You are currently viewing a new version of our website. To view the old version click .
Toxins
  • Review
  • Open Access

11 November 2025

Deoxynivalenol and Its Metabolites: Contamination, Metabolism, and Toxicity

,
,
,
,
,
,
,
,
and
1
College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, China
2
Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471023, China
3
Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471003, China
4
The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, China

Abstract

Deoxynivalenol (DON), a toxic secondary metabolite produced by Fusarium graminearum in infected cereal crops, has emerged as a major global contaminant in food and feed due to its stable physicochemical properties and resistance to degradation during conventional processing. This contamination poses a serious threat to livestock production and animal health. This review provides a comprehensive overview of the current status of DON contamination, its transmission through the food chain, metabolic pathways in animals, and the comparative toxicity of its metabolites. Furthermore, we analyze DON-induced toxic effects, including acute toxicity, cytotoxicity, immunotoxicity, neurotoxicity, gastrointestinal toxicity, and hepatotoxicity. By integrating domestic and international regulatory thresholds with current mitigation strategies, we highlight future research directions focusing on biodegradation technologies and genetic regulation approaches to alleviate DON contamination in livestock feeds. Advancing efficient DON-degradation strategies could open new avenues for sustainable feed management and mycotoxin detoxification technologies.
Key Contribution:
This review comprehensively analyzed the global contamination status, metabolic pathways, and mechanisms of multi-organ toxicity of deoxynivalenol, while also evaluating related mitigation strategies. Furthermore, it elucidated future research directions for its effective management in the fields of animal feed and food safety.

1. Introduction

Mycotoxins are toxic secondary metabolites produced by filamentous fungi that frequently contaminate grains, feed, and food commodities, thereby posing a significant threat to global agricultural security and human health []. Among them, deoxynivalenol (DON), a representative type B trichothecene mycotoxin, has garnered substantial attention in food safety and livestock production research due to its high prevalence and potent biological toxicity. The primary DON-producing species, Fusarium graminearum and Fusarium culmorum, synthesize and accumulate this toxin in wheat, maize, barley, and other crops under favorable temperature and humidity conditions []. Owing to its stable physicochemical properties (Table 1), DON resists degradation through conventional food-processing methods, facilitating its persistence and transmission across the feed supply chain to livestock and humans [,].
Table 1. Physicochemical properties of deoxynivalenol.
The deleterious effects of DON in animals are mediated through multifaceted mechanisms, including acute toxicity, growth retardation, disruption of the intestinal barrier, immunosuppression, and hepatotoxicity []. Studies indicate that swine exhibit markedly higher sensitivity to DON compared to poultry and ruminants. Chronic exposure to DON can lead to a reduction in feed conversion efficiency by 15–30%, resulting in annual economic losses exceeding USD 1 billion in the global livestock industry due to DON contamination [,]. Despite the establishment of regulatory limits for DON in animal feed across numerous countries, the global contamination rate of cereal remains alarmingly high.
Recent research has underscored concerns regarding the potential for in vivo reactivation of DON metabolites. While certain metabolites demonstrate reduced toxicity, DON-3-glucoside (D3G) is susceptible to enzymatic cleavage by gut microbiota, resulting in the release of the parent toxin and an increase in systemic toxicity. Additionally, the immunomodulatory effects of DON—ranging from immunosuppression to hyperactivation—and its intricate interactions with the gut microbiota are not yet fully elucidated, impeding the development of targeted mitigation strategies. This review provides a examination of the physicochemical properties and prevalence of DON and its metabolites, their metabolic pathways in animals, and the mechanistic associations between DON exposure and adverse effects on animal health.

2. Contamination in Food and Feed

2.1. Regulatory Maximum Limits for DON in Food and Feed

DON is among the most prevalent mycotoxins globally, with widespread contamination in cereals and animal feed, posing a significant threat to global food security and sustainable livestock production []. Regions with intensive grain production, such as China, Japan, Canada, and the Americas, are at a heightened risk of DON contamination. Grains compromised by DON not only suffer significant reductions in nutritional quality but also pose serious health risks to humans and animals, either through direct consumption or through bioaccumulation within the food chain []. To address the hazards associated with DON, international regulatory bodies have established stringent regulatory maximum limits for DON in both feed and food (shown in Table 2). Nevertheless, there remain inconsistencies in these standards across different countries and regions, which are influenced by variations in agricultural practices, dietary habits, and risk assessment methodologies. For example, China enforces a DON limit of 1000 µg/kg in feed for susceptible species such as pigs and calves, while the European Union imposes a more stringent limit of 200 µg/kg for cereal products intended for infants. It is noteworthy that higher tolerance limits for DON are typically applied to ruminant feeds, due to the partial detoxification capacity of the rumen microbiota through microbial biotransformation.
Table 2. Regulatory maximum limits for DON in food and feed in selected different countries and regions.

2.2. DON Contamination

DON is predominantly detected in animal feeds and cereals, including oats, maize, wheat, and barley, and is prevalent in regions such as East Asia, Northern and Central Europe, Central America, North America, and South Africa []. The highest levels of DON have been reported in wheat samples from specific areas in China []. In a Shanghai-based study, DON was detected in 85.8% of wheat flour samples []. In a survey conducted in Hebei Province from 2011 to 2013, DON was detected in 91.5% of wheat samples, with a median concentration of 154 µg/kg and a maximum level reaching 1130 µg/kg []. Similarly, in Shandong Province, the contamination rate of DON in wheat flour was as high as 97.2% []. Beyond China, significant DON contamination has also been reported in other regions. For instance, in Europe, contamination rates approached 100% in German silage maize and Polish complete feed [,]. In the United States, DON was detected in 94% of swine feed samples and in 88.2% of corn silage samples, with certain cases exceeding 5000 µg/kg [,]. Additionally, in Brazil, DON was detected in 77.9% of wheat flour samples, with the highest concentration reported at 2794.63 µg/kg []. The occurrence of DON contamination exhibits notable regional and ingredient specificity (Table 3). These high contamination rates may be attributed for Fusarium growth in temperate climates. Contamination rates were generally higher in silage and piglet feed compared to processed grains microbial biotransformation in the rumen, indicating that feed ingredients are particularly vulnerable to secondary contamination during storage and fermentation. The global economic impact of DON contamination on livestock is substantial, with Asia and Europe bearing a relatively high burden.
Table 3. Main Raw Material Sources and Contamination Levels of DON.

2.3. Transmission in the Food Chain

DON is predominantly synthesized by Fusarium spp., with its biosynthesis being markedly influenced by environmental conditions. Research indicates that Fusarium spp. can readily infect cereal crops such as wheat, maize, and barley under optimal temperature and humidity conditions, continuing to produce DON throughout their growth phase [,]. The distribution of DON within plant tissues is notably heterogeneous. In wheat, DON is primarily concentrated in the bran layer, leading to significantly higher concentrations in wholemeal flour compared to refined flour []. The data show that whole grain products generally contain higher DON concentrations than refined flour products, which is consistent with the preferential accumulation of DON in the bran layer. In maize, DON levels are frequently higher in the stem than in the ear, and variations in environmental conditions across different years can result in distinct differences in DON accumulation by altering the pattern of Fusarium graminearum infestation. This heterogeneous distribution has direct implications for the safety risk assessment of silage and processing by-products [].
Furthermore, DON enters the livestock production chain through the consumption of contaminated crops used as feed ingredients. DON is partially absorbed by the digestive tract of animals, after which its parent compound and metabolites are distributed to tissues such as the liver and kidneys via the bloodstream and subsequently excreted through urine and feces. Nonetheless, trace amounts of DON and its metabolites may persist in muscle tissue, liver, and dairy products, ultimately entering the human food chain through consumption of animal-derived products such as meat, eggs, and milk [,]. The transfer of DON within the food chain is further complicated by the effects of food processing. Due to its high chemical stability, DON remains present throughout grain harvest, storage, and transport. It can persist into subsequent food-processing stages. Conventional processing techniques result in only minimal reductions in DON levels, and concentrations in processing by-products may be elevated due to water loss. Epidemiological studies indicate that exposure to DON can result in acute toxicity, while chronic low-dose exposure is associated with immunosuppression, intestinal barrier damage, and neurological dysfunction, thereby posing a significant risk to both human and animal health [,].

3. Biotransformation and Metabolites of DON

3.1. Secondary Metabolites of DON

DON undergoes metabolism in organisms through various pathways, resulting in the formation of secondary metabolites with diverse structures. The primary secondary metabolites of DON currently identified include 3-Acetyl-DON, 15-Acetyl-DON, DON-3-glucoside (D3G), de-epoxylated-DON (DOM-1), and 3-epi-DON, among others (Figure 1) [].
Figure 1. Structures and enzymatic formation pathways of DON and its metabolites.
Both 3-acetyl-DON and 15-acetyl-DON, acetylated derivatives of DON commonly found in contaminated food and feed, can be deacetylated by intestinal microorganisms to revert to DON, thereby exhibiting acute and chronic toxicities similar to the parent compound []. D3G, a detoxification product resulting from the glycosylation of DON in plants, possesses low inherent toxicity. However, it may be hydrolyzed by intestinal flora to release free DON, potentially increasing the risk of toxicity []. DOM-1 is generated through microbial or enzyme-catalyzed de-epoxidation of DON and displays markedly reduced toxicity due to the loss of the epoxy group, which lowers its affinity for the ribosome and consequently its inhibition of protein synthesis []. Furthermore, 3-epi-DON is produced through isomerization, resulting in an altered stereochemical conformation that diminishes its interactions with biomolecules, thereby exhibiting reduced cytotoxicity [].
The formation mechanisms of DON secondary metabolites are intricate, involving multiple pathways such as microbial transformation, plant detoxification, and animal metabolism. Microorganisms modify the structure of DON through enzymatic processes including acetylation, deacetylation, de-epoxidation, and isomerization, leading to the production of corresponding metabolites [,,]. In plants, DON is conjugated with glucose by glycosyltransferases to form D3G, thereby reducing its toxicity. In animals, liver metabolizing enzymes facilitate de-epoxidation, glucuronidation, and sulfonation processes, generating various metabolites and enhancing the excretion of DON.
Compared to DON, DOM-1 and 3-epi-DON exhibit lower toxicity and have reduced inhibitory, induction of oxidative stress, and apoptosis-promoting effects on cell proliferation [,]. From a molecular perspective, DON robustly activates the MAPK signaling pathway, leading to inflammatory responses and apoptosis. In contrast, this pathway’s activation is markedly reduced by the presence of DOM-1, indicating that the toxic effects of DON are intricately linked to structural modifications []. These observations underscore the necessity of integrating toxicity assessments of secondary metabolites with their chemical characterization and metabolic dynamics to comprehensively elucidate their potential impacts on organisms.

3.2. Metabolism of DON in the Organism

3.2.1. Humans

Human exposure to DON occurs primarily through the consumption of contaminated cereals and cereal-based products []. Following ingestion, DON is efficiently absorbed in the gastrointestinal tract []. The major metabolic pathway involves glucuronidation, catalyzed by uridine diphosphate-glucuronosyltransferases (UGTs) in the liver and other tissues, leading to the formation of DON-glucuronide conjugates, such as DON-15-glucuronide and DON-3-glucuronide []. Furthermore, the human intestinal microbiota hydrolyzes the plant conjugate DON-3-glucoside (DON-3G) to release free DON, and in certain individuals, further transforms it into the de-epoxy metabolite DOM-1, which exhibits lower toxicity []. DON and its metabolites are rapidly excreted, predominantly via the kidneys, with approximately 64% of ingested DON and 58% of DON-3G recovered in urine within 24 h post-administration []. Consequently, urinary DON and its glucuronide conjugates, particularly DON-15-glucuronide, serve as robust biomarkers for assessing human exposure. Numerous biomonitoring studies conducted globally in regions such as China, Belgium, Italy, and the United Kingdom have employed these biomarkers for exposure assessment, revealing that certain population subgroups, especially children, may experience exposure levels exceeding the established tolerable daily intake (TDI) [,,,,,].

3.2.2. Monogastric Animals

Swine are highly sensitive to DON. The porcine gastrointestinal tract rapidly and almost completely absorbs this mycotoxin, resulting in high systemic bioavailability. Following oral intake, DON is primarily absorbed in the proximal small intestine. Plasma concentrations peak approximately 4.1 h post-feeding, with an elimination half-life (t1/2) of about 5.8 h []. Inside the body, DON is predominantly metabolized in the liver and kidneys. This process, catalyzed by UGTs, leads to the formation of DON-glucuronide conjugates (DON-GlcA). DON-GlcA represents the major form in plasma and serves as a suitable biomarker for assessing DON exposure []. Furthermore, DON and its metabolite de-epoxy-DON can be detected in serum, liver, bile, and urine. Their concentrations demonstrate a significant linear increase corresponding to rising dietary DON levels []. Renal excretion is the primary elimination route for DON in swine. Up to 43.2% of the ingested dose can be excreted as the parent toxin in urine, while fecal excretion is considerably lower (approximately 3.0%) []. The strong correlation between DON intake and its concentrations in both serum and urine confirms these matrices as reliable indicators for monitoring DON exposure in swine [].
Compared with swine, poultry exhibit lower sensitivity to DON, which is attributed to their distinct metabolic pathways. The intestinal absorption of DON in poultry is limited, with an oral bioavailability of only 19.3%. In broilers, DON is rarely detected in its parent form in plasma; instead, it is rapidly and extensively converted to its major metabolite, DON-3-sulfate (DON-3S). Studies have shown that after 42 days of being fed diets containing 5 mg/kg or 15 mg/kg DON, DON-3S was detectable in the plasma and excreta of broilers. However, hepatic deposition was observed only in the high-dose group (15 mg/kg). Therefore, DON-3S can serve as a specific biomarker for DON exposure in various biological matrices (plasma, excreta, liver) in poultry []. Apart from DON-3S, excreta contained only minimal amounts of the parent DON, and no de-epoxidation metabolite (DOM-1) was detected. Poultry excrete DON rapidly, primarily via feces, with a reported biorecovery (based on DON-3S and DON) of 74–106% in broilers. The conversion of DON to DON-3S is considered a detoxification mechanism, as in vitro studies indicate that the toxicity of DON-3S to ribosomes is nearly 2000 times lower than that of DON []. The relative tolerance of poultry to DON is likely due to their efficient conversion of DON into the less toxic DON-3S and its rapid elimination. Chronic DON exposure in broilers may induce intestinal adaptive changes, such as increased jejunal length and reduced villus height; however, nutrient retention remains unaffected, suggesting the presence of physiological adaptation mechanisms [].

3.2.3. Ruminants

Ruminants exhibit relatively low sensitivity to DON, primarily due to the potent degradation capacity of rumen microbiota. Under normal rumen function, rumen microbes efficiently convert ingested DON into the nearly non-toxic de-epoxy metabolite DOM-1, with conversion rates reaching 81–99%. Consequently, DOM-1 is the primary compound detected in the blood and milk of ruminants, while the systemic bioavailability of DON itself is very low (e.g., only 5.9–9.9% in sheep) []. However, this detoxification capability is highly dependent on a healthy rumen environment. In vitro studies indicate that when rumen pH drops to 5.8 (simulating subacute ruminal acidosis, SARA), DON degradation is significantly inhibited. This is particularly evident in rumen fluid from non-lactating cattle with lower microbial activity, where the degradation rate can decrease to approximately 40% []. High-starch diets predispose the rumen to pH depression, which not only reduces DOM-1 production but also increases the absorption of unmetabolized DON. These changes are associated with decreased total ruminal volatile fatty acid concentrations and elevated serum inflammatory markers (e.g., TNF-α) in dairy cows []. In summary, maintaining healthy rumen function is crucial for protecting ruminants against the adverse effects of DON. Under conditions of SARA or impaired rumen microbial activity, DON may escape degradation, leading to increased post-ruminal exposure and potential health risks.

4. Effects of DON on the Health of the Organism

4.1. Toxic Effects of DON

The health impacts of DON on animals are multifaceted. By binding to ribosomes, it can trigger cellular stress and apoptosis, and disrupt intestinal tight junctions. This leads to a series of toxic effects including cytotoxicity, immunosuppression, intestinal barrier damage, and hepatotoxicity, ultimately resulting in growth retardation and economic losses in livestock and poultry (Figure 2) [].
Figure 2. Toxic mechanisms, impacts, and integrated control strategies of DON.

4.1.1. Acute Toxicity

The acute toxicity of DON is primarily manifested by rapid physiological responses following high-dose exposure in both humans and animals, inducing symptoms such as vomiting, diarrhea, dizziness, and disorders of the central nervous system, with severity influenced by species, sex, age, and exposure route [,]. In humans, acute exposure through contaminated food, particularly during cereal-related outbreak events, has been clinically associated with gastroenteritis characterized by vomiting, abdominal pain, and diarrhea []; findings from Chinese epidemiological studies further substantiate the emetic potential of DON in human populations []. This emetic property, which earns DON the common name “vomitoxin,” has been rigorously quantified in sensitive animal models such as the pig. It has been demonstrated in mouse studies that exposure to 1 and 2.5 mg/kg·bw of DON can induce acute anorexia and concurrently elevate plasma levels of the intestinal hormones CCK, PYY, GIP, and GLP-1 within 3 h []. Among livestock, pigs exhibit the highest sensitivity, achieving peak plasma levels within 30 min post-ingestion of contaminated feed, with vomiting occurring approximately 10 min thereafter [,]. Studies have reported the minimum emetic dose (MED) in pigs weighing 9–10 kg to be 0.05 mg/kg body weight (BW) via intraperitoneal injection and 0.1–0.2 mg/kg BW via oral gavage []. In contrast, ruminants demonstrate greater tolerance due to the de-epoxygenation process facilitated by rumen microorganisms, with poultry displaying intermediate sensitivity. The acute toxicity of DON has direct implications for livestock production, manifesting as reduced feed intake, growth inhibition, and organ damage, all of which collectively result in economic losses [,].

4.1.2. Cytotoxicity

DON exerts its effects by binding to the 60S subunit of eukaryotic ribosomes, thereby inhibiting peptidyltransferase activity, disrupting protein synthesis, and activating the MAPK signaling pathway. This activation induces a “ribosomal stress response”, culminating in apoptosis []. In livestock, intestinal epithelial cells and immune cells are particularly susceptible to DON’s effects. Specifically, the viability of porcine small intestinal epithelial cells (IPEC-J2) is significantly compromised upon exposure to 1 μM DON, with cell membrane rupture and vacuolization observed at concentrations exceeding 8 μM []. Similarly, in human cells such as keratinocytes, DON exposure significantly decreases cell viability and induces oxidative stress, mitochondrial damage, and cell death through apoptosis and pyroptosis []. DON exhibits dose-dependent cytotoxicity, disrupting cell membrane integrity, inflicting DNA damage, altering cellular morphology, impairing protein synthesis, and ultimately inducing apoptosis. Additionally, DON contributes to the formation of mitotic-positive micronuclei, which can further exacerbate organ damage [].

4.1.3. Immunotoxicity

DON functions as a bi-directional modulator of the immune system, inducing nucleotoxic stress and inhibiting the synthesis of RNA, DNA, and proteins []. The immunotoxic effects of DON are modulated by various factors, including exposure dose, duration, and host physiology. These effects are characterized by an imbalance between humoral and cellular immunity, abnormal serum IgA levels, increased lymphocyte apoptosis, and disruptions in the expression of pro-inflammatory factors [,]. At the molecular level, DON activates the endoplasmic reticulum stress-related signaling pathway (ATF3/DDIT3) and mediates mitochondria-dependent apoptosis through the activation of the MAPK phosphorylation cascade as well as hematopoietic cell kinase (Hck), ultimately inhibiting the proliferation and differentiation of immune cells []. At low concentrations (<1 μM), DON enhances host defense against pathogens, as evidenced by the up-regulation of T-cell activating factors, enhancement of helper T-cell function, and increased intestinal IgA secretion []. When the concentration of DON exceeds 1 μM, its immunosuppressive effects became predominant, resulting in the dysregulation of interleukin-mediated immunoglobulin secretion. Additionally, DON inhibited the expression of inducible nitric oxide synthase (iNOS) in intestinal epithelial cells through the proteasomal pathway, leading to intestinal inflammation and an elevated risk of infection []. Furthermore, acetylated derivatives and bound-state metabolites of DON exhibit similar immunomodulatory activities. Notably, deoxynivalenol-3-glucoside (DOM) can specifically activate humoral immune responses due to its preserved ability to bind to lymphocyte surface receptors []. The immunotoxicity of 3-acetyldeoxynivalenol (3ADON) and 15-acetyldeoxynivalenol (15ADON) is significantly lower than that of DON. In contrast, deoxynivalenol-3-glucoside (D3G) exhibits negligible immunotoxicity, as it does not induce cytokine release nor trigger a nucleotoxic stress response [].

4.1.4. Neurotoxicity

The neurotoxic mechanism of DON is intricate, involving interactions across multiple pathways and resulting in multi-target damage. Research has demonstrated that DON can traverse the blood–brain barrier and exert direct effects on the central nervous system, altering its permeability and inducing dysfunction in both neuronal and glial cells. The toxicological effects of DON are primarily manifested through mitochondria-mediated apoptosis, oxidative stress, calcium signaling dysregulation, neuroinflammation, glial cell injury, and disruption of neurotransmitter systems. DON induces oxidative stress and neuronal apoptosis by activating the mitochondrial apoptotic pathway, characterized by an imbalance in Bax/Bcl-2, the release of cytochrome C, and the caspase-9/caspase-3 cascade, while also modulating the MAPK signaling pathway. Additionally, the activation of caspase-8 further amplifies the apoptotic effects via the mitochondrial pathway [,]. Regarding oxidative stress, exposure to DON markedly increased reactive oxygen species (ROS) levels, resulting in lipid peroxidation, protein carbonylation, and DNA damage. This exposure also precipitated a collapse in mitochondrial membrane potential and disrupted neurotransmitter metabolism by interfering with Ca2+ homeostasis, particularly affecting the Ca2+/CaM/CaMKII signaling pathway []. DON prompts microglia to secrete pro-inflammatory cytokines, such as TNF-α, and activates inflammatory responses in astrocytes, thereby intensifying central nervous system (CNS) inflammation []. Additionally, Drp-1 (Dynamin-related protein 1)-mediated mitochondrial hyperfission and the activation of autophagy further contribute to neuronal degenerative pathology []. Furthermore, DON disrupts the neurotransmitter system by significantly reducing levels of dopamine and gamma-aminobutyric acid while increasing concentrations of serotonin and norepinephrine. This neurotransmitter imbalance is closely linked to appetite suppression, gastrointestinal dysfunction, and behavioral abnormalities [].

4.1.5. Enterotoxicity

The intestinal tract serves as the primary target organ for DON in both humans and animals after consumption of contaminated food or feed, leading to morphological damage, impaired nutrient absorption, and compromised barrier function. Chronic exposure thus contributes to significant intestinal pathology. For example, in young rats given 10 mg/g DON for 28 days, jejunal villi exhibited shortening, fusion, and increased epithelial apoptosis []. Similarly, broilers and pigs fed 10 mg/kg DON for 8 weeks showed dose-dependent reductions in villus height, increased crypt depth, and decreased villus-to-crypt ratio, alongside marked downregulation of tight junction proteins, indicating impaired barrier integrity []. Notably, chronic exposure to DON at a human-relevant dose (10 μg/kg bw/day) in mice disrupts multiple aspects of intestinal homeostasis, encompassing microbiota composition, immune response, and epithelial integrity, which bridges the toxic effects observed in livestock to potential human health risks []. DON also markedly alters the gut microbiota. In weaned piglets, DON reduced Firmicutes abundance and increased Actinobacteria, indicating dysbiosis []. Weaned rabbits exposed to high DON levels showed decreased microbial diversity, further destabilizing the gut microenvironment []. Moreover, DON impairs epithelial repair by inhibiting proliferation and inducing apoptosis. Piglets chronically exposed to low-dose DON had fewer jejunal and ileal epithelial cells and a higher proportion of apoptotic cells, suggesting disrupted cellular homeostasis aggravates intestinal dysfunction [,]. In summary, evidence from diverse animal models, including those relevant to humans, demonstrates that DON-induced intestinal toxicity involves structural damage, barrier defects, microbial imbalance, and loss of cellular homeostasis, collectively endangering host health.

4.1.6. Hepatotoxicity

As a central organ involved in metabolism, detoxification, and immunoregulation, the structural and functional integrity of the liver is vital for maintaining overall physiological homeostasis. Research has demonstrated that exposure to DON can significantly impact liver morphology and function, as evidenced by alterations in the ultrastructure of hepatocytes. In porcine hepatocytes treated with DON, numerous short, dilated vesicles were observed in both the rough and smooth endoplasmic reticulum, suggesting potential interference with intracellular protein synthesis and lipid metabolism []. In animal models, histopathological analyses of the livers of broilers and pigs fed with high doses of DON revealed inflammatory cell infiltration in liver tissue, accompanied by increased liver injury []. The deleterious effects of DON on the developing liver are notably pronounced, with pathological alterations such as the accumulation of lipid vesicles in hepatic tissue and a marked suppression of splenic immune function observed in the chick embryo exposure model []. At the molecular level, both in vitro and in vivo studies have elucidated that DON exacerbates hepatic injury by inducing inflammatory responses, activating oxidative stress pathways, and promoting apoptosis in hepatocytes. For instance, short-term exposure to DON significantly diminishes the viability of HepG2 cells and exerts direct toxic effects on primary hepatocytes and liver tissue []. Furthermore, animal studies have demonstrated that DON exposure disrupts hepatic redox homeostasis, as indicated by increased lipid peroxidation products and decreased activity of antioxidant enzymes, alongside the aberrant expression of pro-inflammatory factors [,]. These findings collectively elucidate the multi-faceted mechanisms of DON-induced hepatic toxicity, providing a critical foundation for assessing its health risks and developing targeted prevention and mitigation strategies.

4.2. Effects on Biochemical Indicators

DON significantly influences serum biochemical indices across a broad spectrum of animal species, exhibiting effects that are both consistent and species-specific. Serum biochemical markers are crucial for evaluating the homeostasis of the internal environment and organ functionality in animals, with aberrant changes often indicative of metabolic disturbances or tissue damage.
In pigs, DON exerts significant dose-dependent effects on biochemical indices. Research has demonstrated that prolonged consumption of low-dose DON (12 μg/kg body weight) by prepubertal sows led to reductions in blood alanine aminotransferase levels, erythrocyte counts, platelet, and leukocyte counts, thereby disrupting blood homeostasis []. Conversely, when healthy growing pigs were fed a high dose of DON (12 mg/kg), there was an increase in blood urea nitrogen, alkaline phosphatase, and alanine aminotransferase levels, alongside a decrease in serum L-valine, glycine, and other amino acid concentrations. This was accompanied by a reduction in antioxidant enzyme activities, suggesting that DON can induce metabolic disorders and oxidative stress []. Recent experimental studies on piglets have corroborated that diets contaminated with DON markedly decrease average daily weight gain and feed intake. These diets also suppress the activities of antioxidant enzymes, including serum immunoglobulin G, catalase, and superoxide dismutase, and downregulate the expression of anti-inflammatory cytokines such as IL-4 and IL-10. Conversely, there is an upregulation of pro-inflammatory cytokines, including TNF-α, interleukin-6, and interleukin-12 [].
Parallel investigations in the poultry sector have demonstrated the dose-dependent toxicity of DON. In chicks administered a diet with 10 mg/kg DON until 35 days of age, there was a significant reduction in serum alanine aminotransferase (ALT) activity, alongside significant increases in cholesterol and triglyceride levels. These findings indicate disruptions in lipid metabolism and a suppression of the humoral immune response to viral vaccines []. Lower doses (5 mg/kg) of DON did not affect growth performance but reduced serum creatine kinase levels and altered small intestine morphology. In contrast, higher doses (15 mg/kg) not only reduced body weight gain and feed efficiency but also decreased serum cholesterol levels. Additionally, higher doses led to increased absolute and relative weights of the thymus and gasterocysts, along with a reduction in colon weights []. These findings indicate that DON exerts a dose-dependent effect on the biochemical indices of chickens, involving the comprehensive regulation of lipid metabolism, organ development, and immune function, with more pronounced toxic effects at higher doses.
In fish, the impact of DON on biochemical parameters varies considerably among species and exposure conditions. For instance, the intake of DON (1.15–6 mg/kg feed) by Atlantic salmon (Salmo salar) resulted in dose-dependent decreases in plasma total protein, albumin, cholesterol, and triglyceride levels, as well as a reduction in alkaline phosphatase activity []; in red tilapia (Oreochromis niloticus × O. mossambicus), total plasma protein and lipid indices did not show significant changes after ingestion of DON-containing diets (0.07–1.15 mg/kg), but focal necrosis and cytoplasmic vacuolization were observed in liver tissues []; rainbow trout (Oncorhynchus mykiss) showed significant reductions in plasma glucose, cholesterol, and ammonia levels, as well as a decrease in the mean hemoglobin content of erythrocytes (MCH), when exposed to DON (2 mg/kg feed) []. These differences are closely related to factors such as fish species sensitivity, toxin exposure concentration and experimental design.
Overall, DON exerts widespread effects on the serum biochemical indices of various species, including pigs, poultry, and fish, by disrupting metabolic enzyme activities, inducing oxidative stress, compromising intestinal barriers, and suppressing immune functions. The toxic effects of DON are closely associated with both the exposure dose and duration.

4.3. Reproductive Effects

DON poses a serious threat to the reproductive health of various mammals. In males, DON exposure can induce testicular toxicity, manifested as exfoliation of sperm cells, decreased sperm quality and concentration, and increased malformation rates []. The mechanism is primarily associated with DON-induced oxidative stress, which activates the assembly of the NLRP3-ASC-Caspase 1 inflammasome, leading to pyroptosis of Leydig cells. This results in testosterone deficiency and elevated serum levels of FSH and LH []. Moreover, DON disrupts the integrity of the blood-testis barrier (BTB) by downregulating the expression of junctional proteins such as Occludin, Connexin 43, and N-cadherin. These alterations, together with inflammatory responses and hormonal imbalance, ultimately interfere with spermatogenesis []. In females, DON exerts detrimental effects on ovarian morphology, causing degeneration of oocytes and granulosa cells, interstitial edema, and a reduction in the number of normal follicles at various developmental stages, thereby impairing folliculogenesis []. Such ovarian damage is accompanied by alterations in serum FSH levels and inflammatory responses, with susceptibility exhibiting age-specific differences between prepubertal and adult females []. During pregnancy, maternal DON exposure also adversely affects placental structure and immune function, characterized by downregulation of junctional proteins, elevated levels of inflammatory cytokines (IFN-γ, IL-6, IL-4), and reduced fetal survival rates []. In summary, current evidence indicates that DON-induced reproductive toxicity is mainly mediated by interconnected mechanisms including oxidative stress, inflammatory responses, apoptosis, and pyroptosis, highlighting the necessity for further risk assessment and the development of targeted intervention strategies.

5. Strategies for DON Contamination Control: Biodegradation and Genetic Regulation

Deoxynivalenol, a prevalent mycotoxin in grains and feed, seriously threatens livestock and human health. Conventional detoxification methods face challenges including limited efficacy, nutrient loss, and hazardous by-products. Biodegradation and genetic regulation have thus gained attention as promising alternatives due to their efficiency, specificity, and environmental safety. This section reviews microbial and enzymatic biodegradation approaches alongside genetic strategies such as gene editing and transgenic technology, offering a theoretical basis for integrated DON control.

5.1. Biodegradation Technology

Biodegradation technology primarily utilizes microorganisms and their specific enzymes to convert deoxynivalenol (DON) into less toxic or non-toxic metabolites, representing an effective strategy for in situ detoxification of DON in feed and food products. Numerous studies have demonstrated the capacity of various bacterial strains to efficiently degrade DON. For instance, Devosia insulae A16 has been shown to oxidize DON to 3-keto-DON under aerobic conditions, achieving a degradation rate of 88% within 48 h []. Strains such as Bacillus sp. HN117 and N22 are capable of degrading DON even at high concentrations (up to 500 mg/L), yielding less toxic metabolites including M-DOM, norDON E, and 9-hydroxymethyl DON lactone []. Furthermore, Lactobacillus rhamnosus MY-1 exhibited a high DON degradation rate of 93.34%, and in mouse models, it significantly improved growth performance and intestinal health []. Fungal species such as Aspergillus tubingensis NJA-1 achieved a 94.4% biotransformation rate of DON via hydrolysis []. In the case of yeast, Saccharomyces cerevisiae boulardii CNCM I-1079 was reported to counteract DON-induced activation of NF-κB and p38 MAPK inflammatory pathways, while also restoring lipid metabolism and antioxidant pathways []. At the enzymatic level, Devosia mutans 17-2-E-8 employs a two-enzyme system comprising DepA and DepB to catalyze the epimerization of DON to 3-epi-DON. In this process, DepB functions as an NADPH-dependent dehydrogenase, reducing 3-keto-DON to the final product, 3-epi-DON []. Additionally, the soil bacterium Sphingomonas sp. S3-4, along with the recombinant aldo-keto reductase AKR18A1, catalyzes the oxidation of DON to 3-oxo-DON, which is subsequently converted into the non-phytotoxic 3-epi-DON, facilitating DON degradation []. Collectively, these findings underscore the considerable potential of microbial and enzymatic approaches for the biodegradation of DON, highlighting their promising applications in enhancing food and feed safety.

5.2. Gene Regulation Technology

Beyond biodegradation methods, genetic regulation strategies have emerged as pivotal approaches for mitigating DON contamination, acting through direct targeting of mycotoxin biosynthetic pathways or enhancement of host resistance mechanisms. In Fusarium graminearum, key transcription factors Tri6 and Tri10 govern DON biosynthesis by regulating the expression of TRI genes, including TRI5. Concurrently, a long non-coding RNA (RNA5P) exerts cis-repressive effects on TRI5 expression and DON production []. Furthermore, the zinc-finger transcription factor FgSfp1 coordinates DON synthesis by modulating ribosome biogenesis and post-transcriptional RNA modification; its disruption results in a 95.4% reduction in DON production []. CRISPR-based functional genomics screening has identified the host gene MDH2 as a central mediator of the cytotoxicity of multiple Fusarium toxins, including DON. Knockout of MDH2 enhances cellular toxin tolerance []. In wheat, specific SNPs in the UDP-glucosyltransferase genes TaUGT-2B and TaUGT-3B have been shown to confer resistance to Fusarium head blight and reduce DON accumulation by modulating their expression during early pathogen infection []. Collectively, these genetic and molecular insights not only deepen our understanding of DON toxicity and biosynthesis but also provide critical targets and practical strategies for developing novel control measures through gene editing, marker-assisted breeding, and transgenic technologies.

6. Conclusions and Outlook

In light of the global prevalence and persistent threat of deoxynivalenol (DON) contamination in grains and feed, this review synthesizes current understanding of its contamination status, species-specific metabolic pathways, and multifaceted toxicity mechanisms—including immunotoxicity, neurotoxicity, enterotoxicity, hepatotoxicity, and reproductive impairment—while also evaluating existing mitigation strategies such as microbial detoxification, enzymatic degradation, and genetic regulation. Looking forward, future efforts should prioritize elucidating threshold effects of DON during critical physiological stages, refining real-time detection and differentiated regulatory standards, and advancing integrated solutions combining biodegradation agents, resistant crop varieties, and precision feeding management. By bridging mechanistic insights with practical interventions, we can ultimately mitigate DON’s impact on animal health and foster the sustainable development of livestock production systems.

Author Contributions

Conceptualization, Y.L. and R.W.; methodology, S.L.; formal analysis, H.Z.; data curation, B.W. and S.C.; writing—original draft preparation, Y.L.; writing—review and editing, R.G., L.W., X.X. and Y.X.; supervision, K.D.; project administration, K.D.; funding acquisition, K.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research is sponsored by the National Natural Science Foundation of China (grant no. 32072771).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mishra, S.; Srivastava, S.; Dewangan, J.; Divakar, A.; Kumar Rath, S. Global occurrence of deoxynivalenol in food commodities and exposure risk assessment in humans in the last decade: A survey. Crit. Rev. Food Sci. Nutr. 2019, 60, 1346–1374. [Google Scholar] [CrossRef] [PubMed]
  2. Yuan, J.; Sun, C.; Guo, X.; Yang, T.; Wang, H.; Fu, S.; Li, C.; Yang, H. A rapid Raman detection of deoxynivalenol in agricultural products. Food Chem. 2017, 221, 797–802. [Google Scholar] [CrossRef] [PubMed]
  3. Li, B.; Duan, J.; Ren, J.; Francis, F.; Li, G. Isolation and Characterization of Two New Deoxynivalenol-Degrading Strains, Bacillus sp. HN117 and Bacillus sp. N22. Toxins 2022, 14, 781. [Google Scholar] [CrossRef] [PubMed]
  4. Fan, H.; Ren, Z.; Xu, C.; Wang, H.; Wu, Z.; Rehman, Z.U.; Wu, S.; Sun, M.A.; Bao, W. Chromatin Accessibility and Transcriptomic Alterations in Murine Ovarian Granulosa Cells upon Deoxynivalenol Exposure. Cells 2021, 10, 2818. [Google Scholar] [CrossRef]
  5. Sobrova, P.; Adam, V.; Vasatkova, A.; Beklova, M.; Zeman, L.; Kizek, R. Deoxynivalenol and its toxicity. Interdiscip. Toxicol. 2010, 3, 94–99. [Google Scholar] [CrossRef]
  6. Lancova, K.; Hajslova, J.; Kostelanska, M.; Kohoutkova, J.; Nedelnik, J.; Moravcova, H.; Vanova, M. Fate of trichothecene mycotoxins during the processing: Milling and baking. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2008, 25, 650–659. [Google Scholar] [CrossRef]
  7. Wu, Q.; Kuča, K.; Humpf, H.U.; Klímová, B.; Cramer, B. Fate of deoxynivalenol and deoxynivalenol-3-glucoside during cereal-based thermal food processing: A review study. Mycotoxin Res. 2017, 33, 79–91. [Google Scholar] [CrossRef]
  8. Zhang, H.; Wang, B. Fates of deoxynivalenol and deoxynivalenol-3-glucoside during bread and noodle processing. Food Control 2015, 50, 754–757. [Google Scholar] [CrossRef]
  9. Wolf, C.E.; Bullerman, L.B. Heat and pH alter the concentration of deoxynivalenol in an aqueous environment. J. Food Prot. 1998, 61, 365–367. [Google Scholar] [CrossRef]
  10. Voss, K.A.; Snook, M.E. Stability of the mycotoxin deoxynivalenol (DON) during the production of flour-based foods and wheat flake cereal. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2010, 27, 1694–1700. [Google Scholar] [CrossRef]
  11. Milani, J.; Maleki, G. Effects of processing on mycotoxin stability in cereals. J. Sci. Food Agric. 2014, 94, 2372–2375. [Google Scholar] [CrossRef]
  12. Vanhoutte, I.; De Tender, C.; Demeyere, K.; Abdallah, M.F.; Ommeslag, S.; Vermeir, P.; Saeger, S.; Debode, J.; Meyer, E.; Croubels, S.; et al. Bacterial Enrichment Cultures Biotransform the Mycotoxin Deoxynivalenol into a Novel Metabolite Toxic to Plant and Porcine Cells. Toxins 2021, 13, 552. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, S.S.; Li, Y.H.; Lin, M.F. Chronic Exposure to the Fusarium Mycotoxin Deoxynivalenol: Impact on Performance, Immune Organ, and Intestinal Integrity of Slow-Growing Chickens. Toxins 2017, 9, 334. [Google Scholar] [CrossRef] [PubMed]
  14. Zhai, X.; Qiu, Z.; Wang, L.; Luo, Y.; He, W.; Yang, J. Possible Toxic Mechanisms of Deoxynivalenol (DON) Exposure to Intestinal Barrier Damage and Dysbiosis of the Gut Microbiota in Laying Hens. Toxins 2022, 14, 682. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, Y.; Zhang, M.; Li, K.; Zhang, C.; Tian, H.; Luo, Y. Investigation of Deoxynivalenol Contamination in Local Area and Evaluation of Its Multiple Intestinal Toxicity. Toxins 2024, 16, 353. [Google Scholar] [CrossRef]
  16. Alassane-Kpembi, I.; Gerez, J.R.; Cossalter, A.M.; Neves, M.; Laffitte, J.; Naylies, C.; Lippi, Y.; Kolf-Clauw, M.; Bracarense, A.P.L.; Pinton, P.; et al. Intestinal toxicity of the type B trichothecene mycotoxin fusarenon-X: Whole transcriptome profiling reveals new signaling pathways. Sci. Rep. 2017, 7, 7530. [Google Scholar] [CrossRef]
  17. Xue, R.; Li, S.; Zou, H.; Ji, D.; Lv, M.; Zhou, P.; Wei, Z.; Zhang, Z.; Cao, Y. Melatonin alleviates deoxynivalenol-induced apoptosis of human granulosa cells by reducing mutually accentuated FOXO1 and ER stress. Biol. Reprod. 2021, 105, 554–566. [Google Scholar] [CrossRef]
  18. Ma, R.; Zhang, L.; Liu, M.; Su, Y.T.; Xie, W.M.; Zhang, N.Y.; Dai, J.F.; Wang, Y.; Rajput, S.A.; Qi, D.S.; et al. Individual and Combined Occurrence of Mycotoxins in Feed Ingredients and Complete Feeds in China. Toxins 2018, 10, 113. [Google Scholar] [CrossRef]
  19. Cai, Y.T.; McLaughlin, M.; Zhang, K. Advancing the FDA/Office of Regulatory Affairs Mycotoxin Program: New Analytical Method Approaches to Addressing Needs and Challenges. J. AOAC Int. 2020, 103, 705–709. [Google Scholar] [CrossRef]
  20. Eskola, M.; Kos, G.; Elliott, C.T.; Hajšlová, J.; Mayar, S.; Krska, R. Worldwide contamination of food-crops with mycotoxins: Validity of the widely cited ’FAO estimate’ of 25. Crit. Rev. Food Sci. Nutr. 2020, 60, 2773–2789. [Google Scholar] [CrossRef]
  21. Teixido-Orries, I.; Molino, F.; Femenias, A.; Ramos, A.J.; Marín, S. Quantification and classification of deoxynivalenol-contaminated oat samples by near-infrared hyperspectral imaging. Food Chem. 2023, 417, 135924. [Google Scholar] [CrossRef]
  22. Gruber-Dorninger, C.; Jenkins, T.; Schatzmayr, G. Global Mycotoxin Occurrence in Feed: A Ten-Year Survey. Toxins 2019, 11, 375. [Google Scholar] [CrossRef]
  23. Zhao, J.Y.; Cheng, T.X.; Xu, W.J.; Han, X.M.; Zhang, J.; Zhang, H.Y.; Wang, C.; Fanning, S.; Li, F.Q. Natural co-occurrence of multi-mycotoxins in unprocessed wheat grains from China. Food Control 2021, 130, 108321. [Google Scholar] [CrossRef]
  24. Xu, A.; Yu, S.; Li, Y.; Liu, H.; Yan, Z.; Wu, A.; Peng, S.; Liu, N. Total Deoxynivalenol Contamination of Wheat Products and Coarse Grains in Shanghai, China: Occurrence and Health Risk Assessment. Foods 2024, 13, 3373. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, Y.; Lu, Y.; Wang, L.; Chang, F.; Yang, L. Occurrence of deoxynivalenol in wheat, Hebei Province, China. Food Chem. 2016, 197, 1271–1274. [Google Scholar] [CrossRef]
  26. Li, F.; Jiang, D.; Zhou, J.; Chen, J.; Li, W.; Zheng, F. Mycotoxins in wheat flour and intake assessment in Shandong province of China. Food Addit. Contam. Part B Surveill. 2016, 9, 170–175. [Google Scholar] [CrossRef]
  27. Birr, T.; Jensen, T.; Preusske, N.; Sonnichsen, F.D.; De Boevre, M.; De Saeger, S.; Hasler, M.; Verreet, J.A.; Klink, H. Occurrence of Fusarium Mycotoxins and Their Modified Forms in Forage Maize Cultivars. Toxins 2021, 13, 110. [Google Scholar] [CrossRef]
  28. Twarużek, M.; Skrzydlewski, P.; Kosicki, R.; Grajewski, J. Mycotoxins survey in feed materials and feedingstuffs in years 2015-2020. Toxicon 2021, 202, 27–39. [Google Scholar] [CrossRef]
  29. Pack, E.D.; Weiland, S.; Musser, R.; Schmale, D.G. Survey of zearalenone and type-B trichothecene mycotoxins in swine feed in the USA. Mycotoxin Res. 2021, 37, 297–313. [Google Scholar] [CrossRef]
  30. Weaver, A.C.; Weaver, D.M.; Adams, N.; Yiannikouris, A. Co-Occurrence of 35 Mycotoxins: A Seven-Year Survey of Corn Grain and Corn Silage in the United States. Toxins 2021, 13, 516. [Google Scholar] [CrossRef]
  31. Silva, M.V.; Pante, G.C.; Romoli, J.C.Z.; de Souza, A.P.M.; Rocha, G.; Ferreira, F.D.; Feijó, A.L.R.; Moscardi, S.M.P.; de Paula, K.R.; Bando, E.; et al. Occurrence and risk assessment of population exposed to deoxynivalenol in foods derived from wheat flour in Brazil. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2018, 35, 546–554. [Google Scholar] [CrossRef]
  32. Zhang, D.; Zhao, L.; Chen, Y.; Gao, H.; Hua, Y.; Yuan, X.; Yang, H. Mycotoxins in Maize Silage from China in 2019. Toxins 2022, 14, 241. [Google Scholar] [CrossRef] [PubMed]
  33. Schollenberger, M.; Jara, H.T.; Suchy, S.; Drochner, W.; Müller, H.M. Fusarium toxins in wheat flour collected in an area in southwest Germany. Int. J. Food Microbiol. 2002, 72, 85–89. [Google Scholar] [CrossRef] [PubMed]
  34. Trucksess, M.W.; Ready, D.W.; Pender, M.K.; Ligmond, C.A.; Wood, G.E.; Page, S.W. Determination and survey of deoxynivalenol in white flour, whole wheat flour, and bran. J. AOAC Int. 1996, 79, 883–887. [Google Scholar] [CrossRef] [PubMed]
  35. Cano-Sancho, G.; Gauchi, J.P.; Sanchis, V.; Marin, S.; Ramos, A.J. Quantitative dietary exposure assessment of the Catalonian population (Spain) to the mycotoxin deoxynivalenol. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2011, 28, 1098–1109. [Google Scholar] [CrossRef] [PubMed]
  36. Okorski, A.; Milewska, A.; Pszczolkowska, A.; Karpiesiuk, K.; Kozera, W.; Dabrowska, J.A.; Radwinska, J. Prevalence of Fusarium fungi and Deoxynivalenol Levels in Winter Wheat Grain in Different Climatic Regions of Poland. Toxins 2022, 14, 102. [Google Scholar] [CrossRef]
  37. Pei, P.; Xiong, K.; Wang, X.; Sun, B.; Zhao, Z.; Zhang, X.; Yu, J. Predictive growth kinetic parameters and modelled probabilities of deoxynivalenol production by Fusarium graminearum on wheat during simulated storing conditions. J. Appl. Microbiol. 2022, 133, 349–361. [Google Scholar] [CrossRef]
  38. Thammawong, M.; Okadome, H.; Shiina, T.; Nakagawa, H.; Nagashima, H.; Nakajima, T.; Kushiro, M. Distinct distribution of deoxynivalenol, nivalenol, and ergosterol in Fusarium-infected Japanese soft red winter wheat milling fractions. Mycopathologia 2011, 172, 323–330. [Google Scholar] [CrossRef]
  39. Guo, Y.; Lu, T.; Shi, J.; Li, X.; Wu, K.; Xiong, Y. Identification of Deoxynivalenol and Degradation Products during Maize Germ Oil Refining Process. Foods 2022, 11, 1720. [Google Scholar] [CrossRef]
  40. Song, X.; Qiao, L.; Chang, J.; Dou, X.; Zhang, X.; Pi, S.; Xu, C. Dietary supplementation with selenium nanoparticles-enriched Lactobacillus casei ATCC 393 alleviates intestinal barrier dysfunction of mice exposed to deoxynivalenol by regulating endoplasmic reticulum stress and gut microbiota. Ecotoxicol. Environ. Saf. 2022, 248, 114276. [Google Scholar] [CrossRef]
  41. Liao, Y.; Peng, Z.; Xu, S.; Meng, Z.; Li, D.; Zhou, X.; Zhang, R.; Shi, S.; Hao, L.; Liu, L.; et al. Deoxynivalenol Exposure Induced Colon Damage in Mice Independent of the Gut Microbiota. Mol. Nutr. Food Res. 2023, 67, e2300317. [Google Scholar] [CrossRef] [PubMed]
  42. 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] [PubMed]
  43. Lu, Q.; Luo, J.Y.; Ruan, H.N.; Wang, C.J.; Yang, M.H. Structure-toxicity relationships, toxicity mechanisms and health risk assessment of food-borne modified deoxynivalenol and zearalenone: A comprehensive review. Sci. Total Environ. 2022, 806, 151192. [Google Scholar] [CrossRef] [PubMed]
  44. Feizollahi, E.; Roopesh, M.S. Mechanisms of deoxynivalenol (DON) degradation during different treatments: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 5903–5924. [Google Scholar] [CrossRef]
  45. Li, F.; Jin, J.; Rietjens, I.; Xing, F. Interindividual Differences in In Vitro Human Intestinal Microbial Conversion of 3-Acetyl-DON and 15-Acetyl-DON. Toxins 2022, 14, 199. [Google Scholar] [CrossRef]
  46. Li, K.; Wang, L.; Yu, D.; Yan, Z.; Liu, N.; Wu, A. Cellobiose inhibits the release of deoxynivalenol from transformed deoxynivalenol-3-glucoside from Lactiplantibacillus plantarum. Food Chem. 2022, 4, 100077. [Google Scholar] [CrossRef]
  47. Ruhnau, D.; Hess, C.; Doupovec, B.; Grenier, B.; Schatzmayr, D.; Hess, M.; Awad, W. Deepoxy-deoxynivalenol (DOM-1), a derivate of deoxynivalenol (DON), exhibits less toxicity on intestinal barrier function, Campylobacter jejuni colonization and translocation in broiler chickens. Gut Pathog. 2021, 13, 44. [Google Scholar] [CrossRef]
  48. Li, X.Z.; Hassan, Y.I.; Lepp, D.; Zhu, Y.; Zhou, T. 3-keto-DON, but Not 3-epi-DON, Retains the in Planta Toxicological Potential after the Enzymatic Biotransformation of Deoxynivalenol. Int. J. Mol. Sci. 2022, 23, 7230. [Google Scholar] [CrossRef]
  49. Wu, L.; Wang, B. Transformation of deoxynivalenol and its acetylated derivatives in Chinese steamed bread making, as affected by pH, yeast, and steaming time. Food Chem. 2016, 202, 149–155. [Google Scholar] [CrossRef]
  50. Zhang, H.J.; Sun, J.; Zhang, Y.; Lu, M.B.; Sun, L.J.; Li, W.X.; Hu, X.X.; Wang, B.J. Retention of deoxynivalenol and its derivatives during storage of wheat grain and flour. Food Control 2016, 65, 177–181. [Google Scholar] [CrossRef]
  51. Bryla, M.; Ksieniewicz-Wozniak, E.; Waskiewicz, A.; Szymczyk, K.; Jedrzejczak, R. Co-occurrence of nivalenol, deoxynivalenol and deoxynivalenol-3-glucoside in beer samples. Food Control 2018, 92, 319–324. [Google Scholar] [CrossRef]
  52. Guerrero-Netro, H.M.; Barreta, M.H.; Costa, E.; Goetten, A.; Dupras, R.; Mills, L.; Koch, J.; Portela, V.M.; Price, C.A.; Chorfi, Y. Effects of the mycotoxin metabolite de-epoxy-deoxynivalenol (DOM-1) on embryo development and sperm motility in cattle. J. Appl. Toxicol. 2021, 41, 1180–1187. [Google Scholar] [CrossRef] [PubMed]
  53. Qu, R.; Jiang, C.; Wu, W.; Pang, B.; Lei, S.; Lian, Z.; Shao, D.; Jin, M.; Shi, J. Conversion of DON to 3-epi-DON in vitro and toxicity reduction of DON in vivo by Lactobacillus rhamnosus. Food Funct. 2019, 10, 2785–2796. [Google Scholar] [CrossRef] [PubMed]
  54. Springler, A.; Hessenberger, S.; Reisinger, N.; Kern, C.; Nagl, V.; Schatzmayr, G.; Mayer, E. Deoxynivalenol and its metabolite deepoxy-deoxynivalenol: Multi-parameter analysis for the evaluation of cytotoxicity and cellular effects. Mycotoxin Res. 2017, 33, 25–37. [Google Scholar] [CrossRef]
  55. Mengelers, M.; Zeilmaker, M.; Vidal, A.; De Boevre, M.; De Saeger, S.; Hoogenveen, R. Biomonitoring of Deoxynivalenol and Deoxynivalenol-3-glucoside in Human Volunteers: Renal Excretion Profiles. Toxins 2019, 11, 466. [Google Scholar] [CrossRef]
  56. Chen, L.; Yu, M.; Wu, Q.; Peng, Z.; Wang, D.; Kuča, K.; Yao, P.; Yan, H.; Nüssler, A.K.; Liu, L.; et al. Gender and geographical variability in the exposure pattern and metabolism of deoxynivalenol in humans: A review. J. Appl. Toxicol. 2017, 37, 60–70. [Google Scholar] [CrossRef]
  57. Gratz, S.W.; Duncan, G.; Richardson, A.J. The human fecal microbiota metabolizes deoxynivalenol and deoxynivalenol-3-glucoside and may be responsible for urinary deepoxy-deoxynivalenol. Appl. Environ. Microbiol. 2013, 79, 1821–1825. [Google Scholar] [CrossRef]
  58. Vidal, A.; Claeys, L.; Mengelers, M.; Vanhoorne, V.; Vervaet, C.; Huybrechts, B.; De Saeger, S.; De Boevre, M. Humans significantly metabolize and excrete the mycotoxin deoxynivalenol and its modified form deoxynivalenol-3-glucoside within 24 hours. Sci. Rep. 2018, 8, 5255. [Google Scholar] [CrossRef]
  59. Deng, C.; Li, C.; Zhou, S.; Wang, X.; Xu, H.; Wang, D.; Gong, Y.Y.; Routledge, M.N.; Zhao, Y.; Wu, Y. Risk assessment of deoxynivalenol in high-risk area of China by human biomonitoring using an improved high throughput UPLC-MS/MS method. Sci. Rep. 2018, 8, 3901. [Google Scholar] [CrossRef]
  60. Gerding, J.; Cramer, B.; Humpf, H.U. Determination of mycotoxin exposure in Germany using an LC-MS/MS multibiomarker approach. Mol. Nutr. Food Res. 2014, 58, 2358–2368. [Google Scholar] [CrossRef]
  61. Heyndrickx, E.; Sioen, I.; Huybrechts, B.; Callebaut, A.; De Henauw, S.; De Saeger, S. Human biomonitoring of multiple mycotoxins in the Belgian population: Results of the BIOMYCO study. Environ. Int. 2015, 84, 82–89. [Google Scholar] [CrossRef]
  62. Wells, L.; Hardie, L.; Williams, C.; White, K.; Liu, Y.; De Santis, B.; Debegnach, F.; Moretti, G.; Greetham, S.; Brera, C.; et al. Deoxynivalenol Biomarkers in the Urine of UK Vegetarians. Toxins 2017, 9, 196. [Google Scholar] [CrossRef]
  63. De Santis, B.; Debegnach, F.; Miano, B.; Moretti, G.; Sonego, E.; Chiaretti, A.; Buonsenso, D.; Brera, C. Determination of Deoxynivalenol Biomarkers in Italian Urine Samples. Toxins 2019, 11, 441. [Google Scholar] [CrossRef] [PubMed]
  64. Martins, C.; Torres, D.; Lopes, C.; Correia, D.; Goios, A.; Assunção, R.; Alvito, P.; Vidal, A.; De Boevre, M.; De Saeger, S.; et al. Deoxynivalenol exposure assessment through a modelling approach of food intake and biomonitoring data—A contribution to the risk assessment of an enteropathogenic mycotoxin. Food Res. Int. 2021, 140, 109863. [Google Scholar] [CrossRef] [PubMed]
  65. Dänicke, S.; Valenta, H.; Döll, S. On the toxicokinetics and the metabolism of deoxynivalenol (DON) in the pig. Arch. Anim. Nutr. 2004, 58, 169–180. [Google Scholar] [CrossRef] [PubMed]
  66. Lauwers, M.; Croubels, S.; Letor, B.; Gougoulias, C.; Devreese, M. Biomarkers for Exposure as A Tool for Efficacy Testing of A Mycotoxin Detoxifier in Broiler Chickens and Pigs. Toxins 2019, 11, 187. [Google Scholar] [CrossRef]
  67. Dänicke, S.; Brüssow, K.P.; Valenta, H.; Ueberschär, K.H.; Tiemann, U.; Schollenberger, M. On the effects of graded levels of Fusarium toxin contaminated wheat in diets for gilts on feed intake, growth performance and metabolism of deoxynivalenol and zearalenone. Mol. Nutr. Food Res. 2005, 49, 932–943. [Google Scholar] [CrossRef]
  68. Goyarts, T.; Dänicke, S. Effects of deoxynivalenol (DON) on growth performance, nutrient digestibility and DON metabolism in pigs. Mycotoxin Res. 2005, 21, 139–142. [Google Scholar] [CrossRef]
  69. Panisson, J.C.; Wellington, M.O.; Bosompem, M.A.; Nagl, V.; Schwartz-Zimmermann, H.E.; Columbus, D.A. Urinary and Serum Concentration of Deoxynivalenol (DON) and DON Metabolites as an Indicator of DON Contamination in Swine Diets. Toxins 2023, 15, 120. [Google Scholar] [CrossRef]
  70. Riahi, I.; Marquis, V.; Pérez-Vendrell, A.M.; Brufau, J.; Esteve-Garcia, E.; Ramos, A.J. Effects of Deoxynivalenol-Contaminated Diets on Metabolic and Immunological Parameters in Broiler Chickens. Animals 2021, 11, 147. [Google Scholar] [CrossRef]
  71. Schwartz-Zimmermann, H.E.; Fruhmann, P.; Dänicke, S.; Wiesenberger, G.; Caha, S.; Weber, J.; Berthiller, F. Metabolism of deoxynivalenol and deepoxy-deoxynivalenol in broiler chickens, pullets, roosters and turkeys. Toxins 2015, 7, 4706–4729. [Google Scholar] [CrossRef]
  72. Yunus, A.W.; Blajet-Kosicka, A.; Kosicki, R.; Khan, M.Z.; Rehman, H.; Böhm, J. Deoxynivalenol as a contaminant of broiler feed: Intestinal development, absorptive functionality, and metabolism of the mycotoxin. Poult. Sci. 2012, 91, 852–861. [Google Scholar] [CrossRef]
  73. Dänicke, S.; Brezina, U. Kinetics and metabolism of the Fusarium toxin deoxynivalenol in farm animals: Consequences for diagnosis of exposure and intoxication and carry over. Food Chem. Toxicol. 2013, 60, 58–75. [Google Scholar] [CrossRef] [PubMed]
  74. Debevere, S.; Cools, A.; Baere, S.; Haesaert, G.; Rychlik, M.; Croubels, S.; Fievez, V. In Vitro Rumen Simulations Show a Reduced Disappearance of Deoxynivalenol, Nivalenol and Enniatin B at Conditions of Rumen Acidosis and Lower Microbial Activity. Toxins 2020, 12, 101. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed]
  76. Deng, Y.; You, L.; Wang, X.; Wu, W.; Kuca, K.; Wu, Q.; Wei, W. Deoxynivalenol: Emerging Toxic Mechanisms and Control Strategies, Current and Future Perspectives. J. Agric. Food Chem. 2023, 71, 10901–10915. [Google Scholar] [CrossRef]
  77. Aihara, R.; Ookawara, T.; Morimoto, A.; Iwashita, N.; Takagi, Y.; Miyasaka, A.; Kushiro, M.; Miyake, S.; Fukuyama, T. Acute and subacute oral administration of mycotoxin deoxynivalenol exacerbates the pro-inflammatory and pro-pruritic responses in a mouse model of allergic dermatitis. Arch. Toxicol. 2020, 94, 4197–4207. [Google Scholar] [CrossRef]
  78. Seyed Toutounchi, N.; Braber, S.; Van’t Land, B.; Thijssen, S.; Garssen, J.; Kraneveld, A.D.; Folkerts, G.; Hogenkamp, A. Exposure to Deoxynivalenol During Pregnancy and Lactation Enhances Food Allergy and Reduces Vaccine Responsiveness in the Offspring in a Mouse Model. Front. Immunol. 2021, 12, 797152. [Google Scholar] [CrossRef]
  79. Gonya, S.; Kallmerten, P.; Dinapoli, P. Are Infants and Children at Risk of Adverse Health Effects from Dietary Deoxynivalenol Exposure? An Integrative Review. Int. J. Environ. Res. Public Health 2024, 21, 808. [Google Scholar] [CrossRef]
  80. Pestka, J.J.; Smolinski, A.T. Deoxynivalenol: Toxicology and potential effects on humans. J. Toxicol. Environ. Health B Crit. Rev. 2005, 8, 39–69. [Google Scholar] [CrossRef]
  81. Yue, J.; Guo, D.; Gao, X.; Wang, J.; Nepovimova, E.; Wu, W.; Kuca, K. Deoxynivalenol (Vomitoxin)-Induced Anorexia Is Induced by the Release of Intestinal Hormones in Mice. Toxins 2021, 13, 512. [Google Scholar] [CrossRef] [PubMed]
  82. EFSA Panel on Contaminants in the Food Chain (CONTAM); Knutsen, H.K.; Alexander, J.; Barregard, L.; Bignami, M.; Bruschweiler, B.; Ceccatelli, S.; Cottrill, B.; Dinovi, M.; Grasl-Kraupp, B.; et al. Risks to human and animal health related to the presence of deoxynivalenol and its acetylated and modified forms in food and feed. EFSA J. 2017, 15, e04718. [Google Scholar] [CrossRef] [PubMed]
  83. Forsyth, D.M.; Yoshizawa, T.; Morooka, N.; Tuite, J. Emetic and refusal activity of deoxynivalenol to swine. Appl. Environ. Microbiol. 1977, 34, 547–552. [Google Scholar] [CrossRef] [PubMed]
  84. Holanda, D.M.; Kim, S.W. Mycotoxin Occurrence, Toxicity, and Detoxifying Agents in Pig Production with an Emphasis on Deoxynivalenol. Toxins 2021, 13, 171. [Google Scholar] [CrossRef]
  85. Lafleur Lariviere, E.; Zhu, C.; Sharma, A.; Karrow, N.A.; Huber, L.A. The effects of deoxynivalenol-contaminated corn in low-complexity diets supplemented with either an immune-modulating feed additive, or fish oil on nursery pig growth performance, immune response, small intestinal morphology, and component digestibility. Transl. Anim. Sci. 2022, 6, txac068. [Google Scholar] [CrossRef]
  86. Hooft, J.M.; Bureau, D.P. Deoxynivalenol: Mechanisms of action and its effects on various terrestrial and aquatic species. Food Chem. Toxicol. 2021, 157, 112616. [Google Scholar] [CrossRef]
  87. Awad, W.A.; Aschenbach, J.R.; Zentek, J. Cytotoxicity and metabolic stress induced by deoxynivalenol in the porcine intestinal IPEC-J2 cell line. J. Anim. Physiol. Anim. Nutr. 2012, 96, 709–716. [Google Scholar] [CrossRef]
  88. Chen, J.; Zhou, Z.; Wu, N.; Li, J.; Xi, N.; Xu, M.; Wu, F.; Fu, Q.; Yan, G.; Liu, Y.; et al. Chlorogenic acid attenuates deoxynivalenol-induced apoptosis and pyroptosis in human keratinocytes via activating Nrf2/HO-1 and inhibiting MAPK/NF-κB/NLRP3 pathways. Biomed. Pharmacother. 2024, 170, 116003. [Google Scholar] [CrossRef]
  89. Singh, S.; Banerjee, S.; Chattopadhyay, P.; Borthakur, S.K.; Veer, V. Deoxynivalenol induces cytotoxicity and genotoxicity in animal primary cell culture. Toxicol. Mech. Methods 2015, 25, 184–191. [Google Scholar] [CrossRef]
  90. Wu, W.; Zhou, H.R.; Pan, X.; Pestka, J.J. Comparison of Anorectic Potencies of the Trichothecenes T-2 Toxin, HT-2 Toxin and Satratoxin G to the Ipecac Alkaloid Emetine. Toxicol. Rep. 2015, 2, 238–251. [Google Scholar] [CrossRef]
  91. Doi, K.; Uetsuka, K. Mechanisms of mycotoxin-induced neurotoxicity through oxidative stress-associated pathways. Int. J. Mol. Sci. 2011, 12, 5213–5237. [Google Scholar] [CrossRef] [PubMed]
  92. Liao, Y.; Peng, Z.; Chen, L.; Nüssler, A.K.; Liu, L.; Yang, W. Deoxynivalenol, gut microbiota and immunotoxicity: A potential approach? Food Chem. Toxicol. 2018, 112, 342–354. [Google Scholar] [CrossRef] [PubMed]
  93. Bonnet, M.S.; Roux, J.; Mounien, L.; Dallaporta, M.; Troadec, J.D. Advances in deoxynivalenol toxicity mechanisms: The brain as a target. Toxins 2012, 4, 1120–1138. [Google Scholar] [CrossRef] [PubMed]
  94. Xiong, S.; Li, X.; Zhao, C.; Gao, J.; Yuan, W.; Zhang, J. The Degradation of Deoxynivalenol by Using Electrochemical Oxidation with Graphite Electrodes and the Toxicity Assessment of Degradation Products. Toxins 2019, 11, 478. [Google Scholar] [CrossRef]
  95. Zhang, J.; You, L.; Wu, W.; Wang, X.; Chrienova, Z.; Nepovimova, E.; Wu, Q.; Kuca, K. The neurotoxicity of trichothecenes T-2 toxin and deoxynivalenol (DON): Current status and future perspectives. Food Chem. Toxicol. 2020, 145, 111676. [Google Scholar] [CrossRef]
  96. Cao, L.; Jiang, Y.; Zhu, L.; Xu, W.; Chu, X.; Zhang, Y.; Rahman, S.U.; Feng, S.; Li, Y.; Wu, J.; et al. Deoxynivalenol Induces Caspase-8-Mediated Apoptosis through the Mitochondrial Pathway in Hippocampal Nerve Cells of Piglet. Toxins 2021, 13, 73. [Google Scholar] [CrossRef]
  97. Wang, X.; Chen, X.; Cao, L.; Zhu, L.; Zhang, Y.; Chu, X.; Zhu, D.; Rahman, S.U.; Peng, C.; Feng, S.; et al. Mechanism of deoxynivalenol-induced neurotoxicity in weaned piglets is linked to lipid peroxidation, dampened neurotransmitter levels, and interference with calcium signaling. Ecotoxicol. Environ. Saf. 2020, 194, 110382. [Google Scholar] [CrossRef]
  98. Faeste, C.K.; Solhaug, A.; Gaborit, M.; Pierre, F.; Massotte, D. Neurotoxic Potential of Deoxynivalenol in Murine Brain Cell Lines and Primary Hippocampal Cultures. Toxins 2022, 14, 48. [Google Scholar] [CrossRef]
  99. Mishra, S.; Kapoor, R.; Sushma; Kanchan, S.; Jha, G.; Sharma, D.; Tomar, B.; Rath, S.K. Deoxynivalenol Induces Drp-1-Mediated Mitochondrial Dysfunction via Elevating Oxidative Stress. Chem. Res. Toxicol. 2024, 37, 1139–1154. [Google Scholar] [CrossRef]
  100. Jacobson, M. On the Infectious Causes of Neonatal Piglet Diarrhoea—A Review. Vet. Sci. 2022, 9, 422. [Google Scholar] [CrossRef]
  101. Jia, B.; Lin, H.; Yu, S.; Liu, N.; Yu, D.; Wu, A. Mycotoxin deoxynivalenol-induced intestinal flora disorders, dysfunction and organ damage in broilers and pigs. J. Hazard. Mater. 2023, 451, 131172. [Google Scholar] [CrossRef] [PubMed]
  102. 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] [PubMed]
  103. Liu, M.; Zhang, L.; Chu, X.H.; Ma, R.; Wang, Y.W.; Liu, Q.; Zhang, N.Y.; Karrow, N.A.; Sun, L.H. Effects of deoxynivalenol on the porcine growth performance and intestinal microbiota and potential remediation by a modified HSCAS binder. Food Chem Toxicol. 2020, 141, 111373. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, C.; Huang, L.; Wang, P.; Liu, Q.; Wang, J. The Effects of Deoxynivalenol on the Ultrastructure of the Sacculus Rotundus and Vermiform Appendix, as Well as the Intestinal Microbiota of Weaned Rabbits. Toxins 2020, 12, 569. [Google Scholar] [CrossRef]
  105. Li, D.; Ma, H.; Ye, Y.; Ji, C.; Tang, X.; Ouyang, D.; Chen, J.; Li, Y.; Ma, Y. Deoxynivalenol induces apoptosis in mouse thymic epithelial cells through mitochondria-mediated pathway. Environ. Toxicol. Pharmacol. 2014, 38, 163–171. [Google Scholar] [CrossRef]
  106. Cheat, S.; Pinton, P.; Cossalter, A.M.; Cognie, J.; Vilariño, M.; Callu, P.; Raymond-Letron, I.; Oswald, I.P.; Kolf-Clauw, M. The mycotoxins deoxynivalenol and nivalenol show in vivo synergism on jejunum enterocytes apoptosis. Food Chem. Toxicol. 2016, 87, 45–54. [Google Scholar] [CrossRef]
  107. Skiepko, N.; Przybylska-Gornowicz, B.; Gajecka, M.; Gajecki, M.; Lewczuk, B. Effects of Deoxynivalenol and Zearalenone on the Histology and Ultrastructure of Pig Liver. Toxins 2020, 12, 463. [Google Scholar] [CrossRef]
  108. Moon, Y.; Kim, H.K.; Suh, H.; Chung, D.H. Toxic alterations in chick embryonic liver and spleen by acute exposure to Fusarium-producing mycotoxin deoxynivalenol. Biol. Pharm. Bull. 2007, 30, 1808–1812. [Google Scholar] [CrossRef]
  109. Hasuda, A.L.; Person, E.; Khoshal, A.K.; Bruel, S.; Puel, S.; Oswald, I.P.; Bracarense, A.; Pinton, P. Deoxynivalenol induces apoptosis and inflammation in the liver: Analysis using precision-cut liver slices. Food Chem. Toxicol. 2022, 163, 112930. [Google Scholar] [CrossRef]
  110. Bracarense, A.P.F.L.; Basso, K.M.; Da Silva, E.O.; Payros, D.; Oswald, I.P. Deoxynivalenol in the liver and lymphoid organs of rats: Effects of dose and duration on immunohistological changes. World Mycotoxin J. 2017, 10, 89–96. [Google Scholar] [CrossRef]
  111. de Souza, M.; Baptista, A.A.S.; Valdiviezo, M.J.J.; Justino, L.; Menck-Costa, M.F.; Ferraz, C.R.; da Gloria, E.M.; Verri, W.A., Jr.; Bracarense, A. 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]
  112. Zielonka, L.; Gajecka, M.; Tarasiuk, M.; Gajecki, M. The effects of dietary deoxynivalenol (DON) on selected blood biochemical and hematological parameters in pre-pubertal gilts. Pol. J. Vet. Sci. 2015, 18, 223–231. [Google Scholar] [CrossRef]
  113. Wu, L.; Liao, P.; He, L.; Ren, W.; Yin, J.; Duan, J.; Li, T. Growth performance, serum biochemical profile, jejunal morphology, and the expression of nutrients transporter genes in deoxynivalenol (DON)-challenged growing pigs. BMC Vet. Res. 2015, 11, 144. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, L.; Wang, Y.; Wang, W.; Wang, L.; Shi, J.; Cheng, J.; Zhang, J.; Li, A.; He, B.; Fan, Z. Effects of Deoxynivalenol Detoxifier on Growth Performance, Blood Biochemical Indices, and Microbiota Composition of Piglets. Int. J. Mol. Sci. 2025, 26, 2045. [Google Scholar] [CrossRef] [PubMed]
  115. Ghareeb, K.; Awad, W.A.; Zebeli, Q.; Bohm, J. Deoxynivalenol in chicken feed alters the vaccinal immune response and clinical biochemical serum parameters but not the intestinal and carcass characteristics. J. Anim. Physiol. Anim. Nutr. 2016, 100, 53–60. [Google Scholar] [CrossRef] [PubMed]
  116. Riahi, I.; Marquis, V.; Ramos, A.J.; Brufau, J.; Esteve-Garcia, E.; Perez-Vendrell, A.M. Effects of Deoxynivalenol-Contaminated Diets on Productive, Morphological, and Physiological Indicators in Broiler Chickens. Animals 2020, 10, 1795. [Google Scholar] [CrossRef]
  117. Bernhoft, A.; Hogasen, H.R.; Rosenlund, G.; Moldal, T.; Grove, S.; Berntssen, M.H.G.; Thoresen, S.I.; Alexander, J. Effects of dietary deoxynivalenol or ochratoxin A on performance and selected health indices in Atlantic salmon (Salmo salar). Food Chem. Toxicol. 2018, 121, 374–386. [Google Scholar] [CrossRef]
  118. Tola, S.; Bureau, D.P.; Hooft, J.M.; Beamish, F.W.; Sulyok, M.; Krska, R.; Encarnacao, P.; Petkam, R. Effects of Wheat Naturally Contaminated with Fusarium Mycotoxins on Growth Performance and Selected Health Indices of Red Tilapia (Oreochromis niloticus × O. mossambicus). Toxins 2015, 7, 1929–1944. [Google Scholar] [CrossRef]
  119. Matejova, I.; Modra, H.; Blahova, J.; Franc, A.; Fictum, P.; Sevcikova, M.; Svobodova, Z. The effect of mycotoxin deoxynivalenol on haematological and biochemical indicators and histopathological changes in rainbow trout (Oncorhynchus mykiss). Biomed. Res. Int. 2014, 2014, 310680. [Google Scholar] [CrossRef]
  120. Sprando, R.L.; Collins, T.F.; Black, T.N.; Olejnik, N.; Rorie, J.I.; Eppley, R.M.; Ruggles, D.I. Characterization of the effect of deoxynivalenol on selected male reproductive endpoints. Food Chem. Toxicol. 2005, 43, 623–635. [Google Scholar] [CrossRef]
  121. Ruan, Y.B.; Liu, X.H.; Jiang, J.Z.; Nie, T.; Ma, J. Leydig cells pyroptosis in testis mediates deoxynivalenol-induced male reproductive toxicity in mice. Sci. Total Environ. 2024, 954, 176432. [Google Scholar] [CrossRef]
  122. Cao, Z.; Huang, W.; Sun, Y.; Li, Y. Deoxynivalenol induced spermatogenesis disorder by blood-testis barrier disruption associated with testosterone deficiency and inflammation in mice. Environ. Pollut. 2020, 264, 114748. [Google Scholar] [CrossRef] [PubMed]
  123. Gerez, J.R.; Desto, S.S.; Bracarense, A. Deoxynivalenol induces toxic effects in the ovaries of pigs: An ex vivo approach. Theriogenology 2017, 90, 94–100. [Google Scholar] [CrossRef] [PubMed]
  124. Lemos, G.A.A.; Gerez, J.R.; Costa, J.B.; Venâncio, E.J.; Souza, M.; Favaron, P.O.; Greghi, J.R.; Gloria, E.M.; Staurengo-Ferrari, L.; Verri, W.A.; et al. Deoxynivalenol induces ovarian damage and uterine changes in prepubertal and adult mice. Toxicon 2024, 251, 108123. [Google Scholar] [CrossRef]
  125. Toutounchi, N.S.; Braber, S.; Land, B.V.; Thijssen, S.; Garssen, J.; Folkerts, G.; Hogenkamp, A. Deoxynivalenol exposure during pregnancy has adverse effects on placental structure and immunity in mice model. Reprod. Toxicol. 2022, 112, 109–118. [Google Scholar] [CrossRef]
  126. Wang, G.; Wang, Y.; Ji, F.; Xu, L.; Yu, M.; Shi, J.; Xu, J. Biodegradation of deoxynivalenol and its derivatives by Devosia insulae A16. Food Chem. 2019, 276, 436–442. [Google Scholar] [CrossRef]
  127. Yao, J.; Chen, S.; Li, Y.; Liao, C.; Shang, K.; Guo, R.; Chen, J.; Wang, L.; Xia, X.; Yu, Z.; et al. Unveiling a Novel Antidote for Deoxynivalenol Contamination: Isolation, Identification, Whole Genome Analysis and In Vivo Safety Evaluation of Lactobacillus rhamnosus MY-1. Foods 2024, 13, 2057. [Google Scholar] [CrossRef]
  128. He, C.; Fan, Y.; Liu, G.; Zhang, H. Isolation and identification of a strain of Aspergillus tubingensis with deoxynivalenol biotransformation capability. Int. J. Mol. Sci. 2008, 9, 2366–2375. [Google Scholar] [CrossRef]
  129. Alassane-Kpembi, I.; Pinton, P.; Hupé, J.F.; Neves, M.; Lippi, Y.; Combes, S.; Castex, M.; Oswald, I.P. Saccharomyces cerevisiae Boulardii Reduces the Deoxynivalenol-Induced Alteration of the Intestinal Transcriptome. Toxins 2018, 10, 199. [Google Scholar] [CrossRef]
  130. Hassan, Y.I.; He, J.W.; Perilla, N.; Tang, K.; Karlovsky, P.; Zhou, T. The enzymatic epimerization of deoxynivalenol by Devosia mutans proceeds through the formation of 3-keto-DON intermediate. Sci. Rep. 2017, 7, 6929. [Google Scholar] [CrossRef]
  131. He, W.J.; Zhang, L.; Yi, S.Y.; Tang, X.L.; Yuan, Q.S.; Guo, M.W.; Wu, A.B.; Qu, B.; Li, H.P.; Liao, Y.C. An aldo-keto reductase is responsible for Fusarium toxin-degrading activity in a soil Sphingomonas strain. Sci. Rep. 2017, 7, 9549. [Google Scholar] [CrossRef] [PubMed]
  132. Huang, P.; Yu, X.; Liu, H.; Ding, M.; Wang, Z.; Xu, J.R.; Jiang, C. Regulation of TRI5 expression and deoxynivalenol biosynthesis by a long non-coding RNA in Fusarium graminearum. Nat. Commun. 2024, 15, 1216. [Google Scholar] [CrossRef]
  133. Sun, S.; Yu, D.; Guo, M.; Tang, M.; Yan, Z.; Sun, W.; Wu, A. The transcription factor FgSfp1 orchestrates mycotoxin deoxynivalenol biosynthesis in Fusarium graminearum. Commun. Biol. 2024, 7, 1584. [Google Scholar] [CrossRef]
  134. Shi, W.T.; Yao, C.P.; Liu, W.H.; Cao, W.Y.; Shao, W.; Liao, S.Q.; Yu, T.; Zhu, Q.F.; Chen, Z.; Zang, Y.J.; et al. An fusaric acid-based CRISPR library screen identifies MDH2 as a broad-spectrum regulator of Fusarium toxin-induced cell death. J. Hazard. Mater. 2024, 480, 135937. [Google Scholar] [CrossRef] [PubMed]
  135. Sharma, P.; Gangola, M.P.; Huang, C.; Kutcher, H.R.; Ganeshan, S.; Chibbar, R.N. Single Nucleotide Polymorphisms in B-Genome Specific UDP-Glucosyl Transferases Associated with Fusarium Head Blight Resistance and Reduced Deoxynivalenol Accumulation in Wheat Grain. Phytopathology 2018, 108, 124–132. [Google Scholar] [CrossRef] [PubMed]
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.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.