<<

Louisiana State University LSU Digital Commons

LSU Historical Dissertations and Theses Graduate School

1999 Development of Immunochemical Methods for Gossypol Analysis. Xi Wang Louisiana State University and Agricultural & Mechanical College

Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_disstheses

Recommended Citation Wang, Xi, "Development of Immunochemical Methods for Gossypol Analysis." (1999). LSU Historical Dissertations and Theses. 7021. https://digitalcommons.lsu.edu/gradschool_disstheses/7021

This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Historical Dissertations and Theses by an authorized administrator of LSU Digital Commons. For more information, please contact [email protected]. INFORMATION TO USERS

This manuscript has been reproduced from the microfilm master. UMI films the text directly from the original or copy submitted. Thus, some thesis and dissertation copies are in typewriter face, while others may be from any type of computer printer.

The quality of this reproduction is dependent upon the quality of the copy submitted. Broken or indistinct print, colored or poor quality illustrations and photographs, print bleedthrough, substandard margins, and improper alignment can adversely affect reproduction.

In the unlikely event that the author did not send UMI a complete manuscript and there are missing pages, these will be noted. Also, if unauthorized copyright material had to be removed, a note will indicate the deletion.

Oversize materials (e.g., maps, drawings, charts) are reproduced by sectioning the original, beginning at the upper left-hand comer and continuing from left to right in equal sections with small overlaps. Each original is also photographed in one exposure and is included in reduced form at the back of the book.

Photographs included in the original manuscript have been reproduced xerographically in this copy. Higher quality 6” x 9” black and white photographic prints are available for any photographs or illustrations appearing in this copy for an additional charge. Contact UMI directly to order.

Bell & Howell Information and Learning 300 North Zeeb Road, Ann Arbor, Ml 48106-1346 USA 800-521-0600

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. DEVELOPMENT OF IMMUNOCHEMICAL METHODS FOR GOSSYPOL ANALYSIS

A Dissertation

Submitted to the Graduate Faculty of the Louisiana State University and Agricultural and Mechanical College in partial fulfillment of the requirements for the degree of Doctor of Philosophy

in

The Department of Food Science

by Xi Wang B.S., Wuxi Institute of Light Industry, , 1991 M.S., Wuxi Institute of Light Industry, China, 1994 August, 1999

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. UMI Number: 9945748

UMI Microform 9945748 Copyright 1999, by UMI Company. All rights reserved.

This microform edition is protected against unauthorized copying under Title 17, United States Code.

UMI 300 North Zeeb Road Ann Arbor, MI 48103

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ACKNOWLEDGMENTS

The author wishes to express her sincere appreciation to her present advisor, Dr.

Leslie C. Plhak, Department of Food Science, Louisiana State University, for her

guidance, assistance, and inspiration during this study.

Sincere thanks are given to Dr. Peter J. Wan (USDA, New Orleans, LA), Dr.

Douglas L. Park (Professor and Head of Department of Food Science, LSU), Dr.

Witoon Prinyawiwatkul (Department of Food Science, LSU) and Dr. William

Wischusen (Department of Biological Science) for serving on the dissertation

committee and for their valuable advice.

Grateful appreciation is expressed to Dr. Michael K. Dowd (USDA, New

Orleans, LA), Dr. Millard C. Calhoun (Texas A&M, College Station), Dr. Maren

Hegsted (Department of Human Ecology, LSU), and Laurie A. Henderson (Animal

Care Facility, LSU) for their valuable assistance and suggestions. Special thanks are

given to research colleagues, Eun-Sun Park and Chih-Chun Huang, for their friendship

and assistance.

Sincere appreciation is given to Incorporated (Raleigh, NC) for financial

support.

Finally, the author would like to express her gratitude to her husband, Feng

Chen, her daughter, Rachel W. Chen, her parents and her sisters for their love, moral

support and encouragement.

ii

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. TABLE OF CONTENTS

ACKNOWLEDGMENTS------ii

LIST OF TABLES------v

LIST OF FIGURES------vi

LIST OF ABBREVIATIONS------viii

ABSTRACT ------xi

CHAPTER 1 INTRODUCTION AND REVIEW OF LITERATURE------1 1.1: Overview of cotton and cottonseed products ------1 1.2: Gossypol ------3 1.2.1: Occurrence of gossypol ------3 1.2.2: Physiochemical properties of gossypol ------5 1.2.3: Utilization and physiological properties of gossypol ------12 1.2.4: Toxicity and detoxification of cottonseed products ------14 1.2.5: Gossypol analysis ------19 1.3: Theory of enzyme immunoassay ------22 1.3.1: Enzyme immunoassay ------22 1.3.2: Antibody structure ------24 1.3.3: Antibody-antigen (Ab-Ag) interactions ------26 1.3.4: Polyclonal antibody (PAb) and monoclonal antibody (MAb) production ------26 1.4: Research objectives ------32

CHAPTER2 MATERIALS AND METHODS------34 2.1: Materials------34 2.2: Methods ------36 2.2.1: Instrumentation ------36 2.2.2: Modeling and statistical analysis ------37 2.2.3: Preparation of experimental solutions ------38 2.2.4: Preparation of immunogen ------39 2.2.5: Preparation of solid-phase conjugates ------41 2.2.6: Measurement of gossypol groups in conjugates ------41 2.2.7: Polyclonal antibody (PAb) production ------42 2.2.7.1: Rabbit immunization ------42 2.2.7.2: Antibody capture noncompetitive ELISA for screening sera-- 42 2.2.7.3: Optimization of ELISA ------43 2.2.8: Monoclonal antibody (MAb) production ------44 2.2.8.1: Immunization of mice and serum test ------44 2.2.8.2: Myeloma cell culture ------45

iii

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2.2.8.3: Recovery of splenocytes ------46 2.2.8.4: Production of hybridomas ------46 2.2.8.5: Culture, screening, and selection of hybridomas ------46 2.2.8.6: Cell cloning ------47 2.2.8.7: In vitro production of monoclonal antibodies (MAb) ------49 2.2.8.8: Purification of monoclonal antibodies (MAb) ------49 2.2.8.9: Freezing, storage and thawing of cell lines ------49 2.2.9: Crystallization of isomeric gossypol- ------50 2.2.10: Preparation of various forms of gossypol ------50 2.2.11: Antibody capture competitive ELISA ------51 2.2.12: Gossypol analysis in cottonseed products ------52

CHAPTER 3 RESULTS AND DISCUSSION------54 3.1: Effects of reducing agent and pH on production of protein-gossypol conjugates—54 3.2: Production of polyclonal antibodies (PAb) ------59 3.2.1: Monitoring PAb production ------59 3.2.2: Optimization of ELISA for PA b ------59 3.2.2.1: Effects of microtiter plates on immobilization of BSA-G 59 32.2.2: Selection of assay buffers ------62 3.2.2.3: Effects of incubation time and temperature on interactions of Ab-Ag ------69 3.2.2.4: Effects of solvents on antibody capture noncompetitive ELISA------72 3.2.2.5: Effects of Ab and BSA-G concentrations ------75 3.2.3: Effects of immunogen preparation method on PAb production 75 3.3: Production of monoclonal antibodies ------78 3.3.1: Mouse immunization ------78 3.3.2: Production and selection of hybridomas ------80 3.3.3: Correlation between cell growth and antibody production ------81 3.4: Effects of reaction pH and structure of derivatizing reagents on gossypol derivatization ------82 3.5: Isolation of (+/-)-gossypol isomers ------86 3.6: Evaluation of antibody specificity using different forms of gossypol in an antibody capture competitive ELISA ------86 3.7: Application of ELISA for the analysis of gossypol in cottonseed products ------100 3.8: Future research ------107

CHAPTER4 SUMMARY AND CONCLUSIONS------109

REFERENCES------111

APPENDICES------123

VITA ------129

iv

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. LIST OF TABLES

1. Cottonseed production and main producers ------1

2. Gossypol groups per BSA in BSA-G conjugates ------55

3. Competition using gossypol-lysine as competitor ------80

4. Detection limit for antibodies using gossypol derivatives as competitors ------97

5. Gossypol contents of cottonseed products ------101

v

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. LIST OF FIGURES

1. Chemical structure of gossypol ------4

2. Tautomers of gossypol ------7

3. Antibody capture competitive ELISA ------23

4. Antibody basic unit structure ------25

5. Diagrammatic representation of the production of PAb (polyclonal antibodies) from rabbits------28

6. Diagrammatic representation of the production of MAb (monoclonal antibodies) from m ice ------30

7. The HAT selection system ------31

8. Gossypol-protein conjugate formation via a Schiff base intermediate ------40

9. MALDI-TOF-MS spectrograms of BSA and BSA-G conjugates ------56

10. MALDI-TOF-MS spectrograms of BSA and BSA-G conjugates ------57

11. MALDI-TOF-MS spectrograms of BSA and BSA-G conjugates ------58

12. Antibody production during immunization period from rabbit #16 ------60

13. Antibody production during immunization period from rabbit #15 ------61

14. Comparison of microtiter plates on ELISA for rabbit #16 PAb ------63

15. Comparison of microtiter plates on ELISA for rabbit #15 PAb ------64

16. Comparison of blockers on ELISA for rabbit #15 PAb ------66

17. Effect of dilution buffer on ELISA for rabbit #16 PAb ------67

18. Effect of dilution buffer on ELISA for rabbit #15 PAb ------68

19. Effect of incubation time and temperature on Ab-Ag interactions for rabbit #16 PAb------70

20. Effect of incubation time and temperature on Ab-Ag interactions for rabbit #15 PAb------71

vi

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 21. Effect of acetone concentration on ACN-ELISA ------73

22. Effect of N,N-dimethylformamide concentration on ACN-ELISA ------74

23. ELISA checkerboard for rabbit #16 PAb ------76

24. ELISA checkerboard for rabbit #15 PA b ------77

25. Antibody production from mice after four immunizations ------79

26. The growth rate of MAb cell line ------83

27. MAb production during culture ------84

28. Absorption spectra of gossypol and gossypol derivatives in 10% methanol ------85

29. HPLC chromatograms of (+/-)-gossypol isomers ------87

30. ELISA inhibition curves using gossypol and different gossypol derivatives for rabbit #15 PAb------89

31. ELISA inhibition curves using additional gossypol derivatives for rabbit #15 PAb------90

32. ELISA inhibition curves using different gossypol derivatives for MAb ------91

33. ELISA inhibition curves using different gossypol derivatives for MAb ------92

34. ELISA inhibition curves using different gossypol derivatives for MAb ------93

35. Effects of gossypol storage time on ELISA inhibition curves for rabbit #15 PAb - 95

36. Effects of gossypol storage time on ELISA inhibition curves for MAb ------96

37. Comparison of (+/-)-gossypol derivatives in ACC-ELISA for MAb ------98

38. Comparison of (+/-)-gossypol derivatives in ACC-ELISA for MAb ------99

39. Total Gossypol in cottonseed samples analyzed by ELISA (10% methanol for sample delivery) and AOCS official method ------102

40. Free Gossypol in cottonseed samples measured using ELISA (10% methanol for sample delivery) and AOCS official method ------104

41. Free Gossypol in cottonseed samples determined using ELISA (10% acetone for sample delivery) and AOCS official method ------106

vii

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. LIST OF ABBREVIATIONS

A/Ao relative absorbance (see Section 2.2.2)

Ab antibody

Abs. absorbance

ABTS 2,2’-azino-bis (3-ethylbenthiazoline-6-sulfonic acid)

ACC-ELISA antibody capture competitive ELISA

Ag antigen

AOCS American Oil Chemists’ Society

ATP adenosine triphosphate

BG Bound Gossypol

BSA bovine serum albumin

BSA-G bovine serum albumin-gossypol conjugate

ca about

ACN-ELISA antibody capture noncompetitive ELISA

CSM cottonseed meal

CV coefficient of variation

DMF N,N-dimethylformamide

DMSO dimethylsulfoxide

DNA deoxyribonucleic acid

DNFB dinitrofluorbenzine

El enzyme immunoassay

EI-MS electron impact-MS

ELISA enzyme linked immunosorbent assay

viii

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. FDA Food and Drug Administration

FG Free Gossypol

FIA fluoroimmunoassay

g gram

GLC gas-liquid chromatography

HAT hypoxanthine, aminopterin and thymidine

HB high binding

HBX high binding extra

HGPRT hypoxanthine guanine phosphoribosyle transferase

HIV human immunodeficiency virus

HPLC high performance liquid chromatography

h hour(s)

I50 concentration of analyte giving 50% reduction in absorbance

Ig immunoglobulin (can be IgM, IgG, etc.)

IR infrared spectroscopy

K.eq equilibrium constant

LPH Limulus polyphemus hemolymph

LPH-G Limulus polyphemus hemolymph-gossypol conjugate

MAb monoclonal antibodies

MALDI-TOF matrix-assisted laser desorption ionization-time of flight

min minute(s)

MS mass spectrometry

MW molecular weight

ix

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. MWCO molecular weight cut off

nm nanometer

NMR nuclear magnetic resonance

NZW New Zealand White

°C degree Celsius

PAb polyclonal antibodies

PBS phosphate-buffered saline

PBST phosphate-buffered saline with Tween 20

PEG polyethylene glycol

PVC polyvinylchloride

RIA radioimmunoassay

RPMI 1640 Rosewell Park Memorial Institute 1640

RT room temperature

sc. subcutaneous

TG Total Gossypol

TLC thin layer chromatography

TNBS trinitrobenzinesulfonic acid

Tween 20 polyoxyethylene-20-sorbitanmonolaurate

UV ultraviolet

pi microliter

pM micromolarity

pm micrometer

x

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ABSTRACT

Gossypol is a reactive polyphenolic compound present in the cotton plant.

During cottonseed processing, gossypol can react with other compounds to form bound

gossypol. Gossypol analyses using chemical methods do not always agree with its

bioavailability to animal possibly because of various bound forms of gossypol with

different stabilities in vivo and the (+/-)-gossypol enantiomers with different biological

activities. The objectives of this research were to produce anti-gossypol antibodies and

use antibodies to study gossypol chemistry and to analyze gossypol in cottonseed

samples and cottonseed by-products.

Polyclonal antibodies were produced from rabbits after they were immunized

with gossypol-LPH (.Limulus polyphemus hemolymph). Monoclonal antibodies were

developed by fusion of NS-1 myelomas and immune mouse splenocytes, and the

hybridomas secreting anti-gossypol antibodies were selected and cloned.

Immunoassay parameters were evaluated for their effects on antibody capture

noncompetitive ELISA (ACN-ELISA). It was found that microtiter plates, time and

temperature of incubation for antibody-antigen interactions, and reagent concentrations

for dilution of both primary and secondary antibodies could greatly influence the signal

to noise ratios in ACN-ELISA.

(+)- Gossypol and (-)-gossypol isomers were isolated by crystallization from

acetone and derivatized with L-(+)-lysine and L-2-amino-l-propanol. (+/-)-Racemic

gossypol was derivatized with L-(+)-lysine, L-(+)-arginine, ethanolamine, L-2-amino-l-

propanol, 3-amino-1-propanol, L-glutamine, L-(-)-tryptophan, L-tyrosine and para-

amino-benzoic acid.

xi

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Different forms of gossypol were used in an antibody capture competitive-

ELISA to evaluate antibody sensitivity and specificity. Approximately 0.2 pg/ml to

over 125 pg/ml of gossypol equivalents caused 50% inhibition of binding (I 50) of rabbit

PAb to the solid-phase, and I 50 values ranged from 7.4 to 55 pg/ml for MAb cell lines,

when (+/-)-racemic gossypol derivatives were used competitively. I 50 values from 7.5

to 13.5 pg/ml were obtained for (+)- and (-)-isomeric gossypol derivatives as

competitors for MAb. Underivatized gossypol as competitor gave much higher I 50 value

for PAb and showed no competition for MAb.

Polyclonal antibodies were evaluated for gossypol analyses of cottonseed

products in an antibody capture competitive ELISA. The results showed good

correlation (r2=0.96) for Free Gossypol analyses and poor correlation (r2=0.43) for Total

Gossypol analyses between ELISA and AOCS official methods under the conditions of

the methods.

xii

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CHAPTER 1

INTRODUCTION AND REVIEW OF LITERATURE

1.1: Overview of cotton and cottonseed products

Cotton has long been known as nature’s unique food and fiber plant. It is

produced worldwide in tropical and subtropical regions. The People’s Republic of

China is the largest producer of cottonseed, followed by the United States and India

(Table 1).

Table 1. Cottonseed production (million metric tons) and main producers (USDA, 1998a).

Cottonseed (million metric tons) Countries Average Forecast 91/92-96/97 1997/98

China 8.05 7.72

United States 6.16 6.60

♦FSU-12 3.69 3.15

India 4.57 5.33

Pakistan 3.29 3.05

Brazil 0.86 0.65

Others 6.67 8.57

*FSU-12 represents former Soviet Union-12

World demand for cotton is growing at the rate of about two million bales a year

and if the U.S. maintains its share of world production, the U.S. will be producing about

1

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 20 million bales by the year 2000. This trend in cotton production also applies to

cottonseed production. Cottonseed ranks second to that of soybean in terms of world’s

oilseed production, but only ranks sixth in world production of edible oils

because some cottonseeds are used for animal feed, such as feed for cattle and dairy

cows (USDA, 1998b).

Cottonseed, like other oilseeds, is processed by either expression or extraction

methods. There are four different milling methods that have been used for cottonseed:

(1) hydraulic pressing uses the high-pressure expression to remove oil, (2) screw

pressing is another mechanical expression method, which produces high temperature

that renders protein less soluble, (3) prepressing-solvent extraction is a combine action

of modified screw pressing and solvent extraction process that can supply a better

quality of meal proteins than screw processing, and (4) direct solvent extraction

removes the oil by using hexane or other solvents and provides a high quality protein

feed.

The composition of cottonseed, which includes oil, protein, carbohydrates,

phosphorous compounds and minerals, varies considerably depending on species,

variety, and environment. Oil is embedded in the tissue of cottonseed as droplets

(Markman, 1968).

Cottonseed averages about 45% hull and linters, and 55% kernel. The

cottonseed kernel is a pointed ovoid body approximately 8-12 mm in length, in which

there are innumerable dark spots. These dark spots are pigment glands and are unique

to cottonseed. The major component of the pigment glands is free gossypol. The

processing method affects the quantity of free gossypol and the protein value of

2

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. cottonseed meals (CSM). During screw pressing, most of the free gossypol binds to

amino acids of proteins and other compounds, which lowers the nutritional value of the

protein. The newer methods such as prepress solvent extraction and direct solvent

extraction produce CSM with high quality protein, but they considerably increase the

content of free gossypol.

The presence of gossypol limits the application of cottonseed protein for food

and feed. In the United States, any cottonseed protein products intended for human use

must contain no more than 450 ppm free gossypol as set by FDA (1974). The Protein

Advisory Group of the United Nations Food and Agriculture and World Health

organizations (FAO/WHO) has set limits of 600 ppm of free gossypol and 12,000 ppm

total gossypol for human consumption. Also, some caution was suggested when using

both cottonseed meal and whole cottonseed in dairy rations due to the presence of the

yellow polyphenoiic pigment gossypol (Poore and Rogers, 1998).

1.2: Gossypol

1.2.1: Occurrence of gossypol

Gossypol was first discovered and isolated as a crude pigment by Longmore in

1886 from cottonseed oil foots, a mixture of precipitated soaps and gums produced in

the refining of crude cottonseed oil with sodium hydroxide.

The chemical formula for gossypol C 30H30O8 was first established by Clark in

1927. The whole structure of gossypol was verified in 1958 by Edwards when gossypol

was synthesized and shown to be 1,1', 6,6', 7,7'-hexahydroxy-3,3'-dimethyl-5,5'-

diisopropyl-2,2'-binaphthyl-8,8'-dialdehyde (Figure 1). A comprehensive review of the

3

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 00

X Figure Chemical 1. structure of gossypol O

ua CO in.

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. discovery, determination of structure, and chemistry of gossypol was published by

Adams et al. (1960) and Markman (1968).

Gossypol, as it exists in cottonseed, is contained in pigment glands and

constitutes 20-40% of their weight and results in 0.4 to 1.7% of whole kernel. Gossypol

glands are found not only in the seeds of cotton but also in some other parts, such as the

bark of cotton plant roots, leaves, seed hulls, and flowers. However, the gossypol

content varies widely, depending on the species and varieties of cotton plant, on

climatic and soil conditions of the region, water supply, agrotechnical treatment, and in

particular, on the amount and composition of fertilizers used (Markman, 1968;

Stansbury et al., 1954). The changes of gossypol content during different stages of

maturity also have been reported (Caskey and Gallup, 1931; Gallup, 1927, 1928). The

gossypol content of cotton plant is of importance chiefly in related to some of its unique

characteristics and its effect on the use of cottonseed meal as an animal feed, the oil in

food products, and the cottonseed flour for human consumption.

1.2.2: Physiochemical properties of gossypol

Gossypol is a chiral molecule because of steric hindrance to rotation about the

intemaphthyl bond. Cass et al. (1991) measured gossypol enantiomer ratio by HPLC

analysis after conversion to the Schiff base diastereoisomers and found that an

enantiomeric excess of (+)-gossypol was in each variety of Gossypium arboreum,

G.herbaceum and G.hirsutum, and (-)-gossypol was in excess in each variety of

G.barbadense investigated. The results agreed with that from Zhou and Lin (1988).

Gossypol contains polar groups (6 hydroxyl and 2 aldehyde groups) making it

soluble in most organic solvents, such as methanol, ethanol, isopropanol, butanol,

5

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ethylene glycol, dioxane, diethyl ether, acetone, ethyl acetate, chloroform, carbon

tetrachloride, ethylene dichloride, phenol, pyridine, melted naphthalene, and heated

vegetable oil. It is less soluble in glycerine, cyclohexane, benzene, gasoline, and

petroleum ether. The presence of 2 heavy dialkylnaphthalene groups makes it insoluble

in water (Markman, 1968).

In 1960, Adams et al. proposed that gossypol existed in three tautomeric forms:

aldehyde (a), hemiacetal (b) and ketol (c) (Figure 2) in different solvents. The use of

nuclear magnetic resonance (NMR), mass spectral analysis and UV spectrometry has

illustrated its structure changes in various solutions. In basic solvent systems, gossypol

existed mainly as ketol, and in ordinary inert solvents and acidic conditions, gossypol

existed mainly in the aldehyde forms (Stipanovic, et al., 1973), while in polar solvents

such as dimethyl sulfoxide (DMSO) with alkali condition, the hemiacetal form occurred

in dynamic equilibrium with the aldehyde form (Abdullaev et al., 1990).

Gossypol is markedly reactive due to the reactivity of carbonyl and phenols, so

it has been the compound of the greatest concern in cottonseed. When removing the oil

from the cottonseed, most gossypol reacts with other compounds in cottonseed. This

results in undesirable color and a low value protein in cottonseed meal. Some gossypol

will remain in the oil. This will be removed during refining but will increase the oil

waste (Jones, 1979; Pons et al., 1951).

In order to better describe gossypol in cottonseed products, the terms “Free

Gossypol”, “Bound Gossypol” and “Total Gossypol” are used, respectively. Free

Gossypol (FG) is defined as gossypol and gossypol derivatives in cottonseed products

that can be extracted by 70% aqueous acetone (AOCS, Ba 7-58). Bound Gossypol

6

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure Figure 2. Tautoniers of gossypol

7

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (BG), formed during cottonseed processing by reaction of gossypol with other

compounds, is not soluble in aqueous acetone. Total Gossypol (TG) is defined as

gossypol and gossypol derivatives, both FG and BG, that can react with 3-amino-1-

propanol in dimethylformamide solution (AOCS, Ba 8-78). This AOCS method

(AOCS, Ba 8-78) only measures gossypol, gossypol analogs and gossypol derivatives

that have an available aldehyde moiety for the derivatization reaction. It should be

noted that there might be a fraction of gossypol that is unavailable for this reaction and

therefore is not included in the TG value. FG and TG are determined empirically, and

BG is determined mathematically (BG=TG-FG)

Some evidence indicates that the majority of gossypol is in the form of Schiff

bases from the condensation between aldehydic groups of gossypol and e-amino groups

of proteins during cottonseed processing (Cater, 1968; Lyman et al., 1959). However,

this chemical complex alone can not fully account for gossypol behavior. Gossypol

may be chelated by iron in cottonseed products to form insoluble metal complexes

(Muzaffaruddin and Saxena, 1966), may form gossypol polymers (Anderson et al.,

1984), or may be oxidized (Scheiffele and Shirley, 1964). The phenolic groups may

react to form esters and ethers with other carboxylic compounds and phenols in

cottonseed plants. Two types of reactive functional groups in gossypol: aldehydes and

phenols, may give rise to several forms of bound gossypol.

In past years, much work has been done on the Schiff base reaction between

gossypol and amino groups. During cottonseed processing, the moist heat treatment of

cottonseed kernels catalyzes the chemical reaction between gossypol and the free amino

groups of cottonseed protein. Soluble gossypol (FG) in aqueous acetone was thus

8

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. converted to insoluble Bound Gossypol (BG). Baliga (1956) found that lysine accounted

for most of the free amino groups of cottonseed protein and that reaction of gossypol with

cottonseed protein occurred mainly with e-amino groups of lysine, thus the lysine

availability decreased to about one-half of the original value (82.9% to 48.7%) when the

purified protein was allowed to react with gossypol. Involvement of arginine in the

reaction has also been reported (Martinez et al., 1961). In 1979, Damaty and Hudson

studied the chemical nature of interaction using selective proteolysis, gel filtration and

amino acid analysis and concluded that lysine and the more hydrophobic amino acid in

cottonseed protein were easily to take part in the formation of insoluble material. In 1988,

Reddy and Rao studied the interaction between gossypol and gossypin, congossypin and

glycinin using a difference spectral method and found that the interaction was completely

reversible, and suggested that hydrophobic and ionic interactions were involved in the

reaction of gossypol with proteins. Later, Strem-Hansen et al. (1989) studied the

interaction of gossypol with amino acids and peptides using circular dichroism (CD) and

nuclear magnetic resonance (NMR) and gave evidence that hydrophobic interaction may

be responsible for a significant proportion of the interaction between gossypol and

proteins. This opinion was also supported by the finding that gossypol bound

competitively at the bilirubin binding site on albumin (Royer and Vander Jagt, 1983). This

site is known to be linked with many hydrophobic residues, with only one or two

positively charged amino acids being present. The total binding energy of the complex

would be the sum of the hydrophobic interaction and Schiff base formation.

Another investigation (Lyman et al., 1959) of gossypol-protein complexes

revealed that when gossypol combined with crystalline BSA or cotton protein, the point of

9

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. attachment is the e-amino group of lysine. The molar ratios of free e-amino groups to

gossypol varied according to the experimental conditions, but averaged about 1.5 moles of

lysine being bound to 1 mole of gossypol. At low concentrations of gossypol, the molar

ratio of lysine to gossypol was 2:1, indicating that both of the reactive aldehyde groups of

gossypol were linked to lysine. Sedimentation velocity studies of these gossypol-protein

complexes suggested the presence of from 1 to 4 different compounds, thus indicating that

gossypol probably formed a cross-link between two or more protein molecules.

Later, model complexes of gossypol with amino acids (lysine, asparagine,

glutamine, and glycine), peptides (hippuryl-L-lysine, L-alanyl-L-lysine, glycyl-L-lysine,

L-histidyl-L-lysine) and purified proteins (glandless cottonseed protein, cottonseed

globulin, insulin) were synthesized and partially characterized (Cater, 1968), and it was

concluded that the rate of reaction of gossypol with amino acids increased with an increase

of pH (5.7 to 7.5), and was shown to be related to the distance of the amino group from the

carboxyl group within molecule ( e.g. the e-amino group is more reactive than an a-amino

group of an amino acid). Whaley et al. (1984a and 1984b) studied the reaction of (+)-

gossypol with BSA, human serum albumin, lactate , malate dehydrogenase,

alkaline phosphatase, lysozyme, protamine and poly-L-lysine and measured the products

using circular dichroism (CD) and found that (+)-gossypol bound to albumin with the

same affinity as (+/-)-gossypol. The reactions between gossypol and BSA, glandless

cottonseed meal and glanded cottonseed meal also were studied by Couch and Thomas

(1976).

A Schiff base is a relatively labile bond that is readily reversed by hydrolysis in

aqueous solution. It can be chemically stabilized by reduction. The formation of a

10

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Schiff base is enhanced at alkaline pH values, but is still not completely stable unless

reduced to a secondary or tertiary amine linkage (Hermanson, 1995). A number of

reducing agents can be used to convert the Schiff base into a secondary amine. The

addition of sodium borohydride or sodium cyanoborohydride will result in reduction of

the Schiff base intermediate into a relatively stable secondary amine. Both borohydride

and cyanoborohydride have been used for reductive amination purposes, but

borohydride will reduce the reactive aldehyde groups to hydroxyls at the same time

when it converts Schiff bases present to secondary amines. Cyanoborohydride, by

contrast, is a milder reducing agent that is at least five times milder than borohydride in

reductive amination processes. Cyanoborohydride does not reduce aldehydes, but it is

very effective for Schiff base reduction (Lane, 1975; Hermanson, 1995). Thus, higher

yields of conjugate formation can be obtained using cyanoborohydride instead of

borohydride for the stabilization of Schiff base products.

The available lysine in BSA, glandless cottonseed meal, solvent extracted

cottonseed meal and gossypol complexes of these materials has been determined by

different chemical methods, such as the dinitrofluorobenzine (DNFB), sodium borohydride

(NaBH*), and trinitrobenzenesulfonic acid (TNBS) methods. However, it was found that

different methods could give different results (Couch and Thomas, 1976). By measuring

available lysine, Anderson et al. (1984) found that most gossypol was attached to other

groups (not lysine) in the proteins or had formed large gossypol polymers, binding to very

few lysine groups. This opinion is different from previous reports (Baliga, 1956; Cater,

1968; Lyman etal., 1959; Martinez etal., 1961).

11

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The reactions have also been studied between gossypol and other amine

compounds such as para-amino-benzoic acid, y-amino butyric acid, anisidine and (+)-l-

phenylethylamine. Interested readers can find the related articles published by Gdaniec

(1994) and Cater (1968).

1.2.3: Utilization and physiological properties of gossypol

Gossypol is one of the secondary metabolites defined as compounds that are not

ubiquitous and that do not play an indispensable role in plants that synthesize them. In

the natural environment, plant phenolic metabolites play an important role as

allelochemicals. They function as signals between plants, between plants and

symbiontic or pathogenic organisms to protect plants against microbes or insects

(Heller, 1993). Being a polyphenolic compound, gossypol is an anti-oxidant

(Mar km an, 1968).

Gossypol and many of its derivatives are reactive, so gossypol may serve for the

synthesis of various useful organic compounds with unique structures. For example,

gossypol pitch, obtained from cottonseed oil soapstock by distillation, is used in the

production of core binder agent to improve road surfaces. In addition, taking advantage

of its optically active nature, the (+)-enantiomer of gossypol has been shown to be a

useful CD (circular dichorism) probe to study interactions with human and bovine

serum albumin (Whaley et al., 1984a and 1984b).

Gossypol also has a great number of pharmacological activities. Gossypol has

been studied extensively as a potential male contraceptive agent in several mammalian

species, including rats (Hadley et al., 1981; Lin et al., 1980; Lin et al., 1985), mice

(Coulson et al., 1980), monkeys (Shandilya et al., 1982), hamsters (Matlin et al., 1985;

12

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Waller et al., 1984), rabbits (Chang et al., 1980), bulls (Arshami and Ruttle, 1988) and

human (National Coordinating Group on Male Antifertility Agents, 1978). The basis of

this gossypol-induced infertility is reduction in sperm counts, abnormal sperm structure,

and loss of sperm forward motility (Chang et al., 1980; Lin et al., 1980; Shandilya et al.,

1982) and it was found that (-)-gossypol is more active in antifertility function than (+)-

gossypol (Matlin et al., 1985).

Lin et al. (1989 and 1993) reported that gossypol inhibited the replication of

human immunodeficiency virus-type 1 (HIV-1) and found the (-)-enantiomer of

gossypol to be more inhibitory compared to the (+)-enantiomer. Gossypol is capable of

inhibiting the growth of a variety of cell lines derived from epithelial tumors (Benz et

al., 1988; Benze et al., 1991; Wu et al., 1989), breast carcinoma and esophageal cancer

(Hu et al., 1994; Gilbert et al., 1995) and transplantable tumors (Rao et al., 1985).

These disruptions include inhibition of cytoplasmic and mitochondrial enzymes

involved in energy production (Ueno et al., 1988) and uncoupling of oxidative

phosphorylation (Flack et al., 1993; Abou-Donia and Dieckert, 1974). Depletion of

cellular adenosine triphosphate (ATP) has been demonstrated in cultured tumor cells

(Keniry et al., 1989). Gossypol also inhibits key nuclear enzymes responsible for DNA

replication and repair, including DNA polymerase a (Rosenberg et al., 1986) and

topoisomerase II, and block DNA synthesis in HeLa cells (Wang and Rao, 1984).

These properties make gossypol a potential antineoplastic agent.

Gossypol also can inhibit glutamic, malic and alcohol . It was

found that (-)-gossypol is 13 times more active in inhibiting the enzyme than (+)-

gossypol and (-)-gossypol is 3 times more active in inhibiting amebic culture growth

13

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (Gonzalez-Garza et al., 1992; Gonzdlez-Garza et al., 1993). The studies on the

inhibition of rabbit sperm acrosomal enzymes by gossypol indicated that azocoll

proteinase, acrosin, neuraminidase, and aryksukfatase were significantly inactivated by

gossypol at 12-76 pM, and hyaluronidase, (i-glucuronidase, and acid phosphatase were

inhibited at higher concentration of gossypol at 380 pM (Yuan et al., 1995).

Gossypol has general antifimgal activities with LD 50 values from 20 to 100 ppm

of pure gossypol (Bell, 1967) and has inhibitory effect on microorganisms including

aerobic sporeformers and lactobaciili and some yeasts (Margalith, 1967). It also has

been suggested to be of value in the treatment Chagas disease (Montamat et al., 1982).

Another important aspect of gossypol and its derivatives is that they could lower plasma

cholesterol levels (Shandilya et al., 1982). In 1994, Acheduma et al. reported that

gossypol and related derivatives might in some way contribute to the lowering of

cholesterol level in rats. Later, Nwoha (1995) measured serum constituents of gossypol

treated, protein-malnourished Wistar rats and found that the administration of gossypol

decreased the levels of serum cholesterol.

1.2.4: Toxicity and detoxification of cottonseed products

Cottonseed is an important protein supplement for livestock. Cottonseed meal, a

by-product of the cottonseed oil industry, contains about 40-50% crude protein and is a

good protein resource for cattle. Cottonseed has been utilized as a feed for livestock

since the early 19th century.

A number of investigators have indicated that gossypol is toxic to monogastric

animals and also to young ruminants. The most common toxic effect of gossypol is

cardiac irregularity which causes death of the animal, because gossypol prevents the

14

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. liberation of oxygen from oxyhemoglobin (Menaul, 1923). The toxicological effects of

gossypol have been classified in three levels, which are: ( 1 ) acute doses causing

circulatory failure, (2) subacute doses causing pulmonary edema, and (3) chronic doses

causing symptoms of ill health and malnutrition (Abou-Donia, 1976). The doses for

each level are different for different species.

Since gossypol exhibits this dose response, there are some restrictions for the

safe use of cottonseed meal as livestock feed. A study using male rats (Eagle and

Davies, 1958) showed that oral LD 50 is from 1061 to 2170 mg/kg administering

gossypol as cottonseed pigment glands (27.0 to 37.8% of gossypol), and oral LD 50 is

from 2200 to 2600 mg/kg with pure gossypol (ca 100% gossypol). The cottonseed

pigment glands seemed more toxic than pure gossypol for rats in this study. El-

Nockerashy et al. (1963) had similar findings in studying the acute oral toxicity for rats

by feeding cottonseed pigment glands, gossypol, diaminogossypol and gossypurpurin

with LD50 1120, 2570, 3270 and 6680 mg/kg body weight, respectively. Calhoun et al.

(1990b) compared toxicity of gossypol acetic acid and Free Gossypol in cottonseed

meal and pima cottonseed by feeding lambs, and found that oral administration of

gossypol acetic acid is much more toxic to young lambs than feeding equivalent

amounts of Free Gossypol from cottonseed meal or cottonseed. Gossypol in different

status may have different stabilities in the whole digestive tracts of either ruminants or

nonruminants. Gossypol in unbroken glands may be protected by the gland envelope,

and the other ingredients in the diet may also affect the gossypol stability. For rainbow

trout (Herman, 1970), 95 ppm of Free Gossypol in the diet caused histological changes

in the liver and kidney; 100 ppm could cause pathological changes; 290 ppm could

15

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. suppress growth; at 531 ppm level, fish lost weight for a period of time and suffered

severe reduction of hematocrit, hemoglobin and plasma proteins. A study using channel

catfish (eight weeks old) showed that diets containing approximately 0.14% Free

Gossypol from cottonseed meal or gossypol acetate added in meal could depress

growth. A level of 0.09% or less Free Gossypol in the diet seemed to be safe if all

essential amino acids were in balance (Dorsa and Robinnette, 1982). Swine showed

toxicosis when fed 200 to 400 ppm Free Gossypol in the diet and it was suggested that

Free Gossypol should be less than 100 ppm for growing and fattening swine (Haschek

etal., 1989).

Cattle were found to be more tolerant to gossypol than young calves due to the

action of the rumen (Morgan, 1989). Lindsey et al. (1980) studied the physiological

responses of lactating cows to gossypol from cottonseed meal and suggested that

detoxification occurred in mature ruminants consuming cottonseed meal containing

high Free Gossypol. It was demonstrated that rumen microorganisms were responsible

for the detoxification of gossypol and it was proposed that gossypol formed a gossypol-

protein complex (gossypol-microbial protein) with soluble protein in the rumen liquor

which was not absorbed in the digestive tract. This complex was very stable during

enzymatic hydrolysis (Reiser and Fu, 1962). The microbial population in the rumen is

regulated by the ecological balance of conditions that tend to prevail there (Van Soest,

1982). The higher microbial yield characteristic of the rumen may be important in

supplying enough protein to capture the gossypol.

However, if gossypol content is too high in the diet, it will suppress the rumen’s

ability to detoxify. It was found that 348 to 414 mg of Free Gossypol per day would

16

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. cause congestive heart failure in adult goats (East et al., 1994) and 8 mg free and 222mg

of Total Gossypol (in extracted cottonseed meal)/kg body weight per day could cause

physical and hematological changes at 2 weeks for cows (Lindsey et al., 1980).

Because of the toxicity of gossypol to animals, a glandless variety of cotton was

developed in the early 1960s. It was believed that the value of cottonseed oil and meal

would be improved if gossypol were not present. However, this glandless strain is

easily susceptible to insect attack, and attracts field mice and other rodents (Stipanovic

et al., 1986; Lusas and Jividen, 1987). Dilday (1986) developed a cotton plant through

an interspecific cross of tetraploid (2N=52) Gossypium hirsutum x diploid L. (2N=26)

G. sturtianum Willis. This plant showed gossypol glands in vegetative foliar and

fruiting tissues but not in the seed. However, this approach needs study to prove the

stability and yeild for this plant.

Another approach for gossypol removal is by a flotation process based on the

specific gravity difference or by centrifugal force, such as liquid cyclone process which

is a physical separation of intact pigment glands (Gardner et al., 1976).

Chemical methods are commonly used methods for gossypol detoxification.

Bressani et al. (1964) reported that an alkaline pH of cooking, associated with calcium

ions, was important in reducing Free Gossypol and Total Gossypol in cottonseed flour

used for human foods. The addition of calcium increased the effectiveness of the

gossypol-iron complex formation, resulting in full protection from gossypol toxicity.

Iron sulfate is an inexpensive source of Fe that could also reduce gossypol intoxication

in nonruminant animals (Ullrey, 1966).

17

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Kemmerer et al. (1966) reported that addition of iron sulfate to feed could

prevent yolk discoloration in cold-stored eggs caused by gossypol. Muzaffaruddin and

Saxena (1966) showed that a 1:1 molar ratio of ferric iron to gossypol formed an iron

gossypol complex, in which the two perihydroxyl groups of gossypol were the most

plausible sites where the ferric irons chelated. Barraza et al. (1991) investigated the

efficacy of iron sulfate and feed pelleting to detoxify Free Gossypol in cottonseed diets

for dairy calves. Ferrous sulfate added to diets for swine contained 244 and 400 ppm

Free Gossypol (FG) at a molar ratio of 0.5:1 for iron to gossypol, gave partial

detoxification and a 1:1 ratio gave complete detoxification. The addition of

phospholipids in cottonseed meal or cottonseed meat coupled with cooking could

eliminate some Free Gossypol (FG) and improve the protein quality (Yannai and

Bensal, 1983).

Recently, Calhoun (1999) and Wan (1999) studied the effects of different

processing methods on the gossypol availability through feeding lambs and cattle.

Gossypol toxicity was influenced by the methods of cottonseed processing, dietary

concentrations of iron, calcium, and the presence of other toxic and

chemicals found in cottonseed, as well as the amount and duration of gossypol

consumption.

It has been believed for a long time that BG is physiologically inactive and FG

is physiologically active. The FG content of cottonseed products however is not a good

predictor of the cottonseed product toxicity (Calhoun et al., 1990a; Calhoun, 1996; Eagle

and Davies, 1958; Eagle et al., 1956), and the measurements of BG, FG and TG (Total

Gossypol) are also insufficient. We would like to propose the BG fraction is not always

18

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. entirely inactive depending on the mechanism(s) of gossypol binding that occurs during

cottonseed processing, the type of animal consuming the gossypol and physical form of

the feed.

1.2.5: Gossypol analysis

Several methods for the quantitative determination of gossypol in cottonseed, oil,

press-cake, and meal have been reviewed by Markman (1968). These include gravimetric

methods, volumetric methods, colorimetric, spectrophotometric, polarographic and

luminescent methods.

Gossypol can be derivatized to form a trimethylsilyl (TMS) derivative and

determined by gas-liquid chromatography (Raju and Cater, 1967). Near-infrared

reflectance, which has been used in many applications for composition analysis (Norries

et al., 1976), also offers a possibility for the measuring gossypol content (Birth and

Ramey, 1982). The structural analysis of gossypol, gossypolone and correlated

derivatives were studied using electron impact-mass spectrometry (EI-MS) and infrared

spectrometry. Mass spectra and IR spectra obtained from these derivatives may aid in

the identification of compounds that are found in association with gossypol (Phillip and

Hedin, 1990).

The most common methods for gossypol analysis include high performance

liquid chromatography (HPLC) (Botsoglou, 1991; Chamkasem, 1988; Hron et al., 1990;

Stipanovic et al., 1988) and AOCS official methods (AOCS, Ba 7-58 and Ba 8-78).

Both methods measure either “Total Gossypol” or “Free Gossypol” defined by AOCS

official methods (Ba 7-58 and Ba 8-78). Total Gossypol (TG) is gossypol and gossypol

derivatives, both FG (Free Gossypol) and BG (Bound Gossypol), in cottonseed products

19

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. that can react with 3-amino-1-propanol in dimethylformamide solution to form a

diaminopropanol complex. Free Gossypol (FG) is gossypol or gossypol analogs which

can be extracted by 70% aqueous acetone. High performance liquid chromatography

(HPLC) has been used to determine gossypol and other individual aldehydes

in seed, leaves and flower buds, and in processed oils and meals (Chamkasem, 1988;

Hron et al., 1990; Nomeir and Abou-Donia, 1982; Stipanovic et al., 1988). The

gossypol determined using HPLC is not correlated with the results obtained from AOCS

official methods. The AOCS official method measures all the gossypol and gossypol

derivatives having available aldehydic groups. The AOCS official method may lead to

false readings due to the presence of non-gossypol aldehydic-containing compounds

that react with coloring agent (Stipanovic et al., 1984).

Most toxicity studies for animals with gossypol have been based on the AOCS

official methods and HPLC methods. Some questions exist about the validity of

gossypol analytical procedures because of inconsistencies between determined gossypol

content and the biological activities of the samples (Calhoun et al., 1990a; Calhoun,

1996; Eagle and Davies, 1958; Eagle, et al., 1956). One explanation for this

phenomenon could be the association of various compounds with Free Gossypol

yeilding different physiological activities (Calhoun et al., 1990a; Calhoun, 1996; Eagle

and Davies, 1958; Eagle, et al., 1956), and the existence of (+/-)- two gossypol

enantiomers, having different biological activities. Another reason may be that

different forms of bound gossypol have different stabilities in vitro and in vivo

(Calhoun, 1996), possibly because different cottonseed processing conditions favor

20

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. various forms of bound gossypol. The animals selected for bioactivity experiments also

would affect the results because of differing digestive environments.

Results related to available gossypol must be evaluated and interpreted

carefully, considering the dose and route of administration, the possibility of other toxic

materials, and the degree of inactivation of administered gossypol before and/or after

administration by reaction with components of the diet (Yu, 1987).

Because of the complicated reactivity of gossypol in cottonseed products and its

apparently variable physiological properties, specificity and accuracy of gossypol

analysis becomes more important. This is especially important when one considers

potential new applications. At the same time, the determination of FG, BG and TG is

insufficient, and it is also necessary to determine the forms in which gossypol exists and

their relationship with bioactivity.

For the reasons explained above, gossypol screening requires an improved

analytical method that could facilitate fast and accurate determination and improve the

management, study and utilization of cottonseed products for feed or food. Because of

the selectivity of antibody binding, an immunochemical method would possibly

simplify and improve current gossypol analyses. The acceptability of immunoassays is

increasing for pesticide and other environmental analyses (Abouzied et al., 1993; Chen

et al., 1995). The antibody specific binding make it possible to analyze crude extracts,

possibly both soluble and insoluble analytes such as matrix-bound gossypol. A low

detection limit is another advantage because this allows one to analyze a small sample

that has been sufficiently diluted to remove any interfering compounds. Monoclonal

antibodies (MAb) may be able to distinguish different forms of bound of gossypol and

21

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (+/-)-isomers of gossypol, and therefore an assay based on these antibodies may provide

greater accuracy of analysis.

1.3: Theory of enzyme immunoassay

1.3.1: Enzyme immunoassay

Immunoassays are analytical methods that are based on the specificity of

antibody-antigen (Ab-Ag) interaction. There are two classes of immunoassays

including isotopic immunoassay such as radiaoimmunoassay (RIA) and nonisotopic

immunoassay including fluoroimmunoassay (FIA) and enzyme immunoassay (El)

(Dixon, 1994). Isotopic immunoassay is based on competition for antibody between a

radioactive indicator antigen and unlabeled antigen in the test sample, and nonisotopic

immunoassays are different from isotopic immunoassays, mainly due to the type of

label used, and the means of endpoint detection. Enzyme immunoassays employ

enzyme markers as means of amplifying and visualizing the primary Ab-Ag binding

reaction. This method is widely used in quantitative and qualitative determination

(Nakamura et al., 1986). Enzyme tags are easy to handle, relatively inexpensive and

stable. Also, they can convert a colorless substrate to a colored product giving sensitive

and easy to interpret endpoints. This method, when one of the binding elements is

attached to a solid support, is termed an enzyme-linked immunosorbent assay (ELISA).

The use of a solid support enables easy separation of the unbound fraction from the

bound fraction.

An Ab capture indirect competitive ELISA will be used in this study. The basic

steps involved in this assay are shown in Figure 3. First, a standard amount of antigen

is immobilized on the surface of microtiter plate. Then, sample antigen to be analyzed

22

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. A____ A

Sample Ab AA B

Enzyme labeled 2nd Ab

Enzyme substrate

Figure 3. Antibody capture competitive ELISA (enzyme linked immunosorbent assay) A. Ag is immobilized on a plastic surface. B. Competition occurs for Ab binding sites between the immobilized Ag and added sample Ag. C. After removing unbound materials, the Abs bound to immobilized Ag are detected by adding peroxidase labeled 2nd Ab. D. After removing excess reagents, peroxidase substrate is added and color is developed.

23

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. or standard solutions along with a known amount of the antibodies are added. Free

antigen competes with immobilized antigen for antibodies. When the free antigen

concentration is higher, less Ab will bind to immobilized antigen. After washing away

the unbound materials, all remaining antibodies are detected by adding enzyme labeled

2nd Ab (e.g. goat anti-rabbit IgG and goat anti-mouse IgG+IgM, conjugated to enzyme).

After removing the excess reagents, enzyme substrate is added, and color is measured.

The amount of color produced is inversely proportional to amount of antigen from the

sample or standard solution. The color measured is greatest, when free antigen is 0. In

this case, the assay is called Ab capture indirect non-competitive ELISA.

1.3.2: Antibody structure

Antibodies are globular proteins commonly referred to as immunoglobins (Ig).

Structurally, antibodies are composed of one or more copies of a characteristic unit that

can be visualized as a Y shape. The unit consists of four polypeptide chains: two

identical heavy chains and two identical light chains (Figure 4). Each arm containing a

heavy chain and light chain, contains a site that is responsible for antigen binding. The

variable regions of the amino acid sequence in this portion provide the basis for the Ab-

Ag binding and are responsible for Ab specificity (Rittenburg, 1990).

Antibodies can be divided into five classes, IgA, IgG, IgE, IgM, and IgD based

on their structures. Among the immunoglobins above, IgM is the first antibody

produced when an animal is exposed to an immunogen and it exists as a pentamer in

serum. IgG is a monomer that is normally produced later in the immune response, it

makes up about 89% of the total Ig in the serum, and it is the most common type of

antibody employed in immunoassay (Rittenburg, 1990).

24

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure Figure 4. Antibody basic unit structure

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1.3.3: Antibody-antigen (Ab-Ag) interactions

The interaction between Ab and Ag is noncovalent and reversible. The binding

includes hydrogen binding, electrostatic, Van Der Walls and hydrophobic interactions.

The binding of antibody to antigen follows basic thermodynamic principles of any

reversible biomolecular interaction. The equilibrium equation can be defined as:

_ [Ab- Ag]

eq ‘ [Ab]\Ag]

Where Keq is the equilibrium constant; [Ab], [Ag], and [Ab-Ag] are the molar

concentrations of the free Ab, free Ag and Ab-Ag complex in the equilibrium status,

respectively. For the reversible equilibrium reaction, temperature, pH, and buffers

affect the affinity constant, Keq. The time taken to reach equilibrium depends on the

diffusion rate.

1.3.4: Polyclonal antibody (PAb) and monoclonal antibody (MAb) production

When higher vertebrates are exposed to foreign materials, antibodies are elicited

as part of their immune response. The immune response involves many interactions of

a large numbers of specialized cells: macrophages, T lymphocytes and B-lymphocytes

(Harlow and Lane, 1988b). When an animal is exposed to an immunogen with an

appropriate immunization schedule, B cells will be activated and will proliferate and

differentiate into plasma and memory cells. Plasma cells produce large amounts of

antibody. Memory cells are long-lived cells that remain in circulation and are primed

upon subsequent exposures to the antigen. The differentiation of B cells leads to the

production of higher affinity Ab, when the animal is exposed to the antigen again.

26

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Considerations need to be made before immunization, such as choice of animal,

form of immunogen, type of adjuvant, dose and boosts. Gossypol (MW 518) is not

large enough to elicit Ab, so it must first be conjugated to a carrier protein to become an

immunogen. The ability of the immunogen to elicit the desired immune response

depends on the amount of hapten as well as its orientation. The presence of spacer alkyl

chains, such as e-amino group of lysine induces a greater immune response. Too long

or too short spacer arms would limit the exposure of the analyte for antibody production

and decrease the assay sensitivity (Chen et al., 1995). The purified immunogen

combined with an immune stimulant (adjuvant) can enhance the immune response. The

application of adjuvants could slow down the release of immunogen, give rise to a high

antibody response and induce an increased circulation of lymphocytes to allow greater

contact between antigen and antigen-reactive cells to increase the antibody production

(Harlow and Lane, 1988a). Boosts can improve the maturation and differentiation of B

cells to produce high affinity antibodies.

If blood from an immunized animal is the source of Ab that contains a

heterogeneous population of Ab, this is referred to as polyclonal antibodies (PAb).

Each antibody population differs in both its affinity and its specificity from others,

because each population represents the secreted products from a single stimulated

lymphocyte and its clonal progeny (Hum and Chantler, 1980). Usually rabbits are the

first choice for PAb production, since they are cheap, easy to care for, robust, and easy

to bleed. Figure 5 shows the steps involved to produce PAb by immunizing a rabbit.

Compared to PAb, MAb are more specific, because they are derived from a

cloned lymphocyte cell. MAb can be produced after immunizing mice with

27

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Immunogen

I

H Immunization

I

Serum collection (jl I II )

Figure 5. Diagrammatic representation of the production of PAb (polyclonal antibodies) from rabbits

28

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. immunogen. The immunized mouse spleen has a high density of B cells. Each B-cell

clone contains the genes to produce one type of antibody. The fused cells between

myeloma cells and splenocytes, called hybridoma cells, have the characteristics of both.

After fusion, the hypoxanthine-aminopterin-thymidine (HAT) system is usually used to

select the hybridomas (Kohler and Milstein, 1975). In this system, aminopterin (A) is

used to block the main biosynthetic pathway for nucleic acid production. The mutant

myelomas, lacking the enzyme hypoxanthine guanine phosphoribosyle transferase

(HGPRT), will multiply in the absence of aminopterin, but die out in HAT medium.

Normal cells can continue to synthesize nucleic acids using the salvage pathway when

hypoxanthine and thymidine are provided. Spleen cells, being primary cells, however,

will not grow very long in culture without special stimuli and conditions. Fused

hybridomas can grow in HAT medium. Selected hybridomas are screened for the

production of Ab with desired characteristics and positive cells are cloned. Each

positive cell line may have different specificity, so MAb with different specificities can

be obtained. The Figure 6 shows the basic steps for MAb production and Figure 7

shows the details of HAT selection system.

There are two different ways to produce large amounts of MAb from a secreting

hybridoma line: by growing the cell line in culture or by growing it in animal as a tumor

(Zola, 1987b). For the first method, the antibody is produced by maintaining the

hybridomas in culture and splitting the culture every 2 to 4 days. Alternatively, the

hybridoma is injected into intraperitoneal of an appropriate animal and grows as a

transplantable myeloma, secreting large amounts of immunoglobulin in peritoneal

cavity as ascitic fluid. The ascitic fluid is harvested as the source of MAb. In general,

29

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Immunization

£ a Splenocytes in spleen

•• Myeloma

Cell fusion and HAT selection i

fvhybridoma i ran screening i

t cloning

Ab, Ab, Ab„

Figure 6 . Diagrammatic representation of the production of MAb (monoclonal antibodies) from mice

30

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Salvage Salvage De novo nonviable (in tissue culture) HAT selection Salvage hybridoma Cell fusion De novo myeloma B cell nonviable viable Figure 7. The HAT (hypoxanthine-aminopterin-thymidine) selection system (Adapted from Harlow and Lane, 1988)

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. tissue culture supernatant is not contaminated by other antibodies, but can only be

produced at relatively low concentrations. Supernatants can be used for most serology.

Ascites produced by animals may be contaminated with other immunoglobulins made

by the host, but can be produced in higher quantities. Ascitic fluid is usually used for

immunochemical characterization or isolation of antigens. This is common method of

commercial MAb production.

One of the principal advantages of a hybridoma culture is that it provides a

potentially permanent source of Ab. Once a cell line of interest is obtained, the cells

can be preserved indefinitely using cryopreservation. The temperature of liquid

nitrogen is -196°C. In order to reduce damage caused by the formation of ice crystals,

cells are slowly cooled down and a cryoprotective medium (for example,

dimethylsulfoxide in serum) is used.

MAb used in immunoassay have some advantages over PAb: their specificity of

binding, their homogeneity, and their ability to be produced in unlimited quantities.

One unique advantage of hybridoma production is that impure antigens can be used to

produce specific antibodies. Because hybridomas are single-cell cloned prior to use,

monospecific antibodies can be produced after immunizations with complex mixtures of

antigens.

1.4: Research objectives

The reactivity of gossypol has made study and understanding of its nature

difficult. Besides natural (+)- and (-)-isomers, there are various forms of bound

gossypol. Different forms of gossypol with different stabilities and biological activities

lead to the inconsistant results between chemical analyses and animal availability and

32

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. bioactivity studies. We are proposing to develop and apply immunochemical methods

for gossypol anlalysis. Because of the selectivity and specificity of Ab-Ag interaction,

immunochemical methods may provide a potential tool to study gossypol forms and

their availabilities. The specific objectives were to:

1. Produce protein-gossypol conjugates to immunize rabbits and mice for the

production of polyclonal and monoclonal anti-gossypol antibodies.

2. Use characterized antibodies to study the chemistry of gossypol interactions with

amino and amine compounds in model systems, and improve our knowledge of the

forms and bioactivity of bound gossypol.

3. Adapt and optimize the above antibodies in an immunoassay to rapidly and

accurately measure the quantity and/or quality of gossypol in conttonseed products.

33

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CHAPTER 2

MATERIALS AND METHODS

2.1: Materials

Bovine serum albumin (BSA), Limulus polyphemus hemolymph (LPH),

, ammonium carbonate, ammonium chloride, trypan blue, sodium

pyruvate (100 mM), sterile cell culture penicillin-streptomycin (with 10,000 units

penicillin and 1 0 mg streptomycin per ml), sterile fetal calf serum, oxalic acid, 2 ,2 ’-

azino-bis (3-ethylbenthiazoline-6-sulfonic acid) (ABTS), Polyoxyethylene-20-

sorbitanmonolaurate (Tween 20), N,N-dimethylformamide, ammonium sulfate,

phosphate (monobasic), ethanolamine, L-2-amino-l-propanol, 3-amino-1-

propanol, gossypol, L-glutamine, L-(-)-tryptophan, L-tyrosine, para-amino-benzoic

acid, sterile dimethylsulfoxide (DMSO), cellulose dialysis tubing sacks (MWCO

12,000), agarose (type VII: low gelling temperature), goat anti-mouse IgG+IgM (H+L)

peroxidase conjugated, Rosewell Park Memorial Institute 1640 (RPMI 1640) and

AvidChrom-Protein A Antibody Purification Kit (with HEPES buffer, generated buffer,

serum-binding buffer and elution buffer) were purchased from Sigma Chemical Co. (St.

Louis, MO). Citric acid (monohydrate granular) was from Mallinckrodt Inc. (Paris,

KY). L-(+)-lysine and L-(+)-arginine were purchased from Acros Organics (Geel,

Beljium, NJ). Sodium cyanoborohydride was from Aldrich Chemical Company, Inc.

(Milwaukee, WI). Goat anti-rabbit IgG (H+L) peroxidase conjugated, Freund’s

incomplete adjuvant and Freund’s complete adjuvant were purchased from Calbiochem-

Novabiochem Corp. (La Jolla, CA). The Ribi adjuvant for mouse immunization was

34

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. purchased from Ribi Immunochem Research, Inc. (Hamilton, MT). Polyethylene glycol

1500 (PEG-1500) was purchased from Boehringer Mannheim GmbH, Germany. HT

and HAT were bought from Gibco BRL (Grand Island, NY) as lyophilized

reconstitutable as lOOx sterile solutions (10 mM sodium hypoxanthine and 1.6 mM

thymidine and with 40 pM aminopterin in HAT only). Acepromazine maleate, atropine

sulfate SA, xylazine and ketamine were provided by Animal Care Facility at the

Louisiana State University. The myeloma cell line (NS-1) was a non-secreting clone of

P3x63Ag8 (American Type Culture Collection, Rockvillie, MD).

Single frosted autoclaved microscope slides (25x75xlmm) were purchased from

Sigma Chemical Co. (St. Louis, MO). Disposable microcapillary pipets (25 (al) were

purchased from Kimble (Toledo, Ohio). Flexible polyvinylchloride (PVC) microtiter

plates were from Dynatech Laboratories Inc. (Alexandria, VA). Dynex Immulon® 1 B,

Immulon® 2 HB and Immulon® 4 HBX plates were obtained from Dynex Technologies,

Inc. (Chantilly, VA). Whatman No.l and No.2 filter papers were bought from Whatman

International Ltd. (Maidston, England). Needles (25G5/8, 18G11/2, 20G11/2) were

from Becton Dickinson Labware (Franklin Lakes, NJ). Cottonseed product samples

were gifts from M. Calhoun and sample ID number (2241, 2817, 2822, 2819, 2820,

2814 and 2813) assigned in the Nutrition/Toxicology Laboratory at the Texas A&M

University Agricultural Center. Free Gossypol and Total Gossypol were previously

measured using AOCS official methods, and (+)- and (-)-gossypoI isomers were

measured using HPLC by M. Calhoun (Texas A&M University Agricultural Center,

San Angelo, TX). Two New Zealand white (NZW) rabbits and five mice (BALB/c)

were housed and cared by the Animal Care Facility at the Louisiana State University.

35

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Sterile plasticware used for fusion, cloning and tissue culture included 96-well

and 24-well covered tissue culture plates (Costar, Cambridge, MA), 60x15mm and

100x20mm tissue culture dishes (Becton Dickinson Labware, Franklin Lakes, NJ), 15

ml and 50 ml disposable polypropylene centrifuge tubes, and 200pl universal pipet tips

(Coming Inc., Coming, NY), 1ml pipet tips (dot Scientific Inc., Burton, MI), sterile

solution basins (Labcor Products, Inc., Frederick, MD), 2.5ml and 12.5 ml eppendorf

tips (Brinkman Instrument Inc., Westury, NY), 0.22 pm and 0.45 pm disposable

filterwares (Nalge Company, Rochester, NY), and Millex-GS 0.22 pm flter unit (Sigma

Co., St. Louis, MO).

2.2: Methods

2.2.1: Instrumentation

The shaker for ELISA was Bamstead/Thermolyne, Maxi-Mix III™ (Dubyque,

Iowa). Cottonseed product samples were ground in a coffee mill (BRAUN, model

KSM2, Mexico). Mixing usually was performed on the vortex (Scientific Industries,

Inc., Bohemia, NY). Pre-coated thin layer chromatography (TLC) plastic sheets PEI-

Cellulose (MC/B Manufacturing Chemists, Inc., Cincinnati, Oh) were used in

monitoring the reactions of gossypol derivatization. TLC solvent system was the

mixture of ethyl-acetate: 2-propanol: H 2O (65: 24: 11 v/v) and the spots were visualized

under BLAK-Ray Lamp (Model UVL-56, Upland, CA). Cells were grown in a water-

jacketed CO2 incubator (Forma Scientific, Inc., Maijetta, Ohio) and viewed using an

inverted microscope (Nikon, model TMS, Japan). Cell counting was performed on a

Neubauer counting champer (Fisher). Tissue culturing operations were done in a

biological safety cabinet from ENV Service, Inc. (Minneapolis, MN). Clone selection

36

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. from soft agar was performed in a laminar flow hood. All autoclaving was performed at

121°C for 15 min. Matrix-assisted laser desorption/ionization-time of flight-mass

spectrometry (MALDI-TOF-MS) was used to measure the number of gossypol groups

in protein gossypol conjugates.

2.2.2: Modeling and statistical analysis

Enzyme-linked immunosorbent assays (ELISAs) were carried out in 96-well

plates and read at wavelength 405 nm in a SPECTRAmax Plus microtiter plate reader

(Molecular Devices Corporation, Sunnyvale, CA). The software package Softmax

(Molecular Devices) was used for fitting the sigmoidal standard curve based on a four-

parameter logistic method of Rodbard (1981) as an empirical way to describe the dose

response curve in ELISA. The parameter a represents the response at zero concentration

of gossypol. The d is the response at “infinite” concentration of gossypol. The

parameter c is the gossypol concentration giving 50% reduction (halfway between a and

d, called I50 value). The parameter b is the curvature parameter which determines the

steepness of the curve. Those four parameters a, b, c and d are obtained by the method

of least squares of observed data using the software package Softmax (Molecular

Devices). X in the equation is the gossypol concentration, and Y is the corresponding

absorbance unit. After calibration process, the curve is used to determine the

concentrations of the unknowns.

a - d y = ------1 +(x/cf

Graphs were plotted as Log 10 of equivalent gossypol concentration vs. relative

absorbance (A/Ao). Where A is the corresponding absorbance at certain gossypol (or

37

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. gossypol-derivative) concentration; Ao is absorbance when gossypol (gossypol-

derivative) concentration is 0 .

Four parameter sigmoidal equation was used to calculate I 50 values based on the

least square errors of the observed data. Comparison of I 5 0 values for different gossypol

derivatives were made using Tukey’s Studentized Range Test with significance level of

95%.

2.2.3: Preparation of experimental solutions

Phosphate buffered saline (PBS) solution was prepared by dissolving 18 g NaCl,

2.22 g disodium hydrogen phosphate, and 0.6 g potassium dihydrogen phosphate in 1.9

L of distilled water, and adjusting the pH to 7.3 with IN NaOH, then bringing the

volume to 2.0 L. For phosphate buffered saline-Tween 20 (PBST) solution, Tween 20

(1.0 g) was added into 2 L PBS solution. NH 4 CI solution (0.8% NH 4 CI) was made by

adding 0.8 g of NH 4 CI into 100 ml of distilled water and was autoclaved. Trypan blue

(0.4%) stain solution was prepared by adding 0.4 g of trypan into 100 ml of PBS

solution. ABTS substrate solution was made by adding 10 mg 2,2’-azino-bis(3-

ethylbenthiazoline- 6 -sulfonic acid) (ABTS), 8 pi hydrogen peroxide (30%) solution in

24 ml of 0.1 M citrate buffer (pH 3.8). BSA solution (0.33%) was made by adding

BSA 200 mg into 60 ml of PBS. Dimethylsulfoxide (DMSO) solution ( 8 %) was

prepared by mixing 8 ml of DMSO and 92 ml of sterile fetal calf serum aseptically.

Sterile L-glutamine solution (200 mM) was prepared by adding 3.00g of L-glutamine in

100 ml of PBS and filtered through 0.22 irni of filter kit.

The following media were prepared for the culture of myelomas and

hybridomas. RPMI-1640 medium was prepared by dissolving RPMI 1640 powder

38

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (1.04%) and sodium bicarbonate (0.2%) in distilled water and filtered through 0.45 pm

filter kit in biological safety cabinet. Serum-free RPMI consisted of RPMI 1640

medium with the addition of L-glutamine (2 mM), sodium pyruvate (1 mM) and

penicillin-streptomycin ( 1 0 0 units penicillin and 0 .1 mg streptomycin per ml).

Complete RPMI-20 was made by adding 20% (v/v) calf serum in serum-free RPMI and

complete RPMI-10 was made by adding 10% of calf serum to serum-free RPMI. HT or

HAT medium was complete PRMI-20 with addition of HT (100 p.M sodium

hypoxanthine and 16 pM of thymidine) or HAT (HT with 0.4 pM aminopterin). Spent

medium was prepared by growing myeloma cells in complete RPMI-10 for 2-4 days,

centrifuging the cell suspension at 250 x g for 5 min and passing the supernatant

through 0.45 pm filter kit. Conditioned medium was prepared with spent medium plus

20% fetal calf serum, L-glutamine (2 mM), sodium pyruvate (1 mM) and penicillin-

streptomycin ( 1 0 0 units penicillin and 0 .1 mg streptomycin per ml).

2.2.4: Preparation of Immunogen

A scheme for the production of LPH-G conjugates (Limulus polyphemus

hemolymph-gossypol) via a Schiff base intermediate is shown in Figure 8 . Two LPH-G

conjugates were produced as follows:

LPH-Ga: Gossypol (23.5 mg) was dissolved in 6 ml methanol. LPH (24.5 mg)

was dissolved in 6 ml PBS buffer, then the two solutions were mixed and reacted with

continuous stirring for 48 h at 5°C in the dark. The whole mixture was filtered through a

Whatman No. 1 filter paper and washed using ethyl ether to remove unreacted gossypol.

The product collected on the filter was air dried and stored at 5°C with desiccation.

39

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CD C

*c a o erf u V) Z um as CQ« 2 Crf

JSo i VI

erf I _c 2fS ‘S X oUi a. . . Gossypol-protein conjugate formation via a Schiff base intermediate 8 Figure Figure

o a / to>■» Oc o \ a

40

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. LPH-G8: Gossypol (24.5 mg) was dissolved in 7 ml methanol and mixed with

24.5 mg LPH dissolved in 25 ml PBS and reacted with continuous stirring for 48 h at

RT under mild nitrogen flow with adding 105.6 mg of sodium cyanoborohydride. After

the reaction, the whole mixture was dialyzed in 1 L of 8 M urea for 24 h, 4 L of 50 mM

ammonium carbonate for 24 h and finally 4 L of 25 mM of ammonium carbonate for 24

h, respectively, then the product was lyophilized.

2.2.5: Preparation of solid-phase conjugates

BSA-G conjugates (Bovine serum albumin-gossypol) were produced via Schiff

bases as follows:

a. Gossypol (7.8 mg) was dissolved in 2 ml of ethanol. BSA (50 mg) was

dissolved in 25 ml PBS. These two solutions were mixed with 90 mg of sodium

cyanoborohydride (NaBHjCN) and reacted for 48 h in the dark under N 2 at RT, then

dialyzed in same way as described for immunogen LPH-G8.

b. Gossypol samples (3.9, 7.8, 15.6 mg) were each dissolved in 2 ml methanol.

Each was mixed with 15 ml of BSA solution (containing 50 mg BSA) plus 60 mg

NaBHaCN. The reaction conditions and purification were performed in same way as

(a).

To observe the effect of reducing agent, controls were also prepared without the

addition of NaBHsCN.

2.2.6: Measurement of gossypol groups in conjugates

The number of gossypol groups in protein gossypol conjugates were measured

using Matrix-assisted laser desorption/ionization-time of flight-mass spectrometry

(MALDI-TOF-MS). All samples were prepared for MALDI analysis by mixing 2,5-

41

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. dihydroxybenzoic acid (2,5-DHB) matrix solution with dried BSA-G to give mole ratio

104:1 — 103 :1. The analyte-matrix mixture was applied to the sample probe and was air-

dried. A linear, time lag focusing TOF mass spectrometer was used to collect all mass

spectra at averaging of 290(350) laser shots.

2.2.7: Polyclonal antibody (PAb) production

2.2.7.1: Rabbit immunization

Two female NZW rabbits (#16 and #15) were immunized with 1.0 mg of

conjugate (LPH-GA and LPH-G8, respectively) in 2 ml of PBS/Freund’s complete

adjuvant (1:1, v/v). Rabbit #16 was first immunized at age 6 months and boosts were

given monthly. Rabbit #15 was immunized beginning at age 9 months and boosts were

given on months 10, 15 and 17 using the same concentration of conjugate in Freund’s

incomplete ajuvant. Blood samples were taken 2 weeks after each boost and transferred

to sterile vacutainers, allowed to clot half an hour at room temperature, centrifuged at

16,000 x g for 10 minutes. The sera were tested for anti-gossypol antibodies and stored

frozen. Preimmune sera were used as negative control.

Two weeks after the last immunization, the rabbits were euthanized by injection

with atropine sulfate SA (0.05 ml/kg, sc.) and acepromazine maleate (0.5 ml/kg, sc.).

After 10 min, the rabbits were injected with xylazine (5 ml/kg, sc.) and ketamine (50

mg/kg, sc.), then the rabbits were bled using cardiac puncture and the sera collected and

stored as described above.

2.2.7.2: Antibody capture noncompetitive ELISA for screening sera

An antibody-capture noncompetitive ELISA (ACN-ELISA) was used to monitor

the presence of anti-gossypol antibodies in rabbit sera. BSA-G in PBS (10 pg/ml) was

42

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. prepared and a 100 pi aliquot was added to each well of an Immulon 2 HB microtiter

plate and incubated at 5°C overnight (coating). The solution was removed by inverting

the plate with a quick flick of the wrist, and each well was filled with 2 0 0 pi /well of

1% BSA in PBS (blocking). Following an incubation of 30 min at 37°C, the solution

was removed and wells were washed with 3 x 200 pi of PBST, incubating each wash

for 5 min at RT with shaking. An aliquot of 10% methanol in PBS (50 pl/well) was

added coupled with 50 pl/well of diluted antisera or preimmune sera (1st Ab: diluted

1/1000 with 1% BSA in PBST) and incubated for 30 min at 37°C. After washing away

unbound reagents as described above, the bound antibodies were detected by the

addition of 100 pl/well of 1/10,000 diluted goat anti-rabbit IgG peroxidase conjugated

(2nd Ab) in 1%BSA in PBST, incubated for 30 min at 37°C and then washed with 3 x

200 pi of PBST as described as above. ABTS substrate was added (100 pi /well) and

peroxidase activity was measured as absorbance at 405 run after 30 min of incubation at

RT.

2.2.7.3: Optimization of ELISA

Immulon IB, Immulon 2 HB, immulon 4 HBX and PVC microtiter plates were

compared for their abilities to immobilize solid phase conjugate (coating) in ACN-

ELISA. The blocking buffers (1% BSA, 1% gelatin, 1% ovalbumin and 1% powder

milk), 1st Ab and 2nd Ab dilution buffers (1%, 0.5%, 0.1% or 0.05% BSA in PBST) and

incubation time (30 min, 1 h and 2 h) and temperature (37°C and RT) were investigated

for their effects on ELISA. Acetone and DMF at different concentrations and 10%

methanol incubated with diluted PAb were compared for their effects on ACN-ELISA.

43

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Checkerboard ELISA was used to optimize the concentrations of BSA-G and

antisera from rabbit #16 and #15. A two dimensional titer determination was performed

on the plate with decreasing concentration of coating solution BSA-G rowwise, ranging

from 0.01 pg/ml to 100 pg/ml from top to bottom overnight at 5°C (coating). After

removing the coating solution by inverting the plate, blocking solution 200 n 1/well of

1% BSA in PBS was added. After 1 h incubation at RT (2 h @ RT for rabbit #15 PAb),

the solution was removed and washed with 3 x 200 pi PBST. Fifty pi of 10% methanol

and serially diluted antisera (diluted 1/500 to 1/8000 in 0.5% BSA in PBST, 50 ^1/well)

were added respectively from left to right across columns (non-competitive step).

Following lh incubation at RT (2 h @ RT for rabbit #15 PAb), the unbound materials

were washed away with 3 x 200 |d PBST, and 100 pl/well of 2nd Ab (goat anti-rabbit

IgG peroxidase conjugated) at 1/10,000 dilution in 0.5% BSA was added. After 1 h

incubation at RT (2 h @ RT for rabbit #15 PAb), the excess reagents were removed and

washed with 3 x 200 pi PBST, and 100 pi/ well of ABTS substrate was added. The

absorbance at 405nm was measured after 30 min incubation at RT. The optimum

combination of antiserum and coating solution were chosen both dilution curves were

steep and gave an absorbance between 1 to 2.

2.2.8: Monoclonal antibody (MAb) production

2.2.8.1: Immunization of mice and serum test

Preimmune blood samples (retro-orbital) were taken from five mice (one month

old, female, BALB/c). Mouse #0, #1 and #2 were immunized with 1.0 mg of LPH-GA

conjugate in 2 ml of PBS/Ribi Adjuvant System (1/1, v/v), and mouse #3 and #4 were

immunined with LPH-G8 in the same way. Injections were performed as 0.1 ml

44

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. subcutaneous and 0.1 ml intraperitoneal. Boosts were given in a similar manner on 28,

49, 70 and 91 days after the first immunization. Blood samples were taken 1 week after

each boost and transferred to sterile vacutainers, allowed to clot half an hour at RT,

centrifuged at 16,000 x g for 10 minutes. The sera were collected and tested for anti-

gossypol antibodies using ACN-ELISA.

Microtiter plate was coated (100 pi of 5 pg/ml BSA-G), blocked (200 pi of 1%

BSA) and washed (3 x 200 pi of PBST) as described as above. Fifty pi of 10% of

methanol in PBS and 50 pi diluted serum (diluted 1/1000 in 0.5% BSA in PBST) were

added to microtiter plate. Following 2 h incubation at RT, the unbound material was

removed and washed with 3 x 200 pi in PBST, and 100 pi of anti-mouse IgG+IgM

peroxidase conjugated at 1/3,000 dilution in 0.5% BSA was added. After 2 h

incubation at RT, excess reagent was washed away and 100 pi of ABTS substrate was

added. The absorbance was measured at 405 nm after 30 min incubation at RT.

2.2.8.2: Myeloma cell culture

Myeloma cells (NS-1) were cultured in sterile tissue culture dishes using

complete RPMI-10 at 37°C with 5% CO 2 and at 90-100% relative humidity. Cells were

subcultured every 2-3 days, using 1/5 to 1/20 dilution to keep cell density from 105 to

106 cells/ml. Cells were used for fusion two days after subculturing. After

centrifugation at 250 x g for 5 min, the cells were suspended in 15 ml of RPMI medium,

and centrifuged again. Washing was performed twice, then cells were combined and

resuspended in 2.5 ml of RPMI medium and kept on ice until fusion. Meanwhile, the

cell suspension was counted at 1/10 dilution in 0.25% trypan blue in PBS, using a

Neubauer counting chamber.

45

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2.2.8.3: Recovery of splenocytes

After four immunizations, the mouse #2, exhibiting the highest titer of anti-

gossypol antibodies, was immunized again. Three days later, this mouse was sacrificed

by cervical dislocation, the spleen was aseptically removed, mashed with two single

frosted autoclaved microscope slides, and resuspended in a tissue culture dish

containing 10 ml RPMI medium, and transferred to a centrifuge tube. Cells were

centrifuged at 250 x g for 5 min and washed twice with 10 ml PRMI. After the second

centrifuge, the cells were resuspended in 4 ml of 0.8% NH4CI for 3 min to lyse the red

cells, then centrifuged at 250 x g for 5 min. After discarding the upper-layer, the cells

were resuspended in 7 ml of RPMI medium, counted and immediately used for fusion.

2.2.8.4: Production of hybridomas

Myeloma (10 7) cells were combined with spleen cells (108). The mixture was

mixed on the vortex at low speed, centrifuged at 800 x g for 5 min and the supernatant

was discarded. One ml of 50 % PEG 1500 in HEPES buffer was added to the cells with

gentle mixing over a minute, then mixed on vortex for a few second, and left for another

minute. One ml of RPMI was slowly added and following adding another 9 ml of

RPMI in next 3 min, then the cells were centrifuged. After discarding the supernatant,

the cells were resuspended in 90 ml of HAT medium, and 150 pi of aliquot was added

into each well of 7 sterile 96-well tissue culture plates with doing a few control using

myeloma cells in each plate.

2.2.8.5: Culture, screening, and selection of hybridomas

Fused cells were incubated at 37°C, 5% CO 2 with 90-100% relative humidity.

Seven days after fusion, the cells were fed 100 pi of HT medium per well. Hybridomas

46

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. were checked daily after feeding, looking for colonies and yellowing of the medium.

When the medium became yellow and the colonies were visible over 30% of the base of

the individual wells, the supernatants from these wells were screened for Ab production

using ACN-ELISA and ACC-ELISA.

Fifty pi of 10% methanol (for ACN-ELISA) or gossypo 1-derivatives (for ACC-

ELISA) (100 to 250 ng/ml of gossypol equivalents) was incubated with 50 pi of each

supernatant on microtiter plates that were previously coated (100 pi of 5 pg/ml of BSA-

G), blocked (200 pi of 1% BSA) and washed (3 x 200 pi of PBST). The tests were

performed as same as described in section 2.2.8.1 for mouse PAb test.

Cells with supernatants that showing positive in both assays were transferred

into 24-well microtiter plates and fed with about 0.5 ml of complete RPMIR-20 to each

well. After three days, the cells were transferred into 65x10 mm tissue culture dishes

and cultured for 2-4 days, then transferred into 100x20 mm tissue culture dishes.

2.2.8.6: Cell cloning

Once the colonies have been identified as producing anti-gossypol antibodies,

the positive cells were cloned using soft agar and limiting dilution methods.

Soft agar method: A 2.4% agarose medium was prepared by weighing 2.4 g

agarose in 100 ml of 0.15 M NaCl solution, heating to 70°C until dissolved. The clear

solution was distributed into 10 scintillation vials, capped, autoclaved and stored at 5°C

until required.

On the day of cloning, the agarose medium was melt at 37°C, and 2 ml was

transferred into 10 ml of pre-warmed (37°C) conditioned medium to make a 0.48%

47

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. agarose solution (called soft agar). The soft agar was dispensed into 6-polypropylene

tubes with 2 ml for each tube.

One hundred pi of serial uncloned cell dilutions (1/2) in conditioned medium

was added into each of above polypropylene tubes. They were thoroughly mixed on a

vortex mixer and dispensed (0.5 ml /well) into a 24-well microtiter plate with different

dilution for each column. Cells in soft agar were incubated at 37°C with 5 % CO 2 and

90-100% humidity.

After 8-12 days growth, clones were selected from soft agar under an inverted

microscope placed in a laminar flow hood. A mouth-pipeting system was employed by

connecting a sterile 25 pi disposable micropipet, pipet/hose adapter, sterile tubing, two

0.22 pm filter units and a mouth piece (Plhak and Spoms, 1994). Selected cells were

transferred into 200 pi complete RPMI-20 medium in a sterile 96-well tissue culture

plate. They were incubated at 37°C with 5% CO 2 and 90-100% humidity and

subcultured every 2-4 days to keep cell density from 104 to 106 cells/ml (Hum and

Chantler, 1980). Supernatants were screened and positive cells were selected for further

recloning.

Further cloning was performed using a limiting dilution method. Positive cells

were expanded gradually, through 2- and 10-ml culture. Then the cells were diluted

with conditioned medium and plated into 96 well tissue culture plate at 10 cells/well (2

rows), 3 cells/well (3 rows) and 1 cell/well (3 rows). After 9-15 days, the visible

colonies were tested for antibody production. The positive cells were selected from the

highest dilution showing cell growth and cultured.

48

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The cloning procedure was performed three or four times using either soft agar

or limiting dilution methods or both to ensure stability of the cell lines.

2.2.8.7: In vitro production of monoclonal antibodies (MAb)

Antibody was produced by maintaining the hybridomas in complete RPMI-20

and splitting the culture every 2 to 3 days. The supernatant was collected for the assays.

2.2.8.8: Purification of monoclonal antibodies (MAb)

The tissue culture cell suspensions were collected and centrifuged at 250 x g for

5 min to remove the hybridomas. The supernatant from each cell line was transferred

into a beaker and was kept in a cold water bath (0°C). Ammonium sulfate (35 g/100 ml

supernatant) was then slowly (10-15 min) added to obtain 55% saturation with regular

stirring and left to stir for another 30 min. The mixture was centrifuged at 9,000 x g at

2°C for 15 min, and the supernatant was discarded and the protein precipitated was

resuspended in about 2-4 pellet volumes of PBS buffer. This solution was dialyzed

against 3x1500 ml of PBS buffer for 12 h to remove ammonium sulfate. The dialyzed

solution was stored at -20°C.

2.2.8.9: Freezing, storage and thawing of cell line

Healthy cells, approximately 106 to 107 cells, were centrifuged at 250 x g for 5

min, resuspended in 1 ml of 8% dimethylsulfoxide in calf serum and 0.5 ml aliquots

were placed into cryogenic vials. These vials were wrapped in several layers of paper,

and placed into -80°C of freezer for two months, then transferred into a liquid N 2 (-

196°C).

Cells were thawed quickly after removal from liquid tank N 2 in 37°C water until

the ice has just melted. The cells were asceptically transferred into 10 ml of RPMI-20

49

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. and mixed thoroughly, then centrifuged at 250 x g for 5 min. Washing was performed

twice to remove DMSO. After the final centrifugation, cells were resuspended in

complete RPMI-20 medium, and cultured.

2.2.9: Crystallization of isomeric gossypol-acetic acid

Crystals of gossypol-acetic acid (Dowd et al., 1998) were formed from acetone

by dissolving gossypol acetic acid into acetone at 1:2.6 (w/w) ratio and crystallization

was performed by storing the solution at -20°C to perform primary nuclei. After

melting most of the precipitated material by mild heating, the crystallization was done

in 4°C.

When crystals grew big enough, they were washed with 10% and 20% acetone

in hexane solutions (v/v) to remove the precipitated gossypol, and each crystal was

placed in a different vial. Isolated crystals were dissolved in acetone, and a small

portion was tested using HPLC after formation of diastereoisomer with (+)- 2-amino-1-

propanol in DMF for 30 min at 95°C. HPLC (Waters 2690, separations module),

performed using a reversed-phase C l8 column (mobile phase: 80% acetonitrile and

20% phosphate buffer pH=3.0) with UV detection at 254 nm, was conducted at USDA,

Southern Regional Research center, New Orleans, LA. After determination, the (+)-

gossypol or (-)-gossypol isomers were combined into 2 separate vials, respectively. The

final products were reanalyzed using HPLC, vacuum dried and stored at 5°C under

desiccation.

2.2.10: Preparation of various forms of gossypol

Gossypol solution and different gossypol derivatives with amino acids and

amine compounds were made as follows:

50

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Derivatizing solutions (0.1M) of L-(+)-Iysine, L-(+)-arginine, ethanolamine, L-

2-amino-l-propanol, 3-amino-1-propanol, para-amino-benzoic acid, L-glutamine, L-(-)-

tryptophan and L-tyrosine were prepared in PBS. Gossypol (2.5 mg/ml) was dissolved

in methanol. Gossypol derivatives were made by mixing 1 part of gossypol solution

and 9 parts of each derivatizing solution, adding 0.3% sodium cyanoborohydride for 2 h

at RT. The reaction was followed by TLC and the absorption spectra of products were

also measured. (+)- and (-)-gossypol isomer derivatives were prepared with L-(+)-

lysine and L-2-amino-l-propanol, respectively, as described above. Gossypol solution

(250 ng/ml) was prepared by diluting 2.5 mg/ml of gossypol in PBS.

2.2.11: Antibody capture competitive ELISA

An antibody capture competitive ELISA (ACC-ELISA) was used for evaluating

the specificity of antibodies to racemic gossypol derivatives, racemic gossypol at

different storage time, and (+)- and (-)-gossypol derivatives.

BSA-G in PBS (5 pg/ml) was coated on the microtiter plate, blocked and

washed as described previously.

For rabbit #15 PAb, 50 pi of serially diluted (1/10) gossypol derivative solutions

with 50 pi diluted serum (diluted 1/1000 with 0.5% BSA in PBST) were added from the

top to bottom row on the plate. The following procedures were same as section 2.2.7.2

for checkerboard titration.

For MAb, 50 pi of serially diluted gossypol derivative solutions (1/2.5 dilution

for 7G2F2G3C6, 1/10 dilution for 6G7D11F10F2F8 and 6G7D11F10F2H3) with MAb

supernatant (50 pi) were added and incubated for 2 h at RT. The following steps

followed section 2.2.8.1 for mouse serum test.

51

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Gossypol solution freshly made and the solution stored in refrigerator for 21

days were compared in ACC-ELISA as competitors for MAb (6G7D11F10F2F8 and

6G7D11F10F2H3) and PAb.

2.2.12: Gossypol analysis in cottonseed products

Cottonseed product samples were ground in a coffee mill for 40 seconds.

During grinding, the start button was released 3 times, each time for 3 seconds.

Method A:

Sample preparation:

For Free Gossypol (FG) analysis, 0.5 g ground sample was accurately weighed

into a 250 ml screw-capped volumetric flask, which had a glass bead-covered bottom.

Fifty ml of 70 % aqueous acetone was added, and the flask was mechanically shaken for

1 h. The slurry was filtered through Whatman No. 2 filter paper. The filtrate was

purged under mild nitrogen flow for 5 h to remove the acetone. Gossypol derivatization

was performed by adding 5 ml of methanol and 219 mg of L-(+)-lysine with NaBHsCN

and bringing the volume up to 50 ml with 0.1 M L-(+)-lysine. The mixture was

mechanically shaken for 2 h at RT.

For Total Gossypol (TG) analysis, a ground sample (about 0.5g) was accurately

weighted into a 50 ml stopered glass bottle. Two ml of methanol and 18 ml of 0.1 M L-

(+)-lysine with NaBHaCN were added and the bottle was vigorously shaken for 2 h at

RT.

Standard solution: Gossypol-lysine derivative was prepared by mixing L-(+)-

lysine and gossypol solution with addition of sodium cyanoborohydride (section

2.2.10).

52

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. For the determination of gossypol, an ACC-ELISA was employed using

Immulon 2 HB plates. Standards (gossypol-lysine complex) and samples were analyzed

together on the same plate. Fifty pi of gossypol-lysine (in series of dilutions from 250

pg/ml to 0.001 pg/ml in 10% methanol) or prepared samples (1/10 and 1/100 dilutions

diluted using 10% methanol) with 50 pi of diluted rabbit #15 PAb (1/1000) were added

into pre-coated (5 pg/ml of BSA-G) microtiter plates. The following procedures were

same as section 2.2.11.

Method B:

Free Gossypol (FG) analysis: FG was extracted from ground samples (about

0.5g) using 70% aqueous acetone and filtered through Whatman No. 2 filter paper and

the first 5 ml of filtrate was discarded (AOCS, Ba 7-58). Three ml of the remaining

filtrate was transferred into a 20 ml scintillation vial, and 18 ml of 0.12 M of L-(+)-

lysine with NaBHsCN was added. The reaction was performed at RT for 2 h with

vigorous shaking.

Standard solution: 32.6 mg of gossypol-acetic acid was treated the same as

described above.

For the determination of gossypol, an ACC-ELISA was employed using

Immulon 2 HB plates. Fifty pi of gossypol-lysine standard solutions (in series of

dilutions from 250 pg/ml to 0.001 pg/ml) or prepared samples (1/10 or 1/100 dilutions

diluted using 10% acetone) with 50 pi of diluted PAb (1/1000) were added into pre­

coated (5 pg/ml of BSA-G) microtiter plates. The following procedures were same as

section 2.2.11.

53

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CHAPTER 3

RESULTS AND DISCUSSION

3.1: Effects of reducing agent and pH on production of protein-gossypol conjugates

Molecules below 5,000 daltons usually are not effective immunogens

(Crowther, 1995a). Gossypol (MW 518) is not large enough to elicit a significant

immune response, therefore it must be conjugated to a carrier protein to be used as an

immunogen. The carrier protein must be of a high molecular weight (typically greater

than 20,000), and phylogenically unrelated to the animal species in which the antisera

are to be raised (Jenner and Law, 1996). For anti-gossypol antibody production, LPH

(Limulus Polyphemus hemolymph), a hemolymph protein (high MW) from horseshoe

crab was selected as a carrier protein.

In order to immobilize gossypol on the surface of microtiter plates, gossypol

was bound to another protein BSA (bovine serum albumin) to be used as immobilized

antigen (Ag) during immunoassay. BSA contains 59 lysines with 30 to 35 of these

available for derivatization with aldehydic groups (Hermanson, 1995).

Gossypol (carbonyl compounds) react very readily and reversibly with amino

groups in proteins to form Schiff bases. The reaction is pH-dependent with the greatest

reaction efficiency at pH 9-10 (Hermanson, 1995; Conkerton and Frampton, 1959).

Gossypol, however, can be rapidly attacked in alkaline solution by atmospheric oxygen

(Scheiffele and Shirley, 1964). The pH of the reaction was therefore controlled around

pH 7.3 and the products formed were stabilized with sodium cyanoborohydride, which

can convert the Schiff base to a more stable secondary amine (Hermanson, 1995).

54

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. MALDI-TOF-MS (Matrix-assisted laser desorption ionization-time of flight-

mass spectrometry) provides a great versatility in MS analyses by allowing formation of

large ionic species from thermally labile compounds. Its impressive mass range could

surpass 106 daltons (Spoms and Abell, 1996). Our results showed that LPH-G

conjugates could not be detected by using MALDI, probably because of the extremely

large aggregate (MW3,000,000) that LPH forms (Szurdoki et al., 1995). MALDI-TOF-

MS results (Figure 9, Figure 10 and Figure 11) for BSA-G indicated that the reaction

was favored in the presence of reducing agent NaBHjCN with the ratio of

gossypol/BSA in conjugates 2 times higher than that without NaB^CN (Table 2).

Table 2. Gossypol groups per BSA in BSA-G conjugates

Mole ratio used in reaction Gossypol/BSA in conjugate * (Gossypol/BSA) With NaBH3CN Without NaBH3CN

42/1 11.5 -

20/1 10.8 6.14

10/1 9.5 4.5

20/1 (ethanol) 6 3 ♦Apparent average molecular weights of BSA (bovine serum albumin) and BSA-G (bovine serum albumin-gossypol) conjugates were determined using MALDI-TOF-MS (Matrix-assisted laser desorption ionization-time of flight-mass spectrometry), and gossypol groups per BSA were determined by calculation.

In addition to confirming the production and stabilization of our gossypol-

protein conjugates, we may extrapolate these findings to ruminant vs. nonruminant

metabolism. The stabilization of gossypol-protein complexes by the reducing agent

NaBHsCN could explain why ruminant animals are less susceptible to gossypol

intoxication than monogastric animals. The rumen is a highly reducing environment,

55

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. without prohibited reproduction Further owner. copyright the of permission with Reproduced

Counts 1000 -time of flight-mass spectrometry) spectrograms of BSA (bovine serum serum (bovine BSA of spectrograms spectrometry) flight-mass of -time Figure 9. MALDI-TOF-MS (Matrix-assisted laser desorption ionization ionization desorption laser (Matrix-assisted MALDI-TOF-MS 9. Figure albumin) and BSA-G (bovine serum albumin-gossypol) conjugates albumin-gossypol) serum (bovine BSA-G and albumin) BSA-G/NaBH3CN(10/l mole ratio for reaction) for ratio mole BSA-G/NaBH3CN(10/l BSA-G (10/1 mole ratio for reaction) for ratio mole (10/1 BSA-G 3000 Mass(m/z) 56 007000 5000 68776.5 1356.7 Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. without prohibited reproduction Further owner. copyright the of permission with Reproduced

Counts 1000 Figure 10. MALDI-TOF-MS (Matrix-assisted laser desorption ionization ionization desorption laser (Matrix-assisted 10.MALDI-TOF-MS Figure -time of flight-mass spectrometry) spectrograms of BSA (bovine serum serum (bovine BSA of spectrograms spectrometry) flight-mass of -time albumin) and BSA-G (bovine serum albumin-gossypol) conjugates albumin-gossypol) serum (bovine BSA-G and albumin) BSA-G/NaBH3CN(20/l mole ratio for reaction) for ratio mole BSA-G/NaBH3CN(20/l BSA-G (20/1 mole ratio for reactio for ratio mole (20/1 BSA-G 3000 Mass(m/z) 57 5000 7000 66431 69615.6 72052 Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. without prohibited reproduction Further owner. copyright the of permission with Reproduced

Counts 00 00 00 7000 5000 3000 1000 Figure 11. MALDI-TOF-MS (Matrix-assisted laser desorption ionization ionization desorption laser (Matrix-assisted MALDI-TOF-MS 11. Figure BSA-G/NaBH3CN (20/1 mole ratio for reaction in ethanol in reaction for ratio mole (20/1 BSA-G/NaBH3CN -time of flight-mass spectrometry) spectrograms of BSA (bovine serum serum (bovine BSA of spectrograms spectrometry) flight-mass of -time albumin) and BSA-G (bovine serum albumin-gossypol) conjugates albumin-gossypol) serum (bovine BSA-G and albumin) BSA-G (20/1 mole ratio for reaction in ethanol) ethanol) in reaction for ratio mole (20/1 BSA-G Mass(m/z) 58 66431 | 3 69459.4 | having a redox potential of -0.4 volts (Brock et al., 1994) with a high microbial protein

supply (Van Soest, 1982). High protein content increases the chances for binding free

gossypol and the reducing environment favors stabilization of protein-gossypol Schiff

base intermediates. Moreover, gossypol contains two types of potentially reactive

functional groups, aldehydes and phenols. The condensation of gossypol with other

phenols and formation of insoluble metal complexes may be also involved in gossypol

detoxification in ruminants (Muzaffaruddin and Saxena, 1966; Ramaswamy and

O’Connor, 1968).

3.2: Production of polyclonal antibodies (PAb)

3.2.1: Monitoring PAb production

Figure 12 and Figure 13 show the increase in antibody over the immunization

period, tested using an antibody-capture noncompetitive ELISA (ACN-ELISA). The

increase reflected either a greater concentration or a higher binding affinity of anti-

gossypol antibodies in the rabbit serum over time. Both sera showed the highest titers

after four immunizations.

3.2.2: Optimization of ELISA for PAb

Several parameters, including microtiter plates, blocking buffers, 1st Ab and 2nd

Ab dilution buffers, incubation time and temperature, acetone and N,N-

dimethylformamide solvents, concentrations of coating solution (BSA-G) and antisera,

were investigated for their effects on ELISA.

3.2.2.1: Effects of microtiter plates on immobilization of BSA-G

The first step of an indirect ELISA is to directly coat BSA-G on the plastic plate

surfaces by passive adsorption. The rate of coating BSA-G to plates depends on the

59

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 10.5 9.5 Age (months) Arrows represent immunizations and squares represent bleeds. 6.5 7.5 8.5 Figure Antibody 12. production during immunization period from rabbit #16. 6 0.4 0.4 - 0.6

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Age (months) Arrows represent immunizations and squares represent bleeds. Figure 13. Antibody production during immunization period from rabbit #15 9 9 9.5 10.5 15.5 17.5 0 1.4 1.2 0.8 0.6 0.4 0.2

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. temperature, the BSA-G concentration and the type of solution. The higher

temperature, the greater the rate of adsorption (Crowther, 1995b). The most usual

regimens involve incubating at 4°C overnight or at RT for 1-3 hours. Considering the

instability of gossypol, increasing temperature may have a deleterious effect on the

antigen gossypol, so we selected 4°C overnight for the coating step. Under this

condition, different microtiter plates were compared including flexible

polyvinylchloride (PVC) and treated plates, such as Immulon 1 B, Immulon 2 HB and

Immulon 4 HBX. Of these plates, BSA-G produced different signal intensities

depending on the plates used (Figure 14 and Figure 15). Immulon 2 HB and 4 HBX

were found to have the highest signal to noise ratio (absorbance when 10 |ig/ml of

BSA-G used divided by absorbance when 0 pg/ml of BSA-G used) when compared to

Immulon 1 B or flexible PVC plates. According to the manufacturer (Dynex

Technologies Inc.), Immulon 2 HB was irradiated to provide high binding affinity for

proteins primary with hydrophilic tendencies and Immulon 4 HBX was treated to

improve protein adsorption in a hydrophilic environment. Immulon 1 B is suggested for

adsorption of proteins in a hydrophobic environment. In this study, BSA-G was

dissolved in PBS, so the greater adsorption on Immulon 2 HB or Immulon 4 HBX is

reasonable.

3.2.2.2: Selection of assay buffers

There are a wide variety of buffers used in ELISA, including those for coating,

blocking, primary and secondary antibody dilutions, washing and peroxidase substrate.

The solutions used for each stage of the ELISA are important for successful assay

development.

62

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 [BSA-G], pg/ml 0.1 1B (3.43) 1B Immulon 2HB (7.69) 2HB Immulon 0.01 (1.36) po|yvinylchloride Flexible — Immulon 0 The numbers shown in parentheses are signal to noise ratios (Abs.@ pg/ml/ 10 Abs.@ 0 pg/ml). 0.5 0.6 0.4 0.3 0.2 were coated with BSA-G (bovine serum albumin-gossypol) solutions and serum was diluted 1/1000. Figure 14. Comparison ofmicrotiter plates on ELISA for rabbit #16 PAb (polyclonal antibodies). Plates

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 0.1 [BSA-G], |ig/ml 0.01 ■*— Flexible polyvinylcliloride (4.03) polyvinylcliloride Flexible ■*— IB (1.76) -♦—Immulon 2HB (5.34) Immulon ■*— 4HBX (5.18) Immulon *— 0 The numbers shown in parentheses are signal to noise ratios (Abs.@ 10 pg/ml/ Abs.@ 0 pg/ml). 0.4 . 0.2 were coated with BSA-G (bovine serum albumin-gossypol) solutions and serum was diluted 1/1000. Figure 15. Comparison ofmicrotiter plates on ELISA for rabbit #15 PAb (polyclonal antibodies). Plates cn o < X> n O

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. High background can be caused by the nonspecific interaction of assay

components with the solid-phase. Nonspecific adsorption of protein can take place with

any available plastic sites not occupied by solid-phase reagent or by non-specific

binding to other protein reagents used in the assay. Blocking reagents were applied to

minimize this problem. They act by competing with nonspecific factors for available

sites. The criterion to choose a good blocker is to yield the highest signal to noise ratio.

Four blocking proteins: gelatin, casein, BSA and powdered milk, were compared for

their effects on signal to noise ratios. The results (Figure 16) showed that BSA and

milk gave higher signal to noise ratios than either casein or gelatin at 1% concentration.

Milk powder is much cheaper than BSA, and it was recommended as a promising

candidate for blocking by Vogt (1987). However, it must be used with care in ELISA

systems, because it is a complex mixture containing substances that may interfere with

assay reagents (Vogt, 1987). BSA is relatively pure, well characterized and readily

soluble, making it valuable as a blocking reagent. In this study, BSA was chosen as

blocking protein for these reasons.

Primary and secondary antibodies in dilution buffers containing 1, 0.5, 0.1 or

0.05% BSA in PBST were compared in ACN-ELISA. The results (Figure 17 and 18)

showed that high BSA concentration in PBST is more efficient at reducing the non­

specific binding than lower BSA concentrations, especially for rabbit #16 PAb where

non-specific binding seemed to be a more serious problem. BSA in buffer could

compete with 1st and 2nd Ab for nonspecific binding sites on the immobilized phase.

Higher BSA concentrations gave lower background absorbance and also suppressed the

binding of 1st Ab and/or 2nd Ab to immobilized phase.

65

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. [BSA-G], pg/ml 0.1 10 BSA (9.0) milk (7.7) gelatin (5.8) ovalbumin (7.1) 0.01 0 5 0 1.5 0.5 1/1000. 1/1000. The numbers shown in parentheses are signal to noise ratios (Abs.@ pg/ml/ 10 Abs.@ 0 pg/ml). albumin. Plates were coated with BSA-G (bovine serum albumin-gossypol) solutions and serum was diluted c/) < o ■'3- X) Figure 16. Comparison ofblockers on ELISA for rabbit #15 PAb (polyclonal antibodies). BSA is bovine serum ON 0\

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 [BSA-G], pg/ml 0.1 1%BSA (4.65) 1%BSA 0.5%BSA (4.01) 0.5%BSA (2.23) 0.1%BSA 0.01 0 0 1.5 0.5 1/1000. 1/1000. The numbers shown in parentheses are signal to noise ratios (Abs.@ pg/ml/ 10 Abs.@ 0 pg/ml). (A < o IT> <§> X> albumin. Plates were coated with BSA-G (bovine serum albumin-gossypol) solutions and serum was diluted Figure 17. Effect ofdilution bufferon ELISA forrabbit #16 PAb (polyclonal antibodies). BSA is bovine serum CT\

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 [BSA-G], |ig/ml 0.1 1% BSA(12) 1% 0.5% BSA (12.5) (12.5) BSA 0.5% 0.1% BSA (8.2) (8.2) BSA 0.1% (10.8) BSA 0.05% 0.01 0 2 0 1.5 0.5 1/1000. The numbers shown in parentheses are signal to noise ratios (Abs.@ 10 pg/ml/ Abs.@ 0 pg/ml). albumin. Plates were coated with BSA-G (bovine serum albumin-gossypol) solutions and serum was diluted Figure 18. Effect ofdilution buffer on ELISA for rabbit #15 PAb (polyclonal antibodies). BSA is bovine serum (A < O n O oo

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Detergents are included in washing steps and in 1st and 2nd Ab dilution buffers,

where they act to reduce non-specific binding. They reduce the surface tension of the

incubation media, thus inhibit hydrophobic interactions between proteins (1st Ab with

blocker, 2nd Ab with blocker), between proteins and sample, and between the reagents

and the uncoated surface. Detergent concentration should be kept as low as possible to

minimize the interference with Ab-Ag binding. Tween 20 at 0.5%, 0.1% and 0.05%

were compared in ELISA and they all efficiently decrease the nonspecific binding, so

the lowest concentration of 0.05% was selected.

3.2.2.3: Effects of incubation time and temperature on interactions of Ab-Ag

Incubation time and temperature are two important parameters for ELISA. The

interactions of Ab-Ag are noncovalent and reversible. Ab-Ag reactions in solid-liquid

interface are limited by mass transport or steric interactions (Stenberg and Nygren,

1988). The overall reaction scheme can be expressed as

ka Ag + Ab ** Ab-Ag kd

Where ka is the effective forward association constant and kd is the effective reverse

dissociation constant. The temperature can influence both reaction rate constants (ka

and kd), and the effect is not same for both. Kemeny (1991) found that near maximal

binding could be achieved within 2 hours for most antigens and antibodies. In the

current study, the incubation times (1 h vs. 2 h) and temperature (RT vs. 37°C) for both

of the antibody incubation steps (1st and 2nd antibodies) of ELISA were compared

(Figure 19 and Figure 20). Incubation for 2 h at RT gave greater signal to noise ratios

than either 30 min at 37°C or 1 h at RT. It is important that incubation time is long

69

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 1 [BSA-G], pg/ml 0.1 25°C (a> lh@ 25°C 30min 37°C(5) 2h 0.01 albumin-gossypol) solutions and serum was diluted 1/1000. PAb (polyclonal antibodies). Plates were coated with BSA-G (bovine serum 0 3 0 2 Figure 19. Effect ofincubation time and temperature on Ab-Ag interactions for rabbit #16 1.5 2.5 0.5 c /i < o o

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 [BSA-G],|ig/ml 0.1 25°C 30min (3) 37°C (3) 30min 0.01 albumin-gossypol) solutions and serum was diluted 1/1000. PAb (polyclonal antibodies). Plates were coated with BSA-G (bovine serum 0 3 2 0 .5 Figure 20. Effect ofincubation time and temperature on Ab-Ag interactions forrabbit #15 2.5 0.5

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. enough for Ab-Ag binding, so that the reaction is at or near equilibrium. After this

point, increased incubation time would not significantly affect the amount of colored

product obtained. Nevertheless, time can be shortened at the expense of signal, if a

faster analysis is desired but this would require further testing.

3.2.2.4: Effects of solvents on antibody capture noncompetitive ELISA

Ultimately this assay would be used to measure gossypol in cottonseed products.

For the preparation of gossypol sample and standard, certain solvents must be used for

dissolving gossypol or rupturing glands and this solvent must be compatible with

ELISA. Current available methods for gossypol analysis use either acetone or DMF for

sample preparation. These solvents and methanol were chosen to incubate with the 1st

Ab in an antibody capture noncompetitive ELISA format to predict whether these

solvents could be used in competitive ELISA for sample delivery. As acetone and

DMF concentration increased, serum reactivity was inhibited (Figure 21 and Figure 22).

Acetone concentrations equal to or greater than 10% reduced the reactivity of serum by

46%; DMF concentration, even at 4%, reduced the reactivity of serum by 50%; and

10% methanol gave similar signal as water at 10 pg/ml of BSA-G as coating solution.

Similar effects on the immunoreactivity of antibodies have been reported for organic

solvents (Chen et al., 1995). During the incubation of solvents and antibodies, acetone

and DMF may denature some Ab (proteins) or their binding sites, decreasing the

effective antibody concentration. This could result in lower signals. Therefore,

samples or standard solutions containing acetone or DMF solvents must be diluted in

ELISA to an acceptable range. Further investigation on the effects of these solvents on

the competition ELISA is necessary.

72

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3U0J30V %0t>

suopoy %oe

suopoy o/0Q 3

ouopoy %o I

auopoy %g

3uopoyo/0g'3

suopoy %g3' I

HQ3IAI%0I

OzH Figure Figure 21. Effect of acetone concentration on ACN-ELISA (antibody capture noncompetitive ELISA)

uiu sot1 ® sqy

73

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (antibody (antibody capture noncompetitive ELISA) Figure Figure 22. Effect of N,N-dimethylformamide concentration on ACN-ELISA

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3.2.2.5: Effects of Ab and BSA-G concentrations

The optimal concentrations of Ab and solid-phase conjugates were determined

by checkerboard ELISA (Figure 23 and Figure 24). Increasing the coating antigen,

BSA-G, above 5 pg/ml did not significantly increase the overall absorbance. Therefore,

the optimum concentration of coating antigen conjugate was determined to be 5 pg/ml.

The optimum combination of antiserum and coating solution were chosen where they

gave steep dilution curves and an absorbance between 1 to 2. Serum dilutions of 1/1000

were used in the remaining studies presented in this dissertation.

3.2.3: Effects of immunogen preparation method on PAb production

The results from serum screening and ELISA optimization indicated that sera

from different rabbits showed different behaviors. One reason could be due to animal

to animal variation in immune response. Other reasons may be the chemistry used for

formation of immunogen and immunogen purity (Skerritt, 1995).

The chemistry of immunogen formation plays an important role in determining

assay sensitivity and assay specificity. Careful choice of conjugation to the carrier

protein is necessary (Harrison et al., 1991a and 1991b). The presence of spacer arms

could maximize the exposure of the analyte for antibody production and increase the

assay sensitivity (Chen et al., 1995). In this research, gossypol was bound to e-amino

groups of lysine on the protein. The hapten design and synthesis are critical steps to

elicit a good immune response. Rabbit #16 was injected with LPH-GA without

stabilization, and rabbit #15 was injected with LPH-G8 with the addition of NaBfyCN

for product stabilization. Though substitution ratios could not be detected using

MALDI because of the aggregation tendency of LPH, it can be deduced from the results

75

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 10 5 [BSA-G], pg/ml 0.1 1/500 1/2000 1/4000 1/8000 ^ 0.01 0 3 2 0 1.5 2.5 0.5 Figure 23. ELISA checkerboard for rabbit #16 PAb (polyclonal antibodies). Plates were coated with BSA-G (bovine serum albumin-gossypol) solutions. Different lines represent different PAb dilutions. i/i < ■'T in o X> ON

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 100 10 5 [BSA-G], ng/ml 0.1 1/500 1/4000 1/8000 1/2000 -o- 1/1000 0.01 0 0 3 9 1.5 0.5 2.5 Figure 24. ELISA checkerboard for rabbit #15 PAb (polyclonal antibodies). Plates were coated with BSA-G (bovine serum albumin-gossypol) solutions. Different lines represent different PAb dilutions. C /3 <

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. of BSA-G conjugation that LPH-G8 with stabilization might also have a greater number

of gossypol groups per LPH than LPH-GA. Also, the secondary amino produced by

stabilization would be expected to provide a more flexible linking arm, perhaps making

the hapten more accessible. The incorporation ratio between hapten and carrier protein

LPH is important to get a good immune response. The data from Erlanger (1980)

suggested that incorporation levels on BSA from 8:1 to 25:1 gave a good response. For

different sizes of carrier proteins, different molar substitution ratios may be required.

The packing density is important in the hapten-protein conjugate. Thus, a large carrier

protein should have a greater molar substitution ratio than small proteins to maintain the

same packing density (Jenner and Law, 1996). The substitution ratio may be one reason

to cause the different immune response in this research.

Purification of the crude immunogen to remove non-covalently bound haptens

may have also affected the Ab response. Haptens may be adsorbed to the carrier in a

way which presents an epitope differently than that of the chemically conjugated

material. Injecting an immunogen containing both covalently linked and adsorbed

hapten could result in the generation of a mixed population of antibodies with different

specificities (Jenner and Law, 1996). LPH-GA and LPH-G8 were purified using

different methods which may have given different immunogenic properties.

3.3: Production of monoclonal antibodies

3.3.1: Mouse immunization

After mice were immunized four times, all five mice showed production of anti-

gossypol antibodies by ACN-ELISA (Figure 25). Mouse #2 showed the highest titer of

antibody and this mouse was selected as the spleen cell donor to fuse with myeloma.

78

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Figure Figure 25. Antibody production from mice after four immunizations

sot? ® ‘sqv

79

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3.3.2: Production and selection of hybridomas

Spleen cells (108) were fused with myeloma (107) using sterile PEG-1500 fusion

solution. The fused cells were suspended in HAT selection system to grow the

hybridoma and prevent growth of unfused myeloma. After fusion, the plates were

examined daily for contamination, pH (by color of medium) and colony growth.

After 10-15 days, the cells showing higher signals (72 wells) than control

(myeloma) in ACN-ELISA were transferred into 24-well plates and cultured in RPMI-

20 medium. After three days, supernatants from wells (1A7, 1C6, 1F9, 2G5, 3A5,

3D 10, 4C7, 4D8, 4F2, 5D2, 5C5, 5D7, 3H9, 5H9, 6B10, 6G7, 6H2, 7G2) still showing

positive in ACN-ELISA were tested using ACN- and ACC-ELISA simultaneously.

Cells showing both high absorbance (Ao) in ACN-ELISA and low absorbance (A) in

ACC-ELISA with 100 pg/ml of gossypol-lysine are shown in Table 3. At this stage,

healthy and positive cells from the wells 1C6, 3H9, 5D7, 5H9, 6G7, 6H2 and 7G2 were

selected for cloning.

Table 3. Competition using gossypol-lysine (100 pg/ml of gossypol equivalents) as competitor. A is the absorbance from ACC-ELISA and Ao is the absorbance in ACN- ELISA.

Cell line A/Ao Cell line A/Ao 1A7 0.3695/1.006 5D2 0.5652/2.9805 1C6 0.455/0.819 5C5 0.2817/0.4472 1F9 0.3437/1.7702 5D7 0.623/1.557 3A5 0.3875/0.6945 6G7 0.441/0.797 3D10 0.2833/0.3808 6H2 0.295/0.5050 4C7 0.5126/1.199 7G2 0.655/1.066 4F2 0.317/0.971

80

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The semi-solid agar method was used to clone cells 6G7. Eight days after

cloning, the colonies were picked from soft agar and transferred into 200 pi of RPMI-20

medium in 96-well plate. After culturing and propagation, the cells from 6G7D11,

6G7D2, 6G7D7, 6G7G3, 6G7B6, 6G7D12 showed positive in ACC-ELISA using

lysine-gossypol, arginine-gossypol, ethanolamine-gossypol and 3-amino-1-propanol-

gossypol as competitors (Appendix I). 6G7D11 was selected and recloned using the

limiting dilution method.

Limiting dilution was also used to clone cells from wells 1C6, 3H9, 5D7, 5H9,

6H2 and 7G2. Twelve to fifteen days after cloning, the cells from the highest dilution

were tested using ACN- and ACC-ELISA, and the positive ones (5D7D3, 5D7E3,

5D7F3, 5D7E4, 5D7F4, 5D7G4, 7G2A5, 7G2B5, 7G2A6, 5H9F8, 5H9F2) (Appendix

I) were recloned using limiting dilution. Poisson distribution statistics indicates that if

the most probable cell number per well is 1, then 37% of wells will have no colonies at

all, and it is highly probable that the cells growing within each of the individual wells

are derived from a single parent cell (Zola, 1987a).

After three clonings, 7G2F2G3C6 was obtained, and both 6G7D11F10F2F8 and

6G7D11F10F2H3 were obtained from the thrice cloned cell line 6G7D11F10F2.

3.3.3: Correlation between cell growth and antibody production

Once stable hybrid clones secreting anti-gossypol antibodies were obtained, a

batch MAb was produced and semipurified. The three cell lines were maintained in

RPMI-20 medium and subcutured to keep the cell density within the range of 105-106

cells/ml. Cell growth rate and Ab production of cell line 6G7D11F10F2F8 were

monitored. Cell numbers doubled about every 24 h, and when the cell number reached

81

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 1.4xl06 cells/ml, cells began to die and antibody concentration decreased (Figure 26

and Figure 27). In order to maximize antibody yield from culture, the supernatants

should therefore be collected when cells are at peak of cell density or at around 3 days

for the conditions employed here. The collected supernatants were combined and

concentrated using 55% saturated ammonium sulfate, dialyzed, and stored in a -20°C

freezer.

3.4: Effects of reaction pH and structure of derivatizing reagents on gossypol derivatization

Gossypol derivatives with L-(+)-lysine, L-(+)-arginine, ethanolamine, L-2-

amino-1 -propanol, 3-amino-1-propanol, para-amino-benzoic acid, L-glutamine, L-(-)-

tryptophan and L-tyrosine were made in PBS buffer solution (pH7.3). At their

isoelectric point (pi), amino acids exist as zwitterions (Lehninger et al., 1992).

Gossypol is a weakly acidic compound because of its phenol groups. At pH7.3, both

gossypol and the carboxyl group of an amino acid (pl<3) would bear negative charges

and repel each other. This would slow down the reaction between these compounds.

Therefore, an amino group far from the carboxyl group, such as the e-amino group of L-

(+)-lysine and the amino groups of L-(+)-arginine would be expected to react faster than

an amino group near to the carboxyl group, such as the a-amino groups of L-glutamine,

L-(-)-tryptophan and L-tyrosine. TLC results showed that gossypol disappeared after

10 min at RT in the reaction with L-(+)-lysine and L-(+)-arginine, and the formation of

a gossypol complex resulted in the maximum absorption shifting from 386 nm (for

unreacted gossypol) to 404-406 nm for gossypol-lysine and gossypol-arginine (Figure

28). After 2 h at RT, TLC showed that some gossypol was still underivatized in the

82

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. o 00 Tl“ o Figure Figure 26. The growth rate of MAb cell line (monoclonal antibody cell line: 6G7D11F10F2F8)

, 0 1 x-ou [P3

83

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. oo Figure Figure 27. MAb (monoclonal antibody cell line: 6G7D11F10F2F8) production during culture

urc SOt? © ‘sqv

84

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. •5 obr* v CO> a. a. Gossypol b. L-(+)-lysine-gossypoI c. L-(+)-arginine-gossypol Figure Figure 28. Absorption spectra of gossypol and gossypol derivatives in methanol, 10%

CO 00 CO CM o

soircqjosqv

85

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. reactions of gossypol with L-glutamine, L-(-)-tryptophan, L-tyrosine and para-amino-

benzoic acid under the same conditions.

3.5: Isolation of (+/-)-gossypol isomers

In order to evaluate the antibody specificity for (+)- and (-)-gossypol isomers,

isomeric gossypol was isolated through gossypol crystallization from acetone. Each

individual crystal contains only one isomer (Dowd, 1998). This unique crystallization

property of gossypol makes it valuable for the isolation of extremely pure (+/-)-isomers.

Isolated isomers were identified using HPLC (Figure 29). The results showed that the

purity for combined (+)-gossypol crystals was 99.96% and for combined (-)-gossypol

was 99.64% (Appendix II for area count). Slight contamination (<0.04% for (+)-

gosypol and <0.4% for (-)-gossypol) probably resulted from contamination of the

tweezers used to pick up the crystals.

3.6: Evaluation of antibody specificity using different forms of gossypol in an antibody capture competitive ELISA

It has been known that antibodies produced against a given immunogen will

recognize the compound’s different moieties to different degrees. Racemic gossypol

derivatives, (+)- and (-)-isomeric gossypol derivatives and gossypol (section 2 .2 . 1 0 ) as

standard solutions were used in an antibody capture competitive ELISA (ACC-ELISA)

to evaluate the specificities of the PAb and MAb. Four parameter sigmoidal equations

were used to determine I 50 values based on the least square errors of the observed data.

For the purpose of discussion, the chemical structure of gossypol can be divided into

three recognizable moieties. These include 1) a derivatized naphthyl group, 2) a

naphthyl group without derivatization, and 3) the inter naphthyl bond. For PAb from

rabbit #15, about 0.2 pg/ml of gossypol equivalents caused 50% inhibition of binding

86

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. v) sa co 3.50 4.00 3.50 4.00 CO CO o CM co CM 2.50 3.00 CD 2.50 3.00 O) O) ^ 5 w V | JA S* 1.50 1.50 1.00 1.00 0.50 0.50 ------80 60 2 0 0 0 80 00 60 2 0 60 20 60 2 0 40 ...... 1 1.40 1 1 1.40 1.80 1 1 1 1 0 0 0.80 0.40 0 0 0.00 0 0 0.00 -0.20J -0.20J Figure 29. HPLC (High performance liquid chromatography) chromatograms of(+/-) gossypol isomers

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (I50) when gossypol derivatized with 3-amino-1-propanol, 2-amino-1-propanol,

ethanolamine, L-(+)-lysine, and L-(+)-arginine were used as competitors. These results

indicate that this PAb could not recognize the difference between these four gossypol

derivatives (I5 0 values were not significantly different in Tukey’s Studentized Range

Test with significance level of 95%), possibly because PAb may only recognize a

naphthyl group on gossypol without derivatization. Gossypol and gossypol derivatives

with para-amino-benzoic acid, L-glutamine, L-(-)-tryptophan and L-tyrosine as

competitors, however, gave much higher I50 values (Figure 30 and Figure 31). The Ab

may not recognize free gossypol, since it was raised to bound gossypol during

immunization, no competition between free gossypol and immobilized gossypol. It

may also be feasible that underivatized gossypol (in gossypol solution and gossypol

derivative reaction solutions) covalently binds to antibodies through Schiff base during

the ELISA. This would reduce the effective concentration of the competitor. The

covalently bound antibodies with gossypol still could interact with immobilized

antigens on Ab-Ag binding sites through hydrogen bonds, hydrophobic forces, Van Der

Waals forces and electrostatic forces. These all would decrease the effective

competition between free antigens and immobilized antigens, increasing the I50 values.

Figure 32, Figure 33 and Figure 34 show the inhibition curves for gossypol

derivatives using three different MAb. MAb from different cell lines had different

affinities/specificities to different gossypol derivatives. When gossypol derivatives were

used as competitors, I50 values ranged from 20 to 55 |ig/ml for 7G2F2G3C6, from 8 to

26 pg/ml for 6G7D11F10F2F8, and from 7 to 24 pg/ml for 6G7D11F10F2H3

(Appendix III). The results indicated that MAb with different specificities have

88

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 25 250 2.5 [Gossypol equivalents], pg/ml 0.00025 0.0025 0.025 0.25 are I50 values (theare concentrationsI50 which caused 50% inhibition ofbinding). lysine-gossypol (0.197)lysine-gossypol arginine-gossypol arginine-gossypol (0.209) -propanol-gossypol 3-amino-1 (0.174) 2-amino-1 -propanol-gossypol 2-amino-1 (0.195) gossypol (underivatized) (>250) —ethanolamine-gossypol (0.225) 0 0 0.8 0.6 0.2 0.4 (polyclonal antibodies). Each point is the average of 2 determinations. The numbers shown in parentheses Figure 30. ELISA inhibition curves using gossypol and different gossypol derivatives for rabbit #15 PAb csi t/j <1 o < X X 00 vo

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 12500.000125 0.00125 0.0125 0.125 1.25 12500.000125 12.5 [Gossypol equivalents], pg/ml glutamine-gossypol (>125) glutamine-gossypol (>125) tryptophan-gossypol acid-gossypol(>125) amino-benzoic tyrosine-gossypol (>125) tyrosine-gossypol I50 values (the I50 concentrations which caused 50% inhibition ofbinding). 0.8 0.6 0.4 0.2 antibodies). Each point is the average of 2 determinations. The numbers shown in parentheses are Figure 31. ELISA inhibition curves using additional gossypol derivatives for rabbit #15 PAb (polyclonal c/l in in < o ©

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. [Gossypol equivalents], pg/ml lysine-gossypol (28.31) lysine-gossypol arginine-gossypol (21.51) arginine-gossypol (22.75)ethanolamine-gossypol 2-amino-1-propanol-gossypol (55.05) 3-amino-1-propanol-gossypol (20.86) I50 values (the I50 concentrations which caused 50% inhibition ofbinding). Each point is the average of 2 determinations. The numbers shown in parentheses are Figure 32. ELISA inhibition curves using different gossypol derivatives for MAb (7G2F2G3C6). c/5 c/i IT) SO

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. [Gossypol equivalents], pg/ml 0.25 lysine-gossypol (9.93) argine-gossypol (8.41) ethanolamine-gossypol (10.96) 2-amino-1-propanol-gossypol (21.97) 3-amino-1-propanol-gossypol (26.25) I50 values (the I50 concentrations which caused 50% inhibition ofbinding). - - - 0.8 0.6 0.4 - 0.2 Each point is the average of 2 determinations. The numbers shown in parentheses are m co Figure 33. ELISA inhibition curves using different gossypol derivatives for MAb (6G7D11F10F2F8). ■st < o < X <§> X i SJ so

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 250 I 25 “T 2.5 [Gossypol equivalents], pg/ml 0.25 2-amino-1 -propanol-gossypol (18.63) -propanol-gossypol 2-amino-1 (23.99) 3-amino-1-propanol-gossypol argine-gossypol (8.10) argine-gossypol (7.38) ethanolamine-gossypol ■ lysine-gossypol (8.17) lysine-gossypol ■ I50 values (the concentrationsI50 which caused 50% inhibition ofbinding). Each point is the average of 2 determinations. The numbers shown in parentheses are 0 1 0 0.9 0.8 0.7 0.2 0.1 0.6 0.5 0.4 0.3 Figure 34. ELISA inhibition curves using different gossypol derivatives for MAb (6G7D11F10F2H3). in in < iri o < X>

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. potential to differentiate various forms of bound gossypol formed during cottonseed

processing.

Fresh made gossypol solution (in 10% methanol) was compared with 21 day old

gossypol solution in ACC-ELISA using both MAb (6G7D11F10F2F8 and

6G7D11F10F2H3) and PAb (Figure 35 and Figure 36). Freshly made gossypol as

standard did not show good competition (Iso>250 fig/ml) comparing with 21 day old

gossypol solution (Iso=1.59 (ig/ml) for PAb. Using MAb, however, underivatized

gossypol showed no competition when either 21 day old or freshly made gossypol

solutions were used at similar concentrations that were used with PAb. During storage,

some free aldehydic gossypol may transform into other tautomers such as hemiacetal in

methanol (Abdullaev et al., 1990). Gossypol may be oxidized and/or degraded into

various products with different properties (Markman, 1968; Nomeir and Abou-Donia,

1982). The gossypol derivatives that formed over the 21 days of aging were apparently

reactive to PAb, increasing the effective gossypol concentration for competition. This

resulted in a lower Iso value as compared with freshly prepared underivatized gossypol.

Storage time seemed to have no effect on competition using MAb, possibly because of

their higher specificity. MAb may be able to differentiate Schiff base formed gossypol

derivatives and gossypol tautomers, oxidized forms and degraded forms.

Isomeric (+)- and (-)-gossypol derivatives were also tested in ACC-ELISA to

evaluate the specificity of MAb cell lines (6G7D11F10F2F8 and 6G7D11F10F2H3).

Iso values were not significantly different when (+)-gossypol and (-)-gossypol isomers

were compared using Tukey’s Studentized Range Test with significance level of 95%.

The MAb therefore had a similar affinities or specificities to (+)- and (-)-gossypol

94

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 25 0.25 2.5 250 [Gossypol equivalents], pg/ml 0.0250 freshly made freshly gossypolsolution (>250) 21 21 day old gossypol(1.59)solution I50 values (the concentrationsI50 which caused 50% inhibition ofbinding). . - - 0.8 0.6 0.4 - 0.2 antibodies). Each point is the average of 2 determinations. The numbers shown in parentheses are Figure 35. Effects ofgossypol storage time on ELISA inhibition curves for rabbit #15 PAb (polyclonal co < o ■'3’ § X> VO t-ri

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 25 250 [Gossypol], pg/ml freshly madefreshly gossypolsolution (H3) freshly madefreshly gossypolsolution (F8) 21 21 day old gossypol solution (H3) 21 21 day old gossypolsolution (F8) 0.025 0.25 2.5 0.00250 6G7D11F10F2H3 cell line, respectively. Each point is the average of 2 determinations. 0.8 0.6 0.4 0.2 and 6G7D11F10F2H3). F8 and H3 shown in parentheses represent 6G7D11F10F2F8 cell line and Figure 36. Effects ofgossypol storage time on ELISA inhibition curves for MAb (6G7D11F10F2F8 n c/i t o ■>3- < < XI XI X n v £ > O

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (Figure 37 and Figure 38) and did not differentiate between the two isomers. It seems

that these MAb recognize the naphthyl group and not the inter-naphthyl bond.

A number of statistical approaches have been set up for the definition of

detection limit and limit of quantification in immunoassay. However, many users and

test kit manufacturers do not use statistical methods, and would like to define the limit

detection as the concentration providing either 10% or 15% inhibition of color of

development; some researchers define positive samples as providing color development

of at least 0.1 OD units less than control (Skerritt, 1995). In this study, the

concentration which inhibited color development of 0.1 OD unit comparing with control

was defined as the detection limit (Table 4).

Table 4. Detection limit for antibodies using gossypol derivatives as competitors

Detection limit (pg/ml) Gossypol derivatives Monoclonal antibodies polyclonal antibodies 7G2F2G3 6G7D11F 6G7D11F C6 10F2F8 10F2H3

lysine-gossypol 0.023 8.7 2 .8 1.4

arginine-gossypol 0.03 4.7 2.5 1.4

2 -amino - 1 -propanol-gossypol 0.0025 21.7 4.3 1.7

3 -amino-1 -propanol-gossypol 0.0125 5.5 5.6 4.2

ethanolamine-gossypol 0.05 1 0 .1 3.0 0.9

Usually MAb have better specificity than PAb, so the detection limit is often

higher than those based on PAb (Szurdoki et al., 1995). Surprisingly it was found that

PAb had much lower I 50 values (Iso values for inhibition curves are summarized in

Appendix III) and much lower detection limit than MAb (Table 4) when gossypol

97

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 250 25 2.5 [Gossypol equivalents], p.g/ml 0.250 (+)-gossypol-2-amino-1 -propanol -propanol (10.67) (+)-gossypol-2-amino-1 (+)-gossypol-lysine (12.72) (+)-gossypol-lysine (13.47) 1-lysine (-)-gossypo (-)-gossypol-2-amino-l-propanol (9.69) in parentheses values are(the concentrationsI50 which caused 50% inhibition ofbinding). for MAb (6G7D11F10F2F8). Each point is the average of4 determinations. The numbers shown - - 6 . 0 0.4 - 0.4 0.2 cA t/i < < 43 O 43 43 Figure 37. Comparison of(+/-)- gossypol derivatives in ACC-ELISA (antibody capture competitive-ELISA) oo VO

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. [Gossypol equivalents], pg/ml (-)-gossypol-2-amino-1-propanol (9.09) (+)-gossypol-lysine (10.64) (-)-gossypol-lysine (9.81) (+)-gossypol-2-amino-l-propanol (7.44) in parentheses values are (the concentrationsI50 which caused 50% inhibition ofbinding). 0 0.25 2.5 25 250 for MAb (6G7D11F10F2H3). Each point is the average of4 determinations. The numbers shown 0.4 - t/i t/i in ■sf < < o X X X Figure 38. Comparison of(+/-)- gossypol derivatives in ACC-ELISA (antibody capture competitive-ELISA)

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. derivatives with 3-amino-1-propanol, 2-amino-1-propanol, ethanolamine, L-(+)-lysine,

and L-(+)-arginine were used as competitors. One reason may be that PAb are less

specific, so they cross-react with different forms of gossypol in the reaction mixture

(including gossypol derivatives, oxidized gossypol and degraded gossypol formed

during reaction). MAb may provide a greater specificity to certain forms of gossypol

derivatives in the reaction mixture, therefore requiring higher concentrations of mixed

solutions to compete with the solid-phase comparable to PAb. This would result in a

lower apparent detection limit when testing a mixture of gossypol derivative products.

Ab having broad specificity may be useful in some situations to screen for a family of

compounds. Compound-specific Ab may be preferred for other situations where cross­

reactivity is undesirable. Quite often, high titer PAb give lower detection limits than

MAb for compound mixtures because of PAb cross-reactivity (Skerritt, 1995).

3.7: Application of ELISA for the analysis of gossypol in cottonseed products

The detection limit was much lower for PAb than MAb, so PAb were adopted

first to analyze gossypol in cottonseed samples. L-(+)-lysine was chosen to derivatize

gossypol because of the efficiency of the reaction.

Seven cottonseed product samples with a variety of Free Gossypol (FG) from

0.08% to 1.12% and Total Gossypol (TG) from 1.06 to 1.35% (Table 5) were selected

to be analyzed by ELISA. FG and TG were previously measured (by Dr. M. Calhoun,

Texas A&M University) using AOCS official methods (AOCS, Ba 7-58 and Ba 8-78)

and (+/-)-gossypol isomers were measured using HPLC.

100

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Table 5: Gossypol contents of cottonseed products*

AOCS® HPLC® Sample Treatment description ID* FG% TG% (+)-G (-)-G

2241 Cottonseed meal 0.08 1.06 56.5 43.5

2817 LFFA CS® meats once through expeller 0.33 1.26 57.4 42.6

2822 HFFA CS® meats once through extruder 0.36 1.10 58.2 41.8

2819 HFFA CS meats roasted 20 min 0.91 1.21 58.2 41.8

2820 HFFA CS meats roasted 40 min 0.93 1.26 58.2 41.8

2814 LFFA CS meats roasted 40 min 1.06 1.35 58.2 41.8

2813 LFFA CS meats roasted 20 min 1.12 1.34 57.9 42.1 * samples obtained from M. Calhoun and samp e ID number assignee in the Nutrition/Toxicology Laboratory at the Texas A&M University Agricultural Center, San Angelo, TX; Free Gossypol, Total Gossypol and gossypol isomers measured by M. Calhoun, Texas A &M University; ® low ffee fatty acid cottonseed, ® high free fatty acid cottonseed

For TG analysis by ELISA, samples were extracted and derivatized using L-(+)-

lysine in 10% methanol. Following 2 h reaction at RT, the extracted slurry was

analyzed in ELISA. Results gave poor correlation (r2=0.4332) between the ELISA

method and AOCS official method (AOCS, Ba 8-78), and the values were much lower

from our ELISA method than from the AOCS official method for the seven samples

(Figure 39). One major difference between these two methods is the difference in the

extraction and derivatizing reagent used for sample preparation. The AOCS official

method (AOCS, Ba 8-78) employs 3-amino-1 -propanol in DMF as complex reagent

under 95°C, rather than L-(+)-lysine in 10% methanol at RT in this study. Gossypol is

embedded in the cottonseed gland, and the gland consists of an envelope of 2-3 layers

of thin-wall tablet shaped cells (Markman, 1968). Even if a sample is ground, it may

101

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. ELISA □ AOCS Sample ID average of4 determinations. 2241 2241 2817 2822 2819 2820 2814 2813 sample delivery) and AOCS official method (Ba 8-78). Each point theis Figure 39. Total Gossypol (TG) in cottonseed samples analyzed by ELISA (10% methanol for TG% t o o

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. not break all the glands (wan, 1999). Mild extraction and derivatizing complex reagent,

such as L-(+)-lysine in 10% methanol at RT may not efficiently rupture gossypol

glands. Moreover, differences in sample processing, hardness, fatty acid content,

moisture and purity may influence the grinding efficiency causing the gossypol glands

to rupture differently for different samples. Although these all differences in grinding

efficiency may also be occurring during sample preparation for the AOCS method, they

may not be as apparent because of the strength of the DMF solution. The extracted

slurry was directly used in ELISA, and some bound gossypol with protein and other

compounds may not be released into this slurry. These all could result in lower values.

In order to increase the extraction and derivatization efficiency, samples can be

extracted using 3-amino-1-propanol in dimethylformamide (DMF) solution (AOCS, Ba

8-78). However, because of the interference of this solvent in ELISA, the proportion of

DMF used in ELISA needs to be investigated and controlled to minimize problems.

Our results indicated (Figure 22) that 4% of DMF in non-competitive ELISA could

reduce the reactivity of serum by 50% (abs.=l), so the recommended concentration will

be lower than this level. The effect of levels of DMF on competitive ELISA should also

be investigated. Under ideal conditions, TG measured using ELISA should include all

gossypol and gossypol derivatives. Defining what “all gossypol derivatives” includes

will prove very challenging.

For FG analysis, samples were extracted with 70% aqueous acetone, and

filtered. The filtrate was gently purged using nitrogen gas to remove the acetone, then

methanol with L-(+)-lysine solution was added. After derivatization, the sample was

analyzed in ELISA (Figure 40). FG results obtained by ELISA were lower than FG

103

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 2814 2813 2820 Sample ID point is the average of4 determinations. 2817 2822 2819 □ AOCS (x) ■ ELISA (y) Y=0.5569x-0.0045 r^O.69 2241 methanol for sample delivery) and AOCS official method (Ba 7-58). Each - - - 8 6 Figure 40. Free Gossypol (FG) in cottonseed samples measured using ELISA (10% 2 . . . 0 0 0.4- 0 FG% o •fc.

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. results from AOCS method (Ba 7-58). After removing the acetone, the solutions

became cloudy. Water is a poor solvent for gossypol, therefore some gossypol may

have crystallized from the solution. After adding L-(+)-lysine, gossypol may not have

been completely derivatized. Underivatized gossypol could decrease the effective

gossypol concentration in ACC-ELISA, resulting in lower gossypol values as compared

to the results obtained using the AOCS official method.

Because of the poor solubility of gossypol in water, the above method for FG

analysis was modified. FG was extracted using 70% acetone and filtered (AOCS, Ba 7-

58), and L-(+)-lysine in PBS was slowly added into filtrate to dilute the acetone

concentration from 70% to 10%. Following 2 h reaction at RT, the samples were

analyzed using ELISA. The results showed good correlation (^=0.96) between our

ELISA and AOCS official method (Figure 41, Appendix IV) under the conditions of the

method. However the results from ELISA were approximately two times higher than

those determined using AOCS methods.

It is possible that some gossypol was oxidized and/or degraded during sample

storage and shipment. Loss of the aldehyde groups would result in nondetection by the

AOCS official method (AOCS, Ba 7-58). The ELISA, however, may still detect these

products, depending on their reactivity to the Ab binding sites and their extractabilities.

Free Gossypol measured by ELISA method could therefore be defined as gossypol and

gossypol derivatives which are soluble in 70% acetone and have affinity for the PAb

used in the assay. Bound Gossypol (BG) can be determined mathematically, and BG

can be defined as gossypol derivatives which could not be extracted by 70% acetone but

have specific binding sites for PAb used in the assay.

105

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 8 2813 2814 Sample ID 2822 2819 2820 2817 y=2.0885x rMJ.96 determinations except sample 2241 which is the average of 5 determinations. □ AOCS (x) ■ ELISA (y) sample delivery) and AOCS official method (Ba 7-58). Each point is the average of 2241 Figure 41. Free Gossypol (FG) in cottonseed samples determined using ELISA (10% acetone for 1.5 1.5 - 2.5 - 0.5 - FG% n o O

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3.8: Future research

In this study, anti-gossypol PAb and MAb were successfully produced, and the

model system studies showed that PAb and MAb could recognize gossypol derivatives

formed via Schiff bases. Preliminary research on gossypol analyses of cottonseed

products appeared to be a promising and challenging research.

In order to produce and characterize different MAb cell lines with different

specificities, different forms of relatively pure gossypol and gossypol derivatives

(including gossypol derivatives formed via a Schiff base intermediate and via phenol

hydroxy group condensation, oxidized gossypol, degraded gossypol, (+)- and (-)-

gossypol, and (+)- and (-)-gossypol derivatives) need to be made to produce, screen and

select hybridomas, and to evaluate the affinities/specificities of the Ab produced.

MAb with different specificities should be adapted and optimized in an

immunoassay to rapidly and accurately measure the quantity of gossypol. This could

potentially be adapted to a kit format for use by the cottonseed industry, veterinarians

and plant breeders. The immunoassay results need to be correlated with established

AOCS colorimetric methods, HPLC, and bioavailability measurements using a large

number and variety of samples. The advantages of specific binding between Ab-Ag for

measuring different forms of gossypol, and should be further exploited to improve our

knowledge of the forms and bioactivity of bound gossypol.

MAb having specific affinity to either (+)- or (-)-gossypol could provide an

alternative method to isolate gossypol isomers. The available methods so far reported

to separate gossypol isomers include HPLC method after conversion of gossypol into

diastereomeric Schiff-bases (Matlin, et al., 1987) and crystallization from acetone

107

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. (Dowd, 1998). (+)-Gossypol and (-)-gossypol have different biological activities, and

the more potent anti-spermatogenic, anti-tumor and anti-HIV properties of (-)-gossypol,

have led to a requirement for large quantities of this specific isomer for further

biological studies. Immunoaffinity columns using MAb have been developed

commercially in the United States, the United Kingdom (Scott and Trucksess, 1997)

and Canada for the isolation of many types of analytes.

The current research demonstrated the production of antibodies to gossypol.

The potential uses of this technology are many, from quality control test kits by use in

industry, to research tools to better understand the physiological effects of gossypol.

108

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. CHAPTER 4

SUMMARY AND CONCLUSIONS

The development of immunochemical methods provided a potential tool to study

gossypol chemistry in model systems and to analyze gossypol in cottonseed products.

Protein-gossypol conjugates and gossypol derivatives were produced via Schiff

bases, and the addition of stabilizing reagent NaBHsCN favored Schiff base formation

and increased product yield. High purity (+)- and (-)-gossypol isomers (> 99%) were

obtained through gossypol crystallization from acetone.

Polyclonal antibodies (PAb) against gossypol could be produced when rabbits

were immunized with LPH-gossypol immunogens. The preparation methods for

immunogens were found to greatly affect antibody production. Assay parameters, such

as microtiter plates, incubation time and temperature for Ab-Ag interactions, dilution

buffers for 1 st and 2 nd antibodies, and blocking buffers could greatly influence the

ELISA.

Monoclonal antibodies (MAb) were developed by fusion of immune mouse

splenocytes and NS-1 myelomas under PEG-1500 fusion buffer.

PAb and MAb were used in an antibody capture competitive-ELISA and

different forms of gossypol were used to evaluate the antibodies for their sensitivities

and specificities. Under optimum conditions, approximately 0.2 (ig/ml of gossypol

equivalents caused 50% binding inhibition (I 50) using rabbit PAb, and 7 to 55 (ig/ml of

gossypol equivalents caused 50% binding inhibition of MAb to the solid-phase (BSA-

G), when gossypol derivatives with L-(+)-lysine, L-(+)-arginine, L-2-amino-l-propanol,

109

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. 3-amino-1-propanol and ethanolamine were used competitively. The MAb cell lines

obtained had equal affinity/specificity for (+)- and (-)-gossypol isomeric derivatives.

Underivatized gossypol as competitor gave much higher I 50 values for PAbs and showed

no competition for MAb, indicating that either the antibodies showed low or no

recognition of unbound gossypol or that gossypol reacted non-specifically to reagent

proteins.

Gossypol samples were analyzed using PAb in an antibody capture competitive-

ELISA. Preliminary research indicated that extraction and derivatization methods for

sample delivery could greatly influence the results. After improvement of the method,

Freel Gossypol analysis gave good correlation (^=0.96) between ELISA and AOCS

official methods, so ELISA methods seem to have potential to provide a rapid and

accurate method for gossypol analysis.

110

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. REFERENCES

Abdullaev, N.D., Tyshchenko, A.A., Nazarova, L.P. Ul'chenko, N.T., Yagudaev, M.R. and Glushenkova, A.I. 1990. !H and l3C NMR spectra of transformation products of gossypol in solutions. Chem. Nat. Comp. 2: 129-138.

Abou-Donia M.B. and Dieckert, J.W. 1974. Gossypol: Uncoupling of respiratory chain and oxidative phosphorylation. Life Sci. 14: 1955-1963.

Abou-Donia, M.B. 1976. Physiological effects and metabolism of gossypol. Residue Rev. 61: 125-160.

Abouzied, M.M., Azcona-Plivera, J.I., Yoshizawa, T. and Pestka, J.J. 1993. Production of polyclonal antibodies to the mycotoxin 4,15-Diacetylnivalenol with the carrier adjuvant cholera toxin. Appl. Environ. Microbiol. 59(5): 1264- 1268.

Achedume, A.C., Nwoha, P.U., Adepoju, A.Y. and Abu, K..A. 1994. Biochemical studies of the effect of gossypol consumption on cholesterol and residual glucose in fed and protein-energy deficient rats. Contracept. 50: 391-395.

Adams, R., Geissman, T.A., and Edwards, J.D. 1960. Gossypol, a pigment of cottonseed. Chem. Rev. 60: 555-574.

Anderson, P.A., Sneed, S.M., Skurray, G.R. and Carpenter, K.J. 1984. Measurement of lysine damage in proteins heated with gossypol. J. Agr. Food Chem. 32: 1048- 1053.

Arshami, J. and Ruttle, J.L. 1988. Effects of diets containing gossypol on spermatogenic tissues of young bulls. Theriogenology. 30(3): 507-517.

Baliga, B.P. 1956. The chemical properties and biological significance of gossypol protein complexes. Dissertation. The Agricultural and Mechanical College of Texas, College Station, Texas.

Barraza, M.L., Coppock, C.E., Brooks, K.N., Wilks, D.L., Saunders, R.G. and Laimer, G.W. Jr. 1991. Nutrition, feeding, and calves-iron sulfate and feed pelleting to detoxify free gossypol in cottonseed diets for dairy cattle. Dairy Sci. 74: 3457- 3467.

Bell, A.A. 1967. Formation of gossypol in infected or chemically irritated tissues of gossypium species. Phytopathol. 57: 759-764.

I l l

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Benz, C.C., Keniry, M. and Goldberg, H. 1988. Selective toxicity of gossypol against epithelial tumors and its detection by magnetic resonance spectroscopy. Contracept. 37: 221-228.

Benz, C.C., Lyer, S.B., Asgari, H.S., Matlin, S.A., Aronson, F.R. and Barchowsky, A. 1991. Gossypol effects on endothelial cells and tumor blood flow. Life Sci. 49(12): 67-72.

Birth, G.S. and Ramey, H.H. Jr. 1982. Near-infrared reflectance for analysis of cottonseed for gossypol. Cereal Chem. 59(6): 516-519.

Botsoglou, N.A. 1991. High-performance liquid chromatographic method for the determination of free gossypol in chicken liver. J. Chromatogr. 578: 333-337.

Bressani, R., Elias, L.G., Jarquin, R. and Brham, J.E. 1964. All-vegetable protein mixture for human feeding: effect of cooking mixtures containing cottonseed flour on free gossypol content. Food Tech. 18: 1599-1603.

Brock, T.D., Madigan, M.T., Martinko, J.M. and Parker, J. 1994. Biology of microorganisms. P. 655. Prentice-Hall, Inc., Englewood Cliffs, NJ.

Calhoun, M. C., Huston, J.E., Baldwin, B.C. Jr., Kuhlmann, S.W., Engdahl, B.S. and Bales, K.W. 1990a. Effects of cottonseed meal source and dietary crude protein on performance of early-weaned lambs: with observations on gossypol toxicity. In “Sheep and Goat, Wool and Mohair, 1990 Research Reports. PR 4790”. Texas A & M University System, College Station, TX.

Calhoun, M. C., Huston, J.E., Kuhlmann, S.W., Baldwin, B.C. Jr., Engdahl, B.S. and Bales, K.W. 1990b. Comparative toxicity of gossypol acetic acid and free gossypol in cottonseed meal and Pima cottonseed to lambs. In “Sheep and Goat, Wool and Mohair, 1990 Research Reports. PR 4779”. Texas A & M University System, College Station, TX.

Calhoun, M.C. 1996. Safe levels of cottonseed for cattle. In “Cotton Incorporated Agricultural Research Reports. Summary Reports 1996”. Cotton Incorporated, Raleigh, NC.

Calhoun, M.C. 1999. Communication in “Project Review Meeting”. Texas Agricultural Experiment Station, Texas A&M University System, San Angelo, TX.

Caskey, C. Jr. and Gallup, W.D. 1931. Changes in the sugar, oil, and gossypol content of the developing cotton boll. J. Agric. Res. 42(10): 671-673.

Cass, Q.B., Tiritan, E., Matlin, S.A. and Freire, E.C. 1991. Gossypol enantiomer ratios in cotton seeds. Photochemistry. 30(8): 2655-2657.

112

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Cater, C.M. 1968. Studies on the reaction products of gossypol with amino acids, peptides, and proteins. Dissertation. College of the Texas A & M University.

Chamkasem, K. 1988. Gossypol analysis in cottonseed oil by HPLC. J. Amer. Oil Chem. Soc. 65(10): 1601-1605.

Chang, M.C., Gu, Z.P. and Saksena, S.K. 1980. Effects of gossypol on the fertility of male rats, hamsters and rabbits. Contracept. 21: 461-469.

Chen, T., Dwyre-Gygax, C., Smith, R.S. and Breuil, C. 1995. Enzyme-linked immunosorbent assay for didecyldimethylammonium chloride, a fungicide used by the forest product industry. J. Agr. Food Chem. 43: 1400-1406.

Clark, E.P. 1927. Studies on gossypol. 1. The preparation, purification, and some of the properties of gossypol, the toxic principle of cottonseed. J. Biol. Chem. 75: 725- 739.

Conkerton, E.J. and Frampton, V.L. 1959. Reaction of gossypol with free e-amino groups of lysine in proteins. Arch. Biochem. Biophys. 81, 130-134.

Couch, J.R. and Thomas, M.C. 1976. A comparison of chemical methods for the determination of available lysine in various proteins. J. Agr. Food Chem. 24(5): 943-946.

Coulson, P.B., Snell, R.L. and Parise, C. 1980. Short term metabolic effects of the anti­ fertility agent, gossypol, one various reproductive organs of male mice. Int. J. Androl. 3: 507-518.

Crowther, J.R. 1995a. Basic immunology. Chap. 1. In “ ELISA: Theory and Practice”. Walker, J.M (Ed.). Humana Press, Totowa, New Jersey.

Crowther, J.R. 1995b. Stages in ELISA. Chap. 3. In “ ELISA: Theory and Practice”. Walker, J.M (Ed.). Humana Press, Totowa, New Jersey.

Damaty, S.M. and Hudson, B.J.F. 1979. The interaction between gossypol and cottonseed protein. J. Sci. Food Agric. 30: 1050-1056.

Dilday, R.H. 1986. Development of a cotton plant with glandless seeds, and glanded foliage and fruiting forms. Crop Sci. 26: 639-641.

Dixon, D.E. 1994. Immunoassays. Chap. 33. In “Introduction to the chemical analysis of foods”. Nielsen, S.S. (Ed.). Purdue University, West Lafayette, Indiana.

Dorsa, W.J. and Robinnette, H.R. 1982. Effects of dietary cottonseed meal and gossypol on growth of young channel catfish. Transaction of American Fisheries Society. 111:651-655. 113

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Dowd, M.K. and Thomas, L.M. 1998. The crystal and molecular structure of the gossypol triacetic acid clathrate. In “Proceedings for the 47th oilseed conference”. New Orleans.

Eagle, E. and Davies, D.L. 1958. Effect of long-term storage on acute oral toxicity and gossypol content of cottonseed pigment glands. J. Amer. Oil Chem. Soc. 35: 36- 37.

Eagle, E., Bialek, H.F., Davies, D.L. and Bremer, J.W. 1956. Biological vs. chemical evaluation of toxicity and protein quality of cottonseed meals. J. Amer. Oil Chem. Soc. 33: 15-20.

East, N.E., Anderson, M. and Lowenstine, L.J. 1994. Apparent gossypol-induced toxicosis in adult dairy goats. JAVMA. 204(4): 642-643.

Edwards, J.D. Jr. 1958. Total synthesis of gossypol. J. Amer. Chem. Soc. 80: 3798- 3800.

El-Nockerashy, A.S., Lyman, C.M. and Dollahite, J.W. 1963. The acute oral toxicity of cottonseed pigment glands and intraglandular pigments. J. Amer. Oil Chem. Soc. 40:14-17.

Erlanger, B.F. 1980. The preparation of antigenic hapten-carrier conjugates: a survey. Methods Enzymol. 70: 85-104.

Flack, M.R., Pyle, R.G., Mullen, N.M., Lorenzo, B., Wu, Y.W., Knazek, R.A. Nisula, B.C. and Reidenberg, M.A. 1993. Oral gossypol in the treatment of metastatic adrenal cancer. J. Clin. Endocrinol. Metab. 76(4): 1019-1024.

Food and Drug Administration, 1974. Food Drug Cosmetic Law. 56, 518.94, 172.894.

Gallup, W.D. 1927. The gossypol content and chemical composition of cottonseed during certain periods of development. J. Agric. Res. 34(10): 987-992.

Gallup, W.D. 1928. A chemical study of the development of cotton bolls and the rate of formation of gossypol in the cottonseed. J. Agric. Res. 36(5): 471-480.

Gardner, H.K. Jr., Hron, R.J. Sr. and Vix, H.L.E. 1976. Removal of pigment glands (gossypol) from cottonseed. Cereal Chem. 53: 549-560.

Gdaniec, W. 1994. Crystal structure of the product of a condensation reaction of (+/-)- gossypol with (R)-(+)-l-phenyethylamine. Why do diastereoisomeric diaminogossypols cocrystallize? J. Inclusion Phenom. Mol. Recognit. Chem. 17: 365-376.

114

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Gilbert, N.E., O’Reilly, J.E., Chang, C.J.G., Lin, Y.C. and Brueggemeier, R.W. 1995. Antiproliferative activity of gossypol and gossypone on human breast cancer cells. Life Sci. 57(1): 61-67.

Gonzalez-Garza, M.T., Matlin, S.A., Mata-Cardenas, B.D. and Said-Femandez, S. 1992. Further studies on the in vitro activity of gossypol as antiamebic agent. Arch. Med. Res. 23(2): 69-70.

Gonzalez-Garza, M.T., Matlin, S.A., Mata-Cardenas, B.D. and Said-Femandez, S.S. 1993. Entamoeba histolytica: Inhibition of Malic Enzyme and Alcohol Dehydrogenase by (+/-)-, (+)-, and (-)-gossypol. 24(2): 183-187.

Hadley, M.A., Lin, Y.C. and Dym, M. 1981. Effect of gossypol on reproductive system of male rats. J. Androl. 2: 190-199.

Harlow, E. and Lane, D. 1988a. Monoclonal antibodies. Chap. 6, In “Antibodies-A Laboratory Manual”. Cold Spring Harbor Laboratory.

Harlow, E. and Lane, D. 1988b. Antibody response. Chap.4, In “Antibodies-A Laboratory Manual”. Cold Spring Harbor Laboratory.

Harrison, R.O., Goodrow, M.H. and Hammock, B.D. 1991a. Competitive inhibition ELISA for the s-triazine herbicides: assay optimization and antibody characterization. J. Agr. Food Chem. 39: 122-128.

Harrison, R.O., Goodrow, M.H., Gee, S.T. and Hammock, B.D. 1991b. Hapten synthesis for pesticide immunoassay development. In “Immunoassays for Trace Chemical Analysis”. ACS symposium series 451, American Chemical Society, Washington, DC.

Haschek, W.M., Beasley, V.R., Buck, W.B. and Finnell, J.H. 1989. Cottonseed meal (gossypol) toxicosis in a swine herd. JAVMA. 195(5): 613-615.

Heller, W. 1993. Topics in the biosynthesis of plant phenols. Vol.l. In “Acta horticulturae-Intemational symposium on natural phenols in plant resistance”. Geibel, M., Treutter, D. and Feucht, W. (Eds.). Sellier Druck GmbH, Postfach 1744, 85317 Freising, Germany.

Herman, R.L. 1970. Effects of gossypol on rainbow trout salmon gairdneri Richardson. J. Fish Biol. 2: 293-303.

Hermanson, G.T. 1995. Zero-length cross-linkers. In “Bioconjugate Techniques”. Hermanson, G.T. (Ed.). Pierce Chemical Company, Rockford, IL.

115

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Hron, R.J. Sr., Kuk, M.S. and Abraham, G. 1990. Determination of free and total gossypol by high performance liquid chromatography. J. Amer. Oil Chem. Soc. 67(3): 182-187.

Hu, Y-F., Chang, C.J.G., Brueggemeier, R.W. and Lin, Y.C. 1994. Presence of antitumor activities in the milk collected from gossypol-treated dairy cows. Cancer Lett. 87: 17-23.

Hum, B.A.L. and Chantler, S.M. 1980. Production of reagent antibodies. Methods Enzymol. 70: 104-142.

Jenner, W.N. and Law, B. 1996. Immunogen preparation and purification. Chap. 3. In "Immunoassay- A Practical Guide". Law, B. (Ed.). London. Bristol, PA: Taylor & Francis.

Jones, L.A. 1979. Gossypol and some other terpenoids, flavonoids, and phenols that affect quality of cottonseed protein. J. Amer. Oil Chem. Soc. 56: 727-730.

KOhler, G. and Milstein, C. 1975. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 256: 495-497.

Kemeny, D.M. 1991. Assay optimization and trouble shooting. Chap. 7. In “A Practical Guide to ELISA”. Kemeny, D.M (Ed.). Department of Allergy Guy’s Hospital, London. Pergamon Press.

Kemmerer, A.R., Heywang, V.L., Vavich, M.G. and Sheehan, E.T. 1966. Effect of iron sulfate on egg discoloration caused by gossypol. Poultry Sci. 45: 1025-1028.

Keniry, M.A., Hollander, C. and Benz, C.C. 1989. The effect of gossypol and 6- aminonicotinamide on tumor cell metabolism: a 3lP-magnetic resonance spectroscopic study. Biochem. Biophys. Res. Commun. 164(2): 947-953.

Lane, C.F. 1975. Sodium cyanoborohydride - a highly selective reducing agent for organic functional groups. Synthesis. March, 135-146.

Lehninger, A.L., Nelson, D.L. and Cox, M.M. 1992. Amino acids and peptides. In “Principles of Biochemistry”. Lehninger, A.L., Nelson, D.L. and Cox, M.M. (Eds.). Worth Publishers.

Lin, T.S., Schinazi, R., Griffith, B.P., August, E.M., Eriksson, B.F.H., Zheng, D.K., Huang, L. and Prusoff, W.H. 1989. Selective inhibition of human immunodeficiency virus type 1 replication by the (-) but not the (+) enantiomer of gossypol. Antimicrob. Agents Chemother. 33: 2149-2151.

116

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Lin, T.S., Schinazi, R.F., Zhu, J., Birks, E., Carbone, R., Si, Y., Wu, K., Huang, L. and PrusofF, W.H. 1993. Anti-HIV -1 activity and cellular pharmacology of various analogs of gossypol. Biochem. Pharmacol. 46(2): 251-255.

Lin, Y.C., Fukaya, T., Rikihisa, Y. and Walton, A. 1985. Gossypol in female fertility control: ovum implantation and early pregnancy inhibited in rats. Life Sci. 37(1): 39-47.

Lin, Y.C., Hardley, M.A., Klingenes, D. and Dym, M. 1980. Effects of gossypol on the reproductive system of male rats. Biol. Reprod. 22: 95A.

Lindsey, T.O., Hawkins, G.E. and Guthrie, L.D. 1980. Physiological responses of lactating cows to gossypol from cottonseed meal rations. J. Dairy Sci. 63(4): 562- 573.

Longmore, J. 1886. Cotton-seed oil: Its colouring matter and mucilage, and description of a new method of recovering the loss occurring in the refining process. J. Soc. Chem. Ind. (London) 5: 200-206.

Lusas, E.W. and Jividen, G.M. 1987. Glandless cottonseed: a review of the first 25 years of processing and utilization research. J. Amer. Oil Chem. Soc. 64(6): 839- 854.

Lyman, C. M., Baliga, B.P. and Slay, M.W. 1959. Reaction of proteins with gossypol. Arch. Biochem. Biophys. 84: 486-497.

Margalith, P. 1967. Inhibitory effect of gossypol on microorganisms. Appl. Microbiol. 15(4): 952-953.

Markman, L. 1968. Gossypol Derivatives. Greenberg, D. (Ed.). The U. S. Department of Agriculture and the National Science Foundation, Washington, D. C.

Martinez, W.H., Frampton, V.L. and Cabell, C.A. 1961. Effects of gossypol and raffinose on lysine content and nutritive quality of proteins in meals from glandless cottonseed. J. Agr. Food Chem. 9(1): 64-66.

Matlin, S.A., Belenguer, A., Tyson, R.G. and Brookes, A.N. 1987. Resolution of gossypol: analytical and large-scale preparation HPLC on non-chiral phases. J. High Resolu. Chromatogr Chromatogr. Commun. 10: 86-91.

Matlin, S.A., Zhou, R., Bialy, G., Blye, R.P., Naqvi, R.H. and Lindberg, M.C. 1985. (-)-Gossypol: an active male antifertility agent. Contracept. 31(2): 141-149.

Menaul, P. 1923. The physiological effect of gossypol. J. Agric. Res. 26: 233-237.

117

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Montamat, E.E., Burgos, C., Gerez De Burgos, N.M., Rovai, L.E. and Blanco, A. 1982. Inhibitory action of gossypol on enzymes and growth of trypanosoma cruzi. Sci. 218:288-289.

Morgan, S.E. 1989. Gossypol as a toxicant in livestock. Vet. Clin. North. Am. Food Anim. Pract. 5(2): 251.

Muzaffaruddin, J. and Saxena, E.R. 1966. Physicochemical studies on the composition and stability of metal-gossypol complexes. 1. Fe-H-+ gossypol complex. J. Amer. Oil Chem. Soc. 43: 429-430.

Nakamura, R.M., Voller, A. and Bidwell, D.E. 1986. Handbook of experimental immunology. Chap. 27. In “Immunochemistry”. Weir, D.M., Herzengberg, L.A. and Blackwell, C. (Eds.). Blackwell Scientific Publication, Oxford.

National Coordinating Group on Male Antifertility Agents. 1978. Gossypol-a new antifertility agent for males. Chin. Med. J. 4(6): 417-428.

Nomeir, A.A. and Abou-Donia, M.B. 1982. Gossypol: high-performance liquid chromatographic analysis and stability in various solvents. J. Amer. Oil Chem. Soc. 59(12): 546-549.

Norries, K.H., Bames, R.F., Moore, J.E. and Shenk, J.S. 1976. Predicting forage quality by infrared reflectant spectroscopy. J. Animal Sci. 43(4): 889-897.

Nwoha, P.U. 1995. The blood constituents of gossypol-treated, protein-malnourished wistar rats. Contracept. 52: 249-254.

Official Methods and Recommended Practices of the American Oil Chemists’ Society. 1987. Free gossypol. Ba 7-58. Firestone D. (Ed.). American Oil Chemists’ Society. 1608 Broadmoor, Champaign, Illinois 61821.

Official Methods and Recommended Practices of the American Oil Chemists’ Society. 1987. Total gossypol. Ba 8-78. Firestone D. (Ed.). American Oil Chemists’ Society. 1608 Broadmoor, Champaign, Illinois 61821.

Phillip, V.A. and Hedin, P.A. 1990. Spectral techniques for the structural analysis of the cotton terpenoid aldehydes gossypol and gossypolone. J. Agr. Food Chem. 38: 525-528.

Plhak, L.C. and Spoms, P. 1994. Development and production of monoclonal antibodies for the measurement of solanidine potato glycoaikaloids. Am. Potato J. 71:297-313.

Pons, W.A. Jr., Hoffpauir, C.L. and O’Connor, R.T.T. 1951. Determination of total gossypol pigments in cottonseed oils. J. Amer. Oil Chem. Soc. 28: 8-12.

118

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Poore, M. and Rogers, G.M. 1998. Potential for gossypol toxicity when feeding whole cottonseed, http://www2.ncsu.edu/cal s/an-sci/extension.old/animal/nutri/mhp95- l.htm.

Raju, P.K. and Cater, C.M. 1967. Gas-liquid chromatographic determination of gossypol as the trimethylsilyl ether derivative. J. Amer. Oil Chem. Soc. 6: 465- 466.

Ramaswamy, H.N. and O’Connor, R.T. 1968. Physical and chemical properties of selected metal complex of gossypol. J. Amer. Oil Chem. Soc. 45: 841-844.

Rao, P.N., Wang, Y.C., Lotzva, E., Khan, A.A., Rao, S.P. and Stephens, L.C. 1985. Antitumor effects of gossypol on murine tumors. Cancer Chemother. Pharmacol. 15:20-25.

Reddy, I.M. and Rao, M.S.N. 1987. Effect of chemical modification on the binding of gossypol by gossypin (11s protein) and congossypin (7s protein) of cottonseed. J. Biosci. 12(3): 203-209.

Reiser, R. and Fu, H.C. 1962. The mechanism of gossypol detoxification by ruminant animals. J. Nutr. 76: 215-218.

Rittenburg, J.H. 1990. Fundamental of immunoassay. Chap. 2. In “A Development and Application of Immunoassay for Food Analysis”. Rittenburg, J.H. (Ed.).

Rodbard, D. 1981. Mathematics and statistics of ligand assays: an illustrated guide. In “Ligand Assay”. Langan, J. and Clapp, J.J. (Eds.). Masson Publishing, New York.

Rosenberg, L.J., Adlakha, R.C., Desai, D.M. and Rao, P.N. 1986. Inhibition of DNA polymerase a by gossypol. Biochim. Biophys. Acta. 866: 258-267.

Royer, R.E. and Vander Jagt, D.L. 1983. Gossypol binds to a high-affinity binding site on human serum albumin. FEBS Lett. 157(1): 28-30.

Scheiffele, E.W. and Shirley, D.A. 1964. The oxidation of gossypol. 1. Early stages in the reaction of gossypol and oxygen. J. Org. Chem. 29(12): 3617-3620.

Scott, P.M. and Trucksess, M.W. 1997. Application of immunoaffinity columns to mycotoxin analysis. JAOAC. Int. 80: 941-949.

Shandilya, L., Clarkson, T.B., Adams, M.R. and Lewis, J.C. 1982. Effect of gossypol on reproductive and endocrine functions of male cynomolgus monkeys. Biol. Reprod. 27: 241-252.

119

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Skerritt, J.H. 1995. Analytical aspects of immunoassay of agrochemicals. Chap.l. In "New frontiers in agrochemical immunoassay". Kurtz, D.A., Skerritt, J.H. and Stanker, L. (Eds.). AOCS International. Arlington, VA.

Spoms, P. and Abell, D.C. 1996. MALDI mass spectrometry for food analysis. Trends in food science & technology. 7: 187-190.

Stansbury, M.F., Cucullu, A.F. and Hartog, G.T.D. 1954. Cottonseed content variation: influence of variety and environment on oil content of cottonseed kernels. J. Agr. Food Chem. 2(13): 692-696.

Stenberg, M. and Nygren, H. 1988. Kinetics of antigen-antibody reactions at solid- liquid interfaces. J. Immunol. M. 113:3-15.

Stipanovic, R.D., Altman, D.W., Begin, D.L., Greenblatt, G.A. and Benedict, J.H. 1986. Effects of cotton terpenoid aldehydes on resistance to Heliothis. International Union of Pure and Applied Chemistry, Sixth Int. Congr. Pest. Chem. Ottawa, Canada, Abstract No. 2D/E-05.

Stipanovic, R.D., Altman, D.W., Begin, D.L., Greenblatt, G.A. and Benedict, J.H. 1988. Terpenoid aldehydes in upland : analysis by aniline and HPLC methods. J. Agr. Food Chem. 36: 509-515.

Stipanovic, R.D., Bell, A.A. and Howell, C.R. 1973. Spectral identification of the ketol tautomer of gossypol. J. Amer. Oil Chem. Soc. 50: 462-463.

Stipanovic, R.D., Bell, A.A., O'Brien, D.H. and Lukefahr, M.J. 1977. Heliocide H 2 : an insecticidal sesterterpenoid from cotton (Gossypium). Tetrahedron Lett. 6: 567- 570.

Stipanovic, R.D., Donovan, J.C., Bell, A.A. and Martin, F.W. 1984. Factors interfering in gossypol analysis of okra and glandless cottonseed using direct aniline extraction. J. Agr. Food Chem. 32: 809-810.

StrOm-Hansen, T., Cornett, C. and Jaroszewski, J. W. 1989. Interaction of gossypol with amino acids and peptides as a model of enzyme inhibition. Int. J. Peptide Protein Res. 34: 306-310.

Szurdoki, F., Bekheit, H.K.M., Marco, M.P., Goodrow, M.H. and Hammock, B.D. 1995. Important factors in hapten design and enzyme-linked immunosorbent assay development. Chap.4. In "New frontiers in agrochemical immunoassay". Kurtz, D.A., Skerritt, J.H. and Stanker, L. (Eds.). AOCS International. Arlington, VA.

The United States Department of Agriculture. 1998a. Table 5. Major oilseeds. http://fFas.usda.gov/cotton/circular/98-03/mar98oDdtl.htm

120

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. The United States Department of Agriculture. 1998b. http://ffas.usda.gov/oilseeds/ circular/98-03/

Ueno, H., Sahni, M.K., Segal. S.J. and Koide, S.S. 1988. Interaction of gossypol with sperm macromolecules and enzymes. Contracept. 37(3): 333-341.

Ullrey, D.E. 1966. Requirements, toxic levels and interrelationships of minerals for swine and poultry as related to dietary gossypol. Inactivation of gossypol with mineral salts. In “Proceedings of Proc. Conf’. Nat. Cotton Prod. Assoc., New Orleans, LA.

Van Soest, P.J. 1982. Rumen microbes. In “ Nutritional ecology of the ruminant: ruminant metabolism, nutritional strategies, the cellulolytic fermentation and the chemistry of forages and plant fibers.” Chap. 10. In “Part IV: Gastrointestinal Fermentations”. Comstock Publishing Associates. Ithaca and London.

Vogt, R.F. Jr., Phillips, D.L., Henderson, L.O., Whitfield, W. and Spierto, F.W. 1987. Quantitative differences among various proteins as blocking agents for ELISA microtiter plates. J. Immunol. M. 102: 43-50.

Waller, D.P., Fong, H.H.S., Cordell, G.A. and Soejarto, D.D. 1984. Antifertility effects of gossypol and its impurities on male hamsters. Contracept. 23: 653-660.

Wan, P.J. 1999. Communication in “Project Review Meeting”. The United States Department of Agricultural Research Service, Southern Regional Research Center, New Orleans, LA.

Wang, Y.C. and Rao, P.N. 1984. Effect of gossypol on DNA synthesis and cell cycle progression of mammalian cells in vitro. Cancer Res. 44: 35-38.

Whaley, K.J., Sampath, D.S. and Balaram, P. A. 1984a. Circular dichroism study of (+) gossypol binding to proteins. Biochem. Biophys. Res. Commun. 121(3): 953-959.

Whaley, K.J., Sampath, D.S. and Balaram, P. 1984b. Optically active gossypol as a circular dichroism probe of interactions with serum albumins. Biochim. Biophys. Acta. 801:127-130.

Wu, Y.W., Chik, C.L. and Hnazek, R.A. 1989. An in vitro and in vivo study of antitumor effects of gossypol on human SW-13 adrenocortical carcinoma. Cancer Res. 49: 3754.

Yannai, S. and Bensal, D. 1983. Gossypol in cottonseed products: toxicology and inactivation. Toxicology in the use, misuse and abuse of food, drugs and chemicals. Arch. Toxicol. Suppl. 6: 167-174.

121

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Yu, Y.W. 1987. Probing into the mechanism of action, metabolism and toxicity of gossypol by studying its (+)- and (-)- stereoisomers. J. Ethnopharmacol. 20: 65- 78.

Yuan, Y.Y., Shi, Q.X. and Srivastava, P.N. 1995. Inhibition of rabbit sperm acrosomal enzymes by gossypol. Mol. Reprod. Dev. 40: 228-232.

Zhou, R.H. and Lin, X.D. 1988. Isolation of (-)-gossypol from natural plant. Contracept. 37(3): 239-245.

Zola, H. 1987(a). Making hybridomas. Chap. 3. In “Monoclonal Antibodies: A Manual of Techniques”. CRC Press, Inc. Boca Raton, Florida.

Zola, H. 1987(b). Monoclonal antibodies: production, purification, analysis, quality control, storage and distribution. Chap. 4. In “Monoclonal Antibodies: A Manual of Techniques”. CRC Press, Inc. Boca Raton, Florida.

122

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. APPENDIX I

Competition after 1st cloning using different gossypol derivatives (250 pg/ml of gossypol equivalent) as competitors

Cell line Gossypol-derivatives A/Ao Lysine-g 0.181/0.958 Arginine-g 0.198/0.958 6G7D11 Amino-propanol-g 0.161/0.958 Ethanolamine-g 0.151/0.958

Lysine-g 0.157/0.488

Arginine-g 0.141/0.488 6G7D2 Amino-propanol-g 0.150/0.488 Ethanolamine-g 0.153/0.488 Lysine-g 0.158/0.400 Arginine-g 0.177/0.400 6G7D7 Amino-propanol-g 0.153/0.400

Ethanolamine-g 0.156/0.400 Lysine-g 0.161/0.393

6G7G3 Arginine-g 0.193/0.393 Amino-propanol-g 0.148/0.393 Ethanolamine-g 0.152/0.393 Lysine-g 0.162/0.404

6G7B6 Arginine-g 0.185/0.404 Amino-propanol-g 0.167/0.404 Ethanolamine-g 0.169/0.404 Lysine-g 0.142/0.325

6G7D12 Arginine-g 0.175/0.325 Amino-propanol-g 0.149/0.325 Ethanolamine-g 0.168/0.325 (table cont.)

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Lysine-g 0.268/2.506

5D7D3 Arginine-g 0.333/2.506 Amino-propanol-g 0.198/2.506 Ethanolamine-g 0.186/2.506 Lysine-g 0.285/2.775

5D7E3 Arginine-g 0.256/2.775 Amino-propanol-g 0.199/2.775 Ethanolamine-g 0.186/2.775 Lysine-g 0.170/0.778

5D7F3 Arginine-g 0.155/0.778 Amino-propanol-g 0.154/0.778 Ethanolamine-g 0.149/0.778 Lysine-g 0.252/1.951

5D7E4 Arginine-g 0.223/1.951 Amino-propano 1-g 0.205/1.951 Ethanolamine-g 0.192/1.951 Lysine-g 0.274/1.964

5D7F4 Arginine-g 0.273/1.964 Amino-propanol-g 0.182/1.964 Ethanolamine-g 0.218/1.964 Lysine-g 0.282/1.736

5D7G4 Arginine-g 0.271/1.736 Amino-propanol-g 0.207/1.736 Ethanolamine-g 0.228/1.736 (table cont.)

124

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. Lysine-g 0.182/0.239

7G2A5 Arginine-g 0.173/0.239 Amino-propanol-g 0.160/0.239 Ethanolamine-g 0.171/0.239 Lysine-g 0.178/0.280

7G2B5 Arginine-g 0.178/0.280 Amino-propanol-g 0.175/0.280 Ethanolamine-g 0.178/0.280 Lysine-g 0.189/0.407

7G2A6 Arginine-g 0.181/0.407 Amino-propanol-g 0.179/0.407 Ethanolamine-g 0.184/0.407 7G2F2 Arginine-g 0.131/0.644 5H9F8 Arginine-g 0.136/0.344 Note: g represents gossypol.

125

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. APPENDIX II

Peak results of HPLC (high performance liquid chromatography) for (+)- and (-)- gossypol isomers

Name Area Purity (%) (+)-gossypol: 80089 (-)- 99.63 gossypol (-)-gossypol: 22017381 (+)-gossypol: 9784542 (+)- 99.96 gossypol (-)-gossypol: 3802

126

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. APPENDIX III

I5 0 values for gossypol and gossypol derivatives

I 5 0 Values (pg/ml)

Gossypol derivatives MAb

PAb 7G2F2G3C 6G7D11F10 6G7D11F 6 F2F8 10F2H3

Lysine-gossypol 0.197 28.31 9.93 8.17

Arginine-gossypol 0.209 21.51 8.41 8.10

2-amino-1 -propanol-gossypol 0.195 55.05 21.97 18.63

3-amino-1 -propanol-gossypol 0.174 20.86 26.25 23.99

Ethanolamine-gossypol 0.225 22.75 10.96 7.38

fresh made gossypol >250 >250 >250 >250

21 day old gossypol 1.59 >250 >250 >250

Glutamine-gossypol >125 - --

T yrosine-gossypol >125 - --

T ryptophane-gossypol >125 - --

Para-amino-benzoic acid- >125 - - - gossypol

(+)-gossypol-lysine - - 12.72 10.64

(-)-gossypol-lysine - - 13.47 9.81

(+)-gossypol-2-amino-1 - - - 9.08 7.44 propanol

(-)-gossypol-2-amino-1 - - - 9.53 9.09 propanol

127

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. APPENDIX IV

Comparison of AOCS and ELISA methods for FG analysis

Sample ID* FG% (AOCS:x) FG% (ELISAry)

2241 0.08 0.35

2817 0.33 0.69

2822 0.36 0.9

2819 0.91 2.0

2820 0.93 1.95

2814 1.06 1.94

2813 1.12 2.45 * samples obtained from M. Calhoun (Texas A&M University) and sample ID number assigned in the Nutrition/Toxicology Laboratory at the Texas A&M University Agricultural Center, San Angelo, TX. FG (x) measured by M. Calhoun (Texas A&M University) and FG (y) measured by ELISA method.

128

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. VITA

The author was bom in Shandong Province, People’s Republic of China, on

August 27, 1968. She graduated from the Department of Cereal and Oil Science and

Engineering, Wuxi Institute of Light Industry, with a bachelor’s degree, in 1991. As a

top ranking student, she was recommended for further study as a graduate student in the

same department under Professor Hongsun Diao. Her study area included oilseed

processing, oil modification, unit design, and oilseed by-product utilization (including

vitamin E, tannin, phytic acid, oryzanol and gossypol). In 1995, she was admitted as a

doctoral candidate under Dr. Leslie C. Plhak in the Department of Food Science,

Louisiana State University, with the research on compound analysis using

immunochemical methods. Currently, she is studying in Food Science for the degree of

Doctor of Philosophy, which will be awarded in August 1999.

129

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission. DOCTORAL EXAMINATION AND DISSERTATION REPORT

Candidate: Xi Wang

Major Pield: Food Science

Title of Dissertation: Development of Immunochemical Methods for Gossypol Analysis

Approved:

Mai Professor

( ^ / Dead 'of the"Graduate School

EXAMINING COMMITTEE:

Date of Examination:

April 29, 1999

Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.