Analytical and Preparative Separation Methods of Biomacromolecules PDF
Analytical and Preparative Separation Methods of Biomacromolecules PDF
Analytical and Preparative Separation Methods of Biomacromolecules PDF
0
0.0 10.0 20.0
Time [min] Timc [min]
edited by
Hassan Y. Aboul-Enein
Analytical and
Preparative
Separation Methods
of Biomacromolecules
Analytical and
Preparative
Separation Methods
of BiomacromoIecuIes
edited by
Hassan Y. Aboul-Enein
King Faisal Specialist Hospital and Research Centre
Riyadh, Saudi Arabia
M A R C E L
MARCELDEKKER,
INC.
D E K K E R
Library of Congress Cataloging-in-Publication Data
Headquarters
Marcel Dekker, Inc.
270 Madison Avenue, New York, NY 100 I6
tel: 2 12-696-9000; fax: 2 12-685-4540
The publisher offers discounts on this book when ordered in bulk quantities. For more infor-
mation, write to Special Sales/Professional Marketing at the headquarters address above.
Neither this book nor any part may be reproduced or transmitted in any form or by any
means, electronic or mechanical, including photocopyng, microfilming, and recording, or
by any information storage and retrieval system, without permission in writing from the
publisher.
Hassan Y. Aboul-Enein
Contents
Preface iii
Contributors xi
Bioaffinity Chromatography 99
Jaroslava Turkovd
Index 457
Contributors
* Current affiliarion: Senior Research Scientist, Biochemistry and Bioanalytical Chemistry Depart-
ment. Astra-Draco AB, l a n d , Sweden
xi
xii Contributors
Nona Remo Rama, B.Sc. Research Technician, Biological and Medical Re-
search, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi
Arabia
Contributors xiii
I. INTRODUCTION
and is more time consuming than HPLC methods. Flow rate is limited by the
compressibility of the resin and its nonuniform particle size. These drawbacks
stimulated development of several methods using ion exchange resins for HPLC,
such as Synchropak CM 300 [9], silica-based carboxymethylpolyamide [lo],
Mono S [8,1 I], and the Glycopak resin of the Diamat analyzer (Bio-Rad). These
methods can usually also be used to measure fetal hemoglobin (HbF). They differ
in their sensitivity to interferences such as adducts formed between Hb and
urea, aldehydes, or acetylsalicylic acid and other hemoglobin (Hb) variants
[12,13].
Glycohemoglobin can also be assayed by affinity chromatography using
boronate agarose columns [14]. This method measures HbAlc together with gly-
cohemoglobins, which elute in the HbA, fraction in ion exchange chromatog-
raphy.
In chromatographic methods, abnormal Hb variants may cause erroneous
HbA,, results [15,16], but at the same time they permit identification of individu-
als with such variants. This information is of importance for the interpretation
of the HbA,, results. Therefore it is an advantage if an HPLC method used for
clinical purposes separates not only HbA,, but also other hemoglobin compo-
nents to permit identification of abnormal hemoglobins. The good resolution
of more than 35 frequently encountered human hemoglobins and HbAlc on
PolyCAT A [I71 makes it a useful method for preliminary characterization of
Hb variants. The Diamat method has been shown to separate seven variants [18],
but many overlap partially with the HbA,, peak.
improved the separation and the column life increased up to several thousand
samples with constant resolution.
B. PolyCAT A
PolyCAT A is a weak cation exchanger with polyaspartic acid linked to silica
[20,2]]. This resin has been used for the determination of HbAlC [22,23], of
several minor molecular forms of hemoglobin [24], and for identification of he-
moglobin variants 1171. The separation of HbALC from other Hb components
such as acetylated Hb [13] and HbF depends critically on buffer composition,
particularly on the pH and salt gradient. It is often necessary to make minor
variations in the steepness of the salt gradient in order to get a good separation.
The assay is precise [22] and it depends more on the quality of the chromato-
graphic separation, the level of noise in the spectrophotometer, and the mode of
integration than on volumes injected.
PolyCAT A with bis-tris buffers and sodium acetate gradients has been
used to separate and quantitate many normal and abnormal Hb components
[23,24]. Minor fetal Hb forms can be separated and HbA,c can simultaneously
be quantitated. We have compared the performance of PolyCAT A chromatogra-
phy with a boronate affinity binding assay and the automated Diamat system for
the determination of HbA,c [23]. Elution was achieved with a linear gradient
consisting of buffers A and B at a flow rate of 1.1 mllmin at room temperature.
Buffer A (pH 6.87) contained 10 mmol of bis-tris and 1.0 mmol of KCN per
liter. Buffer B (pH 6.57) contained buffer A plus 200 mmol of NaCl per liter.
The gradient was: time 0-2 min, 21% B; time 16 min, 47% B; time 22 min, 100%
B; time 24 min, 100% B; and time 26 min, 21% B. Integration was performed by
the valley-to-valley method.
When we compared the two cation exchange methods, PolyCAT A and
Diamat, we obtained an acceptable correlation (Fig. 1). The correlation coeffi-
cient was 0.90 but the regression equation (PolyCAT A = 1.06 X Diamat - 3.06)
showed that much lower results were obtained by PolyCAT A. This suggests that
the HbAlc value measured by the Diamat assay includes a background of
2-3% of the total Hb. This might be due to the fact that the Diamat method also
measures carbamylated and acetylated forms of Hb 1121 and possibly
some other derivatives formed in blood during storage, which can be separated
from the HbAlc peak by the use of methods with higher resolution. The Poly-
CAT A assay has been optimized to separate different Hb variants from HbAlc.
However, the difference between the Diamat method and the PolyCAT A assay
cannot be explained only by carbamylated and acetylated Hbs, for which levels
below 0.4% have been reported [12]. The slope of the regression line, 1.06, shows
that the PolyCAT A method reflects differences in glycated Hb in the same way
Turpeinen and Stenman
0 2 4 6 8 1 0 1 2 1 4
Diamat (%)
Figure 1 Correlation between the HbA,, values obtained by PolyCAT A (y) and Diamat
(x).Corresponding regression equation calculated by the standardized principal compo-
nent is y = 1 . 0 6 ~-3.06, r = 0.90.
as the Diamat method. The negative bias of about 2-3% of total Hb is present
at all levels of HbAlc when PolyCAT A is compared to Diamat.
to HbAlc, also HbF and HbA,. The method has been calibrated against the Diamat
method. In practice, however, there seems to be a slight difference between these
systems, with the Variant giving slightly higher results than the Diamat [25].
and 3.9-5.7% [22] have been reported. Slightly higher reference ranges of 5.0-
7.6% [28] and 4.8-6.4% [29] have been reported for boronate affinity chromatog-
raphy methods. Of the glycated Hb adsorbed to boronate affinity columns, about
50% elutes in ion exchange chromatography as HbA,,, about 40% in the HbAo
peak and 10% as HbA,,,, [28]. The values for the PolyCAT A method can be
estimated to correspond to about 60-70% of those obtained by boronate affinity
chromatography. This suggests that the PolyCAT A method measures the gly-
cated component of the HbAlCpeak fairly specifically. A certain change in blood
glucose causes a larger change in glycated Hb measured by affinity chromatogra-
phy than in HbAlc. This is demonstrated by the slope of the correlation, about
1.4-1.5 [24,28] in spite of similar reference values.
HPLC provides a rapid and sensitive means for the examination of,abnormal
hemoglobins. Methods with high sensitivity and specificity have been developed
for screening and confirmation of hemoglobinopathies in newborns [30,31]. Most
methods have used Synchropak CM 300, a silica support with carboxylic acid
residues or PolyCAT A. The screening procedures are designed to rapidly detect
the major variants. However, the resolution of these methods does not permit
differentiation of some commonly encountered hemoglobin variants such as HbE
17.9 H b b ,
-
r
20.4HbA2 ,
-21.1 HbD
19.7 HbE
7- 22.3 HbS
26.7-
25.1 HbC
Figure 2 Cation exchange chromatography of HbF, HbA,, HbE, HbA,, HbD, HbS, and
HbC on PolyCAT A. Time of elution since sample injection is indicated at each peak.
Detection at 415 nm.
Analysis of HbA,, by HPLC
10 - -
5 9.2 Hb Helsinki
from HbA,. The sensitivity to detect hemoglobin variants occurring at low con-
centrations is also poor.
Recently, Ou and Rognemd [17] described a method for Hb variant analysis
on the PolyCAT A column. The good resolution obtained with more than 35
frequently encountered human Hbs also makes it useful for HbAlcdeterminations
and for preliminary identification of Hb variants. Earlier, PolyCAT A was used
for screening of cord blood for hemoglobinopathies [32]. The column has the
resolution necessary for both screening and confirmatory purposes. We have in-
vestigated the use of a 4.6 X 200 mm PolyCAT A column with 5-pm particles
for separation of Hb variants. A typical chromatographic separation of an Hb
mixture containing Ale, F, A,,, E, A,, D, S, and C is shown in Fig. 2. We have
also separated other Hb variants by using bis-tris buffers and an NaCl gradient
(Fig. 3). The baseline resolution permits accurate quantitation of these hemoglo-
bins, which is important for the correct diagnosis of hemoglobinopathies. In addi-
tion, HbA, can be accurately measured. A slight adjustment of the gradient pro-
gram may be necessary for optimal separation of all variants.
Recently, a new automatic HPLC system using a Bio-Rad MA7 polymeric
cation exchange material has been used to screen newborns for sickle cell anemia
8 Turpeinen and Stenman
10 HbAi c
- 15.1 Hb Broussais
17.7 HbAO
20.2 HbA2
15.7 Hb Cemenelum
'
d
1 18.6 HbAO
21.5 HbA2
VI. CONCLUSIONS
HPLC is a widely used technique for the determination of HbAlC although re-
cently developed immunoassays have started competing with HPLC. Since there
is no general agreement on standardization of the methods for determination of
glycated Hbs, HPLC ion exchange methods measuring HbAlc are used to cali-
brate both boronate affinity methods and some new immunochemical methods
[35].Because of its good precision, ion exchange HPLC has been recommended
as the gold standard for measurement of glycohemoglobin. However, it should
be recognized that the presently used reference method is far from ideal because
it measures some unglycated other Hb variants but not all of the glycated ones.
The ongoing standardization of the HbAlcassay methods initiated by the Interna-
tional Federation of Clinical Chemistry aims at developing a new reference
method.
REFERENCES
Goldstein DE, Parker KM, England JD, England JE Jr, Wiedmeyer H-M, Rawlings
SS, et al. Clinical application of glycosylated hemoglobin measurements. Diabetes
1982;3 1 (suppl. 3):70-78.
John WG. Glycated haemoglobin analyses-assessment of within- and between-
laboratory performance in a large UK region. Ann Clin Biochem 1987;24:453-460.
Bodor GS, Little RR, Garrett N, Brown W, Goldstein DE, Nahm MH. Standardiza-
tion of glycohemoglobin determinations in the clinical laboratory: three years of
experience. Clin Chem 1992;38:2414-2418.
Weykamp CW, Penders TJ, Muskiet FAJ, van der Slik W. Effect of calibration
on dispersion of glycohemoglobin values determined by 11 1 laboratories using 21
methods. Clin Chem 1994;40: 138- 144.
Trivelli LA, Ranney HM, Lai H-T. Hemoglobin components in patients with diabe-
tes mellitus. N Engl J Med 1971;284:353-357.
Allen DW, Schroeder WA, Balog J. Observations on the chromatographic heteroge-
neity of normal adult and fetal human hemoglobin: a study of the effects of crystalli-
zation and chromatography on the heterogeneity and isoleucine content. J Am Chem
SOC1958;80:1628-1634.
B i s i E, Abraham EC. New less temperature-sensitive microchromatographic
method for the separation and quantitation of glycosylated hemoglobins using a non-
cyanide buffer system. J Chromatogr 1985;344:8 1-91.
10 Turpeinen and Stenman
tion of glycohemoglobin results and reference values in whole blood studied in 103
laboratories using 20 methods. Clin Chem 1995;41:82-86.
Fliickiger R and Mortensen HB. Glycated Haemoglobins. J Chromatogr 1988;429:
279-292.
Cefalu WT, Wang ZQ, Bell-Farrow A, Kiger FD, Izlar C. Glycohemoglobin mea-
sured by automated affinity HPLC correlates with both short-term and long-term
antecedent glycemia. Clin Chem 1994;40: 1317- 132 1.
Abraham EC, Perry RE, Stallings M. Application of affinity chromatography for
separation and quantitation of glycosylated hemoglobins. J Lab Clin Med 1983;102:
187-197.
Herold DA, Boyd JC, Bruns DE, Emerson JC, Bums KG, Bray RE, et al. Measure-
ment of glycosylated hemoglobins using boronate affinity chromatography. Ann Clin
Lab Sci 1983;13:482-488.
Wilson JB, Headlee ME, Huisman THJ. A new high-performance liquid chromato-
graphic procedure for the separation and quantitation of various hemoglobin variants
in adults and newborn babies. J Lab Clin Med 1983;102:174-186.
Rogers BB, Wessels RA, Ou C-N, Buffone GJ. High-performance liquid chromatog-
raphy in the diagnosis of hemoglobinopathies and thalassemias. Am J Clin Path01
l985;84:67 1-674.
van der Dijs FPL, van den Berg GA, Schemer JG, Muskiet FD, Landman H, Mus-
kiet FAJ. Screening cord blood for hemoglobinopathies and thalassemia by HPLC.
Clin Chem 1992;38: 1864-1 869.
Eastman JW, Wong R, Liao CL, Morales DR. Automated HPLC screening of new-
borns for sickle cell anemia and other hemoglobinopathies. Clin Chem 1996;42:
704-710.
Turpeinen U, Sipila I, Anttila P, Karjalainen U, Kuronen B, Kalkkinen N, et al. Two
a-chain hemoglobin variants, Hb Broussais and Hb Cemenelum, characterized by
cation-exchange HPLC, isoelectric focusing, and peptide sequencing. Clin Chem
1995;41:532-536.
Karl J, Bums G, Engel WD, Finke A, Kratzer M, Rollinger W, et al. Development
and standardization of a new immunoturbidimetric HbA,c assay. Klin Lab 1993;39:
991 -996.
Analysis of Posttranslational
Modifications in Recombinant
Proteins by HPLC and Mass
Spectrometry
Rainer Bischoff* and Bernadette Bouchon
Transgene S.A., Strasbourg, France
I. INTRODUCTION
* Current aflliation:
Senior Research Scientist, Biochemistry and Bioanalytical Chemistry Depart-
ment, Astra-Draco AB, Lund, Sweden.
13
14 Bischoff and Bouchon
ionization (ESI) [4], and matrix-assisted laser desorption ionization (MALDI) [S]
has opened possibilities for obtaining precise molecular mass measurements on
proteins, peptides, oligonucleotides, and oligosaccharides. This development has
enabled especially the growing biotechnology industry to integrate MS into strat-
egies to characterize recombinant proteins in detail and to assure consistent qual-
ity [6-81:
The' combination of efficient separation techniques such as HPLC and capil-
lary electrophoresis (CE) with MS is rapidly becoming an indispensable analyti-
cal tool in many academic and industrial laboratories [9-141. The power of ana-
lyzing separated components by MS results from the fact that the molecular mass
is an inherent property of a given compound. In the case of recombinant proteins,
where the amino acid sequence is generally known, it is possible to compare the
calculated mass with the measured mass and thus to confirm the expected struc-
ture or to detect modifications. More detailed information can be obtained when
analyzing defined proteolytic digests of a protein by either off-line (fraction col-
lection) or on-line HPLC-mass spectrometry (LC-MS). Such analyses permit lo-
calization of the modified site(s) and collection of precise data about the molecu-
lar mass of the modification. Thus it is not surprising that LC-MS studies have
focused in many cases on the characterization of posttranslational modifications
such as glycosylations and phosphorylations as well as on the identification of
modifications that were due to culture, purification, or storage conditions [IS-
221.
The following studies of two recombinant proteins that are being developed
as vaccine candidates against schistosomiasis or Lyme disease illustrate the char-
acterization of posttranslational modifications by combining MS with chromato-
graphic separation techniques. rSmp28, an antigen that was originally isolated
from the parasite Schistosoma mansoni [23], was expressed to high levels in
the cytoplasm of Saccharomyces cerevisiae, whereas rOspA, an outer membrane
protein of Borrelia burgdolferi, the Lyme disease-causing agent, was produced
in Escherichia coli [24]. Both proteins were shown to be heterogeneous due to
posttranslational modifications as determined by isoelectric focusing in the case
of rSmp28 [25] or by MS for rOspA [26,27]. In this chapter we describe the
analytical strategy that led to the characterization of these modifications, stressing
the importance of integrating efficient separation methods such as HPLC with
MS.
membrane confirmed that both of them contained the expected N-terminal se-
quence excluding N-terminal blocking by, for example, formylation or acetyla-
tion [25] in contrast to the presence of an N-acetylalanine in the natural protein
1281. To further characterize the modification, rSmp2817 and rSmp2812 were
separated by anion exchange HPLC using a shallow gradient of potassium chlo-
ride (KCI) at pH 8.55 (0.5 m M KClImin) (Fig. 2). Anion exchange HPLC allowed
separation of three major forms of the protein (rSmp2811, 2, and 3). The nature
of the modification in rSmp2813, which appeared only after purification, has not
been identified in this case but may be related to the binding of glutathione to
the single free cysteine residue (CyslS9)as previously shown [13]. rSmp2811 and
rSmp2812 were baseline-separated and appeared to be homogeneous when ana-
lyzed by isoelectric focusing in an immobilized pH gradient (Fig. 2, insert). It
is noteworthy that rSmp2812 was slowly transformed into rSmp2811 upon incu-
bation at 37°C in water, underscoring the labile nature of the N-terminal modifi-
cation.
T m e (min)
15 20 25 30 35 40
Time (min)
Figure 4 Separation of the modified rSmp28 N-terminal peptide fragment into two iso-
meric components by reversed-phase HPLC. The separated components proved to have
identical amino acid compositions and molecular masses. Both were inaccessible to N-
terminal Edman degradation. The ratio between the earlier and later eluting peptides was
1 :2 as determined by quantitative amino acid composition analysis. (From Ref. 39.)
Bischoff and Bouchon
Ala-Gly-Glu-His-Ile-Lys
Figure 5 Schematic representation of the structure of the unmodified rSmp28 N-termi-
nal peptide fragment showing the expected number of exchangeable hydrogen atoms and
fragmentation sites.
Measured Mass:665.8 Da
Expected Mass @on-deutemted): 653.7 Da
Mass Difference: 12.1 Da
3
-
P)
0 - I -
691.8
I I l . - - l . ~ ~ ~ l ~ l
Figure 6 Deuterium-hydrogen exchange of the nonmodified (A) and modified (B) N-terminal peptide fragments of rSmp28 in DzO
(expected number of exchangeable protons in the nonmodified peptide = 12; see Fig. 5 ) . The measured mass of the nonmodified peptide h,
A
(665.8 Da) differed by 12.1 Da from the expected mass of the nondeuterated form in agreement with its structure, whereas the measured
mass of the modified peptide (689.8 Da) differed by only 10.0 Da from the measured mass of its nondeuterated form. indicating the loss
of two exchangeable hydrogen atoms. (From Ref. 39.)
Figure 7 Tandem mass spectrometry of the unmodified (A and C; doubly charged parent ion: 328 amu) and modified N-terminal peptide
(B and D; doubly charged parent ion: 341 amu) of rSmp28 using the nanoelectrospray technique [29]. (For exact mass values, see Table
1; the region between 180 and 355 amu is not shown.) Panels A and B show a comparison of the fragments obtained in the range from
40 to 180 Da. It is noteworthy that the a , fragment of the modified peptide appeared at the expected mass (mlz = 44.0) as well as at mlz
= 70.0 (Am: +26.0 Da) confirming that Alal was one of the modified amino acid residues. Furthermore, the y", fragment of the modified
peptide occurred at the expected mass (mlz = 147.0) as well as at mlz = 173.2 (Am: +26.2 Da) (Fig. 7B) identifying Lys6 as another
modified site. The abundant immonium ion i,, which is derived from HisJ [30], occurred at the expected value (nzlz = 110.0) and at
mlz = 136.0 (Am: +26.0 Da) In the modified peptide (see Fig. 7B). These results show that cyclization occurred between Ala' and Lys6
as well as with Hisvsee Fig. 8). These results were corroborated by other a, b, and y" ions in the region between 355 and 650 amu, which
showed a +26 Da series in the case of the modified peptide (Fig. 7D) while the unmodified peptide gave the expected fragment ions (Fig.
7C). Fragment ions at 404.5 and 517.6 Da could not be assigned but are related by a Am of 113.1 Da, which corresponds to Ile. (From
Ref. 39.)
Table 1 Comparison of the Measured and Calculated Mass Values for Fragment Ions Obtained by Nanoelectrospray Tandem Mass
Spectrometry of rSmp28 N-Terminal Peptides (see Fig. 7).
Measured mass (Da) Measured mass (Da) Expected
Fragment ion" Nonmodified peptide Modified peptide mass (Da) Fragment
-
"ccording to Ref. 38
Not detected.
Recombinant Proteins and MS 25
its N terminus with acetaldehyde in the case of the hemoglobin P chain [31].
Furthermore, rSmp2812 is not N-terminally "blocked" when analyzed by Edman
degradation excluding modifications such as N-terminal formylation or acetyla-
tion and again underlining the labile nature of the modification. (2) Tryptic diges-
tion of rSmp2812 produced a modified form of the N-terminal hexapeptide
AGEHIK that was blocked to Edman degradation. The observed mass difference
Ala-Gly-Glu-His-lle-Lys-PROTEIN
/
Tryptic Digestion
pH 8.3
cyclization via His
cyclization via Lys
Figure 8 Schematic drawing of the mechanism leading to the generation of two isomeric
cyclic peptides after tryptic digestion of the aldimine-modifiedprotein rSmp2812. (From
Ref. 39.)
Recombinant Proteins and MS 27
S-[2,3-Bis(palmitoyloxy)-(2RS)-propyl]-N-palmitoyl-(R)-cysteinyl ("Pam3Cys")configuration
(A2, A3, A4, ... : amino acids)
sis by GC-MS. The chromatogram (Fig. 10) shows the fatty acid composition of
rOspA, with fatty acids ranging from C14 to C18. The individual mass spectra
of each of these fatty acids show the molecular ion and the typical fragmentation
pattern permitting the identification of saturated fatty acids (C 14: 0, C 16 :0; A
and C) and monounsaturated fatty acids (C 16 : I, C 18 : 1; B and D) [37]. C 16 :0
(65%) and C16: 1 (21%) were the major components of the total fatty acid con-
tent. To ascertain that the fatty acids were attached to the N terminus as expected,
complementary analyses were performed focusing on the lipidated N-terminal
tryptic peptide.
as expected for the most hydrophobic component of the mixture (Fig. 11A). Anal-
ysis by ESMS of the collected fraction permitted localization of the N-terminal
peptide based on its mass and ionization profile (Fig. 11B). Due to the very short
peptidic chain (a single peptide bond, C'K2, Fig. 9), absorption at 205 nm was
very low, and the peptide appeared as a shoulder on the major chromatographic
peak. The purified peptide ( < I pg) was collected for further analysis.
Tryptic peptides are generally characterized during ESMS analysis by the
presence of two ionization positions (The N terminus and the basic C-terminal
residue, except for the C-terminal peptide of any digest). In the case of rOspA,
however, the N-terminal peptide presented only one ionization site due to N-
terminal blockage. The most abundant peak in this spectrum (Fig. 11B) corre-
sponded to a molecular mass of 1036.2 Da (expected mass of the N-terminal
peptide containing three palmitates: 1038.6 Da; Am = -2.4 Da), indicating that
one fatty acid was unsaturated. In addition, several minor components were de-
tectable. Peaks corresponding to plus and minus 28Da (I1 and I11 in Fig. 11B)
were present, indicating different forms of the lipidic moiety, the major one being
a peptide with three C16 fatty acids, whereas the minor forms contain two C16s
plus one C14 (I1 in Fig. 118) and two C16s plus one C18 (111 in Fig. 11B) in
agreement with the GC-MS data. Another peak corresponded to the loss of one
palmitate residue (IV in Fig. 1IB, Am = -238 Da). This could be attributable
to a fragmentation taking place in the source of the mass spectrometer, or most
likely to an incomplete lipidation, as previously described on the same molecule
[26]. The presence of a population of Pam2Cys-modified rOspA molecules that
had been detected in the intact protein was thus confirmed by the analysis of the
N-terminal peptide mixture that contained some Pam2CyrLys (IV in Fig. 1 IS;
expected mass, 800.2 Da; measured mass, 797.8 Da). The measured mass of the
N-terminal tryptic peptide appeared to be 2 Da lower than expected for two
C16:O indicating one unsaturated fatty acid in agreement with the GC-MS data
which showed the presence of about 20% of C16: 1.
To confirm these results, a fast atom bombardment (FAB) mass spectrum
using a magnetic sector mass analyzer was run on approximately 100 ng of the
remaining N-terminal peptides. A complex spectrum was obtained (Fig. 12). As
the sample had remained in solution for several days prior to FABMS analysis
it is likely that the increased complexity was due at least in part to oxidation of
the peptide at the cysteine-lipid thioether moiety. Additional peaks (I1 and I11
in Fig. 12 and Table 2) corresponding to plus and minus 28 Da on [M+H]+ were
present, confirming and extending the ESMS data on the lipidic composition. In
particular, the high mass resolution allowed discrimination of two components
differing in one unsaturation (Ia and Ib in Fig. 12 and Table 2). Fragmentation
(b, fragment) data indicated the presence of lysine in agreement with the expected
structure of the N-terminal peptide containing one cysteine and one lysine resi-
due. Masses corresponding to the loss of one palmitate residue (IV in Fig. 12.
Recombinant Proteins and MS 33
Table 2 Molecular Species Detected in the ES and FAB Mass Spectra of the Lipidated N-Terminal Tryptic Peptide of rOspA
Average mass (Da) Monoisotopic mass (Da)
Theoretical fatty Measured Measured Label in
acid composition Expecteda (ESMS) Am Expecteda (FABMS) Am Proposed structures Figs. 3 and 4
Three C16:O 1038.6 1036.2 -2.4 1037.8 1035.7 -2.1 C16:O-C16:O-C16: 1 Ia
1037.7 -0.1 C16:O-C16:O-C16:O Ib
Two C16:O + One C14:O 1010.6 1008.9 -1.7 1009.8 1007.5 -2.3 C16:O-C16: 1-C14:O I1
Two C16:O + One C18:O 1066.7 1063.7 -3.0 1065.9 1063.6 -2.3 C16:O-C16:O-C18:l 111
Two C16:O 800.2 797.8 -2.4 799.6 797.3 -2.3 C16:O-C16:l IV
"ssuming completely saturated structures.
Recombinant Proteins and MS 35
Am = -238 Da) confirmed that the major fatty acid residue was C16:O. These
results indicated the fatty acid composition of the major components to be C16:
0-C16: 0-C16: 1 and C16: 0-C16: 0-C16: 0 in accordance with results obtained
by GC-MS. Of the two minor components, one had a lipidic composition of C14:
0-C16: 1-C16:0, and the other component had a lipidic composition of C18 : 1-
C16 :0-C 16 :0, according to the GC-MS data.
In conclusion, the use of a combination of GC, normal phase HPLC, and
MS allowed a detailed characterization of the lipidation of rOspA. The heteroge-
neity of the molecule related to its lipidic composition was analyzed using fatty
acid analysis by GC-MS. It indicated that C16:O is the major component as ex-
pected [35] but that other fatty acids including C 16 : 1, C 14:0, and C 18 : 1 were
also present. Purification of the N-terminal peptide and its analysis by ESMS and
FABMS indicated that a minor fraction of rOspA molecules contained a Pam2-
Cys modification and that the major rOspA form corresponded to a peptide having
three C16 residues, two of them being saturated and one being unsaturated. This
structure is consistent with the proposed mode1 for bacterial lipoproteins [35] in
that the major fatty acid component is palmitate.
IV. DISCUSSION
ACKNOWLEDGMENT
The contributions from colleagues in the Yeast and Pilot Development Depart-
ments at Transgene and the Laboratoire de Spectromktrie de Masse Bioorganique
(Universitk Louis Pasteur, Strasbourg) are gratefully acknowledged.
REFERENCE
I. INTRODUCTION
In all of these equations A, B, and C are empirical constants that are depen-
dent on the electrolyte system used and on the size and structure of peptides
investigated. Small peptides may behave as simple organic ions of a compact
structure (tightly would coil); longer peptides may occur in the form of random
coil or linear chain.
In spite of different forms of the above given relation it can be concluded
that the mobility of peptide is directly proportional to its charge and indirectly
proportional to its relative molecular mass with exponent in the range 113-213.
The influence of charge, size, and shape of peptides and proteins on their
mobilities is discussed in detail in some special articles, book chapters, and re-
views [9- 171.
2. Effective Charge
From the physicochemical point of view, peptides are amphoteric (po1y)electro-
lytes or (po1y)ampholytes. They contain in their molecules different types of iono-
genic groups, e.g., carboxyl groups of C-terminal amino acids and of side chains
of aspartic and glutamic acids, amino groups of N-terminal amino acids and of
side chain of lysine, etc. The survey of ionogenic groups present in peptides and
the approximate range of their acid dissociation constants excerpted from the
literature [18-201 are given in Table 1. The effective (net) charge of peptides
(sum of all charges including their signs) is given by the sum of charges of all
ionogenic groups present in the polypeptide chain. If amino acid sequence or at
least amino acid composition of a peptide is known, then from the present iono-
genic groups and from their dissociation constants it is possible to estimate which
groups are dissociated at a given pH and how they conribute to the effective
charge. Generally, a given group is dissociated to 50% in a solution the pH of
which is equal to the pK value of that group. For pH = pK + 1, the group is
90% dissociated and for pH = pK - 1 the group is 10% dissociated.
The amino acid residues of aspartic and glutamic acids are the main contrib-
utors of negative charge, whereas arginine, lysine, and partly histidine are camers
of the positive charge in peptide chain.
In addition to this rough estimation, a more precise, computer program-
based calculation has been developed that allows one to calculate the effective
and specific charges of any peptide whose amino acid sequence is known and
for which the pK values of present ionogenic groups are known or can be esti-
mated [21]. The program is based on the mathematical model of the acid-base
equilibria of a general ampholyte. It is described in detail elsewhere [22]; here
only the main points will be mentioned.
Let us consider a peptide P that can have maximal (positive) charge M and
minimal (negative) charge N. M and N express numbers of elementary charges
including sign, i.e., maximum and minimum are meant in the mathematical sense.
Let P(J) be an ionic form of peptide P with charge J and K(J) is the apparent
dissociation constant of the equilibria between the ionic forms P(J + 1) and P(J):
where J E (N, M), J f 0, H = CH. If molar fractions of all ionic forms P(J) are
calculated, then the effective charge of peptide P, zPzef,
can be determined:
or the ratio of effective charge and relative molecular mass with a general expo-
nent d, called corrected specific charge, z ~ , ~ ~ , ~ ~ ~ :
From the known molar fractions Dp(J,of the individual ionic forms of peptide P
and from their known or estimated ionic mobilities, mp(,,,the effective mobility
of peptide P can be calculated as the weighted sum of ionic mobilities:
where sign(J) = + 1 for J > 0, sign(J) = -1 for J < 0, and sign(J) = 0 for
J = 0.
Equation (12) represents a mathematical definition of effective mobility of
a general ampholytic electrolyte. Mobilities of cations are positive values, mobili-
ties of anions are negative values.
The calculated dependencies of effective, specific, and corrected speci-
fic charges [effective charges divided by relative molecular mass M, with expo-
nent d = 213; see Eq. (1 I)] of simple peptides of diglycine (N = - 1, M = 1,
M, = 132.2, pK(0) = 3.12, pK(- 1) = 8.17) and triglycine [N = - 1, M = 1,
M, = 189.3, pK(0) = 3.26, pK(-1) = 7.911 on pH are shown in Fig. 1.
It is obvious that from these dependencies the useful information for
the selection of pH and composition of background electrolyte (BGE) can
be obtained, e.g., the regions of minimal (negative) and maximal (positive)
charges, regions of strong or weak dependence of effective and specific charges
on pH, and the pH range in which peptide charge is close to zero can be deter-
mined.
An important electromigration characteristic of peptides is their isoelectric
point (PI), i.e., the pH value of a solution in which the effective charge and
consequently the effective mobility is equal to zero. For small oligopeptides con-
taining only few amino acid residues with distant pK values of their ionogenic
groups, the pH range in which effective charge is close to zero is relatively broad
(see Fig. 1) and instead of isoelectric point we are speaking about isoelectric
zone. Isoelectric point differs from the similar characteristic, isoionic point, in
that it is related to a given buffer coniposition in which the measurement of
charge (mobility) is performed. This includes electrostatic interactions with all
the ions present in the solution whereas the isoionic point takes into account only
interactions with protons. The isoelectric and isoionic points are mostly close
values, but generally not identical. Peptide moves in a dc electric field as cation
at pH < pI and as an anion at pH > PI. The isoionic point can be read from the
above given dependence of effective charge on pH (see Fig. 1).
The calculated pH dependence of effective charges and of corrected specific
charges, i.e., effective charges divided by relative molecular mass with exponent
213, for longer peptides with higher number of ionogenic groups. growth hor-
mone-releasing peptides (GHRPs), insulin and desoctapeptide insulin, are pre-
sented in Figs. 2 and 3. Figure 2 shows these dependencies for His'-GHRP (hexa-
peptide with the sequence His-D-Trp-Ah-Trp-~-phe-Lys.NH?and with the
following parameters of calculation: N = 0, M = 3, M, = 873.1. pK(2) = 5.8,
pK(1) = 7.8, pK(0) = 10.4) and for Tyrl-GHRP (hexapeptide with the sequence
Tyr-D-Trp-D-Ala-Trp-~-phe-Lys.NH~ and with the following parameters, N = - 1,
M = 2, M, = 899.1, pK(1) = 7.6, pK(0) = 10.0, pK(-I) = 10.4). Figure 3
shows the pH dependencies of effective and specific corrected charge of hu-
man insulin (two-chain polypeptide consisting of 51-amino-acid residues,
M, = 5750.0, N = - 10, M = 6) and des-B23-30-octapeptide insulin (43-amino-
acid residues, M, = 4837.8, N = -9, M = 5). The accuracy of these calculated
Separations of Peptides by CZE and FFZE
Effective charge
Gly2 -
Gly3 - - - - .
SpeclBc charge
0.0072t '1' ' ' ' ' ' ' ' ' ' ' 4
Figure 1 Dependence of (A) effective charge, (B) specific charge (effective charge di-
vjded by relative molecular mass), and (C) corrected specific charge (effective charge
divided by relative molecular mass with exponent 213). of diglycine, c ~ w Gly2, e and
triglycine, curve Gly3, on pH. Parameters of the calculation, pK values, and relative rnolec-
ular masses are given in the text.
Effective charge
3.0
Hisl-GHRP -
Tyrl-GHRP - - - - -
1.0 -
0.0
!
I
A
,'.
Figure 2 Calculated dependence of (A) effective charge and (B) corrected specific
charge (effective charge divided by relative molecular mass with exponent 213) of growth
hormone-releasing peptides (GHRPs), His'-GHKP and Tyr'-GHRP, on pH. Peptide se-
quences and parameters of the calculation are given in the text.
Effective charge
Insulin -
DO1 ---- -
-4 -
-8 -
A
0 2 4 6 8 10
pH
14
0.0078
Figure 3 Calculated dependence of (A) effective charge and (B) corrected specific
charge (effective charge divided by relative molecular mass with exponent 213) of pig
insulin and desoctapeptide-B23-B30-insulin (DOI) on pH. Parameters of the calculation
are given in the text.
the conclusions derived from the calculated dependencies can be used only as
the first approximation for the selection of the suitable pH of BGE, which can
be further optimized by experimental tests. Similar strategy for the prediction of
peptide charge and mobility from the peptide sequence has been published by
Cifuentes and Poppe [8] who try to predict more precise values of individual
pK values of ionogenic groups present in peptide chain taking into account the
environment of given ionogenic group.
The pH dependence of the effective charge and/or effective mobility of
peptides can also be obtained by other methods, e.g., by acid-base titration curves
[23] or electrophoretic titration curves [24,25]. A large set of mobilities of small
oligopeptides in the pH range 6-10 was determined by capillary isotachophoresis
[26]. Isoelectric point of peptides can be determined by capillary or slab-gel
isoelectric focusing 127-291 or by CZE [30], or it can be calculated theoreti-
cally on the similar bases as the above-described approach for acid-base equi-
libria [3 1,321.
The pH of BGE should be at least 1-2 units distant from p l since close
to p l both electrophoretic mobility and solubility of peptide are relatively low.
The pH of BGE should be taken from the region where the specific charge is
greater than about 2 X e (e is elementary charge unit), i.e., a polypeptide
with relative molecular mass 5000 should possess at least one elementary charge
per molecule. With respect to the fact that effective mobility is directly propor-
tional to the effective charge and indirectly proportional to the relative molecular
mass [see Eqs. (2)-(5)], it is obvious that the difference of peptide mobilities
will be maximal in the same region where the difference of specific charges or
corrected specific charges of these peptides is maximal. Consequently, from this
region of maximal difference of charges the suitable pH of BGE should be
chosen.
For the given example of diglycine and triglycine it is evident that the
suitable pH for their separation is either at low acid region (pH < 3) or at alkaline
pH (pH > 9), since in these regions the differences of specific and corrected
specific charges are maximal. On the basis of these calculations and with respect
to other aspects (see below, 0.5 mol/L acetic acid, pH 2.5, was chosen as BGE
for the separation of these two peptides, which are often used as test mixture
in our laboratory. The record of CZE separation of these two peptides and the
electroosmotic flow marker, phenol, is given in Fig. 4.
Similarly, from the course of pH dependence of corrected specific charge
of GHRP derivatives (see Fig. 2), it follows that the suitable region for their
separation is in the acid pH region and that for pH < 4.5 the difference of cor-
Figure 4 CZE separation of test mixture of diglycine (0.3 mglml), peak 1, triglycine
(0.3 mglml), peak 2, and phenol (0.1 mglml), peak 3, using 0.5 mol1L acetic acid, pH
2.5 as BGE in a home-made CZE device described in Section 1II.B.I. Capillary: i.d. 0.056
mm, total (effective) length 310 (200) mm. Voltage 9.0 kV. current 10.1 FA; AZObr ab-
sorbance at 206 nm; t. migration time.
rected specific charges (effective mobilities) is independent of pH. Separation at
high pH (pH > 11) should be from the point of view of different specific charges
also possible but it cannot be recommended because of hydrolysis of amide
groups at high pH. As follows from the calculated pH dependence of effective
and corrected specific charges of insulin and desoctapeptide insulin (see Fig. 3),
the suitable pH of BGE for CZE separation of these two peptides is between 7
and 9 and from this range the pH of the BGE was chosen for monitoring of
tryptic conversion of pig insulin to desoctapeptide insulin by CZE (see Fig. 9 in
Section 1II.B.1).
According to the predicted suitable pH for peptide separation the composi-
tion of BGE is selected. The main constituent(s) of BGE should possess buffering
capacity at a given pH, i.e., their pK should fulfill the conditions pHBGE= pK
+ 0.5. The ionic strength of the buffer is typically in the range of 10-200 mM
concentrations of the main buffer constituents. At a given pH/ionic strength com-
bination, several buffers can be used in principle. Preference should be given to
the amphoteric, so-called Good-type buffers, which are available for the pH re-
gion 5.5-1 1 and which fulfill the condition of sufficient buffering capacity at
relatively low electric conductivity [39]. The classical buffers such as phosphate,
citrate, and acetate can be used in moderately low pH regions, and borate and
phosphate buffers can be used in weakly alkaline pH range. Common buffers
utilized for peptide separations by CZE and FFZE are given in Table 2.
Of course, the pH of BGE must be selected not only according to pH depen-
dence of effective and specific charge of peptides to be separated, but also with
respect to their solubility, chemical and thermal lability, and biological activity.
These aspects are discussed in more details in the following sections.
D. Detection
1. Optical Detection
Similar to the other classes of compounds, optical detection is the most frequently
used detection mode in CZE separation of peptides. Of the several optical detec-
tion modes used in CZE [57], only (spectro)photometric UV absorption and fluo-
rescence detection will be mentioned here in more detail. Descriptions of the
other modes, used only rarely for peptide detection, say for refractive index and
thermooptical absorbance detection, can be found in the above given review [57].
a. UVAbsorption Detection Thanks to the relatively strong absorption of
peptide bond CO-NH in the short-wavelength UV region, absorption detection
at these wavelengths (200-220 nm) can be used as a universal detection principle.
Special UV absorption detectors have been designed utilizing, for example, high-
frequency excited iodine discharge lamp with emission at 206 nm [58,59] or Zn
or Cd lamps with emission at 214 nm [60], in addition to those utilizing classical
deuterium lamp with continuous light emission in the whole UV region [61].
With the fused silica capillaries detection below 200 nm. up to 185 nm, is also
possible [62]. Due to the higher molar absorption coefficient of peptides at this
region the detection sensitivity is increased, e.g., twofold gain in the signal-to-
noise ratio was obtained by changing the detection wavelength from 2 14 to 200
nm [63], but the choice of BGE is limited to those buffers that do not absorb at
this wavelength. Borate and phosphate buffers are useful in this respect, but many
biological Good's buffers are inappropriate for use below 215 nm. For polypep-
tides separated in the presence of low-UV-absorbing BGE constituents and addi-
tives, 230-nm wavelength provides a good compromise between detectability and
peak-area analysis accuracy on one side and stronger light absorption of the
background electrolyte on the other side.
The UV absorption of the peptide zone in the spectral region 200-220 nm
characterizes the peptide bond quantity, i.e., a longer peptide provides a higher
absorbance signal response at equal molar concentration [64]. Micromolar con-
centrations of peptides are usually detectable by capillary electrophoresis (CE)
with UV absorption detection. Increased sensitivity can be achieved at some spe-
cial designs, as a Z-shaped cell, a bubble cell, or a sleeve cell [61,65,66].
Peptides containing aromatic amino acid residues can be detected more
specifically by UV absorption at 275-280 nm or with a lower sensitivity at 254
nm. The highest absorption coefficient is characteristic for tryptophan residue and
also for tyrosine residue, where significant changes of the absorption coefficient
influenced by pH occur. Less sensitive is the detection of phenylalanine residue.
Although the absorption coefficients and hence the resulting detectability of aro-
matic residue containing peptides at 280 nm is lower than at 206 nm, more free-
dom in selection of BGE composition partially compensates for this drawback.
More information about the quality and quantity of peptides separated and
confirmation of peak homogeneity and identity can be obtained by multiple-
wavelength detection or by full spectrophotometric detection realized by fast
scanning spectrophotometric detection or by diode array detection [61]. Spectral
analysis and the use of spectral libraries and automated library searches can
distinguish closely related peptides, e.g., cyclic and linear forms of synthetic
oligopeptides, and allows identification of individual peaks of peptide maps of
proteins [67].
b. Fluorescence Detection
Laser-Induced Fluorescence Detection of Native Peptides By its inherent
character (measurements are performed with a negligible background signal)
fluorescence detection is more sensitive than absorption detection. In the first
generation of CE devices the fluorescence detectors with classical light sources
(xenon lamps) used in HPLC were adapted for capillary columns [68]. They
suffered from the same drawback as the adapted HPLC UV absorption detectors,
i.e., the amount of light coupled to the analyte zone in the capillary was rather low.
More successful versions of fluorescence detectors use lasers as sources of
excitation light [69-721. Laser-induced fluorescence (LIF) is the most sensitive
detection mode in CZE. With special designs of liquid sheath-flow cuvettes, LIF
detection is approaching the absolute limit-detection of a single molecule [73].
Separations of Peptides by CZE and FFZE 55
portant for peptide mapping and for separation of peptides, i.e., fragments of
proteins originated from specific enzymatic hydrolysis of proteins [20,85]. These
peptide maps are frequently used for the confirmation of known protein sequences
[I021 and for the investigation of posttranslation modification, e.g., phosphoryla-
tion of proteins [103]. ESI is the preferred mode of coupling CE apparatus with
mass analyzer, namely, because of its capability to generate the ions with multiple
charges so that the ion mlz (masslcharge) values for even very large species
(such as polypeptides and proteins) may fall within the limited mlz detection
range of most mass spectrometers. Equally important is the fact that a population
of multiply charged ions having different numbers of charges is created for these
larger ions so that the mlz data can be mathematically deconvoluted to give the
original molecular masses of the species. CE-ESI-MS is now a widely used tech-
nique for separation and identification of closely related peptides, e.g., neuroten-
sin and angiotensin analogs [104], model mixtures of synthetic peptides
[105,106], and naturally occurring biologically active peptides, e.g., enkephalins,
oxytocin, bradykinin, angiotensin, and bombesin at femtomole to attomole levels
[107]. An example of ESI-MS detection of CZE-separated peptide and protein
mixtures is shown in Fig. 5. Combination of CZE with tandem MS, CZE-ESII
MSIMS [log-1101, allowed determination not only of molecular mass of the
whole molecules of proteins and peptides but also of their fragments. From the
collision-induced production-ion spectra the amino acid sequences of CZE-sepa-
rated peptides and proteins were obtained, which allowed their identification by
comparison of these sequences with those in a protein database [109,110]. For
on-line CZE-MSIMS coupling the best signal-to-noise ratio was obtained when
acidic buffers of low ionic strength for CZE peptide separations were applied
[log]. Extremely large number of components of complex peptide mixtures origi-
nating, for example, from protein hydrolysis (peptide mapping) can be resolved
and identified when the MS detector is coupled to an on-line combination of
HPLC and CZE, and in such a way a three-dimensional separation. HPLC-CZE-
MS, is realized [ I l l ] .
The newest version of MS, matrix-assisted laser desorption-ionization mass
spectrometry with time-of-flight mass analyzer (MALDI-TOF-MS), is combined
with CZE separations of peptides and proteins mostly in an off-line mode
[112,113]. Its advantage, i.e., soft ionization and generation of predominantly
singly charged molecular ions of even polypeptide and protein macromolecules
as antibodies [I 141, is used for exact determination of relative molecular mass
with an accuracy of 20.1% and for identification of peptides and proteins isolated
after their CZE separation. In some experimental arrangements the CZE fractions
are deposited directly on the MALDI probes so that individual peaks from electro-
phoreogram are associated with a single sample spot on the probe [112]. MALDI
matrices with high acid concentrations afford enhanced tolerance of CZE buffers
to be used for introducing peptides to the mass analyzer. Keough et al. [115]
mi. 531 A Bradykinln
1 1 (MW 1060)
mlz 749
1
1 S p e n Whale Myoglobln
(MW 17.199)
mtz 1157
1
1
m l z 808
1
Porcine Insulin
I
Horse Myoglobin
(MW 5 7 7 8 ) (MW 16.950)
(M.5H)"
0 10 20 0 10 20 30 40
Time (mln.1 Time (min.)
reported a 250-fmol detection limit for smaller peptides and a 100-fmol off-line
detection limit for a-lactalbumin. The MS detection also enables one to detect
and characterjze absolutely the nonpeptidic part, e.g., the glyco-component,
attached to the peptide chain [ I 161.
with that of fluorescence detection methods. The relative simplicity and low cost
of EC instrumentation, including the ease with which the small electrodes can
be constructed and the associated small currents measured, make the EC methods
particularly attractive for CZE applications [117-1191. Most of EC detection in
CZE are performed in an amperometric mode. The problem of "decoupling" of
small EC potentials (about 1 V) and currents (in the pA range) from the much
larger driving CE voltage (10-30 kV) and current (tens of FA) was solved by
using porous glass joint or porous membrane covered fracture decoupler or by
end-column application of the working electrode [I 18,1201. The major drawback
is the small number of analytically important species that are electroactive at
modest potentials at the carbon electrodes conventionally used, e.g., catechols,
phenols, aromatic amines, and thiols. This is also the reason why EC techniques
have not been applied to the detection of peptides very frequently up to now.
Thiol-containing peptides and amino acids, e.g., glutathione, cysteine, and cys-
tine, were determined by amperometric detection at copper microelectrode [121]
or at gold/mercury amalgam microelectrode [122]. With the latter the detection
limits for glutathione were about 0.5 fmol or 20 nmol/L. Pulsed amperometric
detection with gold fiber microelectrode was used for characterization of glyco-
peptides from recombinant coagulation factor VIIa separated by CZE [123].
An end-column detection cell that can be used for both electrochemical
and conductivity detection in CZE was designed by Huang et al. [I241 and by
Muller et al. [125]. A few other types of conductivity detectors for CZE have
been developed 1126-1281, but mostly they are not used for direct detection of
peptides in CZE. However, conductivity detection can be recommended for CZE
and CITP determination of small organic and inorganic ions, e.g., anionic coun-
tenons (acetates, trifluoroacetates) of basic peptide preparations [21,129,130].
It must be emphasized that both ways of expressing the CZE purity degree
can be used only as approximate and relative criteria of peptide purity since
the molar absorption coefficients of the individual sample components may be
generally different. However, in the case of synthetic peptide preparations most
of the admixtures will have the similar structure to that of the main synthetic
product and consequently also the similar molar absorption coefficients.
Generally, peak height is less suitable for quantitative analysis than peak
area, since peak height is more dependent on the experimental conditions. Stack-
ing or destacking of the sample zone can occur due to concentrating effect of
the BGE constituents or the sample matrix components [150]. Differences in
mobilities of analytes and BGE constituents cause electromigration dispersion of
analyte zones, which also influences their heights.
The peak height-based CZE purity degree is recommended to be used for
CZE-grams with great difference in migration times and with similar peak shape
and width and when the signal-to-noise ratio is too low for precise integration
of peak area. Because of limited linearity between sample amount (concentration)
and peak height the difference in the analyte amounts (concentrations) should be
within one order.
The advantage of peak area-based quantitative evaluation is that the above-
mentioned peak shape distortions do not affect peak area. However, if peak area
is used for quantitative evaluation, then a correction for different migration veloc-
ities of the analytes has to be taken into account. Due to these differences, analytes
with lower migration velocities (mobilities) remain in the detection window for
a longer time than those with higher migration velocities (mobilities). Conse-
quently, they exhibit broader peaks with a larger peak area than the faster moving
analytes, although the physical length of their zones may be the same as that of
the faster moving analytes. The deviation of peak area caused by this effect can
be corrected by dividing the peak area of an analyte by the migration time of
this analyte 11511.
The migration velocity noncorrected peak area-based CZE purity degree
can be used for evaluation of CZE grams where the peaks of some components
are broader not because of slow migration through the detector window but due
to the adsorption of analytes to the capillary wall or due to electromigration dis-
persion.
A further few applications of CZE to peptide analysis are demonstrated.
All of the presented examples were performed on the home-made CZE device
developed in our laboratory [152]. It consists of the untreated fused silica capil-
lary with outer polyimide coating (i.d. 0.056 mm, 0.d. 0.200 mrn, total length
3 10 mm, effective length 200 mm), UV photometric detector at 206 nm, and a
high-voltage power supply. Peptide samples and electroosmotic flow markers
were dissolved in the BGE in the concentration range 0.1 - 1.5 mglml. The sample
was introduced into the capillary manually forming a hydrostatic pressure (50
mm of water column) for the time period 5-20 s. High-voltage power. supply
was utilized in constant voltage mode. Experiments were performed at ambient
temperature 22-24°C without active cooling of the separation compartment.
Application of CZE to the determination of peptide purity is demonstrated
in Fig. 6. It shows CZE analysis of standard preparation of dalargin, a synthetic
hexapeptide with the sequence H-Tyr-D-Ala-Gly-Phe-Leu-Arg-OH, and a deriv-
ative of dalargin, a synthetic hexapeptide with the sequence of H-Tyr-D-Ala-
Gly-D-Phe-Leu-Arg-NH2.Whereas no admixtures were found in the standard pre-
paration, i.e., its CZE purity degree approaches loo%, in the case of dalargin
derivative in addition to the peak of the main synthetic product two admixtures
with lower mobility are present. Peak height-based purity degree of the dalargin
derivative preparation is 86.1% and peak area-based purity is 89.6%.
CZE analysis of insect oostatic hormone fragment, a synthetic octapeptide
with the sequence H-Tyr-Asp-Pro-Ala-Pro-Pro-Pro-Pro-OH (see Fig. 7), shows
the utilization of CZE as a control technique for evaluation of the suitability of
other separation technique, namely reversed-phase HPLC (RP-HPLC), for pre-
parative separation (purification) of a given peptide preparation. Comparison of
CZE analysis of crude synthetic product of insect oostatic hormone and of the
same product purified by RP-HPLC (see Fig. 7) shows that RP-HPLC was a
very efficient method in the purification procedure; it was capable of removing
completely four admixtures (al, a2, a4, a5) of the five admixtures (al-a5) of the
crude synthetic product and to decrease substantially the content of the admixture
a3, but for the complete purification of the preparation further technique has to
be employed or RP-HPLC has to be repetitively used.
The high separation power of CZE is demonstrated by the separation of
Separations of Peptides by CZE and FFZE
IW1
lW1
I , . . . . . . . . . I . . . . . . . . .
human insulin (HI) and its closely related derivatives and fragments, such as HI
with D-phenylalanine in positions 2 4 and 2 5 of B chain, HI with N-phenylacetyl-
protected amino group of lysine in position B29, L-Phe- and D-Phe-B-24,B25-
octapeptide-B23-B30-HI (see Fig. 8).
CZE can b e used not only for qualitative and quantitative microanalysis of
0 20 40 60 80 100 120 la M 180 a00
l i n e Isecl
IW1
0.sssoC , , , , , , , , , , , , , , , , , , , ,d
0 20 40 60 80 100 120 la M 180 ~KI
line lsecl
peptide preparations but also for monitoring of chemical and enzymatic conver-
sions of peptides. Figure 9 shows CZE monitoring of enzymatic cleavage of pig
insulin (PI) by trypsin 150 pg of pig insulin was dissolved in 200 p1 of the
background electrolyte (10 mM tricine, 5.8 mM morpholine, 10 mM tricine, 20
mM NaCl, adjusted by 0.1 M NaOH to pH 8.0) developed earlier for the separa-
tion of PI and desoctapeptide insulin. Composition and pH of the BGE was cho-
sen on the basis of the calculated pH dependencies of effective, specific, and
Separations of Peptides by CZE and FFZE
Figure 8 CZE separation of human insulin (HI) and its derivatives and fragments. Sam-
ple components dissolved in ,BGE in concentration range 0.5-1.5 mglml. BGE: 10 mM
tricine, 5.8 mM morpholine, 20 mM NaCl, pH 8.6 adj. by 0.1 M NaOH. constant current
25 FA, voltage 8.6 kV. Capillary: same as in Fig. 6. 25, Human insulin (HI); 34, [D-Phel-
B24,B25-HI; 39, [~-Phe]-B24,B25-[Lys-N-PhAc]-B29-HI, 40, [Lys-NPhAcI-B29-HI;35,
[~-Phe]-B24,B25-0ctapeptide-B23-B30-H1; 36, [L-Phel-B24,B25-octapeptide-B23-B30-
HI; NA, nicotinamide (electroosmotic flow marker). A:, absorbance at 206 nm: t, migra-
tion time.
0 . m
D RH
omm
Separations of Peptides by CZE and FFZE
Figure 9 CZE analysis of pig insulin (a) and monitoring of the cleavage of pig insulin
by trypsin after 7, 20, 40, and 60 min (b-e). BGE: 10 mM tricine, 5.8 mM morpholine,
20 mM NaC1, pH 8.0 adj. by 0.1 M NaOH, constant current 20.0 PA, voltage 7.3 kV.
Capillary: same as in Fig. 6. For more details, see the text. PI, pig insulin; DOI, desoctapep-
tide-B23-30-insulin; NA, nicotinamide.
apillary Electrophoresis
PVDF Membrane
3MM Filter
papers
Ground Electrode
MEMBRANE
ASSEMBLY
Figure 10 Schematic diagram of the membrane fraction collection for capillary klectro-
phoresis. (From Ref. 174.)
ration of some of the solvent the collected sample component can be reconcen-
trated to the level detectable by other off-line detection methods. For this post-
column off-line detection and/or characterization of the separated peptides,
mostly different modes of MS detection or amino acid and sequence analysis
are used [167-1701.
For the continuous fraction collection in CE it is necessary to use special
designs of the separation capillary in which the electrical circuit is completed
prior to its outlet. This was achieved by the use of a porous glass joint [171], an
on-column frit [172], or on-column microfractures of the capillary [173]. Then
the sample components can be collected at an electrically isolated exit, while
maintaining the electrical connection between high-potential and grounded elec-
trode.
Another adaptation of the CZE system for micropreparative purposes
consists of connection of the outlet end of the capillary with the membrane
assembly [174], as indicated in Fig. 10. This membrane assembly, consisting of
Immobilon transfer membrane (used for protein blotting) and of two
layers of filter paper serving as electrolyte reservoir and stainless steel ground
electrode, rotates and the sample components eluted by electromigration and by
electroosmotic flow from the outlet end of the capillary are adsorbed to the slowly
rotating membrane assembly. Proteins and peptides caught on the Immobilon
membrane are then detected by staining, e.g., with Coomassie blue, and can be
Separations of Peptides by CZE and FFZE 69
in the chamber. At the outlet side of the chamber the separated sample compo-
nents are collected in the fraction collector.
The advantage of FFZE is that it works continuously, in a free solution
under mild conditions in which biological activity of separated substances is pre-
served. FFZE can be applied to preparative separation of a wide range of both
low molecular mass and high molecular mass species and also for the separation
of cells, organelles, and other bioparticles [187,189- 1921.
Because of some problems with thermoconvection and sedimentation in
FFZE processes on the earth, some attempts to perform FFZE experiments in
microgravity conditions inside orbiting space craft have been performed [193-
1951. Successful separation of macromolecules (DNA, proteins) and bioparticles
was achieved in these experiments, but the improvement of the separation effi-
ciency and capacity was less than originally expected.
Miniaturized FFZE device integrated onto a silicon chip has been devel-
oped for continuous microanalysis of biomolecules [196].
The velocities in Eq. (15) can be expressed as the ratio of effective length of the
capillary, l,, and the corresponding migration times, resulting migration time, t,,
electrophoretic migration time, t,,, and electroosmotic migration time, t,,, respec-
tively:
'ef
-
Jr
Veo Vep
CHAMBER
I \ \
\
1 1 \
1 1 \
I \ \
I \ \
\
I \
From the combination of Eqs. (15) and (16), the following relation can be ob-
tained for the electrophoretic velocity in the capillary:
Equation (17) allows one to obtain the electrophoretic velocity from the
data experimentally available from CZE analysis: t, is the resulting migration
Separations of Peptides by CZE and FFZE 73
From Eqs. (19) and (20) the following relation for electrophoretic velocity in the
chamber, v , , ~ can
, be derived:
Using relation (22) the electrophoretic velocity of the charged analyte in the
chamber, v,,,, can be calculated from the experimentally available data, namely,
from the resulting migration distance of the charged analyte. d,, from the mi-
gration distance of the EOF marker, d,,, and from the flow-through time of
BGE, tf.
The EOF velocity in the chamber, v,,,~,can be calculated from the migration
distance of EOF marker, d,,, and from the flow-through time of BGE, t,.
For the description of the correlation between CZE and FFZE it is advantageous
to express the ratio of electrophoretic velocities in the chamber and in the capil-
lary, qe,, and the ratio of EOF velocities in the chamber and in the capillary, q,,.
From Eqs. (17) and (22) the ratio q,, can be expressed as
and from Eqs. (18) and (23) the ratio q,, can be expressed as
Provided that the adsorption of the sample components to the walls of the
separation compartments (both capillary and flow-through chamber) can be ne-
glected it is reasonable to assume that q,, is approximately constant for different
charged components separated by CZE and FFZE under the same separation
conditions. This is a realistic assumption because it means that if the electropho-
retic velocity of component A in FFZE is q times higher than in CZE, then the
electrophoretic velocity of component B, analyzed under the same conditions as
A, will be also q times higher in FFZE than in CZE. Consequently, as follows
from Eq. (24), coefficient q,, (determined for standard component S) can be used
to predict the electrophoretic velocities of sample components (A, B, C) in FFZE,
if their electrophoretic velocities in CZE were determined under the same condi-
tions as the electrophoretic velocity of standard S. A similar conclusion can be
applied for the ratio of EOF velocities in CZE and FFZE, i.e., this ratio can be
considered as a constant if the conditions of CZE and FFZE are the same as they
were in the experiment when EOF velocity was determined.
Knowing the values of ratios, qepand q,, allows us to predict the migration
velocities and migration distances of analytes in FFZE from the data obtained
by their CZE analysis. This fact alone is the core of the procedure for conversion
of analytical CZE separations to preparative FFZE separations.
Similarly, the ratio of EOF velocities in the chamber and in the capillary, q,,, is
obtained:
(6) The resulting migration distances can also be calculated for the other
components of the sample (admixtures A l , A2, . . . and neutral component(s)
N) and their separability in FFZE can be estimated. If the distances of the compo-
nents of interest at the outlet side of the chamber are sufficient for their separation,
then the FFZE separation can be performed under the same conditions as those
used for separation of standard components.
If the predicted distances are not sufficient for the separation of sample
components of interest, then the separation conditions of FFZE, namely, clamp
voltage and/or flow-through time, have to be further optimized. If the predicted
distances are too small and the separation of sample components is not achieved,
then the voltage and/or flow-through time should be increased. If the predicted
distances for the sample components are too long and there is a danger ,that the
fastest component will reach the close vicinity of the ion exchange membrane
separating the separation chamber from the electrode compartment (see Fig. 1 I),
where this component can be damaged or lost because of concentration, pH, and
conductivity nonhomogeneities occurring in this region, then the clamp voltage
and/or flow-through time must be decreased. The migrated distance is approxi-
mately directly proportional to the voltage and to the flow-through time, i.e., p%
prolongation of migration time and r% increasing of voltage will result in (p +
r)% prolongation of migrated distance. Following this rule suitable separation
conditions can be selected.
The above-described procedure allows one to develop suitable separation
conditions in the more economical and faster microscale by CZE. Only then
can the optimized conditions be converted to the preparative scale realized by
continuous FFZE separation.
fraction number
Figure 14 FFZE separation of standard mixture of diglycine (15 mglml), peak 1, tri-
glycine (15 mglml), peak 2, and phenol (5 mglml), peak 3. BGE: 0.5 mol1L acetic acid,
flow-through time 31 min, sample flow rate 1.5 mllh, voltage 3000 V, current 120 mA,
temperature -3°C; Peak height = peak height of the analytes obtained by CZE analyses
of the aliquots of the FFZE fractions; 1',sample inlet position.
of diglycine, triglycine, and phenol was separated (clamp voltage 3000 V, flow-
through time 31 min), were applied also to the separation of the components of
the crude product of [~-Tle~.~]dalargin. The lyophilyzate of the crude synthetic
product (190 mg) was dissolved in 5 ml of BGE (0.5 mol/L acetic acid), centri-
fuged, and applied to FFZE separation. The record of FFZE separation (off-line
W absorption measurement of FFZE fractions) is shown in Fig. 15. Comparing
Fig. 13 and 15, the ''qualitative" similarity of CZE and FFZE separation profiles
can be observed. In addition to the main dalargin product (peak D), the fastest
component F, slowest component S, and noncharged component N can be found
on both records. The differences in relative peak heights are caused by the differ-
ent detection wavelengths in CZE (206 nm) and in FFZE (280 nm). Obviously,
a better separation of sample components is achieved in CZE than in FFZE. This
Table 3 Migration Times, Migration Distances, Electrophoretic and Electroosmotic
Velocities of Standard Components Separated by CZE and FFZE
;' Resulting migration time, t, (migration distance, d,) of phenol is equal to migration time, t,,, (migra-
tion distance, d,,,) of electroneutral EOF marker.
t,, resulting migration time; v,,,,, electrophoretic velocity in CZE; v,,,,, electroosmotic velocity in
CZE; d,, resulting migration distance; v,,,~, electrophoretic velocity in FFZE; v,,,,, electroosmotic
velocity in FFZE; q,, (q,,), ratio of electrophoretic (electroosmotic) velocities in FFZE and CZE.
is quite understandable when taking into account the differences in the experi-
mental conditions of these two methods: more efficient anticonvective stabiliza-
tion and Joule heat transfer in the capillary (i.d. 0.056 mm, wall thickness 0.07
mm) than in the flow-through chamber (rectangular gap 0.5 mm between two
glass plates of the thickness 4 mm), about one order lower separation time and
sample concent~ationin CZE than in FFZE, absence of hydrodynamic flow in
CZE, and a relatively large width of the collected f~action(10.4 mm) in FFZE.
CZE FFZE
fraction number
Figure 16 CZE analysis of fraction 26 (a) and 36 (b) of FFZE separation of [~-Tle'.~]da-
largin preparation presented in Fig. 15. Aliquot of FFZE fraction was directly applied to
CZE analysis. Experimental conditions the same as in Fig. 13. A,,,, absorption at 206 nm;
t, migration time
Separations of Peptides by CZE and FFZE 83
was synthesized by the solid phase method and purified by RP-HPLC 11981. CZE
analysis of the HPLC-purified preparation revealed three admixtures of the main
synthetic product (see Fig. 17a). The admixture indicated in this figure as I was
later on identified as Cys-acetamidomethyl-derivative of octapeptide monomer
of the dimer VI. In HPLC this octapeptide derivative was co-eluted with the
heterodimer VI but in CZE these two peptide species were well separated. For
that reason it was decided to use preparative FFZE for purification of heterodimer
VI. The CZE analysis of FFZE-purified preparation of heterodimer VI confirmed
the high purity of this preparation (see Fig. 17b) and the suitability of FFZE for
separation of the admixtures not removed by HPLC. This is a good example of
complementarity of CZE (FFZE) and HPLC for peptide analysis and preparation
resulting from different separation principles of these methods.
A combination of CZE and FFZE has been applied to analysis and prepara-
tion of some other biopeptides synthesized at our institute, such as growth
hormone-releasing peptide (GHRP) and its analogs and fragments 1152,1991,
derivatives of luteinizing hormone-releasing hormone (LHRH) [200], and B23-
30-octapeptide fragments of insulin [201].
Combined application of CZE and FFZE represents a new systematic ap-
proach to analysis and preparation of synthetic biopeptides. First, CZE is used
for analysis of the peptide preparation and suitable conditions for its analysis
are developed in a microscale minimizing time and material expenses for the
development of the optimized separation conditions. Then, based on the theory
of the correlation between CZE and FFZE, the optimized conditions of CZE
separation are converted to FFZE separation. FFZE separation of peptide prepara-
Figure 17 CZE analysis of eicosapeptide disulfide fragment of human cathepsin D prep-
aration purified by HPLC (a) and by FFZE (b). Experimental conditions the same as in
Fig. 13. VI, main synthetic product-eicosapeptide disulfide fragment of human cathepsin
D. I, octapeptide fragment of eicosapeptide VI. For peptide sequences and other details,
see the text.
tion provides an efficient tool for preparative purification of peptide sample with
the preparative capacity of tens to hundreds of milligrams per hour. The advan-
tage of this method is that it works continuously, in a carrierless medium, under
mild conditions in which the biological activity of separated peptides is retained
and the loss of material minimized. The purity of peptide fractions separated by
FFZE can then be checked by CZE and/or another method. A combination of
more methods based on different separation principles should be preferred be-
cause that way more complete information about peptide purity and identity can
be obtained.
Separations of Peptides by CZE and FFZE
ACKNOWLEDGMENTS
The results presented in this chapter were obtained with the financial support of
the Grant Agency of the Academy of Sciences of the Czech Republic, grant no.
4551 1, and of the Grant Agency of the Czech Republic, grant nos. 20319310718,
20319410698, and 203196lK128, which is greatfully acknowledged. The author
thanks his coworkers from the Laboratory of Electromigration Methods of the
Institute of Organic Chemistry and Biochemistry-Dr. Z.Pmsik, Dr. P. S5zelovi,
Mrs. V. LiBkov6, Mr. J. &gp6nek, and Mgr. M. Machov6-for their cooperation
and assistance in the experimental work and for their help in the preparation of
this chapter.
ABBREVIATIONS
REFERENCES
Adamson NJ, Reynolds EC. Rules relating electrophoretic mobility, charge and
molecular size of peptides and proteins. J Chromatogr B 1997;699:133-147.
Cifuentes A, Poppe H. Behavior of peptides in capillary electrophoresis: effect of
peptide charge, mass and structure. Electrophoresis 1997;18:2362-2376.
Gao JM, Whitesides GM. Using protein charge ladders to estimate the effective
charges and molecular weights of proteins in solution. Anal Chem 1997;69:575-
580.
Chae KS, Lenhoff AM. Computation of the electrophoretic mobility of proteins.
Biophys J 1995;68:ll20-1127.
Stauff J, Jaenicke R. Physikalische chemie der losungen. In: Rauen HM, ed. Bio-
chemisches Taschenbuch. Berlin: Springer-Verlag, 1964:37- 121.
Prusik Z. Peptides and structural analysis of proteins. In: Deyl Z, ed. Electrophore-
sis: a survey of techniques and applications. Pt. B: Applications. Amsterdam: Elsev-
ier, 1983:81-107.
Rickard EC, Towns JK. Applications of capillary zone electrophoresis to peptide
mapping. Meth Enzymol 1996;271 :237-264.
KaSiEka V, Prusik Z. Isotachophoretic analysis of peptides. Selection of electrolyte
systems and determination of purity. J Chromatogr 1989;470:209-22 1.
KaSiEka V. Analysis and preparation of peptides and proteins by isotachophoresis.
PhD thesis (in Czech). Prague: Czechoslovak Academy of Science, 1985.
Mosher RA, Gebauer P, Thormann W. Computer simulation and experimental vali-
dation of the electrophoretic behavior of proteins 3. Use of titration data predicted
by the protein's amino acid composition. J Chromatogr l993;638: 15.5- 164.
Westermeier R. Electrophoresis in Practice: A Guide to Methods and Applications
of DNA and Protein Separations. Weinheim: VCH, 1997.
Nath S, Schutte H, Hustedt H, Deckwer WD. Correlation of migration behavior in
free-flow zone electrophoresis and electrophoretic titration curve. Electrophoresis
1990;11:612-616.
Hirokawa T, Kiso Y, GaS B, Zuskova I, Vacik J. Simulated quantitative and qualita-
tive isotachophoretic indices of 73 amino acids and peptides in the pH range 6.4-
10. J Chromatogr 1993;628:283-308.
Wehr T, Zhu M, Rodriguez-Diaz R. Capillary isoelectric focusing. Meth Enzymol
1996;270:358-374.
Righetti PG, Gelfi C, Chiari M. Isoelectric focusing in capillaries. In: Righetti PC,
ed. Capillary Electrophoresis in Analytical Biotechnology. Boca Raton: CRC Press,
1996:509-539.
Righetti PG, Gelfi C. Isoelectric focusing in capillaries and slab gels: a comparison.
J Cap Elec 1994;1:27-35.
Yao YJ, Khoo KS, Chung MCM, Li SFY. Determination of isoelectric points of
acidic and basic proteins by capillary electrophroesis. Chromatogr A 1994;680:
431-435.
Sillero A, Ribeiro JM. Isoelectric points of proteins: theoretical determination. Anal
Biochem 1989;179:319-325.
Henriksson G, Englund AN, Johansson G, Lundahl P. Calculation of the isoelectric
points of native proteins with spreading of pKa values. Electrophoresis 1995;16:
1377-1380.
Grossman PD. Background concepts. In: Grossman PD, Colburn JC, eds. Capillary
Electrophoresis. San Diego: Academic Press, 1992:3-43.
Botek P. Analytical capillary electrophoresis. In: Churitek J, ed. Advanced Instru-
mental Methods of Chemical Analysis. Praha: Academia, 1993:97- 14 1.
Foret F, Kfivinkovi L, BoEek P. Capillary Zone Electrophoresis. Weinheim: Ver-
lag Chemie, 1993.
Moring SE. Buffers, electrolytes, and additives for capillary electrophoresis. In:
Righetti PC, ed. Capillary Electrophoresis in Analytical Biotechnology. Boca Ra-
ton: CRC Press, 1996:37-60.
Issaq HJ, Janini GM, Chan KC, Elrassi Z. Approaches for the optimization of exper-
imental parameters in capillary zone electrophoresis. Adv Chromatogr 1995;35:
101-169.
Hannig K, Heidrich HK. Free-Flow Electrophoresis. Darmstadt: GIT, 1989.
Reijenga JC, Verheggen TPEM, Martens JHPA, Everaerts FM. Buffer capacity,
ionic strength and heat dissipation in capillary electrophoresis. J Chromatogr A
l996;744: 147- 153.
Nielsen RG, Rickard EC. Method optimization in CZE analysis of hGH tryptic
digest fragments. J Chromatogr 1990;5 16:99-114.
Swedberg SA. Use of non-ionic and zwitterionic surfacants to entrance selectivity
in HPCE. J Chromatogr 1990;503:449-452.
Greve KF, Nashabeh W, Karger BL. Use of zwitterionic detergents for the separa-
tion of closely related peptides by capillary electrophoresis. J Chromatogr A 1994;
680: 15-24.
Matsubara N, Koezuka K, Terabe S. Separation of eleven angiotensin I1 analogs by
capillary electrophoresis with a nonionic surfactant in acidic media. Electrophoresis
1995;16:580-583.
Liu J, Cobb KA, Novotny M. Capillary electrophoretic separations of peptides us-
ing micelle-forming compounds and cyclodextrins as additives. J Chromatogr 1990;
519:189-197.
Kornfelt T, Vinther A, Okafo GN, Camilleri P. Improved peptide mapping using
phytic acid as ion-pairing buffer additive in capillary electrophoresis. J Chromatogr
A 1996;726:223-228.
Matsubara N, Terabe S. Micellar electrokinetic chromatography. Meth Enzymol
l996;270:3 19-34 1.
Matsubara N, Terabe S. Micellar electrokinetic chromatography in the analysis of
amino acids and peptides. In: Righetti PC, ed. Capillary Electrophoresis in Analyti-
cal Biotechnology. Boca Raton: CRC Press, 1996:1.55- 182.
Vindevogel J, Sandra P. Introduction to Micellar Electrokinetic Chromatography.
Heidelberg: Huthig, 1992.
Yashima T, Tsuchiya A, Morita 0. Separation of closely related large peptides by
micellar electrokinetic chromatography with organic modifiers. Anal Chem 1992;
64:298 1-2984.
Ye B, Hadjmohammadi M, Khaledi MG. Selectivity control in micellar electroki-
netic chromatography of small peptides using mixed fluorocarbon hydrocarbon an-
ionic surfactants. J Chromatogr A 1995;692:29 1-300.
Castagnola M. Cassiano L, Messana I, Paci M, Rossetti DV, Giardina B. Effect
7
Voegel PD, Baldwin RP. Electrochemical detection with copper electrodes in liquid
chromatography and capillary electrophoresis. Int Lab 1996;26: 16A- 16M.
O'Shea TJ, Greenhagen RD, Lunte SM, Lunte CE, Smyth MR, Radzik DM, Wata-
nabe N. Capillary electrophoresis with electrochemical detection employing an on-
column Nafion joint. J Chrornatogr l992;593:305-3 12.
Ye JN, Baldwin RP. Determination of amino acids and peptides by capillary elec-
trophoresis and electrochemical detection at a copper electrode. Anal Chem 1994;
66:2669-2674.
O'Shea TJ, Lunte SM. Selective detection of free thiols by capillary electrophoresis
electrochemistry using a gold mercury amalgam microelectrode. Anal Chem 1993;
65:247-250.
Weber PL, Kornfelt T, Klausen NK, Lunte SM. Characterization of glycopeptides
from recombinant coagulation factor VIIa by high-performance liquid chromatog-
raphy and capillary zone electrophoresis using ultraviolet and pulsed electrochemi-
cal detection. Anal Biochem 1995;225: 135- 142.
Huang XH, Zare RN, Sloss S, Ewing AG. End-column detection for capillary zone
electrophoresis. Anal Chem 1991;63:189- 192.
Muller D, Jelinek I, Opekar F, Stulik K. A conductometric detector for capillary
separations. ~lectroanal~sis 1996:8:722-725.
Huang X, Pang TKJ, Gordon MJ, Zare RN. On-column conductivity detector for
capillary zone electrophoresis. Anal Chem 1987;59:2747-2749.
Dasgupta PK, Bao LY. Suppressed conductometric capillary electrophoresis sepa-
ration systems. Anal Chem 1993;65:1003- 101l.
Kar S, Dasgupta PK, Liu HH, Hwang H. Computer-interfaced bipolar pulse con-
ductivity detector for capillary systems. Anal Chem 1994;66:2537-2543.
Jandik P, Bonn G. Capillary Electrophoresis of Small Molecules and Ions. Cam-
bridge: VCH, 1993.
Huang XH, Luckey JA, Gordon MJ, Zare RN. Quantitative analysis of low molecu-
lar weight carboxylic acids by capillary zone electrophoresislconductivity detec-
tion. Anal Chem 1989;61:766-770.
Grossman PD, Wilson KJ, Petrie G, Lauer HH. Effect of buffer pH and peptide
composition on the selectivity of peptide separations by capillary zone electropho-
resis. Anal Biochem 1988;l73:265-270.
Colburn JC. Capillary electrophoresis separations of peptides: practical aspects and
applications. In: Grossman PD, Colburn JC, eds. Capillary Electrophoresis: Theory
and Practice. San Diego: Academic Press, 1992:237-27 1.
Nashabeh W, Greve KF, Kirby D, Foret F, Karger BL, Reifsnyder DH,
Builder SE. Incorporation of hydrophobic selectivity in capillary electrophoresis:
analysis of recombinant insulin-like growth factor I variants. Anal Chem 1994;66:
2148-2154.
Grossman PD, Colburn JC, Lauer HH, Nielsen RG, Riggin RM, Sittampalam GS,
Rickard EC. Application of free-solution capillary electrophoresis to the
analytical scale separation of proteins and peptides. Anal Chem 1989;61: 1186- 1194.
Nielsen RG, Sittampalam GS, Rickard EC. Capillary zone electrophoresis of insulin
and growth hormone. Anal Biochem 1989;177:20-26.
Vinther A, Holm A, Hoeejensen T, Jespersen AM, Klausen NK, Christensen T,
Sorensen HH. Synthesis of stereoisomers and isoforms of a tryptic heptapeptide
fragment of human growth hormone and analysis by reverse-phase HPLC and capil-
lary electrophoresis. Eur J Biochem 1996;235:304-309.
Wan H, Blomberg LC. Enantiomeric separation by capillary electrophoresis of di-
and tripeptides derivatized with 9-fluorenylmethyl chloroformate using vancomycin
as chiral selector. J Microcolumn Sep 1996;8:339-344.
Wan H, Blomberg LC. Enantiomeric and diastereomeric separation of di- and tri-
peptides by capillary electrophoresis. J Chromatogr A 1997;758:303-311.
Sanger-van de Griend CE, Groningsson K, Arvidsson T. Enantiomeric separation
of a tetrapeptide with cyclodextrin: extension of the model for chiral capillary elec-
trophoresis by complex formation of one enantiomer molecule with more than one
chiral selector molecules. J Chromatogr A 1997;782:271-279.
Moore AW, Jorgenson JW. Resolution of cis and trans isomers of peptides con-
taining proline using capillary zone electrophoresis. Anal Chem 1995;67:3464-3475.
Ma S, Kalman F, Kalman A, Thunecke F, Howath C. Capillary zone electrophore-
sis at subzero temperatures: separation of the cis and trans conformers of small
peptides. J Chromatogr A 1995;716: 167- 182.
Survay MA, Goodall DM, Wren SAC, Rowe RC. Oligoglycines and oligoalanines
as tests for modelling mobility of peptides in capillary electrophoresis. J Chro-
matogr 1993;636:81-86.
KaSiEka V, Prusik Z, Mudra P, StEtpinek J. Capillary electrophoresis device with
double UV detection and its application to the determination of effective mobilities
of peptides. J Chromatogr A 1995;709:3 1-38.
Gao JM, Gomez FA, Harter R, Whitesides GM. Determination of the effective
charge of a protein in solution by capillary electrophoresis. Proc Natl Acad Sci
USA l994;9l: 12027- 12030.
Zhang Y, Lee HK, Li SFY. Separation of myoglobin molecular mass markers using
non-gel sieving capillary electrophoresis. J Chromatogr A 1996;744:249-257.
Walbroehl Y, Jorgenson JW. Capillary zone electrophoresis for the determination
of electrophoretic mobilities and diffusion coefficients of proteins. J Microcolumn
Sep 1989;1:41-45.
Rao JH, Whitesides GM. Tight binding of a dimeric derivative of vancomycin with
dimeric L-Lys-D-Ala-D-Ala. J Am Chem Soc 1997;119:10286-10290.
Rao JH, Colton IJ, Whitesides GM. Using capillary electrophoresis to study the
electrostatic interactions involved in the association of ~ - A l a - ~ - Awith
l a vancomy-
cin. J Am Chem Soc 1997;119:9336-9340.
Letourneau DL, Allen NE. Use of capillary electrophoresis to measure dimerization
of glycopeptide antibiotics. Anal Biochem 1997;246:62-66.
Kiivinkovi L, BoCek P. Synergism of capillary isotachophoresis and capillary zone
electrophoresis. J Chromatogr B 1997;689: 13-34.
Goodall DM, Williams SJ, Lloyd DK. Quantitative aspects of capillary electropho-
resis. Trends Anal Chem 1991;10:272-279.
Prusik Z, KaSitka V, Mudra P, StEtpinek J, SmCkal 0 , Hlavieek J. Correlation
of capillary zone electrophoresis with continuous free-flow zone electrophoresis:
application to the analysis and purification of synthetic growth hormone releasing
peptide. Electrophoresis 1990;11:932-936.
Novotny MV, Cobb KA, Liu JP. Recent advances in capillary electrophoresis of
proteins, peptides and amino acids. Electrophoresis 1990;11:735-749.
Separations of Peptides by CZE and FFZE 95
Righetti PG, Gelfi C, Perego M, Stoyanov AV, Bossi A. Capillary zone electropho-
resis of oligonucleotides and peptides in isoelectric buffers: theory and methodol-
ogy. Electrophoresis 1997;18:2145-2153.
Kuhr WG, Monnig CA. Capillary electrophoresis. Anal Chem 1992;64:R389-R407.
Monnig CA, Kennedy RT. Capillary electrophoresis. Anal Chem 1994;66:R280-R3 14.
Stclaire RL. Capillary electrophoresis. Anal Chem 1996;68:R569-R586.
KaSiEka V, Prusik Z. Capillary zone electrophoresis and isotachophoresis of biolog-
ically active peptides. In: Parvez H, Caudy P, Parvez S, Roland-Gosselin P, eds.
Capillary Electrophoresis in Biotechnology and Environmental Analysis. Utrecht:
VSP, 1997:173-197.
Castagnola M, Messana I, Rossetti DV. Capillary zone electrophoresis for the anal-
ysis of peptides. In: Righetti PG, ed. Capillary Electrophoresis in Analytical Bio-
technology. Boca Raton: CRC Press, 1996:239-275.
Li SFY. Capillary Electrophoresis. Amsterdam: Elsevier, 1992.
Rose DJ, Jorgenson JW. Fraction collector for capillary zone electrophoresis. J
Chromatogr 1988;438:23-34.
Lee HG, Desiderio DM. Preparative capillary zone electrophoresis of synthetic pep-
tides: conversion of an autosarnpler into a fraction collector. J Chromatogr A 1994;
686:309-3 17.
Smith AJ. Analytical and micropreparative capillary electrophoresis of peptides.
In: Smith BJ, ed. Protein Sequencing Protocols. Totowa: Humana Press, 1997:91-99.
Boss HJ, Rohde MF, Rush RS. Multiple peptide fraction collection by capillary
electrophoresis with reinjection analysis. Peptide Res 1996;9:203-209.
Herold M, Wu SL. Automated peptide fraction collection in CE. LC GC-Mag Sepa-
ration Sci 1994;12:531-533.
Grimm R, Herold M. Micropreparative single run fraction collection of peptides
separated by CZE for protein sequencing. J Cap Elec 1994;1:79-82.
Lee HG, Tseng JL, Becklin RR, Desiderio DM. Preparative and analytical capillary
zone electrophoresis analysis of native endorphins and enkephalins extracted from
the bovine pituitary: mass spectrometric confirmation of the molecular mass of
leucine enkephalin. Anal Biochem 1995;229:188-197.
Boss HJ, Rohde MF, Rush RS. Multiple sequential fraction collection of peptides
and glycopeptides by high-performance capillary electrophoresis. Anal Biochem
1995;230:123-129.
Bergman AC, Bergman T. Micropreparation of peptides by capillary electrophoresis
for matrix assisted laser desorption mass spectrometry. FEBS Lett 1996:397:45-49.
Bergman T, Agerberth B, Jornvall H. Direct analysis of peptides and amino acids
from capillary electrophoresis. FEBS Lett 1991;283:100- 103.
Wallingford RA, Ewing AG. Amperometric detection of catechols in capillary zone
electrophoresis with normal and micellar solutions. Anal Chem 1988;60:258-263.
Huang X, Zare RN. Continuous sample collection in capillary zone electrophoresis
by coupling the outlet of a capillary to a moving surface. J Chromatogr 19905 16:
185-189.
Fujimoto C, Fujikawa T, Jinno K. Sample collection by a capillary zone electropho-
retic system with an on-column fracture. J High Res Chromatogr 1992;15:201-203.
Cheng YF, Fuchs M, Andrews D, Carson W. Membrane fraction collection for
capillary electrophoresis. J Chromatogr 1992;608:109-116.
96 Kasicka
I. INTRODUCTION
plex assuming that the conditions necessary for the formation of this complex
exist. The binding sites of the immobilized substances must be sterically accessi-
ble even after their coupling to the solid support, and they must not be deformed
by immobilization.
A solid support with a covalently bound affinant is used as the stationary
phase in a chromatographic column. When a crude mixture containing the biolog-
ically active products to be isolated is passed through the column of affinity
adsorbent, then all of the compounds that under given experimental conditions
have no complementary binding site for the immobilized affinity ligand will pass
through unretarded. By contrast, products showing affinity for the insoluble affi-
nant are adsorbed onto the column. They can be released by elution with a solu-
tion of a soluble affinity ligand or by changing of the solvent composition by
so-called deforming buffers (e.g., by a change in pH, ionic strength, temperature,
etc.).
Assuming that a single enzyme of the crude proteins has an affinity for the
specific adsorbent, the equilibrium between the attached affinity ligand L and the
isolated enzyme E is given by the equation:
Dissociation constant:
(1) --
1.5 -
C
a --- -
I
A 280 I
I
low molecular weight inhibitors. Unlike the naturally occumng inhibitors, which
could undergo denaturation because of their protein character, the synthetic low
molecular weight inhibitors are completely stable. The capacity of specific adsor-
bents prepared with these inhibitors can be regenerated virtually without limit,
if a stable solid support and stable bonds between the support and the amino
groups of peptide inhibitors are used.
According to the specificity we distinguish (1) highly specific and (2)
group-specific matrices. The practical utility of specific sorbents increases if, in-
stead of the narrowly specific ligands, a so-called general ligand is used for their
preparation. As is implied by the name, a group-specific matrix prepared in this
Turkova
manner displays an affinity for a more or less large group of biological macromol-
ecules. For example, the enzymes related to the metabolism of aspartic acid show
group-specific adsorption affinity to N-(w-aminohexy1)-L-aspartic acid-Sepha-
rose. Asparaginase, aspartase, aspartate-P-decarboxylase, and asparaginase modi-
fied with tetranitromethane [9] could all be sorbed onto this immobilized affinant.
B. Enzymes
The combination of several complementary binding sites on the surface of the
enzyme molecule permits the formation of a relatively large number of biospec-
Bioaffinity Chromatography 105
ific complexes to be achieved; these complexes can be utilized for efficient isola-
tion as well as for oriented immobilization. Figure 3 shows the surface of an
enzyme molecule covered with several complementary binding sites [lo]. Such
an enzyme could be, for instance, carboxypeptidase Y (CPY) containing the com-
plementary binding site for glycylglycyl-p-aminobenzylsuccinicacid, a specific
inhibitor, which after immobilization was used for the isolation of CPY from
autolyzates of various kinds of yeast. The active sile of the enzyme also contains
a free SH- group and therei'ore can be adsorbed to mercury-Spheron. CPY is a
glycoprotein whose carbohydrate moiety specifically interacts with concanavalin
A, a lectin. If CPY was adsorbed on immobilized concanavalin A followed by
crosslinking with glutaraldehyde, the bound enzyme retained 96% of the native
catalytic activity. The antigenic sites of the enzyme can be determined by investi-
gation of the antigenic structures of the peptide chain in experiments with specific
antibodies to this enzyme. Enzymes whose coenzymes are nucleotides can form
biospecific complexes with nucleotides. Enzymes form complexes with substrates
and their analogs, allosteric effectors, metal ions, etc. The type of complex formed
determines the mode of their action in chemical processes that take place in the
living cell.
The isolation of enzymes by use of immobilized inhibitors is shown in Fig.
I and 2. The catalytic activity of many enzymes depends on the presence of
coenzymes or cofactors. The coenzymes nicotinamide adenine dinucleotide and
0 8 16 24 30
ELUTION VOLUME, m l
NEUTRAL 0
SULFONATE OR
PHOSPHATE 0
affinity labeling, x-ray diffraction, and other techniques have been utilized to
demonstrate the specificity of dye binding to the active sites of proteins.
IgC Fc RECEPTORS
B ANTIGEN RECEPTORS
PROTEIN A
RABBIT IgG ANTIBODIES
\
Fc PORTION
effects of various column parameters, Schott [22] expressed all elution volumes
relative to the elution volume of alanine, which displays no measurable retention
on these columns. The relative elution ratio (V,) is thus obtained as the ratio of !
the elution volume found for the investigated peptide to that for alanine (V, = j
V,,,/V,,,). The peptide-oligonucleotide interaction is then evaluated on the basis 1
1
of the difference in the relative elution volumes obtained by chromatography on
both columns. i
A solid phase triple-helix-mediated affinity capture method was described 1
i.
for the purification of single-stranded M13 DNA for use as template in fluores-
cence-based DNA sequencing reactions by Johnson et al. [23]. In this method,
a biotinylated polypyrimidine oligonucleotide "loop" bound to streptavidin-
1
coated magnetic beads was used to selectively capture single-stranded M13 DNA
from high-titer phage supernatant through the formation of a cooperative and a
/1
polypurine site previously cloned into the M13 vector. I
I
Bioaffinity Chromatography 113
-CO-NN-CO-WBr ---,Mxleophiles
However, a spacer arm should be used when proteins are modified by biotin.
Detailed information on biotin-binding proteins, the preparation of biotin, avidin,
and streptavidin derivatives, assays for avidin and biotin, and applications is
given in Volume 184 of Methods in Enzymology titled Avidin-Biotin Technology,
edited by Wilchek and Bayer [31].
The use of an avidin-biotin complex in bioaffinity chromatography was
the isolation of avidin from egg white on a biocytin-Sepharose column [32]. The
conditions required for its dissociation were extremely drastic requiring elution
by 6 M guanidinium hydrochloride, pH 1.5. In order to decrease the strong affinity
of avidin for biotin, Finn and Hofmann [29] used immobilized succinylated avidin
for the isolation of hormone receptor. Soluble receptor (R) is percolated through
a bioaffinity column to form the complex shown in the center of Fig. 9. The
column is then exhaustively washed to remove contaminating materials. Alterna-
tively, the biotinylated hormone ligand (B-H) can be added to a solution of solubi-
lized receptor to form a soluble complex BHR. Applying a solution containing
this complex to a column of immobilized succinylated avidin (SA) will result in
the formation of the same complex (Fig. 9B). Both of these schemes have been
used for the isolation of insulin receptors from human placenta. Removal of active
insulin receptor can be achieved by eluting the column with acetate buffer, pH
5 containing either 1 M NaCl or biotin.
116 Turkova
Despite the fact Lhal streptavidin is currently about 100 times more expen-
sive than avidin, its use is sometimes justified since immobilized streplavidin
exhibits less nonspecific binding. Avidin is highly positively charged at neutral
pH, with an isoelectric point above 10. Consequently, it binds negatively charged
molecules such as nucleic acids, acid proteins, or phospholipids in a nonspecific
manner. This can result i n nonspecific staining of, for example, the nucleus and
cell membranes. Avidjn is also a glycoprotein and therefore interacts with other
biological molecules such as lectins or other sugar-binding materials via its carbo-
hydrate moiety. The advantage of using streptavidin lies in the fact that it is a
neutral, nonglycosylated protein (pl lower than 7). To decrease its positive
charge, the lysines of avidin can be derivatized by succinylation, acetylation, etc.
A variety of avidin derivatives with average pl values of 7 or lower are now
commercially available. However, the ren~ovalof the carbohydrate residue from
avidin is much more difficult.
One method that can be generalized to link virtually any DNA substrate
by its 3' or 5' end to a chromatography matrix via streptavidin-biotin linkage has
been described by Fishel et al. [33]. Biotin-slreptavidin affinity selection as a
valuable tool permitting the analysis of the RNA components of splicing com-
plexes assembled on a wide variety of pre-mRNA substrates has been reviewed
by Grabowski [34]. A review of the isolation of cell surface glycoproteins by
the use of biotinylated lectin was published by Cook and Buckie [35]. They
demonstrated the use of immobilized streptavidin to obviate dissociation of com-
Bioaffinity Chromatography 117
to purity, functional activity, and viability of separated cells. The most frequently
employed affinants for the isolations of viruses are bound antibodies. An example
is the isolation of Alleutian disease virus from chronically infected mink [41] by
use of Sepharose with coupled IgG. In some cases rather than immobilized lec-
tins, antibodies or Protein A, other specific and reversible cell surface receptor-
ligand interactions can be successfully utilized. As an example one may mention
the isolation of acetylcholine receptor-bearing neuronal cells from chick embryo
sympathetic ganglia by the use of snake venom a-bungarotoxin immobilized on
Sepharose 6MB macrobeads [42]. Information on the use of mentioned and many
other affinants is given in Ref. 7.
of the described requirements does not exist. When a certain affinity ligand is
immobilized an individual solid support and method of coupling must be selected
with respect to its future use.
Application of bioaffinity chromatography resulting from a changeover
from soft gel supports to small rigid particles used in high-performance liquid
bioaffinity chromatography (HPLBAC) has been reviewed by Ohlson et al. [44].
A comparison of HPLBAC with soft gel bioaffinity chromatography (BAC) is
shown in Fig. 10. Mechanically stable, rigid particles, with small and uniform
sizes, provide high flow rates with good mass transfer characteristics, giving over-
all high operational adsorption capacity. Much more favorable mass transport
and adsorption/desorption kinetic behavior with nonporous support has been
demonstrated by Anspach et al. [45]. The elution behavior of proteins on a nonpo-
rous silica-based adsorbent (with an average diameter of 1.4 pm) was investigated
both theoretically and experimentally using human immunoglobulin G and immo-
bilized protein A as the affinity pair by Lee and Chuang [17]. The desorption
rate constant and equilibrium association constant under elution conditions were
found to decrease elution time and improve the shape of the elution peak. How-
ever, the adsorption rate in column chromatography is limited by either slow
intraparticle diffusion for large beads or low axial velocities and high-pressure
0
0.001 m m h a r d -nonporous
hours m i nut es
B. Biopolymers
Water molecules are essential for the structure and function of biologically active
compounds. BAC is therefore generally performed in the aqueous phase and hy-
drophilic biopolymers are usually employed as solid supports, chiefly natural
polysaccharides, such as agarose, dextran, cellulose, and, to a lesser extent, starch.
The modification necessary for the BAC can be carried out in a relatively simply
via their OH groups.
2. Dextran Gels
Dextran is a branched-chain glucose polysaccharide produced in solutions con-
taining sugar by various strains of Leuconostoc mesenteroides. Soluble dextran,
prepared by fractional precipitation with ethanol of partially hydrolyzed crude
dextran, contains more than 90% of a-1,6-glucosidic linkages with 1,2-, 1,3-,
and 1,4-glucoside branching. When crosslinked with 1-chloro-2,3-epoxypropane
in alkaline solution, dextran becomes suitable for chromatography.
The most important producer of dextran gels, supplied under the trade name
Sephadex, is Pharmacia Biotechnology. The gels are very stable to chemical at-
tack. The glucosidic bond is sensitive to hydrolysis at low pH, although it is stable
for 6 months in 0.02 M hydrochloric acid, or for 1-2 h in 0.1 M hydrochloric acid
or 88% formic acid. Aldehyde or carboxyl groups are formed under the effect
of oxidizing agents. Dextran gels can withstand heating in an autoclave at 110°C
(in solution) for 40 min, or at 120°C when dry. Drying and swelling is reversible.
The gels swell to some extent even in ethanol, ethylene glycol, formamide, N,N-
dimethylformamide, and dimethylsulfoxide.
A molecular sieve produced by covalent crosslinking of allyldextran with
N,Nf-methylenebisacrylamidehas been developed by Pharmacia under the trade
name Sephacryl. The advantage of matrix is the good flow rate that results be-
cause the support is exceptionally rigid. Unfortunately, the nonspecific adsorption
is increased over that of other dextran gels. The use of dextran gels is partly
restricted by their rather low porosity. They are widely used without any modifi-
cation as specific sorbents for the isolation of a series of lectins. One example
is the isolation of concanavalin A from jack bean seeds [49].
Bioaffinity Chromatography
C. Synthetic Copolymers
The advantages of HPLBAC, shown in Fig. 10, and the usefulness of the prepara-
tion of biologically active compounds on a pilot or industrial scale are the impetus
behind the continual development of synthetic polymers. The main appeal of
solid supports for these applications is their inherent mechanical stability, which
provides good flow characteristics even under high pressures. They can be oper-
ated at pressures up to 100 psi and usually tolerate a wide pH range. They are
124 Turkova
suitable for affinity ligand immobilization and provide bioaffinity supports with
high capacities. They are biologically inert and because of their inert structure
are not subject to enzymatic or microbial degradation. The chemical Structure of
these supports can be characterized by their polyethylene backbone, which creates
excellent chemical and physical stability. They also contain modifiable side
chains R , , R2, R3, R4:
1. Acrylamide Derivatives
Polyacrylamide gels are produced by copolymerization of acrylamide with the
bifunctional crosslinking agent N,N1-methylenebisacrylamide.The monomers
used in this synthesis are highly toxic and thus should be handled with care. The
main producer of polyacrylamide gels is Bio-Rad under the trade name Bio-Gel
P. This gel is produced with a range of pore sizes, from Bio-Gel P-2 with a
molecular weight exclusion limit of 1800, to Bio-Gel P-300 with a molecular
weight exclusion limit of 400,000. Commercial polyacrylamide beads are pur-
chased in the dry state and are swollen by mixing with water or aqueous solutions
for period of 4-48 h depending on the porosity. Bio-Gel P products are stable
to most eluants used in biochemical studies, including dilute solutions of salts,
detergents, urea, and guanidine hydrochloride, although high concentrations of
these reagents may alter the exclusion limits by up to 10%. The use of media
with pH values outside the range 2- 10 is to be avoided since some hydrolysis of
the amide side groups may occur with the consequent appearance of ion exchange
groups. Polyacrylamide gels are biologically inert and are not attacked by micro-
organisms.
Nonionic synthetic supports are obtained by copolymerization of N-acry-
loyl-2-amino-2-hydroxymethyl-1,3-propane diol with hydroxylated acrylic bi-
functional monomer. Hydrophilic supports under the trade name Trisacryl G F
are manufactured by IBF Reactifs. By strictly controlling the polymerization pro-
cess, it is possible to synthesize a complete line of products with covering a
Bioaffinity Chromatography 125
wide range of molecular weights from 3000 (Trisacryl GF05) to about 20 million
(Trisacryl GF 2000). Trisacryl is characterized by a high degree of hydrophilicity,
due both to the presence of primary alcohol groups and also the secondary amide
function. It can be used under pressures up to 2-3 bar and is not affected by
organic solvents such as alcohols, ketones. dioxane, or chlorinated solvents. It
is stable to low (-20°C) and high (121 "C) temperatures. Denaturing agents have
no effect on the gel because its structure involves no hydrogen bonds. It is also
stable to acidic pH, but less stable to high pH because of the slow hydrolysis of the
amide linkage. Adsorption of lysozyme on the Trisacryl GF 2000 with attached
Cibacron Blue F3GA in comparison with dye affinity sorbents prepared by use
of other solid supports was studied by Anspach et al. [50].
2. Methacrylate Supports
The copolymerization of hydroxyalkyl methacrylate with alkylene dimethacry-
lates gives rise to heavily crosslinked xerogel microparticles which subsequently
aggregate and yield macroporous spheroids. Their structure is shown in Fig. 12.
These gels have some chemical properties in common with agarose [53]. Hydroxy-
alkyl methacrylate supports have been marketed under the trade name Separon
HEMA and is produced by Tessek S.R.O. (Prague, Czech Republic), or under the
trade name Spheron was produced by Lachema (Brno, Czech Republic). These
supports have good chemical and mechanical stability and are stable to heating
for 8 h in 1 M sodium glycolate at 150°C or boiling in 20% hydrochloric acid
for 24 h. They are biologically inert and are not attacked by microorganisms.
They can be employed in organic solvents. HPLBAC of porcine pepsin on Sep-
aron HlOOO modified with E-aminocaproyl-L-phenylalanyl-D-phenylalanine
methyl ester has been described by Turkovi et al. [54].
Hydroxyethyl methacrylate support is also produced under the trade name
Dynospheres by Dyno Particles. Their monodisperse microparticles with a size
range of 0.3-5 pm are promising synthetic polymers for HPLBAC application.
The Japanese firm Toyo Soda Manufacturing Co. (Tokyo, Japan) has devel-
oped a semirigid gel, a copolymer of oligoethylene glycol, glycidyl methacrylate,
and pentaerythrol dimethacrylate under the trade name Toyopearl. The identical
copolymer under the name Fractogel TSK HW type is sold by E. Merck (Darm
stadt, Germany). Optimal conditions for the activation of free hydroxyl groups
on the gel matrix by epichlorohydrin and subsequent immobilization of ligands
were investigated by Matsumoto et al. 1551. They successfully prepared affinity
adsorbents for bioaffinity chromatography of lectins and trypsin. The advantage
of this gel is its pressure stability up to 7 bar. Swelling of dry gels in water is
3-4 mllg for Fractogel TSK HW-65. Its molecular exclusion limit is 5 X 10"
for proteins and lo6 for polyethylene glycols or dextrans. The negligible change
126 Turkova
I
C0
I
C C H ~ C H ~ O ~ IE H 2 C H 2 0 H
-
MACROPORE
MACROSPHEROID
in swelling volume with changing eluents, results i n a very constant gel bed
volume. It may be used from pH 1 to 14. The high chemical stability has led to
applications at high temperature and therefore it may be autoclaved. Its properties
render Fractogel TSK particularly suitable for large-scale industrial application.
Oxirane acrylic beads are obtained by copolymerization of methacrylam-
ide, methylenebismethacrylamide, and allylglycidylether. Due to the nature of
monomers the copolymer has neutral and mostly hydrophilic matrix, with a slight
hydrophobic component due to the methyl groups along the polymer backbone.
The oxirane group content is 1000 pmollg dry beads. The beads are macroporous
and they show a water regain of 2.5 mllg of dry beads. This water regain is
independent of pH (0.5- 12.5) and ionic strength. The beads are morphologically
and chemically stable under these conditions, even if exposed to them for several
weeks. The mechanical stability upon stirring is very good. At present oxirane
acrylic beads are produced under the trademark Epergit by Rohm Pharma GMBH
Bioaffinity Chromatography 127
(Darmstadt, Germany). HPIAC using Eupergit C beads was developed and opti-
mized by Fleminger et al. [56].
During chemical reactions involving one ol' the polymers, certain precau-
tions must be taken in view of the other polymer properties. Thus the gel must
not be exposed to strongly alkaline media, since the amide groups will be hy-
drolyzed to carboxylic acids. The limited heat resistance of the agarose must
equally be respected: Ultrogel AcA must not be exposed to temperatures greater
than a b o ~ 40°C.
~t The preparation of po.lyacrylhydrazidoagarose based on perio-
date oxidation of Sepharose followed by reaction with polyacrylhydrazide has
been described by Miron and Wilchek [58]. These yielded matrices which were
colorless and stable after reduction with sodium borohydride. Polyacrylhydrazi-
doagarose could be used either directly or after further modification with various
additional reactive groups.
F. Inorganic Support
Inorganic supports have been reviewed by Weetall and Lee 1591. They classif ed
these supports into a few major categories: metals, metal oxides, ceramics, and
glasses. They described the preparations, properties, and applications of porous
glass, porous silica, titania, alumina, and zirconia bodies, and iron and nickel
oxides. These supports have an inherent advantage in their rigidity. Since inor-
Bioaffinity Chromatography 129
ganic particles are friable materials, one should never use a stirring bar when
working with these materials. Particles can be separated for washing or assay
by several convenient methods. These include filtration, centrifugation, settling,
aspiration, or magnetic separation. Several of these methods can be utilized for
sizing inorganic particles, particularly when clumping has occurred or when one
wants particles of only a specific size range. Particles may be sized on the basis
of settling times, centrifugation speed, or filter porosity. The best method usually
determined by considering the size range that is desired and choosing the method
most likely to yield the desired particle range.
ports are available commercially in both irregular or spherical shapes, either un-
coated or glycophase-coated. Among them are, for example, silica-based supports
under the trademark LiChrospher Si, LiChrospher Diol, etc. (E. Merck, Darm-
stadt, Germany), Porasil A-F (Waters Associates, Milford, MA, USA), Progel-
TSK columns (Supelco, Bellefonte, PA, USA). Ultraaffinity-EP is one of the
commercially available epoxide silicas available in prepacked columns (Beck-
mann Instruments, Berkeley, CA, USA). Immobilization of the ligand onto such
a matrix can be performed by passing the material through the column slowly,
over a predetermined time period. The use of antichaotropic anions such as phos-
phates and sulfates is recommended because they stabilize the protein during
derivatization and increase their interaction with the activated support. Columns,
cartridges, or kits of a silica activated with epoxide functional bonded phases are
sold under the name Durasphere AS by Alltech Associates (Deerfield, IL, USA).
3. Periodate Oxidation
A simple method for the binding of proteins to insoluble polysaccharides after
their periodate oxidation has been described by Sanderson and Wilson [78]. The
aldehyde formed reacts with the protein. Subsequent reduction with sodium boro-
hydride led to the stabilization of the bonds between the protein and the polysac-
charide, and to the reduction of the residual aldehyde groups. In order to compare
the coupling of ligands to solid supports via presumed aldehyde groups, the
amount of G l y - D , L - Pwas
~ ~ coupled to periodate-oxidized glucose-Separon and
to Separon with attached hexamethylenediamine and activated by glutaraldehyde
in relation to pH [79]. From Fig. 17 it is evident that the two activated matrices
are clearly different. Another big difference between these two types of aldehyde
groups was also found in the stability of bonds with glycyl-D,L-phenylalanine.
Unlike the situation with oxidized glucose residues, in which case the reduction
with NaBH, is necessary, the bond between glutaraldehyde and dipeptide is very
stable. There is no difference between reduced and untreated preparation. In both
variants no release of nitrogen was found after 11 weeks at pH 7.0 and 24°C.
tional derivatives it should be born in mind that side reactions, such as crosslink-
ing of the support, may occur, and the permeability may decrease drastically.
gel by washing with water. The pH range used for the carbodiimide condensation
is 4.7-6.5 and the reaction time is 1.5-72 h at a carbodiimide concentration
of 2-100 mglml. The disadvantage is that carbodiimides are relatively unstable
compounds and must be handled with care because of their toxicity. After the
coupling of the affinant, the reaction ought to be continued to block reactive
groups by carrying out further carbodiimide reactions with glucosamine or 2-
aminoethanol in the case of CH-Sepharose, or with acetic acid as blocking agent
for the amino groups of AH-Sepharose.
Boschetti et al. [88] published a comparative study of soluble carbodiimide
CMC with a condensation agent, N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquino-
line (EEDQ). The latter catalyzes the reaction between spacer arm and ligand by
forming a mixed anhydride on the carboxyl group of the spacer arm. The mixed
anhydride then reacts with the complementary amine to form a stable amide bond.
The mixed anhydride can also react with other nucleophilic groups such as sulfhy-
dry1 and hydroxyl groups. EEDQ must be used in a water-ethanol mixture, which
is an advantage when working with ligands that are only slightly soluble in water.
EEDQ is a suitable condensation agent, being stable, nontoxic, and cheap.
7. Active Esters
Cuatrecasas and Parikh [891 described the preparation of N-hydroxysuccinimide
(NHS) esters of succinylated aminoalkyl agarose derivatives. These active ester
derivatives of agarose, when stored in dioxane, are stable for several months.
These derivatives very rapidly form stable amide bonds (at 4°C) with nonproton-
ated forms of primary aliphatic or aromatic amino groups at pH 6-9. Among the
functional groups of amino acids tested, only sulfhydryl groups compete effec-
tively with the amino groups during the binding reaction.
Using the esterification of the carboxyl groups of CH-Sepharose 4B with
the application of N-hydroxysuccinimide, Pharmacia produces activated CH-
Sepharose 4B. The pH range suitable for binding on this derivative is indicated
by Pharmacia to be 5-10, with an optimum of pH 8. The advantage of lower
pH values is in the decreased ester hydrolysis but with a slower reaction rate.
Buffers that contain amino acids cannot be used (Tris or glycine buffers) in the
coupling reaction. Agarose derivatives containing N-hydroxysuccinimide ester
have been introduced by Bio-Rad under the names Affi-Gel 10 and Affi-Gel 15.
As shown in Fig. 18, Affi-Gel 10 couples proteins best at a pH near or below
their isoelectric point, and Affi-Gel 15 couples proteins best near or above their
isoelectric point. Therefore, when coupling at neutral pH (6.5-7.5), Affi-Gel 10
is recommended for proteins with isoelectric points of 6.5-1 1 (neutral or basic
proteins) and Affi-Gel 15 is recommended for proteins with isoelectric points
below 6.5 (acidic proteins). The difference in coupling efficiency of Affi-Gel 10
and Affi-Gel 15 for acidic and basic proteins can be attributed to interactions
142 Turkova
between the charge on the protein and charge on the gel. Hydrolysis of some of
the active esters during aqueous coupling will impart a slight negative charge to
Affi-Gel 10. This negative charge will attract positively charged proteins (proteins
buffered at a pH below their isoelectric point) and enhance their coupling effi-
ciency. Conversely, the negative charge will repel negatively charged proteins
(proteins buffered at a pH above their isoelectric point) and lower their coupling
efficiency. Affi-Gel 15, due to the tertiary amine incorporated into its arm, has
a slight overall positive charge, and the effects are reversed. Coupling under
anhydrous conditions is the preferred method when this is suitable for the ligand.
Since there is no hydrolysis of active esters in the absence of water, the only
reaction will be that of the ligand with the gel. After the coupling of affinity
ligands the unreacted groups can be eliminated by addition of 0.1 M Tris buffer,
pH 8.
8. Activation with Carbonylating Reagents
Activation of crosslinked agarose with 1,l'-carbonyldiimidazole (CDI) has been
described by Bethell et al. [90]. The activated imidazolylcarbamate supports react
Bioaffinity Chromatography 143
with primary amino groups at pH 8.5-10.0 and are more stable to hydrolysis
compared with N-hydroxysuccinimide ester-activated supports. CDI-activated
agarose has a half-life of more than 14 weeks when stored in dioxane. A further
advantage of using CDI-activated agarose is that the resulting N-alkylcarbamates
are uncharged in normal pH ranges. Optimization of protein immobilization on
CDI-activated diol-bonded silica has been described by Crowley et al. [9]]. CDI-
activated glass matrices are commercially available, e.g., from Pierce Chemical.
9. Triazine Method
The covalent linkage of ligands to hydroxyl-containing supports activated with
cyanuric chloride (2,4,6-trichloro-s-triazine = TCT) was developed by Kay and
Crook [92]. An easily controllable variant for use with macroporous cellulose
bead with TCT was developed by BeneS et al. [93]. Bilkovi et al. [5] used this
method of coupling for the immobilization of the natural chymotrypsin inhibitor
antilysine. The disadvantage of this method is that the activating reagents are
highly toxic.
12. Diazotization
The first attachment of an affinant to cellulose was carried out by means of diazo-
nium groups by Campbell et al. [95]. The affinants are bound by their aromatic
144 Turkova
residues (mainly tyrosine and histidine), but also slowly through their amino
groups. Although this method is not frequently used at present, it offers two
essential advantages: (1) the bound ligand can easily be split off by reduction
with sodium dithionite; (2) this reductive cleavage allows protein-inhibitor conju-
gates to be isolated intact under mild conditions.
more favorably to the extracted polymer than do structural analogs. Wulff and
Vesper [loo] preparecl chromatographic sorbents with chiral cavities for racemic
resolution. For this purpose, with the aid of a chiral template molecule, functional
groups were placed in a highly crosslinked polymer in such a way that they were
present in a chiral cavity in a given stereochemistry. For example, Cnitrophenyl-
a-~-mannoside-2,3;4,6-di-O-(4-~inylphenylboronate) (A) was copolymerized to
a macroporous polymer, from which the template 4-nitrophenyl-a-D-mannopyra-
noside (B) could be split off. These polymers were used for the chromatographic
resolution of the racemate of the template molecule B. Chromatographic resolu-
tion of racemic mixtures of amino acid derivatives by use of molecular imprinting
of amino acid derivatives in macroporous polymers was described by Sellergren
et al. [ l o l l .
Synthesis and characterization of polymeric receptors for cholesterol by
use of MIT was published by Whitcornbe et al. [102]. The polymers obtained
by this method were shown to bind cholesterol with a single dissociation constant,
thus displaying characteristics similar to the true biological receptor or synthetic
host. Mosbach [lo31 informed of an increasing number of applications of MIT.
These include (1) the use of molecularly imprinted polymers as tailor-made sepa-
ration materials; (2) antibody and receptor binding site mimics in recognition and
assay systems; (3) enzyme mimics for catalytic applications; and (4) recognition
elements in biosensors. The stability and low cost of molecularly imprinted poly-
146 Turkova
mers make them advantageous for use in analysis as well as in industrial scale
production and application.
et al. [5 I ] list 34 suppliers, their addresses, and the products including affinity
matrices, activated supports, activation reagents, immobilized ligands, prepacked
columns and accessories, and affinity purification kits. Turkov6 [7] lists addresses
of 90 companies providing activated supports and biospecific adsorbents, along
with examples of their products. However, it was already mentioned that many
suppliers or their commercially available supports have been changing rapidly.
In order for the immobilized affinity ligands to be readily accessible to the binding
sites of biological macromolecules, it is necessary to have more than a solid
support with high porosity. The chemical groups of the affinant that participate
in the interaction with the macromolecular substance must also be sufficiently
remote from the surface of the solid matrix to avoid steric hindrance. The impor-
tance of spacing between the low molecular weight ligand and the surface of the
matrix in bioaffinity chromatography was illustrated by Cuatrecases et al. [I051
in one of the first successful applications of bioaffinity chromatography in the
isolation of enzymes. Figure 20 represents the bioaffinity chromatography of a -
chymotrypsin, both on Sepharose coupled with &-aminocaproyl-D-tryptophan
methyl ester (A) and on Sepharose coupled with D-tryptophan methyl ester (B),
in comparison with chromatography on unsubstituted Sepharose. In the first in-
stance (A), the bound inhibitor has high affinity for a-chymotrypsin and the en-
zyme can be released from the complex only by decreasing the pH of the eluting
buffer. By using 0.1 M acetic acid, pH 3.0, chymotrypsin is eluted in a sharp
peak and the volume of the eluted chymotrypsin does not depend on the volume
of the sample applied to the column. In the second instance (B), the inhibitor
coupled directly on Sepharose has a much lower affinity for the a-chymotrypsin
owing to steric hindrance. In this instance a change of buffer is not necessary
for enzyme elution and, as can be seen from the graph, the enzyme is eluted in
a much larger volume closely after the inactive material. In order to verify that
nonspecific adsorption on the carrier did not take place under the given experi-
mental conditions, chromatography of a-chymotrypsin on an unsubstituted car-
rier was carried out (C).
Studies on the conformation of model peptides in membrane-mimetic envi-
ronments [I061 lead the authors to expect that steric accessibility of the reactive
groups of the affinity ligand not only will be determined by their distance from
the surface of the solid support but will also depend on the interfacial water
region, which does not stabilize the same conformation as does bulk water. The
arrangement of the solvent layers on the surface of the solid support will also
obviously be one of the main factors affecting the penetration of the bound mole-
Turkova
ELUTION VOLUME , ml
Figure 20 Bioaffinity chromatography of a-chymotrypsin on inhibitor Sepharose col-
umns. The columns (50 X 5 mm) were equilibrated and run with 0.05 M Tris-hydrochloric
acid buffer of pH 8.0. Each sample (2.5 mg) was applied in 0.5 ml of the same buffer.
Columns were run at room temperature with a flow rate about 40 ml/h and fractions
containing I ml were collected. The arrows indicate a change of elution buffer (0.1 M
acetic acid, pH 3.0). (A) Sepharose coupled with E-aminocaproyl-D-tryptophanmethyl
ester. (B) Sepharose coupled with D-tryptophan methyl ester. (C) Unsubstituted Sepharose.
The first peaks in A and B were devoid of enzyme activity. (From R Cuatrecasas P, et
al. Proc Natl Acad Sci USA 1968; 639-643.)
cules to the surface of the support. As a logical result, the mode of immobilization
has a greater effect on the interaction of the substances isolated with immobilized
low molecular weight ligands than with high molecular ligands.
Angal and Dean [I 071 studied the effect of matrix on the binding of human
serum albumin to the sulfonated aromatic dye Cibacron Blue 3G-A immobilized
on 10 solid supports. The results are summarized in Table 3. A 16-fold range in
ligand concentration was observed in the amount of dye immobilized to the vari-
ous matrices despite the similarity of the reaction conditions used. The adsorption
was shown to be selective because rabbit, chicken, and bovine albumin did not
bind to the same immobilized dye.
High molecular weight affinity ligands usually offer more possibilities for
Bioaffinity Chromatography 149
is low and the ligand is preferentially bound only to readily accessible sites (Fig.
21, middle). The low density of the affinity ligand is also required to prevent
nonspecific binding. The top part of the figure illustrates the sorption of macro-
molecules, e.g., enzymes, that d o not have a complementary binding site for the
immobilized affinity ligand. This binding is caused by the high density of the
immobilized affinity ligand, permitting the formation of multiple nonspecific
bonds between the macromolecules in solution and the solid phase. These nonspe-
Bioaffinity Chromatography 151
cific bonds allow the compounds present in the mobile phase to bind to the affinity
ligand, the spacer, and the surface of the solid matrix. These multiple nonspecific
bonds may be stronger than a single complementary bond between the isolated
enzyme and immobilized complementary affinity ligand, e.g., an inhibitor. When
the nonspecific multiple bonds are involved in addition to the specific comple-
mentary bond (bottom part of the figure) they increase the strength of the binding
in the specific complex. This results in the elution of an enzyme in several frac-
tions and also gives rise to difficulties in enzyme elution. The multiple nonspecific
bonds may then lead to binding of the enzyme to the immobilized ligand in an
incorrect orientation (bottom part of the figure left). Thus, affinity chromatogra-
phy may yield good results only on a sorbent containing a low-affinity ligand
concentration (middle part of the figure) where the enzyme can bind only via the
complementary bond to the immobilized affinity ligand at a ratio of 1 : 1.
In order to determine experimentally the effect of the concentration of the
immobilized inhibitor on the course of affinity chromatography of proteolytic
enzymes, specific sorbents for carboxylic proteinases containing different con-
centrations of &-aminocaproyl-L-Phe-D-Phe-OMewere prepared by Turkovi et
al. [109]. The inhibitor was coupled to epoxide-containing Separon H 1000; the
resulting concentrations of E-aminocaproyl-L-Phe-D-Phe-OMein pmollg of dry
gel were 0.85, 1.2, 2.5, 4.5, and 155. Solutions of porcine, chicken, or human
pepsin were applied continuously to columns of these affinity sorbents until the
effluent showed the same activity as the solution applied (cf. Fig. 22). On columns
of affinity sorbents containing the inhibitor attached in the concentration range
0.85-4.5 pmollg, in all cases one sharp peak of very active pepsin was achieved.
The amount of desorbed pepsin was calculated from the adsorbance at 278 nm
and proteolytic activity measurement. In contrast, using the affinity sorbent con-
taining the inhibitor at a concentration of 155 pmollg, several pepsin peaks were
seen. This different behavior of the enzyme on affinity sorbents having low and
high amounts of immobilized inhibitor may be due to multiple bonding of the
enzyme molecule and inert proteins, as illustrated in Fig. 21. The amounts of
porcine, chicken, and human pepsins eluted depending on the concentration of
E-aminocaproyl-L-Phe-D-Phe-OMeof the individual affinity sorbents were deter-
mined. From the comparison of individual pepsins it was evident that &-aminoca-
proyl-L-Phe-D-Phe-OMe-Separonis a very good sorbent only for porcine pepsin.
The specific sorbent containing 0.85 pmol of inhibitor per g of dry support sorbed
29.4 mg of porcine pepsin per g of dry sorbent. Using the molecular weight of
pepsin (35,000) it can be calculated that 99% of the inhibitor molecules attached
were involved in the specific complex. As the amount of the affinity ligand
attached increased, the portion of the inhibitor molecules involved in the specific
complex with pepsin decreased sharply. With specific sorbents containing 4.5
pmol inhibitor per g only 26% of the total number of inhibitor molecules attached
FRACTION NUMBER
take part in the sorption of pepsin. In an affinity sorbent with the lowest concen-
tration of the affinity ligand only, all of the molecules of the affinity ligand are
fully available for the formation of the complex with the isolated enzyme.
Liu and Stellwagen [I101 used Cibacron Blue F3GA immobilized at several
dye densities to study the different adsorptions of monomeric octopine dehydro-
genase and tetrameric lactate dehydrogenase. They determined that the half-time
for desorption of lactate dehydrogenase from Cibacron Blue F3GA-Sepharose
CL-6B was nearly identical (27 s) to the mass transfer half-time of the protein
in the matrix. The change in the visible adsorbance of Cibacron F3GA accompa-
nying its complexation with lactate dehydrogenase was used to observe the kinet-
ics of complexation. The results of their experiments indicated that it is chromato-
graphic mass transfer and not the chemistry of complexation that limits zonal
chromatography. This is why the effect of immobilized dye concentration on
protein complexation is usually studied using zonal chromatography.
PROTEIN A
,
TIME min
best be exploited: (1) the concentration of the desired product in the starting
material; (2) the composition of other components in the starting material, along
with its physical and chemical properties; (3) the desired product purity; and (4)
the volume of material to be processed. Each stage of an affinity chromatography
process-adsorption, washing, elution, and regeneration-needs to be optimized
before the process is scaled up. Moreover, each step must be consistent and repro-
ducible. Bioadsorbent stability is an important criterion because the ligands are
often labile biological molecules. Affinity media can lose its effectiveness be-
cause of unstable ligands, microbial contamination, and column clogging due to
the presence of insoluble matter in the sample and in the eluting buffers. Accumu-
lation of denatured protein, lipids, nucleic acids, etc., that are not eluted during
the regeneration process can also limit the lifetime of the column. A preparative
column is exposed to more protein in three or four preparative cycles than an
analytical column is exposed to in two or three hundred cycles. Under these
conditions, maintenance of the affinity media becomes very important. Stringent
clean-in-place procedures are recommended by the media manufactures to pro-
long its lifetime. The feasibility of such measures should be taken into consider-
ation before a purification method is scaled up. Suitable affinity ligands for large-
scale isolations are polyclonal or monoclonal antibodies because they can be
produced against any compound, even if the latter is only partially purified. Fur-
thermore, monoclonal antibodies can be selected with any desired affinity,
thereby making the use of biospecific columns, prepared by their immobilization,
very attractive. Such antibodies show absolute specifity for only one single epi-
tope: the smallest immunologically submolecular group on an antigen. Mono-
clonal antibodies can be produced in large quantities by the hybridoma technol-
ogy developed by Kohler and Milstein [112]. Tarnowski and Liptak [113] used
this antibody for the automated immunosorbent purification of interferon. Large-
scale purification of monoclonal antibody, which recognized a human mela-
noma-associated 250-kDa glycoprotein/proteoglycan,was isolated by Lee et al.
[114] by use of staphylococcal Protein A-Sepharose. As improvements in the
technology of chromatographic support materials are developed, the combination
of the unique selectivity of an affinity interaction along with the improved perfor-
mance of modern supports assures bioaffinity chromatography a commanding
position in the future of large-scale purification. At present, affinity chromatogra-
phy is already being increasingly used in large-scale purification of therapeutic
products.
enzymes are produced by organisms for their own use: in regulated metabolic
processes their low stability, narrow specificity, and strictly defined requirements
are inherently connected with their function. However, it is important to take into
account that after completion of their function in living cells, their denaturation
and hydrolysis by proteinases occurs. For their stabilization in vivo as well as
their function, hydrophobicity plays an important role. However, the contact of
nonpolar amino acids with water is enthalpically disadvantageous and results in
ice-like water structure. Such contact of water with hydrophobic surface clusters
of proteins in vitro decreases protein stability. Hence, reduction of the nonpolar
surface area should stabilize proteins. Shami et al. [I 151 showed the dramatically
increased activity of amylase complexed with their antibodies. Sheriff and co-
workers [I 161 used x-ray crystallography to determine the three-dimensional
structure of antibody-antigen complex by use of antilysozyme Fab and lysozyme.
They demonstrated that more than 80 van der Waals bonds are formed during
the antibody-lysozyme interaction. Formation of the complex resulted in exclu-
sion of all molecules of water. The conclusion of cited data was that oriented
immobilization of enzymes by use of antigen-antibody interaction can result both
in good steric accessibility of the enzyme active site toward high molecular mass
substrates and an increased stability. Moreover, biospecific adsorption of an en-
zyme to a suitable immunosorbent combines the isolation of molecules with their
oriented immobilization.
To confirm this hypothesis, polyclonal antibodies suitable for the oriented
immobilization of chymotrypsin were prepared by chromatography on a bioaffin-
ity matrix that had the enzyme immobilized through its active site. This was
prepared by the attachment of chymotrypsin to pancreatic trypsin inhibitor antily-
sine, covalently linked to bead cellulose (Fig. 24). After periodate oxidation of
their carbohydrate moieties, the isolated antibodies were coupled to a hydrazide
derivative of bead cellulose and used for the sorption of chymotrypsin [5]. The
failure to detect any chymotryptic activity toward N-succinyl-I.-phenylalanyl-p-
nitroanilide showed that the enzyme was immobilized via its active site. No de-
crease in proteolytic activity resulted from incubation of chymotrypsin with spe-
cific antichymotrypsin IgG in a 1 : 1 molar ratio. These isolated antibodies are
thus suited for the preparation of a biospecific affinity matrix bearing immobilized
chymotrypsin oriented such that substrate proteins are accessible to its active
site. The antigenic affinity of the antichymotrypsin antibodies was essentially
unchanged by periodate oxidation of their carbohydrate moieties. Therefore, this
procedure was used to covalently attach isolated antichymotrypsin antibodies to
a hydrazide derivative of beaded cellulose. The molar ratio of biospecifically
adsorbed chymotrypsin molecules to immobilized antibody was 2 : 1, which is
the value predicted for enzyme occupancy of each of the two Fab binding sites
in IgG. The immobilized chymotrypsin retained practically 100% of the native
catalytic activity determined by use of the high molecular substrate, i.e., dena-
tured hemoglobin. Its proteolytic activity has not changed after more than a year.
Bioaffinity Chromatography
G LTA
and therefore was used for the preparation of a highly active immobilized enzyme
by Solomon et al. [ I 191. They prepared the complex of peroxidase and mAB in
solution and attached this imrnunocomplex to immobilized anti-Fc antibodies.
REFERENCES
Kay J, Afting EG, Aoyagi T, Dunn M. The effects of lactoyl-pepstatin and the
pepsin inhibitor peptide on pig cathepsin D. Biochem J 1982;203:795-797.
Bilkovi Z, ChuriEek J, Kuzerovi Z, Turkovi J. Purification of anti-chymotrypsin
antibodies for the preparation of a bioaffinity matrix with oriented chymotrypsin
as immobilized ligand. J Chromatogr 1997;689:273-279.
Winzor DJ. Review: recent developments in quantitative affinity chromatography.
J Chromatogr 1992;597:67-82.
Turkovi J. Bioaffinity Chromatography. 2nd ed. Amsterdam: Elsevier, 1993.
Turkovi J, Seifertovi A. Affinity chromatography of proteases on hydroxyalkyl meth-
acrylate gels with covalently attached inhibitors. J Chromatogr 1978;148:293-297.
Tosa T, Sato T, Sano R, Yamamoto K, Matuo Y, Chibata I. Characteristics and
applications of N-(w-aminohexy1)-L-asparticacid-Sepharose as an affinity adsor-
bent. Biochim Biophys Acta 1974;334:1-1 1.
Turkovi J, Fusek M, Maksimov JJ, Alakhov YuB. Reversible and irreversible im-
mobilization of carboxypeptidase Y using biospecific adsorption. J Chromatogr
1986:315-321.
Trayer IP, Trayer HR. Affinity chromatography of nicotinamide nucleotide-depen-
dent dehydrogenases on immobilized nucleotide derivatives. Biochem J 1974;141:
775-787.
Craven DB, Harvey MJ, Lowe CR, Dean PDG. Affinity chromatography on immo-
bilized adenosine 5'-monophosphate. Eur J Biochem 1974;41:329-340.
Thompson ST, Cass KH, Stellwagen E. Blue dextran-Sepharose: an affinity column
for the dinucleotide fold in proteins. Proc Natl Acad Sci USA 1975;72:669-672.
Biellmann JF, Samama JP, Briinden CI, Eklund H. X-Ray studies of the binding of Ciba-
cron Blue F3GA to liver alcohol dehydrogenase. Ew J Biochem 1979;102:107-1 10.
Vijayalakshrni MA, Bertrand 0, eds. Protein-dye interactions: developments and
applications. New York: Elsevier, 1989.
Phillips TM. Queen WD, More NS, Thompson AM. Protein A-coated glass beads:
a universal support media for high performance immunoaffinity chromatography.
J Chromatogr 1985;327:213-219.
Lee WC, Chuang CY. Performance of pH elution in high-performance affinity chro-
matography of proteins using non-porous silica. J Chromatogr A 1996;721:31-39.
Lis H, Sharon N. Affinity chromatography for the purification of lectins. J Chro-
matogr 1981;215:361-372.
Hatakeyama T, Murakami K, Miyamoto Y, Yamasaki N. An assay for lectin activ-
ity using microtiter plate with chemically immobilized carbohydrates. Anal Bio-
chem 1996;237: 188-1 92.
Kristiansen T. Group-specific separation of glycoproteins. Meth Enzymol 1974;
184:629-641.
Pergami P, Jaffe H, Safar J. Semipreparative chromatographic method to purify
the normal cellular isoform of the prion protein in nondenatured form. Anal Bio-
chem 1996;236:63-73.
Schott H. Affinity Chromatography: Template Chromatography of Nucleic Acids
and Proteins. Chromatographic Science Series. New York: Marcel Dekker, 1984.
Johnson AF, Wang R, Ji H, Chen D, Guilfoyle RA, Smith LM. Purification of
single-stranded M13 DNA by cooperative triple-helix-mediated affinity capture.
Anal Biochem 1996;234:83-95.
160 Turkova
Bioaffinity Chromatography
antibody by means of a cellulose-protein antigen. Proc Natl Acad Sci USA 1951;
37575-578.
Nilsson K, Mosbach K. Immobilization of enzymes and affinity ligands to various
hydroxyl group carrying supports using highly reactive sulfonyl chlorides. Biochem
Biophys Res Commun 1981 ;1O2:449-457.
Porath J, Sundberg L. High capacity chemisorbents for protein immobilization. Na-
ture New Biol 1972;238:26 1-262.
Ngo TT. Facile activation of Sepharose hydroxyl groups by 2-fluoro-l methylpyri-
dinium toluene-4-sulfonate: preparation of affinity and covalent chromatographic
matrices. Bio/Technology 1986;4:134- 137.
Ekberg B, Mosbach K. Molecular imprinting: a technique for producing specific
separation materials. Trends Biotechnol 1989;7:92-96.
Wulff G, Vesper W. Preparation of chromatographic sorbents with chiral cavities
for racemic resolution. J Chromatogr 1978; 167: 17 1 - 186.
Sellergren B, Ekberg B, Mosbach K. Demostration of substrate-and enantio-selec-
tivity by chromatographic resolution of racemic mixtures of amino acid derivatives.
J Chromatogr 1985;347:1 - 10.
Whitcombe MJ, Rodriguez ME. Villar P, Vulfson EN. A new method for the intro-
duction of recognition site functionality into polymers prepared by molecular im-
printing: synthesis and characterization of polymeric receptors for cholesterol. J
Am Chem Soc l995;l l7:7l05-7lll.
Mosbach K. The emerging technique of molecular imprinting and its future impact
on biotechnology. 12th International Symposium on Affinity Interaction, Kalmar.
Sweden, June 15-19, 1997, L15.
Kukongviriyapan V, Poopyruchpong N, Ratanabanangkoon K. Some parameters
of affinity chromatography in the purification of antibody against Naja naja sia-
mensis toxin 3. J Immunol Meth 1982;49:97-104.
Cuatrecasas P, Wilchek M. Single-step purification of avidin from egg white by
affinity chromatography on biocytin-Sepharose columns. Biochem Biophys Res
Commun 1968;33:235-239.
Gierasch LM, Lacy JE, Thompson KF, Rockwell AL, Watnick PI. Conformation
of model peptides in membrane-mimetic environments. Biophys J 1982;37:275-
284.
Angal S. Dean PDG. The effect of matrix on the binding of albumin to immobilized
Cibacron blue. Biochem J 1977;167:301-303.
Clemetson KJ, Pfueller SL, Luscher EF, Jenkins CSP. Isolation of the membrane
glycoproteins of human blood platelets by lectin affinity chromatography. Biochim
Biophys Acta 1977;464:493-508.
Turkovi J, Bliha K, Adamovi K. Effect of concentration of immobilized inhibitor
on the biospecific chromatography of pepsins. J Chromatogr 1982;236:375-383.
Liu YC, Stellwagen E. Accessibility and multivalency of immobilized Cibacron
Blue F3GA. J Biol Chem 1987;262:583-588.
Narayanan SR. Preparative affinity chromatography of proteins. J Chromatogr A
1994;658:237-258.
Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of pre-
defined specificity. Nature 1975;256:495-497.
r
Bioaffinity Chromatography
I. INTRODUCTION
The thymus gland, besides its well-recognized role in the immune system,
plays a role in reproductive biology, particularly in regulating ovarian function.
Studies using neonatally thymectomized mice or congenitally athymic nude mice
( n u h u ) have clearly shown that thymus gland influences ovarian function, al-
though the mechanism for this influence is not completely known [I]. In this
chapter, we shall summarize the experimental evidence that supports the regula-
tion of the ovaries by the thymus gland and the mechanisms that might be in-
volved. We further describe our characterization and partial purification of two
potentially novel thymic factors that directly influence ovarian steroidogenesis
in vitro.
Uzumcu et al.
ian dysgenesis was prevented by transfer of CD4+ CD5 + T cells [15]. Further-
more, autoreactive antibodies to the ovary have been shown in neonatally thymec-
tomized mice [16]. However, these antibodies were detected only after the age
of 30 days, which fails to explain the initial destruction at the ovary prior to day
30. The possible explanation of this failure is that disturbances of the ovary prior
to day 30 are due to the effect of thymectomy at the level of the hypothalamic-
pituitary axis. Specifically, the development of the hypothalamus remains incom-
plete if the thymus is absent or removed at an early age. Therefore, the underde-
veloped hypothalamus cannot release sufficient luteinizing hormone-releasing
hormone (LHRH), which in turn causes gonadotropin deficiency and ovarian dys-
genesis later in the life. Experimental findings using congenitally athymic mice
support this hypothesis. This strain of mice exhibits decreased levels of luteiniz-
ing hormone (LH) and follicle-stimulating hormone (FSH) in the pituitary gland
and in the circulation prior to vaginal opening (i.e., puberty), along with reduced
serum estrogen [17]. In addition, a reduced level of LHRH in hypothalami of
athymic nude mice was observed while the LH response of these animals to
exogenously administered LHRH in vivo was identical to that of their heterozy-
gote (nu/+) littermates [18]. Previously, Rebar and coworkers demonstrated that
reduced LH and FSH levels in athymic mice could be restored to normal by
neonatal thymic implantation 1191. Finally, it has been demonstrated that thy-
mosin fraction-5 (TF-5) and thymosin P4 stimulates LHRH from medial basal
hypothalami from normal cycling female rats superfused in vitro [20]. Thus, the
evidence from neonatally thymectomized and congenitally athymic mice suggests
that the influence of thymus on the reproductive functions is due to at least two
mechanisms:
1. Autoimmune. Removal of the thymus at an early age causes develop-
ment of autoimmune antibodies against functional structures at the
ovary due to the disturbance of the delicate balance at the immune
system.
2. Endocrine. Hormonal interactions between the thymus and hypothala-
mic-pituitary-ovarian axis exist. Since this interaction is disrupted in
neonatally thymectomized or athymic mice, ovarian dysgenesis devel-
ops later in the life.
In addition to these two separate but interrelated mechanisms, interaction
between the thymus and the ovaries may be partly through the direct effect of
secretory products of the thymus (i.e. thymic hormones) on the ovary without
any involvement of hypothalamus-pituitary axis or immune system. A consider-
able body of experimental evidence shows that there is a direct influence of vari-
ous immune products (e.g., interleukins and/or other cytokines) on ovarian func-
tion [21].
Uzumcu et al.
B. Preparation of RGC
Granulosa cells were prepared from 21- to 23-day-old diethylstilbestrol (DES)-
treated Sprague-Dawley female rats as previously described [27]. Briefly, granu-
losa cells were washed and plated at a density of 4 X l o 5 viable cells per well
in 24-well tissue culture plates containing 1 ml serum-free medium. The cells
were first cultured for 24 h at 37°C before TCM treatment.
C. Effect of TCM on Steroidogenesis from RGC
Steroidogenic activity of TCM was tested in time-course and dose-response
studies. TCM stimulated both progesterone (P4) and estradiol (E2) secretion from
Steroidogenic Factors and TCM
E+Zm No FSH A
FSH (100 nglml)
Q
m
Control --
ME HCM TCM Control -ME HCM TCM
10% conditioned media 10% conditioned media
Figure 1 Effect of TCM on basal and FSH-induced P, (A) and E2 (B) secretions from
cultured RGC as determined by radioimmunoassay (RIA). Data points are shown as the
mean 5 SD of picogram quantity of the steroids per microgram of cell protein from qua-
druplicate-culture wells in a representative experiment that was repeated 3 times with
similar results. The control represents the granulosa cells incubated with culture medium
containing no conditioned medium. (Modified from Ref. 28.)
RGC in a time- and dose-dependent manner [27]. Later, we observed 80-and 17-
fold increases in basal and FSH-induced P, secretion from rat granulosa cells
treated with 10% TCM for 48 h. (The treatment length was 48 h throughout the
entire study.) In addition, TCM stimulated basal and FSH-induced E2 secretion
about four- and threefold, respectively [28] (Fig. 1). Similarly, 10% TCM aug-
mented basal and FSH-induced 20a-hydroxyprogesterone (OH-P,) approxi-
mately 40- and 10-fold. Basal and FSH-stimulated aromatase enzyme activity
was also upregulated by TCM. The stirnulatory effect of TCM on steroidogenesis
from RGC was not due to cellular proliferation or growth since the TCM Ireat-
ment increased neither total cellular protein content nor [%]thymidine incorpora-
tion to the RGC [28].
Physicochemicaltreatments
nal volume with distilled water. The samples were clarified by centrifugation,
filtered using 0.45-ym membrane, and 1 ml of TCM concentrate was applied to
TSK-Gel G2000 SW gel filtration column (0.8 X 30 cm; TosoHaas, Philadelphia,
PA, USA) equipped with a TSK-GSW guard column (0.8 X 4 cm; TosoHaas).
A fast protein liquid chromatography (FPLC; Pharmacia, Piscataway, NJ, USA)
system was used to maintain the flow rate at 0.5 mllmin during elution of the
proteins with PBS containing 0.5 M NaCI. The column eluate was monitored at
280 nm using an in-line UV detector and 200-yl fractions were collected. Frac-
tions were stored at -70°C prior to assay. The molecular weight of the fractions
was calculated by reference to the elution positions of ovalbumin (45 kDa), tryp-
sin inhibitor (20.1 kDa), lactalbumin (14.2 kDa), and epidermal growth factor (6
kDa). Similarly, HCM and ME concentrates were also subjected to the gel filtra-
tion FPLC, and the collected fractions were also used as controls. Gel filtration
FPLC of TCM resulted in the elution of several major protein peaks as detected
at 280 nm [29] (Fig. 3).
30 40 50 60 70 80 90 100 110
Fraction numbers
Figure 3 Effect of TCM gel filtration fractions on P4 (A) and E, (e) secretion from
cultured RGC as determined by RIA. The figure also shows the absorbance of the column
eluate at 280 nm (a).In this experiment, control group is the granulosa cells that were
treated with culture medium with no gel filtration fraction. The elution positions of molecu-
lar weight marker proteins (a) ovalbumin (45 m a ) , (b) trypsin inhibitor (20.1 m a ) , (c)
lactalbumin (14.2), and (d) epidermal growth factor (6 kDa) are indicated at the top
(arrows). Data points for the steroids are expressed as picograms per microgram of cell
protein from single well. The experiment was repeated for at least 5 times with similar
results. (Modified from Ref. 29.)
Figure 4 Morphology of the RGC following a I-h treatment with FSH (500 nglml) (A),
TCM-1 (B), and control medium or TCM-22 (C). The cells that were treated with FSH or
TCM-I became rounded (mows) and left finger-like processes (arrowheads) attaching sub-
stratum. The cells that were treated with control medium or TCM-22 were well spread and
polygonal in shape. Magnifications of the micrographs are 720X. (Adapted from Ref. 29.)
Steroidogenic Factors and TCM 175
Uzumcu et al.
Fraction Numbers
Figure 5 Effect of TCM-I and TCM-22 alone or in combination on steroidogenesis
from RGC. Gel filtration fractions containing 90 yl TCM-I (fractim 83-94; triangles)
and TCM-22 (fraction 50-61; squares) were tested individually or 45 yl of successive
fractions from each activity region were combined (i.e., no. 50 with no. 83, no. 5 1 with
no. 84, etc.; circles) and tested for their combined effects on P4 (A) and E, (B) secretion
from RGC. In this experiment, control represents the granulosa cell that were treated with
culture medium containing PBSIO.5 M NaCI. The steroids were measured by RIA in cul-
ture medium and expressed as the mean + SD of picogram quantity of the steroids per
microgram of cell protein in single-culture well (n = 3). (Modified from Ref. 29.)
Steroidogenic Factors and TCM 177
4). The morphological changes that were induced by either FSH or TCM-1 started
approximately 15 min after treatment and were most prominent by 1 h post treat-
ment. The cells started gaining their normal appearance by 12 h and returned to
normal by 48 h post treatment.
Uzurncu et al.
0 10 100 0 10 100
TCM-1 (pllml) TCM-22 (pllml)
sample injection, 0-90% from 50 to 120 min, and 90% from 120 to 130 min. The
flow rate was 1 mllmin throughout, and the chromatogram (Azl4)was archived as
previously described [311. The column eluate was collected into tubes containing
50 p1 0.12510.25 N NaOH to immediately neutralize the TFA. For TCM- 1, frac-
tions of 500 p1 were collected 50 min after sample injection and 10-p1 aliquots
were tested for their stimulation of steroidogenesis or morphological changes in
cultured RGC. For TCM-22, fractions of 1 ml were collected immediately after
sample injection, and 10 p1 was tested for stimulation of P4 and E2secretion from
RGC. The remaining portion of the fractions was stored at -80°C until further
analyzed for peptide sequence of TCM- 1 or re-HPLC of the TCM-22.
80 85 90
2 Fraction numbers
a
-
0.00
0 20 40 60 80 100 120 140 160
l " " l " " l " " l " " l " " l " " l " " ~ ' ' ' ~ ~
50 60 70 80 90 100 110 120 130
Fraction numberslRetention time (min.)
VI. CONCLUSIONS
-'-
2 45 50 55 60 65
3 Fraction numbers
-a
20 30 40 ' 50 60 70 80 90 100
Fraction numbersIRetention time (min.)
ence on the ovaries. When we started our work, the available information sug-
gested that this influence may be through the autoimmune and endocrine
mechanisms described in the Introduction. However, our data and those of others
have highlighted the possibility of a direct influence of the thymus on the ovary
through thymic factors. Aguilera and Romano [22] reported the presence of a
28-kDa factor in thymic reticuloepithelial cell-or thymus-conditioned media that
inhibited hCG-induced P, and E2 production from rat ovarian cell but had no
effect on P4 and E2 production in the absence of hCG. Interestingly, the same
laboratory has recently reported that rat thymus contains a heparin-binding factor
that modulates steroidogenesis in the rat testis 1321. Ledwitz-Rigby and Scheid
[23,33] reported that thymulin, a nanopeptide produced by the thymus gland,
182 Uzumcu et al.
ACKNOWLEDGMENTS
The studies described from our laboratory in this chapter were supported in part
by NIH grant DK 45916, and NATO Scientific Training ProgramIScientific and
Technical Research Council of Turkey
REFERENCES
1. Chapman JC, Michael SD. Hormonal and immunologic interactions between thymus
and ovary. In: Grossman CJ, ed. Endocrinology and Metabolism. Bilateral Commu-
nication Between the Endocrine and Immune Systems. New York: Springer-Verlag,
1994: 12-35.
2. Nishizuka Y, Sakakura T. Thymus and reproduction: sex-linked dysgenesia of the
gonad after neonatal thymectomy in mice. Science 1969;166:753-755.
r
Rebar RW, Morandini IC, Benirschke K, Petze JE. Reduced gonadotropin in athymic
mice: prevention by thymic transplantation. Endocrinology 1980;107:2130-2132.
Rebar RW, Miyake A, Low TLK, Goldstein AL. Thymosin stimulates secretion of
luteinizing hormone-releasing factor. Science 198 1 ;2 14:669-67 1.
Terranova PF, Rice VM. Review: cytokine involvement in ovarian processes. Am
J Reprod Immunol 1997;37:50-63.
Aguilera G, Romano MC. Influence of the thymus on steroidogenesis by rat ovarian
cells in vitro. J Endocrinol 1989; 123:367-373.
Ledwitz-Rigby F, Schneid PC. Serum thymic factor enhances porcine granulosa
cell responsiveness to gonadotropins in vitro. The Eighth Ovarian Workshop on
Regulatory Processes and Gene Expression in the Ovary. Maryville, Tennessee,
USA, July 12-14, 1990.
Lewis VM, Twomey JJ, Bealmear P, Goldstein G, Good RA. Age, thymic involu-
tion, and circulating thymic hormone activity. J Clin Endocrinol Metab 1978;47:
145- 150.
Dardenne M, Savino W, Gagnerault M-C, Itoh T, Bach J-F. Neuroendocrine control
of thymic hormonal production. I. Prolactin stimulates in vivo and in vitro the pro-
duction of thymulin by human and murine thymic epithelial cells. Endocrinology
1989;125:3-12.
Uzumcu M. Lin YC. Factor(s) from thymic cell culture medium conditioned medium
(TCM) stimulate(s) progesterone (P) secretion in cultured rat granulosa cells (abstr).
Biol Reprod 1990;42 (Suppl 1):163.
Uzumcu M, Akira S, Lin YC. Stimulatory effect of thymic factor(s) on steroidogene-
sis in cultured rat granulosa cells. Life Sci 1992;Sl :l2l7- 12.28.
Uzumcu M, Lin YC. Characterization of the stimulatory actions of thymic factor(s)
on basal and gonadotropin-induced steroidogenesis in cultured rat granulosa cells.
Mol Cell Endocrinol 1994;105:209-216.
Uzumcu M, Brigstock DR, Lin YC. Partial purification and characterization of two
non-FSH steroid-modulating factors in rat thymic epithelial cell-conditioned me-
dium (TCM). Dom Anim Endocrinol 1998;15:155- 168.
Soto EA, Kliman HJ, Strauss JF 111, Paavola LG. Gonadotropins and cyclic adeno-
sine 3',Sf-monophosphate (CAMP)alter the morphology of cultures human granulosa
cells. Biol Reprod 1986:34:559-569.
Bray W, Brigstock DR. Raw integrator data storage and retrieval using data acquisi-
tion software. Amer Lab 1994;26:38.
Porras MG, Reyes J, Romano MC. The rat thymus contains a heparin-binding factor
that modulates steridogenesis in the testis. Acta Physiol Pharmacol Ther Latin Am
1996;46:286-293.
Ledwitz-Rigby F, Schneid PC. Serum thymic factor enhances porcine granulosa cell
responsiveness to gonadotropins in vitro. In: Gibori G, ed. Signaling Mechanisms
and Gene Expression in the Ovary. New York: Springer-Verlag, 1991:473-478.
Prepin J. Thymus and thymulin increase the proliferation of oogonia in fetal rat
ovary in vitro. C R Acad Sci Paris Ser. 111 199 1 :3 13:407-4 11.
Prepin J. Fetal thymus and thymulin stimulate in vitro gonocytes in fetal testis in
rats. C R Acad Sci Paris Ser. I11 1993;3l6:4S 1-454.
Prepin J, Le Vigouroux P. Inhibition by TGF-beta I of the in vitro thymulin-stimu-
r
lated proliferation of gonocytes from fetal rat testes. Reprod Fertil Dev 1997;37:
203-206.
Gorospe WC, Fong Y-Y, Spangelo BL. Thymosin fraction-5 modulates steroidogen-
esis and interleukin-6 release by rat granulosa cells in vitro. Endocr J 1993;1:35-
39.
Rosas P, Hinjosa L, Garcia L. Evidence for the participation of the thymus on the
regulation of ovarian development in the prepubertal mouse (abstr). Biol Reprod
1995;52 (Suppl 1):130.
Hinojosa L, Rosa P. Effects of the serum thymic factor (FTS) on the gonaodropin-
induced ovulation in normal and hypothymic mice (abstr). Biol Reprod 1996;54
(Suppl 1):85.
Oikawa M, Dargan C, Ny T, Hsueh AJW. Expression of gonadotropin-releasing
hormone and prothymosin-a messenger ribonucleic acid in the ovary. Endocrinol-
ogy 1990;l27:2350-2356.
Hall AK, Aten R1, Behrman HR. Differential modulation of thymosin genes in the
immature rat ovary by gonadotropins. Mol Cell Endocrinol 1991:79:37-43.
Hall AK, Aten Rl,Behrman IIR. Thymosin gene expression is modulated by preg-
nant mare's serum gonadotropin, human chorionic gonadotropin, and prostaglandin
F2, in the immature rat ovary. Endocrinology 1991;128:951-957.
Determination of Affinity Constants
of Lectins for Sugars by Affinity
Capillary Electrophoresis
Kiyohito Shimura and Ken-ichi Kasai
Teikyo University, Sagamiko, Kanagawa, Japan
I. INTRODUCTION
cable. Equilibrium mixture analysis, which is suitable for a system that shows
slow dissociation kinetics, measures the concentration of the free species in equi-
librium mixtures at different concentrations by zone electrophoresis. This allows
a binding isotherm to be determined and, thus, an estimation of the affinity con-
stant. Mobility change analysis, which is applicable to a system that shows rapid
dissociation kinetics, measures the mobility change of a zone of one component
in a solution of the other under a rapid binding equilibrium. The amount of the
mobility change can be correlated to the mole fraction of the bound species and
also leads to determination of the affinity constant. This latter format of affinity
CE is very useful as a microscale technique for the determination of affinity
constants for systems with relatively weak affinity, e.g., for systems with a disso-
ciation constant larger than the micromolar level.
The principle of the determination of affinity constants by mobility change
analysis has been established through the application of affinity electrophoresis
in gel slabs or rods [2.3]. This type of analysis has several advantages. First, it
is not necessary to determine precisely the concentration of an interacting compo-
nent applied as a zone. Considering the difficulty in the determination of precise
concentration of small quantities of a protein sample, this feature is particularly
valuable. Second, the sample that is applied as a zone need not necessarily be
pure, so long as the peak of the component can be traced. This represents a
big advantage, especially in cases where the protein is unstable and is prone to
denaturation. The simultaneous determination of the affinity constants of a group
of molecules sharing the same binding specificities is also possible, provided all
components can be separated. Third, the amount of an interacting component
applied as a zone can be very small.
In this chapter. we will describe the principle and applications of mobility
change analysis with two examples of the analysis of lectin-sugar interactions
by affinophoresis and affinity probe capillary electrophoresis (APCE). Before we
move to the body of this chapter, it would be useful to briefly review the basics
of CE.
- -
EOF EOF EP
where L (cm) is the distance between the injection end and the detection point,
and E (V cm-') represents the field strength. The polarity of the mobility is de-
fined as negative for movement toward the positive electrode. Thus, in the case
of the negatively charged ion (S) in Fig. 1, p,, is negative and pe, is positive.
Note that pep(cm2 V-' s-' ), a value of fundamental importance in the mobility
change analysis, is inversely related to the experimentally measurable value, t.
Coelectrophoresis of a neutral marker molecule allows the determination of p,
in the capillary and, therefore, pepof charged substances using Eq. (1). This point
will be discussed further in Section 111.
The EOF of a bare silica capillary is not very stable and is greatly affected
by the adsorption of ions, especially proteins. Experimentally, a sudden decrease
in EOF was also experienced for unknown reasons. We used fused silica capillar-
ies coated with succinylpolylysine in experiments described in this chapter. Such
capillaries show very stable EOF.
The mobility of the complex is usually intermediate between those of its compo-
nents. In the case of protein-ligand interactions, the charge on the ligand is a
major source of the mobility change of the protein.
When electrophoresis of A is camed out in a solution of B, the microscopic
electrophoretic mobility of A should discontinuously change between pAand pAB
at the moments of dissociation and association with B. As a result, the observed
macroscopic mobility of A, p, should be an average of the two mobilities
weighted for the time fractions in which it migrates as the free molecule and as
Determination of Affinity Constants by CE 191
where t and to are the detection time of component A in the presence and the
absence of B , respectively, and t, and t,' are the detection time of an electrophore-
sis marker in the presence and the absence of B , respectively. The prime symbol
is attached to show the possible variation of electroosmosis between two runs.
The marker should not interact with B and need not necessarily be electrically
192 Shirnura and Kasai
neutral. Fluctuations in electroosmosis are canceled out by this equation and the
net mobility change of A caused by the interaction with B can be calculated.
8 9 10 12
Time (min)
able to produce a sufficient mobility change of the protein for analysis. We have
introduced the use of charged soluble polymers, such as succinylpolylysine, as
mobile matrices for affinity electrophoresis. We call the charged matrix bearing
ligands the "affinophore" and electrophoresis in its presence "affinophoresis"
[3,4]. The distinctive feature of affinophores is that the portion responsible for
specific binding and that for migration in an electric field are structurally sepa-
rated, i.e., an affinity ligand and a charged affinophore matrix. Therefore, once
a suitable ionic molecule is obtained as an affinophore matrix, the method is
applicable to many binding molecules by changing the affinity ligand.
The principle of affinophoresis was successfully applied to the mobility
change analysis of lectin-sugar interactions. For the determination of affinity
constants of pea lectin, a divalent sugar-binding protein, the use of a monoli-
ganded affinophore is straightforward. The polyliganded affinophores, used in
the previous applications, should interact with such a divalent protein in a more
complicated manner [ I l l . N-Succinylated glutathione was used as an anionic
matrix for a monoliganded affinophore [lo]. The affinity ligand p-aminophenyl
a-D-mannoside was iodoacetylated at its amino group and coupled to the thiol
of the matrix (Fig. 2A). The intermediates and the affinophore were easily purified
by using reversed-phase HPLC. The affinophore caused a mobility change of pea
lectin in a concentration-dependent manner (Fig. 2B). Pea lectin barely migrated
under the present electrophoresis conditions. The interaction with the negatively
Figure 3 A linear plot for the affinophoresis of pea lectin. The plot was made using
the data in Fig. 2B according to Eq. [5]. The slope is the negative value of K, (0.199
mM) and the intercept with the ordinate gives Ap,,,, (-5.35 X cm2 V - ' s-' ). (From
Ref. 10.)
194 Shimura and Kasai
charged affinophore increased its mobility toward the positive end, thus lengthen-
ing its detection time. The result of the affinophoresis was analyzed according
to Eq. (5) based on several assumptions, i.e., equivalency and independency of
the two binding sites of the dimeric lectin, and the same mobility change for the
successive binding of the two affinophores (Fig. 3). The determination of the two
parameters, Kd = 0.199 mM and Ap,,, = -5.35 x cm2 V-I s-' , for the
interaction between the lectin and the affinophore served the basis for the determi-
nation of the affinity constants of the lectin to neutral sugars by competition
experiments, using affinophoresis. It should be noted that Kdrepresents the reac-
tion of each binding site, i.e., a microscopic equilibrium constant.
A neutral sugar to be investigated was added only in the positive electrode
buffer and the affinophore only in the capillary. Since the neutral sugar does not
migrate electrophoretically, it is transferred to the negative end only by EOF
faster than the lectin under the affinophoresis conditions, where the lectin mi-
grates electrophoretically toward the positive end. With the affinophore (B) at a
Time (rnin)
constant concentration, the macroscopic mobility change (Ap,) of the lectin be-
came smaller as the concentration of the neutral sugar (I) increased (Fig. 4). The
competition analysis is basically the same as that in enzyme kinetics. It can be
considered that the K, value in Eq. (4) increased by a factor of 1 +
[I]/Ki than
that without I in an identical relation to Eq. (4), i.e.,
[Bl
Api = W m a x
Kd app + [Bl
The K, ,,,,
an apparent dissociation constant, is in a relation with the K, as
Kd = Kd(l + []]/K,), where K, is the dissociation constant of pea lectin for
I. Since Apmaxhas already been previously obtained, K,,,, can be determined by
the measurement of Ap, according to Eq. (7). The plot of Kdapp/Kd versus [I]
gave lines with a slope of 1/K, (Fig. 5). The K , values obtained by this method
Figure 5 Determination of the dissociation constants of pea lectin for neutral sugars
(I) through competition experiments in the affinophoresis. The affinophoresis of the lectin
was carried out as described for Fig. 2 except for the addition of various concentrations
of neutral sugars to the positive electrode buffer (100 yl). Apparent K, values (K,,,,) were
determined from the observed mobility change (AyJ, the concentration of the affinophore
used ([B]), and the Ap,,,, value using Eq. [7]. The results are plotted according to Kd,I
Kd = [I]/Ki + 1.@, p-aminophenyl a-D-mannoside (0.25 mM); 0, methyl a-D-mannoside
(0.65 mM); A, D-mannose (1.6 mM); A,maltose (2.1 mM); W, methyl a-D-glucoside
(2.4 mM); 0, D-glucose (3.6 mM), where the Ki values determined are given in parenthe-
ses. (From Ref. 10.)
196 Shimura and Kasai
agreed well with those obtained by calorimetry. For example, Ki for methyl a -
mann no side was found to be 0.65 mM and was reported to be 0.61 mM by
calorimetric measurement. For D-mannose, it was found to be 1.6 rnM and was
reported to be 1.3 mM, calorimetrically.
A B
Time (min)
Figure 7 Plots for the determination of the dissociation constants of Con A for neutral
sugars (I). Prior to the analysis of neutral sugar-Con A interaction, the affinity probe-Con
A interaction was characterized by analysis according to Eq. [5] and K, and AF,, were
determined to be 15.5 FM and 1.20 X cm2 V-' s-'. When a neutral sugar was added
to the Con A solution, the mobility change of the affinity probe was reduced, as the result
of the reduction in the number of the free binding sites of Con A available for the interac-
tion with the affinity probe, as shown for methyl mannoside in Fig. 6B. From the reduced
mobility change, the equilibrium concentration of free binding site of Con A ([Con A],
corresponding to [B] in Eq. [4]) in a mixture with the neutral sugar (I) was calculated
according to Eq. [4] using the K, and AF,,, values determined previously. The determina-
tion of [Con A] allowed the calculation of [Con A-I] and [I], since the total concentrations
of Con A and I are known. The plot of [Con A-]]/[Con A] vs. [I] gave lines with slopes
of 1 IK,, where K, = ([Con A] X [I])/[Con A-I]. The plots and the determined values of
K,'s are: (a) methyl a- mann no side (0.104 mM); (b) mann nose (0.89 mM); (c) D-glucose
(4.5 mM); and (d) D-galactose (1.1 M). (From Ref. 12.)
Determination of Affinity Constants by CE
with a manually operated instrument 1121 and 6% (n= 5) with a fully automated
instrument ( 101.
VII. PROSPECTS
REFERENCES
11. Shirnura K, Kasai K. Capillary affinophoresis of pea lectin with polyliganded af-
finophores: A model study of divalent-polyvalent interactions. Electrophoresis 1998;
19:397-402.
12. Shimura K. Kasai K. Determination of the affinity constants of concanavalin A for
monosaccharides by fluorescence affinity probe capillary electrophoresis. Anal Bio-
chern 1995;227: 186- 194.
13. Harrison DJ, Fluri K, Seiler K, Fan Z, Effenhauser CS, Manz A. Micromachining
a miniaturized capillary electrophoresis-based chemical analysis system on a chip.
Science 1993;261:895-897.
Displacement Chromatography
of Biomolecules
Ruth Freitag
ETH Lausanne, Lausanne, Switzerland
I. INTRODUCTION
Time
Time
Time
In the meantime, the question of what constitutes the most versatile approach to
biotechnical downstream processing in the kilogram to ton range awaits its final
answer. Especially at larger scale displacement chromatography, a method
whereby the components are resolved into consecutive zones of the pure and
highly concentrated substances (Fig. 2), may become a serious competitor to
overloaded elution chromatography in the establishment of high-resolution, effi-
cient biopolymer isolation schemes.
Figure 3 Cases of equilibrium isotherms: (A) linear isotherm, (B) favorable nonlinear
isotherm, (C) unfavorable nonlinear isotherm.
206 Freitag
I I
elution volume
Initial condition:
(2)
ci (0, z) = 0 0 5 z 5 L i = 1,2, . . . , n
and
with a and b as substance-specific constants that can be calculated from the single-
component Langmuir isotherms.
This approach is often a good approximation, since many experimentally
recorded isotherms can be fitted to the Langmuir equation. However, the multi-
component Langmuir isotherm model is less useful in biopolymer chromatogra-
phy for reasons given in Section 111.
The effective use of the stationary and mobile phase capacity is among
the most obvious advantages of displacement over elution chromatography in
preparative separations. Contrary to affinity chromatography, several substances
can be purified simultaneously, which is clearly an advantage whenever the feed
contains more than one substance of value. Another major advantage of displace-
ment chromatography, especially in the context of large-scale biopolymer chro-
matography, stems from the fact that the feed, the displacer, and the regenerant
are introduced as simple step functions. Displacement chromatography is there-
fore much easier to operate in a continuous manner than overloaded gradient
elution chromatography. For example, de Carli et al. succeeded in operating a
continuous annular chromatograph in the displacement mode [3].
Displacement Chromatography of Biomolecules
Column
Turbulencies in
'mixing chambers"
/- (injector, detector)
T' Detector
Extraparticular effects
B
Molecular diffus~on Eddy diffus~on
(dlfferent pathways)
Intraparticular effects
Figure 5 Continued.
The ideal model has a long tradition going back to Gliickauf and Tiselius in
the theoretical description of displacement effects. In spite of its acknowledged
limitations, it describes correctly the most important aspects of displacement
chromatography and the influence of parameters like the sample and displacer
concentration on a given separation. This is especially the case under highly
nonlinear conditions and for small molecules.
As outlined above, the goal of a displacement experiment is the separation
of the feed components into consecutive zones of the pure substances, i.e., estab-
lishment of the so-called isotachic state or displacement train. For this to happen
it is necessary that the component isotherms be favorable (convex upward). Most
authors assume Langmuir-type isotherm shapes with good results. According to
the ideal model, under these conditions the displacement train will always evolve
after a certain column length.
Under favorable isotherm conditions the front of a substance zone is self-
sharpening until, in the absence of modifying dispersive effects, a triangular
"peak" is formed, where the concentration jumps suddenly to the maximum
value (shock transition) (Fig. 6). This can be observed in overloaded elution chro-
matography for substances with favorable isotherms. In displacement chromatog-
Freitag
Figure 6 Change in the shape of a given substance's zone under increasingly overloaded
conditions in the case of a favorable isotherm.
raphy, the rear boundary of each zone will also be sharpened, since the normally
diffuse rear end will be pushed up by the sharp front of the next zone. Every
molecule that lags behind will immediately be displaced. The opposite is true
for a molecule that for some reason finds itself ahead of its own bulk zone. It
will be among molecules with a lesser stationary phase affinity than itself and
most likely will be retained until overtaken by its own zone.
Once the displacement train has been established, all substance zones move
at the speed of the displacer front, uD.According to the material balance argument
of DeVault 11 11, the velocity of the latter is given by:
UD =
uo
1 + @(A~DJAcD)
or
uD = uo
1 + @(~D/CD)
Since all zones move at the same speed as the displacer front,
,J, = ,J2 = ,Jg = ... = u D
Equation (6b) applies also to the various compounds, i.e., the ratio q/c for each
compound must be equal to qn/cn. An operating line with steepness qD/cDcan
be drawn to determine the concentration in the individual zones (Fig. 7).
The exact concentration within a given zone of the displacement train can
be calculated by equaling Eq. (6b) for the displacer and the compound and resolv-
ing for the compound's concentration. A "water-shed'' point can be determined
Displacement Chromatography of Biomolecules
Operatlng line
Displacer
6 4 isotherm
Gill C l ~ C~
Mobile phase concentration, c
rlr Displacer
Elution volume
where the operating line becomes tangential to the substance isotherm. In this
case, the rear end of the eluting peak will just be touched by the front of the
displacement train. Substances whose isotherms are not intersected by the op-
erating line elute ahead of the displacement train. The only experimental parame-
ter needed for the treatment of displacement chromatography within the herme-
neutics of the ideal model thus are the equilibrium isotherms of the relevant
substances and the isotherm of the displacer.
Concentration in the individual zones is determined by the component's
isotherm and the displacer concentration. The length of each zone depends on
the original amount of the substance in the feed. By changing the displacer con-
centration, one changes both the speed of the separation [uD = f(cD)] and the
concentration of all substance zones. The concentration in the original feed, on
the other hand, is of little importance. According to the ideal model, this is also
the case for trace components and highly diluted feeds. In practice, dispersive
effects will prevent trace components from reaching their theoretically predicted
concentration maximum; however, the application of displacement chromatogra-
phy to isolate and enrich trace components prior to detailed analysis has been
demonstrated in a couple of real-life applications (see Section V1.F below).
Diluted and complex feeds, which are constantly encountered in the case
of high-value bioproducts such as recombinant proteins from mammalian cell
cultures, are both enriched and concentrated by displacement chromatography.
The fact that a concentration plateau rather than a peak is observed can also be
advantageous. While concentration is often a major goal in the bioseparations,
sometimes unwanted effects such as aggregation, denaturation, or maybe only a
troublesome increase in viscosity are observed as a certain critical concentration
is surpassed. In the elution mode the peak maximum would have to stay below
this critical value. Consequently, the concentration of the pooled fraction would
be considerably lower. In displacement chromatography the entire substance zone
could be kept just below this value resulting in a much higher average concentra-
tion in the pool. In at least two published applications, displacement chromatogra-
phy was used in the downstream process with the specific aim of keeping the
product concentration below the critical level [ 12,131.
Under the ideal conditions defined above, the displacement train will even-
tually form. The parameters of this train can easily be calculated given the equilib-
rium isotherms of the involved substances. Nothing has so far been said about
the developing train or the determination of the actual length required for the
development. Both can be calculated based on the ideal model provided the sys-
tem is characterized by Langmuir-type isotherms. Based on these models,
Gliickauf analyzed displacement phenomena and discussed effects of solute and
displacer concentrations as early as 1935. In the late 1960s, Helfferich et al.
presented the first algorithm for a mathematical description of displacement chro-
Displacement Chromatography of Biomolecules 215
Column Length, cm
0 10 20 30 40 50 60 70
agreement with the experimental results is good. The model has since been ex-
tended to the description of immobilized metal affinity chromatography (IMAC)
WI.
Time
Figure 10 Calculation of the column plate height from experimental results (linear chro-
matography).
Displacement Chromatography of Biomolecules 219
100 I/
1
1
I, min
0 -
0 5 10 1 25 30 1. min 35 .
are merely additions to the dominating axial dispersive effects. As we have also
discussed, this assumption is not always allowed for biopolymers, since the mass
transfer resistance is often several orders of magnitude higher in the case of these
large molecules (small diffusion coefficients). In contrast, certain types of chro-
matography popular in biopolymer separation are characterized by slow sorption
kinetics. Affinity chromatography, for example, quickly becomes surface reaction
limited, which may become a problem even in elution chromatography at elevated
Displacement Chromatography of Biomolecules 221
flow rates. In cases like this, the equilibrium-dispersive model is not capable of
providing helpful simulations. Instead a kinetic model should be used.
In the kinetic models the mass balance equation [Eq. (I)] is combined with
a kinetic equation relating the rate of variation of the concentration of each com-
ponent in the stationary phase to its concentration in both phases and to the equi-
librium concentration in the stationary phase. Solutions to the equations of the
kinetic models are usually obtained numerically as for the equilibrium-dispersive
model. Various models have been proposed, which vary mainly in the choice of
the kinetic rate expressions (see table below).
An important decision concerns which degree of complexity is necessary
in a given situation. As long as the kinetics are not very slow, the various kinetic
models yield similar results, which often resemble the results of the equilibrium-
dispersive model [24]. As the influence of the kinetic terms increases, the various
models yield results of differing accuracy.
Theoretically, a general rate model that explicitly considers all effects will
be the most correct. However, it is also the most complex one and requires the
experimental determination of all involved rate constants. In most cases, how-
ever, a kinetic model that lumps all kinetic effects into a single expression will
suffice.
For increasing mass transfer resistance, such a lumped model predicts a
decrease in the length of the zones in the displacement train and a concomitant
increase in the shock layer thickness. For very small mass transfer coefficients
the plateaus disappear completely and the resolution between successive bands
222 Freitag
with Kd = kA/(l + bdcd'),kl is the film mass transfer coefficient, and 9 is the
characteristic parameter (see Fig. 12 for details).
The shock layer thickness thus depends on the axial dispersion coefficient
and the mass transfer coefficient of the two components, on their separation fac-
tor, and on the concentration and retention factor of the displacer [34]. In a given
train the shock layer thickness depends only on the value of a. It does not depend
on the retention factor or the feed concentration of the components.
C',
Figure 12 Schematic presentation of the shock layer concept. (From Ref. 23.)
Displacement Chromatography of Biomolecules 223
A differentiation Eq. (9) shows that the shock layer thickness would be at
minimum for Kd = 1, i.e., for
k: = 1 + bdcd (104
These predictions are qualitatively correct for any isotherm type provided
its shape is convex and no intersection occurs. The latter, however, is quite a
common phenomenon in biopolymer chromatography, since the adsorption en-
ergy tends to increase with molecular mass, while the saturation capacity de-
creases in the same direction. A large molecule will therefore be characterized
by an isotherm with a steeper initial slope but a lower saturation plateau than a
similar but smaller molecule. The result may be an isotherm crossing leading at
worst to "elution azeotropes," which no column despite its length will be able
to dissolve [38]. Obviously this cannot be accounted for by the competitive
Langmuir-isotherm model, which assumes constant separation factors [7].
Other models such as the LeVan and Vermeulen isotherm derived from
the theory of the ideal adsorbed solution (JAS) [39] and modified Langmuir mod-
els [40] have been suggested to describe the multicomponent adsorption behavior
of complex molecules under linear and nonlinear chromatographic conditions.
Antia and Horvath have used this theory as a basis for their investigation of the
selectivity reversal phenomenon [38]. Langmuir-type single-component iso-
therms were also assumed in their case. They were able to show that the result
of an isotherm crossing is the development of a separation gap in the system
(Fig. 13). The position of the gap depends on the saturation capacities of the two
compounds. If the operating line crosses the isotherms in this gap, no separation
takes place. If it intersects on the right- or the left-hand side of the gap, a separa-
tion of the two compounds by displacement is possible. However, the order of
the substances in the displacement train is opposite in the two cases. If the initial
Freitag
Displacement Chromatography of Biomolecules 227
slope of the isotherm A is higher than that of substance B, whereas the opposite
is true for the saturation capacities, A appears before B in the displacement train
if the operating line intersects on the right-hand side of the separation gap and
B before A if the points of intersection are placed to the left of the gap. The
occurrence of an adsorption azeotrope has been observed experimentally, e.g.,
by Carta and Dinerman for the separation of a-aminobutyric acid and isoleucin
on Dowex 50W-X8 resin [41] and by Kim and Crarner for protein separations
in the immobilized metal affinity chromatography (IMAC) mode 1421. A similar
reason was proposed by Kasper et al. for their inability to resolve a mixture of
antithrombin I11 and bovine serum albumin (BSA) on hydroxyapatite columns
under certain experimental conditions [43].
A. Stationary Phase
Choosing the stationary phase also involves choosing the interaction mode for
the separation. The vast majority of the published protein displacement separa-
tions has been done on an ion exchange column. Displacement separations of
peptides and other smaller biomolecules are mostly carried out in the reversed-
phase mode. In these standard cases a host of guiding examples can be found in
the pertinent literature; a number of the more recent ones will be discussed below
in Section VI. Occasionally other stationary phases have been used, including
immobilized metal affinity chromatography (IMAC), antibody exchange (Abx),
hydroxyapatite, or hydrophobic interaction chromatography (HIC) phases (see
Section VI for details).
Today's high-performance stationary phases are in general not designed
for displacement chromatography. If anything, the differences between stationary
phase materials and columns from different suppliers is even more pronounced
in displacement than in elution chromatography. It is thus highly advisable to
investigate a number of columns before coming to a final decision. Some attention
should be paid to the availability of the bulk material. The column length is an
Displacement Chromatography of Biomolecules 229
Figure 14 Types of stationary phase materials with supposedly improved mass transfer
features: (A) perfusion material (Perfusion chromatography uses a bead with 2 types of
pores; throughpores: 6000-8000 A; diffusive-pores; 500-1500 A; throughpores enable
the eluent to pass the beads; diffusive pores are short and allow faster separation times.)
(9) hyperdiffusion material, (C) continuous bed column. Continuous Bed matrix unifor-
mity minimizes the band broadening seen in conventional packed beds. The non-porous
surface allows extremely fast mass transfer, minimizing band broadening even at high
flow-rates. The fimbriated surface structure of the nodules provides a large surface area
for good binding capacity.
7
Figure 14 Continued ,
6. Mobile Phase
In displacement chromatography the mobile phase is often merely considered as
an inert carrier. Concomitantly the use of conditions that aid strong binding of
the substances and the displacer to the stationary phase are advised. However,
this has not been shown to improve the separation. Instead, low retention factors
may actually improve displacement separations. The theoretically predicted opti-
mum values lay between 1.2 and 2.0 [46]. The influence of the separation factor,
a,on the shock layer thickness between two consecutive substances in the dis-
placement train should be kept in mind. A high value of a helps to achieve good
resolution between consecutive zones. Given the tendency of biopolymers to
show all-or-nothing binding behavior, the deciding factors in choosing the mobile
phase in biodisplacement chromatography are often less chromatographic and
more biological in nature, i.e., prevention of denaturation, interactionlaggrega-
tion, or solubility.
velop and conditions of nonlinearity should prevail during the separation. For
any given sample size there is an optimum column length and vice versa. Dis-
placement chromatography of proteins has been shown on columns as short as
5 cm. Extending the column length beyond the optimum will not improve the
separation any further. Often it is easier to adjust the sample size to the dimen-
sions of a given column rather than optimize the column length for a given sam-
ple. For modern high-performance columns the utilization of up to 80% of the
column's capacity should be possible.
Finally the question to be addressed is whether the full development of the
displacement train is always necessary. While this does maximize the recovery
yield, it does not necessarily correspond to the highest productivity. Throughputs
may, for example, be higher when the column is not long enough to allow the
full development of the displacement train and instead the mixed zones are recy-
cled. The decision will depend on the position of the target molecule in the dis-
placement train and the stability of the molecule.
D. Flow Rate
The optimum flow rate in displacement chromatography will be up to one order
of magnitude lower than in elution chromatography. In almost all cases of bio-
polymer displacement chromatography, this optimun~flow rate will therefore be
much too low to be of practical relevance. Most authors use flow rates between
0. I and 0.5 mllmin in biopolymer displacement chromatography in the semiprep-
arative scale (typical column dimensions cm X mm). At higher flow rates, the
quality of the separation tends to decrease rapidly, although flow rates of several
n~illilitersper minute have occasionally been used with success even with analyti-
cal scale columns [48].
The lower flow rates in displacement chromatography do not necessarily
result in lower throughputs or in an economic disadvantage. In our laboratory,
for example, identical columns were used in the displacement and the overloaded
elution mode for the separation of whey proteins. The preparation of I g of a-
laktalbumin of comparable purity took 5 times as long in the elution than in the
displacement mode in spite of the one order of magnitude higher flow rate in the
elution column (1 mllmin versus 0. I mllmin). The fact that the sample volume
was much smaller in the elution case was largely responsible for this. In addition,
the two whey proteins were concentrated by a factor of 3 in the displacement
case, whereas dilution was observed for the pooled fractions collected from the
elution column. A similar observation was made by Gerstner 1493 for the separa-
tion of oligonucleotides by displacement and elution chromatography. Although
the flow rate was twice as high in the elution than i n the displacement mode and
the feed per run similar (with 15 L the elution column was 3 times as large
as the displacement column), 31 1 runs were required to produce 5 kg of product
in the displacement mode versus 467 runs in case of the elution column. The
Displacement Chromatography of Biomolecules 233
E. Displacer
The preceding subsections dealt with aspects of displacement chromatography,
which are not very different from similar considerations in elution chromatogra-
phy. This is not so in the case of the displacer, which is a unique feature of
displacement chromatography. At the same time, the choice of the displacer has
consequences not only for the success but for the economic soundness of the
final method.
The ideal displacer should have the following features:
It should be nontoxic, stable, detectable, and cheap.
It should combine high solubility in the carrier with a high binding tendency
toward the stationary phase.
Regeneration of the column should nevertheless be possible.
In addition, the displacer should be uniform and its removal from the product
zone possible. For pharmaceutical applications it might even be necessary to
sterilize the material.
Mixtures of small biologicals such as amino acids, peptides, and small pro-
teins (antibiotics, insulin) are usually processed by reversed-phase displacement
chromatography. Hydrophobic substances such as 2-(2-butoxyethoxy)ethanol
(BEE), decyltrimethylammonium bromide, cetyltrimethylammonium bromide
(cetramide), benzyldimethyldodecylammonium bromide, dodecyloctyldimethyl-
ammonium chloride, and palmitic acid are standard displacers for these applica-
tions [2]. Nucleotide and nucleoside mixtures have been separated using a similar
combination between reversed-phase stationary phases and hydrophobic dis-
placers. The separation of oligonucleotides is also possible in the anion exchange
mode using dextran sulfate as displacer [49].
Ion exchange chromatography is very popular in preparative protein chro-
matography because it is known to preserve biological activity to a high degree.
To this day the majority of protein purification schemes contains one or several
ion exchange steps. While hydrophobic interaction chromatography is gaining
ground in preparative elution chromatography, the ion exchange mode is still
almost exclusively used in protein displacement chromatography. The lack of
suitably hydrophobic protein displacers for the hydrophobic interaction mode
may be among the reasons for this, since it is easier to find a highly aqueous
mobile phase-soluble polyion than similar hydrophobic polymer. For ion ex-
change applications, a number of (semi)synthetic polyions such as chondroitin
234 Freitag
Among the host of synthetic and semisynthetic substances that have been
used for protein displacement, few if any have been synthesized with that explicit
goal in mind. Torres and Peterson were among the first to chemically modify
their (high molecular mass CM-D) displacers in order to gain control over the
stationary phase affinity [62]. An interesting approach to displacer design was
recently suggested by two groups. These synthetic displacer molecules mimic
certain features of biopolymers (proteins) such as their solubility behavior in
aqueous solutions. The triblock polymethacrylate-based displacers prepared by
Patrickios et al. [63] using group transfer polymerization carry a sequence of
positively charged groups at one end of the molecule and a sequence of negatively
charged ones at the other. In the middle a neutral, hydrophobic block is created
to separate the two. The resulting polymers show an isoelectric point much like
that of proteins and can be precipitated at the corresponding pH. The authors
claim good results with these substances as displacers for anion exchange dis-
placement chromatography.
A similar approach to the problem of displacer recovery, either to rid prod-
uct zones from contaminating displacer traces or to establish a recycling scheme,
was used by Vogt and Freitag [64]. In this case a copolymer was synthesized by
radical polymerization that showed a lower critical solution temperature (LCST),
a phenomenon that is based on a similar physicochemical basis as the salting-
out effect observed for many proteins. Due to a finely tuned balance between
hydrophobic and hydrophilic groups, LCST polymers are water-soluble at low
temperature, but precipitate rapidly when the temperature is increased and a cer-
tain critical temperature, the LCST, is passed. The exact value of the LCST can
be adjusted from between 10°C and 90°C, whereas the displacing character of
the molecules stays roughly the same. The polymers can be redissolved simply
by lowering the temperature again. The cycle can be run through several hundred
times; no unspecified protein coprecipitation was observed in the investigated
cases. The displacers were used in combination with anion exchange and hy-
droxyapatite columns.
A. Protein Separation
C. Antibiotics
D. Isomers
Displacement chromatography has been repeatedly used by Vigh et al. to separate
optical and structural isomers at high throughputs and concentrations [83]. A
series of homologous displacers of varied affinity for Cyclobond I1 columns (a-
cyclodextrin silica) was introduced by the same group in 1995 [84].
H. Miscellaneous
VII. CONCLUSIONS
VIII. APPENDIX
A. Definitions
ldeal chromatography ldeal chromatography assumes infinite column effi-
ciency, i.e., all possible band broadening effects (mass transfer, reaction kinetics,
axial dispersion) are neglected. The two phases are constantly at equilibrium.
Models based on ideal chromatography allow, for example, study of the influence
of equilibrium thermodynamics alone on a chromatographic separation.
Linear chromatography Linear chromatography is characterized by a lin-
ear isotherm, i.e., the equilibrium concentration of a component in the mobile
phase and the amount adsorbed onto the stationary phase are directly proportional
at all times. The mass transfer processes are also linear. No heat or sorption
effects occur. Parameters such as the viscosity of the fluid phase are independent
of the composition. The mass flow rate is also constant. The sample concentration
has no influence on the peak shape (Gaussian) or the retention time. Both the peak
area and the peak height are proportional to the sample concentration. Analytical
chromatography usually approximates linear conditions.
Nonideal chromatography Nonideal chromatography allows for band
broadening effects, such as axial dispersion, mass transfer and reaction kinetics,
molecular and eddy diffusion. Increasingly complex models have been developed
that take into account some or all of these effects.
Displacement Chromatography of Biomolecules 249
B. Symbols
substance-specific constant (Langmuir isotherm)
substance-specific constant (Langmuir isotherm)
mobile phase concentration, displacer
mobile phase concentration
diffusion coefficient
bulk axial dispersion coefficient (equilibrium-dispersive model)
particle diameter
height equivalent to a theoretical plate (HETP)
Step function
equilibrium constant (KD = kb/(l + bucD)
capacity factor (t, - to)/to
film mass transfer coefficient
column length
apparent plate number of a given column (N = HIL)
stationary phase concentration, displacer
stationary phase concentration
velocity of the displacer front (shock layer)
linear flow velocity (mobile phase)
time
retention time of a solute (usually approximated as the retention time
of the peak maximum)
retention time of an inert tracer (column holdup time)
dimensionless column length
shock layer thickness
separation factor (k;/kj)
phase ratio
characteristic shock layer parameter (see Fig. 12 for details)
duration of feed introduction
C. Abbreviations
Abx antibody exchange
BEE 2-(2-butoxyethoxy)ethanol
1
250 Freitag
REFERENCES
Freitag
I. INTRODUCTION
The adsorptive or interactive forms of HPLC are clearly the most important
ones for users in biotechnology and related fields. In these cases an adsorption-
desorption respectively a partitioning reaction takes place between the substances
to be separated and the sorbent (stationary phase). These interactions can be of
an electrostatic, hydrophobic, but also highly specific (biological complementary
type) character, as indicated by the respective names, such as ion exchange, re-
versed-phase, hydrophobic, pseudoaffinity, and affinity chromatography.
As mentioned above, HPLC is a most important tool for the analytical and
preparative separations of biomolecules, in particular proteins. The complex and
peculiar physicochemical character of these biopolymers has been a constant in-
centive to the development and deeper understanding of the methods that are
used for their analysis and isolation. t
0 : Protein molecule
Figure 1 Stages involved in the trensrer of an rtnalyte molecule from the bulk of thc
mobile phase to the sorbent surface, where the adsorptionldesorption reaction takes place,
and back into the bulk mobile phase. ( I ) Diffusion from the bulk mobile phase through
!he stagnant film around the particle to the outer particle surface; (2) molec~~lar
diffusion
into the pore; (3) adsorption; (4) desorption; (5) molecular diffusion Crorn thc inner pore
surface through the pore space to the outer surface (pore mouth); (6) diffusion through
the stagnant film into the bulk mobile phase.
rous structure. The big pores with a size comparable to the size of the channels
formed by the interparticle space allow mass transport into the particle by convec-
tion rather than diffusion. The channels "divide" the particle into smaller seg-
ments, which is an important factor for the decrease of the distance to be covered
by diffusion by a molecule on its way to the adsorptive surface. Thus the mass
transfer is much more rapid in perfusion than in conventional porous supports.
The second type of pores, which also exists in the perfusion particles, consists of
non-flow-through channels of much smaller diameter and was called "diffusion"
pores by the inventors. It is implied that the majority of the adsorptive surface
is located in the diffusion pores and that the separation takes place there as well,
in accordance with the adsorption-desorption mechanism. Thus, the major ad-
vantage of perfusion chromatography is the improved mass transfer of the sub-
stance from the mobile phase to the stationary one [lo-141. The enhancement
is due to the fact that the solubilized molecules are transferred through the station-
ary phase particle (site of adsorption-desorption) by convection rather than by
molecular diffusion.
Membrane chromatography is often linked or compared to perfusion chro-
matography because some superficial resemblance exists. In membrane chroma-
tography the mobile phase flows through the sorbent layer or, more correctly,
through the single "particle," just as in perfusion chromatography. How-
ever, these two methods cannot be described with the same mathematical equa-
tions. The main difference is that in membrane chromatography the adsorption-
desorption process takes place on the walls of flow-through channels under the
conditions (pressure, shear stress, etc.) defined by the flow rate of the mobile
phase. There are no diffusive pores. Rather than as a column, the membrane can
be imagined as a bundle of capillaries of different diameters but of the same
extremely small length. In membrane chromatography the step of intraparticle
diffusion is absent. So there is no necessity of considering any migration into
the particle by molecular diffusion followed by the adsorption on the inner parti-
cle surface. From the many equations describing diffusional effects in conven-
tional HPLC, only a single one remains relevant in membrane chromatography.
This is the diffusional migration through the boundary layer of liquid close to
the walls of pores. Another important difference between the theory of membrane
chromatography and that of perfusion chromatography is that the desorption act
itself is not considered important in the latter and that perfusion chromatography,
just as conventional chromatography, is modeled by assuming repetition (and
accordingly, averaging of the parameters) of the adsorption-desorption process
as the separation evolves along the column [12-141.
A pronounced improvement of the mass transport kinetics was also ob-
served in another type of HPLC column where totally nonporous particles with
bonded adsorptive phase on their surface were used [15,16]. This type of station-
HPMC of Proteins 261
For practical reasons, the logarithmic form of this equation is normally used:
where M and S are constants defined by the nature of the protein and Q is a
variable value representing the modifier concentration in the eluent.
A very thorough experimental and theoretical investigation of this relation
was camed out by Regnier and coworkers [39-411. In these papers, a theoretical
retention model is constructed by applying the law of mass action to describe the
various interactions in the system, e.g., competition between protein and modifier,
interaction between protein and sorbent or sorbent and modifier. Unfortunately,
this model cannot be extended to the nonlinear range of the adsorption isotherm,
i.e., the region where most preparative chromatography occurs.
Upon analysis of the various equilibria and writing the corresponding equa-
tions, the authors come to the following expression which relates the retention
factor to the concentration of the desorbing agent (modifier):
Since the separation of the substances according to our idea occurs by a one-
step act of adsorption-desorption (more exactly, the single step of desorption),
the initial distribution of the protein molecules at the adsorptive surface located
at the inner walls of the disc pores (channels) is most important. This distribution
is governed by the energy of interaction, which in turn is characterized by the
Z parameter. The desorbed molecules instantaneously leave their "landing
grounds" and are flushed from the disc by the flow of the mobile phase. In this
situation the Z parameter is not averaged as in the case of multiple adsorption-
desorption acts in a column.
For the chromatography over short distances (discs) it is important to exam-
ine the parameter as a function of time and, equivalently, space. The former
corresponds to a separation on a very short column with the efficiency of "one
theoretical plate" [42], while the latter acknowledges the existing of some defi-
nite "length of separation layer" [43,44]-terns that take into account the non-
stationary nature of the initial part of the process of gradient elution in a column.
It is useful to keep these terms in mind when choosing the gradient shape and
comparing it with the real length of separation layer, i.e., the thickness of
the actual disc. Unfortunately, it is not possible to give a full summary of the
published data, which could be interesting for the theory of HPMC. The inter-
ested reader is referred to some of the, in our opinion, most significant papers
[45-491.
ture into your column, then turn it up-side down and elute in your standard linear
gradient. Quite often you will see no difference in the resolution compared to
that obtained when the entire "separation length" is used. In this experiment the
operative thickness of the separation layer is defined by an amount of protein in
the sample.
The traditional understanding of (column) HPLC is that the experimentally
observed separation effect is accumulated during multiple acts of adsorption-
desorption. Within this theoretical framework, the effect of the initial step is
considered as too insignificant to deserve or require an individual evaluation. In
this context the theory of HPMC, as outlined below, may well engender a change
of mind. Unless indicated otherwise, the model described below interprets separa-
tions by disc chromatography as a one-act desorption process even in such cases
where the theoretical separation layer thickness does not exactly correlate with
the physical thickness of the chromatographic disc.
In our development we propose that expression (4) can indeed be used to
describe correctly the experimentally obtained equilibrium adsorption. For conve-
nience's sake we change to a single variable and write the expression for the
retention factor in a reduced form:
Note that the probability is normalized because the sum of the probability of
residence of the desorbed protein in the mobile phase and the probability of resi-
dence of the protein in the stationary phase Pa = k'/(l + k') needs to be 1. One
can correctly estimate the probability of residence of the protein in the mobile
phase at equilibrium as the integral over the probability of a transition of the
protein from the stationary to the mobile phase under nonequilibrium conditions.
The latter, in its turn, is defined as the derivative of P, over Y:
(7) = [ 7 p ( ~ ) d=~
2nlZ
sin (2nlZ)
Taking into account that Z >> 1, we obtain for the first moment:
From the last equation, an expression for the Z parameter can be derived:
Recently it was shown [57] that expressions (12) and (13) reflect a fundamental
property of the process described by Eq. (4).
Combining Eq. (12), for the first moment, with Eq. (7) shows that, given
the typical protein values of Z, the average modifier concentration necessary for
protein desorption is equal to that for which k' = 1. This concentration corre-
sponds to the x axis intercept of a plot of In k' as a function of y~ (Fig. 2). Whether
or not a protein separation according to Eq. (6) is possible depends on the differ-
ences in the x axis intercepts of the corresponding plots and thus only on the Z
parameters of the proteins [57]. Besides that, it follows that all proteins are sepa-
rated at the same k' value. This conclusion was reached before [43], but in another
context.
Since the dispersion can be calculated from Eq. (14) and the Z parameter
from Eq. (15), it follows that the elution peak of a given protein seen after a one-
step desorption process is mathematically described by the probability density, p,
. obtain a more accurate expression for Z would necessitate a
as a function of y ~ To
measurement of the peak shape of the same protein passed through the chromato-
graphic layer in the absence of adsorptive interaction.
It was shown [58] that for processes describable by Eq. (4) or the corre-
Tennikova and Freitag
0
log 11D
Figure 2 Graphic dependence of the fundamental expression of interactive chromatog-
raphy of proteins. The lower "pole" of the crossed straight lines correspond tu the mem-
brane separation [59], whereas the upper ones reject the RP-HPLC results using a differ-
ent kind of displacer 1401.
plot of published experimental data (from Refs. 40 and 59) according to Eq. (14)
yields straight lines. Figure 2 also shows some basic differences between column
and disc protein separations. Obviously, the pole is located at a much lower y
value in disc chromatography, while the distance between the x axis intercepts
(a measure for the resolution) are wider. The latter fact indicates that HPMC may
well be superior in terms of separation power.
where F is the volumetric flow rate, A is the cross-section of the disc, and E is
the porosity.
Equation ( 1 6) does not reflect the fact that the flow velocity in the individual
pores may not necessarily be equal to the overall velocity U, since it will depend
on the size of the pores. From the Hagen-Poiseuille law, we know that the flow
through a tube is proportional to the square of its diameter:
where AP is the pressure drop along the liquid path and q is the dynamic viscosity
of the liquid.
Although Eq. (17) applies exactly only to the flow through a straight cylin-
drical tube of radius r and length L,, it was recently demonstrated that the flow
through a macroporous monolithic layer (disc) can at least be approximated by
this law [17].
HPMC of Proteins
which shows that the pressure drop per unit of linear flow velocity increases
exponentially with decreasing tube diameter. In other words, the larger the pore,
the lower the flow resistance, and the more liquid flows through this pore at a
given pressure.
As a result, the flow through the small pores is very slow and can even
approach zero because the overall back pressure within the cartridge, which
drives the liquid through the porous matrix, is lower than that required to enforce
a flow through the smaller pores. This uneven distribution of flow has the effect
of decreasing the surface area available for the adsorption-desorption process.
Therefore, membranes with a broad pore size distribution are not desirable be-
cause a considerable portion of the pores may not be open to the mobile phase
and thus not participate in the separation. The ideal matrix for disc chromatogra-
phy may thus be assumed to be characterized by large pores and a narrow pore
size distribution.
A very simple description of the effect of pores and pore size distributions
on chromatographic separations can be based on the residence time of the mobile
phase within the membrane tre,and the time scale tfilm of the mass transfer through
the film of stagnant liquid to the pore wall (adsorptive surface). In order to achieve
a good separation, t,, must be larger than t,,,.
The residence time is a simple function of the thickness of the separation
layer L and the flow velocity U:
In the first part of this chapter we tried to formulate the theoretical basis of
HPMC. Our goal was to define this evolving technique within the general frame-
work of adsorption chromatography, but also to convince the newcomer to the
field that it is indeed possible to separate protein mixtures over very short (mi-
crometer to millimeter) separation distances. In the second part of the chapter
we will illustrate the advantages of membrane chromatography using examples
drawn from the day-to-day laboratory practice. The application of HPMC will
be stressed, but other types of membrane chromatography will also be considered.
where w , , is the peak width at half height, t, is the retention time of the tracer,
and 6 is the thickness of the membrane, does not increase for flow rates between
0.1 and 30 mllmin. In spite of the fact that the plate height concept does not
fully apply to membrane chromatography it must be noted that the peak shapes
stay the same in disc chromatography independent of the mobile phase flow rate.
Taking into account that short separation layers cause less back pressure than
conventional columns, very fast separations should become possible in HPMC.
This is indeed the case for ion exchange membrane chromatography as Fig. 4
demonstrates. A mixture of three standard proteins could be resolved equally
HPMC of Proteins
0
0 5 10 15 20 25 30
Flow Rate [mUmin]
Figure 3 Dependency of the plate height, H, calculated from Eq. (22) as a function of
the mobile phase flow rate. Disc, Sartobind Q (Sartorius AG); tracer, lysozyme (1 mg/
ml).
well using mobile phase flow rates of 1 , 3 , 7 , and 40 mllmin, whereas the duration
of the separation was concomitantly reduced from 30 min to 30 s. (Fig 4a). This
is also hue for more complex separations such as that of human serum at 1, 5,
and 10 mllmin (Fig. 4b) [63] and that of a nucleotide mixture at 1 and 10 mll
min (Fig. 4c). This flow rate independence of the separation is not necessarily
found in all varieties of HPMC. While ion exchange chromatography is character-
ized by very fast surface interaction kinetics, other types, e.g., affinity chromatog-
raphy, show much slower reaction kinetics. If the flow rate is increased indiscrim-
inately in these cases, a loss in resolution can sometimes be observed [64], simply
because the separation becomes reaction-limited.
A second question concerns the effect of the flow rate on the capacity of
the disc. Here both the pore size distribution and the construction of the disc
cartridge (mainly the inlet where the incoming flow is distributed equally over
the entire cross-sectional area) are of major importance. While some discs can
be used at flow rates of more than 30 mllmin without significant loss in capacity,
others already lose more than 50% of the low flow rate capacity when the flow
rate is increased to 10 mllmin [65] (Fig. 5). Affinity discs seem again to be more
prone to a loss in capacity than the ion exchange types.
Once the advantage of HPMC over conventional column chromatography
in terms of fast and efficient protein separation has been realized, a number of
Tennikova and Freitag
-
p so
i
0
20 40
[Sek.]
Figure 4 Demonstration of the flow rate dependency of HPMC separations. (a) Separa-
tion of a standard protein mixture (cytochrome c, a-chymotrypsinogen, lysozyme, 1 mgl
ml each); disc, strong cation exchanger (prototype Sartorius AG). (b) Separation of human
serum proteins (total protein 3 mglml) on an anion exchange membrane (prototype Sarto-
rius AG). (c) Separation of a nucleotide mixture (NADINADH, 50 pglml; NADPl
NADPH, 150 pglml) at 1 mllmin and 10 mllmin on a strong cation exchange membrane
(prototype Sartorius AG). (From Ref. 63.)
r
HPMC of Proteins
Figure 4 Continued
Static Capacity
r Dynamic Capacity
0 5 10 15 20 25 30 35 40 45 50 55 60 65
Flow Rate [mllmin]
Figure 5 Influence of flow rate on capacity. (top) Static and dynamic antithrombin I11
binding capacity of a heparin affinity membrane adsorber (prototype Sartorius AG). (From
Ref. 65.) (bottom) Static capacity, strong anionlstrong cation exchanger (both prototype
Sartorius AG). The disc was loaded with human serum albumin until saturation. Afterward
the amount retained was eluted and quantified. (From Ref. 67.)
HPMC of Proteins
n j3-Galaktosidase
200
150
100
50
25
10
Medium
0. Preparative HPMC
Given the particular advantages of HPMC, this mode should also be applicable
to preparative separations. To date the majority of application protocols is given
for small discs (mm x cm range) (see below). Most applicants still tend to use
a single-step interaction mode, much in the tradition of affinity filtration, rather
than exploiting the full advantages of the new stationary phase geometry in chro-
matographic terms. Multistage separation processes are therefore still rare, al-
though they do exist [65,68].
Antithrombin I11 is an anticoagulant with considerable therapeutic value.
It has thus become a target substance for biotechnical production and several
mammalian cell lines exist, which express human antithrombin I11 as a recombi-
nant protein. The recovery of the product from the cell culture supernatant is
made quite complex by the fact that the product is normally found only in minute
amounts (in the case considered here typically 1% of the total protein concentra-
Tennikova and Freitag
Time [rnin]
Time [rnin]
Figure 7 (top) Chromatogram of a cell culture supernatant using a strong anion ex-
change membrane adsorber (prototype Sartorius AG). The product containing fraction is
indicated. (bottom) Electropherogram of the product containing fraction. (From Ref. 67.)
HPMC of Proteins 279
tion) and that large amounts of FCS are normally added to the production environ-
ment (here typically 10%). Lowering the serum concentration often yields a prod-
uct of inferior quality, e.g., low activity, low stability [67]. HPMC was used as
a cheap but high-performance preparative approach to process between 0.1 and
10 L of culture supernatant.
In a first attempt, heparin affinity HPMC was used in combination with an
ultrafiltrationldiafiltration step 1651. While the latter served mainly to concentrate
the protein fraction in the culture supernatant, we hoped to isolate pure antithrom-
bin I11 by the heparin affinity step. Column heparin affinity chromatography is
a standard procedure to isolate antithrombin 111, since the protein shows a pro-
nounced affinity for its cofactor.
The results of the first attempt are summarized in Table 1. Of the original
AT I11 activity, 81% was recovered by the heparin disc. In addition, the product
was concentrated by a factor of 101 during this step. When the protein was ana-
lyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immu-
nodiffusion its purity was established at 64%. Large amounts of both bovine
serum albumin and bovine transferrin as well as traces of bovine immunoglobulin
G and other unidentified proteins were still present. One hundred sixty minutes
was required for the processing of one batch of culture supernatant in this case,
corresponding to 0.7 mglh AT 111.
In order to improve the final purity, ion exchange HPMC steps were inte-
grated in the isolation scheme. It was possible to nearly remove the b-IgG by
a strong anion exchange disc, as was verified by immunoblotting. The final pur-
ity of the antithrombin was 85%, with bovine serum albumin as the major con-
taminant.
Since none of the procedures was capable of removing the bovine serum
albumin totally, a further improvement of the product quality was attempted by
introducing a step for the specific removal of the albumin. Cibacron blue ligands
are known to interact with bovine serum albumin but not with antithrombin. It
was found that the Cibacron blue disc is best used before both the anion exchange
and the heparin disc (Table 2). A cycle required 300 min in this case and yielded
approximately 2 mg of product (0.4 mglh). The final procedure yields a 94%
pure product with a recovery of 72%. Purities of >99.9% were reached only
when the serum concentration in the culture was lowered to 3%. Then, however,
the biological activity of the product suffered [65].
In order to increase productivity, the flow rate over the discs was increased
stepwise from the 2 mllmin (0.12 Llh) used during development to 4.8 Llh. The
maximum flow rate through the heparin MA had to be restricted to 0.18 Llh
during these experiments, since affinity discs show a pronounced flow rate depen-
dency of the dynamic capacity. Nevertheless the cycle time was reduced consider-
ably by this increase of flow rate and the antithrombin throughput concomitantly
increased from 0.55 to 13.7 mglh (Table 3).
The scale-up of HPMC to fully preparative dimensions is also rarely done.
The easiest way to increase the scale of a HPMC separation is to increase the
number of discs involved, either by inserting several discs into a cartridge (mod-
em disc cartridges can usually incorporate varied numbers of discs) or by short-
circuiting several modules. Figure 8 shows such a scale-up by an increase in the
number of discs. By increasing that number from 1 to 6, the total amount of
trypsin inhibitor, a-chymotrypsinogen, and lysozyme separated could be in-
Anion
Cell culture Ultrafiltration/ exchange Heparin
supernatant diafiltration CB-MA MA MA
Table 3 Influence of the Mobile Phase Flow Rate on the Separation of Recombinant
Antithrombin 111 by the Three-Step HPMC Process
\ . . . . . . , . . . , . .
0 2.0 4.0 6.0
Time [min]
creased from 0.25 to 1.2 mg [63]. By keeping the flow rate and the gradient
volume constant, the retention times were kept equal. The resolution does suffer
slightly, since the protein zones are somewhat broader at higher concentration.
An interesting effect was observed when coupling modules to increase the
maximum sample load. When working close to the separation capacity (i.e., the
amount of protein that could still be resolved with a resolution of 1 or better)
the resolution was found to improve whenever two modules were used in series
even though the loading, i.e., the amount of protein per cm2, was kept constant
[69]. In other words, we find a higher separation capacity in the case of two
modules than for a single one. No further improvement is observed when more
than two modules are stacked. The improvement in separation is not due to the
increase in the number of disc layers or simply to an increase in total area, since
two consecutive smaller modules [total of six layers (2 X 3) representing 60 cm']
resolves a given protein mixture better than a single large one (five layers equal
to a total of 100 cm2).
While such an increase in the number of layers can often suffice to carry
a separation from the analytical to the semipreparative scale, a truly large-scale
membrane chromatography requires a more significant increase in area. In this
context HPMC can profit considerably from filtration theory, to which it bears
a certain resemblance. In fact, various forms of filtration have been adapted to
membrane chromatography, including cross-flow and hollow fiber modules. Fig-
ure 9 shows the scale-up of a separation of three proteins (a-chymotrypsinogen,
cytochrome c, lysozyme) from a small module (0.5 cm2) to a large one (130
cm2).The flow (ml/cm2)and the protein load (mg/cm2)were kept constant, while
the volumetric flow rate was increased from 0.2 to 50 ml/min. Given the difficulty
of recording a UV trace for the larger modules, the separation stayed much the
same.
The largest module (fitted this time with 10 layers, each 130 cm2) was also
used in the processing of technical dairy whey, where throughput is an important
point [69]. Five liters of feed solution was passed through this module at a flow
rate of 50 mllmin. Using a two-step NaCl gradient (up to 1 M NaCl), 248 mg
of protein was eluted within 14 min: 116 mg in step 1 (0.1 M NaCl, mostly a -
lactalbumin) and 132 mg in step 2 (0.5 M NaCl, mostly P-lactoglobulin). The
recovery was >70% for both proteins.
C. Mixed-Mode HPMC
Save for a few exceptions, such as hydroxyapatite chromatography or the
Bakerbond ABX phase (Baker Chemical Co., USA), which were especially de-
veloped for antibody purification, mixed-mode interactions are avoided in liquid
chromatography. On the other hand, single-stage chromatography is seldomly
sufficient for preparative protein separation and multistage procedures, where
HPMC of Proteins
several types of interaction are exploited in series are the norm. Controllable
mixed-mode phases may thus become an interesting tool. However, with conven-
tional porous particle-based stationary phases, mixed-mode supports are difficult
to realize in a reproducible, homogeneous, but flexible manner. The number of
(usually prepacked) columns to be offered by the suppliers would increase dra-
matically with the number of possible or desirable combinations. This is radically
different in the case of HPMC where the use of stacks of discs has already become
routine (see above). In this case even the most complex mixed-mode approach
calls only for the insertion of different discs into the cartridge. In fact, an approach
using discs with different interactive groups in one housing is currently being
promoted by a major HPMC supplier (BIA, Slovenia) under the name of combi-
natorial liquid chromatography (CLC).
That the separation by one type of disc is not disturbed by any other type
can be demonstrated in a simple experiment. Interactive discs, e.g., ion exchange
discs, are alternated with inert filtration discs and the chromatograms compared
with those obtained for a cartridge filled only with the ion exchange material. In
our experience there will hardly ever be a difference unless the putatively inert
filter discs are not really inert (in the case of proteins, hydrophobic interactions
may be present).
Figure 10 shows the separation of five standard proteins [(P-lactoglobulin
(IEP: 5.1), conalbumin (IEP: 6.8), a-chymotrypsinogen (IEP: 9.6), lysozyme
(IEP: 10.5), and cytochrome c (IEP: 11.2)] by mixed-mode ion exchange chroma-
tography together with the chromatograms of the single substances on the same
stationary phase [70]. Five strong anion exchanger and five strong cation ex-
changer membrane adsorbers (prototypes Sartorius AG) were alternated in the
cartridge. Note that the separation becomes possible at physiological pH with
the mixed-mode approach, since it is no longer necessary to go to extreme buffer
pH in order to ensure that all proteins carry the same net charge.
Alternating the discs gives a better resolution than using two stacks (Fig.
1 1 ) [70]. As always in ion exchange HPMC, the separation seems to be nearly
independent of the flow rate. Figure 12 shows again the separation of the standard
protein mixture on the alternating discs using flow rates between 2 and 12 mll
min [70]. The gradient volume and shape were kept constant during these experi-
ments. While the separation required 30 min at a flow rate of 1 mllmin it could
be performed in 1.6 min with a flow rate of 12 mllmin.
The mixed-mode approach is not restricted to mixed-mode ion exchange
HPMC. Mixed mode ion exchangelaffinity HPMC is also promising-especially
so because it combines the advantages of affinity chromatography, i.e., the ability
to be highly selective for the target molecule which is often also considerably
concentrated, with that of ion exchange chromatography, e.g., the ability to re-
solve the entire substance mixture. In mixed-mode ion exchangelaffinity HPMC
a single-step separation again becomes possible, which would otherwise have
HPMC of Proteins
required at least two steps [70]. We have seen above that the isolation of the
recombinant anticoagulant antithrombin 111 from cell culture supernatant requires
several chromatographic steps. The removal of bovine serum albumin and bovine
transfemn is especially difficult. The separation of the antithrombin I11 from these
two contaminants requires at least two chromatographic steps based on different
interaction principles. The albumin can be removed by Cibacron blue affinity
chromatography (Fig. 13a), which doesn't help to lower the bovine transfemn
concentration. Bovine transferrin, on the other hand, can be removed-due to
the difference in IEP-on a strong anion exchange chromatography (Fig. 13b).
However, this step does not remove the albumin. Only one possibility has been
reported to at least partially resolve the three-protein mixture and that was immo-
bilized metal affinity disc chromatography (IMADC) using Cu2+ions (Fig. 13c).
In this case, however, antithrombin activity is found in the breakthrough as well
as in one of the later eluting fractions.
By the simple measure of using controlled mixed-mode anion exchange1
affinity interaction, on the other hand, the separation of recombinant antithrombin
Ill, bovine serum albumin, and bovine transfenin becomes possible in a single
1
CHY
CYT LYS
CON
LJ
Alternating MA
\
0.0 20.0
Time [min]
mixed-mode HPMC separation at pH 7.0 (Fig. 13d). Since the bovine serum
albumin may be expected to interact mainly with the affinity membrane adsorber
(i.e., a Cibacron blue disc, prototype Sartorius AG) under these circumstances,
it is not surprising that a comparatively high NaCl step gradient (3 M NaCl) is
required for elution of that particular protein. The antithrombin I11 and the bovine
transferrin, on the other hand, which should adsorb mainly onto the anion ex-
changer MA, are eluted in a linear NaCl gradient up to 1 M NaCl.
VI. EXAMPLES
Of the various types of adsorption chromatography, the two most commonly used
types of HPMC in preparative protein separations are affinity chromatography
("affinity filtration") and ion exchange chromatography. In spite of the potential
HPMC of Proteins
2dmin
4 mVmin
6dmin
8dmin
10 mVmin
Figure 12 Influence of the mobile phase flow rate on mixed-mode protein HPMC.
(From Ref. 70.)
Albumin
_J
I I
0 10 20 30
Time [min.]
0 20 40
Time [min]
Figure 13 Separation recombinant antithrombin 111, bovine serum albumin, and bovine
transferrin. (a) Cibacron blue affinity membrane adsorber. (b) Strong anion exchange
membrane adsorber. (c) Immobilized metal affinity membrane adsorber (Cu2+ions). (d)
Mixed-mode HPMC, using the Cibacron blue membrane adsorber and the strong anion
exchange membrane adsorber. (From Ref. 70.)
HPMC of Proteins
Transferrin
Imidazol
lmMl
r 20
Volume [ml]
BSA
10.0 20.0
Time [min]
Figure 13 Continued
290 Tennikova and Freitag
A. Affinity Filtration
C. Therapeutic Proteins
Factor VIll from human Use of ion exchange discs Strancar et al. 1997 [86]
plasma (also for process monitor-
ing) and scale-up via the
use of a compact porous
tube made from the same
material as the discs
Monoclonal antibodies from Cation exchanger, scale-up Liitkemeyer et al.
cell cultures from disc to hollow fiber 1992 [87]
system, process up to har-
vest from 100 L bioreac-
tor, recoveries >96%
HPMC of Proteins 293
Human tumor necrosis Recombinant protein pro- Lukas et al. 1994 [24]
factor-a duced in E. coli, product
electrophoretically pure,
anion exchange HPMC
Recombinant antithrombin Ion exchange and heparin Liitkemeyer et al.
111, monoclonal antibod- affinity disc chromatogra- 1993 1881
ies from cell cultures phy, scale-up to hollow
fiber system (2400 cm2)
in case of ion exchanger,
coupling chemistry for he-
parin
Serum and plasma proteins Scale-up from 10- to 50- Josic et al. 1992 [22]
mm disc (weak anion ex-
changer), comparison to
column chromatography
Annexins from liver plasma Anion exchange disc, sepa- Josic et al. 1994 (231
membranes, monospecific ration of annexins from
polyclonal antibodies each other in 10 min,
comparison of disc and
cellulose fiber modules,
immobilization of an-
nexins to epoxy-acti-
vated discs for antibody
purification
Monoclonal antibodies from Thiophilic membranes, Finger et al. 1995
cell culture membrane stacks and
cross-flow module, inter-
pretation of the flux be-
havior according to filtra-
tion theory
Kistler and Nitschmann's Weak anion exchanger Lacoste-Bourgeacq et al.
fraction IV of blood 1991 [90]
plasma
Recombinant antithrombin Multistage disc chromatog- Reif and Freitag
111 from cell culture raphy process (affinity 1994 16.51
and ion exchange chroma-
tography), scale-up to 10
L scale
Whey proteins, permeate Comparison to column chro- Splitt et al. 1996 [69]
components matography, mixed-mode
approach, influence of
the process parameters
(flow rate, pH), scale-up
considerations
294 Tennikova and Freitag
D. Endotoxin Removal
The lipopolysaccharides, which are a major component of the cell membrane of
gram-negative bacteria, are powerful endotoxins that cause a violent reaction of
the immune system of mammals upon entering the bloodstream. Pharmaceuticals
and especially injectables need to be endotoxin-free. The removal of endotoxins
is not an easy task and chromatographic methods have been suggested. HPMC
may present itself as a fast and efficient detoxification (depyrogenation) method.
E. Process Monitoring
Investigation of the Mem- 30- and 15-mg of protein Gerstner et al. 1992 [95]
Sep system for analytical were purified within 3
and preparative applica- min when a cation ex-
tions change MemSep 1010
system was operated in
the step and linear gradi-
ent mode, respectively
Investigation of kinetics of Theoretical treatment, appli- Nachman 1992 [46]
immunoaffinity mem- cation of the results to
brane chromatography the isolation of biothera-
peutics
Model for membrane affin- Mathematical model for the Suen and Etzel
ity chromatography simulation and the design 1992 [96]
of dead-end affinity fil-
tration
Modeling of disc chroma- Model and experimental Briefs and Kula
tography verification for various 1992 [97]
limiting conditions, appli-
cation to protein purifica-
tion and scale-up
Investigation of mass trans- Development of a mathe- Sarfert and Etzel
fer limitations matical model to describe 1997 [62]
various processes during
protein separation by
HPMC, film mass trans-
fer resistance is found to
be the decisive factor
296 Tennikova and Freitag
G. Miscellaneous
VII. CONCLUSIONS
ACKNOWLEDGMENTS
The authors gratefully acknowledge Dr. Michael Tennikov for the fruitful discus-
sion,regarding the preparation of this chapter.
REFERENCES
HPMC of Proteins
I. INTRODUCTION
can be summarized as follows: (1) preparation of the appropriate gene; (2) incorpo-
ration of the gene into a vector; (3) transfer of the vector carrying the gene into
a recipient (host); (4) selection and cloning of the host cells carrying the gene;
(5) expression of the gene; and (6) extraction of the protein [5 I]. Each recombi-
nant protein has its own distinct chemical, physical, and biochemical properties.
In selecting an expression system for the production of a recombinant pro-
tein, the potential use of the protein is a determining factor. The size, complexity,
posttranslational modifications, and ease of production and purity of that particu-
lar protein are considerations in selection. Research into phenotypical changes
occurring in host cells necessitates an expression system closely imitating a pro-
tein's natural source [17]. The four major cell types currently employed for pro-
tein production include bacterial cells, yeast, insect cells, and mammalian cells.
When choosing which expression system to use, conservation of the biological
activity (proper folding) of the expressed recombinant protein as well as isolation
(purification) of this protein should be considered [17,63].
Escherichia coli and Bacteroides subtilis are the two most widely used
bacterial cell systems. The advantages of bacterial expression systems .are (1)
their simplicity and (2) their rapid and large yields at low cost. Bacterial cells
can also secrete the recombinant protein into the culture medium facilitating puri-
fication. One disadvantage is that the proteins often fail to fold properly, forming
insoluble inclusion bodies of biologically inactive proteins. While some small
proteins extracted from these inclusion bodies can be successfully refolded, most
larger proteins cannot. Bacterial cells also lack necessary enzymes for posttransla-
tional modifications required for proper functioning of the protein [17,63]. E.
coli was the expression system used to produce the first commercial recombinant
protein drug, human insulin 1181.
E. coli expression systems offer the option of fusion protein expression
[1,32,59]. Fusion proteins are a combination of the gene product of interest (usu-
ally eukaryotic) and a second "carrier" gene product (usually prokaryotic)
[32,63]. The fusion helps to stabilize the protein in the bacteria. The fusion protein
expression strategy allows good translation initiation, overcomes instability, and
yields a high degree of expression. The fusion partner also facilitates generic
protein purification methods and helps prevent formation of inclusion bodies.
The four main gene fusion expression systems in E. coli include Staphylococcus
protein A (SPA), Schistosomajaponicum glutathione S-transferase (GST), E. coli
maltose-binding protein (MBP), and E. coli thioredoxin [32].
Yeast can be grown as rapidly and economically as bacteria. Yeast can
also achieve high cell densities and allow secretion into the culture medium,
thereby facilitating purification. An advantage of yeast over bacteria is ability
to perform many posttranslational modifications found on human proteins. One
potential disadvantage in using yeast cells is the presence of proteases that de-
grade foreign proteins. Proteolytic degradation can be prevented by constructing
HPLC Purification of Recombinant Proteins 303
yeast strains in which the genes for these proteases have been deleted 117,631.
A subunit vaccine for hepatitis B virus (HBV) using only a surface protein to
initiate the immune response has been successfully produced in yeast [61]. The
major advantage is lack of infection risk.
A third, relatively new expression system uses baculovirus to insert foreign
proteins into insect cells. Advantages are abundant expression, correct folding,
and posttranslational modifications similar to those in mammalian cells. One dis-
advantage is the higher cost of culturing insect cells over that of bacteria and yeast.
Cost is still less than that for culturing mammalian cells [63]. A vaccine for the
autoimmune deficiency syndrome (AIDS) virus produced from a recombinant HIV
envelope protein by this method has been approved for clinical evaluation [3].
Finally, mammalian cells have been found to be the best place to produce
mammalian proteins. Mammalian cell systems are used for both transient and
stable expression. Transient expression is used to verify the biological activity
of engineered proteins and rapidly evaluate different vectors for the stable expres-
sion systems. In contrast, stable mammalian cell expression is more time consum-
ing but is necessary for large-scale production of proteins in mammalian cells.
The versatility of protein production possible from mammalian cells is the reward
for time and effort spent on the processes of selection, amplification, and growth
of stable cell lines. Thus far, there are no examples of proteins that cannot be
made in mammalian cells [13,63]. Tissue plasminogen activator (tPA) was the
first commercially available drug produced from a stably integrated mammalian
cell culture [48]. Factor VIII, a protein required for normal clotting of blood, has
also been efficiently produced in a stably integrated mammalian cell culture [45].
If an expression system conserves the biological activity of the recombinant
protein, the next consideration is whether this biological activity will be further
conserved following purification of the protein [53]. Currently, purification of
recombinant proteins generally utilizes the same purification techniques em-
ployed for separating other proteins from cell culture media. The large number
of proteins produced by the biotechnology revolution have also driven advances
in analytical techniques. High-performance liquid chromatography (HPLC) and
other analytical scale chromatographic techniques are essential to the achieve-
ment of the objectives of this revolution.
The techniques classically used for the purification and characterization of
proteins are ultracentrifugation, size exclusion, gel filtration, ion exchange, affin-
ity chromatography, electrophoresis, and radioimmunoassay 16,461. However,
these methods are generally time consuming and difficult to automate. The advent
of HPLC brought a new dimension to the purification and analysis of proteins. The
major advance was the use of microparticulate packing materials, which permitted
the use of high pressure. The cost of moving from traditional liquid chromatography
(LC)methods to HPLC was more than offset by improvements in speed, resolution,
sensitivity of chromatographic separations, and ease of sample recovery [ I 11. Ad-
304 Smith et al.
A large number of different fusion proteins have been isolated using a variety
of HPLC columns and solvent systems (Table 1). The following are illustrative
as there are many more examples.
Extracellular Klyushnichenko VE, Wulfson Insulin-containing proteins TSK G3000SW (300 X Reagents used = acetonitrile,
AN. Recombinant human insu- 7.5 mm) methanol, sodium phosphate,
lin-11. Size-exclusion HPLC phosphoric acid (all esp. pure),
of biotechnological precursors. water purified on Milli-Q, so-
Factqrs influencing retention dium dodecyl sulfate, guani-
and selectivity. Pure Appl dine hydrochloride.
Chem 1993;65(10):2265-2272.
Patrick JS, Lagu AL. Determina- Human proinsulin fusion protein TSK DEAE-5PW (anion Anion exchange-"Loading"
tion of recombinant human pro- (ChPI) expressed in recombi- exchange); TSK SP- mobile phase A = 20 mM
insulin fusion protein produced nant E. coli 5PW (cation ex- Tris in 7 M urea (pH 7.0).
in Escherichia coli using oxida- change); Zorbax GF- "Eluting" mobile phase B =
tive sulfitolysis and two-dimen- 250 (size exclusion) 20 mM Tris and 0.5 M sodium
sional HPLC. Anal Chem sulfate in 7 M urea (pH 7.0).
1992;64(5):507-5 11. Cation exchange-Mobile phase
A = 40 mM monobasic potas-
sium phosphate in 7 M urea
(pH 3.5). Mobile phase B =
0.5 M in sodium sulfate, 40
mM in monobasic potassium
phosphate, and 7 M in urea
with pH 3.5.
Size exclusion-Mobile A phases
used in ion exchange: (1) 20 V)
mM Tris in 7 M urea (pH 3
-
5
7.0) & (2) 40 mM monobasic s
potassium phosphate in 7 M (P
urea (pH 3.5). p
Gearing DP, et al. Production of Leukemia inhibitory factor (LIF) 300-A Brownlee C8 RP 5 min linear gradient to 40% ace-
I
leukemia inhibitory factor in [initially expressed as a fusion tonitrile in 0.1% TFA followed w
Escherichia coli by a novel
procedure and its use in main-
product with glutathione S-
transferase, purified on glutathi-
by a 60 min linear gradient to
60% acetonitrile in 0.1% TFA.
5
taining embryonic stem cells one-agarose affinity matrix and
released from the matrix by
2*
+
in culture. Biotechnology ii'
1989;7(11):1157-1161. cleavage with thrombin]
O'Keefe DO, et al. Use of mono- 1) transforming growth factor-a Hy-Tach nonporous CIS Linear gradient of 34-64% aceto-
3
bromobimane to resolve two re- Pseudomonas aeruginosa exo- nitrile in 0.1% TFA
combinant proteins by re- toxin A 40 (TGFa-PE4O). a re- 0,
versed-phase high-performance combinant protein synthesized
liquid chromatography based in E. coli
on their cysteine content. Chro- 2) PE40, an M, = 40,000 C-ter-
matography 1992;627(1-2): minal fragment of TGFa-
137-143. PE40, which copurifies with
-5'
TGFa-PE40.
Swoboda I, et al. Isofoms of Bet Proteolytic fragments of purified Waters pBondapak C18 Linear gradient of acetonitrile
v 1, the major birch pollen al- natural Bet v 1 (nBet v l ) and [solvent A, 0.1% (vlv) TFA in
lergen, analyzed by liquid chro- Recombinant nonfusion Bet v water; solvent B, 0.07% (vlv)
(n
matography, mass spectrome- l a (rBet v la) [Bet v 1 = ma- TFA in acetonitrile; 0-40% B
try, and cDNA cloning. J Biol jor allergen of birch pollen] in 120 min]
Chem 1995;270(6);2607-2613.
lntracellular Sharma SK, et al. Metal affinity Recombinant HIV-1 reverse tran- C4 (Vydac) RP. 300-A 0-70% gradient of 0.1% TFAI
chromatography of recombi- scriptase containing a human pore size, 4.6 mm X H,O to 0.1% TFAICAN in 40
nant HIV- 1 reverse tran- renin-cleavable metal binding 15 cm min
scriptase containing a human domain
renin cleavable metal binding
domain. Biotechnol Appl Bio-
chem 1991;14(1):69-81.
Iwakura M, et al. Dihydrofolate Chemically synthesized polypep- Inertsil-ODs 5-pm col- Equilibrated with 0.1% TFA in
reductase as a new "affinity tides of 5-44 amino acids. ex- umn (RP) 15% acetonitrile. Linear gradi-
handle." J Biochem (Tokyo) pressed in E. coli as fusion pro- ent of acetonitrile up to 50%.
1992;lll(I):37-45. teins which show dihydrofolate 0
4
reductase (DHFR) activity.
Table 1 Continued w
0
OD
Protein
classification Ref. Substance purified Column type Solvent system
Surface Welling GW, et al. Isolation of Integral membrane protein of Sen- Mono Q HR 5/5 24-min gradient from 0.15 M to
membrane Sendai virus F protein by dai virus (a paramyxovirus of 1.5 M sodium chloride in 0.02
anion-exchange high-perfor- mice): F (fusion protein. M, = M sodium phosphate (pH 7.2)
mance liquid chromatography 6500) containing 0.1% Triton X- 100.
in the presence of Triton X
100. J Chromatogr 1983:266:
629-632.
Welling GW, et al. Isolation of Integral membrane proteins of TSK 4000SW (gel filtra- TSK 4000 SW-isocratic elution
detergent-extracted Sendai vi- Sendai virus (a paramyxovirus tion); Mono Q HR 5/ with 50 mM sodium phos-
rus proteins by gel-filtration, of mice): HN (hemagglutinin- 5 (ion exchange); C1 phate, pH 6.5, containing 0.1%
ion-exchange and reversed- neuraminidase. M, = 68,000) (reversed-phase) SDS.
phase high-performance liquid and F (fusion protein. M, = Mono Q HR 5/5-24-min gradi-
chromatography and the effect 6500) ent from 20 mM Tris-HC I , pH
on immunological activity. J 7.8, containing 0. 1% Triton X-
Chromatogr 1984:297:101- 100 to 0.5 M sodium chloride
109. in the same buffer.
CI-25-min gradient of 25% ace-
tonitrile in water with 0.05%
TFA to 75% acetonitrile in wa-
ter with 0.05% TFA.
Welling GW. Purification strate- Integral membrane proteins of TSK 4OOOSW (size ex- TSK 4000SW-elution with 0.1 %
gies for Sendai virus mem- Sendai virus (a paramyxovirus clusion); Mono Q HR SDS in 50 mM sodium phos-
brane proteins. J Chromatogr of mice): HN (hemagglutinin- 5/5 (anion exchange); phate (pH 6.5).
1987:397:165-174. neuraminidase, M, = 68,000) TSK Chelate-5PW Mono Q HR 5/5-gradient from V)
and F (fusion protein, M, = (metal chelate affin- 0 to 0.5 M sodium chloride in 3
:i'
6500) ity); Phenyl 5PW-RP 20 mM Tris-HC1 (pH 7.8). con- s
(reversed-phase) taining 0.1% (w/w) decyl- ?!
PEG.
TSK Chelate-5PW--
FIRSTPROGRAM: gradient from 0
to 0.1 M glycine in 20 mM
Tris-HCI (pH 8.0), containing
0.5 M sodium chloride and
0.1 % decyl-PEG (buffer A).
[Equilibrarion-with huffer A.
Then loaded with 0.2 M zinc
chloride and equilibrated agam
with buffer A]
SECOND PROGRAM: Gradient
from buffer A to huffer B.
[Both buffers contained 0.2 M
sodium acetate (pH 7.0) and
0.1% (wlw) decyl-PEG. Also,
buffer A contained 0.5 M so-
dium chloride, and huffer B
contained 0.5 M ammonium
chloride.]
[Equilibration-with buffer A.
Then loaded with 0.2 M zinc
chloride in buffer A and 10 ml
of buffer B. Before sample ap-
plication, column reequili-
brated with buffer A].
Phenyl 5PW-RP-24-min gradi-
ent from 15% to 75% acetoni-
rile in water containing 0.05%
TFA.
Table 1 Continued
2
Protein 0
classification Ref. Substance purified Column type Solvent system
Welling-Wester S, et al. Effect of Integral membrane proteins of Two tandem-linked Su- Following eluents were used: 6
detergents on the structure of Sendai virus (a paramyxovirus perose 6HR 10130 M guanidine hydrochloride in
integral membrane proteins of of mice): HN (hemagglutinin- (300 X 10 mm id.) 50 mM sodium phosphate (pH
Sendai virus studied with size- neuraminidase, M, = 68,000) size exclusion col- 6.5); 0.25% deoxycholate in
exclusion high-performance liq- and F (fusion protein, M, = umns 10 mM sodium phosphate (pH
uid chromatography and mono- 6500) 8.1); 0.1% Brij 35 in 50 mM
clonal antibodies. J Chro- sodium phosphate (pH 6.5);
matogr 1988;443:255-266. 0.1% triethylamine (pH 3.0)
with 0.1% decyl polyethylene
glycol-300; 20% acetonitrile
in 50 mM sodium phosphate
(pH 6.5); 0.1% lauryldimethy-
lamine oxide in 50 mM so-
dium phosphate (pH 6.5);
0.25% taurocholate in 10 mM
sodium phosphate (pH 7.4);
0.03% Tween 20 in 50 mM so-
dium phosphate (pH 6.5).
0.1% decyl polyethylene gly-
col-300 in 50 mM sodium
phosphate (pH 6.5); 0.1% SDS
in 50 mM sodium phosphate
(pH 6.5); 0.25% CHAPS in
100 mM sodium phosphate
V,
(pH 6.5); 0.1 % octylglucoside
3
in 50 mM sodium phosphate =
(pH 6.5); and 0.05% sarkosyl
in 10 m~ T ~ ~ S - H( Cp~7.5)
~ %
supplemented with 0.6 M so-
diurn chloride.
Van Ede J, et al. Comparison of Integral membrane proteins of Mono Q HR 515 (anion Anion exchange-gradient from V
non-ionic detergents for extrac-
tion and ion-exchange high-per-
Sendai virus (a paramyxovirus
of mice): HN (hemagglutinin-
exchange); Zorbax Bi-
oseries GF 450 or
0 to 0.5 M sodium chloride in 5
20 mM Tris-HCI (pH 7.8) con-
formance liquid chromatogra- neuraminidase, M, = 68,000)
and F (fusion protein, M, =
TSK G4000SW (size taining 0.1% (wlw) of deter- 5.
6
phy of Sendai virus integral exclusion) gent. 5'
membrane proteins. J Chro- 6500) Size exclusion-mobile phase %
matogr l989;476:3 19-327. was 50 mM sodium phosphate
a
(pH 6.5) containing 0.1% SDS.
Welling-Wester S, et al. Compari- Integral membrane proteins of Ion erchange: Ion exchange-Linear 12-min 2.
son of ion-exchange high-per- Sendai virus (a paramyxovirus Mono Q HR 515 (80- gradient from 20 mM Tris-HC1
0
formance liquid chromatogra- of mice): HN (hernagglutinin- nm pores). TSK (pH 7.8), containing 0.1% de- 0
phy columns for purification neuraminidase, M, = 68,000) DEAE-NPR(nonpo- tergent, to 0.5 M sodium chlo- 3
of Sendai virus integral mem- and F (fusion protein, M, = rous); and Zorbaw ride in the same buffer. [Pre- 5
brane proteins. J Chromatogr
1989;1988;476:477-485.
6500) BioSeries SAX
(30-nm pores)
ceded by isocratic elution for 5
min]
5
*
V
Size exclusion: Size exclusion-elution with 50 7
virus integral membrane proteins [64]. Extracted proteins were subjected to ion
exchange HPLC using four different columns (MA7Q, Zorbax BioSeries SAX,
Mono Q, and PL-SAX). The amount of protein in the extracts and fractions after
anion exchange HPLC was determined by size exclusion HPLC on a Polyol Si-
500 column, and the relative recoveries of protein were similar for all four anion
exchange columns. A final study on the purification of Sendai virus proteins by
this group examined the effect of different amounts of the nonionic detergents
ClOE5 and C12E5 in the eluents from Mono Q HR 515 ("classical system")
and Mono Q PC 1.615 (Smart system) ion exchange columns [71]. They found
that a detergent concentration of less than 0.026-0.05% allows the integral mem-
brane proteins of Sendai virus to remain on the column. The authors used size
exclusion HPLC to determine the yield from both classical and Smart anion ex-
change HPLC systems and found that the Smart system had the advantage that
only one-tenth of the sample amount needed for classical HPLC is required to
obtain comparable results. A disadvantage of the Smart system was the relatively
long time required for equilibrium of the column with different detergents.
A tandem column procedure using an I.D. Brownlee C4 300-A 7-pm pre-
column coupled to a Vydac C4 300-A 5-pm column isolated a recombinant ma-
laria circumsporozoite fusion protein [13]. Specifically, the isolated recombinant
protein R32Leu-Arg was a 32-tetrapeptide sequence from the immunodominant
repeat region of the malaria circumsporozoite protein of Plasmodium falciparum
(R32) linked to the dipeptide Leu-kg. An affinity column containing Ricinus
cummunis agglutin (RCA 120) was used to purify N-linked oligosaccharides of
the human immunodeficiency virus recombinant envelope glycoprotein gp120
produced in Chinese hamster ovary (CHO) cells [39].
Extracellular Fassina, Giorgio, et al. High human endothelin ( 1-2 1) re- ABI Aquapore 30 X 2.1 Linear acetonitrile gradient
yield expression and purifica- leased from human big endo- mm i.d. RP; ABI (0.1% TFA) from 3% to 60%
tion of human endothelin-l . thelin (1-37) Aquapore RP-8 HPLC
Protein Expression Purif 1994;
5(6):559-568.
Hiraoka. Osamu, et al. Forma- mCRH-G-CSF complex [a cyto- TSK gel G3000SW (gel Equilibrated with 20 rnM so-
tion of l : l complex of the cy- kine receptor homologous filtration) dium phosphate buffer, pH
tokine receptor homologous re- (CRH) region of the murine 7.0, containing 0.2 M NaC1.
gion of granulocyte colony- granulocyte colony-stimulating Eluted with the same solution.
stimulating factor receptor factor (G-CSF) receptor ex-
with ligand. Biosci Biotechnol pressed by an E. coli maltose-
Biochem 1995;59(12):2351- binding protein (MBP) fusion
2354. system.]
Intracellulnr Hoener zu Bentrup, Kerstin, et Fragments generated by cleaving Vydac C4 Acetonitrile gradient (0-70%) in
al. Maltose transport in Aero- a periplasmic maltose-binding 0- 1% TFA
rnonas hydrophila: purifica- protein from Aeromonas h!-
tion, biochemical characteriza- drophila with CNBr
tion and partial protein
sequence analysis of a peri-
plasmic maltose-binding pro-
tein. Microbiology (Reading,
U. K.) 1994;140(4):945-951.
I
Kim. Do Hyung, et al. Expres- HIV- I protease Mono S HR/5 Equilibrated with the Mono S 'CI
sion and purification of HIV-1 equilibration buffer (50 mM
protease utilizing a maltose MES, 1 mM EDTA, 1 mM D
binding protein. Mol Cells
1994;4(1):79-84.
Dm, 10% glycerol, pH 6.2).
NaCl gradient (0-0.1 M for 5
5.
min, and 0.1 -0.4 M for 30 nl
e.
min). 0
3
Martinez, Aurora, et al. Expres- Fusion protein MBP-(FxJhPAH HiLoad Superdex 200 HR Mobile phase consisted of 20
sion of recombinant human [a recombinam human phenyl- (size exclusion) mM Na-Hepes and 0.2 M 0,
phenylalanine hydroxylase as alanine hydroxylase (hPAH) NaC1, pH 7.0 a
fusion protein in Escherichia fused through the target se-
W
coli circumvents proteolytic quences of the restriction pro-
degradation by host cell prote- tease factor Xa to the C-termi-
ases. Isolation and character- nal end of E. coli MBP]
ization of the wild-type en-
zyme. Biochem J 1995;306(2):
589-597. ;c
Surface Quadri, Luis EN, et al. Charac- Camobacteriocin B2 immunity C8-Vydac (10 X 250 Gradient from 38.5% to 45.5%
!?.
3
membrane terization of the protein confer- protein (CbiB2) mm, 10-ym particle of acetonitrile in 0.1 % TFA cn
ring immunity to the antimicro- size, 3 0 0 . ~pore size)
bial peptide carnobacteriocin
B2 and expression of carno-
bacteriocins B2 and BMI. J
Bacterial 1995;177(5):1144-
1151.
Smith et al.
Vakharia, Vikram N.. et al. Syn- Pheromone biosynthesis-activat- 4.6-mm-diam. Aquapore 4.6-mm-dian1.-gradient of ace-
thetic pheromone biosynthesis ing neuropeptide (PBAN) RP-300 column; 2.1-mm tonitrile rising from 10% at
activating neuropeptide gene gene product derived from ex- diam. Aquapore RP-300 0.67%/min, in 0.05 M so-
expressed in a haculovirus ex- tracellular fraction of 5B 1-4 columns dium phosphate, pH 6.0;
pression system. Insect Bio- (an insect cell line) infected 2.1-mm-diam.
chem Mol Biol 1995;25(5): with vINV-4 (a recombinant -gradient of isopropanol rising
583-589. baculovirus) from 10% at O.5%/min in
0.1% TFA;
g r a d i e n t of acetonitrile in
0. I % heptafluorobutyric acid;
-gradient of acetonitrile in
0.1% TFA.
lntracellular Xue, Hong, et al. Purification of Bacillus suhrilis tRNATQgene Vydac C4-derivatized silica Eluted with 60 min linear gradi-
hyperexpressed Bacillus subti- product cloned in E. coli ent from buffer A (10 mM so-
lis tRNATrp cloned in Esche- dium phosphate, pH 5.5, 1 M
richia coli. J Chromatogr, Bio- sodium formate, 8 mM
med Appl 1993;613(2):247- MgC1,) to buffer B (10 mM
255. sodium phosphate: pH 5.5,
10% methanol), followed by
isocratic elution with 100%
buffer B for 20 min.
Yamagata, S., et al. Overexpres- Saccharomyces cerevisiae DEAE-SPW (ion ex- Not given
sion of the Sac-charomyces MET 17lMET25 gene product change); G3000SW (gel
cerevisiae MET17lMET25 O-acetylserine-O-acetylhomo- filtration)
gene in Escherichia coli and serine sulfiydrylase (OAS-
comparative characterization OAH) expressed in E. coli;
of the product with O-ace- OAS-OAH from yeast
tylserine-0-acetylhomoserine
sulfhydrylase of the yeast.
Appl Microbial Biotechnol
1994;42(1):92-99.
Nuclear Lillehoj, Erik P., et al. Virion- Tax (transregulatory) protein of Linear gradient of 0-1 00% aque-
associated transregulatory pro- HTLV- 1) from HUT- 102 ous acetonitrile/O.l% TFA
tein of human T-cell leukemia cells
virus type I. AIDS Res Hum
Retroviruses 1992;8(2):237-
244.
Surface Bhown, Ajit S., et al. Purifica- Gag gene products of avian type I2'I gel pem~eationcolumns Mixture of acetic acid/propanoll
membrane tion and characterization of C retroviruses highly purified water (20: 15:
the gag gene products of 65)
avian-type C retroviruses by
high-pressure liquid chroma-
tography. Anal Biochem
1981;112(1):128-134.
Kolbe, Hanno V.J., et al. Isola- Recombinant partial gag gene Sulfoethyl aspartamide; Sulfoethyl asparramide-Equili-
tion of recombinant partial product p18 (HIV- I,,) [= Nucleosil C4; Vydac brated with a blank gradient;
gag gene product p 18 (HIV- membrane-associated struc- 218TP54 C18 nonlinear gradient from 0 to
1Bru) from Escherichia coli. tural protein of HIV- I ] 100% Cat Ex-B buffer [40
J Chromatogr 1989;476:99- mM sodium phosphate (pH
112. 7.0)-1 M sodium chloride] in
CatEx-A buffer [20 mM so-
dium phosphate (pH 7.0)-40
mM sodium chloride] fol-
lowed by return to 100% Cat
Ex-A buffer.
Nucleosil C4-Nonlinear gradi-
ent from 90% eluent R1 -A
(0.1% TFA in Milli-Q water)
to 90% eluent RP I -B [0. 1%
TFA in acetonitrile-Milli-Q
water (70: 30. vlv)] and then
back to 90% RPI-A.
Vydac 218TP54 Cl8-Nonlin-
ear gradient from 99% eluent
RPI-A to 100% eluent RPI -B
and then back to 99% RPI-A.
322 Smith et al.
B. lntracellular Proteins
Xue et al. hyperexpressed the gene product from Bacillus subtilis tRNATQcloned
in E. coli [73]. Vydac CCderivatized silica was used to isolate the gene product.
Overexpression of the Saccharomyces cerevisiae MET17lMET25 gene in,E. coli
was conducted by Yamagata et al. [74]. Following overexpression, the gene prod-
uct was characterized and compared with 0-acetylserine-0-acetylhomoserine
sulfhydrylase from yeast. Both ion exchange and gel filtration HPLC were per-
formed on the two proteins to determine behavioral differences. Results implied
that no differences in molecular size and electric charge exist. It should be noted
that in this case HPLC was used for characterization instead of purification.
D. Nuclear Proteins
HUT-102 cells were engineered to produce a virion-associated transregulatory
protein of human T-cell leukemia virus type 1 [33]. A C4 RP column and a linear
gradient of 0-100% aqueous acetonitrile with 0.1% TFA was used to isolate the
transregulatory protein.
HPLC Purification of Recombinant Proteins
V. DISCUSSION
Since the advent of recombinant DNA techniques in the 1970s [5 11, a great num-
ber of different proteins have been produced in a variety of expression systems
[63]. As biotechnology progresses, molecular biologists are producing an ever
greater variety and quantity of recombinant proteins. As the examples in Tables
1-3 illustrate, the purification of recombinant proteins is accomplished using
standard protein purification techniques that rely on the ability to separate pro-
teins on the basis of molecular size, electrical charge, hydrophobicity, or affinity
for a specific monoclonal antibody. Examination of purification strategies for
different protein classifications, i.e., extracellular, intracellular, surface mem-
brane, and nuclear, suggests that there is no overall preferred method for any
given protein type. The studies reviewed show that combinations of ion exchange,
size exclusion, and RP-HPLC techniques have been used to purify each of these
protein categories. Two studies report the use of affinity HPLC to purify surface
membrane proteins [39,68]. The affinity method may be particularly suited for
surface membrane protein purification. This technique can exploit the unique
biological specificity of the protein-ligand interactions [ I I], which frequently
occur at the cell surface in in vivo systems, e.g., the interaction between a carbo-
hydrate moiety and a lectin [39]. The majority of methods for purifying surface
membrane proteins required a detergent in the solvent system. Detergents are
amphipathic molecules whose hydrophilic head and hydrophobic tail allow them
to compete with the phospholipids in the membrane lipid bilayer [36], thereby
freeing the membrane protein of interest from the bilayer [65]. Extraction of
membrane proteins with nonionic detergents, such as Triton X- 100, decylpolye-
thylene glycol (DecylPEG), and octylglucoside, preserves the biological activity
of the protein. Ionic detergents, such as SDS and naturally occurring deoxycho-
late and taurodeoxycholate bile salts, tend to irreversibly denature proteins. How-
ever, transmembrane protein hydrophobic regions have a greater affinity for this
type of detergent [65].
In many biological applications, retention of conformational structure and
the attendant biological activity is important. For example, because the biological
activity of human growth hormone (hGH) is preserved upon production in a bac-
terial expression system, recombinant techniques can be used to provide this hor-
mone for hGH-deficient children. Previously, the purification method involved
painstaking extraction of the hormone from the pituitary of human cadavers.
Studies by Nishi et al. and Knudtzon show that the therapeutic effects of pituitary
hGH are successfully duplicated by recombinant hGH [29,41].
Retention of the structure-activity relationship in various recombinant cy-
tokines has allowed the large-scale production of these molecules for patient ther-
apy. The biological activity of recombinant interferon-y (IFNy) has been shown
to supplement the activity of endogenous IFNy following injection into patients
324 Smith et al.
with malignant tumors [75], and IFNy has been successfully administered for the
treatment of chronic myeloproliferative syndromes and the prevention of chronic
granulomatous disease [52]. Also, recombinant granulocyte colony-stimulating
factor (G-CSF) and granulocyte-macrophage CSF (GM-CSF) stimulate granulo-
poiesis after chemotherapy or bone marrow transplantation and mobilize marrow
stem cells for peripheral blood stem cell transplantation.
Numerous interleukins retain their biological activity and therapeutic po-
tential when produced as recombinant proteins. Biologically active interleukin-
12 (IL-12) has been produced in vitro and in vivo by recombinant techniques
[5] and has been shown to provide effective therapy against tumors and infections
[7]. Recombinant human (rh) IL-11 has been used clinically to preserve the integ-
rity of gastrointestinal mucosa during cancer treatment regimens [49]. The cyto-
kine synthesis-inhibiting action of IL-I0 is duplicated in rhIL-10 and is therapeu-
tic for patients with myelomonocytic leukemia. Likewise, the pleiotropic bio-
logical activities of IL-6 on B cells, T cells, and hematopoietic progenitors are
preserved in recombinant IL-6 [26] and serve to inhibit advanced renal cell can-
cer [56]. Lastly. the activity of rhIL-2 provides effective inhibition of growth of
breast cancer [34].
Purifying fusion proteins whose synthetic sequences retain their original
conformational structures and attendant biological activities is another technical
challenge that promises advances in patient therapy. For example, antibody-
cytokine fusion proteins combine the unique targeting ability of antibodies with
the multifunctional activities of cytokines. Becker et al. produced antibody-IL-
2 fusion proteins by fusing a sequence coding for human IL-2 to genes encoding
antibodies [2]. Their use of antibodies targeted the IL-2 to the tumor site effec-
tively and eradicated human hepatic and pulmonary melanoma metastases in Se-
vere Combined Immune Deficiency (SCID) mice.
Immunotoxins, fusion proteins made up of a toxin and a monoclonal anti-
body, also offer an attractive approach to cancer therapy [63]. O'Boyle et al.
purified an immunotoxin combining a type 1 ribosome inactivating protein with
a murine monoclonal antibody reactive with a polymorphic determinant of class
2 HLA-DR histocompatibility leukocyte antigen (HLA) on human lymphoma
cells [42]. Such immunotoxins are advantageous because the antibody delivers
the toxin specifically to the target cell.
The use of mammalian cells for protein production has been favored be-
cause it is often difficult to retain the desired biological activity when therapeutic
proteins are produced in bacteria and yeast. Accordingly, recent efforts have fo-
cused on the development of numerous mammalian cell lines to host proteins
produced by recombinant techniques [43].
The field of chromatography is vital to recombinant technology. In particu-
lar, HPLC has played an important role in the purification of recombinant pro-
HPLC Purification of Recombinant Proteins 325
teins. Technological advances, e.g., the development of new detergents, will con-
tinue to provide new avenues for the use of HPLC.
REFERENCES
Knudtzon J. Growth hormone therapy of short stature. Acta Paediatr Scand 1986;
75(3):353-361.
Kolbe HVJ, Jaeger F, Lepage P, Roitsch C, Lacaud G, Kieny MP, Sabatie J, Brown
SW, Lecocq JP, Girard M. Isolation of recombinant partial gag gene product p18
(HIV-1 Bru) from Escherichia coli. J Chromatogr l989;476:99- 112.
Kornberg A. Why purify enzymes? In: Deutscher MP, ed. Methods in Enzymology.
Vol 182. Guide to Protein Purification. San Diego: Academic Press, 1990.
LaVallie ER, McCoy JM. Gene fusion expression systems in Escherichia coli. Curr
Opin Biotech 1995;6:501-506.'
Lillehoj EP, Alexander SS. Virion-associated trans-regulatory protein of human T-
cell leukemia virus type I. AIDS Res Hum Retroviruses 1992;8(2):237-244.
Liu DL, Yang MQ, Eberhardt J, Persson B. Repeated immunotherapy using intratu-
moural injection with recombinant interleukin-2 and tumour-infiltrating lymphocytes
inhibits growth of breast cancer and induces apoptosis of tumour cells. Cancer Lett
1996;103(2):131-136.
Luckow VA, Summers MD. Trends in the development of baculovirus expression
vectors [review]. Bio Technology 1988;6:47-55.
Martin DW. Membranes: In: Martin DW, Mayes PA, Rodwell VW and Granner DK,
eds. Harper's Review of Biochemistry. 20th ed. Los Altos, California: Lange, 1985.
Martinez A, Knappskog PM, Olafsdottir S, Doskeland AP, Eiken HG, Svebak RM,
Bozzini M, Apold J, Flatmark T. Expression of recombinant human phenylalanine
hydroxylase as fusion protein in Escherichia coli circumvents proteolytic degrada-
tion by host cell proteases. Isolation and characterization of the wild-type enzyme.
Biochem J 1995;306(2):589-597.
Marumoto Y, Sato Y, Fujiwara H, Sakano K, Saeki Y, Agata M, Furusaw M, Maeda
S. Hyperproduction of polyhedrin-IGF I1 fusion protein in silkworm larvae infected
with recombinant Bombyx mori nuclear polyhedrosis virus. J Gen Virol 1987;68(10):
2599-2606.
Mizuochi T, Spellman MW, Larkin M, Solomon J, Basa LJ, Feizi T. Structural
characterization by chromatographic profiling of the oligosaccarides of human im-
munodeficiency virus (HIV) recombinant envelope glycoprotein gp120 produced in
Chinese hamster ovary cells. Biomed Chromatogr 1988;2(6):260-270.
Mueller S, Senn H, Gsell B, Vetter W, Baron C, Boeck A. The formation of diselen-
ide bridges in proteins by incorporation of selenocysteine residues: biosynthesis and
characterization of (Se)2-thioredoxin. Biochemistry 1994;33(11):3404-3412.
Nishi Y, Masuda H, Nishimura S, Kihara M, Suwa S, Tachibana K, Takeda M,
Okada Y, Matsuda I. Isolated human growth hormone deficiency due to the hGH-I
gene deletion with (type IA) and without (the Israeli-type) hGH antibody formation
during hGH therapy. Acta Endocrinol 1990;122(2):267-271.
O'Boyle KP, Colletti D, Mazurek C, Wang Y, Ray SK, Diamond B, Rosenblum
MG, Epstein AL, Shochat D, Dutcher JP, Wiernik PH, Klein RS. Potentiation of
antiproliferative effects of monoclonal antibody Lym-1 and immunoconjugate Lym-
1-gelonin on human Burkitt's lymphoma cells with gamma-interferon and tumor
necrosis factor. J Immunother 1995;18(4):221-230.
Ogez JR, Builder SE. Downstream processing of proteins from mammalian cells.
Bioprocess Techno1 1990; lO:393-416.
328 Smith et al.
O'Keefe DO, Lee AL, Yamazaki S. Use of monobromobimane to resolve two re-
combinant proteins by reversed-phase high-performance liquid chromatography
based on their cysteine content. Chromatography 1992;627(1-2):137-143.
Paborsky LR, Fendly BM. Fisher KL, Lawn RM, Marks BJ, McCray G, Tate KM,
Vehar GA and Gorman CM. Mammalian cell transient expression of tissue factor
for the production of antigen. Protein Eng 1990;3:547-553.
Papadoyannis IN, ed. HPLC in Clinical Chemistry. New York: Marcel Dekker, 1990,
Chap. 19.
Patrick JS, Lagu AL. Determination of recombinant human proinsulin fusion protein
produced in Escherichiu coli using oxidative sulfitolysis and two-dimensional
HPLC. Anal Chem 1992;64(5):507-5 11.
Pennica D, Holmes WE, Kohr WJ, Harkins RN, Vehar GA, Ward CA, Bennett WF,
Yelverton E, Seeburg PH, Heyneker HL, Goeddel DV, Collen D. Cloning and ex-
pression of human tissue-type plasminogen activator cDNA in E. coli. Nature 1983;
301:214-221.
Peterson RL, Bozza MM, Dorner AJ. Interleukin-l l induces intestinal epithelial cell
growth arrest through effects on retinoblastoma protein phosphorylation. Am J Pa-
tho1 1996;149(3):895-902.
Quadri LEN, Sailer M, Terebiznik MR, Roy KL, Vederas JC, Stiles ME. character-
ization of the protein conferring immunity to the antimicrobial peptide carnobacteri-
ocin B2 and expression of carnobacteriocins B2 and BM1. J Bacteriol 1995;177(5):
1144-1151.
Richmond S, publisher. Scrip Biotechnology Made Simple, 4th ed. Richmond, Sur-
rey, UK: PJB Publications, 1991.
Robak T. Cytokiny w leczeniu chorob krwi (Cytokines in the treatment of blood
diseases). Acta Haematol Pol 1995;26(2 Suppl 1):72-78.
Sadana A. Inactivation of proteins and other biological macromolecules during chro-
matographic methods of bioseparation. Bioseparation 1992;3:145-165.
Schoneich C, Hiihmer AFR, Rabel SR, Stobaugh JF, Bigelow DJ, Williams
TD. Separation and analysis of peptides and proteins. Anal Chem 1995;67(12):
155R.
Schoneich C, Kivok SK, Wilson GS. Rabel SR, Stobaugh JF, Williams TD, Vander-
velde DG. Separation and analysis of peptides and proteins. Anal Chem 1993;65(12):
67R.
Schuler M, Peschel C, Schneller F, Fichtner J, Farber L, Huber C, Aulitzky WE.
Immunomodulatory and hematopoietic effects of recombinant human interleuhn-6
in patients with advanced renal cell cancer. J Interferon Cytokine Res 1996;16(11):
903-91 0.
Sharma SK, Evans DB, Vosters AF, McQuade TJ, Tarpley WG. Metal affinity chro-
matography of recombinant HIV-I reverse transcriptase containing a human renin
cleavable metal binding domain. Biotechnol Appl Biochem 1991;14(1):69-8 1.
Swoboda I, Jilek A, Ferreira F, Engel E, Hoffmann-Sommergruber K, Scheiner 0 ,
Kraft D, Breiteneder H, Pittenauer E. Isoforms of Bet v I, the major birch pollen
allergen, analyzed by liquid chromatography, mass spectrometry, and cDNA clon-
ing. J Biol Chem l995;270(6):2607-2613.
Uhlin M, Moks T. Gene fusions for purpose of expression: an introduction. In:
HPLC Purification of Recombinant Proteins 329
Goeddel DV, ed. Methods in Enzymology. Vol 185. Gene Expression Technology.
San Diego: Academic Press, 1990.
Vakharia VN, Raina AK, Kingan TG, Kempe TG. Synthetic pheromone biosynthesis
activating neuropeptide gene expressed in a baculovirus expression system. Insect
Biochem Mol Biol 1995;25(5):583-589.
Valenzuela P, Medina A, Rutter WJ, Ammerer G. Hall BD. Synthesis and assembly
of hepatitis B virus surface antigen particles in yeast. Nature 1982;298:347-350.
Van Ede J, Nijmeijer JRJ, Welling-Wester S, Oervell C and Welling GW. Compari-
son of non-ionic detergents for extraction and ion-exchange high-performance liquid
chromatography of Sendai virus integral membrane proteins. J Chromatogr 1989;
476~319-327.
Watson JD, Gilman M, Witkowski J, Zoller M. Recombinant DNA, 2nd ed. New
York: Scientific American Books, W.H. Freeman 1992.
Welling GW, Hiemstra Y, Feijlbrief M, Oervell C, Van Ede J, Welling-Wester S.
Comparison of detergents for extraction and ion-exchange high-performance liquid
chromatography of Sendai virus membrane proteins. J Chromatogr 1992;599(1-2):
157-162.
Welling GW, Van der Zee R, Weeling-Wester S. HPLC of membrane proteins. In:
Gooding KM, Regnier FE, eds. HPLC of Biological Macromolecules. New York:
Marcel Dekker, 1990.
Welling GW, Nijmeijer JRJ, Van der Zee R, Groen G, Wilterdink JB, Welling-
Wester S. Isolation of detergent-extracted Sendai virus proteins by gel-filtration, ion-
exchange and reversed-phase high-performance liquid chromatography and the ef-
fect on immunological activity. J Chromatogr 1984;297: 10 1 - 109.
Welling GW, Groen G, Welling-Wester S. Isolation of Sendai virus F protein by
anion-exchange high-performance liquid chromatography in the presence of Triton
X 100. J Chromatogr 1983;266:629-632.
Welling GW, Slopsema K, Welling-Wester S. Purification strategies for Sendai virus
membrane proteins. J. Chromatogr 1987;397:165- 174.
Welling-Wester S, Haring RM, Laurens H, Oervell C, Welling GW. Comparison
of ion-exchange high-performance liquid chromatography columns for purification
of Sendai virus integral membrane proteins. J Chromatogr 1989;476:477-485.
Welling-Wester S, Kazemier B, Oervell C, Welling GW. Effect of detergents on the
structure of integral membrane proteins of Sendai virus studied with size-exclusion
high-performance liquid chromatography and monoclonal antibodies. J Chromatogr
1988;443:255-266.
Welling-Wester S, Feijlbrief M, Koedijk DGAM, Braaksma MA, Douma BRK,
Welling GW. Effect of different amounts of the nonionic detergents C10E5 and
C12E5 present in eluents for ion-exchange high-performance liquid chromatography
of integral membrane proteins of Sendai virus. J Chromatogr 1993;646(1):37-44.
Wisniewski R. Principles of the design and operational considerations of large scale
high performance liquid chromatography (HPLC) systems for proteins and peptides
purification. Bioseparation 1992;3:77-143.
Xue H, Shen W, Wong JTF. Purification of hyperexpressed Bacillus subtilis
tRNATrp cloned in Escherichia coli. J Chromatogr Biomed Appl 1993;613(2):247-
255.
330 Smith et al.
I. INTRODUCTION
Over the last few years, significant data have accumulated concerning compo-
nents present in the seminal plasma such as hormones, enzymes, peptides, and
cytokines [I-41. This progress has led us to the discovery of other components
in seminal plasma, not only in humans but in animals. We have achieved a greater
understanding of the functional properties of these components, especially semi-
nal plasma proteins in the male reproductive organs, its role in male fertility,
and its immunological effects in vitro. Still, the physiochemical and functional
properties of most of these proteins in seminal plasma remain unclear. Therefore,
further investigation should be established to determine the complexities of these
components present in seminal plasma and whether it has a role in the immunopa-
thogenesis of diseases in the male and female reproductive systems. The protein
makeup of seminal plasma appears to be extremely susceptible to variation on
account of large number of factors, such as species, age, frequency of semen
collection, the nature of ejaculates, the early ejaculates which show a protein
profile different from that of the later ejaculates, the hormonal status of the indi-
vidual, the condition of storage of semen, and possibly other factors such as diet,
weather, environmental and emotional factors including stress. Unlike the other
332 Haq et al.
body Ruids that are readily accessible for sampling and study, generation of se-
men, and therefore of seminal plasma, cannot be ordered about. The seminal
plasma of all species is rich in proteins but some of the proteins are not indigenous
to seminal plasma in that they may originate from serum. The proteins may also
leak into the seminal plasma from dead spermatozoa or spermatozoa damaged
during centrifugation or handling of the semen. Furthermore, some of the proteins
of seminal fluid may be bound to the spermatozoa immediately after ejaculation
and thus no longer be available in seminal plasma. The proteins in seminal plasma
are contributed from secretions of various male reproductive glands, e.g., epididy-
mis, seminal vesicles, prostate, and Cowper's and coagulating glands. The
remaining proteins found in the seminal plasma have been divided into the fol-
lowing groups: proteolytic enzymes; glycolytic enzymes; nucleolytic enzymes;
other enzymes; hormones and growth factors; antifertility factors; immunosup-
pressive factors; androgen-binding proteins; inhibin; immunoglobulins; and other
nonenzymatic, nonhormonal proteins.
II. PROTEINS
Proteins are fundamental components of all living cells and include many sub-
stances, such as enzymes, hormones, and antibodies, that are necessary for the
proper functioning of an organism. Proteins serve as the structural pattern of the
protoplasms from enzymes, hormones, chromosomes, and cell components. They
constitute about 15% of the protoplasms, are colloidal in nature, and are formed
of large molecules of great complexity and variety. These proteins are built up
by peptide linkages of amino acids with the peptide bonds between the amino
acid group (NH,) of one amino acid and the acid group (COOH) of the adjacent
one. They are made up of 20 amino acids as tripeptide and of several amino
acids as polypeptide. The human body produces thousands of different proteins,
including those that act as catalysts, those that are localized where they regulate
the flow of material in and out of the cell, those that are soluble or membrane-
bound, those that can interconvert different types of energy, those that recognize
foreign materials and microbes, those that are implicated in the repair of injury,
those that respond to biological stress, and those with purely structural roles.
Proteins can be structural (intracellular and extracellular proteins); catalytic (en-
zymes); vectoreal (transport proteins); regulatory (determine the rate at which
other proteins are made).
Any deficiencies and excess of these proteins can contribute to human dis-
eases. Significant studies have been done to determine chemical properties that
can be derived from the composition and distribution of amino acids. But still,
the relationship between the chemical compositiion, structure, and the function
of seminal plasma proteins is obscure and intriguing.
Isolation of Human Seminal Plasma Proteins 333
during ejaculation and expel their contents into the ejaculatory ducts, thus wash-
ing the spermatozoa from the urethra.
nates and raises intracellular CAMP in leukocytes, which may contribute to the
immunosuppressive effects of seminal plasma. Immunosuppression may attribute
to the presence of a protein similar to pregnancy-associated Protein A [27]. The
presence of polyamines like spermin and spermidine has been reported and that
could also contribute to immunosuppression of seminal plasma [28]. Immunosup-
pression might also be due to the seminal nucleases and proteases in seminal
plasma [5]. Following is a brief list of important proteins of human seminal
plasma that have been studied extensively:
P-Microseminoprotein [29]
Sperm-binding proteins [30,3 11
Protein kinase inhibitor [32]
P-Endorphin [33]
Calcium-binding protein (calmodulin) [34]
Zinc-binding proteins [35]
Ion-binding proteins [36]
Placental proteins [37]
Since proteins are really large molecules, in order to investigate their structure
and function, isolation and fractionation processes are required. There is no single
or simple way to purify all proteins. Procedures useful in the purification of one
protein may result in the denaturation of another (Fig. 1).
We were able to fractionate human seminal plasma using DEAE Sephadex
A-50 ion exchange columns [44]. Semen was obtained from healthy donors and
seminal plasma was recovered by ultracentrifugation. Seminal plasma was dia-
lyzed against PBS at 4°C overnight and then applied onto a DEAE-Sephadex
A-50 ion exchange column using different salt concentrations in phosphate buffer
pH 6.0 (Fig. 2). Forty men either fertile or under investigation for infertility
donated semen, which was collected by masturbation into sterile plastic contain-
ers after 2-3 days of abstinence from ejaculation. The mean age of the test sub-
jects was 34.2 -C 8 (20-45) years. Semen analysis was performed by using Cell-
soft Automated Semen Analyzer (Cryo Resources Ltd., New York). All patients
(normospermic 20-100 million sperms/ml) included in this study attended the
IVF and Infertility Clinics of King Faisal Specialist Hospital and Research Cen-
tre, Riyadh (Saudi Arabia). All of the semen samples were centrifuged at 10,000g
at 4°C for 15 min to remove sperm and the clear supernatant was stored at -70°C
until used.
The exchanger chosen was DEAE-Sephadex A-50 because it was reported
to provide better results than DEAE-cellulose in the purification of proteins [45].
Two hundred fifty milligrams (25 ml) protein from normospermic human seminal
plasma was applied to a XK 50130 (Pharmacia, Piscataway, NJ, USA) column
Haq et al.
I Ultracentrifugatian I
I Protein Concentration I
( Chromatography I I 6
. ~~
1 Centrifueation of Fractions II
L
6
Dialysis of Fraclions
C
store at -70°C
I I I I
- 75
TUBE NUMBER
100
Figure 3 SDS-PAGE of marker proteins and human seminal plasma and its fractions
in 12.5% gel. Lane I , the marker proteins (from top): phosphorylase b (97 kDa), bovine
serum albumin (66 kDa), ovalburnin (43 kDa), carbonic anhydrase (3 1 kDa), trypsin inhib-
itor (20 kDa), and lysozyme ( I 4 kDa). Lanes PI -P7 represent seminal plasma obtained
from DEAE-Sephadex A-50 ion exchange chromatography. Lane 2, the marker proteins
(hom top): myosin (200 kDa), P-galactosidase ( I 16 kDa), phosphorylase b (97 kDa), bo-
vine serum albumin (66 kDa), and ovalbumin (43 kDa) [44].
Characterization of seminal plasma protein was carried out by using a fast protein
liquid chromatography (FPLC) system. Superose HR I0130 column (Pharmacia)
was used for the elution of the protein. A total of 100 ~1 (0.2 pglml) of protein
from seminal plasma was applied to the column [0.5 M phosphate buffer (KHPO,
+ Na2HP04)with 0.14 M NaCI]. The pH was adjusted to 7.3 by use of orthophos-
phoric acid. The buffer was degassed, filtered through 0.2-pm disposable filters,
and used as mobile phase. The flow rate was 0.2 mllmin and the fraction size
fixed at 0.5 ml. Ovalbumin (43 kDa) was used as a reference protein to confirm
the performance of the Superose column. These experiments of characterization
by FPLC and SDS-PAGE were carried out in the laboratory of Professor J. P.
Bouvet in the Institute Pasteur in Paris. The elution profile of a 20-kDa protein
as detected by FPLC is shown in Fig. 4. The results of SDS-PAGE are shown
in Fig. 5.
START
Figure 4 Elution profile of a 20-kDa protein of human seminal plasma by using FPLC
(Superose column HR 10130); 0.5 M phosphate buffer (pH 7.3) with 0.14 M NaCl was
used.
Figure 5 SDS-PAGE (12.5% gel) of marker proteins, whole human seminal plasma,
and a 20-kDa protein. Lane M represents protein markers (from top); phosphorylase b
(97 m a ) , bovine albumin (66 m a ) , ovalbumin (43 kDa), carbonic anhydrase (31 ma),
trypsin inhibitor (20 kDa), and lysozyme (14 kDa). Lane WS represents the whole human
seminal plasma and lane P represents the 20-kDa protein of human seminal plasma.
SP Fractions
WA + I O O U Q I ~ I SP WA + 40ug1m1 SP PMA + I O U Q I ~ I SP
Figure 6 Effect of Seminal plasma (SP) and its fractions (P1 to P7) on phagocytic
response to phorbol myristate acetate (PMA). The x axis represents the whole SP and
its different fractions (Pl-P7), whereas the y axis represents the relative phagocytic
index [44].
Table 1 Effect of Human Seminal Plasma Protein and Its Peaks on Phagocytic
Activity of Monocytes and Granulocytes by Flow Cytometry.
Monocytes Granulocytes
Mean fluorescent Mean fluorescent
channel number channel number
% Total Phagocytic % Total Phagocytic
Sample Phagocytosis population cells Phagocytosis population cells
Control 69 38 1 548 81 686 836
Whole 69 409 586 69 510 736
Peak 3 59 333 560 53 314 584
Peak 4 60 349 582 60 397 656
Peak 7 67 408 609 56 455 800
whole seminal plasma were stimulatory (RPI > loo%), whereas peaks 2, 3, 4,
and 7 were suppressive (RPI < 100%). These results are shown in Fig. 6. The
percent phagocytosis effect of peaks 3, 4, and 7 on monocytes and granulocytes
was also studied by using flow cytometry (Table 1).
Figure 7 Effect of human seminal plasma (SP) and its fractions on mixed lymphocyte
reaction (MLR). SP represents the whole seminal plasma while P1-P7 represent peaks
obtained from DEAE-Sephadex A-50 ion exchange chromatography. The x axis represents
protein concentration (pg) and the y axis the index [44].
studies done by Naz and Kaplan [60] on the detection of tumor necrosis factor-
a (TNFa), interleukin- 1P (IL- LP), interferon-y (IFNy), and interleukin-6 (IL-6)
in human seminal plasma. IL-6 was significantly higher in infertile men compared
to those of fertile. TNFa and IL-1 P were not detected, whereas IFNy was detected
but the difference between the levels of fertile and infertile were not significant.
IL-6 could be associated with infertility. Production of IFNy was determined and
showed to be present in higher amounts in the seminal plasma of infertile donors
[6 11. Recently, the concentrations of various cytokines (IL-1P, IL-2, IL-6, NIL-2,
srIL-6) in seminal plasma have been studied [62]. According to these researchers,
several lines of evidence indicate that cytokines are involved in male infertility.
These cytokines are secreted by different parts of the male genital tract and may
exert effects on steroidogenesis, spermatogenesis, and sperm functions.
IX. CONCLUSION
REFERENCES
Stites DP, Erickson RP. Suppressive effect of seminal plasma on lymphocytes acti-
vation. Nature 1975;253:727-729.
Caldini AL, Orlando C, Barni T, Messeri G, Pazzegli M, Baldi E. Seris M. Measure-
ment of transfemn in human seminal plasma by chemiluminescent method. Clin
Chem l986;32: 153- 156.
Bryan MKO, Baker HWG, Saunders JR, Kirszbaum L, Walker ID. Hudson P, Liu
DY, Clew MD, Apice AFD, Murphy M. Human seminal clusterin (SP-40,40). Isola-
tion and characterization. J Clin Invest 1993;85:1477- 1486.
Choi NH, Tobe T, Hara K, Yoshida H, Tomita M. Sandwich ELISA assay for quanti-
tative measurement of SP-40.40 in seminal plasma and serum. J Immunol Meth
1990;131:159-163.
Shivaji S, Scheit KH, Bhargava PM. Proteins of Seminal Plasma. New York: John
Wiley and Sons, 1990.
Kitamura M, Namiki M, Matsumiya K, Tanaka K, Matsumoto M, Hara T, Kiyohara
H, Okabe M, Okuyama A, Seya T. Membrane cofactor protein (CD46) in seminal
plasma is a prostasome-bound with complement regulatory activity and measles vi-
rus neutralizing activity. Immunology 1995;84:626-632.
Skibinski G, RElly RW, Harrison CM, McMillan LA, James K. Relative immuno-
suppressive activity of human seminal prostaglandins. J Reprod Immunol 1992;22:
189-195.
Iborra A, Morte C, Fuentes P, Garcia-Framis V, Andolz P, Martinez P. Human sperm
coating antigen from seminal plasma origin. Am J Reprod lmmunol 1996;36: 1 18-
125.
Dimitrova D, Kehayov I, Kyukechiev S. Purification and characterization of sperm-
coating antigen identified by monoclonal antibody. Andrologia 1993;25:271-277.
Nocera M, Chu TM. Transforming growth factor beta as an immunosuppressive
protein in human seminal plasma. Am J Reprod Immunol 1993;30:1-8.
Yie SM, Lobb DK, Clark DA, Younglai EV. Identification of transforming growth
factor-alpha like molecule in human seminal plasma. Fertil Steril l994;6: 129- 135.
Robert M, Gagnon C. Sperm motility inhibitor from human seminal plasma: associa-
tion with semen coagulum. Human Reprod 1995; 10:2192-2 197.
Ohkubo I, Tada T, Ochiai Y, Ueyama H, Eirnoto T, Sasaki M. Human seminal
plasma beta-microseminoprotein: its purification, characterization, and immunohis-
tochemical localization. Int J Biochem Cell Biol 1995;27(6):603-611.
Paradisi R, Pession A, Bellavia E, Focacci M, Flamigni C. Characterization of hu-
man sperm antigens reacting with antisperm antibodies from autologous sera and
seminal plasma in a fertile population. J Reprod Immunol 1995;28:61-73.
Deperthes D, Chapdelaine P, Tremblay RR, Brunet C, Berton J, Hebert J, Lazure
C, Dube JY. Isolation of prostatic kallikrein hK2, also known as hGK- I, in human
seminal plasma. Biochim Biophys Acta 1995; 1245:311-31 6.
Kekow J, Wachsman W, McCutchan JA, Gross WL, Zacchariah M, Carson DA,
Lozt M. Transforming growth factor-p and suppression of humoral immune re-
sponses in HIV infection. J Clin Invest l991;87: 1010- 1016.
Zheng X, Geiger M, Ecke S, Bielek E, Donner P, Eberspacher U, Scheuning WD,
Binder BR. Inhibition of acrosin by protein C inhibitor and localization of protein
C inhibitor to spermatozoa. Am J Physiol 1994;267:C466-472.
350 Haq et al.
Sedor J, Callahan HJ, Perussia B, Laftime EC, Hirsch IH. Soluble Fc gamma RIII
(CD 16) and immunoglobulin infertility. J Androl 1993;14:187- 193.
Chvapil M, Stankova L, Bernhard DS, Zukoski CF, Drach GW. Effect of prostatic
fluid and its fraction on some function of peritoneal macrophages. Invest Urol 1977;
15:173-179.
Mukherjee DC, Agrawal AK, Manjunathn R, Mukherjee AB. Suppression of epidid-
ymal sperm antigenicity in the rabbit by uteroglobulin and transglutaminase in vitro.
Science l983;219:989-991.
Witkin SS, Richards JM, Bangiovanni AM, Zelikovsky G. An IgG-Fc binding pro-
tein in seminal fluid. Am J Reprod Immunol 1983;3:23-27.
Kelly RW. Immunosuppressive mechanisms in semen implications for contracep-
tion. Human Reprod 1995;10: 1686-1693.
Bischof P, Marin-Du-Pan R, Lauber K, Girard JP, Herrmann WL, Sizonenko PC.
Human seminal plasma contains a protein that shares physiochemical, immunochem-
ical, immunosuppressive properties with pregnancy-associated plasma protein-A. J
Clin Endocrinal Metab 1993;56:359-362.
Byrd WJ, Jacobs DM, Amoss MS. Synthetic polyamines added to cultures con-
taining bovine sera reversibly inhibit in vitro parameters of immunity. Nature 1977;
267:621-623.
Akiyama K, Yoshioka Y, Schimid K, Offner GD, Troxler RF, Tsuda R, Hara M.
The amino acid sequence of human beta microseminoprotein. Biochim Biophys Acta
1985;829:288-294.
Abrescia P, Lombardi G, De Rosa M, Quagliozzi L, Guardiola J, Metafora S. Identi-
fication and preliminary characterization of a sperm-binding protein in normal hu-
man semen. J Reprod Fertil 1985;73:71-77.
Metafora S, Lombardi G, De Rosa M, Quagliozzi L, Ravagnan G, Peluso G, Abres-
cia P. A protein family immunorelated to a sperm-binding protein and its regulation
in human semen. Gamete Res 1987;16:229-241.
Freedman MF, Kopf GS. Characterization of a seminal plasma associated inhibitor
of human seminal plasma protein. Biol Reprod 1985;32:322-332.
Chan SY, Tang LC. Effect of reserpine on fertilizing capacity of human spermato-
zoa. Contraception 1984;30:363-369.
Marcum J, Dedman MJR, Brinkley BR, Means AR. Control of microtubule assem-
bly-disassembly by calcium-dependent regulatory protein. Proc Natl Acad Sci USA
1978;75:3771-3775.
Kavanagh J. Zinc binding properties of human prostatic tissue, prostatic secretion
and seminal fluid. J Reprod Fertil 1983;68:359-363.
Robert TK, Boettcher B. Identification of human sperm coating antigen. J Reprod
Fertil 1969;18:347-350.
Sinosich M, Seppala JM, Saunders DM, Grudzinskas G. Pregnancy-associated
plasma protein-A and placental protein in human seminal plasma. Ann NY Acad
Sci 1985;442:287-292.
Leblond E, Desnoyers L, Manjunath P. Phosphorylcholine-binding proteins from
the seminal fluids of different specie share antigenic determinants with the major
proteins of bovine seminal plasma. Mol Reprod Dev 1993;34:443-449.
Calvete JJ, Mann K, Sanz L, Raida M, Topfer-Petersen E. The primary structure of
Isolation of Human Seminal Plasma Proteins 351
56. Templeton AA, Cooper I, Kelly RW. Prostaglandin concentrations in the semen of
fertile men. J Reprod Fertil l978;52: 147-150.
57. Witkins SS. Failure of sperm-induced immunosuppression: association with anti-
sperm antibodies in women. Am J Obstet Gynecol 1989;160:1166-1168.
58. Baker HWG, Clarke GN. Hudson B. Treatment of sperm autoimmunity in men. Clin
Reprod F e d l983;2:55-71.
59. Evans CH, Lee TS, Flugelman AA. Spermine-directed immunosuppression of cervi-
cal carcinoma cell sensitivity to a majority of lymphokine-activated killer lympho-
cyte cytotoxicity. Nat Immun 1995;14: 157-163.
60. Naz RK, Kaplan P. Increased levels of IL-6 in seminal plasma of infertile men. J
Androl 1994;15:220-227.
61. Paradisi R, Capelli M, Mandini M, Bellavia E, Flamigni C. Increased levels of inter-
feron-gamma in seminal plasma of infertile men. Andrologia 1996;28:157-161.
62. Dousset B, Hussenet F, Daudin M, Bujan L, Foliguet B, Nobet P. Seminal cytokine
concentrations (IL-lbeta, IL-2, IL-6, sR IL-6), semen parameters and blood hor-
monal status in male infertility. Hum Reprod 1997;12: 1476-1479.
Isolation, Purification, and
Structural Study of Allergenic
Proteins
Jean-Pierre Dandeu
lnstitut Pasteur, Paris, France
I. INTRODUCTION
Allergy, from the Greek "allos" (other) and "ergeon" (action), can be defined as a
separate behavior in comparison with the classical immune response. It i s a modifica-
tion of the human or animal organism reactivity to aparticular immunogen, the ''aller-
gen.'' The molecule responsible for the sensitization was clearly defined at the begin-
ning of the twentieth century by Clemens von Pirquet and then by Paul Portier and
Charles Richet in their works on anaphylaxis. Either for sensitization or desensitiza-
tion an allergen can act at a very low concentration. To become sensitized to an
allergen one must be genetically programmed and able to synthesize immunoglobu-
lins of the particular IgE class in response to an antigen. However, an antigen can
only be considered as an allergen when it is able not only to induce an IgE response
but also to subsequently provoke an anaphylactic orland inflammatory reaction. De-
sensitization is a peculiar immunotherapy that leads the patient to synthesize IgG
instead of IgE in response to repeated injections of very low doses of allergen that
arepractically unable to induce any anaphylactic reaction. In this case, the best mole-
cule to be used should be one whose structure would have been modified to be nonre-
active with the specific IgE present on the mast cells and thus to prevent the cascade
of events leading to the inflammation process by inducing an IgG response.
Allergens are biological molecules such as proteins, glycoproteins, or poly-
saccharides originating from insects, plants, or animals. A biological extract often
contains several allergens. The relative molecular mass of these molecules is gener-
354 Dandeu
ally low and ranges between 10,000 and 50,000. Allergens are frequently qualified by
the terms major or minor allergens. These are referred to their particular frequencies of
IgE reactivity in a group of patients clinically allergic to these allergens but are never
referred to their concentrationsin a biological extract.
Nowadays many allergens have been isolated and purified. The genes of great-
est importance have been cloned and the corresponding recombinant proteins synthe-
sized. Both natural allergens and recombinant allergens are rather useful to improve
our knowledge on IgE synthesis and regulation but also to advance the field of allergy
diagnosis and therapy. We have personally isolated two allergens from cat, one from
horse, and one from a house dust mite. In this chapter we would like to point out the
usefulness of chromatographic methods in allergen purification. One of the most
important techniques generally used to prepare allergenic extracts was first described
by Guibert et al. [I], which essentially consists in an aqueous extraction. From this
extract the allergenic material was first concentrated by acetone precipitations at 25%
and 75%, then by an additional precipitation at 75% of saturation in ammonium
sulfate.
We do not pretend to examine all of the work done on so large a topic and we
have voluntarily limited this review to a comprehensive summary of our personal
results. Mention should be made before any description of chromatographic processes
is given that a combination of crossed line immunoelectrophoresis and rocket line
immunoelectrophoresis, two techniques previously described by Axelsen et al. [2],
are particularly helpful not only for following the degree of purity of a given antigen1
allergen but also for demonstrating whether the different antigens are bearing com-
mon or different epitopes.In rabbit, the majority of natural proteins are good immuno-
gens and give rise to high IgG antibody titers. Thus, it is easy to identify and follow
the purification of a particular antigen using a rabbit antiserum against the crude
material from which the allergen of interest will be purified. Each step of the purifica-
tion process can be followed up by immunoelectrophoresis. The allergenicity of the
protein can be determined by testing the ability to bind specific human IgE antibodies
present in the sera of sensitized patients. To this end, either crossedradioimmunoelec-
trophoresis, western blot, or enzyme-linked immunosorbent assay (ELISA) can be
performed. If sodium dodecyl sulfate-polyacrylamide gel electrophoreses (SDS-
PAGE) remains the most classical tool to define homogeneity of a proteinic fraction
or to determine its relative molecular mass, it is sometimes necessary to perform
chromatofocusing or/and capillary electrophoresis for a better characterization of
the molecule of interest.
Several authors have demonstrated the presence of allergens in horse hair and
dandruff and have isolated several proteins of different molecular weights [4-
Isolation of Allergenic Proteins 355
71. The raw material we used to isolate and purify horse allergens was principally
obtained during grooming of healthy animals. After an aqueous extraction active
material was concentrated by a classical salting-out process using ammonium
sulfate followed by an exhaustive dialysis against water not only to eliminate
ammonium sulfate but to select molecules of a relative molecular mass above
10,000. For efficient concentration and storage the resulting solution was lyophi-
lized. The most classical way to separate proteins or glycoproteins undoubtedly
remains size exclusion chromatography (SEC), which can sometimes lead to al-
most pure active fractions. This is particularly true when the relative molecular
mass of the protein of interest, i.e., Equ c l , is suspected to be around 25,000, as
found in the pic SII (Fig. 1). Since ion exchange chromatography, previously
used by other authors, was not totally efficient in spite of our several attempts
to improve the technique and no more success was obtained with immobilized
by Porath et al. [12] and Hjerten et al. 1131. If HIC has been shown useful in
purifying enzymes 114,151, hormones [ 161, and fragments from IgM 1171, until
now it has never been used to isolate and purify allergenic molecules. Using
a column packed with phenyl-Superose (Pharmacia Sweden) an almost 100%
purification of the horse major allergen Equ c l was achieved. Elution of the
adsorbed molecules according to the order of increasing hydrophobicity was per-
formed by decreasing the antichaotropic salt concentration. i.e., ammonium sul-
fate [18]. For analytical purpose this was done in a linear gradient while for
preparative ones a stepwise elution was carried out (Fig. 2). Physicochemical,
biochemical, and immunochemical analyses stated that Equ c l prepared in that
manner is a pure protein consisting of a single peptide chain with a relative molec-
ular mass of 20,000 and a pI = 3.9. Its partial microsequencing suggeqted that
it could belong to the lipocalin family. This was further confirmed by cDNA
cloning and sequencing of the Equ c l gene [19].
Several proteins derived from the domestic cat Felix doiwsricus causes allergic
disorders. The most important one, previously designated Cat I, was first de-
scribed by Ohman [21]. Isolation and purification of this tnajor feline allergen,
recently named Fel d l , was achieved by affinity chromatography using polyclonal
and monoclonal antibody 121,221. If this method is somewhat efficient it unfortu-
nately gives a poor yield. Fel d l , which is an acidic protein with a native molecu-
lar mass of 35,000, is composed of two chains that are not covalently bound and
can be easily dissociated. Each monomer, M, 17,000, has an equivalent antigenic
and allergenic potency. Fel d l is particular1y present in house dust [21,23] from
which it can be extracted by water. Since Fel d l is an acidic protein with pI 3.8,
an anion exchanger was used to fractionate the ammonium sulfate-precipitated
fraction previously prepared according to the method described above. An analyt-
ical chromatography started with an isocratic elution followed by a linear gradient
of molarity showed that three fractions can be obtained, each at a very specific
concentration of salt. After that result a stepwise elution was defined and an
enriched fraction of Fel d I, Q2, was eluted at 1 M NaCl (Fig. 3). This partially
purified allergen was then loaded onto a copper chelate column. The chromato-
gram (Fig. 4) shows that five fractions were obtained following a stepwise elution
at different salt concentrations. All fractions were itnmunologically tested, and
the one eluted during the isocratic step appeared as a pure Fel d l preparation.
A second fraction, Q2Cu2, which showed a higher affinity for the copper ions,
also appeared to contain Fel d 1 but rather contaminated. Thus, we concluded that
there were two species of this allergenic protein, which were the monomer and
Dandeu
NaCl
Molarity
the dimer, respectively (Fig. 5). These results lead us to assume that the number
of exposed metal binding sites on the protein able to bind to the copper ions was
greater on the dimer than on the monomer.
This type of experiment underlines the usefulness of affinity chroma-
tography not only to purify allergenic proteins but also to study their struc-
tures.
Previous studies showed that several allergens can be extracted from cat pelts
[25-271. The major one, Fel d l [21], was isolated and purified by us [20]. Some
Dandeu
l lmn
The major allergen from Dermatophagoides pteronyssinus mite was first isolated
and purified about 10 years ago [33-351. It was shown to be a single peptide
with some traces of carbohydrates. Its attachment to the cysteine proteases family
of enzymes appeared from its mRNA [36,37] and cDNA [38] sequences. A fusion
protein produced by a cDNA clone was effectively shown to react with specific
rabbit IgG raised against natural Der p l . As the yield of pure protein from bacte-
rial lysate was reported to be very low, we defined in our laboratory a convenient
process to prepare Der p l from a mite culture using a simple chromatographic
method. Ion exchange chromatography, already effective with a lot of proteins,
was shown to be really efficient to purify Der p l on a large scale. This process
essentially consists of applying an ammonium sulfate fraction obtained from a
partially purified mite culture extract to a Mono Q column (Pharmacia, Uppsala,
Sweden). The protein of interest was eluted during the first isocratic part of the
run, pic Q1 (Fig. 9). All of the controls applied to the so obtained fraction stated
that it was a highly purified Der p l preparation with a real homogeneity in SDS-
NaCl
Molarity
!
f1
1 2 3 4 5 6 7 8 9 1 0
Migration distance(crn)
-
Figure 10 Electropheretogram obtained by capillary electrophoresis. (From Ref. 32.)
PAGE and an M, of 25,500-a result very close to that reported by other workers
[33,37]. Our Der p l preparation also appeared to be highly homogeneous in capil-
lary electrophoresis (Fig. 10) and in chromatofocusing (Fig. 1l), the most critical
methods of analysis available at that time. At the level of the 1-25 N-terminal
peptide sequence no differences were shown when compared with the same pep-
tide of the cDNA-coded Der p l .
ACKNOWLEDGMENTS
All figures were reprinted with permission from Elsevier Science NL, Sara Burg-
erhartstraat 25, 1055 KV Amsterdam, The Netherlands.
REFERENCES
Cloning and sequencing reveal the major horse allergen Equ cl to be a glycoprotein
member of the lipocalin superfamily. J Biol Chem 1996;271:32951-32959.
Dandeu J-P, Rabillon J, Beltrand M-J, Lux M, Duval R, David B. Immobilized metal
ion affinity chromatography for the purification of Fel d I, a cat major allergen, from
a house-dust extract. J Chromatogr 1990;512: 177-88.
Leitermann K, Ohman JL. Cat allergen I: Biochemical, antigenic and allergenic
properties. J Allergy Clin Immunol 1984;74:147-1 53.
Chapman M, Aalberse RC, Brown MJ, Platts-Mills TAE. Monoclonal antibodies to
the major feline allergen Fel d l . 11. Single step affinity purification of Fel d l , N-
terminal sequence analysis, and development of a sensitive two-site immunoassay
to assess Fel d 1 exposure. J Immunol 1988;140312-8 18.
Ohman JL, Lorusso JR, Lewis S, Cat allergen content of commercial house dust
extract: comparison with dust extracts from cat-containing environment. J Allergy
Clin Immunol 1987;79:955-959.
Dandeu J-P, Rabillon J, Guillaume J-L, Camoin L, Lux M, David B. Isolation and
purification of cat albumin from cat serum by copper ion affinity chromatography:
further analysis of its primary structure. J Chromatogr 1991;539:475-484.
Ohman JL Jr, Lowell FC, Bloch KJ. Allergens of mammalian origin: characteri-
zation of allergen extracted from cat pelts. J Allergy Clin Immunol 1973;*:231-
241.
Ohman JL Jr, Lowell FC, Bloch KJ. Allergens of mammalian origin: 111. Properties
of a major feline allergen. J Immunol 1974;113: 1668-1677.
Andersson MC, Baer H. Allergenically active components of cat allergen extracts.
J Immunol 1981; 127:972-975.
Lowenstern H, Lind P, Weeke B. ldentification and clinical significance of allergenic
molecules of cat origin. Allergy 1985;40:430-44 1.
Ohman JL, Sundin B. Standard~zedallergenic extracts derived from mammals. Clin
Rev Allergy 1987;5:37-47.
Goubran-Botros H, Gregoire C, Rabillon J, David B, Dandeu J-P. Cross-antigenicity
of horse serum albumin with dog and cat albumins: study of three short peptides
with significant inhibitory activity towards specific human IgE and IgG antibodies.
Immunology 1996;88:340-347.
Andersson L, Sulkowski E, Porath J. Purification of commercial human albumin on
immobilized IDA-NiZ+.J Chromatogr 1987;421: 141- 146.
Dandeu J-P, Rabillon J, Gullaume J-L, Camoin L, Lux M, David B. Isolation of
Der pl, the Dermatophagoides pteronyssinus major mite allergen, from a crude mite
culture extract, purification by ion-chromatography, and comparison between the
material obtained and a cDNA-coded Der pl. J Chromatogr 1992;599:105-111.
Chapman MD, Platts-Mills TAE. Purification and characterization of the major aller-
gen from Dermatophagoides pteronyssinus-antigen PI. J Immunol 1980;125:587-
592.
Krilis S, Baldo BA, Basten A. Antigens and allergens from the common house dust
mite Dermatophagoides pteronyssinus. Part 11. Identification of the major IgE-bind-
ing antigens by crossed radioimmunoelectrophoresis. J Allergy Clin Immunol 1984;
74:142-146.
Lind P, Lowenstein H. Identification of allergens in Denriatophagoides pteronyssi-
Isolation of Allergenic Proteins 367
I. INTRODUCTION
In 1972, Huglin's collection [I] on light scattering from polymer solutions was
published. This was a full 2 years before Ouono and Kaye's classic paper [2]
reporting the combination of gel permeation chromatography (GPC, often called
size exclusion chromatography or SEC) and light scattering (IS). Although
Moore [3] had developed the GPC concept a decade earlier and James Waters
had launched his company a few years after that, nowhere in Huglin's text are
to be found references to the GPC concept. Indeed, Beckman did not introduce its
low-angle laser light scattering (LALLS) instrument until 1972, so this powerful
technique would remain but a curiosity until Chromatix licensed the concept from
Beckman and began their "missionary" work. Nevertheless, even without the
ability to fractionate samples before making LS measurements, LS techniques
themselves had reached a high degree of sophistication. Perhaps this was best
summarized for the field of biopolymers by Burchard and Cowie [4] in their
chapter entitled "Selected topics in biopolymeric systems." They stated that "an
attempt has been made to present a wide variety of examples which illustrate
most effectively the breadth of application of light scattering in this field. The
versatility of the technique is probably unique in providing information in depth
on biopolymers, and while it is by no means the only method in use, one hopes
it will become obvious to the unconverted that to neglect light scattering would
be to proceed under a distinct disadvantage." Although the numbers of "uncon-
369
370 Wyatt
verted" are still large not only in the field of biopolymers but in the broad areas
of organic polymers as well, the introduction of the DAWN instruments (which
implemented the concept of multiangle light scattering, or MALS, combined with
GPC) in the mid 1980s has begun to change those numbers. Referring back to
the various chapters in Huglin, it is of interest to note that huge areas of applica-
tion have yet to be examined by the combined GPCIMALS approach. It is the
object of this chapter to continue this "conversion" process while presenting some
important new results for biopolymers.
Despite the increased importance ascribed to LS methods during the past few
years, there remains a great deal of uncertainty and misunderstanding about the
subject. A few summary remarks are in order. We begin with the basic equation
used most frequently to describe the relation between what is measured and what
is derived. Equation (1) is the result popularized by Zimm [ 5 ] in the development
of his now classical technique by which he was able to generate key molecular
parameters, i.e., the weight average molar mass, Mw, mean square radius, (r:),
and second virial coefficient, A*, from measurements made on an unfractionated
sample comprised generally of an unknown distribution of molar masses and
sizes.
where
nated solution and solvent, respectively. The mean square radius is given by Eq.
(5) below, where the distances r, are measured from the molecule's center of
mass to the mass element m,:
Equation (5) applies to a single molecule, but the quantity measured from an
unfractionated ensemble is actually a light scattering average of the mean square
radii present in the sample. See Section IV, below, for further details.
Equation (1) is derived, to order c2, by Zimm from the more general [5,6]
reciprocal form:
In Eq. (6), A, is the third virial coefficient and Q(8) is a special form factor, of
magnitude always less than unity for finite 8, that describes additional light-scat-
tering contributions from the interacting molecules. At 8 = 0, both P and Q = 1.
Equations (1) and (6) are based on several approximations. First is the
assumption that the solvated molecules are well described by the Rayleigh ap-
proximation [6], i.e., that the refractive index (RI) of the molecules differs imper-
ceptibly from that of the solvent and that the solvated molecules do not affect
the phase of the (plane) wave of (polarized) light incident on the solution. Then
there are the assumptions that the concentration of the solute molecules is "van-
ishingly small" and that the molecules interact with each other at a single point.
Although the concentrations of samples after GPC fractionation are generally
very small, the assumption that very large molecules may interact only in the
single contact point approximation seems somewhat unreasonable. Remarkably,
the validity of these assumptions spans a surprisingly broad range of molar
masses.
Refemng to Eq. (I), we see some other important results of measurements
at very low concentrations. When the term 2A2c may be dropped at very low
concentrations, the mean square radius may be derived independently of any
knowledge of the concentration c or molar mass M. (See also Section IV for a
discussion of further limitations.) In this limit, Eq. (1) reduces to
K*c - 1
- -[I + a , sin' 812 - a2sin4 812 + ...I
R(8) MP(8) M
where a , is defined by Eq. (4).
Wyatt
Letting
Since the intercept at 5(0°) = 1/M,, the mean square radius is readily calculated
from the ratio of the slope to intercept. And that ratio is independent of M, and
c! Remember that 1/M, is calculated from the intercept, i.e., using the extrapo-
lated excess Rayleigh ratio, R(OO). So it does not matter what M, is actually
calculated: Set c = any arbitrary constant and one will generate an arbitrary M,.
Both M, and c drop out of the calculation. The important term is R(OO)and that
is obtained by extrapolation using mensured data.
Since the earliest days of on-line light scattering [2] there has been a considerable
amount of confusion as to the correct means for extracting the molar mass. At
vanishingly small concentrations and in the limit as the scattering angle, at which
the measurement is made. approaches zero, the excess Rayleigh ratio divided by
the product K*c becomes exactly equal to the weight average molar mass. This
is clearly seen from Eq. (7). In this manner, were the scattering angle taken small
enough, one could extract the molar mass of each eluting slice without having
to extrapolate the measurements following the methods developed by Zimm [ 5 ] .
The Beckman instrument (eventually developed further and marketed as the
Chromatix KMX6) made measurements at angles as low as 3" which, for all
intents, could be equated to 0" with only minor error up to molar masses well
into the millions.
Measurements at a single angle, of course, are insufficient to generate the
corresponding mean square radii, so the LALLS technique can at best generate
molar mass with no subsequent details of conformation or branching. The low-
angle measurement has other problems as well. In particular, the presence of any
dust or debris from column shedding results in additional signals throughout the
elution profile that often swamps the molecular signals themselves. Figure 1
shows the strip chart recording by Berkowitz [7] of the familiar broad polystyrene
standard NBS 706 illustrating the type of spike noise characteristic of such mea-
surements. (The Berkowitz paper describes various methods to suppress this
noise and try to restore the peak.) Figure 2 presents a three-dimensional plot of
MALS Combined with HPLC
TIME - TllE
Figure 1 Strip chart recording from raw LALLS data from N B S 706 and NBS 705.
the same standard showing the variation of the noise with scattering angle for a
separation in toluene. The data of Fig. 2, however, were obtained with a DAWN-
DSP system with a lowest angle collected around 24" in contrast to a 3" collec-
tion for the LALLS device. The point to emphasize here is that as the scattering
angle decreases, the associated noise generally increases. A scattering angle
closer to 0" does not necessarily mean that the extracted molar mass will be any
the more accurate. Indeed, there are some unexpectedly erroneous results that
can occur by ignoring the significance of the errors associated with the data col-
lected at any angle. Let us examine in more detail the extraction of molar mass
(and root mean square, or rms, radii, when attainable) from light scattering mea-
surements.
Figure 3 shows a set of MALS chromatograms at 16 angles (plus the Re-
fractive Index, RI, detector) for an aqueous GPC separation of a nominal 400
kglmol pullulan "standard." The actual scattering angles accessible (together
with their corresponding sin2 812 values) with a DAWN K5 glass cell are listed
in Table 1. The measurements of Fig. 3 were achieved without detector 1 (3.3").
The chromatography, as indicated by the "noise" at the smaller scattering angles,
is of rather poor quality, yet even the smallest scattering angle measured (14.5")
is considerably larger than the 3" data collected from the KMX6. The value of
sin2 812, however, is very small relative to the range of angles measured. Figure
4 (top) shows a Zimm plot for the single slice indicated by the vertical line in
the overlay plot (bottom) of the RI and 90" LS signals. For chromatographic
separations, the concentration at a given slice is assumed to be so small that the
second virial coefficient may be set equal to zero with negligible error. Thus the
Zimm plot has only a single concentration. The second virial coefficient may be
entered manually if required; however, since chromatographic separations are
occurring at a single concentration for each slice, the second virial coefficient
cannot be derived directly. Note that for rms radii up to about 40 nm, the plot
of K*c/R(B) versus sin2 812 is linear. The weighted fit of the data points to a
straight line will then yield the molar mass (intercept at sin2 812 = 0) and the
mean square radius (slope divided by the intercept times a constant). The ques-
tions that now might be asked include: How will these two quantities vary with
the lowest angle measured? Will they be more (or less) precise as the position
of the lowest angle measured is varied? Indeed, does the low angle even add
additional information?
A least-squares fit of a set of data points to a straight line is a straightfor-
P
Wyatt
ward process. The minimization of the sum of the squares with respect to the
slope a and intercept b (= 1 IM) yields the pair of equations:
and
Y2 - Y I
a =- and b =
x2y1 - yzxl
X2 - X, x2 - X I
Wyatt
The choice of a very small angle as one of the two points through which
the straight line is to be constructed indicates, from Eqs. (14) and (15), that the
error in the derived molar mass, M, will increase as the lower angle goes to zero.
For noisy chromatography, the fluctuations of R(sin2 01/2)will increase as 0 +
0. Accordingly, if only two angles are chosen for a LS measurement, the selected
angles should be as far away from 0" as practical.
For the case of three angles, where the lowest angle measured is also ex-
pected to have a correspondingly less precise value than either of the other two,
replacing only the lowest angle measured with an even smaller angle in no way
implies that the final values will be more precise. On the contrary, keeping mea-
surements away from 0" will generally increase the precision of the determina-
tion. Furthermore, as should be evident from Eqs. (10) and ( l l ) , the fit to the
straight line will now be a true least-squares fit and the weighted data will result
in the "best" fit being selected.
As might be expected, as the number of angles included in the final calcula-
tions is increased, the precision of both molar mass and mean square radius is
increased. Table 2 presents the relative precision of these final values for the case
of two (very low angle and 90°), three (approximately 40°, 90°, and 140°), and
many (see Table 1) angles for the example of Figs. 2 and 3.
One of the possibilities associated with making LS measurements with too
few angles lies in the hope of extending the range accessible by assuming a model
of the molecules measured. For certain types of models, the form factor, P(0),
is known a priori so there is no need to fit the data to a straight line or higher
MALS Combined with HPLC
order polynomial for the subsequent determination of molar mass and rms radius.
Of course, such an assumption flies somewhat in the face of the concept of an
"absolute" measurement. Nevertheless, we see frequent use of this assumption
either directly or implicit in some software programs. The latter assumption is
insidious in that data are processed and results calculated without the possibility
of user intervention. If a random coil model is built into the software without
the user's knowledge, then there is no basis left for the user to understand why,
for example, the results contradict the results of other measurements such as
thermal analysis. Let us examine the random coil model and its dual-angle imple-
mentation in greater detail.
For a random coil molecule, the form factor P(8) takes on the simple closed
[5] form:
where
where P(0) is given by Eq. (16). Equation (19) has two unknowns, M u and
(ri). and thus again like the straight line fit of Eqs. (12), the number of data are
equal to the number of degrees of freedom. The average values of R(0) measured
at 0 , and O2 are the only data available for this fit, i.e., R(0,) and R(02). The
nonlinear Eq. (19) in the two unknowns. M, and (ri), has (at best) a single soh-
tion. Let the two measured excess Rayleigh ratios be 5, = R(0,) and c2 = R(0?).
Equation (19) is best solved first by eliminating M u i.e.:
where
One would generally begin by solving Eq. (20) for (ri) which value is then substi-
tuted in Eq. (16) to obtain P(0) at the angle for which R(0) has the smallest
standard deviation. This value and the corresponding R(0) are then substituted
in Eq. (19) to solve for M,. The most "accurate" value of M, derived from Eq.
(19) would best be calculated at the angle with the least noise (the largest, 02),
once (ri) had been calculated from Eq. (20). But note that the nonlinear calcula-
tions generating (ri) from Eq. (20) will have a precision that depends on the
measured quantities 5, = R ( 0 , ) andc2 = R(02).To maintain the greatest preci-
sion in this derivation of (ri), we see again that if only data at two angles are to
be collected, the angles should be chosen as far as possible from the small, noisier,
forward angles. Once a model is chosen, the smaller angle fits to that model add
to the errors of the final results as the experimental uncertainty of those data.
The precision of such a determination will depend not only on how small the
noise is at the angles selected but on the number of different angles measured
as well. Once again, the best way to fit a model such as the random coil model
is to extract the key physical constants by fitting weighted data in a least-squares
sense. Thus one must use as many angles as possible and certainly at least three.
In closing this section, it is worth remarking that traditional methods for
determining functional parameters by comparing with experimentally derived
data require that the number of such data exceed substantially the number of
MALS Combined with HPLC 381
functional parameters or degrees of freedom. Only on this basis can one perfom1
a "chi-squared" goodness-of-fit analysis and thereby establish the quality of the
selected model (function) by which the data are governed. Any model must be
tested at points throughout its range of application. With only two data points
and two angles, the results generated will depend always on the model chosen.
One often reads that the rms radius measured by light scattering [8] is actually
a z-average radius. (The misnomer "radius of gyration" is frequently found in
the literature to describe this same quantity.) It is important to understand just
what these so-called moments of the distributions mean since they play an impor-
tant role for a variety of analyses, not the least of which is the determination of
a sample's polydispersity. In addition, concepts such as the measurement and
quantitation of branching require that the moments be identical within each suit-
ably fractionated slice of a GPC separation. We begin by considering the MALS
measurement of a polydisperse sample. We may rewrite Eq. (7), at vanishingly
small concentrations and small angle, to obtain the excess Rayleigh ratio mea-
sured from the suspension:
where M, is the weight-average [see Eq. (25)] molar mass, c = Cc,, and K =
2molh0,as before. In the limit as 0 + 0, the MALS measurement yields the
weight-average molar mass, i.e., R(OO)= K*M,c. Note that the constant K* has
been factored out from the summations over the species present based on the
further assumption that the polymer is a homopolymer, or at the very least that
the dnldc value is the same for all species present (including, for example, homo-
geneous copolymers). In the event that a copolymer or polymer blend has been
separated, the mean square radius is not so easily extracted. A more detailed
discussion of the modifications to the LS formulation may be found in the paper
by Benoit and Froelich [9].
Wyatt
by definition. In other words, the z-average mean square radius has been dejned
by means of a weighting involving both mass and concentration. Let us examine
this and related moments more carefully.
The mass moments of a distribution are given traditionally by
for the number-average molar mass, where ni is the number of molecules of mass
M, and the summations are over all of the i species present; the concentration c,
of the ith species therefore is equal to Mini;
for the z-average molar mass. Of course, Eqs. (24) and (25) have an associated
physical meaning in terms of numbers and weights. Each corresponds also to an
absolute mass measurement technique. Light scattering is the absolute method
of choice to measure the weight-average molar mass while osmometry is the
absolute technique of choice to measure the number-average molar mass of a
sample directly. Equation (26) corresponds to the quantity measured by the other
absolute measurement technique, sedimentation equilibrium. The subscript "z"
has its origin in the German word for centrifuge, Zentrijuge. From Eq. (26) it is
a simple matter to write expressions for the z + 1, z + 2, etc., moments. Note
how these "moments" are related. In particular, each higher moment corresponds
to "the next higher weighting" of both numerator and denominator by the factor
M;.
MALS Combined with HPLC 383
The number- and weight-average values of the mean square radius, (ri),
are easily written in terms of their definitions i.e.:
for the number-average mean square radius, i.e., the mean square radius of each
species is weighted by the number, ni, of such members present. So far so good,
but to calculate the weight-average mean square radius we must weight the mean
square radius by the concentration, ci, of each species present, i.e.:
produces three equations [(30), (28), and (29)] that display the same type of mo-
ment relationship shown in Eqs. (24), (25), and (26). However, note that once
again the formal moments [6] of the mean square radii are with respect to the
masses MI and not the mean square radii. Note that for the special case of a
random coil structure at the theta point, M I a (ri). Replacing (r;) by M, in Eqs.
(30), (28), and (29) yields Eqs. (24) through (26), respectively. In order to empha-
size that the size moment measured by light scattering is not a true z average,
384 Wyatt
except for the rare occasion that the molecules being measured are random coils
in a theta solvent, the term LS average should be used in the future, viz.
I HEPARIN
x'
sample. Both show the mass elution curves reversing their downward trends and
turning upward at larger elution volumes. In both cases the mobile phase was
aqueous. Note that these results were obtained via a MALS measurement com-
bined with an RI detector to monitor the concentration. The RI elution curves
are shown in the background. The types of mass elution calculated from MALS
would not have been calculated from standard GPC plus column calibration. Only
an absolute measurement would have disclosed this unusual elution behavior
since calibration techniques invariably require a set of standards that produce
decreasing molar mass with increasing elution.
A variety of explanations have been presented for the non-GPC effects seen
here, though without MALS they are rarely detected. Among them are column
interactions and the presence of microgels. The former effects are akin to affinity
chromatographic interactions. The latter are based on the fact that highly compact
structures with higher molar mass would be expected to elute at later times be-
cause their hydrodynamic size, for their greater mass, could be considerably
smaller than their noncompact companions. But irrespective of the possible expla-
nation, the mere presence of these fractions requires that they be included in the
mass distributions reported as they often represent a considerable fraction of the
386 Wyatt
sample on column. The most powerful means of presenting the results of a GPC
separation is to produce a differential weight fraction distribution. From such
a result, the cumulative distribution in addition to the various moments of the
distribution may be calculated. The differential weight fraction distribution also
represents the most direct means by which samples may be differentiated and
column performance evaluated. Let us look at these distributions in greater detail,
especially when applied to the results of Figs. 5 and 6.
The differential mass fraction distributions are generally presented as a
distribution in log,, M rather than M. This permits the details of distributions
spanning several orders of magnitude to be clearly visible. Perhaps most impor-
tantly, this type of presentation arose because the calibration curves are generally
prepared with standards spanning a few orders of magnitude. An ideal GPC col-
umn set will separate linearly with respect to the logarithm of the hydrodynamic
size. Since the logarithm of the hydrodynamic radius is proportional to the loga-
rithm of the molar mass for linear molecules, the presentation of distributions in
these terms has become an accepted practice. Let us review the definitions of
these quantities.
Following Shortt 11I],we define W(M) as the cumulative weight fraction
of molecules in the selected peak whose mass is less than M. This is the total
mass of molecules of mass less than M divided by the total mass of the molecules
present. Thus:
For normal separations by the GPC mechanism (size exclusion), the term
dW/dV may be replaced by the concentration detector's response. Thus if the
concentration detector's baseline subtracted response is h(V), then dW/dV =
-h(v)/I h(V)dV, the integral representing the sum over all contributing concen-
trations. The differential weight fraction has thus been replaced [I I] by the nor-
malized concentration fraction. Note the negative sign which corresponds to the
1.ox1oF 1.OXI o7
Molar Mass (glrnol)
Figure 7 Differential mass fraction distribution for the polyacrylamide sample of Fig.
5 formed by re-sorting the slice collected data into bins in log,, M.
388 Wyatt
fact that the variation of the slope of the "calibration curve," d(log,, M)/dV, is
negative since larger volumes correspond to smaller molar masses. The differen-
tial ("log base 10") mass fraction is then written as
If the term d(log,, M)/dV is linear, then the differential fraction, x(M) is just a
rescaled concentration curve. As long as d(logloM)/dV remains monotonically
decreasing, the formalism of Eq. (37) is reasonable. However, when the variation
of d(logloM)/dV becomes zero and even reverses its slope, such as shown in
Figs. 5 and 6, the results are both difficult to understand and could be in error.
For example, in Fig. 5, a simple fifth-order polynomial does not even fit the data,
so that the results based on Eq. (37) may be in considerable error because the
larger mass fractions that elute irregularly (non-GPC) are not accorded their
proper contributions to the distribution of masses present in the sample. Let us
examine these results in more detail.
-.-
*
+
+ * PAM91-1
+ Norm = Log
3rd order
i
,2.0-
.-0
4- -
0
F -
L
E -
.-m
-
3
.- 1.0-
3
4-
C
2 -
w
E -
n
0.0- I 1 1 1 1 1
1 .OXI o6 1.ox1o7
Molar Mass (g/rnol)
Figure 8 Differential mass fraction distribution for the polyacrylamide sample of Fig.
5 using a third-order fit to the mass elution data. Note the double values about the point
of reversal of molar mass versus elution volume, i.e., where d ( l ~ gM)/dV
,~ = 0.
MALS Combined with HPLC 389
We begin again with the formalism of Eq. (34). At each elution volume
V, there is a corresponding mcasurcd value of M as well as a baseline corrected
concentration response h(V). The procedure to generate the differential weighted
log base 10 mass fraction, x(M), begins weighting the calculated mass M, by
(log, 10) h(V,)/C[h(V,)AV,],where the sum is taken over all i slices present in
the selected peak being processed. These values are then re-sorted into mass bins
divided in terms of a log,, M scaling. In this manner, similar mass contributions
at different elution volumes would be added together. Applied to the data of Fig.
5, for example, this procedure yields the very discontinuous result of Fig. 7.
Because of the uncertainty associated with the calculation of molar mass (the
ASTRA program calculates the precision of each measurement reported), one
might well consider distributing the mass fraction value into a range of bins rather
than into a single mass bin. Thus if the standard deviation of the calculated mass
is AM, then the mass fraction might be distributed following a Gaussian distribu-
tion over a range of bins spanning at least +2AM. Even if this procedure were
successful, the resulting distributions would still appear strange, and for good
PAMY 1
Norm = Log
3rd order
PAM91-2
1.OXI o6 1.OXI o7
Molar Mass (glrnol)
Figure 9 Overlay of the resorted data of Fig. 7 with the analytically fit data of Fig. 8.
390 Wyatt
Volume (mL)
Figure 10 The root mean square radius as a function of elution volume for the sample
of Fig. 5.
MALS Combined with HPLC 391
I .ox106 I .ox1o7
Molar Mass (glrnol)
Figure 11 The conformation plot for the polyacrylamide data of Figs. 5 and 10.
the point of the rms radius reversal. Figure 11 presents the conformation plot for
this molecular sample, which appears quite normal for molecules eluting before
the reversal point, yet which appears very aberrant and distorted for those frac-
tions eluting later in the so-called non-GPC mode. Matsumoto [I21 was the first
to associate the unusual appearance of the conformation plot with the onset of
microgel formation. Whatever the source, the MALS confirms that there is pres-
ent a fraction of the sample whose conformation is distinctively different than
that of a linear or even slightly branched polymer. Conformation plots and differ-
ential mass fraction distributions tell much about samples whose elution from
GPC columns don't follow the rules.
tially unity. In other words, a protein such as bovine serum albumin is produced
at a single molar mass only as determined by the corresponding genetic code.
Naturally such proteins can form aggregates whose presence in various biologi-
cals can be immunogenic. Thus the detection of dimers, trimers, and higher order
aggregates can become an important task for the quality control of many types
of commercially sold biologicals.
Let us consider a few examples of the power of MALS combined with
differential refractive index (DRI) detection. In a recent paper by Riggs's group
at the University of Texas [13], the use of MALS with a DRI detector produced
the values for the broad range of values shown in Table 3. The values calculated
from the MALS measurements (DAWN-DSP operating at the He-Ne laser wave-
length 632.8 nm) are contrasted to the values calculated based on the expected
base pair sequences. A single value of dnldc (0.19) was used in their measure-
ments using a phosphate buffer saline mobile phase and two Toya Soda GPC
columns described in the article.
Light scattering measurements are particularly useful for proteins whenever
a DRI detector can be used due to the near-constancy of dnldc. This is in contrast
to reversed-phase chromatography where a UV detector must be used because
of the varying refractive index of the mobile phase. This in turn requires the
measurement of the corresponding UV extinction coefficient for each protein
species. For certain closely related proteins, where a single extinction coefficient
may be chosen, the light scattering results for determining molar masses of sepa-
rated species is equally impressive. Figure 12 shows the UV chromatogram from
a set of isolated wheat proteins whose corresponding sequence values are shown
in Table 4. These proteins were separated by reversed-chromatography using a
gradient of acetyl nitrate in water over the range of 0 to 15%. What is particularly
significant about this type of separation is that the elution sequence is not gener-
0 5 10 15
Volume (mL)
Figure 12 UV chromatogram from a set of isolated wheat proteins whose molar mass
and size were calculated from MALS measurements. See Table 4 for details.
ally in any particular order. There is no means to calibrate the columns used for
reversed-phase separation as is often the case for GPC separations.
Figure 13 illustrates an important element of MALS combined with a con-
centration-sensitive detector. Shown here are three differential mass fraction dis-
tributions [cf. Eq. (37)] of specially processed recombinant DNA-produced hya-
loronic acid. Although all overlap broadly with each other, the direct comparison
of their differential mass fractions permits an easy discrimination between them.
Finally, the process of adding polyethylene glycol (PEG) to proteins (so-
called pegylation), which has become of considerable importance as a time re-
lease drug delivery process, produces a copolymer whose core is a pure, monodis-
perse protein molecule. By iteratively calculating the copolymer mass at each
elution volume (note that each component has a different d d d c value), staff
- Refractometer
Signal overlaid
Figure 14 Variation of molar mass with elution volume for a pegylated protein sample.
Used a self-consistent iterative method to calculate the number of PEGIprotein.
MALS Combined with HPLC 395
member Robert Paulson in an unpublished Application Note [14] has been able
to calculate not only the total molar mass of each eluting fraction but the number
of PEG molecules (molar mass about 15,000) attached to each protein molecule
(molar mass about 150,000), as illustrated in Fig. 14.
ACKNOWLEDGMENTS
This chapter is based on a paper presented at the Waters GPC Symposium, Sep-
tember 1996, San Diego, CA. The exceptional laboratory results, especially the
data shown in Figs. 10 through 12, were made under the direction of Dr. Michelle
H. Chen. Other individuals who made contributions include Dr. Greg Cauchon,
Lena Nilsson, and David Villalpando.
REFERENCES
1. Huglin MB, ed. Light Scattering from Polymer Solutions. London: Academic Press,
1972.
2. Ouano AC. Kaye W. J Polym Sci. A-l 1974; 12:1151.
396 Wyatt
I. INTRODUCTION
The term connective tissue growth factor (CTGF) was first used in 1991 to de-
scribe a mitogenic and chemotactic factor for fibroblasts that was produced by
human umbilical vein endothelial cells (HUVECs) in vitro [ I 1. The primary trans-
lational products of both human CTGF (hCTGF) and porcine CTGF (pCTGF)
are predicted to comprise 349 residues, the first 26 of which are a presumptive
signal peptide [1,2]. Mouse CTGF (mCTGF; also termedjsp-12 or PIG-M2) is
predicted to comprise 348 residues, of which the first 25 are a hydrophobic signal
peptide [3,4]. Secreted forms of CTGF from all three species are thus predicted
to comprise 323 residues, 38 of which are conserved cysteine residues. Metabolic
labeling and immunoprecipitation studies of cell lysates and conditioned medium
have shown that CTGF proteins of approximately 38 kDa are produced and se-
creted in various cell cultures [2,5,6]. CTGF, which is encoded by a transforming
growth factor-P (TGFP)-inducible immediate early gene [3], has been strongly
implicated in a variety of fibrotic disorders [7-I I], wound healing [12-141, em-
bryonic development [ 5 ] , and uterine function [2,15]. Although the 38-kDa form
of CTGF was initially implicated in many of these processes, low-mass forms
of CTGF of M, 10,000-20,000 have since been discovered in uterine secretory
fluids [ 151 and fibroblast conditioned medium [6].
397
398 Brigstock
6. Chromatography
1. Cation Exchange Chromatography
ULF were applied at 3.5 mllmin to a Bio-Rex 70 cation exchange column
(5 X 6 cm; Bio-Rad Laboratories, Richmond, CA, USA) which was then washed
with 1000 ml of PBS containing 0.2 M NaCl and subsequently developed with
a 500-ml gradient of 0.2-2.0 M NaCl in PBS. These procedures were performed
at 4°C. Fractions (10-ml) were collected during salt gradient elution of bound
proteins and were assayed for their ability to stimulate [3H]thymidineincorpora-
tion in Balblc 3T3 cells. The 0.3-0.7 M NaCl elute was selected for further
analysis by heparin affinity FPLC.
Purification and Characterization of CTGF
C. Protein Sequencing
The HPLC-purified IOkDa protein was evaporated to dryness using a SpeedVac
concentrator, redissolved in SDS-PAGE sample buffer, subjected to SDS-PAGE,
transferred to nitrocellulose, and located by staining the blot with Coomassie
R250. The protein was then excised and subjected to N-terminal microsequencing
on a model 470A gas phase sequenator (Applied BioSystems, Foster City, CA,
USA). Phenylthiohydantoin derivatives in each cycle were separated using C18
RP-HPLC. The sequence was assigned using Applied BioSystems sequence anal-
ysis software, with independent verification by an experienced operator with no
knowledge of the likely identity of the sample.
E. Bioassay
Column fractions were tested for their stimulation of [3H]thymidine incorpora-
tion into the DNA of quiescent Balblc 3T3 cells. Neutralization studies were
performed by coincubation of the 0.8 M NaCl heparin column eluate with
25 pglml anti-PDGF IgG (Upstate Biotechnology Inc., Lake Placid, NY, USA)
or 10 pllml basic fibroblast growth factor (bFGF) antiserum ("77R," kindly
donated by Dr. M. Klagsbrun, Children's Hospital, Boston, MA, USA). PDGF
and bFGF standards were from R&D Systems (Minneapolis, MN, USA) and Life
Technologies (Grand Island, NY).
Purification and Characterization of CTGF
Ill. RESULTS
A. Ion Exchange Chromatography and Heparin
Affinity FPLC
When ULF were applied to a Bio-Rex 70 column, the flow-through fraction was
shown to contain high-mass forms of epidermal growth factor (EGF) that stimu-
lated DNA synthesis in 3T3 cells [22]. However, material which bound to the
column and was eluted by 0.3-0.7 M NaCl was also shown to be mitogenic for
3T3 cells (Fig. 1). Affinity chromatography of this eluate using EconoPac heparin
NaCl (M)
A 0.0
0 10 20 30 40 50
Fraction Number
columns resulted in the elution of four peaks of growth factor activity over a
0.2-2.0 M NaCl gradient (Fig. 2). The first activity peak was eluted by 0.5 M
NaCl and shown to contain a PDGF-like factor [23]. The second activity peak
was eluted by 0.8 M NaCl and contained a novel factor that was termed HBGF-
0.8 prior to its definitive identification as a 10-kDa form of CTGF [15]. The third
activity peak was eluted by approximately 1 M NaCl and contained heparin-
binding EGF-like growth factor (HB-EGF) and pleiotrophin (PTN) [23,24]. The
fourth activity peak was eluted by 1.6 M NaCl and was due to the presence of
bFGF [I 5,23,25].
To further purify 10-kDa CTGF, fractions containing the 0.8 M NaCl eluate
from the EconoPac heparin column were pooled, diluted, and subjected to a sec-
ond step of heparin affinity FPLC using a TSK heparin column. A peak of growth
factor activity was subsequently eluted by 0.8 M NaCl (Fig. 3), which in some
separations could be resolved into two microheterogeneous forms of 10-kDa
CTGF [15]. To show that 10-kDa CTGF was immunologically distinct from
NaCl (M)
0.2 0.65 1.1 1.55 2.0
0 10 20 30 40
Fraction Number
Figure 2 First-step heparin affinity FPLC. Mitogenic fractions from cation exchange
chromatography of 900 ml of ULF from 22 animals were pooled, diluted to three-fold,
and applied to an EconoPac heparin column, which was then washed with 10 m120 mM
Tris-HClf0.2 M NaC1/0.1% CHAPS (pH 7.4) and developed with a 40-ml gradient of
0.2-2.0 M NaCl in 20 mM Tris-HC1/0.1% CHAPS (pH 7.4). Fractions (1-ml) were col-
lected during NaCl gradient elution of the bound proteins and tested at 15 yllrnl for their
stimulation of [3H]thyrnidineincorporation in Balblc 3T3 cells. The figure shows the iden-
tity of various heparin-binding growth factors that were separated from one another.
Purification and Characterization of CTGF
NaCl (M)
0.2 0.65 1.1 1.55 2.0
300 1 I I 1 0.60 1
Fraction Number
Figure 3 Second-step heparin affinity FPLC. Nine individual samples of ULF (7.15-L
total volume from 206 pigs) were individually processed by cation exchange and EconoPac
heparin affinity chromatography as shown in Figs. 1 and 2. The 0.8 M NaCl eluates from
each heparin affinity purification were pooled, diluted, and applied to a TSK heparin col-
umn which was washed with 10 ml of 20 mM Tris-HC110.2 M NaCl (pH 7.4) and devel-
oped with a 40-ml gradient of 0.2-2.0 M NaCl in 20 mM Tris-HCI (pH 7.4). Fractions
(I-ml) were collected during NaCl gradient elution of the bound proteins and tested at 5
pllml for their stimulation of ["Hlthymidine incorporation in Balblc 3T3 cells.
bFGF or PDGF and was not contaminated by either of these factors, it was incu-
bated with neutralizing antisera to PDGF or bFGF. As shown in Table 1, CTGF
mitogenic activity was unaffected by either of these antisera, which were nonethe-
less very effective in antagonizing the activity of their respective ligands.
8. Reversed-Phase HPLC
Isolation of 10-kDa CTGF to homogeneity was achieved by performing two steps
of C, RP-HPLC on heparin-purified samples. Results from the second HPLC step
are presented in Fig. 4 that show the elution of a single peak of growth factor
activity at 45 min (28% acetonitrile). The elution profile of the mitogenic activity
was directly correlated with that of a single peak of protein, which was of M,,
10,000 as assessed by SDS-PAGE. Based on silver staining of SDS polyacryl-
amide gels, no proteins other than the 10-kDa polypeptide were present in any
of the biologically active fractions (Fig. 4).
Brigstock
Figure 4 Second-step RP-HPLC. Two peak active fractions from first-step HPLC puri-
fication (data not shown) were subjected to second-step HPLC purification as described
in "Materials and Methods." The figure shows the elution of protein as measured at 3-14
nm and the ability of 20 FI of selected fractions to stimulate 3T3 cell DNA synthesis after
drying and reconstitution in 10 pl PBSIO.I% BSA. The inset shows a silver-stained SDS
polyacrylamide gel of 50-yl aliquots of successive fractions containing the peak of rnito-
genic activity.
406 Brigstock
mitogenic activity in the 0.8 M NaCl eluate after two rounds of heparin affinity
FPLC showed that ULFs from pigs on day 11 of pregnancy contained approxi-
mately 5 times the level of CTGF-like activity as ULF from pigs on day 11 of
the estrous cycle (Fig. 7).
Figure 5 Heat and acid treatment of 10-kDa CTGF. Heparin-purified 10-kDa CTGF
was incubated at pH 7.4 for 30 min at room temperature (a),pH 7.4 for 30 min at 56°C
(U), pH 7.4 for 5 min at 100°C (m), or pH 1.5 for 3 min at room temperature (0).Sanlples
were assayed for their stimulation of 3T3 cell DNA synthesis at equivalent doses. Values
shown are mean +SD of triplicate determination of [%]thymidine incorporation.
No IOkDa CTGF
addition 20pllrnl
Figure 6 Stimulation of pig endometrial cell DNA synthesis by 10-kDa CTGF. Primary
cultures of pig endometrial cells were treated for 18 h with 20 pllml heparin-purified
10-kDa CTGF (a concentration that induced maximal stimulation of DNA synthesis in
Balblc 3T3 cells) after which they were incubated with I pCi/ml ['Hlthymidine for 6 h.
Brigstock
Fract~onNumber
Figure 7 Heparin affinity FPLC of ULF from day 11 pregnant or cycling pigs. ULF
from day I I pregnant (@) or cycling (0) pigs (n = 6 per group) were subjected to two
rounds of heparin affinity purification essentially as shown in Figs. 2 and 3 except that
no Bio-Rex purification was performed and the first and second heparin affinity steps were
performed using heparin-Sepharose and EconoPac heparin columns, respectively. The
figure shows the stimulation of 3T3 cell DNA synthesis by selected fractions from the
second heparin affinity step when assayed at 2.5 pllml on 3T3 cells. The levels of CTGF
in ULF were 45.1 units in pregnant animals versus 8.8 units in nonpregnant animals, where
one unit is the amount of each growth factor per ml that was required to elicit half the
['Hjthymidine incorporation as 20% calf serum.
translational product (i.e., the N terminus of 10-kDa CTGF; see Table 2) was
subjected to heparin affinity FPLC. Although the pI of this peptide is the same
as that of PDGF (9.6) which is eluted from heparin affinity columns by 0.5 M
NaCl 133,341, the CTGF peptide required 0.7 M NaCl for elution from a TSK
heparin column (Fig. 8). These results showed that the peptide bound more
strongly to heparin than could be explained by its isoelectric point alone and
were suggestive of the presence of specific heparin-binding determinants in the
peptide.
IV. DISCUSSION
A. Heparin Affinity Purification of CTGF
Over the last 15 years, heparin affinity chromatography has proven a particularly
powerful means by which a variety of polypeptide growth factors have been
characterized and purified. These include acidic FGF (aFGF), bFGF, HB-EGF,
Purification and Characterization of CTGF
Fraction Number
PTN, vascular endothelial growth factor, keratinocyte growth factor, and amphi-
regulin [35-411. Since many of these factors exhibit different affinities for hepa-
rin, the relative binding of these factors to heparin has been a particularly useful
criterion for characterizing growth factor of unknown identity, especially when
used in conjunction with immunological (e.g., western blot) and biological (e.g.,
activity, target cell specificity) assays. Indeed, CTGF was first recognized as a
fibroblast rnitogen in uterine secretory fluids in 1989 as a result of its atypical
chromatographic behavior on heparin-agarose [25]. Key features that were subse-
quently used to distinguish it from other well-characterized growth factors (in-
cluding FGFs with which it was initially thought to be related [25]) were its !I)
elution from heparin by 0.8 M NaCI, (2) molecular mass of 10,000 as determined
by nondenaturing gel filtration and SDS-PAGE, (3) target cell specificity (mito-
genic for fibroblasts and smooth muscle cells but not endothelial cells), (4) heat
and acid lability, (5) potentiation of bFGF-, EGF-, PDGF-, or IGF-induced mito-
genic activity, and (6) lack of reactivity with PDGF or bFGF antisem (these
results and [15]). However, it was not until the development of this purification
protocol that the factor was isolated and definitively identified as a 10-kDa form
of CTGF. Heparin affinity chromatography not only played an important role
in the initial purification and characterization of 10-kDa CTGF, but also in the
separation of two microheterogeneous forms of the protein that differed by the
410 Brigstock
for the degree of heparin binding observed since 10-kDa CTGF has a lower iso-
electric point than PDGF (PI 9.6) yet requires a higher salt concentration than
PDGF for elution from heparin (0.8 M NaCl versus 0.5 M NaC1). As we have
previously reported, residues 250-255 resemble a heparin-binding consensus se-
quence and there is a very high proportion (50%) of basic amino acids (K, R)
in the 17-residue region CTGF[251-2671 1151. Evidence that the N terminus of
10-kDa CTGF may be involved in heparin binding is illustrated by ( I ) the require-
ment for 0.7 M NaCl to elute the peptide CTGF[247-2601 from a TSK heparin
column, which, by comparison to PDGF, is a higher salt concentration than would
be predicted from its pI alone; (2) the observation that CTGF[247-2601 binds
[3H]heparin more strongly than peptides that flank this sequence 1151; and (3)
the finding that a form of 10-kDa CTGF that commences at G ~ u ~exhibits
''~ greater
heparin binding than the form described here, which commenced at G ~ (and u ~ ~ ~
thus differs by the presence of a single acidic residue at its N terminus) [15].
While these data suggest that residues 247-260 are involved in heparin binding,
analysis of [3H]heparinbinding by a panel of peptides that spanned the C-terminal
103 residues of CTGF suggested that additional domains, including residues 305-
328, may also be involved in the interaction between 10-kDa CTGF and heparin
1151. While it has been proposed that residues 206-214 of CTGF resemble a
binding motif for sulfated glycoconjugates 1261, the involvement, if any, of this
domain in mediating interactions between larger forms of CTGF (e.g., the full-
length 38-kDa CTGF protein) and heparin has yet to be clarified.
D. Conclusions
Heparin affinity chromatography has played a key step in the isolation of native
CTGF from uterine secretions and recombinant CTGF from baculovirus expres-
sion systems. While the interaction of CTGF with heparin has been exploited for
the purification of 10-kDa and 38-kDa forms of CTGF, this property appears to
be the result of the presence of specific heparin binding motifs within the CTGF
molecule and may have physiological consequences in that heparin is a regulator
of CTGF biological activity. Future studies will undoubtedly focus on additional
structural and functional aspects of heparin binding by CTGF, including the pos-
sible role that cell surface heparan sulfate proteoglycans play in regulating the
bioavailability of CTGF and its binding and activation of cell surface receptors.
ACKNOWLEDGMENTS
I am grateful to Christy Steffen and Greg Kim for excellent technical help, Bill
Pope for providing timed cyclic and pregnant pigs, John Lowbridge for protein
412 Brigstock
sequencing, and Brad Baker for peptide synthesis. This work was supported by
NIH Grant HD30334 awarded to D.R.B.
REFERENCES
Bradham DM. Igarashi A, Potter RL, Grotendorst GR. Connective tissue growth
factor: a cysteine-rich mitogen secreted by human vascular endothelial cells is re-
lated to the SRC-induced immediate early gene product CEF-10. J Cell Biol 1991;
1 14:1285- 1294.
Harding PA, Surveyor GA, Brigstock DR. Characterization of pig connective tissue
growth factor (CTGF) cDNA, mRNA and protein from uterine tissue. DNA Se-
quence 1998;8:385-390.
Brunner A, Chinn J, Neubauer M, Purchio AF. Identification of a gene family regu-
lated by transfom~inggrowth factor-p. DNA Cell Biol 199 1 ;10:293-300.
Ryseck R-P, Macdonald-Bravo H, Mattei M-G, Bravo R. Structure, mapping and
expression of fisp- 12, a growth-factor-inducible gene encoding a secreted cysteine-
rich protein. Cell Growth Differ 1991;2:225-233.
Kireeva ML, Latinic BV, Kolesnikova TV, Chen C-C, Yang GP, Abler AS, Lau
LF. Cyr61 and Fispl2 are both ECM-associated signalling molecules: activities. me-
tabolism, and localization during development. Exp Cell Res 1997:233:63-77.
Steffen CL, Ball-Mirth DK, Harding PA, Bhattacharyya N, Pillai S, Brigstock DR.
Characterization of cell-associated and soluble forms of connective tissue growth
factor (CTGF) produced by fibroblast cells in vitro. Growth Factors 1998;15:199-
213.
Frazier KS, Grotendorst GR. Expression of connective tissue growth factor mRNA
in the fibrous stroma of mammary tumors. Int J Biochem Cell Biol 1997;29:153- 161.
Igarashi A, Nashiro K, Kikuchi K, Sato S, Ihn H, Grotendorst GR, Takehara K.
Significant correlation between connective tissue growth factor gene expression and
skin sclerosis in tissue sections from patients with systemic sclerosis. J Invest Der-
mat01 1995; 105:280-284.
Igarashi A, Nashiro K, Kikuchi K, Sato S, Ihn H, Fujimoto M, Grotendorst GR,
Takehara K. Connective tissue growth factor gene expression in tissue sections from
localized scleroderma, keloid, and other fibrotic skin disorders. J Invest Dermatol
1996;106:729-733.
Oemar BS, Werner A,, Gamier J-M, Do, D-D, Godoy N, Nauck M, Marz W, Rupp
J, Pech M, Luscher TF. Human connective tissue growth factor is expressed in ad-
vanced atherosclerotic lesions. Circulation 1997;95:83 1-839.
Shinozaki M, Kawara S, Hayashi N, Kakinuma T, Igarashi A, Takehara K. Induction
of subcutaneous tissue fibrosis in newborn mice by transforming growth factor P-
simultaneous application with basic fibroblast growth factor casues persistent fibro-
sis. Biochem Biophys Res Commun 1997;237:292-296.
Igarashi A, Okochi H, Bradham DM. Grotendorst GR. Regulation of connective
tissue growth factor gene expression in human skin fibroblasts and during wound
repair. Mol Biol Cell 1993;4:637-645.
13. ~ a m m e MS,
s Lieske JC, Pawar S, Spargo BH, Toback FG. Calcium oxalate mono-
Purification and Characterization of CTGF 413
hydrate crystals stimulate gene expression in renal epithelial cells. Kidney Int 1995;
48:501-509.
Pawar S, Kartha S, Toback FG. Differential gene expression in migrating renal epi-
thelial cells after wounding. J Cell Physiol 1995;165:556-565.
Brigstock DR, Steffen CL, Kim GY, Vegunta RK, Diehl JR, Harding PA. Purifica-
tion and characterization of novel heparin-binding growth factors in uterine secretory
fluids: identification as heparin-regulated 10,000-M,, forms of connective tissue
growth factor. J Biol Chem 1997;272:20275-20282.
Frazier K, Williams S, Kothapalli D, Klapper H, Grotendorst GR. Stimulation of
fibroblast cell growth, matrix production, and granulation tissue formation by con-
nective tissue growth factor. J Invest Dermatol 1996;107:404-411.
Anderson LL. Growth, protein content, and distribution of early pig embryos. Anat
Rec 1978;190:143-154.
Geisert RD, Brookbank JW, Roberts RM, Bazer FW. Establishment of pregnancy
in the pig: 11. Cellular remodelling of the porcine blastocyst during elongation on
day 12 of pregnancy. Biol Reprod l982;27:94 1-955.
Perry JS, Heap RB, Burton RD, Gadsby JE. Endocrinology of the blastocyst and
its role in the establishment of pregnancy. J Reprod Fert Suppl 1976;25:85-104.
Perry JS, Rowlands IW. Early pregnancy in the pig. J Reprod Fertil 1962;4: 175-
188.
Wray W, Boulikas T, Wray V. Hancock R. Silver staining of proteins in polyacryl-
amide gels. Anal Biochem I98 1;118:197-203.
Brigstock DR, Kim GY, Steffen CL, Liu A, Vegunta RK, Ismail NH. High molecular
weight forms of epidermal growth factor in pig uterine secretions. J Reprod Fertil
1996;lO8:3 13-320.
Kim GY, Besner GE, Steffen CL, McCarthy DW, Downing MT, Luquette MH,
Abad MS, Brigstock DR. Purification of heparin-binding EGF-like growth factor
from pig uterine luminal flushings and its production by endometrial tissues. Biol
Reprod l995;52:56 1-571.
Brigstock DR, Kim GY. Steffen CL. Pig uterine fluid contains the developmentally-
regulated neurotrophic factor, pleiotrophin. J Endocrinol 1996;148: 103- 1 1 I.
Brigstock DR, Heap RB. Brown KD. Polypeptide growth factors in uterine tissues
and secretions. J Reprod Fert 1989;85:747-758.
Bork P. The modular architecture of a new family of growth regulators related to
connective tissue growth factor. FEBS Lett 1993;327:125-1 30.
O'Brien TP, Yang GP, Sanders L, Lau LF. Expression of cyr61, a growth factor-
inducible immediate early gene. Mol Cell Biol 1990;10:3569-3577.
Jay P, Bege-Lefranc JL, Marsollier C. Mejean C, Taviaux S. Berta P. The human
growth factor-inducible immediate early gene, CYR61, maps to chromosome Ip.
Oncogene 1997;14:1753- 1757.
Simmons DL, Levy DB, Yannoni Y, Erickson RL. Identification of a phorbol ester-
repressive v-src-inducible gene. Proc Natl Acad Sci USA 1989;86: 1 178- 1 182.
Joliot V. Martinerie C, Dambrine G, Plassiart G, Brisac M, Crochet J, Perbal B.
Proviral rearrangements and overexpression of a new cellular gene (nov) in myelo-
blastosis-associated virus type 1-induced nephroblastomas. Mol Cell Biol 1992;12:
10-21.
Martinerie C, Huff V, Joubert I, Badzioch M, Saunders G, Strong L, Perbal B. Struc-
414 Brigstock
tural analysis of the human nov proto-oncogene and expression in Wilms tumors.
Oncogene 1994;9:2279-2732.
Ying Z. Ling ML. Isolation and characterization of xnov, a Xenopus laevis ortholog
of the chicken nov gene. Gene 1997;171:243-248.
Shing Y, Folkman J, Butterfield C, Murray J, Klagsbrun M. Heparin affinity: purifi-
cation of a tumor-derived capillary endothelial cell growth factor. Science 1984;233:
1296-1299.
Besner GE, Higashiyama S, Klagsbrun M. Isolation and characterization of a macro-
phage-derived heparin-binding growth factor. Cell Regu 1990;1:811-8 19.
Lobb RR, Harper JW, Fett JW. Purification of heparin-binding growth factors. Anal
Biochem 1986;154:1-4.
Rosenthall RA, Megyesi JF, Henzel WJ, Ferrara N, Folkman J. Conditioned medium
from mouse sarcoma 180 cells contains vascular endothelial growth factor. Growth
Factors 1990;4:53-59.
Cook PW, Mattox PA, Keeble WW, Pittelkow MR, Plowman GD, Shoyab M, Adel-
man JP, Shipley GD. A heparin sulfate-regulated human keratinocyte autocrine fac-
tor is similar or identical to amphiregulin. Mol Cell Biol 1991;11:2547-2557.
Shoyab M, Plowman GD. Purification of amphiregulin from serum-free conditioned
medium of TPA-treated cell lines. Meth Enzymol 1991 ;198:213-221.
Klagsbrun M. The fibroblast growth factor family: Structural and biological proper-
ties. Prog Growth Factor Res 1989;1:207-235.
Higashiyama S, Lau K, Besner G, Abraham JA, Klagsbrun M. Structure of heparin-
binding EGF-like growth factor: multiple forms, primary structure and glycosylation
of the mature protein. J Biol Chem 1992;267:6205-6212.
Milner PG, Li Y-S, Hoffman RM, Kodner CM, Siege1 NR, Deuel TF. A novel 17
kDa heparin-binding growth factor (HBGF-8) in bovine uterus: Purification and N-
terminal amino acid sequence. Biochem Biophys Res Commun 1989;165:1096-
1 103.
Caday CG, Klagsbrun M, Fanning PJ, Mirzabegian A, Finkelstein SP. Fibroblast
growth factor (FGF) levels in the developing brain. Dev Brain Res 1990;52:241-
246.
Weich HA, Iberg N, Klagsbrun M, Folkman J. Expression of acidic and basic fibro-
blast growth factors in human and bovine vascular smooth muscle cells. Growth
Factors 1990;2:3 13-320.
Slalom Chromatography
A New Hydrodynamic-Based
Chromatographic Mode Applicable to
Size-Dependent Separation and
Physicochemical Analysis of Large
DNA Molecules
I. INTRODUCTION
Chromatography is one of the best research tools for the biosciences. It makes
it possible to analyze biomolecules and to obtain the most valuable and important
information on them, such as their chemical, physicochemical, and biochemical
properties. At the same time, it affords complete separation of components.
Therefore, the more sophisticated the chromatographic mode, the higher the qual-
ity of the information obtained. For example, affinity chromatography reveals
the nature of the active site of enzymes; gel permeation chromatography discloses
the size of biomolecules. Moreover, development of high-performance liquid
chromatography (HPLC) has greatly enhanced the utility of chromatography by
improving speed, sensitivity, reproducibility, and accuracy.
Use of chromatography as an intelligent tool has not been adequately ap-
preciated in the field of nucleic acid research because of several inherent difficul-
ties [I]. Though size-dependent separation of large polynucleotides such as DNA
fragments must be the first step of not only the majority of basic analyses but also
416 Hirabayashi and Kasai
a variety of applications, the chromatographic method has rarely been applied for
such a purpose, in contrast to the protein research field where gel permeation
chromatography has always been indispensable. This is mainly because gel per-
meation chromatography is extremely inefficient for the separation of DNA frag-
ments in comparison with agarose gel electrophoresis, which has been much more
popular and efficient, as described below.
First, in gel permeation chromatography, separation of component mole-
cules must take place within a relatively small volume, i.e., the elution volume
of a salt subtracted by the flow-through volume. This corresponds to the volume
of the liquid retained in the stationary phase, in other words, the volume of the
liquid retained in the pores of the packing particles. It is difficult to expect high
resolution of many components in such a small volume. Second. target macro-
molecules must diffuse into the pores of packing particles and an equilibrium
state must be reached between the moving phase and the stationary phase. The
time required to reach an equilibrium state becomes considerably long for large
molecules due to their small diffusion constant, and this also reduces the resolu-
tion. Third, most DNA fragments are usually too large to be applied to conven-
tional gel permeation media. Even a small DNA fragment has a Stokes radius
considerably larger than that of proteins of similar molecular weight. Fourth, the
possible risk of cleavage of long DNA molecules due to shear force has also
made researchers reluctant to use chromatographic separation. However, in spite
of these difficulties, to make full use of chromatography in the field of nucleic
acids would seem to be meaningful. Chromatography is often complementary to
electrophoresis in terms of both preparative and analytical purposes, and some-
times it can provide unique results. In the field of nucleic acid research, analytical
scale chromatography serves both analytical and preparative purposes because if
only a trace amount of a DNA or RNA fragment is separated it can be easily multi-
plied by polymerase chain reaction (PCR) and recombinant DNA techniques.
We recently discovered a new chromatographic mode that enables the sepa-
ration of DNA fragments according to their size [2,3]. This method, firstly discov-
ered by chance, requires only an ordinary HPLC system and a commercially
available column packed with small, rigid, spherical beads, such as those used
for high-performance gel permeation. Strikingly, the order of elution was found
to be opposite to that expected for gel permeation chromatography, i.e., larger
fragments were eluted later than smaller ones. It is not surprising that longer
DNA fragments would be retarded more if they interacted with the column pack-
ing. The amazing thing, though, is that no evidence could be found of interaction
between them. Moreover, separation patterns significantly depended on the flow
rate and the size of the packing particles, but not on their pore size or chemical
nature. DNA fragments were retarded without any attractive force provided by
the column packings. Therefore. it is a quite unusual phenomenon and cannot
Slalom Chromatography 417
discuss in detail various unusual phenomena that led us to the above hypothetical
mechanism.
Kawasaki, Japan) were used. Their exclusion limits (for protein, M, 5 X lo5)
were the same, but the particle sizes were different. One column was packed
with 5-pm particles (Fig. 2a, Asahipak GFT-5 10) and the other with 9-pm parti-
cles (Fig. 2b, Asahipak GS-5 10). Both columns separated eight fragments in the
order of smaller to larger. This order was completely opposite to that expected
for gel permeation chromatography. Moreover, though the pore sizes were the
same, the column packed with 5-pm particles was superior for the separation of
smaller fragments (less then 20 kbp), whereas larger fragments (more than 20
kbp) were better separated by the column packed with 9-pm particles. It is evident
that separation occurred by a mechanism other than the gel permeation mode
because the order of elution was opposite and the size range depended not on
the pore size but on the particle size.
Figure 3 shows the separation pattern of HindIII-digested h phage DNA
(hlHindII1) on columns packed with 5-pm and 9-pm particles. Again, the 5-pm
packing gave a better resolution for fragments of less than 10 kbp. Such experi-
ments were carried out more systematically using four columns (average particle
diameters of 5.0,9.0, 13.1, and 19.1 pm, Asahipak GS-3 10 series), and the results
are summarized in Fig. 4 [4]. The relative retention time, defined as the ratio of
Elution Volume ( ml
Figure 4 Dependence of relative retention times of DNA fragments on their size. Pack-
ings of different particle size (5, 9, 13, and 19 ~ m were
) used, and different flow rates
were applied. Relative retention time was plotted as a function of DNA size (kbp). Flow
rates were 0.3 (a),0.6 (A), and 1.2 mllmin (m).
17, 15-30,23-40, and 35-50 kbp for the 5-, 9-, 13-, and 19-pm packings, respec-
tively.
rates DNA fragments according to their physical length and not to their molecular
weight.
a diameter of -2 pm in its most contracted state. The random coil is very flexible,
and its shape changes without a break between the contracted and slightly ex-
tended forms by an external force provided by Brownian motion of water mole-
cules. The speed of the intramolecular movement (expansion and contraction)
can be as fast as - 15 pm/s, though the migration velocity of the whole molecule
(center of gravity) is only 1 ym/s [7]. However, in the presence of laminar flow,
the DNA molecule is stretched. As the flow rate increases, its shape changes
gradually from an ellipsoid to a solenoid one, then to a thick filament one, and
finally to a thin filament one.
DNA molecules are too large to permeate into the pores of packing particles
and can only pass through the narrow channels between particles. Since these
channels are extremely tortuous, when the moving phase is forced to move, DNA
fragments have to turn very frequently. It will be more difficult for longer DNA
strands to turn quickly, and this will result in retardation of longer DNA strands.
A simple calculation shows that DNA fragments turn about 36,000 times if ap-
plied to a column of 300-mm length packed with 10-ym packing particles. Under
these conditions, a 23-kbp fragment turns as frequently as 70 times per second,
when a flow rate of 0.6 ml/min is applied. If we use smaller packing particles,
the channels become narrower and more tortuous; and also, the velocity gradient
of laminar flow becomes steeper, which makes shear force stronger. At a flow
rate of 1.2 ml/min, in a column of 5-ym particles, the fragment has to turn 72,000
times at a frequency of 280 times per second. On each turn, DNA strands would
be exposed to a significant frictional force against the solvent, and this force
would increase with the increase in DNA length, so that larger DNA molecules
are retarded.
As to the effect of flow rate, at a flow rate of close to zero all DNA frag-
ments are in a random coil state; as none of them are retarded, they are eluted
together in the flow-through fraction. Application of a faster flow rate has at least
dual effects. First, it increases the frequency of turn. Second, it increases the
velocity gradient of the laminar flow. This generates stronger shear force and
consequently raises average end-to-end distance of DNA strands, and resulting
in stronger frictional force between the DNA strands and the solvent (Fig. 5).
These considerations may explain why the use of smaller packing and a higher
flow rate is advantageous for the separation of smaller molecules.
This new chromatographic mode is attributed to the combination of several
hydrodynamic phenomena, i.e., stretch by laminar flow of DNA fragments that
otherwise assume a random coil state because of entropic force; frequent turning
of extremely long molecules along the curved surface of packing particles; and
an extremely complicated flow pattern due to the channels made between packing
particles, which may cause continuous perturbation of the three-dimensional
shape of DNA. Although the explanation presented here is undoubtedly oversim-
plified, it should be a useful model in elucidating the precise mechanism of slalom
chromatography.
Slalom Chromatography
Narrow
Flow Rate
l==s Wide
Figure 5 An explanation of the effect of the flow rate and particle size. For simplicity,
the channel between closely packed particles is considered as a capillary. As the flow rate
is increased, velocity gradient of the moving phase is generated due to laminar Row. In
a narrow capillary, the velocity gradient is steep, and then a DNA molecule is subjected
to a strong shear force and stretched to a greater extent. As a result, the DNA molecule is
retarded greatly due to the slalom effect. On the contrary, in a wide capillary, no significant
velocity gradient is generated especially in the central part of the cross-section. Therefore,
the DNA molecule is not subjected to a strong shear force, stays as a more compact form,
and goes through the capillary without significant retardation.
The utility of this new procedure is not limited to size analysis. This method will
undoubtedly provide us valuable information on DNA molecules almost equiva-
lent to those gel filtration has provided on the properties of proteins. Moreover,
426 Hirabayashi and Kasai
this hydrodynamics-based method has potential to analyze not only the sizes
of DNA molecules but also physicochemical properties, such as conformation,
topology, and rigidity. Preliminary experiments showed that it can distinguish
the difference in the topology of circular DNA molecules because different flow
rate dependencies were observed for supercoiled, relaxed, and single-stranded
DNAs. These results might be attributed to their rigidity or special three-dimen-
sional structure. Therefore, it will be possible to separate and analyze DNA mole-
cules according to their physicochemical properties. If the efficiency of slalom
chromatography becomes much higher, isolation of topoisomers of supercoiled
DNAs according to the extent of coiling might become possible.
67 5
REFERENCES
I. INTRODUCTION
This chapter was adapted from X. T. Wang et a]., Prep Biochem 1994: 24: 237-250. Copyright
0 1994 by Marcel Dekker, Inc.
432 Mateescu et al.
function as copper carrier. There is a recently growing interest for genetic dis-
eases characterized by a C P deficiency or by a defective loading of copper into
CP. Wilson's, Menkes', Parkinson's, Alzheimer's, and, as recently found, hemo-
siderosis are diseases related to C P deficiency.
Despite the abundant literature linking C P to several pathological condi-
tions, little research has been done on its possible therapeutic applications, proba-
bly because of its high susceptibility to proteolysis and instability, even during the
purification steps. In fact, structural aspects of CP, mainly related to the molecular
characteristics and the copper content, have been controversy (mostly due from
its high susceptibility at proteolysis). Also controversial was its complex physio-
logical role (antioxidantlprooxidant) related to the molecular integrity of the mol-
ecule, as recently reported [ l l l.
The "blue copper" center of CP has a characteristic absorption band at
610 nm and is electronic paramagnetic resonance (EPR)-detectable. It is now
accepted that C P contains six copper atoms per molecule and that CP structure
consists of six domains [12]-surprisingly, in a configuration closed to that of
clotting Factor VIII. Three copper atoms are aggregated in a cluster which is the
blue copper center of ceruloplasrnin. Two others form a diamagnetic pair. The
last one is paramagnetic (EPR-detectable). An absorbancy ratio Ab10nmlA280nm =
0.040 was considered in the literature as characteristic for the homogeneous stan-
dard pure enzyme.
Previously reported was a new single-step chromatographic method for the
fast ceruloplasmin purification [13], starting directly from plasma, which leads
to a purified, electrophoretically homogeneous CP. The procedure is based on
the highly selective retention of C P on the aminoethyl (AE)-agarose, a new chro-
matographic material (not commercially available), realized in our laboratories
113- 151. This single-step purification procedure described by Calabrese et al.
[14] is very fast, requiring only plasma dilution (about 20 times), followed by
AE-agarose chromatography and a final concentration of the purified CP. Despite
the speed at the laboratory scale, for scaling-up purposes, handling of large vol-
umes (dozens of liters) of diluted plasma is somewhat cumbersome. Furthermore,
when large plasma volumes are used, some clotting phenomena can occur during
the chromatographic step, producing a decrease in flow rate, difficulties in regen-
eration, and partial loss of the chromatographic material. At the same time, han-
dling of large protein volumes for the final concentration is also cumbersome.
These limitations can be completely overcome by the recently reported purifica-
tion procedure [16], here described.
Bovine serum amine oxidase (SAO), also known as benzylamine oxidase
belongs to the semicarbazide-sensitive amine oxidase (SSAO), is a copper en-
zyme (EC 1.4.3.6) catalyzing the oxidative deamination of various biogenic pri-
mary amines, with release of aldehydes, ammonia, and hydrogen peroxide. Con-
sidering the toxicity of aldehyde and H 2 0 2 ,SAO was suggested as an antitumoral
agent [I71 with an important cytotoxicity-inducing activity [18]. Recent data
Chromatographic Separation of CP and SAO 433
showed that SAO has a multifunctional role as regulator of polyamine level [I 91,
as cardioprotective agent [20,21], and as modulator of ionic channel [22]. It was
also shown that SAO substrate specificity is not limited only to small molecular
biogenic amines such as spermine and spermidine, but is extended to polylysine
and some peptides or proteins [23]. This suggested the hypothesis that SAO can
be involved in the process of protein posttranslational modification [23]. The
enzyme was first isolated in crystalline form by Yamada and Yasunobu [24].
Presently, several purification procedures are available. An affinity separation of
SAO on AH-Sepharose was described by Svenson and Hynning [25] and a two-
step procedure (ammonium sulfate and affinity chromatography on AH-Sepha-
rose followed by elution with aniline SAO inhibitor) was reported by Vianello
et al. 1261. The method described by Mondovi et al. [27] (ammonium sulfate
precipitation followed by chromatography on AH-Sepharose and Con A-Sepha-
rose) leads to high SAO-specific activities (0.33 EUImg). Since the AH-Sepha-
rose is no longer commercially available, it has been substituted with Q-Sepha-
rose 1281. The procedure usually leads to a purified SAO [27] with a slightly
lower specific activity (0.28 EUImg). In some chromatographic runs on Q-Sepha-
rose, the peaks of ceruloplasmin and bovine serum amine oxidase were partly
overlapped, thus lowering the purification yield and the specific SAO activity.
This can be satisfactorily improved by elimination of C P from the plasma protein
preparation before passing it through a Q-Sepharose column, leading to higher
specific activities and yields.
SAO and ceruloplasmin exhibit several common features. Both are serum
copper proteins of a relative high molecular mass: 180,000 Da (two monomers
of 90 kDa) for SAO and 132,000 Da for CP. Both enzymes are involved in
oxidative processes and both are glycoproteins (6-12% carbohydrate). Both pro-
teins have antioxidant properties and act as cardioprotective [20,29] and neuro-
modulatory agents [9,22]. Furthermore, the two proteins exhibit, to a certain
extent, a similar behavior toward some chromatographic materials (e.g.,
Q-Sepharose and AH-Sepharose), often leading to interference during the purifi-
cation procedures. A joint purification procedure leading to higher purity parame-
ters or yields should therefore be of interest to laboratories working in the field
of copper proteins.
In this study, a joint chromatographic purification procedure of C P and
SAO is described and the results are compared with independent CP [14] and
SAO [27] purification procedures.
II. EXPERIMENTAL
A. Chromatographic Material
Aminoethyl agarose is a chromatographic material (not commercially available)
obtained in laboratory by the treatment of agarose beads (i.e., Superose 6B, Phar-
434 Mateescu et al.
1. Collection of Plasma
A volume of 10 L of bovine blood was collected at the slaughterhouse, mixed
with 1 L of 2.5% sodium citrate solution and centrifuged at 3000g for 20 rnin,
retaining the plasma. The crude CP-specific activity is usually about 8.14 X
EUImg and SAO specific activity of about 2.4 X to 7.0 X EUImg.
+ Bo- blood
(centrifuged at 3000g)
t
Bovine plasma (.) Dilntion
(about 20 limes)
1 I.
Ib) Precipitation
with (NH+)2 SO+ AE-Agar~se
I Con A-Sephamse
(PH 7-21
1.A. = 0.33
SAO SIO
KDa Standard
D. SAO Purification
The SAO separation method according to Mondovi et al. [27], slightly modified
[B. Mondovi and 0 . Befani, unpublished data, 19921 as is now currently applied
(without the AE-agarose column), was carried out in parallel with the joint puri-
fication here described (Fig. I), starting from the same bovine plasma prepared
as described above.
Electrophoresis
The degree of homogeneity as well as the molecular integrity and characteristics
of purified CP and SAO were evaluated by SDS-PAGE fast electrophoresis (Phast
System, Pharmacia-LKB), in the presence of 5% P-mercaptoethanol (Fig. 2).
"efined as the ratio: specific activity of purified enzymelspecific activity in the starting material.
hLower starting volume than for the combined method, since the required 1 :20 dilution leads to
4 L (difficult to handle in the chromatographic run).
Source: From Ref. 16.
step" method [14]. However, the recovery appears higher for the initial single-
step procedure (50.6 mg CP yielded from 200 mL plasma versus 123 mg CP
from 2000 mL plasma). Considering the amount of CP in plasma (200-300 mg/
L), the CP recovery by the single-step procedure [14] is very high (250 mg/L).
On the other hand, the modified method, despite a lower recovery (60-65 mg
CPIL), seems more advantageous because dealing with smaller volumes yields
a higher CP amount (120-125 mg) per separation run.
The characteristics of CP purified by the modified method (including am-
monium sulfate precipitation) are better than those obtained by the single-step
procedure. For instance, the specific activity is three times higher (0.70 versus
0.23 EU/mg) while the purity expressed by the spectral parameters (A610/A280)
are improved by more than 25% (higher than 0.057 versus 0.045). The improve-
ment in CP purification can be related to the partial elimination of contaminants
as well as of clottable proteins (in fact, no clotting phenomena were observed
during the AE-agarose chromatographic run). Minimizing the sample story, the
risk of protein degradation is limited. In fact we suppose, following a reexamina-
tion of CP spectral properties (EPR [14]), that CP purified by chromatography
on AE-agarose is closer to its real native structure than commercial CP obtained
by other methods.
This method can also allow the CP immobilization directly on this specific
chromatographic support during its purification [33]. The conjugation of CP with
440 Mateescu et al.
Current Combined
Purification characteristics method method
Starting material volume (mL plasma)
Total SAO activity (EU) of starting plasma
Total protein amount (mg) in plasma
Specific SAO activity in plasma (EUImg protein)
Specific activity (EUIrng) of purified SAO
Purification factora
Purity (A481dA280)
Total purified SAO protein (mg)
Total activity (enzyme units) of purified SAO
Yield (recovered EU) (%)
"efined as the ratio: specific activity of purified enzymelspecific activity in the starting material.
Source: From Ref. 16.
Chromatographic Separation of CP and SAO 441
[27], the elution profile (Fig. 3a) shows a slight overlap between the CP and SAO
peaks. As a result of the additional AE-agarose step provided by the combined
CP-SAO separation procedure, the chromatographic interference by CP is
avoided (Fig. 3b). With a previous AE-agarose column. the SAO specific activity
after the first Q-Sepharose purification step was increased by about 35% (the
result of four different separation runs) when compared with the current method
(without the AE-agarose step).
As shown in Fig. 1, differently from the previous separation procedure, the
Con A-Sepharose step was carried out after the second Q-Sepharose column. The
advantage of this inversion is that the use of a second Q-Sepharose column leads
to a major increase in specific activity, with a simultaneous decrease in the vol-
ume to be handled in the last Con A-Sepharose step.
We are not considering the joint CP and SAO purification method presented
here as a new method for either CP or SAO purification. However, it is worth
mentioning that the mutually improving effect generated by associating the
two methods of the independent C P and SAO purification procedures into a joint
method for C P and SAO purification leads to better characteristics for each puri-
fied protein. The improvement is more important for the CP purification, as it
leads to several fold increases in different characteristics (specific activity and
the total activity units obtained by a separation run) and to a better molecular
integrity as reflected by the A,,,,/A2,, ratio. The increased yield of SAO of more
than 25% by the joint purification technique is also useful.
The current procedures for the SAO purification [27] require dialysis and
concentrations between each chromatographic step. In the modified method, dial-
ysis and concentration between the chromatographic columns can be eliminated
and replaced by simple dilution (1 : 1) of the enzyme solution with one volume
of the buffer used to equilibrate the subsequent column. This improvement allows
a faster purification run, leading to higher yields, increased specific activities,
and ameliorated spectral characteristics. For instance, with these modifications,
the yield is increased (100 to 115 mg SAO from 10 L of blood). The specific
enzymatic activity is 0.32 to 0.35 EU/mg and the absorbancy ratio A4xonn,/A280nm
= 0.013 (denoting a high quality of the enzyme). At the same time, the eluent
volumes were reduced. For example, the eluent volume of the first Q-Sepharose
column was reduced from 1800 mL to 600 mL (while maintaining the initial and
final gradient concentrations) and thus the method is faster and easier to apply.
The modified method is therefore time saving (only 3 days for the whole separa-
tion instead of 4-5 days for the current procedure) and leads to improved yields
and quality of SAO. Apart from the mutual improvement effects, better yields,
and molecular characteristics, the combined purification procedure may be of
interest for large-scale or industrial preparations, since the same inexpensive bo-
vine plasma source and preliminary precipitation as well as the AE-agarose steps
are common steps for both C P and SAO purification.
Mateescu et al.
ACKNOWLEDGMENTS
REFERENCES
Calabrese L, Mateescu MA, Riccio P-L. Carbonaro M, Natoli G, Mondovi B. Single step
chromatographic purification of plasma ceruloplasmin. Italian Patent 479 14A86 (1986).
Wang XT, Dumoulin MJ, Befani 0, Mondovi B, Mateescu MA. Joint chromato-
graphic purification of bovine serum ceruloplasmin and amineoxidase. Prep Bio-
chem 1994;24:237-250.
Mondovi B, Gerosa P, Cavaliere R. Studies on the effect of polyamines and their
products on Ehrlich ascites tumours. Agents Actions 1982; 1 :450-45 1.
Agostinelli E, Bates DA, Przybytkowski E, Mateescu MA, Mondovi B. Cytotoxicity
of polyamines in Chinese hamster ovarian (CHO) cells in the presence of serum
amine oxidase. Life Chemistry Reports 1991;Y:193-204.
Mondovi B, Riccio P, Agostinelli E. The biological functions of amine oxidases and
their reaction products: an overview. Adv Exp Med Biol 1988;250:147-161.
Mateescu MA, Dumoulin MJ, Wang XT, Nadeau R, Mondovi B. A new physiologi-
cal role of copper amine oxidases: cardioprotection against reactive oxygen interme-
diates. J Physiol Pharmacol 1997;48 Suppl 2: 110-121.
Mondovi B, Wang XT, Pietrangeli P, Wang R, Nadeau R, Mateescu MA. New as-
pects on the physiological role of copper amineoxidases. Curr Top Med Chem 1997;
2:3 1-43,
Wu L, Mateescu MA, Wang XT, Mondovi B. Wang R. Modulation of K ' channel
currents by serum amineoxidase in neurons. Biochem Biophys Res Commun 1996;
220:47--52.
Wang XT, Pietrangeli P, Mateescu MA, Mondovi B. Extended substrate specificity
of serum amine oxidase: possible involvement in protein posttranslational modifica-
tion. Biochem Biophys Res Commun 1996;223:91-97.
Yamada H, Yasunobu KT. Purification, crystallization and properties of plasma
monoamine oxidase. J Biol Chem l962;237: 151 1- 1516.
Svenson A. Hynning PA. Preparation of amine oxidase from bovine serum by affin-
ity chromatography on aminoheyxyl-Sepharose. Prep Biochem 1981;11:99-108.
Vianello F, Di Paolo ML, Zennaro L, Stevanato R, Rigo A. Isolation of amine oxi-
dase from bovine plasma by a two-step procedure. Protein Expression Puri 1992;3:
362-367.
Mondovi B, Turini P, Befani 0, Sabatini S. Purification of bovine plasma amine
oxidase. Meth Enzymol 1983;94:3 14-3 18.
Susan-Janes M, David MV, Wemmer D, Smith AJ, Kaur S, Maltby D, Burlingam
AL, Klinman JP. A new redox cofactor in eukaryotic enzymes: 6-hydroxydopa at
the active site of bovine serum amine oxidase. Science l990;248:98 1-987.
Atanasiu R, Gouin L, Mateescu MA, Cardinal R, Nadeau R. Class 111 antiarrhythmic
effects of ceruloplasmin on rat heart. Can J Physiol Pharmacol 1996;74:652-656.
Osaki S, Johnson DA, Frieden E. The possible significance of the ferrous oxidase activity
of ceruloplasmin in normal human serum. J Biol Chem 1966;241:2746-275 1.
Tabor CA, Tabor H, Rosenthal SM. Purification of amine oxidase from beef plasma.
J Biol Chem 1954;208:645-665.
Bradford M. A rapid and sensitive method for the quantitation of microgram of protein
utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248-254.
Mateescu MA, Fortier G, Neidhart S, Roger S. Selective immobilization of cerulo-
plasmin on the chromatographic material used for its purification. Chromatographia
1988;26:110-121.
Fast, Single-Step Affinity
Chromatography Purification
of Hemoglobin
Mircea-Alexandru Mateescu and Wilfrid Jacques
Universite du Quebec a Montreal, Quebec, Canada
I. INTRODUCTION
Hemoglobin (Hb), the main oxygen carrier, is a tetramer in which each monomer
( a , , a,, p,; and p,) contains a porphyrin prosthetic group. Initiated in 1937, the
long path to understanding Hb biochemistry led to high-resolution X-ray Hb anal-
ysis (Perutz, 1968), which was a landmark achievement for the formulation of
the "Perutz mechanism" based on the stereochemistry of the cooperative effects
in Hb oxygenation [I]. One of the most complex proteins, Hb is also the most
studied over the last 30 years. Since oxygen exchange is controlled by the revers-
ible steric configuration of the protein, Hb has been nominated as an "honorary
enzyme" (Voet and Voet [2]).
Hemoglobin is not a glycoprotein, but it can be easily glycosylated with
free glucose, by a nonenzymatic mechanism. About 6% of the erythrocytary Hb
is glycosylated in normal subjects. In diabetes the glucose level can be three
times higher [3,4], leading, with different rates, to glycated forms HbA,,, HbAlh,
and HbAI,. More knowledge of interactions of Hb with sugars is of interest since
the oxygenation-deoxygenation capacity of the glycated Hb is reduced by 20-
25%. Furthermore, the effects of the alteration of oxygen transport function are
enhanced by the relatively high half-life of Hb, up to 120 days. It was also
Part of this work was adapted from W. Jacques and M. A. Mateescu, Anal. Lett. 1993;26:875-886.
Copyright O 1993 by Marcel Dekker, Inc.
446 Mateescu and Jacques
reported [5] that, unexpectedly, the velocity of glycosylation of the amine termi-
nal group of the P subunit located on the allosteric center of Hb is three times
higher than the glycosylation rate of other amine groups on the Hb surface. Since
the mechanism is nonenzymatic, the kinetics is in disagreement with the statistical
distribution of amine groups susceptible to glycosylation.
On the basis of the peculiar kinetics of glycosylation, we have advanced
the hypothesis of a selective recognition center for sugars. Previously [6,7], we
have described specific interactions of Hb with some polysaccharides as CL-
amylose (CLA) and CL-agarose, while practically no interaction has been ob-
served with CL-dextran (Sephadex). All of these chromatographic materials are
obtained by the polysaccharide chain treatment with epichlorohydrin [8,9], lead-
ing to swellable three-dimensional polymeric structures. The fact that Hb is re-
tained by CL-amylose (main chains based on glucose units linked by a-1,4-glyco-
sidic bonds) and not by Sephadex (main chains based on glucose units linked by
a-1,6-glycosidic bonds) suggests that Hb can selectively recognize and differenti-
ate between the two types of linkages characteristic for each polymer. At the
same time it was shown that Hb can recognize the CL-agarose (main chains
based on galactose and 3,6-anhydro-I--galactose), indicating that both the type
of linkage and the saccharidic units forming the carbohydrate polymer are impor-
tant [7]. Furthermore, the study described specific interactions of Hb with mono-
and disaccharides. The competitive elution of Hb retained by affinity on CL-
arnylose and on CL-agarose (Sepharose CL-4B) chromatographic materials, with
different sugars as competitive "affinants," showed that glucose, fructose, and
cellobiose are poor eluants, whereas lactose is a good competitive eluant, sug-
gesting that it better interacts with Hb and, more important, that the interaction
is selective [7].
Another related study reported Hb separation by specific affinity chroma-
tography retention on CL-arnylose, with a capacity of 4.5-10 mg Hblml gel bed,
followed by desorption with deforming eluants, as NaCl [6]. The CL-amylose
material was previously used in exclusion chromatography [9] and in affinity
chromatography for a-amylase separation [ 101.
This new aspect on the Hb properites can offer a better understanding of
Hb-carbohydrate and Hb-glycoprotein interactions and, at the same time, a pos-
sibiltiy to purify Hb directly from erythrocytary lysate, by a single-step chromato-
graphic procedure on crosslinked amylose columns [6]. These Hb separation ex-
periments were carried out using commercial bovine Hb (Sigma Chemical Co.)
solutions.
Hemoglobin and its derivatives are currently isolated by various ion ex-
change chromatographic techniques. However, several difficulties have been re-
ported, mainly related to the Hb contamination with various compounds, and
often a rechromatography is required. Thus, alternative procedures have been
elaborated based on the Hb elution from the starch gel after the electrophoretic
Purification of Hemoglobin 447
II. EXPERIMENTAL
A. Materials and Reagents
Agarose-based chromatographic materials (Sepharose C L 4 B , Sepharose CL-6B,
Superose 12 Prep. Grade, Sephadex G-25, and Sephadex G-100) were from Phar-
macia LKB (Uppsala, Sweden). Commercial bovine hemoglobin (lyophilizedpow-
der), bovine serum albumin (BSA), and Coomassie brillant blue R-250 were from
Sigma Chem. Co (St. Louis, Missouri). CL-amylose (CLA) is a semisynthetic po-
lyglucose chromatographic material realized by amylose crosslinking with epichl-
orohydrin [6,7,9]. As the Sephadex gel, CLA was here used almost for comparison
in terms of Hb interactions with agarose-based materials. Human blood was ob-
tained from the H6pital Charles-Lemoyne, Longueuil (Qukbec), Canada.
and the suspension was centrifuged at 12,000g for 20 min at 4"C, retaining the
supernatant.
0 2 4 6 8 10 12 14 16 18 20 22
Elution Volume (mL)
Figure 1 Typical chromatographic (A) and electrophoretic (B) separation profiles of a
mixture of bovine serum albumin (BSA) (1 mglml) and commercial hemoglobin (Hb)(l
mglml) on a Superose 12 column (4 ml gel bed). The absorbances A,,, (-o-) and A280n,
(-) yielded for the first peak a ratio value A,,,,IA,s,,,,,,, = 0.21 and for the retained Hb
A40an,lA280,,= 5.2. The sample volume was 2 ml. SDS-PAGE was run for standard molec-
ular weight (Jane l), Hb and BSA mixture before separation (lane 2), nonretained (ascribed
to BSA) fraction (lane 3), and retained Hb eluted from the CLA column (lane 4).
Purification of Hemoglobin 451
solution. In the case of Hb associated with other proteins, e.g., with BSA in equal
concentrations in aqueous samples, a selective retention of Hb on the Superose
12 column was observed, while a total elution of the nonretained BSA was found
in the first peak (Figure 1).
No Hb retention was observed in identical chromatographic conditions on
Sephadex G-25 or Sephadex G- 100 (crosslinked dextran based on a-1,6-glucosi-
dic bonds). These data clearly indicated that Hb can specifically recognize and
be retained at the level of galactose and 3,6-anhydro-L-galactose sequences of
the Superose 12 chromatographic material. The Hb was eluted in a single step,
with a 0.9% NaCl solution (Figure 1A). It is worth noting that, being sensitive
to ionic strength, Hb can be desorbed at even lower NaCl concentrations. The
yield of Hb recovery was higher than 85%.
The BSA was chosen as a model contaminant because this protein, like
Hb, is not a glycoprotein, and presents a molecular weight of 68 kDa, close to
that of Hb (64 m a ) . The presence of small amounts of Hb in the first peak of
BSA or other contaminant proteins, even if the column is not overloaded, can
be ascribed to the presence of some modified forms in the commercial Hb (be-
yond the methemoglobin and the dimers, which can be associated with Hb in
commercial preparations). In fact, the presence of dimers (32 kDa) was confirmed
as a minor peak in the commercial Hb, by exclusion chromatoraphy of Sephadex
G-100, eluted after the major tetrameric Hb peak (64 m a ) .
The SDS-PAGE runs (Fig. 1B) clearly indicated the specific retention of
Hb on Superose 12, whereas BSA is not retained at all. In fact, only Hb recovered
from the column was visualized on lane 4 as a single band corresponding to the
Hb monomer (16 kDa) and another minor one for the dimer (32 kDa).
On the basis of this specific Hb retention onto the Superose 12 column,
the proposed affinity chromatography method allows the single-step purification
of Hb, directly from erythrocytary lysate. A selective chromatographic retention
of erythrocytary Hb (Figure 2) from a crude heterogeneous extract obtained by
red cell hemolysis in water (1 : 11 vlv), was found. As in the case of commercial
Hb, recovery of the erythrocytary Hb can be easily realized by direct elution with
physiological 0.9% NaCl solution (Figure 2A).
The absorbancy ratio A406nm/A280nm was considered (beyond SDS-PAGE) a
criterion to assess the purity of Hb eluted from the column. The purification
parameters are presented in Table 2. For all of the purified Hb recovered peaks,
the A,MnmIA2,0,m ratio values were 4.3 (as for the standard commercial Hb) or
even higher, up to 5.3, indicating an improvement of the degree of purity of
the commercial Hb. The first nonretained peaks (contaminants and eventually
overloaded Hb) present lower ratio values, due to various contaminants absorbing
at 280 nm. The retention capacity and the elution profiles were obtained with
small columns (1 X 5 cm), but the scaling-up process with bigger columns (50-
to 100-ml gel bed or more) allows the large-scale purification of Hb from blood
Mateescu and Jacques
Hb
dime r
24000
Hb
'8'0° monomer
B
0 6 12 18 24 30 36 42 48 54 60
Elution Volume (mL)
Figure 2 Typical chromatographic (A) and electrophoretic (B) profiles of the erythrocy-
tary separation on a Superose 12 column (3-ml gel bed). The absorbances (-0.) and
A,,,, (-) yielded for the erythrocyte lysate a ratio value Am,,lnlA2xonm= 3.7 and for the
retained Hb A41KII,,,IA2X0,,,II = 4.5. SDS-PAGE was run for standard molecular weight (lane
I), heterogeneous erythrocytary hemolysate before separation (lane 2). nonretained frac-
tion (lane 3), and rctained Hb eluted from the Superose 12 column (lane 4). The separation
conditions are presented In the "Materials and Methods" section.
"For the commercial Hb (Sigma Chemical Co.) before the column, the A,IA,,,, ratio was 4.3. The
data represent a mean of four experimental values.
Purification of Hemoglobin 453
sources. The Superose 12 columns were repeatedly used for tens of cycles without
significant alteration of the retention capacity. A condition for the column to
work is that it be thoroughly washed first with 0. I M NaOH and then with HzO,
from one cycle to another (until conductivity in washing eluate decreases at less
than 15-30 mmho). However, after more than 50 cycles, and under certain condi-
tions, the mechanical properties could be partially affected by alkaline (0.1 M
NaOH) washings between the affinity cycles.
The erythrocytary Hb recovered from CLA-40 column (Figure 2B) was
electrophoretically (SDS-PAGE) homogeneous, obtaining a strong band charac-
teristic for Hb monomer (16 kDa) and another minor band for the Hb dimer (32
kDa) only, as for the purified commercial Hb (Figure 1 B, lanes 2 and 4). Dissocia-
tion into monomers is due to the presence of SDS in the electrophoretical gel
and has been reported by other groups [16].
The molecular integrity of purified Hb (64 kDa) was confirmed by exclu-
sion chromatography on Sephadex G-100, where a unique peak was observed at
the same elution volume (V,) generating a K, = 0.17, close to that found for
BSA (68 kDa).
The specific affinity interaction of bovine Hb with CL-agarose (Superose
12) and with CL-amylose was comparable with that found for human Hb; the
elution profiles (data not shown) were similar to those of bovine Hb. Furthermore,
the affinity retention on Superose 12 and on CL-amylose seems to be specific
for Hb and also for some related proteins. Globin, the heme-free hemoglobin
monomer, was shown to interact moderately with the CL-amylose, but to a lesser
extent than Hb [14], whereas heme is not retained at all. The similarities between
the affinity behavior of human and bovine Hb on agarose chromatographic mate-
rials, here mentioned, as well as previous data [14] showing affinity interaction
of horse heart myoglobin with the CL-agarose, suggest that the interaction with
these polysaccharides can be a general property of hemoglobins and related pro-
teins of various species. The selective sugar recognition of hemoglobins and re-
lated proteins appears to be irrespective of the origin (human or bovine hemoglo-
bin and horse heart myoglobin). The retention capacities of CL-amylose for
myoglobin (18.8 kDa) was of 20 mglg dry support [14], a value of about 25%
of the retention capacity of 86 mglg dry support for the tetrameric bovine Hb
(64 kDa). Since myoglobin's structure is very close to the Hb monomers (16
kDa), it is possible to suppose a unique type of sugar recognition center on the
Hb molecule. This supposition fits with the data of Haney and Bunn [5]. on the
higher rate and yield of nonenzymatic glycosylation of one P subunit, leading to
an excess of HbAl, glycosylated form when compared to the forms HbA,,, and
HbAlb.
The advantage of the method described here is that no other plasma or
erythrocytary proteins interact, under the experimental conditions presented,
with these CL-amylose and CL-agarose chromatographic materials that are not
454 Mateescu and Jacques
ACKNOWLEDGMENTS
REFERENCES