Next Article in Journal
Role of Diagnostic Nerve Blocks in the Goal-Oriented Treatment of Spasticity with Botulinum Toxin Type A: A Case–Control Study
Previous Article in Journal
A Guide to the Clinical Management of Vipera Snakebite in Italy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification of a Novel Aflatoxin B1-Degrading Strain, Bacillus halotolerans DDC-4, and Its Response Mechanisms to Aflatoxin B1

1
College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
2
Engineering Research Center of Food Fermentation Technology, Liaoning, Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, China
3
Greens SCI. & TECH. Development Co., Ltd., Tangshan 063299, China
*
Authors to whom correspondence should be addressed.
Current address: Development and Reform Bureau of Linze County, Zhangye 734200, China.
Toxins 2024, 16(6), 256; https://doi.org/10.3390/toxins16060256
Submission received: 30 April 2024 / Revised: 24 May 2024 / Accepted: 27 May 2024 / Published: 31 May 2024
(This article belongs to the Section Mycotoxins)

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.
Key Contribution: In this study, a novel AFB1-degrading strain was identified as Bacillus halotolerans DDC-4, and the active components of this strain were thermostable extracellular proteins. Transcriptomic analysis indicated that the alpha/beta hydrolase-encoding gene might act as a novel candidate gene for AFB1 degradation, and inhibition of nucleic acid synthesis was the main toxicological effect of AFB1.

1. Introduction

Aflatoxins are a group of noxious difuran coumarin derivatives and are mainly produced by the Aspergillus species (e.g., As. flavus and As. parasiticus) [1]. They primarily spread through contamination of various foodstuffs (e.g., nuts, corn, and oil by-products) during crop growth, harvest, and storage [2,3,4]. Moreover, this toxin can barely be degraded naturally because of its high stability. Thus, approximately five billion people are at the risk of chronic exposure to aflatoxin worldwide [5]. Among the identified aflatoxins, aflatoxin B1 (AFB1) is regarded the most toxic, carcinogenic, and mutagenic because of the C8-C9 double bond of the difuran ring and the lactone ring within the coumarin ring [6].
In the last decade, several physical, chemical, and biological approaches have been reported for AFB1 degradation [5]. Compared with other methods, biodegradation is the most promising alternative because of its high specificity, eco-friendliness, and harmlessness to nutritional and organoleptic properties of food [6]. Until now, many AFB1-degrading strains have been identified, such as Bacillus subtilis UTBSP1 [7], Pseudomonas putida [8], Mycobacterium smegmatis mc2 155 [9], Rhodococcus pyridinivorans [10], As. niger FS10 [11], Zygosaccharomyces rouxii [12], Armillariella tabescens [13], and Trametes versicolor [14]. However, the response mechanisms related to AFB1 toxicity, degradation, and adaptation in degrading strains remain unknown.
Degrading strains chiefly mediate AFB1 degradation by producing enzymes that convert this toxin into less toxic or nontoxic metabolites. Most of the reported degrading enzymes are oxidoreductases, including oxidase (e.g., aflatoxin oxidase enzyme, AFO [13] and laccases [15]), peroxidase (e.g., manganese peroxidase, MnP) [16], and reductases (e.g., F420/H2-dependent reductases) [9]. The degradation mechanisms of oxidase and peroxidase to AFB1 are mainly oxidation and hydroxylation reactions. The major chemically active location for these reactions is the difuran ring due to the presence of a double bond in conjugation with an oxygen atom [17]. AFO from Ar. tabescens and MnPs from the white-rot fungus such as Phanerochaete sordida YK-624 could oxidize the furan ring of AFB1 to 8,9-epoxide formation, further forming 8,9-dihydrodiol through hydrolysis [16,18]. A conversion of AFB1 to AFQ1 is also a common degradation pathway, which was found in laccase of Lac2 from the white-rot fungus [19], CotA laccase from B. licheniformis [20], and dye-decolorizing peroxidase type B [21]. The major targets of reductases are unsaturations in furan and lactone rings and the α-β unsaturated carbonyl group [17]. F420/H2-dependent reductases identified from M. smegmatis could reduce α,β-unsaturated esters of AFB1 [9].
As determined by the structures of AFB1 and degradation products, hydrolysis, demethylation, demethoxylation, and decarbonylation reactions are also involved in the degradation mechanisms [17]. In many AFB1-degrading strains, hydrolysis of the lactone ring has been reported as a starting point [17]. After hydrolysis, the presence of the α-β unsaturated in the product increases its chemical activity, leading to a series of degradation reactions, including decarboxylation and the cleavage of the cyclopentenone ring, which convert AFB1 to AFD1 and further to AFD2 [8]. Moreover, some demethylated, demethoxylated, and decarbonylated products were also found in the biodegraded products of T. versicolor [14] and Tetragenococcus halophilus CGMCC 3792 [22]. However, the enzymes involved in the above reactions have not been identified. Therefore, more AFB1-degrading enzymes, especially of new types, need to be identified. This will contribute to efficient AFB1 degradation through genetic engineering methods.
With the advancements of high-throughput sequencing and bioinformatics, omics technologies offer a new in-depth insight into the response mechanism, which will help identify more genes encoding degrading enzymes [23]. Xu et al. identified a gene encoding the novel zearalenone degradation-associated thioesterase from B. amyloliquefaciens H6 through transcriptomic analysis [24]. On investigating the detoxification mechanism of R. pyridinivorans GF3 in response to thraquinone-2-sulfonate (ASA-2) through transcriptomic analysis, Wang et al. found that cytochrome P450 and short-chain dehydrogenase/reductase are involved in ASA-2 degradation [25]. By performing transcriptomic analysis, Wei et al. explored the response mechanism of Cryptococcus podzolicus Y3 under ochratoxin A stress [26]. Protein processing in C. podzolicus Y3 was inhibited by ochratoxin A, and C. podzolicus Y3 improved the excision repair pathway to protect genetic information.
In this study, 12 AFB1-degrading strains were screened from moldy maize, moldy rice, and strains stored in our laboratory (isolated from Chinese traditional fermented foods). Among them, strain DDC-4 exhibited the highest degradation activity and was identified as B. halotolerans through physiological, biochemical, and molecular methods. The active component and its characteristics were explored, and transcriptomic analysis was performed to explore the response mechanisms of strain DDC-4 to AFB1. Several candidate AFB1-degrading genes, especially the previously ignored alpha/beta hydrolase (arylesterase) gene, were mined.

2. Results

2.1. Isolation and Identification of AFB1-Degrading Strains

The modified Hormisch medium, containing coumarin as the sole carbon source, was used for obtaining potential AFB1-degrading strains [27]. Coumarin is the basic molecular structure of aflatoxin B1 with lower price and more secure. The strains that could grow on modified Hormisch medium have the ability to utilize coumarin as their carbon source indicating they might also be able to degrade aflatoxin B1 [28]. In total, 12 strains were isolated from various sources, namely 7 strains from moldy rice (i.e., ZYX1–ZYX7, respectively), 3 strains from moldy maize (i.e., DC-1, DC-3, and DC-5, respectively), and 2 strains from among those stored in our lab (i.e., DDC-1 and DDC-4, respectively). Of the strains isolated, strain DDC-4 exhibited the highest AFB1 degradation rate of 76.30% ± 2.18% after 72 h of incubation at 37 °C, which was significantly higher than other strains (Figure 1).
During the physiological and biochemical tests, strain DDC-4, a Gram-positive rod bacterium, displayed the typical characteristics of Bacillus species (Table 1). The strain could use glucose, arabinose, xylose, mannitol, gelatin, starch, casein, citrate, and malonate and reduce nitrate and grow at pH 5.7, but it could not grow in the presence of lysozymes. Moreover, it exhibited certain temperature adaptability (30 °C–50 °C) and salt resistance (up to 10% (w/v) NaCl). Additionally, strain DDC-4 exhibited catalase activity but no phenylalanine dehydrolase or tryptophanase activity. Meanwhile, an approximately 1500 bp product was amplified from the genomic DNA of DDC-4, and a neighbor-joining tree was constructed based on the results of the 16S rRNA gene sequence analysis (Figure 2). Compared with the outgroup Metabacillus galliciensis, Bacillus species were grouped together in a single cluster. Strain DDC-4 and B. halotolerans ATCC 25096T were clustered into the same clade with 100% sequence similarity. Thus, strain DDC-4 was recognized as B. halotolerans. To the best of our knowledge, this study is the first to report B. halotolerans as an AFB1-degrading strain.

2.2. AFB1 Degradation by the Active Component of Strain DDC-4 and Its Characteristics

We here investigated whether the fermentation broth, cell-free supernatant, cell suspension, and cell lysate can cause AFB1 degradation (Figure 3A). Overall, the cell-free supernatant removed 55.04% ± 2.60% of AFB1 after 72 h incubation, whereas the cell suspension and cell lysate were almost unable to remove AFB1, with their degradation rates being −1.88% ± 8.46% and 4.44% ± 0.52%, respectively, dramatically lower than that of the cell-free supernatant. This indicated that AFB1 removal by strain DDC-4 predominantly depended on degradation rather than on absorption. The cell-free supernatant was the main active component of strain DDC-4 during degradation. These findings are consistent with those of Bacillus species (e.g., B. licheniformis CFR1, B. subtilis UTBSP1, B. velezensis DY3108, and B. amyloliquefaciens WF2020) [7,29,30,31]. Although the cell-free supernatant had a major role in degradation, the degradation rate with the supernatant was significantly lower than that with the fermentation broth. Therefore, AFB1 could be speculated to exert an induction effect on the degradation activity of strain DDC-4. In other words, the expression level of genes encoding degradation-associated extracellular metabolites might be augmented under AFB1 stress to reduce AFB1-induced damage. To verify this hypothesis, we evaluated the induction effect of AFB1. The degradation rate of the cell-free supernatant increased to 68.08% ± 4.11% after induction. This rate was significantly higher than that of the noninduction group (50.49% ± 8.26%) and was almost the same as that of the fermentation broth (Figure 3B).
The degradation rate of the cell-free supernatant dramatically decreased to 10.60% ± 8.61% and 39.37% ± 1.18% after SDS and proteinase K pretreatments, respectively (Figure 4A). This might be because the structure of the protein in the supernatant was destroyed. By contrast, the degradation rate exhibited no decrease but increased slightly after heat treatment. The degradation rate increased more significantly when the heat treatment was prolonged. We further investigated the effect of incubation temperature on degradation activity. Similarly, the degradation rate significantly increased at 30 °C–60 °C, from 18.51% ± 1.34% to 79.24% ± 3.67%, and remained stable (approximately 80%) at 60 °C–90 °C (Figure 4B). According to these results, thermostable proteins or perhaps enzymes with a broad temperature adaptability present in the supernatant were involved in AFB1 degradation, and their activities were activated by heat treatment. Similar results have been observed in some AFB1-degrading strains, such as B. velezensis DY3108 [30], B. shackletonii L7 [32], and P. aeruginosa N17-1 [33], which have made them more advantageous in industrial applications.
Considering the enzyme activity loss during freeze–drying, although the cell-free supernatant was pH sensitive, it could still degrade AFB1 within 5–10 pH (Figure 4C). The maximum degradation rate of 45.11% ± 1.99% was observed at pH 7, while decreased significantly as the pH increased or decreased because of the impaired activity of the enzymes in the supernatant. Compared to acid, the supernatant had a stronger tolerance to alkalis. The degradation rate decreased to 30.63% ± 2.75% at pH 8 and decreased to 22.07% ± 4.22% at pH 6. The degradation rates at pH 5, pH6, pH 9 and pH 10 were not significantly different. Moreover, the activity of the cell-free supernatant was almost lost at pH 4 and pH 11, with their degradation rates being 1.63% ± 0.09% and 6.33% ± 1.92%, respectively, which were significantly lower than others.
Additionally, the effects of metal ions on degradation by the cell-free supernatant were evaluated (Figure 4D). Cu2+ dramatically enhanced the degradation rate to 95.88% ± 1.51%, whereas Zn2+ and Fe3+ exerted no significant effect. However, Li+, Ni+, Mg2+, and Ca2+ inhibited the degradation activity to a certain extent. Furthermore, the influence of the copper concentration revealed that the degradation rate increased sharply within the range of 0–10 mM Cu2+ and decreased slightly afterward (Figure 4E). Thus, Cu2+ might act as an activator or membrane stabilizer or an electron transfer medium for enzymes to stimulate AFB1 degradation activity, comparatively similar to the results obtained in other AFB1-degrading strains, including B. amyloliquefaciens WF2020 [31], B. velezensis DY3108 [30], and B. shackletonii L7 [32]. When 10 mM Cu2+ was added, 41.56% ± 2.02% AFB1 was degraded in the initial 6 h and 95.45% ± 1.81% AFB1 was degraded after 48 h incubation (Figure 4F). This indicated that the supernatant caused relatively rapid degradation. Moreover, the supernatant decreased the AFB1 content in the moldy maize powder from 6.39 ± 0.43 μg/kg to 2.96 ± 0.92 μg/kg (the degradation rate was 53.77% ± 14.42%, Table 2), which demonstrates that the cell-free supernatant of strain DDC-4 can be a potential tool for handling moldy grains.
Overall, the active components of strain DDC-4 were thermostable extracellular proteins. AFB1 induced the expression of genes encoding these proteins, and Cu2+ and heat treatment increased the activity of these proteins.

2.3. GO Term and KEGG Pathway Enrichment Analyses

To reveal the molecular response of DDC-4 to AFB1, transcriptomic analysis was performed. Q20 and Q30 values for each sample were greater than 98% and 95%, respectively (Table S1). More than 95% of the clean reads were mapped to the reference genome (Table S2), which indicated the reliability of the RNA sequencing results. The distance between the treated and untreated samples was significant (Figure S1). In total, 165 upregulated and 284 downregulated differentially expressed genes (DEGs; Figure S2), mapped to 27 and 32 GO terms (Figure S3), respectively, were identified after AFB1 treatment.
The upregulated DEGs were significantly enriched in 10 GO terms, including the histidine catabolic process, the histidine catabolic process to glutamate and formamide, the histidine catabolic process to glutamate and formate, and developmental process (Figure 5A). All the top three GO terms were related to the histidine catabolic process, with all the rich factors (the ratio of the enriched DEGs to total transcripts) being 1.00. Similar results were observed in the KEGG pathway enrichment analysis. The upregulated DEGs were significantly enriched in the histidine metabolism pathway (Figure 6A). Following AFB1 treatment, the expression of genes encoding histidine ammonia-lyase (EC: 4.3.1.3, encoded by the gene RS11215, HutH), urocanate hydratase (EC: 4.2.1.49, encoded by the gene RS11210, HutU), imidazolonepropionase (EC: 3.5.2.7, encoded by the gene RS11205), formimidoylglutamase (EC: 3.5.3.8, encoded by the gene RS11200), and aldehyde dehydrogenase DhaS (EC: 1.2.1.3, encoded by the gene RS20930, DhaS) was significantly upregulated to varying degrees (Figure 7 and Figure 8A). Among them, gene RS20930 was the most highly expressed, and the transcripts per million (TPM) values in the samples untreated and treated with AFB1 were 5215 and 11,948, respectively. The expression level of gene RS11215 showed the most significant difference between untreated and treated samples with the log2 fold change being 2.11. The induced histidine metabolism-related genes promoted the conversion of histidine to glutamate, a precursor for glutathione synthesis. Glutathione possibly participates in AFB1 degradation by binding to AFB1 or intermediate products, which is consistent with the results of Qiu et al. [34]. Furthermore, the number of DEGs enriched in the developmental process was the highest, as determined through the GO term enrichment analysis (Figure 5A). DEGs in this process were predominantly related to sporulation (Table S3), possibly because sporulation in strain DDC-4 was promoted under AFB1-induced stress.
The downregulated DEGs were significantly enriched in 22 GO terms, including de novo IMP biosynthesis, IMP biosynthesis, IMP metabolism, purine nucleobase biosynthesis, purine nucleoside monophosphate biosynthesis, purine ribonucleoside monophosphate biosynthesis, and purine-containing compound biosynthesis (Figure 5B). The top three GO terms were all related to the IMP metabolic process, with all the rich factors being >0.75. Nearly all genes associated with the de novo IMP biosynthesis process (including genes RS06555, RS06560, RS06565, RS06570, RS06575, RS06580, RS06585, RS06590, RS06595, RS06600, RS06605, and RS06610) were inhibited to varying degrees (Figure 8B and Table 3). Among them, the expression of gene RS06585, RS06610, RS06590, RS06595, and RS06605 were dramatically inhibited by AFB1, with the log2 fold change being −2.63, −2.53, −2.39, −2.36, and −2.28, respectively. IMP serves as a precursor of AMP and GMP during de novo purine nucleobase biosynthesis. Although de novo pyrimidine nucleobase biosynthesis was not significantly inhibited, the expression of genes encoding the enzyme (carbamoyl phosphate synthase, encoded by the genes RS03965 and RS03960) involved in the first-step reaction of this process was dramatically downregulated, with the log2 fold change of the expression level being approximately −5. The expression of genes encoding hypoxanthine/guanine permease (encoded by the gene RS06640, PbuG) and uracil permease (encoded by the gene RS01625, PyrP), which might transport raw materials for the salvage pathway, was also downregulated. Similarly, the top three KEGG pathways with the highest number of enriched DEGs were purine metabolism, the two-component system, and ABC transporters, respectively (Figure 6B). These results indicated that AFB1 significantly inhibited nucleotide synthesis in strain DDC-4.

2.4. Identification and Expression Analysis of Potential Degrading Genes

The reported AFB1-degrading enzymes were primarily oxidoreductases. Meanwhile, hydrolase may be involved in AFB1 degradation from the degradation product perspective. According to our results, eight genes encoding oxidoreductases and six genes encoding hydrolases were induced following AFB1 treatment (Table 4). Among these genes, the gene RS11000 (aldo/keto reductase-encoding gene) was the most highly expressed (Figure 8C,D), and the TPM values in the samples untreated and treated with AFB1 were 305 and 930, respectively. Aldo/keto reductase, short-chain dehydrogenase/reductases (SDR) family oxidoreductase, and alpha/beta hydrolase (arylesterase), encoded by the genes RS11000, RS07845, and RS04140, respectively, possibly destroyed the lactone ring within the coumarin ring of AFB1 to decrease its toxicity and mutagenicity [35]. Moreover, other oxidoreductases and hydrolases might be involved in AFB1 degradation (Table 4), but their potential action sites need to be further investigated.
Genes RS11000, RS07845 and RS04140 were selected for the qRT-PCR analysis (Figure 9). The expression trend of these genes was consistent with the RNA-seq results, which confirmed the credibility of the transcriptomic analysis results.

3. Discussion

3.1. AFB1-Degrading Strains and Functional Genes

Many AFB1-degrading strains have been identified. Of them, the B. subtilis group was more sought after by researchers because of its potential probiotic characteristics and antibacterial action against Aspergillus species [30,31]. To our best knowledge, although several strains, including B. subtilis UTBSP1 [7], B. licheniformis CFR1 [29], B. shackletonii L7 [32], B. velezensis DY3108 [30], B. amyloliquefaciens WF2020 [31], and B. albus YUN5 [1], degrade aflatoxins, this study is the first to identify B. halotolerans to degrade AFB1. Consistent with the results of most reports about the B. subtilis group, the extracellular proteins of strain DDC-4 were chiefly responsible for AFB1 degradation. When activated with 10 mM Cu2+, 95.45% AFB1 (initial concentration: 1 μg/mL) was degraded by the extracellular proteins at 48 h, which was comparable to the results obtained with B. velezensis DY3108 (initial concentration: 0.5 μg/mL, >90%, 24 h) [30], B. amyloliquefaciens WF2020 (initial concentration: 2 μg/mL, ~100%, 48 h) [31], B. licheniformis CFR1 (initial concentration: 0.5 μg/mL, >90%, 24 h) [29], and B. subtilis UTBSP1 (initial concentration: 2.5 μg/mL, 78.39%, 72 h) [7] and higher than those obtained with B. shackletonii L7 [32] and B. subtilis JSW-1 [36]. Additionally, different from the heat-labile proteins of B. licheniformis CFR1 [29] and B. subtilis UTBSP1 [7], the active extracellular proteins of strain DDC-4 were thermostable. Furthermore, the degradation rate remained at approximately 80% at 90 °C, which was higher than those of B. amyloliquefaciens WF2020 [31] and B. shackletonii L7 [32], but slightly lower than that of B. velezensis DY3108 [30]. This facilitated the proteins in maintaining catalytic stability in a harsh industrial environment. The active extracellular proteins could remove 53.77% AFB1 from the moldy maize powder and is thus a promising agent for handling AFB1-contaminated food in the industry.
Although few proteins with a degradation ability have been isolated from the B. subtilis group, the response mechanism of this group to aflatoxins has not been completely reported. A 22-kDa heat-stable unidentified extracellular protein was purified from the cell-free supernatant of B. shackletonii L7 [32]. CotA laccase from B. licheniformis ANSB82 could transform AFB1 to aflatoxin Q1 and epi-aflatoxin Q1 [20]. Bacilysin biosynthesis oxidoreductase (BacC) from B. subtilis UTB1 was involved in AFB1 degradation by reducing the α,β-unsaturated ester between the lactone rings of AFB1 [37]. However, mass spectrometry of degradation products revealed that the difuran and lactone rings of AFB1 were all destroyed. Six and eight major degraded products were identified in the reaction mixture of AFB1 coincubated with B. albus YUN5 [1] and B. subtilis [14], respectively. Four major degraded products were detected in the B. sp. H16v8 and B. sp. HGD9229 cocultures [38]. This suggests that in addition to oxidoreductase, other types of enzymes, particularly esterase, are involved in AFB1 degradation. According to Pereyra et al., N-acyl-homoserine lactonase might contribute to AFB1 degradation [35]. However, not all AFB1-degrading strains of the B. subtilis group could produce this enzyme. In the present study, the transcriptomic analysis was performed to identify the previously neglected gene-encoding alpha/beta hydrolase (arylesterase) as the candidate gene for AFB1 degradation. This study provides a novel insight about AFB1-degrading enzymes. Alpha/beta hydrolase is a class of enzymes having similar structures and diverse functions, including esterase, lipase, proteases, and other hydrolytic enzymes [39]. Among the enzymes, arylesterase possibly targets the ester bond of AFB1 and thus cleaves its lactone ring to reduce its toxicity and mutagenicity.
The gene RS11000 encodes for aldo/keto reductase, which might destroy the lactone ring in AFB1 by reducing the keto group to the OH group. The gene RS07845 encodes for the SDR family oxidoreductase that has a broad substrate specificity. After cloning the CgSDR gene from Candida guilliermondii, Xing et al. found that recombinase transformed patulin into non-toxic E-ascladiol [40]. Thus, the SDR family oxidoreductase in this study was speculated to cleave the lactone ring in AFB1 following the reduction reaction catalyzed by aldo/keto reductase. Similar to the results of Xu et al., Cu2+ possibly serves as an electron transfer medium in redox reactions that boosts degradation activity [32]. Furthermore, all the aforementioned proteins of strain DDC-4 belonged to the general stress protein, which could confer advantages to bacteria under stress, such as salt, osmosis, oxidative damage, and freezing [41]. In this study, the expression of genes RS11000 and RS07845 was significantly upregulated under AFB1 stress, which might allow the strain to survive in the presence of the toxicological effects of AFB1 because these genes are associated with AFB1 degradation.
Glutathione exerted its detoxification effect on AFB1 by binding to it or its intermediate products, and this was first observed in mammals. In a reaction mixture of AFB1 coincubated with A. niger FS10, Qiu et al. analyzed AFB1 degradation products through triple quadrupole-linear ion trap-mass spectrometry (Q-Trap-MS) coupled with LightSight™ software (Version 2.2.1) [34]. They found that glutathione formed AFB2-GOH (C27H31N3O13) with AFB1 to modify the toxicity site of AFB1. As mentioned above, glutathione might participate in the AFB1 degradation of strain DDC-4. As glutamate is a precursor for glutathione synthesis, the conversion of histidine to glutamate was promoted in the AFB1-treated samples (Figure 10).
Several potential mycotoxins degradation genes were also selected by transcriptomic analysis due to their upregulated expression in the present of mycotoxins, such as short-chain aryl-alcohol dehydrogenase for patulin degradation [42] and carboxypeptidase A4 for ochratoxin A degradation [26]. However, the specific function of these genes in mycotoxins degradation still needs to be validated by heterologous expression. The degradation mechanism will be revealed by analysis of degradation products of recombinant protein expressed in host strain. The encoding gene of an acyl coenzyme A thioester hydrolase in B. amyloliquefaciens H6 was selected from upregulated genes under zearalenone stress by transcriptomic analysis [24]. The recombinant protein was expressed in Escherichia coli. The purified recombinant protein could convert zearalenone to the less toxic metabolites by cleaving the lactone bond and breaking down its macrolide ring [24]. More experiments will be carried out in our future study.

3.2. Toxicological Effect of AFB1 on Nucleic Acid Synthesis

On measuring the incorporation of me-[3H] thymidine and 6-[14C] orotic acid into DNA and RNA, respectively, Butler and Neal found that AFB1 inhibited nucleic acid synthesis [43]. Numerous subsequent studies have supported this viewpoint. However, the underlying molecular mechanism remains unclear. We here conjectured that AFB1 inhibited nucleic acid synthesis in strain DDC-4 through two hypothetical pathways (Figure 10). First, AFB1 inhibited nucleotide synthesis in strain DDC-4. As shown previously, AFB1 significantly inhibited de novo nucleotide biosynthesis by suppressing the expression level of genes encoding enzymes involved in this process. Moreover, the salvage pathway might be inhibited by reducing the transportation of raw materials. Second, AFB1 inhibited DNA replication in strain DDC-4. The process of DNA replication is divided into three stages: initiation, extension, and termination. At the beginning of extension, short RNA fragments (called primers), which are synthesized by primase, are acted as a starting point for DNA polymerase III. After termination, the primers are removed by ribonuclease H and DNA polymerase I (Figure S4). In this study, the expression level of genes encoding ribonuclease H, including ribonuclease HI (encoded by the gene RS20480) and ribonuclease HIII (encoded by the gene RS17285), were inhibited by AFB1; thereby, DNA replication was inhibited.
AFB1 was bioactivated by cytochrome P450 to generate the intermediate AFB1-8,9-epoxide [44]. This intermediate product was then attacked by N7 of guanine to form trans-8,9-dihydro-8-(N7-guanyl)-9-hydroxyaflatoxin B1 (AFB1-N7-Gua). This was considered as the main AFB1–DNA adduct causing mutations (Figure 10). Nucleotide excision repair (NER) is a pivotal player in removing AFB1–DNA damage in both bacterial and mammalian systems [44]. In prokaryotes, the Uvr system is involved in NER. In the present study, the expression level of the UvrD gene was upregulated after AFB1 treatment, whereas that of the UvrABC gene remained almost unchanged.
Altogether, AFB1 inhibited the synthesis of nucleotides, including purines and pyrimidines, and DNA replication (Figure 10). Cytochrome P450-mediated mutations may increase following AFB1 treatment. In the case of the resistance and adaptation to AFB1, the expression of genes encoding the potential AFB1-degrading enzyme was upregulated, and sporulation in strain DDC-4 was promoted.
Although several potential AFB1-degrading enzymes were selected, further validation of their function is needed. In the future, we will obtain the aforementioned enzymes through heterologous expression and purification. Degradation activity of the enzymes will be verified, the structures of the degradation products will be determined, and the safety of degradation products will be evaluated.

4. Conclusions

In this study, a novel AFB1-degrading strain was isolated and identified as B. halotolerans DDC-4 (belonging to the B. subtilis group). The active components of this strain were thermostable extracellular proteins or enzymes with a wide temperature adaptability. More than 90% AFB1 was degraded by the proteins or enzymes when Cu2+ was added. Thus, after adequate purification, these enzymes or proteins could serve as promising agents for AFB1 biodegradation in the food industry. To our best knowledge, this study is the first to explore response mechanisms of the B. subtilis group to AFB1 through transcriptomic analysis. Inhibition of nucleic acid synthesis was the primary toxicological effect of AFB1 on strain DDC-4. To survive under this stress, sporulation was promoted in the bacteria and the expression of genes encoding these degradation-related enzymes were induced. The genes encoding alpha/beta hydrolase (arylesterase), aldo/keto reductase, and SDR family oxidoreductase were selected as candidate genes for AFB1 degradation. Our study will be helpful to reveal the degradation mechanism of AFB1 and provide more options for handling AFB1-contaminated food.

5. Materials and Methods

5.1. Isolation of AFB1-Degrading Strains

First, 10 g moldy maize and 10 g moldy rice were separately diluted in 90 mL sterile distilled water and incubated in water bath shaker (Guangdong Foheng Instrument Co., Ltd., Dongguan, China) at 37 °C with continuous shaking (150 rpm) for 72 h. The samples were serially diluted to 10−7 with sterile distilled water. Aliquots (150 µL) of each dilution or strains stored in our lab (isolated from Chinese traditional fermented foods) were spread on plates containing modified Hormisch medium (HM: 0.1% coumarin, 0.05% KNO3, 0.05% (NH4)2SO4, 0.025% KH2PO4, 0.025% MgSO4·7H2O, 0.0005% CaCl2, 0.0003% FeCl3·6H2O, 2% agar) [27]. Each plate was cultured at 37 °C for 7 days. Visible single colonies were isolated and transferred to fresh HM plates. The aforementioned process was repeated 3–5 times until pure isolates were obtained.
To test the AFB1 degradation activity, each pure isolate was inoculated in Luria-Bertani (LB) medium, cultivated overnight at 37 °C with continuous shaking (150 rpm), and diluted to an optical density at 600 nm (OD600) of 0.4. The medium was modified to maintain neutrality during fermentation. Then, 500 μL of each dilution was added to the modified LB medium (1% peptone, 1% NaCl, 0.5% yeast extract, 0.1% KH2PO4). Fermentation was carried out at 37 °C for 48 h by shaking. Then, 960 μL of the fermentation broth was co-incubated with 40 μL of 25 μg/mL AFB1 (Yuanye Bio-Technology Co., Ltd., Shanghai, China) in the dark at 37 °C for 72 h with shaking (150 rpm). Sterile modified LB medium containing AFB1 was used as the control. The supernatant was recovered through centrifugation at 4500 rpm for 10 min at room temperature. Subsequently, 650 μL of the supernatant was mixed with 350 μL methanol, and residual AFB1 was analyzed using the ELISA kit (Youlong Biotech Co., Ltd., Shanghai, China). According to kit instruction, the cross-reactivity ration with similar toxin AFB2, AFG1, and AFG2 was 13%, 1.9%, and 5.7%, respectively, indicating the kit could specifically detect AFB1. The AFB1 degradation rate was calculated as follows:
Y = X 1 X 2 / X 1 × 100 %
where X1 is the residual AFB1 in the control, X2 is the residual AFB1 in the sample, and Y is the AFB1 degradation rate (%).

5.2. Identification of Strain DDC-4

Strain DDC-4 was identified through physiological and biochemical tests and 16S rRNA gene sequencing. The physiological and biochemical tests were conducted using the specified reagents (Haibo Biotechnology Co., Ltd., Qingdao, China). Meanwhile, genomic DNA was extracted using the E.Z.N.A Bacterial DNA Kit (Omega Bio-tek. Inc., Norcross, GA, USA). The 16S rRNA-coding gene was amplified through PCR by using the universal primer pair 16S-F and 16S-R (Table S1) [45], sequenced by General Biosystems Co., Ltd. (Chuzhou, China), aligned with sequences found on the EzBioCloud server [46], and deposited in the NCBI GenBank with accession number OQ306542. A phylogenetic tree was constructed with MEGA software (version 6.0) using the neighbor-joining method [47].

5.3. AFB1 Degradation by the Cell-Free Supernatant, Cell Suspension, and Cell Lysate

Strain DDC-4 was fermented as mentioned above. After fermentation for 48 h, 2 mL fermentation broth was centrifuged at 4500 rpm for 10 min at room temperature to separate the cell-free supernatant and cells. The cells were washed with 2 mL phosphate buffer saline (PBS: 137 mM NaCl, 2.7 mM KCl, 4.5 mM Na2HPO4, 1.4 mM KH2PO4) twice and resuspended in 2 mL PBS. Then, the solution was divided into two fractions. One fraction was processed without any treatment (namely, cell suspension). The other fraction was disintegrated through ultrasonication (Sonics, Newtown, Connecticut, USA, 50% of maximum amplitude, subjected to ultrasound for 3 min with a 5 s interval between two 3 s processing) in the ice bath and centrifuged at 10,000 rpm for 2 min at 4 °C to obtain the supernatant (namely, cell lysate). The obtained cell-free supernatant and cell lysate were separately filtered through a 0.45 µm filter. Then, 960 μL each of the fermentation broth, cell-free supernatant, cell suspension, and cell lysate was separately coincubated with 40 μL of 25 μg/mL AFB1. The sterile modified LB medium or PBS containing AFB1 served as the control. Residual AFB1 in each sample was determined as described previously. The AFB1 degradation rate was calculated using the aforementioned formula.

5.4. Induction Effect of Degradation by AFB1

Strain DDC-4 was fermented as mentioned above. The fermentation broth was divided into four fractions, namely fraction A, fraction B, fraction C, and fraction D. To investigate the induction effect of AFB1 on degradation by comparing the degradation rate of the cell-free supernatant induced by AFB1, the cell-free supernatant uninduced by AFB1, and the fermentation broth; the AFB1 addition concentration and incubation time were the same as those used while determining the degradation rates of the fermentation broth.
Fraction A (induced by AFB1): 960 μL of the fermentation broth was treated with 40 μL of 25 μg/mL AFB1 in the dark at 37 °C for 72 h with shaking (150 rpm). The supernatant was collected through centrifugation at 4500 rpm for 10 min at room temperature, filtered through a 0.45 µm filter, and coincubated with 40 μL of 25 μg/mL AFB1 in the dark at 37 °C for 72 h with shaking (150 rpm).
Fraction B (without induction): 960 μL of the fermentation broth was treated with 40 μL sterile distilled water in the dark at 37 °C for 72 h with shaking (150 rpm). The supernatant was treated in the same manner as fraction A.
Fraction C: 960 μL of the fermentation broth was treated with 40 μL of 25 μg/mL AFB1 in the dark at 37 °C for 72 h with shaking (150 rpm).
Fraction D: 960 μL of the sterile modified LB medium was coincubated with 40 μL of 25 μg/mL AFB1 in the dark at 37 °C for 72 h with shaking (150 rpm).
Residual AFB1 in each fraction was determined as described above. The AFB1 degradation rate was calculated as follows:
Y i = C + D A / C + D × 100 %
Y u = D B / D × 100 %
where A is the residual AFB1 in Fraction A, B is the residual AFB1 in Fraction B, C is the residual AFB1 in Fraction C, D is the residual AFB1 in Fraction D, Yi is the AFB1 degradation rate of the induction group (%), and Yu is the AFB1 degradation rate of the noninduction group (%).

5.5. Effects of Heat, SDS, and Proteinase K Treatments on AFB1 Degradation by the Cell-Free Supernatant

The cell-free supernatant was prepared as mentioned above. To investigate the effects of heat, SDS, and proteinase K treatments, the cell-free supernatant was treated with boiling water for 10 and 30 min, 1% SDS in the dark for 24 h, and 1 mg/mL proteinase K in the dark for 24 h, respectively. The degradation experiment was conducted as mentioned above. The sterile modified LB medium containing AFB1 was used as the control.

5.6. Effects of Incubation Conditions on AFB1 Degradation by the Cell-Free Supernatant

The cell-free supernatant was prepared as mentioned above. To demonstrate the effects of temperature, the supernatant containing AFB1 was incubated at different temperatures (30 °C, 40 °C, 50 °C, 60 °C, 70 °C, 80 °C, and 90 °C) without shaking in the dark for 72 h. In the pH test, the supernatant was freeze-dried, redissolved in an equal volume of different buffers (citrate buffer (pH 4 and 5), phosphate buffer (pH 6, 7 and 8), and sodium carbonate/sodium bicarbonate buffer (pH 9, 10, and 11)), and coincubated with AFB1 in the dark at 37 °C for 72 h with shaking. Regarding metal ions, the supernatant was added to 10 mM each of Li+ (LiCl), Ni2+ (NiSO4), Cu2+ (CuSO4), Mg2+ (MgCl2), Ca2+ (CaCl2), Zn2+ (ZnSO4), Mn2+ (MnCl2), and Fe3+ (FeCl3) and coincubated with AFB1 in the dark at 37 °C for 72 h with shaking. The influence of the copper concentration (1, 5, 10, and 15 mM) and that of incubation times (6, 12, 18, 24 h, 36, 48, and 72 h) with 10 mM Cu2+ on AFB1 degradation were also determined. The residual AFB1 in each sample was determined as mentioned above, and the sterile modified LB medium substituted the supernatant in the control.

5.7. Application of the Cell-Free Supernatant to Remove AFB1 from the Moldy Maize Powder

After the moldy maize powder was sterilized, 5 g of the powder was mixed with 10 mL cell-free supernatant of strain DDC-4 and 10 mM Cu2+ and incubated for 48 h. The AFB1 content was analyzed using the ELISA kit.

5.8. RNA Extraction and Sequencing

First, 960 μL of the fermentation broth was treated separately with 40 μL of 25 μg/mL AFB1 and 40 μL sterile distilled water in the dark at 37 °C for 72 h with shaking. Three independent biological replicates were used for each treatment. The cells were obtained through centrifugation at 5000× g for 10 min. Total RNA was extracted using the Total RNA Extractor Kit (Sangon Biotech Co., Ltd., Shanghai, China). RNA quality and integrity was detected through 1% agarose gel electrophoresis, and the RNA concentration was determined using the NanoDrop (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The concentration and quality of RNA met the requirements for libraries construction (Table S5). rRNAs were removed using the Ribo-off rRNA Depletion kit (Vazyme Biotech Co., Ltd., Nanjing, China). cDNA libraries were constructed using the VAHTS™ Stranded mRNA-seq Library Prep Kit for Illumina® (Vazyme Biotech Co., Ltd., China). Library quality was examined through 8% polyacrylamide gel electrophoresis. The libraries were sequenced on the DNBseq-T7 (BGI Genomics Co., Ltd., Shenzhen, China) platform (Sangon Biotech Co., Ltd., China) to obtain raw reads. The clean reads were acquired using the Trimmomatic program (version 0.36) for data processing. After the reads were evaluated for quality, the clean reads were mapped to the reference genome of B. halotolerans ZB201702 from the NCBI database (https://www.ncbi.nlm.nih.gov/assembly/GCF_004006435.1/?shouldredirect=false, accessed on 9 January 2019) using the Bowtie2 program (version 2.3.2). The transcriptome sequencing data were stored in the Sequence Read Archive (https://dataview.ncbi.nlm.nih.gov/object/PRJNA917813?reviewer=jk3r3v6iq68bb7bch1o9esdkn9, created on 4 January 2023).

5.9. GO Term and KEGG Pathway Enrichment Analyses

Heatmaps were constructed using the gplots package in R to present the distance between the samples. Transcripts per million (TPM) values were calculated using the featureCounts program (version 1.6.0) to reflect the gene expression level. Differentially expressed genes (DEGs) between the samples untreated and treated with AFB1 were selected using the DESeq2 (version 1.12.4) package in R while considering |log2 fold change| > 1 and q value < 0.05 as the filtering criteria. Functions of DEGs were annotated by referring to bioinformatics databases, including the Nonredundant Protein, Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and Cluster of Orthologous Groups of Proteins databases. GO term and KEGG pathway enrichment analyses were performed using topGO (version 2.24.0) and the clusterProfiler (version 3.0.5) package in R, respectively. The significance level was determined using the q value (<0.05). The expression of the selected genes was presented in the heatmaps constructed using the pheatmap package in R.

5.10. Quantitative Real-Time PCR

To validate the RNA-seq results, genes RS11000, RS07845, and RS04140 were selected as target genes and examined through quantitative real-time PCR (qRT-PCR). Primers were designed using Premier 6 (Table S1). Total RNA was transcribed into cDNA using the PrimeScript™ 1st Strand cDNA Synthesis Kit (Takara, Dalian, China). qRT-PCR was performed using SYBR® Premix Ex TaqTM (Takara, Dalian, China) on the 7500 Real-Time PCR System (ABI, Foster City, CA, USA). Relative expression levels of the target genes were normalized by the expression levels of the internal control gene (16S rRNA) and quantified using the ΔΔCt method. Three independent biological replicates were used.

5.11. Statistical Analysis

All assays were conducted in triplicate. The study results are expressed as mean ± SD and analyzed conducting Duncan’s multiple comparison test (p < 0.05) with SPSS software (version 22.0.0.0).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/toxins16060256/s1, Table S1: Statistics of the read alignments in the RNA-Seq study, Table S2: The reads mapped to the reference genome, Table S3: DEGs enriched in developmental process, Table S4: Primers used in this study, Table S5: The concentration and quality of RNA, Figure S1: Heatmap of distance between samples, Figure S2: Volcano plot of DEGs, Figure S3: GO term enrichment classification of DEGs, Figure S4: DNA replication.

Author Contributions

Conceptualization, J.G.; methodology, J.G.; formal analysis, J.G. and H.Z.; investigation, H.Z., Y.Z., X.H., and Y.L.; resources, R.W.; writing—original draft preparation, J.G.; writing—review and editing, S.L. and R.W.; visualization, J.G. and H.Z.; project administration, J.G.; funding acquisition, J.G. and S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant (LJKZ0654) from The Educational Department of Liaoning Province, a grant (22-322-3-39) from the Shenyang Bureau of Science and Technology and the Scientific Research Project of Shenyang Agricultural University (880418067).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article and Supplementary Material, further inquiries can be directed to the corresponding authors.

Conflicts of Interest

Author Hanlu Zhang was employed by the company Greens SCI. & TECH. Development Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Kumar, V.; Bahuguna, A.; Lee, J.S.; Sood, A.; Han, S.S.; Chun, H.S.; Kim, M. Degradation mechanism of aflatoxin B1 and aflatoxin G1 by salt tolerant Bacillus albus YUN5 isolated from ‘doenjang’, a traditional Korean food. Food Res. Int. 2023, 165, 112479. [Google Scholar] [CrossRef]
  2. Owolabi, I.O.; Karoonuthaisiri, N.; Elliott, C.T.; Petchkongkaew, A. A 10-year analysis of RASFF notifications for my-cotoxins in nuts. Trend in key mycotoxins and impacted countries. Food Res. Int. 2023, 172, 112915. [Google Scholar] [CrossRef]
  3. Focker, M.; van Asselt, E.D.; Berendsen, B.J.A.; van de Schans, M.G.M.; van Leeuwen, S.P.J.; Visser, S.M.; Van der Fels-Klerx, H.J. Review of food safety hazards in circular food systems in Europe. Food Res. Int. 2022, 158, 111505. [Google Scholar] [CrossRef]
  4. Ahmad, M.; Han, Z.; Kong, Q. Aflatoxin in peanuts and maize: An overview on occurrence, regulations, prevention, and control methods. World Mycotoxin J. 2023, 16, 99–114. [Google Scholar] [CrossRef]
  5. Guo, Y.P.; Zhao, L.H.; Ma, Q.G.; Ji, C. Novel strategies for degradation of aflatoxins in food and feed: A review. Food Res. Int. 2021, 140, 109878. [Google Scholar] [CrossRef] [PubMed]
  6. Kumar, V.; Bahuguna, A.; Ramalingam, S.; Dhakal, G.; Shim, J.J.; Kim, M. Recent technological advances in mechanism, toxicity, and food perspectives of enzyme-mediated aflatoxin degradation. Crit. Rev. Food Sci. 2022, 62, 5395–5412. [Google Scholar] [CrossRef]
  7. Farzaneh, M.; Shi, Z.Q.; Ghassempour, A.; Sedaghat, N.; Ahmadzadeh, M.; Mirabolfathy, M.; Javan-Nikkhah, M. Aflatoxin B1 degradation by Bacillus subtilis UTBSP1 isolated from pistachio nuts of Iran. Food Control 2012, 23, 100–106. [Google Scholar] [CrossRef]
  8. Samuel, M.S.; Sivaramakrishna, A.; Mehta, A. Degradation and detoxification of aflatoxin B1 by Pseudomonas putida. Int. Biodeter. Biodegr. 2014, 86, 202–209. [Google Scholar] [CrossRef]
  9. Taylor, M.C.; Jackson, C.J.; Tattersall, D.B.; French, N.; Peat, T.S.; Newman, J.; Briggs, L.J.; Lapalikar, G.V.; Campbell, P.M.; Scott, C.; et al. Identification and characterization of two families of F420H2-dependent reductases from Mycobacteria that catalyse aflatoxin degradation. Mol. Microbiol. 2010, 78, 561–575. [Google Scholar] [CrossRef]
  10. Prettl, Z.; Dési, E.; Lepossa, A.; Kriszt, B.; Kukolya, J.; Nagy, E. Biological degradation of aflatoxin B1 by a Rhodococcus pyridinivorans strain in by-product of bioethanol. Anim. Feed. Sci. Tech. 2017, 224, 104–114. [Google Scholar] [CrossRef]
  11. Xu, D.; Wang, H.X.; Zhang, Y.Z.; Yang, Z.D.; Sun, X.L. Inhibition of non-toxigenic Aspergillus niger FS10 isolated from Chinese fermented soybean on growth and aflatoxin B1 production by Aspergillus flavus. Food Control 2013, 32, 359–365. [Google Scholar] [CrossRef]
  12. Zhou, G.H.; Chen, Y.J.; Kong, Q.; Ma, Y.X.; Liu, Y. Detoxification of aflatoxin B1 by Zygosaccharomyces rouxii with solid state fermentation in peanut meal. Toxins 2017, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  13. Xu, T.T.; Xie, C.F.; Yao, D.S.; Zhou, C.Z.; Liu, J.S. Crystal structures of aflatoxin-oxidase from Armillariella tabescens reveal a dual activity enzyme. Biochem. Bioph Res. Commun. 2017, 494, 621–625. [Google Scholar] [CrossRef] [PubMed]
  14. Suresh, G.; Cabezudo, I.; Pulicharla, R.; Cuprys, A.; Rouissi, T.; Brar, S.K. Biodegradation of aflatoxin B1 with cell-free extracts of Trametes versicolor and Bacillus subtilis. Res. Vet. Sci. 2020, 133, 85–91. [Google Scholar] [CrossRef] [PubMed]
  15. Alberts, J.F.; Gelderblom, W.C.A.; Botha, A.; van Zyl, W.H. Degradation of aflatoxin B1 by fungal laccase enzymes. Int. J. Food Microbiol. 2009, 135, 47–52. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, J.Q.; Ogata, M.; Hirai, H.; Kawagishi, H. Detoxification of aflatoxin B1 by manganese peroxidase from the white-rot fungus Phanerochaete sordida YK-624. FEMS Microbiol. Lett. 2011, 314, 164–169. [Google Scholar] [CrossRef] [PubMed]
  17. Ranjith, A. Metabolites and degradation pathways of microbial detoxification of aflatoxins: A review. Mycotoxin Res. 2024, 40, 71–83. [Google Scholar] [CrossRef]
  18. Cao, H.; Liu, D.L.; Mo, X.M.; Xie, C.F.; Yao, D.S. A fungal enzyme with the ability of aflatoxin B1 conversion: Purification and ESI-MS/MS identification. Microbiol. Res. 2011, 166, 475–483. [Google Scholar] [CrossRef] [PubMed]
  19. Zhou, Z.; Li, R.; Ng, T.B.; Lai, Y.; Yang, J.; Ye, X. A new laccase of Lac 2 from the white rot fungus Cerrena unicolor 6884 and Lac 2-mediated degradation of aflatoxin B1. Toxins 2020, 12, 476. [Google Scholar] [CrossRef]
  20. Guo, Y.P.; Qin, X.J.; Tang, Y.; Ma, Q.G.; Zhang, J.Y.; Zhao, L.H. CotA laccase, a novel aflatoxin oxidase from Bacillus licheniformis, transforms aflatoxin B1 to aflatoxin Q1 and epi-aflatoxin Q1. Food Chem. 2020, 325, 126877. [Google Scholar] [CrossRef]
  21. Loi, M.; Renaud, J.B.; Rosini, E.; Pollegioni, L.; Vignali, E.; Haidukowski, M.; Sumarah, M.W.; Logrieco, A.F.; Mulè, G. Enzymatic transformation of aflatoxin B1 by Rh_DypB peroxidase and characterization of the reaction products. Chemosphere 2020, 250, 126296. [Google Scholar] [CrossRef] [PubMed]
  22. Li, J.; Huang, J.; Jin, Y.; Wu, C.; Shen, D.; Zhang, S.; Zhou, R. Aflatoxin B1 degradation by salt tolerant Tetragenococcus halophilus CGMCC 3792. Food Chem. Toxicol. 2018, 121, 430–436. [Google Scholar] [CrossRef] [PubMed]
  23. Qiu, J.R.; Chen, Y.X.; Zhang, L.Q.; Wu, J.Z.; Zeng, X.H.; Shi, X.G.; Liu, L.M.; Chen, J.F. A comprehensive review on enzymatic biodegradation of polyethylene terephthalate. Environ. Res. 2024, 240, 117427. [Google Scholar] [CrossRef]
  24. Xu, L.P.; Sun, X.L.; Wan, X.H.; Li, H.; Yan, F.B.; Han, R.L.; Li, H.; Li, Z.J.; Tian, Y.D.; Liu, X.J.; et al. Identification of a Bacillus amyloliquefaciens H6 thioesterase involved in zearalenone detoxification by transcriptomic analysis. J. Agric. Food Chem. 2020, 68, 10071–10080. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, X.L.; Lu, H.; Li, Q.S.; Zhou, Y.; Zhou, J.T. Comparative genome and transcriptome of Rhodococcus pyridinivorans GF3 for analyzing the detoxification mechanism of anthraquinone compounds. Ecotox Environ. Safe 2022, 237, 113545. [Google Scholar] [CrossRef] [PubMed]
  26. Wei, M.L.; Dhanasekaran, S.; Yang, Q.Y.; Ngea, G.L.N.; Godana, E.A.; Zhang, H.Y. Degradation and stress response mechanism of Cryptococcus podzolicus Y3 on ochratoxin A at the transcriptional level. LWT-Food Sci. Technol. 2022, 157, 113061. [Google Scholar] [CrossRef]
  27. Hormisch, D.; Brost, I.; Kohring, G.W.; Giffhorn, F.; Kroppenstedt, R.M.; Stackebrandt, E.; Farber, P.; Holzapfel, W.H. Mycobacterium fluoranthenivorans sp. nov., a fluoranthene and aflatoxin B1 degrading bacterium from contaminated soil of a former coal gas plant. Syst. Appl. Microbiol. 2004, 27, 653–660. [Google Scholar] [CrossRef] [PubMed]
  28. Guan, S.; Ji, C.; Zhou, T.; Li, J.-X.; Ma, Q.-G.; Niu, T.-G. Aflatoxin B1 degradation by Stenotrophomonas Maltophilia and other microbes selected using coumarin medium. Int. J. Mol. Sci. 2008, 9, 1489–1503. [Google Scholar] [CrossRef] [PubMed]
  29. Rao, K.R.; Vipin, A.V.; Hariprasad, P.; Appaiah, K.A.A.; Venkateswaran, G. Biological detoxification of aflatoxin B1 by Bacillus licheniformis CFR1. Food Control 2017, 71, 234–241. [Google Scholar] [CrossRef]
  30. Shu, X.; Wang, Y.T.; Zhou, Q.; Li, M.H.; Hu, H.; Ma, Y.H.; Chen, X.; Ni, J.; Zhao, W.W.; Huang, S.W.; et al. Biological degradation of aflatoxin B1 by cell-free extracts of Bacillus velezensis DY3108 with broad pH stability and excellent thermostability. Toxins 2018, 10, 330. [Google Scholar] [CrossRef]
  31. Chen, G.J.; Fang, Q.A.; Liao, Z.L.; Xu, C.W.; Liang, Z.B.; Liu, T.; Zhong, Q.P.; Wang, L.; Fang, X.; Wang, J. Detoxification of aflatoxin B1 by a potential probiotic Bacillus amyloliquefaciens WF2020. Front. Microbiol. 2022, 13, 891091. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, L.; Ahmed, M.F.E.; Sangare, L.; Zhao, Y.J.; Selvaraj, J.N.; Xing, F.G.; Wang, Y.; Yang, H.P.; Liu, Y. Novel aflatoxin-degrading enzyme from Bacillus shackletonii L7. Toxins 2017, 9, 36. [Google Scholar] [CrossRef]
  33. Sangare, L.; Zhao, Y.J.; Folly, Y.M.E.; Chang, J.H.; Li, J.H.; Selvaraj, J.N.; Xing, F.G.; Zhou, L.; Wang, Y.; Liu, Y. Aflatoxin B1 degradation by a Pseudomonas Strain. Toxins 2014, 6, 3028–3040. [Google Scholar] [CrossRef]
  34. Qiu, T.Y.; Wang, H.M.; Yang, Y.; Yu, J.; Ji, J.; Sun, J.D.; Zhang, S.; Sun, X.L. Exploration of biodegradation mechanism by AFB1-degrading strain Aspergillus niger FS10 and its metabolic feedback. Food Control 2021, 121, 107609. [Google Scholar] [CrossRef]
  35. Pereyra, M.L.G.; Martínez, M.P.; Cavaglieri, L.R. Presence of aii homologue genes encoding for N-Acyl homoserine lactone-degrading enzyme in aflatoxin B1-decontaminating Bacillus strains with potential use as feed additives. Food Chem. Toxicol. 2019, 124, 316–323. [Google Scholar] [CrossRef]
  36. Xia, X.S.; Zhang, Y.; Li, M.Y.; Garba, B.; Zhang, Q.; Wang, Y.; Zhang, H.Y.; Li, P.W. Isolation and characterization of a Bacillus subtilis strain with aflatoxin B1 biodegradation capability. Food Control 2017, 75, 92–98. [Google Scholar] [CrossRef]
  37. Afsharmanesh, H.; Perez-Garcia, A.; Zeriouh, H.; Ahmadzadeh, M.; Romero, D. Aflatoxin degradation by Bacillus subtilis UTB1 is based on production of an oxidoreductase involved in bacilysin biosynthesis. Food Control 2018, 94, 48–55. [Google Scholar] [CrossRef]
  38. Wang, L.; Huang, W.; Sha, Y.; Yin, H.C.; Liang, Y.; Wang, X.; Shen, Y.; Wu, X.Q.; Wu, D.P.; Wang, J.S. Co-cultivation of two Bacillus strains for improved cell growth and enzyme production to enhance the degradation of aflatoxin B1. Toxins 2021, 13, 435. [Google Scholar] [CrossRef]
  39. Qian, Z.; Fields, C.J.; Yu, Y.; Lutz, S. Recent progress in engineering alpha/beta hydrolase-fold family members. Biotechnol. J. 2007, 2, 192–200. [Google Scholar] [CrossRef]
  40. Xing, M.Y.; Chen, Y.; Li, B.Q.; Tian, S.P. Characterization of a short-chain dehydrogenase/reductase and its function in patulin biodegradation in apple juice. Food Chem. 2021, 348, 129046. [Google Scholar] [CrossRef]
  41. Bernhardt, J.R.; V Lker, U.; V Lker, A.; Antelmann, H.; Schmid, R.; Mach, H.; Hecker, M. Specific and general stress proteins in Bacillus subtilis—A two-deimensional protein electrophoresis study. Microbiology 1997, 143, 999–1017. [Google Scholar] [CrossRef] [PubMed]
  42. Yang, C.; Li, M.; Peng, B.-Z. Transcriptomic analysis reveals the metabolic mechanism of patulin by Saccharomyces cerevisiae during fermentation. LWT-Food Sci. Technol. 2021, 149, 111808. [Google Scholar] [CrossRef]
  43. Butler, W.H.; Neal, G.E. Mode of action and human health aspects of aflatoxin carcinogenesis. Ann. Nutr. L’alimentation 1977, 31, 949–956. [Google Scholar] [CrossRef] [PubMed]
  44. Bedard, L.L.; Massey, T.E. Aflatoxin B1-induced DNA damage and its repair. Cancer Lett. 2006, 241, 174–183. [Google Scholar] [CrossRef] [PubMed]
  45. Edwards, U.; Rogall, T.; Blocker, H.; Emde, M.; Bottger, E.C. Isolation and direct complete nucleotide determination of entire genes-Characterization of a gene coding for 16S ribosomal RNA. Nucleic Acids Res. 1989, 17, 7843–7853. [Google Scholar] [CrossRef] [PubMed]
  46. Yoon, S.H.; Ha, S.M.; Kwon, S.; Lim, J.; Kim, Y.; Seo, H.; Chun, J. Introducing EzBioCloud: A taxonomically united database of 16S rRNA gene sequences and whole-genome assemblies. Int. J. Syst. Evol. Micr 2017, 67, 1613–1617. [Google Scholar] [CrossRef]
  47. Kumar, S.; Stecher, G.; Tamura, K. MEGA7: Molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol. Biol. Evol. 2016, 33, 1870–1874. [Google Scholar] [CrossRef]
Figure 1. The aflatoxin B1 (AFB1) degradation rates of isolated strains co-incubated with AFB1 in the dark at 37 °C for 72 h with shaking. Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Figure 1. The aflatoxin B1 (AFB1) degradation rates of isolated strains co-incubated with AFB1 in the dark at 37 °C for 72 h with shaking. Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Toxins 16 00256 g001
Figure 2. Neighbor-joining tree reconstructed using 16S rRNA gene sequences from the EzBioCloud server by MEGA software version 6.0. Metabacillus galliciensis was used as an outgroup. Numbers at branches indicate bootstrap values (>50%) from 1000 replicates.
Figure 2. Neighbor-joining tree reconstructed using 16S rRNA gene sequences from the EzBioCloud server by MEGA software version 6.0. Metabacillus galliciensis was used as an outgroup. Numbers at branches indicate bootstrap values (>50%) from 1000 replicates.
Toxins 16 00256 g002
Figure 3. Degradation of AFB1 by different components of strain DDC-4 in the dark at 37 °C for 72 h (A), and AFB1-induced enhancement effect (B). Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Figure 3. Degradation of AFB1 by different components of strain DDC-4 in the dark at 37 °C for 72 h (A), and AFB1-induced enhancement effect (B). Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Toxins 16 00256 g003
Figure 4. The influences of different factors on AFB1 degradation by the cell-free supernatant of strain DDC-4. The influence of heat, SDS and proteinase K treatments (A), temperature (B), pH (C), metal ions (D), copper concentration (E), and incubation time (F) are shown. Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Figure 4. The influences of different factors on AFB1 degradation by the cell-free supernatant of strain DDC-4. The influence of heat, SDS and proteinase K treatments (A), temperature (B), pH (C), metal ions (D), copper concentration (E), and incubation time (F) are shown. Each value is presented as the mean ± SD (n = 3). Different letters represent significant differences between species (p < 0.05).
Toxins 16 00256 g004
Figure 5. GO term enrichment analysis of upregulated (A) and downregulated (B) differentially expressed genes (DEGs). The circle size indicates the number of DEGs enriched in each pathway. The Q value indicates the significance of enrichment, increasing from blue to red. Rich factor represents the ratio of the enriched DEGs to total transcripts in this pathway. *, represents the DEGs in significantly enriched pathways.
Figure 5. GO term enrichment analysis of upregulated (A) and downregulated (B) differentially expressed genes (DEGs). The circle size indicates the number of DEGs enriched in each pathway. The Q value indicates the significance of enrichment, increasing from blue to red. Rich factor represents the ratio of the enriched DEGs to total transcripts in this pathway. *, represents the DEGs in significantly enriched pathways.
Toxins 16 00256 g005
Figure 6. KEGG pathway enrichment analysis of upregulated (A) and downregulated (B) DEGs. The circle size indicates the number of DEGs enriched in each pathway. The Q value indicates the significance of enrichment, increasing from blue to red. Rich factor represents the ratio of the enriched DEGs to total transcripts in this pathway. *, represents the DEGs in significantly enriched pathways.
Figure 6. KEGG pathway enrichment analysis of upregulated (A) and downregulated (B) DEGs. The circle size indicates the number of DEGs enriched in each pathway. The Q value indicates the significance of enrichment, increasing from blue to red. Rich factor represents the ratio of the enriched DEGs to total transcripts in this pathway. *, represents the DEGs in significantly enriched pathways.
Toxins 16 00256 g006
Figure 7. The pathway of histidine metabolism. The red rectangle indicates the enzyme-encoding gene induced by AFB1. The reaction substrates and products of these enzymes are also shown.
Figure 7. The pathway of histidine metabolism. The red rectangle indicates the enzyme-encoding gene induced by AFB1. The reaction substrates and products of these enzymes are also shown.
Toxins 16 00256 g007
Figure 8. Expression patterns of DEGs in the pathway of histidine metabolism (A), DEGs in the pathway of ‘de novo’ IMP biosynthetic process (B), upregulated oxidoreductase encoding genes (C), and upregulated hydrolase encoding genes (D). Different colors represent different expression levels (increasing from green to red). U and T indicate AFB1-untreated and -treated samples, respectively.
Figure 8. Expression patterns of DEGs in the pathway of histidine metabolism (A), DEGs in the pathway of ‘de novo’ IMP biosynthetic process (B), upregulated oxidoreductase encoding genes (C), and upregulated hydrolase encoding genes (D). Different colors represent different expression levels (increasing from green to red). U and T indicate AFB1-untreated and -treated samples, respectively.
Toxins 16 00256 g008
Figure 9. Relative expression levels of gene RS11000 (A), RS07845 (B), and RS04140 (C) between AFB1-treated and -untreated samples based on qRT-PCR analysis. U and T represent AFB1-treated and -untreated samples, respectively. 16S rRNA was used as an internal control. Each value is presented as the mean ± SD (n = 3). *, representssignificant differences between species (p < 0.05).
Figure 9. Relative expression levels of gene RS11000 (A), RS07845 (B), and RS04140 (C) between AFB1-treated and -untreated samples based on qRT-PCR analysis. U and T represent AFB1-treated and -untreated samples, respectively. 16S rRNA was used as an internal control. Each value is presented as the mean ± SD (n = 3). *, representssignificant differences between species (p < 0.05).
Toxins 16 00256 g009
Figure 10. Proposed response mechanisms of strain DDC-4 to AFB1. Up- and downregulated encoding genes were displayed in red and green fillings, respectively.
Figure 10. Proposed response mechanisms of strain DDC-4 to AFB1. Up- and downregulated encoding genes were displayed in red and green fillings, respectively.
Toxins 16 00256 g010
Table 1. Physiological and biochemical characteristics of strain DDC-4.
Table 1. Physiological and biochemical characteristics of strain DDC-4.
ItemsDDC-4
Gram stainGram-positive rod
Moveability+
Voges-Proskauer+
Oxidation of
glucose+
arabinose+
xylose+
mannitol+
Hydrolysis of
gelatin+
starch+
casein+
Growth on
citrate +
lysozyme
5 °C
10 °C
30 °C+
40 °C+
50 °C+
55 °C
65 °C
NaCl (2%)+
NaCl (5%)+
NaCl (7%)+
NaCl (10%)+
pH 5.7+
Phenylalanine dehydrolase
Catalase activity+
Nitrate reduction+
Malonate+
Indole
“+” and “−” indicates that the result is positive and negative, respectively.
Table 2. The content of aflatoxin B1 (AFB1) in moldy maize powder.
Table 2. The content of aflatoxin B1 (AFB1) in moldy maize powder.
Sample NameThe Content of AFB1 (μg/kg)
Initial6.76 ± 0.85 a
Sterilization6.39 ± 0.43 a
Treatment2.96 ± 0.92 b
Initial: moldy maize powder without treatment; sterilization: moldy maize powder was sterilized in an autoclave; treatment: moldy maize powder was mixed with the cell-free supernatant of strain DDC-4 and 10 mM Cu2+ for 48 h after sterilization.
Table 3. Differentially expressed genes (DEGs) enriched in the ‘de novo’ purine nucleobase biosynthetic process.
Table 3. Differentially expressed genes (DEGs) enriched in the ‘de novo’ purine nucleobase biosynthetic process.
Gene idGene NameGene Description
RS06555PurDphosphoribosylamine-glycine ligase
RS06560PurHIMP cyclohydrolase
RS06565PurNphosphoribosylglycinamide formyltransferase
RS06570RS06570phosphoribosylformylglycinamidine cyclo-ligase
RS06575RS06575amidophosphoribosyltransferase
RS06580PurLphosphoribosylformylglycinamidine synthase subunit PurL
RS06585PurQphosphoribosylformylglycinamidine synthase subunit PurQ
RS06590PurSphosphoribosylformylglycinamidine synthase subunit PurS
RS06595RS06595phosphoribosylaminoimidazolesuccinocarboxamide synthase
RS06600PurBadenylosuccinate lyase
RS06605PurK5-(carboxyamino)imidazole ribonucleotide synthase
RS06610PurE5-(carboxyamino)imidazole ribonucleotide mutase
Table 4. Genes encoding oxidoreductase and hydrolase induced by AFB1 in strain DDC-4.
Table 4. Genes encoding oxidoreductase and hydrolase induced by AFB1 in strain DDC-4.
Gene idGene NameGene Description
RS16210RS16210cytochrome ubiquinol oxidase subunit II
RS16215RS16215cytochrome ubiquinol oxidase subunit I
RS02280AhpAbiofilm-specific peroxidase AhpA
RS11000RS11000aldo/keto reductase
RS07845RS07845SDR family oxidoreductase
RS04960RS04960NAD(P)H-dependent oxidoreductase
RS05120RS05120NAD(P)H-dependent oxidoreductase
RS02275YkuVthiol-disulfide oxidoreductase YkuV
RS03385RS03385NUDIX hydrolase
RS19530RS19530alpha/beta hydrolase (haloalkane dehalogenase)
RS05750RS05750amidohydrolase
RS04140RS04140alpha/beta hydrolase (arylesterase)
RS02520RS02520glycoside hydrolase family 18 protein
RS00295RS00295poly-gamma-glutamate hydrolase family protein
Potential degrading genes are shown in red.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guo, J.; Zhang, H.; Zhao, Y.; Hao, X.; Liu, Y.; Li, S.; Wu, R. Identification of a Novel Aflatoxin B1-Degrading Strain, Bacillus halotolerans DDC-4, and Its Response Mechanisms to Aflatoxin B1. Toxins 2024, 16, 256. https://doi.org/10.3390/toxins16060256

AMA Style

Guo J, Zhang H, Zhao Y, Hao X, Liu Y, Li S, Wu R. Identification of a Novel Aflatoxin B1-Degrading Strain, Bacillus halotolerans DDC-4, and Its Response Mechanisms to Aflatoxin B1. Toxins. 2024; 16(6):256. https://doi.org/10.3390/toxins16060256

Chicago/Turabian Style

Guo, Jia, Hanlu Zhang, Yixuan Zhao, Xiaoxu Hao, Yu Liu, Suhong Li, and Rina Wu. 2024. "Identification of a Novel Aflatoxin B1-Degrading Strain, Bacillus halotolerans DDC-4, and Its Response Mechanisms to Aflatoxin B1" Toxins 16, no. 6: 256. https://doi.org/10.3390/toxins16060256

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop