Next Article in Journal
Snake Venomics and Antivenomics of Cape Cobra (Naja nivea) from South Africa: Insights into Venom Toxicity and Cross-Neutralization Activity
Previous Article in Journal
Combining mKate2-Kv1.3 Channel and Atto488-Hongotoxin for the Studies of Peptide Pore Blockers on Living Eukaryotic Cells
Previous Article in Special Issue
Possible Reaction Mechanisms Involved in Degradation of Patulin by Heat-Assisted Cysteine under Highly Acidic Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crosstalk between Mycotoxins and Intestinal Microbiota and the Alleviation Approach via Microorganisms

1
Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
2
Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
*
Author to whom correspondence should be addressed.
Toxins 2022, 14(12), 859; https://doi.org/10.3390/toxins14120859
Submission received: 31 October 2022 / Revised: 30 November 2022 / Accepted: 2 December 2022 / Published: 6 December 2022

Abstract

:
Mycotoxins are secondary metabolites produced by fungus. Due to their widespread distribution, difficulty in removal, and complicated subsequent harmful by-products, mycotoxins pose a threat to the health of humans and animals worldwide. Increasing studies in recent years have highlighted the impact of mycotoxins on the gut microbiota. Numerous researchers have sought to illustrate novel toxicological mechanisms of mycotoxins by examining alterations in the gut microbiota caused by mycotoxins. However, few efficient techniques have been found to ameliorate the toxicity of mycotoxins via microbial pathways in terms of animal husbandry, human health management, and the prognosis of mycotoxin poisoning. This review seeks to examine the crosstalk between five typical mycotoxins and gut microbes, summarize the functions of mycotoxins-induced alterations in gut microbes in toxicological processes and investigate the application prospects of microbes in mycotoxins prevention and therapy from a variety of perspectives. The work is intended to provide support for future research on the interaction between mycotoxins and gut microbes, and to advance the technology for preventing and controlling mycotoxins.
Key Contribution: Cross-talk between mycotoxins and bacteria is a massive and complex subject that requires extensive research. This review summarizes the research on the crosstalk between mycotoxins and microbiota in recent years and provides new ideas for controlling the harm of mycotoxins by micro-organisms.

1. Introduction

Mycotoxin is a naturally occurring substance produced by fungi. Consumption of low concentrations of mycotoxins in animals would result in severe hazardous symptoms [1]. The first credible evidence of the hazardous of mycotoxins effects dates back to the 11th century when ergot intoxication caused widespread human and animal poisoning and even mortality in Europe [2]. With detection and analysis technology advances, hundreds of mycotoxins have been discovered [3]. Five of these mycotoxins, aflatoxin B1 (AFB1), deoxynivalenol (DON), zearalenone (ZEA), fumonisin B1 (FB1), and ochratoxin A (OTA), have historically been major objects of mycotoxin study because of their high detection rates and significant toxicities in feed raw materials and foodstuffs [4]. Due to the extensive prevalence of fungi in the environment, grains have been contaminated with mycotoxins during the growth process, and almost all agricultural commodities are susceptible to fungi infection and the production of mycotoxins if improper storage [5]. Mycotoxins not only lower livestock productivity and result in significant economic losses, but also pose a threat to human health because of their accumulation along the food chain. Severe clinical symptoms occur during animal breeding with mycotoxin-contaminated feed, including diarrhea, liver and kidney damage, pulmonary edema, vomiting, bleeding, and tumors [6,7,8,9,10,11]. Additionally, mycotoxins have a synergistic effect, typically including a combination of toxins, which results in human and animal poisoning upon consumption [12]. This complicates the toxicological mechanism of mycotoxins.
The gut serves as the first line of defense and protection against mycotoxins and the first location of mycotoxins’ absorption into the body [13,14]. The gut microbiota plays a critical role in forming the intestinal barrier and maintaining intestinal homeostasis [15]. With the introduction of new concepts such as the brain-gut axis, liver-gut axis, and kidney-gut axis [16,17,18,19], as well as the widespread use and advancement of microbial sequencing technology, an increasing number of studies are focusing on the impact of mycotoxins in intestinal microbiota. Meanwhile, scientists are investigating the role of intestinal microbial changes in the process of mycotoxin poisoning and detoxification. The deleterious effects conferred by mycotoxins-induced microbial alterations would be brought to the forefront for investigating toxicological effects and response of the host.
Interpreting the crosstalk between various mycotoxins and microbes will be crucial for mycotoxin control and prevention. The crosstalk mechanisms between AFB1, DON, ZEA, FB1 and OTA with intestinal microbes are described in this review. The potential for microbial applications in mycotoxin hazardous mitigation is also discussed with present viewpoints. This paper aims to shed light on the interaction between various mycotoxins and microbes, discover new toxicological processes for mycotoxins, and identify potential treatment targets.

2. Toxicity of Mycotoxins to Intestinal Epithelial Cells

As the “transit point” of animals ingesting mycotoxins, intestinal epithelial cells are the first barrier where mycotoxins contact the animal body [14]. Numerous animal studies have demonstrated that mycotoxins such as DON, ZEA, FB, OTA and AFB1 trigger direct epithelial cell damage (Table 1). The most noticeable symptom is that mycotoxin directly limits the growth and structural destruction of the small intestinal villi, the outer wall consisting of a single layer of epithelial cells [20,21,22,23]. Recently, studies on the mechanism of directive injury of mycotoxins on the intestinal epithelium were well investigated. By studying changes in the physiological functions of intestinal cells exposed to mycotoxins, researchers initially explored the direct toxicological effects of mycotoxins in the absence of gut microbiota and have achieved considerable progress.

2.1. DON

The disclosure of the direct toxicological effects of DON on the intestinal epithelium has made great progress, and a systematic explanation mechanism has been constructed from multiple perspectives. It is assumed that the detrimental effects of DON on intestinal epithelial cells are mediated predominantly through the following three pathways: (Ι) DON activates DAO, up-regulates NF-κB signaling pathway, increases the levels of inflammatory cytokines in the intestine, and ultimately mediates intestinal epithelial apoptotic [24,25,26]. (II) DON decreases the expression of the trefoil factor family (TFFs) of peptides (a type of bioactive substance that could govern tissue regeneration, improve barrier function, and decrease proinflammatory expression) via triggering the MAPK signaling pathway, hence inhibiting intestinal epithelial cell self-repair. This mechanism has been validated in human intestinal cell line HT29-16E and swine intestinal explants [24,27]. (III) Recently, a new perspective on the enterotoxicity of DON has also been proposed. It was revealed that DON can also decrease the stability of intercellular compact proteins in the intestinal epithelium [28]. Along with accelerating tight junction protein degradation in the fusion medium, DON would activate the p38 (MAPK) signaling pathway, resulting in the swallowing and degradation of Occluding and ZO-1 in lysozyme, and eventually destroying the small intestine villus structure and increasing intestinal permeability. Collectively, DON can activate immune pathways and induce inflammatory responses in the intestinal epithelium. And it can activate lysosomes to engulf connexins between intestinal epithelial cells, resulting in structural collapse. In addition, DON also inhibits the self-repair process of the intestine, which eventually leads to the death and autophagy of intestinal epithelial cells.

2.2. ZEA

ZEA generates oxidative stress at the cellular level and increases lactate dehydrogenase activity, further impairing the organism’s scavenging of reactive oxygen species (ROS) and increasing the amount of oxidative stress in intestinal cells [29,30]. In addition, ZEA also mediates the activation of NLRP3 inflammasome in mouse intestine, which in turn promotes the level of Caspase-1, up-regulates inflammatory cytokines, leads to the expansion of inflammatory cells in intestinal epithelial cells, and produces apoptosis [31]. On the other hand, ZEA also leads to aberrant G2/M transition in IPEC-J2 cells by disrupting the cell cycle signaling system, consequently reducing cell proliferation and producing intestinal epithelial damage [32]. Intestinal epithelial cells have the shortest cell cycle and are the fastest-renewing somatic cells. Therefore, the perturbation of cell cycle signals by ZEA will greatly inhibit the development and self-repair process of the intestinal epithelium.

2.3. OTA

The mechanism of OTA cytotoxicity in the intestinal epithelium is mostly based on the production of ROS and the stimulation of apoptosis-regulating genes. Wang et al. [33] found that OTA may generate reactive oxygen species (ROS) in IPEC-J2 cells, which elevated the activity of the Ca2+ and MLCK Signaling pathways and ultimately resulted in barrier malfunction and destruction. Comparative transcriptomics demonstrated that OTA enhanced the expression of apoptosis-related genes such as casp3, cdc25B and egr1 in Caco-2 cells, elucidated the genome-wide biological reaction perspective of OTA controlling intestinal epithelial damage [34]. The researchers also explained the toxicological mechanism of OTA on intestinal epithelial cells from various aspects. It is worth noting that OTA appears to be a dose-dependent amplifier of apoptotic signaling to intestinal exposed-epithelial cells. OTA showed a perturbation of functional gene expression of human intestinal cells at a very low dose (0.0005 μg/mL).

2.4. FB1

FB1 regulates the secretion of mucin by stimulating the ERK phosphorylation pathway, lowering the expression level of intestinal tight junction protein, and inhibiting the viability of IPEC-J2 cells [35]. Mucin secreted by intestinal epithelial goblet cells is a glycoprotein composed of mucopolysaccharides that protects intestinal cells. The network of mucins makes it difficult for chemical irritants, digested foods, toxins, and bacteria to pass through, protects the intestinal epithelium from damage, and prevents pathogens from binding to the intestinal epithelium. The digestion of mucin by FB1 is likely to be an important inducement for it to enter the body and cause multi-organ toxicity. It is worth noting that FB1 could regulate the mouse intestinal aryl hydrocarbon receptor (AHR), the constitutive androstane receptor (CAR), the pregnane X receptor (PXR), and downstream target genes (CYP450s) to disrupt nuclear xenobiotic receptor (NXR) homeostasis, and meanwhile induce intestinal villus and epithelial layer shedding, intestinal gland atrophy, and necrosis [36].

2.5. AFB1

There are few studies on the mechanism of AFB1 intestinal epithelial toxicity. By observing the ultrastructure of intestinal epithelial cells exposed to AFB1, the researchers found that AFB1 is significantly toxic to the organelles of intestinal epithelial cells. AFB1 causes mitochondrial vacuolization in small intestinal cells, leads to the disappearance of mitochondrial cristae, junctional complexes, and terminal reticulum, and then induces apoptosis [37]. Notably, AFB1 also reduced the ratio of goblet cells in epithelial cells in this study. This result indicates that FB1 and AFB1 also seem to have certain effects on the differentiation of intestinal epithelial cells, but whether the underlying mechanism is the toxic effect on goblet cells or the direct induction of the cell differentiation process remains to be further investigated. In addition, researchers have revealed that AFBI exposure increases p42/44 (MAPK) phosphorylation in Caco-2 cells, inhibiting tight junction protein synthesis between epithelial cells, increasing intestinal permeability, and weakening the intestinal barrier [38].
In conclusion, based on the results of direct mycotoxin exposure assays on intestinal epithelial cells, mycotoxins usually directly inhibit the normal division and proliferation of intestinal epithelial cells and cause apoptosis through modulation of cell signaling pathways.

3. Crosstalk between Mycotoxins and Intestinal Microbiota

Regarding the critical function of the gut microbiota in maintaining gut homeostasis, the enterotoxicity of mycotoxins in animals is inseparable from the interaction with the intestinal microbiota. In recent years, increasing experiments have focused on gut microbiota changes under the stress of mycotoxin exposure (Table 2). Mycotoxins have been proven to disrupt intestinal microbiota community homeostasis, and the underlying mechanisms have been gradually explored.

3.1. DON

It is now widely recognized that DON and OTA can exacerbate harmful effects by altering intestinal bacteria. Exposure to DON at either long-term low-dose or short-term medium-to-high doses significantly perturbed the gut microbial composition of animals. When using pigs as experimental animal models, DON can directly affect the variety of gut microbiota. In the short term, the composition of fecal bacteria may be drastically altered [39,40]. In addition to perturbing the gut microbial community as a whole, DON operates synergistically with pathogenic bacteria, aggravating the disease process in the host. Ruhnau et al. [41] proved that DON collaborated with Campylobacter jejuni to enhance intestinal load, disrupt the intestinal barrier, and expedite intestinal pathogen translocation into the liver and kidney. In addition, DON is also demonstrated to enhance the relative abundance of Clostridium perfringens in the jejunum of broilers, thereby increasing the digestive load [42]. Meanwhile, DON enhanced the abundance of pathogenic Escherichia coli in the colon and exacerbated the Escherichia-induced intestinal cell DNA damage [43,44]. Metagenomic analysis revealed that DON severely disrupted the intestinal microbiota in mice and impaired biosynthesis and repair functions. A high dose of DON damages the nerve sphingolipid, protein digestion and absorption, and lipoic acid metabolism pathways [45].

3.2. OTA

Regarding the perturbing effect of OTA on gut microbes, many studies have shown that OTA had a substantial effect on the relative number of Firmicutes, increased the relative abundance of Bacteroides, and decreased the β diversity of the intestinal microbiota [46]. Notably, Wang et al. [47] explored the OTA-induced alterations via intestinal microbiota in its toxicological mechanism and estimated that OTA-mediated liver inflammation was microbially dependent. OTA increased the relative abundance of lipopolysaccharide-producing bacteria (Bacteroidetes), elevated the intestinal LPS load, weakened the intestinal barrier, and ultimately induced liver inflammation via LPS-specific activation of the liver TLR4/MyD88 signaling pathway. The researchers constructed a pseudo-sterile animal model with antibiotics, and the fecal bacteria transplantation experiment proved that OTA-induced changes in gut microbes are an important incentive for OTA-induced liver validation.

3.3. ZEA

When animals were exposed to ZEA alone, no substantial changes in their gut bacteria occurred, and ZEA had no discernible influence on the composition of intestinal microbes. By stimulating the RhoA/ROCK pathway, ZEA tended to inhibit microbial involvement in intestinal mucin formation [48,49,50]. These results suggest that ZEA may have little effect on the overall microbial community in the gut, but through its effect on the mucus layer of the intestinal epithelium, it is likely to have some effects on the parasitic bacteria that adhere to mucin or decompose mucin. This series of changes may further negatively affect the construction of the intestinal barrier. Studies have shown that ZEA has a significant impact on the overall functional gene level of gut microbes. ZEA inhibits intestinal microorganism glycerophospholipid metabolism, which may be one of the reasons contributing to ZEA-induced ovarian reproductive damage [51,52].
The combined presence of more than two mycotoxins frequently occurs in naturally moldy cereals and grains. Therefore, it is more applicable for investigation in combined mycotoxins impact on gut microbiota. As ZEA combined with DON, the organization of the intestinal microbiota was drastically altered [53]. High doses of DON and ZEA changed the number of proteins involved in microbial metabolism, genetic processing, and oxidative stress responses (related to the ribosome and pentose phosphate pathways), resulting in structural modification of the gut microbiome [54]. Additionally, combined ZEA and DON suppressed costimulatory molecule expression on intestinal CD4+ T cells and il-4R-mediated Th2 cell development, hence impairing intestinal resistance and pathogen clearance [56]. These results reflected the additive effect among mycotoxins, whereas, in reality, multiple mycotoxins are often combined and generate in grains and feeds. ZEA has a high detection rate in grains, therefore the combined effects of other toxins and ZEA, and the harmful effects of other mixed mycotoxins on intestinal microbes are urgently needed to advance research.

3.4. AFB1

AFB1 exposure also induces gut microbial dysfunction. Fermentation of carbohydrates and proteins and amino acids to produce short-chain fatty acids (SCFA) is an important function of intestinal microorganisms. SCFA produced by intestinal microorganisms is not only an important source of energy for intestinal cells but also drives the absorption of nutrients and hormone production in the intestine. After four weeks of AFB1-exposed, SCFA, pyruvate-related pathways, amino acids, bile acids, and long-chain fatty acid metabolic pathways were all eliminated in the intestinal bacteria of rats. The efficiency of the host digestive system, energy supply, intestinal immunity, neurotransmitter synthesis, and enterohepatic crosstalk was also affected [55]. When studying the hazards of exposure to mycotoxins, researchers tend to overlook this indirect effect of gut microbial dysbiosis. Little is known about changes in gut microbial communities under mycotoxin exposure. As an important “organ” that performs host digestion, metabolism, absorption, and immune processes, the gut microbiome deserves more attention. There is little experimental evidence on the changes of intestinal microorganisms in animals exposed to FB1, therefore we do not summarize FB1 related information in this part. With the increasing amount of research focusing on changes in the gut microbiome upon exposure to mycotoxins, the understanding of the role of the gut microbial community under toxin perturbation will become improved. This will also contribute to a more complete revelation of the underlying toxicological mechanisms of mycotoxins.

4. Alleviate Mycotoxins Harm by Microbiota

As the crosstalk between mycotoxins and animal gut microbiota discussed, mycotoxin-induced damage in the host organism is partially attributed to microbiota-mediated enterotoxicity and pathological changes. Thus, strengthening the intestinal barrier by modulation of the gut microbiota may represent a unique strategy for minimizing the harm caused by mycotoxins.
At the present, there are three primary strategies for mitigating the harm caused by mycotoxins transmitted by microorganisms: (Ι) directly degrade mycotoxins in food through microbial pretreatment and reduce toxin intake; (II) by increasing probiotic colonization, the formation of bacterial-toxin complexes is inhibited, thereby reducing toxin absorption in the intestinal tract; and (III) by regulating the intestinal microecology with probiotics or prebiotics, remodeling the intestinal microflora, enhancing the intestinal barrier, relieving intestinal toxicity, and reducing toxin penetration.
As several recent pieces of literature reviewed the mycotoxin degradation by microorganisms, we summarized the topic of mycotoxin degradation by microbiota in Section 4.1. This review focused here on the alleviation of mycotoxins in the host by reshaping and modulating intestinal microorganisms via microbiota adhesion effect or supplemental nutrients etc.

4.1. Alleviate Mycotoxins Harm by Microbiota Degradation

With the development and utilization of microbial culturomics and sequencing technology, an increasing number of microorganisms may be cultured individually in the laboratory. Recently, an increasing number of microorganisms have been shown to degrade mycotoxins with high efficiency and specificity.
We list the microbes that can directly and efficiently degrade mycotoxins (Table 3). With the advancement of food fermentation technology and the pre-storage treatment process, it is anticipated that the direct elimination of toxins by microorganisms will become increasingly prevalent.
While microbial degradation of mycotoxins has the advantage of being highly efficient and specific, it also has drawbacks, including limited degradation efficiency of mycotoxins complexes, severe pretreatment conditions, and complex fermentation products.

4.2. Alleviate Mycotoxins Harm by Microbiota Adhesion Effect

Mycotoxins in feed and food are typically in the form of mycotoxins-glycoconjugates generated from plants. These conjugates break down hardly in vivo and are often not digested in the stomach or small intestine, resulting in large intestinal colon toxicity. Increased colonization of high-adhesion probiotics results in the formation of bacteria-toxin complexes in the intestinal segment, which effectively reduces toxin absorption in the large intestine [73]. Lactic acid bacteria adsorption capacity for mycotoxins is a prominent study area in recent studies, owing to their distinctive secretory mucin layer with strong adhesion and their ease of cultivation [74].
The adsorption capacity of ZEA by lactic acid bacteria was first reported in 2017 and examined the durability of the ZEA-lactic acid bacteria combination. It was found that ZEA had a 68.2 percent adsorption capacity on lactic acid bacteria and that 15.8 percent of ZEA remained adsorbed in the complex particles after three bleaching cycles. The potential of lactic acid bacteria as ZEA bioactive adsorbents was demonstrated in this study [75]. With the advancement of microbial culturomics, increasing isolated Lactobacillus species were assessed for the adsorption impact, and the FTIR (Fourier Transform Infrared) technique was utilized to determine the mechanism of microbial adsorption. Lactobacillus plantarum has been found to limit the bioavailability of ZEA in the gastrointestinal system by adhesion, hence reducing the genotoxicity and nephrotoxicity of ZEA [76].
ZEA was absorbed by L. plantarum BCC 47723 with the effect of hydrophobic adsorption rather than electrostatic adsorption. The adsorption efficacy of L. plantarum could be increased further by altering the bacterial cell structure via heat treatment [77].
High-performance liquid chromatography analysis indicated that Lactobacillus paracei might decrease ZEA to α-Zol and β-Zol, resulting in less harmful byproducts [78]. Król et al., found that Lactococcus lactis is capable of neutralizing ZEA via bacterial proteins and deprotonated carboxyl groups in peptidoglycans (asparagine and glutamine) [79].
Except for ZEA, lactic acid bacteria were also capable of adsorbing AFB1 and DON [79,80]. It is demonstrated that Lactobacillus Plantarum could adsorb 82% of AFB1 in vitro and that the resulting complex retained high stability after five washes [81]. It was also reported that Lactobacillus para cel live cells bind AFB1 98 percent of the time and dramatically lower AFB1 levels in the serum of rats exposed to the toxin [82]. Lactobacillus para cel LHZ-1, isolated from yogurt, the cell wall had the highest rate of DON absorption, up to 40.7 percent, supporting that the S protein released by the lactobacillus cell wall had a major role in adhesion [83].

4.3. Alleviate Mycotoxins Harm by Shaping the Microbiota

As shown in the table below, many studies have demonstrated the detoxification effect of additional supplemental nutrients such as non-flavonoid polyphenols, flavonoids, dietary fibers, terpenoid carotenoids, and fatty acids on the toxic effects of mycotoxins (Table 4); the crosstalk between the aforementioned substances and gut microbes, however, was not considered. The gut microbiota is an important link that maintains the host’s metabolic function, immune function, and nutrient absorption. Recently, growing studies have revealed the effects of nutrients on gut microbes and entire metabolic processes passing through the digestive tract. Combining the potential mechanisms of nutrients to mitigate mycotoxins hazards, as well as the interactions between nutrients and gut microbes, will help us to clarify the possibility of gut microbes as potential targets for antagonizing mycotoxins.

4.3.1. Non-Flavonoid Phenolic

Polyphenolic compounds (such as curcumin, caffeic acid, resveratrol, etc.) have an intense alleviation effect on damages caused by mycotoxins.
Curcumin is known as a polyphenolic compound with pharmacological activities, such as antioxidative, anti-inflammatory, and antibacterial. Previous studies revealed that curcumin supplementation attenuated liver damage induced by AFB1 exposure in chickens, mice, and rats and reduced liver inflammation in OTA-exposed ducks [84,85,86,87,88]. Increasing pieces of evidence demonstrated that the hepatotoxicity of AFB1 and OTA is microbe-dependent. AFB1 and OTA disrupt animal intestinal microbial homeostasis, increase the relative abundance of lipopolysaccharide-producing Bacteroides, and ultimately induce liver inflammation in animals [47,148]. As a natural herbal polyphenol, curcumin has been reported in several studies to modulate the composition of gut microbes. Curcumin could increase the relative abundance of Lactobacillus in the intestine of mice, and reduce the relative abundance of Shigella Enterobacter and pathogenic Bacteroides, reducing lipopolysaccharide production to alleviate the process of metabolic endotoxemia [149,150]. In addition, curcumin significantly up-regulates the relative abundance of the butyric acid-producing bacteria Butyricicoccus in the rat gut, and the species Butyricicoccus repair intestinal barrier damage and further improve liver lipids by degrading fermented proteins and carbohydrates to produce short-chain fatty acids [151].
As a powerful antioxidant, resveratrol effectively alleviates liver injury in mice exposed to AFB1 and can improve serum immune indexes in rats exposed to ZEA. Notably, multiple studies have demonstrated the protective effect of resveratrol on the gut of weaned piglets exposed to DON. Significantly, resveratrol up-regulated the relative abundance of SCFA-producing bacteria, such as Butyricicoccus, Ruminococcus_1, Roseburia and Adlercreutzia in rat gut, and increased the level of SCFA in feces [152,153]. During the weaning stage of piglets, the level of secondary bile acids in the intestine was up-regulated due to dietary changes, resulting in disturbance of the intestinal microbiota and intestinal barrier [154,155]. Long-term exposure to DON also induces bile duct hyperplasia and tissue damage in piglets [156]. Interestingly, studies have reported that resveratrol could mediate the synthesis and recycling of bile acids by affecting gut microbes. Resveratrol increases the relative abundance of Lactobacillus and Bifidobacterium, thereby increasing the activity of bile salt hydrolase. And reducing the concentration of bile acids in the ileum. Meanwhile, it has also been found to up-regulate the intestinal FXR signaling pathway and enhance the intestinal barrier [157]. Therefore, resveratrol is likely to mediate the repair effect of gut microbes on the gut of weaned piglets exposed to DON.
Caffeic acid is a hydroxycinnamic acid-related organic compound with both phenolic hydroxyl and acrylic functional groups. It possesses antioxidant, immunomodulatory, and anti-inflammatory properties. A recent study showed that caffeic acid alleviates AFB1-induced kidney damage in rats. Dietary supplementation of caffeic acid significantly reduced the relative abundance of Bacteroidetes and Turicibacter and increased the abundance of Alistipes, Dubosiella and Akkermansia [158,159]. Alistipes were shown to produce SCFA, and Akkermansia is a novel probiotic that could effectively reduce the potential risk of colitis by regulating the secretion of mucin [160,161]. Therefore, gut microbiota may be potential targets for caffeic acid to exert its detoxification effect.

4.3.2. Flaudio-Videoonoids

Flaudio-videoonoids substances (e.g., anthocyanin, gallic acid, quercetin and baicalin and their derivatives) are also shown detoxification effects against mycotoxins.
Anthocyanin is a water-soluble plant pigment. Toxicological studies showed that anthocyanins alleviate the damage caused by AFB1 in broilers and the damage caused by ZEA in mice. As polyphenols naturally extracted from plants, anthocyanins not only play an antioxidant role, but also significantly change the composition of animals’ gut microbes. Anthocyanin supplementation reduces intestinal inflammation by increasing the relative abundance of Lactobacillus, Bifidobacterium, Lachnospira and Ruminococcus, which in turn increases SCFA production, improves intestinal barrier and mucus production, and reduces the potential risk of intestinal inflammation [162,163].
Gallic acid is not only a polyphenolic compound present in plants, but also one of the important metabolites of gut microbes [164]. Gallic acid alleviates the reproductive toxicity of ZEA-exposed mice and the toxic effect of OTA on catfish. Adding gallic acid to the diet significantly upregulates gut microbial diversity in dogs and bees [165,166]. Recent studies have shown that ZEA is likely to induce reproductive toxicity by disrupting the blood-testis barrier [167]. The protective effect of probiotics on the blood-testis barrier has gradually been discovered and clarified by researchers. Proteobacteria have been reported to increase the probability of semen hyperviscosity, while Lactobacillus improves mitochondrial activity and alleviates oxidative stress in sperm cells [168]. Interestingly, the relative abundance of probiotics such as Lactobacillus and Prevotaceae were significantly up-regulated and the level of Proteobacteria was significantly decreased after gallic acid was added to the diet of colitis model rats [169]. Therefore, gallic acid is likely to be able to alleviate the reproductive toxicity caused by ZEA by regulating the composition of gut microbes, and its antioxidant properties also resist the oxidative stress caused by ZEA.
In recent studies, researchers have revealed the alleviation effect of quercetin on metabolic diseases such as hyperlipidemia, hyperglycemia, and obesity [170,171,172]. Quercetin also effectively alleviates OTA-induced immune toxicity in chickens and nephrotoxicity in rats exposed to OTA. As gut microbes may be an important bridge for quercetin to regulate energy metabolism disorders, the effect of quercetin on intestinal microbes is also an inevitable consideration in the process of alleviating mycotoxins. Studies have shown that quercetin is effective against microbial dysbiosis, restores gut microbiota in antibiotics-treated mice, and significantly increases the relative abundance of butyrate-producing bacteria [173]. Similarly, a dietary quercetin supplementation in pigs experiment revealed that when quercetin was added to piglet feed, quercetin significantly upregulated Akkermansia muciniphila in the gut, as well as the relative abundance of SCFA-producing bacteria Clostridium butyricum, Clostridium celatum and Prevotella copri [174].
Baicalin is a naturally occurring glycoside that is derived from baicalein and glucuronic acid. Baicalin and its copper-zinc compounds effectively alleviate intestinal inflammation in weaned piglets exposed to DON and protect the liver and kidney toxicity in broilers exposed to ZEA. Baicalin with dietary intake is converted into baicalenin with gut microbes’ fermentation [175,176]. As baicalenin is absorbed into the host mechanism through the intestinal tract, baicalenin and baicalinit could be converted into each other under the catalysis of phase II enzymes, and phase II enzymes also have detoxification effects [177,178]. Therefore, the interaction between baicalin and gut microbes is also an important part of its detoxification effect. Dietary supplementation with baicalin monomer significantly altered gut microbial composition and induced changes in fecal bile acid profiles, according to the findings of the researchers. Moreover, baicalin inhibited the appearance of the bile acid-specific receptor FXR and increased intestinal barrier toxicity resistance [179]. This study accurately depicts the regulatory effect of baicalin on the intestinal microbiota and bile acid system and paves the way for the discovery of a more profound detoxification mechanism.

4.3.3. Dietary Fiber

Many studies have shown that dietary fiber and its metabolites (oligosaccharides) also have a detoxifying effect on mycotoxins. Dietary fiber affects the peristalsis and digestion process of the intestine, and most of them are not digested in the foregut. Dietary fibers are converted into small molecular substances and absorbed by the host after the hindgut degradation and fermentation by intestinal microorganisms [180]. As the main active ingredient in moringa oleifera leaves, moringa oleifera polysaccharide significantly reduced the relative abundance of Bacteroides and Helicobacter pylori in gut microbes [122]. The protective effect of algal polysaccharides, fucoidan, and Alginate oligosaccharides on animal intestines is mainly due to their promotion of SCFA production by bacteria in the hindgut [181]. At present, the interaction effect of polysaccharides and oligosaccharides with intestinal microbes in the hindgut is still unclear. It is necessary to further design sterile or pseudo-sterile animal experiments to verify the role of intestinal microbes in the process of dietary fiber alleviating the toxicity of mycotoxins.

4.3.4. Terpenoid Carotenoids

Studies have shown that two terpenoid carotenoids, lycopene, and astaxanthin, also have a significant detoxification effect on mycotoxins. Lycopene, in particular, has a relieving effect on damage with AFB1, DON and ZEA exposure. Studies show that additional dietary lycopene remodels the gut microbiome of mice with colitis, significantly reducing the relative abundance of Proteobacteria and increasing the relative abundance of Bifidobacterium and Lactobacillus [182]. In other studies, additional supplementation of astaxanthin in feed directly reduces the functional gene abundance that is associated with inflammation in gut microbes [183]. By regulating the abundance of functional gene fragments in intestinal microorganisms, the antagonistic effect of mycotoxin was indirectly achieved.

4.3.5. Fatty Acids

Cinnamic acid and its derivatives, as well as sodium butyrate, have also shown detoxification effects on mycotoxins in a large number of animal tests. Studies in rats and chickens showed that cinnamic acid derivative ferulic acid can alleviate liver damage caused by AFB1 [98]. Laurate supplementation in the feed also shows the alleviation of broiler growth performance by the damage of FB1 [121]. A recent study in DON revealed that sodium butyrate could renovate intestinal barrier damage. SCFA plays a pivotal role in the construction of the intestinal barrier [112]. After the supply of SCFA precursors, the production of SCFA by intestinal microorganisms is also greatly promoted, and the abundance of SCFA-producing bacteria also increased. It is easier to become the dominant flora under this condition, and further, exert the biosynthetic function of SCFA.

4.3.6. Microelement

Many studies have shown that mycotoxins can be detoxified well by supplementing feed with the trace element selenium. For poultry exposed to AFB1, supplementation with low doses of selenium is effective in reducing AFB1 damage to the liver and thymus of poultry [101,102]. Either selenium or yeast selenium supplementation helped to alleviate kidney damage in mice exposed to ZEA [129,135]. Yeast selenium has a strong antagonistic effect on both hepatotoxicity and nephrotoxicity in chickens exposed to OTA [145,146]. It has been demonstrated that selenium nanoparticles are effective in suppressing the relative abundance of Enterococcus cecorum in the intestinal tract of chickens [184]. Of note here is the selenium conjugate, yeast, which, as we mentioned earlier, is widely used for the adsorption and degradation of mycotoxins. Thus, in the process of the natural product yeast selenium acting to mitigate the toxicity of mycotoxins, yeast also plays a part in the adsorption and degradation of mycotoxins.

4.4. Microbiota-Guided Direct Regulatory Strategy

In addition to intestinal toxicity, mycotoxins have complicated toxicological pathways that include hepatotoxicity, nephrotoxicity, neurotoxicity, and immunotoxicity. The concepts of the gut-liver axis, gut-brain axis, and gut-kidney axis have been proposed in recent years, prompting researchers to reconsider the role of mycotoxin-mediated intestinal microbiological disorder in extra-intestinal organ injury and pointing to a new direction for elucidating mycotoxins’ novel toxicological mechanism. It can be seen that numerous researchers have attempted to modify the gut microbiota directly through microbial community regulation.
Interestingly, certain lactic acid bacteria have a considerable protective effect against DON-induced intestinal damage. For example, Lactobacillus rhamnosus treatment boosts the liver antioxidant capacity, blocks the NRF2 signaling pathway, and increases butyrate synthesis by up-regulating the Buk and But genes, thereby inhibiting the IRE1/XBP1 signaling pathway and protecting mice from DON damage [185,186]. In vitro studies revealed that Lactobacillus plantarum supernatant was proven to significantly recover the loss of intestinal goblet cells induced by DON, and to strengthen the architecture of intestinal villi [187]. Whereas Lactobacillus plantarum cells have the potential to rearrange the intestinal community homeostasis disrupted by DON, and down-regulate the expression of apoptotic genes to ameliorate intestinal cell death and inflammation generated by DON [188]. Interestingly, Lactobacillus rhamnosus was unable to adsorb DON, but it was able to inhibit the expression of CCL20, IL-1β, TNFα, IL-8, IL-22 and IL-10 via MAP kinase and therefore withstand the intestinal inflammation generated by DON [189]. Notably, several probiotics were utilized to mitigate the toxicity of DON, and the effect of mending the intestinal barrier and restoring phylum-level bacteria abundance was shown in both mouse and piglet models [190,191].
The mechanism by which OTA induces liver inflammation via intestinal bacteria was detailed above, demonstrating that the LPS and TLR4 signaling pathways are critical targets of the OTA toxicological mechanism. Xia et al., revealed that melatonin successfully reversed the OTA-induced increase of Bacteroidetes abundance, greatly lowering LPS accumulation in the gut and liver, and relieving OTA-induced liver inflammation [192]. Additionally, other studies have demonstrated that astaxanthin and selenium-rich yeast can modulate fecal barrier function and the TLR4/MyD88 signaling pathway, block the OTA-mediated NF-κB signaling pathway, reduce OTA-induced intestinal toxicity, and repair the intestinal barrier [193,194]. Transcriptomics results showed that Bacillus subtilis CW14 would activate the toll-like receptor signaling pathway to protect the ZO-1 protein, and minimize OTA-induced cell apoptosis by down-regulating the death receptor gene and up-regulating the DNA repair gene [195].
There is still a dearth of investigation of precise pathogenic pathways related to gut microorganisms of AFB1 and ZEA. As a result, the prevention and treatment of these two mycotoxins are primarily focused on intestinal barrier development and preservation of gut microbial balance. Chen et al., found that Bacillus amylolitica B10 dramatically increased intestinal tight junction protein expression in mice while decreasing the relative abundance of Bacteroides and Bacteroidetes [196]. Wang et al. demonstrated that Bacillus cereus BC7 effectively normalized ZEA-induced disturbances in the intestinal microbiota, and significantly increased lactobacillus abundance for microbiome homeostasis in mice, thereby reversing abnormal histological phenotypes in the uterus, ovaries, and liver exposed to ZEA [197].

5. Conclusions

Cross-talk between mycotoxins and bacteria is complicated, and requires systematic and extensive studies. Researchers have finally been able to decipher this mysterious black box because of advancements in microbiome culture and sequencing technology. It is through the digestive system that mycotoxins enter the body, and the intestinal microbe is the primary vector for the crosstalk effect that occurs with them. In addition to broadening our horizons in understanding the toxicological mechanisms of mycotoxins, investigating the cross-talk relationship between mycotoxins and microorganisms can also provide us with more valuable and potential therapeutic targets that alleviate the toxic effects of mycotoxins. As the depth and breadth of research increases, more mechanisms to mitigate mycotoxicosis through microorganisms will be revealed and applied to bioengineering production and animal husbandry.

Author Contributions

Conceptualization, W.W.; writing—original draft, W.W. and D.X.; writing—review and editing, W.W., D.X. and Q.M.; project administration, W.W. and L.Y.; funding acquisition, W.W. and L.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Guangdong Province Natural Science Funds for Distinguished Young Scholar (2022B1515020016), National Science Fund Project of China (32072751), National Key Research Program (2021YFD1300404), The National Science Fund for Outstanding Young Scholars (32222080), Natural Science Foundation of Guangdong Province (2019B1515210012), China Agriculture Research System (CARS-42-15), Modern Agricultural Industrial Technology System Innovation Team of Guangdong Province (2021KJ137).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bills, G.F.; Gloer, J.B. Biologically active secondary metabolites from the fungi. Microbiol. Spectr. 2016, 4, 6. [Google Scholar] [CrossRef] [PubMed]
  2. Bonnichon, P.; Graff, J.P. Petit Saint Antoine, Parisian hospice for the treatment of ergotism from the 11th to the 16th Century. Hist. Sci. Med. 2010, 44, 405–408. [Google Scholar] [PubMed]
  3. Du, K.; Wang, C.; Liu, P.; Li, Y.; Ma, X. Effects of dietary mycotoxins on gut microbiome. Protein Pept. Lett. 2017, 24, 397–405. [Google Scholar] [CrossRef]
  4. Streit, E.; Naehrer, K.; Rodrigues, I.; Schatzmayr, G. Mycotoxin occurrence in feed and feed raw materials worldwide: Long-term analysis with special focus on Europe and Asia. J. Sci. Food Agric. 2013, 93, 2892–2899. [Google Scholar] [CrossRef]
  5. Pinotti, L.; Ottoboni, M.; Giromini, C.; Dell’Orto, V.; Cheli, F. Mycotoxin contamination in the EU feed supply chain: A focus on cereal byproducts. Toxins 2016, 8, 45. [Google Scholar] [CrossRef] [Green Version]
  6. Gelderblom, W.C.; Kriek, N.P.; Marasas, W.F.; Thiel, P.G. Toxicity and carcinogenicity of the Fusarium moniliforme metabolite, fumonisin B1, in rats. Carcinogenesis 1991, 12, 1247–1251. [Google Scholar] [CrossRef] [PubMed]
  7. Hoffmann, V.; Jones, K.; Leroy, J. Mitigating aflatoxin exposure to improve child growth in Eastern Kenya: Study protocol for a randomized controlled trial. Trials 2015, 16, 552. [Google Scholar] [CrossRef] [Green Version]
  8. Nejdfors, P.; Ekelund, M.; Jeppsson, B.; Weström, B.R. Mucosal in vitro permeability in the intestinal tract of the pig, the rat, and man: Species- and region-related differences. Scand. J. Gastroenterol. 2000, 35, 501–507. [Google Scholar] [CrossRef] [PubMed]
  9. Pestka, J.J. Deoxynivalenol: Mechanisms of action, human exposure, and toxicological relevance. Arch. Toxicol. 2010, 84, 663–679. [Google Scholar] [CrossRef]
  10. Stockmann-Juvala, H.; Savolainen, K. A review of the toxic effects and mechanisms of action of fumonisin B1. Hum. Exp. Toxicol. 2008, 27, 799–809. [Google Scholar] [CrossRef]
  11. Zinedine, A.; Soriano, J.M.; Moltó, J.C.; Mañes, J. Review on the toxicity, occurrence, metabolism, detoxification, regulations and intake of zearalenone: An oestrogenic mycotoxin. Food Chem. Toxicol. 2007, 45, 1–18. [Google Scholar] [CrossRef] [PubMed]
  12. Peraica, M.; Radić, B.; Lucić, A.; Pavlović, M. Toxic effects of mycotoxins in humans. Bull. World Health Organ. 1999, 77, 754–766. [Google Scholar] [PubMed]
  13. Adegbeye, M.J.; Reddy, P.; Chilaka, C.A.; Balogun, O.B.; Elghandour, M.; Rivas-Caceres, R.R.; Salem, A. Mycotoxin toxicity and residue in animal products: Prevalence, consumer exposure and reduction strategies—A review. Toxicon 2020, 177, 96–108. [Google Scholar] [CrossRef] [PubMed]
  14. Liew, W.P.; Mohd-Redzwan, S. Mycotoxin: Its Impact on Gut Health and Microbiota. Front. Cell. Infect. Microbiol. 2018, 8, 60. [Google Scholar] [CrossRef] [Green Version]
  15. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 61, 1. [Google Scholar] [CrossRef] [Green Version]
  16. Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [Green Version]
  17. Chen, Y.Y.; Chen, D.Q.; Chen, L.; Liu, J.R.; Vaziri, N.D.; Guo, Y.; Zhao, Y.Y. Microbiome-metabolome reveals the contribution of gut-kidney axis on kidney disease. J. Transl. Med. 2019, 17, 5. [Google Scholar] [CrossRef] [Green Version]
  18. Quigley, E. Microbiota-Brain-Gut Axis and Neurodegenerative Diseases. Curr. Neurol. Neurosci. Rep. 2017, 17, 94. [Google Scholar] [CrossRef]
  19. Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
  20. Tomaszewska, E.; Rudyk, H.; Dobrowolski, P.; Donaldson, J.; Świetlicka, I.; Puzio, I.; Kamiński, D.; Wiącek, D.; Kushnir, V.; Brezvyn, O.; et al. Changes in the Intestinal Histomorphometry, the Expression of Intestinal Tight Junction Proteins, and the Bone Structure and Liver of Pre-Laying Hens Following Oral Administration of Fumonisins for 21 Days. Toxins 2021, 13, 375. [Google Scholar] [CrossRef]
  21. Feng, G.D.; He, J.; Ao, X.; Chen, D.W. Effects of maize naturally contaminated with aflatoxin B1 on growth performance, intestinal morphology, and digestive physiology in ducks. Poult. Sci. 2017, 96, 1948–1955. [Google Scholar] [CrossRef] [PubMed]
  22. Ricci, F.G.; Terkelli, L.R.; Venancio, E.J.; Justino, L.; Dos Santos, B.Q.; Baptista, A.; Oba, A.; de Oliveira Souza, B.D.; Bracarense, A.; Hirooka, E.Y.; et al. Tryptophan Attenuates the effects of OTA on intestinal morphology and local IgA/IgY production in broiler chicks. Toxins 2020, 13, 5. [Google Scholar] [CrossRef]
  23. de Souza, M.; Baptista, A.; Valdiviezo, M.; Justino, L.; Menck-Costa, M.F.; Ferraz, C.R.; da Gloria, E.M.; Verri, W.A.; Bracarense, A., Jr. Lactobacillus spp. reduces morphological changes and oxidative stress induced by deoxynivalenol on the intestine and liver of broilers. Toxicon. 2020, 185, 203–212. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, S.; Zhang, C.; Wang, X.; Yang, J.; Wu, K.; Zhang, J.; Zhang, B.; Yang, A.; Qi, D. Deoxynivalenol Inhibits Porcine Intestinal Trefoil Factors Expression in Weanling Piglets and IPEC-J2 Cells. Toxins 2019, 11, 670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wang, X.C.; Zhang, Y.F.; Cao, L.; Zhu, L.; Huang, Y.Y.; Chen, X.F.; Chu, X.Y.; Zhu, D.F.; Ur Rahman, S.; Feng, S.B.; et al. Deoxynivalenol Induces Intestinal Damage and Inflammatory Response through the Nuclear Factor-κB Signaling Pathway in Piglets. Toxins 2019, 11, 663. [Google Scholar] [CrossRef] [Green Version]
  26. Dąbrowski, M.; Jakimiuk, E.; Gajęcka, M.; Gajęcki, M.T.; Zielonka, Ł. Effect of deoxynivalenol on the levels of toll-like receptors 2 and 9 and their mRNA expression in enterocytes in the porcine large intestine: A preliminary study. Pol. J. Vet. Sci. 2017, 20, 213–220. [Google Scholar] [CrossRef] [Green Version]
  27. Graziani, F.; Pinton, P.; Olleik, H.; Pujol, A.; Nicoletti, C.; Sicre, M.; Quinson, N.; Ajandouz, E.H.; Perrier, J.; Pasquale, E.D.; et al. Deoxynivalenol inhibits the expression of trefoil factors (TFF) by intestinal human and porcine goblet cells. Arch. Toxicol. 2019, 93, 1039–1049. [Google Scholar] [CrossRef]
  28. Li, E.; Horn, N.; Ajuwon, K.M. Mechanisms of deoxynivalenol-induced endocytosis and degradation of tight junction proteins in jejunal IPEC-J2 cells involve selective activation of the MAPK pathways. Arch. Toxicol. 2021, 95, 2065–2079. [Google Scholar] [CrossRef]
  29. Shen, T.; Miao, Y.; Ding, C.; Fan, W.; Liu, S.; Lv, Y.; Gao, X.; De Boevre, M.; Yan, L.; Okoth, S.; et al. Activation of the p38/MAPK pathway regulates autophagy in response to the CYPOR-dependent oxidative stress induced by zearalenone in porcine intestinal epithelial cells. Food Chem. Toxicol. 2019, 131, 110527. [Google Scholar] [CrossRef]
  30. Gu, A.; Yang, L.; Wang, J.; Li, J.; Shan, A. Protective effect of glutamine and alanyl-glutamine against zearalenone-induced intestinal epithelial barrier dysfunction in IPEC-J2 cells. Res. Vet. Sci. 2021, 137, 48–55. [Google Scholar] [CrossRef]
  31. Fan, W.; Lv, Y.; Ren, S.; Shao, M.; Shen, T.; Huang, K.; Zhou, J.; Yan, L.; Song, S. Zearalenone (ZEA)-induced intestinal inflammation is mediated by the NLRP3 inflammasome. Chemosphere 2018, 190, 272–279. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, X.; Yu, H.; Fang, H.; Zhao, Y.; Jin, Y.; Shen, J.; Zhou, C.; Zhou, Y.; Fu, Y.; Wang, J.; et al. Transcriptional profiling of zearalenone-induced inhibition of IPEC-J2 cell proliferation. Toxicon 2019, 172, 8–14. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, H.; Zhai, N.; Chen, Y.; Fu, C.; Huang, K. OTA induces intestinal epithelial barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca2+-mediated MLCK activation. Environ. Pollut. 2018, 242, 106–112. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, X.; Gao, Y.; Yan, Q.; Bao, X.; Zhao, S.; Wang, J.; Zheng, N. Transcriptome Analysis of Ochratoxin A-Induced Apoptosis in Differentiated Caco-2 Cells. Toxins 2019, 12, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Chen, Z.; Chen, H.; Li, X.; Yuan, Q.; Su, J.; Yang, L.; Ning, L.; Lei, H. Fumonisin B (1) damages the barrier functions of porcine intestinal epithelial cells in vitro. J. Biochem. Mol. Toxicol. 2019, 33, e22397. [Google Scholar] [CrossRef]
  36. Li, X.; Cao, C.; Zhu, X.; Li, X.; Wang, K. Fumonisins B1 exposure triggers intestinal tract injury via activating nuclear xenobiotic receptors and attracting inflammation response. Environ. Pollut. 2020, 267, 115461. [Google Scholar] [CrossRef]
  37. 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]
  38. Gao, Y.; Li, S.; Wang, J.; Luo, C.; Zhao, S.; Zheng, N. Modulation of Intestinal Epithelial Permeability in Differentiated Caco-2 Cells Exposed to Aflatoxin M1 and Ochratoxin A Individually or Collectively. Toxins 2017, 10, 13. [Google Scholar] [CrossRef] [Green Version]
  39. Vignal, C.; Djouina, M.; Pichavant, M.; Caboche, S.; Waxin, C.; Beury, D.; Hot, D.; Gower-Rousseau, C.; Body-Malapel, M. Chronic ingestion of deoxynivalenol at human dietary levels impairs intestinal homeostasis and gut microbiota in mice. Arch. Toxicol. 2018, 92, 2327–2338. [Google Scholar] [CrossRef]
  40. Le Sciellour, M.; Zemb, O.; Serviento, A.M.; Renaudeau, D. Transient effect of single or repeated acute deoxynivalenol and zearalenone dietary challenge on fecal microbiota composition in female finishing pigs. Animal 2020, 14, 2277–2287. [Google Scholar] [CrossRef]
  41. Ruhnau, D.; Hess, C.; Grenier, B.; Doupovec, B.; Schatzmayr, D.; Hess, M.; Awad, W.A. The mycotoxin Deoxynivalenol (DON) promotes campylobacter jejuni multiplication in the intestine of broiler chickens with consequences on bacterial translocation and gut integrity. Front. Vet. Sci. 2020, 7, 573894. [Google Scholar] [CrossRef] [PubMed]
  42. Guo, F.; Wang, F.; Ma, H.; Ren, Z.; Yang, X.; Yang, X. Study on the interactive effect of deoxynivalenol and Clostridium perfringens on the jejunal health of broiler chickens. Poult. Sci. 2021, 100, 100807. [Google Scholar] [CrossRef] [PubMed]
  43. Lucke, A.; Böhm, J.; Zebeli, Q.; Metzler-Zebeli, B.U. Dietary Deoxynivalenol contamination and oral lipopolysaccharide challenge alters the cecal microbiota of broiler chickens. Front. Microbiol. 2018, 9, 804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Payros, D.; Dobrindt, U.; Martin, P.; Secher, T.; Bracarense, A.P.; Boury, M.; Laffitte, J.; Pinton, P.; Oswald, E.; Oswald, I.P. The food contaminant Deoxynivalenol exacerbates the genotoxicity of gut microbiota. mBio 2017, 8, e00007-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wang, J.J.; Zhang, R.Q.; Zhai, Q.Y.; Liu, J.C.; Li, N.; Liu, W.X.; Li, L.; Shen, W. Metagenomic analysis of gut microbiota alteration in a mouse model exposed to mycotoxin deoxynivalenol. Toxicol. Appl. Pharmacol. 2019, 372, 47–56. [Google Scholar] [CrossRef] [PubMed]
  46. Izco, M.; Vettorazzi, A.; de Toro, M.; Sáenz, Y.; Alvarez-Erviti, L. Oral Sub-chronic Ochratoxin A Exposure Induces Gut Microbiota Alterations in Mice. Toxins 2021, 13, 106. [Google Scholar] [CrossRef]
  47. Wang, W.; Zhai, S.; Xia, Y.; Wang, H.; Ruan, D.; Zhou, T.; Zhu, Y.; Zhang, H.; Zhang, M.; Ye, H.; et al. Ochratoxin A induces liver inflammation: Involvement of intestinal microbiota. Microbiome 2019, 7, 151. [Google Scholar] [CrossRef]
  48. Li, P.; Yang, S.; Zhang, X.; Huang, S.; Wang, N.; Wang, M.; Long, M.; He, J. Zearalenone Changes the Diversity and Composition of Caecum Microbiota in Weaned Rabbit. BioMed Res. Int. 2018, 2018, 3623274. [Google Scholar] [CrossRef] [Green Version]
  49. Zhang, W.; Zhang, S.; Wang, J.; Shan, A.; Xu, L. Changes in intestinal barrier functions and gut microbiota in rats exposed to zearalenone. Ecotoxicol. Environ. Saf. 2020, 204, 111072. [Google Scholar] [CrossRef]
  50. Cieplińska, K.; Gajęcka, M.; Dąbrowski, M.; Rykaczewska, A.; Lisieska-Żołnierczyk, S.; Bulińska, M.; Zielonka, Ł.; Gajęcki, M.T. Time-dependent changes in the intestinal microbiome of gilts exposed to low zearalenone doses. Toxins 2019, 11, 296. [Google Scholar] [CrossRef]
  51. Tan, S.; Ge, W.; Wang, J.; Liu, W.; Zhao, Y.; Shen, W.; Li, L. Zearalenone-induced aberration in the composition of the gut microbiome and function impacts the ovary reserve. Chemosphere 2020, 244, 125493. [Google Scholar] [CrossRef]
  52. Li, Y.; Yi, J.; Zeng, Q.; Liu, Y.; Yang, B.; Liu, B.; Li, Y.; Mehmood, K.; Hussain, R.; Tang, Z.; et al. Zearalenone exposure mediated hepatotoxicity via mitochondrial apoptotic and autophagy pathways: Associated with gut microbiome and metabolites. Toxicology 2021, 462, 152957. [Google Scholar] [CrossRef] [PubMed]
  53. Reddy, K.E.; Kim, M.; Kim, K.H.; Ji, S.Y.; Baek, Y.; Chun, J.L.; Jung, H.J.; Choe, C.; Lee, H.J.; Kim, M.; et al. Effect of commercially purified deoxynivalenol and zearalenone mycotoxins on microbial diversity of pig cecum contents. Anim. Biosci. 2021, 34, 243–255. [Google Scholar] [CrossRef] [PubMed]
  54. Saenz, J.S.; Kurz, A.; Ruczizka, U.; Bünger, M.; Dippel, M.; Nagl, V.; Grenier, B.; Ladinig, A.; Seifert, J.; Selberherr, E.; et al. Metaproteomics Reveals Alteration of the Gut Microbiome in Weaned Piglets Due to the Ingestion of the Mycotoxins Deoxynivalenol and Zearalenone. Toxins 2021, 13, 583. [Google Scholar] [CrossRef] [PubMed]
  55. Zhou, J.; Tang, L.; Wang, J.; Wang, J.S. Aflatoxin B1 Disrupts Gut-Microbial Metabolisms of Short-Chain Fatty Acids, Long-Chain Fatty Acids, and Bile Acids in Male F344 Rats. Toxicol. Sci. 2018, 164, 453–464. [Google Scholar] [CrossRef] [PubMed]
  56. Cai, G.; Liu, S.; Zhong, F.; Gu, J.; Yuan, Y.; Zhu, J.; Zhu, G.; Liu, Z.; Zou, H.; Bian, J. Zearalenone and deoxynivalenol inhibited IL-4 receptor-mediated Th2 cell differentiation and aggravated bacterial infection in mice. Toxicol. Appl. Pharmacol. 2021, 415, 115441. [Google Scholar] [CrossRef]
  57. Xu, L.; Eisa Ahmed, M.F.; Sangare, L.; Zhao, Y.; Selvaraj, J.N.; Xing, F.; Wang, Y.; Yang, H.; Liu, Y. Novel Aflatoxin-Degrading Enzyme from Bacillus shackletonii L7. Toxins 2017, 9, 36. [Google Scholar] [CrossRef]
  58. Ali, S.; Hassan, M.; Essam, T.; Ibrahim, M.A.; Al-Amry, K. Biodegradation of aflatoxin by bacterial species isolated from poultry farms. Toxicon 2021, 195, 7–16. [Google Scholar] [CrossRef]
  59. González Pereyra, M.L.; Di Giacomo, A.L.; Lara, A.L.; Martínez, M.P.; Cavaglieri, L. Aflatoxin-degrading Bacillus sp. strains degrade zearalenone and produce proteases, amylases and cellulases of agro-industrial interest. Toxicon 2020, 180, 43–48. [Google Scholar] [CrossRef]
  60. Pourmohammadi, K.; Sohrabi, M.; Hashemi, S.; Amiri, M.J. A kinetic analysis of the aflatoxin detoxification potential of lactic acid bacteria in Terxine (a cereal-based food). FEMS Microbiol. Lett. 2021, 368, fnab104. [Google Scholar] [CrossRef]
  61. Rodriguez, H.; Reveron, I.; Doria, F.; Costantini, A.; De Las Rivas, B.; Muňoz, R.; Garcia-Moruno, E. Degradation of ochratoxin a by Brevibacterium species 2011. J. Agric. Food Chem. 2011, 59, 10755–10760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Chang, X.; Wu, Z.; Wu, S.; Dai, Y.; Sun, C. Degradation of ochratoxin A by Bacillus amyloliquefaciens ASAG1. Food Addit. Contam. Part A 2015, 32, 564–571. [Google Scholar] [CrossRef]
  63. Shi, L.; Liang, Z.; Li, J.; Hao, J.; Xu, Y.; Huang, K.; Tian, J.; He, X.; Xu, W. Ochratoxin A biocontrol and biodegradation by Bacillus subtilis CW 14. J. Sci. Food Agric. 2014, 94, 1879–1885. [Google Scholar] [CrossRef] [PubMed]
  64. Keawmanee, P.; Rattanakreetakul, C.; Pongpisutta, R. Microbial reduction of fumonisin B1 by the new isolate serratia marcescens 329-2. Toxins 2021, 13, 638. [Google Scholar] [CrossRef] [PubMed]
  65. Deepthi, B.V.; Poornachandra Rao, K.; Chennapa, G.; Naik, M.K.; Chandrashekara, K.T.; Sreenivasa, M.Y. Antifungal attributes of Lactobacillus plantarum MYS6 against Fumonisin producing fusarium proliferatum associated with poultry feeds. PLoS ONE 2016, 11, e0155122. [Google Scholar] [CrossRef] [Green Version]
  66. Nazareth, T.M.; Luz, C.; Torrijos, R.; Quiles, J.M.; Luciano, F.B.; Mañes, J.; Meca, G. Potential application of lactic acid bacteria to reduce Aflatoxin B (1) and Fumonisin B (1) occurrence on corn kernels and corn ears. Toxins 2019, 12, 21. [Google Scholar] [CrossRef] [Green Version]
  67. Martinez Tuppia, C.; Atanasova-Penichon, V.; Chéreau, S.; Ferrer, N.; Marchegay, G.; Savoie, J.M.; Richard-Forget, F. Yeast and bacteria from ensiled high moisture maize grains as potential mitigation agents of fumonisin B (1). J. Sci. Food Agric. 2017, 97, 2443–2452. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, W.C.; Hsu, T.C.; Cheng, K.C.; Liu, J.R. Expression of the Clonostachys rosea lactonohydrolase gene by Lactobacillus reuteri to increase its zearalenone-removing ability. Microb. Cell Fact. 2017, 16, 69. [Google Scholar] [CrossRef]
  69. Liu, F.; Malaphan, W.; Xing, F.; Yu, B. Biodetoxification of fungal mycotoxins zearalenone by engineered probiotic bacterium Lactobacillus reuteri with surface-displayed lactonohydrolase. Appl. Microbiol. Biotechnol. 2019, 103, 8813–8824. [Google Scholar] [CrossRef]
  70. Risa, A.; Krifaton, C.; Kukolya, J.; Kriszt, B.; Cserháti, M.; Táncsics, A. Aflatoxin B1 and Zearalenone-Detoxifying Profile of Rhodococcus Type Strains. Curr. Microbiol. 2018, 75, 907–917. [Google Scholar] [CrossRef]
  71. Shen, W.; Liu, Y.; Zhang, X.; Zhang, X.; Rong, X.; Zhao, L.; Ji, C.; Lei, Y.; Li, F.; Chen, J.; et al. Comparison of Ameliorative Effects between Probiotic and Biodegradable Bacillus subtilis on Zearalenone Toxicosis in Gilts. Toxins 2021, 13, 882. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, J.; Qin, X.; Guo, Y.; Zhang, Q.; Ma, Q.; Ji, C.; Zhao, L. Enzymatic degradation of deoxynivalenol by a novel bacterium, Pelagibacterium halotolerans ANSP101. Food Chem. Toxicol. 2020, 140, 111276. [Google Scholar] [CrossRef] [PubMed]
  73. Daud, N.; Currie, V.; Duncan, G.; Busman, M.; Gratz, S.W. Intestinal hydrolysis and microbial biotransformation of diacetoxyscirpenol-α-glucoside, HT-2-β-glucoside and N-(1-deoxy-d-fructos-1-yl) fumonisin B (1) by human gut microbiota in vitro. Int. J. Food Sci. Nutr. 2020, 71, 540–548. [Google Scholar] [CrossRef]
  74. Boekhorst, J.; Helmer, Q.; Kleerebezem, M.; Siezen, R.J. Comparative analysis of proteins with a mucus-binding domain found exclusively in lactic acid bacteria. Microbiology 2006, 152, 273–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Vega, M.F.; Dieguez, S.N.; Riccio, B.; Aranguren, S.; Giordano, A.; Denzoin, L.; Soraci, A.L.; Tapia, M.O.; Ross, R.; Apás, A.; et al. Zearalenone adsorption capacity of lactic acid bacteria isolated from pigs. Braz. J. Microbiol. 2017, 48, 715–723. [Google Scholar] [CrossRef] [PubMed]
  76. Belgacem, H.; Ben Salah-Abbès, J.; Ezzdini, K.; Abdel-Wahhab, M.A.; Zinedine, A.; Abbès, S. Lactobacillus plantarum MON03 counteracts zearalenone génotoxicty in mice: Chromosome aberrations, micronuclei, DNA fragmentation and apoptotique gene expression. Mutat. Res. Genet. Toxicol. Environ. Mutagenesis 2019, 840, 11–19. [Google Scholar] [CrossRef]
  77. Adunphatcharaphon, S.; Petchkongkaew, A.; Visessanguan, W. In Vitro mechanism assessment of Zearalenone removal by plant-derived Lactobacillus plantarum BCC 47723. Toxins 2021, 13, 286. [Google Scholar] [CrossRef]
  78. Złoch, M.; Rogowska, A.; Pomastowski, P.; Railean-Plugaru, V.; Walczak-Skierska, J.; Rudnicka, J.; Buszewski, B. Use of Lactobacillus paracasei strain for zearalenone binding and metabolization. Toxicon 2020, 181, 9–18. [Google Scholar] [CrossRef] [PubMed]
  79. Król, A.; Pomastowski, P.; Rafińska, K.; Railean-Plugaru, V.; Walczak, J.; Buszewski, B. Microbiology neutralization of zearalenone using Lactococcus lactis and Bifidobacterium sp. Anal. Bioanal. Chem. 2018, 410, 943–952. [Google Scholar] [CrossRef] [Green Version]
  80. Abbès, S.; Ben Salah-Abbès, J.; Jebali, R.; Younes, R.B.; Oueslati, R. Interaction of aflatoxin B1 and fumonisin B1 in mice causes immunotoxicity and oxidative stress: Possible protective role using lactic acid bacteria. J Immunotoxicol. 2016, 13, 46–54. [Google Scholar] [CrossRef]
  81. Jebali, R.; Abbès, S.; Salah-Abbès, J.B.; Younes, R.B.; Haous, Z.; Oueslati, R. Ability of Lactobacillus plantarum MON03 to mitigate aflatoxins (B1 and M1) immunotoxicities in mice. J. Immunotoxicol. 2015, 12, 290–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Nikbakht, E.; Jamaluddin, R.; Redzwan, S.M.; Khalesi, S. Oral administration of Lactobacillus casei Shirota can ameliorate the adverse effect of an acute aflatoxin exposure in Sprague Dawley rats. Int. J. Vitam. Nutr. Res. 2018, 88, 199–208. [Google Scholar] [CrossRef] [PubMed]
  83. Zhai, Y.; Hu, S.; Zhong, L.; Lu, Z.; Bie, X.; Zhao, H.; Zhang, C.; Lu, F. Characterization of deoxynivalenol detoxification by Lactobacillus paracasei LHZ-1 isolated from yogurt. J. Food Prot. 2019, 82, 1292–1299. [Google Scholar] [CrossRef] [PubMed]
  84. Li, S.; Liu, R.; Xia, S.; Wei, G.; Ishfaq, M.; Zhang, Y.; Zhang, X. Protective role of curcumin on aflatoxin B1-induced TLR4/RIPK pathway mediated-necroptosis and inflammation in chicken liver. Ecotoxicol. Environ. Saf. 2022, 233, 113319. [Google Scholar] [CrossRef]
  85. Wang, Y.; Liu, F.; Liu, M.; Zhou, X.; Wang, M.; Cao, K.; Jin, S.; Shan, A.; Feng, X. Curcumin mitigates aflatoxin B1-induced liver injury via regulating the NLRP3 inflammasome and Nrf2 signaling pathway. Food Chem. Toxicol. 2022, 161, 112823. [Google Scholar] [CrossRef]
  86. Wan, F.; Tang, L.; Rao, G.; Zhong, G.; Jiang, X.; Wu, S.; Huang, R.; Tang, Z.; Ruan, Z.; Chen, Z.; et al. Curcumin activates the Nrf2 Pathway to alleviate AFB1-induced immunosuppression in the spleen of ducklings. Toxicon 2022, 209, 18–27. [Google Scholar] [CrossRef]
  87. Jin, S.; Yang, H.; Wang, Y.; Pang, Q.; Jiao, Y.; Shan, A.; Feng, X. Dietary Curcumin Alleviated Aflatoxin B1-Induced Acute Liver Damage in Ducks by Regulating NLRP3-Caspase-1 Signaling Pathways. Foods 2021, 10, 3086. [Google Scholar] [CrossRef]
  88. Jin, S.; Yang, H.; Jiao, Y.; Pang, Q.; Wang, Y.; Wang, M.; Shan, A.; Feng, X. Dietary Curcumin Alleviated Acute Ileum Damage of Ducks (Anas platyrhynchos) Induced by AFB1 through Regulating Nrf2-ARE and NF-κB Signaling Pathways. Foods 2021, 10, 1370. [Google Scholar] [CrossRef]
  89. Owumi, S.E.; Irozuru, C.E.; Arunsi, U.O.; Oyelere, A.K. Caffeic acid protects against DNA damage, oxidative and inflammatory mediated toxicities, and upregulated caspases activation in the hepatorenal system of rats treated with aflatoxin B (1). Toxicon 2022, 207, 1–12. [Google Scholar] [CrossRef]
  90. Chen, S.J.; Gan, L.; Guo, Y.C.; Tian, L.X.; Liu, Y.J. Changes in growth performance, aflatoxin B (1) residues, immune response and antioxidant status of Litopenaeus vannamei fed with AFB (1)-contaminated diets and the regulating effect of dietary myo-inositol supplementation. Food Chem. 2020, 324, 126888. [Google Scholar] [CrossRef]
  91. Ali Rajput, S.; Sun, L.; Zhang, N.; Mohamed Khalil, M.; Gao, X.; Ling, Z.; Zhu, L.; Khan, F.A.; Zhang, J.; Qi, D. Ameliorative Effects of Grape Seed Proanthocyanidin Extract on Growth Performance, Immune Function, Antioxidant Capacity, Biochemical Constituents, Liver Histopathology and Aflatoxin Residues in Broilers Exposed to Aflatoxin B1. Toxins 2017, 9, 371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Rajput, S.A.; Zhang, C.; Feng, Y.; Wei, X.T.; Khalil, M.M.; Rajput, I.R.; Baloch, D.M.; Shaukat, A.; Rajput, N.; Qamar, H.; et al. Proanthocyanidins Alleviates AflatoxinB₁-Induced Oxidative Stress and Apoptosis through Mitochondrial Pathway in the Bursa of Fabricius of Broilers. Toxins 2019, 11, 157. [Google Scholar] [CrossRef] [Green Version]
  93. Sakamoto, M.I.; Murakami, A.E.; Fernandes, A.M.; Ospina-Rojas, I.C.; Nunes, K.C.; Hirata, A.K. Performance and serum biochemical profile of Japanese quail supplemented with silymarin and contaminated with aflatoxin B1. Poult. Sci. 2018, 97, 159–166. [Google Scholar] [CrossRef]
  94. Sarker, M.T.; Wan, X.; Yang, H.; Wang, Z. Dietary Lycopene Supplementation Could Alleviate Aflatoxin B (1) Induced Intestinal Damage through Improving Immune Function and Anti-Oxidant Capacity in Broilers. Animals 2021, 11, 3165. [Google Scholar] [CrossRef]
  95. Xu, F.; Wang, P.; Yao, Q.; Shao, B.; Yu, H.; Yu, K.; Li, Y. Lycopene alleviates AFB (1)-induced immunosuppression by inhibiting oxidative stress and apoptosis in the spleen of mice. Food Funct. 2019, 10, 3868–3879. [Google Scholar] [CrossRef]
  96. Yu, K.; Zhang, J.; Cao, Z.; Ji, Q.; Han, Y.; Song, M.; Shao, B.; Li, Y. Lycopene attenuates AFB (1)-induced renal injury with the activation of the Nrf2 antioxidant signaling pathway in mice. Food Funct. 2018, 9, 6427–6434. [Google Scholar] [CrossRef]
  97. Rajput, S.A.; Shaukat, A.; Wu, K.; Rajput, I.R.; Baloch, D.M.; Akhtar, R.W.; Raza, M.A.; Najda, A.; Rafał, P.; Albrakati, A.; et al. Luteolin Alleviates Aflatoxin B (1)-Induced apoptosis and oxidative Stress in the Liver of Mice through activation of Nrf2 signaling pathway. Antioxidants 2021, 10, 1268. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, X.; He, Y.; Tian, J.; Muhammad, I.; Liu, M.; Wu, C.; Xu, C.; Zhang, X. Ferulic acid prevents aflatoxin B1-induced liver injury in rats via inhibiting cytochrome P450 enzyme, activating Nrf2/GST pathway and regulating mitochondrial pathway. Ecotoxicol. Environ. Saf. 2021, 224, 112624. [Google Scholar] [CrossRef]
  99. Guo, Y.; Balasubramanian, B.; Zhao, Z.H.; Liu, W.C. Marine algal polysaccharides alleviate aflatoxin B1-induced bursa of Fabricius injury by regulating redox and apoptotic signaling pathway in broilers. Poult. Sci. 2021, 100, 844–857. [Google Scholar] [CrossRef] [PubMed]
  100. Abdel-Daim, M.M.; Abdeen, A.; Jalouli, M.; Abdelkader, A.; Megahed, A.; Alkahtane, A.; Almeer, R.; Alhoshani, N.M.; Al-Johani, N.S.; Alkahtani, S.; et al. Fucoidan supplementation modulates hepato-renal oxidative stress and DNA damage induced by aflatoxin B1 intoxication in rats. Sci. Total Environ. 2021, 768, 144781. [Google Scholar] [CrossRef]
  101. Liao, S.; Shi, D.; Clemons-Chevis, C.L.; Guo, S.; Su, R.; Qiang, P.; Tang, Z. Protective role of selenium on aflatoxin b1-induced hepatic dysfunction and apoptosis of liver in ducklings. Biol. Trace Elem. Res. 2014, 162, 296–301. [Google Scholar] [CrossRef] [PubMed]
  102. Chen, K.; Shu, G.; Peng, X.; Fang, J.; Cui, H.; Chen, J.; Wang, F.; Chen, Z.; Zuo, Z.; Deng, J.; et al. Protective role of sodium selenite on histopathological lesions, decreased T-cell subsets and increased apoptosis of thymus in broilers intoxicated with aflatoxin B1. Food Chem. Toxicol. 2013, 59, 446–454. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, F.; Wang, Y.; Zhou, X.; Liu, M.; Jin, S.; Shan, A.; Feng, X. Resveratrol Relieved Acute Liver Damage in Ducks (Anas platyrhynchos) Induced by AFB1 via Modulation of Apoptosis and Nrf2 Signaling Pathways. Animals 2021, 11, 3516. [Google Scholar] [CrossRef]
  104. Li, Y.; Ma, Q.G.; Zhao, L.H.; Wei, H.; Duan, G.X.; Zhang, J.Y.; Ji, C. Effects of lipoic acid on immune function, the antioxidant defense system, and inflammation-related genes expression of broiler chickens fed aflatoxin contaminated diets. Int. J. Mol. Sci. 2014, 15, 5649–5662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zha, A.; Tu, R.; Cui, Z.; Qi, M.; Liao, S.; Wang, J.; Tan, B.; Liao, P. Baicalin-Zinc complex alleviates inflammatory responses and hormone profiles by microbiome in Deoxynivalenol induced piglets. Front. Nutr. 2021, 8, 738281. [Google Scholar] [CrossRef] [PubMed]
  106. Zha, A.; Yuan, D.; Cui, Z.; Qi, M.; Liao, S.; Liao, P.; Tan, B. The Evaluation of the Antioxidant and Intestinal Protective Effects of Baicalin-Copper in Deoxynivalenol-Challenged Piglets. Oxid. Med. Cell. Longev. 2020, 2020, 5363546. [Google Scholar] [CrossRef] [Green Version]
  107. Liao, P.; Li, Y.; Li, M.; Chen, X.; Yuan, D.; Tang, M.; Xu, K. Baicalin alleviates deoxynivalenol-induced intestinal inflammation and oxidative stress damage by inhibiting NF-κB and increasing mTOR signaling pathways in piglets. Food Chem. Toxicol. 2020, 140, 111326. [Google Scholar] [CrossRef]
  108. Rajput, S.A.; Liang, S.J.; Wang, X.Q.; Yan, H.C. Lycopene Protects Intestinal Epithelium from Deoxynivalenol-Induced Oxidative Damage via Regulating Keap1/Nrf2 Signaling. Antioxidants 2021, 10, 1493. [Google Scholar] [CrossRef]
  109. Qiu, Y.; Yang, J.; Wang, L.; Yang, X.; Gao, K.; Zhu, C.; Jiang, Z. Dietary resveratrol attenuation of intestinal inflammation and oxidative damage is linked to the alteration of gut microbiota and butyrate in piglets challenged with deoxynivalenol. J. Anim. Sci. Biotechnol. 2021, 12, 71. [Google Scholar] [CrossRef]
  110. Huang, Y.; Zheng, C.; Song, B.; Wang, L.; Xiao, H.; Jiang, Z. Resveratrol Ameliorates Intestinal Damage Challenged with Deoxynivalenol Through Mitophagy in vitro and in vivo. Front. Vet. Sci. 2021, 8, 807301. [Google Scholar] [CrossRef]
  111. Hong, Q.; Li, X.; Lin, Q.; Shen, Z.; Feng, J.; Hu, C. Resveratrol Improves Intestinal Morphology and Anti-Oxidation Ability in Deoxynivalenol-Challenged Piglets. Animals 2022, 12, 311. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, S.; Zhang, C.; Yang, J.; Wang, X.; Wu, K.; Zhang, B.; Zhang, J.; Yang, A.; Rajput, S.A.; Qi, D. Sodium butyrate protects the intestinal barrier by modulating intestinal host defense peptide expression and gut microbiota after a challenge with deoxynivalenol in weaned piglets. J. Agric. Food Chem. 2020, 68, 4515–4527. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, J.Y.; Lin, H.L.; Wang, Z.; Zhang, S.W.; Huang, D.G.; Gao, C.Q.; Yan, H.C.; Wang, X.Q. Zinc L-Aspartate enhances intestinal stem cell activity to protect the integrity of the intestinal mucosa against deoxynivalenol through activation of the Wnt/β-catenin signaling pathway. Environ. Pollut. 2020, 262, 114290. [Google Scholar] [CrossRef]
  114. Zhou, J.Y.; Lin, H.L.; Qin, Y.C.; Li, X.G.; Gao, C.Q.; Yan, H.C.; Wang, X.Q. l-Carnosine protects against Deoxynivalenol-induced oxidative stress in intestinal stem cells by regulating the Keap1/Nrf2 signaling pathway. Mol. Nutr. Food Res. 2021, 65, e2100406. [Google Scholar] [CrossRef]
  115. Rajput, S.A.; Shaukat, A.; Rajput, I.R.; Kamboh, A.A.; Iqbal, Z.; Saeed, M.; Akhtar, R.W.; Shah, S.; Raza, M.A.; El Askary, A.; et al. Ginsenoside Rb1 prevents deoxynivalenol-induced immune injury via alleviating oxidative stress and apoptosis in mice. Ecotoxicol. Environ. Saf. 2021, 220, 112333. [Google Scholar] [CrossRef]
  116. Liu, Z.H.; Xie, W.W.; Zan, G.X.; Gao, C.Q.; Yan, H.C.; Zhou, J.Y.; Wang, X.Q. Lauric acid alleviates deoxynivalenol-induced intestinal stem cell damage by potentiating the Akt/mTORC1/S6K1 signaling axis. Chem. Biol. Interact. 2021, 348, 109640. [Google Scholar] [CrossRef] [PubMed]
  117. Liao, S.; Tang, S.; Tan, B.; Li, J.; Qi, M.; Cui, Z.; Zha, A.; Wang, Y.; Yin, Y.; Sun, P.; et al. Chloroquine improves Deoxynivalenol-induced inflammatory response and intestinal mucosal damage in piglets. Oxid. Med. Cell. Longev. 2020, 2020, 9834813. [Google Scholar] [CrossRef] [PubMed]
  118. Zhou, J.Y.; Wang, Z.; Zhang, S.W.; Lin, H.L.; Gao, C.Q.; Zhao, J.C.; Yang, C.; Wang, X.Q. Methionine and its hydroxyl analogues improve stem cell activity to eliminate Deoxynivalenol-induced intestinal injury by reactivating Wnt/β-Catenin signaling. J. Agric. Food Chem. 2019, 67, 11464–11473. [Google Scholar] [CrossRef] [PubMed]
  119. Wu, M.; Xiao, H.; Ren, W.; Yin, J.; Tan, B.; Liu, G.; Li, L.; Nyachoti, C.M.; Xiong, X.; Wu, G. Therapeutic effects of glutamic acid in piglets challenged with deoxynivalenol. PLoS ONE 2014, 9, e100591. [Google Scholar] [CrossRef]
  120. Li, T.; Huang, S.; Wang, J.; Yin, P.; Liu, H.; Sun, C. Alginate oligosaccharides protect against fumonisin B1-induced intestinal damage via promoting gut microbiota homeostasis. Food Res. Int. 2022, 152, 110927. [Google Scholar] [CrossRef]
  121. Fortuoso, B.F.; Galli, G.M.; Griss, L.G.; Armanini, E.H.; Silva, A.D.; Fracasso, M.; Mostardeiro, V.; Morsch, V.M.; Lopes, L.; Santos, R.; et al. Effects of glycerol monolaurate on growth and physiology of chicks consuming diet containing fumonisin. Microb. Pathog. 2020, 147, 104261. [Google Scholar] [CrossRef] [PubMed]
  122. Gbore, F.A.; Adewumi, F. HAmeliorative potential of Moringa Leaf Meal on nutrient digestibility of rabbits fed Fumonisin B (1)-contaminated diets. Toxicon 2021, 201, 164–168. [Google Scholar] [CrossRef] [PubMed]
  123. Lin, X.; Zhu, L.; Gao, X.; Kong, L.; Huang, Y.; Zhao, H.; Chen, Y.; Wen, L.; Li, R.; Wu, J.; et al. Ameliorative effect of betulinic acid against zearalenone exposure triggers testicular dysfunction and oxidative stress in mice via p38/ERK MAPK inhibition and Nrf2-mediated antioxidant defense activation. Ecotoxicol. Environ. Saf. 2022, 238, 113561. [Google Scholar] [CrossRef]
  124. Gao, X.; Xiao, Z.H.; Liu, M.; Zhang, N.Y.; Khalil, M.M.; Gu, C.Q.; Qi, D.S.; Sun, L.H. Dietary silymarin supplementation alleviates Zearalenone-induced hepatotoxicity and reproductive toxicity in rats. J. Nutr. 2018, 148, 1209–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Toda, K.; Uno, S.; Kokushi, E.; Shiiba, A.; Hasunuma, H.; Matsumoto, D.; Ohtani, M.; Yamato, O.; Shinya, U.; Wijayagunawardane, M.; et al. Fructo-oligosaccharide (DFA III) feed supplementation for mitigation of mycotoxin exposure in cattle-clinical evaluation by a urinary zearalenone monitoring system. Toxins 2018, 10, 223. [Google Scholar] [CrossRef] [Green Version]
  126. Zhu, W.; Ge, M.; Li, X.; Wang, J.; Wang, P.; Tai, T.; Wang, Y.; Sun, J.; Shi, G. Hyperoside attenuates zearalenone-induced spleen injury by suppressing oxidative stress and inhibiting apoptosis in mice. Int. Immunopharmacol. 2022, 102, 108408. [Google Scholar] [CrossRef] [PubMed]
  127. Xu, J.; Li, S.; Jiang, L.; Gao, X.; Liu, W.; Zhu, X.; Huang, W.; Zhao, H.; Wei, Z.; Wang, K.; et al. Baicalin protects against zearalenone-induced chicks liver and kidney injury by inhibiting expression of oxidative stress, inflammatory cytokines and caspase signaling pathway. Int. Immunopharmacol. 2021, 100, 108097. [Google Scholar] [CrossRef] [PubMed]
  128. Virk, P.; Al-Mukhaizeem, N.; Bin Morebah, S.H.; Fouad, D.; Elobeid, M. Protective effect of resveratrol against toxicity induced by the mycotoxin, zearalenone in a rat model. Food Chem. Toxicol. 2020, 146, 111840. [Google Scholar] [CrossRef]
  129. Long, M.; Yang, S.; Zhang, W.; Zhang, Y.; Li, P.; Guo, Y.; Wang, Y.; Chen, X.; He, J. The Influence of selenium yeast on hematological, biochemical and reproductive hormone level changes in kunming mice following acute exposure to Zearalenone. Biol. Trace Elem. Res. 2016, 174, 362–368. [Google Scholar] [CrossRef] [PubMed]
  130. Long, M.; Yang, S.; Zhang, Y.; Li, P.; Han, J.; Dong, S.; Chen, X.; He, J. Proanthocyanidin protects against acute zearalenone-induced testicular oxidative damage in male mice. Environ. Sci. Pollut. Res. Int. 2017, 24, 938–946. [Google Scholar] [CrossRef] [PubMed]
  131. Long, M.; Yang, S.H.; Han, J.X.; Li, P.; Zhang, Y.; Dong, S.; Chen, X.; Guo, J.; Wang, J.; He, J.B. The Protective Effect of Grape-Seed Proanthocyanidin Extract on Oxidative Damage Induced by Zearalenone in Kunming Mice Liver. Int. J. Mol. Sci. 2016, 17, 808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Boeira, S.P.; Funck, V.R.; Borges Filho, C.; Del’Fabbro, L.; de Gomes, M.G.; Donato, F.; Royes, L.F.; Oliveira, M.S.; Jesse, C.R.; Furian, A.F. Lycopene protects against acute zearalenone-induced oxidative, endocrine, inflammatory and reproductive damages in male mice. Chem. Biol. Interact. 2015, 230, 50–57. [Google Scholar] [CrossRef] [PubMed]
  133. Owumi, S.E.; Bello, S.A.; Najophe, S.E.; Nwozo, S.O.; Esan, I.O. Coadministration of gallic acid abates zearalenone-mediated defects in male rat’s reproductive function. J. Biochem. Mol. Toxicol. 2022, 36, e22940. [Google Scholar] [CrossRef] [PubMed]
  134. Abdel-Tawwab, M.; Khalil, R.H.; Diab, A.M.; Khallaf, M.A.; Abdel-Razek, N.; Abdel-Latif, H.; Khalifa, E. Dietary garlic and chitosan enhanced the antioxidant capacity, immunity, and modulated the transcription of HSP70 and Cytokine genes in Zearalenone-intoxicated European seabass. Fish Shellfish Immunol. 2021, 113, 35–41. [Google Scholar] [CrossRef]
  135. Zhang, Y.; Hu, B.; Wang, M.; Tong, J.; Pan, J.; Wang, N.; Gong, P.; Long, M. Selenium Protects against Zearalenone-induced oxidative stress and apoptosis in the mouse kidney by inhibiting endoplasmic reticulum stress. Oxid. Med. Cell. Longev. 2020, 2020, 6059058. [Google Scholar] [CrossRef]
  136. Del Fabbro, L.; Jesse, C.R.; de Gomes, M.G.; Borges Filho, C.; Donato, F.; Souza, L.C.; Goes, A.R.; Furian, A.F.; Boeira, S.P. The flavonoid chrysin protects against zearalenone induced reproductive toxicity in male mice. Toxicon 2019, 165, 13–21. [Google Scholar] [CrossRef]
  137. Abdelrahman, R.E.; Khalaf, A.; Elhady, M.A.; Ibrahim, M.A.; Hassanen, E.I.; Noshy, P.A. Quercetin ameliorates ochratoxin A-Induced immunotoxicity in broiler chickens by modulation of PI3K/AKT pathway. Chem. Biol. Interact. 2022, 351, 109720. [Google Scholar] [CrossRef]
  138. Abdel-Wahhab, M.A.; Aljawish, A.; El-Nekeety, A.A.; Abdel-Aziem, S.H.; Hassan, N.S. Chitosan nanoparticles plus quercetin suppress the oxidative stress, modulate DNA fragmentation and gene expression in the kidney of rats fed ochratoxin A-contaminated diet. Food Chem. Toxicol. 2017, 99, 209–221. [Google Scholar] [CrossRef] [PubMed]
  139. Longobardi, C.; Damiano, S.; Andretta, E.; Prisco, F.; Russo, V.; Pagnini, F.; Florio, S.; Ciarcia, R. Curcumin Modulates Nitrosative Stress, Inflammation, and DNA Damage and Protects against Ochratoxin A-Induced Hepatotoxicity and Nephrotoxicity in Rats. Antioxidants 2021, 10, 1239. [Google Scholar] [CrossRef] [PubMed]
  140. Damiano, S.; Longobardi, C.; Andretta, E.; Prisco, F.; Piegari, G.; Squillacioti, C.; Montagnaro, S.; Pagnini, F.; Badino, P.; Florio, S.; et al. Antioxidative effects of curcumin on the hepatotoxicity induced by Ochratoxin A in rats. Antioxidants 2021, 10, 125. [Google Scholar] [CrossRef] [PubMed]
  141. Ruan, D.; Wang, W.C.; Lin, C.X.; Fouad, A.M.; Chen, W.; Xia, W.G.; Wang, S.; Luo, X.; Zhang, W.H.; Yan, S.J.; et al. Effects of curcumin on performance, antioxidation, intestinal barrier and mitochondrial function in ducks fed corn contaminated with ochratoxin A. Animal 2019, 13, 42–52. [Google Scholar] [CrossRef] [PubMed]
  142. Damiano, S.; Andretta, E.; Longobardi, C.; Prisco, F.; Paciello, O.; Squillacioti, C.; Mirabella, N.; Florio, S.; Ciarcia, R. Effects of curcumin on the renal toxicity induced by Ochratoxin A in rats. Antioxidants 2020, 9, 332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Li, L.; Chen, Y.; Jiao, D.; Yang, S.; Li, L.; Li, P. Protective Effect of Astaxanthin on Ochratoxin A-Induced Kidney Injury to Mice by Regulating Oxidative Stress-Related NRF2/KEAP1 Pathway. Molecules 2020, 25, 1386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Cui, G.; Li, L.; Xu, W.; Wang, M.; Jiao, D.; Yao, B.; Xu, K.; Chen, Y.; Yang, S.; Long, M.; et al. Astaxanthin protects Ochratoxin A-induced oxidative stress and apoptosis in the heart via the Nrf2 pathway. Oxid. Med. Cell. Longev. 2020, 2020, 7639109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Li, K.; Cao, Z.; Guo, Y.; Tong, C.; Yang, S.; Long, M.; Li, P.; He, J. Selenium Yeast Alleviates Ochratoxin A-induced apoptosis and oxidative stress via modulation of the PI3K/AKT and Nrf2/Keap1 signaling pathways in the kidneys of chickens. Oxid. Med. Cell. Longev. 2020, 2020, 4048706. [Google Scholar] [CrossRef]
  146. Li, P.; Li, K.; Zou, C.; Tong, C.; Sun, L.; Cao, Z.; Yang, S.; Lyu, Q. Selenium Yeast Alleviates Ochratoxin A-Induced Hepatotoxicity via Modulation of the PI3K/AKT and Nrf2/Keap1 Signaling Pathways in Chickens. Toxins 2020, 12, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Abdel-Wahhab, M.A.; Aljawish, A.; Kenawy, A.M.; El-Nekeety, A.A.; Hamed, H.S.; Abdel-Aziem, S.H. Grafting of gallic acid onto chitosan nano particles enhances antioxidant activities in vitro and protects against ochratoxin A toxicity in catfish (Clarias gariepinus). Environ. Toxicol. Pharmacol. 2016, 41, 279–288. [Google Scholar] [CrossRef]
  148. Liu, S.; Kang, W.; Mao, X.; Ge, L.; Du, H.; Li, J.; Hou, L.; Liu, D.; Yin, Y.; Liu, Y.; et al. Melatonin mitigates aflatoxin B1-induced liver injury via modulation of gut microbiota/intestinal FXR/liver TLR4 signaling axis in mice. J. Pineal Res. 2022, 73, e12812. [Google Scholar] [CrossRef] [PubMed]
  149. Xu, X.; Wang, H.; Guo, D.; Man, X.; Liu, J.; Li, J.; Luo, C.; Zhang, M.; Zhen, L.; Liu, X. Curcumin modulates gut microbiota and improves renal function in rats with uric acid nephropathy. Ren. Fail. 2021, 43, 1063–1075. [Google Scholar] [CrossRef]
  150. Islam, T.; Koboziev, I.; Albracht-Schulte, K.; Mistretta, B.; Scoggin, S.; Yosofvand, M.; Moussa, H.; Zabet-Moghaddam, M.; Ramalingam, L.; Gunaratne, P.H.; et al. Curcumin Reduces Adipose Tissue Inflammation and Alters Gut Microbiota in Diet-Induced Obese Male Mice. Mol. Nutr. Food Res. 2021, 65, e2100274. [Google Scholar] [CrossRef]
  151. Li, R.; Yao, Y.; Gao, P.; Bu, S. The Therapeutic Efficacy of Curcumin vs. Metformin in Modulating the Gut Microbiota in NAFLD Rats: A Comparative Study. Front. Microbiol. 2020, 11, 555293. [Google Scholar] [CrossRef] [PubMed]
  152. Zhuang, Y.; Huang, H.; Liu, S.; Liu, F.; Tu, Q.; Yin, Y.; He, S. Resveratrol improves growth performance, intestinal morphology, and microbiota composition and metabolism in mice. Front. Microbiol. 2021, 12, 726878. [Google Scholar] [CrossRef] [PubMed]
  153. Liao, J.X.; Chen, Y.W.; Shih, M.K.; Tain, Y.L.; Yeh, Y.T.; Chiu, M.H.; Chang, S.; Hou, C.Y. Resveratrol butyrate esters inhibit BPA-induced liver damage in male offspring rats by modulating antioxidant capacity and gut microbiota. Int. J. Mol. Sci. 2021, 22, 5273. [Google Scholar] [CrossRef] [PubMed]
  154. Lin, S.; Yang, X.; Yuan, P.; Yang, J.; Wang, P.; Zhong, H.; Zhang, X.; Che, L.; Feng, B.; Li, J.; et al. Undernutrition shapes the gut microbiota and bile acid profile in association with altered gut-liver FXR signaling in weaning pigs. J. Agric. Food Chem. 2019, 67, 3691–3701. [Google Scholar] [CrossRef]
  155. van der Meer, Y.; Gerrits, W.J.; van den Bosch, M.; Holst, J.J.; Moreto, M.; Buurman, W.A.; Kulik, W.; van Kempen, T.A. Chenodeoxycholic acid reduces intestinal permeability in newly weaned piglets. J. Anim. Sci. 2012, 90, 302–304. [Google Scholar] [CrossRef] [Green Version]
  156. Weaver, A.C.; See, M.T.; Hansen, J.A.; Kim, Y.B.; De Souza, A.L.; Middleton, T.F.; Kim, S.W. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ health and immune status during chronic exposure. Toxins 2013, 5, 1261–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Chen, M.L.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.D.; Zhang, Q.Y.; Mi, M.T. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. mBio 2016, 7, e2210-5. [Google Scholar] [CrossRef] [Green Version]
  158. Zhang, Z.; Wu, X.; Cao, S.; Wang, L.; Wang, D.; Yang, H.; Feng, Y.; Wang, S.; Li, L. Caffeic acid ameliorates colitis in association with increased Akkermansia population in the gut microbiota of mice. Oncotarget 2016, 7, 31790–31799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Wan, F.; Zhong, R.; Wang, M.; Zhou, Y.; Chen, Y.; Yi, B.; Hou, F.; Liu, L.; Zhao, Y.; Chen, L.; et al. Caffeic Acid Supplement Alleviates Colonic Inflammation and Oxidative Stress Potentially Through Improved Gut Microbiota Community in Mice. Front. Microbiol. 2021, 12, 784211. [Google Scholar] [CrossRef] [PubMed]
  160. Radka, C.D.; Frank, M.W.; Rock, C.O.; Yao, J. Fatty acid activation and utilization by Alistipes finegoldii, a representative Bacteroidetes resident of the human gut microbiome. Mol. Microbiol. 2020, 113, 807–825. [Google Scholar] [CrossRef]
  161. Zhang, T.; Li, Q.; Cheng, L.; Buch, H.; Zhang, F. Akkermansia muciniphila is a promising probiotic. Microb. Biotechnol. 2019, 12, 1109–1125. [Google Scholar] [CrossRef] [Green Version]
  162. Verediano, T.A.; Stampini Duarte Martino, H.; Dias Paes, M.C.; Tako, E. Effects of Anthocyanin on Intestinal Health: A Systematic Review. Nutrients 2021, 13, 1331. [Google Scholar] [CrossRef]
  163. Tolnai, E.; Fauszt, P.; Fidler, G.; Pesti-Asboth, G.; Szilagyi, E.; Stagel, A.; Konya, J.; Szabo, J.; Stundl, L.; Babinszky, L.; et al. Nutraceuticals Induced Changes in the Broiler Gastrointestinal Tract Microbiota. mSystems 2021, 6, e01124-20. [Google Scholar] [CrossRef]
  164. Kadosh, E.; Snir-Alkalay, I.; Venkatachalam, A.; May, S.; Lasry, A.; Elyada, E.; Zinger, A.; Shaham, M.; Vaalani, G.; Mernberger, M.; et al. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 2020, 586, 133–138. [Google Scholar] [CrossRef]
  165. Yang, K.; Deng, X.; Jian, S.; Zhang, M.; Wen, C.; Xin, Z.; Zhang, L.; Tong, A.; Ye, S.; Liao, P.; et al. Gallic acid alleviates gut dysfunction and boosts immune and antioxidant activities in puppies under environmental stress based on microbiome-metabolomics analysis. Front. Immunol. 2021, 12, 813890. [Google Scholar] [CrossRef] [PubMed]
  166. Geldert, C.; Abdo, Z.; Stewart, J.E.; Arathi, H.S. Dietary supplementation with phytochemicals improves diversity and abundance of honey bee gut microbiota. J. Appl. Microbiol. 2021, 130, 1705–1720. [Google Scholar] [CrossRef]
  167. She, J.; Feng, N.; Zheng, W.; Zheng, H.; Cai, P.; Zou, H.; Yuan, Y.; Gu, J.; Liu, Z.; Bian, J. Zearalenone Exposure Disrupts Blood-Testis Barrier Integrity through Excessive Ca2+-Mediated Autophagy. Toxins 2021, 13, 875. [Google Scholar] [CrossRef] [PubMed]
  168. Feng, T.; Liu, Y. Microorganisms in the reproductive system and probiotic’s regulatory effects on reproductive health. Comput. Struct. Biotechnol. J. 2022, 20, 1541–1553. [Google Scholar] [CrossRef]
  169. Li, Y.; Xie, Z.; Gao, T.; Li, L.; Chen, Y.; Xiao, D.; Liu, W.; Zou, B.; Lu, B.; Tian, X.; et al. A holistic view of gallic acid-induced attenuation in colitis based on microbiome-metabolomics analysis. Food Funct. 2019, 10, 4046–4061. [Google Scholar] [CrossRef] [PubMed]
  170. Hosseini, A.; Razavi, B.M.; Banach, M.; Hosseinzadeh, H. Quercetin and metabolic syndrome: A review. Phytother. Res. 2021, 35, 5352–5364. [Google Scholar] [CrossRef]
  171. Marunaka, Y.; Marunaka, R.; Sun, H.; Yamamoto, T.; Kanamura, N.; Inui, T.; Taruno, A. Actions of Quercetin, a Polyphenol, on Blood Pressure. Molecules 2017, 22, 209. [Google Scholar] [CrossRef] [Green Version]
  172. Eid, H.M.; Haddad, P.S. The antidiabetic potential of quercetin: Underlying mechanisms. Curr. Med. Chem. 2017, 24, 355–364. [Google Scholar] [CrossRef]
  173. Shi, T.; Bian, X.; Yao, Z.; Wang, Y.; Gao, W.; Guo, C. Quercetin improves gut dysbiosis in antibiotic-treated mice. Food Funct. 2020, 11, 8003–8013. [Google Scholar] [CrossRef] [PubMed]
  174. Xu, B.; Qin, W.; Xu, Y.; Yang, W.; Chen, Y.; Huang, J.; Zhao, J.; Ma, L. Dietary quercetin supplementation attenuates diarrhea and intestinal damage by regulating gut microbiota in weanling piglets. Oxid. Med. Cell. Longev. 2021, 2021, 6221012. [Google Scholar] [CrossRef]
  175. Dey, P. Gut microbiota in phytopharmacology: A comprehensive overview of concepts, reciprocal interactions, biotransformations and mode of actions. Pharmacol. Res. 2019, 147, 104367. [Google Scholar] [CrossRef] [PubMed]
  176. Kang, M.J.; Ko, G.S.; Oh, D.G.; Kim, J.S.; Noh, K.; Kang, W.; Yoon, W.K.; Kim, H.C.; Jeong, H.G.; Jeong, T.C. Role of metabolism by intestinal microbiota in pharmacokinetics of oral baicalin. Arch. Pharm. Res. 2014, 37, 371–378. [Google Scholar] [CrossRef] [PubMed]
  177. Noh, K.; Kang, Y.; Nepal, M.R.; Jeong, K.S.; Oh, D.G.; Kang, M.J.; Lee, S.; Kang, W.; Jeong, H.G.; Jeong, T.C. Role of intestinal microbiota in baicalin-induced drug interaction and its Pharmacokinetics. Molecules 2016, 21, 337. [Google Scholar] [CrossRef] [Green Version]
  178. Shin, H.S.; Lee, H.J.; Pyo, M.C.; Ryu, D.; Lee, K.W. Ochratoxin A-induced hepatotoxicity through Phase I and Phase II reactions regulated by AhR in liver cells. Toxins 2019, 11, 377. [Google Scholar] [CrossRef] [Green Version]
  179. Yan, X.; Zhang, Y.; Peng, Y.; Li, X. The water extract of Radix scutellariae, its total flavonoids and baicalin inhibited CYP7A1 expression, improved bile acid, and glycolipid metabolism in T2DM mice. J. Ethnopharmacol. 2022, 293, 115238. [Google Scholar] [CrossRef] [PubMed]
  180. Zheng, L.X.; Chen, X.Q.; Cheong, K.L. Current trends in marine algae polysaccharides: The digestive tract, microbial catabolism, and prebiotic potential. Int. J. Biol. Macromol. 2020, 151, 344–354. [Google Scholar] [CrossRef] [PubMed]
  181. Shannon, E.; Conlon, M.; Hayes, M. Seaweed Components as Potential Modulators of the Gut Microbiota. Mar. Drugs 2021, 19, 358. [Google Scholar] [CrossRef] [PubMed]
  182. Zhao, B.; Wu, J.; Li, J.; Bai, Y.; Luo, Y.; Ji, B.; Xia, B.; Liu, Z.; Tan, X.; Lv, J.; et al. Lycopene alleviates DSS-induced colitis and behavioral disorders via mediating microbes-gut-brain axis balance. J. Agric. Food Chem. 2020, 68, 3963–3975. [Google Scholar] [CrossRef] [PubMed]
  183. Wu, L.; Lyu, Y.; Srinivasagan, R.; Wu, J.; Ojo, B.; Tang, M.; El-Rassi, G.D.; Metzinger, K.; Smith, B.J.; Lucas, E.A.; et al. Astaxanthin-shifted gut microbiota is associated with inflammation and metabolic homeostasis in mice. J. Nutr. 2020, 150, 2687–2698. [Google Scholar] [CrossRef] [PubMed]
  184. Gangadoo, S.; Bauer, B.W.; Bajagai, Y.S.; Van, T.T.H.; Moore, R.J.; Stanley, D. In Vitro growth of gut microbiota with selenium nanoparticles. Anim. Nutr. 2019, 5, 424–431. [Google Scholar] [CrossRef] [PubMed]
  185. Bai, Y.; Ma, K.; Li, J.; Li, J.; Bi, C.; Shan, A. Deoxynivalenol exposure induces liver damage in mice: Inflammation and immune responses, oxidative stress, and protective effects of Lactobacillus rhamnosus GG. Food Chem. Toxicol. 2021, 156, 112514. [Google Scholar] [CrossRef]
  186. Lin, R.; Sun, Y.; Mu, P.; Zheng, T.; Mu, H.; Deng, F.; Deng, Y.; Wen, J. Lactobacillus rhamnosus GG supplementation modulates the gut microbiota to promote butyrate production, protecting against deoxynivalenol exposure in nude mice. Biochem. Pharmacol. 2020, 175, 113868. [Google Scholar] [CrossRef]
  187. Maidana, L.G.; Gerez, J.; Pinho, F.; Garcia, S.; Bracarense, A. Lactobacillus plantarum culture supernatants improve intestinal tissue exposed to deoxynivalenol. Exp. Toxicol. Pathol. 2017, 69, 666–671. [Google Scholar] [CrossRef]
  188. Yang, X.; Liang, S.; Guo, F.; Ren, Z.; Yang, X.; Long, F. Gut microbiota mediates the protective role of Lactobacillus plantarum in ameliorating deoxynivalenol-induced apoptosis and intestinal inflammation of broiler chickens. Poult. Sci. 2020, 99, 2395–2406. [Google Scholar] [CrossRef]
  189. García, G.R.; Payros, D.; Pinton, P.; Dogi, C.A.; Laffitte, J.; Neves, M.; González Pereyra, M.L.; Cavaglieri, L.R.; Oswald, I.P. Intestinal toxicity of deoxynivalenol is limited by Lactobacillus rhamnosus RC007 in pig jejunum explants. Arch. Toxicol. 2018, 92, 983–993. [Google Scholar] [CrossRef]
  190. Wang, S.; Hou, Q.; Guo, Q.; Zhang, J.; Sun, Y.; Wei, H.; Shen, L. Isolation and characterization of a Deoxynivalenol-degrading bacterium Bacillus licheniformis YB9 with the capability of modulating intestinal microbial flora of mice. Toxins 2020, 12, 184. [Google Scholar] [CrossRef]
  191. Li, F.; Wang, J.; Huang, L.; Chen, H.; Wang, C. Effects of adding Clostridium sp. WJ06 on intestinal morphology and microbial diversity of growing pigs fed with natural Deoxynivalenol contaminated wheat. Toxins 2017, 9, 383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Xia, D.; Yang, L.; Li, Y.; Chen, J.; Zhang, X.; Wang, H.; Zhai, S.; Jiang, X.; Meca, G.; Wang, S.; et al. Melatonin alleviates Ochratoxin A-induced liver inflammation involved intestinal microbiota homeostasis and microbiota-independent manner. J. Hazard. Mater. 2021, 413, 125239. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, Y.; Zhao, S.; Jiao, D.; Yao, B.; Yang, S.; Li, P.; Long, M. Astaxanthin Alleviates Ochratoxin A-Induced Cecum Injury and Inflammation in Mice by Regulating the Diversity of Cecal Microbiota and TLR4/MyD88/NF-κB Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 8894491. [Google Scholar] [CrossRef] [PubMed]
  194. Yang, S.; Li, L.; Yu, L.; Sun, L.; Li, K.; Tong, C.; Xu, W.; Cui, G.; Long, M.; Li, P. Selenium-enriched yeast reduces caecal pathological injuries and intervenes changes of the diversity of caecal microbiota caused by Ochratoxin-A in broilers. Food Chem. Toxicol. 2020, 137, 111139. [Google Scholar] [CrossRef] [PubMed]
  195. Peng, M.; Liu, J.; Liang, Z. Probiotic bacillus subtilis CW14 reduces disruption of the epithelial barrier and toxicity of ochratoxin A to Caco-2 cells. Food Chem. Toxicol. 2019, 126, 25–33. [Google Scholar] [CrossRef]
  196. Chen, J.; Lv, Z.; Cheng, Z.; Wang, T.; Li, P.; Wu, A.; Nepovimova, E.; Long, M.; Wu, W.; Kuca, K. Bacillus amyloliquefaciens B10 inhibits aflatoxin B1-induced cecal inflammation in mice by regulating their intestinal flora. Food Chem. Toxicol. 2021, 156, 112438. [Google Scholar] [CrossRef]
  197. Wang, Y.; Zhang, J.; Wang, Y.; Wang, K.; Wei, H.; Shen, L. Isolation and characterization of the Bacillus cereus BC7 strain, which is capable of zearalenone removal and intestinal flora modulation in mice. Toxicon 2018, 155, 9–20. [Google Scholar] [CrossRef]
Table 1. Direct toxicity of mycotoxins to intestinal epithelial cells.
Table 1. Direct toxicity of mycotoxins to intestinal epithelial cells.
Mycotoxin Exposure TreatmentSample Types Toxicity to Intestinal Epithelial CellsReference
DON, 0.5 μM, incubation 6 or 12 hPig, intestinal epithelial cells (IPEC-J2)Activates diamine oxidase (DAO), Significantly decreased expression levels of TFF2, TFF3, and Claudin-4 genes[24]
DON, 1300 and 2200 μg/kg feed, 60 d Pig, duodenal epithelial cellsActivates DAO, Low-dose group: endoplasmic reticulum swelling, irregular chromatin distribution; high-dose group: chromatin condensation, nuclear pyknosis, mitochondrial swelling and vacuolization[25]
DON, 1008 μg/kg feed, 42 dPig, Cecal epithelial cellsDecreases numbers of immune cells TLR2 and TLR4 in cecal epithelial cells, up-regulated NFκB signaling pathway[26]
DON, 0.1, 1, 10, 100 μM, incubation 10–14 dHuman, colon cancer cells (HT29-16E cells) and colorectal adenocarcinoma cells, (CACO-2 cells)Dose-dependently inhibits the expression of TFF family genes by regulating the expression of protein kinase R and MAP kinase (MAPK) p38 and ERK1/2[27]
DON, 2 μM, incubation 24 hPig, intestinal epithelial cells (IPEC-J2)Decreases the protein stability and accelerates the degradation of TJP in the lysosome[28]
ZEA, 6 and 8 μg/mL, incubation 12, 24 and 36 hPig, intestinal epithelial cells (IPEC-J2)Up-regulates ROS, causing oxidative stress[29]
ZEA, 20 μg/mL, incubation 24 hPig, intestinal epithelial cells (IPEC-J2)Up-regulates ROS, causing oxidative stress[30]
ZEA, 6 and 8 μg/mL, incubation 24 hPig, intestinal epithelial cells (IPEC-J2) and mice, peritoneal macrophagesIncreases NLRP3 inflammasome expression and cytokine release [31]
ZEA, 40 μM, incubation 24 hPig, intestinal epithelial cells (IPEC-J2)Inhibits cell proliferation and causes intestinal cell damage[32]
OTA, 5, 10, 20, 40, 80 μM, incubation 12 hPig, intestinal epithelial cells (IPEC-J2)Induces ROS generation causes barrier dysfunction, and disrupts tight junctions[33]
OTA, 0.0005, 0.005 and 4 μg/mL, incubation 48 hHuman, colorectal adenocarcinoma cells, (CACO-2 cells)Perturbs functional gene expression and induces apoptosis in a dose-dependent manner[34]
FB1, 10, 25 and 50 μg/mL, incubation 24 or 48 hPig, intestinal epithelial cells (IPEC-J2)Inhibits cell proliferation and damages tight junction proteins[35]
FB1, 5 mg/kg feed, 42 dMice, duodenal epithelial cellsCauses epithelial cells of duodenal villi to slough off and partial necrosis of intestinal glands[36]
AFB1, 0.6 mg/kg feed, 21 dChicken, intestinal epithelial cellsPathological changes in the ultrastructure of duodenal mitochondria, complete shedding of microvilli on the surface of the jejunum, reduce the number of mitochondria[37]
AFB1, 0.12 and 12 μM, incubationHuman, colorectal adenocarcinoma cells, (CACO-2 cells)Disrupts gut-tight junction proteins[38]
Table 2. Effects of mycotoxins exposure on gut microbiota.
Table 2. Effects of mycotoxins exposure on gut microbiota.
Mycotoxin Exposure TreatmentAnimal ModelsToxicity to Gut MicrobiotaReference
DON, 10 μg/kg BW, 1–280 dMiceUp-regulates the relative abundance of pathogens highly associated with chronic intestinal diseases at the phylum, family, and genus levels.[39]
DON, 3.02 mg/kg feed, 14–20 d and 35–41 dPigThis leads to altered fecal microbiota composition and microbial biological functions associated with mycotoxin detoxification[40]
DON, 5 mg/kg feed, 1–35 dChickenPromotes the Campylobacter jejuni colonization and translocation in intestinal epithelial cells[41]
DON, 5 mg/kg feed, 1–21 dChickenManipulates microbiota community composition and metabolite production, disrupts host metabolic processes[42]
DON, 2.5, 5 and 10 mg/kg feed, 1–35 dChickenDisrupts the composition of the cecal microbiota and reduces the microbial diversity[43]
DON, 2 and 10 mg/kg BW, 1–28 dRatExacerbates the genotoxicity of Escherichia coli[44]
DON, 1 and 5 mg/kg, every other day for 1–14 dMiceAlters the composition of gut microbiota, affects microbial biosynthetic and degradative functions, and further contributes to host metabolic dysfunction[45]
OTA, 0.21, 0.5 and 1.5 mg/kg BW, 1–28 dMiceDisrupts the structure and diversity of gut microbial communities[46]
OTA, 235 μg/kg BW, 1–21 dDuckDisrupts gut microbial composition and lipopolysaccharide biosynthesis function[47]
ZEA, 400, 800 and 1600 μg/kg BW, 1–28 dRabbitAffects the cecal microbiota balance and reduces the abundance of bacteria with important metabolic functions[48]
ZEA, 0.2, 1 and 5 mg/kg BW, 1–28 dRatDisrupts the integrity and function of the mucus layer, induces imbalance of gut microbiota[49]
ZEA, 5, 10 and 15 μg/kg BW, 1–42 dPigAffects the colony counts of intestinal microbiota[50]
ZEA, 20 and 40 μg/kg BW, 1–14 dMiceInhibits the glycerophospholipid metabolic pathway of gut microbiota[51]
ZEA, 20 mg/kg BW, 1–21 dMiceDisrupts microbial metabolism of lipid molecules and organic acids[52]
ZEA, 0.8 mg/kg feed and DON, 8 mg/kg feed, 1–28 dPigThe combination of DON and ZEA disrupts the gut microbial composition[53]
ZEA, 1.36 mg/kg feed and DON, 0.87 g/kg feed, 1–28 dPigAlters gut microbial composition and downregulates abundance of the microbial ribosome and pentose phosphate pathway functional genes[54]
AFB1, 5, 25, and 75 μg/kg BW, 1–28 dRatDisrupts gut microbiota-dependent organic acid metabolism[55]
Table 3. The degradation rate of mycotoxins by different microorganisms.
Table 3. The degradation rate of mycotoxins by different microorganisms.
MycotoxinMicrobiotaCulture ConditionsDegradation RateReference
AFB1Bacillus shackletonii37 °C, 72 h92.1%[57]
Pseudomonas fluorescens37 °C, 72 h99%[58]
Bacillus sp. Strains37 °C, 72 h58–96.9%[59]
lactobacillus plantarum37 °C, 24 h56%[60]
OTABrevibacterium30 °C, 6 d100%[61]
bacillus amyloliquefaciens. ASAG131 °C, 72 h99.7%[62]
Bacillus. CW1430 °C, 24 h97.6%[63]
FB1S. marcescens 329-225 °C, 24 h37%[64]
L. plantarum MYS630 °C, 30 d61.7%[65]
lactobacillus plantarum CECT 74925 °C, 15 d90.6%[66]
Lactobacillus brevis18–25 °C, 141 d90%[67]
ZEAE. coli-Lactobacillus shuttle vector pNZ300414 h99.3%[68]
Lactobacillus reuteri37 °C, 4 h100%[69]
R. percolatus JCM 1008728 °C, 7 d90%[70]
Bacillus subtilis ANSB01G37 °C, 48 h100%[71]
DONPelagibacterium halotolerans ANSP10140 °C, 6 h81%[72]
Table 4. Compounds that mitigate the effects of mycotoxins.
Table 4. Compounds that mitigate the effects of mycotoxins.
Mycotoxin Exposure TreatmentAntidote TreatmentAnimal ModelsReference
AFB1, 1 mg/kg feed, 1–28 dCurcumin, 300 mg/kg feed, 1–28 dChicken[84]
AFB1, 0.75 mg/kg BW, 1–30 dCurcumin, 200 mg/kg BW, 1–30 dMice[85]
AFB1, 0.75 mg/kg BW, 1–21 dCurcumin, 400 mg/kg feed, 1–21 dDuck[86]
AFB1, 0.75 mg/kg BW, at 70 dCurcumin, 500 mg/kg feed, 1–70 dDuck[87]
AFB1, 0.75 mg/kg BW, at 70 dCurcumin, 500 mg/kg feed, 1–70 dDuck[88]
AFB1, 50 μg/kg BW, 1–12 dCaffeic acid, 40 mg/kg BW, 1–12 dRat[89]
AFB1, 168.3 μg/kg BW, 1–58 dMyoinositol, 527.9 mg/kg feed, 1–58 dLitopenaeus vannamei[90]
AFB1, 1 mg/kg feed,1–28 dProanthocyanidin, 250 mg/kg feed, 1–28 dChicken[91]
AFB1, 1 mg/kg feed,1–28 dProanthocyanidin, 250 mg/kg feed, 1–28 dChicken[92]
AFB1, 500 μg/kg feed,1–60 dSilymarin, 500 mg/kg feed, 1–60 dJapanese quail[93]
AFB1, 100 µg/kg feed, 1–42 dLycopene, 200 mg/kg feed, 1–42 dChicken[94]
AFB1, 0.75 mg/kg BW, 1–31 dLycopene, 5 mg/kg BW, 1–31 dMice[95]
AFB1, 0.75 mg/kg BW, 1–31 dLycopene, 5 mg/kg BW, 1–31 dMice[96]
AFB1, 0.75 mg/kg BW,1–28 dLuteolin, 50 mg/kg BW,1–28 dMice[97]
AFB1, 300 μg/kg BW, 1–42 dFerulic acid, 120 mg/kg BW,1–42 dRat[98]
AFB1, 100 μg/kg feed, 1–28 dMarine-algal polysaccharides, 2500 mg/kg feed, 1–28 dChicken[99]
AFB1, 100 mg/kg BW, 1–14 dFucoidan, 200 mg/kg BW, 1–14 dRat[100]
AFB1, 0.1 mg/kg BW, 1–28 dSelenium, 1 mg/kg BW, 1–28 dDuck[101]
AFB1, 0.3 mg/kg feed, 1–21 dSelenium, 0.6 mg/kg feed, 1–21 dChicken[102]
AFB1, 0.3 mg/kg BW, at 70 dResveratrol, 500 mg/kg feed, 1–70 dDuck[103]
AFB1, 74 μg/kg BW, 1–21 dLipoic acid, 300 mg/kg feed, 1–21 dChicken[104]
DON, 4 mg/kg feed, 1–14 d 0.5% Baicalin-Zinc complex feed, 1–14 d Weaned piglets[105]
DON, 4 mg/kg feed, 1–21 d Baicalin-Copper, 5 g /kg feed, 1–21 d Weaned piglets[106]
DON, 4 mg/kg feed, 1–14 d 0.1% Baicalin feed, 1–14 dWeaned piglets[107]
DON, 3 mg/kg BW, 1–12 dLycopene, 10 mg/kg BW, 1–12 dMice[108]
DON, 3.8 mg/kg feed, 1–28 dResveratrol, 300 mg/kg feed, 1–28 dWeaned piglets[109]
DON, 3.8 mg/kg feed, 1–28 dResveratrol, 300 mg/kg feed, 1–28 dWeaned piglets[110]
DON, 2.65 mg/kg feed, 1–21 dResveratrol, 300 mg/kg, 1–21 dWeaned piglets[111]
DON, 4 mg/kg feed, 1–28 d0.2% Sodium butyrate feed, 1–28 d Weaned piglets[112]
DON, 2 mg/kg BW, 4–9 dZinc L-Aspartate, 20 mg/kg BW, 1–6 dMice[113]
DON, 3 mg/kg BW, 1–10 dl-Carnosine, 300 mg kg BW, 1–10 dMice[114]
DON, 3 mg/kg BW, 1–15 dGinsenoside Rb1, 50 mg/kg BW, 1–15 dMice[115]
DON, 2 mg/kg BW, 1–10 dLauric acid, 10 mg/kg BW, 1–10 dMice[116]
DON, 1 mg/kg BW, 11–17 dChloroquine, 10 mg/kg BW, 4–10 dWeaned piglets[117]
DON, 2 mg/kg BW, 4–8 dMethionine, 300 mg /kg BW, 1–11 dMice[118]
DON, 4 mg/kg feed, 1–36 d2% glutamic acid feed, 1–36 dWeaned piglets[119]
FB1, 1 mg/kg BW, 1–28 dAlginate oligosaccharides, 200 mg/kg BW, 1–28 dMice[120]
FB1, 400 ppb feed, 1–42 dGlycerol monolaurate, 4 mg/kg feed, 1–42 dChicken[121]
FB1, 5 mg/kg feed, 1–42 dMoringa leaf, 20 g/kg feed, 1–42 dRabbit[122]
ZEA, 0.25 mg/kg feed, 8–14 dBetulinic acid, 0.5 mg/kg feed, 1–14 dMice[123]
ZEA, 20 mg/kg feed, 1–42 dSilymarin, 500 mg/kg feed, 1–42 dRat[124]
ZEA, 0.27 mg/kg feed, 1–14 dFructo oligosaccharide10 g/d feed, 1–14 dCattle[125]
ZEA, 40 mg/kg feed, 1–30 dHyperoside, 100 mg/kg feed, 1–30 dMice[126]
ZEA, 5 mg/kg feed, 1–7 dBaicalin, 80 mg/kg feed, 5–7 dChicken[127]
ZEA, 2 mg/kg feed, 1–21 dResveratrol, 5 mg/kg feed, 1–21 dRat[128]
ZEA, 40 mg/kg BW, 13–14 dSelenium yeast, 0.25 g/kg BW 1–14 dMice[129]
ZEA, 40 mg/kg BW, 11 dProanthocyanidin, 75 mg/kg BW, 1–10 dMice[130]
ZEA, 40 mg/kg BW, 6–7 dProanthocyanidin, 100 mg/kg BW, 1–5 dMice[131]
ZEA, 40 mg/kg BW, 11 dLycopene, 20 mg/kg BW, 1–10 dMice[132]
ZEA, 100 µg/kg BW, 1–28 dGallic acid, 40 mg/kg BW, 1–28 dRat[133]
ZEA, 0.725 mg/kg feed, 1–14 dGarlic 30 g/kg and chitosan 10 g/kg feed, 1–14 dEuropean seabass[134]
ZEA, 40 mg/kg BW, 1–28 dSelenium, 0.4 mg/kg BW, 1–28 dMice[135]
ZEA, 40 mg/kg BW, 11 dChrysin, 20 mg/kg BW, 1–10 dMice[136]
OTA, 0.5 mg/kg feed, 1–42 dQuercetin, 0.5 g/kg feed, 1–42 dChicken[137]
OTA, 3 mg/kg BW, 1–21 dQuercetin, 50 mg/kg BW, 1–21 dRat[138]
OTA, 0.5 mg/kg BW, 1–45 dCurcumin, 100 mg/kg BW, 1–45 dRat[139]
OTA, 0.5 mg/kg BW, 1–45 dCurcumin, 100 mg/kg BW, 1–45 dRat[140]
OTA, 2 mg/kg feed, 1–21 dCurcumin, 400 mg/kg feed, 1–21 dDuck[141]
OTA, 0.5 mg/kg BW, 1–14 dCurcumin, 100 mg/kg BW, 1–14 dRat[142]
OTA, 5 mg/kg BW, 1–21 dAstaxanthin, 100 mg/kg BW, 1–21 dMice[143]
OTA, 5 mg/kg BW, 1–21 dAstaxanthin, 100 mg/kg BW, 1–21 dMice[144]
OTA, 50 μg/kg BW, 1–21 dSelenium yeast, 0.4 mg/kg BW, 1–21 dChicken[145]
OTA, 50 μg/kg BW, 1–21 dSelenium yeast, 0.4 mg/kg BW, 1–21 dChicken[146]
OTA, 1 mg/kg BW, 1–21 dGallic acid, 280 mg/kg BW, 1–21 dCatfish[147]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xia, D.; Mo, Q.; Yang, L.; Wang, W. Crosstalk between Mycotoxins and Intestinal Microbiota and the Alleviation Approach via Microorganisms. Toxins 2022, 14, 859. https://doi.org/10.3390/toxins14120859

AMA Style

Xia D, Mo Q, Yang L, Wang W. Crosstalk between Mycotoxins and Intestinal Microbiota and the Alleviation Approach via Microorganisms. Toxins. 2022; 14(12):859. https://doi.org/10.3390/toxins14120859

Chicago/Turabian Style

Xia, Daiyang, Qianyuan Mo, Lin Yang, and Wence Wang. 2022. "Crosstalk between Mycotoxins and Intestinal Microbiota and the Alleviation Approach via Microorganisms" Toxins 14, no. 12: 859. https://doi.org/10.3390/toxins14120859

APA Style

Xia, D., Mo, Q., Yang, L., & Wang, W. (2022). Crosstalk between Mycotoxins and Intestinal Microbiota and the Alleviation Approach via Microorganisms. Toxins, 14(12), 859. https://doi.org/10.3390/toxins14120859

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