Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 757
Filter
1.
Toxins (Basel) ; 16(6)2024 May 31.
Article in English | MEDLINE | ID: mdl-38922150

ABSTRACT

Aflatoxin B1 (AFB1) contamination is a food safety issue threatening human health globally. Biodegradation is an effective method for overcoming this problem, and many microorganisms have been identified as AFB1-degrading strains. However, the response mechanisms of these microbes to AFB1 remain unclear. More degrading enzymes, especially of new types, need to be discovered. In this study, a novel AFB1-degrading strain, DDC-4, was isolated using coumarin as the sole carbon source. This strain was identified as Bacillus halotolerans through physiological, biochemical, and molecular methods. The strain's degradation activity was predominantly attributable to thermostable extracellular proteins (degradation rate remained approximately 80% at 90 °C) and was augmented by Cu2+ (95.45% AFB1 was degraded at 48 h). Alpha/beta hydrolase (arylesterase) was selected as candidate AFB1-degrading enzymes for the first time as a gene encoding this enzyme was highly expressed in the presence of AFB1. Moreover, AFB1 inhibited many genes involved in the nucleotide synthesis of strain DDC-4, which is possibly the partial molecular mechanism of AFB1's toxicity to microorganisms. To survive under this stress, sporulation-related genes were induced in the strain. Altogether, our study identified a novel AFB1-degrading strain and explained its response mechanisms to AFB1, thereby providing new insights for AFB1 biodegradation.


Subject(s)
Aflatoxin B1 , Bacillus , Aflatoxin B1/metabolism , Bacillus/metabolism , Bacillus/genetics , Biodegradation, Environmental , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
Int J Mol Sci ; 25(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38928160

ABSTRACT

Aflatoxin B1 (AFB1) contamination is a serious threat to nutritional safety and public health. The CotA-laccase from Bacillus licheniformis ANSB821 previously reported by our laboratory showed great potential to degrade AFB1 without redox mediators. However, the use of this CotA-laccase to remove AFB1 in animal feed is limited because of its low catalytic efficiency and low expression level. In order to make better use of this excellent enzyme to effectively degrade AFB1, twelve mutants of CotA-laccase were constructed by site-directed mutagenesis. Among these mutants, E186A and E186R showed the best degradation ability of AFB1, with degradation ratios of 82.2% and 91.8% within 12 h, which were 1.6- and 1.8-times higher than those of the wild-type CotA-laccase, respectively. The catalytic efficiencies (kcat/Km) of E186A and E186R were found to be 1.8- and 3.2-times higher, respectively, than those of the wild-type CotA-laccase. Then the expression vectors pPICZαA-N-E186A and pPICZαA-N-E186R with an optimized signal peptide were constructed and transformed into Pichia pastoris GS115. The optimized signal peptide improved the secretory expressions of E186A and E186R in P. pastoris GS115. Collectively, the current study provided ideal candidate CotA-laccase mutants for AFB1 detoxification in food and animal feed and a feasible protocol, which was desperately needed for the industrial production of CotA-laccases.


Subject(s)
Aflatoxin B1 , Bacillus licheniformis , Bacterial Proteins , Laccase , Aflatoxin B1/metabolism , Bacillus licheniformis/genetics , Bacillus licheniformis/metabolism , Bacillus licheniformis/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Laccase/metabolism , Laccase/genetics , Mutagenesis, Site-Directed , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Saccharomycetales
3.
Food Chem ; 455: 139862, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38833866

ABSTRACT

Laccase mediators possess advantage of oxidizing substrates with high redox potentials, such as aflatoxin B1 (AFB1). High costs of chemically synthesized mediators limit laccase industrial application. In this study, thin stillage extract (TSE), a byproduct of corn-based ethanol fermentation was investigated as the potential natural mediator of laccases. Ferulic acid, p-coumaric acid, and vanillic acid were identified as the predominant phenolic compounds of TSE. With the assistance of 0.05 mM TSE, AFB1 degradation activity of novel laccase Glac1 increased by 17 times. The promoting efficiency of TSE was similar to ferulic acid, but superior to vanillic acid and p-coumaric acid, with 1.2- and 1.3-fold increases, respectively. After Glac1-TSE treatment, two oxidation products were identified. Ames test showed AFB1 degradation products lost mutagenicity. Meanwhile, TSE also showed 1.3-3.0 times promoting effect on laccase degradation activity in cereal flours. Collectively, a safe and highly efficient natural mediator was obtained for aflatoxin detoxification.


Subject(s)
Laccase , Zea mays , Laccase/metabolism , Laccase/chemistry , Zea mays/chemistry , Zea mays/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Aflatoxin B1/chemistry , Aflatoxin B1/metabolism , Oxidation-Reduction , Plant Extracts/chemistry , Fermentation , Coumaric Acids/chemistry , Coumaric Acids/metabolism
4.
Int J Biol Macromol ; 273(Pt 1): 133115, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38871108

ABSTRACT

Aflatoxin B1 (AFB1) contamination of oils is a serious concern for the safety of edible oil consumers. Enzyme-assisted detoxification of AFB1 is an efficient and safe method for decontaminating oils, but pristine enzymes are unstable in oils and require modifications before use. Therefore, we designed a novel and magnetically separable laccase-carrying biocatalyst containing spent-mushroom-substrate (SMS)-derived biochar (BF). Laccase was immobilized on NH2-activated magnetic biochar (BF-NH2) through covalent crosslinking, which provided physicochemical stability to the immobilized enzyme. After 30 days of storage at 4 °C, the immobilized laccase (product named "BF-NH2-Lac") retained ~95 % of its initial activity, while after five repeated cycles of ABTS oxidation, ~85 % activity retention was observed. BF-NH2-Lac was investigated for the oxidative degradation of AFB1, which exhibited superior performance compared to free laccase. Among many tested natural compounds as mediators, p-coumaric acid proved the most efficient in activating laccase for AFB1 degradation. BF-NH2-Lac demonstrated >90 % removal of AFB1 within 5.0 h, while the observed degradation efficiency in corn oil and buffer was comparable. An insight into the adsorptive and degradative removal of AFB1 revealed that AFB1 removal was governed mainly by degradation. The coexistence of multi-mycotoxins did not significantly affect the AFB1 degradation capability of BF-NH2-Lac. Investigation of the degradation products revealed the transformation of AFB1 into non-toxic AFQ1, while corn oil quality remained unaffected after BF-NH2-Lac treatment. Hence, this study holds practical importance for the research, knowledge-base and industrial application of newly proposed immobilized enzyme products.


Subject(s)
Aflatoxin B1 , Charcoal , Corn Oil , Enzymes, Immobilized , Laccase , Laccase/metabolism , Laccase/chemistry , Enzymes, Immobilized/metabolism , Enzymes, Immobilized/chemistry , Charcoal/chemistry , Aflatoxin B1/chemistry , Aflatoxin B1/metabolism , Corn Oil/chemistry , Porosity , Recycling
5.
J Agric Food Chem ; 72(26): 14481-14497, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38897919

ABSTRACT

Aflatoxin B1 (AFB1), the most toxic and harmful mycotoxin, has a high likelihood of occurring in animal feed and human food, which seriously affects agriculture and food safety and endangers animal and human health. Recently, natural plant products have attracted widespread attention due to their low toxicity, high biocompatibility, and simple composition, indicating significant potential for resisting AFB1. The mechanisms by which these phytochemicals resist toxins mainly involve antioxidative, anti-inflammatory, and antiapoptotic pathways. Moreover, these substances also inhibit the genotoxicity of AFB1 by directly influencing its metabolism in vivo, which contributes to its elimination. Here, we review various phytochemicals that resist AFB1 and their anti-AFB1 mechanisms in different animals, as well as the common characteristics of phytochemicals with anti-AFB1 function. Additionally, the shortcomings of current research and future research directions will be discussed. Overall, this comprehensive summary contributes to the better application of phytochemicals in agriculture and food safety.


Subject(s)
Aflatoxin B1 , Agriculture , Food Contamination , Phytochemicals , Aflatoxin B1/metabolism , Aflatoxin B1/chemistry , Phytochemicals/chemistry , Phytochemicals/metabolism , Phytochemicals/pharmacology , Animals , Humans , Food Contamination/analysis , Food Contamination/prevention & control , Inactivation, Metabolic , Food Safety , Food Technology
6.
J Hazard Mater ; 474: 134822, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38850943

ABSTRACT

The disturbed gut microbiota is a key factor in activating the aflatoxin B1 (AFB1)-induced liver pyroptosis by promoting inflammatory hepatic injury; however, the pathogen associated molecular pattern (PAMP) from disturbed gut microbiota and its mechanism in activating liver pyroptosis remain undefined. By transplanting AFB1-originated fecal microbiota and sterile fecal microbial metabolites filtrate, we determined the association of PAMP in AFB1-induced liver pyroptosis. Notably, AFB1-originated sterile fecal microbial metabolites filtrate were more active in triggering liver pyroptosis in mice, as compared to parental fecal microbiota. This result supported a critical role of the metabolic homeostasis of gut microbiota in AFB1-induced liver pyroptosis, rather than an injurious response to direct exposure of AFB1 in liver. Among the gut-microbial metabolites, pipecolic acid and norepinephrine were proposed to bind TLR4 and NLRP3, the upstream proteins of pyroptosis signaling pathway. Besides, the activations of TLR4 and NLRP3 were linearly correlated with the concentrations of pipecolic acid and norepinephrine in the serum of mice. In silenced expression of TLR4 and NLRP3 in HepG2 cells, pipecolic acid or norepinephrine did not able to activate hepatocyte pyroptosis. These results demonstrated the necessity of gut microbial metabolism in sustaining liver homeostasis, as well as the potential to provide new insights into targeted intervention for AFB1 hepatotoxicity.


Subject(s)
Aflatoxin B1 , Gastrointestinal Microbiome , Liver , NLR Family, Pyrin Domain-Containing 3 Protein , Norepinephrine , Pipecolic Acids , Pyroptosis , Animals , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Pyroptosis/drug effects , Gastrointestinal Microbiome/drug effects , Pipecolic Acids/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Norepinephrine/metabolism , Hep G2 Cells , Male , Mice, Inbred C57BL , Toll-Like Receptor 4/metabolism , Mice , Feces/microbiology
7.
Ecotoxicol Environ Saf ; 279: 116449, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38759532

ABSTRACT

Over the past few years, there has been growing interest in the ability of insect larvae to convert various organic side-streams containing mycotoxins into insect biomass that can be used as animal feed. Various studies have examined the effects of exposure to aflatoxin B1 (AFB1) on a variety of insect species, including the larvae of the black soldier fly (BSFL; Hermetia illucens L.; Diptera: Stratiomyidae) and the housefly (HFL; Musca domestica L.; Diptera: Muscidae). Most of these studies demonstrated that AFB1 degradation takes place, either enzymatic and/or non-enzymatic. The possible role of feed substrate microorganisms (MOs) in this process has thus far not been investigated. The main objective of this study was therefore to investigate whether biotransformation of AFB1 occurred and whether it is caused by insect-enzymes and/or by microbial enzymes of MOs in the feed substrate. In order to investigate this, sterile and non-sterile feed substrates were spiked with AFB1 and incubated either with or without insect larvae (BSFL or HFL). The AFB1 concentration was determined via LC-MS/MS analyses and recorded over time. Approximately 50% of the initially present AFB1 was recovered in the treatment involving BSFL, which was comparable to the treatment without BSFL (60%). Similar patterns were observed for HFL. The molar mass balance of AFB1 for the sterile feed substrates with BSFL and HFL was 73% and 78%, respectively. We could not establish whether non-enzymatic degradation of AFB1 in the feed substrates occurred. The results showed that both BSFL and substrate-specific MOs play a role in the biotransformation of AFB1 as well as in conversion of AFB1 into aflatoxin P1 and aflatoxicol, respectively. In contrast, HFL did not seem to contribute to AFB1 degradation. The obtained results contribute to our understanding of aflatoxin metabolism by different insect species. This information is crucial for assessing the safety of feeding fly larvae with feed substrates contaminated with AFB1 with the purpose of subsequent use as animal feed.


Subject(s)
Aflatoxin B1 , Animal Feed , Biotransformation , Diptera , Houseflies , Larva , Animals , Aflatoxin B1/metabolism , Houseflies/metabolism , Animal Feed/analysis , Tandem Mass Spectrometry
8.
J Agric Food Chem ; 72(23): 13371-13381, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38809574

ABSTRACT

The enzymatic biodegradation of mycotoxins in food and feed has attracted the most interest in recent years. In this paper, the laccase gene from Bacillus swezeyi was cloned and expressed in Escherichia coli BL 21(D3). The sequence analysis indicated that the gene consisted of 1533 bp. The purified B. swezeyi laccase was analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis -12% with an estimated molecular weight of 56.7 kDa. The enzyme is thermo-alkali-tolerant, displaying the optimal degradation of zearalenone (ZEN) and aflatoxin B1 (AFB1) at pH 8 and 9, with incubation temperatures of 55 and 50 °C, respectively, within 24 h. The degradation potentials of the 50 µg of the enzyme against ZEN (5.0 µg/mL) and AFB1 (2.5 µg/mL) were 99.60 and 96.73%, respectively, within 24 h. To the best of our knowledge, this is the first study revealing the recombinant production of laccase from B. swezeyi, its biochemical properties, and potential use in ZEN and AFB1 degradation in vitro and in vivo.


Subject(s)
Aflatoxin B1 , Bacillus , Bacterial Proteins , Enzyme Stability , Laccase , Recombinant Proteins , Zearalenone , Laccase/genetics , Laccase/metabolism , Laccase/chemistry , Aflatoxin B1/metabolism , Aflatoxin B1/chemistry , Zearalenone/metabolism , Zearalenone/chemistry , Bacillus/enzymology , Bacillus/genetics , Bacillus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/chemistry , Hydrogen-Ion Concentration , Temperature , Molecular Weight , Escherichia coli/genetics , Escherichia coli/metabolism , Cloning, Molecular , Alkalies/metabolism , Alkalies/chemistry
9.
Sci China Life Sci ; 67(7): 1468-1478, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38703348

ABSTRACT

Dietary exposure to aflatoxin B1 (AFB1) is harmful to the health and performance of domestic animals. The hepatic cytochrome P450s (CYPs), CYP1A1 and CYP2A6, are the primary enzymes responsible for the bioactivation of AFB1 to the highly toxic exo-AFB1-8,9-epoxide (AFBO) in chicks. However, the transcriptional regulation mechanism of these CYP genes in the liver of chicks in AFB1 metabolism remains unknown. Dual-luciferase reporter assay, bioinformatics and site-directed mutation results indicated that specificity protein 1 (SP1) and activator protein-1 (AP-1) motifs were located in the core region -1,063/-948, -606/-541 of the CYP1A1 promoter as well as -636/-595, -503/-462, -147/-1 of the CYP2A6 promoter. Furthermore, overexpression and decoy oligodeoxynucleotide technologies demonstrated that SP1 and AP-1 were pivotal transcriptional activators regulating the promoter activity of CYP1A1 and CYP2A6. Moreover, bioactivation of AFB1 to AFBO could be increased by upregulation of CYP1A1 and CYP2A6 expression, which was trans-activated owing to the upregulalion of AP-1, rather than SP1, stimulated by AFB1-induced reactive oxygen species. Additionally, nano-selenium could reduce ROS, downregulate AP-1 expression and then decrease the expression of CYP1A1 and CYP2A6, thus alleviating the toxicity of AFB1. In conclusion, AP-1 and SP1 played important roles in the transactivation of CYP1A1 and CYP2A6 expression and further bioactivated AFB1 to AFBO in chicken liver, which could provide novel targets for the remediation of aflatoxicosis in chicks.


Subject(s)
Aflatoxin B1 , Chickens , Cytochrome P-450 CYP1A1 , Cytochrome P-450 CYP2A6 , Liver , Promoter Regions, Genetic , Sp1 Transcription Factor , Transcription Factor AP-1 , Animals , Aflatoxin B1/metabolism , Chickens/metabolism , Liver/metabolism , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/genetics , Cytochrome P-450 CYP2A6/metabolism , Cytochrome P-450 CYP2A6/genetics , Transcriptional Activation
10.
DNA Repair (Amst) ; 139: 103695, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795603

ABSTRACT

The base excision repair (BER) pathway is a precise and versatile mechanism of DNA repair that is initiated by DNA glycosylases. Endonuclease VIII-like 1 (NEIL1) is a bifunctional glycosylase/abasic site (AP) lyase that excises a damaged base and subsequently cleaves the phosphodiester backbone. NEIL1 is able to recognize and hydrolyze a broad range of oxidatively-induced base lesions and substituted ring-fragmented guanines, including aflatoxin-induced 8,9-dihydro-8-(2,6-diamino-4-oxo-3,4-dihydropyrimid-5-yl-formamido)-9-hydroxyaflatoxin B1 (AFB1-FapyGua). Due to NEIL1's protective role against these and other pro-mutagenic lesions, it was hypothesized that naturally occurring single nucleotide polymorphic (SNP) variants of NEIL1 could increase human risk for aflatoxin-induced hepatocellular carcinoma (HCC). Given that populations in South Asia experience high levels of dietary aflatoxin exposures and hepatitis B viral infections that induce oxidative stress, investigations on SNP variants of NEIL1 that occur in this region may have clinical implications. In this study, the most common South Asian variants of NEIL1 were expressed, purified, and functionally characterized. All tested variants exhibited activities and substrate specificities similar to wild type (wt)-NEIL1 on high-molecular weight DNA containing an array of oxidatively-induced base lesions. On short oligodeoxynucleotides (17-mers) containing either a site-specific apurinic/apyrimidinic (AP) site, thymine glycol (ThyGly), or AFB1-FapyGua, P206L-NEIL1 was catalytically comparable to wt-NEIL1, while the activities of NEIL1 variants Q67K and T278I on these substrates were ≈2-fold reduced. Variant T103A had a greatly diminished ability to bind to 17-mer DNAs, limiting the subsequent glycosylase and lyase reactions. Consistent with this observation, the rate of excision by T103A on 17-mer oligodeoxynucleotides containing ThyGly or AFB1-FapyGua could not be measured. However, the ability of T103A to excise ThyGly was improved on longer oligodeoxynucleotides (51-mers), with ≈7-fold reduced activity compared to wt-NEIL1. Our studies suggest that NEIL1 variant T103A may present a pathogenic phenotype that is limited in damage recognition, potentially increasing human risk for HCC.


Subject(s)
DNA Glycosylases , DNA Repair , Polymorphism, Single Nucleotide , DNA Glycosylases/metabolism , DNA Glycosylases/genetics , DNA Glycosylases/chemistry , Humans , Aflatoxin B1/metabolism , DNA Damage , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/enzymology , Substrate Specificity , Liver Neoplasms/genetics , Liver Neoplasms/enzymology
11.
Appl Microbiol Biotechnol ; 108(1): 348, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809353

ABSTRACT

Mycotoxin production by aflatoxin B1 (AFB1) -producing Aspergillus flavus Zt41 and sterigmatocystin (ST) -hyperproducer Aspergillus creber 2663 mold strains on corn and rice starch, both of high purity and nearly identical amylose-amylopectin composition, as the only source of carbon, was studied. Scanning electron microscopy revealed average starch particle sizes of 4.54 ± 0.635 µm and 10.9 ± 2.78 µm, corresponding to surface area to volume ratios of 127 1/µm for rice starch and 0.49 1/µm for corn starch. Thus, a 2.5-fold difference in particle size correlated to a larger, 259-fold difference in surface area. To allow starch, a water-absorbing powder, to be used as a sole food source for Aspergillus strains, a special glass bead system was applied. AFB1 production of A. flavus Zt41 was determined to be 437.6 ± 128.4 ng/g and 90.0 ± 44.8 ng/g on rice and corn starch, respectively, while corresponding ST production levels by A. creber 2663 were 72.8 ± 10.0 µg/g and 26.8 ± 11.6 µg/g, indicating 3-fivefold higher mycotoxin levels on rice starch than on corn starch as sole carbon and energy sources. KEY POINTS: • A glass bead system ensuring the flow of air when studying powders was developed. • AFB1 and ST production of A. flavus and A. creber on rice and corn starches were studied. • 3-fivefold higher mycotoxin levels on rice starch than on corn starch were detected.


Subject(s)
Oryza , Starch , Zea mays , Oryza/chemistry , Zea mays/chemistry , Starch/metabolism , Aspergillus/metabolism , Aspergillus flavus/metabolism , Aflatoxin B1/biosynthesis , Aflatoxin B1/metabolism , Sterigmatocystin/biosynthesis , Sterigmatocystin/metabolism , Microscopy, Electron, Scanning , Particle Size , Mycotoxins/metabolism , Mycotoxins/biosynthesis , Glass
12.
Food Chem ; 452: 139541, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38718457

ABSTRACT

Identifying aflatoxin-detoxifying probiotics remains a significant challenge in mitigating the risks associated with aflatoxin contamination in crops. Biological detoxification is a popular technique that reduces mycotoxin hazards and garners consumer acceptance. Through multiple rounds of screening and validation tests, Geotrichum candidum XG1 demonstrated the ability to degrade aflatoxin B1 (AFB1) by 99-100%, exceeding the capabilities of mere adsorption mechanisms. Notably, the degradation efficiency was demonstrably influenced by the presence of copper and iron ions in the liquid medium, suggesting a potential role for proteases in the degradation process. Subsequent validation experiments with red pepper revealed an 83% reduction in AFB1 levels following fermentation with G. candidum XG1. Furthermore, mass spectrometry analysis confirmed the disruption of the AFB1 furan ring structure, leading to a subsequent reduction in its toxicity. Collectively, these findings establish G. candidum XG1 as a promising candidate for effective aflatoxin degradation, with potential applications within the food industry.


Subject(s)
Aflatoxin B1 , Food Contamination , Geotrichum , Probiotics , Aflatoxin B1/metabolism , Aflatoxin B1/chemistry , Aflatoxin B1/analysis , Probiotics/metabolism , Probiotics/chemistry , Geotrichum/metabolism , Geotrichum/chemistry , Food Contamination/analysis , Fermentation , Capsicum/chemistry , Capsicum/metabolism , Capsicum/microbiology , China
13.
Food Microbiol ; 121: 104524, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38637086

ABSTRACT

Aspergillus flavus colonization on agricultural products during preharvest and postharvest results in tremendous economic losses. Inspired by the synergistic antifungal effects of essential oils, the aims of this study were to explore the mechanism of combined cinnamaldehyde and nonanal (SCAN) against A. flavus and to evaluate the antifungal activity of SCAN loading into diatomite (DM). Shriveled mycelia were observed by scanning electron microscopy, especially in the SCAN treatment group. Calcofluor white staining, transmission electron microscopy, dichloro-dihydro-fluorescein diacetate staining and the inhibition of key enzymes in tricarboxylic acid cycle indicated that the antifungal mechanism of SCAN against A. flavus was related to the cell wall damage, reactive oxygen species accumulation and energy metabolism interruption. RNA sequencing revealed that some genes involved in antioxidation were upregulated, whereas genes responsible for cell wall biosynthesis, oxidative stress, cell cycle and spore development were significantly downregulated, supporting the occurrence of cellular apoptosis. In addition, compared with the control group, conidia production in 1.5 mg/mL DM/cinnamaldehyde, DM/nonanal and DM/SCAN groups were decreased by 27.16%, 48.22% and 76.66%, respectively, and the aflatoxin B1 (AFB1) contents decreased by 2.00%, 73.02% and 84.15%, respectively. These finding suggest that DM/SCAN complex has potential uses in food preservation.


Subject(s)
Acrolein/analogs & derivatives , Aldehydes , Antifungal Agents , Aspergillus flavus , Antifungal Agents/pharmacology , Antifungal Agents/metabolism , Aflatoxin B1/metabolism , Food Preservation
14.
Microbiol Res ; 283: 127710, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593581

ABSTRACT

Aflatoxin B1 (AFB1), a highly toxic secondary metabolite produced by Aspergillus flavus, poses a severe threat to agricultural production, food safety and human health. The methylation of mRNA m6A has been identified as a regulator of both the growth and AFB1 production of A. flavus. However, its intracellular occurrence and function needs to be elucidated. Here, we identified and characterized a m6A methyltransferase, AflIme4, in A. flavus. The enzyme was localized in the cytoplasm, and knockout of AflIme4 significantly reduced the methylation modification level of mRNA. Compared with the control strains, ΔAflIme4 exhibited diminished growth, conidial formation, mycelial hydrophobicity, sclerotium yield, pathogenicity and increased sensitivity to CR, SDS, NaCl and H2O2. Notably, AFB1 production was markedly inhibited in the A. flavus ΔAflIme4 strain. RNA-Seq coupled with RT-qPCR validation showed that the transcriptional levels of genes involved in the AFB1 biosynthesis pathway including aflA, aflG, aflH, aflK, aflL, aflO, aflS, aflV and aflY were significantly upregulated. Methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) analysis demonstrated a significant increase in m6A methylation modification levels of these pathway-specific genes, concomitant with a decrease in mRNA stability. These results suggest that AflIme4 attenuates the mRNA stability of genes in AFB1 biosynthesis by enhancing their mRNA m6A methylation modification, leading to impaired AFB1 biosynthesis. Our study identifies a novel m6A methyltransferase AflIme4 and highlights it as a potential target to control A. flavus growth, development and aflatoxin pollution.


Subject(s)
Aflatoxins , Aspergillus flavus , Humans , Aspergillus flavus/genetics , Aflatoxin B1/genetics , Aflatoxin B1/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Hydrogen Peroxide/metabolism , RNA, Messenger/metabolism , Aflatoxins/genetics , Aflatoxins/metabolism
15.
Int J Food Microbiol ; 417: 110693, 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38653122

ABSTRACT

Aspergillus flavus is a fungus notorious for contaminating food and feed with aflatoxins. As a saprophytic fungus, it secretes large amounts of enzymes to access nutrients, making endoplasmic reticulum (ER) homeostasis important for protein folding and secretion. The role of HacA, a key transcription factor in the unfolded protein response pathway, remains poorly understood in A. flavus. In this study, the hacA gene in A. flavus was knockout. Results showed that the absence of hacA led to a decreased pathogenicity of the strain, as it failed to colonize intact maize kernels. This may be due to retarded vegetable growth, especially the abnormal development of swollen tips and shorter hyphal septa. Deletion of hacA also hindered conidiogenesis and sclerotial development. Notably, the mutant strain failed to produce aflatoxin B1. Moreover, compared to the wild type, the mutant strain showed increased sensitivity to ER stress inducer such as Dithiothreitol (DTT), and heat stress. It also displayed heightened sensitivity to other environmental stresses, including cell wall, osmotic, and pH stresses. Further transcriptomic analysis revealed the involvement of the hacA in numerous biological processes, including filamentous growth, asexual reproduction, mycotoxin biosynthetic process, signal transduction, budding cell apical bud growth, invasive filamentous growth, response to stimulus, and so on. Taken together, HacA plays a vital role in fungal development, pathogenicity and aflatoxins biosynthesis. This highlights the potential of targeting hacA as a novel approach for early prevention of A. flavus contamination.


Subject(s)
Aflatoxins , Aspergillus flavus , Fungal Proteins , Gene Expression Regulation, Fungal , Transcription Factors , Unfolded Protein Response , Zea mays , Aspergillus flavus/genetics , Aspergillus flavus/pathogenicity , Aspergillus flavus/metabolism , Aspergillus flavus/growth & development , Fungal Proteins/genetics , Fungal Proteins/metabolism , Aflatoxins/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Zea mays/microbiology , Virulence , Aflatoxin B1/biosynthesis , Aflatoxin B1/metabolism , Endoplasmic Reticulum Stress
16.
Food Chem ; 449: 139240, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38599109

ABSTRACT

The study reports the efficacy of nanofabricated citronellal inside the chitosan biopolymer (NeCn) against Aspergillus flavus growth, aflatoxin B1 (AFB1) production, and active ingredient biodeterioration (Piperine) in Piper longum L. The prepared NeCn was characterized by Scanning Electron Microscopy (SEM), Dynamic Light Scattering (DLS), and Fourier Transform Infrared Spectroscopy (FTIR). The results revealed that the NeCn exhibited distantly improved antifungal (1.25 µL/mL) and AFB1 inhibition (1.0 µL/mL) compared to free Cn. The perturbances in membrane function, mitochondrial membrane potential, antioxidant defense system, and regulatory genes (Ver-1 and Nor-1) of AFB1 biosynthesis were reported as probable modes of action of NeCn. The NeCn (1.25 µL/mL) effectively protects the P. longum from A. flavus (78.8%), AFB1 contamination (100%), and deterioration of Piperine (62.39%), thus demonstrating its potential as a promising novel antifungal agent for food preservation.


Subject(s)
Acyclic Monoterpenes , Aflatoxin B1 , Aspergillus flavus , Chitosan , Piper , Aflatoxin B1/metabolism , Aspergillus flavus/drug effects , Aspergillus flavus/growth & development , Aspergillus flavus/metabolism , Chitosan/chemistry , Chitosan/pharmacology , Piper/chemistry , Biopolymers/chemistry , Biopolymers/pharmacology , Acyclic Monoterpenes/pharmacology , Acyclic Monoterpenes/chemistry , Aldehydes/pharmacology , Aldehydes/chemistry , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Food Preservation/methods , Monoterpenes/pharmacology , Monoterpenes/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology
17.
J Agric Food Chem ; 72(11): 5955-5965, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38451160

ABSTRACT

The worldwide prevalence of Aflatoxin B1 (AFB1), which contaminates feedstock and food, is on the rise. AFB1 inhibits testosterone (T) biosynthesis, but the mechanism is not yet clear. By establishing in vivo and in vitro models, this study found the number of Leydig cells (LCs), T content, and the expression of T biosynthesis key enzymes were suppressed after AFB1 treatment. AFB1 exposure also increased reactive oxygen species (ROS) and promoted mitochondrial injury and mitochondrial pathway apoptosis. Moreover, the AMPK signaling pathway was activated, and using an AMPK inhibitor relieved apoptosis and the suppressed T biosynthesis key enzymes of LCs caused by AFB1 through regulating downstream p53 and Nur77. Additionally, adding ROS intervention could inhibit AMPK activation and alleviate the decreased T content caused by AFB1. In summary, AFB1 promotes the apoptosis of LCs and inhibits T biosynthesis key enzyme expression via activating the ROS/AMPK signaling pathway, which eventually leads to T synthesis disorder.


Subject(s)
AMP-Activated Protein Kinases , Aflatoxin B1 , Mice , Male , Animals , Reactive Oxygen Species/metabolism , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Signal Transduction , Testosterone , Apoptosis , Oxidative Stress
18.
BMC Vet Res ; 20(1): 108, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38500117

ABSTRACT

BACKGROUND: Camel milk and silymarin have many different beneficial effects on several animal species. Meanwhile, Aflatoxins are mycotoxins with extraordinary potency that pose major health risks to several animal species. Additionally, it has been documented that aflatoxins harm the reproductive systems of a variety of domestic animals. The present design aimed to investigate the impact of aflatoxin B1 (AFB1) on rat body weight and reproductive organs and the ameliorative effects of camel milk and silymarin through measured serum testosterone, testes pathology, and gene expression of tumor necrosis factor (TNF-α), luteinizing hormone receptor (LHR), and steroidogenic acute regulatory protein (StAR) in the testes. A total of sixty mature male Wister white rats, each weighing an average of 83.67 ± 0.21 g, were used. There were six groups created from the rats. Each division had ten rats. The groups were the control (without any treatment), CM (1 ml of camel milk/kg body weight orally), S (20 mg silymarin/kg b. wt. suspension, orally), A (1.4 mg aflatoxin/kg diet), ACM (aflatoxin plus camel milk), and AS (aflatoxin plus silymarin). RESULTS: The results indicated the positive effects of camel milk and silymarin on growth, reproductive organs, and gene expression of TNF-α, LHR, and StAR with normal testicular architecture. Also, the negative effect of AFB1 on the rat's body weight and reproductive organs, as indicated by low body weight and testosterone concentration, was confirmed by the results of histopathology and gene expression. However, these negative effects were ameliorated by the ingestion of camel milk and silymarin. CONCLUSION: In conclusion, camel milk and silymarin could mitigate the negative effect of AFB1 on rat body weight and reproductive organs.


Subject(s)
Aflatoxins , Silymarin , Male , Rats , Animals , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Silymarin/pharmacology , Camelus , Milk , Tumor Necrosis Factor-alpha/metabolism , Rats, Wistar , Testis/metabolism , Testosterone/metabolism , Body Weight
19.
J Hazard Mater ; 468: 133836, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38394902

ABSTRACT

Global aflatoxin B1 (AFB1) contamination is inevitable, and it can significantly damage testicular development. However, the current mechanism is confusing. Here, by integrating the transcriptome, microbiome, and serum metabolome, we comprehensively explain the impact of AFB1 on testis from the gut-metabolism-testis axis. Transcriptome analysis suggested that AFB1 exposure directly causes abnormalities in testicular inflammation-related signalling, such as tumor necrosis factor (TNF) pathway, and proliferation-related signalling pathways, such as phosphatidylinositide 3-kinases-protein kinase B (PI3K-AKT) pathway, which was verified by immunofluorescence. On the other hand, we found that upregulated inflammatory factors in the intestine after AFB1 exposure were associated with intestinal microbial dysbiosis, especially the enrichment of Bacilli, and enrichment analysis showed that this may be related to NLR family pyrin domain containing 3 (NLRP3)-mediated NOD-like receptor signalling. Also, AFB1 exposure caused blood metabolic disturbances, manifested as decreased hormone levels and increased oxidative stress. Significantly, B. licheniformis has remarkable AFB1 degradation efficiency (> 90%). B. licheniformis treatment is effective in attenuating gut-testis axis damage caused by AFB1 exposure through the above-mentioned signalling pathways. In conclusion, our findings indicate that AFB1 exposure disrupts testicular development through the gut-metabolism-testis axis, and B. licheniformis can effectively degrade AFB1.


Subject(s)
Bacillus licheniformis , Testis , Male , Humans , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Metabolome
20.
Chem Biol Interact ; 392: 110923, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38382706

ABSTRACT

Aflatoxin B1 (AFB1) is the most toxic mycotoxin and a proven human carcinogen that requires metabolic activation, known by cytochrome P450 (CYP) 1A2 and 3A4. Previous evidence showed that AFB1 is activated by human recombinant CYP1A1 expressed in budding yeast. Yet, the toxicity, in particular the genotoxicity of the reactive metabolites formed from AFB1 remains unclear. Humans could be exposed to both AFB1 and benzo(a)pyrene (BaP) simultaneously, thus we were interested in their combined genotoxic effects subsequent to metabolic activation by CYP1A1. In this study, molecular docking of AFB1 to human CYP1A1 indicated that AFB1 is valid as a substrate. In the incubations with AFB1 in human CYP1A1-expressed microsomes, AFM1 as a marking metabolite of AFB1 was detected. Moreover, AFB1 induced micronucleus formation in a Chinese hamster V79-derived cell line and in a human lung epithelial BEAS-2B cell line, both expressing recombinant human CYP1A1, V79-hCYP1A1 and 2B-hCYP1A1 cells, respectively. Immunofluorescence of centromere protein B stained micronuclei was dominant in AFB1-treated BEAS-2B cells exposed to AFB1, suggesting an aneugenic effect. Moreover, AFB1 elevated the levels of ROS, 8-OHdG, AFB1-DNA adduct, and DNA breaks in 2B-hCYP1A1 cells, compared with those in the parental BEAS-2B cells. Meanwhile, AFB1 increased CYP1A1, RAD51, and γ-H2AX protein levels in 2B-hCYP1A1 cells, which were attenuated by the CYP1A1 inhibitor bergamottin. Co-exposure of AFB1 with BaP increased 8-OHdG, RAD51, and γ-H2AX levels (indicating DNA damage). In conclusion, AFB1 could be activated by human CYP1A1 for potent aneugenicity, which may be further enhanced by co-exposure to BaP.


Subject(s)
Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System , Animals , Humans , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Aflatoxin B1/toxicity , Aflatoxin B1/metabolism , Benzo(a)pyrene/toxicity , Aneugens , Molecular Docking Simulation , Mammals/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL