Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Fungi (Basel) ; 9(1)2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36675850

ABSTRACT

Screening for microorganisms that inhibit aflatoxin production from environments showed that Penicillium citrinum inhibited aflatoxin production by Aspergillus parasiticus. The inhibitory substance in the culture medium of P. citrinum was confirmed to be citrinin (CTN). RT-PCR analyses showed that CTN did not inhibit expressions of aflatoxin biosynthetic genes (aflR, pksL1, and fas-1) of A. parasiticus, whereas feeding experiments using A. parasiticus showed that CTN inhibited the in vivo conversion of dihydrosterigmatocystin to AFB2·AFG2. These results suggest that CTN inhibits a certain post-transcriptional step in aflatoxin biosynthesis. CTN in the culture medium of A. parasiticus was found to be decreased or lost with time, suggesting that a certain metabolite produced by A. parasiticus is the cause of the CTN decrease; we then purified, characterized, and then analyzed the substance. Physico-chemical analyses confirmed that the metabolite causing a decrease in CTN fluorescence was kojic acid (KA) and the resulting product was identified as a novel substance: (1R,3S,4R)-3,4-dihydro-6,8-dihydroxy-1-(3-hydroxy-6-(hydroxymethyl)-4-oxo-4H-pyran-2-yl)-3,4,5-trimethyl-1H-isochromene-7-carboxylic acid, which was named "CTN-KA adduct". Our examination of the metabolites' toxicities revealed that unlike CTN, the CTN-KA adduct did not inhibit aflatoxin production by A. parasiticus. These results indicate that CTN's toxicity was alleviated with KA by converting CTN to the CTN-KA adduct.

2.
J Fungi (Basel) ; 9(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36675872

ABSTRACT

Our previous work showed that citrinin (CTN) produced bay Penicillium citrinum inhibited the production of aflatoxin by Aspergillus parasiticus. We also reported that CTN was non-enzymatically converted to a novel CTN-KA adduct with kojic acid (KA) in aqueous condition. We herein observed that unlike CTN, the CTN-KA adduct does not show antimicrobial activity against Escherichia coli or Bacillus subtilis or any cytotoxic effect on HeLa cells, suggesting that CTN was detoxified by KA by the formation of the CTN-KA adduct. To examine the function of KA production by fungi, we isolated A. parasiticus mutants with impaired KA production. When the mutants were incubated in either liquid or agar medium supplemented with CTN, they were more susceptible to CTN than the wild KA-producing strain. The same results were obtained when we used the A. oryzae KA-producing strain RIB40 and KA-non-producing strains. When KA was added to the CTN-containing agar medium, the inhibition of growth by CTN was remarkably mitigated, suggesting that the production of KA protected the fungal growth from CTN's toxicity. We also observed that CTN enhanced the production of KA by A. parasiticus as well as A. oryzae strains. Reverse transcription-PCR showed that CTN enhanced the expression of KA biosynthetic genes (kojA, kojR, and kojT) of A. parasiticus. However, the enhancement of KA production with CTN was repressed by the addition of α-tocopherol or butylated hydroxy anisole, suggesting that KA production is enhanced by oxidative stress via the formation of reactive oxygen species caused by CTN. In contrast, α-tocopherol did not affect inhibition of AF production as well as fungal growth by CTN, suggesting that the regulation of these inhibitions with CTN might be different from that of KA production. We propose a regulation scheme of CTN for each of KA production, AF production, and fungal growth in A. parasiticus.

3.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887494

ABSTRACT

In the biosynthesis of aflatoxin, verA, ver-1, ordB, and hypA genes of the aflatoxin gene cluster are involved in the pathway from versicolorin A (VA) to demethylsterigmatocystin (DMST). We herein isolated each disruptant of these four genes to determine their functions in more detail. Disruptants of ver-1, ordB, and hypA genes commonly accumulated VA in their mycelia. In contrast, the verA gene disruptant accumulated a novel yellow fluorescent substance (which we named HAMA) in the mycelia as well as culture medium. Feeding HAMA to the other disruptants commonly caused the production of aflatoxins B1 (AFB1) and G1 (AFG1). These results indicate that HAMA pigment is a novel aflatoxin precursor which is involved at a certain step after those of ver-1, ordB, and hypA genes between VA and DMST. HAMA was found to be an unstable substance to easily convert to DMST and sterigmatin. A liquid chromatography-mass spectrometry (LC-MS) analysis showed that the molecular mass of HAMA was 374, and HAMA gave two close major peaks in the LC chromatogram in some LC conditions. We suggest that these peaks correspond to the two conformers of HAMA; one of them would be selectively bound on the substrate binding site of VerA enzyme and then converted to DMST. VerA enzyme may work as a key enzyme in the creation of the xanthone structure of DMST from HAMA.


Subject(s)
Aflatoxin B1/biosynthesis , Aspergillus/metabolism , Fungal Proteins/metabolism , Sterigmatocystin/analogs & derivatives , Xanthones/chemistry , Aspergillus/genetics , Fungal Proteins/genetics , Multigene Family , Sterigmatocystin/biosynthesis
4.
Toxins (Basel) ; 10(12)2018 12 05.
Article in English | MEDLINE | ID: mdl-30563113

ABSTRACT

The dichlorvos-ammonia (DV-AM) method is a simple but sensitive visual method for detecting aflatoxigenic fungi. Here we sought to develop a selective medium that is appropriate for the growth of aflatoxigenic fungi among soil mycoflora. We examined the effects of different concentrations of carbon sources (sucrose and glucose) and detergents (deoxycholate (DOC), Triton X-100, and Tween 80) on microorganisms in soils, using agar medium supplemented with chloramphenicol. The results demonstrated that 5⁻10% sucrose concentrations and 0.1⁻0.15% DOC concentrations were appropriate for the selective detection of aflatoxigenic fungi in soil. We also identified the optimal constituents of the medium on which the normal rapid growth of Rhizopus sp. was completely inhibited. By using the new medium along with the DV-AM method, we succeeded in the isolation of aflatoxigenic fungi from non-agricultural fields in Fukui city, Japan. The fungi were identified as Aspergillus nomius based on their calmodulin gene sequences. These results indicate that the new medium will be useful in practice for the detection of aflatoxigenic fungi in soil samples including those from non-agricultural environments.


Subject(s)
Aspergillus/isolation & purification , Culture Media/pharmacology , Rhizopus/isolation & purification , Aflatoxins/metabolism , Ammonia , Aspergillus/drug effects , Aspergillus/physiology , Deoxycholic Acid/pharmacology , Detergents/pharmacology , Dichlorvos , Glucose/pharmacology , Microbiological Techniques , Octoxynol/pharmacology , Polysorbates/pharmacology , Rhizopus/drug effects , Rhizopus/physiology , Soil Microbiology , Sucrose/pharmacology
5.
Toxins (Basel) ; 10(7)2018 06 27.
Article in English | MEDLINE | ID: mdl-29954142

ABSTRACT

The dichlorvos⁻ammonia (DV⁻AM) method is a sensitive method for distinguishing aflatoxigenic fungi by detecting red (positive) colonies. In this study, the DV⁻AM method was applied for the isolation of aflatoxigenic and atoxigenic fungi from soil samples from a maize field in Mexico. In the first screening, we obtained two isolates from two soil subsamples of 20 independent samples and, in the second screening, we obtained two isolates from one subsample of these. Morphological and phylogenic analyses of the two isolates (MEX-A19-13, MEX-A19-2nd-5) indicated that they were Aspergillus flavus located in the A. flavus clade. Chemical analyses demonstrated that one isolate could produce B-type aflatoxins, while the other produced no aflatoxins. These results demonstrate that the DV⁻AM method is useful for the isolation of both aflatoxigenic and atoxigenic Aspergilli.


Subject(s)
Aflatoxins/analysis , Aspergillus/isolation & purification , Zea mays/microbiology , Aflatoxins/metabolism , Ammonia , Aspergillus/genetics , Aspergillus/metabolism , Dichlorvos , Environmental Monitoring , Mexico , Phylogeny , Soil Microbiology
6.
Appl Microbiol Biotechnol ; 99(24): 10681-94, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26300294

ABSTRACT

Aflatoxins (AFs) are carcinogenic and toxic secondary metabolites produced mainly by Aspergillus flavus and Aspergillus parasiticus. To monitor and regulate the AF contamination of crops, a sensitive and precise detection method for these toxigenic fungi in environments is necessary. We herein developed a novel visual detection method, the dichlorvos-ammonia (DV-AM) method, for identifying AF-producing fungi using DV and AM vapor on agar culture plates, in which DV inhibits the esterase in AF biosynthesis, causing the accumulation of anthraquinone precursors (versiconal hemiacetal acetate and versiconol acetate) of AFs in mycelia on the agar plate, followed by a change in the color of the colonies from light yellow to brilliant purple-red by the AM vapor treatment. We also investigated the appropriate culture conditions to increase the color intensity. It should be noted that other species producing the same precursors of AFs such as Aspergillus nidulans and Aspergillus versicolor could be discriminated from the Aspergillus section Flavi based on the differences of their phenotypes. The DV-AM method was also useful for the isolation of nonaflatoxigenic fungi showing no color change, for screening microorganisms that inhibit the AF production by fungi, and for the characterization of the fungi infecting corn kernels. Thus, the DV-AM method can provide a highly sensitive and visible indicator for the detection of aflatoxigenic fungi.


Subject(s)
Aflatoxins/metabolism , Ammonia , Aspergillus flavus/metabolism , Aspergillus nidulans/metabolism , Dichlorvos , Microbiological Techniques/methods , Staining and Labeling/methods , Color
7.
Front Microbiol ; 6: 78, 2015.
Article in English | MEDLINE | ID: mdl-25713569

ABSTRACT

GI-VII-6 is a chromosomally integrated multidrug resistance genomic island harbored by a specific clone of Salmonella enterica serovar Typhimurium (S.Typhimurium). It contains a gene encoding CMY-2 ß-lactamase (bla CMY-2), and therefore contributes to extended-spectrum cephalosporin resistance. To elucidate the significance of GI-VII-6 on adaptive evolution, spontaneous mutants of S. Typhimurium strain L-3553 were selected on plates containing cefotaxime (CTX). The concentrations of CTX were higher than its minimum inhibition concentration to the parent strain. The mutants appeared on the plates containing 12.5 and 25 mg/L CTX at a frequency of 10(-6) and 10(-8), respectively. No colonies were observed at higher CTX concentrations. The copy number of bla CMY-2 increased up to 85 per genome in the mutants, while the parent strain contains one copy of that in the chromosome. This elevation was accompanied by increased amount of transcription. The bla CMY-2 copy number in the mutants drastically decreased in the absence of antimicrobial selection pressure. Southern hybridization analysis and short-read mapping indicated that the entire 125 kb GI-VII-6 or parts of it were tandemly amplified. GI-VII-6 amplification occurred at its original position, although it also transposed to other locations in the genome in some mutants, including an endogenous plasmid in some of the mutants, leading to the amplification of GI-VII-6 at different loci. Insertion sequences were observed at the junction of the amplified regions in the mutants, suggesting their significant roles in the transposition and amplification. Plasmid copy number in the selected mutants was 1.4 to 4.4 times higher than that of the parent strain. These data suggest that transposition and amplification of the bla CMY-2-containing region, along with the copy number variation of the plasmid, contributed to the extensive amplification of bla CMY-2 and increased resistance to CTX.

8.
Fungal Genet Biol ; 49(9): 744-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22750656

ABSTRACT

Aspergillus parasiticus produces the minor aflatoxins M(1) (AFM(1)), M(2) (AFM(2)), GM(1) (AFGM(1)), and GM(2) (AFGM(2)), as well as the major aflatoxins B(1) (AFB(1)), B(2) (AFB(2)), G(1) (AFG(1)), and G(2) (AFG(2)). Feeding of A. parasiticus with aspertoxin (12c-hydroxyOMST) caused AFM(1) and AFGM(1), and cell-free experiments using the microsomal fraction of A. parasiticus and aspertoxin caused production of AFM(1), indicating that aspertoxin is a precursor of AFM(1) and AFGM(1). Feeding of the same fungus with O-methylsterigmatocystin (OMST) caused AFM(1) and AFGM(1) together with AFB(1) and AFG(1); feeding with dihydroOMST (DHOMST) caused AFM(2) and AFGM(2) together with AFB(2) and AFG(2). Incubation of either the microsomal fraction or OrdA enzyme-expressing yeast with OMST caused production of aspertoxin together with AFM(1) and AFB(1). These results demonstrated that the OrdA enzyme catalyzes both 12c-hydroxylation reaction from OMST to aspertoxin and the successive reaction from aspertoxin to AFM(1). In contrast, feeding of the fungus with AFB(1) did not produce any AFM(1), demonstrating that M-/GM-aflatoxins are not produced from B-/G-aflatoxins. Furthermore, AFM(1) together with AFB(1) and AFG(1) was also produced from 11-hydroxyOMST (HOMST) in feeding experiment of A. parasiticus, whereas no aflatoxins were produced when used the ordA deletion mutant. These results demonstrated that OrdA enzyme can also catalyze 12c-hydroxylation of HOMST to produce 11-hydroxyaspertoxin, which serves as a precursor for the production of AFM(1) and AFGM(1). The same pathway may work for the production of AFM(2) and AFGM(2) from DHOMST and dihydroHOMST through the formation of dihydroaspertoxin and dihydro-11-hydroxyaspertoxin, respectively.


Subject(s)
Aflatoxins/biosynthesis , Aspergillus/enzymology , Fungal Proteins/metabolism , Aflatoxins/chemistry , Aspergillus/genetics , Aspergillus/metabolism , Biocatalysis , Biosynthetic Pathways , Fungal Proteins/chemistry , Fungal Proteins/genetics , Sequence Deletion
10.
Biosci Biotechnol Biochem ; 75(1): 181-4, 2011.
Article in English | MEDLINE | ID: mdl-21228470

ABSTRACT

Five carboxin-resistant mutants from Aspergillus oryzae were characterized by the sensitivities of their mycelial growth and succinate dehydrogenase (SDH) activity to carboxin and three related fungicides. Despite a significant resistance to carboxin, exhibited by all the mutants, their patterns of sensitivity to the other fungicides was distinct. This provides clues to the molecular interaction between SDH and these fungicides.


Subject(s)
Aspergillus oryzae/drug effects , Aspergillus oryzae/genetics , Carboxin/toxicity , Drug Resistance, Fungal/genetics , Fungicides, Industrial/toxicity , Mutation , Aspergillus oryzae/enzymology , Mycelium/drug effects , Mycelium/enzymology , Mycelium/genetics , Succinate Dehydrogenase/metabolism
11.
Appl Microbiol Biotechnol ; 90(2): 635-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21153813

ABSTRACT

In aflatoxin biosynthesis, aflatoxins G(1) (AFG(1)) and B(1) (AFB(1)) are independently produced from a common precursor, O-methylsterigmatocystin (OMST). Recently, 11-hydroxy-O-methylsterigmatocystin (HOMST) was suggested to be a later precursor involved in the conversion of OMST to AFB(1), and conversion of HOMST to AFB(1) was catalyzed by OrdA enzyme. However, the involvement of HOMST in AFG(1) formation has not been determined. In this work, HOMST was prepared by incubating OrdA-expressing yeast with OMST. Feeding Aspergillus parasiticus with HOMST allowed production of AFG(1) as well as AFB(1). In cell-free systems, HOMST was converted to AFG(1) when the microsomal fraction, the cytosolic fraction from A. parasiticus, and yeast expressing A. parasiticus OrdA were added. These results demonstrated (1) HOMST is produced from OMST by OrdA, (2) HOMST is a precursor of AFG(1) as well as AFB(1), and (3) three enzymes, OrdA, CypA, and NadA, and possibly other unknown enzymes are involved in conversion of HOMST to AFG(1).


Subject(s)
Aflatoxins/biosynthesis , Aspergillus/enzymology , Genes, Fungal , Sterigmatocystin/analogs & derivatives , Cell-Free System/metabolism , Fungal Proteins , Gene Deletion , Gene Expression Regulation, Fungal , Multigene Family , Saccharomyces cerevisiae/genetics , Sterigmatocystin/chemistry
12.
Fungal Genet Biol ; 46(3): 221-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211038

ABSTRACT

Three reactions from hydroxyversicolorone to versicolorone, from versiconal hemiacetal acetate to versiconol acetate, and from versiconal to versiconol are involved in a metabolic grid in aflatoxin biosynthesis. This work demonstrated that the same reductase of Aspergillus parasiticus catalyzes the three reactions. The gene (named vrdA) encoding the reductase was cloned, and its sequence did not show homology to any regions in aflatoxin gene cluster. Its cDNA encoding a 38,566Da protein was separated by three introns in the genome. Deletion of the vrdA gene in A. parasiticus caused a significant decrease in enzyme activity, but did not affect aflatoxin productivity of the fungi. Although the vrdA gene was expressed in culture conditions conducive to aflatoxin production, it was expressed even in the aflR deletion mutant. These results suggest that the vrdA is not an aflatoxin biosynthesis gene, although it actually participates in aflatoxin biosynthesis in cells.


Subject(s)
Aflatoxins/biosynthesis , Aspergillus/enzymology , Fungal Proteins/metabolism , Oxidoreductases/metabolism , Amino Acid Sequence , Anthraquinones/metabolism , Aspergillus/genetics , Base Sequence , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Gene Deletion , Introns , Molecular Sequence Data , Molecular Weight , Multigene Family , Oxidoreductases/chemistry , Oxidoreductases/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid
13.
Fungal Genet Biol ; 46(1): 67-76, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18992352

ABSTRACT

Mutants exhibiting resistance to the fungicide, carboxin, were isolated from Aspergillus oryzae, and the mutations in the three gene loci, which encode succinate dehydrogenase (SDH) B, C, and D subunits, were identified to be independently responsible for the resistance. A structural model of the SDH revealed the different mechanisms that confer carboxin-resistance in different mutations. The mutant AosdhB gene (AosdhB(cxr)) was further examined for possible use as a transformant selection marker. After transformation with AosdhB(cxr), carboxin-resistant colonies appeared within 4 days of culture, and all of the examined colonies carried the transgene. Insertion analyses revealed that the AosdhB(cxr) gene was integrated into AosdhB locus via homologous recombination at high efficiency. Furthermore, AosdhB(cxr) functioned as a successful selection marker in a transformation experiment in Aspergillus parasiticus, suggesting that this transformation system can be used for Aspergillus species.


Subject(s)
Aspergillus oryzae/drug effects , Aspergillus oryzae/genetics , Carboxin/pharmacology , Drug Resistance, Fungal/genetics , Fungicides, Industrial/pharmacology , Transformation, Genetic , Aspergillus oryzae/growth & development , Carboxin/metabolism , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Mutation , Succinate Dehydrogenase/metabolism
14.
Fungal Genet Biol ; 45(7): 1081-93, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18486503

ABSTRACT

The nadA gene is present at the end of the aflatoxin gene cluster in the genome of Aspergillus parasiticus as well as in Aspergillus flavus. RT-PCR analyses showed that the nadA gene was expressed in an aflatoxin-inducible YES medium, but not in an aflatoxin-non-inducible YEP medium. The nadA gene was not expressed in the aflR gene-deletion mutant, irrespective of the culture medium used. To clarify the nadA gene's function, we disrupted the gene in aflatoxigenic A. parasiticus. The four nadA-deletion mutants that were isolated commonly accumulated a novel yellow-fluorescent pigment (named NADA) in mycelia as well as in culture medium. When the mutants and the wild-type strain were cultured for 3 days in YES medium, the mutants each produced about 50% of the amounts of G-group aflatoxins that the wild-type strain produced. In contrast, the amounts of B-group aflatoxins did not significantly differ between the mutants and the wild-type strain. The NADA pigment was so unstable that it could non-enzymatically change to aflatoxin G(1) (AFG(1)). LC-MS measurement showed that the molecular mass of NADA was 360, which is 32 higher than that of AFG(1). We previously reported that at least one cytosol enzyme, together with two other microsome enzymes, is necessary for the formation of AFG(1) from O-methylsterigmatocystin (OMST) in the cell-free system of A. parasiticus. The present study confirmed that the cytosol fraction of the wild-type A.parasiticus strain significantly enhanced the AFG(1) formation from OMST, whereas the cytosol fraction of the nadA-deletion mutant did not show the same activity. Furthermore, the cytosol fraction of the wild-type strain showed the enzyme activity catalyzing the reaction from NADA to AFG(1), which required NADPH or NADH, indicating that NADA is a precursor of AFG(1); in contrast, the cytosol fraction of the nadA-deletion mutant did not show the same enzyme activity. These results demonstrated that the NadA protein is the cytosol enzyme required for G-aflatoxin biosynthesis from OMST, and that it catalyzes the reaction from NADA to AFG(1), the last step in G-aflatoxin biosynthesis.


Subject(s)
Aflatoxins/biosynthesis , Aspergillus/enzymology , Fungal Proteins/metabolism , Aflatoxins/analysis , Aspergillus/chemistry , Aspergillus/genetics , Aspergillus/metabolism , Biosynthetic Pathways , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Pigments, Biological/chemistry , Pigments, Biological/genetics , Pigments, Biological/metabolism , Sequence Deletion
15.
Biosci Biotechnol Biochem ; 72(3): 724-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18323645

ABSTRACT

When 10 strains of lactic acid bacteria were incubated with 5'-hydroxyaverantin (HAVN), a precursor of aflatoxins, seven of them converted HAVN to averufin; the same reaction is found in aflatoxin biosynthesis of aflatoxigenic fungi. These bacteria had a dehydrogenase that catalyzed the reaction from HAVN to 5'-oxoaverantin (OAVN), which was so unstable that it was easily converted to averufin. The enzyme was purified from Lactobacillus brevis IFO 12005. The molecular mass of the enzyme was 100 kDa on gel filtration chromatography and 33 kDa on SDS polyacrylamide gel electrophoresis (SDS-PAGE). The gene encoding the enzyme was cloned and sequenced. The deduced protein consisted of 249 amino acids, and its estimated molecular mass was 25,873, in agreement with that by time of flight mass spectrometry (TOF MS) analysis. Although the deduced amino acid sequence showed about 50% identity to those reported for alcohol dehydrogenases from L. brevis or L. kefir, the commercially available alcohol dehydrogenase from L. kefir did not convert HAVN to OAVN. Aspergillus parasiticus HAVN dehydrogenase showed about 25% identity in amino acid sequence with the dehydrogenase and also with these two alcohol dehydrogenases.


Subject(s)
Aflatoxins/biosynthesis , Alcohol Dehydrogenase/genetics , Ketone Oxidoreductases/genetics , Levilactobacillus brevis/enzymology , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/isolation & purification , Amino Acid Sequence , Anthraquinones/metabolism , Catalysis , Cloning, Molecular , Ketone Oxidoreductases/chemistry , Ketone Oxidoreductases/isolation & purification , Molecular Sequence Data
16.
Plant J ; 55(2): 212-23, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18363783

ABSTRACT

The RIN gene encodes a putative MADS box transcription factor that controls tomato fruit ripening, and its ripening inhibitor (rin) mutation yields non-ripening fruit. In this study, the molecular properties of RIN and the rin mutant protein were clarified. The results revealed that the RIN protein accumulates in ripening fruit specifically and is localized in the nucleus of the cell. In vitro studies revealed that RIN forms a stable homodimer that binds to MADS domain-specific DNA sites. Analysis of binding site selection experiments revealed that the consensus binding sites of RIN highly resemble those of the SEPALLATA (SEP) proteins, which are Arabidopsis MADS box proteins that control the identity of floral organs. RIN exhibited a transcription-activating function similar to that exhibited by the SEP proteins. These results indicate that RIN exhibits similar molecular functions to SEP proteins although they play distinctly different biological roles. In vivo assays revealed that RIN binds to the cis-element of LeACS2. Our results also revealed that the rin mutant protein accumulates in the mutant fruit and exhibits a DNA-binding activity similar to that exhibited by the wild-type protein, but has lost its transcription-activating function, which in turn would inhibit ripening in mutant fruit.


Subject(s)
DNA, Plant/metabolism , Fruit/metabolism , Gene Expression Regulation, Plant/physiology , Plant Proteins/metabolism , Solanum lycopersicum/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Amino Acid Sequence , Base Sequence , Fruit/cytology , Solanum lycopersicum/cytology , Molecular Sequence Data , Mutation , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation/physiology
17.
Appl Environ Microbiol ; 71(6): 2999-3006, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15932995

ABSTRACT

In the aflatoxin biosynthetic pathway, 5'-oxoaverantin (OAVN) cyclase, the cytosolic enzyme, catalyzes the reaction from OAVN to (2'S,5'S)-averufin (AVR) (E. Sakuno, K. Yabe, and H. Nakajima, Appl. Environ. Microbiol. 69:6418-6426, 2003). Interestingly, the N-terminal 25-amino-acid sequence of OAVN cyclase completely matched an internal sequence of the versiconal (VHOH) cyclase that was deduced from its gene (vbs). The purified OAVN cyclase also catalyzed the reaction from VHOH to versicolorin B (VB). In a competition experiment using the cytosol fraction of Aspergillus parasiticus, a high concentration of VHOH inhibited the enzyme reaction from OAVN to AVR, and instead VB was newly formed. The recombinant Vbs protein, which was expressed in Pichia pastoris, showed OAVN cyclase activity, as well as VHOH cyclase activity. A mutant of A. parasiticus SYS-4 (= NRRL 2999) with vbs deleted accumulated large amounts of OAVN, 5'-hydroxyaverantin, averantin, AVR, and averufanin in the mycelium. These results indicated that the cyclase encoded by the vbs gene is also involved in the reaction from OAVN to AVR in aflatoxin biosynthesis. Small amounts of VHOH, VB, and aflatoxins also accumulated in the same mutant, and this accumulation may have been due to an unknown enzyme(s) not involved in aflatoxin biosynthesis. This is the first report of one enzyme catalyzing two different reactions in a pathway of secondary metabolism.


Subject(s)
Aflatoxins/biosynthesis , Anthraquinones/metabolism , Aspergillus/enzymology , Hydro-Lyases/metabolism , Amino Acid Sequence , Aspergillus/genetics , Gene Expression Regulation, Fungal , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Molecular Sequence Data , Pichia/enzymology , Pichia/genetics , Recombinant Proteins
18.
Appl Environ Microbiol ; 71(6): 3192-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15933021

ABSTRACT

The pathway oxoaverantin (OAVN) --> averufin (AVR) --> hydroxyversicolorone (HVN) --> versiconal hemiacetal acetate (VHA) is involved in aflatoxin biosynthesis, and the cypX and moxY genes, which are present in the aflatoxin gene cluster, have been previously suggested to be involved in this pathway. To clarify the function of these two genes in more detail, we disrupted the genes in aflatoxigenic Aspergillus parasiticus NRRL 2999. The cypX-deleted mutant lost aflatoxin productivity and accumulated AVR in the mycelia. Although this mutant converted HVN, versicolorone (VONE), VHA, and versiconol acetate (VOAc) to aflatoxins in feeding experiments, it could not produce aflatoxins from either OAVN or AVR. The moxY-deleted mutant also lost aflatoxin productivity, whereas it newly accumulated HVN and VONE. In feeding experiments, this mutant converted either VHA or VOAc to aflatoxins but did not convert OAVN, AVR, HVN, or VONE to aflatoxins. These results demonstrated that cypX encodes AVR monooxygenase, catalyzing the reaction from AVR to HVN, and moxY encodes HVN monooxygenase, catalyzing a Baeyer-Villiger reaction from HVN to VHA as well as from VONE to VOAc. In this work, we devised a simple and rapid method to extract DNA from many fungi for PCR analyses in which cell disruption with a shaker and phenol extraction were combined.


Subject(s)
Aflatoxins/biosynthesis , Aspergillus/enzymology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Oxygenases/genetics , Oxygenases/metabolism , Anthraquinones/metabolism , Aspergillus/genetics , Aspergillus/growth & development , DNA, Fungal/analysis , DNA, Fungal/isolation & purification , Gene Deletion , Gene Expression Regulation, Fungal , Polymerase Chain Reaction
19.
Appl Environ Microbiol ; 70(12): 7466-73, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15574949

ABSTRACT

Aflatoxins are potent carcinogenic and toxic substances that are produced primarily by Aspergillus flavus and Aspergillus parasiticus. We found that a bacterium remarkably inhibited production of norsolorinic acid, a precursor of aflatoxin, by A. parasiticus. This bacterium was identified as Achromobacter xylosoxidans based on its 16S ribosomal DNA sequence and was designated A. xylosoxidans NFRI-A1. A. xylosoxidans strains commonly showed similar inhibition. The inhibitory substance(s) was excreted into the medium and was stable after heat, acid, or alkaline treatment. Although the bacterium appeared to produce several inhibitory substances, we finally succeeded in purifying a major inhibitory substance from the culture medium using Diaion HP20 column chromatography, thin-layer chromatography, and high-performance liquid chromatography. The purified inhibitory substance was identified as cyclo(L-leucyl-L-prolyl) based on physicochemical methods. The 50% inhibitory concentration for aflatoxin production by A. parasiticus SYS-4 (= NRRL2999) was 0.20 mg ml(-1), as determined by the tip culture method. High concentrations (more than 6.0 mg ml(-1)) of cyclo(L-leucyl-L-prolyl) further inhibited fungal growth. Similar inhibitory activities were observed with cyclo(D-leucyl-D-prolyl) and cyclo(L-valyl-L-prolyl), whereas cyclo(D-prolyl-L-leucyl) and cyclo(L-prolyl-D-leucyl) showed weaker activities. Reverse transcription-PCR analyses showed that cyclo(L-leucyl-L-prolyl) repressed transcription of the aflatoxin-related genes aflR, hexB, pksL1, and dmtA. This is the first report of a cyclodipeptide that affects aflatoxin production.


Subject(s)
Achromobacter denitrificans/metabolism , Aflatoxins/antagonists & inhibitors , Aspergillus/drug effects , Peptides, Cyclic/pharmacology , Achromobacter denitrificans/classification , Achromobacter denitrificans/genetics , Achromobacter denitrificans/growth & development , Aflatoxins/biosynthesis , Anthraquinones/metabolism , Aspergillus/metabolism , Culture Media , Fungal Proteins/genetics , Fungal Proteins/metabolism , Molecular Sequence Data , Peptides, Cyclic/biosynthesis , Peptides, Cyclic/chemistry , Peptides, Cyclic/isolation & purification , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
20.
Appl Environ Microbiol ; 70(6): 3593-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15184162

ABSTRACT

In aflatoxin biosynthesis, the pathway for the conversion of 1-hydroxyversicolorone to versiconal hemiacetal acetate (VHA) to versiconal (VHOH) is part of a metabolic grid. In the grid, the steps from VHA to VHOH and from versiconol acetate (VOAc) to versiconol (VOH) may be catalyzed by the same esterase. Several esterase activities are associated with the conversion of VHA to VHOH, but only one esterase gene (estA) is present in the complete aflatoxin gene cluster of Aspergillus parasiticus. We deleted the estA gene from A. parasiticus SRRC 2043, an O-methylsterigmatocystin (OMST)-accumulating strain. The estA-deleted mutants were pigmented and accumulated mainly VHA and versicolorin A (VA). A small amount of VOAc and other downstream aflatoxin intermediates, including VHOH, versicolorin B, and OMST, also were accumulated. In contrast, a VA-accumulating mutant, NIAH-9, accumulated VA exclusively and neither VHA nor VOAc were produced. Addition of the esterase inhibitor dichlorvos (dimethyl 2,2-dichlorovinylphosphate) to the transformation recipient strain RHN1, an estA-deleted mutant, or NIAH-9 resulted in the accumulation of only VHA and VOAc. In in vitro enzyme assays, the levels of the esterase activities catalyzing the conversion of VHA to VHOH in the cell extracts of two estA-deleted mutants were decreased to approximately 10% of that seen with RHN1. Similar decreases in the esterase activities catalyzing the conversion of VOAc to VOH were also obtained. Thus, the estA-encoded esterase catalyzes the conversion of both VHA to VHOH and VOAc to VOH during aflatoxin biosynthesis.


Subject(s)
Aflatoxins/biosynthesis , Anthraquinones/metabolism , Aspergillus/enzymology , Carboxylic Ester Hydrolases/metabolism , Fungal Proteins , Aspergillus/genetics , Aspergillus/isolation & purification , Carboxylic Ester Hydrolases/genetics , Culture Media , Dichlorvos/pharmacology , Gene Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...