Next Article in Journal
Trimethylamine-N-Oxide (TMAO) as a Rising-Star Metabolite: Implications for Human Health
Next Article in Special Issue
Dietary Tea Polyphenols Alleviate Acute-Heat-Stress-Induced Death of Hybrid Crucian Carp HCC2: Involvement of Modified Lipid Metabolisms in Liver
Previous Article in Journal
Short-Term Supplementation with 100% Bilberry Products and Its Effects on Body Composition and Lipid Profile in Overweight/Obese Women
Previous Article in Special Issue
Toll-like Receptor Activation Remodels the Polyamine and Tryptophan Metabolism in Porcine Macrophages
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies

1
Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
2
Yuelushan Laboratory, Changsha 410128, China
3
Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
*
Authors to whom correspondence should be addressed.
Metabolites 2025, 15(4), 219; https://doi.org/10.3390/metabo15040219
Submission received: 25 February 2025 / Revised: 17 March 2025 / Accepted: 20 March 2025 / Published: 24 March 2025
(This article belongs to the Special Issue Intestinal Health and Metabolites in Farm Animals)

Abstract

:
Background: Mycotoxins, which are secondary metabolites produced by fungi, are prevalent in animal feed and pose a serious risk to the healthy growth of livestock and poultry. Methods: This review aims to conclude current knowledge on the detrimental effects of mycotoxins on animal health and to demonstrate the potential of plant extracts as a means to counteract mycotoxin toxicity in feed. A systematic review of the literature was conducted to identify studies on the impact of mycotoxins on livestock and poultry health, as well as research into the use of plant extracts as feed additives to mitigate mycotoxin effects. Studies were selected based on their relevance to the topic, and data were extracted regarding the mechanisms of action and the efficacy of plant extracts. Results: Excessive mycotoxins in feed can lead to reduced appetite, impaired digestion, and general health issues in animals, resulting in decreased food intake, slowed weight gain, and instances of acute poisoning. Plant extracts with antioxidant, anti-inflammatory, and anti-mutagenic properties have shown the potential to improve production efficiency and reduce the toxic effects of mycotoxins. Conclusion: This comprehensive review not only consolidates the well-documented adverse effects of mycotoxins on animal health but also introduces a novel perspective by highlighting the potential of plant extracts as a promising and natural solution to counteract mycotoxin toxicity.

1. Types of Mycotoxins and Current Status of Feed Contamination

Mycotoxins, which are toxic compounds produced by specific fungi, pose a serious threat of contamination to a wide range of food sources. In agricultural products, molds are capable of producing more than 300 types of mycotoxins, many of which are considered major threats to human and animal health, with toxic and carcinogenic effects on the immune system, liver, kidneys, genes, and reproductive system [1]. The presence of these toxins in feed involves multiple complex steps, including fungal growth, toxin production, contamination pathways, absorption, distribution, and ultimate toxic effects. Ranging from hepatotoxicity to immunotoxicity, these toxins severely impact animal health, and due to their thermal stability, they can survive during feed processing, posing a persistent threat to animal productivity (Table 1). Therefore, a deep understanding of the mechanism of action of mycotoxins is crucial for developing effective prevention and control strategies to mitigate their polluting impact. Against this backdrop, measures for the prevention and control of mycotoxins are vital, and post-contamination detoxification technologies are equally critical. Recently, plant extracts containing flavonoids, terpenoids, and phenolics have become the focus of research on mycotoxin control and detoxification, with the potential of these natural compounds lying in their ability to interfere with the absorption and bioavailability of toxins, potentially reducing the toxicity of mycotoxins [2].

1.1. Types and Chemical Stability of Mycotoxins

Most mycotoxins are produced by toxigenic molds, such as Aspergillus, Penicillium, Fusarium, Claviceps, and Grapevine, and certain species of endophytic fungi. At present, more than 300 kinds of mycotoxins are known, among which aflatoxins (AFs), deoxynivalenol (DON), zearalenone (ZEA), fumonisins (FBs), and ochratoxin-A (OTA) are serious hazards to animal husbandry. They are widely distributed in cereals, such as maize and rice, and nuts, such as pistachios. These mycotoxins have stable physical and chemical properties and are resistant to high temperatures during conventional feed processing (including heating). For example, aflatoxin B1 (AFB1) is stable at most feed processing temperatures and has a boiling point of 268 °C [3]. DON remains chemically stable through processing, storage, and even cooking, which makes it difficult to eliminate [4].
Table 1. Five common mycotoxins in feed and their mechanisms of toxicology.
Table 1. Five common mycotoxins in feed and their mechanisms of toxicology.
Mycotoxin TypesSourcesToxicity CharacteristicsMechanismsReference
AFs Aspergillus flavus and Aspergillus parasiticusHighly toxic, hepatotoxic and carcinogenicInhibits protein synthesis, causes cell apoptosis, and increases the risk of hepatic carcinogenesis[5]
DONFusarium graminearumFood refusal, vomiting, growth retardation, and immunosuppressionActivates the intracellular stress response, leading to apoptosis[6]
ZEAFusarium graminearum and Fusarium rhubarbSimulating estrogen, pseudopregnancy, vaginitis, and infertilityActivates estrogen receptors and affects the development of reproductive organs[7]
FBsFusarium sp.Neurotoxicity, liver and kidney injuries, immunosuppression, and reproductive disordersInhibits sphingolipid biosynthesis and disrupts cell membrane structure[8]
OTAAspergillus spp. and Penicillium spp.Kidney injury, immunosuppression, and growth retardationInhibits protein synthesis and causes cell death[9]

1.2. Widespread Occurrence of Mycotoxin Contamination

Mycotoxin contamination is a major challenge to feed safety globally, affecting 60–80% of agriculture products. The prevalence, co-occurrence patterns, and regulatory exceedances are listed in Table 2. A quarter of the world’s food and feed crops are susceptible to mycotoxins, according to the FAO (The Food and Agriculture Organization). The European survey in 2021 showed that 98.5% of nearly 1200 feed samples contained at least one mycotoxin, 86.1% contained two or more mycotoxins, and 77% of cereals such as wheat and barley were detected with novel mycotoxins [10]. AFs are widely present in the feed of Indian dairy cattle, with AFs detected in 59% of concentrated feeds, 44% exceeding the US limit of 20 μg/kg, and 58% exceeding the EU limit of 5 μg/kg [11]. The study conducted by Zhao et al. in China from 2018 to 2020 found that the average content of AFB1 in feed and whole feed was 1.2–27.4 μg/kg, the incidence rate was 81.9–100%, and 0.9% of the raw materials were contaminated with AFB1 exceeding the Chinese safety standard concentration [12]. Wei Hao et al. ’s study, covering 9392 feed samples in China from 2017 to 2021, found that trichothecene B and FB contamination were the most common in new season maize, with detection rates as high as 84.04% and 87.16%, respectively [13]. Zhang Yong and his colleagues analyzed 1025 feed samples and found that the detection rates of AFB1, ZEN, and DON reached 95.99%, 98.54%, and 100%, respectively. Particularly striking, 89.85% of the samples contained these three mycotoxins, revealing a widespread problem of multiple contamination in 2021 [14]. Cargill Animal Nutrition analyzed over 350,000 results from 145,000 raw material samples collected from more than 150 feed mills, farms, and raw material storage sites in 2023, which indicated that the top three in terms of prevalence and risk level were DON, FBs, and ZEA [15]. Based on a globally extensive survey, it has been revealed that climate is a crucial factor in determining the areas contaminated by mycotoxins. In several regions of South Asia, Sub-Saharan Africa, and Southeast Asia, rainfall or significant temperature variations during the critical period of grain development are the decisive factors for the concentration of mycotoxin contamination. A large number of samples (64%) were co-contaminated by ≥two types of mycotoxins [16]. A study examined data from 717 wheat fields across Norway, Sweden, Finland, and the Netherlands. The observational data included details on flowering dates, the length of time between flowering and harvest, the resistance of wheat to Fusarium infection, and the concentration of deoxynivalenol (DON) in relation to various climate variables critical to wheat cultivation, such as relative humidity, temperature, and rainfall. The findings highlighted that the pattern of DON contamination in wheat in Northwestern Europe is influenced by climatic changes [17]. Taken together, mycotoxin contamination poses a global challenge that requires continuous surveillance.
Table 2. Mycotoxin contamination in feed: prevalence, co-occurrence patterns, and regulatory exceedances.
Table 2. Mycotoxin contamination in feed: prevalence, co-occurrence patterns, and regulatory exceedances.
Mycotoxin TypeCountry/RegionStudy Period/YearSample Size/ScopeContamination LevelsDetection Rate (%)Exceedance of Limits
Multiple mycotoxinsEurope2021~1200 feed samples98.5% with ≥1 mycotoxin; 86.1% with ≥2 mycotoxins; 77% cereals with novel mycotoxins98.5 (any)-
AFsIndia-Dairy cattle feedAF detected in 59% of concentrated feeds5944% exceeded US limit (20 μg/kg); 58% exceeded EU limit (5 μg/kg)
AFB1China2018–2020Various feedsAverage content: 1.2–27.4 μg/kg; incidence: 81.9–100%81.9–1000.9% of materials exceeded the Chinese safety standard
Trichothecene B and FBsChina2017–20219392 feed samplesMost common in new season maize84.04 (trichothecene B); 87.16 (FBs)-
AFB1, ZEN, and DONChina20211025 feed samplesCo-contamination prevalence95.99 (AFB1); 98.54 (ZEN); 100 (DON)89.85% of samples contained all three mycotoxins
DON, FBs, and ZENGlobal (Cargill data)2023>145,000 samples from 150+ sitesTop 3 prevalent mycotoxins by risk level-Global contamination patterns observed

1.3. Effects and Toxicology of Five Common Mycotoxin Contaminants on Livestock and Poultry

Ingestion of feed contaminated with these mycotoxins commonly induces poisoning in animals. Low concentrations impair animal growth performance and immune function, resulting in liver and kidney dysfunctions, intestinal syndrome, digestive tract inflammation, and reproductive dysfunction, while high concentrations cause acute death in animals [18,19]. In addition, the residues of mycotoxins in meat, eggs, milk, and other products have become a major hazard to the safety of animal-derived food [20]. The effective management and mitigation of mycotoxin contamination in food and feed play a crucial role in improving animal performance and ensuring the safety of the human food chain. The pathological characteristics of different animals exposed to these five common mycotoxins in diets are listed in Table 3.
AFs, as a high-risk and prevalent mycotoxin, pose significant threats to livestock health and productivity [21]. Aflatoxin mainly affects the livers of animals, and chronic exposure can cause fatty liver degeneration and liver tissue necrosis. The characteristics of poisoning include depression, loss of appetite, weight loss, stagnating movement, and decreased immunity. Liver damage was particularly significant, which was manifested as liver enlargement, fragile texture, and brown color and, in severe cases, accompanied by liver fibrosis and pathological structural changes [22,23,24,25]. Although pigs are slightly less sensitive to aflatoxin than birds, their liver damage is more serious, and the ingestion of contaminated feed by sows can be transmitted to piglets through milk, leading to growth retardation or death [26,27]. In addition, the ingestion of aflatoxin-contaminated diets by cows can destroy their intestinal barrier function, affect their immune response, and weaken their defense against pathogens while impairing their cellular and humoral immune mechanisms and reducing their resistance to viral infections and parasites [28].
ZEA, as a kind of estrogen substance, mainly affects the function of the reproductive system and can cause estrogen hyperemia in animals. The intake of ZEA during pregnancy can cause abortion, stillbirths, and malformation and has negative effects on the central nervous system, such as nausea, chills, headache, confusion, and coordination disorders [7]. Gilts are particularly sensitive to ZEA, and the typical characteristic of poisoning is vulvovaginitis with abnormal mammary gland development, while boars show decreased libido, testicular atrophy, and decreased semen quality [29]. Compared with swine, poultry has a higher tolerance to ZEA due to its efficient hepatic and intestinal circulation, rapid excretion capacity, and specific physiological characteristics, such as microbial transformation, differential enzyme activities, and low estrogen receptor binding capacity [30]. Ruminants are less susceptible to ZEA, which may be attributed to their rumen microorganisms degrading ZEA.
Table 3. Pathological characteristics of different animals exposed to five common mycotoxins in diets.
Table 3. Pathological characteristics of different animals exposed to five common mycotoxins in diets.
Mycotoxin TypesEffects on Monogastric AnimalEffects on PoultryEffects on RuminantsEffects on Aquatic Livestock
AFsGrowth retardation, decreased feed utilization, depression, anorexia, acute liver disease, and immunosuppressionSubcutaneous hemorrhage, smaller eggs, reduced yolk weight, and reduced fertilization and hatching rateReduced resistance of cows to viruses and parasitesLiver necrosis, decreased feed intake, and weight loss were observed
ZEAPseudoestrus, vaginitis, abortion, stillbirth in gilts, testicular atrophy in boars, and decreased semen quality in giltsThe ovaries were atrophic, the egg production rate was decreased, and the fertilization rate of breeding eggs was decreasedThe rumen degrades 90% of ZEA and generates the more toxic zearalenolDecreased fecundity, ovulation disorders, and infertility
DONDecreased feed intake, intestinal damage, vomiting, and food refusalInvasion of the digestive tract, decreased feed intake, food refusal, and reduced egg production rateUnder stress, the risk of poisoning is increasedDestroying the integrity of the gill structure
FBsGrowth arrest, liver tissue damage, reproductive impairment, and immunosuppressionReduced egg production, poor feather growth, oral ulcers, and neurological disordersWeight loss, immunosuppression, and liver toxicityInhibit growth and cause pathological damage
OTAWeight loss, growth retardation, and liver and kidney lesionsIncomplete eggshell calcification, high rate of egg breaking, and subcutaneous hemorrhageAnorexia, diarrhea, difficulty gaining weight, and decreased milk productionNervous system and respiratory toxicities
DON has been identified as one of the most dangerous natural food contaminants by the FAO and WHO. After the ingestion of feed contaminated with DON, livestock will show signs of anorexia, vomiting, diarrhea, fever, dyskinesia, and delayed response. In severe cases, it can lead to blood system damage and even death [31]. Pigs are the most sensitive to DON, showing obvious loss of appetite, food refusal, vomiting, and intestinal damage. Every 1 mg/kg of DON in the diet can reduce the weight gain of pigs by 8% [32]. Moreover, 2 mg/kg of DON in the diet significantly decreased the digestibility of essential amino acids, impaired the absorption of nutrients, and reduced the utilization rate of energy and nutrition [33]. Birds have a high tolerance to DON; however, high doses of DON also reduce their feed intake and growth performance [34,35]. Ruminants show a high tolerance to DON; likewise, beef cattle and sheep can tolerate up to 21 mg/kg of DON intake [36,37,38].
FBs commonly cause pulmonary edema; damage the liver, kidneys, and intestine; affect the reproductive health of boars; reduce sperm quality; and interfere with the ovarian function of sows [39,40,41,42]. For example, 2 μg/kg injection of FB1 into the air cell of chicken eggs can significantly reduce the number of chicken embryos and cause bleeding [43]. The air chamber experiment of chicken embryos incubated for 72 h after injection of different doses of FB1 showed that the mortality rate of chicken embryos was up to 100% [44].
The action mechanism of OTA on ruminants and monogastric animals is different: the former is degraded by microorganisms in the rumen, while the latter is absorbed directly in the intestine [45]. OTA mainly attacks the kidney, showing nephrotoxicity in all monogastric animals, resulting in kidney atrophy, enlargement, and pallor [46]. Experiments have shown that piglets fed rice cultures contaminated with OTA (0.2 to 0.6 mg/kg body weight per day) or pure toxin (2.0 or 1.0 mg/kg body weight per day) suddenly develop depression and reduced feed intake, resulting in weight loss, followed by diarrhea, anorexia, and dehydration [47]. After consuming OTA-contaminated feed, 65% of OTA was absorbed in the GI tract, and the remaining OTA was deposited in the kidney, resulting in weight loss, low feed efficiency, and weight gain [48]. Physiological symptoms of OTA toxicity in poultry include fatigue, reduced feed consumption, developmental delay, poor oviposition, reduced plumage, and extreme mortality at high dietary concentrations, with weight gain reduced in a dose-dependent manner when infected at rates of 0.5 to 29.4 mg/kg for 7 to 60 days in experimental studies. Reduced feed intake was observed between 21 and 60 days of 0.5–4 mg/kg OTA feeding, whereas egg production, hatchability, shell thickness, and egg production were severely affected between 28 and 84 days of 0.5–4 mg/kg OTA feeding in the diet [48,49]. In cattle, microbes in the rumen degrade low doses of OTA to nontoxic forms. Higher doses of OTA ingestion by cattle cause anorexia, diarrhea, difficulty gaining weight, and rapid loss of milk production [50].
In conclusion, different types of mycotoxins have different hazards to different livestock and poultry, and prevention and control can be focused according to the category of livestock and poultry.

2. Mechanisms and Targets of Feed Mycotoxins That Harm the Health and Growth of Livestock and Poultry

2.1. AFs

AFs are the most frequent mycotoxins in human food and animal feed. Their fat-soluble properties promote AFs to invade organisms through the skin, respiratory tract, and digestive tract, especially the digestive tract is the main pathway for the absorption of feed mycotoxins [51,52]. According to a previous study, about half of AFB1 from feed is absorbed in the duodenum and then transported to the target tissues through the blood circulation and lymphatic system [53].
The liver plays a central role in the biotransformation of AFs, in which AFB1 undergoes hydroxylation, epoxidation, hydration, and α-dimethylation catalyzed by the reductase system in the liver microsome and cytoplasm to be converted to less toxic derivatives. This conversion process includes reversible and irreversible reaction types [54,55,56].
Aflatoxin B1 is converted to a more active B1–8, 9-epoxide catalyzed by the hepatic microsomal P450 enzyme system. This epoxide can form stable adducts to DNA and proteins, for example, by covalent bonding at position C8 to DNA guanine N7 and binding to lysine residues, interfering with genetic information transmission and protein synthesis. It can induce genetic mutations, malformations, and carcinogenic effects [57,58].
By promoting the high-frequency mutation of the p53 gene, aflatoxin B1 not only eliminates its tumor suppressor function, but also activates the anti-apoptotic pathway, drives cell proliferation disorders, and then causes tumor formation, showing the typical characteristics of oncogenes. These mutant p53 proteins also tend to build stable complexes with oncogenic proteins, prolong their intracellular stability and over-express, and accelerate the process of cell carcinogenesis [59].
As a powerful immunosuppressant, aflatoxin has multiple effects, not only hindering the normal development of immune organs but also weakening host defense by inhibiting humoral and cellular immune mechanisms and increasing the sensitivity to bacterial, viral, and parasitic infections [60,61]. Aflatoxin also inhibits protein synthesis through DNA/RNA binding, resulting in thymic degeneration, the depletion of lymphocytes, and the impairment of liver and macrophage functions, accompanied by the suppression of complement C4 and T-lymphokine production [62,63]. At the level of humoral immunity, aflatoxin interferes with the activity of RNA polymerase and the synthesis of immunoglobulin, thus showing immunotoxicity [52]. In terms of cellular immunity, aflatoxin B1 further weakened the cellular immune response by downregulating the mRNA expression of IL-2 and IFN-γ in hepatocytes [64].

2.2. ZEA

The absorption and biotransformation sites of ZEA are the gastrointestinal tract and liver. Its metabolic pathway can be summarized as a two-stage process: The first stage involves the hydroxylation reaction. Under the catalysis of steroid hydrogenase and 3α/3β-hydroxysteroid deoxy enzyme, ZEA undergoes ketone group reduction at the C-6 position to produce α- and β-ZEA isomers and then undergoes double-bond reduction at the C11-C12 position to form α- and β- ZEA. In the second stage, uridine diphosphate glucuronosyltransferase (UDP-glucuronosyltransferase) catalyzes glucuronic acid conjugation of these compounds, resulting in the resultant conjugates being cleared from the body through different excretion pathways [65,66,67].
Studies have shown that ZEA and its derivatives ZOL and ZEL can specifically and competitively bind to the ERα and ERβ isoforms of the ER (estrogen receptor) in the cytoplasm, mimicking the mechanism of 17-β-estradiol. The structure of the phenolic ring in ZEA and its derivatives ZOL and ZEL show consistency with the binding domain of the A ring of estradiol in the active site. Notably, as a full agonist of ERα and a partial agonist of ERβ, the modulatory effects of ZEA on estrogenic activity are mainly mediated through ERα. After binding, the ER-ZEA complex is transported into the nucleus and interacts with estrogen response elements (EREs) to regulate target gene transcription and protein synthesis, interfere with cell proliferation and growth, and display typical estrogen-like activities [68,69,70].
The mechanism by which ZEA promotes ROS production may involve structural changes in mitochondria. As the main source of ROS, ZEA can reduce mitochondrial membrane potential, mitochondrial membrane damage, permeability transition, and cytochrome c oxidase inactivation in a dose-dependent manner. The inhibition of immunoglobulin secretion may result from the direct inhibition of B cells, reducing immunoglobulin production, given that plasma cells are responsible for immunoglobulin secretion after differentiation from B cells upon antigen stimulation [71,72].

2.3. DON

DON is absorbed primarily in the duodenum, which, thanks to its small-molecule properties, allows for passive transport into the osmotic bloodstream. Generally, DON interferes with the function of 5-hydroxytryptamine and catecholamine receptors in the small intestine and interferes with the normal peristaltic rhythm, resulting in a series of digestive disorders, such as nausea, vomiting, loss of appetite, and diarrhea [73].
Studies have shown that DON can reduce the transepithelial electrical resistance (TEER) of intestinal epithelial cell monolayer in a dose-dependent manner. In addition, DON downregulated the expression of tight junction proteins (ZO-1, E-cadherin, Occludin, and Claudin) in the intestinal epithelium both in vitro and in vivo, which may be regulated by the activation of the MAPK pathway [74,75]. DON has also been found to significantly increase the expressions of TNF-α, IL-1β, IFN-γ, IL-6, and other pro-inflammatory factors in the jejunums and ilea of piglets, affect cytokine and immune function, and damage the intestinal mucosal barrier [76]. Additionally, DON affects mucosal SIgA secretion and further damages the host defense against pathogens, indicating the adverse effects of DON on intestinal mucosal immunity [77].

2.4. FBs

The gastrointestinal absorption of FBs in pigs is limited, and the absorption rate of fumonisin-B1 (FB1) is only about 4%. The accumulation of FB1 was mainly distributed in the liver, kidneys, and spleen in pigs, and its distribution pattern was significantly affected by the dose and duration of exposure [78]. Specifically, during low-dose, long-term exposure, FB1 accumulated the most in the lungs, followed by the liver and kidneys. On the other hand, when a high dose was taken for a short period, the accumulation of the toxin in the liver was the first, followed by the kidneys, heart, and lungs, showing the regulation of the dose and time on the distribution of the toxin [79].
FB1, which is structurally similar to sphingosine, can inhibit the activity of N-acyltransferase, thereby interfering with sphingomyelin synthesis [80]. Sphingolipids, as a key component of the cell membrane, are involved in the regulation of cell life activities. FB competes with sphingosine N-2 acyltransferase due to its structural similarity, hindering sphingolipid metabolism and causing cell damage [81]. In addition, by interfering with sphingolipid-mediated cell signaling, FB1 can affect the activities of protein kinase C, phosphorylase D, and AP-1 transcription factors and interfere with cell adhesion, differentiation, proliferation, and even carcinogenesis [82,83].
Several studies have suggested that FB1 may mediate toxicity and carcinogenicity through oxidative stress. For example, when HepG2 cells were treated with 50 μM FB1 for 0, 12, and 24 h, the ROS content in treated cells was significantly higher than that in blank cells [42]. When 2.5 mg/kg FB 1 was exposed to the colon of mice, antioxidant enzymes such as SOD-1, SOD-2, GR, and GADPH in the FB 1-treated group were significantly lower than those in the control group. The contents of CYTP450, TRX, HSP70, and HSP90 were significantly higher than those in the control group [84].

2.5. OTA

OTA is mainly absorbed in the small intestine segment and undergoes a biotransformation process in the liver and kidneys of pigs [85]. After entering the blood, the OTA binds serum proteins in a bivalent form and maintains its long-term toxic effect through interaction with biomacromolecules and reabsorption in the kidneys and hepatic–intestinal circulation [86].
OTA inhibits protein synthesis by competitively binding to tRNA synthesis sites and affects phenylalanine access, thereby inhibiting t-RNA function [87]. Its promotion of lipid peroxidation may be related to the activation of NADPH and the enhancement of ascorbate-dependent lipid peroxidation, leading to the accumulation of malondialdehyde [88]. In the main target organ, the kidneys, OTA inhibits renal enzyme activity through the phenylalanine group, affecting renal function. OTA can also cause liver peroxidation, leading to hepatocyte apoptosis and dysfunction [89]. Its cytotoxic mechanism may involve competitive inhibition of mitochondrial carrier proteins, blocking mitochondrial phosphate transport and the respiratory chain and affecting mitochondrial function. In addition, OTA inhibits protein synthesis, reduces immunoglobulin secretion, reduces the activity of immune factors, and inhibits the proliferation and apoptosis of immune cells [90].

3. Study on the Application of Plant Extracts in Mycotoxin Poisoning

3.1. The Development Potential of Plant Extracts

Since ancient times, plants have played a central role in the prevention and treatment of diseases, a tradition that continues to this day. Despite the remarkable advances in modern medicine, many people in developing regions still rely on traditional remedies to cope with health problems, both for cultural heritage, economic, and efficacy reasons [91]. According to the World Health Organization, about 80% of the world’s population relies on traditional medicine for their basic medical needs, demonstrating the universal value of plant medicine [92]. Recent studies have witnessed a resurgence of interest in medicinal products derived from plants in both developed and developing societies, highlighting the vast resources and geographical diversity of plants for medicinal use, as well as the translation of traditional wisdom into modern medicines derived from plants [93].

3.2. Source and Composition of Plant Extracts

Plant extracts are the products of physical extraction or biological fermentation of the whole or parts of one or more natural plants as raw materials. They are usually derived from the roots, stems, leaves, flowers, fruits, or seeds of plants and contain a variety of phytochemicals. According to the main active ingredients, they can be divided into alkaloids, glycosides, flavonoids, organic acids, polysaccharides, polyphenols, and volatile oils [94]. Saponins are widely distributed in higher plant kingdoms and play important roles in Chinese herbal medicine, such as ginseng, Astragalus membranaceus, Panax notoginseng, and jujube seed. They show a variety of pharmacological activities, including anti-cancer, anti-tumor, antipyretic, detoxification, etc., which can play a beneficial role in the immunity, antioxidant digestive, and metabolic functions of livestock and poultry, thereby improving the performance of livestock and poultry [95]. Flavonoids, which are widely distributed in plants, play key roles in plant growth and development and have antibacterial and disease-prevention functions [96]. As widely distributed non-amino acid compounds, organic acids are widely present in plants with acidic characteristics, especially in fruits. Su Hong et al. used the Box–Behnkenken design to optimize the compound biological preservative of citric acid, thymol, and sodium alginate and provided a natural and efficient microbial inhibition scheme [97]. Polysaccharides are mainly distributed in plants, microorganisms, and marine organisms, especially in plant tissues. ZHAI et al. showed that pomegranate peel polysaccharides can significantly improve the activity of antioxidant enzymes in mice with CCI4-induced liver damage, suggesting their protective effect [98]. Hu Aijun’s team extracted polysaccharides from chickpeas with the assistance of ultrasound, which proved that both non-starchy and acidic polysaccharides had free radical scavenging ability, emphasizing the potential of polysaccharides in biological protection and treatment [99]. Polyphenols are a class of secondary metabolites that are widely present in plants, and most representatives of these compounds are characterized by interactions with reactive oxygen species [100]. In summary, the active components of the plant extracts were proven to have significant antioxidative and tissue-protective effects, showing potential to ameliorate mycotoxin-induced damage. Polyphenolic compounds, flavonoids, and other bioactive components in plant extracts can prevent the absorption of mycotoxins or reduce their biological activity by binding to mycotoxin molecules. Phenolics from beetroot may interfere with the absorption and bioavailability of mycotoxins in the gastrointestinal tract, potentially reducing their toxicity [101]. Pumpkin extract plays a significant role in mitigating the toxic effects of aflatoxin B1 and Ochratoxin A on neuronal differentiation. Specifically, the addition of pumpkin extract in contaminated breads significantly reduced the bio-accessibility of aflatoxin B1 and Ochratoxin A [102]. Moreover, some extracts can enhance the detoxification capacity in animals by regulating the activities of detoxifying enzymes. This study indicates that the phenolic compounds in algal extracts can aid in the detoxification of rats damaged by AFB1 [103].

3.3. Application of Plant Extracts

Plant extracts are rich in bioactive components with anti-mutagenic, antioxidant, and anti-cancer properties, which may effectively reduce the toxicity of mycotoxins. Their antioxidant mechanism involves scavenging free radicals, protecting cell membranes and macromolecules from free radical damage, and maintaining cell integrity [104]. Experimental research has substantiated that the elevated polyphenol content found in edamame and various common bean varieties enhances their antioxidant characteristics. The bioactive compounds present in these beans have exhibited potent antifungal and antitoxic properties, suggesting the capacity of legumes to counteract aflatoxin contamination by leveraging the antioxidative attributes of phenolic compounds [105]. Mutagenicity resistance is reflected in reducing gene variation induced by mutagens and maintaining genetic stability [106]. This study highlights the antioxidant and anti-mutagenic potential of Piper nigrum. The ability of phytocompounds to interact with DNA might reduce the interaction of mutagens and could be one of the possible mechanisms of the anti-mutagenic activity of P. nigrum extract [107]. On the anti-cancer side, plant extracts can activate and induce detoxification enzymes, interfere with key oncogenic signaling pathways (such as PI3K/Akt and Raf/MEK/ERK), and suppress tumor progression [108].
Interestingly, bioactive compounds in plants have been used as additives to prevent fungal growth and mycotoxin contamination in food and feed, thereby reducing the risk of mutagenicity and carcinogenicity of mycotoxins (Table 4). For instance, the potent inhibitory effect of essential oil on a variety of molds, including Aspergillus flavus and Aspergillus niger, and their toxins has been verified. Kocic-Tanackov et al. further demonstrated that a high concentration of Carum carvi L. essential oil treatment can completely block the production of aflatoxin [109]. It has also been reported that calabash extract, especially the polar extract with a high ferulic acid composition, has shown potential in inhibiting toxigenic fungi and reducing mycotoxin accumulation [110]. In addition, the high efficiency of plant-derived phenols such as chlorophyll in limiting the proliferation of Fusarium verticillioides and the production of its toxin, FB1, provides a solid theoretical basis for the development of biosafety strategies to prevent the penetration of harmful mycotoxins into the food chain [111]. Phenolic substances contained in soybeans have been proven to strengthen the defense system of antifungals and inhibit the formation of aflatoxin [112]. Shah Zaman conducted experimental tests to demonstrate the potential resistance of certain plant species (C. cyminum). The findings revealed a statistically significant and robust negative correlation between average mycotoxins and phytochemical concentrations [113].
Table 4. Overview of studies on the reduction of mycotoxin toxicity by a compound.
Table 4. Overview of studies on the reduction of mycotoxin toxicity by a compound.
Plant ExtractClassificationExperimental SubjectMechanismReferences
CurcuminPolyphenolDucksMiceIncreased jejunal tight junction protein mRNA and protein levels to protect the intestinal barrier and mitochondria from OTA-induced damageRegulation of Nrf2/p53-mediated apoptosis pathway and NF-kB/MLCK-mediated TJ pathway alleviates intestinal epithelial barrier damage induced by DON in mice[114,115]
ResveratrolPolyphenolIntestinal cellsActivation of the protein kinase-dependent pathway regulates IL-6 and IL-8 secretion to promote the assembly of claudin-4 in tight junction complexes to prevent DON-induced barrier dysfunction[116]
DihydromyricetinFlavoneIPEC-J2Alleviates cell damage caused by DON through its antioxidant activity, anti-inflammatory activity, or regulation of metabolic pathways[117]
Grape seed proanthocyanidin extractGlycosideRatsGSPE can alleviate the oxidative stress induced by AFB1 and significantly improve the immune damage induced by AFB1 in mice[118]
Red orange and lemon extractGlycosideRatsRLE attenuates OTA kidney injury caused by oxidative stress[119]
Flavonoid-rich fractions from Chromolena odorataFlavoneRatsAfb1-induced liver and intestinal injuries were ameliorated by changing the levels of pro-inflammatory cytokines, TNF-α, and IL-1β[120]
Ferulic acidOrganic acidRatsUpregulation of tight junction proteins, downregulation of ROCK, competition for CYP450 enzyme, and activation of GST attenuate AFB1-induced duodenal barrier injury in rats[121]
QuercetinPolyphenolMiceQuercetin alleviates intestinal injury induced by DON in mice by inhibiting the TLR4/NF-κB signaling pathway and ferroptosis[122]
Hericium mushroom polysaccharidePolysaccharideIPEC-J2It can significantly protect IPEC-J2 cells from DON-induced oxidative stress, inhibit DON-induced apoptosis, and reduce the production of reactive oxygen species (ROS)[123]
TheophyllineAlkaloidPigletsTo improve the intestinal barrier function and reduce inflammation, immunosuppression, and oxidative stress in piglets challenged with DON by regulating NF-κB/MAPK signaling pathway[124]
Melaleuca alternifoliaEssential oilSilver catfishElevated levels of ROS, LOOH, and PC in plasma and liver were avoided; in addition, TTO treatment attenuated aflatoxin-related liver injury[125]
Studies have also found that phenol-rich ginger extract (GE) can effectively reduce oxidative stress and liver injury caused by AFB1, and this protective effect may be related to the activation of the Nrf2/HO-1 signaling pathway [126]. A study demonstrated the protective effect of Tunisian radish extract (TRE) against immunosuppression induced by aflatoxin ZEN in Balb/c mice by quantifying flavonoids and isothiocyanates in TRE and measuring its antioxidant capacity [127]. Subsequently, onion, garlic extract, and eugenol were found to have significant effects on the growth and toxin synthesis of aflatoxin, especially the excellent toxin inhibition performance of onion extract [128]. In addition, carnosic acid (CA), a key polyphenol in rosemary, shows a powerful free radical scavenging effect and effectively alleviates AFB1-induced cytotoxicity and oxidative stress at the cellular level [129]. In addition, the antioxidant mechanism of total curcumin (TCMN) has been verified to significantly improve the adverse effects of AFB1 on broking chickens, restore serum biochemical indicators, and enhance the antioxidant capacity of the body, providing scientific evidence for the biological activities of the above plant extracts [130].
Scutellaria baicalensis root extract has been verified to specifically block the metabolic pathway of AFB1 in rat and human liver microsomes and reduce the generation of harmful metabolites by inhibiting CYP1A1/2 enzyme activity [131]. FWGE showed an efficient effect on alleviating the oxidative stress caused by DON and T-2 toxin in IPEC-J2 cells, reflecting its cytoprotective performance [132]. The ability of green tea extract to inhibit AFB1-induced transformation of liver cancer was confirmed across models, highlighting the value of anti-cancer components in water-soluble and ethanol extracts [133]. Agave plant extracts, especially flower parts, can significantly inhibit the production of aflatoxin and cyclopidonic acid [134]. The study of Capsantal and its mixture (Capsantal FS-30-NT) revealed its complex regulatory effects on the growth and toxin production of aflatoxin, and a low temperature was identified as the key intervention point [135]. In addition, studies have revealed that green tea extract and coumarins can regulate AFB1 metabolism in piglets and enhance detoxification, highlighting the positive utility of antioxidants in this process [136].
Currently, plant extracts have also achieved good application effects as alternatives in antimicrobial and detoxifying medications, such as common Chinese herbal medicines like licorice, bupleurum, coptis, balloon flower, isatis root, pulsatilla, and forsythia. A mixture of oregano and thyme, along with two commercial inoculants, was fermented. After 90 days of fermentation, it was indicated that the herbal extracts of oregano and thyme could be used to reduce the mycotoxin concentrations and improve the hygienic quality of corn silage [137]. The extract of Houttuynia cordata has been proven to completely replace certain antibiotics in animal diets, such as colistin sulfate or doxycycline calcium, and has been applied in large- and medium-sized enterprises for several years [138].

4. Limitations and Prospects

The increasing demand for plant extracts and their compounds has raised concerns about the safety, toxicity, and quality of these products. Therefore, a thorough screening of their toxicological properties is necessary. Future applications of plant extracts against mycotoxins should prioritize the development of techniques for producing safe and stable extracts. This includes comparing the similarities and differences among the active constituents of plant extracts that demonstrate effective detoxification properties and validating their precise detoxification mechanisms. Additionally, research should investigate the use of material coatings to deliver the extracts to targeted sites of action. It is also essential to determine the optimal concentrations for the application of these extracts under different conditions to minimize costs.
Research on plant extracts in the field of anti-mycotoxins has shown potential, but there are still several key gaps:
(1)
Most studies focus on superficial effects, such as antioxidation and anti-inflammation, but there is insufficient analysis of the molecular targets for detoxification, such as key enzyme inhibition and signaling pathway regulation, as well as the toxin metabolism pathways. There is a lack of systematic validation at the multi-omics level.
(2)
Active components (such as polyphenols and flavonoids) are prone to degradation under the influence of light, heat, and pH, and some extracts may cause unknown toxicities or interact with feed components. A long-term toxicological evaluation system needs to be established.
(3)
The extracts have low absorption rates and rapid metabolisms in animals, making it difficult to target the sites of toxin action (such as the intestines and liver). There is an urgent need to develop encapsulation technologies (such as nanocarriers) to improve delivery efficiency.
(4)
Existing research mostly focuses on single toxins, whereas actual contamination often involves multiple mycotoxins coexisting. The synergistic detoxification effects and mechanisms of plant extracts on complex contamination have not yet been clarified.
(5)
The extraction process of high-purity active components is complex and costly, and it is limited by the regional and seasonal availability of plant resources. There is a need to optimize low-cost, sustainable industrial production schemes.
(6)
There are no uniform quality control standards for the active components of different extracts, and there is a lack of research on the dose-effect relationship between active ingredients and detoxification effects, which affects the reliability of practical applications.
In the future, it will be necessary to combine metabolomics, targeted delivery technologies, and interdisciplinary collaboration to break through the aforementioned bottlenecks and promote the transformation of plant extracts from basic research to industrial application.

Author Contributions

Conceptualization, X.Z., K.Y. and Q.J.; investigation, X.Z., J.C., X.M., X.T. and Q.J.; methodology, X.Z., P.L., K.Y. and Q.J.; visualization, X.Z., P.L., K.Y. and Q.J.; project administration, J.C., X.M., X.T., B.T., P.L., K.Y. and Q.J.; writing—original draft, X.Z. and Q.J.; writing—review and editing, X.Z., J.C., X.M., X.T., B.T., P.L., K.Y. and Q.J.; funding acquisition, P.L., K.Y. and Q.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Key Research and Development Program (2023YFD1301003), the National Natural Science Foundation (32302794; 32372926), the “Shen Nong Scholar Funding of Hunan Agricultural University”, “the Key Research fund from the Science and Technology Department of Yunnan Province (202202AE090032)”, the Earmarked Fund for China Agriculture Research System (CARS-37), and the “China Agriculture Research System of MOF and MARA”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Khan, R. Mycotoxins in food: Occurrence, health implications, and control strategies-A comprehensive review. Toxicon 2024, 248, 108038. [Google Scholar] [CrossRef]
  2. Makhuvele, R.; Naidu, K.; Gbashi, S.; Thipe, V.C.; Adebo, O.A.; Njobeh, P.B. The use of plant extracts and their phytochemicals for control of toxigenic fungi and mycotoxins. Heliyon 2020, 6, e05291. [Google Scholar] [CrossRef] [PubMed]
  3. Raters, M.; Matissek, R. Thermal stability of aflatoxin B1 and ochratoxin A. Mycotoxin Res. 2008, 24, 130–134. [Google Scholar] [CrossRef] [PubMed]
  4. Li, Y.; Gao, H.; Wang, R.; Xu, Q. Deoxynivalenol in food and feed: Recent advances in decontamination strategies. Front. Microbiol. 2023, 14, 1141378. [Google Scholar] [CrossRef]
  5. Awuchi, C.G.; Ondari, E.N.; Nwozo, S.; Odongo, G.A.; Eseoghene, I.J.; Twinomuhwezi, H.; Ogbonna, C.U.; Upadhyay, A.K.; Adeleye, A.O.; Okpala, C.O.R. Mycotoxins’ Toxicological Mechanisms Involving Humans, Livestock and Their Associated Health Concerns: A Review. Toxins 2022, 14, 167. [Google Scholar] [CrossRef]
  6. Murtaza, B.; Wang, L.; Li, X.; Nawaz, M.Y.; Saleemi, M.K.; Khatoon, A.; Yongping, X. Recalling the reported toxicity assessment of deoxynivalenol, mitigating strategies and its toxicity mechanisms: Comprehensive review. Chem.-Biol. Interact. 2024, 387, 110799. [Google Scholar] [CrossRef]
  7. Lv, Q.; Xu, W.; Yang, F.; Wei, W.; Chen, X.; Zhang, Z.; Liu, Y. Reproductive Toxicity of Zearalenone and Its Molecular Mechanisms: A Review. Molecules 2025, 30, 505. [Google Scholar] [CrossRef]
  8. Kamle, M.; Mahato, D.K.; Devi, S.; Lee, K.E.; Kang, S.G.; Kumar, P. Fumonisins: Impact on Agriculture, Food, and Human Health and their Management Strategies. Toxins 2019, 11, 328. [Google Scholar] [CrossRef]
  9. Banahene, J.C.M.; Ofosu, I.W.; Odai, B.T.; Lutterodt, H.E.; Agyemang, P.A.; Ellis, W.O. Ochratoxin A in food commodities: A review of occurrence, toxicity, and management strategies. Heliyon 2024, 10, e39313. [Google Scholar] [CrossRef]
  10. Zhang, Z.; McCullough, C. Survey on the Content of Mycotoxins in Newly Harvested Feeds and Forages in Europe, 2021. Today Pig Ind. 2022, 1, 66–68. [Google Scholar]
  11. Patyal, A.; Gill, J.P.S.; Bedi, J.S.; Aulakh, R.S. Assessment of aflatoxin contamination in dairy animal concentrate feed from Punjab, India. Environ. Sci. Pollut. Res. Int. 2021, 28, 37705–37715. [Google Scholar] [CrossRef] [PubMed]
  12. Zhao, L.; Zhang, L.; Xu, Z.; Liu, X.; Chen, L.; Dai, J.; Karrow, N.A.; Sun, L. Occurrence of Aflatoxin B(1), deoxynivalenol and zearalenone in feeds in China during 2018–2020. J. Anim. Sci. Biotechnol. 2021, 12, 74. [Google Scholar] [CrossRef]
  13. Hao, W.; Guan, S.; Li, A.; Wang, J.; An, G.; Hofstetter, U.; Schatzmayr, G. Mycotoxin Occurrence in Feeds and Raw Materials in China: A Five-Year Investigation. Toxins 2023, 15, 63. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Y.; Yang, Y.; Qi, S.; Zhou, J.; Wang, C.; Zheng, W.; Lei, Y.; Ji, C. Survey on the Mycotoxin Contamination in Domestic Feed. and Feed. Ingredients in 2021. Feed. Ind. 2022, 43, 55–58. [Google Scholar] [CrossRef]
  15. Cargill’s 2023 Global Mycotoxin Report. Available online: https://mycotoxins.com/home (accessed on 6 February 2024).
  16. Gruber-Dorninger, C.; Jenkins, T.; Schatzmayr, G. Global Mycotoxin Occurrence in Feed: A Ten-Year Survey. Toxins 2019, 11, 375. [Google Scholar] [CrossRef]
  17. van der Fels-Klerx, H.J.; Goedhart, P.W.; Elen, O.; Börjesson, T.; Hietaniemi, V.; Booij, C.J. Modeling deoxynivalenol contamination of wheat in northwestern Europe for climate change assessments. J. Food Prot. 2012, 75, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  18. Tian, M.; Feng, Y.; He, X.; Zhang, D.; Wang, W.; Liu, D. Mycotoxins in livestock feed in China—Current status and future challenges. Toxicon 2022, 214, 112–120. [Google Scholar] [CrossRef]
  19. Yang, C.; Song, G.; Lim, W. Effects of mycotoxin-contaminated feed on farm animals. J. Hazard. Mater. 2020, 389, 122087. [Google Scholar] [CrossRef]
  20. Ivanovics, B.; Gazsi, G.; Reining, M.; Berta, I.; Poliska, S.; Toth, M.; Domokos, A.; Nagy, B., Jr.; Staszny, A.; Cserhati, M.; et al. Embryonic exposure to low concentrations of aflatoxin B1 triggers global transcriptomic changes, defective yolk lipid mobilization, abnormal gastrointestinal tract development and inflammation in zebrafish. J. Hazard. Mater. 2021, 416, 125788. [Google Scholar] [CrossRef]
  21. Mahato, D.K.; Lee, K.E.; Kamle, M.; Devi, S.; Dewangan, K.N.; Kumar, P.; Kang, S.G. Aflatoxins in Food and Feed: An Overview on Prevalence, Detection and Control Strategies. Front. Microbiol. 2019, 10, 2266. [Google Scholar] [CrossRef]
  22. Liu, X.; Kumar Mishra, S.; Wang, T.; Xu, Z.; Zhao, X.; Wang, Y.; Yin, H.; Fan, X.; Zeng, B.; Yang, M.; et al. AFB1 Induced Transcriptional Regulation Related to Apoptosis and Lipid Metabolism in Liver of Chicken. Toxins 2020, 12, 290. [Google Scholar] [CrossRef] [PubMed]
  23. Loi, M.; Fanelli, F.; Liuzzi, V.C.; Logrieco, A.F.; Mulè, G. Mycotoxin Biotransformation by Native and Commercial Enzymes: Present and Future Perspectives. Toxins 2017, 9, 111. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, F.; Groopman, J.D.; Pestka, J.J. Public health impacts of foodborne mycotoxins. Annu. Rev. Food Sci. Technol. 2014, 5, 351–372. [Google Scholar] [CrossRef]
  25. Benkerroum, N. Chronic and Acute Toxicities of Aflatoxins: Mechanisms of Action. Int. J. Environ. Res. Public Health 2020, 17, 423. [Google Scholar] [CrossRef] [PubMed]
  26. Rawal, S.; Kim, J.E.; Coulombe, R., Jr. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Res. Vet. Sci. 2010, 89, 325–331. [Google Scholar] [CrossRef]
  27. Silvotti, L.; Petterino, C.; Bonomi, A.; Cabassi, E. Immunotoxicological effects on piglets of feeding sows diets containing aflatoxins. Vet. Rec. 1997, 141, 469–472. [Google Scholar] [CrossRef]
  28. Jiang, Y.; Ogunade, I.M.; Vyas, D.; Adesogan, A.T. Aflatoxin in Dairy Cows: Toxicity, Occurrence in Feedstuffs and Milk and Dietary Mitigation Strategies. Toxins 2021, 13, 283. [Google Scholar] [CrossRef] [PubMed]
  29. Tassis, P.D.; Reisinger, N.; Nagl, V.; Tzika, E.; Schatzmayr, D.; Mittas, N.; Basioura, A.; Michos, I.; Tsakmakidis, I.A. Comparative Effects of Deoxynivalenol, Zearalenone and Its Modified Forms De-Epoxy-Deoxynivalenol and Hydrolyzed Zearalenone on Boar Semen In Vitro. Toxins 2022, 14, 497. [Google Scholar] [CrossRef]
  30. Wu, K.; Ren, C.; Gong, Y.; Gao, X.; Rajput, S.A.; Qi, D.; Wang, S. The insensitive mechanism of poultry to zearalenone: A review. Anim. Nutr. (Zhongguo Xu Mu Shou Yi Xue Hui) 2021, 7, 587–594. [Google Scholar] [CrossRef]
  31. Hooft, J.M.; Bureau, D.P. Deoxynivalenol: Mechanisms of action and its effects on various terrestrial and aquatic species. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2021, 157, 112616. [Google Scholar] [CrossRef]
  32. Dersjant-Li, Y.; Verstegen, M.W.; Gerrits, W.J. The impact of low concentrations of aflatoxin, deoxynivalenol or fumonisin in diets on growing pigs and poultry. Nutr. Res. Rev. 2003, 16, 223–239. [Google Scholar] [CrossRef] [PubMed]
  33. Holanda, D.M.; Yiannikouris, A.; Kim, S.W. Investigation of the Efficacy of a Postbiotic Yeast Cell Wall-Based Blend on Newly-Weaned Pigs under a Dietary Challenge of Multiple Mycotoxins with Emphasis on Deoxynivalenol. Toxins 2020, 12, 504. [Google Scholar] [CrossRef]
  34. Metayer, J.P.; Travel, A.; Mika, A.; Bailly, J.D.; Cleva, D.; Boissieu, C.; Guennec, J.L.; Froment, P.; Albaric, O.; Labrut, S.; et al. Lack of Toxic Interaction Between Fusariotoxins in Broiler Chickens Fed throughout Their Life at the Highest Level Tolerated in the European Union. Toxins 2019, 11, 455. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, J.H.; Park, G.H.; Han, G.P.; Kil, D.Y. Effect of feeding corn distillers dried grains with solubles naturally contaminated with deoxynivalenol on growth performance, meat quality, intestinal permeability, and utilization of energy and nutrients in broiler chickens. Poult. Sci. 2021, 100, 101215. [Google Scholar] [CrossRef] [PubMed]
  36. Dong, J.N.; Zhao, Z.K.; Wang, Z.Q.; Li, S.Z.; Zhang, Y.P.; Sun, Z.; Qin, G.X.; Zhang, X.F.; Zhao, W.; Aschalew, N.D.; et al. Impact of deoxynivalenol on rumen function, production, and health of dairy cows: Insights from metabolomics and microbiota analysis. J. Hazard. Mater. 2024, 465, 133376. [Google Scholar] [CrossRef]
  37. Loh, Z.H.; Ouwerkerk, D.; Klieve, A.V.; Hungerford, N.L.; Fletcher, M.T. Toxin Degradation by Rumen Microorganisms: A Review. Toxins 2020, 12, 664. [Google Scholar] [CrossRef]
  38. Wang, G.; Du, Y. The Impact of Vomitoxin on Ruminant Animals. Feed. Ind. 2016, 37, 58–61. [Google Scholar] [CrossRef]
  39. Li, W.; He, Y.; Zhao, H.; Peng, L.; Li, J.; Rui, R.; Ju, S. Grape Seed Proanthocyanidin Ameliorates FB(1)-Induced Meiotic Defects in Porcine Oocytes. Toxins 2021, 13, 841. [Google Scholar] [CrossRef] [PubMed]
  40. Gbore, F.A. Reproductive organ weights and semen quality of pubertal boars fed dietary fumonisin B1. Anim. Int. J. Anim. Biosci. 2009, 3, 1133–1137. [Google Scholar] [CrossRef]
  41. Wang, Y.; Quan, H.; Li, X.; Li, Q.; Haque, M.A.; Shi, Q.; Fu, Q.; He, C. Contamination With Fumonisin B and Deoxynivalenol Is a Threat to Egg Safety and Contributes to Gizzard Ulcerations of Newborn Chickens. Front. Microbiol. 2021, 12, 676671. [Google Scholar] [CrossRef]
  42. Singh, M.P.; Kang, S.C. Endoplasmic reticulum stress-mediated autophagy activation attenuates fumonisin B1 induced hepatotoxicity in vitro and in vivo. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2017, 110, 371–382. [Google Scholar] [CrossRef] [PubMed]
  43. Henry, M.H.; Wyatt, R.D. The toxicity of fumonisin B1, B2, and B3, individually and in combination, in chicken embryos. Poult. Sci. 2001, 80, 401–407. [Google Scholar] [CrossRef]
  44. Zacharias, C.; van Echten-Deckert, G.; Wang, E.; Merrill, A.H., Jr.; Sandhoff, K. The effect of fumonisin B1 on developing chick embryos: Correlation between de novo sphingolipid biosynthesis and gross morphological changes. Glycoconj. J. 1996, 13, 167–175. [Google Scholar] [CrossRef]
  45. Denli, M.; Perez, J.F. Ochratoxins in feed, a risk for animal and human health: Control strategies. Toxins 2010, 2, 1065–1077. [Google Scholar] [CrossRef]
  46. Tao, Y.; Xie, S.; Xu, F.; Liu, A.; Wang, Y.; Chen, D.; Pan, Y.; Huang, L.; Peng, D.; Wang, X.; et al. Ochratoxin A: Toxicity, oxidative stress and metabolism. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2018, 112, 320–331. [Google Scholar] [CrossRef]
  47. Stoev, S.D.; Paskalev, M.; MacDonald, S.; Mantle, P.G. Experimental one year ochratoxin A toxicosis in pigs. Exp. Toxicol. Pathol. Off. J. Ges. Fur Toxikol. Pathol. 2002, 53, 481–487. [Google Scholar] [CrossRef] [PubMed]
  48. Liu, W.C.; Pushparaj, K.; Meyyazhagan, A.; Arumugam, V.A.; Pappuswamy, M.; Bhotla, H.K.; Baskaran, R.; Issara, U.; Balasubramanian, B.; Mousavi Khaneghah, A. Ochratoxin A as an alarming health threat for livestock and human: A review on molecular interactions, mechanism of toxicity, detection, detoxification, and dietary prophylaxis. Toxicon 2022, 213, 59–75. [Google Scholar] [CrossRef] [PubMed]
  49. Murugesan, G.R.; Ledoux, D.R.; Naehrer, K.; Berthiller, F.; Applegate, T.J.; Grenier, B.; Phillips, T.D.; Schatzmayr, G. Prevalence and effects of mycotoxins on poultry health and performance, and recent development in mycotoxin counteracting strategies. Poult. Sci. 2015, 94, 1298–1315. [Google Scholar] [CrossRef]
  50. Mobashar, M.; Hummel, J.; Blank, R.; Südekum, K.H. Ochratoxin A in ruminants—A review on its degradation by gut microbes and effects on animals. Toxins 2010, 2, 809–839. [Google Scholar] [CrossRef]
  51. Kumar, V.; Bahuguna, A.; Ramalingam, S.; Dhakal, G.; Shim, J.J.; Kim, M. Recent technological advances in mechanism, toxicity, and food perspectives of enzyme-mediated aflatoxin degradation. Crit. Rev. Food Sci. Nutr. 2022, 62, 5395–5412. [Google Scholar] [CrossRef]
  52. Jaćević, V.; Dumanović, J.; Alomar, S.Y.; Resanović, R.; Milovanović, Z.; Nepovimova, E.; Wu, Q.; Franca, T.C.C.; Wu, W.; Kuča, K. Research update on aflatoxins toxicity, metabolism, distribution, and detection: A concise overview. Toxicology 2023, 492, 153549. [Google Scholar] [CrossRef] [PubMed]
  53. Kumagai, S. Intestinal absorption and excretion of aflatoxin in rats. Toxicol. Appl. Pharmacol. 1989, 97, 88–97. [Google Scholar] [CrossRef]
  54. Min, L.; Fink-Gremmels, J.; Li, D.; Tong, X.; Tang, J.; Nan, X.; Yu, Z.; Chen, W.; Wang, G. An overview of aflatoxin B1 biotransformation and aflatoxin M1 secretion in lactating dairy cows. Anim. Nutr. (Zhongguo Xu Mu Shou Yi Xue Hui) 2021, 7, 42–48. [Google Scholar] [CrossRef]
  55. Deng, J.; Zhao, L.; Zhang, N.Y.; Karrow, N.A.; Krumm, C.S.; Qi, D.S.; Sun, L.H. Aflatoxin B(1) metabolism: Regulation by phase I and II metabolizing enzymes and chemoprotective agents. Mutat. Res. Rev. Mutat. Res. 2018, 778, 79–89. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, L.; Huang, Q.; Wu, J.; Wu, W.; Jiang, J.; Yan, H.; Huang, J.; Sun, Y.; Deng, Y. The metabolism and biotransformation of AFB(1): Key enzymes and pathways. Biochem. Pharmacol. 2022, 199, 115005. [Google Scholar] [CrossRef]
  57. Rushing, B.R.; Selim, M.I. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 124, 81–100. [Google Scholar] [CrossRef]
  58. Eaton, D.L.; Williams, D.E.; Coulombe, R.A. Species Differences in the Biotransformation of Aflatoxin B1: Primary Determinants of Relative Carcinogenic Potency in Different Animal Species. Toxins 2025, 17, 30. [Google Scholar] [CrossRef]
  59. Su, J.J.; Ban, K.C.; Li, Y.; Qin, L.L.; Wang, H.Y.; Yang, C.; Ou, C.; Duan, X.X.; Lee, Y.L.; Yang, R.Q. Alteration of p53 and p21 during hepatocarcinogenesis in tree shrews. World J. Gastroenterol. 2004, 10, 3559–3563. [Google Scholar] [CrossRef] [PubMed]
  60. Saha Turna, N.; Comstock, S.S.; Gangur, V.; Wu, F. Effects of aflatoxin on the immune system: Evidence from human and mammalian animal research. Crit. Rev. Food Sci. Nutr. 2024, 64, 9955–9973. [Google Scholar] [CrossRef]
  61. He, X.N.; Zeng, Z.Z.; Wu, P.; Jiang, W.D.; Liu, Y.; Jiang, J.; Kuang, S.Y.; Tang, L.; Feng, L.; Zhou, X.Q. Dietary Aflatoxin B1 attenuates immune function of immune organs in grass carp (Ctenopharyngodon idella) by modulating NF-κB and the TOR signaling pathway. Front. Immunol. 2022, 13, 1027064. [Google Scholar] [CrossRef]
  62. Lyman, B.A.; Erki, L.; Biedrzycka, D.W.; Devlin, T.M.; Ch’ih, J.J. Modification of protein synthetic components by aflatoxin B1. Biochem. Pharmacol. 1988, 37, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  63. Pai, M.R.; Bai, N.J.; Venkitasubramanian, T.A. Aflatoxin induced inhibition of protein synthesis. Toxicon 1978, 16, 283–287. [Google Scholar] [CrossRef]
  64. Han, S.H.; Jeon, Y.J.; Yea, S.S.; Yang, K.H. Suppression of the interleukin-2 gene expression by aflatoxin B1 is mediated through the down-regulation of the NF-AT and AP-1 transcription factors. Toxicol. Lett. 1999, 108, 1–10. [Google Scholar] [CrossRef]
  65. Yang, S.; Zhang, H.; Sun, F.; De Ruyck, K.; Zhang, J.; Jin, Y.; Li, Y.; Wang, Z.; Zhang, S.; De Saeger, S.; et al. Metabolic Profile of Zearalenone in Liver Microsomes from Different Species and Its in Vivo Metabolism in Rats and Chickens Using Ultra High-Pressure Liquid Chromatography-Quadrupole/Time-of-Flight Mass Spectrometry. J. Agric. Food Chem. 2017, 65, 11292–11303. [Google Scholar] [CrossRef] [PubMed]
  66. Pfeiffer, E.; Kommer, A.; Dempe, J.S.; Hildebrand, A.A.; Metzler, M. Absorption and metabolism of the mycotoxin zearalenone and the growth promotor zeranol in Caco-2 cells in vitro. Mol. Nutr. Food Res. 2011, 55, 560–567. [Google Scholar] [CrossRef]
  67. Ropejko, K.; Twarużek, M. Zearalenone and Its Metabolites-General Overview, Occurrence, and Toxicity. Toxins 2021, 13, 35. [Google Scholar] [CrossRef]
  68. Tatay, E.; Espín, S.; García-Fernández, A.J.; Ruiz, M.J. Estrogenic activity of zearalenone, α-zearalenol and β-zearalenol assessed using the E-screen assay in MCF-7 cells. Toxicol. Mech. Methods 2018, 28, 239–242. [Google Scholar] [CrossRef]
  69. Zhou, J.; Zhao, L.; Huang, S.; Liu, Q.; Ao, X.; Lei, Y.; Ji, C.; Ma, Q. Zearalenone toxicosis on reproduction as estrogen receptor selective modulator and alleviation of zearalenone biodegradative agent in pregnant sows. J. Anim. Sci. Biotechnol. 2022, 13, 36. [Google Scholar] [CrossRef]
  70. Takemura, H.; Shim, J.Y.; Sayama, K.; Tsubura, A.; Zhu, B.T.; Shimoi, K. Characterization of the estrogenic activities of zearalenone and zeranol in vivo and in vitro. J. Steroid Biochem. Mol. Biol. 2007, 103, 170–177. [Google Scholar] [CrossRef]
  71. Fan, W.; Shen, T.; Ding, Q.; Lv, Y.; Li, L.; Huang, K.; Yan, L.; Song, S. Zearalenone induces ROS-mediated mitochondrial damage in porcine IPEC-J2 cells. J. Biochem. Mol. Toxicol. 2017, 31, e21944. [Google Scholar] [CrossRef]
  72. Feng, Y.Q.; Zhao, A.H.; Wang, J.J.; Tian, Y.; Yan, Z.H.; Dri, M.; Shen, W.; De Felici, M.; Li, L. Oxidative stress as a plausible mechanism for zearalenone to induce genome toxicity. Gene 2022, 829, 146511. [Google Scholar] [CrossRef] [PubMed]
  73. Gershon, M.D. 5-Hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 14–21. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. Zhao, X.; Dong, B.; Friesen, M.; Liu, S.; Zhu, C.; Yang, C. Capsaicin Attenuates Lipopolysaccharide-Induced Inflammation and Barrier Dysfunction in Intestinal Porcine Epithelial Cell Line-J2. Front. Physiol. 2021, 12, 715469. [Google Scholar] [CrossRef]
  76. Gao, H.; Liu, L.; Zhao, Y.; Hara, H.; Chen, P.; Xu, J.; Tang, J.; Wei, L.; Li, Z.; Cooper, D.K.C.; et al. Human IL-6, IL-17, IL-1β, and TNF-α differently regulate the expression of pro-inflammatory related genes, tissue factor, and swine leukocyte antigen class I in porcine aortic endothelial cells. Xenotransplantation 2017, 24, e12291. [Google Scholar] [CrossRef]
  77. Macpherson, A.J.; Geuking, M.B.; Slack, E.; Hapfelmeier, S.; McCoy, K.D. The habitat, double life, citizenship, and forgetfulness of IgA. Immunol. Rev. 2012, 245, 132–146. [Google Scholar] [CrossRef] [PubMed]
  78. Fodor, J.; Balogh, K.; Weber, M.; Miklós, M.; Kametler, L.; Pósa, R.; Mamet, R.; Bauer, J.; Horn, P.; Kovács, F.; et al. Absorption, distribution and elimination of fumonisin B(1) metabolites in weaned piglets. Food Addit. Contam. Part. A Chem. Anal. Control Expo. Risk Assess. 2008, 25, 88–96. [Google Scholar] [CrossRef] [PubMed]
  79. Szabó, A.; Szabó-Fodor, J.; Kachlek, M.; Mézes, M.; Balogh, K.; Glávits, R.; Ali, O.; Zeebone, Y.Y.; Kovács, M. Dose and Exposure Time-Dependent Renal and Hepatic Effects of Intraperitoneally Administered Fumonisin B1 in Rats. Toxins 2018, 10, 465. [Google Scholar] [CrossRef]
  80. Chen, J.; Wen, J.; Tang, Y.; Shi, J.; Mu, G.; Yan, R.; Cai, J.; Long, M. Research Progress on Fumonisin B1 Contamination and Toxicity: A Review. Molecules 2021, 26, 5238. [Google Scholar] [CrossRef]
  81. Li, H.; Wang, M.; Kang, W.; Lin, Z.; Gan, F.; Huang, K. Non-cytotoxic dosage of fumonisin B1 aggravates ochratoxin A-induced nephrocytotoxicity and apoptosis via ROS-dependent JNK/MAPK signaling pathway. Toxicology 2021, 457, 152802. [Google Scholar] [CrossRef]
  82. Burgess, K.M.; Renaud, J.B.; McDowell, T.; Sumarah, M.W. Mechanistic Insight into the Biosynthesis and Detoxification of Fumonisin Mycotoxins. ACS Chem. Biol. 2016, 11, 2618–2625. [Google Scholar] [CrossRef] [PubMed]
  83. Hannun, Y.A.; Obeid, L.M. Sphingolipids and their metabolism in physiology and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 175–191. [Google Scholar] [CrossRef] [PubMed]
  84. Kim, S.H.; Singh, M.P.; Sharma, C.; Kang, S.C. Fumonisin B1 actuates oxidative stress-associated colonic damage via apoptosis and autophagy activation in murine model. J. Biochem. Mol. Toxicol. 2018, 32, e22161. [Google Scholar] [CrossRef]
  85. Schrenk, D.; Bignami, M.; Bodin, L.; Chipman, J.K.; Del Mazo, J.; Grasl-Kraupp, B.; Hogstrand, C.; Hoogenboom, L.R.; Leblanc, J.C.; Nielsen, E.; et al. Risks for animal health related to the presence of ochratoxin A (OTA) in feed. EFSA J. Eur. Food Saf. Auth. 2023, 21, e08375. [Google Scholar] [CrossRef]
  86. Hagelberg, S.; Hult, K.; Fuchs, R. Toxicokinetics of ochratoxin A in several species and its plasma-binding properties. J. Appl. Toxicol. JAT 1989, 9, 91–96. [Google Scholar] [CrossRef]
  87. Creppy, E.E.; Størmer, F.C.; Kern, D.; Röschenthaler, R.; Dirheimer, G. Effects of ochratoxin A metabolites on yeast phenylalanyl-tRNA synthetase and on the growth and in vivo protein synthesis of hepatoma cells. Chem.-Biol. Interact. 1983, 47, 239–247. [Google Scholar] [CrossRef]
  88. Huang, X.; Gao, Y.; Li, S.; Wu, C.; Wang, J.; Zheng, N. Modulation of Mucin (MUC2, MUC5AC and MUC5B) mRNA Expression and Protein Production and Secretion in Caco-2/HT29-MTX Co-Cultures Following Exposure to Individual and Combined Aflatoxin M1 and Ochratoxin A. Toxins 2019, 11, 132. [Google Scholar] [CrossRef]
  89. Al-Anati, L.; Petzinger, E. Immunotoxic activity of ochratoxin A. J. Vet. Pharmacol. Ther. 2006, 29, 79–90. [Google Scholar] [CrossRef]
  90. Aleo, M.D.; Wyatt, R.D.; Schnellmann, R.G. Mitochondrial dysfunction is an early event in ochratoxin A but not oosporein toxicity to rat renal proximal tubules. Toxicol. Appl. Pharmacol. 1991, 107, 73–80. [Google Scholar] [CrossRef]
  91. Sofowora, A.; Ogunbodede, E.; Onayade, A. The role and place of medicinal plants in the strategies for disease prevention. Afr. J. Tradit. Complement. Altern. Med. AJTCAM 2013, 10, 210–229. [Google Scholar] [CrossRef]
  92. Mumtaz, A.; Ashfaq, U.A.; Ul Qamar, M.T.; Anwar, F.; Gulzar, F.; Ali, M.A.; Saari, N.; Pervez, M.T. MPD3: A useful medicinal plants database for drug designing. Nat. Prod. Res. 2017, 31, 1228–1236. [Google Scholar] [CrossRef] [PubMed]
  93. Pandita, D.; Pandita, A.; Wani, S.H.; Abdelmohsen, S.A.M.; Alyousef, H.A.; Abdelbacki, A.M.M.; Al-Yafrasi, M.A.; Al-Mana, F.A.; Elansary, H.O. Crosstalk of Multi-Omics Platforms with Plants of Therapeutic Importance. Cells 2021, 10, 1296. [Google Scholar] [CrossRef]
  94. Sarker, S.D.; Nahar, L. An introduction to natural products isolation. Methods Mol. Biol. 2012, 864, 1–25. [Google Scholar] [CrossRef] [PubMed]
  95. Rao, A.V.; Gurfinkel, D.M. The bioactivity of saponins: Triterpenoid and steroidal glycosides. Drug Metab. Drug Interact. 2000, 17, 211–235. [Google Scholar] [CrossRef]
  96. Cao, Y.; Xie, L.; Liu, K.; Liang, Y.; Dai, X.; Wang, X.; Lu, J.; Zhang, X.; Li, X. The antihypertensive potential of flavonoids from Chinese Herbal Medicine: A review. Pharmacol. Res. 2021, 174, 105919. [Google Scholar] [CrossRef] [PubMed]
  97. Su, H.; Shen, L.; Bi, S.; Zhang, X.; Guo, R.; Liu, H. The Preservation Effect of Composite Biopreservatives Combined with Ice Temperature Storage on Redfin Tetrodotoxin. J. Fish. Sci. 2019, 43, 688–696. [Google Scholar]
  98. Zhai, X.; Zhu, C.; Zhang, Y.; Sun, J.; Alim, A.; Yang, X. Chemical characteristics, antioxidant capacities and hepatoprotection of polysaccharides from pomegranate peel. Carbohydr. Polym. 2018, 202, 461–469. [Google Scholar] [CrossRef]
  99. Hu, A.; Li, Y.; Li, Z.; Wang, H.; Zhao, C.; Ma, L. Ultrasonic Extraction of Non-starch Polysaccharides from Chickpeas and Their Antioxidant Activities. China Food Addit. 2018, 6, 133–138. [Google Scholar]
  100. Bolat, E.; Sarıtaş, S.; Duman, H.; Eker, F.; Akdaşçi, E.; Karav, S.; Witkowska, A.M. Polyphenols: Secondary Metabolites with a Biological Impression. Nutrients 2024, 16, 2550. [Google Scholar] [CrossRef]
  101. Paula, L.; Ana, J.G.; Hannu, P.; Carlos, M.J.; Eeva-Riikka, V.; Cristina, J. Role of red beetroot in bread for reducing mycotoxin risks: Bioavailability of beetroot polyphenols and betalains with ochratoxin a, aflatoxin B1 and zearalenone in Caco-2 cells. Food Chem. 2025, 465, 142036. [Google Scholar] [CrossRef]
  102. Frangiamone, M.; Alonso-Garrido, M.; Font, G.; Cimbalo, A.; Manyes, L. Pumpkin extract and fermented whey individually and in combination alleviated AFB1- and OTA-induced alterations on neuronal differentiation invitro. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2022, 164, 113011. [Google Scholar] [CrossRef]
  103. Marrez, D.A.; Badr, A.N.; El-Bahrawy, A.; Naeem, M.A. Algal extracts evaluation as an Antitoxicity sustainable solution against aflatoxin B(1) toxicity in rat tissues. Toxicon 2024, 250, 108098. [Google Scholar] [CrossRef] [PubMed]
  104. Ahmad Nejhad, A.; Alizadeh Behbahani, B.; Hojjati, M.; Vasiee, A.; Mehrnia, M.A. Identification of phytochemical, antioxidant, anticancer and antimicrobial potential of Calotropis procera leaf aqueous extract. Sci. Rep. 2023, 13, 14716. [Google Scholar] [CrossRef] [PubMed]
  105. León-Cortés, D.; Arce-Villalobos, K.; Bogantes-Ledezma, D.; Irías-Mata, A.; Chaves-Barrantes, N.; Vinas, M. Anti-aflatoxin potential of phenolic compounds from common beans (Phaseolus vulgaris L.). Food Chem. 2025, 469, 142597. [Google Scholar] [CrossRef]
  106. Makhafola, T.J.; Elgorashi, E.E.; McGaw, L.J.; Verschaeve, L.; Eloff, J.N. The correlation between antimutagenic activity and total phenolic content of extracts of 31 plant species with high antioxidant activity. BMC Complement. Altern. Med. 2016, 16, 490. [Google Scholar] [CrossRef]
  107. Zahin, M.; Bokhari, N.A.; Ahmad, I.; Husain, F.M.; Althubiani, A.S.; Alruways, M.W.; Perveen, K.; Shalawi, M. Antioxidant, antibacterial, and antimutagenic activity of Piper nigrum seeds extracts. Saudi J. Biol. Sci. 2021, 28, 5094–5105. [Google Scholar] [CrossRef]
  108. Manogaran, P.; Beeraka, N.M.; Paulraj, R.S.; Sathiyachandran, P.; Thammaiappa, M. Impediment of Cancer by Dietary Plant-derived Alkaloids Through Oxidative Stress: Implications of PI3K/AKT Pathway in Apoptosis, Autophagy, and Ferroptosis. Curr. Top. Med. Chem. 2023, 23, 860–877. [Google Scholar] [CrossRef]
  109. Maurya, A.; Kumar, S.; Singh, B.K.; Chaudhari, A.K.; Dwivedy, A.K.; Prakash, B.; Dubey, N.K. Mechanistic investigations on antifungal and antiaflatoxigenic activities of chemically characterised Carum carvi L. essential oil against fungal infestation and aflatoxin contamination of herbal raw materials. Nat. Prod. Res. 2022, 36, 4569–4574. [Google Scholar] [CrossRef]
  110. Abdel-Razek, A.G.; Badr, A.N.; Alharthi, S.S.; Selim, K.A. Efficacy of Bottle Gourd Seeds’ Extracts in Chemical Hazard Reduction Secreted as Toxigenic Fungi Metabolites. Toxins 2021, 13, 789. [Google Scholar] [CrossRef]
  111. Beekrum, S.; Govinden, R.; Padayachee, T.; Odhav, B. Naturally occurring phenols: A detoxification strategy for fumonisin B1. Food Addit. Contam. 2003, 20, 490–493. [Google Scholar] [CrossRef]
  112. Silva, B.; Souza, M.M.; Badiale-Furlong, E. Antioxidant and antifungal activity of phenolic compounds and their relation to aflatoxin B1 occurrence in soybeans (Glycine max L.). J. Sci. Food Agric. 2020, 100, 1256–1264. [Google Scholar] [CrossRef] [PubMed]
  113. Zaman, S.; Khan, N.; Zahoor, M.; Ullah, R.; Bari, A.; Sohail. Phytochemical-mediated regulation of aflatoxigenic fungi contamination in a shifting climate and environment. Environ. Geochem. Health 2024, 46, 272. [Google Scholar] [CrossRef] [PubMed]
  114. 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. Anim. Int. J. Anim. Biosci. 2019, 13, 42–52. [Google Scholar] [CrossRef]
  115. Cao, Z.; Gao, J.; Huang, W.; Yan, J.; Shan, A.; Gao, X. Curcumin mitigates deoxynivalenol-induced intestinal epithelial barrier disruption by regulating Nrf2/p53 and NF-κB/MLCK signaling in mice. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2022, 167, 113281. [Google Scholar] [CrossRef]
  116. Ling, K.H.; Wan, M.L.; El-Nezami, H.; Wang, M. Protective Capacity of Resveratrol, a Natural Polyphenolic Compound, against Deoxynivalenol-Induced Intestinal Barrier Dysfunction and Bacterial Translocation. Chem. Res. Toxicol. 2016, 29, 823–833. [Google Scholar] [CrossRef] [PubMed]
  117. Long, H.; Xin, Z.; Zhang, F.; Zhai, Z.; Ni, X.; Chen, J.; Yang, K.; Liao, P.; Zhang, L.; Xiao, Z.; et al. The cytoprotective effects of dihydromyricetin and associated metabolic pathway changes on deoxynivalenol treated IPEC-J2 cells. Food Chem. 2021, 338, 128116. [Google Scholar] [CrossRef]
  118. Long, M.; Zhang, Y.; Li, P.; Yang, S.H.; Zhang, W.K.; Han, J.X.; Wang, Y.; He, J.B. Intervention of Grape Seed Proanthocyanidin Extract on the Subchronic Immune Injury in Mice Induced by Aflatoxin B1. Int. J. Mol. Sci. 2016, 17, 516. [Google Scholar] [CrossRef]
  119. Damiano, S.; Iovane, V.; Squillacioti, C.; Mirabella, N.; Prisco, F.; Ariano, A.; Amenta, M.; Giordano, A.; Florio, S.; Ciarcia, R. Red orange and lemon extract prevents the renal toxicity induced by ochratoxin A in rats. J. Cell. Physiol. 2020, 235, 5386–5393. [Google Scholar] [CrossRef]
  120. Akinrinmade, F.J.; Akinrinde, A.S.; Amid, A. Changes in serum cytokine levels, hepatic and intestinal morphology in aflatoxin B1-induced injury: Modulatory roles of melatonin and flavonoid-rich fractions from Chromolena odorata. Mycotoxin Res. 2016, 32, 53–60. [Google Scholar] [CrossRef]
  121. Wang, X.; Yang, F.; Na, L.; Jia, M.; Ishfaq, M.; Zhang, Y.; Liu, M.; Wu, C. Ferulic acid alleviates AFB1-induced duodenal barrier damage in rats via up-regulating tight junction proteins, down-regulating ROCK, competing CYP450 enzyme and activating GST. Ecotoxicol. Environ. Saf. 2022, 241, 113805. [Google Scholar] [CrossRef]
  122. Ye, Y.; Jiang, M.; Hong, X.; Fu, Y.; Chen, Y.; Wu, H.; Sun, Y.; Wang, X.; Zhou, E.; Wang, J.; et al. Quercetin Alleviates Deoxynivalenol-Induced Intestinal Damage by Suppressing Inflammation and Ferroptosis in Mice. J. Agric. Food Chem. 2023, 71, 10761–10772. [Google Scholar] [CrossRef]
  123. Qin, T.; Liu, X.; Luo, Y.; Yu, R.; Chen, S.; Zhang, J.; Xu, Y.; Meng, Z.; Huang, Y.; Ren, Z. Characterization of polysaccharides isolated from Hericium erinaceus and their protective effects on the DON-induced oxidative stress. Int. J. Biol. Macromol. 2020, 152, 1265–1273. [Google Scholar] [CrossRef]
  124. Tang, M.; Yuan, D.; Liao, P. Berberine improves intestinal barrier function and reduces inflammation, immunosuppression, and oxidative stress by regulating the NF-κB/MAPK signaling pathway in deoxynivalenol-challenged piglets. Environ. Pollut. 2021, 289, 117865. [Google Scholar] [CrossRef] [PubMed]
  125. de Freitas Souza, C.; Baldissera, M.D.; Descovi, S.; Zeppenfeld, C.; Eslava-Mocha, P.R.; Gloria, E.M.; Zanette, R.A.; Baldisserotto, B.; Schafer da Silva, A. Melaleuca alternifolia essential oil abrogates hepatic oxidative damage in silver catfish (Rhamdia quelen) fed with an aflatoxin-contaminated diet. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2019, 221, 10–20. [Google Scholar] [CrossRef]
  126. Vipin, A.V.; Rao, R.; Kurrey, N.K.; KA, A.A.; Venkateswaran, G. Protective effects of phenolics rich extract of ginger against Aflatoxin B(1)-induced oxidative stress and hepatotoxicity. Biomed. Pharmacother. Biomed. Pharmacother. 2017, 91, 415–424. [Google Scholar] [CrossRef]
  127. Ben Salah-Abbès, J.; Abbès, S.; Houas, Z.; Abdel-Wahhab, M.A.; Oueslati, R. Zearalenone induces immunotoxicity in mice: Possible protective effects of radish extract (Raphanus sativus). J. Pharm. Pharmacol. 2008, 60, 761–770. [Google Scholar] [CrossRef]
  128. Bilgrami, K.S.; Sinha, K.K.; Sinha, A.K. Inhibition of aflatoxin production & growth of Aspergillus flavus by eugenol & onion & garlic extracts. Indian. J. Med. Res. 1992, 96, 171–175. [Google Scholar]
  129. Costa, S.; Utan, A.; Speroni, E.; Cervellati, R.; Piva, G.; Prandini, A.; Guerra, M.C. Carnosic acid from rosemary extracts: A potential chemoprotective agent against aflatoxin B1. An in vitro study. J. Appl. Toxicol. JAT 2007, 27, 152–159. [Google Scholar] [CrossRef]
  130. Gowda, N.K.; Ledoux, D.R.; Rottinghaus, G.E.; Bermudez, A.J.; Chen, Y.C. Antioxidant efficacy of curcuminoids from turmeric (Curcuma longa L.) powder in broiler chickens fed diets containing aflatoxin B1. Br. J. Nutr. 2009, 102, 1629–1634. [Google Scholar] [CrossRef]
  131. Kim, B.R.; Kim, D.H.; Park, R.; Kwon, K.B.; Ryu, D.G.; Kim, Y.C.; Kim, N.Y.; Jeong, S.; Kang, B.K.; Kim, K.S. Effect of an extract of the root of Scutellaria baicalensis and its flavonoids on aflatoxin B1 oxidizing cytochrome P450 enzymes. Planta Medica 2001, 67, 396–399. [Google Scholar] [CrossRef]
  132. Pomothy, J.M.; Pászti-Gere, E.; Barna, R.F.; Prokoly, D.; Jerzsele, Á. The Impact of Fermented Wheat Germ Extract on Porcine Epithelial Cell Line Exposed to Deoxynivalenol and T-2 Mycotoxins. Oxidative Med. Cell. Longev. 2020, 2020, 3854247. [Google Scholar] [CrossRef] [PubMed]
  133. Qin, G.Z. Effects of green tea extract on the development of aflatoxin B1-induced precancerous enzyme-altered hepatocellular foci in rats. Zhonghua Yu Fang Yi Xue Za Zhi Chin. J. Prev. Med. 1991, 25, 332–334. [Google Scholar]
  134. Sánchez, E.; Heredia, N.; García, S. Inhibition of growth and mycotoxin production of Aspergillus flavus and Aspergillus parasiticus by extracts of Agave species. Int. J. Food Microbiol. 2005, 98, 271–279. [Google Scholar] [CrossRef] [PubMed]
  135. Santos, L.; Kasper, R.; Sardiñas, N.; Marín, S.; Sanchis, V.; Ramos, A.J. Effect of Capsicum carotenoids on growth and aflatoxins production by Aspergillus flavus isolated from paprika and chilli. Food Microbiol. 2010, 27, 1064–1070. [Google Scholar] [CrossRef]
  136. Tulayakul, P.; Dong, K.S.; Li, J.Y.; Manabe, N.; Kumagai, S. The effect of feeding piglets with the diet containing green tea extracts or coumarin on in vitro metabolism of aflatoxin B1 by their tissues. Toxicon 2007, 50, 339–348. [Google Scholar] [CrossRef]
  137. Vaičiulienė, G.; Bakutis, B.; Jovaišienė, J.; Falkauskas, R.; Gerulis, G.; Bartkienė, E.; Klupšaitė, D.; Klementavičiūtė, J.; Baliukonienė, V. Effects of Ethanol Extracts of Origanum vulgare and Thymus vulgaris on the Mycotoxin Concentrations and the Hygienic Quality of Maize (Zea mays L.). Silage. Toxins 2022, 14, 298. [Google Scholar] [CrossRef]
  138. Jin, L.; Yang, J. Research Progress on the Antioxidant Properties of Plant Extracts and Their Application in Antibiotic-Free Animal Feed. China Anim. Husb. Mag. 2020, 56, 29–34. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, X.; Chen, J.; Ma, X.; Tang, X.; Tan, B.; Liao, P.; Yao, K.; Jiang, Q. Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies. Metabolites 2025, 15, 219. https://doi.org/10.3390/metabo15040219

AMA Style

Zhang X, Chen J, Ma X, Tang X, Tan B, Liao P, Yao K, Jiang Q. Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies. Metabolites. 2025; 15(4):219. https://doi.org/10.3390/metabo15040219

Chicago/Turabian Style

Zhang, Xiangnan, Jiashun Chen, Xiaokang Ma, Xiongzhuo Tang, Bie Tan, Peng Liao, Kang Yao, and Qian Jiang. 2025. "Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies" Metabolites 15, no. 4: 219. https://doi.org/10.3390/metabo15040219

APA Style

Zhang, X., Chen, J., Ma, X., Tang, X., Tan, B., Liao, P., Yao, K., & Jiang, Q. (2025). Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies. Metabolites, 15(4), 219. https://doi.org/10.3390/metabo15040219

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