:h‘fl. .r. ,2 . . u an... . ".3: {1.91.}, 1|. 713.74- at: I vii-74‘!“ I” 1 I30. I." 1:63.: £7.an Rump—fix (AQQQ) HIGAN S ATE LB S " Illlmulliifim minimliu'iifiix‘tutmmtl \ 1 3129301751 1 47 This is to certify that the dissertation entitled Recombinant Antibodies and Peptide Mimotopes to FusariumiMycotoxins presented by Qiaoping Yuan has been accepted towards fulfillment of the requirements for Ph.D. degree in Botany and Plant Pathology - Environmental Toxicology \ Major professor new 9%” MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 LIBRARY Michigan State University PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINE return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE ma clumps-m4 RECOMBINANT ANTIBODIES AND PEPTIDE MIMOTOPES TO FUSARIUM MYCOTOXINS By Qiaoping Yuan A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Botany and Plant Pathology, and Institute of Environmental Toxicology 1998 ABSTRACT RECOMBINANT ANTIBODIES AND PEPTIDE MIMOTOPES TO FUSARIUM MYCOTOXINS By Qiaoping Yuan Zearalenone, fumonisins and trichothecene mycotoxins are a group of secondary metabolites produced by toxigenic strains of Fusarium fungi. These mycotoxins are toxic to both human and animals; Some are also phytotoxic. Modern immunology and molecular biology provide novel approaches to develop low-cost antibodies and alternative antigens with desirable affinity and specificity for mycotoxin detection, neutralization, and detoxification as well as the improvement in plant resistance to mycotoxin associated plant diseases. In this thesis, phage display technology was explored to develop recombinant antibodies and peptide mimotopes for Fusarium mycotoxins. Firstly, the heavy and kappa light- chain variable region genes of an anti-zearalenone hybridoma cell line were isolated by polymerase chain reaction and joined by a DNA linker encoding peptide (Gly4Ser)3 as a single-chain Fv (scFv) DNA fragment. The scFv DNA fragment was cloned and expressed as a fusion protein with an antibody recognition sequence (E- tag) and phage M13 coat protein 93p in Escherichia coli TG1. To facilitate the cloning of genes for variable heavy chain, variable kappa light chain, and the assembly of scFv DNA fragment, a new phage display vector, pEY.5, was also constructed. Through affinity panning-elution, high affinity scFv phage antibodies were enriched and selected. Nucleotide sequence analyses revealed that the VH portion of the cloned scFv antibody contributed more to the specificity than the VK portion. The potential of using the soluble scFv antibody for routine screening of zearalenone and its analogues was demonstrated. With a similar approach, attempts were made to generate scFv antibodies from mice immunized with fumonisin B,-cholera toxin conjugate immunized mice. However, no specific scFv antibody clone was selected. An alternative approach was suggested for the cloning of functional FB1 specific scFv antibodies. Secondly, the anti-zearalenone scFv DNA fragment was cloned into a plant transformation vector and transferred into Ambidopsis immature seeds by vacuum infiltration via Agrobacterium tumefaciens mediated plant transformation. Functional anti-zearalenone scFv plantibody was expressed. The plantibody exhibited a high zearalenone-binding affinity comparable to bacteria-produced scFv antibody and the parent monoclonal antibody. Finally, from a phage displayed random 7-mer peptide library, two highly homologous heptapeptide sequences, SWGPFPF and SWGPLPF, were identified that specifically bound a deoxynivalenol-specific monoclonal antibody. In contrast to deoxynivalenol’s toxic effects, the peptide did not elicit apoptosis in bone marrow cells, neither did it inhibit protein synthesis in a cell-free translation system. Rather, the peptide mimotope showed an antagonism to deoxynivalenol-induced protein synthesis inhibition. These peptide mimotopes were also evaluated as immunochemical reagents for deoxynivalenol immunoassay. To my dear parents, my wife, and my lovely twin daughters iv ACKNOWLEDGMENTS I am greatly grateful to my major advisor, Dr. L. Patrick Hart, for his advice, guidance, encouragement, assistance, and understanding throughout the development of this dissertation. I am also indebted to Dr. James J. Pestka, Dr. John E. Linz, and Dr. Dennis W. Fulbright for their invaluable suggestions, advice, and for being members of my graduate guidance committee. I wish to thank Dr. Hui-Ren Zhou, Dr. James R. Clarke, Ms. Rebecca Uzarski, Mr. Wenqi Hu, and many others from Dr. Hart’s and Dr. Pestka’s laboratories for their invaluable suggestions, practical help and friendship. Acknowledgment also goes to Dr. Shenyang He and his students for their help with plant transformation and to Mr. Frank E. Klein at the Neogen Company for his help in making deoxynivalenol conjugates. Most importantly, I would like to thank my wife Youlan Chen, and my daughters, Jing Yuan and Lin Yuan, for their love, support, patience, and understanding during all of these years. I am also very grateful to my father, Junxi Yuan, my mother, Meichun Xiang, my brother Xuewen Yuan and my sister Hongxia Yuan, for their continuous love and support in my life. TABLE OF CONTENTS LIST OF TABLES ....................................................................................................... x LIST OF FIGURES .................................................................................................... xi LIST OF SYMBOLS AND ABBREVIATIONS .......................................................... xvii INTRODUCTION .................................................................................................................... 1 PART I. LITERATURE REVIEW ............................................................................. 4 MYCOTOXINS ...................................................................................................... 5 Zearalenone .................................................................................................... 6 Fumonisins .................................................................................................... 12 Tn'chothecenes .............................................................................................. 16 Analytical methods for mycotoxin detection .................................................. 19 RECOMBINANT ANTIBODIES AND PEPTIDE MIMOTOPES ........................... 22 Native antibody .............................................................................................. 22 Recombinant antibody and phage display ..................................................... 23 Peptide mimotopes ........................................................................................ 29 PLANTIBODY ..................................................................................................... 31 PARTII. MOLECULAR CLONING. EXPRESSION AND CHARACTERIZATION OF A FUNCTIONAL SINGLE-CHAIN Fv (scFv) ANTIBODY TO THE MYCOTOXINZEARALENONE ................... 36 ABSTRACT ......................................................................................................... 37 INTRODUCTION ................................................................................................. 39 MATERIALS AND METHODS ............................................................................ 41 Materials, strains and general methods ......................................................... 41 mRNA isolation .............................................................................................. 43 cDNA synthesis, PCR amplification and scFv cloning ................................... 43 ScFv antibody expression and panning-elution selection .............................. 46 Competitive indirect ELISA (Cl-ELISA) ........................................................ 48 SDS-PAGE and Western blot analysis .......................................................... 50 Sequence analysis ........................................................................................ 51 CI-ELISA for spiked corn extracts ................................................................ 51 Nucleotide sequence accession numbers ..................................................... 52 vi PAI RESULTS ......................................................................................................... 53 PCR amplification .......................................................................................... 53 Phage display of scFv antibody and panning-elution selection ..................... 55 Soluble scFv antibody production and characterization ................................ 56 SDS-PAGE and Western blot analyses ........................................................ 60 Sequence analysis ........................................................................................ 60 Application of scFv OY1 .5toa spiked corn extract ....................................... 64 DISCUSSION ..................................................................................................... 67 PART III. ATTEMPTS TO CLONE SINGLE-CHAIN FV (SCFV) ANTIBODIES AGAINST FUMONISIN B1 FROM IMMUNIZED MICE ............................. 71 ABSTRACT ......................................................................................................... 72 INTRODUCTION ................................................................................................. 73 MATERIALS AND METHODS ............................................................................ 76 General procedures ....................................................................................... 76 Preparation of immunogens .......................................................................... 76 Animal immunization ..................................................................................... 77 ELISA ............................................................................................................ 79 Spleen mRNA isolation and cDNA synthesis ................................................ 80 PCR amplification and scFv cloning .............................................................. 80 Affinity selection of phage-displayed anti-PB, scFv antibodies ...................... 81 RESULTS AND DISCUSSION ............................................................................ 84 Mouse immunization and polyclonal antibodies ............................................ 84 Construction of phage-displayed scFinbrary ................................................ 86 Affinity selection of phage-displayed scFv antibodies ................................... 88 CONCLUSION .................................................................................................... 92 PART IV. EXPRESSION OF A FUNCTIONAL ANTI-ZEARALENONE SINGLE-CHAIN FV ANTIBODY IN TRANSGENIC ARABIDOPSIS PLANTS .................................................................................................. 93 ABSTRACT ......................................................................................................... 94 INTRODUCTION ................................................................................................. 95 METHODS AND MATERIALS ............................................................................ 97 General .......................................................................................................... 97 Constmction of scFv cloning vector ............................................................... 97 vii Anti-zearalenone scFv antibody cloning in pEY.5 ........................................ 99 Construction of plant scFv expression plasmids ........................................... 99 Plant transformation ................................................................................ 100 Expression of scFv plantibody and ELISA analysis ..................................... 101 Protein analysis ........................................................................................... 102 ScFv antibody cytolocation with immunogold labeling ................................. 103 RESULTS .......................................................................................................... 105 Cloning of anti-zearalenone scFv in pEY.5 ................................................. 105 Expression of anti-zearalenone scFv plantibody ......................................... 107 The effects of methanol and low pH on plantibody ..................................... 1 1 1 Localization of anti-zearalenone scFv plantibody ........................................ 113 DISCUSSION .................................................................................................... 115 PART V. IDENTIFICATION OF MIMOTOPE PEPTIDES WHICH BIND TO THE MYCOTOXIN DEOXYNIVALENOL-SPECIFIC MONOCLONAL ANTIBODY ............................................................... 1 19 ABSTRACT ....................................................................................................... 120 INTRODUCTION ............................................................................................... 122 MATERIALS AND METHODS .......................................................................... 126 Reagents ..................................................................................................... 126 Phage-display peptide library and amplification .......................................... 128 Phage selection by panning-elution selection ............................................. 128 Titration and amplification of panning-elution selected phage ..................... 131 Competitive ELISA with phage-displayed peptide ...................................... 131 DNA sequencing ......................................................................................... 132 Translational fusion with bacterial alkaline phosphatase (AP) .................... 133 CD-ELISA with DONPEP—AP fusion protein ................................................ 134 CD-ELISA using peptide C430 and its HRP conjugate ............................... 134 Immunization of animals with C430-BSA conjugate .................................... 135 Cytotoxicity and effects on protein synthesis in vitm ................................... 135 RESULTS .......................................................................................................... 138 Panning-elution selection of specific phage ............................................... 138 Application of DON mimotope peptide in competitive ELISA ...................... 140 CD—ELISA for DON spiked wheat extract ................................................... 145 Immunization with C430-BSA conjugate ..................................................... 147 Cytotoxicity and effects on protein synthesis in vitro .................................. 147 DISCUSSION .................................................................................................... 152 viii SUMMARY ............................................................................................................. 1 58 APPENDICES: BUFFERS AND MEDIA ................................................................ 159 APPENDIX A: COMMONLY USED BUFFERS ............................................. 160 APPENDIX B: COMMONLY USED MEDIA ................................................. 163 LIST OF REFERENCES ....................................................................................... 164 ix Table 1.1. Table 1.2. Table 2.1. Table 2.2. Table 4.1 . Table 5.1. LIST OF TABLES The genes and proteins of fd bacteriophage ...................................... 25 Antibody-derived molecules produced in transgenic plants ................ 35 Sequences of oligonucleotide PCR primers for PCR amplification of V", VK, and linker DNA ............................................. 45 Comparison of cross-reactivities between scFv antibody and mAb toward zearalenone analogues ................................................. 59 Effect of pH environment on antibody binding activity ...................... 1 12 50% binding inhibition of DON-HRP and C430-HRP by DON and C430 in competitive ELISA with monoclonal antibody 6F5 coated in microtiter wells ............................................................... 143 LIST OF FIGURES Figure 1.1. Structures of zearalenone and analogues showing structural similarities with estradiol .................................................................... 7 Figure 1.2. Structures of known fumonisins and Altemaria altemata f. sp. lycopetsici (AAL) toxin, showing stmctural similarities with sphingosine (Norred, 1993) ............................................................ 14 Figure 1.3. Structure and numbering system of some naturally identified trichothecene mycotoxins (modified from Ueno, 1983) .................. 17 Figure 1.4. Antibody model structures showing subunit composition and domain distribution along the polypeptide chains. Fragments generated by proteolytic cleavage and [or recombinant technology appear in the shaded area (Phannacia Biotech) .......... 24 Figure 2.1. Preparation of zearalenone immunogens through oxime chemistry (based on Thouvenot and Morfin, 1983) ......................... 42 Figure 2.2. Panning-elution selection procedure for anti-zearalenone scFv phage in zearalenone-ovalbumin (ZEN-OVA) coated Immunlon- 4 microtitetwells .............................................................................. 47 Figure 2.3. VH and VK amplification and assembly of scFv. Lanes are: (1): VH marker (pUC18NH, EcoR I/Xba I digested, Pharrnarcia Biotech); (2): VH PCR product; (3): VK PCR product; (4): Linker PCR product; (5): Linker-VK PCR product; (6): VH-Linker-VK (scFv) PCR product; (7): scFv marker (pUC18/A10B, Sfi l/ Not I digested, Pharrnacia Biotech) ...................................................... 54 Figure 2.4. pQY1 construct. PCR amplified scFv was cloned into Sfil and Not I sites of pCANTAB5E, a pUC119 based vector from Phannacia Biotech. The linker DNA sequence was 5'GGTGGAGGCGG‘I'I'CAGGCGGAGGTGGCTCTGGCGGTG GCGGATCG-3', which encoded a short peptide [(Gly,Ser)3] ....................................................................................... 55 Figure 2.5. Zearalenone-binding activities of recombinant phage following each round of panning-elution selection measured by Cl-ELISA in zearalenone-ovalbumin coated Immulon-4 microtiter wells. Bound phages were detected by horseradish peroxidase conjugated sheep anti-M13 antibody .............................................. 57 xi Figure Figure Figure Fbum I FIgUfI Figure Figure 2.6. Sensitivity of soluble scFv antibodies tested in CI-ELISA in zearalenone-ovalbumin coated Immulon-4 microtiter wells. QY1.1 and QY1.2 ( no dilution) were from preselection; QY1.5 (5x dilution) was from first round of panning-elution selection; QY1.12 (10 x dilution) was from second round of panning- elution selection; mAb (supernatant of hybridoma cell culture) was diluted 5 times. Bound soluble scFv antibodies were detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse IgG. A: Absorbance reading at 405 nm. B: Percent of binding inhibition determined by dividing sample absorbance by control absorbance x 100 ............................................................................................. 58 Figure 2.7. SDS-PAGE gel and Western blot analysis of scFv expression. (A): Supematants of QY1 .1 (lane 1; 0.3 ml), QY1.2 (lane 2; 0.3 ml), QY1.12 (Iane3; 0.3 ml) and QY1.5 (lane 4; 0.6 ml) concentrated in 10% trichloroacetic acid were subjected to SDS- PAGE and stained with Coomassie brilliant blue G-250. (B): Western blot ofduplicate SDS/PAGE gel ......................................... 61 Figure 2.8. Deduced amino acid sequences of VH and VK in scFv QY1.5. Complementary-determining regions (CDR) and framework regions (FW) are indicated ................................................................ 63 Figure 2.9. Zearalenone sensitivities in spiked corn extracts with monclonal antibody and soluble scFv antibody QY1.5 measured by CI- ELISA in zearalenone-keyhole limpet hemocyanin conjugate coated lmmulon-4 microtiter wells. Both scFv QY1.5 and mAb were diluted 1:4 in PBS containing 10% nonfat dry milk. Zearalenone in ground corn was extracted with 5 volumes (wt/vol) of methanol-water (70:30) and the extracts were then diluted 1:1 with PBS .......................................................................... 65 Figure 2.10. Effects of methanol on the binding of mAb and scFv QY1.5 to zearalenone-keyhole limpet hemocyanin conjugate. lmmulon-4 microtiter wells were coated with zearalenone-keyhole limpet hemocyanin conjugate. Bound soluble scFv antibody was detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse IgG. Bound monoclonal antibody was detected by horseradish peroxidase conjugated goat anti-mouse IgG .......................................................................... 66 Figure 3.1. Preparation of fumonisin B1 conjugates with glutaraldehyde linkage via a free amino group (Pestka et al., 1995) ........................ 78 xii Fig Fig Fig Fic Fig Figure 3.2. Polyclonal anti-FB, antibody production from mice immunized with FB1-cholera toxin (FB1-CT) and FB1-keyhole limpet hemocyanin (FB,-KLH) conjugates (Each group had one representative out of eight mice). Sera were obtained one week after the second boost with the immunogens. A: Titration of antisera measured in FB,-ovalbumin (FB,-OVA) conjugate coated Immulon-4 microtiter wells. Bound mouse antibodies were detected by horseradish peroxidase conjugated goat anti- mouse lgG. B: Competitive indirect ELISA for FB1 with the antisera (diluted 1:60.000 and 1:3,200 respectively for FB,-KLH and FB,-CT immunized mice) ........................................................... 85 Figure 3.3. PCR amplification of VH and VK genes from mouse splenic mRNA and the assembly of scFv. Lanes are: (1): Linker PCR product; (2): VK PCR product; (3): VH PCR product; (4): Linker- VK PCR product; (5):VH-Linker-VK (scFv) PCR product; Lane M: 100-bp ladder DNA marker (Gibco BRL) ............................................ 87 Figure 3.4. Phage titers after panning-elution selection of the phage library. Standard procedure was: coating: 1 pg/well of FB,-OVA conjugates or OVA alone (in control wells); phage incubation: 60 min; bound phage elution: by PBS or 100 jig/"II of FB1 in PBS. Modifications were different conjugates for coating (A), phage incubation time (B), and FB1 concentration for elution (C) .............. 89 Figure 4.1. Map of phage-display vector pEY.5. (A). Restriction map with unique restriction sites marked. (B). The nucleotide sequence of linker (bold sequence) and the VH and VK cloning sites (underlined)in pEY.5 ............................................................... 98 Figure 4.2. Sensitivity and specificity of anti-zearalenone scFv antibodies measured by competitive indirect ELISA using zearalenone- ovalbumin coated wells. Bound soluble scFv antibody was detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse IgG. EY.5HL3 was produced in E. coli and plantibody was produced in Arabidopsis ................................................................ 106 Figure 4.3. The transgene in plant transformation constructs pQY4.1-1 (A) and pQY4.4-1 (B). 3582: Can 35S promoter with a duplicated enhancer; sp: plant pathogenicity—related protein PR-1b signal peptide sequence; T,m and Tm: transcription lamination sequences for tch and nos respectively; Pm: nos promoter, Kn': kanamycin resistance gene .................................................... 108 xiii I Figure 4 Figure . Figure Figure FIgUre Figure Figure Figure 4.4. The germination of Arabidopsis tansformant seeds on kanamycin selection medium plates. A. Seeds from un- transformed control plants; B. Seeds from plants transformed with pQY4.4-1, targeting scFv antibody to the apoplastic space with signal peptide sequence. C. Seeds from plants transformed with pQY4.1-1 , targeting scFv antibody to the cytoplasm ............... 109 Figure 4.5. SDS-polyacrylamide gel electrophoresis and Western blot analysis of anti-zearalenone scFv plantibody expression. Soluble proteins of Arabidopsis from un-transformed plants (lane 1), vector pKYLX71::35S2 transformed plants (lane 2), and pQY4.4-1 transformed plants (lane 3) were subjected to SDS- PAGE and stained with Coomassie brilliant blue G-250 (A). Western blot of duplicate SDS-polyacrylamide gel detected by anti-E tag antibody (B). Lane M: protein molecular size marker with 10KDa-ladder ........................................................................ 110 Figure 4.6. Effect of methanol on the binding of anti-zearalenone monoclonal antibody (mAb) and bacterial scFv antibody (EY.5HL3) and plant scFv antibody (plantibody) to immobilized zearalenone-KLH conjugate ........................................................... 112 Figure 4.7. Cytolocation of anti-zearalenone scFv plantibody by immunogold labeling and transmission electron microscopy. Leaf sections from plants expressing anti-zearalenone scFv plantibodies (A, B) and non-transformed control plants (D) were detected by mouse anti-E tag antibody followed by goat anti- mouse lgG gold conjugate. (C): Leaf sections from non- transforrned control plants were pre-incubated with anti- zearalenone scFv antibody, then with mouse anti-E tag antibody followed by goat anti-mouse IgG gold conjugate. CW, cell wall; CY, cytoplasm, ICS, intercellular space; Arrows: gold labeling; Magnification x 39,000 ............................................................... 114 Figure 5.1 . Preparation of deoxynivalenol (DON) conjugates via 3-hydroxyl group (Pestka et al., 1995) ............................................................ 127 Figure 5.2. Panning-elution selection procedure for deoxynivalenol (DON) mimotopes in anti-DON monoclonal antibody coated Immulon-4 microtiter wells ............................................................................... 130 Figure 5.3. Enrichment of mAb 6F5 specific phage. Phage were titered after elution with DON (100 pglml) from mAb 6F5 coated lmmulon-4 microtiter wells or from mouse IgG coated Immulon—4 microtiter wells in each round of panning-elution selection ............................................................ 139 xiv Figure Figure FIgUI'I Figur Flgu Flgu FIgL FIQL Figure 5.4. Competition between phage-displayed mimotope peptides and DON for binding to immobilized mAb 6F5 in CD-ELISA. Various concentrations of free DON competed with an equal volume of phage-displayed peptides (at constant concentration) for binding to immobilized mAb 6F5. Bound phage peptide was detected by horseradish peroxidase conjugated sheep anti-M13 IgG and then measured by absorbance. DON-HRP was included as a positive control ................................................................................ 141 Figure 5.5. Competition between synthetic peptide C430 and DON for binding to mAb 6F5. (A). DON and synthetic peptide C430 compete with DON-HRP for binding to immobilized mAb 6F5. (B). DON and synthetic peptide C430 compete with C430-HRP for binding to immobilized anti-DON mAb 6F5 ................................ 142 Figure 5.6. Competition between individual amino acids in synthetic peptide C430 and DON measured with CD-ELISA. Amino acids (at various concentrations) competed with DON-HRP for binding to immobilized mAb 6F5 ...................................................................... 143 Figure 5.7. CD-ELISA with DONPEP-AP fusion protein. Binding of DONPEP—AP fusion protein to immobilized mAb 6F5 was inhibited by free DON. DON-HRP was included as a positive control ............................................................................................. 144 Figure 5.8. Application of C430-HRP and DONPEP-AP in DON immunoassay (CD-ELISA) in a DON-spiked wheat extract. Immulon-4 microtiter wells were coated with mAb 6F5 antibody. DON-HRP was included as a positive control ................................ 146 Figure 5.9. The specificity of antibody from C430-BSA immunized mice. DON and C430 at various concentrations were used to inhibit the binding of antisera to immobilized DONPEP.2 phage. Bound mouse antibodies were detected by horseradish peroxidase conjugated goat anti-mouse lgG and measure by absorbance .................................................................................... 148 Figure 5.10. Cytotoxicity to bone marrow cells after 18 hr incubation with synthetic peptide C430 co-incubated with (white bar) or without (hatched bar) DON (3.4 uM). Cell viability was determined using a MTI' conversion assay .............................................................. 149 Figure 5.1 1 . Effects of DON (3.4 pM) and synthetic peptide C430 (3.4 uM) on protein synthesis in vitro with rabbit reticulocyte Iysate. The translation template was y-globulin mRNA (2 pg). “+” or “-”: with or without added, respectively .............................................. 151 XV Figure 5. Figure 5.12. Possible mechanisms for the binding of DON mimotope peptides to mAb 6F5 and for the immunogenicity of C430-BSA. In Model 1, DON and DONPEP might bind to different portions at the antigen binding pocket of mAb 6F5. After immunization with DONPEP, only anti-DONPEP antibody is produced. In Model 2, DON and DONPEP bind to mAb 6F5 in the same fashion. But after immunization with DONPEP, only the epitope which is different from DON elicits antibody response .................... 154 ABTS AP“. 3‘ BCle BSA: CD-E CDR: Cl-El CT: c DON EDTI ELEl ELIS FB1 : GC: HPLI HRP ICSQ: IPTC KLH. MBS MS: NBT OVA PAG PCR PBS PEP DNP PMS SCH LIST OF SYMBOLS AND ABBREVIATIONS ABTS: 2,2-azinobis(3—3-ethylbenzthiazoline sulfonic acid). AP: alkaline phosphatase BCIP: 5-bromo-4-chloro-3-indolyphosphate-p-toluidine. BSA: bovine serum albumin. CD-ELISA: competitive direct enzyme-linked immunosorbent assay. CDR: complemetarity-determining region. CI-ELISA: competitive indirect enzyme-linked immunosorbent assay. CT: cholera toxin. DON: deoxynivalenol. EDTA: ethylenediaminetetraacetate. ELEM: equine leukoencephalomalacia. ELISA: enzyme-linked immunosorbent assay. FB1 :fumonisin B1. FW: framework region. 60: gas chromatography. HPLC: high performance liquid chromatography. HRP : horseradish peroxidase. I050: the concentrations required for inhibition of binding by 50% , IPTG: isopropyI-[i-D-thiogalactopyranoside. KLH: keyhole limpet hemocyanin. mAb: monoclonal antibody. MBS: m-maleimidobenzoyI-N-hydroxysuccinimide ester. MS: mass spectroscopy. NBT: nitroblue tetrazolium. OVA: ovalbumin. ' PAGE: polyacrylamide gel electrophoresis. PCR: polymerase chain reaction. PBS: phosphate-buffered saline. PEP: porcine pulmonary edema. pNPP: p-nitrophenyl phosphate. PMSF: phenylmethylsulfonyl fluride. RACE: rapid amplification of cDNA ends. scFv: single-chain Fv fragment. SDS: sodium dodecyl sulfate. TBS: Tris-buffered saline. TCA: trichloroacetic acid. TLC: thin layer chromatography. TMB: 3,3',5,5'-tetramethyl-benzidine. VH: variable region of heavy chain. Vk: variable region of kappa light chain. VL: variable region of light chain. ZEN: zearalenone. xvii INTRODUCTION Mycotoxins are a group of secondary metabolites produced by toxigenic strains of fungi. Fusarium is one of the major fungal genera that contains species capable of mycotoxin production. Zearalenone, fumonisins and trichothecenes are Fusarium mycotoxins that are toxic to both human and animals and are commonly found in grain and grain products infected by Fusarium spp. The presence of these toxins in agricultural commodities is primarily governed by environmental and biological factors. A major means of eliminating mycotoxins from human and animal food is to detect and divert contaminated raw materials from feed and finished food use. Mycotoxin detection can be carried out using either conventional chemical methods such as thin layer chromatography (TLC), gas chromatography (60), high performance liquid chromatography (HPLC), and mass spectroscopy (MS) or immunoassay techniques such as enzyme-linked immunosorbent assay (ELISA). Immunoassay is commonly preferred to conventional chemical methods because of its high specificity, sensitivity, simplicity, rapidity, and applicability both in the field and the laboratory. Consequently, immunoassays for zearalenone, fumonisins, and the trichothecene deoxynivalenol (DON) have been developed in our laboratory with both polyclonal and monoclonal antibodies. However, the limits of detection with the existing antibodies to fumonisin and deoxynivalenol are not sufficiently adequate for screening these compounds at <1 ppm in conventional immunoassays, while detection at $0.1 ppm would be desirable in order for the agrifood industry to anticipate problems in a growing season. Therefore, production of higher affinity and low-cost specific antibodies against these Fusarium mycotoxins is desirable. Immunoassay for deoxynivalenol encounters further problems because chemical conjugation of DON to a carrier protein or an enzyme has low efficiency and the conjugates are weakly immunogenic. Therefore, alternative reagents are needed to replace deoxynivalenol as immunogens and immunochemical reagents. With newly developed phage display technology, a specific clone for antibody, or protein, or peptide can be selected from large repertoires because the DNA sequence is linked with its expression product in a single phage particle. This provides a powerful tool to clone mycotoxin antibody genes and to search proteinaceous mimics for mycotoxins. Once the specific antibodies to mycotoxins are cloned, the genes can also be introduced into plants. These plant-produced antibodies (plantibodies) might be very useful to neutralize mycotoxins through passive immunization of animals by feeding and to improve plant resistance against mycotoxin-associated plant diseases. The research in this dissertation was undertaken to address the above rationales and is presented in five parts. Part I (Literature Review) reviews Fusarium mycotoxins especially zearalenone, fumonisins and trichothecenes including their chemical structures, toxicity, natural occurrence, and detection. The development of recombinant antibodies, peptide mimotopes and plantibodies is also reviewed. Part II (Molecular Cloning, Expression and Characterization of A Functional Single- chain Fv Antibody to the Mycotoxin Zearalenone) describes in detail the molecular during 1 (scFv) a dmes zearaler (Attemp‘. lmmunig from m (Expresil Transge ' anlizea of a neii Mimoioi Monoclc mimotoi The am SOme C cloning (phage display and expression in bacteria) of a functional single-chain Fv (scFv) antibody to the mycotoxin zearalenone from a hybridoma cell line. The use of the scFv antibody for ELISA development and the application of ELISA for zearalenone detection in spiked corn samples are also demonstrated. Part III (Attempts to Clone Single-Chain Fv Antibodies Against Fumonisin B1 from lmmunized Mice) explains efforts to produce scFv antibodies with phage display from mice immunized with fumonisin B1-cholera toxin conjugate. Part IV (Expression of A Functional Anti-Zearalenone Single-Chain Fv Antibody in Transgenic Arabidopsis Plants) describes in detail the expression of a functional anti-zearalenone scFv antibody in transgenic Arabidopsis plants. The construction of a new phage-display cloning vector is also described. Part V (Identification of Mimotope Peptides Which Bind to the Mycotoxin DeoxynivalenoI-Specific Monoclonal Antibody) describes the identification of two deoxynivalenol mimotope peptide sequences from a phage-display random 7-mer peptide library. The application of these peptide mimotopes in deoxynivalenol immunoassay and some of their bio-activities are also described. PART I. LITERATURE REVIEW N, apart oi Casale, dunng g dunng l includin biologi at haw comm do notd In som foods (1 effects: and car I animals pathOQ body of VITUIen 1989; SChO/br MYCOTOXINS Mycotoxins are a chemically diverse group of toxic compounds produced as a part of secondary metabolism in a wide variety of filamentous fungi ( Pestka and Casale, 1990). They often contaminate agricultural commodities priorto harvest and during storage. These natural toxicants are elaborated in plant tissue infected during the growing season or later in storage environments by common fungi including species of Aspergillus, Penicilium and Fusarium. Environmental and biological factors, particularly the regional weather during the growing season and at harvest, determine the degree to which a mycotoxin will contaminate a commodity (CAST, 1989). Because mycotoxins are resistant to food processing and do not degrade at high temperatures, they enter the human and animal food supply. In some cases, mycotoxins are passed on through animal feed to meat and poultry foods (Pestka et al., 1995). Human and animal mycotoxicoses include diverse effects such as gastroenteritis, hypetestrogenism, kidney dysfunction, neurotoxicity and cancer (Pestka and Casale, 1990). Although mycotoxins are defined as fungal secondary metabolites toxic to animals, many of them are also phytotoxic and serve as virulence factors for plant pathogenic fungi (Desjardins and Hohn, 1997). For example, there is a growing body of evidence indicating that trichothecenes, including DON, can enhance the virulence of plant-pathogenic species of Fusarium on plant hosts (Adams and Hart, 1989; Desjardins et al., 1989; 1992; Bruins et al., 1993; Wang and Miller, 1988; Scholbrock et al., 1992; Atanassov et al., 1994; Proctor et al., 1995). Fumonisins have a concen Fusani zeanue Mach, uthofl mycou Zearai undec lNodur and C Woddi Warns zeara have also been shown to cause necrosis and other disease symptoms at low concentrations and are associated with high level of virulence in pathogenic Fusarium (Lamprecht et al., 1994; Desjardins et al., 1995). The mycotoxin zearalenone has also been reported to cause certain phytotoxicities (Vianello and Maori, 1981; Wakulinski, 1989; Nedelnik, 1993). Among the most important mycotoxins are zearalenone, fumonisins, trichothecenes, aflatoxins, and ochratoxins (Pohland, 1993). The first three mycotoxins are produced by Fusarium and will be reviewed in this chapter. Zearalenone Production and occurrence. Zearalenone [6-(10-hydroxy-6-oxo-trans-1- undecenyIFB-resorcylic acid-lactone] (Figure 1.1) is an estrogenic mycotoxin produced by the genus Fusarium after infection of corn and small grains (Pestka and Casale, 1990). Species of Fusarium are extremely common in nature and worldwide in distribution. They are active pathogens infecting a wide variety of host plants. Since the first finding of zearalenone in Fusarium-infected corn in 1960s, zearalenone and its analogs, along with several other mycotoxins (such as deoxynivalenol, nivalenol and fumonisins), have been detected as secondary metabolites of Fusarium species in many mold contaminated grains and fruits, including corn, wheat, barley, oats, sorghum, sesame (Mirocha et al., 1977a), soybean (Shotwell et al., 1977), cottonseed, peanut (Price et al., 1993), walnut (Jemmali and Mazerand, 1980), rice, rapeseed (Chakrabarti and Ghosal, 1987), banana (Chakrabarti and Ghosal, 1986), tomato (El-Morshedy and Aziz, 1995), OH OH 0 Me 00". H0 o H Zearalenone Estradiol OH 0 Me OH 0 Me Ho OH H' OH a—Zearalenol B-Zearalenol OH 0 Me OH 0 Me m H “OH H' OH a—Zearalanol (Zeranol) B-Zearalanol Figure1.1. Structures of zearalenone and analogues showing structural similarities with estradiol. I\l beets (Bosch and Mirocha, 1992), and amaranth grain (Bresler et al., 1991 ). Corn is the most common grain contaminated with zearalenone and its derivatives (Wood, 1992). The occurrence of zearalenone ranged from 0.1 to 8 ppm in corn samples from several surveys in the United States (Stahr et al., 1981; Cote et al., 1984; Abbas et al., 1986; Hart, 1987). Normally, zearalenone production is promoted by high humidity and low temperatures, as is common in the upper Midwest during autumn harvest (Coulombe, 1993). Zearalenone production is also correlated with plant density, soil fertility, insect damage, mechanical damage during harvest, and improper storage conditions (Mirocha et al., 1977a; Sutton et al., 1980; Montani et al., 1988). Zearalenone, in spite of its large lactone ring, is surprisingly stable to hydrolytic cleavage (Mirocha et al., 19773). It is also relatively stable during the processing of feed and foods. Zearalenone and its analogs enter the food chain directly through contaminated grain-based food and animal feed. Once zearalenone and its derivatives are produced in grains, they will remain in the grain-based feed or food. In two separate surveys of processed grain-based products (breakfast cereals, popcorn, snack foods, corn meal, crackers/cookies/muffin mixes) conducted during 1985 and 1989, the same relative concentrations of zearalenone (average 19 to 20 uglg )were detected (Warner and Pestka, 1987; Abouzied et al., 1991 ). Additionally, the use of zeranol (Ralgro)(a-zearalanol) (a synthetic derivative of zearalenone) (Figure 1.1) as a growth-promoting agent in beef cattle and sheep provides another route to enter the food chain (Sundlof and Strickland, 1986). Zeranol has been found in the edible tissues (such as muscle, liver, kidney, and fat) of the animals implanted with this growth-promoting agent (Dixon and Russell, 1986) Distribution, elimination and metabolism. When ingested orally by animals, zearalenone appears to be fairly rapidly absorbed through the gut into the circulatory system (Dailey et al., 1980; Olsen et al., 1985a). Zearalenone can then be transported to various organs and tissues. The liver appears to be the main organ of deposition upon absorption (Chichila et al., 1988). Zearalenone is also distributed into adipose tissue, ovary, utenls, testicle, and other sites (Ueno et al., 1977; Appelgren et al., 1982). Zearalenone can be rapidly eliminated from an animal. Following a single oral dose of zearalenone in rats, about 90% of the dose was excreted in feces after 48 hr, while the remaining 10% was excreted in urine (Smith, 1982). Zearalenone and metabolites were excreted mainly in the free form with conjugates being found only in urine. Using different oral doses of zearalenone in rats, Fitzpatrick et al. (1988) found that dose had little effect on metabolites formed, or excretion route. In one study, Pandey et al. (1990) showed that UDP-glucuronyltransferase induction by phenol barbital increased urinary excretion of conjugated a—zearalenol. A more recent study with administration of [3H] zearalenone showed that the biological half-life of zearalenone in plasma was 86 hr for intravenously or orally dosed immature pigs, while only 3.3 hr for intravenously dosed animals after bile removal (Biehl et al., 1993). It provided evidence for extensive biliary secretion and enterohepatic cycling of zearalenone and metabolites in pigs. Besides the route of feces and urine, a small portion of zearalenone can be excreted into milk as al. an SBJ vul Ch mo Oil Ani re pl (Mil conjugates (Prelusky et al., 1990). Zearalenone is metabolized mainly in the liver through dehydrogenation and glucuronide conjugation (Kiessling and Pettersen, 1978; Mirocha et al., 1981 ; Olsen et al., 1981; Bock et al., 1987). Considerable differences between species exist in the ability to reduce zearalenone (Olsen and Kiessling, 1983; Pompa et al., 1986). Among five species examined, swine had one of the greatest abilities to form the more highly estrogenic a-zearalenol (Olsen et al., 1985b). In some cases, sulfo- conjugates of zearalenone and its analogs were also found at low levels (Olsen et al., 1986; Bories et al., 1991). Toxicity. With their chemical similarity to estradiol (Figure 1 .1 ), zearalenone and its derivatives bind to mammalian estrogen receptors in reproductive and other sensitive tissues (Hurd, 1977). When fed to animals, zearalenone causes hyperestrogenism with symptoms such as enlargement of the uterus and nipples, vulvar swelling, vaginal prolapse, and infertility (Hidy et al., 1977; Mirocha and Christensen, 1974). Swine are the most sensitive of the large domestic animals and most commonly affected with hyperestrogenism on the farm although bovine hyperestrogenism due to zearalenone is also suspected (Mirocha et al., 1977b). Other farm animals affected by zearalenone are dairy cattle, chickens and turkeys. Animals experimentally affected are rats, mice, guinea pigs, monkeys and lambs (Mirocha et al., 1977). Embryotoxicity and teratogenic effects have also been reported in some animals after consumption of zearalenone-contaminated feeds (Miller et al., 1973; Sharma et al., 1974; Shreeve et al., 1978; Chang et al., 1979; Becci et al., 1982). Zearalenone and its derivatives also affect the immune system 10 anabo probe, zearal Sheep of animals. They are capable of inhibiting mitogen-stimulated lymphocyte proliferation (Forsell and Pestka, 1985). Dietary exposure to 10 ppm of zearalenone for two weeks decreases the resistance of mice to Listeria monocytogenes (Pestka et al., 1987). Zeranol could induce acute hepatotoxicity and, subsequently, hepatic carcinogenesis in Armenian hamster (Coe et al., 1992). More recently, DNA adducts were also found in the kidney, liver and ovary of female mice treated with zearalenone (Pfohl et al., 1995; Grosse et al., 1997). Using the closely related a-zearal'enol, the no-honnonal effect level for zearalenone was estimated to be 50-60 ug/kg body weight per day, based on a 90- day oral study in ovariectomized monkeys with vaginal comification as the endpoint (Griffin et al., 1984; Lindsay, 1985; Kuiper-Goodman et al., 1987). Dividing the non- honnonal effect level by a safety factor of 500, which takes into account inter— and intraspecies differences and uncertainties in the experiment model, Kuiper- Goodman et al. (1987) gave an estimated Tolerable Daily Intake (T DI) of 100 ng/kg body weight per day for humans. Although zearalenone and its derivatives are toxic at high doses, they act as anabolic hormones enhancing growth when used at the proper time and in the proper amounts. Notably, zeranol (Ralgro) (a-zearalanol), a synthetic derivative of zearalenone, has been approved as a growth-promoting agent in beef cattle and sheep (Umberger, 1975). 11 Fumon produce F. proif sorghui F. moni causati 1904). ' leukoer charact hemiso obsew Cher 199 DTOC an 6 al., 1 Cam; Fumonlsins Historical background. Fumonisins are a group of secondary metabolites produced by Fusarium moniliforme (Sheeldon) (Benzuidenhout et al., 1988) and F. proliferatum (Ross et al.,1990). These fungi are commonly found on corn, sorghum, and other grain commodities throughout the world (Marasas et al., 1984). F. moniliforme, which was firstly described by Sheldon (1904), was implicated as a causative agent of “moldy corn poisoning” in animals in the United States (Peter, 1904). This mold has been associated with an equine poisoning known as equine leukoencephalomalacia (ELEM) (Ross, 1994). This is a neurotoxicosis characterized by multifocal liquefactive necrosis in the white matter of cerebral hemispheres (Marasas et al., 1988b). Although the disease symptoms have been observed since the previous century (Marasas, 1986), its causative agent [fumonisin B1 (FB,)] was not isolated and identified until 1988 (Benzuidenhout et al., 1988; Marasas et al., 1988b). As a consequence, interest in fumonisins has increased leading to extensive studies on their occurrence, distribution, production, toxicity, detection, and chemistry (Ross, 1994). Six different fumonisins, fumonisin A,, A2, B1, B2, Ba, and B,, have been chemically characterized (Figure 1.2) (Bezuidenhout et al., 1988; Cawood et al., 1991; Plattner et al., 1992). FB1 is the most toxic and predominant fumonisin produced by F. moniliforme (Gelderblom et al., 1988a), and has been confirmed as an etiologic agent of fatal animal diseases such as ELEM in horses (Kellerrnan et al., 1990), porcine pulmonary edema (PEP) in pigs (Harrison et al., 1990), and liver cancer in rats ( Gelderbolm et al., 1991). In addition, epidemiological studies 12 inc 3C pe be de of Bi all WI' be indicate that fumonisins are associated with human esophageal cancer in South Africa (Marasas et al., 1988a). Chemistry. Fumonisins are diesters of 14, 15-propane-1,2,3-tricarboxylic acid and either 2-acetylamino- or 2-amino-12,16-dimethyl-3,5,10,14,15- pentahydroxy-icosane or its C-10 deoxy analogue (Figure 1 .2) (Bezuidenhout et al., 1988). Chemical structures of these toxins are similar to that of Altemaria altemata f. sp. lycopersici (AAL) toxin and sphingosine (Figure 1.2) (Norred, 1993). The biosynthetic pathways through which fungi produce fumonisins have only been partially elucidated (Norred, 1993). When Plattner and Shackelford (1992) fed deuterium-labeled methionine to F. moniliforme cultures, they observed high levels of deuterium incorporation in FB1 at the methyl groups on the C-12 and C-16 position of fumonisin backbone. Based on 13C-labeled acetate feeding experiment, Blackwell et al. (1994) concluded that the backbone of fumonisins was synthesized by the fungus through condensation of acetyl coenzyme A and serine. However, more work is still required to fully elaborate the biosynthetic pathway of the fumonisins. Natural occurrence. F. moniliforme, the fumonisin-producing fungus, commonly occurs in the world and it has been isolated in many countries (Bacon and Nelson, 1994; Doko et al., 1995). This fungus is primarily found as a corn contaminant, but can also be found on several grain commodities such as sorghum, wheat, rice, and cat and other agricultural products such as beans, peanuts, sugar beats, and bananas (Bacon and Nelson, 1994). 13 FUMONISINS R1 R2 R3 COOH FAl OH OH CH2CO cocnzICchzcoon FA2 H OH CH2CO I R, 0,, F81 OH OH H 20 1CH3 F82 H OH H F83 OH H H 21 CH3 0 22 H3 1 NHR3 FB4 H H H COCHzCHCHzCOOH OOH COOH cocr—rzcncnzcoon OH on AAL TOXIN W CH3 on CH3 OH NHz OH SPHINOSINE chzm NH2 Figure 1.2. Structures of known fumonisins and Altemaria altemata f. sp. chopersici (AAL) toxin, showing structural similarities with sphingosine (Norred, 1993). 14 C storage reporter aerated product FB, at It conditi 1990). caused Maras the hor oi the I ELEM body \ Sufien ELEM Edem, alooj et at, bIOSYn Conditions required for optimal production of fumonisins in the field or storage are only partially known (Bacon and Nelson, 1994). Bars et al. (1994) reported that a temperature of 20°C, 32% moisture content of corn media, and aerated atmosphere (cotton-stoppered flasks) were the optimum condition of FB1 production by F. moniliforme in laboratory experiments. F. moniliforme can produce F B1 at level of 300 to 17,000 ppm when cultured on solid corn media at the optimum conditions for 2 to 12 weeks (Bars et al., 1994, Nelson et al., 1991, Alberts et al., 1990) Toxicity. Equine leukoencephalomalacia (ELEM) is the most studied disease caused by consumption of F. moniliforme contaminated corn (Norred, 1993; Marasas et al., 1988b). ELEM develops over a several-week period, during which the horse may refuse feed, becomes lethargic and is disoriented. The progression of the condition includes ataxia, profuse sweating, convulsions, and finally death. ELEM has been reproduced by intravenous administration of 7 doses (0.125mg/kg body weight) of purified FB1 over 10 days (Marasas et al., 1988b). The horses suffered convulsions and were found to have brain lesions similar to those seen in ELEM cases (Merrill et al., 1995). Porcine pulmonary edema (PPE) which develops hydrothorax and lung edema usually resulting in death was particulariy prevalent in the United States when pigs were fed contaminated corn (Norred, 1993). Pigs that received pure FB1 at 0.4 mg/kg body weight were dead on day 5, with typical PPE lesions (Harrison et al., 1990). Haschek et al. (1992) suggested that the disruption of sphingolipid biosynthesis by FB1 causes membranes damage in hepatocytes and release into 15 the blood stream. Pumonary intravascular macrophages (PlMs) which phagocytize the membrane fragments are especially rich in the lungs of pigs. This initiates the PlMs to release enzymes and other mediators, which increase capillary permeability in the lung tissue and lead to edema (Haschek et al., 1992; Merril et al., 1995). In addition, fumonisins can cause nephrotoxicity, hepatotoxicity and hepatocarcinogenicity in some experimental animals (Jaskiewicz et al., 1987; Gelderblom et al., 1991; 1992; Riley et al., 1994; Voss et al., 1995). More importantly, human esophageal cancer in the areas of Southern Africa and China has been correlated with the consumption of corn contaminated by F. moniliforme (Marasas et al., 1988a; Marasas, 1995; Cheng et al., 1985; Chu and Li, 1994). Trichothecenes Occurrence. Trichothecenes are a group of structurally similar sesquiterpenoid metabolites (Figure 1.3) produced mainly by Fusarium spp. (Bamburg and Strong, 1971 ; Bamburg, 1983; Ueno, 1983; Betina, 1989a) . The first known member of this group, trichothecin, was originally discovered as an antifungal antibiotic in 1948 (Freeman and Morrison, 1948; 1949). Since that time over 80 trichothecenes have been isolated and characterized (Betina, 1989b). But only a few members of the trichothecenes, such as deoxynivalenol (DON, vomitoxin), nivalenol (NIV), T-2 toxin, HT-2 toxin, and diacetoxyscirpenol (DAS) are detected as major natural contaminants in cereal grains (Betina, 1989b). 16 ----R 3 1 4t”-H R2 Trichothecene R1 R2 R3 R4 R5 T—2 toxin OH OAC OAC H OOCCH2CH(CH3)2 HT-2 toxin OH OH OAC H OOCCH2CH(CH3)2 T-2 tetraol OH OH OH H OH Diacetoxscirenol OH OAC OAC H H Scirpenetriol OH OH OH H H Deoxynivalenol OH H OH OH O Nivalenol OH OH OH OH O Fusarenon-X OH OAC OH OH O Figure1.3. Structure and numbering system of some naturally identified trichothecene mycotoxins (modified from Ueno, 1983). 17 enviro Wet a trichc Uenc of de Sch red SUI Production of trichothecenes in cereal grains is primarily dictated by environmental conditions (Vesonder et al., 1978; Ueno, 1983; Cote et al., 1984). Wet and cool seasons in the spring and early summer favor Fusarium infection and trichothecene production in the field (T uite et al., 1974; Shotwell et al., 1977; 1983; Ueno, 1983; CAST, 1989). In a year when wheat scab is epidemic, the occurrence of deoxynivalenol could be detected up to12 ppm in bulk grain samples (Hart and Schabenberger, 1998). Toxicity. Symptoms in acute experimental trichothecene poisoning are reduced weight gain, anorexia, dermal irritation, abortion and hematological lesions such as leukopenia, anemia and lack of bone marrow development (Ueno, 1987; Rotter et al., 1996). Major human outbreaks of trichothecene toxicoses have been reported in Russia, Japan, China and India (Beardall and Miller, 1994). Consumption of trichothecene—contaminated grains causes headaches, chills, severe nausea and vomiting, diarrhea, visual disturbances and fatal hemorrhage in humans (Watson et al., 1984). Trichothecenes also modulate immune function and their ingestion over prolonged periods may reduce the immune resistance to infectious diseases, facilitate tumor growth by impairing immune surveillance (Pestka and Bondy, 1994), and cause autoimmune disease, as demonstrated in experimental deoxynivalenol-induced IgA glomerulonephritis (Dong et al., 1991; Dong and Pestka, 1993; Rasooly et al., 1994; Rasooly and Pestka, 1994). 18 Analytical methods for mycotoxin detection The presence of mycotoxins in foods and feeds is potentially hazardous to the health of humans and animals. Although it is difficult to remove mycotoxins from human and animal diets, it is possible to decrease the risk of exposure through a rigorous program for monitoring these toxins in foods and feeds and diversion of the contaminated materials (Chu, 1992; Pestka et al., 1995). In general, analysis of mycotoxins in food and feed is a difficult task because only trace amounts of the toxins are present in the sample. The analysis is further complicated by the uneven distribution of mycotoxins in the sample and by sample matrix interference. Normally, the suspected contaminated commodity is first subjected to a rigorous sampling program (Campbell et al., 1986). The toxin is then extracted from the sample and subjected to a very extensive cleanup treatment to remove interfering substances before analysis is conducted. Current analytical methods for mycotoxins include thin layer chromatography (TLC), high performance liquid chromatography (HPLC), gas chromatography (GC), mass spectroscopy (MS), immunoassay and bioassay. For mycotoxins that contain a chromophore group with fluorescent properties, such as zearalenone, sample extracts after cleanup are generally subjected either to TLC or to HPLC to separate different structurally related compounds and contaminants (Chu, 1992; Richard et al., 1993). Identification of the toxin is made by comparing the retention factor (Rf) values or retention time in the chromatogram with the standard, whereas quantitative information is obtained from fluorescence intensity or absorbance of the elution peak (or spot). Some 19 mycotoxins, such as trichothecenes and fumonisins, do not have well-defined absorption maxima or potential for fluorescence under ultraviolet or visible light. Therefore, derivatization reagents are required to provide a fluorescent characteristic for their detection (Chu, 1992; Richard et al., 1993). An effective TLC or HPLC method can detect zearalenone, fumonisins and deoxynivalenol at 50 to 300 ppb (Richard et al., 1993). Higher sensitivity and selectivity of mycotoxin analysis can be achieved by using GC and GC/MS techniques although they require laborious sample cleanups, and expensive instruments (Pestka, 1988, 1994; Chu, 1991). Immunoassays have been widely used for mycotoxin analysis at the pg and ng levels (Pestka et al., 1995). The most common immunoassay, enzyme-linked immunosorbent assay (ELISA), involves competition between a free mycotoxin in a sample extract with an enzyme-labeled mycotoxin for an antibody binding site. Mycotoxins can be analyzed directly in a liquid system such as milk and solid substrates can be extracted with methanol-water and analyzed directly or after dilution. Because mycotoxins alone are not antigenic, early studies focused on the development of methods for conjugation of mycotoxins to a protein or polypeptide carrier and optimization of conditions for antibody production in rabbits and in other animals (polyclonal antibody). With the advances in hybridoma technology, monoclonal antibodies against many important mycotoxins, including zearalenone, fumonisin B,, and deoxynivalenol, were also generated (Pestka et al., 1995). Compared with other methods, mycotoxin immunoassays have several advantages 20 for rapid field tests, including high specificity, sensitivity, facile sample preparation, and ease of use (Pestka et al., 1995). 21 RECOMBINANT ANTIBODIES AND PEPTIDE MIMOTOPES The wide application of mycotoxin immunoassay has led to a great demand for immunochemical reagents including low-cost antibodies with high affinity and defined specificity. With the advances in the field of recombinant antibody technology, it is now theoretically feasible to engineer low cost antibodies for mycotoxins with desirable affinity and specificity characteristics by using recombinant DNA technology to manipulate the basic domain structure of the immunoglobulin molecule and to express it in bacteria (Hoogenboom et al., 1992; Lerner et al., 1992). Peptide mimotopes to mycotoxins may also be selected from a random peptide library and used as alternative immunochemical reagents. Native antibody An immunoglobulin (lg) molecule is composed of two heavy (H) and two light (L) protein chains (Figure 1.4). These protein chains contain defined Variable (V), Diversity (D, heavy chain only), Joining (J) and Constant (C) regions. The variable region is positioned at the amino-terminus and is composed of alternating framework (Fw) and hypervariable or complemetarity—determining region (CDRs). The VL and VH regions constitute the basic antibody binding site (paratope) which is dictated by the CDRs and, to a lesser extent, the Fw region. This domain structure encoded by variability (V) genes facilitates the extensive gene rearrangement that occur in B cells during in vivo immune selection. When newly generated B cells bind antigens via the lg receptor, they differentiate (with T cell 22 help) to either short-lived lg secreting plasma cells or long-lived memory cells that are involved in secondary/tertiary responses. During the selection process, highly frequent somatic mutations in the rearranged lg genes of the memory B cells greatly improve antibody affinity. Recombinant antibody and phage display Recombinant antibody technology involves rearrangement or assembly of antibody V-genes, surface display of antibody, affinity selection, affinity maturation and soluble antibody production (Winter at al., 1994). Initially, antibody genes were taken from hybridomas, cloned into plasmid vectors and expressed as fragments in bacteria (Better et al., 1988) or as complete antibodies in mammalian cells (Oi et al., 1983). But screening specific antibody from large combinatorial repertoires was a difficult task. In 1985, a report was published describing the in-frame insertion of a DNA fragment encoding a section of the restriction endonuclease (5le into the gene of a minor capsid protein (93p) of the filamentous bacteriophage fd (Smith, 1985). The filamentous fd bacteriophage is male specific, containing circular single-strand DNA (Model and Russel, 1988) . The phage genome (approximately 6500 nucleotides in length) encodes 10 proteins (Table 1.1). An important feature of filamentous fd phage is their ability to package longer genomes than the wild-type DNA, simply by further addition of 98p subunits. Fusion phages have been generated with insertions into either the minor capsid protein g3p for large insertions, or the major capsid 23 Figure 1.4. Antibody model structures showing subunit composition and domain distribution along the polypeptide chains. Fragments generated by proteolytic cleavage and /or recombinant technology appear in the shaded area (Phannacia Biotech). 24 Table 1.1. The genes and proteins of fd bacteriophage Gene Protein size Copy number Protein function 1 35-40 KDa few/cell NS membrane protein; interacts with host thioredoxin; required for assembly. 2 46KDa ~103/cell Site- and (+) strand-specific endonuclease! topoisomerase; required for replication of RF. X 12 KDa ~500/cell N-terminal fragment of gene 2; required for replication of RF. 3 42 KDa 5/particle At one end of particle; required for correct morphogenesis of unit-length particles; N-terminal part binds to F pilus of host cell. 4 49KDa few/cell NS membrane protein; required for assembly. 5 10KDa ~105lcell Major structural protein during replication; controls expression of 92p; binds to DNA; replaced by g8p during assembly; controls switch from RF replication to progeny (+) strand synthesis. 6 12KDa ~5/particle At the same end of particle as g3p; involved in attachment and morphogenesis. 7/9 3.5KDa ~5 of each At opposite end of particle to gSp/g6p; involved in [particle assembly; 8 5KDa 2,700-3,000/ Major coat protein. particle 25 protein g8p for short peptide insertions (Smith, 1993). In these “fusion phages’, the DNA encoding the insert protein is packaged as an integral part of the phage genome, while the chimeric protein product is expressed on the surface of the bacteriophage particle, allowing the simultaneous selection of a gene and its protein. Since 1985, this ingenious system linking both the DNA Sequence and its expression product in a single phage particle has resulted in a rapidly expanding technology with diverse applications (O’Neil and Hoess, 1995; Hill and Stockley, 1996) A rapidly expanding application of phage display technology has stemmed from the discovery that antibody fragments can be presented on the surface of ' bacteriophage in a biologically active form (McCafferty et al., 1990) and the repertoires of heavy- and light-chain variable region genes can be built by using polymerase chain reaction (PCR) (Orlandi et al., 1989). In the last decade, many antibody or antibody fragment genes have been cloned with phage display. Antibody phage display is accomplished by fusing the coding sequence of the antibody variable (V) regions to the amino terminus of the phage minor coat protein 93p (McCafferty et al., 1990). Expression of the fusion product and its subsequent incorporation into the mature phage coat results in the antibody being presented on the phage surface, while its genetic material resides within the phage particle. This linkage between antibody genotype and phenotype allows the enrichment of antigen-specific phage antibodies, using immobilized or labeled antigen. In a screening procedure, called “biopanning” (Marks et al., 1991), phage that display a relevant antibody will be retained on the surface coated with antigen, while non- 26 adherent phage will be washed away. Bound phages can be recovered from the surface, re-infected into bacteria and re-grown for further enrichment and, eventually, for binding analysis (Marks et al., 1991; Hawkins et al., 1992; Yuan et aL,1997) Affinity and specificity in phage-displayed antibodies can be further enhanced by mimicking B cell somatic mutation via in vitro mutagenesis or by shuffling a repertoire of in vivo somatically mutated V-genes (Hoogenboom et al., 1992). The CDR regions are often the targets for antibody in vitro mutation and maturation. Using “CDR-walking”, Yang et al. (1995) created independent libraries with mutations in one of four CDR regions, combined the selected variants in one clone, and obtained a 420-fold increase in affinity. The CDR3 of the heavy chain was demonstrated to be most important for in vitro affinity maturation (Thompson et al., 1996; Schier et al., 1996; Yang et al., 1995). Chain shuffling is the sequential replacement of VH and VL region genes with repertoires of V-genes (Marks et al., 1992a). Chain shuffling of V-genes has been used successfully to obtain a 300-fold increase in the affinity of an anti-hapten antibody, originally isolated from a naive human scFv library (Marks et al., 1992b). The source of the V-genes for chain shuffling can be a library of light or heavy chain V-genes from non-immunized animals (naive library) (Marks et al., 19923), from animals immunized with an antigen of interest (Kang et al., 1991) , or from other positive clones in the initial library (Clackson et al., 1991). Several types of antibody constmct have been displayed including the variable region of single heavy chains, and single-chain Fv fragments (scFv) (Hill 27 and Stockley, 1996). Fab fragments have also been displayed in a form in which either the light or heavy chain is expressed as a fusion on the bacteriophage surface with the complementary heavy or light chain associated via its secretion into the periplasm of phage-infected cells (Barbas et al., 1991; Hoogenboom et al., 1991) Compared to Fab and Fv fragments, single-chain Fv fragments fold readily in the correct conformation and have improved stability because the variable heavy (VH) and variable light chain (VL) domains are covalently joined by a polypeptide linker (Raag and Whitlow, 1995). Generally, peptide sequences from known protein structures are used as linkers, whose lengths and conformations are compatible with bridging the variable domains of an scFv without serious steric interference (Bird et al., 1988; Takkinen et al., 1991; Mallender and Voss, 1994). The most widely used linker designs have sequences consisting primarily of stretches of Gly and Ser residues for flexibility, sometimes with charged residues such as Glu and Lys interspersed for solubility (Bird et al., 1988; Huston et al., 1988; Whitlow et al., 1993). The stability of a particular scFv form depends on a number of factors, in addition to the nature of the V,,,/VL interface. These include the presence of antigen, the length of the linker, the solvent system, and temperature. Longer linkers were found to stabilize scFv antibodies (Whitlow et al., 1993; Holliger et al., 1993), while some organic solvents, such as ethylene glycol (Kortt et al., 1994), ethanol (Essig et al., 1993), and methanol (Yuan et al., 1997) were showed to destabilize scFvs. Some strategies generally applicable to protein stabilization may be used to 28 stabilize the scFv (Raag and Whitlow, 1995). These include the engineering of stronger hydrogen bonding interaction (Pantoliano et al., 1989), disulfide bonds (Glockshuber et al., 1990; Btinkmann et al., 1993; Luo et al., 1995; McCartney et al., 1995), metal binding sites (Wade et al., 1993), increasing buried hydrophobic areas, and adding or improving electrostatic interactions (Jones et al., 1992). The majority of scFv applications involve a bifunctional task in which scFv binding to a target antigen allows high specific delivery of a second effector function (Raag and Whitlow, 1995). They might also be used as diagnostic reagents. Antibody fragments, specific for an important pollutant or a toxicant, could have considerable potential for the detection and removal of trace levels of contaminants. For example, a cloned antibody fragment to the herbicide paraquat has been shown to be capable of removing paraquat from aqueous environments (Graham et al., 1995). Antibody fragments with catalytic activities have also been developed using phage display (McCafferty et al., 1994; Janda et al., 1994; Baca et al., 1997). Peptide mimotopes The physical linkage between a vast library of variants with various binding activities to genes encoding each variant from phage display also permits the identification of peptides with certain binding functions. In 1990, three groups simultaneously reported the construction and screening of highly diverse libraries of unstructured random peptides (6-12 residues) displayed on fd phage or the related phage M13 as g3p fusions (Scott and Smith, 1990; Cwirla et al., 1990; Devlin et al., 1990). In a complementary strategy, peptide libraries fused to the 29 major coat protein, g8p, have also been used for epitope mapping (Felici et al., 1991). The increased valency of 98p fusions permits selection of lower-affinity ligands, while g3p library generally produce higher affinity ligands. Peptide libraries can be structurally constrained by flanking the randomized segment with a pair of cysteines which form a disulfide bond (McLafferty et al., 1993; McConnell et al., 1994; Giebel et al., 1995). Cycllzation can mimic the native structural context of epitope sequences by ‘freezing out” unproductive conformations. Structurally-constrained peptide libraries are valuable in identifying structural epitopes (Hoess et al., 1994). In the last 8 years, random peptide libraries displayed on phage have been extensively used to select for peptide ligands specific for a variety of targets (antibodies, receptors, enzymes, etc.) by using multiple cycles of in vitro selection (biopanning) and in vivo amplification (Parrnley and Smith, 1988; Smith and Scott, 1993). Phage-display libraries have also been used to identify peptide mimics of non-peptide ligands (Devlin et al., 1990; Scott et al., 1992; Oldenburg et al., 1992; Hoess et al., 1993; Adey et al., 1995). 30 PLANTIBODIES Antibodies historically have been considered as part of mammalian defense systems. With the advances in antibody gene cloning and plant genetic transformation, however, the ability to produce antibodies and antibody fragments in plants presents some very interesting possibilities. These so-called “plantibodies” can be produced on an agricultural scale for therapeutic or diagnostic use (Smith 1996). They also provide the opportunity to manipulate plant traits or reduce pathogenic infections in plants through antibody-mediated modifications of antigen activity in planta (Hiatt, 1990; Schots et al., 1992; Tavladoraki et al., 1993; Voss et al., 1995; Whitelam, 1995). The binding and retention capacity of antibodies could also be used for the isolation and processing of environmental contaminants, a process known as biofiltration (Hiatt, 1990). For expression of complete antibodies or Fab fragments, in one approach, light- and heavy-chain genes were separately introduced into plants. Plants expressing light and heavy chains were then crossed and assembled antibodies were detected in the F1 generation (Hiatt et al., 1989; Ma et al., 1994; Ma et al., 1995). Alternatively, double transformations were performed or the genes for expressing light and heavy chains were cloned into one expression cassette (De Neve et al., 1993; Dilring et al., 1990; Voss et al., 1995). The expression of a functional scFv antibody in plants is relatively simple because it requires only correct folding and it does not have to be assembled like complete antibodies. 31 Antibodies have been successfully expressed in plants both intracellulariy and intercellulariy. Full length antibodies in algal cytoplasm can form correctly folded antigenic binding sites even though the relatively high reducing conditions in the cytoplasm do not allow formation of disulfide bonds (Stieger et al., 1991). However, functional full length antibodies in the cytoplasm have not yet been reported in higher plants. It is possible that the cytoplasm of algae is less reducing and contains the chaperonins necessary for folding of full-size immunoglobulins (Smith, 1996). In contrast, the expression of scFv antibodies in the plant cytoplasm has been more successful. Some groups have reported successful expression of scFv antibodies in the cytoplasm (Owen et al., 1992; Tavladoraki et al., 1993), although others failed to get any antibody accumulation unless an endoplasmic reticulum (ER) retention signal (a tetrapeptide KDEL) was included (Schouten et al., 1996). The production of secreted antibodies in plants normally requires a signal sequence, although the origin of the signal sequence does not appear to be critical, since plant, mouse, and yeast sequences can be used (DlJring et al., 1990; Hein et al., 1991). The signal sequences not only direct the antibody fragments to plant secretory pathways but also stabilize the antibody and increase the yield (Hiatt et al., 1989; Firek et al., 1993). Although plant cell walls are believed to have exclusion limits of between 17 and 60 KDa (Carpita et al., 1979; Tepfer and Taylor, 1981; De Wilde et al., 1996), the results of De Wilde et al. (1996) suggest that the mesophyll cell walls of Arabidopsis are permeable to intact IgG molecules, which have a molecular weight of 146 KDa. These findings are of particular interest to any application where extracellular antigen binding is desired. When a suitable signal 32 sequence was used, antibody was also targeted into plant nuclei (De Neve et al., 1993). Antibody can also be targeted to specific plant organs. Fiedler and Conrad (1995) directed active scFv molecules into tobacco seeds and stored them stably for one year at room temperature. Such a system would be valuable for long-term storage and easy delivery of large quantities of antibodies for passive immunization. As in mammals, y—heavy chain expressed in plant has N-linked glycosylation, in which a high-mannose core oligosaccharide is attached to the asparagines contained within the canonical Asn-X-Ser/Thr sequence (Hein et al., 1991). However, the composition of terminal residues on the plant glycan appears to be distinct from that in mammals. The differences in glycosylation patterns of plant antibodies had no effect on antigen binding or specificity (Hein et al., 1991). However, for human therapy, the presence of plant-specific glycan might increase the immunogenicity of the recombinant antibody. Since the first plantibody was expressed in tobacco (Hiatt et al., 1989), a number of antibodies or antibody fragments have been successfully produced in plants for various purposes (Table 1 .2). Full length antibodies have been expressed in plants with the aim of producing large quantities for use in immunotherapy. Indeed, the production of multimeric secretory antibodies (Ma et al., 1995) and the ability to store antibodies long-term in seeds or other organs (Fiedler and Conrad, 1995) showed great promise in immunotherapy and passive immunization by using plantibody. In an effort to confer viral resistance to plants, Tavladoraki et al. (1993) expressed an scFv antibody against the artichoke mottled crinkle vims in the 33 h\ In A. cytoplasm of tobacco. These transgenic plants were specifically protected from viral attack, in that they had a reduced incidence of infection and showed delayed symptom development ( Tavladoraki et al., 1993). The possibility of using full length antibodies to interfere with fungal pathogenesis in the apoplast has also been examined (van Engelen et al., 1994). With the intracellular expression of specific antibody fragments in plants, plant physiological traits such as phytochrome- mediated seed germination (Owen et al., 1992) and abscisic acid-controlled physiology (Artsaenko et al., 1995; Phillips et al., 1997), have successfully been altered. The feasibility of producing specific antibody or antibody fragments in plants allows us to speculate that the expression of mycotoxin specific antibody in plants may be useful to reduce plant and animal diseases associated with the mycotoxin by antibody-mediated neutralization and degradation. Table 1.2. Antibody—derived molecules produced in transgenic plants Antibody form Antigen Plant species Reference Single domain Substance P Nicotiana Benvenuto et al., 1991 (sAb) (neuropeptide) Single chain Fv Phytochrome Nicotiana Firek et al., 1993; (scFv) Owen et al., 1992 Single chain Fv Artichoke mottled crinkle Nicotiana Tavladoraki et al., 1993 (scFv) virus coat protein Single chain Fv Beet necrotic yellow vein Nicotiana Fecker et al., 1996 (scFv) virus coat protein Single chain Fv Abscisic acid Nicotiana Artsaenko et al., 1995 (scFv) Phillips et al., 1997 Fab; IgG (1:) Human creatine kinase Nicotiana De Neve et al., 1993 Arabidopsis IgG (1:) Transition-state analogue Nicotiana Hiatt et al., 1989; Hein et al., 1991 lgG (x) Fungal cutinase Nicotiana van Engelen et al., 1994 lgG (x/k) TMV Nicotiana Voss et al., 1995 lgG (x) and lgG/A Streptococcus mutants Nicotiana Ma et al., 1994 hybrids adhesin Secretory lgA/G S. mutants adhesin Nicotiana Ma et al., 1995 IgM (k) NP (4-hydroxy-3- Nicotiana Dtlring et al., 1990 nitrophenyl) acetyl hapten lgG-like lgM Nematode stylet Nicotiana Baum et al., 1996 secretions Rosso et al., 1996 35 PART II MOLECULAR CLONING, EXPRESSION AND CHARACTERIZATION OF A FUNCTIONAL SINGLE-CHAIN Fv (scFv) ANTIBODY TO THE MYCOTOXIN ZEARALENONE 36 ABSTRACT MOLECULAR CLONING, EXPRESSION AND CHARACTERIZATION OF A FUNCTIONAL SINGLE-CHAIN Fv (scFv) ANTIBODY TO THE MYCOTOXIN ZEARALENONE The heavy and kappa light-chain variable region genes of an anti- zearalenone hybridoma cell line (2GB—6E3-2E2) were isolated by polymerase chain reaction (PCR) and joined by a DNA fragment encoding the linker peptide (Gly.,Ser)3 to generate a single-chain Fv (scFv) DNA fragment. The scFv DNA fragment was cloned into a phagemid (pCANTAB5E) and expressed as a fusion protein with an antibody recognition sequence (E-tag) and phage M13 coat protein 93p in Escherichia coli TG1. In the presence of helper phage M13K07, the scFv fusion protein was displayed on the surface of recombinant phage. High affinity scFv phage antibodies were enriched through panning-elution selection in microtiter wells coated with zearalenone-ovalbumin conjugate. The selected recombinant phages were used to infect E. coli HB2151 for the production of soluble scFv antibodies. One selected clone (pQY1.5) in HB2151 secreted a soluble scFv antibody (QY1.5) with high zearalenone binding affinity (IC50=14 ng/ml), similar to that of the parent monoclonal antibody in a competitive indirect ELISA. However, scFv QY1.5 exhibited higher cross-reactivity with zearalenone analogues and had increased sensitivity to methanol destabilization when compared with the parent monoclonal antibody. Nucleotide sequence analyses revealed that the light chain portion of scFv QY1.5 had a nucleotide sequence identity of 97% to a mouse gerrnline gene 37 VK23.32 in mouse kappa variable light chain subgroup V, whereas the heavy chain nucleotide sequence was classified as mouse heavy chain subgroup Ill (D) but without any closely related members having highly homologous complementary- deterrnining regions (CDRs) sequences. The potential of soluble scFv QY1.5 for routine screening of zearalenone and its analogues was demonstrated with zearalenone spiked corn extracts. A portion of the research presented here has been published (Yuan, 0., Clarke, J. R., Zhou, H. R., Linz, J. E., Pestka, J. J., and Hart, L. P. 1997. Molecular cloning, expression and characterization of a functional single-chain Fv antibody to the mycotoxin zearalenone. Appl. Environ. Microbiol. 63:263-269). 38 INTRODUCTION Zearalenone [6-(10-hydroxy-6-oxo-trans—1-undecenyl)-|3-resorcylic acid lactone] (Figure 1.1) is a mycotoxin produced by members of the genus Fusarium after infection of corn and small grains (Mirocha et al., 1971; 1977; Hart et al., 1982). When fed to animals, the compound causes hyperestrogenism with symptoms such as enlargement of the uterus and nipples, vulvar swelling, vaginal prolapse, and infertility (Mirocha and Christensen, 1974; Hidy et al., 1977). As a consequence of these estrogenic effects, there is a need for routine screening of agricultural commodities used for human and animal consumption. Methods for the analysis of zearalenone in food and feeds include thin-layer chromatography (TLC) (Gimeno, 1983; Scott et al., 1978; Swanson et al., 1984), gas-liquid chromatography (Scott et al., 1978; Thouvenot and Morfin, 1979), high- pressure liquid chromatography ( Scott et al., 1978; James et al., 1982; Turner et al., 1983) and more recently immunoassays (T houvenot and Morfin, 1983; Liu et al., 1985; Warner et al., 1986; Dixon et al., 1987; Teshima et al., 1990; Bennett et al., 1994). Compared with other methods, immunoassays have several advantages for rapid field tests including high specificity, sensitivity, facile sample preparation and ease of use (Pestka et al., 1982). However, the development of antibodies requires the use of animals, specialized cell culturing facilities, and an extensive commitment of time and labor. Production of monoclonal antibodies from hybridoma cell lines also requires specialized cell culture facilities and usually is time consuming. 39 Advances in the field of recombinant antibody technology provide an alternative means to engineer low cost antibodies with desirable affinity and specificity by enabling one to manipulate the basic domain structure of the immunoglobulin (lg) molecule. One of the most successful approaches is to display scFv antibodies on filamentous phage (McCafferty et al., 1990; Barbas et al., 1991 ; Clackson et al., 1991; Marks et al., 1991; 1992; Griffiths et al., 1993). ScFv is an antigen-binding protein, composed of an immunoglobulin heavy chain variable domain (VH) and a light chain variable domain (Vk or V,) joined together by a flexible peptide linker (Figure 1.4). When expressed with phage protein g3p (fd 93 protein of phage M13K07) as a fusion protein, a high affinity scFv producing phage clone can be enriched by a procedure called "panning" (Marks et al., 1991 ; Hawkins et al., 1992). In this part, we describe the molecular cloning, expression and characterization of a functional scFv with high specificity and sensitivity to zearalenone and its analogues, and the application of this scFv to the analysis of zearalenone in spiked corn samples. 40 MATERIALS AND METHODS Materials, strains and general methods All chemicals and organic solvents were reagent grade or better. Zearalenone was generously supplied by lntemational Minerals and Chemicals Corp. (T erre Haute, IN). Zearalenone-ovalbumin and -keyhole limpet hemocyanin (KLH) conjugates were prepared as described by Dixon et al. (1987) (Figure 2.1 ). Anti-zearalenone hybridoma cell line 2GS-6E3-2EZ (Dixon et al., 1987) was kindly supplied by M. A. Abouzied (Neogen Company, Lansing, MI). Plasmid pCANTAB5E, E. coli TG1 and HB2151, M13K07 helper phage, horseradish peroxidase conjugated sheep anti-M13 antibody and mouse anti-E tag antibody were obtained from Phannacia Biotech (Piscataway, NJ). Cell culture components and reagents, goat anti-mouse lgG peroxidase and rabbit anti-mouse lgG alkaline phosphatase were purchased from Sigma Chemical Co. (St. Louis, MO). Fetal bovine serum, oligo(dT) cellulose columns and Not I restriction enzyme were supplied by Gibco BRL (Gaithersburg, MD). PCR amplification primers were synthesized by Gibco BRL(Gaithersburg, MD). The restriction enzyme Sfi I was purchased from New England BioLabs (Beverly, MA). All DNA manipulations, if not described, were carried out by standard procedures (Sambrook et al., 1989). 41 OH 0 Me O HO O Carboxymethoxylamine Zearalenone hemihydrochloride Zearalenone-oxime 3 HO—N NHS (RI-N=C=N—R1) 2— EDC ’\ NOCHz| —O— N OH ZRNHz (Protein) 140* I II NOCHzZ—N—R + HO—N Zearalenone-protein conjugte 2 Figure 2.1. Preparation of zearalenone immunogens through oxime chemistry (based on Thouvenot and Morfin, 1983). 42 mRNA isolation Hybridoma cell line 263-6E3-2EZ producing anti-zearalenone antibody (Dixon et al., 1987) was cultured in macrophage conditioned DMEM medium containing 20% (v/v) fetal bovine serum supplemented with 100 U/ml penicillin/streptomycin in a 5% CO2 humidified chamber. After the cells were grown to a density of 10° cells/ml, they were pelleted by centrifugation at 1000 x g for 5 min. For total RNA isolation, the pelleted cells were homogenized in RNA STAT-60 (TEL-TEST "B”, Inc., Friendswood, TX) at 5x10° cells/ml, followed by chloroform extraction and isopropanol precipitation. mRNA was purified by affinity chromatography with an oligo (dT)—cellulose column according to manufacturer instructions. cDNA synthesis, PCR amplification and scFv cloning First strand cDNA was synthesized from mRNA template with M-Mulv reverse transcriptase and random hexadeoxyribonucleotide [pd(N),,] primers (Phannacia Biotech, Piscataway, NJ). The variable regions of heavy chain (VH) and kappa light chain (VK) were amplified from first strand cDNA using Taq DNA polymerase with 30 cycles of PCR (94°C, 1 min; 55°C, 1 min; 72°C, 2 min). The primers used in the PCR amplification were based on Orlandi et al. (1989) and Clackson et al. (1991), and modified for facilitating ligase-free UDG-ligation (Nisson et al., 1991 ) (Table 2.1). VHBACK and du-VHFOR were used for amplification of V“; dU-VKBACK and VKFOR were used for amplification of VK. A 93—bp DNA linker (Phannacia Biotech, Piscataway, NJ), containing a sequence encoding a short 43 bL re f0 pr flexible peptide, (Gly4Ser)3, was also amplified with LINKBACK and du-LINKFOR. After gel purification with a QIAEX gel extraction kit (QIAGEN Inc., Chatsworth, CA), 25ng linker DNA and 75ng VK DNA were mixed and treated with 1 unit uracil DNA glycosylase (UDG) (USB, Cleveland, OH) at 37°C in a 20ul reaction with 1x PCR buffer for 1.5 hr. The UDG treated fragments were then scaled up to a 40pl PCR reaction without primers and cycled 15 times (94°C, 1 min; 60°C, 1 min; 72°C, 2 min) for joining the linker DNA with VK DNA. The joined linker-VK DNA was amplified in a 100pl PCR reaction (94°C, 1 min; 60°C, 1 min; 72°C, 2 min) for 30 cycles with 75 pmol each VKFOR and du-LINKBACK. Gel-purified linker-VK DNA fragment (40 ng) was joined with VH DNA (40 ng) into scFv (VH-Iinker-VK) DNA fragment in a 20 pl PCR fill-in reaction after UDG treatment. The assembled scFv DNA was amplified with 75 pmol each VHBACK and VKFOR in a 100 pl PCR amplification reaction for 30 cycles as described above. Finally, the assembled scFv products were gel- purified and reamplified with restriction site tagged primers (RS Primers Mix) (Phannacia Biotech, Piscataway, NJ) to append an Sfil site on the 5' end and a Notl site on the 3' end of the scFv DNA. The scFv DNA products were digested with Sfil and Notl restriction enzymes, gel purified and then ligated into the phagemid pCANTAB5E (digested with these same enzymes). 44 Table 2.1: Sequences of oligonucleotide PCR primers for PCR amplification of V", VK, and linker DNA* VHBACK: 5' AGGTSMARCTGCAGSAGTCWGG-3' dU-VHFOR: 5' UGAGGAUACGGUGACCGUAGUACCUUGGCCCC-3' dU-VKBACK: 5' GACAUCGAGCUCACUCAGUCUCCA—S' VKFOR: 5' CCG'ITI'BAKYTCCARC'ITKGTSCC—3' LINKBACK: 5' GGTACTACGGTCACCGTATCCTCAGGTG-3' dU-LINKBACK: 5' GGUACUACGGUCACCGUAUCCUCAGGUG-3' dU-LINKFOR: 5' AGACUGAGUGAGCUCGAUGUCCGAUCC-3' *: U=deoxyuracil; R=A or G; Y=C or T; M=A or C; K=T or G; S=C or G; W=A or T; B=T or C or G; 45 ScFv antibody expression and panning-elution selection To display scFv as a fusion protein with E-tag and M13 g3p (there is an amber stop codon between E-tag and fd 93 in pCANTAB5E), the ligated products were used to transform E. coli TG1 competent cells which is an amber suppressor strain (supE). Transformed cells were plated on SOB medium (Sambrook et al., 1989) containing 100 pg/ml ampicillin and 2% glucose and incubated at 30°C overnight. Colonies were pooled and infected with M13K07 helper phage in 2x YT medium (Sambrook et al., 1989) containing 100 pg/ml ampicillin and 50 pg/ml kanamycin to rescue the phagemid with its scFv gene inserts and to display scFv fusion protein on the surface of the recombinant phages. The recombinant phages from the supernatant were filtered through a 0.45 pm-pore-size filter. For panning-elution selection (Figure 2.2), wells of sterile polystyrene lmmulon-4 microtiter plates (Dynatech Laboratories, Inc., Chantilly, VA) were coated overnight (4°C) with 100 pl of zearalenone-ovalbumin conjugate (10 pg/ml) in 0.05 M carbonate-bicarbonate buffer (pH 9.6). Plates were washed four times by filling each well with 300 pl of 0.05% (v/v) Tween-20 in 0.01 M PBS (PBS-T) and aspirating the contents. Non-specific binding was blocked by addition of 300 pl “blocking buffer" [1% ovalbumin (w/v) dissolved in PBS (PBS-OVA)] to each well for 1 hr at 37 °C and then washing 6 times with PBS-T. Recombinant phages (about 5 x 1010 pfulml) were then added to the walls (100 pllwell) of a zearalenone- ovalbumin coated plate. The plates were incubated for 2 hr at 37°C followed by ten washes with PBS and ten washes with PBS-T. One hundred microliters of free 46 Fic lie-Infection and re-empllficetlon Figure 2.2. Panning-elution selection procedure for anti-zearalenone scFv phage in zearalenone-ovalbumin (ZEN-OVA) coated lmmunlon-4 microtiter wells. 47 268 OOH vvnl syn vver affii 68C liEi FHBJ gfll fliej adc 0(3c set; thrc detr (3or and "Her zearalenone (0, 0.01, 0.1, 1.0, 5.0, 10.0, 15.0, and 50.0 pg/ml) diluted with PBS containing 1.5% methanol were added to the wells and incubated at 37 °C for 1 hr with rotary shaking (150 rpm) to elute the bound phages. The zearalenone eluted phages were collected and passed through a 0.45 pm-pore-size Iow-protein-binding syringe filter (Gelman Sciences, Ann Arbor, MI). These zearalenone specific phages were used to reinfect E. coli TG1 cells for subsequent rounds of selection. The affinities of recombinant phage antibodies from pooled or individual colonies after each round of selection was tested with a modified Cl-ELISA described below. Recombinant phages from selected clones were used to infect E. coli HBZ151 for production of soluble scFv antibodies. Briefly, the infected E. coli HB2151 cells were cultured in SB medium (35 g/l tryptone, 20 g/l yeast extract, 5 g/l NaCl) supplemented with 100 pglml ampicillin at 30°C with 250 rpm shaking until they reached an A600 of 0.5. lsopropyl-B-D-thiogalactopyranoside (IPTG) was then added to 1 mM final concentration and the cells were incubated on a shaker at 30 °C overnight. Supematants containing the extracellular soluble scFv antibodies were separated from the cell pellets by centrifugation at 1500 x g for 15 min, and filtered through a 0.45 pm-pore-size filter. The affinity of soluble scFv antibodies was detected with a modified Cl-ELISA described below. Competitive Indirect ELISA (Cl-ELISA) Several Cl-ELISAs were developed and used to determine the sensitivities and specificities of the scFv antibodies (phage-associated or soluble forms). Briefly, microtiter plates were coated and blocked as described above. Next, 50 pl of 48 zearalenone standard (or analogues) at various concentrations dissolved in PBS containing 1% methanol was added to the coated wells and simultaneously incubated with 50 pl of phage-associated (about 5 x 1010 pfulml in 2x YT medium) or soluble scFv antibody (various dilutions with PBS-OVA) or parent monoclonal antibody (diluted 1:5 with PBS-OVA) from the supernatant of hybridoma cell culture for comparison. The plates were incubated for 1 hr at 37 °C then washed 6 times with PBS-T. The amount of bound phage-associated scFv antibody was determined by the addition of 100 pl anti-M13-HRP conjugate diluted 1:2500 with PBS-OVA. The amount of soluble scFv antibody bound was determined by incubation with 100 pl mouse anti-E tag (diluted 122500 with PBS-OVA) at 37°C for 1 hr followed by washing (six times) and a subsequent incubation with goat anti-mouse lgG HRP conjugate (diluted 1:500 with PBS-OVA). The bound monoclonal antibody was also detected by the addition of 100 pl goat anti-mouse lgG HRP conjugate (diluted 1:500 with PBS-OVA). The plates were incubated for 1 hr at 37°C and then washed 6 times with PBS-T. The amounts of anti-M13-HRP and goat anti-mouse IgG HRP conjugates bound were assessed by the addition of 100 pl of 2,2'-azinobis(3- ethylbenzthiazoline sulfonic acid) (ABTS) substrate (Pestka et al., 1982). The plates were incubated for 15 min for color development and the absorbances were determined at 405 nm with a microtiter plate reader (Molecular Devices Corporation, Menlo Park, CA). 49 ar fol ins tOIl Vis SDS-PAGE and Western blot analysis Supematants, containing the extracellular soluble scFv antibodies in SB medium from E. coli HB2151 cell culture, were precipitated with trichloroacetic acid (T CA) at a final concentration of 10% (w/v) on ice for 20 min. After centrifugation in a microcentrifuge at 14,000 rpm for 10 min, the pellet was resuspended in 15 pl of 0.5 M Tris buffer (pH 8.0), heated for 5 min at 95°C after adding 5 pl loading buffer (4 x), and subjected to SDS-12% polyacrylamide gel electrophoresis with a 0.5 mm thick gel in a Bio-Rad Mini-Protein l| Eletrophoresis System (Bio-Rad Laboratories, Hercules, CA). Prestained low range SDS-PAGE standards (Bio-Rad laboratories) were used to calibrate protein mobilities. Two identical gels were run in parallel. After separation, the protein bands in one gel were stained with Coomassie brilliant blue G-250, those in the other gel were transferred to a nitrocellulose membrane (Schleicher & Schuell, Kneene, NH) in a Multiphor II Electrophoresis System (LKB Produkter AB, Bromma, Sweden). The transblotted membrane was probed with anti-E tag antibody (about 8 pg/ml in PBS-OVA containing 0.05% Tween 20) followed by incubation with rabbit anti-mouse lgG alkaline phosphatase conjugate (Sigma Chemical Company, St. Louis, MO) as described by manufacturer’s instructions. Nitroblue tetrazolium and 5-bromo-4-chloro-3- indolyphosphate-p- toluidine (NBT/BCIP)(Pierce, Rockford, IL) were used as the substrate for visualization. 5O Sequence analysis Plasmid DNA of anti-zearalenone producing clones was isolated from E. coli HBZ151 by alkaline lysis. VH and VK DNA portions in the plasmid were sequenced on both strands with the pCANTAB5 sequence primer set (Phannacia Biotech, Piscataway, NJ) by using Taq cycle sequencing and dye terminator chemistry at the MSU Sequencing Facility. Sequence comparisons were performed by searching the Kabat Database at the Internet Web site (http:/limmuno.bme.nwu.edu). Cl-ELISA for spiked corn extracts The procedures for extraction of corn and spiking were similar to those described by Liu et al. (1985). Briefly, zearalenone-free homogeneous ground corn was extracted with five volumes (w/v) of methanol-water (70:30) for 5 min, with constant stirring. The mixture was immediately filtered through a Whatman No. 4 filter paper and the filtrate was spiked directly with zearalenone, or the filtrate was diluted 2 times with 1x PBS and then spiked with zearalenone. The spiked extracts were then subjected to Cl-ELISA using the soluble scFv antibodies and monoclonal antibody in a similar procedure as described above with the following modifications. (i) Since anti-E-tag antibody, which was used as a second antibody to detect the bound scFv antibodies, exhibited nonspecific binding to methanol treated carrier protein ovalbumin, zearalenone-KLH conjugate was used in the Cl-ELISA for methanol extracted corn samples. (ii) Coated wells were blocked by PBS containing 10% nonfat dry milk. (iii) Antibodies were diluted in PBS containing 10% 51 nor pFC Nu Ul’l nonfat dry milk. The spiking experiment was repeated once with the same procedure. Nucleotide sequence accession numbers The sequences determined for VH and VK from this clone are available under GenBank database accession numbers U74671 and U74672, respectively. 52 RESULTS PCR amplification VH and VK cDNA from hybridoma cell line 2GS-6E3—2EZ were amplified by PCR and assembled into an scFv encoding DNA fragment (Figure 2.3). Amplification of VK generated a major DNA fragment with the expected length (about 324 bp), while VH generated an expected 340 bp fragment and two other closely migrating fragments. The PCR assembly of linker-Vk produced two major closely migrating fragments (lane 5 in Figure 2.3) with approximately the expected size plus a minor band much larger than the expected product. The two major linker-Vk fragments, which might be generated by alternative priming of the linker DNA with its nearly identical repeats, were collectively isolated, purified, and then PCR assembled with VH products (the mixture of three major bands in lane 2 of Figure 2.3) to form the scFv fragments in lane 6 of Figure 2.3. For the assembly of scFv, a preliminary experiment with a one step PCR fill-in reaction failed to join VH, linker, and VK . The modified two-step procedure described in Materials and Methods greatly improved the joining. 53 Figul 1234567 scFv "" 750bp Figure 2.3. VH and VK amplification and assembly of scFv. Lanes are: (1): VH marker (pUC18NH, EcoR l/Xba | digested, Pharmarcia Biotech); (2): VH PCR product; (3): VK PCR product; (4): Linker PCR product; (5): Linker-VK PCR product; (6): VH-Linker-VK (scFv) PCR product; (7): scFv marker (pUC18/A1OB, Sfi l/ Not I digested, Pharrnacia Biotech). Phag town wdht used stop I fluap flfllE aHou wens eune phag after PUC FigL Phage display of scFv antibody and panning-elution selection Two major scFv DNA fragments in lane 6 of Figure 2.3 were collectively isolated, purified, digested, then ligated into pCANTABSE, and translationally fused with the E-tag DNA fragment (Figure 2.4). The resulting library, named pQY1, was used to transform E. coli TG1 cells (amber suppression strain), in which the amber stop codon between the E-tag DNA sequence and fd 93 was read through, allowing the production of scFv E-tag-g3p fusion protein. In the presence of helper phage (M13K07), scFv fusion products were displayed on the recombinant phage tips, allowing for affinity selection with panning-elation. In lmmulon-4 microtiter plate wells coated with zearalenone-ovalbumin, the bound recombinant phages were eluted by free zearalenone at a minimum concentration of 1 pg/ml. The eluted phages were used for elution in the sequential round of panning-elution selection after re-amplification. TAG (amber stop codon) 133 Figure 2.4. pQY1 construct. PCR amplified scFv was cloned into Sfil and Nail sites of pCANTAB5E, a pUC119 based vector from Phannacia Biotech. The linker DNA sequence was 5'GGTGGAGGCGGTTCAGGCGGAGGTGGCTCTGGCGGTG GCGGATCG-3', which encoded a short peptide [(Gly4Ser)3]. 55 low : a ole (Figl bind rour rour eac isol: am; iron dist binr fror Sol bet SOIL r90 bInt Of 5 IPT Prior to affinity selection, the pooled recombinant phage library exhibited very low zearalenone-binding activity (Figure 2.5). After one round of affinity selection, a clear enrichment for scFv clones expressing zearalenone-binding was observed (Figure 2.5). The lC50 (concentrations of free zearalenone required for inhibition of binding by 50%) for preselection phage, first round selected phage and second round selected phage were >1000, 31, and 28 ng/ml, respectively. The second- round selection mainly increased yield of functional scFv fusion protein. Following each round of panning-elution selection, individual clones (in TG1) were randomly isolated and the scFv DNA insert in these clones was confirmed by PCR amplification and restriction digestion with Sfil and Notl. Three individual clones from each selection group were rescued with helper phage M13K07 for scFv phage display. When the resultant recombinant phages were tested for zearalenone binding in CI-ELISA, all three preselection clones were negative whereas those from first and second round selection were positive. Soluble scFv antibody production and characterization In the nonsuppressor strain, E. coli HB2151(supE'), the amber stop codon between E- tag and fd 93 in scFv clones is recognized as a stop codon and a soluble scFv-E tag fusion protein is produced as a consequence. Three individual recombinant phage clones (the same clones tested above for zearalenone binding) from each selection group were used to infect HB2151. The expression of scFv E-tag fusion protein in the resultant HB2151 clones was induced by IPTG, and soluble scFv antibodies were secreted to the culture supematants. 56 Fig -O—pre-selection —A— lst round selection 0.8 ~ + 2nd round selection 0.6 - A405 0.4 i 0.2 .. 1 10 100 1000 10000 Zearalenone (ng/ml) Figure 2.5. Zearalenone-binding activities of recombinant phage following each round of panning-elution selection measured by CI-ELISA in zearalenone-ovalbumin coated lmmulon—4 microtiter wells. Bound phages were detected by horseradish peroxidase conjugated sheep anti-M13 antibody. 57 A405 0 Fig Figure 2.6. 120 -<>-(IY1.I ‘x‘QYl-Z 100 - +QY1.12 +QY1.12 -O— . -O—QY1.5 QY1 5 -HD-nukb 80- 60- Percent of inhibition l 10 100 1000 I 10 100 1000 Zearalenone (ng/ml) Zearalenone (ng/ml) Sensitivity of soluble scFv antibodies tested in Cl-ELISA in zearalenone-ovalbumin coated Immulon-4 microtiterwells. QY1 .1 and QY1.2 ( no dilution) were from preselection; QY1.5 (5x dilution) was from first round of panning-elution selection; QY1.12 (10 x dilution) was from second round of panning-elution selection; mAb (supernatant of hybridoma cell culture) was diluted 5 times. Bound soluble scFv antibodies were detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse lgG. A: Absorbance reading at 405 nm. B: Percent of binding inhibition determined by dividing sample absorbance by control absorbance x 100. 58 Table 2.2: Comparison of cross-reactivities between scFv antibody and mAb toward zearalenone analogues mAb scFv QY1.5 Analogue IC50 Cross-reactivity* IC50 Cross-reactivity (no/ml) (%) (no/ml) (%) Zearalenone 17 100 14 100 a-zearalenol 66 26 17 82 B-zearalenol 159 1 1 54 26 a-zearalanol 212 8 23 62 B—zearalanol 175 10 54 26 *: Cross-reactivity defined as (lemmbm)/( leo mm”) x 100 %. 59 In In The zearalenone-binding pattern of soluble scFv antibodies from these nine HB2151 clones was the same as their parent phage scFv antibodies. Cl-ELISA results for four typical scFv clones are shown in Figure 2.6A. Two of the scFv antibodies (QY1.5 and QY1.12 from first round and second round selection, respectively) exhibited zearalenone-binding affinities similar to those of the parent monoclonal antibody (Figure 2.6B). leo for scFv QY1.5, QY1.12 and their parent monoclonal antibody were 14, 35, 17 ng/ml, respectively. Compared with the parent monoclonal antibody, however, soluble scFv QY1.5 exhibited similar sensitivity but higher relative cross-reactivities to hydroxylated zearalenone analogues (Table 2.2). SDS-PAGE and Western blot analyses An approximately 32 KDa scFv-E tag fusion protein was expressed from HB2151/pQY1.2, HB2151/pQY1.12 and HB2151/pQY1.5, while HB2151/pQY1.1 expressed a smaller protein, which probably lacked an E tag and could not be detected by anti-E tag antibody (Figure 2.7). Among the two positive zearalenone binding clones, HB2151/pQY1.12 expressed higher yields of scFv than HB2151/pQY1.5; however, the affinity of its scFv antibody (QY1.12) for zearalenone appeared slightly lower than that of QY1.5 (Figure 2.5B). Sequence analysis Clone pQY1.5, producing scFv with the highest affinity for zearalenone in HB2151, was chosen for DNA sequencing. The deduced amino acid sequences of VH and VK in pQY1.5 are shown in Figure 2.8. According to the amino acid 60 Figure 2.7. SDS-PAGE gel and Western blot analysis of scFv expression. (A): Supematants of QY1.1 (lane 1; 0.3 ml), QY1.2 (lane 2; 0.3 ml), QY1.12 (Iane3; 0.3 ml) and QY1.5 (lane 4; 0.6 ml) concentrated in 10% trichloroacetic acid were subjected to SDS-PAGE and stained with Coomassie brilliant blue G-250. (B): Western blot of duplicate SDS/PAGE gel. 61 sequences, the VH was classified as mouse heavy chain subgroup lll (D), while VK fell into mouse kappa light chain subgroup V (12) (Kabat et al., 1991). The sequences of VH and VK were screened against the entries of lg heavy chain and lg kappa light chain respectively in the Kabat Database. The VK sequence was closely related to that of a genomic DNA clone VK23.32, a member with no identified binding specificity in gerrnline gene VK23 family (Lawler et al., 1989). There was 97% nucleotide sequence identity between VK of pQY1.5 and VK23.32 with only 9 nucleotide differences, resulting in 4 amino acid changes. Among the 9 changed nucleotides, 4 were in the framework region (FW) 1, and were attributed to the use of a degenerate VKBACK primer, 3 were in FW3, and a single silent nucleotide change (causing no amino acid sequence change) occurred in both complementary- detennining regions (CDR) 2 and 3. This implied that V, 23.32 might be the germ line gene for Vk of scFv QY1.5. Thus, few somatic mutations in the kappa light chain occurred during the in vivo affinity maturation of its parent anti-zearalenone antibody. However, the sequences of CDRs (especially CDR2 and CDR3) in the VH of pQY1.5 were not related to any known members in the database. The closest VH sequence was that of L3 10A (Kettleborough et al., 1994), an antibody with specificity to human epidermal growth factor receptor. But there was only 68% and 47% sequence identity between scFv OY1.5 and L3 10A in their VH CDRs at the nucleotide and amino acid levels, respectively, although they had 96% identity in the FWs at both nucleotide and amino acid levels. 62 DlELTc GIPSR] Figul A. VH sequence: FWl CDRI FW2 CDR2 MA QVKLQESGGGLVQPGGSLKLSCAASGFTF S NYGMS WVRQTPDKRLEFVA NINGNGGKTYYPGSVKG FW3 CDR3 FW4 RFTISRDNAKNTLYLQMSSLKSEDTAMYYCVR VAFDGYYDDF WGQG'ITVTVSS B. VK sequence: FWl CDR] FWZ CDR2 DIELTQSPATLSVTPGDRVSLSC RASQSISDYLH WYQQKSHESPRLLIK YASQSIS FW3 CDR3 FW4 GIPSRFSGSGSGSDFTLSINSVEPEDVGVYYC QNGHSFPPT FGGGTKLEIK Figure 2.8. Deduced amino acid sequences of VH and VK in scFv QY1.5. Complementary-determining regions (CDR) and framework regions (FW) are indicated. 63 AP QY mc EL 28 to 26 3F Application of scFv QY1.5 to a spiked corn extract Using zearalenone-KLH conjugate coated microtiter plates, soluble scFv QY1.5 showed higher sensitivity to zearalenone in corn extracts than its parent monoclonal antibody when the final methanol concentration was 17.5% in the Cl- ELISA solution (Figure 2.9). Corn extracts containing as little as 10 ng zearalenone/ml caused a visually distinct inhibition of absorbance with soluble scFv OY1.5 in Cl-ELISA. However, for undiluted corn extract (35% methanol in the final CI-ELISA solution), soluble scFv QY1 .5 failed to detect any level of zearalenone in Cl-ELISA because zearalenone specific binding of QY1.5 was eliminated by methanol at concentrations higher than 25% in the final Cl-ELISA solution (Figure 2.10). In contrast, the parent monoclonal antibody exhibited much highertolerance to methanol destabilization (Figure 2.10). Regardless, scFv QY1.5 did bind zearalenone with high specificity and sensitivity at practical levels of methanol concentration (less than 20%). Thus, the scFv antibody described here should be applicable for routine screening of zearalenone in corn and perhaps other cereal grains. 100 Percent of inhibition Figure 2.9. 80‘ 60~ 4o~ 20- 0.1 l 10 100 1000 10000 Zearalenone (ng/ml) Zearalenone sensitivities in spiked corn extracts with monclonal antibody and soluble scFv antibody OY1.5 measured by Cl-ELISA in zearalenone-keyhole limpet hemocyanin conjugate coated Immulon-4 microtiter wells. Both scFv QY1.5 and mAb were diluted 1:4 in PBS containing 10% nonfat dry milk. Zearalenone in ground corn was extracted with 5 volumes (wt/vol) of methanol-water (70:30) and the extracts were then diluted 1:1 with PBS. 65 A405 0 I I I I I I 0 5 10 15 20 25 3O 35 Methanol concentration in test well, % Figure 2.10. Effects of methanol on the binding of mAb and scFv QY1.5 to zearalenone-keyhole limpet hemocyanin conjugate. lmmulon-4 microtiter wells were coated with zearalenone-keyhole limpet hemocyanin conjugate. Bound soluble scFv antibody was detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse lgG. Bound monoclonal antibody was detected by horseradish peroxidase conjugated goat anti-mouse I96. 66 from and succ and ' anut anflt fort proc i~hk very dev. Dior mR "Tel 96o 98v an” all DISCUSSION This study describes the successful isolation and cloning of VH and VK genes from a zearalenone-specific hybridoma, their soluble expression as an scFv protein, and their use in a zearalenone Cl-ELISA. To our knowledge, this is the first successful example of the development of a recombinant antibody for mycotoxins, and we think it could serve as a model for development and construction of novel antibodies for mycotoxins and other potentially toxic residues. When a hybridoma is used as a mRNA source for construction of scFv antibody, it is most desirable to use a cell line that produces high-affinity antibody for the target hapten or antigen. The hybridoma cell line we used in this study produced monoclonal antibody with high affinity (le0=17 ng/ml) for zearalenone, which made the cloning of high affinity scFv antibody possible. In contrast, only a very low sensitive scFv antibody specific for the mycotoxin fumonisin B, was developed in a previous study because, in part, the hybridoma cell line used produced a lower affinity antibody specific to fumonisin B, (Zhou et al., 1996). Although hybridomas producing high affinity antibodies are good sources of mRNA for the facile cloning of desired variable region (V region) genes, additional irrelevant transcripts are possible. Thus, it was notable that amplification of the VH gene from hybridoma 2G3-6E3-2EZ, which produces anti-zearalenone antibody, gave three different PCR products. One of these products may have been the amplified product from the fusion partner, myeloma cell line NS-1, which produces an aberrant V,, transcript with a 50 nucleotide deletion at FW3-CDR3 boundary 67 (Thammana, 1994). Other irrelevant V regions might possibly result from non- productive rearrangements. Practically, a hybridoma may contain several different cell populations as a result of recombination during extended culture after limiting dilution. So V regions which are productive but do not recognize the antigen of interest may also be isolated. Additionally, modifications could be generated in the PCR amplification and assembly because of the use of degenerate primers and Taq DNA polymerase with low fidelity. Affinity selection was therefore necessary for isolation of the variable regions with the desired specificity. Like the scFv antibodies before panning elution selection in this study, the fumonisin B, specific recombinant scFv antibody (Zhou et al., 1996), selected only by panning without affinity elution selection (through hapten elution), were 10 to 100 times less sensitive compared with anti-fumonisin B, polyclonal or monoclonal antibodies. Clackson et al. (1991) also showed that affinity elution with free antigen or hapten elution after panning greatly enriched clones producing high affinity scFv antibody. Since only a few species of mRNA transcripts from a specific hybridoma cell line contain V regions, a few rounds of panning-elution selection should select the desired V regions. In this study, a functional anti-zearalenone scFv protein with high affinity was isolated after only one round of panning-elution selection. Sequential selections mainly appeared to enrich clones with a higher yield of scFv protein, not necessarily improving the affinity. Theoretically, a scFv will have similar specificity and sensitivity as its parent immunoglobulin molecule. In this study, soluble scFv antibody QY1.5 exhibited an affinity to zearalenone similar to that of its parent monoclonal antibody, but it was 68 showr these at al.. partic gene and SBQI reve seqr COV to t of s effi of de ml at shown to have higher cross-reactivity to hydroxylated zearalenone analogues. Since these analogues are naturally occurring toxic metabolites of zearalenone (Mirocha et al., 1981; Hagler et al., 1979), the ability of the scFv antibody to detect them is particularly advantageous when detection of these analogues is desirable. In general, a change in specificity and sensitivity could result from sequence changes and conformation differences between scFv and the monoclonal antibody. Sequence changes may occur during the scFv construction procedures, including reverse transcription, PCR amplification, enzyme digestion and ligation. Even if the sequences of V,, and VK in the scFv are the same as those naturally occuring in an lgG molecule, their conformations may also differ because scFv has V,, and VK covalently coupled by a flexible peptide linker while natural lgG molecule does not. ScFv QY1 .5 also differed from its parent monoclonal antibody in its sensitivity to the destabilizing influence of methanol, which drastically decreased the binding of scFv QY1.5 to zearalenone. Muller et al. (1994) observed a similar destabilizing effect of glycerol on scFv, which promoted the dissociation of two variable domains of scFv protein but did not affect the domain structure of scFv. In contrast, destabilization was not observed for Fab fragments and whole monoclonal antibody molecules in that study. It was believed that the constant domains in Fab or monoclonal antibody may provide additional stabilization of the molecular structure at the antigen binding site. Some general strategies for protein stabilization may be used to stabilize scFv protein by strengthening the interactions between VH and VK domains. These include engineering of stronger hydrogen bonds, disulfide 69 bonds, metal binding sites, and increasing the hydrophobic area or electrostatic interactions (Raag and Whitlow, 1995). It was notable that the VH sequence of scFv QY1.5 exhibited numerous sequence differences from sequenced members in the antibody database. As might be expected, the changes are clustered in the CDRs, especially in CDR2 and CDR3. This implies that CDR2 and CDR3 in the V,, of scFv QY1.5 may play an important role in determining the specificity for zearalenone binding. In contrast, the VK in scFv QY1.5 may contribute less to the specificity, since there were few differences in its sequence compared with its possible germ line gene. The latter may have a broad range of specificities without somatic mutations. As immunoassays become a more common method forthe routine screening of mycotoxins and other natural toxicants in food and agriculture products, larger supplies of low-cost high affinity antibodies will be needed. The recombinant antibody technique provides a promising alternative for producing low-cost antibodies with the desirable specificity and sensitivity. The recombinant antibody scFv QY1.5 has high sensitivity and specificity to zearalenone and its analogues, and should be readily applicable to the routine screening for these mycotoxins. 70 PART III ATTEMPTS TO CLONE SINGLE-CHAIN FV (SCFV) ANTIBODIES AGAINST FUMONISIN B, FROM IMMUNIZED MICE 71 ABSTRACT ATTEMPTS TO CLONE SINGLE-CHAIN FV (SCFV) ANTIBODIES AGAINST FUMONISIN B, FROM IMMUNIZED MICE High affinity (le0, 0.27 pg fumonisin B, lml) mouse polyclonal antibodies were readily generated after immunization with fumonisin B, -cholera toxin (FB,-CT) conjugate but not produced with fumonisin B,-keyhole limpet hemocyanin (FB,-KLH) conjugate immunization. From the splenic cDNA of FB,-CT immunized mice, variable region genes of heavy- (VH) and kappa light-chain (VK) were successfully amplified by PCR with previously designed primers (Yuan et al., 1997). The V,, and VK genes were assembled with linker DNA to form scFv DNA fragments by PCR and cloned in a phage-display vector. However, panning-elution selection from this scFv phage-display library did not result in enrichment of PB, specific phage clones. Different FB, conjugates, phage incubation times and F3, concentrations in the eluting solution were investigated for their effects on specific binding and elution. No conditions were found suitable for FB, specific scFv phage selection in the panning-elution selection procedure. The soluble form of scFv proteins from approximately 300 randomly selected individual clones did not show any FB, specific binding activity either. An alternative method is suggested to clone functional FB, specific scFv antibodies. 72 INTRODUCTION Fumonisins are toxic secondary metabolites produced by certain species of Fusarium during pathogenesis on corn and sorghum (Sydenham et al., 1990). Interest on fumonisins has intensified recently because of its cytotoxic properties and carcinogenic potential (T hiel et al., 1991; Gelderblom et al., 1993). Six different fumonisins, fumonisin A,, A,, B,, B,,, B,,, and B,, have been chemically characterized (Figure 1.2) (Bezuidenhout et al., 1988; Cawood et al., 1991; Plattner et al., 1992). Fumonisin B, (FB,) is the most toxic and primary fumonisin produced by Fusarium moniliforme (Gelderblom et al., 1988b). FB, produces characteristic effects in different animal species, including hepatic cancer in rats, brain lesions in horses (equine leukoencephalomalacia, ELEM), and pulmonary lesions in pigs (porcine pulmonary edema, PPE). Elimination of fumonisins from human and animal food requires detection and diversion of contaminated raw materials from feed and finished food use. Commonly used chemical analytical methods for detecting fumonisins are thin layer chromatography (TLC), high performance liquid chromatography (HPLC), and mass spectroscopy (MS). Fumonisins do not absorb ultraviolet light and do not fluoresce; therefore, derivatization reactions are required before detecting the toxins with chromatographic techniques (Geldeblom et al., 1988a; Shephard at al., 1990; Cawood et al., 1991; Rottinghaus et al., 1992; Scott and Lawrence, 1992). Regardless of sensitivity and selectivity, these chemical analytical methods are generally expensive, complex, and time consuming (Pestka et al., 1995). More 73 recently, immunoassays have proven to be useful in screening for mycotoxins. Compared to the chemical analytical methods, immunoassays have several advantages for rapid field tests, including high specificity, sensitivity, facile sample preparation, and ease of use (Pestka et al., 1995). As a consequence, both polyclonal (Azcona-Olivera et al., 1992a) and monoclonal (Azcona-Olivera et al., 1992b) antibodies to FB, have been developed and applied to the enzyme-linked immu nosorbent assays (ELISAs) (Elissalde et al., 1995; F ukuda et al., 1994; Pestka et al., 1994a; Tejada-Simon et al., 1995). Such developments have lead to a great demand for antibodies in the fumonisin immunoassays. However, the limit of detection with most current antibodies is not sufficiently adequate for screening fumonisins at <1 ppm in conventional ELISA or multianalyte assays. Furthermore, the development of these natural antibodies requires the use of animals, specialized cell culturing facilities, and an extensive commitment of time and labor. It is now theoretically feasible to engineer low cost antibodies with desirable affinity and specificity characteristics by using recombinant DNA technology to manipulate the basic domain structure of immunoglobulin molecule (Hoogenboom et al., 1992; Lerner et al., 1992). Using filamentous phage to display antibodies, antibody fragments can now be engineered from antibody V-gene repertoires (McCafferty et al., 1990; Clackson et al., 1991; Marks et al., 1991) and high affinity clones can be selected by a procedure called “panning” (Marks et al., 1991; Hawkins et al., 1992). 74 Previously, several single-chain Fv clones were developed in our laboratory using mRNA from either spleen cells of mice immunized with FB,-bovine serum albumin (FB,-BSA) conjugate or from an existing hybridoma cell line that produces anti-FB, antibody (Zhou et al., 1996). However, these scFv antibodies had very low affinities to fumonisins and were even 10 to 100 times less sensitive than the parent natural antibodies. The failure to develop high affinity scFv antibodies to fumonisins in the previous study might be due to (i) the use of a hybridoma cell line or immunized mice which produced low affinity antibodies; (ii) only panning was used in the selection, without affinity elution. Based on the successful development of anti-zearalenone scFv antibodies (Yuan et al., 1997), it is desirable to use a cell line or immunized mice that produce high-affinity antibodies to start scFv fragments cloning. For the selection of specific scFv antibodies, it is also critical to conduct an affinity elution by free hapten after the regular panning. In this study, the possibility of developing high affinity scFv antibodies to fumonisin B, was investigated by using (i) spleen mRNA from immunized mice that produce high affinity antibodies to fumonisin 3,; (ii) affinity elution with free fumonisin B, after the regular panning. 75 MATERIALS AND METHODS General procedures All chemicals and organic solvents were reagent grade or better. Fumonisin B,, cholera toxin (CT), ovalbumin (OVA), bovine albumin (BSA), glutaraldehyde, goat anti-mouse lgG peroxidase and rabbit anti-mouse lgG alkaline phosphatase were purchased from Sigma Chemical Co. (St. Louis, MO). lnject" keyhole limpet hemocyanin (KLH) was purchased from Pierce Chemical Company (Rockford, IL). Plasmid pCANTAB5E, E. coli TG1 and HB2151, M13K07 helper phage, horseradish peroxidase conjugated sheep anti-M13 antibody and mouse anti-E tag antibody were obtained from Pharmacia Biotech (Piscataway, NJ). Oligo(dT) cellulose columns and Not I restriction enzyme were supplied by Gibco BRL (Gaithersburg, MD). PCR amplification primers were synthesized by Gibco BRL(Gaithersburg, MD). Restriction enzyme Sfil was purchased from New England BioLabs (Beverly, MA). All DNA manipulations, if not described, were carried out by standard procedures (Sambrook et al., 1989). Preparation of Immunogens Fumonisin B, was conjugated to KLH (FB,-KLH), cholera toxin (FB,-CT), ovalbumin (FB,-OVA), and bovine semm albumin (FB,-BSA) with glutaraldehyde linkage via a free amino group (Figure 3.1) as described by Azcona-Olivera et al. (1992a). FB,-KLH and FB,-CT conjugates were used as immunogens, and FB,- OVA and FB,-BSA conjugates were used as solid-phase antigens for affinity 76 selection. Briefly, the coupling reaction was carried out at 4°C in 0.01 M phosphate- buffered saline (pH7.4) (PBS). F3, was added to a 1-mglml suspension of protein at a molar ratio of 50:1 (toxin/protein), and then an equal volume of glutaraldehyde (2% vol/vol) was added dropwise with constant stirring. After 1 hr, the reaction was stopped and the bond stabilized by reduction via the addition of sodium borohyd ride to a final concentration of 10 mg/ml. One hour later, the mixture was dialyzed for 72 hr (three changes) against 0.01 M PBS (pH7.4). The conjugates then were aliquoted in fractions of 1 mg (total protein) and stored at -20°C until required. Animal Immunization Female BALB/c mice (7 to 8 weeks of age) were immunized by intraperitoneal injections. Mice immunized with FB,-KLH conjugate received three 100-pg doses of immunogen at 2-week intervals. The first injection consisted of 0.1 ml of conjugate in saline mixed with (1 :1 volume ratio) Freund’s complete adjuvant. The second and third injections consisted of 0.1 ml of conjugate in saline mixed with (1 :1 volume ratio) Freund’s incomplete adjuvant. Mice immunized with FB,-CT were injected three times (10-day interval) with 10 pg of immunogen dissolved in 0.2 ml of PBS. Ether-anesthetized mice were bled from the eye retrobulbar plexus, and serum was obtained after overnight incubation of blood at 4°C and centrifugation at 1,000 x g for 10 min. 77 COCHzCI-lCHzCOOH H P t ' 33'” ro etn ($th (glutaraldehyde) CHO COOH cocrizcncnzcoon 20 22 2' CH3 0 N—cr-rz—(CHm -—CH2—N ——Protein cocuzcr-lcnzcoon H Figure 3.1. Preparation of fumonisin B, conjugates with glutaraldehyde linkage via a free amino group (Pestka et al., 1995). 78 ELISA For antiserum titration, wells of polystyrene microtiter plates (lmmunolon-4- Removawells) (Dynatech Laboratories, Inc., Chantilly, VA) were coated overnight at 4°C with 10 pg/ml (100 pllwell) of FB,-OVA, FB,-BSA or FB,-KLH in 0.1 M sodium carbonate-bicarbonate buffer (pH 9.6). Plates were washed four times with 300 pl of PBS. The wells were blocked for 1 hr at 37°C with 320 pl of 1% (wt/vol) OVA, BSA or KLH in PBS and then washed four times with 300 pl of PBS. Serially diluted serum (100 pl) was then added to each well and incubated for 1 hr at 37°C. Unbound antibodies were removed by washing six times with 0.1% (vol/vol) Tween 20 in PBS (PBS-T), and 100 pl of goat anti-mouse immunoglobulin G peroxidase conjugate (diluted 1/2000 in 1% OVA-PBS or 1% BSA-PBS) was added to each well. Plates were incubated for 1 hr at 37°C and washed six times with PBS-T. The bound peroxidase was determined by the addition of 100 pllwell of TMB substrate (Sigma Chemical Company) and incubation at 37°C for 15 min. The reaction was stopped by adding 100 pllwell of 10% H2804. The absorbance at 450nm was determined with a microtiter plant reader (Molecular Device Corporation, Menlo Park, CA). A competitive indirect ELISA (Cl-ELISA) was used to assess the presence of FB, specific antibodies in mouse sera. Microtiter plates were coated and blocked as described above. A 50-pl volume of F B, (at various concentrations) dissolved in PBS was added over FB,-OVA (or FB,-BSA) coated wells, followed by addition of a 50-pl volume of antiserum (appropriately diluted in 1% OVA-PBS or 1% BSA- PBS) and incubated at 37°C for 1 hr. Bound antibodies were determined as 79 described above. Relative antibody affinity was arbitrarily designated as the toxin concentration required to inhibit antibody binding by 50% (I050). Spleen mRNA Isolation and cDNA synthesis Total cellular RNA was extracted by homogenizing the murina spleen tissue of immunized mice in RNA STAT-60 (T EL-TEST Inc., Friendswood, TX). The mRNA was purified by affinity chromatography on oligo (dT)—cellulose column according to the manufacturer’s instructions. First-strand cDNA was synthesized from the mRNA template with Moloney murina leukemia virus reverse transcriptase and random hexadaoxyribonucleotida [pd(N),,] primers (Phannacia Biotech). Random hexamers were used because they eliminated the need for lg-spacific primers or oligo (dT) primers that require synthesis of a long cDNA for obtaining the sequences encoding the variable heavy or light chain. PCR amplification and scFv cloning The variable regions of heavy chain (V,,) and kappa light chain (VK) were amplified from the first-strand cDNA as described previously (Yuan at al., 1997). Briefly, the variable regions of heavy chain (V,,) were amplified by using Taq DNA polymerase and VHBACK and du-VHFOR as primers (T able 2.1). The kappa light chains (VK) were amplified with dU-VKBACK and VKFOR as primers (T able 2.1). A 93—bp DNA linker (Phannacia Biotech, Piscataway, NJ), containing a sequence encoding a short flexible peptide, (Gly,Sar)3, was also amplified with LINKBACK and du-LINKFOR (Table 2.1). A two-step procedure described previously (Yuan at 80 al., 1997) was used for PCR assembly of scFv DNA fragment. The assembled scFv products were gel-purified and reamplifiad with restriction site tagged primers (RS Primers Mix) (Pharmacia Biotech) to add an Sfrl site on the 5' and and a Notl site on the 3' and of the scFv DNA. The scFv DNA products were then digested with Sfil and Notl restriction enzymes, gel purified and ligated into the phagemid pCANTAB5E (digested with these same enzymes). Affinity selection of phage-displayed antl-FB, scFv antibodies E. coli TG1 competent cells, an amber suppressor strain (supE ), were transformed by the ligated DNA products above and plated on SOB medium (Sambrook at al., 1989) containing 100 pg/ml ampicillin and 2% glucose, and incubated at 30°C ovamight. Colonies were pooled and infected with M13K07 helper phage as described previously (Yuan at al., 1997) to display scFv fusion protein on the surface of the recombinant phage. The recombinant phages from the supernatant were filtered through a 0.45 pm-pore-siza filter (Gelman Sciences) For panning-elution selection, wells of sterile polystyrene lmmulon-4 microtiter plates (Dynatech Laboratories, Inc., Chantilly, VA) were coated overnight (4 °C) with 100 pllwell of FB,-OVA or FB,-BSA or FB,-KLH conjugate (10 pg/ml) in 0.05 M carbonate-bicarbonate buffer (pH 9.6). The control wells were coated with the carrier proteins (10 pg/ml) alone. Plates were washed four times by filling each well with 300 pl of 0.01 M PBS and aspirating the contents. Non-specific binding was blocked by incubation at 37°C for 1 hr with 300 pl of 1% OVA, BSA or KLH (wlv) dissolved in PBS and then washing 6 times with 0.05% (v/v) Tween-20 in 0.01 81 M PBS (PBS-T). Recombinant phages (about 10° to 10'° pfulml) were then added (100 pllwell) The plates were incubated for various times (10 to 60 min) at 37 °C followed by ten washes with PBS-T and incubation with 300 pllwell of PBS at 37°C for 1 hr. The wells were then washed another ten times with PBS-T. One hundred microliter of free FB, (10, 50 and 100 pglml) diluted with PBS was added to the walls and incubated at 37 °C for 1 hr with rotary shaking (150 rpm) to elute the bound phages. PBS was used as a control alution reagent. The eluted phages were collected and passed through a 0.45 pm-pore-siza Iow-protein-binding syringe filter (Gelman Sciences), and were used to reinfect E. coli TG1 cells for phage titration and raamplification. Recombinant phages from selected clones were used to infect E. coli HB2151 for production of soluble scFv antibodies. Briefly, the infected E. coli HB2151 cells were cultured in SB medium (35 g/l tryptone, 20 g/l yeast extract, 5 g/l NaCI) supplemented with 100 pglml ampicillin at 30°C with 250 rpm shaking until they reached an A600 of 0.5. lsopropyl-B-D-thiogaIactopyranoside (IPTG) was then added to 1 mM final concentration and the cells were incubated on a shaker at 30°C overnight. Supematants containing the extracellular soluble scFv proteins were separated from the cell pellets by centrifugation at 1500 x g for 15 min, and filtered through a 0.45 pm-pore-size filter. The sensitivities and specificities of the scFv antibodies (phage-associated or soluble forms) were datenninad with ELISA as described previously (Yuan at al., 1997). Briefly, microtiter plates were coated and blocked as described above. Next, 50 pl of PB, standard (at various concentrations) dissolved in PBS was added to the coated wells and simultaneously incubated with 82 50 pl of phage-associated (about 5 x 1010 pfulml in 2x YT medium) or soluble scFv antibody (various dilutions with 1% OVA-PBS or 1% BSA-PBS or PBS-2% non-fat dry milk). A monoclonal antibody (diluted 1:20.000) was included for comparison. The plates were incubated for 1 hr at 37 °C then washed 6 times with PBS-T. The amount of bound phage-associated scFv antibody was datenninad by the addition of 100 pl sheep anti-M13—HRP conjugate diluted 122500. The amount of soluble scFv antibody bound was determined by incubation with 100 pl mouse anti-E tag (1 pglml) at 37°C for 1 hr followed by washing (six times) and a subsequent incubation with goat anti-mouse lgG HRP conjugate (diluted 122000). The bound monoclonal antibody was also detected by the addition of 100 pl goat anti-mouse lgG HRP conjugate. The plates were incubated for 1 hr at 37°C and then washed 6 times with PBS-T. The amount of anti-M13—HRP and goat anti-mouse IgG HRP conjugates bound in the walls was assessed by the addition of 100 pl of 3,3',5,5'- tetramathyI-benzidina (T MB) at 37°C for 15 min. The absorbance was read at 450 nm with a microtiter plate reader (Molecular Devices Corporation, Menlo Park, CA) after the color development was stopped with 100 pllwell of 10% H2804. 83 RESULTS AND DISCUSSION Mouse immunization and polyclonal antibodies Eight mice were immunized and boosted with FB,-KLH immunogen and another eight mice were injected intraperitoneally with FB,-CT conjugate. Each mouse was bled one week after every boost and its samm was prepared and titrated. Titers of the mouse antisera after the second boost were detected in an indirect ELISA format (Figure 3.2A). The specificity and sensitivity of the antisera were determined with a competitive indirect ELISA (Figure 3.2B). Concentration of FB, required for 50% binding inhibition with antisera from FB,-CT immunized mice (approximately 0.27 pglml) were much lower than that from FB,-KLH immunized mice (>10 pglml) (Figure 3.2B). Like FB,-BSA conjugate (Azcona-Olivera at al., 1992a), the conjugate of FB,-KLH was generally ineffective in the production of PB, antibodies in mice. The apparent high antibody titers in mice immunized with FB,- KLH might be due to the presence of antibodies that recognize the toxin-carrier bridge or a conjugate by-product, but which are not inhibited by free F3, in CI- ELISA. In contrast to immunization with FB,-KLH, immunization with low FB,-CT doses resulted in the rapid production of PB, specific antibodies with high relative affinity in all treated animals. Compared with the FB,-KLH immunogen, the effectiveness of FB,-CT conjugate in the production of FB, antibodies might be explained on the basis of the CT adjuvant effect (Azcona-Olivera at al., 1992a). A. B. -o—FB1-KLH +FB1-CT E E til 13 a a 5 E i i O 4 I I 1T g g 400 800 1600 3200 6400 0 10 100 1000 10000 Antisera dilution factor (x) Competitive FB1 . nglml Figure 3.2. Polyclonal anti-FB, antibody production from mice immunized with FB,-cholera toxin (FB,-CT) and FB,-keyhole limpet hemocyanin (FB,- KLH) conjugates (Each group had one representative out of eight mice). Sara were obtained one week after the second boost with the immunogens. A: Titration of antisera measured in FB,-ovalbumin (FB,-OVA) conjugate coated Immulon-4 microtiter wells. Bound mouse antibodies were detected by horseradish peroxidase conjugated goat anti—mouse IgG. B: Competitive indirect ELISA for FB, with the antisera (diluted 1:60.000 and 1:3,200 respectively for FB,-KLH and FB,-CT immunized mice). 85 Construction of phage-display scFv library Mice immunized with FB,-CT conjugate were sacrificed one week after the second boost and their spleens were homogenized in RNA STAT-60 (T EL-TEST Inc.) After mRNA was purified by affinity chromatography on oligo (dT)-cellulose column, first-strand cDNA was synthesized with Moloney murine leukemia virus reverse transcriptase and random hexadaoxyribonucleotida [pd(N),,] primers (Phannacia Biotech). PCR amplification from the mRNA yielded a VH DNA fragment with a size close to the expected 340 bp (lane 3 in Figure 3.3) and a VK DNA fragment with a size close to the expected 324 bp (lane 2 in Figure 3.3). A linker DNA (lane 1 in Figure 3.3) was also produced from a template containing sequence encoding short peptide (Gly,Ser)4 by PCR amplification with LINKBACK and du- LINKFOR as primers. The DNA fragments of VK and linker were successfully joined by PCR assembly (lane 4 in Figure 3.3) and then joined with V,, DNA forming the scFv DNA fragments (lane 5 in Figure 3.3). After addition with an Sfil site on the 5' and and a Notl site on the 3' end, the assembled scFv DNA products were digested with Sfil and Notl restriction enzymes, gel purified and ligated into the phagemid pCANTAB5E. The ligated products were introduced into E.coIiTG1 competent cells by chemical transformation. Approximately 10,000 transforrnants were produced for the construction of scFv phage-display library. Restriction analysis of the recombinant phagemid DNA isolated from the transformants showed an expected size of scFv insert DNA in pCANTAB5E. 86 Figure 3.3. PCR amplification of VH and VK genes from mouse splenic mRNA and the assembly of scFv. Lanes are: (1 ): Linker PCR product; (2): VK PCR product; (3): VH PCR product; (4): Linker-VK PCR product; (5):V,,-Linker-VK (scFv) PCR product; Lane M2100-bp ladder DNA marker (Gibco BRL). 87 Affinity selection of phage-display scFv antibodies In a preliminary experiment, the newly constructed recombinant scFv phage library was incubated in FB,-OVA (1 pg/well) coated wells for FB, specific binding and in OVA coated wells as a negative control of nonspecific binding. After incubation for 60 min with the scFv phage library and extensive washing with PBS- T, FB, (100 pglml) or PBS were used to elute the bound phages from the wells. After five rounds of panning-alution selection, the number of phages eluted by FB, from FB,-OVA coated wells did not differ significantly (less than one order difference) from that eluted by PBS. On the other hand. FB, eluted approximately the same number of phages from both FB,-OVA coated wells and OVA coated walls. This indicated that FB,-specific scFv phages were not enriched in this panning-alution selection. To search for a suitable panning-alution selection procedure for specific binding and elution. the effects of different FB,-carrier protein conjugates (i.e. FB,- OVA. FB,-BSA. and FB,-KLH. Figure 3.4 A), scFv phage library incubation time (Le. 60. 20, and 10 min, Figure 3.4 B) and FB, concentration (i.e.10, 50, and 100 pglml, Figure 3.40) in the elution solution were investigated. No significant enrichment for FB,-specific scFv phage (phage eluted by FB, from FB,-carrier proteins coated walls) was found in any condition tested (Figure 3.4). Although the number of phages eluted by FB, (50, 100 pglml in the elution solution in most cases) from FB,- OVA or FB,-BSA coated walls was greater than that eluted by PBS. the difference appeared not due to the presence of FB, specific scFv phages because the number 88 lure \W .m \\\\\\\\\\\\\\m a s\\\\\\\\\\\\\\. m \\\\\\\\\\\\\\\\\\\\\\\s /////////////// 3.. m r///////// mm 7//////////////////////// b I F o m m m m s\\\\\\\\\\\\\\\\\\\s w m s\\\\\\\\\\\\\\\\\\\\\\s m \\\\\\\\\\\\\\\\\\\\\\\\ a ////////////////// 3.. a /////////////////// mm %////////////// m .. ..... .. .m m Wmm s\\\\\\\\\\\\\\\\\\\\\\\s u m. \\\\\\\\\\\\\\\\\\\\\\\ m \\\\\\\\\\\\\\\\\\\\\\ .1w .1 ///////////////////////////, m. m x////////////////////// m w ///////////////////// m mm s .m 2s a gas qfi «4 _ _ _ a. _ _ _ wmwwww wwwwwww wwwwm eEBEv conga oo§m . e523 oust 355 9528 ounce 35$. 10 F8, used for alution. pglml phage incubation: 60 min; bound so phage alution: by PBS or 100 pglml of FB, in PBS. Modifications 89 100 Standard procedure was: coating: 1 pg/well of FB,-OVA conjugates were different conjugates for coating (A), phage incubation time (B). and FB, concentration for alution (C). Figure 3.4. Phage titers after panning-alution selection of the phage library. or OVA alone (in control wells); of phages ( dmhutoth phage hbrz ooncentrati specific ell nonspecdm 34) Afle identificati I0 FB, was Wmepa phage pl; round of g washed, Soluble s ELKSA. Al Phage lit: sDecific SeqUenr absence of phages eluted by FB, from the FB,-carrier protein conjugates coated walls was similar to that eluted by FB, from wells coated only carrier proteins. Reducing the phage library incubation time in the coated walls, or decreasing the FB, concentration in the elution solution did not improve the differentiation between specific alution (phages eluted by FB, from FB,-conjugate coated wells) and nonspecific alution (phages eluted by F B, from carrier proteins coated wells) (Figure 3.4). After failing to enrich F B, specific scFv phages by panning-alution selection. identification of individual clones with soluble scFv proteins that specifically bound to FB, was also investigated. Two hundred seventy randomly selected individual phage plaques from the unselected original scFv phage library and 39 individual phage plaques selected after four rounds of panning-alution selection (In each round of selection. phages were incubated in FB,-OVA coated walls for 60 min, washed, and eluted with 100 pglml FB,) were used to infect E. coli HBZ151 for soluble scFv protein production. None of the isolates bound FB, specifically in ELISA. Although the mice whose splenic mRNAs were used to construct the scFv phage library in this study produced relatively high affinity antibodies to FB, , no FB, specific scFv antibody clone has been selected from this scFv phage library. Sequence changes during the scFv construction procedure and conformation differences between scFv protein and the native antibodies may cause changes in specificity and sensitivity for the scFv proteins. These changes may result in the absence of FB, specific clones in the original scFv phage library. Even if there were 90 a few clones specifically binding to FB, in the library. the high degree of nonspecific binding of the phage library to the carrier proteins alone plus nonspecific alution activity by FB, in the panning-alution selection make the enrichment for FB, specific phages impossible. Considering that phage panning selection also failed to generate any scFv antibody clone with high affinity to FB, from an scFv phage library constructed from a hybridoma cell line producing FB, specific monoclonal antibody (Zhou at al., 1996). an alternative way to clone a functional FB, scFv antibody is needed. For example, the VH and V,_ can be cloned using PCR-based RACE (Rapid Amplification of cDNA Ends) technology with primers which are independent of the variability within the V regions (Ruberti at al., 1994) and with high fidelity enzymes such as Pfu DNA polymerase. They are then assembled into an scFv fragment and tested for their activities. The specificity and sensitivity of the cloned scFv antibody could be improved further by mutagenesis or chain shuffling (Hoogenboom at al., 1992) or CDR-grafting (Smith at al., 1995; Jung and Pluckthun. 1997) 91 CONCLUSION Mouse polyclonal antibodies to FB, were readily generated after immunization with FB,-CT conjugate but not produced with FB,-KLH conjugate immunization. The relative affinity (measured by FB, concentration required for 50% binding inhibition) of mouse polyclonal antibodies to FB, was 0.27 pglml. From the splenic cDNA of mice that were immunized with FB,-CT. variable region genes of heavy- and kappa light-chain were successfully amplified by PCR with previously designed primers (Yuan at al., 1997). The VH and VK genes were assembled with a linker DNA to form scFv DNA fragments by PCR and cloned into a phage-display vector. However. panning-alution selection from this scFv phage-display library did not result in any enrichment of FB, specific phages. Different FB,-protein conjugates, phage incubation times, and FB, concentration in the elution solution were tested for their effects on specific binding and alution. No conditions were found suitable for FB, specific scFv phage selection in the panning-alution procedure. The soluble form of scFv protein from approximately 300 randomly selected individual clones did not show FB, specific binding activity either. An altamative method is needed to clone functional FB, specific scFv antibodies. 92 PART IV EXPRESSION OF A FUNCTIONAL ANTI-ZEARALENONE SINGLE-CHAIN FV ANTIBODY IN TRANSGENIC ARABIDOPSIS PLANTS 93 ABSTRACT EXPRESSION OF A FUNCTIONAL ANTI-ZEARALENONE SINGLE-CHAIN FV ANTIBODY IN TRANSGENIC ARABIDOPSIS PLANTS The efficacy of cloning a recombinant mycotoxin antibody in plants was tested using Arabidopsis as a model. An anti-zearalenone scFv DNA fragment was first cloned in the newly constructed phage-display vector (pEY.5) and then recloned in the plant transformation vector, pKYLX71::35$2. After transformation, constructs of anti-zearalenone scFv were introduced into immature Arabidopsis seeds via Agrobacterium tumafacians mediation by vacuum infiltration. Only plants transforrnad with construct containing a PR-1 b signal peptide sequence produced transgenic offspring. The anti-zearalenone scFv plantibody from these transgenic plants bound zearalenone with a high affinity ( leo, 11.2 ng/ml) comparable to bacteria-produced scFv antibody and the parent monoclonal antibody (mAb) from hybridoma cell culture. By electron microscopic immunogold labeling, the presence of anti-zearalenone scFv was mainly detected in the cytoplasm and only occasionally outside the cell. Like bacteria-produced scFv antibody. anti- zearalenona scFv plantibody exhibited greater sensitivity to methanol destabilization than the parent monoclonal antibody did. The sensitivity of anti-zearalenone scFv plantibody to acidic disassociation was similar to bacterial-produced scFv antibody and mAb. Expression of specific plantibodies in crops might be useful to neutralize mycotoxins in animal feeds and to reduce mycotoxin-associated plant diseases. 94 INTRODUCTION Zearalenone [6-(10-hydroxy-6-oxo-trans-1-undecanyl)—l3-rasorcylic acid lactone] is a mycotoxin produced by members of the genus Fusarium after infection of corn and small grains ( Mirocha at al., 1971; 1977; Hart at al.. 1982). When fed to animals. the compound causes hyperestrogenism with symptoms such as enlargement of the uterus and nipples, vulvar swelling, vaginal prolapse. and infertility (Hidy at al.. 1977; Mirocha at al.. 1974). Recently. we developed a single-chain Fv (scFv) antibody with high affinity to zearalenone (Yuan at al., 1997). Our experience and others (Kramer and Hock, 1996; Dr. Chu in UW, personal communication) showed that scFv assembly from VH , linker and VK DNA fragments by PCR is one of the most problematic steps in scFv cloning. The assembly often results in undetectable amplificates or undefined DNA amplified products with various sizes. To facilitate the cloning and chain shuffling, and to increase the efficiency of scFv assembly from V,, and VK cDNA fragments, a new phage-display vector was constructed by replacing a 52-bp Sfi llNot I fragment in pCANTAB5E (Phannacia Biotech, Piscataway, NJ )with a DNA fragment encoding the (GIy,Sar)3 linker peptide plus restriction sites which rarely exist in V gene regions. To confirm the feasibility of the new vector in scFv antibody cloning. an anti-zearalenone scFv DNA fragment was cloned and expressed with this new vector in Escherichia coli. In the last ten years. the expression of specific antibodies or antibody fragments in plant has attracted great interest (reviewed by Hiatt and Ma, 1992; 95 Conrad an and has sl (lavladore Ansaenkc mycotoxir used the ArabiclOp zearalen of a SOIL Conrad and Fiedler, 1994; Ma and Hein. 1995; Whitelam, 1995; 1996; Smith, 1996) and has shown some potential in improving plant resistance against pathogens (T avladoraki at al., 1993) and altering plant metabolic pathways (Owen at al., 1992; Artsaenko at al.. 1995). To explore the possibility of using plantibodies to neutralize mycotoxin through passive immunization of animals by feeding. as a first step, we used the newly cloned anti-zearalenone scFv DNA fragment to transform Arabidopsis plants. In this report, we demonstrated that the expression of anti- zaaralenone scFv gene in transgenic Arabidopsis plants leads to the accumulation of a soluble scFv plantibody with high affinity to the mycotoxin zearalenone. 96 Gen purt note pror Co METHODS AND MATERIALS General All chemicals and solvents were reagent grade or better. Chemicals were purchased from Sigma Chemical Company (St. Luis. MO) unless otherwise noted. All DNA manipulations, if not described, were carried out by standard procedures (Sambrook at al., 1989). Construction of scFv cloning vector A DNA fragment (Pharmacia Biotech, Piscataway, NJ), encoding a short peptide containing the structurally flexible peptide linker (Gly,Ser)3, was amplified by polymerase chain reaction (PCR) with Taq DNA polymerase using a sense primer (5'-TCTATGCGGCCCAGQCGQQQGGCAQTAGTGTCACCGTC-3') containing Sfi l and Spa I restriction sites (undaflined) and an antisense primer (5'-AGCACCT§C§§CC§QCTGAGTGAGQTQGATGTCCGATCC—S') containing Not I and Sec I sites (underlined). After electrophoresis in 1.5% low melting point aQarosa gal (Boehringar Mannheim Biochemicals, Indianapolis, IN), the amplified fr’agment was excised from the gel and purified with QIAEXII gel extraction kit (Q IAGEN. Chatsworth, CA). The purified DNA fragment was then digested with 817 I and Not I and ligated into the same enzyme digested pCANTAB5E (Pharmacla Biotech). creating pEY.5 (Figure 4.1). 97 Ci‘i Flc Sac I (2404) so: (2318?” I (2332' Nil (2416 BspE 1 (2442) A Hind m (2235) m . Stop Codon (2470) ' PM I (2233) Barn 1 (2578) All III (1878) a 1 (3359) pEY.5 "a” ' (3555’ (4565 bp) EcoRI (3689) Natl (3850) Dre m (4126) S“ | (505) BMAI(4129) B. Xho I (319) “5177251?“ (7) 2314 I 60W GGC AQT AQT GTC ACC GTC TCC TCA GGT GGA GGC GGT TCA GGC GGA GGT GGC TCT GGC GGT GGC GGA TCG GAC ATC 2424 l GAG CTC ACT CAG GCG GCC GCA E tag-Amber stop codon -fd gene 3 Figure 4.1 . Map of phage-display vector pEY.5. (A). Restriction map with unique restriction sites marked. (B). The nucleotide sequence of linker (bold sequence) and the VH and VK cloning sites (underlined) in pEY.5. 98 Anti-1 eara VH _, clone pQY attac led p TGA GGA, AT( rAC ‘ GSI GTC for ‘ 'K DI\ GTI :TCC CAI ICTT DN time will thee | 3 Id Ilg Yle ding for the p anl lntr pl) iviou Cl' aract Cl 'nSII bi with p' 'Ymi Anti-zearalenone scFv antibody cloning in pEY.5 VH and VK DNA fragments were ra-amplifiad from an anti-zearalenone scFv clone pQY1.5 phagemid DNA (Yuan at al.. 1997) using the following restriction site attached primers. The primers used for VH DNA re-amplification were: VHFORY (5'- TGAGGAGACGGTGACACTAQTGCCTTGGC-3') and VHBACKY (5'- ATGACTCGCGGCCQAGQQGGCCATGGCCSAGGTSMARCTGCA GSAGTCWGG-3'); S= C or G; M=A or C; R=A or G; W=A or T. The primers used for VK DNA ra-amplification were: VKBACKY (5'-TCGGACATC§A§CTQACTCA GTCTCC-3') and VKFORY (5'-GAGTCA‘I'I'CT§CGQCQGCCCG‘ITI’BAKYT C CARCTTKGTSCC-3'); B=T or C or G; K=T or G; Y=C or T. The re-amplified VH DNA fragments were digested with Sfi l and Spa l and ligated into pEY.5 digested with the same enzymes. The ligated products were then digested with Sac I and Not I and ligated with the same enzyme digested VK re-amplified DNA fragments. yielding pEY.5HL. The pEY.5HL phagemid DNA was introduced into E. coIiTG1 cell forthe production of recombinant phages in the presence of helper phage M13KO7 and into E. coli HB2151 for soluble scFv antibody production as described previously (Yuan et al., 1997). The soluble scFv antibody from E. coli cultures was Characterized with ELISA as described (Yuan at al.. 1997). construction of plant scFv expression plasmids The anti-zearalenone scFv DNA fragment was amplified from a zearalenone biriding positive clone, pEY.5HL3 (reconstructed in pEY.5 from pQY1.5), by DOIymerase chain reaction (PCR) using Pfu DNA polymerase (Stratagane. La Jolie. 99 CA ) GTG CCC amp with yielc frorr cree HITII sigr seq p81 Fin wa cre Dl— m; PI; it; Be ar CA ) with the following primers: Sense primer ( 5'-TAT&_C§_GTATGGCCCAG GTGAAACTGC-3') containing an Sst ll site and antisense primer (5'- CCCTQTAGACCAGACGTTAGTAAATGAA—S') containing an Xba I site. The amplified DNA fragment was isolated from a low melting point agarose gel. purified with QIAEXII, digested with Xba I. and ligated into Hinc lI/Xba l -digested pUC19, yielding pQY2. For expression without a signal peptide. 3 Hind Ill-Xba I fragment from pQY2 was ligated into the Hind Ill-Xba l sites of pKYLX71::35S2 directly, creating pQY4.1-1. For expression with a plant protein signal sequence. a 110-bp Hind Ill/Sst ll fragment containing tobacco pathogenesis-related protein PR-1b signal peptide (sp) sequence from pSlG3. a clone with the PR-1b signal peptide sequence (Gopalan at al., 1996) ligated in the EcoR V site of Stratagene’s pBIuescript SK, was ligated into the Hind Ill-Sst ll sites of pQY2. generating pQY3. F inally, a Hind Ill-Xba I fragment containing the sp-scFv fusion fragment from pQY3 was ligated into the Hind III-Xba I sites of pKYLX71::3582 (Maiti at al., 1993), creating pQY4.4-1. Both pQY4.1-1 and pQY4.4-1 were introduced from E. coli DHSa into Agrobacterium tumefacians strain GV3850 (Zambryski at al., 1983) by triparental mating (Figurski and Heliniski, 1979). P lant transformation Arabidopsis thaliana acotype Columbia (Col. GLABROUS1) plants were transformed using a vacuum infiltration protocol similar to the one described by Bechtold at al. (1993). Mature seeds from these infiltrated plants were germinated and selected on plant nutrient agar plates (Bechtold at al., 1993) supplemented with 100 50 pglml kanamycin. Kanamycin resistant seedlings were transferred into soil and leaves from two-month-old kanamycin resistant plants were tested for scFv antibody binding activity with ELISA. Expression of scFv plantibody and ELISA analysis lmmulon-4 microtiter walls (Dynatech Laboratories, Inc., Chantilly, VA) were coated with 1 pglwall of zearalenone-OVA (ZEN-OVA) conjugate in 100 pl 0.05M carbonate-bicarbonate coating buffer (pH9.4) at 4°C overnight. The wells were washed and blocked as described previously (Yuan at al.. 1997). Two to three leaves from two-month-old transgenic plants were cut and ground in a 1.5 ml microcentrifuge tube on ice with a pestle. After centrifugation at 13,000 rpm in a microcentrifuge for 10 min at 4°C, the sap (supernatant) was collected. Fifty microliters of the leaf sap was diluted 1:1 (vol/vol) with 2% non-fat dry milk in PBS and added to each well, followed by incubation at 37°C for 1 hr. After washing 6 times with 320 pllwell of PBS-0.1% Tween 20, 100 pllwell of mouse anti-E tag antibody (1 pglml) was added. followed by addition of the goat anti-mouse lgG HRP conjugate (diluted 1:2000 in 2% non-fat dry milk in PBS). Finally, 100 pllwell of TMB substrate (Sigma Chemical Company) was added and incubated at 37°C for 1 5 min. The reaction was stopped by adding 100 pllwell of 10% H2804. The absorbances at 450nm were determined with a microtiter plant reader (Molecular Device Corporation, Menlo Park, CA). For competitive ELISA, 50 pllwell of free zearalenone at various concentrations (0. 1. 10, 100, and 1000 ng/ml in PBS containing 1% methanol) was 101 adr (frc bor SCI 09 an pl- inc ba KL WE Pr pr Di added into zearalenone-OVA coated wells. then 50 pllwell of diluted (1 :49) plant sap (from GV3850/pQY4.4-1 transformed plants) was added. After 1 hr incubation. the bound plantibodies were detected in the same way as described above. For comparison. a CI-ELISA with diluted (1 :9) bacterial anti-zearalenone scFv (produced from E. coli HBZ151/pEY.5HL3) was performed simultaneously. Methanol effects on the binding activity were measured for mAb. bacterial scFv antibody and scFv plantibody ( diluted 10 folds in 10 % non-fat milk-PBS) as described previously (Yuan at al., 1997). The effect of the pH environment on these antibodies was also tested. The antibodies were diluted (1 :5 vol/vol) in pH 2.3 and pH 6.3 (adjusted with 0.05 M citric acid and 0.05 M NazHPO, ) buffer solution, and incubated at 4°C, overnight. A portion of the pH 2.3 treated sample was adjusted back to pH 6.3 and incubated at room temperature for one hour. To zearalenone- KLH conjugate (1 pglwall) coated walls, 100 pllwell of pH treated antibody samples was added. Bound antibodies were determined as described above. Protein analysis Proteins in plant leaf sap from independent transgenic plants were separated in a 12% SDS-polyacrylamide gel. Two identical gels were run at the same time. After electrophoresis, one gel was stained in Coomassie brilliant blue G-250. The proteins in another gel were transferred to a nitrocellulose membrane (Schleicher & Schuell, Keene, NH). Mouse anti-E tag antibody, goat anti-mouse IgG alkaline phosphatase conjugate (Sigma Chemical Company, St. Louis. MO) and NBT/BCIP 102 substrate similar to ScFv an S transgen glutarald tempera 1% osiu followed embedcl Ultrathir substrate (Pierce, Rockford. IL) were used to detect plantibodies in a procedure similar to that previously described (Yuan at al., 1997). ScFv antibody cytolocation with immunogold labeling Segments ( approximate 0.5 x 1.0 mm) of leaves from 2-month-old transgenic Arabidopsis plants were fixed by vacuum infiltration in 2% glutaraldelhyde in 0.1 M sodium cacodylate buffer (pH 7.2) for 2 hr at room temperature. After 3 washes in the above buffer, the samples were post-fixed with 1% osium tatroxide in 0.1 M sodium cacodylate buffer at room temperature for 2 hr, followed by dehydration with a graded acetone series. The samples were then embedded by infiltration with Resin “Spurtol” (Spurr, 1969; Kushida, 1974). Ultrathin sections were cut and supported on formvar-coated grids. The grids containing ultrathin sections were pretreated by incubation in a saturated aqueous solution of sodium metaperiodata as described by Bendayan and Zollinger (1983) After washed throughly in distilled water, the pretreated grids were blocked for 1 hr at room temperature in 20 mM Tris-buffered saline (T BS, pH 7.5). containing 0.05 % Tween 20 . 1 % wt/vol bovine serum albumin and 0.01 % NaN3. The samples on the grids were then incubated with the primary antibody (mouse anti-E tag antibody) diluted in the blocking buffer for 2 hr at room temperature. In the case of testing for the cross-reactivity of anti-zearalenone scFv antibody to Arabidopsis. samples from non-transformed control plants were incubated with anti-zearalenone scFv antibody first, then with mouse anti-E tag antibody. After four 5 min washings with TBS. the grids were incubated with goat 103 anti tem dist rins al., anc Nel the anti-mouse lgG gold conjugate (10 nm gold particle) (1: 20 dilution) for 2 hr at room temperature. After four 5 min washings with TBS and two 5 min washings in distilled water, the grids were post-stained with 1 % uranyl acetate for 30 min, rinsed in distilled water, and then stained with Hanaichi’s lead solution (Hanaichi at al., 1986) for 5 min. The grids were then rinsed in 20 mM NaOH and distilled water and examined with a CM10 Transmission Electron Microscope (Philips, The Netherlands). Immunogold labeling of sections of non-transformed Arabidopsis thaliana ecotypa Columbia was performed as a control. 104 (Noni expn anufl nevi zeat 5096 INDY then chai not sep scF rnu scF RESULTS Cloning of anti-zearalenone scFv In pEY.5 To determine the feasibility of cloning vector pEY.5 for scFv antibody expression. the DNA fragments of VH and VK from pQY1.5 (Yuan at al., 1997) were amplified with the above primers and ra-cloned into pEY.5. In E. coli HB2151 , one new clone (pEY.5HL3) produced soluble scFv antibody with high affinity to zearalenone (Figure 4. 2). The IC50. concentration of free zearalenone required for 50% inhibition of binding, was 15.5 nglml, similar to that previously described for pQY1.5. Either V,, or VK alone cloned in pEY.5 is in frame with E-tag and 93p, therefore this vector is also suitable for selecting single domain Fv antibodies and chain shuffling. However, either V,, or VK alone from pQY1.5 (Yuan at al.. 1997) did not show any zearalenone binding activity when they were cloned into pEY.5 separately (data not shown). indicating that both VH and VK were important for the scFv binding to zearalenone. It was notable that E.coli HB2151/pEY.5HL3 grew much slower than HB2151/pEY.5, which suggests the functional anti-zearalenone scFv expression may affect the growth of the E. coli host cells. 105 100 90 80 70 60 50 4O 3O 20 10 -o—EY.5HL3 +Plantibody Inhibition of binding, % I I O 1 10 100 1000 Zearalenone, ng/mL Figure 4.2. Sensitivity and specificity of anti-zearalenone scFv antibodies measured by competitive indirect ELISA using zearalenone- ovalbumin coated wells. Bound soluble scFv antibody was detected by mouse anti-E tag antibody, followed by horseradish peroxidase conjugated goat anti-mouse lgG. EY.5HL3 was produced in E. coli and plantibody was produced in Arabidopsis. 106 Expression of anti-zearalenone scFv plantibody The anti-zearalenone scFv DNA fragment from pEY.5HL3 was re-cloned into a plant transformation vector to generate pQY4.1-1 and pQY4.4-1(Figure 4.3). About 1% of 20,000 seeds from plants transformed with GV3850/pQY4.4-1 were kanamycin resistant. whereas plants transformed with GV3850/pQY4.1-1 produced no kanamycin resistant seedlings from approximately 20,000 seeds (Figure 4.4). In the putative transgenic plants (kanamycin resistant) transformed with GV3850/pQY4.4-1. approximately 20% of the tested plants produced functional antibody for zearalenone as determined by ELISA. Western analysis showed that GV3850/pQY4.4-1 transformed Arabidopsis plants produced a 32-KDA scFv plantibody protein (Figure 4.5), similar in size to the scFv antibody produced in E. coli (Yuan et al., 1997). The specificity and sensitivity to zearalenone of the plantibody was determined by CI-ELISA (Figure 4.2). The plantibody had an IC50 of 11.2 ng/mI for zearalenone, showing similar affinity to zearalenone as the scFv antibody produced from bacteria and monoclonal antibody from hybridoma cell culture (Yuan at al., 1997). From each of the T0 transgenic Arabidopsis plants, 15 to 20 T, offspring seedlings were tested with ELISA for zearalenone binding. Some plants had an approximately 1 :1 segregation ratio of functional anti-zearalenone scFv plantibody producing offspring to non-producing offspring; others had a 1:0 segregation ratio. This suggests that both single and double integrations of plant chromosomes by the anti-zearalenone scFv DNA fragment occurred. In the T2 generation, all plants 107 Figure 4.3. Hind 35Sz P... A A A A A A A A A A A A A A 9 9 Anti-zearalenone scFv E-tag III Sst II Xba I ((666 :D ........................... m l9i9999995‘ ‘A‘J‘JA4’ ‘6‘ 999999 P... :1) sp Tu Tu: 32553252223552isisisizisibfl H I 99999 9 9 9 9 ¢ 6 C 6 ¢ 6 ¢ G 9 9 9 9 9 9 9 9 9 AAAAAAAA 1c: Anti-zearalenone scFv E-tag The transgene in plant transformation constructs pQY4.1-1 (A) and pQY4.4-1 (B). 3582: Can 35S promoter with a duplicated enhancer; sp: plant pathogenicity-related protein PR-1 b signal peptide sequence; Tim and Tm: transcription termination sequences for rch and nos respectively; Pm: nos promoter; Kn': kanamycin resistance gene. 108 Figure 4.4. The germination of Arabidopsis tansformant seeds on kanamycin selection medium plates. A. Seeds from un-transformed control plants; B. Seeds from plants transformed with pQY4.4-1, targeting scFv antibody to the apoplastic space with signal peptide sequence. C. Seeds from plants transformed with pQY4.1-1, targeting scFv antibody to the cytoplasm. 109 Figure 4.5. SDS-polyacrylamide gel electrophoresis and Western blot analysis of anti-zearalenone scFv plantibody expression. Soluble proteins of Arabidopsis from un-transformed plants (lane 1), vector pKYLX71::35$2 transformed plants (lane 2), and pQY4.4-1 transformed plants (lane 3) were subjected to SDS-PAGE and stained with Coomassie brilliant blue G-250 (A). Western blot of duplicate SDS-polyacrylamida gal detected by anti-E tag antibody (B). Lane M: protein molecular size marker with 10KDa-ladder. 110 produced anti-zearalenone scFv plantibody without segregation. This indicates that the anti-zearalenone scFv antibody gene was stably integrated into the genome of Arabidopsis. The effects of methanol and low pH on plantibody As shown previously. anti-zearalenone scFv antibody from bacteria exhibited greater sensitivity to methanol destabilization than the parent monoclonal antibody (Yuan at al., 1997). To test the effects of post-translational modifications on the stability of scFv antibodies, anti-zearalenone scFv plantibody was compared with scFv antibody produced in E. coli and the parental monoclonal antibody for their stabilities in methanol and in acidic environment. The scFv plantibody from pQY4.4- 1 transformed Arabidopsis plants was also shown very sensitive to methanol destabilization (Figure 4.6), whereas the parent monoclonal antibody exhibited tolerance to much higher concentrations of methanol. In the low pH environment (pH 2.3), however, all three antibodies (monoclonal antibody. bacterial scFv antibody and scFv plantibody) lost binding activity to zearalenone (T able 4.1). When the pH was adjusted back to neutral (pH 6.3), the binding activity recovered. 111 E 1.6 - O z; *5 1.2 *- 0 § -D-mAb “E 0.8 ~~ —o—EY.5HL3 § +Plantibody 0.4 ~— 0 I l l I l I 7 Methanol in the well, % Figure 4.6. Effect of methanol on the binding of anti-zearalenone monoclonal antibody (mAb) and bacterial scFv antibody (EY.5HL3) and plant scFv antibody (plantibody) to immobilized zearalenone-KLH conjugate. Table 4.1. Effect of pH environment on antibody binding activity. Anti-zearalenone Relative binding activity in different pH environment, %* antibody pH 6.3 pH 2.3 pH 2.3 than pH 6.3 mAb 100 0 99 Bacterial scFv 100 0 98 scFv plantibody 100 0 89 *: The binding activity at pH 6.3 was set as 100%. 112 Localization of anti-zearalenone scFv plantibody Transgenic Arabidopsis plants expressing anti-zearalenone scFv antibody were processed and labeled for electron microscopic immunolocalization as described in materials and methods. Gold particle deposition indicate the presence of scFv antibody mainly in the cytoplasm with a few labeling signal in the intercellular spaces (Figure 6A,B). Deposition was not found in the non-transformed control plant samples (Figure 4.70). However. incubation of non-transformed plant sections with the anti-zearalenone scFv antibody than with mouse anti-E tag antibody followed by goat anti-mouse lgG gold conjugate caused many labeling signals evenly distributed in the cytoplasm (Figure 4.7C). indicating that anti- zearalenone scFv antibody can bind to Arabidopsis cell components. 113 Figure 4.7. Cytolocation of anti-zearalenone scFv plantibody by immunogold labeling and transmission electron microscopy. Leaf sections from plants expressing anti-zearalenone scFv plantibodies (A, B) and non- transfonned control plants (D) were detected by mouse anti-E tag antibody followed by goat anti-mouse lgG gold conjugate. (C): Leaf sections from non-transformed control plants were pre-incubated with anti-zearalenone scFv antibody. than with mouse anti-E tag antibody followed by goat anti-mouse IgG gold conjugate. CW, cell wall; CY, cytoplasm; ICS, intercellular space; Arrows: gold labeling; Magnification x 39,000. 114 DISCUSSION In this investigation, an scFv plantibody with high affinity to zearalenone was produced from stable transgenic Arabidopsis plants . To our knowledge, this is the first report of an anti-mycotoxin antibody gene expressed in plants. Like other scFv plantibodies (Bruyns at al.. 1996), this anti-zearalenone scFv plantibody had similar antigen-binding properties as bacteria-produced scFv antibody. The anti-zearalenone scFv DNA fragment was first cloned in a newly constructed vector. This new vector, pEY.5, maintains the advantageous features of the original vector pCANTAB5E , such as an inducible lac promoter, a widely used linker (Gly,Ser)3, a highly specific E-tag, an amber stop codon allowing the switch between expression of membrane anchored scFv-93p fusion and soluble scFv simply by changing the expression host. It also allows the direct cloning of VH and VK, with the introduction of two rare cutting restriction enzyme sites (Spa l and Sac | ). In the Kabat Database of Sequences of Proteins of Immunological Interest (http://immuno.bme.nwu.edu). the Spa I recognition sequence was found in less than 1% of 4701 known murine VH chains and 1% of 2492 known murine VK chains. Sac I has a slightly higher cutting frequency, but accounts for only 4.5% and 2.3% of known V,, and V, chains, respectively. This indicates that the majority of variable region cDNAs will be free from intemal cutting. We tested the feasibility of this new vector by cloning and expressing an anti-zearalenone scFv in E. coli. By utilizing the Sfi l/Spa l or Sac l/Not I sites. this new vector will also facilitate chain shuffling. 115 which will be useful for cloning compatible variable chains and improving antigen binding ability (Kang at al., 1991; Marks at al., 1992a). For plant transformation, two constructs of the anti-zearalenone scFv were made by fusion with or without the N-tenninal signal peptide from tobacco pathogenesis related protein PR-1 b, which has been used to secrete foreign proteins to the plant apoplast (Dixon at al., 1991; Lund and Dunsmuir, 1992). The PR-1b signal sequence was included in the expectation that it would target the fusion protein to the endoplasmic reticulum where the signal peptide would be cleaved and the scFv protein transported to the apoplast via the default pathway (Danecke at al., 1990). Plants transfonnad with this signal sequence containing construct produced many transfonnants expressing scFv plantibody with high affinity to zearalenone. However, electron microscopic immunolocalization indicated the presence of the scFv plantibody mainly in the cytoplasm. Only low level of labeling signals existed in the intercellular space and was limited to those parts of intercellular space that were filled with electron-dense material. The labeling signals in the cytoplasm might reflect a binding activity of anti-zearalenone scFv to some plant cell components. As a matter of fact, immunogold labeling showed that antl- zearalanone scFv antibody binds to non-transformed Arabidopsis cell components in the cytoplasm. Although some antibodies or antibody fragments fused with plant signal sequences have been confirmed by immunogold electron microscopy to be secreted through the cell wall into intercellular spaces. despite limitations in cell wall porosity (De Wilde at al., 1996), others were retained in endoplasmic reticulum compartment or in the cytoplasm (Dllring at al.. 1990; Artsaenko at al., 1995; 116 Schouten at al., 1997). The construct without signal sequence failed to generate any transgenic plants in this study. One explanation might be that high accumulation of anti-zearalenone scFv in the plant cytoplasm and the sequential binding to plant cell components was toxic to the host plant cell. As previously reported. anti-zearalenone scFv antibody produced from bacteria had greater sensitivity to methanol destabilization than its parent mAb. One of the possibilities may be the differences between prokaryota and eukaryote in protein post-translational modifications, especially glycosylation. N—linked glycosylation of proteins in plants is very similar to the glycosylation process in mammals (Jones and Robison. 1989; Sturm at al., 1987). Possible plant post- translational changes in addition to glycosylation include fatty acylation. phosphorylation. sulfation. or modifications that change the pl of the protein (Jonas and Robinson, 1989). The comparison between antibody produced in bacteria and plants may help elucidate the role of post-translational modifications in stabilizing antibody. Unfortunately. it was not possible to test the hypothesis that differences between anti-zearalenone scFv antibody and its parent mAB in methanol sensitivity reflect structural differences derived from post-translational modifications because the anti-zearalenone scFv lacks the canonical Asn-X-Ser/Thr signal sequence for glycosylation. Some mycotoxins not only cause toxic effects to animal and humans, but also serve as virulence factors for the producing fungi in causing plant diseases. The mycotoxin zearalenone has been reported to cause certain phytotoxicities (Wakulinski, 1989; Vianello and Macri, 1981; Nedalnik. 1993). The expression of 117 anti-zearalenone antibody in plants might be useful to reduce the severity of plant diseases associated with this mycotoxin. Antibodies have been used successfully as antidotes for some low molecular weight toxins through passive immunization (reviewed by Baud at al., 1997). However, large quantities of antibody are generally required for passive immunization in vivo. The prospect of harvesting antibody on an agricultural scale offers the potential for economic production of almost limitless amounts of antibody. The production of antibody in edible plants makes it possible to deliver large amount of toxin specific antibody through food and feed. Feed-delivered antibody could theoretically bind zearalenone in the intestine providing it can survive passage through the acidic stomach environment. When the anti-zearalenone antibody was subjected to low pH. the mAb and scFv antibody from both bacteria and plants lost their binding ability. However, all three antibodies recovered their binding activity when the pH was adjusted back to neutral (pH 6.3). This suggests that the pH affects on the binding activity is not irreversible denaturation. Thus. providing these antibodies survive the proteolysis in the gut, they could recover binding to zearalenone in the small intestine, where the pH is close to neutral. Whether the anti-zearalenone plantibody reported in this study will neutralize zearalenone in animal feed and reduce toxicity will be determined in the future. The expression of mycotoxin specific plantibody may also be useful in reducing mycotoxin-associated plant diseases. Especially, the catalytic plantibodies expressed in crops may degrade mycotoxins when they are produced. 118 PART V IDENTIFICATION OF MIMOTOPE PEPTIDES WHICH BIND TO THE MYCOTOXIN DEOXYNIVALENOL-SPECIFIC MONOCLONAL ANTIBODY 119 ABSTRACT IDENTIFICATION OF MIMOTOPE PEPTIDES WHICH BIND TO THE MYCOTOXIN DEOXYNIVALENOL-SPECIFIC MONOCLONAL ANTIBODY A monoclonal antibody (mAb) 6F5 that recognizes the mycotoxin deoxynivalenol (DON. vomitoxin) was used to select for peptides that mimic the mycotoxin by employing a library of filamentous phage which display random 7-mar peptides on their surface. Two phage clones were selected from the random peptide phage-display library and found to code for the amino acid sequences SWGPFPF and SWGPLPF. These clones were named DONPEP.2 and DONPEP.12, respectively. A competitive enzyme-linked immunosorbent assay (ELISA) strongly suggested that these two phage-displayed peptides bound to mAb 6F5 specifically in the deoxynivalenol binding site. The amino acid sequence of DONPEP.2 plus a structurally flexible linker at the C-terminus. i.e. SWGPFPFGGGSC. was synthesized and tested for its ability to bind to mAb 6F5. This synthetic peptide (C430) and deoxynivalenol competed with each other for mAb 6F5 binding. When translationally fused with bacterial alkaline phosphatase. DONPEP.2 bound specifically to mAb 6F5 while retaining enzyme activity. The potential use of these peptides as immunochemical reagents in deoxynivalenol immunoassay was evaluated with a deoxynivalenol-spiked wheat extract. When peptide C430 was conjugated to bovine serum albumin, it elicited antibodies in mice and rabbits that were specific only to the peptide C430 but not to deoxynivalenol. In an in vitro translation system containing rabbit reticulocyte lysate. the synthetic peptide C430 did not inhibit protein synthesis but did show antagonism toward deoxynivalenol- 120 induced protein synthesis inhibition. These data suggest that the peptides selected in this study bind to both mAb 6F5 and to the ribosoma at the same sites as deoxynivalenol does, but possibly in a slighter different orientation. 121 INTRODUCTION Deoxynivalanol (DON, vomitoxin, Figura1.3) is one of the sesquiterpene mycotoxins classified as 12,13-epoxy-trichothecenas (Mirocha at al.. 1977). It occurs naturally in infected corn (Hart at al.. 1982; Mirocha at al.. 1976), small grains (Hart and Braselton, 1983; Naish and Cohen, 1981), and mixed feeds (Mirocha at al., 1976). DON is mainly produced by the fungus Gibbaralla zaae (Schwein) Patch (anamorph =Fusarium graminaarum Schwabe). At the cellular level, the main toxic effect of DON is inhibition of protein synthesis via binding to the ribosome and by interfering with peptidyltransferase (Ueno, 1983; Thompson and Wannemacher, 1986; Betina, 1989b). In animals. DON can cause anorexia and emesis (vomiting) (Mirocha at al., 1976; Rotter at al.. 1996); Other toxic effects of DON include skin irritation, hemonhaging. hematological changes, human lymphocyte blastogenesis impairment. radiomimetic effects, apoptosis (cytotoxicity) and immunotoxicity (Ueno, 1983; Forsell and Pestka. 1985; Forsell at al.. 1986; Pestka at al.. 1987; Pestka at al., 1994b; Pestka and Bondy, 1994; Ueno at al., 1995; Rotter et al., 1996). A major means for eliminating DON from human and animal food. like for most other mycotoxins. is to detect and divert contaminated raw materials from feed and finished food use. Conventional methods for DON detection (Pathre and Mirocha. 1977; Scott, 1982). including gas chromatography, gas chromatography- mass spectrometry, high-pressure liquid chromatography, and thin-layer ch romatography. involve considerable sample preparation, are time-consuming. and 122 require technical expertise. Compared with other methods, immunoassays have several advantages for rapid field tests, including high specificity, sensitivity, facile sample preparation, and ease of use (Pestka at al., 1995). Following the development of a reliable monoclonal antibody to DON (Casale at al., 1988), immunological methods, mainly enzyme-linked immunosorbent assay (ELISA). have been used widely for detection of DON (Pestka at al.. 1995). Such developments have led to a great demand for specific antibodies and related immunochemical reagents for the assay. However. chemical conjugation of DON to a carrier protein or an enzyme has low efficiency because it involves extensive modification and blocking stages and causes substantial bridge-group interferences and un-wanted cross-reactions (Casale et al., 1988; Wilkinson at al., 1992; Xiao at al., 1995; Pestka at al.. 1995). Secondly, when DON is conjugated to a carrier protein. it is weakly immunogenic. Finally. since DON is toxic and is included as standard and conjugate in immunoassay. it potentially poses a toxicity risk to the kit user. A possible altamative to the use of mycotoxins as immunochemical reagents would be to develop protein or peptide mimics that serve the same function. One approach for this is through generation of anti-idiotypa antibodies (Chanh at al., 1989; 1990; 1991;1992; Hsu and Chu, 1994; Chu at al.. 1995). which mimic the surface structure of low-molecular-weight biological toxins. Sometimes the sensitivity of original antibody can be improved via generation anti-anti-idiotype antibody (Chanh at al., 1990). However. these techniques are time consuming and costly. A new technique, namely phage display, has been developed more recently. In this procedure, foreign peptides and proteins can be genetically fused to the N- 123 terminus of the minor coat protein 93p of the filamentous phage fd. resulting in display of the peptides and proteins on the surface of the virion (Smith. 1985). By insertion of random oligonucleotide sequences into the 93 gene of filamentous phage fd. phage display provides a means of constructing extensive peptide libraries that may be screened to select peptides with specific affinities or activities (Cwirla et al., 1990; Devlin et al., 1990; Scott and Smith, 1990). Phage-displayed short peptide libraries have been widely used in a number of applications (Cortese at al.. 1995; Lowman, 1997; Katz, 1997). including epitope mapping (Scott and Smith. 1990; Cwirla at al., 1990), mapping of protein-protein contacts (Hong and Boulanger, 1995), identification of protease substrate (Smith at al., 1995). identification of integrin (O’Neil at al., 1992) and other receptors (Doorbar and Winter, 1994), antagonists (Pierce at al., 1996), and ligands (Katz, 1997). Few studies, however, have used this technology to select peptide mimics of non- proteinaceous chemicals with the exception of Iectin (Oldenburg at al., 1992; Scott at al., 1992), biotin (Weber et al., 1992). and carbohydrate (Hoess at al., 1993). Given the feasibility of selecting peptides that mimic non-proteinaceous chemicals, we were interested in determining whether peptide mimics of low- molecular-weight mycotoxins could be identified. Once selected, these peptide mimics might be useful in mycotoxin immunoassay and for development of new antibodies against mycotoxins. Here we describe two closely related heptapeptide mimotopes for DON selected from a phage-display random peptide library and their application to the analysis of DON by ELISA. The mimotope peptide was also 124 compared with DON for cytotoxicity in cell culture and in vitro protein synthesis inhibition. 125 MATERIALS AND METHODS Reagents All inorganic chemicals and organic solvents were reagent grade or better. Ovalbumin (OVA) (crude), bovine serum albumin (BSA)(fraction V), 3,3',5,5'- tetramethyl-benzidine (TMB), phenylmethylsulfonyl fluoride (PMSF). polyoxyethylana sorbitan monolaurata (Tween 20). deoxynivalenol (DON). mouse immunoglobulin G. and goat anti-mouse IgG peroxidase conjugate were purchased from Sigma Chemical Co. (St. Louis, MO). Dimethyl sulfoxide (DMSO) and methanol were purchased from J. T. Baker Inc. (Phillipsburg, NJ). Instant nonfat dry milk ( Commander Foods, Inc., Syracuse. NY) was purchased from a local grocery store. DON-BSA, DON-OVA, and DON-horseradish peroxidase (DON-HRP) conjugates at DON’s 3-hydroxyl group were made though an activated N- hydroxysuccinimide ester as described by Casale at al. (1988) (Figure 5.1) and kindly provided by Frank E. Klein (Neogen Corporation, Lansing, MI). Sheep anti- M13 horseradish peroxidase (HRP) conjugate was purchased from Phannacia Biotech Inc. (Piscataway, NJ). m-MaleimidobenzoyI-N-hydroxysulfosuccinimide ester (Sulfo-MBS) and p-nitrophenyl phosphate disodium salt tablets (pNPP) was purchased from Pierce Chemical Company(Rockford. IL). Anti-DON monoclonal antibody from hybridoma cell line 6F5 was prepared as described previously (Casale at al., 1988). 126 H3C . his) 0 CH3 0 H9C4._\B/ / L---0—C(CHZ)2C —-OH DON-boronate || 0 i PM 0 CH3 0 H9C4_\B/ DON-boronate-HS 0—C(CH2)2C —OH l i f”3l 0” CH3 OH DON-HS l ....o—c(cnz)2cl —Protein Figure 5.1. DON-HS-Protein conjugate Preparation of deoxynivalenol (DON) conjugates via 3-hydroxyl group (Pestka at al., 1995). 127 Phage-display peptide library and amplification A phage-display heptapeptide library with 2 x 10" sequence complexity (random peptide 7-mers fused to the minor coat protein 93p of the filamentous coliphage M13) was purchased from New England Biolabs. Inc. (Beverly, MA). The library was amplified by adding 5 pi (= 1011 pfu) recombinant M13 phage from the stock library into 100 ml of Escherichia coli ER2537 (New England Biolabs. Inc.) (grown in LB medium with 250 rpm shaking at 37°C to 0060,, approximately 0.4)and incubated 6.5 hr at 37°C with shaking at 250 rpm. After the culture was centrifuged at 1,200 x g for 20 min. the supernatant containing amplified recombinant M13 phage was collected and filtered through an Acrodisc Syringe Filter (pore size 0.45pm, low protein binding) (Gelman Science, Ann Arbor, MI). The titer of the recombinant phage was determined by plaque counting after infection of E. coli ER2537 with serial 10-fold dilutions of the phage in LB top agar and plating on LB agar plates. Phage selection by panning-alution selection One hundred microliters of mAb 6F5 (15 pglml) in 0.01M phosphate buffered saline (PBS) (pH 7.4) was dispensed into each well of disposable lmmulon-4 microtiter strips (Dynatech Laboratories, Inc., Chantilly, VA). The antibody was dried onto the wells in a forced-air oven at 40°C. overnight. Blank wells were coated with an equal concentration of mouse immunoglobulin G. The wells were washed 4 times by filling each well with 300 pl PBS and aspirating the contents. Nonspecific binding was blocked by incubating 320 pl of 10% non-fat dry milk dissolved in PBS 128 (10% milk-PBS) in each well for 1 hr at 37°C and then washing 4 times with PBS. For the panning-alution selection (Figure 5.2), the recombinant phage-display peptide library (diluted with 10% milk-PBS to approximately 1010 pfulml) was then added into the walls (100 pllwell) and incubated at 37°C for 1 hr. The wells were washed 20 times by filling each well with approximately 300 pl PBS containing 0.1% (vol/vol) Tween 20 (PBS-T) followed by incubation with 300 pllwell of PBS at 37°C with shaking at 150 rpm for 1 hr. The wells were washed again 10 times with PBS-T (300 pllwell). To elute the bound phages in the microtiter walls. 100 pl of DON (100 pglml in PBS containing 1% methanol) or PBS containing 1% methanol was added to each well and incubated at 37°C with shaking at 150 rpm for 1 hr. The eluted phages were then collected from the microtiter wells and used to reinfect E. coli ER2537 cells for phage titering and amplification. After four rounds of panning- elution selection, individual plaques were picked from LB plates and used to infect 2.5 ml of E. coli ER2537 culture described above. After 6 hr incubation at 37°C with shaking at 250 rpm, the culture was centrifuged at 10.000 x g for 10 min and the supernatant containing recombinant phages was collected. The binding activity to mAb 6F5 was tested for each individual phage clone by ELISA. The bound phages in mAb 6F5 coated microtiter walls was determined by incubation with 100 pllwell of sheep anti-M13 HRP conjugate (5000 times diluted in 10% non-fat dry milk-PBS) at 37°C for 1 hr followed by incubation with 100 pllwell of TMB substrate at 37°C for 15 min. The absorbance was read at 450nm after stopping the reaction with 100 pllwell of 10% H260... 129 Re-Infectlon and re-ampllficatlon Figure 5.2. Panning-alution selection procedure for deoxynivalenol (DON) mimotopes in anti-DON monoclonal antibody coated lmmulon-4 microtiter walls. 130 Titration and amplification of panning-elutlon selected phages A 10-fold dilution series of the selected phages was made in LB medium and 100 pl of the diluted phages was mixed with 100 pl of E. coli ER2537 (grown in LB with shaking (250 rpm) at 37°C to OD$00 approximately 0.2) and incubated at room temperature for 30 min for infection. The infected E. coli ER2537 calls were mixed with 4 ml LB top agar and spread onto LB agar plates. After solidification, the plates were incubated at 37°C overnight and plaques were counted. To amplify the phages, approximately 2 ml of phage solution eluted from monoclonal antibody 6F5 coated walls was added into 50 ml E. coli ER 2537 (grown in LB with shaking at 250 rpm at 37°C to 0060,, 0.2) in a 250 ml flask and incubated for 6 hr at 37°C with shaking (250 rpm). The supernatant from this culture was collected after centrifugation at 10,000 x g for 10 min. In order to avoid the interference of a high concentration of free DON in the phage solution. 100 pl of the supernatant containing amplified phages was added into 50 ml E. coli ER 2537 for re- amplification as described above. The re-amplified phages, containing approximately 8 nglml of DON, was collected after centrifugation and used for a subsequent round of panning-alution selection. Competitive ELISA with phage-displayed peptlde For competitive direct ELISA (CD-ELISA) with phage-displayed peptide, microtiter walls were coated with mAB 6F5 and blocked as described for panning- alution selection. Various concentrations (0-10.000 nglml in 1% methanol-PBS) of DON were mixed with an equal volume of phage-displaying peptide (diluted 10 131 times in 10% non-fat dry milk-PBS). The mixtures were added into mAB 6F 5 coated microtiter wells at 100 pllwell and incubated at 37°C for 1 hr. After washing 6 times with PBS-T (300 pllwell), the bound recombinant phages were determined by incubation with 100 pllwell of sheep anti-M13 HRP conjugate (diluted 1:5000 in 10% non-fat dry milk-PBS) at 37°C for 1 hr. Bound enzyme was determined as described above. For comparison, 50 pllwell of DON-HRP was also mixed with 50 pllwell of DON at various concentrations (0-10.000 nglml in 1% methanol-PBS) and incubated in mAB 6F5 coated microtiter wells at 37°C for 1 hr. Bound enzyme was determined as described above. For competitive indirect ELISA (Cl-ELISA) with phage-displayed peptide, 100 pllwell of phage-displayed peptide was dispensed into each well of disposable lmmulon-4 microtiter strips and dried onto the wells in a forced-air oven at 40°C overnight. Plates were washed and blocked as in the CD-ELISA. Various concentrations (0-10,000 nglml in 1% methanol-PBS) of DON (50 pllwell) were added into the wells followed by 50 pllwell of anti-DON mAB 6F5 (10 pglml in 10% non-fat dry milk-PBS). Wells were incubated at 37°C for 1 hr. After washing 6 times with PBS-T, the bound anti-DON mAB 6F5 was determined by incubation with goat anti-mouse IgG HRP conjugate (diluted 2000 times with 10% non-fat dry milk-PBS) at 37°C for 1 hr. Bound enzyme was determined as described above. DNA sequencing After confirmation of the specific binding to mAB 6F5 by ELISA, 10 ml of recombinant phage (1011 pfulml in LB) from each positive phage clone was used for 132 single stranded DNA (ssDNA) isolation with QlAprep Spin M13 Kit (QIAGEN Inc., Chatsworth, CA). The ssDNA was sequenced with -28 glll sequencing primer and -96 glll sequencing primer (New England Biolabs. Inc.) by Taq cycle sequencing and dye terminator chemistry at the Michigan State University Sequencing Facility. Translational fusion with bacterial alkaline phosphatase (AP) A 179 bp DNA fragment encoding the 93p signal peptide, DON mimotope peptide and first 17 amino acids of M13 phage g3p protein was amplified by polymerase chain reaction (PCR) with Pfu DNA polymerase using a sense primer (5'-GCCAAGCTTAGATC'I'I'GGAGCCT'ITFITTI'GGAG-3') and an anti-sense primer (5'-CCGGTCGACCTGTATGGGA‘I'I'T'I’GCTAAACAACT—3'). After gel purification, the amplified DNA was digested with 89! II and Sal l and cloned into BamH I-SaII digested leP5 (Carrier at al., 1995) to generate plasmid pOY7. The ligated product was used to transform E. coliDH11S competent cells (Gibco BRL. Gaitharsburg. MD), generating DH11S/pQY7. For DONPEP-AP fusion protein production, DH1IS/pQY7 was grown in SB supplemented with ampicillin (100 pglml) and 2 % glucose at 30°C with shaking (250 rpm) to absorbance (600nm) (A600) approximately 0.5. After the culture was centrifuged at 1000 x g for 10 min and the medium was discarded. the cells than were resuspended in SB containing ampicillin (100 pglml) and 1 mM isopropyI-B-D-thiogalctopyranoside (IPTG) and incubated with shaking (250 rpm) at 30°C overnight. The supernatant containing the extracellular soluble DONPEP-AP fusion protein was separated from the cell pellets by centrifugation at 7.000 x g for 15 min and filtered through a 0.45 pm-pore-size 133 filter. The periplasmic DON PEP-AP fusion protein was extracted by suspending the bacterial cells (1 :5 vol/vol) in lysis buffer (50 mM Tris-HCI, pH 8.0, 20% sucrose. 10 mM EDTA . 0.1 mg/ml of lysozyma. 0.5 mM PMSF), and left on ice for 1 hr with agitation before centrifugation for 30 min at 7,000 x g. The supernatant was filtered through a 0.4 pm porous filter. The activity of DONPEP-AP fusion protein was determined by CD-ELISA as described below. CD-ELISA with DONPEP-AP fusion protein Microtiter wells were coated with mAB 6F5 and washed as described above. Various concentrations (0-10,000 nglml in 0.05M Tris buffered saline. pH7.4) of DON were mixed with an equal volume of DONPEP-AP fusion protein (periplasmic extract diluted 1:7 with 2% non-fat dry milk-TBS). The mixtures (100 pllwell) were added into each mAB 6F5 coated microtiter well and incubated at 37°C for 1 hr. After washing 6 times with 300 pl TBS containing 0.1% Tween 20 (TBS-T). the bound DONPEP-AP was datenninad by incubation with pNPP substrate solution at 37°C for 45 min. The absorbance was read at 405 nm. CD-ELISA using peptide C430 and its HRP conjugate One DON mimotope peptide plus a structurally flexible linker and a cysteine residue, NHZ- SWGPFPFGGGSC-COOH, was synthesized via Fmoc chemistry at Bio-Synthesis, Inc. (Lewisville, TX) and was named C430. The synthetic peptide C430, at various concentrations. was used to compete with DON-HRP for binding to mAB 6F5 in CD-ELISA. C430 was also chemically conjugated to HRP with a 134 sulfo-MBS cross linker. For CD-ELISA with C430-HRP conjugate, the procedure was the same as CD-ELISA with DON-HRP described above, except that C430- HRP (diluted 1:5000 in blocking buffer) replaced DON-HRP. Immunization of animals with C430-BSA conjugate The synthetic peptide C430 was conjugated to bovine serum albumin (BSA) with a sulfo-MBS cross linker. The conjugate (approximately 29 : 1 molar ratio of peptide C430 to carrier protein BSA) was used to inject 7-weaks-old BALB/c female mice intraperitoneally or 6-month-old New Zaaland white rabbits subcutaneously. Initial injections for mice were 100-200 pg of C430-BSA conjugate in 200 pl of saline-Fraund’s complete adjuvant (1 :1 ). followed at 3-weak intervals by boosts of 100-200 pg conjugate in 200 pl saline-Freund’s incomplete adjuvant(1 :1). The initial injection for rabbits was 1 mg of C430-BSA conjugate in 1 ml of saline-Fraund’s complete adjuvant (1 :1 ). followed at 4-week intervals by boosts of 250 pg conjugate in 1 ml saline-Fraund’s incomplete adjuvant (1 :1). Animals were bled 1 week after each boost. Antisera were screened for specific binding in phage-displayed DONPEP.2 coated walls by CI-ELISA using C430 or DON as competitive antigens. Cytotoxicity and effects on protein synthesis in vitro B6C3F1 mice (10-weeks-old) were euthanized by cervical dislocation and femurs were removed. Bone marrow cells were flushed from the femurs using a 1 cc syringe and 25 gauge needle. Erythrocytes were Iysed with 0.83% ammonium chloride. Call number and viability were determined by trypan blue dye exclusion 135 using a hemacytometer. The cells were cultured at 1 X 10" cells/ml in 96 well flat bottom plates in RPMl-1640 in a 5% CO2 humidified incubator. RPMl-1640 was supplemented with 100 U/ml penicillin, 100 pglml streptomycin, 50 pM 2- mercaptoethanol, 1 mM sodium pyruvate, 1 mM non-essential amino acids, 2 mM glutamine and 10% fetal bovine serum. DON and synthetic peptide C430 were diluted in RPMI-1640. Duplicate cultures were treated. After 18 hr of exposure to DON and synthetic peptide C430, cell viability was determined using an MTT conversion assay as described by Marin et al. (1996). To determine the effects on protein synthesis, DON or synthetic peptide C430 (0 or 3.4 pM in the final 50 pl reaction) was added to 30 pl biotin translation mix (Boehringer Mannheim Corporation, Indianapolis, IN) containing reticulocyte lysate, 10 pmol biotin-lysine-tRNA'”, 42 pM amino acids without lysine, sperrnidine, energy mix, dithiothreitol, 83 mM K-acetate, and 0.83 mM Mg-acetate. The mixtures (46 pl) were incubated on ice for 10 min, followed by the addition of 4 pl of y-globulin mRNA (0.5 pg/pl). The final reaction (50 pl) was incubated at 30°C for 1 hr. The translated protein samples were stored at -80°C. For Western blot analysis, 3 pl of protein translated in vitro were boiled for 10 min after mixing with SDS-PAGE loading buffer. The samples then were loaded into a mini SDS-10% polyacrylamide gel and subjected to electrophoresis at 80 V for 2 hr. The newly synthesized proteins were detected by electrotransfer to a PVDF transfer membrane (DuPont NEN Research Products, Boston, MA), followed by incubation with streptavidin-peroxidase conjugate (Boehringer Mannheim Corporation). Bound enzyme was visualized by incubation with SuperSignal" 136 ULTRA chemiluminescent substrate (Pierce Chemical Company) and exposing a Kodak XAR5 autoradiography film (Kodak, Rochester, NY). 137 RESULTS Panning-alution selection of specific phages After each round of panning-elution selection, the numbers of eluted phages were monitored (Figure 5.3). Enrichment for the specific phages with affinity to mAb 6F5 was observed after two rounds of panning-elution selection. The phages with affinity to mAb 6F5 were enriched approximately six orders of magnitude after the fourth round of panning-elution. Fifteen individual phage plaques from the fourth round of panning-elution selected phages were randomly isolated to infect E. coli ER2537 for phage reamplification. Each of the 15 phage isolates was tested for their binding activity to mAb 6F5 by ELISA. Five of these phage isolates, named as DONPEP.1, DONPEP.2, DONPEP.3, DONPEP.4, and DONPEP.12, showed binding activity to mAb 6F5. Single stranded phage DNA was isolated and the nucleotide sequences were determined for each of the five positive phage isolates. Four of five sequenced isolates had identical DNA sequence (5'-AG'|‘|'GGGGTCCTITTCCG'|'TT—3') encoding the consent peptide sequence SWGPFPF, while isolate DONPEP.12 differed by 4 nucleotides (5'-1§TTGGGGTCC§_QTTCCTTTT-3') resulting in a change of the fifth amino acid from F5 to L5. 138 h4011513186 coated [:1 mAb 6F5 coated 101° — 0 06 04 102 — 10° 1.55.3 6820 owns—m 2nd 3rd 4th lst Round of biopanning selection microtiter wells or from mouse IgG coated lmmulon-4 microtiter wells 139 elution with DON (100 pglml) from mAb 6F5 coated lmmulon-4 in each round of panning-elution selection. Figure 5.3. Enrichment of mAb 6F5 specific phage. Phage were titered after Application of DON mimotope peptide in competitive ELISA To demonstrate whether the positive recombinant phage peptides actually mimic the epitope recognized by mAB 6F5 or just bind non-specifically to the surface of the antibody molecule outside the antigen binding site, two different positive phage clones were tested for binding to mAB 6F5 in a CD-ELISA (Figure 5.4). Binding of phage clone DONPEP.2 and DONPEP.12 to immobilized mAB 6F5 was competitively inhibited by free DON. This strongly suggested that these two phage clones were binding to the antigen binding site of the monoclonal antibody, mimicking, in part, the structural epitope of DON. To confirm that the sequence obtained from the phage library binds specifically to mAB 6F 5 independently of phage structural context, synthetic peptide C430 was tested for binding in CD-ELISA. Binding of DON-HRP to immobilized mAB 6F5 was inhibited by free C430 (Figure 5.5A) while the binding of C430-HRP conjugate to immobilized mAB 6F5 was conversely inhibited by free DON (Figure 5.58). This suggested that the peptide was by itself sufficient for binding to the antibody, independent of the phage structural context. When C430 was used to compete DON-HRP or C430-HRP for binding to mAB 6F5, the leo's were 0.64 to 0.8 pM, whereas 3.4 pM of free DON was required to reach 50% inhibition of DON- HRP or C430-H RP binding to the same antibody (Table 5.1). This indicates a higher affinity of mAB 6F5 for C430 than for DON. in a similar CD-ELISA, none of the individual amino acids (at concentrations up to 34 pM) in C430 showed any significant binding inhibition to DON-HRP (Figure 5.6). This suggests that the 140 Figure 5.4. Absorbance at 450 nm 0.8 -~ 0.6 4- 0-4 ‘“ +00»an +Phagc DONPEP.2 0,2 __ +Phage DONPEP.12 0 l i ‘r 0 10 100 1000 10000 Competitive DON, ng/ml Competition between phage-displayed mimotope peptides and DON for binding to immobilized mAb 6F5 in CD-ELISA. Various concentrations of free DON competed with an equal volume of phage- displayed peptides (at constant concentration) for binding to immobilized mAb 6F5. Bound phage peptide was detected by horseradish peroxidase conjugated sheep anti-M13 lgG and then measured by absorbance. DON-HRP was included as a positive control. 141 100 100 °\o —D—DON °\° 90 - -D-DON g3 30‘ +C430 g3 80‘ +080 =5 'e 70 ‘ f5 60 - g 60 - “5 “a 50 ‘ g 40 - g 40 ~ :g :e 30 - E 20 « E 20 . 10 - o 0 ~ 0 0.01 0.1 1 10 100 o 0,01 0,1 1 10 100 Competitor, PM Competitor, pM Figure 5.5. Competition between synthetic peptide C430 and DON for binding to mAb 6F5. (A). DON and synthetic peptide C430 compete with DON- HRP for binding to immobilized mAb 6F5. (B). DON and synthetic peptide C430 compete with C430-H RP for binding to immobilized anti- DON mAb 6F5. 142 Table 5.1. 50% binding inhibition of DON-HRP and C430-HRP by DON and C430 in competitive ELISA with monoclonal antibody 6F5 coated in microtiter wells. Enzyme Binding competitor ( pM) conjugate DON C430 DON-HRP 3.43 0.64 C430-HRP 3.47 0.8 Absorbance at 450 nm Figure 5.6. 0'8 +Ser (S) +Trp (W) +Gly (G) -o—Pro (P) 0.4 ~ —o—Phe (F) +Cys (C) 0 . T r . 0.001 0.01 0.1 1 10 100 Competitive amino acid, pM Competition between individual amino acids in synthetic peptide C430 and DON measured with CD-ELISA. Amino acids (at various concentrations) competed with DON-HRP for binding to immobilized mAb 6F5. 143 100 9O - +DONPEP-AP 8O - -D—DON-HRP 7o — 60 J 50 - 40~ Inhibition of binding, % 30‘ 20~ 10- 1 0.001 0.01 0.1 1 10 100 Competitive DON, pM l l l l T l rrrrrr t It IT Figure 5.7. CD-ELISA with DONPEP-AP fusion protein. Binding of DONPEP-AP fusion protein to immobilized mAb 6F5 was inhibited by free DON. DON-HRP was included as a positive control. 144 sequence in C430 is important for the specific binding to mAB 6F5, while the individual amino acids can not bind to mAB 6F5 specifically. The DONPEP-AP fusion protein collected from the culture supernatant or periplasmic extract showed alkaline phosphatase activity and had a similar specific binding to mAB 6F5 as DON-HRP (Figure 5.7). This suggests that the DON mimotope peptide sequence is structurally stable in a different protein structural context. CD-ELISA for DON spiked wheat extract To test the feasibility of DON mimotope peptide sequences as immunochemical reagents for a DON immunoassay in food and feed, a CD-ELISA was performed for a DON spiked wheat extracts. Both C430-HRP conjugate and DONPEP-AP fusion protein showed binding activity to immobilized mAB 6F5 in the wheat extract (Figure 5.8). All three conjugates had a similar linear inhibition curve at DON concentrations ranging from 0.1 pglml to 10 pglml in the wheat extract. However, a slightly lower absorbance was observed in the CD-ELISA with C430- HRP and DONPEP-AP in wheat extract than in PBS buffer, indicating that the wheat extract interfered with the binding of the mimotope peptide to anti-DON mAB 6F5 to some degree. 145 Inhibition of binding, % Figure 5.8. 100 (D O -D- DON-HRP -I- DONPEP-HRP 0 10 100 1000 10000 Spiked DON (nglml) Application of C430-HRP and DONPEP-AP in DON immunoassay (CD-ELISA) in a DON-spiked wheat extract. Immulon-4 microtiter wells were coated with mAb 6F5 antibody. DON-HRP was included as a positive control. 146 Immunization with C430-BSA conjugate If the selected peptides represent an true image of the DON surface structure, they might be able to elicit an immune response similar to that of the original DON epitope, and therefore they might serve as alternative antigens for DON. When rabbits and mice were immunized with C430-BSA conjugate, both produced antibody specific to the DON mimotope peptide sequence after the second injection of C430-BSA. After four injections, the antiserum showed a strong antibody response against the DON mimotope peptide sequence (Figure 5.9). A concentration of 0.39 pM of synthetic peptide C430 caused 50% inhibition of antiserum binding to immobilized phage-displayed peptide. However, binding of antisera to immobilized phage-displayed peptide was not inhibited by free DON in solution, indicating that antibodies in these immunized animals were not specific for DON. Cytotoxicity and effects on protein synthesis in vitro One of DON’s effects is to cause cytotoxicity through cell apoptosis (Pestka et al., 1994b). To test if the DON mimotope peptide is cytotoxic, synthetic peptide C430 was compared with DON for their cytotoxic effects to bone marrow cells. As expected, DON at 3.4 pM caused 40-60% cell death after 18 hr incubation (Figure 5.10). Synthetic peptide C430, however, did not show any adverse effects on the viability of bone marrow cells in culture at concentrations up to 34 pM (Figure 5.10). When combined with DON, C430 at any of the tested concentrations did not 147 Figure 5.9. Absorbance at 450 nm O .......l “he... ..... ..... ........ 0.001 0.01 0.1 1 10 100 Competitor, pM The specificity of antibody from C430-BSA immunized mice. DON and C430 at various concentrations were used to inhibit the binding of antisera to immobilized DONPEP.2 phage. Bound mouse antibodies were detected by horseradish peroxidase conjugated goat anti-mouse lgG and measure by absorbance. 148 34 120 x 5:52., :8 Synthetic peptide C430, pM peptide C430 co-incubated with (white bar) or without (hatched bar) DON (3.4 pM). Cell viability was determined using a MTT conversion Figure 5.10. Cytotoxicity to bone marrow cells after 18 hr incubation with synthetic assay. 149 significantly increase or decrease cell viability caused by DON, suggesting that no significant synergism or antagonism between DON mimotope peptide and DON in causing bone marrow cell death. In a cell free translation system, at a concentration 3.4 pM, DON significantly inhibited new protein synthesis, while C430 showed no inhibition (Figure 5.11). However, protein synthesis was not inhibited by DON when mixed with C430 both at 3.4 pM. This suggests that C430 was antagonistic to the inhibitory effects on protein synthesis caused by DON. 150 Figure 5.11. Effects of DON (3.4 pM) and synthetic peptide C430 (3.4 pM) on protein synthesis in vitro with rabbit reticulocyte lysate. The translation template was y-globulin mRNA (2 pg). “+" or ‘-": with or without added, respectively. 151 DISCUSSION In a search for DON mimotope peptides, we screened a phage-display random- heptapeptide library and identified two closely related peptide sequences that specifically bind with high affinity to anti-DON mAb 6F5. To our knowledge, this is the first example of a mimotope peptide for a nonproteinaceous, low-molecular- weight mycotoxin. In a competitive ELISA, the binding of the selected peptides to mAb 6F5 could be competed by DON, while DON binding to the same antibody was also competed by the synthetic peptide, strongly suggesting that these clones bind to the same antigen-binding site of the monoclonal antibody. The demonstration that these peptides can be applied to ELISA has several implications. From a safety standpoint, it is advantageous to use a nontoxic peptide as an immunochemical reagent to replace toxic DON in the immunoassay. Second, the conjugation of peptide to peroxidase or other proteins will be much easier than the conjugation of DON to the same enzyme. Finally, by genetically fusing the peptide with reporter enzyme such as alkaline phosphatase, chemical conjugation can be avoided. Although many mimotope peptides for protein antigens have been isolated from phage-displayed peptide libraries, only a few examples exist where peptides were selected as mimics to non-proteinaceous chemicals, such as lectins and carbohydrates (Oldenburg et al., 1992; Scott et al., 1992; Hoess et al., 1993). It is interesting to compare the peptide sequences selected with mAb 6F5 to other peptide sequences known to interact with carbohydrate binding sites. One can ask 152 whether there are general features common to these sequences. Our sequences are similar to those carbohydrate mimics in that there is a clear preference for aromatic groups and proline residues (Oldenburg et al., 1992; Scott et al., 1992; Hoess et al., 1993). This is not unreasonable since these amino acids resemble sugar moieties in their size and cyclic shape. Furthermore, the proline may serve an important structural role by orienting the aromatic side chains spatially in a manner similar to the branched carbohydrates they mimic. if a small peptide completely mimics the epitope of a certain antigen, it can elicit antibodies directed against native antigen whose epitope the peptide represents after the peptide is conjugated on a suitable carrier protein or displayed on phage (Amon, 1991; Minenkova et al., 1993; Di-Marzo et al., 1994; Motti et al., 1994). But peptides mimicking discontinuous epitopes were unable to elicit antibodies specific to the original antigen (Felici et al., 1993). Although our selected peptides and DON competed with each other for binding to mAB 6F5 and C430 can antagonize DON’s inhibitory effects in an in vitro protein synthesis system, no specific antibodies for DON were produced from C430-BSA conjugate immunized animals when high affinity antibodies to the peptide were detected in these animals. It is not clear whether the selected peptides mimic the surface structure of DON in some respects or rather bind to the cleft by an entirely different mechanism (Figure 5.12). The latter possibility has been noted in the case of streptavidin-binding peptides discovered with a phage-display library (Weber et al., 1992), where the peptides associate with the binding site in a quite different fashion than the natural ligand, biotin. Turcatti et al. (1997) also observed that non-peptide antagonist and 153 Model 1 Model 2 Binding to mAb 6F5 Binding to mAb 6F5 Generation of DONPEP antibody Generation of DONPEP antibody after immunized with DON PEP after immunized with DONPEP Figure 5.12. Possible mechanisms for the binding of DON mimotope peptides to mAb 6F5 and for the immunogenicity of C430-BSA. In Model 1, DON and DONPEP might bind to different portions at the antigen binding pocket of mAb 6F5. After immunization with DONPEP, only anti- DONPEP antibody is produced. In Model 2, DON and DONPEP bind to mAb 6F5 in the same fashion. But after immunization with DONPEP, only the epitope which is different from DON elicits antibody response. 154 peptide agonists were vastly different when bound to the NK1 receptor. Another possibility is that more than one epitope may exist in the peptide and the moiety which mimics DON has very poor immunogenicity (Figure 5.12). This proposed mechanism is supported by the observation that specific antibody for DON was rarely detected in large numbers of animals immunized with DON carrier protein conjugates (Casale etal., 1988). The toxic effects of trichothecenes are presumably related to their protein synthesis inhibition. The inhibitory activity of trichothecenes is due to their ability to bind the 608 subunit of eukaryotic ribosomes and to interfere with peptidyltransferase activity (Bamburg, 1983; Ueno, 1983; Thompson and Wannemacher, 1986; Betina, 1989b; Rotter et al., 1996). Stoichiometricaly, one trichothecene molecule binds one ribosome (Middlebrook and Leatherrnan, 1989). Presumably peptides will only be agonists if they undergo the same specific contacts and interactions with the ribosome as DON. The mimotope peptides selected from phage-display library in this study, however, showed an antagonism to DON-induced protein synthesis inhibition. This suggests that the synthetic peptide may bind to the same site in ribosome as DON does, but in a different fashion. It is commonly accepted that native ligands, agonists, or antagonists can partly bind to their receptors (Strader et al., 1995) although an inhibitory active site might be structurally distinct from the binding site. it is notable, however, that in a cell culture model, C430 did not potentiate or attenuate DON in causing cell death. This may be due to the difficulty of the peptide crossing the cell membrane (Burton et al., 1996; Firth et al., 1997). DON, with a 4-fold lower 155 molecular weight, absence of charged groups, and specific structure, may be more readily absorbed into the cell (Witt and Pestka, 1990). In addition to the toxic effects to animals and humans, there is a growing body of evidence indicating that trichothothecenes, including DON, can enhance the virulence of plant-pathogenic species of Fusarium on plant hosts (Adams and Hart, 1989; Desjardins et al., 1989:1992; Bmins et al., 1993; Wang and Miller, 1988; Scholbrock et al., 1992; Atanassov et al., 1994; Proctor et al., 1995). These trichothecene-producing Fusarium fungi cause a broad range of plant diseases including head and seedling blight of small grains such as wheat and rye, ear and stalk rot of maize, stem rot of carnation, and seedling blight and root rot of a number of other plant species, including beans, clover, and tomato (Cook, 1981; Kommedahl and Windels, 1981). If the fungal virulence associated with DON is mediated through protein synthesis inhibition and the mimotope peptides presented here are antagonistic to DON activity in plant, a transgenic plant expressing the peptides (may need to be fused into another protein) might show reduced levels of wheat head scab and other plant diseases caused by DON-producing pathogenic Fusarium. Previously, synthetic peptide combinatorial libraries have been used to identify bioactive peptides against plant pathogens (Marcos et al., 1995; Blondelle and Houghten, 1996; Reed et al., 1997). Some antimicrobial peptides have been expressed in transgenic plants (De Bolle et al., 1996). Thus, phage-display peptide libraries may provide a new and innovative approach in developing antimicrobial peptides to control plant diseases. 156 In summary, we have identified two closely related mimotope peptides for DON and demonstrated their potential application in DON immunoassay. We also demonstrated an antagonistic activity of the mimotopes to DON-induced protein synthesis inhibition. Further understanding of the difference between the mimotope peptides and DON in the structure-function relationships might help in elucidation of the mechanisms of DON toxicity, and lead to prevention of plant and animal diseases associated with this mycotoxin. 157 SUMMARY In this dissertation, phage—display technology was explored to develop recombinant antibodies and peptide mimotopes for Fusarium mycotoxins. Firstly, a functional scFv antibody with high affinity to the mycotoxin zearalenone was cloned and expressed in E. coli. A new phage-display vector which facilitates the cloning of genes for variable heavy chain, variable kappa light chain, and the assembly of scFv DNA fragment, was also constructed. However, attempts to generate scFv antibodies to the mycotoxin fumonisin B1 from fumonisin B1-cholera toxin conjugate immunized mice failed due to inefficient selection. Secondly, the anti-zearalenone scFv DNA fragment was transferred into Arabidopsis and functional anti-zearalenone scFv plantibody with high affinity to mycotoxin zearalenone was expressed. Finally, two highly homologous heptapeptide sequences from a phage-displayed random 7-mer peptide library were identified for specifically binding a deoxynivalenol-specific monoclonal antibody. The mimotope peptide appeared to be nontoxic and showed antagonistic to mycotoxin deoxynivalenol. 158 APPENDICES: BUFFERS AND MEDIA 159 APPENDIX A: COMMONLY USED BUFFERS Bacteria alkaline lysis buffer for mini-preparations of plasmid DNA (Sambrook et al., 1989): Solution I 50 mM glucose 25 mM Tris.Cl (pH 8.0) 10 mM EDTA (pH 8.0) Autoclaved and stored at 4°C Solution ll (fresh prepared) 0.2 N NaOH (freshly diluted from a 10 N stock) 1% SDS Solution ill 5 M potassium acetate 60 ml glacial acetic acid 11.5 ml H20 28.5 ml 0.05 M carbonate-bicarbonate coating buffer (Sigma Chemical Company, St. Louis, MO): 1.59 g Na.‘,CO3 2.93 g NaHCOa dissolved in 1 liter Milli-Q water adjust pH to 9.6, if necessary stored at 4°C Citrate buffer (pH 4.0) for ABTS substrate preparation: Add 9.6 g of citric acid monohydrate to 500 ml of Milli-Q water. Adjust the pH to 4.0 with NaOH. Scale up to 1 liter and check pH. Autoclave and store at 4°C. DNA electrophoresis gel-loading buffer (Sambrook et al., 1989): Buffer type i (6 x) 0.25% bromophenol blue 0.25%xylene cyanol FF 40% (wlv) sucrose in water 4°C storage 160 Buffer type II (6 x) 0.25% bromophenol blue 0.25%xylene cyanol FF 15% Ficoll (Type 400; Pharmacia) in water room temperature storage Buffer type ill (6 x) 0.25% bromophenol blue 0.25%xylene cyanol FF 30% glycerol in water 4°C storage Buffer type iv (6 x) 0.25% bromophenol blue 40% (wlv) sucrose in water 4°C storage Phosphate-buffered saline (PBS) (Sambrook et al., 1989): To 8 g of NaCl, 0.2 g of KCI, 1.44 g of Na,HPO,, 0.24 g of KHZPO4, add Milli- Q water to a total volume of 1 liter. Adjust the pH to 7.4 with NaOH or HCI if necessary and autoclave. 0.05 M phosphate-citrate buffer for TMB substrate preparation (Sigma Chemical Company, St. Louis, MO): Dissolve 3.65 g of NazHPO, and 2.55 g of citric acid in Milli-Q water, scale up the volume to 500 ml. Adjust pH to 5.0, if necessary. Protein SDS-PAGE gel-loading buffer (2 x) (Sambrook et al., 1989) 100 mM Tris.CI (pH 6.8) 200 mM dithiothreitol (add just before the buffer is used) 4% SDS (electrophoresis grade) 0.2% bromophenol blue 20% glycerol room temperature storage (without dithiothreitol) SDS-PAGE running buffer (5 x stock) (Sambrook et al., 1989): 15.1 g Tris base 94 g glycine 5 9 SDS (electrophoresis-grade) add Milli-Q water to 1 liter 161 SDS-PAGE protein transfer buffer 12.1 g Tris base 14.4 g glycine add Milli-Q water to 1 liter Tris-buffered saline (TBS) (Boehringer Mannheim Corporation, Indianapolis, IN ): Add 6.05 g Tris base (50mM), 8.76 g NaCI (150 mM) to 800 ml Milli-Q water, adjust pH to 7.5 with HCI or NaOH, scale up to a total volume of 1 liter with Milli-Q water. Tris-EDTA (TE) buffer (Sambrook et al., 1989): pH 7.4: 10mM Tris.Cl (pH7.4) 1mM EDTA (pH8.0) pH 7.6: 10mM Tris.Cl (pH7.6) 1mM EDTA (pH8.0) pH 8.0: 10mM Tris.Cl (pH8.0) 1mM EDTA (pH8.0) Tris-acetate electrophoresis buffer (T AE, 50 x stock) (Sambrook et al., 1989y 242 g Tris base 57.1 ml glacial acetic acid 100 ml 0.5 M EDTA (pH 8.0) scale up to 1 liter with Milli-Q water 162 APPENDIX B: COMMONLY USED MEDIA LB medium (Luria-Bertani medium) (Sambrook et al., 1989) 10 g bacto-tryptone 5 g bacto-yeast extract 10 9 NaCl dissolved in Milli-Q water, adjust pH to 7.4 scale up to 1 liter, autoclave. Minimal medium (Phannacia blotech): 6 g NazHPO, (dibasic) 3 g KH,,PO4 1 g NH,C| Dissolved in Milli-Q water (500 ml), adjust pH to 7.4, autoclave; In a separate bottle, autoclave 15 g of Bacto-agar in 492 ml Milli-Q water, autoclave. Add 1 ml of1 M MgCl2 .6H20, 1 mi of 1 M CaCl2.2H20, 1 ml of 1 M thiamine hydrochloride and 5 ml of 20% glucose (all autoclave or filtration sterilized). Then combine with above salts. SB medium (Phannacia blotech): 35 g bacto-tryptone 20 g bacto-yeast extract 5 9 NaCl Dissolved in Milli-Q water, scale up to 1 liter, adjust pH to 7.4, autoclave. SOB medium (Sambrook et al., 1989): 20 g bacto-tryptone 5 g bacto-yeast extract 0.5 9 NaCl 10 ml 250 mM KCI Dissolved in Milli—Q water, adjust pH to 7.4, scaled up to 990 ml with Milli-Q water. Autoclave, after cool down to 60°C, add 10 ml of 1 M MgCl,. 2 X YT medium (Sambrook et al., 1989): 16 g bacto-tryptone 10 g bacto-yeast extract 5 9 NaCl Dissolved in Milli-Q water, adjust pH to 7.4, scale up to 1 liter, autoclave. 163 LIST OF REFERENCES Abbas, H.K., Mirocha, C.J., and Tuite, J. 1986. Natural occurrence of deoxynivalmol, 15-acetyl-deoxynivalenol, and zearalenone in refusal factor corn stored since 1972. Appl. Environ. Microbiol. 51:841-843. Abouzied, M.M., Azcona, J.l., Braselton, W.E., and Pestka, J.J. 1991. lmmunochemicai assessment of mycotoxins in 1989 grain foods: Evidence for deoxynivalenol (vomitoxin) contamination. Appl. Environ. Microbiol. 57:672-677. Adams, 6.0. and Hart, LP. 1989. The role of deoxynivalenol and 15- acetyldeoxynivalenol in pathogenesis by Gibberelle 2939, as elucidated through protoplast fusions between toxigenic and nontoxigenic strains. Phytopathology 79:404-408. Adey, N.B., Mataragnon, A.H., Rider, J.E., Carter, J.M., and Kay, B.K. 1995. Characterization of phage that bind plastic from phage-displayed random peptide libraries. Gene 156:27-31. Alberts, J.F., Gelderblom, W.C., Thiel, P.G., Marasas, W.F., van, SD, and Behrend, Y. 1990. Effects of temperature and incubation period on production of fumonisin B1 by Fusarium moniliforme. Appl. Environ. Microbiol. 56:1 729-1733. Appelgren, L.E., Arora, R.G., and Larsson, P. 1982. Autoradiographic studies of [5H] zearalenone in mice. Toxicol. 25:243-253. Amon, R. 1991. Synthetic peptides as the basis for vaccine design. Mol. Immunol. 28:209-21 5. Artsaenko, O., Peisker, M., zur Nieden, U., Fiedler, U., Weiler, E.W., Muntz, K., and Conrad, U. 1995. Expression of a single-chain Fv antibody against abscisic acid creates a wilty phenotype in transgenic tobacco. Plant J. 8:745-750. Atanassov, 2., Nakamura, C., Mori, N., and Kaneda, C. 1994. Mycotoxin production and pathogenicity of Fusarium species and wheat resistance to Fusarium head blight. Can. J. Bot. 72:161-167. Azcona-Olivera, J.l., Abouzied, M.M., Plattner, R.D., Norred, WP, and Pestka, J.J. 19923. Generation of antibodies reactive with fumonisins B1 , 32, and B3 by using cholera toxin as the carrier-adjuvant. Appl. Environ. Microbiol. 58:169- 1 73. Azcona-Olivera, J.l., Abouzied, M.A., Plattner, RD, and Pestka, J.J. 1992b. Production of monoclonal antibodies to the mycotoxins fumonisin B,, B,, and 8,. J. Agri. Food Chem. 40:531-534. 164 Baca, M., Scanlan, T.S., Stephenson, RC, and Wells, J.A. 1997. Phage display of a catalytic antibody to optimize affinity for transition-state analog binding. Proc. Natl. Acad. Sci. USA 94:10063-10068. Bacon, CW. and Nelson, PE. 1994. Fumonisin production in corn by toxigenic strains of Fusarium moniliforme and Fusarium proliferatum. J. Food Protect. 57:514-521. Bamburg, JR. and Strong, FM. 1971 . 12,13-epoxitrichothecenes. pp. 207-292. In S. Kadis, A. Ciegler, and S.L. Aji (eds.) , Microbial Toxins, Vol. VII. Academic Press, New York, NY. Bamburg, JR. 1983. Biological and biochemical actions of trichothecene mycotoxins. pp. 41-110. In F.E. Hahn (ed.) , Progress in Molecular and Subcellular Biology, Vol. 8. Springer-Vedag, Berlin, Germany. Barbas, C.F., Kang, A.S., Lerner, RA, and Benkovic, SJ. 1991. Assembly of combinatorial antibody libraries on phage surfaces: the gene Ill site. Proc. Natl. Acad. Sci. USA 88:7978-7982. Bars, J.L., Bars, P.L., Dupuy, J., and Boudra, H. 1994. Biotic and abiotic factors in fumonisin B,production and stability. J. AOAC lntemational 77:517-521. Baud, F.J.,Borron,S.W.,Scherrmann,J.M.,and Bismuth, C.1997.Acritical review of antidotal immunotherapy for low molecular weight toxins. Current antidotes and perspectives. Arch. Toxicol. Suppl. 19:271-287. Baum, T.J., Hiatt, A., Parrott, W.A., Pratt, L.H., and Hussey, RS. 1996. Expression in tobacco of a functional monoclonal antibody specific to stylet secretions of the root-knot nematode. Moi. Plant-Microb. interact. 9:382-387. Beardall, J.M. and Miller, JD. 1994. Diseases in humans with mycotoxins as possible causes. pp. 487-539. In J.D. Miller and H.L. Trenholm (eds.) , Mycotoxins in Grain: Compouds Other Than Aflatoxin. Eagan Press, St. Paul, MN. Becci, P.J., Johnson, W.D., Hess, P.G., Gallo, M.A., Pareet, RA, and Taylor, J.M. 1982. Combined two-generation reproduction-teratogenesis study of zearalenone in the rat. J. Appl. Toxicol. 2:201-206. Bechtold, N., Ellis, J., and Pelletier, G. 1993. In planta Agrobacterium mediated gene transfer by infiltration of adult Arabidopsis thaliana plants. CR Acad. Sci. Ser. III Sci. 316:1194-1199. Bennett, G.A., Nelsen, TC, and Miller, BM. 1994. Enzyme-linked immunosorbent assay for detection of zearalenone in corn, wheat, and pig feed: collaborative study. J. AOAC. Int. 77:1500-1509. 165 Benvenuto, E., Ordas, R.J., Tavazza, R., Ancora, G., Biocca, S., Cattaneo, A., and Galeffi, P. 1991. 'Phytoantibodies": A general vector for the expression of immunoglobulin domains in transgenic plants. Plant Mol. Biol. 17:865-874. Betina, V. 1989a. Bioactive Molecules Vol.9: Mycotoxins: Chemical, Biological and Environmental Aspects. Elsevier Science Publishing Company, lnc., New York, NY. Betina, V. 1989b. Structure-activity relationships among mycotoxins. Chem. Biol. Interact. 71:105-146. Better, M., Chang, C.P., Robinson, RR, and Horwitz, AH. 1988. Escherichia coli secretion of an active chimeric antibody fragment. Science 240:1041-1043. Bezuidenhout, S.C., Gelderblom, W.C.A., Gorst-Allman, C.P., Horak, R.M., Marasas, W.F.O., Spiteller, G., and Vleggaar, R. 1988. Structure elucidation of the fumonisins, mycotoxins from Fusarium moniliforme. J. Chem. Soc. Chem. Commun. 743-745. Biehl, M.L., Prelusky, D.B., Koritz, G.D., Hartin, K.E., Buck, W.B., and Trenholm, H.L. 1993. Biliary excretion and enterohepatic cycling of zearalenone in immature pigs. Toxicol. Appl. Pharmacol. 121 :1 52-1 59. Bird, R.E., Hardman, K.D., Jacobson, J.W., Johnson, 8., Kaufman, B.M., Lee, S.- M., Lee, T., Pope, H.S., Riordan, 6.8., and Whitlow, M. 1988. Single-chain antigen-binding proteins. Science 242:423-426. Blackwell, B.A., Miller, JD, and Savard, ME. 1994. Productionofcarbon 14-labeled fumonisin in liquid culture. J. AOAC lntemational 77:506-511. Blondelle, SE. and Houghten, RA. 1996. Novel antimicrobial compounds identified using synthetic combinatorial library technology. Trends. Biotechnol. 14:60- 65. Bock, K.W., Lilienblum, W., Fischer, G., Schirrner, G., and Bock-Henning BS 1987. The role of conjugation reactions in detoxication. Arch. Toxicol. 60:22-29. Bories, G.F., Perdu-Durand, E.F., Sutra, J.F., and Tulliez, J.E. 1991. Evidence for glucuronidation and sulfation of zeranol and metabolites (taleranol and zearalenone) by rat and pig hepatic subfractions. Drug Metab. Dispos. 19:140-143. Bosch, U. and Mirocha, C.J. 1992. Toxin production by Fusarium species from sugar beets and natural occurrence of zearalenone in beets and beets fibers. Appl. Environ. Microbiol. 58:3233-3239. Bresler, G., Vaamonde, G., and Brizzio, S. 1991 . Natural occurence of zearalenone and toxicogenic fungi in amaranth grain. Int. J. Food Microbiol. 13:75-80. 166 Brinkmann, U., Reiter, Y., Jung, S.H., Lee, B., and Pastan, l. 1993. A recombinant immunotoxin containing a disulfide-stabilized Fv fragment. Proc. Natl. Acad. Sci. USA 90:7538-7542. Bmins, M.B.M., Karsai, l., Schepers, J., and Snijders, C.H.A. 1993. Phytotoxicity of deoxynivalenol to wheat tissue with regard to in vitro selection for Fusarium head blight resistance. Plant Sci. 94:195-206. Bmyns, A.M., De, J.G., De, N.M., De, W.C., Van, MM, and Depicker, A. 1996. Bacterial and plant-produced scFv proteins have similar antigen-binding properties. FEBS Lett. 386:5-10. Burton, P.S., Conradi, R.A., Ho, N.F., Hilgers, A.R., and Borchardt, RT. 1996. How structural features influence the biomembrane permeability of peptides. J. Pharm. Sci. 85:1336-1340. Campbell, A.D., Whitaker, T.B., Pohland, A.E., Dickens, J.W., and Park, D.L. 1986. Sampling, sample preparation, and sampling plans for foodstuffs for mycotoxin analysis. Pure Appl. Chem. 58:305 Carpita, N., Sabularse, D., Montezinos, D., and Delmer, DP. 1979. Determination of the pore size of cell walls of living plant cells. Science 205:1144-1147. Carrier, A., Ducancel, F., Settiawan, N.B., Cattolico, L., Maillere, B., Leonetti, M., Drevet, P., Menez, A., and Boulain, JC. 1995. Recombinant antibody- alkaline phosphatase conjugates for diagnosis of human lgGs: application to anti-HBsAg detection. J. Immunol. Methods 181 :177-186. Casale, W.L., Pestka, J.J., and Hart, LP. 1988. Enzyme-linked immunosorbent assay employing monoclonal antibody specific for deoxynivalenol (vomitoxin) and several analogues. J. Agric. Food Chem. 36:663-668. CAST. 1989. Mycotoxins: Economic and Health Risks. Task Force Report No.116. Council for Agricultural Science and Technology, Ames, Iowa. (GENERIC) Ref Type: Serial (Book,Monograph) Cawood, M., Gelderblom, W.C., Vleggaar, R., Behrend, Y., Thiel, P.G., and Marasas, W.F.O. 1991. Isolation of fumonisin mycotoxins: A quantitative approach. J. Agri. Food Chem. 39:1958-1962. Chakrabarti, D.K. and Ghosal, S. 1986. Occurrence of free and conjugated 12,13- epoxytrichothecenes and zearalenone in banana fruits infected with Fusarium moniliforme. Appl. Environ. Microbiol. 51:217-219. Chakrabarti, D.K. and Ghosal, S. 1987. Mycotoxins produced by Fusarium oxysporum in the seeds of Brassica campestn's during storage. Mycopathologia 97:69-75. 167 Chang, K., Kurtz, H.J., and Mirocha, C.J. 1979. Effect of the mycotoxin zearalenone on swine reproduction. Am. J. Vet. Res. 40:1260-1267. Chanh, T.C., Huot, R.I., Schick, MR, and Hewetson, J.F. 1989. Anti-idiotypic antibodies against a monoclonal antibody specific for the trichothecene mycotoxin T-2. Toxicol. Appl. Pharmacol. 100:201-207. Chanh, T.C., Rappocciolo, G., and Hewetson, J.F. 1990. Monoclonal anti-idiotype induces protection against the cytotoxicity of the trichothecene mycotoxin T- 2. J. Immunol. 144:4721-4728. Chanh, T.C., Siwak, EB, and Hewetson, J.F. 1991. Anti-idiotype-based vaccines against biological toxins. Toxicol. Appl. Pharmacol. 108:183-193. Chanh, T.C., Kennedy, R.C., and Hewetson, J.F. 1992. Anti-idiotype vaccines in toxicology. lntemat. J. Clin. Lab. Res. 22:28-35. Cheng, S.J., Jiang, Y.Z., Li, M.H., and Lo, H2. 1985. A mutagenic metabolite produced by Fusarium moniliforme isolated from Linxia county, China. Carcinogenesis 6:903-905. Chichila, T.M., Silvestre, D., Covey, TR, and Henion, JD. 1988. Distribution of zeranol in bovine tissues determined by selected ion monitoring capillary gas chromatography/mass spectrometry. J. Anal. Toxicol. 12:310-318. Chu, SC. 1991. Development and use of immunoassays in detection of the ecologically important mycotoxins. In D. Bhatnagar, E.B. Lillihoj, and D.K. Arora (eds.) , Handbook of Applied Mycology Vol. V. Mycotoxin. Marcel Dekker, Inc., New York, NY. Chu, F.S. 1992. Recent progress on analytical techniques for mycotoxins in feedstuffs. J. Anim. Sci. 70:3950-3963. Chu, PS. and Li, G.Y. 1994. Simultaneous occurrence of fumonisin B1 and other mycotoxins in moldy corn collected from the People's Republic of China in regions with high incidences of esophageal cancer. Appl. Environ. Microbiol. 60:847-852. Chu, F.S., Huang, X., and Maragos, CM. 1995. Production and Characterization of Anti-idiotype and Anti-anti-idiotype Antibodies against F umonisin B-1. J. Agric. Food Chem. 43:261-267. Clackson, T., Hoogenboom, H.R., Griffiths, AD, and Winter, G. 1991. Making antibody fragments using phage display libraries. Nature 352:624-628. Coe, J.E., Ishak, K.G., Ward, J.M., and Ross, M.J. 1992. Tamoxifen prevents induction of hepatic neoplasia by zeranol, an estrogenic food contaminant. Proc. Natl. Acad. Sci. USA 89:1085—1089. 168 Conrad, U. and Fiedler, U. 1994. Expression of engineered antibodies in plant cells. Plant Mol. Biol. 26:1023—1030. Cook, R.J. 1981. Fuan'um diseases of wheat and other small grains in North America. pp. 39-52. In P.E. Nelson, T.A. Toussoun, and R.J. Cook (eds.), Fusarium: Diseases, Biology, and Taxonomy. The Pennsylvania State University Press, University Park, PA. Cortese, R., Monaci, P., Nicosia, A., Luzzago, A., Felici, F., Galfre, G., Pessi, A., Tramontano, A., and Sollazzo, M. 1995. Identification of biologically active peptides using random libraries displayed on phage. Curr. Opin. Biotechnol. 6:73-80. Cote, L.M., Reynolds, J.D., Vesonder, R.F., Buck, W.B., Swanson, S.P., Coffey, R.T., and Brown, DC. 1984. Survey of vomitoxin-contaminated feed grains in midwestem United States, and associated health problems in swine. J. Am. Vet. Med. Assoc. 184:189-192. Coulombe, RA. 1993. Symposium: Biological action of mycotoxins. J. Dairy Sci. 76:880-891. Cwirla, S.E., Peters, E.A., Barrett, R.W., and Dower, W.J. 1990. Peptides on phage: a vast library of peptides for identifying ligands. Proc. Natl. Acad. Sci. USA 87:6378-6382. Dailey, R.E., Reese, R.E., and Brouwer, A. 1980. Metabolism of [“0] zearalenone in laying hens. J. Agric. Food Chem. 28:286-291. De Bolle, M.F., Osborn, R.W., Goderis, l.J., Noe, L., Acland, D., Hart, C.A., Torrekens, S., Van Leuven F., and Broekaert, W.F. 1996. Antimicrobial peptides from Mirabilis jelapa and Amaranthus caudatus: expression, processing, localization and biological activity in transgenic tobacco. Plant Mol. Biol. 31 :993-1008. De Neve, M., De Loose, M., Jacobs, A., Van Houdt, H., Kaluza, B., Weidle, U., Van Montagu, M., and Depicker, A. 1993. Assembly of an antibody and its derived antibody fragment in Nicotiana and Arabidopsis. Transgenic. Res. 2:227-237. De Wilde, C., De Neve, M., De Rycke, R., Bruyns, A.M., De Jaeger, G., Van Montagu, M., Depicker, A., and Engler, G. 1996. Intact antigen-binding MAK33 antibody and F-ab fragment accumulate in intercellular spaces of Arabidopsis thaliana. Plant Sci. 114:233-241. Denecke, J., Botterrnann, J., and Deblaere, R. 1990. Protein secretion in plant cells can occur via the default pathway. Plant Cell 2:51-59. 169 Desjardins, A.E., Spencer, G.F., Plattner, RD, and Beremand, MN. 1989. Furanocoumarin phytoalexins, trichothecene toxin, and infection of Pastinaca sativa by Fusarium sporotrichioides. Phytopathology 79:170-175. Desjardins, A.E., Hohn, T.M., and McCormick, SP. 1992. Effect of gene disruption of trichodiene synthase on the virulence of Gibberella pulican's. Mol. Plant- Microbe Interact. 5:214-222. Desjardins, A.E., Plattner, R.D., Nelsen, TC, and Leslie, J.F. 1995. Genetic analysis of fumonisin production and virulence of Gibberella fujikuroi mating pepulation A (Fusarium moniliforme) on maize (Zea mays) seedlings. Appl. Environ. Microbiol. 61:79-86. Desjardins, A.E. and Hohn, TM. 1997. Mycotoxins in plant pathogenesis. MoI.Plant-Microbe Interact. 10: 1 47-1 52. Devlin, J.J., Panganiban, LC, and Devlin, PE. 1990. Random peptide libraries: a source of specific protein binding molecules. Science 249:404-406. Di-Marzo, V.F., Willis, A.E., Boyer, T.C., Appella, E., and Perham, RN. 1994. Structural mimicry and enhanced immunogenicity of peptide epitopes displayed on filamentous bacteriophage. The V3 loop of HIV-1 gp120. J. Mol. Biol. 243:167-172. Dixon, D.E., Warner, R.L., Ram, B.P., Hart, LR, and Pestka, J.J. 1987. Hybridoma cell line production of a specific monoclonal antibody to the mycotoxins zearalenone and a-zearalenol. J. Agric. Food Chem. 35:122-126. Dixon, D.C., Cutt, JR, and Klessig, OF. 1991. Differential targeting of the tobacco PR-1 pathogenesis-related proteins to the extracellular space and vacuoles of crystal idioblasts. EMBO J. 10:1317-1324. Dixon, SN. and Russell, K.L. 1986. Radioimmunoassay of the anabolic agent zeranol. IV. The determination of zeranol concentrations in the edible tissues of cattle implanted with zeranol (Ralgro). J. Vet. Pharmacol. Ther. 9:94-100. Doko, M.B., Rapior, S., Visconti, A., and Schjoth, E. 1995. Incidence and levels of fumonisin contamination in maize genotype grown in Europe and Africa. J. Agric. Food Chem. 43:429-434. Dong, W., Sell, J.E., and Pestka, J.J. 1991. Quantitative assessment of mesangial immunoglobulin A (lgA) accumulation, elevated circulating lgA immune complexes, and hematuria during vomitoxin-induced lgA nephropathy. Fundam. Appl. Toxicol. 1 7: 1 97-207. Dong, W. and Pestka, J.J. 1993. Persistent dysregulation of IgA production and lgA nephropathy in the B6C3F1 mouse following withdrawal of dietary vomitoxin (deoxynivalenol). Fundam. Appl. Toxicol. 20:38-47. 170 Doorbar, J. and Winter, G. 1994. Isolation of a peptide antagonist to the thrombin receptor using phage display. J. Mol. Biol. 244:361-369. During, K., Hippe, S., Kreuzaler, F., and Schell, J. 1990. Synthesis and self- assembly of a functional monoclonal antibody in transgenic Nicotiana tabacum. Plant Mol. Biol. 15:281-293. El-Morshedy, MM. and Aziz, H.H. 1995. Effects of fenamiphos, carbofuran, and aldicarb on zearalenone production by toxigenic Fusarium spp. contaminating roots and fruits of tomato. Bull. Environ. Contam. Toxicol. 54:514-518. Elissalde, M.H., Kamps-Holtzapple, C., Beier, R.C., Plattner, R.D., Rowe, L.D., and Stanker, L.H. 1995. Development of an improved monoclonal antibody- based ELISA for fumonisin B,,3 and use of molecular modeling to explain observed detection limits. Food Agric. lmmunol. 7:109-122. Essig, N.Z., Wood, J.F., Howard, A.J., Raag, R., and Whitlow, M. 1993. Crystallization of single-chain Fv fragments. J. Mol. Biol. 234:897-901. Fecker, L.F., Kaufmann, A., Commandeur, U., Commandeur, J., Koenig, R., and Burgerrneister, W. 1996. Expression of single-chain antibody fragments (scFv) specific for beet necrotic yellow vein virus coat protein or 25 kDa protein in Escherichia coli and Nicotiana benthamiana. Plant Mol. Biol. 32:979-986. Felici, F., Castagnoli, L., Musacchio, A., Jappelli, R., and Cesareni, G. 1991. Selection of antibody ligands from a large library of oligopeptides expressed on a mutivalent exposition vector. J. Mol. Biol. 222:301-310. Felici, F., Luzzago, A., Folgori, A., and Cortese, R. 1993. Mimicking of discontinuous epitopes by phage-displayed peptides, II. Selection of clones recognized by a protective monoclonal antibody against the Bordetella pertussis toxin from phage peptide libraries. Gene 128:21-27. Fiedler, U. and Conrad, U. 1995. High-level production and long-term storage of engineered antibodies in transgenic tobacco seeds. Bio/Technol. 13:1090- 1093. Fiedler, U., Phillips, J., Artsaenko, O., and Conrad, U. 1997. Optimization of scFv antibody production in transgenic plants. lmmunotechnology. 3:205-216. F igurski, D. and Helinski, DR. 1979. Replication of a origin-containing derivative of plasmid RK2 dependent on a plasmid function provided in trans. Proc. Natl. Acad. Sci. USA 76:1648-1652. Firek, S., Draper, J., Owen, M.R.L., Gandecha, A., Cockbum, B., and Whitelam, GO. 1993. Secretion of a functional single-chain Fv protein in transgenic tobacco plants and cell suspension cultures. Plant Mol. Biol. 23:861-870. 171 Firth, K.L., Brownel, H.L., and Raptis, L. 1997. Improved procedure for electroporation of peptides into adherent cells in situ. BioTechniques 23:644-646. Fitzpatrick, D.W., Arbuckle, L.D., and Hassen, AM. 1988. Zearalenone metablism and excretion in the rat: effect of different doses. J. Environ. Sci. Health (B) 23:343-354. Forsell, J.H. and Pestka, J.J. 1985. Relation of 8-ketotrichothecene and zearalenone analog structure to inhibition of mitogen-induced human lymphocyte blastogenesis. Appl. Environ. Microbiol. 50:1304-1307. Forsell, J.H., Witt, M.F., Tai, J.H., Jensen, R., and Pestka, J.J. 1986. Effects of 8- week exposure of the B6C3F1 mouse to dietary deoxynivalenol (vomitoxin) and zearalenone. Food Chem. Toxicol. 24:213-219. Freeman, 6.6. and Morrison, RI. 1948. Trichothecin: An antifungal metabolic product of Trichothecium roseum Link. Nature 162:30-33. Freeman, 6.6. and Morrison, RI. 1949. The isolation and chemical properties of trichothecin, an antifungal substance from Trichothecium roseum Link. Biochem. J. 44:1-5. Fukuda, S., Nagahara, A., Kikuchi, M., and Kumagai, S. 1994. Preparation and characterization of anti-fumonisin monoclonal antibodies. Biosci. Biotechnol. Biochem. 58:765-767. Gelderblom, W.C., Jaskiewic, K., and Marasas, W.F. 1988a. Cancer promoting potential of different strains of Fusarium moniliforme in short-term cancer initiation/promotion assay. Carcinogenesis 9:1405-1409. Gelderblom, W.C., Jaskiewicz, K., Marasas, W.F., Thiel, P.G., Horak, R.M., Vleggaar, R., and Kriek, N.P. 1988b. Fumonisins—novel mycotoxins with cancer-promoting activity produced by Fusarium moniliforme. Appl. Environ. Microbiol. 54:1806-1811. Gelderblom, W.C., Kriek, N.P., Marasas, W.F., and Thiel, RC. 1991. Toxicity and carcinogenicity of the Fusarium moniliforme metabolite, fumonisin BI , in rats. Carcinogenesis 12:1247-1251 . Gelderblom, W.C., Semple, E., Marasas, W.F., and Farber, E. 1992. The cancer- initiating potential of the fumonisin B mycotoxins. Carcinogenesis 13:433- 437. Gelderblom, W.C.A., Cawood, M.E., Snyman, S.D., Vleggaar, R., and Marasas, W.F. 1993. Strucutre-activity relationships of fumonisins in short term carcinogenesis and cytotoxicity assays. Food Chem. Toxicol. 31:407-414. 172 Giebel, L.B., Cass, R.T., Milligan, D.L., Young, D.C., Arze, R., and Johnson, CR. 1995. Screening of cyclic peptide phage libraries identifies ligands that bind streptavidin with high affinities. Biochemistry 34:15430-15435. Gimeno, A. 1983. Rapid thin layer chromatographic determination of zearalenone in corn, sorghum, and wheat. J. Assoc. Off. Anal. Chem. 66:565-569. Glockshuber, R., Malia, M., Pfitzinger, l., and Pluckthun. A. 1990. A comparison of strategies to stabilize immu noglobulin Fv-fragments. Biochemistry 29:1362- 1367. Gopalan, 8., Bauer, D.W., Alfano, J.R., Loniello, A.O., He, S.Y., and Collmer, A. 1996. Expression of the Pseudomonas syringae avimlence protein AvrB in plant cells alleviates its dependence on the hypersensitive response and pathogenicity (Hrp) secretion system in eliciting genotype-specific hypersensitive cell death. Plant Cell 8:1095-1105. Graham, B.M., Porter, A.J.R., and Harris, W.J. 1995. Cloning expression and characterisation of a single-chain antibody fragment to the herbicide paraquat. J. Chem. Technol. Biotechnol. 63:279-289. Griffin, T. 8., Singh, A. R., and Coulston, F. 1984. No hormonal effect in non-human primates of oral zeranol. Report to the lntemational Minerals and Chemical Company. Terre Haute, IN. Griffiths, A.D., Malmqvist, M., Marks, J.D., Bye, J.M., Embleton, M.J., McCafferty, J., Baier, M., Holliger, K.P., Gorick, B.D., Hughes, J.N., and et, a. 1993. Human anti-self antibodies with high specificity from phage display libraries. EMBO J. 12:725-734. Grosse, Y., Chekir, G.L., Huc, A., Obrecht, P.S., Dirheimer, G., Bacha, H., and Pfohl, LA. 1997. Retinol, ascorbic acid and alpha-tocopherol prevent DNA adduct formation in mice treated with the mycotoxins ochratoxin A and zearalenone. Cancer Lett. 114:225-229. Hagler, W.M., Mirocha, C.J., Pathre, S.V., and Behrens, JG. 1979. Identification of the naturally occurring isomer of zearalenol produced by Fusarium roseum 'Gibbosum' in rice culture. Appl. Environ. Microbiol. 37:849-853. Hanaichi, T., Sato, T., lwamoto, T., Malavasi-Yamashiro, J., Hoshino, M., and Mizuno, N. 1986. A stable lead by modification of Sato's method. J. Electron Microsc. 35:304-306. Harrison, L.R., Colvin, B.M., Greene, J.T., Newman, LE, and Cole-JR, J. 1990. Pulmonary edema and hydrothorax in swine produced by fumonisin 81, a toxic metabolite of Fusarium moniliforme. J. Vet. Diagn. Invest. 2:217-221. Hart, L.P., Braselton, W.E., and Stebbins, TC. 1982. Production of zearalenone and deoxynivalenol in commercial sweet corn. Plant Dis. 66:1133-1135. 173 Hart, LP. and Braselton-WE, J. 1983. Distribution of vomitoxin in dry milled fractions of wheat infected with Gibberella zeae. J Agric. Food Chem. 31:657-659. Hart, L. P. 1987. Analysis of 1986 corn and wheat crops for deoxynivalenol and zearalenone. Michigan State University Report of Swine Research. pp.156. Hart, LP. and Schabenberger, O. 1998. Variability of vomitoxin in truckloads of wheat in a wheat scab epidemic year. Plant Dis. 82:625-630. Haschek, W.M., Motelin, G., Ness, D.K., Harlin, K.S., Hall, W.F., Vesonder, R.F., Peterson, R.E., and Beasley, V.R. 1992. Characterization of fumonisin toxicity in orally and intravenously dosed swine. Mycopathologia 117:83-96. Hawkins, R.E., Russell, SJ, and Winter, G. 1992. Selection of phage antibodies by binding affinity. Mimicking affinity maturation. J. Mol. Biol. 226:889-896. Hein, M.B., Tang, Y., McLeod, D.A., Janda, K.D., and Hiatt, A. 1991. Evaluation of immunoglobulins from plant cells. Biotechnol. Prog. 7:455-461. Hiatt, A., Cafferkey, R., and Bowdish, K. 1989. Production of antibodies in transgenic plants. Nature 342:76-78. Hiatt, A. 1990. Antibodies produced in plants. Nature 344:469-470. Hiatt, A. and Ma, J.K. 1992. Monoclonal antibody engineering in plants. FEBS Lett. 307:71-75. Hidy, P.H., Baldwin, R.S., Greasham, R.L., Keith, R.L., and McMullea, JR 1977. Zearalenone and some derivatives: production and biological activities. Adv. Appl. Microbiol. 22:59-82. Hill, HR. and Stockley, PG. 1996. Phage presentation. Mol. Microbiol. 20:685-692. Hoess, R., Brinkmann, U., Handel, T., and Pastan, I. 1993. Identification of a peptide which binds to the carbohydrate-specific monoclonal antibody B3. Gene 128:43-49. Hoess, R.H., Mack, A.J., Walton, H., and Reilly, TM. 1994. Identification of a structural epitope by using a peptide library displayed on filamentous bacteriophage. J. Immunol. 153:724-729. Holliger, P., Prospero, T., and Winter, G. 1993. Diabodies: small bivalent and bispecific antibody fragments. Proc. Natl. Acad. Sci. USA 90:6444-6448. Hong, SS. and Boulanger, P. 1995. Protein ligands of the human adenovirus type 2 outer capsid identified by biopanning of a phage-displayed peptide library on separate domains of wild-type and mutant penton capsomers. EMBO J. 14:4714-4727. 174 Hoogenboom, H.R., Griffiths, A.D., Johnson, K.S., Chiswell, D.J., Hudson, P., and Winter, G. 1991. MuIti-subunit proteins on the surface of filamentous phage: methodologies for displaying antibody (Fab) heavy and light chains. Nucleic Acids Res. 19:4133-4137. Hoogenboom, H.R., Marks, J.D., Griffiths, A.O., and Winter, G. 1992. Building antibodies from their genes. immunol. Rev. 130:41-68. Hsu, K.H. and Chu, F .S. 1994. Production and characterization of anti-idiotype and anti-anti-idiotype antibodies from a monoclonal antibody against aflatoxin. J. Agric. Food Chem. 42:2353-2359. Hurd, RN. 1977. Structure-activity relationship in zearalenones. pp. 379-392. In J.V. Rodricks, C.W. Hesseltine, and MA. Mehlman (eds.) , Mycotoxins in Human and Animal Health. Pathotox Pub., Inc., Forest Park, IL. Huston, J.S., Levinson, D., Mudgett, H.M., Tai, M.S., Novotny, J., Margolies, M.N., Ridge, R.J., Bruccoleri, R.E., Haber, E., Crea, R., and Opperrnann, H. 1988. Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proc. Natl. Acad. Sci. USA 85:5879-5883. James, L.J., McGirr, LG, and Smith, T.K. 1982. High pressure liquid chromatography of zearalenone and zearalenols in rat urine and liver. J. Assoc. Off. Anal. Chem. 65:8-13. Janda, K.D., Lo, C.H., Li, T., Barbas, C.F., Wirsching, P., and Lerner, RA. 1994. Direct selection for a catalytic mechanism from combinatorial antibody libraries. Proc. Natl. Acad. Sci. USA 91:2532-2536. Jaskiewicz, K., Van Rensburg, S.J., Marasas, W.F.O., and Gelderblom, W.C.A. 1987. Carcinogenicity of Fusarium moniliforme culture material in rats. J. Natl. Cancer Inst. 78:321-325. Jemmali, M. and Mazerand, C. 1980. [The presence of zearalenone or F2 in commercial walnuts]. Ann. Microbiol.(Paris) 1318:319-321 . Jones, R.L. and Robinson, 0.6. 1989. Protein secretion in plants. Tansley Rev. 17:567-588. Jones, E.Y., Davis, S.J., Williams, A.F., Harios, K., and Stuart, DJ. 1992. Crystal structure at 2.8A resolution of a soluble form of the cell adhesion molecule CDZ. Nature 360:232-239. Jung, 8. and Pluckthun. A. 1997. Improving in vivo folding and stability of a single- chain Fv antibody fragment by loop grafling. Protein Eng. 10:959-966. 175 Kabat, E.A., Wu, T.T., Perry, H.M., Gottesman, KS, and Foeller, C. 1991. Sequences of Proteins of lmmu nological interest. US. Department of Health and Human Services, Bethesda, MD. Kang, A.S., Jones, T.M., and Burton, DR. 1991. Antibody redesign by chain shuffling from random combinatorial immunoglobulin libraries. Proc. Natl. Acad. Sci. USA 88:11120-11123. Katz, BA. 1997. Structural and mechanistic determinants of affinity and specificity of ligands discovered or engineered by phage display. Annu. Rev. Biophys. Biomol. Struct. 26:27-45. Kellerrnan, T.S., Marasas, W.F., Thiel, P.G., Gelderblom, W.C., Cawood, M., and Coetzer, J.A. 1990. Leukoencephalomalacia in two horses induced by oral dosing of fumonisin B1. Onderstepoort. J. Vet. Res. 57:269-275. Kettleborough, C.A., Ansell, K.H., Allen, R.W., Rosell, V.E., Gussow, DH, and Bendig, MM. 1994. Isolation of tumor cell-specific single-chain Fv from immunized mice using phage-antibody libraries and the re-constniction of whole antibodies from these antibody fragments. Eur. J. Immunol. 24:952- 958. Kiessling, K.-H. and Patterson, H. 1978. Metabolism of zearalenone in rat liver. Acta Pharmacol. Toxicol. 43:285-290. Kommedahl, T. and Windels, CE. 1981. Root-, stalk-, and ear-infecting Fusarium species on corn in the USA. pp. 94-103. In P.E. Nelson, T.A. Toussoun, and R.J. Cook (eds.) , Fusarium: Diseases, Biology, and Taxonomy. The Pennsylvania State University Press, University Park, PA. Kortt, A.A., Malby, R.L., Caldwell, J.B., Gruen, L.C., lvancic, N., Lawrence, M.C., Hewlett, G.J., Webster, R.G., Hudson, P.J., and Colman, PM. 1994. Recombinant anti-sialidase single-chain variable fragment antibody. Characterization, formation of dimer and higher-molecular-mass multimers and the solution of the crystal structure of the single-chain variable fragment/sialidase complex. Eur. J. Biochem. 221 :151-157. Kramer, K. and Hock, B. 1996. Recombinant single-chain antibodies against 3- triazines. Food Agric. lmmunol. 8:97-109. Kuiper-Goodman, T., Scott, RM, and Watanabe, H. 1987. Risk assessment of the mycotoxin zearalenone. Regul. Toxicol. Pharmacol. 7:253—306. Kushida, H. 1974. Letten A new method for embedding with a low viscosity epoxy resin "Quetol 651'. J. Electron Microsc. 23:197. Lamprecht, S.C., Marasas, W.F., Alberts, J.F., Cawood, M.E., Gelderblom, W.C.A., Shephard, G.S., Thiel, P.G., and Calitz, F.J. 1994. Phytotoxicity of fumonisins and TA-toxin to corn and tomato. Phytopathology 84:383-391. 176 Lawler, A.M., Keamer, J.F., Kuehl, M., and Gearhart, P.J. 1989. Early rearrangements of gene encoding murine immunoglobulin k chains, unlike genes encoding heavy chains, use variable gene segments dispersed throughout the locus. Proc. Natl. Acad. Sci. USA 86:6744-6747. Lerner, R.A., Kang, A.S., Bain, J.D., Burton, DR, and Barbas, CF. 1992. Antibodies without immunization. Science 258:1313-1314. Lindsay, D.G. 1985. Zeranola ”nature-identical" oestrogen? Food Chem. Toxicol. 23:767-774. Liu, M.T., Ram, B.P., Hart, LP, and Pestka, J.J. 1985. Indirect enzyme-linked immunosorbent assay for the mycotoxin zearalenone. Appl. Environ. Microbiol. 50:332-336. Lowman, HB 1997. Bacteriophage display and discovery of peptide leads for dmg development. Annu. Rev. Biophys. Biomol. Struct. 26:401-424. Lund, P. and Dunsmuir, P. 1992. A plant signal sequence enhances the secretion of bacterial ChiA in transgenic tobacco. Plant Mol. Biol. 18:47-53. Luo, D., Mah, N., Krantz, M., Wilde, K., Wishart, D., Zhang, Y., Jacobs, F., and Martin, L. 1995. Vl-linker-Vh orientation-dependent expression of single chain Fv-containing an engineered disulfide-stabilized bond in the framework regions. J.Biochem.Tokyo. 118:825-831. Ma, J.K.C., Lehner, T., Stabila, P., Fux, CL, and Hiatt, A. 1994. Assembly of monoclonal antibodies with lgG1 and lgA heavy chain domains in transgenic tobacco plants. Eur. J. Immunol. 24:131-138. Ma, J.K.C., Hiatt, A., Hein, M., Vine, N.D., Wang, F., Stabila, P., van, D.C., Mostov, K., and Lehner, T. 1995. Generation and assembly of secretory antibodies in plants. Science 268:716-719. Ma, J.K.C. and Hein, MB. 1995. Plant antibodies for immunotherapy. Plant Physiol. 109:341-346. Mallender, W.D. and Voss, E.W.Jr. 1994. Construction, expression, and activity of a bivalent bispecific single-chain antibody. J. Biol. Chem. 269:199-206. Marasas, W. F., Nelson, P. E., and Toussoun, T. A. 1984. Toxigenic Fusarium species: Identity and Mycotoxicology. Pennsylvania State University Press, University Park, PA. Marasas, W. F. 1986. Fusarium moniliforme: A mycotocicological miasma. 6th lntemational lUPAC Symposium on Mycotoxins and Phytotoxins, Pretoria, South Afica, July 22-25, 1985. pp.19-28. Elsevier Science Publishing Company, Inc., Amsterdam, The Netherlands. 177 Marasas, W.F., Jaskiewic, K., Venter, F.S., and Van Schalkwyk, D.J. 1988a. Fusarium moniliforme contamination of maize in esophageal cancer areas in Transkei. S. Afr. Med. J. 74:110-114. Marasas, W.F., Kellennan, T.S., Gelderblom, W.C., Coetzer, J.A., Thiel, P.G., and van-der Lugt, J.J. 1988b. Leukoencephalomalacia in a horse induced by fumonisin B1 isolated from Fusarium moniliforme. Onderstepoort. J. Vet. Res. 55:197-203. Marasas, W.F. 1995. Fumonisins: their implications for human and animal health. Nat. Toxins 3:193-198. Marcos, J.F., Beachy, R.N., Houghten, R.A., Blondelle, SE, and Perez, PE. 1995. Inhibition of a plant virus infection by analogs of melittin. Proc. Natl. Acad. Sci. USA 92:12466-12469. Marin, M.L., Wong, S.-S., and Pestka, J.J. 1996. Increased lL-1, lL-6 and TNFa secretion and mRNA levels in WEHl-3 cells exposed to cyclopiazonic acid. Toxicol. 1 14:67-79. Marks, J.D., Hoogenboom, H.R., Bonnert, T.P., McCafferty, J., Griffiths, AD, and Winter, G. 1991. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J. Mol. Biol. 222:581-597. Marks, J.D., Griffiths, A.D., Malmqvist, M., Clackson, T.P., Bye, J.M., and Winter, G. 1992a. By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology N.Y. 10:779-783. Marks, J.D., Hoogenboom, H.R., Griffiths, AD, and Winter, G. 1992b. Molecular evolution of proteins on filamentous phage. Mimicking the strategy of the immune system. J. Biol. Chem. 267:16007-16010. McCafferty, J., Griffiths, A.D., Winter, G., and Chiswell, D.J. 1990. Phage antibodies: filamentous phage displaying antibody variable domains. Nature 348:552-554. McCafferty, J., Fitzgerald, K.J., Eamshaw, J., Chiswell, D.J., Link, J., Smith, R., and Kenten, J. 1994. Selection and rapid purification of murine antibody fragments that bind a transition-state analog by phage display. Appl. Biochem. Biotechnol. 472157-171 . McCartney, J.E., Tai, M.S., Hudziak, R.M., Adams, G.P., Weiner, L.M., Jin, D., Stafford, W.F., Liu, S., Bookman, M.A., Laminet, AA, and et, a. 1995. Engineering disulfide-linked single-chain Fv dimers [(st')2] with improved solution and targeting properties: anti-digoxin 26-10 (st‘)2 and anti-c-erbB-2 741 F8 (st')2 made by protein folding and bonded through C-terminal cysteinyl peptides. Protein Eng. 8:301-314. 178 McConnell, S.J., Kendall, M.L., Reilly, T.M., and Hoess, RH. 1994. Constrained peptide libraries as a tool for finding mimotopes. Gene 151:115-118. McLafferty, M.A., Kent, R.B., Ladner, R.C., and Markland, W. 1993. M13 bacteriophage displaying disulfide-constrained microproteins. Gene 128:29- 36. Merrill, A.H.Jr., Wang, E., Schroeder, J.J., Smith, E.R., Yoo, H.-S., and Riley, RT. 1995. Distruption of sphingolipid metabolism in the toxicity and carcinogenicity of fumonisins. pp. 429-443. In M. Eklund, J.L. Richard, and K. Mise (eds.) , Molecular Approaches to Food Safety: Issues Involving Toxic Microorganisms. Alskan, Inc., Fort Collins, Corolado. Middlebrook, J.L. and Leathennan, D.L. 1989. Binding of T-2 toxin to eukaryotic cell ribosomes. Biochem. Pharmacol. 38:3103-3110. Miller, J.K., Hacking, A., Harrison, J., and Gross, V.J. 1973. Stillbirths, neonatal mortality, and small litters in pigs associated with ingestion of Fusarium toxin by pregnant sows. Vet. Rec. 93:555-559. Minenkova, 0.0., llyichev, A.A., Kishchenko, GP, and Petrenko, VA. 1993. Design of specific immunogens using filamentous phage as the carrier. Gene 128:85-88. Mirocha, C.J., Christensen, CM, and Nelson, CH 1971. F-2 (zearalenone) estrogenic mycotoxin from Fusarium. pp. 107-138. In S. Kadis, A. Ciegler, and SJ. Ayl (eds.) , Microbial Toxins, Vol. 7. Academic Press, New York, NY. Mirocha, OJ. and Christensen, CM. 1974. Oestrogenic mycotoxins synthesized by Fusarium. pp. 129-148. In I.F.H. Purchase (ed.) , Mycotoxins. Elsevier Scientific publishing Co., Amsterdam, The Netherlands. Mirocha, C.J., Pathre, S.V., Schauerhamer, B., and Christensen, CM. 1976. Natural occurrence of Fusarium toxins in feedstuff. Appl. Environ. Microbiol. 32:553-556. Mirocha, C.J., Pathre, S.V., and Christensen, C.M. 1977a. Zearalenone. pp. 345- 364. In J.V. Rodricks, C.W. Hesseltine, and MA. Mehlman (eds.) , Mycotoxins in Human and Animal Health. Pathotix Pub., Inc., Forest Park, IL. Mirocha, C.J., Pathre, S.V., and Christensen, C.M. 1977b. Chemistry of Fusarium and Stachybotrys mycotoxins. In T.D. Wyllie and LG. Morehouse (eds.) , Mycotoxic Fungi, Mycotoxins, Mycotoxicoses: An Encylopedic Handbook. Mycotoxic Fungi and Chemistry of Mycotoxins. Marcel Dekker, New York. 179 Mirocha, C.J., Pathre, S.V., and Robison. TS. 1981. Comparative metabolism of zearalenone and transmission into bovine milk. Food Cosmet. Toxicol. 19:25-30. Model, P. and Russel, M. 1988. Filamentous bacteriophage. pp. 375-456. In R. Calendar (ed.) , The Bacteriophages. Vol. 2. Plenum Press, New York, NY. Montani, M.L., Vaamonde, G., Resnik, S.L., and Buera, P. 1988. Influence of water activity and temperature on the accumulation of zearalenone in corn. Int. J. Food Microbiol. 6:1-8. Motti, C., Nuzzo, M., Meola, A., Galfre, G., Felici, F., Cortese, R., Nicosia, A., and Monaci, P. 1994. Recognition by human sera and immunogenicity of HBsAg mimotopes selected from an M13 phage display library. Gene 146:191-198. Muller, J.D., Nienhaus, G.U., Tetin, S.Y., and Voss, E.W. 1994. Ligand-binding to anti-flurescyl antibodies stability of the antigen-binding site. Biochemistry 33:6221-6227. Nedelnik, J. 1993. Phytotoxicity of some Fusarium spp. toxins to clover plants. Ochrana Rostlin 29:69-76. Neish, GA. and Cohen, H. 1981. Vomitoxin and zearalenoen production by Fusarium graminearum from winter wheat and barley in Ontario. Can. J. Plant Sci. 61 :81 1-815. Nelson, P.E., Plattner, R.D., Shackelford, DD, and Desjardins, A.E. 1991. Production of fumonisins by Fusarium moniliforme strains from various substrates and geographic areas. Appl. Environ. Microbiol. 57:2410-2412. Nisson, P.E., Rashtchian, A., and Watkins, PC 1991. Rapid and efficient cloning of Alu-PCR products using uracil DNA glyosylase. PCR Methods Appl. 1:120-123. Norred, WP. 1993. Fumonisins—mycotoxins produced by Fusarium moniliforme. J. Toxicol. Environ. Health 38:309-328. O'Neil, K.T. and Hoess, RH. 1995. Phage display: protein engineering by directed evolution. Curr. Opin. Struct. Biol. 5:443-449. O'Neil, K.T., Hoess, R.H., Jackson, S.A., Ramachandran, N.S., Mousa, SA, and DeGrado, W.F. 1992. Identification of novel peptide antagonists for GPllb/l I la from a conforrnationally constrained phage peptide library. Proteins 14:509- 515. Oi, V.T., Morrison, S.L., Herzenberg, LA, and Berg, P. 1983. lmmunoglobulin gene expression in transformed lymphoid cells. Proc. Natl. Acad. Sci. USA 80:825-829. 180 Oldenburg, K.R., Loganathan, D., Goldstein, l.J., Schultz, P.G., and Gallop, MA. 1992. Peptide ligands for a sugar-binding protein isolated from a random peptide library. Proc. Natl. Acad. Sci. USA 89:5393-5397. Olsen, M., Pettersen, H., and Kiessling, K.-H. 1981. Reduction of zearalenone to zearalenol in female rat liver by 3a-hydroxysteroid dehydrogenase. Acta Pharmacol. Toxicol. 48:157-161 . Olsen, M. and Kiessling, K.-H. 1983. Species difference in zearalenone-reducing activity in subcellular fractions of liver from female domestic animals. Acta Pharmacol. Toxicol. 52:287-291. Olsen, M., Malmlof, K., Pettersen, H., Sandholm, K., and Kiessling, K.-H. 1985a. Plasma and urinary levels of zearalenone. Acta Pharmacol. Toxicol. 56:239- 243. Olsen, M., Pettersen, H., Sandholm, K., and Kiessling, K.-H. 1985b. Quantitative liquid chromatographic method using fluorescence detection for determining zearalenone and its metabolites in blood plasma and urine. J. Assoc. Off. Anal. Chem. 68:632-635. Olsen, M., Mirocha, C.J., Abbas, H.K., and Johansson, B. 1986. Metabolism of high concentrations of dietary zearalenone by young male turkey poults. Pout. Sci. 65:1905-1910. Oriandi, R., Gussow, D.H., Jones, PT, and Winter, G. 1989. Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc. Natl. Acad. Sci. USA 86:3833-3837. Owen, M., Gandecha, A., Cockbum, B., and Whitelam, G. 1992. Synthesis of a functional anti-phytochrome single-chain Fv protein in transgenic tobacco. Bio/Technol. 10:790-794. Pandey, A., Hassen, A.M., Benedict, DR, and Fitzpatrick, D.W. 1990. Effect of UDP-glucuronyltransferase induction on zearalenone metabolism. Toxicol. Lett. 51 :203-21 1. Pantoliano, M.W., Whitlow, M., Wood, J.F., Dodd, S.W., Hardman, K.D., Rollence, M.L., and Bryan, PM 1989. Large increase in general stability for subtilisin BPN' through incremental changes in the free energy of unfolding. Biochem. 28:7205-7213. Parrnley, SF. and Smith, GP. 1988. Antibody-selectable filamentous fd phage vectors: affinity purification of target genes. Gene 73:305-318. Pathre, S.V. and Mirocha, C.J. 1977. Assay methods for trichothecenes and review of their natural occurrence. pp. 229-58. In: Rodricks.JV., et al.(ed.) Mycotoxins. Pathotox.Publ., Park Forest South. 181 Pestka, J.J., Li, Y.K., and Chu, ES. 1982. Reactivity of aflatoxin B,, antibody and aflatoxin B1 modified DNA and related metabolites. Appl. Environ. Microbiol. 44:1159-1165. Pestka, J.J., Tai, J.H., Wrtt, M.F., Dixon, DE, and Forsell, J.H. 1987. Suppression of immune response in the B6C3F1 mouse after dietary exposure to the Fusarium mycobtins deoxynivalenol (vomitoxin) and zearalenone. Food Chem.ToxicoI 25:297-304. Pestka, J.J. 1988. Enhanced surveillance of foodbome mycotoxins by immunochemical assay.J. Assoc. Off. Anal. Chem. 71 :1075-1081. Pestka, J.J. and Casale, W. 1990. Natural occuring fungal toxins. pp. 613-638. In M.S. Simmons and J. Nriagu (eds.) , Food Contamination from Environmental Sources. John Willey and Sons, Ltd, New York, NY. Pestka, J.J. 1994. Application of immunology to the analysis and toxicity assessment of mycotoxins. Food Agric. Immunol. 6:219-234. Pestka, J.J., Azcona-Olivera, J.l., Plattner, R.D., Minervini, F., Doko, MB, and Visconti, A. 1994a. Comparative assessment of fumonisin in grain-based foods by ELISA, GC-MS. and HPLC. J. Food Protect. 57:169-172. Pestka, J.J., Yan, D., and King, L.E. 1994b. Flow cytometric analysis of the effects of in vitro exposure to vomitoxin (deoxynivalenol) on apoptosis in murine T, B and lgA+ cells. Food Chem. Toxicol. 32:1125-1136. Pestka, J.J. and Bondy, G.S. 1994a. Immunotoxic effects of mycotoxins. pp. 339- 358. In J.D. Miller and H.L. Trenholm (eds.) , Mycotoxins in Grain: Compounds Other Than Aflatoxin. Eagan Press, St. Paul, MN. Pestka, J.J. and Bondy, G.S. 1994b. Mycotoxin-induced immune modulation. pp. 162-182. In J.H. Dean, M.l. Luster, A.E. Munson, and l. Kimber (eds.), Immunotoxicology and Immunopharrnacology. Raven Press, New York. NY. Pestka, J .J., Abouzied, MN, and Sutikno 1 995. lmmu nological asays for mycotoxin detection. Food Technol. 49:120-128. Peter, A. T. 1904. A fungus disease in corn. Agric. Exp. Stn. Nebraska, 17th Ann. Rep. pp.13-22. Pfohl, L.A., Chekir, G.L., and Bacha, H. 1995. Genotoxicity of zearalenone, an estrogenic mycotoxin: DNA adduct formation in female mouse tissues. Carcinogenesis 16:2315-2320. 182 Phillips, J., Artsaenko, O., Fiedler, U., Horstmann, C., Mock, H.P., Muentz, K., and Conrad, U. 1997. Seed-specific immunomodulation of abscisic acid activity induces a developmental switch. EMBO J. 16:4489-4496. Pierce, H.H., Adey, N., and Kay, B.K. 1996. Identification of cyclized calmodulin antagonists from a phage display random peptide library. Moi. Divers. 1 :259- 265. Plattner, RD. and Shackelford, DD. 1992. Biosynthesis of labeled fumonisins in liquid cultures of Fusarium moniliforme. Mycopathologia 117:17-22. Plattner, R.D., Weisleder, D., Shackelford, D.D., Peterson, RP, and Powell, R.G. 1992. A new fumonisin from solid cultures of Fusarium moniliforme. Mycopathologia 117:23—28. Pohland, A.E. 1993. Mycotoxin in review. Food Addit. Contam. 10:17-28. Pompa, G., Montesissa, C., Di-Lauro, F.M., and Fadini, L. 1986. The metabolism of zearalenone in subcellular fractions from rabbit and hen hepatocytes and its estrogenic activity in rabbits. Toxicol. 42:69-75. Prelusky, D.B., Scott, P.M., Trenholm, H.L., and Lawrence, GA. 1990. Minimal transmission of zearalenone to milk of dairy cows. J. Environ. Sci. Health B 25287-103. Price, W.D., Lovell, R.A., and McChesney, D.G. 1993. Naturally occurring toxins in feestuffs: Center for veterinary medicine perspective. J. Anim. Sci. 71 :2556- 2562. Proctor, R.H., Hohn, T.M., and McCormick. SP. 1995. Reduced virulence of Gibberella zeae caused by disruption of a trichothecene toxin biosynthetic gene. Mol. Plant-Microbe Interact. 8:593-601. Raag, R. and Whitlow, M. 1995. Single-chain Fvs. FASEB J. 9:73-80. Rasooly, L., Abouzied, M.M., Brooks, K.H., and Pestka, J.J. 1994. Polyspecific and autoreactive IgA secreted by hybridomas derived from Peyer's patches of vomitoxin-fed mice: characterization and possible pathogenic role in IgA nephropathy. Food Chem. Toxicol. 32:337-348. Rasooly, L. and Pestka, J.J. 1994. Polyclonal autoreactive IgA increase and mesangial deposition during vomitoxin-induced IgA nephropathy in the BALB/c mouse. Food Chem. Toxicol. 32:329-336. Reed, JD, Edwards, D.L., and Gonzalez, CF. 1997. Synthetic peptide combinatorial libraries: a method for the identification of bioactive peptides against phytopathogenic fungi. Moi. Plant-Microbe Interact. 10:537-549. 183 Richard, J.L., Bennett, G.A., Ross, PF, and Nelson, PE. 1993. Analysis of naturally occurring mycotoxins in feedstuffs and food. J. Anim. Sci. 71 :2563- 2574. Riley, R.T., Hinton, D.M., Chamberiain, W.J., Bacon, C.W., Wang, E., Merrill-AH, J., and Voss, K.A. 1994. Dietary fumonisin B1 induces disruption of sphingolipid metabolism in Sprague-Dawiey rats: a new mechanism of nephrotoxicity. J. Nutr. 124:594-603. Ross, P.F., Nelson, P.E., Richard, J.L., Osweiler, G.D., Rice, L.G., Plattner, RD, and Wilson, TM. 1990. Production of fumonisins by Fusarium moniliforme and Fusarium proliferatum isolates associated with equine leukoencephalomalacia and a pulmonary edema syndrome in swine. Appl. Environ. Microbiol. 56:3225-3226. Ross, PF. 1994. What are we going to do with this dead horse? J. Assoc. Off. Anal. Chem. 69:699-703. Rosso, M.N., Schouten, A., Roosien, J., Borst-Vrenssen, T., Hussey, R.S., Gommers, F.J., Bakker, J., Schots, A., and Abad, P. 1996. Expression and functional characterization of a single chain Fv antibody directed against secretions involved in plant nematode infection process. Biochem. Biophys. Res. Commun. 220:255-263. Rotter, B.A., Prelusky, DB, and Pestka, J.J. 1996. Toxicology of deoxynivalenol (vomitoxin). J. Toxicol. Environ. Health 48:1-34. Rottinghaus, G.E., Coatney, CE, and Minor, H.C. 1992. A rapid, sensitive thin layer chromatography procedure for the detection of fumonisin B1 and B2. J. Vet. Diagn. Invest. 4:326-329. Ruberti, F., Cattaneo, A., and Bradbury, A. 1994. The use of the RACE method to clone hybridoma cDNA when V region primers fail. J. Immunol. Methods 173:33-39. Sambrook, J., Fritsch, E.F., and Maniatis, T.A. 1989. Molecular Cloning: A Laboratory Manual. Cold Spring Habor Laboratory, Cold Spring Harbor, NY. Schier, R., McCall, A., Adams, G.P., Marshall, K.W., Merritt, H., Yim, M., Crawford, R.S., Weiner, L.M., Marks, 0., and Marks, JD. 1996. Isolation of picomolar affinity anti-c-erbB-2 single-chain Fv by molecular evolution of the complementarity determining regions in the center of the antibody binding site. J. Mol. Biol. 263:551-567. Scholbrock, L.L., Fleener, B.K., and Berry, J.A. 1992. Comparison of wheat seedling leaf reactions to deoxynivalenol in relation to wheat scab (Fusarium graminearum) resistance classes. Phytopathology 82:1167 184 Schots, A., De Boer, J., Schouten, A., Roosien, J., Zilverentant, J.F., Pomp, H., Bouwman-Smits, L., Overrnars, H., Gommers, F .J., Visser, B., Stiekma, W.J., and Bakker, J. 1992. ”Plantibodies": A flexible approach to design resistance against pathogens. Neth. J. Plant Pathol. 98:183-191. Schouten, A., Roosien, J., van, E.F., de, J.G., Borst, V.A., Zilverentant, J.F., Bosch, 0., Stiekema, W.J., Gommers, F.J., Schots, A., and Bakker, J. 1996. The C- terrninal KDEL sequence increases the expression level of a single-chain antibody designed to be targeted to both the cytosol and the secretory pathway in transgenic tobacco. Plant Mol. Biol. 30:781-793. Schouten, A., Roosien, J., de Boer J.M., Wilmink, A., Rosso, M.N., Bosch, 0., Stiekema, W.J., Gommers, F.J., Bakker, J., and Schots, A. 1997. Improving scFv antibody expression levels in the plant cytosol. FEBS Lett. 415:235— 241. Scott, J.K. and Smith, GP. 1990. Searching for peptide ligands with an epitope library. Science 249:386-390. Scott, J.K., Loganathan, D., Easley, R.B., Gong, X., and Goldstein, l.J. 1992. A family of concanavalin A-binding peptides from a hexapeptide epitope library. Proc. Natl. Acad. Sci. USA 89:5398-5402. Scott, PM. 1982. Assessment of quantitative methods for determination of trichothecenes in grains and grain products. J.Assoc.Off.Anal.Chem. 65:876-883. Scott, P.M., Panalaks, T., Kanhere, S., and Miles, W.F. 1978. Determination of zearalenone in comflakes and other com-based foods by thin layer chromatography, high pressure liquid chromatography, and gas-liquid chromatography. J. Assoc. Off. Anal. Chem. 61:593-600. Scott, PM. and Lawrence, GA. 1992. Liquid chromatography determination of fumonisin B1 and B2 in corn and corn products. J. AOAC lntemational 75:829-834. Shanna, V.D., Wilson, R.F., and Williams, LE. 1974. Reproductive performance of female swine fed corn naturally molded or inoculated with Fusarium roseum, Ohio isolates B and C. J. Anim. Sci. 38:598-602. Sheldon, J. L. 1904. A com mold (Fusarium moniliforme n. sp). Agric.Exp.Stn.Nebraska, 17th Ann.Rep. pp.23-32. Shephard, G.S., Sydenham, E.W., Thiel, P.G., and Gelderblom, WC. 1990. Quantitative determination of fumonisin B1 and BZ by high performance liquid chromatography with fluorescence detection. J. Liq. Chromatogr. 1 3:2077-2087. 185 Shotwell, O.L., Goulden, G.A., Bennett, G.A., Plattner, RD, and Hesseltine, CW. 1977. Survey of 1975 wheat and soybeans for aflatoxin, zearalenone, and ochratoxin. J. Assoc. Off. Anal. Chem. 60:778-783. Shotwell, O.L. and Hesseltine, CW. 1983. Five year study of mycotoxins in Virginia wheat and dent com. J. Assoc. Off. Anal. Chem. 66:1466-1469. Shreeve, B.J., Patterson, D.S.P., Roberts, BA, and Wrathall, A.E. 1978. Effect of moldy feed containing zearalenone on pregnant sows. Br. Vet. J. 134:421- 427. Smith, G.P. 1985. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science 228:1315-1317. Smith, GP. 1993. Surface display and peptide libraries. Gene 128:1-2. Smith, GP. and Scott, J.K. 1993. Libraries of peptides and proteins displayed on filamentous phage. Methods Enzymol. 217:228-257. Smith, J.W., Tachias, K., and Madison, EL. 1995. Protein loop grafting to construct a variant of tissue-type plasminogen activator that binds platelet integrin alpha llb beta 3. J. Biol. Chem. 270:30486-30490. Smith, MD. 1996. Antibody production in plants. Biotechnol.adv. 14:267-281. Smith, M.M., Shi, L., and Navre, M. 1995. Rapid identification of highly active and selective substrates for stromelysin and matrilysin using bacteriophage peptide display libraries. J. Biol. Chem. 270:6440-6449. Smith, T.K. 1982. Dietary influences on excretory pathways and tissue residues of zearalenone and zearalenols in the rat. Can. J. Physiol. Pharmacol. 60:1444-1449. Spurr, AR. 1969. A low-viscosity epoxy resin embedding medium for electron microscopy. J. Ultrastruct. Res. 26:31-43. Stahr, H.M., Ross, PF, and Obioha, W. 1981. Some mycotoxin levels in fann- stored corn. J. Agric. Food Chem. 29:207 Stieger, M., Neuhaus, G., Momma, T., Schell, J., and Kreuzaler, F. 1991. Self assembly of immunoglobulins in the cytoplasm of the alga Acetabulan’a meditenanea. Plant Sci. 73:181-190. Strader, C.D., Fong, T.M., Graziano, MP, and Tota, MR. 1995. The family of G- protein-coupled receptors. Faseb Journal 9:745-754. 186 Sturm, A., Kuik, A.V., Vliegenthart, J.F.G., and Crispeels, M.J. 1987. Structure, position, and biosynthesis of the high mannose and the complex oligosaccharide side chains of the bean storage protein phaseolin. J. Biol. Chem. 262:1 3392-13403. Sundlof, SF. and Strickland, C. 1986. Zearalenone and zeranol: Potential residue problems in livestock. Vet. Hum. Toxicol. 28:242-250. Sutton, J.C., Baliko, W., and Funnel, H.S. 1980. Relation of weather variables to incidence of zearalenone in corn in southern Ontario. Can. J. Plant Sci. 60:149 Swanson, S.P., Corley, R.A., White, D.G., and Buck, W.B. 1984. Rapid thin layer chromatographic method for determination of zearalenone and zearalenol in grains and animal feeds. J. Assoc. Off. Anal. Chem. 67:580-582. Sydenham, E.W., Thiel, P.G., Marasas, W.F., Shepard, G.S., Van Schalkwyk, D.J., and Koch, KR. 1990. Natural occurrences of some Fusarium mycotoxins in corn from low and high esophageal cancer prevalence areas of the Transkei. South Africa. J. Agri. Food Chem 38:1900-1903. Takkinen, K., Laukkanen, M.-L., Sizmann, D., Alfthan, K., lmmonen, T., Vanne, L., Kaartinen, M., Knowles, J.K.C., and Teeri, T.T. 1991. An active single-chain antibody containing a cellulase linker domain is secreted by Escherichia coli. Protein Eng. 4:837-841. Tavladoraki, P., Benvenuto, E., Trinca, S., De, M.D., Cattaneo, A., and Galeffi, P. 1993. Transgenic plants expressing a functional single-chain Fv antibody are specifically protected from vims attack. Nature 366:469-472. Tejada-Simon, M.V., Marovatsanga, L.T., and Pestka, J.J. 1995. Comparative detection of fumonisin by HPLC, ELISA, and immuno-cytochemical localization in Fusarium culture. J. Food Protect. 58:666-672. Tepfer, M. and Taylor, I.E.P. 1981 . The permeability of plant cell wall as measured by gel filtration chromatography. Science 213:761-763. Teshima, R., Kawase, M., Tanaka, T., Hirai, K., Sato, M., Sawada, J.-l., lkebuchi, H., lchinoe, M., and Terao, T. 1990. Production and characterization of a specific monoclonal antibody against mycotoxin zearalenone. J. Agric. Food Chem. 38:1618-1622. Thammana, P. 1994. Characterization of a deletion in the P3 myeloma heavy chain V region sequence in the non-secreting cell line NS1. Mol. lmmunol 31 :77- 78. . 187 Thiel, P.G., Shepard, G.S., Sydenham, E.W., Marasas, W.F., Nelson, PE, and Wilson, TM. 1991. Levels of fumonisin B, and B2 in feeds associated with confirmed cases of equine leukoencephalomalacia. J. Agri. Food Chem 39:109-1 11. Thompson, W.L. and Wannemacher-RW, J. 1986. Stnicture-function relationships of 12,13-epoxytrichothecene mycotoxins in cell culture: comparison to whole animal lethality. Toxicon 24:985-994. Thompson, J., Pope, T., Tung, J.S., Chan, C., Hollis, 6., Mark, G., and Johnson, KS. 1996. Affinity maturation of a high-affinity human monoclonal antibody against the third hypervariable loop of human immunodeficiency vims: use of phage display to improve affinity and broaden strain reactivity. J. Mol. Biol. 256:77-88. Thouvenot, DR. and Morfin, RF. 1979. Quantitation of zearalenone by gas-liquid chromatography on capillary glass columns. J. Chromatogr. 170:165-173. Thouvenot, DR. and Morfin, RF. 1983. Radioimmunoassay for zearalenone and zearalanol in human serum: production, properties, and use of porcine antibodies. Appl. Environ. Microbiol. 45:16-23. Tuite, J., Shaner, G., Rambo, 6., Foster, 6., and Caldwell, R.W. 1974. The Gibberella ear of epidemics of corn in Idiana in 1965 and 1972. Cereal Sci. Today 19:238-241. Turcatti, G., Zoffmann, S., Lowe, J.A., Drozda, S.E., Chassaing, G., Schwartz, T.W., and Chollet, A. 1997. Characterization of non-peptide antagonist and peptide agonist binding sites of the NK1 receptor with fluorescent ligands. J. Biol. Chem. 272:21167-21175. Turner, G.V., Phillips, T.D., Heidelbaugh, N.D., and Russell, L.H. 1983. High pressure liquid chromatographic determination of zearalenone in chicken tissues. J. Assoc. Off. Anal. Chem. 66:102-104. Ueno, Y., Ayaki, S., Sato, N., and Ito, T. 1977. Fate and mode of action of zearalenone. Ann. Nutr. Aliment. 31:935-948. Ueno, Y. 1983a. General toxicology. pp. 135-146. In Y. Ueno (ed.) , Developments in Food Science IV: Trichothecens, Chemical, Biological and Toxicological Aspects. Elsevier, Amsterdam, Netherlands. Ueno, Y. 1983b. DeveIOpments in Food Science lV: Tricothecenes: Chemical, Biological and Toxicological Aspects. Elsevier Science Publisher, Amsterdam, The Netheriands. 188 Ueno, Y. 1987. Trichothecenes in food. pp. 123-147. In P. Krogh (ed.) , Mycotoxins in Foods. Academic Press, New York, NY. Ueno, Y., Umemori, K., Niimi, E., Tanuma, S., Nagata, S., Sugamata, M., lhara, T., Sekijima, M., Kawai, K., Ueno, l., and et, a. 1995. Induction of apoptosis by T-2 toxin and other natural toxins in HL-60 human promyelotic leukemia cells. Nat. Toxins 3:129-137. Umberger, E.J. 1975. Products marketed to promote growth in food-producing animals: steroid and hormone products. Toxicol. 323-21. van Engelen, F.A., Schouten, A., Molthoff, J.W., Roosien, J., Salinas, J., Dirkse, W.G., Schots, A., Bakker, J., Gommers, F .J., Jongsma, M.A., Bosch, 0., and Stiekema, W.J. 1994. Coordinate expression of antibody subunit genes yields high levels of functional antibodies in roots of transgenic tobacco. Plant Mol. Biol. 26:1701-1710. Vesonder, R.F., Ciegler, A., Rogers, R.F., Burbridge, K.A., Borhast, R.J., and Jensen, AH. 1978. Survey of 1977 crop year preharvest corn for vomitoxin. Appl. Environ. Microbiol. 36:886-888. Vianello, A. and Macri, F. 1981 . Effect of zearalenone (F-2) on pea (Pisum sativum cultivar Alaska) stem, maize (Zea mays cultivar DeKalb XL 342) root and rat liver mitochondria. Planta 153:443-446. Voss, A., Niersbach, M., Hain, R., Hirsch, H.J., Liao, Y.C., Kreuzaler, F., and Fischer, R. 1995. Reduced virus infectivity in N. tabacum secreting a TMV- specific full-size antibody. Mol. Breeding 1:39-50. Voss, K.A., Chamberlain, W.J., Bacon, C.W., Herbert, R.A., Walters, DB, and Norred, WP 1995. Subchronic feeding study of the mycotoxin fumonisin B1 in B6C3F1 mice and Fischer 344 rats. Fundam. Appl. Toxicol. 24:102-110. Wade, W.S., Koh, J.S., Han, N., Hoekstra, D.M., and Lerner, RA. 1993. Engineering metal coordination sites into the antibody light chain. J. Am. Chem. Soc. 115:4449-4456. Wakulinski, W. 1989. Phytotoxicity of the secondary metabolites of fungi causing wheat head fusariosis (head blight). Acta Physiologiae Plantarum 11:301- 306. Wang, Y2. and Miller, JD. 1988. Effects of Fusarium gramineanrm metabolites on wheat tissue in relation to Fusarium head blight resistance. J. Phytopathol. 122:1 18-125. 189 Warner, R., Ram, B.P., Hart, LP, and Pestka, J.J. 1986. Screening for zearalenone in corn by competitive direct enzyme-linked immunosorbent assay. J. Agric. Food Chem. 34:714-717. Warner, R. and Pestka, J.J. 1987. ELISA survey of retail grain-based food products for zearalenone and aflatoxin B1. J. Food Protect. 50:502-503. Watson, S.A., Mirocha, C.J., and Hayes, A.W. 1984. Analysis for trichothecenes in samples from Southeast Asia associated with ”yellow rain“. Fundam. Appl. Toxicol. 42700-71 7. Weber, P.C., Pantoliano, MW, and Thompson, L.D. 1992. Crystal stmcture and ligand-binding studies of a screened peptide complexed with streptavidin. Biochemistry 31:9350—9354. Whitelam, 6.0. 1995. The production of recombinant proteins in plants. J. Sci. Food Agric. 6821-9. Whitelam, 6.0. and Cockbum, W. 1996. Antibody expression in transgenic plants. Trends in Plant Science 12268-272. Whitlow, M., Bell, B.A., Feng, S.L., Filpula, D., Hardman, K.D., Hubert, S.L., Rollence, M.L., Wood, J.F., Schott, M.E., Milenic, DE, and et, a. 1993. An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability. Protein Eng. 62989-995. Wilkinson, A.P., Ward, CM, and Morgan, M.R.A. 1992. Immunological analysis of mycotoxins. pp. 185-225. In H.F. Linskens and J.F.Jackson (eds.) , Modern Methods of Plant Analysis, New Series. Plant Toxin Analysis. Springer- Verlag, Berlin,Germany. Winter, 6., Griffiths, A.D., Hawkins, R.E., and Hoogenboom, HR. 1994. Making antibodies by phage display technology. Annu. Rev. Immunol. 12:433-455. Witt, MP and Pestka, J.J. 1990. Uptake of the naturally occurring 3-alpha-hydroxy isomer of T-2 toxin by a murine B cell hybridoma. Food Chem. Toxicol. 28:21-28. Wood, GE. 1992. Mycotoxins in foods and feeds in the United States. J. Anim. Sci. 7023941 -3949. Xiao, H., Clarke, J.R., Marquardt, RR, and Frohlich, AA 1995. Improved methods for conjugating selected mycotoxins to carrier proteins and dextran for immunoassays. J. Agric. Food Chem. 2092 190 Yang, W.P., Green, K., Pinz, S.S., Briones, A.T., Burton, DR, and Barbas, CF. 1995. CDR walking mutagenesis forthe affinity maturation of a potent human anti-HlV-1 antibody into the picomolar range. J. Mol. Biol. 2542392-403. Yuan, Q., Clarke, J.R., Zhou, H.R., Linz, J.E., Pestka, J.J., and Hart, LP. 1997. Molecular cloning, expression, and characterization of a functional single- chain Fv antibody to the mycotoxin zearalenone. Appl. Environ. Microbiol. 632263-269. Zambryski, P., Joos, H., Gentello, C., Leemans, J., Van Montagu, M., and Schell, J. 1983. Ti-plasmid vector for the introduction of DNA into plant cells without altering their normal regeneration capacity. EMBO J. 222143-2150. Zhou, H., Pestka, J.J., and Hart, LP. 1996. Molecular cloning and expression of recombinant antibody against Fumonisin B,. J. Food Protect. 5921208-1212. 191