PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. DATE DUE I DATE DUE DATE DUE MI} I 2 2 L! 1 (I 1/” WWW“ THE FUNCTION, ACCUMULATION AND LOCALIZATION OF THE NOR-l PROTEIN INVOLVED IN AFLATOXIN BIOSYNTHESIS; THE FUNCTION OF THE I'IUP GENE ASSOCIATED WITH SPORULATION IN ASPERGHL US PARASITICUS BY RENQIN G ZIIOU 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 and Multidisciplinary Graduate Program in Environmental Toxicology 1997 ABSTRACT THE FUNCTION, ACCUMULATION AND LOCALIZATION OF THE NOR-l PROTEIN INVOLVED IN AFLATOXIN BIOSYNTHESIS; THE FUNCTION OF THE HUP GENE ASSOCIATED WTTH SPORULATION IN ASPER GILL US PARASI 77 C US BY Renqing Zhou Aflatoxin contamination can cause public health and economic problems in many areas of the world. A study of the mechanisms regulating aflatoxin biosynthesis and the biology of the aflatoxigenic fungus Aspergillus parasiticus may play a role in controlling aflatoxin contamination. ' To study the function of the Nor-l protein, which is directly involved in aflatoxin Br biosynthesis in A. parasitic-us, a nor-1 cDNA was expressed in Escherichia coli. The resulting Nor-1c protein was evaluated in an enzyme activity assay. To study the acammlation of the Nor-l protein in A. parasiticus, polyclonal antibodies were raised against the Nor-1c protein and were used to monitor the accumulation of the Nor-1 protein. A nor-llGUS reporter construct was also used to monitor the activity of the nor-l promoter. Cell fi’actionationN/estem blot analysis and in situ immunolocalization experiments were used to study the localization of the Nor-l protein. A nor-I/GU S reporter construct was used to localize the activity of the nor-1 promoter. Gene disruption was used to study the function of the fluP gene which encodes a putative polyketide synthase in A. parasiticus. The data presented in this dissertation suggest that: The Nor-l protein is a ketoreductase which converts the aflatoxin B1 pathway intermediate norsolorinic acid to averantin. The accumulation of the Nor-1 protein is regulated temporally in liquid media and temporally/spatially on solid media. The Nor-1 protein is mainly localized in the cytosol of the vegetative cell. The Nor-l protein is localized in developing conidial head, conidial stalks, vegetative hyphal cells, and mature conidia in the order fi'om the highest to the nondetectable level, respectively. The localization of the Nor-l protein and the localization of nor-1 promoter activity follow a temporal and spatial pattern and are correlated to the process of spomlation in A. pausiticms grown on solid media. Disruption of fluP resulted in slow growth with cotton-like hyphae, no sporulation, and reduced aflatoxin accumulation suggesting that fluP is associated with the vegetative cell growth, sporulation, and in turn indirectly influences the accumulation of aflatoxin in A. parasiticus. This work is dedicated to my aunt (Zhou Chunlan), my uncle (Ma Zhichun), my grand mother (Cong Guicang) and my sister (Ma Lijun) for their emotional and financial support during the most difficult time in my life; and to my daughter (Zhou Ymgchiao) for her understanding and self sacrifice during my seven-year absence, working and studying in the United States. iv ACKNOWLEDGMENTS I would like to deeply acknowledge the academic and technical guidance and advice of Dr. John E. Linz throughout my graduate study at Michigan State University. It was his continuous encouragement and inspiration that helped me succeed in these studies. I am very grateful to receive the advice and help from Dr. James J. Pestka, Dr. William G. Heiferich, and Dr. L. Patrick Hart who have graciously served on my guidance committee. Special thanks are also expressed to Matt Rarick for his assistance in sequence analysis, and last but not least to Dr. Tzong-Shoon Wu, Dr. Ludmila Rose, Dr. N‘mee Mahanti and all of my colleagues in the Food Microbiology Laboratory, the Department of Food Science and Human Nutrition (Michigan State University) for their technical expertise. TABLE OF CONTENTS LIST OF TABLES .................................................................................. x LIST OF FIGURES .................................................................................. CHAPTERI .xi 1 LITERATURE REVIEW ............................................................................. 5 PART 1.MYCOTOXIN BIOSYNTHESIS IN ASPERGEL US ................ 5 SECONDARY METABOLISM... ...5 FUNCTIONS or SECONDARY METABOLI‘I'ES......................................' 1...... .' .' .....7 BIOSYNTHESIS OF MYCOTOXINS BY ASPERGILLUS Biosynthesis Of Isoprenoid Mycotoxrns Isopentenyl Pyrophosphate... . Hexahydropolyprenols... Biosynthesis Of Amino Acrd-Isoprenord Mycotoxms Funntremorgrn Biosynthesis Of Polyketrde Mycotoxms ............................................... Pamlm... Stengmatocystm, Stengmatrn, ”and Autocystrn ................................. Biosynthesis Of Amino Acid-Polyketide Mycotoxins” Ochratoxin... Biosynthesis Of Polypeptide Mycotoxms ............................................ Dicoumarol... . Gliotoxin... .... TryptoqurvalrneM Biosynthesis Of Isoprenord-Polyketide Mycotoxrns Fumagillin 8 ll .....11 .....13 ......13 .....13 ...16 16 ......18 ......18 ...19 21 22 ......22 ...24 24 ......25 ......25 ......27 .....29 ......32 .....32 ...32 TerritremB... . ............34 Biosynthesis Of Amino Ac1d-Isopreno1d-Polyket1de Mycotoxms ................. 36 Cyclopiazonic Acid... . . .. 36 Epilog.... 37 TOXICITY OF AFLATOXIN S ................................................................ 37 BIOSYNTHESIS OF AFLATOXIN B1 .................................................... 42 Biosynthesis Of Aflatoxin B1.. 42 Genes Involved In The Aflatoxin B1 Pathway ................................... 51 Purification OfEnzymes Involved In The AFBr Pathwa... 56 Regulation Of Aflatoxin Biosynthesis. . 56 BIOCONTROL OF AFLATOXIN CONTAMINATION .............58 Possible Function Of Aflatoxins ......................................................... 58 Species OfTheAqoergilIusflavus Group... 58 BiocontrolOfAflatoxin Contamination... ... 60 Perspectives On The Use Of Fungal Biocontrol Agents .................................... 64 CHAPTER2 ENZYMATIC FUNCTION OF THE NOR-l PROTEIN INVOLVED IN AFLATOXIN B1 BIOSYNTHESIS IN ASPERGHLUS PARASIHCUS.... . . .........67 ABSTRACT ................................................................................... 67 MATERIALS AND METHODS .. 71 BacterialStrain, Fungal Strams,andCultureCond1trons .........71 Nucleotide Sequence Analysis... ........71 Construction of pMN1,a pMAL-c2-nor-l-cDNA Expression Vector ......... 73 Preparation of the Nor-ICIMBP Fusion Protein and the Nor-lo Protein ...... 73 Fungal Culture and Crude Protein Extract Preparation.....................74 lmmunoafinity Ptnification of the Native Nor-1 Protein ........................... 75 Western Blot Analysis... 75 NorsolorinicAcid (NA)Preparatron 76 Averantin (AVN) Preparation... .7.......6 Enzyme Activity Assay of the Nor-lolMBP Fusion Protein and the Nor-1c protein .................................................................. 77 UV/VIS, Mass, and NMR Spectroscopy of NA and AVN... ............. 77 Aflatoxin Analysis by TLC and ELISA78 RESULTS ........................................................................................... 78 Sequence Analysis ofthe nor-l cDNA78 Preparation of the Nor-ldBMP Fusion Protein and the Nor-1c Protein ................................................................................... 82 Immunoafinity Purification ofthe Native Nor-1 Protein 82 Enzyme Activity of the Nor-le/BMP Fusion Protein and the Nor-1c Protein .................................................................................... 82 UV/VIS, Mass, and NMR Spectra ofNA85 UV/VISMass,andN1VlRSpectraofAVN.......89 AFBr Accumulation m A. parasiticus SU-l and A. parasztzcus ANor-1.90 CHAPTER 3 THE ACCUMULATION OF THE NOR-l PROTEIN AND THE ACTIVITY OF THE NOR-l PROMOTER DURING AFLATOXIN Br BIOSYNTHESIS IN ASPERGEL US PARASITICUS .......................................................................... 95 ABSTRACT ........................................................................................................ 95 INTRODUCTION ............................................................................ 95 MATERIALS AND METHODS ............................................................. 98 Bacterial Strain, Fungal Strains, and Growth Media ........................... 98 Fungal Culture, Dry Weight Determination, and Analysis of Fungal Morphology .................................................................................... 99 Temporal and Regional Collection of Samples from Solid Growth Medium ............................................................................ 99 Crude Protein Extract Preparation... .100 Preparation and Purification of Polyclonal Antrbodres 101 Western BlotAnalysisoftheNative Nor-1 Protein" .......102 Aflatoxin Analysis By TLC and ELISA.102 Quantitative Assay of B-Glucuronidase Activity ................................... 103 RESULTS ...................................................................................... 103 Titer and Specificity of Polyclonal Antibodies... ... ..103 The Accumulation of the Nor-l Protein and the Accumulation of AFB1 in A. parasiticus SU-l Grown in YES Liquid Medium .............. 105 The Accumulation of the Nor-l Protein and the accumulation of AFBr in A. parasiticus SU-l in Nutritional Shift Assay ....................... 107 Morphology of A. pm'asiticus SU-l grown on YES Agar .................. 109 Morphology of Development and the Accumulation of AFBr at theColonervelinA.parasiticus Grown onYESAgar ................ 111 The Accumulation of the Nor-1 Protein, the Activity of the nor-l Promoter, and the Accumulation of Aflatoxin in A. parasiticus SU-l Grown on YES Agar ...................................... 112 DISCUSSION ................................................................................ l 16 CHAPTER4 LOCALIZATION OF NOR-l PROTEIN ACCUMULATION AND LOCALIZATION OF nor-l PROMOTER ACTIVITY DURING AFLATOXIN Bl BIOSYNTHESIS IN ASPERGHLUS PARASHTCUS..........124 ABSTRACT ...................................................................................................... 124 Fungal Strains and Culture Media... 127 Cell Fractronatron128 viii Western Blot Analysis of the Nor-1 Protein in Cell Fractions .................... 130 Analysis of Marker Enzyme Activities ........................................................ 131 Nor-1 Protein Localization by Immunofluorescence Microscopy ............. 133 Semiquantitative Petri Dish Assay for nor-l/GUS Activity (Overlay Procedure) ................................................................................. 134 RESULTS... .. ...134 Distributions of Marker Enzymes and the Nor-1 Protein 111 "Cell Fractrons 147 Subcellular Immunolocalization of the Nor-1 Protein... ...147 In Situ Localization of nor-1 Promoter Activity in a Colony of" A. parasiticus ........................................................................................... 148 DISCUSSION ............................................................................................................ 152 CHAPTERS ISOLATION AND ANALYSIS OF HUP, A GENE ASSOCIATED WITH HYPHAL GROWTH AND SPORULATION IN ASPERGEL US PARASIHCUS ............... 159 ABSTRACT ..................................................................................................... 159 INTRODUCTION... .. 159 MATERIALS AND METHODS .161 Bacterial Strain, Fungal Strains and Culture Condrtrons ..........161 Fungal Culture, Dry Weight Determination, Growth Rate Measurement, and Aflatoxin Analysis ............................................. 161 Temporal and Regional Collection of Samples fi'om Solid Medium ..... 163 Plasmid Preparation and Protoplast Transformation ................................. 163 Nucleotide Sequence Analysis and Amino Acid Sequence Comparison....164 Isolation and Analysis of Genomic DNA and Total RNA164 Analysis of the Codrng Region offluP 165 Construction of a Disruption Vector pPRS ...............168 Transformation of A. parasiticus NR-3 and Disruption of fluP ................. 173 fluP Transcript Accumulation 1n the erd-Type Strain and thefluP-Disnrpted Transformant... 181 Aflatoxin B1 Accumulation 1n the erd-Type Strain and thefluP-Disrupted Transformant... 182 CONCLUSIONS 192 APPENDICES APPENDIX A. CLONING OF THREE EcoRI DNA FRAGMENT S FROM COSMID Nor-A AND ANALYSIS OF RELATED TRANSCRIPTS ............ 194 APPENDIX B. IDENTIFICATION OF VERSICONAL CYCLASE GENE ......... 195 APPENDIX C. A MODIFIED PROCEDURE FOR CRUDE EXTRACT PREPARATION FROM FUNGAL CELLSl96 ix LIST OF TABLES Table 3-1. Titers and specificities of anti Nor-lc/MBP PAb ..................................... 105 Table 3-2. The accumulation of aflatoxin Bl in A. parasiticus SU-l in nutritional shifi Table 3-3. The morphological characteristics and the acmnmlation of AFB: in A. parasitic-w SU-l gown on YES agar112 Table 3-4. The accumulation of the Nor-l protein, the activity of nor-l/GUS (GUS activity), and the accumulation of AFB] in A. parasiticus SU-l gown on YES agar ............................................................................... 115 Table 4-1. Distributions of marker enzymes and the Nor-l protein in cell fi'actions obtained by Potter homogenization and difi‘erential centrifitgation..........l38 Table 4-2. Distributions of marker enzymes and the Nor-l protein in total organelle and the cytosol fractions obtained by protoplast pelmeabilimion and ceuuifitgation. ............................................................ 139 Table 5-1. Mycelial dry weight and aflatoxin Bl accumulation in A. pwasitz’cus NR-3 and the fluP-disrupted transformant gown on YES agar ................ 183 LIST OF FIGURES Figure 1-1. Representatives of mycotoxins produced by species of Aspergillus........ . . . .. Figure 1-2. Biosynthesis of isopentenyl pyrophosphate from acetyl CoA... Figure 1-3. Biosynthesis of hexahydropolyprenols in A3pergill113fi1miga1113... . . . . . . . . . Figure 1-4. Biosynthesis ofaflatrem inAspergiIIusflavus................ Figure 1-5. Biosynthesis offumitremorgin A inA.spergiIlu3fi1migalu3......... Figure 1-6. Biosynthesis of sulochrin in A3pergill1131erre113................. Figure l-7.Biosynthesis ofpatulininPeniciIliumpa111h1m...................................... Figure 1-8. Biosynthesis of citrinin in Aspergillus candzdus Figure 1-9. Biosynthesis of sterigmatocystin, sterignatin, and austocystin in Avergillus ver31'color .......................................................... Figure 1-10. Biosynthesis of ochratoxin in Aspergillus ochraceu3 Figure 1-11. Biosynthesis of cytochalasin B in Aspergillus clava1113 .................... Figure 1-12. Biosynthesis ofdicoumarol inA.spergiIlu3fi1miga1113...... Figure 1-13. Biosynthesis ofgliotoxin in Trichodema vmdz Figure 1-14. Biosynthesis of tryptoquivaline M in Aspergillusfianigatus.................. Figure 1-15. Biosynthesis ofpenicillin in Aspergillus nidulam......... 28 Figure l-16. Biosynthesis ofaustin inAspergiIlus 1131113... Figure 1-17. Biosynthesis offumagillin in Aspergillusfimigatus............. 10 12 12 14 ..15 15 .20 .20 20 23 23 .26 .26 28 .33 .33 Figure 1-18. Biosynthesis ofterritrem B inAspergillus 1erre113............ 35 Figure 1-19. Biosynthesis of cyclopiazonic acid in some species of Aspergillus. . . ........35 Figure 1-20. Structures ofAflatoxin B1, Bz, Gr, and 62 41 Figure 1-21. Origin of the 16 skeletal carbon atoms of aflatoxin Br ........................ 41 Figure 1-22. Aflatoxin B1 biosynthetic pathway in Aspergillus parasmcus43 Figure 1-23. Restriction endonuclease and transcript map of cosmid Nor-A.................53 Figure 2-1. Proposed multiple alternative pathways which convert NA to AVF. ............ 69 Figure 2-2. Construction of the expression vector pMN 1 ......................................... 72 Figure 2-3. Nucleotide and predicted amino acid sequence of the S’and 3’-ends of nor-1 cDNA in expression vector pMN179 Figure 2-4. SDS-PAGE detection of the Nor- 1 c/MBP fusion protein and the Nor-1c protein ................................................................... 80 Figure 2-5. Western blot analysis of the native Nor-l protein in A. pw'asiricus SU-l ......... 81 Figure 2-6. Enzyme activity of the Nor-l/MBP filSlOll protein and the Nor-1c protein ......... 83 Figure 2-7. UV/VIS spectroscopy of norsolorinic acid (NA) and averantin (AVN) ........... 86 Figure 2-8. Mass spectroscopy ofnorsolorinic acid (NA) and averantin (AVN)...............87 Figure 2-9. NMR spectroscopy of norsolorinic acid (NA) and averantin (AVN). . . . .........88 Figure 3-1. Western blot analysis demonstrating the specrficrty of PAb from different levels ofpurification.... ......104 Figure 3-2. Western blot analysis of the accumulation of the Nor-1 protein in A. para31‘11‘c113 SU-l gown in YES liquid medium106 Figure 3-3. The accumulation of AFBi and mycelial dry weight of A pw'asiticus SU-l gowninYES liquid medium... .. ... 108 Figure 3-4. Western blot analysis of the accumulation of the Nor-1 protein in A. parasiticus SU-l in PMS-GMS nutritional shifi assay... 110 xii Figure 3-5. Western blot analysis of the accumulation of the Nor-1 protein in A. pamsitr’cus SU-l gown on YES agar... Figure 3-6. TLC analysis of the accumulation of aflatoxins in A.para31’11'cus SU-l gown on YES agar. Figure 4-1. Western blot analysis of the Nor-1 protein in cell fractions of A. parasitic“ .................................................................................... Figure 4-2.1mnnmolocalization of the Nor-1 protein in hyphalm cells of A. pm'asm'cus SU-l gown in YES liquid medium... Figure 4-3. Inummolocalization of the Nor-l protein in A. paras’ticus SU-l gown onYESagar... ...................................................................................... Figure 4-4. Imrmmolocalization of the Nor-l protein in A. parasificus SU-l by laser scanning microscopy (LSM) .................................................................... Figure4-5. Schematicofa conidiophore ofAmergiIlusparasiticus..... FigunMInsimanalysisofnw-l promoteractrvrtywrthmacolonyof A. pry-1131110113 gown on solid medium... Figure$LAminoacid sequencecomparisonofflquithtwoPKS genesandoneFAS gene ............................................................................................................ Figure 5-2. A schematic ofthe 9.1-kb SacI genomic DNA fragnent containing the fl11P gene in A3pergr’ll113 par-03mm ................................................... Figure 5-3. The fluP-disruption vector pPKS and a schematic description of the expected restriction map ofgenomic DNA in thefluP-disrupted transformant........ Figure 5-4. Southern blot analysis of genomic DNA in A. parasitic-113 NR-3 and the fluP-disrupted transfonnant .................................................................. 113 ...114 136 ...140 142 144 146 ...149 166 168 169 ....171 Figure 5-5. Morphology of A. parasitic-113 NR—3 and the fluP-disrupted transformant gown on coconut agar medium (CAM) or on potato dextrose agar (PDA) ..... 174 Figure 5-6. Transcript accumulations of fluP, the B-tubulin gene, and the ver-rA gene in A. parasiticus. .. ................................................................................... Figure 5-7. Transcript accumulations of fluP in A. parantrcus NR-3 and the fluP-drsrupted transformant... xiii ....177 ...179 INTRODUCTION Aflatoxins are extremely toxic mycotoxins produced by certain strains of Aspergillus parasitic-113, A. flaws, and A. 1101111113. These ubiquitous fungi gow on a variety of food and feed crops and may cause serious aflatoxin contamination. Aflatoxin contamination poses a threat to public health and in turn causes economic problems in many areas of the world. Therefore, reducing or eliminating aflatoxin contamination is an urgent need. Myresearchisonepartofalongtermresearchprojectaimedattheeliminationof prebarvest contamination of crops by aflatoxin Bl (AFBl). The work is designed to provide an understanding of the mechanisms which regulate aflatoxin biosynthesis at the molecular level and an understanding of the general biology of the toxigenic firngi (e.g. regulatory mechanisms involved in hyphal gowth, hyphal differentiation, and sporulation). This information may lead to the development of new fimgicides, the improvement of agonomic practices, the development of resistant crops (by genetic engineering or classical breeding), and the development of biocontrol agents (such as nontoxigenic competitive fungi) for control of aflatoxin contamination. In the currently accepted general pathway of aflatoxin biosynthesis (Figure 1-22) (Bhatnagar e1 at, 1992; Dutton, 1988; Yabe etal, 1993), the decaketide—derived norsolorinic acid (NA) is the earliest stable pathway intermediate isolated (Dutton, 1988). NA is converted to averufin (AVF) via one or more alternative pathways by converting NA to averantin (AVN) first or to averufanin (AVNN) first (Figure 2-1) (Bennett and Cluistensen, 1983; Button, 1988; McCormick e1 11]., 1987; Yabe et al., 1991b) Disruption of the nor-1 gene resulted in NA accumulation and a significant reduction of aflatoxin in an aflatoxigenic strain of A. parasitim (Trail et at, 1994). Therefore the nor-1 gene may function in one of these alternative pathways. Analysis of the predicted amino acid sequence of nor-1 provided evidence that nor- 1 encodes a ketorcductase (Trail e1al., 1994). Comparison ofthe chemical structure ofNA and AVN (Figure 2-1) demonstrated that the 5’-keto goup of NA is converted to the 5’- hydroxyl goup of AVN suggesting that this reaction is catalyzed by a ketoreductase. The accunmlation of nor-l transcript is subjected to temporal regulation in liquid arlmre (Skory et al., 1993; Trail e1 01., 1994). Aflatoxins have been found to be concentrated in spores and sclerotia (Wicldow and Cole, 1982; Cotty, 1988). These data mggest that the accumulation of the Nor-1 protein is also subjected to temporal regulation in liquid culture and the localization of the Nor-1 protein is correlated to the process of sporulation. Based on thebackgound data, three hypotheses related to theNor-l protein are proposed. (1) The Nor-l protein encoded by nor-l is a ketoreductase which converts NA to AVN in the AFBr pathway. (2) The accumulation of the Nor-l protein is subjected to temporal regulation in liquid media and to temporal and spatial regulation on solid media. (3) The Nor-l protein resides either in the cytosol or in organelles. The localization of the Nor-1 protein is correlated to the process of sporulation in A. marine-113 gown on solid media. Previous data showed that a 680-bp genomic DNA fiagnent fiom A. par-1131110113 was able to hybridize to a cDNA fi'agnent of a polyketide synthase (PKS) gene which encodes the B- ketoacyl synthase functional domain of the 6-methylsalicylic acid synthase (MSAS) complex in Penicifliwnpatuhan (Becketal, 1990) As expected, the deduced amino acid sequence fi'om the 680-bp DNA fi'agnent was similar (72% identity) to that of the B-ketoacyl synthase functional domain of MSAS. A search of the EMBL and GenBank data base h‘braries showed that the 680-bp DNA fi'agnent is also very similar to a variety of other PKS genes (35.3% to 70. 8% identity in 216 amino acid overiap compared with 6 PKS genes). Because aflatoxins are derived from a polyketide pathway (Kurtzman e101. , 1987), the producing fungi are predicted to have a polyketide synthase (PKS) related to aflatoxin biosynthesis. ItmshufiaflyhypothenzedflntflqumdbcmvohedmaflMmdnbiosymhedsbecause there had been no published report on identifying patulin and/or its precursor 6-methylsalicylic acid produced by A. parasiticus. Polykides are secondary metabolites and are often involved in regulation of development or difi‘erentiation (Summers et al., 1995; Revill et al., 1995; Keller and Adams, 1995). Therefore an alternative hypothesis was that this PKS-hke gene (fluP) containing the 680-bp DNA fiagment might be related to some kind of secondary metabolism (such as pignent synthesis, hyphal gowth and/or sporulation). In order to test these hypotheses, four goals were proposed: (1) to determine the enzymatic function of the Nor-1 protein; (2) to determine the accmnulation pattern of the Nor-1 protein; (3) to localize the accumulation of the Nor-1 protein; and (4) to test the firnction of the fluP gene. Fourexperimentsweredesigredtofiilfillthefourgoalsandintumtoaddressthefour hypotheses. (1) The Nor-1c protein was produced in a host bacterium transformed with a nor- 1 cDNA expression vector. The Nor-1c protein was used to test the enzymatic function of the native Nor-1 protein in the AFB! pathway. (2) Polyclonal antibodies (PAb) were raised against the Nor-1c protein and used to monitor the accumulation of the native Nor-1 protein in A. pausitiws gown in liquid media and on solid media A nor-l/GUS reporter construct was used to monitor the activity of the nor-1 promoter (which could indirectly indicate the accunmlation of the Nor-1 protein during aflatoxin biosynthesis) (3) Cell fiactionation plus Western blot analysis and in situ irnmunolocalization were used to localize the Nor-1 protein at thecellularlevel andthebyphal level. Anor-l/GUS reporter constructwasusedto localize the activity of the nor-l promoter (which could indirectly indicate the localintion of the Nor-1 protein during aflatoxin biosynthesis) at the hyphal level and at the colony level (4) Southern blot and Northern blot analyses were used to clone the fluP gene. Gene disruption was used to arnlyzethefimctionoftheflngene. CHAPTER] LITERATURE REVIEW My dissertation focuses attention on two genes. The first is the nor-l gene which is involved in afiatoxin Br (AFBr) biosynthesis. The second is a suspected polyketide synthase (PKS) gene, the fluP gene, which is associated with gowth and sporulation and indirectly influences AFBr biosynthesis in the filamentous firngus Aspergillus para31‘tic113. AFBr is just one of the numerous mycotoxins produced by Aspergillus. The first part of this review is a general introduction to mycotoxin biosynthesis in Aspergillus, which includes a brief discussion of secondary metabolism and its possible functions and several biosynthetic pathways for representative mycotoxins produced by species of Aspergillus. This dissertation is a part of a long term research project desigied to provide an understanding of the regulation of aflatoxin biosynthesis which may lead to the development of biocontrol agents for control of aflatoxin contamination. The second part of this review includes a discussion of aflatoxin toxicity and biosynthesis, and biocontrol of aflatoxin contamination. PART 1. MY COTOXIN BIOSYNTHESIS IN ASPERGHL US SECONDARY METABOLISM Mycotoxins are a large and highly varied goup of natural products referred to as secondary metabolites. Chemical compounds are synthesized and degaded by means of a series of chemical reactions mediated by enzymes in the living organism. These processes are known as metabolism which is comprised of catabolism (degadation) and anabolism (synthesis). All organisms possess very similar primary metabolic pathways by which they synthesize and degade certain biochemicals such as sugars, amino acids, common fatty acids and nucleotides to generate energy and to build the polymers derived from them (polysaccharides, proteins, lipids, RNA, and DNA). Compounds derived fiom primary metabolic pathways are essential for the survival and well-being of the organism and are called primary metabolites. Most organisms also utilize other metabolic pathways to produce compounds which appear to have no apparent utility. These compounds are called secondary metabolites. The pathways of synthesis and degadation of secondary metabolites constitute secondary metabolism. These pathways are perhaps only activated during particular stages of gowth and development or during periods of stress caused by factors such as nutritional limitation or microbial attack. Most secondary metabolites are produced only by a specific species or a small number of closely related species. For example, aflatoxins have been found to be produced only by certain strains of three Aspergillus species. Primary metabolites have the same or similar structures such as the 20 common anrino acids, RNA and DNA, while secondary metabolites have a wide diversity of structures. Their unusual chemical structures include B-lactam rings, cyclic peptides containing ‘unnatural’ and non-protein amino acids, unusual sugars and nucleosides, unsaturated bonds of polyacetylenes and polyenes and large macrolide rings. Examples of some of the well known secondary metabolites with unusual structures include ephedrine (for respiratory ailments), ricinine (a purgative), and salicin (aspirin is a synthetic analogue) which are medicines; strychnine, coniline, and rotenone which are poisons; morphine (fiom opium), tetrahydrocanabinol (from marijuana), and cocaine which are narcotics and hallucinogens; caffeine which is a stimulant; geraniol (from rose oil), linalol (fiom lavender), cinamaldehyde (fi'om cinamon), eugenol (fi'om cloves) and diallyl disuphide (fi'om garlic) which are perfumes and spices. The organisms which can produce secondary metabolites are usually considered to be limited to bacteria, firngi, and plants. But some scientists believe that all organisms including humans can produce secondary metabolites. For example, defensins, which are endogenous antibiotic peptides produced by human leukocytes may be considered as secondary metabolites (Ganz etal., 1985). There is no unanimous definition of secondary metabolism. However, one proposed definition may provide some hints about the nature of secondary metabolism. Davies (1992) proposed that primary metabolism enables an organism to produce the next generation. Secondary metabolism is responsible for interactions between the organism and its environment and is concerned with what is going on outside the producing organism, rather than events going on inside. FUNCTIONS OF SECONDARY METABOLITES The role of secondary metabolites for the producing organisms is still an area of vigorous debate, but there is a gowing consensus that secondary metabolism benefits the organism and this benefit usually reflects the nature of the metabolites themselves (Davies, 1992). Davies has proposed ten potential biological firnctions of secondary metabolites: (l) competitive weapons against other organisms including bacteria, firngi, plants, amoebae, and insects; (2) metal-transporting agents; (3) plant-microbe symbiosis; (4) nematode-microbe symbiosis; (5) insect-microbe symbiosis; (6) sexual hormones (pheromones); (7) difi'erentiation efl‘ectors between and within cells; (8) excretion of unwanted products; (9) products of ‘selfish’ DNA; (10) reserve pool of new biochemical pathways. BIOSYNTHESIS OF MY COTOXIN S BY ASPERGEL US Fungal secondary metabolites are relatively small molecules characterized not only by their structural diversity (Turner and Aldridge, 1983), but also by their diversity of biological activity which includes antibiotic activity, phytotoxicity, animal toxicity and a very large array of physiological and pharmacological activities in a mammalian system. The toxic secondary metabolites produced by firngi are called mycotoxins. The diversity of mycotorn'ns produced by Aspergillus was reviewed by Moss (1977, 1994). The precursors of mycotoxins produced by Aspergr'llus species are certain amino acids, acetyl CoA, and isopentenyl pyrophosphate. The polymers of the three basic building units are polypeptides (fi'om amino acids), polyketides (from acetyl CoA), and isoprenoids (fi‘om isopentenyl pyrophosphate). Acetyl CoA is also the precursors of some amino acids and isopentenyl pyrophosphate. Not only can the mycotoxins from Asvergillus be derived fi'om the three kinds of precursors respectively, but also fi'om all the possible combinations of the three kinds of precursors. The broad range of mycotoxins produced by Aspergillus are contained in seven goups: (l) isoprenoids; (2) amino acid-isoprenoids; (3) polyketides; (4) amino acid-polyketides; (5) polypeptides; (6) is0prenoid-polyketides; (7) amino acid-isoprenoid-polyketides. Some of the well known mycotoxins produced by Aspergillus are listed in Figure 1-1 along with their likely producers (may not be the major producers). In addition to the mycotoxins listed in Figure 1-1, many other Aspergillus mycotoxins (each with one producer) have been identified such as sulochrin (A. 1erre113), dicoumarol (A. fi1migatr13), aflatrem (A. flaws), fumitremorgin (A. frmrr’gatus), emodin (A. alutaceus), kojic acid (A. a1111ace113), neoaspergillic acid (A. alutaceus), penicillic acid (A. 111111042113), secalonic acid A (A. alutaceus), gandidufin (A. candidus), terphenyllin (A. candidu3), xanthoascin (A. cardidres), ascladiol (A. clavatu3), clavatol (A. clava1113), kotanine (A. clava1113), aflavinin (A. flaws), aspergillic acids (A. flavr13), paspalinin (A. flaws), firmigaclavines (A. fi1miga1113), firmigatin (A. fi1m1'ga1113), fumitoxins (A. fimrr'gatus), spinulosin (A. flanigatus), verruculogen (A. fimrr’gatus), naphthoquinones (A. oryzae), nigagillin (A. niger), aspergillomarasmin (A. oryzae), 3-nitropropionic acid (A. oryzae), oryzacidin (A. oryzae), nidulatoxin (A. sydawii ), giseofirlvin (A. sydowii), terrein (A. terreus), terreic acid (A. 1erre113), terretonin (A. 1erre113), terredionol (A. terre113), austarnid (A. 1131113), wentilacton (A. wentir), and physicon (A. wenriz) (Frisvad, 1988). With some exceptions such as 3-nitropropionic acid fi'om A. oryzae, the biosynthetic pathways of most mycotoxins (as for many other secondary metabolites) in A3perg1'll113 10 ....................................................................................... gacid anary metabohsm Isopentenyl Acetyl CoA Amino acids pyrophOSPhate (Malonyl CoA) . 1 Isoprenoids _, Polyketides Polypeptides (1)1 :Hexahydro-E polyprenols --------------- (6) (3)" "(4) (5) EFumagillin i Patulin Ochratoxin iGliotoxin 5 (A. fi1miga1113), (A. sclerofiorum),§ (A. alliaceus), :(A. famigatus), 5Austin 5 Austocystins 5 5 Aspochalasin 5Malforrnins ' 5(A. 1131113), 5 (A. 1131113), 5 E A. microcysticus),§ (A. niger), iTerritrems Citreoviridin Cytochalasin E, Xanthocillin 5 (A. 1erre113), 5 (A. terreus), 5 5 (A. clavatus), 5 A. chevalieri), 5 {Andibenins E Citrinin 1:; Sphingofrmgins 5Trytoquivaline 5 5(A. wm’ecolor); (A. 1erre113), :_.(A fumigatus) i ( A c lava1113), . r, """""""""""" , 5 Sterigmatocysting' """""""""""""" :Penicillin E (A nidulam), """""" (A nidulans) E Asteltoxin (A. 31ella1113), § Aflatoxins (A. flaws), 5 Mevinolin (A. terrens), 5 Maltoryzine (A. niger), Monacolin (A. terreus). Figure 1-1. Representatives of mycotoxins produced by species of Aspergillus. ll involve two distinct phases. The first phase is the polymerization phase during which the precursor molecules are linked together by enzyme complexes such as polyketide synthases. The second phase is the modification phase during which the products of the polymerization phase are modified by enzymes which carry out hydroxylations, oxidative cleavages and rearrangements leading to the remarkable diversity of mycotoxins produced by Aspergillus. Although hexahydropolyprenol and penicillin may not be considered as mycotoxins, their biosynthetic pathways have been well studied (Manitto, 1981; Baldwin and Abraham, 1988), and these two secondary metabolites are typically used as examples to discuss isoprenoid mycotoxin biosynthesis and polypeptide mycotoxin biosynthesis. Biosynthetic pathways of each of the seven goups of Aspergillus mycotoxins are described with one or more examples. The best studied biosynthetic pathway is that utilized for AFBr biosynthesis which is discussed separately. Biosynthesis Of Isoprenoid Mycotoxins Secondary metabolites containing a carbon backbone comprised of five-carbon isoprene [CH2C(CH3)CHCH2), 2-methylbutadiene] units are called terpenes. If the carbon backbone of a terpene is altered by addition or loss of carbon atoms, or by other modifications, such a modified compound is called an isoprenoid. Isopentenyl pyrophosphate. All isoprenoids originate from isopentenyl pyrophosphate which is formed fiom acetyl CoA through an important intermediate mevalonic acid (Figure 1-2). 12 cm H OH HJC —C0A _, mow 2 / O—P—O—P—O OH II ll 0 O 3 Acetyl CoA Mevalonic acid Isopentenyl pyrophosphate Figure 1-2. Biosynthesis of isopentenyl pyrophosphate fi'om acetyl CoA. >5 0 o n o-r'i-o-i'i-ori —. \ O" n-3 Isopentenyl pyrophosphate Hexahydropolyprenols Figure 1-3. Biosynthesis of hexahydropolyprenols in Aspergillusfimigatus. 13 Hexahydropolyprenols. A large goup of isoprenoids are the polyisoprenes which are made up of more than 10 isoprene units, bound head to tail. Polyisoprenes which exist in the alcohol form are called polyprenols. Among the polyprenols, hexahydropolyprenols were isolated fi'om Aspergillus fumigams and their synthetic pathway (Figure 1-3) was described by Manitto (1981). Biosynthesis Of Amino Acid-Isoprenoid Mycotoxins Most isoprenoids produced by Aspergr'llus species are not mycotoxins, but many amino acid-isoprenoids are mycotoxins. A large goup of tremorgenic mycotoxins, such as aflatrem and firmitremorgin, are isoprenoids with an irrdole nucleus provided by tryptophan. Aflatrem. Afiatrem was the first mycotoxin with tremorgenic activity isolated from the mycelium and the sclerotium of a strain of A. W by Wilson and Wilson (1964) and its structure was then elucidated by Gallagher and Wilson (1978). Later, other aflatrem- related tremorgenic compounds from Aspergillus were isolated, such as paspalinine (Gallagher et al., 1980), aflavarin, B-aflatrem (T ePaske et a1. , 1992), sulpinine, secopenitrern B, and aflatrem B (Laakso e1al., 1992). Indeed, paspalinine, which may be a precursor of aflatrem, was isolated fiom an aflatrem-producing isolate of A. flaws along with the very unusual indole-isopentenyl pyrophosphate metabolites aflavinine and dihydroxyaflavinine (Cole e1 111., 1981). The biosynthetic pathway of aflatrem in A. flaws was proposed by Steyn and Vleggaar (1985). Aflatrem is formed fi'om tryptophan, l4 Tryptophan Paspalinine o o Ho-i'i-o-ij-o 5 on on W my. BAcetyl 00A H O O W-fiQ-g-Gfi \ o o Paspaline 0H 0H 0 o I yl Mega ”WWW“ on or om momma/l o o u 1 0-1 Naif-oi- Mrrr .. °" °" 2 Isopenten pyrophosm 2 Dimethylally pyrophosphate Y' o L 12 Acetyl CoA J Aflatrem Figure. 1-4. Biosynthesis of aflatrem in Aspergillusflaws 15 aegis Funltrernorg'm C o 3 Dimsthylally pyrophosphate Fmitramorgin A Tryptophan+Prolina M-A—o-B-ou .— SAeetleoA 3Wmflmm Figure 1-5. Biosynthesis of firmitremorgin A in Aspergillusfimigatus. 7 Malonyl-COA + Acetyl-00A / cu m- @2233 ”a o o c on c on Polyketide (01s) Emodin-S-anthrone on m o 0 WI?“ ”W“ ”etc "’c'° ° on "ac-O 0 0H on 0 0H Sulochrin Questin Emodin Figure 1-6. Biosynthesis of sulochrin in quergillus 1erre113. l6 geranylgeranyl pyrophosphate, and isopentenyl perphosphate with the loss of a methyl goup during the formation of the polycyclic structure (Figure 1-4). Fumitremorgin. During a survey of toxigenic food-borne firngi in Japan, Yamazaki et a1. (1971) found metabolites fi'om certain strains of Aspergr'llus fiam‘gatus which caused vigorous tremor and convulsions in experimental animals. Two indole-containing metabolites, desigrated firmitremorgin A and B, were isolated fiom extracts of this firngus. These tremorgenic metabolites are a family of compounds derived fi'om diketopiperazine. Based on feeding experiments in A. fianigatus with [“Cl-labeled tryptophan, proline and mevalonic acid, Yamazaki e1 11!. (1980) proposed that fumitremorgin C is synthesized from two amino acids, tryptophan and proline, to which a C5 unit from dimethylally pyrophosphate is added. Funritremorgin A is synthesized by adding two more C, units fi'om dimethylallyl pyrophosphate (Figure 1-5). Their discovery and structural characterization have been concisely reviewed by Yamazaki (1980). A derivative of fumitremorgin C was also found in A. fimrigatus (Abraham and Arfinann, 1990). Biosynthesis Of Polyketide Mycotoxins Polyketides may represent the largest goup of secondary metabolites (Weiss and Edward, 1980). These structurally diverse compounds typically contain a carbon backbone with oxygen atoms at alternate positions. The name ‘polyketide’ was coined more than 100 years ago by Collie and Myers (1893). They imagined that poly-B-keto compounds could be produced by treatment of polyacetyl compounds with weak alkali. Biochemical support for this idea was provided by Birch and Donovan (1953). According 17 to the isotopic labeling pattern of several fungal metabolites, they proposed that polyketides must be formed fiom acetic and malonic acids by a process similar to the biosynthesis of fatty acids. Polyketide chain gowth, however, is different fiom fatty acid biosynthesis, because it lacks the faithfirl removal of each B-keto goup by a process necessary in fatty acid biosynthesis. This hypothesis to explain the mechanism of polyketide synthesis was further supported by the application of the isotopic labeling method, and later by the development of sophisticated nuclear magretic resonance spectroscopic techniques (Simpson, 1987; Vederas, 1987). Knowledge of the enzyrnology of the polyketide synthase (PKS) enzymes was obtained very “slowly prior to 1985 because of the dificulty encountered in enzyme assay and purification. Only three PKS’s were purified by 1985: 6-methylsalicylic acid synthase (6- MSAS) from the firngus Penicilliun patulum (Dimroth et at, 1970), naringenin chalcone synthase (NCS) fi'om the parsley plant Petroselinum horren3e (Kreuzaler et al. , 1979), and resveratrol (stilbene) synthase from the peanut plant Arachis hjpohaea (Schoppner and Kindl, 1984). Studies on these three enzymes established the basic characteristics of PKS, even though they displayed distinctly difl‘erent properties. The synthase of 6-MSA is a large (800 kDa), tetrarneric multifirnctional protein to which all the substrates are covalently attached, whereas NCS is a homodirner composed of 42 kDa subunits that acts on the CoA esters of the substrates and lacks a firnctional equivalent to an acyl carrier protein (ACP). From 1986 until late 1996, several other PKS’s or their components have been identified and/or purified. The multienzyme of the erythromycin-producing polyketide synthase, DEBSl, DEBS2 and DEB S3 (6-deoxyerythronolide B synthase) 18 were identified and purified from Saccharopolyspora erythraea (Cafl‘rey et aI. , 1992). A proposed component of the dimeric polyketide synthase (actinorhodin acyl carrier protein- dependent malonyltransferase) for the antibiotic actinorhodin was purified from Streptomyces coelicolor A3(2) (Revill et al., 1995). With advanced molecular biology techniques, other PKS’s or their components were expressed fi'om cDNAs, and then purified. Acyl carrier proteins (ACPs) of the polyketide synthases for the aromatic antibiotics actinorhodin, ganaticin, frenolicin and oxytetracycline were expressed in and purified fi’om Escherichia coli (Crosby et a1. , 1995). The biosyntheses of sulchrin, patulin, citrinin, sterignatocystin, sterignatin, and autocystin in some species of Aspergillus have been well studied and are taken as examples to discuss the charateristics of polyketide biosynthesis. Sulochrin. Sulochrin is a derivative of a goup of mycotoxins called ergochromes which were first isolated fi'om ergot, a name for the sclerotium of Claviceps purpurea. Ergochrome BB was isolated from Aspergillus ochraceus, and ergochrome AC and ergochrome BC fi'om Amergr‘llus aculea1113. All of the known ergochromes are dimers of the monoxanthones which are synthesized fi'om polyketide precursors. The structure and the possible biosynthetic pathway of sulochrin (Figure 1-6) were described by Franck (1980). Patulin. Patulin and penicillic acid are synthesized primarily by Penicillium and Amergillus species. The biosyntheses of these two secondary metabolites are very similar. Only the biosynthesis of patulin is discussed in this section. Patulin was first discovered by Birkinshaw et a1. (1943) and its chemical structure was identified by Woodward and Singh 19 (1949). The toxicity of patulin has led to an increased awareness of its occurrence in fresh apple juice which originates from brown rot of apples contaminated with Penicillium mm Link. The production of patulin by Aspergillus clavatus gowing on spent malted barley was implicated in the poisoning of cattle when this material was used as a feed additive. Birch e1 11!. (1955) demonstrated the biosynthetic pathway of 6- methylsalicylic acid (6-MSA) in experiments with [1-“C]-labeled acetate fed to Penicilliran pa1ulr1m. 6-MSA is converted to patulin by a complex sequence of oxidation, cleavage and rearrangements which form the second stage in the biosynthesis of patulin. Zamir (1980) confirmed that 6-MSA is converted to gentisaldehyde, then phyllostine which is the substrate for the oxidative cleavage reaction and the rearrangement which results in the formation of patulin. The major steps are outlined in Figure 1-7. The biosynthesis of patulin is a clear example of the two stages in the production of a secondary metabolite. The polymerization of three molecules of malonyl CoA with a molecule of acetyl CoA, during which a reduction and dehydration occur, leads to a tetraketide derivative which readily cyclizes to form 6-methylsalicylic acid (6-MSA). Although a relatively small molecule, 6-MSA has a special place in the history of our understanding of polyketide biosynthesis. Citrinin. Citrinin, first isolated fi'om Penicillin»: citrinum by Hetherington and Raistrick (1931), is produced by numerous Penicilliwn and Aspergillus species (Rodig er a]. , 1966). Citrinin exhibited marked antibiotic activity in vitro, but its nephrotoxigenic properties prevented therapeutic application (Krogh et al. , 1973 ). The biosynthesis of citrinin was determined initially by Birch et al. (1958). Addition of sodium [1-“C]-labeled 20 0'1 0 3MalonyCoA Ho 20-1 0“ °°=” ° : . -—> .__..o MW -—- CPD 0'1 on 0 H0420 Acetyl G-MSA Gentisaldehyde Phylloetlne Patulln Figure 1-7. Biosynthesis of patulin in Penicilliwn patr1111m. Pentaketlde ‘ W COA 3 Methionlne Cltrlnln o on 0 on meteoA+4MannvICOA-~ dim mam “mm o C 0 o o 3 ea; J4 arouncoorr Figure 1-8. Biosynthesis of citrinin in Aspergillus candidus. Starlgmatocyatln 0 H: a o coca / AcatleoA 0 HO OH + —.—D —->—. b OMannyCoA c on c on \b 4 I on on 0 on on 0 0H AustocystinF SWM" Figure 1-9. Biosynthesis of sterignatocystin, sterignatin, and austocystin in Aspergillus versicolor. 21 acetate to cultures of Aspergillus candidus gave ["C]-citrinin. Gererally, during the building of a polyketide chain, a number of additional goups can be added, especially to the reactive mythylene goups, providing another source of diversity in structure. In the biosynthesis of citrinin, three additional C1 goups are added to the pentaketide chain from the biochemically active one carbon pool involving methionine (Figure 1-8). The common occurrence of C-methyl goups in polyketide-derived metabolites produced by firngi is often provided by malonyl CoA. These additional C1 goups are frequently further oxidized. Thus, one of the three additional branched carbons in citrinin is oxidized to a carboxylic acid goup. Sterigmatocystin, sterigmatin, and autocystin. Sterignatocystin (Figure 1-9), a xanthone which contains an angularly firsed bisdihydrofuran system, was originally isolated from the nrycelium of Aspergillus versicolor Tiraboschi and proved to be hepatocarcinogenic in rats (Hatsuda and Kuyama, 1954). Sterignatocystin is an intermediate in the biosynthetic pathway leading to the aflatoxins. Sterignatocystin is synthesized from a decaketide, which is probably derived from hexanoyl CoA with the subsequent addition of seven more malonyl CoA units (Townsend et aL, 1984). With the development of an understanding of the molecular biology of aflatoxin biosynthesis,‘ several genes and enzymes involved in sterignatosystin synthesis have been identified (Skory e1 11]., 1992; Keller e1 11]., 1994; Yu and Leonard, 1995; Keller et 01., 1995). Sterignatin (Figure 1-9), a metabolite isolated from A. versicolor, was the first metabolite found with a linearly fused xanthone and bisdihydrofirran moiety (Harnasaki et at, 1973). Steyn and Vleggaar (197 5) obtained other linear xanthones from corn cultures 22 of A. 1131113. The linear xanthones, called austocystins, fi'equently contain a chlorine atom, or an isopentyl side chain. They are essentially substituted derivatives of sterignatin produced (along with sterignatocystin) by A3perg1'II113 versicolor. They are believed to share the early pathway of aflatoxin biosynthesis (Harnasaki e1 (11., 1973). A key intermediate in this pathway is the postulated benzophyenone carboxylic acid (BCA) (Figure 1-9). Rotation about bond (11) would lead to the sterignatocystins, whereas no rotation or rotation about bond (b) would lead to sterignatin and the austocystins. Biosynthesis Of Amino Acid-Polyketide Mycotoxins This goup of mycotoxins is synthesized first by forming a polyketide backbone to which amino acids are added. Ochratoxin and cytochalasin B are two examples of amino acid-polyketide mycotoxins produced by Aspergillus. Ochratoxin. Ochratoxin is a nephrotoxic metabolite of some species of Aspergillus. Ochratoxin A is also a weak antibiotic (Heller e1 111., 1975). The discovery of toxigenic strains of Aspergillus ochraceus (Scott, 1965) led to the isolation and structural elucidation of ochratoxin A and B. The biosynthesis of ochratoxin A in Aspergillus ochraceus was studied in feeding experiments by Seary e1 al. (1969) with ["C]-labeled phenylalanine, by Steyn et a1. (1970) with [“C]-labeled acetate and methionine, by Wei et a]. (1971) with [“Cl]-labeled sodium chloride, and by Maebayashi et a1. (1972) with [13C]-labeled forrnate. These studies made it clear that the biosynthesis of ochratoxin A (Figure 1-10) consists of the generation of a pentaketide which is modified by the addition 23 Methionine muonyICoA Mofiggflo mmTwmn me Ochratoxin A Figure 1-10. Biosynthesis of ochratoxin in Aspergillus ochraceus. Phenyl- \ alanine o 3 Methionine 5:0 0 O O o ! €0.13 0 o —. ——.——. Oetakatide k Cytochalasin E Acetyl CoA 7 Malonyl CoA «j- 7 Acetyl CoA 7 Biotin-COOH Figure 1-11. Biosynthesis of cytochalasin E in Aspergillus clava1113. 24 of a single methyl goup fi'om the C1 pooL This additional methyl goup is oxidized to a carboxyl goup through which L-phenylalanine is linked by an amide bond. Cytochalasin E. The discovery of cytochalasans was the result of the observation that the culture filtrate of certain microorganisms produced morphological changes in hyphal cells of test firngi and unusual efi‘ects in mammalian cells in tissue culture (Rothweiler and Tamm, 1966). Cytochalasin B, one of more than 20 cytochalasans, was found in certain species of Asperillus (Buchi et a1., 1973). Cytochalasans are characterized by a highly substituted perhydroisoindolone goup, to which is fused a macrocyclic ring (which is either a carbocyclic, a lactone, or a cyclic carbonate). The structure of cytochalasin B was determined by Buchi et al. (1973). A detailed account of the biosynthetic pathway leading to cytochalasans was provided by Tarnm (1980). Here, cytochalasin E produced by A. clava1113 (Buchi e1 01., 1973) is utilized as an example to describe a possible biosynthetic pathway for cytochalasans. The proposed pathway of cytochalasin B synthesis starts with the formation of an octaketide which is produced fi'om one molecule of acetyl CoA and 7 molecules of malonyl CoA, followed by addition of a phenylalanine to form an amino acid- polyketide complex to which three C1 goup fiom methionine are added. The synthesis is terminated by formation of an epoxide (Figure 1-1 1). Biosynthesis Of Polypeptide Mycotoxins Anrino acids are the precursors of several distinct families of secondary metabolites including polypeptides, cyclic polypeptides, amino acid-polyketides, and amino acid- isoprerroids. The last two goups have already been described. Some mycotoxins 25 produced by Aspergillus are synthesized using one or two amino acids as building blocks. Coumarin and related compounds are the derivatives of one moleclue of L-phenylalanine or one molecule of L-tyrosine in several species of Aspergillus. Aspergillic acid is synthesized fi'om one molecule of leucine and one molecule of isoleucine in Aspergillus flaws. Many amino acid-derived mycotoxins or antibiotics are synthesized from more than two anrino acids such as dicoumarol, gliotoxin, tryptoquivaline M, and penicillin. Dicoumarol. During fermentation of hay, Aspergillus fianigatms transforms coumarin into dicoumarol which is a powerful blood anticoagulant and can cause fatal hemorrhage in cattle eating the spoiled bay. The immediate precursor of dicoumarol is 4- hydroxycoumarin which is derived from either L-phenylalanine or 1..-tyrosine. The methylene goup which connects the two 4-hydroxycoumarins is derived fiom formaldehyde which is also produced during fermentation. The possible biosynthetic pathway of dicoumarol in Aspergillusfimigatus (Luckner, 1990) is shown in Figure 1-12. Gliotoxin. Gliotoxin was originally described as a metabolite with antifirngal activity. The producing organism was subsequently identified as Gliocladium fimbriatwn Gilman & Abbott (Webster and Lomas, 1964). Gliotoxin is also produced by several species of Penicillium and Aspergillus such as A. chevalieri (Wilkinson and Spilsbury, 1965). Gliotoxin belongs to a large goup of toxins called epipolythiodioxopeperazines with phenylalanine, tryptophan, or tyrosine and sometimes alanine as their precursors, and with methionine or cysteine as their sulfur suppliers. Gliotoxin causes genomic DNA degadation preferentially in certain blood cell types including T lymphocytes and 26 KW“ m.“ L-Tyrosine L-Phenyla\lanine on can -— \ng p-Hydroxymelilotic acid acoumaric acid Melilotic acid OH Formaldehyde 1° B-Ketomelilotic acid 4—Hydroxycoumarin Dicoumarol Figure 1-12. Biosynthesis of dicourrrarol in Aspergillusflmigatus. H o C1420"! °Hs H2014 CHzG'I HOOCY‘JH; CHzO-l Glrotoxrn Sarita Figure 1-13. Biosynthesis of gliotoxin in T richodemra viridi. 27 macrophages. Gliotoxin has previously been used to treat murine allogeneic bone marrow prior to transplantation into irradiated recipients, and in this situation the drug prevents development of gaft-versus-host disease. This goup of toxins includes hyalodendrin, aranotin, epicorazine, sporidesnrin, chactocin, verticillin, chetomin, sirodesrnin, and a large number of their derivatives. An understanding of the gliotoxin biosynthetic pathway has been partially achieved (Figure 1-13). Suhadolnik and Chenoweth (1958) were the first to demonstrate that phenylalanine provides the reduced indole nucleus of gliotoxin in T richodenna viridi. Almost 20 years later, Kirby et a]. (197 8) confirmed that cyclo-(Irphenylalanyl-L-seryl) is an intermediate in gliotoxin biosynthesis in T richoderma viridi. For the biosynthetic mechanism for introduction of sulfirr into gliotoxin, no firm conclusions have been reached. Cysteine ( possibly in combination with pyridoxal) is suggested for the sulfirr donor, and a dehydrodioxopiperazine is suggested for the sulfur acceptor. Consecutive introduction of two thiol goups would be suficient for oxidative cyclization to an epidisulfide compound (Kirby and Robins, 1980). The biosynthesis of gliotoxin is dealt with in some detail by Kirby and Robins (1980). Gliotoxin was the first mold metabolite to be recognized as belonging to the diketopiperazines with polysulfirr bridges. Tryptoquivaline M. Two tremor-causing metabolites, tryptoquivaline and tryptoquivalone, were isolated from a sample of rice implicated in the death of a child in Thailand, and then demonstrated to be produced in a toxigenic strain of Aspergillus clavatus by Clardy e1 11]. (1975). Later other tryptoquivaline-related metabolites were isolated from Aspergillus fi1m1'gatr13 (Buchi e1 11!. , 1977; Yamamki et a1. , 1979). 28 "2N *1 U “ac-CHOW! HOW Vuln- Maw “'6 OIK. 03:5,. cacao . Mr 0 p o p o p (1?” ”firm" 0&1; 0%; 012:?“ O 7W Alanhe Deoxynortryptovalne Tryptoqulvalone TWIN!“ M Figure 1-14. Biosynthesis of tryptoquivaline M in A3perg1‘llusfi1m1’ga1113. “WW L-arphuminoadiprc acid L-valine We“ ach L-cystelne Phenylacetyl CoA Mmfig” IpnA an’sf $0500Yfi9< LLD-ACV lsopenicillin N Penicillin 6 Figure 1-15. Biosynthesis of penicillin in Aspergillus nidulans. 29 The basic precursors involved in biosynthesis of these complex molecules are probably the amino acids tryptophan, valine, alanine and anthranilic acid, the latter being also an intermediate in the shikimate pathway to tryptophan. In this section tryptoquivaline M is utilized as an example to describe the possible pathway of tryptoquivaline biosynthesis in A3pergillr13fi1m1‘ga1113 (Y amazaki et al. , 1979). Synthesis appears to be initiated to form a tripeptide composed of anthranilic acid, tryptophan, and valine. Deoxynortryptoquivalone may be the first metabolite formed in the pathway of tryptoquivaline by addition of another amino acid alanine. The oxidation of dexoynortryptoquivalone results in the formation of tryptoquivalone which is converted to nortryptoquivaline. Nortryptoquivaline is then converted to tryptoquivaline M by simple epirnerization (Figure 1-14). Penicillin. The discovery of penicillin has a special place in the history of human medicine. In September 1928, Alexander Fleming, a Scottish bacteriologist working in Almroth Wright’s Inoculation Department at St. Mary’s Hospital, London, returned from holiday to his laboratory and found a contamination of mold (Penicillium n01a111m) in a plate culture of staphylococci. The gowth of the bacterial colony was inhibited around the mold. The inhibition was later shown to be caused by the production of a substance by the mold to which Fleming gave the name “penicillin”. His discovery of penicillin was published in 1929. Penicillin belongs to a goup of antibiotics with a common B—lactam ring. Today, 69 years later, the understanding of penicillin (and other B-lactam compounds) biosynthesis has been reached not only at the chemical and biochemical level, but also at the genetic and molecular levels. The main interest is with Penicillium chrysogeman and Acremonium chrysogenum which have been the commercial sources of 30 penicillin and other B-lactams. But in the late 1940s, screening of a wide range of fungi for new antibiotics led to the detection of penicillin produced by Aspergillus nidulans (Dulaney, 1947). Although this has never been of commercial sigrificance, A. nidulam has been utilized as a model organism for genetic and molecular studies of penicillin biosynthesis. The biosynthetic pathway for penicillin G in A. nidulans fi'om amino acid precursors is shown in Figure 1-15. The primary metabolites which are the starting point for penicillin biosynthesis are the three amino acids L-or-aminoadipic acid, L-cysteine, and 1..-valine. The first step is the formation of the tripeptide 5-(L-a-aminoadipyl)-L-cysteinyl-D—valine (ACV) catalyzed by the single large enzyme ACV synthetase (ACVS). The tripeptide is cyclized by is0penicillin N synthetase (IPNS), also called ACV cyclase, in an oxidative reaction in which four hydrogen atoms are removed fi'om ACV, and one molecule of oxygen is consumed (Baldwin and Abraham, 1988). Isopenicillin N is modified by the enzyme acyltransferase (acyl CoA:6-amino-penicillanic acid acyltransferase, ACT). This enzyme removes the aminoadipyl side chain, and replaces it by phenoxyacetyl to form penicillin G. Streptonwces clabvuligerus contains isopenicillin epirnerase which converts isopenicillin to penicillin N. Certain fungi and many bacterial species further modify the lactam ring by 5 the sequential action of expandase and hydroxylase activities to form decacetylcephalosporin C. This is converted into either cephalosporins or cephamycins, depending on the produing organism. 31 Delta-Ha-aminoadipylylfcysteinyl-D-valine synthetase (ACVS) was first isolated fi'om Aspergillus nidulans and studied by Van Liempt et al. (1989), and further analyzed by MacCabe et al. (1991). Isopenicillin synthetase (IPNS) was first isolated and studied by Baldwin and Abraham (1988) and was further studied by Roach et a1. (1995) and Blackburn et al. (1995). Acyl CoAz6-amino-penicillanic acid acyltransferase (ACT) was purified fi'om A. nidulans (Whiteman e1 111., 1990; Montenego e1 11]., 1990). The first B-lactam biosynthetic gene to be cloned was the isopenicillin synthetase (IPNS) gene of Amergillus chorsogenum. This gene was identified using oligonucleotides based on a partial sequence of the purified enzyme (Samson e1 111., 1985). Availability of this gene quickly led to the isolation of the IPNS gene, 1'pnA, of A. nidulans (Ramon er al. , 1987) by heterologous hybridization. The discovery that a cephamycin biosynthetic gene cluster from the Gram-negative bacterium, F lavobacterium spp. SCI and 154, hybridized to the IPNS gene of P. chrysogenum, enabled the identification of the region containing the 11ch gene encoding the 5-(L-or-arninoadipyl)—L-cysteinyl-D-valine synthetase (ACVS) inA. 1111111111113 (Bumham e1 111., 1989). Later the ach gene was positively identified in A. nidulans by MacCabe et a1. (1991). The isolation of the penDE gene encoding acyl CoAz6-amino-penicillanic acid acyltransferase (ACT) was initially achieved in P. chrysogenum (Barredo et aL, 1989), using oligonucleotides desigred fiom the N-terminal amino acid sequence of the purified enzyme. The equivalent gene, acyA, of A. nidulans was isolated using the P. chrysogenum penDE gene as a probe (Montenego et al. , 1990). The Aspergillu3 nidulans rrpeA locus was identified by MacCabe et a1. (1990). The locus consists of three contiguous genes in the order of ach-ipnA-acyA and is required for 32 penicillin biosynthesis. These three genes, which are clustered in chromosome V1 (3.0 Mb), were cloned fi'om A. nidulans. Beach gene is expressed as a single transcript from a separate promoter (Martin and Gutierrez, 1995). Biosynthesis Of Isoprenoid-Polyketide Mycotoxins Some mycotoxins isolated fiom Aspergillus contain both isoprenoid and polyketide moieties. Turner and Aldridge (1983) listed a number of isoprenoid-polyketide mycotoxins produced by Aspergillus. Austin, firmagillin, and territrem B belong to this goup of mycotoxins. Austin. Austin, a polyketide-isoprenoid mycotoxin, was first isolated fi'om Aspergillus 1131113 by Cherral et a]. (1976). Austin was later found in A. 1erre113 by Springer et a1. (1979). The biosynthetic pathway of austin in Aspergillus 1131113 was initially studied by Wicnienski (1979) and proposed by Simpson er al. (1982). The major step in the biosynthesis is the preliminary coupling of the sesquiterpene precursor farnesyl pyrophosphate and the methylated polyketide. The three methyl goups come from methionine. Rearrangement of this hypothetical intermediate leads to the formation of austin (Figure 1-16). . Fumagilliu. Furnagillin, first isolated from Aspergillus fi1m1'gatr13, was recogrized as an antibiotic agent (Hoza, 1966). At high doses firmagillin was lethal and caused toxic alteration in the liver, the kidney (Lauren et a1. , 1989), and the respiratory epithelium (Anritani e1 11]., 1995). Fumagillin is a potent anti-angiogenic compound (I-Iorsman at al, 1995; Nishimura e1 111., 1996). Fumagillin is another example of a secondary metabolite 33 MGM 3M8bnleoAc—3Bloth-COOH'I'3AOCMCOA Figure 1-17. Biosynthesis of fumagillin in Aspergillusfimigams. 34 assembled fiom two distinct pools of precursor intermediates, one is polyketide and the other is isoprenoid. Birch and Hussain (1969) showed that in A. firmigatus the decantetraendioic acid moiety, which is one of the two precursors of firmagillin, has a polyketide origin and that the terminal carboxyl goup represents the original carboxyl end of the polyketide. They proposed an hypothesis for the origin of the unusual nucleus of fumagillin from farnesyl pyrophosphate via a'bergamotene intermediate. This hypothesis was later supported by the detailed studies of Cane and Levin (1976) on the biosynthesis of ovalicin of Pseudeurotium ovalis Stolk. Fumagillin is an intermediate in the ovalicin biosynthetic pathway. The proposed biosynthetic pathway of firmagillin is shown in Figure 1-17. Territrem B. Territrem A and territrem B were first identified in stored unhulled rice contaminated with Aspergillus ten-e113 in Taiwan by Chung et a1. (1971). Ling (1976) demonstrated that both toxins induce tremor and convulsions in mice. Territrems do not contain nitrogen. This is the most distinctive feature of territrems which sets them apart fi'om the known N-containing tremorgenic mycotoxins. Other members of this family of compounds have been identified and characterized by Ling er a]. (1984), Peng et a]. (1985), and Ling (1994). For example, territrem C was isolated fi'om the chloroform extract of rice cultures of Aspergillus terreus 23-1, which also produces territrems A and B. Ling et a1. (1984) also studied acute toxicity and some physicochernical properties of territrems. Territrems contain isoprenoid and polyketide moieties. The biosynthetic pathways of both territrem A and territrem B have been studied in Aspergillus terreus 35 OO Fameeyl pyrophosphate o 45-0-1"; -oa 0 on on Isopentenyl pyrophosphate W0 0' 9 o 41-04-011 5" 5H Hexaketide Gennyl pyrophosphate 9 9 > , 95, 9 -P-O-P-O-1 -o-r.>-ou 6" on Isopentenyl pyrophosphate Dlmathylally pyrophosphate L W9 Figure 1-18. Biosynthesis of territrem B in Aspergillu3 1erre113. O I HN o ; 0'1 cc?“ ° \ °° 49-011 a. Dimethylallypyrophosphate """°""" - / 9 9 H 7 f-o-g-ou amateur _. 0401-1 Cycloplazonic acid Isopentenyl pyrophosphate ' Figure 1-19. Biosynthesis of cyclopiazonic acid in some species of Aspergillus. 36 (Kuo and Yun, 1988; Lee et aL, 1992). The major step in the biosynthesis of territrem B is the combination of one molecule of hexaketide and one molecule of farnesyl perphosphate. Famesyl pyrophosphate is formed from one molecule of isopentenyl pyrophosphate and one molecule of dimethylally pyrophosphate (Figure 1-18). Biosynthesis Of Amino Acid-Isoprenoid-Polyketide Mycotoxins This goup of mycotoxins contains all the three basic precursors: amino acid, polyketide, and isoprenoid components. Cyclopiazonic acid belongs to this goup of mycotoxins. Cyclopiazonic acid. Cyclopiazonic acid is produced by strains of Aspergillus flaws, A. oryzae and A. versicolor, as well as several species of Penicillium (Luk et a]. , 1977; Gallagher e1 (11., 1978; Richard and Gallagher, 1979; Dorner, 1983; Domer et al., 1984; Trucksess e1 01., 1987; Lee and Hagler, 1991). The biosynthesis of cyclopiazonic acid was reviewed by Holzapfel (1980). The molecule is synthesized in some species of Aspergillu3 (Figure 1-19) possibly from tryptophan and an acetyl CoA-derived diketide to yield or-acetyl y—(B-indolyl) methyltetramine acid to which is added a C5 dimethylallyl goup with subsequent cyclization to yield cyclopiazonic acid. Because the C5 dimethyallyl goup is derived fi'om isopentenyl pyrophosphate, cyclopiazonic acid is composed of the three basic precursors: tryptophan an amino acid, a deketide, and an isopentenyl pyrophosphate. Two of the enzymes involved in the later steps of this pathway were isolated and partially characterized (Steenkamp et a1, 1973) and the stereochenristry of the cyclization was thoroughly studied (Steyn e1 111., 197 5a). 37 Epilos The biosynthesis of mycotoxins in Aspergillus introduced in this review represents a small piece of the whole picture of the enormous chemical diversity and the complex biosyntheses of mycotoxins of this genus. In addition to the challenges still ofi‘ered by the mycotoxins of Aqrergillus to chemists and biochernists, their potential biological activity including mammalian toxicity also challenge the medical and toxicological fields. One important challenge now is to understand the molecular biology of the biosyntheses of this goup of mycotoxins. PART 2. AFLATOXINS TOXICITYOFAFLATOXINS AmmamsewndmynwtabohtessynfledmdbycatmnMOqurergIhufhmA paun'ticus, andA. 1101111113(Gormanetal., 1992;Kurtzmanetal., 1987). Thesefirngiare ubiqrutwsandfiequanlygowonavafiayoffoodmdfeedaopsandcauseaflatordn contaminatoninmanyareasintheworld (Blourrt, 1961;]elineke1al, 1989;Ellise1 111., 1991). Peands,cottonseed,comandteermtsarethemajorcropsafi‘ectedinfl1eUSA. AfiatordnsarebasicaflyclassifiedhrtotheBoereriesbasedontheircoloroffiuorescence under Iongwave (360 nm) UV light (B for bhre, and G for geen). Afiatoxin Br (AFBr), afiatoxin B2 (AFBz), aflatoxin Gr (AF Gr), and afiatordn G2 (AFGz) are the four major afiatordnsproducedbyfirngiinnatm'e. ItislikclythataflatordcosiserdstedformanyyearspriortotheepizooficoutbreakinBritain inl960knownasTurkedeisease(Blount,1961)inwhichmorethan100,000turkeyswere 38 destroyed with acute hepatotoxicity caused by afiatoxin-contaminated feed. However, that dramaficornbreakdemonsu'atedtheseriousnessofafiatordncomanfinafion, andledtothe recogrition that afiatoxin contamination is both an economic and a public health problem in my areas of the world. Asobsavedinfieldoutbreaks,acutestructuralandfirnctional damagetotheliver(suchas fatty infiltration, necrosis, and extensive bile duct proliferation), the major target organ for afiatothhavebewreponedmmoahbomtorymfimalsmdmsevaaldomesficanhnal species(BusbyandWogan, 1984). Humanexposuretoaflatordnoccurreddirectlythrough ingestion ofcontarninated crops and resulted in fatal aflatoxicosis in India (Krishnamachari et at, 1975) and in West Afiica (Ngindu er al., 1982). Inaddifionmaanehepatotorddty,afiatordmmesuongnnnagmsmdcarcmogeus Aflatoxins have been demonstrated to be potent mutagens in a variety of test systems causing difi‘erent mutations. For example, aflatoxins have been demonstrated to cause mutations in: bacteriophage (Goze et al., 1975) and plasmids (Wood et al., 1987; Courtemanche and Anderson, 1994); bacteria (Raina e1 (11., 1980; Lowery et at, 1983; Refolo e1 01., 1987; Foster et at, 1988); Drosophila melanogaster (Lamb and Lilly, 1971; Graf e1 (11., 1984); cotton leaf-worm (Abdou e1 111., 1984); the chick embryo (Bloom, 1984); monkeys (Adgigitov e1 11]., 1984); mice (Umeda et at, 1977;.Macgegor, 1979); and human cells (Kaden et al., 1987). The types of mutations linked to aflatoxin exposure include substitutions, fi'arneshifts, sister chromatid exchanges, chromosome aberrations, DNA single-strand breaks, nricronucleus formation, and lethal mutations. 39 The experimmtal data, which have been obtained from different animals ingesting AFBl such as rat, rainbowtrout, duck, rhesus monkey and tree shrew, demonstrate that AFBl is the most potent naturally occurring carcinogen (Adarnson e1 11]., 1973; Carnaghan, 1965; Gopalan et at, 1972; Reddy e1 11]., 1976; Sinnhuber e1 01., 1968; Tilak, 1975) in these experimental systems. For example, early experiments with laboratory rats demonstrated that AFBl is a renal carcinogen (Epstein et al., 1969). A small incidence of colon carcinomas resulted when AFBr wasadmhusuatedtochha'mtseitheromflyorviaflredietMogmmdNewbene, 1967). Administration of AFB1 elicited tumors in the central and peripheral nervous systems of pregnant rats (Goerttler et al., 1980). Sieber et at (1979) found that long term exposure of AFBr to monkeys induced hepatocelluar carcinomas, osteogerric sarcomas, bladder papillary carcinoma, gall bladder adeno-carcinomas, and pancreatic carcinomas. Emerimental evidence has linked AFBl to human lung cancer (Hayes etal, 1984; Mace e1 111., 1994). Epiderniologically, AFBl also has been suggested as a synergistic factor along with hepafifisthusOiBWinthegenerafionofhumanpfimaryhvercancerQLC),andhuman hepatocellular carcinoma (HCC) (Groopman et at, 1988). Very recently it was demonstrated tint HBV and AFBl played a synergistic role in the development of hepatocellular carcinoma in tree shrews (Yan e1al., 1996). Afiatoxinscancauseavariety ofothertoxic efi‘ects. AFBl experirnentallyhasbeen shown to be a potent teratogen in rat, hamster, and chick embryo (Butler et at, 1966; Elis and DiPaolo, 1967; Venett e1 at, 1973). Afiatoxicosis altered the reproductive efliciency of both male and female domestic animals, particularly poultry (Ottinger and Doerr, 1980). Afiatoxins havepdmfialtordcefi‘easonmehmnmesystanmmhnalsremlfingmsrscepubflitym 40 diseases caused by bacteria, fungi, and viruses (Edds et a1, 1973; Richard et al., 1973; Giambrone et at, 1985). Herrold (1969) reported that AFBl caused megalocytosis in the prordmaltubulesinthekidney. Hemorrhagickidney syndromethatcomrnonlyoccursin chickensinAfiicahasbeeulinkedtoAFBl (Datiillaetal, 1987). Inwlturedlddneycelllines, AFBr induced mitochondrial degeneration and loss of microvilli (Y oneyama et at, 1987). The possible efi‘ects of AFBl on nervous system function were proposed by finding this toxin in tissue samples fi'om patients with Reye’s syndrome, a pediatric disease characterized by cerebral edema and neuronal degeneration (Chaves-Carballo er al., 1976). In rats, AFBl cmsedadeaeaseofsamomnmdhsmetabohtembrainmdalsomhibhedbrainstem tryptophan metabolism (Coulombe e1 111., 1985). The generally accepted biochemical mechanism of AFBl toxicity is its biotransformation to the AFBl-8,9 epoxide by cytochrome Pea-dependent epoxidation of the terminal firran ring double bond (Essignann e1 111., 1982; Stark, 1980; Soman and Wogan, 1993). AFBl-8,9 epoxide is a potent electrophilic molecule which can form adducts with protein and DNA. The protein adducts may result in abnormal fimction and cause acute toxicity, and DNA adducts may cause mutagenicity and carcinogenicity. In addition, formation of fi'ee radicals from AFBl suggests a possible role of fi'ee radical metabolites in the carcinogenicity (Kodarna et at, 1990). As early as the late 19705, Autrup et a]. (1979) reported that colon tissues from rat and harm were capable of activating AFBl to form DNA adducts. later it was demonstrated that AFBl-8,9 epoxide combines mainly to the N-7 of guanine to form DNA adducts (Essignarm, 41 00 on Aflatoxin 32 O O OCH3 O O OCH; Aflatoxin G2 Figure 1-20. Structures of Aflatoxin B1, B2, Gr, an G2. CHBCOZH CHsSCHzCI-IZOH(NHZ)C02H ACETATE METI-IIONINE Figure 1-21. The origin of the 16 skeletal carbon atoms of aflatoxin B1. 42 er al, 1982). These DNA adducts have been proven to be mutagenic in many in vitro mutagen-detecting systems such as bacteria, firngi, drosophila, and mammalian cells. For example, mutations in codon 61 of the Ha-ras proto-oncogene have been shown to occur with a high fi'equency in AFBr-induced liver tumors in the CF] mouse (Bauer-Hofinan e1 111., 1990). Another major mutational hot spot for DNA adduct formation by AFBl in human primary liver cancer (PLC) and human hepatocellular carcinoma (HCC) was demonstrated to be codon 249 in the p53 tumor suppresser gene (Puisieux e1 01,1991). Experimental data demonstrated that rmrtations in p53 occur later in tumor development, and therefore suggested that these mutations may act as tumor promoters not as tumor initiators (Nose et al., 1993). AFBl can also induce specific mutations which activate another proto-oncogene c-ki-ras (Soman and Wogan, 1993). Because of the threat of aflatoxin contamination to public health the US Food and Drug Administration set action levels of 20 ppb for human foods (except for milk which is 0.5 ppb) and 20to 300 ppbinmostanirnal feeds(Labuza, 1983). BIOSYNTHESIS OF AFLATOXIN Bl Biosynthesis Of Allatoxin B1 Afiatoxins are polycyclic, unsaturated compounds consisting of a coumarin mrcleus flanked byabisfirran system containing eitherafilranring(Bl, G1 series) oradihydrofirranring (82, G2 series) on one side, and either a cyclopentanone (B series), or a six-member lactone (G-series) 43 mortar! CO MabnleoA o ,5“,me ,_j_ ,...,wvm °L4__ Eagle“, TMalonleoA 7c02 o o o o . we» . \_ a we Ema/‘chs EmSJk/V‘CBJ o o o o 8.5 HexanoleoA Decaltetide ouoouou err, on 0110011011 01100110 "misfit. are... ‘3. WTW \ on c on Avarantin (AVN) Norsolorinic acid (NA) ° 0110011 no oucu / Avezufanln $1.3] , on ‘ 016". (MIN) / "° 0 on c on on c on Veniconal W1 acetate ...-(3:100... ,cec ,. Averufin (AVF) ° 1'4-lydroxyveraicolorone (HVA) 0110011 01100" ouoonv WU.— ._ "o 0° "0 0° HO oouorr ° 0 Versioolorin A (VA) Versioolorin 3 (VB) Venisonal (VAL) o c on c on 0 can. 0 o o _, o _. o o o ecu. 0 ° OCH: ° °°"' Demetlrylaterig- Sterigmatocystin O-methylsterigma- Aflatox in 81 (AF 31) matocyatin (ST) tocystrn (DMST) (OM37) Figure 1-22. The aflatoxin B1 biosynthetic pathway in Aspergillus parasiticus. on the other side (Figure 1-20). Most researchers in the field of aflatoxin biosynthesis have focused on aflatoxin Bl (AFBr) because it is the most toxic and abundant aflatoxin produced by fungi. The , currently accepted AFBl biosynthetic pathway begins with acetyl CoA and malonyl CoA to form a proposed polyketide precursor, which is then converted sequentially to several pathway intermediates in the order of norsolorinic acid (NA), averantin (AVN), averufanin (AVNN), averufin (AVF), versiconal hemiacetal acetate (VHA), versiconal (VAL), versicolorin B (VB), versicolorin A (VA), demethylsterignatocystin (DMST), sterignatocystin (ST), O-methyl sterignatocystin (OMT), and finally aflatoxin B1 (AFBr) (Figure 1-22) (Dutton 1988; Bhatnager e1 111., 1992; Yabe and Hamasaki, 1993). The established pathway is the result of a combination of the use of Aspergillus mutants blocked in the AFBl pathway in certain steps, radiolabeled-precursor feeding experiments, enzyme isolation (purification) and activity determination, and chemical structure determination by nuclear magnetic resonance (NMR) spectroscopy and mass spectroscopy. Determination of the structure (especially the radiolabeled atom distribution and the stereochemical configuration) of any potential precursor and the final product is a very important requirement. Without accurate structure determination, all the 5 other experimental results are not fillly explainable. Therefore, any comprehensive review on AFBl biosynthesis should include structure analysis. However, in this brief review, only the most important experiments related to structure determination will be mentioned. Steyn e1 al.(l980) presented an excellent review on structure determination related to AFBl biosynthesis. 45 According to the chemical structure of the starter units, the intermediates and the final product, and the demonstrated or speculated nature of the biochemical reactions involved, the biosynthesis of AFB1 can be divided into seven distinct stages: (1) the fatty acid stage in which it is proposed that a hexanoyl CoA is synthesized with one acetyl CoA and two malonyl CoA as the building units; (2) the polyketide stage in which a proposed decaketide is formed (by adding another seven malonyl CoA to the hexanoyl CoA in a manner similar to fatty acid synthesis but without the complete reduction, dehydration, and reduction cycles) and finished by formation of a stable anthraquinone norsolorinic acid; (3) the internal ketal stage in which the six-carbon side chain of norsolorinic acid is modified (through averantin, and averufanin) to form the internal ketal of averufin; (4) the dihydrobisfuran stage in which the ketal of averufin is firrther modified by oxidative reorganization (in the order of versiconal hemiacetal acetate, versiconal, versicolorin B) to form versicolorin A which contains an internal ketal structure; (5) the anthraquinone- xanthone conversion stage in which the anthraquinone versicolorin A undergoes a complex set of oxidative cleavage and deoxygenation reactions 5 to form the xanthone demethylsterignatocystin; (6) the methylation stage in which demethylsterignatocystin is modified by methylation to form sterignatocystin and in turn O-methylsterignatocystin. (7) the xanthone-coumarin conversion stage in which the xanthone structure of O- methylsterignatocystin undergoes oxidative cleavage, subsequent cyclization and loss of one carbon atom to form the coumarin nucleus of afiatoxin Br. Acetyl CoA, malonyl CoA, and C-1 of methionine are the carbon sources of AFB1. The study of the sources of the carbons in aflatoxin Br was started in the' late 1960’s. Among many studies, the experiments conducted by Biollaz et a1. (1970) established conclusively that AFBl is synthesized with acetate units as the starter. AFBl, prepared by separately feeding of [l-"C]- and [2-"C]-acetate to an A. flaws culture, was analyzed by step degadation. The origin of the 16 skeletal carbon atoms was determined (Figure 1- 21). The nearly equal distribution of the labeled carbon atoms at all the centers suggested that AFBl is derived from a single, highly rearranged polyketide chain. This hypothesis was firrther supported (Hsieh and Mateles, 1970; Hsieh and Mateles, 1971). In the study by Biollaz et al. (1970), [methyl-“C]-methionine was found to specifically label the methoxy carbon atom of AFBl. Later, the development of [13C] NMR spectroscopy, which reduced the time used in these studies, confirmed that acetate is the source of carbon for AFBl (Steyn et al., 1975b; Pachler et al.,1976; Gorst-Allman et al., 1978). The first stage was proposed to occur via a fatty acid biosynthetic pathway to form a hexanoate (Townsend et al., 1984; Brown et al., 1996). The second stage was proposed to occur through a polyketide synthetic process to form a decaketide (Lee et al., 1971; Peng and Leonard, 1995; Chang etal., 1995). The third stage is the internal ketal stage in which a proposed decaketide goes through norsolorinic acid, averantin, and averufanin to form averufin (an internal ketal). The anthraquinones norsolorinic acid (NA) (Conrsoi) (Lee et al., 1971), averantin (AVN) (Conzoo‘I) (Birkinshaw etal., 1966), and averufin (AVF) (canrror) (Donkersloot er al., 1972), are each produced by blocked mutant strains derived fiom wild-type A. parasiticus. Each compound is derived from a Car-decaketide, and was demonstrated to be an intermediate in the AFBl pathway by feeding experiments. Bennett et a1. (1980) 47 demonstrated the transformation of [“C]-AVN into AVF, but not into NA in A. parasiticr13. Therefore AVN was placed after NA and before AVF in the AFBl pathway. McCormick et a]. (1987) found AVF in wild-type A. para31'11'c113. In studies with blocked mutants they demonstrated ["Cj-radiolabeled averufanin (AVNN) accumulated in AVF but not AVN. The results demonstrated that AVNN is a precursor in the AFBl pathway afier AVN and before AVF. With improved I-IPLC, Yabe et al. (1991a) suggested that 5’-hydroxyaverantin may be an intermediate between AVN and AVNN. The fourth stage is the dihydrobisfirran stage in which averufin goes through versiconal hemiacetal acetate, versiconal, and versicolorin B (C) to form versicolorin (a dihydrobisfirran). Lin et a1. (1973) first demonstrated that averufin (AVF) was converted into AFBl by A. pwasiticus. Yao and Hsieh ( 1974) treated wild-type A. pamsiticr13 with the insecticide dichlorvos (dimethyl 2,2-dichlorovinyl phosphate) which presumably acts as a specific enzyme inhibitor, and found AFBl production was inhibited but versiconal hemiacetal acetate (VHA) accumulated. In the presence of dichlorvos, AVF was converted into VHA. Therefore AVF lies before VHA in the AFBl pathway. Several mechanisms have been proposed for the rearrangement of averufin (AVF) to versiconal hemiacetal acetate (VHA). Among them an epoxide rearrangement is consistent with experimental evidence. Nidurufin (2’-hydroxyaverufin) is a natural metabolite of Amergillus nidulans (Auch and Holzapfel, 1970). Hydroxylation at C-2 of averufin could convert averufin into nidurufin. A rearrangement of nidurufin followed by a Baeyer- Villiger oxidation (Townsend at al., 1982) was proposed to convert nidurufin into versiconal hemiacetal acetate. 48 In feeding studies without dichlorvos treatment, VHA was converted to AFBl in wild- type A. parasiticu3. In feeding studies with dichlorvos, versiconal (VAL), versicolorin B (VB), and versicolorin A (VA), (but not VHA) were converted into AFBl (Y ac and Hsieh, 1974; Fitzell et al., 1977; Hsieh et al., 1978). Therefore VHA was before VAL, ' VB and VA in the AFBl pathway. Hsieh et a1. (1989) and Anderson and Chung (1990) found that VHA was converted to versiconal (VAL) which was further converted to versicolorin C (VC) by A. parasiticus. Lin and Anderson (1992) and Silva et al. (1996) demonstrated that a versiconal cyclase purified from A. parasiticus catalyzes the dehydration of VAL to VB or versicolorin C (V C). Indeed versicolorin C (V C) is the. racemate of VB. Yabe .et al. (1991b) found that VB was converted into VA in A. pm'asiticus by a desaturase activity. Townsend et at (1988) suggested that 1’-hydroxy-versicolorone (HVN) may be an intermediate between AVF and VHA. Yabe and Hamasaki (1993) suggested that, in the AFBl pathway, the conversion of AVF to VA goes through a metabolic gid involving versiconal hemiacetal acetate, versiconol acetate, versiconol, and versiconal. In the fifih stage, versicolorin A (an anthraquinone) is converted to demethylsterignatocystin (a xanthone). Hsieh et al. (197 8) found that ["C]-labeled versicolorin A (VA) was eficiently converted to sterigmatocystin (ST) by A. versicolor. Thomas (1965) proposed that the conversion of versicolorin A to sterignatocystin could occur by oxidative decarboxylation and elimination of an acetate-derived methyl carbon atom. This hypothesis was further supported by the [13C]-NMR spectrum data of sterignatocystin and aflatoxin Bl (Pachler et al., 1976). Jeenah and Dutton (1983) found 49 that. a VA-accumulating mutant of A. parasiticus was able to convert sterignatocystin to both O-methylsterignatocystin (OMST) and AFBl suggesting that VA is an earlier precursor than ST in the AFBl pathway. Yabe et a1. (1989) found that demethylsterigmatocystin (DMST) was converted to AFBl; and in the presence of S- adenosyl-[methyl-‘lflmethionine, DMST was first converted into ST, and then to OMST in A. parasi11'c113. According to recent data, VA is the last anthroquinone and DMST is the first-xanthOne in the AFBl pathway. DMST is clearly an intermediate between VA and ST. At this stage the anthraquinone VA is converted into the xanthone DMST through a possible oxidative cleavage and deoxygenation (Y abe et al. , 1989). In the sixth stage, demethylsterignatocystin is methylated to form sterignatocystin which in turn is methylated to form O-methylsterigmatocystin. Singh and Hsieh (1976) demonstrated that A. pam3111c113 was capable of converting (“cl-sterigmatocystin (31) into AFBl. Bhatnagar er a1. (1987) found an isolate of A. parasiticus which accumulated O-methylsterignatocystin (OMST) without AFBl. When ["Cj-OMST was fed to the mycelia of amutant of A. parasiticus, ["C]-AFB1 was produced and when ["C]-ST was fed to the same mutant, [“C]-OMST was produced. Cleveland and Bhatnager (1987), and Bhatnagar and Cleveland (1988) demonstrated that, in A. parasiticu3, a postrnicrosomal activity (PMA) catalyzed the conversion of ST to OMST, and a microsomal activity (MA) catalyzed the conversion of OMST to AFBl. These results suggest that OMST is an intermediate in the AFBl pathway between ST and AFBl. In the seventh stage, O-methylsterignatocystin is converted to aflatoxin B1. In this stage, the xanthone structure of O-methylsterignatocystin undergoes oxidative cleavage, 50 subsequent cyclization and loss of one carbon atom to form the coumarin nucleus of aflatoxin Bl. This is the complete current picture of AFB1 biosynthesis which is generally accepted. Based on the experimental data that vesicolorin A (VA) was a precursor of AFB1 (not of AFB:) and versicolorin B (VB) was a precursor of both AFBl and AFB2, combined with the fact that VB was converted to VA, McGuire et al. (1989) and Yabe et al. (1991b) proposed that VB was the branch point leading both to AFBl and AFB2 in the aflatoxin biosynthesis pathway. Lee et a]. (1975) found two distinct O-methyltransferase activities in aflatoxin biosynthesis. Based on their experiments, Cleveland e1 al.(l987) and Cleveland (1989) suggested that AFBl/AFGl and AFB2/AFG2 are synthesized separately. This idea was filrther confirmed. Yabe et a1. (1989) demonstrated that A. parasiticus NIAH-26, which does not normally produce aflatoxins, was able to convert demethylsterignatocystin (DMST) to aflatoxins Br and G1 (not to B2 and G2); and to convert dihydrodemethyl- sterignatocystin (DHDMST) to aflatoxins B2 and G2 (not Br and G1). They also demonstrated that O-methyltransferase I converts DMST and DHDMST into sterigmatocystin and dihydrosterignatocystin, respectively; and O-methyltransferase II converts sterignatocystin and dihydrosterignatocystin into O-methylsterignatocystin (OMST) and dihydro-O-methylsterignatocystin (DHOMST), respectively. These data support the idea that AFBl/AFGl and AFBz/AFG2 are synthesized separately. Henderberg et a]. (1988) demonstrated that [um-sterignatocystin was converted to AFGl in wild-type A. flaws and A. parasiticus but ["C]-AFBl was not. These data 51 suggested that AFGl may not be derived from AFBl; while AFBl and AFGl may share a common pathway at least until sterignatocystin formation. The relationship between of AFBl/AFGl and AFB2/AF G2 in aflatoxin biosynthesis then may be summarized as follows: the four aflatoxins share a common pathway in their biosynthesis up to versicolorin B. After versicolorin B, one branch leads to AFBl/AFGl, the other to AFBz/AFG2. AFBl and AFGl share a common pathway up to sterignatocystin; while AFB2 and AF G2 share a common pathway up to dihydrosterignatocystin. Genes Involved In The Allatoxin Br Pathway In order to understand the regulatory mechanisms involved in aflatoxin biosynthesis, a very important approach is to clone and sequence the relevant pathway genes. Woloshuk et a1. (1989), Horng er a1. (1990), and Skory et a1. (1990) developed transformation systems for A. flaws and A. parasiticus, respectively, which have been demonstrated to be useful tools to introduce exogenous DNA into these fungi to identify, isolate, and characterize the genes related to aflatoxin biosynthesis. Genes can be isolated by the use of heterologous sequences to screen DNA libraries, by the use of PCR primers to amplify desired genes from genomic DNA, by the use of antibodies raised against purified proteins to probe DNA expression libraries, or by the use of complementation tests with relevant mutants and appropriate selectable markers. The first aflatoxin pathway gene isolated, named nor-1, is involved in the conversion of norsolorinic acid (NA) to averantin (AVN) in the aflatoxin pathway, and was cloned by 52 genetic complementation of the aflatoxin blocked mutant strain, A. parasiticus B62 (nor- 1, niaD, brn-l), with a cosmid library which was constructed by inserting wild type A. parasiticus genomic DNA into a vector containing a nitrate reductase gene (niaD) as a selectable marker (Chang et al., 1992). Another gene, named vet-1 which encodes an activity associated with the conversion of versicolorin A to sterignatocystin, was cloned similarly by genetic complementation of another aflatoxin blocked mutant, A. parasiticus CS 10 (ver-l, pyrG, wh-l), with a cost genomic DNA library and the homologous gene (pyrG) encoding orotidine monophosphate decarboxylase for the selection of transformants (Skory e1 al., 1992). Payne et a1. (1993) cloned a gene called 1117-2 from A. flaws by using an aflatoxin defective strain and a genomic DNA hbrary fiom aflatoxigenic A. flaws in a cosmid vector containing the pyr-4 gene of Neurosprora crassa. Complernentation of the mutated afl-2 gene in a double mutant containing 1177-2 and a lesion resulting in accumulation of norsolorinic acid suggested that the product of the gene has a regulatory firnction (Payne et a1. , 1993) Another regulatory gene (apa-Z, which is very similar to 11/7-2) has been cloned fiom A. parasiticus by transformation of strains with a cosmid containing both nor-1 and ver-l (Chang et al., 1993) The two regulatory genes, apa-2 and 011-2, are both called aflR Yu er al. (1993) isolated a methyltransferase- related gene (omt-l) by the use of a polyclonal antibodies raised against a 40-kDa methyltransferase to screen a cDNA library from wild type aflatoxigenic A. parasiticus Even though there are no data showing accumulation of a stable intermediate (e. g a polyketide between acetate and norsolorinic acid), recent molecular data support the hypothesis that AFBl originates from a polyketide. 53 Figure 1-23. Restriction endonuclease and transcript map of cost Nor-A. Cosmid Nor-A was cloned from genomic DNA of the wild-type Aspergillus par-1131110113. Restriction enzymes: E, EcoRI; X, XbaI; H, HindIII; B, BamHI; Genes: pyrG (selective marker in the cosmid), encoding orotidine monophosphate decarboxylase; pksA, encoding a polyketide synthase (including acyl carrier protein, B-ketoacyl carrier protein synthase, acyltransferase, and thioesterase firnctional domains); nor-l, encoding norsolorinic acid reductase; fa3-2A, encoding a fatty acid synthetase or-subunit (including B-ketoreductase, enoylreductase, and acyl carrier protein functional domains); fa3-1A(11vm8), encoding a fatty acid synthetase B-subunit (including enoyl reductase and malonyl/palmityl transferase functional domains); qflk involved in regulation of AFBl biosynthesis; ver-lA. encoding an activity associated with the conversion of versicolorin A to sterignatocystin; Amp (selective marker), encoding B-lactamase; nor-A, encoding a reductase activity; 31cV, encoding a cytochrome P-450 protein. Symbols: narrow straight line, detected transcript with unknown firnction; narrow straight line with abbreviation below, deduced functional domain fi'om DNA sequence; narrow straight line with number below and gene name above, detected transcript with possible known firnction; Abbreviations: ACP, acyl carrier protein; KS, B-ketoacyl ACP synthase; KR B-ketoreductase; DH, dehydratase; ER, enoylreductase; MP, malonyl-palmityl transferase. 54 sameness e. 3 Pizza e. on fleecim 6.2 can: an... a..: . ea... and (Hum. I It . I are. so <75. E». 3.3 5.3 To: <31 use m ..m :2 new in.” find 8:..— efid 6.3. ...—ad. cacao $56 $55 .236 2.5 oz . «186 in... a..." en... es.. ea... can .25 3236 moses «2.5 $56 «...an 2:55 Figure 1-23. 55 Disruption of the pics-A gene in A. pamsiticus resulted in a strain which lost both the ability to produce aflatoxins and the ability to accumulate norsolorinic acid and all other intermediates in the aflatoxin pathway (Feng and Leonard, 1995; Chang etal, 1995; Trail et al., 1995). Sequence analysis showed four firnctional domains in this gene, acy carrier protein, B-ketoacyl carrier protein synthase, acyltransferase, and thioesterase, all of which are usually present in polyketide synthases and fatty acid synthetases Disruption of a gene (fas-IA) physically located between nor-l and ver-l in A. parasiticus B62 (nor-l , niaD, bmA), which normally accumulates norsolorinic acid, resulted in a new set of mutants which were unable to accumulate norsolorinic acid and AFBl (Mahanti et al., 1996). The predicted amino acid sequence of this gene showed a high level of identity with extensive regions in the enoyl reductase and malonyl/palmityl transferase functional domains in the B-subunit of yeast fatty acid synthetase suggesting that fas-lA encodes a novel fatty acid synthetase which synthesizes part of the polyketide backbone of AFB1. Therefore, fas-lA and pksA are clearly involved in AFBl biosynthesis, function before nor-1 in the AFBl pathway, and possibly work together at the fatty acid stage and polyketide stage in the AFBl pathway. A gene named nor-A was identified when an Aspergillus parasiticus cDNA library was screened with monoclonal antibody raised against a purified 43-kDa protein demonstrating norsolorinic acid reductase activity (Cary et al., 1996). Northern blot and Western blot analyses showed that norA transcript and protein are present only when the firngus was gown in medium conducive to aflatoxin biosynthesis norA is located between ver-l and nor-l in the aflatoxin gene cluster, a region which is in part located in a cosrrrid named Plenty l.v2_....e$ ...2<..n- ..Fxmv-z.. .».——...L::..I.._ ...... ......— _.....: .—J ...... .....r < .A— .... .2) v. .>. .ru 56 Nor-A (Figure 1-23 ), isolated fi'om A. parasiticus. The deduced amino acid sequence of norA had 49% anrino acid identity with an aryl-alcohol dehydrogerrase gene fi'om Phanerochaete chysosparium. The function of norA in the AFBl pathway is not clear. Purification Of Enzymes Involved In The Allatoxin Br Pathway PudfyhgmzymesflmwalyzeaflatordnsymhensfiomAflamorpranfiwsaude mycdidMactsisndeasybecausethemzymesmpresahhmlafivdylowwnMafiom and are extremely short lived (Dutton, 1988). Mashaly et a1. (1988) purified an enzyme (70 kDa) showing activity in converting sterignatocystin to aflatoxin B1. Bhatnagar et at (1988) and Keller etal (1993) have purified two methyltransferases (168 kDa and 40 kDa) to homogeneity, both enzymes catalyze conversion of sterignatocystin (ST) to O- methylsterigmatocystin (OMST). A 38 kDa reductase that catalyzes the reduction of NA to AVN was purified by Bhatnagar and Cleveland (1990); an isozyme (43 kDa) ofthe reductase also has been purified to homogeneity (Bhatnagar, unpublished observation). Another NA reductase (140 kDa) was also isolated (Chuturgoon and Dutton, 1991). Lin and Anderson (1992) purified versiconal cyclase (72 kDa) catalyzing the dehydration of versiconal to versicolorin B or versicolorin C Studies on protein isolation, purification, and enzymatic reacfionmggeuflmdifi‘aunmzymesudmmemmecatdyficfimcfionmybeisolaedfiom pertinent cells (Bhatnagar etal, 1991). Regulation 01' Allatoxin Biosynthesis The regulation of aflatoxin biosynthesis has been studied at difl‘erent levels by many researchers. Many environmental factors can influence aflatoxin biosynthesis. The three 57 most important factors controlling aflatoxin formation in field crops are relative humidity, moisture, and temperature (Russell, 1979; Wicklow, 1990; Gorman et al., 1992). At the biochemical level, studies on the regulation of aflatoxin biosynthesis started in the 1970’s. One of the early important studies was conducted by Shih and Marth (1974). They investigated the influence of sodium azide on AFBl biosynthesis and found that, at low concentration, sodium azide selectively inhibits the respiratory system of the mold, enhances production of AFBl and total lipid, and promotes [ 1-"C]-glucose incorporated into aflatoxin. Shih and Marth concluded that the decrease in oxidative respiration during late logarithmic gowth favored an accumulation of acetate (acetyl CoA) and NADPH via the Embden-Meyerhof pathway. Later, aflatoxin biosynthesis was found to be influenced by many other biochemical factors, such as the cellular energy status (Buchanan e1 al., 1987), the intracellular level of cyclic AMP (Khan and Venkitasubramanian, 1986), the cellular redox state (Bhatnagar et al., 1986), and glycolytic activity (Gupta e1al., 1977). Aflatoxin biosynthesis was also influenced by the level of glucose (Buchanan and Lewis, 1984), the levels of other carbohydrates (Abdollahi and Buchanan, 1981), and the pH value of the gowth medium (Cotty, 1988). In the 1990’s, aflatoxin biosynthesis was found to be regulated at the transcriptional level (Skory et al., 1993; Trail et al., 1995). A regulatory gene, aflR, for aflatoxin biosynthesis was characterized (Woloshuk e1_al., 1994). DNA sequence analysis showed that the aim gene possesses a binuclear zinc finger DNA-binding domain suggesting that the product of aflR is a regulatory protein (Chang e1 al. , 1995). Overexpression of qflR relieved nitrate inhibition of aflatoxin biosynthesis (Chang et al., 1995). 58 BIOCONTROL OF AFLATOJGN CONTAMINATION Possible Function Of Aflatoxins Aflatoxins may provide a mechanism for a toxigenic firngus to survive in the environment. Aflatoxins were found to be concentrated in both the conidium and the sclerotium of aflatoxigenic strains (\Vrcklow and Cole, 1982; Cotty, 1988). The presence of aflatoxins in sclerotia and conidia may have a long term survival value, because aflatoxins are toxic to a variety of predators of firngi, especially insects (erletts and Bullock, 1992). Aflatoxins may have survival value only for the producing firngus in the field. There is not a clear cut relationship between sclerotia and aflatoxin production. Some fungal strains produce aflatoxins but not sclerotia and vice versa (Bennett et al. , 1979). Aflatoxin production is highly variable within the overall A. flaws goup. Aflatoxin production can vary by as much as a million-fold among isolates fiom the same species (Clevstrom and Ljunggen, 1985). A. parasiticus and A. 1101111113 produce both the B- series and G-series of aflatoxins (Kurtzrnan et al., 1987). A. flaws is observed to produce only the B-series of aflatoxins (Samson and Frisvad, 1991). The ability to produce aflatoxin is highly conserved among most wild type strains of A. parasiticus and A. flaws (Domer et al., 1984). This ability is easily lost in culture (Cotty, 1989). Therefore, aflatoxin may only have survival value for the aflatoxigenic fungi in the field. Species Of TheAspagiHus Flaws Group One primary requirement for studies on the field populations of members of Aspergillus 59 flavlls goup is the ability to classify species ofthe A. flaws goup. There are many taxonomic schemes used to classify species in the A. flavus goup (Klich and Pitt, 1988; Samson and Frisvad, 1991). The standard for classifying a species proposed by Papa (1986) may be the best one. He proposed that a species represents a collection of strains which behave as clonal organisms with the exception of occasional parasexuality between members of the same vegetative compatibility goup (V CG). Parasexuality can be observed in the laboratory only in the same VCG. According to King and Stransfield (1985), clonal organisms are a goup of identical organisms descended from a single common ancestor by mitosis. How to determine if a goup of organisms is originated fi'om a single common ancestor? One standard is based on DNA polymorphism. Analyzing DNA polymorphism, Bayman and Cotty (1993) proposed that the A. flaws goup consists of four species which are Aspergillus flaws, A. parasitic-113, A. 1101111113, and A. tamarii. Among the four, no aflatoxigenic isolates have been obtained fi'om A. tamarii. Within each of the three aflatoxin producing species, there is geat variability. Bayman and Cotty (1991) further classified each species into Vegetative Compatibility Groups (V CGs). Each species is composed of several Vegetative Compatibility Groups (V CGs) (Papa, 1986). Physiological and morphological traits such as enzyme production, plant virulence and sclerotial morphology, are much more consistent within a VCG (Bayman and Cotty, 1993). By using this classification scheme as a tool, scientists now know the vast diversity of strains within the Aspergillus flavus goup (Papa, 1986; Bayman and Cotty, 1991), the vast diversity in ecological niches (Brown et al., 1991), the vast diversity of difl‘erent 5.0 regional population structures (Bayman and Cotty, 1991), and the vast diversity of changing populations caused by the agricultural practices (Shearer e1 01. , 1992). The distributions of the A. flaws goup fungi are very complex. Almost all species from the A. flaws goup are better adapted to a soil environment, but certain species are better adapted to niches above the soil. For example, A. parasiticus occurs more fiequently on peanuts than on corn (Hill et al., 1985). Sometimes all species within the goup may occur on the same crop or in the same field (Cotty, 1992b). Biocontrol Of Aflatoxin Contamination Eventhough aflatoxirrsareproduced onlybycertainspecies ofAmergiIIusflavus, A. paran’ticus, andA. 1101111113, these firngi are ubiquitous and fiequently gow on a variety of food and feed crops and cause aflatoxin contamination in many areas in the world (Blourrt, 1961; Ellis etal, 1991; Jelinek et al, 1989). AflmordnwmmmmonisdifiimhtodimefiommefoodchainwithomWfomsof treatment. These compounds are resistant to normal food processing such as millingand cooking, and some of their bio-metabolites are also very toxic and mutagenic such as AFMl in cow milk. Therefore aflatoxin contamination causes serious Nth problems, and in turn resultsineconomicproblerns. Therehavebeenseveralstrategiescurrentlyinuscorproposed for reducing or eliminating aflatoxin fiom contaminated food and feed such as screening/detection, removal/decontamination, or altered AFBl metabolism/DNA adduct formation (Clru, 1991; Ellis et al., 1991; Park and Liang, 1993). On the other hand, strategies formdudng/dinfinafingaflflordnmnmnfinafionattheprehmvestlevdmybebata 61 approaches. These strategies include improving agononric practices (such as irrigation and fimgicide application) and classical breeding progarns (such as selecting aflatoxin-resistant strains of crops). Thwe strategies of aflatoxin decontamination and contamination prevention arestillveryusefirlandarewidelyused. Withthedevelopment ofmodernmolecular techniques, studies on the mechanism ofaflatoxin biosynthesis at the molecular level has been an active fours area. Such studies not only can provide information for improving agonomic practices and breeding progams, for developing safe and specific ‘aspergicides’, for developing genetically engineered crops, but also can provide new biocontrol agents by knocking out genes involved specifically in aflatoxin biosynthesis to generate genetically stable nontordgeuicstrainsofAspergillus. Thesestrainsirrtheorymaybesafelyutilizedintlrefieldto mducemdhfinfleaflatordnwmmmmonoffoodorfeedaopsbybiologcalmrdusion (biocontrol). Therefore the following brief review will focus on the possrbility of developing such biocontrol agents. , The idea of using biocontrol agents to control aflatoxin contamination is that nonaflatoxigenic strains or mutants of the A. flaws goup can compete with the toxigenic strains in the field for gowing space and nutrition, and/or by the production of interfering compounds to reduce or eliminate aflatoxin contamination. It is possible to obtain nonaflatoxigenic strains for use as biocontrol agents. Aflatoxin production is highly variable (up to a million-fold) within the overall A. flaws goup. Many strains cannot produce detectable aflatoxins (Clevstrom and Ljunggen, 1985). Over the centuries, industrial strain selection has reduced fungal toxicity and increased fungal traits associated with both product quality and emcient fermentation in products 62 using Amergilli. Therefore, it is also possible to identify nonaflatoxigenic isotates in the field. Alternatively, nonaflatoxigenic strains can be created by the use of molecular techniques to genetically disrupt specific genes only in aflatoxin biosynthesis. However, not all nonaflatoxigenic strains are capable of reducing aflatoxin contamination when inoculated in the field (Cotty, 1992a). Following selection, the co-inoculation of both the producing and nonproducing strains should be tested both in the geenhouse and in the field. It is safe to use nonaflatoxigenic strains as biocontrol agents. The inoculation of nonaflatoxigenic strains or mutants in the field may cause some concern about the danger of those potential pathogens to humans. Indeed, in many areas people respire high concentrations ofAspergiII113 spores but without noticeable disease. For example, in the koji, baking, and brewing industries workers over several generations have been exposed to very high concentrations of spores throughout their working years with a very low incidence of disease (Barbesgaard et al. , 1992). It is important to note that inoculation of nonaflatoxigenic strains in the field can result in A. flaws population with altered composition, but without increased population size (Cotty, 1992c). Indeed, inoculation of nonaflatoxigenic strains in the field may provide the opportunity to improve the overall safety of fungal populations by reducing human exposure to aflatoxin through both dietary and respiratory routes. Aflatoxins can cause several physiological efl‘ects on the host crops and on domestic animals (Mclean e1 01., 1992; Robens and Richard, 1992). Reducing the aflatoxigenic population of fungi in the field can also reduce the physiological or toxic efi‘ects on aflatoxins of crops and domestic animals. 63 Normal agricultural practices generate a very large quantity of organic materials contaminated with A. flaws goup fungi in the field. Because those materials consist of crop remnants, corn cobs, gin trash, inoculation of the A. flaws goup filngi occurs naturally without our notice. The main difference between natural inoculation and artificial inoculation is that in the latter only nonaflatoxigenic strains are inoculated. Inoculation of nonaflatoxigenic strains in the field generally will not increase the infection level of crops Crop infection by the A. flavus goup ofien happens when crops are wounded or under the stress of nutrition limitation. Aflatoxins are not required for the producing fungi to infect the host crops (Cotty, 1989). Infection is more heavily dependent on host predisposition and the environment. than on the number of A. flaws. Therefore nonaflatoxigenic strains can compete with the aflatoxigenic strains for areas which are infection-predisposed in the crops. In three-year tests on cotton, inoculation of nonaflatoxigenic strains in the field has not resulted in an increase of infection rates (Cotty, 1992c). It is efi‘ective to use nonaflatoxigenic strains as biocontrol agents. The advantage of inoculation of nonaflatoxigenic strains of A. flavus over other types of microbial biocontrol agents is that nonaflatoxigenic strains are adapted to similar environmental conditions as aflatoxigenic strains. Other potential agents, such as bacteria, may be inactive under the hot, dry conditions associated with aflatoxin contamination (Brown e1 al., 1992). The A. flaws goup fungi are associated with crops in the field during crop development and remain associated with the crop during harvest, storage and processing. 64 Inoculation of nonaflatoxigenic strains in the field both before and after harvest has been shown to provide protection fiom aflatoxin contamination of corn (Brown et aI. , 1991). Nonaflatoxigenic strains or mutants of the A. flaws goup have been proven to reduce aflatoxin production and crop contamination by competition for gowing space and nutrition, and/or by the production of interfering compounds (Cotty, 1990; Shantha e101. , l 990). Perspectives On The Use Of Fungal Biocontrol Agents Based on the above discussion, the possibility of using nontoxigenic firngi as biocontrol agents to reduce or eliminate aflatoxin contamination is promising. However there are still some questions which need to be answered. One is the biology of a firngal population in the field and the other is the firnction of aflatoxin to the producing organism. Because of the tremendous reproductive and dispersal abilities of the A. flaws goup and the influence of different cr0ps and difi‘erent soils on the population structure of the firngus, scientists still do not know the change in population structure over time, or the mechanism of natural selection in the firngal population. Therefore studies on one goup of firngi have often turned out to have limited application to other goups (Cotty and Bayman, 1993 ). Even though there is much research left to be done, the preliminary results of using the nonaflatoxigenic strains to control the aflatoxigenic strains are very positive. For example, in geenhouse and field experiments, in which cotton bolls and corn ears were inoculated with combinations of different strains of the A. flavus goup, nonaflatoxigenic strains 65 reduced aflatoxin contamination by 80 to 90% (Cotty, 1990; Brown et al., 1991). Domer e1 01. (1992) demonstrated that nonaflatoxigenic A. parasiticus strains reduced aflatoxin contamination when concentrated mycelia/spore suspensions were applied to developing peanuts. Those procedures are both reliable and economic, and are active in the field under hot, dry conditions that are near optimal for the aflatoxin production by the A. flaws group. As discussed above, it is possible to identify nontoxigenic isolates in the field as biocontrol agents. Alternatively, nontoxigenic biocontrol agents (strains) can be created by the use of molecular techniques to genetically disrupt specific genes (gene-knock-out strategy) involved in aflatoxin biosynthesis. The gene-knock-out biocontrol agents may share the same niche with toxigenic strains in the ecosystem, and therefore, may be more effective in the field than the natural nontoxigenic isolates. An understanding of the regulation of aflatoxin biosynthesis and the general biology of the toxigenic fungus (e.g. regulatory mechanisms involved in hyphal gowth, hyphal difl‘erentiation, and sponrlation) may also lead to the development of new fungicides, the improvement of agononric practices, the development of resistant crops (by genetic engineering or classical breeding), and the development of biocontrol agents (such as nontoxigenic competitive fungi) for control of aflatoxin contamination. In the following chapters, the enzymatic filnction of the Nor-l protein, the pattern of Nor-1 protein accumulation in A. parasiticus gown in liquid media and on solid media, the localization of Nor-l protein accumulation and the localization of nor-1 promoter activity during aflatoxin biosynthesis, and the firnction of the flap gene which is associated 66 with hyphal development and the process of sporulation and indirectly influences the accumulation of aflatoxin in A. parasiticus are presented. CHAPTERZ ENZYMATIC FUNCTION OF THE NOR-l PROTEIN INVOLVED IN AFLATOXIN Br BIOSYNTHESIS IN ASPERGEL US PARASHYCUS ABSTRACT: To study the function of the nor-l gene, which is directly involved in aflatoxin B1 (AFBl) biosynthesis Aspergillus pw'asiticus, a nor-1 cDNA was expressed in Weinbcoli. TheresultingNor—lcproteinwasusedinanenzymeactivity assaytotestthe function ofthe nor-1 gene in the AFB! pathway. The results confirmed that the Nor-1c protein is a ketoreductase which converts the AFBl pathway intermediate norsolorinic acid (NA) to averantin (AVN) in the presence of NADPH and a supernatant fiaction (105,000 x g) obtained fiom E. coli DHSor. The results suggest that an unidentified cofactor(s) may be necessary for the function of the native Nor-l protein and that the nor-l gene is involved in only one of multiple proposed pathways for the conversion of NA to averufin (AVF) in the AFB: pathway in A. parasiziaw. INTRODUCTION The anthraquinones norsolorinic acid (NA) (CmHlsoy), averantin (AVN) (ConzoO'l), and averufin (AVF) (CmeO-I) (Figure 2-1) are each produced by blocked mutant strains of the wild-type A. parasiticus, and were demonstrated to be intermediates in the aflatoxin 67 68 Br (AFBl) pathway by feeding experiments (Lee et al., 1971; Birkinshaw et al., 1966; Donkersloot et al., 1972; Dutton, 1988). NA is converted to AVF via one or more proposed alternative pathways by converting NA to either AVN or to averufanin (AVNN) initially (Frgm'e 2—1) (Bennett and Christensen, 1983; Button, 1988; McCormick et al., 1987; Yabe er al., 1991b). The nor-l gene is physically located in the AFB] pathway gene cluster (Figure 1-23) in the genome of A. parasiticus suggesting that nor-l is involved in AFBl biosynthesis. Many genes physically arranged in a cluster in the genome of fungi are found to be involved in the same biosynthetic pathway (Timberlake and Barnard, 1981; Feitelson er a1. , 1985; Chater, 1992; Kimura and Tsuge, 1993; Hohn et al., 1993; Rainey et al., 1993; Ueda et al., 1993; Keller and Adams, 1995) for eficient expression (F eitelson et al., 1985). Disruption of the nor-1 gene in a toxigenic strain of A. parasiticus (SU-l) resulted in significant NA accmrmlation and significant reduction of aflatoxin accumulation (Trail et al., 1994) suggesting that nor-l is directly involved in AFBl biosynthesis inA. parasitic-tar. Analysis of the proposed amino acid sequence of nor-l provided evidence that this gene encodes a ketoreductase (Trail et al., 1994). Comparison of the chemical structure of NA and AVN (Figure 2-1) suggests that the 5’-keto group of NA can be converted to the S’-hydroxyl group of AVN by a ketoreductase. Therefore it is reasonable to hypothesize that the Nor-l protein encoded by nor-l can convert NA to AVN in the AFB: pathway and that the nor-1 gene is involved in one branch of proposed alternative patlmays for conversion of NA to AVF. To prove this hypothesis requires isolation of the native Nor-l protein. 69 O 5'-Hydroxyaverantin (HAVN) v v v H H \ H H H3 __ Ho H Ho CH’ 0 o Averufanin (AVNN) Averufin (AVF) Figure 2-1. Proposed multiple alternative pathways which convert NA to AVF (Bennett and Christensen, 1983; Button, 1988; McCormick er al., 1987; and Yabe etal, 1991). 70 Several NA reductase (38, 140, and 43 kDa) activities have been purified to homogeneity fiom toxigenicA. parasiticus (Bhatnagar and Cleveland, 1990; Chuturgoon and Dutton, 1991; Bhatnagar, unpublished data) which can convert NA to AVN. But these three NA reductases donothavethesamemolecularmassastheNor-l protein(3l kDadeduced fromtheDNA sequence of nor-1). These data suggest that: (l) the Nor-l protein is extremely unstable; (2) the fungal cell contains ememdy low levels of the Nor-1 protein; and/or (3) the Nor-l protein needs a cofactor(s) (in addition to NADPH) to perform the reductase activity and the cofactor(s)waslostduringpurification. Thesedataalsosuggestthatanaltemativestrategyis required to determine the fimction of the Nor-1 protein. A nor-l cDNA cloning and expression strategy was chosen in this study. Thegoal ofthisstudywasto determinetheenzymaticfimction oftheNor-lcprotein. The Nor-1c protein was produced in a host bacterium transformed with a nor-l cDNA expression vector. The enzymatic function ofthe Nor-1c protein was tested to analyze the fimction ofthe nor-1 gene in the AFB: pathway. The results confirmed that the Nor-1c protein is a ketoreductase which converts NA to AVN in the presence ofNADPH and a supernatant fiaction (105,000 x g) obtained fiom Escherichia coli DHSa. The data also suggest that an unidentified cofactor(s) may be necessary for the native Nor-l protein fiinction and that the nor-1 gene is involved in only one of multiple proposed pathways of NA conversion to AVF. 71 MATERIALS AND METHODS Bacterial Strain, lfimgal strains, and Culture Conditions. Plasmids were amplified in EWI'I'Chid coli DHSa F' °[PendA1 hem 7(rx" mid) upE44 thi-I recAI gyrA (Nal') relA IA(IacZYA-argF)mgg (m801acZAM15)] using standard methods (Ausubel et at, 1987). Amergillus parasiticus NRRL 5862 (ATCC 56775, SU-l) (American Type Culture Collections) was used as the aflatoxin-producing wild-type strain. AWIIus parasiticus ATCC 56774, which accumulates averantin, was used as a source of averantin. A nor-l disrupted strain of A.‘ parasiticus (Trail er al., 1994) (designated ANor-l) was used for NA preparation. An aflatoxin—inducing liquid growth medium consisting of 2% (w/v) yeast extract and 6% (w/v) sucrose (pH 5.8) (YES broth) was used to grow mycelia at 29°C in the dark with constant shaking (150 rpm) for protein preparation and aflatoxin extraction. YES solid medium [YES broth plus 1.5% (w/v) Bacto-agar] was used for spore preparation. Fungal strains were maintained as fiozen spore stock suspensions at -80°C. Spore stock wspensions were prepared by suspending spores in 20% (v/v) glycerol. Nucleotide Sequence Analysis. A nor-1 cDNA (with an EcoRI adapter at the 5'-end and an Mrol adapter at the 3'-end) was cloned into the EcoRI/Mrol sites of pBluescriptSKII' (Stratagene Cloning Systems, La Jolla, CA) resulting in pNOR which was kindly provided by Dr. Perng-Kuang Chang (USDA. ARS. SRRC). Nucleotide sequence analysis of the nor-l cDNAwasperfonnedbythedideoxychaintemfinafionmethodwithSequenaseH (Biochemical Corp., Cleveland, Ohio) as described in the manufacturer's instructions. The clonedinsertwassequencedonbothstrandswiththefiandTSprimers 72 fl ColEl origin pMNl , 7731hps ' maIE I malE-nor-l cDNA fusion anI nor-l cDNA Figure 2-2. Construction of expression vector pMN 1. An EcoRl/Sall fi‘agment (1040 bp) containing a nor-l cDNA was cloned into expression vector pMAL-c2. The nor-l cDNA in the resulting expression vector pMNl was fiised with the MIPS gene encoding maltose biding protein (MBP) in the same reading flame to form a maIE/nor-l-cDNA firsion which was expressed as a Nor-l c/MBP fiision protein. 73 supplied in the kit. In order to resolve the ambiguities in the initial sequence data, one internal l7-nucleotide primer (5’GTA'I'I‘TGGTCACCGGGG3’) was also used for sequwcing. Construction of pMNl, a pMAL—cz-nor-l-cDNA Expression Vector. The nor-l cDNA fi'agment was removed from pNOR using the restriction enzymes BamHI and mi and cloned into the BamI-II/Kanl sites of the expression vector pQE31 (QIAexpress, QIAGEN Inc. Chatsworth, CA). The nor-l cDNA was removed again fi'om pQE31 with the restriction enzymes EcoRI and SalI and cloned into the EcoRI/Sall sites of the expression vector pMAL- c2 (New England Biolabs Inc., Beverly, MA). The resulting vector was cut with EcoRI, and the protruding ends were filled with dN'I'Ps by the Klenow fiagment of -E. coli DNA polymerase I to create blunt ends. Blunt-end ligation created a new expression vector called pMNl (Figure 2-2) in which themr-l cDNAwas fused in framewiththe 3’-end ofthe maIE gene (encoding the maltose-binding protein) in pMAL-c2. Preparation of the Nor-lc/MBP Fusion Protein and the Nor-1c Protein. The expression vector pMNl was transformed into E. coli DHSa using standard methods (Ausubel et at, 1987). The Nor-lclMBP firsion protein and the Nor-1c protein were prepared following the instructions of the Protein Fusion & Pmification System (New England Biolabs Inc. Beverly, MA). The resulting proteins were concentrated to approximately 1 mg/ml in an Amicon Centriprep concentrator (Amicon Inc., Beverly, MA). Cleavage of the fiision protein with protease factor Xa and separation of the Nor-1c protein fiom MBP by afinity chromatography were carried out according to the manufacturer’s instructions (New England Biolabs Inc., Beverly, MA). 74 . Fungal Culture and Crude Protein Extract Preparation. For culture of the fungus in liquid media, a stock spore suspension was inoculated into liquid media with a final spore concentration of 1.5x105 spores/ml, and gown at 29°C in the dark with constant shaking (150 rpm). Mycelia were collected by filtration, and were either used immediately, or flow under liquid nitrogen and stored at -80°C until use. The dry weight of mycelia gowing in liquid media was determined after drying the mycelium in a preweighed flask overnight at 80°C. To generate a crude cell extract, mycelia were gound under liquid nitrogen with a mortar and pestle to produce a fine powder. The flow mycelial powder was quickly transferred to a 50-ml centrifuge tube containing precooled extraction bufi‘er (PEB; a volume approximately 5 times the mass of cell material) consisting of 0.01 M Tris-HCl (pH 8.0), 0.14 M NaCl, and 0.025% (w/v) NaNs. Just before mixing with mycelia, the following chemicals were added to PEB: 20 mM phenylrnethylsulfonyl fluoride (PMSF), 0.4 TIU (trypsin inhibition unit)/ml aprotinin, 10 mM iodoacetarnide, 4 mM 2-mercaptoethanol, 10 mM EDTA, and 5% (v/v) glycerol. Cell debris was removed by centrifugation two times at 1,000 x g for 10 min. The resulting supernatant fiaction was further centrifuged at 10,000 x g for 1 h to generate a crude extract (10,000 x g supernatant). For a protein concentration assay, sodium dodecyl sulfate (SDS) was added to the crude extract [to a 2% (w/v) final concentration] . and was boiled at 100°C for 5 min. The protein concentration was then determined with the Bio-Rad Protein Assay reagent (Bio-Rad Lab., Hercules, CA) according to the manufacturer’s instructions. 75 Immunoafl'mity Purification of the Native Nor-l Protein. Afinity column preparation using CNBr-activated Sepharose 4B (Sigma, St. Louis, MO) and polyclonal antibody (PAb) raised against the Nor-ldMBP fusion protein (Chapter 3. Preparation and Purification of Polyclonal Antibodies) was carried out according to published standard methods (Ausubel et al., 1995c). The basic procedure for aflinity purification of the Nor-1 protein was established by Dr. R. Mehigh in our laboratory. Briefly, an afinity coltunn (1.2 x 5.5 cm) containing 4.5 mlSepharose-4Bresinconjugatedwith l mgPAbwasequilibratedwithhighsaltphosplmte- bufl‘ered saline (HS-PBS) consisting of 390 rnM NaCl, 10 mM NazI-IPO4, and 10 rnM NaI-IzPO4 (pH 7.2). Twenty mg of crude extract (5 mg/ml) fi'om A. parasitic” SU-l or ANor-l (nor-l disrupted strain) cultured in YES liquid medium for 60 h was loaded onto the column. ThecohnnnwaswashedwithHS—PBSunfiltheODnat-mretumedtothebaseline ThenativeNor-l proteinwaselutedwithO.1 MglycineHCl (pH2.5)andcollectedwitha fiaction collector. Bach fiction (2 ml) was neutralized immediately with 0.3 ml 1 M Tris-HCl (pH 8.0). Fractions were pooled and dialyzed against three changes of low salt phosphate- bufi‘ered saline (LS-PBS) consisting of 130 mM NaCl, 10 rnM NW, and 10 mM NaHzP04(pH7.2)at4°Covera36hperiod. Western Blot Analysis. Proteins were resolved by SDS«PAGE (polyacrylamide gel electrophoresis) and blotted to a polyvinylidene difluoride (PVDF) membrane (Du Porrt Co., Boston, MA) using published standard methods (Ausubel et al., 1995b). Irrmmnodetection wascarriedoerithachemiluminescerrtdetectionldt(Schleicher& Schuell, Keene,NH) according to the manufacturer‘s instructions. 76 Nomlorinic Acid (NA) Preparation. Conidia (about 1x10° ) from a nor-1 disrupted snairLpr'asificusANor-l, wereinoculatedintoZLonES liquidmedimnandculturedfor 2daysat29°Cwithconstantshaldng(150rpm)inthedark. Theculturewasthenincubated under stationary conditions at 30°C in the dark for an additional 13-day period of time. The mycelia (red color) were collected by filtration and a portion (250 g wet weight) was extracted with300mlacetonethreetimes. ‘I‘heredpurpleextractsohrtionwasdriedwithaRotovapor R110 (Brinlonarmlnstrumentslnc. Westbury,NY) at 65°C. Theresulting solid materialswere extracted with 100 ml chloroform. The chloroform extract was concentrated with a Rotovapor R110at72°C(5-6mlfinalvolumelefi)andextractedagainwith100mlacetone. TheNAin theacaone-amwmnplered)wasfiuflrapmifiedbynuunngprepmafivethinhya clnomatogaphyCI'LCMPIG’ SILICAGEL60A, 20x20cm,WhatmanInc.Clifton,NJ)with chlorofomr-acetone (9:1) as the developing system. The NA spot (brown color) on the TLC platewasscraped ofi‘witharazorbladeandtheNAwasextractedwithchloroform. Averantin (AVN) Preparation Averantin was prepared following the method developed by Bennett et al. (1980) with modifications. A freeze-dried hyphal culture of A. parasitic-us ATCC 56774, which accumulates averantin, was reconstituted according to ATCC's instructions. Corridia (about 1x10”) ofstrain ATCC 56774 were inoculated into 2 L of YES liquidmediurn,andculhuedwithconstarfishaldng(150rpm)for2daysat29°Chrthedark followed by incubation without shaking for an additional l3-day period of time at 29°C in the dark. Approximately 210 g (wet weight) of orange mycelia were collected by filtration and extracted with acetone until colorless. Water was added to make a 30% (v/v) acetone solution. The acetone solutionwaswashed with hexane (1:1 ratio), and the orange pignents 77 were then partitioned fiom the acetone into chloroform (l :1 ratio). The chloroform extract was concentrated to 5 ml at room temperature, and then further purified by preparative TLC (HPK SILICA GEL 60 A 20 x 20 cm, Whatrnam Inc. Clifton, NJ) with chloroform-acetone (9: 1) as the developing system. The AVN spot (yellow-brown color) on the TLC plate was scrapedofi‘witharazorbladeandtheAVNwasextractedwithchlorofonn. Enzyme Activity Assay of the Nor-lc/MBP Fusion Protein and the Nor-1c Protein. Theenzymaticassaywas conducted accordingto apublished method (Yabeetal,1991b)with modifications. The reaction mixture included an appropriate quantity of protein (70 to 105 pg), 90 M NA, 0.23 rnM NADPH, 90 rnM KHzP04 (pH 7.5), and 10% (v/v) glycerol in a totalvohrmeoflOOuL Reactionswereconductedat37°C inthedarkforappropriateperiods oftime(30to90min),andthenstoppedbyadding900ulethylacetate. Theethylacetate extract was air dried and the residue was extracted by adding 400 ul chloroform. The reaction products in chloroform extract were resolved by TLC (HPK SILICA GEL 60 A, 10 x 10 cm, Whatmam Inc. Clifton, NJ) with benzene-ethyl acetate (7:3) as the developing system. UV/VIS, Mass, and NMR Spectroscopy of NA and AVN. UV/VIS spectroscopy was conducted using a UV/Visible light spectrophotometer (CARY 3E, Varian, Australia Pty. Ltd., Australia). Mass spectrometry was carried out on a double-focusing mass spectrometer (IEOL AX-SOSH, Varian, Australia Pty. Ltd., Australia) with the help of Dr. B. Charnberlin (Department of Biochemistry, Michigan State University, East Lansing MI) under the following conditions: electron energy, 70 eV; scan range, m/z 45-600. The 1H proton nuclear magnetic resonance (NMR) spectroscopy was performed with a NMR spectrometer (VX12- 500, Varian, Australia Pty. Ltd., Australia) with the help of Dr. K. Johnson (Department of 78 Chemistry, Michigan State University, East Lansing, MI) under the following conditions: proton probe at 80 mHz; range, 0 to 12 ppm; solvent, deuterated dimethyl sulfoxide (d6DMSO). AflatoxinAnalysisbyTLCand ELISA. Aflatoxinswereextractedwith lOrnlof chloroform fi'om mycelial liquid culture medium (not including mycelium) (1 ml) or from mycelial solid culture medium (including mycelium) (1 mg). Chloroform extracts were tested either semiquantitatively by TLC (for total aflatoxins) with chloroforrn—acetone (95:5) as the developing system, or quantitatively by enzyme-linked irnrnunosorbent assay (ELISA) (for AFBr). The direct competitive ELISA was carried out according to the method ofPestka et at (1980) using anti-AFBr antibodies kindly provided by Dr. J. Pestka (Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI). RESULTS Sequence Analysis of the nor-1 cDNA. Nucleotide sequence analysis (Figure 2-3) showed that the nor-1 cDNA was missing 6 nucleotides (ATGAAC) fiom the protein-coding region at 5'-end of the proposed transcript beginning with the translation initiation codon (ATG) based on a comparison with the nor-1 genomic DNA sequence (T rail et al., 1994). It msthoughtthatthisshort deletionrnightnothaveasignificantefl‘ectontheNor—lcprotein activity. Oneofthethreereadingfi'amesofthemr-l cDNAencoded acomplete openreading fiameand had three in-fi'ame stop codons at the 3'-end which in theory could geneate proteins withpredicted molecularrnassof31 kDa, 34 kDa, or38 kDaifreadthrough oftheprimary translation termination codon occurred. 79 Factor Xa cutting sit: ( nor-1 cDNA 5'... ATG AAC) l DNA 5':de....§ FAQ fig AQG ATT TOA GAA TTA ATTOGG OAO GAGGGATOA CTI' AGO CAG CAO GATCM N-AA 001 E G R l S E L I R H E G S L S O H D Q DNAS': GAGAGGCTCTOTAOGCOATAOCGGGATGGACOGOOTGAGGAGAOGGTGTAT'ITGGTOAOC MMERLSTPYRDGPPEFTVYLVT N—AA: 041— 230 DNAS': TGG TAG GAO GOT AGT GAO GACGMGOG OAG TOA TATGTT TOT GAG ACGAGCAGA MOGTG N-AA2281WS1DASDDEAQSYVSETSRNV DNAS’: OTG GOG TGG AGO TGG TOO AGA MG GOG COG TGA GGT COT GGG TTC GTG TOG COG COG CTT N—MI301LAWSWSRKARSZGPGFVSRRL DNAS': TOG OTC CAT CAG TTO GTG TAT GAO CTT TIC GGT OTT TTC OGT TCO GTC TGT 'l'l'A GGO TOT NM321$LDQFVYDLFGLFRSVCLGS DNAS': OACMGATAMACCAMTTGAMCOTAAOGTI'OGTTTTOAMAMMAAMMAMAAAC 3411-! K I K P N #3 N L T F V F K K K K K K T DNAS‘: TOG A66 666 GGC COG GTA COO CGG GTC DC T A N-AA: 318 R G G P V P R Figure 2-3. Nucleotide and predicted amino acid sequence of the 5’ and 3’-ends of nor-1 cDNA in expression vector pMNl. The underlined nucleotides represent the carboxyl terminus of the maltose binding protein encoded by maIE (in the expression vector pMAL-c2) which is fused in-fi'ame with the nor-1 cDNA. The nucleotides in italics were introduced during vector construction. The nucleotides in bold represent the nor-1 cDNA. The nucleotides in parentheses (ATGAAC) were missing fiom the protein-coding region at 5'-end of the proposed transcript beginning with the translation initiation codon (ATG) based on a comparison with the nor-1 genomic DNA sequence. The factor Xa cutting site is indicated by an arrow. The two Nor-1c proteins (starting fiom the factor Xa cutting site, 32 and 36 kDa) are predicted to be derived fiom translation termination at the first two stop codons (marked #1 at residue 282, and #2 at residue 311), respectively. The two Nor-1c proteins are therefore predictedtolacktwoarnino acids(M, andT)which are replacedby8 amino acid(I, S, E, L, 1, IL H, and E) at the amino terminus. N-AA indicates the N-terrninus of the amino acid sequence. 80 97* m a“ «78‘ _ to .....— 66‘ ~ 9* 74 ., ..., 45-- Q. """’ EL? .- «42 ...... "-3 r..- *36 31—b - ‘c 22—. - Figure 2-4. SDS-PAGE detection of the Nor-IcIMBP firsion protein and the Nor-1c protein. Expression vector pMN 1 containing the nor-l cDNA fused with maIE encoding maltose binding protein (MBP) was transformed into Escherichia coli DHSor. Under iSOphenylthiogalactoside (IPTG) induction, two Nor-lclMBP fusion proteins were produced in the bacterial host. Lanes: 1, molecular mass standard (kDa); 2, total crude extract (10,000 x g supernatant) from E. coli DHSor; 3, total crude extract from E. coli DHSor transformed with expression vector pMNl; 4, two Nor-lc/MBP fusion proteins (74 kDa, and 78 kDa) purified by affinity chromatogaphy; and 5, two Nor-1c proteins (32 kDa, and 36 kDa) and MBP (42 kDa) obtained by factor Xa cutting of the two Nor- 1c/MBP fusion proteins. 81 ¢- Nor-1 Figure 2-5. Western blot analysis of the native Nor-l protein in A. parasiticus SU- 1. Each lane contained 10 pg protein. The primary antibody was the IgG fraction of the antiserum raised against the Nor-ldMBP fusion protein (Chapter 3) (10 ug/ml). Lanes: 1, total crude extract (10,000 x g supernatant) fiom the nor-l-disrupted strain A. pw'asiticus ANor-l cultured in YES liquid medium for 60 h; 2 and 3, total crude extract fiom the wild-type A. parasiticus SU—l cultured in YES liquid medium for 48 h and 60 h, respectively; 4, the native Nor-1 protein (31 kDa) purified with anti-Nor-lc/MBP firsion protein PAb aflinity column from the total crude extract of A. parasiticus SU-l cultured in YES medium for 60 h. 82 . Preparation of the Nor-ldMBP Fusion Protein and the Nor-1c Protein. The express'onvectorpMNl wastransformedintoE coliDHSor. Afier2hinductionwith isophenylthiogalactoside (IPT G), two Nor-ldMBP fusion proteins (approximately 74, and 78 kDa)wereproducedinthehostcdlandwaedetectedbySDS-PAGE(Figne24). The firsimpmtdnswaepnifiedbynnhoserednammychmmatogaphyandanwhhpmtease factorXamrltinghrtheappearanceofthemaltosebhrdingproteinMZ kDa)andtwoNor-1c proteins(approximately32kDa, and36kDa)(Figure2-4). Immunoaflinity Purification of the Native Nor-l Protein. Twenty mg of crude extract obtained fi‘om the aflatoxin-producing strainA. pen-asia’als SU-l, or fi'om the nor-l disrupted strainpr'asiticw; ANor-l, was used to purify the native Nor-1 protein by imrnunoaflinity chromatogaphy. The concentrated Nor-1 protein, resolved by SDS-PAGE, was detected by Westernblotanalysis. Onlyonemajorproteinbandcorrespondingtothe sizepredictedforthe Nor-l protein was detected by the IgG fiaction of the anti-Nor-lc/MBP firsion protein serum (Chapter 3) in the wild-typeA. parasiticus SU-l (Figure 2-5, lane 4). This protein band was notdetectedusingthe sameantibodiesinthenor-l disrupted strainprusiticusANor-l (Figure 2-5, lane 1). Using the same antibodies, in addition to the expected Nor-1 protein, severalotherproteinsweredetected inthecr'udeextract ofA. pm'asr'tr'cus SU-l (Figure 2-5, lanes 2, 3) suggesting that cross-reactivity occurred. Enzyme Activity of the Nor-ldMBP Fusion Protein and the Nor-1c Protein. The imnnmoafinity purified Nor-ldMBP fusion protein, the Nor-ICIMBP fusion protein in the crude extract (10,000 x g supernatant) ofE coli cells containing expression vector pMNl, and thehmumaflimtypmifiedNor-lcpmtdnwereassayedfortheuabflhymwnvatNAm 83 Figure 2-6. Enzyme activity assay of the Nor-lc/MBP fusion protein and the Nor-1c protein. All reactions were conducted at 37°C in the dark. The reaction products were resolved by TLC with benzene-ethyl acetate (7:3) as the developing system. The photo of reactions 1 through 9 was taken under white light. The photo of reactions 10 through 18 was taken under UV light. Reactions: 1, NA standard; 2, AVN standard; 3, NADPH plus NA control. *, crude extract (10,000 x g supernatant) of E. coli DHSot; v, pellet (105,000 x g) of crude extract from E. coli DHSor; 4, supernatant (105,000 x g) of crude extract fiom E. coli DHSa; o, 35 ug anti-Nor-lc/MBP firsion protein PAb; 0, Nor-1dMBP fusion protein was stored for 4 months at -80°C; + or -, containing or not containing the component in the same row in the “Reaction components” column. team :eom :8 mm m n ma): :cuum . .0 mg 2.32 =8 ..m mg zoomsw n n ma): Am :06 ..N mg :8 ..w o _ LoZ :8 m =8 .m 85 AVN (Figure 2-6). In the presence of NADPH, only the crude extract fi'om E coli DH5a containing the Nor-lc/MBP fusion protein (reaction 6), the mixture of the afinity purified Nor- lc proteinandtheE colicell crudeextract (reaction 8), andthe mixture ofthe afinitypurified Nor-1c/MBP firsion protein and the E coli cell crude extract (reaction 10) completely convertedNA(90uM)toacompoundthatco—migatedwithAVNuponTLCanalysis. Neither the afinity purified Nor-1c protein (not shown) nor the control E coli extract (reaction 4) converted NA to AVN in the presence of NADPH. These data suggested that E coli provided some essential cofactor(s) required for NA reductase activity. The cofactor(s) was foundinthe 105,000 xgsuper'natant (reaction 12)andnotinthe 105,000 xgpellet oftheE coliextract (reaction 11). NADPH, E coli cell extract, andE coli cell extract containing MBP were unable to convert NA to AVN (reactions 3, 4, and 5). Anti-Nor-lc/MBP fusion protein PAb inhibited the Nor-ldMBP activity (reaction 13) suggesting that the enzymatic activity originated fiorn the Nor-1c protein. The freshly prepared Nor-lclMBP firsion protein finished the NA-AVN conversion in 30 min and the stored (at —80°C for 4 months) Nor-lc/MBP fusion protein finished the same conversion in 50 min (reactions 14 to 18) suggesting that storage reduced the Nor-1c protein activity. UVIVIS,Mass,andNMRSpectra ofNA. To confinntheidentityofthecompomrd usedasthesrbsuatemthemzymeacfivityassay,thecompoundwaspmifiedmtwo consecutiveTLCsteps. Thepurified compound, whichhadthesameRfvahre(0.45)with benzene-ethyl acetate (7:3) as the developing system and the same color (brown under white light)asthestandardNA,wasscrapedfromtheTLCplateandthenanalyzedbyUV/VISCUV A 4 .9990:I 3 .0999J 2 .9099- 1.8880- 9.009 , ' . . 209.99 399.99 499.99 599.99 B 3 .8880- Fignre 2-7. UV/VIS spectroscopy of norsolorinic acid (NA) and averantin (AVN). A: UV/VIS spectrum (200-600 nm) of NA (rn ethanol). 3. max (EtOHXe): 235nm(24,500), 269nm(16,900), 284mn(18,600), 297nm(19,900), 314nm(22,900), and 465nm(7,760). B: UV/VIS spectrum (200-600 nm) of AVN (in ethanol). 9e mx(EtOH)(a): 222nm(26,784), 262nm(15,810), 298nm(20,386), 315nm(22,022), 453 nm(6,658). 87 * A 2.. J 88-1 J t ...: l ‘ L P 48: 3?? 31,8 ; 3 4 ‘ Z . 3 1 l O 50 100 150 200 250 300 358 "/2 100 83‘ i r en 1 . . l 49- 395 . 20‘ Z 7 ' , 3 1 r ‘ l' 373.,“ is ‘T’ ’ 62:?3.‘ laLE?§1 l'lfignll IIlaLnxgrnna'1..-11-Aaa?3:aaa gilllllll [In-galalfixlsi Ian'.-a.'xx-3;?l6n-;-.x .1 an. i 300 328 349 360 383 403 "/2 Figure 2-8. Mass spectroscopy of norsolorinic acid (NA) and averantin (AVN). A: The massspectrumofthepurifiedNA [relativeintensity(%): molewlarionatm/2370;fiagments at m/z 327, 314, 299, 272]. B: The mass spectrum ofthe enzymatic end product AVN [relative intensity (“/o): molecular ion at m/z 371; fragments at m/z 354, 325, 311, 297, and 285]. 88 DMSO cam. 5.15 conm, 3.53 Figure 2-9. NMR spectroscopy of norsolorinic acid (NA) and averantin (AVN). A: NMR spectrum of NA (in d6DMSO) (6pprn): CH3(6'), 0.88(3H); CH2(4'5'), 130(2H); CHZC“). 1-60(2H); 012(2). 281(2H); CH(7). 6-61(1H); CH(5). 7-l3(1H); CH(4). 7220K)- B: NMR spectrum of the end product AVN (in d6DMSO) (fippm): CH3(6’), 0.85(3H); CH2(3',4',5'), l.30(6I-I); CI-I2(2'), 1.84(2H); COH(1'), 3.53(1H); CH(1'), 5.15(1H); H(7), 653(111);H(5). 7-19(1H); 11(4). 7220“). 89 and visrhle light), mass, and NMR spectroscopy. The six maximum absorbence values of the WMS spectrum (200-600 nm) of the purified compound in ethanol (Figure 2-7A) were consistent with the data for NA reported by Cole and Cox (1981) [A max (EtOHXa): 235nm(24,500), 269mn(l6,900), 284nm(18,600), 297nm(19,900), 314nm(22,900), and 465nrn(7,760)]. The mass spectrum (Figure 2-8A) and the NMR spectrum (Figure 2-9A) of thepurified compoundwerealso consistentwiththepublisheddataforNA(ColeandCox 1981’) [mass spectrum (relative intensity (%): molecular ion at m/z 370; fiagments at m/z 327, 314, 299, 272; NMR spectrum (in d6DMSO, 8pprn): CH3(6'), 0.88(3H); CH2(4',5'), 130(21‘1); CH2(3'). 1-60(2H); CH2(2'). 2-81(2H); CH(7). 6-6l(1H); (311(5). 7-13(1H); CH(4). 7.22(1I-I)]. UV/VIS,Mass,andNMRspectraofAVN. Toidentifytheendproductinthe conversion of NA by the Nor-lc/MBP fusion protein, the enzymatic reaction was scaled up 10 fold. The enzymatic end product, which had the same Rf value (0.37) with benzene-ethyl acetate (7:3) as the developing system and the same color (yellow under white light ) as the standardAVNon'I'LC,werescrapedfi'omthe'ILCplateandthenanalyzedbyUV/VISCUV and visible light), mass, and NMR spectroscopy. The five maximum absorbence values of the WMS spectrum (200-600 nm) of the end product in ethanol (Figure 2-7B) were consistent with AVN data reported by Bennett et at (1980) [A mx(EtOI-I) (a): 222nm(26,784), 262nm(15,810), 298nm(20,386), 315nm(22,022) 453 nm(6,658)]. The mass spectrum (Figure 2-8B) of the end product was consistent with AVN data published by Bennett et al. (1980) [relative intensity (%): molecular ion at m/z 372; fiagments at m/z 354, 325, 311, 309, 297, and 285] with one exception (molecular ion at m/z 371, not at m/z 372 as expected). The NMR 90 spectrum of the end product in d6DMSO (Figure 2-9B) was consistent with the data for AVN published by Bennett et at (1980) [(5ppm): CHB(6'), 0.85(3H); CHZ(3’,4',5'), 1.30(2H); CH2(2'), 1.74(2H); COH(1’), 3.53(1H); CH(l’), 5.15(lI-I); CH(7), 6.S3(1H); CH(S), 7.02 (1H); CH(4), 7.02(1H)]. Two “new peaks” appeared in the NMR spectrum of AVN. One was COH(1'), 3.53(8ppm); another CH(l'), 5.15(6ppm). These two new peaks resulted fi'orn keto-reduction by the addition of two more hydrogen atoms. The standard AVN purified fiom thefimgal snainAptrr'asitiatsATCC 56774was shownto luvethe sameUV/VIS, mass, and NMRspectraasAVNgeneratedintheNAreductasereaction. AFBr Accumulation in A. pansiticus SU-I and A. panniticus ANor-I. Direct competitive ELISA analysis was utilized to quantify AFBr accumulation in the wild-type A. pen-annals SU-l and the nor-1 disrupted transformarrt, A. pen-asiticus ANor-l, arltured in YESliquidmedium. After2,and5daysofculture, approximately25 foldmoreAFBr was produced by the wild-type A. pw'asr'ticus SU-l (4.55 i- 0.02 mg, and 6.8 :t: 0.03 mg of AFBl per garn of dry mycelial mass, respectively) when compared with the nor-l disrupted transforrrrant A. pw'asr'ricus ANor-l (0.18 :t 0.02 mg, and 0.27 :l: 0.03 mg, respectively). DISCUSSION The data presented in this study demonstrated that the Nor-1c protein (or the Nor-lclMBP firsion protein) is able to convert the aflatoxin Br pathway intermediate NA to AVN but only in the presence of NADPH and a cofactor(s) obtained fi'om E coli DHSot. This suggests that the rmtive Nor-1 protein is an NADPH dependent NA ketoreductase requiring an unidentified 91 cofactor(s) and that nor-1 encodes an activity involved in one of multiple proposed pathways for the conversion ofNA to AVF in the AFBl pathway inA. par-annals SU-l. The Nor-1c protein has a ketoreductase activity capable ofconverting NA to AVN in vitro. TheUVNIS, mass, anthrIRspectrmndataconfirmedthatthesubstratewasNAandtheend product was AVN. The conversion of the 1'-keto goup of NA to the l’-hydroxyl goup of AVNandtherequirementofNADPHintheenzymaticconversion confirmedthattheactivity involved in this conversion is a NADPH dependent NA ketoreductase activity. The conclusion thumlytheNor-lcpmtdnmmiskaoreduaaseacfivuymmemzynwacfivhyassaywas deduced fi'om the following three observations: (1) the conversion of NA to AVN was inhibited by the addition of the anti-Nor-lc/MBP firsion protein PAb when the Nor-1c protein was involved in the enzyme activity assay, (2) NADPH, the E coli protein plus NADPH, and the MBP plus the E coli protein and NADPH were unable to convert NA to AVN; and. (3) analysis of the proposed amino acid sequence of nor-l provided evidence that the Nor-1c proteinwasaketoreductase (Trail eral, 1994). Bhatnagar and Cleveland (1990) proved that the conversion of NA to AVN is reversible in the presence of a 38-kDa reductase plus NADP. This may also be true for the Nor-1c protein, butthereversereactionwasnot conductedinthecurrent study. The Nor-1c protein showed NA ketoreductase activity only in the presence of a cofactor(s) fi'omE coli DHSa suggesting that a cofactor(s) may be also necessary for the native Nor-1 protein to show the NA ketoreductase activity. A 38-kDa reductase (Bhatnagar and Cleveland, 1990), a 43-kDa isozyme of the reductase (Bhatnagar, unpublished observation), and a 140-kDa reductase (also called dehydrogenase) (Chuturgoon and Dutton, 1991) have 92 been purified to homogeneity, and are reported to convert NA to AVN. Surprisingly the purification of the 31-kDa native Nor-l protein encoded by nor-1 has not been reported. This obsavafionmaybeetplaimblewiththedatapresentedmthisstudfiseebdow). Western blot analysis using PAb raised against the Nor-le/MBP firsion protein showed that:flreacarmrflafionofa3l-kDapmtdnhadapattanconsistentwiflrAFBr accumulationin A. parturition: SU-l (Chapter 3); the 31-kDa protein purified fiom a crude extract ofA. parasitic” SU-l byafinitychrormtographywasdetectedasasingleband; andthe31-kDa protein was not detected in a crude extract fi'om the nor-l disrupted strain ANor-l. These observations suggest that the 31-kDa protein is the native Nor-1 protein encoded by the nor-1 gene. But, the nor-1c protein alone does not show the reductase activity. Therefore, it is possible that a cofactor(s) is needed for the activity of the native Nor-1 protein. The cofactor(s)mayhavebeenlostdmingpurificationstepinprevious studiesresultingininability to purify Nor-1. Such a cofactor(s) was possibly provided by E coli DHSa in this study. Alternatively, because polyketide synthase (PKS) and fatty acid synthetase (FAS) enzymes often firnction in multisubunit complexes, it is possible that the native Nor-1 protein is a part of a multiple polypeptide complex required for AFBl biosynthesis. Therefore, only when it is mchtdedmdheldinafirncfionalconfigmafiondhecflyassodatedwhhthecomplag doesthe native Nor-l protein have the reductase activity. It is hypothesized that certain low mass element(s) in an E coli supematant (105,000 x g ) substitute for a required cofactor or eliminate the requirement for protein/protein interaction to help the Nor-1 protein to show reductaseactivity. Fmtherresearchiswarranted. 93 Thenar-l geneiscorrfirmed tobedirecdyinvolvedhrAFBrbiosynthesisinAparan'fic-ras, an observation which was reported previously (Skory et al., 1993; Trail et at, 1994). ELISA datainthisstudyshowedthatthenw-ldisruptedstrainANor-l reducedtheaccunmlationof AFBrbyapproxirnatelyZSfold. TheNor-lcproteinencodedbythenar-l cDNAwasableto convertNAtoAVN,andbothNAandAVNhavebeenproventobetheintermediatesin AFBl pathway(Bennett etaL, 1980;Yabe etaL, 1993). ThemassandNMRspectroscopicdatastronglysuggestthatNA(370Da)isconvertedto AVN(372Da)(the l'oketogoupisreducedtohydroxylgoup), eventhoughthetheoretical molewlarmassofAVNis372Da,buttheobservedmassspectrumofAVNhadabasepeak atm/z371,notatm/z3729sexpected. OfienionsCM—l)*arldsonretimes(1\d-2)*,(M-3)*are observedintheprocessofmassspectroscopyduetolossesofhydrogenatom(s)fi'omthe molecular ion M. Sometimestheion (M-1)*ismuch moreabundantthanthemolecular ion M(RoseandJohnstone, 1982)asoccurredherewithAVN. TheNMRspectrumdataofNA wasalsoconsistentwiththeliteratme(ColeandCox,1981),withoneexception.Norsolorinic addmflnsresmrchwasprepmedudmomaaymflimfionaepfliueforeflnNMRspewtm ofNAinthisstudynotonlyhadamajorpeakatZS5ppmwhichoriginatedfiomDMSO,but alsohadapeakat3.46ppmwhichoriginatedfiomwatdzTheNMRdataofAVNweremore complicated. TheerrzyrnaficelidproducLAVN,andtheAVNpreparedfiomA.pa-asificus ATCC56774mdnsaudyhad-flxsmneNMRspchumThisNMRspem‘mnwasmnfigau withthedatareportedbyBennettetal. (1980),butwaslessconsistentwiththedatareported by Birkinshaw et al (1966) [(8ppm): CHB(6'), 1.00(3H); CHZ(3’,4',5'), l.40(21-I);CH2(2'), 94 1.90(2H); CH(7), 6.15(1H); CH(S), 6.80(1H); CH(4), 6.82(1H)]. These observed difl‘erences couldrenrltfiomdifl‘eremquafityofsolvemsanddifi‘ereminsumnm Insurnmary, thedatapreserrtedinthisstudyconfirmedthattheNor—lcproteinisa ketoreductase which converts the AFBl pathway intermediate norsolorinic acid (NA) to averatin (AVN) in the presence ofNADPH and a supernatant fraction (105,000 x g) obtained fi'omE coliDI-ISG. Thissuggeststhatanuniderrtified cofactor(s)maybenecessaryforthe nativeNor-l proteinfirnctionandthatthem—l geneisirrvolvedinonly one ofrmrltiple proposedpathwaysfortheNAconversiontoaverufin(AVF)intheAFBr pathwayinA. pw'asiticus. CHAPTER3 THE ACCUMULATION OF THE NOR-l PROTEIN AND THE ACITVITY OF THE NOR-l PROMOTER DURING AFLATOXIN B1 BIOSYNTHESIS IN ASPERGEL US PARASIHCUS ABSTRACT: Two difi‘erent procedures were utilized to study the expression of the nor-l gene during aflatoxin Br biosynthesis in Aspergillus parasiticus. First, polyclonal antibodies, which were raised against the Nor-1c protein (expressed in Escherichia coli fiom a nor-1 cDNA), were used to monitor the accumulation of the native Nor-1 protein. Second, a nor-l/GUS reporter construct was used to monitor the activity of the nor-1 promoter. The results showed that the accumulation of the Nor-1 protein (in liquid media and on solid media) and the activity of the nor-1 promoter (on solid media) are regulated in the same pattern as the accumulation of afiatoxin and are also closely correlated to the development of sporulation in A. pw'asiticus. The results also suggested that the expression of nor-1 is regulated in a temporal and spatial pattern in A. parasiticus gown on sold media. INTRODUCTION A major focus of current studies on aflatoxin biosynthesis in Aspergillus parasiticus is the regulation of the expression of the genes involved in this complex pathway. Northern 95 96 blot. analysis demonstrated that the expression of nor-1, which is directly involved in aflatoxin B1 biosynthesis, is regulated at the level of transcript accumulation in the toxigenic strain A. parasitism SU-l gown in liquid media (Skory et al., 1993; Trail et al. , 1994; Trail 01 al., 1995). However, the expression of nor-1 can also be studied at the level of the accumulation of the Nor-l protein and at the level of the activity of the nor-l promoter in A. parasiticus. Condifimsofmrdgancfimggnwingmfiquidmediamevaydifi‘erentfiomthenatural environmentalconditionsofthe sarnefungigowinginsoil oronhost plantsinthefield. For example, sporulation usually cannot be observed in liquid media. Therefore, studies conducted infiqlfidmediamaynotcompletdyrefiecttheregflafionofthegenesinvolvedinafiatoxin biosynthesis in toxigenic fungi gown on solid media. Aflatoxins have been found in high concentrations in spores (\Vrcklow and Cole, 1982; Wrcklow and Shotwell, 1983; Cotty, 1988; Bayman and Cotty, 1990). Mycelial gowth rate wasndrcedunsponranonandadmdnaecrnudanonweetotnlyinnbnednammmd A. parasiticus by addition of 2.0% (v/w) phosphates to Sabouraud dextrose agar (Lebron et al., 1989). These data suggest that the regulation of aflatoxin biosynthesis may be correlated to sponrlation. Basedonthesedataandobservations, themajorhypothesisinthisstudyisthatthe acclmmlationoftheNor-l protein(in liquidmediaandon solid media) andtheactivityofthe nor-l promoteractivity(on solid media) areregulatedinthesamepatternastheaccunmlation of aflatoxin and are correlated to the deveIOpment of sporulation in A. pw'asificus. 97 The data presented in Chapter 2 demonstrated that the Nor-1c protein was expressed in E coli fiom a nor-1 cDNA, the Nor-1 protein had the same molecular mass .as that predicted fi'om the nor-l gene sequence, and the Nor-1 protein had the enzymatic activity . necessary to convert norsolorinic acid (NA) to averantin (AVN), two intermediates in the AFBl pathway. These data suggested that the Nor-1c protein and the Nor-1 protein may have very similar, if not identical, amino acid sequences, and therefore, have the same inununogenicity and the same anitgenicity. Hence, a subsidiary hypothesis is that polyclonal antibodies (PAb) raised against the Nor-1c protein would be useful to monitor the accumulation of the Nor-l protein. The Escherichia coli uidA gene encoding B—glucuronidase (GUS) has been used in fungi as a reliable reporter for gene expression at the level of promoter activity (Monke and Schafer, 1993). Therefore, the activity of nor-1 promoter can be monitored by a nor-1 promoter/GUS construct (nor-l/GUS) providing another tool to address the major hypothesis. Accordingly, two experimental procedures are proposed to address the major hypothesis in this study. First, polyclonal antibodies (PAb) will be raised against the fusion of the Nor-1c protein and the maltose binding protein (MBP) (N or-lc/MBP). The PAb will be used to detect the accumulation of the Nor-1 protein. Second, a GUS reporter construct will be used to monitor the activity of the nor-l promoter. The accumulation of the Nor-1 protein and the activity of the nor-1 promoter will be compared with the pattern of afiatoxin accumulation and the development of sponrlation during aflatoxin biosynthesis. 98 The data presented in this study showed that the accumulation of the Nor-1 protein (in liquid media and on solid media) and the activity of the nor-1 promoter (on solid media) are regulated in the same pattern as the accumulation of aflatoxin and are closely correlated to the development of sporulation in A. parasiticus. The data also suggested that the expression of nor-1 is regulated in a temporal and spatial pattern in A. parasiticus gown on sold media. MATERIALS AND METHODS Bacterial Strain, Fungal Strains, and Growth Media. Plasmids were amplified in Escherichia coli DHSa F' °[F'endAI hstI 7(rK-rnK+)supE44 thi-l recAI gyrA (Nal‘) relAI (IacZYA-argF) 0169 (m801acZ AM15)] using standard methods (Ausubel et al. , 1987). Aspergillus parasiticrts NRRL 5862 (ATCC 56775, SU-l) was used as the aflatoxin-producing wild-type strain. An A. parasiticus transformant (designated nor- 1/GUS) containing pAPGUSNN, a reporter construct containing the nor-1 promoter fused with the E coli uidA gene encoding B-glucuronidase (kindly provided by David Wilson in our laboratory) was used to monitor the activity of nor-l promoter. In this transformant, pAPGUSNN was inserted at the 5’-end of the chromosomal nor-1 locus. Fungal strains were maintained as frozen spore stock suspensions [in 20% (v/v) glycerol] at -80°C. Aflatoxin-inducing media, YES broth [2% (w/v) yeast extract, 6% (w/v) sucrose, pH 5.8] and YES agar [YES broth containing 1.5% (w/v) Bacto-agar] were used to gow mycelia for protein preparation and aflatoxin extraction. 99 Fungal Culture, Dry Weight Determination, and Analysis of Fungal Morphology. For gowth of fungi on solid medium, 5 ul of a spore stock suspension (1.5 x 10° spores/ml) was inoculated onto the center of YES agar media and gown at 29°C in the dark. Mycelia were collected using a spatula to scrape the mycelia from the surface of agar. The collected mycelia were either used immediately or flow under liquid nitrogen and stored at -80°C until use. The dry weight of mycelia gowing on solid media was determined by a filtration/drying method described by Olsson and Jennings (1991). For gowth of firngi in liquid media, a stock spore suspension was inoculated into liquid media with a final spore concentration of 1.5 x 105 spores/ml and gown at 29°C with constant shaking (150 rpm) in the dark. Mycelia were collected by filtration and were either used immediately or fiozen under liquid nitrogen and stored at -80°C until use. The dry weight of mycelia gown in liquid media was determined by drying the mycelial pad in a preweighed flask overnight at 80°C before weighing. A nutritional shift culture assay using nonaflatoxin-inducing peptone-rnineral salts (PMS) medium and aflatoxin-inducing glucose—mineral salts (GMS) medium was performed according to Wiseman and Buchanan (1987). Briefly, spores were inoculated into PMS medium (1.5 x 10’ spores/ml) and incubated for 60 h and then shifted into GMS medium for a 60-h period of incubation. The morphological characteristics of a colony gown on YES agar were examined under a dissection microscope. Temporal and Regional Collection of Samples From Solid Growth Medium. Corridia (7.5x 10’)wereinoculatedatthecenteronESagarinPeuidishes. Colonieswiththe same gowthratewereselected. Forcoflecfingsmnplesfiomthecmtalregionatleast4cultures 100 wereneeded. Twodaysaiterinocrrlafiontheareacoveredbythemycelimn (infourPetri dishes)wasmarkedwithacircleanddesignatedthe‘centralregion’. Two days(4days, 5 days, or6days)afierinoarlafion,themycelimnwhichcoveredthecenfialregion ofthefirst (second, third, or fourth) Petri dish was collected as the ‘2-day’ (‘4—day’, ‘S-day’, or ‘6-day’) ‘central-region’ sample C2 (C4, C5, or C6). For collecting samples fiom the middle region, at least3 cultureswereneeded. TwodaysaflerinoarlafiontheareacoveredbythemyceliumCm threePetridishes)wasmarkedwithadrcle (the2-daymark). Fourdaysafierinoculation, the areacoveredbythemyceliurn(rnthreePetridishes)wasmarkedwithasecondcircle(the4- daymark). TheareabetweentheZ-daymarkandthe4—daymarkwasdesignatedthe ‘middle region. Fourdays(5 days, or6days)afierinoculation,themyceliawhichcoveredthemiddle region of the first (second, or third) Petri dish was collected as the ‘4-day’ (‘5-day’, or ‘6-day’) ‘middle-region’ sample M4 (M5, or M6). For collecting samples from the peripheral region, at least2arltureswereneeded. Fourdaysafierinoarlafiontheareacoveredbythemyceliumfin twoPetridishes)wasmarkedwithacircle(the4-daymark). Theareabetweenthe4-daymark andthedrcularedgeoftheagarwasdesignatedthe ‘peripheralregion’. Five 5 days(or6 days)aflerinoculation, themyceliawhichcoveredtheperipheral regionofthefirst (or second) Petri dish was collected as the ‘5-day’ (or ‘6-day’) ‘peripheral region’ sample P5 (or P6). The sampling procedure is shown schematically in Figure 3-5B. Crude Protein Extract Preparation. Crude protein extract preparation fiom fungal cells was conducted as described in Chapter 2. Preparation and Purification of Polyclonal Antibodies. A nor-1 cDNA was cloned into the expression vector pMAL-c2 resulting in pMN 1 . The Nor-lc/MBP (fusion 101 protein) was produced in E coli transformed with pMNl. The Nor-lclMBP was cut with factor Xa to yield the Nor-1c protein and the MBP. The Nor-lclMBP and the Nor-1c protein were purified by afinity chromatogaphy as described in Chapter 2. The antigen-adjuvant mixture (100 pl of Hunter ’3 TiterMaxTM #R-l adjuvant plus 120 ug of the Nor-lclMBP in 50 ul of 10 mM phosphate-bufl‘ered saline) was prepared according to the manufacturer’s instructions (Cthx Corporation, Atlanta, GA) and was used for primary injection and boosts. The anti-Nor-lc/MBP serum of the rabbit was prepared according to Ausubel et al.(1995a). Briefly, the antigen-adjuvant emulsion was injected into the rabbit subcutaneously at four difi‘erent sites on the rabbit's back. The rabbit was boosted subcutaneously 4 weeks later after the primary injection and reboosted 2 weeks after the initial boost. The titer ofthe antiseum was estimated by indirect ELISA according to Ausubel et al.(l995a) using the Nor-lolMBP and the Nor-1c protein as coating antigens respectively. Briefly, a microtiter plate was coated with 10 ug/ml antigen and blocked with 0.25% (w/v) bovine serum albumin (BSA). The antigen-coated and BSA-blocked wells of the microtiter plate were incubated with serial dilutions of the antiserum (10 to 10° dilution). The wells were then incubated with goat anti-rabbit-IgG antibodies conjugated with alkaline phosphatase (Signa, St. Louis, MO). The activity of the captured alkaline phosphatase was determined according to the manufacturer's instructions. The dilution of the antiserum which showed 2 times the alkaline phosphatase activity of the preimmune serum in ELISA assay was defined as the titer. The IgG fiaction of the antiserum was precipitated with 33% (v/v) saturated ammonium sulfate (SAS). The double-purified polyclonal antibodies (PAb) were obtained by passing 102 a portion of the IgG fiaction twice through an afinity column containing MBP plus crude protein (10,000 x g supernatant) of E coli DHSa as ligands. The triple-purified PAb were obtained by passing a portion of the double—purified PAb through a second amnity column containing the Nor-1c protein as the ligand. Aflinity column preparation using CNBr-activated Sepharose 4B (Signa, St. Louis, MO) and ligand proteins (either a mixture of MBP and E coli DHSa protein in a 1:1 ratio, or Nor-1c protein alone) was carried out accordingtoapublishedstandardmethod(Ausubeletal, 1995c). Theprocedureforafinity pmificationofPAbwascaniedoutaccordingtothemethod established byDr. R. Mehighin our lab with modifications (Chapter 2, Immunoafiinity purification of the Nor-l protein). Five mg of the IgG fiaction (or the double purified PAb) (5 mg/ml) was loaded onto the aflinity column for purification of the double purified PAb (or the triple purified PAb). Western Blot Analysis of the Native Nor-1 Protein. Protein, separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), was blotted to a polyvinylidene difluoride (PVDF) membrane (Du Pont Co., Boston, MA) using a standard publishedmethod(Ausubeletal, 1995b). Irmmmodctectionwascaniedoutwitha dramhumnescandetecfionldt(Schleidrer&SdmdLKeeneNFDaccordingtoflle marmfactm’er’s instructions. Afiatoxin Analysis by TLC and ELISA. Aflatoxins were extracted for 1- h at room temperature with chloroform (10 ml) either fiom mycelial liquid gowth media (not including mycelia) (1 ml) or fiom mycelial solid gowth media (including mycelia) (1 g). Chloroforrn-extracted aflatoxins were tested either semiquantitatively by thin layer chromatogaphy (TLC) with chloroforrn-acetone (95:5) as the developing system, or 103 quantitatively by enzyme-linked irnmunosorbent assay (ELISA). For ELISA, only aflatoxin B1 (AFBl) was tested according to the method of Pestka et al. (1980) using anti- AFBl antibodies kindly provided by Dr. J. Pestka (Department of Food Science and Human Nutrition, Michigan State University, East Lansing , Nfl). Quantitative Assay of B-Glucuronidase Activity. Quantitative analysis of B- glucuronidase activity was performed by spectrophotometry as described by Jefi‘erson et al. (1986), and Tada et al. (1991) using p-nitrophenylglucuronide as the substrate. One unit was defined as the amount (nanomole) of p-nitrOphenol produced in one min at 37°C by 1 mg protein (nmol/min per mg protein). RESULTS Titer and Specificity of Polyclonal Antibodies. The titer of the anti-Nor-lc/MBP serum was determined by ELISA. The titers of antisera from the first and second boost were similar (450,000 and 500,000). The irnmunogen used for antibody production in this study was the Nor-lclMBP composed of the MBP (42 kDa) and the Nor-1c protein (31 kDa). Because the serum contained antibodies against both portions of the fusion protein, the titer which was determined using the Nor-lclMBP as the antigen was higher than the titer which was determined using the Nor-1c protein as the antigen. In Western blot analysis of crude fungal cell extracts, the IgG fiaction detected at least 7 protein bands (Figure 3-1). The double-purified PAb detected two protein bands (Figure 3-2). The triple-purified PAb detected only one major protein band (Figure 3-1) with a 104 ‘— Nor-l Figure 3-1. Western blot analysis demonstrating the specificity of PAb fiom different levels of purification. All crude extracts (except lane 5) were obtained from A. parasiticus SU-l grown in YES liquid medium. Each lane contained 10 [lg crude protein. Lanes: 1, crude extract (from the 60-h sample) was probed by the triple-purified PAb (10 [lg/ml); 2, 3, and 4, crude extracts from the 60-h, 54-h, and 48-h samples, respectively, were probed with the IgG fiaction (10 ug/ml); 5, crude extract from nor-l disrupted strain of A. parasiticus gown in YES liquid medium (for 60 h) was probed with the IgG fraction (10 uglml). The molecular mass of the Nor-l protein (top band) was 31 kDa. 105 Table 3-1. Titers and specificities of anti Nor-lc/MBP PAb. Samples Titer against the Nor- Titer against the Specificity lclMBP Nor-1c protein (cross reaction) Antiserum from primary injection 200,000 50,000 > 7 bands Antiserum from first boost 450,000 110,000 > 7 bands Antiserum fiom second boost 500,000 120,000 > 7 bands Double-purified PAb 2 bands Triple-purified PAb 1 band Notes: (I). The antiserum fiom primary injection was obtained 4 weeks afier the primary immunization. The antiserum fiom the first boost and the second boost were obtained 2 weeks after the first boost and the second boost respectively. (2). The titers of antisera were estimated using either the Nor-lclMBP, or the Nor-1c protein as the antigen. (3). The relative specificity was estimated by Western blot analysis. (4)ThespedfidtyoftheanfisamnwaseflnfledusingtheIgGfiacfioaneuanblot analysis. size of 31 kDa which was the expected size of the Nor-1 protein. The 31-kDa protein band was also detected by both the IgG fiaction and the double-purified PAb. The relative specificities of the IgG fiaction, the double-purified PAb, and the triple-purified PAb (as measured by the degee of cross reactivity) are summarized in Table 3-1. The Accumulation of the Nor-l Protein and the Accumulation of AFB1 in A. pm'asiticus SU-l Grown in YES Liquid Medium. The pattern of the accurmrlation of the Nor-1 protein was analyzed in A. parasiticus SU-l (an aflatoxin-producing strain) gown in YESliquidmedium(anaflatoxin-inducingmedimn). Westernblotanalysisofcrudeextracts fiom cells harvested at the appropriate time points showed that the Nor-1 protein was not 106 <— Nor-l 24h 36h 48h 60h 72h 84h 96h 108h Figure 3-2. Western blot analysis of the accumulation of the Nor-l protein in A. parasiticus SU—l gown in YES liquid medium. Each lane contained 10 ug crude protein. Lanes: 1 through 8, crude extract from firngal cells gown for 24 h, 36 h, 60 h, 72 h, 84 h, 96 h, and 108 h, respectively, and probed by the double-purified PAb (10 rig/ml). The molecular mass of the Nor-1 protein (top band) was 31 kDa. 107 detectedat24h ofincubation, butwasdetected from 36 hto 108 hofincubation (Figure 3-2). The maxirmrm level ofthe Nor-1 protein appeared between 36 h and 72 h ofincubation and decreased fi'om 72 h to 108 h ofincubation. ELISA analysis ofthe growth medium showed thatAFBr beganto accmnulateafier24hofinaibation(Figure3-3). Themaximurnrateof AFBraccunmlationoccurredbetween36hto60hofincubation (fiom 19to 121 mg/L). AFBl accmmrlationremainedatalmostthesamelevel fiom60hto 108 hofincubation. The maxirmrmrateofaccunmlation oftheNor-l proteinoccurredatthesametimeasthemaxinnnn rate ofAFBr accumulation. The Accumulation of the Nor-l Protein and the Accumulation of AFB1 in A. parasiticus SU-l in Nutritional Shift Assay. A nutritional shift assay was conducted to menarre the accumulation ofthe Nor-1 protein inA. pwasiticras SU-l gown in a nonaflatoxin- inducingPMSmediumandthenshifiedinto aflatoxin-inducingGMSmedium. Afier60hof incubation in PMS no Nor-l protein was detected by Western blot analysis (Figure 3-4). The Nor-1proteinwasdetectedasearlyas6hafierashififi’omPMStoGMS. Thecorresponding accumulationofAFBr inthegowthmedimnwasdeterminedbyEIJSACl‘able 3-2). Afierthe mrtritional shifi, AFBl increased more slowly inthe fresh PMS (fiom 0.13 to 2.42 mg/L) than inthefi'eshGMS (fiorn 0.13 to 129.80 mg/L). Inthenutritional shift assay, thepattern ofthe accumulation of the Nor-1 protein was consistent with the pattern of the accrmrulation of AFB1. 108 i .s ..a G O N O 90 N u-l 3 [iill[lilllliillllll|lllilllTTlllil Time vs AF 81 Time vs dry weight L l N 0 AF B1 production (mg/L) 8 Dry mycelial weight (glflask) I I I I I I 0 12 24 36 48 60 72 84 96108 Culture time (hour) Figure 3-3. The accumulation of AFB1 and mycelial dry weight in A. parasiticus SU-l gown in YES liquid medium. The concentration of AFB1 was assayed by ELISA in samples taken 12 h, 24 h, 36 h, 60 h, 72 h, 84 h, 96 h, and 108 h after inoculation. The mycelial dry weight was estimated at the same time points. 109 Table 3-2. The accumulation of aflatoxin Br in A. parasiticus SU-l in nutritional shifi assay. Incubationtimeaftera AFByinPMSafiershifi: AFBrinGMSaftershifl: shifl fi'om PMS (h) mean t SE (mg/L) mean :1: SE (mg) 0 0.13 10.09 0.13 $0.09 6 0.16 :t: 0.07 7.89 :t 0.16 12 0.59 :l: 0.06 25.36 i 0.25 24 1.06 :i: 0.07 45.47 :1: 0.28 48 2.12 :l: 0.08 115.36 :1: 1.23 60 2.42 :t 0.11 129.80 :t 1.26 Note: A. parasiticus SU-l was gown in a nonaflatoxin-inducing PMS medium for 60 h and then shifted into aflatoxin-inducing GMS medium. Each value represents the mean i standard error (SE) of triple tests. Morphology of A. parasiticus SU-l Grown on YES Agar. The morphology of A. musicals SU-l gown on YES agar was observed under a dissection microscope. Irmnediatelyafierinoculation, theareaoccupiedbytheinoculated sporeswasirregular. After one day of incubation, the germination of individual spores was randomly oriented. Afier two daysofincubation, awhiteroundcolony(0.5-l cmindiameter)appearedwithawhitecircular bandofsubstratehyphae(0.1- 0.2cminwidth)atthemarginandawhiteromldareaofaerial hyphae(0.3 -0.6cmindiameter)atthecentralregion. Afierthreedaysofincubation, the white circular band of substrate hyphae (at the margin of the colony) moved outwards followed byanadjacentwhitecircularbandofaerialhyphae(0.2-0.4cminwidth). Ayellow-geen roundareaofimmaturespores(l.4- 1.6 crnindiarneter) appeared atthecentralregion. After faudaysofhmbafionmewlfitedrmhrbarflofmbmhyphaeandflnadjacauwlnte circular band of aerial hyphae continued to move outwards followed by a yellow-green circular 110 ‘— Nor-l G6 G12 G24 G48 G60 P60 Figure 3-4. Western blot analysis of the accumulation of the Nor-1 protein in A. parasiticus SU—l in PMS-GMS nutritional shift assay. Each lane contained 10 pg crude protein and was probed by the double-purified PAb (10 rig/ml). Lanes: 1, 2, 3, 4, and 5, crude extract from cells collected alter incubation in GMS for 6 h, 12 h, 24 11, 4811, and 60 h (G6, GIZ, 624, G48, and G60), respectively, alter a shifi from a 60-h incubation in PMS (P60); 6, crude extract from cells collected afier a 60-h period of incubation in PMS (0 h in GMS). The molecular mass of the Nor-l protein (top band) was 31 kDa. 111 band ofinrrnature spores (1.0 - 2.0 cm in width). Abrown-geen round area ofmature spores (1.0-2.0cmindiameter) appearedatthecentralregionofthePetridish. Afierfivedaysof incubation, the white circular band of substrate hyphae reached the edge of the agar, the adjacanwhhedmdmbandofaaidhyphaeandtheydlow-geendraflmbmdofhmnamm sporesconfirmedtomoveoutwardsfollowedbyabrown-geencirmlarband ofmature spores (1.5-2.5 crninwidth). Thehyphaeinthecentralregionstartedtodegenerate. Aflersixdays ofharbafionthebmdofwbsfifiehyphaemdthebandofaaialhyphaedisappearedThe bandofimmaturesporesandthebandofmature sporesmoved outwardsandabrownround areaofdegenelatedhyphae(0.2- 0.5 crnindiarneter) appearedatthecenter. Morphological Development and the Accumulation of AFB1 at the Colony Level in A. paradtiaasGrownonYESAgar. SporeswereinoculatedontothecenteronESagarina Petri dish and samples (C2, C4, C5, C6, M4, M5, M6, P5, and P6) were collected as described (Materials and Methods) and shown schematically in Figure 3-5B. The accunmlation of AFBl in each sample was determined by ELISA. The morphological characteristics and the accurmllation of AFB1 in each samples were summarized in Table 3-3. By comparing the morphological characteristics with the accumulation of AFBl, a pattern was found. In the gowing colony, the concentration of AFBl increased from the region with substrate hyphae plus aerial hyphae (3.2 mg/g), to the region with substrate hyphae plus aerial hyphae plus irmnature spores (4.3-4.7 mg/g), to the region with irmnature spores plus mature spores (5.8- 7.4 mg/g), to the region with mature spores (7.4—9.0 mg/g), and to the region with mature spores plus degenerated hyphae (7.9 mg/g) in a colony. The pattern of the accumulation 112 Table 3-3. The morphological characteristics and the accumulation of AFB1 in A. pm'as'iticus SU-l gown on YES agar. Samples Collecting Incubation Morphological characteristics AFBl region time(day) (mm CZ Central 2 Substrateandaerialhyphae 3.2:t0.8 C4 Central 4 Immatureandmaturespores 5.8:0.4 C5 Central 5 Mathew 7.4:03 C6 Central 6 Maturespores, anddegemratedhyphae 1910.7 M4 Middle 4 Wandaerialhyphae,andimmatmegxxes 4.31:0.5 M5 Middle 5 Inunatureandmaturespores 7.43:0.6 M6 Middle 6 Maulregtores 9.01: 1.2 P5 Peripheral 5 swarmeandauialhyphmandimmatmeqrores 4.7:tO.4 P6 Peripheral 6 Imrmtureandmatmespores 6.91:0.3 Notes: (1) Conidia were inoculated at the center of YES agar in a Petri dish. Samples: Samples C2, C4, C5, and C6 were collected fi'om the central region 2 days, 4 days, 5 days, and 6 days after inoculation; samples M4, M5, and M6 were collected from the middle region 4 days, 5 days, and 6 days afier inoculation; samples P5, and P6 were collected from the peripheral region 5 days, and 6 days alter inoculation. A schematic of the sample collection procedure is shown in Figure 3-5B. (2) The accumulation of AFB1 (mg/g dry filngal mass) was determined by ELISA (Materials and Methods). (3) Each value represents the mean i standard error (SE) of three independent tests. of AFB1 in the gowing colony was correlated to the morphological development of sponrlation in A. parasiticus SU-l. The Accumulation of the Nor-l Protein, the Activity of the nor-1 Promoter, and the Accumulation of Aflatoxin in A. parasiticus SU-l Grown on YES Agar. Fungal spores were inoculated onto the center of YES agar in a Petri dish. Samples (C2, C4, C5, C6, M4, M5, M6, P5, and P6) were collected as described (Materials and Methods) and shown schematically in Figure 3-5B. Western blot analysis of crude mycelial extracts 113 Nor-l P6 P5 M6 M5 M4 CG C5 C4 C2 P5, P6 C4, C5, C6 C2, C4, C5, C6 I a Figure 3-5. Westem blot analysis of the accumulation of the Nor-l protein in A. parasiticus SU-l gown on YES agar. Each lane contained 10 ug crude protein and was probed with the double-purified PAb (10 rig/ml). A: Lanes: C2, C4, C5, and C6, samples collected from the central region 2 days, 4 days , 5 days , and 6 days afier inoculation; M4, M5, and M6, samples collected from the middle region 4 days, 5 days, and 6 days after inoculation; P5, and P6, samples collected from the peripheral region 5 days , and 6 days afler inoculation. B: A schematic of the sample collection procedure. 114 «AFB1 e—AFG1 C2 C4 CS C6 M4 M5 M6 P6 P5 Figure 3-6. TLC analysis of the accumulation of aflatoxins in A. parasiticus SU-l gown on YES agar. Aflatoxins from each sample (mycelia and agar) were extracted with chloroform and assayed by TLC with chloroform-acetone (95:5) as the developing system. Fluorescence was detected with long wavelength UV light. A. Lanes: C2, C4, C5, C6, M4, M5, M6, P5, and P6 are samples collected as explained in the legend of Figure 3-5. 115 Table 3-4. .3932 3a ”3382 E 393mg ..3 3:383 803 83 8233 cc 5803 7.82 2: mo 333$ 023$ 05 3a 0.5. no 55. me $885 038.2 2:. Amv sense: e5 6636 <23 .3 season... as 83388 in... 3 A5805 we be 58385 .388 m8 _ .3 use». 3 £8 25 E 30:35 33323.50 3.08285 250:8 05 8 3.5% mg «we: 25 63:38 05 8 evacuees—waeofiofie .& men: 283.? 3 3a.“. 98 A3236 3 580.3». .3 3n33e we bfisouofieaoonm 3 35.83% 83 33:8 mDG A9 .33“ “33335 8.5 we Ammv Coho vac—Be. H :38 2: A32. £38m 782 38x3 $3352 2...? seem ANV .mm .m oaewfi E ESQ—m mm 8:385 330:8 295m 2: Co 03823 < .3353: 28 $332 5 ...—303 ease—=85 .53 «use 0 3a .943 n common Eco—Eton 2: 80¢ 33:8 033 e.— 28 .mm 3382 Hose—305 .63 as e 3a is m .93 v 5&8 22:8 2: 88% 33:3 203 e2 e3 .32 ...: 338nm Bose—305 33 996 e 3a .996 m .33 v .33 N common .833 05 Sec 386:8 203 90 3e .mU JO .NU 8.95m u3.95am ..3—c 3m a 5 came mm> .«o 82.8 05 an 33:83 063 £350 a "332 $3 an an 82 an a Zn 2 ”new 3.. 2. when 32 38 33853.29: 55. no a .3 to a 5. S a Se 3. a I. ....c a 3. he a as no a 3. ed a an no a «a $5 5:22:32. .95. 3:: 38. : «a: one: 242 we 2 : a 3 2 a a: a new miniseefl 22.8 £5 a an a 8. S 2 a. 2 : so... 5833 ..e 3.5.... 9529.: £89:— 782 c.— 2 32 m2 .2 so no 5 8 8.95m «awe mm; :o 859% 73m maniacach .V E .92 mo cog—3:508 05 23 65333 3:3 .88an 7.6: 05 Co 3338 05 .5895 7.82 05 .«o ease—3830a 2E. arm 63:. 116 showed that the Nor-1 protein (31 kDa) was present in all the samples (Figure 3-5A). In the central region, the Nor-1 protein appeared 2 days after inoculation (C2), reached the highest level 4 days after inoculation (C4), and declined 5 days and 6 days after inoculation (C5 and C6). In the middle region, the Nor-l protein appeared 4 days alter inoculation (M4), and increased 5 days and 6 days after inoculation (MS and M6). In the peripheral region, the Nor-1 protein appeared 5 days after inoculation (PS), and increased 6 days after inoculation (PG). For each sample, the GUS activity was measured spectrophotometrically. The accumulation of aflatoxins was determined by TLC (Figure 3-6). The relative intensities of the Nor-l protein in Western blot and AFB1 on TLC were determined with an EPSON ES-IOOOC scanner (EPSON Accessory, Singapore) and quantitated with Sigma Gel software. These results (along with the accumulation of AFB1 measured by ELISA) are listed in Table 3-4 to facilitate comparison. The quantity of AFB: in ELISA analysis and the relative quantity of AFB1 in TLC analysis in each sample was consistent with the relative quantity of Nor-l protein in Western blot analysis (Figure 3-5), and was consistent with the level of nor-l promoter activity in the GUS assay. DISCUSSION The data presented in this study demonstrated that the PAb raised against the Nor- lc/MBP were suficiently specific for detection of the (native) Nor-1 protein by Western blot analysis. The data showed that the accumulation of the Nor-1 protein (in liquid media and on solid media) and the activity of the nor-l promoter (on solid media) are consistent with the pattern of the accumulation of aflatoxin and are closely correlated to the morphological development of sporulation in A. parasiticus. The data also suggested that 117 the expression of nor-l is regulated in a temporal and spatial pattern in A. pm'asiticus grown on solid media. The first step in this study was to prove that anti-Nor-lc/MBP PAb were suficiently specific to detect the Nor-1 protein in Western blot analysis. This step was necessary not only because the imrnunogen used in this study was a fission protein composed of the Nor- lc protein (instead of the Nor-l protein) and MBP, but also because the fusion protein was likely contaminated with the bacterial protein. It was not surprising that the antiserum specificity was low (indicated by the cross reactivity in Western blot analysis). Therefore the antiserum could not be used in Western blot analysis of the Nor-1 protein without validation. The only major protein band detected by the triple-purified PAb not only had the expected size of the Nor-l protein (31 kDa) but also was detected by the IgG fi'action and the double-purified PAb in crude mycelial extracts fi'om the wild-type A. parasiticus SU—l. The same protein band was not detected in crude mycelial extracts from the nor-l disrupted strain. Therefore the double-purified PAb (or even the IgG fiaction) could be used to monitor the accumulation of the Nor-l protein in Western blot analysis. A 28-kDa protein band was also detected by the IgG fraction and the double-purified PAb, but was not detected by the triple-purified PAb. One possible explanation is that the 28-kDa protein and the Nor-l protein have similar antigenicity. One fraction of the total PAb (raised against the Nor-lc/MBP) may have a higher afinity to the 3 l-kDa Nor-1 protein and a lower afinity to the 28-kDa protein. Another fraction may have a lower amnity to the 3 l-kDa Nor-1 protein but a higher amnity to the 28-kDa protein. Therefore, if an excess of the double-purified PAb was used for further purification using 118 amnity chromatography, the binding sites for the Nor-1c protein on the afinity column could be saturated by the PAb fiaction which had the higher afinity to the 31-kDa Nor-l protein. This fi'action was then purified (triple-purified PAb). This could be one possible reason that the double-purified PAb detected both the Nor-1 protein and the 28-kDa protein, but the triple-purified PAb detected only the Nor-1 protein in Western blot analysis. The pattern of the 28-kDa protein accumulation was consistent with the pattern of the Nor-l protein accumulation. It is possible that the 28-kDa protein is also involved in the AFB1 pathway. The 28-kDa protein and the Nor-l protein may be synthesized at the same time and combine with each other in order to increase the eficiency of aflatoxin synthesis. The combination of the two proteins may stabilize the 28-kDa protein. Therefore, when the nor—1 gene is disrupted, there is no Nor-l protein existing and the 28- kDa protein is rapidly degraded. The temporal pattern of the accumulation of the Nor-1 protein and the accumulation of AFB: are closely correlated in A. parasiticus grown in liquid medium. In batch fermentation (YES liquid medium), the accumulation of the Nor-1 protein and the accumulation of AFB1 increased simultaneously and quickly during late log-phase of the fiingal growth and reached a maximum level at stationary phase. The level of the accumulation of the Nor-1 protein declined at approximately the same time that the accumulation of AFB1 reached its peak level. In the nutritional shift assay, the accumulation of the Nor-l protein and the accumulation of AFB1 began simultaneously and increased quickly a short time after the ll9 shift fi'om the nonafiatoxin-inducing PMS medium to the aflatoxin-inducing GMS medium. The Nor-l protein accumulation data were consistent with the nor-1 transcript accumulation data previously reported (Skory et al., 1993; Trail et al., 1994; Trail at al., 1995). In PMS medium, no Nor-1 protein was detected, but AFB: was detected. This apparently inconsistency may be explained in two possible ways. In the nutritional shift culture, the AFB: concentration in PMS medium was much lower than in GMS medium after the shift. Therefore, the corresponding Nor-l protein in PMS could be too low to be detectable in Western blot analysis in this study. The second possible explanation may come fi'om the result of nor-l disruption. The disruption of the nor-l gene resulted in an accumulation of norsolorinic acid (NA) and a significant decrease in aflatoxin accumulation (Trail etal, 1994). Thisresult suggeststhatasmall quantityofAFB: couldbe synthesizedandacctmmlatedwithoutthefimction oftheNor-l proteininPMS medium. The data from liquid batch fermentation and the nutritional shift assay confirmed the observed expression pattern of the nor-l gene (at the level of transcript accumulation) in A. pamsiticus grown in liquid media previously reported (Skory et al., 1993; Trail at al., 1994; Trail at al., 1995) and demonstrated that the accumulation of the Nor-l protein can provide an alternative measurement for the expression of nor-l. Therefore, it is also reasonable to predict that the accumulation of the Nor-l protein can be used as a measurement for the expression of nor-l in A. parasiticus grown on solid media. The accumulation of the Nor-l protein was higher at 72 h than at 36 h in YES liquid medium, but the rate of AFB1 accumulation between 60 h and 72 h was much lower than 120 between 36 h and 48 h. One possible explanation is that at 36 h most of the Nor-1 protein was active, while at 72 h most of the Nor-l protein was inactive possibly because of the absence of NADPH (Chapter 1, Regulation of aflatoxin biosynthesis) or other unknown factors (Chapter 2, Discussion). An alternative explanation comes fi'om the observation that the A. flaws group is not very eficient at degrading aflatoxins under normal conditions (Doyle and Marth, 1978). It is possible that when aflatoxin concentrations reach high levels in the medium, fungal cells are forced to eficiently degrade aflatoxins to survive. AFB: could be synthesized at the same rate in the period fi'om 60 h to 70 h and inthe period from 36 h to 48 h because the similar accumulation of the Nor-1 protein was observed in the two periods of time. But in the period fi'om 60 h to 70 h, a certain amount of AFB: was possibly degraded. This degradation could result in the much lower rate of AFB: accumulation. The morphological development at the colony level of A. parasiticus grown on solid media in a Petri dish appears to be controlled by genetics and environmental influences. Here, morphological development at the colony level is defined as the development of the shape and color of a colony and distributions and colors of substrate hyphae, aerial hyphae, immaturespores, maturespores, anddegenerated hyphalcellsinthecolony. Regardlessofthe irregulargeometricalshapeoftheareaocwpiedbytheinoculated sporesandtherandom orientation of the germination of individual spores, the resulting colony and the concentric regions of distinct morphology in the colony are almost always nearly circular. Nutrients in YESagarareevenlydistributed. Nutrientsincoconutagararenotalwaysevenlydismbuted becauseitmaycontainlargepiecesofcocomrtflakeswithdifi‘erentsizes(Chapter4).The 121 nearly circular shape of the colony and the concentric regions of distinct morphology in the colonyappearedonbothmedia. Butthesetwomediadohavedifi‘erentinfluencesonthe colors and widths of concentric circular regions of distinct morphology in the colony. These observations suggested that the morphological development of A. parasiticrrs is controlled by genetic and environmental influence at the colony level. In another words, morphological developmentappearstoberegulatedatthecolonylevel. Sporulation is one example of morphological development. The Aspergillic m‘dulansfluG geneisnecessaryforthesynthesisofasmalldifirsiblefactorwhichmaybeanextracellular sigrfldirecfingasemdsponflafionmdpehapsomeraspecmofmlonygrowmaeeand Adams, 1994a, 1994b, and 1996). This observation suggests that sporulation is regulated at the colony level by cell-cell communication. The accumulation of the Nor-1 protein, the activity of the nor-l promoter, and the accumulation of aflatoxin are consistent with each other and are possibly regulated at the colony level through an unknown mechanism which is closely related to the morphological development of sporulation in A. parasitic-us grown on YES agar. Because of the inherent dificulties encountered in the separation of substrate hyphae, aerial hyphae, immature spores, and mature spores, the procedure of collecting samples from YES agar used in this study was a crude approach to achieve this separation. Nevertheless, the accumulation of the Nor-l protein and the activity of the nor-1 promoter (indicated by GUS activity) increased and decreased at the same time (temporal level) in A. pw'asiticus SU-l grown on YES agar. Higher levels of the Nor-l protein accumulation and the nor-l promoter activity were detected in regions containing conidiophores and immature conidia than in 122 regions containing only substrate hyphae and aerial hyphae (spatial regulation). Decreased levels of the Nor-l protein accumulation and the nor-l promoter activity were observed in regions containing only mature spores and degraded hyphal cells. In the center of the colony, the accumulation of AFB: increased (per unit of dry weight) at the same time that the accumulation of the Nor-l protein (or GUS protein) decreased. This phenomenon may result fi'om the fact that AFB: is much more stable than the Nor-l protein. The value for AFB: accumulation likely represents a true measurement of accumulative. The value for Nor-1 protein accumulation represents synthesis and degradation. In the center of the colony, when degenerated hyphae were not able to synthesize new Nor-l protein, the accumulation of the Nor-1 protein decreased but the accumulation of AFB: still increased. These patterns were consistent with the pattern of the relative intensity and distribution of blue color (indicating the activity of nor-l/GUS) shown on a colony using the same nor- l/(RIS reporter construct with S-bromo-4~chloro-3 -indolyl glucuronide as the substrate (Chapter 4). Also consistent with these observations, immunolocalization data presented later in this dissertation (Chapter 4) showed that more Nor-1 protein accumulated in conidiophores and conidia than in vegetative cells. Further, when a sporulation related gene (fluP) was disrupted in an aflatoxin-producing strain of A. parasiticus, fungal hyphae gown on solid media did not sporulate and AFB: remained at the same low level during a 6—day period of incubation (Chapter 5). Because sporulation appeared to be regulated at the colony level and a close correlation between sporulation and the accumulation of aflatoxin (as well as the accumulation of the Nor-1 protein and the activity of the nor-1 promoter) was observed, the accumulation of 123 the .Nor-l protein and the activity of the nor-l promoter may also be regulated at the colony level. The correlation between aflatoxin biosynthesis and sporulation in toxigenic A. pausitr’cus needs further study. Certain sponrlation-related pignents with the basic structure of norsolorinic acid were detected in the Awergillus genus (Steyn et at, 1980). Spore pigments from A. nidulans and A. parasiticus also appear to be produced via polymerization of polyketide precursors (Brown et al., 1993; Brown and Salvo, 1994). A development-related gene wA in A. nidulans possibly encodes a polyketide (Mayorga and Timberlake, 1992). The A. parasiticus polyketide synthase gene pksA is required for AFB: biosynthesis (Chang et al., 1995). Therefore, it is possible that a common signal(s) at an early stage of development triggers both aflatoxin synthesis and sporulation. When sporulation is inhibited in liquid media, only aflatoxin synthesis occurs in A. parasiticus. When the mechanism of aflatoxin synthesis is absent in nontoxigenic fungi or mutation(s) of the genes involved in aflatoxin synthesis occurs in toxigenic fungi, only sporulation happens- In summary, the data presented in this study showed that the accumulation of the Nor- ] protein (in liquid media and on solid media) and the activity of the nor-1 promoter (on solid media) are regulated in the same pattern as the accumulation of aflatoxin and are closely correlated to the morphological development) of sponrlation in A. parasiticus. These results also suggested that the expression of nor-1 is regulated in a temporal and spatial pattern in A. parasiticus gown on solid media. CHAPTER4 LOCALIZATION OF NOR-l PROTEIN ACCUMULATION AND LOCALIZATION OF nor-1 PROMOTER ACTIVITY DURING AFLATOXIN Bl BIOSYNTHESIS IN ASPERGILL US PARASITICUS ABSTRACT: Anti-Nor-lc protein polyclonal antibodies were used in immunolocalimtion experiments to localize the accumulation of the Nor-l protein (at the cellular and 11pr level) and a nor-l promoter/GU S (nor-l/GUS) reporter construct was used to localize the activity of the nor-1 promoter (at the hyphal and colony level) during aflatoxin B1 biosynthesis in Aspergillus pamsitr'cus gown in liquid and/or on solid gowth media. The Nor-l protein was mainly localized in the cytosol of vegetative hyphae (although a sigrificant quantity appeared to be associated with particles of unknown composition) either in liquid gowth medium using cell fiactionation/Western blot procedure and in situ innmmolocalintionprocedmesorfiomsolidgowthmedimnusinganinsim imrmrnolocalization procedure. On solid gowth media, the highest level of the Nor-1 protein wasassociatedwithimmatureconidia higherlevelswereassociatedwithconidialstalkslower levelswereassociatedwithvegetativehyphae, andnonewasassociatedwithmatureconidia(at thehyphallevel). Onsolidgrowthmediimthehighestlevelofflreactivityofthemr-l promoter was detected in conidial heads, higher levels in conidial stalks, and lower levels in vegetative hyphae (at the hyphal level). At the colony level, the activity of the nor-1 promoter 124 125 wasmtdetededhregonswmaMngomymbsuatehyphaeormatumconidiaMW detected in regions containing aerial hyplne (higher levels) and/or irmnature conidia (the highest level). These data mggest that localization of Nor-l protein accumulation and localization of nor-l promoter activity are closely correlated to the process of sporulation in a colony. Therefore the regulation of nor-l gene expression may be regulated at the colony level dmingaflatoxinbiosymlresisirrApar-asiticusonsolidmedia. INTRODUCTION Manystudieshavebeenconductedwithagoal ofunderstandingthegenesandtheproteins involved in afiatoxin biosynthesis (Chapter 1). Little effort, however, has bear directed toward definingwhereaflatordnbiosymhesisoccursatthecelhrlarlevel, atthehyphal level (vegetative hyphae, conidial heads, conidial stalks, and conidia), and at the colony level (regions in a colony corrtairringsubstatehyphaeaerialhyphaeimmamreconidia, and/ormatureconidiaina colony). SeveralenzymeactivitiesinvolvedinaflatordnB: (AFB1) syntlresishavebeendetectedin the cytosol while others are associated with organelles (Bhatnagar et at, 1989; Yabe et at, 1989; Yabe et at, 1993; Yabe and Hamasaki, 1993; Matsushima et al., 1994). These data wggwmmmfiomhedsomedniscaniedommdifimwmrhrwmpamm Therefore, aflatoxin pathway enzymes or intermediates may be transferred fi'orn one cellular compartment to another during aflatoxin biosynthesis. Westernblotanalysesandnor—l/GUS assayspresentedinChapterB suggestthatthe regulation of Nor-1 protein accumulation and the regulation of nor-1 promoter activity are 126 closelycon‘elatedtotbeprowssofspomlationatthecolonylevel. Anunderstandingofthe localintion ofNor-lproteinacarmulationandthelocalimtion ofmr-l promoteractivitymay remhmabeaamdastmdingofflrewndafionbaweardresemolocdimfionsandthe processofspomlationduringaflatordnbiosynthesis. T'hisinformationmaydirectlyorindirectly leadtothedevelopmentofsafe, specificfirngicidesornaturalplantproductswhichcanblock flnmuacdhdunansfaofAFBrpathwayenzymesorhnamedimesfiomomwnmamnmtto another,md/orhnafaewiflrflnregdafionofaflatordnbiosyndrefisatthewlonylwdmdin truninhibitAFBrbiosynthesisinpr'asr'tims. Twohypothesesareproposedinthisstudy:(l)theNor-lproteinresideslocallyincertain organelles or in the cytosol; (2) localization of Nor-l protein accumulation and localimtion of nor-l promoteractivityareclosely correlated to theprocessofsponrlationduringaflatordn biosyrrthesisinpr'au'ticw. ThedfiapresemdmChapta-BflsodanonsuatedthmmepolydonflmmbodiesGAb) raisedagainsttheNor—lc proteinweresuficiently specifictodetectthenativeNor—l protein and that regulation of Nor-l protein accumulation and regulation of nor-1 promoter activity wereconsistentwiththepattemofAFBraccunmlation. Thereforetwogoalsareproposedin thisstudy:(l)localizetheaccurmrlationoftheNor-l proteinattlrecelhrlarlevelandatthe hyphallevelusingthePAbraisedagainsttheNor-lcprotein;(2)10calizetheactivityofthe nor-1 promoter at the hyphal level and at the colony level using a nor-UGUS reporter construct. Thelocalizationdatapresented inthis study demonstrated that the Nor-l proteinwas mainly localized in the cytosol of vegetative hyphae. The highest level of the Nor-l protein was 127 associated with immature conidia, higher levels were associated with conidial stalks, lower levdswaeassodaedwithvegaafivehyphaeandmnewasassodaedwnhmaunewmdia The highest level ofnor-l promoter activity was detected in conidial heads, higher levels in conidial stalks, and lower levels in vegetative hyphae. At the colony level, the activity of the nor-l promoter was not detected in regions of a colony containing only substrate hyphae or matumconidiahnwasdeteaedmregonscomairfingaaialhyphaeafighalevds)or immature conidia (highest level). These data suggest that localization of Nor-l protein accumulation and localimtion of nor-1 promote activity are closely conelated to the process of sponflafionandhmcefliesepmcessmaybemgdatedamewlonylwddufingaflatordn biosynthesis in A. pw-asiticus on solid media. MATERIALS AND METHODS Fungal Strains and Culture Media. Awergilhzspamsificus NRRL 5862 (ATCC 56775, SU-l) was used as the aflatoxin-producing wild-type strain. An A. manuals strain, nor- 1/GUS, which contains the nor-llGUS reporter construct (Chapter 3, Materials and Methods), was used to monitor nor-l promoter activity. Fungal strains were maintained as fiozen spore stock suspensions in 20% (v/v) glycerol at -80°C (Chapter 2, Materials and Methods). Aflatoxin-inducing liquid gowth medium consisting of 2% (w/v) yeast extract, and 6% (w/v) sucrose (pH 5.8) (YES broth), and solid gowth medium YES agar [YES broth plus 1.5% (w/v) Bacto—agar] were used to gow fungal cells for localization of Nor-l protein accurmrlation and for localintion of nor-l promoter activity. 128 Fungal Culture. For culture offirngal colonies on solid medium, 5 ul ofa stock spore suspension (1.5 x106 spores/ml) was inoculated onto the center onES agar and incubated at 29°Cinthedarkforappropriateperiodsoftime. Forfirngalgowthinliquidmediurrtaspore stockwspmsimwasmoadatedhnoYESbrothwafimlspomconcanrafionoflfixw’ spores/ml. Thewlunewashrarbatedat29°Cwithconstamflnldng(150rpm)mflredarkfor appropriateperiodsoftime. Myceliawerecollectedbyfiltration. Forirnrrarnolocalization,the collectedmyceliawereusedinrrnediately. ForWestanblotarmlysisthecollectedmycelia wacdfluusedhmnedimdyorfiomundafiqifidmmgmmdstoredat-SWCunfiluse For fimgalshdeufluneflremethodaccordhigtoflanis(l986)wasmodifiedasfoflows A mia'oscopeslidewasplacedonflrecerna'ofaPenidishcomairnngYESagar. Twoblocksof YESagar(appmrdnmdy200nnn3)waephcedattwosepmatelocafionsontheslide A vohrmeof2ulofsporestocksuspension(l.5xlO‘spores/ml)wasinoarlatedoneachsideof theYESagarblockexceptthebottomandthetopsidesfollowedbyplacemartofacover glassontopoftheYESagarblock. 'I‘lreslideswereincrrbatedat29’Cforappropriateperiods oftinreinthedark. Theslidesandthecoverglasseswithhyphaeattachedwereused immediatelyforirmmmolocalimtionstudies. Cell Eactionation. Two protocols were used for cell fractionation. Protocol 1. Cell breakage by Potter homogenization of protoplasts followed by differential centrifugation. Thisprotocolwascaniedoutat4°C according toMulleretal(1991)with certainmodifications. Briefly,mycelia,gowninYESliquidmedimn,wereharvestedby Monmfifiereaflfighyphflmflwaswadwdmdcewimmfingmfl‘awndsfingofom M Tris-Cl (pH 8.0), 0.14 M NaCl, and 0.025% (w/v) NaN3 (T SA). Portions (20 g wet weight) 129 ofthewashed hyphalmatwereresuspended in 100 nrl ofcell-wall-digestion bufl‘er consisting of 1.5% (w/v) Novozyme 234, 0.1 M citric acid (pH 6.0), and 0.6 MKCl for 4 h at 30°C with gentleshaking. Theprotoplastswerecollectedbypassagethrougha29nnnnylonmeshand washed three times with protoplast-washing bufl‘a' consisting of 2-(N-morpholino) ethane sulphorric acid (pH 7.5) and 0.6 M KCl (MES). The resulting protoplasts were resuspended with modified MES supplemented with 1.0 mM phenylmethylsulfonyl fluoride (PMSF), 0.4 nypsininln‘bitionmrit/mlaprofininSdeithiotlneitoLandO.6MKCL Theresuspended protoplastsweredisruptedbyaPoflerhomogerfizedAuflrerHThomasCo.) Celldebriswas removedbycentrifirgationtwiceatl,000ngor 10min. Theresultingcelllysatewas desigrated the PH attract (obtained by Potter homogenization). The PH extract was fiacfionfledflodifi‘aanpmddefiacfiomandafinalwpanmamfiacfionbydifi‘amfial centrifirgation. The1,000xgpelletfraction(PL)wasobtainedbycentrifirgationofthePH extractatl,000ngor20minfollowedbythreewasheswithl\4ES. Tasmanian (NU)wasobmmedfiomthepoa-l,000xgmpanmmfiacfimbycamifirgafimm6,000xg for20minfollowedbythreewasheswithMES. ThenntochondnalfiacfionMDwas obtainedfi'omthepostnuclear supernatant fiactionbycentrifiigationfor 30minat20,000xg followedbythreewasheswithMES. Thelysosomalfiaction(LS)wasobtainedfiomthe postmitochondrial supernatant fiactionbycentrifirgation for 30 min at 50,000 ngollowedby threewasheswithMES. ThemicrosomalfiactionMC)waspreparedfi'omthepostlysosomal supernatantfiactionbycentriflrgationforlhat100,000ngollowedbythreewasheswith MES. The rrbosomal fiaction (RS) was prepared fiom the postmicrosomal supernatant fi'action by centrifugation at 140,000 x g for l h followed by three washes with MES. The 130 postribosomalsupernatamfiomthisstepwassavedasthe 140,000 x gcytosol fiaction (CS). Forproteinconcentrationdetermination,aknownvolumeofeachfiactionwasmixedwithan equal vohrrne of 4% (w/v) sodium dodecyl sulfate (SDS) bufl‘er and boiled for 5 min. Then drewmemafimwasdeenfinedwithmeBieRadpmtenassaykiflBieRadIabomofies, Hecules, CA) according to the mamfacmrer’s instructions. PmtoeolLSeparationoforganeflesandtbecytosolfractionbypmtoplast permeabil'nation. Protoplasts were prepared as described above. Permeabilization of protoplasts and separation of the organelle and cytosol fractions separation wee conducted at 4°C accordingtothemethodsodea etaL (1993). Briefly, protoplastswerewashedusing thefollowingschenezaSminwashwithbufl’el [250mMsorbitoL20mMHEPES-KOH (pH6.8), 150mMpotassirmracetatemrd5mMmagresiumacetate];a3nfinwashwnhbufi'e 2(50mMsorbitoLand5anagresiumacetate);a5minwashwithbufi‘e3(2.0M pomsimnaceateZSOmMsorbnoLandSangredmnaceate);andaSnnnwashwith bufl‘e4(250'mMsorbitol,and5mMmagnesiumacetate). Afieeachwashthecellswere harvestedbycentrifirgationfor45 secat13,000xg. Thecombinedsupematantsfiomthefour washesweethecytosolfiacfionmdtherenflfingpdlesweetheorgmeflefiacdon The proteinconcentrationofeachfiactionwasdeterminedasdescribedabove. Thesetwofiactions werecalledthePPextracts(obtainedfiompemeabilizedprotoplasts). ’ WesternBlotAnalysisoftheNor-l ProteininCellFractions. Proteinsineachcell fi'action wee separated by SDS-polyacrylamide gel electrophoresis (PA(E) and blotted to a polyvirrylidene difluoride (PVDF) membrane (Du Pont Co., Boston, MA) using standard published methods (Ausubel er al., 1995b). The double-purified anti-Nor—lc protein polyclonal 131 mfibodies(PAb)(Chapte3)weeusedasthepfinmryanfibodymWeuenblmanalysis hummodeecfionofthepfimmyarmbodywascmfiedomthachenfihmnnescendeecfion kit (Schleiche & Schuell, Keene, NH) according to the mamrfacturer‘s instructions. The rdafivethsityofNor-lpmtdnheachhnehWeflenbManalysiswasdderfinedby scanning the X-ray autoradiograph with an EPSON ES-lOOOC scanner (EPSON Accessory, Singapore)andquantitatedwithSignagelsoftware. AnalysisofMarkerEnzymeActivities. Themeasm'emertoftheactivityofsuccinate dehydrogenasewascarried out according to Singe eta]. (1965) withmodifications. This mahodwasongrrallyusedwmethewtdmnmmwmchebytheuseofmercess unknownamomrtofsuccinatedehydrogenase. Inthecurrentstudy,thereactionwasusedto deennnemcdnatedehydrogenaseacfivitybyflreuseofanercessknownmnwmof succinate. Thereactionwasoptimizedandcarriedoutfor30and60minatroomternperature inatotalvolumeof3mlconsistingof0.5mlof0.3MTrisbufi‘e(pH7.6),0.1mlof0.lM potassiumfenicyanide, 0.3 nil of 0.01M succinate, and 0.05 mg of protein. The optical density ofthereactionmixturewasmeasuredat450nm. Oneunitofsuccinatedehydrogenasewas definedasflreamoumOfenzymecausingadeeeaseoflnnnolewcdnatemlmhr. Themeasurement ofcatalase activitywasconducted asdescribedbyCohen etal (1970). Thereactionwasoptimizedandcaniedoutat4°Cfor1,5,and20mininatotalvolumeof9 ml consisting of] nrl of 2% (v/v) H202 solution in 0.01M phosphate bufi‘e (pH 7.0), and 0.02 mgofproteirr. Thereactionwasstoppedbyadditionoflmlof6NH2804followedby additionof7mlof0.01NKMnO.. Theresidualeozwasmeasuredbymeamring 132 absorbanceat480nm. Oneunitofcatalasewasdefinedasthearnountofproteinrequiredto liberate halfthe peroxide oxygen from a hydrogen peoxide solution in 100 s (Luck, 1965). The activity of acid phosphatase was measured with a commercial assay kit (Signa Chenical Co., St. Louis, MO) according to the manufacture’s instructions. The reaction was carried out for30minat37°C inatotalvolumeofS ml consistingof0.5 mlof4mg/mlp- nitrophenyl phosphate, 5.0 ml of 0.1 N NaOI-I, and 0.02 mg of protein. The measurement is based on the hydrolysis of p-nitropherryl phosphate by the acid phosphatase yielding p- nitrophenol. Under alkaline conditions, p-nitrophenol is converted to a yellow complex readily nreasured by absorbance at 410 nm. The activity of acid phosphatase (Signa units/ml) was calculated according to the manufacturer’s instructions. The activity of glucose—6—phosphatase was measured according to the method of Harpe (1965) with modifications to optimize the reaction conditions. The reaction was canied out at 37°C for 30 min in a total volume of0.3 ml consisting of0.1 ml ofO.l M citrate buffer (pH 6.5), 0.1 ml of 0.1 M glucose-6-phosphate, and 0.03 mg of protein. The reaction was stopped by addition of 1 ml of 10% (w/v) trichloroacetic acid. After centrifugation, 1 ml of supernatant was mixed with 5 ml of2 rnM molybdate solution and 1 ml ofreducing agent (40 mM 1- amino-Z-naphthol-4-sulphonic acid) for 15 to 30 nrin at room temperature. The optical density was measured at 660 nm. One unit of glucose-6-phosphatase was defined as the quantity of enzyme required to liberate 1 mole phosphate fiom glucose-6-phosphate in l min. The activity of glucose-6—phosphate dehydrogerrase was carried out according to Niehaus and Dilts (1984). The reaction was canied out at 30°C from 1 to 30 nrin in a total volume of 2.5 ml consisting of 2.2 ml of 0.5 mM glucose-6-phosphate, 25 u] of 25 rnM NADP, 25 pl of 133 25 .mM phenazine methosulfate, 200 pl of 5 mM 3-(4,5-dimethylthiazol-2-y|)-2,5- diphenylteflamlhnnbronfideando.5mgofprotein. Thereactionwasmonitoredby measuringabsorbanceat578nm. Oneunitwasdefinedastheamomrtofenzymethat catalyzes the NADP-dependent oxidation of 1 mrnole of glucose-6-phosphate in 1 nrin. Nor-1 Protein Localization by Immunofluorescence Microscopy. Hyphae gown in YEShqrfidmediumweewfleaedbyfilmfionmdrewspmdedmfieshYESfiqrMmedimn (1 mgwethyphaeinSmlliquidmedium). AvolumeofSOulofthehyphal arspensionwas droppedontoaslidepreneatedwith2°/o(w/v)glycinesolutionandairdried. Thefirngal cells onflreslideeithefiomliqiriderlnneorfiomslideadmrewerefixed for4hatroom ternpeature or 16 h at 4°C in I-listochoice Tissue Fixative (Amresco, Solon, OH). For partial digesfimofflwcdlwalfimgflcdlsweewbjeaedtoacdlwafldigesfimmzymesohfion consisting of 1.5% (w/v) Novozyme 234 in wate for 1 h at 30°C. The cell membrane ofthe hyphalcellwaspermeabilizedwitho.l% (w/v) saponininTris-bufl‘eed saline (TBS) consisting of 100 rnM Tris-Cl (pH 7.5) and 0.9% (w/v) NaCl for 30 rrrin at room temperature. The hyphae were the: exposed to the triple-purified anti-Nor-lc protein polyclonal antibodies (PAb) (see Chapter 3) [10 ug /ml in modified TBS supplenented with 0.1% (w/v) saponin and 1% (w/v) bovine serumalbumin(BSA)] at room temperature for 1 h. This incubationwas followedbythreerinseswithTBS for 10mineach. The slide preparationwasthenincubated at room temperature with labeled secondary antibodies [a 100-fold dilution of fluorescein isothiocyanate (FITC)-conjugated anti-rabbit IgG (Signa Chemical Co., St. Louis, MO)] for 1 h. Afieafinalwashwithwater,thepreparationwasairdried,mountedon slideswith Signa Diagrostics mounting medium (Signa Chemical Co., St. Louis, MO), and examined under a 134 Nikon Labophot fluorescent microscope or a Zeiss 10 lase scanning microscope (LSM) to obseve FIT C fluorescence. LSM micrographs wee taken with the help of Dr. J. H. Whallon (Lase Scanning Microscope Laboratory, Michigan State Univesity, East Lansing, MI). Semiquantitative Petri Dish Assay for ml/GUS Activity (Overlay Procedure). The seniquantitativeinsitumeasurenert ofGUSactivitywasconducted accordingtoMbnkeand Schafer(1993) with certainmodifrcations. Briefly, melted agar [12% (w/v) inwater] was kept at 45°C in a wate bath and used as the overlay agar. X-glucuronide (5-bromo-4-chloro-3- indolyl glucuronide, X-gluc, Sigma Chemical Co., St. Louis, MO) was dissolved in dimethylsulfoxide (DMSO) (0.5 mg/ul), and added to 5 ml of the overlay agar at a final concentration of 0.3 mg/ml (substrate/agar) and irnrrrediately poured onto a Petri dish containingamycelial colonywhichhadbeerrinarbated fortheappropriateperiods oftime. ThePetridishwasgenlyshakenundlflreerfirecolonywascoveedwiththeovelayagar. Thedishwithovelaywasincubatedatroomtenperaurreuntilthebhre colorflillydeveloped (W3 h) RESULTS Distributions of Marker Enzymes and the Nor-l Protein in Cell Fractions. Cell fiactionation of mycelial cells was conducted using two methods: (1) disruption of fungal protoplasts using a Potter homogenizer followed by difi‘eential certrifirgation (the PH method) and (2) separation of the cytosol fi'om organdles by protoplast permeabilization followed by centrifugation (the PP method). The resulting cell fractions wee subjected to marke erzyme assay, to Western blot analysis for the Nor-l protein, and to protein concentration 135 determination. The results of Western blot analysis for the Nor-1 protein are shown in Figure 4-1. Distributions of marke enzymes, the pecentage of the Nor-1 protein (measured by the relative intensity of the Nor-1 protein band in the Western blot autoradiogaph), and the pecerrtage of the total protein in each fraction obtained by the PH method and the PP method are summarized in Table 4-1 and Table 4-2, respectively. Succinate dehydrogenase (DH) was used as a mitochondrion marke, glucose-6-phosphatase (G-6-phosphatase) as a errdoplasrrric reticulum (ER) marker, catalase as a peoxisome marke, and acid phosphatase as a lysosome marke (Muller et at, 1991). Glucose-6-phosphate dehydrogenase (G6PDH) was used as a cytosol marke (Vida et at, 1993). High pecentage ofeach organelle marker erzyme was detected in the cytosol fiaction using the PH method (Table 4-1) (succinate DH, 16.4%; G-6-phosphatase 50.6%; catalase, 39.0%; and acid phosphatase, 64.0%). Thee data suggest that the PH method disrupts a certain fiaction of each organelle. Succinate DH (mitocondrion marker) activity was also detected in the mitochondrial fiaction (32.5%) and the lysosomal fraction (20.1%). Some overlap in localization of the two markers was expected because mitochondia and lysosomes have similar sedirnertation coeficients (10‘ to 105 5). Analyzing the data for the organelle fiaction only, the highest level of G-6—phosphatase (ER marke) was detected in the microsomal fraction (24.5%). The highest level of catalase (peoxisome marke) was detected in the lysosomal fraction (20.6%) and the mitochondrial fiaction (20.5%) because the sedimentation coeficient of peroxisomes (5 x 10‘ s) falls within 136 Figure 4-1. Western blot analysis of the Nor-1 protein in cell fiactions of A. pm'asitr'cus. Mycelial cells were gown in YES liquid medium for 60 h with constant shaking (150 rpm) at 29°C in the dark. Cell fractions were obtained from the wild-type strain SU-l (except sample NA‘). The proteins in each fiaction were resolved by SDS-PAGE and then transferred to a PVDF membrane. Each lane was loaded with 10 ug protein. The membrane was probed with the double-purified PAb (10 pig/ml). A: Cell fiactions were obtained using a Potter homogenizer followed by differential centrifugation (the PH method). Lanes: 1 (CT), cytosol fiaction; 2 (RB), free ribosomal fraction; 3 (MC), microsomal fraction; 4 (LY), lysosomal fiaction; 5 (MT), mitochondrial fiaction; 6 (NU) nuclear fiaction; 7 (1K), 1,000 x g pellet from crude extract; 8 (NA‘), crude extract from a norsolorinic acid (NA)-accumulating strain in which nor-1 was disrupted; 9 (WL), crude extract from the wild-type strain SU-l. B: Cell fractions were obtained by protoplast permeabilization followed by centrifugation (the PP method). Lanes: 1 (CT), cytosol fiaction; 2 (OG), total organelle fiaction; 3 (NA‘), crude extract from an NA- accumulating strain in which nor-1 was disrupted. 137 Figure 4-1. ‘- Nor-1» CT RB MC LY MT NU 1K NA+ WL CT 06 NA+ 138 682—2: 2 388.3: 33. :8: fig 820 “8:8“... H 82: 2: 83 28> :8m 3.: .==o:8: 8889828 2: .8 ax: 5088 :8: 2: x 08— :80 E .388 782 2: .8 8:32: 23 .8 ER 2.: v + :88: 33082.8 2: .8 flex: 5088 18:23 x 88. 28 E 8088 782.8 £2.82 2:3 65 7.82 An: .23. 286 2886293 a: .583 25.. a5 8 882 .882 2. a? .888 888.38 as as: as. as a 885 732 a 2.55 23 + 2.82323 83 333 2: .8 A83»: wficcoemotoo 2: 80:: 28— 28 E .388 7828 £882 23 x 8. 5352: .9552: 7.82 A3 68:8: :80 E .388 :8: 2: .8 EB 23 + 80:8: 28 E 388 88: 23 ”ax; £088 88H. 2: ”882 S. n ...? to a we 3 a 3 ...o s. cc 3 a 2 no a no o.o a od g 782 no a new 2.2 3 a 2.. 9n a n: 8 a 8 3 a to 3 a 3. 22.25 93.28 752 E n 36 ad a no no a E to a 8 2 a 3: 3 a 8 3 n no :8 52% ask u... « Se 3 a ...N 3 a Z. 2 a _.: 3. a ...2 3 a Na 3 a No g 3.288% 23. on a can 2 a a.“ 2 a 8 2 a 8.8 S a. no” 2 a E 3 a ...o g 8228 3 a.. wow 2 a 8 2: H n: E a 2.. Z « .... no a 8 3 a 3 g 8288828820 2 a I: Z a as S. a n: ...m a I: 3 a 2m 2 a a.» S a 3 g 3282328 22605 885 qumonE oESEBE 0888.3 8528;00:5— m=2u=2 8:9: M x coo; 8:8,: :00 o: 2: 8 8 a _ a x .o: 8:22:50 80:84:32.8 388%: :8 8:35.880: :88: 3 3:58 80:8: =8 2 £088 782 2: :8 88:28 828:. .8 80:39:25 .7v 2.8:. Table 4-1. 139 Table 4-2. Distributions of marker enzymes and the Nor-l protein in total organelle and the cytosol fiactions obtained by protoplast permeabilization and centrifugation. Total organelles Cytosol Glucose-6—phosphatedehydrogenase (%) 3.0: 0.8 97.0:t2.6 Succinate dehydrogenase (%) 89. 4 i 5. 4 10. 6 :I: 4. 2 Catalase (%) 85. 7 :1: 4. 6 14. 3 t 10. 5 Totalprotein(%) 13.0117 87.0:t11.7 Nor-l relative intensity 15. 0 :t 3. 9 100 Nor-l(%) 2.2:t0.7 91812.3 Notes: (1) Total protein (%): (the total protein in total organelles or in the cytosol) + (the sum ofthe total protein in total organelles and the cytosol). (2) Nor-l (relative) intensity: 100 x [the intensity of Nor-l protein in one lane (from total organelles or the cytosol fi'action) on the Western blot autoradiograph] + (the intensity of the Nor-1 protein in the cytosol on the same Western blot autoradiograph) (Figure 4-lB). (3) Nor-l (%): {[the intensity of Nor-1 protein in one lane (from total organelles or the cytosol fi'action)] x [the total protein (%) of total organelles or the cytosol fi'actionI} + { the sum of [theintensityofNor-l proteinineachlanex thetotal protein (%) ofthe corresponding fi'action1}. (4) Each value was the mean :t standard error (SE) from tests performed in triplicate. the range of sedimentation coeficients of lysosomes and mitochondria (104 to 105 s). The highest level of acid phosphatase (lysosome rmrker) was detected in the mitochondrial fi'action (12.4%) and the lysosomal fiaction (11.7%) because of the same reason as mentionw above. These data suggest that difl‘erential cerm'ifirgation method can successfully fi'actionate organeflesaccordingmthdrsedhnauafioncoefidanseventhoughahrgemmtbaof organelles were destroyed by the PH method. By the PP method, 97.0% of GGPDH (cytosol marker) was detected in the cytosol fi'action while 89.4% of succinate DH (mitochondrion marker) and 85.7% of catalase (peroxisome marker) were detected in the organelle fraction (total organelles). In comparison, 83.6% of 140 Figure 4-2. Irmnunolocalizafion of the Nor-l protein in hyphal cells of A. musiticw SU-l grown in YES liquid medium. Mycelial cells were incubated in YES liquid medium for 60 h with constant shaking (150 rpm) at 29°C in the dark. The triple-purified PAb was used as the primary antibodies (10 113/ml). The secondary antibodies (FIT C-coniugated anti-rabbit IgG) were diluted loo-fold. Photographs (magnification, 200 x): Al, fluorescence micrograph of SU-l (the wild-type) grown for 48 h; A2, sample Al under white light; Bl, fluorescence micrograph ofa nor-l-dismpted strain grown for 48 h; 32, sample Bl under white light; Cl, fluorescence micrograph of SU-l (the wild-type) grown for 18 h; C2, sample Cl under white light. 14] Figure 4-2. 142 Figure 4-3. Immlmolocelimtion ofthe Nor-l protein inA. pa'an'ticw SU-l gown on YES agar. IheflmgtswasinwbatedonYESagarat29°C for60hinthedarlaThe triple-purified PAb was used as the primary antibodies (10 ug/ml). The secondary antibodies (FITC- conjugated anti-rabbit IgG) were diluted loo-fold. Pbotogaphs (magnification, 280 x): Al, fluorescence nncrogaph of SU-l (the wild-type); A2, sample A1 under white light; Bl, fluorescence microgaph ofa typical hyphal cell SU-l (the wild-type); B2, sample B1 under whitelight;Cl,fluorescencemicrogaphofanor-l disruptedstrain;C2,sampleCl under 143 Figure 4-3. 144 Figure“. Inmmolocalization oftheNor-l protein inA. pmuw'ficus SU-l bylaserscanning microscopy(LSM). MycelialcellsweregrowninYES liquidmediumat29°Cinthedarkfor 48 h with constant shaking (150 rpm). The triple-purified PAb was used as the primary antibodies (10 ug/ml). The secondary antibodies (FIT C-conjugated anti-rabbit IgG) were diluted loo-fold. Pbotogaphs (magnification, 350 x): A, hyphal cells under white light; B, fluorescence micrograph of sample A; C, fluorescence micrograph (the lower part) taken in the X-Yplaneandthephi—zsectionmicmgaph(theupperpart)takenalongtheZaxisand throughasflaigmwhitefinesdeaedmthelowerpan(seedetailsinDiswssion) 145 Figure 4.4. C=689 I=218 200l=70 (2:435 I=Q44 F0 L488 C=425 I=454 F0 L488 146 Vesicle Vegetative cell Foot cell / Figure 4-5. Schematic of a conidiophore of Aspergillus maritime The process of sporulation in A. parasiticus starts with a substrate somic hyphal cell which is called the . foot cell. The foot cell branches to give rise the conidiophore which is a long, erect. hyphal cell terminating in a bulbous vesicle. As the multinucleate vesicle develops, a large number of conidiogenous cells, which are called phialides (or sterignata), are produced over the vesicle. One or two layers of phialides (the first layer of phialides is sometimes called metula) may be produced. As phialides reach maturity, they begin to form conidia at their tips one below the other in chains (Alexopoulos and Mims, 1979; Kale et al., 1994). The conidiophore does not include the foot cell and conidia. To facilitate description of the localization of Nor-l protein accumulation and the localization of Nor-1 promoter activity, the vesicle with phialides attached is called “the conidial head”; the long and erect structure of the conidiophore is called the conidial stalk in this study. The schematic figure of a conidiophore was drawn according to previous descriptions (Alexopoulos and Mims, 1979; Kale et al., 1994) and scanning electron microgaphs (Tsuneda, 1983). 147 succinate DH and 61.0% ofcatalase were detected in the organelle Won by the PH method. msmggestflmfewaorganefleswaedesuoyedbythePPmahodthanthePHmdhod The majority of Nor-l protein was detected in the cytosol fraction (95.4%) and a trace Wwasddeaedmthemdear,numchondfiaLlymsomaLnfiaosomaLandnbosomal fi'actions (0.8%, 2.4%, 0.6%, 0.2%, and 0.6%, respectively) by the PH method. The majority of the Nor-l protein was detected in the cytosol fiaction (97.8%) and a trace amount was detectedinthe(total)organellefiaclion(2.2°/o)bythePPmethod. Thesedatasuggestthatthe Nor-l proteinisnottightlyassodatedwithcellularorganelles. Subcellular Immunolocalization of the Nor-l Protein. The Nor-l protein was localized insiminAparasiticusgowninliquidmedimnandon solidmedimnusingafluorescent antibodyprobe. TheappearanceofgeenfluorescencewresenceofFHC-labeledsecondary antrbody) was used as an indicator of the level of Nor-1 protein acammlation (Figure 4-2). Usingthisinurnmolocalimtiontechnique, mostoftheNor-l proteinappearedtobelocalizedin thecytosol andasigrificantquantityofNor—l proteinwasfoundto associatewith“particles” of uncharacterized compositions in hyphal cells florn the wild-type strain A. parasitic“ SU-l gowninYES liquidmediumfor48h(Frgure4—2A). Incontrast,thefluoresccncewasnot detectedintwonegativecontrols, anor-l mutant (genedisruption) ofiApaasificusgownin YES liquidmediumfor48 h(Frgure4-ZB) arrdflrewfld-typeApmariticras SU-l gownin YES liquid medium for 18 h (Chapter 2) (Figure 4~2C). More information was obtained fl'om thefungusgownon solid media. Theimmatm‘econidium showedthemostintensivegneen fluorescence (during photogaphy, the most intensive geen color artifactually appeared yellow); while the corresponding conidial stalk showed stronger fluorescence than that 148 observed in the vegetative hyphal cell (Figure 4-3A). In ageernent with the data flom liquid culture, the majority of Nor-1 protein was localized in the cytosol. However, a significant quantitywasalsofoundtoassociatewith‘particles”inhyphalcellsofthewild-typeA. parasiticus SU-l gown on YES solid medium for 60 h (slide culture) (Figure 4-3B). The nor- 1 disrupted rrrutant (negative control) gown on YES solid medium (slide culture) for 60 h did not show detectable fluorescence (Figure 4-3C). Laser seaming microscopy (LSM) was used to collect more information about the subcellular location of the Nor-1 protein. Micrographs ofhyphalcellsfi‘ompr'asiticusSU-l gowninYES liquidnwdiumfor48 hweregenerated by LSM using white light (figure 4—4A), fluorescence (Figure 4-43), and fluorescence in phi-z section (Figure 4-4C). The phi-z section represents the optical section taken in the vertical Z plane(theupperpart ofFrgure4-4C) whichpassesthrough a straight line (thewhitelinewhich passes through a selected fluorescent “particle” shown in the lower part of Figure 4-4C) in the micrograph taken in the X-Y plane (the lower part of Figure 4-4C). The position and the shapeofthe fluorescent“particle”inthe phi-z section indicatedthatitwaslocatedwithinthe cellandnot stucktotheoutsidesurface. Aschematic ofthe strucmreandthedevelopment of a conidiophore are shown in Figure 4-5 for reference. In Situ Localization of nor-l Promoter Activity in a Colony of A. Wars. The close correlation between nor-1 promoter activity and the process of sporulation in A. parasiticwgownon solidmediawasinitiallyobservedusinginvitroanalysis(Chapter3). In orderto confirmthe invitro data, in situ localization ofnor-l promoteractivitywasconducted in a fungal colony. An A. pw-asificus strain, nor-llGUS (the same strain as used in 149 Figure4-6. Insituanalysisofnor-l promoteractivitywithinacolonyoprmasiacusgown onsolidmedium.Thefimgusinallthecolonies(exceptcolonyB)wasaA.pausificus transformantcontainingamr-l/GUSreporterconstruct. Thefimgusincolonwaasthe wild-type SU-l which did not contain the nor-l/GUS reporter construct. All colonies were hmbatedonYESagarmPeuidislresmdthmovaiaidwimmovaiayagarwmmfinga chrornogenic GUS substrate X-glucuronide. Photogaphs: A and B (magnification, 0.7 x), coloniesgownfor3days(colonyAanchadthesamemorphologybeforebeingoverlaid); C (magrification, 5 x), part of a colony gown for3 days; D (magnification, 30x), vegetative hyphalcells,andhyphalcellmasses(bhre)ofacolonygownfor5days;E(magnification,25 x),conidialheadsandconidiaofacolonygownfor4days. 150 Figure 4-6. 151 Chapter 3) which contains a nor-l/GUS reporter construct integated at the nor-1 locus (Chapter3),wasincubatedon YES solidmedimnforappropriateperiods oftimeandthen overlaid with melted agar (45°C) containing a chromogenic B-glucuronidase substrate (5- bromo-4-chloro-3-indolyl glucuronide). Accumulation of a blue colored end product generated by B-glucuronidase indicates B-glucuronidase activity. The blue color was observed inallareasofthecolonyexceptattheoutermarginmgrre4-6A). Nobluecolorwas observed in the control colony of the wild—type SU-l which did not contain the nor-l/GUS reporter construct (figure 4-6Bl The following describes GUS activity in a section (Figure 4—6C with a higha magnification than Figure 4—6A) of a colony of the nor-l/GUS strain (gown for 3 days) proceeding from the outer margin gadually toward the center. No blue color was detectable in the peripheral region (0.1-0.4 cm in width) in which only substrate hyphae were observed. An blue color was observedintheadjacent region(0.l-0.4cminwidth)inwhichaerial hyphaeweredominant. A more intense blue color was observed in the next adjacent region (0.8-1.0 cm in width) in which immature conidia were dominant. A less intense blue (or no blue) color was observed atthecenter(0.04-0.06cmindiameter)inwhichonlymatureconidiawerefound.Theinsitu datawaeconsistauwiththemvimdata(Chapta3)wlflchwggestedthatmr-l promoter acfivitywasdoselycorrelatedtoflrepmcessofsponflafioninpr'asifials. At certain stages of gowth on solid media (e.g. afler 5 days of incubation), some vegetative hyphae formed tightly interwoven round structures which showed an intense blue color (Figure 4-6D). When a colony was overlaid with the overlayer agar (containing X-glucuronide) and was observed underadissectionmicroscopeatlS-nrinfinreirrtervalsthebhrecolorappearedflrstinthe 152 conidialheathhconidiaaflachedsecondintheconidialstallgandlastinthevegetative hyphal cell (Figure 4-6E). DISCUSSION Threetentafivecondusionscanbedmwnfi‘omflredatapresentedinflfissmdy: (l)the Nor-l proteinwaslocalizedmainlyinflrecytosolofthehyphalcefl;(2)theNor-l proteinwas foundminmrauuewmdhandwmdidheadswmdidaalksvegaafivehyphaeandm conidia in the order flom the highest level to the lowest level (undetectable); and (3) the localizationofnor-l promoteractivityiscloselyconelatedtotheprocessofspomlationinA. parasitic-us SU—l gownon solidmedia. Theprimarypurposeofcellfi'actionationinthissmdywastocheckiftheNor-l proteinis mdosedmormongyaswdaedwnhcatdnmbcdmmpmfidesororgandlesfihemahods used for sample preparation for cell fractionation was a major factor which influenced the reliabilityofsubcellularlocalizationoftheNor-l proteininthisstudy. Oneofthe most widely used methods for determimtion ofthe effectiveness ofcell fractionation is the bioassay for specific marker enzymes. The marker enzyme assay suggested that Potter homogenization (the PH method) destroyedalargemrmberoforganelles. However,themarkerenzymeassayalso suggested flmdifl‘aarfialcamifirgafionatecluflqueMcedbytheNobdlaureaterat Claude, efl‘ecfivdysepmateddifl‘aemmbceflrflarparfidesaccordingtothdrdifl‘aem sedimentation coeficients when used afier sample preparation by the PH method. The protoplast permeabilization (PP) method was shown to be more efl‘ective in preserving theintegrityofcellularorganelles. 'I'hismethodcanremovethecytosolfl‘omthepermeabilized 153 cell and maintain organelle associations with the permeabilized cell (Vida et al, 1993). The bufl‘ers used to remove cytosol contain physiological levels of magnesium and potassium ions, whichamreqtfiredtomahnainorganefleassodafionsudththepermeabifizedcdl Wrthshort fimeandlowspwdcaflifirgafionfiecytowlmflpmneabflimdcdbueeflecfivdysepmfled (Vida etal, 1993). For example, glucose-6-phosphate dehydrogenase (G6PDH) has been established as a usefirl cytosolmarkerinAmergiIIus (archasA. niger,A. mduIans, A. apergillus)byseveral researchers (Carson and Cooney, 1990; Broek et al., 1995; Vida et al., 1993). The high percentage(97%)ofG6PDH detectedinthecytosolfiactionbythePPmethodinthisstudy was similar to the data (98%) of Vida er al (1993) and Broek et a1. (1995). Higher percentages of succinate DH (mitochondrion marker) and of catalase (peroxisome marker) weredetectedintheparficlefiactionbythePPmethodthanbythePHmethod. Similar percentages ofthe Nor-1 protein were detected in the cytosol fiction by either the PH method (96%) orthe PP method (98%) suggesting the Nor-1 protein is not enclosed in or strongly associated with a particle(s) or an organelle(s). Ifthe Nor-1 protein were enclosed in or strongly associated with a particle(s) or an organelle(s), it might be predicted that a higher percentage of the Nor-l protein would have been detected in the cytosol by the PH method thanthePPmethodbecausemoreorganellesweredesfloyedbythePHmethodthanthePP mahod. The data did not support this hypothesis. Even thoughthedata suggestthattheNor-l protein is not enclosed in or strongly associated with a particle(s) or an organelle(s), the highest concentration of Nor-1 protein was detected in the free ribosomal flaction by the PH method suggesting that a significant quantity of the Nor-1 154 protein is associated with flee ribosomes or with particles (possible the complex consisting of proteins involved in the AFB1 pathway) with significant mass which comigrated with free n‘bosomesdmingdifl‘erentialcentrifirgation. 'I'heimrmmolocalimtiondatasupportedanassociation of the Nor-1 proteinwithaparticle(s) orsomesort. Forexample,inmycelialcellsgowninYESliquidmediumorgownonYES solidmedium,mostoftheNor-l proteinappearedtobelocalizedinthecytosolbuta significant quantity appeared to be associated with particles by imnnmolocalization. This observation was confirmed by Laser Scanning Microscopy (LSM). Comparing the micrograph takeninflreX-Yplanewiththernicrogaphtakenhrtheverficallplane,afluorescentparticle waslocalizedinsidethecell. ThererrmhfingNor-lproteinwlrichwasnotassociatedwith particles, could in theory spread throughout the cytosol Whar the phi-z section was observed inareaswheretheNor-l proteinwasnotattachedtoanyparticles,thereshouldbea“srnear” withweakfluorescmcemdflmhdgnofme“mreaf’flmgmez-ardsduecfionshmfldbe approxirmtelyequaltothecrosssectioml diarneterofthehyphalcell. IfalloftheNor-l proteinweresimplyattachedtothesurfaceofthecell,thephi-zsectionwouldhaveshowna circle offluorescence with no “smear”. Inthe actual experiment, the phi-z section showed that the Nor-l protein, when it was not associated with any particle(s) or organelle(s), was extensivelydism'butedthroughoutthecytosol. TheobsavafimflratasigfificmflqumfityoftheNor-lpmtdnappearedwbeassodaed whh“parfldes”ofhrgemassbymsimmmmolocdinfionmflrstappeammbemmnsistmt withthedataobtainedbythePHmethodandthePPmethodbywhichonlyalittlearnountof the Nor-l protein was localized in particles or organelles. One possible explanation which fits 155 the experimental data is that the Nor-1 protein is associated with “particles” of small mass. 'l'hesesmallparticlesareintrnnlooselyassociatedwithparticlesororganellesofrmrchlarger mass which are detected in situ immunolocalimtion. Such an association could be destroyed during protoplast homogenization and protoplast permeabilization. Because of limitations of resolution and magnification, fluorescence microscopy and laser scanning microscopy cannot distinguishsmallorganelles suchasn'bosomesandperoxisomes. EveniftheNor-l proteinwas assodatedwithsmaflparfideswhichdism‘hnedeaensivdymthechtheNm-l protein would still seem to be localized in the cytosol. However, a larger fiaction ofthe small particles nfighranainassodmeduimmehrgepmfideswhmcdbwaefixedunmedimdyafiahmvefl forinsituimnurnolocalimtion. Thishypothesiscanbeaddressedmoreeasilybyelectron microscopy. TheNor-l protein maybe associated with small particles which may still reside primarily in thecytosolforanimportantreason. AsanearlyintermediateintheAFBrpathway, norsolorinic acid could be a precursor for other secondary metabolites. A goup of pignents withthebasic structure ofnorsolorinic acid andagoup ofxanthoneswiththebasicstmcture of sterigmatocystin were detected in the germsAmergillus in the 1960’s and 1970’s (Steyn et al., 1980). Norsolorinic acid, which does not possess a bisfuran ring, does not exhibit genotoxicity. Versicolorin A (VA) and other later intermediates (after VA) in the AFBr pathway, which possess a bisfuran ring, are mutagenic and genotoxic (Mori er al., 1985). Therefore the earlier nontoxic or less toxic intermediates (like norsolorinic acid) in the AFBl pathwaymaymtbemmpamnanalizedandnmymkepmtmothabiosynflwficpmhways The 156 more toxic intermediates may be compartmentalized in order to protect the producing cell fiom toxicity and to increase the eficiency of aflatoxin biosynthesis. The localization of Nor-1 protein accumulation is closely correlated to the process of sporulation in A. pamiticus SU-l gown on YES agar. Immunolocalimtion data showed that the conidial head, on which immature conidia were dominant, showed the highest level of the Nor-1 protein, the conidial stalk showed a higher level of the Nor-l protein than the vegetative hyphalcdkarrdmatureconidia, whichdidnotattachtotheconidialhead, showedundetectable Nor-1 protein. Thesedatawereconsistemwithflieinviuodataforflreacamndafionofthe Nor-1 protein, the accumulation of aflatoxin, and the activity of the nor-1 promoter presented previously in this dissertation (Chapter 3). The localimtion of nor-l promoter activity was also closely correlated to the process of sporulation in A. par-milieu: SU-l gown on YES agar. The in vivo localization of nor-l promoter activity in a colony was monitored by the nor-l/GU S reporter construct (blue color). Areas of the colony containing substrate hyphae showed no detectable blue color, areas containing aerial hyphae showed an intense blue color, areas containing immature conidia showed a more intense blue color, and areas containing mature conidia showed dark-green color which was the color of wild-type conidia (during photogaphy the dark-geen color artifactually turned to dark-brown). The pattern of appearance of blue color was consistent with the process of sporulation. Sporulation initiates with a foot cell (at the substrate level) whichgeneratesanaerialhyphalcell. Thisaerialhyphalcellthengoestln'oughaseriesof morphological steps including a conidial stalk, a conidial head with immature conidia, mauu'e 157 conidia attached to the conidial head, to mature conidia falling off the conidial head. Therefore the localization of the nor-1 promoter activity was consistent with the process of sporulation. The correlation between the localization of nor-1 promoter activity and the process of sporulation was firrther supported by observation of the blue color development in individual conidiophores under a dissection microscope at 15-min time intervals. The conidial head with immature conidia was first to show the blue color, the conidial stalk second, and the vegetative hyphalcelllast. 'I'hisdifl‘erenceintheappearanceofthemr—l promoteractivityindifl'erent location could be due to two possible reasons: (1) real difl‘erences of the nor-l promoter activityand GUS activityin the conidial head, the conidial stalk, andthevegetative hyphal cell; or (2) difi‘erences in substrate permeability. A demonstration of the second hypothesis is difiarhbecmrsembsfiatepermeabflhyisdeterfinedbyatleasttwofactors:thestructureof ceflwaflandceflmanbmneofflrefimgusandthechenficalandphysicalnatumofthe substrate. Howeverthefirsthypothesisissupponedbyinsimimnumoloealizafiondata Further research is required to confirm this observation. Sclerotia (surviving structures) are relatively massive structures composed entirely of vegaafivecdlswhichafisebympeatedhyphalbmdfingmdhnenselyunawomAt maturity, sclerotia contain a mixture of darkly pignented and hyaline (coloriess) hyphae (Deacon, 1980). Accumulation of aflatoxins in sclerotia was observed in some strains of A. flaws and A. parasiticus (Wreklow and Shotwell, 1983). In this study, relatively massive structures composed entirely of vegetative cells, which showed an intense blue color, were observed wggesting that these intensely stained blue colored masses may be developing or irmnature sclerotia. A wild range of environmental and internal factors influence the initiation 158 and subsequent development of sclerotia. For example, internal morphological factors have been suggested to be involved in the initiation of sclerotia (Chet and Herris, 1975). Therefore, it is possrble that aflatoxin gene expression is corregulated with two difl‘erent developmental processes (sporulation and scelrotial development) in A. parasitic-us. In summary, the localization data suggest that the Nor-l protein resides mainly in the cytosol of vegetative hyphae and that localintion of Nor-1 protein accumulation and the localimtion ofnar-l promote activity are closely correlated to the process ofsporulation inA. parasiticras. ThesnidiesinflfischapterpresanprdMnarydataonthembcdhflmlocalimfion ofNor-l protein. Theinsitu subcellularimmunolocalimtiondataneedtobeconfirmedbythe use of electron microscopy. CHAPTERS ISOLATION AND ANALYSIS OF HUP, A GENE ASSOCIATED WITH HYPHAL GROWTH, HYPHAL DEVELOPMENT AND SPORULATION IN ASPERGEL US PARASIUCUS ABSTRACT: To study a gene (fluP) encoding a putative polyketide synthase, a disruption vector was constructed and transformed into Amergillus parasiticus. Disruption of fluP resulted in slow gowth with cotton-like hyphae, no sporulation, and reduced aflatoxin accumulation. The data suggest that fluP is associated with vegetative cell gowth, sporulation, and in turn indirectly influences aflatoxin accumulation in A. parasiticus. INTRODUCTION Research on the molecular biology of aflatoxin biosynthesis may lead to development of novel methods to reduce or eliminate aflatoxin contamination. However research on the general biology of the toxigenic fungi encompassing areas such as the, regulatory mechanisms involved in hyphal gowth, hyphal difi‘erentiation, and sporulation is also usefirl. Blocking or interfering in any of these processes would help to eliminate or reduce aflatoxin accumulation. Studies on the genes involved in these basic cell fimctions may also provide useful information for the development of new fungicides, improvement of agonomic practices, development of resistant crops (by genetic engineering or classical 159 160 breeding), and development of biocontrol agents (such as nontoxigenic Competitive firngi) for control of aflatoxin contamination. Unfortunately, relatively little research data have been published related to this area. Because aflatoxins are derived from a polyketide pathway (Kurtzman er al., 1987), the producing fungi are predicted to have a polyketide synthase (PKS) related to aflatoxin biosynthesis. Previous studies showed that a 680-bp CIaI/HindIII DNA fiagment from an aflatoxin-producing strain, A. parasitials SU-l, hybridized to a 1.5-kb cDNA fiagment containing the B—ketoacyl synthase functional domain of 6-methylsalicylic acid synthase (MSAS) complex fiom Penicilliwn patulwn (Beck et at, 1990). As expected, the deduced ammaddsequencefiomthe680-prlaUHbrdeNAfingnanwasvaysinflhr(72% identity)tothatofthe B-ketoacyl synthasefimctional domain ofMSAS ofP.patqun. Itwas originally hypothesizedthatthegene containingthe680~bp DNAfiagnrerrtfromAparasiticus amdesaPKSwfichishvoNedhaflflordnbioWefisbecmmemwbfisheddflamggefled thatpamfinmd/mhsprewmor6-methylsahcyficaddisproducedmprasificus Onthe other hand, it is also known that certain polyketides are closely related to cell difl‘erentiation, development, and sporulation (Summers er al., 1995; Revill et al., 1995; Keller and Adams, 1995). Therefore a reasonable alternative hypothesis is that the gene containing the 680-bp DNA fiagment is a PKS gene involved in either aflatoxin biosynthesis, cell gowth, cell difl‘erentiation, or sporulation in A. parasiticus. The data presented in this chapter demonstrated that disruption of the suspected PKS gene (fluP) resulted in slow gowth with cotton-like hyphae, no sporulation, and reduced aflatoxin accumulation. This suggests that fluP is associated with hyphal cell gowth, hyphal cell 161 development, and sponrlation, and in turn indirectly influences aflatoxin synthesis in A. parasiticus. MATERIALS AND METHODS Bacterial Strain, Fungal strains, and Incubation Conditions. Plasmids were amplified in Escherichia coli DHer Fem/emu] W1 7(rK' mK+)supE44 thi—I recAI gyrA (Nair) relAI A(lacZYA-argF) Ul69 (m8OIacZAM15)] using standard methods (Ausubel et al., 1987). Aspergillus parasiticus NRRL 5862 (ATCC 567755, SU-l) was used as the aflatoxin-producing wild-type strain. A. pw'asr'ticus NR-3, a nitrate reductase (encoded by the m'aD gene) deficient strain derived fi'om A. parasiticus SU-l (Horng er al., 1990), was used as the host stain for fluP disruption. Fungal strains were maintained as stock spore suspension at -80°C in 20% (v/v) glycerol. Czapek-Dox (Difco laboratories, Detroit, MI) agar (CDA) medium containing sodium nitrate as the sole nitrogen source was used for selection of niaD” transformants. Aflatoxin-inducing media, coconut agar medium (CAM) (Arseculeratne et al. , 1969), potato dextrose agar (PDA) medium (Difco Laboratories, Detroit, MI), and yeast extract-sucrose (YES) liquid medium and YES agar (Chapter 4) were used to gow fungal mycelia for genomic DNA and total RNA preparation, for aflatoxin extraction, and for observation of morphology. Reddy’s medium (Reddy er a1. , 1971), a chemically defined and aflatoxin-inducing medium, was also used to gow mycelia for RNA preparation. Fungal Culture, Dry Weight Determination, Growth Rate Measurement, and Aflatoxin Analysis. For gowth of fungi on solid medium, 5 ul of a stock spore 162 suspension (1.5 x 10‘ spores/ml) were inoculated onto the center of agar media and incubated at 29°C in the dark. For the non-sporulating fluP-disrupted strain, hyphae were removed fi'om a flufl'y colony with a toothpick and transferred onto the center of agar media and incubated at 29°C in the dark. The quantity of hyphae transferred was determined by scraping the hyphae fi'om the toothpick, resuspending in 1 ml of YES liquid medium, and spreading the hyphal suspension on CAM Petri dishes. The number of colonies which gew on each Petri dish was between 50 and 200. The differences in inoculum level did not appear to influence the gowth rate of the colonies. The majority of the colonies (95%) initiated fiom toothpick-transfer had the same gowth rate. Colonies initiated either from spore-suspension (more than 95% showed the same gowth rate) or from hyphal-transfer with difl‘erent gowth rates (compared with the majority of the colonies prepared by the same procedure) were eliminated from the analysis. Mycelial samples were collected by using a spatula to scrape mycelia fi'om the surface of agar. The collected mycelia were either used immediately, or fi'ozen under liquid nitrogen and stored at -80°C until use. The dry weight of mycelia gown on solid medium was determined by the filtration-drying method of Olsson and Jennings (1991). For gowth of fungi in liquid medium, a stock spore suspension was inoculated into liquid medium to a final spore concentration of 1.5x10s spores/ml, and gown at 29°C in the dark with constant shaking (150 rpm). Mycelia in liquid media were collected by filtration through Miracloth (Calbiochem-Novabiochern Corporation, La Jolla, CA), and were either used immediately, or frozen under liquid nitrogen and stored at -80°C until use. 163 Colony gowth rate was measured by the time (in days) which the colony took to cover the surface of agar medium in a Petri dish or by the mycelial dry weight (mg) per Petri dish. AflatoxinBr wasanalyzedbyusingdirectcompetifiveenzyme—linkednmmrneabsorbem assay (ELISA) according to the method of Pestka (1980). Aflatoxin Br monoclonal antibodies usedinthisstudywereldndlyprovidedbyDr. J. Pestka(DepartmentofFood Scienceand Human Nutrition, Michigan State University, East Lansing, MI ). Temporal and Regional Collection of Samples from Solid Medium. To measure aflatoxin acctmrulation in wild-type A. pm'asiticus NR-3 gown on YES agar in a Petri dish, mycdialsampleswaecoflectedaccordhrgtoaspecifictemporalandregional scheduleas described previously (Chapter 3, Materials and Methods). Briefly, samples were collected fromthecentralregiononESagarZ, 4, 5,and6daysafierinoculation; fi'omthemiddle region4, 5,and6daysafierinoculation;andfi'omtheperipheral region 5,and6daysafler inoculation. For collecting samples of the fluP-disrupted transformant gown on YES agar, the sameregional schedulewasusedbutthetimepointswerechangedto 6,12,15, and 18 days alter inoculation, because the colony of wild-type NR-3 took 6 days to cover the YES agar in a Petri dish, while the fluP-disrupted transfonnant took 18 days to cover the same area. Plasmid Preparation and Protoplast Transformation. Plasmid minipreparation was performedbytheboilingmethod, andlarge-scalepreparationwasperfonnedbythealkaline lysis procedure (Ausubel et al., 1987). Transformation of protoplasts of A. pausitr‘cus NR-3 with the disruption vector was conducted using minor modifications of the polyethylene glycol method (Oakley et at, 1987) as previously described (Skory et at, 1990). 164 Nucleotide Sequence Analysis and Amino Acid Sequence Comparison. Nucleotide sequawedatawereanalyzedwiththeWuconsinGeneficsComprneerupPadmge Comparisons of the predicted amino acid sequence to EMBL and GenBank databases hbraries wereconductedwith’I'FastAandGapandalignedwithPileup fromtheWisconsin Genetics Computer Group Package. Isolation and Analysis of Genomic DNA and Total RNA. Fungi mycelia collected fiom eitherliquidmediaorsolid samplewaeusedforDNAandRNAisolationandpmification. Genomic DNA was isolated and purified by a published modification (Skory et al., 1990) of a phenol-chloroform protocol developed for mammalian DNA isolation and purification (Ausubel et a1, 1995d). Total RNA was isolated and purified using the hot-phenol method as previously described (Ausubel etal,1995e). Restriction endonucleases utilized in analysis of DNAwerepm‘chasedfiomBoehringerMannheirnBiochemicals (Indianapolis, IN) orNew England Biolabs (Beverly, MA) and were used according to the marnrfircturer’s instructions. Northern (RNA) and Southern (DNA) hybridization analyses were conducted using published procedures (Ausubel et al, 1995c, 1995f) with the modified hybridization bufl'er and conditiomrecommended by Stratagene Cloning Systems (LaJolla, CA). Approximatelle pg ofgenomicDNAor30ugoftotalRNApersamplewereseparatedbyagarosegel dchophorefisandflwnMnsfenedbycapflhwacfiontoanylonmanbmnemym‘ manbrane, SchleicherandSclurellInc,Keene,NI-I)which servedasasolid support. Radio- labeledDNAprobesweregeneratedwith[32P]-dCTP(DuPontCo.,Boston,MA)andthe Random Primed DNA labeling Kit (Boehring Mannheim Biochemical, Indianapolis, IN) accordingtothemamrfactrua’sinstructions. Afterthefinalwashthemembraneswere 165 exposed to X-ray film (Dodak-XARS, Eastman Kodok Co., Rochester, NY) at -80°C. DNA probes(specifiedinthefigurelegends)wereusedinbothSouthernandNorthernanalysis. RESULTS AnalysisoftheCodingRegionoffluP. InaprevimrssuidyconrhactedbyRRasoolyin our laboratory, a 680-bp CIaI/HindlII DNA fi'agnent from A. parasiticras was shown to hybridizetoal.5-kb cDNAfi'agmentcontainingtheB-ketoacylsynthasefirnctional domain of 6-methylsalicylic acid synthase (MSAS) complex fiom Penicilliran patulin» (Beck et at, 1990). Inflfisstudy,ascarchofEMBLmrdGeankdambasesshowedflratthemnmoadd sequawearwdedbyflie680-prIaUHmaIHDNAfiagnauwasalsosinfllarmflicB- ketoacylsynthasefimctional domain ofother6PKS genes (35%to7l% identity overa216- arnino-acidspan). ToillusuateanaminoacidsequencecomparisonoffluPtotwoPKSgenes andonefattyacidsynthetase(FAS)geneisshowninFigm'eS-l. Northernblotanalysiswas usedtoidentilytheappmrdmatesizeandlocationofthecodingregionoffluP. Initially,the 680-prIaI/HindlIIDNAfi'agmentwasusedasaprobetohybridizetototalRNAfi'omA. maritime: and an approximately 6-kb transcript was detected. Several genomic DNA fi'agnents (in vector B-E2 fi'om a phage A genomic DNA library of A. parasiticrw) flanking bothsidesofthe680-prIaI/Hincfl11DNAfi'agrnentwerethenusedasprobesinNorthem blot analysis (Figure 5.2) Each probe (0.9.rtb SacI/SphI, 0.3-1d) SohI/Psll, 3.0-kb PstI, and 166 Figures-1. AminoacidsequencecomparisonofflquithtwoPKSgenesandoneFASgene AP,anfinoaddsequence(AAS)anodedbyflre680—bpflagnemofflreAwgilhrs pw'as'ia'cusfluP gene; PP, AAS of the N-terminus of the Penicllr’ran patulran 6-methylsalisylic acid synthase gene (PKS gene); AN, AAS of the N-termimrs of the developmentally regulated A. WwAgeneQKS-like gene); YSC, AASoftheN—terminus oftheSacdrmanyc-es cerevia’aefattyacidsyntbasedenefiASgene).Theidentityandsimilarityare72%and86% betweenAPandPP,35%and58%betweenAPandAN,and13%and39%betweenAPand YSC. 167 Figure 5-1. 168 Clal SphI Pstl SphI Sacl Pstl SM Pstl HindlII SacI _l__ 61 | Genomic DNA Probes I II III IV V VI Figure 5-2. Aschernatic ofthe 9.1-kaachenomicDNAfi'agment containingtheflngene inAmergillrerrasiricus. DNA‘probesusedinNorthernanalysisto definethetranscribed region of fluP: I, 0.9-kb SacI/Sphl; II, 0.3-kb SphI/Pstl; III, 3.0-kb PstI; IV, 1.3-kb Pstl; V, 0.68-kb CIaI/Hr'nrflll; VI, 2.9-kb HindIII/Sacl. The arrow represents the proposed direction of transcription of fluP. 1.3-kb PstI fi'agment) except the 2.9-kb HindIII/Sacl fi'agnent detected the same size nanscriptasthatdetectedbythe680-prIaIlHindflIDNAfiagment. Sincethededuced amino acid sequence ofthe 680-bp CIaI/Hindlll DNA fragment was similar to the B-ketoacyl synthasefimctioml domain locatedintheN-termirmsofMSAS inP.parqun (Becketal, 1990),itwaspredictedthatfluPshouldbetranscribedflomrighttolefiasshowninFigm'eS- 2. The 6.1-kb .Sohl DNA fragnerrt, which contains the 0.3-lib SphI/Pstl, 3.0-kb Pstl, 1.3-kb P511, and 1.5-1d) Par/5pm fi'agments, was predicted to encompass the 5’-end plus its flanking region and most ofthe 3’-end offluP, and therefore could be used for disruption vector construction. Construction of a Disruption Vector pPKS. The 6.1-kb SphI DNA fiagnent was cloned into the .Sphl site of pUC19 resulting in plasmid pUCl9—6. The disruption plasmid, pPKS, was constructed by insertion of a 6.5-kb PvuII fi'agment from pUNH (Figure 5-3), containing a functional copy of the nitrate reductase gene, niaD, into the SmaI site of pUCl9-6. 169 Fignn5-3.TlnfhrP-dimrpfionveampPKSandascbanaficdesaipfionofflwameaed resnictionmapofgenomicDNAinthefluP-disruptedu'ansfonmnt. Thesolidblacklineisthe 6.1-kb .Sohlfi'agmentcontainingmost offluP. A6.5-kbfragnent,whichwascutofl‘fl'om plasrnideNHwasinsertedintothemiddleofthefhrchne.Theshadedline(2.7kb)inthe 6.5 kb fi'agnent is afirnctional copy of the nitrate reductase gene (niaD). Abbreviations: Sp, SuhLScSacLB,Banm;H,Hmn;smsnaL 170 Figure 5-3. SphI 15.0 kb HindIII 0.8 kb BamHI 0.9 kb PstI 1.5 kb 12.3 kb BamHI SphI HindIIIZ-B kb 11.? kb PstI HindIII 4.0 kb BamHI4.5 kb 9.8 kaacI 9.0 kl) BamHI pPKS was cut with Sphl and the linearized disruption vector was transformed into NR-3 c = * thch-I niaD P \ Double cross-over is?" m 1 P Sir The fluP gene in the genome 11E ip Ecfi l lirScrI fl :lfiSp SjE niaD — The disrupted fluP gene in the genome 2kb 171 Figure 5-4. Southern blot analysis of genonric DNA in A. parasiticus NR-3 and the quP- - disrupted transformant. Ten ug ofgenomic DNA from each sample was digested with SacI (Sa), SphI (Sp), Hindm (Hi), BamI-II (Ba), separated by agarose gel electrophoresis, and transferredtoaNytranmembrane. A3.0-kastIintemal DNAfragmentofflquasusedasa probe. A: disrupted, sample fiom the fluP-disrupted transformant (fluP). B: and D: wid, samplefi'omthewild-typeUIuP‘). C: mixed, samplefiomthecolonywiththefluP/fluP“ genotype. Themnnbersontheleflsideofeachpanelisfliemoleuflarsizestandardladder (unit: kb). 172 Figure 5-4. Sa Sp Hi Ba Sa Sp Hi Ba Sp Ba Hi Sp Ba Hi “1:56”: ”1in 1‘ l A: disrupted B: wild C: mixed D: wild 173 Transformation of A. parasiticus NR-3 and Disruption of fluP. Plasmid pPKS was linearized by cutting with the restriction enzyme Sphl to generate an approximately 12-kb DNA fiagment. The linear disruption vector was transformed into protoplasts of A. paramicus NR-3 (Figure 5-3). Nitrate-utilizing transformants (niaD‘) were selected on Cmpdc-Doxagar(CDA)comahnngsodhunmuateasthesolemnogensomceandthen nansfaredomococomnagmmedimn(CAM)mwnduaaqualhafivesaemforaflatordn accrmmlation. All colonies (more than 300) transferred to CAM produced aflatoxins. The selection oftrarrsformants containing the disruptedfluP gene was then conducted by Southern blot analysis. Southern blot analysis of genomic DNA isolated from more than 200 111'er+ transformants identified two transformants (isolates 10 and 11) in whichfluP was disrupted (Figure 5-4). Wild-type colonies gown on CAM (Figure S-SA) or on potato dextrose agar (PDA) (Figure 5-5B) gew with normal morphology of hyphae and spores. However colonies of disrupted msfommmsgewwhhnmchlessaaialhyphaemidhadudddydispasedsporesonPDA (Figure S-SC) or with cotton-like (flufl'y) hyphae and no spores on CAM (Figure S-SD). Thereforethisgene fiomApmudtiaaswastentativelynamedfluP for_fl__ufl‘ygene. Wild-type hyphae and cotton-like hyphae of the disrupted transformant gown on CAM are shown in Figures S-SE and 5-5F. When the hyphae ofisolates 10 and 11 were transferred onto CAM (secondary isolates), about 10% of the colonies recovered the wild-type phenotype and wild- type genotype (fluF, nondisruptedfluP gene) (Figure 5-43 and 5-4D). Another 20 % of the secondaryisolatesgewasamixtureofnormalphenotypeandabnormal phenotype(cotton- like hyphae with no spores) and contained both the non-disruptedfluP allele and the disrupted 174 Figure 5-5. Morphology of A. maritime: NR-3 and the fluP-disrupted transformants gown on coconut agar medium (CAM) or on potato dextrose agar (PDA). Pictures: Al (magrification, 0.7 x), a colony of NR-3 on CAM; A2 (magnification, 1.4 x), an enlargement of part of A1; B1 (magrification, 0.7 x), a colony of NR-3 on PDA; B2 (magnification, 1.4 x), an enlargement of part of B1; C1 (magnification, 0.7 x), a colony of the fluP-disrupted transformant on PDA; C2 (magnification, 1.4 x), an enlargement of part of C1; D1, a colony of the fluP-disrupted transformant on CAM, D2 (magnification, 1.4 x), an enlargement of part of D1; E (magnification, 20 x), hyphae of the wild-type (NR-3) on CAM; F (magnification, 20 x), hyphae of the fluP-disrupted transformant on CAM. Figure 5-5. 175 176 Figure 5-5 (cont’d). D1 DZ 177 Figure 5-6. Transcript accumulations of fluP, the B-tubulin gene, and the ver-I A gene in A. parasiticus. Total RNA was isolated from cells gown in Reddy medium (Reddy er al, l971)for24h,60h,and72h,respectively. ThirtyugoftotalRNAineachlanewas separatedbyagarosegel electrophoresisandtransferred onto aNytranmembrane. PKSO‘IaP): a ls-kadIhnernalfiagnerrtofflquasused asaprobe. var-I: a0.6-kb CIaI/EcoRI internal fragment of the Mar-IA gene was used as a probe. B-Tubulin: a 2.0-kb HiszH/Bamm internal fi'agnent ofthe B-tubulin gene was used as a probe. 178 Figure 5—6. 24h 60h 72h 24h 60h 72h &Ol(b—> 21)Kb—> 1A Kb—> PKS ver-1 B-Tubulin 179 Figure 5-7. Transcript accumulations of fluP in A. parasiticus NR-3 and the fluP-disrupted transformant. Thirty ug of total RNA was separated by agarose gel electrophoresis, and transferred onto aNytran membrane. A 1.3-kastI internal fragment offlquasused asa probe. Lane panel: A: l, 2 and 3, genomic DNA isolated from fluP-disrupted isolate ll, 10, and fi'om NR-3, respectively, gown on CAM for 72 h; B: l, genomic DNA isolated fiom fluP-disrupted isolate gown on YES agar for 4 days; 2 and 3, genomic DNA isolated fiom NR-3 gownonYES agarfor2 and4 days, respectively, 4and5, genomic DNAisolatedNR- 3 gowninYES liquidmediumforZand4days, respectively. 180 Figure 5-7. <—6 kb-* 181 fluP allele (fluP‘lfluP) (Figure 5-4C). About 70 % the secondary isolates maintained only the abnormal phenotype (cotton-er hyphae with no spores) and genotype (disrupted-fluP allele only). The pattern of phenotype and genotype in the secondary isolates on CAM (the CAM pattern) was repeatable from one transfer to the next transfer. Interestingly, when the hyphae of isolates 10 and 11 were transferred onto PDA, the colonies gew with much less aerial hyphaeandhadwidelydispersedsporesmgure 5-5C). ThispatterniscalledthePDApattem. When the spores produced on PDA were inoculated onto either CAM or PDA, all the colonies showed normal phenotype and normal genotype. When the hyphae gowing on PDA were inoculated onto CAM or PDA, the CAM or PDA pattern appeared again. fluP Transcript Accumulations in the Wild-Type Strain and the fluP-D’mrupted Transformant. Total RNA extracted fi'om wild-type mycelia gown in selective Reddy’s liquid medium (Reddy et al., 1971, an aflatoxin-inducing chemically defined medium), was subjected to Northern hybridization analysis. A 6.0-kb transcript was detected in the 24-h, 60- h, and 72-h samples using a 1.3-kb Ps'tI internal fragment of fluP as a probe (Figure 5-6, PKS). In contrast, a 1.1-kb transcript was only detected in the 60-h, and 72-h (not in the 24-h) samples using a 0.6-kb ClaI/EcoRI internal fi‘agment of var-1A gene (directly involved in aflatoxin biosynthesis in A. mariticus) as a probe (Figure 5-6, ver-l). This pattern of transcript accumulation of ver-IA was similar to that seen in previous studies (Trail et at, 1995). As a control, a 2.0-kb transcript was detected in the 24-h, 60-h and 72-h samples using a 2.0-kb HirrdIH/BamHI intemal fi‘agnent of the B-tubulin gene (a house keeping gene) as a probe (Figure 5-6, B-Tubulin). The transcript of the B-tubulin gene appeared in geater abundance in the 24—h sample than that in the 72-h sample, a pattern seen in previous studies 182 (Trail etal, 1995). In contrast, the transcript offluP appeared in less abundance in the 24-h sample than that in the 72-h sample. Also, the 6.0-kb transcript was not detected in the two fluP-disrupted transformants (figure 5-7A, lanes 1 and 2), but was detected in the wild-type cells gown on CAM for 72 h (Figure 5-7A; lane 3). Normally, sporulation can occur only on solid gowth media, therefore if fluP is associated with sporulation the corresponding transcfipt onsofidmediaiserpeaedtoappearingeaterabundanceflunfliatinfiqiudmedia Total RNA ofthefluP-disrupted transformant was extracted fiom the firngus gown in YES liquid mediumandYES solidmedium. Attwotimepoints, thefluPtranscriptisolatedfi'omcells gown on solid media appeared in approximately 3-fold geater abundance (relative intensity on Northern blot analysis is 100) than transcript (relative intensity on Northern blot analysis is 35) isolated fi'om cells gown in liquid media (Figure 5-7B). Aflatoxin Bi Accumulations in the Wild-Type Strain and the fluP-Disrupted Transformant. In order to determine if fluP had an efl‘ect on aflatoxin accumulation, the hyphae of the fluP-disrupted transformant and the wild-type (NR-3) were inoculated onto YES agar. YES agar was used to measure aflatoxin accumulation in this study for two reasons: (1) the fluP-disrupted transformant gew on YES agar with similar morphology to the CAM pattern (fluffy-colony with no sporulation); (2) YES is the primary medium used to study aflatoxin accumulation in this dissertation (Chapter 2, 3, and 4). The hyphae of NR-3 took about 6 days, while the hyphae of the fluP-disrupted transformant took about 18 days to coverthe surface ofthe gowth mediumin aPetri dish Thequantityofhyphaetransferreddidnotappearto afl‘ectthehyphalgowthrateinthis experiment. Mycelial samples were collected according to the specific temporal and regional schedule as described previously (Materials and Methods). The dry weight of the mycelia 183 Table 5-1. .mtmz 0996:? 05 .Lfix. ”fiancee“: voqummvtma§05 2 $3.39 .88. 838.82.. 8....» mo 5% .88 23:8... H 58:. o... 2...? seem Am. .8882 ...a nausea. <28 .... Began :3 .928 8.33888 2... 5 A8282 28 8.882 ... 288.. 83 coca—.82.. .38 93.. w. 98 .996 m. ..3 996 o ...... .98.. m comma. 88in»... 2.. 80.... 882.8 £28.30... 8...... 98.8 m. 98 .98.. m. .9§o N. .3 9.3 e vs. .98.. m .98.. v comma. 882... o... :8... 388:8 £38.80... 8%. A998 w. .28 .98.. m. .953 N. .98.. o ..3 98.. 0 can .33 m .938 v .98.. N comma. .8280 85 89c 888:8 883 Amaiuggomce. evasamfitmak. o... .«o ..3 mamz 25.2...» o... «o 8.9.8... 389?. 8.7.... t8m ... 8w“ mm.» co ESE 8.83 88200 A: .882 ...3... 3...: 883.62 33.8 2.2.... 23...... e 2 as... m .. a o ... e ... a . .n . .2 a e .93 an n a .2 e a n ... .8 e .m a e .... e 2 .8889. .....36 ...3: c.2342 33.2 «.33... 8.3.? n 2 3.2.8.... 8..."... ...3: 33.8. ”.28.". 3...... 3.3... e ... 28.2 «..3... ...3..." 123.8 ...3... 2:5... ...3; n 2 28.2 “..3... 3...: 23.2 3:": “.3: 3.3.. e N. 38.2 e..“: 2...: 3:3... «.23.... «...3... 3.3.. o 2 .380 e..“: ...3: ...3”... ...3...“ Team... «...3... m 2 .880 ...3”: ...3...“ ”...3...” ...3; You: ...3... e N. .838 ...3; 3:”: «.3: 2:3... ...32... ....«m... N o .880 ......x .....x ......x .....x ......x .....x ......x .....x 8.8.8 .32. .5 use... .92 .229»... 8...... as. 2.92. an En... on... 8.8.30 ease .o 8.8... came mm; ..o gew 28.53.28: Baafivdak. o... 98 642 «acuteg .V ... 88838.88 .m 5.68%. 98 2.383 Ev .2832 .—.m 03:. 184 and. the accumulation of aflatoxin B1 (AFB1) (from the growth medium and mycelia) of each mycelial sample were determined (Table 5-1). The overall accumulation of AFB1 (per Petri dish) in NR-3 was approximately lO-fold higher than that of the fluP-disrupted transformant, while AFB: accumulation per gram of mycelium of NR-3 (7.0 ug/mg) was 2-fold higher than that of the fluP-disrupted transformant (3 .2 ug/mg). The overall growth rate of the wild-type was 3-fold faster than the fluP-disrupted transformant if the time to cover the Petri dish was used as the indicator, and 4-fold faster if the dry weight per Petri dish was used as the indictor suggesting that fluP may directly or indirectly control hyphal grth rate. The AFB: accumulation per gram of dry weight of mycelium was nearly the same (3.2 ug/mg) in all areas from a colony of the fluP-disrupted transformant which did not sporulate. However, in NR-3, AFB: accumulation per mg of dry mycelium (3 .2 ug/mg) in the central region 2 days after inoculation (no sporulation) was almost the same as that of the fluP-disrupted transformant, but increased quickly in the same region when sporulation appeared [for example 4 days (5.8 pig/mg) and 5 days (7.4 ug/mg ) after inoculation in the central region]. It was observed that the dry weight of mycelium and aflatoxin accumulation of the fluP-disrupted transformant started to decrease 15 days after incubation in the central region. These data suggested that mycelia and aflatoxin degraded after long incubation times. DISCUSSION Based on the data collected in this study, it is reasonable to conclude that fluP is 185 associated with hyphal growth, hyphal development and sporulation, and in turn indirectly influences aflatoxin accumulation in A. parasiticus. The disruption of fluP resulted in cotton-like hyphae with no spores and reduced growth rate, and the recovery of the wild-type allelefluPresultedinnomial hyphaewithnormal sporesandnonnalgrowthratesupportingthe proposed fianction of fluP in A. parasiticus. Other evidence which supports the association of fluP with hyphal cell development and sporulation is derived form the comparison of fluP transcript level in liquid and solid media. Normally, sporulation occurs only on solid media. Therefore, if fluP is associated with sponflafionflreconespondhgnansaiptlevdonsofidmediamaybeerpectedtoappearin greaterabtmdancethanthatinliquidmedia. Atthetwotimepointsanalyzed,thetranscripts fiomsolidmediadidappearingreaterabundancefllanthoseinfiqtndmedia However,the factthatfluPisexpressedinliquidmediamayalsomggeststhattlfisgeneisassociatedwith growth in general. Thepresenceorabsenceofthem’aD selectablemarkerhadnoobservableinfluenceonthe growth rate ofthefluP—disrupted transformant on YES or CAM. When hyphae, derived from SU-l (niaDYfluP‘) and the niaD mutant NR-3 (m'aDlfluF) of A. parasiticus were inoculated onto YES or CAM, there was no observable differences in growth rate between the two strains. ItislikelythatbecauseCAMandYES containmanydifl‘erentaltemativenitrogen sources, the two strains did not have to use nitrate reductase (encoded by niaD) for normal growth. However, when hyphae derived fi‘om SU-l (muDYfluP‘) and the fluP-disrupted transformant (m’aD‘lfluP) of A. parasiticus were inoculated onto YES or CAM, there was a great observable difl'erence in growth rate between the two strains. Therefore, the reduced 186 growth rate observed for the fluP-disrupted transformant (niaDY/IuP) on nonselective YES or CAM is the result of disruption of the fluP gene. The pattern of expression of the fluP gene and the B-tubulin gene were different during sporulation. Microtubules are one of the 3 types of filamentous structures (cytoskeleton) found in all enkaryotic cells (the other two are microfilarnents and intermediate filaments). In filamentous firngi, not only are microtubules necessary for the formation of cytoskeleton in general, but also involved in the formation of the mitotic spindle during mitosis. Microtubules consist of or-tubulin and B-tubulin. May et al. (1987) May and Morris (1988) suggested that B-tubulin participates in conidial development in Aspergillus nidulans by demonstrating that the expression of B-tubulin gene was developmentally regulated and directly correlated with the appearance of conidiating cell types. A large increase occurred in the rate of B-tubulin synthesis and there was an accumulation of the corresponding mRNA biosynthesis during sporulation in the filamentous firngus Blastocladiella emersonii (Da Silva and Juliani, 1988). Temperli et al. (1990) demonstrated that cytoplasmic microtubules arise from a nucleus-associated center (their density was highest near the nucleus) and extend into the proximal cytoplasm during mitosis by in situ immunolocalization using anti-B-tubulin monoclonal antibody. These data suggest that the increased accumulation of B-tubulin during sporulation is the result of the increased activity of mitosis in which B—tubulin is involved in the formation of the mitotic spindle. As mentioned above, fluP is also thought to be associated with sporulation in A. parasiticus. A comparison of the transcript level between the B-tubulin gene and fluP may 187 reveal more information about the function of fluP. For example, in liquid media, the accumulation ofthetranscript ofthe B—tubulingene decreased fi'om 24 to 72 h ofincubation, wlfiledm'hrgthesmnefimepaiodflreacanmflafionoffluPUanscfiptmcreased Thereappear to be at least two reasonable explanations for this observation. First, this experiment was conducted in liquid media and sporulation was inhibited. Therefore the requirement for a large ammefB-MmaymtbenecessmybemuseB-Maasasflnhrfldingmfitofmnofic spindleduringsporulation (notasasignal). Secondandmoreimportantly,notonlymayfluP (nuyencodeaéngbeassodMedwhhsponflafithdsonmybeassodfiedwithotha biologicalprocessesintheproducingfimgus. 'I'hereforeeventhoughsporulationisinhibited otherbiologicalprocessesstillneedtheexpressionoffluP. ThesedatasuggestthatfluP firnctions at an early stage of vegetative cell difl‘erentiation. The fluP gene isolated from A. parasiticus is firnctionally difl‘erent fi'om brIA, fluG, and flbA in Aspergillus nidulans. The developmental regulatory gene brIA in A. m'dukms is likely involved in controlling the switch from growth to development (Clutterbuck, 1969; Boylan er al., 1987; Lee and Admas, 1994b). But DNA sequence analysis predicted that brIA encodes a protein similar to a number of plant phytochromes (Grifith et al., 1994) which are very different fi'om polyketide synthase. Lee and Adams (1994a) described the fluG gene which is required for the activation of brlA expression and conidiophore development. TheFlquroteinispresentatnearlyconstantlevelsthroughouttheA. m’dulcmslifecycle. Sequence analysis of fluG predicted that a 864-codon fluG open reading frame shares 28% identity with prokaryotic glutamine synthetase I (Lee and Adams, 1994a). The M meducesflufiyhyphaewimmsporeslheflqumphmotypeiswppressedwhm 188 M mutant colonies are grown adjacent to colonies ofthe wild-type strain even ifthe two shainsareseparatedbydialysismembranewitha6—8kDacutofi‘(Adams etal, 1992). Another flufi‘y gene, flbA was found to be required in conidiophore developmart prior to activation of brIA. A 3.0-kb mRNA corresponding to flbA is present at low levels throughout the A. m’dukms asexual life cycle (Lee and Adams, 1994b). flbA encodes an Aspergillus nidrdam' regulator of G protein signaling (RGS) domain protein (Y u at al., 1996). The phamtypicdefeasoftheflbAddefionnmtamscouldbepmfiaflyranediedbygmwth onhigh osmolarity medium (e.g. 0.8 M NaCl) (Lee and Adams, 1994b). However, the fluP-disrupted phenotype of A. paran’ticus could not be suppressed whar being grown next to the wild-type orberemediedbygrowthonhighosmolaritymedium. ThereforefluPappearstobedifl‘erent fiom brIA, fluG, and flbA in Aspergillus nichdans with regard to their association with sporulation suggesting that fluP may be a new class of genes involved in fungal development. 'IheproteinerrcodedbyfluPirrprasiticusislikelyaPKSandthecorrespondingend product is likely a polyketide. Beck et a1. (1990) cloned the MSAS gene together with its flanking sequences (total 7131 bp) fi'om Penicilliwn patulum. Vifrthin this sequence the MSAS gene was identified as a 5322-bp long open reading flame. The transcript of fluP is about 6.0 kb in length which is similar in size to the transcript derived from the MSAS gene in P. patulum. A comparison of the deduced amino acid sequence of the 680—bp DNA fiagment of the fluP gene showed a great deal of similarity with B—ketoacyl synthase firnction domain of MSAS gene and other PKS genes. 189 The disrupted-fluP allele is not stable. Twenty percent of the colonies derived from cotton- like hyphae “reverted” to the wild genotype and phenotype suggesting that the disrupted fluP allele was genetically unstable. It is possible that reversion to the wild-type fluP allele couldoccurbycertaingeneticmechanisms(suchasasimpledeletionoftheinsertedDNA fiagment containing m'aD). It is also possible that the transferred hyphae were contaminated with hyphae and/or spores which contained wild-type nuclei although the data would argue that this scenario is unlikely. For example, if the transferred hyphae were contaminated with hyphae and/or spores which contained wild-type nuclei, the initial colony would predominately consist of wild-type hyphae with wild-type spores because of the faster growth rate of the wild-type hyphae. Similarly, the possibility that the transferred hyphae were heterokaryons is also not consistent with the data. Heterokaryons may exist immediately afler transformation, but serial nrycelial transferring should reduce the possrbility of a heterokarybn significantly. In- the unlikely event that a heterokaryon survived serial transfer, the colony which originated fi'om a heterokaryon would have normal hyphae and spores at the center of the colony. The normal hyphae (with spores) would continue to appear during colony growth. The cotton-like flufiy mycelium would only form a sector. The actual morphology of the colony observed suggests that neither of these possibilities is likely to be true. Therefore, the most reasonable explanation for the genetic instability of the knockout fluP appears to be genetic recombination within the fluP—disrupted allele to restore the wild-type allele. CAM and PDA had difi‘erent influences on the recovery of the wild-typefluP phenotype in the fluP-disrupted transformant. When hyphae of the fluP-disrupted transformant were 190 transferred onto CAM, 10% of the resulting colonies recovered the wild-type morphology and contained only the wild-typefluP allele WP); 20% gew as a mixture ofnomral hyphae plus cotton-like hyphae and contained the wild-type fluP allele and the disrupted fluP allele (fluP‘lfluP); 70% gew with only the cotton-like hyphae and contained only the disrupted fluP allele(fluP). Incontrast,whenhyphae ofthefluP-disrupted transformantweretransferred ontopotato dextroseagar(PDA), onlyonetypeofcolonyappeared. Such coloniesgewwith nmchlessaerialhyphaeandhadwidelydispersed flquporea IhaeforethefluPallelein disrupted transformants could “revert” to the wild-typefluP allele on either CAM or PDA, but in difl‘erent patterns. The fluP gene does not appear to be directly involved in aflatoxin biosynthesis. In liquid media (Reddy’s medium), the fluP transcript was detected in the 24-h sample while the transaiptofwr-lA(aswellasothergenesintheAFBr pathway), whichisdirectlyinvolvedin aflatoxin biosynthesis, was not detected in the 24-h sample (but detected in the 60-h sample). Thisdifi‘erencesuggeststhattheflngeneisnotdirectlyinvolvedinaflatoxinbiosynthesis. The decrease in AFBr accumulation of the fluP-disrupted transformant gown on YES agar could result from lack of sporulation and reduced gowth rate (decreased cell mass). Even though the overall AFB1 accumulation (per Petri dish) of the wild-type was approrn'mately lO-fold higher than that of the fluP—disrupted transformant, the accumulation of AFB1 per mg dry weight in the central region after 2 days incubation (which lacked sporulation) was essentially the same as the accumulation in any region of the fluP-disrupted transformant. In the wild-type, AFB1 accumulation per gam dry weight increased quickly only when sporulation appeared. Conidiophores and developing 191 conidia have been found to contain the highest concentration of Nor-1 protein (Chapter 4) and Ver-lA protein in a gowing colony of wild type on solid medium (Liang and Linz, unpublished data). These two proteins are directly involved in AFB1 biosynthesis. Therefore, fluP may directly influence vegetative cell differentiation and sporulation, which in turn influences aflatoxin biosynthesis in A. parasiticus. Future studies onfluPirrAparasiticusmayinclude: (a) acompleteanalysisofthe mdeofidesequenceofflqududingthededucedamumacidsequammdcompmison bawearflqudotherPKngeshNohedinhyphalgowthmdsponflafiminoflrafimgm narrow down the possrble product of fluP; (b) identification of the protein product of M; and (c) a study of the mechanism ofthe regulation of expression offluP. These studies may provide useful information for the development of new firngicides, improved agonomic practices, resistant crops (by genetic engineering or classical breeding), and biocontrol agents (such as nontoxigenic competitive fimgi) for control of aflatoxin contamination. In summary, disruption of fluP resulted in slow gowth with cotton-like hyphae, no sporulation, and reduced aflatoxin accumulation. The data suggest that fluP is associated with vegetative cell gowth, sporulation, and in turn indirectly reduces aflatoxin accumulation in A. parasiticras. CONCLUSIONS The data presented in this dissertation provided a further understanding of the mechanisms of regulation of aflatoxin biosynthesis and the general biology at the molecular level in the toxigenic filamentous fimgus Aspergillus parmiticus. These information may lead to the development of new fungicides, the improvement of agonomic practices, the development of resistant crops (by genetic engineering or classical breeding), and the development of biocontrol agents (such as nontoxigenic competitive fungi ) for control of aflatoxin contamination. 'I'hedataconfinnedthattheNor-lcprotein, derivedfromthenor-l cDNA, isakcto— reductase which converts the AFB1 pathway intermediate norsolorinic acid (NA) to averantin (AVN) in the presence of NADPH plus a factor(s) obtained fiom Eschreichia loci DHSor. The dataalso arggestthat anunidentified cofactor(s) mayalsobenecessaryforthenativeNor—l proteinfimctionandthatthenor-l geneisinvolvedinonlyonebranchoftheproposed alternative pathways for conversion ofNA to averufin (AVF). Polyclonal antibodies (PAb) raised against the Nor-1c protein were proven to be usefirl to monitor the accumulation of the native Nor-l protein. A nor-l/GUS reporter construct was also used to monitor the activity of the nor-1 promoter in A. parasiticus. The data liquid media, and to temporal and spatial regulation on solid media; and that the activity of the nor-l promoter is subjected to temporal and spatial regulation on solid media. 192 193 The data obtained by cell fractionation/Western blot analysis and in situ immrmolocalization suggest that the Nor-l protein is mainly localized in the cytosol of the fungal vegetative cell; the highest level of the Nor-l protein is associated with the developing irmnature conidium, high levels are associated with the conidiophore, lower levels are associated with the vegetative hyphalcelLandnoneisassociatedwiththematureconidium. Indicatedbyanor-l/GUS reporter construct, the localization of the activity of the nor-l promoter follows a temporal and spafialpatterninafugalcolony. Thedataalso arggestthatthelocalizationofNor-l protein accunmlationandthelocalizationofnor-l promoteractivityarecorrelatedtotheprocessof sporulation in A. parasiticus. Disruption of fluP, a putative polyketide synthase gene, resulted in reduced gowth rate, flufi‘y-mycelium (cotton-like hyphae), no sporulation, and reduced aflatoxin accumulation suggesting that fluP is associated with vegetative cell gowth, sporulation, and in turn indirectly influences aflatoxin synthesis in A. pwasiticus. APPENDICES APPENDIX A APPENDIX A CLONING OF THREE EeoRI DNA FRAGMENTS FROM COSMID Nor-A AND ANALYSISOFTHEIRTRANSCRIPTS Three difl‘erent but adjacent EcoRI DNA fi'agments (in the order of 2.8-kb, 4.0-kb and 4.6- kb) fi‘om an aflatoxin gene cluster (cloned in cosmid Nor-A, Figure 1-23) in Aspergillus pwasiticushavebeentriedtobeclonedandmapped. Onlytwo(the2.8-kbEcoRIfiagnent, andflre4.0-kbEcoRIfiagnem)offlremweresuccessfifllyclonedandmapped Butthethird one(a4.6-kbEcoRIfiagment)wasunabletobeclonedforunknownreasoninatleastfive roundsofexperiments. In Northern blot analysis, a 7.5-kb transcript was detected using the 2.8-kb anRI DNA fragmentasaprobe,anda7.5-kbtranscriptplusa6.5-kbfianscriptweredetectedusingthe 4.0-kbEmRIDNAfiagnemasapmbeinApmusifiarsgowninanaflatordn-indudng medium. Theacanmrlafionsofthetwouanscriptshadthesamepattemasthatofthe accrrnudafionsofmr—larrdver-lfiarrscriptsinApm'asiticus. Thesedatasuggestthatthetwo transcripts were related to or involved in aflatoxin biosynthesis. 194 APPENDIX B APPENDIX B IDENTIFICATION OF VERSICONAL CY CLASE GENE Isolation and identification of the gene encoding versiconal cyclase (converting versiconal to versicolorinBintheAFBrpathway)havebeentriedforaperiodoftime. Thisexperimentwas startedwithaknownaminoacidsequenceofversiconalcyclase. Amixtureofl8-mer mrcleotidesandambrtureofl7-mermrcleotideswerepreparedaccordingtoapartofthe aminoacidsequenceofversiconalcyclase. Boththe18mer'sandthel7merswereusedas probestoprobetheAFBrgeneclusterclonedincosmidNor-A. A400-prcoRI/X7ral fiagment(ina4.0-kbEcoRIDNAfiagment)incosmidNor—Awasfoundtohybridizethe18- merprobesinSouthernblotanalysis. This400—bpfiagnentwassequenced. Sequencedata slwwedmaflfisfiagnanwmainedasirmursequaweasomofthe18maprobesThedaa suggest the gene encoding versiconal cyclase was not included in cosmid Nor-A. 195 APPENDIX C APPENDIX C A MODIFIED PROCEDURE FOR CRUDE EXTRACT PREPARATION FROM FUN GAL CELLS CertainProteinsinvolvedinAFBrbiosymhesisareenremelyunstable. Thereforethe qualityofcrudeermactfiomtoxigenicfimgalcellsisumallyone ofthemostimportantfirctors whichinfluenceflreremltsofenzymaficassayorWesternblotanalysis Grinding firngal cells under liquid nitrogen followed by addition of extraction bufl‘er containing protease inhibitors is a common protocol used by many researchers. Two ofthe commonly used protease inhibitors are phenol methylsulfonyl fluoride (PMSF) and aprotinin. PMSF is quickly inactivated in the aqueous solution, and aprotinin can be precipitated at low temperature. Grinding fungal cells usually cannot destroy all organelles and most proteases are enclosed in organelles. Therefore, the procedure described above cannot get all proteases inhibited. The crude extract prepared in this way is stored in the freezer (at -80°C) and later is taken out and thawed for use. The freeze- and-thaw procedure will destroy the intact organelles and proteases inside will be released. This phenomenon has been ignored in most of published literature. To get a better result of enzymatic assay or Western blot analysis, protease inhibitors can be added again afier each fieeze-and-thaw step as soon as possible. 196 LIST OF REFERENCES LIST OF REFERENCES Abdollahi, A., and R. L. Buchanan. 1981. Regulation of aflatoxin biosynthesis: induction of aflatoxin production by various carbohydrates. J. Food Sci. 46: 633-635. Abdou, R. F., S. E. Megalla, and S. G. Azab. 1984. Mutagenic efl‘ects of aflatoxin B1 and G1 on the Egyptian cotton leaf-worm, Spodoptera Iittoralis (Boisd.). Mycopatholo. 88: 23-26. Abraham, W. R., and H. A. Arl'manu. 1990. 12,13-dihydroxy-fumitremorgin C from Aspergillusfimigatus. Phytochem. 29: 1025-1026. Adams, T. IL, W. A. Ride, L. N. Yager, and B. N. Lee. 1992. Isolation of a gene required for progammed initiation of development by Aspergillus nidulans. Mol. Cell. Biol. 12: 3827-3833. AdamsomRH.P.Conea,andD.W.Dalgard.1973.0ccurrenceofaprimaryliver careinomainaRhesusmonkeyfedaflatoxin Br. J. NatL CancerInst. 50: 549-553. Adgigitov, F. 1., L. P. Kosichenko, P. G. Popandopulo, D. A. Gubeladze, and Z. A. Djemilev. 1984. Frequency of chromosome aberrations in bone marrow of monkeys and their F1 after aflatoxin Br injection. Exp. Pathol. 26: 163-169. Alexopoulos, C. J. and Mims C. W. (Eds). 1979. Introductory mycology. pp. 292-293. John Wiley & Sons. New York, New York. Amitani, R., G. Taylor, E. N. Elais, C. Llewellyn-Jones, J. Mitchell, F. Kuze, P. J. Cole, and R. Wilson. 1995. Purification and characterization of factors produced by Aspergillusfirmigatus which afi‘ect human ciliated respiratory epithelium. Infect. Immun. 63: 3266-3271. Anderson, J. A., and C. II. Chung. 1990. Conversion of versiconal acetate to versiconal and versicolorin C in extracts fiom Aspergillus parasiticus. Mycopatholo. 110: 31-35. Arseculeratne, S. N., L. M. de Silva, S. Wijesundera, and C. H. Bauduuatha. 1969. Coconut as a medium for the experimental production of aflatoxin. Appl. Microbiol. 18: 88-94. 197 198 Aucamp, P. J., and C. W. Holzapfel. 1970. Polyhydroxyanthraquinones from Aspergillus versicolor, Aspergillus nidulans and Bipolaris spp.: their significance in relation to biogenetic theories on aflatoxin Br. S. Afi'. Chem. Inst. J. 23: 40-56. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1987. Escherichia coli, plasmids, and bacteriophages, in “Current protocols in molecular biology”. pp. 1.0.3-l.15.7. John Wiley & Sons, New York. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995a. Preparation of polyclonal antisera, in “Current protocols in molecular biology”. pp. 11.12.1-11.13.3. John Wiley & Sons, New York. Ausubel. F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995b. One-dimensional gel electrophoresis of protein, in “Current protocols in molecular biology”. pp. 1021-10218. John Wiley & Sons, New York. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995c. Immunoafinity chromatography, in “Current protocols in molecular biology”. pp. 10.1.1-10.l 1.6. John Wiley & Sons, New York. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995d. Preparation of genomic DNA fi'om mammalian tissue, in “Current protocols in molecular biology”. pp. 2.2.1-1.2.2. John Wiley & Sons, New York. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995e. Preparation and analysis of RNA, in “Current protocols in molecular biology”. pp. 4.0.3-4.lO.10. John Wiley & Sons, New York. Ausubel, F. M., R. Brent, R. E. Kingston, D. D. Moore, J. G. Seidman, J. A. Smith, and K. Struhl (Eds). 1995f. Analysis of DNA sequence by blotting and hybridization, in “Current protocols in molecular biology”. pp. 6.0.3 -6. 12.7. John Wiley & Sons, New York. Autrup,H.,J. M.Essigmann,R. G. Croy,B. F. Trump, G. N. Wogan, and C. C. Harr’n. 1979. Metabolism of aflatoxin B1 and identification of the major aflatoxin Br-DNA adducts formedincultmedhrmanbronclmsandcolon. CancerRea39: 694-698. Baldwin, J. E., and E. Abraham. 1988. The biosynthesis of penicillins and cephalosporins. Nat. Prod. Rep. J. Curr. Dev. Bio. Org. Chem. 5: 129-145. Barbesgaard, P., H. P. Heldt-Hansen, and B. Diderichsen. 1992. On the safety of Aspergillus oryzae: a review. Appl. Microbiol. Biotechnol. 36: 569-5 72. 199 Barredo, J. L., P. van Solingen, B. Din, E. Alvarez, J. M. Cantoral, A. Kattevilder, E. B. Smaal, M. A. Groenen, A. E. Veenstra, and J. F. Martin. 1989. Cloning and characterization of the acyl-coenzyme A: 6-aminopenicillanic-acid-acyltransferase gene of Penicillin»: charsogenum. Gene 83: 291-3 00. Bauer-Hofmann, R., A. Buehmann, A. S. Wright, and M. Schwarz. 1990. Mutations in the Ha-ras proto-oncogene in spontaneous and chemically induced liver tumours of the CF 1 mouse. Carcinogenesis 11: 1875-1877. Bayman, P., and P. J. Cotty, 1990. Triadirnenol stimulates aflatoxin production by Awaits-flaws in vitro. Mycol. Res. Cambridge, Cambridge University Press. 94: 1023- 1025. Bayman, P., and P. J. Cotty. 1991. Vegetative compatibility and genetic diversity in the Aspergillus flaws population of a single field. Can. J. Bot. 69: 1007-1711. Bayman, P., and P. J. Cotty. 1993. Genetic diversity in Aspergillus flaws. association wtith aflatoxin production and morphology. Can. J. Bot. 71: 23-31. Beck, J., S. Riplra, A. Siegner, E. Schiltz, and E. Schweizer. 1990. The multifunctional 6-methylsalicylic acid synthase gene of Penicillium patulum. Its gene structure relative to that of other polyketide synthases. Eur. J. Biochem. 192: 487-498. Bennett, J. W., and S. B. Christensen. 1983. New perspective on aflatoxin biosynthesis. Adv. Appl. Microbiol. 29: 53-92. Bennett, J. W., L. S. Lee, S. M. Shoss, and G. H. Boudreaux. 1980. Identification of averantin as an aflatoxin Br precursor: placement in the biosynthetic pathway. Appl. Environ. Microbiol. 39: 835-839. Bennett, J. W., P. C. Horowitz, and L. S. Lee. 1979. Production of sclerotia by aflatoxigenic and nonaflatoxigenic strains of Aspergillus flaws and A. parsiticus. Mycol. 71: 415-422. ‘ Bhatnagar, D., A. H. Ullah, and T. E. Cleveland. 1988. Purification and characterization of a methyltransferase fi'om Aspergillus parasiticus SRRC 163 involved in aflatoxin biosynthetic pathway. Prep. Biochem. 18: 321-349. Bhatnagar, D., and T. E. Cleveland. 1988. Fate of the methyl group during the conversion of sterigmatocystin into O-methylsterigmatocystin and aflatoxin Br by cell-fiee preparations of Aspergillus parasiticus. Biochimie. 70: 743-747. Bhatnagar, D., and T. E. Cleveland. 1990. Purification and characterization of a reductase fi'om Aspergillus parasiticus SRRC 2043 involved in aflatoxin biosynthesis. FASEB J. 4: A2164. 200 Bhatnagar, D., S. P. McCormick, 1.. S. Lee, and R. A. Hill. 1987. Identification of O- methylsterigmatocystin as an aflatoxin B1 and Gr precursor in Aspergillus parasiticus. Appl. Environ. Microbiol. 53: 1028-1033. Bhatnagar, D., T. E. Cleveland, and D. G. L Kingston. 1991. Enzymological evidence for separate pathways for aflatoxin Br and Bz biosynthesis. Biochem. 30: 4343- 4350. Bhatnagar, D., T. E. Cleveland, and E. B. Lillehoj. 1989. Enzymes in aflatoxin Br biosynthesis: strategies for identifying pertinent genes. Mycopatholo. 107: 75-83. Bhatnagar, R. L., S. Ahmad, K. G. Mukerji, and T. A. Subramanian. 1986. Pyridine nucleotides and redox state regulation of aflatoxin biosynthesis in Aspergillus pwasifim NRRL 3240. J. Appl. Bacteriol. 60: 135-141. Bhatnager, D., K. C. Erhlich, and T. E. Cleveland. 1992. Oxidation-reduction reactions in biosynthesis of secondary metabolites, in “Handbook of applied mycology” (Bhatnagar, D., E. B. Lillehoj, and D. K. Aroa Eds). 5: 255-286. Marcel Dekker Inc. New York. Biollaz, M., G. Biichi, and G. Milne. 1970. The biosynthesis of aflatoxin. J. Amer. Chem. Soc. 92: 1035-1043. Birch, A. J., and F. W. Donovan. 1953. Studies in relation to biosynthesis. L Some possible routes to derivatives of orcinol and phloroglucinol. AustraL J. Chem. 6: 360- 368. Birch, A. J., and S. F. Hussain. 1969. Studies in relation to biosynthesis. 38. A preliminary study of fumagillin. J. Chem. Soc. Perkin. L 1: 1473-1474. Birch, A. J., P. Fitton, E. Pride, A. J. Ryan, H. Smith, and W. B. Whalley. 1958. Studies in relation to biosynthesis. Part XVII. Sclerotiorin, citrinin, and citomycetin. J. Chem. Soc. IV: 4576-4581. Birch, A. J., R. A. Massy-Westropp, and C. J. Moye. 1955. Studies in relation to biosynthesis VII. 2-Hydroxy-6-methylbenzoic acid in Penicillium griseofidvum Dierckx. Aust. J. Chem. 8: 539-544. Birkinshaw, J. 11., A. Bracken, S. E. Michael, and H. Raistriek. 1943. Patulin in the common cold. Collaborative research on a derivative of Penicillin»: patulin» Bainier. Lancet 245: 625-634. Birkinshaw, J. E., J. C. Roberts, and P. Rofl'ey. 1966. Studies in mycological chemistry. XIX. ”Product B" (averantin) [1,3,6,8-tetrahydroxy-2-(1-hydroxyhexyl)-anthraquinone], a pigment fi'om Aspergillus versicolor (Vuillemin) Tiraboschi. J. Chem. Soc. Perkin. 1. 9: 855-857. 20] Blackburn, J. M., J. D. Sutherland, and J. E. Baldwin. 1995. A heuristic approach to the analysis of enzymatic catalysis: reaction of 6-(1.-a-aminoadipoyl)-L-cysteinyl-D-a- aminobutyrate and 6-(Ira-aminoadipoyl)-L-cysteinyl-D-allylglycine catalyzed by isopenicillin N synthase isozymes. Biochem. 34: 7548-7562. Bloom, S. E. 1984. Sister chromatid exchange studies in the chick embryo and neonate: actions of mutagens in a developing system. Basic Life Sci. 29: 509-533. Blount, W. P. 1961. Turkey "X" disease. J. Brit. Turkey Fed. 9: 52-54. Boylan, M. T., P. M. Mirabito, C. E. Willett, C. R. Zimmerman, and W. E. Timberlake. 1987. Isolation and physical characterization of three essential conidiation genes from Aspergillus nidulans. Mol. Cell Biol. 7: 3113-3118. Brown, D. W., F. M. Hauser, R. Tommasi, S. Corlett, and J. J. Salvo. 1993. Structural elucidation of a putative conidial pigment intermediate in Aspergillus parasiticus. Tet. Lett. 34: 419-422. Brown, D. W., J. H. Yu, H. S. Kelkar, M. Fernandes, T. C. Neshitt, N. P. Keller, T. H. Adams, and T. J. Leonard. 1996. Twenty-five coregulated transcripts define a sterigmatocystin gene cluster in Aspergillus nidulans. Proc. Natl. Acad. Sci. 93: 1418- 1422. Brown, D. W., and J. J. Salvo. 1994. Isolation and characterization of sexual spore pigments from Aspergillus nidulans. Appl. Environ. Microbiol. 60: 979-983. Brown, R. L., P. J. Cotty, and T. E. Cleveland. 1991. Reduction in aflatoxin content of maize by atoxigenic strains of Aspergillus flaws. J. Food Prot. 54: 623-626. Brown, R. L., T. E. Cleveland, P. J. Cotty, and J. E. Mellon. 1992. Spread of Aspergillus flaws in cotton bolls, decay of intercarpellary membranes and production of fungal pectinases. Phytopathol. 82: 462-467. Buchanan, R. L., and D. F. Lewis. 1984. Regulation of aflatoxin biosynthesis: effect of glucose on activities of various glycolytic enzymes. Appl. Environ. Microbiol. 48: 306- 310. Buchanan, R. L., S. B. Jones, W. V. Gerasimowicz, L. L. Zaika, H. G. Stahl, and L. A. Ocker. 1987. Regulation of aflatoxin biosynthesis: assessment of the role of cellular energy status as a regulator of the induction of aflatoxin production. Appl. Environ. Microbiol. 53: 1224-1231. Buchi, G., K. C. Luk, B. Kobbe, and J. M. Townsend. 1977. Four new mycotoxins of Aspergillus clavatus related to tryptoquivaline [collected from mold-damaged rice] related to trptoquivaline. J. Org. Chem. 42: 244-246. 202 Buchi, G., Y. Kitaura, S. S. Yuan, H. E. Wright, J. Clardy, A. L. Demain, T. Ginsukon, N. Hunt, and G. N. Wogan. 1973. Structure of cytochalasin B, a toxic metabolite of Aspergillus clavatus. J. Amer. Chem. Soc. 95: 5423-5425. Burnham, M. K. R., A. J. Earl, J. H. Bull, D. J. Smith, and G. Turner. 1989. DNA encoding ACV synthetase. European Patent Application 8831165.0. Busby, W. F. Jr., and G. N. Wogan. 1984. Aflatoxins, in “Chemical carcinogens” (Searle, C. E. Ed). pp. 945-1136. American Chemial Soceity. Washington D.C. Butler, W. H., and J. S. Wigglesworth. 1966. Efi‘ects of afiatoxin Br on the pregnant rat. Br. J. Exp. Pathol. 47: 242-247. Button, M. F. 1988. Enzymes and aflatoxin biosynthesis. MicrobioL Rev. 52: 274-295. Caffrey, P., D. J. Bevitt, J. Staunton, and P. F. Leadlay. 1992. Identification of DEBS l, DEBS 2 and DEBS 3, the multienzyme polypeptides of the erythromycin-producing polyketide synthase fi'om Saccharopolyspora erythraea. FEBS Lett. 304: 225-228. Cane, D. E., and R. H. Levin. 1976. Application of carbon-13 magnetic resonance to isoprenoid biosynthesis. II. Ovalicin and the use of doubly labeled mevalonate. J. Am. Chem. Soc. 98: 1183-1188. Carnaghan, R. B. A. 1965. Hepatic tumors in ducks fed a low level of toxin groundnut meaL Nature 208: 308. Carson, D. B., and J. J. Cooney. 1990. Microbodies in fungi: a review. J. Ind. Microbiol. 6: 1-8. Cary, J. W., M. Wright, D. Bhatnagar, R. Lee, and F. S. Chu. 1996. Molecular characterization of an Aspergillus parasiticus dehydrogenase gene, norA, located on the aflatoxin biosynthesis gene cluster. Appl. Environ. Microbiol. 62: 360-3 66. Chang, P.-K., K. C. Ehrlich, J. J. Yu, D. Bhatnagar, and T. E. Cleveland. 1995. Increased expression of Aspergillus parasiticus q/IR, encoding a sequence-specific DNA- binding protein, relieves nitrate inhibition of aflatoxin biosynthesis. Appl. Environ. Microbiol. 61: 2372-2377. Chang, P.-K., C. D. Skory, and J. E. Linz. 1992. Cloning of a gene associated with aflatoxin Br biosynthesis in Aspergillus parasiticus. Curr. Genet. 21: 231-233. Chang, P.-K., J. W. Cary, D. Bhatnagar, T. E. Cleveland, J. W. Bennett, J. E. Linz, C. P. Woloshik, and G. A. Payne. 1993. Cloning of the Aspergillus paratr'cicus ape-2 gene associated with the regulation of aflatoxin biosynthesis. Appl. Environ. Microbiol. 59: 3273-3279. 203 Chang, P.-K., J. W. Cary, J. Yu, D. Bhatnagar, and T. E. Cleveland. 1995. The Aspergillus parasiticus polyketide synthase gene pksA, a homolog of Aspergillus nidulans wA, is required for aflatoxin B1 biosynthesis. MGG Mol. Gen. Genet. 248: 270-277. Chater, K. F. 1992. Genetic regulation of secondary metabolic pathways in Streptomyces, in “Secondary metabolites: their function and evolution. Ciba. Found. Symp. 171” (Chadwick, D. J. and J. E. Whelan Eds). pp.144-156. John Wiley & Sons, New York. Chaves—Carballo, E., R. D. Ellefson, and M. R. Gomez. 1976. An aflatoxin in the liver of a patient with Raye-Johnson syndrome. Mayo. Clin. Proc. 51: 48-50. Chet, L, and Henis Y. 1975. Sclerotia! morphogenesis in firngi. Annu. Rev. Phytopathol. 13: 169-192. Chexal, K. K., J. P. Spinger, J. Clardy, R. J. Cole, J. W. Kirksey, J. W. Dorner, H. G. Cutler, and B. J. Strawter. 1976. Austin, a novel polyis0prenoid mycotoxin fiom Aspergillus astus [letter]. J. Am. Chem. Soc. 98: 6748. Chu, F. S. 1991. Mycotoxins: food contamination, mechanism, carcinogenic potential and preventive measures. Mutat. Res. 259: 291-306. Chung, C. H., K. H. Ling, S. S. Tung, and T. C. Tung. 1971. Study on fimgi of the stored unhulled rice to Taiwan. (2) Aflatoxin Br like compounds from the culture of Aspergillus genus. J. Formosan Med. Assoc. 70: 258-266. Chuturgoon, A. A., and M. F. Dutton. 1991. The afinity purification and characterization of a dehydrogenase from Aspergillus parasitic-us involved in aflatoxin Br biosynthesis. Prep. Biocherrr. 21: 125-140. Clardy, J., J. P. Springer, G. Buchi, K. Matsuo, and R. Wightman. 1975. Letter: Tryptoquivaline and tryptoquivalone, two tremorgenic metabolites of Aspergillus clavaius. J. Am. Chem. Soc. 97: 663-665. Cleveland, T. E. 1989. Conversion of dihydro-O-methylsterigmatocystin to aflatoxin B2 byAspergiIlusparariticua Arch. Environ. Contam. Toxicol. 18: 429-433. Cleveland, T. E., and D. Bhatnagar. 1987. Individual reaction requirements of two enzyme activities, isolated fi'om Amergillus parasiticus, which together catalyze conversion of sterigmatocystin to aflatoxin Br. Can. J. Microbiol. 33: 1108-1112. Cleveland, T. E., D. Bhatnagar, C. J. Foell, and S. P. McCormick. 1987. Conversion of a new metabolite to aflatoxin B: by Aspergillus parasiticus. Appl. Environ. Microbiol. 3: 2804-2807. 204 Clevstrom, G. and H. Ljunggren. 1985. Aflatoxin formation and the dual phenomenon in Aspergillus flaws Link. Mycopatholo. 92: 129-139. Clutterbuck, A. J. 1969. A mutational analysis of conidial development in Aspergillus nidulans. Genet. 63: 317-327. Cohen, G, D. Dembiec, and J. Marcus. 1970. Measurement of catalase activity in tissue extracts. Anal. Biochem. 34: 30-38. Cole, R. J., and R. H. Cox (Eds). 1981. Handbook of toxic fungal metabolites. pp.111- 112. Academic Press Inc. New York. Cole, R. J., J. W. Dorner, J. P. Springer, and R. H. Cox. 1981. Indole metabolites fiom a strain of Aspergillus flaws. J. Afiic. Food Chem. 29: 293-295. Collie, N., and W. S. Myers. 1893. The formation of orcinol and other condensation products fi'om dehydracetic acid. J. Chem. Soc. 63: 122-128. Cotty, P. J. 1988. Aflatoxin and sclerotial production by Aspergillusflavus, influence on pH. Phytopathol. 78:1250-1253. Cotty, P. J. 1989. Virulence and cultural characteristics of two Aspergillusflavus strains pathogenic on cotton. Phytopathol. 79: 808-814. Cotty, P. J. 1990. Efi‘ect of atoxigenic strains of Aspergillus flavus on aflatoxin contamination of developing cottonseed. Plant Dis. 64: 233-23 5. Cotty, P. J. 1992a. Use of native Aspergillus flaws strains to prevent aflatoxin contamination. United States Patent 5,171,686. Cotty, P. J. 1992b. Soil populations of Aspergillus flaws group fungi in agricultural fields in Alabama, Arizona, Louisianan, and Mississippi. Phytophathol. 82: 1064. Cotty, P. J. 1992c. Aspergillus flaws, wild intruder or domesticated fiee loader, in “Aflatoxin elimination whorkshiop” (Robens, J. F. Ed). pp. 28. Agricultural Research Service, Beltsville. Cotty, P. J., and P. Bayman. 1993. Competitive exclusion of a toxigenic strain of Aspergillusflavus by an atoxigenic strain. Phytopathol. 83: 1283-1287. Coulomhe, R. A. Jr., and R. P. Sharma. 1985. Efi‘ect of repeated dietary exposure of afiatoxin B1 on brain biogenic amines and metabolites in the rat. Toxicol. Appl. Pharmacol. 80: 496-501. 205 Courtemanche, C., and A. Anderson. 1994. Shuttle-vector mutagenesis by aflatoxin Br in human cells: effects of sequence context on the supF mutational spectrum. Mutat. Res. 306: 143-151. Crosby, J., D. H. Sherman, M. J. Bibb, W. P. Revill, D. A. Hopwood, and T. J. Simpson. 1995. Polyketide synthase acyl carrier proteins fi'om Streptomyces: expression in Escherichia coli, purification and partial characterisation. Biochim. Biophys. Acta 1251: 32-42. Da Silva, A. M., and M. H. Juliani. 1988. Regulation of tubulin and actin synthesis and accumulation during Blastocladiella emersonii development. Cell Difi‘er. 24: 45-54. Dafalla,R.,Y.M.Hassan,andS.E.I.Adam.1987.Fattyhemorrhagicliverandlddney syndmmembreedingherscausedbyaflatordnBrmrdheatsnessmflreSudanVet. HumToxicol. 29: 252-254. Davies, J. 1992. Introduction, in “Secondary metabolites: their firnction and evolution. Ciba. Found. Symp. 171” (Chadwick, D. J. and J. E. Whelan Eds). pp.l-2. John Wiley & Sons, New York. De Duve, C., and H. Beaufay. 1981. A short history of tissue fi'actionation. J. Cell Biol. 91: 293 s-2993. Deacon, J. W. (Ed). 1980. Sclerotia, in “Introduction to modern mycology”. pp. 60-61. John Wiley & Sons. New Yowlg New Yowk. Dimroth, P., H. Walter, and F. Lynen. 1970. Biosynthesis of 6-methylsalicylic acid. Eur. J. Biochem. 13: 98-110. Donkersloot, J. A., R. L Matele, and S. S. Yang. 1972. Isolation of averufin fi'om a mutant of Aspergillus parasiticus impaired in aflatoxin biosynthesis. Bioche. Biophys. Res. Comm. 47:1051-1055. Dorner, J. W. 1983. Production of cyclopiazonic acid by Aspergillus tamarii Kita. Appl. Environ. Microbiol. 46: 143 5-1437. Dorner, J. W., R. J. Cole, and P. D. Blankenship. 1992. Use of a biocompetitive agent to control preharvest aflatoxin in drought stressed peanuts. J. Food Prot. 55: 888- 892. Dorner, J. W., R. J. Cole, and U. L. Diener. 1984. The relationship of Aspergillus flaws and Aspergillus parastiticus with reference to production of aflatoxins and cyclopiazonic acid. Mycopatholo. 87:13-15. 206 Doyle, M. P., and E. H. Marth. 1978. Aflatoxin is degraded by mycelia fiom toxigenic and nontoxigentic strains of Aspergilli grown on different substrates. Mycopatholo. 63: 145-153. Dulaney, E. L. 1947. Some aspects of penicillin production by Aspergillus nidulans. MycoL 39: 570-582. Dutton, M. F. 1988. Enzymes and aflatoxin biosynthesis. Microbiol. Rev. 52: 274- 295. Edda, G. T., K. P. C. Nair, and C. F. Simpson. 1973. Efl‘ect of aflatoxin Br on resistance in poultry against cecal coccidiosis and Marek's disease. Am. J. Vet. Res. 34: 819-826. Elis. J., and J. A. DiPaolo. 1967. Aflatoxin Br. Induction of malformations. Arch. Pathol. 83: 53-57. Ellis, W. O., J. P. Smith, B. K. Simpson, and J. H. Oldham. 1991. Aflatoxin in food: occurrence, biosynthesis, efi‘ects on organisms, detection, and methods of control. Crit. Rev. Food Sci. Nutr. 30: 403-439. Epstein,S.M.,B.Bartus,andE.Farber.1969.Renalepithelialneoplasmsinducedinmale Wrstarrats by oral aflatoxin B1. CancerRes. 29:1045-1050. Essigmann, J. M., R. G. Croy, R. A. Bennett, and G. N. Wogan. 1982. Metabolic activation of aflatoxin Br: patterns of DNA adduct formation, removal, and excretion on relation to carcinogenesis. Drug. Metab. Rev. 13: 581-602. Feitelson, J. S., F. Malpartida, and D. A. Hopwood. 1985. Genetic and biochemical characterization of the red gene cluster of Streptomyces coelicolor A3(2). J. Gen. Microbiol. 131: 2431-2441. Feng, G. H., and T. J. Leonard. 1995. Characterization of the polyketide synthase gene (plle) required for aflatoxin biosynthesis in Aspergillus parasiticus. J. Bacteriol. 177: 6246-6254. Fitzell, D. L., R. Singh, D. P. Hsieh, and E. L. Motell. 1977. Nuclear magnetic resonance identification of versiconal hemiacetal acetate as an intermediate in aflatoxin biosynthesis. J. Agric. Food Chem. 25: 1193-1197. Foster, P. L., J. D. Groopman, and E. Eisenstadt. 1988. Induction of base substitution mutations by aflatoxin Br is chB dependent in Escherichia coli. J. Bacteriol. l7 0: 3415-3420. 207 Franck, B. 1980. The biosynthesis of the ergochromes, in “The biosynthesis of mycotoxins: a study in secondary metabolism”( Steyn, P. S. Ed). pp. 157-191. Academic Press. New York. Frisvad, J. C. 1988. Fungal species and their specific production of mycotoxins, in “ Introduciton to food-borne fungi” (Samson, R. A., and E. S. van Reenen-Hoekstra Eds). pp. 23 9-249. 3rd Ed. Centraalbureau voor Schimmelcultures, Baarn, The Netherlands. Gallagher, R. T., and B. J. Wilson. 1978. Aflatrem, the tremorgenic mycotoxin fi'om Aspergillusflams. Mycopatholo. 66: 183-185. Gallagher, R. T., J. Finer, J. Clardy, A. Leutwiler, F. Weibel, W. Acklin, and D. Arigoni. 1980. Paspalinine, a tremorgenic metabolite fiom Claviceps paspali Stevens and Hall. Tet. Lett. 21: 235-238. Gallagher, R. T., J. L. Richard, H. M. Stahr, and R. J. Cole. 1978. Cyclopiazonic acid production by aflatoxigenic and non-aflatoxigenic strains of Aspergillus flavus. Mycopatholo. 66: 31-36. Ganz, T., M. E. Selsted, D. Szklarek, S. S. Harwig, K. Daher, D. F. Bainton, and R. L Lehrer. 1985. Defensins. Natural peptide antibiotics of human neutrophils. J. Clin. Invest. 76: 1427-1435. Giambrone, J. J., U. L. Diener, N. D. Davis, V. S. Panangala, and F. J. Hoerr. 1985. Efi‘ects of purified aflatoxin on broiler chickens. Poult. Sci. 64: 852-858. Goerttler, K., H. Lohrke, H. J. Schweizer, and B. Hesse. 1980. Efi‘ects of aflatoxin Br on pregnant inbred Sprague-Dawley rats and thdr F 1 generation. A contribution to transplacental carcinogenesis. J. Natl. Cancer. Inst. 64: 1349-1354. Gopalan, C., P. G. Tulpule, and D. Krishnamurthi. 1972. Induction of hepatic carcinoma with aflatoxin in the Rhesus monkey. Food Cosmet Toxicol. 10: 519-521. German, D. P., M. S. Kang, T. E. Cleveland, and R. L. Hutchinson. 1992. Field aflatoxin production by Aspergillusflavus and A. parasiticus on maize kernels. Euphytica (Netherlands) 61: 187-191. Gorst-Allman, C. P., P. S. Steyn, P. L. Wessels, and B. de Scott. 1978. Carbon-13 nuclear magnetic resonance assignments and biosynthesis of veriscolorin A in Aspergillus parasiticus. J. Chem. Soc. Perkin.Trans. L 9: 961-964. Goze, A., A. Sarsin, Y. Moule, and R. Devoret. 1975. Induction and mutagenesis of prophage lambda in Escherichia coli K12 by metabolites of aflatoxin Br. Mutat. Res. 28: 1-7. 208 Graf, U., F. E. Wurgler, A. J. Katz, H. Frei, H. Juon, C. B. Hall, and P. G. Kale. 1984. Somatic mutation and recombination test in Drosophila melanogaster. Environ. Mutagen. 6: 153-188. Griffith, G. W.., G. L Jenkins, E. Milner-Whitc, and A. J. Clutterbuck. 1994. Homology at the amino acid level between plant phytochromes and a regulator of asexual sporulation in Emericella (Aspergillns) nidulans. Photochem. photobiol. 59: 252-256. Groopman, J. D., L. G. Cain, and T. W. Kensler. 1988. Aflatoxin exposure in human populations: measurements and relationship to cancer. Crit. Rev. Toxicol. 19: 113-145. Gupta, S. K., K. K. Maggon, and T. A. Venkitasubramanian. 1977. Regulation of aflatoxin biosynthesis. L Comparative study of mycelial composition and glycolysis in aflatoxigen and nonaflatoxigenic strains. Microbios. 18: 27-33. Hamasaki, T., K. Matsui, K. Isono, and Y. Hatsuda. 1973. A new metabolite fiom Aspergillns versicolor. Agric. Biol. Chem. 37: 1769-1770. Harper, A. E. 1965. Glucose-6-phosphatase, in “Methods of enzymatic analysis” (Bergmeyer, H. U. Ed). pp. 788-792. Academic Press, New York. Harris, J. L. 1986. Modified method for fimgal slide culture. J. Clin. Microbiol. 24: 460-461. Hatsuda, Y., and S. Kuyama. 1954. Studies on metabolic products of Aspergillns versicolor, I. Cultivation of Aspergillns versicolor, isolation and purification of metabolic products. J. Agric. Chem. Soc. Jpn. 28: 989-993. Hayes, R. B., J. P. Van Nieuwenhuize, J. W. Raatgever, and F. J. W. Ten-Kate. 1984. Aflatoxin exposures in the industrial setting: an epidemological study of mortality [Dutch oilpress workers]. Food Chem. Toxicol. 22: 39-43. Heller, K., C. Schultz, R. Loser, and R. Roschenthaler. 1975. Furnitremorgin A from Aspergillnsfirmigatns. Chem. Pharm. Bull. Tokyo, 28: 245-254. Henderberg, A., J. W. Bennett, and L. S. Lee. 1988. Biosynthetic origin of aflatoxin Gr: confirmation of sterigmatocystin and lack of confirmation of aflatoxin Br as precursors. J. Gen. Microbiol. 134: 661-667. Herrold, K. M. 1969. Aflatoxin induced lesions in Syrian hamsters. Br. J. Cancer. 23: 655- 660. Hetherington, A. C., and H. Raistrick. 1931. Studies in the biochemistry of microorganisms. Part XIV. On the production and chemical constitution of a new yellow coloring matter, citrinin, produced from glucose by Penicillin»: ctirinnm Thom. Phil. Trans. R. Soc. London, Ser. B. 220: 269-295. 209 Hill, R. A., D. M. Wilson, W. W. McMillian, N. W. Widstrom, R. J. Cole, T. H. Sanders, and P. D. Blankenship. 1985. Ecology of the Aspergillns flavns group and aflatoxin formation in maize and groundnut, in “Trichothecenes and other mycotoxins” (Lacey, J. Ed). pp. 79-95. John erey & Sons, New York. Hohn, T. M., S. P. McCormick, and A. E. Desjardins. 1993. Evidence for a gene cluster involving trichothecene-pathway biosynthetic genes in F nsarin»: sporotrichioides. Curr. Genet. 24: 291-295. Holzapfel, C. W. 1980. The biosynthesis of cyclopiazonic acid and related tetramic acids, in “The biosynthesis of mycotoxins: a study in secondary metabolism” (Steyn, P. S. Ed). pp. 327-355. Academic Press, New York. Horng, J. S., P.-K. Chang, J. J. Pestka, and J. E. Linz. 1990. Development of a homologous transformation system for Aspergillns parasiticns with the gene encoding nitrate reductase. Mol. Gen. Genet. 224: 294-296. Horsman, M. R., A. A. Khalil, D. J. Chaplin, and J. Overgaard. 1995. The ability of nicotinamide to inhibit the growth of a C3H mouse mammary carcinoma. Acta Oncol. 34: 443-446. Hoza, J. 1966. Secondary reactions of some new antibiotics. Fumagillin. Lek. Veda. Zahr. 8: 156-157. Hsieh, D. P., and R. L Mateles. 1970. The relative contribution of acetate and glucose to aflatoxin biosynthesis. Biocherrr. BiOphys. Acta 208: 482-486. Hsieh, D. P., and R. L Mateles. 1971. Preparation of labeled aflatoxins with high specific activities. Appl. Microbiol. 22:79-83. Hsieh, D. P., C. C. Wan, and J. A. Billington. 1989. Conversion of versiconal acetate to versiconal and versicolorin C in extracts from Aspergillns parasiticns. Mycopatholo. 107: 121-126. Hsieh, D. P., R. Singh, R. C. Yao, and J. W. Bennett. 1978. Anthraquinones in the biosynthesis of sterigmatocystin by Aspergillns versicolor. Appl. Environ. Microbiol. 35: 980-982. Jeenah, M. S., and M. F. Dutton. 1983. The conversion of sterigmatocystin to O- methylsterigmatocystin and aflatoxin Br by a cell-free preparation. Biochem. Biophys. Res. Commun. 116: 1114-1118. Jefferson, R. A., S. M. Burgess, and D. Hirsh. 1986. B-Glucuronidase fiom Escherichia coli as a gene-firsion marker. Proc. Natl. Acad. Sci. 83: 8447-8451. 210 Jelinek, C. F, A. E. Pohland, and G. E. Wood. 1989. Worldwide occurrence of mycotoxin in foods and feeds-an update. J. Assoc. Anal. Chem. 72: 223-230. Kaden, D. A., K. M. Call, P. M. Leong, E. A. Komives, and W. G. Thilly. 1987. Killing and mutation of human lymphoblast cells by aflatoxin B1: evidence for an inducible repair response. Cancer Res. 47: 1993-2001. Kale, S. P., D. Bhatnagar, and J. W. Bennett. 1994. Isolation and charaterization of morphological variants of Amergillsns parasiticns deficient in secondary metabolite production. Mycol. Res. 98: 645-652. Keller, N. P., and T. H. Adams. 1995. Analysis of a mycotoxin gene cluster in Aspergillns nianans. SAAS Bull. Biochem. BiotechnoL 8:14-21. Keller, N. P., H. C. Dischinger Jr., D. Bhatnagar, T. E. Cleveland, and A. H. J. Ullah. 1993. Purification of a 40-kilodalton methyltransferase active in the aflatoxin biosynthetic pathway. Appl. Environ. Microbiol. 59: 479-484. Keller, N. P., N. J. Kantz, and T. H. Adams. 1994. Aspergillns nidulans verA is required for production of the mycotoxin sterigmatocystin. Appl. Environ. Microbiol. 60: 1444-1450. Keller, N. P., S. Segner, D. Bhatnagar, and T. H. Adams. 1995. stcS, a putative P-450 monooxygenase, is required for the conversion of versicolorin A to sterigmatocystin in Amergillns nidulans. Appl. Environ. Microbiol. 10: 3628-3632. Khan, S. N. and T. A. Venkitasubramanian. 1986. Regulation of aflatoxin biosynthesis: effect of adenine nucleotides, cyclic AMP and N-6, O-2'-dibutyryl cych AMP on the incorporation of (1-"C)-acetate into aflatoxins by Aspergillns parasiticns NRRL-3240. J. Environ. Sci. Health. B. 211: 67-85. Kimura, N., and T. Tsuge. 1993. Gene cluster involved in melanin biosynthesis of the filamentous fungus Alternaria alternata. J. Bacteriol. 175: 4427-443 5. King, R. C., and W. D. Stanst'reld. 1985. “A dictioany of gnetics”. pp. 76. Oxford Univrsity Press, New York. ' Kirby, G. W., and D. J. Robins. 1980. The biosynthesis of gliotoxin and related epipolythiodioxopiperazines in T richoderma viride, in “The biosynthesis of mycotoxins: a study in secondary metabolism” (Steyn, P. S. Ed). pp. 301-326. Academic Press, New York. Kirby, G. W., G. L. Patrick, and D. J. Robins. 1978. Cyclo-(Lphenylalanyl-L-seryl) as an intermediate in the biosynthesis of gliotoxin. J. Cherrr. Soc. Perkin Trans. L 1: 1336- 1338. 211 Klich, M. A., and J. L Pitt. 1988. Differentiation of Aspergillns flavns from A. parsiticns and other closely related species. Trans. Brit. Mycol. Soc. 91: 99-108. Kodama, M., F. Inoue, and M. Akao. 1990. Enzymatic and non-enzymatic formation of fiee radicals fi'om aflatoxin Br. Free Radic. Res. Commun. 10: 137-142. Kreuzaler, F., H. Ragg, W. Heller, R. Tesch, L Witt, D. Hammer, and K. Hahlbrock. 1979. Flavanone synthase from Petroselinnm hortense. Molecular weight, subunit composition, size of messenger RNA, and absence of pantetheinyl residue. Eur. J. Biochem. 99: 89-96. Krishnamachari, K. A., R. V. Bhat, V. Nagarajan, and T. B. Tilak. 1975. Hepatitis due to aflatoxicosis. An outbreak in Western India. Lancet 1: 1061-1063. Krogh, P., B. Hald, and E. J. Pedersen. 1973. Occurrence of ochratoxin A and citrinin in cereals associated with mycotoxic porcine nephropathy. Acta Pathol. Microbiol. Scand. B. Microbiol. Irnmunol. 81: 689-695. Kuo, H. L., and W. P. Yun. 1988. Biosynthesis of territrems by Aspergillns ten-ens. Appl. Environ. Microbiol. 54: 585-5 87. Kurtzman, C. P., B. W. Horn, and C. W. Hesseltine. 1987. A new aflatoxin-producing species related to Aspergillnsflaws and Aspergillns tamarii. Antonie Van Leeuwenhoek 53: 147-158. Laakso, J. A., M. R. TePaske, P. F. Dowd, J. B. Gloer, and D. T. Wicklow. 1992. Sulpinine, secopenitrem B and alfatrem B antiinsectan metabolites. U. S. Dep. Agric. Pat. 5,130,236: 1. Labuza, T. P. 1983. Regulation of mycotoxins in food. Ames: International Association of Milk, Food, and Enviromnental Sanitarians. J. Food Prot. 46: 260-265. Lamb, M. J., and L. J. Lilly. 1971. Induction of recessive lethals in Drosophila melanogaster by aflatoxin Br. Mutat. Res. 11: 430-433. Lauren, D. J., A. Wishkovsky, J. M. Groff, R. P. Hedrick, and D. E. Hinton. 1989. Toxicity and pharmacokinetics of the antibiotic firmagillin in yearling rainbow trout (Salmo gairdnerr). Toxicol. Appl. Pharmacol. 98: 444-453. Iebron, C.J., R. A. Molins, H. W. Walker, A. A. Kraft, and H. M. Stahr. 1989. Inhibition of grth and aflatoxin production of Aspergilli in medium containing phosphates. J. Food Prot. 52: 4-6. 212 Lee, B. N., and T. H. Adams. 1994a. The Aspergillns nidulansflnG gene is required for production of an extracellular developmental signal and is related to prokaryotic glutamine synthetase L Gene. Dev. 8: 641-651. Lee, B. N., and T. H. Adams. 1994b. Overexpression of flbA, an early regulator of Aspergillns nidulans asexual sporulation, leads to activation of brlA and premature initiation of development. Mol. Microbiol. 14: 323-334. Lee, B. N., and T. H. Adams. 1996. FlnG and flbA firnction interdependemly to imtr' 'ate conidiophore development in Aspergillns nidnlrms through brlA beta activation. EMBO J. 15: 299-309. Lee, L. S., J. W. Bennett, A. F. Cucullu, and J. B. Stanley. 1975. Two distinct O- methyltransferases in aflatoxin biosynthesis. J. Agric. Food Chem. 23: 1132-1134. Lee, L. S., J. W. Bennett, L. A. Goldblatt, and R. E. Lundin. 1971. Norsolorinic acid fiom a mutant strain of Aspergillns parasiticns. J. Amer. Oil Chem. Soc. 48: 93-94. Lee, S. S., F. C. Peng, C. M. Chiou, and K. H. Ling. 1992. NMR assignments of territrems A B, and C and the structure of MB2, the major metabolite of territrem B by rat liver nricrosomal fraction. J. Nat. Prod. 55: 251-255. Lee, Y. J., and W. M. Hagler Jr. 1991. Aflatoxin and cyclopiazonic acid production by Aspergillnsflavus isolated from contaminated maize. J. Food Sci. 56: 871-872. Lin, B. K., and J. A. Anderson. 1992. Purification and properties of versiconal cyclase from Aspergillns parasiticns. Arch. Biocherrr. Biophys. 293: 67-70. Lin, M. T., D. P. Hsieh, R. C. Yao, and J. A. Donkersloot. 1973. Conversion of averufin into aflatoxins by Aspergillns parasiticns. Biochem. 12: 5167-5171. Ling, K. H. 1976. Study on mycotoxins contaminated in food in Taiwan. (2) Tremor inducing compounds fi'om Aspergillns terrens. Proc. Natl. Aci. Council, Republic of China 9: 121-129. Ling, K. H. 1994. Territrems, tremorgenic mycotoxins isolated fiom Aspergillns terrens J. Toxicol. Toxin Rev. 13: 243-252. Ling, K. H., H. H. Lion, C. M. Yang, and C. K. Yang. 1984. Isolation, chemical structure, acute toxicity, and some physicocherrrical properties of territrem C fi'om Awergillns terrens. Appl. Environ. Microbiol. 7: 98-100. Lowery, C., T. C. Orton, and R. C. Garner. 1983. Metabolic activation and bacterial mutagenicity of aflatoxin Br in two difl‘erent test systems. Toxicol. 29: 131-141. 213 Liick, H. 1965. Catalase, in “Methods of enzymatic analysis” (Bergrneyer, IL U. Ed). pp. 885-894. Academic Press, New York. Luckner, M. 1990. Secondary products derived from L-phenylalanine and L-tyrosine (phenylpropanoinds), in “Secondary metabolism in microorganisms, plants, and animals” (Luckner, M. Ed). pp. 361-417. 3rd Ed. Springer-Verlag. New York Luk, K. C., B. Kobbe, and J. M. Townsend. 1977. Production of cyclopiazonic acid by Aspergillnsflavns Link. Appl. Environ. Microbiol. 33: 211-212. MacCabe, A. P., H. van Liempt, H. Palissa, S. E. Unkles, M. B. R. Riach, E. Pfeifer, H. von Dohren, and J. R. Kinghorn. 1991. 5—(L-a-Aminoadipyl)—L-cysteinyl-D-valine synthetase from Aspergillns nidulans. Molecular characterization of the ach gene encoding the first enzyme of the penicillin biosynthetic pathway. J. Bio. Chem. 266: 12646-12654. MacCabe, A. P., M. B. R. Riach, S. E. Unkles, and J. R. Kinghorn. 1990. The Aspergillns nidulans npeA locus consists of three contiguous genes required for penicillin biosynthesis. EMBO J. 9: 279-287. Mace, K., F. J. Gonzalez, L R. McConnell, R. C. Garner, 0. Avanti, C. C. Harris, and A. M. Pfeifer. 1994. Activation of promutagerrs in a human bronchial epithelial cell line stably expressing human cytochrome P-450 1A2. MoL Carcinog. 11: 65-73. Macgregor, J. T., D. H. Gould, A. D. Mitchell, and G. P. Sterling. 1979. Mutagenicity tests of diflubenzuron in the nricronucleus test in mice, the L5178Y mouse lymphoma forward mutation assay, and the Ames Salmonella reverse mutation test. Mutat. Res. 66: 45-53. Maebayashi, Y., K. Miyaki, and M. Yamazaki. 1972. Application of ‘3C-NMR to the biosynthetic investigations. I. Biosynthesis of ochratoxin A. Chem. Pharm. Bull. (Tokyo) 20: 2172-2175. Mahanti, N., D. Bhatnagar, J. W. Cary, J. Joubran, and J. E. Linz 1996. Structure and function of fas-l A, a gene encoding a putative fatty acid synthetase directly involved in aflatoxin biosynthesis in Aspergillns parasiticns. Appl. Environ. Microbiol. 62: 191- 195. Manitto, P. 1981. The isoprenoids, in “Biosynthesis of natural products” (Manitto, P. Ed). pp. 213-313. John Wiley & Sons, New York. Martin, J. F., and S. Gutierrez. 1995. Genes for B-lactam antibiotic biosynthesis. Antonie van leeuwenhoek 67: 181-200. 214 Mashaly, R. L, S. L. Habib, S. A. el Deeb, M. H. Salem, and M. M. Safwat. 1988. Isolation, purification and characterization of enzyme(s) responsible for conversion of sterigmatocystin to aflatoxin Br. Z. Lebensm. Unters. Forsch. 186: 118-124. Matsushima, K., Y. Ando, T. Hamasaki, and K. Yabe. 1994. Purification and characterization of two versiconal hemiacetal acetate reductases involved in aflatoxin biosynthesis. Appl. Environ. Microbiol. 60: 2561-2567. May, G. S., M. L. Tsang, H. Smith, S. Fidel, and N. R. Morris. 1987. Aspergillns nidulans B-tubulin genes are unusually divergent. Gene. 55: 231-243. May, G. S., N. R. Morris. 1988. Developmental regulation of a conidiation specific beta- tubulin inAspergilIns nidulans. Dev. Biol. 128: 406-414. Mayorga, M. E., and W. E. Timberlake. 1992. The developmentally regulated Aspergillns nidulans wA gene encodes a polypeptide homologous to polyketide and fatty acid synthases. Mol. Gen. Genet. 235: 205-212. McCormick, S. P., D. Bhatnagar, and L. S. Lee. 1987. Averufanin is an aflatoxin Br precursor between averantin and averufin in the biosynthetic pathway. Appl. Environ. Microbiol. 53: 14-16. McGuire, S. M., S. W. Brobst, T. L. Graybill, K. Pal, and C. A. Townsend. 1989. Partitioning of tetrahydro- and dihydro-bisfuran formation in atlatoxin biosynthesis defined by cell-fiee and direct incorporation experiments. J. Am. Chem. Soc. 111: 8308-8309. McLean , M., P. Berjak, M. P. Watt, and M. F. Dutton. 1992. The efl‘ects of aflatoxin B1 on immature germinating maize (Zea mays) embroyos. Mycopathol. 119: 181-190. Miinkc, E., and W. Schfifer. 1993. Transient and stable gene expression in the fungal maize pathogen Cochliobolns heterostrophus after thransformation with the B-glucuronidase (GUS) gene. Mol. Gene. Genet. 241: 73-80. Montenegro, E., J. L. Barredo, S. Gutierree,'B. Dies, E. Alvarez, and J. F. Martin. 1990. Cloning, characterization of the acyl-CoA: 6-amino penicillanic acid acyltransferase gene of Aspergillns nidulans and linkage to the isopenicillin N synthase gene. MoL Gen. Genet. 221: 322-330. Mori, H., J. Kitamura, S. Sugie, K. Kawai, and T. Hamasaki. 1985. Genotoxicity of firngal metabolites related to aflatoxin Br biosynthesis. Mutat. Res. 143: 121-125. Moss, M. O. 1977. Aspergillns mycotoxins, in “Genetics and physiology of Aspergillns” (Simth, J. E., and J. A. Pateman Eds). pp. 499-524. Academic Press, London. 215 Moss, M. O. 1994. Biosynthesis of Aspergillns toxins-non-aflatoxins. in “The genus Aspergillns” (Powell, K. A., A. Renwick, and J. F. Peberdy Eds). pp. 29-50. Plenum Press, New York. Muller,W.H.,T.P.van derKrift,A.J.J.Krouwer,H.A.B.Wosten,L.H.M.Voort, E. B. van der Smaal, and A. J. Verkleij. 1991. Localization of the pathway of the penicillin biosynthesis in Penicilliwn chrysogennm. MBO J. Eur. MoL Biol. Organ. 10: 489-495. Ngindu, A., B. K. Johnson, P. R. Kenya, J. A. Ngira, D. M. Ocheng, H. Nandwa, T. N. Omondi, A. J. Jansen, W. Ngare, J. N. Kaviti, D. Gatei, and T. A. Siongok. 1982. Outbreak of acute hepatitis caused by aflatoxin poisoning in Kenya. Lancet. 1: 1346- 1348. Niehaus, W. G. Jr, and R. P. Dilts Jr. 1984. Purification and characterization of glucose-6- phosphate dehydrogenase fiom Anaergillns parasiticns. Arch. Biochem. Biophys. 228: 113- 119. Nishimura, Y., R. Murata, and M. Hiraoka. 1996. Combined efi‘ects of an angiogenesis inhibitor (TNP-470) and hyperthermia. Br. J. Cancer. 73: 270-427. Nose, H., F. Imazeki, M. Ohto, and M. Omata. 1993. p53 gene mutations and 17p allelic deletions in hepatocellular carcinoma fi'om Japan. Caner 72: 355-360. Oakley, B. R., J. E. Rinehart, B. L. Mitchell, C. E. Oakley, C. Carmona, G. L. Gray, and G. S. May. 1987. Cloning, mapping and molecular analysis of the pyrG (orotidine-5'- phosphate decarboxylase) gene of Aspergillns nidulans. Gene 61: 385-3 99. Olsson, S., and D. H. Jennings. 1991. A glass fiber filter technique for studying nutrient uptake by fungi: the technique used on colonies grown on nutrient gradients of carbon and phosphorus. Exp. Mycol. 15: 292-301. Ottinger, M. A., and J. A. Doerr. 1980. The early influence of afiatoxin upon sexual maturationinthe male Japanese quail. Poult. Sci. 59: 1750-1754. Pachler, K. G., P. S. Steyn, R. Vleggaar, and P. L. Wessels. 1976. Carbon-l3 nuclear magnetic resonance assignments and biosynthesis of aflatoxin Br and sterigmatocystin. J. Chem. Soc. Perkin. L 11: 1182-1189. Papa, K. E. 1986. Heterokaryon incompatibility in Aspergillns flavns. Mycol. 78: 98- 101. Park, D. L., and B. Liang. 1993. Perspectives on aflatoxin control for human food and animal feed. Trends Food Sci. Technol. 4: 334-342. 216 Payne, G. A., G. J. Nystrom, D. Bhatnagar, T. E. Cleveland, and C. P. Woloshuk. 1993. Clonig of the afl-2 gene involved in aflatoxin biosynthesis fiom Aspergillns falvns. Appl. Environ. Mirobiol. 59:156-162. Peng, F. C., K. H. Ling, Y. Wang, and G. H. Lee. 1985. Isolation, chemical structure, acute toxicity, and some physicochemical properties of territrem B from Aspergillns terrens. Appl. Environ. Microbiol. 49: 721-723. Pestka, J. J, P. K. Gaur, and F. S. Chu. 1980. Quantitation of aflatoxin Br and aflatoxin Br antibody by an enzyme-linked irnmunosorbent microassay. Appl. Environ. Microbiol. 40: 1027-1031. Puisieux, A., S. Lim, J. Groopman, and M. Ozturk. 1991. Selective targeting of p53 gene mutational hotspots in human cancers by etiologically defined carcinogens. Cancer Res. 51: 6185-6189. Raina, V., H. L. Gurtoo, M. Berrigan, and B. Paigen. 1980. Genetic dependence of hepatic microsome—mediated depression of aflatoxin Br activation to mutagens in Ames Salmonella whimnrinm TA-98 system. Biocherrr. Biophys. Res. Commun. 94: 737-743. Rainey, P. B., C. L. Brodey, and K. Johnstone. 1993. Identification of a gene cluster encoding three high-molecular-weight proteins, which is required for synthesis of tolasin by the mushroom pathogen Psendomonas tolaasii. Mol. Microbiol. 8: 643 -652. Ramon, D., L. Carramolino, C. Patina, F. Sanchez, and M. A. Penalva. 1987. Cloning and characterization of the isopenicillin N synthetase gene mediating the formation of the beta-lactam ring in Aspergillns nidulans. Gene 57 : 171-181. Reddy, J. K., D. J. Svoboda, and M. S. Rao. 1976. Induction of liver tumors by aflatoxin Br in the tree shrew (Tnpaia glis), a nonhuman primate. Cancer Res. 36: 151- 160. Reddy, T. V., L. Viswanathan, and T. A. Venkitasubramanian. 1971. High aflatoxin production on a chemically defined medium. [Amergilhzs parasiticns]. AppL Microbiol. 22: 393-3 96. Refolo, L. M., C. B. Bennett, and M. Z. Humayun. 1987. Mechanisms of frameshifi mutagenesis by aflatoxin Br-2,3-dichloride. J. Mol. BioL 193: 609-636. Revill, W. P., M. J. Bibb, and D. A. Hopwood. 1995. Purification of a malonyltransferase fiom Streptomyces coelicolor A3(2) and analysis of its genetic determinant. J. Bacteriol. 177: 3946-3952. Richard, J. L., A. C. Pier, S. J. Cysewski, and C. K. Graham. 1973. Efi‘ect of aflatoxin and aspergillosis on turkey poults. Avian Dis. 17: 111-121. 217 Richard, J. L., and R. T. Gallagher. 1979. Multiple toxin production by an isolate of Aspergillnsflam. Mycopatholo. 67: 161-163. Roach, P. L., C. J. Schofield, J. E. Baldwin, L J. Clifton, and J. Hajdu. 1995. Crystallization and preliminary X-ray diffraction studies on recombinant isopenicillin N synthase fi'om Aspergillns nidulans. Protein Sci. 4: 1007- 1009. Robens, J. F., and J. L. Richard. 1992. Aflatoxins in animal and human health. Rev. Environ. Contam. Toxicol. 127: 69-94. Rodig, O. R., L. C. Ellis, and L T. Glover. 1966. The biosynthesis of citrinin in Penicillin»: citrinnm. I. Production and degradation of citrinin. Biochem. 5: 2451-2458. Rose, M. E., and R. A. W. Johnstone. 1982. Structure elucidation, in “Mass sepctrometry for chemists and biochemists”. pp. 185-246. Cambridge University Press, Cambridge, Great Britain. Rothweiler, W., and C. Tamm. 1966. Isolation and structure of phomin. Experientia 22: 750-752. Russell, T. E. 1979. Field conditions which influence aflatoxin Aspergillns flavns production in cottonseed. Prepr. Pap. Oilseed Process Clin. New Orleans, Oilseed Processing Clinic 28: 44-45. Samson, R. A., and J. C. Frisvad. 1991. Current taxonomic concepts in Penicillin»: and Aspergillns. Dev. Food Sci. 26: 405-439. Samson, S. M., R. Belagaje, D. T. Blankenship, J. L. Chapman, D. Perry, P. L. Skatrud, R. M. Van Frank, E. P. Abraham, J. E. Baldwin, and S. W. Queener. 1985. Isolation, sequence determination and expression in Escherichia coli of the isopenicillin N synthetase gene fi'om Cephalosporinm acremoninm. Nature 318: 191-194. Schoppner, A., and H. Kindl. 1984. Purification and properties of a stilbene synthase fiom induced cell suspension cultures of peanut. J. Biol. Chem. 259: 6806-6811. Scott, D. B. 1965. Toxigenic fungi isolated fi'om cereal and legume products. Mycopathol. Mycol. Appl. 25: 213-222. Scary, J. W., N. D. Davis, and U. L. Diener. 1969. Biosynthesis of ochratoxin A. Appl. Microbiol. 18: 622-627. Shantha, T., E. R. Rati, and T. N. B. Shankar. 1990. Behaviour of Aspergillns flaws in presence of Aspergillns niger during biosynthesis of aflatoxin B1. Antonie Van Leeuwenhoek 58: 121-127. 218 Shearer, J. F., L. E. Sweets, N. K. Baker, and L. H. Tiffany. 1992. A study of Aspergillnsflavns/parasiticns in Iowa crop fields: 1988-1990. Plant. Dis. 76: 19-22. Shih, C. N., and E. H. Marth. 1974. Aflatoxin formation, lipid synthesis, and glucose metabolism by Aspergillns parasiticns during incubation with and without agitation. Biochinr. Biophys. Acta 338: 286-296. Sieber, S. M., P. Correa, D. W. Dalgard, and R. H. Adamson. 1979. Induction of osteogenic sarcomas and tumors of the hepatobiliary system in nonhuman primates with aflatoxin Br. Cancer Res. 39: 4545-4554. Silva, J. C., R. E. Minto, C. E. Barry, K. A. Holland, and C. A. Townsend. 1996. Isolation and characterization of the versicolorin B synthase gene fiom Aspergillns parasiticns. Expansion of the aflatoxin Br biosynthetic gene cluster. J. Biol. Chem. 271: 13600-13608. Simpson, T. J. 1987. The biosynthesis of polyketides. Nat. Prod. Rep. 4: 339-376. Simpson, T. J., D. J. Stenzel, A. J. Bartlett, E. O'Brien, and J. S. E. Holker. 1982. Studies on fungal metabolites. 3. l3C-carbon isotopes NMR (nuclear magnetic resonance) spectral and structural studies on austin and new related meroterpenoids from Aspergillns nstns, Aspergillns variecolor, and Penicillin»: diversn»: Fungi. J. Chem. Soc. Perkin. Trans. 1. 11: 2687-2692. Singer, T. P., P. Bernath, and C. J. Lusty. 1965. Succinate, in “Methods of enzymatic analysis” (Bergrneyer, H U. Ed). pp. 340-345. Academic Press, New York. Singh, R., and D. P. H. Hsieh. 1976. Enzymatic conversion of sterigmatocystin into aflatoxin B: by cell-free extracts of Aspergillns parasiticns. Appl. Environ. Microbiol. 31: 743 -745. Sinnhuber, R. 0., J. H. Wales, J. L. Ayres, R. H. Engebrecht, and D. L. Amend. 1968. Dietary factors and hepatoma in rainbow trout (Salmo gairdneri). L Aflatoxins in vegetable protein feedstufi‘s. J. Natl. Cancer. Inst. 41: 711-718. Skory, C. D., J. S. Horng, J. J. Pestka, and J. E. Linz. 1990. Transformation of Aspergillns pw'asiticns with a homologous gene (pyrG) involved in pyrimidine biosynthesis. Appl. Environl. Microbiol. 56: 3315-3320. Skory, C. D., P.-K. Chang, and J. E. Linz. 1993. Regulated expression of the nor-l and ver-l genes associated with aflatoxin biosynthesis. Appl. Environ. Microbiol. 59: 1642- 1646. Skory, C. D., P.-K. Chang, J. Cary, and J. E. Linz. 1992. Isolation and characterization of a gene from Aspergillns parasiticns associated with the conversion of 219 versicolorin A to sterigmatocystin in aflatoxin biosynthesis. Appl. Environ. Microbiol. 58: 3527-3 53 7. Soman, N. R., and G. N. Wogan. 1993. Activation of the c-ki-ras oncogene in aflatoxin- Br-induced hepatocellular carcinoma and adenoma in the rat: detection by denaturing gradient gel electrophoresis. Pro. Natl. Acad. Sci. 90: 2045-2049. Springer, J. P., J. W. Dorner, R. J. Cole, and R. H. Cox. 1979. Terretonin, a toxic compound from Aspergillns terrens. J. Org. Chem. 44: 4852-4854. Stark, A. 1980. Mutagenicity and carcinogenicity of mycotoxins: DNA binding as a possible mode of action. Annu. Rev. Microbiol. 34: 23 5-262. Steenkamp, D. J., J. C. Schabort, and N. P. Ferreira. 1973. B-cyclopiazonate oxidocyclase from Penicillin»: cyclopinm, III, preliminary studies on the mehanism of action. Biochim. Biophys. Acta 309: 440-456. Steyn, P. S., and R. Vleggaar. 1975. Dihydrofirro[3’,2’,4,5] firro [3,2-b]xanthenones: the structures of austocystins G, H, and I. J. S. Afi'. Chem. Inst. 28: 375-377. Steyn, P. S., and R. Vleggaar. 1985. Tremorgenic mycotoxins. Fortschr. Chem. Org. Naturst. 48: 1-80. Steyn, P. S., C. W. Holzapfel, and N. P. Ferreira. 1970. The biosynthesis of the ochratoxins, metabolites of Aspergillns ochracens. Phytochem. 9: 1977-1983. Steyn, P. S., R. Vleggaar, and P. L. Wessels. 1980. The biosynthesis of aflatoxin and its congeners, in “The biosynthesis of mycotoxins: a study in secondary metabolism”(Steyn, P. S. Ed). pp. 105-155. Academic Press, New York. Steyn, P. S., R. Vleggaar, N. P. Ferreira, G. W. Kirby, and M. J. Varley. 1975a. Steric course of proton removal during the cyclistation of beta-cyclopiazonic acid in Penicillin»: cyclopinm. ChemCommun. 12: 465-466. Steyn, P. S., R. Vleggaar, P. L. Wessels, and B. de Scott. 1975b. Biosynthesis of aflatoxin Br from [2-‘3c1 and [1,2-‘3C]-acetate [Aspergillns flavns, Aspergillns parasiticns, flingr’]. J. Chem. Soc. Chem. Comm. 6:193-195. Suhadolnik, R. J., and R. G. Chenoweth. 1958. Biosynthesis of gliotoxin. L Incorporation of phenylalanine-l-and-Z-“C. J. Am. Chem. Soc. 80: 4391-4392. Summers, R. G., A. Ali, B. Shen, W. A. Wessel, and C. R. Hutchinson. 1995. Malonyl-coenzyme A: acyl carrier protein acyltransferase of Sireptonwces glancescens: a possible link between fatty acid and polyketide biosynthesis. Biochem. 34: 9389-9402. 220 Tada, S., K. Gomi, K. Kitamoto, K. Takahashi, G. Tamura, and S. Hara. 1991. Construction of a fusion gene comprising the Taka-amylase A promoter and the Escherichia coli B-glucuronidas gene and analysis of its expressin in Aspergllns oryzae. Mol. Gen. Genet. 229: 301-306. Tamm, C. H. 1980. The biosynthesis of the cytochalasans, in “The biosynthesis of mycotoxins: a study in secondary metabolism” (Seytn, P.S. Ed). pp. 269-299. Academic Press, New York. Temperli, E., U. P. Roos, and H. R. Hohl. 1990. Actin and tubulin cytoskeletons in germlings of the oomycete firngus Phytophthora infestans. Eur. J. Cell Biol. 53: 75-88. TePaske, M. R., J. B. Gloer, D. T. Wicklow, and P. F. Dowd. 1992. Aflavarin and beta-aflatrem: new anti-insectan metabolites from the sclerotia of Aspergillns flaws. J. Nat. Prod. 55: 1080-1086. Thomas, R. 1965. Biosynthetic pathways involving ring cleavage, in “Biogenesis of antibiotic substances” (Vanek, Z., and Z. Hostalek Eds). pp. 115-168. Academic Press, New York. Tilak, T. B. 1975. Induction of cholangiocarcinoma following treatment of a Rhesus monkey with aflatoxin. Food Cosmet Toxicol. 13: 247-249. Timberlake, W. E., and E. C. Barnard. 1981. Organization of a gene cluster expressed specifically in the asexual spores of Aspergillns nidnlans Fungi. Cell 26: 29-3 7. Townsend, C. A., K. A. Plancan, K. Pal, and S. W. Brobst. 1988. Hydroxyversicolorone: isolation and characterisation of a potential intemediate in aflatoxin biosynthesis. J. Org. Chem. 53: 2472-2477. Townsend, C. A., S. B. Christensen, and K. Trantwein. 1984. Hexanoate as a starter unit in polyketide biosynthesis. J. Amer. Chem. Soc. 106: 3868-3 869. Townsend, C. A., S. B. Christensen. and S. G. Davis. 1982. Bisfirran formation in aflatoxin biosynthesis: the role of versiconal acetate. J. Am. Chem. Soc. 104: 6154-6155. Trail, F., N. Mahanti, M. Rarick, R. Mehigh, S. H. Liang, R. Zhou, and J. E. Linz. 1995. Physical and transcriptional map of an aflatoxin gene cluster in Aspergillns parasiticns and firnctional disruption of a gene involved early in the aflatoxin pathway. Appl. Environ. Microbiol. 61: 2665-2673. Trail, F., P.-K. Chang, J. Cary and J. E. Linz. 1994. Structural and functional analysis of the nor-1 gene involved in the biosynthesis of aflatoxins by Aspergillns pwasiticns. Appl. Environ. Microbiol. 60: 4078-4085. 221 Trucksess, M. W., P. B. Mislivec, K. Young, V. R. Bruce, and S. W. Page. 1987. Cyclopiazonic acid production by cultures of Aspergillns and Penicillin»: species isolated fiom dried beans, corn meal, macaroni, and pecans. J. Assoc. Ofi‘. Anal. Chem. 70: 123- 126. Tsuneda, A. (Ed). 1983. Fungal morphology and ecology. pp. 196. Tokyo Press Co., Ltd. Tokyo, Japan. Turner, W. B., and D. C. Aldridge (Eds). 1983. Fungal metabolites II. pp. 1-631. Academic Press, London. Ueda, K., K. Miyake, S. Horinouchi, and T. Beppu. 1993. A gene cluster involved in aerial mycelium formation in Streptomyces grisens encodes proteins similar to the response regulators of two-component regulatory systems and membrane translocators. J. Bacteriol. 175: 2006-2016. Umcda, M., T. Tsutsui, and M. Saito. 1977. Mutagenicity and inducibility of DNA single-strand breaks and chromosome aberrations by various mycotoxins. Gann 68: 619- 625. Van den Brock, P., T. Goosen, B. Wennckes, and H. van den Brock. 1995. Isolation and characterization of the glucose-6-phosplmte dehydrogenase encoding gene (gsdA) fiom AWIIns niger. MoL Gen. Genet. 247: 229.239. Van Liempt, H., H. von Doehrcn, and H. Kleinkauf. 1989. 5—(L-a-aminoadipyl)-L- cysteinyl-D-valine synthetase fi'om Aspergillns nidnlans. The first enzyme in penicillin biosynthesis is a multifunctional peptide synthetase. J. Biol. Chem. 264: 3680-3684. Vederas, J. C. 1987. The use of stable isotopes in biosynthetic studies. Nat. Prod. Rep. 4: 277-337. Verrett, M. J., J. Winbush, E. F. Reynaldo, and W. F. Scott. 1973. Collaborative study of the chicken embryo bioassay for aflatoxin Br. Ass. Ofic. Anal. Chem. J. 56: 901-904. Vida, T. A., G. Huycr, and S. D. Emr. 1993. Yeast vacuolar proenzymes are sorted in the late Golgi corrrplex and transported to the vacuole via a prevacuolar endosome-like comparMent. J. Cell. Biol. 121: 1245-1256. Webster, J., and L. Lomas. 1964. Does Yhichodenna viride produce gliotoxin and viridin? 'Ihans. Brit. Mycol. Soc. 47: 535-540. ’ Wei, R. D., F. M. Strong, and E. B. Smallcy. 1971. Incorporation of chlorine-36 into ochratoxin A. Appl. Microbiol. 22: 276-277. 222 Weiss U., and J. M. Edwards. (Eds). 1980. The biosynthesis of aromatic compounds. John Wiley & Sons, New York. Whiteman, P. A., E. P. Abraham, J. E. Baldwin, M. D. Fleming, C. J. Schofield, J. D. Sutherland, and A. C. Willis. 1990. Acyl coenzyme A: 6-aminopenicillanic acid acyltransferase fi'om Penicillin»: chrysogenn»: and Aspergillns nidnlans. FEBS Lett. 262: 342-344. Wicklow, D. T. 1990. Environmental factors and other stresses important in the production of aflatoxin in corn. USDA, Agricultural Research Service. 83: 53-61. Wicklow, D. T., and O. 1.. Shotwell. 1983. Intrafilngal distnbution of aflatoxins among conidia and sclerotia of Amergillns flaws and Aspergillns parasiticns Can. J. Microbiol. 29: 1-5. Wicklow, D. T., and R. J. Cole. 1982. Tremorgenic indole metabolites and aflatoxins in sclerotia of Aspergillns flavns, an evolutionary perspective. Can. J. Bot. 60: 525-528. Wicnienski, N. A. 1979. Preliminary studies of the biosynthesis of austin. Govt. Reports Announcements & Index (GRA&I), Issue 23. Wilkinson, S. and J. F. Spilsbury. 1965. Gliotoxin fi'om Aspergillns chevalieri (Mangin) Thom et Church. Nature 206: 619. Willetts, H. J., and S. Bullock. 1992. Developmental biology of sclerotia. Mycol. Res. Cambridge 96: 801-816. Wilson, B. J., and C. H. Wilson. 1964. Toxin from Aspergillns flavns: production on food materials of a substance causing tremors in mice. Science 144: 177-178. Wiseman, D. W., and R. L. Buchanan. 1987. Determination of glucose level needed to induce aflatoxin production in Aspergillns parasiticns. Can. J. Microbiol. 33: 828-830. Wogan, G. N., and P. M. Newbemc. 1967. Dose-response charateristics of aflatoxin Br carcinogenesisintherat. CancerRes. 27: 2370-2376. Woloshuk, C. P., E. R. Seip, G. A. Payne, and C. R. Adkins. 1989. Genetic transformation system for the aflatoxin-producing firngus Aspergillns flavns. Appl. Environ. Microbiol. 55: 86-90. Woloshuk, C. P., K. R. Foutz, J. F. Brewer, D. Bhatnagar, T. E. Cleveland, and G. A. Payne. 1994. Molecular characterization of aflR, a regulatory locus for aflatoxin biosynthesis. Appl. Environ. Microbiol. 60: 2408-2414. 223 Wood, M. L., J. R. Smith, and R. C. Garner. 1987. Aflatoxin Br activation to a plasmid mutagen by a chemical model of cytochrome P-450. Mutat Res. 176:11-20. Woodward, R. B., and G. Singh. 1949. The structure of patulin. J. Am. Chem. Soc. 71: 758-759. Yabe, K., and T. Hamasaki. 1993. Stereochemistry during aflatoxin biosynthesis: cyclase reaction in the conversion of versiconal to versicolorin B and reacemization of versiconal hemiacetal acetate. Appl. Environ. Microbiol. 59: 2493-2500. Yabe, K., Y. Ando, and T. Hamasaki. 1991a. Desaturase activity in the branching step between aflatoxins Br and Gr and aflatoxins B: and G2. Agric. Biol. Chem. 55:1907- 191 1. Yabe, K., Y. Ando, J. Hashirnoto, and T. Hamasaki. 1989. Two distinct O- methyltransferases in aflatoxin biosynthesis. AppL Environ. Microbiol. 55: 2172-2177. Yabe, K., Y. Matsuyama, Y. Ando, H. Nakajima, and T. Hamasaki. 1993. Stereochemistry during aflatoxin biosynthesis: conversion of norsolorinic acid to averufin. AppL Environ. Microbiol. 59: 2486-2492. Yabe, K., Y. Nakamura, H. Nakajima, Y. Ando, and T. Hamasaki. 1991b. Enzymatic conversion of norsolorinic acid to averufin in aflatoxin biosynthesis. AppL Environ. Microbiol. 57: 1340-1345. Yamazaki, M. 1980. The biosynthesis of neurotropic mycotoxins, in “The biosynthesis of mycotoxins: a study in seconde metabolism” (Steyn, P. S. Ed). pp. 193-222. Academic Press, New York. Yamazaki, M., E. Okuyama, and Y. Maebayashi. 1979. Isolation of some new tryptoquivaline-related metabolites from Aspergillns fumigatns. Chem. Pharm. BulL Tokyo, 27: 1611-1617. Yamazaki, M., H. Fujimoto, and T. Kawasaki. 1980. Chemistry of tremorogenic metabolites. I. F unritremorgin A fiom Aspergillns firmigains. Chem. Pharm. Bull. (Tokyo) 23: 245-254. Yamazaki, M., S. Suzuki, and K. Miyaki. 1971. Tremorgenic toxins fiom Aspergillns fianigatns Fres. Chem. Pharm. Bull. Tokyo. 19: 1739-1740. Yan, R. Q., J. J. Su, D. R. Huang, Y. C. Gan, C. Yang, and G. H. Huang. 1996. Human hepatitis B virus and hepatocellular carcinoma. 11. Experimental induction of hepatocellular carcinoma in tree shrews exposed to hepatitis B virus and aflatoxin Br. J. Cancer. Res. Clin. Oncol. 122: 289-295. 224 Yao, R. C., and D. P. H. Hsieh. 1974. Step of dichlorvos inhibition in the pathway of aflatoxin biosynthesis. Appl. Microbiol. 28: 52-57. Yoneyama, M., R. P. Sharma, and Y. Y. Elsner. 1987. Efi‘ects of mycotoxins in cultured kidney cells: cytotoxicity of aflatoxin Br in Madin-Darby and primary fetal bovine kidney cells. Ecotoxical. Environ. Safety 13: 174-184. Yu, J. H., J. Wieser, and T. H. Adams. 1996. The Aspergillns flbA RGS domain protein antagonizes G protein signaling to block proliferation and allow development. EMBO J. 15: 5184-5190. Yu, J. H., and T. J. Leonard. 1995. Sterigmatocystin biosynthesis in Aspergillns nidnlans requires a novel type I polyketide synthase. J. Bacteriol. 177: 4792-4800. Yu, J., J. W. Cary, D. Bhantnagar. T. E. Cleveland, N. P. Keller, and F. S. Chu. 1993. Cloning and characterization of a cDNA fi‘om Aspergillns parasiticns encoding an O-mcthyltransferase involved in aflatoxin biosynthesis. Appl. Environ. Microbiol. 59: 3564-3571. Zamir, L. O. 1980. The biosynthesis of patulin and penicillic acid, in “The biosynthesis of mycotoxins: a study in secondary metabolism”( Steyn, P. S. Ed). pp. 223-268. Academic Press, New York.