Skip to main content

Nutritional impact of mycotoxins in food animal production and strategies for mitigation

Abstract

Mycotoxins are toxic secondary metabolites produced by filamentous fungi that are commonly detected as natural contaminants in agricultural commodities worldwide. Mycotoxin exposure can lead to mycotoxicosis in both animals and humans when found in animal feeds and food products, and at lower concentrations can affect animal performance by disrupting nutrient digestion, absorption, metabolism, and animal physiology. Thus, mycotoxin contamination of animal feeds represents a significant issue to the livestock industry and is a health threat to food animals. Since prevention of mycotoxin formation is difficult to undertake to avoid contamination, mitigation strategies are needed. This review explores how the mycotoxins aflatoxins, deoxynivalenol, zearalenone, fumonisins and ochratoxin A impose nutritional and metabolic effects on food animals and summarizes mitigation strategies to reduce the risk of mycotoxicity.

Introduction

A rapidly growing world population and increased standard of living in many countries has driven increasing demand for high quality food. The livestock industry contributes to global food supply and human society by converting animal feed, which is generally made from low-value agricultural products or by-products to high value animal-derived food products that are rich in nutrients such as meat, eggs, and milk. For food animals to achieve their genetically selected production potential, nutrients must be made available and partitioned towards productive functions. The overall availability of these resources is determined by food intake, gastrointestinal digestion, and absorption [1], thus limiting factors of nutrient availability could constrain animal performance.

Mycotoxins are secondary metabolites produced by filamentous fungi as coping strategies under environmental pressure and functioning as fitness factors to enhance pathogenicity, aggressiveness and/or virulence the virulence of fungi [2, 3]. Mycotoxins are among one of the most significant hazards to the feed supply chain and pose a threat to feed industries worldwide with a direct impact on feed safety, animal health and productivity, human health via animal by-products, economies and international trade [4,5,6]. These toxic compounds are commonly detected as natural contaminants in a variety of agricultural commodities of plant origin, especially in cereal grains, and are therefore often detected in animal feeds containing corn, soybean, and wheat [5, 7,8,9], but can also be present in silage, haylage and pasture [10,11,12]. Major mycotoxins can also be found in animal-derived products such as eggs, meat, milk and milk by-products with varying residual concentrations due to carry-over from animals that have consumed contaminated feeds [13, 14]. Natural co-occurrence of mycotoxins with additive, antagonistic, or synergistic is more common in foods and feeds than single mycotoxin contaminants [15, 16]. The ingestion of mycotoxin-contaminated feeds by animals can cause both acute and chronic toxicity. However, a major problem associated with mycotoxin contamination in livestock production is the chronic effects on growth, production and immune function associated with a range of metabolic, physiological, and immunological disturbances in animals induced by the ingestion of low levels of a single or combined mycotoxins, which could result in economic losses [4, 17]. This review specifically focuses on the nutritional effects of mycotoxins on food animals. The mycotoxins most commonly addressed in this review include aflatoxins (AF), deoxynivalenol (DON), zearalenone (ZEN), fumonisins (FUM) and ochratoxin A (OTA) based on their prevalence, toxicity and relevance to food animals. In addition, the main mitigation strategies for mycotoxin contamination are presented in this review.

Major mycotoxins

Aflatoxins are produced pre- and post-harvest, mainly by Aspergillus flavus and A. parasiticus [7]. Among the AF family, AFB1 is the most commonly occurring and is considered to be the most potent carcinogenic toxin known to both animals and humans [5]. A wide variety of agricultural commodities including corn, wheat, and rice can be contaminated with AF [7]. Two common climate conditions associated with tropical and subtropical regions favor AF contamination of crops; high temperature and low humidity facilitate pre-harvest production and accumulation of AF in the growing plants [18, 19], on the other hand, exposure to high temperatures and high moisture leading up the harvest and during crop storage also favors fungal growth and AF production [20].

The trichothecenes (TRC) are produced predominately by the Fusarium species [5], which usually infect crops and produce mycotoxins in the field, generally in association with a cool and excessively wet conditions [8]. Deoxynivalenol is produced mainly by F. graminearum and F. culmorum and is the predominate TRC considering it is the most widely distributed and the most frequently detected mycotoxin in cereal grains such as corn, wheat, barley, and oats worldwide. Exposure to DON has been reported to have negative impacts on animals, especially pigs, which makes it a highly relevant mycotoxin to livestock husbandry even though it is among the least acutely toxic TRC in the family [21]. DON can interact with the neural dopaminergic system after ingestion and cause nausea [22]. It can also inhibit protein synthesis [23].

Zearalenone is another major mycotoxin primarily produced by F. graminearum, but can also be produced by F. culmorum, F. cerealis and F. equiseti. Zearalenone contaminates cereals worldwide [21].

The FUM are produced by several species of Fusarium among which F. verticillioides and F. proliferatum are the main producers [21]. High temperatures and low precipitation around corn silking facilitate fungal infection and subsequent FUM contamination [20]. Fumonisins structurally resemble sphingolipids and can inhibit de novo sphingolipid biosynthesis by potently inhibiting ceramide synthases [24]. The most prevalent member of the FUM family is fumonisin B1 (FB1) [25].

Ochratoxins (OT) are produced by various A. spp. and Penicillium spp. The most abundant and harmful of the OT is OTA, which is primarily produced by A. ochraceus and P. verrucosum [21, 26]. Ochratoxin A inhibits protein synthesis, disrupts cell-cycle progression, and induce DNA adducts. It also inhibits ATP production and induce production of reactive oxygen and nitrogen species in mitochondria [27,28,29].

Due to the global occurrence of these mycotoxins and animal health risks concerning the intake of mycotoxins, the level of these mycotoxins in cereals and feed are currently regulated in a number of countries around the world [30]. Table 1 summarized the global occurrence of these mycotoxins as well as action levels and advisory/guideline limits set by European Commission, United States Food and Drug Administration and Canadian Food Inspection Agency for maize and maize products as well as finished feed for the animal species commonly addressed in the present review.

Table 1 Permitted/guidance levels of regulatory bodies for selected mycotoxins

Nutritional impact of major mycotoxins

Carbohydrates, lipids, and proteins are the major constituents of feed and serve as fuel molecules for animals as well as building blocks for growth and development of essential cellular components. The digestion of these nutrients and the subsequent absorption of the digestive end-products make it possible for cells and tissues to utilize them for proper functionality. Efficient utilization of these macronutrients is crucial to food animal production. Non-ruminant animals are generally considered more susceptible to nutritional effects of mycotoxins compared to ruminants [31, 32]. As a result, more research has been dedicated to poultry and swine than to ruminants.

Feed intake

Feed intake is one factor affecting nutrient availability [33]. Effects of various mycotoxins on feed intake have been documented for various animal models (Table 2). However, limited data on FUM and OTA has been reported compared to AF, DON and ZEN. Effects of AF and DON on feed intake have been reportedly pronounced over a wide range of concentrations compared with ZEN and FUM (Table 2). However, concentrations of individual mycotoxins used in the reviewed studies that affected feed intake were all above the permitted/guidance levels of regulatory bodies where available (Table 1). The concentration of AF that reportedly reduced feed intake in poultry was as low as 0.04 mg/kg of feed [34] in broilers; whereas the lowest observed concentration of DON in pigs was 1.7 mg/kg of feed [53]. Moreover, co-occurrence of various mycotoxins has also been reported to reduce feed intake. With the concentrations of each individual mycotoxin respectively in accordance with legislated levels (Table 1), combinations of DON (5 mg/kg of feed) + FUM (20 mg/kg of feed) + ZEN (5 mg/kg of feed) was reported to result in reduced feed intake in broiler chickens, which was more pronounced than DON (5 mg/kg feed) alone [49]. Synergistic effects of combined DON (1 mg/kg) + ZEN (0.27 mg/kg) on feed intake were also reported in piglets compared with either DON or ZEN alone [52]. Furthermore, a large number of reviewed articles used either artificially contaminated experiential diets or purified powder rather than naturally contaminated diets (Table 2).

Table 2 Effects of selected major mycotoxins on feed intake

The reduced feed intake has been reportedly associated with feeding behavior changes in animals, such as reduced meal frequency and size, slower feeding rate [55, 72,73,74]. Different mechanisms of feed refusal may be involved. For example, the increased anorexia responses induced by TRC especially DON has been well documented mostly in mouse/mink model [75], but has been confirmed with pig models. DON can reduce feed intake by direct regulation of anorexigenic pathways in central nervous system after crossing blood-brain barrier in pigs [75, 76] or indirect peripheral regulations such as stimulating secretion of gut-satiety hormones such as peptide YY and cholecystokinin in pigs [56, 77, 78]. Other mechanisms such as mycotoxin-induced gut dysbiosis [79] and secretion of inflammatory mediators [80] could also contribute to behavior changes and feed refusal in animals.

Although mycotoxin-induced anorexia has been evidenced, especially for DON, several studies have reported progressive disappearance of negative effects on feed intake over time in pigs either continuously on DON-contaminated diets [54] or on a normal DON-free diets after initial exposure to DON in the experiments [81], with the depression in feed intake being most severe usually within 2 weeks after exposure [54, 55, 58, 77, 81]. The former scenario suggested that animals appeared to show tolerance to the presence of DON in the diet [54, 55, 58]. Although such adaptive mechanisms of pigs have not been fully understood, this could be linked to the possible alteration in intestinal microbes to favor detoxification of DON in pigs. However, piglets fed with diets contaminated with combined low-concentration DON (1 mg/kg feed) and ZEN (0.27 mg/kg feed) were reported to fail to recover after the mixture was withdrawn from the diets [52]. High concentration of FUM (58 mg/kg feed) also resulted in the failure of recovery in pigs compared to control group on mycotoxin-free diet [60].

The feed intake of ruminants has not been observed to be affected by DON and ZEN [68,69,70], whereas it was reportedly reduced by the mixture of AFB1, OTA and ZEN in lactating goats [47].

Nutrient digestibility

Reduced ability of food animals to efficiently utilize their feed has been documented (Table 3). Several reviewed articles reported decreased digestibility of dry matter, gross energy and/or metabolizable energy, crude protein/amino acids, crude fat of animals after exposure to either AF, DON or FUM alone as well as combination of multiple Fusarium mycotoxins in poultry and pigs (Table 3). The reduced digestibility of neutral detergent fiber was also reported after lactating cows were exposed to Fusarium mycotoxins below the EU maximum levels [68]. Previous studies indicated that lower levels of FUM (5.5 mg/kg feed) [59] in broiler chickens and ZEN (1 mg/kg feed) in pigs [64], respectively, had no effects on digestibility of dry matter, crude protein and gross energy. Most of concentrations that negatively affected nutrient digestibility were above legislated maximum levels (Tables 1 and 3). However, several studies respectively reported that reduced nutrient digestibility in poultry, pigs and dairy cows was observed after animals were exposed to DON or FUM alone [85] or combinations of Fusarium mycotoxins [59, 68, 85] at concentrations in compliance with the regulations (Table 1). There is variability of results reported regarding the effects of different mycotoxins on nutrient digestibility, which could be due to the factors outlined in Table 4.

Table 3 Effects of selected major mycotoxins on nutrient digestibility
Table 4 Factors contributing to viability of results in the literature

Digestive and absorptive processes

Nutrient digestion and absorption are vital for energy maintenance and nutrient homeostasis and require a network of biochemical reactions and biomolecules in various tissues to ensure requirements for productive purposes are met. The intestine is the renowned site for nutrient digestion and absorption. Distinct finger-like projections in the small intestine called villi increase the absorptive surface area by extending into the lumen. The structure and functionality of the intestine epithelium is maintained by continuous renewal and differentiation of intestinal epithelial cells arising from crypt stem cells [91]. As chyme or digesta passage through the small intestine, brush border (BB) enzymes including oligopeptidases, lipase and oligosaccharidases, are responsible for the final stage of luminal nutrient digestion prior to nutrient absorption. These enzymes can also further hydrolyze the fraction of undigested nutrient oligomers following buccal, gastric, and pancreatic digestion [92]. Following digestion, uptake and absorption of glucose,amino acids and fats is facilitated by several transporter proteins located within brush border membranes [93, 94]. Although, the primary site of nutrient absorption takes place in the small intestine [95], the contribution of the pancreas should not be overlooked. The pancreas can synthesize digestive enzymes such as α-amylase, lipase and proteolytic enzymes that empty into the duodenum; these enzymes respectively digest starches, fats, and proteins [96].

Morphology of the pancreas and the intestine

The integrity of tissues is crucial to their functionality. Despite the importance of pancreas, the effects of mycotoxins on pancreatic function are limited. AFB1 reportedly alters morphology of pancreas in poultry [97, 98], and increased the relative weight of pancreas in ducks and broiler chickens [99, 100]. However, Matur et al. [101] did not observe any effects of AFB1 on relative pancreas weight in breeder hens, but the authors suggested that was due to the low concentration of AFB1 that was used in their study.

Several studies have demonstrated that mycotoxins altered the intestinal morphology. The intestinal villus height and crypt depth, and sometimes the ratio of villus height to crypt depth (H/D), are used as morphological indicators of the likely digestive and absorptive capacity of the small intestine; an increase in H/D ratio corresponds to an increase in digestion and absorption [102]. AFB1 at concentrations of 0.15 mg/kg, 1.2 mg/kg and 2 mg/kg feed reportedly reduced villus height, increased the crypt depth in ducks [41], laying hens [84] and broiler chickens [103], respectively. Ochratoxin A at low level of 0.05 mg of OTA/kg body weight/day also decreased H/D ratio in broiler chickens [104]. The alteration in morphology of intestine has also been observed in broiler chickens [49] and turkeys [105] after birds exposed to 5 mg DON/kg feed and 4.5 mg DON/kg feed, respectively; both concentrations were in accordance with legislated levels (Table 1). In terms of pigs, it has been previously reported that DON above 3 mg/kg feed could result in shorter villi [54, 57, 106]. However, this concentration range of DON was all above permitted levels regulated by the legislative bodies (Table 1). Data on effects of FUM and ZEN on the intestine is limited. Previous study indicated that co-occurrence of FUM (6 mg/kg feed) + DON (3 mg/kg feed) rather than FUM (6 mg/kg feed) alone affected villus height or crypt depth, suggesting synergistic effects of these two mycotoxins [89]. Metayer et al. [49] reported that FUM (20 mg/kg) and ZEN (0.5 mg/kg) within EU regulated levels (Table 1) increased the crypt depth in broiler chickens and the mixture of DON (5 mg/kg) + FUM (20 mg/kg) and ZEN (0.5 mg/kg) also altered intestinal morphology in broiler chickens.

The intestinal epithelium is the first site of exposure following mycotoxin ingestion and may be exposed to higher concentrations than other tissues [17]. Changes in villus height reflect the balance between intestinal epithelial cells (IEC) proliferation and apoptosis [89, 107], which is in line with findings that several mycotoxins can cause oxidative stress induced IEC apoptosis and cell cycle arrest both in vivo and in vitro [108,109,110]. Previous studies suggested effects of mycotoxins on the intestinal morphology may appear to be section-specific, with effects on duodenum and jejunum being more pronounced than ileum [49, 57, 89, 111]. This could be due to the majority of the ingested mycotoxins being absorbed in the upper part of the intestine [17].

Digestive enzymes

Different studies have reported an increase in activity of pancreatic α-amylase, lipase, trypsin, and chymotrypsin across several poultry species such as broilers, breeder hens and ducks after exposure to different AFB1 levels [42, 101, 112]. These authors suggested that the increased activities of α-amylase and lipase were abnormal and pathologic, which may be ascribed to increased pro-enzyme released from the injured pancreas [42, 101, 112]. The results from previous study also suggested that effects of AF on pancreatic enzymes could be concentration- and time-dependent [112]. For example, the activity of α-amylase was elevated as the level of AF increased after 2 weeks animals were on the test diets [112]. Moreover, AF at 2.8 mg/kg feed started to induce increased activity of α-amylase and lipase on d 14; whereas trypsin activity was not affected until d 35 in the experiment [112].

Only limited studies have analyzed the activity of pancreatic digestive enzymes that has been secreted into small intestine, however, contradictory effects were reported. For example, Han et al. [99] reported increased enzyme activities of lipase and α-amylase in the duodenum in ducks exposed to 0.04 mg AFB1/kg feed. Increased level of proenzymes released from injured pancreas could possibly account for this finding [99]; the increased activities may also be due to a compensatory effect of the birds to meet their nutrient needs in response to reduced feed intake [42]. In contrast, Matur et al. [101] observed decreased activities of decreased α-amylase and lipase in breeder hens fed diet containing 0.1 mg AF/kg feed [101]. The authors suggested there could be a secretion problem into duodenum from damaged pancreas [101].

With regards to BB digestive enzymes, Applegate et al. [84] reported that activity of intestinal maltase exhibited hermetic response pattern. Specifically, the activity was increased in laying hens by feeding up to 1.2 mg/kg of purified AF and declined at 2.5 mg/kg [84]. These findings could be explained by the phenomenon called “hormesis”, which is an adaptive beneficial effects that occurs in cells or living organisms after exposure a low concentration of a chemical agent or environmental factor that is damaging at higher concentrations [113]. It is considered an adaptive compensatory process following an initial disruption in homeostasis [113], and its detection is highly dependent on experimental design [114]. Biphasic effects of AFB1 on body weight [115] and immune response [116] in chickens has been reported. Several other studies consistently reported no effects of AFB1 on sucrase and maltase activities in poultry [41, 42, 83]. Moreover, both DON (1 mg/kg feed) and ZEN (1.04 mg/kg feed) were respectively reported to decrease the activities of sucrase, maltase and lactase in pig intestine [65, 117]. The results from previous study could indicate that DON selectively affect enzyme activities in different sections of the intestine, implicating effects of DON on BB enzymes could be segment-dependent in the intestine [117].

Nutrient uptake

Dietary nutrients can only be utilized by different animal tissues after they have been transported across the intestinal epithelium and entered systemic circulation. Different studies have been carried out to investigate the effects of mycotoxins on glucose and amino acid transport across the small intestine using short-circuit current (Isc) measurement, which is a measure of ion transmembrane flux [118], and is a good indicator of sodium-dependent glucose and/or amino acid transport [17]. Awad et al. [119] reported that the Isc induced by the addition of glucose was reduced in broiler chickens fed with a DON-contaminated diet containing 10 mg/kg, suggesting disrupted glucose uptake induced by DON. Similar inhibitory effects of DON on glucose-induced Isc were also observed in laying hens in other studies [120, 121]. This inhibitory effects of DON on Isc could be attributed to its strong inhibition on sodium-dependent glucose co-transporter (SGLT1) [121], which could be supported by the reduced mRNA expression of SGLT1 and the facilitated glucose transporter GLUT2 observed in broiler chickens fed diets naturally contaminated with 1 mg/kg and 5 mg DON/kg feed later in another study [50]. Recently, downregulation of mRNA expression of facilitated glucose transporter GLUT1 and several amino acid transporter including peptide transporter (PepT1) and Heavy chain corresponding to the b0,+ transport system (rBAT) was reported in broiler chickens after exposure to 4-10 mg DON/kg feed [111, 122]. In pigs exposed to 2.86 mg DON/kg, mRNA expression of PepT1 and SLUT1 was not reportedly affected [54].

Aflatoxins also appeared to affect glucose and amino acid transport across the small intestine. Exposure to 1.5 mg/kg AFB1 had no effect on mRNA expression of SGLT1 and GLUT2 in broiler fed diets artificially contaminated with AFB1; however, an increase in mRNA expression of several amino acid transporters including b0,+AT, EAAT3, PepT1, rBAT, yLAT1, and yLAT2 was observed after exposure to AFB1, which might be a compensatory response for amino acid deficiency and impaired protein activities; this may also suggest an increased requirement for amino acid absorption for the subsequent protein synthesis [83].

These results suggested that inhibitory effects of DON and AFB1 on mRNA expression of glucose and animal acid transporters could contribute to the adverse nutritional impact on food animals. However, more relevant studies need to be carried out to better understand mycotoxin-induced nutrient uptake disruption.

Nutrient metabolism by gut microbiota

The intestine, especially large intestine, is a complex ecosystem comprised of trillions of microbes. These microbes play multifaceted role in maintaining health, including providing nutrients, metabolizing complex food sources and toxins, and facilitating normal development of neonatal intestinal immune function and its maintenance throughout life [123,124,125]. Microbiota residing in the large intestine are mainly responsible for the digestion of dietary substrates that escaped proximal digestion in the gastrointestinal tract; such digestion also provides nutrients and energy sources like short chain fatty acids, essential amino acids and vitamins to the host [126, 127]. Recently, a study conducted by Wu et al. [111] using 16S rRNA gene amplicon sequencing demonstrated that exposing broiler chickens to DON at 10 mg/kg significantly decreased the abundance of caecal microbiota, namely the Proteobacteria (phylum level), Escherichia and Cc-115 (genus level), as well as the Escherichia coli (species level) that are beneficial to nutrient utilization processes such as digestion and absorption of protein, lipid and carbohydrates. Contamination of DON also reportedly tended to reduce other microbes including Lactobacillus and Prevotella (genus level), Ruminococcus bromii, Desulfovibrio, C21_c20, and Eubacterium dolichum (species level).

Nutritionally similar to intestinal microbes, the ruminal microbiota also has the capacity to convert recalcitrant fibrous plant material into assimilable energy and nutrients for ruminant species, and also contributes to rumen epithelium development and establishment of the immune system [128]. Different studies have been conducted in vitro with rumen fluid to evaluate the nutrient metabolism in rumen. Boguhn et al. [129] reported DON at 5 mg/kg in the diet failed to alter the ability of rumen microbes to ferment organic matter and carry out protein synthesis. In contrast, another study using rumen fluid containing DON at 40 mg/kg reported a reduction in gas production, ammonia-N and volatile fatty acids (VFA) concentrations [130]. AFB1 in rumen fluid at various concentrations has also been reported to reduce digestibility of DM, gas production and concentrations of ammonia-N and VFA [131,132,133].

Taken together, data suggest that mycotoxins can negatively impact feed intake, nutrient digestion, and absorption, thus making nutrients less available for animals to utilize for productive purposes (Fig 1).

Fig. 1
figure 1

Schematic diagram depicting physiological impact of mycotoxins on different organs affecting nutrient utilization

Differences in susceptibility to mycotoxin exposure

Mycotoxin-related impacts reported in the literature vary greatly. This could be ascribed to several factors that has been summarized in Table 4. Among which, species, age and sex of animals could be major contributing factors accounting for differential susceptibility of animals to mycotoxin exposure from the following aspects.

Microbial transformation of mycotoxins in the gastrointestinal tract

Microbial metabolism of mycotoxins could affect bioaccessibility of parent mycotoxins in animals of different species. Maresca et al. [134] proposed that localization of resident microbiota in the gastrointestinal tract play a role in resistance of animals to mycotoxins. This author categorized animals into two groups: 1) animals having large numbers of microbes located before and after the small intestine (polygastric animals such as ruminants that have microbes in the rumen and in their colon, and birds that have microbes in their crop and cecum), and 2) monogastric species including humans, pigs and rodents that have a large number of microbes located only after the small intestine. On this basis, ruminants are more resistant to mycotoxins compared to monogastric animals in part because most mycotoxins are transformed to less or non-toxic derivatives by microbes in the rumen [31, 32]. This could be exemplified by the transformation of DON and OTA by rumen microbes into their less toxic derivatives deepoxy-deoxynivalenol (DOM-1) and OTA-α, respectively [135]. Resistance of poultry to DON is another case in point. Poultry species are thought to be less susceptible to DON than swine [118, 134] because microbes in the crop transform DON to its less toxic derivative DOM-1 and deepoxy-DON-3-sulfate (DOM-3-sulfate) [134, 136]; this reduces the amount of DON reaching the lower gut. Absence of pre-intestinal microbial transformation in pigs means a larger amount of DON reaches the lower porcine gut.

In addition to the localization of microbiota, the microbial diversity could also be a contributing factor to age-dependent mycotoxicity. The effects of DON and FB1 have been reported to be more pronounced in young pigs [137], which lack microbial diversity. Indeed, the microbial complexity is thought to increase as animals age [138, 139].

Mycotoxin metabolism in animals

The mechanisms by which mycotoxins are metabolized by animals following absorption across the intestinal epithelium could also contribute to species-specific sensitivity to mycotoxins. Detoxification processes that biotransform mycotoxins involve phase I and phase II metabolizing enzymes present both in the intestine and the liver [134, 140]. These biotransformation pathways vary across species, in part, due to species-specific types and activities of metabolizing enzymes [118, 141, 142]. The domestic turkey for example, is one of the most susceptible farm animals to AFB1, and this susceptibility is mainly associated with a combination of efficient hepatic bioactivation of AFB1 to its highly toxic metabolite exo-AFB1-8,9-epoxide (AFBO) by cytochromes P450 (CYP) enzymes, members of the phase I metabolizing enzymes and subsequent deficient inactivation and detoxification of AFBO through conjugation mediated by the phase II enzymes hepatic glutathione–S transferases (GST). In contrast, pigs are more resistant to AFB1 toxicity in comparison to turkeys due to their efficient conjugation of AFBO by GST and subsequent excretion of these AFBO conjugates [140, 141]. Interestingly, in the context of ZEN, pigs are more susceptible to ZEN compared with poultry due to greater production of the more toxic α-ZOL metabolite produced by CYP and rather low expression of UDP-glucuronosyltransferases (UGT) phase II metabolizing enzymes that are responsible for subsequent conjugation of α-ZOL and its inactivation [143].

The differences in mycotoxin metabolizing mechanisms could also explain age-dependent difference in mycotoxin toxicity. Since the effect of mycotoxins on growth was reported to be greater in younger animals of pigs and poultry compared to older animals [144, 145], it is possible that differences in detoxification activity and quantity of hepatic enzymes could contribute to the age-dependent differences in sensitivity to mycotoxins. In support of this, it has been reported that activity of CYP enzymes that are responsible for metabolizing AFB1 is inversely related to age with regards to young poults and chickens [140]. Also, in rats it has been reported that lower levels of AF–glutathione transferase conjugate can be detected in young animals compared to their adult counterparts, suggesting lower capacity for detoxification [145]. Collectively, these studies suggest the liver of young animals is less efficient at biotransforming mycotoxins for elimination, which could contribute to higher susceptibility of young animals to mycotoxicosis. The activities of metabolizing enzymes could also be sex dependent. Studies showed that there were sex-dependent differences in the activities of certain CYP enzymes in human liver and mice [146,147,148,149], which suggests animals of different sexes used in the experiments could lead to varying response to mycotoxin exposure.

Other factors

Other factors may also contribute to age- and species-specific susceptibility to mycotoxins. Variation in the number and affinity of oestrogen receptors for ZEN may affect susceptibility to this mycotoxin. For example, pigs and sheep are the highly susceptible species, and immature animals are generally considered to be more susceptible than adults [14]. Different transport mechanisms and cellular uptake within renal tissue has also been demonstrated to affect OTA nephrotoxicity [14]. Also, gastrointestinal transit time can influence exposure duration of the gut to unabsorbed or poorly absorbed mycotoxins such as FB1 [17, 32]. Induction of oxidative stress by mycotoxins have been shown to be implicated in their toxicities [150, 151]. Upon ingestion of mycotoxin, the ability of animals to alleviate oxidative stress may affect their susceptibility to mycotoxin toxicity. Antioxidant capacity has been reported to vary depending on age and sex [152,153,154], which could also be a contributing factor to possible variations resulting from different ages and sexes used in the studies.

Mycotoxin risk management

Prevention strategies

Mycotoxin contamination can occur at any stage of feed supply chain including crop cultivation, harvest, storage and distribution of crops and compound feeds. Preventative measures should be taken to minimize mycotoxin contamination. Predicting the risk of mycotoxin contamination pre-harvest in cereal crops is a useful and effective tool for pre-harvest and post-harvest mycotoxin management [155]. These predictive models usually use an empirical or mechanistic approach to quantify mycotoxins, and some of them have been implemented in agricultural sectors across Europe to support food source decision making for farmers [156]. Recently, a machine learning approach has been incorporated to build mycotoxin prediction models [156, 157], which could be a promising contribution to mycotoxin control.

Good agricultural practices are one of the primary pre-harvest strategies for mycotoxin prevention are mainly performed at the level of crop cultivation. These practices could include implementing breeding programs for selecting more mycotoxin-resistant plants, crop rotation, soil, and irrigation management, use of registered fungicides and insecticides for control of mild and insect infestations [21]. Application of non-aflatoxigenic Aspergillus flavus has been accelerated as biological control to mitigate pre-harvest aflatoxin contamination over the past years [158]. Post-harvest storage management is also crucial to counteract mycotoxin contamination. Techniques, such as maintaining low moisture levels (less than 15%) and low temperature in the storage environment as well as preserving the integrity of whole grains, are crucial to controlling the level of fungi and mycotoxin contaminants [159].

Decontamination strategies

When prevention is not achievable, it is important to apply strategies to mitigate mycotoxin contamination of feed ingredients or compound feeds. Mitigating mycotoxins should be carried out at an integrated level throughout the supply chain to ensure mycotoxin concentrations in feed are compliant with legislated maximum tolerated/recommended tolerance levels, and therefore feed products are considered safe for animal consumption. Detection of mycotoxins in the cereals and animal feeds using different technologies is critical for monitoring mycotoxin occurrence and mitigation [160]. In terms of efficiency and feasibility, mitigation approaches should include mycotoxin removal and inactivation strategies that do not lead to the production of toxic residues or jeopardize the nutritive value and other desirable parameters, such as palatability, of products [161]. It should also be noted that mitigation needs to be simple and inexpensive to perform. Different remediation strategies that have been deployed in an attempt to mitigate the risk of mycotoxin contamination in feed and their effectiveness, as well as limitations, have been assessed below.

Physical techniques

Dilution

Dilution involves mixing mycotoxin-contaminated and uncontaminated grains to achieve a total mixture containing mycotoxin concentrations below the legislated maximum tolerated levels/recommended tolerance levels [4, 162]. Dilution is a simple and widely used economical approach for mitigating the risk of mycotoxin contamination in feed. However, success of this approach will depend on the degree of the contamination and the availability of uncontaminated grain sources. In some countries, such as European Union, this practice is no longer permitted [4, 162].

Grain cleaning and sorting

Certain steps of grain processing contribute to the decontamination of mycotoxins. Unprocessed cereal grains are normally received in bulk and often contain undesired materials such as dust, foreign materials and interior kernels [163, 164]. Broken and damaged kernels in these bulk loads usually contain most of the mycotoxin contaminants [164]. In large-scale feed manufacturing, the grain cleaning and sorting is applied to mechanically remove dust, foreign materials and interior kernels from healthy grains, mainly based on the lower density of potentially infected grains and feed contaminants [162, 163]. This cleaning and sorting practice have also been shown to reduce contamination of DON and some other mycotoxins such as nivalenol, T-2 and HT-2 in wheat and wheat cultivars [165, 166], as well as the contamination of AF [164, 167]. However, the reduction of mycotoxins by cleaning processes could be highly variable [168].

Milling is another grain processing step that could potentially mitigate mycotoxin contamination. The by-products derived from milling are used as raw ingredients in animal feed. Milling processes have been reported to redirect existing mycotoxins into different milled fractions rather than reduce mycotoxin contamination [162, 169, 170]. Fractions derived from outer layer of kernels tend to have higher mycotoxin concentrations than inner parts through milling processes, since outer parts of grains are more easily contaminated with mycotoxins [165, 166], therefore, depending on the milled fractions, milling could also result in reduced mycotoxin concentrations.

Thermal methods

Although most mycotoxins are generally thermally stable compounds, processes such as crumbling, pelleting and extrusion during feed manufacturing, combining high-speed shearing and superheated steaming, can reduce mycotoxin concentration, but these processes do not completely eliminate mycotoxins [163, 171]. These processes have been shown to reduce concentrations of AF, FUM, DON and ZEN [159, 162]. The degree of reduction depends mainly on several factors, including types of mycotoxins, initial mycotoxin concentration, exposure temperature and duration at high temperature, degree of heat penetration, moisture content and pH, among others [171,172,173]. In general, temperatures higher than 150 °C, long exposure time, high moisture content and low initial mycotoxin concentration all result in greater reduction in mycotoxin concentration [159, 171]. However, with many influencing factors involved, the effects of thermal processing on mycotoxin reduction can be quite variable [162, 163]. It may contribute to mycotoxin mitigation, but it alone is not sufficient for mitigation of exposure risk.

Chemical techniques

Chemical agents

A wide variety of chemical agents have been found to be effective to reduce the concentration of several mycotoxins in different commodities, including acids, bases, chlorinated substances, sodium metabisulfite, ammonia and dry ozone. However, toxic metabolites can be generated by chemical treatment and the nutritional value, and the palatability of the feed can be diminished. Handling of these chemicals also poses a potential risk to workers [30, 174]. The application of chemical agents for decontamination is currently not authorized within the EU and US [30, 162].

Irradiation

For many stored cereals, irradiation is used as an approach to reduce or eliminate fungi and other potential pathogens infecting the grains, and it can partially eliminate mycotoxins [21, 164]. Three sources of ionizing radiation are authorized in food or feed manufacturing in Europe, including gamma-radiation, X-rays, and electron beams [162]. It was reported that gamma-irradiation reduced AFB1 content in maize and chicken feed, respectively, and an increase in irradiation dose showed better reducing effect [175, 176]. There are, however, some concerns about use of irradiation, including public concern about the safety of ionizing irradiation, changes in nutritional value and added food processing cost [162, 177].

Mycotoxin-detoxifying agents as feed additives

Depending on the mode of actions, mycotoxin-detoxifying agents (MDA) can be categorized into biotransforming agents (BA) and adsorbing agents (AA) according to a scientific reported submitted to ESFA [178]. The BA, such as bacteria/fungi or enzymes, degrade mycotoxins into less toxic metabolites, whereas the AA migrate mycotoxin contamination by decreasing mycotoxin bioavailability, which leads to a reduction in mycotoxin uptake and distribution to the blood and target organs.

Mycotoxin biotransforming agents

Mycotoxin biotransforming agents include microorganisms or their enzymes, which biologically transform mycotoxins into non- or less-toxic metabolites via routes such as hydrolysis, de-epoxidation, acetylation, oxidation, ring/side chain cleavage, and glycosylation by acting molecular signatures on each mycotoxin that confer their toxic effects [30, 179]. Mycotoxin biotransforming microorganisms have been isolated from the environment (i.e., soil, cereal grains and insects) and resident gastrointestinal microbes from animals [174]. Their detoxifying capabilities have been reported to be effective for different mycotoxins such as AFB1, ZEN, OTA and FB1 [180, 181]. The Eubacterium BBSH 797 bacterial strain, originally isolated from bovine rumen fluid was the first microbe used as an MDA. This MDA is able to detoxify TRC and is now used commercially for mitigation [30, 182]. Results of animal trials have shown that Eubacterium BBSH 797 significantly reduce the adverse effects of DON on sows and dairy cows, and T-2 toxin on growing broiler chickens [179]. Trichosporon mycotoxinivorans is a yeast strain that has also been thoroughly investigated and it is also commercially used as an MDA. This yeast, isolated from the hind-gut of the termite Mastotermes darwiniensis can detoxify both ZEN and OTA [179, 183]. Application of enzymes that can biologically degrade mycotoxins is an attractive alternative to microorganisms as they catalyze chemical reactions in a highly specific and efficient manner and offer advantages in terms of safety and ease of handing compared to viable microorganisms but are restricted to a specific mycotoxin substrate [180]. Several enzymes being identified and reported to have the capacity to biologically degrade FUM, AFB1, OTA, ZEN and DON [180, 181]. An ESFA-approved enzyme-based feed additive has been used commercially, and shows efficient degradation of fumonisins in pigs and avian species [184, 185]

Certain criteria that have to be fulfilled for the effective use of microorganisms and enzymes make their industrial application quite complicated and limited. For example: 1) isolated microbial strains should be non-pathogenic; 2) the detoxification process should yield non- or less-toxic compounds compared to the parent mycotoxin, and the efficacy of these MDA including BA needs be assessed and proved using specific biomarkers for certain mycotoxin [184]; 3) the degradation processes should occur rapidly, and the BA should be able to survive, adapt and be stable under different oxygen conditions and pH levels in the complex environment of the gastrointestinal tract; 4) the organoleptic and nutritive properties of the feed should be preserved; 5) lastly, viability of isolated microorganisms, and detoxification activity of microbes and enzymes, should be maintained through feed processing methods and be stable in the final commercial products [180, 186]. Several other time-consuming and complex processes may also limit the industrial application of enzymes. These include degrading enzyme identification and characterization, detoxifying-enzyme isolation as well as the involvement of molecular engineering and structure-function modifications of native enzymes by targeted or random mutagenesis, since native enzymes usually do not respond to each distinctive requirement of a perfect industrial enzyme [180, 186].

Mycotoxin adsorbing agents

The other class of MDA is mycotoxin adsorbing agents or adsorbents (AA), which help to alleviate the harmful effects of mycotoxins in livestock and poultry through direct binding to mycotoxins; this decreases their bioavailability or reduces their intestinal absorption, promotes the formation of mycotoxin-adsorbent complexes, and their consequent excretion via the fecal route [178].

Mycotoxin adsorbents are divided into inorganic and organic AA. Inorganic AA, also known as mineral adsorbents, are mainly phyllosilicates of the clay mineral group and also include tectosilicates like zeolites and activated charcoal. Inorganic AA are considered first-generation AA [180]. The most significant AA among phyllosilicates group are bentonite, montmorillonite, hydrated sodium aluminosilicates and smectite. The binding capability of inorganic AA depends on the physio-chemical structures of both AA and mycotoxins; this includes the charge distribution of AA and mycotoxins, surface area and pore size of the AA, polarity, and shape of the mycotoxins [159, 187]. Some mineral AA such as bentonites, zeolites and activated charcoal have been reported to adsorb ZEN, OTA, FUM and DON in vitro, however, in vivo confirmation studies are lacking [30, 54]. Most inorganic AA have been recognized as efficient binders of AF, as supported by in vivo studies, but they appear to have very limited capability of binding to other mycotoxins such as ZEN, TRC and FUM [30, 180, 188, 189]. Unfortunately, mineral AA are also known to adsorb micronutrients and have negative effects on the bioavailability of vitamins, amino acids, and minerals in feed [188, 189]. Mineral AA also have ecological disadvantages since the degradation of bound mycotoxins after they have been excreted is relatively slow [162].

In an attempt to overcome this inefficacy of inorganic AA, organic adsorbents originating from cell wall components of microorganisms have been developed as second-generation AA [180]. Cell wall components from Saccharomyces cerevisiae yeast strain are commonly used organic AA. The major functional fractions of yeast cell wall (YCW) responsible for mycotoxin binding include β-D-glucan and mannan oligosaccharides, which bind to mycotoxins via hydrogen bonding and van-der-Waal forces [188, 190, 191]. The proportion of the functional organic AA varies with the microbial strains and processing [192, 193], and differences in product purity and supplemental concentration can lead to different efficacy since their affinity to mycotoxins is reversible and saturable [192]. Compared to its inorganic counterparts, the YCW has exhibited greater capacity of binding to a wider spectrum of mycotoxins such as DON, ZEN, OTA and AFB1, and alleviating negative effects of mycotoxins has been investigated in numerous scientific publication both in vitro [28, 190, 191, 194, 195] and in vivo using poultry, pigs and ruminants [28, 45, 192, 196,197,198,199,200]. Another advantage of the YCW products is that they are biodegradable, and therefore the toxin-binder complexes do not accumulate in the environment after being excreted in the feces [188].

Conclusions and future directions

Feed is the only source of energy and nutrients for farm animals. Sound practice in nutrition is required for animals to achieve their genetically selected production potential. Various mycotoxins and their mixtures can lead to negative nutritional outcomes, which could limit nutrient and energy availability to farm animals, thus, resulting in a sub-optimal production performance. However, it is worth noting that nutritional impact should not be the only criterion for assessment of overall mycotoxicity to food animals as it is well documented mycotoxins could lead to other adverse effects in various organs and systems such as gastrointestinal tract, liver or kidney, as well as the nervous, reproductive and immune systems in food animals, in some cases without affecting growth performance [201].

Moreover, in modern highly intensive and large scaled livestock production, mycotoxin contamination in feeds is only one of the various challenges that food animals face, such as environmental, nutritional non-infectious or infectious stressors [202]. As a result, mycotoxins could pose a cconfounding effect on animal production. For example, it has been reported that exposure to most Fusarium mycotoxins increases animal susceptibility to infectious diseases such as coccidiosis in poultry and swine respiratory diseases [203].

Although various mycotoxin management measures are available, there is still room for improvement given the challenges encountered in mycotoxin management. More effort should be made on assessment of emerging and modified mycotoxin occurrence and their toxicity (toxicokinetic and toxicodynamics), as such data are limited. Moreover, global mycotoxin regulations currently focus on major toxins, and are based on toxicity of individual mycotoxins, and have not considered potential additive or synergistic toxicity of combined mycotoxins. Effort should also be made to develop predictive models capable of predicting contamination from wider spectrum of mycotoxins and including climate change scenarios. Moreover, there is continuous need for development of novel detection and decontamination strategies for effective mycotoxin risk management. Mycotoxin risk management should be an integration of efforts that include risk assessment, establishment of regulatory options, as well as decontamination mitigation methods.

Availability of data and materials

Not applicable.

Abbreviations

AA:

Adsorbing agents

ADE:

Apparent digestible energy

AF:

Aflatoxins

AFB1 :

Aflatoxin B1

AFBO:

Exo-AFB1-8,9-epoxide

BA:

Biotransforming agents

BB:

Brush border

CP:

Crude protein

CYP:

Cytochromes P450 enzymes

DM:

Dry matter

DON:

Deoxynivalenol

EFSA:

European Food Safety Authority

FB1 :

Fumonisin B1

FUM:

Fumonisins

GE :

Gross energy

GST:

Gutathione–S transferases

H/D:

Ratio of villus height to crypt depth

IEC:

Intestinal epithelial cells

Isc:

Short-circuit current

LOAEL:

Lowest-observed-effect-level

MDA:

Mycotoxin-detoxifying agents

NA:

Not applicable

NS:

Not significantly affected

NOEL:

No-observed-effect-level

OM:

Organic matter

OT:

Ochratoxins

OTA:

Ochratoxin A

SGLT1:

Sodium-dependent glucose co-transporter

TRC:

Trichothecenes

UGT:

UDP-glucuronosyltransferases

VFA:

Volatile fatty acids

YCW:

Yeast cell wall

ZEN:

Zearalenone

References

  1. Baumgard LH, Collier RJ, Bauman DE. A 100-Year Review: Regulation of nutrient partitioning to support lactation. J Dairy Sci. 2017;100:10353–66.

    Article  CAS  PubMed  Google Scholar 

  2. Venkatesh N, Keller NP. Mycotoxins in conversation with bacteria and fungi. Front Microbiol. 2019;10:403.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Bai GH, Desjardins AE, Plattner RD. Deoxynivalenol-nonproducing fusarium graminearum causes initial infection, but does not cause disease spread in wheat spikes. Mycopathologia. 2002;153:91–8.

    Article  CAS  PubMed  Google Scholar 

  4. Bryden WL. Mycotoxin contamination of the feed supply chain: Implications for animal productivity and feed security. Anim Feed Sci Technol. 2012;173:134–58.

    Article  CAS  Google Scholar 

  5. Santos Pereira CC, Cunha S, Fernandes JO. Prevalent mycotoxins in animal feed: occurrence and analytical methods. Toxins. 2019;11:290.

    Article  PubMed Central  CAS  Google Scholar 

  6. Wu F. Global impacts of aflatoxin in maize: trade and human health. World Mycotoxin J. 2015;8(2):137–42.

    Article  CAS  Google Scholar 

  7. Alshannaq A, Yu J-H. Occurrence, toxicity, and analysis of major mycotoxins in food. Int J Environ Res Public Health. 2017;14:632.

    Article  PubMed Central  CAS  Google Scholar 

  8. Pinotti L, Ottoboni M, Giromini C, Dell’Orto V, Cheli F. Mycotoxin contamination in the EU feed supply chain: A focus on cereal byproducts. Toxins. 2016;8:45.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Smith M-C, Madec S, Coton E, Hymery N. Natural co-occurrence of mycotoxins in foods and feeds and their in vitro combined toxicological effects. Toxins. 2016;8:94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Andersen B, Phippen C, Frisvad JC, Emery S, Eustace RA. Fungal and chemical diversity in hay and wrapped haylage for equine feed. Mycotoxin Res. 2020;36:159–72.

    Article  CAS  PubMed  Google Scholar 

  11. Penagos-Tabares F, Khiaosa-ard R, Nagl V, Faas J, Jenkins T, Sulyok M, et al. Mycotoxins, phytoestrogens and other secondary metabolites in Austrian pastures: Occurrences, contamination levels and implications of Geo-climatic factors. Toxins. 2021;13:460.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Reisinger N, Schürer-Waldheim S, Mayer E, Debevere S, Antonissen G, Sulyok M, et al. Mycotoxin occurrence in maize silage—A neglected risk for bovine gut health? Toxins. 2019;11:577.

    Article  CAS  PubMed Central  Google Scholar 

  13. Adegbeye MJ, Reddy PRK, Chilaka CA, Balogun OB, Elghandour MMMY, Rivas-Caceres RR, et al. Mycotoxin toxicity and residue in animal products: Prevalence, consumer exposure and reduction strategies – A review. Toxicon. 2020;177:96–108.

    Article  CAS  PubMed  Google Scholar 

  14. Völkel I, Schröer-Merker E, Czerny C-P. The Carry-over of mycotoxins in products of animal origin with special regard to its implications for the European Food Safety Legislation. Food Nutr Sci. 2011;2:852–67.

    Google Scholar 

  15. Palumbo R, Crisci A, Venâncio A, Cortiñas Abrahantes J, Dorne J-L, Battilani P, et al. Occurrence and co-occurrence of mycotoxins in cereal-based feed and food. Microorganisms. 2020;8:74.

    Article  PubMed Central  Google Scholar 

  16. Weaver AC, Weaver DM, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Grenier B, Applegate T. Modulation of intestinal functions following mycotoxin ingestion: Meta-analysis of published experiments in animals. Toxins. 2013;5:396–430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Battilani P, Toscano P, Van der Fels-Klerx HJ, Moretti A, Camardo Leggieri M, Brera C, et al. Aflatoxin B1 contamination in maize in Europe increases due to climate change. Sci Rep. 2016;6:24328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kebede H, Abbas HK, Fisher DK, Bellaloui N. Relationship between Aflatoxin contamination and physiological responses of corn plants under drought and heat stress. Toxins. 2012;4:1385–403.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Gruber-Dorninger C, Jenkins T, Schatzmayr G. Global mycotoxin occurrence in feed: A Ten-Year survey. Toxins. 2019;11:375.

    Article  CAS  PubMed Central  Google Scholar 

  21. Agriopoulou S, Stamatelopoulou E, Varzakas T. Advances in occurrence, importance, and mycotoxin control strategies: Prevention and detoxification in foods. Foods. 2020;9:137.

    Article  CAS  PubMed Central  Google Scholar 

  22. Pestka JJ. Deoxynivalenol: Mechanisms of action, human exposure, and toxicological relevance. Arch Toxicol. 2010;84:663–79.

    Article  CAS  PubMed  Google Scholar 

  23. Rocha O, Ansari K, Doohan FM. Effects of trichothecene mycotoxins on eukaryotic cells: A review. Food Addit Contam. 2005;22:369–78.

    Article  CAS  PubMed  Google Scholar 

  24. Riley RT, Merrill AH. Ceramide synthase inhibition by fumonisins: a perfect storm of perturbed sphingolipid metabolism, signaling, and disease. J Lipid Res. 2019;60:1183–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li L, Chen W, Li H, Iqbal J, Zhu Y, Wu T, et al. Rapid determination of fumonisin (FB1) by syringe SPE coupled with solid-phase fluorescence spectrometry. Spectrochim Acta A Mol Biomol Spectroscopy. 2020;226:117549.

    Article  CAS  Google Scholar 

  26. Mannaa M, Kim KD. Influence of temperature and water activity on deleterious fungi and mycotoxin production during grain storage. Mycobiology. 2017;45:240–54.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Mally A. Ochratoxin A and mitotic disruption: Mode of action analysis of renal tumor formation by ochratoxin A. Toxicol Sci. 2012;127:315–30.

    Article  CAS  PubMed  Google Scholar 

  28. Pfohl-Leszkowicz A, Hadjeba-Medjdoub K, Ballet N, Schrickx J, Fink-Gremmels J. Assessment and characterisation of yeast-based products intended to mitigate ochratoxin exposure using in vitro and in vivo models. Food Addit Contam Part Chem Anal Control Expo Risk Assess. 2015;32:604–16.

    Article  CAS  Google Scholar 

  29. Sorrenti V, Di Giacomo C, Acquaviva R, Barbagallo I, Bognanno M, Galvano F. Toxicity of ochratoxin A and its modulation by antioxidants: A review. Toxins. 2013;5:1742–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Vila-Donat P, Marín S, Sanchis V, Ramos AJ. A review of the mycotoxin adsorbing agents, with an emphasis on their multi-binding capacity, for animal feed decontamination. Food Chem Toxicol. 2018;114:246–59.

    Article  CAS  PubMed  Google Scholar 

  31. Fink-Gremmels J. The role of mycotoxins in the health and performance of dairy cows. Vet J. 2008;176:84–92.

    Article  CAS  PubMed  Google Scholar 

  32. Gallo A, Giuberti G, Frisvad JC, Bertuzzi T, Nielsen KF. Review on mycotoxin issues in ruminants: Occurrence in forages, effects of mycotoxin ingestion on health status and animal performance and practical strategies to counteract their negative effects. Toxins. 2015;7:3057–111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Huber K. Invited review: Resource allocation mismatch as pathway to disproportionate growth in farm animals – prerequisite for a disturbed health. Animal. 2018;12:528–36.

    Article  CAS  PubMed  Google Scholar 

  34. Liu N, Ding K, Wang J, Deng Q, Gu K, Wang J. Effects of lactic acid bacteria and smectite after aflatoxin B1 challenge on the growth performance, nutrient digestibility and blood parameters of broilers. J Anim Physiol Anim Nutr. 2018;102:953–61.

    Article  CAS  Google Scholar 

  35. Rashidi N, Khatibjoo A, Taherpour K, Mohammad A-G, Shirzadi H. Effects of licorice extract, probiotic, toxin binder and poultry litter biochar on performance, immune function, blood indices and liver histopathology of broilers exposed to aflatoxin-B1. Poult Sci. 2020;99:5896–906.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tavangar P, Gharahveysi S, Rezaeipour V, Irani M. Efficacy of phytobiotic and toxin binder feed additives individually or in combination on the growth performance, blood biochemical parameters, intestinal morphology, and microbial population in broiler chickens exposed to aflatoxin B1. Trop Anim Health Prod. 2021;53:335.

    Article  PubMed  Google Scholar 

  37. Śliżewska K, Cukrowska B, Smulikowska S, Cielecka-Kuszyk J. The effect of probiotic supplementation on performance and the histopathological changes in liver and kidneys in broiler chickens fed diets with aflatoxin B1. Toxins. 2019;11:112.

    Article  PubMed Central  CAS  Google Scholar 

  38. Jia R, Ma Q, Fan Y, Ji C, Zhang J, Liu T, et al. The toxic effects of combined aflatoxins and zearalenone in naturally contaminated diets on laying performance, egg quality and mycotoxins residues in eggs of layers and the protective effect of Bacillus subtilis biodegradation product. Food Chem Toxicol. 2016;90:142–50.

    Article  CAS  PubMed  Google Scholar 

  39. Akande TO, Salami TK, Salako AO. Nutrient stability in mould-infested feed and mitigating effect of dietary supplemental vitamins in brown laying hens. Trop Anim Health Prod. 2020;52:293–9.

    Article  PubMed  Google Scholar 

  40. Wan XL, Yang ZB, Yang WR, Jiang SZ, Zhang GG, Johnston SL, et al. Toxicity of increasing aflatoxin B1 concentrations from contaminated corn with or without clay adsorbent supplementation in ducklings. Poult Sci. 2013;92:1244–53.

    Article  CAS  PubMed  Google Scholar 

  41. Feng GD, He J, Ao X, Chen DW. Effects of maize naturally contaminated with aflatoxin B1 on growth performance, intestinal morphology, and digestive physiology in ducks. Poult Sci. 2017;96:1948–55.

    Article  CAS  PubMed  Google Scholar 

  42. Chen X, Murdoch R, Zhang Q, Shafer DJ, Applegate TJ. Effects of dietary protein concentration on performance and nutrient digestibility in Pekin ducks during aflatoxicosis. Poult Sci. 2016;95:834–41.

    Article  CAS  PubMed  Google Scholar 

  43. Rauber RH, Dilkin P, Giacomini LZ, de Almeida CAA, Mallmann CA. Performance of turkey poults fed different doses of Aflatoxins in the diet. Poult Sci. 2007;86:1620–4.

    Article  CAS  PubMed  Google Scholar 

  44. Diaz GJ, Cortés A, Botero L. Evaluation of the ability of a feed additive to ameliorate the adverse effects of aflatoxins in turkey poults. Br Poult Sci. 2009;50:240–50.

    Article  CAS  PubMed  Google Scholar 

  45. Sun Y, Park I, Guo J, Weaver AC, Kim SW. Impacts of low level aflatoxin in feed and the use of modified yeast cell wall extract on growth and health of nursery pigs. Anim Nutr. 2015;1:177–83.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Pu J, Yuan Q, Yan H, Tian G, Chen D, He J, et al. Effects of chronic exposure to low levels of dietary Aflatoxin B1 on growth performance, apparent total tact digestibility and intestinal health in pigs. Animals. 2021;11:336.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Huang S, Zheng N, Fan C, Cheng M, Wang S, Jabar A, et al. Effects of aflatoxin B1 combined with ochratoxin A and/or zearalenone on metabolism, immune function, and antioxidant status in lactating dairy goats. Asian-Australas J Anim Sci. 2017;31:505–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Peillod C, Laborde M, Travel A, Mika A, Bailly JD, Cleva D, et al. Toxic effects of fumonisins, deoxynivalenol and zearalenone alone and in combination in ducks fed the maximum EU tolerated level. Toxins. 2021;13:152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Metayer J-P, Travel A, Mika A, Bailly J-D, Cleva D, Boissieu C, 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.

    Article  CAS  PubMed Central  Google Scholar 

  50. Awad WA, Vahjen W, Aschenbach JR, Zentek J. A diet naturally contaminated with the Fusarium mycotoxin deoxynivalenol (DON) downregulates gene expression of glucose transporters in the intestine of broiler chickens. Livest Sci. 2011;140:72–9.

    Article  Google Scholar 

  51. Travel A, Metayer J-P, Mika A, Bailly J-D, Cleva D, Boissieu C, et al. Toxicity of fumonisins, deoxynivalenol, and zearalenone alone and in combination in turkeys fed with the maximum European Union–tolerated level. Avian Dis. 2019;63:703–12.

    Article  PubMed  Google Scholar 

  52. Jia R, Liu W, Zhao L, Cao L, Shen Z. Low doses of individual and combined deoxynivalenol and zearalenone in naturally moldy diets impair intestinal functions via inducing inflammation and disrupting epithelial barrier in the intestine of piglets. Toxicol Lett. 2020;333:159–69.

    Article  CAS  PubMed  Google Scholar 

  53. Sayyari A, Framstad T, Krogenæs AK, Sivertsen T. Effects of feeding naturally contaminated deoxynivalenol diets to sows during late gestation and lactation in a high-yield specific pathogen-free herd. Porc Health Manag. 2018;4:26.

    Article  Google Scholar 

  54. Mwaniki AW, Buis QR, Trott D, Huber L-A, Yang C, Kiarie EG. Comparative efficacy of commercially available deoxynivalenol detoxifying feed additives on growth performance, total tract digestibility of components, and physiological responses in nursery pigs fed diets formulated with naturally contaminated corn. Transl Anim Sci. 2021;5:1–13.

    Article  CAS  Google Scholar 

  55. Serviento AM, Brossard L, Renaudeau D. An acute challenge with a deoxynivalenol-contaminated diet has short- and long-term effects on performance and feeding behavior in finishing pigs. J Anim Sci. 2018;96:5209–21.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Jia R, Sadiq FA, Liu W, Cao L, Shen Z. Protective effects of Bacillus subtilis ASAG 216 on growth performance, antioxidant capacity, gut microbiota and tissues residues of weaned piglets fed deoxynivalenol contaminated diets. Food Chem Toxicol. 2021;148:111962.

    Article  CAS  PubMed  Google Scholar 

  57. Van Le Thanh B, Lessard M, Chorfi Y, Guay F. The efficacy of anti-mycotoxin feed additives in preventing the adverse effects of wheat naturally contaminated with Fusarium mycotoxins on performance, intestinal barrier function and nutrient digestibility and retention in weanling pigs. Can J Anim Sci. 2015;95:197–209.

    Article  CAS  Google Scholar 

  58. Wellington MO, Bosompem MA, Petracek R, Nagl V, Columbus DA. Effect of long-term feeding of graded levels of deoxynivalenol (DON) on growth performance, nutrient utilization, and organ health in finishing pigs and DON content in biological samples. J Anim Sci. 2020;98(12):1–9.

    Article  Google Scholar 

  59. Zou Y, Yang ZB, Yang WR, Jiang SZ, Zhang GG, Chi F. Effect of purified zearalenone on nutrient digestibility in broilers fed 2 levels of fumonisin from naturally contaminated corn (Zea mays). J Appl Poult Res. 2012;21:251–8.

    Article  CAS  Google Scholar 

  60. Rao Z-X, Tokach MD, Dritz SS, Woodworth JC, DeRouchey JM, Goodband RD, et al. Efficacy of commercial products on nursery pig growth performance fed diets with fumonisin contaminated corn. Transl Anim Sci. 2020;4:1–12.

    Article  CAS  Google Scholar 

  61. Mazur-Kuśnirek M, Antoszkiewicz Z, Lipiński K, Fijałkowska M, Purwin C, Kotlarczyk S. The effect of polyphenols and vitamin E on the antioxidant status and meat quality of broiler chickens fed diets naturally contaminated with ochratoxin A. Arch Anim Nutr. 2019;73:431–44.

    Article  PubMed  Google Scholar 

  62. Sakthivelan SM, Sudhakar Rao GV. Effect of ochratoxin A on body weight, feed intake and feed conversion in broiler chicken. Vet Med Int. 2010;2010:590432.

  63. Gan F, Hou L, Lin Z, Ge L, Liu D, Li H, et al. Effects of selenium-enriched probiotics on ochratoxin A-induced kidney injury and DNMTs expressions in piglets. Res Vet Sci. 2021;139:94–101.

    Article  CAS  PubMed  Google Scholar 

  64. Jiang SZ, Yang ZB, Yang WR, Wang SJ, Liu FX, Johnston LA, et al. Effect of purified zearalenone with or without modified montmorillonite on nutrient availability, genital organs and serum hormones in post-weaning piglets. Livest Sci. 2012;144:110–8.

  65. Liu X, Xu C, Yang Z, Yang W, Huang L, Wang S, et al. Effects of dietary zearalenone exposure on the growth performance, small intestine disaccharidase, and antioxidant activities of weaned gilts. Animals. 2020;10:2157.

    Article  PubMed Central  Google Scholar 

  66. Su Y, Sun Y, Ju D, Chang S, Shi B, Shan A. The detoxification effect of vitamin C on zearalenone toxicity in piglets. Ecotoxicol Environ Saf. 2018;158:284–92.

    Article  CAS  PubMed  Google Scholar 

  67. Raju MVLN, Rao SVR, Panda AK. Interaction effects of sunflower oil and aflatoxin at graded levels in diet on performance, serum and tissue biochemical profile, organ weights and immuneresponse in broiler chicken. Trop Anim Health Prod. 2021;53:317.

    Article  CAS  PubMed  Google Scholar 

  68. Gallo A, Minuti A, Bani P, Bertuzzi T, Cappelli FP, Doupovec B, et al. A mycotoxin-deactivating feed additive counteracts the adverse effects of regular levels of Fusarium mycotoxins in dairy cows. J Dairy Sci. 2020;103:11314–31.

    Article  CAS  PubMed  Google Scholar 

  69. McKay ZC, Averkieva O, Rajauria G, Pierce KM. The effect of feedborne Fusarium mycotoxins on dry matter intake, milk production and blood metabolites of early lactation dairy cows. Anim Feed Sci Technol. 2019;253:39–44.

    Article  CAS  Google Scholar 

  70. Winkler J, Kersten S, Meyer U, Engelhardt U, Dänicke S. Residues of zearalenone (ZEN), deoxynivalenol (DON) and their metabolites in plasma of dairy cows fed Fusarium contaminated maize and their relationships to performance parameters. Food Chem Toxicol. 2014;65:196–204.

    Article  CAS  PubMed  Google Scholar 

  71. Santos EV, Fontes DO, Benfato MS, Hackenhaar FS, Salomon T, Jacob DV, et al. Mycotoxin deactivator improves performance, antioxidant status, and reduces oxidative stress in nursery pigs fed diets containing mycotoxins. J Anim Sci. 2021;99(8):1–8.

    Google Scholar 

  72. Girardet C, Bonnet MS, Jdir R, Sadoud M, Thirion S, Tardivel C, et al. The food-contaminant deoxynivalenol modifies eating by targeting anorexigenic neurocircuitry. PLoS One. 2011;6(10):e26134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Müller LKF, Silva ASD, Bottari NB, Santurio J, Morsch VM, Piva MM, et al. Effects of fed mycotoxin contaminated diets supplemented with spray-dried porcine plasma on cholinergic response and behavior in piglets. An Acad Bras Ciênc. 2019;91:e20180419.

    Article  PubMed  CAS  Google Scholar 

  74. Wang A, Schwean-Lardner K, Hogan NS. Feed preference and feeding behaviours in grower broilers fed diets containing wheat naturally contaminated with Fusarium mycotoxins. Br Poult Sci. 2019;60:309–16.

    Article  CAS  PubMed  Google Scholar 

  75. Terciolo C, Maresca M, Pinton P, Oswald IP. Review article: Role of satiety hormones in anorexia induction by trichothecene mycotoxins. Food Chem Toxicol. 2018;121:701–14.

    Article  CAS  PubMed  Google Scholar 

  76. Gaigé S, Bonnet MS, Tardivel C, Pinton P, Trouslard J, Jean A, et al. c-Fos immunoreactivity in the pig brain following deoxynivalenol intoxication: Focus on NUCB2/nesfatin-1 expressing neurons. NeuroToxicology. 2013;34:135–49.

    Article  PubMed  CAS  Google Scholar 

  77. Li R, Li Y, Su Y, Shen D, Dai P, Li C. Short-term ingestion of deoxynivalenol in naturally contaminated feed alters piglet performance and gut hormone secretion. Anim Sci J. 2018;89:1134–43.

    Article  CAS  PubMed  Google Scholar 

  78. Zheng W, Ji X, Zhang Q, Du W, Wei Q, Yao W. Hydrogen-Rich water and lactulose protect against growth suppression and oxidative stress in female piglets fed Fusarium toxins contaminated diets. Toxins. 2018;10:228.

    Article  PubMed Central  CAS  Google Scholar 

  79. Peng Z, Chen L, Xiao J, Zhou X, Nüssler AK, Liu L, et al. Review of mechanisms of deoxynivalenol-induced anorexia: The role of gut microbiota. J Appl Toxicol. 2017;37:1021–9.

    Article  CAS  PubMed  Google Scholar 

  80. Lebrun B, Tardivel C, Félix B, Abysique A, Troadec J-D, Gaigé S, et al. Dysregulation of energy balance by trichothecene mycotoxins: Mechanisms and prospects. NeuroToxicology. 2015;49:15–27.

    Article  PubMed  CAS  Google Scholar 

  81. Kong C, Shin SY, Park CS, Kim BG. Effects of feeding barley naturally contaminated with Fusarium mycotoxins on growth performance, nutrient digestibility, and blood chemistry of gilts and growth recoveries by feeding a non-contaminated diet. Asian-Australas J Anim Sci. 2015;28:662–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Verma J, Swain B, Johri T. Effect of various levels of aflatoxin and ochratoxin A and combinations thereof on protein and energy utilisation in broilers. J Sci Food Agric. 2002;82:1412–7.

    Article  CAS  Google Scholar 

  83. Chen X, Naehrer K, Applegate TJ. Interactive effects of dietary protein concentration and aflatoxin B1 on performance, nutrient digestibility, and gut health in broiler chicks. Poult Sci. 2016;95:1312–25.

    Article  CAS  PubMed  Google Scholar 

  84. Applegate TJ, Schatzmayr G, Pricket K, Troche C, Jiang Z. Effect of aflatoxin culture on intestinal function and nutrient loss in laying hens. Poult Sci. 2009;88:1235–41.

    Article  CAS  PubMed  Google Scholar 

  85. Liu JD, Doupovec B, Schatzmayr D, Murugesan GR, Bortoluzzi C, Villegas AM, et al. The impact of deoxynivalenol, fumonisins, and their combination on performance, nutrient, and energy digestibility in broiler chickens. Poult Sci. 2020;99:272–9.

    Article  CAS  PubMed  Google Scholar 

  86. Jo H, Kong C, Song M, Kim BG. Effects of dietary deoxynivalenol and zearalenone on apparent ileal digestibility of amino acids in growing pigs. Anim Feed Sci Technol. 2016;219:77–82.

    Article  CAS  Google Scholar 

  87. Zeebone YY, Kovács M, Bóta B, Halas V. Effects of dietary fumonisins on nutrients digestibility in weanling pigs. Acta Fytotech Zootech. 2020;23:23–8.

    Article  Google Scholar 

  88. Wang JP, Chi, Kim IH. Effects of montmorillonite clay on growth performance, nutrient digestibility, vulva size, faecal microflora, and oxidative stress in weaning gilts challenged with zearalenone. Anim Feed Sci Technol. 2012;178:158–66.

    Article  CAS  Google Scholar 

  89. Bracarense A-PFL, Lucioli J, Grenier B, Pacheco GD, Moll W-D, Schatzmayr G, et al. Chronic ingestion of deoxynivalenol and fumonisin, alone or in interaction, induces morphological and immunological changes in the intestine of piglets. Br J Nutr. 2012;107:1776–86.

    Article  CAS  PubMed  Google Scholar 

  90. Wang D, Lindemann MD, Estienne MJ. Effect of folic acid supplementation and dietary protein level on growth performance, serum chemistry and immune response in weanling piglets fed differing concentrations of aflatoxin. Toxins. 2020;12:651.

    Article  CAS  PubMed Central  Google Scholar 

  91. Mowat AM, Agace WW. Regional specialization within the intestinal immune system. Nat Rev Immunol. 2014;14:667–85.

    Article  CAS  PubMed  Google Scholar 

  92. Hooton D, Lentle R, Monro J, Wickham M, Simpson R. The secretion and action of brush border enzymes in the mammalian small intestine. Rev Physiol Biochem Pharmacol. 2015;168:59–118.

    Article  CAS  PubMed  Google Scholar 

  93. Iqbal J, Hussain MM. Intestinal lipid absorption. Am J Physiol Endocrinol Metab. 2009;296:E1183–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kiela PR, Ghishan FK. Physiology of Intestinal Absorption and Secretion. Best Pract Res Clin Gastroenterol. 2016;30:145–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Duca FA, Waise TMZ, Peppler WT, Lam TKT. The metabolic impact of small intestinal nutrient sensing. Nat Commun. 2021;12:903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Leung PS, Ip SP. Pancreatic acinar cell: Its role in acute pancreatitis. Int J Biochem Cell Biol. 2006;38:1024–30.

    Article  CAS  PubMed  Google Scholar 

  97. Groseva N, Valchev I, Hristov T, Lazarov L, Kanakov D, Marutsova V, et al. Investigatio on pancreas morphology in turkey broilers with experimental aflatoxicosis B. Tradit Mod Vet Med. 2018;3:100–8.

    Google Scholar 

  98. Şimşek N, Ergun L, Ergun E, Alabay B, Essiz D. The effects of experimental aflatoxicosis on the exocrine pancreas in quails (Coturnix coturnix japonica). Arch Toxicol. 2007;81:583–8.

    Article  PubMed  CAS  Google Scholar 

  99. Han X-Y, Huang Q-C, Li W-F, Jiang J-F, Xu Z-R. Changes in growth performance, digestive enzyme activities and nutrient digestibility of cherry valley ducks in response to aflatoxin B1 levels. Livest Sci. 2008;119:216–20.

    Article  Google Scholar 

  100. Raju DMVLN, Rao SVR, Radhika K, Panda AK. Effect of amount and source of supplemental dietary vegetable oil on broiler chickens exposed to aflatoxicosis. Br Poult Sci. 2005;46:587–94.

    Article  CAS  PubMed  Google Scholar 

  101. Matur E, Ergul E, Akyazi I, Eraslan E, Cirakli ZT. The effects of Saccharomyces cerevisiae extract on the weight of some organs, liver, and pancreatic digestive enzyme activity in breeder hens fed diets contaminated with aflatoxins. Poult Sci. 2010;89:2213–20.

    Article  CAS  PubMed  Google Scholar 

  102. Montagne L, Pluske JR, Hampson DJ. A review of interactions between dietary fibre and the intestinal mucosa, and their consequences on digestive health in young non-ruminant animals. Anim Feed Sci Technol. 2003;108:95–117.

    Article  Google Scholar 

  103. Jahanian E, Mahdavi AH, Asgary S, Jahanian R. Effects of dietary inclusion of silymarin on performance, intestinal morphology and ileal bacterial count in aflatoxin-challenged broiler chicks. J Anim Physiol Anim Nutr. 2017;101:e43–54.

    Article  CAS  Google Scholar 

  104. Tong C, Li P, Yu L-H, Li L, Li K, Chen Y, et al. Selenium-rich yeast attenuates ochratoxin A-induced small intestinal injury in broiler chickens by activating the Nrf2 pathway and inhibiting NF-KB activation. J Funct Foods. 2020;66:103784.

    Article  CAS  Google Scholar 

  105. Devreese M, Girgis GN, Tran S-T, De Baere S, De Backer P, Croubels S, et al. The effects of feed-borne Fusarium mycotoxins and glucomannan in turkey poults based on specific and non-specific parameters. Food Chem Toxicol. 2014;63:69–75.

    Article  CAS  PubMed  Google Scholar 

  106. Wu L, Liao P, He L, Ren W, Yin J, Duan J, et al. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Wang X, Zhang Y, Zhao J, Cao L, Zhu L, Huang Y, et al. Deoxynivalenol induces inflammatory injury in IPEC-J2 cells via NF-κB signaling pathway. Toxins. 2019;11(12):733.

  108. Fouad A, Ruan D, El-Senousey H, Chen W, Jiang S, Zheng C. Harmful efects and control strategies of aflatoxin B1 produced by Aspergillus flavus and Aspergillus parasiticus strains on poultry: Review. Toxins. 2019;11:176.

    Article  CAS  PubMed Central  Google Scholar 

  109. Xu R, Karrow NA, Shandilya UK, Sun L-H, Kitazawa H. In-vitro cell culture for efficient assessment of mycotoxin exposure, toxicity and risk mitigation. Toxins. 2020;12:146.

    Article  CAS  PubMed Central  Google Scholar 

  110. Yang X, Gao Y, Yan Q, Bao X, Zhao S, Wang J, et al. Transcriptome analysis of ochratoxin A-induced apoptosis in differentiated Caco-2 cells. Toxins. 2020;12:23.

    Article  CAS  Google Scholar 

  111. Wu S, Liu Y, Duan Y, Wang F, Guo F, Yan F, et al. Intestinal toxicity of deoxynivalenol is limited by supplementation with Lactobacillus plantarum JM113 and consequentially altered gut microbiota in broiler chickens. J Anim Sci Biotechnol. 2018;9:74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Marchioro A, Mallmann AO, Diel A, Dilkin P, Rauber RH, Blazquez FJH, et al. Effects of aflatoxins on performance and exocrine pancreas of broiler chickens. Avian Dis. 2013;57:280–4.

    Article  CAS  PubMed  Google Scholar 

  113. Mattson MP. Hormesis defined. Ageing Res Rev. 2008;7:1–7.

    Article  CAS  PubMed  Google Scholar 

  114. Calabrese EJ. Evidence that hormesis represents an “overcompensation” response to a disruption in homeostasis. Ecotoxicol Environ Saf. 1999;42:135–7.

    Article  Google Scholar 

  115. Diaz G, Calabrese E, Blain R. Aflatoxicosis in chickens (Gallus gallus): An example of hormesis? Poult Sci. 2008;87:727–32.

    Article  CAS  PubMed  Google Scholar 

  116. Yunus AW. Aflatoxin B1 in affecting broiler’s performance, immunity, and gastrointestinal tract: A review of history and contemporary issues. Toxins. 2011;3:566–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Martínez G, Diéguez SN, Fernández Paggi MB, Riccio MB, Pérez Gaudio DS, Rodríguez E, et al. Effect of fosfomycin, cynara scolymus extract, deoxynivalenol and their combinations on intestinal health of weaned piglets. Anim Nutr. 2019;5:386–95.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Pinton P, Oswald IP. Effect of deoxynivalenol and other Type B trichothecenes on the Intestine: A review. Toxins. 2014;6:1615–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Awad WA, Böhm J, Razzazi-Fazeli E, Hulan HW, Zentek J. Effects of deoxynivalenol on general performance and electrophysiological properties of intestinal mucosa of broiler chickens. Poult Sci. 2004;83:1964–72.

    Article  CAS  PubMed  Google Scholar 

  120. Awad WA, Rehman H, Böhm J, Razzazi-Fazeli E, Zentek J. Effects of luminal deoxynivalenol and L-proline on electrophysiological parameters in the jejunums of laying hens. Poult Sci. 2005;84:928–32.

    Article  CAS  PubMed  Google Scholar 

  121. Awad WA, Bohm J, Razzazi-Fazeli E, Zentek J. In vitro effects of deoxynivalenol on electrical properties of intestinal mucosa of laying hens. Poult Sci. 2005;84:921–7.

    Article  CAS  PubMed  Google Scholar 

  122. Santos RR, Oosterveer-van der Doelen MAM, Tersteeg-Zijderveld MHG, Molist F, Mézes M, Gehring R. Susceptibility of broiler chickens to deoxynivalenol exposure via artificial or natural dietary contamination. Animals. 2021;11(4):989.

  123. Liew W-P-P, Mohd-Redzwan S. Mycotoxin: Its impact on gut health and microbiota. Front Cell Infect Microbiol. 2018;8:60.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:k2179.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Wu H-J, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3:4–14.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Reddy DN. Role of the normal gut microbiota. World J Gastroenterol. 2015;21:8787–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Rinttilä T, Apajalahti J. Intestinal microbiota and metabolites—Implications for broiler chicken health and performance. J Appl Poult Res. 2013;22:647–58.

    Article  Google Scholar 

  128. Cammack KM, Austin KJ, Lamberson WR, Conant GC, Cunningham HC. Tiny but mighty: The role of the rumen microbes in livestock production. J Anim Sci. 2018;96:752–70.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Boguhn J, Neumann D, Helm A, Strobel E, Tebbe CC, Dänicke S, et al. Effects of concentrate proportion in the diet with or without Fusarium toxin-contaminated triticale on ruminal fermentation and the structural diversity of rumen microbial communities in vitro. Arch Anim Nutr. 2010;64:467–83.

    Article  CAS  PubMed  Google Scholar 

  130. Jeong JS, Lee JH, Simizu Y, Tazaki H, Itabashi H, Kimura N. Effects of the Fusarium mycotoxin deoxynivalenol on in vitro rumen fermentation. Anim Feed Sci Technol. 2010;162:144–8.

    Article  CAS  Google Scholar 

  131. Jiang Y, Yang HJ, Lund P. Effect of aflatoxin B1 on in vitro ruminal fermentation of rations high in alfalfa hay or ryegrass hay. Anim Feed Sci Technol. 2012;175:85–9.

    Article  CAS  Google Scholar 

  132. Khodabandehloo M, Malecky M, Aliarabi H, Saki AA, Alipour D. In vitro evaluation of aflatoxin B1 effect on gas production and ruminal fermentation parameters. Iran J Vet Res. 2019;20:263–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Singh R, Park S, Koo JS, Balasubramanian B. Influence of various concentrations of aflatoxin B1 on in vitro rumen fermentation of a buffalo diet. Korean J Agric Sci. 2020;47:131–8.

    CAS  Google Scholar 

  134. Maresca M. From the gut to the brain: Journey and pathophysiological effects of the food-associated trichothecene mycotoxin deoxynivalenol. Toxins. 2013;5:784–820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Loh ZH, Ouwerkerk D, Klieve AV, Hungerford NL, Fletcher MT. Toxin degradation by rumen microorganisms: A review. Toxins. 2020;12:664.

    Article  CAS  PubMed Central  Google Scholar 

  136. Schwartz-Zimmermann HE, Fruhmann P, Dänicke S, Wiesenberger G, Caha S, Weber J, et al. Metabolism of deoxynivalenol and deepoxy-deoxynivalenol in broiler chickens, pullets, roosters and turkeys. Toxins. 2015;7:4706–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Pierron A, Alassane-Kpembi I, Oswald IP. Impact of two mycotoxins deoxynivalenol and fumonisin on pig intestinal health. Porc Health Manag. 2016;2:21.

    Article  Google Scholar 

  138. Celi P, Cowieson AJ, Fru-Nji F, Steinert RE, Kluenter A-M, Verlhac V. Gastrointestinal functionality in animal nutrition and health: New opportunities for sustainable animal production. Anim Feed Sci Technol. 2017;234:88–100.

    Article  Google Scholar 

  139. Pourabedin M, Zhao X. Prebiotics and gut microbiota in chickens. FEMS Microbiol Lett. 2015;362:15.

    Article  CAS  Google Scholar 

  140. Monson M, Coulombe R, Reed K. Aflatoxicosis: Lessons from toxicity and responses to aflatoxin B1 in poultry. Agriculture. 2015;5:742–77.

    Article  CAS  Google Scholar 

  141. Deng J, Zhao L, Zhang N-Y, Karrow NA, Krumm CS, Qi D-S, et al. Aflatoxin B1 metabolism: Regulation by phase I and II metabolizing enzymes and chemoprotective agents. Mutat Res Mutat Res. 2018;778:79–89.

    Article  CAS  Google Scholar 

  142. Martignoni M, Groothuis GMM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol. 2006;2:875–94.

    Article  CAS  PubMed  Google Scholar 

  143. Fink-Gremmels J, Malekinejad H. Clinical effects and biochemical mechanisms associated with exposure to the mycoestrogen zearalenone. Anim Feed Sci Technol. 2007;137:326–41.

    Article  CAS  Google Scholar 

  144. Andretta I, Kipper M, Lehnen CR, Hauschild L, Vale MM, Lovatto PA. Meta-analytical study of productive and nutritional interactions of mycotoxins in broilers. Poult Sci. 2011;90:1934–40.

    Article  CAS  PubMed  Google Scholar 

  145. Andretta I, Kipper M, Lehnen CR, Hauschild L, Vale MM, Lovatto PA. Meta-analytical study of productive and nutritional interactions of mycotoxins in growing pigs. Anim Int J Anim Biosci Camb. 2012;6:1476–82.

    Article  CAS  Google Scholar 

  146. Yang X, Zhang B, Molony C, Chudin E, Hao K, Zhu J, et al. Systematic genetic and genomic analysis of cytochrome P450 enzyme activities in human liver. Genome Res. 2010;20:1020–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Mačak-Šafranko Ž, Sobočanec S, Šarić A, Balog T, Šverko V, Kušić B, et al. Cytochrome P450 gender-related differences in response to hyperoxia in young CBA mice. Exp Toxicol Pathol. 2011;63:345–50.

    Article  PubMed  CAS  Google Scholar 

  148. Parkinson A, Mudra DR, Johnson C, Dwyer A, Carroll KM. The effects of gender, age, ethnicity, and liver cirrhosis on cytochrome P450 enzyme activity in human liver microsomes and inducibility in cultured human hepatocytes. Toxicol Appl Pharmacol. 2004;199:193–209.

    Article  CAS  PubMed  Google Scholar 

  149. Wolbold R, Klein K, Burk O, Nüssler AK, Neuhaus P, Eichelbaum M, et al. Sex is a major determinant of CYP3A4 expression in human liver. Hepatology. 2003;38:978–88.

    Article  CAS  PubMed  Google Scholar 

  150. da Silva EO, Bracarense APFL, Oswald IP. Mycotoxins and oxidative stress: where are we? World Mycotoxin J. 2018;11:113–34.

    Article  Google Scholar 

  151. Mavrommatis A, Giamouri E, Tavrizelou S, Zacharioudaki M, Danezis G, Simitzis PE, et al. Impact of mycotoxins on animals’ oxidative status. Antioxidants. 2021;10:214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Giergiel M, Lopucki M, Stachowicz N, Kankofer M. The influence of age and gender on antioxidant enzyme activities in humans and laboratory animals. Aging Clin Exp Res. 2012;24:6.

    Google Scholar 

  153. Miyagi SJ, Brown IW, Chock JM-L, Collier AC. Developmental changes in hepatic antioxidant capacity are age-and sex-dependent. J Pharmacol Sci. 2009;111:440–5.

  154. Puppel K, Nałȩcz-Tarwacka T, Kuczyńska B, Gołȩbiewski M, Kordyasz M, Grodzki H. The age of cows as a factor shaping the antioxidant level during a nutritional experiment with fish oil and linseed supplementation for increasing the antioxidant value of milk. J Sci Food Agric. 2012;92:2494–9.

    Article  CAS  PubMed  Google Scholar 

  155. Chhaya RS, O’Brien J, Cummins E. Feed to fork risk assessment of mycotoxins under climate change influences - recent developments. Trends Food Sci Technol. 2021. https://doi.org/10.1016/j.tifs.2021.07.040.

  156. Liu C, Manstretta V, Rossi V, Van der Fels-Klerx HJ. Comparison of three modelling approaches for predicting deoxynivalenol contamination in winter wheat. Toxins. 2018;10:267.

    Article  PubMed Central  Google Scholar 

  157. Camardo Leggieri M, Mazzoni M, Battilani P. Machine Learning for predicting mycotoxin occurrence in maize. Front Microbiol. 2021;12:782.

    Article  Google Scholar 

  158. Moore GG. Practical considerations will ensure the continued success of pre-harvest biocontrol using non-aflatoxigenic Aspergillus flavus strains. Crit Rev Food Sci Nutr. 2021. https://doi.org/10.1080/10408398.2021.1873731.

  159. Kabak B, Dobson ADW, Var I. Strategies to prevent mycotoxin contamination of food and animal feed: A review. Crit Rev Food Sci Nutr. 2006;46:593–619.

    Article  CAS  PubMed  Google Scholar 

  160. Weaver AC, Adams N, Yiannikouris A. Invited review: Use of technology to assess and monitor multimycotoxin and emerging mycotoxin challenges in feedstuffs. Appl Anim Sci. 2020;36:19–25.

    Article  Google Scholar 

  161. Pankaj SK, Shi H, Keener KM. A review of novel physical and chemical decontamination technologies for aflatoxin in food. Trends Food Sci Technol. 2018;71:73–83.

    Article  CAS  Google Scholar 

  162. Peng W-X, Marchal JLM, van der Poel AFB. Strategies to prevent and reduce mycotoxins for compound feed manufacturing. Anim Feed Sci Technol. 2018;237:129–53.

    Article  CAS  Google Scholar 

  163. Čolović R, Puvača N, Cheli F, Avantaggiato G, Greco D, Đuragić O, et al. Decontamination of mycotoxin-contaminated feedstuffs and compound Feed. Toxins. 2019;11:617.

    Article  PubMed Central  CAS  Google Scholar 

  164. Karlovsky P, Suman M, Berthiller F, De Meester J, Eisenbrand G, Perrin I, et al. Impact of food processing and detoxification treatments on mycotoxin contamination. Mycotoxin Res. 2016;32:179–205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Lancova K, Hajslova J, Kostelanska M, Kohoutkova J, Nedelnik J, Moravcova H, et al. Fate of trichothecene mycotoxins during the processing: Milling and baking. Food Addit Contam Part Chem Anal Control Expo Risk Assess. 2008;25:650–9.

    Article  CAS  Google Scholar 

  166. Tibola CS, Fernandes JMC, Guarienti EM. Effect of cleaning, sorting and milling processes in wheat mycotoxin content. Food Control. 2016;60:174–9.

    Article  CAS  Google Scholar 

  167. Pascale M, Logrieco AF, Graeber M, Hirschberger M, Reichel M, Lippolis V, et al. Aflatoxin reduction in maize by industrial-scale cleaning solutions. Toxins. 2020;12:331.

    Article  CAS  PubMed Central  Google Scholar 

  168. Cheli F, Pinotti L, Rossi L, Dell’Orto V. Effect of milling procedures on mycotoxin distribution in wheat fractions: A review. LWT Food Sci Technol. 2013;54:307–14.

    Article  CAS  Google Scholar 

  169. Janić Hajnal E, Mastilović J, Bagi F, Orčić D, Budakov D, Kos J, et al. Effect of wheat milling process on the distribution of Alternaria toxins. Toxins. 2019;11:139.

    Article  PubMed Central  CAS  Google Scholar 

  170. Vanara F, Scarpino V, Blandino M. Fumonisin distribution in maize dry-milling products and by-products: Impact of two industrial degermination systems. Toxins. 2018;10:357.

    Article  PubMed Central  CAS  Google Scholar 

  171. Bullerman LB, Bianchini A. Stability of mycotoxins during food processing. Int J Food Microbiol. 2007;119:140–6.

    Article  CAS  PubMed  Google Scholar 

  172. Castells M, Pardo E, Ramos AJ, Sanchis V, Marín S. Reduction of ochratoxin A in extruded barley meal. J Food Prot. 2006;69:1139–43.

    Article  CAS  PubMed  Google Scholar 

  173. Kabak B. The fate of mycotoxins during thermal food processing. J Sci Food Agric. 2009;89:549–54.

    Article  CAS  Google Scholar 

  174. Awad WA, Ghareeb K, Böhm J, Zentek J. Decontamination and detoxification strategies for the Fusarium mycotoxin deoxynivalenol in animal feed and the effectiveness of microbial biodegradation. Food Addit Contam Part A. 2010;27:510–20.

    Article  CAS  Google Scholar 

  175. Ghanem I, Orfi M, Shamma M. Effect of gamma radiation on the inactivation of aflatoxin B1 in food and feed crops. Braz J Microbiol. 2008;39:787–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Herzallah S, Alshawabkeh K, Fataftah AA. Aflatoxin decontamination of artificially contaminated feeds by sunlight, γ-radiation, and microwave heating. J Appl Poult Res. 2008;17:515–21.

    Article  CAS  Google Scholar 

  177. Calado T, Venâncio A, Abrunhosa L. Irradiation for mold and mycotoxin control: A review. Compr Rev Food Sci Food Saf. 2014;13:1049–61.

    Article  CAS  Google Scholar 

  178. Boudergue C, Burel C, Dragacci S, Favrot M-C, Fremy J-M, Massimi C, et al. Review of mycotoxin-detoxifying agents used as feed additives: mode of action, efficacy and feed/food safety. EFSA Support Publ. 2009;6:22E.

    Google Scholar 

  179. Vanhoutte I, Audenaert K, De Gelder L. Biodegradation of mycotoxins: Tales from known and unexplored worlds. Front Microbiol. 2016;7:561.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Zhu Y, Hassan YI, Watts C, Zhou T. Innovative technologies for the mitigation of mycotoxins in animal feed and ingredients—A review of recent patents. Anim Feed Sci Technol. 2016;216:19–29.

    Article  CAS  Google Scholar 

  181. Haque MA, Wang Y, Shen Z, Li X, Saleemi MK, He C. Mycotoxin contamination and control strategy in human, domestic animal and poultry: A review. Microb Pathog. 2020;142:104095.

    Article  CAS  PubMed  Google Scholar 

  182. He J, Zhou T, Young JC, Boland GJ, Scott PM. Chemical and biological transformations for detoxification of trichothecene mycotoxins in human and animal food chains: a review. Trends Food Sci Technol. 2010;21:67–76.

    Article  CAS  Google Scholar 

  183. Devreese M, De Backer P, Croubels S. Different methods to counteract mycotoxin production and its impact on animal health. Vlaams Diergeneeskd Tijdschr. 2013;82(4):180–90.

  184. EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP). Statement on the establishment of guidelines for the assessment of additives from the functional group ‘substances for reduction of the contamination of feed by mycotoxins’. EFSA J. 2010;8:1693.

    Article  Google Scholar 

  185. EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP), Rychen G, Aquilina G, Azimonti G, Bampidis V, de Lourdes Bastos M, et al. Safety and efficacy of fumonisin esterase (FUMzyme®) as a technological feed additive for all avian species. EFSA J. 2016;14:e04617.

    Google Scholar 

  186. Loi M, Fanelli F, Liuzzi V, Logrieco A, Mulè G. Mycotoxin biotransformation by native and commercial enzymes: Present and future perspectives. Toxins. 2017;9:111.

    Article  PubMed Central  CAS  Google Scholar 

  187. Galvano F, Piva A, Ritieni A, Galvano G. Dietary strategies to counteract the effects of mycotoxins: A review. J Food Prot. 2001;64:120–31.

    Article  CAS  PubMed  Google Scholar 

  188. Jouany JP. Methods for preventing, decontaminating and minimizing the toxicity of mycotoxins in feeds. Anim Feed Sci Technol. 2007;137:342–62.

    Article  CAS  Google Scholar 

  189. Huwig A, Freimund S, Käppeli O, Dutler H. Mycotoxin detoxication of animal feed by different adsorbents. Toxicol Lett. 2001;122:179–88.

    Article  CAS  PubMed  Google Scholar 

  190. Yiannikouris A, André G, Buléon A, Jeminet G, Canet I, François J, et al. Comprehensive conformational study of key interactions involved in zearalenone complexation with β-d-Glucans. Biomacromolecules. 2004;5:2176–85.

    Article  CAS  PubMed  Google Scholar 

  191. Yiannikouris A, André G, Poughon L, François J, Dussap C-G, Jeminet G, et al. Chemical and conformational study of the interactions involved in mycotoxin Ccmplexation with β-d-Glucans. Biomacromolecules. 2006;7:1147–55.

    Article  CAS  PubMed  Google Scholar 

  192. Liu N, Wang JQ, Jia SC, Chen YK, Wang JP. Effect of yeast cell wall on the growth performance and gut health of broilers challenged with aflatoxin B1 and necrotic enteritis. Poult Sci. 2018;97:477–84.

    Article  CAS  PubMed  Google Scholar 

  193. Wielogórska E, MacDonald S, Elliott CT. A review of the efficacy of mycotoxin detoxifying agents used in feed in light of changing global environment and legislation. World Mycotoxin J. 2016;9:419–33.

    Article  CAS  Google Scholar 

  194. Cavret S, Laurent N, Videmann B, Mazallon M, Lecoeur S. Assessment of deoxynivalenol (DON) adsorbents and characterisation of their efficacy using complementary in vitro tests. Food Addit Contam Part A. 2010;27:43–53.

    Article  CAS  Google Scholar 

  195. Oh S-Y, Quinton VM, Boermans HJ, Swamy HVLN, Karrow NA. In vitro exposure of Penicillium mycotoxins with or without a modified yeast cell wall extract (mYCW) on bovine macrophages (BoMacs). Mycotoxin Res. 2015;31:167–75.

    Article  CAS  PubMed  Google Scholar 

  196. Díaz-Llano G, Smith TK. The effects of feeding grains naturally contaminated with Fusarium mycotoxins with and without a polymeric glucomannan adsorbent on lactation, serum chemistry, and reproductive performance after weaning of first-parity lactating sows. J Anim Sci. 2007;85:1412–23.

    Article  PubMed  CAS  Google Scholar 

  197. Firmin S, Gandia P, Morgavi DP, Houin G, Jouany JP, Bertin G, et al. Modification of aflatoxin B1 and ochratoxin A toxicokinetics in rats administered a yeast cell wall preparation. Food Addit Contam Part A. 2010;27:1153–60.

    Article  CAS  Google Scholar 

  198. Mohaghegh A, Chamani M, Shivazad M, Sadeghi AA, Afzali N. Effect of esterified glucomannan on broilers exposed to natural mycotoxin-contaminated diets. J Appl Anim Res. 2017;45:285–91.

    Article  CAS  Google Scholar 

  199. Naveed S, Chohan K, Jabbar M, Ditta Y, Ahmed S, Ahmad N, et al. Aflatoxin M1 in Nili-ravi buffaloes and its detoxification using organic and inorganic toxin binders. J Hell Vet Med Soc. 2018;69:873–8.

    Article  Google Scholar 

  200. Weaver AC, See MT, Kim SW. Protective effect of two yeast based feed additives on pigs chronically exposed to deoxynivalenol and zearalenone. Toxins. 2014;6:3336–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Awuchi CG, Ondari EN, Ogbonna CU, Upadhyay AK, Baran K, Okpala COR, et al. Mycotoxins affecting animals, foods, humans, and plants: Types, occurrence, toxicities, action mechanisms, prevention, and detoxification strategies—A revisit. Foods. 2021;10:1279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Lee IK, Kye YC, Kim G, Kim HW, Gu MJ, Umboh J, et al. Stress, nutrition, and intestinal immune responses in pigs — A review. Asian Australas J Anim Sci. 2016;29:1075–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Antonissen G, Martel A, Pasmans F, Ducatelle R, Verbrugghe E, Vandenbroucke V, et al. The impact of Fusarium mycotoxins on human and animal host susceptibility to infectious diseases. Toxins. 2014;6:430–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We appreciate the work of all colleagues on this topic. The authors have not stated any conflicts of interest.

Funding

This work has been funded by Natural Sciences and Engineering Research Council of Canada and Alltech Inc, KY, US [532378-18].

Author information

Authors and Affiliations

Authors

Contributions

RX prepared the original draft. EK and RX structured the manuscript content. RX, EK, AY, LS and NK contributed to editing, revision and finalization of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Niel A. Karrow.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

AY is the employee of Alltech lnc. USA. The authors have no conflict of interest to disclose.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, R., Kiarie, E.G., Yiannikouris, A. et al. Nutritional impact of mycotoxins in food animal production and strategies for mitigation. J Animal Sci Biotechnol 13, 69 (2022). https://doi.org/10.1186/s40104-022-00714-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-022-00714-2

Keywords