55.5.3; ., 1!...» :l THESIS :lslllll lllllllllllllllllllllllllll‘lllH 3 1293 014 This is to certify that the dissertation entitled Novel Approaches for the Immunoassay of Fusarium Myootoxins presented by Sutikno has been accepted towards fulfillment of the requirements for Envimmnental Toxicology Dr. James J. Pestka Major professor July 18, 1995 MS U i: an Affirmative Action/Equal Opportunity Institution 0-12771 LIQRARY Michigan $tate Unlverstty PLACE N RETURN BOXto remove this checkout from your recent. TO AVOID FINES Mum on or More data duo. MSU I. An Affirmative ActkWEqual Opportunity lnstltulon W ”31.1 NOVEL APPROACHES FOR THE IMMUNOASSAY OF FUSARIUM MYCOTOXINS By Sufikno A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Food Science and Human Nutrition 1995 The fu metabolites p! are common!“ pmduCts. The dictated by em almost imposs Prevented thro WWW USI.’ chromatograpm immu"OSOfbem sensitive. In I; l applmllOn of al monodonal, ra:| were produced . .l as a ”Diem Q ABSTRACT NOVEL APPROACHES FOR THE IMMUNOASSAY OF FUSARIUM MYCOTOXINS By Sutikno The fumonisin and trichothecence mycotoxins are a group of secondary metabolites produced by toxigenic strains of Fusan'um fungi. These mycotoxins are commonly toxic to both human and animals and found in agricultural products. The presence of these toxins in agricultural commodities are primarily dictated by environmental and biologic factors. Since controlling these factors is almost impossible, mycotoxin entry into human and animal foods is typically prevented through detection and diversion. Although mycotoxin detection is performed using conventional methods such as high performance liquid chromatography (HPLC), immunoassay techniques such as enzyme-linked immunosorbent assay (ELISA) are inexpensive, simple, rapid, specific, and sensitive. In this thesis, several new approaches for the generation and application of antibodies to Fusarium mycotoxins were explored. Firstly, mouse monoclonal, rabbit and sheep polyclonal antibodies against fumonisin B1 (F81 ) were produced via a novel immunization procedure with keyhole limpet hemacyanin as a protein carrier. These antibodies were used to improve the sensitivity and malty of E senstivlly and s mnpettive die this ELISA pro HPLC. In addi- tat they are 5 Secondly. atte' deoxynlvalendl serum afbumlr‘, competitive ELIE DON MPG-trim W to the 835 ”bosom; : W. thes specificity of ELISAs that were previously developed in our laboratory. ELISA sensitivity and specificity were much higher when F81 -sheep antisera were used in competitive direct ELISA. FB1 detection in Fusan‘um culture, and corn products with this ELISA provided approximately two fold higher F81 estimates than that with HPLC. In addition, ELISAs had a strong positive relationship with HPLC. suggesting that they are suitable for fumonisins screening from human and animal foods. Secondly, attempts to generating high affinity and specificity antibodies against deoxynivalenol (DON) by immunizing mice and rabbits with a variety of DON-bovine serum albumin conjugates were made to improve sensitivity of previous DON competitive ELISA . High titer antisera were produced but they could not be used in DON competitive ELISAs. Finally, attempts were made to generate antibodies smcific to the hichothecene—yeast ribosomal binding site by immunizing mice with 808 ribosomal subunits. Although all animals exhibit high antiserum titers for the ribosomes, these antisera were not effective in a variety of ELISA configurations to measure “total load” trichothecenes in foods. To my parents and my grandparents for their moral support and prayer. To my wife Zuli and my daughter, Wulan, for their support, patience, kindness, and understanding. l am g advice. guide this dissertat: and Dr. L Pat graduate gun? I WiSh ti Olivera for the Dioduction of m SPeCial i daughter. Tlti understanding c Finally, almarltum, my ll‘sSplre me to University and , ACKNOWLEDGMENTS I am greatly grateful to my major advisor, Dr. James J. Pestka, for his advice, guidance, assistance, and understanding throughout the development of this dissertation. I am also indebted to Dr. John E. Linz, Dr. William Helferich, and Dr. L. Patrick Hart for their invaluable suggestion and being members of my graduate guidance committee. I wish to thanks to Dr. Mohamed M. Abouzied and Dr. Juan I. Azcona- Olivera for their invaluable suggestion and practical help especially during the production of monoclonal antibodies. Special acknowledgment goes to my wife, Siti Zulaikah Sutikno, and my daughter, Titiek Wulandari Sutikno, for their patience, supports, and understanding during the course of my graduate program. Finally, my appreciation extends to my late father, bapak Soewito almarhum, my grandmother, Eyang Musini, and my brothers and sisters who ‘ inspire me to study and work very hard during my stay in Michigan State University and to be a better Moeslim. LIST OF TAE LIST OF FIG INTRODUCI PART I. RE FUSA RIUL Introduc: Fumonisi Tn’chothe CONVENT Cleanup . Detection IMMUNOAS Antibody ‘ MYCOfoxm Applicator Commemt, PART II DEV F Uttt PRO; ABSTRACT BaCkgI’QUr. Analytical . “momsln alIOnale' TABLE OF CONTENTS LIST OF TABLES .............................................................................................. ix LIST OF FIGURES ............................................................................................ xi INTRODUCTION ................................................................................................. 1 PART I. REVIEW OF FUSARIUM-MYCOTOXIN IMMUNOASSAY ................... 4 FUSARIUM MYCOTOXINS ............................................................................. 5 Introduction ................................................................................................... 5 Fumonisins ................................................................................................... 9 Trichothecenes ........................................................................................... 14 CONVENTIONAL ANALYTICAL METHODS .................................................. 28 Cleanup of trichothecenes prior to analytical detection ................................ 28 Detection of trichothecenes ........................................................................ 30 IMMUNOASSAYS FOR FUSARIUM MYCOTOXINS ..................................... 38 Antibody production .................................................................................... 39 Mycotoxin immunoassay format .................................................................. 49 Application of mycotoxin immunoassays in foods ........................................ 54 Commercial mycotoxin immunoassay kits for foods .................................... 57 PART II DEVELOPMENT AND APPLICATION OF AN ELISA FOR FUMONISIN B1 IN FUNGAL CULTURES, CORN AND CORN PRODUCTS ....................................................................................... 61 ABSTRACT ................................................................................................... 62 INTRODUCTION ........................................................................................... 63 Background. ............................................................................................... 63 Analytical methods ...................................................................................... 66 Fumonisin antibodies .................................................................................. 68 Rationale. ................................................................................................... 69 vi MATERIAL Chemical Conluga‘. abbtlr Direct E“ Veratox‘: Com-$3 HPLC. Statistics RESULTS Rabbit p: Mouse lr Compaq [knecno Detecttc IMRSJK CONCLU: PARTIH r r ABSTR INTRQ{ Natur TOXK DON Rail: MATE] one Den Ck» Ih‘Ql Rat ELJ MATERIALS AND METHODS ........................................................................ 71 Chemical and reagents ............................................................................... 71 Conjugation of fumonisin 81 to protein ........................................................ 72 Rabbit immunization ................................................................................... 73 Hybridoma production ................................................................................. 74 Indirect ELISA ............................................................................................. 76 Direct ELISA ............................................................................................... 77 Veratox® .................................................................................................... 78 ELISAGRAM ............................................................................................... 79 Fusan'um cultures ....................................................................................... 8O Com-sample extraction and clean up .......................................................... 81 HPLC .......................................................................................................... 82 Statistics ..................................................................................................... 84 RESULTS AND DISCUSSION ....................................................................... 85 Rabbit polyclonal antibodies ....................................................................... 85 Mouse immunization and monoclonal antibodies ........................................ 86 Comparison among different antibodies ...................................................... 87 Detection of FBt-like compounds ................................................................ 89 Detection of F81 in Fusan'um cultures ......................................................... 90 Detection of F B1 in corn and corn product .................................................. 91 CONCLUSION ............................................................................................... 98 PART III PRODUCTION OF DEOXYNIVALENOL (VOMITOXIN) ANTIBODIES ................................................................................... 122 ABSTRACT ................................................................................................. 123 INTRODUCTION ......................................................................................... 124 Natural occurrence. .................................................................................. 124 Toxicity ..................................................................................................... 126 DON detection .......................................................................................... 129 Rationale .................................................................................................. 1 30 MATERIALS AND METHODS ....................................................................... 132 Chemical and reagents ............................................................................ '. 132 Derivation of DON ..................................................................................... 132 Conjugation of DON derivatives to protein .................................................. 134 Mouse immunization .................................................................................. 134 Rabbit immunization ................................................................................. 136 ELISA ....................................................................................................... 137 vii RESULTS DONder liouseir Rabbnir WSCUSSU Ethider flan) CONCLUS PARTIV. ABSTTLAC NTRODI Tmmm muso Ramm MATE R I} Cheflm Yamu Ffibosc T-Z t0): Mame Induec RESULT RIstC IHCEIr CONCLL APPENDI) APPENDlx APPENDI): RESULTS ............................................................................................. DON derivatization ...................................................................... I333 Mouse Immunization ............. 139 Rabbit immunization 14o DISCUSSION ............................................................................ 141 DON denvatlzatlon '''''''''' 141 Antibody production 141 CONCLUSION .............................................................................................. 143 PART IV. PRODUCTION OF ANTIBODIES AGAINST TRICHOTHECENE- YEAST RIBOSOMAL BINDING SITE ........................................... 148 ABSTRACT ...................................................... 149 INTRODUCTION ......................................................................................... 1 50 Trichothecene mycotoxins ........................................................................ 150 Ribosomal binding as a toxicity mechanism ................................................. 151 Rationale and objective .............................................................................. 154 MATERIALS AND METHODS ...................................................................... 156 Chemicals and reagents ............................................................................. 156 Yeast production ........................................................................................ 157 Ribosome preparation ................................................................................ 157 T-2 toxin-ribosome binding assay ............................................................... 159 Mouse immunization .................................................................................. 160 Indirect ELISA ........................................................................................... 161 RESULTS AND DISCUSSION ..................................................................... 163 Ribosome preparation ............................................................................... 163 Mice immunization ..................................................................................... 164 CONCLUSION .............................................................................................. 167 APPENDIX A: MEDIA FOR HYBRIDOMA PRODUCING MONOCLONAL ANTIBODIES .......................................................................... 172 APPENDIX B: SAMPLE LISTS OF CORN AND CORN PRODUCTS ............... 175 APPENDIX C: IMMUNOLOGICAL ASSAYS FOR MY COTOXIN DETECTION . 179 LIST OF REFERENCES ........................................................................................ 188 viii “av-fl W". I Table 1.1. Table 1.2. Table 1.3. h Table 1.4. Table 1.5. Table 1.6. Table 1.7_ Table 2.1. Table 2.2. Table 1.1. Table 1.2. Table 1.3. Table 1.4. Table 1.5. Table 1.6. Table 1.7. Table 2.1. Table 2.2. Table 2.3. Table 2.4. Table 2.5. LIST OF TABLES Types Of economic losses and costs associated with mycotoxin contamination in foods and feeds (CAST, 1989) ................................ 8 Natural occurrence of fumonisins in cereals and cereal products 15 Natural occurrence of trichothecene deoxynivalenol (DON) and nivalenol (NIV) in cereals and cereal products ................................ 22 Summary of trichothecene-induced toxicoses ................................. 26 Retention factor (Rf) values of some trichothecene mycotoxins on TLC plates using silica gel as absorbent (adapted from Ueno, 1980) .................................................................................... 32 Selected Fusarium mycotoxin immunoassays in foods .................... 55 Commercial Immunoassay Kits for Mycotoxins available as of June 1993 (Pestka et al., 1995) ...................................................... 58 Sensitivity of Cl-and CD-ELISAS for F81 using monoclonal antibodies prepared from ascites fluid ........................................... 106 Comparison of competitive F81 ELISA using mouse-monoclonal antibodies prepared from ascites fluid, rabbit-and sheep- polyclonal antibodies ..................................................................... 1 10 Cross reactivity of FB1 sheep antisera toward fumonisin analogues. .................................................................................... 112 Comparison of FBt concentrations in Fusarium corn cultures detected by HPLC and CD-ELISA methods ................................. 115 Comparison of FBI recoveries from spiked-com samples containing FB1 of 100 to 3,000 uglg using raw methanolic extracts and SAX-cleaned extracts as determined by HPLC, Veratox® ', and CD-ELISA using F81 Sheep antisera (Neogen ’ Corp., Lansing, MI) ....................................................................... 116 ix Table 2.6. 031.0 . Table 2.7. .‘T m A Table 2.8. I0 0 Q3. Table 3.1. 81 Table 5.1. Table 5.2_ h Table 5_ 3_ '7 Table 2.6. Comparisons of F 81 concentrations in 14 com-food samples by HPLC, Veratox® ', and CD-ELISA using FBt sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox® ' and CD-ELISA to HPLC results ........................................................... 117 Table 2.7. Comparisons of F81 concentrations in 28 Italian feed samples by HPLC, Veratox® ', and CD-ELISA using F81 sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox® " and CD-ELISA to HPLC results ........................................................... 118 Table 2.8. Comparisons of F B1 concentrations in fresh com 3 samples by HPLC, Veratox® '. and CD-ELISA using F81 sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox® b and CD-ELISA to HPLC results ........................................................... 120 Table 3.1. Structures of deoxynivalenol (DON) analogs (Adapted from Ueno, 1983) .................................................................................. 125 Table 5.1. Number and description of food samples bought in retail supermarket in Mid Michigan, in September, 1991. ....................... 175 Table 5.2. Number and description of Italian feed samples. ........................... 176 Table 5. 3. Number and description of fresh corn sample harvested from five counties in Michigan in Summer, 1994' .................................. 177 Figure 1.1. ‘ Figure 1.2 Figure 1 3 Figure 14 Figure 1_ Figure 1 5 “WE 1.7. l I 5 LIST OF FIGURES Figure 1.1. Factors affecting occurrence of mycotoxins in the human food Chain (Pestka and Casale, 1990) ...................................................... 6 Figure 1.2. Structures of known fumonisins and of Alteman’a altemata f. sp. chopersici (AAL) toxin, showing structural similarities with Sphingosine (Norred, 1993) ............................................................. 11 Figure 1.3. Structure and numbering system of some naturally identified trichothecene mycotoxins (modified from Ueno, 1983). ................... 18 Figure 1.4. Classification of trichothecene mycotoxins based on their Chemical structure (modified from Ueno, 1980). .............................. 19 Figure 1.5. Preparation of immunogen for (left) fumonisins and (right) deoxynivalenol (DON) (Pestka et al., 1995) .................................... 43 Figure 1.6. Process of antibody production in vivo (left) and using gene technology (right). Step 1: rearrangement or assembly of germ line V-genes; step 2: surface displaying of antibody; step 3: antigen—driven or affinity selection; step 4: affinity maturation; step 5: production of soluble antibody or antibody fragment (Adapted from Vlfinter et al., 1994) ................................................................... 48 Figure 1.7. Competitive ELISA for mycotoxins (adapted from Pestka, 1988). In direct ELISA, enzyme-labeled mycotoxins (ENZ) are simultaneously incubated with free mycotoxins (D)over solid- phase bound antibodies (AB). Concentration of free mycotoxins is inversely related to bound enzyme-labeled mycotoxins and can be measured quantitatively using spectrophotometer after color development by the addition of enzyme substrate. In indirect competitive ELISA, mycotoxin specific antibodies (AB) compete with free mycotoxins (CI) to solid-phase mycotoxin- carrier protein (CP) conjugate. Second antibodies (ab-ENZ) which have been labeled with enzyme are then added to determine total antibody bound. Concentration of free toxin is inversely related to bound enzyme-labeled antibodies. ................... 51 Figure 1.8. ELISAGRAM procedure for mycotoxins (Pestka, 1991). ................. 52 xi Figure Zl Figure 2 2 Figure 2 3 Figure 274 Figure 2 Figure Figure 2.1. ELISA titration of rabbit polyclonal FBI antibodies. Sera were obtained two weeks after the second injection with FBt-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate indirect ELISA. Open symbols are direct ELISA. R1, R2 and R3 refer to rabbit number. The letter i indicates immunized and p indicates preimmun ........................................................................................ 99 Figure 2.2. Competitive inhibition ELISA for FB1 using different rabbit polyclonal antibodies. Sera were obtained two weeks after the second injection with FBt-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate CI-ELISA. Open symbols are CD-ELISA. R1, R2 and R3 refer to rabbit number. .................................................. 100 Figure 2.3. Competitive inhibition ELISA for F81 using mouse sera. Sera were obtained ten days after the third subcutaneous (SC) injection with FBt-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate CI-ELISA. Open symbols are CD-ELISA. Subscript 1.4 refers to mouse number. ............................................................... 101 Figure 2.4. Competitive inhibition ELISA for FB1 using mouse sera. Sera were obtained ten days after the third intraperitoneal (IP) injection with FBI-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate CI-ELISA. Open symbols are CD-ELISA. Subscript 1-4 refers to mouse number. ............................................................... 102 Figure 2.5. Competitive indirect ELISA curves for FB1 using hybridoma supematants. Supematants were obtained from hybridoma cells producing FBI antibodies before cloning. Each data point represents the mean value of duplicate measurements. ............... 103 Figure 2.6. ELISA titration of monoclonal F81 antibodies prepared from ascites fluid. Each data point represents the mean value of duplicate measurements. Filled symbols indicate Cl-ELISA. Open symbols are CD-ELISA. 0105 clone had no titer when determined with CD-ELISA. .......................................................... 104 Figure 2.7. Competitive inhibition ELISA for F81 using monoclonal antibodies prepared from ascitic fluid. Data points represent the mean value of duplicate measurements. Filled symbols were CI- ELISA. Open symbols were CD-ELISA. Q1CS clone had no titer when determined with CD—ELISA. .......................................... 105 xii Figure 2': I Figure 2 E Figure 2.1 .' FIQUTe 2 Figure 2.8. Competitive direct ELISA curves for FB1 using FBt sheep anti sera (Neogen Corp., Lansing, MI). F8, was dissolved in extractant [10% methanol (vol/vol.) in distilled water], fresh corn extract numbers 37 and 56 at a dilution of 1:35). Each data point represents the mean value of triplicate measurements ......... 107 Figure 2.9. Competitive direct ELISA curves for F81 using F 8, sheep anti sera (Neogen Corp., Lansing, MI). FBI was dissolved in extractant [10% methanol (vol/vol.) in distilled water], fresh corn extract numbers 37 and 56 at a dilution of 1:200). Each data point represents the mean value of triplicate measurements. ............................................................................. 108 Figure 2.10. Comparison of competitive inhibition ELISA for F81 mouse- monoclonal antibodies, rabbit-and sheep-polyclonal antibodies, and Veratox". Monoclonal antibodies (N205 MAB and 205 MAB) were prepared from ascitic fluids. N205 MAB was generated in the current study using F BLKLH immunogen, while 205 was produced by Azcona-Olivera (1992a) using F81- Cholera toxin immunogen. R3 refers to rabbit number 3. Sheep antiserum was from a single lot supplied by Neogen Corp. (Lansing, MI). Veratox’ is an ELISA kit for FB1 based on CD- ELISA and is produced and supplied by Neogen Corp. (Lansing, MI). Monoclonal antibodies were determined with Cl- ELISA, but polyclonal antibodies were measured with CD- ELISA. Each data point represents the mean t standard errors of the mean (n = 4, two duplicate measurements). Absorbencies at 0 nglml F81 ranged from 0.675 to 1.250 ............. 109 Figure 2.11. Competitive direct ELISA standard curves for F3. , F82 and F83 , using F81 sheep antisera. Sheep antiserum was from Single lot supplied by Neogen Corp. (Lansing, MI). Each data point represents the average value of triplicate determination ....... 111 Figure 2.12. A typical “broad range “ standard curve used for HPLC analyses of Fusarium corn cultures. Each data point represents the average value of two determinations ....................................... 113 Figure 2.13. A typical “low range “standard curve used for HPLC analyses of corn and corn products. Each data point represents the average value of two determinations ............................................. 114 xiii FgueB‘ Fuwe32 Fgweafl Flgure 3 FlgUre 4 Figure 3.1. Indirect ELISA titration of mouse antibodies obtained ten days after the third injection with BSA-HS-DON conjugate. The first injection was given at a dose of 50 and 100 uglmouse. The second and third injections were given at a dose of 25 and 50 jug/mouse. Each data point represents the mean :t standard error of the mean (n = 6, duplicate measurements from three mice). lP indicates peritoneal injection. SC indicate subcutaneous injection. 50 and 100 indicate dose (ug) of the first injection. ................................................................................ 144 Figure 3.2. Indirect ELISA titration of mouse antibodies obtained ten days after the second (SI) and third immunizations (T I) with BSA-HS- DON conjugate. The first immunization was given via intrasplenic deposition at a dose of 20 uglmouse. The second and third immunization were given via intraperitoneal (IP) and subcutaneous (SC) injections at a dose of 10 and 50 ug, respectively. Each data point represents the mean :I: standard error of the mean (n = 8, duplicate measurements from four mice). . .......................................................................................... 145 Figure 3.3. Direct ELISA titration of rabbit antisera. Hunter's adjuvant indicates that rabbits were given a two Sites intramuscular injection with an emulsion of Hunter Titer Max and BSA-HS- DON conjugate at a dose of 100 pg per rabbit for both the first and second injections. Freund’s adjuvant indicates that rabbits were given a ten site subcutaneous injection with an emulsion of Freund’s adjuvant and BSA-HS-DON conjugate at a dose of 500 ug and 250 pg per animal for the first and second immunization, respectively. Antisera were obtained four weeks after the second injection with BSA-HS-DON conjugate. Each data point represents the mean 1 standard error of the mean (n = 6, duplicate measurements from three rabbits). ......................... 146 Figure 3.4. Types of protein-deoxynivalenol conjugation. ................................ 147 Figure 4.1. Competitive indirect ELISA for “trichothecene load” using yeast ribosomes for coating microtiter plates. Trichothecene ribosomal binding site specific antibodies (AB) compete with trichothecenes (T) for the trichothecene binding Site (TBS). Second antibodies (SAB) which have been labeled with enzyme (ENZ) are then added to determined total bound antibody (AB). “Trichothecene load” is inversely related to the bound enzyme- Iabeled antibodies and can be measured quantitatively using spectrophotometer after color development by the addition of enzyme substrate. ........................................................................ 168 xiv Flt Figure 4.2. Inhibition of T-2 toxin to the association of 3H-T-2 toxin to yeast ribosomes. Each data point represents the mean value of triplicate measurements. One hundred ul of yeast ribosomes (0.4 mglml standard buffer) were reacted with 10 pl of 3H-T-z toxin (0.2 uCiIml) in the presence of 10 ul of serially diluted (0 to 1000 ng) non radiolabeled T-2 toxin and 60 ul of precooled alcohol. As a control the same amount of radiolabeled toxin was mixed with the standard buffer without ribosomes. After ribosome-bound 3H-T-2 toxin was separated by centrifugation, radioactivity (DPM) of 100 pl of the supernatant was measured by liquid scintillation counting. DPM values of control solution and 0 nglml supernatant were 16,026 and 8,577, respectively. Percent inhibition = [(DPM value of certain ng T-2 toxin lml supernatant - DPM value of 0 nglml supematant)l(DPM value of control solution - DPM value of 0 nglml supematant)] x 100%. ......... 169 Figure 4.3. Typical indirect ELISA titration of mouse antisera. The mouse antisera were obtained ten days after the third subcutaneous or intraperitoneal injection with song of 608 or 808 yeast ribosomal subunit emulsified with F reund’s adjuvant. Each data point represents the mean :I: standard error of the mean (n=6, duplicate measurements of three mice). IP, and SC indicate intraperitoneal and subcutaneous injections, respectively. 803 and 608 indicates 808 and 608 yeast ribosomal subunits, respectively. ................................................................................. 170 Figure 4.4. Typical indirect ELISA titration of mouse antisera. The mouse antisera were obtained ten days after the third subcutaneous injection with song of 808 yeast ribosomal subunit mixed with different adjuvants. Each data points represents the mean a: standard error of the mean (n=4, duplicate measurements of two mice). 808 indicates 80$ yeast ribosomal subunit. FA, CT, and T-2 indicate Freund’s adjuvant, Cholera toxin, and T-2 toxin, respectively. .......... . ............................................................. 171 P'9 303: deter chror C‘iron' tech/tit commc applical H(Never CUItures' , HPLC me INTRODUCTION Mycotoxins are a group of secondary metabolites produced by toxigenic strains of fungi. One fungal genus that contains Species capable of mycotoxin production is Fusarium. Among the Fusarium mycotoxins that are toxic to both human and animals and commonly found in grain and grain products are fumonisins and trichothecenes. The presence of these toxins in agricultural commodities are primarily dictated by environmental and biologic factors. Since controlling both environmental and biological factors is almost impossible, prevention of mycotoxin entries into human foods and animal feeds is commonly accomplished via detection of the toxins in agricultural commodities. Mycotoxin detection can be carried out using either conventional methods such as thin layer Chromatography (TLC), gas chromatography (GC), high performance liquid Chromatography (HPLC), and mass spectroscopy (MS) or immunoassay techniques such as enzyme-linked immunosorbent assay (ELISA). ELISAS are commonly preferred to conventional methods because of simplicity, rapidity, and applicability both in fields and laboratories. ELISAS for fumonisin 81 (FB1) have been developed in our laboratory. However, when these ELISA systems are used to detect F31 in fusarium corn cultures, corn and corn products, they provide much higher F81 estimates than HPLC methods. One possible explanation for these observations is that F 81 tl‘BTG' and c t“. r anti" tncbot corn-b detecti ELISA higher. becaust compett toxicity. investigg agncuhu TI rationale: FUSan'um ELISA f0: (Fromm-C aQainst Tr mICOIOXIn: 2 antibodies used in these ELISAS are not completely specific to fumonisins and, therefore, may have reacted with other compounds found in Fusarium cultures, and contaminated corn. To overcome this problem, it is desirable to produce antibodies which have higher affinity and specificity to the toxins. Our laboratory has also developed an ELISA for deoxynivalenol trichothecene. When this ELISA was used to detect deoxynivalenol (DON) in com-based foods, it had a relatively high detection limit (1000 ppb). A lower detection limit of an ELISA can be generated when DON antibodies used in the ELISA have a higher affinity and specificity to DON. Therefore, production of higher affinity and specificity antibodies against DON is desirable. Trichothecenes are a potent inhibitor of eukaryotic protein synthesis because they bind to a common site on the 603 ribosomal subunit. These toxins compete with each others for the ribosomal binding site in proportion to their toxicity. Production of antibodies Specific to the binding site would enable investigators to develop an ELISA that can assess total ”trichothecene load” in agricultural commodities and, thus enhance food safety. The research in this dissertation was undertaken to address all above rationales and this dissertation was divided into four parts: Part I (Review of Fusarium Mycotoxin Immunoassay); Part II (Development and Application of an ELISA for Fumonisin B1 in Fungal Cultures, Corn and Corn Products); Part III (Production of Deoxynivalenol Antibodies); and Part IV (Production of Antibodies against Trichothecene Yeast Ribosomal Binding Site). Part I reviews Fusarium mycotoxins especially fumonisins and trichothecenes including their chemical structures in detail ’5 develop" com Cult; antibodre respect“ 3 structures, toxicity, natural occurrence, and detection methods. Part II describes in detail: the production of F B1 antibodies, the use of the antibodies for ELISA development, and the application of the ELISA for detection of F81 in Fusarium corn cultures, corn and com product. Part III and IV explain efforts to produce antibodies against deoxynivalenol and trichothecene yeast ribosomal binding Site, respectively. PART I. REVIEW OF FUSARIUM-MYCOTOXIN IMMUNOASSAY Introductir My: part oi se contamin; and Mos: products Ne l0 comm: environmt usual/y 0c favor certa.i 1983'. Shob environment; The human foc MIcolo lllllllan and fa lldude: (1) “II/30mm, (2 litterial or _ FUSARIUM MYCOTOXINS Introduction Mycotoxins are a very diverse group of toxic compounds produced as a part of secondary metabolism in a wide variety of filamentous fungi which often contaminate agricultural commodities prior to harvest and during storage (Smith and Moss, 1985). Peanut, corn, feeds, wheat and cereals are five agricultural products which most often have mycotoxin problems (Hesseltine, 1986). Natural occurrence of mycotoxin in foods and feeds varies from commodity to mmmodity, year to year and region to region and are strongly dictated by environmental factors (CAST, 1989). Trichothecene mycotoxin contamination usually occurs during cold and wet season, or in extreme drought years which favor certain fungus infections and toxin production (Vesonder et al., 1978; Ueno, 1983; Shotwell et al., 1985; Tanaka et al., 1988a; CAST, 1989). Effects of environmental conditions and genetic factors on the occurrence of mycotoxins in the human food Chain are illustrated in Figure 1.1. Mycotoxin-contaminated products can produce adverse effects on both human and farm animals when the products are consumed. The effects may include: (1) acute toxicity and death following high level exposures of a mycotoxin, (2) lower growth rate, impaired immunity, greater susceptibility to bacterial or parasitic infection, (3) decreased milk and egg production, . HI & #\ era/22% OOH! Biological Environmental Harvesting Factors Factors Susceptibile Temperature Crop Maturity Crop + ' Moisture ' Temperature Compatible. Mechanical Injury Moisture _ Toxigenic Insect/Bird Damage Detection/Diversion Fungus Fungus Storage Temperature Moisture ‘ Detection/Diversion Distribution- Processing “ Detection/Diversion Humans < . Animals Animal Products Figure 1.1. Factors affecting occurrence of mycotoxin in the human food Chain (Pestka and Casale, 1990) (4) reduced I location eta 1989). Con: livestock prod The e livestock log heath lCAS‘ human food Cosmetic A; established m leVel: (CAST~198 diatoms, . melamine C bwnmsa all? (300 p bleeding cat-ill AmOng zearalenOne‘ a first ”7’98 mic; especially mm 7 (4) reduced reproductive efficiency, or (5) Chronic toxicity such as cancer formation after prolonged exposure to small quantities of the mycotoxins (CAST. 1989). Consequently, mycotoxins cause economic losses to farmers and livestock producers (Hesseltine, 1986). The economic losses (Table 1.1) are derived not only from crop and livestock losses, but also from regulatory action to protect human and animal health (CAST, 1989). In the United States, for example, the amount of aflatoxins in human foods and animal feeds has been regulated. Under the Food, Drug, and Cosmetic Act, Section 402(a)(1), the US. Food and Drug Administration (FDA) has established acceptable aflatoxin levels in agricultural commodities by establishing action levels that allow for the removal of violative lots from interstate commerce (CAST, 1989). The action levels of human foods are 20 part per billion (ppb) total aflatoxins, except for milk which has an action level of 0.5 ppb for aflatoxin M (a metabolite of aflatoxin 81). The action level of aflatoxins for feeds is 20 ppb, except for cottonseed meal used in feeds (300 ppb), corn used for finishing (feedlot) beef cattle (300 ppb), com destined for finishing swine (200 ppb), and feeds used for breeding cattle, breading swine, and mature poultry (100 ppb)(CAST, 1989). Among the most important mycotoxins are fumonisins, trichothecenes, zearalenone, aflatoxins, and ochratoxins (Hesseltine, 1986; Pohland, 1993). The first three mycotoxins are produced by Fusan‘um spp. Fusarium mycotoxins especially fumonisins and trichothecenes will be reviewed in this chapter. Bearer ____________.__ Farmers a“: stool orOC-JC Fooc am ‘e rartdle’s :1 at prices Sevemmer 8 Table 1.1. Types of economic losses and costs associated with mycotoxin contamination in foods and feeds (CAST, 1989) Bearer Economic losses and costs Farmers and live Stock producers Food and feed handlers, distributor and processors Government Consumers (human or animal) - Outright food and feed loss. - Contaminated crops provide less income and may lead to potential losses of outlet. - Reduced productivity of livestock from (1) lower quantity and quality of animal products, (2) smaller letters, (3) reduced work output, (4) loss of pregnancy, (5) reduced feed efficiency, (6) impaired resistance to disease, and (7) loss of vaccination efficacy. - Less income from products refused, condemned, or sold at discount. — Increased storage, transport, and packing costs on such products. - Potential loss of market, trading reputation, and raw material source. - Increased costs due to litigation (may exceed cost of product), surveillance, and control. - Lower foreign exchange earnings from reduced exports. - Increased cost involved in shipment, sampling, and analyses of exported goods that are subsequently refused import entry; potential loss of overseas outlets. - Increased costs of detoxification or reconditioning abroad. - Increase costs for food or feed imports; staple food subsidies. - Increased costs of surveillance and mntrol. - Increased costs for expenditures on human and animal health facilities and activities. - Increased costs involved in training and extension programs. - Consumption may lead to impaired health and productive capacity. - Lack of food may lead to undemutrition or higher food prices resulting from outside purchase of foods or feeds. - Possible medical and veterinary costs associated with the above conditions in previous two statements. - Possible consumer-initiated litigation costs. Furllonisins Histo metabtllites 19881. and‘ lound on 0 lila'asas 8' Sheldon (1! agent of ‘rr mold hasb equine le; reurotoxioc matter of r Slurptoms tausatrve a {Benzuidenl in fumonisrn. dIStnbUIIOII, p FBI is ,- Mon/l/fome (G agent gi (am a WWW/”Toni Ill 93/69/5194) 1, Fumonisins Historical background. Fumonisins are a group of secondary metabolites produced by Fusarium moniliforme (Sheldon)(Benzuidenhout et al., 1988), and Fusarium proliferatum (Ross et al., 1990). These fungi are commonly found on corn, sorghum, and other grain commodities throughout the world (Marasas et al., 1984a). Fusarium monilifonne, which was firstly described by Sheldon (1904) as Fusarium monilifonne Sheldon, was implicated as a causative agent of “moldy corn poisoning” in animals in the United State (Peter, 1904). This mold has been associated with poisoning in equine Species that is now known as equine leukoencephalomalacia (ELEM) (Ross, 1994). This disease is a neurotoxicosis that is 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 81 (F B1 )] 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). F81 is the most toxic and predominant fumonisin produced by Fusarium moniliforme (Gelderblom et al., 1988b), and has been confirmed as an etiologic agent of fatal animal diseases such as ELEM in horses (Kellerman et al., 1990), porcine pulmonary edema (PPE) in pigs (Harrison et al., 1990), and liver cancer in rats (Gelderblom et al., 1991). In addition, epidemiological studies indicate that fumonisins are associated with human esophageal cancer in South Africa (Marasas et 5 given in Part I Chem add and ei‘. pentahydrox Chemical st troopers/cl studies. to were isoia‘ Marasas ( al., 1991: been rept whereas t a methyl I fresco r. Madman The 0th been p, 1994) SDI/d SI 10 (Marasas et al., 1988a). A detailed review of fumonisin toxicity and detection is given in Part II. 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 dery analogue (Bezuidenhout et al., 1988). Chemical structures of these toxins are similar to that of Alteman'a altemata f. Sp. chopersici (AAL) toxin and sphingosine (Figure 1.2)(Norred, 1993). In early studies, four fumonisins (FA1, FA2, F81 and FB2 ) (Bezuidenhout et al., 1988) were isolated and identified by the South African researchers led by Dr. W.F.O. Marasas (Norred, 1993). Additional chemical structures for F83, FB4 (Cawood et al., 1991; Plattner et al., 1992) and F01 (Branham and Plattner, 1993) have now been reported. The A-series fumonisin are acetylated at the amino group whereas the B-fumonisins have a free amine (Figure 1.2). F01 is F B1 which lost a methyl group at C-1 position; thus F01 is a diester of 13, 14-propane-1,2,3- tricarboxylic acid and 1-amino-11, 15-dimethyl-2, 4, 9, 13, 14-pentahydroxy— nonadecane (Branham and Plattner, 1993). The biosynthetic pathways through which fungi produce fumonisins have only been partially elucidated (Norred, 1993). When Fusarium spp. are cultured on solid (corn) substrates, FB1 is produced predominantly by Fusan'um moniliforme strains, but some strains of Fusarium proliferatum produce FB2 or F33 at higher concentrations than F81 (Nelson et al., 1994; Visconti and Doko, 1994). Solid substrate media are not conductive for the study of fumonisin FULIC SPI “the 1.2 11 FUMONISINS (itOOH COCH,CHCH,COOI~I \ R. R; R. I . . FA. OH OH CH,CO “- “ or, H OH Waco ”ONFHEMUTI“.OCTI' I ,, NHR, Pei. OH OH H “04'? CH’ 8‘ e. t. a. H mmcwr ‘ H FB. .OH H H FB. I-I - H H ('ZOOH COOLOIOLCOOII ' ' on OH AAL TOXIN W C“. CH CH» 0H NH. OH SPHINGOSINE /\/\/\/\/\/\/W :.' " . ' NH; FIgure 1.2. Structures of known fumonisins and of Alteman‘a altemata f. sp. chopen’sici (ALL) toxin, showing structural similarities with spingosine (Norred, 1993). A libs Cultu the m Sin/tar tidings shines; 581/76, [3 12 biosynthesis, and production of fumonisins in liquid media must be performed (Plattner and Shackelford, 1992; Blackwell et al., 1994). When Plattner and Shackelford (1992) fed deuterium-labeled methionine to Fusarium monilifonne cultures, they observed high levels of deuterium incorporation into F81 at the methyl groups on the C-12 and C-16 position of the fumonisin backbone. Since the'structure of fumonisins is Similar to that of sphingosine, these investigators speculated that biosynthesis of a fumonisin backbone is also Similar to that of sphingosine. Sphingosine is synthesized through condensation of linolyl- coenzyme A and alanin, whereas the fumonisin backbone is synthesized via condensation of palmitoyl coenzyme A and serine (Plattner and Shackelford, 1992) However, the above hypothesis was contested by Blackwell et al. (1994) who fed 13C-Iabeled acetate to liquid cultures of Fusarium moniliforme to determine the location of labeled carbon atoms in the radiolabeled fumonisin. They observed that the methyl group (C-2) of acetate resulted in enrichment of C- 20, 18, 16, 14, 12, 10, 8, 6, and 4 while the carbonyl group (C-1) of acetate labeled C-19, 17, 15, 13, 11, 9, 7, 5, and 3. When they added methionine to the cultures, F81 production increases up to 12-fold, with the S-methyl group from the methionine exclusively enriching positions C-21 and C22. Those resutls were Similar to the results of Plattner and Shackelford (1992). Based on these findings, Blackwell et al. (1994) concluded that the backbone of fumonisins was synthesized by the fungus through condensation of acetyl coenzyme A and serine, rather than modification of palmitic acid as stated by Plattner and reo an 1 FBI non/i ion it than t rarities 11051 I lie 13 Shackelford (1992). More work is still required to fully elaborate the biosynthetic pathway of the fumonisins. Natural occurrence. Fusarium monilifomIe , the fumonisin-producing fungus, commonly occurs in the world and it has been isolated from many countries including Australia, Brazil, Canada, Central America, China, Croatia, Egypt, German, Hong Kong, India, Indonesia, Israel, Italy, Jamaica, Japan, Nepal, New Zealand, Peru, Philippines, Poland, Portugal, Romania, South Africa, Taiwan, the United State, Turkey and Zambia (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 oat and on other agricultural products such as beans, peanuts, sugar beats, and bananas (Bacon and Nelson, 1994). Natural occurrence of fumonisins has been documented in a number of countries (Table 1.2). 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, a 32% moisture content of com media, and an aerated atmosphere (cotton-stoppered flasks) were the optimum condition of F Bi production by Fusarium monilifomie in laboratory experiments. Fusarium monilifomie that was isolated from European fresh corn produced FB1 up to 300 ppm when cultured on solid corn media at the optimum conditions for 12 days (Bars et al., 1994). Other Fusarium moniliforme isolates produce higher quantities (6400 ppm) of FB1 when cultured on the same media (Nelson et al., 1991). More higher F 81 (17,000 ppm) was produced when Fusarium moniliforme f05’t’t lnterr 0f tor. ttreat 14 MRC 826 was cultured on corn media at 20°C for 13 weeks (Alberts, et al., 1990) Trichothecenes Historical background. Trichothecenes are a group of structurally similar sesquiterpenoid metabolites produced mainly by Fusarium spp. (Bamburg and strong, 1971; Bamburg, 1983; Ueno, 1983; Betina, 1989). The first known member of this group, trichothecin, was originally dismvered as an antifungal antibiotic in 1948 (Freeman and Morrison, 1948; 1949). Since that time over eighty trichothecenes have been isolated and characterized (Betina, 1989). Interest in trichothecenes has increased over the years since their first discovery. The former Soviet Union was the first country to conduct an extensive research on these toxins in attempts to identify and elucidate the causative agents of mycotoxicoses in the 1930’s following outbreaks of alimentary toxic aleukia (ATA) and stachybotriotoxicoses (Ueno, 1980; Bamburg, 1983). Japan followed shortly thereafter because this country also suffered from a trichothecene-mediated disease known as red-mold (akakabi in Japan) disease. Interest in mycotoxins in the Western worId did not emerge until the appearance of “turkey x disease”, which killed thousands of turkeys in the United Kingdom in the early 19605 (CAST, 1989). The etiologic agent of the “turkey x disease” was feed contaminated by Aspergillus flavus which produced aflatoxins (Spensley, r963) ‘1: IV Table 1.2. Natural occurrence of fumonisins in cereals and cereal products CROP GRAIN MEAN IN POSITIVES, PPM COUNTRY YEAR PRODUCTS (POSITIVES/SAMPLES) REFERENCE F B, FB2 _A_rgentina 1991 com 2.88 (17/17) 1.14 (17/17) Sydenham et al., 1993 Canada 1990 cornmeal 0.05(1/2) nd (0/2) Sydenham et al., 1991 China: Linxian 1989 corn 0.87 (13/27) 0.45 (3/27) Yoshizawa et al., 1994 Cixian 1991 moldy corn 93.13 (4/4) - Chu and Li, 1994 Shaggqui 1989 corn 0.89 (5/20) 0.33 (1/20) Yoshizawa et al., 1994 Shangqui 1991 moldy corn 5500(5/5) - Chu and Li, 1994 Croatia 1992 corn 0.02 (11/19) 0.01 (4/19) Doko et al., 1995 mi 1990 cornmeal 2.38 (2/2) 0.60 (ZIZ) Sydenham et al., 1991 Italy 1989- corn 0.38 (26/26) 0.14 (13l26) Doko et al., 1995 1991 1991 corn feeds 1.14 (23/25) 0.30 (13/25) Minervini et al., 1992 Peru 1990 cornmeal 0.66 (1/2) 0.14 (1/2) Sydenham etal.,1991 Poland 1992 com 0.02 (2/7) 0.01 (1/7) Doko et al., 1995 Portugal 1992 corn 1.03 (9/9) 1.21 (8/9) Doko et al., 1995 Romania 1992 corn 0.01 (3/6) 0.01 (1/6) Doko et al., 1995 South Africa 1985 com 1.60 (12/12) 0.50 (10l12) Sydenham et al., 1990 1985 moldy corn 29.3(12/12) 7.55(12/12) - - 1989 corn 1.53 (5/6) 0.42 (5/6) Rheeder et al., 1992 1989 moldy corn 53.74(6/6) 1368(6/6) - - 1989 export corn 0.29 (28/68) 0. 13 (1 0/68) Rheeder et al., 1994 1990 cornmeal 0.14 (46/52) 0.08 (11/52) Sydenham et al., 1991 1990 corn grits 0.13 (10/18) 0.09 (4/18) -"- 1990 comflakes nd (013) nd (0/3) - - Switzerland 1986- corn 2.88 (7/7) 0.24 (7/7) Stack and Eppley, 1992 1991 USA 1989 corn 0.64 (7/7) 0.18 (6/7) Sydenham et al., 1991 1990 cam 55.40 (616) 15.08 (7/7) Stack and Eppley, 1992 scree_niflgs 1990 com foods 0.43 (25/36) 0.14 (18/36) - - 1990 corn food' 0.41 (4/4) 0.15 (3/4) Sydenham et al., 1991 1990 cornmeal 1.05 (15/16) 0.30 (13/16) - - 1990 corn grits 0.60 (10/10) 0.38 (5/10) -"- 1990 comflakes nd (0/2) nd (0/2) - - 1991 cornmeal 0.09 (2/7) nd (0/7) Pittet et al., 1992 1991 corn grits 0.26 (34/55) 0.10 (13I55) - — 1991 comflakes 0.06 (1/12) nd (0/12) -”— 1991 sweet corn 0.07 (1/7) nd (0!?) -"- 1991 poultry feed 0.24 (6/22) 0.09 (2/22) - - 1991 corn foods 0.55 (11/13) 0.13 (10/13) Stack and Eppley, 1992 Zambia 1992 Corn 0.18 (20/20) 0.05 (15/20) Doko et al., 1995 nd indicate not detected. - indicate not analyzed. -”- same as above tin: argued IIEW an contamin 'l’9llow ra The alienating natural occ WI. Bas Weed tr toll In hum a r 16 Interest in trichothecenes arose world wide in September, 1981 after the United States indirectly accused the Soviet Union, \ertnam and Laos governments of employing these mycotoxins as chemical warfare agents (Bamburg, 1983; Watson et al., 1984). The accusation was based on several findings. Firstly, analyses of blood, urine, and internal tissues of “yellow rain” attack victims revealed extremely high levels of trichothecenes (Watson et al., 1984). Secondly, a high level (up to 20 times higher than any recorded natural outbreak) of trichothecene mycotoxins was also found in a Single leaf and stem sample taken from a region of Kampuchea where “yellow rain” was reported (Seagrave, 1981). Thirdly, normal background levels of these toxins were essentially undetectable and natural occurrence of trichothecenes in Southeast Asia was absent (Bamburg, 1983). However, a group of researchers disagreed with the above findings. They argued that “yellow rain” was merely naturally occurring bee feces where fungus grew and produced trichothecenes. In addition, consumption of trichothecene- contaminated foods might have yielded similar symptoms to the symptoms of “yellow rain” attack victims (Schiefer, 1988). The controversy resulted in intensive research activities directing towards elucidating many aspects of trichothecene mycotoxins including their detection, natural occurrence, toxicological, biological and biochemical action (Bamburg, 1983). Based on this and other research over the last five decades, it is now recognized that trichothecene mycotoxins are etiologic agents of mycotoxicoses both in human and animals (Ueno, 1980; 1983; Bamburg, 1983; CAST, 1989). that 13 I Bet natu toxm posit numb Betina dassifr 1.4). t toxin ar deoxynn such asr beMeenc The generally 5 Cllloroform’ l97l.’ Ueno, aIcohol den homologue leadlly 83%“ 17 Chemistry. Trichothecenes all have tetracyclic, sesquiterpenoid structure that includes a six-member oxygen containing ring, an epoxide group in the 12, 13 position, and an olefinic bond in the 9, 10 position (Ueno, 1980; 1983; Bamburg, 1983; Betina, 1989). Structure and numbering system of some naturally identified trichothecene myCotoxins are illustrated in Figure 1.3. These toxins possess oxygen-containing substitutes at one or more C-3, 4, 7, 8, and 15 positions. The substitutes may be hydroxyl, esterified hydroxyl, keto (carbon number 8 only), or epoxide (carbons number 7, 8 only) groups (Ueno, 1983; Betina, 1989). Based on trichothecene structural characteristics, Ueno (1980; 1983) classified trichothecenes into 4 different groups (group A, B, C and D; Figure 1.4). Group A trichothecenes have hydroxyl or acetoxy substitutes, such as T-2 toxin and trichodermin. Group B trichothecenes are 8-keto derivatives such as deoxynivalenol and nivalenol. Group C trichothecenes are 7,8 epoxide derivatives such as crotocin. Group D includes trichothecenes containing a macrocyclic ring between carbon number 4 and 5 such as verrucarin A and roridin A.(Ueno, 1980). Trichothecenes are colorless, crystalline, optically active solids, and are generally soluble in non-polar solvents such as acetone, ethylacetate, and chloroform, but less soluble in polar solvents (e.g. water) (Bamburg and Strong, 1971; Ueno, 1980). Trichothecenes are more stable in solid condition. Their alcohol derivatives have a higher solubility in water than their esterified homologue (Betina, 1989). Under a mild alkaline solution the ester groups are readily saponified and result in less acylated derivatives or parent alcohol forms Ilia Sci Deo (v01 Nive 11155 Figure 18 j-" Ezicnognecene R1 R2 R3 R4 R5 T-Z toxin on OAC OAC H ooccu,ca (CH3) , arr-2 toxin on on OAC H ooccnzcn (C8,) , T-Z tetraol OH OH OH H OH Diacetoxyscirpenol OH OAC OAC H H Scirpenetriol OH OH OH H H Deoxynivalenol . (vomitoxin) OH H OH OH O Nivalenol OH OH OH' OH O Fusarenon-x OH OAC OH OH 0 Figure 1.3. Structure and numbering system of some naturally identified trichothecene mycotoxins (modified from Ueno, 1983). Figure I. 19 0 R1 11’ RI Group D Figure 1.4. Classifiwsion of trichothecene mycotoxins based on their chemml structure (modified from Ueno,1980). 20 (Ueno, 1980). The double bond at carbon 9, 10 can be catalytically reduced to a dihydro derivative, and the corresponding alcohol groups are then easily oxidized to ketone or aldehyde functional groups. Natural occurrence. Only a few members of the trichothecenes, such as deoxynivalenol (DON), nivalenol (NIV),T-2 toxin (T-2), HT-2 toxin, and diacetoxyscirpenol (DAS) are detected as natural contaminants in cereal grains although over 80 members of these toxins have been characterized in the laboratory (Betina, 1989). Production of trichothecenes in cereal grains is mainly dictated by environmental conditions (Vesonder et al., 1978; Ueno, 1983; Cote et al., 1984). Wet and cool seasons favor Fusarium infection and trichothecene production in the field (Tuite et al.,1974; Shotwell et al., 1977, 1983; Ueno, 1983; CAST, 1989). Tuite et al. (1974) reported that during the unusually cool and wet summer of 1972, corn produced in the United States was heavily infected with Fusarium gram/nearum, and caused outbreaks of feed refusal and emesis in swine. DON was believed to be an etiologic agent for these outbreaks. Shotwell et al. (1977, 1983) observed Similar effects of weather on Fusarium contamination in wheat, grown in VIrginia from 1975 to 1980. The weather was unusually cold and rainy during the 1975 growing season, but during the following 5 years (1976 to 1980) such weather did not occur. These investigators found zearalenone in 19 of 42 samples from wheat grains harvested in the cool and wet growing season; however, they did not find the toxin during the 1976-1980 harvest period. Later, DON was also detected in the zearalenone-positive samples (Shotwell and () 21 Hesseltine, 1983). “Red mold disease” in Japan that is caused by Fusarium gramineanrm and Fusarium nivale (Ueno, 1983), also occurred after a long rainy season in 1963 and in 1970. Similar weather conditions resulted in “scabby wheat” (soft and shriveled wheat and often with a pink discoloration) in the United States and Canada (CAST, 1989). This “scabby wheat” was caused by Fusarium gram/nearum which produced DON (Shotwell et al., 1985; Tanaka et al., 1988a; Fernandez et al., 1994). The natural occurrence of DON as well as NIV in cereal and cereal products has been surveyed by several investigators in several countries and the results are summarized in Table 1.3. Human and animal mycotoxicoses. Outbreaks of human and animal diseases associated with consumption of trichothecene-contaminated foods and feeds occurs in many countries usually after a cold and wet harvest season (Bamburg, 1983). Although such outbreaks have been reported to take place Since the late 19'h century (Bamburg, 1983; Ueno, 1983), elucidation of trichothecenes as an etiological agent in mycotoxicoses was not reported until 1972. At that time, Hsu et al. (1972) demonstrated that 2 ppm of T-2 toxin, a trichothecene produced by Fusarium tn'cinctum, was detected in moldy corn associated with illness and death of lactating cows. Since that time other trichothecenes such as diacetoxyscirpenol, nivalenol, fusarenon-X, deoxynivalenol and HT-2 toxin have also been found naturally in feedstuffs and in cereal grains (Betina, 1989).- 22 Table 1.3. Natural occurrence of trichothecene deoxynivalenol (DON) and nivalenol (NIV) in cereals and cereal products CROP GRAIN MEAN IN POSITIVES, PPM COUNTRY YEAR PRODUCTS (POSITIVES/SAMPLES) REFERENCE DON NIV Argentina 1983 wheat 0.02(3l20) nd (0/20) Tanaka et al., 1988b 1983 barley 024(18/20) 0.03(15/20) - - 1983 corn 011(2/20) nd (0/20) - - Austria 1983 wheat 0.36(3/4) 0.03(3/4) Tanaka et al., 1988b Bulgaria 1983 wheat 0.21 (1/2) 0.03(1/2) Tanaka et al., 1988b Canada 1980 wheat 030(72/72) - Scott et al., 1981 1980- wheat 1 .26(9/10) 0.02(4/10) Tanaka et al., 1988a 1984 1984 corn 0.96(1I1) 0.01(1l1) - - 1982 rye 0.20(1/1) 0.01(1l1) Tanaka et al., 1988b China 1984 wheat 1.71 (1 [5) 6.64(1/5) Ueno et al., 1986 1985 wheat flour 0.19(7/7) nd (0”) - - 1984 wheat 4.28(4/4) 016(3/4) Tanaka et al., 1988b England 1984 wheat 0.03(20/31) 0.10(17/31) Tanaka et al., 1988b Finland 1987- cereal 0.13 - Hietanlemi and 1978 (246/268) Kumpulainen, 1991 France 1984 wheat 0.09(1/2) 0.04(2/2) Tanaka et al., 1988b Germany 1984 wheat 0.71(2/8) 0.27(0/8) Tanaka et al., 1988b 1984 barley 0.19(2/13) 0.04(1/13) - - 1984 cat 0.14(4/10) 1.45(1/10) -"- 1984 sonan nd (0/1) nd (0/1) -'- 1984 rye 0.41(4/22) 0.01(4/22) - - 1987 barley 0..40(43/44) 0.01(5/44) Muller and Schwardorf, 1993 1984 rye flour 0.17(1/1) 0.01(1l1) - - Greece 1984 wheat 0.01(1l1) 0.01(1l1) Tanaka et al., 1988b Hungary 1984 wheat 0.67(2/2) 0.01(1l1) - - India 1989 sorghum nd (0/150) - Ramakrisna et al., 1 990 1989 cam nd (0/102) - -”-- 1989 wheat 0.31 (1/58) - -"- 1989 whole - -”- wheat flour 438(1 1/37) 1989 feed sample nig01102) - - - Italy 1984 wheat 012(1/12) nd (0/12) Tanaka et al., 1988b 1984 barley 0.19(2/5) 0.02(1/5) - - 1984 corn 0.40(2l3) nd (0/3) -”- 1984 cat nd (015) nd (0/5) -"- nd indicates not detected. -"- indicates the same as above. - indicates not analyzed. Table 1.3. Continued 23 CROP GRAIN MEAN IN POSITIVES, PPM DON NIV Japan 1983 wheat 0.02(4/6) 0.39(6/6) Tanaka et al., 1985. 1983 barley 0.25(5/5) 0.71 (5/5) - - Korea 1983 wheat 0.01(2/10) 0.14(9/10) Lee et al., 1985 1983 rye 0.01(5/5) 0.08(5/5) Lee et al., 1985 1984 wheat 0.02(5/9) 053(9/9) Lee et al., 1986 1983 barley 012(26/28L 055(28/28) Lee et al., 1985 1984 barley 012(31/31) 0.50(31/31) Lee et al., 1986 1989 barley 0.26(2/11) 0.30(3/11) Park et al. 1991 1989 rice nd (0/8) nd (0/8) - - 1989 corn 0.62(1/3) 035(1/3) —'- 1989 millet 0.34 (1/6) 023(1/6) - — 1990 barley 0.19(24/27) 1.11(27I27) Park et al., 1992 1993 barley 0.17(?/39) 1.01 (7/39 Kim et al., 1993 1993 corn 0.31(?/46) - - - Nepal 1984 wheat 0.06(1/1 0) 0.07(5/10) Tanaka et al., 1988b 1984 barley nd (0/4) 0.02(1/4) - - 1984 cat I'Id (0/7) 0.02(4/7) -"- 1984 rice nd (0/9) 0.02(2/9) -”- 1983 rye nd (0/2) nd 0/2) -"- 1984 com 0.54(3/9) 0.89(6/9) - - Netherlands 1984 cereals 0.22(26/29) - Tanaka et al., 1990 1988- feeds 0.63(19/95) - Veldman et al., 1992 1989 Poland 1984 wheat 010(13/48) 0.05(37/48) Tanaka et al., 1988b 1984 barley 0.39(1/6) 0.08(3/6) Tanaka et al., 1988b Pmm 1984 wheat nd (0/4) nd (0/4) Tanaka et al., 1988b Scotland 1984 wheat 0.03(1/20 nd (0/2) Tanaka et al., 1986 1984 barley 0.04(5/8) 0.39(3@ - - S. Africa 1987 wheat 0.87(3/3) - Sydenham et al., 1989 Sweden 1984 wheat nd (0/1) nd (0/1) Tanaka et al., 1988b Taiwan 1984 wheat 056(9/12) 0.07(6/12) Ueno et al., 1986 1985 barley 0.08(4/4) 0.63(4/4) - - 1985 wheat 0.25(3/10) 0.02(4/10) - - USA 1977 corn 230(24/52) - Vesonder et al., 1978 1981 cam 3.1(274/342) - Cote et al., 1984 1982 wheat 1.7891/33) - Hagler et al., 1984 1982 wheat 1 .17(45/161) - Shotwell et al., 1985 9le Table 1.3. Continued 24 CROP GRAIN MEAN IN POSITIVES, PPM COUNTRY YEAR PRODUCTS (POSITIVESISAMPLES) REFERENCE DON NIV 1984 wheat 0.59(75/123) - Wood and Carter, ' 1989 1984 corn 0.39(61/92) - -”- 1984 dairy grain 0.10(60/101) - Whitlow et al., 1986 1 985 wheat 0.49( 57/ 1 24) - Wood and Carter, 1 989 1985 corn 047(32/106) - - - 1989 human food 4.00(46/92) - Abouzied et al., 1991 1991 wheat 1.57(17/81) - Fernandez et al., 1 994 1990 corn 240(13/99) - Price et al., 1993 1990 winter 2.4(201/207) - - - wheat 1990 sping wheat 0.9(1 201206) - - - USSR 1984 cat nd (0/2) nd (0/2) Ueno et al., 1986 1984 spice nd (0/3) nd (0/3) Tanaka et al., 1988b 1986 wheat 0.59(6/140 - Tutellyan et al., 1990 1987 wheat 0.26(14/90) - - - 1988 wheat 1 .13(14162) - - - Yemen 1983 sorghum nd (0/6L 0.09(1/6) Tanaka et al., 1988b 1984 wheat 0.00(1/7) nd (0/7) - - 1984 barley 0.02(2/3) 0.01(2/3) -”- 1984 com 0.01(1/12) nd (0/12) -"- 1984 sorghum nd (0/5) nd (05) -”- 1984 soybean nd (0/2) ndJOQ) -"- 1984 sesame nd (0/7) nd (0/7) -"- nd' indicates not detected. -"- indicates the same as above. - indicates not analyzed. 25 Severe mycotoxicosis induced by trichothecene mycotoxins include alimentary toxic aleukia (ATA) in Russia (Joffe, 1962; 1965), red-mold disease in Japan (Bamburg et al., 1969), moldy corn diseases in The United States (Bamburg et al., 1969; Hsu et al., 1972), staggering grain toxicosis in Eastern Siberia (Bamburg, 1983), dendrochiotoxicosis in Russia (Bamburg, 1983), vomiting and feed refusal in the United States and other countries (Vesonder et al., 1973), been hull poisoning in Japan (Ueno, 1980), and gastrointestinal disorders in Kasmir Valley, India (Bhat et al., 1989). Symptoms, victims, and etiologic agents of these mycotoxicosis are summerized in Table 1.4. 26 coca. Esocxc: 85:38 5... ococofifiou 550 can w: ._oco.m>_c 295cm Em. on. c. .322 coo. one ._oco.m>_c>xooo Somme Sauces”. of «$er 6.00 .329 coo. .oc...Eo> can cmEs... can ac.._Eo> 35w ..oco9.om>xo.oom.a Sam 23.28:... 6:6. €382.65 653» .w: on: ~-...I .536. «9 83.285 .0? Eco $0.5m .cozoEEmcc. Exm .oaoctoEoI acm 2.60 on. E. E8 >222 35m .66 ._oco_m>_c>xooa .o.m>.E Esteem”. .8532... cocoa c. 3.3 .6562... .2568. Sateen”: .o...E on. c. ountoEo. 5...:35 .322 com. 2955 429.92. commas 83 8585055 EEmocEmS m .59.; Serum dozoEEmcc. Exm £9.th . ac...Eo> Em. docs... 208-com 35m 68 .82 6.38. WP... .538. 83.39% Emma .93sz bofisamo. .BoEoE econ oaoSm .EEoo .32 N-bmocooo....ozo..= 2.3.88ch Scan .0 8.8358 .933 .995 2.98: ace manna c. .ctmmEomoa .omoqm .502; .65.: 685.089.. 29:2: .wEoQOxao. 2.2. 3.33 #3.. 6.698903 .mobeEoEanm .n. c22£2co>o .moctofi cosmEEmcc. Exm .uc.._Eo> cmEaz 0.36. bo.coE..< $3.. macaw c. 82552 85937.6 .953 .mc...Eo> .momamc .aEEm Em. £323. .05 282.022.... 53on Sateen“. >259 .655. .m..._..o 65525 69:? scooped: ace cmEax 9.598% 83 82:65 .3332 use 5.832.! 2.95". 33588.30 8.33.56 «8.85 303:6... so; $80.38. 3035658505.: co .cmEezm .1 992. 27 .0023 “um—@800 6.08. NF .22 .200 46.0.: 60.. 0050000 0.0:. 0. 5200.0 .00. 0000005020.... .000 2.3.2003”. 000.: 000.5 .0 0005.3. .0 05.00. m 0.00 .0502 0053.. 30.30.52.006 33 .c.x0. .__00 020: 0000.. NP: .2368? 0.00.0 0000 0:0 .0 02.900000“. 6002:0505 60.0030. 0. .uc.com.00 003 .c.x0. «F .30 53.00030. 2.3.. 0000 00.5 ..coEm>0E 5.32.0 60.8.... 8.0.... 00.0: ..3c.cmmm 0005 00.5038) «E3000. 0.00.5950. 6.0.3.300. 0053; 0.003". c. 0.0 53 0:0 0.0.5.. $500595... 3.3.0000“. 6000:0505 600ch .5000 x230 .0500 .030: 00.x0.0.:00.0cmo 005m 00.002000 .00....0300 002.0 20020 0055.0 0.000950. 0083: 0:0 0:0 E803... «0:00.000 02.00 $0.000 6.0030930. .3090 .9260 .0530 .0002... 6.003... c. 0.00 33 0.0 .1 5.8.8.8 .m. 0.500300% $0.. .326 c. 000E880: .co..0EEm..c. 0.05 .0200 .080... 0.x0.0>..00>:00.m 00:.» 0.03020 00:000. 0:0 5.8.00»: 039.3“. 00.00.50.000 05805.6 08:05 0000038.. 50> 835.80 .3 0.33 CONVENTIONAL ANALYTICAL METHODS The presence of trichothecenes as well as fumonisins in foods and feedstuffs is potentially hazardous to human and animal health (Ueno, 1983; CAST, 1989; Norred, 1993; Pohland,1993) and cause economic loss to farmers and livestock producers (CAST, 1989). Therefore, elimination of the mycotoxins from foods and feeds is very crucial. Although, prevention and elimination of mycotoxins from human and animal diets is very difficult, detection and diversion of contaminated raw materials from feed and food use can reduce the toxin in the diets (CAST, 1989; Pestka et al, 1995). Conventional methods for mycotoxin detection include thin layer chromatography (TLC), high performance liquid chromatography (HPLC), gas chromatography (GC) and mass spectroscopy (MS) (Pestka, 1994). Purification and detection of fumonisins will be reviewed in Part II. In the following sections purification and detection of trichothecenes are described. Cleanup of trichothecenes prior to analytical detection. Analyses of trichothecene mycotoxins in foods and feeds involve sample extraction and a very extensive cleanup to purify the mycotoxins before detection is made (Snyder, 1986; Chu, 1991; Gilbert, 1993). Based on trichothecene solubility in various solvents, Snyder (1986) classified trichothecene mycotoxins 28 go: go: a (T mix pr 0 9f: are [ml 10) EX. an, al, 29 into two groups. Group I toxins which have one or more acetyl groups, such as T- 2 toxin, HT-toxin, diacetoxyscirpenol, are very soluble in nonpolar solvent, such as chloroform, methylene chloride, ethylacetate, diethyl ether, and acetone. Group II toxins, which have no acetyl groups (e.g. DON, NW, and T-2 tetraol) are highly soluble in polar solvents, such as ethanol, methanol, water, aqueous methanol and aqueous acetonitrile because of stronger polar side groups in the group II molecules. Therefore, nonpolar solvents are suitable for extraction of group I trichothecenes, and polar ones for group II toxins (Snyder, 1986). When a mixture of group I and II compounds is extracted, this author suggested using a mixture of acetylacetate and acetonitrile as a solvent system. However, this procedure requires an extensive purification to remove the acetylacetate from the group I toxins. (Snyder, 1986). Polar solvents such as methanol or acetonitrile in combination with water are now commonly used for extraction of mycotoxins from sample matrices because of increasing use of reverse-phase liquid chromatography column and immunoassay (Whitaker et al., 1986). In addition, those solvent systems are less toxic and less expensive than those used early. Furlong et al. (1995), for example, used a mixture of methanol and aqueous 4% KCI solution at ratio of 9:1 (vol.lvol.) for extraction of trichothecenes from wheat. Meanwhile, a mixture of acetonitrile and water at a ratio of 3:1 has also been used to extract DON, NIV and zearalenone from cereal grains (Tanaka et al., 1985; 1986; 1988a,b; Park et al., 1991; 1992; Muller and Schwadorf, 1993). 30 After extraction, trichothecenes can be separated from their crude extracts via liquid/liquid or solid/liquid partitioning techniques (Snyder, 1986; Chu, 1991). An example of a liquid/liquid partition solvent system used for T-2 toxin separation is a mixture of methanol/ethylacetate/chIoroform at a ratio of 1:122 (vol.lvol.lvol.) (Hsu et al., 1972). A mixture of acetonitrile and petroleum ether at a ratio of 1:1 (vol.lvol.) is also used in the separation of T-2 toxin or DAS (Mirocha et al., 1976). In solid/liquid partition systems, columns which are used for trichothecene cleanup include ferric gel (Mirocha et al., 1976), activated charcoal (Morooka et al., 1972), activated charcoal-alumina (Romer, 1984), florisil and silica gel (Tanaka et al., 1985), and charcoalzaluminazcelite at a ratio of 7:5:3 (wIw/w) (Eppley et al., 1986; Trucksess et al., 1986; 1987; Fernandez et al., 1994). The availability of higher capacity and more effectively controlled size absorption packing materials led to the development of a smaller but more efficient column chromatography (Chu, 1991). Such smaller columns including SAX (Thiel et al., 1991), Amberlite IRC 50 (Shepherd and Gilbert, 1986), Sep-Pak (Mirocha et al., 1989) are now commonly used for mycotoxin purification, instead of using large columns such as silica gel or using solvent partition methods (Chu, 1991). The following step after purification is mycotoxin detection which can be performed with TLC, GC, HPLC, or MS methods (Pestka et al., 1995). Detection of trichothecenes TLC has always been a favorite method for analyses of mycotoxins because of its simplicity and low cost (CAST, 1989). Most of the TLC studies 31 usually employ silica gel as an absorbent. The thickness of the thin layer is generally 0.25 mm and various solvent systems are used as developing agents (Ueno, 1983). Retention factor (Rf) values of some trichothecene mycotoxins are summarized in Table 1.4. Derivatization reactions are required when TLC is applied to trichothecenes because most trichothecene toxins do not have useful absorption or potential for fluorescence under ultraviolet or visible light (Ueno, 1983). Compounds used as derivatization reagents include sulfuric acid (Ueno et al., 1973; Gimeno, 1979), p-anisaldehyde (Scott et al., 1970), 4-(p- nitrobenzyl)piridine (NBP) (Takitani et al., 1979), nicotinamide-Z-acetalpyridine (Sano et al., 1982), and aluminum chloride (Romer, 1986; Trucksess et al., 1987; Fernandez et al., 1994). Use of a certain derivatization reagent depends on the objectives of trichothecene analysis, the kinds of trichothecene mycotoxins to be analyzed, and the methods of toxin purification (Ueno, 1983). This author suggested to use sulfuric acid, aluminium chloride, or nicotinamide-Z- acetylpiridine for analysis sensitivity; aluminium chlon'de, NBP, or nicotinamide—Z- acetylpin‘dine for toxin selectivity; sulfuric acid, aluminium chloride, or NBP for procedure simplicity; aluminium chloride, or nicotinamide—2-acetylpiridine for derivative stability. For general purposes, aluminium chloride or NBP is most frequently used as a derivatization reagent (Ueno, 1983). Color development using sulfuric acid, or p-anisaldehyde reagents is performed by spraying the reagents on a TLC plate and then heating at 100- 130°C for about 20 minutes (Scott et al., 1970; Ueno et al., 1973). When sulfuric 32 - - - mm . .0 . 00.0 .00 50.0 000.0 800.020.0800 - - - - - 00.0 $0 - - .000_0>.0_...0000._.0.. vmd mvd 5‘0 - - 3.0 5.0 - - 800.020.3805 «00 .010 5.0 omwd - 00.0 3.0 00.0 -050 0000000003... .00 60 No.0 - - 00.0 00.0 mm .0 - .000.0>.z - - - - 00.0 - - - - 0.000.002... - - - - m . .0 - . - - 000.800.0005 0 030.0 00.0 00.0 5.0 0 3.20 0 r .0 N00 000 - wmmd 006. N-.. mmd :0 0.0 09.0 000 E0 00.0 - r90 0.08. NF... «00 mmd m . .0 mm . .0 - 00.0 00.0 - 00.0 8.00.0802 - - - .N .0 0 - - 0 .0000. «P .00000.0m>x0.000.0 - - - - . .00 00.0 - - 06.30.00. 00.0 00.0 5.0 05.0 v . .0 00.0 «0.0 - Evd .0000..0m>xo.000.0 - - - .w r .0 - - - - 000.0 .00000.0w>x0.000000.2 - - - {0.0 - - - - 000.0 80.000000 - - - - 00.0 - - - - .0.00300> - . - - 5.0 - - - - 0.0000002... - . - - x. F .0 - . - - 800000000... < 030.0 ._ v. e _ 0 n. m 0 0 529m .5200 .000. .0000 .00.. 00.0000. .00000000 00 .00 02.0 00.03 00.0.0 0.. .. 00 00.8.8.6. 000000.000... 0.000 .0 m03.0> cm. 8.00. 00:00.0”. .0... 0.00.. |( L ‘1 II‘Q“.P 33 005000 .00 00.00.00. .. .. .0000 .0 0.02.0205 2.0. .0 0.0.000...0.0..0000.0.E.o.0.200 00.00.00. .. .0000 .0 0.02.9.0... 2.0. .0 0.0.000.>0.0H.00000.000.”8.0.0.200 00.00.00. v. .0 .0 202.05 2.0. .0 00000000000350 00.00.00. 0 .0 .0 302.03 2.0. .0 053.200.000.20 00.00.00. . .0 :00 .0 202.03 2.0. .0 00.:.o.0>00..0..000~000 0.00.00. I .0 .00 .0 302.03 2.0. .0 000.002.580.200 00.00.00. . .3 .0 202.0... 2.0. .0 002000003000 00.00.00. m 5.0200000. .00 000 .000 .0<.0 .000 .0 302.0... 2.0. .0 .0000.00.HE.0.0.0_00 0.00.00. 0 000 .o .0 .< ”00.0.0.6 .00200 - v0.0 000 00.0 - - 0.00.0.0 - .00 - - - - 6020.0 0 030.0 00.0 - - - - 00.0 0 0.2.00 00.0 - - - .00 0.20 < 0.2.0.". 00.0 - - - - 00.0 0 0..003..0> 00.0 - - - 00.0 .00 0 0..000..0> 00.0 - - - 00.0 00.0 < 0..003..0> 0 030.0 I 0 n. m 0 < 50.0..» .5200 82.28 .3 038 34 acid is used, group A and B trichothecenes generate a grayish black color, and a brown color, respectively. The detection limit of this system is approximately 0.25 pg per spot (Ueno et al., 1973). In addition, under 360 nm UV light the group A give a blue fluorescent color with a detection limit of 0.05 pg per spot (Ueno et al., 1973). The same detection limits is also obtained when p-anisaldehyde is used as a derivatization reagent, but group A trichothecenes give a pinkish violet color and the group B trichothecenes yield a yellowish brown color(Scott et al., 1970). Takitani et al. (1979) reported that a detection limit of 0.02-0.2 pg per spot was possible by using NBP as a derivatization reagent. A blue-violet color for trichothecenes was observed under long wave UV light after a TLC plate was sprayed with NBP solution, heated at 150°C for about 30 minutes, cooled at room temperature, and then dipped in tetraethylenepentamine solution. The blue-violet color was a compound which was formed through N-alkylation of N-atom on the piridine ring of the NBP reagent with the epoxy group of trichothecenes. These investigators suggested that this method was applicable to all trichothecene mycotoxins having a characteristic of 12, 13-epoxy group. More sensitive and specific TLC methods were developed by Nelis and Sensheimer (1981). They used nicotinamide as an alkylating reagent to produce fluorescent derivatives. An epoxide-containing compound such as trichothecenes is added to the solution of nicotinamide, a ketone (e.g. acetophenone), and alcoholic KOH. A blue fluorescent color is developed by the addition of formic acid at ambient conditions. These researchers reported that this system had a 35 detection limit of 0.1-2.0 ng per spot. If compared to NBP method, this technique is better because it is approximately 100 folds more sensitive. High-performance thin layer chromatography (HPTLC) has also been applied to detection of DON, fusarenon-X, and NIV in cereal grains (Trucksess et al. 1987). After purification, the toxins are spotted on a TLC plate which has been previously impregnated with aluminum chloride. The plate is then developed in two sequential solvent systems, firstly in chloroform:acetone:2- propanol at ratio of 8:1:1(vol.lvol./vol.), and secondly in the same solvent mixture but at a 14:3:3 ratio. After air drying, heating at 120°C for 8 minutes, and cooling at room temperature, the plate is observed under long-wave UV light. The toxins appear as blue fluorescent spots. Rf values of DON, fusarenon-X, and NIV are 0.5, 0.4, and 0.1 respectively. Average recovery of these three toxins added to cereal grains at levels of 100 and 200 ppb was 83%, and the detection limit of this system was 50 pbb (Trucksess et al., 1987). Fernandez et al. (1994) also used this technique for detection of DON in 1991 US. winter and spring wheat and they found that the limit of determination was 40 ppb, and average recoveries of DON-spiked wheat samples at levels of 200, 400, and 800 ppb were 83, 82 and 72%, respectively. Trichothecenes can be analyzed either individually (Lauren and Greenhalg, 1987) or collectively (Lauren and Agnew, 1991) with HPLC methods. Lauren and Agnew (1991) developed a multitoxin screening method for Fusarium mycotoxins using HPLC techniques. They hydrolyzed trichothecenes (DON, NIV, scirpentriol, and T-2 tetraol) to form the toxin parent alcohol. This parent 36 alcohol is then detected with HPLC using a longwave UV detector. Recoveries of parent alcohol spiked into wheat and maize extracts at a level of 0.5 ppm ranged from 55 to 103%. Levels of 50 ppb or less were detectable by this method. Meanwhile, Lauren and Greenhalg (1987) observed a detection limit of 50-100 ppb for DON and NIV without hydrolysis. A higher detection limit (up to 0.02 ppb for DON and NIV) was achieved when HPLC was equipped with a fluorescent detector (Gilbert, 1991). This system involves post column heating with alkali to generate formaldehyde which is then derivatized with methyl acetoacetate and ammonium acetate in a second reaction coil (Gilbert, 1991). Trichothecene have frequently been analyzed by GC methods (Ueno, 1983; Snyder, 1986). GC analysis require derivatization of trichothecenes to form either trimethylsilyl (TMS) or heptafluorobutyril (HFB) derivatives, prior to detection with a capillary column which is equipped with a flame ionization detector (FlD) (Kamimura et al., 1981; Trucksess et al., 1987) or an electron capture detector (ECD) (Ware et al., 1984). Detection limits of GC—FID are 200 ppb and 100 ppb for group A and B trichothecenes, respectively (Kamimura et al., 1981). Detection limits of approximately 80 ppb for group A trichothecenes and 2 ppb for group B trichothecenes are achieved when ECD-GC is used (Ware et al., 1984). GC analyses are generally preferred to HPLC methods because GC has a greater sensitivity and specificity. The greater separating capacity of GC compared with that of HPLC also enables researchers to monitor a greater number of trichothecenes in the same extracts. In addition, 60 when coupled a q: sou; met M8 the Clea GC 37 with MS analysis can simultaneously detect and confirm trichothecene mycotoxins in a food or feed sample (Gilbert, 1993). In earlier investigations, MS detection methods were not considered to be a quantitative analytical tool for mycotoxins. However, a high resolution MS coupled with computer search and integration capabilities has led to new MS methods for both identification and quantification of mycotoxins (Plattner, 1986). MS methods are preferable for trichothecene analyses since they do not require the compounds to have a chromophore (Chu, 1991). In some cases, extensive clean-up steps are not necessary for MS and a detection limit of tandem GCIMSIMS systems is 1 ppb for T—2 toxin in urine and blood (Mirocha, et al., 1989) In summary, analysis of trichothecene mycotoxins using a TLC method are rapid, simple and low cost. This method, however, is generally low in sensitivity and selectivity (Bamburg, 1983; Ueno, 1983). Higher sensitivity and selectivity of trichothecenes analysis can be achieved by using GC, HPLC, and MS techniques although they require laborious sample cleanups, and expensive instruments (Pestka, 1988; 1994; Chu, 1991). IMMUNOASSAYS FOR FUSARIUM MYCOTOXINS Fusarium mycotoxins are a group of secondary metabolites produced by toxigenic strains of Fusarium including Fusan‘um monilifomie that produces fumonisins and Fusarium gramineamm that produces DON (Betina, 1989; Bacon and Nelson, 1994). Fusan'um mycotoxins especially DON and fumonisins are toxic to both human and animals and commonly contaminate cereal and cereal products (Bamburg, 1983; Tanaka et al., 1988b; Marasas et al., 1988a; Sydenham et al., 1991). Elimination of mycotoxins from human and animal foods is commonly accomplished via detection and diversion mycotoxin-contaminated raw materials from feed and finished food use (Pestka, 1994). Mycotoxin detection can be carried out using either conventional methods such as TLC, GC, HPLC and MS methods or immunoassay techniques such as ELISA. ELlSAs are commonly preferred to conventional methods because of simplicity, rapidity, and applicability both in the field and laboratory (El-Nakib et al, 1981). The basis of mycotoxin immunoassays involves competition between a free mycotoxin and a labeled mycotoxin for an antibody binding site (Pestka, 1988). The purpose of the following review is to discuss Fusarium-mycotoxin immunoassays with specific emphasis on antibody production, mycotoxin immunoassay format, application of mycotoxin immunoassays in foods, and commercial mycotoxin immunoassay kits for foods. 38 rx 39 Antibody production Antibodies or immunoglobulins are glycoproteins that are produced and secreted into the body fluids by B lymphocytes of animals in response to a foreign chemical (antigen) significantly different from the animal’s own chemical (Candlish, 1991). lmmunogenicity refers to the capacity of an antigen to induce an animal’s immune response and usually dependent on its chemical structure and its size (Pestka et al., 1995). Production of a highly specific antibody in an animal is mainly influenced by the type of antigens, dose and route of immunizations, the number and types of accessory cells such as macrophages that initially interact with antigen and induce lymphocyte activation, and the nature of responding lymphocytes (Abbas et al., 1994). Generally, immunogen must be degradable and must have an epitope that can bind to the cell-surface antibody of a B virgin cell. It must also have at least one site that can be recognized simultaneously by a class II protein and by a T-cell receptor, thus facilitating cell- to-cell communication between helper T cells and B cells can occur (Harlow and Lane, 1988). Compounds smaller than 3000 daltons (hapten) may be able to bind to the surface antibody of B cells, but may not have suitable site for the 3 simultaneous binding of a class II protein and a T-cell receptor. Most Fusarium mycotoxins are of low molecular weight (250-500 dalton) (Betina, 1989), and thus are not immunogenic by themselves (Chu,'1986). This problem can be overcome through conjugation of the toxins to larger immunogenic molecules (carriers). 4O Mycotoxin conjugation. Mycotoxin conjugation is a reaction of a mycotoxin and a carrier protein to form a mycotoxin-protein carrier conjugate so that the mycotoxin becomes immunogen. Carrier molecules that are commonly used for mycotoxin conjugation are capable of imparting immunogenicity to covalently coupled mycotoxins or haptens. These include bovine serum albumin (BSA), chicken ovalbumine (OA), and keyhole limpet hemacyanin (KLH), and cholera toxin (CT) (Chu, 1986; Harlow and Lane, 1988). BSA is very soluble and a good protein carrier (Harlow and Lane, 1998). These authors reported that BSA has 30-35 lysines, 19 tyrosines, 35 cysteines, 39 aspartic acids, and 59 glutamic acids that are available for coupling. Meanwhile, CA has 20 lysines, 10 tyrosines, 6 cysteines, 14 aspartic acids, and 33 glutamic acid residues. KLH (MW 4.5x 105 to 1.3x107), a copper containing protein, belongs to a group of non—heme proteins called hemacyanins which are found in arthropods and molluscs (Senozan et al., 1981). KLH is isolated from the mollusc Megathura creulata. ln Tris buffer, pH 7.4, this protein exists in 5 five states of aggregation which have sediment coefficients of 102$, 130$, 150$, 1708, and 1868 (Senozan et al., 1981; Herckovits, 1988). It will reversibly dissociate to subunits when pH changes moderately. Both lowering and raising of pH will cause dissociation (Herckovits, 1988) and at pH 8.9 it will completely dissociate to subunits. Each subunit contains oxygen binding sites which can bind to two copper atoms per molecule oxygen in KLH (Senozan et al., 1981; Herckovits, 41 1988). Unlike BSA, KLH is sometimes likely precipitated during coupling due to its large size, and this can make difficult in some cases (Harlow Lane, 1988). Cholera toxin (CT) (MW 86,000) is a protein exotoxin produced by Vibrio cholerae and has been shown to have strong and oral systemic adjuvant properties when co-administered with unrelated antigen (Liang et al, 1978; Lycke and Holmgren, 1986). CT consists of two types of subunits, a single ’heavy’ subunit of molecular weight 28,000 which noncovalently attached to a 58,000- MW aggregate of ’Iight’ subunits (Holmgren, 1981). CT has several advantages when used for generating antibodies (Azcona-Olivera et al., 1992a; Abouzied et al., 1993). Firstly, the procedure was rapid and yield a good quality of antibodies than standard protocols when the toxin was applied to generate F B1 antibody. Secondly, CT might be an humane alternative to Freund’s adjuvant since abscesses, ulcers, or granulomas at injection sites which usually appeared after F reund’s adjuvant immunization, are not observed after CT injection. Thirdly, because low doses of CT-immunogen yields a rapid and strong antibody response, CT would be valuable when mycotoxin availability is limited. Conjugation of a mycotoxin to a protein carrier can be achieved following chemical reaction of functional groups present on the mycotoxin with functional groups present on the carrier (Chu, 1986). Mycotoxins which already contain reactive groups such as fumonisin 81 , 32, and 83 can be coupled directly to protein molecules by using glutaraldehyde as a protein linker (Figure 1.5) (Azcona-Olivera, 1992a,b; Fukuda et al., 1994; Usleber et al., 1994). However, generation of protein conjugates for mycotoxins which do not contain functional 42 groups such as DON (Casale et al., 1988) is much more complex because a reactive group has to be introduced by chemical synthesis to the toxins prior to the conjugation reaction (Figure 1.5). A reactive group commonly introduced to trichothecenes is carboxylic acid which is performed by reacting the toxins with bifunctional acid anhydrides, such as succinic or glutaric anhydrides at the presence of a catalyst such as 4,N,N- dimethylaminopyn'dine or pyridine. Hemisuccinate and hemiglutarate of T2 toxin (Chu et al., 1979), diacetylscirpenol (Chu et al., 1984a), 3-acetyl DON (Kemp et al., 1986; Usleber et al., 1991), and DON (Usleber et al., 1993), as well as hemisuccinate of DON triacetate (Zhang et al., 1986), DON (Casale et al., 1988; Usleber et al., 1991), and 4, 15-diacetylnivalenol (Abouzied et al., 1993) have been prepared prior to the conjugation of the toxins to protein carriers. Conjugation of hemisuccinic or hemiglutan'c trichothecenes to protein carriers is usually accomplished using a mixed-anhydride (MA) or an activated ester (AE) method. In MA method the trichothecene derivatives are generally activated to their corresponding anhydride by isobutyl chloroforrnate in dry tetrahydrofuran and triethylamine and simultaneously coupled with protein (Gendloff et al., 1986); where as, in the AE method, N-hydroxysuccinimide and dicyclohexylcarbodiimide in dimethylforrnamide solution are used to activate the derivatives prior to conjugation process (Kitagawa et al., 1981). 43 I 3‘“; H non-om :10 m1. (‘ q 1” . ' g " our ir-cl,-(cu,),~¢'|;“l""“" {. O—BSA DON-E3834 Figure 1.5. Preparation of immunogen for (left) fumonisins and (right) deoxynivalenol (DON) (Pestka et al., 1995) 44 Polyclonal antibodies. After successful production, characterization, and purification of a mycotoxin-protein conjugate, suitable animal species are then immunized with the purified conjugate (Chu, 1986; Harlow and Lane, 1988). These latter authors suggest the use of rabbits, goats, or sheep for polyclonal antibodies and mice for monoclonal antibodies because only mice which have tumor cell lines for the efficient fusion of plasma cells. Multiple-site intracutaneous immunizations of rabbits with 100—1000 pg of mycotoxin-protein conjugate which has been mixed with an adjuvant are commonly performed to generate antibodies (Hariow and Lane, 1988). The conjugate is usually emulsified with an oil-base adjuvant containing killed Mycobacten’um such as “complete” Freund’s adjuvant at a ratio of 1:1 (vol/vol.) to allow slow release of the immunogen and nonspecifically induce immune response (Usleber, 1994). “Complete” Freund’s adjuvant is used for primary immunization and “incomplete” (not containing killed Mycobacten’um) one for subsequent injections which are usually given in 2-4 week intervals. High titer rabbit antisera commonly can be generated as early as 4 weeks after the initial injection (Zang et al., 1986; Wang and Chu, 1991; Abouzied et al., 1993; Usleber et al., 1993; 1994). Rabbit antisera contain antibodies which are generated by multiple B-cell clones and vary in specificity; therefore, they are considered polyclonal (Pestka et al., 1995). A major advantage of polyclonal antibodies is that they are easy and inexpensive to produce, and often contain a subclone of high affinity antibody (Usleber et al., 1994). However, the quality of polyclonal antisera in terms of affinity, 45 specificity as well as physical and chemical stability varies from bleeding to bleeding and from rabbit to rabbit (Pestka et al., 1995). As a consequence, use of these antibodies for constniction of commercial kits with defined performance characteristics is difficult. This difficulty can be eliminated when monoclonal antibodies are used. Monoclonal antibodies. Production of monoclonal antibodies involves animal immunization and fusion of splenocytes with a tumor cell line (Kohler and Milstein, 1975). lntraperitoneal or subcutaneous immunizations of female BALBIc mice with 5-50 pg mycotoxin-protein conjugate per injection are generally used to induce B cells which an synthesize the mycotoxin-specific antibodies (Azcona- Olivera, 1992b; Fukuda et al., 1994). The immunizations are usually given three times at two weeks intervals. The first and second injections were with an emulsion of adjuvant and the conjugate, and the final one was with the conjugation without adjuvant. Four days later, the mice producing mycotoxin-specific antibodies are killed and their spleen cells which contain high numbers of antibody-secreting B A cells are fused with myeloma cells (mouse cells which have all cellular processes necessary for the secretion of antibody but have no ability to produce antibody) using polyethylene glycol (Galfre and Milstein, 1981). During fusion, only about 1% of the starting cells are fused and only 1 in 105 form viable hybrids (Harlow and Lane, 1988). In tissue cultures the cells from immunized mouse spleens can not grow but the unfused myeloma cells can adapt and grow well (Harlow and Lane, 1988). These unfused cells have to be eliminated, 46 so they do not block the growth of the hybrid cells. Unfused myeloma cells, in which the hypoxanthine-guanine phosphoribosyl transferase gene (HPRT) in the salvage nucleotide synthesis pathway has been mutated, are killed by the addition of hypoxanthine, aminopterin and thymidine to the culture media (Azcona-Olivera, 1992b). Aminopterin will block de novo nucleotide synthesis pathway and force every cell in the culture to synthesize its purine nucleotides via salvage pathway. Under these conditions, cells containing a non functional HPRT protein such as myeloma cells will die and hybrids between spleen B cells with a functional HPRT and myeloma cells with a non-functional HPRT will continue to grow by using hypoxanthine and thymidine for producing their nucleotides (Hariow and Lane, 1988) Supematants of the hybridoma cultures can be screened for the presence of the mycotoxin-specific antibodies by competitive indirect enzyme-linked immunosorbent assay (Cl-ELISA) (Azcona-Olivera et al., 1992b). The selected cultures are then successively scaled up and cloned by limiting dilution at 0.5-1 cellMell (Goding, 1980). An immortalized subclone that consistently secretes antibody of desired affinity, specificity, and performance characteristics is then ' expanded to produce a large amount of monoclonal antibodies. Although monoclonal antibody generation is expensive, time consuming and requires for tissue culture facilities, these antibodies tend to exhibit a low degree of interassay Variation and give highly reproducible results (Pestka et al., 1995). Therefore, they are suitable for use in the development of commercial kits. 47 Recombinant antibodies. Beside using hybridoma technology, antibodies can be produced through gene recombinant technology which was developed recently. The gene technology involves reanangement or assembly of germ line V— genes, surface display of antibody, affinity selection, affinity maturation and soluble antibody production (Figure 1.6, Winter et 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). Later, Orlandi et al. (1989) isolated antibody genes directly form lymphocytes of immunized animals, and then propagated the genes by using universal primers and polymerase chain reaction (PCR) prior to expression the genes in bacteria. In addition, by building restriction sites into these primers, the amplified DNA can be cloned directly for expression in bacteria (Milstein, 1990). Like hybridoma technology, the later approach still relies on animal immunization to generate antigen specific lymphocyte cells. The hybridoma technology an immortalize these cells (Kohler and Milstein, 1975), while the gene technology can immortalize their genes (Milstein,1990). In future, it may become possible to generate antibody without animal immunization. Antibody genes are taken from unimmunized animal, and then amplify using universal primer and PCR (Orlandi et al., 1989). Prior to expression in bacteria, the genes can be readily manipulated by cutting and pasting of restriction fragments (Milstein, 1990) or by site directed mutagenesis (Better et al., 1983) to construct a new and desired antibodies. 48 Unrearranged V-genes ‘ Steps 1.2 Steps 1,2 ‘ Rearranged V-genes Assembled V-genes in phage(mid) ‘ Step 3 5‘99 3 * Figure 1.6. Process of antibody production in vivo (left) and using gene technology (right). Step 1: rearrangement or assembly of germ line V-genes; step 2: surface displaying of antibody; step 3: antigen-driven or affinity selection; step 4: affinity maturation; step 5: production of soluble antibody or antibody fragment (Adapted from VWnter et al., 1994). 49 Mycotoxin immunoassay format A number of immunoassay formats have been developed for mycotoxin analysis. Initially, competitive radioactive immunoassays have been developed for mycotoxins, at which a mixture of a constant amount of a radiolabeled mycotoxin and mycotoxin standards or unknown samples is incubated with specific antibodies (Pestka et al., 1995). Radioactivity of unbound radiolabeled mycotoxins in the solution is then measured after a complex of antibodies and radiolabeled toxin is removed from solution. Amount of the toxin in a sample is reversely related with unbound radiolabeled in the solution. Because of inherent problems with radioactive reagents, enzyme-linked immunosorbent assays (ELISAS) were subsequently developed on the procedures of Engvall and Perlman (1971). Solid phase supports used for mycotoxin ELISA include microtiter plates, beads, tubes, and dipsticks (T archa, 1991). These solid supports are most commonly made from polystyrene because of its low cost, easy process, optical clarity and high-protein binding (Tarcha, 1991). Among solid phase supports, polystyrene microtiter plates have been most commonly used because of supporting technology, such as removable stn'ps, multiwell pipettes, automated washers, and spectrophotometers (Pestka, 1994). ELISA procedures commonly applied for mycotoxin detection are competitive direct ELISA (CD-ELISA) and the Cl-ELISA (Figure 1.7). In the CD- ELISA, an enzyme-labeled mycotoxin and a free mycotoxin from samples are 5O incubated together over a solid phase-bound mycotoxin antibody (Pestka, 1988). This assay is based on the competition between free and the labeled mycotoxins. A concentration of mycotoxin in sample is inversely related to a complex of enzyme-labeled mycotoxin and the bound antibody. After the addition of an appropriate enzyme substrate, the bound antibody as well as sample mycotoxin concentrations can be calculated based on the intensity of color development which is measured spectrophotometrically. In the Cl—ELISA, a solid-phase mycotoxin-protein conjugate competes with a free mycotoxin for binding to a soluble-mycotoxin specific antibody. A second antibody which has been labeled with an enzyme is then used to determine total bound antibodies and free mycotoxin concentration. Recently, Pestka (1991) developed immunoassay format, called ELISAGRAM, by combining the sensitivity and selectivity of competitive ELISA with the capability of high-performance thin-layer chromatography (HPTLC) to separate structurally related mycotoxins (Figure 1.8). ELISAGRAM involves coating nitrocellulose (NC) membrane with mycotoxin-specific monoclonal antibodies, separation of mycotoxins by HPTLC, blotted the HPTLC with the NC, incubation of NC with mycotoxin-enzyme conjugate to identify mycotoxins bound to the monoclonal antibodies, detection of bound enzyme conjugate with a precipitating substrate, and visual or densitometric assessment of inhibition bands indicative of cross-reacting mycotoxins. This format has successfully been used to detect zearalenones and aflatoxins, and the result is similar to the competitive ELISA (Pestka, 1991). A major advantage of this procedure is that detection and Direct “ , {0% .-‘ $0, 0 Figure 1.7. Competitive ELISA for mycotoxins (adapted from Pestka, 1988). In direct ELISA, enzyme-labeled mycotoxins (ENZ) are simultaneously incubated with free mycotoxins (U)over solid-phase bound antibodies (AB). Concentration of free mycotoxins is inversely related to bound enzyme-labeled mycotoxins and can be measured quantitatively using spectrophotometer after color development by the addition of enzyme substrate. In indirect competitive ELISA, mycotoxin specific antibodies (AB) compete with free mycotoxins (D) to solid-phase mycotoxin-carrier protein (CP) conjugate. Second antibodies (ab- ENZ) which have been labeled with enzyme are then added to determined total antibody bound. Concentration of free toxin is inversely related to bound enzyme-labeled antibodies. 52 1. SEPARATE HAPTENS ON CHANNELED HPTLC SILICA GEL PLATE (I) a IMMUNOSPECIFIC TRANSFER: FILTER PAPER (field ANTIBODY-GOA NC (-) HPTLC (-) ' 3' $503330». HRP m ’ OONJUGATE (8) 4. INCUBATE NC WITH PRECIPITATING HRP SUBSTRATE. IDENTIFY HAPTENS AS INHIBIT ORY BANDS 5. PERFORM SCANNING DENSITOMETRY :1' r5: ‘0." " -‘ - 1,-2. . I," D A . .3 . ~ - ~ , c .‘..L " ,' -, a .-]a' c.’ . 2 ' ‘ .0 . .‘ . "\r' 9_ .193. . ': 0,“. ‘x i. -‘.- vr .. A' ' ’2‘. ."f'; .~ "' .- . ' " . t . .0 .' . D .:‘ ‘.? ‘.‘ :3. t : ’3‘ ‘0 ,' '. ‘0. a." 0. I ‘ z" ‘. ‘3‘. | 9. ..~‘._.. “a" Q .'. . ' ‘5. 0", . ‘. .0. “ . . u . . u- '0. . ¢ . . '0; O a. ' .0 a _' I ' I f f A. . K ‘ . Figure 1.8. ELISAGRAM procedure for mycotoxins (Pestka, 1991). 53 confirmation of multiple haptens can be performed simultaneously with a single cross-reactive antibody. More recently, a Computer—Assisted-MuItianalyte Assay System (CAMAS) has been devised by Abouzied et al. (1993) for simultaneously detection of fumonisins, aflatoxins and zearalenones. In this format, monoclonal antibodies for each of the toxins are immobilized as multiple lines on nitro cellulose membrane strips and sectored into hydrophobic compartments to minimize reagent use. A modified ELISA is performed at which free mycotoxins and enzyme-labeled mycotoxins compete for binding to the nitrocellulose-bound antibodies. By addition of a precipiting substrate, line-color intensity which is inversely related to mycotoxin concentration is developed. The color intensity can be measured quantitatively using a camera, video monitor, and microcomputer equipped with a video digitizing board. An advantage of this approach is that detection of several mycotoxins man be performed simultaneously in less than 30 minutes (Abouzied et al., 1993). In addition, assay data can be recorded in the minicomputer hard disk. Other immunochemical formats called “hit and run” assay for T-2 toxin (Warden et al., 1987) and immunochromatography for group A trichothecenes (Chu and Lee, 1989) have been also developed. In the “hit and run” assay, a T2 toxin column is equilibrated with fluoresce isothiocyanate (FITC)-Iabeled Fab fragments of lgG T-2 toxin antibodies. Samples containing T-2 toxin are injected to the column so the toxin binds to FlTC-Fab fragments. After washing, FITC- Fab—T-2 toxin complexes are eluted and then the toxin is determined in a 54 standard flow through fluorometer (Warden, et al., 1987). In the immunochromatography approach, which is a combination of HPLC and ELISA methods, group A trichothecenes are separated on C"; reversed-phase column. Individual fractions eluted from the column are assessed by ELISA using mycotoxin-specific antibodies. This approach can both identify and determine the concentration of individual group A trichothecene and its detection limit is 2 ng (Chu and Lee, 1989). Application of mycotoxin immunoassays in foods Use of immunoassay techniques for mycotoxin detection in foods and feeds is generally preferred to conventional methods, such as TLC, HPLC, GC, or MS because of lower costs, easier execution and quicker result (Pestka, 1988). Antibodies against important Fusarium mycotoxins such as fumonisins, trichothecenes, zearalenones have been produced and used for development of radioactive immunoassay (RIA) and ELISA methods. Both methods have been successfully used for mycotoxin screenings in a diverse array of foods and feeds. Selected examples of reported immunoassay for Fusarium mycotoxins are summarized in Table 1.6. An aqueous system is required for immunoassays because antibody and enzyme conjugate activities. Originally, extraction by standard procedures, evaporation, and reconstitution in aqueous buffer have to be done prior to mycotoxin analysis in solid foods such as grain and grain products (Pestka, 1988). In some cases, a column clean up is also performed to obtain higher 55 Table 1.6. Selected Fusarium mycotoxin immunoassays in foods Toxin Format Food analyzed Detection Reference limit (ppb) Fumonisin B, ELISA Buffer solution 50.0 Azcona-Olivera et al., 1992a,b ELISA Feed 250.0 -”- ELISA Buffer solution 0.6 Usleber et al., 1994 ELISA Corn 10.0 -”- ELISA Buffer solution 100 Fukuda et al., 1994 ELISA Foods 200 Pestka et al., 1994 Trichothecenes: Acetyldeoxynivalencl ELISA Rice 1.0 Kemp et al., 1986 15~Acetyldeoxy- ELISA Wheat 50.0-100.0 Usleber et al., 1993 nivalenol 4,15-Diacetyl- ELISA Buffer solution 5.0 Abouzied et al., 1993 deoxynivalenol Diacetoxyscirpenol ELISA Culture 16.0 Hack et al., 1989 DON RIA Corn, wheat 20.0 Xu et al., 1986 ELISA Corn 200.0 Casale et al., 1988 ELISA Grain-based 1000.0 Abouzied et al., 1991 food ELISA Buffer solution 1.0 Usleber et al., 1991 Nivalenol tetraacetate RIA Buffer solution 5.0 Wang and Chu, 1991 Roridin A ELISA Feed 5.0 Martbauer et al., 1988 T-2 toxin RIA Corn, wheat 0.1 Lee and Chu, 1981 a RIA Milk 2.5 Lee and Chu, 1981b ELISA Corn 50.0 Gendloff et al., 1984 ELISA Corn, wheat 2.5 Pestka et al., 1981a ELISA Milk 0.2 Fan et al., 1984 ELISA Wheat 0.5 Chiba et al., 1988 ELISA Cereal grains 100 Vetro et al., 1994 Zearalenone ELISA Corn 1.0 Warner and Pestka, 1986 ELISA Grain-based 2.5 Warner and Pestka, food 1987 ELISA and RIA indicate enzyme-linked immunoassay and radioimmunoassay, respectively. 56 sensitivity (Pestka et al., 1981a). However, since mycotoxin-horseradish peroxidase conjugate as well as solid-phase bound antibodies remain stable in 35% (vol/vol.) methanol in water (Ram et al., 1986), mycotoxin immunoassays can be directly applied to crude extracts of food samples (Usleber et al., 1994). In a liquid system, such as milk, mycotoxins can be detect directly with immunoassay approach although cleanup with a Sep—Pak or affinity column can increase the detection limit of immunoassays (Pestka et al., 1981 b). Sample matrix interference should be considered carefully when detecting mycotoxins in foods or feeds using immunoassay techniques (Laamanen and Veijalainen, 1992). These authors reported that certain substances in food sample might affect color development. When they detected T-2 toxin in millet grains with competitive ELISA assays, 75% of the grain gave higher optical density (OD) values than the negative control. However, a lower OD value than the negative control was obtained when the system applied to fermented foods, processed foods or feed stuffs. This problem could be eliminated by retesting the samples at higher dilution (Laamanen and Veijalainen, 1992). Incorporating toxin-free sample extracts during standard curve preparations can also minimize the false positive or negative reactions (Ram et al., 1986). Since reaction between antibodies and antigens occurs optimally at neutral pH, sample pH must also be considered prior to mycotoxin immunoassays (Pestka et al., 1995). 57 Commercial mycotoxin immunoassay kits for foods Extensive research in ELISA has been performed since its discovery in 1971 (Engvall and Perrnann, 1971) and the results demonstrate that ELISA methods are feasible and reliable for mycotoxin analysis in foods and feeds (Chu, 1986). As a consequence, a number of immunoassay kits have been developed and marketed in the United States as a tool for monitoring mycotoxins in foods and feeds (Table 1.7). Generally, commercial immunoassay kits have worked well both in laboratories and in the field (Azer and Cooper, 1991; Domer et al., 1993). Some have been evaluated and approved by several professional organizations including Association of Official Analytical Chemists (AOAC) International (Park et al., 1989 a,b). To facilitate evaluation and certification of rapid test kits used for safety screening of foods, the AOAC Research Institute has been set up recently (Pestka et al., 1995). In October, 1992, this organization signed a Memorandum of Understanding with the US. Department of Agriculture’s Federal Grain Inspection Service (FGIS) concerning the Test Kit Performance Testing Program (Pestka, et al., 1995). Aflatoxin test kits have been evaluated using FGIS protocols and certified to claim “Performance Tested in Accordance with Standards Established by FGIS for Test Kits to Detect Aflatoxin Residues in Grain and Grain Products”. More recently, two DON ELISAs have been similarly certified by FGIS. Although commercial immunoassay kits have been certified by FGIS, they must be evaluated critically prior to adopting the systems. Criteria for adoption of 58 Nm. 0.0.0903“. .0.0E0.03.. 0:3 0>..0...003O 0.00 00.0. 0.. 0.0 0.03.00 >..0...< .0 0.0.0903”. 100200.03”. .0000. «.030 0...... 02.050030 000. 0.30000 0.00 00.0 0.. 0.0 >>.2 H4.0.00 .0 0.xo.0..< 0000000 ..0..0000 00000. .0 .030.> 000. 0.30000 .E00 00.0 00 0.. 003.. ”(0:0 .0 0.6.0:... 000000.00 00.....00 N0 0.x0.0_.< 0.00-000: 0.000000 .030.> 000002.00 0.00 0.0. 0 0.0.0. .00 >0... €0.00 .0 0.x0.0_.< .0090 0..0 000. 00002. 0.00 00.0 00-0. 0.000 3.2 H{wim— 00000.0.00N -..- 000. 0000.5 0.00 00.0 000. 0.000 >>.2 €030 006. war -.- 000. 00003 .E00 000 00-0.. 0.00 3.2 H0.0...0 0.3.0.000 _.0..0000 00000. (030 .0 .030.> 000. 0000.... 0.00 00.0 00-0. 0.000 3.2 H(0:0 0.0.00E3n. 00.....00 0.00.30: ...0..0000 00000. <0...0 .0 .030.> 000. .000; 0.00 00.0 00-0. 0.000.. .55. €030 0.x0..0.0> _0>0.00< 00..0< .02“. 0 0.30000 .E00 00.0 070 0.0 >>s. H(0.0m. 00.x0.0..< .000.00-..0< .0.0E0.03.. 0.0.. 00.00.0030 2...... 00.0. 0. .0 0E3_00 3.2.... .5. 0.6.0.... .0>0.000 00.0.... ._0>0.00< 02.0.0. .020. 0.54. 00.....00 0.00.5100: 00.00.2030 0...... 02.00.0030 000. ..30000 0.00 00. n 0. 0. _. 0E3_00 >._0...< 00.x0.0..< 000.0.... 06.00.. 02.0.. .2: 0<0< 0.00 .08 0.00-000... 0.0.0000 .030.> 000009.00 0.00 00.0. 0 .00. .00 030 ”(030 00.6.0.3 00 .0. .0 0..< .00.30.E. .3. .00... .80. .0... 0.00EE00 co..00..00< >0000>000 0.0>_00< 02.00.00 .0000“. 0.0.0000. .0. .00.. 9.00. .._0 .0 02.000. 000. 003.. .0 00 0.00..0>0 00.06.0002 .0. 05. 000000200... 0.0.0.0000 N... 0.00... .., . ~05! 00‘.._... —o’ 59 ..0. . 3.00-80. 0.0.0 .02 00.05.50 .0... .x..8..0<.0 .. .. .0000-.~.-00. .0000 .80.... .00.... 0.000... ..0001000000. «003 .0... 090.00.... .000. 5.00. . ..0000-v0~-000 9.9.... 0.00.. 0.. 00.000. .050 00082 . .0500 00 00.00 00.00.00. -.- 20280000. 0026.0? 000 0.0.0 0..0...3E 0.00.00. 35. 000 0.2 .. 59.800000. 00.3 .<.2 .0._.>.0..000 0.005.. 0.0000006 00000. 0000.. ...2 0008. ..0 00.08005 000.000.... 0 ...03 0.00..0>0 .00..0E0 .0 00000.30 00 00000 0 02.00.06 0.00.00 003.00. .00 0000 0...... 0.0200... . 5.0280000. 5000000208600. 00.00.00. Sm ...0000003EE. 00.00-00.000 00.00.00. Sm. 000 (0.00 0.00.9030 0...... 02.0...0030 .000... .E00 00.0. 0. 0.000 0E3_00 0.0.0.... 00000.0.00N .. .00.0_0.00N 000. 0000.... 0.00 00.0 00 0.000 >>.2 H<0...0 00000.0.00N 000. 0000.... 0.00 00.0 00 000 >>.... H....0...0 008. «F 000. ..000... 0.00 00.0 om 0.000 >>.2 H<0...0 0.x0..0.o> .0000. (0:0 0...... 02.0...0030 000. 0000.... .E00 000 0v 00 >22 H0.0.00 ..2 00.0.0.2 0000.00 0.003.003 000. 000002.00 ..0000. (030 0...... .0>..0...0030 0.30000 0.00 000 0. 0.0 >>s. H....0...0 00.5.00... ...x0.0.0> -.- .0002. 0.00 00.. 0.. 0000 >>.>. U0.0.00 00000.0.00N . 0...... 00.. ov 00 >22 U0.030 ..2 0.06.0.0... 000. 000009.00 0 (030 .0000. 4.030 0...... .02.0...0030 0.30000 0.00 00.. 0v 0.0 >>s. H....0_..0 .0 0.5.0.0... 00.0000 0.0.0903: 0....> 02.0...0030 0.00 000. 0. 00 00.3.8 3.0.0.... 0.5.0.000 .. 0.0.0.000 0.00 000-000 0. 0.00 0.2 H0.0...0 0.00 00.00.... 0. 00. 0.2 H0.0...0 0.00 000-000 0. 0.0 0.2 H...0...0_ 0...... 00.00.... 0. 0.0 0.2 H<0...0 .5. 00.0.0.2 ..0..0000 .030.> 0.30000 0.00 000-000 0. 0.3-0.0 0.2 H{01.0 0008.00... ..000.00-N0 00.30.00 n..0. .00... .000. .0... 0.000.000 00..00..00< ..00000000 0.0200... 02.00.00 .0E.ou. 0.03.00... ..v. .00.. 003:..000 8.. 0.00... 60 a mycotoxin immunoassay include: detection limits and sensitivity ranges, requirements for rapid screening and/or quantitation, mycotoxin specificity, effectiveness of recommended extraction procedures, field stability, inter-and intra- assay reproducibility (Pestka, 1988). 61 PART II DEVELOPMENT AND APPLICATION OF AN ELISA FOR FUMONISIN 81 IN FUNGAL CULTURES, CORN AND CORN PRODUCTS ABSTRACT DEVELOPMENT AND APPLICATION OF AN ELISA FOR FUMONISIN 81 IN FUNGAL CULTURES, CORN AND CORN PRODUCTS By Sutikno Mouse monoclonal antibodies, rabbit and sheep polyclonal antibodies against fumonisin B1 (FB1 ) were readily generated after immunization with keyhole limpet hemacyanin (KLH) as a protein carrier. When these antibodies were used in competitive inhibition enzyme-linked immunosorbent assay (ELISA), the sheep antisera had the highest affinity and were the most specific to FB1 as compared to the mouse monoclonal and rabbit polyclonal antibodies. Cross reactivities of these sheep antisera toward fumonisin B1 , Bz , and 83 were 100, 24 and 30%, respectively. When competitive direct ELISA (CD—ELISA) employing these FB1 sheep antisera was used to detect F31 in Fusarium corn culture, corn and com products, it yielded approximately two fold higher FB1 levels than high performance liquid chromatography (HPLC). Its perfomance was much better than a monoclonal antibody based CD—ELISA that was previously developed in our laboratory. This CD-ELISA had a strong positive relationship with HPLC, suggesting that it would be useful for screening human and animal foods from fumonisins. 62 INTRODUCTION Background. Fumonisins are toxic secondary metabolites, found worldwide that are produced by Fusarium monilifonne and F. pmliferatum. These toxins are elaborated by the fungi in corn in the field and during storage (Bezuidenhout et al 1988; Gelderblom et al., 1988a). Six different fumonisins, fumonisin A1, A2, B1, 82, 33, and B4, have been chemically characterized(Figure 1.2) (Bezuidenhout et al., 1988; Cawood et al., 1991; Plattner et al., 1992). Fumonisin 8, (F81) is the most toxic and primary fumonisin produced by Fusarium moniIifonne (Gelderblom et al., 1988b). F81 produces characteristic effects in different species, including hepatic cancer in rats, brain lesions in horses (equine leukoencephalomalacia, ELEM), and pulmonary lesions in pigs (porcine pulmonary edema, PPE). Vifilson et al. (1985) observed hepatic cancer in rats that were fed with corn naturally contaminated with the Fusarium moniliforme for extended periods (up to 176 days). Marasas et al. (1984b) also found liver tumors in rats by feeding Fusarium monilifonne culture materials. Gelderblom et al. (1988b) performed experiments to investigate the toxic metabolite of Fusarium moniliforme. Through chromatographic procedures, these investigators identified the water-soluble fraction had potential tumor promoting capacity. Bezuidenhout et al. (1988) and Gelderblom et al. (1988b) isolated fumonisin compounds from 63 64 the water-soluble material and determined their structure. Similarly, Voss et al. (1989) developed a rat bioassay for hepatotoxic lesions that were produced by feeding Fusan‘um monilifonne cultures or naturally contaminated corn. They found that a water-extractable, rather than a chloroform-extractable one, was a causal agent of both hepatocellular carcinoma in rats and brain lesions in horses. ELEM is a neurotoxic disease that occurs only in Equidae and is characterized by multifocal liquefactive necrosis of cereme white matter (Marasas et al., 1988b). ELEM has also been demonstrated in separate experiments in which horses were administered purified FB1 either orally (Kellerman et al., 1990) or intravenously (Marasas et al., 1988b). Clinical signs begin with lethargy, head pressing and no appetite, and over a period of days progresses to convulsions and death (Ross et al., 1991). Other signs of intoxication are liver damage and altered serum sphingosinezsphinganine ratios (Wang et al., 1991). Plattner et al. (1990) identified F81 in extracts obtained from hepatotoxic and ELEM cases. PPE is an unusual disease in pig, which is characterized by recumbency and death (Norred and Voss, 1994). Outbreaks of PPE have occurred in Georgia (Colvin and Harrison, 1992), and several Midwestern states (Osweiler et al., 1992; Ross et al., 1992). These outbreaks were associated with the consumption of corn screenings that were contaminated with F. monilifonne (Harrison et al., 1990). When purified F81 is injected intravenously into pigs at a dose level of 0.40 mglkg body weight for 4 days, pigs die by day 5 and have lung edema and hydrothorax similar to pigs diagnosed with PPE. PPE does not occur when pigs are either injected with a smaller dose of F31 (0.17 mglkg body weight) or with FBz at a dose level of 0.30 65 mglkg body weight (Harrison et al., 1990). These observations strongly suggest that F81 is a causative agent of PPE. This finding has been confirmed by other investigators by feeding pigs with com contaminated with FB1 (Osweiler et al., 1992; Riley et al., 1993). Hascheck et al. (1992) reproduced PPE in pigs by injection of purified FB1. These investigators suggested a possible mechanism for induction of PPE where by F81 disrupts sphingolipid biosynthesis in the liver. This leads to damaged hepatocyte membranes, which are released into the bloodstream. These membrane fragments are phagocytized by pulmonary intravascular macrophages which then release enzymes and other mediators wpable of increasing mpillary permeability in the lung with subsequent edema. The fact that PPE has not been found in other species, perhaps because of much lower levels of pulmonary macrophages in other species (Winkler, 1988). Correlations between Fusarium monilifonne food contamination and human esophageal cancer have been reported in South Africa (Marasas et al., 1988a) and in China (Cheng et al., 1985) suggesting that fumonisins are a possible etiologic agent for this disease. Sydenham et al. (1990) surveyed corn from areas with high and low rates of human esophageal cancer in Transkei, South Africa. Samples taken from the high rate cancer area had higher levels of Fusarium monilifonne and fumonisins. The authors suggested that consumption of corn contaminated with Fusan‘um moniliforme may be a factor in the high rate of human esophageal cancer. 66 Analytical methods Commonly used methods for detecting and quantitating fumonisins are thin layer chromatography (TLC), high performance liquid chromatography (HPLC), and mass spectroscopy (MS). Fumonisins do not absorb ultraviolet light and do not fiuoresce; therefore, derivatization reactions are required before detecting the toxins with chromatographic techniques. In order to visualize fumonisins on TLC plates, Gelderblom et al., (1988a) and Cawood et al. (1991) used 0.5% p- anisaldehyde which was able to react with amino group of fumonisins for generating reddish purple compounds when heated to 120°C. Retention factor (Rf) values of F81 and F82 developed on normal phase silica TLC (Merck, Art. 5554) in chloroform:methanol:acetic acid (60:35:10) are 0.32 and 0.52, respectively (Ackerman, 1991). By spraying with fluorescamine, Rottinghaus et al.(1992) visualized F81 and F8; on reverse-phase C18 TLC plate developed in methanol:4% aqueous KCI (3:2). They observed the toxins under longwave ultraviolet light. F 81 and F B: appeared as bright yellow—green fluorescent bands at Rf values of 0.5 and 0.1, respectively. The detection limit of this procedure was 0.1 ppm in corn and its recoveries from spiked corn samples averaged more than 80%. In general, TLC methods are qualitatively useful and simple but they are not accurate for quantifying fumonisins. More accurate and lower detection limits for fumonisin quantification can be achieved using HPLC which can be performed via formation of a maleyl derivative that absorbed at 250 nm (Gelderblom et al., 1988a). The maleyl derivative procedure was initially developed by Siler and Gilchrist (1982) for 67 analysis Altemaria altemata f. sp. Lucopersici (ALL) toxin which had a chemical structure similar to fumonisins. Briefly, the toxin is dissolved in 0.1 M sodium carbonate solution pH 9.2, and then maleic anhydride crystals are slowly added over a period of 5 minutes. At least a 10:1 molar ratio of maleic anhydride crystals to toxin is used in this procedure to ensure complete derivatization. Prior to HPLC analysis, the maleylation mixtures are adjusted to pH 6-7 by dilution with the mobile phase, 0.05M KH2PO42methanol (3:7, pH 3.5), or by addition of hydrochloric acid. This procedure is good for detecting fumonisins in corn cultures with Fusan’um monilifonne , which generally have a high (> part per thousand, ppt) level of the toxins. lt, however, does not work for detecting fumonisins in naturally contaminated corn (Norred, 1993). HPLC sensitivity can be increased by formation of fumonisin-fluorescent derivatives with o—phthaldialdehyde (OPA)(Shephard et al., 1990). OPA derivatization should be performed between 1 and 2 minutes prior to HPLC analysis due to the CPA—derivative stability (Shephard et al., 1990). Detection limits of this procedure are approximately 50 nglg for F81 and 100 nglg for F82. Scott and Lawrence (1992) developed a liquid chromatographic method using 4- - fluoro-7-nitrobenzofurazan (NBD-F) which yielded moderately stable derivatives and had detection limits of 1 nglg of F81 and F82. However, derivatization with NBD-F required many steps and was time consuming. Gas chromatography-mass spectrophotometry (GC-MS) has also been applied to detecting fumonisins in corn and feeds (Plattner et al 1990). In this method, an extract of sample is hydrolyzed in 1N potassium hydroxide at 60°C for 68 1 hour followed by acidification with 0.5N HCl to form aminopentol backbones which are then analyzed with GC-MS. GC-MS is highly selective and sensitive although it requires relatively expensive equipment. Regardless of sensitivity and selectivity, HPLC and GC-MS methods are expensive, complex, and time consuming. As an alternative to these methods, immunoassays have already been proven to be useful in screening for other mycotoxins (Pestka, 1988). Fumonisin antibodies Previously, both mouse polyclonal and monoclonal antibodies against fumonisin 81, 82 and 83 were produced in our laboratory by using cholera toxin (CT) as carrier and adjuvant (Azcona-Olivera et al., 1992a,b). Concentrations of fumonisin 81, 82 and 83in buffer solution required for 50% binding inhibition were 260, 300, and 650 nglml respectively for polyclonal antibodies with Cl-ELISA, and 630, 1800, and 2300 nglml, respectively for monoclonal antibodies with CD- ELISA. Recently, antibodies against fumonisins have been produced by several other investigators. Fukuda et al. (1994) generated murine monoclonal antibodies against F81 by using ovalbumin and keyhole limpet hemacyanin (KLH) as a protein carrier. When these monoclonal antibodies were used in a Cl-ELISA, the amount of F81 required for 50% inhibition ranged from 65 to 255 nglml. F81- rabbit polyclonal antibodies have also been produced using a F81-KLH conjugate (Usleber et al., 1994). When these rabbit antibodies were used in CD-ELISA, 69 only 0.6 ng/ml'of F8, was required to inhibit 50% of F81 peroxidase conjugate binding to the solid-phase-bound antibodies. Rationale. An HPLC procedure approved by AOAC and IUPAC after throughout collaborative studies has been the primary method used to detect F81. This method, however, requires laborious sample extraction, cleanup, and elusions, before finally detection. Several ELISA approaches have also been developed to date (Azcona-Olivera et al., 1992a,b; Pestka et al., 1994; Tejada-Simon, 1994; Fukuda et al., 1994; Usleber et al., 1994). The ELlSAs are preferable to HPLC because of their simplicity, ease of sample preparation, and use of stable reagents (El-Nakib, et al, 1981). However, detection of F81 in food samples by CD-ELISA technique using monoclonal antibodies produced by Azcona-Olivera (1992b) provided much higher (30 fold) fumonisin estimates than GC-MS or HPLC method (Pestka et al., 1994). The CD-ELISA yielded even higher(up to 400 fold) fumonisin estimates in Fusarium moniliforme cultures (T ejada-Simon, 1994) than that for HPLC. One possible explanation for these observations is that the F81 antibody generated by using cholera toxin (CT) as the carrier-adjuvant is not completely specific to fumonisins and, therefore, may have reacted with other compounds found in Fusarium moniliforme cultures and contaminated grains. To overcome these problems, it is desirable to produce antibodies which have higher affinity and specificity to the toxins. By using KLH as a protein carrier, higher affinity and specificity rabbit polyclonal antibodies against F81 could be generated 70 (Usleber et al., 1994). In addition, by utilizing hybridoma technique, monoclonal antibodies which have higher specificity and affinity could be generated. Based on the early findings, l hypothesized that use of KLH as a protein carrier, polyclonal approaches and hybridoma techniques could result in the development of higher affinity F81-specific antibodies. Specifically, the purposes of this study were to: (1) produce both polyclonal and monoclonal fumonisin antibodies using KLH as a protein carrier; (2) compare the applicability of these antibodies in ELISA; (3) apply the optimal ELISA techniques for detecting F81 in Fusarium corn cultures, corn and corn products; and (4) to compare the optimized ELISA to the existing reference HPLC method. MATERIALS AND METHODS Chemical and reagents All organic solvents and inorganic chemical were of reagent grade or better. Ovalbumin (OA) (chicken egg albumin grade III; fraction Vll), Tween 20, 2,2’-azinobis(3-ethylbenzthiazolinesulfonic acid)(A8TS), hydrogen peroxide, horseradish peroxidase (HRP)(fraction VI), sodium borohydride, glutaraldehyde, penicillin/streptomycin solution (pen/strep) (100,000 units/ml), sodium pymvate, polyethylene glycol (MW 1450) (PEG), hypoxanthine, aminopterin, thymidine, pristane, and dimethyl sulfoxide were purchased from Sigma Chemical Co. (St. Louis, MO). Bovine serum albumin (BSA)(Albumin, bovine fraction V) was obtained from Amresco (Solon, Ohio). Dulbecco’s Modified Eagle’s Medium (DMEM), NCTC supplemental medium, and fetal bovine serum (F88) were obtained from Gibco Laboratories (Grand Island, NY). Tissue culture plasticware was purchased from Corning Laboratory Science Co. (Corning, NY). The myeloma cell line P3INS1l1-Ag4-1 (NS 1) (ATCC TIB 18) was purchased from the American Type Culture Collection (Rockville, MD). Macrophage conditioned media (MCM) was prepared as described by Sugasawara et al. (1985). Sheep polyclonal antibodies used in this study were developed jointly by our laboratory and Neogen Corp. (Lansing, MI). 71 72 Conjugation of fumonisin B1 to protein Fumonisin 81 (F81) was conjugated using glutaraldehyde to KLH (Pierce, Rockford, IL) for use as immunogen and to ovalbumin (fraction Vll) for use as a solid-phase antigen for indirect ELISAs by the method of Avrameas and Temynck (1969). The conjugation reaction was carried out at 4°C in 0.01M phosphate- buffered (pH 7.4) saline (PBS). F 81 was added to 1 mglml suspension of carrier protein at a protein/toxin weight ratio of 5:1 for FBi-KLH, and of 3.411 for F 81-OA. An equal volume of 2% (vol.lvol.) glutaraldehyde in water was then added dropwise with constant stirring. After 1 hour, the reaction was stopped by adding sodium borohydride to a final concentration of 10 mglml. One hour later, the mixture was dialyzed for 72 hours (three changes) against 0.01M PBS pH 7.2. Both KLH and OA conjugates were aliquoted in fractions of 1 mg (total protein), lyophilized, and stored at -2o°c. F81 was conjugated to horseradish peroxidase (F81-HRP) at an HRP/F81 weight ratio of 8:1 and used in the competitive direct ELISA by the periodate method (Nakane and Kawaoi, 1974). Two ml of HRP solution (8 mglml distilled water) was reacted with 400 pl of fresh 0.1M sodium periodate (0.21 9 I10 ml distilled water) for 20 minutes at room temperature to activate the HRP. The solution was then dialyzed twice against 3 liters of 1mM sodium acetate buffer pH 4.4 overnight at 4°C. Activated HRP was added into the F81 solution (2 mg F81 in 200 pl of 50% acetonitrile in water) and 20 pl of fresh 1M sodium carbonate (1.06 gl10 ml distilled water) was added dropwise to the solution. The conjugation reaction was carried out for 2 hours at room temperature and then 200 pl of 73 sodium borohydride (4 mglml distilled water) was added dropwise. After incubation for 2 hours at 4°C, the conjugate was dialyzed twice against 6 liter 0.85% (wt/vol.) sodium chloride overnight, lyophilized in small portions, and stored at -20°C. Rabbit immunization Three Female New Zealand rabbits (Charles River Laboratories, Wilmington, MA) were initially given a ten site subcutaneous (sc) injection with F81-KLH conjugate. This injection consisted of 1.0 ml of the conjugate (0.5 mg F81-KLHIml) in a 1:1 ratio of saline and Freund’s “complete“ adjuvant (Difco Laboratory, Detroit, Michigan). Two weeks later, the rabbits were boosted subcutaneously with 0.5 ml (0.5 mglml) of the conjugate in saline and Freund’s “incomplete” adjuvant (Difco Laboratory, Detroit, Michigan) in the ratio of 1 :1(vol.lvol.). Every other week after the second injection, rabbits were bled from lateral marginal earn vein. Two weeks after the fifth bleeding, rabbits were given the second boost as the first one and two weeks later, the animal were sacrificed and their blood was collected. Serum was obtained after overnight incubation of blood at 4°C and centrifugation at 1,000 x g for 15 min. Rabbit immunoglobulins were purified by 33 percent saturation with ammonium sulfate (Hebert et al., 1973). Serum titer and antibody specificity were then determined by ELISA as described below. 74 Hybridoma production Eight female BALB/c mice (6 to 8 weeks of age, Charles River Laboratories, Wilmington, MA) were immunized by intraperitoneal (i.p.) and subcutaneous (s.c.) routes. Mice were immunized three times with F81 -KLH conjugates at two-week intervals. The first immunization consisted of 200 pl (0.25 pg F81-KLHI pl) of conjugate in a 1:1 (vol.lvol.) ratio of PBS and Freund’s complete adjuvant. For the second and the third injections, mice received 5-50 pg F81-KLH conjugate which was mixed with an equal volume of Freund’s incomplete adjuvant for ip injection or with an equal volume of saline for sc injection. Ten days after the second and the third immunizations, ether- anesthetized mice were bled from the tail vein and the blood was collected with a heparinized tube. The blood was incubated at 4°C overnight and then centrifuged at 1000 x g for 15 minutes to obtain mouse plasma. Titer and antibody specificity were then determined by both direct and indirect ELISAs. Mice producing F81- specific and high titer antibody were chosen for fusion. At 4 days prior to the fusion, the mice were injected intravenously (iv) in the lateral tail vein with 4 pg of F81-KLH conjugate in saline. Hybridoma production was performed by a modification of the procedure of Galfre and Milstein (1981). Spleen cells (1 x 10") from an immunized mouse were fused with NS-1 myeloma cells (1 x 107) using PEG. Fused cells were resuspended in 100 ml of complete DMEM medium supplemented with 1% NCTC (vol.lvol.), 10 mM sodium pyruvate, 100 units/ml of pen/strep solution, 20% (vol.lvol.) FBS, and 20% (vol.lvol.) MCM, and then distributed into 875 wells of 75 96-well plates and incubated for 24 hours at 37°C in a humid atmosphere of 5% CO2 in air. One half of the supernatant from each well was removed and replaced with an equal volume of HAT medium (complete medium with hypoxanthine, aminopterin, and thymidine). This operation was repeated every 3 days during a 2 week period, after which time HAT medium was eliminated gradually and replaced by HT medium (the same composition of HAT but without aminopterin). Supematants of hybridoma cultures were tested for the presence of F81 specific antibody by Cl-ELISA. Cultures that produced a F81 specific antibody were successively scaled up and cloned by limiting dilution at 0.5-1 cell/well (Goding, 1980). Subclones yielding desirable antibody activity were then isolated and stored in FBS-dimethyl sulfoxide (9:1) under liquid nitrogen. Mass production of F81 monoclonal antibodies was done by expansion of the selected subclones. Antibodies were purified and concentrated from cell-free culture supematants by precipitation with 50% saturated ammonium sulfate (Hariow and Lane,1988) For large scale production of monoclonal antibodies in vivo, mice were injected with 0.5 ml of pristane (2,6,10,14 tetramethylpentadecanoic acid) intraperitoneally (Potter, 1972). After 7-10 days, the mice were immunized with 5x105 to 5x10‘5 hybridoma cells from an actively growing culture which had been centrifuged and resuspended in 0.5 ml 20% F BS-DMEM. After mouse-abdominal swelling (usually 7-10 days after hybridoma injection), ascitic fluid was tapped with an 18 gauge needle, and then purified by 45-50% saturation of ammonium sulfate (Harlow and Lane, 1988). 76 Indirect ELISA Indirect ELISA was performed by a modification of the procedure of Azcona-Olivera et al. (1992a) and used to determine serum titers. Briefly, wells of polystyrene microtiter plates (lmmunolon 4, Dynatech Laboratories, Alexandria, VA) were coated overnight (at 4°C) with 100 pl of F81-OA (5 pglml) in 0.1 M sodium carbonate-bicarbonate buffer (pH 9.6). Plates were washed six times by filling each well with 300 pl of 0.02% (vlv) Tween 20 in PBS (PBS-Tween) and aspirating the contents. Nonspecific binding was blocked by filling the wells with 300 pl of 1% (wt/vol.) bovine serum albumin in PBS 0.01 M (BSA-PBS), pH 7.2. After incubating for 30 minutes at 37°C, the plate was washed six times with PBS-Tween. Fifty pl of serially diluted mouse serum was added to each well and incubated at 37°C for 1 hour. Wells of serially diluted preimmune serum were used as controls. Unbound antibodies were removed by washing six times with PBS-Tween, and 100 pl of goat anti-mouse lgG peroxidase conjugate (2 pglml, in BSA-PBS, Cappel Laboratories, West Chester, PA) was added to each well. The plate was incubated for 30 minutes at 37°C and washed eight times with PBS- Tween. Bound peroxidase was determined with ABTS substrate (1 ml of 35 mg ABTS/15 ml distilled water mixed with 11 ml of citrate buffer pH 4 and 8 pl hydrogen peroxide) as described previously by Pestka et al. (1982). Absorbance at 405 nm was read with a Vmax Kinetic Microplate Reader (Molecular Devices Corporation, Menlo Park, CA). Titer of each serum was arbitrarily designed as 77 the maximum dilution that yielded twice or greater absorbence as the same dilution nonimmune control serum. A competitive indirect ELISA (Cl-ELISA) was used to verify specificity of antibodies in sera toward F 81 during the course of immunization and to determine those tissue culture wells which contained hybridomas secreting desirable antibody, following fusion and cloning. Briefly, microtiter plates were coated and blocked as described in the indirect ELISA procedure, and then 50 pl of standard F8, (0 - 5000 nglml) dissolved in PBS was simultaneously incubated with 50 pl of appropriate dilution of antibody or culture supernatant over the FBi-OA solid phase for 1 hour at 37°C. The assay was then completed as described above. Direct ELISA Direct ELISA was used to determine the titer of animal sera (Azcona- Olivera et al., 1992a). Plates were coated (125 pllwell) with serial dilution of preimmune and immune sera in 0.1 M sodium carbonate-bicarbonate buffer (coating buffer pH 9.6), then incubated overnight by drying at 40°C in a conventional oven. After washing and blocking, 50 pl of F8. -HRP (2 pglml, in BSA-PBS) was added to each well. After 1 hour of incubation at 37°C, plates were washed and bound peroxidase was determined as described above. A competitive direct ELISA (CD-ELISA) was used for F81 detection in fungus cultures, corn and corn products. Briefly, microtiter plates with 96 wells were coated and blocked as described in the direct ELISA, and then 50 pl of serially (0 to 25 nglml) diluted F81 standard in 10% methanol or of appropriately 78 diluted samples in 10% methanol were simultaneously incubated with 50 pl of HRP-F81 (2 pglml in 1% BSA-PBS) for one hour at 37°C. The assay was then completed as described in the indirect ELISA. Veratox® Veratox® (Neogen Corp, Lansing, MI) is an ELISA kit for fumonisin test based on CD-ELISA and was developed jointly between our laboratory and Neogen Corp. Veratox® was used to detect F 81 in corn and corn products and performed according to its instructions. Briefly, 100 pl of serially diluted F81 standard (0-20 pglml) or diluted samples was put into red-marked microtiter wells and then simultaneously added with 100 pl of conjugate solution from the blue- labeled bottle. This solution was mixed by pipetting the liquid up and down 3 times using an 8 or 12 -channel pipetor. One hundred pl of the liquid mixture was transferred into coated-antibody micro titer wells and incubated for 15 minutes at room temperature. Every 30 seconds, the wells were swirled back and forth carefully to mix the solution. After 15 minutes, the solution was decanted. The wells were washed 5 times by filling/decanting with distilled water, and then added 100 pl substrate solution from the green-labeled bottle to develop color. The color development was then stopped by addition of 100 pl of stop reagent from the red-labeled bottle. Absorbance at 650 nm was read with a Vmax Kinetic Microplate Reader (Molecular Devices Corporation, Menlo Park, CA). 79 ELISAGRAM ELISAGRAM was used to determine F83 and other compounds in food and culture samples which are also bound to FBi-antibodies. It was performed as described by Pestka (1991) with modification. Briefly, fumonisin B1 in samples was separated using silica gel 60 thin layer chromatography (Kieselgel 60, 0.063- 0.200 mm, Merck, 'SA) with chloroformzmethanolzacetic acid (60:30:10) as developing solvents. Nitrocellulose membranes (0.45 pm; Schlicher and Schuell, Keene, NH) were coated with F81 antibodies by soaking them in F81 antibody solution over night. The TLC plate was sprayed with PBS and then blotted with the FBi-antibody-coated nitrocellulose (NC) membranes. The AB-coated NC membranes were then soaked in FB1-HRP solution (2pglml in 1% BSA-PBS) and incubated at room temperature for 10 minutes. NC sheets were washed with PBS containing 0.2% Tween 20, and then rinsed briefly in distilled water. For color development, the NC sheets were incubated for 10-20 minutes at 25°C with 10 ml of dioctylsodium sulfosuccinate (DONS)I3,5,3’,5’-tetramethylbenzidine (TMB) substrate prepared as described by Koch et al. (1985). The reaction was stopped by washing the NC in distilled water and then incubating for 10 minutes in stopping reagent solution (100 mg DONS dissolved in 13 ml of ethanol and made up to 50 ml with distilled water). After drying between filter paper, the white spots on the NC indicating the presence of F81 toxin in the samples were observed. 80 Fusarium cultures Fusarium cultures used in this study were M-5986 F. moniliforme, isolated from samples that produced PPE (Ross et al., 1990); and M-5982 F. moniliforme, isolated from samples that produced ELEM (Ross et al., 1990). In addition, F. gramineamm W-8, which did not produce fumonisins, but rather deoxynivalenol and zearalenone (Marasas et al., 1984a), was used as a negative control. These cultures were obtained from the Fusarium Research Center at Pennsylvania State University (University Park, PA). Fusarium monilifomie cultures were inoculated onto petri dishes with V-8 juice agar medium (Stevens, 1974). Each liter of V-8 juice agar medium contained 200 ml of V-8 juice, 3 g of calcium carbonate, and 20 g of agar (Jackson and Bennet, 1990). These plates were incubated for 10-14 days at room temperature on an alternating light-dark schedule. To prepare spore suspensions, a loopfull of conidia from F. monilifomie petri dish cultures was transferred to V-8 juice agar slant-tubes which were then incubated at room temperature for 10-14 days on an alternating light-dark schedule. Spore suspensions were obtained by washing the slant tubes with sterile distilled water. Unlike Fusarium monilifonne spores, conidial suspensions of F.graminearum W-8 were prepared by transferring the cultures into an ,150—ml Erlenmeyer flask containing 40 ml of autoclaved CMC medium (15 g of carboxymethyl cellulose, 1 g of ammonium nitrate, 1 g of potassium phosphate, 0.5 g of magnesium sulfate heptahydrate, 1 g of yeast extract in 1 liter of distilled water) (Stevens, 1974). The flasks were then incubated at 25°C with shaking at 81 220 rpm. Cultures were checked between 3 and 5 days for macroconidia production. Conidial suspensions were obtained by filtering CMC medium through 4 layers of sterile cheesecloth to remove mycelia. Prior to inoculation, 250-ml Erlenmeyer flasks containing 40 g of ground corn and 11 ml of distilled water were autoclaved for 30 min. After autoclaving, an additional 11 ml of sterile distilled water containing 107 conidia was added (Stevens, 1974). The flask cultures were incubated in the dark at 25°C for 14 to 28 days. Corn cultures were extracted with 5 ml/g of acetonitrilezwater (1:1) by soaking for 2-3 hours with mixing every half hour (Plattner et al., 1992). Suspensions were filtered through Whatman # 1 filter paper (Whatman Ltd., Maidstone, UK) and the filtrate was tested for fumonisin 81 by ELISA and HPLC. Com-sample extraction and clean up Com extraction was performed by a modification of the procedure of Thiel et al. (1993). Food, feed, or fresh corn samples were finely ground with Waring Blender Model 1042 (VVinsted, Connecticut). A subsample (5 g) was put in 50 ml plastic-conical tubes, and 25 ml of 75% (vol.lvol.) methanol in distilled water . was added. The tubes were then shaken (American Rotator V, R4140) at 200 rpm for 20 minutes. The solution was centrifuged at 10009 for 10 minutes, and filtered through Whatman # 4 filter paper (Whatman Ltd., Maidstone, UK). The filtrates were diluted with 10% methanol at ratio of 1:40 or higher before analyzing for fumonisin 81 with CD-ELISA and Veratox® (Neogen Corp., Lansing, MI, 48912). 82 For some experiments, filtrates were cleaned up by passage through a strong anion exchange (SAX) column (Bond-Elute SAX cartridges, 3 cc capacity containing 500 mg sorbent; Varian, Harbor City, CA 90710) before analyzing with HPLC. Briefly, pH of the filtered extract was checked and adjusted, if necessary, with 0.1 M KOH to pH 5.8-6.5 before running through the SAX column. SAX columns were attached to 16-port vacuum manifold (Alltech, State College, PA) and conditioned by washing successively, first with 5 ml methanol and then with 5 ml of 75% (vol.lvol.) methanol in water at flow rate of no more than 2 mllmin. While maintaining the flow rate, 10 ml of the substrate was applied to the column. The column was then washed with 8 ml of 75% methanol (vol.lvol.) in water, followed by 3 ml methanol. Fumonisins were eluted with 10 ml of 1% (vol.lvol.) acetic acid in methanol, at flow rate of no more than 1 mllminute (at atmospheric pressure). The eluent was collected in 15 ml conical tubes. One half of that (5 ml) was evaporated in 4 ml capacity vials under stream of nitrogen at about 60°C. The vial was then capped and stored in 4°C until HPLC analysis. Prior to F81 detection with HPLC, the purified sample residue in the 4 ml vial was redissoved with 100 pl methanol. HPLC Measurement of F 81 by HPLC was performed according to the previously described reference method (Shephard et al., 1990 and Sydenham et al., 1992). The liquid chromatography system used in this study consisted of lsco Model 83 2300 HPLC pump with an injector valve (Valco valve)(Lincoln, NE); H-S3 C13 #316 stainless steel packed 013 column (Perkin Elmer, Norwalk, Co) 0258-0178 reverse phase (3.30m); Hewlett Packard model HP 3392A integrator (Avondale, PA); Linear Instruments Fluor fluorescence detector LG. 304 (Reno,NE) fitted with a 3.1 pl flow cell, 500 psi (34 atm) maximum pressure and set at 334 nm (excitation) and 440 nm (emission) and slit widths of 12 nm or similar. The mobile phase was methanol:0.1M sodium dihydrogen phosphate (13.89 NaH2PO4.H2O in 1 liter distilled water)(66:34) that adjusted to pH 3.4 with ortho- phosphoric acid and filtered through 0.22 pm water GV membrane (Milipore Corporation, Bedford, MA). Before running the mobile phase, the C13 column was washed approximately for 2 hours with degassed absolute methanol at a flow rate of 1.0 mllminute and then with 25% (vol.lvol.) methanol in water for approx. 2 hours at the same flow rate. The mobile phase was pumped at 1.5 mllminute when analyzing samples. After analyzing all samples, the column was washed with 25% (vol.lvol.) methanol in water then with absolute methanol for approximately two hours each at a flow rate of 1.0 mllminute. OPA derivatizing reagent was prepared by dissolving 40 mg O- phthaldialdehyde in 1 ml methanol and diluted with 5 ml 0.1 M sodium borate (3.8 g Na2B4O-, in 100 ml dH2O) and 50 pl of 2-mercaptoethanol. The solution was stored no more than one week at room temperature in the dark. Purified sample residues in a 4 ml vial were dissolved in 100 pl methanol. Fumonisin standards were serially diluted (0.5 to 100 pglml) in methanol. Twenty five pl fumonisin standard or samples were transferred to the base of small test 84 tubes and mixed with 225 pl OPA reagent. Less than 2 minutes after mixing, 10 pl of the mixture was injected into the HPLC system. Statistics Linear regression analyses were used to correlate ELISA and HPLC data. A linear correlation coefficient (r) equaling to 1 indicates there is a perfect positive relationship between the ELISA and HPLC methods, and rclose to -1 indicates a strong negative correlation; whereas, a correlation coefficient close to 0 indicates no relationship between two variables (Steel and Torrie, 1980). The P value is the probability of being wrong in concluding that there is a true association between the variable. The smaller the P value, the greater the probability that the variables are correlated. Sigma Plot® (Scientific Graph System, Version 1.00 for Windows, Jandel Scientific, San Rafael, CA) was used to perform linear regression analyses. RESULTS AND DISCUSSION Rabbit polyclonal antibodies Three rabbits were immunized and boosted with FB1-KLH immunogen. Each rabbit was bled from a lateral marginal ear vein and its serum was prepared and stored. Titers of the rabbit polyclonal antibodies in a direct ELISA format were much lower (more than 3,200) as compared to that in an indirect ELISA format (more than 64,000) (Figure 2.1). Concentrations of F81 required for a 50% inhibition of antibody binding were much lower (approximately 100 nglml) in CD-ELISA than that in Cl-ELISA (more than 10,000 nglml) (Figure 2.2). In a direct ELISA format, antibodies were bound to a solid phase and then enzyme- labeled antigen (FB1-HRP) was added to detect and Quantify the antibodies. Since F81 was conjugated to HRP directly without using glutaraldehyde as “a bridge”, it is possible that FB1-HRP conjugate was reacted with only F 81 specific antibodies. On the other hand, unlike in FB1-HRP conjugation, glutaraldehyde was used as “a bridge” in both F81-OA and FBi-KLH conjugations. Therefore, glutaraldehyde-0A might have acted as an epitope for glutaraldehyde specific antibodies to produce non-specific binding as well as specific antibodies binding to F81. If this happened, then both specific and non-specific binding between rabbit antisera and the coating protein would be detected upon addition of the second enzyme-labeled antibodies (goat-anti-rabbit lgG-HRP). In that case, an 85 86 indirect ELISA format would yield greater color development, and subsequently higher titer than the direct ELISA. However, F81 required to inhibit 50% of the antibody binding may be much more higher in Cl-ELISA as compared to that in CD-ELISA. Mouse immunization and monoclonal antibodies Four mice were injected intraperitoneally and the other four were immunized subcutaneously with FBi-KLH conjugates. All immunized mice produced F81 specific antibodies as early as 4 weeks after initial exposure of F 81- KLH immunogen. Competitive inhibition ELISAs for F81 using mouse antisera were determined with both Cl- and CD-ELISA formats (Figures 2.3 and 2.4). Like F81 rabbit antibodies binding, mouse antiserum binding was inhibited by F81 to a greater extent in CD-ELISA than in Cl-ELISA. CD-ELISA results were used to select optimal mice for fusion. Mice were sacrificed and their spleen cells were fused with myeloma (NS1) cells. The fused cells were then cultured into 875 wells of 96-well plates. Hybridomas were detected in 821 wells, indicating a fusion efficiency [(number of wells with growing colonies/number of wells seeded) x 100%] of 94 %. This fusion efficiency was similar to that observed by Azcona-Olivera et al. (1992b). After screening with CI-ELISA using F81-BSA coated plates, more than 20 wells were identified as FB1-antibody producers. Of these, seven cell lines (A703, 8103, B1G5, 82F7, B406, B4E3, B4611) remained stable during scale- up prior to cloning. Cl-ELISA standard curves of these cell lines are shown in 87 Figure 2.5. Concentrations of F81 required for a 50% inhibition of antibody binding ranged from 90 to 2000 nglml. The two best cell lines (82F7, B4D6) were cloned by limiting dilution twice and from these cultures, four cell lines (N205, Q1C5, QZC9, R185) were chosen for large scale antibody production in ascitic fluid. Mice were injected with activer growing hybridoma cells (N2C5, Q1C5, 0209, R185) after intraperitoneal injection with pristane. After mouse-abdominal swelling, ascitic fluids of F81 -monoclonal antibodies were tapped and purified. Titers and competitive inhibition using these antibodies were then analyzed with both CD- and Cl-ELISA. Titers exceeded 40,000 with CI-ELISA and were between 500 to 1,000 with CD-ELISA (Figure 2.6). Concentrations of F81 required for a 50% inhibition of antibody binding for the clones ranged from 50 to 100 nglml with CI-ELISA and from 400 to 800 nglml with CD-ELISA (Table 2.1). Comparison among different antibodies F81 Rabbit polyclonal and N2C5 monoclonal antibodies were compared to the 205 monoclonal antibody which was previously generated by Azcona-Olivera (1992b) using F 81 cholera immunogen and to sheep antisera developed against FB1-KLH conjugate (Neogen Corp., Lansing, MI). As previously noted, CD-ELISA for F81 using rabbit polyclonal antibodies was more sensitive than CI-ELISA (Figure 2.2). However, the opposite was true for the monoclonal antibodies (Figure 2.7). In order to ascertain which F81 antibodies had the highest avidity, CD-ELISA standard curves for F81 polyclonal antisera and CI-ELISA standard curves for F81 monoclonal antibodies were 88 generated (Figure 2.10). Concentrations of F8, required for 50% inhibition of antibody binding were 6, 80, 100, and 200 nglml for sheep, rabbit, N2C5, and 205, respectively (Table 2.2). Thus, the sheep antisera contained F81 specific antibodies with the highest affinity. These results support the hypothesis of this study [use of KLH as a protein carrier, polyclonal approaches and hybridoma techniques could result in the development of higher affinity F81-specific antibodies than the 205 antibodies which were previously produced by Azcona- Olivera et al. (1992a,b) in our laboratory]. The affinity of F81 polyclonal antisera was higher than that of the monoclonal antibodies (Table 2.2). This observation may be related to several possibilities. Firstly, when producing monoclonal antibodies, hybridomas secreting F81 antibodies with lower affinity, instead of with higher affinity, might be chosen during hybridoma screening. Secondly, hybridomas secreting higher affinity F81 antibodies might not have been stable and died before screening. Thirdly, the affinity of F8. antibodies might be affected by animal species since sheep, rabbits and mice produced F81 antibodies with different affinities. This current study result agreed with the findings of Usleber et al. (1994) and Fukuda - et al. (1994) who also generated F81 antibodies using KLH as a protein carrier. The former researchers produced FB1-rabbit polyclonal antibodies that required only 0.6 nglml F81 for 50% inhibition with CD-ELISA. Meanwhile, the latter researchers generated F81 monoclonal antibodies that required a higher concentration of F81 (65 nglml) for 50% inhibition with CI-ELISA. 89 Since the F81 sheep antisera contained FB1-specific antibodies with the highest affinity, they were utilized in CD-ELISA for detection of F81 concentrations in fungal corn cultures, corn and corn products. CD-ELISA standard curves for F81 , F82 and F83 using these sheep antisera were graphically shown in Figure 2.11. Cross reactivities of the sheep antibodies towards F81, F82, and F83 [determined as (nglml of F81 required for 50% inhibition)l(ng/ml of fumonisin analogue required for 50% inhibition) x 100] were 100, 24 and 30%, respectively (Table 2.3). Detection of FB1-like compounds Several investigators reported that ELISA methods tended to give higher F 81 estimates than HPLC methods. Minervini et al. (1992) found ELISA methods yielded higher F81 estimates than HPLC when applied to Italian feed samples. Elevated estimates for F81 in feeds by ELISA as compared to HPLC have also been found in foods (Pestka et al., 1994) and in Fusarium corn cultures (Tejada- Simon, 1994). The latter investigator reported that ELISA methods yielded much higher F81 estimates (up to 400 times) than HPLC. She proposed that one reason for the different F 81 estimates between the two analytical methods might have resulted from the presence of compounds structurally similar to F81 in sample extracts which were detectable by ELISA, but not by HPLC. Detection of the F81-Iike compounds in corn and corn products might be performed by ELISAGRAM. F81 monoclonal antibodies (205) produced by Azcona-Olivera et al., (1992b) and F81 -sheep antisera developed jointly by our 90 laboratory and Neogen Corp. (Lansing, MI) were used in the ELISAGRAM. Before analyzing samples, a standard of F81 (2.5 pglml) was used to test whether the ELISAGRAM system worked or not for F81 toxin. Twenty pl of the F81 standard was directly spotted on silica gel thin layer chromatography, and then transferred onto FB1-antibody-coated nitrocellulose membrane (NC). The NC was incubated with F 81 -HRP, washed and finally incubated in dioctylsodium sulfosuccinate (DONS)I3,5,3’,5’-tetramethylbenzidine (T MB) substrate for color development. However, a white dot on the NC did not appear. This indicated that F81 standard did not bind to a coating antibody, so the coating antibody reacted with FB1-HRP which subsequently produced green color after incubation in HRP-substrate solution. Similar observations occurred when 10 pg F81 standard was directly spotted onto the NC. Thus, the ELISAGRAM that worked well for zearalenones and aflatoxins (Pestka, 1991) did not work at all for F81 toxin. Since F81 easily dissolved in water, the toxin might have run off during washing or blotting before reacting with coating F81 specific antibodies on the NC. As a result, a white dot on the NC was not developed although a relatively high quantity of F81 standard (10 pg) was spotted on the NC. Thus the ELISAGRAM would not be useful for detecting putative F 81 -like compounds. Detection of F81 in Fusarium cultures F81 analysis was performed by HPLC and CD-ELISA using F81 -sheep antisera (Neogen Corp., Lansing, MI). F. graminearum (FW-8), F. proliferatum (M-5956), and F. moniliforme (M-5958) were grown in solid (ground corn) media. 91 Non FB1-producing F. graminearum (FW-8) and uninoculated samples were used as negative controls. The corn cultures were harvested at 2, 3, 4, and 5 weeks, and then extracted with 50% acetonitrile, filtered, and subsequently analyzed with HPLC and CD-ELISA. Standard curves for CD-ELISA and for HPLC analyses were generated and shown graphically in Figures 2.10 and 2.12, respectively. Based on these standard curves, F 81 concentrations in Fusarium corn cultures were calculated and the results were tabulated in Table 2.4. F 81 concentrations in the cultures ranged from 0 to 220 pglg with HPLC and from 0 to 759 pglg with ELISA. In both negative controls, F 81 was not detected by either HPLC or ELISA. However, when this ELISA was used to assay F81 in fumonisins producing cultures, it yielded F81 estimates higher (mean = 2.8-fold) than HPLC method (Table 2.4). These current study results were a vast improvement over the findings of Tejada-Simon (1994) who found that ELISA methods yielded much higher F81 estimates (up to 400-fold) than HPLC. The improvement was likely caused by the sheep antiserum which had higher affinity and specificity than 205 monoclonal antibodies used by Tejada-Simon (1994) (Table 2.2). Detection of F8. in corn and corn product Corn and corn products analyzed in this study consisted of 77 fresh com, 14 com food and 28 com feed samples. Ground corn samples were extracted with 75% (vol.lvol.) methanol in distilled water. After filtering through Whatman #4 filter paper (Whatman Ltd., Maidstone, UK), the raw methanolic extracts were 92 diluted at a 1:35 and then analyzed by CD-ELISA using FB1- sheep antisera (Neogen Corp, Lansing, MI). Although detection of F81 in the food and feed extracts at a dilution of 1:35 with the CD—ELISA could be performed successfully, a problem was found when analyzing F 81 in the fresh corn extracts. After color development, some samples had a higher color intensity (higher 0.0.) than the zero F81 standard solution. CD-ELISA standard curves for F81 in extractant [10% methanol (vol.lvol.) in distilled water], and raw methanolic extracts of fresh corn sample numbers 37 and 56 (at a dilution of 1:35) were graphically shown in Figure 2.8. All standard curves had relatively the same pattern, but different 00. indicating that the raw corn extracts contained an interfering material which was able to generate non specific binding. Several possibilities might relate to this observation. First, the corn extracts might contain peroxidase which then reacted with peroxidase substrate (ABTS) resulting increase of color intensity. Second, the corn extracts might contain protein compounds which could bind to both sheep antisera and FBi-HRP resulting higher color intensity alter the addition of ABTS. Third, the pH of the corn substrate might favor to the reaction of sheep antisera with F81 -HRP or with peroxidase-containing compounds which subsequently increased color intensity. Fourth, the corn extracts might contain non protein compounds which could increase color intensity. Several efforts have been performed to solve the above problem. The corn extracts was heated and then centrifuged to inactivate peroxidase and to eliminate protein compounds from the corn extracts. Addition of NaCI to the corn extracts was also performed to precipitate the interfered proteins. Measurements 93 of corn substrate pH and adjustment of the pH to neutral were also carried out to eliminate the effect of the sample pH. All of these efforts, however, did not solved the problem indicating that the interfered compounds were not peroxidase or proteins. The problem was eliminated after the corn extracts were diluted up to 200 times or higher with 10% methanol in distilled water. By diluting sample extracts, Laamanen and Veijalainen (1992) were also able to eliminate a similar problem when they analyzed T-2 toxin in milled grains with an ELISA method. After diluting fresh corn extracts up to 200 times, CD—ELISA standard curves for _ F8. in the extractant and in the diluted corn extracts had relatively the same 0.0. and pattern (Figure 2.9). Beside the CD-ELISA, Veratox® (Neogen Corp, Lansing, MI) and HPLC were also used to detect F81 in fresh com, com products and spiked-com samples. After sample extraction and filtration, raw methanolic extracts were analyzed for F81 using both CD-ELISA and Veratox® methods. Some raw extracts were cleaned up with SAX columns and then analyzed by HPLC. A standard curve of HPLC (Figure 2.13) and standard curves of CD-ELISA and Veratox® (Figure 2.10) for analyzing F81 in corn and corn products were then developed. Based on these standard curves, F81 concentrations in com-spiked samples, corn, and corn products were calculated. Alter calculating F81 concentrations in com-spiked samples containing F81 of 100 to 3000 nglg, the F81 concentrations in the samples were divided by the added F 81 concentrations and then multiplied by 100% to determine percent recovery of the mycotoxin (Table 2.5). Percent recoveries of F81 in SAX- 94 columned samples with HPLC ranged from 68.8 to 84.7 percent (average = 74.1) (Table 2.5) indicating some F81 might loss during extraction and SAX-column purification. These recovery results were relatively similar to the findings of Stack and Eppley (1992) who analyzed F81 in SAX-columned extracts of com-spiked samples containing F81 of 500 to .2000 nglg with HPLC, and found percent recoveries of F 81 in the range of 63.2 and 71.5 percent. Meanwhile, by using the same method Usleber et al. (1994) reported that percent recoveries of the same toxin in corn samples spiked with 50 nglg and 500 nglg F81 were 63.2% and 65.6%, respectively. When both Veratox® and CD-ELISA were used to analyze F81 in the com-spiked samples, percent recoveries of F 81 in the raw methanolic extracts were higher than that in SAX-cleaned extracts (Table 2.5), indicating that some F81 might have lost during SAX column purification. F81 could not be detected in SAX-clean extracts from the samples which were spiked with only 100 or 200 nglg F81 (Table 2.5) because all of the spiked toxin might have lost during purification. Percent recoveries of the spiked F81 in the raw methanolic corn extracts determined with CD-ELISA and Veratox® ranged from 48.5 to 127.2% (Table 2.5). This variability might result from two possibilities. Firstly, the presence of F81 in the corn extracts might not be distributed evenly during spiking, so some samples yielded high recoveries and the other ones provided low recoveries. Secondly, the F81 -extraction efficiencies from corn containing different amounts of F81 might differ. Because of high variability, these percent 95 recoveries were not used to adjust the concentration of F81 in corn and corn products. F81 concentrations in 14 food samples ranged from <0.04 to 7.11 pglg (part per million, ppm), 0.13 to 9.59 ppm and <0.02 to 10.16 ppm when detected by HPLC, Veratox® , and CD-ELISA, respectively (Table 2.6). The ratio of Veratox® and CD-ELISA results to HPLC results ranged from 1.0 to 1.5 (Table 2.6). These ratio results were significant improvement over the findings of Pestka et al. (1994) who reported that the ELISA estimates were higher (up to 30-fold) than HPLC estimates. As in Fusarium corn cultures, sheep antiserum used in this currentstudy had greater affinity and higher sensitivity than 205 monoclonal antibodies used by Pestka et al. (1994) (Table 2.2). These undoubted contributed to the analytical improvement. These results indicate that both Veratox® and CD-ELISA were suitable to routinely screen com-based foods from F81. The levels of F81 in Italian feed samples ranged from <0.04 to 4.99 ppm, 0.34 to 7.63 ppm and 0.27 to 8.43 ppm when measured by HPLC, Veratox®, and CD-ELISA, respectively (Table 2.7). Veratox® and CD-ELISA analyses yielded ‘ F 81 estimates approximately twice higher (mean = 2.24 and 1.93 respectively) than HPLC method. Regression analyses between HPLC and Veratox® and between HPLC and CD-ELISA had linear correlation coefficients (r) of 0.961 and 0.951, respectively (Table 2.7) indicating that HPLC had a strong positive relationship with both Veratox® and CD-ELISA methods. These current study results were much better than the finding of Minervini et al. (1992) who reported 96 that the correlation coefficient between HPLC and ELISA was only 0.240, indicating no relationship at all between ELISA and HPLC methods. Like the above mentioned assays of Fusarium cultures and foods, the sheep antiserum was critical to ELISA improvement. Seventy seven of fresh corn samples were first analyzed by Veratox® to screen for samples contaminated with F81. All positive samples and approximately half of the negative samples were reanalyzed by Veratox®, C0- ELISA and HPLC. Analysis of positive fresh corn samples, Veratox® and CD- ELISA yielded higher F81 estimates (mean = 2.85 and 1.54, respectively) (Table 2.8) than HPLC. However, the three methods yielded the same results when applied to negative fresh corn samples (Table 2.8). Linear correlation coefficients (r) between HPLC and Veratox® and between HPLC and CD-ELISA were 0.933 and 0.836, respectively (Table 2.8) indicating that HPLC correlated well with both Veratox® and CD-ELISA methods. Thus, both Veratox® and CD-ELISA can be useful for routinely screening fresh corn from F81 . CD-ELISA developed in this current study was much better than the previous CD-ELISA that was developed by Azcona-Olivera et al. (1992a,b) and used by Minervini et al. (1992), Pestka et al. (1994) and Tejada-Simon (1994) for analyses of F 81 in feeds, foods, and fungal cultures, respectively. However, when this “new" CD-ELISA was used to detect F81 in corn cultures, corn foods, corn feeds and fresh corn, it still provided higher F81 levels (means = 2.8, 1.3, 1.9, and 1.5, respectively) as compared to those obtained by HPLC (Tables 2.4, 2.6, 2.7, 2.8). This disagreement might relate to several possibilities. Firstly, HPLC 97 detected only F81 ; whereas, CD-ELISA detected not only F81 but also F82 and F 83 since F81 sheep antisera used in the CD-ELISA cross-reacted with F82 and F83 (Table 2.3). Secondly, the sample extracts might contain F81 -similar compounds which were detectable by CD-ELISA but not by HPLC. The F81 - sheep antisera might not be completely specific for F81, so they can possibly detect other related compounds. Thirdly, because it contains an amine (-NH2) functional group, F81 might react with protein compounds prior to or during extraction to form F 81 -protein conjugates that cross react with F81 -sheep antisera, and thus increase the response of the CD-ELISA without affecting HPLC detection. CONCLUSION Mouse monoclonal antibodies, rabbit and sheep polyclonal antibodies against F81 were readily generated after immunization with keyhole limpet hemacyanin (KLH) as a protein tamer. The sheep antisera had the highest affinity and were most specific to F 81 as compared to the mouse monoclonal and rabbit polyclonal antibodies when they used in competitive inhibition ELISA Cross reactivities of these sheep antisera toward fumonisin B1 , 82 , and 83 were 100, 24 and 30%, respectively. C03ELISA using these F81 sheep antisera provided approximately two fold higher F B. estimates than HPLC when used for detection of F 81 in Fusarium corn cultures, corn and corn products. This was much better than CD-ELISA previously developed in our laboratory In addition, this CD-ELISA as well as Veratox® correlated well with HPLC methods, suggesting that it is suitable for routinely screening Fusarium cultures, corn and corn products from F 83 toxin. 98 99 0.0. (405 NM) ’4 ,5“ if” /x) // ,////’D // o’ // A // / // / (I -O-— /U/ // // — ' RFi // / // / // __D—- R24 / // K/ V // —-A—-- R3,; / / / /// / / / // / / / ///// “0‘ R110 / / / ./// V 324’ / / / A/// —<>— R,-p / / / I Figure 2.1. ANTIBODY DILUTION ELISA titration of rabbit polyclonal F81 antibodies. Sera were obtained two weeks after the second injection with FB1-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate indirect ELISA. Open symbols are direct ELISA. R1, R2 and R3 refer to rabbit number. The letter i indicates immunized and p indicates preimmun. 100 90 80 70-1 PERCENT INHIBITION 30—l 20— 101 Figure 2.2. 100 60— 50 IIT ITWI IIII TTTI TTIW T Ijfl ITIT IITI TIITTIII l l l 100 1000 10000 PB, (nglml) Competitive inhibition ELISA for F81 using different rabbit polyclonal antibodies. Sera were obtained two weeks after the second injection with FBi-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate Cl-ELISA. Open symbols are CD-ELISA. R1, R2 and R3 refer to rabbit number. 101 100 BO—I 704 8 I IIrI TllI‘rIIrIITI PERCENT INHIBITION 8 1 1o— IITj IIII VIII IPWTIIII IIII F8, (nglml) Figure 2.3. Competitive inhibition ELISA for F81 using mouse sera. Sera were obtained ten days after the third subcutaneous (SC) injection with FB1-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate Cl-ELISA. Open symbols are CD-ELISA. Subscript 1-4 refers to mouse number. 102 100 IIII IIII 00— TTTI N c I ITIVIIII PERCENT INHIBITION S l Ir—II—ITITI IIII 10“ ITII ITII 111 l 1 111111 I 141L111 I l 100 1000 10000 FE, (nglml) Figure 2.4. Competitive inhibition ELISA for F81 using mouse sera. Sera were obtained ten days after the third intraperitoneal (IP) injection with FB1-KLH immunogen. Each data point represents the average value of duplicate measurements. Filled symbols indicate Cl-ELISA. Open symbols are CD-ELISA. Subscript 1-4 refers to mouse number. 103 110 IIrI too— IIII I—I—TI 00-4 st 0 l PERCENT INHIBITION IIII IIII IIII IfiIITIIII IITI ITIT ITrI IIII '10 111111 1 1 111111 J 1 111111 1 1 111111 10 100 1000 10000 FB1 (nglml) Figure 2.5. Competitive indirect ELISA curves for F81 using hybridoma supematants. Supematants were obtained from hybridoma cells producing F 81 antibodies before cloning. Each data point represents the mean value of duplicate measurements. 104 10-— T I T I 2.5 —l FT If ZD'~ I I I I 1A5— o.o. (405 NM) I I I j L0-— I I I I 05-— I T I j 00-— _I N2C5 R185 10‘ Figure 2.6. 10‘ 1o4 104 ANTIBODY DILUTION ELISA titration of monoclonal F81 antibodies prepared from ascites fluid. Each data point represents the mean value of duplicate measurements. Filled symbols indicate Cl-ELISA. Open symbols are CD-ELISA. Q1C5 clone had no titer when determined with CD- ELISA. 105 100 t E so .1 : . N2C5 so—: A R185 : v 70 __ o1cs : I ozcs 2 3°“- 0 _ E _ m _- 5 5° - g l- i— I E 4o— 0 E Q 0 N a 8 I I rWIIfiTWI ITIj IITI rIII .10 1111 1 1 L11111 1 1 111111 1 111L111 1 1 111111 1 10 100 1000 10000 FB1 (nglml) Figure 2.7. Competitive inhibition ELISA for F81 using monoclonal antibodies prepared from ascitic fluid. Data points represent the mean value of duplicate measurements. Filled symbols were Cl-ELISA. Open symbols were CD-ELISA. Q1CS clone had no titer when determined with CD-ELISA. 106 Table 2.1. Sensitivity of Cl-and CD-ELISAs for F81 using monoclonal antibodies prepared from ascites fluid Amount of F 81 required for 50% inhibitiona (nglml) Clone Cl-ELISA CD-ELISA N2C5 50 400 R1 85 60 700 Q1C5 90 -" QZC9 100 800 a Data from Figure 2.7 b No titer in this assay. 107 1.2 — I. ~ v CORN EXTRACT#56(1:35) 1.0 I A CORN EXTRACTII 37 (1:35) - 0 EXTRACTANT 0.0 — g _ no J” 8 0.0 RI : o 0.4 — 0.2 a o.o___jJ11I 1 _1 111111 1 1 111111 0.0 0.1 1.0 10.0 100.0 F31 ("9"”) Figure 2.8. Competitive direct ELISA curves for F81 using F81 sheep anti sera (Neogen Corp., Lansing, MI). F8, was dissolved in extractant [10% methanol (vol.lvol.) in distilled water], fresh corn extract numbers 37 and 56 at a dilution of 1:35). Each data point represents the mean value of triplicate measurements. 108 1.2 - L. r v CORN EXTRACT If 50 (1:200) . 1.0 —-h A CORN EXTRACT 11 37 (1:200) - 0 EXTRACTANT 0.0 — / 0.6 -i ‘\ 0.0. (405 NM) T I I I 0.4 — 0.2 I \ 001111 4 1111111 1 1 111111 1 1111111 1 1111_11 I 0.0 0.1 1 .0 10.0 100.0 FB1 (nglml) IITW I r I I Figure 2.9. Competitive direct ELISA curves for F81 using F81 sheep anti sera (Neogen Corp, Lansing, MI). F81 was dissolved in extractant [10% methanol (vol.lvol.) in distilled water], fresh corn extract numbers 37 and 56 at a dilution of 1:200). Each data point represents the mean value of triplicate measurements. 109 110 100 - 90— 80 PERCENT INHIBITION '5’ 8 3 S S 3‘ 1 1 1 1 1 .5 O L a I IITT O VERATOX ITII C SHEEP IIIW A R: IIIV V 2D5 MAB IIII CI R1 BS MAB IIII ITII IIII ITTI TIII IIII TTFT .\ . 1 1 1 11111 J1 1 L1111L 111 1 11111 1 1 111111 1 1 111111 1 1 1 11111 I d O O 0 I I I I 10.0 1 00.0 1 000.0 1 0000.0 FB, (nglml) Q P .5 .L C Figure 2.10. Comparison of competitive inhibition ELISA for F8, mouse- monoclonal antibodies, rabbit-and sheep-polyclonal antibodies, and Veratox®. Monoclonal antibodies (N205 MAB and 205 MAB) were ‘ prepared from ascitic fluids. N205 MAB was generated in the current study using F81-KLH immunogen, while 205 was produced by Azcona-Olivera (1992a) using F81-0holera toxin immunogen. R3 refers to rabbit number 3. Sheep antiserum was from a single lot supplied by Neogen Corp. (Lansing, MI). Veratox" is an ELISA kit for F81 based on CD-ELISA and is produced and supplied by Neogen Corp. (Lansing, MI). Monoclonal antibodies were determined with Cl- ELISA, but polyclonal antibodies were measured with CD-ELISA. Each data point represents the mean :1: standard errors of the mean (11 = 4, two of duplicate measurements). Absorbencies at 0 nglml F 81 ranged from 0.675 to 1.250. 110 Table 2.2. Comparison of competitive F 81 ELISA using mouse-monoclonal antibodies prepared from ascites fluid, rabbit-and sheep-polyclonal antibodies F81 concentration Absorbance value . required for 50% for 0 ng F 81 lml Antibody Inhibition“ (nglml) ELISA format 205b 200 1.10 Cl-ELISA N205° 100 1 .25 Cl-ELISA R3° 80 0.79 CD-ELISA Sheep" 6 0.74 CD-ELISA Veratox' 6 0.68 CD-ELISA " Data from Figure 2.8. b Monoclonal antibodies produced by Azcona-Olivera (19928) using F 81-0holera toxin immunogen. ° Monoclonal antibodies generated in the current study using F B1-KLH immunogen . d Rabbit polyclonal antibodies from rabbit number 3. e Sheep polyclonal antibodies from Neogen Corp. (Lansing, MI). ' Veratox is an ELISA kit for F81 based on CD—ELISA and is produced and supplied by Neogen Corp. (Lansing, MI). 111 100 IIII 90—1 ITII 80— 0 F32 IIrI 70— VIII 004 504 IIIrTrfirI PERCENT INHIBITION 30-1 ITIIfiITfi 20- 10« TPIIHIII o 1 111111 1 _1 J_1_1111 1 1 11111—11 1 10 100 F8, (nglml) Figure 2.11. Competitive direct ELISA standard curves for F81 , F82 and F83 , using F81 sheep antisera. Data for sheep antisera were provided by Dr. Mohamed M.Abouzied (Neogen Corp., Lansing, MI). Each data point represents the average value of triplicate determination. 112 Table 2. 3. Cross reactivity of F81 sheep antisera toward fumonisin analogues. Fumonisin Fumonisin concentration required Cross reactivity (%)° analogue for 50% inhibition8 (nglml) F 81 5.5 100 FB2 22.9 24 F83 18.3 30 a Data from Figure 2.9. b Cross reactivity defined as (nglml of F81 required for 50% inhibition)l(nglml of fumonisin analogue required for 50% inhibition) x 100. 113 600 . v - 5.384x + 2.224 50° "I E - 0.999 400 — g 300 — .- _ 5 r- 2 200 -— L. 100 _ 1. o .— r- 1 1 1 1 1 1 1 1 1 1 1 1 11 1 1 1 1 1 1 1 1 1 1 r 1 1 l 1 0 20 40 00 00 100 120 F81 (uglmi) Figure 2.12. A typical “broad range “ standard curve used for HPLC analyses of Fusarium corn cultures. Each data point represents the average value of two determinations. 114 1000 "' Y I 01.789 X + 3.991 800 — I" 3 0.998 600 — 400 -— AREA 0: 1000) 200 - 1111 41111111 1111 11114114 111L 0 2 4 6 8 10 12 F8, (nglml) Figure 2.13. A typical “low range “ standard curve used for HPLC analyses of corn and corn products. Each data point represents the average value of two determinations 115 Table 2.4. Comparison of F81 concentrations in Fusarium corn cultures detected by HPLC and CD-ELISA methods SAMPLE HPLC (pglg) CD—ELISA (pg/g)“ RATIO OF ELISA TO HPLC Controlj’, wk. 2 <0.04° <0.02h - Control, wk. 3 <0.04 <0.02 - Control, wk. 4 <0.04 <0.02 - Control, wk. 5 <0.04 <0.02 - FW-B c, wk. 2 <0.04 <0.02 - FW-8, wk. 3 <0.04 <0.02 - FW-8, wk. 4 <0.04 <0.02 - FW—8, wk. 5 <0.04 <0.02 - 101-5955 ‘1, wk. 2 7.0 19.3 2.8 M-5956, wk. 3 33.9 30.3 0.9 M-5956, wk. 4 9.7 26.1 2.7 M-5956, wk. 5 21.4 39.7 1.9 M-5958 °, wk. 2, R1 23.8 58.1 2.4 M-5958, wk. 3, R1 9.8 27.4 2.8 M-5958, wk. 4, R1 17.9 58.1 3.2 M-5958, wk. 5, R1 24.3 76.2 3.1 M-5958, wk. 2, R2 8.9 24.2 2.7 M-5958, wk. 3, R2 220.0 759.0 3.5 M-5958, wk. 4, R2 <0.04 <0.02 <0.02- M-5958, wk. 5, R2 25.7 109.0 4.2 MEAN :I: SEM r 2.8 :1 0.9 ' CD-ELISA using F81 sheep antisera (Neogen Corp., Lansing, MI); Linear regression analyses between HPLC (X) and CD-ELISA (Y) at P<0.05 yielded an equation line of Y = 3.428X - 7.640 and linear correlation coefficient (r )=0.992. b Uninoculated samples. " F. proliferatum cultures (M-5956). ' Standard errors Of the means. " CD-ELISA detection limit (20 nglg) ° Fusarium graminearum cultures(FW-B). ° Fusarium moniliforme cultures (M-5958). 9 HPLC detection limit (0.04 nglg) wk. 2, 3, 4, and 5 refer to week number 2, 3, 4, and 5 respectively at which Fusarium cultures were harvested. R1 and R2 refer to replication number 1 and 2, respectively. 116 Table 2.5. Comparison of F81 recoveries from spiked-com samples containing F81 of 100 to 3,000 pglg using raw methanolic extracts and SAX- Cleaned extracts as determined by HPLC, Veratox® ', and CD—ELISA using F 8. sheep antisera (Neogen Corp., Lansing, MI) PERCENT RECOVERIES " of F8. in F8. RAW METHANOLIC EXTRACTS SAX-CLEANED EXTRACTS ADDED nglg Veratox® CD-ELISA HPLC Veratox® CD-ELISA 100 86.3 3 24.0 nd 68.8 3 4.2 nd nd 200 127.2 3 31.8 48.5 3 3.3 71.2 3 12.4 nd nd 600 80.5 3 12.4 70.3 3 13.3 84.7 3 7.2 56.5 3 6.4 50.2 3 9.1 1,000 71.5 3 6.8 70.0 3 2.3 73.6 3 9.9 50.2 3 10.7 58.7 3 9.6 3,000 60.5 3 6.4 50.1 3 4.5 72.1 3 5.4 51.6 3 9.1 38.8 3 1.7 MEAN° 85.2 3 16.3 59.7 3 5.9 74.1 3 7.8 45.2 3 16.3 49.2 3 6.8 a Veratox® Fumonisin Test, Neogen Corp.(Lansing, Ml). " (F81 found/F81 added) x 100%. ° Calculated from positive recoveries. nd indicates no F81 detected (Detection limits of Veratox® and CD-ELISA were 0.02 pglg) ; Thus, recovery could not be calculated. Each data point represents the mean 3: SEM (n = 3). SEM = standard errors of the means. 117 Table 2.6. Comparisons Of F81 concentrations in 14 com-food samples by HPLC. Veratox® ‘, and CD-ELISA using F81 sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox®a and CD- ELISA to HPLC results SAMPLE FUMONISIN B1 (p919) FOUND” BY RATIOS TO HPLC ° NUMBER “ HPLC Veratox®' CD-ELISA Veratox®” CD-ELISA - 41 <0.04' 0.13 0.02' - - 38 <0.04 0.15 0.29 - - 43 <0.04 0.17 0.12 - - 48 <0.04 0.14 0.07 - - 33 0.34 0.44 0.41 1.3 1.2 32 0.42 0.58 0.59 1.4 1.4 25 0.64 0.94 0.85 1.5 1.3 10 0.75 0.96 1.16 1.3 1.5 8 0.92 0.97 1.28 1.1 1.4 7 1.10 1.24 1.08 1.1 1.0 22 1.21 1.61 1.20 1.3 1.0 6 1.79 2.35 2.25 1.3 1.3 21 2.27 3.21 2.40 1.4 1.1 H9 7.11 9.59 10.16 1.3 1.4 MEAN 1.18 1.61 1.56 1.31 1.26 smo 1.84 2.47 2.59 0.12 0.20 ' Veratox® Fumonisin Test, Neogen Corp. (Lansing, MI). b Linear regression analyses between HPLC (X) and Veratox® (Y1) and between HPLC and CD- ELISA (Y2 ) at P < 0.05 yielded equation lines of Y, = 1.473X - 0.485; r= 0.992 and Y2 = 1.370X - 0.356; r= 0.983. c Veratox® or CD-ELISA results divided by HPLC results. ‘ Ranked according to HPLC results; the sample descriptions were on Appendix (Table 5.1). ‘ HPLC detection limit (40 nglg). ' CD-ELISA detection limit (20 nglg). ’ Standard errors of the means. 118 Table 2.7. Comparisons of F 81 concentrations in 28 Italian feed samples by HPLC, Veratox® ‘, and CD-ELISA using F81 sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox®a and CD- ELISA to HPLC results SAMPLE FUMONISIN B1 (pglg) FOUND” BY THE RATIOS OF HPLC‘ NUMBERd HPLC Veratox®' CD-ELISA Veratox®“ CD-ELISA F18 <0.04' 0.34 0.51 - - F20 <0.04 0.49 0.27 - - F8 <0.04 0.51 0.87 - — F25 0.24 0.62 0.41 2.6 1.7 F3 0.11 0.40 0.38 3.6 3.4 F22 0.21 0.43 0.15 2.1 0.7 F1 0.22 0.95 0.61 4.3 2.7 F16 0.24 0.56 0.68 2.3 2.8 F11 0.34 0.90 0.74 2.6 2.2 F24 0.36 0.72 0.88 2.0 2.5 F23 0.38 1.25 0.74 3.3 2.0 F30 0.42 0.67 0.32 1.6 0.8 F13 0.44 1.01 0.58 2.3 1.3 F10 0.50 1.07 0.74 2.2 1.5 F29 0.62 1.12 0.79 1.8 1.3 F4 0.67 2.23 1.87 3.3 2.8 F7 0.79 1.62 2.85 2.0 3.6 F28 0.94 1.16 1.44 1.2 1.5 F9 0.94 3.05 2.30 3.2 2.4 F2 1.00 1.69 0.88 1.7 0.9 F14 1.07 2.06 1.73 1.9 1.6 F27 1.16 2.37 1.66 2.0 1.4 F19 1.24 2.40 1.93 1.9 1.6 119 Table 2.7. (Con’d) SAMPLE FUMONISIN B1 (pg/g) FOUND” BY THE RATIOS OF HPLC‘ NUMBER“ HPLC Veratox®“ CD-ELISA Veratox®“ CD-ELISA F17 1.31 2.37 2.60 1.8 2.0 .33, 1.39 2.17 1.96 1.6 1.4 F26 1.67 2.96 3.84 1.8 2.3 F15 3.71 5.01 8.43 1.3 2.3 F12 4.99 7.63 7.87 1.5 1.6 MEANI 1.00 1.86 1.86 2.24 1.93 SEMO 1.11 1.60 2.10 0.77 0.77 'Veratox® Fumonisin Test, Neogen Corp. (Lansing, MI). b Linear regression analyses between HPLC (X) and Veratox ® (Y1) and between HPLC (X) and CD-ELISA (Y2 ) at P < 0.05 yielded equation lines of Y. = 1.010X + 0.532; r= 0.961 and Y2 =1.300X + 0.160; r= 0.951). °Veratox® or CD-ELISA results divided by HPLC results. ° Ranked according to HPLC results, and the sample descriptions were on Appendix (T able 5.2). ° HPLC detection limit (0.04 nglg). ' Mean of positive samples according to HPLC results. 9 Standard errors Of the means 120 Table 2.8. Comparisons of F 81 concentrations in fresh com 3 samples by HPLC, Veratox® 3, and CD-ELISA using F 81 sheep antisera (Neogen Corp., Lansing, MI) and the ratios of Veratox®b and CD- ELISA to HPLC results SAMPLE FUMONISIN B. (pglg) FOUND6 BY THE RATIOS OF HPLC‘I NUMBER“ HPLC Veratox®” CD-ELISA Veratox®” CD-ELISA P14 <0.04' <0.02“ <0.02 " - - P15 <0.04 <0.02 <0.02 - - P19 <0.04 <0.02 <0.02 - - P21 <0.04 <0.02 <0.02 - - P25 <0.04 <0.02 <0.02 - - P27 <0.04 <0.02 <0.02 - - P28 <0.04 <0.02 <0.02 - - P31 <0.04 <0.02 <0.02 - - P32 <0.04 <0.02 <0.02 - - P37 <0.04 <0.02 <0.02 - - P46 <0.04 <0.02 <0.02 - - P5 <0.04 <0.02 <0.02 - - P50 <0.04 <0.02 <0.02 - - P51 <0.04 <0.02 <0.02 - - P52 <0.04 <0.02 <0.02 - - P53 <0.04 <0.02 <0.02 - - P54 <0.04 <0.02 <0.02 - - P55 <0.04 <0.02 <0.02 - - P56 <0.04 <0.02 <0.02 - — P58 <0.04 <0.02 <0.02 - - P59 <0.04 <0.02 <0.02 - - P62 <0.04 <0.02 <0.02 - - P67 <0.04 <0.02 <0.02 - - P7 <0.04 <0.02 <0.02 - - P74 <0.04 <0.02 <0.02 - - P9 <0.04 <0.02 <0.02 - - P17 <0.04 <0.02 <0.02 - - P26 <0.04 <0.02 <0.02 - - P63 <0.04 <0.02 <0.02 - - P42 0.04 0.27 0.26 7.1 6.8 P61 0.05 0.26 0.16 5.2 3.1 P80 0.05 0.25 <0.02 4.6 0.0 121 Table 2.8. (Con’d) SAMPLE FUMONISIN B, (11919) FOUND6 BY THE RATIOS OF HPLC“ NUMBER' HPLC Veratox®° CD—ELISA Veratox®° CD-ELISA P10 0.08 0.16 <0.02h 2.0 0.0 P12 0.10 0.28 <0.02 2.8 0.0 P57 0.10 0.23 <0.02 2.2 0.0 P8 0.12 0.21 <0.02 1.7 0.0 P1 0.15 0.31 0.15 2.0 1.0 P66 0.25 0.75 0.60 3.0 2.4 P29 0.28 0.52 0.34 1.9 1.2 P45 030 0.47 0.86 1.5 2.8 P20 0.47 0.76 0.88 1.6 1.9 P44 0.70 0.92 0.56 1.3 0.8 MEANi 0.20 0.41 0.29 2.85 1.54 SEMl 0.21 0.23 0.33 1.75 1.95 ' Harvested in five counties of Michigan in Summer 1994. Samples were supplied by Dr. Patrick L. Hart (105 Pesticide Research Center, Michigan State University, Ml, 48824). All samples were first analyzed with CD-ELISA; and then all positive samples as well as randomly selected negative samples were analyzed with HPLC, Veratox®, and CD-ELISA. . ” Veratox® Fumonisin Test, Neogen Corp. (Lansing, MI). ° Linear regression analyses between HPLC (X) and Veratox® (Y3) and between HPLC (X) and CD-ELISA (Y2 ) at P < 0.05 yielded equation lines of Y1 = 1.127X + 0.039; r= 0.933 and Y2 = 0.973X + 0.008; r = 0.836. dVeratox® or CD-ELISA results divided by HPLC results. ° Ranked according to HPLC results, and the sample descriptions were on Appendix (Table 5.3). F 9- " Detection limit of HPLC (40 nglg), Veratox® (20 nglg), CD-ELISA (20 nglg), respectively. ' Mean Of positive samples. jStandard errors Of the means. 122 PART III PRODUCTION OF DEOXYNIVALENOL (VOMITOXIN) ANTIBODIES ABSTRACT PRODUCTION OF DEOXYNIVALENOL (VOMITOXIN) ANTIBODIES By Sutikno Deoxynivalenol (DON) was reacted with hemisuccinate (HS) for 8 hours to form DON-HS derivatives. These derivatives were conjugated to bovine serum albumin (BSA) and then used as an immunogen for producing DON specific antibodies. Twenty female BALBIC mice were immunized intrasplenically, intraperitoneal, or subcutaneously and 6 female New Zealand rabbits were injected intramusculariy with the immunogen. All animals apparently produced high titer antisera. However, these antisera could not be used in competitive ELISAs for DON and also did not cross react with either 3-acetyl-DON, 15-acetyl-DON, nivalenol, 4,15—diacetylnivalenol or fusarenone—X. 123 INTRODUCTION Natural occurrence. Deoxynivalenol (DON, vomitoxin) is a trichothecene mycotoxin, produced by Fusan'um graminearum and Chemically determined to be 301, 7a, 15-trihydroxy- 12, 13-epoxytrichotheC-9-en-8-one (Table 3.1)(Yoshizawa and Morooka, 1973). DON was first isolated in Japan from Fusarium-infected barley (Morooka et al., 1972). This toxin was also found in the United States by Vesonder et al. (1973) who analyzed Northwest Ohio corn infected with Fusan‘um. These investigators tested the presence of the emetic compound by intubation of each fraction of their isolation procedures into pigs to induce vomiting at which they isolated and Characterized DON as an active compound. Because of its emetic effect on swine, DON was also named vomitoxin (Vesonder et al., 1973). As a consequence of these findings, surveys for the presence of DON in foods and feeds as well as testing for its toxicological and immunosuppresive effects began. Surveys for the presence of DON in foods especially grain and grain products have been done by several investigators in several countries and the results are summarized in Table 1.3. DON has been found worldwide and is a major contaminant in cereal grains (Tanaka et al., 1988; Hietaniemi and Kumpulainen, 1991). DON production occurs when environmental conditions in 124 125 Table 3.1. Structures of deoxynivalenol (DON) analogs (Adapted from Ueno, 1 983) Compound R1 R2 R3 R4 R5 DON OH H OH OH =O DON-3-HS OHS' H OH OH =O DON-15-HS OH H OHS' OH =0 3—Acetyl-DON OAc" H OH OH =0 15-Acetyl-DON OH H OAc" OH =0 Nivalenol 0H 0H 0H 0H =0 4,15—Diacetylnivalenol OH OAo" OAob OH =0 Fusarenon-X 0H OAC" 0H 0H =0 ‘ T-2 toxin 0H 0aCb Oacb H 0Ip OHS', OAc", Olp° indicate 000(0H2)2000H, 0000H3, and 000H2CH(0H3)2, respectively. 126 the field are low temperature and high humidity (Cote et al., 1984; Vesonder et al.,1978; Mills, 1982), although this toxin has also been found in high temperature regions (Richardson et al., 1985). Vesonder et al. (1978) reported that cold and wet weather tends to delay harvest and subsequently molds on the crop grow continuously, and thus produce high quantities of DON. In some cases, however, prolonged delay of harvest can yield a decline in DON concentration in grains left in the field, possibly due to reaction with plant components or metabolism by plant! fungal enzymes (Scott et al., 1984). In North America, the areas most often contaminated by DON are the midwestem region of the United states (Vesonder, 1983) and the eastern region of Canada (Seaman, 1982). Toxicity DON is much less toxic than other trichothecene mycotoxins such as T-2 toxin, HT-2 toxin, diacetoxyscirpenol, nivalenol, and fusarenon-X (Scott et al., 1980). DON was found to be 60-fold less toxic than T-2 toxin and two fold less toxic than nivalenol when the trichothecene toxicity is measured using lethal dose 50 (L053, an amount of toxin required to kill 50% population) of brine shrimp larvae (Scott et al., 1980). In mice, the L050 of DON is 49 mglkg body weight (intraperitoneal) and 78 mglkg body weight (oral) (Forsell et al., 1987). The toxicity of this toxin may be due to protein synthesis inhibition (Ueno, 1983). Consumption of high quantities of DON (at L053, or higher) results in Classical acute symptoms of trichothecene toxicity ranging from necrosis of the intestinal tract, bone marrow and lymphoid tissues as well as lesions in kidney 127 and heart, to death (Forsell et al., 1987; and Robbana-Bamat et al., 1987). Pathological Changes include lesions and degeneration of the stomach and small intestine mucosa, enlargement and edema of mesenteric lymph nodes, vascular congestion and depletion of all lymphoid organs and liver (Cote et al., 1985; Robbana-Bamat et al., 1987). At lower doses (— 1.0 —1 A sc-100 g“ I ,,, 0.8 4 c L 3. _ D: L o 1- 0.6 a 0.4 — 0.2 — C 0.0 1 1111111 1 I 111111 I 111L111 1 1 1L1111 1 1 I 10‘ 10‘ 10" 10:2 10'1 ANTISERUM DILUTION Figure 3.1. Indirect ELISA titration of mouse antibodies obtained ten days after the third injection with BSA-HS-DON conjugate. The first injection was given at a dose of 50 and 100 pglmouse. The second and third injections were given at a dose of 25 and 50 pglmouse. Each data point represents the mean :I: standard error of the mean (n = 6, duplicate measurements from three mice). IP indicates peritoneal injection. SC indicate subcutaneous injection. 50 and 100 indicate dose (pg) of the first injection. W‘s 145 1.2 TTIV 1.0 — fl I I I 08 O IiTI I 06 I 0.0. (405 NM) 0.4 m I I I I 0.2 - I I I I . SI-IP ‘ SI-SC 5 O TI-IP T A TI-SC J O / I .11. 11111 1 1111111 1 1 1111111 Figure 3.2. 1 1 10‘1 10:2 10'1 ANTISERUM DILUTION Indirect ELISA titration of mouse antibodies Obtained ten days after the second (SI) and third immunizations (T I) with BSA-HS-DON conjugate. The first immunization was given via intrasplenic deposition at a dose of 20 pglmouse. The second and third immunization were given via intraperitoneal (IP) and subcutaneous (SC) injections at a dose of 10 and 50 p9, respectively. Each data point represents the mean 1: standard error of the mean (n = 8, duplicate measurements from four mice). 146 1.2 I I I I 1.0 — I I I I on I I I I I 06— O 0.0. (405 NM) I I I I 0.4 -1 I I I I 0.2 — I I I T 0 HUNTER‘S ADJUVANT O FREUND'S ADJUVANT 0.0 Figure 3.3. 104 104 ANTISERUM DILUTiON Direct ELISA titration of rabbit antisera. Hunter's adjuvant indicates that rabbits were given a two sites intramuscular injection with an emulsion of Hunter Titer Max and BSA-HS-DON conjugate at a dose of 100 pg per rabbit for both the first and second injections. Freund’s adjuvant indicates that rabbits were given a ten site subcutaneous injection with an emulsion of Freund’s adjuvant and BSA-HS-DON conjugate at a dose of 500 pg and 250 pg per animal for the first and second immunization, respectively. Antisera were obtained four weeks after the second injection with BSA-HS-DON conjugate. Each data point represents the mean 1 standard error of the mean (n = 6, duplicate measurements from three rabbits). maxi—— 147 Compound R1 R2 R3 R4 R5 DON OH H OH OH =0 A O-HS-BSA H OH OH =0 8 OH H OH OH -O-CMO-BSA Figure 3.4. Deoxynivalenol (DON) and its conjugates. BSA-3-O-hemisuccinyl- DON (A) and BSA-8-O-carboxymethyl-oxime-DON (8). BSA, HS, and 0M0 indiwte bovine serum albumin, hemisuccinyl, and carboxymethyl-oxime, respectively. 148 PART IV. PRODUCTION OF ANTIBODIES AGAINST TRICHOTHECENE-YEAST RIBOSOMAL BINDING SITE ABSTRACT PRODUCTION OF ANTIBODIES AGAINST TRICHOTHECENE-YEAST RIBOSOMAL BINDING SITE 31/ Sutikno Both pure 608 and 808 ribosomal subunits were isolated from trichodennin- sensitive yeast and used for the production of antibodies against trichothecene ribosomal binding site. Twelve female BALBIC mice were immunized with either the 608 or 808 ribosomal subunits. Another ten female BALBIC mice were injected with the mixture of the 808 subunits with different adjuvants. All mice apparently produced high titer antisera. These antisera were used to develop a competitive indimct enzyme-linked immonosorbent assay (CI-ELISA) for detection of trichothecenes. However, this Cl-ELISA could not detect trichothecene because the antisera were not specific to the trichothecene ribosomal binding site. 149 INTRODUCTION Trichothecene mycotoxins Trichothecenes are a group Of structurally similar sesquiterpenoid metabolites produced mainly by toxigenic strains of Fusarium, and by some toxigenic strains of Trichothecium, Trichoderma, Acremonium, Cylindrocarpon, Myrothecium and Stachybotrys (Bamburg, 1983; Ueno, 1983; Committee on Protection Against Mycotoxin, 1983; Betina, 1989). These mycotoxins, especially deoxynivalenol (DON) and nivalenol (NIV) are found as natural contaminants in agricultural commodities throughout the world (T anaka et al., 1988a). The presence of DON and NIV trichothecenes in cereal and cereal products in several countries has been surveyed and the results were summarized in Table 1.3. Trichothecenes can cause a wide range of toxicoses to both animals and human (I' able 1.4) and cause large economic losses to farmer and livestock producers (Table 1.1). ’ The presence of trichothecenes in agricultural products in the field and during storage is mainly dictated by environmental factors and less importantly by genetic factors (Figure 1.1). Controlling the Climatic and biological factors that contribute to the presence Of the mycotoxins in agricultural commodities is almost impossible (Pestka, 1988). Thus, detection and diversion are the most important means for preventing the mycotoxin entry into food chains. 150 151 Immunoassay methods to detect the presence of mycotoxins in agricultural commodities have recently gained wide acceptance because of sensitivity, specificity, simplicity, and rapidity (Chu, 1991; Pestka et al., 1994a). lmmunoassays for individual trichothecenes, such as T-2 toxin, DON, diacetoxyscirpenol, NIV, and roridin A, have been developed to date (Table 1.5). Immunoassay kits for T-2 toxin, and DON have also been produced and marketed in the United States (Table 1.6). However, immunoassays that are capable Of detecting trichothecenes as a group in agricultural commodities have not yet developed. Ribosomal binding as a toxicity mechanism Trichothecenes are potent inhibitors of eukaryotic protein synthesis (Bamburg and Strong, 1971) because they bind to a common site on the 608 ribosomal subunit (Carrasco et al., 1973; Barbacid, and Vazquez, 1974; Cundliffe et al., 1974; Schindler et al., 1974; Cannon et al., 1976; McLaughlin et al., 1977). Occupancy of this binding site by some trichothecenes, such as T-2 toxin and NIV results in inhibition of protein synthesis at an initiation stage (Smith et al., 1975), whereas other trichothecenes including trichodennin lead to block elongation and termination _ stages by interfering with the peptidyl transferase on the ribosomes (Cundliffe et al., 1974) In order to elucidate the binding site, Schindler et al. (1974) induced mutations in a trichodennin-sensitive yeast strain (Saccharomyces cerevisiae A224A) and then isolated a trichodennin-resistant strain (S. cerevisiae CLP-1). By comparing the ribosomal components from both strains, these authors reported that 152 a mutant, which was resistant to the action of trichodennin, has an altered component on its 608 ribosomes. The gene determining the altered 608 ribosomal component is a single, recessive nuclear gene located on the right arm of Chromosome XV and named tcm1 (Grant et al., 1976). By Cloning yeast genes for trichodennin resistance and ribosomal protein L3 (the largest ribosomal protein), Fried and Warner (1981) isolated tcm1 gene and found that the gene coded for ribosomal protein L3. The nucleotide sequence of this tcm1 gene (ribosomal protein L3) has also been determined (Schultz and Friesen, 1983). The binding characteristics between trichothecenes and eukaryotic ribosomes have been studied by several investigators. Jimenez and Vazques (1975) used [acetyl-“0] trichodennin to study quantitative binding of trichodennin to ribosomes from trichodennin sensitive(Y133) and trichodennin resistant (T R1) yeasts. They found that Ym ribosomes bound to trichodennin with a dissociation constant (Kd) of 0.99 pM while those from the resistant one (T R1) bound to trichodennin with a Kd of 15.4 pM. Using similar techniques, Barbacid and Vazquez (1974) found that trichodennin bound to the peptidyl transferase center of trichodennin-sensitive yeast ribosomes with a Kd of 1.8 pM. and to 60$ subunit of the yeast ribosome with Kd Of 1.4 pM. Cannon et al. (1976) studied the binding affinity of the same toxin to different states Of the sensitive yeast ribosomes (polyribosome and “run Off” ribosome). They found that the binding affinity between trichodennin and the “run Off ribosome (Kd = 0.72 pM) is approximately threefold higher than that between trichodennin and the polyribosome (Kd = 2.1 pM). 153 Factors. affecting binding of trichothecenes and eukaryotic ribosomes have also been studied. Changes in ionic concentrations (from 0 to 300 mM of ammonium ion; from 0 to 25 mM of Mng), pH (from 6 to 8), and ethanol concentration (from 0 to 40% vol.lvol) do not significantly affect the binding between trichodennin and the peptidyl transferase center of yeast ribosomes although the activity of the center is much affected by ionic concentrations (Barbacid and Vazquez, 1974). Time and temperature of the reaction greatly affect the binding between T-2 toxin trichothecene and the yeast ribosomes (Middlebrook and Leathemnan, 1989). When incubated at 37°C, the association or 3H-T-2 with ribosome is biphasic. In the initial phase, binding is rapidly increased and by 30 minutes a plateau equilibrium state is achieved. After that a second phase occurs in which the binding gradually decreases to the base line value at a half-time of 2.5 hours. However, 3H-T-2-n'bosome binding at 4°C is much slower, taking 24-28 hours to reach completion, and very stable, requiring 3 weeks to decay 50%. When T-2 is prebound to ribosomes at 4°C and then incubated at 37°C, a significant degradation process begins after a time lag of approximately 4 hours (Middlebrook and Leathennan, 1989). A ribosome has only one binding site and can bind to only one molecule of trichothecene (Barbacid and Vazquez, 1974; Middlebrook and Leatherrnan, 1989). Trichothecenes as well as anisomycin compete with each other for the ribosomal binding site (Barbacid and Vazquez, 1974; Cannon et al., 1976; Middlebrook and Leatherrnan, 1989). Interestingly, competition among trichothecenes for the binding «I... 154 site appears to be proportional to their toxicity (Committee on Protection Against Mycotoxin, 1983; Middlebrook and Leathennan, 1989). Rationale and objective Trichothecenes bind specifically to yeast ribosomes. These toxins compete with each other for the ribosomal binding site in proportion to their toxicity. Production of antibodies specific to the binding site would enable researchers to develop ELISAs which an assess total “trichothecene load” in agricultural commodities and , thus would enhance food safety. Maximum binding between trichothecenes and yeast ribosomes is achieved by 30 minutes when incubated at 37°C. Thus, developing indirect ELISA using yeast ribosomes for coating microtiter plates (Figure 4.1) might be feasible. In such an assay, trichothecenes and trichothecene-ribosomal binding site antibodies would be incubated together over a solid-phase ribosomal binding site. Trichothecenes would compete with the antibodies for the ribosomal binding site. The total bound antibodies could be then detected by a second antibody which has been labeled with an enzyme. After the addition of an appropriate enzyme substrate, the bound antibody could be calculated based on the intensity of color development which is measured spectrophotometrically. The “trichothecene load” would be inversely related to a complex of bound enzyme-labeled antibodies. Based on the above literature study, I hypothesized that antibodies against trichothecene-yeast ribosomal binding site could be generated by immunization mice with either 608 or 808 yeast ribosomes. The specific Objectives of this study were to 155 produce antibodies against trichothecene-yeast ribosomal binding site and use these antibodies to develop indirect ELISA for trichothecenes as a group. MATERIALS AND METHODS Chemicals and reagents All organic solvents and inorganic chemicals were of reagent grade or better. Trichodennin sensitive S. cerevisiae (ATCC 46913) was obtained from American Type Culture Collection (Rockville. Maryland). This strain was maintained at 30°C on YEPD plates and grown in liquid medium at this temperature. Liquid YEPD media were prepared by autoclaving of a mixture solution of 10 9 of yeast extract (Difco Laboratories, Detroit MI), 20 g of bactO-peptone (Difco 0118, Difco Laboratories, Detroit MI), and 20 g of dextrose in 1 liter of distilled water. Ovalbumin (OA) (Chicken egg albumin grade III; fraction VII), Tween 20, 2,2'-azinobis(3- ethylbenzthiazolinesulfonic acid)(A8TS), hydrogen peroxide, T-2 toxin, and dithiothreitol were purchased from Sigma Chemical CO. (St. Louis, MO). Goat anti-mouse immunoglobulin G (lgG)-horseradish peroxidase conjugate was obtained from Cappel Laboratories (West Chester, PA). Tritium labeled T-2 toxin (3H-T-2) was kindly supplied by Dr. Fun Sun Chu (University of Wisconsin, Madison, Wisconsin). Methofane (methoxyflurane, inhalation anesthetic for veterinary) was purchased from Pitman-Moore Inc. (Mundelein, IL). 156 1 57 Yeast production Yeast was grown by the procedure of Treadgill et al. (1986). Briefly, colonies were picked from plates and inoculated into several 10 ml liquid YEPD media (cultures). After 10-18 hours of growthxfour of these cultures were used to inoculate four separate 150 ml cultures, which were incubated at 30°C with slow (approximately 150 rpm) shaking overnight. These cultures were examined microscopically for possible bacterial contamination and two were selected and used to inoculate two separate two liter cultures. These cultures were incubated at 30°C with slow shaking until the cell solution reached an A333 of 2.0. The grown culture was then cooled slowly to 8°C to induce ribosome run-Off. Cells were harvested and washed twice in distilled water with centrifugation at 10,000 g for 5 minutes at 2°C. Ribosome preparation Yeast ribosomes were prepared by the procedure of Battaner and Vazquez (1971) with slight modifiwfion. Briefly, each yeast preparation (approximately 100 9 wet weight of yeast cells) was resuspended in 300 ml of buffer containing 100 mM- TriS/HCI. pH 7.4, 12.5 mM-magnesium acetate, 80 mM KCI, and 5 mM dithiothreitol. Cells were then disrupted by four passages through a French Pressure Cell (Aminoo) at 700 lbirrn2 (48.3 MPa), and the lysates were centrifuged at 20,0009 for 15 minutes at 2°C. The supernatant fractions were then centrifuged again at 20,0009 for 30 minutes at 4°C to Clear any remaining debris and kept at 4°C before ultracentrifugation. 158 Clean supernatant fractions were sedimented by centrifugation in a Sorvall RC-60 ultracentrifuge (DU Pont Company, \Nlllington, Delaware) using T865.1 rotor at 45,000 rpm (150,0009) for 120 minutes. For washing, the pellet (crude ribosomes) was suspended in 24 volumes of washing buffer (10 mM Tris-HCl buffer, pH 7.4, 500 mM NH4CI, 100 mM Mg acetate, 5 mM dithiothreitol). An aliquot (5 ml) Of ribosomes was layered on 6 ml Of a discontinuous sucrose gradient in washing buffer (3 ml 20%, w/v sucrose in the bottom and 3 ml 5%, w/v in the top) and then sedimented by centrifugation at 150.0009 for 8 hours. The ribosome pellet was resuspended in about 25 volumes Of the standard buffer (10 mM Tris-HCl buffer, pH 7.4, 50 mM ‘NH4CI, 5 mM Mg acetate, 5 mM dithiothreitol). After centrifugation at 10,0009 for 10 minutes, the pellet was discarded, and the supernatant containing the 808 ribosomes was finally adjusted with the standard buffer to give a concentration of approximately 30 mg ribosomes/ml, aliquoted, and stored at -80°C. For preparation of 608 ribosome subunits, 1 ml Of the 808 ribosomes was dialyzed for 2 hours against two liter of dialyzing buffer (10 mM Tris-HCI buffer, pH 7.4, 50 mM NH4CI, 0.2 mM magnesium acetate, 5 mM dithiothreitol) at 4°C. Linear sucrose gradients were prepared by dissolving sucrose in the dialyzing buffer and . slowly layered into 11-ml tubes of the T8651 rotor (Du Pont Company, Willington, Delaware). The linear gradient consisted of 1 ml of each (from bottom to top) 25%, 20.5%, 17.0%, 13.0%, 9.5% and 7% sucrose (% sucrose was measured with ABBEL-3L refiactometer, Milton Roy 00., Rochester, NY). Three ml of 4% sucrose in dialyzing buffer, 1 ml of ribosome solution (15 mglml), and then 1 ml of the . dialyzing buffer were gently layered onto this linear gradient. 159 After 4 hours of centrifugation at 122.0009 (40,000 rpm of the T8651 rotor), the supernatant was fractionated and their absorbency was measured using Spectronic 601 spectrOphotometer (Milton Roy 00., Rochester, NY). The pellet (containing enriched 60$ subunits) was resuspended in the dialyzing buffer (5 ml) and centrifuged at 101,8009 for 3.5 hours. The resultant supernatant was discarded, and the 608 subunit pellet was rinsed and resuspended in standard buffer, aliquoted and stored at -80°C. T-2 toxin-ribosome binding assay Ethanol precipitation techniques used to determine T-2 toxin—ribosome binding were performed based on the procedure of Femandez-Munoz et al. (1971) with slight modifimfion. Briefly, 100 pl of ribosome solution (0.4 pg ribosome/ml of 10 mM Tris-HCl buffer, pH 7.4, 50 mM NH4CI, 5 mM Mg acetate, 5 mM dithiothreitol) was mixed with 10 pl Of standard buffer (10 mM Tris—HCl buffer, pH 7.4, 50 mM NH4CI, 5 mM Mg acetate, 5 mM dithiothreitol) and 10 pl of 3H-T-2 solution (0.2 pCilml). After incubation at room temperature for 30 minutes, this mixture was mixed with 60 pl precooled absolute alcohol and then incubated at 4°C for 30 to 90 minutes. Ribosome-bound 3H-T-2 was separated from the solution by centrifugation at 30009 for 20 minutes at 4°C. One hundred (100) pl of the supernatant was carefully removed and mixed with 4 ml Of scintillation cocktail (Research Products lntemational Corp.) into scintillation vials. Radioactivity was determined by liquid scintillation counting using a Packard TRICARB 4430 Liquid Scintillation System (Packard Instrument InC., Downers Grove, IL). 160 The resUltant value provided an estimate for the concentration of the labeled toxin in free solution at equilibrium. For control, the total radioactivity was determined in dupliwte samples in which ribosomes were omitted. The difference between the total radioactivity and the radioactivity in free solution gave an estimate for the concentration of ribosome-bound compound. Competitive binding assay was used to determine whether nonradiolabeled T-2 toxin or mouse sera competed with tritium labeled T-2 toxin for trichothecene- ribosome binding site. Briefly, 100 pl of ribosome solution (0.4 pg ribosome/ml of standard buffer) was mixed with 10 pl of serially diluted (0-1000 nglml) T-2 toxin standard solution or 10 pl of appropriately diluted mouse sera. After incubation at room temperature for 30 minutes, 10 p1 Of 3H-T-2 solution (0.2 110le) was added and the mixture was incubated for another 30 minutes at room temperature. The binding assay was then completed as described above. Mouse immunization Twelve female BALBIC mice (6 to 8 weeks of age, Charles River Laboratories, Wilmington, MA) were immunized by intraperitoneal (i.p.) and subcutaneous (s.C.) routes. The mice were immunized four times with an emulsion of 608 or 808 subunit of yeast ribosomes with Freund’s adjuvant at a ratio of 1:1 (vol.lvol.) at two-week intervals. The first immunization was performed at week zero at a dose of 50 p9 ribosome/mouse and the ribosome was emulsified with Freund’s complete adjuvant. The second, third and fourth injections were performed at week 2, 4 and 6 at a dose of 25 p9 161 ribosomes/mouse and Freund’s incomplete adjuvant was used to emulsify the ribosomes. Ten days after the second, third and fourth immunizations, methoxyflurane-anesthetized mice were bled from the tail vein and the blood was collected with a heparinized tube. The blood was incubated at 4°C overnight and then centrifuged at 10009 for 15 minutes to Obtain mouse plasma. Antibody titer and specificity were then determined by indirect ELISAS as described below. Indirect ELISA. Indirect ELISA was performed by a modification of the procedure of Azcona-Olivera et al. (1992a) and used to determine serum titers. Briefly, wells of polystyrene microtiter plates (lmmunolon 4, Dynatech Laboratories, Alexandria, VA) were coated overnight (at 4°C) with 100 pl of ribosomes (5 pglml) in standard buffer (10 mM Tris-HCl buffer, pH 7.4, 50 mM NH40l, 5 mM Mg acetate, 5 mM dithiothreitol). Plates were washed four times by filling each well with 300 pl of the standard buffer and aspirating the contents. Nonspecific binding was blocked by filling the wells with 300 pl of 1% (wt/vol) ovalbumin in the standard buffer. After incubating for 30 minutes at 37°C, the plates were washed four times with the standard buffer. Fifty pl of serially diluted mouse serum was added to each well and incubated at 37°C for 30 minutes. Wells Of serially diluted preimmune serum were used as control. Unbound antibodies were removed by washing four times with the same buffer and 100 pl of goat anti-mouse IgG peroxidase conjugate (2 pglml of the ovalbumin solution) was added to each well. The plates were incubated for 30 minutes at 37°C, washed eight times with the standard buffer 162 and then rinsed twice with distilled water. Bound peroxidase was determined with ABTS substrate [1 ml of 35 mg 2,2’-azinobis(3-ethylbenzthiazolinesulfonic acid)/15 ml distilled water mixed with 11 ml of Citrate buffer pH 4 and 8 pl hydrogen peroxide] as described previously by Pestka et al., (1982). Absorbance at 405 nm was read with a Vmax Kinetic Microplate Reader (Molecular Devices Corporation, Menlo Park, CA). Titer of each serum was arbitrarily designed as the maximum dilution that yielded twice or greater absorbance as the same dilution nonimmune control serum. A competitive indirect ELISA (Cl-ELISA) was used to test the potential for T-2 to block the binding of antibody to yeast ribosomes. verify specificity of antibodies in sera toward T-2 toxin. Briefly, microtiter plates were coated and blocked as described in the indirect ELISA procedure, and then 50 pl of serially diluted (0-10,000 nglml) T-2 standard solution was simultaneously incubated with 50 pl of appropriate dilution of antibodies over the ribosome solid phase for 30 minutes at 37°C. The assay was then completed as described above. RESULTS AND DISCUSSION Ribosome preparation Four liter yeast cultures were prepared by inoculating trichothecene- sensitive yeast into 4 liter liquid medium and then incubating it at 30°C with slow shaking for one week. These cultures were harvested by centrifugation at 10,0009 for 5 minutes and resulted in approximately 80 g wet weight yeast cells. From these, approximately 50 m9 of 808 ribosomal subunit and 30 mg of 608 ribosomal subunit were prepared. RiboSomal purity was checked by calculating the values of ribosomal absorbance at 260 nm (A233)divided by that at 280 nm (A233). The A233! A233 values of the 808 and 608 ribosomal subunits were 2.08 and 1.95, respectively, indicating that the ribosomal subunits were pure since proteins including ribosomes are considered as pure or uncontaminated if the value ofA2331 A233 is greater than 1.80 (Spedding, 1990). Tritium labeled T-2 toxin was used in ribosome binding assays. 3H-T-2 was incubated with yeast ribosomes diluted in the standard buffer. As a control the same amount of the radiolabeled toxin and the standard buffer were mixed without the ribosomes. After separation of ribosome-bound 3H-T-2 by centrifugation, radioactivity Of the supernatant was measured. It was found that supernatant from ribosome supernatant had a lower radioactivity values (disintegration per minute, DPM) than that fiom the control solution indicating that some of the 3H-T-2 bound to 163 164 the ribosomes. This suggested that the trichothecene—ribosomal binding site was still active. When these ribosomes were reacted with the radiolabeled toxin in the presence of free T-2 toxin, the association of the 3H-T-2 to the ribosomes was inhibited by the free toxin (Figure 4.2). These ribosomes, which still had the capability to bind to T-2 toxin, were used to immunize female BALBIC mice. Mice immunization Initially, twelve mice were immunized and boosted intraperitoneally and subcutaneously with an emulsion of 808 or 608 yeast ribosomal subunits and Freund’s adjuvant. Ten days alter the booster injection, the mice were bled. The blood was incubated at 4°C overnight and then centrifuged at 10009 for 15 minutes to obtained mouse plasma. The mouse antisera were analyzed by indirect ELISA to determine their titers and specificities. These antisera had high titers (Figure 4.3) indicating that they could recognized solid phase bound ribosomes. However, these antibodies could not inhibit the association of trichothecenes to yeast ribosomes when used in Cl-ELISA. In addition, when these antibodies were incubated together with ribosomes and tritium labeled T-2 toxin during binding assay, they could not inhibit the association of the radiolabeled toxin to the ribosomes. Thus, these antibodies were not applicable for assays of “trichothecene load” in food samples. In subsequent experiments, ten mice were immunized subcutaneously (SC) using 803 ribosomal subunit because in the previous experiments, SC immunization yielded higher titer antibodies (Figure 4.3). Percent inhibition of T-2 toxin to the association of tritium labeled T-2 toxin to 808 ribosomal subunit was similar to that to 165 the 608 one (Figure 4.2). Moreover, preparation of the 808 subunit was much simpler than that of the 608 one. In this immunization, the 808 ribosomal subunit was mixed either with T-2 toxin (T -2—80$), Freund’s adjuvant (FA-808), Cholera toxin (CT-80$), or with a mixture of FA and CT (FACT-808) prior to animal immunization. All mice apparently produced antisera with high titers (Figure 4.4). However, these antisera could not inhibit the reaction Of trichothecenes and yeast ribosomes when they were used either in competitive indirect ELISA or in binding assays. Animal immunizations with 808 yeast ribosomal subunits either alone or mixing with adjuvants could not induce the production Of antisera that could be utilized in competitive inhibition ELISA for trichothecenes. This may relate to two possibilities. Firstly, trichothecene-ribosomal binding site might be modified during immunogen preparation or during metabolism in animal body, and thus antibodies that were specific to the binding site were not generated. Secondly, the antibodies specific to the binding site might have been produced by the immunized animals but in a small proportion as compared to non specific polyclonal antibodies. As a result, trichothecene inhibition to the specific antibodies were undetectable when these antisera were used in CI-ELISA for trichothecene detection. Thus, these results did ' not support the hypothesis of this current study. Direct injection of specific proteins or specific oligopeptides for the binding site into animals, rather than “whole” ribosomes, might be capable to stimulate the desired specific antibodies against the binding site. The specific proteins for trichothecene-ribosomal binding site could be isolated by comparing ribosomal components from trichodennin-sensitive yeast to that from trichodennin-resistant 166 one. Schindler et al., (1974) found an alteration of 608 ribosomal subunit in the trichodennin-resistant mutant. The alteration was associated with ribosomal protein L3 which was coded by tcm1 genes (Grant et al., 1976; Fried and Warner, 1981). Thus, injection ribosomal protein L3 from trichodennin-sensitive yeast into animals may be as an alternative to produce antibodies against the ribosomal binding site. Another alternative is immunization of specific oligopeptides for the binding site. The nucleotide sequence of tcm1 gene that encodes ribosomal protein L3 in trichodennin-resistant yeast strains has been determined (Schultz and F riesen, 1983). By comparing this nucleotide sequence and the nucleotide sequence of a gene encoding protein L3 In trichodennin sensitive yeast strains, specific nucleotide sequence for trichothecene-ribosomal binding site could be isolated. From this sequence, polypeptides could be constructed in vitro and might be used for the production of specific antibodies against trichothecene-yeast ribosomal binding site. CONCLUSION Both pure 608 and 808 ribosomal subunits have been isolated from trichodennin-sensitive yeast and used for the production of antibodies which were specific to trichothecene ribosomal binding site. These ribosomal subunits (either alone or after mixing with different adjuvants) were used to immunize 22 female BALBIC mice. All mice apparently produced high titer antisera. However, these antisera were not able to inhibit the association of trichothecenes and yeast ribosomes when they were used either in Cl-ELISAs or in binding assays for trichothecenes. Thus, the production of antibodies that were specific to trichothecene-ribosomal binding site could not be accomplished through animal immunization with either 608 or 808 subunits of yeast ribosomes 167 168 Figure 4.1. Competitive indirect ELISA for “trichothecene load” using yeast ribosomes for coating microtiter plates. Trichothecene ribosomal binding site specific antibodies (AB) compete with trichothecenes (T) for the trichothecene binding site (TBS). Second antibodies (SAB) which have been labeled with enzyme (ENZ) are then added to determined total bound antibody (AB). ”Trichothecene load” is inversely related to the bound enzyme-labeled antibodies and can be measured quantitatively using spectrophotometer after color development by the addition Of enzyme substrate. 169 110 100 - so 00% 70— 50— 40— PERCENT INHIBITION 30— 20— . 803 RIBOSOMAL SUBUNIT A 603 RIBOSOMAL SUBUNIT I I I I T I I I I I I -100 0 100 200 300 400 500 600 700 000 900 1000 1100 Figure 4.2. T-2 TOXIN (nglml) Inhibition of T-2 toxin to the association of °H-T-2 toxin to yeast ribosomes. Each data point represents the mean value of triplicate measurements. One hundred pl of yeast ribosomes (0.4 mglml standard buffer) were reacted with 10 pl of 3H-T-2 toxin (0.2 uCi/ml) in the presence of 10 pl -of serially diluted (0 to 1000 ng) non radiolabeled T-2 toxin and 60 pl of precooled alcohol. As a control the same amount of radiolabeled toxin was mixed with the standard buffer without ribosomes. After ribosome-bound 3H-T-2 toxin was separated by centrifugation, radioactivity (DPM) of 100 pl of the supernatant was measured by liquid scintillation counting. DPM values Of control solution and 0 nglml supernatant were 16,026 and 8,577, respectively. Percent inhibition = [(DPM value of certain n9 T-2 toxin lml supernatant - DPM value of 0 nglml supematant)l(DPM value of control solution - DPM value of 0 nglml supematant)] x 100%. 170 2.0 )— 1-3 -_ 0 sc-80s Z O lP-80s . —” ~ 1 . 1 ° - A sc-sos I A IP-sos 1.4 —- _ E : -- .3 1.2 — 3. : d. : 0 1.0 — 0.8 ---h “ 1 0.6 _‘- /‘\/ 0.4 —- ‘ 1 I L I 1 I 1 1 l 1 1 1 I 1 I 10‘1 103 ANTISERA DILUTION Figure 4.3. Typical indirect ELISA titration of mouse antisera. The mouse antisera were obtained ten days after the third subcutaneous or intraperitoneal injection with 50pg of 608 or 808 yeast ribosomal subunit emulsified with Freund’s adjuvant. Each data point represents the mean 1: standard error of the mean (n=6, duplicate measurements Of three mice). IP, and SC indicate intraperitoneal and subcutaneous injections, respectively. 808 and 608 indicates 808 and 608 yeast ribosomal subunits, respectively. 171 2.0 1.8 J D i O 1.6 — IIII d . 1 IVI TI 4\. 1.2 —“‘ I 80s ONLY FA - 80s ' . 0 CT - 30$ / 1- A FACT - 808 IITI 1.0 - 0.0. (405 nm) rIII 08— O A T-2 - 008 0.6 — IIjfiIITI 0.4 H 0.2 — jIIIIIII 10‘ . 10*2 ANTISERA DILUTION Figure 4.4. Typical indirect ELISA titration of mouse antisera. The mouse antisera were obtained ten days after the third subcutaneous injection with 50pg of 808 yeast ribosomal subunit mixed with different adjuvants. Each data points represents the mean :I: standard error of the mean (n=4, duplicate measurements of two mice). 808 indicates 80$ yeast ribosomal subunit. FA, CT, and T-2 indicate F reund’s adjuvant, cholera toxin, and T—2 toxin, respectively. APPENDICES 172 APPENDIX A: MEDIA FOR HYBRIDOMA PRODUCING MONOCLONAL ANTIBODIES 1. Chemical and reagents All organic solvents and inorganic Chemical were of reagent grade or better. Penicillin/streptomycin solution (pen/strep) (100,000 units/ml), sodium pyruvate, polyethylene glycol (MW 1450) (PEG), hypoxanthine, aminopterin, thymidine, pristane, and dimethyl sulfoxide were purchased from Sigma Chemical CO. (St. Louis, MO). Dulbecco’s Modified Eagle’s Medium (DMEM, in powder form for 1 l medium/bottle), NCTC supplemental medium, and fetal bovine serum (PBS) were Obtained from Gibco Laboratories (Grand Island, NY). Tissue culture plasticware was purchased from Corning Laboratory Science 00. (Corning, NY). The myeloma cell line P3INS1/1-Ag4-1 (NS 1) (ATCC TIB 18) was purchased from the American Type Culture Collection (Rockville, MD). Macrophage conditioned media (MCM) was prepared as described by Sugasawara et al. (1985) II. MEDIA Dulbecco's modified eagle's medium (DMEM) One liter Of distilled water was poured into a 2 liter Erlenmeyer flask. Powder of DMEM was added to the flash and stirred with a magnetic rod until the powder was dissolved. Sodium bicarbonate (3.7 gll) was weighed and added so that the medium color Changed from orange to red. pH of the medium was measured and adjusted to neutrality (pH 7.0) either with 1N NaOH or 1N HCI. Usually the medium was slightly basic so that 4-5 ml of 1N HCI was added. Prior to sterilization with filter sterilized (Nalgene disposable filter unit catalog number. 155-0020; volume: 150 mL; memb mat type: C.A./T.A; color code: yellow, pore size: 0.2 mm), 10 ml Of NCTC suplement and 10 ml of sodium pyruvate were added to the neutral medium so that the DMEM contained 1% NCTC and 10 mm sodium pyruvate. The sterilized DMEM medium was stored in a sterilized and labeled 500 ml bottle at 4°C and tested for its steril' by incubating two separte 1 ml of the medium into wells of a 24-well plate at 37 C in an incubator containing 5% 002 for 2 days. If the medium was contaminated by microorganims, the medium has to be refiltered, and then rechecked for its sterility again. 173 Maintenace medium Twenty ml of fetal bovine serum (FBS), and 1 ml of Pen/Strep (PIS) solution were added to 80 ml of the sterilizied DMEM medium to make 20% PBS-DMEM. The 20% FBS-medium was filter sterilized (Nalgene disposable filter unit catalog number. 155-0020; volume: 150 mL; memb mat type: CAITA; color code: yellow, pore size: 0.2 mm) and stored at 4°C. One hundred x stock solution of hypoxanthine-thymidine (HT) One hundred x stock solution of HT was prepared by suspending 166.1 mg of hypoxanthine (H) and 38.75 mg of thymidine (T) in 50 mL of distilled water. The H and T were dissolved by adding (0.1N or 1.0N) NaOH dropwise until H and T were solubilized. The volume was adjusted to 100 ml with distilled water, filter sterilized, aliquoted in 10 ml, and stored at -20°C. Fifty x stock solution of HT Fifty x stock solution of HT was prepared by adding an equal volume of 100 x Stock solution HT to an equal volume of the sterilized DMEM and stored at 4°C. Aminopterin stock solution Aminopterin stock solution was prepared by adding 17.6 mg of aminopterin (A) to 100 ml of distilled water and stored at -20°C in aliquots of 10 mlltube. Fifty x Stock Solution of HAT Fifty x Stock Solution of HAT was prepared by adding 50 ml of 100 x stock solution of HT and 5 ml of aminopterin stock solution to 45 ml of the sterilized DMEM, filter sterilized, and stored at 4°C in aliquots of 12.5 mlltube. HAT medium HAT medium was prepared by adding 20 ml of PBS, 1.25 ml of PIS, and 2.4 ml of 50 x HAT stock to 100 ml of the sterilizied DMEM, filter sterilized, and stored at 4°C. 174 HT Medium HT Medium was prepared by adding 20 ml of FBS, 1.25 ml of PIS, and 2.4 ml of 50 x stock solution of HT to 100 ml of the sterilized DMEM, filter sterilized, and stored at 4°C. Macrophage Conditioned Medium (MCM) Five female BALBIC mouse (6 to 8 weeks of age, Charles River Laboratories, Wilmington, MA) were sacrificed, dipped in 70% ethanol in water to sterilize their whole bodies, and peeled back their skin to expose their peritoneal lining. Ten ml of 20% PBS-DMEM was injected to mouse’s peritoneal cavity using an 18 gauge needle. The peritoneal cavity was tapped several times to dissolve macrophage cells into DMEM which was then drew off using the same needle. The DMEM containing macrophage cells was centrifuged at 450 x 9 for 7 minuted. The cells (sediment) were resuspend in 40 ml of 20% FBS—DMEM, and placed into two large T-flasks (20 ml each). After incubation at 37°C in an incubator containing 5% CO2 for 3 days, the medium was collected, and the cells were rated with 20 ml of fresh 20% F BS-DMEM medium. The medium was harvested 2 more times at 3 day intervals. The medium was stored at -20°C. Ten or 20% macrophage-HT medium Ten or 20% macrophage-HT medium was prepared by adding 10 or 20 ml of MCM to 90 or 80 ml of HT medium, filter sterilized, and stored at 4°C. Cloning medium Cloning medium was prepared by adding 20 ml of MCM to 80 ml of 20% FBS-DMEM, filter sterilized, and stored at 4°C. Lysing buffer Lysing buffer was prepared by adding 8.29 9 of NH4CI, 1.0 g of KHCO3 and 0.037 g of EDTA tO 1 liter of double distilled water, filter sterilized, and stored at room temp. 175 APPENDIX B: SAMPLE LISTS OF CORN AND CORN PRODUCTS Table 5.1. Number and description of food samples bought in retail supermarket in Mid Michigan, in September, 1991. SAMPLE: DESCRIPTION # 6 Self-rising white, corn meal 7 Yellow corn meal 8 Self-rising white, com meal mix 10 Yellow corn meal 21 Self-rising white corn meal 22 Self rising white corn meal mix 24 Corn tortilla mix 25 White yellow plain enriched corn meal 32 Yellow corn meal 33 White corn meal 38 Seven grain cereals 41 Corn cereals 43 Corn flakes 48 Corn meal H9 Corn meal 176 Table 5.2. Number and description of Italian feed samples. SAMPLE DESCRIPTION SAMPLE DESCRIPTION NUMBER NUMBER F1 Feed for goat F17 Feed for laying Chicken F2 Feed for milk cow F18 Feed for swine F3 Complementary feed for horse F19 Feed for swine (45% maize) F4 Feed for calf F20 Starter feed for swine F5 Feed for Chicken F21 Feed for swine F7 Flour for pig F22 Prestarter feed for swine F8 Feed for swine on phase Of growth F23 Feed for pregnant cow F9 Feed for milk cow F24 Feed for Chicken F10 Feed for swine F25 Silage maize F11 Feed for milk cow F26 Feed for swine F12 Flour of maize F27 Feed for laying chicken F13 Feed for swine F28 Feed for swine F14 Feed for milk cow F29 Feed for buffalo F15 Feed for calf (50% maize, 50% F30 Feed for calf barley) F16 Feed for rabbit F31 Feed for calf 177 Table 5. 3. Number and description of fresh corn sample harvested from five counties in Michigan in Summer, 1994a SAMPLE NAME AGE (DAYS) LOCATION NUMBER P1 CaLgill 3777 ' 98 Montcalm P2 Cagill 4327 104 Montcalm P3 Dekalb 0K 527 102 Montcalm P4 Pioneer 3293 1 13 Monroe P5 Pioneer 3394 110 Montcalm P6 Amcom 5930 110 Cass P7 Callahan 07337 97 Cass P8 Callahan C7446X 103 Cass P9 Cargill 3777 98 Cass P10 Carfll 7777 1 15 Cass P1 1 Counflmard 432 100 Cass P12 Dekalb 0K 471 97 Cass P13 Dekalb 0K 560 106 Cass P14 Mycogen 6060 107 Cass P15 Peyco 614 98 Cass P16 Pioneer 3293 1 13 Cass P1 7 Pioneer 3525 1 06 Cass P18 Renk 646PT 105 Cass P19 Rusb XR-1727 106 Cass P20 Callahan 07252 107 Huron P21 Cargill 3777 98 Huron P22 Cargill 4327 105 Huron P23 Cargill 5547 106 Huron P24 Country Mark 432 100 Huron P25 Dairy cand Stealth 1205 105 Huron P26 Decald 0K 471 97 Huron P27 Dekalb 0K 527 102 Huron P28 Mycogen 3440 93 Huron P29 NK 4242 100 Huron P30 Northrup King N K4242 100 Huron P31 Peyco 614 98 Huron P32 Pioneer 3394 1 10 Huron P33 Pioneer 3525 106 Huron P34 Renk RK 657 106 Huron P35 Rupp XR 1727 106 Huron P36 Amcorn 5930 1 10 Monroe P37 Callahan 07337 97 Monroe P38 Callahan C7446X 103 Monroe P39 Cargill 7777 1 15 Monroe 1 78 Table 5.3. (Cont’d) SAMPLE NAME AGE (DAYS) LOCATION NUMBER P40 Country Mark 432 100 Monroe P41 Dekalb 0K 569 106 Monroe P42 Dekalb OK 471 97 Monroe P43 Mycogen 6060 107 Monroe P44 Northrup King K4242 - Monroe P45 Peyco 614 98 Monroe P46 Pioneer 3525 106 Monroe P47 Rank 646 PT 105 Monroe P48 Rupp XR 1727 106 Monroe P49 Cargill 3777 91 - Montcalm P50 Cargill 5877 108 Montcalm P51 Country Mark 432 100 Montcalm P52 . Decald 0K 471 97 Montcalm P53 Junflg2672 108 Montcalm P54 Mycogen 3440 93 Montcalm P55 NK 4242 100 Montcalm P56 Peyco 614 98 Montcalm P57 Pioneer 3394 1 10 Montcalm P58 Pioneer 3525 106 Montcalm P59 Rank 657 106 Montcalm P60 Renk RK 657 106 Montcalm P61 Rupp 1727 106 Moutcalm P62 Callaahan 7446 x 103 Saginaw P63 Callahan 07337 97 Saginaw P64 Cargill 3777 98 Saginaw P65 Cargfl 4277 102 Saginaw P66 Cargill 6677 1 10 Saginaw P67 Clba 4394 107 Saginaw P68 Country Mark 432 100 Saginaw P69 Dekalb 0K 471 97 Saginaw P70 Dekalb 0K 569 106 Saginaw P71 Mycogen 6970 107 Saginaw P72 NK 4242 100 Saginaw P73 NKX 423 105 Saginaw P74 Peyco 614 98 Saginaw P75 Pioneer 3394 1 10 Saginaw P76 Rank 646 PT 105 Saginaw P77 Rupp XR 1677 106 Saginaw ‘ kindly supplied by Dr. Patrick L. Hart (105 Pesticide Research Center, Michigan State University, Ml, 48824. APPENDIX C IMMUNOLOGICAL ASSAYS FOR MY COTOXIN DETECTION 179 Immunological Assays for Mycotoxin Detection Enzyme—linked immunosorbent assays have been success-fully applied to the screening of mycotoxins in a diverse array of foods James J. Pestka. Mohamed N. Abouzied. and Sutikno D MYCOTOXINS ARE TOXIC SEC- ondary metabolites produced by molds that often contaminate agricultural stao plea such as corn, wheat, and peanuts rior .to harvest and during storage. ese compounds have a wide array of chemical structures and are produced by common field and storage fungi, includ- many s 'es of Aspergt'llus, Perri. Mycotoxinscanelicitavariety oftosic symptoms in humans and animals, - ing from gastroenteritis to cancer (Tab e 1). For example, the aflatoxins were identified in the early 19603 as etiologic agents of hepatotoxicity and hepatic cancer in hark poults and rainbow trout. respects y (Pestka and Casale. 1990). Strrct regulations were subse- quently established for aflatoxins in food inmanycountriesbecsuseot’the poten- tial for similar effects in humans. Besides direct concerns over human health. aflatoxins and other mycotoxins have major economic impact on live- stock productivity as a result of lower quantrtyand quality of animal produgsd, smaller litters, infertility, reduced 1' ' ' ' resistance to. dis- Whetheram foodielargelyd‘ixotaid“l talandbiologicalfactorsfi’ig. 1).par- tiarlarlytheregionalweatherduringa means ehminatmgmycotonnstrmn hmnansndanimalt’oodntodetectend ntaminatedrswmaterialsfipm Tabla I—Mejor Mycotoxins sndtheeroxlc Effectshemerirmrdanimdmodsb Mycotoxin Tealeeflect M AflsroeinsB..02.Gr.Ga Livsrtosicitysndcsnosr WWW. m AflatoaLiM. Liversoaicirysndcsnosr Milt Cydooissonicsa’d Musdsnesrosimorsl Cerium lesions Fm l ‘ ., ' Com pdmonsryedsms Odrstoain WM.” monuments cats PM Medicaid" mumps}: net-staid" Tridmmirdufing Fesdreltrsstdermea. Madman-11w Winni- orslerdGIIesions. MuiMT-Z W rosin Zearalenone Mamas."- Column!" Mutations utnwheagcottonseed.andother mimosorbentassaymfl‘heseand 1 ssvrellasclinicalsamplestrom otheressaysheveheenmsdeavailable htunansandanimalsAseriesot’esten- comm ' {fortherspidusayofmy- sive cleanup steps involving liquid-liq- ootoxins in cod. uidpartition.columncleanup,andevap- Key considerations in the orationarethereforerequiredtoover- meatand ' ' 01' ' im- come these interferences. '11: munoassaystot’oodmanalydsoin- proceduresarstime-consmningand cludemethodot’an generation. costlyandofteninvolveuseofharmful immunoassayt’ormats. to solvents. food andysigandevalustion criteria {or Researchinitietedinthelatelm oommercialtesthits. roposed the application of immuno- ghemical rocedures commonly medindm toriestotheanal- ysis of aflatoxin B; (AFB and other mycotoxins (Chu and 1977; LangoneandVanVunakisJWG .Theae assaystypicallyinvolvethecompetition between a free woman in a sample extractant! a edmycotoxin {oran antr’bodybindingsiteAl initially basedonrsdioimmunoasay sub- sequent research has established the feasibility of using enzyme-linked im- Author Pestka. a Professional Member d [FT isProtessor.andauthorsAhoozisdand ' State University.EastLansing.hfl48824.Sendre- print requests to author Pestka. Generation of Mycotoxin Antibodies Antibodieamrhnmmoglobulimarea 10:3 of glycoproteins that are pro- d asa foreiflzolewles in the ham and 1988). This ' the interaction responserequlres among white blood cells (leukocytes) knownasBcelIaToelhandmacro- phagethatculminsteintheterminal difl'erentiation of the B cells into anti- body-eecretingplasmaoells. muneresponseandisdepend Chemicalstructnreanditsabilitytope recognisedasforeignmaterialtlhemm- imum moleatluweightofanrmmuno- genis3,000-5,000. Because mycotoxins are oflow molecular ’ ht (300409). they must be earnings to a‘camer 180 Tabe—EconomiefiffectsofMyco— tordncontamhsdcnhlood. W MCASTHSOQI Harder/miter International trade geroteinsuchasbovineserumalbuminto Imm Imogemc. . Often. preparation of a suitable my- cotoxin immunogen is the rate-limiting step in the development of an immu- noassay. Standard hapten conjugation techniques used for mycotoxins have been reviewed by Chu (1986). lithe my- cotoxin doesn’thsve a reactive up for conjugation. it must be derivatized. [for example, generation of protein conju- gate {or the mycotoxin fumonisin, (Asco- na-Oliver'setal..19923.b)srm lym- volves the use of glutaraldehyde ° conjugs o eoxynrval Casalee al.. 1988) is much more dificult because cchng’ stages (Fig. . nee-ally, e same conjugation techniques used for immrmcgen preparation can be applied to link to to enzyme markers forthe Mlongssthereaction conditions do not denature the enzyme. In some cases. undesirable side reac- tions can occur during chemical conju- 'onandcanresultinantibodrestothe - roducts (Gendlofl' et al., 1986). An- ‘es may also react with mycotoxin plus bridge bridge group. plus carrier protem, or the carrier protein di- rectly. Thus, when ’ ° and evaluating mycotoxin antibodies for im- munoassay, conjugation reaction proto- HARVESTING an Molsture STORAGE Temperature Detection/Diversion iii HUMANS ; 59. I—FectorsAffectir-Iglllycosoxln Oeewreneshdrelcoddrshfiomfcaeandthsefe (1990! colsmustbecarefullyselectedandap— propriate controls utilised. . A key aspect in antibody development 18 the site of chemical conjugation For example, many approaches have been used to roduce antibodies with differ- ent specrficities for the aflatoxin family (F111. 3). Those portions of the aflatoxin m enile which project distally from the conjugation site are said to be immuno- dorrrmont, became the resultant anti- bodies will exhibit the highest degree of recognition {or these moieties. Thus. any metabohc precursor or analogues which mimic this immunodominant re- gronwillberecognisedbyanantibody generated against the t toxin. Cross-reactivity can assessed using RIAorELISAcompetitioncur-veawhere the levels required for 50 96 inhibition of marker ligand ’ ' are as the bastsofcomparjson ig.4).Rarelyare these competrtron curves superimpos- able; thus, an analogue typically cross reacts to a greater or lesser extent than the parent toxin. Although the presence ofsuch analogues in a sample may ren- der a quantitative ass to the level of semiquantitative, a high level of cross- reactivi can beveryuseful. ashasbeen o for the screening of fumonisins (Ascona-Olivera et al.. 1992a. b), zearale- nones (Dixon et al., 1987; Warner and Pestka. 1986) and the aflatoxins (Dixon et al.. 1988). The most straightforward approach to generation of antibodies is the multi pie-site immunization of rabbits with too-1,000 pg of mycotoxin—protein con- jugateUaa leantiserumcouldbeob— tunedinHmdAaiticalfsctorinthis immunizationproceaaistheuseofan oil-based adjuvant MW Mycobacterium such as the ’s Complete type” to allow slow release of the immunogen and nonspecifically stimulate the immune nae. More recently. we have utilized very low levels of cholera toxin conju- its of filrsrgrsdm (Axons-Olivera at 19920, a trichothecene myco- toxin (Abouzied et al.. 1993). Although the mechanism“) by which choleratoxrn (CT) exerts the potent adjuvant efl’ect in the immune system is not fully under- stood. it has.been shown that CTcon- comrtantly stimulates interleulnn' -1 pro- duction and antigen presentation (Bro- FFRR' ’Anv ’oo‘- fnnn V'hl Ode-- - Immunological Assays (continued) mander et al.. 1991). The advantages to this a proach are severalfold. First, the p urejs rapid and yield; quality ann'bodieczlm cor; panson poorer results 'eved standard tocols. Second. since no thimpairmentwasobserved at the concentrationsused inthiswork. mt be a humane alternative to . 'sadyumtwhichtypicallygives rise tooa. ulcers. or granulomas atthermectionsiteAndthrrd.theuse ofCT is also valuable when mycotoxin availability is limited. since relatively 181 uircdto yre- low doses of immunogen are induce a rapid and strong anti spouse. Rabbit antisera contain antibodies generated by multiple B-cell clones. Thesevarym ’ city andareconsid- cred yelonal. A moor advantage of l antisera are that they are of ‘ afinity and inexpensive to pro- duce. However,inherentvariability£rom lottolotmakesitdifimlttometbem in commercial kits with defined perfor- mancecharacteristica.Hybridomas have therefore been developed by fusions of immunized mouse spleen cells with a myeloma cell line to secrete reagent- mhéty monoclonal antibodies. A maJo' r ° vantagetothisapproachisthere— quirement for a tissue-culture facility, high cost. and time efl'ort involved. Mycotoxin Immunoassay Formats A number of immunoassay formats have been devised for mycotoxin analy- sis. Initially, competitive RIAs were used whereby specific antibody is incubated with a constant amount of radiolabeled figZ—Preparationotlrmnuiogensbr wmwmmwloom 122 $600 rsmocv—sssauanv 1995 . #4:»! ' o - ' . Vi toxininthepresenceofstandardorun— known sample and then various proce- duresareusedtoremove the toxinanti- body complex from solution. The amount oftoxin in a sample is inversely' related to the amount of radiolabeled (imbound) toxin in solution. Because of ' inherent problems with radioactivity. titive assaysbasedonELISAGEIn- andPerlman, 1971) were devised. thdirectand indirectassays (Fig.5) have been applied to mycotoxin detec- tion. Microtiter plates. beads. and Terasaki plates have been used as solid- 182 phase support for ELISA (Pestka et al., 1980' Pestka and Chu. 1984). High-pro- tein-binding polystyrene microtiter plates have been most widely used be- cause they ofl'er an extensive support technglzfy, ,including removable stripe. multi pipettes, automated washers. and spectrophotometers. Membranes are an alternative solid phase that have been employed for yea-no or threshold tests in cups, cards, and di 'cks. We have successfully em- pl nitrocellulose membranes in a Computer-Assisted-Multianalyte Assay System (CAMAS) for fum afla- toxins, and zearalenones (Abouzied and Pestka, 1994). Monoclonal anti bodies for each of these toxins are immobilized as multiple lines on niuocellulose' mem- brane strips and sectored into hydro- phobic compartments to minimize use oiments. A modified ELISA rs wand whereby tree m horseradish peroxi beled myco- toxins compete for binding to them cellulose-bound aneta'bggiea. Color inten- sr 0 lines cm a precipitating tEstate is inversely related to myco- NOD-CC. MOO-0.... >625: 0:8? w~w~w§9 me?” ”30:63? ”.2005? canteen-m fia3—prWinmtlondaMW ISA W“? l o e a Pereeat Ilealrael Mose 8 mam—Mummy» mmumham med Wlbwnrmwmnm MMdWZklass Wmmfimtafl‘flbd— M irradiated with mp- gaead and. 9 over Wane-born! ”MAB. Tosh concantratimlsh- Mutatedtobandenrymaconlw gateandrhmcanbecalcdaredmdavel- Ubourdmtbody. Tmmb Watered ao borndenzyme conju- gate FEBRUARY 1995—FOOD TEONOLOGY 123 lmmunological Assays (continued) 183 Monitor | Z NOW Antibody lines Hg. BMWMMMhWWWWIU mammmnmslum 1. SEPARATE HAPTENS ON CHANNELED HPTLC SILICA GEL PLATE (I) 2. IMMUNOSPECIFIC TRANSFER: r g FILTER PAPER (F38) —_ ANflBODYCOA NC_(-) _ HPTLC (-) 8 8 8' 3. meme _ WITH HAPTEN-HRP CONJUGATE (8) 4. INCUBATE NC WITH PRECIPI‘I'ATING HRP SUBSTRATE. IDENTIFY HAPT'ENS AS INHIBITORY BANDS 5. PERFORM SCANNING DENSITOMETRY fig. 7—EUSAGRAM Procedure for Mycotoxins. From Pestka (I991) 124 F000 TECHNOLOGY—FEBRUARY 3995 toxin concentration. Line density can be quantitatively assessed using a camera, video monitor, and microcomputer equipped with a video digitizing ' board (Fig. 6). The assay can be used to deter- mrne range values for the various myco- toxinsinexuactsofspikedcominless than30minandrecordthemonthemi- comm uter hard disk. Ano er immunoblot approach, called ELISAGRAM, has been devised that combines the sensitivity and selectivity of competitive ELISA with the capacity of high-performance thin-layer chroma- tography (HPTLC) to separate struc- turally related mycotoxins (Pestka, 1991). The procedure (Fig. 7) involves separation of mycotoxin by HPTLC, blotting of the HPTLC plate with nitro- cellulose (NC) coated with mycotoxin- specific monoclonal antibody, incuba- tion of NC with mycotoxin-enzyme con- ‘ugate to identify unreacted antibody inding sites, detection of bound en- zyme conjugate with a precipitating sub. strata, and visual or densitometric as- sessment of inhibition bands indicative of a cross-reacting mycotoxin. The tech- nique has been applied to two major mymtoxin families, the zearalenones and aflatoxins. Multiple standard curves for the zearalenones and the aflatoxins can also be constructed using scanning deusitometry. Cross-reactivity in ELIS- AGRAM curves are analogous to that found in com titive ELISA. This pro- cedure could widely applicable to the simultaneous quantitation and confir- mation of multiple ha tens with a single cross-reactive anti ‘ y. Mycotoxin antibodies have also been i “figs: 1’ ”I r “d ' l or or examp e, aflatoxins can 3% to an afinity column and then desorbed for subse- quent derivatization and fluorescence measurement (Trucksess et aL, 1991). This approach requires fluorescent my- cotoxin derivatives and additional in- strumentation (fluorometer). Another :Eproach is to quantitate mycotoxin in e afinity column eluates by chrolmgnawsnphy (renewed' by u. Immunoassay of Mycotoxins in Foods Antibodies (polyclonal and mono- clonal) havebeeumadetothemajor known mnotoxins, and both ELISAs andRIAs vebeensuceeasfully applied to the ' of mycotoxins in a diverse arrayof oodaTable 3 givease~ lected examples of reported immunoaso says for the aflatoxins, ochratoxins, tri- chothecenea, fumonisins, and other my- cotoxins, many of which can detect picogram or nanogram levels of toxin. ago mainf gmwu? protein struc-. 0 anti an enzyme co gate, immunoassays have to be can"; out in an aqueous system. In a liquid system such as milk, mycotoxins can lie analyzed directly (Pestka et al., 1981a), although the limit of detection can be Increased by cleanup and concentration —Text continued on page 127 184 Tabla 3—Selocted WWI Assays tor Mycotoxins In Foods . " Unit 0' Toxin Fervent Food analyzed detection Mm Allatcno‘n a. an Com. wheat. man s nglg EI-Naltb et al. (1981) butter ELSA Corn. wheat. pom 3 nglg EI—Nakb at al. (1981) m Pesriut butter 2.5 nglg Mortimer et at (1988) Corn. cotton-seed 2.5 nglg Dixon at al. (ISBBI Barisy 0.1 nglml. Ramaluishna at al. I1990I Allatoxins B. ELISA Peanut butter 0.3 nglg Morgan at al. and G; (1986” Aflatatin M. RIA Mat 0.5 nglg Pestka at d. (1981!» ELISA Milt 0.3 nglg Pestka et al. (ISBIbl 0.3 nglg Fan et al. (1984) 12 pglg Nieuwenhot at al. (1990) AC Milk so pglg Hansen (1990) Cydopiucnic add ELISA Buffet 30 pg/assay Heron arr! Weilel’ 1991 Ergot altaloid ELSA Wheat 10 nglmL Shelby and Kelsy 1992 Fumonisins ELISA Feed 250 nglg Arcana-Olivera at al. , (19928. bl firsamchromanone LC-ELISA Wheat. barley 5 only Yu and Chu I199“ Octratoxin ELSA Bartay 1.0 nglml. Rarnaluishne at al. (1990! 5.0 nglg Candish at al. I1988I 0.1 nglg Morgan et al. (1983) Pig kidney 0.5 nglg Morgan at al. (1986!) Wheat. meat. 1.0 nglg Sato at al. (1987) please Chicken meat. 0.1 nglml. Kama et al. wheat flow. (1989. 1990! porcine plasma. bovine sera PR Toxin RIA Grease 50 jag/9 Wei and Chu (1988) Ribr'atarin RIA Cdtta‘es 0.1 pg Davis and Stone I197” Sterigmatocystin ELISA Barley 0.01 pg et al. “98“ Trichothecenes: W ELSA Rim 1 nglg Kemp at d. (1985, valenol OW ELSA Crltm 16 09/01. M at al. (1989) scirpencl Vllhoat 0.3 ug/mL ms at al. "983i W RIA Corn. wheat 20 nglg Xu et al. IISOGI ELISA Corn 200 nglrnL Casale et al. (1988l Wheat 0.1 ng/assay Mile at al. (1990) W load I pglg Abfuasd at al. 199“ Nivalenol ELISA Barley 0.1 nglassay Itebudti et al. (199“ Roridn A E.SA Feed 5 nglml. W et al. (1988) 7-2 toxin ma Com. m 0.1 nglg Lea and cm (1981!) Mir 2.5 nglg Lee and Our (1981” ELSA Com 50 nglg W at d. (1984) Com. when 2.5 nglg Pestka et al. (1981“ .. MI: 0.2 nglg Fan Ct UL (ISM \Nhoot " 0.5 nglg Chin et al. (1938) Zearalenone ELISA Contestants“ Inn/ml. LiuetaLI1985l Corn 1 nglg Warner and Pestka "986) We 2.5 nglg Warner and Pestka toods (1987) FEBRUARY 1995—FOOD TECHNOLOGY 125 ' Immunological Assays (continued) 185 ru4wmmmmmm~um 1993 . m Annals “lees-y tum: mu m who» end- w W m EZ-Saaen‘ M ELSkWIi- 5-20 10 5.00-1.50 Mm Moses/hi ahead M”. WW6- 0.5 10 5.00-7.50 Mir Mons/III ahead Ocular-in mm 5 I0 5M7“ Corn Mose-Ital ahead 1’4 train 8.5K: Mae'- 11.5 IO 5.00-7.50 Corn Vin-I. Mal ahead leer-alarm» 85k” 50 10 5.00-1.50 Corn Mose-Ital ahead atePrube‘ m5, Whey 5.20 5 10.00 (hum Moan/MW“ “5.10. W ELSkCcp 5.10.20 5 4.00 mumps-nut “WWW 2000' hut-.eenonaeed. “WWW endIeeda One-Step M5. ELISA: 5 40 1.00 Commune-an Quantitative: withELSAreeder EUSA' W Nahum endIeede MM. ELSA: 0.5 ‘0 LI» Mi szhhwm Zeualarlene euse: 100 so I.” Cam.“ Muhammad-r w W M am 5 5.10 3.50 Comm _Vaerder&SAree¢er.m m Mind USOA-EGSaMAOACF‘nt Actienw W’ ELSA: 1M 11-20 5.50 CeruwheetJeerl ”www.mflel Hand m We.“ W fiancnia‘n 845k 500 12-20 5.50 Market.” thalerELISAreed-Jleaeflel. M We.“ Ode-train am 20 12-20 5.75 “EM“ Narmada.“ m 74w 85k 500 ".30 5.50 Calumet.” Via-deraISAreaa.oeaeIIel W W ELISA: 250 ”-30 5.50 Commune “casement.“ w Veratu' Anetta-r. EM 5 I5 4.00 Canoe-Ina. Wanna”. W mm mm MM. EBA: 0.15 40 5.50 “It WWW“. W Der-m6- flJSk :00 10 5.50 Can't-eat.“ WWW“. “and w 7-2 EBA: 50 30 em Contented Mariam”. herd-rune m 250 so em Mme.“ wmmnea. W Dosage. m5. EBA: 0.5 45 4.00 Canoe-numbed Wanna” W W Anetta-rial. White 1.5 45 em “umber! Warned-r.” .Afleeeet’ Anne-“m Meal- 1.0 to 1.00 Commie“ WWW. mun WWW“ ' WWMACW Alec-“n". Med- 0.‘ 10 tom MI; Warm «an M m Meal- 2 15 tom Cern WWW .9.) W W m Meal- 5 10 10.00 Corn WWW m M leeralerrerre Med- “ 10 ION Corn.“ WWW m Whammwm unmet-WM“ mummznrswmum fia—mmuzmm WMRMHGWSIW Wmmumrzm "Ila. L!“ m em os-nnoaau M m MA 0214: was» 126 'Fooo TECHNOLOGY—FEBRUARY 1995 186 on a Sep-Pak or afinity column. Food samples were 0 ' ’ y extracted with a solvent stan protocols, evapo- rated. reconstituted in an aqueous bone: for assay. However, based on our initial observation that mycotoxin- horseradish peroxidase and solid-phase antibodies retain suficient stability for ELISA when incubated with as much as 35% (w/vol) methanol (Ram et 1986a). a direct apgsroach was develo whereby solid su trates are blended with methanol-water extraction solvent and the extract analyzed directly or af- ter dilution. Immunoassay and various chromato- graphic methods for mycotoxin detec- tion in foods are usually comparable when they are performed in the research laboratory (Ram et al., 1986a. b; Chu et al., 1987). However, sometimes toxin- free food extracts m interftrilre ‘lirtlh mycotoxin-enzyme ' ingto eso i - phase antibody and therefore yield a low falseepositive response when compared to a standard curvzgrepared in extrac- tion solvent with b er. Samples can be diluted more extensively to eliminate this interference, but this will decrease sensitivity. Alternatively. interference can be mmimized by incorporating tox- in-free sample extracts dunng standard- curve preparation. Another factor that must be considered is sample H, which must sometimes be adjusted, prior to immunoassay, since antibodyhantigen binding occurs optimally at neutral pH. Commercial Mycotoxin Immunoassays Theaboveresearchhasledtothede- velopment of a number of commercial kits that have been marketed in the United States for food safety verifica- tion (Table 4). Commercial immunoas- say kits have generall performed well in routme' anatllyeee informed in the labo- ratory and e 5 d (Kocltsow and Tan- ner, I990; Azer and Cooper, 1991; Domer et al., 1993). However, when liar-wits et al. (1993) recalculated the precision per- fonnance parameters of collaborative studies for mycotoxins through 1991. they found that ELISA had somewhat poorer precision than thin-layer chro- ma pby and liquid chromatogra- phy. us. when adopting a commercial immunoassay for rapid testing of myco- tonns.food analysts mustcritically evalo ustethegeteminlightoftheirspecific needs an the limitations of the assay (Table 5). ha ed organisations ve provul' leadership in evaluating these tests. Of- ficial First Action approval has been given for detection of aflatoxins by AOAC International (formerly Associa- tion of Oficial Analytical Chemists) in various commodities. using microtiter- well (Park et al.. 1989a. b; Patsy st 1992), cup (Trucksess et al.. 1989). afinity-column (Trucksess et al., 1991) tests. The US. Dept. of Agriculture’s Federal Grain InspectionService (FGIS) has tested and approved a number of kits for qualitative screening and quan- titation of aflatoxins (Table 4). Table 5—Suggested Criteria IorAdop- o'onolsMyeoroxrn .San- marizedfiomPssdrslIQOBl LImltsofdetsctionsndaensitivity 0090 Wmfanpidsaeering ‘ snd/orqusntitation s 'fi’l Effectiveness of recommended ex- traction procedure Sample thrwd'aput Field stabiity Inter- and he's-assay reproducbility To further facilitate kit evaluation. AOAC International has created the AOAC W Instlitute with th; char-gee uatingan certrfymg’ ° rapi test 'ts used for safety screening of foods. During summer 1993, this Insti- tute evaluated aflatoxin test kits in con- junction with :d Memoranggrm 109g; ndetrl; standing tn Octo w: £958 whiéfinT ' P the Test Kit ormance estmg rogram or test kits that detect aflatoxins in grain. Upon successful evaluation using protocols by. FGIS,testkitssreoertifiedtoclaim “Performance Tested in Accordance ‘1 with Standards Established by FGIS for ~‘ Test Kits to Detect Aflatoxin Residues in Grain and Grain Products."‘More re-= cently. two deoxynivalenol ELISAs have been similarl certified by FGIS. It is anticipated t food analysts will be assisted by similar evaluation studies that focus on mycotoxins of potential health and economic significance, such as deoxynivalenol. the fum0nisins, och- ratoxins. and zearalenone. References mepmu. 1994.8i- multaneomscreeningoffumoniein Bushe- texin Br. and searalenom by line immuno- blot: A computer-assisted multanalyte as- say system (CAMAS). J. AOAC Intl. 77: ‘ L Ascona. J.l.. Brasel . m W£.sndP JJ. 1991. lmmunochesno icalaaseasment mycotoxinsin1989grain foods: Evidence for deoxynivalenol (vomi- toxin) contamination. Appl. Environ. Mi- 5.273333; .11., Abouzied, MM. Pie 0 0 w net. RD. and Pestka. J.J. 1992a. Produc- tion of monoclonal antibodies to the m otoxinsfumonisinsBl.B?.and83.J.A: ric. Food Chem. 40: 531-634. Assam-Olivera. J Abouzied,’ ner. RD. N 1992b. ' Olivera. .. Yoshizawa, T.. and Pestka, JJ. 1993. Pro- duction of antibodies to the tn- . 4.15-diaeetylmvale- Aser.M.andCooper.(1199LDetermination ofaflatoxinsinfoodsusingl-IPLCanda commercial ELISA system J. Food Pro- BromarfdezgtgglmgremJ andL N. 03o 1991. Cholera toxin stimtllstu m ductionandenhancesan ’ presentation by macrophages in vitro. . Immunol. 146: 2908-29 Candlisb.A.A.G.. Stimson. W11. and Smith, J.E. 1988. Determination of ochratoxin A by monoclonal antibody-based enzyme im- munosorbent may. J. Assn. Ofie. Anal. Cagfialemglljsgg’lestka. JJ and Hart. LP e. c. e 1988. Enzymeolinked immunosorbent as. snag employing monoclonal antibody spe- ' c for deoxynivalenol (vomitoxin) and several analogues. J.Agrie. Food Chem. 36: CAST. 1989. Mycotoxins—Economic and health risks. Task Force Report No. 116. Council for Agricultural Science and Tech- nology. Ames. Iowa. Chiba. .. Kawamura. 0.. Kaji. PL. Ohtani. K. Nagayama. S.. and Ueno. Y. 1988. A semio tive enzymeolinked immunosorbent assay for detection of T-2 toxin with monoclonal antibody. Food Addit. Contam. 6: 629-639. Chi Fflm "" 37‘ m“. °" m}. “a; em ysis mycotoxin. “ . cotoxins and Phycotoxim,” 6th Intl. IUPAC Symp. on Mycotoxins and PhyootoxinaPre- toria. South Africa. July 22-25, 1985. ed. P. Step: and R Vleggaar. pp. 277-32. Chu. .S. 1992. Recent progress on analytical techni use for mycotoxins in feedstufl's. J. Animal Sci. 70: 3950-8963. Chu.t£.§.d and (long. 1977.8Productae‘: of an y It?“ toxin 1. App]. Vi- ron. Micro '01. Ch 17.8,!" rfimfi'amxe u. Y-C. 1987. proved enzymelinked im- munosorbent assay for aflatoxin B; in agri- cultural commoditieaJ.Asan.Ofic.Anal. Chem.70: 854-857. DavigRMandShneSSImProduetion ofantirubratoxinantibod snditeuseins redioimmtmoasaay for rubratoxin B. “Jr copatbologyG‘l: 29-31. Dixon. DE, Ram, PB, Hart, LP. and Pestks.JJ.1987.Hybridomacelllinepso- duction ofaspecificmonoclonalantibody totbemycotoximsearalenonsandalphe- searslenol. J. Agric. Food Chem. 35: 122- 126. ‘ D P JJ 8' ' MM... m. .1, ems: Hart. LP. 1988. Production of sensitive monoclonal antibodies to aflatoxin B and agatoxinMgandtbeirap° ° toELISA 1’ ads 37 field laboratories. J or toxin at ° . BAlaAkizbhg- 73:5,; JJ and Ch RS. issL'hbemmmzm'd tion “51me “h, in corn. ea peanut enzyme- linked immunosorbent assay and solid phase rsdioimmttnoassay. J Assn. Oflr. Anal. Cnhegd6; 1077-on 97 W almsn. . 1 I. m mam.“ ““’- mm we r rail... M“:&Pm§ mifm" an. Anindirectenzymedinked immutlnosorbent ass: for T-2 toxin in biological fluids. J. P fll’rotect.g7: sol-333.3 8.? audio 0 E-H-v Mk1- -e "m -e andHarLLPJSSLDetectionofT-Ztolin influariumsporotrichioides infectedcern by e e-linked immunosorbent assay. Appl. Viron. Microbiol. 47: 1161- 163. —Continued on next page am.- -.mmr r3 _ a. -- -.- '49.. Immunological Assays (continued) Gendlofl'. 13.11.. Casale. W.L.. Ram.B. P. .Tai. HJ... Pestka. J.J.. and llart. LP. 1986. Hapten-proteinconj tespreparedbythe mixed anhydrid e me ode: Cross reactive antibodies tn heterologous antisera. J. lm- munol. Methodol. 92 15-20. Hsck.R.Klafi'er.U.. andTerplan. G. 1989. A monoclonal antibody to the trichothecene mycotoxin diacetoxyscirpenol. Lett. Appl. Microbiol. 8: 71—76. Hslnau. S. and Weiler. RW.1991.Determi nation of the mycotoxin cyclopiasonic acid enzyme immunoassayJ .Agric Food to. 39: 1887-1891. Hansen, JJ. 1990. Afinity column clean up and direct fluorescence measurement of aflatoxin M1 in raw milk. J. Food Protect. 53: 75—77. Harlow. EandLane.D. 1988AntibodiescA laboratory manual. Cold Sfiring Harbor lymbfrstory. Cold Spring arbor. New or Hesseltine, CW. n198;}. Global signigcgn‘ce of mycotoxins. " ycotoxins an yco- toxins.” 6th IntL IUPAC Symp. on Myco- toxins’hnd Phycotoxins. Pretoria South Africa.July22-25. 1985. ed. PS. Steynand RVleggaar. pp. 1- 18. Elsevier Scientific Pub. 00.. Amsterdam. The Netherlands. Horwitx. W.. Albert. R. and Nesheim. S. 1993. Reliability of mycotoxin assays: An te. J. AOAC Intl. 76: 461-491. Ike i.H.. Teshims.R.Hirai. K..Sato.M.. Ichinoe. M.. and Tom. '1‘. 1990. Produc- tion and characterization of monoclonal antibodies to nivalenol tetraacetate and their application to enzyme-linked Immu- noassay of nivalenol. Biol. Chem. Hoppe- Seyler 371(1): 31-36. Kawamura.0.. Sato S.. Kaji. 11.. N ayama. S..Ohtani.l(.. Chi ...enoJ..andU .1989. Asensitive ELISA of ochratoxin A based onmonoclonal antibodies. Toxicon27: 887- Sato. S. Nagura.M.. Kishim- WWI. Sato. 8.. Uds.T.. Ito. Y.. mUean.L1990. ELISAfordetection andstnveyofochretoxininlivestockeers and mixed feeds. Food Agric. Immunol. 2(3): 135-144. MILAN-L. Mills, EW. C.. and Morgan. 1966. Ensyme-linked' Immunosor- pliedtoriceJ. Sci.FoodAgric.3‘l: 888-894. Kocltxow. D. and Tanner. S.1990.Com tive evalmtion of commercially available sflstoxintestmethodaJ.Assn.Ofic.Anal. Chem. 73: 584-589. J.J. and Van Vunakis. H.1976. toxin B; specific antibodies and their use in radioimmunoassay. J. Natl. Cancer Inst. 56: 591-595. Lee.S. andChu. F.S. 1981a. Radioimmum ss ofT-2toxinincornandwheatJ.Assn. c. Anal. Chem. 64: 156-161. Lee. S. and Chu. F.S. 1981b. Radioimmu- noaasa ofT-2toxin' tn biological fluidaJ. Assn. c. Anal. Chem. 64: 684-688. Liu.M.T..Ram.B.P.. Hart.L.P.. andPestka. J.J. I985. Indirect enzyme-linked immu- noassa for the m sear-ale enone. Appl. viron. MicrobioL 50. 332-336. Mennon. J.and andJohnaon. E. 1985. F mamthdamNewScientist 105(144 446 . Msrtlbauer. 8.. Garcia. M. and fTe I988. oregmsc Enxyme tmmunoaasa rocydic trichothecene rorichnor A- Produc- roperttesand use of rabbit antibod- ies. AEpK‘L Environ. Microbiol. 54: 225-230. “Em!“ C.. Johnston. ilélglé. linemp. H.A.. M011!!! M R A enzyme. linked immunosorbent assa 30%;:de to the "13de of wheat. Agric.42 225-233. 128 FOOD TECflNOLOGY—FEBRUARY 1995 187 Mills. E.N...C Alcick. S.M.. Lee.H.A..and Mo an. M. RA. 1990. AnELISAfordeoxy- niv enol tn wheat novel hapten derivation procedures. F Agric. lmmu- Mnol. 2(3): 109-118. MEWS. M.RA.. McNerney. R. and Chan. $1983. Enzyme-linked immunosor- bent assay of ochratoxin A in barley. J. Assn. Ofic. Anal. Chem. 66: 1481-1484. Mo M.RA.. McNemey. R. Chan. II. .S.. and Anderson. RH. 1986a. Ochrs- toxin Ain ig kidney determined by en- e. link immunosorbentaesay. J. Assn. fie. Anal. Chem. 37: 475-480. Morivn. M.RA.. K A.S.. and Chan. .3. 1986b. P uction of antisera against sterigmatocystin hemiacetal and its potential for use in an enzyme-linked immunosorbent assay for sterigmatocystin in barley. J. Sci. Food Agric. 37: 873-880. Mo an. M.RA.. . A.S.. and Chen. H. .S. 1986c. Aflatoxin determination in peanut butter by enzyme-linked immu- nosorbent assay. J. Sci. Food Agric. 37: 908-914. Mortimer. D. N.. Shepherd. M.J.. GilberLJ.. andM organ. M.RA. 1988. A survey of the occurrence of aflatoxin B; tn peanut but- ters by enzyme-linked ° ..amunosorhent as- say. Food Addit. Contam. 5: 127-13?. Nieuwenhof. F.F.J.. Roolwerf.J.D.. and Van- Den-Beden. J.W. 1990. Evaluation of an enzyme immunoassay for the detection of aflatoxin M1 in milk usi antibody-coated polystyrene beads. Mil wiseenschaft 45: 584-588. Park. D. L. Miller. BM. WHart. P.. Yang.“ 6.. McVey. J.. Page.S. ..Pestka.J Brown. LH ”aligns-linked tmmu- noeorbent assay for screening aflatoxin tn products and mixed feed: Col- laborativestudy. J.Aasn.OfIi<-_AnaLChem. 326-332. Park. D.L.. Miller. B.M.. Nesheim. 3.. Truck- sess.M.W..Veki A.. Bidigare.B.. McVey .11.. and Brown..1989b. Visual and semiquantitetive spectrophotometric W method for figautoxin B .234 cor}: peanut products: ow-up y Assn. Ofic. Anal. Chem.72 72638-643. Petey. A.L.. Sharman. M.. and Gilbert. J. :39?“ Wm: M muff: ‘"’ tn peanut enzyme- im- munosorbent assay: Collaborative study. J. AOAC IntL 75: 693-697. Pestka. J.J. 1991. High ormance thin layer chromatography GRAM: Ap- plication of s multi-hapten tmmunoassay toanalysisof thesearalenoneand aflatoxin mycotoxin familiesJ. Immunol. Methodol. 136: 177-183. Pestka.J.J. andCaaale.W. L 1990. Naturally occurring f al toxins. In “Food Contam- ination from vironmental Sources.”ed. M.S. Simmons andJ. Nriagu. pp. 613-638 John Wiley and Sons Ltd, New York. Pestka. J.J. and Chu. F.S. 1984.Enzyme- linked immunosorbent ease of to issuing nylo'n beadsand "91%;; soltdp Pestka. J.J.. Gaur. RR. and Chu. FS. 1980. Quantitation of aflatoxin B1 and aflatoxin B1 antibody by an linked tmmu- nosorbent microsssa . ppl. Environ. Mi- crobiol. 40: 1027-1031. Pestka.J.J.. Li.Y.. Harder. W.0.. and Chu. FS.1981a. Comparison of radioimmunoas- say and enzyme-linked immunosorbent as- say for determ aflatoxin M; tn milk. J. AmOficAnal. hem.64:294—-301. . Pestka.J.J.. Lee. 8.34141“. H-PalMIChu. RS. 1981a. Enzyme-linked immunosorbent assay for T-2 toxin. J. Am. Oil Chem. Soc. 58: 940A-944A. Pestka.J.J.. Li.Y. K..Harder.W. 0.. andChu. F.S. 1981b. Comparison of radioimmunoas- say and enzyme-linked immunosorbent as- say for determin aflatoxin M1 tn milk. J. Assn. OfieAnal. em. 64: 294-301. Ram. B...P Hart.L.P.. Cole.RJ.. andPestks. J.J. 1986a. A research note: Application of ELISAtoretailsurveyofaflatoxinBrin gut butter. J. Food Protect. 49: 792- Ram. B.P.. Hart. LP. Shotwell. OJ... and Pestka. J.J. 1986b. Analysis of aflatoxtn B1 in naturally contaminated corn and cotp e-linked immunosorbent assay: Comparison with thin layer chroma- tographyandhighperformancel uidchro- matography. J. AssrLOfieAnal. 69: Ramakrishna.N.. Lacey. J ..Candlish. A.A.G.. Smith. J. E..and Goodhrsnd. LA. 1990. Monoclonal antibodyobased enzyme linked immunosorbent assay of aflatoxin B;. T-2 toxin. and ochratoxin A to barley. J. Assn. Ollie. Anal. Chem. 73: 71-76. Sato. S.. Nagayams. S.. szamura. 0.. and Ueno.Y. 1987. Detection ofocl-ratoxinAin meat. wheat. and plasma by an enzyme- linked immunosorbent assay. Proc. Japan. . Assn. Mycotoxicol. 26: 47-50. Shelby. RA. and Kelley. V .C. 1992. Detection of ergot alkaloids from Cleric-cps spp.. in aaglricultural products by oompetttive ISA a monoclonal antibody. J. Fwood em. 40: 1090-1092. W..Stack. M.E..N WM. Park. DL..sndPohland.A.E.1989 cynic-linked immunosorbent assay of alla- toxinng. BalfldG. incormcottoneeed. peanuts. peanut butter. and poultry feed: Collaborative study. J. Assn. 05c. Anal. Chem. 72 957-962. Trucksess. M.W.. Stack. M.E.. Nesheim. 8.. Page. S.W.. and Albert. RH. 1991. lmmu- noafinity column can led with solution fluorometry or liq chromatography postcolumn derivatiution for determtns- tion of aflatoxins in corn. peanuts. and peanut butter: Collaborative study. J. Ann. 06c. Anal. Chem. 74: 81-88. Warner. R and Pestka.J.J.1986. Screening for zearalenone tn corn hycom tive di- rect enzyme-linked Miami.“ J. Agric. Food Chem. 34: 714-717. survey of retail grain- -haeed food products for zearalenone and aflatoxin B1. J. Food Protect. 50: 502—503. Wei. RD. andChu. F5. 1988. Productionand characterisation of antibody against PR toxin. J. Food Protect. 51: 463-466. Xu. Y.C_.. Zhang. 6.8.. and Chu. RS. 1986. ioimmunoassay of vslenol in wheatandcornJ. AmnOficAnaLChem. 69:967-969. Yu. J. and Chu. RS. 1991. lmmunochro- matographyof fttssrochromanonemyeo- anal AmOfieAnaLChekaGSS- Updated November 1994 from a paper gdn tedduring the IFT Toxicology and ety Evaluation Division posium “Im- munoassay Applications touchIII attheAnnual Meetingofthelnstitutsof {'33: Technologists. Chicago. nLJuly 10—14. —Edited Neil H. Met-melee ' Senior Associate ' an. .... LIST OF REFERENCES LIST OF REFERENCES Abbas, A.K., Lichtm'an, A.H., and Pober, J.S. 1994. Cellular and molecular immunology. W.B. Saunders Company, Philadelphia Abouzied, M.A., Azcona-Olivera, J.l., Braseleton, W.E., Pestka, J.J. 1993. Immunochemical assessment of mycotoxins in 1989 grain foods: Evidence for deoxynivalenol (vomitoxin) contamination. Appl. Environ. Microbiol. 57(3):672-677. Abouzied, M.A., Azcona-Olivera, J.l., Yoshizawa, T., Pestka, J.J. 1993. Production of polyclonal antibodies to the trichothecene mycotoxin 4,15- diacetylnivalenol with the carrier-adjuvant cholera toxin. Appl. Environ. Microbiol. 59(5): 1264-1268. Ackermann, T. 1991. Fast thin-layer chromatography systems for fumonisin isolation and identification. J. Appl. Toxicol. 11(6):451. Alberts, J.F., Gelderblom, W.C.A., Thiel, P.G., Marasas, W.F.O., Van Schalkwyk, D.J., and Behrend, Y. 1990. Effort of temperature and incubation period on production of fumonisin 81 by Fusarium monilifonne. Appl. Environ. Microbiol. 56: 1 729-1733. Avrameas, S and Temynck, T. 1969. The cross-linking of proteins with glutaraldehyde and its use for the preparation of immunoadsorbents. Immunochemistry 6:53-66. Azcona-Olivera, J.l., Abouzied, M.M., Plattner, R.D., and Pestka, J.J. 1992b. Production of monoclonal antibodies to the mycotoxins fumonisin-B1, fumonisin-82, and fumonisin-Ba; J. Agric. Food Chem. 40:531-534. Azcona-Olivera, J.l., Abouzied, M.M., Plattner, R.D., Norred, WP, and Pestka, J.J. 1992a. Generation of antibodies reactive with fumonisins B1, 32, and B3 by using cholera toxin as the carrier adjuvant. Appl. Environ. Microbiol. 58:169-173. Azer, M., and Cooper, C. 1991. Determination of aflatoxins in foods using HPLC and a commercial ELISA system. J. Food Protect. 54:291-294. 188 189 Bacon, CW, and Nelson, PE. 1994. Fumonisin production in corn by toxigenic strains of Fusarium moniliforme and Fusan‘um meiferatum. J. Food Protec. 57(6):514-521. Bamburg, J.R., 1983. Biological and biochemical actions of trichothecene mycotoxins, in “Progress in molecular and suboelllular biology: (ed. F.E. Hahn) vol. 8, pp. 41-110. Springer-Venag, Berlin. Bamburg, J.R., and Strong, FM. 19°71. 12, 13-epoxitrichothecenes, in Microbial toxins” (Eds. S. Kadis, A. Ciegler, and S.L. Aji) vol. Vll, pp. 207-292. Academic Press, New York. Bamburg, J.r., Strong, FM, and Smalley, EB, 1969. Toxins from moldy cereals J. Agric. Food Chem. 17:443-450. Barbacid, M., and Vazquez, D. 1974. Binding of [acetyl 14C] trichodennin to peptidyl transferase center of eukaryotic ribosome. Eur. J. Biochem 44: 437-444. Bars, J.L., Bars, P.L., Dupuy, J.. and Boudra, H. 1994. Biotic and abiotic factors in fumonisin B1 production and stability. J. AOAC lntemational, 77(2):517- 521. Battaner, E and Vazquez, D. 1971. Preparation of active 608 and 408 subunit from yeast ribosomes. Methods Enzymol. 20:446-449. Betina, V. 1989. Bioactive molecules volume 9, Mycotoxins: Chemical, biological and environmental aspects. Elsevier Science Publishing Company, Inc., New York. Better, M., Chang, C.P., Robinson, RR, and Horwitz, A.H., 1988. Escherichia coli secretion of an active chimeric antibody fragment, Science, 240:1041- 1043. Bezuidenhout, S.C., Gelderblom, W.C.A., Gorst-Allman, GP, 1988. Structure elucidation of the fumonisins, mycotoxins from Fusarium moniliforme, J. Chem. Soc., Chem. Commun.: 743-745. Bhat, R.V., Ramakrishna, Y., Beedu, SR, and Munshi, KL. 1989. Outbreak of trichothecene mycotoxicosis associated with consumption of mould- damaged wheat products in Kashmir Valley. The Lancet, January 7: 35-37. Blackwell, B.A., Miller, JD, and Savard, ME. 1994. Production of carbon 14- labeled fumonisin in liquid culture. J. AOAC lntemational, 77(2):506-511. Branham , BE, and Plattner, RD 1993. Isolation and characterization of a new fumonisin from liquid cultures of Fusarium moniliforme . J. Natural Products 56(9):1630-1633. 190 Candlish, A.A.G., 1991. The determination of mycotoxins in animal feeds by biological methods, In: Mycotoxyns and animal foods (Eds. J.E. Smith and RS. Henderson), pp. 224-246. CRS Press, Inc., Boston. Cannon, M., Jimenez, A, and Varquez, D. 1976. Competition between trichodermin and several other sesquiterpene antibiotics for binding to their receptor site(s) on eurkaryotic ribosomes Biochem. J. 160: 137-145. Carrasco, L., Barbacid, M., and Vazquez, D. 1973. The trichodennin group of antibiotics, inhibitor of peptide bond formation by eukaryotic ribosomes. Biochem. Biophys. Acta. 312: 368-376. 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(3):663-668. CAST, 1989. Mycotoxins: Economic and helath risks. Task Force Report No. 116. Council for Agricultural Science and Technology, Ames, Iowa. Cawood, M.’E., Gelderblom, W.C.A., Vleggaar, R., Behrend, Y., Thiel, P.G., and Marasas, W.F.O. 1991. Isolation of fumonisin mycotoxins: A quantitative approach. J. Agric. Food. Chem. 39:1958-1962. Cheng, S.J., Jiang,Y.Z., Li, M.H., and Lo, H1. 1985. A mutagenic metabolite produced by Fusarium moniliforme isolated from Linxia county, China. carcinogenesis, 62903-905 Chiba, J., Kawamura, 0., Kaji, H., Ohtani, K, Nagayama, S. and Ueno, Y. 1988. A sensitive enzyme-linked immunosorbent assay for detection of T-2 toxin with monoclonal antibody. Food Addit. Contam. 5:629-639. Chu, F.S., 1986. Resent studies on immunochemical analysis of mycotoxins. In: Mycotoxins and phycotoxins, 6th Intl. IUPAC Symp. on Mycotoxins and Phycotoxins, Pretoria, South Africa, July 22-25, 1985 (Ed. P. Steyn ans R. Vleggaar), pp. 277-292. Chu, SC. 1991. Development and use of immunoassays in detection of the ecologically important mycotoxins. In: Handbook of Applied Mycology, Vol. V . Mycotoxin (Eds. D. Bhatnagar, E.B. Lillihoj, and 0K. Arora), Marcel Dekker Inc., New York.. Chu, SC. 1992. Recent progress on analytical techniques for mycotoxins in feedstuffs. J. Anim. Sci. 70:3950-3963. Chu, F.S., Grossman, S., Wei, R.D., and Mirocha C.J., 1979. Plroduction of antibody against T-2 toxin. Appl. Environ. Mircrobiol. 37:104-108. In. . 2.12:1 191 Chu, F.S., Chen Liang, M.Y., and Zhang, 6.8., 19843. Production and characterization of antibody against diacetoxyscirpenol. Appl. Environ. Microbiol. 48:777-780. Chu, F.S., and Lee, RC, 1989. lmmunochromatography of group A trichothecene mycotoxins. Food Agric. lmmunol. 1:127-131. Chu, F.S., and Li, CY 1994. Simultaneous occurrence of fumonisin B1 and other mycotoxins in moldy corn collected from the People’s Republic of China in Region with high incidences of esophageal cancer. Appl. Environ. Microbiol. 60(3):847-852. Colvin, B.M., and Harrison, LR. 1992. Fumonisin-induced pulmonary edema and hydrothorax in swine. Mycopathologia 117:79-82. Colvin,B.M., Cooley, A.J., and Beaver, R.W. 1993. Fumonisins toxicosis in swine: Clinical and pathologic finding. J.Vet. Diagn. Invest 5:232-241. Committee on Protection Against Mycotoxins, 1983. Protection against trichothecene mycotoxins, National Academic Press, Washington, DC. Cote, L.M., Beasby, V.R., Bratich, P.M., Swanson, S.P., Shivaprasad, H.L., and Buck, W.B., 1985. Sex-related reduwd weight gains in growing swine fed diets containing deoxynivalenol. J. Animal Sci. 61:942-950. Cote, L.M., Reynolds, J.D., Vesonder, R.F., Buck, W.B., Swanson, S.P., Coffey, RT, and Brown, DC. 1984. Survey of vomitoxin-contaminated feed grains in Midwestern United State and associated health problems in swine. J. Amer. Vet. Assoc. 198(2):189-192. Cunliffe, E., and Davies, J. 1977. Inhibition of initiation, elongation, and termination of eukaryotic protein synthesis by trichothecene fungal toxins. Antimicrob. Agents Chemother. (11):491-499. Cunliffe, E., Cannon, M., and Davies, J. 1974. Mechanism of inhibition of eukaryotic protein synthesis by trichothecene fungal toxins. Proc. Natl. Acad. Sci.USA (71):30-34. Doko, M.B., Rapior, S., Visconti, A.. and Schjoth, E. 1995. Incidence and levels of fumonisin contamination in maize genotypes 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 (lg A) accumulation, elevated circulating lgA immune complexes, and hematuria during vomitoxin-induced lg A nephrophaty. F undam. Appl. Toxicol. 17:197-207. 192 Domer, J.W.,‘ Blankenship, PD, and Cole, R.J., 1993. Performance of two immunochemical assays in the analysis of peanuts for aflatoxin at 37 field laboratories. J. AOAC lntemational, 76:637-643. Doyle, T.W., and Bradner, W.T. 1980. Trichothecenes. In: Anticancer agents based on natural product models. Medicinal chemistry (Eds. J.M. Cassay, J.D. Douros) Vol. 16, pp. 43-72. Academic Press, New York . El-Nakib, O., Pestka, J.J. and Chu, PS. 1981. Determination of aflatoxin B1 in com, wheat and peanut butter by enzyme-linked immunosorbent assay and solid phase radioimmunoassay. J Assoc. Off. Anal. Chem. 64:1077-1082. Engvall, E., and Pennann, P., 1971. Enzim-linked immunosorbent assay (ELISA). Quantitative assay of protein immunoglobulin G; lmmunochemistry 8:871. Eppley, R.M., Truckesess, M.W., Neshiem, S., Thorpe, CW, and Pohland, AB. 1986. This layer chromatographic method for deerrnination of deoxynivalenol in wheat: Colaborative study. J. Assoc. Off. Anal. Chem. 69:37-40. Fan, T.S.L., Zhang, GS. and Chu, PS. 1984. An indirect enzyme-linked immunosorbent assay for T-2 toxin in biological fluids. J. Food Prot. 47:964- 968. Fernandez, C., Stack, ME, and Musser, SM. 1994. Determination of deoxynivalenol in 1991 US. winter and spring wheat by high-pannonnance thin-layer chromatograaphy. J. AOAC lntemational, 77(3):628-630. Femandez-Munoz, R., Monro, RE, and Vazquez 1971. Ribosomal peptidyltransferase: Binding of inhibitors. Methods Enzymol. 20:481-490. Forgacs, J.. and Car1l, W.T., 1962. Mycotoxicoses. Adv.Vet.Sci.7:273-282. Forsell, J.H., Jensen, R., Tai, J.H., Witt, M., Lin, W.S., and Pestka, J.J. 1987. Comparison of acute toxicities of deoxynivalenol (vomitoxin) and 15- acetyldeoxynivalenol in the B6CSF1 mouse. Fd. Chem. Toxic. 25:155-162. Forsell, J.H., Witt, M., Jensen, R., Tai, J.H., Jensen, R., and Pestka, J.J. 1986. Effects of 8-week exposure of the B603F 1 mouse to dietary deoxynivalenol (vomitoxin) and zearalenone. Food Chem. Toxicol. 24:213- 219. Freeman, G.G., and Morrison, RI 1948. Tnchothecin: An antifungal metabolic product of T. roseum Link. Nature, 162230-33. E_ . 193 Freeman, G.G., and Morrison, RI 1949. The isolation and chemical properties of trichothecin, an antifungal substance from Trichothecium roseum Link. Biochem. J. 4421-5. Fried, HM. and Wamer, JR. 1981. Cloning of yeast gene for trichodennin resistance and ribosomal protein L3. Proc. Natl Acad. USA 78: 238-242. Fukuda, S., Nagahara, A., Kikuchi, M., and Kumagai, S. 1994. Preparation and characterization of anti-fumonisin monoclonal antibodies. Biosci. Biotech. Biochem, 58(4):765-767. Furlong, EB, and Scares, L.M.V., 1995. Gas chromatograpic method for quantitation and confirmation of trichothecenes in wheat. J. AOAC lntemational, 78(2):386-390. Galfre, G. and Milstein, C. 1981. Preparation of monoclonal antibodies: strategies and procedures: Methods Enzymol., 73:3-46. Gelderblom, W.C.A., Jaskiewic, K., Marasas, W.F.O., 1988a. Fumonisins-novel mycotoxins with cancer-promoting activity produced by Fusarium moniliforme. App. Environ. Microbiol. 54:1806-181 1. Gelderblom, W.C.A., Marasas, W.F.O., Jaskiewicz, K., Combrinck, S. and Van Schalkwyk, D.J. 1988b. Cancer promoting petential of different strains of Fusarium monilifonne in short-term cancer initiation/promotion assay, Carcinogenesis 9(8): 1405-1409. Gelderblom, W.C.A., Kriek, N.P.J., Marasas, W.F.O., and Thiel, P.G., 1991. Toxicity and carcinogenicity of the Fusarium monilifonne metabolite, fumonisin 81 , in rats. Carcinogensis, 12:1247-1251. Gelderblom, W.C.A., Marasas, W.F.O., Vleggaar, R., Thiel, P.G., and Cawood, M.E.,1992a. Fumonisins-Isolation, chemical characterization and biological effects. Mycopathologia 1 17:1 1-16. Gelderblom, W.C.A., Semple, E., Marasas, W.F.O., and Farber, E. 1992b. The cancer-initiating potential of the fumonisin-B mycotoxins. Carcinogenesis 13:433-437. Gendloff, E.H., Pestka, J.J., Swanson, SP. and Hart, LP. 1984. Detection of T-2 toxin in Fusarium sporotn’chioides infected corn by enzyme-linked immunosorbent assay. Appl. Environ. Microbiol. 47:1161-1163. Gendloff, E.H., Casale, W.L., Ram, B.P., Tai, J.H., Pestka, J.J., and Hart, LP. 1986. Hapten-protein conjugates prepared by the mixed anhydride method. J. Immunol. Methods, 92:15-20. 9'1 K. .‘JX14...’ . 194 Gilbert, J. 1993. Recent advances in analytical methods for mycotoxins. Food Addit. Contam. 10(1):37-48. Goding, J.W. 1980. Antibody production by hybridoma. J. Immunol. Methods, 39:285-308. Grant, P.G., Schinder, D., and Davies, J.E. 1976. Mapping of trichodennin resistance in Saccharomyces cerevisae: A genetic locus for a component of the 608 ribosomal subunit Genetic 83: 667-673. Greene, D.M., Bondy, G.S., Azcona-Olivera, J.l., and Pestka, J.J. 1994. Role of gender and strain in vomitoxin-induced dysregulation of IgA production and IgA nephropathy in the mouse. J. Toxic. Environ. Health, 43(1):37-50. Hack, R., Klaffer, U. and Terplan, G. 1989. A monoclonal antibody to the trichothecene mycotoxin diacetoxyscirpenol. Lett. Appl. Microbiol. 8:71-76. Hagler, W.M., Tyczkowska, K., and Hamilton, PB, 1984. Simultaneous occurrence of deoxynivalenol, zearalenone, and aflatoxin in 1982 scabby wheat from the midwestem United States. Appl. Environ. Microbiol, 47:151-154. Harlow, E. and Lane, D. 1988. Antibodies: A laboratory manual. Cold Spring Harbor Laboratory, New York. Harrison, L.R., Colvin, B.M., Greene, J.T., 1990. Pulmonary edema and hydrothorax in swine produced by fumonisin B1, a toxic metabolite of Fusarium monilifonne. J. Vet. Diagn. Invest. 22217-221. Hascheck,W.M., Motelin, G., Ness,D.K., Harlin, KS, Hall, W.F., Vesonder, R.F., Peterson, RE, and Beasley, V.R. 1992. Characterization of fumonisin toxicity in orally and intravenously dosed swine. Mycopathologia 117:83- 96. Hebert, G.A., Pehlman, PL, and Pittman, B. 1973. Determination of the optimal ammonium sulfate concentration for the fractionation of rabbit, sheep, horse and goat antisera. Appl. Microb. 37:26-36. Hersckovits, T. 1988. Recent aspect of the subunit organization and dissociation of hemocyanins. Comp. Biochem. Physiol. 91B2597-611. Hesseltine, CW. 1986. Global significance of mycotoxins, in: “Mycotoxin and phycotoxins', 6th Intl. IUPAC Symp. on Mycotoxins and Phycotoxins, Pretoria, South Africa, July 22-25, 1985, ed. P.S. Steyn and R. Vleggaar, pp. 1-18. Elsevier Science Publishing Company Inc., Amsterdam, The Netherlands. 195 Hietaniemi, V., and Kumpulainen, J., 1991. Contents of Fusarium toxins in Finnish and imported grains and feeds. Food addit. contam., 8:171-182. Holmgren, J. 1981. Actions of cholera toxin and the prevention and treatment of cholera. Nature, 292:413-417. Hsu, l.C., smalley, E.B., Strong, FM, and Ribelin, W.E. 1972. Identification of T- 2 toxin in moldy corn associated with a lethal toxicoses in dairy cattle. Appl. Microbiol. 24:684-690. Jackson, MA. and Bennet, GA. 1990. Production of fumonisin B1 by Fusan'um monilifonne NRRL 13616 in submered culture. Appl. Environ. Microbiol. 56(8):2296-2298. Jaskiewicz, K., Van Rensburg, S.J., Marasas, W.F.O., and Genderblom, WC 1987. Carcinogenicity of Fusarium monilifon'ne culture material in rats. J.Natl. Cancer Inst. 78(2):321-325. Jimenez, A., and Vazquez, D., 1975. Quantitative binding of antibiotics to ribosomes from a yeast mutant altered on the peptidyl-transferase center. Eur. J. Biochem. 54:483-492. Joffe, AZ. 1962. Biological properties of some toxic fungi isolated from ovemintered cereals. Mycopathol. Mycol.Ale. 16:201-221. Joffe, AZ. 1965. Toxin production by cereal fungi causing toxic alimentary aleukia in man, In: Mycotoxins in foodstuffs (ed.G.N. Wagon), pp.77-85. MIT Press, Cambridge, MA. Kamimura, H., Nishijima, M., Yasuda, K., Saito, lbe, A., Nagayama, T., Ushiyama, H. and Naoi, Y. 1981. Simultaneous detection of several Fusarium mycotoxin in cereals, grains, and foodstuffs. J. Assoc. Off. Anal. Chem. 64(5): 1067-1073. Kellerrnan, T.S., Marasas, W.F.O., Thiel, PG. 1990. Leukoencephalomalacia in two horses induced by oral dosing of fumonisin B1. Onderstepoort J. Vet. Res. 57:269-275. Kemp, H.A., Mills, E.W.C., and Morgan, M.R.A.; 1986; Enzyme-linked immunosorbent assay of 3-acetyldeoxynivalenol applied to rice; J.Sci. Food Agric., 37:888-894. Kim, J.C., Kang, H.J., Lee. D.H., Lee, Y.W., and Yoshizawa, T. 1993. Natural occurrence of Fusarium mycotoxins (trichothecenes and zearalenone) in barley and corn in Korea. Appl. Environ. Microbiol. 59(11):3798-3802. Kitagawa, T., Shimozono, T., Aikawa, T, Yoshida, T., and Nishimura, H. 1981. Preparatiion and characterization of hetero bifunctional crosslinking reagents for protein modifications. Chem. Phann. Bull., 29:1130—1135. 196 Koch, C., Skjodt, K., and Laursen, I. 1985. A simple immunoblotting method after separation of proteins in agarose gel. J. Immunol. Methods. 84:271-278. Kohler, G., and Milstein, C.. 1975. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495-497. Laamanen, l., and Veijalainen, P. 1992. Factor affecting the results of T-2 mycotoxin ELISA asay. Food Addit. Contam., 9(4):337-343. Lacey, J. 1986. Factors affecting mycotoxin production. In: Mycotoxin and phyxotoxins. (eds. P.S. Steyn and R. Vleggaar), pp.65-76. Elsevier Scientific Publishing 00., Amsterdam, The Netherlands. Lauren, DR. and Greenhalg, R. 1987. Simultaneous analysis of nivalenol and deoxinevalenol in cereals by liquid chromatography. J. Assoc. Off. Anal. Chem. 70:479-483. Lauren, DR, and Agnew, MP. 1991. Multitoxin screening method for Fusarium mycotoxins in grains. J.Agric. Food Chem. 39:502-507. Lee, S. and Chu, F.S. 1981a. Radioimmunoassay of T-2 toxin in corn and wheat J. Assoc. Off. Anal. Chem. 64:156-161. Lee, S. and Chu, F .S. 1981b. Radioimmunoassay of T-2 toxin in biological fluids. J. Assoc. Off. Anal. Chem. 64:684—688. Lee, U.S., Jang, H.S., Tanaka, T., Hasegawa, A.. Oh, Y.J., and Ueno, Y. 1985. The coexistence of the Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone in Korean cereals harvested in 1983. Food Addit. Contam. 2: 185-192. Lee, U.S., Jang, H.S., Tanaka, T., Hasegawa, A.. Oh, Y.J., Cho, C.M., Sugiura, Y., and Ueno, Y. 1986. Further survey of the Fusarium mcotoxin in Korea cereals. Food Addit. Contam. 3:253-261. Liang, X,. Lamm, ME, and Nedrud, JG, 1978. Cholera toxin as a mucosal adjuvant. Glutaraldehyde treatment dissociates adjuvanticity from toxicity. J. Immunol. 143:484-490. Lycke, N. and Holmgren, J. 1986. Strong adjuvant properties of cholera toxin on gut mucosal immmune response to orally presented antigens. lmmunoloy 59:301-308. Marasas, W.F.O. 1982. Mycotoxicological investigations on corn produced in esophageal cancer areas in Transkei. In Cancer of the Esophagus; Pfeiffer, C.J. Ed. CRC Press; Bota Raton, FL.;pp:29-40. 197 Marasas, W.F.O., 1986. Fusarium monilifonne : A mycotocicological miasma. In : “Mycotoxin and phycotoxins”, 6th Intl. IUPAC Symp. on Mycotoxins and Phycotoxins, Pretoria, South Africa, July 22-25, 1985, ed. F.S. Steyn and R. Vleggaar, pp. 19-28. Elsevier Science Publishing Company Inc, Amsterdam, The Netherlands. Marasas, W.F.O., ., Jaskiewicz, K., Venter, F.S., and Van Schalkwyk, D.J. 1988a. Fusarium moniliforme contamination of maize in esophageal cancer areas in Transkei. SAMJ Vol. 74, 6 August, 110-114. Marasas, W.F.O., Kellennan, T.S., Gelderblom, W.C.A.,Coetzer, J.A.W., Thiel, PF. and Van Der Lugt, J.J. 1988b. Leukoencephalomalacia in a horse induced by fumonisin B1 isolated from Fusarium monilifonne. Onderstepoort J. Vet. Res. 55:197-203. Marasas, W.F.O., Nelson, PE, and Toussoun, T.A. 1984a. Toxigenic Fusarium species: Identity and Mycotoxicology; Pennsylvania State University Press; University Park, PA. Marasas, W.F.O., Kriek, N.P.J., Fincham, J.E., and Van Rensburg, S.J., 1984b. Primary liver cancer and esophageal basal cell hyperplasia in rats caused by Fusarium moniliforme . Int. J. Cancer, 34:383-387. Martlbauer, E., Gareis, M., and Terpan, G. 1988. Enzyme immunoassay for the macrocyclic trichothecene roridin A: production, properties, and use of rabbit antibodies. Appl. Environ. Microbiol. 54:225-230. McLaughlin, C.S. Vaughan, M.H., Cambel, I.M., Wei, C.M., Stafford, ME. and Hansen, BS. 1977. Inhibitor of protein synthesis by trichothecene. In: Mycotoxin in Human and Animal Health (Eds. Roderick, J.V., Hesseltine, CV, and Mehllman, MA), pp. 262-273, Pathotox Publishers, Park Forest South, Illinois. Middlebrook, J.L. and Leathennan, D.L., 1989. Binding of T-2 toxin to eukaryotic cell - ribosomes. Biochemical Pharmacology, 38(18):3103-3110. Mills, J.T., 1982. Development of fusaria and fusariotoxins on cereal grains in storage. Can. J. Plant Pathol. 4:217-218. Milstein, C. 1990. The croonian lecture, 1989. Antiboy: a paradigm for the biology of molecular recognition. Proc R. Soc. Lond. Biol. 239:1-16. Minervini, F., Bottalico, C.. Pestka, J., and Visconti, A. 1992. On the occurance of fumonisins in feeds in Italy; Atti Della Societa Italiana Delle Scienze Veterinarie (volume XLVI):1365-1 368. ir 33mgi‘x. - _ o.~ v-‘- "'ifl 198 Mirocha, C.J., Pathre, S.V., Schauerhamer, B., and Christensen, CM, 1976. Natural occurrence of Fusarium toxins in feedstuff. Appl. Environ. Microbiol. 32(4):553-556. Mirocha, C.J., Pawlosky, R.J., and Abbas, H.K. 1989. Analysis of T-2 toxin in a biological matrix using multiple reacting monitoring. Arc. Environ. Contam. Toxicol. 18:349-355. Mooroka, N., Uratsuji, N., Yoshizawa, T., and Yamamota, H., 1972. Studies on the toxic substances in barley infected with Fusarium spp. J. Food Hyg. Soc. Jpn. 13:368-375. Morrissey, RE. and Vesonder, RF, 1985. Effect of deoxynivalenol (vomitoxin) on fertility, pregnancy, and postnatal development of Sprague-Dawley rats. Appl. Env. Microb. 49:1062-1066.. Muller, HM. and Schwardorf, K 1993. Natural occurrence of Fusarium toxin in barley grown in a Southwestern area of Germany. Bull. Environ. Contam. Toxicol. 51 :532-537. Nakane, PK. and Kawaoi, A., 1974. Peroxidase-labeled antibody: a new methods of conjugation. J. Histochem. Cytochem. Vol.22(12):1084-1091. Nelis, H.J., and Sinsheimer. 1981. A sensitive fiuorometric procedure for the determination of aliphatic epoxides under physiological condition. Anal. Biochem. 115:151-157. Nelson, P.E., Juba, J.H., Ross, RF, and Rice, LG. 1994. Fumonisin production by Fusarium species on solid substrates. J. AOAC lntemational. 77(2):522-524. Nelson, P.E., Platter, R.D., Shackelford, DD, and Deohardins, A.E.; 1991. Production of fumonisins by Fusarium monilifonne strains form various substrates and geographic areas. Appl. Environ. Microbiol. 57:2410-2412. Nilsson, B.O., Svalander, PC, and Larson, A. 1987. Immunization of mice and rabbits by intrasplenic deposition of nanogram quantities of protein attached to Sepharose beads or nitrocellulose paper strips. J. Immunol. Methods, 99:67-75. Norred, WP. 1993. Fumonisins-mycotoxin produced by Fusarium moniliforme, J. Toxicol. and environ. Health, 38:309-328. Norred, WP. and Voss, K.A. 1994. Toxicity and role of fumonisins in animal diseases and human esophageal cancer. J. Food Protec. 57(6):522-527. :7." on: imam-a: ‘T-‘_‘ . n-_' 199 Oi, V.T., MoniSon, S.L., Herzenberg, LA, and Berg, P. 1983. Immunoglobulin gene expression in transformed lymphoid cells. Proc. Natl. Acad. Sci. USA, 80:825- 829. Orlandi, R., Gussow, D.H., Jones. PT, and Winter, G. 1989. Cloning immnoglobulin variable domains for expression by the polymerase chain reaction. Proc. Natl. Acad. Sci. USA, 86:3833-3837. Osweiler, G.D., Ross. P.F., VViIson, T.M., Nelson, P.E., Witte, S.T. Carson.T.L., Rice, LG, and Nelson. HA. 1992. Characterization of an epizootic of pulmonary edema in swine associated with fumonisin in corn screenings. J.Vet.Diagn. Invest. 4:53-59. Park, D.L., Miller, B.M., Hart, P., Yang, G., McVey, J.L., Page, S.W., Pestka, J., and Brown, L.H. 1989a. Enzyme-linked immunosorbent assay for screening aflatoxin in cottonseed products and mixed feed: Colaborative study. J. Assn. Off. Anal. Chem. 72:326-332. Park, D.L., Miller, B.M., Neshheim, S., Trucksess, M.W., Vekich, A, Bidigare, B., McVey, J.L., and Brown, L.H. 1989b. Visual and semiquantitative spectrophotometric screening method for aflatoxin B in corn and peanut products: Follow-up study. J. Assn. Off. Anal. Chem. 72:638-642. Park, K.J., Park, AR, and Lee, Y.W. 1992. Natural occurrence of Fusarium mycotoxins of the 1990 barley crop in Korea. Food Addit. Contam. 9(6):639-645. Pestka, J.J., 1988. Enhanced surveillance of foodbome mycotoxins by immunochemical assay. J. Assoc. Off. Anal. Chem.; 71(6):1075—1081. Pestka, J.J. 1991. High performance thin layer chromatography ELISAGRAM: Application of a multihapten immunoassy to analysis of the zearalenone and aflatoxin mycotoxin familics. J. Immunol. Methods. 136:177-183. Pestka, J.J. 1994. Application of immunology to the analysis and toxicity assessment of mycotoxins. Food and Agric. immunol. 6:219-234. Pestka, J.J. and Bondy, GS. 1990. Alteration of immune fuction following dietay mycotoxin exposure. Can. J. Physiol. Pharmacol, 68:1009-1016. Pestka, J.J., and Casale, W., 1990. Natural occuring fungal toxins. In Food contamination from environmental sources (eds. M.S. Simmons and J Nriagu), John Willey and Sons, Ltd., New York. pp. 613-638. Pestka, J.J. and Dong, W., 1993. Persistent dysregulation of IgA production of lgA nephropathy in the BGC3F1 mouse following withdrawal of dietary vomitoxin (deoxynivalenol). Fundam. and Appl. Toxicol., 20 (1):38-47. 200 Pestka, J.J. and Dong, W., 1994. Progressive serum IgE elevation in the B6C3F1 mouse following withdrawal of dietary vomitoxin (deoxynivalenol). F undam. and Appl. Toxicol., 22:314-316. Pestka, J.J., Lee, S.S., Lau, HP, and Chu, F.S.; 1981a; Enzyme-linked immunosorbent assay for T-2 toxin; J. Am. Oil Chem. Soc, 58:940A-944A. Pestka, J.J., Li, Y., Harder, W.O., and Chu, F.S., 1981b. Comparison of radioimmunoassay and enzye-linked immunosorbent assay for determining aflatoxin M in milk. J. Assn. Offs. Anal. Chem. 64:294-301. Pestka, J.J., Li, Y.K, and Chu, F.S., 1982. ReactMty of aflatoxin 82. antibody with aflatoxin B1 modified DNA and related metabolites. Appl. Environ. Microbiol. 44:1159—1165. Pestka, J.J., Tai, J.H., Witt, M.F., Dixon, DE, and Forsell, J.H. 1987. Suppression of immune response in the B603F1 mouse after dietray exposure to the Fusarium mycotoxins deoxynivalenol (vomitoxin) and zearalenone. Fd. Chem. Toxic. 25:297-304. Pestka, J.J., Moorrnan, MA, and Warner, R.L. 1989. Dysregulation of lgA production and lgA nephropathy induced by the trichothecene vomitoxin. Food Chem. Toxicol., 27:361-368.Pestka, J.J. Dong, W., Warner, R.L., Rasooly, L., and Bondy, GS. 1990. Effect of dietary administration of the trichothecene comitoxin (deoxynivalenol) on IgA and lgG secretion by Peyr‘s Patch and splenic lymphocytes. Food. Chem. Toxicol.28:693-699. Pestka, J.J., Azcona-Olivera, J.l., Plattner, R.D., Minervini, F., Doko, MB, and \fisconti, A. 1994. Comparative assessment of fumonisin in grain-based foods by ELISA, GC-MS, and HPLC. J. Food Protection 57(2):169-172. Pestka, J.J., Abuzied, MM, and Sutikno. 1995. lmmunologically-based assays for mycotoxin detection. Food Tech. 21120-128. Peter, AT. 1904. A fungus disease in corn. Agric. Exp. Stn. Nebraska, 17th Ann. Rep. pp. 13-22. Pittet, A., Parisod, V., and Schellenberg, 1992. Occurrence of fumonisin B1 , and B2, in com-based products from the Swiss market. J. Agric. Food Chem. 40(8): 1 352-1354. Plattner, RD. 1986. Mass spectrometry-mass spectrometry as a tool for mycotoxin analysis. In: Cole, R.J. (Ed.) Modern Method in the Analysis and Structural Elucidation of Mycotoxins, pp. 393-414. Academic Press. New York. 201 Plattner, R.D., Norred, W.P., Bacon, C.W., Voss, K.A., Peterson, R., Shackelford, DD, and Weisleder, D. 1990. A method of detection of fumonisins in corn samples associated with field cases of equine leukoencephalomalacia. Mycologia 82:698-702. Plattner, R.D., and Schackelford, DD, 1992. Biosynthesis of labeled fumonisins in liquids cultures of Fusarium moniliforme . Mycopathologia, 117:17-22. Plattner, R.D., Weisleder, D., Shackelford, D.D., Peterson, R., and Powell, R.G. 1992. A new fumonisin from solid cultures of Fusarium moniliforme. Mycopathologia 1 17:23-26. Pohland, A.E. 1993. Mycotoxin in review. Food Addit. Contam. 10(1):17-28. Potter, M. 1972. Immunoglobulin-producing tumors and myeloma proteins of mice. Physiol. Rev. 52:631-719. Price, W.D., Lovell, RA, and McChesney, 0.6., 1993. Natural occurring toxins in feedstuffs: Center for Veterinary Medicine Perspective. J. Anim. Sci. 71 :2556-2562. Ram, B.P., Hart, L.P., Shotwell, O.L., and Pestka, J.J. 1986. A research note: Application of ELISA to retail survey of aflatoxin B1 in peanut butter. J. Food Protect. 49:792-795.. Ramakrishna, Y., Bhat, RV, and Vasanthi, S., 1990. Natural occurance of mycotoxins in staple foods in India. J. Agric. Food Chem. 38218574859. Rheeder, J.P., Marasas, W.F.O., Thiel, P.G., Sydenham, E.W., Shephard, GS, and Van Schalkwyk, DJ. 1992. Fusarium moniliforme and fumonisins in corn in relation to human esophageal cancer in Transkei. Phytopathology, 82:129-135. Rheeder, J.P., Sydenham, E.W., Marasas, W.F.O., Thiel, P.G., Shephard, G.S., Schlechter, M., Stockenstrom, S., Cronje, D.E., nad Vrljoen, J.H. 1994. Ear-rot fungi and mycotoxins in South African corn of the 1989 crop exported to Taiwan. Mycopathologia 127:35-41. Riley, R.T., An, N.H., showker, J.L., Yoo, H.S., Norred, W.P., Chamberlain, W.J., Wang, E., Merrill Jr, A.H., Montelin, G., Beasley, V.R., and Haschek, WM. 1993. Alteration of tissue and serum sphinganine to sphingosine ratio: An early biomarker of exposure to fumonisin-containing feeds in pigs. Toxicology and Appl. Marmacol. 118:105-112. Robbana-Bamat, S., Lafarge-Frayssinet, C., Cohen, H., Neish, GA, and Frayssinet, C.. 1988. Immunosuppressive properties of deoxynivalenol . Toxicology, 48:155-166. 202 Robbana-Bamat, S., London-Rosa, B., Cohen, H., Lafarge-Frayssinet, C., Neish, GA, and Frayssinet, C., 1987. Protein synthesis inhibition and cardiac lesions associated with deoxynivalenol ingestion in mice. Food Addit. and Contam. 4:49-55. Romer, TR. 1986. Use of small charcoal/alumina cleanup columns in determination of trichothecene mycotoxins in foods and feeds. J. Assoc. Off. Anal. Chem. 69(4):699-703. Ross, PF, 1994. What are we going to do with this dead horse?, J. AOAC lntemational. '77(2):491-494. Ross, P.F., Nelson, P.E., Richard, J.L. 1990. Production of fumonisin by Fusarium moniliforme and Fusarium proliferatum isolates associated with equine leukoencephalomalacia and pulmonary edema syndrome in swine. Appl. Environ. Microbiol. 56:3225-3226. Ross, P.F., Rice, L.G., Osweiler, G.D., Nelson, P.E., Richard, J.L., and V\filson, TM, 1992. A review and update of animal toxicoses associated with fumonisin-contaminated feeds and production of fumonisins by Fusarium isolates. Mycopatholodia 1 17: 1 09-1 14. Ross, P.F., Rice, L.G., Osweiler, G.D., Wilson, T.M., Owens, D.L., Nelson, HA, and Richard, J.L. 1991. Concentrations of fumonisin B1 in feeds associated with animal health problems. Mycopathologia 114:129—135. Rottinghaus, G.E., Coatney, C.E., Minor, HQ 1992. A rapid, sensitive thin layer chromatography procedure for the detection fo fumonisn B1 and B2. J. Vet. Diagn. Invest. 4:326-329. Sano, A., Asabe, Y., Takitani, S., Ueno, Y., 1982. Fluorodensitometric determination of trichothecene mycotoxins with nicotinamide and 2- acetylpyridine on a silica gel layer. J. Chromatogr. 235:257-265. Schiefer, H3. 1988. The differential diagnosis: Are there natural explanations for what is called ‘yellow-rain” and its alleged effects? Comment Toxicol. 2:51- 62. Schindler, D., Grant, P., and Davies, J. 1974. Trichoderrnin resistance-mutation affecting eukaryotic ribosomes. Nature 248: 535-536. Schroeder, H.W., and Hein, H, Jr. 1975. A note on zearalenone in grain sorghum. Cereal Chem. 52:751-752. Schultz, L.D., and Friesen, JD, 1983. Nucleotide sequence of the tcm1 gene (ribosomal protein L3) of Saccaromyces cerevisiae. J. Bactriology, 155(1):8-14. 203 Scott, P.M., Lawrence, J.W., and Walbeek, W.V., 1970. Detection of mycotoxins by thin-layer chromatography: Application to screening of fungal extracts. Appl. Microbiol. 20(5):839-842. Scoot, P.M., Harwig, J., and Blanchfield. 1980. Screening Fusarium strains isolated from ovenrvintered Canadian grains for trichothecenes. Mycopathologia, 72: 175-180. Scott, P.M., Lau, P.Y., and Kanhere, SR, 1981. Gas chromatography with electron capture and mass spectrometric detection of deoxynivalenol in wheat and other grains. J. Assoc. Off. Anal. Chem. 64:1364-1370. Scott, P.M., Nelson, K., Kanhers, S.R., Karpinski, S.F., Hayward, 8., Neish, GA, and Teich, AH. 1984. Decline in deoxynivalenol (vomitoxin) concentrations in 1983 Ontario winter wheat before harvest. App. Environt. Microbiol. 48:884-886. Scott, PM. and Lawrence , GA. 1992. Liquid chromatographic determination of fumonisin B1 and B2 in corn and corn products. J. AOAC lntemational 75(5):829-834. Seagrave, S., 1981. Yellow rain. New York: Evans. 1981. Seaman, W.L. 1982. Epidemiology and control of mycotoxigenic fusaria on cereal grains. Can. J. Plant Pathol. 4:187-190. Senozan, N.M., Landmm, J., Bonaventura, J.. and Bonaventura, C., 1981. Hemocyanin of the giant keyhole limpet, Megathura crenulata. ln Invertebrate oxygen binding proteins: structure, active sites and function. (eds. J. Lammy and J. Lammy), Marcel Dekker, New York. pp. 703-717 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, W.C.A. 1990. Quantitative determination of fumonisins B1 and B2 by high performance liquid chromatography with fluorescence detection. J. Liq. Chromatogr. 1 322077-2087. Shepherd, M.J. and Gilbert, J. 1986. Method for the analysis in maize of the Fusarium mycotoxin monilifonnin empoying ion-paimg extraction and high performance liquid chromatography. J. Chomatogr. 358:415-422. Shotwell, O.L., Goulden, M.L., Bennett, G.A., Plattner, R.D., and Hesseltine, CW. 1977. Survey of 1975 wheat and soybeans for aflatoxin, zearalenone and ochratoxin. J. Assoc. Off. Ana. Chem, 60:718-783. 204 Shotwell, O.L., and Hesseltine, CW. 1983. Five year study of mycotoxins in Virginia wheat and dent corn. J. Assoc. Off. Ana. Chem., 66:1466-1469. Shotwell, O.L., Bennett, G.A., Stubblefield, R.D., Shannon, G.M., Kwolek, W.F., and Plattner, RD. 1985. Deoxynivalenol in hard red winter wheat: relationship between toxin levels and factors that could be used in grading. J. Assoc. Off. Anal. Chem. 68:954-957. Siler, DJ, and Gilchrist, DC. 1982. Determination of host-selective phytotoxins from Alteman‘a altemata f. sp. chopersici as their maleyl derivatives by high-performance liquid chromatography. J. chromatogr. 238:167-173. Smith, J.E., and Moss, MO. 1985. Mycotoxins :Fonnation, analysis, and significance. John Willy & Sons, New York. Smith, K.E., Cannon, M., Cundliff, E., 1975. Inhibition at initiation level of eukaryotic protein synthesis by T-2 toxin. F EBS Lett. 5028-12. Snyder, AP. 1986. Qualitative, quantitative and technological aspects of the trichOthecene mycotoxins. J. Food. Protect. 49(7):544-569. Spedding, G.G., 1990. Ribosomes and protein synthesis: A practical approach, IRL Press at Oxford University Press, Oxford, England. p. 5. Spensley, PC. 1963. Aflatoxin, the active principle in turkey 'x’ disease. Endeavor, 22:75-79. Stack, ME, and Eppley, RM. 1992. Liquid chromatograpic determintion of fumonisins B1 and B2 in corn and corn products. J. AOAC lntemational 75(5):834-837. Steel, R.G.D., and Torrie, J.H. 1980. Principles and procedures of statistics: A biometrical approach. Second edition, pp. 272-284. McGraw-Hill Book Company, New York. Steven, RB. 1974. Mycology guidebook. Mycology guidebook committee, Mycological Society of America. University of Washington Press, Seattle. Sugasawara, R.J., Cahoon, BE, and Karu, A.E. 1985. The influence of murine macrophage-conditioned medium on cloning efficiency, antibody synthesis and growth rate of hybridomas. J.lmmunol. Methods. 79:263-275. Sydenham, E.W., Thiel, P.G., Marasas, W.F.O., and Nieuwenhuis, J.J. 1989. Occurrence of deoxynivalenol and nivalenol in Fusarium graminearum infected undergrade wheat in South Africa. J. Agric. Food Chem. 37:922- 926. 205 Sydenham, E.W., Thiel, P.G., Marasas, W.F.O., Shephard, GS, and Van Schalkwyk, DJ. 1990. Natural occurrence of some Fusarium mycotoxins in corn from low and high asaphageal cancer prevalence areas of the Transkei, Southern Africa. J.Agir. Food Chem. 38(10):1900-1903. Sydenham, E.W., Gordon, S., Shephard, G.S., Thiel, P.G., Marasas, W.F.O., and Stockenstrom, S., 1991. Fumonisin contamination of commercial com- based human foodstuffs. J. Agric. Food Chem. 39:2014-2018. Sydenham, E.W., Gordon, S., Shephard, G.S., Pieter G., Thiel, P.G., Marasas, ' W.F.O., Rheeder, J.P., Sanhueza, C.E.P., Gonzalez, H.H.L., and Resnik, S.L., 1993. Fumonisins in Argentinian field-trial corn. J. Agric. Food Chem. 41:891-895. Takitani, S. Asabe, Y., Kato, T., Suzuki, M., Ueno, Y. 1979. “Spectrodensitometric determination of trichothecene mycotoxins with 4-(p- nitrobenzyl)pyridine on silica gel thin layer chromatography". Chromatographia, 172:335-342. Tanaka, T., Hasegawa, A., Matsuki, Y., lshii, K, and Ueno, Y. 1985. Improved methodology for the simultaneous detection of the Fusarium mycotoxins deoxynivalenol and nivalenol in cereals. Food. Addit. Contam. 22125-137. Tanaka, T., Hasegawa, A., Matsuki, Y., Lee, U.S., and Ueno, Y. 1986. A limited survey of Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone in 1984 UK-harvested wheat and barley. Food Addit. Contam. 32247-252. Tanaka, T., Hasegawa, A.. Yamamoto, S.,Sugiura, Y., and Ueno, Y. 1988a. A case report on a minor contamination of nivalenol in cereals harvested in canada. Mycopathologia, 101 2157-160. Tanaka, T., Hasegawa, A., Yamamoto, S., Lee, U.S., Sugiura, Y., and Ueno, Y. 1988b. Worldwide contamination of cereals by the Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone. 1. Survey of 19 countries. J. Agric. Food Chem. 26:979-983. Tanaka, T., Yamamoto, S., Hasegawa, A., Aoki, N., Besling, J.R., Sugiura, Y., and Ueno, Y. 1990. A survey of the natural occurrence of Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone, in cereal harvested in the Netherlands. Mycopathologia, 110219-22. Tarcha, P.J., 1991. The chemical properties of solid-phases and their interaction with proteins, In: lmmunochemistry of solid-phase immunoassay (Ed. J.E. Butler), CRC Press, Boca Rtaton, Florida. 206 Tejada-Simon, M.V.; 1994. Comparative detection of fumonisin by HPLC, ELISA, and immunocytochemical localization in Fusarium Cultures; and polyclonal antibody production for ergosterol; Master Thesis, Department of Food Science and Human Nutrition, Michigan State University. Thiel, P.G., Gordon, S., Shephard, G.S., Sydenham, E.W., Marasas, W.F.O., Neson, PE, and Wilson, T.W., 1991. Levels of fumonisin B1, B2 in feeds associated with confirmed cases of equine leukoencephalomalacia, J. Agric. Food. Chem 392109-111. Thiel, P.G., Sydenham, E.W., Shephard, GS, and Schalkwyk, JV. 1993. Study of the reproducibility characteristics of a liquid chromatographic method for the determination of fumonisins B1 and B2 in com2IUPAC collaborative study. J. AOAC lntemational 76(2):361-366. Treadgill, G.J., Conrad, R.G.; Changchien, L.; and Cannon, M. 1986. Applicationof high-performance liquid chromatography to the purification and characterization of ribosomal protein L-3 from trichodennin-resistant yeast mutants. Biochem. J. 237: 421-426. TnJcksess, M.W., 1995. Committee on natural toxins, J. AOAC international, 78(1):135-141. Trucksess, M.W., Flood, M.T., and Page, S.W., 1986. This layer chromatographic determination of deoxynivalenol in processed grain products. J. Assoc. Off. Anal. Chem. 69(1):35-36. Truckscss, M.W., Flood, M.T., Mossoba, MM, and Page, S.W., 1987. High- perforrnance thin-layer chromatographic determination of deoxynivalenol, fusarenon-X, and nivalenol in bartey, com, and wheat. J. Agric. Food Chem. 35(4):445-448. Tuite, J. 1979. Field and storage conditions for the production of mycotoxins and geographic distribution of some mycotoxin problems in the United states. pp.19-39. Proceedings of a symposium, July 13, 1978, Michigan State University. Interactions of mycotoxins in animal production. National Academic of Sciences, Washington, DC. Tuite, J.. Shaner, G., Rambo, G., Foster, G., and Caldwell, R.W. 1974. The Gibberella ear of epidemics of corn in Indiana in 1965 and 1972. Cereal Sci. Today, 19:238-241. Tutelyan, V.A., Eller, K.l., Sobolev, V.S., Pinenova, V.V., Zasharova, LP. and Muzychenko, NJ. 1990. A survey of the occurrence of deoxynivalenol in wheat from 1986-1988 harvests in the USSR. Food Addit. Contam., 7:521- 525.. 207 Ueno, Y. 1980. Trichothecene mycotoxins, mycology, chemistry, and toxicology. Adv. Nutr. Sci. 31301-353. Ueno, Y. 1983. Tricothecenes: Chemical, biological and toxicological aspects; Elsevier Science Publisher B.V., Amsterdam, the Nedernald. Ueno,, Y., Sato, N., lshii, K., Sakai, K., Tsunoda, H. and Enomoto, M. 1973. Biological and chemical detection of trichothecene mycotoxins of Fusarium species. Appl. Microbiol. 25:699-704. Ueno, Y., Lee, U.S., Tanaka, T., Hasegawa, A., and Matsuki, Y. 1986. Examination of of chinese and U.S.S.R. cereals for the Fusarium mycotoxins nivalenol, deoxynivalenol, and zearalenone. Toxicon 24:618- 621. Usleber, E., Martlbauer, E., Dietrich, R., and Terplan, G. 1991. Direct enzyme- linked immunosorbent assays for the detection of the 8-ketotrichothecene mycotoxin deoxynivalenol, 3-acetyldeoxynivalenol, and 15-acetyl- deoxynivalenol in buffer solutions. J. Agric. Food Chem. 3922091-2095. Usleber, E., Schineider, E., Martlbauer, E., and Terplan, G. 1993. Two formats of enzyme immunoassay for 15-acetyldeoxynivalenol applied to wheat. J. Agric. Food Chem. 41:2019-2023. Usleber, E., straka, M., and Terplan, G., 1994. Enzyme immunoassy for fumonisin Bl applied to com-based food. J.Agric. Food Chem, 42:1392-1396. Van-Ness, J., Laemmli, UK, and Pettijohn, DE, 1984. Immunization in vitro and production of monoclonal antibodies specific to insoluble and weakly immunogenic proteins. Proc. Natl. Acad. Sci. USA, 81:7897-7901. Veldman, A., Borggreve, G.J., Mulders, E.J., and Lagemaat, D. V.D. 1992. Occurrence of the mycotoxins ochratoxin A, zearalenone, and deoxynivalenol in feed components. Food Addit. Contam. 9(6):647-655. Vesonder, R. F. 1983. Natural occurrence in North America, pp. 210-217. In: Trichothecenes. Chemical, Biological, and Toxicological aspects. (ed. Y. Ueno), Development in Food Sci. 4, Elsevier, Amsterdam. Vesonder, R.F., Ciegler, A., Jensen, AH. 1973. Isolation of the emetic principle from Fusarium-infected corn. Appl. Microbiol. 26:1008-1010. Vesonder, R.F., Ciegler, A., Rogers, R.F., Burbridge, KA., Borhast, R.J., and Jensen, AH. 1978. Survey of 1977 crop year preharvest corn for vomitoxin. Appl. Environ. Microbiol. 36:886-888. Vesonder, R.F., Ciegler, A., Rohwedder, W.K., and Eppley, R. 1979. Re- examination of 1972 Midwest corn for vomitoxin. Toxicon. 17:658-660. 208 Vesonder, R., Peterson, R., Plattner, R., and Weisleder, D. 1990. Fumonisin B1 : Isolation from corn cultures and purification by high performance liquid chromatography. Mycotoxin Res. 6:85-88. Vetro, l.B., Gyongyosi, A., and Solti. 1994. Monoclonal antibody-based enzyme- linked immunosorbent assay fo Fusarium -2 toxin and zearalenone toxin in cereals. App. Environ. Microbiol. 60(2):729-731. Visconti, A. and Doko, MB, 1994. Survey of fumonisin production by Fusarium isolated from cereals in Europe. J. AOAC. 77(2):546-550. Voss, K.A., Norred, W.P., Plattner, R.D., and Bacon, CW. 1989. Hepatotoxicity and renal toxicity in rats of corn samples associatedd with field cases of equine leukoencephalomalacia. Food Chem. Toxicol. 117297-104. Wang E.W., Norred, P., Bacon, X.E., Riley, RT, and Merrill Jr., AH 1991. Inhibition of sphingolipid biosyntheses by fumonisins: implications for diseases associated with Fusarium moniliforrne. J. Biol. Chem. 266214486- 14490. Wang, CR, and Chu, F.S. 1991. Production and characterization of antibodies against nivalenol tetraacetate. App. Environ. Microbiol. 57(4):1026-1030. Warden, B.A., Allam, K., Sentissi, A.. Cecchini, DJ, and Giese, R.W. 1987. Repetitive hit and run fluoroimmunoassay for -2 toxin. Anal. Biochem. 162:263-265. Ware, G.M., Cannan, A., Francis, 0., and Kuan, S. 1984. Gas chromatographic detenninaton of deoxynivalenol in wheat with electron capture detection. J. Assoc. Off. Anal. Chem. 67:731-734. Warner, R. and Pestka, J.J. 1986. Screening for zearalenone in corn by competitive direct enzyme-linked immunosorbent assay. J. Agric. Food Chem. 342714-717. Warner, R. and Pestka, J.J. 1987. ELISA survey of retail grain-based food products for zearalenone and aflatoxin B1. J. Food Prot. 50(6):502-503. Watson, S.A., Mitocha, C.J., and Hayes, A .W.; 1984. Analysis for trichothecenes in sample from southeast asia associated with “yellow rain”. Fund. Appl. Toxic. 42700-717. Whitaker, T.B., Dickens, J.W., and Giesbrecht, PG. 1986. Optimum methanol concentration and solvent/peanut ratio for extraction of aflatoxin from raw peanuts by modified AOAC methods. J. Assoc. Off. Anal. Chem. 69:508- 510. 209 Whitlow, L.W., Nebel, R.L., and Behlow, RF. 1985. Mycotoxin survey results. Proceedings of the dairy fieldmen and sanitarians conference, February 18-19, 1985, pp.61-62. Whitlow, L.W., Nebel, R.L., Behlow, R.F., Nagler, W.M., and Brownie, C.F.G., 1986. A survey of North Carolina dairy feeds for mycotoxins. Dairy Fieldmen and sanitarians conference proceedings, Februay 17-18, 1986. pp.34-35. Whitlow, L.W. and Hagler, W.M., 1987. The association of productivity losses in dairy cows with deoxynivalenol. Proceedings from a paper presented at a symposium on Recent Developments in the study of mycotoxins, Rosemont, Illinois, Desember 17, 1987. Wilson, T.M., Nelson, PE, and Knepp. CR. 1985. Hepatic neoplastic nodules, adenofibrosis, and cholangiocarcinomas in male Fischer 344 rats fed corn naturally contaminated with Fusarium moniliforme. Carcinogenesis 621155- 1160. Wilson, T.M., Ross, P.F., Rice, L.G., Osweiler, G.D., Nelson, H.A., Owens, D.L., Plattner, R.D., Reggiardo, C., and Noon, TH. 1990. Fumonisin B1 levels associated with an epizootic of equine leukoencephalomalacia. J. Vet. Diagnost. Invest. 22213-216. Winkler, GC 1988. Pulmonary intravascular macrophages in domestic animal species: Review of structural and fuctional properties. Am.J.Anat. 1812217- 234. Winter, G., Griffiths, A.D., Hawkins, RE, and Hoogenboom, HR. 1994. Making antibodies by phage display technology. Annu. Rev. Immunol. 122433455. Wood, GE, and Carter, C. 1989. Limited survey of deoxynivalenol in wheat and corn in the United States. J. Assoc. Off. Anal. Chem. 72:38-40. Xu, Y.C., Zhang, GS. and Chu, F.S. 1986. Radioimmunoassay of deoxynivalenol in wheat and corn. J. Assoc. Off. Anal. Chem. 69:967-969. Yong, J.C., Fulcher, R.G., Hayhoe, J.H., Scott, PM, and Dexter, J.E. 1984. Effect of milling and baking on deoxynivalenol (vomitoxin) content of eastern canadian wheats. J. Agric. Food Chem., 32:659-664.. Yoshizawa, T. and Morooka, N. 1973. Deoxynivalenol and its monoacetate: new mycotoxins from Fusarium roseum and moldy barley. Agric. Biol. Chem. 3722933-2934. Yoshizawa, T. and Morooka, N. 1974. Studies on the toxic substances in the infected cereals (Ill). Acute toxicity of new trichothecene mycotoxins: deoxynivalenol and its monoacetate. J. Food Hyg. Soc. Jpn. 15:261-269. 210 Yoshizawa, T., Yamashita, A., and Luo, Y. 1994. Fumonisin occurrence in corn from high- and low-risk areas for human esophagela cancer in china. App. Environ. Microbiol. 60(5): 1626-1629. Zhang, G.S., Li, S.W., and Chu, F.S., 1986. Production and characterization of antibody against deoxynivalenol triacetate. J. Food Protect. 49(5)2336-339. L 9 narrow STATE UNIV. IBR iiiiililliiilliilliiiIiiIiiiiliilliilliili 312930141723 RIES 85