Skip to main content

Full text of "Basic lipid methodology"

See other formats


LIBRARY  OF  THE 

UNIVERSITY  OF  ILLINOIS 

AT  URBANA-CHAMPAIGN 


*!CULTUR£ 


!"he  person  charging  this  material  is  re- 
ponsible  £or~ks  return  to  the  library  from 
/hich  it  wa,s_ withdrawn  on  or  before  the 
atest  Date  statnped  befow. 

Theft,  mutilation,  and  underlining  of  books 
are  reasons  for  disciplinary  action  and  may 
result  in  dismissal  from  the  University. 

UNIVERSITY    OF     ILLINOIS     LIBRARY    AT    URBANA-CHAMPAIGN 


N0£4s«8> 


JUU 


J£££3"$ffl 


0CI-2£4983 


*s& 


18  «93 

|t)3l|o4 

Qtg-jj  1083    ^P  14  2001 


L161  — O-1096 


Digitized  by  the  Internet  Archive 

in  2012  with  funding  from 

University  of  Illinois  Urbana-Champaign 


http://www.archive.org/details/basiclipidmethod19john 


—  5 


&*■■&£■ 


OH 

CH2OH  OH, 


/    CH?OH  OH 

f    co'NH' 


j  ,  university  of  fafwrar 

""'""  AGRICbi, 


NH 
CH-TW  _1     9H20H  .  /  ^^^^^^CH- 

C02H 


■ffSlo 


it     "W.-1 


CH2 

CH3- CO-NH  HC  -  OH  *Hfi  U**^  ^ 

HC-OH 

ch2oh  AUG  24  137; 

ON,, 

basic  |JD. , 

Hd  methodology 

by  Patricia  V.Johnston 


oh 

ch2oh  n  OH„ 

—  O  /     CH2OH 


CH3CO-NH  HC-OH 
1 
HC-OH 

CH2OH 


OH 

CHoOH  OH 

-  •     CH2OH 


HO 

CH3CO-NH  HC-OH 
HC-OH 


OH 

HO 


basic  lipid  methodology 

Patricia  V.Johnston 

Assistant  Professor  of  Food  Science 


Special  Publication  19 

January,  1971 

College  of  Agriculture 

University  of  Illinois  at  Urbana-Champaign 


INTRODUCTION 


T 


-he  size  and  diverse  nature  of  the  group  of  compounds  known  as 
L  the  lipids  preclude  the  coverage  of  their  chemistry  to  any  extent  in 
elementary  courses  in  organic  chemistry;  this  vast  group  is,  in  general, 
considered  to  be  a  field  of  study  in  itself.  This  publication  has  been  pre- 
pared as  a  laboratory  handbook  for  those  with  little  previous  experience 
in  lipid  analysis.  Therefore,  while  a  knowledge  of  basic  organic  chemis- 
try is  assumed,  previous  knowledge  of  lipid  chemistry  is  not.  The  first 
chapter  is  an  introduction  to  lipid  chemistry.  It  includes  definitions, 
classification,  and  basic  structures  of  lipids.  The  rest  of  the  book  deals 
with  the  preparation  and  care  of  samples  for  lipid  analysis  and  pro- 
cedures for  the  analysis  of  commonly  occurring  lipids.  A  knowledge  of 
newer  techniques  basic  to  the  study  of  lipids  is  not  assumed;  therefore, 
the  book  also  serves  as  an  introduction  to  column,  thin-layer,  and  gas- 
liquid  chromatography. 

Numerous  references  to  original  research  papers  are  not  given  since 
this  work  is  not  intended  to  be  an  exhaustive  account  of  all  the  methods 
available  for  the  analysis  of  lipids.  Rather,  it  is  a  personal  account  of 
methods  tried,  tested,  and  known  to  work.  A  few  references  to  research 
papers  are  included  where  pertinent,  and  a  selected  bibliography  is 
designed  to  lead  the  interested  reader  to  a  wider  range  of  methods 
and  applications.  Analyses  of  only  the  more  commonly  encountered 
lipids  are  described,  but  the  Suggested  Further  Readings  includes  some 
works  dealing  with  the  analysis  of  the  less  common  lipid  subclasses. 


Patricia  V.  Johnston  is  Research  Assistant 
Professor  in  the  Children's  Research  Center  and 
Assistant  Professor  of  Food  Science.  She  is  also 
a  member  of  the  Nutritional  Sciences  Faculty. 
Developmental  neuro chemistry  and  neurobiology 
arc  Dr.  Johnston's  main  research  interests.  She 
is  especially  interested  in  pre-  and  postnatal  con- 
ditions, such  as  malnutrition  and  drug  therapy, 
which  may  affect  the  development  of  the  hptd- 
rich  myelin  sheath  of  nerve  cells. 
This  book  was  written  in  part  while  Dr.  Johnston 
was  supported  by  funds  from  the  Hatch  NC74 
committee. 
The  figure  on  the  cover  is  a  mono  sialoganglio  side. 


TABLE  OF  CONTENTS 

Introduction ii 

I.  LIPIDS:  DEFINITIONS,  CLASSIFICATION, 
NOMENCLATURE I 

Simple  lipids 1 

Neutral  glycerides 1 

Esters  of  fatty  alcohols 3 

Derived  lipids 4 

Glyceryl  ethers 8 

Phosphoglycerides 8 

Phosphatidic  acids,  phosphatidyl  glycerols,  and 

polyglycerophosphatides 10 

Phosphatidyl  ethanolamines,  cholines,  serines,  and  inositols.  .  .  10 

Lysophosphoglycerides 12 

Plasmalogens 12 

Phosphonolipids 13 

Sphingolipids 14 

Ceramides 14 

Sphingomyelin 15 

Glycolipids:  cerebrosides   and   sulfatides    15 

Gangliosides 16 

II.  PREPARATION  AND  HANDLING  OF  SAMPLES  FOR 

ANALYSIS  OF  LIPID  CONSTITUENTS 19 

General  techniques  in  lipid  chemistry 

Prevention  of  oxidation 19 

Elimination  of  possible  contaminants 20 

Unwanted  emulsions  and  other  hazards  in  lipid  chemistry   ...  21 

Extraction  of  lipids  from  various  sources 22 

From  blood  serum 22 

From  erythrocytes 24 

From  brain 25 

From  other  sources 26 

Removal  of  nonlipid  contaminants  from  extracts 

Aqueous  washing 26 

Treatment  on  cellulose  and  Sephadex  columns 27 

III.  COLUMN  CHROMATOGRAPHY 30 

Solid-liquid  adsorption  chromatography 30 

Liquid-liquid  partition  chromatography 31 

Preparation  of  columns 31 

Silicic  acid  columns    34 

Initial  separation:  neutral  lipids  from  phospho- 

and  glycolipids 35 

Separation  of  neutral  lipids 35 

Separation  of  polar  lipids 36 

Florisil  columns 37 


Diethylaminoethyl  (DEAE)  and  trimethylaminoethyl  (TEAE) 

cellulose  columns 37 

Separation  of  lipid  samples  into  acidic  and 

nonacidic  fractions 39 

A  general  elution  scheme 39 

IV.  THIN-LAYER  AND  PAPER  CHROMATOGRAPHY 42 

Thin-layer  chromatography 42 

Preparation  of  thin-layer  chromatographic  plates 42 

Detection  of  lipids  on  chromatograms 45 

Separation  of  neutral  lipids 48 

Argentation  thin-layer  chromatography 51 

Separation  of  phospholipids  and  glycolipids 52 

Quantitative  thin-layer  chromatography 53 

Paper  chromatography 59 

V.  GAS-LIQUID  CHROMATOGRAPHY 61 

Instrumentation 61 

Qualitative  analysis 66 

Quantitative    analysis 69 

Chemical  modification  of  compounds  for  analysis  by  GLC 75 

Analysis  of  methyl  esters  of  fatty  acids 75 

Analysis  of  fatty  aldehydes 78 

Analysis  of  glycerides 81 

Analysis  of  other  lipids  and  components  derived  from  lipids.  .  .  82 

Pyrolysis-GLC 82 

VI.  PROCEDURES  FOR  THE  DETERMINATION  OF  SPECIFIC 

ELEMENTS,  FUNCTIONAL  GROUPS,  AND  LIPID  CLASSES  84 

Determination  of  organic  phosphorus 84 

Determination  of  cholesterol 

By  spectrophotometry 85 

By  gas-liquid  chromatography 87 

Determination  of  glycolipid  sugars 

By  using  anthrone 89 

By  gas-liquid  chromatography 90 

Determination  of  N-acetylneuraminic  acid  (to  determine  gangliosides) 

By  using  resorcinol 90 

By  gas-liquid  chromatography 91 

Determination  of  plasmalogens 

By  colorimetry     92 

By  two-dimensional  thin-layer  chromatography 93 

Determination  of  the  amount  of  trans  double  bond 

By  infrared  spectrophotometry 94 

By  gas-liquid  chromatography 97 

Suggested  Further  Readings 99 

Index 100 


I.  Lipids:  Definitions,  Classification, 
Nomenclature 


The  term  lipid  describes  a  large  group  of  compounds  of  a  chem- 
ically diverse  nature.  This  book  employs  a  simple  definition  and 
classification  system,  as  no  system  has  been  universally  accepted.  Lipids 
may  be  denned  as  compounds  found  in  living  organisms  and  generally 
insoluble  in  water  but  soluble  in  organic  solvents.  There  are  exceptions 
to  this  definition  since  some  lipids  are  sparingly  soluble  in  water  while 
others  are  soluble  in  a  very  limited  number  of  organic  solvents.  In 
general,  however,  this  definition  holds. 

Three  major  lipid  classes  exist,  namely  the  simple  lipids  and  the  two 
general  groups  of  polar  lipids,  the  glycerophosphatides  and  the  sphingo- 
lipids.  We  shall  consider  each  of  these  classes  individually.  Examples  of 
the  structural  formulae  of  the  lipids  mentioned  are  shown. 

SIMPLE  LIPIDS 

These  compounds  comprise  the  neutral  lipids  such  as  the  glycerides, 
esters  of  fatty  alcohols,  and  lipids  derived  from  these  compounds  by 
alkaline  or  acid  hydrolysis. 

Neutral  Glycerides 

The  neutral  glycerides  are  fatty  acid  esters  of  glycerol,  and  although 
the  most  abundant  are  the  triglycerides,  mono-  and  diglycerides  do  occur 
naturally.  The  glycerides  are  named  according  to  the  position  of  the 
fatty  acid  substituent  on  the  glycerol  moiety;  thus  two  isomers  of  mono- 
and  diglycerides  exist.  In  a  triglyceride  all  the  glyceride  hydroxy! 
groups  are  esterified. 

0 
<  H2C-OH  H2C-0-C-R 


0  "2, 

/3     HC-O-C-R  HC-OH 

I  i 

<L'  H2C-OH  H2C-OH 

monoglyceride,  /?-form  monoglyceride,  a-form 


LIPIDS:  DEFINITIONS 


0 

II 

H2C  -  0  "  C  "  R, 

i         o 

II 


HC  -  0  -  C  -  R2 

i 
H2C-OH 

diglyceride,  «,/3-form 


0 

ii 

H2C-0-C-  R, 
I 
HC  -  OH 
,        o 

H2C  -  0  "  C  "  R2 


diglyceride,  «,«'-form 


H2C-0-C-R, 
,         0 

HC-0-C-R2 
i 

H0C-O-C-R3 
11 
0 

triglyceride 


The  carbons  of  the  glycerol  moiety  are  often  designated  1,  2,  and  3 
instead  of  a,  p,  and  a'.  Ri,  R2,  and  R3  refer  to  the  alkyl  chain  of  the 
esterifying  fatty  acids.  In  general,  glycerides  are  named  by  the  trivial 
name  for  the  fatty  acids.  For  example,  if  stearic  acid,  CH3(CH2)i6- 
COOH,  is  the  esterifying  fatty  acid,  we  can  have: 


H2C  "OH 
,  0 

HC  -0-  C(CH2),6CH3 
1 
H2C  "OH 

2  (or  /3)-monostearin 


0 
H2C-0-C(CH2)l6CH3 

HC  "OH 
1 

H2C-0H 

1  (or  «)-monostearin 


0 
H2C-0-C(CH2),6CH3 

1         0 
HC-0-C(CH2),6CH3 

1 
H2C-0H 

1,2  (or  a,/?)-distearin 


0 
H2C-0-C(CH2)l6CH3 

1 
HC-OH 

«  0 

H2C-0-C(CH2)l6CH3 

1,3  (or  a,a')-distearin 


H2C-0-C(CH2),6CH3 

HC-0-C(CH2),6CH3 
1 

H2C-0-C(CH2),6CH3 
11 
0 


tristearm 


CLASSIFICATION,  NOMENCLATURE  3 

_  When  the  fatty  acid  composition  is  mixed,  the  position  of  each  fatty 
acid  is  specified  when  known.  Thus,  introducing  oleic  CH3(CH2)7- 
CH  =  CH(CH2)7COOH  and  palmitic  CH3(CH2)14  COOH  acids, 
we  can  have  as  examples: 


H2C-0-C(CH2)l4CH3 

HC  -0  "  C(CH2)7 CH=  CH(CH2)7CH3 

0 
H2C*0H 

l-palmitoyl-2-olein 


H2C-0-C(CH2),4CH3 

HC-0-C(CH2)l4CH3 
I 

H2C-0-C(CH2)7CH  =  CH(CH2)7CH3 

0 

1,2-dipalmitoyl  olein 


H2C-0-C(CH2)7CH=  CH(CH2)7CH, 


HC-OH 
•  0 


H2C-0-C(CH2),4CH3 
l-oleoyl-3-palmitin 


H2C-0-C(CH2),4CH3 
I  0 

HC-0-C(CH2),6CH3 

H2C  -  0  -  C(CH2)7CH=  CH(CH2)7CH3 
6 

l-palmitoyl-2-stearoyl-3-olein,  or 
(since  the  1  and  3  positions  are 
equivalent)  l-oleoyl-2-stearoyl- 
3-palmitin 


When  the  fatty  acids  are  more  complicated  and  not  generally  known 
by  any  trivial  name,  they  must  of  course  be  named  systematically.  The 
generally  accepted  nomenclature  for  fatty  acids  is  described  under 
Derived  Lipids  (p.  4). 

Esters  of  Fatty  Alcohols 

Alcohols  that  are  relatively  insoluble  in  water  but  are  soluble  in 
organic  solvents  fall  under  the  class  of  fatty  (or  lipid)  alcohols.  This 
class,  therefore,  includes  long-chain  aliphatic  alcohols  (more  than  8  car- 
bons), aromatic  alcohols  such  as  cholesterol,  and  other  steroids,  includ- 
ing the  vitamin  D  group.  Vitamins  A  and  E  are  also  lipid  alcohols,  but 
because  of  the  vast  literature  devoted  to  the  vitamins  we  shall  not 
discuss  them  further  here.  Cholesterol  is  the  alcohol  with  which  we  shall 
be  most  concerned.  Cholesterol  exists  in  nature  in  the  esterified  state  and 
in  the  free  form.  The  stearic  acid  ester  of  cholesterol  is  shown  (p.  4). 

The  esters  of  long-chain  alcohols  and  fatty  acids  are  known  as 
waxes;  natural  waxes  such  as  beeswax  and  wool  waxes  are  mixtures  of 


LIPIDS:  DEFINITIONS 


CH3  CH3 

CH"CH2"CH2*CH2"CH 
CH3 


CH3(CH2),6C-0^N/<^ 

cholesterol  ester 

these  esters.  Trace  amounts  of  other  components,  such  as  hydrocarbons, 
are  also  found  in  natural  waxes. 

Derived  Lipids 

If  the  products  obtained  on  hydrolysis  of  a  lipid  are  soluble  in 
organic  solvents  and  relatively  insoluble  in  water,  they  are  termed 
derived  lipids.  From  our  point  of  view,  the  most  important  are  the  fatty 
acids.  We  shall  now  discuss  their  structure  and  nomenclature  in  some 
detail.  For  the  most  part,  the  fatty  acids  in  mammalian  tissues  and  body 
fluids  are  straight  chain  and  contain  an  even  number  of  carbon  atoms. 
Hydroxy,  keto,  and  branch  chain  fatty  acids  also  occur;  indeed,  the 
nervous  system  of  mammals  contains  considerable  amounts  of  2(a)- 
hydroxylated  fatty  acids.  Most  fatty  acids  found  in  tissues  are  in  ester 
or  amide  linkages,  but  small  amounts  of  free  fatty  acids  (FFA)  do 
occur  and  are  of  great  importance  metabolically  both  in  supplying 
energy  and  in  lipid  biosynthesis. 

It  is  usual  to  divide  the  fatty  acids  into  two  main  classes,  saturated 
and  unsaturated.  Since  aliphatic  acids  are  regarded  as  derivatives  of  the 
hydrocarbons  which  have  the  same  number  of  carbon  atoms,  the  names 
of  fatty  acids  are  derived  from  the  appropriate  parent  hydrocarbon. 
Saturated  fatty  acids  are  named,  according  to  the  modified  Geneva 
system,  by  replacing  the  terminal  "e"  of  the  parent  hydrocarbon  name 
with  the  suffix  "oic."  Thus,  the  saturated  fatty  acid  that  is  related  to  the 
hydrocarbon  octane,  CH3(CH2)GCH3,  is  known  as  octanoic  acid. 
Most  fatty  acids,  however,  were  known  and  named  before  the  Geneva 
convention,  and  the  use  of  their  non-systematic  ("trivial")  names  per- 
sists. The  accompanying  tables  of  fatty  acids,  therefore,  include  the 
trivial  names  as  well  as  the  systematic  names.  Some  commonly  oc- 
curring saturated  fatty  acids  are  shown  in  Table  1. 

The  simplest  unsaturated  fatty  acids  have  the  empirical  formula 
CnH2n_202;  that  is,  they  contain  only  one  double  bond.  They  are  named 
by  replacing  the  "e"  of  the  corresponding  unsaturated  hydrocarbon  with 
the  suffix  "oic";  thus  one  has  octenoic  acid,  decenoic  acid,  and  so  on. 
These  fatty  acids  are  termed  "monoenoic."  Fatty  acids  with  two,  three, 
four,  five,  and  more  double  bonds  are  named  by  taking  the  stem  of  the 


CLASSIFICATION,  NOMENCLATURE 
Table  7.  —  Some  Saturated  Fatty  Acids 


Systematic 


Trivial  name 


Formula 


Butanoic 

Hexanoic 

Octanoic 

Decanoic 

Dodecanoic 

Tetradecanoic 

Hexadecanoic 

Octadecanoic 

Eicosanoic 

Docosanoic 

Tetracosanoic 


Butyric 

Caproic 

Caprylic 

Capric 

Laurie 

Myristic 

Palmitic 

Stearic 

Arachidic 

Behenic 

Lignoceric 


CH3(CH2)2COOH 
CHaCCH^COOH 
CH3(CH2)6COOH 
CH3(CH2)8COOH 
CH3(CH2)10COOH 
CH3(CH2)12COOH 
CH3(CH2)14COOH 
CH3(CH2)16COOH 
CH3(CH2)18COOH 
CH3(CH2)20COOH 
CH3(CH2)22COOH 


name  of  corresponding  hydrocarbon,  octa-,  deca-,  etc.,  and  adding  the 
appropriate  ending:  "-dienoic"  (2  double  bonds),  "-trienoic"  (3  double 
bonds),  etc.  Fatty  acids  with  multiple  double  bonds  are  referred  to 
collectively  as  polyunsaturated  fatty  acids  (PUFA)  and  individually 
as  dienoic,  tnenoic,  tetraenoic,  pentaenoic,  and  hexaenoic  fatty  acids 
(see  Table  2).  A  name  must  also,  of  course,  designate  both  the  position 
of  the  double  bonds  along  the  chain  and  their  geometric  configuration: 
cis  or  trans.  Double  bonds  are  assumed  to  be  cis,  unless  a  statement  is 
made  to  the  contrary.  This  convention  has  been  adopted  because  most 
naturally  occurring  fatty  acids  have  their  double  bonds  in  the  cis  con- 
figuration; moreover,  most  common  trans  isomers  have  a  different  trivial 
name  which  is  used,  so  the  problem  is  often  completely  circumvented. 
For  example,  octadecenoic  acid  with  a  double  bond  on  the  9th  carbon 
has  both  cis  and  trans  forms.  The  cis  form  has  the  trivial  name  of 
oleic  acid,  and  the  trans  form  is  known  as  elaidic  acid.  Similarly,  when 


HC(CH2)7CH3 
ii 

HC(CH2)7COOH 
cis  form :  oleic  acid 


CH3(CH2)7CH 
H 

HC(CH2)7C00H 
trans  form:  elaidic  acid 


both  double  bonds  of  octadecadienoic  acid  are  trans,  the  acid  is 
termed  linolelaidic  acid.  While  trans  fatty  acids  are  rare  as  natural 
constituents,  they  do  occur  naturally  in  some  plants  and  animals-  more- 
over, when  included  in  the  diet,  they  are  deposited  in  body  tissues  (1). 
They  are,  therefore,  nutritionally  significant.  A  further  discussion  of 
trans  fatty  acids  is  included  in  the  description  of  their  analysis  (see 
p.  94) .  J        v 

While  the  naming  of  geometric  isomers  is  relatively  straightforward, 
the  designation  of  double  bond  position  is  confused  by  the  fact  that 


LIPIDS:  DEFINITIONS 


CM 


X 

DC 

DC 

o 

c 
c 

O 

U 

o 

O 

II 

u 

u 

DC 

'"JJ 

u 

^JJ 

U 

DC 

DC 

£ 
u 

u 

£ 

U 

X 

E 

u 

X 

X 

o 

U 

£ 

u 

u 

■M 

o 

II 

u 

II 

II 

X 

0) 

u 

E 

DC 

E 

X 

u 

U 

£ 

u 

a: 

E 
u 

£ 

u 

£ 
u 

u 

£ 
u 

DC 

X 

~^~~X 

X 

o 

o 

O 

u 

u 

DC 
U 
II 

u 

u 

o 

o 

O 

II 

ii 

II 

II 

u 

u 

u 

X 

X 

E 

X 

CI 

'"n 

M 

u 

u 

E 

u 

£ 
u 

DC 
U 

U 

u 

DC 

u 

DC 

u 

X 

u 

u 

u 

£ 
u 

£ 
u 

X 

K 

ffi 

dc 

E 

DC 

DC 

u 

U 

*u 

u 

u 

U 

U 

II 

•2  II 

-  II 

II 

II 

II 

II 

X 

*X 

£DC 

DC 

DC 

II 

X 

X 

u 

u 

U 

U 

u 

X 

u 

u 

•a 

r~ 

s~7y 

'"TJ 

-"> 

* — j 

' — J 

u 

£ 
u 

' — J 

' — J 

X 

dc 

£ 

X 

X 

DC 

£ 

u 

u 

u 

u 

u 

u 

u 

* — * 

v — 

X 

£ 

DC 

ffi 

X 

£ 

£ 

£ 

u 

u 

U 

u 

u 

u 

u 

u 

u 

u 

<v 

c 

CJ 

o 

y 

y 
'5 

o 

s 

_y 

is 

0 

e 

o 

>—" 

*s 

"c5 

c 

J 

c 

CO 

cu 

o 

U3 

J 

?- 

*J 

u 

O 

C 

u 

'5 

c 

c 
.22 

U 

0> 

0) 

"o 

_y 

"C 

'5 

— 

a 

U 

0) 

"E 

ca 

c 
'C 

0) 

c 

u 

u 

'o 
c 
<v 

u 

'-5 

CO 

o 

u 

09 

y 

'o 

c 

"o 

CO 

o 

<v 
-o 

co" 

(J 

O 

0) 

-v 

4J 

OS 

O 

y 

CD 

u 

<u 

co 
X 

S3 
o 

0) 

CO 

u 

<v 

"O 
O 

o 

6\ 

CO 

U 

o 

CN 

o 

o 

cn" 

o 
u 

•r— 1 

X 

o 

*— 4 

ot 

y—t 

00 

oo" 

o» 

On 

s 

Os 

o 

Os 

LC 

LO 

.** 

.<^ 

a 

,^ 

<^> 

to 

«o 

<^> 

k 

•*» 

•"- 

•«» 

2 

<^ 

<o 

vo 

VJ 

<o 

o 

CLASSIFICATION,  NOMENCLATURE  7 

several  conventions  exist.  To  be  able  to  follow  the  literature,  one  must 
know  all  the  conventions.  In  the  simplest  procedure  the  terminal  car- 
boxyl  group  carbon  is  number  1  and  the  rest  of  the  chain  is  2,  3,  4,  etc. 
Thus  palmitoleic   acid  named  systematically  becomes  9-hexadecenoic 

9  1 

acid,  CH3(CH2)5CH  =  CH(CH2)7COOH.  However,  especially  in 
older  literature,  lengthier  conventions  are  used.  The  Greek  letter  A  is 
used  to  indicate  the  presence  of  double  bonds,  and  both  carbon  atoms 
participating  in  the  bonds  are  indicated.  Palmitoleic  acid  then  be- 
comes A9»10-hexadecenoic  acid.  A9-hexadecenoic  acid  is  also  used. 

There  is  a  growing  tendency  to  use  a  shorthand  designation  for  fatty 
acids.  This  is  very  useful  especially  when  long  lists  of  fatty  acids  are 
given  as,  for  example,  in  the  complete  gas  chromatographic  analysis  of 
the  fatty  acids  of  natural  lipids.  In  this  convention  the  Greek  letter  o> 
is  used  to  refer  to  the  terminal  carbon  atom  away  from  the  carboxyl 
group.  The  position  of  the  double  bond  is  then  indicated  with  respect 
to  the  o)  carbon.  Palmitoleic  acid  is,  therefore,  the  w  6-hexadecenoic  acid. 
The  length  of  the  carbon  chain  and  the  number  of  double  bonds  is  indi- 
cated by  the  shorthand  form  C16:l,  where  the  number  before  the  colon 
gives  the  chain  length  and  the  number  after  the  colon  indicates  the 
number  of  double  bonds.  Palmitoleic  acid  is,  therefore,  designated 
C16:lo)6. 

When  there  is  more  than  one  double  bond  a  question  arises ; 
namely,  is  it  sufficient  to  define  the  position  of  only  one  double  bond 
with  respect  to  the  w  carbon  atom?  To  answer  this,  we  must  con- 
sider still  another  way  of  classifying  fatty  acids.  Polyenoic  acids  may 
be  classified  as  conjugated  or  unconjugated  (nonconjugated  is  also 
used)  depending  on  the  relative  position  of  the  double  bonds.  If  the 
double  bonds  are  separated  by  one  or  more  single-bonded  carbon  atoms, 
—  C  =  C—  Cn  —  C  =  C— ,  the  acid  is  said  to  be  unconjugated. 
When  double-bonded  carbon  atoms  are  adjacent  to  each  other,  —  C 
=  C  —  C  =  C  —,  the  acid  is  termed  conjugated.  Fatty  acids  from 
mammalian  sources  are  usually  unconjugated.  Double  bonds  are  usually 
separated  by  one  single-bonded  carbon  atom;  that  is,  by  a  single  meth- 
ylene ( —  CH2  —  )  group.  It  is,  therefore,  perfectly  correct  in  such  cases 
to  define  double  bond  position  by  reference  to  the  first  double  bond  from 
the  a)  carbon  atom.  Thus,  the  commonly  occurring  mammalian  fatty 
acids,  5,  8,  11-eicosatrienoic  acid  and  5,  8,  11,  14-eicosatetraenoic 
(arachidonic)  acid,  can  be  referred  to  as  20:3  w  6  and  20:4  w  6  respec- 
tively. If,  however,  the  double  bonds  are  conjugated,  then  reference 
must  be  made  to  this  fact.  If  the  acid  is  unconjugated  but  double  bonds 
are  separated  by  more  than  one  single-bonded  carbon  atom,  then  clearly 
one  of  the  lengthier  descriptions  of  the  structure  must  be  used. 

The  fatty  acids  listed  in  Tables  1  and  2  occur  widely  in  nature,  as  do 
numerous  others.  Linoleic  acid  is  of  great  importance  to  mammals:  it 


8  LIPIDS:  DEFINITIONS 

must  be  included  in  their  diets  to  prevent  a  deficiency  syndrome  since 
they  cannot  synthesize  it.  For  this  reason  linoleic  acid  is  called  an  essen- 
tial fatty  acid  (EFA).  Arachidonic  and  y-linolenic  acids,  synthesized  in 
mammals  from  linoleic  acid,  are  also  sometimes  termed  essential  fatty 
acids  since  they  protect  and  cure  the  EFA  deficiency  syndrome. 

Hydroxy,  keto,  branched  chain,  and  cyclic  fatty  acids  all  exist.  Ex- 
amples of  such  acids  and  a  source  of  each  are  shown  in  Table  3.  The 
only  hydroxy  fatty  acids  of  importance  in  mammals  are  the  2(a)- 
hydroxy  substituted  fatty  acids  found  in  cerebrosides  and  sulfatides 
(see  p.  15).  Some  hydroxy  and  branched  chain  acids  may  be  ingested 
by  man  in  oils  from  plant  sources  ( such  as  ricinoleic  acid  in  castor  oil) , 
and  small  quantities  of  hydroxy  and  keto  acids  may  also  be  present  in 
man's  diet  due  to  oxidative  breakdown  of  fats  (2,3). 

Glyceryl  Ethers 

Neutral  lipids  with  ether  or  vinyl  ether  linkages  have  been  isolated 
and  characterized.  In  these  lipids  an  ester-linked  fatty  acid  of  a  glyc- 
eride  is  replaced  by  a  vinyl  ether  («,/?  unsaturated)  linkage  or  an 
ether  linkage.  Thus,  one  may  have  a  aj-l-(alkenyl)  ether  of  a  2,3- 
diacyl  glycerol  with  the  general  formula: 


H2Ct  0  ~  CH  =  CH  4  R,     vinyl  ether  linkage 


R2-C-0-CH 

>  u 

H2C-0-C-R3 

and  glycerol  ethers  with  the  formula : 


o      , 

ii 


H2C  TOtR|         ether  linkage 


Rz-C-0-CH       Q 

H2C-0-C-R3 

These  lipids  are  of  importance  in  fish  and  many  invertebrates  (4). 

PHOSPHOGLYCERIDES 

Phosphoglycerides  are  defined  as  lipids  which,  on  hydrolysis,  pro- 
duce derived  lipids  plus  inorganic  phosphate  and  glycerol. 


CLASSIFICATION,  NOMENCLATURE 


'3. 


:8 

< 

B 

u. 

u 

I 

c 
c 

.c 

0 

u 


X 

o 

u 

a: 


o 


X 

o 
o 
o 

X   X 

o-o 

CSJ 

X 

o 

X 

o 


X 

o 
o 
o 


X 

o 

X 

o 


X 

o 

<M 

X 

o 

X   X 

o-o 

JO 

X 

o 

io 

X 

o 


4, 

21 

CO  bo 

X 

o 
o 
o 
^cvj 

CSJ 

X 

o 

cv) 

X 

o 

X 

o 

II 

X 

o 

X 

o 

II 

X 

o 

X 

o 

II 

X 

o 

J? 

CSJ 

X 

o 


as  o 


^a 


o 

2 

O 
O 

£ 

u 


X 

o 
o 
o 

CM 

X 

O     w 
X   X 

to 

X 

o 


X 

o 
o 
o 

"*J 

X 

o 

CSJ  <-> 
X     ii 

?    X 


CO 


o 

Ih 


y 

"3 

c 

3 


JO  LIPIDS:  DEFINITIONS 

Phosphatide  Acids,  Phosphatidyl  Glycerols, 
and  Polyglycerophosphatides 

The  simplest  phosphoglycerides  are  the  phosphatidic  acids.  In  these 
lipids  two  of  the  glycerol  —OH  groups  are  esterfied  with  fatty  acyl 
groups  and  the  third  with  phosphoric  acid  (see  also  phosphonolipids,  p. 
13).  Closely  related  to  the  phosphatidic  acids  are  the  phosphatidyl  glyc- 
erols and  the  polyglycerophosphatides.  Cardiolipin,  a  polyglycerophos- 
phatide,  occurs  mainly  as  a  mitochondrial  lipid. 

9  o 

*  0     H2C-0-C-R,  0     H2C-0-C-R, 


H 


••  '  H2C-OH 

r-n-ru  ci 


a  r2-c-o-ch    0  r2-c-o-ch  ^         H 


it 


A'  H2C-0-P-OH  H2C-0-P-0-CH2 

OH  OH 

phosphatidic  acid  phosphatidyl  glycerol 

0 

0 


H2C-0-C-R, 
0  ,  H2C-0-P-0-CH2    Q 

R2-C-0"CH  HC'OH   0H     HC-O-C-R3 

H2C-0"  P-0-CH2  H2C-0-C-R4 


1  11 

OH  0 

cardiolipin 

Phosphatidyl  Ethanolamines,  Cholines,  Serines,  and  Inositols 

This  subclass  contains  some  of  the  most  abundantly  occurring  phos- 
pholipids. In  these  compounds  the  phosphoric  acid  group  of  the  parent 
compound,  a  phosphatidic  acid,  is  linked  to  either  ethanolamine,  choline, 
serine,  or  myoinositol.  At  least  three  subclasses  of  phosphoinositide 
exist,  differing  in  the  number  of  phosphoric  acid  residues  they  contain. 

Phosphatidyl  choline  is  known  by  the  generally  accepted  trivial 
name  of  lecithin.  Phosphatidyl  ethanolamine,  however,  should  not  be 
termed  "cephalin,"  as  this  trivial  name  originally  referred  to  a  mixture 
and  has  since  lost  its  meaning. 

It  will  be  noted  that  the  phosphoglycerides  are  amphiphilic  com- 
pounds; that  is,  one  end  of  the  molecule  consists  of  a  hydrophobic  region 
of  long-chain  alkyl  groups  while  the  other  end  is  hydrophilic,  consisting 
of  the  ionic  phosphoryl  group  and  its  esterifying  molecule.  Note  also 
that  the  /?  carbon  of  the  glycerol  moiety  is  asymmetrical.  Phosphoglyc- 


CLASSIFICATION,  NOMENCLATURE 


77 


N+HZ 


Q       H2C-0-C-R, 
R2  "  C  -  0  -  CH       n 

H2C-0-P-0-CH2-CH2 
0 

phosphatidyl  ethanolamine 

0 
0     H2C-0-C-R, 

R2-C-0"CH 

H2C-0-P-0-CH2-CH2-N4(CH3)3 

g 

phosphatidyl  choline 


0 
ii 


0 
H2C-0-C-R, 


R2-C-0-CH       0 

1  H 

H2C  -  0  "  P  -  0  -  CH2  -CH  -  COO' 
i  i 

g  nh3 

phosphatidyl  serine 

brides  have  been  found  to  have  the  same  stereochemical  configuration  as 
^-glycerophosphate.  They  are,  therefore,  more  correctly  termed  L-«- 
phosphatidyl  enthanolamine,  and  so  on.  In  general,  unsaturated  fatty 
acids  estenfy  the  p  position  in  all  phosphoglycerides  and  saturated  fatty 
acids  estenfy  the  a  position. 

The  three  commonly  occurring  phosphoinositides  are  shown  below 
and  on  page  12.  The  monophosphorus  compound,  phosphatidyl  inositol, 


H         OH  R2"C"°-CH2 

> L  '        ° 

>h/5h     ?Kh 


fy         h/)-P-q 
OU         r\Lj  0 


hc-o-c-r- 


I 
-CH, 


OH       OH 

phosphatidyl  inositol 


0 

RfC-O-CHo 
H        OH 
=°f0) 1    H 

hN^         H/O-P-O-CH 
OH      OH       0 

diphosphoinositide 


HC"0-C-R2 


12  LIPIDS:  DEFINITIONS 


H 

OH  OH 

triphosphoinositide 

occurs  in  skeletal  and  cardiac  muscle ;  the  di-  and  triphosphoinositides 
constitute  a  considerable  proportion  of  the  phospholipids  in  brain  and 
occur  only  as  trace  amounts  in  other  tissues. 

Lysophosphoglycerides 

In  lysoglycerophosphatides  only  one  alcoholic  group  is  esterified  by 
a  fatty  acid;  the  other  one  is  free.  The  only  common  naturally  occurring 
member  of  this  subclass  is  lysophosphatidyl  choline  (lysolecithin), 
which  usually  has  the  /3-OH  group  free. 

Lysolecithin  occurs  to  an  appreciable  extent  in  blood  serum.  Between 
6  and  10  percent  of  the  total  lipid  phosphorus  in  human  blood  serum  is 
lysolecithin  (5).  There  is  evidence  for  the  occurrence  of  the  monoacyl 
compounds  of  all  the  other  phosphoglycerides  and  for  isomeric  forms 
in  which  the  a-OH  group  is  free. 

0 

HgC-O-C-R, 

i 
HC-OH^ 

I  9  + 

H2C-0-P-CH2-CH2-N(CH3)3 

0 

lysolecithin 

Plasmalogens 

Plasmalogens  are  monovinyl-ether,  monofatty-acyl  phosphoglyc- 
erides. They  are  known  generally  by  the  trivial  name  plasmalogens  and 
occur  in  a  wide  range  of  tissues  in  most  species.  Typically,  the  fatty  acyl 
group  at  the  a  position  is  replaced  by  an  alkenyl  group  so  that  in  place 
of  the  ester  linkage  there  is  a  vinyl  ether  linkage.  The  most  abundant 
plasmalogen  in  a  tissue  is  usually  the  ethanolamine  derivative. 

Plasmalogens  are  sometimes  named  by  replacing  the  terminal  "yl"  of 
the  phosphatidyl  analog  by  "al,"  so  that  we  have  phosphatidal  ethanol- 
amine, phosphatidal  choline,  etc.  This  method,  however,  is  subject  to 
error  due  to  the  close  similarity  in  the  names  and  has  not,  therefore, 
found  universal  acceptance.  It  is  preferable  when  referring  to  plasma- 


CLASSIFICATION,  NOMENCLATURE  73 


n     H2CtO-CH=CHiR, 


R2  -C-O-CH 


H2C-0-P-0-CH2-CH2-NH2 
0 

ethanolamine  plasmalogen 

logens  to  use  either  "ethanolamine  plasmalogen"  or  "vinyl  ether  phos- 
phatidyl ethanolamine."  These  terms  clearly  distinguish  the  plasma- 
logens  from  the  diacyl  phosphoglyceride.  Still  another  type  of  phospho- 
glyceride occurs.  This  is  the  relatively  rare  alkoxy  glycerophosphatide 
in  which  the  a  ester  linkage  is  replaced  by  an  ether  linkage. 

0     HaC-O-R, 
R2 -C-O-CH     0 

H2C-0-P-0-CH2-CH2-NH2 
OH 

alkoxy  phosphatidyl  ethanolamine 

Phosphonolipids 

Some  years  ago  an  entirely  new  group  of  phosphorus-containing 
lipids  was  discovered  by  two  groups  of  investigators,  one  studying  the 
lipids  of  the  sea  anemone,  Anthopleura  elegantissima  (6),  the  other 
studying  the  proteolipid  fraction  of  ciliate  protozoa  of  sheep  rumen  (7). 
This  group  of  lipids  contains  phosphonic  acid  in  place  of  phosphoric 
acid.  The  first  clues  to  the  existence  of  these  lipids  came  with  the  isola- 

O 
tion  of  2-aminoethylphosphonic  acid    (OH)2  —  P— CH2  —  CH2  —  NH2, 

O 
suggesting  that  this  replaced  2-aminoethylphosphoric  acid  (OH)2  —  P 
—  O  —  CH2  —  CH2  —  NH2  in  some  lipids.  A  typical  glycerol  phosphono- 
lipid  would,  therefore,  have  the  structure: 

0 
H2C-0-C-R, 

0  . 

11 

R2  "C-O-CH 

0 

H2C-0-P-CH2-CH2-NH2 

OH 

dialkyl  glyceryl-(2-aminoethyl)-phosphonate 


j  4  LIPIDS:  DEFINITIONS 

Many  types  of  phosphonolipids  have  now  been  isolated  and  char- 
acterized, including  sphingolipid  and  plasmalogen  classes. 

Phosphonolipids  have  also  been  synthesized;  Baer  and  Stanacev 
(8  9)  have  prepared  glycerol  phosphonolipids  of  the  type  on  page  13 
and  of  a  type  in  which  the  glycerol  is  bound  directly  to  the  phosphorus 
by  a  carbon-phosphorus  bond. 

0 
0     H2C-0-C-R, 

R2-C-0-CH0 

H2C-P-0-CH2-CH2-NH2 

OH 

More  recently  Chacko  and  Hanahan  (10)  have  synthesized  the 
monoether  phosphatidyl  aminoethylphosphonate  and  have  confirmed 
the  presence  of  both  mono-  and  diester  phosphonolipids  in  Tetrahymena 
pyriformis  (aciliate). 

SPHINGOUPIDS 

Sphingolipids  are  compounds  which  on  hydrolysis  yield  sphingosine 
(or  a  closely  related  compound),  derived  lipids,  and  water  soluble 
products.  Unlike  the  complex  lipids  discussed  so  far,  this  group  does 
not  contain  the  glycerol  carbon  skeleton.  Rather,  the  common  link  is 
sphingosine,  an  amino  alcohol  with  the  following  structure: 

3         2         I 

CH3(CH2)I2CH  =  CH-CH-CH-CH 

OH  NH2    OH 

sphingosine 

Sphingosine  carbons  are  numbered  starting  with  the  primary  hy- 
droxyl  group.  The  configuration  of  the  molecule  at  C2  is  D,  and  the 
relationship  of  C3  to  C2  is  erythro.  The  double  bond  has  the  trans  con- 
figuration. The  systematic  name  for  sphingosine  is,  therefore,  D- 
ery^ro-l,3-dihydroxy-2-amino-4-*raw.y-octadecene. 

Molecules  closely  related  to  sphingosine,  and  which  replace  it  m  some 
sphingolipids,  include  dihydrosphingosine  (fully  saturated  sphingosine) 
and  C20  homologues  of  sphingosine. 

Ceramides 

The  simplest  and  most  fundamental  sphingolipids  are  called  ce- 
ramides. In  these  compounds  the  amino  group  of  sphingosine  (or  a 
sphingosine-related  compound)  is  in  an  amide  linkage  with  a  fatty  acid. 
Amide,  ether,  and  vinyl  ether  linkages  are  stable  to  alkali  while  ester 


CLASSIFICATION,  NOMENCLATURE  15 

CH3  (CH2)|2  CH  =  CH-CH-CH-CH2 
OH   NH   OH 
C^O 
R 

ceramide 

linkages  are  readily  broken  by  alkali.  This  difference  is  used  to  ad- 
vantage in  analytical  procedures. 

Sphingomyelin 

Sphingomyelin  is  a  phosphorus-  and  choline-containing  sphingo- 
lipid.  The  primary  alcohol  group  of  a  ceramide  is  joined  via  an  ester 
linkage  to  phosphoric  acid  which  is  in  turn  esterified  with  choline. 
Some  10  percent  of  the  lipid  phosphorus  in  brain  is  sphingomyelin; 
kidney,  spleen,  erythrocytes,  and  plasma  are  also  rich  in  sphingomyelin. 

CH3(CH2),2CH  =  CH-CH-CH-CH2 

•     »     •    9  + 

OH    NH     0-  P-0-CH2-CH2-NT(CH3)3 

•  ■ 

c  =  o     9 

1 

R 

sphingomyelin 

Glycolipids:  Cerebrosides  and  Sulfatides 

In  the  general  group  of  lipids  called  glycolipids,  or  glycosyl  ce- 
ramides,  the  primary  hydroxyl  group  of  a  ceramide  is  linked  by  a  glyco- 
sidic  bond  to  a  monosaccharide  or  oligosaccharide  chain.  A  cerebroside 
is  a  ceramide  linked  to  a  monosaccharide,  usually  galactose.  Cerebro- 
sides are  important  constituents  of  nervous  tissue;  they  are  major  con- 
stituents of  the  myelin  sheath.  Nervous  tissue  cerebrosides  are  of  two 
types:  those  containing  normal  fatty  acids  and  those  containing  2- 
hydroxy  fatty  acids.  A  typical  cerebroside  containing  lignoceric  acid  is 
known  by  the  trivial  name  "kerasin"   (shown  below).  When  the  fatty 


CH3(CH2),2CH=CH-CH-CH-CH2 
OH  NH  6 

C=0 

1 

(CH2)22 
CH, 


ceramide  galactoside  (kerasin) 


7  6  LIPIDS:  DEFINITIONS 

acid  constituent  is  2-hydroxytetracosanoic  (cerebronic)  acid,  the  cere- 
broside  is  called  "phrenosin."  The  cerebrosides  containing  9-tetraco- 
senoic  (nervonic)  and  2-hydroxy-9-tetracosenoic  (oxynervonic)  acids 
are  known  as  "nervon"  and  "oxynervon"  respectively. 

Glycolipids  of  other  types  are  found  in  spleen,  liver,  plasma,  and 
erythrocytes.  These  include  ceramide-dihexosides,  in  which  a  ceramide 
is  linked  to  a  galactose  disaccharide  or  a  glucose-galactose  disaccharide ; 
ceramide-trihexosides;  and  ceramide-tetrahexosides. 

Sulfatides  are  sulfate  esters  of  cerebrosides  in  which  the  sulfate 
group  is  found  on  C3  of  the  galactose  moiety.  Like  the  cerebrosides,  the 
sulfatides  are  abundant  in  nervous  tissues.  These  sulfatides  also  occur 
with  2-hydroxy  fatty  acid  constituents,  but  to  a  lesser  extent  than  do 
nervous  tissue  cerebrosides.  In  other  tissues,  such  as  kidney,  sulfatides 
of  ceramide  dihexosides  have  been  found. 


CH3(CH2)J2CH=CH-CH-CH-CH2 

OH  NH  0 

i 

c=o 

R 


sulfatide  (cerebroside  sulfate) 

Sulfatides   should  not  be   confused  with   sulfolipids,   a   group   of 
plant  lipids  containing  sulfonic  acid  and  having  the  general  formula: 

0 

KLC-O-C-R, 
0        i  OH        H 


\H 

'oh 


CH2 

0=S=0 

I 
OH 


sulfolipid 


Gangliosides 

Gangliosides  are  a  complex  group  of  glycosphingolipids  which  dif- 
fer from  the  other  glycolipids  in  containing  sialic  acids.  Sialic  acids  are 


CLASSIFICATION,  NOMENCLATURE  17 

COOH 

C  =  0 

i 

CH2 
0     H-C-OH 
CH3-C-NH-C-H 
HO-C-H 

H  -C-OH 

1 

H-C-OH 
H2C-0H 
D  (  —  )N-acetylneuraminic  acid 

N-acyl  derivatives  of  neuraminic  acid;  the  common  constituent  of 
gangliosides  is  N-acetylneuraminic  acid. 

High  levels  of  gangliosides  occur  in  nervous  tissue;  they  are  be- 
lieved to  be  located  specifically  in  the  neurons.  Other  gangliosides,  con- 
taining N-glycoylneuraminic  acid,  have  been  found  in  spleen  and  in  red 
blood  cell  stroma. 

The  gangliosides  of  brain  have  been  most  extensively  studied  and 
have  been  separated  into  three  main  types  —  mono-,  di-,  and  trisialo- 
gangliosides  —  according  to  their  sialic  acid  content.  The  major  mono- 
sialoganglioside  has  been  shown  to  have  the  following  structure: 

OH 

CHoOH  _  OH 


/     UHoOH  „  OH,,,   M, 

£343- 


0 
C0'NH/        J    CH2OH 
C02H     ' 


CH2OH  HC"0H 


OH 


I     UM2UM 


CH3CO-NH  HC-OH 

HC-OH 

CH2OH 


A  group  of  relatively  recently  discovered  compounds,  the  prosta- 
glandins, are  also  classified  as  lipids.  The  study  of  these  substances  is 
rapidly  developing  into  a  separate  field,  so  they  are  not  considered  in 
this  text.  A  review  of  the  chemistry  of  the  prostaglandins  is  included 
in  the  list  of  Suggested  Further  Readings  (p.  99). 


78  LIPIDS:  DEFINITIONS 


REFERENCES 


1.  Johnston,  P.  V.,  O.  C.  Johnson,  and  F.  A.  Kummerow   (1957)  /.  Nutrition 
65:  13. 

2.  Perkins,  E.  G.  (1960)  Food  Tech.  14:  508. 

3.  Schultz,  H.  W.,  ed.   (1962)  Symposium  on  Foods:  Lipids  and  Their  Oxida- 
tion. Avi  Publishing  Co.,  Inc.,  Westport,  Conn. 

4.  Hilditch,  T.  P.,  and  P.   N.  Williams   (1964)    The  Chemical  Constitution   of 
Natural  Fats;  4th  ed.,  p.  670.  John  Wiley  &  Sons,  Inc.,  New  York. 

5.  Williams,  J.  H.,  M.  Kuchmak,  and  R.  F.  Witter  (1965)  Lipids  1:  89. 

6.  Kittredge,  J.  S.,  E.  Roberts,  and  D.  G.  Simonsen  (1962)  Biochem.  1:  624. 

7.  Horiguchi,  M.,  and  M.  Kandatsu  (1959)  Nature  184:  901. 

8.  Baer,  E.,  and  N.  Z.  Stanacev  (1964)  /.  Biol.  Chem.  239:  3209. 

9.  (1965)  /.  Biol.  Chem.  240:  44. 

10.  Chacko,  G.  H.,  and  D.  J.  Hanahan  (1969)  Biochim.  Biophys.  Acta  176:   190. 

11.  Kuhn,  R.,  and  H.  Wiegandt  (1963)  Chem.  Ber.  96:  866. 


II .   Preparation  and  Handling  of  Samples 
for  Analysis  of  Lipid  Constituents 


During  the  extraction  of  lipids  from  tissues  and  their  subsequent 
handling  during  analysis  the  lipid  chemist  must  constantly  battle 
his  two  worst  enemies,  oxidation  and  contamination.  We  cannot  over- 
stress  the  need  to  set  up  conditions  in  the  laboratory  that  will  prevent 
oxidation  of  the  lipids  and  intrusion  by  contaminants  (lipid  and  non- 
lipid). It  is  here  that  the  inexperienced  lipid  analyst  most  frequently 
fails.  Before  discussing  extraction  procedures  we  shall  consider  some 
basic  rules  to  follow  when  working  with  lipids. 

GENERAL  TECHNIQUES  IN  LIPID  CHEMISTRY 

Prevention  of  Oxidation 

When  exposed  to  air  the  unsaturated  moieties  of  lipids  rapidly 
oxidize,  a  process  that  is  accelerated  by  light.  It  is  absolutely  essential 
that  the  lipids  be  manipulated  in  an  oxygen- free  environment.  This  is 
generally  achieved  by  carrying  out  operations  under  an  atmosphere  of 
nitrogen.  A  lipid  laboratory  should  have  nitrogen  supplies  at  every  point 
where  lipids  are  to  be  handled.  This  can  be  accomplished  by  running 
several  branch  lines  of  polyethylene  tubing  from  a  tank  (or  tanks)  of 
nitrogen.  The  nitrogen  flow  can  be  regulated  by  constricting  the  tubing 
with  clamps  or  by  inserting  Teflon  stopcocks  or  flow  regulators  at 
suitable  points.  Glass  stopcocks  should  not  be  used,  as  the  grease  from 
them  may  get  blown  down  the  lines  and  contaminate  samples.  Several 
examples  of  when  and  how  to  use  nitrogen  follow: 

1.  When  filtering  a  solution  containing  lipids,  attach  a  line  from 
the  nitrogen  to  the  stem  of  a  funnel  and  invert  over  the  filtration  ap- 
paratus, letting  a  gentle  flow  of  nitrogen  continue  until  filtration  is 
complete. 

2.  When  placing  lipid  samples  in  a  dessicator  to  dry,  first  flush 
out  the  dessicator  with  nitrogen,  evacuate,  and  repeat  the  process.  When 
releasing  the  vacuum  in  the  dessicator,  do  so  by  letting  in  nitrogen,  not 
air.  Any  apparatus  in  which  lipid  is  placed  should  be  fitted  with  three- 
way  stopcocks  so  that  it  can  be  flushed  through  with  nitrogen. 

3.  Other  manipulations  of  the  lipid  can  be  carried  out  under  a  plastic 
"tent"  within  which  a  slight  positive  pressure  of  nitrogen  is  maintained. 
Setups  of  this  type  can  readily  be  made  by  purchasing  plastic  "glove 
bags,"  which  are  available  from  a  number  of  supply  houses. 

4.  When  dealing  with  lipid  dissolved  in  small  volumes  of  solvent, 
it  is  often  convenient  to  remove  the  solvent  under  a  stream  of  nitrogen. 

79 


20  PREPARATION  AND  HANDLING 

For  this  purpose  a  manifold  (a  tube  having  several  outlets)  attached 
to  a  nitrogen  line  is  very  useful.  The  nitrogen  stream  should  be  directed 
through  disposable  glass  pipets,  which  can  be  discarded  after  use  with 
one  sample. 

In  general,  lipid  samples  should  be  blanketed  by  nitrogen  during  all 
operations.  This  is  not  so  necessary  when  the  lipid  is  in  an  atmosphere 
saturated  with  solvent  vapor.  If  lipid  or  tissues  have  to  be  stored  for 
a  while,  they  should  be  frozen  rapidly  and  kept  in  their  containers  at 
— 20°C  (or  below)  in  a  plastic  bag  flushed  out  with  nitrogen. 

In  addition  to  using  a  nitrogen  atmosphere  it  is  often  convenient  to 
add  to  the  lipid  an  antioxidant  such  as  2,6-di-t-butyl-/>-cresol,  more 
commonly  called  butylated  hydroxytoluene  (BHT).  The  usual  addition 
of  BHT  is  0.1  percent  of  the  weight  of  the  lipid. 

Lipids  should  never  be  stored  in  chloroform-methanol  solution  as 
this  leads  to  breakdown  of  phosphoglycerides. 

Additional  hazards  of  breakdown  occur  during  removal  of  tissues 
from  animals  and  during  maceration  of  the  sample.  Here,  lipolytic 
enzymes  may  be  activated  and  break  down  some  of  the  lipids.  It  is, 
therefore,  essential  that  dissection  and  maceration  are  carried  out  as 
rapidly  as  possible. 

Elimination  of  Possible  Contaminants 

Lipids  and  compounds  which  mimic  lipids  in  their  chromatographic 
behavior  are  likely  to  enter  samples  in  a  number  of  ways.  The  only 
way  to  avoid  artifacts  and  contaminants  is  to  ensure  that  all  laboratory 
personnel  abide  by  a  set  of  rigid  rules.  The  following  list  of  suggested 
rules  illustrates  the  vast  number  of  sources  of  contamination. 

1.  All  reagent  grade  solvents  should  be  redistilled  to  remove  non- 
volatile impurities.  Distillation  should  be  carried  out  in  glass  apparatus 
and  solvents  stored  in  glass  containers.  Chloroform  should  be  stabilized 
by  adding  0.25  percent  (by  vol.)  methanol  and  stored  in  a  dark  bottle. 

2.  Corks,  rubber  stoppers,  plastic  film  wraps,  etc.,  should  not  be 
used  to  close  containers  containing  lipid  samples,  especially  when  these 
materials  may  come  in  contact  with  solvent  vapors.  Rubber  tubing 
should  not  be  used.  Contact  of  solvent  with  any  tubing  should  be 
avoided. 

3.  The  use  of  grease  on  stopcocks  and  vacuum  systems  should  be 
avoided.  Teflon  stopcocks  are  the  best  solution.  Good  ground  glass 
apparatus  on  rotary  vacuum  evaporators  and  similar  equipment  will 
usually  produce  good  leakless  seals  without  the  use  of  grease. 

4.  Personnel  should  be  trained  never  to  put  their  fingers  inside 
vessels  to  be  used  for  lipid  work.  Fingerprints  are  a  rich  source  of  lipids. 

5.  Smoking  near  chromatographic  equipment  should  be  forbidden 
as  tobacco  smoke  is  a  rich  source  of  chromatographic  artifacts. 


OF  SAMPLES  FOR  ANALYSIS  21 

6.  Vapors  from  vacuum  pumps  and  other  apparatus  should  be 
vented  into  a  hood. 

7.  If  at  all  possible,  glassware  should  be  washed  in  chromic  acid 
and  rinsed  thoroughly  in  deionized  water. 

8.  Finally,  it  is  good  practice  in  the  lipid  laboratory  to  run  blanks 
of  procedures  to  check  for  possible  artifacts  and  contaminants. 

Rouser  et  al.  (1)  have  demonstrated  the  presence  of  an  impressive 
number  of  potential  contaminants  found  in  a  lipid  laboratory. 

Unwanted  Emulsions  and  Other  Hazards  in  Lipid  Chemistry 

The  beginner  in  lipid  chemistry  encounters  hazards  in  addition  to 
those  of  oxidation  and  contamination.  Invariably  he  will  experience 
the  formation  of  unwanted  and  seemingly  intractable  emulsions.  As  we 
shall  see  later,  procedures  in  lipid  analysis  frequently  call  for  the  par- 
titioning of  lipids  and  nonlipids  between  aqueous  and  organic  sol- 
vent phases.  The  very  property  of  lipids  which  makes  them  important 
in  the  assembly  of  biological  membranes,  namely  their  amphiphilic  na- 
ture, means  that  they  are  frequently  good  emulsifiers.  As  a  consequence, 
the  beginner  finds  that  phases  refuse  to  separate;  often  there  is  a  band 
of  emulsified  material  where  the  interface  should  be,  or  the  whole  sys- 
tem becomes  emulsified.  Emulsions  are  easier  to  prevent  than  to  cure. 
Prevention  is  therefore  emphasized. 

Whenever  a  procedure  calls  for  the  intermixing  of  aqueous  and  non- 
aqueous phases,  this  should  be  achieved  by  swirling  the  contents  of  the 
container  (usually  a  separatory  funnel)  and  inverting  the  vessel  several 
times.  Vigorous  shaking,  especially  when  phospholipids  are  present, 
will  form  emulsions.  Emulsions  also  readily  form  when  soaps  are  pres- 
ent, such  as  when  a  soap  solution  is  being  extracted  with  ether  or  hexane 
to  remove  non-saponifiable  lipids.  Generally,  however,  swirling  and 
inverting  the  vessel  rather  than  shaking  it  vigorously  will  prevent  the 
formation  of  hard-to-break  emulsions. 

If  emulsions  are  formed,  there  are  several  techniques  for  breaking 
them.  Should  the  whole  system  form  a  very  permanent-looking  emul- 
sion, centrifugation  (at  600  to  700  X  g)  will  break  it  most  quickly. 
Other  emulsion-breaking  techniques  are  generally  useless  and  a  waste 
of  time.  If  only  part  of  the  system  is  emulsified  and  the  interface  can 
be  detected,  the  emulsion  can  often  be  dispersed  by  using  a  disposable 
pipet  to  add  a  little  ethanol  at  the  interface.  Adding  a  salt,  such  as 
sodium  sulfate,  and  gently  swirling  will  also  tend  to  break  emulsions. 
Generally,  however,  centrifugation  gives  clean  and  quick  results  and 
is  the  preferred  method. 

Numerous  other  tedious  difficulties  and  potential  dangers  (to  per- 
sonnel as  well  as  to  samples)  are  encountered  in  the  lipid  laboratory. 
In  general  these  are  discussed  in  the  text  as  the  situations  arise.  A 
common   problem   should,   however,   be   mentioned   immediately.    Fre- 


22  PREPARATION  AND  HANDLING 

quently,  a  beginner  complains  that  results  are  not  reproducible.  There 
are  many  possible  reasons  for  this,  but  the  adoption  of  one  simple 
rule  often  solves  the  problem.  Lipids  must  never  be  sampled  from  the 
solid  or  semi-solid  state  unless  efforts  have  been  made  to  homogenize 
the  sample.  Lipids  extracted  from  natural  materials  are  not  homoge- 
neous: they  crystallize  and  solidify  at  different  rates  and  different 
temperatures  so  that  all  parts  of  the  sample  are  not  necessarily  the 
same.  The  sample  can  be  homogenized  by  grinding  it  under  nitrogen 
in  a  mortar,  but  it  is  more  common  and  more  effective  to  dissolve  the 
lipids  in  a  known  volume  of  solvent  and  to  take  aliquots  from  this. 

EXTRACTION  OF  LIPIDS  FROM  VARIOUS  SOURCES 

Samples  from  which  lipids  are  to  be  extracted  should  be  as  fresh 
as  possible.  If  tissues  dissected  from  animals  cannot  be  extracted  imme- 
diately, they  should  be  frozen  by  plunging  them  into  liquid  nitrogen. 
They  should  then  be  stored  at  —  20° C  or  below  in  closed  containers 
that  have  been  flushed  out  with  nitrogen.  Blood  samples  should  not 
be  drawn  unless  it  is  certain  that  they  can  be  treated  immediately. 

It  is  not  possible  to  describe  in  detail  an  extraction  procedure  that 
is  applicable  to  all  types  of  material.  In  changing  from  one  source  to 
another,  some  modification  of  procedure  is  usually  necessary.  Some 
general  remarks,  however,  can  be  made  about  maceration  of  samples. 

If  the  sample  is  no  larger  than  a  few  grams,  it  can  be  ground  most 
efficiently  with  a  tissue  homogenizer  of  the  Potter  Elvehjem  or  Ten- 
broech  type.  These  homogenizers  consist  of  ground  glass  tubes  con- 
taining closely  fitting  pestles.  They  can  be  used  for  grinding  by  hand, 
or  the  pestle  can  be  turned  mechanically  by  attaching  it  to  a  high  speed 
motor.  The  tube  should  be  placed  in  ice  while  grinding.  Larger  samples 
can  be  handled  in  Waring  Blendors,  or  by  using  a  mortar  and  pestle. 

The  choice  of  the  macerating  method  depends  to  a  great  degree  on 
the  toughness  of  the  sample.  When  dealing  with  tough  tissues  such  as 
skin  or  samples  rich  in  connective  tissue,  it  is  best  to  freeze  the 
sample  in  a  steel  mortar  in  liquid  nitrogen.  A  sharp  blow  with  a  steel 
pestle  or  hammer  will  fragment  the  material,  which  can  then  be  ground 
to  a  fine  powder  in  the  frozen  state  and  extracted. 

From  Blood  Serum 

Blood  should  be  drawn  by  venipuncture  or,  in  the  case  of  small 
animals,  by  puncturing  the  heart  or  a  suitable  large  blood  vessel.  The 
blood  should  be  drawn  into  hypodermic  syringes  and  immediately  trans- 
ferred to  centrifuge  tubes.  Centrifugation  (at  550  to  700  X  g)  to  obtain 
the  serum  should  be  carried  out  in  a  refrigerated  centrifuge  at  4°C. 
The  serum  should  be  straw-colored  and  not  show  any  signs  of  hemoly- 


OF  SAMPLES  FOR  ANALYSIS  23 

sis.  Some  investigators  allow  blood  to  clot  before  they  draw  off  the 
serum,  a  process  that  may  take  several  hours.  This  is  not  a  good 
practice,  since  the  lipids  will  oxidize. 

Several  procedures  for  the  extraction  of  lipids  from  serum  have 
been  described  in  the  literature,  all  of  them  similar.  The  one  given 
here  is  a  minor  modification  of  the  method  used  by  Williams  et  al.  (2). 

The  serum  is  extracted  with  chloroform-methanol  using  a  ratio  of 
serum: chloroform: methanol  of  5:6:12  v/v/v.  The  appropriate  vol- 
ume of  methanol  is  placed  in  a  glass-stoppered  Erlenmeyer  flask.  The 
serum  is  then  added  slowly,  with  mixing,  to  the  methanol.  The  chloro- 
form is  added  and  the  flask  is  placed  in  a  water  bath  at  55 °C  for  30 
minutes.  The  solution  is  filtered  immediately  through  a  Buchner  funnel 
and  the  residue  is  washed  with  water: methanol: chloroform  5:6:12 
v/v/v,  using  1ml  of  wash  solution  per  ml  of  original  serum.  The  com- 
bined filtrates  are  allowed  to  separate  into  two  phases  in  a  separatory 
funnel  at  4°C.  The  lower  layer  contains  the  lipid.  The  top  aqueous  layer 
should  be  free  of  lipid ;  this  should  be  checked  by  concentrating  the 
upper  layer,  running  TLC  plates  (see  Chapter  4),  and  looking  for  neu- 
tral lipids  and  phospholipids  (see  p.  45). 

The  solvent  in  the  lower  layer  is  then  removed  under  vacuum.  It  is 
preferable  to  carry  this  out  with  a  good  vacuum  pump  rather  than  a 
water-type  pump ;  this  makes  the  application  of  heat  unnecessary.  If 
this  is  not  possible,  the  heat  should  be  kept  to  a  minimum.  The  evapo- 
rating system  (rotary  type  evaporator,  flash  evaporator,  etc.)  should 
be  fitted  so  that  nitrogen  can  be  introduced  into  the  system  for  con- 
trolling pressure  and  releasing  the  vacuum. 

The  lipid  obtained  should  be  redissolved  in  a  minimum  amount  of 
chloroform-methanol  and  if  necessary  refiltered.  Although  the  serum 
lipid  is  essentially  subjected  to  a  Folch-type  washing  procedure  (3)  in 
the  above  routine,  some  nonlipid  material  may  still  be  carried  through. 
Redissolving  the  sample  in  chloroform-methanol  solution  generally 
eliminates  this. 

The  lipid  sample  is  then  placed  in  a  dessicator  over  potassium  hy- 
droxide or  some  other  dessicant  and  dried  under  vacuum.  As  noted 
earlier,  the  dessicator  should  be  flushed  out  with  nitrogen  and  evacuated 
a  couple  of  times.  When  the  weight  of  lipid  is  known,  an  antioxidant 
such  as  BHT  can  be  added  at  the  level  of  0.1  percent  of  the  weight  of 
lipid.  This  weighing  procedure  is  probably  best  when  only  very  small 
amounts  of  lipids  are  being  handled  and  when  prevention  of  waste  is 
imperative.  There  is  the  danger  that  the  lipid  may  get  exposed  to  air 
during  weighing.  It  is  essential  to  flush  out  containers  with  nitrogen  and 
stopper  them  tightly  during  this  time.  When  wastage  is  not  an  issue, 
the  weighing  procedure  outlined  by  Rouser  et  al.  (4)  is  recommended: 
the  lipid  is  dissolved  in  a  known  volume  of  solvent  and  an  aliquot  (50  to 
200  fx\)  is  removed  and  weighed  on  a  microbalance.  This  procedure  is 


24  PREPARATION  AND  HANDLING 

not  suitable  if  the  lipid  sample  contains  short-chain  volatile  fatty  acids 
or  their  methyl  esters. 

Some  authorities  suggest  adding  the  antioxidant  to  the  extracting 
solvents  so  that  the  lipid  is  protected  throughout  the  extraction  pro- 
cedure. This  is  a  useful  technique  if  one  knows  approximately  the 
amount  of  lipid  that  will  be  extracted  so  that  the  appropriate  amount 
of  antioxidant  can  be  added  to  the  solvents.  This  procedure,  however, 
necessitates  either  the  determination  of  the  amount  of  antioxidant  in 
the  sample  or  the  accurate  measurement  of  the  solvents  in  order  to  ob- 
tain the  weight  of  lipid. 

As  always  with  lipids,  analyses  should  be  completed  as  soon  as  pos- 
sible. Lipids  to  which  antioxidant  has  been  added,  however,  can  be 
stored  in  glass  containers  and  under  nitrogen  at  —  20° C  or  below  with 
little  decomposition  over  fairly  long  periods. 

From  Erythrocytes 

It  is  somewhat  more  difficult  to  extract  the  lipids  from  erythrocytes 
(or  erythrocyte  stroma  preparations)  than  from  serum.  First,  there  is 
a  tendency  for  the  erythrocytes  to  form  a  rubbery  mass  that  is  diffi- 
cult to  break  up,  making  consequent  extraction  inefficient.  This  can 
be  prevented  as  follows.  Place  the  appropriate  amount  of  methanol  in 
any  conventional  tissue  homogenizer,  add  the  cells,  and  homogenize. 
Then  add  the  correct  volume  of  chloroform  and  homogenize  again. 
One  extraction  with  this  solvent  mixture  will  not  remove  the  lipids 
completely.  The  number  of  extractions  and  the  variations  in  the  solvent 
mixture  will  depend  on  the  tissue  involved.  Rouser  et  al.  (4)  have  pro- 
posed an  exhaustive  extraction  sequence  (the  figures  in  parentheses 
refer  to  the  milliliters  of  solvent  per  gram  of  fresh  tissue) : 

1.  Chloroform-methanol,  2:1  v/v,  twice  (20ml,  then  10ml) 

2.  Chloroform-methanol,  1:2  v/v  (10ml) 

3.  Chloroform-methanol,  7:1  saturated  with  28  percent  (by  weight) 
aqueous  ammonia1  (10ml) 

4.  Chloroform-methanol-glacial   acetic   acid,   8:4:3   v/v/v    (10ml) 

5.  Chloroform-methanol-concentrated  hydrochloric  acid,  200:100:1 
v/v/v  (10ml) 

This  complete  extraction  sequence  is  not  required  for  efficient  ex- 
traction from  most  tissues.  In  most  cases  the  first  three  systems  suffice, 
and  frequently  efficient  extraction  is  obtained  by  extraction  with 
chloroform: methanol  2:1  v/v  only.  In  the  case  of  erythrocytes,  extrac- 
tion with  the  first  three  solvents  is  recommended. 

1  Commercial  preparations  of  28%  aqueous  ammonia  usually  contain  non- 
volatile solids,  largely  silicates.  To  avoid  this  contamination,  ammonia  can  be 
bubbled  from  a  cylinder  into  ice-cold  distilled  water  in  a  plastic  container,  in 
which  the  solution  is  then  stored. 


OF  SAMPLES  FOR  ANALYSIS  25 

The  combined  solvent  extracts  will  contain  nonlipid  material.  This 
can  be  removed  using  the  Folch  washing  procedure  (3).  In  this  method 
0.2  volumes  of  water  (or  a  salt  solution)  is  added  to  the  combined  sol- 
vents in  a  separatory  funnel  and  the  nonlipid  material  passes  into  the 
aqueous  phase.  This  procedure  is  widely  used  but  is  not  entirely  satis- 
factory since  some  loss  of  lipid  occurs.  As  an  alternative,  most  nonlipid 
material  can  be  removed  by  one  of  the  other  methods  described  below. 

The  combined  solvents  freed  of  nonlipids  and  containing  the  erythro- 
cyte lipids  are  removed  under  vacuum  as  previously  described,  and  the 
lipid  sample  is  dried  and  weighed. 

When  working  with  erythrocytes,  fresh  cells  must  be  used,  since 
in  aging  erythrocytes  the  lipids  are  rapidly  oxidized  by  the  catalytic 
effect  of  heme.  This  is  also  the  reason  for  removing  nonlipid  material 
from  the  lipid  sample  and  adding  an  antioxidant. 

From  Brain 

Brain  can  be  efficiently  extracted  with  chloroform-methanol  2:1 
v/v.  Fresh  or  frozen  brain  should  be  placed  in  a  suitable  homogenizer  or 
mortar  and  extracted  three  times  with  chloroform-methanol  2:1  v/v, 
using  20ml  per  gram  of  brain  the  first  time  and  then  10ml  per  gram 
the  remaining  two  times. 

Brain  contains  many  gangliosides  that  partition  mainly  into  the 
top  (aqueous)  phase  in  the  Folch  washing  procedure.  Some  ganglio- 
sides usually  remain  in  the  bottom  phase,  however,  and  any  gangliosides 
entering  the  top  phase  may  carry  other  lipid  with  them.  The  method  is, 
therefore,  not  reproducible  and  if  a  total  extract  of  brain  lipids  is  re- 
quired, aqueous  washing  should  not  be  used. 

Redissolving  the  lipid  after  removing  the  solvent  is  another  way  of 
removing  nonlipid  material.  Provided  that  the  solvent  is  removed  at  a 
low  temperature  and  two  phases  exist  during  evaporation,  any  carried- 
over  protein  will  be  denatured.  The  solvents  containing  the  extracted 
lipids  plus  nonlipid  contaminants  should  be  placed  in  a  suitable  flask 
for  the  evaporator  system  being  used.  If  two  phases  are  not  present, 
add  water  until  two  phases  are  formed.  Remove  the  solvents  under 
vacuum,  applying  just  enough  heat  via  a  water  bath  to  prevent  the 
formation  of  ice  on  the  outside  of  the  flask.  The  dried  crude  extract 
obtained  can  then  be  re-extracted  three  times  with  chloroform-meth- 
anol 2:1  v/v  (1ml  per  lOOmg  of  lipid)  and  the  insoluble  residue  re- 
moved by  filtration  through  a  sintered  glass  filter  (medium).  Re- 
extraction  and  filtration  can  be  carried  out  at  room  temperature. 

Nonlipid  material  can  also  be  removed  by  one  of  the  column  chroma- 
tographic procedures  described  later  (pp.  27-29).  When  the  final  solvent 
is  removed  and  the  lipid  dried,  great  caution  must  be  used  in  subsequent 
handling.  Brain  lipid  is  rich  in  highly  unsaturated   (penta-  and  hexa- 


26  PREPARATION  AND  HANDLING 

enoic)    fatty  acids,  which  oxidize  rapidly.   An  antioxidant  should  be 
used,  and  the  lipid  must  be  carefully  protected  from  air. 

From  Other  Sources 

The  extraction  procedure  employed  for  brain  is  suitable  for  ex- 
tracting lipids  from  many  soft  tissues  such  as  liver,  spleen,  kidney, 
adipose  tissue,  etc.  With  other  tissue,  however,  individual  problems 
are  encountered  and  modifications  must  be  made.  Nerves,  skin,  heart, 
and  muscle  are  not  easily  homogenized  in  the  ground  glass  homoge- 
nizers  of  the  Potter-Elvehjem  type.  Special  handling  such  as  grinding  in 
a  mortar  or  cutting  up  in  a  Waring  Blendor  may  be  necessary.  The 
most  useful  technique  with  many  tougher  tissues  is  to  grind  the  solidly 
frozen  tissue  to  a  powder  as  previously  described  (p.  22).  Plant  sources, 
particularly  some  leaves  and  seeds,  frequently  present  similar  problems 
and  more  severe  grinding  procedures  must  be  employed. 

If  no  established  extraction  procedure  is  available  for  a  particular 
material,  it  should  be  subjected  to  the  exhaustive  extraction  sequence 
previously  described.  Each  solvent  system  should  be  evaporated  off 
separately  in  order  to  determine  how  much  lipid  it  extracted.  In  any 
event,  solvent  systems  containing  acid  should  be  evaporated  separately 
as  the  presence  of  acid  may  hydrolyse  labile  lipids.  Finally,  it  may  be 
necessary  to  subject  the  homogenized  material  to  acid  or  alkaline  hy- 
drolysis and  to  check  the  hydrolysate  for  the  presence  of  fatty  acids. 
This  will  ascertain  if  any  very  tightly  bound  lipid  remains  unextracted. 
For  example,  the  fatty  acids  in  feces  are  present  in  the  form  of  soaps ; 
it  is  therefore  necessary  to  treat  feces  with  dilute  (10  to  20  percent) 
hydrochloric  acid  before  extracting.  Problems  of  a  similar  nature  are 
associated  with  some  microbial  sources  in  which  fatty  acids  are  found 
to  be  associated  with  glycoproteins.  These  sources  of  lipid  constitute 
separate  problems  in  themselves  and,  as  special  cases,  are  beyond  the 
scope  of  this  book. 

REMOVAL  OF  NONLIPID  CONTAMINANTS  FROM  EXTRACTS 

Aqueous  Washing 

As  noted  earlier,  hazards  are  associated  with  this  procedure,  and  in 
certain  cases,  for  instance  when  total  brain  lipid  is  required,  its  use  is 
unwise.  Although  simpler,  more  reliable  methods  are  now  available, 
aqueous  washing  still  enjoys  wide  acceptance,  however,  and  objections 
to  it  are  probably  somewhat  overstated.  The  method  as  described  here 
applies  to  the  combined  2:1  v/v  chloroform-methanol  extracts. 

In  this  procedure  the  crude  chloroform-methanol  extract  is  washed 
with  0.2  its  volume  of  water  or  a  suitable  salt  solution.  It  is  necessary 
to  prepare  solutions  known  as  "pure  solvents  upper  phase"  and  "pure 
solvents   lower   phase."   These    are    prepared    by   mixing   chloroform, 


OF  SAMPLES  FOR  ANALYSIS  27 

methanol,  and  water  (8:4:3  v/v/v)  in  a  separatory  funnel.  On  standing, 
two  phases  separate.  These  are  collected  and  stored  in  glass  bottles. 
The  upper  phase  consists  of  chloroform-methanol-water,  approximately 
30:48:47  v/v/v,  and  in  the  lower  phase  the  proportions  are  86:14:1 
v/v/v.  The  phases  may  be  prepared  by  using  the  above  proportions  if 
preferred.  A  "pure  solvents  upper  phase"  that  contains  0.02  percent 
CaCl2,  0.017  percent  MgCl2,  0.29  percent  NaCl,  or  0.37  percent  KC1 
serves  as  the  salt  solution  wash.  The  salt  solutions  may  be  prepared 
either  by  shaking  the  correct  amount  of  salt  with  "pure  solvents  upper 
phase"  or  by  replacing  the  water  during  the  preparation  of  "pure 
solvents  upper  phase"  with  the  following  aqueous  solutions:  0.04  per- 
cent CaCl2,  0.034  percent  MgCl2,  0.58  percent  NaCl,  or  0.74  percent 
KG.  Using  a  salt  solution  rather  than  pure  water  reduces  loss  of  lipids 
into  the  upper  phase.  Usually  a  crude  lipid  extract  already  contains 
salts,  which  helps  conditions  somewhat,  but  additional  salts  may  im- 
prove the  distribution  required.  Gangliosides,  for  example,  found 
mainly  in  the  upper  phase  in  all  systems,  will  be  almost  entirely  elimi- 
nated from  the  lower  phase  by  a  CaCl2  wash.  This  wash  should  not  be 
used,  however,  if  insoluble  calcium  salts  are  likely  to  be  formed. 

The  washing  procedure  is  the  same  for  both  the  aqueous  and  the  salt 
wash.  Washing  is  carried  out  in  a  separatory  funnel  if  the  phases  are 
to  be  allowed  to  separate  by  standing,  or  in  a  centrifuge  tube  if  phase 
separation  is  to  be  achieved  more  rapidly  by  centrifugation  (at  approxi- 
mately 600  X  g). 

"The  crude  extract  is  mixed  thoroughly  with  0.2  its  volume  of  water 
or  one  of  the  salt  solutions.  When  the  biphasic  system  is  obtained,  as 
much  of  the  upper  phase  as  possible  is  removed  by  siphoning.  The  inter- 
face is  then  rinsed  three  times  with  small  amounts  of  "pure  solvents 
upper  phase."  The  rinsing  should  be  done  carefully  so  as  to  avoid  dis- 
turbing the  lower  phase.  After  the  last  rinse,  the  lower  phase  and  any 
unremoved  rinse  solution  are  combined  into  one  phase  by  adding  meth- 
anol. The  solution  can  then  be  diluted  with  methanol  to  any  desired 
volume,  or  the  solvent  can  be  removed  under  vacuum  and  the  lipid  dried. 

Treatment  on  Cellulose  and  Sephadex  Columns 

Rouser  et  al.  (5)  describe  a  cellulose  column  chromatographic  pro- 
cedure which  removes  water-soluble  nonlipid  contaminants  from  crude 
lipid  extracts.  Gangliosides  are  eluted  with  the  nonlipid  fraction. 

The  column  size  chosen  will  depend  on  sample  size.  The  procedure 
described  below  is  suitable  for  a  100  to  500mg  sample. 

A  glass  column  2.5cm  internal  diameter  (i.d.)  X  20  to  30cm  long  is 
packed  with  20g  of  Whatman  standard  grade  ashless  cellulose  powder 
suspended  in  methanol- water  1:1  v/v  (see  p.  31  for  hints  on  packing 
glass  columns).  The  packed  bed  is  washed  at  a  flow  rate  of  3  ml/min 
with  the  following  set  of  solvents:  methanol-water  1:1  v/v  (7  column 


28  PREPARATION  AND  HANDLING 

volumes2),  chloroform-methanol  1:1  v/v  (3  column  volumes),  and 
chloroform-methanol  9:1  v/v  saturated  with  water  (4  column  volumes). 

The  sample,  100  to  500mg,  is  added  to  the  column  in  5  to  10ml  of 
the  last  washing  solution.  Two  fractions  are  then  eluted:  the  first,  with 
chloroform-methanol  9:1  v/v  saturated  with  water  (20  column  vol- 
umes), contains  lipids  minus  the  gangliosides,  if  present;  the  water- 
soluble  nonlipid  contaminants  and  the  gangliosides  are  then  eluted  with 
12  column  volumes  of  methanol-water  9: 1  v/v. 

Sephadex,  a  cross-linked  dextran  gel,  can  also  be  used  to  separate 
lipids  and  water-soluble  nonlipids.  Procedures  have  been  described  by 
Wells  and  Dittmer  (6)  and  by  Siakotos  and  Rouser  (7).  The  latter 
method  is  described  below. 

Four  solvent  systems  are  required  (all  proportions  are  by  volume) : 

1.  Chloroform-methanol  19:1,  saturated  with  water 

2.  850ml  chloroform-methanol  19:1  plus  170ml  glacial  acetic  acid 
plus  about  25ml  water  (to  saturate) 

3.  850ml  chloroform-methanol  9:1  plus  170ml  glacial  acetic  acid 
plus  about  42ml  water  (to  saturate) 

4.  Methanol-water  1 : 1 

When  preparing  solvent  mixtures  1,  2,  and  3,  shake  them  vigor- 
ously and  allow  them  to  separate  in  a  separatory  funnel.  The  lower 
phase  of  each  is  used  for  chromatography.  When  adding  the  water  to 
mixtures  1,  2,  and  3,  do  so  slowly  so  that  the  mixtures  are  slightly 
undersaturated. 

Sephadex  (G-25,  coarse,  beaded,  Pharmacia  Fine  Chemicals,  New 
Jersey)  is  placed  in  methanol-water  1:1  v/v.  The  "fines"  in  the  material 
are  removed  by  decantation  and  dissolved  gases  are  removed  by  gentle 
suction  from  a  vacuum  pump.  This  takes  about  1  minute.  After 
equilibrating  at  room  temperature  for  several  hours,  the  gel  is  again 
degassed  and  poured  into  a  suitably  sized  column.  For  samples  of  50  to 
250mg,  a  column  1cm  i.d.  X  10  to  30cm  is  appropriate.  A  packed 
2.5cm  i.d.  X  30cm  column  will  accommodate  several  grams  of  sample. 
To  prevent  the  gel  from  floating  in  the  solvents  add  a  2.5  to  5cm  layer 
of  clean  sand  above  it. 

Transfer  the  sample  onto  the  column  in  solvent  mixture  1  and  cover 
the  top  of  the  sand  with  a  plug  of  washed  glass  wool.  For  the  smaller 
samples  on  the  1cm  i.d.  X  10cm  columns  elute  with  the  following  vol- 
umes of  the  solvents:  1,  25ml;  2,  50ml;  3,  25ml;  and  4,  50ml.  The  flow 
rate  should  be  3ml  per  minute.  Larger  samples  up  to  5g  on  the  2.5cm 
i.d.  X  30cm  columns  should  be  eluted  with  20  times  the  above  volumes. 


2  A  column  volume  (sometimes  called  the  bed  volume)  is  the  volume  of  the 
column  occupied  by  the  adsorbent. 


OF  SAMPLES  FOR  ANALYSIS  29 

Depending  on  the  complexity  of  the  lipid-nonlipid  mixture,  the  four 
fractions  may  contain  a  variety  of  substances.  The  following  is  a  com- 
prehensive list  of  the  substances  found  after  chromatography  of 
samples  from  various  sources  (5). 

Fraction  1:  Hydrocarbons;  mono-,  di-,  and  triglycerides;  sterols; 
sterol  esters;  waxes  ;  all  phosphoglycerides  (including  lyso  compounds)  ; 
sulfatides;  cerebrosides;  sulfolipids;  free  fatty  acids;  glycosyldiglycer- 
ides;  unconjugated  bile  acids;  conjugated  bile  acids  (glycine,  taurine)  ; 
and  uncharacterized  nonlipid  compounds.  The  unconjugated  bile  acids 
are  eluted  whether  they  are  applied  as  salts  or  free  acids  and  the 
conjugated  when  applied  in  the  free  acid  form  (4).  The  bile  acids  and 
related  steroids  are  not  covered  in  this  text;  any  major  biochemical 
text  (such  as  8)  gives  accounts  of  the  steroids. 

Fraction  2:  Gangliosides;  glycine  conjugated  bile  acids  (applied  as 
salts)  ;  some  acidic  phosphatides  (in  "altered"  form)  ;  urea;  and  other 
materials  soluble  in  organic  solvents. 

Fraction  3:  Traces  of  gangliosides;  taurine-conjugated  dihydroxy- 
cholanic  (deoxy  and  chenodeoxy)  acids  (when  applied  as  salts);  some 
amino  acids;  and  other  uncharacterized  organic  substances. 

Fraction  4:  Taurocholate  and  most  water  soluble  nonlipids  such  as 
salts,  amino  acids,  sugars,  etc. 

Note  that,  for  the  purposes  of  the  types  of  analyses  covered  in  this 
text,  Fraction  1  contains  the  bulk  of  the  lipids,  while  Fraction  2  is  rich 
in  gangliosides. 

This  procedure  is  the  most  efficient  and  reliable  method  now  avail- 
able for  the  removal  of  water-soluble  nonlipids  from  crude  lipid  ex- 
tracts. It  is  also  relatively  simple  to  set  up  and  use.  Columns  can  be 
reused  after  allowing  them  to  stand  in  methanol-water  1:1  (solvent 
mixture  4)  for  about  48  hours,  then  washing  with  100  to  500ml  of  sol- 
vent mixture  1  immediately  before  use. 

REFERENCES 

1.  Rouser,  G.,  G.  Kritchevsky,  M.  Whatley,  and  C.  F.  Baxter  (1965)  Lipids 
1 :  107. 

2.  Williams,  J.  H.,  M.  Kuchmak,  and  R.  F.  Witter  (1965)  Lipids  1:  89. 

3.  Folch,  J.,  M.  Lees,  and  G.  H.  Sloane-Stanley  (1957)  /.  Biol.  Chem.  226:  497. 

4.  Rouser,  G.,  G.  Kritchevsky,  and  A.  Yamamoto  (1967)  in  Lipid  Chromato- 
graphic Analysis,  ed.  G.  V.  Marinetti ;  vol.  1,  p.  99.  Marcel  Dekker,  Inc., 
New  York. 

5.  Rouser,  G.,  C.  Galli,  and  G.  Kritchevsky  (1965)  /.  Am.  Oil  Chem.  Soc.  42: 
404. 

6.  Wells,  M.  A.,  and  J.  C.  Dittmer  (1963)  Biochem.  2:  1259. 

7.  Siakotos,  A.  N.,  and  G.  Rouser  (1965)  /.  Am.  Oil  Chem.  Soc.  42:  913. 

8.  Mahler,  H.  R.,  and  E.  H.  Cordes  (1966)  Biological  Chemistry.  Harper  & 
Row,  New  York. 


III.   Column  Chromatography 


The  chromatographic  technique  has  been  used  by  organic 
chemists  as  a  separation  method  for  many  years.  Early  develop- 
ments were  confined  to  column  and  paper  chromatography  and  the 
types  of  separations  achieved  were  limited.  However,  in  1952  the  first 
gas-liquid  chromatograph  (1)  was  introduced,  and  in  1958  Stahl  (2) 
described  his  development  of  thin-layer  chromatography.  Rapid  devel- 
opment of  these  two  techniques,  together  with  improved  applications 
of  column  chromatography,  has  revolutionized  the  field  of  lipid 
analysis.  Xew  and  improved  methods  are  still  being  reported  in  some 
abundance,  from  which  we  have  selected  a  few  well-tested  procedures 
in  each  area  of  chromatography.  These  can  be  applied  with  good  re- 
producibility to  the  analysis  of  mixtures  of  neutral,  phospho-,  and 
glycolipids.  Separations  of  the  rarer  lipids  and  of  complex  lipids  such  as 
gangliosides  still  present  some  difficulties,  as  does  the  separation  of 
individual  molecular  species  of  lipid  classes.  The  major  lipids  of  blood 
serum,  say,  can  be  analyzed,  including  their  fatty  acid  and  fatty  alcohol 
patterns,  provided  that  the  indicated  procedures  are  followed  strictly 
and  the  lipids  are  carefully  protected  from  oxidation  and  contamination. 
This  introduction  to  chromatographic  techniques  is  geared  to  that  level 
of  achievement.  It  should,  however,  be  stressed  that  in  many  respects 
chromatography  is  very  much  an  art,  requiring  practice  before  effective 
separations  can  be  obtained. 

Chromatography  includes  any  technique  in  which  compounds  are 
physically  separated  by  differential  distribution  between  two  phases, 
one  of  the  phases  being  stationary  and  the  other  moving.  Stationary 
phases  are  either  solid  or  liquid  and  moving  phases  either  liquid  or 
gaseous. 

In  column  chromatography  the  stationary  phase  is  packed  in  a  glass 
column  onto  which  the  sample  is  introduced ;  the  moving  phase  is 
liquid.  Separation  is  achieved  by  percolating  suitably  constituted  sol- 
vents through  the  bed  of  stationary  phase.  Column  chromatography 
can  be  subdivided  into  two  main  types  according  to  the  nature  of  the 
stationary  phase:  solid-liquid  adsorption  chromatography  and  liquid- 
liquid  partition  chromatography. 

SOLID-LIQUID  ADSORPTION  CHROMATOGRAPHY 

This  type  of  chromatography  is  based  on  the  differences  in  affinity 
of  compounds  for  the  solid  adsorbent  that  serves  as  the  stationary  phase. 
The  relative  affinity  of  compounds  for  the  phase  depends  upon  the  na- 
ture of  their  polar  groups  and  to  some  extent  upon  the  van  der  Waals 
forces  exerted  by  their  nonpolar  groups.  The  greatest  forces  involved 

30 


COLUMN  CHROMATOGRAPHY  31 

in  the  affinity  for  the  adsorbent  are  those  due  to  polar  and  ionic  groups. 
This  means  that  this  type  of  chromatography  is  useful  for  separating 
lipid  mixtures  which  differ  in  the  number  and  type  of  their  polar  groups. 
Thus,  broad  separations  of  the  neutral  lipids  from  the  main  types  of 
polar  lipids  can  readily  be  carried  out.  Separations  within  lipid  classes 
can  also  be  achieved.  Many  adsorbents  have  been  employed,  among 
them  alumina,  magnesium  oxide,  urea,  Florisil  (a  magnesia  silica  gel), 
silicic  acid,  diethylaminoethyl  cellulose  (DEAE),  and  others.  The  most 
efficient  and  versatile  separations  to  date  have  been  obtained  using 
DEAE.  Silicic  acid  is,  however,  very  useful  for  separation  of  lipid 
classes,  and  Florisil  is  especially  useful  for  the  separation  of  cerebro- 
sides.  The  procedures  given  utilize  these  three  main  adsorbents. 

A  typical  adsorption  chromatography  set-up  is  shown  in  Figure  1. 
This  is  the  simplest  form  of  apparatus  which  should  be  employed.  Note 
the  Teflon  stopcocks  (to  avoid  use  of  grease).  Also  note  the  nitrogen 
line  so  that  a  slight  positive  pressure  of  nitrogen  can  be  maintained  over 
the  system. 

LIQUID-LIQUID  PARTITION  CHROMATOGRAPHY 

Liquid-liquid  partition  chromatography  is  essentially  a  counter- 
current  distribution  process.  In  this  method,  separation  depends  on  the 
relative  solubility  characteristics  of  the  compounds  with  respect  to  the 
stationary  and  moving  phases  of  the  column.  Two  types  of  liquid-liquid 
columns  are  in  general  use,  one  in  which  the  more  polar  liquid  is  station- 
ary (partition  type)  and  the  other  in  which  the  less  polar  liquid  is 
stationary  (reversed-phase  partition  type).  Liquid-liquid  partition  chro- 
matography has  been  used  largely  for  the  separation  of  fatty  acids. 
Separations  have  been  obtained  according  to  unsaturation,  chain  length, 
and  geometrical  isomerism.  One  of  the  major  uses  of  this  method  (es- 
pecially when  preparative  gas  chromatography  is  not  available)  is  the 
large-scale  preparative  separation  of  fatty  acids. 

We  shall  be  concerned  only  with  adsorption  chromatography.  Before 
describing  some  separations  it  is  essential  that  we  consider  the  basic 
principles  of  column  packing  and  preparation. 

PREPARATION  OF  COLUMNS 

Separation  and  reproducibility  depend  on  a  number  of  factors,  the 

most  important  of  which  are  packing  of  the  column,  pretreatment  of  the 

;  adsorbent,  shape  and  dimensions  of  the  column,  rate  of  elution,  quality 

I  of  the  eluting  solvents,  and  load  of  sample  in  relation  to  weight  of 

adsorbent.   Temperature   and   humidity  also   influence   reproducibility. 

It  is  usually  best  to  introduce  the  adsorbent  into  the  column  in  the 

form  of  a  thin  slurry  or  suspension.  The  slurry  is  prepared  by  adding 

the  desired  weight  of  packing  material  to  a  suitable  volume  of  solvent. 

The  solvent  used  is  generally  the  one  with  which  the  adsorbent  is  to  be 


32 


COLUMN  CHROMATOGRAPHY 

nitrogen  line 


paper  disc 
or  sand 


receiver 


solvent  reservoir 


—  Teflon  stopcock 


solvent  (moving  phase) 


column  packing 
(stationary  phase) 


glass  wool 


Teflon  stopcock  'H  f""^ 


eluate 


Apparatus  for  adsorption  column  chromatography. 


(Fig.  1 


COLUMN  CHROMATOGRAPHY  33 

prewashed  or  pretreated;  in  some  instances,  the  first  eluting  solvent 
may  be  used.  A  plug  of  glass  wool  should  first  be  placed  in  the  shoulder 
of  the  column  (Fig.  1).  As  the  adsorbent  is  poured  in  and  allowed  to 
settle,  the  flow  of  solvent  through  the  column  is  kept  constant.  This  pro- 
cedure prevents  the  formation  of  "channels"  in  the  column,  a  common 
hazard  when  columns  are  packed  dry.  Channel  formation  is  quite  dis- 
astrous in  column  chromatography,  since  it  leads  to  very  inefficient 
separation  and  overlapping  of  fractions.  Applying  pressure  to  pack 
columns  is  a  dangerous  practice  and  generally  does  not  prevent  channel- 
ing. If  pressure  is  applied  to  a  glass  column,  which  is  hardly  designed 
as  a  pressure  vessel,  the  glass  may  break  and  possibly  cause  injuries. 

Pretreatment  of  the  adsorbent  varies  according  to  the  adsorbent 
being  used.  In  the  examples  which  follow,  therefore,  pretreatment  is, 
dealt  with  individually.  It  is  essential  that  a  recommended  pretreatment 
is  carried  out,  as  frequently  this  step  endows  the  adsorbent  with  its 
particular  characteristics. 

The  shape  and  length  of  columns  affect  separation;  column  dimen- 
sions, therefore,  should  always  be  stated.  It  has  been  demonstrated  that 
longer,  narrower  columns  usually  give  better  separations  than  shorter, 
wider  ones  (3).  If  a  column  is  tried  and  it  is  found  that  some  fractions 
overlap,  increasing  the  column  length  often  solves  the  problem.  Ex- 
tremely long  columns  are,  of  course,  inconvenient;  increasing  the  length 
beyond  40cm  is  not  recommended. 

Once  elution  is  started,  a  flow  of  nitrogen  through  the  column 
(preferably  via  a  proper  flow  control)  can  be  maintained  as  an  anti- 
oxidation  precaution. 

While  investigators  invariably  recommend  a  particular  sample  load 
for  best  resolution,  the  same  load  may  not  be  optimal  in  all  laboratories. 
Slight  differences  in  the  adsorbent  as  well  as  in  environmental  factors 
such  as  laboratory  temperature  and  humidity  may  affect  resolution. 
Therefore,  even  though  a  method  may  be  well  established,  it  may  be 
necessary  to  change  some  dimensions  of  the  procedure  if  resolution  is 
not  satisfactory.  This  is  done  by  applying  different  loads  to  a  particular 
column  until  the  best  resolution  is  obtained.  Ideally,  and  in  the  interests 
of  saving  time,  one  should  strive  to  ascertain  the  maximum  load  that 
can  be  applied  to  the  smallest  column  (that  is,  the  column  with  the 
smallest  elution  volume)  to  give  satisfactory  resolution  of  fractions. 
The  composition  of  fractions  and  the  degree  of  resolution  obtained  are 
very  readily  checked  by  thin-layer  chromatography.  This  technique  and 
its  use  as  an  adjunct  to  column  chromatography  are  discussed  in  the 
next  chapter. 

The  quantitative  analysis  of  samples  by  column  chromatography  can 
be  accomplished  in  a  number  of  ways,  depending  upon  the  composition 
of  the  sample  and  the  needs  of  the  investigator.  It  is  usual  to  analyze 
each  tube  of  eluate  for  a  suitable  element  or  functional  group  and  to 


34  COLUMN  CHROMATOGRAPHY 

plot  its  concentration  versus  tube  number.  Each  tube  should  contain  a 
constant  volume  of  eluate  (usually  5  or  10ml),  which  may  be  collected 
manually  or  by  one  of  the  many  available  automatic  fraction  collectors. 
In  making  a  preliminary  separation  of,  say,  cholesterol  and  glycolipids 
from  phospholipids,  each  tube  would  be  analyzed  for  phosphorus. 

Once  the  elution  pattern  has  been  established,  an  appropriate  large 
volume  of  eluate  may  be  collected,  concentrated  to  a  known  volume, 
and  checked  by  thin-layer  chromatography  for  composition.  A  known 
aliquot  can  then  be  analyzed  for  the  appropriate  element  or  compound 
and  the  total  amount  in  the  fraction  calculated.  Contents  of  each  lipid 
can  be  reported  in  a  variety  of  ways,  such  as  mg  lipid  phosphorus  per  g 
of  total  lipid,  or  per  g  of  wet  —  or  dry  —  tissue.  Examples  of  the  cal- 
culation of  such  expressions  are  given  in  the  final  chapter.  Sometimes 
merely  a  weight  of  a  fraction  may  be  required ;  this  may  be  done  di- 
rectly by  evaporating  the  solvents  to  a  known  volume,  taking  an  aliquot, 
removing  the  solvent,  and  weighing  on  a  microbalance.  Thin-layer 
chromatographic  checks  of  fraction  content  should  still  be  made  for 
every  run. 

Silicic  Acid  Columns 

For  some  time  silicic  acid  and  alumina  were  the  only  adsorbents  used 
for  the  chromatography  of  lipids.  Both  adsorbents  have  to  some  extent 
been  supplanted  by  others,  but  silicic  acid  is  still  used,  especially  for 
initial  broad  separations.  Fine-degree  resolution  requires  rechroma- 
tography  on  other  adsorbents. 

Early  separations  on  silicic  acid  were  done  using  fine  mesh  prepara- 
tions. As  a  consequence,  flow  rates  were  very  slow  and  the  procedures 
time-consuming.  Reproducibility  of  results  on  silicic  acid  columns  is 
not  easily  achieved  since  different  batches  of  the  adsorbent  may  vary 
considerably  in  their  properties.  To  some  extent,  these  disadvantages 
can  be  overcome  by  using  one  of  the  commercially  available  silicic  acids 
that  have  been  manufactured  with  a  view  to  maintaining  a  uniform 
standard  grade  (for  example,  Adsorbosil,  Applied  Science  Laboratories, 
and  Unisil,  Clarkson  Chemical  Company).  Using  these  preparations 
in  coarser  mesh  sizes  overcomes  the  problem  of  slow  flow  rates.  Using 
standardized  silicic  acid  preparations  also  gives  better  reproducibility, 
although  reproducibility  between  one  laboratory  and  another  remains 
difficult.  The  procedures  that  follow  are,  therefore,  generalized.  Each 
investigator  must  standardize  procedures  according  to  his  individual 
needs,  and  each  lot  of  adsorbent  must  be  separately  assessed. 

Usually  the  better  grades  of  silicic  acid  do  not  require  pretreatment, 
but  they  should  be  activated  at  120° C  if  they  have  been  in  a  moist 
atmosphere.  Activation  is  necessary  to  remove  some  of  the  absorbed 
moisture,  which  could  affect  the  adsorption  properties  of  the  silicic 
acids. 


COLUMN  CHROMATOGRAPHY  35 

The  column  in  general  use  is  2.5cm  i.d.  and  10  to  20cm  long;  the 
sample  loading  for  separations  of  neutral  from  phospholipid  classes  is 
about  10  to  30mg  per  g  adsorbent. 

The  mechanisms  involved  in  silicic  acid  chromatography  and  the 
elution  patterns  of  lipids  on  this  adsorbent  have  been  discussed  in  great 
detail  by  Wren  (4).  Wren  gives  an  expanded  version  of  Trappe's 
"eluotropic  series"  (solvents  listed  in  order  of  their  eluting  power). 
The  solvents  as  listed  by  Wren  are:  methanol;  ethanol;  1-propanol; 
acetone;  methyl  acetate;  ethyl  acetate;  ether;  dichloromethane;  ben- 
zene; toluene;  1,1-dichloroethane ;  1,1,2,2-tetrachloroethane;  chloro- 
form; trichloroethylene;  carbon  tetrachloride;  cyclohexane ;  and 
petroleum  ether  of  various  boiling  ranges.  Of  these  solvents  those  most 
frequently  used  are  methanol,  ether,  chloroform,  and  petroleum  ether. 
Wren  (4)  also  listed  the  approximate  order  in  which  one  may  expect 
lipids  to  be  eluted  from  a  silicic  acid  column:  hydrocarbons,  esters 
(other  than  sterol  esters  and  glycerides),  sterol  esters,  fatty  aldehydes, 
triglycerides,  long-chain  alcohols,  fatty  acids,  quinones,  sterols,  di- 
glycerides,  monoglycerides,  glycolipids,  lipamino  acids,  bile  acids,  gly- 
cerophosphatidic  acids,  inositol  lipids,  phosphatidyl  ethanolamines, 
lysophosphatidyl  ethanolamines,  phosphatidyl  cholines,  sphingomyelins, 
lysolecithins. 

Initial  separation:  neutral  from  phospholipids  and  glycolipids. 
When  attempting  to  separate  naturally-occurring  lipids,  it  is  best  to 
start  by  carrying  out  a  crude  separation  into  neutral  lipids,  phospho- 
lipids, and  glycolipids.  Like  all  the  column  chromatographic  procedures 
described  here,  this  is  done  using  a  stepwise  elution.1  The  sample  (100 
to  150mg  per  g  of  adsorbent)  is  added  to  a  column  of  coarse  mesh 
silicic  acid.  Elution  is  carried  out  first  with  chloroform  (10  column 
volumes  for  a  2.5cm  i.d.  X  5cm  column).  This  elutes  neutral  lipid. 
Ten  column  volumes  of  chloroform-methanol  1:1  v/v  or  of  methanol 
alone  will  elute  the  phospholipids.  If  the  lipid  sample  contains  a  lot  of 
glycolipids,  an  intermediate  elution  with  acetone  (30  to  50  column 
volumes)  will  elute  these  as  a  separate  fraction  before  the  phospho- 
lipids. Cardiolipin  will  be  largely  eluted  in  an  acetone  fraction  along 
with  the  glycolipids.  The  three-step  elution  is  very  useful  for  the  pre- 
liminary separation  of  nervous  tissue  lipids,  in  which  the  first  fraction 
will  consist  almost  entirely  of  cholesterol. 

Separation  of  neutral  lipids.  Numerous  solvent  systems  have 
been  used  for  the  separation  of  neutral  lipids  on  silicic  acid.  The  choice 
of  procedure  will  depend  on  the  composition  of  the  fraction  and  the 

1  Better  resolutions  can  often  be  obtained  by  using  gradient  elution,  which 
frequently  overcomes  tailing  of  one  lipid  fraction  into  another;  however,  step- 
wise elution  is  simpler  and  needs  no  special  equipment.  Automatic  equipment  for 
the  mixing  of  solvent  gradients  can  be  obtained  commercially.  Some  references 
to  gradient  elution  procedures  occur  in  the  text. 


36  COLUMN  CHROMATOGRAPHY 

relative  proportions  of  the  constituents.  A  procedure  to  suit  most  needs 
should  be  found  among  the  following: 

Hydrocarbons  (saturated)  can  be  eluted  first  with  light  petroleum 
ether,  hexane,  or  Skelly  B.  Unsaturated  hydrocarbons  such  as  squalene 
will  usually  be  the  tail  of  this  fraction. 

Esters  of  fatty  acids,  cholesterol  esters,  and  wax  esters  are  eluted 
together  with  1  percent  diethyl  ether  in  petroleum  ether  (5).  Esters 
containing  hydroxylated  fatty  acids  require  a  more  polar  solvent  for 
their  elution,  a  fact  that  can  be  used  to  separate  nonhydroxylated  and 
hydroxylated  fatty  acids  and  their  esters.  For  example,  pentane  with 
3  percent  diethyl  ether  elutes  nonhydroxylated  methyl  esters,  and 
pentane  with  20  percent  diethyl  ether  elutes  the  hydroxylated  form  (6). 
Free  acids  can  be  separated  into  nonhydroxylated  and  hydroxylated 
groups  by  eluting  first  with  benzene,  then  with  benzene-diethyl  ether 
9:1  v/v  (7). 

Fatty  acid  esters  which  differ  in  degree  of  unsaturation  can  be 
separated  on  silicic  acid  by  various  procedures.  Goldfine  and  Bloch  (8) 
and  Erwin  and  Bloch  (9)  achieved  separation  of  fatty  acids  according 
to  degree  of  saturation  by  treating  the  methyl  esters  with  mercuric 
acetate  and  chromatographing  the  mercuric  adducts  on  silicic  acid.  The 
saturated  esters  were  eluted  with  pentane-ether  95 : 5  v/v,  the  monoenoic 
esters  with  ethanol-ether  50:50,  the  dienoic  with  ethanol-acetic  acid 
99:2,  the  trienoic  with  ethanol-acetic  acid  99:1,  and  the  tetraenoic  and 
polyenoic  esters  with  ethanol-acetic  acid  95:5. 

Silicic  acid  impregnated  with  silver  nitrate  has  been  used  by  DeVries 
(10)  to  separate  saturated  and  unsaturated  fatty  acid  methyl  esters  and 
their  geometric  isomers.  DeVries  (11)  also  employed  this  absorbent  to 
separate  triglycerides  according  to  their  degree  of  unsaturation.  See 
also  pages  51-52. 

Triglycerides  separate  from  wax  and  sterol  esters  on  silicic  acid 
by  eluting  with  3  to  5  percent  ether  in  petroleum  ether.  Free  fatty  acids 
and  fatty  alcohols  are  eluted  after  triglycerides  with  a  slightly  more 
polar  solvent,  but  tailing  into  the  triglyceride  fraction  is  usual.  Diglyc- 
erides  elute  with  20  to  60  percent  ether  in  petroleum  ether,  and  mono- 
glycerides  elute  with  ether  or  chloroform  (5).  Monoglyceryl  ethers 
accompany  the  monoglyceride  fraction. 

A  hazard  is  encountered  when  chromatographing  monoglycerides  on 
silicic  acid:  both  the  1-  and  2-isomers  isomerize.  Diglycerides  do  not 
isomerize  (5,  13).  Isomerization  of  monoglycerides  is  prevented  by 
using  silicic  acid  impregnated  with  10  percent  w/w  boric  acid  (12).  On 
this  packing,  the  1-  and  2-monoglycerides  can  be  efficiently  separated. 

Separation  of  polar  lipids.  Polar  lipids  cannot  be  completely 
separated  on  silicic  acid,  but  some  separations  are  possible  by  eluting 
with  increasing  amounts  of  methanol  in  chloroform.  Much  better  reso- 


COLUMN  CHROMATOGRAPHY  37 

lutions,  however,  can  be  obtained  using  diethylaminoethyl  cellulose 
(DEAE),  described  below. 

Fiorisil  Columns 

Florisil  (Floridin  Co.,  Pennsylvania  Glass  Sand  Corp.,  2  Gateway 
Center,  Pittsburgh,  Pennsylvania)  is  a  synthetic  magnesium  silicate. 
It  has  a  number  of  uses  in  lipid  separation,  the  chief  one  being  the  sepa- 
ration of  cerebrosides,  sulfatides,  and  ceramides.  Florisil  has  the  dis- 
advantage, however,  that  its  chromatographic  properties  are  changed 
when  traces  of  water  are  present.  This  problem  may  be  overcome  by 
drying  the  adsorbent  and  by  using  very  dry  solvents.  Solvents  may  be 
dried  by  adding  5  percent  2,2-dimethoxypropane  to  them  as  described 
below.  A  useful  procedure  for  the  preparation  of  ceramide,  cerebrosides, 
and  sulfatides,  described  by  Rouser  et  al.  (14),  is  outlined  here. 

One  pound  of  Florisil  is  washed  on  a  sintered-glass  filter  (medium 
porosity)  with  8  bed-volumes  of  water.  It  is  then  activated  at  100°C 
for  6  hours  and  cooled  without  exposure  to  air.  The  appropriate 
amount  of  Florisil  is  quickly  weighed  and  1  percent  (by  weight)  water 
is  added.  Let  the  mixture  stand  for  an  hour  or  more  in  a  closed  con- 
tainer so  that  the  water  and  adsorbent  will  equilibrate.  Mix  the  equili- 
brated adsorbent  with  chloroform  containing  5  percent  (by  volume) 
2,2-dimethoxypropane.  This  slurry  is  then  poured  into  a  chromato- 
graphic tube. 

A  10.2cm  i.d.  column  is  used  for  the  separation  of  ceramides, 
cerebrosides,  and  sulfatides  from  about  3g  of  brain  lipid.  The  lipid 
sample  is  placed  on  the  column  in  chloroform  containing  5  percent  2,2- 
dimethoxypropane.  The  eluting  solvents,  which  must  all  also  contain 
5  percent  2,2-dimethoxypropane,  are: 

1.  Chloroform,  10  column  volumes 

2.  Chloroform-methanol  19:1  v/v,  10  column  volumes 

3.  Chloroform-methanol  70:30  v/v,  20  column  volumes 

The  first  fraction  will  consist  of  cholesterol  and  any  less  polar  lipids. 
The  second  will  be  ceramides,  and  the  third,  cerebrosides  and  sulfa- 
tides. The  flow  rate  of  solvents  should  be  about  50ml  per  minute.  If  the 
phosphatides  are  to  be  collected,  they  can  be  eluted  last  with  20  column 
volumes  of  chloroform-methanol  2:1  v/v,  saturated  with  water.  The 
ceramides  can  be  purified  by  preparative  thin-layer  chromatography 
(see  next  chapter),  and  the  cerebrosides  and  sulfatides  can  be  sepa- 
rated on  a  DEAE  column. 

Diethylaminoethyl  (DEAE)  and  Triethylaminoethyl  (TEAE) 
Cellulose  Columns 

DEAE  cellulose  gives  the  most  satisfactory  column  chromatographic 
separations  of  complex  lipid  mixtures.    Separation  is  achieved  either 


38  COLUMN  CHROMATOGRAPHY 

by  ion  exchange  or  by  differences  in  polarity  of  nonionic  groups.  The 
elution  characteristics  of  DEAE  are  most  easily  understood  if  lipids  are 
considered  as  being  in  three  groups  —  nonionic,  nonacidic  ionic,  and 
acidic.  Nonionic  lipids  are  eluted  according  to  relative  polarity.  The 
least  polar  lipids  such  as  the  sterols,  sterol  esters,  glycerides,  and  hydro- 
carbons are  eluted  with  chloroform.  Three  to  5  percent  methanol  in 
chloroform  elutes  cerebrosides  and  glycosyl  diglycerides.  All  of  the 
nonionic  and  ionic  nonacidic  lipids  are  eluted  with  increasing  amounts 
of  methanol  in  chloroform.  The  acidic  lipids,  however,  are  not  eluted 
with  chloroform-methanol  or  methanol  unless  an  acid,  base,  or  salt  is 
added  to  the  solvent. 

The  development  of  DEAE  cellulose  column  chromatography  is 
due  largely  to  Rouser  and  his  co-workers,  who  have  compiled  an  ex- 
tensive review  and  detailed  description  of  all  aspects  of  this  method 
(14).  Here  we  shall  deal  with  preparation  and  pretreatment  of  columns 
and  typical  elution  sequences  for  the  more  common  lipids.  For  varia- 
tions and  separations  of  more  complex  mixtures,  the  Rouser  article 
should  be  consulted. 

The  DEAE  cellulose  usually  recommended  is  Selectacel  DEAE 
cellulose  regular  grade  (Brown  Co.,  Berlin,  New  Hampshire).  This  is 
a  coarse  grade,  which  is  usually  best  for  lipid  separations.  The  DEAE 
must  be  washed  before  use,  as  it  contains  impurities.  Washing  can  be 
done  on  a  sintered-glass  or  Buchner  funnel,  using  about  3  bed  volumes 
of  acid  or  base.  The  DEAE  is  washed  first  with  IN  hydrocloric  acid, 
rinsed  with  water  to  a  neutral  pH,  then  washed  with  0.1N  aqueous 
potassium  hydroxide  followed  by  a  water  rinse  to  neutral  pH  again. 
After  washing,  the  DEAE  is  converted  to  the  acetate  form  by  washing 
it  with  3  bed  volumes  of  glacial  acetic  acid.  The  adsorbent  is  air-dried, 
then  dried  to  constant  weight  in  a  vacuum  dessicator  over  potassium 
hydroxide.  Thorough  drying  is  important.  The  dry  DEAE  is  left  over- 
night in  glacial  acetic  acid  and  finally  is  added  to  the  column  as  a  slurry 
in  glacial  acetic  acid.  The  excess  acid  is  blown  through  the  column  with 
nitrogen,  and  the  adsorbent  is  patted  down  gently  with  a  glass  rod.  A 
30cm  column,  1.0  to  4.5cm  i.d.,  is  a  useful  size.  The  packing  should  be 
about  20cm  high.  After  packing,  the  bed  of  adsorbent  is  washed  with 
3  bed  volumes  of  methanol,  3  bed  volumes  of  chloroform-methanol  1:1, 
and  3  to  5  bed  volumes  of  chloroform.  The  packing  should  not  be  al- 
lowed to  run  dry  at  any  time.  The  packed,  washed  column  should  then 
be  tested.  The  recommended  testing  procedure  is  as  follows: 

To  a  column  (2.5cm  i.d.  X  20cm)  add  a  solution  of  10  to  30mg  of 
cholesterol  in  5  to  10ml  of  chloroform.  Collect  10ml  volume  fractions 
using  chloroform  as  the  eluting  solvent  (flow  rate  about  3ml/min.)  and 
test  each  fraction  for  cholesterol.  The  best  test  is  to  add  5  drops  of 
acetic  anhydride  and  1  drop  of  concentrated  sulfuric  acid  to  1ml  of 
fraction.  A  green-blue  color  indicates  cholesterol.  The  column  is  judged 
satisfactory  if  cholesterol  first  appears  in  tube  7  to  9.  If  satisfactory, 


COLUMN  CHROMATOGRAPHY  39 

the  remainder  of  the  cholesterol  can  be  eluted  with  chloroform  and 
the  sample  applied. 

There  are  many  possible  elution  schemes  depending  on  the  separa- 
tions desired.  Many  elution  sequences  have  been  described  by  Rouser 
and  co-workers  (14,  15).  Two  of  more  general  use  are  described  below. 

Separation  of  lipid  samples  into  acidic  and  nonacidic  fractions 
(14).  Using  a  6cm  long  and  4.5cm  i.d.  column  of  DEAE  prepared  in 
chloroform-methanol  2:1  v/v,  apply  the  sample  in  the  same  solvent, 
then  elute  with  the  following  sequence  at  a  flow  rate  of   10ml/min.: 

1.  Chloroform-methanol  2:1  v/v  (4  column  volumes) 

2.  Methanol  (10  column  volumes) 

3.  Chloroform-methanol  4:1  v/v  made  0.01  to  0.05M  with  respect  to 
ammonium  acetate,  to  which  is  added  20ml  of  fresh  28  percent  w/w 
aqueous  ammonia2  per  liter  (10  column  volumes) 

4.  Methanol  (10  column  volumes) 

The  first  two  fractions  will  contain  all  nonacidic  lipids  plus  salts.  The 
third  fraction  will  contain  acidic  lipids  plus  salts,  and  the  fourth  will 
contain  salts  and  possibly  traces  of  lipids.  The  nonacidic  lipids  include 
all  the  neutral  lipids  and  nonionic  glycolipids  (like  the  cerebrosides  and 
phospholipids)  that  lack  a  negative  charge.  Acidic  lipids  include  all  those 
having  only  negatively  charged  groups,  such  as  fatty  acids  and  cere- 
broside  sulfates,  and  those  lipids  that  have  at  least  one  more  negative 
group  than  positive  groups,  such  as  phosphatidyl  serine. 

A  general  elution  scheme  (14).  This  procedure  utilizes  a  20cm 
high,  2  to  5cm  i.d.  column  of  DEAE  prepared  in  a  slurry  with  choloro- 
form.  The  sample  is  applied  in  chloroform,  and  the  following  solvents 
are  used  at  a  flow  rate  of  3ml/min. 

1.  Chloroform  (10  column  volumes) 

2.  Chloroform-methanol  9:1  v/v  (9  column  volumes) 

3.  Chloroform-methanol  7:3  v/v  (9  column  volumes) 

4.  Chloroform-methanol  1:1  v/v  (9  column  volumes) 

5.  Methanol  (10  column  volumes) 

6.  Chloroform-glacial  acetic  acid  3:1  v/v  (10  column  volumes) 

7.  Glacial  acetic  acid  (10  column  volumes) 

8.  Methanol  (4  column  volumes) 

9.  Chloroform-methanol-ammonium  salt  (as  described  in  step  3  of 
the  preceding  procedure)  (10  column  volumes) 

10.  Methanol  (10  column  volumes) 

Composition  of  the  resulting  fractions  can  be  expected  to  be: 

1.  Neutral  lipids 

2.  Cerebrosides,  lysophosphatidyl  choline,  phosphatidyl  choline, 
sphingomyelin,  and  mono-  and  diglycosyl  diglycerides 

2  See  footnote,  p.  24. 


40  COLUMN  CHROMATOGRAPHY 

3.  Ceramide,  aminoethylphosphonate,  ceramide  dihexosides  and 
polyhexasides,  and  phosphatidyl  ethanolamine 

4.  Ceramide  polyhexosides,  lysophosphatidyl  ethanolamine,  and  un- 
characterized  oxidation  products 

5.  Oxidation  products  and  salts 

6.  Free  fatty  acids,  glycine  conjugated  bile  acids,  and  unconjugated 
bile  acids 

7.  Phosphatidyl  serine 

8.  Lipid-free  fraction 

9.  Cardiolipid,  phosphatide  acid,  phosphatidyl  glycerol,  phos- 
phatidyl inositol,  sulfolipid,  sulfatides,  and  salts 

10.  Salts  and  traces  of  lipid 

Of  course,  not  all  the  lipids  listed  above  are  likely  to  be  present  in  one 
sample,  and  in  most  cases  fractions  will  contain  only  one  or  two  major 
components.  Thus,  a  sample  of  serum  lipid  would  yield  the  following 
fractions: 

1.  Neutral  lipids 

2.  Lysophosphatidyl  choline,  phosphatidyl  choline,  and  sphingo- 
myelin 

3.  Phosphatidyl  ethanolamine 

4.  Phosphatidyl  serine 

5.  Phosphatidyl  inositol 

The  first  fraction  could  readily  be  separated  on  a  silicic  acid  column 
into  glycerides,  sterol  esters,  and  free  fatty  acid  fractions.  Separation  of 
the  second  fraction  would  be  best  achieved  by  chromatography  on  a 
silicic  acid-silicate  column.  The  latter  column  is  prepared  by  treating 
silicic  acid  with  aqueous  ammonia.  Silicic  acid  (100  to  200  mesh)  is 
placed  on  a  coarse  sintered  glass  filter  and  washed  first  with  3  bed 
volumes  of  6N  hydrochloric  acid,  then  with  5  bed  volumes  of  water. 
The  adsorbent  is  then  heated  at  120°C  for  6  hours  and  cooled  without 
exposure  to  air.  Using  a  2.5cm  i.d.  column,  a  slurry  of  25g  of  the 
silicic  acid  is  prepared  in  40  to  100ml  chloroform-methanol  1:1  v/v  and 
4ml  of  28  percent  aqueous  ammonia.  The  chromatography  tube  is  filled 
to  a  height  of  10cm.  Before  the  sample  is  added,  the  column  is  washed 
with  4  column  volumes  of  chloroform.  The  fraction  containing  phos- 
phatidyl choline,  lysophosphatidyl  choline,  and  sphingomyelin  (50  to 
75mg)  is  added  to  the  column  in  about  5ml  of  chloroform. 

The  following  elution  sequence  is  then  applied: 

1.  Chloroform-methanol  4:1  plus  1.0  percent  (by  volume)  water 
(8  column  volumes) 

2.  Chloroform-methanol  4:1  plus  1.5  percent  (by  volume)  water 
(11  column  volumes) 

3.  Methanol  plus  2  percent  (by  volume)  water  (5  column  volumes) 

Fraction  1  will  contain  the  phosphatidyl  choline,  fraction  2  the  sphin- 
gomyelin, and  fraction  3  the  lysolecithin. 


COLUMN  CHROMATOGRAPHY  41 

Even  the  most  complex  mixtures  of  lipids  can  be  separated  by 
chromatography  on  DEAE  when  followed  by  suitable  rechromatog- 
raphy  of  mixed  fractions  on  silicic  acid,  silicic  acid-silicate,  Florisil, 
DEAE,  or  TEAE  columns.  Rouser  et  al.  (14,  15)  should  be  consulted 
for  elution  sequences  suited  to  separation  of  complex  mixtures. 

The  triethylaminoethyl  (TEAE)  cellulose  differs  from  DEAE  in 
having  a  greater  capacity  for  lipids  that  have  carboxyl  groups  as  their 
only  ionic  group.  Thus,  it  is  an  ideal  choice  for  the  separation  of  mix- 
tures containing  large  proportions  of  fatty  acids,  gangliosides,  and  bile 
acids.  TEAE  is  usually  used  in  the  hydroxyl  form,  and  elution  se- 
quences similar  to  those  used  with  DEAE  are  generally  employed  (14). 

REFERENCES 

1.  James,  A.  T.,  and  A.  J.  P.  Martin  (1952)  Biochem.  J.  50:  679. 

2.  Stahl,  E.  (1958)  Chem.  Ztg.  82:  323. 

3.  Hagdahl,  L.   (1961)  in  Chromatography,  ed.   E.   Heftmann ;   p.  56.   Reinhold 
Publ.  Corp.,  New  York. 

4.  Wren,  J.  J.  (1960)  /.  Chromatog.4:  173. 

5.  Hirsch,  J.,  and  E.  H.  Ahrens,  Jr.  (1958)  /.  Biol.  Chem.  233:  311. 

6.  Fulco,  A.  J.,  and  J.  F.  Mead  (1961)  /.  Biol.  Chem.  236:  2416. 

7.  Kishimoto,  Y.,  and  N.  S.  Radin  (1963)  /.  Lipid  Res.  A:  130. 

8.  Goldfine,  H.,  and  K.  Bloch  (1961)  /.  Biol.  Chem.  236:  2596. 

9.  Erwin,  J.,  and  K.  Bloch  (1963)  /.  Biol.  Chem.  238:  1618. 

10.  DeVries,  B.  (1963)  /.  Am.  Oil  Chem.  Soc.  40:  184. 

11.  (1964)  /.  Am.  Oil  Chem.  Soc.  41 :  403. 

12.  Thomas,  A.  E.,  J.  E.  Scharoun,  and  H.  Ralson  (1965)  /.  Am.  Oil  Chem.  Soc. 
42 :  789. 

13.  Mattson,  F.  H.,  and  R.  A.  Volpenhein  (1962)  J.  Lipid  Res.  3:  281. 

14.  Rouser,  G.,  G.  Kritchevsky,  and  A.  Yamamoto  (1967)  in  Lipid  Chromato- 
graphic Analysis,  ed.  G.  V.  Marinetti;  vol.  1,  p.  99.  Marcel  Dekker,  Inc., 
New  York. 

15.  Rouser,  G.,  G.  Kritchevsky,  D.  Heller,  and  E.  Lieber  (1963)  /.  Am.  Oil 
Chem.  Soc.  40 :  425. 


IV.  Thin -Layer  and  Paper  Chromatography 


THIN-LAYER  CHROMATOGRAPHY 

Although  the  basic  principle  of  thin-layer  chromatography  (TLC) 
was  first  described  many  years  ago,  it  did  not  become  a  popular  tech- 
nique until  Stahl  (1)  reported  his  standardization  of  procedures. 

In  TLC  the  adsorbent  is  spread  in  a  thin,  even  layer  on  a  glass 
plate,  and  the  components  of  an  applied  sample  are  separated  by  allow- 
ing a  suitably  composed  solvent  mixture  to  move  through  the  adsorbent. 
The  technique  is  simple  and,  in  many  cases,  more  rapid  than  others. 
The  speed  with  which  many  separations  can  be  achieved  make  TLC  a 
most  useful  adjunct  to  column  chromatography.  It  is  relatively  simple 
to  apply  a  concentrated  aliquot  of  solvent  from  a  column  chromato- 
graphic fraction  to  a  TLC  plate,  develop  the  plate,  and  by  suitable  de- 
tecting agents  determine  the  composition  of  the  fraction.  TLC  is  also 
useful  in  many  instances  as  a  purification  procedure.  Moreover,  TLC 
enjoys  wide  popularity  as  an  analytical  tool  in  its  own  right;  frequently 
the  analysis  of  very  small  samples,  which  would  be  extremely  tedious 
or  impossible  by  other  procedures,  can  be  readily  achieved  by  TLC. 
The  technique  is  not  without  hazards,  however,  and  they  will  be  noted 
as  each  stage  of  TLC  procedure  is  discussed. 

Preparation  of  Thin-Layer  Chromatographic  Plates 

Two  sizes  of  glass  plates  are  in  general  use:  4cm  X  20cm  and  20cm 
X  20cm.  While  plates  can  be  obtained  commercially  as  matched  sets, 
it  is  usually  more  economical  to  have  plates  cut  from  plate  glass  and 
smooth  their  edges.  Plates  should  then  be  marked,  on  the  basis  of 
evenness  of  thickness,  as  matched  sets  (5  large  plates  per  set  is  useful 
for  most  spreading  devices). 

The  plates  should  be  thoroughly  cleaned.  Just  prior  to  use  they 
should  be  cleaned  with  a  suitable  solvent,  such  as  chloroform  or  ben- 
zene, to  remove  any  traces  of  grease  that  would  interfere  with  the 
spreading  of  the  adsorbent. 

Several  types  of  adsorbent  are  available.  Some  satisfactory  ones 
for  universal  use  are  the  Merck  Silica  Gel  G  (with  and  without  calcium 
sulfate  as  binder),  Merck  Silica  Gel  H,  and  Supelcosil  12  B  (Supelco, 
Inc.).  Rouser  and  co-workers  (2)  recommend  the  use  of  Silica  Gel  G 
plain  (no  binder)  plus  10  percent  by  weight  magnesium  silicate  (Al- 
legheny Industrial  Chem.  Co.,  P.O.  Box  786,  Butler,  New  Jersey)  for 
chromatography  of  polar  lipids. 

Sometimes  it  may  be  preferable  to  wash  adsorbents  before  use  since 
impurities  in  them  may  interfere   with   subsequent  analyses.   A  bulk 

42 


THIN-LAYER  CHROMATOGRAPHY 


43 


washing  procedure  for  Silica  Gel  H,  as  described  by  Parker  and  Peter- 
son (3),  is  recommended.  The  Silica  Gel  H  (125g)  is  weighed  and 
placed  in  a  Buchner  funnel  on  2  pieces  of  Whatman  No.  2  filter  paper. 
Over  this  is  poured  1  liter  of  a  mixture  of  formic  acid  (98  to  100  per- 
cent), chloroform,  and  methanol  1:2:1  v/v/v.  This  is  sucked  through 
by  water  pump  vacuum,  and  the  bed  of  Silica  Gel  H  is  then  washed 
with  500ml  of  deionized  water.  The  adsorbent  is  then  spread  out  on 
an  enamel  or  heavy  aluminum  foil  tray  and  dried  in  an  oven  at  110  to 
120° C  for  48  hours.  The  lumps  should  be  broken  up  after  24  hours. 
When  preparing  a  slurry  from  washed  adsorbent,  mix  thoroughly  in 
a  Waring  Blendor. 

The  one  disadvantage  of  washing  procedures  of  this  type  is  that 
some  characteristics  of  the  gel  change,  and  frequently  it  is  difficult  to 
prepare  smooth,  even  plates.  Usually  it  is  more  convenient  to  wash  the 
adsorbent  when  it  has  already  been  plated.  This  is  done  by  allowing  a 
solvent  system  to  develop  beyond  the  planned  solvent  front.  Chloroform- 
methanol-water,  65:25:4  v/v/v  is  a  suitable  solvent  in  most  cases. 

While  TLC  plates  can  be  prepared  by  dipping  and  spraying,  by  far 
the  most  successful  and  reproducible  method  is  spreading,  using  a  com- 
mercial spreader  and  tray  such  as  the  Desaga  ( Brinkmann  Instruments, 
Inc.,  115  Cutter  Mill  Road,  Great  Neck,  Long  Island,  N.Y.).  The  plates 
are  aligned  on  the  tray  and  a  slurry  of  the  adsorbent  is  placed  in  the 
spreader  at  one  end  of  the  board  (Fig.  2).  The  slurry  will  vary  in  con- 
sistency according  to  the  thickness  of  the  layer  desired.  A  suitable 
thickness  for  general  use  is  0.25mm.  A  fixed  thickness  0.25mm 
spreader  is  available;  otherwise,  the  variable  thickness  spreader  is  set 
accordingly.  A  slurry  is  prepared  by  mixing  20g  of  adsorbent 
thoroughly  in  about  65ml  water.  Note  (Fig.  2)  that  a  small  plate  is 
placed  at  each  end  of  the  board  to  allow  for  run-out  from  the  spreader 


J 

t 

small  plate 
(4cm  x  20cm) 

A  thin-layer  chromatography  tray  and  spreader. 


small  plate 

(Fig.  2) 


44  THIN-LAYER  CHROMATOGRAPHY 

when  it  is  opened  and  closed.  These  end  plates  are  unevenly  spread  and 
therefore  are  not  used.  The  lever  on  the  spreader  is  turned  through  90° 
and  the  spreader  is  moved  steadily  across  the  plates  to  the  end,  where 
the  lever  is  again  turned  through  90°  to  close.  The  speed  with  which 
the  spreader  moves  across  the  plates  affects  the  thickness  of  the  layers, 
and  some  practice  is  required  before  plates  of  uniform  thickness  are 
obtained.  After  the  slurry  has  been  applied,  the  spreader  is  removed 
and  washed.  The  plates  are  then  vibrated  slightly  by  bouncing  the  tray 
up  and  down  several  times.  This  helps  to  even  out  the  layers.  The  plates 
are  air  dried  and  then  dried  at  120°C  for  30  minutes  to  an  hour  in  a 
clean  atmosphere  free  of  vapors.  It  is  best  to  prepare  and  use  TLC 
plates  under  standardized  conditions ;  otherwise,  separations  will  not 
be  reproducible  from  one  day  to  the  next.  If  the  atmosphere  is  very 
humid,  it  may  be  necessary  to  treat  plates  in  a  humidifying  chamber 
to  ensure  a  more  constant  water  content. 

Before  applying  samples  to  plates,  the  edges  should  be  made  uni- 
form by  stripping  the  first  4  or  5mm  of  adsorbent  in  a  straight  line.  If 
plates  are  to  be  developed  in  only  one  dimension,  it  is  wise  to  divide  the 
plate  into  lanes  by  scoring  through  the  adsorbent.  This  prevents  lateral 
spread  of  components  towards  each  other. 

Samples  may  be  applied  either  as  spots  or  streaks.  They  are  usually 
applied  with  Hamilton  Syringes  of  10  to  50/xl  capacity  (Hamilton  Co., 
P.O.  Box  307,  Whittier,  Calif.).  It  is  usual  to  apply  samples  as  a  short 
streak  (1cm)  made  by  overlapping  several  small  spots  or  as  a  single 
discrete  spot.  If  TLC  is  being  used  to  prepare  pure  samples  ("prepara- 
tive TLC"),  the  sample  is  applied  as  a  large  streak  made  by  overlapping 
spots  straight  across  a  large  (20cm  square)  plate  to  within  1cm  of  the 
edges,  or  by  using  a  commercially  available  "Streaker"  (Applied  Sci- 
ence, Inc.,  State  College,  Penn.).  The  sample  load  may  vary  consider- 
ably and  good  results  still  be  obtained.  One  may  plate  as  little  as  0.5/xg 
of  a  single  component  (on  a  0.25mm  plate)  and  still  be  able  to  detect 
it.  A  load  of  1500/xg  on  a  0.25mm  plate  (to  be  developed  in  2  dimen- 
sions) is  not  uncommon,  although  loads  of  250  to  500/ig  are  more  usual. 
With  preparative  TLC  plates  (layer  thicknesses  of  0.5  to  1mm),  several 
milligrams  may  be  plated. 

As  a  precaution  against  oxidation,  samples  should  be  plated  under  a 
flow  of  nitrogen.  Application  chambers  through  which  nitrogen  can  be 
passed  are  available  commercially;  however,  one  can  easily  be  made 
from  Plexiglas  (Fig.  3).  A  box  about  25cm  square  and  3cm  deep  is 
made  with  an  inlet  and  outlet  for  a  nitrogen  line.  A  narrow  (1cm  or 
less)  slot  is  made  in  the  lid  to  coincide  with  a  line  about  1cm  from  the 
bottom  of  a  plate  placed  in  the  chamber.  A  sliding  cover  is  fitted  over 
the  slot  so  that  the  plate  can  gradually  be  covered  as  the  sample  is  ap- 
plied. A  good  flow  of  nitrogen  is  allowed  to  pass  through  the  chamber 
during  plating. 


THIN-LAYER  CHROMATOGRAPHY  45 


nitrogen  out  <*—mmm'         "  77 

/  /  /■■^■r-^ —  nitrogen  in 

//  /  sliding  cover 

Chamber  for  applying  samples  to  TLC  plates  under  a  flow  of  nitrogen. 

(Fig.  3) 

Chromatograms  are  developed  in  glass  chambers,  in  wide-mouth 
Erlenmeyer  flasks  (for  small  plates),  or  in  special  "S-chambers."  The 
most  commonly  used  are  glass  chambers  10^"  X  2j4"  X  IO1/2" 
(Brinkmann  Instruments,  Inc.,  115  Cutter  Mill  Road,  Great  Neck,  Long 
Island,  N.Y.;  Analabs,  Inc.,  P.O.  Box  1501,  North  Haven,  Conn. 
06473;  and  other  suppliers).  The  sides  of  the  chambers  should  be  lined 
with  Whatman  No.  3  paper;  just  before  use,  250ml  of  the  solvent  to 
be  used  for  developing  the  chromatogram  should  be  placed  in  the 
chamber,  tilting  the  chamber  back  and  forth  to  saturate  the  paper 
with  the  solvent.  A  heavy  glass  lid  is  used  without  a  grease  seal. 

For  routine  checking  of  column  chromatography  fractions,  it  is 
often  convenient  to  have  a  series  of  1 -liter  wide-mouth  Erlenmeyer 
flasks  covered  with  one-half  of  a  Petri  dish.  Each  flask  will  accom- 
modate two  4cm  X  20cm  plates.  S-chambers  are  commercially  available 
vessels  which  sandwich  plates  within  a  narrow  space,  thus  providing  an 
area  uniformly  and  highly  saturated  with  the  solvent  vapor.  This  type 
of  chamber  is  frequently  used  with  the  commercially  available  pre- 
coated  plates  ("Prekotes,"  Applied  Science,  Inc.)  or  adsorbent  coated 
plastic  sheets  (Eastman  Kodak).  The  S-chambers  generally  give  the 
most  consistent  results. 

Detection  of  Lipids  on  Chromatograms 

Treatment  of  developed  plates  with  a  variety  of  reagents  allows 
visualization  of  separated  components  and  possible  identification  of 
specific  lipid  functional  groups.  When  combined  with  the  chroma- 
tography of  standards  developed  under  the  same  conditions,  detection 
of  specific  groups  in  most  cases  allows  the  identification  of  individual 
lipids. 

There  are  three  general  ways  in  which  separated  components  may 
be  visualized.  These  methods  show  the  position  of  the  spots  but  give 
little  or  no  further  information  about  the  compound.  These  procedures 
are  as  follows: 


46  THIN-LAYER  CHROMATOGRAPHY 

1.  Spray1  with  a  fluorescent  reagent  such  as  rhodamine  or  2,2- 
dichlorofluorescein  and  view  under  ultraviolet  light.  Protective  glasses 
must  be  worn  when  using  UV.  The  rhodamine  reagent  is  either 
Rhodamine  6G  or  B,  0.05  percent  by  weight  in  96  percent  ethanol,  and 
the  dichlorofluorescein  is  0.2  percent  by  weight  in  90  percent  ethanol. 
The  plate  must  be  viewed  while  still  wet  with  the  spray  reagent. 

2.  Spray  with  a  solution  of  55  percent  by  weight  sulfuric  acid  and 
0.6  percent  by  weight  potassium  dichromate  and  char  in  an  oven  at 
120°C. 

3.  Expose  to  iodine  vapor  in  a  closed  chamber. 

Method  2,  the  charring  procedure,  is  fairly  specific  for  lipids, 
but  carbohydrates  also  react.  As  little  as  0.5/xg  of  a  compound  may  be 
detected  in  this  way.  However,  there  is  the  disadvantage  that  the  lipids 
are  destroyed  and  cannot  be  recovered  for  further  investigations. 
Method  1,  especially  the  dichlorofluorescein,  is  the  method  of  choice 
if  the  lipids  are  to  be  recovered  for  further  analysis.  Unfortunately, 
visualization  of  small  amounts  of  phospholipids  is  difficult  with  this 
method.  Method  3  is  excellent  in  most  cases  but  depends  upon 
the  presence  of  unsaturated  lipids,  the  reaction  involved  being  that  of 
iodine  with  double  bonds.  Some  saturated  nitrogenous  lipids  (for  ex- 
ample, phosphatidyl  serine)  will  take  up  iodine,  but  most  other  satu- 
rated lipids  will  not.  Since  most  natural  lipids  contain  some  unsaturated 
moieties,  this  procedure  can  be  widely  used.  Iodine  vapor  should  not  be 
used  when  lipids  are  to  be  recovered  for  fatty  acid  analysis  since  some 
iodine  may  remain  associated  with  double  bonds  and  lead  to  erroneous 
results. 

Sprays  that  can  be  used  to  detect  the  presence  of  specific  elements 
or  groups  and,  therefore,  the  presence  of  lipid  classes  are  listed  below. 

Acid-molybdate  spray  for  detection  of  phosphorus-containing  lipids. 
This  spray  consists  of  5ml  of  60  percent  perchloric  acid,  10ml  IX 
hydrochloric  acid,  and  25ml  of  4  percent  w/v  ammonium  molybdate. 
Make  up  to  100ml  with  distilled  water.  Spray  and  heat  in  oven  at  110°C. 
Blue  spots  on  gray  background  are  given  by  phosphorus-containing 
lipids.  Sensitivity  is  about  1/xg. 

Ninhy drin  spray  for  detection  of  lipids  containing  free  amino  groups. 
A  positive  reaction  to  this  spray  is  obtained  with  phosphatidyl  ethanol- 
amine  and  serine,  their  lyso  derivatives,  and  any  other  lipid  with  a 
primary  amine  group. 

The  spray  is  a  0.2  percent  solution  of  ninhydrin  (1,2,3-indantrione- 
hydrate)  in  n-butanol-10  percent  aqueous  acetic  acid  95:5  v/v.  Spray 
and  heat  in  oven  at  110°C.  Purple  spots  indicate  the  presence  of  free 

1  A  number  of  spraying  devices  are  available  commercially  and  prepacked 
sprays  can  be  purchased.  One  useful  spraying  aid  is  the  Sprayon-jet  pack  power 
unit  (Sprayon  Products,  Inc.,  Cleveland,  Ohio).  Note:  Use  all  sprays  in  a  hood. 


THIN-LAYER  CHROMATOGRAPHY  47 

amino  groups.  Phosphatidyl  serine  does  not  always  react  well  unless 
some  acetic  acid  (from  a  developing  solvent)  remains  on  the  plate; 
hence,  the  plate  should  be  sprayed  soon  after  development. 

Dragendorff  reagent  for  choline-containing  lipids.  Positive  results  are 
given  by  phosphatidyl  choline,  its  lyso  derivative,  sphingomyelin,  and 
related  compounds. 

A  solution  of  8.0g  bismuth  subnitrate  in  20  to  30ml  30  percent  nitric 
acid  (density  1.18)  is  added  slowly,  with  stirring,  to  a  solution  of  28g 
potassium  iodide  and  1ml  6N  hydrochloric  acid  in  approximately  5ml 
water.  The  dark  precipitate  obtained  will  redissolve  giving  an  orange- 
red  solution.  Cool  this  to  5°C  and  filter.  Make  up  to  100ml  with  dis- 
tilled water.  This  is  the  stock  solution.  If  stored  in  a  dark  bottle  and 
refrigerated,  the  stock  is  stable  for  1  to  3  months.  To  make  the  spray 
reagent,  combine  the  following  in  the  given  order:  20ml  water,  5ml 
6N  hydrochloric  acid,  2ml  stock  solution,  and  5ml  6N  sodium  hydrox- 
ide. If  the  precipitate  of  bismuth  hydroxide  does  not  redissolve  even  on 
shaking,  add  a  few  drops  of  6N  hydrochloric  acid.  The  spray  solution 
is  stable  for  10  to  14  days  in  a  refrigerator.  Spray  heavily.  Positive  is 
a  dark  orange  on  a  yellow  background.  Sensitivity  is  lO^g. 

2,4-dinitrophenylhydrazine  sptay  for  free  aldehyde  and  keto  groups. 
A  positive  reaction  is  given  by  plasmalogens,  but  the  spray  is  not  very 
sensitive.  The  SchifT  reagent  spray  is  better  (see  below). 

Spray  heavily  with  a  0.5  percent  solution  of  2,4-dinitrophenlhydra- 
zine  in  2N  hydrochloric  acid.  Yellow  or  orange  spots  are  positive. 

Schiff  reagent  spray  for  plasmalogens.  SchifT  reagent  is  prepared  by 
dissolving  lg  basic  fuchsin  and  lOg  sodium  metabisulnte  in  10ml  con- 
centrated hydrochloric  acid  and  100ml  water.  This  solution  is  treated 
for  1  hour  with  charcoal,  filtered,  and  made  up  to  500ml  with  distilled 
water.  This  stock  should  be  colorless.  It  is  stable  in  a  refrigerator  for 
several  months.  The  spray  reagent  consists  of  250ml  0.05  percent 
sodium  metabisulnte,  2.5ml  0.05  M  mercuric  chloride  (1.35g  in  100ml 
water),  and  2.5ml  stock  SchifT  reagent.  Spray  heavily  and  allow  several 
minutes  for  the  reaction.  Mauve  spots  are  positive. 

Diphenylamine  spray  for  glycolipids.  Positive  results  with  this  spray 
are  given  by  all  glycolipids,  such  as  the  cerebrosides. 

The  spray  consists  of  20ml  of  a  10  percent  (by  weight)  ethanolic 
solution  of  diphenylamine,  100ml  concentrated  hydrochloric  acid,  and 
80ml  glacial  acetic  acid.  Spray  and  heat  for  5  to  10  minutes  at  100  to 
120° C.  Blue-gray  spots  on  a  light  gray  background  are  positive.  Sen- 
sitivity is  within  the  range  of  4.0  to  10.0(ug. 

a-naphthol  spray  for  glycolipids.  This  spray  gives  positive  results  with 
sterols  and  steroid  conjugates,  as  well  as  with  glycolipids. 

The  spray  consists  of  10.5ml  of  a  15  percent  (by  weight)  solution 
of    a-naphthol    in   ethanol,    6.5ml    concentrated    sulfuric    acid,    50.5ml 


48  THIN-LAYER  CHROMATOGRAPHY 

ethanol  and  4ml  water.  Heat  for  3  to  6  minutes  at  100  to  110°C.  Spots 
of  various  colors  are  positive. 

Resorcinol  spray  for  gangliosides.  Two  grams  of  resorcinol  are  dis- 
solved in  100ml  distilled  water.  Ten  ml  of  this  solution  are  added  to 
80ml  of  concentrated  hydrochloric  acid  containing  0.25ml  of  0.1M 
copper  sulfate,  and  the  resulting  solution  is  made  up  to  100ml  with 
distilled  water.  This  spray  reagent  is  stable  for  about  one  week  in  the 
refrigerator.  Spray  and  heat  in  oven  at  110  to  120°C  in  closed  jar. 
Gangliosides  give  violet-blue  reaction;  other  glycolipids  go  yellow. 

Other  spraying  methods.  Many  other  sprays  for  specific  groups  can 
be  applied  to  TLC  plates.  Generally  any  spray  reagent  used  in  paper 
chromatography  can  be  applied  with  little  or  no  modification  to  TLC 
plates.  The  TLC  plate  has  an  advantage  over  the  paper  chromatogram 
in  that  corrosive  sprays  can  be  used.  The  very  corrosive  procedure  of 
spraying  with  the  sulfuric  acid-chromate  solution  has  the  advantage  that 
observation  of  the  spots  during  charring  may  provide  additional  infor- 
mation. Sterols,  such  as  cholesterol,  and  various  bile  acids  undergo  bril- 
liant color  changes,  from  green  through  red-purple,  before  blackening. 

As  with  paper  chromatograms,  if  radioactively  labeled  lipids  are 
used,  spots  may  be  detected  by  exposing  the  plates  to  X-ray  film.  In 
these  cases  a  permanent  record  is  obtainable  in  the  form  of  an  X-ray 
print.  Otherwise,  results  can  be  documented  by  copying  the  patterns 
into  a  notebook  or  by  taking  photographs  with  a  Polaroid  camera. 

Whatever  visualization  and  detection  techniques  are  employed,  it 
is  absolutely  essential  that  standard  lipids  are  run  under  the  same  con- 
ditions. Runs  should  also  be  made  using  more  than  one  solvent  system 
before  identifications  are  made.  Identification  should  never  be  made 
on  the  basis  of  RF  values2  alone  since  separations  may  vary  with  dif- 
ferences in  humidity,  batch  of  adsorbent,  etc.  If  the  sample  contains 
rare  lipids  or  is  derived  from  a  source  not  previously  examined  by 
other  investigators,  any  unusual  lipids  should  be  assigned  an  identity 
only  when  additional  aids  have  been  employed.  In  such  cases  it  is 
necessary  to  isolate  enough  of  the  component  in  pure  form  to  be  ex- 
amined by  mass  spectrometry,  infrared  spectroscopy,  and  other  ana- 
lytical aids. 

Separation  of  Neutral  Lipids 

The  choice  of  solvent  systems  for  the  development  of  chromato- 
grams is  quite  wide  and  depends  on  the  adsorbent  used,  the  tempera- 
ture and  humidity  conditions  in  the  laboratory,  and  the  complexity 
of  the  mixture  to  be  resolved. 

In  general,  mixtures  of  low  boiling  petroleum  hydrocarbons  con- 
taining various  amounts  of  diethyl  ether  and  benzene  will   separate 

distance  from  origin  to  component  spot 


Rf  value  = 


distance  from  origin  to  solvent  front 


THIN-LAYER  CHROMATOGRAPHY 


49 


most  neutral  lipid  mixtures.  Neutral  lipids  are  readily  separated  from 
the  polar  in  a  number  of  ways.  In  many  cases,  development  of  the 
chromatogram  with  a  more  polar  solvent  mixture  such  chloroform- 
methanol-water  65:25:4  (all  mixtures  are  given  by  volume)  will  move 
the  neutral  lipids  to  or  near  the  solvent  front,  whereas  the  polar  lipids 
will  lag  behind  to  varying  degrees. 

If  free  fatty  acids,  cerebrosides,  and  cardiolipin  are  present,  how- 
ever, there  will  be  fraction  overlap.  In  these  cases  the  technique  of 
multiple  development  is  useful.  With  this  procedure,  the  plate  is  de- 
veloped with  the  chloroform-methanol-water  system  to  a  height  of 
about  10cm,  instead  of  the  usual  15cm  employed  with  average  plates. 
The  plate  is  then  dried  and  redeveloped  in  the  same  direction  with  a  less 
polar  solvent,  such  as  hexane-ether  4:1,  to  a  height  of  15cm  (Fig.  4). 
This  solvent  will  hardly  move  the  polar  lipids,  but  will  redistribute  the 


neutral 
lipids 


polar 
lipids 


<^-  2nd  solvent  front 
(15cm  from  origin) 


r  |i 


1st  solvent  front 
(10cm  from  origin) 

-  2nd  solvent  system 


1st  solvent  system 


origin 


Separation  of  polar  from  neutral  lipids  by  multiple  development  technique. 
(1)  Developing  solvent  allowed  to  rise  to  height  of  10cm  (chloroform-metha- 
nol-water 65:25:4).  (2)  Developing  solvent  developed  to  height  of  15cm 
(hexane-ether  4:1).  Adsorbent,  Silica  Gel  G.  (Fig.  4) 


50 


THIN-LAYER  CHROMATOGRAPHY 


t 

t 

f 

t 

• 

t 

1 

• 

y 

I 

• 

• 

• 

• 

» 

# 

I 

» 

» 

• 

r 

• 

1 

2 

3 

4 

5 

6 

7 

8 

9 

10 

II 

12 

solvent  front 


origin 


Separation  of  some  neutral  lipids.  Solvent  system  hexane-diethyl  ether-gla- 
cial acetic  acid  90:10:1. 

(1)  9-octadecene,  (2)  oleyl  alcohol,  (3)  oleylaldehyde,  (4)  oleic  acid,  (5) 
methyl  oleate,  (6)  cholesterol  oleate,  (7)  monolein,  (8)  diolein,  (9)  triolein, 
(10)  cholesterol,  (11)  tristearin,  (12)  phosphatidyl  choline.  Note  that  the 
polar  lipid  (12)  and  monolein  (7)  do  not  move  from  the  origin.  Adsorbent, 
Silica  Gel  G.  (Fig.  5) 


neutral  lipids  in  the  5cm  band  above  the  polar  lipids.  It  should  be  noted 
here  that  if  the  lipids  are  to  be  recovered  for  further  analysis,  especially 
for  fatty  acid  and  fatty  alcohol  analyses,  the  plates  must  be  dried  in  a 
box  through  which  there  is  a  flow  of  nitrogen. 

Figure  5  shows  the  separation  of  some  neutral  lipids.  The  solvent 
system  is  hexane-diethyl  ether-glacial  acetic  acid  90:10:1.  The  glacial 
acetic  acid  in  the  system  prevents  tailing  of  the  more  acidic  components. 
Similar  separations  can  be  obtained  using  a  system  without  the  acid  and 
with  differing  proportions  of  hexane-ether.  The  more  polar  the  system 
(that  is,  the  more  diethyl  ether,  or  acid,  or  both,  it  contains),  the  further 
it  will  move  the  relatively  more  polar  materials. 

Isomeric  mono-  and  diglycerides  can  be  separated  by  TLC.  As  we 
previously  noted  (p.  36),  1-  and  2-monoglycerides  can  be  separated  on 
columns  impregnated  with  boric  acid.  Similarly,  these  isomers  may  be 
separated  on  Silica  Gel  G  TLC  plates  which  have  been  impregnated 
with  ammonium  borate.  The  solvent  system  is  chloroform-acetone  96:4. 
The  1,2-  and  the  1,3-diglycerides  may  be  separated  on  Silica  Gel  G  by 


THIN-LAYER  CHROMATOGRAPHY  51 

using  petroleum  hydrocarbon  (boiling  range  40  to  60° C) -diethyl  ether 
70:30. 

Numerous  other  systems  are  available  for  special  separations  of 
neutral  lipid  mixtures.  Many  of  the  earlier  ones  were  reviewed  by 
Mangold  and  Malins  (4),  and  more  recently  by  Renkonen  and 
Varo  (5). 

Argentation  Thin-Layer  Chromatography 

Fatty  acids,  their  methyl  esters,  cholesterol  esters,  and  glycerides 
can  be  separated  according  to  degree  of  unsaturation  by  TLC  on  silica 
gel  impregnated  with  silver  nitrate.  The  cis  and  trans  isomers  of  some 
fatty  acids  can  also  be  separated  in  this  way. 

Silver  nitrate  plates  can  be  made  in  several  ways.  Some  investiga- 
tors prepare  plates  by  spraying  silica  gel  plates  with  a  10  percent 
solution  of  silver  nitrate  in  aqueous  ethanol.  "Prekotes"  or  other  com- 
mercially available  plates  can  be  impregnated  with  silver  nitrate  by 
dipping  the  plates  into  a  10  to  12  percent  solution  of  silver  nitrate. 
The  most  reliable  procedure,  however,  is  to  prepare  the  silica  gel 
slurry  in  silver  nitrate  solution  rather  than  water  and  to  spread  the 
plates  in  the  usual  manner.  The  following  formula  will  generally  give 
satisfactory  plates,  although  it  may  be  necessary  to  adjust  to  the  pre- 
vailing humidity  conditions. 

Weigh  25g  silver  nitrate  and  dissolve  in  200ml  distilled  water. 
Weigh  60g  silica  gel  and  prepare  a  slurry  using  136ml  of  the  silver 
nitrate  solution.  Make  sure  that  the  slurry  is  very  thoroughly  mixed 
by  using  a  Waring  Blendor  or  by  shaking  vigorously  in  a  stoppered 
flask  for  about  10  minutes.  Prepare  plates  in  the  usual  way;  dry  and 
store  plates  in  a  dark  place. 

These  plates  can  be  spotted  in  dull  light ;  however,  they  should  be 
developed  in  a  dark  jar  or  the  developing  chambers  should  be  placed 
in  a  dark  place.  Depending  upon  the  separations  desired,  hexane-diethyl 
ether  in  varying  proportions  will  be  found  suitable  in  most  cases.  Figure 
6  illustrates  the  separation  of  several  cholesterol  esters  on  silver  ni- 
trate impregnated  Silica  Gel  G.  The  plates  were  developed  with  hexane- 
ether  93:7  for  monoenoate  separation  and  93:17  for  dienoates. 
To  separate  the  more  unsaturated  compounds  (trienoates,  tetraenoates, 
etc.),  increasing  quantities  of  ether  in  hexane  must  be  used.  For 
example,  a  mixture  of  saturated,  monoene,  diene,  triene,  tetraene- 
pentaene-hexaene  (polyene  fraction)  of  fatty  acid  methyl  esters  can 
be  separated  into  5  bands  by  using  60:40  hexane-ether,  although  there 
may  be  slight  overlap.  Lipids  can  be  recovered  from  the  scraped  off 
silica  gel  by  extraction  with  moist  diethyl  ether.  Rechromatography  on 
silver  nitrate  impregnated  plates  will  yield  purified  lipid  fractions. 

Solvent  systems  similar  to  those  described  above  can  be  used  to 
separate  various  glyceride  mixtures  on  the  basis  of  unsaturation.  Each 


52 


THIN-LAYER  CHROMATOGRAPHY 


1 

1 

# 

1 

I 



1 

1 

> 

1 

l 

l 

f 

ft 

1 

l 

I 

t 

# 

a 

b 

c 

d 

e 

f 

a 

b 

c 

d 

e 

Plate  1 :  monoenoates 


Plate  2:  dienoates 


Thin-layer  chromatography  of  cholesterol  esters  on  Silica  Gel  G  impregnated 
with  silver  nitrate.  Plate  1:  (a)  mixture  of  cholesterol  palmitate,  elaidate, 
oleate;  (b)  stearolate  [CH3(CH2)7C  =  C(CH2)7COOH  is  stearolic  acid]; 
(c)  mixture  of  elaidate,  oleate,  stearolate;  (d)  trans  and  cis  vaccenate; 
(e)  trans  and  cis  erucate;  (f)  trans  and  cis  petroselenate.  Plate  2:  (a) 
linoleate;  (b)  9-cis- 12- trans  octadecadienoate;  (c)  linelaidate;  (d)  mixture 
of  a,  b,  and  c;   (e)   9-trans-12-c/s  octadecadienoate.   (After  Sgoutas,  IS.) 

(Fig.  6) 


investigator  will,  however,  find  some  trial  runs  necessary  in  order  to 
ascertain  the  particular  solvent  systems  suited  to  his  conditions. 

Separation  of  Phospholipids  and  Glycolipids 

Numerous  solvent  systems  for  one-dimensional  TLC  of  phospho- 
lipids and  glycolipids  have  been  reported.  Among  those  that  give 
satisfactory  separations  of  mixtures  containing  most  of  the  common 
phospholipids  and  cerebrosides  are:  (a)  chloroform-methanol-28  per- 
cent aqueous  ammonia  75:25:4  (on  Silica  Gel  G) ;  (b)  chloroform- 
propionic  acid-n-propanol-water  10:10:10:4  or  2:2:3:1  (on  Silica  Gel 
G  plates  prepared  in  5  percent  ammonium  nitrate);  (c)  chloroform- 
methanol-acetic  acid- water  25:15:4:2  (on  Silica  Gel  G  plain  prepared  in 
0.001M  sodium  carbonate). 

For  routine  checking  of  column  chromatography  fractions  and  for 
many  analyses,  unidimensional  systems  are  suitable.  If,  however,  it 
is  desired  to  separate  all  the  phospholipids  and  glycolipids  of  a  multi- 
component  mixture  or  to  characterize  a  lipid  mixture  from  a  new 
source,  then  two-dimensional  TLC  must  be  used.  Furthermore,  it  is 
highly  desirable  that  the  number  and  identity  of  components  in  a 
complex  mixture  be  established  with  more  than  one  set  of  developing 
solvents.  Several  such  systems  have  been  developed  by  Rouser  et  al.  (2). 


THIN-LAYER  CHROMATOGRAPHY  53 

In  two-dimensional  TLC  the  sample  is  spotted  in  one  corner  of  a 
20cm  X  20cm  plate  and  developed  in  one  direction  with  one  solvent 
system;  then,  after  removal  of  the  first  solvent,  the  plate  is  turned 
through  90°  and  developed  in  the  second  direction  with  solvent  2.  The 
adsorbents  used  may  be  any  commonly  used  TLC  adsorbent.  The 
chromatogram  maps  shown  in  Figure  7  (p.  54)  illustrate  separations 
obtained  by  using  pairs  of  solvent  systems. 

In  the  separations  shown  in  Figure  7,  the  gangliosides  are  present 
as  one  spot.  However,  gangliosides  can  be  separated  into  mono-,  di-, 
tri-,  and  tetrasialogangliosides  by  using  several  solvent  systems.  The 
system  described  by  Kuhn  and  Wiegandt  (<5)  gives  good  separation  of 
the  major  gangliosides.  The  solvent  system  is  propanol-water  7:3  (on 
Silica  Gel  G).  Chloroform-methanol-water  60:35:8  (on  Silica  Gel  G) 
also  gives  good  separation  of  gangliosides  (7).  Leedeen  (8)  has  de- 
scribed a  multiple  development  system  that  gives  good  resolution  of 
major  gangliosides;  double  length  (40cm)  plates  and  two  sucessive 
runs  of  chloroform-methanol-2.5N  ammonia  60:40:9  are  used. 

Quantitative  Thin-Layer  Chromatography 

There  are  several  possible  approaches  to  quantitative  analysis  by 
TLC.  These  approaches  can  be  divided  into  four  main  groups. 

1.  Direct  analysis  on  the  TLC  plate. 

a.  Charring  under  standard  conditions  and  subjecting  the  plate 
to  photodensitometry. 

b.  Direct  radio  scanning  (when  radioactively  labeled  lipids  are 
being  handled). 

c.  Neutron  activation  analysis. 

2.  Gravimetric  analysis  by  recovery  of  the  lipids  from  each  spot. 

3.  Analysis   for  specific  elements   or   functional  groups   on   lipids 
extracted  from  the  adsorbent. 

4.  Analysis    for   specific    elements   or    functional   groups    on    lipid 
plus  adsorbent. 

In  the  first  group,  charring  followed  by  densitometry  has  been  used 
successfully  by  a  number  of  investigators.  This  method  is,  however, 
often  difficult  and  tedious  to  standardize.  Standard  lipids  must  be 
charred  and  separate  curves  prepared  for  the  densitometry  of  each. 
The  size  of  spots  for  each  lipid  should  be  fairly  consistent  from  day 
to  day,  which  is  often  difficult  to  achieve.  Moreover,  charring  should 
ideally  convert  the  lipids  quantitatively  to  carbon,  but  this  is  not  easily 
achieved  in  practice.  Other  detection  methods  besides  charring  have 
been  successful.  Such  methods  are  usually  specific  for  lipids  containing 
one  specific  group;  for  example,  staining  with  chlorox-benzidine  re- 
agent is  specific  for  amide  groups.  Sphingolipids  have  been  analyzed 
by  densitometry  after  having  been  stained  by  this  method  (9). 


54 


THIN-LAYER  CHROMATOGRAPHY 


2* 

in 

c    ^ 

c 

"#     a>| 

% 

<fr 

solvent  1 


o_  * 

• 

ID 

iv 

V 

A     C\J*    GO 

inf    ~ 

V* 

<fr 

CO 

V 

■*-> 
03 


U 

cd 

s 


cu 

s 


+ 


-     rS 


••  CM     i    /~v 

rt   rt  5  fe 

P<Ph     I 

rt  S  O 

£      g 

.2  £  ° 

w)    1    t— 1 
rt  T3  -G 

6 '2  u 

G    y  N 

•  S  S 
o 

S3" 

a »° 

rt   •• 

w   bflco 

<u   G  *■* 

O  iS  .2 

3  d  1 

w  II  g 

I  s * 
<..g 

is  I 

to   rt   rt 

gri      (UN 

g  o   c   •• 

43    c    C  CM 
4>    <u    o  S 

*  13  * 


c  E  °  "S 

.G     *-"     r«     rt 

■9 .2 -3  £ 


-  8  11 J2 

rt   O     .    u 

8  "3* 

'      CD  CO.* 

*  ^   G  ~ 
H    co    rt    bfl 


>* 

T3 


.-     CO 
CO     ft 

.2^ 

,G    g, 

bfl  o 

G    co 
rt    >» 


rt 

C 

o 
a 

(0 

J  "ft 

o  >»"55  o 

S£  o  >, 

S|  8m 

T3 


>>  fa  o.: 
3  rt  a  rt 

+j  <u  <u  j- 
rt  T3  T3  CX 
«G  •£  'B  co 
a  £    £    O 

O  u  u.  o» 
.G    <U    <U  -ST 

ftOUQ 


O  1-4  CM  CO  rj-  vft 


G 

rt 


G         G 

r  *s 
Mil 

O   u   rt 
G    rt  J! 


0   G    0 
•J   G 

^G   >.  cu   o   10 


^  <~  T3  tJ  T3  >» 

to  ^-v  *43  '43  • ^3  c 

*>  J3    c«    rt  rt  o 

*j   >.  ft  ftft  c 

rt   r?  w   co  co  .5 

3   g  J3  J3  jc  a 

C/3  13  Ph  pL|  Ph  c/3 


m 


VO  ts  00  o> 


G^J 

G    rt 


Ph       -    G 

.G"  co  .G 

— '     <L)     Q, 

—  ^;g 
2  'C  o 

J    (US 
•H  CM 


'S  rt   co 

!>  "-•-"  T3 

co  ^v'G 

4>  J    rt 

^2  o  ^ 

<u  u    «u 

J- ,  -o    «u 

cu  >,  u 


CO 


THIN-LAYER  CHROMATOGRAPHY  55 

Radio  scanning  of  TLC  plates,  using  a  proportional  gas-flow  counter 
tube,  is  generally  successful  but  is  of  limited  value. 

Neutron  activation  analysis  has  been  tried  and  promises  to  be  of 
great  value  in  the  analysis  of  very  small  quantities  of  lipids.  This 
approach  awaits  further  development. 

Gravimetric  analysis  is  not  generally  a  reliable  approach.  It  is 
often  adequate  on  a  preparative  TLC  scale  when  large  quantities  are 
being  chromatographed  and  recovered.  At  lower  levels,  however,  it  is 
often  difficult  to  recover  lipids  completely  from  the  adsorbent,  par- 
ticularly polar  lipids.  It  is  not  recommended  as  an  analytical  procedure. 

The  third  approach  suffers  the  same  disadvantage  as  gravimetric 
analysis  in  that  complete  or  consistent  recovery  of  lipids  from  the 
adsorbent  is  frequently  difficult  and  tedious. 

The  fourth  approach  is  less  cumbersome  than  other  methods  and, 
in  general,  is  the  best  procedure.  It  is  essential,  however,  that  pre- 
cautions are  taken  against  interference  with  the  analysis  by  the  adsor- 
bent itself,  by  impurities  in  it,  or  by  reagents  used  for  localization  of 
the  spots.  Exposing  plates  to  iodine  vapor  and  outlining  spots  with  a 
needle  is  ideal  in  many  cases.  The  iodine  resublimes  fairly  rapidly  and 
the  spots  can  then  be  removed  either  by  scraping  or  aspirating  off. 
This  method  cannot  be  used  if  the  lipids  are  being  removed  for  analysis 
of  their  fatty  acid  moieties,  since  some  iodine  may  remain  irreversibly 
added  to  double  bonds.  In  such  cases,  it  is  best  to  localize  spots  by 
spraying  with  one  of  the  fluorescent  dyes  like  2,7-dichlorofluorescein. 

After  the  spots  are  removed  and  put  into  suitable  containers,  many 
analyses  can  be  carried  out  directly  on  the  adsorbent  plus  lipid.  For  ex- 
ample, phosphorus  analyses  for  the  relative  proportions  of  phospho- 
lipids can  be  carried  out  in  the  presence  of  adsorbent  directly  after 
digestion  of  the  lipid.  Detailed  instructions  for  determination  of  organic 
phosphorus  and  other  analytical  procedures  are  given  in  Chapter  6. 
Procedures  for  the  analysis  of  fatty  acids  and  fatty  aldehydes  of  lipids 
recovered  from  TLC  plates  are  given  in  the  chapter  on  gas-liquid 
chromatography. 

A  scheme  for  analysis  of  phosphoglycerides.  Lipids  from  natural 
sources  are  complicated  mixtures  of  different  molecular  species.  A 
triglyceride  preparation  that  contains  only  two  different  fatty  acids 
could  actually  be  a  composition  of  six  different  species.  Thus,  with 
fatty  acids  A  and  B,  we  can  have  triglycerides  AAA,  BBB,  AAB, 
BBA,  ABA,  and  BAB.  Similarly,  with  a  phosphoglyceride  containing 
only  two  fatty  acids,  we  could  have  four  species:  AA,  BB,  AB,  BA. 
With  increasing  frequency,  answers  to  the  role  in  biological  systems 
demand  that  we  know  the  pattern  of  the  existing  molecular  species. 
In  the  case  of  glycerides,  there  is  already  a  vast  amount  of  knowl- 
edge about  determining  structural  patterns,  and  references  to  pertinent 
methods  and  results  are  given  in  Suggested  Further  Readings.  In  the 


56  THIN-LAYER  CHROMATOGRAPHY 

case  of  polar  lipids,  however,  only  in  the  last  few  years  has  progress 
been  made  in  the  analysis  of  molecular  species.  Renkonen  (10) 
demonstrated  that  much  could  be  learned  about  the  molecular  species  of 
many  natural  phosphatides  by  examining  the  diglyceride  parts  of  these 
molecules  by  thin-layer  chromatography.  Kuksis  and  Marai  (11)  ex- 
tended this  approach  to  include  examination  of  the  derived  diglyceride 
acetates  by  gas-liquid  chromatography.  They  first  applied  this  technique 
to  the  determination  of  the  complete  structure  of  Qgg  yolk  lecithins. 
More  recently  Kuksis  et  al.  (12)  have  shown  that  the  method  can  be 
applied  successfully  to  the  determination  of  the  molecular  species  of 
the  lecithins  from  rat  heart,  kidney,  and  plasma.  The  outline  of  their 
procedure  (summarized  in  Fig.  8)  is  as  follows: 

1.  The  phosphoglycerides  to  be  studied  are  isolated  and  purified 
by  column  or  preparative  thin-layer  chromatography.  Aliquots  of  the 
isolated  pure  phosphoglycerides  are  taken  for  immediate  transmethyla- 
tion. The  methyl  esters  of  the  fatty  acids  thus  obtained  are  analyzed 
by  gas-liquid  chromatography  (see  Chapter  5  for  details  of  GLC). 

2.  Separate  aliquots  of  the  pure  phospholipids  are  converted  directly 
to  the  diglycerides  by  dropping  the  removed  silica  gel  bands  into  a  buf- 
fered phospholipase  C  solution  (10).  The  incubations  are  continued 
for  30  to  60  minutes  at  28  to  30 °C  under  a  layer  of  diethyl  ether. 

3.  At  the  end  of  digestion  the  phases  are  separated  and  further 
extractions  with  ether  are  made.  The  combined  solvents  are  evaporated. 

4.  The  diglycerides  obtained  are  purified  by  TLC  on  Silica  Gel 
H  (Merck  &  Co.)  using  petroleum  ether-diethyl  ether- formic  acid 
120:40:3  v/v/v  as  the  developing  solvent. 

5.  The  diglycerides  are  recovered  from  the  scraped-ofr*  bands  (using 
2,7-dichlorofluorescein  for  location)  by  elution  with  chloroform. 

6.  The  diglycerides  are  dissolved  in  pyridine  and  converted  to  the 
diglyceride  acetates  by  treatment  with  acetic  anhydride  at  room  tem- 
perature for  12  hours.  If  the  original  lipid  sample  is  400  to  500mg, 
the  diglycerides  obtained  should  be  dissolved  in  about  0.1ml  pyridine 
and  treated  with  0.25  to  0.50ml  acetic  anhydride. 

7.  The  diglyceride  acetates  are  separated  according  to  degree  of 
unsaturation  by  TLC  on  0.25mm  thick  Silica  Gel  G  plates  (20cm  X 
20cm)  containing  20  percent  silver  nitrate.  About  10  to  15mg  of  ma- 
terial is  applied  as  a  band  and  the  plate  is  developed  twice  in  the  same 
direction  with  a  solution  of  0.7  or  0.8  percent  methanol  in  chloroform. 
The  bands  are  located  using  the  fluorescein  reagent.  The  individual 
bands  are  recovered  by  eluting  with  chloroform-methanol  9:1  v/v  to 
which  5  percent  water  has  been  added. 

8.  The  eluates  are  reduced  to  dryness  under  nitrogen  and  subjected 
to  gas-liquid  chromatographic  analysis. 


THIN-LAYER  CHROMATOGRAPHY 


57 


Sen 
<V    o    <-> 

rt  5°  ^ 
^  a>  •+-> 


cu  £  52        ^ 


b    CO    ctf 


■-* 


^ 

^ 


c 
U 

O 


CO 


CO 

cj 


*       2 ' 

«+h         CM 


> 


Jo 

di  a; 

CO 

CO      CO 

iD 

O    aj 

TJ 

J^    0, 

as 

<U 

a 

—    >v 

"*-'    G 

^  S  "S  g 

rt  *"  *+* 


<\>    as   O   <u ' 
co  M    G 


^-^ 

^        CM                 co 

CO 

•£         T3                 co 

<U         ,-,         C                                        ^ 

u 

6  g  *          * 

.5  «4-»   eo               ™ 

a  w  g            r  ^ 

*2'|               ^ 

fi  B  «         o 

a  co  o 

4h     D     (^ 

rt 

c 

XI 

u 

G 

rt 

rt 

rt 

a 

u 

~ 

<u 

QJ 

co  T3 

5 

^ 

o 

u 

eo 

'•^   CO 

buo 

oj 

"55 

G 

G 

k3 

r*l 

"55 

eg 

c3 

•- • 

4-J 

G 

CO 

G 

rt 

D 

.  .— 

cu 

u 

Rj 

h-1 

« 

^-, 

o 

U 

CO 

Diagram  of  analysis  scheme  for  phosphoglycerides. 


(Fig.  8) 


58  THIN-LAYER  CHROMATOGRAPHY 

9.  The  mixture  of  diglyceride  acetates  (from  step  6)  is  analyzed  by 
gas-liquid  chromatography  in  order  to  obtain  the  overall  molecular 
weight  distribution. 

10.  Further  information  is  obtained  by  subjecting  the  diglyceride 
acetates  to  hydrolysis  by  pancreatic  lipase,  which  selectively  removes 
the  fatty  acid  in  the  a(l) -position.  The  free  fatty  acids  and  mono- 
glycerides  obtained  are  removed  separately  after  thin-layer  chroma- 
tography as  described  for  the  diglycerides  (step  7). 

11.  The  positional  distribution  of  the  fatty  acids  in  the  original 
mixture  is  determined  by  hydrolysis  with  phospholipase  A,  which  re- 
moves the  fatty  acids  in  the  f3 (2) -position ;  the  fatty  acids  are  then 
converted  to  their  methyl  esters  and  analyzed  by  gas  liquid  chroma- 
tography. This  allows  for  verification  of  the  positional  distribution  of 
the  fatty  acids  derived  from  the  lipase  digestion. 

The  two  enzymic  incubations  in  this  procedure  are  carried  out  as 
follows : 

The  lipase  digestion  of  the  diglyceride  acetates.  Fifty  to  lOOmg  of 
sample  is  added  (in  a  5ml  screw  cap  vial)  to  a  predetermined  weight 
of  pancreatin  (Steapsin,  Nutritional  Biochemical  Corp.).  The  amount 
of  enzyme  chosen  is  that  which  hydrolyses  50  percent  of  a  given  weight 
of  diglyceride  acetate  in  1  to  2  minutes.  One  ml  of  1M  trishydroxy- 
methylamino  methane  (pH  8.0),  0.1ml  22  percent  calcium  chloride,  and 
0.25ml  0.1  percent  bile  salts  solution  are  added  to  sample  plus  pancreatin. 
The  vial  is  warmed  in  a  water  bath  at  40° C  for  1  minute.  The  cap  is 
then  screwed  on  tightly  and  the  vial  is  shaken  for  the  time  required; 
for  50  percent  hydrolysis,  this  is  usually  1  to  2  minutes.  At  the  end  of 
reaction  the  contents  are  acidified  with  6  N  HCL  and  extracted  with 
ether. 

The  phospholipase  A  digestion  of  the  phosphoglyceride.  From  each 
sample,  150  to  200mg  of  phosphoglyceride  is  dissolved  in  100ml  diethyl 
ether  and  treated  with  1ml  of  0.1  percent  rattlesnake  venom  (Crotalus 
adamenteus)  in  0.005M  CaCl2  solution.  The  solution  is  allowed  to 
stand  overnight,  after  which  the  precipitate  is  removed  by  centrifuga- 
tion  and  washed  once  with  diethyl  ether.  The  ether  phase  should  be 
free  of  phosphorus  (it  should  contain  only  free  fatty  acid)  if  the 
reaction  is  complete. 

This  scheme,  therefore,  supplies  the  following  information: 

1.  The  total  fatty  acid  composition  of  the  phosphoglyceride  (step 
A  in  Fig.  8). 

2.  The  overall  molecular  weight  distribution  of  the  diglyceride 
acetates  derived  from  the  phosphoglyceride  (steps  B,  C,  D). 


PAPER  CHROMATOGRAPHY  59 

3.  The  overall  molecular  weight  distribution  and  proportional 
contribution  of  the  diglyceride  acetates  according  to  their  degree  of 
unsaturation  (steps  B,  C,  E). 

4.  The  overall  composition  and  positional  placement  of  the  fatty 
acids  in  the  derived  diglyceride  acetates  (steps  B,  C,  G,  H). 

5.  The  fatty  acid  composition  of  position  2  in  the  original  phospho- 
glyceride  (steps  I  &  J),  a  verification  of  the  information  derived  in  4. 

From  the  above  information  it  is,  therefore,  possible  to  deduce  the 
position  of  the  fatty  acids  in  the  phosphoglyceride  and  to  describe  the 
molecular  species  present.  The  method  allows  for  cross-checking  in 
that  the  total  fatty  acids  composition  of  the  original  phospholipid 
(under  5  above)  should  be  the  same  as  that  of  the  derived  diglyceride 
acetates  (under  4  above) . 

The  conditions  suitable  for  the  GLC  analysis  steps  in  the  above 
scheme  are  to  be  found  in  the  next  chapter  (see  pp.  76-78,  82). 

PAPER  CHROMATOGRAPHY 

Early  attempts  to  apply  paper  chromatography  to  the  resolution 
of  naturally  occurring  lipid  mixtures  were  only  partially  successful. 
It  was  not  until  chromatography  on  impregnated  papers  was  introduced 
that  more  successful  separations  were  made.  Papers  have  been  treated 
in  various  ways,  including  acetylation  and  impregnation  with  tetralin 
and  formalin.  The  most  successful  and  widely  used  procedure  is  chro- 
matography on  paper  impregnated  with  silicic  acid,  a  technique  de- 
veloped mainly  by  Marinetti  (14)  and  his  co-workers.  This  approach 
enjoyed  wide  popularity  for  some  time  until  the  development  of  TLC 
procedures,  which  are  more  rapid  and  more  versatile  than  paper  chro- 
matography and  allow  a  wider  range  of  sample  loading.  Paper  chroma- 
tography is,  however,  still  popular  in  a  number  of  laboratories  for 
some  special  purposes.  A  number  of  reviews  give  details  of  paper 
chromatographic  procedures  (14, 15, 16). 

One  special  use  of  paper  chromatography  in  lipid  analysis  should 
be  mentioned  here,  the  method  of  phosphatide  analysis  introduced  by 
Dawson  in  1954.  Very  mild  hydrolysis  is  used  to  deacylate  phosphatides, 
after  which  the  water  soluble  phosphorus-containing  compounds  are 
analyzed  by  paper  chromatography.  Again,  after  the  introduction  of 
TLC  this  method  declined  in  popularity.  It  does,  however,  offer  su- 
perior resolution  of  the  acidic  phosphatides.  If  a  detailed  analysis  of 
previously  uncharacterized  lipids  is  being  undertaken,  this  method  pro- 
vides a  valuable  adjunct  to  TLC  and  column  procedures  for  the  analy- 
sis of  intact  lipids.  Dawson  (17)  has  recently  reviewed  this  procedure 
in  detail  and  discusses  those  areas  in  which  it  offers  special  advantages. 


60  THIN-LAYER  CHROMATOGRAPHY 

REFERENCES 

1.  Stahl,  E.  (1958)  Chem.  Ztg.  82:  323. 

2.  Parker,  R,  and  N.  F.  Peterson  (1965)  /.  Lipid  Res.  6:  455. 

3.  Mangold,  H.  K.,  and  D.  C.  Malins  (1960)  /.  Am.  Oil  Chem.  Soc.  37:  383. 

4.  Renkonen,  O.,  and  P.  Varo   (1967)   in  Lipid  Chromatographic  Analysis,  ed. 
G.  V.  Marinetti;  vol.  1,  p.  41.  Marcel  Dekker,  Inc.,  New  York. 

5.  Rouser,  G.,  G.  Kritchevsky,  and  A.  Yamamoto    (1967)   in  Lipid  Chromato- 
graphic Analysis,  vol.  1,  p.  99. 

6.  Kuhn,  R.,  and  H.  Wiegandt  (1963)  Chem.  Ber.  96:  866. 

7.  Wagner,  H.,  L.  Horhammer,  and  P.  Wolff  (1961)  Biochem.  J.  334:  175. 

8.  Leedeen,  R.  (1966)  /.  Am.  Oil  Chem.  Soc.  43:  57. 

9.  Austin,  J.  H.  (1966)  /.  Neurochem.  10:  921. 

10.  Renkonen,  O.  (1966)  Lipids  1:  160. 

11.  Kuksis,  A.,  and  L.  Marai  (1967)  Lipids  2:  217. 

12.  Kuksis,  A.,  W.  C.  Breckenridge,  L.  Marai,  and  O.  Stachnyk  (1969)  /.  Lipid 
Res.  10 :  25. 

13.  Sgoutas,  D.  S.  (1968)  Biochim.  Biophys.  Acta  164:  317. 

14.  Marinetti,  G.  V.  (1962)  /.  Lipid  Res.  3:  1. 

15.  (1964)   in  New  Biochemical  Separations,  ed.  A.  T.  James  and  L.  G. 

Morris ;  p.  339.  Van  Nostrand,  Princeton,  NJ. 

16.  Kates,  M.  (1967)  in  Lipid  Chromatographic  Analysis,  vol.  1,  p.  1. 

17.  Dawson,  R.  M.  C.  (1967)  in  Lipid  Chromatographic  Analysis,  vol.  1,  p.  163. 


V.   Gas-Liquid  Chromatography 


So  far  we  have  considered  chromatographic  techniques  in  which 
the  moving  phase  was  a  liquid  and  the  stationary  phase  a  solid. 
In  gas-liquid,  or  gas-liquid  partition,  chromatography  (GLC),  the 
moving  phase  is  a  gas  and  the  stationary  phase  is  a  liquid.  The  liquid 
is  spread  over  an  inert  solid  support,  usually  a  flux-calcined,  diato- 
maceous  earth  of  fairly  small  particles.  Separation  takes  place  in  a 
series  of  partitions  whereby  the  sample  goes  into  solution  in  the  liquid 
phase  and  is  subsequently  revaporized.  The  length  of  time  it  takes  a 
component  to  emerge  from  a  GLC  column  depends  upon  the  affinity 
it  has  for  the  liquid  phase  and  upon  its  boiling  point.  Thus  components 
are  separated  because  of  differences  in  their  affinity  for  the  adsorbent 
and  differences  in  their  boiling  points. 

INSTRUMENTATION 

A  typical  GLC  apparatus  consists  of  the  following  units: 

carrier  gas  — »  injector  — ->  column  packed  — >  detector  — >  signal 
(moving  system         with  solid  sup-        system  recorder 

phase)  port  coated  with 

liquid  phase; 

column  enclosed 

in  oven 

The  various  components  of  an  injected  sample  separate  into  discrete 
bands  in  the  gas  flow  and  move  through  the  column  at  different  veloc- 
ities. As  they  emerge,  they  pass  through  a  detector  system  and  a  signal 
is  sent  to  a  millivolt  recorder.  The  record  so  obtained  is  termed  the 
chromatogram.  A  typical  chromatogram  is  shown  in  Figure  9. 

The  time  lapse  measured  between  sample  injection  and  the  apex 
of  a  component  peak  is  termed  the  retention  time.  Frequently,  a  small 
positive  or  negative  recorder  response  at  the  point  of  injection  makes 
a  suitable  start  for  measurement.  If  this  response  is  not  obtained, 
measurements  may  be  made  starting  at  the  apex  of  the  solvent  peak. 
The  distance  may  be  measured  in  any  units  (cm.,  inches);  the  reten- 
tion time  is  then  calculated  from  this  measurement  and  the  speed  of 
the  recorder  chart.  Before  discussing  the  routines  for  making  qualita- 
tive and  quantitative  analyses,  each  unit  of  the  typical  GLC  instrument 
will  be  considered  in  a  little  more  detail. 


67 


62 


GAS-LIQUID  CHROMATOGRAPHY 

solvent 


injection    — i     ^ ^j 

of  sample  i  |  I 

retention 
time  for  A 

A  typical  gas  chromatogram. 


time' 


(Fig.  9) 


Carrier  gases.  The  selection  of  carrier  gas  is  dependent  upon  the 
type  of  detector,  as  shown  here: 


Detector 

Flame  ionization 
Thermal  conductivity 
/3-ionization 
Electron  capture 


Carrier  gas 

Nitrogen,  helium,  argon 
Hydrogen,  helium 
Argon 
Argon 


The  carrier  gas  must  be  pure,  inert,  and  dry.  It  is  best  to  dry  the 
gas  by  passing  it  through  a  molecular  sieve,  which  can  be  reacti- 
vated periodically  by  heating  at  approximately  350°C  for  four  hours 
with  the  carrier  gas  flowing  through  it.  Impure  or  wet  carrier  gas  will 
lead  to  such  problems  as  the  appearance  of  "ghost"  peaks,  decreased 
column  life,  baseline  drift,  and  noise. 

The  carrier  gas  should  pass  through  a  flow  regulator,  as  carrier 
gas  flow  rates  must  be  adjusted  to  achieve  optimum  efficiency.  Optimum 
flow  rates  will  vary  from  column  to  column. 


GAS-LIQUID  CHROMATOGRAPHY  63 

Injection  system.  The  usual  injection  system  consists  of  a  heated 
injection  port  with  a  self -sealing  rubber  or  silicone  septum  through 
which  the  sample  can  be  introduced  into  the  gas  stream  with  a  micro- 
liter syringe.  Most  injection  ports  allow  for  projection  of  the  column 
into  the  injection  area  so  that  on-column  injection  is  possible. 

The  injection  port  temperature  must  be  controlled  and  must  be 
high  enough  to  vaporize  samples.  The  injection  system  should  be  leak- 
proof  and  must  be  so  designed  as  to  prevent  back  flow  into  the  carrier 
gas  inlet.  The  velocity  of  gas  flow  through  the  injection  port  must 
sweep  the  vaporized  sample  rapidly  into  the  rest  of  the  system. 

Column  technology.  This  is  the  most  important  part  of  the  chroma- 
tograph.  Problems  can  often  be  traced  to  a  faulty  column.  The  column 
consists  of  three  parts:  the  container,  which  may  be  metal  (copper, 
stainless  steel)   or  glass;  the  solid  support;  and  the  stationary  phase. 

For  some  analyses,  including  the  analysis  of  sterols,  a  glass  column 
and  injection  port  are  essential,  since  metal,  especially  copper,  may 
catalyze  sample  breakdown.  In  general,  however,  stainless  steel  is 
satisfactory. 

The  purpose  of  the  solid  support  is  to  hold  the  liquid  phase.  The 
support  can,  however,  influence  events  occurring  within  the  column 
by  adsorbing  components  at  active  sites  or  by  catalyzing  reactions  due 
to  trace  metal  impurities.  The  former  problem  is  alleviated  by  treating 
the  support  with  a  silylating  reagent,  such  as  bis-trimethylsilylacetamide 
(BSA).  Acid  washing  will  remove  metals. 

The  choice  of  column,  solid  support,  and  liquid  phase  varies  with 
the  separations  being  attempted.  The  catalogs  of  all  leading  suppliers 
generally  recommend  specific  packings  for  various  types  of  separations. 
In  the  sections  that  follow,  suggestions  for  column  packings  are  in- 
cluded in  the  discussions  of  the  various  separations. 

Length  of  column  may  vary  from  a  few  inches  to  about  one  mile. 
The  latter  extreme  is  the  case  for  capillary  or  Golay  columns,  in  which 
the  support  and  liquid  phase  are  merely  coated  on  the  inside  of  capil- 
lary tubing.  Such  columns  are  especially  useful  in  some  tricky  situations 
involving  geometric  isomer  separations.  For  general  use,  columns  are 
18  inches  to  6  feet  long  and  U-shaped,  straight,  or  coiled,  depending 
on  the  instruments. 

The  temperatures  at  which  columns  are  used  depends  on  the  com- 
pounds being  analyzed  but  above  all  on  the  stability  of  the  stationary 
phase.  The  maximum  temperature  at  which  various  phases  are  stable 
is  reported  by  suppliers.  This  is  a  most  important  consideration  since 
excessive  bleed  of  liquid  phase  from  the  column  or  its  breakdown  will 
lead  to  problems,  including  fouling  of  the  detector  system.  The  per- 
centage of  liquid  phase  used  varies  from  as  little  as  1  percent  to  as 
much  as  30  percent;  most  analyses,  however,  are  achieved  with  con- 
centrations of   1   to   10  percent.  Before  use,  a  new  column  is  always 


64  GAS-LIQUID  CHROMATOGRAPHY 

"bled  out"  (usually  overnight  or  for  24  hours)  to  rid  the  column  sup- 
port of  excess  phase.  This  is  generally  done  with  the  column  detached 
from  the  detector. 

Columns  may  be  operated  isothermally  for  analysis  at  a  predeter- 
mined optimum  temperature,  or  the  temperature  may  be  programmed 
to  rise  from  a  given  lower  temperature  to  a  given  maximum  tempera- 
ture during  each  run.  The  rate  of  rise  of  temperature  may  vary  from 
3  to  18°C  per  minute.  Temperature  programming  is  especially  useful 
when  samples  with  a  wide  range  of  boiling  points  are  being  analyzed 
or  when  samples  contain  components  that  elute  over  a  long  period  of 
time.  Programming  is  not  merely  a  time  saver,  however;  it  also  im- 
proves the  general  symmetry  of  the  recorded  peaks.  As  we  shall  see, 
this  is  a  big  advantage  in  quantitative  analysis.  The  longer  a  com- 
ponent remains  on  a  column,  the  broader  its  band  becomes  as  it  dif- 
fuses during  passage  through  the  column.  The  result  is  loss  of  peak 
response,  since  small  amounts  of  the  diffused  band  (with  and  against 
the  direction  of  carrier  gas  flow)  are  not  detected.  Instead,  the  peak 
may  appear  as  a  long  hump  on  the  baseline.  Temperature  programming 
reduces  the  length  of  time  that  high-boiling  and  slow-eluting  compo- 
nents remain  on  the  column,  and  the  decrease  in  diffusion  leads  to 
sharp,  well-defined  peaks. 

Detector  systems.  The  separation  achieved  by  the  column  must  be 
translated  into  an  electrical  signal  which  can  be  fed  into  a  recorder 
and  used  for  qualitative  and  quantitative  analysis.  The  translation  is 
done  by  a  detector.  The  "perfect  detector,"  one  which  responds  linearly 
and  identically  to  any  amount  of  any  type  of  compound,  does  not  exist. 
Each  detector  has  a  finite  range  within  which  response  changes  linearly 
with  the  quantity  of  components.  This  is  called  the  linear  dynamic 
range  (LDR)  of  the  detector.  If  the  detector  is  used  outside  its  LDR, 
the  peak  shapes  become  distorted  and  reproducibility  decreases.  Detec- 
tor responses  also  depend  on  the  functional  groups  on  the  component 
being  analyzed.  An  equal  number  of  moles  of  a  hydrocarbon  and  a 
fatty  acid  methyl  ester  will  not  give  the  same  signal.  Some  calibration 
of  the  detector  response  is  required  for  every  compound  being  analyzed 
quantitatively.  These  and  other  factors  involved  in  detector  technology 
will  be  discussed  for  each  type  of  detector  considered. 

Of  the  many  kinds  of  detectors,  the  most  commonly  used  are  the 
thermal  conductivity  cells  and  the  ionization-type  detectors.  Thermal 
conductivity  (TC)  detectors  detect  changes  of  thermal  conductivity  in 
the  carrier  gas  when  it  becomes  diluted  by  components  in  the  sample. 
The  more  highly  conducting  the  carrier  gas  is,  the  greater  will  be  the 
thermal  conductivity  change  when  a  component  enters  the  TC  cell. 
Hydrogen  and  helium  are  recommended  carrier  gases.  When  the 
thermal  conductivity  of  the  component  is  less  than  that  of  the  carrier 
gas,  sensitivity  is  good  but  will  diminish  as  the  thermal  conductivity 


GAS-LIQUID  CHROMATOGRAPHY  65 

approaches  that  of  the  carrier  gas ;  response  will  be  a  negative  peak  if 
the  thermal  conductivity  of  the  carrier  gas  is  exceeded. 

A  filament  heated  by  direct  current  is  usually  the  varying  resistance 
element.  Two  of  these  elements  or  two  matched  pairs  of  elements  are 
placed  in  two  streams  of  carrier  gas;  they  are  called  the  reference  and 
the  sensor  elements.  The  temperature  of  the  elements  depends  on  the 
carrier  gas  conductivity,  its  flow  rate,  and  the  temperature  of  the  detec- 
tor block.  To  decrease  noise  and  increase  cell  life,  filament  detector 
cells  must  be  run  as  cool  as  possible,  consistent  with  the  boiling  points  of 
the  sample  components. 

The  typical  circuitry  for  a  TC  detector  is  a  Wheatstone  bridge  used 
as  a  temperature  sensing  bridge.  When  a  component  dilutes  the  carrier 
gas  and  passes  with  it  over  the  sensor  element,  the  element  heats  up 
and  its  resistance  increases.  As  a  consequence,  there  is  an  out-of- 
balance  signal  between  the  sensor  element  and  the  reference  element 
(over  which  pure  carrier  gas  flows).  This  signal  is  transposed  to  a 
voltage  output  and  fed  to  the  recorder. 

The  most  popular  type  of  TC  detector  employs  the  hot  wire  element. 
This  type  of  detector  has  a  wide  temperature  range,  does  not  destroy 
the  component  (which  can,  therefore,  be  collected  and  fed  into  other 
instruments  such  as  infrared  spectrophotometers  or  mass  spectrom- 
eters), and  has  no  selectivity  so  far  as  any  particular  type  of  com- 
pound is  concerned.  If  conditions  are  favorable,  a  hot  wire  detector 
can  measure  as  little  as  0.1  fig. 

The  thermistor,  another  type  of  varying  resistance  element,  is  in- 
frequently used  now  except  for  gas  analysis.  It  is  very  efficient  in  this, 
measuring  in  the  parts  per  million  range.  Sensitivity  decreases  above 
150°C. 

Ionization  detectors  have  more  complicated  circuitry  than  TC  detec- 
tors, are  more  trouble  to  maintain,  and,  since  they  show  selectivity  of 
response,  require  more  detailed  calibration  and  sample  handling.  They 
are,  however,  very  sensitive,  giving  in  some  cases  full-scale  recorder 
deflection  in  response  to  1  picogram  (10~12  g)  of  component. 

The  flame  ionization  detector  (FID)  is  one  of  the  most  popular 
detectors.  It  measures  the  minute  current  developed  when  a  combustible 
material  in  carrier  gas  enters  a  hydrogen-air  flame.  A  d.c.  potential 
is  applied  across  the  flame  by  a  pair  of  electrodes.  As  the  component 
burns,  the  electrodes  collect  the  charged  species  and  the  resulting  cur- 
rent is  amplified  by  an  electrometer  and  led  to  a  recorder.  Routine  use 
allows  analysis  of  10  nanograms  (1  nanogram  =  10~9  g),  and  under 
favorable  conditions  0.1  nanogram  can  be  measured.  Since  response 
is  a  function  of  the  number  of  carbon  atoms  and  the  groups  in  which 
they  appear,  the  detector  must  be  standardized  and  calibrated  to  handle 
a  specific  type  of  sample.  This  detector  has  some  degree  of  selectivity 
since  poorly  combustible  materials  such  as  carbon  tetrachloride  give 
weak  signals;  the  detector  is  also  not  suitable  for  the  analysis  of  many 


66  GAS-LIQUID  CHROMATOGRAPHY 

gases.  However,  the  FID  detector  is  not  so  easily  damaged  as  the  TC 
detector  and  is  readily  cleaned. 

Another  type  of  ionization  detector  is  the  Argon,  Beta  Ray,  or 
Alpha  Ray  type.  The  usual  name  is  the  Argon  Ionization  Detector, 
since  argon  is  the  commonly  used  carrier  gas  for  these  detectors;  Beta 
Ionization  Detector  is  also  frequently  used,  whether  the  ionizing  source 
emits  /3-  or  a-particles.  In  this  detector,  radiation  from  an  isotope 
source  ionizes  a  small  percentage  of  the  carrier  gas  (argon)  atoms, 
giving  rise  to  an  "ionization"  or  a  "standing"  current.  The  isotope 
sources  are  strontium  90  or  tritium  (/3-particle  emitters)  or  radium 
226  (an  a  emitter).  A  few  argon  atoms  receive  less  than  enough  energy 
to  be  ionized  but  enough  to  displace  an  electron  from  its  normal  orbit 
or  shell  —  about  11.6  electron  volts.  Such  atoms  remain  in  their  meta- 
stable  state  until  something  collides  with  them.  If  one  collides  with  a 
sample  component  that  needs  less  than  11.6  ev  to  be  ionized,  ionization 
will  take  place  and  another  electron  is  set  free  in  the  ionization  cell. 
If  a  polarizing  voltage  is  applied  across  the  cell,  these  ionized  sample 
molecules  will  increase  the  ionization  (or  "standing")  current.  Increas- 
ing the  cell  voltage  increases  the  number  of  metastable  atoms  as  fol- 
lows: the  electrons  composing  the  ionization  current  accelerate  more 
rapidly;  the  kinetic  energy  of  the  electrons  is  transferred  to  the  argon 
atoms  during  collisions,  resulting  in  more  transfers  of  11.6  ev ;  and 
more  metastable  atoms  are  formed  which,  in  turn,  ionize  more  sample 
molecules.  Therefore,  raising  the  cell  voltage  (and  hence  the  number 
of  metastable  argon  atoms  per  unit  volume)  results  in  a  higher  ioniza- 
tion efficiency  for  sample  molecules.  As  with  other  detectors,  changes 
in  ionization  current  are  fed  to  a  recorder. 

Still  another  type  of  ionization  detector  is  in  common  use  for 
certain  types  of  analysis:  the  Electron  Capture  (or  Attachment) 
Detector  (ECD).  Here  /^-particles  generate  an  atmosphere  of  electrons 
and  positive  ions.  The  electrons  are  collected  at  the  cell  anode.  There 
is  a  low  voltage  on  the  electrodes  when  only  carrier  gas  is  in  the  cell. 
If  sample  molecules  which  readily  accept  electrons  enter  the  cell,  the 
low-energy  electrons  may  attach  themselves  to  the  molecules.  Negative 
ions  of  sample  components  and  positive  argon  ions  combine  much 
faster  than  electrons  and  positive  ions,  hence  components  which  have 
a  high  electron  affinity  reduce  the  primary  ionization  current.  This 
detector  is  much  more  selective  than  any  we  have  discussed  so  far, 
particularly,  for  example,  towards  halogenated  compounds.  For  this 
reason,  the  ECD  is  much  used  by  investigators  measuring  small  quan- 
tities of  chlorine-containing  pesticides.  ECD  sensitivities  are  in  the 
picogram  range. 

QUALITATIVE  ANALYSIS 

If  it  is  desired  to  analyze  completely  unknown  samples,  GLC  is 
not  a  suitable  technique  when  used  alone ;  other  techniques  such  as 


GAS-LIQUID  CHROMATOGRAPHY  67 

infrared  and  mass  spectroscopy  must  also  be  used.  One  must,  there- 
fore, have  some  idea  of  the  type  of  compound  being  investigated.  The 
only  definitive  property  of  a  compound  measured  by  GLC  is  its  reten- 
tion time,  and  that  only  to  the  extent  that  all  conditions  are  carefully 
standardized  during  the  chromatographic  run.  Temperature,  gas  flows, 
etc.,  should  be  controlled  carefully;  if  temperature  is  programmed, 
the  rate  of  rise  should  be  uniform.  Also,  the  retention  time  should  be 
measured  from  some  reproducible  point,  for  example,  as  we  previously 
noted,  from  the  solvent  peak.  The  start  may  be  taken  as  the  leading 
edge  of  the  solvent  peak  or  its  apex,  whichever  is  more  convenient. 
With  a  TC  detector  the  air  peak  is  frequently  a  suitable  starting  point. 
Retention  time  (RT)  itself  varies  with  several  instrument  parameters, 
including  column  temperature,  column  length,  flow  rate  of  carrier  gas, 
type  of  liquid  phase,  loading  of  sample,  and  weight  of  column  packing. 
This  makes  RT  not  too  reliable  a  figure  for  correlating  data  obtained 
at  different  times  or  in  different  laboratories.  A  more  reliable  param- 
eter is  relative  retention  time  (RRT),  the  ratio  of  the  RT  of  a  known 
standard  component  to  the  RT  of  the  sample. 

If  one  has  some  preliminary  information  about  the  unknown  ob- 
tained by  other  means  (having  determined  functional  groups  by  spot 
tests,  infrared  analysis,  etc.),  one  may  proceed  with  some  GLC  analy- 
sis. For  example,  suppose  that  preliminary  analysis  indicates  that  the 
sample  is  a  mixture  of  aliphatic  alcohols:  then  the  number  of  com- 
ponents, their  chain  length,  etc.  can  be  determined  by  GLC.  Chain 
lengths  are  determined  by  utilizing  the  fact  that  for  various  members 
within  a  chemical  class  (the  n-alcohols,  the  ethers,  the  ketones,  the  fatty 
acids,  etc.)  log  RT  varies  linearly  with  molecular  weight.  Thus,  if  one 
chromatographs  a  series  of  saturated  n-alcohols  on  a  suitable  phase 
(silicone  or  carbowax),  measures  retention  time,  and  plots  log  RT 
against  the  number  of  carbon  atoms  in  the  alcohol,  a  straight  line  is 
obtained  (Fig.  10).  A  series  of  straight  line  graphs  can  be  obtained 
from  the  mono-,  di-,  and  tri-unsaturated  species,  and  so  on.  Thus,  the 
possession  of  a  series  of  standards  enables  one  to  test  the  unknown 
for  fit  on  one  of  the  curves.  Ideally,  unknowns  and  standards  should 
be  run  on  the  same  day  and,  of  course,  under  standardized  conditions 
of  column,  flow  rate,  and  temperature. 

In  addition  to  determining  functional  groups  by  conventional  means, 
it  is  often  possible  to  use  GLC  data  to  find  out  what  type  of  compound 
one  is  dealing  with.  This  requires  collecting  RRT  data  for  a  large  num- 
ber of  functional  group  classes  on  at  least  two  standard  columns,  such 
as  silicone  and  carbowax.  Because  of  the  linearity  of  homolog  retention 
on  the  two  columns,  a  plot  similar  to  that  in  Figure  11  can  be  obtained. 
Thus,  by  injecting  an  unknown  on  two  columns,  a  compound  can  be 
identified  that  fits  several  of  the  prepared  standard  plots. 

The  utility  of  GLC  is  enhanced  when  combined  with  a  number  of 
other  analytical  procedures;   in  particular,   chromatographing  an  un- 


68 


GAS-LIQUID  CHROMATOGRAPHY 


c 
o 

c 

u 
bJD 

o 


number  of  carbon  atoms 
Results  of  chromatography  of  homologs. 


(Fig.  10) 


c 
o 

H 


esters 


ketones 


alcohols 


RRT  on  carbowax  column 
Chromatography  of  homologs  on  two  columns. 


(Fig.  11) 


known  before  and  after  a  specific  chemical  treatment  designed  to  remove 
or  modify  specific  groups  yields  information  in  the  form  of  missing  and 
enlarged  peaks.  Treating  the  sample  vapor  with  sodium  metal  will,  for 
example,  leave  only  hydrocarbons  and  ethers;  hydrogenation  (with 
platinum  oxide  as  a  catalyst)  will  saturate  unsaturated  compounds ; 
treatment  with  hydrochloric  acid  will  leave  only  neutral  and  acidic 
compounds. 

In  summary,  GLC  alone  is  never  useful  for  the  characterization  of 
complete  unknowns;  used  in  conjunction  with  other  analytical  tech- 


GAS-LIQUID  CHROMATOGRAPHY  69 

niques,  it  can  be  a  useful  aid.  For  qualitative  analysis,  GLC  is  most 
useful  when  linked  to  such  analytical  tools  as  infrared  and  mass 
spectrometers.  In  such  cases,  characteristic  "fingerprints"  of  each 
GLC  peak  (on  different  columns)  can  be  obtained,  and  frequently 
quite  complex  unknowns  can  be  characterized. 

QUANTITATIVE  ANALYSIS 

Gas  chromatography  is  an  extremely  powerful  tool  for  separation 
and  quantitation  of  complex  mixtures.  It  has  revolutionized  lipid  chem- 
istry in  the  last  15  years.  It  is,  for  example,  possible  with  GLC  to 
analyze  mixtures  of  dozens  of  fatty  acids  in  a  relatively  short  time, 
an  analysis  which  in  the  past  would  have  taken  months  and  even  then 
probably  would  not  have  included  trace  components. 

Certain  rules  must  be  rigidly  observed  during  quantitative  analysis 
by  GLC,  and  time  must  be  taken  to  standardize  conditions.  A  little 
time  spent  in  the  beginning  will  pay  dividends  later  in  the  form  of 
smooth-running,  routine,  accurate  analyses. 

The  first  essential  is  investment  in  a  set  of  highly  purified  standards. 
These  standards  should  be  analyzed  to  determine  the  linear  dynamic 
range  and  response  of  the  instrument.  Correction  factors  should  then 
be  calculated  for  all  types  of  columns  to  be  used.  Standards  should  be 
run  periodically  to  check  for  changes  in  the  instrument;  they  should 
always  be  run  whenever  a  new  column,  new  detector,  or  any  new 
electronics  part  is  fitted. 

Before  standardization  of  methods  can  begin,  a  method  of  peak 
measurement  must  be  selected.  The  two  methods  available  are  peak 
height  and  peak  area. 

The  measurement  of  peak  height  is  the  easier  method  provided 
that  clear,  sharp  peaks  are  routinely  obtained.  In  practice,  this  is  seldom 
achieved  for  all  compounds  except  when  programmed  temperature  is 
employed.  Peak  height  measurement  under  other  conditions  requires 
much  standardization  and  is  not  recommended. 

A  number  of  techniques  for  measuring  peak  area  are  available: 

(a)  Disc  integrator.  This  is  an  electromechanical  device  attached  to  the 
recorder.  Over  the  bottom  10  percent  of  the  recorder  chart  paper,  the 
integrator  pen  travels  with  a  speed  proportional  to  the  displacement  of 
the  recorder  pen.  This  technique  gives  an  accuracy  slightly  better  than 
that  in  method  (d)  described  below.  It  is  especially  useful  when  peaks 
are  asymmetrical.  It  has  the  added  advantage  that  a  permanent  record 
of  quantitation  is  available  on  the  original  chromatogram.  Peaks  must 
stay  on  scale  with  this  method,  so  integrator  settings  must  be  adjusted 
during  a  run  or  must  be  predetermined. 

(b)  Electronic  integrator.  These  integrators  usually  integrate  peaks 
and  print  out  peak  area  and  retention  time.  This  is  the  quickest  and  most 


70  GAS-LIQUID  CHROMATOGRAPHY 

accurate  method  of  peak  area  measurement.  Usually  these  devices 
correct  for  baseline  drift,  determine  areas  of  peaks  that  are  incom- 
pletely resolved,  and  operate  independently  of  the  recorder  so  that  off- 
scale  peaks  are  still  measured.  Not  unexpectedly,  these  devices  are 
quite  costly. 

(c)  Planimetry.  Peak  area  may  be  determined  by  tracing  the  peak 
periphery  with  a  planimeter,  a  device  that  mechanically  integrates  the 
peak  and  records  the  area  digitally  on  the  planimeter  dial.  This  tech- 
nique can  be  quite  accurate,  depending  upon  the  operator's  skill. 

(d)  "Height  times  width  at  one-half  height"  method.  Using  this  pro- 
cedure one  must  first  construct  a  baseline  by  drawing  a  line  with  a 
ruler  across  the  bottom  of  a  peak,  making  the  best  connection  between 
the  peak's  leading  and  trailing  edge.  The  peak's  height,  half-height,  and 
width  at  one-half  height  are  determined.  The  area  is  the  product  of 
the  height  and  the  width  at  one-half  height.  In  other  words,  it  is  as- 
sumed that  the  peak  is  essentially  a  triangle.  The  usual  formula  for 
area  of  a  triangle  (1/2  bh)  is  not  used,  since  the  length  of  the  base 
of  a  broad  peak  is  often  difficult  to  determine.  This  method  works  best 
when  peaks  are  well  resolved  and  fairly  sharp  and  symmetrical.  In- 
creasing the  speed  of  the  recorder  chart  often  improves  peaks  for 
this  method. 

(e)  Triangulation.  In  this  method  a  triangle  is  constructed  by  drawing 
tangents  to  the  peak  sides.  The  apex  of  the  triangle  will  appear  above 
the  peak  apex,  but  this  allows  for  the  area  lost  by  drawing  tangents  to 
the  sides.  The  base  of  the  triangle  is  defined  by  the  intercept  of  the 
tangents  with  the  base  line  drawn  across  the  bottom  of  the  peak.  The 
area  of  the  peak  is  determined  by  the  usual  formula  for  area  of  a 
triangle,  J/2  bh.  Trouble  is  encountered  with  this,  as  with  the  previous 
procedure,  when  peaks  are  asymmetrical  or  when  peaks  are  incom- 
pletely resolved  (the  recorder  pen  does  not  return  to  the  baseline  be- 
tween peaks).  One  way  to  overcome  this  is  to  measure  the  peak  width 
at  two  points  and  calculate  the  area  according  to  the  formula: 

area  =  l/2  X  height  X  (width    at   0.2   height  +  width   at    0.8   height). 

This  procedure  is  said  to  compensate  for  peak  asymmetry  and  provide 
a  more  accurate  area. 

(f)  "Paper  doll"  technique.  In  this  method  the  peaks  are  cut  out  and 
weighed.  This  gives  good  reproducibility  and  is  excellent  for  asymmetric 
peaks.  The  disadvantages  are  that  the  chromatogram  is  destroyed  and 
the  method  is  time  consuming. 

Standardization  Procedures  for  Quantitative  Analysis 

Purity  of  standard  compounds  should  first  be  assessed.   As  with 
all  analyses,  the  standards  should  be  run  on  two  columns,  one  with  a 


GAS-LIQUID  CHROMATOGRAPHY  71 

polar  liquid  phase  (such  as  ethylene  glycol  succinate,  EGS)  and  one 
with  a  nonpolar  phase  (SE-30  or  an  Apiezon).  If  the  component  gives 
a  full-scale  deflection  on  the  recorder  chart  without  the  appearance 
of  impurity  peaks  and  if  it  has  the  correct  RRT  (on  both  columns), 
checked  by  referring  to  the  literature,  then  it  is  about  99  percent  pure. 
This,  of  course,  assumes  that  impurities  separate  from  the  main  com- 
ponent and  is  why  at  least  two  columns  should  be  used  to  assess  purity. 
If  this  condition  is  satisfied,  then  10  times  the  amount  can  be  chro- 
matographed  to  establish  99.9  percent  purity.  Generally  speaking,  99  per- 
cent purity  is  acceptable  and  the  above  procedure  is  adequate,  provided 
that  the  standards  have  been  purchased  from  one  of  the  established 
suppliers.  If  the  source  of  the  standard  is  in  some  doubt,  however, 
then  establishing  purity  by  GLC  alone  is  insufficient.  It  must  be  re- 
membered that  the  impurity  may  not  be  revealed  by  GLC.  In  such 
cases,  it  is  advisable  to  use  other  approaches;  for  example,  TLC  in 
two  different  systems  may  be  used  to  determine  if  only  a  single  com- 
ponent is  detected.  If  a  lot  of  material  is  available,  the  compound 
should  be  examined  by  some  other  procedure  such  as  characteristic 
infrared  spectra,  etc. 

In  the  case  of  lipids  it  is  seldom  necessary  to  go  to  other  analytical 
aids  to  assess  purity  when  the  source  is  one  of  the  well-known  lipid 
suppliers  (such  as  Applied  Science  Laboratories,  Mann  Chemical  Cor- 
poration, Sigma  Chemical  Corporation,  Supelco,  Inc.).  These  suppliers 
state  the  purity  of  their  products  and  are  on  the  whole  reliable.  Often 
chromatographic  proof  of  purity  is  supplied  with  the  more  expensive 
standards.  However,  spot  GLC  checks  should  be  run.  If  the  compound 
is  rare,  detailed  proof  of  purity  should  be  obtained. 

In  place  of  the  check  on  RRT's  from  the  literature,  another  prop- 
erty, called  the  Carbon  Number  (CN)  may  be  used.  Within  reasonable 
limits  CN's  are  accurately  reproducible  for  a  particular  liquid  phase. 
The  CN  of  a  compound  is  found  in  the  following  way.  Using  fatty 
acid  methyl  esters  as  an  example,  assume  that  at  least  four  standard 
esters  have  been  run  on  a  polar  (EGS)  and  nonpolar  (SE-30)  column 
and  that  the  retention  times  have  been  plotted  on  semilog  graph  paper 
versus  the  number  of  carbon  atoms  (here  called  the  CN)  in  the  ester. 
Figure  12  shows  two  typical  graphs.  If  another  ester,  say  methyl 
oleate,  is  now  run  on  a  column  and  its  RT  calculated,  its  CN  can  be 
read  off  the  graph  as  shown  by  the  dotted  line.  Lists  of  CN's  are 
found  in  the  literature  and  can  be  used  both  to  help  establish  purity 
of  standards  and  to  identify  fatty  acids  in  a  series  being  analyzed.  It 
is  advisable  to  establish  the  CN  on  both  a  polar  and  a  nonpolar  column. 

Once  purity  of  standards  is  established,  the  Detector  Response 
(DR)  and  Linear  Dynamic  Range  (LDR)  should  be  determined. 

Detector  response  must  be  determined  so  that  the  relationship 
between  the  amount  of  sample  injected  and  the  response  of  the  detector 


72 


GAS-LIQUID  CHROMATOGRAPHY 


o 


bJD 
O 


carbon  number 


carbon  number 


(a)  on  EGS 


fbJ  on  SE-30 


Determining  carbon  numbers  (CN's)  by  chromatography  on  two  columns. 

(Fig.  12) 


is  known.  Unless  good  DR  is  obtained,  analysis  of  small  samples  is 
not  possible.  DR  varies  with  detector  type,  column,  instrument  make, 
and  type  of  compound.  To  determine  DR,  a  weighed  mixture  of  stan- 
dards A,  B,  C,  and  D  is  chromatographed.  The  weight  injected  must 
be  known,  and  dilution  of  the  mixture  in  solvent  and  measurement  of 
injected  volume  (using  a  10/aI  Hamilton  Syringe)  must  be  accurate. 
The  injection  should  be  repeated  several  times  to  check  for  repro- 
ducibility. The  injection  should  be  done  steadily,  but  quickly,  and  the 
septum  on  the  injection  port  must  be  leak  free.  After  obtaining  the 
chromatogram,  the  area  of  each  peak  is  determined.  The  response  of 
the  detector  to  each  compound  per  unit  weight  is  then  determined.  The 
adjusted  area  of  detector  response  is  calculated  from  the  relationship 
DR  =  area  -4-  weight.  The  response  ratio  is  then  calculated  by  selecting 
one  compound  as  having  a  response  of  unity.  Suppose  compound  B  is 
our  response  standard.  If  one  microgram  of  B  gave  peak  area  of  352 
units  (352  units  per  /xg),  then  we  divide  the  other  DR's  by  352  in  order 
to  obtain  the  ratio  of  response  per  jug  to  compound  B.  Alternatively,  a 
factor  can  be  obtained  by  dividing  the  other  DR's  by  the  DR  of  the 
compound  selected  as  unity.  Peak  areas  obtained  on  the  chromatogram 
can  then  be  corrected  for  variation  in  detector  response  by  multiplying 
by  the  correction  factor  (or  the  response  ratio)   obtained. 

The  determination  of  response  factors  is  time  consuming,  but  in 
lipid  chemistry  some  short  cuts  are  usually  possible.  For  example, 
many  compounds  of  the  same  carbon  length  will  have  the  same  response 
factor.  While  the  response  factor  for  stearic  (C18:0)  acid  may  differ 
from  that  of  oleic  (C18:l)  acid,  other  unsaturated  acids  of  the  same 
chain  length  may  give  the  same  response  factor  correction  as  oleic  acid. 

Linear  Dynamic  Range  refers  to  the  sample  sizes  that  can  ap- 
propriately be  used  on  a  given  instrument,  from  the  minimum  detect- 


GAS-LIQUID  CHROMATOGRAPHY 


73 


concentration 
Typical  linear  dynamic  response  curve  for  a  given  instrument. 


(Fig.  13) 


able  quantity  to  the  point  of  overload.  Ideally,  if  1  jug  of  a  sample 
gives  a  peak  area  of  10  units,  10  /xg  should  give  100  units.  Also,  a  plot 
of  response  versus  concentration  should  pass  through  zero.  In  most 
cases  these  requirements  are  met,  but  the  instrument's  LDR  must  be 
checked.  A  linearity  curve  will  show  the  concentrations  at  which  the 
system  becomes  nonlinear.  In  addition,  any  interaction  of  the  sample 
with  the  system  (sample  loss)  will  show  up. 

A  typical  LDR  curve  is  shown  in  Figure  13.  Point  B  is  the  mini- 
mum detectable  level  (response  X  5  times  the  noise  level);  point  C  is 
the  upper  limit  of  linearity  (frequently  due  to  column  overload);  D 
is  a  typical  point  at  which  the  concentration  will  not  give  valid  results. 
An  LDR  curve  which  approximates  an  S-shape  and  has  a  narrow 
range  (say  from  A  to  B)  indicates  sample  adsorption  by  the  column 
or  other  parts  of  the  instrument.  This  type  of  LDR  curve  indicates  a 
need  for  silylation  of  the  column,  use  of  a  glass  rather  than  a  metal 
column,  or  both.  If  the  curve  crosses  the  Y-axis  above  zero,  this  can 
indicate  either  response  when  no  sample  is  injected  or  sample  measure- 
ment error  on  the  part  of  the  operator.  If  more  than  one  effect  is  present 
at  one  time  —  say  adsorption  plus  sample  measurement  error  —  a  very 
distorted  curve  will  be  produced,  and  all  possibilities  must  be  con- 
sidered. It  is  usually  wise  to  consider  sample  measurement  error  first 
before  changing  column  material. 

Now  that  DR  and  LDR  have  been  determined,  one  of  the  following 
standardization  techniques  can  be  chosen:  internal  normalization,  ex- 
ternal standardization,  or  internal  standardization. 

Internal  normalization.  In  this  procedure  the  quantity  of  a  com- 
ponent in  a  mixture  is  expressed  as  a  percentage  of  the  total  area  of 
the  chromatogram.  Thus,  if  the  sum  of  the  areas  of  all  the  peaks  is 
100  and  component  A  gives  an  area  of  10,  then  the  area  percentage 


74  GAS-LIQUID  CHROMATOGRAPHY 

for  A  is  10  percent.  In  calculating  areas,  the  response  factors  for  each 
component  should  be  used  to  correct  the  areas.  This  technique  is  in 
common  use  and  has  a  number  of  advantages  —  mainly,  speed  and 
independence  from  sample  size.  There  are,  however,  several  disad- 
vantages: first,  all  peaks  must  be  measured;  second,  absolute  measure- 
ments rely  on  the  assumption  that  the  entire  sample  chromatographs ; 
and  third,  for  complete  accuracy  response  factors  for  all  peaks  should 
be  known.  In  practice  these  disadvantages  are  not  very  serious.  Often 
in  lipid  chemistry  one  is  attempting  to  completely  analyze  a  series  of 
monoglycerides,  fatty  acid  methyl  esters,  etc.,  so  one  wishes  to  measure 
all  peaks  anyway.  Such  samples  are  usually  prepurified  (by  TLC  or 
some  other  method),  and  all  components  usually  chromatography  In- 
deed, it  is  common  practice  to  convert  peak  area  percentages  into  moles 
using  the  molecular  weight  —  which  assumes  that  everything  chro- 
matographs. Usually  this  is  safe,  but  the  investigator  should  ascertain 
that  no  adsorption  is  occurring. 

Internal  normalization  is  the  most  commonly  used  technique  in 
GLC  of  fatty  acid  methyl  esters  and  of  dimethyl  acetals  (and  other 
derivatives)  of  fatty  aldehydes;  it  is  also  used  in  other  lipid  GLC  work. 

External  standardization.  This  technique  demands  that  a  number  of 
analyses  of  both  the  standard  and  sample  be  made  and  averages  deter- 
mined. The  concentration  of  the  standard  should  be  near  that  of  the 
sample  or  should  be  the  same  compound  as  the  sample  so  that  no 
response  factor  is  required.  Samples  of  known  size  must  be  injected; 
in  practice  this  is  hard  to  achieve,  and  the  procedure  is  generally  used 
only  when  a  single  peak  is  being  analyzed.  It  is  not  recommended  as 
a  quantitative  procedure  for  any  of  the  lipid  analyses  discussed  later 
in  this  section. 

Internal  standardization.  This  method  is  the  best  for  obtaining 
accurate  determinations  by  GLC  and  it  is  highly  recommended  when- 
ever conditions  permit  its  use.  An  example  of  internal  standardization 
in  cholesterol  analysis  by  GLC  is  discussed  in  Chapter  6  (p.  87). 

In  this  method,  a  known  amount  of  a  substance  that  is  not  present 
in  the  sample  is  added  to  an  aliquot  of  the  sample.  The  area  of  the 
peak  of  the  added  standard  is  determined  and  compared  with  the  area 
of  the  sample  component  of  interest.  When  the  component  is  1  to  100 
percent  of  the  sample,  it  is  usual  to  use  a  weight  of  internal  standard 
of  about  10  percent  of  the  weight  of  the  component;  for  trace  amounts, 
however,  the  standard  should  be  in  the  same  concentration  range  as 
the  components. 

It  is  not  always  possible  to  use  internal  standardization  since  suitable 
substances  for  standards  are  not  always  available.  The  standard  chosen 
must  fit  certain  specifications.  It  must  elute  from  the  column  well 
separated  from  all  sample  components  but  near  to  them.  It  must  have 


GAS-LIQUID  CHROMATOGRAPHY  75 

functional  groups  similar  to  that  of  the  sample  component  or  be  an 
appropriate  hydrocarbon.  It  must  be  stable  under  the  analytical  con- 
ditions and  not  react  with  sample  components.  Finally,  it  must  be 
sufficiently  nonvolatile  and  stable  to  permit  storage  in  solution  for  a 
significant  period  of  time. 

The  first  steps  in  internal  standardization  are  preparing  the 
standard  solution  and  determining  the  response  factor  (RF)  for  the 
sample  compound  relative  to  the  internal  standard.  A  mixture  of  the 
component  to  be  analyzed  and  the  internal  standard  is  prepared,  and 
the  RF  is  calculated  as  follows: 

_     weight  of  internal  standard  X  area  of  component  peak 
weight  of  component  X  area  of  internal  standard  peak 

Next,  a  sample  mixture  is  prepared  by  adding  a  known  amount 
of  the  internal  standard  to  a  known  amount  of  the  sample  containing 
the  component  of  interest.  This  mixture  is  then  chromatographed 
under  the  same  conditions  as  the  standard  and  the  percent  of  the 
component  by  weight  is  calculated  by  the  equation: 

component  area  X  internal  standard  weight  X  100 
^  RF  X  internal  standard  area  X  sample  weight 

Following  the  selection  of  an  internal  standard,  it  is  necessary  to 
determine  the  response  characteristics  of  standard  mixtures.  A  response 
versus  concentration  curve  should  be  reproducible  and  preferably 
linear.  The  linearity  plot  should  be  linear  over  a  wide  concentration 
range  and  should  finally  plateau.  In  preparing  this  curve,  the  concen- 
tration of  the  internal  standard  is  kept  constant  and  only  the  concen- 
tration of  the  sample  is  varied. 

CHEMICAL  MODIFICATION  OF  COMPOUNDS  FOR  ANALYSIS 
BY  GAS-LIQUID  CHROMATOGRAPHY 

Many  lipids  can  now  be  analyzed  by  GLC  in  their  unmodified  state. 
Frequently,  however,  the  preparation  of  a  more  volatile  derivative, 
or  one  which  has  certain  functional  groups  blocked,  or  both,  leads  to 
more  efficient  chromatography.  Furthermore,  chromatography  of  both 
the  original  and  the  chemically  modified  form  will  provide  additional 
information  about  a  sample  and  may  serve  as  a  check  on  quantitative 
procedures. 

Analysis  of  Methyl  Esters  of  Fatty  Acids 

Fatty  acids  are  generally  analyzed  as  their  methyl  esters,1  although 
free  fatty  acids  can  be  chromatographed.  The  method  of  methyl  ester 

1  Other  derivatives  are  employed ;  however,  not  enough  is  known  about  the 
quantitation  of  yields  to  rank  these  as  competitors  with  methyl  esters. 


76  GAS-LIQUID  CHROMATOGRAPHY 

preparation  varies  with  the  source  of  material.  Transesterification  with 
acidic  methanol  is  in  common  use.  This  approach  gives  the  methyl 
ester  of  all  the  fatty  acids  in  lipids  from  biological  sources  whether 
they  are  in  amide  (sphingolipid)  or  ester  (glyceride  and  phosphoglycer- 
ide)  linkages.  Numerous  procedures  have  been  reported  and  a  partial 
list  of  suitable  reagents  follows. 

1.  1  to  10  percent  sulfuric  acid  in  methanol  at  60  to  100°C  for  1 
to  16  hours. 

2.  1  to  10  percent  hydrochloric  acid  I    .     --  ,.      Al  , 
or  5  percent  sulfuric  acid               j  +   2,2-dimethoxypropaue   (as 

a  water  scavenger)  in  methanol  at  50  to  70°C  for  2  to  4  hours. 

3.  3  to  14  percent  boron  trifluoride  in  methanol  at  60  to  100°C  for  1 
minute  to  16  hours. 

Some  workers  carry  out  reactions  by  refluxing  (under  nitrogen), 
while  others  seal  the  reaction  mixture  in  screw  cap  vials  or  glass 
ampules  and  heat  in  a  water  bath.  Definitely  avoid  using  hydrochloric 
acid-methanol,  since  this  reacts  to  yield  artifacts  which  mimic  fatty 
acid  esters  on  GLC  (1). 

The  method  we  recommend  uses  4  percent  sulfuric  acid  in  methanol 
in  a  sealed  ampule  at  90°C  for  2  hours.  Ten  to  500mg  of  sample  are 
placed  in  a  10ml  glass  ampule  and  an  appropriate  volume  (0.5  to  10ml) 
of  H2S04-methanol  is  added.  The  ampule  is  cooled  in  dry  ice-acetone 
and  sealed  in  an  oxygen  flame.  It  is  then  heated  for  2  hours  in  a  water 
bath  at  90°C  (Caution!  Do  not  heat  above  this  temperature  or  in  an 
oven  as  the  ampules  will  explode).  This  procedure  can  be  used  directly 
on  spots  removed  from  a  TLC  plate  if  a  noninterfering  detection  spray 
has  been  used.  After  heating,  the  ampule  is  again  cooled  in  dry  ice- 
acetone,  opened,  and  2  to  5ml  water  are  added  dropwise  until  two 
phases  appear.  The  methyl  esters  are  extracted  with  hexane  or  diethyl 
ether  (3X3  volumes). 

If  there  is  doubt  about  the  purity  of  the  methyl  ester  preparations, 
they  should  be  purified  by  TLC  (in  the  presence  of  an  antioxidant). 
For  example,  if  total  lipid  from  an  animal  source  is  transmethylated, 
the  methyl  esters  must  be  purified  by  TLC  since  cholesterol  derivatives 
that  mimic  fatty  acid  esters  on  GLC  may  appear ;  moreover,  aldehydes, 
derived  from  plasmalogens,  will  form  dimethylacetals  under  acid 
methanolysis  conditions.  These  must  be  separated  from  the  methyl 
esters  by  TLC  or  other  means  (see  pages  78-79). 

Generally,  a  good  instrument  will  give  a  good  resolution  of  the 
methyl  esters  of  most  natural  fatty  acid  mixtures,  and  numerous  column 
packings  are  available  for  this  analysis.  If  good  standards  are  avail- 
able and  semilog  plots  of  RT  versus  carbon  number  are  drawn  (see 
pages  71-72),  all  major  components  can  usually  be  identified.  The  ideal 
way  to  characterize  complex  mixtures  is  to  make  a  preliminary  separa- 
tion by  TLC  first  and  then  gas-chromatograph  each  fraction  separately ; 


GAS-LIQUID  CHROMATOGRAPHY  77 

for  example,  mixtures  may  be  separated  into  saturated  and  unsaturated 
fractions  on  silver  nitrate  impregnated  plates  as  described  earlier.  If 
only  small  quantities  of  material  are  available,  however,  this  approach 
may  involve  too  many  manipulations,  and  some  losses  may  occur. 

A  previously  uncharacterized  mixture  should  be  chromatographed 
on  both  a  polar  column,  such  as  ethylene  glycol  succinate,  and  a  non- 
polar  one,  such  as  one  of  the  Apiezon  or  SE-30  greases.  On  a  polar 
column,  it  is  usual  for  unsaturated  fatty  acid  esters  to  elute  after  the 
saturated  ones  of  the  same  carbon  chain  length,  and  branched  chain 
esters  are  eluted  just  before  the  straight  chain  ester  of  the  same  carbon 
chain  length.  On  nonpolar  columns,  unsaturated  esters  precede  the 
saturated  esters  of  the  same  carbon  chain  length. 

Some  coincident  elutions  often  occur,  and  on  many  columns  some 
unsaturated  acids,  especially  those  with  many  double  bonds,  may  elute 
at  the  same  time  as  a  long-chain  saturated  acid.  Uncharacterized  mix- 
tures must  be  checked  for  this  possibility.  If  such  mixtures  were  not 
initially  separated  by  TLC,  the  sample  should  be  treated  in  methanol 
with  platinum  oxide  as  a  catalyst2  to  hydrogenate  the  fatty  acid  esters. 
The  mixture  should  be  chromatographed  both  before  and  after  hydro- 
genation  and  on  more  than  one  kind  of  column.  The  appearance  of 
new  peaks,  the  change  in  size  of  old  peaks,  the  disappearance  of  old 
peaks,  and  so  forth  will  provide  information  about  the  presence  of 
any  unsaturated  species. 

The  presence  of  hydroxy  fatty  acid  methyl  esters,  eluted  from 
many  types  of  columns  under  certain  conditions,  can  confuse  identi- 
fication of  peaks.  Mixtures  containing  hydroxylated  fatty  acids  (for 
example,  cerebroside  and  sulfatide  fatty  acids)  should  be  separated 
into  hydroxylated  and  nonhydroxylated  ester  fractions  by  one  of  the 
TLC  or  column  chromatographic  procedures  described  on  pages  36 
and  48.  Derivatives  of  the  hydroxylated  esters  can  be  prepared  in 
which  the  free  hydroxy  groups  are  blocked.  GLC  of  these  derivatives  — 
as  trifluoroacetates,  methyl  ethers,  or  trimethylsilyl  ethers  —  gives  a 
much  more  rapid  analysis  and  better  peak  symmetry.  The  third  of 
these  derivatives  is  highly  recommended  and  easily  prepared.  Most 
suppliers  of  GLC  accessories  now  offer  a  variety  of  "silylating  kits," 
and  while  these  are  relatively  expensive,  they  generally  pay  for  them- 
selves in  the  form  of  time  saved.  Usually  all  the  investigator  does  is 
add  a  small  volume  of  the  silylating  agent;  after  a  short  waiting  period, 

2  Preferably,  the  hydrogen  should  be  supplied  via  a  manometer  so  that  hy- 
drogenation  can  be  continued  until  there  is  no  further  uptake  of  hydrogen.  The 
mixture  should  be  stirred  with  a  magnetic  stirrer  during  the  reaction.  A  simple, 
safe,  and  inexpensive  hydrogenation  apparatus  has  been  designed  by  Applied 
Science  Laboratories,  Inc.,  State  College,  Pa.  16801.  The  apparatus  is  not  offered 
for  sale,  but  it  is  described  in  Applied  Science's  Gas -Chromatography  Newsletter 
10,  No.  3  (1969).  Further  details  can  be  obtained  from  the  Biochemicals  Depart- 
ment at  Applied  Science. 


78  GAS-LIQUID  CHROMATOGRAPHY 

the  mixture  can  be  chromatographed  directly  without  having  to  extract 
the  derivative.  If  the  hydroxy  groups  are  sterically  hindered,  it  may 
be  necessary  to  heat  the  reaction  mixture. 

While  many  positional  isomers  of  fatty  acid  esters  separate  under 
average  conditions,  geometric  isomers  do  not.  Cis  and  trans  isomers 
can  be  separated  efficiently  with  Golay  (capillary)  columns  (see  p.  97). 
Another  approach  is  to  convert  the  isomers  to  hydroxylated  acids  by 
oxidizing  with  osmium  tetroxide  (2).  The  hydroxylated  compounds 
are  then  derivatized  by  one  of  the  procedures  mentioned  above  and 
chromatographed.  After  such  treatment,  the  erythro  and  threo  deri- 
vatives separate  on  the  usual  columns  used  for  methyl  ester  GLC. 

Quantitation  of  methyl  ester  analyses  may  be  carried  out  using  any 
of  the  peak  measuring  techniques  previously  described.  Generally,  it 
is  wise  to  obtain  response  correction  factors  for  all  the  saturated  esters 
and  for  at  least  one  unsaturated  ester  of  each  chain  length. 

For  details  of  the  GLC  of  some  rare  fatty  acids,  such  as  those  con- 
taining acetylenic  bonds,  Lipid  Chromatographic  Analysis,  vol.  1, 
should  be  consulted  (see  Suggested  Further  Readings). 

Fatty  Aldehydes 

Usually  the  lipid  chemist  encounters  fatty  aldehydes  in  the  form 
of  their  dimethyl  acetals  (DMA's),  derived  from  the  acid  hydrolysis  of 
plasmalogens.  While  aldehydes  can  be  analyzed  by  GLC  in  their  underi- 
vatized  form,  this  is  not  a  wise  procedure.  On  standing,  even  for 
short  periods,  aldehydes  readily  undergo  condensation  and  polymeriza- 
tion, reactions  that  are  accelerated  under  alkaline  conditions.  It  is  prefer- 
able to  obtain  aldehydes  in  a  stable  form,  such  as  the  DMA,  or  to 
convert  them  to  this  or  other  stable  derivatives  as  soon  as  possible.  The 
DMA  is  the  most  satisfactory  derivative  since  it  is  readily  formed  in 
quantitative  yield  and  is  very  stable  under  neutral  and  alkaline  condi- 
tions. Furthermore,  if  lipid  mixtures  are  subjected  to  acid  hydrolysis, 
fatty  acids  (whether  in  ester  or  amide  linkages)  are  converted  to 
methyl  esters  and  plasmalogen  aldehydes  are  converted  to  DMA's  all 
in  one  step.  Only  a  simple  separation  procedure  must  be  employed,  and 
both  sets  of  derivatives  can  be  subjected  to  GLC. 

H+  /0CH3 

R-CH0+2CH30H-*R-CH  +H20 

^0CH3 

dimethyl  acetal  (DMA) 

The  methyl  esters  and  DMA's  may  be  separated  either  by  TLC  on 
silica  gel  using  benzene  as  the  developing  solvent  or  by  taking  advan- 
tage of  the  fact  that  DMA's  are  stable  in  alkaline  solution  whereas 


GAS-LIQUID  CHROMATOGRAPHY  79 

methyl  esters  are  not.  In  the  latter  procedure,  the  methyl  ester-DMA 
mixture  is  refluxed  with  0.5N  methanolic  sodium  hydroxide  for  two 
hours.  The  methyl  ester  fatty  acids  will  be  converted  to  sodium  salts 
(soaps)  and  the  DMA's  will  remain  unchanged.  On  cooling,  the  solu- 
tion is  diluted  with  water  (1  vol.)  and  the  DMA's  are  extracted  three 
times  with  hexane  (1  vol.).  The  extract  is  washed  with  water-ethanol- 
3N  sodium  hydroxide  40:10:1  v/v/v  (5).  The  combined  hexane  ex- 
tracts are  dried  over  sodium  sulfate,  the  solvent  is  removed,  and  the 
acetals  are  reserved  for  GLC.  The  soaps  in  the  water  layer  are  con- 
verted to  free  fatty  acids  by  acidifying  the  solution  with  IN  hydro- 
chloric acid.  The  free  fatty  acids  are  then  extracted  with  hexane,  and, 
after  drying,  the  solvent  is  removed.  The  fatty  acids  may  then  readily 
be  reconverted  to  the  methyl  esters  by  any  of  the  usual  procedures 
(see  p.  76). 

Other  derivatives  of  long-chain  aldehydes  that  are  useful  for 
GLC  analysis  are  the  alcohol,  the  alcohol  acetate,  and  the  acid.  Far- 
quhar  (3)  used  all  these  derivatives,  in  addition  to  the  DMA's,  in  his 
detailed  study  of  the  fatty  aldehydes  of  erythrocyte  plasmalogens.  He 
prepared  the  free  aldehydes  by  dissolving  the  DMA's  in  90  percent 
acetic  acid  1:30  w/v  and  adding  1  drop  of  a  saturated  solution  of 
mercuric  chloride.  The  mixture  was  heated  in  a  sealed  ampule  (under 
nitrogen)  for  8  to  24  hours  at  37 °C.  The  aldehydes  were  recovered  as 
follows:  1  volume  of  water  was  added;  the  solution  was  neutralized 
with  3N  sodium  hydroxide  and  extracted  four  times  with  petroleum 
ether;  the  extracts  were  washed  once  with  water-ethanol-3N  sodium 
hydroxide  40:10:1  v/v/v  and  dried  over  sodium  sulfate.  The  solvent 
was  then  removed  and  the  aldehydes  were  converted  to  derivatives  as 
soon  as  possible. 

The  alcohols  were  prepared  by  reducing  3  to  30mg  of  the  free 
aldehyde  with  10ml  of  3  percent  lithium  aluminum  hydride  anhydrous 
ether  kept  at  — 20°C  for  2  hours.  ''Anhydrous"  is  stressed  as  the  re- 
action of  lithium  aluminum  hydride  with  water  is  extremely  violent. 
The  reaction  was  stopped  by  adding  5ml  of  water  drop  by  drop  to 
the  reaction  mixture  which  was  still  at  — 20°C.  Five  ml  of  ethanol 
were  added,  the  supernatant  ether  was  removed,  and  the  lower  layer 
was  extracted  5  times  with  petroleum  ether  at  room  temperature.  The 
combined  extracts  were  washed  with  water-ethanol  4:1  v/v,  dried,  and 
the  solvent  removed. 

One  to  lOmg  of  the  fatty  alcohols  were  converted  to  the  acetates  by 
dissolving  in  9ml  of  acetic  anhydride-pyridine  3:6  v/v  in  a  glass- 
stoppered  tube.  The  tube  was  kept  at  37 °C  for  15  minutes  and  shaken 
occasionally.  Five  ml  of  water  were  added  and  the  acetates  were  re- 
covered by  extracting  3  times  with  5ml  petroleum  ether. 

Conversion  of  the  free  aldehydes  to  the  fatty  acid  is  of  limited 
usefulness  since  the  usual  oxidation  procedures  destroy  the  unsaturated 


80  GAS-LIQUID  CHROMATOGRAPHY 

aldehyde.  Farquhar  (3)  employed  alkaline  silver  oxide  as  the  oxidizing 
agent.  Some  information,  however,  can  be  derived  from  the  fatty  acids 
obtained  from  the  aldehydes  derived  from  the  DMA  mixture  before 
and  after  hydrogenation.  The  DMA's  can  be  hydrogenated  in  methanol 
using  platinum  oxide  as  a  catalyst.  It  is  usually  necessary  to  add  chloro- 
form to  the  reaction  mixture  (up  to  20  parts  by  volume)  to  keep  the 
DMA's  in  solution.  The  fatty  acid  derivatives  are  generally  prepared 
only  when  additional  structural  information  is  sought.  For  most  pur- 
poses, the  preparation  of  the  DMA's  or  the  alcohol  acetates  suffices 
for  the  GLC  analysis  of  fatty  aldehydes.  If  fatty  acid  derivatives  are 
used,  they  can  be  converted  to  their  methyl  esters  for  GLC  analysis. 

A  number  of  liquid  phases  are  suited  to  the  GLC  analysis  of  DMA's 
and  alcohol  acetates.  It  is  most  important  to  remember  that  acidic 
columns  must  not  be  employed,  as  the  DMA's  will  readily  be  hydrolyzed 
in  acid  conditions.  Farquhar  used  ethylene  glycol  adipate  (EGA)  and 
Apiezon  M  for  the  analysis  of  the  acetates,  the  DMA's,  and  the  methyl 
esters.  Other  useful  phases  are  ethylene  glycol  succinate  (EGS), 
ethylene  glycol  succinate-silicone  copolymer  (EGSSX),  Reoplex  400, 
and  Apiezon  L.  Usually  10  to  15  percent  EGA,  EGS,  EGSSX  (polar) 
packings  or  10  to  12.5  percent  nonpolar  packings  is  employed.  Alkaline 
washed  supports  are  recommended.  Temperatures  used  with  average 
columns  (6  to  10  ft.  X  1/6 -inch)  are  about  150°C  for  polar  phases  and 
190° C  with  nonpolar  phases. 

Aldehydes  are  not  readily  available  in  pure  form,  so  standards  for 
these  analyses  can  present  a  problem.  It  is  usual  to  use  relative  retention 
data  and  to  refer  all  retention  times  to  the  normal  C18  saturated  hydro- 
carbon (octadecane)  or,  preferably,  the  corresponding  16-carbon  deriv- 
ative of  hexadecanal  (palmitaldehyde).  The  palmitaldehyde  is  available 
in  a  relatively  pure  state  as  its  bisulfite.  The  bisulfite  is  converted  to  the 
DMA  by  heating  with  4  percent  sulfuric  acid  in  methanol,  or  the  free 
aldehyde  is  obtained  by  aqueous  acid  hydrolysis.  The  DMA  preparation 
can  be  freed  from  aldehyde  by  shaking  the  extract  with  a  saturated  solu- 
tion of  sodium  metabisulfite.  The  DMA  extract  should  also  be  washed 
with  an  alkaline  wash  solution  (water-ethanol-3N  sodium  hydroxide 
40:10:1  v/v/v)  according  to  Farquhar's  procedure  to  prevent  acid- 
catalyzed  breakdown.  Retention  times  of  components  are  stated  as  rela- 
tive to  hexadecanal,  hexadecanal  acetate,  hexadecanal  DMA,  or  methyl 
hexadecanoate,  according  to  the  derivative  chromatographed,  since 
long-chain  aldehyde  GLC  data  is  generally  so  reported  in  the  literature ; 
this  permits  ready  comparisons  with  the  work  of  others. 

On  polar  columns,  the  order  of  elution  of  fatty  aldehydes  and  their 
derivatives  of  the  same  carbon  chain  length  (unbranched)  is:  free 
aldehyde,  DMA,  methyl  ester  of  fatty  acid,  alcohol  acetate,  free  alcohol. 
On  a  nonpolar  column  the  order  is:  free  alcohol,  methyl  ester,  DMA, 
and  alcohol  acetate.  Free  aldehyde  is  omitted  from  the  last  series  be- 


GAS-LIQUID  CHROMATOGRAPHY  81 

cause  according  to  Farquhar,  it  was  not  eluted  from  an  alkaline  treated 
nonpolar  column.  More  recently,  Gray  (4)  has  reported  aldehyde 
chromatography  on  untreated  Apiezon  L,  but  under  these  conditions 
breakdown  of  DMA's  occurred. 

Peak  area  calculations  may  be  made  by  any  of  the  methods  previ- 
ously described. 

Glycerides 

The  beginner  in  gas  chromatography  can,  after  a  short  practice 
period,  expect  to  achieve  reasonably  successful  analysis  of  uncompli- 
cated fatty  acid  and  fatty  aldehyde  mixtures.  This  is  not  so,  however, 
in  the  case  of  glyceride  analysis  since  the  situation  is  complicated  by  the 
high  molecular  weights  of  the  compounds  and  by  the  complexity  of 
the  molecular  species  found  in  naturally  occurring  glycerides.  Glyceride 
analysis  by  GLC  is  still  in  its  infancy.  The  first  practical  demonstration 
of  glyceride  analysis  (5)  by  GLC  was  shown  one  decade  after  the  intro- 
duction of  GLC  analysis.  Although  glyceride  GLC  analysis  is  beyond 
the  scope  of  this  book,  we  can  appropriately  include  a  brief  account  of 
the  difficulties  and  approaches  to  success  in  this  area. 

So  far  we  have  considered  GLC  analyses  that  can  be  readily  ac- 
complished using  isothermal  conditions.  In  general,  the  efficiency 
achieved  under  isothermal  operation  is  not  conducive  to  satisfactory 
analysis  of  glycerides.  The  efficiency  of  a  GLC  column  isothermally 
operated  is  given  by  the  number  of  "theoretical  plates"  (a  term  allied 
to  distillation  theory),  for  which  the  expression  is:  N  =  I6(tr/w)2. 
N  is  the  number  of  theoretical  plates,  tr  is  the  residence  time  of  the 
component  in  the  column,  and  w  is  the  width  of  the  peak.  An  efficient 
column  gives  peaks  of  narrow  width.  If  band  diffusion  is  large  and 
broad  peaks  result,  programmed  temperature  is  called  for.  A  column 
operated  isothermally  under  linear  temperature  programming  conditions 
may  often  give  5  or  6  times  the  apparent  number  of  theoretical  plates. 
This  improvement  is  very  important  to  the  chromatographer  of 
glycerides.  The  best  conditions  determined  so  far  for  glycerides  are 
low  liquid  phase  concentrations;  narrow,  short  columns;  and  tempera- 
ture programming  from  200  to  350°C.  At  temperatures  above  350° C 
thermal  cracking  may  result. 

Triglycerides  can  be  chromatographed  directly,  but  in  the  interests 
of  increased  stability  and  volatility,  mono-  and  diglycerides  should  be 
converted  to  suitable  derivatives  such  as  acyl  esters  or  silyl  ethers. 
Isopropylidene  and  benzylidene  derivatives  have  also  been  used. 

The  usual  columns  employed  for  glyceride  analysis  (triglycerides 
or  derivatized  mono-  and  diglycerides)  are  the  silicone  polymers  (Se- 
30),  polysiloxane  polymers  (JXR,  Applied  Science  Laboratories,  Inc.), 
and  fluoroalkyl  silicone  gums  (Dow-Corning).  The  percentage  of 
liquid  phase  employed  does  not  usually  exceed  3  percent  and  is  fre- 
quently below  1  percent. 


82  GAS-LIQUID  CHROMATOGRAPHY 

Because  column  "bleed"  at  high  temperatures  is  considerable,  instru- 
ments with  "dual  column  operation"  are  preferable.  Dual  column  opera- 
tion usually  compensates  for  baseline  drift  due  to  bleed  by  splitting  the 
effluent  stream  and  using  dual  compensating  flame  ionization  detectors. 

Unless  some  preliminary  separations  are  performed,  chromatography 
of  glycerides  yields  peaks  of  mixed  molecular  species,  generally  classi- 
fied according  to  the  total  number  of  carbon  atoms  they  contain.  If 
detailed  molecular  species  analyses  are  sought,  then  mixtures  must  be 
subjected  to  preliminary  separations  by  column  and  thin-layer  chroma- 
tography. Mixtures  must  be  separated  not  only  into  mono-,  di-,  and 
triglyceride  fractions,  but  also  into  fractions  based  on  the  degree  of 
unsaturation.  This  may  be  achieved  by  argentation  chromatography. 

Acetate  derivatives.  The  determination  of  phosphoglyceride  struc- 
ture has  been  discussed  previously  (Chapter  4,  p.  55).  This  analytical 
procedure  included  the  preparation  and  analysis  of  the  diglyceride 
acetates  derived  from  the  phospholipids.  These  acetate  derivatives  are 
readily  prepared  at  room  temperature  by  dissolving  about  5mg  glyceride 
in  1ml  dry  pyridine  (distilled  over  barium  oxide)  in  a  tight  screw 
cap  vial  and  adding  0.5ml  acetic  anhydride.  The  mixture  is  allowed  to 
stand  overnight,  after  which  excess  reagent  is  removed  by  evaporation 
under  vacuum.  Alternatively,  diglyceride  analysis  may  use  silyl  ether 
derivatives  prepared  with  one  of  the  available  silylation  reagents. 
Whichever  derivative  is  chosen,  the  analysis  can  be  carried  out  (iso- 
thermally  or  by  programmed  temperature  operation)  on  column  pack- 
ings previously  mentioned  (for  example,  1  to  3  percent  SE-30, 
3   ft.  X  i/8-inch  column)   at  temperatures  between   180  and  300°C. 

The  preceding  comments  merely  cover  some  of  the  major  points  re- 
garding GLC  of  glycerides.  The  reader  interested  in  pursuing  this  field 
is  referred  to  the  Suggested  Further  Readings. 

Other  Lipids  and  Components  Derived  from  Lipids 

Other  nonpolar  lipids,  such  as  the  glyceryl  ethers,  can  also  be  sub- 
jected to  GLC  analysis  usually  as  trifluoracetate  or  silyl  ether 
derivatives. 

Glycerol,  amino  alcohols  such  as  sphingosine  and  related  compounds, 
and  carbohydrates  derived  from  lipids  can  also  be  analyzed  by  GLC 
generally  in  derivatized  form.  References  to  the  GLC  analysis  of  non- 
lipid moieties  of  lipid  molecules  are  given  in  Suggested  Further 
Readings. 

PYROLYSIS-GLC 

The  GLC  analysis  of  the  products  of  pyrolysis  has  been  used  ad- 
vantageously in  the  study  of  hydrocarbons.  Recently,  several  investiga- 
tors have  started  to  explore  the  use  of  pyrolysis-GLC  as  an  approach 
to   the   determination   of   phospholipid   structure.   Kuksis,   Marai,   and 


GAS-LIQUID  CHROMATOGRAPHY  83 

Gornall  (6)  noted  that  serum  lecithins  undergo  pyrolysis  in  a  flash 
evaporator  (at  280  to  300°C)  attached  to  a  GLC  apparatus.  They 
tentatively  identified  the  chromatographed  peaks  as  the  propenediol 
diesters,  which  under  most  conditions  have  the  same  retention  times  as 
the  corresponding  diglycerides.  Perkins  and  Johnston  (7)  subjected 
a  number  of  phosphoglycerides  to  pyrolysis-GLC  and  made  a  mass 
spectral  study  of  the  products.  All  the  phosphoglycerides  were  found  to 
cleave  at  the  phosphate  ester  bond,  and  the  GLC  peaks  obtained  had 
retention  times  the  same  as  those  of  the  corresponding  diglycerides. 
The  mass  spectral  data  confirmed  the  elimination  of  the  phosphate  ester 
group  and  showed  that  the  products  obtained  were  the  dehydrated  di- 
glycerides, that  is,  the  diacylesters  of  propenediol.  A  different  approach 
has  been  used  by  Horning,  Casparrini,  and  Horning  (8),  who  subjected 
phospholipids  to  silylation  with  bis-trimethylsilylacetamide  (BSA)  and 
trimethylchlorosilane  (TMCS)  and  injected  the  derivatives  into  the 
gas  chromatograph.  They  identified  the  products  of  the  phosphoglycer- 
ides as  the  corresponding  dehydrated  trimethylsilyl  derivatives  of  the 
diglycerides.  These  investigators  also  found  that  the  phosphate  ester 
group  was  eliminated  from  silylated  sphingomyelin  on  thermal  cracking, 
and  that  trimethylsilyl  derivatives  of  ceramides  amenable  to  GLC  and 
GLC-mass  spectrometry  studies  could  be  obtained. 

Further  studies  are  needed  to  explore  the  possibility  of  adapting 
these  findings  to  the  analysis  of  polar  lipids  by  GLC. 

REFERENCES 

1.  Johnston,  P.  V.,  and  B.  I.  Roots  (1964)  /.  Lipid  Res.  5 :  477. 

2.  Wood,  R.,  E.  L.  Bever,  and  F.  Snyder  (1966)  Lipids  1 :  399. 

3.  Farquhar,  J.  W.  (1962)  /.  Lipid  Res.  3 :  21. 

4.  Gray,  G.  M.  (1967)  in  Lipid  Chromatographic  Analysis,  ed.  G.  V.  Marinetti ; 
vol.  1,  p.  401.  Marcel  Dekker,  Inc.,  New  York. 

5.  Kuksis,  A.,  and  M.  J.  McCarthy  (1962)   Can.  J.  Biochem.  Physiol.  40:  679. 

6.  Kuksis,  A.,  L.  Marai,  and  D.  A.  Gornall  (1967)  /.  Lipid  Res.  8:  352. 

7.  Perkins,  E.  G.,  and  P.  V.  Johnston  (1969)  Lipids  4:  301. 

8.  Horning,  M.  G.,  G.  Casparrini,  and  E.  C.  Horning  (1969)  /.  Chromatogr.  Sci. 
7 :  267. 


VI.  Procedures  for  the  Determination 

of  Specific  Elements,  Functional  Groups, 
and  Lipid  Classes 


In  this  chapter  the  determination  of  specific  elements,  functional 
groups,  and  individual  lipids  are  described  in  detail.  These  pro- 
cedures allow  the  investigator  to  determine  the  amount  of  specific  lipids 
(cholesterol,  gangliosides)  and  specific  lipid  classes  (total  phospholipid, 
glycolipid,  etc.)  in  lipid  mixtures.  They  can  also  be  applied  to  the 
quantitative  analysis  of  lipids  separated  by  column,  thin-layer,  and 
paper  chromatographic  procedures. 

In  keeping  with  all  former  accounts  in  this  text,  this  chapter  is  not 
a  compilation  of  all  available  methods  for  these  determinations.  The 
methods  presented  are  selected  on  the  basis  that  the  author  or  a  col- 
league has  had  considerable  experience  with  them  and  has  found  them 
to  be  satisfactory. 

DETERMINATION  OF  ORGANIC  PHOSPHORUS 

The  method  described  is  a  modification  of  Bartlett's  (1)  procedure. 
Reagents:      concentrated  sulfuric  acid 

70  percent  perchloric  acid 

ammonium  molybdate,  0.26  percent  aqueous  solution 

disodium  hydrogen  phosphate  (for  standard  curve) 

Fiske-Subbarow  Reagent 
Equipment:  pyrex  test  tubes 

water  bath  and  sand  bath 

spectrophotometer  reading  at  800  to  820m/x 

Fiske-Subbarow  Reagent:  To  80ml  of  15  percent  sodium  bisulfite 
add  0.2g  purified  l-amino-2-naphthol-4-sulfonic  acid  and  0.4g  an- 
hydrous sodium  sulfite,  while  stirring.  Filter  and  store  in  brown  bottle. 
Make  fresh  solution  each  week. 

Procedure:  To  a  test  tube  containing  0.5  to  20.0/xg  of  lipid  phos- 
phorus add  0.5ml  concentrated  sulfuric  acid.  Digest  the  lipid  by  placing 
the  tube  in  a  sand  bath  at  250° C  for  3  hours.  Complete  the  digestion  by 
adding  3  drops  (approx.  0.15ml)  70  percent  perchloric  acid  and  heat 
at  250°C  for  a  further  30  minutes.  Mix  the  contents  of  the  tube  oc- 
casionally. After  digestion  is  complete  (solutions  should  be  clear), 
cool  the  tubes  and  add  9.1ml  of  0.26  percent  ammonium  molybdate  and 
0.4ml   Fiske-Subbarow   reagent.   After  the   solution   has   been   mixed 

84 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  85 

thoroughly  (preferably  with  a  Vortex  mixer),  heat  the  tubes  in  a  bath 
of  boiling  water  for  10  minutes,  then  cool.  Read  the  blue  color  developed 
at  800  or  820mjn;  the  latter  is  preferred  if  available.  Zero  the  instrument 
with  a  prepared  reagent  blank. 

Prepare  a  standard  curve  0.2  to  30/ig  of  phosphorus,  using  potassium 
dihydrogen  phosphate. 

Application  of  method  to  spots  on  TLC  plates.  This  procedure  can 
also  be  applied  directly  to  adsorbent  plus  lipid  removed  from  a  TLC 
plate.  The  digestion  will  go  to  completion  in  the  presence  of  adsorbent 
(preferably  Silica  Gel  H)  and  remaining  traces  of  solvents  such  as  n- 
butanol.  The  solution  will  clear  when  digestion  with  perchloric  acid 
is  complete. 

Before  deciding  on  a  loading  for  a  TLC  plate,  the  total  lipid  phos- 
phorus should  be  determined  and  a  trial  TLC  plate  should  be  run  to 
determine  how  many  phospholipids  are  present.  The  relative  concen- 
trations of  the  phospholipids  may  be  judged  by  charring.  A  suitable 
loading  can  then  be  selected.  A  load  which  gives  about  0.5  to  15.0/xg 
lipid  phosphorus  per  spot  is  ideal ;  however,  the  method  will  detect 
amounts  from  as  little  as  0.1  to  0.2/xg  up  to  25.0/xg. 

After  developing  the  plate,  spot  an  amount  of  total  lipid  containing 
5  to  10/Ag  phosphorus  on  a  part  of  the  plate  untouched  by  solvent,  and 
remove  the  spot  to  make  a  total  lipid  phosphorus  check.  Also  remove  a 
clear  area  of  equal  size  to  the  blank.  Spots  are  best  visualized  by  ex- 
posing the  plate  to  iodine  vapor  in  a  closed  jar,  after  the  solvents  have 
evaporated.  Spots  of  interest  can  be  outlined  with  a  needle,  removed 
either  by  aspiration  or  scraping,  and  placed  in  test  tubes.  Remove  clear 
areas  of  equal  size  and  place  in  test  tubes  to  use  as  sample  blanks.  The 
procedure  is  carried  out  as  described  above,  except  that  after  color 
development  the  tubes  must  be  centrifuged  at  about  600  X  g  and  the 
solutions  decanted  into  clean  tubes  to  free  them  of  the  silica  gel.  The 
color  is  read  in  the  usual  way,  subtracting  adsorbent  blank  optical 
density  from  the  sample  before  reading  off  the  standard  curve.  Blank 
readings  above  optical  density  of  0.03  to  0.04  are  undesirable  and  indi- 
cate contamination  of  either  the  adsorbent  or  reagents  with  a  phos- 
phorus containing  compound.  To  avoid  contamination,  wash  all  glass- 
ware in  chromic  acid  and  rinse  thoroughly  in  deionized  water.  It  may 
prove  necessary  to  wash  the  Silica  Gel  H  as  described  on  pp.  42-43. 

DETERMINATION  OF  CHOLESTEROL 

By  Spectrophotometry 

The  procedure  is  that  of  Zak,  Luz,  and  Fisher  (2). 

Reagents:  ferric  chloride  stock  solution  —  700g  ferric  chloride 
(FeCl3  *  6H20)  in  glacial  acetic  acid  in  a  100ml 
volumetric  flask;  dilute  to  mark  and  mix  well. 


86  DETERMINATION  OF  SPECIFIC  ELEMENTS 

ferric  chloride  working  solution  —  stock  solution  diluted 
1:10  v/v  with  glacial  acetic  acid. 

cholesterol  stock  standard  —  lOOmg  pure  cholesterol 
in  glacial  acetic  acid  in  100ml  volumetric  flask;  dilute 
to  mark. 

digitonin  solution  —  lg  digitonin  in  50ml  ethanol,  di- 
luted with  distilled  water  to  100ml  in  volumetric  flask, 
acetone-alcohol  1 : 1  v/v 
acetone 

concentrated  sulfuric  acid 
Equipment:  spectrophotometer  and  1cm  cuvets 
conical  centrifuge  tubes,  15ml 
test  tubes,  15ml 

Procedure:  Prepare  a  standard  curve  by  diluting  1ml  of  the  ferric 
chloride  stock  solution  and  1ml  of  the  cholesterol  stock  standard  with 
glacial  acetic  acid  to  10ml  in  a  volumetric  flask.  Pipet  1.0,  2.0,  and  3.0ml 
of  this  standard  into  test  tubes,  diluting  the  1.0  and  2.0ml  fractions  to 
3.0ml  with  the  ferric  chloride  working  reagent.  Prepare  one  blank  tube 
of  3ml  ferric  chloride  working  reagent.  Carefully  layer  2ml  of  concen- 
trated sulfuric  acid  over  each  solution,  then  mix  well.  When  the  solu- 
tions have  cooled  to  room  temperature,  measure  absorbancies  at  560m//. 
against  the  blank. 

To  determine  serum  cholesterol  levels,  pipet  0.2ml  of  serum  into  a 
10ml  volumetric  flask  containing  approximately  5ml  of  acetone-alcohol 
solution.  Dilute  to  mark  with  acetone-alcohol  and  shake  vigorously 
to  extract  the  cholesterol.  Filter  mixture  through  Whatman  Xo. 
41  filter  paper,  keeping  a  watch  glass  over  the  funnel.  Pipet  5ml  of 
filtrate  into  conical  centrifuge  tube  for  determination  of  free  cholesterol 
and  2.5ml  of  filtrate  into  a  test  tube  for  total  cholesterol  determination. 
Evaporate  contents  of  test  tube  to  dryness  and  contents  of  centrifuge 
tube  to  0.5  to  1.0ml. 

To  test  tube  add  3ml  of  ferric  chloride  working  solution  to  dissolve 
residue.  Layer  in  2ml  of  concentrated  sulfuric  acid  and  mix  solution 
well.  Wait  15  minutes  for  color  development,  then  measure  the  absor- 
bance  against  a  blank  at  560m/,t. 

To  the  centrifuge  tube  add  0.5ml  of  digitonin  solution,  wait  15 
minutes,  then  centrifuge  at  600  to  700  X  g  for  10  minutes.  Decant  the 
supernatant.  Add  4ml  of  acetone  to  disperse  the  precipitate,  tapping  the 
tube  until  the  precipitate  is  homogenous,  then  centrifuge  again  for  10 
minutes.  Decant  the  wash  solution.  Invert  tube  on  absorbent  paper 
to  drain.  Add  ferric  chloride  (3ml)  and  sulfuric  acid  (2ml)  as  de- 
scribed above,  wait  15  minutes,  and  read  absorbance  at  560m/x. 

The  method  has  been  shown  to  give  stoichiometric  final  color  re- 
action with  amounts  of  cholesterol  up  to  lOOOmg  per  100ml.  The  range 
of  analysis  can  be  extended  by  appropriate  dilution  of  the   original 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  87 

sample.  While  the  method  has  been  described  for  analysis  of  serum 
cholesterol  levels,  it  can,  of  course,  be  adapted  to  the  analysis  of  the 
cholesterol  content  of  any  sample  of  total  lipid.  All  that  is  required  is 
dilution  of  the  original  sample  to  give  a  cholesterol  content  within  the 
range  0  to  lOOOmg  per  100ml. 

By  Gas-Liquid  Chromatography 

The  method  described  utilizes  cholestane  as  an  internal  GLC  stan- 
dard (3). 

Reagents:      cholesterol 

cholestane 

95  percent  ethanol 

diethyl  ether 

hexane 

chloroform 

potassium  hydroxide 
Equipment:  18mm  X  150mm  test  tubes 

serum  caps  to  fit  test  tubes 

centrifuge  tubes,  15ml,  screw  cap 

20ml  vials 

5  and  10ml  pipets 

graduated  pipets 

gas  chromatograph  equipped  with  glass  column,  glass 
inlet  system,  and  flame  ionization  detector 

Procedure:  Prepare  the  following  standard  solutions: 

1.  Ratio  Solution.  Weigh  lOOmg  cholesterol  and  lOOmg  cholestane. 
Dilute  to  100ml  with  chloroform.  Mix  and  store  in  refrigerator.  These 
compounds  are  stable  for  several  weeks. 

2.  Potassium  hydroxide.  Weigh  out  3.3g  potassium  hydroxide  and 
dilute  to  10ml  with  distilled  water.  Prepare  fresh  daily. 

3.  Internal  standard.  Weigh  lOOmg  cholestane  and  dilute  with 
chloroform  to  100ml.  Subdivide  solution  into  10ml  portions.  Store  in  re- 
frigerator in  tightly  stoppered  containers. 

Operating  conditions  for  gas  chromatograph : 

Column.    18-inch  glass  packed  with  3.8  percent  SE-30  on  Diatoport 

S.    (Note:   This  column  is  recommended  in  the  original 

procedure,  but  other  columns  are  also  suitable,   such  as 

1  to  2  foot  columns  packed  with  1  to  3  percent  SE-30  on 

various  supports.) 
Column  temperature.  200  to  230° C,  depending  on  column  packing, 

etc.  Other  settings  will  vary  from  instrument  to  instrument 

and  must  be  determined  individually. 
Detector.  A  flame  ionization  detector  is  recommended  because  its 

response   is   usually  linear  over   a   wide   range.    Use   of 


88  DETERMINATION  OF  SPECIFIC  ELEMENTS 

another  kind  of  detector  will  probably  necessitate  prepar- 
ing a  correction  curve  for  the  specific  detector. 

Determination  of  total  serum  cholesterol 

1.  Measure  0.2ml  serum  into  test  tube. 

2.  Add  4.7ml  95  percent  ethanol. 

3.  Add  0.3ml  33  percent  aqueous  potassium  hydroxide  solution. 

4.  Stopper  tube  and  mix  well. 

5.  Place  in  water  bath  at  55 °C  for  15  minutes. 

6.  Remove  tube  and  cool. 

7.  Add  5ml  distilled  water. 

8.  Add  10ml  hexane  and  allow  to  stand  till  two  phases  appear. 

9.  Remove  a  5ml  aliquot  of  the  upper  (hexane)  phase  and  trans- 
fer to  a  3-dram  screw-cap  vial. 

10.  Use  a  stream  of  nitrogen  and  a  water  bath  to  evaporate  the 
hexane  aliquot  to  dryness. 

11.  Add  0.2ml  of  the  internal  standard  solution  to  the  dry  residue. 

12.  Cap  vial  and  mix  well;  inject  about  3/xl  of  mixture  into  gas 
chromatograph  (exact  size  of  sample  is  not  important  when  internal 
standardization  is  used  —  see  p.  74) . 

Analysis  on  GLC  will  take  7  to  15  minutes,  depending  on  the 
column. 

Determination  of  free  cholesterol 

1.  Measure  0.2ml  serum  into  15ml  screw  cap  centrifuge  tube. 

2.  Add  9ml  95  percent  ethanol. 

3.  Add  3ml  diethyl  ether. 

4.  Shake  and  allow  to  stand  for  5  minutes. 

5.  Centrifuge  (600  to  700  X  g). 

6.  Pour  supernatant  into  20ml  vial. 

7.  Evaporate  residue  to  dryness  as  previously  described. 

8.  Add  0.2ml  of  internal  standard  solution. 

9.  Inject  about  3/xl  into  gas  chromatograph;  GLC  analysis  time  will 
be  the  same  as  for  total  cholesterol. 

Interpretation  of  chromatograms.  First  obtain  a  chromatogram  by 
injecting  Zjx\  of  the  cholestane-cholesterol  standard  solution  and  calcu- 
late the  ratio  either  by  using  peak  height  or  peak  area,  thus: 

cholestane  peak  height  (area)  _  „ 
cholesterol  peak  height  (area) 

To  calculate  total  cholesterol  in  the  sample,  measure  peak  heights 
(or  areas),  multiply  cholesterol  peak  height  by  the  ratio,  and  divide  by 
the  cholestane  peak  height.  Since  in  the  original  procedure  5ml  of 
hexane  was  withdrawn  from  the  mixture,  the  value  must  be  multiplied 
by  2.  Thus,  the  calculation  for  total  cholesterol  in  a  sample  becomes: 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  89 

cholesterol  peak  height  x  R  x  2  =  ^  cholesterol  (mg/lQOml) 
cholestane  peak  height 

To  determine  free  cholesterol,  follow  the  same  calculation  procedure 
as  for  total  cholesterol.  Since  the  whole  sample  is  used  in  the  analysis 
for  free  cholesterol,  the  calculation  will  be: 

cholesterol  peak  height  x  R  =  frge  cho,estero,  (mg/100ml) 
cholestane   peak  height 

The  ratio  R  should  be  determined  from  standards  daily,  as  slight 
changes  in  column  conditions  may  affect  it.  It  must  also  be  remembered 
that  if  changes  in  sensitivity  factors  have  been  made  during  the  run, 
these  must  be  taken  into  account  in  the  calculations. 

THE  DETERMINATION  OF  GLYCOLIPID  SUGARS 

By  Using  Anthrone 

The  method  described  is  that  of  Radin  et  al.  (4,  5). 

Reagents:  anthrone  stock  solution  —  2  percent  anthrone  (re- 
crystallized)  in  sulfuric  acid;  age  4  hours  at  room 
temperature  (stable  for  2  weeks  in  refrigerator), 
anthrone  working  solution  —  stock  solution  diluted  14 
times  with  sulfuric  acid- water  9:5  v/v;  make  up  just 
before  use. 

galactose  standards  —  dry  galactose  at  100°C  over 
phosphorus  pentoxide  and  dissolve  in  water  to  give  a 
concentration  of  20mg  per  ml ;  store  this  solution  in 
polyethylene  bottle  in  refrigerator;  prepare  standards 
by  evaporating  appropriate  volumes  to  dryness  in 
colorimeter  tubes. 

hydrolytic  solvent  —  ethanol-concentrated  hydrochloric 
acid  74:63  v/v. 

Equipment:  colorimeter 

screw-cap  tubes,  10  to  15ml 
constant  temperature  bath  set  at  58° C 

Procedure : 

1.  Evaporate  a  sample  solution  containing  0.2  to  1.7mg  cerebroside 
(or  other  galactolipid)  to  dryness  in  a  screw-cap  tube. 

2.  Add  3ml  hydrolytic  solvent  and  place  in  water  bath  at  58° C 
for  3  hours. 

3.  Add  5ml  toluene  (as  an  anti-splashing  agent)  and  evaporate  to 
dryness  under  vacuum. 

4.  Add  10ml  water  and  2ml  chloroform,  cap  the  tube,  and  centrifuge 
at  600  to  700  X  g. 


90  DETERMINATION  OF  SPECIFIC  ELEMENTS 

5.  Transfer  duplicate  2ml  aliquots  to  colorimeter  tubes,  add  1ml 
toluene,  and  evaporate  to  dryness. 

6.  Add  5ml  of  the  anthrone  working  solution  and  mix  well. 

7.  Develop  the  color  (green)  by  heating  for  6  minutes  at  100°C  or 
16  minutes  at  90°C. 

8.  Read  the  color  at  625m^  against  a  blank  anthrone  solution. 

9.  Prepare  a  standard  curve  from  evaporated  aliquots  of  standard 
galactose  solution  (as  described  above)  treated  as  in  steps  5  through  8. 

If  the  complete  fatty  acid  composition  of  the  glycolipids  is  known, 
the  average  molecular  weight  can  be  determined  and  the  galatose  con- 
tent can  be  used  to  calculate  moles  of  glycolipid.  If  this  information  is 
not  available,  conversion  of  sugar  content  to  quantity  of  glycolipid  can 
be  calculated  on  the  basis  of  expected  average  sugar  content  for  the 
particular  glycolipid. 

By  Gas-Liquid  Chromatography 

Carbohydrates  liberated  from  glycolipids  can  readily  be  analyzed  by 
GLC.  Preparation  of  the  trimethylsilyl  derivatives  of  sugars  is  quite 
easily  and  rapidly  achieved  with  the  commonly  used  silylating  agents 
such  as  hexamethyldisilazane  and  trimethylchlorosilane  in  dry  pyridine. 
Quantitative  yields  of  the  polytrimethylsilyl  derivatives  can  usually  be 
obtained  within  a  few  minutes  at  room  temperature.  The  derivatives 
can  be  separated  on  short  columns  packed  with  nonpolar  phases  such  as 
SE-30  (2  to  3  percent)  at  temperatures  about  140°C  to  160°C.  Mix- 
tures of  sugars  can  be  quantitatively  determined  by  using  the  internal 
normalization  procedure,  and  a  single  sugar  can  be  analyzed  by  using 
an  appropriate  internal  standard.  However,  while  the  preparation  of 
derivatives  and  their  GLC  analysis  are  relatively  simple  matters,  the 
quantitative  liberation  of  the  sugars  from  the  glycolipids  presents  diffi- 
culties. Under  some  hydrolytic  conditions  (such  as  aqueous  acid), 
some  sugars  may  degrade.  These  difficulties  have  been  discussed  in  de- 
tail recently  by  Sweeley  and  Vance  (6).  The  best  method  available  at 
present  appears  to  be  anhydrous  methanolysis :  the  products  liberated 
from  most  glycolipids  under  these  conditions  are  equilibrated  anomeric 
mixtures  of  relatively  stable  methyl  glycosides  that  can  be  readily  con- 
verted to  derivatives  for  GLC  analysis. 

DETERMINATION  OF  N-ACETYLNEURAMINIC  ACID 
(IN  GANGLIOSIDES) 

By  Using  Resorcinol 

The  method  described  is  from  Svennerholm  (7,8). 

Reagents:      hydrochloric  acid,  density  1.19  (at  least  36.4  percent), 

Fe+++  less  than  0.0001  percent. 

0.1M  solution  of  copper  sulfate. 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  91 

resorcinol  stock  solution  —  2g  resorcinol  in  100ml  de- 
ionized  water  (stable  for  months  in  refrigerator), 
resorcinol  working  solution  —  10ml  stock  solution 
added  to  80ml  concentrated  hydrochloric  acid  that  con- 
tains 0.25ml  of  the  0.1M  copper  sulfate  solution  (stable 
one  week  in  refrigerator). 

blank  —  same  as  working  solution,  without  resorcinol. 
Equipment:  spectrophotometer 
centrifuge 
water  bath 
test  tubes,  10  to  15ml 

Procedure:  Three  2ml  samples  containing  5  to  30/xg  of  N-acetyl- 
neuraminic  acid  are  pipetted  into  test  tubes.  Two  of  the  samples  are  the 
unknowns  in  duplicate,  and  the  third  is  the  blank.  To  the  two  unknowns 
add  2ml  of  the  working  resorcinol  reagent,  and  to  the  third  tube  add 
2ml  of  blank  sample  reagent.  Prepare  a  standard  curve  using  N-acetyl- 
neuraminic  acid  in  the  range  0  to  30^.  Prepare  as  for  samples  using 
the  0/xg  tube  as  the  blank.  Heat  the  tubes  for  15  minutes  in  a  bath  of 
boiling  water.  Cool  and  place  in  ice  bath.  Extract  the  color  with  4ml 
of  a  solution  of  n-butylacetate-n-butyl  alcohol,  85:15  v/v.  Shake  well 
and  allow  to  settle  in  the  cold,  or  transfer  to  centrifuge  tube  and  centri- 
fuge at  300  to  400  X  g  in  a  cold  room  or  a  refrigerated  centrifuge.  Com- 
plete this  part  of  the  procedure  within  1  hour  of  the  heating  step.  Read 
absorbance  at  580m//.  in  1cm  cells.  Subtract  absorbance  of  blank  sample 
from  test  samples  and  read  /xg  of  N-acetylneuraminic  acid  from  curve. 
If  greater  sensitivity  is  desired  (0.05  to  0.50/xM  of  N-acetylneuraminic 
acid),  the  color  may  be  read  in  50mm  microcells. 

If  this  procedure  is  applied  to  gangliosides  that  have  been  sepa- 
rated into  mono-,  di-,  and  trisialogangliosides  (compounds  containing 
1,  2,  and  3  moles  of  N-acetylneuraminic  acid  per  mole  of  ganglioside), 
then  the  N-acetylneuraminic  acid  content  may  readily  be  used  to  calcu- 
late the  weight  of  individual  gangliosides.  A  typical  preparation  of  total 
gangliosides  extracted  from  mammalian  brain  contains  24  to  27  percent 
N-acetylneuraminic  acid. 

This  procedure  can  be  applied  to  glycolipids  containing  N-glycolyl- 
neuraminic  acid  in  place  of  N-acethylneuraminic  acid;  however,  either 
a  standard  curve  using  the  N-glycolyl  compound  must  be  used  or  the 
absorbancies  must  be  corrected  for  the  30  percent  greater  molar  absor- 
bancy  index  of  the  glycolyl  derivative.  Values  read  from  the  N-acetyl- 
neuraminic acid  standard  curve  would  have  to  be  multiplied  by  0.77. 

By  Gas-Liquid  Chromatography 

The  hydroxyl  groups  on  N-acetylneuraminic  acid  can  readily  be 
silylated  to  yield  a  compound  amenable  to  GLC  analysis.  Thus,  the 


92  DETERMINATION  OF  SPECIFIC  ELEMENTS 

methyl  ester  of  N-acetylneuraminic  acid,  obtained  on  methanolysis  of 
gangliosides,  can  be  converted  to  the  trimethylsilyl  derivative  with 
any  of  the  available  silylating  reagents.  This  product  can  be  subjected 
to  GLC  on  a  column  such  as  2.5  to  3  percent  SE-30.  For  quantitative 
determinations  it  is  necessary  to  use  the  internal  standard  technique 
(p.  87),  as  described  for  the  GLC  determination  of  cholesterol.  The 
choice  of  a  suitable  internal  standard  for  N-acetylneuraminic  acid 
determinations  has  not  been  extensively  studied.  It  appears,  however, 
that  a  sugar  such  as  mannitol  or  a  suitable  amino  sugar  should  prove 
satisfactory. 

THE  DETERMINATION  OF  PLASAAALOGENS 

By  Colorimetry 

The  method  described  is  that  of  Gray  and  Macfarlane  (9).  It  is  a 

two-stage  reaction  procedure  in  which  the  aldehyde  is  first  split  off  the 

plasmalogen  and  then  condensed  with  fuchsin  reagent  to  give  a  color. 

Reagents:      fuchsin  reagent  —  dissolve  lg  rosaniline  hydrochloride 

in  700ml  boiling  water;  filter,  cool;  add  5.0g  sodium 

metabisulfite  and  100ml  of  IN  HC1  and  make  up  to 

1  liter  with  water;  decolorize  for  48  hours  and  store  at 

2°C  in  a  dark,  glass-stoppered,  narrow-necked  bottle. 

sulfite    water  —  0.5    percent    sodium    metabisulfite    in 

0.1N  HC1. 

octan-2-ol  (capryl  alcohol),  low  ketone  grade, 
dimethyl  acetal  of  palmitaldehyde,  prepared  from  com- 
mercially available  palmitaldehyde  bisulfite  (see  p.  80). 
acetic  acid- water  90: 10  v/v. 
Equipment :  spectrophotometer 

15ml  centrifuge  tubes,  glass-stoppered 

Procedure: 

1.  To  determine  total  aldehyde  in  lipid  samples,  prepare  a  stock 
standard  solution  by  dissolving  48mg  palmitaldehyde  dimethyl  acetal 
in  20ml  chloroform ;  prepare  a  working  standard  by  diluting  to  100ftg 
palmitaldehyde  per  ml.  Prepare  a  standard  curve  of  palmitaldehyde  in 
the  range  5  to  SOfig.  Place  suitable  amounts  of  the  working  standard  in 
centrifuge  tubes  and  remove  solvent.  Add  0.5ml  90  percent  acetic  acid 
and  store  at  50°C  for  45  minutes,  prepare  one  blank).  Add  2.0ml 
fuchsin  reagent,  and  after  20  minutes  at  room  temperature  add  2.0ml 
sulfite  water  and  5.0ml  capryl  alcohol.  Shake  vigorously  and  centrifuge 
for  4  minutes.  Read  the  optical  density  of  the  colored  layer  against  the 
blank  at  546mia.  Now  place  an  amount  of  lipid  sample  containing  5  to 
45 fig  of  aldehyde  in  centrifuge  tubes  and  procede  as  with  the  working 
standard. 

2.  Determine  the  phosphorus  content  of  the  samples   (see  p.  84). 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  93 

The  ratio  of  moles  of  phosphorus  to  moles  of  aldehyde  in  a  plasma- 
logen  is  unity.  The  ratio  of  moles  of  aldehyde  in  the  sample  (from  step 
1)  to  the  moles  of  phosphorus  in  the  sample  (from  step  2),  multiplied 
by  100,  gives  the  "Plasmalogen  Value,"  or  the  percent  of  phosphorus 
that  is  present  in  the  sample  as  plasmalogen. 

By  Two-Dimensional  Thin-Layer  Chromatography  (10) 

This  method  is  based  on  the  specific  hydrolysis  of  plasmalogens  to 
the  2-acyl  lysophosphoglyceride  in  the  presence  of  a  mercuric  chloride 
spray  reagent. 

Reagents:      chloroform-methanol-water  60:35:8  v/v/v 

chloroform-methanol- water-acetic  acid  65:43:3:1   v/v 

petroleum  ether  (bp.  40  to  60°  C) -diethyl  ether-acetic 

acid  80:20:1  v/v/v 

aqueous  ammonia  (sp.  gr.  0.880) 

5mM  mercuric  chloride  in  deionized  water 

18N  sulfuric  acid 
Equipment:  20cm  X  20cm  TLC  plates 

TLC  spreader  and  board 

Silica  Gel  H  (E.  Merck  A.-G.,  Darmstadt,  Germany) 

10ml  stoppered  tubes 

oven  at  180°C,  situated  under  extractor  fan 

Procedure:  Prepare  TLC  plates  (about  500m//,  thick)  using  a  Silica 
Gel  H  slurry  prepared  in  ion-free  water.  Before  activating  plates  at 
110°C,  wash  the  adsorbent  by  allowing  chloroform-methanol-water 
(60:35:8)  to  ascend  to  the  top  of  the  plate.  This  removes  material  which 
interferes  with  charring  by  sulfuric  acid.  Silica  gel  washed  in  bulk  may 
be  used,  but  remember  that  separations  will  vary  and  the  variations 
must  be  ascertained  in  advance  for  each  silica  gel  used. 

Duplicate  lipid  samples  (containing  0.2  to  0.5/xg  atom  of  phos- 
phorus) are  plated  as  1cm  bands,  one  2cm  in  from  the  left  and  the 
other  3cm  in  from  the  right-hand  edge.  Develop  the  plate  in  the  freshly 
prepared  chloroform-methanol- water-acetic  acid  65:43:3:1  v/v  to  8cm 
from  the  top  of  the  plate.  After  removing  the  solvent,  redevelop  the 
plate  to  the  top  using  petroleum  ether  (b.p.  40  to  60°C) -diethyl  ether- 
acetic  acid  80:20:1  v/v/v.  This  solvent  mixture  causes  the  lipids,  other 
than  phospholipids  and  glycolipids,  to  migrate  to  a  position  above  the 
first  solvent  front.  The  residual  acetic  acid  left  on  the  plate  after  de- 
velopment is  neutralized  by  supporting  the  chromatogram  above  an 
aqueous  ammonia  solution  (sp.  gr.  0.880)  in  a  sealed  dish  for  5  minutes. 
The  excess  ammonia  is  then  drawn  off  under  vacuum  (0.5mm  mercury 
or  less)  for  30  minutes.  The  left-hand  lipid  track  is  then  sprayed  with 
5mM  mercuric  chloride  while  the  remainder  of  the  chromatogram  is 
screened.  The  plate  is  turned  through  an  angle  of  90°  and  the  second 
solvent    front    is   marked    at    5cm    from   the   top    of    the   plate.    The 


94  DETERMINATION  OF  SPECIFIC  ELEMENTS 

plate  is  then  reactivated  by  evacuating  at  0.5mm  mercury  or  less 
over  dark  blue  self-indicating  silica  gel  for  one  hour.  The  mercuric 
chloride-treated  lipids  are  developed  in  the  second  dimension  with 
chloroform-methanol-water  60:35:8  v/v/v.  The  chromatogram  is  then 
dried,  sprayed  with  18N  sulfuric  acid,  and  charred  at  180°C  for  1 
hour  in  an  oven  situated  beneath  an  extractor  fan  to  ensure  removal 
of  volatilized  sulfuric  acid  and  mercuric  chloride. 

The  charred  areas  are  removed  and  placed  in  10ml  stoppered 
tubes.  Appropriate  blank  areas  (corresponding  to  large,  medium,  and 
small  lipid  spots)  are  also  taken  and  are  added  to  tubes  containing 
evaporated  aliquots  (0.1ml)  of  a  standard  phosphate  solution  equivalent 
to  0.05/xM  of  phosphorus.  Total  phosphorus  is  then  determined  on  all 
samples  and  on  blank  plus  standard  samples.  The  phosphorus  deter- 
mination procedure  used  by  the  originator  of  this  method  was  that  of 
Sloane-Stanley  and  Eldin  (11),  but  other  procedures,  such  as  the  one 
previously  described  (pp.  84-85),  can  be  applied. 

The  total  lipid  phosphorus  content  of  the  original  diacyl  and  mono- 
vinylether-monoacyl  phospholipid  mixture  plus  the  2-acyl  lysophos- 
pholipid  left  after  hydrolysis  with  mercuric  chloride  is  determined. 
These  values  for  percent  phosphorus  can  be  converted  to  amounts  of 
phospholipid  by  multiplying  by  an  appropriate  factor.  The  factors  can 
be  determined  from  the  actual  fatty  acid  composition  of  each  phos- 
pholipid as  determined  by  GLC.  However,  for  many  purposes  the 
factors  given  by  Williams  et  at.  (10)  for  human  serum  phospholipids 
may  be  sufficiently  accurate: 

phosphatidyl  choline  26.2 

phosphatidyl  ethanolamine  25.0 

phosphatidyl  serine  25.7 

lysophosphatidyl  choline  17.5 

lysophosphatidyl  ethanolamine  16.0 

If  the  fatty  acid  composition  of  the  lipids  differs  markedly  from  that 
of  serum  phospholipids  and  would  mean  fairly  large  differences  in 
molecular  weight,  then  factors  for  the  lipids  being  analyzed  must  be 
determined. 

When  the  percentages  of  the  intact  and  the  2-acyl  lysophospho- 
lipids  are  known,  the  percent  plasmalogen  can  be  determined  by  the 
difference  between  the  two. 

DETERMINATION  OF  THE  AMOUNT  OF  TRANS  DOUBLE  BOND 

By  Infrared  Spectrophotometry 

Compounds  with  isolated  trans  double  bonds  exhibit  an  absorption 
band  in  the  infrared  due  to  a  C-H  deformation  about  the  trans  double 
bond.  This  absorption  band  has  its  maximum  at  about  10.3ju  and  can 
be   used   to   determine   quantitatively   the   isolated   trans   double   bond 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  95 

content  of  compounds.  The  procedure  outlined  below  is  the  one  de- 
scribed in  The  American  Oil  Chemists'  Society  Handbook  of  Official 
and  Tentative  Methods,  vol.  1.  The  method  is  not  applicable  to  lipids 
that  contain  large  quantities  of  conjugated  unsaturation,  to  compounds 
that  contain  functional  groups  that  modify  the  intensity  of  the  C-H 
deformation  about  the  trans  bond,  nor  to  compounds  which  give  rise 
to  absorption  bands  near  10.3//,.  For  example,  long-chain  fatty  acids 
with  less  than  15  percent  isolated  trans  isomers  must  be  converted  to 
their  methyl  esters  for  analysis,  because  efficient  correction  for  an 
absorption  band  of  the  carboxyl  group  at  10.6//,  is  not  possible  at  that 
concentration  of  trans. 

Equipment:  any  infrared  spectrophotometer  covering  the  region  9 
to  11//,  (900  to  1150cm-1)  with  a  wavelength  scale  read- 
able to  0.01^  and  fitted  with  a  cell  compartment  for 
holding  0.2  to  2.0cm  cells. 

fixed   thickness   absorption   cells   with    NaCl    or    KBr 
windows  from  0.2  to  2.0cm. 

Reagents:      carbon  disulfide  (dry,  ACS) 

standards    of    elaidic    acid,    methyl    elaidate,    and    tri- 
elaidin 

Procedure:  Weigh  200mg  of  standard;  place  standard  and  sample 
into  a  10ml  volumetric  flask.  Dilute  to  volume  with  CS2  and  mix. 
Transmittance  at  the  trans  absorption  maximum  should  be  20  to  70 
percent;  if  not,  use  a  different  sample  weight  or  cell  thickness. 

Using  matched  absorption  cells,  fill  one  cell  with  carbon  disulfide 
and  the  other  with  sample  or  standard  solution.  Place  the  cells  in  the 
reference  and  sample  beam  holders  in  the  spectrophotometer.  Measure 
the  transmittance  or  absorbance  over  the  range  9  to  1 1/x.  Different  in- 
struments will  require  different  programs  in  order  to  obtain  a  satis- 
factory curve.  Once  a  satisfactory  program  is  obtained,  all  subsequent 
measurements  must  be  made  using  the  identical  program  conditions. 

From  the  charts  obtained  with  the  standards  and  samples,  read  the 
transmittance  at  10.36//,.  Convert  to  absorbance  and  calculate  absorp- 
tivities.  Draw  a  base  line  on  the  charts  from  10.10/x  to  10.65//,  for  acids, 
from  10.20^1  to  10.59/x  for  methyl  esters,  and  from  10.05/x  to  10.67//. 
for  triglycerides  (see  Fig.  14).  Measure  the  distance  from  the  zero 
line  of  the  recorder  chart  to  the  absorption  peak  (distance  ab),  cal- 
culate the  fractional  transmission  (be)  as  the  distance  to  the  absorp- 
tion peak  (ab)  divided  by  the  distance  to  the  base  line  (ac),  convert 
to  absorbance,  and  calculate  the  "background  corrected  absorptivity." 
If  the  chart  paper  is  calibrated  in  absorbance,  subtract  the  absorbance 
at  the  base  line  (c)  from  the  absorbance  at  the  10.3//,  maximum  (b)  to 
get  the  absorbance  of  the  sample. 

Calculate  percent  trans  isomer  as  elaidic  acid,  methyl  elaidate,  or 
trielaidin  as  follows: 


96 


DETERMINATION  OF  SPECIFIC  ELEMENTS 


100 


u 


0  9  10 

wavelength  (microns) 
Infrared  absorption  of  rrans  unsaturation  in  esters. 


(Fig.  14) 


FUNCTIONAL  GROUPS  AND  LIPID  CLASSES  97 

percent  trans  =  absorptivity  (A/bc)  of  sample  X  100 

absorptivity  of  elaidic  acid,  methyl  elaidate,  or  trielaidin 

Where  A  =  absorbance  =  log  100/7 
b  =  internal  cell  length  in  cm 
c  =  concentration  of  sample  in  g/liter 
T  =  percent  transmission 

By  Gas  Chromatography 

Equipment:  Any  gas  chromatograph  equipped  to  use  capillary 
columns.  Stainless  steel  capillary  columns  (length  50m,  i.d.  0.254mm) 
coated  with  an  8  percent  w/v  solution  of  EGSS-X  (Applied  Science 
Laboratories,  Inc.)  in  methylene  chloride.  A  polar  wetting  agent  such 
as  Alkaterge  T  (an  amine  surfactant)  is  added  to  the  packing  solution 
to  give  a  concentration  of  0.2  percent.  Suitable  prepacked  columns  also 
can  be  purchased  from  Perkin  Elmer  Company. 

Conditions  for  analysis:  These  are  the  conditions  used  by  Lavoue 
and  Bezard  (13),  who  employed  a  Barber  Colman  Model  10  chro- 
matograph equipped  with  a  Sr90  ionization  detector.  Suitable  operating 
conditions  for  other  instruments  and  detectors  must  be  determined 
by  individual  investigators. 

Column  temperature:  186°  to  187°C 

Flow  rate  at  column  outlet:  0.2  to  0.3  ml/min. 

Injector  temperature:  300° C 

Carrier  gas  pressure  at  inlet  to  injector:  8  to  10  psi 

"Split"  ratio:  about   1:100   (The  injected  sample  is  split  into  two 

streams  so  that  only  a  portion  of  the  sample  enters  the 

column.) 
Amount  of  solution  injected:  about  2jA 
Rate  of  back  flush  (scavenger)  :  60  ml/min. 

The  authors  found  that  in  addition  they  had  to  employ  a  shunt 
(flow  rate  8  ml/min.)  to  reduce  the  dead  volume  between  the  column 
outlet  and  the  inlet  to  the  detector.  This  seems  to  be  necessary  when 
using  capillary  columns  with  an  argon  ionization  detector  that  has  a 
large  internal  volume  relative  to  the  volume  eluted  at  the  column  outlet. 
This  problem  is  not  encountered  with  a  flame  ionization  detector. 

The  detector  temperature  was  220° C  and  the  carrier  gas  was  argon. 

Using  this  set  of  conditions,  the  authors  were  able  to  determine 
efficiently  the  composition  of  mixtures  of  methyl  esters  of  oleic,  elaidic, 
9-trans-\2-trans,  and  9-cis-l2-cis  linoleic  acids. 

The  one  disadvantage  of  this  procedure  is  the  use  of  the  polar 
column  packing,  which  has  under  these  conditions  a  very  short  life 
(5  to  10  days).  Separation  of  many  cis  and  trans  isomers  can  be 
achieved  using  a  nonpolar  column  packing  such  as  an  Apiezon  grease. 
If  suitable  conditions  for  separation  can  be  found  using  a  nonpolar 
column  then  it  is  clearly  wiser  to  use  nonpolar  packings. 


98  DETERMINATION  OF  SPECIFIC  ELEMENTS 

REFERENCES 

1.  Bartlett,  G.  R.  (1959)  /.  Biol.  Chem.  234:  466. 

2.  Zak,  B.,  D.  A.  Luz,  and  M.  Fisher  (1957)  Am.  J.  of  Med.-Technol.  Sept.-Oct., 
p.  283. 

3.  F  &  M  Scientific  (Div.  of  Hewlett-Packard)  Bull.  No.  116,  "A  rapid  method 
for  serum  cholesterol  analysis  by  gas  chromatography." 

4.  Radin,  N.S.,  J.  R.  Brown,  and  F.  B.  Lavin   (1956)  /.  Biol.  Chem.  219:  972. 

5.  Radin,    N.    S.    (1958)    in   Methods   of  Biochemical   Analysis,   vol.   6,   p.    162. 
Interscience  Publishers,  New  York. 

6.  Sweeley,  C.  C,  and  D.  E.  Vance  (1967)  in  Lipid  Chromatographic  Analysis, 
ed.  G.  V.  Marinetti;  vol.  1,  p.  465.  Marcel  Dekker,  Inc.,  New  York. 

7.  Svennerholm,  L.  (1957)  Biochim.  Biophys.  Acta  24:  604. 

8.  (1963)  /.  Nenrochem.  10:  613. 

9.  Gray,  G.  M.,  and  M.  G.  Macfarlane  (1958)  Bio  chem.  J.  70 :  409. 

10.  Owens,  K.  (1966)  Biochem.  J.  100:  354. 

11.  Sloane-Stanley,  G.  H.,  and  A.  K.  Eldin  (1962)  Biochem.  J.  85:  40. 

12.  Williams,  J.  H.,  M.  Kuchmak,  and  R.  F.  Witter  (1966)  Lipids  1:  89. 

13.  Lavoue,  G.,  and  J.  Bezard  (1969)  /.  Chromatogr.  Sci.  7 :  375. 


Suggested  Further  Readings 

Classification  of  Lipids 

Deuel,  H.  J.,  Jr.  (1951)  The  Lipids;  vol.  1:  chemistry.  Interscience 
Publishers,  New  York. 

Lipid  Structure 

Chapman,  D.  (1965)  The  Structure  of  Lipids  by  Spectroscopic  and 
X-ray  Techniques.  John  Wiley  &  Sons,  New  York. 

Chemistry  of  Fatty  Acids 

Markley,  K.  S.  (1954,  1968,  1969)  Fatty  Acids;  parts  1,  2,  and  3. 
Interscience  Publishers,  New  York. 

Lipid  Nomenclature 

"The  Nomenclature  of  Lipids."  A  document  for  discussion  sponsored 
by  the  IUPACIUB  Commission  of  Biochemical  Nomenclature  (1967) 
J.  Lipid  Res.  8:  523. 

Phosphonolipids  (a  review) 

Kittredge,  J.  S.,  and  E.  Roberts  (1969)  "A  Carbon-Phosphorus  Bond 
in  Nature."  Science  164:  37. 

Gangliosides  (a  review) 

Leedeen,  R.   (1966)   "The  Chemistry  of  Gangliosides:   a  Review."  /. 

Am.  Oil  Chem.  Soc.  43:  67. 

Prostaglandins  (a  review) 

Samuelson,  B.  (1968)  in  Regulatory  Functions  of  Biological  Mem- 
branes, ed.  J.  Jarnefelt;  B.B.A.  Library,  vol.  11.  American  Elsevier 
Publishing  Co.,  Inc.,  New  York. 

Physiological  Chemistry  of  Lipids 

Masoro,  E.  J.  (1968)  Physiological  Chemistry  of  Lipids  in  Mammals. 
W.  B.  Saunders  Co.,  Philadelphia. 

Chromatography  (Column,  Thin-Layer,  and  Gas-Liquid)  of  Com- 
mon and  Uncommon  Lipid  Classes:  Detailed  Accounts  by  Experts 
in  Individual  Fields 

Marinetti,  G.  V.,  ed.  (1967)  Lipid  Chroinato graphic  Analysis;  vol.  1 
and  2.  Marcel  Dekker,  Inc.,  New  York. 


99 


INDEX 


cerebrosides,  see  glycolipids 
cholesterol,     determination     of,     85 ; 

structure  of,  4 
complex      lipids,      see      gangliosides, 

glycolipids,   phosphoglycerides,   and 

sphingolipids 
column   chromatography,   preparation 

of  columns  for,  31;  separation  of 

neutral  lipids  by,  35;  separation  of 

polar  lipids  by,  36 

dimethylacetals,  78 
see  also  plasmalogens 

esters,  see  methyl  esters 

fatty  acids,  column  chromatographic 
separation  of,  36;  determination  of 
trans  double  bond  in,  94;  GLC 
analysis  of,  75 ;  methyl  esters  of, 
35-37,  48,  51,  75-76;  nomenclature 
of,  4;  structures  of,  5-7,  9;  separa- 
tion on  TLC  of,  48 
fatty  aldehydes,  see  dimethylacetals 
fatty  alcohols,  analysis  of,  79;  defi- 
nition of,  3 
see  also  cholesterol 

gangliosides,  column  chromatographic 
separation  of,  29,  37 ;  determination 
of  N-acetylneuraminic  acid  in,  90; 
structure  of,  17;  TLC  analysis  of, 
53 

gas-liquid  chromatography  (GLC), 
carrier  gases,  columns,  and  detector 
systems  for,  62-66;  chemical  modi- 
fication of  compounds  for,  75 ; 
qualitative  analysis  by,  66;  quanti- 
tative analysis  by,  69 

glycerides,  column  chromatographic 
separation  of,  36;  GLC  analysis  of, 
81 

glycerophosphatides,  see  phosphoglyc- 
erides 

glycolipids,  analysis  of  sugar  in,  89; 
column  chromatographic  separation 
of,  35,  37;  definition,  15;  structures 


of,    15-17;   TLC   separation   of,   52\ 
see  also  gangliosides 
glyceryl  ethers,  structure  of,  8 

lipids,  contamination  of,  20,  26;  defi- 
nition  of,    1 ;   extraction   from  tis-J 
sues  of,  22;  oxidation  of,  19 

methyl  esters  of  fatty  acids,  column 
chromatographic  separation  of,  35- 
37;  GLC  analysis  of,  75;  prepara- 
tion of,  76;  TLC  separation  of,  48, 
51 

N-acetylneuraminic  acid,  determina- 
tion in  gangliosides  of,  90;  struc- 
ture of,  17 

paper  chromatography,   59 

phosphoglycerides,  column  chromato- 
graphic separation  of,  35,  37;  defi- 
nition of,  8;  determination  of 
phosphorus  in,  84;  structures  of, 
10-12;   TLC  separation  of,   52 

phosphonolipids,  13-14 

plasmalogens,    analysis    of,    92;    defi- 
nition of,  12;  structure  of,  13 
see  also  dimethylacetals 

sphingosine,  14 

sphingolipids,  definition  of,  14;  struc- 
tures of,  14-17 

see    also    complex    lipids,    ganglio- 
sides, and  glycolipids 

sialic  acid,  see  N-acetylneuraminic 
acid 

simple  lipids,  definition  of,  1 
see     also     cholesterol,     glycerides, 
fatty  acids,  and  fatty  alcohols 

thin-layer  chromatography  (TLC), 
lipid  detection  sprays  for,  45;  pre- 
paration of  plates  for,  42;  separa- 
tion of  glycolipids  and  phospholip- 
ids by,  52,  55;  separation  of 
neutral  lipids  by,  48;  quantitative 
analysis  by,  53 

waxes,  definition  of,  3 


100 


. 


HHHH8BBBBH