gray  and  white 
matter:  Effects  of  normal  aging.  J  Magn 
Reson  Imaging  41:27 '6-284,  1999a;. 

Pfefferbaum,  A.;  Sullivan,  E.;  Adalsteinsson, 
A.;  Spielman,  D.;  and  Lim,  K.O.  Low  N- 


504 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


acetyl  aspartate  and  high  choline  gray 
matter  signal  in  Alzheimer's  disease: 
Relationships  to  cognitive  functions.  Arch 
Gen  Psychiatry  56:185-192,  1999£. 

Pfefferbaum,  A.;  Sullivan,  E.V.;  Hedehus, 
M.;  Moseley,  M.;  and  deCrespigny,  A. 
Disruption  of  brain  white  matter  fiber 
coherence  in  alcoholics  observed  with 
diffusion  tensor  imaging  (abstract). 
Alcohol  Clin  Exp  Res  23:68A,  1999c. 

Rao,  V.L.R,  and  Butterworth,  R.F. 
Thiamine  phosphatases  in  human  brain: 
Regional  alterations  in  patients  with 
alcoholic  cirrhosis.  Alcohol  Clin  Exp  Res 
19:523-526,  1995. 

Raz,  N.;  Gunning,  F.M.;  Head,  D.; 
Dupuis,  J.H.;  McQuain,  J.;  Briggs,  S.D.; 
Loken,  W.J.;  Thornton,  A.E.;  and  Acker, 
J.D.  Selective  aging  of  the  human  cere- 
bral cortex  observed  in  vivo:  Differential 
vulnerability  of  the  prefrontal  gray  matter. 
Cereb  Cortex  7:268-282,  1997. 

Regier,  D.A.;  Farmer,  M.E.;  Rae,  D.S.; 
Locke,  B.Z.;  Keith,  S.J.;  Judd,  L.L.;  and 
Goodwin,  F.K.  Comorbidity  of  mental 
disorders  with  alcohol  and  other  drug 
abuse — results  from  the  Epidemiologic 
Catchment  Area  (ECA)  Study.  JAMA 
264:2511-2518,  1990. 

Riley,  E.P.;  Mattson,  S.N.;  Sowell,  E.R.; 
Jernigan,  T.L.;  Sobel,  D.F.;  and  Jones, 
K.L.  Abnormalities  of  the  corpus  callosum 
in  children  prenatally  exposed  to  alcohol. 
Alcohol  Clin  Exp  Res  19:1198-1202,  1995. 

Riley,  J.N.,  and  Walker,  D.W.  Morphological 
alterations  in  hippocampus  after  long-term 
alcohol  consumption  in  mice.  Science 
201:646-648,  1978. 

Roman,  P.,  and  Blum,  T.  Life  transitions, 
work,  and  alcohol:  An  overview  and  pre- 
liminary data.  Alcohol  Clin  Exp  Res  16: 
149-158,  1992. 


Ron,  M.A.;  Acker,  R.W.;  Shaw,  G.K.;  and 
Lishman,  W.A.  Computerized  tomogra- 
phy of  the  brain  in  chronic  alcholism:  A 
survey  and  follow-up  study.  Brain 
105:497-514,  1982. 

Sachs,  PL;  Russell,  J.A.G.;  Christman, 
D.R;  and  Cook,  B.  Alteration  of  regional 
cerebral  glucose  metabolic  rate  in  non- 
Korsakoff  chronic  alcoholism.  Arch 
Neurol  44:1242-1251,  1987. 

Schlaepfer,  T.E.;  Harris,  G.J.;  Tien,  A.Y.; 
Peng,  L.W.;  Lee,  S.;  Federman,  E.B.; 
Chase,  G.A.;  Barta,  P.E.;  and  Pearlson, 
G.D.  Decreased  regional  cortical  gray 
matter  volume  in  schizophrenia.  Am  J 
Psychiatry  151:842-848,  1994. 

Schleifer,  S.;  Keller,  S.;  LaFarge,  S.; 
Dhaibar,  Y.;  Shiflett,  S.;  and  Eckholdt,  H. 
Human  immunodeficiency  virus-type  1 
infection  in  an  inner- city  alcohol  treat- 
ment program.  Alcohol  Clin  Exp  Res 
20:75-80,  1996. 

Schroth,  V.G.;  Remmes,  U.;  and 
Schupmann,  A.  Brain  shrinking  in  chronic 
alcoholism:  CT  follow-up  study  in  65 
patients.  Eortschritte  Roentgenst  142:363- 
369,  1985. 

Schroth,  G.;  Naegele,  T.;  Klose,  U.; 
Mann,  K;  and  Petersen,  D.  Reversible 
brain  shrinkage  in  abstinent  alcoholics, 
measured  by  MRI.  Neuroradiology 
30:385-389,  1988. 

Seitz,  D.;  Widmann,  U.;  Seeger,  U.; 
Nagele,  T.;  Mann,  K;  and  Grodd,  W. 
Localized  proton  MR  spectroscopy  of  the 
cerebellum  in  detoxifying  alcoholics. 
Alcohol  Clin  Exp  Res  23:158-163,  1999. 

Shear,  P.K.;  Jernigan,  T.L.;  and  Butters, 
N.  Volumetric  magnetic  resonance  imag- 
ing quantification  of  longitudinal  brain 
changes  in  abstinent  alcoholics.  Alcohol 
Clin  Exp  Res  18:172-176,  1994. 


505 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Shear,  P.K.;  Sullivan,  E.V.;  Lane,  B.;  and 
Pfefferbaum,  A.  Mammillary  body  and 
cerebellar  shrinkage  in  chronic  alcoholics 
with  and  without  amnesia.  Alcohol  Clin 
Exp  Res  20:1489-1495,  1996. 

Shimamura,  A.P.;  Jernigan,  T.L.;  and 
Squire,  L.R.  KorsakofPs  syndrome: 
Radiological  (CT)  findings  and  neuropsy- 
chological correlates.  J  Neurosci  8:4400- 
4410,  1988. 

Sowell,  E.R.;  Jernigan,  T.L.;  Mattson, 
S.N.;  Riley,  E.P.;  Sobel,  D.F.;  and  Jones, 
K.L.  Abnormal  development  of  the  cere- 
bellar vermis  in  children  prenatally  exposed 
to  alcohol:  Size  reduction  in  lobules  I-V. 
Alcohol  Clin  Exp  Res  20:31-34,  1996. 

Spielman,  D.M.;  Glover,  G.H.;  Macovski, 
A.;  and  Pfefferbaum,  A.  Magnetic  resonance 
spectroscopic  imaging  of  ethanol  in  the 
human  brain:  A  feasibility  study.  Alcohol 
Clin  Exp  Res  17:1072-1077,  1993. 

Squire,  L.R,  and  Zola,  S.M.  Memory, 
memory  impairment,  and  the  medial 
temporal  lobe.  Cold  Spring  Harb  Symp 
Quant  Biol  61:185-195,  1996. 

Squire,  L.R.;  Amaral,  D.G.;  and  Press, 
G.A.  Magnetic  resonance  imaging  of  the 
hippocampal  formation  and  mammillary 
nuclei  distinguish  medial  temporal  lobe 
and  diencephalic  amnesia.  /  Neurosci 
10:3106-3117,1990. 

Stout,  J.;  Ellis,  R;  Jernigan,  T.;  Archibald, 
S.;  Abramson,  I.;  Wolfson,  T.;  McCutchan, 
J.;  Wallace,  M.;  Atkinson,  J.;  and  Grant, 
I.  Progressive  cerebral  volume  loss  in 
human  immunodeficiency  virus  infection: 
A  longitudinal  volumetric  magnetic  reso- 
nance imaging  study.  Arch  Neurol 
55:161-168,  1998. 

Strittmatter,  W.J.,  and  Roses,  A.D. 
Apolipoprotein  e  and  Alzheimer's  disease. 
Ann  Rev  Neurosci  19:53-77,  1996. 


Sullivan,  E.V.;  Mathalon,  D.H.;  Zipursky, 
R.B.;  Kersteen-Tucker,  Z.;  Knight,  R.T.; 
and  Pfefferbaum,  A.  Factors  of  the 
Wisconsin  Card  Sorting  Test  as  a  test  of 
dorsolateral  prefrontal  cortical  function  in 
schizophrenia  and  alcoholism.  Psychiatry 
Res  46:175-199,  1993. 

Sullivan,  E.V.;  Marsh,  L.;  Mathalon, 
D.H.;  Lim,  K.O.;  and  Pfefferbaum,  A. 
Anterior  hippocampal  volume  deficits  in 
nonamnesic,  aging  chronic  alcoholics. 
Alcohol  Clin  Exp  Res  19:110-122,  1995#. 

Sullivan,  E.V.;  Rosenbloom,  M.;  Deshmukh, 
A.;  Desmond,  J.E.;  and  Pfefferbaum,  A. 
Alcohol  and  the  cerebellum:  Effects  on 
balance,  motor  coordination,  and  cogni- 
tion. Alcohol  Health  Res  World  19:138- 
141,  19956. 

Sullivan  E.V.;  Deshmukh,  A.;  Desmond, 
J.E.;  Mathalon,  D.H.;  Rosenbloom,  M.J.; 
Lim,  K.O.;  and  Pfefferbaum,  A.  Contribution 
of  alcohol  abuse  to  cerebellar  volume 
deficits  in  men  with  schizophrenia. 
Archives  of  General  Psychiatry,  in  press. 

Sullivan,  E.V.;  Deshmukh,  A.;  Desmond, 
J.E.;  Lim,  K.O.;  and  Pfefferbaum,  A. 
Cerebellar  volume  decline  in  normal 
aging,  uncomplicated  alcoholism,  and 
Korsakoff  syndrome:  Relationship  to 
ataxia.  Neuropsychol,  in  press. 

Sullivan,  E.V.;  Marsh,  L.;  Mathalon, 
D.H.;  Lim,  K.O.;  and  Pfefferbaum,  A. 
Relationship  between  alcohol  withdrawal 
seizures  and  temporal  lobe  white  matter 
volume  deficits.  Alcohol  Clin  Exp  Res 
20:348-354,  1996^. 

Sullivan,  E.V.;  Marsh,  L.;  Shear,  P.K.;  Lim, 
K.O.;  and  Pfefferbaum,  A.  Hippocampal 
but  not  cortical  volumes  distinguish  amnesic 
and  nonamnesic  alcoholics  (abstract). 
Abstracts:  Society  for  Neuroscience 
22:1865.  Washington  DC,  November 
16-21,  19966. 


506 


Brain  Vulnerability  to  Alcoholism:  Neuroimaging  Studies 


Sullivan,  E.V.;  Deshmukh,  A.;  Desmond, 
J.E.;  Shear,  P.K.;  Lim,  K.O.;  Pfefferbaum, 
A.  Cerebellar  volume  deficits  and  neuropsy- 
chological function  in  alcoholics  (abstract). 
Alcohol  Clin  Exp  Res  22:63 A,  1998#. 

Sullivan,  E.;  Mathalon,  D.H.;  Lim,  K.O.; 
Marsh,  L.;  and  Pfefferbaum,  A.  Patterns 
of  regional  cortical  dysmorphology  distin- 
guishing schizophrenia  and  chronic  alco- 
holism. Biol  Psychiatry  43:118-131,  1998/?. 

Sullivan,  E.V.;  Lane,  B.;  Rosenbloom, 
M.J.;  Desmukh,  A.;  Desmond,  J.;  Lim, 
K.O.;  and  Pfefferbaum,  A.  In  vivo  mam- 
millary  body  volume  deficits  in  amnesic 
and  nonamnesic  alcoholics.  Alcohol  Clin 
Exp  Res  23(10):  1629-1636,  1999. 

Sullivan,  E.V.;  Rosenbloom,  M.J.;  Lim, 
K.O.;  and  Pfefferbaum,  A.  Longitudinal 
changes  in  cognition,  gait  and  balance  in 
abstinent  and  relapsed  alcoholic  men: 
Relationships  to  changes  in  brain  structure. 
Neuropsychology,  in  press. 

Sullivan,  W.  A  note  on  the  influence  of 
maternal  inebriety  on  the  offspring.  Journal 
of  the  Mental  Sciences  45:489-503,  1899. 

Tanabe,  J.L.;  Amend,  D.;  Schuff,  N.;  Di 
Sclafani,  V.;  Ezekiel,  F.;  Norman,  D.;  Fein, 
G.;  and  Weiner,  M.W.  Tissue  segmentation 
of  the  brain  in  Alzheimer  disease.  Am  J 
Neuroradiol  18:115-123,  1997. 

Tedeschi,  G.;  Bertolino,  A.;  Righini,  A.; 
Campbell,  G.;  Raman,  R.;  Duyn,  J.H.; 
Moonen,  C.T.W.;  Alger,  J.R.;  and  Di 
Chiro,  G.  Brain  regional  distribution 
pattern  of  metabolite  signal  intensities  in 
young  adults  by  proton  magnetic  reso- 
nance spectroscopic  imaging.  Neurology 
45:1384-1391,  1995. 

Thomasson,  H.S.  Gender  differences  in 
alcohol  metabolism:  Physiological  responses 
to  ethanol.  In:  Galanter,  M.,  eds.  Recent 
Developments  in  Alcoholism,  Vol  12: 


Alcoholism  and  Women.  New  York: 
Plenum  Press,  1995.  pp.  163-180. 

Torvik,  A.  Wernicke's  encephalopathy: 
Prevalence  and  clinical  spectrum.  Alcohol 
Alcohol  Suppl  1:381-384,  1991. 

Urenjak,  J.;  Williams,  S.R.;  Gadian,  D.G.; 
and  Noble,  M.  Specific  expression  of  N- 
acetylaspartate  in  neurons,  oligodendro- 
cyte-type-2  astrocyte  progenitors,  and 
immature  oligodendrocytes  in  vitro.  / 
Neurochem  59:55-61,  1992. 

Urenjak,  J.;  Williams,  S.R.;  Gadian,  D.G.; 
and  Noble,  M.  Proton  nuclear  magnetic 
resonance  spectroscopy  unambiguously 
identifies  different  neural  cell  types.  / 
Neurosci  13:981-989,  1993. 

Victor,  M.,  and  Brausch,  C.  The  role  of 
abstinence  in  the  genesis  of  alcoholic 
epilepsy.  Epilepsia  8:1-20,  1967. 

Victor,  M.;  Adams,  R.D.;  and  Collins, 
G.H.  The  Wernicke-Korsakoff  Syndrome. 
Philadelphia:  FA.  Davis,  1971. 

Victor,  M.;  Adams,  R.D.;  and  Collins, 
G.H.  The  Wernicke-Korsakoff  Syndrome 
and  Related  Neurologic  Disorders  Due  to 
Alcoholism  and  Malnutrition.  2d  ed. 
Philadelphia:  FA.  Davis,  1989. 

Volkow,  N.D.;  Hitzemann,  R;  Wang, 
G.J.;  Fowler,  J.S.;  Burr,  G.;  Pascani,  K.; 
Dewey,  S.L.;  and  Wolf,  A.P.  Decreased 
brain  metabolism  in  neurologically  intact 
healthy  alcoholics.  Am  J  Psychiatry 
149:1016-1022,  1992. 

Volkow,  N.D.;  Wang,  G.J.;  Hitzemann, 
R;  Fowler,  J.S.;  Overall,  J.E.;  Burr,  G.; 
and  Wolf,  A.P.  Recovery  of  brain  glucose 
metabolism  in  detoxified  alcoholics.  Am  J 
Psychiatry  151:178-183,  1994. 

Volkow,  N.D.;  Wang,  G.J.;  Begleiter,  H.; 
Hitzemann,  R.;  Pappas,  N.;  Burr,  G.; 
Pascani,  K.;  Wong,  C;  Fowler,  J.S.;  and 


507 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Wolf,  A. P.  Regional  brain  metabolic 
response  to  lorazepam  in  subjects  at  risk 
for  alcoholism.  Alcohol  Clin  Exp  Res 
19:510-516,  1995. 

Walker,  D.W.;  Barnes,  D.E.;  Zornfetzer, 
S.F.;  Hunter,  B.E.;  and  Kubanis,  P. 
Neuronal  loss  in  hippocampus  induced  by 
prolonged  ethanol  consumption  in  rats. 
Science  209:711-713,  1980. 

Wang,  G.J.;  Volkow,  N.D.;  Roque,  C.T.; 
Cestaro,  V.L.;  Hitzemann,  R.J.;  Cantos, 
E.L.;  Levy,  A.V.,  and  Dhawan,  A.P.  Func- 
tional importance  of  ventricular  enlargement 
and  cortical  atrophy  in  healthy  subjects 
and  alcoholics  as  assessed  with  PET,  MR 
imaging,  and  neuropsychologic  testing. 
Radiology  186:59-65,  1993. 

Wehrli,  R.W.;  MacFall,  J.R;  Shutts,  D.; 
Breger,  R.;  and  Herfkens,  RJ.  Mechanisms 
of  contrast  in  NMR  imaging.  /  Comput 
Assist  Tomogr  8:369-380,  1984. 

West,  J.R.;  Goodlett,  C.R.;  Bonthius, 
D.J.;  Hamre,  K.M.;  and  Marcussen,  B.L. 
Cell  population  depletion  associated  with 
fetal  alcohol  brain  damage:  Mechanisms 
of  BAC-dependent  cell  loss.  Alcohol  Clin 
Exp  Res  14:813-818,  1990. 

Wik,  G.;  Borg,  S.;  Sjogren,  I.;  Wiesel, 
FA.;  Blomqvist,  G.;  Borg,  J.;  Greitz,  T.; 
Nyback,  PL;  Sedvall,  G.;  Stone- Elan der, 
S.;  and  Widens,  L.  PET  determination  of 
regional  cerebral  glucose  metabolism  in 
alcohol-dependent  men  and  healthy  con- 
trols using  uC-glucose.  Acta  Psychiatr 
Scand  78:234-241,  1988. 

Wilkinson,  DA.  Neuroradiologic  investi- 
gations of  alcoholism.  In:  Tarter,  R.E., 
and  Van  Thiel,  D.H.,  eds.  Alcohol  and  the 
Brain:  Chronic  Effects.  New  York:  Plenum, 
1985.  pp.  183-215. 

Wilkinson,  DA.,  and  Carlen,  P.L.  Rela- 
tionship of  neuropsychological  test  perfor- 


mance to  brain  morphology  in  amnesic  and 
non-amnesic  chronic  alcoholics.  Acta  Psychiatr 
Scand  62  (Suppl  286):89-101,  1980. 

Wilsnack,  R.W.;  Wilsnack,  S.C.;  and 
Klassen,  A.D.  Women's  drinking  and 
drinking  problems:  Patterns  from  a  1981 
national  survey.  Am  J  Pub  Health 
74:1231-1238,  1984. 

Witt,  E.,  and  Goldman -Rakic,  P. 
Intermittent  thiamine  deficiency  in  the 
rhesus  monkey.  I.  Progression  of  neuro- 
logical signs  and  neuroanatomical  lesions. 
Ann  Neurol  13:376-395,  1983. 

York,  J.L.,  and  Welte,  J  W.  Gender  com- 
parisons of  alcohol  consumption  in  alco- 
holic and  nonalcoholic  populations.  / 
Stud  Alcohol  55:743-750,  1994. 

Zimmerberg,  B.,  and  Mickus,  L.A.  Sex 
differences  in  corpus  callosum — influence 
of  prenatal  alcohol  exposure  and  maternal 
undernutrition.  Brain  Res  537:115- 
122,  1990. 

Zimmerberg,  B.,  and  Scalzi,  L.V. 
Commissural  size  in  neonatal  rats:  Effects 
of  sex  and  prenatal  alcohol  exposure.  IntJ 
Neurosci  7 ':81-86,  1989. 

Zipursky,  RB.;  Lim,  K.O.;  and  Pfefierbaum, 
A.  MRI  study  of  brain  changes  with  short 
term  abstinence  from  alcohol.  Alcohol 
Clin  Exp  Res  13:664-666,  1989. 

Zipursky,  R.B.;  Lim,  K.O.;  Sullivan,  E.V.; 
Brown,  B.W.;  and  Pfefferbaum,  A. 
Widespread  cerebral  gray  matter  volume 
deficits  in  schizophrenia.  Arch  Gen 
Psychiatry  49:195-205,  1992. 

Zipursky,  R.B.;  Seeman,  M.V.;  Bury,  A.; 
Langevin,  R.;  Wortzman,  G.;  and  Katz, 
R.  Deficits  in  gray  matter  volume  are 
present  in  schizophrenia  but  not  bipolar 
disorder.  Schizophr  Res  26:85-92,  1997. 


508 


Chapter  15 

Human  Brain  Dysfunction  Secondary 

to  Alcohol  Abuse:  Suggestions  for 

New  Research  Initiatives 


George  Fein,  Ph.D.,  Daniel  Fletcher,  Ph.D., 
and  Victoria  Di  Sclafani,  M.P.H. 

KEY  WORDS:  AOD  (alcohol  or  other  drug)  abuse;  brain  damage;  electroen- 
cephalography; neuroimaging;  behavior;  chronic  AODE  (effects  of  AOD  use, 
abuse,  and  dependence);  risk  factors;  dysfunction;  gender  differences;  aging;  ado- 
lescent; morbidity;  evoked  potential;  literature  review 


Electrophysiological ,  neuropsycholog- 
ical,  and  structural  and  functional 
neuroimaging  methods  hold  great 
promise  for  obtaining  new  insights 
into  brain  dysfunction  secondary  to 
chronic  alcohol  abuse.  These  comple- 
mentary approaches  have  the  potential 
to  advance  the  currently  limited  state 
of  knowledge  regarding  ( 1 )  structural 
damage  to  the  brain  pursuant  to 
chronic  alcohol  exposure,  (2)  func- 
tional brain  changes  associated  with 
or  independent  of  structural  damage 
(or  both),  and  (3)  the  specific  behav- 
ioral effects  resulting  from  these  struc- 
tural and  functional  alterations. 


WHAT  ARE  THE  MOST 
IMPORTANT  QUESTIONS 
REGARDING  BRAIN 
DYSFUNCTION 
RELATED  TO  CHRONIC 
ALCOHOL  ABUSE? 

Which  Brain  Systems  Are 
Most  Vulnerable  to  the 
Morbid  Effects  of  Chronic 
Alcohol  Abuse? 

It  is  important  to  improve  the  evi- 
dence linking  structural  damage, 
functional  alterations,  and  the  specific 
behavioral  effects  of  chronic  alcohol 


G.  Fein,  Ph.D.,  is  a  senior  scientist  at  Neurobehavioral  Research  Inc.,  45  Gable  Court,  San  Rafael, 
CA  94903.  D.  Fletcher,  Ph.D.,  is  a  student  in  the  School  of  Veterinary  Medicine  at  the  University  of 
California,  Davis.  V.  Di  Sclafani,  M.P.H.,  is  a  research  scientist  at  Neurobehavioral  Research  Inc. 


509 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


abuse.  Performance  on  most  behavioral 
tasks  involves  a  complex  interplay  of  a 
number  of  brain  systems.  Further  work 
is  needed  to  define  the  specific  brain 
processes  and  systems  affected  by 
chronic  alcohol  abuse.  Such  work  could 
begin  with  a  review  of  the  behavioral 
literature  to  determine  tasks  that  con- 
sistently reveal  robust  performance 
decrements  in  long-term  alcoholics. 
This  should  be  followed  up  with  care- 
ful behavioral,  electrophysiological,  and 
functional  imaging  investigations  of 
performance  on  such  tasks  in  non- 
alcohol-abusing  subjects  to  elucidate 
the  constituent  operations  and  brain 
systems  involved  in  performing  these 
tasks.  This  approach  would  provide  a 
normative  database  useful  in  defining 
abnormalities  in  alcoholic  patients. 

It  is  very  possible  that  alcohol- 
related  changes  will  be  subtle,  reflect- 
ing "bias"  effects  within  the  neural 
systems  that  contribute  to  task 
performance.  In  other  words,  the  sys- 
tems and  associated  sites  of  brain 
activation  may  be  unchanged  in 
chronic  alcoholic  patients,  but  the 
relative  contribution  and  timing 
of  activation  of  the  constituent 
brain  systems  may  be  altered. 
Electrophysiological  methods  can 
make  a  unique  contribution  to  such 
investigations  as  a  result  of  their  very 
fine  temporal  resolution. 

What  Alcohol  Use-Related 
Factors  Influence  Brain 
Morbidity  Secondary  to 
Alcohol  Abuse? 

1 .  What  is  the  relative  role  of  nutritional 
(primarily  thiamine)  deficiency  and 


impaired  liver  function  in  the 
development  of  brain  dysfunction 
secondary  to  alcohol  abuse?  Is 
there  a  diagnostic  entity  of 
alcoholic  dementia  separate  from 
that  of  dementia  secondary 
to  alcohol  abuse-related  nutri- 
tional deficiency  or  impaired 
liver  function  (Diamond  and 
Messing  1994;  Emsley  et  al. 
1996)?  How  would  one  differenti- 
ate these  conditions  from  each 
other  and  from  brain  changes  asso- 
ciated with  coexisting  Alzheimer's 
disease  (AD)  or  primary  cere- 
brovascular disease? 

2.  Is  brain  dysfunction  primarily  related 
to  the  effects  of  alcohol  consump- 
tion or  to  the  occurrence  of  episodes 
of  withdrawal?  A  fundamental 
question  is  whether  brain  dys- 
function is  caused  by  the  direct 
toxic  effects  of  heavy  alcohol  use  or 
whether  it  is  caused  by  the  episodes 
of  alcohol  withdrawal  (which  result 
in  kindling  in  animals  and 
humans).  This  is  a  core  issue  with 
very  little  data. 

3.  Is  the  development  of  tolerance  to 
the  acute  effects  of  alcohol  associated 
(either  positively  or  negatively) 
with  the  degree  of  brain  dysfunc- 
tion secondary  to  alcohol  abuse? 

4.  What  is  the  role  of  the  pattern  of 
drinking  behavior  on  the  develop- 
ment of  brain  dysfunction?  This 
issue  relates  to  the  relative  impor- 
tance of  total  alcohol  consumption 
versus  the  maximum  alcohol  dose 
(i.e.,  the  brain  dysfunction  atten- 


510 


Brain  Dysfunction  Secondary  to  Alcohol  Abuse 


dant  to  chronic  regular  drinking 
versus  binge  drinking). 

What  Other  Factors 
Influence  Brain  Morbidity 
Secondary  to  Alcohol  Abuse? 

Other  factors  that  may  influence  brain 
morbidity  secondary  to  alcohol  abuse 
include  genetics,  gender,  age,  brain 
functional  reserve,  and  comorbidity. 
These  factors  are  discussed  in  the  fol- 
lowing paragraphs. 

Are  there  genetically  transmitted 
vulnerabilities  to  alcohol  abuse-related 
brain  morbidity?  There  is  extensive 
compelling  evidence  for  a  genetic  pre- 
disposition to  develop  alcoholism  (e.g., 
Pfefferbaum  et  al.  1991;  Cadoret  et 
al.  1995;  Crabbe  et  al.  1998).  It  is  pos- 
sible that  there  may  also  be  a  genetic 
predisposition  to  develop  brain  damage 
secondary  to  the  toxic  effects  of  chronic 
alcohol  abuse.  Moreover,  such  a 
genetic  predisposition  may  be  quite 
independent  of  the  genetic  predisposi- 
tion to  develop  alcoholism. 

How  does  gender  affect  the  severity 
and  persistence  of  brain  morbidity  sec- 
ondary to  alcohol  abuse?  Most  research 
on  cognitive  function  in  recovering 
alcoholics  has  used  samples  of  male 
veterans  in  inpatient  and  outpatient 
treatment  programs.  The  relative  vulner- 
ability of  women  to  cognitive  deficits 
associated  with  chronic  alcohol  abuse 
has  received  only  limited  study. 
Although  most  studies  report  the 
same  pattern  and  degree  of  deficits  in 
alcoholic  women  as  in  alcoholic  men 
(Sparadeo  et  al.  1993),  gender-related 
differences  have  been  reported  (Leber 
et  al.  1981).  Some  of  these  studies  have 
determined  that  the  pattern  of  deficits 


is  the  same  in  female  alcoholics  as  in 
male  alcoholics,  but  is  more  severe 
among  women  (Acker  1986;  Bergman 
1987),  or  that  deficits  comparable  to 
those  suffered  by  male  alcoholics  are 
present  in  women  after  shorter  drink- 
ing histories  and  lower  average  daily 
consumptions  (Acker  1986).  Some 
studies  have  found  additional  deficits 
in  female  alcoholics,  such  as  in  verbal 
abstraction  ability  (Hatcher  et  al. 
1977).  Other  studies  that  compare  cog- 
nitive functioning  of  alcoholic  women 
and  men  show  better  performance 
among  the  women  in  visuospatial  abil- 
ities (Fabian  et  al.  1984)  and  in  both 
verbal  and  visual  short-term  memory 
(Sparadeo  et  al.  1993).  It  is  clear  that 
additional  research  on  the  central  ner- 
vous system  (CNS)  effects  of  chronic 
alcohol  abuse  in  women  is  needed;  a 
particularly  important  question  is 
whether  the  modulating  effects  of  age 
on  the  CNS  morbidity  of  chronic  alco- 
hol abuse  differ  in  men  and  women. 

Why  is  brain  morbidity  greater  in 
the  older  alcoholic?  The  effect  of  aging 
on  brain  dysfunction  secondary  to 
alcohol  abuse  has  been  a  long-debated 
topic.  There  is  an  extensive  literature 
demonstrating  that  brain  morbidity 
due  to  alcohol  abuse  is  greater  in  the 
older  compared  with  the  young  or 
middle-aged  alcoholic,  even  after 
adjustments  are  made  for  duration 
and  amount  of  alcohol  consumption 
(e.g.,  Goldman  et  al.  1983;  Grant  et  al. 
1984;  Fein  et  al.  1990;  Pfefferbaum  et 
al.  1992).  The  generally  accepted 
"age-related  vulnerability"  model  postu- 
lates that  abusive  drinking  during  old 
age  has  greater  effects  on  the  brain 
than  does  comparable  drinking  during 


511 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


young  adulthood  or  middle  age.  In 
this  model,  an  older  alcoholic  who 
became  abstinent  in  old  age  would  have 
more  brain  damage  than  an  alcoholic 
of  the  same  age  who  became  abstinent 
in  middle  age,  assuming  comparable 
duration  and  amount  of  drinking. 

An  alternative  model,  the  "cumula- 
tive effects"  model,  postulates  the  aging 
process  and  the  duration  and  amount 
of  abusive  drinking  as  additive  effects 
underlying  the  greater  brain  morbidity 
in  the  older  alcoholic.  In  this  model, 
brain  structure  and  function  of  two 
older  abstinent  alcoholics  would  be 
comparable  regardless  of  the  age  at 
which  abstinence  was  attained  (assum- 
ing comparable  durations  and  total 
amounts  of  alcohol  consumed).  This 
cumulative  effects  model  maintains 
that  the  young  or  middle-aged  brain 
is  able  to  compensate  for  the  damage 
done  by  alcohol  abuse.  However, 
even  in  such  individuals,  the  brain 
damage  caused  by  abusive  drinking  in 
earlier  life  will  become  unmasked  as 
compensatory  brain  processes  are  dimin- 
ished due  to  the  normal  aging  process. 
There  are  no  data  in  the  literature  to 
test  the  differential  predictions  of  these 
two  models  (e.g.,  data  on  the  brain 
status  of  older  individuals  who  were 
alcoholic  into  middle  age,  but  who  have 
been  abstinent  for  10  years  or  more) 
because  research  to  date  has  focused 
only  on  recently  abstinent  individuals 
(usually  those  in  treatment  through 
the  first  year  or  so  of  abstinence). 

The  cumulative  effects  model  and  the 
age-related  vulnerability  model  have 
very  different  implications.  The  age- 
related  vulnerability  model  suggests 
that  the  long-term  consequences  of 


alcohol  abuse  are  attenuated  if  a  person 
stops  abusing  alcohol  before  old  age 
(when  the  brain  is  much  more  vulner- 
able to  lasting  damage  secondary  to 
alcohol  abuse).  The  cumulative  effects 
model  suggests  that  functional  dam- 
age will  be  revealed  as  a  person  ages 
(when  the  brain  can  no  longer  com- 
pensate for  the  underlying  damage) 
no  matter  when  he  or  she  stopped 
abusing  alcohol. 

Are  there  subject  factors  that  denote 
a  brain  functional  reserve  mitigating 
the  effects  of  chronic  alcohol  abuse7. 
Might  genetic  factors  and/ or  parental 
alcohol  abuse  result  in  a  decreased  func- 
tional reserve,  amplifying  the  morbid 
effects  of  chronic  alcohol  use7.  The  concept 
of  brain  functional  reserve  was  devel- 
oped in  the  AD  literature  to  describe 
normally  "unused"  or  "extra"  brain 
capacity.  In  the  AD  literature,  functional 
reserve  has  been  estimated  using  a  mea- 
sure of  premorbid  brain  volume,  pos- 
tulating a  greater  number  of  neurons 
and  synapses  available  in  the  larger 
brain.  Head  circumference  or  intracra- 
nial volume  quantified  on  magnetic 
resonance  images  or  computed 
tomography  (CT)  images  is  usually 
used  to  estimate  premorbid  brain  vol- 
ume (since  mature  skull  size  is  driven 
by  brain  growth,  a  process  mostly 
complete  by  the  end  of  the  second 
year  of  life).  AD  patients  with  larger 
head  circumference  or  intracranial  vol- 
ume on  magnetic  resonance  or  CT 
images  had  a  later  onset  of  disease,  or 
a  lessened  severity  of  cognitive  impair- 
ment, or  both  (Schofield  et  al.  1995; 
Graves  et  al.  1996;  Mori  et  al.  1997). 

In  the  first  study  (that  we  know  of) 
to  investigate  the  concept  of  functional 


512 


Brain  Dysfunction  Secondary  to  Alcohol  Abuse 


reserve  outside  the  AD  field,  Di  Sclafani 
and  colleagues  (1998),  in  our  laboratory, 
used  MRI  to  quantitate  the  intracranial 
volume  of  cocaine -dependent  individ- 
uals and  dually  addicted  cocaine-  and 
alcohol-dependent  persons  abstinent 
approximately  3  months.  Premorbid 
brain  size  accounted  for  more  than  20 
percent  of  the  variance  in  global  cog- 
nitive impairment  in  both  groups. 
The  investigators  suggested  that  func- 
tional reserve  may  be  a  general  pro- 
tective mechanism  of  the  brain  that 
has  consequences  for  the  severity  of 
expression  of  cerebral  disease  or  insult 
throughout  life. 

In  addition  to  the  very  large  effect 
of  functional  reserve  on  neuropsycho- 
logical performance  in  abstinent  alcohol- 
and/or  other  drug- dependent  (AOD- 
dependent)  subjects,  Di  Sclafani  and 
colleagues  (1998)  also  observed  a  trend 
for  AOD-abusing  subjects  to  have  a 
smaller  intracranial  volume  than  control 
subjects.  It  is  possible  that  the  inter- 
section of  negative  prenatal  and  early 
childhood  factors  on  brain  growth  and 
the  genetic  predisposition  to  alcoholism 
and  addiction  may  result  in  a  synergism. 
The  parents  of  AOD-dependent  sub- 
jects may  have  been  AOD-dependent 
themselves;  therefore  both  prenatal  and 
early  childhood  environments  for  such 
individuals  may  not  be  conducive  to 
maximal  brain  growth.  Furthermore, 
children  of  AOD-dependent  parents 
have  a  greater  probability  of  becom- 
ing AOD-dependent  themselves.  In 
that  case,  in  the  context  of  possible 
early  developmental  deficits,  the  mor- 
bid effects  of  AOD  dependence  may 
be  a  "second  strike"  situation  for  the 
brain.  Moreover,  such  individuals  may 


be  even  more  vulnerable  to  a  "third 
strike"  later  in  life  secondary  to  cerebral 
diseases  such  as  AD,  or  even  the  declines 
associated  with  normal  aging. 

Are  there  developing  brain  systems  in 
adolescents  that  are  particularly  vul- 
nerable to  the  morbid  effects  of  chronic 
alcohol  abuse?  There  are  a  plethora  of 
studies  underscoring  the  importance 
of  late  brain  maturational  processes 
during  adolescence  (Benes  et  al. 
1994;  Pfefferbaum  et  al.  1994;  Giedd 
et  al.  1996a,  1996£,  1996c)-  Since 
many  people  begin  to  drink  heavily 
during  adolescence,  it  is  important  to 
determine  if  and  how  alcohol  exposure 
affects  brain  maturation  during  this 
period.  Neuropsychological,  electro- 
physiological, and  in  vivo  imaging 
techniques  can  be  used  to  monitor  the 
processes  of  brain  maturation  in  adoles- 
cence. In  this  regard,  it  has  been  demon- 
strated that  both  age-related  structural 
changes  and  changing  patterns  of 
brain  activation  can  be  observed  during 
late  childhood  and  adolescence.  How- 
ever, the  most  informative  studies  would 
require  analysis  of  changing  patterns 
of  brain  activation  associated  with  per- 
formance of  specific  tasks,  so  that 
alcohol-related  alterations  in  the  devel- 
opmental trajectories  could  be  investi- 
gated in  young  alcohol  abusers,  and 
so  that  such  changes  could  be  linked 
to  specific  behavioral  effects  and  out- 
comes. Work  by  Rakic  and  others 
(Huttenlocher  1979;  Rakic  et  al. 
1986;  Huttenlocher  and  de  Courten 
1987;  Zecevic  and  Rakic  1991)  sug- 
gests that  peripubertal  changes  in 
synaptic  density  are  dramatic,  and 
these  changes  probably  have  important 
and  long-lasting  functional  implications. 


513 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


There  is  also  evidence  that  gonadal 
steroid  hormones  may  play  a  large  role 
in  these  final  "sculpting"  processes 
within  the  brain  (e.g.,  Forget  and 
Cohen  1994;  Panzica  et  al.  1996; 
Rubinow  and  Schmidt  1996).  Alcohol 
exposure,  either  directly  or  through 
interactions  with  hormonal  effects, 
may  result  in  particularly  resilient 
alterations  within  neural  circuits. 
Carefully  designed  electrophysiological 
and  functional  imaging  studies  of  these 
phenomena  would  be  invaluable. 

How  does  chronic  alcohol  abuse-asso- 
ciated brain  dysfunction  manifest  in 
persons  with  comorbid  disorders  who 
have  independent  brain  morbidities? 
This  is  a  crucially  important  issue  given 
the  high  comorbidity  of  alcoholism 
with  psychiatric  disorders  and  with  abuse 
of  other  substances.  The  effects  of 
alcoholism  on  brain  dysfunction  in  per- 
sons with  comorbidity  reflect  at  least  two 
processes.  First,  there  are  the  additive 
(and  possibly  synergistic)  direct  effects 
of  alcohol  abuse  and  the  comorbid 
disorder  (e.g.,  schizophrenia,  HIV 
infection,  cocaine  or  other  drug  abuse) 
on  brain  function.  Second,  alcoholism 
is  associated  with  denial,  reduced  care 
seeking,  and  reduced  compliance  with 
treatment.  This  would  result  in  less 
efficacious  treatment  of  the  comorbid 
disorder,  with  corresponding  greater 
morbidity  secondary  to  the  comorbid 
disorder.  Most  research  studies  focus- 
ing on  these  comorbid  disorders 
exclude  alcohol  abusers.  The  National 
Institute  on  Alcohol  Abuse  and  Alco- 
holism (NIAAA)  should  form  alliances 
with  the  institutes  that  have  direct 
responsibility  for  research  on  these 
comorbid  disorders  to  expand  their 


research  to  include  subjects  who 
chronically  abuse  alcohol. 

What  Are  the  Mechanisms 
Involved  in  Brain  Recovery 
From  the  Effects  of  Chronic 
Alcohol  Abuse? 

Studies  of  the  process  of  brain  recov- 
ery from  dysfunction  associated  with 
chronic  alcohol  abuse  may  need  to 
wait  for  progress  in  the  areas  mentioned 
above.  The  mechanisms  of  recovery 
from  brain  dysfunction  secondary  to 
chronic  alcohol  abuse  may  also  differ 
across  brain  systems  and  with  comor- 
bid and  modulating  factors. 

HOW  CAN 

ELECTROPHYSIOLOGICAL 
METHODS  UNIQUELY 
CONTRIBUTE  TO 
INVESTIGATIONS  OF 
ALCOHOL  ABUSE- 
RELATED  BRAIN 
DYSFUNCTION? 

Electrophysiological  methods  have 
advantages  over  other  neuroimaging 
techniques  in  terms  of  temporal  resolu- 
tion, ease,  and  cost-effectiveness  of  data 
acquisition,  and  the  ability  to  collect 
data  during  the  performance  of  complex 
cognitive  tasks.  The  ability  to  collect 
electrophysiological  data  during  com- 
plex cognitive  tasks  is  particularly  useful 
in  studies  of  the  effects  of  chronic  and 
acute  alcohol  exposure.  Correlating  the 
electrophysiological  responses  of  the 
brain  with  performance  on  specific 
cognitive  tasks  can  yield  insight  into 
alcohol  effects  unobtainable  with 
other  functional  imaging  methods  where 
the  type  of  stimuli  and  tasks  are  more 


514 


Brain  Dysfunction  Secondary  to  Alcohol  Abuse 


severely  constrained  by  the  imaging 
equipment  and  procedures.  In  addition, 
the  injured  brain  is  more  likely  to  dis- 
play measurable  deficiencies  when 
stimulated  rapidly,  and  the  increased 
temporal  resolution  possible  in  elec- 
trophysiological studies  allows  studies 
using  such  paradigms.  Finally,  electro- 
physiological methods  have  particular 
promise  in  studies  of  the  genetic  factors 
in  alcoholism  (and  other  disorders) 
because  of  the  high  heritability  of  event- 
related  potentials  (ERPs)  and  electroen- 
cephalographic  (EEG)  characteristics 
(e.g.,  van  Beijsterveldt  and  Boomsma 
1994;  Sorbel  et  al.  1996;  van  Beijster- 
veldt et  al.  1996).  Positron  emission 
tomography  (PET)  yields  functional 
information  on  a  scale  measured  in 
seconds  to  tens  of  seconds  depending  on 
the  tracer  used,  and  functional  magnetic 
resonance  imaging  (fMRI)  resolves 
brain  activity  on  a  scale  measured  in 
hundreds  of  milliseconds  to  seconds.  In 
contrast,  electrophysiological  recordings 
can  resolve  activity  on  a  millisecond  time 
scale.  Once  the  electrodes  are  attached, 
electrophysiological  data  acquisition 
can  easily  be  done  while  a  subject  per- 
forms tasks  or  responds  to  sensory  stim- 
uli in  any  modality.  Sensory  inputs 
and  information-processing  demands 
can  be  manipulated  and  the  effects  of 
these  manipulations  observed  in  the 
neural  responses  obtained. 

There  are  two  major  categories  of 
brain  electrophysiological  data:  spon- 
taneous EEG  signals  and  ERPs.  ERPs 
reflect  processes  time-locked  to  sen- 
sory stimuli  or  to  specific  information- 
processing  operations  (e.g.,  orienting 
to  a  stimulus  or  preparing  for  a  motor 
response).  They  are  deterministic  and 


repeatable,  and  are  usually  averaged 
over  several  occasions  to  increase  the 
signal-to-noise  ratio.  This  reduces  the 
effect  of  spontaneous  brain  activity, 
which  is  nondeterministic  and  repre- 
sents the  "background"  activity  of  the 
brain.  Because  spontaneous  EEG  sig- 
nals are  nondeterministic  and  ERPs 
are  deterministic  signals,  they  must  be 
interpreted  using  different  analytical 
and  statistical  techniques. 

Spontaneous  EEG  recordings  can 
be  used  to  examine  alertness,  patterns 
of  regional  brain  activation,  and  the 
integrity  and  efficiency  of  connectivity 
between  brain  regions  as  these  measures 
are  affected  by  experimental  manipu- 
lations, disease  states,  or  acute  or 
chronic  AOD  administration.  Similarly, 
ERPs  can  be  used  to  study  brain  sensory 
or  cognitive  processes  and  systems  as 
they  are  affected  by  experimental  manip- 
ulations, disease  states,  or  acute  or 
chronic  AOD  administration. 

Although  electrophysiological  meth- 
ods offer  insights  into  alcohol  abuse- 
associated  brain  dysfunction  not  obtain- 
able with  other  techniques,  their  use- 
fulness can  be  improved  through  further 
technological  developments.  Most 
studies  to  date  have  relied  on  charac- 
terization of  the  ERPs  at  a  single  elec- 
trode location.  However,  this  single - 
channel  waveform  analysis  suffers  from 
a  number  of  limitations.  Because  of 
individual  variation  in  head  shape  and 
brain  structure,  the  same  neural 
source  does  not  yield  a  maximal  ERP 
at  the  same  scalp  location  in  all 
subjects.  Measures  of  the  amplitude 
and  latency  of  the  component  at  a 
single  scalp  location  vary  across  sub- 
jects in  how  well  they  represent  the 


515 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


amplitude  and  latency  of  the  underly- 
ing neural  source. 

The  difficulties  presented  by  individ- 
ual variation  in  head  shape  and  brain 
structure  are  compounded  by  the  fact 
that  scalp  responses  are  rarely  the  result 
of  activation  of  a  single  region  of  the 
brain.  Usually,  ERPs  reflect  the  activity 
of  many  spatially  separated  neural 
sources,  which  are  activated  asynchro- 
nously and  simultaneously.  Indeed, 
even  a  single  peak  in  an  ERP  can  be 
the  result  of  several  distinct  neural 
generators.  This  is  hypothesized  to  be 
the  case  with  the  P300  response, 
which  appears  to  have  at  least  two 
components,  P3A  and  P3B.  This  situ- 
ation makes  interpretation  of  group 
differences  in  ERPs  extremely  com- 
plex, because  a  change  in  a  given  peak 
may  reflect  a  change  in  a  single  neural 
generator  or  in  multiple  generators. 
Several  methods  have  been  developed 
that  attempt  to  individually  character- 
ize the  contributions  of  specific  neural 
generators  and  to  yield  measures  that 
can  be  compared  across  subjects.  Fur- 
ther development  of  these  methods 
would  strengthen  the  usefulness  of 
ERP  measures  in  studies  of  alcohol 
abuse-related  brain  dysfunction. 

EEG  coherence  and  phase  are  mea- 
sures of  the  integrity  and  speed  of 
communication  between  brain  regions 
that  can  be  used  to  investigate  alcohol 
effects  on  white  matter.  A  major 
methodological  problem  has  cast  the 
results  of  much  of  the  previous  coher- 
ence literature  into  doubt.  We  have 
shown  (Fein  et  al.  1988)  that  common- 
reference  EEG  recordings  (which 
make  up  over  90  percent  of  the  data  in 
all  published  studies  to  date)  can  yield 


inaccurate  coherence  estimates,  which 
depend  both  on  the  actual  coherence 
between  the  two  regions  of  interest 
and  the  power  and  phase  of  the  activity 
at  the  reference  electrode.  Reference - 
free  methods  have  been  developed 
which  do  not  have  these  problems.  An 
investment  in  coherence  studies  using 
this  newer  methodology  is  warranted. 
The  full  promise  of  electrophysio- 
logical methods  can  be  achieved  through 
their  use  in  conjunction  with  neu- 
roimaging  studies.  PET  imaging  or 
fMRI  can  be  used  to  determine  locality 
of  brain  sources,  and  EEG  data  can 
examine  the  activity  of  those  sources 
at  a  much  higher  temporal  resolution. 
MRI  or  CT  imaging  can  be  used  to 
construct  extremely  accurate  three- 
dimensional  representations  of  the 
shape  and  physical  characteristics  of 
the  brain,  and  EEG  data  collected 
from  co-registered  spatially  distributed 
electrodes  can  be  used  to  examine  the 
activity  of  cortical  sources.  That  activ- 
ity can  then  be  correlated  with  specific 
structures  identified  from  the  three- 
dimensional  representation  of  the 
brain.  Although  a  layering  of  methods 
will  provide  information  that  each 
method  alone  cannot  (indeed,  convolved 
methods  often  produce  a  synergy  of 
information),  electrophysiological 
methods  will  remain  a  unique  tool  in  the 
alcohol  research  armamentarium  because 
of  their  unparalleled  temporal  resolution. 

CONCLUSION 

It  is  vitally  important  that  NIAAA 
provide  a  framework  for  research  into 
alcohol  abuse-related  brain  dysfunction 
in  the  next  decades  by  defining  the 


516 


Brain  Dysfunction  Secondary  to  Alcohol  Abuse 


essential  questions  to  be  addressed.  A 
mandate  is  needed  for  the  establishment 
of  a  normative  database  (including 
behavioral,  structural,  and  functional 
indices)  of  brain  systems  affected  by 
alcohol  abuse.  An  investment  in 
improved  methods  for  combining 
behavioral,  structural,  and  functional 
approaches  is  warranted,  and  investi- 
gations using  a  multifaceted  approach 
should  be  encouraged. 

ACKNOWLEDGMENTS 

Preparation  of  this  chapter  was  supported 
by  NIAAA  grant  ROl  AA11311  and 
National  Institute  on  Drug  Abuse 
grant  ROl  DA09463. 

REFERENCES 

Acker,  C.  Neuropsychological  deficits  in 
alcoholics:  The  relative  contributions  of 
gender  and  drinking  history.  Br  J  Addict 
81:395-403,  1986. 

Benes,  F.M.;  Turtle,  M.;  Khan,  Y.;  and 
Farol,  P.  Myelination  of  a  key  relay  zone 
in  the  hippocampal  formation  occurs  in 
the  human  brain  during  childhood,  ado- 
lescence, and  adulthood.  Arch  Gen 
Psychiatry  51:477-484,  1994. 

Bergman,  H.  Brain  dysfunction  related  to 
alcoholism:  Some  results  from  the  KAR- 
TAD  project.  In:  Parsons  O.A.;  Butters, 
N.;  and  Nathan,  P.,  eds.  Neuropsychology 
of  Alcoholism:  Implications  for  Diagnosis 
and  Treatment.  New  York:  Guilford 
Press,  1987.  pp.  21-45. 

Cadoret,  RJ.;  Yates,  W.R;  Troughton, 
E.;  Woodworth,  G.;  and  Stewart,  M.A. 
Adoption  study  demonstrating  two  ge- 
netic pathways  to  drug  abuse.  Arch  Gen 
Psychiatry  52:42-52,  1995. 


Crabbe,  J.C.;  Belknap,  J.K.;  Metten,  P.; 
Grisel,  I.E.;  and  Buck,  KJ.  Quantitative 
trait  loci:  Mapping  drug  and  alcohol- 
related  genes.  Adv  Pharmacol 42:1033- 
1037,  1998. 

Di  Sclafani,  V.;  Clark,  H.;  Tolou-Shams, 
M.;  et  al.  Premorbid  brain  size  as  a  deter- 
minant of  functional  reserve  in  recently 
abstinent  crack-cocaine  and  crack- cocaine/ 
alcohol  dependent  subjects.  J  Int 
Neuropsychol  Soc  4:559-565,  1998. 

Diamond,  I.,  and  Messing,  RO.  Neurologic 
effects  of  alcoholism.  West  J  Med  161:279- 
287,  1994. 

Emsley,  R;  Smith,  R.;  Roberts,  M.; 
Kapnias,  S.;  Pieters,  H.;  and  Maritz,  S. 
Magnetic  resonance  imaging  in  alcoholic 
KorsakofPs  syndrome:  Evidence  for  an 
association  with  alcoholic  dementia. 
Alcohol  Alcohol  31:479-486,  1996. 

Fabian,  M.S.;  Parsons,  O.A.;  and  Sheldon, 
M.D.  Effects  of  gender  and  alcoholism  on 
verbal  and  visual-spatial  learning.  /  Nerv 
MentDis  172:16-20,  1984. 

Fein,  G.;  Raz,  J.;  Brown,  F.F.;  and  Merrin, 
E.L.  Common  reference  coherence  data 
are  confounded  by  power  and  phase  effects. 
Plectroencephalogr  Clin  Neurophysiol  69: 
581-584,  1988. 

Fein,  G.;  Bachman,  L.;  Fisher,  S.;  and 
Davenport,  L.  Cognitive  impairments  in 
abstinent  alcoholics.  West  J  Med  152:531- 
537,  1990. 

Forget,  H.,  and  Cohen,  H.  Life  after  birth: 
The  influence  of  steroid  hormones  on  cere- 
bral structure  and  function  is  not  fixed 
prenatally.  Brain  Cogn  26:243-248,  1994. 

Giedd,  J.N.;  Rumsey,  I.M.;  Castellanos,  F.X.; 
et  al.  A  quantitative  MRI  study  of  the 
corpus  callosum  in  children  and  adoles- 
cents. Brain  Res  Dev  Brain  Res91: 
274-280,  1996/?. 


517 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Giedd,  F.N.;  Snell,  J.W.;  Lange,  N.;  et  al. 
Quantitative  magnetic  resonance  imaging 
of  human  brain  development:  Ages  4-8. 
Cereb  Cortex  6:551-560,  1996£. 

Giedd,  J.N.;  Vaituzis,  A.C.;  Hamburger, 
S.D.;  et  al.  Quantitative  MRI  of  the  tem- 
poral lobe,  amygdala,  and  hippocampus  in 
normal  human  development:  Ages  4-18 
years.  JComp  Neurol  366:223-230, 1996c. 

Goldman,  M.S.;  Williams,  D.L.;  and 
Klisz,  D.K.  Recoverability  of  psychologi- 
cal functioning  following  alcohol  abuse: 
Prolonged  visual-spatial  dysfunction  in 
older  alcoholics.  /  Consult  Clin  Psychol 
51:370-378,1983. 

Grant,  I.;  Adams,  K.M.;  and  Reed,  R 
Aging,  abstinence,  and  medical  risk  fac- 
tors in  the  prediction  of  neuropsychologic 
deficit  among  long-term  alcoholics.  Arch 
Gen  Psychiatry  41:710-718,  1984. 

Graves,  A.B.;  Mortimer,  J.A.;  Larson, 
E.B.;  Wenzlow,  A.;  Bowden,  J.D.;  and 
McCormick,  W.C.  Head  circumference  as 
a  measure  of  cognitive  reserve:  Association 
with  severity  of  impairment  in  Alzheimer's 
disease.  Br  J  Psychiatry  169:86-92,  1996. 

Hatcher,  E.M.;  Jones,  M.K.;  and  Jones, 
B.M.  Cognitive  deficits  in  alcoholic 
women.  Alcoholism  1:371-377,  1977. 

Huttenlocher,  P.R.  Synaptic  density  in 
human  frontal  cortex — developmental 
changes  and  effects  of  aging.  Brain  Res 
163:195-205,  1979. 

Huttenlocher,  P.R.,  and  de  Courten,  C. 
The  development  of  synapses  in  striate 
cortex  of  man.  Hum  Neurobiol  6:1-9,  1987. 

Leber,  W.R;  Jenkins,  R.L.;  and  Parsons, 
O.A.  Recovery  of  visual-spatial  learning 
and  memory  in  chronic  alcoholics.  /  Clin 
Psychol  37:192-197,  1981. 


Mori,  E.;  Hirono,  N.;  Yamashita,  H.;  et 
al.  Premorbid  brain  size  as  a  determinant 
of  reserve  capacity  against  intellectual 
decline  in  Alzheimer's  disease.  Am  J 
Psychiatry  154:18-24,  1997. 

Panzica,  G.C.;  Garcia-Ojeda,  E.;  Viglietti- 
Panzica,  C.;  Thompson,  N.E.;  and  Ottinger, 
MA.  Testosterone  effects  on  vasotocinergjc 
innervation  of  sexually  dimorphic  medial 
preoptic  nucleus  and  lateral  septum  during 
aging  in  male  quail.  Brain  Res  712:190- 
198, 1996. 

PfefFerbaum,  A.;  Ford,  J.M.;  White, 
P.M.;  and  Mathalon,  D.  Event-related 
potentials  in  alcoholic  men:  P3  amplitude 
reflects  family  history  but  not  alcohol 
consumption.  Alcohol  Clin  Exp  Res 
15:839-850,1991. 

PfefFerbaum,  A.;  Lim,  K.O.;  Zipursky, 
R.B.;  et  al.  Brain  gray  and  white  matter 
volume  loss  accelerates  with  aging  in 
chronic  alcoholics:  A  quantitative  MRI 
study.  Alcohol  Clin  Exp  Res  16:1078- 
1089, 1992. 

PfefFerbaum,  A.;  Mathalon,  D.H.;  Sullivan, 
E.V.;  Rawles,  J.M.;  Zipursky,  R.B.;  and 
Lim,  K.O.  A  quantitative  magnetic  reso- 
nance imaging  study  of  changes  in  brain 
morphology  from  infancy  to  late  adult- 
hood. Arch  Neurol  51:874-887,  1994. 

Rakic,  P.;  Bourgeois,  J. P.;  Eckenhoff, 
M.F.;  Zecevic,  N.;  and  Goldman-Rakic, 
P.S.  Concurrent  overproduction  of  synapses 
in  diverse  regions  of  the  primate  cerebral 
cortex.  Science  232:232-235,  1986. 

Rubinow,  D.R.,  and  Schmidt,  P.J. 
Androgens,  brain,  and  behavior.  Am  J 
Psychiatry  153:974-984,  1996. 

Schofield,  P.;  Mosesson,  R;  Stern,  Y.;  and 
Mayeux,  R  The  age  at  onset  of  Alzheimer's 
disease  and  an  intracranial  area  measure- 
ment. Arch  Neurol  52:95-98,  1995. 


518 


Brain  Dysfunction  Secondary  to  Alcohol  Abuse 


Sorbel,  J.;  Morzorati,  S.;  O'Connor,  S.; 
Li,  T.K.;  and  Christian,  J.C.  Alcohol 
effects  on  the  heritability  of  EEG 
spectral  power.  Alcohol  Clin  Exp  Res 
20:1523-1527,  1996. 

Sparadeo,  F.R.;  Zwick,  W.;  and  Butters, 
N.  Cognitive  functioning  of  alcoholic 
females:  An  exploratory  study.  Drug 
Alcohol  Depend  12:143-150,  1993. 

van  Beijsterveldt,  C.E.,  and  Boomsma, 
D.I.  Genetics  of  the  human  electroen- 


cephalogram (EEG)  and  event-related 
brain  potentials  (ERPs):  A  review.  Hum 
Genet  94:319-330, 1994. 

van  Beijsterveldt,  C.E.;  Molenaar,  P.C.; 
de  Geus,  E.J.;  and  Boomsma,  D.I. 
Heritability  of  human  brain  functioning 
as  assessed  by  electroencephalography. 
Am  J  Hum  Genet  58:562-573,  1996. 

Zecevic,  N.,  and  Rakic,  P.  Synaptogenesis 
in  monkey  somatosensory  cortex.  Cereb 
Cortex  1:510-523,  1991. 


519 


SUBCOMMITTEE  REPORT 


Chapter  16 

Report  of  a  Subcommittee  of  the 

National  Advisory  Council  on  Alcohol 

Abuse  and  Alcoholism  on  the  Review 

of  the  Extramural  Research  Portfolio 

for  Neuroscience  and  Behavior 


KEY  WORDS:  AODD  (AOD  [alcohol  and  other  drug]  use  disorder);  research; 
neurology  (field);  nervous  system;  brain;  AODE  (effects  of  AOD  use,  abuse,  and 
dependence);  behavior;  theory  ofAODU  (AOD  use,  abuse,  and  dependence); 
AOD  sensitivity;  neuroimaging;  endocrine  system;  biological  adaptation;  adoles- 
cence; sleep;  cognition;  research  funding;  report 


EXECUTIVE  SUMMARY 

The  National  Institute  on  Alcohol  Abuse 
and  Alcoholism  (NIAAA)  Subcommittee 
of  the  National  Advisory  Council  on 
Alcohol  Abuse  and  Alcoholism  on  the 
Review  of  the  Extramural  Research 
Portfolio  for  Neuroscience  and  Behavior 
met  May  11-13,  1998.  The  charge  to 
the  subcommittee  was  to  examine  the 
appropriateness  of  the  breadth,  coverage, 
and  balance  of  the  neuroscience  and 
behavior  research  portfolio,  identifying 
research  areas  that  are  well  covered  and 
others  that  are  either  underinvestigated 
or  otherwise  warrant  significantly 
increased  attention.  The  subcommit- 
tee was  asked  also  to  provide  specific 
advice  and  guidance  on  the  scope  and 


direction  of  NIAAA's  extramural 
research  activities  in  the  neuroscience 
and  behavior  area. 

The  subcommittee  consisted  of 
two  NIAAA  Advisory  Council  co-chairs 
and  an  advisory  group  of  17  individuals. 
Sixteen  of  these  individuals  have  demon- 
strated expertise  in  alcohol-related  areas, 
and  three  individuals  have  demon- 
strated expertise  in  non-alcohol- 
related  areas  (see  Appendix  A). 

The  review  process  was  initiated  by 
having  experts  (see  Appendix  B)  in 
neuroscience  and  behavior  prepare 
written  assessments  of  the  state  of 
knowledge,  gaps  in  knowledge,  and 
research  opportunities.  NIAAA  program 
staff  (see  Appendix  C)  presented  the 
current  extramural  portfolio,  categorized 


523 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


into  the  areas  of  basic  neuroscience 
research,  molecular  adaptive  responses, 
neuroendocrine  system,  studies  of  the 
acute  and  chronic  effects  of  alcohol  in 


behavioral/structural  deficits  in 
humans.  All  information  was  shared 
with  experts,  selected  NIAAA  staff, 
and  the  co- chairs  and  advisory  group 


animals  and  in  humans,  and  cognitive/     before  the  meeting. 


Table  1.  NIAAA  FY97  Awards  in  Neuroscience  and  Behavior. 


Dollar  Amount 

Type  of  Award 

Number 

(thousands) 

Research  project  grants 

121 

21,677 

Research  centers 

4a 

6,744 

Research  careers 

17 

1,218 

Research  training 

29 

2,012 

Total 

171 

31,651 

No. 


%  of  Total 
Amount 


22 
29 

28 

57 
25b 


17 
30 
21 
35 
20b 


aOf  the  14  NIAAA  centers,  4  are  exclusively  neuroscience  and  behavior.  Seven  of  the  14  centers  have  neuroscience  and 
behavior  components;  5  of  these  estimate  that  a  50  percent  of  the  research  budget  of  the  center  is  invested  in 
neuroscience  and  behavior. 
"Percentage  of  all  NIAAA  awards  in  FY97. 


Table  2.  NIAAA  FY97  Research  Project  Grants  and  Research  Careers  Awards  in  Neuroscience  and 

Behavior. 

Number  of 

%of 

Category 

Awards 

FY97  Awards* 

Basic  neuroscience 

Research  project  grants 

30 

25 

Research  careers 

5 

29 

Molecular  adaptive  responses 

Research  project  grants 

22 

18 

Research  careers 

7 

41 

Neuroendocrine  system 

Research  project  grants 

14 

11 

Research  careers 

0 

0 

Acute  and  chronic  behavioral  effects  of  alcohol 

Animals 

Research  project  grants 

35 

29 

Research  careers 

2 

12 

Humans 

Research  project  grants 

8 

7 

Research  careers 

2 

12 

Cognitive/behavioral/structural  deficits  in 

humans 

Research  project  grants 

12 

10 

Research  careers 

1 

6 

'Percentage  of  FY97  awards  in  neuroscience  and  behavior. 

524 


Subcommittee  Report 


At  the  meeting  in  May  1998, 
experts  and  NIAAA  program  staff 
made  abbreviated  presentations  of  their 
material  followed  by  discussion  among 
all  of  the  participants,  including  rep- 
resentatives from  other  Institutes  of 
the  National  Institutes  of  Health 
(NIH)  and  guests  (see  Appendix  D). 
After  completing  this  process,  the 
co-chairs  and  advisory  group,  with 
input  from  the  experts,  delineated  the 
following  list  of  research  priorities,  in 
order  of  importance: 

1.  Increase  emphasis  on  mechanisms 
associated  with  various  models  of  neu- 
roadaptation  (sensitization,  tolerance, 
dependence,  withdrawal,  recovery), 
at  multiple  levels  of  analysis — that  is, 
molecular,  cellular,  and  whole  ani- 
mal, including  human  and  nonhu- 
man  primates. 

2.  Emphasize  new  genetic  techniques 
that  have  great  potential  for  under- 
standing the  effects  of  alcohol  on 
brain  and  behavior.  Techniques 
include  transgenics,  knockouts, 
knockins,  antisense  oligonucleotides, 
tissue-specific  expression,  and  viral- 
mediated  gene  transfer.  Enhanced 
development  and  availability  of 
these  genetically  altered  animals  may 
have  to  be  subsidized  by  NIAAA. 

3.  Increase  emphasis  on  delineating  the 
cellular  and  molecular  mechanisms 
of  neuropathophysiology  and  neuro- 
pathology resulting  from  exposure 
to  alcohol.  This  should  be  accom- 
plished in  areas  of  the  brain  previ- 
ously demonstrated  to  be  highly 
sensitive  to  the  effects  of  alcohol,  such 


as  hippocampus,  frontal  cortex,  cere- 
bellum, accumbens,  hypothalamus, 
and  ventral  tegmental  area. 

4.  Some  of  the  research  questions  in  neu- 
roscience  and  behavior  require  col- 
laborations by  experts  in  a  number 
of  different  disciplines.  Moreover, 
involving  the  best  and  most  appro- 
priate scientists  often  requires 
interinstitution  collaboration.  Conse- 
quently, NIAAA  is  encouraged  to 
use  appropriate  funding  mecha- 
nisms that  will  facilitate  such  col- 
laborative studies. 

Other  areas  mentioned,  in  order  of 
importance,  included: 

5.  Support  multisite,  prospective  longi- 
tudinal studies  designed  to  examine 
vulnerability  and  protective  effects  of 
alcohol  on  brain  and  behavior.  Spec- 
ified individuals  should  be  examined 
longitudinally  with  the  techniques 
of  imaging,  electrophysiology,  and 
neuropsychology.  Alcohol,  other 
drug,  and  neuroendocrine  challenges 
should  be  conducted  when  ethi- 
cally appropriate. 

6.  Support  studies  of  the  effects  of  alco- 
hol on  intracellular  signaling  sys- 
tems. This  area  is  underrepresented 
in  the  portfolio,  especially  given  the 
recent  explosion  of  information  on 
intracellular  signaling. 

7.  Increase  the  number  of  animal  neu- 
robehavioral  phenotypes  studied,  such 
as  animal  analogs  of  impulsive  or 
disinhibited  behaviors  and  their 
interactions  with  alcohol. 


525 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


8.  Develop  and  enhance  availability  of 
new  imaging  techniques,  with  consid- 
eration of  a  dedicated  imaging  center. 

These  8  areas  of  importance  were 
derived  from  a  list  of  22.  The  remain- 
ing 14  are  listed  by  topic  without 
regard  to  priority. 

General  Considerations 

•  There  should  be  an  increased  empha- 
sis on  training  in  the  neurosciences, 
especially  mentored  training. 

•  Alcohol-related  ligands  and  reagents 
should  be  developed  and  their 
availability,  at  reduced  cost,  should 
be  increased. 

•  NIAAA  should  assist  investigators 
in  gaining  access  to  alcohol -related 
investigational  new  drugs. 

•  An  informatics  type  of  database  for 
alcohol  research  should  be  devel- 
oped, including  information  on 
molecular,  cellular,  genetic,  behav- 
ioral, and  pathologic  effects  of 
alcohol.  The  database  would  be 
designed  to  provide  information  in 
a  format  that  would  be  useful  to 
investigators  addressing  specific 
and  complex  questions. 

•  Support  for  alcohol-related  neuro- 
endocrine research  at  all  levels  of 
analysis  should  be  enhanced.  Repre- 
sentative topics  include  responses  to 
stress  and  alcohol-neuroendocrine- 
immune  interactions. 

•  Dose-response  relationships  are  par- 
ticularly informative.  The  effects  of 
moderate  doses  of  alcohol  on  the 
central  nervous  system  are  underin- 
vestigated,  particularly  in  animal 
models  and  humans. 


Molecular 

and  Cellular  Studies 

•  Effects  of  alcohol  on  lipid-protein 
interactions  should  be  emphasized; 
effects  of  alcohol  on  lipids  alone  are 
of  more  limited  value.  Particular 
emphasis  should  be  placed  on  the 
identification  of  proteins  with  rea- 
sonable sensitivity  to  alcohol  that 
can  readily  be  studied  in  the  presence 
of  different  lipids. 

•  A  program  for  the  structural  analyses 
of  alcohol-protein  interactions, 
including  molecular  modeling  as 
well  as  physical  measurements, 
should  be  developed.  This  could 
be  accomplished  by  establishing 
dedicated  groups  with  equipment 
and  facilities  or  by  supplementing 
existing  grants  to  allow  incorpora- 
tion of  structural  initiatives. 


Animal  Models 

•  The  use  of  nonhuman  primate 
models  should  be  increased,  includ- 
ing studies  of  the  effects  of  alcohol 
on  adolescent  and  aged  animals. 

•  Support  for  neurobiological  studies 
of  acute  and  chronic  alcohol  expo- 
sure in  adolescent  animals  should 
be  increased. 

Human  Studies 

•  An  alcoholic  brain  bank  should  be 
established,  perhaps  by  supple- 
menting an  existing  bank.  Accurate 
information  on  diagnoses,  current 
drug  use,  and  concomitant  medical 
conditions  is  essential.  Antemortem 


526 


Subcommittee  Report 


brain  structure-function  studies 
would  be  valuable. 

•  Studies  of  alcohol-related  nosology 
and  concomitant  comorbidity 
with  other  psychiatric  and  brain  dis- 
orders, including  dementia,  would 
be  informative. 

•  Emphasis  on  alcohol  and  sleep 
research  should  be  increased. 

•  Studies  of  neurobiological  factors 
associated  with  relapse  are  important. 

Additional  gaps  in  knowledge  and 
research  opportunities  were  deter- 
mined by  experts  in  each  of  the  areas 
covered  and  are  listed  in  the  relevant 
sections  of  this  chapter. 

OVERVIEW  OF  THE 
NEUROSCIENCE 
AND  BEHAVIORAL 
RESEARCH  PROGRAM 

Walter  A.  Hunt,  Ph.D. 

This  overview  provides  information  on  the 
overall  programmatic  balance  of  the  port- 
folio, with  all  analyses  based  on  awards 
made  in  fiscal  year  1997  (FY97).  Spe- 
cific aspects  of  the  portfolio  will  be  pre- 
sented in  other  sections  of  this  chapter. 

The  overall  portfolio  consists  of  121 
grants  at  a  total  cost  of  $21,677,142 
(see  table  1).  The  balance  of  neurosci- 
ence  grants  to  behavior  grants  is  almost 
equal.  Most  of  the  research  funded  is 
basic  research,  with  very  little  of  the  port- 
folio considered  applied  research. 
Those  few  grants  that  are  applied 
research  relate  to  the  development  of 
medications  at  the  preclinical  or  early 
clinical  level. 

About  80  percent  of  the  grants, 
representing  77  percent  of  the  dollar 


amount  of  the  awards,  used  animals; 
about  20  percent,  representing  23 
percent  of  the  dollar  amount  of  the 
awards,  used  humans.  About  70  percent 
of  the  grants  were  in  vivo  studies,  repre- 
senting 68  percent  of  the  dollar  amount 
of  the  awards.  Animal  and  human  stud- 
ies represent  62  percent  of  the  number 
of  grants  awarded,  with  molecular  and 
cellular  studies  reflecting  the  remainder 
in  approximately  equal  amounts. 

The  following  list  of  priorities  rep- 
resents common  themes  among  vari- 
ous initiatives: 

•  Identify  relevant  targets  of  alcohol 
in  the  brain. 

•  Identify  neural  circuits  underlying 
the  behavioral  effects  of  alcohol. 

•  Determine  what  neurobiological 
actions  of  alcohol  are  responsible 
for  its  acute  and  chronic  behavioral 
effects. 

•  Determine  how  the  effects  of  alcohol 
on  the  brain  and  behavior  contribute 
to  the  development  of  alcoholism. 

•  Determine  neurobiological  and  behav- 
ioral factors  that  render  adolescents 
more  vulnerable  than  adults  to  abuse 
alcohol  and  become  alcoholic. 

•  Develop  prototypic  compounds  for 
potential  therapeutic  development. 

The  last  recommendation  evolves 
from  the  need  to  translate  basic 
research  findings  into  clinical  applica- 
tions. To  that  end,  a  Medications 
Development  Working  Group  was 
recently  formed  to  determine  if 
research  findings  from  basic  preclinical 
work  could  provide  a  basis  for  devel- 
oping potential  agents  for  clinical  trials 
to  treat  alcoholism.  A  short-term  goal 


527 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


is  a  workshop  to  bring  together  individ- 
uals from  academia,  industry,  and  gov- 
ernment to  build  bridges  among  the 
groups,  review  the  process  by  which 
compounds  are  developed  into  drugs  for 
clinical  trials,  and  solicit  ideas  about  how 
NIAAA  can  help  facilitate  this  process. 


TRAINING  AND  CAREER 
DEVELOPMENT  IN 
NEUROSCIENCE  AND 
BEHAVIORAL  RESEARCH 

Walter  A.  Hunt,  Ph.D. 

Training  of  new  investigators  is  sup- 
ported through  several  mechanisms, 
depending  on  the  educational  level  and 
experience  of  the  applicant.  Much  of 
the  program  is  based  on  individual  fel- 
lowships and  training  grants.  The 
remainder  of  the  program  supports 
scientist  development  awards  at  differ- 
ent levels  of  experience. 

Overall  Training  Program 

Forty-six  competitive  and  noncom- 
petitive awards  were  funded  in  FY97. 
Of  those,  19  were  individual  fellow- 
ships, 10  were  training  grants,  and 
17  were  scientist  development 
awards.  Thirty  awards  supported  neu- 
roscience research  and  16  supported 
behavioral  research. 

Individual  Fellowships 

The  balance  between  predoctoral  and 
postdoctoral  awards  is  fairly  even,  with 
a  slightly  greater  number  for  predoc- 
toral awards.  Based  on  NIH  policy, 
the  commitment  to  individual  fellow- 
ships relative  to  training  grants  should 
be  at  least  15  percent  of  the  available 


funds.  In  FY97,  the  figure  for  NIAAA 
was  19  percent. 

Only  one  of  the  awards  is  for  a 
potential  clinical  researcher.  Four 
involve  neurotransmitter  receptors, 
three  use  genetic  approaches,  two 
study  ontogenetic  differences  in  the 
effects  of  alcohol,  three  examine  the 
motivational  effects  of  alcohol,  four 
are  on  the  central  control  of  alcohol 
reinforcement,  one  is  on  stress  and 
neurosteroids,  and  two  are  minority 
predoctoral  fellowships. 

Institutional  Training  Grants 

The  10  training  grants  support 
57  slots  for  21  predoctoral  and  34 
postdoctoral  trainees.  The  relative 
balance  between  neuroscience  train- 
ing slots  and  behavioral  training 
slots  is  almost  equal.  Three  grants 
educate  trainees  in  using  genetic 
approaches  to  discover  the  mecha- 
nisms underlying  actions  of  alcohol 
on  the  brain,  4  deal  with  basic 
neurochemical  mechanisms,  2  with 
neurotoxic  actions  of  alcohol,  1  with 
brain  imaging,  and  3  with  behavioral 
studies.  Two  training  grants  primarily 
use  human  subjects. 

Scientist  Development  Awards 

Of  the  17  awards,  most  were  men- 
tored  awards,  and  almost  half  were  to 
junior,  mentored  investigators.  All  of 
the  mentored  awards  support  investi- 
gators in  neuroscience  research.  Only 
two  awards  support  investigators  in 
clinical  research.  Most  of  the  investi- 
gators study  basic  actions  of  alcohol 
on  neurotransmitter  receptors  or  sig- 
nal transduction  systems.  Two  grants 
have  behavioral  components. 


528 


Subcommittee  Report 


Recruitment 

and  Future  Directions 

Recruitment  efforts  involve  staff 
contacts  with  potential  applicants 
at  scientific  meetings.  In  addition,  site 
visits  of  NIAAA- funded  training  grants 
were  undertaken  to  introduce  trainees 
to  the  grant  process  and  inform 
them  about  the  benefits  of  individual 
fellowships  and  mentored  scientist 
development  awards.  These  visits 
resulted  in  increased  applications  but 
have  been  discontinued  because  of  lim- 
ited travel  funds. 

The  training  program  is  fairly  mature, 
with  most  mechanisms  of  training  ade- 
quately represented.  However,  increased 
training  of  clinical  investigators  could 
provide  an  additional  pool  of  needed 
scientists  to  pursue  alcohol  research 
with  human  subjects. 

MOLECULAR  AND 
CELLULAR  EFFECTS 
OF  ALCOHOL 

Acute  Actions  of  Alcohol 
on  Specific  Neural  Targets 

Neurotransmission,  Receptors, 
Transporters,  Modulators, 
Ion  Channels  and  Signal 
Transduction:  Gaps  in  Knowledge 
and  Research  Opportunities 

David  M.  Lovinger,  Ph.D. 

•  A  greater  emphasis  should  be  placed 
on  describing  alcohol-induced 
alterations  in  the  molecular  struc- 
ture and  dynamics  of  neural  pro- 
teins that  are  sensitive  to  the 
effects  of  alcohol. 


•  Analysis  of  relationships  between  alco- 
hol sensitivity  and  protein  domain 
structure  should  be  encouraged  in 
order  to  characterize  the  molecular 
sites  of  alcohol  actions. 

•  Changes  in  protein  function  asso- 
ciated with  exposure  to  alcohol  are 
important  to  delineate. 

•  It  is  important  to  determine  alco- 
hol effects  on  synaptic  transmission 
using  techniques  that  infer  a  pre-  or 
post-synaptic  locus  of  effect.  Specific 
synaptic  proteins  should  be  identified. 

•  A  greater  emphasis  should  be  placed 
on  discerning  the  effects  of  alcohol 
on  the  neurophysiology  of  the 
ventral  tegmental  area,  amygdala, 
hypothalamus,  cerebellum,  and 
prefrontal  cortex. 

Lipid  Involvement  in  the  Acute 
Actions  of  Alcohol  in  the  Nervous 
System:  Gaps  in  Knowledge  and 
Research  Opportunities 

Steven  N.  Treistman,  Ph.D. 

•  Increased  emphasis  on  protein 
function  as  the  measure  of  lipid 
perturbations  will  be  informative. 

•  The  function  of  a  particular  protein 
should  be  well  characterized  before 
attempts  are  made  to  assess  pertur- 
bations of  function  by  alcohol.  Ideally, 
this  should  include  information  in 
native  membranes  as  well  as  in  arti- 
ficial bilayers. 

•  Reasonable  (physiological)  concen- 
trations of  alcohol  should  be  used 
in  determining  its  effects. 

•  Studies  should  be  integrated  across 
multiple  levels  of  analysis:  (a)  a  target 
protein  of  known  behavioral  or  physio- 
logical relevance  to  alcohol  action 


529 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


should  be  selected;  (b)  effects  of  lipid 
perturbation  and  possibly  modulation 
of  lipid  composition  on  the  protein  in 
its  native  environments  should  be 
determined;  (c)  the  target  protein 
should  be  capable  of  being  reconsti- 
tuted into  a  simplified  lipid  environ- 
ment for  examining  alcohol  effects; 
and  (d)  a  cloned  protein  should  be 
selected  to  permit  expression  studies 
in  a  variety  of  native  membranes  and 
to  enable  mutagenesis  studies. 
•  The  study  of  the  significant  role  of 
lipids  in  the  compensatory  responses 
of  cells  exposed  to  alcohol  should 
be  continued. 

NIAAA  Basic  Neuroscience 
Research  Portfolio 

Yuan  Liu,  Ph.D. 

In  FY97,  the  NIAAA  Basic  Neuroscience 
Research  Program  funded  35  extramural 
grants  for  a  total  of  $5.5  million.  Of 
these,  30  were  basic  research  project 
grants,  and  5  were  research  careers 
awards  (see  table  2). 

This  program  supports  a  wide  variety 
of  investigations  that  explore  different 
effects  of  alcohol  on  the  brain  at  multi- 
ple levels  of  analysis  and  use  a  number 
of  different  neuroscience  techniques. 
Several  different  molecular  targets  are 


being  examined,  as  are  different  mech- 
anisms hypothesized  to  underlie  the 
actions  of  alcohol  in  the  brain.  Tables  3, 
4,  and  5  show  the  distribution  of  grants 
by  level  of  analysis,  molecular  target,  and 
brain  region,  respectively.  In  table  3, 
the  categories  are  not  mutually  exclu- 
sive: some  investigators  are  studying  the 
effects  of  alcohol  at  multiple  levels.  Table 
4  shows  that  identifying  the  molecular 
targets  and  elucidating  the  mechanisms 
underlying  the  interactions  between  alco- 
hol and  these  targets  has  been  the  research 
focus  of  more  than  half  of  the  basic  neu- 
roscience research  portfolio.  As  shown  in 
table  5,  identifying  the  cellular  targets  of 
alcohol  and  their  distribution  in  the  brain 
is  another  major  focus  of  research. 

More  than  half  of  the  total  awards  in 
the  portfolio  examine  the  acute  effects 
of  alcohol  on  the  brain,  about  one -third 
study  the  chronic  effects,  and  the  remain- 
der investigate  both  short-  and  long-term 
effects.  Only  a  few  research  projects  study 
directly  the  mechanisms  underlying 
various  interactions  phases  of  alcohol- 
related  problems,  including  tolerance  and 
withdrawal;  the  majority  of  projects  study 
the  direct  action  between  alcohol  and 
candidate  targets.  About  80  percent  of 
investigators  use  multidisciplinary 
approaches,  primarily  a  combination  of 


Table  3.  NIAAA  Basic 

Neuroscience  Grant  Distribution 

by 

Level 

of  Analysis. 

Level  of  Analysis 

Number 

%  of  Total 

Molecular 

18 

51 

Cellular 

7 

20 

Neural  circuitry 

3 

9 

Miscellaneous 

7 

20 

Total 

35 

100 

530 


Subcommittee  Report 


electrophysiological  and  molecular  bio- 
logical techniques.  Nearly  80  percent  of 
the  research  teams  use  in  vitro  prepa- 
rations. Among  the  projects  studying 
molecular  targets  of  alcohol,  about  80 
percent  of  the  experiments  use  recom- 
binant receptor-channel  proteins. 

Research  Areas 

Molecular  Level  (18 grants) 

Alcohol,  unlike  most  other  highly 
abused  substances,  does  not  act  on  a 
single  specific  target  in  the  brain.  Instead, 
it  interacts  with  many  targets,  such  as 
neurotransmitter  receptors  and  voltage  - 
gated  ion  channels  on  nerve  cells.  A 
major  challenge  confronting  molecular 
studies  is  the  lack  of  uniform  effects  of 
alcohol  on  various  cell  types  in  different 


regions  in  the  brain.  Several  complemen- 
tary working  hypotheses  address  the 
cellular  and  regional  specificity  of  alco- 
hol. The  first  postulates  that  alcohol 
directly  interacts  with  specific  amino 
acid  residues  or  domains  of  the  target 
proteins.  The  second  proposes  that 
expression  of  particular  receptor  sub- 
unit  combinations  contribute  to  the 
different  degrees  of  alcohol  sensitivity 
in  different  neurons.  A  more  recent 
hypothesis  suggests  that  posttransla- 
tional  modifications,  such  as  the  phos- 
phorylation status  of  a  target  protein, 
can  determine  sensitivity  to  alcohol. 

Direct  Molecular  Action  Sites  (8 
grants).  The  majority  of  research  pro- 
jects within  this  category  are  attempting 
to  identify  the  sites  of  alcohol  action 
on  target  molecules,  including  voltage- 


Table  4.  NIAAA  Basic  Neuroscience  Grant  Distribution 

by 

Molecular  Targets 

of  Alcohol. 

Molecular  Target 

Number 

%  of  Total 

Acetylcholine  receptors 

4 

22.2 

GABAA  receptors 

4 

22.2 

NMDA  receptors 

5 

27.8 

Glycine  receptor 

1 

5.6 

5-HT3  receptor 

1 

5.6 

CA++  channels 

2 

11.1 

K+  channels 

1 

5.6 

Total 

18 

100.0 

Table  5.  NIAAA  Basic  Neuroscience  Grant  Distribution  by 

Studies  of  Brain 

Regions. 

Brain  Region 

Number 

%  of  Total 

Amygdala 

1 

2.8 

Cortex 

2 

5.7 

Cerebellum 

2 

5.7 

Hippocampus 

12 

34.3 

Mesolimbic  system 

8 

22.8 

>  4  Brain  regions 

7 

20.0 

N/A 

13 

37.1 

Total 

45 

128.5 

531 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


gated  Ca++  and  K+  channels  and 
acetylcholine,  gamma-aminobutyric 
acid  type  A  (GABAA),  N-methyl-D- 
aspartate  (NMDA),  and  glycine  recep- 
tors. One  working  hypothesis  is  that 
alcohol  alters  channel  kinetics,  such  as 
channel  open  time,  channel  closing  time, 
and  desensitization  status  of  molecular 
targets.  Other  investigators  study  protein 
targets  in  their  native  environments.  The 
majority  use  a  combination  of  electro- 
physiological and  molecular  biological 
approaches  to  test  alcohol  effects  on 
recombinant  proteins  expressed  in 
Xenopus  oocytes  or  transfected  mam- 
malian cell  lines.  Two  research  groups 
introduced  a  state-of-the-art  approach — 
the  chimeric  receptor  technique — to 
alcohol  research.  Chimeric  receptors 
consist  of  complementary  parts  of  two 
target  proteins  that  give  opposite 
responses  to  alcohol.  By  using  a  well- 
designed  set  of  chimeras,  the  process 
of  locating  the  sites  of  action  of  alcohol 
can  be  significantly  accelerated. 

There  has  been  a  long  debate  over 
the  question  of  whether  lipid,  protein, 
or  the  interface  between  them  is  the  site 
of  action  of  alcohol.  Although  increasing 
evidence  suggests  that  membrane  pro- 
teins are  the  direct  targets,  the  role  of 
the  lipid  environment  in  mediating 
effects  of  alcohol  is  likely  to  be  impor- 
tant. Two  projects  are  attempting  to 
further  clarify  this  challenging  and 
confusing  issue.  Both  projects  use 
preparations  that  incorporate  well- 
characterized  protein  candidates  into 
a  lipid  bilayer.  In  this  environment, 
each  lipid  component  is  known  and 
can  be  individually  manipulated. 

Subunit  Composition  (8  grants). 
Both  voltage-gated  and  ligand-gated 


channel  proteins  are  composed  of 
multiple  subunits.  The  putative  mole- 
cular structure  of  the  superfamily  of 
ligand-gated  channels/neurotransmit- 
ter  receptors  is  a  pentamer  consisting 
of  five  identical  or  distinct  subunits.  It 
is  evident  that  the  number  of  possible 
combinations  of  subunits  is  quite 
large.  In  addition,  an  individual  neu- 
ron can  express  multiple  types  of  a 
given  receptor,  based  on  the  combina- 
tion of  different  subunits.  When  con- 
sidering the  many  different  regions  in 
the  brain,  the  patterns  of  expression  of 
those  subunit  compositions  can  be 
exceedingly  complex. 

Currently,  eight  research  groups 
supported  by  this  program  are  testing 
the  "subunit  composition"  hypothesis. 
The  two  major  approaches  that  are 
investigating  the  structure-function 
relationship  use  either  (a)  recombinant 
proteins  produced  in  various  expres- 
sion systems  or  (b)  natural  proteins 
expressed  in  intact  neurons.  Several 
research  groups  are  taking  advantage  of 
some  of  the  newest  pharmacological 
tools.  The  binding  selectivity  to  subunits 
of  these  agonists  or  antagonists  appears 
to  correlate  with  alcohol  specificity  of 
certain  subunits.  Other  investigators 
are  using  different  approaches,  such  as 
specific  antibodies  against  the  candidate 
subunits.  A  novel  technique — the  single 
cell  reverse  transcription-polymerase 
chain  reaction  (RT-PCR)  method — 
has  recently  been  introduced  to  alcohol 
research.  This  sophisticated  method, 
when  properly  used,  can  identify  the 
profile  of  the  subunits  at  the  messenger 
RNA  level  in  an  identified  cell. 

Protein  Modifications  (2  grants). 
Previous  observations  suggest  that 


532 


Subcommittee  Report 


phosphorylation/dephosphorylation 
status  of  some  target  proteins,  such  as 
GABAA,  NMDA,  and  5-hydroxytrypt- 
amine  type  3  (5-HT3)  receptors, 
might  be  important  for  understanding 
the  interactions  between  alcohol  and 
these  targets.  Two  research  projects  are 
exploring  this  hypothesis  by  applying 
a  combination  of  electrophysiological, 
biochemical,  and  molecular  biological 
techniques  on  recombinant  receptors. 

Cellular  Level  (7 grants) 

Two  projects  are  examining  the 
effects  of  alcohol  at  the  single  cell 
level.  One  project  is  using  the  brain 
slice  preparation  and  electrophysio- 
logical recording  techniques  to  study 
the  acute  effects  of  alcohol.  A  second 
study  is  using  both  brain  slice  and  in 
vivo  preparations  to  explore  the 
chronic  effects  of  alcohol  on  the  spon- 
taneous activity  of  dopaminergic  cells 
in  the  ventral  tegmental  area. 

Most  projects  are  focused  at  the 
synaptic  level.  For  example,  one  inves- 
tigator is  using  an  in  vitro  NMDA 
synapse  model  to  identify  cellular 
mechanisms  underlying  the  develop- 
ment of  acute  tolerance  to  alcohol. 
Another  investigator  is  using  both  tra- 
ditional and  organotypic  slice  prepara- 
tions to  explore  the  alteration  of 
voltage-gated  Ca++  channels  following 
acute  and  chronic  alcohol  exposure. 

Several  projects  are  investigating 
the  effects  of  alcohol  on  synaptic  plas- 
ticity. One  project  is  testing  the 
hypothesis  that  subcortical  inputs  to 
the  dentate  gyrus  mediate  the  acute 
actions  of  alcohol  on  both  short-  and 
long-term  plasticity  in  hippocampus. 
Another  project  is  exploring  a  possi- 


ble role  for  the  amygdala  in  mediating 
the  acute  effects  of  alcohol  on  synap- 
tic plasticity.  One  study  uses  hip- 
pocampal  activity-related  spatial 
memory  as  a  model  system  to  test  the 
effects  of  alcohol  on  different  age 
groups  of  animals. 

Neural  Circuitry  Level  (3 grants) 

Research  at  the  neural  circuitry  level 
has  not  yet  been  intensively  pursued. 
Investigators  supported  by  this  pro- 
gram have  recently  developed  several 
novel  methods  that  are  further  exten- 
sions of  the  traditional  extracellular 
single-unit  recording  techniques.  One 
uses  a  multielectrode,  single-unit 
recording  apparatus  that  can  perform 
simultaneous  recordings  of  neuronal 
activities  from  different  areas  of  a 
defined  neural  circuit  during  a  specific 
behavioral  paradigm.  This  allows  the 
analysis  of  spatial/temporal  pattern 
changes  of  neuronal  firing  related  to 
alcohol-induced  behaviors.  Another 
new  method  combines  in  vivo  micro- 
dialysis  with  electrophysiological  and 
behavioral  techniques.  This  provides  a 
means  of  locally  delivering  alcohol 
and  other  pharmacological  agents  to 
individual  neurons,  simultaneously 
recording  neuronal  activity,  and  collect- 
ing neurotransmitters  released  from 
the  same  neuron  in  real  time,  during 
ongoing  behaviors. 

Recommendations 

Molecular  Level 

Direct  Molecular  Action  Sites 

•   Emphasize  less-studied  molecular 
targets  (e.g.,  serotonin  receptors, 


533 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


peptide  neurotransmitters,  neuro- 
transmitter transporters). 

•  Identify  action  sites  of  alcohol  at 
the  level  of  single  amino  acids. 

•  Examine  channel  kinetics  of  target 
proteins  at  the  single-channel  level. 

•  Study  well-characterized  protein 
targets  in  a  known  lipid  environment. 

Subunit  Composition 

•  Emphasize  studies  of  natural  receptors 
in  intact  neurons,  rather  than  focusing 
specifically  on  recombinant  receptors. 

•  Identify  subunit  composition  pro- 
files by  single-cell  RT-PCR  and  by 
subunit-specific  antibodies. 

•  Characterize  the  role  of  subunit 
composition  as  a  mediator  of  alco- 
hol's effects  using  antisense  tech- 
niques and  inducible  knockout, 
tissue-specific  knockout  techniques. 

Protein  Modifications 

•  Examine  a  larger  variety  of  protein 
kinase  pathways. 

•  Investigate  the  role  of  protein 
phosphorylation  and  alcohol  inter- 
actions, using  phosphorylation, 
state -specific  antibodies,  and  genet- 
ically engineered  animals. 

•  Explore  mechanisms  other  than 
phosphorylation  modifications. 

Cellular  Level 

•  Investigate  mechanisms  of  alcohol's 
actions  on  neuronal  firing  rate  at 
molecular  and  cellular  levels. 

•  Determine  the  effects  of  alcohol 
on  synaptic  transmission  (e.g., 
neurotransmitter  release  stat- 
istics and  quantal  efficiency,  presy- 
naptic modification,  postsynaptic 
regulation). 


•  Characterize  the  effects  of  alcohol 
at  integrated  levels  (e.g.,  interac- 
tions between  excitatory  and 
inhibitory  receptors  at  single-cell 
and  pathway  levels). 

•  Study  the  effects  of  alcohol  on  synap- 
tic plasticity,  using  methods  such  as 
minimal  stimulation,  stimulation- 
induced  miniature  potential,  and 
quantal  analysis. 

•  Identify  effects  of  alcohol  on 
synaptic  plasticity  at  locations 
other  than  hippocampal  NMDA- 
dependent  long-term  potentiation 
(LTP)  (e.g.,  non-NMDA-depen- 
dent  hippocampal  LTP,  cerebellar 
long-term  depression  [LTD] 
and  LTP,  and  synaptic  plasticity 
in  other  areas  of  the  brain,  includ- 
ing the  mesolimbic  system, 
basal  ganglia,  thalamic  relays, 
and  neocortex). 

Neural  Circuitry  Level 

•  Use  single-  or  multiple  single-unit 
recordings  in  behaving  animals  to 
associate  effects  of  alcohol  directly 
to  behavior. 

•  Use  combinations  of  neurochemical 
(e.g.,  iontophoresis,  microdialysis,  and 
voltammetry)  and  electrophysiological 
recordings  in  behaving  animals  to 
associate  neurochemical  and  neuro- 
physiological  events  with  alcohol- 
induced  behaviors. 

•  Encourage  the  use  of  sophisticated 
optical  recording  techniques 
and  voltage-sensitive  and  CA++ 
sensitive  dyes  in  brain  slice  prepara- 
tions to  study  the  activity  of  large 
neural  networks  under  the  influence 
of  alcohol,  as  well  as  studies  of  intrin- 
sic optical  activity  using  the  newest 


534 


Subcommittee  Report 


computer  algorithms  coupled  with 
modern  photorecording  techniques. 
Use  computational  modeling  meth- 
ods in  combination  with  physiolog- 
ical and  behavioral  empirical 
studies  to  explore  how  neural  cir- 
cuitry synchronizes  functions  dur- 
ing alcohol-induced  behaviors. 


Effects  of  Alcohol  on  the 
Neuroendocrine  System:  Gaps 
in  Knowledge  and  Research 
Opportunities 

Catherine  Rivier,  Ph.D. 

Despite  the  involvement  of 
corticotropin- releasing  factor  (CRF)  in 
many  alcohol-related  disorders,  and 
the  fact  that  this  peptide  appears  to  be 
a  reinforcer  of  drug  abuse,  the  num- 
ber of  studies  supported  by  NIAAA 
that  investigate  its  synthesis,  release, 
and  effects  on  endocrine  function  in 
general,  and  the  hypothalamic-pitu- 
itary-adrenal  (HPA)  axis  in  particular, 
is  quite  low. 

•  A  greater  emphasis  should  be  placed 
on  studies  investigating  the  func- 
tional interactions  between  CRF 
and  neurotransmitters  involved  in 
drug- seeking  behavior,  reinforce- 
ment, and  relapse. 

•  Exposure  of  adult  mice  with  a  con- 
ditional null  mutation  for  the  CRF 
or  CRF  receptor  gene  to  alcohol 
would  provide  valuable  informa- 
tion. 

•  Exposure  of  pregnant  dams  lacking 
the  CRF  or  CRF-R1  gene  to 
alcohol  could  provide  useful  infor- 
mation on  the  endocrine,  behav- 


ioral, autonomic,  and  immune 
pathologies  observed  in  fetal 
alcohol  syndrome. 
Studies  of  the  functional  interac- 
tions between  alcohol  and  nitric 
oxide/carbon  monoxide,  and 
between  alcohol  and  nuclear 
regulatory  factor-KB  (NF-kB), 
are  important. 

A  good  model  of  isolated  cells  (either 
primary  culture  or  immortalized 
cells)  that  produce  CRF  is  needed. 
The  development  of  potent  CRF 
antagonists  that  are  long-lasting 
and  receptor  specific  should  be 
encouraged. 

NIH-distributed  reagents  for  the 
measurement  of  plasma  adrenocor- 
ticotropic hormone  (ACTH)  and 
corticosterone  levels  in  rodents  are 
urgently  needed. 

It  would  be  useful  to  have  access  to 
different  strains  of  alcohol -prefer- 
ring rats  and  mice. 


Alcohol  and  the 
Neuroendocrine  Portfolio 

Samir  Zakhari,  Ph.D. 

In  FY97,  the  neuroendocrine  port- 
folio consisted  of  14  grants  totaling 
$2.1  million.  Except  for  1  human 
study,  all  of  the  grants  study  alcohol 
effects  on  the  endocrine  system  using 
experimental  animals.  Five  of  the  14 
grants  focus  on  the  HPA  axis,  5  on 
the  hypothalamic-pituitary-gonadal 
(HPG)  axis,  2  on  the  endocrine 
regulation  of  immune  function,  1  on 
hormonal  regulation  of  alcohol 
metabolism,  and  1  on  enkephalin 
gene  expression. 


535 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Twelve  of  the  14  grants  are  catego- 
rized as  basic  research;  the  other  2  are 
considered  applied  research.  Two  grants 
focus  on  response  to  stress,  and  all  14 
grants  study  alcohol  at  the  cellular  and 
molecular  level. 

Research  Areas 

HPA  Axis  (5 grants) 

Alcohol  and  Stress:  Interactive 
Effects.  Prenatal  alcohol  exposure 
elicited  HPA  axis  hyperresponsiveness 
and  compromised  immune  integrity  in 
adult  male  and  female  rats. 

AlcoholTnterleukin  Interactions  on 
the  HPA  Axis.  Prenatal  exposure  to 
alcohol  perturbed  HPA  responsiveness  to 
interleukin-l(3  (IL-1(3)  by  blunting  the 
ACTH  response  in  immature  (3  week) 
male  and  female  rats,  but  conversely  pro- 
duced a  potentiating  effect  when  these 
same  animals  reached  adulthood. 

Alcohol  Effects  on  Opiomelano- 
cortinergic  Regulation.  Under  non- 
stressful  conditions,  moderately  high 
blood  alcohol  levels  temporarily  activated 
the  HPA  axis,  with  concomitant  activa- 
tion of  the  forebrain  opiomelanocortin- 
ergic  neuronal  system. 

HPA  Axis  and  Alcoholism.  The 
HPA  dynamics  was  different  in  nonal- 
coholic people  with  a  family  history  of 
alcoholism  (FHP)  than  in  nonalcoholic 
subjects  without  a  family  history  of 
alcoholism  (FHN). 

Alcohol  and  Neuroendocrine  Func- 
tion: Oxytocin  Expression.  Alcohol 
inhibited  the  secretion  of  oxytocin,  which 
may  play  a  role  in  the  development  of 
tolerance  to  alcohol.  Oxytocin  secretion 
was  reduced  during  acute  intoxication, 
but  not  during  alcohol  withdrawal. 


Chronic  alcohol  administration  inhib- 
ited oxytocin  secretion  in  male  rats 
but  not  in  female  rats. 

HPG  Axis  (5 grants) 

Pubertal  Alcohol  and  Male  Repro- 
duction. Acute  alcohol  administration 
to  35-day-old  (prepubertal),  45-day- 
old  (midpubertal),  or  55-day-old  (late 
pubertal)  male  rats  caused  depression 
of  testosterone  and  luteinizing  hormone 
(LH)  levels  in  the  two  older  groups; 
coadministered  naltrexone  reversed  the 
testosterone  but  not  the  LH  depres- 
sion. Chronic  alcohol  administration 
to  male  rats  (45  and  55  days  old)  that 
were  subcutaneously  implanted  with  a 
pellet  of  naltrexone  2  days  before 
being  offered  a  liquid  diet  containing 
alcohol  (36  percent  of  total  calories) 
for  14  days  showed  similar  effects. 

Alcohol  Testicular  Effects.  Alcohol 
decreased  testosterone  secretion  and 
testicular  interstitial  fluid  formation  in 
rats.  This  effect  was  not  mediated  by 
endogenous  opioids  or  nitric  oxide. 

Alcohol  and  Female  Rodent  Repro- 
duction. A  single  dose  of  alcohol 
given  to  female  rats  nearly  obliterated 
serum  proestrum  LH  levels,  and 
serum  estradiol  and  progesterone  lev- 
els fell  to  half  of  the  control  values. 

Neuroendocrine  Effects  of  Alcohol 
on  Puberty.  In  vivo  and  in  vitro  inves- 
tigations have  demonstrated  that  insulin- 
like growth  factor- 1  (IGF-1)  is  crucial  to 
the  onset  of  puberty  in  females,  and 
that  alcohol  impedes  the  physiological 
responses  of  the  brain  and  ovary  to  IGF- 
1.  Alcohol-induced  depression  of  LH 
release  arises  from  a  decrease  in  pro- 
staglandin E2  formation.  Leptin  also 
induces  the  prepubertal  release  of  LH; 


536 


Subcommittee  Report 


peripherally  administered  leptin  reverses 
the  depressed  LH  secretion  by  alcohol. 
Alcohol  also  perturbs  the  nitric  oxide/ 
nitric  oxide  synthase  system  at  specific 
phases  of  the  reproductive  cycle  dur- 
ing puberty,  consistent  with  a  role  in 
ovulation  and  luteal  formation. 

Alcohol  and  Hyperprolactinemia. 
The  human  disorder  gynecomastia, 
observed  in  some  alcoholic  men,  which 
arises  from  elevated  plasma  levels  of 
the  pituitary  prolactin  hormone  (PRL), 
is  being  studied  in  a  rat  model.  Stud- 
ies focus  on  elucidating  the  cellular 
mechanisms  (presumptively  involving 
inhibition  of  transforming  growth  fac- 
tor beta  1  [TGF-(31]  in  the  pituitary) 
of  hyperprolactinemia. 

Endocrine  Regulation  of  Immune 
Function  (Igrants) 

Immunosuppression  in  a  Binge-Drink- 
ing Model.  A  single  high  dose  of  alcohol 
in  the  mouse  produced  peak  corticos- 
terone  levels  reaching  10  times  basal 
levels.  Humoral  immune  function  was 
suppressed,  expression  of  IL-1(3,  inter- 
leukin-2,  and  interleukin-4  were  com- 
promised in  the  spleen,  and  resident 
B-cell  population  was  reduced.  RU  486 
(a  glucocorticoid  antagonist)  reversed 
the  suppressed  antibody  and  cytokine 
responses.  Exogenous  administration 
of  corticosterone  mimicked  some,  but 
not  all,  of  the  effects  produced  by  a  high 
dose  of  alcohol,  suggesting  that  addi- 
tional components  are  contributing  to 
the  immunosuppression  produced  by 
high-dose  alcohol  administration. 

Natural  Killer  (NK)  Cells  and  Binge 
Drinking.  Alcohol  suppresses  basal  and 
induced  NK  cell  activity.  The  sup- 
pressed NK  activity  arises  pardy  via  a 


glucocorticoid- based  mechanism  and 
partly  via  the  perturbation  of  the  bal- 
ance between  Thl  and  Th2  cells. 

Endocrine  Regulation  of  Alcohol 
Metabolism  (1  grant) 

Dihydrotestosterone  (DHT)  suppresses 
alcohol  dehydrogenase  (ADH)  transcrip- 
tion in  hepatocytes,  possibly  explaining 
the  higher  alcohol  elimination  rates  in 
women.  The  suppression  of  rat  liver 
ADH  activity  by  DHT  was  associated 
with  a  decrease  in  ADH  protein. 

Effects  of  Alcohol  on  Enkephalin  Gene 
Expression  (1  grant) 

The  ultimate  goal  of  this  grant  is  to  study 
the  role  of  enkephalins  in  alcohol-seeking 
behavior;  the  immediate  focus  is  on 
the  mechanism  by  which  members  of 
the  steroid-retinoid  receptor  super- 
family  modulate  expression  of  the  pre- 
proenkephalin  gene. 

Recommendations 

More  research  is  encouraged  in  the  fol- 
lowing areas:  (1)  alcohol  and  neuroim- 
munomodulation,  (2)  role  of  peptides 
in  alcohol  intake,  (3)  alcohol  and  the 
hypothalamic-pituitary-thyroid  axis,  and 
(4)  alcohol  and  growth  hormone. 

Molecular  and  Cellular 
Adaptive  Responses  to 
Chronic  Alcohol  Exposure 

Neuroadaptation:  Gaps  in  Knowledge 
and  Research  Opportunities 

Paula  L.  Hoffman,  Ph.D.,  A.  Leslie 
Morrow,  Ph.D.,  Tamara  J.  Phillips,  Ph.D., 
and  George  R.  Siggins,  Ph.D. 

•    It  is  important  to  establish  the  rela- 
tionship between  observed  changes 


537 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


and  neurochemical  and  molecular 
changes  after  chronic  alcohol  treatment 
and  the  occurrence  of  neuroadaptive 
events  such  as  tolerance  and  depen- 
dence. Studies  are  needed  that  are 
interdisciplinary  and  translational — 
studies  at  the  molecular  and  cellular, 
brain  slice,  and  whole  animal  levels, 
as  well  as  human  studies.  Examples 
include  (1)  studies  of  synaptic  neuro- 
physiology and  neural  circuitry  after 
chronic  alcohol  exposure,  using  tech- 
niques to  separate  pre-  and  postsynap- 
tic changes;  use  of  models  such  as 
slices  and  explants,  with  intact  neuro- 
physiology, and  use  of  animal  models 
to  observe  neuronal  ensemble  activity; 
(2)  studies  of  molecular  mechanisms 
of  changes  in  receptors/ion  channels 
after  chronic  alcohol  treatment,  includ- 
ing subunit  composition,  posttrans- 
lational  modification,  and  receptor 
localization;  (3)  studies  of  functional 
importance  of  changes  in  gene  expres- 
sion for  neuroadaptation,  with  a  focus 
on  gene  expression  in  neural  circuits 
that  affect  pharmacological  responses 
to  alcohol;  and  (4)  studies  of  the  inter- 
connections among  various  signal 
transduction  systems  that  influence  cell 
survival,  differentiation,  and  responses 
to  stress  and  other  external  stimuli, 
and  the  importance  of  these  systems 
for  neuroadaptation  to  alcohol. 

•  It  is  necessary  to  determine  the  rela- 
tionship between  tolerance  or  sensi- 
tization to  the  reinforcing/aversive 
effects  of  alcohol  and  alcohol  intake. 
A  corollary  is  whether  tolerance  or 
sensitization  to  the  reinforcing  effects 
of  alcohol  does,  in  fact,  develop. 

•  Increased  emphasis  should  be 
placed  on  the  application  of  simple 


(e.g.,  invertebrate)  models  that 
have  been  used  to  study  learning 
and  memory,  to  the  study  of  alcohol- 
induced  neuroadaptation  (toler- 
ance and  dependence). 

•  Neuroadaptation  after  moderate  drink- 
ing (events  taking  place  in  the  brain 
during  the  transition  from  moderate  to 
abusive  drinking)  should  be  explored. 
Biochemical  and  behavioral  studies  of 
the  effects  of  the  full  range  of  alco- 
hol doses  and  concentrations  should 
be  carried  out,  to  determine  thresh- 
olds for  neuroadaptive  effects  of 
alcohol,  and  J-  or  U-  shaped  dose- 
response  curves. 

•  Tools  are  needed  to  study  neu- 
roadaptation, including  genetic 
models  such  as  transgenics,  knock- 
outs, selected  lines,  recombinant 
inbreds,  and  congenics,  in  order 
to  test  hypotheses  of  mechanisms 
of  neuroadaptation  and  identify 
genes  involved  in  these  processes. 
Animal  models  should  also 
include  nonhuman  primates  for 
behavioral  studies. 

•  Emphasis  should  be  placed  on 
application  of  discoveries  regarding 
molecular  and  neurochemical 
mechanisms  of  neuroadaptation  to 
alcohol  to  the  treatment  and  inter- 
vention arena. 

Neurotoxicity:  Gaps  in  Knowledge 
and  Research  Opportunities 

Fulton  T.  Crews,  Ph.D. 

•  It  is  important  to  determine  the 
region-specific  neurotoxic  effects 
accompanying  extended  alcohol 
exposure  as  well  as  those  accompa- 
nying withdrawal. 


538 


Subcommittee  Report 


Although  the  loss  of  white  matter 
has  been  documented  to  accom- 
pany chronic  alcohol  exposure, 
there  has  been  relatively  little 
research  into  mechanisms. 
It  is  recommended  that  a  human 
alcoholic  brain  bank  be  established 
in  the  United  States. 
Although  women  represent  approx- 
imately 25  percent  of  alcoholics 
and  may  suffer  greater  pathology, 
they  have  been  understudied. 
Moreover,  data  regarding  the  role 
of  gender  might  provide  important 
fundamental  insights  into  mecha- 
nisms of  brain  damage. 
A  significant  gap  in  knowledge 
is  the  relationship  between 
acute  excessive  stimulation  of  neu- 
rons by  glutamate  and  delayed 
neuronal  death. 

Why  specific  brain  regions  are 
particularly  sensitive  to  chronic 
alcohol  exposure  is  an  important 
question  that  needs  to  be 
answered. 

Oxidative  stress  is  increased 
in  brain  by  alcohol  and  is  often 
postulated  to  contribute  to  brain 
damage.  There  are,  however, 
relatively  few  data  for  or  against 
this  hypothesis. 

An  important  area  of  research  is 
to  determine  the  role  alcoholic 
brain  damage  plays  in  the  progres- 
sion to  alcoholism,  recovery  from 
alcoholism,  and  other  behaviors 
associated  with  alcoholism. 
Understanding  the  relationship 
of  neuropathology  to  behavioral 
pathology  is  essential  and  funda- 
mental to  improving  prevention 
and  treatment. 


Molecular  Neuropharmacology 
Portfolio 

Robert  W.  Karp,  Ph.D. 

To  identify  the  primary  effects  of 
chronic  alcohol  exposure,  investiga- 
tors studying  neuroadaptation  and 
neurotoxicity  have  examined  alcohol- 
induced  molecular  changes  in  cellular 
components.  Some  of  these  studies  can 
be  performed  in  intact  animals,  thereby 
permitting  correlation  with  alcohol- 
induced  changes  in  systemic  neural 
function  and  behavior.  Other  studies  can 
be  performed  only  in  cultured  cells. 
Although  studies  in  cultured  cells  pro- 
vide information  about  some  molecu- 
lar processes  that  cannot  be  assayed  in 
intact  animals,  it  is  often  difficult  to 
evaluate  their  behavioral  significance. 
Moreover,  for  some  measures  that  can 
be  made  in  both  cultured  cells  and 
whole  animals,  different  changes  are 
observed  in  these  two  experimental  sys- 
tems. For  this  reason,  many  investigators 
try  to  confirm  cellular  observations  in 
intact  animals  whenever  possible. 

Research  Areas 

Table  6  shows  the  distribution  of 
molecular  neuropharmacology  awards 
by  research  area. 

Withdrawal 

NIAAA  supports  a  number  of  studies, 
most  of  them  in  intact  animals,  of 
molecular  changes  associated  with 
withdrawal  from  chronic  alcohol 
exposure.  The  largest  group  of  these 
studies  is  directed  at  GABAA  receptors, 
focusing  on  changes  in  brain -regional 
distribution  and  pharmacological 
properties  (including  interactions  with 


539 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


neurosteroids),  and  the  underlying 
causes  of  such  changes.  The  second 
largest  group  of  studies  focuses  on 
similar  issues  related  to  NMD  A  recep- 
tors. Other  studies  examine  5-HT 
receptors  and  their  downstream  sig- 
naling (especially  their  relationship  to 
anxiety  during  withdrawal),  non- 
NMDA  glutamate  receptors,  Fos-like 
immunoreactivity,  mechanism  of  up- 
regulation  of  voltage -gated  Ca++  chan- 
nels, and  free  radical  accumulation 
due  to  oxidative  stress.  Of  the  11  pro- 
jects in  this  category,  4  include  studies 
on  cultured  cells. 

Tolerance 

NIAAA  supports  studies  of  a  variety 
of  molecular  changes  induced  by 
chronic  alcohol  exposure  in  various 
types  of  neuronal  cell  lines.  Although 
the  precise  relationships  of  these  changes 
to  alcohol-induced  physiological  changes 
in  animals  are  not  yet  understood, 
they  could  potentially  be  related  to 
tolerance  or  dependence.  These  studies 
include  heterologous  desensitization 
of  signaling  from  purine  receptors, 
intracellular  translocation  of  protein 


kinase  A,  global  changes  in  gene 
expression,  and  the  normal  physiolog- 
ical role  of  a  phosducin-like  protein, 
whose  levels  are  raised  in  response  to 
chronic  alcohol  exposure. 

Neurotoxicity 

Most  of  the  studies  of  molecular 
changes  associated  with  neurotoxicity 
involve  a  combination  of  experiments 
on  both  intact  animals  and  culture 
cells.  The  largest  group  of  such  studies 
focuses  on  alcohol- induced  changes  in 
brain-regional  distribution  of  neuro- 
trophic factors  and  their  receptors,  the 
role  of  neurotrophic  factors  in  protec- 
tion from  alcohol-induced  disruption 
of  calcium  homeostasis,  and  the  mech- 
anism of  enhancement  of  neuro- 
trophin-induced  neurite  outgrowth  by 
alcohol.  Another  group  of  studies  is 
directed  at  NMDA  receptors,  focusing 
on  changes  in  brain-regional  distribu- 
tion and  pharmacological  properties, 
underlying  causes  of  such  changes, 
and  changes  in  downstream  signaling 
(especially  induction  of  nitric  oxide  syn- 
thase). Two  studies  are  concerned  with 
alcohol's  enhancement  of  pro-oxidant- 


Table  6.  NIAAA  FY97  Awards 

in  Molecular  Neuropharmacology. 

Study  Area 

Number 

Dollar  Amount 

Withdrawal 

11 

1,694,000 

GABA 

5 

871,000 

NMDA 

2 

283,000 

Other 

4 

540,000 

Tolerance 

5 

1,351,000 

Neurotoxicity 

9 

1,761,000 

Neurotrophins 

3 

706,000 

NMDA 

3 

532,000 

Oxidative  Stress 

2 

356,000 

Other 

1 

167,000 

Total 

25 

4,806,000 

540 


Subcommittee  Report 


induced  membrane  lipid  peroxidation 
and  changes  in  gene  expression.  Finally, 
individual  projects  study,  variously,  the 
mechanisms  of  alcohol-induced  changes 
in  the  levels  and/or  activities  of  the 
Na-Ca  exchanger  and  metabotropic 
glutamate  receptors. 

Recommendations 

The  projects  described  above  study 
molecular  changes  induced  by  alcohol 
in  intact  animals  and/or  cultured 
cells.  Alcohol-induced  molecular 
changes  can  be  correlated  with  behav- 
ioral and  physiological  changes  in  ani- 
mals, whereas  molecular  changes  in 
cultured  cells  cannot  be  directly  corre- 
lated. Investigators  typically  hypothe- 
size that  the  molecular  changes  they 
observe  actually  mediate  particular 
behavioral  and  physiological  changes. 
Since  chronic  alcohol  exposure  induces 
a  multitude  of  molecular,  physiological, 
and  behavioral  changes,  correlative 
evidence  is  insufficient  to  prove  that  a 
particular  molecular  change  mediates 
a  particular  physiological  or  behavioral 
change.  Emphasis  should  be  placed  on 
experiments  in  which  an  investigator 
specifically  blocks  either  the  alcohol- 
induced  molecular  change  under 
study,  or  the  function  of  the  molecule 
whose  disposition  is  changed  by  alco- 
hol, and  then  observes  whether  this 
intervention  also  blocks  the  behavioral 
change  under  study.  Alternatively,  the 
investigator  can  induce  the  molecular 
change  by  some  means  other  than  alco- 
hol treatment  and  observe  whether 
the  behavioral  or  physiological  change 
still  occurs.  Only  about  one-fifth  of 
the  projects  described  above  attempt 
to  perform  such  an  intervention. 


ADDICTION  AND  OTHER 
BEHAVIORS  IN  ANIMAL 
MODELS 

Basic  Behavioral  Effects  and 
Underlying  Neurocircuitries 
of  Alcohol:  Gaps  in  Knowledge 
and  Research  Opportunities 

Kathleen  A.  Grant,  Ph.D. 

•  The  initiation  of  alcohol  seeking 
needs  additional  exploration  with 
new  paradigms  using  animal  models. 
There  is  also  a  lack  of  studies  inves- 
tigating the  possibility  of  protecting 
the  individual  from  the  develop- 
ment of  alcohol-seeking  behavior. 

•  Additional  studies  are  needed  to 
characterize  the  interrelationships 
among  oral  self-administration  of 
alcohol  and  preference  condition- 
ing studies. 

•  Studies  are  needed  to  understand 
the  apparent  differences  between 
rats  and  mice  in  sensitivity  to  alco- 
hol's rewarding  effects  in  the  place 
conditioning  paradigm. 

•  Sophisticated  neuroscientific 
procedures  should  be  combined 
with  sophisticated  behavioral 
procedures. 

•  As  candidate  genes  become  identified 
and  gene  products  known,  there  will 
be  a  need  to  have  ligand  development 
to  target  potential  sites  of  action  in 
rats,  monkeys,  and  humans. 

•  The  use  of  monkey  models  for  non- 
invasive imaging  procedures  related 
to  alcohol  abuse  should  be  increased. 
Monkeys  are  important  models 
because  of  their  compliance  in  drink- 
ing excessive  quantities  of  alcohol, 


541 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


neuroanatomy,  brain  size,  complex 
behavior,  neuroendocrine  system, 
and  longevity. 

•  Cloning  techniques  of  nuclear  trans- 
plantation from  adult  monkey  cells 
should  at  least  be  given  thoughtful 
consideration.  Such  approaches 
could  be  used  to  address  the 
genetic  basis  of  complex  behavioral 
responses  associated  with  the 
development  of  alcohol  abuse  and 
alcoholism. 

•  Gender  differences  in  the  behav- 
ioral neuroscience  of  alcohol  using 
animal  models  are  understudied. 

•  Understanding  the  role  of  stress  in 
alcohol's  behavioral  effects  requires 
more  sophisticated  approaches. 

•  Depression  is  a  risk  factor  that  ani- 
mal models  in  alcohol  abuse  have 
not  addressed  extensively. 

•  The  age  at  which  individuals  start 
regular,  heavy  use  of  alcohol  has 
recently  been  reported  to  predict 
the  occurrence  of  alcohol  depen- 
dence. The  macaque  monkey  has  at 
least  a  12-month  adolescent  phase, 
which  allows  a  window  of  opportu- 
nity to  design  appropriate  experi- 
mental manipulations. 

•  The  concurrent  use  of  alcohol  and 
other  drugs  of  abuse  has  received 
limited  attention. 

Neuroadaptive  Changes  in 
Neurotransmitter  Systems 
Mediating  Alcohol-Induced 
Behaviors:  Gaps  in  Knowledge 
and  Research  Opportunities 

Friedbert  Weiss,  Ph.D. 

•  A  systematic  research  effort  at  the 
behavioral,  neurochemical,  cellular, 


and  molecular  levels  will  be  needed 
to  identify  and  characterize  neu- 
roadaptive changes  and  homeosta- 
tic  disturbances  during  protracted 
withdrawal,  and  to  determine  their 
motivational  significance  in  appro- 
priate models  of  alcohol-seeking 
behavior  and  relapse. . 

•  It  is  essential  to  develop  new,  or 
modify  existing,  animal  models  of 
self- administration  suitable  for  the 
longitudinal  monitoring  of  neu- 
roadaptive or  homeostatic  changes 
during  the  development  of  chronic 
alcohol  drinking  and  over  the 
course  of  withdrawal  and  interven- 
ing periods  of  abstinence. 

•  Effective  procedures  or  models  are 
needed  that  permit  investigation  of 
behavioral  plasticity  such  as  the 
development  of  associations 
between  alcohol's  subjective 
rewarding  effects  and  relevant  envi- 
ronmental stimuli  that  may  trigger 
relapse  during  abstinence. 

•  It  is  important  to  examine  the  role  of 
stressful  stimuli  on  the  reinstatement 
of  alcohol-seeking  behavior  at  differ- 
ent stages  of  the  protracted  abstinence 
phase  and  to  determine  whether 
vulnerability  to  relapse  becomes 
exacerbated  with  repeated  with- 
drawal and  abstinence  episodes. 

•  Research  strategies  need  to  be  devel- 
oped to  identify  neural  or  molecu- 
lar mechanisms  mediating  the 
switch  or  transition  from  nonde- 
pendent  social  drinking  to  a  state 
of  dependence. 

•  There  is  a  need  to  better  model 
various  aspects  of  alcoholism  in 
laboratory  animals.  This  includes, 
in  particular,  voluntary  drinking 


542 


Subcommittee  Report 


models  that  promote  spontaneous 
and  persistent  intake  of  high 
alcohol  concentrations  or  volumes 
without  prior  need  to  induce 
dependence.  Such  models  will 
represent  an  important  step  toward 
the  need  for  studying  critical 
issues  such  as  the  mechanisms 
underlying  the  switch  from  nonde- 
pendence  to  dependence,  and 
the  reinforcement  contingencies 
that  maintain  alcohol  consumption 
in  dependent  individuals. 
There  is  a  need  to  study  neuro- 
transmitter circuitries  and  interac- 
tions mediating  alcohol  reward 
in  the  dependent  and  postdepen- 
dent  state.  Although  there  is 
increasing  evidence  that  the  acute 
reinforcing  actions  of  alcohol 
depend  on  multiple  neurochemical 
systems  and  their  interactions,  lit- 
tle if  anything  is  known  about 
these  mechanisms  in  dependent 
individuals.  In  this  context,  it  will 
also  be  beneficial  to  incorporate 
multiple  systems  approaches  in 
medication  development  efforts 
and  to  examine  the  therapeutic 
efficacy  of  combinations  of  rele- 
vant pharmacological  agents. 
It  is  important  to  clarify  the  role  of 
dopamine  in  alcohol  reinforcement. 
This  need  involves  both  a  better 
understanding  of  mechanistic  ques- 
tions such  as,  for  example,  how  alco- 
hol activates  mesolimbic  dopamine 
transmission  and  a  better  under- 
standing of  the  precise  role  of 
dopamine  in  various  aspects  of 
alcohol-seeking  behavior. 
An  important  emerging  issue  is  the 
role  of  sensitization  in  alcohol 


reinforcement,  genetic  preference, 
and  dependence.  In  particular,  the 
following  questions  will  require 
clarification:  (1)  does  alcohol  sen- 
sitization augment  the  reinforcing 
efficacy  or  potency  of  alcohol;  (2) 
does  alcohol  sensitization  promote 
a  heightened  motivational  state 
with  increased  alcohol-seeking 
behavior  ("craving")  without  nec- 
essarily altering  the  reinforcing  effi- 
cacy of  alcohol;  (3)  is  alcohol 
sensitization  a  correlate  of  aversive 
or  side  effects  of  repeated  alcohol 
intoxication;  and  (4)  if  so,  is  alco- 
hol sensitization  negatively  linked 
with  alcohol  preference  or  vulnera- 
bility to  abuse? 

It  is  important  to  better  under- 
stand the  mechanisms  of  kindling 
or  sensitization  of  withdrawal 
seizures  at  the  molecular,  cellular, 
and  biochemical  levels.  This 
includes  efforts  to  define  sensitiza- 
tion of  psychological  components 
of  withdrawal  (e.g.,  anxiety,  affec- 
tive changes),  to  characterize  pos- 
sible changes  in  the  subjective 
perception  of  alcohol's  intoxicating 
actions  (i.e.,  alcohol's  discrimina- 
tive stimulus  effects),  to  determine 
whether  multiple  alcohol  with- 
drawal experiences  alter  the  rein- 
forcing properties  of  alcohol,  and 
to  examine  potential  changes  in 
susceptibility  to  alcohol  neurotoxi- 
city and  associated  cognitive 
impairments.  Finally,  it  is  impor- 
tant to  examine  whether  condition- 
ing factors  contribute  to  the 
kindling  phenomenon. 
The  field  would  benefit  from  the 
development  of  genetic  models  of 


543 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


alcohol-seeking  behavior  selected 
on  the  basis  of  motivational 
measures  such  as  operant  respond- 
ing or  even  progressive-ratio  per- 
formance during  dependence. 

Adolescent  Period:  Gaps  in 
Knowledge  and  Research 
Opportunities 

Linda  P.  Spear,  Ph.D. 

•  Further  research  is  needed  to  exam- 
ine how  sensitivity  to  various  effects 
of  alcohol  differs  in  adolescents  from 
other  aged  organisms,  and  to  explore 
the  neural  mechanisms  underlying 
these  ontogenetic  effects. 

•  Research  to  examine  the  reinforc- 
ing efficacy  of  alcohol  and  other 
reinforcers  during  adolescence  is 
particularly  critical,  given  evidence 
for  alterations  during  adolescence 
in  forebrain  regions  modulating 
the  reward  efficacy  of  reinforcing 
drugs. 

•  Stress  effects  on  alcohol  self- admin- 
istration during  adolescence  should 
be  examined,  along  with  funda- 
mental research  examining  the  rela- 
tionships among  hormonal, 
behavioral,  and  neural  responses  to 
stressors  throughout  ontogeny. 


•  In  addition  to  further  research 
examining  normal  brain  function  in 
adolescence,  research  is  needed  to 
determine  factors  that  trigger  onto- 
genetic changes  in  brain  function, 
alcohol  sensitivity,  and  responsivity 
to  stressors  during  the  adolescent 
period. 

•  It  is  critical  to  determine  whether 
early  exposure  to  alcohol  increases 
the  likelihood  for  later  alcohol 
problems,  and  if  so,  why  such  early 
exposure  should  be  so  predictive. 

Portfolio  of  Studies  of  the 
Addiction  Process  and  Other 
Behaviors  in  Animal  Models 

Ellen  D.  Witt,  Ph.D. 

Animal  studies,  representing  about  $6 
million,  are  focused  largely  on  neural 
mechanisms  of  sensitization,  tolerance, 
dependence,  withdrawal,  relapse,  and 
more  recently  the  reinforcing  and 
hedonic  effects  of  alcohol  that  can  lead 
to  alcohol-seeking  behavior  and  exces- 
sive drinking.  Animal  models  have 
also  been  constructed  to  study  innate 
neural  and  behavioral  traits  as  well  as 
the  acute  and  chronic  effects  of  alcohol 
on  behaviors  such  as  learning,  memory, 
and  aggression. 


Table  7.  NIAAA  FY97  Animal  Portfolio. 

Category 

Number 

Dollar  Amount 

Basic  behavioral  models/nondependent 

24 

3,850,412 

Neuroadaptive  models/dependence 

4 

630,790 

Basic  behavioral  and  dependence  models 

4 

798,483 

Adolescent  models 

4 

520,924 

Methodology  development 

2 

182,256 

Total 

38 

5,982,865 

544 


Subcommittee  Report 


The  animal  portfolio  has  been 
divided  conceptually  into  five  broad 
scientific  categories,  as  shown  in  table 
7.  Each  broad  category  is  further  sub- 
divided to  provide  a  more  detailed 
picture  of  the  number  and  types  of 
animal  models  being  studied.  This 
breakdown  is  based  on  the  specific 
aims  of  each  grant.  Since  a  single 
grant  may  be  investigating  more  than 
one  paradigm  or  neurotransmitter  sys- 
tem, a  grant  could  be  counted  more 
than  one  time  in  this  analysis.  There- 
fore, the  number  of  grants  (not  dol- 
lars) is  indicated  for  each  area  of  study 
described  in  the  text  below. 

Basic  Behavioral  Effects  and 
Underlying  Neural  Circuitry  With 
Limited  Alcohol  Exposure  (Low  to 
Moderate/Nondependent ) 

Research  Areas 

Alcohol-Seeking  Behavior/Hedonic 
Effects,  Neural  Circuitry, 
Neurotransmitters 

Self- Administration:  Operant  Rein- 
forcement, Two-Bottle  Choice  (7 
grants).  These  grants  are  investigating 
the  neural  circuitry  and  neurotrans- 
mitter pathways  underlying  excessive 
alcohol  drinking  using  either  operant 
self- administration  paradigms  or  two- 
bottle  choice.  All  seven  grants  use 
microinjection  and/or  microdialysis 
techniques  to  look  at  the  regulation  of 
oral  self-administration  by  various 
neurotransmitter  systems  in  specific 
regions  of  the  cortico-mesolimbic 
reward  pathway. 

Drug  Discrimination  (4  grants). 
These  grants  are  investigating  multi- 


ple receptor  mechanisms  that  mediate 
the  discriminative  stimulus  effects  of 
alcohol — that  is,  the  internal  subjective 
effects  that  are  reinforcing  and  main- 
tain drinking. 

Brain  Stimulation  Reward  (2 
grants).  Reductions  in  brain  stimulation 
reward  thresholds  are  used  to  identify 
brain  areas  that  mediate  reinforcement 
after  drug  administration,  using  selected 
lines  of  alcohol-preferring  rats. 

Plus  Maze  (2  grants).  The  plus  maze 
is  used  to  measure  alcohol's  anxiolytic 
effects  in  selected  lines  of  alcohol- 
preferring  and  -nonpreferring  rats. 

Place  Preference,  Taste  Aversion  (3 
grants).  These  grants  are  studying  the 
neural  circuits  and/or  neurotransmit- 
ter systems  mediating  the  hedonic  or 
aversive  properties  of  initial  alcohol 
exposure  using  place  and  taste  condi- 
tioning paradigms. 

Tolerance/Acute  Withdrawal 

Place  Preference  /Aversion,  Operant 
Self- Administration,  Drug  Discrimina- 
tion (2  grants).  One  project  is  investi- 
gating whether  alcohol's  aversive 
effects  are  reduced  and  its  hedonic 
effects  are  enhanced  as  tolerance 
develops  to  alcohol's  hypothermic  effects. 
A  second  project  is  investigating  acute 
withdrawal  12-30  hours  after  a  single 
high  dose  of  alcohol,  referred  to  as 
"ethanol  delayed  effect"  (EDE)  or 
"hangover,"  and  determining  whether 
physiological  and  subjective  effects  of 
EDE  are  similar  to  phase  shift  effects 
associated  with  jet  lag  or  work  shifts. 

Environmental  Factors,  Taste,  Gender 

Operant  Self-Administration  (6 
grants).  Many  social  and  environmental 


545 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


factors  such  as  stress  (social  status), 
schedules  of  reinforcement,  taste  factors, 
and  temperature  influence  the  estab- 
lishment of  alcohol-seeking  behavior. 
Four  studies  are  investigating  environ- 
mental factors  in  an  operant  reinforce- 
ment paradigm  to  evaluate  reinforcing 
efficacy  of  alcohol  alone  or  multiple 
reinforcers  that  differ  in  taste  (alcohol 
vs.  sucrose  or  alcohol  vs.  alcohol/ 
sucrose).  Two  grants  are  examining 
gender  differences,  hormonal  regulation, 
and  effects  of  social  status  on  operant 
self- administration  of  alcohol.  One 
study  is  investigating  the  role  of  ambient 
temperature  in  altering  alcohol's  hedonic 
effects  as  measured  by  taste  and  place 
conditioning  and  self- administration. 

Conditioned  Stimuli,  Incentive 
Motivation  (3  grants).  Previously  neutral 
environmental  stimuli  can  acquire  moti- 
vational properties  of  a  primary  reinforcer 
(alcohol)  that  contribute  to  alcohol  seek- 
ing. These  grants  are  examining  the  role 
of  conditioned  stimuli  (previously  neutral 
stimuli  paired  with  alcohol)  in  rein- 
stating or  maintaining  alcohol  self- 
administration  following  extinction. 

Acute/Chronic  Brain  and  Behavioral 
Effects  (4<0rants) 

These  grants  are  investigating  the 
acute  and  chronic  (nondependent) 
effects  of  alcohol  on  cognition  and 
other  behaviors,  and  their  underlying 
neuroanatomies,  neurophysiological, 
and/or  neurochemical  substrates. 

Innate  Behavioral  Characteristics 
(Disinhibition,  Challenge)  (3 grants) 

These  grants  use  selected  rat  lines  (pre- 
ferring/nonpreferring)  to  study  innate 
patterns  of  behavior  (disinhibition  and 


electrophysiological  parameters)  that  may 
predispose  to  alcoholism  and  their  under- 
lying neurobiological  mechanisms. 

Recommendations 

Behavioral  Paradigms/Phenotypes 

More  studies  are  needed  in  the  areas  of 
tolerance  and  acute  withdrawal  and  devel- 
opment of  temperament  phenotypes. 

Neurotransmitter  and  Brain  Regions 

More  grants  are  needed  to  study  addi- 
tional sites  in  the  prefrontal  cortex, 
amygdala,  hippocampus,  and  hypo- 
thalamus, and  the  interactions  of  the 
neurotransmitter  NMD  A,  5-HT,  and 
the  cholinergic  system. 

Species/Gender 

Only  four  grants  are  investigating  gen- 
der differences  as  part  of  their  specific 
aims.  Since  women  are  more  susceptible 
to  the  toxic  effects  of  alcohol  and  the 
menstrual  cycle  phase  alters  sensitivity 
of  the  subjective  effects  of  alcohol, 
more  studies  are  needed  on  the  neural 
mechanisms  of  gender  differences  in 
the  subjective  and/or  reinforcing 
effects  of  alcohol. 

Neuroadaptive  Changes  in 
Neurotransmitters  Mediating 
Alcohol-Induced  Behaviors: 
Sensitization,  Relapse,  Dependence, 
Repeated  Withdrawals 

Research  Areas 

Sensitization  (2  grants) 

Sensitization  is  the  opposite  response 
to  tolerance,  that  is,  the  effects  of  the 


546 


Subcommittee  Report 


same  dose  of  a  drug  become  more  pro- 
nounced with  repeated  exposure.  Two 
grants  are  studying  neuroadaptive 
changes  underlying  sensitization. 

Dependence,  Abstinence/Relapse  Models, 
Neural  Mechanisms  (5 grants) 

Dependence  Models:  Self-Adminis- 
tration, Brain  Stimulation  Reward, 
Incentive  Motivation  (2  grants). 
These  grants  are  examining  models 
of  dependence  produced  by  repeated 
withdrawals  from  liquid  diet  or  vapor 
inhalation.  Both  grants  use  microdialy- 
sis  and/or  microinjection  techniques. 

Prolonged  Abstinence,  Relapse, 
"Deprivation  Effect"  (3  grants). 
These  grants  are  exploring  models  of 
relapse  in  chronically  exposed  but  not 
dependent  animals.  It  has  been  shown 
that  when  abstinence  is  imposed  after 
chronic  exposure  to  alcohol,  drinking  pat- 
terns temporarily  increase.  This  has  been 
referred  to  as  the  "deprivation  effect." 

Long-Term  Effects  on  Brain 
and  Behavior  (I  grant) 

This  grant  is  investigating  the  neural 
mechanisms  of  memory  impairments 
produced  by  long-term  chronic  expo- 
sure to  alcohol. 

Neurobiological  Mechanisms  of 
Protracted  Withdrawal  (1  grant) 

This  grant  is  exploring  the  role  of  5- 
HT  in  mediating  the  symptoms  of 
alcohol  withdrawal. 

Recommendations 

Behavioral  Paradigms/Phenotypes 

More  applications  are  needed  to  develop 
models  of  dependence,  craving,  relapse, 


and  withdrawal,  as  well  as  long-term 
neurobiological  consequences. 

Neurotransmitter  Systems 
and  Brain  Areas 

Because  the  cortico-mesolimbic  reward 
system  involves  complex  interconnections 
among  many  structures  in  addition  to 
the  ventral  tegmental  area,  nucleus 
accumbens,  and  "extended"  amygdala, 
more  grants  are  needed  to  study  all  the 
relevant  transmitter  systems  and  their 
interactions  in  the  cortico-mesolimbic 
reward  system,  including  the  prefrontal 
cortex  and  hippocampus. 

Species/Gender 

In  that  females  have  been  shown  to  be 
differentially  sensitive  to  alcohol's  effects 
across  the  menstrual  cycle,  more  studies 
are  needed  on  gender  differences  in 
craving,  development  of  dependence, 
and  relapse. 

Adolescent  Period:  Biological  Basis 
for  Vulnerability  and  Underlying 
Neurobiological  Mechanisms 

Research  Areas 

Neurobiological  and  Behavioral 
Mechanisms  and  Consequences  of 
Adolescent  Drinking 

Self-Administration  and  Neuro- 
transmitter Systems  (2  grants).  These 
grants  are  investigating  whether  charac- 
teristics of  the  dopamine  and  serotonin 
systems  present  in  adults  are  also  pre- 
sent in  young  animals  and  contribute  to 
excessive  drinking  in  the  young.  Both 
grants  use  microdialysis,  high-pressure 
liquid  chromatography,  and  autoradiog- 
raphy procedures  to  determine  if  there 


547 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


are  differences  in  regional  5-HT  and 
dopamine  content,  regional  densities 
of  5-HTj  and  5-HT2  sites,  and  binding 
of  5-HT1A  and  D2  sites  in  animals  15, 
25,  and  35  days  of  age.  Brain  regions 
of  interest  are  the  cerebral  cortex,  hippo- 
campus, striatum,  hypothalamus,  accum- 
bens,  and  olfactory  tubercle.  Both  grants 
are  using  selected  lines  (P,  NP,  HAD, 
LAD)  to  compare  phenotypic  differences 
in  these  measures. 

Development  of  Tolerance /Sensitivity, 
Low- Dose  Stimulatory  Effect  (3  grants). 
These  grants  are  looking  at  the  presence 
of  and/or  ontogeny  of  alcohol  respon- 
siveness by  measures  of  low- dose  alcohol 
stimulation,  acute  and  rapid  tolerance  to 
alcohol's  sedative  effects  (sleep  time), 
hypothermia,  and  suppression  of  startle, 
as  well  as  differences  in  emotionality  and 
cognitive  functions  (plus  maze  and 
Morris  Water  Maze  learning).  Three 
grants  are  using  rodents  and  two  are 
using  selected  lines  (P,  NP,  HAD,  LAD). 

Environmental  Factors  and  Con- 
sumption (Free  Choice  and  Operant 
Responding)  (1  grant).  This  grant  is 
examining  the  ontogeny  of  drinking 
after  weaning  (day  22  of  age)  and  the 
effects  of  various  factors  on  this  devel- 
opment, such  as  housing  conditions, 
taste  aversion  conditioning,  and  phar- 
macological interventions  (fluoxetine, 
methylphenidate,  and  buspirone). 
Selected  lines  of  rats  are  used  (P,  NP, 
HAD,  LAD). 

Long-Term  Behavioral  Consequences 
of  Adolescent  Drinking  (2  grants). 
These  grants  are  studying  cognitive  and 
emotional  changes  in  adult  animals  from 
selected  lines  (P,  NP,  HAD,  LAD)  that 
were  exposed  to  alcohol  during  adoles- 
cence, such  as  orienting  to  a  novel 


environment,  plus  maze  performance, 
and  Morris  Water  Maze  performance. 
Early  Experience  and  Later  Response 
to  Alcohol  (3  grants).  These  grants  are 
investigating  the  effects  of  early  expo- 
sure to  alcohol  on  later  responsiveness 
to  alcohol.  All  grants  use  rodents,  and 
one  grant  uses  selected  lines. 

Recommendations 

Behavioral  Paradigms 

This  is  a  relatively  new  initiative  in  the 
portfolio.  There  is  now  evidence  in 
humans  that  early  alcohol  exposure 
(especially  during  periods  of  early  child- 
hood, late  childhood,  and  adolescence) 
correlates  with  the  development  of  alco- 
holism in  adulthood.  More  studies  are 
needed  to  develop  paradigms  of  alcohol 
seeking  (self- administration  and  drug 
discrimination)  during  the  adolescent 
period  to  study  the  underlying  mecha- 
nisms of  ontogeny  of  alcoholism. 

Neurotransmitters 

Only  one  study  is  examining  neurochem- 
ical markers  of  alcoholism  vulnerability 
in  selected  lines  of  alcohol-preferring  and 
-nonpreferring  rats.  The  neurotrans- 
mitter systems  targeted  are  dopamine 
and  5-HT.  Clearly,  studies  of  other 
neurotransmitter  systems  are  needed, 
particularly  their  role  in  the  initiation  of 
alcohol-seeking  behavior. 

Species/Gender 

All  four  grants  are  using  rats,  and  two  are 
using  selected  lines.  Although  two  are 
studying  both  male  and  females,  gender 
difference  is  not  a  specific  aim.  More 
studies  are  needed  to  examine  gender 
differences.  Furthermore,  primate 


548 


Subcommittee  Report 


models  are  ideal  because  of  their  rela- 
tively longer  adolescent  period. 

Medications  Development 

The  underlying  goal  of  NIAAA's  basic 
neuroscience  research  program  is  to 
understand  the  neurobiological  mecha- 
nisms of  alcohol's  effect  in  order  to 
develop  treatments  for  alcoholism,  partic- 
ularly medications.  However,  for  the  grants 
discussed  in  this  section,  at  least  one 
specific  aim  is  to  test  specific  compounds, 
via  systemic  injection,  for  their  ability 
to  reduce  drinking,  prevent  relapses, 
or  reduce  withdrawal  symptoms. 

Research  Areas 

Operant  Responding  and  Free-Choice 
Consumption  (4jjrants) 

These  studies  are  using  self- administra- 
tion paradigms  (operant  reinforcement 
paradigms  and  two-bottle  consumption) 
to  explore  the  effectiveness  of  several 
receptor  antagonists  as  potential  thera- 
peutic agents  for  reducing  alcohol  intake. 

Protracted  Withdrawal  (1  grant) 

This  grant  is  studying  the  effectiveness 
of  various  serotonergic  compounds 
(buspirone,  a  5-HT1A  partial  agonist; 
mianserin,  a  5-HT2A/2C  antagonist; 
methysergide,  a  nonselective  5-HT 
antagonist;  and  ICS  205-930,  a  5-HT3 
antagonist)  in  blocking  withdrawal 
symptoms  as  measured  by  drug  discrim- 
ination and  plus  maze  paradigms. 

Recommendations 

The  most  frequently  studied  compounds 
for  reducing  drinking  are  the  opiate 
antagonists.  Clearly,  more  candidates 
for  potential  therapeutic  agents  are 


needed,  drawing  from  all  the  relevant 
neurotransmitter  systems.  Another  rec- 
ommendation is  that  more  standardized 
testing  procedures  for  the  potential 
agents  be  developed. 

Methodology  Development  (2  grants) 

One  grant  is  establishing  procedures 
to  measure  the  effects  of  alcohol  on 
carbohydrate  metabolism  and  rates  and 
synthesis  of  amino  acid  neurotransmit- 
ter (glutamate,  GAB  A,  and  aspartate) 
in  the  brain  of  fully  intact,  conscious 
rats  using  13C  nuclear  magnetic  reso- 
nance spectroscopy.  Another  grant  is 
developing  an  ultrasensitive  mass  spec- 
trometric  procedure  for  the  analysis  of 
neurosteroids  in  tissue  samples  and 
microdialysates  of  select  brain  areas  of 
freely  moving  rats  after  acute  and 
chronic  alcohol  administration  and 
during  withdrawal. 

Recommendations 

Techniques  that  promote  an  understand- 
ing of  the  underlying  neural  circuitry  of 
the  addiction  process  and  their  relation- 
ship to  behavior  should  be  encouraged. 

STUDIES  OF  ACUTE 
AND  CHRONIC  EFFECTS 
OF  ALCOHOL  IN  HUMANS 

Studies  of  the  Acute  Effects 
of  Alcohol  on  Cognition 

AND  iMPULSIVITY/DlSINHrBrTORY 

Behavior:  Gaps  in  Knowledge 
and  Research  Opportunities 

Peter  R.  Finn,  Ph.D. 

•    Research  should  be  undertaken  on  the 
effects  of  early  exposure  to  alcohol 


549 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


(preadolescent/adolescent)  and  the 
development  of  disinhibited  traits 
using  longitudinal  designs. 
It  is  important  to  conduct  cross- 
sectional  studies  of  the  effects  of 
alcohol  challenge  on  disinhibited/ 
impulsive  behavior  (and  factors 
associated  with  such  behavior), 
using  multimethod  measurements 
to  capture  the  multidimensional 
nature  of  behavioral  disinhibition 
and  controlling  for  important 
sources  of  individual  differences 
such  as  preexisting  disinhibited 
traits,  family  history  of  alcoholism 
(and  antisocial  behavior),  drinking 
history,  and  level  and  limb  of  the 
blood  alcohol  curve. 


Clinical  Neuroscience 
Studies  of  Behaviors 
Associated  With  Alcohol 
Consumption  in  Alcoholism: 
Gaps  in  Knowledge  and 
Research  Opportunities 

John  H.  Krystal,  M.D. 

•  More  emphasis  should  be  placed 
on  developing  drugs  that  block  the 
euphoric  effects  of  alcohol. 

•  NIAAA  should  assist  investigators 
in  gaining  access  to  alcohol-related 
investigational  new  drugs. 

•  Increased  emphasis  should  be 
placed  on  studying  the  neurobiol- 
ogy of  triggers  for  relapse  to  alco- 
hol use,  particularly  alcohol-related 
cues;  priming  effects  of  alcohol 
consumption  on  subsequent  drink- 
ing; and  the  interactive  effects  of 
stress  (negative  mood  induction) 
and  alcohol  cues. 


•  Alcohol  research  should  more  fully 
integrate  functional  neuroimaging 
techniques  to  facilitate  the  study  of 
neural  circuitry  underlying  craving 
and  factors  related  to  alcohol  con- 
sumption. 

HPA  Axis:  Changes  and  Risk 
for  Alcoholism:  Gaps  in 
Knowledge  and  Research 
Opportunities 

Gary  Wand,  M.D. 

•  It  is  important  to  better  understand 
the  relationships  between  specific 
neurotransmitter  input  to  hypothal- 
amic CRH  neurons  as  a  function  of 
family  history  of  alcoholism  and  to 
estimate  their  relevance  to  vulnerabil- 
ity for  alcoholism.  More  specifically, 
dose -response  neuropharmacological 
studies  are  needed  that  activate  or 
block  5-HT,  GABA,  and  opioid 
input.  It  is  important  that  physio- 
logical and  behavioral  responses  be 
examined  in  these  studies. 

•  Relationships  between  HPA  and 
alcohol-seeking  behavior  should  be 
investigated.  In  rodent  models,  empha- 
sis should  be  on  relationships  among 
corticotropin-releasing  hormone 
(CRH)/cortisol  states,  mesolimbic 
neurochemistry,  and  alcohol-seeking 
behavior.  In  humans,  there  should 
be  comparisons  of  the  entire  HPA  axis 
between  alcohol-  and  nonalcohol- 
dependent  individuals  as  a  function 
of  family  history  of  alcoholism. 

•  It  would  be  useful  to  determine 
whether  (1)  the  magnitude  of  HPA 
axis  activity  during  acute  withdrawal 
predicts  relapse  rates;  (2)  supplements 


550 


Subcommittee  Report 


with  CRH,  ACTH,  and/or  glucocor- 
ticoids during  early  abstinence  decrease 
mood  disturbances  and  attenuate 
relapse;  and  (3)  effectiveness  of  opioid 
receptor  antagonists  in  treating 
alcoholism  relates  to  ability  to  stim- 
ulate HPA  axis. 

•  It  is  important  to  determine  if  indi- 
viduals at  increased  risk  for  the 
future  development  of  alcoholism 
have  enhanced  opioidergic  sensitiv- 
ity to  alcohol  exposure. 

•  The  effects  of  stress  dampening  by 
alcohol  should  be  studied  in  a  family 
history  design  using  components  of 
the  HPA  axis  as  end-point  measures. 

•  Future  studies  should  more  clearly  de- 
fine the  relationships  between  alcohol- 
associated  alterations  to  HPA  axis 
function  and  immune  system  function. 

Acute  and  Chronic  Effects 
of  Alcohol  on  Human  Sleep: 
Gaps  in  Knowledge  and 
Research  Opportunities 

Cindy  L.  Ehlers,  Ph.D. 

•  There  is  a  clear  need  to  increase  fund- 
ing in  the  area  of  alcohol  and  sleep. 

•  It  is  important  to  determine  if  chronic 
alcohol  intake  produces  prolonged 


sleep  disturbances  and  increased 
risk  for  relapse. 

It  is  not  known  what  brain  mecha- 
nisms underlie  alcohol-induced 
sleep  disturbances. 
It  would  be  valuable  to  determine 
the  risk  factors  for  sleep  disturbance 
and  excessive  alcohol  consumption. 
It  would  be  useful  to  investigate 
gender-related,  alcohol-associated 
sleep  disturbances. 
It  is  important  to  determine  if  new 
therapies  that  target  sleep  distur- 
bances can  be  effective  in  prevent- 
ing relapse. 


Portfolio  of  Studies  of  Acute  and 
Chronic  Effects  of  Alcohol  in 

Humans 

Ellen  D.  Witt,  Ph.D. 

In  the  scientific  content  analysis  of 
the  portfolio  on  the  acute  and 
chronic  effects  of  alcohol  in  humans 
described  below,  each  broad  scientific 
category  is  further  subdivided  to 
give  more  detail  on  the  number  and 
types  of  human  paradigms  being 
studied.  Table  8  shows  the  number 
and  dollar  value  of  grants  for  these 
human  studies. 


Table  8.  NIAAA  FY97  Grants  for  Studies  of  Acute  and  Chronic  Effects  of  Alcohol  in  Humans. 


Category 


Number 


Dollar  Amount 


Acute  cognitive  and  behavioral  effects 
Mechanisms  of  alcoholic  behavior 
Sleep 
Total 


348,967 

550,772 

202,466 

1,102,205 


551 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Acute  Cognitive  and  Behavioral  Studies 

Research  Areas 

Acute  Effects  of  Alcohol  on  Stress- 
Dampening  Response/ Aggressive 
Behavior  (Igrants) 

These  grants  are  studying  the  acute 
behavioral  effects  of  alcohol.  One  grant 
is  examining  the  effects  of  alcohol  on 
aggressive  responding  in  women  in  a 
laboratory  setting.  The  other  grant  is 
examining  the  ability  of  alcohol  to 
reduce  stress/anxiety  using  "stress 
dampening"  techniques  (i.e.,  alcohol's 
effects  on  the  magnitude  of  physiolog- 
ical responses)  and  a  "cognitive  appraisal" 
model  of  stress  and  emotion. 

Innate  Behaviors:  Antisocial 
Personality,  Behavioral  Activation/ 
Inhibition/ Aggression  (2  grants) 

These  grants  are  investigating  the  rela- 
tionship between  innate  characteristics, 
such  as  antisocial  personality  and/or 
temperament  (behavioral  activation 
and  inhibition),  and  alcohol-related 
behaviors  (i.e,  consumption  and/or 
alcohol-induced  aggression).  One 
grant  is  investigating  the  relationship 
between  antisocial  personality,  alco- 
hol-induced aggression,  and  serotonin 
function.  The  other  grant  is  investi- 
gating the  biobehavioral  mechanisms 
(i.e.,  strength  of  the  behavioral  inhibi- 
tion and  activation  systems)  mediating 
the  relationship  between  impulsive 
personality,  disinhibited/antisocial 
personality,  and  alcohol  abuse. 

Recommendations 

There  is  a  long  history  of  research 
on  the  acute  effects  of  alcohol  on  cogni- 


tive processes,  but  these  studies  have 
largely  focused  on  memory,  divided 
attention,  reaction  time,  and  visuomo- 
tor  skills.  The  currently  funded 
research  is  investigating  how  alcohol's 
effects  on  cognitive  processes  may 
have  a  role  in  increasing  drinking  or 
other  alcohol-related  behaviors,  such 
as  aggression.  More  research  is  need- 
ed in  other  areas,  such  as  alcohol's 
effects  on  executive  functioning,  a  cog- 
nitive construct  involving  the  cogni- 
tive regulation  of  behavior,  and  how 
that  may  increase  drinking.  More 
research  is  also  needed  on  the  role  of 
temperament  and/or  personality  char- 
acteristics in  contributing  to  excessive 
drinking  and  other  alcohol-related 
behaviors. 

Mechanisms  of  Alcoholic  Behaviors 

Research  Areas 

Neurotransmitter  and  Neurohormonal 
Mechanisms:  Behavioral 
Psychopharmacology  (3 grants) 

These  grants  are  examining  the  role 
of  neurotransmitters  and  neuropep- 
tides in  alcohol  reinforcement  and 
craving  in  alcoholics.  One  grant  is 
studying  the  interactive  contributions 
of  noradrenergic  and  serotonergic  sys- 
tems to  alcohol  craving  in  detoxified 
alcoholics  using  cue  reactivity  para- 
digms. A  second  grant  is  evaluating 
the  role  of  a  glutamate  antagonist 
(ketamine)  at  the  NMDA  receptor 
in  producing  subjective  effects  similar 
to  alcohol.  A  third  grant  is  study- 
ing opioidergic  mechanisms  of  rein- 
forcement for  coexisting  alcohol  and 
nicotine  use. 


552 


Subcommittee  Report 


Neurotransmitter  Mechanisms: 
Imaging  Studies  (1  grant) 

This  grant  is  studying  whether  the 
dopamine  system  is  abnormal  in  alco- 
holics, the  functional  consequences  of 
these  abnormalities,  and  the  effects  of 
detoxification.  Using  positron  emission 
tomography  (PET)  imaging  and  mul- 
tiple tracer  methods,  this  grant  is  eval- 
uating the  dopamine  system  in 
alcoholics  in  two  phases:  the  dopamine 
system  at  rest  and  during  pharmaco- 
logical activation. 

Recommendations 

While  many  animal  studies  are  investigat- 
ing neural  circuitry  and  neurochemical 
mechanisms  of  alcohol- motivated  behav- 
iors, such  as  reinforcement,  few  studies 
are  exploring  neurochemical  mecha- 
nisms in  humans.  With  the  advent  of  new 
imaging  technologies  such  as  PET,  and 
the  discovery  of  ligands  and  receptor  ago- 
nists and  antagonists,  more  studies  are 
needed  to  investigate  neural  mecha- 
nisms of  alcohol-motivated  behaviors 
in  humans. 

Effects  of  Alcohol 
on  Sleep  (1  grant) 

This  grant  is  investigating  whether  dis- 
ruption of  sleep  continuity  and/or  loss 
of  slow  wave  sleep  leads  to  greater 
elevations  in  sympathetic  nervous  sys- 
tem activity  in  African  American  alco- 
holics, which  in  turn  results  in  impaired 
immune  function  and  increased  risk 
for  disease. 

Recommendations 

Sleep  disturbance  is  a  common  prob- 
lem during  withdrawal  and  abstinence, 


which  could  contribute  to  relapse  to 
drinking.  More  research  is  needed  to 
understand  the  underlying  neural  mech- 
anisms of  sleep  disturbances  in  abstinent 
alcoholics,  and  whether  potential  medica- 
tions will  ameliorate  these  sleep  disorders. 


COGNITIVE/BEHAVIORAL/ 
STRUCTURAL  DEFICITS 


Neuropsychological 
Sequelae  of  Chronic 
Alcoholism:  Gaps  in 
Knowledge  and  Research 
Opportunities 

Marlene  Oscar-Berman,  Ph.D. 

•  Additional  research  is  needed  to 
clarify  the  separate  and  combined 
roles  of  malnutrition  and  alcohol 
neurotoxicity  in  brain  damage  and 
functional  impairments. 

•  Additional  studies  would  be  useful 
on  the  enhanced  susceptibility  to 
alcoholism-associated  deficits  in 
women  and  the  elderly. 

•  Future  research  should  include  lon- 
gitudinal or  retrospective  methods 
to  evaluate  the  likelihood  of  sub- 
group vulnerability. 

•  Specific  evaluation  of  nonhuman  ani- 
mal models  of  alcohol-associated  cog- 
nitive deficits  should  be  encouraged. 

•  It  is  important  to  establish  uniform 
positive  diagnostic  criteria  for  sub- 
types of  brain-impaired  alcoholics 
(e.g.,  KorsakofPs  syndrome  vs. 
alcoholic  dementia);  this  can  be 
approached  most  effectively  with 
the  aid  of  sophisticated  neurological 
and  neurobehavioral  techniques. 


553 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


•  Followup  postmortem  examination 
of  brains  of  well-studied  alcoholics 
should  be  encouraged  for  clues  about 
neurotransmitter  abnormalities,  as 
well  as  analyses  of  injury  at  the  cel- 
lular level. 

•  Particular  problems  in  need  of  future 
research  effort  include  the  following: 
neurobehavioral  and  brain  functional/ 
structural  recovery  with  abstinence; 
extent  of  multimodal  sensory  and 
perceptual  deficits;  nature  of  the 
loss  of  emotional  and  motivational 
functioning  among  subgroups  of 
alcoholics;  and  specific  contribu- 
tions of  frontal  system  dysfunction 
to  alcoholic  symptomatology. 

Human  Brain  Dysfunction 
Secondary  to  Alcohol 
Abuse:  Gaps  in  Knowledge 
and  Research  Opportunities 

George  Fein,  Ph.D. 

•  It  is  important  to  determine  which 
brain  systems  are  most  vulnerable 
to  the  morbid  effects  of  chronic 
alcohol  abuse. 

•  What  alcohol  use-related  factors 
influence  brain  morbidity  secondary 
to  alcohol  abuse,  including  nutritional 
deficiency,  number  of  episodes  of 
withdrawal,  development  of  tolerance, 
and  role  of  pattern  of  drinking? 

•  Other  factors  influencing  brain 
morbidity  secondary  to  alcohol 
abuse  should  also  be  studied, 
including  genetically  transmitted 
vulnerabilities,  gender,  age,  vari- 
ability in  brain  functional  reserve  as 
measured  by  intracranial  volume, 
adolescence,  and  comorbidity. 


What  are  the  mechanisms  involved 
in  recovery  of  the  brain  from  the 
effects  of  chronic  alcohol  abuse? 
Electrophysiological  methods  should 
continue  to  be  used  because  of  the 
advantages  over  other  neuroimaging 
techniques  in  terms  of  temporal 
resolution,  ease,  and  cost- effectiveness 
of  data  acquisition,  and  the  ability 
to  collect  data  during  the  performance 
of  complex  cognitive  tasks. 
It  is  important  to  combine  behav- 
ioral, structural,  and  functional 
measures  of  brain  function. 


Neuroimaging  Studies 

of  Brain  Vulnerability 

to  Alcoholism:  Gaps 

in  Knowledge  and  Research 

Opportunities 

Edith  V.  Sullivan,  Ph.D. 

•  Even  though  alcohol  abuse  and 
dependence  are  prevalent  in  adoles- 
cents as  well  as  older  individuals, 
alcohol's  adverse  effects  on  the 
adolescent  brain  are  unknown. 

•  It  is  unknown  whether  adequate 
nutritional  supplements  improve 
the  structural  condition  of  the 
brain  of  alcoholics. 

•  There  is  a  paucity  of  data  on  the 
effects  of  alcoholism  on  the  brains 
of  women. 

•  The  mechanisms  underlying  morpho- 
logical recovery  are  still  unknown. 

•  Increased  emphasis  should  be  placed 
on  in  vivo  metabolite  imaging 
because  of  its  possible  use  in  helping 
to  define  disease  progress,  assess  effi- 
cacy of  treatment,  and  track  alcohol- 
related  changes  across  the  lifespan. 


554 


Subcommittee  Report 


•  To  examine  the  extent  that  toler- 
ance may  be  genetically  determined, 
it  should  be  possible  to  use  mag- 
netic resonance  spectroscopy  (MRS) 
to  compare  estimates  of  nonab- 
sorbed  alcohol  in  nonalcoholics 
(low  alcohol-consuming  individu- 
als) who  have  a  positive  family  his- 
tory for  alcoholism  to  those  with  a 
negative  family  history. 

•  Although  the  functional  signifi- 
cance of  brain  volume  loss  in  terms 
of  the  commonly  observed  alco- 
holism-associated cognitive  behav- 
ioral deficits  has  been  difficult 
to  demonstrate,  functional  mag- 
netic resonance  imaging  (fMRI) 
has  the  potential  to  identify  areas 
throughout  the  brain  that  are 
activated  during  performance  of 
specific  components  of  cognitive 
operations. 

•  Diffusion  anisotropy  offers  a  poten- 
tial metric  for  assessing  the  integrity 
of  white  matter  throughout  the 
course  of  alcoholism. 

•  It  would  be  useful  to  examine 
brain  function  and  psychiatric 
comorbidity  in  alcoholics  in  a  sys- 
tematic manner. 

•  It  is  important  to  determine  whether, 
with  abstinence,  alcoholics  whose 
brains  show  recovery  of  tissue  vol- 
ume are  the  individuals  who  also 
show  improvement  in  their  cogni- 
tive and  motor  abilities. 

•  It  needs  to  be  established  whether 
alcoholism-associated  brain  volume 
deficits  are  accompanied  by  predictable 
neuropsychological  performance. 

•  The  question  of  gene -environment 
interaction  with  respect  to  apolipo- 
protein  genotype  and  alcohol  in 


the  development  of  dementia  and 
accelerated  brain  volume  loss  in 
alcoholics  has  yet  to  be  addressed. 

•  The  extent  to  which  the  frontal 
lobes  and  cerebellum  recover  in 
structure  or  function  remains 
unknown  despite  their  importance 
to  problem  solving,  contextual 
memory,  and  execution  and  learn- 
ing of  procedures. 

•  Controlled  studies  of  alcoholics  who 
remain  abstinent  for  years  in  com- 
parison with  those  who  drink  for 
years  are  needed. 

•  No  quantitative  in  vivo  studies  have 
been  conducted  to  examine,  even 
cross-sectionally,  potential  alcohol- 
gene  interactions  on  brain  morphol- 
ogy. These  studies  are  essential  to 
determine  the  pattern  and  extent  of 
potentially  preexisting  differences  in 
individuals  genetically  predisposed 
to  alcohol  addiction  compared  with 
those  not  so  disposed. 

Portfolio  of  Studies  on  Cognitive, 
Behavioral,  and  Structural  Deficits 
in  Humans 

Ellen  D.  Witt,  Ph.D. 

Human  studies  investigate  the  conse- 
quences of  acute  and  chronic  alcohol 
on  cognition  and  other  behaviors,  as 
well  as  the  underlying  structural 
changes  associated  with  the  behavioral 
deficits  using  state-of-the-art  imaging 
technologies.  The  neural  mechanisms 
of  alcohol-motivated  behaviors,  such  as 
craving,  have  also  been  studied  in 
humans,  but  to  a  lesser  extent.  Table  9 
shows  the  number  and  dollar  value  of 
grants  for  studies  of  cognitive,  behav- 
ioral, and  structural  deficits  in  humans. 


555 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


Brain  Damage,  Cognitive/Motor 
Dysfunction:  Affected  Brain  Areas, 
Contributions  of  Malnutrition, 
Influences  of  Polysubstance  Abuse, 
Aging,  Gender 

Research  Areas 

Neurobehavioral/Cognitive/Motor 
Dysfunction  (8 grants) 

These  grants  are  investigating  cogni- 
tive and  motor  deficits  and  their  un- 
derlying neural  substrates  in  chronic 
alcoholics  and  Korsakoff  patients,  us- 
ing neuropsychological,  neurocogni- 
tive,  neurophysiological,  and/or 
magnetic  resonance  imaging  (MRI) 
techniques. 

Motor  Functioning  (2  grants). 
These  grants  are  targeting  alcohol's 
effects  on  the  cerebellum.  One  grant 
uses  Pavlovian  techniques  (delayed 
classical  conditioning  tasks  involving 
heart  rate,  galvanic  skin  response 
(GSR)  conditioning,  eyeblink  condi- 
tioning, and  extinction)  to  dissociate 
structures  in  the  medial  temporal  lobe 
(amygdala)  and  cerebellum  in  mediat- 
ing associative  learning  deficits.  The 
other  grant  is  quantifying  regional 
volumes  of  the  cerebellar  hemispheres 
and  vermis  with  MRI,  using  a  com- 
ponent-process approach  to  assess 
motor  coordination  and  motor  skill 


learning,  and  determining  structure- 
function  relationships  between  spe- 
cific motor  processes  and  regional 
cerebellar  volumes. 

Attention,  Memory,  Executive 
Processes  (4  grants).  Two  of  these 
grants  study  attention  and/or  mem- 
ory processes  using  cognitive  tests. 
One  grant  is  studying  deficits  in  ex- 
plicit and  implicit  conceptual  memory 
in  chronic  alcoholic  and  Korsakoff  pa- 
tients. The  second  grant  is  evaluating 
cognitive  efficiency  (principally  atten- 
tion and  memory)  in  subtypes  of  al- 
coholics classified  on  the  basis  of 
other  drug  use  and  abuse  (marijuana, 
stimulants). 

Two  other  grants  are  studying 
attention,  memory,  and  executive 
functioning  in  conjunction  with 
event-related  potential  (ERP)  and/or 
fMRI  to  understand  the  underlying 
neural  mechanisms  of  these  deficits  in 
alcoholics.  One  of  these  grants  uses 
fMRI  to  study  localized  brain  activa- 
tion during  performance  of  auditory 
and  visual  working  memory  tasks. 
The  other  grant  uses  ERP  and  elec- 
troencephalographic  techniques 
to  study  attention  and  memory 
processes  in  abstinent  alcoholics 
classified  along  several  variables,  in- 
cluding type  I  and  type  II  alcoholism, 
family  history  of  alcoholism,  and 


Table  9.  NIAAA  FY97  Grants  for  Studies  of  Cognitive,  Behavioral,  and  Structural  Deficits 
in  Humans. 


Category 


Number 


Dollar  Amount 


Brain  damage/cognitive/motor  deficits 

Methodology  development 

Total 


12 

1 

13 


3,173,813 

103,482 

3,277,295 


556 


Subcommittee  Report 


the  relationship  between  antisocial 
behavior  and  neurophysiological 
characteristics. 

Affective  (Emotional)  and 
Conative  (Intentional)  Functions  (2 
grants).  These  grants  are  investigating 
emotional  and  intentional  abnormali- 
ties in  chronic  alcoholics  (with  and 
without  Korsakoff  s  syndrome)  and 
whether  these  changes  are  mediated 
by  right  frontal  or  bilateral  frontal 
lobe  pathology. 

Brain  Metabolic  Changes  and  Tolerance 
and  Long-Term  Abstinence  (2 grants) 

One  grant  is  using  MRS,  which  al- 
lows noninvasive  quantification  in 
vivo  of  brain  metabolites,  to  investi- 
gate the  mechanism  of  alcohol- 
induced  tolerance  in  humans. 
Another  grant  is  using  MRS  to  char- 
acterize the  longitudinal  course  of 
metabolic  changes  (i.e,  the  ratio  of 
visible  choline  to  the  neuronal  marker 
N-acetylaspartate)  in  the  brains  of  ab- 
stinent alcoholics. 

Effect  of  Aging  and/or  HTVon  Cognitive 
Functioning  in  Alcoholics  (5 grants) 

Alcohol  and  Aging  (4  grants).  One 
grant  is  using  structural  MRI,  electro - 
physiology,  and  neuropsychological 
assessments  to  critically  evaluate 
two  opposing  models  of  central  ner- 
vous system  effects  of  chronic  alcohol 
abuse  as  they  interact  with  age 
and  gender.  Three  grants  are  con- 
tinutions  of  ongoing  research  on 
the  interaction  between  alcohol  and 


aging.  Two  of  these  are  investigating 
emotional,  attentional,  and  inten- 
tional processes  in  alcoholics  and  age- 
matched  control  subjects  to  evaluate 
the  ways  in  which  the  behavioral  con- 
sequences of  aging  and  alcoholism  are 
parallel,  divergent,  and/or  interactive. 
Another  grant  is  a  continuation  of  an 
ongoing  longitudinal  study  of  alco- 
holic and  control  women  using  MRI, 
ERP,  and  neuropsychological  tests  to 
identify  cross-sectional  patterns  of 
sparing  and  loss,  their  interaction 
with  age,  and  comparability  to  find- 
ings in  alcoholic  men. 

Alcohol  and  HIV  (1  grant).  This 
grant  is  examining  the  effect  of 
alcohol  consumption  on  neuropsycho- 
logical function  across  stages  of  HIV 
infection. 

Recommendations 

More  research  is  needed  in  relating 
cognitive  deficits  produced  by  chronic 
alcoholism  to  excessive  drinking  and 
the  inability  to  benefit  from  treatment. 

Methodology  Development  (1  grant) 

This  grant  is  developing  a  method  to 
induce  and  maintain  a  steady-state 
concentration  of  alcohol  in  the  brain 
and  blood  while  multiple  dependent 
measures  are  assessed. 

Recommendations 

Technologies  that  improve  our  un- 
derstanding of  the  neural  mechanisms 
of  alcoholism  and  improve  treatment 
are  encouraged. 


557 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


APPENDIX  A: 
SUBCOMMITTEE  FOR 
REVIEW  OF 
NEUROSCIENCE  AND 
BEHAVIOR  PORTFOLIO 

Co -Chairs 

Henri  Begleiter,  M.D.,  Ph.D. 
Department  of  Psychiatry 
Health  Science  Center  at  Brooklyn 
450  Clarkson  Ave.,  Box  1203 
Brooklyn,  NY  11203-2098 

Catherine  Rivier,  Ph.D. 
The  Salk  Institute 
10010  North  Torrey  Pines  Rd. 
La  Jolla,  CA  92037-1099 


Experts  in  Alcohol- 
Related  Areas 

Ivan  Diamond,  M.D.,  Ph.D. 

Department  of  Neurology/Gallo  Center 

University  of  California,  San  Francisco 

San  Francisco  General  Hospital 

Bldg.  1,  Room  101 

1001  Potrero  Ave. 

San  Francisco,  CA  94110-3594 

Adron  Harris,  Ph.D. 

Department  of  Pharmacology,  C-236 

University  of  Colorado  HSC 

4200  East  Ninth  Ave. 

Denver,  CO  80262-0236 

Harold  Kalant,  M.D.,  Ph.D. 
Department  of  Pharmacology 
University  of  Toronto 
Medical  Science  Bldg. 
Toronto,  ON 
Canada  M5S  1A8 

George  Koob,  Ph.D. 

Department  of  Neuropharmacology 

The  Scripps  Research  Institute,  CVN-7 

10550  North  Torrey  Pines  Rd. 

La  Jolla,  CA  92037 


Ting-Kai  Li,  M.D. 

Indiana  University  School  of  Medicine 

Emerson  Hall  421 

545  Barnhill  Dr. 

Indianapolis,  IN  46202-5124 

Elias  Michaelis,  M.D.,  Ph.D. 
Department  of  Pharmacology  and 

Toxicology 
Malott  Hall 
University  of  Kansas 
Lawrence,  KS  66045-2505 

Oscar  Parsons,  Ph.D. 

University  of  Oklahoma  HSC 

Rogers  Bldg.,  Suite  410 

800  N.E.  15th  St. 

Oklahoma  City,  OK  73104-1602 

Adolf  Pfefferbaum,  M.D. 
SRI  International 
333  Ravenswood  Ave. 
Menlo  Park,  CA  94025 

Bernice  Porjesz,  Ph.D. 
Department  of  Psychiatry 
Health  Science  Center  at  Brooklyn 
450  Clarkson  Ave.,  Box  1203 
Brooklyn,  NY  11203-2098 

Herman  Samson,  Ph.D. 

Department  of  Physiology/Pharmacology 

Wake  Forest  University  School  of 

Medicine 
Medical  Center  Blvd. 
Winston-Salem,  NC  27157-1083 

Boris  Tabakoff,  Ph.D. 
Department  of  Pharmacology,  C-236 
University  of  Colorado  HSC 
4200  East  Ninth  Ave. 
Denver,  CO  80262-0236 

Maharaj  Ticku,  Ph.D. 
Department  of  Pharmacology 
University  of  Texas  HSC 
7703  Floyd  Curl  Dr. 
San  Antonio,  TX  78284-7764 

Nora  Volkow,  M.D. 
Department  of  Medicine 
Brookhaven  National  Laboratory 
Upton,  NY  11973 


558 


Subcommittee  Report 


Gary  Wand,  M.D. 
Division  of  Endocrinology 
The  Johns  Hopkins  University 
School  of  Medicine,  Ross  863 
720  Rutland  Ave. 
Baltimore,  MD  21205 


Experts  in  Non-Alcohol- 
Related  Areas 

Robert  Freedman,  M.D. 
Department  of  Psychiatry 
University  of  Colorado  HSC 
Box  C-268-71 
4200  East  Ninth  Ave. 
Denver,  CO  80262 

Peter  Kalivas,  Ph.D. 
Department  of  VCAPP 
Washington  State  University 
Pullman,  WA  99154 

Steven  Paul,  M.D. 
Lilly  Research  Laboratories 
Lilly  Corporate  Center 
893  Delaware  Ave. 
Indianapolis,  IN  46285 


APPENDIX  B:  EXPERTS 
IN  NEUROSCIENCE 
AND  BEHAVIOR 

Fulton  T.  Crews,  Ph.D. 

University  of  North  Carolina  School 

of  Medicine 
Center  for  Alcohol  Studies 
Thurston-Bowles  Bldg.,  CB  7178 
Chapel  Hill,  NC  27599-7178 

Cindy  Ehlers,  Ph.D. 

The  Scripps  Research  Institute,  CVN-14 

10550  North  Torrey  Pines  Rd. 

La  Jolla,  CA  92037 

George  Fein,  Ph.D. 
Department  of  Psychiatry 
University  of  California 
Parnassus  Ave. 
San  Francisco,  CA  94143 


Peter  R.  Finn,  Ph.D. 
Department  of  Psychology 
Indiana  University 
1101  East  10th  St. 
Bloomington,  IN  47405-7007 

Kathleen  A.  Grant,  Ph.D. 
Department  of  Physiology  and 

Pharmacology 
Wake  Forest  University  School  of 

Medicine 
Medical  Center  Blvd. 
Winston-Salem,  NC  27157-1083 

Paula  L.  Hoffman,  Ph.D. 
Department  of  Pharmacology,  C-236 
University  of  Colorado  Health  Sciences 

Center 
4200  East  9th  Ave. 
Denver,  CO  80262-0001 

John  H.  Krystal,  M.D. 

Yale-VA  Alcoholism  Research  Center 

Psychiatry  Service  (116-A) 

VA  Connecticut  Healthcare  System 

950  Campbell  Ave. 

West  Haven,  CT  06516 

David  M.  Lovinger,  Ph.D. 

Department  of  Molecular  Physiology  and 

Biophysics 
Vanderbilt  University  School  of  Medicine 
702  Light  Hall 
Nashville,  TN  37232-0615 

A.  Leslie  Morrow,  Ph.D. 

University  of  North  Carolina  School  of 

Medicine 
Center  for  Alcohol  Studies 
3027  Thurston-Bowles  Bldg.,  CB  7178 
Chapel  Hill,  NC  27599-7178 

Marlene  Oscar-Berman,  Ph.D. 

Division  of  Psychiatry 

Boston  University  School  of  Medicine 

715  Albany  St. 

Boston,  MA  02118 

Catherine  Rivier,  Ph.D. 
The  Salk  Institute 
10010  North  Torrey  Pines  Rd. 
La  Jolla,  CA  92037-1099 


559 


NIAAA's  Neuroscience  and  Behavioral  Research  Portfolio 


George  R.  Siggins,  Ph.D. 

The  Scripps  Research  Institute,  CVN-12 

10550  North  Torrey  Pines  Rd. 

La  Jolla,  CA  92037 

Linda  P.  Spear,  Ph.D. 

Department  of  Psychology 

Center  for  Developmental  Psychobiology 

Binghamton  University 

Binghamton,  NY  13902-6000 

Edith  V.  Sullivan,  Ph.D. 

Department  of  Psychiatry 

Stanford  University  School  of  Medicine 

410  Quarry  Rd. 

Stanford,  CA  94305-5717 

Steven  N.  Treistman,  Ph.D. 
Department  of  Pharmacology 
University  of  Massachusetts  Medical 

Center 
55  Lake  Ave.  North 
Worcester,  MA  01655 

Gary  Wand,  M.D. 
Division  of  Endocrinology 
The  Johns  Hopkins  University 
School  of  Medicine,  Ross  863 
720  Rutland  Ave. 
Baltimore,  MD  21205 

Friedbert  Weiss,  Ph.D. 
Department  of  Neuropharmacology 
The  Scripps  Research  Institute,  CVN-15 
10550  North  Torrey  Pines  Rd. 
La  Jolla,  CA  92037 


APPENDIX  C:  NIAAA 
PROGRAM  STAFF 

Walter  A.  Hunt,  Ph.D. 

Neurosciences  and  Behavioral  Research 

Branch 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 


Robert  W.  Karp,  Ph.D. 
Neurosciences  and  Behavioral  Research 

Branch 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 

Yuan  Liu,  Ph.D. 

Neurosciences  and  Behavioral  Research 

Branch 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 

Ellen  D.  Witt,  Ph.D. 

Neurosciences  and  Behavioral  Research 

Branch 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 

Samir  Zakhari,  Ph.D. 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 


APPENDIX  D:  NIAAA  STAFF, 
REPRESENTATIVES  FROM 
OTHER  NIH  INSTITUTES, 
AND  GUESTS 

Megan  Adamson,  M.D. 
Office  of  Collaborative  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Faye  Calhoun,  D.P.A. 
Office  of  Collaborative  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Mary  Dufour,  M.D.,  M.P.H. 
Deputy  Director,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Michael  J.  Eckardt,  Ph.D. 
Office  of  Scientific  Affairs,  NIAAA 
6000  Executive  Blvd.,  Suite  409 
Bethesda,  MD  20892-7003 


560 


Subcommittee  Report 


Joanne  Fertig,  Ph.D. 

Division  of  Clinical  and  Prevention 

Research,  NIAAA 
6000  Executive  Blvd.,  Suite  505 
Bethesda,  MD  20892-7003 

Richard  K.  Fuller,  M.D. 

Division  of  Clinical  and  Prevention 

Research,  NIAAA 
6000  Executive  Blvd.,  Suite  505 
Bethesda,  MD  20892-7003 

Klaus  Gawrisch,  Ph.D. 

Laboratory  of  Membrane  Biochemistry 

and  Biophysics,  NIAAA 
12420  Parklawn  Dr.,  Room  116 
Bethesda,  MD  20892-8115 

Thomas  Gentry,  Ph.D. 
Office  of  Collaborative  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Enoch  Gordis,  M.D. 
Director,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Daniel  Hommer,  M.D. 

Laboratory  of  Clinical  Studies,  NIAAA 

Bldg.  10,  Room3C114 

Bethesda,  MD  20892-1256 

Nancy  Hondros 

Planning  and  Financial  Management 

Branch,  NIAAA 
6000  Executive  Blvd.,  Suite  412 
Bethesda,  MD  20892-7003 

William  M.  Lands,  Ph.D. 
Office  of  the  Director,  NIAAA 
6000  Executive  Blvd.,  Suite  400 
Bethesda,  MD  20892-7003 

Burt  Litman,  Ph.D. 

Laboratory  of  Membrane  Biochemistry 

and  Biophysics,  NIAAA 
12420  Parklawn  Dr.,  Room  116 
Bethesda,  MD  20892-8115 


Raye  Litten,  Ph.D. 

Division  of  Clinical  and  Prevention 

Research,  NIAAA 
6000  Executive  Blvd.,  Suite  505 
Bethesda,  MD  20892-7003 

Steve  Long,  M.A. 
Office  of  Policy  Analysis,  NIAAA 
6000  Executive  Blvd.,  Suite  405 
Bethesda,  MD  20892-7003 

Matthew  McGue,  Ph.D. 
Department  of  Psychology 
University  of  Minnesota 
Elliot  Hall,  Room  N-2 18 
75  East  River  Rd. 
Minneapolis,  MN  55455 

Antonio  Noronha,  Ph.D. 
Office  of  Scientific  Affairs,  NIAAA 
6000  Executive  Blvd.,  Suite  409 
Bethesda,  MD  20892-7003 

Carrie  L.  Randall,  Ph.D. 

Department  of  Psychiatry  and  Behavioral 

Science 
Medical  University  of  South  Carolina 
171  Ashley  Ave. 
Charleston,  SC  29425 

Norman  Salem,  Jr.,  Ph.D. 

Laboratory  of  Membrane  Biochemistry 

and  Biophysics,  NIAAA 
12420  Parklawn  Dr.,  Room  116 
Bethesda,  MD  20892-8115 

Jules  Selden,  D.V.M.,  Ph.D. 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 

Martin  K.  Trusty,  M.S. 

Office  of  Planning  and  Resource 

Management,  NIAAA 
6000  Executive  Blvd.,  Suite  412 
Bethesda,  MD  20892-7003 

Ernestine  Vanderveen,  Ph.D. 
Division  of  Basic  Research,  NIAAA 
6000  Executive  Blvd.,  Suite  402 
Bethesda,  MD  20892-7003 


561 


NIAAA'' s  Neuroscience  and  Behavioral  Research  Portfolio 


Kenneth  Warren,  Ph.D. 
Office  of  Scientific  Affairs,  NIAAA 
6000  Executive  Blvd.,  Suite  409 
Bethesda,  MD  20892-7003 

Forrest  Weight,  M.D. 

Laboratory  of  Molecular  and  Cellular 

Neurobiology,  NIAAA 
12420  Parklawn  Dr.,  Room  118 
Bethesda,  MD  20892-8115 


Herbert  Weingartner,  Ph.D. 
National  Institute  on  Drug  Abuse 
Bldg.  31,  Room  1B58 
Bethesda,  MD  20892-1250 


562 


NOTES 


NOTES 


NOTES 


NOTES 


N 


I   LliRARy 

|H 

™    Amazing  Research. 
Amazing  Help. 


http://nihlibrary.nih.gov 


10  Center  Drive 

Bethesda,  MD  20892-1150 

301-496-1080 


3  1496  00823  7078 


NIH  Publication  No.  00-4520 
National  Institutes  of  Health 
Printed  2000