Next Article in Journal
The Diplodia Tip Blight Pathogen Sphaeropsis sapinea Is the Most Common Fungus in Scots Pines’ Mycobiome, Irrespective of Health Status—A Case Study from Germany
Previous Article in Journal
Testing Practices for Fungal Respiratory Infections and SARS-CoV-2 among Infectious Disease Specialists, United States
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Aflatoxin Biosynthesis, Genetic Regulation, Toxicity, and Control Strategies: A Review

by
Rahim Khan
1,
Farinazleen Mohamad Ghazali
1,*,
Nor Ainy Mahyudin
2,3 and
Nik Iskandar Putra Samsudin
1,4
1
Department of Food Science, Faculty of Food Science and Technology, University Putra Malaysia, Serdang 43400, Malaysia
2
Department of Food Service and Management, Faculty of Food Science and Technology, University Putra Malaysia, Serdang 43400, Malaysia
3
Laboratory of Halal Science Research, Halal Products Research Institute, University Putra Malaysia, Serdang 43400, Malaysia
4
Laboratory of Food Safety and Food Integrity, Institute of Tropical Agriculture and Food Security, University Putra Malaysia, Serdang 43400, Malaysia
*
Author to whom correspondence should be addressed.
J. Fungi 2021, 7(8), 606; https://doi.org/10.3390/jof7080606
Submission received: 9 July 2021 / Revised: 19 July 2021 / Accepted: 22 July 2021 / Published: 27 July 2021
(This article belongs to the Section Fungal Pathogenesis and Disease Control)

Abstract

:
Aflatoxins (AFs) are highly toxic and cancer-causing compounds, predominantly synthesized by the Aspergillus species. AFs biosynthesis is a lengthy process that requires as minimum as 30 genes grouped inside 75 kilobytes (kB) of gene clusters, which are regulated by specific transcription factors, including aflR, aflS, and some general transcription factors. This paper summarizes the status of research on characterizing structural and regulatory genes associated with AF production and their roles in aflatoxigenic fungi, particularly Aspergillus flavus and A. parasiticus, and enhances the current understanding of AFs that adversely affect humans and animals with a great emphasis on toxicity and preventive methods.

1. Introduction

Aflatoxins (AFs) are secondary metabolites predominantly synthesized by Aspergillus flavus and A. parasiticus. They are highly toxic, mutagenic, carcinogenic, immunosuppressive compounds with severe detrimental effects on the human liver [1]. AFs contamination in food products is a worldwide issue and a possible risk to human and animal health [2,3]. The threat of AFs to human and animal health was first recognized after their identification as a causal agent of turkey X infection in poultry in the UK. The toxin was detected in feeds, and its properties and biological impacts were then investigated [4]. The term AF was given to the toxin since it was produced by A. flavus. In tropical and subtropical regions, billions of people were impacted by AFs adversely by consuming contaminated foods and water [5]. AF exposure is closely related to increased risk of hepatocellular carcinoma (HCC), AIDS, stunting, and malnutrition in children in America, Asia, and Africa [6,7,8,9,10,11,12,13,14]. Contamination of corn, peanuts, rice, and cottonseed with AF has been linked to agricultural losses and increased liver cancer incidence in Central America, Africa, and Asia. The study on the mechanism of AF production directly influences our capacity to diminish AF adulteration in the food supply chain.
Consequently, AF production has been developed into the most extensively studied biological activity. In the early 1990s, molecular biologists began to pay attention to AF biosynthesis, and the primary genes responsible for AF-production (nor-1 and ver-1) were identified and transcribed [15]. Later, the complete gene cluster for AF was identified [16,17]. Thus far, several genes, proteins, and regulatory mechanisms have been extensively investigated. Thus, the AF’s biosynthetic pathway helped to develop an outline for the production of mycotoxins and metabolic pathways in eukaryotic organisms. This paper will review the characterization and functions of structural genes involved in the production of AFs, genetic regulation and toxicity of AFs, and serval novel methods developed over the last few decades to minimize humans’ vulnerability to AFs in high-risk communities.

2. Biosynthetic Pathway of Aflatoxins

Following the revelation of Turkey X disease, researchers began studying AF biosynthesis by developing ultraviolet variants [18]. Different researchers recently characterized the entire 75-kb cluster on chromosome 3′s subtelomeric locus [19]. Although the gene cluster of A. flavus is similar to A. parasiticus in terms of sequencing, they are markedly different in deletion, ranging between 0.8 kb (L-strain) and 1.5 kb (S-strain). This deletion extends from the 5′ end of aflF, aflU to the whole 279 bp intergenic loci, preventing A. flavus from producing AFG1 and AFG2. DNA analysis revealed that strains of A. flavus and A. parasiticus exhibit approximately 96% affinity for this gene cluster [20]. Research on A. parasiticus identification found that 30 genes are located in this gene cluster [21]. The genes and enzymes associated with the AF biosynthesis pathway in A. parasiticus are presented in Figure 1. Two substrates contribute to AF biosynthesis, known as 1-Acetyl-CoA and 9-Malonyl-coA. Here, we will discuss the genes, encoding proteins, and precursors involved in AF production.
Figure 2 demonstrates each phase of the AF biosynthesis pathway. Norsolorinic acid (NOR) is the primary step of the AF biosynthesis pathway.

2.1. Synthesis of Norsolorinic Acid (NOR)

Three proteins, including fatty acid synthase α (aflA), fatty acid synthase β (aflB), and polyketide synthase (aflC), are responsible for the production of NorS. NorS plays a vital role in the synthesis of the hexanoyl primer through integrating with malonyl-CoA molecules. Afterward, the hexanoyl primer is moved to the region of β-ketoacyl synthase [24] and combined with malonyl-CoA to form norsolorinic acid anthrone (NAA). Due to its high reactivity, this metabolite is rapidly converted into NOR by NAA oxidase [25]. NOR, an essential metabolite synthesized in the AF’s biosynthetic pathway, exhibits a red–orange color in mutant strains of aflD (nor-1) of A. parasiticus [26].

2.2. NOR Conversion to Averantin (AVN)

AflD, a ketoreductase, reduces the NOR 1’-keto group to the AVN 1′-hydroxyl group [27]. Even though its role is defined, the mutant strain of aflD does not always result in AVN formation. The other processes contributing to this reduction remain unknown at this point.

2.3. AVN Conversion to 5′-Hydroxyaverantin (HAVN)

AflG, a monooxygenase of cytochrome P450, catalyzes the breakdown of the 5′-keto group of AVN to the 5′-hydroxyl group of HAVN [28].

2.4. HAVN Conversion to Averufin (AVF)

The HAVN dehydrogenase facilitates the dehydrogenation of the HAVN’s 5′-hydroxyl group to 5′-oxide group of oxoaverantin (OAVN) [29]. The deleted aflH mutant consistently demonstrates its ability to synthesize OAVN, suggesting the involvement of other potential mechanisms. In contrast, aflK is an OAVN cyclase that catalyzes the dehydration of 5′-oxide of OAVN to form the 2′-5′ AVF [30].

2.5. AVF Conversion to Versiconal Hemiacetal Acetate (VHA)

Being a cytochrome P450 oxidoreductase, aflV can reduce the hydride group of AVF [31]. The projected compound becomes hydrated, while aflI presumably functions as an oxidoreductase [32]. On the other hand, aflW monooxygenase is vital for incorporating the O2 atoms within the 4’-5′ ketone groups of HAVN, forming VHA.

2.6. VHA Conversion to Versiconal (VAL)

AflJ, an esterase enzyme that stimulates VHA acetate eradication, results in converting the latter into VAL [33].

2.7. VAL Conversion to Versicolorin-B (VERB)

AflK, a cyclase that catalyzes the cyclodehydration of VAL into VERB [23]. This is a crucial phase in the AF’s biosynthetic pathway as the closure of the bisfuran ring occurs at this stage. Additionally, it serves as the final precursor for the biosynthetic pathways of AFB1-AFG1 and AFB2-AFG2.

2.8. VERB Conversion to Versicolorin A (VERA)-AFB1-AFG1 Pathway

AflL, a monooxygenase of cytochrome P450, is responsible for converting the tetrahydrofuran ring to a dihydrobisfuran ring [34].

2.9. VERA Conversion to Demethylsterigmatocystin (DMST) and VERB Conversion to Dihydro Demethylsterigmatocystin (DHDMST)

AflM, aflN, aflY, and aflX are putative enzymes involved in DMST formation in the biosynthetic pathway of AFB1-AFG1 [35]. The same enzymatic steps have been suggested in the biosynthetic pathway of AFB2-AFG2, but using VERB as a substrate rather than VERA, resulting in DHDMST formation. The discrepancy amid DMST-DHDMST is comparable to that of VERA-VERB, owing to the bisfuran double bond.

2.10. DMST Conversion to Sterigmatocystin (ST) and DHDMST Conversion to Dihydrosterigmatocystin (DHST)

AflO, an O-methyltransferase, is responsible for transmitting the S-adenosylmethionine methyl group, DMST hydroxyl group, and synthesis of DHDMST to ST and DHST based on biosynthetic pathways [36].

2.11. ST Conversion to O-Methylsterigmatocystin (OMST) and DHST Conversion to Dihydro-O-Methylsterigmatocystin (DHOMST)

AflP is a second O-methyltransferase of AF biosynthesis appropriate for ST and DHST substrates [37]. Strains of A. nidulans preclude the synthesis of AF as they lack the aflP orthologue [38].

2.12. OMST Conversion to AFB1 and DHOMST Conversion to AFB2

AflQ, another monooxygenase of cytochrome P450, transforms OMST into AFB1 [39]. Yu [40] suggested a comprehensive metabolic pathway in which aflQ is replicated in C-11 hydroxylation, while aflLa may serve as a source of O2 for the keto-tautomer 11-hydroxy of OMST. These reactions might result in the formation of 370 da metabolites. On the other hand, it is assumed that AflMa is responsible for demethylating the A-ring and might work in conjunction with a cytochrome P450 as a final phase of the AF biosynthesis pathway.

2.13. Bis. OMST Conversion to AFG1 and DHOMST Conversion to AFG2

The 370-da metabolites could serve as substrates in aflU oxidation, which results in the synthesis of AFG1 and AFG2 [41]. Thus, NadA and aflF could be suitable candidates for enhancing aflU activity in the production of AFG1 and AFG2.

3. Genetic Regulation of Aflatoxin Biosynthesis

The above-mentioned phases of AF production are regulated by certain specific transcription factors such as aflR and aflS and some general transcription factors.

3.1. AflR, a Specific Transcription Factor

AflR is the ninth gene of the AF biosynthetic cluster that encodes the Cys6Zn2 transcriptional factor required for AF production. Figure 3 represents the structure of the aflR transcriptional factor.
The N-terminal part of the aflR (C6 cluster) includes the NLD (Nuclear Localization Domain), which is required for aflR movement from the cytoplasm to the nucleus [42], while the linkage portion may contribute to DNA-binding affinity. The DNA sequence is 11 bp long (5′-TCGSWNNSCGR-3′), featuring the highest binding affinity for 5′-WCGSNNNSCGA-3′. These aflR-binding loci are typically located at the 200 bp exterior to the translation start point of the AF gene. Upstream of the aflR gene’s translation start point, a partial aflR binding site indicates autoregulation. Other binding sites of diverse DNA binding proteins in the same intergenic region show that different regulatory networks could regulate the expression of aflR. Price et al. [43] analyzed 40% transcriptomes of A. parasiticus in its wild-type and aflR-mutated strains that cannot generate AFs. They discovered that the aflR mutant lacks most of the AF genes in the cluster except for aflF, Ma, N, and Na.

3.2. AflS, a Putative Transcription Factor

AflS is the 10th gene in the cluster of AF biosynthesis pathways, sharing a similar intergenic region with aflR. Although the knockout mutants demonstrated that aflS is needed for AF’s synthesis, its exact role is yet to be determined. The three possible functions of aflS are as follows:
  • It may operate as an aflR coactivator [44], although its deletion has little effect on aflR transcript levels.
  • It strongly affects the early genes involved in AF production [45].
  • AflS mutants inhibit the aflC, aflD, aflM, and aflP’s transcription by up to 20 times yet do not affect the expression of aflR. In contrast, other researchers ruled out the effects of aflS on aflM and aflP’s expression.
  • It is vital for LaeA to target a particular gene cluster. Furthermore, it is sensitive to temperature during incubation; henceforth, the expressions of aflS and aflR were increased by 24 times at 30 °C compared to 37 °C [46].

3.3. General Transcription Regulators

Seven well-known general transcriptional regulators control the biosynthetic mechanism of AFs. Each pathway is crucial to our research as it explains how specific genes of AFs are expressed or inhibited. A complex network of proteins regulates the synthesis of secondary metabolites of fungi [47]. Figure 4 shows the three essential pathways that regulate AF production.
The heterotrimeric G-protein pathways (G proteins) are general transcription regulators linked to the plasmid membrane of the cell and function as transduction impulses in reaction to foreign stimuli to maintain the cell’s physiologic conditions. The G proteins have three subunits—α, β, and γ—that abandon their function once reassembled into a trimeric form (Figure 4). The instigation occurs because of GTP binding to the G subunit. Regarding AF biosynthesis, it has been demonstrated that two subunits of Gα (GanB and FadA) prevent ST/AF synthesis in the presence of GTP through the suppression of aflR activity [48,49]. Nevertheless, it was shown that the Gβγ subunits (SfaD and GpgA) stimulate ST synthesis, implying that the G protein subunits analyzed have distinct functions in ST biosynthesis [50].
Moreover, the response to Reactive Oxygen Species (ROS) is a second transcription regulator. Figure 4 illustrates a suggested mode of action for such reactions. YapA gene’s mutation increases AF production, indicating that YapA may act as an inhibitor of ROS buildup. It was discovered that in the presence of ROS, four DNA-binding transcriptional factors, such as MsnA, AtfB, and AP-1/SrrA complexes, entangle to the specified DNA to stimulate AF production by boosting AF genes [51]. Similarly, the light-sensitive complex (VeA, VelB, and LaeA) is a third transcription regulator (Figure 5) that exhibits a low amount of VeA activity in light inside the cytoplasm. Nonetheless, VeA expression increased in the dark and was carried into the nucleus via the importer α carrier (KapA) [52]. Therefore, LaeA should be bound to a VeA/VelB compound to have an inhibitory impact over HepA. HepA is a spatial adaptor that plays a vital role in chromatins’ molecular compounds [53,54]. The HepA’s suppression prevents the transition of heterochromatin towards euchromatin in the aflR region [55].
Additionally, the ppoABC genes contain three distinct putative fatty acid oxygenases involved in producing oxylipins by fungi (Figure 6) [56]. The VeA, hydroxylated linoleic (psiα), and oleic acid (psiβ) proteins are thought to be involved in the shift from sexual to asexual reproduction in fungi [57]. A dual deletion in the ppoABC gene resulted in the inhibition of ST production, but a single loss of ppoB enhanced ST accumulation (Figure 6).
Recent research indicates that diverse Ppo oxygenases may result in the aggregation of oxylipin exterior to the cell of fungi and may stimulate the activity of G-proteins. Three additional general transcription regulators are activated in response to external stimuli (Figure 7), which are briefly discussed here. CreA is a zinc transcription factor that responds to carbon supply by triggering metabolic activities [58,59]. Additional characterization is necessary to have an in-depth insight into the fundamental processes. Likewise, AreA is the zinc transcriptional factor regulating the nitrogen pathway [60]. The aflS-aflR intergenic region has an AreA-binding site that may induce AF production.
Likewise, PacC is a transcription factor that negatively regulates the ST’s biosynthetic pathway in A. nidulans under alkaline conditions [61]. Its inactivation is pH-dependent and may be reversed under acidic conditions. In addition to the above-mentioned regulatory mechanisms, other processes, including the production site and excretion process, may affect AF synthesis.

4. Aflatoxins Toxicity

AFs are the most significant food safety concern since they are widely distributed in foods and feeds and are highly toxic. AFs carcinogenicity has long been linked with the liver, where they produce transitional metabolites; however, recent epidemiological and animal trials revealed that they were carcinogenic to organs other than the liver, including the pancreas and kidneys, bones, bladder, and central nervous system [62]. Other than that, other AF-induced long-term health impacts include anemia, malnutrition illnesses, retardation in physical and mental growth, and nervous system maturation. Despite these challenges, their modes of action need further clarity [63].

4.1. Chronic Aflatoxicosis

The consumption of AF-contaminated foods is typically linked with HCC and bile duct hyperplasia [64]; however, other organs, including kidneys, the viscera, lung, bladder, and bone, were also found to develop cancer once exposed to AFs. AFs also cause lung [65] and skin [66] cancer mainly through inhalation and direct contact. Other complications resulting from AF consumption include immunosuppression, mutagenicity, teratogenicity, and cytotoxicity in mammals, particularly in rats and humans [67]. Furthermore, AFs are associated with nutritional disorders, including kwashiorkor and growth faltering, possibly influencing the accumulation of iron, zinc, vitamin B, protein synthesis, and other enzyme activities [68,69]. Lower doses of AFs are often detrimental to the health, productivity, and reproduction of livestock and increased vulnerability to infections. Despite the insidious property of chronic aflatoxicosis, its health impacts are more catastrophic and costlier than acute infections. Chronic aflatoxicosis with hepatitis B (HB) has been reported to increase AFB1 potency up to 60-fold. According to the latest IARC Global Cancer Observatory statistics, over 841,080 new liver cancer cases and 781,631 fatalities were recorded in 2018. It equates to the age-standardized frequency of 9.3 per 100,000 persons and a fatality ratio of 93%. It is the fifth most prevalent malignancy and the primary cause of cancer-related deaths. The continents of Asia and Africa consistently produce the newest incidents, with 64,779 (7.7%) and 609,596 (72%) cases, accounting for over 80% of the total cases globally. AFB1 alone was expected to induce 25,200–155,000 infections each year [70,71], of which almost 40% of cases were reported in Africa, whereby AF-induced liver cancers are responsible for one-third of all liver cancers [72]. China holds the world’s highest rate of liver cancer at a country level, most of which were reported in the country’s southern part, where dietary AF exposure and HB chronic diseases are prevalent [73,74,75,76,77].

4.1.1. Immunotoxicity

An increased prevalence and severity of infectious diseases and extended healing time with reduced vaccine effectiveness have established that AFs impair the innate and adaptive immune system [78,79,80,81]. Some recent studies have reported that AFBO interacts with innate and immune-competent cells in the body, influencing their reproduction and generation of immune response mediators, impeding the establishment of adaptive and innate immune systems. Research conducted for observing the toxicity mechanisms in animals and humans has discovered the immunotoxicity of AFB1 on human cell lines in highly exposed regions of Ghana [82,83]. Alternatively, some researchers have examined the immunotoxicity of AFs rather than AFB1 [84,85,86,87]. Meanwhile, a general agreement exists that low to medium levels of AFB1 may not have detrimental effects on the immune system, although cell-mediated immunity is highly susceptible to AFs compared to hormonal immunity [88,89].

4.1.2. Innate Immunity

The breakdown of physiological barriers, such as epidermal and gastrointestinal mucosal tissues with pathogen invasion, has been shown in vitro and in vivo studies. For example, animal skin contact with AFB1 has been reported inducing various lesions, including intra-epidermal vesicle production and squamous cell carcinoma [90,91,92,93]. Pigs fed with a mixture of AFB1 and AFB2 for 28 days developed irritation and cutaneous ulcers on the nose, lips, and labial commissure of the mouth. Another study has shown that AFs impair the intestinal mechanical barrier integrity by intervening with the cell cycle or disrupting epithelial cells and tight junctions, cementing them together. These results have recently been supported by research in which a broiler chicken was fed with a feed comprising 0.6 mg AFB1/kg for 21 days and reported various structural and functional variations in the gastrointestinal tract, such as the contraction of mitochondria and depletion of absorptive cell goblets [94,95]. Such alterations drastically change the intestine’s ability to absorb nutrients and the innate immune response that protects against the invasion of pathogens and toxins. The impacts of AFs on immune cells, including macrophages, monocytes, natural killers, and dendritic cells, have been well established.
Additionally, AFB1 and AFM1 have been found to decrease the feasibility, multiplication, and necrosis of macrophages and cytokines’ production, including TNF-a and IL-1 [96,97,98,99]. Recently, autophagy was reported to influence the innate immune system, particularly M1-type macrophages, which are involved in inflammation responses induced by proinflammatory cytokines. Feeding research has also shown a reduced complement behavior in livestock and poultry at varying levels [100].

4.1.3. Adaptive Immunity

The inhibition of adaptive immunity following AF’s exposure is well documented, demonstrating improved susceptibility of exposed hosts to contagious agents and weakened vaccine defense [101,102]. The epidemiological research demonstrated that vaccination failed to protect poultry from bronchitis [103] and Newcastle disease once exposed to AFs. The same types of suppressive impacts have been observed in swine, in which vaccine treatment did not defend them against E. rhusiopathiae once exposed to AFB1 [104]. It is also reported that reduced replication, activation, and lymphocyte activity are critical elements of humans’ adaptive immunity. Dose and time-dependent apoptotic impacts have been seen in human blood cells after being incubated with 3.12–2000 g/L of AFB1 solution for 2–72 h [105,106]. Recent research also found that AFB1 and AFM1 substantially improved the IL-8 activity, which is connected with innate immunity.
Similarly, in humans, a high level of AFB1 is closely associated with reduced lymphocyte percentages and plays an essential role in immunization and inflammatory responses to microbial infections. In addition, previous studies’ results indicate that AFB1 inhibits cell-mediated immunity in human beings, weakening their tolerance to infection [107]. It is noteworthy, however, that humoral immunity and cell-mediated immunity might not always be distinguishable. For instance, the deregulation of dendritic cell proliferation and expression of TLRs can affect both innate and adaptive immunity since such antigen-producing cells serve as critical intermediaries for both forms of the immune response [108,109].

4.1.4. Teratogenicity

AF exposure to pregnant females or animals may inhibit the growth and development of embryos in the womb, leading to different health problems and pathological outcomes [110]. In Asian and African countries, mothers are highly exposed to AFs; they transmit AFs to their fetuses through blood circulation. AFs and their resultant biomarkers (AF metabolites, AF-DNA, and AF-albumin adducts) were found in both fetal cord and mother’s blood samples [111,112]. Hence, it is inferred that AFs and their metabolites are passed to the fetus metabolized by the same pathway as adults [113]. As a result, maternal risk factors greatly influence fetal growth, causing weight loss and premature delivery. An inverse correlation between birth weight and the number of suitable biomarkers in umbilical cord blood samples has been established in humans and animals [114,115,116].
On the other hand, very few studies have correlated AF consumption in pregnant ladies with early delivery and miscarriages [117]. Apart from the above stated detrimental health impacts, AF contaminated meals during pregnancy impair pregnant women’s well-being and expose their fetuses to indirect risks of congenital anomalies, including impeding placental growth, stillbirth, miscarriage, and premature birth. Additionally, AFs interfere with the availability of iron, selenium, and vitamins and result in anemia and low fetal development, or premature childbirth. However, data are scarce on the relationship between AF exposure and inflammation-related anemia among pregnant females. The data on dose, procedures and sensitivity to AFs exposure in pregnant women need further studies to improve pregnancy and delivery safety.

4.1.5. Malnutrition

Along with the essential toxicological impacts discussed above, AFs cause various other detrimental health effects by overlapping processes and risk factors, including malnutrition disorders (stunting), delayed physical and mental growth, fertility problems, and nervous system disorders [118,119]. Malnutrition has garnered enormous attention because of its detrimental effects on children worldwide, especially in underdeveloped nations wherein kids suffer food scarcity. To be specific, one must ensure that kids obtain physiological and cognitive maturity and are ready for adulthood as responsible and productive persons. AF exposure deprives children of these vital micronutrients and often enhances their vulnerability to AFs, which they usually detoxify with the help of endogenous antioxidants [120,121,122]. Consequently, exposed children can experience development defects beginning from the gestational phase, resulting in stunted growth and delayed physiological and psychological development. The stunted growth in kids under the age of five in African nations has been linked to chronic exposure of AFs since they depend upon indigenous agriculture items such as corn, peanuts, and derivatives as staple foods [123]. Protein-energy malnutrition illnesses, including kwashiorkor and marasmic kwashiorkor, have also been linked to the higher level of AF exposure in various African nations [124,125,126,127]. A study on malnourished Sudanese children with kwashiorkor and marasmic kwashiorkor reported that their serum and urine samples had a substantially greater concentration of AFB1 than children undernourished with marasmus. The researchers concluded that kwashiorkor was related to the long-term exposure of AFs, owing to the liver injury or an etiological element of such sickness that has not yet been identified.

4.1.6. Neurodegenerative Diseases

Apart from the well-established detrimental health impacts of AFs, there is a growing realization indicating that long-term AF exposure may often lead to neurodegenerative diseases. The AFBO and ROS synthesized by the CY450 enzyme and AF-induced oxidative stress interact with active molecules in neurons, inhibiting lipid and protein production and damaging fatty and polypeptide molecules. Additionally, AFs have been found to impair the mitochondrial activity of neurons, which results in apoptosis [128]. Furthermore, the discovery of AFs in kwashiorkor-deceased children’s brain tissues and their relation to cerebellar edema suggests that AFs can cross the brain–blood barriers, and penetrate the neurological system. The epidemiological research on the neurotoxicity of AFs has found AFs in human and animal nervous systems. In addition to oxidative stress, AFs encourage neurodegenerative issues by degenerating immunocompetent cells’ immune responses and producing proinflammatory situations in the brain and spinal cord.

4.2. Acute Toxicity

Acute toxicity is predominantly linked to AFs protein adducts since they inhibit enzymes involved in metabolic pathways, protein production, DNA replication, and immune responses. Moreover, there is a mounting indication that AFs phospholipid adducts are the primary cause of disruption and dysfunction of neurons, mitochondria, and endoplasmic reticulum [129,130]. Moreover, enhanced DNA fragmentation is a significant impact of acute aflatoxicosis reported in mouse testicles given a daily dosage of 2000 mg of AFB1 for three weeks [131]. However, a recent report investigating acute aflatoxicosis of AFB1 in chickens indicated that AF-dihydrodiol is an essential compound involved in acute aflatoxicosis as it produces AF-albumin adducts [132]. Furthermore, it is proposed that AFB2a and AFB1-phase-I metabolites contribute to acute toxicity. Other than that, AFB2a has been shown to interact with cell phospholipids and proteins, producing lipid and protein adducts and acute aflatoxicosis [133]. Notably, chronic exposure of AFs may cause similar impacts to acute toxicity; however, such impacts could be mitigated by detoxifying phase-2 enzymes and antioxidative protection pathways and DNA repairment to avoid gene mutations. On the contrary, these toxins progressively accumulate with continual exposure to small doses and develop into liver cancer such as a typical chronic exposure effect. Thus, acute aflatoxicosis can occur with a sharp accentuation of most of the harms listed above in a short period if the dosage was massive [134,135].

5. Strategies for Aflatoxin Mitigation

In Central America, Asia, and Africa, populations consume a high concentration of AF in their diets, as corn is a staple crop in these regions. Therefore, eradicating or reducing AF contamination is critical from a public health and economic perspective. Due to advancements in AF research over the past few decades, several novel techniques have been developed to mitigate human exposure to AFs in populations with an increased threat of aflatoxicosis; some of these strategies have been tried in real-life situations in developed nations. The primary approach is to employ genetically modified (GM) Bt corn to inhibit AF infection. The second strategy involves using NovaSil clay as a dietary supplement to aid in the absorption of AFs within the digestive system, limiting its solubility. Thirdly, non-aflatoxigenic strains of A. flavus (AF) are used to competitively eradicate the harmful aflatoxigenic strains of A. flavus (AF+) in the field. Finally, although still in its infancy, the last method explores the usage of plant-based volatile compounds to prevent AF corruption in seeds during storage.

5.1. Bt Corn

Bt corn is a GM crop containing genes from Bacillus thuringiensis (soil-borne bacteria), encoding for Cry; the Cyt protein endotoxin, exhibiting many insecticide activities towards lepidopteran coleopteran pests, which often inhabit cereal crops [136]. Several Bt crops have been developed by inserting the B. thuringiensis gene, encoding endotoxins with an expanded pesticide range, and improved manifestations [137,138]. Other than corn, other GM crops include wheat, cotton, rice, peanuts, tomato, tobacco, and walnuts. Notably, immunological and metabolomics studies revealed that inclusion of the Bt protein, endotoxin Cry1Ab to swine and rats had a minimal allergic effect and slightly changed metabolomic markers and IL-6, IL-4, and CD (+) t cells production [139]. Pest invasion is closely associated with spore transmission of fungi and plant injury, resulting in a rise in fungal colonization and mycotoxin buildup.
Nevertheless, it is believed that fungal mycotoxins, such as AF, are critical for fungal protection against fungivores and other pathogens [140]. Nevertheless, another potential consequence of mycotoxins’ existence in commercial crops is that they could act as pesticides for the host plants. Nonetheless, lessening insects’ infiltration by the use of GM Bt corn provides a possible approach for reducing humans’ exposure to food-related mycotoxins. While field research in South Africa revealed a ten times reduction in fumonisin B1 (FB1) levels in Bt corn relative to non-Bt corn, the efficacy tests for Bt corn to a lower AF level in field crops exhibited varying outcomes [141]. This discrepancy could be elucidated by the presence of Bt-resistant pests on corn. Henceforth, the trials that produced adverse outcomes indicate that further study is needed to comprehend the complex mechanism of preventing AF contamination in field crops [141]. Surprisingly, the Bt Cry1Ab protein manifestation within the corn genotype did not increase AF resilience among developed testcrosses [142]. Nevertheless, the testing of attained hybrids against diverse insect stresses is yet to be conducted.

5.2. Biocontrol

The use of competitive non-aflatoxigenic strains of A. flavus and A. parasiticus has shown tremendous success in the biocontrol of AF contamination in both pre-and post-harvest crops [143]. Numerous field studies, especially corn, peanuts and cotton, have consistently shown a substantial reduction in AF production ranging from 70 to 90% when non-aflatoxigenic strains of A. flavus were used [144,145,146]. Recently, the US Environmental Protection Agency (EPA) has registered two products derived from non-aflatoxigenic strains as bio-pesticides to reduce AF contamination in cotton and peanut crops in different states of America [144]. This approach employs non-aflatoxigenic strains to competitively exclude aflatoxigenic strains that compete for agricultural resources in the same niche [147]. Cotty [148] examined the potential of AF36 (non-aflatoxigenic strains of A. flavus) for reducing AF contamination in cottonseed and corn, and found that it substantially decreased AF levels when co-inoculated with aflatoxigenic strains. Another non-aflatoxigenic strain, A. flavus NRRL21882 (known as Afla-Guard), was tested for AF mitigation that was highly efficient in reducing AF production in both pre-and post-harvest stages. Likewise, some other nontoxigenic A. flavus strains (CT3 and K49) were assessed in the US and were effective in reducing AF levels in corn [149]. In Africa, a non-aflatoxigenic strain (BN30) significantly reduced AF contamination in corn when co-inoculated with an aflatoxigenic S-strain [150,151]. In Australia, the use of non-aflatoxigenic strains of A. flavus markedly reduced (approximately 95%) AF contamination in peanuts [152]. In China, a non-aflatoxigenic strain of A. flavus (AF051) has lowered the population of aflatoxigenic A. flavus by up to 99% in peanut fields [153].

5.3. Clay

Clinical trials have been performed in Africa to assess the effectiveness and safety of the food supplement called NovaSil [154]. NovaSil clay comprises an extremely disinfected clay that serves as a mycotoxin absorbent in the digestive tract. NovaSil clay considerably decreased the AFB1-albumin adducts’ amount in blood when orally taken every day before each meal for three months. After three months of its application, the amounts of AFM1 were reduced by up to 60% in urine. These findings proposed that NovaSil could be used to inhibit AF-related harmful effects during prolonged dietary exposure to clay, but not for a short interval of time (one month). Other toxic compounds, such as polycyclic aromatic hydrocarbons, were also absorbed by the clay with no side effects on the liver and kidney functions [154].

5.4. Plants Volatiles

Despite the positive results obtained with the techniques mentioned earlier, several barriers in adopting these novel approaches for AF reduction in commercial crop production remain in developing nations of the world. For instance, there is some reluctance in growing GM crops. Additionally, industrial biohazards are related to handling vast quantities of fungal spores and the unviability of clay application as an everyday food supplement. Consequently, innovative, safe, and feasible approaches must supplement the established methods that present effectiveness and acceptability. Previous studies have proven that plant-based volatile compounds such as CO2 [155], ethylene [156,157], crotyl alcohol [158,159], cotton-leaf volatiles [160], and corn-based volatiles [161,162] may hinder AF’s biosynthesis. Plant-based volatile compounds in controlling AF contamination in crops are highly beneficial from a food safety perspective.

6. Conclusions

We have made significant progress in understanding the mechanism of AF biosynthesis, control, and adverse effects of AFs on human health. The main obstacle of existing and future research will be identifying diverse regulatory system members that relate AF biosynthesis to the unrest of cell metabolism, particularly oxidative stress. Additionally, missing enzymes in AF biosynthesis must be identified to put the puzzles of AF biosynthesis together. The analysis of the AF biosynthetic pathway resulted in the development of a safety system that defends humans from AF’s detrimental effects. Conversely, in developing countries where foodborne AFs are a source of dietary exposure, there is a need for safe, economical, and practical methods to minimize AF contamination in food.

Author Contributions

F.M.G., supervision, project administration, and funding acquisition; R.K., methodology, writing—original draft preparation; N.I.P.S., conceptualization and methodology assistance; N.A.M., review, resources, and data curation. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Geran Inisiatif Putra University Putra Malaysia, UPM/GP/2017/9568800.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors want to acknowledge the financial contribution of the Ministry of Science, Technology, and Innovation (MOSTI), Malaysia, for funding this research under the Science Fund (grant number: 05-01-04-SF0750).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benkerroum, N. Chronic and acute toxicities of aflatoxins: Mechanisms of action. Int. J. Environ. Res. Public Health 2020, 17, 423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Pickova, D.; Ostry, V.; Malir, F. A recent overview of producers and important dietary sources of aflatoxins. Toxins 2021, 13, 186. [Google Scholar] [CrossRef]
  3. Khan, R.; Ghazali, M.F.; Mahyudin, N.A.; Samsudin, N.I.P. Morphological characterization, and determination of aflatoxigenic and non-aflatoxigenic Aspergillus flavus isolated from sweet corn kernels and soil in Malaysia. Agriculture 2020, 10, 450. [Google Scholar] [CrossRef]
  4. Krulj, J.; Đisalov, J.; Bočarov-Stančić, A.; Pezo, L.; Kojić, J.; Vidaković, A.; Solarov, M.B. Occurrence of aflatoxin B1 in Triticum species inoculated with Aspergillus flavus. World Mycotoxin J. 2018, 11, 247–257. [Google Scholar] [CrossRef]
  5. Khan, R.; Ghazali, F.M.; Mahyudin, N.A.; Samsudin, N.I.P. Chromatographic analysis of aflatoxigenic Aspergillus flavus isolated from Malaysian sweet corn. Separations 2021, 8, 98. [Google Scholar] [CrossRef]
  6. Raiola, A.; Tenore, G.C.; Manyes, L.; Meca, G.; Ritieni, A. Risk analysis of main mycotoxins occurring in food for children: An overview. Food Chem. Toxicol. 2015, 84, 169–180. [Google Scholar] [CrossRef]
  7. Gong, Y.Y.; Watson, S.; Routledge, M.N. Aflatoxin exposure and associated human health effects, a review of epidemiological studies. Food Saf. 2016, 4, 14–27. [Google Scholar] [CrossRef] [Green Version]
  8. Lauer, J.M.; Natamba, B.K.; Ghosh, S.; Webb, P.; Wang, J.S.; Griffiths, J.K. Aflatoxin exposure in pregnant women of mixed-status of human immunodeficiency virus infection and rate of gestational weight gain: A Ugandan cohort study. Trop. Med. Int. Health 2020, 25, 1145–1154. [Google Scholar] [CrossRef]
  9. Mitchell, N.J.; Bowers, E.; Hurburgh, C.; Wu, F. Potential economic losses to the US corn industry from aflatoxin contamination. Food Addit. Contam. A 2016, 33, 540–550. [Google Scholar] [CrossRef]
  10. Matumba, L.; Kimanya, M.; Chunga-Sambo, W.; Munthali, M.; Ayalew, A. Probabilistic dietary based estimation of the burden of aflatoxin-induced hepatocellular carcinoma among adult Malawians. World Mycotoxin J. 2019, 12, 409–419. [Google Scholar] [CrossRef]
  11. Verheecke, C.; Liboz, T.; Anson, P.; Diaz, R.; Mathieu, F. Reduction of aflatoxin production by Aspergillus flavus and Aspergillus parasiticus in interaction with Streptomyces. Microbiology 2015, 161, 967–972. [Google Scholar] [CrossRef] [PubMed]
  12. Lee, H.J.; Ryu, D. Worldwide occurrence of mycotoxins in cereals and cereal-derived food products: Public health perspectives of their co-occurrence. J. Agric. Food Chem. 2017, 65, 7034–7051. [Google Scholar] [CrossRef]
  13. Hao, S.; Hu, J.; Song, S.; Huang, D.; Xu, H.; Qian, G.; Huang, K. Selenium alleviates aflatoxin B1-induced immune toxicity through improving glutathione peroxidase 1 and selenoprotein S expression in primary porcine splenocytes. J. Agric. Food Chem. 2016, 64, 1385–1393. [Google Scholar] [CrossRef]
  14. Xu, Y.; Gong, Y.Y.; Routledge, M.N. Aflatoxin exposure assessed by aflatoxin albumin adduct biomarker in populations from six African countries. World Mycotoxin J. 2018, 11, 411–419. [Google Scholar] [CrossRef] [Green Version]
  15. Sadhasivam, S.; Britzi, M.; Zakin, V.; Kostyukovsky, M.; Trostanetsky, A.; Quinn, E.; Sionov, E. Rapid detection, and identification of mycotoxigenic fungi and mycotoxins in stored wheat grain. Toxins 2017, 9, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Caceres, I.; Khoury, A.A.; Khoury, R.E.; Lorber, S.; Oswald, I.P.; Khoury, A.E.; Bailly, J.D. Aflatoxin biosynthesis and genetic regulation: A review. Toxins 2020, 12, 150. [Google Scholar] [CrossRef] [Green Version]
  17. Keller, N.P. Fungal secondary metabolism: Regulation, function and drug discovery. Nat. Rev. Microbiol. 2019, 17, 167–180. [Google Scholar] [CrossRef]
  18. Benkerroum, N. Retrospective and prospective look at aflatoxin research and development from a practical standpoint. Int. J. Environ. Res. Public Health 2019, 16, 3633. [Google Scholar] [CrossRef] [Green Version]
  19. Zeng, H.; Cai, J.; Hatabayashi, H.; Nakagawa, H.; Nakajima, H.; Yabe, K. VerA gene is involved in the step to make the xanthone structure of demethylsterigmatocystin in aflatoxin biosynthesis. Int. J. Mol. Sci. 2020, 21, 6389. [Google Scholar] [CrossRef]
  20. Ehrlich, K.C.; Mack, B.M. Comparison of expression of secondary metabolite biosynthesis cluster genes in Aspergillus flavus, A. parasiticus, and A. oryzae. Toxins 2014, 6, 1916–1928. [Google Scholar] [CrossRef]
  21. Amare, M.G.; Keller, N.P. Molecular mechanisms of Aspergillus flavus secondary metabolism and development. Fungal Genet. Biol. 2014, 66, 11–18. [Google Scholar] [CrossRef] [PubMed]
  22. Buitimea-Cantúa, G.V.; Buitimea-Cantúa, N.E.; del Refugio Rocha-Pizaña, M.; Rosas-Burgos, E.C.; Hernández-Morales, A.; Molina-Torres, J. Antifungal and anti-aflatoxigenic activity of Heliopsis longipes roots and affinin/spilanthol against Aspergillus parasiticus by downregulating the expression of aflD and aflR genes of the aflatoxins biosynthetic pathway. J. Environ. Sci. Health B 2020, 55, 210–219. [Google Scholar] [CrossRef]
  23. Conradt, D.; Schätzle, M.A.; Haas, J.; Townsend, C.A.; Müller, M. New insights into the conversion of versicolorin A in the biosynthesis of aflatoxin B1. J. Am. Chem. Soc. 2015, 137, 10867–10869. [Google Scholar] [CrossRef] [Green Version]
  24. Herbst, D.A.; Townsend, C.A.; Maier, T. The architectures of iterative type I PKS and FAS. Nat. Prod. Rep. 2018, 35, 1046–1069. [Google Scholar] [CrossRef] [Green Version]
  25. Frisvad, J.C.; Hubka, V.; Ezekiel, C.N.; Hong, S.B.; Nováková, A.; Chen, A.J.; Houbraken, J. Taxonomy of Aspergillus section Flavi and their production of aflatoxins, ochratoxins, and other mycotoxins. Stud. Mycol. 2019, 93, 1–63. [Google Scholar] [CrossRef]
  26. Moon, Y.S.; Kim, H.M.; Chun, H.S.; Lee, S.E. Organic acids suppress aflatoxin production via lowering expression of aflatoxin biosynthesis-related genes in Aspergillus flavus. Food Control 2018, 88, 207–216. [Google Scholar] [CrossRef]
  27. Wu, Y.Z.; Lu, F.P.; Jiang, H.L.; Tan, C.P.; Yao, D.S.; Xie, C.F.; Liu, D.L. The furofuran-ring selectivity, hydrogen peroxide-production, and low Km value are the three elements for highly effective detoxification of aflatoxin oxidase. Food Chem. Toxicol. 2015, 76, 125–131. [Google Scholar] [CrossRef] [PubMed]
  28. Yabe, K.; Hatabayashi, H.; Ikehata, A.; Zheng, Y.; Kushiro, M. Development of the dichlorvos-ammonia (DV-AM) method for the visual detection of aflatoxigenic fungi. Appl. Microbiol. Biotechnol. 2015, 99, 10681–10694. [Google Scholar] [CrossRef] [PubMed]
  29. Jahanshiri, Z.; Shams-Ghahfarokhi, M.; Allameh, A.; Razzaghi-Abyaneh, M. Inhibitory effect of eugenol on aflatoxin B1 production in Aspergillus parasiticus by downregulating the expression of significant genes in the toxin biosynthetic pathway. World J. Microbiol. Biotechnol. 2015, 31, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  30. Sakuno, E.; Wen, Y.; Hatabayashi, H.; Arai, H.; Aoki, C.; Yabe, K.; Nakajima, H. Aspergillus parasiticus cyclase catalyzes two dehydration steps in aflatoxin biosynthesis. Appl. Environ. Microbiol. 2005, 71, 2999–3006. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, B.; Han, X.; Bai, Y.; Lin, Z.; Qiu, M.; Nie, X.; Yuan, J. Effects of nitrogen metabolism on growth and aflatoxin biosynthesis in Aspergillus flavus. J. Hazard. Mater. 2017, 324, 691–700. [Google Scholar] [CrossRef]
  32. Li, S.; Muhammad, I.; Yu, H.; Sun, X.; Zhang, X. Detection of aflatoxin adducts as potential markers and the role of curcumin in alleviating AFB1-induced liver damage in chickens. Ecotox. Environ. Saf. 2019, 176, 137–145. [Google Scholar] [CrossRef]
  33. Kolawole, O.; Meneely, J.; Petchkongkaew, A.; Elliott, C. A review of mycotoxin biosynthetic pathways: Associated genes and their expressions under the influence of climatic factors. Fungal Biol. Rev. 2021, 37, 8–26. [Google Scholar] [CrossRef]
  34. Hosseini, H.M.; Pour, S.H.; Amani, J.; Jabbarzadeh, S.; Hosseinabadi, M.; Mirhosseini, S.A. The effect of Propolis on inhibition of Aspergillus parasiticus growth, aflatoxin production, and expression of aflatoxin biosynthesis pathway genes. J. Environ. Health Sci. Eng. 2020, 18, 297. [Google Scholar] [CrossRef] [PubMed]
  35. Liang, D.; Xing, F.; Selvaraj, J.N.; Liu, X.; Wang, L.; Hua, H.; Liu, Y. Inhibitory effect of cinnamaldehyde, citral, and eugenol on aflatoxin biosynthetic gene expression and aflatoxin B1 biosynthesis in Aspergillus flavus. J. Food Sci. 2015, 80, 2917–2924. [Google Scholar] [CrossRef]
  36. Keller, N.P. Translating biosynthetic gene clusters into fungal armor and weaponry. Nat. Chem. Biol. 2015, 11, 671–677. [Google Scholar] [CrossRef] [Green Version]
  37. Gallo, A.; Knox, B.P.; Bruno, K.S.; Solfrizzo, M.; Baker, S.E.; Perrone, G. Identification and characterization of the polyketide synthase involved in ochratoxin A biosynthesis in Aspergillus carbonarius. Int. J. Food Microbiol. 2014, 179, 10–17. [Google Scholar] [CrossRef] [PubMed]
  38. Hathout, A.S.; Aly, S.E. Biological detoxification of mycotoxins: A review. Ann. Microbiol. 2014, 64, 905–919. [Google Scholar] [CrossRef]
  39. Rao, K.R.; Vipin, A.V.; Venkateswaran, G. Mechanism of inhibition of aflatoxin synthesis by non-aflatoxigenic strains of Aspergillus flavus. Microb. Pathog. 2020, 147, 104280. [Google Scholar]
  40. Yu, J. Current understanding on aflatoxin biosynthesis and future perspective in reducing aflatoxin contamination. Toxins 2012, 4, 1024–1057. [Google Scholar] [CrossRef] [Green Version]
  41. Callicott, K.; Cotty, P. Method for monitoring deletions in the aflatoxin biosynthesis gene cluster of Aspergillus flavus with multiplex PCR. Lett. Appl. Microbiol. 2015, 60, 60–65. [Google Scholar] [CrossRef]
  42. Tang, M.C.; Zou, Y.; Watanabe, K.; Walsh, C.T.; Tang, Y. Oxidative cyclization in natural product biosynthesis. Chem. Rev. 2017, 117, 5226–5333. [Google Scholar] [CrossRef]
  43. Price, M.S.; Yu, J.; Nierman, W.C.; Kim, H.S.; Pritchard, B.; Jacobus, C.A.; Bhatnagar, D.; Cleveland, T.E.; Payne, G.A. The aflatoxin pathway regulator aflR induces gene transcription inside and outside of the aflatoxin biosynthetic cluster. FEMS Microbiol. Lett. 2006, 255, 275–279. [Google Scholar] [CrossRef] [Green Version]
  44. Faustinelli, P.C.; Palencia, E.R.; Sobolev, V.S.; Horn, B.W.; Sheppard, H.T.; Lamb, M.C.; Arias, R.S. Study of the genetic diversity of the aflatoxin biosynthesis cluster in Aspergillus section Flavi using insertion/deletion markers in peanut seeds from Georgia, USA. Mycologia 2017, 109, 200–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Li, R.; Oliver, R.A.; Townsend, C.A. Identification and characterization of the sulfazecin monobactams biosynthetic gene cluster. Cell Chem. Biol. 2017, 24, 24–34. [Google Scholar] [CrossRef] [Green Version]
  46. Ojiambo, P.S.; Battilani, P.; Cary, J.W.; Blum, B.H.; Carbone, I. Cultural and genetic approaches to managing aflatoxin contamination: Recent insights provide opportunities for improved control. Phytopathology 2018, 108, 1024–1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Alkhayyat, F.; Yu, J.H. Upstream regulation of mycotoxin biosynthesis. Adv. Appl. Microbiol. 2014, 86, 251–278. [Google Scholar] [PubMed]
  48. Chan, P.; Han, X.; Zheng, B.; DeRan, M.; Yu, J.; Jarugumilli, G.K.; Wu, X. Autopalmitoylation of TEAD proteins regulates the transcriptional output of the Hippo pathway. Nat. Chem. Biol. 2016, 12, 282–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Pfannenstiel, B.T.; Zhao, X.; Wortman, J.; Wiemann, P.; Throckmorton, K.; Spraker, J.E.; Lim, F.Y. Revitalization of a forward genetic screen identifies three new regulators of fungal secondary metabolism in the genus Aspergillus. MBio 2017, 8, e01246-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Scala, V.; Giorni, P.; Cirlini, M.; Ludovici, M.; Visentin, I.; Cardinale, F.; Battilani, P. LDS1-produced oxylipins are negative regulators of growth, conidiation, and fumonisin synthesis in the fungal maize pathogen Fusarium verticillioides. Front. Microbiol. 2014, 5, 669. [Google Scholar] [CrossRef]
  51. Hong, S.Y.; Roze, L.V.; Wee, J.; Linz, J.E. Evidence that a transcription factor regulatory network coordinates oxidative stress response and secondary metabolism in Aspergilli. Microbiologyopen 2013, 2, 144–160. [Google Scholar] [CrossRef]
  52. Cary, J.; Han, Z.; Yin, Y.; Lohmar, J.; Shantappa, S.; Harris-Coward, P.; Arroyo-Manzanares, N. Transcriptome analysis of Aspergillus flavus reveals the veA-dependent regulation of secondary metabolite gene clusters, including the novel aflavarin cluster. Eukaryot. Cell 2015, 14, 983–997. [Google Scholar] [CrossRef] [Green Version]
  53. Alvarez-Dominguez, J.R.; Bai, Z.; Xu, D.; Yuan, B.; Lo, K.A.; Yoon, M.J.; Chen, S. De novo reconstruction of adipose tissue transcriptomes reveals long non-coding RNA regulators of brown adipocyte development. Cell Metab. 2015, 21, 764–776. [Google Scholar] [CrossRef] [Green Version]
  54. Martín, J.F. Key role of LaeA and velvet complex proteins on expression of β-lactam and PR-toxin genes in Penicillium chrysogenum: Cross-talk regulation of secondary metabolite pathways. J. Ind. Microbiol. Biotechnol. 2017, 44, 525–535. [Google Scholar] [CrossRef]
  55. Reyes-Dominguez, Y.; Bok, J.W.; Berger, H.; Shwab, E.K.; Basheer, A.; Gallmetzer, A.; Scazzocchio, C.; Keller, N.; Strauss, J. Heterochromatic marks are associated with the repression of secondary metabolism clusters in Aspergillus nidulans. Mol. Microbiol. 2010, 76, 1376–1386. [Google Scholar] [CrossRef] [Green Version]
  56. Fischer, G.J.; Keller, N.P. Production of cross-kingdom oxylipins by pathogenic fungi: An update on their role in development and pathogenicity. J. Microbiol. 2016, 54, 254–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Han, J.E.; Seo, M.J.; Shin, K.C.; Oh, D.K. Production of 10R-hydroxy unsaturated fatty acids from hemp seed oil hydrolyzate by recombinant Escherichia coli cells expressing PpoC from Aspergillus nidulans. Appl. Microbiol. Biotechnol. 2016, 100, 7933–7944. [Google Scholar] [CrossRef] [PubMed]
  58. Alam, M.A.; Kamlangdee, N.; Kelly, J.M. The CreB deubiquitinating enzyme does not directly target the CreA repressor protein in Aspergillus nidulans. Curr. Genet. 2017, 63, 647–667. [Google Scholar] [CrossRef] [PubMed]
  59. Peng, M.; Khosravi, C.; Lubbers, R.J.; Kun, R.S.; Pontes, M.V.A.; Battaglia, E.; de Vries, R.P. CreA-mediated repression of gene expression occurs at low monosaccharide levels during fungal plant biomass conversion in a time and substrate-dependent manner. Cell Surf. 2021, 7, 100050. [Google Scholar] [CrossRef]
  60. Mannaa, M.; Kim, K.D. Influence of temperature and water activity on deleterious fungi and mycotoxin production during grain storage. Mycobiology 2017, 45, 240–254. [Google Scholar] [CrossRef]
  61. Bills, G.F.; Gloer, J.B. Biologically active secondary metabolites from the fungi. Microbiol. Spectr. 2016, 4, 4–6. [Google Scholar] [CrossRef]
  62. Fouad, M.A.; Ruan, D.; El-Senousey, K.H.; Chen, W.; Jiang, S.; Zheng, C. Harmful effects and control strategies of aflatoxin B1 produced by Aspergillus flavus and Aspergillus parasiticus strains on poultry: Review. Toxins 2019, 11, 176. [Google Scholar] [CrossRef] [Green Version]
  63. Kourousekos, G.D.; Theodosiadou, E. Effects of aflatoxins on male reproductive system: A review. J. Hell. Vet. Med. Soc. 2015, 66, 201–210. [Google Scholar] [CrossRef] [Green Version]
  64. McGlynn, K.A.; Petrick, J.L.; El-Serag, H.B. Epidemiology of hepatocellular carcinoma. Hepatology 2021, 73, 4–13. [Google Scholar] [CrossRef] [PubMed]
  65. Alshannaq, A.F.; Gibbons, J.G.; Lee, M.K.; Han, K.H.; Hong, S.B.; Yu, J.H. Controlling aflatoxin contamination and propagation of Aspergillus flavus by a soy-fermenting Aspergillus oryzae strain. Sci. Rep. 2018, 8, 1–14. [Google Scholar] [CrossRef] [PubMed]
  66. Marchese, S.; Polo, A.; Ariano, A.; Velotto, S.; Costantini, S.; Severino, L. Aflatoxin B1, and M1: Biological properties and their involvement in cancer development. Toxins 2018, 10, 214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Klvana, M.; Bren, U. Aflatoxin B1–formamidopyrimidine DNA adducts relationships between structures, free energies, and melting temperatures. Molecules 2019, 24, 150. [Google Scholar] [CrossRef] [Green Version]
  68. Turner, P.C. The molecular epidemiology of chronic aflatoxin driven impaired child growth. Scientifica 2013, 2013, 152879. [Google Scholar] [CrossRef] [Green Version]
  69. WHO (World Health Organization). Evaluation of Certain Contaminants in Food: Eighty-Third Report of the Joint FAO/WHO Expert Committee on Food Additives; World Health Organization: Geneva, Switzerland, 2017. [Google Scholar]
  70. Klingelhöfer, D.; Zhu, Y.; Braun, M.; Bendels, M.H.; Brüggmann, D.; Groneberg, D.A. Aflatoxin–publication analysis of a global health threat. Food Control 2018, 89, 280–290. [Google Scholar] [CrossRef]
  71. Liu, Y.; Wu, F. Global burden of aflatoxin-induced hepatocellular carcinoma: A risk assessment. Environ. Health Perspect. 2010, 118, 818–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Habibi, N.; Nassiri-Toosi, M.; Sharafi, H.; Alavian, S.M.; Shams-Ghahfarokhi, M.; Razzaghi-Abyaneh, M. Aflatoxin B1 exposure and the risk of hepatocellular carcinoma in Iranian carriers of viral hepatitis B and C. Toxin Rev. 2019, 38, 234–239. [Google Scholar] [CrossRef]
  73. Gibb, H.; Devleesschauwer, B.; Bolger, P.M.; Wu, F.; Ezendam, J.; Cliff, J.; Zeilmaker, M.; Verger, P.; Pitt, J.; Baines, J. World Health Organization estimates of the global and regional disease burden of four foodborne chemical toxins, 2010: A data synthesis. F1000Reserch 2015, 4, 1393. [Google Scholar] [CrossRef]
  74. Yao, J.G.; Huang, X.Y.; Long, X.D. Interaction of DNA repair gene polymorphisms and aflatoxin B1 in the risk of hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2014, 7, 6231–6244. [Google Scholar] [PubMed]
  75. Vartanian, V.; Minko, I.G.; Chawanthayatham, S.; Egner, P.A.; Lin, Y.C.; Earley, L.F.; Makar, R.; Eng, J.R.; Camp, M.T.; Li, L. NEIL1 protects against aflatoxin-induced hepatocellular carcinoma in mice. Proc. Natl. Acad. Sci. USA 2017, 114, 4207–4212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Long, X.D.; Huang, H.D.; Xia, Q. The polymorphism of XRCC3 codon 241 and the hotspot mutation in the TP53 gene in hepatocellular carcinoma induced by aflatoxin B1. J. Tumor 2014, 2, 272–277. [Google Scholar]
  77. Long, X.D.; Zhao, D.; Wang, C.; Huang, X.Y.; Yao, J.G.; Ma, Y.; Wei, Z.H.; Liu, M.; Zeng, L.X.; Mo, X.Q.; et al. Genetic polymorphisms in DNA repair genes XRCC4 and XRCC5 and aflatoxin B1-related hepatocellular carcinoma. Epidemiology 2013, 24, 671–681. [Google Scholar] [CrossRef]
  78. Arafa, M.; Besheer, T.; El-Eraky, A.M.; Abo El-khair, S.M.; Elsamanoudy, A.Z. Genetic variants of XRCC1 and risk of hepatocellular carcinoma in chronic hepatitis C patients. Br. J. Biomed. Sci. 2019, 76, 64–69. [Google Scholar] [CrossRef]
  79. Coppock, R.W.; Christian, R.G.; Jacobsen, B.J. Aflatoxins. In Veterinary Toxicology, 3rd ed.; Gupta, R.C., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 983–994. [Google Scholar]
  80. Raafat, N.; Emam, W.A.; Gharib, A.F.; Nafea, O.E.; Zakaria, M. Assessment of serum aflatoxin B1 levels in neonatal jaundice with glucose-6-phosphate dehydrogenase deficiency: A preliminary study. Mycotoxin Res. 2021, 37, 109–116. [Google Scholar] [CrossRef]
  81. Mohsenzadeh, M.S.; Hedayati, N.; Riahi-Zanjani, B.; Karimi, G. Immunosuppression following dietary aflatoxin B1 exposure: A review of the existing evidence. Toxin Rev. 2016, 35, 121–127. [Google Scholar] [CrossRef]
  82. Barany, A.; Guilloto, M.; Cosano, J.; De Boevre, M.; Oliva, M.; De Saeger, S.; Mancera, J.M. Dietary aflatoxin B1 (AFB1) reduces growth performance, affecting growth axis, metabolism, and tissue integrity in juvenile gilthead sea bream (Sparus aurata). Aquaculture 2021, 533, 736189. [Google Scholar] [CrossRef]
  83. Gashaw, M. Review on Mycotoxins in Feeds: Implications to livestock and human health. J. Agric. Res. Dev. 2016, 5, 137–144. [Google Scholar]
  84. Mohammadi, A.; Mehrzad, J.; Mahmoudi, M.; Schneider, M. Environmentally appropriate level of aflatoxin B1 dysregulates human dendritic cells through signaling on key toll-like receptors. Int. J. Toxicol. 2014, 33, 175–186. [Google Scholar] [CrossRef] [PubMed]
  85. Jolly, P.E. Aflatoxin: Does it contribute to an increase in HIV viral load? Future Microbiol. 2014, 9, 121–124. [Google Scholar] [CrossRef] [PubMed]
  86. Shirani, K.; Zanjani, B.R.; Mahmoudi, M.; Jafarian, A.H.; Hassani, F.V.; Giesy, J.P.; Karimi, G. Immunotoxicity of aflatoxin M1: As a potent suppressor of innate and acquired immune systems in a subacute study. J. Sci. Food Agric. 2018, 98, 5884–5892. [Google Scholar] [CrossRef] [PubMed]
  87. Bianco, G.; Russo, R.; Marzocco, S.; Velotto, S.; Autore, G.; Severino, L. Modulation of macrophage activity by aflatoxins B1 and B2 and their metabolites aflatoxins M1 and M2. Toxicon 2012, 59, 644–650. [Google Scholar] [CrossRef]
  88. Chaytor, A.C.; See, M.T.; Hansen, J.A.; de Souza, A.L.P.; Middleton, T.F.; Kim, S.W. Effects of chronic exposure of diets with reduced aflatoxin concentrations deoxynivalenol on growth and immune status of pigs1. J. Anim. Sci. 2011, 89, 124–135. [Google Scholar] [CrossRef]
  89. Adhikari, M.; Negi, B.; Kaushik, N.; Adhikari, A.; Al-Khedhairy, A.A.; Kaushik, N.K.; Choi, E.H. T-2 mycotoxin: Toxicological effects and decontamination strategies. Oncotarget 2017, 8, 33933–33952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Giles, A.J.; Hutchinson, M.K.N.; Sonnemann, H.M.; Jung, J.; Fecci, P.E.; Ratnam, N.M.; Gilbert, M.R. Dexamethasone-induced immunosuppression: Mechanisms and implications for immunotherapy. J. Immunother. Cancer 2018, 6, 1–13. [Google Scholar] [CrossRef] [PubMed]
  91. Valtchev, I.; Koynarski, T.; Sotirov, L.; Nikolov, Y.; Petkov, P. Effect of aflatoxin B1 on moulard duck’s natural immunity. Pak. Vet. J. 2015, 35, 67–70. [Google Scholar]
  92. Yunus, A.W.; Razzazi-Fazeli, E.; Bohm, J. Aflatoxin B1 in affecting broiler’s performance, immunity, and gastrointestinal tract: A review of history and contemporary issues. Toxins 2011, 3, 566–590. [Google Scholar] [CrossRef] [Green Version]
  93. Doi, K.; Uetsuka, K. Mechanisms of mycotoxin-induced dermal toxicity and tumorigenesis through oxidative stress-related pathways. J. Toxicol. Pathol. 2014, 27, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Wangia, R.N.; Tang, L.; Wang, J.S. Occupational exposure to aflatoxins and health outcomes: A review. J. Environ. Sci. Health C 2019, 37, 215–234. [Google Scholar] [CrossRef]
  95. Chaudhary, Z.; Rehman, K.; Akash, M.S.H. Mechanistic Insight of Mycotoxin-Induced Neurological Disorders and Treatment Strategies. Environ. Contam. Neurol. Disord. 2021, 125–146. [Google Scholar] [CrossRef]
  96. Yin, H.; Jiang, M.; Peng, X.; Cui, H.; Zhou, Y.; He, M.; Zuo, Z.; Ouyang, P.; Fan, J.; Fang, J. The molecular mechanism of G2M cell cycle arrest induced by AFB1 in the jejunum. Oncotarget 2016, 7, 35592–35606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wang, F.; Zuo, Z.; Chen, K.; Gao, C.; Yang, Z.; Zhao, S.; Li, J.; Song, H.; Peng, X.; Fang, J.; et al. Histopathological injuries, ultrastructural changes, and depressed TLR expression in the small intestine of broiler chickens with aflatoxin B1. Toxins 2018, 10, 131. [Google Scholar] [CrossRef] [Green Version]
  98. Chatterjee, D.; Ghosh, P. Sub-cytotoxic concentration of aflatoxin B2 prevents NO-mediated Increased mitochondrial membrane potential and intracellular killing of Candida albicans in macrophages. Adv. Life Sci. 2012, 2, 52–56. [Google Scholar] [CrossRef] [Green Version]
  99. Ma, J.; Liu, Y.; Guo, Y.; Ma, Q.; Ji, C.; Zhao, L. Transcriptional profiling of aflatoxin B1-induced oxidative stress and inflammatory response in macrophages. Toxins 2021, 13, 401. [Google Scholar] [CrossRef]
  100. Brown, R.; Priest, E.; Naglik, J.R.; Richardson, J.P. Fungal Toxins and Host Immune Responses. Front. Microbiol. 2021, 12, 697. [Google Scholar] [CrossRef]
  101. Lin, S.; Gao, P.; Li, Q.; Zhang, Y.; Hu, J.; Cai, M.; Zhou, P. Aflatoxin influences achalasia symptomatology. Mol. Med. Rep. 2020, 21, 1276–1284. [Google Scholar] [CrossRef] [Green Version]
  102. Malvandi, A.M.; Mehrzad, J.; Saleh-Moghaddam, M. Biologically relevant doses of mixed aflatoxins B and G up-regulate MyD88, TLR2, TLR4 and CD14 transcripts in human PBMCs. Immunopharmacol. Immunotoxicol. 2013, 35, 528–532. [Google Scholar] [CrossRef]
  103. Chen, X.; Horn, N.; Cotter, P.F.; Applegate, T.J. Growth, serum biochemistry, complement activity, and liver gene expression responses of Pekin ducklings to graded levels of cultured aflatoxin B1. Poult. Sci. 2014, 93, 2028–2036. [Google Scholar] [CrossRef]
  104. Monson, M.S.; Coulombe, R.A.; Reed, K.M. Aflatoxicosis: Lessons from toxicity and responses to aflatoxin B1 in poultry. Agriculture 2015, 5, 742–777. [Google Scholar] [CrossRef] [Green Version]
  105. Pierron, A.; Alassane-Kpembi, I.; Oswald, I.P. Impact of mycotoxin on immune response and consequences for pig health. Anim. Nutr. 2016, 2, 63–68. [Google Scholar] [CrossRef] [PubMed]
  106. Shahabi-Ghahfarokhi, B.; Gholami-Ahangaran, M.; Dehkordi, M. Aflatoxin effect on humoral and mucosal immune responses against infectious bronchitis vaccine in broilers. Thai Vet. Med. 2016, 46, 149–153. [Google Scholar]
  107. Mikami, O.; Yamaguchi, H.; Murata, H.; Nakajima, Y.; Miyazaki, S. Induction of apoptotic lesions in the liver and lymphoid tissues and modulation of cytokine mRNA expression by acute exposure to deoxynivalenol in piglets. J. Vet. Sci. 2010, 11, 107–113. [Google Scholar] [CrossRef] [Green Version]
  108. Alpsoy, L.; Kotan, E.; Tatar, A.; Agar, G. Protective effects of selenium against sister chromatid exchange induced by AFG1 in human lymphocytes in vitro. Hum. Exp. Toxicol. 2011, 30, 515–519. [Google Scholar] [CrossRef] [PubMed]
  109. Mamo, F.T.; Abate, B.A.; Tesfaye, K.; Nie, C.; Wang, G.; Liu, Y. Mycotoxins in Ethiopia: A review on prevalence, economic and health impacts. Toxins 2020, 12, 648. [Google Scholar] [CrossRef]
  110. Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells, and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef] [PubMed]
  111. Ishikawa, A.T.; Hirooka, E.Y.; Alvares, E.; Silva, P.L.; Bracarense, A.P.F.R.L.; Flaiban, K.K.M.D.C.; Akagi, C.Y.; Kawamura, O.; Costa, M.C.D.; Itano, E.N. Impact of a single oral acute dose of aflatoxin B1 on liver function/cytokines and the lymphoproliferative response in C57Bl/6 mice. Toxins 2017, 9, 374. [Google Scholar] [CrossRef] [Green Version]
  112. Smith, L.E.; Prendergast, A.J.; Turner, P.C.; Humphrey, J.H.; Stoltzfus, R.J. Aflatoxin exposure during pregnancy, maternal anemia, and adverse birth outcomes. Am. J. Trop. Med. Hyg. 2017, 96, 770–776. [Google Scholar] [CrossRef] [Green Version]
  113. Maleki, F.; Abdi, S.; Davodian, E.; Haghani, K.; Bakhtiyari, S. Exposure of infants to aflatoxin M1 from mother’s breast milk in Ilam, Western Iran. Osong Public Health Res. Perspect. 2015, 6, 283–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Khlangwiset, P.; Shephard, G.S.; Wu, F. Aflatoxins and growth impairment: A review. Crit. Rev. Toxicol. 2011, 41, 740–755. [Google Scholar] [CrossRef]
  115. Watson, S.; Moore, S.E.; Darboe, M.K.; Chen, G.; Tu, Y.K.; Huang, Y.T.; Gong, Y.Y. Impaired growth in rural Gambian infants exposed to aflatoxin: A prospective cohort study. BMC Public Health 2018, 18, 1–9. [Google Scholar] [CrossRef] [PubMed]
  116. Partanen, H.A.; El-Nezami, H.S.; Leppänen, J.M.; Myllynen, P.K.; Woodhouse, H.J.; Vähäkangas, K.H. Aflatoxin B1 transfer and metabolism in human placenta. Toxicol. Sci. 2010, 113, 216–225. [Google Scholar] [CrossRef]
  117. Kyei, N.N.; Boakye, D.; Gabrysch, S. Maternal mycotoxin exposure and adverse pregnancy outcomes: A systematic review. Mycotoxin Res. 2020, 36, 243–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Shuaib, F.M.; Jolly, P.E.; Ehiri, J.E.; Yatich, N.; Jiang, Y.; Funkhouser, E.; Person, S.D.; Wilson, C.; Ellis, W.O.; Wang, J.S. Association between birth outcomes and aflatoxin B1 biomarker blood levels in pregnant women in Kumasi, Ghana. Trop. Med. Int. Health 2010, 15, 160–167. [Google Scholar] [CrossRef] [PubMed]
  119. Khan, R.; Ghazali, F.M.; Mahyudin, N.A.; Samsudin, N.I.P. Biocontrol of aflatoxins using non-aflatoxigenic Aspergillus flavus: A literature review. J. Fungi. 2021, 7, 381. [Google Scholar] [CrossRef]
  120. Hayashi, A.; Dorantes-Aranda, J.; Bowman, P.J.; Hallegraeff, G. Combined cytotoxicity of the phycotoxin okadaic acid and mycotoxins on intestinal and neuroblastoma human cell models. Toxins 2018, 10, 526. [Google Scholar] [CrossRef] [Green Version]
  121. El Khoury, D.; Fayjaloun, S.; Nassar, M.; Sahakian, J.; Aad, Y.P. Updates on the effect of mycotoxins on male reproductive efficiency in mammals. Toxins 2019, 11, 515. [Google Scholar] [CrossRef] [Green Version]
  122. Obuseh, F.A.; Jolly, P.E.; Kulczycki, A.; Ehiri, J.; Waterbor, J.; Desmond, R.A.; Preko, P.O.; Jiang, Y.; Piyathilake, C.J. Aflatoxin levels, plasma vitamins A and E concentrations, and their association with HIV and hepatitis B virus infections in Ghanaians: A cross-sectional study. J. Int. AIDS Soc. 2011, 14, 53. [Google Scholar] [CrossRef]
  123. Zhao, L.; Feng, Y.; Deng, J.; Zhang, N.Y.; Zhang, W.P.; Liu, X.L.; Rajput, S.A.; Qi, D.S.; Sun, L.H. Selenium deficiency aggravates aflatoxin B1-induced immunotoxicity in chick spleen by regulating six selenoprotein genes and redox/inflammation/apoptotic signaling. J. Nutr. 2019, 149, 894–901. [Google Scholar] [CrossRef]
  124. Knipstein, B.; Huang, J.; Barr, E.; Sossenheimer, P.; Dietzen, D.; Egner, P.A.; Rudnick, D.A. Dietary aflatoxin-induced stunting in a novel rat model: Evidence for toxin-induced liver injury and hepatic growth hormone resistance. Pediatr. Res. 2015, 78, 120–127. [Google Scholar] [CrossRef] [Green Version]
  125. Monyo, E.S.; Njoroge, S.M.C.; Coe, R.; Osiru, M.; Madinda, F.; Waliyar, F.; Thakur, R.P.; Chilunjika, T.; Anitha, S. Occurrence and distribution of aflatoxin contamination in groundnuts (Arachis hypogaea L.) and population density of aflatoxigenic Aspergilli in Malawi. Crop Prot. 2012, 42, 149–155. [Google Scholar] [CrossRef] [Green Version]
  126. Onyemelukwe, G.; Ogoina, D.; Ibiam, G.E.; Ogbadu, G.H. Aflatoxins in body fluids and Nigerian children with protein-energy malnutrition. Afr. J. Food Agric. Nutr. Dev. 2012, 12, 6553–6566. [Google Scholar]
  127. Ayelign, A.; Woldegiorgis, A.Z.; Adish, A.; De Boevre, M.; Heyndrickx, E.; De Saeger, S. Assessment of aflatoxin exposure among young children in Ethiopia using urinary biomarkers. Food Addit. Contam. A 2017, 34, 1606–1616. [Google Scholar] [CrossRef]
  128. Soriano, J.M.; Rubini, A.; Morales-Suarez, M.; Merino-Torres, J.F.; Silvestre, D. Aflatoxins in organs and biological samples from children affected by kwashiorkor, marasmus, and marasmic-kwashiorkor: A scoping review. Toxicon 2020, 185, 174–183. [Google Scholar] [CrossRef] [PubMed]
  129. McMillan, A.; Renaud, J.B.; Burgess, K.M.; Orimadegun, A.E.; Akinyinka, O.O.; Allen, S.J.; Sumarah, M.W. Aflatoxin exposure in Nigerian children with severe acute malnutrition. Food Chem. Toxicol. 2018, 111, 356–362. [Google Scholar] [CrossRef] [PubMed]
  130. Shen, H.; Liu, J.; Wang, Y.; Lian, H.; Wang, J.; Xing, L.; Zhang, X. Aflatoxin G1-induced oxidative stress causes DNA damage and triggers apoptosis through MAPK signaling pathway in A549 cells. Food Chem. Toxicol. 2013, 62, 661–669. [Google Scholar] [CrossRef] [PubMed]
  131. Abdel-Wahhab, M.A.; El-Nekeety, A.A.; Hathout, A.S.; Salman, A.S.; Abdel-Aziem, S.H.; Sabry, B.A.; Jaswir, I. Bioactive compounds from Aspergillus niger extract enhance the antioxidant activity and prevent the genotoxicity in aflatoxin B1-treated rats. Toxicon 2020, 181, 57–68. [Google Scholar] [CrossRef] [PubMed]
  132. Rushing, B.R.; Selim, M.I. Structure and oxidation of pyrrole adducts formed between aflatoxin B2a and biological amines. Chem. Res. Toxicol. 2017, 30, 1275–1285. [Google Scholar] [CrossRef]
  133. Zamir-Nasta, T.; Razi, M.; Shapour, H.; Malekinejad, H. Roles of p21, p53, cyclin D1, CDK-4, estrogen receptor alpha in aflatoxin B1-induced cytotoxicity in testicular tissue of mice. Environ. Toxicol. 2018, 33, 385–395. [Google Scholar] [CrossRef] [PubMed]
  134. Diaz, G.J.; Murcia, H.W. An unusually high production of hepatic aflatoxin B1-dihydrodiol, the possible explanation for the high susceptibility of ducks to aflatoxin B1. Sci. Rep. 2019, 9, 8010. [Google Scholar] [CrossRef] [PubMed]
  135. Colakoglu, F.; Donmez, H.H. Effects of aflatoxin on the liver and shielding effectiveness of esterified glucomannan in Merino rams. Sci. World J. 2012, 2012, 462925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Fan, Y.; Liu, L.; Zhao, L.; Wang, X.; Wang, D.; Huang, C.; Zhang, J.; Ji, C.; Ma, Q. Influence of Bacillus subtilis ANSB060 on growth, digestive enzyme and aflatoxin residue in Yellow River carp fed diets contaminated with aflatoxin B1. Food Chem. Toxicol. 2018, 113, 108–114. [Google Scholar] [CrossRef] [PubMed]
  137. Peng, Z.; Chen, L.; Zhu, Y.; Huang, Y.; Hu, X.; Wu, Q.; Nüssler, A.K.; Liu, L.; Yang, W. Current major degradation methods for aflatoxins: A review. Trends Food Sci. Technol. 2018, 80, 155–166. [Google Scholar] [CrossRef]
  138. Bediako, K.A.; Ofori, K.; Offei, S.K.; Dzidzienyo, D.; Asibuo, J.Y.; Amoah, R.A. Aflatoxin contamination of groundnut (Arachis hypogaea L.): Predisposing factors and management interventions. Food Control 2019, 98, 61–67. [Google Scholar] [CrossRef]
  139. Theumer, M.G.; Henneb, Y.; Khoury, L.; Snini, S.P.; Tadrist, S.; Canlet, C.; Audebert, M. Genotoxicity of aflatoxins and their precursors in human cells. Toxicol. Lett. 2018, 287, 100–107. [Google Scholar] [CrossRef]
  140. Rohlfs, M. Fungal secondary metabolite dynamics in fungus–grazer interactions: Novel insights and unanswered questions. Front. Microbiol. 2015, 5, 2–5. [Google Scholar] [CrossRef] [Green Version]
  141. Moral, J.; Garcia-Lopez, M.T.; Camiletti, B.X.; Jaime, R.; Michailides, T.J.; Bandyopadhyay, R.; Ortega-Beltran, A. Present status, and perspective on the future use of aflatoxin biocontrol products. Agronomy 2020, 10, 491. [Google Scholar] [CrossRef] [Green Version]
  142. Valencia-Quintana, R.; Sánchez-Alarcón, J.; Tenorio, M.G.; Deng, Y.; Waliszewski, S.M.; Valera, M.Á. Preventive strategies aimed at reducing the health risks of aflatoxin B1. Toxicol. Environ. Health Sci. 2012, 4, 71–79. [Google Scholar] [CrossRef]
  143. Bandyopadhyay, R.; Ortega-Beltran, A.; Akande, A.; Mutegi, C.; Atehnkeng, J.; Kaptoge, L.; Cotty, P. Biological control of aflatoxins in Africa: Current status and potential challenges in the face of climate change. World Mycotoxin J. 2016, 9, 771–789. [Google Scholar] [CrossRef] [Green Version]
  144. Dorner, J.W. Biological control of aflatoxin contamination of crops. J. Toxicol. Toxin Rev. 2004, 23, 425–450. [Google Scholar] [CrossRef]
  145. Bhatnagar-Mathur, P.; Sunkara, S.; Bhatnagar-Panwar, M.; Waliyar, F.; Sharma, K.K. Biotechnological advances for combating Aspergillus flavus and aflatoxin contamination in crops. Plant Sci. 2015, 234, 119–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Khan, R.; Ghazali, F.M.; Mahyudin, N.A.; Samsudin, N.I.P. Co-Inoculation of aflatoxigenic and non-aflatoxigenic strains of Aspergillus flavus to assess the efficacy of non-aflatoxigenic strains in growth inhibition and aflatoxin B1 reduction. Agriculture 2021, 11, 198. [Google Scholar] [CrossRef]
  147. Zhou, L.; Wei, D.D.; Selvaraj, J.N.; Shang, B.; Zhang, C.S.; Xing, F.G.; Liu, Y. A strain of Aspergillus flavus from China shows potential as a biocontrol agent for aflatoxin contamination. Biocontrol Sci. Technol. 2015, 25, 583–592. [Google Scholar] [CrossRef]
  148. Cotty, P.J. Influence of field application of an aflatoxigenic strain of Aspergillus flavus on the populations of A. flavus infecting cotton bolls and on the aflatoxin content of cottonseed. Phytopathology 1994, 84, 1270–1277. [Google Scholar] [CrossRef]
  149. Abbas, H.K.; Zablotowicz, R.M.; Bruns, H.A.; Abel, C.A. Biocontrol of aflatoxin in corn by inoculation with non-aflatoxigenic Aspergillus flavus isolates. Biocontrol Sci. Technol. 2006, 16, 437–449. [Google Scholar] [CrossRef]
  150. Cardwell, K.F.; Henry, S.H. Risk of exposure to and mitigation of the effect of aflatoxin on human health: A West African example. J. Toxicol. Toxin Rev. 2004, 23, 217–247. [Google Scholar] [CrossRef]
  151. Roze, L.V.; Hong, S.Y.; Linz, J.E. Aflatoxin biosynthesis: Current frontiers. Annu. Rev. Food Sci. Technol. 2013, 4, 293–311. [Google Scholar] [CrossRef]
  152. Pitt, J.I.; Hocking, A.D. Mycotoxins in Australia: Biocontrol of aflatoxin in peanuts. Mycopathologia 2006, 162, 233–243. [Google Scholar] [CrossRef]
  153. Yin, Y.N.; Yan, L.Y.; Jiang, J.H.; Ma, Z.H. Biological control of aflatoxin contamination of crops. J. Zhejiang Univ. Sci. B 2008, 9, 787–792. [Google Scholar] [CrossRef] [PubMed]
  154. Phillips, T.D.; Afriyie-Gyawu, E.; Williams, J.; Huebner, H.; Ankrah, N.A.; Ofori-Adjei, D.; Wang, J.S. Reducing human exposure to aflatoxin through the use of clay: A review. Food Addit. Contam. 2008, 25, 134–145. [Google Scholar] [CrossRef]
  155. Roze, L.V.; Chanda, A.; Wee, J.; Awad, D.; Linz, J.E. Stress-related transcription factor atfB integrates secondary metabolism with oxidative stress response in Aspergilli. J. Biol. Chem. 2011, 286, 35137–35148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Wee, J.; Day, D.M.; Linz, J.E. Effects of zinc chelators on aflatoxin production in Aspergillus parasiticus. Toxins 2016, 8, 171. [Google Scholar] [CrossRef] [Green Version]
  157. Grintzalis, K.; Vernardis, S.I.; Klapa, M.I.; Georgiou, C.D. Role of oxidative stress in sclerotial differentiation and aflatoxin B1 biosynthesis in Aspergillus flavus. Appl. Environ. Microbiol. 2014, 80, 5561–5571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Alinezhad, S.; Kamalzadeh, A.; Shams-Ghahfarokhi, M.; Rezaee, M.B.; Jaimand, K.; Kawachi, M.; Razzaghi-Abyaneh, M. Search for novel antifungals from 49 indigenous medicinal plants: Foeniculum vulgare and Platycladus orientalis as strong inhibitors of aflatoxin production by Aspergillus parasiticus. Ann. Microbiol. 2011, 61, 673–681. [Google Scholar] [CrossRef]
  159. Chang, P.K.; Scharfenstein, L.L.; Mack, B.; Yu, J.; Ehrlich, K.C. Transcriptomic profiles of Aspergillus flavus CA42, a strain that produces small sclerotia, by decanal treatment and after recovery. Fungal Genet. Biol. 2014, 68, 39–47. [Google Scholar] [CrossRef] [PubMed]
  160. Gómez, J.V.; Tarazona, A.; Mateo-Castro, R.; Gimeno-Adelantado, J.V.; Jiménez, M.; Mateo, E.M. Selected plant essential oils and their main active components, a promising approach to inhibit aflatoxigenic fungi and aflatoxin production in food. Food Addit. Contam. A 2018, 35, 1581–1595. [Google Scholar] [CrossRef]
  161. Singh, G.; Bhattacharyya, R.; Das, T.K.; Sharma, A.R.; Ghosh, A.; Das, S.; Jha, P. Crop rotation and residue management effects on soil enzyme activities, glomalin, and aggregate stability under zero tillage in the Indo-Gangetic Plains. Soil Till. Res. 2018, 184, 291–300. [Google Scholar] [CrossRef]
  162. Kluge, E.R.; Mendes, M.C.; Faria, M.V.; Santos, L.A.; Santos, H.O.D.; Szeuczuk, K. Effect of foliar fungicide and plant spacing on the expression of lipoxygenase enzyme and grain rot in maize hybrids. Acta Sci. Agron. 2017, 39, 407–415. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Organization of gene clusters of AFs’ biosynthesis pathway [21].
Figure 1. Organization of gene clusters of AFs’ biosynthesis pathway [21].
Jof 07 00606 g001
Figure 2. Biosynthetic pathway of AFs [22,23].
Figure 2. Biosynthetic pathway of AFs [22,23].
Jof 07 00606 g002
Figure 3. Genetic regulation of AF biosynthesis.
Figure 3. Genetic regulation of AF biosynthesis.
Jof 07 00606 g003
Figure 4. Different upstream elements affecting the AF/ST gene cluster (source: the author).
Figure 4. Different upstream elements affecting the AF/ST gene cluster (source: the author).
Jof 07 00606 g004
Figure 5. The velvet complex model (source: the author).
Figure 5. The velvet complex model (source: the author).
Jof 07 00606 g005
Figure 6. Ppo effects on ST production (source: the author).
Figure 6. Ppo effects on ST production (source: the author).
Jof 07 00606 g006
Figure 7. Ecological factors affecting AF/ST production. Dots signify links that have yet to be proven (source: the author).
Figure 7. Ecological factors affecting AF/ST production. Dots signify links that have yet to be proven (source: the author).
Jof 07 00606 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Khan, R.; Ghazali, F.M.; Mahyudin, N.A.; Samsudin, N.I.P. Aflatoxin Biosynthesis, Genetic Regulation, Toxicity, and Control Strategies: A Review. J. Fungi 2021, 7, 606. https://doi.org/10.3390/jof7080606

AMA Style

Khan R, Ghazali FM, Mahyudin NA, Samsudin NIP. Aflatoxin Biosynthesis, Genetic Regulation, Toxicity, and Control Strategies: A Review. Journal of Fungi. 2021; 7(8):606. https://doi.org/10.3390/jof7080606

Chicago/Turabian Style

Khan, Rahim, Farinazleen Mohamad Ghazali, Nor Ainy Mahyudin, and Nik Iskandar Putra Samsudin. 2021. "Aflatoxin Biosynthesis, Genetic Regulation, Toxicity, and Control Strategies: A Review" Journal of Fungi 7, no. 8: 606. https://doi.org/10.3390/jof7080606

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