Next Article in Journal
Two Distinct Faces of Vitamin C: AA vs. DHA
Next Article in Special Issue
An Evaluation of the Anti-Carcinogenic Response of Major Isothiocyanates in Non-Metastatic and Metastatic Melanoma Cells
Previous Article in Journal
Val16A SOD2 Polymorphism Promotes Epithelial–Mesenchymal Transition Antagonized by Muscadine Grape Skin Extract in Prostate Cancer Cells
Previous Article in Special Issue
Protective Role of Natural and Semi-Synthetic Tocopherols on TNFα-Induced ROS Production and ICAM-1 and Cl-2 Expression in HT29 Intestinal Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impact of Mycotoxins on Animals’ Oxidative Status

by
Alexandros Mavrommatis
1,
Elisavet Giamouri
1,
Savvina Tavrizelou
1,
Maria Zacharioudaki
1,
George Danezis
2,3,
Panagiotis E. Simitzis
4,
Evangelos Zoidis
1,
Eleni Tsiplakou
1,
Athanasios C. Pappas
1,*,
Constantinos A. Georgiou
2,3 and
Kostas Feggeros
1
1
Laboratory of Nutritional Physiology and Feeding, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece
2
Chemistry Laboratory, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
3
FoodOmics GR Research Infrastructure, Agricultural University of Athens, 11855 Athens, Greece
4
Laboratory of Animal Breeding and Husbandry, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece
*
Author to whom correspondence should be addressed.
Antioxidants 2021, 10(2), 214; https://doi.org/10.3390/antiox10020214
Submission received: 30 December 2020 / Revised: 20 January 2021 / Accepted: 26 January 2021 / Published: 1 February 2021
(This article belongs to the Special Issue Feature Papers in Antioxidants in 2020)

Abstract

:
Mycotoxins appear to be the “Achilles’ heel” of the agriculture sector inducing enormous economic losses and representing a severe risk to the health of humans and animals. Although novel determination protocols have been developed and legislation has been implemented within Europe, the side effects of mycotoxins on the homeostatic mechanisms of the animals have not been extensively considered. Feed mycotoxin contamination and the effects on the antioxidant status of livestock (poultry, swine, and ruminants) are presented. The findings support the idea that the antioxidant systems in both monogastrics and ruminants are challenged under the detrimental effect of mycotoxins by increasing the toxic lipid peroxidation by-product malondialdehyde (MDA) and inhibiting the activity of antioxidant defense mechanisms. The degree of oxidative stress is related to the duration of contamination, co-contamination, the synergetic effects, toxin levels, animal age, species, and productive stage. Since the damaging effects of MDA and other by-products derived by lipid peroxidation as well as reactive oxygen species have been extensively studied on human health, a more integrated monitoring mechanism (which will take into account the oxidative stability) is urgently required to be implemented in animal products.

1. Introduction

Mycotoxins are particularly toxic substances, which are mostly produced by three genera of fungi: Aspergillus, Penicillium, and Fusarium [1]. Mycotoxins have been characterized as secondary metabolites of low molecular weight [1,2]. They could be pathogenic or simply saprophytic and can show up on many food categories [2,3], either liquid or solid as well as on animal feed [1,2]. Contamination by mycotoxins can affect both human and animal health [1,2,3]. Some fungi can produce multiple mycotoxins [1]. In Table 1, the major fungi and produced mycotoxins are presented. The existence of mycotoxins has an outstanding repercussion on global economy and international commerce [1].
One of the most studied groups of mycotoxins is aflatoxins (AF), which are produced by the fungal genus Aspergillus, notably A. flavus, A. parasiticus, A. ochraceus, A. carbonarius, A. niger (lower significance), and seldomly by A. pseudotamari [3,4,5,6]. Favorable weather conditions are important for the production of aflatoxins, commonly apparent in tropical and sub-tropical countries [3]. Known compounds generated by A. flavus are aflatoxin B1 (AFB1), B2 (AFB2), G1 (AFG1), and G2 (AFG2) [2,3]. An in vivo metabolite found in animals fed AFB1 contaminated feed is M1 (AFM1) with known carry over to animal products like milk, eggs, and tissues [3]. Aflatoxins have been linked to human liver carcinogenesis and health issues in various animals that can lead to high mortality rates, especially in poultry [1,2,3].
Ochratoxins include several types, namely ochratoxin A, B, and C. Ochratoxin A (OTA), which can be found in several foods is the most profuse and damaging one. OTA are produced by fungal species belonging to Aspergillus and Penicillium genera, most notably A. ochraceus, A. niger, A. carbonarius, A. westerdijkiae, A. glaucus, A. melleus, A. alliaceus, A. auricomus, P. verrucosum, and P. viridicatum [1,5,6] (Table 1). Environmental conditions that favor OTA production are variable and depend on fungal genus [6]. Ochratoxins are toxic for the immune-, nervous system, and kidneys for both animals and humans [1,2,6]. Furthermore, they are responsible for DNA damage, cancer, and teratogens [2,6].
Zearalenone (ZEA) is produced mostly by the Fusarium genus. Specifically, ΖΕA is synthesized by the following fungal species: F. graminearum, F. roseum, F. culmorum, F. equiseti, F. cerealis, F. verticillioides, F. incarnatum, F. crookwellense, and F. semitectum [2] (Table 1). The great interest in their study lies in the fact that they are naturally occurring estrogens [6] affecting humans and several animal species. Swine is the mostly affected species by ZEA. It is known than ZEA causes hormonal disorders and is associated with human breast cancer [1,6]. Additionally, ZEA presence in feed during pregnancy may reduce the survival rate of the embryo [1]. Moreover, ZEA provokes vulvar dilatation and redness, decreases sperm quality, and rectal prolapse [1]. Zearalenone consumption by dairy cows can be excreted into milk [6].
Fumonisins (FUM) are produced mainly by Fusarium species such as F. verticillioides, F. proliferatum, and also A. niger [2,6] (Table 1). FUM are divided into groups: A, B, C, and P. From these groups, only six fumonisins have been recognized: two from group A (FA1, FA2) and four from group B (FB1, FB2, FB3, FB4) [6]. FB1, FB2, and FB3 can be found in nature, but only FB1 turns up in high percentage and constitutes the most toxic one. The FUM health effects on humans have not yet been investigated. On the other hand, surveys have shown that fumonisins cause serious damage in animal heath such as cancer, especially in the liver and kidney [2,6]. Additionally, FUM contamination causes an illness in horses called leukoencephalomalacia (ELEM) and a syndrome known as pulmonary edema and hydrothorax in pigs [1,2]. It is also known that FB’s cause damage on neural and liver tissues in fish [1].
Trichothecenes are a large family of chemically related mycotoxins produced by various fungal genera such as Fusarium, Myrothecium, Trichoderma, Trichothecium, Cephalosporium, Verticimonosporium, and Stachybotrys [2]. Trichothecenes are divided in four groups: A, B, C, and D. Most notably, group A included the most toxic compounds (T-2 and HT-2 toxins, diacetoxiscirpenol) and group B consisted of deoxynivalenol and nivalenol [6]. In addition, F. langsethiae, F.poae, F. sporotrichioides, F equiseti, and F. acumninatum are responsible for mycotoxins, which belong in group A [2]. Deoxynivalenol (DON) is produced by F. graminearum, F. culmorum, F. cerealis (F. croockwellense), F. sporotrichioides, F. poae, F. tricinctum, and F. acuminatum [2,6]. HT-2 and T-2 toxicity associated with growth retardation, myelotoxicity, hematotoxicity, and sepsis on contact sites [2]. DON intake causes damage in the digestive system and reduction in food appetite [6]. Nivalenol (NIV) is produced by F. cerealis, F. crookwellense, F.poae, F. nivale, F. culmorum, and F. graminearum. NIV intake causes serious damage on bone marrow, intestinal membranes, and lymphoid organs as well as erythropenia, leycopenia, hemorrhage, and diarrhea [6].
Sterigmatocystin (STC) is produced by more than 50 fungal species with the main source that of Aspergillus [7,8,9], notably Aspergillus flavus, A. parasiticus, A. versicolor, A. nidulans, and A. versicolor [7,10]. Sterigmatocystin has been accused of tumorigenicity, hepatocellular carcinomas, angiosarcomas in brown fat, lung adenomas, and hemangiosarcomas in the liver and pulmonary adenomas in both human and animals [7,11]. As far as animals are concerned, surveys have shown that STC is hepatoxic in both poultry and pigs, nephrotoxic in poultry, and extremely toxic in fishes. In cattle, bloody diarrhea, which can even lead to death, has been observed [7,11]. It is noteworthy that this mycotoxin group can be found not only in food and animal feedstuff, but also in indoor environments to wet building and housing materials [11,12].
Ergot alkaloids (EAs) have gained considerable attention with more than 50 compounds to have been identified including, but not limited to ergometrine, ergotamine, ergosine, ergocristine, ergocryptine, ergocornine, and the corresponding inine epimers [13,14,15]. EAs are produced by the Claviceps and Epichloë fungus [13,14,15]. Ergot alkaloids have been accused of neurotoxicity, weight reduction, and imbalance of the endocrine system (alter some hormones’ normal levels) [13,14].
Furthermore, Phomopsins are significant mycotoxins, although scarce data exist on their effects [16]. Phomopsins have been classified into five groups: A, B, C, D, and E, of which the most toxic is group A [16]. These mycotoxins are produced by Diaporthe toxica (Phomopsis leptostromiformis) [16,17]. Experimental results showed that these modified polypeptides are nephrotoxic, immunotoxic, hematotoxic, genotoxic, and carcinogenic [16]. Additionally, these mycotoxins are hematotoxic for animals and cause liver cancer in rats [16]. Phomopsin intake seems to be the cause of Lupinosis in shepherd animals [17].
Citrinin (CTN/CIT) is produced by three different genera of fungi: Aspergillus, Penicillium, and Monascus [9,12,18,19]. Several research data have described the toxicity of this secondary metabolite in the kidneys, liver, and the immune system [18,19,20]. Furthermore, citrinin may cause DNA damage and cancer [19]. It is noteworthy that this polyketide mycotoxin is detected in various herbal medicines [9,19].
Enormous interest exists in Altenaria mycotoxin research because of their frequent presence in food and feed [19,21]. Altenaria species are synthesized by Altenaria fungi and produce more than 70 toxins, but only a few have been featured as mycotoxins both for humans and animals [9,19,21]. Some complexes include, but are not limited to altenuene, tenuazonic acid, alternariol monomethyl ether, and alternariol [9,21]. Research results have shown that Altenaria compounds are phytotoxic, fetotoxic, genotoxic, and responsible for mutagenicity and teratogenicity in both humans and animals (especially mice, chicken, and dogs) [19,21,22].
Mycotoxin contamination of food has enormous economic and commercial implications [1,3,23,24,25]. Several times, the whole grain batch may need to be destroyed, more often maize, wheat, rye, barley, and oat [1]. Surveys have shown that economic losses can range from hundreds of million to billion dollars per year [25]. According to the FAO, financial damage amounts to twenty-five percent of the global harvest [3]. It is important to note, that these data were modified due to population growth from year to year, which causes an increase in production to meet mankind’s needs, so the level of mycotoxins increased due to the extension of storage time [24]. Loss incidents due to mycotoxins have been noticed in over a hundred countries [25].
Responsible fungal genera differ from country to country because of environmental conditions that favor species production [23]. The presence of mycotoxins reduces the crop quantity and quality as well as livestock production [3,23]. Moreover, economic losses can be indirect such as treatments that humans and animals may need after infected food consumption and reduction in the final product price due to the raw materials used [3,23]. Moreover, business related to crops may fail or lose profits [23]. An extra cost is that contaminated crops cannot be used as seeds the next year [23]. The most important commercial implications are losses of foreign exchange earnings due to mycotoxin contamination of food [23]. According to the literature, mycotoxins can be found in both human food and in animal feed. It is noticeable that the same fungal genera are responsible for the appearance of different mycotoxins. The results of the surveys are listed in Table 1.
Legislation and regulations on mycotoxins are continually evolving, since countries recognize that addressing mycotoxin contamination in feeds and foods will portray plenty of advantages such as decrease health-care expenses, improvement of the international trade, and promotion of sustainability [26]. Due to the aforementioned detrimental effects of mycotoxins, the European Union has set certain limits via regulations on their content in food and feed to preserve the health of both citizens and animals (EC, 1126/2007; EC, 165/2010; EC, 574/2011; EC, 594/2012; EC, 212/2014, in addition, the European Union provided recommendation EC, 2016/1319). The regulations and recommendations are presented in Table 2. Several other countries have set maximum mycotoxin limits. U.S. regulations concerning aflatoxin contaminated foods for humans are 10-fold higher than that of the EU, while those of India are 15-fold [26].
The detrimental effects of mycotoxins on livestock may result in morbidity and mortality [19,27]. Mycotoxins are capable of inducing several imbalances in the cellular level, resulting in acute or chronic health issues on animals. The induction of oxidative stress and generation of reactive oxygen species (ROS) may be a major trigger of detrimental outcomes [28]. Cells produce ROS and reactive nitrogen species (RNS) as a result of their physiological metabolism or as signaling molecules capable of regulating substantial homeostatic functions [29]. Another crucial role of ROS emerges through the phagocytic properties of immune cells [30]. It seems that there are two faces of ROS, redox signaling and oxidative stress, contributing to both physiological and pathological conditions. Within the range of low to moderate concentration levels, ROS regulate normal cell function, whilst at their highest levels, their biochemical instability damages cell component such as lipids, proteins, and DNA, resulting in oxidative stress [31]. However, the organism regulates an efficient network of mechanisms to counteract the detrimental effect of ROS, which includes enzymatic or endogenous and non-enzymatic (exogenous) factors [32].
Imbalance between free radicals generated by mycotoxins and the antioxidant defense system triggers a cascade of damaging effects [28]. More specifically, in vitro studies have reported that AFB1 induced downregulation of antioxidant enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase, resulting in increased lipid peroxidation by-products (malondialdehyde; MDA) and a severe decrease in the levels of the predominant exogenous antioxidant compound, namely, reduced glutathione (GSH) [28]. Liver and kidneys are considered the main organs of the mycotoxins’ metabolism. Proper hepatic function is evaluated through several laboratory analyses including, but are not limited to, the determination of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Increased ALT and/or AST activities are indicative of hepatocellular injury [33]. Plenty of factors predominantly affect the cellular function and homeostatic balance in animals affected by mycotoxins. These factors include, but are not limited to animal species, time of exposure, age, production stage, and toxin co-contamination (synergetic effect). The most determinant factors seem to be contamination level, type of mycotoxin, and time of exposure (acute or chronic). Under this context, it has been observed that the antioxidant defense of cows could be triggered under a short time of exposure to AFB1 (3–7 days), resulting in increased SOD activity as a response to oxidative challenge [34,35]. Moreover, long-term exposure of cows up to nine weeks to AFB1 manifested in low SOD activity, total antioxidant capacity, GSH-Px, and high levels of plasma MDA [36]. This study aims to bridge the detrimental effect of mycotoxin contamination in livestock with the antioxidant status of animal organisms, which is related to the products’ oxidative stability.

2. The Effect of Mycotoxins on the Antioxidant Status of Pigs and Poultry

Mycotoxins are considered one of the main contaminants in animal diets and their presence might damage livestock health [38,39]. The intensive rearing of poultry and swine might pose a risk for animal health and production because of the high consumption of cereals and oilseeds, which are more likely to contain mycotoxins [40,41,42]. Mycotoxins affect several organs such as the gastrointestinal system, liver, and immune system, and in general reduce productivity. Although one mycotoxin might be harmful for animals, the presence of more can be more toxic due to their synergism. Some of the most common species that can be found in feeds are AF, OTA, ZEA, FUM, DON, and T-2 toxin.
The toxic effect of mycotoxins can lead to oxidative stress (OS) and the generation of free radicals [43,44]. The increased number of free radicals in accordance with the malfunction of antioxidant system damages DNA, proteins, and lipids [45]. Oxygen free radicals and antioxidants are produced normally by cells in a balanced range. Exterior parameters can promote the generation of oxidative stress and an overproduction of free radicals [46], causing an imbalance in the homeostasis mechanism of the cells. Disruptions of the antioxidant system and excess generation of free radicals may lead to oxidative stress [47]. Valco et al. [48] stated that oxidative stress exists when the antioxidant capacity of a cell is overtaken due to the over production of reactive oxygen species (ROS), like the hydroxyl radical (HO), perhydroxyl radical (HOO•−), superoxide anion (O2•−), and RNS including nitric oxide (NO). The excessive number of ROS species might cause an alteration or a generation of several intracellular mechanisms that oxidate DNA, proteins, and membrane lipids. Cell death is more likely if lipid peroxidation occurs, indicating the serious consequences of the toxicity of mycotoxins [49]. It is not clear if mycotoxins induce lipid peroxidation by triggering free radical production or by undermining the antioxidant defense. In order to tackle this situation, cells use primary and secondary enzymatic systems to avoid excessive damage [48]. Antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR), and glutathione peroxidase (GSH-Px) compose a primary system to cope with free radicals or create a mechanism with glutathione (GSH).
Nutritional stress factors are responsible for negative effects in cell homeostasis. Mycotoxins are such kinds of factors and seem to have a negative impact on antioxidant enzyme function (Table 3). Galvano et al. [50] reported that AF are one of the most dangerous mycotoxin species and evaluated several dietary strategies to counteract the effects of mycotoxins. Alterations may occur depending on the mycotoxin species, the dose, and the duration of exposure, or in the presence of other antioxidants. An antioxidant enzyme may increase, if an oxidative stress occurs, or decrease, depending on the action of the mycotoxins.
Several studies have been conducted to examine the effects of several mycotoxin species on antioxidant enzymes in poultry and swine (Table 3). Results by Chen et al. [51] indicated the generation of oxidative stress in the spleen of chickens after the consumption of AFB1, as shown by the decreased levels of antioxidant enzymes such as GSH-Px, GR, CAT, and levels of malondialdehyde (MDA) and GSH. In agreement with the aforementioned study, Shahid et al. [52] found an increased rate in MDA, while total superoxide dismutase (T-SOD), GSH-Px, CAT, GR, and glutathione-S transferase (GSTs) were decreased in samples of liver and serum, as an effect of the contamination of one-day-old chick diets with a 1 mg AFB1/kg diet. These decreased activities of the above enzymes might lead to the generation of hydroxyl radicals, which play an important role in the lipid peroxidation process [53].
In another study [54], broilers that consumed a diet with 1 ppm aflatoxin B1 gained less weight, had lower feed intake, and at the same time, the activity of SOD was increased while that of CAT was decreased compared to the control group. MDA levels in serum were higher in broilers fed aflatoxin. Moreover, the control group had a lower activity of AST and ALT compared to the AFB1 group. Additionally, blood glucose was decreased and both cholesterol and triglycerides in the AFB1 group were increased. Similarly, Eraslan et al. [55] reported that exposure to AF at high doses caused lipid peroxidation in broilers. Li et al. [56] reported the effect of adding OTA in the diet of broilers in the inclusion rate of 50 μg/kg. MDA levels in kidneys were increased while the total antioxidant capacity (TAC) was decreased and levels of SOD, CAT, and GSH were markedly lower than in the control group. It was suggested that OTA induced the production of reactive oxygen, resulting in oxidative stress in the kidneys of chickens.
Yang et al. [33] carried out an in vivo and in vitro trial feeding broilers with a diet contaminated with T-2 and HT-2 mycotoxins. In the in vivo experiment, a reduction in the body weight and weight gain were observed, and the feed conversion ratio was worse compared to the control. These results were most prominent in the group with the highest concentration of toxins (4 mg/kg T-2 and 0.667 mg/kg HT-2). In the in vitro trial, a reduction of the GSH concentration in cells incubated with increasing concentrations of T-2 and HT-2 mycotoxins was reported (Table 3). Moreover, ALT/AST, GSH-Px, CAT, and SOD activities as well as MDA concentration were increased compared to the control. These results suggest that oxidative stress might be induced by the combination of these two toxins and is dose-related. Similar results were reported by Oskoueian et al. [57] after the in vitro application of AFB1 mycotoxin in the hepatocytes of five-week-old roosters. Antioxidant enzymes were negatively affected and at the same time, MDA levels increased. Similarly, Dvorska et al. [58] reported that the presence of the T-2 toxin in the broilers diet at 8.1 mg/kg fed for 3 weeks resulted in a decrease in the concentration of selenium, α-tocopherol, carotenoids, ascorbic acid, Se-dependent glutathione peroxidase (Se-GSH-Px), and reduced glutathione in the liver.
Studies have been conducted to determine the consequences of mycotoxins in the antioxidant status of pigs. Pigs are the most sensitive animals toward the products of aflatoxins, and damage in the liver and gut were observed after their consumption [59,60]. For instance, Thanh et al. [61] carried out an experiment with 6-kg weaned piglets that were fed diets contaminated with DON or/and ZEA. The control group contained 0.8 mg/kg DON and the contaminated diet contained 3.1 mg/kg of DON and 1.8 mg/kg of ZEA. The combination of DON-ZEA did not have any impact on the performance parameters for pigs, but induced oxidative stress. This was affirmed by the high level of MDA in the plasma and SOD in the liver. Antioxidant enzymes and GSH concentrations in plasma and liver were not affected. On the other hand, Sun et al. [62] studied the effect of naturally fed contaminated corn with aflatoxins (20 μg/kg) and FUM in pigs (6.02 ± 0.83 kg BW). Growing performance parameters were not significantly different between the control and the treatment group. MDA concentration was not affected by the presence of aflatoxin in the diet. Da Silva et al. [63] studied the intestinal explants of pigs after exposure to FB1 and/or DON in the treatments: DON 10 μM, FB1 70 μM, DON 10 μM + FB1 70 μM. From the results, it can be summarized that GSH was lower in treatments with one mycotoxin or a combination compared to the control group.
Reduced activity of the enzymes CAT, SOD, GSH-Px in plasma and organs was observed when weaned pigs were fed with 320 ppb of pure AFB1 [64]. The total antioxidant capacity also decreased as an effect of AFB1. Ren et al. [65] used porcine splenic lymphocytes and treated them with different concentrations of DON, ZEA, and their combination. SOD, CAT, GSH-Px, and GSH decreased when lymphocytes were exposed in DON or ZEA even in the lowest doses, when compared with the control group. In the group of DON and ZEA combination, antioxidant enzymes were lower than in the groups of DON or ZEA separately. In agreement with the previous studies, MDA increased in the exposed groups and was higher in the combination group.
Antioxidants of natural origin may protect against the toxic effects of mycotoxins by increasing the function of antioxidant enzymes [66] and the total antioxidant capacity in broilers against those contaminated with AFB1 feeds in broilers [54].

3. The Effect of Mycotoxins on the Antioxidant Status of Ruminants

Within the ruminant sector, feed contamination with mycotoxins results in crucial economic losses and food safety concerns. The economic impact of mycotoxins could either be directly through the rejection of the contaminated animal products occurring in reduced revenues, or indirectly via the animal’s long-term health exposure. Specifically, contaminated animals often showed severe immunosuppression, leading them to infection susceptibility or preventative vaccination failure. In this section, the aim was to expand on the current knowledge of the mycotoxin effects on ruminant health through examining the potential burden of immune and antioxidant systems.
The effects of mycotoxins in ruminants are not as severe as in monogastric animals, since rumen microbiome is able to metabolize and biotransform some toxins, however, without necessarily eliminating its whole compound load [67]. Hence, it could be hypothesized that ruminants are less susceptible to mycotoxins than monogastrics. However, it has also been mentioned that certain mycotoxins cause direct toxicity in rumen microbes, first and foremost to the cellulolytic [68]. More specifically, fusaric acid has been shown to exert an inhibitory effect against Ruminococcus albus and Methanobrevibacter ruminantium, predominant rumen microbes that contribute to cellulose degradation and hydrogen neutralization within the rumen, respectively [69]. Since ruminant physiology is strongly dependent on rumen microbiome and their dynamic biochemical procedures, the direct effect of mycotoxins on the rumen habitat activates a domino effect of physiological imbalances. Such imbalances have been previously summarized by Gallo et al. [70] where AFB1, DON, Gliotoxin, and Patulin negatively affected rumen dry matter and NDF digestibility. A severe decrease in rumen fiber digestion activity may enhance the utilization of high fermentable carbohydrates, which along with a suppression of rumen pH, results in MDA escalation and total antioxidant capacity inhibition in blood and tissues [71]. Furthermore, mycotoxin negatively affects the microbial protein synthesis within the rumen, resulting in a negative protein balance (NPB) [68]. During the demanding peripartum period, ruminants are able to catabolize their muscle tissue in order to be supplied with essential amino acids (AA) to fulfil their high protein demands [72]. Muscle hypercatabolism causes a significant increase in RNS production that in turn disrupts the oxidative equilibrium [73,74]. This unfavorable condition is further burdened with the presence of AFB1, since it has been observed that the inclusion of AFB1 in lactating cows significantly decreases the feed intake [70]. In addition, high genetic merit cows may be in a negative energy balance (NEB) during the prepartum period and early lactation with an inability to meet their high energy and nutrient requirements, leading to lipid mobilization and in turn, increased formation of beta-hydroxybutyrate (BHBA) and non-esterified fatty acids (NEFA). Indeed, the NEB induced by undernutrition in pregnant sheep results in increased levels of H2O2 and MDA, while CAT and SOD activities were observed suppressed in both maternal and fetal livers, indicating a severe oxidative stress [75]. Considering the aforementioned, mycotoxin contamination in high performance ruminants and during their transition period can further burden their cellular homeostasis. In addition to the important role of rumen in volatile fatty acid production, the liver has a critical role in the metabolism of glucose, lipid and nitrogen metabolism, ketogenesis, immune function, ammonia circulating, hormone catabolism, and vitamin and mineral metabolism. Proper liver function is reflected in the activity of several enzymes, most notably AST and ALT. Increased AST activity is linked to oxidative burst since the cell damage is related to free radical production [61].
AFB1, OTA, and ZEA are considered to be the predominant mycotoxins in agricultural products [76]. Huang et al. [77] tested the aforementioned mycotoxin contamination in the diet of dairy goats, reporting an intense oxidative burst (Table 4). The combination of 50 μg AFB1, 100 μg OTA, and 500 μg ZEA/kg dry matter intake (DMI) significantly increased the MDA serum concentration, decreased the total antioxidant capacity, and decreased the activities of SOD and GSH-Px [77]. These metabolic alterations portray an increase in ROS production and foremost in the superoxide anion and hydrogen peroxidase as precipitated by their corresponding neutralization mechanisms (SOD, GSH-Px). It could be hypothesized that the formation of the above unstable radicals oxidized the polyunsaturated fatty acids (PUFA) of the cells’ phospholipid membranes and MDA and other by-products were produced. On a cellular level, the activities of AST, ALT, alkaline phosphatase (ALP), and total bilirubin (TBIL) were increased, reflecting severe damage either on the hepatocytes or on the cell membranes’ permeability [78]. In addition to the detrimental role of ROS on cell membranes, findings on interleukin 6 (IL-6) concentration indicate further imbalances in the immune system. Specifically, the increase in IL-6 might be attributed to the regulatory effect of mitogen activate protein kinase (MAPK), which is triggered by ROS and promote signaling for pro-inflammatory response [79,80].
Both level and type of mycotoxin are substantial parameters of the severity of toxicity. For instance, the combination of 50 μg AFB1 and 100 μg OTA was more toxic than that of 50 μg AFB1 and 500 μg ZEA/kg DMI, despite the fact that there were higher concentration levels of ZEA compared to OTA [77]. In agreement, Mohamed et al. [81] fed sheep with diets that were contaminated with 50 μg AFB1 and 100 μg OTA/kg DMI during the peripartum period. A significant milestone in this study was related to the survival rates of lambs, which dropped from 100% to 50% when mycotoxins were added to the animals’ experimental diet. This observation may be correlated to the aforementioned reports of the neutralization role of rumen microbiome toward mycotoxins. Since the newborn’s stomachs are still not developed and the microbe’s colonization is still in progress, the animals are prone to the deleterious effect of toxins in the same extent as monogastrics. The response of oxidative indices and antioxidant enzyme activities in the aforementioned study by Mohamed et al. [81] were relatively comparable to a previous work by Huang et al. [77], indicating a pronounced negative effect of AFB1 and OTA on the oxidative balance in small ruminants. In this context, Wang et al. [82] investigated the effect of 100 μg AFB1/kg in 60 day-old lamb diets reporting no mortality, while body weight gain was decreased approximately to half. These alterations in productive features can be attributed to immense oxidative damage, as demonstrated by glutathione and glutathione dependent enzymes in the liver and duodenal mucosa of lambs. Specifically, aflatoxicosis decreased GSH concentration and GSTs and GR activities in both tissues, suggesting an inefficiency in the neutralized formed ROS and an incapability in detoxifying cells from xenobiotics (GSTs) [83]. A comparable experimental trial was conducted by Nayakwadi et al. [84] in goat kids (2–3 month-old) by contaminating diets with 10 or 20 ppm T-2 toxin. In the same way, the lipid peroxidation index in the liver, intestine, kidneys, and spleen were significantly increased by the addition of mycotoxins. Observing the results of these studies, discrepancies were revealed related to the activities of CAT and SOD, indicating that depending on stress impact, antioxidant enzymes are differentially modulated (Table 4). Different mycotoxins may create different stress factors. Nayakwadi et al. [84] reported a lipid peroxidation rate caused by the T-2 toxin [82], which was less pronounced compared to that of AFB1, ZEA, and OTA [85]. It is worth mentioning that ROS production at low levels or in short intervals may trigger an upregulation of antioxidant enzymes due to increased demands for detoxification [31]. However, oxidative stress is well-justified as a result of either the generation of a higher concentration of ROS or decreased production of antioxidants within the cells [86].
More recently, Wang et al. [87] provided cows with two levels of mycotoxin contaminated cottonseed (Level 1: AFB1 + 80.13 μg ZEA/daily/cow; Level 2: 40.16 μg AFB1 + 160.26 μg ZEA/daily/cow), exceeding the limits of the EU for an interval of 14 days. There were no significant alterations in the antioxidant enzymes and oxidative indices, except for a decrease in the gamma-glutamyl transpeptidase (GGT) within the lower level of contamination. GGT is considered an indicator of liver function in ruminants [88] and its modulation in serum portray that liver function was negatively affected by contaminated cottonseed, although it is unclear why the levels did not change significantly in the higher dosage. Authors have suggested that the lack of immense biochemical alterations were attributed to the short experimental period. Another study by Wang et al. [89] using the same experimental subjects, but this time with higher levels of AFB1 (20 or 40 μg AFB1/kg DMI; approximately 400 and 800 μg AFB1/day) for a shorter time schedule (seven days), reported that MDA concentrations in serum was significantly increased while SOD activity was suppressed in high contaminated levels. In conclusion, the results of these studies showed that AFB1 contamination in cow diets was able to induce an immediate oxidative imbalance, whilst higher contamination levels further portrayed significant importance relative to their effects.
Experimental studies by Xiong et al. [36,90] described an aflatoxicosis effect in late and early lactation stage of dairy cows (Table 4). The results of both trials were comparable, showing increased levels of MDA in serum while the activity of GSH-Px was suppressed. However, TAC, SOD activity, and the concentrations of IgA and IgG were further decreased even within the lower contamination level (20 μg AFB1/kg DMI) in the case of the long-term contamination experimental trial (nine weeks) in early lactating cows [36]. Discrepancies over time, like those reported, may be attributed to the accumulation of mycotoxins in the liver and kidneys of cows [91] compared to those contaminated for a shorter period of time. In addition, another substantial factor may be correlated to the imbalance of the antioxidant and immune systems, which are further burdened during the peripartum period; moreover, such toxicity finds the organism in a more unfavorable condition [90]. A more recent study by Elgioushy et al. [92] observed increased values of serum MDA and CAT activity while GSH-Px activity was decreased in cattle recharging a naturally contaminated diet with AFB1 (range from 5 to 20 ng/mL).
Another two in vivo studies in mid-lactating dairy cows were conducted by providing higher levels of aflatoxin (100 μg AFB1/kg DMI) in the TMR ration [34] or ingested through rumen canula [35]. In both studies, SOD concentration in serum was reported to be higher in aflatoxic animals compared to the control group ones, suggesting a response of cellular mechanism as a protection against the oxidative damage. On the other hand, in studies in dairy cows with lower levels of aflatoxins but for a longer contamination interval, SOD was found to be suppressed [36]. It seems that mycotoxin levels and duration of exposure may induce different effects on the antioxidant system. Specifically, it could be hypothesized that mycotoxin contamination induces a severe oxidative condition that in the first stage could trigger endogenous antioxidant mechanisms and increase enzyme activity, as observed by Weatherly et al. [34] and Sulzberger et al. [35], while longer contamination trials such as those by Xiong et al. [36] diminished the antioxidant defenses, resulting in the suppression of antioxidant enzymes.
Regarding the in vitro studies, a recent work by Pauletto et al. [93] on bovine hepatocytes reported a significant increase in MDA concentration in incubated cells with AFB1, while their transcriptional profile of antioxidant enzymes (CAT, SOD, GSH-Px) were not affected. Two comparable studies conducted in vitro in bovine mammary epithelia cells that were treated with DON (0.25 μg/mL) for 24 h showed an increase in the concentration of MDA while TAC and GSH portrayed a decrease [94,95]. More specifically, Wang et al. [94] reported a lower activity of the SOD enzyme and Zhang et al. [95] observed the same trend in transcript levels (SOD1 and SOD2). In both experimental trials, despite the negative oxidative status that was observed, immunomodulating genes related to pro-inflammatory responses were reported in higher expression levels, suggesting a cytokine storm. Finally, a study by Bernabucci et al. [96], which incubated the peripheral mononuclear blood cells in aflatoxins and fumonisin medium in cows, reported an increase in the levels of MDA concentration and downregulated the signaling levels of SOD and GPX1.
Taking into account the aforementioned reports, ruminants may be less susceptible to the negative effects of mycotoxins since death occurrence is rarely observed in adult animals, despite the exceeding levels administered in trials. On the other hand, the majority of the current knowledge supports the idea that severe oxidative stress is induced by mycotoxin contamination. These assumptions should aid us in reconsidering the “innocent” term relative to the minimal susceptibility of ruminants toward mycotoxins, since the toxicity is further transferred to humans through dairy products and meat consumption. Without overlooking the detrimental consequences of mycotoxin metabolites in animal foods, additional deleterious molecules may be present in the case of mycotoxin contaminated ruminants such as alkanes, MDA, and 4-hydroxy-2-(E)-Nonenal. More specifically, it has been confirmed that MDA can modify double-stranded DNA by the formation of amino-iminopropene crosslinks between the NH2 groups of a guanosine base and the NH2 group of the complementary cytosine base [97]. In addition, MDA also has carcinogenic properties, based on experimental studies on rats and mice [98]. Therefore, within Europe following Commission Regulation (EC) No. 165/2010, the industry should determine the milk for mycotoxin contamination. In future, corresponding rules and policies should be implemented for lipid peroxidation products given their well-documented disastrous consequences for consumer health.

4. Prevention Strategies and Detoxification Technologies for the Mitigation of Mycotoxins in Animal Diets

Diet contamination with mycotoxins is a global problem that leads to livestock illnesses, severe economic losses, and adverse human health effects. Apart from the fact that the peri-harvest strategies should be in agreement with the good agricultural practices, much attention has been paid to develop innovative detoxification methods during the recent decades. The efficiency of the above approaches generally depends on the initial contamination levels, the achieved inactivation rate, their regular application possibilities, their safety, and their cost [100,101,102].

4.1. Good Agricultural Practices

Plant selection or breeding programs for mycotoxin resistance, appropriate use of fungicides–insecticides, crop rotation, proper soil and irrigation management, transportation, and packaging are the most important preventive measures against the contamination of animal feedstuffs by mycotoxins [100,103]. Moreover, the selection of the optimal harvesting period and the avoidance of mechanical injury results in a reduction of fungal infection in the field and as a result, the mycotoxin levels are determined at low levels in the harvested crop [104]. Proper pest management and storage conditions (duration, temperature, humidity) and regular commodity inspection through an appropriate control strategy also minimizes the extent of contamination by mycotoxins. Insects and rodents could act as carriers of fungi spores leading to their excessive proliferation and spread [103,105] Rapid turnover of feed within the animal unit also reduces mycotoxin production, since less time is available for fungal growth and toxin production [100].

4.2. Physical Detoxification Techniques

In the case of moderate to light mycotoxin contamination, physical methods such as sorting, winnowing, washing, milling, and floating could contribute in reducing mycotoxin levels by removing the more heavily contaminated particles [106,107]. Furthermore, the subjection of crops to rapid drying immediately after harvesting significantly reduces their moisture level and intercepts fungal growth and proliferation [108]. Thermal treatment such as the high temperatures used in frying, roasting, toasting, and extrusion have promising effects on reducing the mycotoxin content of a feed [109]. Irradiation using medium or long wavelength UVA and UVB also remove mycotoxins without severe adverse effects on organoleptic properties, but the high cost of irradiation units and the safety concerns related with its application have prevented its regular use [110,111].

4.3. Chemical Detoxification Techniques

Acids, alkalis, organic acids, and oxidizing agents have already been used with the intention to modify the bioavailability of mycotoxins [112]. Reaction of mycotoxins with bases such as ammonia and sodium hydroxide, or ozone and hydrogen peroxide may also result in the structural changes of mycotoxins and lead to their transformation into other compounds, the toxicity of which should be assessed [107,109]. Parameters that should be taken into consideration before the application of a chemical detoxification method are their safety, cost, efficiency, and the extent to which the nutritional content or the organoleptic properties of the feed are negatively affected [100]. During the recent years, nanomaterials such as selenium-, zinc oxide-, or copper-nanoparticles have been used as mycotoxin binders, leading to their removal [113].

4.4. Biological Detoxification Techniques

Fungi causing mycoses can be separated into two major categories, namely primary and opportunistic pathogens [38]. Primary pathogens affect healthy organisms with competent immune systems, while opportunistic pathogens make use of a compromised immune system of the host [38]. Fungi can be transmitted vertically and horizontally into plants and crops. During the horizontal infection, fungal endophytes are contagiously spread through ascospores and this transmission can be inhibited by the application of certain fungicides. Vertical transmission of the endophytic hyphae into seeds and seedlings is associated with the transmission of the fungus from generation to generation and is also very important, since these hyphae cannot be controlled by fungicides, they are neither latent nor dormant, but physiologically active and comprise the reservoir from which infection and toxin biosynthesis are activated [114]. Biology-based methods are therefore developed and are generally considered as safe and efficient without negative implications on the sensory attributes of the treated material and on the environment. The strategies of using naturally existing microorganisms including bacteria and yeasts or bioactive materials such as enzymes or polypeptides that biodegrade mycotoxins and alleviate their toxic effects have gained ground during the recent decades. The first method consists of the development of nontoxigenic strains of fungi that preclude or decrease the growth of their closely related toxigenic strains through the principle of competitive exclusion [115]. These bio-control strains can be applied directly to soil, but the most effective way is by combining the desired strain with a carrier/substrate such as a small grain before planting that provides a competitive advantage against toxigenic fungi [116]. On the other hand, specific enzymes can also accelerate chemical reactions in an efficient way and biodegrade mycotoxins [101]. Parameters that affect the effectiveness of a biological detoxification method are the stability of these agents at a variety of external conditions, the ease of their production, the safety of the detoxification metabolites, and the economic feasibility of such methods [101].

4.5. Feed Additives

The adsorption and bio-inactivation of mycotoxins via ingested feed additives has been extensively studied in livestock. Several substances such as lucerne, zeolites, bentonite, and bleaching clays act as mycotoxin-binding agents and prevent intestinal adsorption of the toxin by the animal through its diet. In detail, the above additives form stable complexes with mycotoxins, resulting in a reduction of their bioavailability [50,117]. Their effectiveness is related to the structures of both the binders and the mycotoxins (charge distribution, polarity, pore size, surface area) [50]. Among the problems of this approach is the risk of decreasing dietary vitamins, amino acids, and mineral availability. In order to overcome these constraints, biomass that contains yeast, lactic acid bacteria, and conidia of Aspergillus is used as a second-generation binder by providing numerous potential sites for mycotoxin attachment and ensuring improved tolerance by the animals due to its nature [118]. Potential adsorbents should possess improved binding ability against a wide range of mycotoxins, high adsorption capability, and limited binding to nutrients [101].
Dietary supplementation with natural antioxidants significantly delays or inhibits feed oxidation and protects cellular membranes, proteins, lipids, and nucleic acids against the toxic effects of mycotoxins [119,120,121]. Many vitamins such as vitamins A, E, and C have the potential to act as free radical scavengers and alleviate the negative implications of oxidative stress. In brief, the antioxidant properties of vitamin A rely on the prevention of mutagenic epoxides from binding to DNA, the inhibition of toxic substances, and the increase in levels of antioxidant enzymes (GSH and GSH-Px) [121]. Vitamin C is a powerful antioxidant that acts as a scavenger of oxygen- and nitrogen-based free radicals contributing to a delay in the lipid peroxidation rate and prevention of the nitrozation of the target molecules and regulation of antioxidant enzymes [121]. Vitamin E is a potent chain-breaking antioxidant that is capable of scavenging ROS and terminating free-radical chain reactions [122].
Dietary inclusion of carotenoids (i.e., crocin) in the diet of mice restored normal levels of biochemical parameters in the liver and kidney that were deteriorated by mycotoxin patulin [123] and alleviated ZEN-induced toxicity [124]. In a study with mice, the ameliorative effect of curcumin on lipid peroxidation in the liver and kidney induced by aflatoxin were demonstrated [125]. Accordingly, in a study with pigs, phytic acid has been shown to exert beneficial effects on the small intestine (jejunum), alleviating the changes induced by the mycotoxins DON and FB1 and protecting cells against oxidative stress [63]. Finally, several minerals like zinc and selenium are capable of protecting against mycotoxins as shown in several studies [58,126,127]. Most notably, organic selenium and modified glucomannans exerted a protective effect against the antioxidant depletion of avian liver due to T-2 toxicity [58]. Zinc was able to reduce the cytotoxicity of OTA via inhibition of oxidative and DNA damage and via regulation of the expression of several zinc-associated genes [126].

5. The Biotransformation of Mycotoxins and Presence in Animal Products

The passage of mycotoxins or their metabolites into animal products through the contaminated diet is an issue of great importance for the consumers, but also the market. There is a variation in tissue deposition of the above toxins among farm animals that is attributed to differences in their absorption and metabolism. In general, the accumulation of mycotoxins and their metabolites in animal muscle tissues is low, often below detection limits due to their intense metabolism in the liver [128,129,130]. Blood, kidney, and liver contain higher levels of mycotoxins and their metabolites than muscles and adipose tissue. As a result, special attention should be given if these offal are consumed [129,131,132]. Human exposure to mycotoxins through the consumption of meat products could be a result of aging or other processes such as dry-curing and the application of mycotoxin contaminated spices (e.g., nutmeg, peppers, coriander, and paprika). Mold species belonging to the genus Penicillium and Aspergillus are usually isolated in cured, fermented, or ripened meats and contribute to the acquisition of the organoleptic properties of these products. On the other hand, development of toxigenic fungi poses a great hazard for human health related with mycotoxin synthesis on these substrates [133].
The carry-over of mycotoxins through egg consumption has also been examined. As shown, residues of aflatoxins and their metabolites were lower than the detection limits [134] or their determined levels were 5-fold lower (<1 μg/kg) than the maximum residue limits (MRL) set by the EU [135] in eggs produced by hens fed with diets contaminated with these mycotoxins. Aflatoxins were also detected in egg and chicken meat samples from Pakistan, but their levels were also lower than the above MRL; the highest concentrations of these mycotoxins were found in liver [136].
In general, multi-exposure of humans to mycotoxins via milk consumption is observed. AFM1 is the hydroxylated derivative of AFB1 and is the most usual mycotoxin determined in milk due to its resistance in heat. Its permissive levels are 0.05 μg/kg milk in EU and is related to carcinogenic and mutagenic properties. Apart from AFM1, aflatoxins M2, B1, B2, G1, G2, OTA, FB1, ZEA, or their metabolites are also found in milk samples. Although several factors affect mycotoxin biotransformation in milk such as their molecular weight and lipophilicity, diet (forage–concentrate ratio), feed intake, digestion rate, animal health and productivity, season, and environmental conditions, the carry-over of the majority of them is limited and does not negatively affect human health according to the literature [137,138]. As stated previously, rumen plays an important role as a barrier against various mycotoxins in milk-producing animals as a significant number of them are inactivated or metabolized into less toxic forms. However, some of them may pass the rumen unchanged or be converted into metabolites that retain toxicity (i.e., AFM1) and pose a risk for human health. During recent years, the co-existence of several mycotoxins in milk that could affect their toxicity due to additive or synergistic effects has also been examined [139]. At the same time, the carry-over of mycotoxins into milk is usually examined in healthy animals with an intact blood–milk barrier. However, various systemic diseases and mammary infections might alter the functionality of this barrier, and hence transmission rates may be higher in daily practice [140].
Lactation stage is a parameter that mainly appears to influence AFM1 levels in cow milk; samples from early lactation have 3–3.5-fold higher AFM1 content compared to that of late lactation [141]. This seasonal trend in the levels of mycotoxins in milk is possibly related with the prolonged storage required for cattle feeds at early lactation, providing favorable conditions for fungal growth [142]. AFM1 is mainly determined in the casein fraction of milk, resulting in 3-fold and 5-fold higher levels in soft and hard cheeses, respectively, compared to the milk from which they were produced [143]. On the other hand, fermentation during yoghurt production significantly decreased AFM1 levels as a result of low pH, the formation of organic acids, and the presence of Lactobacillus sp. [144].
The majority of the data that exist on the effects of the ingestion of mycotoxin contaminated diets on the quality characteristics of the derived products is for eggs. In poultry, turkeys and ducks are the most sensitive species to AF and when they are fed with AF contaminated diets, they produce small eggs of poor quality and pigmentation, possibly as an effect of fat deposition in the liver, which impairs lipid metabolism and pigment deposition in yolk [102,134]. At the same time, reduced values for shape index, color [145], shell thickness, and strength [135] were observed in laying hens fed aflatoxin contaminated diets. Egg weight, relative yolk weight, albumen height, and Haugh unit were also decreased in laying hens fed with AF and DON contaminated diets [146]. Feeding broilers with an OTA contaminated diet resulted in decreased dressing percentage, carcass fat content, and breast meat water holding capacity and increased liver relative weight and longer small intestine and caeca [147]. It can be concluded that animal product quality is of paramount importance, thus both prevention strategies and detoxification technologies should be implemented.

6. Conclusions

Mycotoxins, for decades, have been an important threat for the livestock and agriculture sector. The present study concluded that mycotoxin contamination induced severe oxidative stress responses on both monogastrics (poultry, swine) and ruminants (sheep, goats, cows), which may be related to the products’ oxidative stability and shelf life. In future, a more holistic approach should be implemented on the mycotoxin problem without focusing only on meeting the current legislation and regulations. Nowadays, the current health crisis, the COVID-19 pandemic, has changed our perspective on health issues to a more holistic point of view, espousing ever more the One Health Concept, recognizing the connection between people, animals, plants, and the environment.

Author Contributions

Conceptualization, A.C.P., E.T., E.Z., G.D., P.E.S., C.A.G., and K.F.; Writing-original draft preparation. A.M., E.G., S.T., M.Z., A.C.P., E.T., E.Z., G.D., P.E.S., C.A.G., and K.F.; Writing-review and editing, A.M., E.G., S.T., M.Z., A.C.P., E.T., E.Z., G.D., P.E.S., C.A.G., and K.F.; Visualization, A.M., E.G., S.T., M.Z., A.C.P., E.T., E.Z., G.D., P.E.S., C.A.G., and K.F.; Supervision and project administration, A.C.P., E.T., E.Z., G.D., P.E.S., C.A.G., and K.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vila-Donat, P.; Marín, S.; Sanchis, V.; Ramos, A. A review of the mycotoxin adsorbing agents, with an emphasis on their multi-binding capacity, for animal feed decontamination. Food Chem. Toxicol. 2018, 114, 246–259. [Google Scholar] [CrossRef] [Green Version]
  2. Agriopoulou, S.; Stamatelopoulou, E.; Varzakas, T. Advances in Occurrence, Importance, and Mycotoxin Control Strategies: Prevention and Detoxification in Foods. Foods 2020, 9, 137. [Google Scholar] [CrossRef]
  3. Ayofemi Olalekan Adeyeye, S. Aflatoxigenic fungi and mycotoxins in food: A review. Crit. Rev. Food Sci. Nutr. 2019, 60, 709–721. [Google Scholar] [CrossRef]
  4. Rushing, B.; Selim, M. Aflatoxin B1: A review on metabolism, toxicity, occurrence in food, occupational exposure, and detoxification methods. Food Chem. Toxicol. 2014, 124, 81–100. [Google Scholar] [CrossRef] [PubMed]
  5. Taniwaki, M.; Pitt, J.; Magan, N. Aspergillus species and mycotoxins: Occurrence and importance in major food commodities. Curr. Opin. Food Sci. 2018, 23, 38–43. [Google Scholar] [CrossRef] [Green Version]
  6. Patriarca, A.; Fernández Pinto, V. Prevalence of mycotoxins in foods and decontamination. Curr. Opin. Food Sci. 2017, 14, 50–60. [Google Scholar] [CrossRef]
  7. EFSA Panel on Contaminants in the Food Chain (CONTAM). Scientific Opinion on the Risk for Public and Animal Health Related to the Presence of Sterigmatocystin in Food and Feed. EFSA J. 2013, 11, 3254. [Google Scholar] [CrossRef]
  8. Ráduly, Z.; Szabó, L.; Madar, A.; Pócsi, I.; Csernoch, L. Toxicological and Medical Aspects of Aspergillus-Derived Mycotoxins Entering the Feed and Food Chain. Front. Microbiol. 2020, 10, 2908. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, L.; Dou, X.W.; Zhang, C.; Logrieco, A.; Yang, M.H. A Review of Current Methods for Analysis of Mycotoxins in Herbal Medicines. Toxins 2018, 10, 65. [Google Scholar] [CrossRef] [Green Version]
  10. Caceres, I.; Al Khoury, A.; El Khoury, R.; Lorber, S.; Oswald, I.P.; El Khoury, A.; Atoui, A.; Puel, O.; Bailly, J.-D. Aflatoxin Biosynthesis and Genetic Regulation: A Review. Toxins 2020, 12, 150. [Google Scholar] [CrossRef] [Green Version]
  11. Nieto, C.H.D.; Granero, A.M.; Zon, M.A.; Fernández, H. Sterigmatocystin: A Mycotoxin to Be Seriously Considered. Food Chem. Toxicol. 2018, 118, 460–470. [Google Scholar] [CrossRef] [PubMed]
  12. Vidal, A.; Mengelers, M.; Yang, S.; De Saeger, S.; De Boevre, M. Mycotoxin Biomarkers of Exposure: A Comprehensive Review. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1127–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Beuerle, T.; Benford, D.; Brimer, L.; Cottrill, B.; Doerge, D.; Dusemund, B.; Farmer, P.; Fürst, P.; Humpf, H.; Mulder, P.P.J. Scientific Opinion on Ergot Alkaloids in Food and Feed. EFSA J. 2012, 10, 2798. [Google Scholar]
  14. Hojnik, N.; Cvelbar, U.; Tavčar-Kalcher, G.; Walsh, J.L.; Križaj, I. Mycotoxin Decontamination of Food: Cold Atmospheric Pressure Plasma versus “Classic” Decontamination. Toxins 2017, 9, 151. [Google Scholar] [CrossRef]
  15. Berthiller, F.; Cramer, B.; Iha, M.H.; Krska, R.; Lattanzio, V.M.T.; MacDonald, S.; Malone, R.J.; Maragos, C.; Solfrizzo, M.; Stranska-Zachariasova, M.; et al. Developments in Mycotoxin Analysis: An Update for 2016–2017. World Mycotoxin J. 2018, 11, 5–32. [Google Scholar] [CrossRef] [Green Version]
  16. EFSA Panel on Contaminants in the Food Chain. Scientific Opinion on the Risks for Animal and Public Health Related to the Presence of Phomopsins in Feed and Food. EFSA J. 2012, 10, 2567. [Google Scholar] [CrossRef]
  17. Książkiewicz, M.; Wójcik, K.; Irzykowski, W.; Bielski, W.; Rychel, S.; Kaczmarek, J.; Plewiński, P.; Rudy, E.; Jędryczka, M. Validation of Diaporthe toxica resistance markers in European Lupinus angustifolius germplasm and identification of novel resistance donors for marker-assisted selection. J. Appl. Genet. 2020, 61, 1–12. [Google Scholar] [CrossRef] [Green Version]
  18. EFSA Panel on Contaminants in the Food Chain (CONTAM). Scientific Opinion on the Risks for Public and Animal Health Related to the Presence of Citrinin in Food and Feed. EFSA J. 2012, 10, 2605. [Google Scholar]
  19. Li, P.; Su, R.; Yin, R.; Lai, D.; Wang, M.; Liu, Y.; Zhou, L. Detoxification of Mycotoxins through Biotransformation. Toxins 2020, 12, 121. [Google Scholar] [CrossRef] [Green Version]
  20. Ingenbleek, L.; Sulyok, M.; Adegboye, A.; Hossou, S.E.; Koné, A.Z.; Oyedele, A.D.; Kisito, C.S.K.J.; Koreissi Dembélé, Y.; Eyangoh, S.; Verger, P.; et al. Regional Sub-Saharan Africa Total Diet Study in Benin, Cameroon, Mali and Nigeria Reveals the Presence of 164 Mycotoxins and Other Secondary Metabolites in Foods. Toxins 2019, 11, 54. [Google Scholar] [CrossRef] [Green Version]
  21. EFSA Panel on Contaminants in the Food Chain. Scientific Opinion on the Risks for Animal and Public Health Related to the Presence Of Alternariatoxins in Feed and Food. EFSA J. 2011, 10, 2407. [Google Scholar]
  22. Puntscher, H.; Kütt, M.L.; Skrinjar, P.; Mikula, H.; Podlech, J.; Fröhlich, J.; Marko, D.; Warth, B. Tracking emerging mycotoxins in food: Development of an LC-MS/MS method for free and modified Alternaria toxins. Anal. Bioanal. Chem. 2018, 410, 4481–4494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Wegulo, S. Factors Influencing Deoxynivalenol Accumulation in Small Grain Cereals. Toxins 2012, 4, 1157–1180. [Google Scholar] [CrossRef] [PubMed]
  24. Mannaa, M.; Kim, K.D. Influence of Temperature and Water Activity on Deleterious Fungi and Mycotoxin Production during Grain Storage. Mycobiology 2017, 45, 240–254. [Google Scholar] [CrossRef]
  25. Pinotti, L.; Ottoboni, M.; Giromini, C.; Dell’Orto, V.; Cheli, F. Mycotoxin Contamination in the EU Feed Supply Chain: A Focus on Cereal Byproducts. Toxins 2016, 8, 45. [Google Scholar] [CrossRef]
  26. Udomkun, P.; Wiredu, A.N.; Nagle, M.; Bandyopadhyay, R.; Müller, J.; Vanlauwe, B. Mycotoxins in Sub-Saharan Africa: Present situation, socio-economic impact, awareness, and outlook. Food Control 2017, 72, 110–122. [Google Scholar] [CrossRef]
  27. Fraeyman, S.; Croubels, S.; Devreese, M.; Antonissen, G. Emerging Fusarium and Alternaria Mycotoxins: Occurrence, Toxicity and Toxicokinetics. Toxins 2017, 9, 228. [Google Scholar] [CrossRef] [Green Version]
  28. Da Silva, E.O.; Bracarense, A.P.F.L.; Oswald, I.P. Mycotoxins and oxidative stress: Where are we? World Mycotoxin J. 2018, 11, 113–134. [Google Scholar] [CrossRef]
  29. Collin, F. Chemical Basis of Reactive Oxygen Species Reactivity and Involvement in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 95. [Google Scholar] [CrossRef] [Green Version]
  30. Dupré-Crochet, S.; Erard, M.; Nüβe, O. ROS production in phagocytes: Why, when, and where? J. Leukoc. Biol. 2013, 94, 657–670. [Google Scholar] [CrossRef]
  31. Schieber, M.; Chandel, N.S. ROS Function in Redox Signaling and Oxidative Stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Moussa, Z.; Judeh, Z.M.; Ahmed, S.A. Nonenzymatic Exogenous and Endogenous Antioxidants. In Free Radical Medicine and Biology; IntechOpen: Rijeka, Croatia, 2019. [Google Scholar]
  33. Yang, L.; Yu, Z.; Hou, J.; Deng, Y.; Zhou, Z.; Zhao, Z.; Cui, J. Toxicity and oxidative stress induced by T-2 toxin and HT-2 toxin in broilers and broiler hepatocytes. Food Chem. Toxicol. 2015, 87, 128–137. [Google Scholar] [CrossRef] [PubMed]
  34. Weatherly, M.E.; Pate, R.T.; Rottinghaus, G.E.; Roberti Filho, F.O.; Cardoso, F.C. Physiological responses to a yeast and clay-based adsorbent during an aflatoxin challenge in Holstein cows. Anim. Feed Sci. Technol. 2018, 235, 147–157. [Google Scholar] [CrossRef]
  35. Sulzberger, S.A.; Melnichenko, S.; Cardoso, F.C. Effects of clay after an aflatoxin challenge on aflatoxin clearance, milk production, and metabolism of Holstein cows. J. Dairy Sci. 2017, 100, 1856–1869. [Google Scholar] [CrossRef] [PubMed]
  36. Xiong, J.L.; Wang, Y.M.; Zhou, H.L.; Liu, J.X. Effects of dietary adsorbent on milk aflatoxin M1 content and the health of lactating dairy cows exposed to long-term aflatoxin B1 challenge. J. Dairy Sci. 2018, 101, 8944–8953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Malachová, A.; Sulyok, M.; Beltrán, E.; Berthiller, F.; Krska, R. Optimization and validation of a quantitative liquid chromatography–tandem mass spectrometric method covering 295 bacterial and fungal metabolites including all regulated mycotoxins in four model food matrices. J. Chromatogr. A 2014, 1362, 145–156. [Google Scholar] [CrossRef] [Green Version]
  38. Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [Green Version]
  39. Wu, Q.H.; Wang, X.; Yang, W.; Nüssler, A.K.; Xiong, L.Y.; Kuca, K.; Dohnal, V.; Zhang, X.J.; Yuan, Z.H. Oxidative stress-mediated cytotoxicity and metabolism of T-2 toxin and deoxynivalenol in animals and humans: An update. Arch. Toxicol. 2014, 88, 1309–1326. [Google Scholar] [CrossRef]
  40. Smith, J.E.; Anderson, R.A. Mycotoxins and Animal Foods; CRC Press: Boca Raton, FL, USA, 1991. [Google Scholar]
  41. Smith, J.E.; Lewis, C.W.; Anderson, J.G.; Solomons, G.L. Mycotoxins in Human Nutrition and Health; European Commission: Brussels, Belgium, 1994. [Google Scholar]
  42. Charoenpornsook, K.; Kavisarasai, P. Mycotoxins in animal feedstuff of Thailand. Curr. Appl. Sci. Technol. 2006, 6, 25–28. [Google Scholar]
  43. Adhikari, M.; Negi, B.; Kaushik, N.; Adhikari, A.; Al-Khedairy, A.A.; Kaushik, N.K.; Choi, E.H. T-2 mycotoxin: Toxicological effects and decontamination strategies. Oncotarget 2017, 8, 33933–33952. [Google Scholar] [CrossRef] [Green Version]
  44. Wang, X.; Wu, Q.; Wan, D.; Liu, Q.; Chen, D.; Liu, Z.; Matinez-Larranaga, M.R.; Martinez, M.A.; Anadon, A.; Yuan, Z. Fumonisins: Oxidative stress-mediated toxicity and metabolism in vivo and in vitro. Arch. Toxicol. 2016, 90, 81–101. [Google Scholar] [CrossRef] [PubMed]
  45. Assi, M. The differential role of reactive oxygen species in early and late stages of cancer. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017, 313, R646–R653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Young, I.S.; Woodside, J.V. Antioxidants and health and disease. J. Clin. Pathol. 2001, 54, 176–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Halliwell, B.; Whiteman, M. Measuring reactive species and oxidative damage in vivo and in cell culture: How should you do it and what do the results mean? Br. J. Pharmacol. 2004, 142, 231–255. [Google Scholar] [CrossRef] [Green Version]
  48. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.D.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  49. Mezes, M.; Barta, M.; Nagy, G. Comparative investigation on the effect of T-2 mycotoxin on lipid peroxidation and antioxidant status in different poultry species. Res. Vet. Sci. 1999, 66, 19–23. [Google Scholar] [CrossRef]
  50. Galvano, F.; Piva, A.; Ritieni, A.; Galvano, G. Dietary strategies to counteract the effects of mycotoxins: A review. J. Food Prot. 2001, 64, 120–131. [Google Scholar] [CrossRef]
  51. Chen, J.; Chen, K.; Yuan, S.; Peng, X.; Fang, J.; Wang, F.; Cui, H.; Chen, Z.; Yuan, J.; Geng, Y. Effects of aflatoxin B1 on oxidative stress markers and apoptosis of spleens in Broilers. Toxicol. Ind. Health 2016, 32, 278–284. [Google Scholar] [CrossRef]
  52. Shahid, A.R.; Sun, L.; Zhang, N.; Khalil, M.M.; Gao, X.; Ling, Z.; Zhu, L.; Khan, F.A.; Zhang, J.; Qi, D. Ameliorative Effects of Grape Seed Proanthocyanidin Extract on Growth Performance, Immune Function, Antioxidant Capacity, Biochemical Constituents, Liver Histopathology and Aflatoxin Residues in Broilers Exposed to Aflatoxin B1. Toxins 2017, 9, 371. [Google Scholar]
  53. Halliwell, B.; Chirico, S. Lipid peroxidation: Its mechanism, measurement, and significance. Am. J. Clin. Nutr. 1993, 57, 715S–725S. [Google Scholar] [CrossRef] [Green Version]
  54. Shridhar, M.; Suganthi, R.U.; Thammiaha, V. Effect of dietary resveratrol in ameliorating aflatoxin B1 -induced changes in broiler birds. J. Anim. Physiol. Anim. Nutr. 2014, 99, 1094–1104. [Google Scholar] [CrossRef] [PubMed]
  55. Eraslan, G.; Akdoúan, M.; Yarsan, E.; Þahündokuyucu, F.; Eþsüz, D.; Altintaþ, L. The Effects of Aflatoxins on Oxidative Stress in Broiler Chickens. Turk. J. Vet. Anim. Sci. 2005, 29, 701–707. [Google Scholar]
  56. Li, K.; Cao, Z.; Guo, Y.; Tong, C.; Yang, S.; Long, M.; Li, P.; He, J. Selenium Yeast Alleviates Ochratoxin A-Induced Apoptosis and Oxidative Stress via Modulation of the PI3K/AKT and Nrf2/Keap 1 Signaling Pathways in the Kidneys of Chickens. Oxidat. Med. Cell. Longev. 2020, 12, 143. [Google Scholar]
  57. Oskoueian, E. Cytoprotective effect of palm kernel cake phenolics against aflatoxin B1- induced cell damage and its underlying mechanism of action. BMC Complement. Altern. Med. 2015, 15, 392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Dvorska, J.E.; Pappas, A.C.; Karadas, F.; Speake, B.K.; Surai, P.F. Protective effect of modified glucomannans and organic selenium against antioxidant depletion in the chicken liver due to T-2 toxin-contaminated feed consumption. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2007, 145, 582–587. [Google Scholar] [CrossRef]
  59. Hussein, H.S.; Brasel, J.M. Toxicity, metabolism and impact of mycotoxins on humans and animals. Toxicology 2001, 167, 101–134. [Google Scholar] [CrossRef]
  60. Weaver, A.C.; See, M.T.; Hansen, J.A.; Kim, Y.B.; De Souza, A.L.P.; Middleton, T.F.; Kim, S.W. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ, health, and immune status during chronic exposure. Toxins 2013, 5, 1261–1281. [Google Scholar] [CrossRef] [Green Version]
  61. Thanh, B.V.L.; Lemay, M.; Bastien, A.; Lapointe, J.; Lessard, M.; Chorfi, Y.; Guay, F. The potential effects of antioxidant feed additives in mitigating the adverse effects of corn naturally contaminated with Fusarium mycotoxins on antioxidant systems in the intestinal mucosa, plasma, and liver in weaned pigs. Mycotoxin Res. 2016, 32, 99–116. [Google Scholar] [CrossRef]
  62. Sun, Y.; Park, I.; Guo, J.; Weaver, A.C.; Kim, S.W. 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, 177–183. [Google Scholar] [CrossRef]
  63. Da Silva, E.O.; Gerez, J.R.; Hohmann, M.S.N.; Verri, W.A.; Bracarense, A.P.F. Phytic acid Decreases Oxidative Stress and Intestinal Lesions Induced by Fumonisin B1 and Deoxynivalenol in Intestinal Explants of Pigs. Toxins 2019, 11, 18. [Google Scholar] [CrossRef] [Green Version]
  64. Taranu, I.; Marin, D.E.; Palade, M.; Pistol, G.C.; Chedea, V.S.; Gras, M.A.; Rotar, C. Assessment of the efficacy of a grape seed waste in counteracting the changes induced by aflatoxin B1 contaminated diet on performance, plasma, liver and intestinal tissues of pigs after weaning. Toxicon 2019, 162, 24–31. [Google Scholar] [CrossRef] [PubMed]
  65. Ren, Z.; Deng, H.; Deng, Y.; Liang, Z.; Deng, J.; Zuo, Z.; Hu, Y.; Shen, L.; Yu, S.; Cao, S. Combined effects of deoxynivalenol and zearalenone on oxidative injury and apoptosis in porcine splenic lymphocytes in vitro. Exp. Toxicol. Pathol. 2017, 69, 612–617. [Google Scholar] [CrossRef] [PubMed]
  66. Sorrenti, V.; Di Giacomo, C.; Acquaviva, R.; Barbagallo, I.; Barbagallo, M.; Galvano, F. Toxicity of ochratoxin A and its modulation by antioxidants: A review. Toxins 2013, 5, 1742–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Upadhaya, S.D.; Sung, H.G.; Lee, C.H.; Lee, S.Y.; Kim, S.W.; Jo, K.J.; Ha, J.K. Comparative study on the aflatoxin B1 degradation ability of rumen fluid from Holstein steers and Korean native goats. J. Vet. Sci. 2009, 10, 29–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Rodrigues, I. A review on the effects of mycotoxins in dairy ruminants. Anim. Prod. Sci. 2014, 54, 1155–1165. [Google Scholar] [CrossRef]
  69. May, H.D.; Wu, Q.; Blake, C.K. Effects of the Fusarium spp. mycotoxins fusaric acid and deoxynivalenol on the growth of Ruminococcus albus and Methanobrevibacter ruminantium. Can. J. Microbiol. 2000, 46, 692–699. [Google Scholar] [CrossRef]
  70. Gallo, A.; Giuberti, G.; Frisvad, J.C.; Bertuzzi, T.; Nielsen, K.F. 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, 3057–3111. [Google Scholar] [CrossRef]
  71. Guo, Y.; Xu, X.; Zou, Y.; Yang, Z.; Li, S.; Cao, Z. Changes in feed intake, nutrient digestion, plasma metabolites, and oxidative stress parameters in dairy cows with subacute ruminal acidosis and its regulation with pelleted beet pulp. J. Anim. Sci. Biotechnol. 2013, 4, 31. [Google Scholar] [CrossRef] [Green Version]
  72. Bell, A.W.; Burhans, W.S.; Overton, T.R. Protein nutrition in late pregnancy, maternal protein reserves and lactation performance in dairy cows. Proc. Nutr. Soc. 2000, 59, 119–126. [Google Scholar] [CrossRef] [Green Version]
  73. Jackson, M.J.; Pye, D.; Palomero, J. The production of reactive oxygen and nitrogen species by skeletal muscle. J. Appl. Physiol. 2007, 102, 1664–1670. [Google Scholar] [CrossRef] [Green Version]
  74. Kuhla, B.; Nürnberg, G.; Albrecht, D.; Görs, S.; Hammon, H.M.; Metges, C.C. Involvement of Skeletal Muscle Protein, Glycogen, and Fat Metabolism in the Adaptation on Early Lactation of Dairy Cows. J. Proteome Res. 2011, 10, 4252–4262. [Google Scholar] [CrossRef] [PubMed]
  75. Xue, Y.; Guo, C.; Hu, F.; Zhu, W.; Mao, S. Undernutrition-induced lipid metabolism disorder triggers oxidative stress in maternal and fetal livers using a model of pregnant sheep. FASEB J. 2020, 34, 6508–6520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Solfrizzo, M.; Gambacorta, L.; Visconti, A. Assessment of multi-mycotoxin exposure in Southern Italy by urinary multi-biomarker determination. Toxins 2014, 6, 523–538. [Google Scholar] [CrossRef] [PubMed]
  77. Huang, S.; Zheng, N.; Fan, C.; Cheng, M.; Wang, S.; Jabar, A.; Wang, J.Q.; Cheng, J.B. Effects of aflatoxin B1combined with ochratoxin A and/or zearalenone on metabolism, immune function, and antioxidant status in lactating dairy goats. Asian Australas J. Anim. Sci. 2018, 31, 505–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ozer, J.; Ratner, M.; Shaw, M.; Bailey, W.; Schomaker, S. The current state of serum biomarkers of hepatotoxicity. Toxicology 2008, 245, 194–205. [Google Scholar] [CrossRef] [PubMed]
  79. Son, Y.; Kim, S.; Chung, H.T.; Pae, H.O. Reactive Oxygen Species in the Activation of MAP Kinases. Methods Enzymol. 2013, 528, 27–48. [Google Scholar] [PubMed]
  80. Costa-Pereira, A.P. Regulation of IL-6-type cytokine responses by MAPKs. Biochem. Soc. Trans. 2014, 42, 59–62. [Google Scholar] [CrossRef] [PubMed]
  81. Mohamed, M.; Abd El-Hafeez, A.; Ibrahim, E.; Abd El Mola, A. Ameliorating effects of organic and inorganic mycotoxin binders on the performance of Ossimi sheep. Egypt. J. Sheep Goats Sci. 2019, 14, 33–48. [Google Scholar]
  82. Wang, J.; Lin, L.; Jiang, Q.; Huang, W. Effect of supplemental lactic acid bacteria on growth performance, glutathione turnover and aflatoxin B1 removal in lambs. Czech J. Anim. Sci. 2019, 64, 272–278. [Google Scholar] [CrossRef] [Green Version]
  83. Dasari, S.; Ganjayi, M.S.; Oruganti, L.; Balaji, H.; Meriga, B. Glutathione s-transferases detoxify endogenous and exogenous toxic agents-mini review. J. Dairy Vet. Anim. Res. 2017, 5, 157–159. [Google Scholar] [CrossRef] [Green Version]
  84. Nayakwadi, S.; Ramu, R.; Kumar Sharma, A.; Kumar Gupta, V.; Rajukumar, K.; Kumar, V.; Shirahatti, P.S.; Rashmi, L.; Basalingappa, K.M. Toxicopathological studies on the effects of T-2 mycotoxin and their interaction in juvenile goats. PLoS ONE 2020, 15, e0229463. [Google Scholar] [CrossRef]
  85. Schuster, A.; Hunder, G.; Fichtl, B.; Forth, W. Role of lipid peroxidation in the toxicity of T-2 toxin. Toxicon 1987, 25, 1321–1328. [Google Scholar] [CrossRef]
  86. Chaudhari, M.; Jayaraj, R.; Santhosh, S.R.; Rao, P.V.L. Oxidative damage and gene expression profile of antioxidant enzymes after T-2 toxin exposure in mice. J. Biochem. Mol. Toxicol. 2009, 23, 212–221. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, Q.; Zhang, Y.; Zheng, N.; Zhao, S.; Li, S.; Wang, J. The biochemical and metabolic profiles of dairy cows with mycotoxins-contaminated diets. PeerJ 2020, 8, e8742. [Google Scholar] [CrossRef] [PubMed]
  88. Osorio, J.S.; Trevisi, E.; Ji, P.; Drackley, J.K.; Luchini, D.; Bertoni, G.; Loor, J.J. Biomarkers of inflammation, metabolism, and oxidative stress in blood, liver, and milk reveal a better immunometabolic status in peripartal cows supplemented with Smartamine M or MetaSmart. J. Dairy Sci. 2014, 97, 7437–7450. [Google Scholar] [CrossRef] [Green Version]
  89. Wang, Q.; Zhang, Y.; Zheng, N.; Guo, L.; Song, X.; Zhao, S.; Wang, J. Biological System Responses of Dairy Cows to Aflatoxin B1 Exposure Revealed with Metabolomic Changes in Multiple Biofluids. Toxins 2019, 11, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Xiong, J.L.; Wang, Y.M.; Nennich, T.D.; Li, Y.; Liu, J.X. Transfer of dietary aflatoxin B1 to milk aflatoxin M1 and effect of inclusion of adsorbent in the diet of dairy cows. J. Dairy Sci. 2015, 98, 2545–2554. [Google Scholar] [CrossRef] [PubMed]
  91. Capraro, J.; Rossi, F. The effects of ochratoxin A on liver metabolism. Med. J. Nutr. Metab. 2012, 5, 177–185. [Google Scholar] [CrossRef]
  92. Elgioushy, M.M.; Elgaml, S.A.; El-Adl, M.M.; Hegazy, A.M.; Hashish, E.A. Aflatoxicosis in cattle: Clinical findings and biochemical alterations. Environ. Sci. Pollut. Res. 2020, 27, 35526–35534. [Google Scholar] [CrossRef]
  93. Pauletto, M.; Giantin, M.; Tolosi, R.; Bassan, I.; Barbarossa, A.; Zaghini, A.; Dacasto, M. Curcumin Mitigates AFB1-Induced Hepatic Toxicity by Triggering Cattle Antioxidant and Anti-inflammatory Pathways: A Whole Transcriptomic In Vitro Study. Antioxidants 2020, 9, 1059. [Google Scholar] [CrossRef]
  94. Wang, J.; Jin, Y.; Wu, S.; Yu, H.; Zhao, Y.; Fang, H.; Shen, J.; Zhou, C.; Fu, Y.; Li, R.; et al. Deoxynivalenol induces oxidative stress, inflammatory response and apoptosis in bovine mammary epithelial cells. J. Anim. Physiol. Anim. Nutr. 2019, 103, 1663–1674. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, J.; Wang, J.; Fang, H.; Yu, H.; Zhao, Y.; Shen, J.; Zhou, C.; Jin, Y. Pterostilbene inhibits deoxynivalenol-induced oxidative stress and inflammatory response in bovine mammary epithelial cells. Toxicon 2021, 189, 10–18. [Google Scholar] [CrossRef] [PubMed]
  96. Bernabucci, U.; Colavecchia, L.; Danieli, P.P.; Basiricò, L.; Lacetera, N.; Nardone, A.; Ronchi, B. Aflatoxin B1 and fumonisin B1 affect the oxidative status of bovine peripheral blood mononuclear cells. Toxicol. In Vitro 2011, 25, 684–691. [Google Scholar] [CrossRef]
  97. Papastergiadis, A.; Fatouh, A.; Jacxsens, L.; Lachat, C.; Shrestha, K.; Daelman, J.; Kolsteren, P.; Van Langenhove, H.; De Meulenaer, B. Exposure assessment of Malondialdehyde, 4-Hydroxy-2-(E)-Nonenal and 4-Hydroxy-2-(E)-Hexenal through specific foods available in Belgium. Food Chem. Toxicol. 2014, 73, 51–58. [Google Scholar] [CrossRef] [PubMed]
  98. Saieva, C.; Peluso, M.; Palli, D.; Cellai, F.; Ceroti, M.; Selvi, V.; Bendinelli, B.; Assedi, M.; Munnia, A.; Masala, G. Dietary and lifestyle determinants of malondialdehyde DNA adducts in a representative sample of the Florence City population. Mutagenesis 2016, 31, 475–480. [Google Scholar] [CrossRef] [PubMed]
  99. Pate, R.T.; Cardoso, F.C. Injectable trace minerals (selenium, copper, zinc, and manganese) alleviates inflammation and oxidative stress during an aflatoxin challenge in lactating multiparous Holstein cows. J. Dairy Sci. 2018, 101, 8532–8543. [Google Scholar] [CrossRef] [PubMed]
  100. Bryden, W.L. Mycotoxin contamination of the feed supply chain: Implications for animal productivity and feed security. Anim. Feed Sci. Technol. 2012, 173, 134–158. [Google Scholar] [CrossRef]
  101. Zhu, Y.; Hassan, Y.I.; 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. [Google Scholar] [CrossRef]
  102. Haque, M.A.; Wang, Y.; Shen, Z.; Li, X.; Saleemi, M.K.; He, C. Mycotoxin contamination and control strategy in human, domestic animal and poultry: A review. Microb. Pathog. 2020, 142, 104095. [Google Scholar] [CrossRef]
  103. Munkvold, G.P. Cultural and genetic approaches to managing mycotoxins in maize. Annu. Rev. Phytopathol. 2003, 41, 99–116. [Google Scholar] [CrossRef]
  104. Rachaputi, N.; Krosch, S.; Wright, G.C. Management practices to minimise pre-harvest aflatoxin contamination in Australian peanuts. Aust. J. Exp. Agric. 2002, 42, 595–605. [Google Scholar] [CrossRef]
  105. Hell, K.; Cardwell, K.F.; Setamou, M.; Poehling, H.M. The influence of storage practices on aflatoxin contamination in maize in four agroecological zones of Benin, West Africa. J. Stored Prod. Res. 2000, 36, 365–382. [Google Scholar] [CrossRef]
  106. Neme, K.; Mohammed, A. Mycotoxin occurrence in grains and the role of postharvest management as a mitigation strategies. A review. Food Control 2017, 78, 412–425. [Google Scholar] [CrossRef]
  107. He, J.; Zhou, T. Patented techniques for detoxification of mycotoxins in feeds and food matrices. Recent Pat. Food Nutr. Agric. 2010, 2, 96–104. [Google Scholar] [CrossRef] [PubMed]
  108. Lanyasunya, T.P.; Wamae, L.W.; Musa, H.H.; Olowofeso, O.; Lokwaleput, I.K. The risk of mycotoxins contamination of dairy feed and milk on smallholder dairy farms in Kenya. Pak. J. Nutr. 2005, 4, 162–169. [Google Scholar]
  109. Karlovsky, P.; Suman, M.; Berthiller, F.; De Meester, J.; Eisenbrand, G.; Perrin, I.; Oswald, I.P.; Speijers, G.; Chiodoni, A.; Recker, T.; et al. Impact of food processing and detoxification treatments on mycotoxin contamination. Mycotoxin Res. 2016, 32, 179–205. [Google Scholar] [CrossRef]
  110. Calado, T.; Venâncio, A.; Abrunhosa, L. Irradiation for mold and mycotoxin control: A review. Compr. Rev. Food Sci. Food Saf. 2014, 13, 1049–1061. [Google Scholar] [CrossRef] [Green Version]
  111. Wu, Q.; Kuča, K.; Humpf, H.U.; Klímová, B.; Cramer, B. Fate of deoxynivalenol and deoxynivalenol-3-glucoside during cereal-based thermal food processing: A review study. Mycotoxin Res. 2017, 33, 79–91. [Google Scholar] [CrossRef]
  112. Adebo, O.A.; Molelekoa, T.; Makhuvele, R.; Adebiyi, J.A.; Oyedeji, A.B.; Gbashi, S.; Adefisoye, M.A.; Ogundele, O.M.; Njobeh, P.B. A review on novel non-thermal food processing techniques for mycotoxin reduction. Int. J. Food Sci. Technol. 2021, 56, 13–27. [Google Scholar] [CrossRef]
  113. Abd-Elsalam, K.A.; Hashim, A.F.; Alghuthaymi, M.A.; Said-Galiev, E. Nanobiotechnological strategies for toxigenic fungi and mycotoxin control. In Food Preservation; Academic Press: Cambridge, MA, USA, 2017; pp. 337–364. [Google Scholar]
  114. Bacon, C.W.; Yates, I.E.; Hinton, D.M.; Meredith, F. Biological control of Fusarium moniliforme in maize. Environ. Health Perspect. 2001, 109, 325–332. [Google Scholar]
  115. Cleveland, T.E.; Dowd, P.F.; Desjardins, A.E.; Bhatnagar, D.; Cotty, P.J. United States Department of Agriculture—Agricultural Research Service research on pre-harvest prevention of mycotoxins and mycotoxigenic fungi in US crops. Pest Manag. Sci. 2003, 59, 629–642. [Google Scholar] [CrossRef] [PubMed]
  116. Dorner, J.W. Biological control of aflatoxin contamination of crops. J. Toxicol. Toxin Rev. 2004, 23, 425–450. [Google Scholar] [CrossRef]
  117. Jouany, J.P. Methods for preventing, decontaminating and minimizing the toxicity of mycotoxins in feeds. Anim. Feed Sci. Technol. 2007, 137, 342–362. [Google Scholar] [CrossRef]
  118. Huwig, A.; Freimund, S.; Käppeli, O.; Dutler, H. Mycotoxin detoxication of animal feed by different adsorbents. Toxicol. Lett. 2001, 122, 179–188. [Google Scholar] [CrossRef]
  119. Wu, Q.; Wang, X.; Nepovimova, E.; Wang, Y.; Yang, H.; Li, L.; Zhang, X.; Kuca, K. Antioxidant agents against trichothecenes: New hints for oxidative stress treatment. Oncotarget 2017, 8, 110708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Strasser, A.; Carra, M.; Ghareeb, K.; Awad, W.; Böhm, J. Protective effects of antioxidants on deoxynivalenol-induced damage in murine lymphoma cells. Mycotoxin Res. 2013, 29, 203–208. [Google Scholar] [CrossRef]
  121. Diplock, A.T.; Charleux, J.L.; Grozier-Willi, G.; Kok, F.J.; Rice-Evans, C.; Roberfroid, M. Functional food science and defence against reactive oxidative species. Br. J. Nutr. 1998, 80 (Suppl. 1), S77–S112. [Google Scholar] [CrossRef] [Green Version]
  122. Rock, C.L.; Jacob, R.A.; Bowen, P. Update on the biological characteristics of the antioxidant micronutrients: Vitamin C, vitamin E, and the carotenoids. J. Am. Diet. Assoc. 1996, 96, 693–702. [Google Scholar] [CrossRef]
  123. Boussabbeh, M.; Salem, I.B.; Belguesmi, F.; Neffati, F.; Najjar, M.F.; Abid-Essefi, S.; Bacha, H. Crocin protects the liver and kidney from patulin-induced apoptosis in vivo. Environ. Sci. Pollut. Res. 2016, 23, 9799–9808. [Google Scholar] [CrossRef]
  124. Salem, I.B.; Boussabbeh, M.; Neffati, F.; Najjar, M.F.; Abid-Essefi, S.; Bacha, H. Zearalenone-induced changes in biochemical parameters, oxidative stress and apoptosis in cardiac tissue: Protective role of crocin. Hum. Exp. Toxicol. 2016, 35, 623–634. [Google Scholar] [CrossRef]
  125. Verma, R.J.; Mathuria, N. Curcumin ameliorates aflatoxin-induced lipid-peroxidation in liver and kidney of mice. Acta Pol. Pharm. 2008, 65, 195–202. [Google Scholar] [PubMed]
  126. Zheng, J.; Zhang, Y.; Xu, W.; Luo, Y.; Hao, J.; Shen, X.L.; Yang, X.; Li, X.; Huang, K. Zinc protects HepG2 cells against the oxidative damage and DNA damage induced by ochratoxin A. Toxicol. Appl. Pharmacol. 2013, 268, 123–131. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, X.; Zuo, Z.; Zhao, C.; Zhang, Z.; Peng, G.; Cao, S.; Hu, Y.; Yu, S.; Zhong, Z.; Deng, J.; et al. Protective role of selenium in the activities of antioxidant enzymes in piglet splenic lymphocytes exposed to deoxynivalenol. Environ. Toxicol. Pharmacol. 2016, 47, 53–61. [Google Scholar] [CrossRef] [PubMed]
  128. Matrella, R.; Monaci, L.; Milillo, M.A.; Palmisano, F.; Tantillo, M.G. Ochratoxin A determination in paired kidneys and muscle samples from swines slaughtered in southern Italy. Food Control 2006, 17, 114–117. [Google Scholar] [CrossRef]
  129. Bailly, J.D.; Guerre, P. Mycotoxins in meat and processed meat products. In Safety of Meat and Processed Meat; Springer: New York, NY, USA, 2009; pp. 83–124. [Google Scholar]
  130. Dall’Asta, C.; Galaverna, G.; Bertuzzi, T.; Moseriti, A.; Pietri, A.; Dossena, A.; Marchelli, R. Occurrence of ochratoxin A in raw ham muscle, salami and dry-cured ham from pigs fed with contaminated diet. Food Chem. 2010, 120, 978–983. [Google Scholar] [CrossRef]
  131. Gareis, M.; Scheuer, R. Ochratoxin A in meat and meat products. Arch. Lebensm. 2000, 51, 102–104. [Google Scholar]
  132. Perši, N.; Pleadin, J.; Kovačević, D.; Scortichini, G.; Milone, S. Ochratoxin A in raw materials and cooked meat products made from FUM -treated pigs. Meat Sci. 2014, 96, 203–210. [Google Scholar] [CrossRef]
  133. Montanha, F.P.; Anater, A.; Burchard, J.F.; Luciano, F.B.; Meca, G.; Manyes, L.; Pimpão, C.T. Mycotoxins in dry-cured meats: A review. Food Chem. Toxicol. 2018, 111, 494–502. [Google Scholar] [CrossRef]
  134. Zaghini, A.; Martelli, G.; Roncada, P.; Simioli, M.; Rizzi, L. Mannanoligosaccharides and aflatoxin B1 in feed for laying hens: Effects on egg quality, aflatoxins B1 and M1 residues in eggs, and aflatoxin B1 levels in liver. Poult. Sci. 2005, 84, 825–832. [Google Scholar] [CrossRef]
  135. Jia, R.; Ma, Q.; Fan, Y.; Ji, C.; Zhang, J.; Liu, T.; Zhao, L. 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, 142–150. [Google Scholar] [CrossRef]
  136. Iqbal, S.Z.; Nisar, S.; Asi, M.R.; Jinap, S. Natural incidence of aflatoxins, ochratoxin A and zearalenone in chicken meat and eggs. Food Control 2014, 43, 98–103. [Google Scholar] [CrossRef]
  137. EFSA Panel on Contaminants in the Food Chain; Alexander, J.; Autrup, H.; Bard, D.; Carere, A.; Costa, L.G.; Cravedi, J.-P.; Di Domenico, A.; Fanelli, R.; Fink-Gremmels, J.; et al. Opinion of the scientific panel on contaminants in the food chain on a request from the commission related to zearalenone as undesirable substance in animal feed. EFSA J. 2004, 89, 1–35. [Google Scholar]
  138. EFSA Panel on Contaminants in the Food Chain; Alexander, J.; Autrup, H.; Bard, D.; Carere, A.; Costa, L.G.; Cravedi, J.-P.; Di Domenico, A.; Fanelli, R.; Fink-Gremmels, J.; et al. Opinion of the scientific panel on contaminants in food chain on a request from the commission related to fumonisins as undesirable substances in animal feed. EFSA J. 2005, 235, 1–32. [Google Scholar]
  139. Flores-Flores, M.E.; Lizarraga, E.; de Cerain, A.L.; González-Peñas, E. Presence of mycotoxins in animal milk: A review. Food Control 2015, 53, 163–176. [Google Scholar] [CrossRef]
  140. Fink-Gremmels, J. Mycotoxins in cattle feeds and carry-over to dairy milk: A review. Food Addit. Contam. Part A 2008, 25, 172–180. [Google Scholar] [CrossRef] [Green Version]
  141. Veldman, A.; Meijs, J.A.C.; Borggreve, G.J.; Heeres-van, D.T. Carry-over of aflatoxin from cows’ food to milk. Anim. Sci. 1992, 55, 163–168. [Google Scholar] [CrossRef]
  142. Becker-Algeri, T.A.; Castagnaro, D.; de Bortoli, K.; de Souza, C.; Drunkler, D.A.; Badiale-Furlong, E. Mycotoxins in bovine milk and dairy products: A review. J. Food Sci. 2016, 81, 544–552. [Google Scholar] [CrossRef] [Green Version]
  143. Ardic, M.; Karakaya, Y.; Atasever, M.; Adiguzel, G. Aflatoxin M1 levels of Turkish white brined cheese. Food Control 2009, 20, 196–199. [Google Scholar] [CrossRef]
  144. Govaris, A.; Roussi, V.; Koidis, P.A.; Botsoglou, N.A. Distribution and stability of aflatoxin M1 during production and storage of yoghurt. Food Addit. Contam. 2002, 19, 1043–1050. [Google Scholar] [CrossRef]
  145. Pandey, I.; Chauhan, S.S. Studies on production performance and toxin residues in tissues and eggs of layer chickens fed on diets with various concentrations of aflatoxin AFB1. Br. Poult. Sci. 2007, 48, 713–723. [Google Scholar] [CrossRef]
  146. Lee, J.T.; Jessen, K.A.; Beltran, R.; Starkl, V.; Schatzmayr, G.; Borutova, R.; Caldwell, D.J. Effects of mycotoxin-contaminated diets and deactivating compound in laying hens: 2. Effects on white shell egg quality and characteristics. Poult. Sci. 2012, 91, 2096–2104. [Google Scholar] [CrossRef] [PubMed]
  147. 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, 431–444. [Google Scholar] [CrossRef] [PubMed]
Table 1. Selected mycotoxins, producing fungal species, and contaminated food/feed.
Table 1. Selected mycotoxins, producing fungal species, and contaminated food/feed.
MycotoxinMycotoxin-Producing FungiFood/Feed MaterialReferences
AflatoxinsAspergillus flavusMaize, peanuts, wheat, rice, sorghum, pistachio, ground nuts, tree nuts (almonds, walnut, hazelnut, brazil nuts), cottonseed, spices (cumin, black pepper, chili pods/powder), dried fruits (figs, raising, currant, sultanas, plums, date, apricots), cereals, soybean, cocoa, milk, milk products, meat, feeds[1,2,3,4,5,6,25]
Aspergillus parasiticus, Aspergillus ochraceus, Aspergillus carbonarius,Maize, peanuts, brazil nuts, cocoa[3,5]
Aspergillus niger
OchratoxinsAspergillus ochrareusBarley, wheat, maize, cereals, dried vine fruits, wine, grapes, coffee, cocoa, cheese, feeds[1,2,5,6,25]
Aspergillus nigerCoffee, grapes, maize[5]
Aspergillus carbonariusMaize, cereals, dried vine fruits, wine, grapes, coffee, cocoa, cheese[2,5]
Penicillium verrucosumBarley, wheat, cereals, dried vine fruits, wine, grapes, coffee, cocoa, cheese, feeds[1,2,3,6]
Penicillium viridicatumcereals, dried vine fruits, wine, grapes, coffee, cocoa, cheese, feeds[1,2,6,25]
ZearaleoneFusarium graminearumWheat, maize, cereal product, barley, feeds[1,2,6,25]
Fusarium culmorum, Fusarium equiseti, Fusarium cerealis, Fusarium verticillioides, Fusarium incarnatumWheat, maize, cereal product, barley, rye[2,6]
FumonisinsFusarium verticillioides, Fusarium proliferatumMaize products, sorghum, asparagus, feeds[1,2,25]
T-2 and HT-2Fusarium langsethiae, Fusarium sporotrichioidesMaize, wheat, barley, oat, rye[2,6]
DeoxynivalenolFusarium graminearum, Fusarium culmorum, Fusarium cerealis, Fusarium sporotrichioides, Fusarium poae, Fusarium tricinctum, and Fusarium acuminatumMaize, wheat, barley, oat, cereal, cereal product, feeds[1,2,6]
NivalenolFusarium crookwellense, Fusarium poae, Fusarium nivale, Fusarium culmorum, and Fusarium graminearumMaize, wheat, barley[3,6]
SterigmatocystinAspergillus flavus, Aspergillus parasiticus, Aspergillus versicolor, Aspergillus nidulans and Aspergillus versicolorWheat, oats, ryes, barley, buckwheat, grain-based products, breakfast cereals, cooking oils, sorghum, maize on the cob, maize-based thickeners, maize syrup, polenta, tacos, tinned sweet maize, popcorn and maize snacks, cheese, nuts (peanut, hazelnuts), coffee beans, fresh fruits and sterilized fruits (grapes, plums, apples, pears, bananas and oranges), fruit juices (apple juices, blackcurrant juice and cherry juice), green vegetables and canned vegetables, beer, spices, animal feed[7,11,20,37]
Ergot alkaloidsClaviceps purpurea, Claviceps fusiformis, Claviceps africana, Neotyphodium spp.,Rye, rye-containing commodities, wheat, triticale, barley, millet, oat, grains, grass[2,14,24]
AlternariaAlternaria alternata, Alternaria tenuissima, Alternaria arborescensGrain based products, all cereal grains, fruit and fruit products, vegetables and vegetable products, oilseeds, beer, wine[2,6,24]
Table 2. Focus on European legislation and major regulations and recommendations regarding the maximum levels of mycotoxins in foods and feeds.
Table 2. Focus on European legislation and major regulations and recommendations regarding the maximum levels of mycotoxins in foods and feeds.
MycotoxinFoodstuffsMaximum Levels (μg/kg)EU Commission Regulations or Recommendations
Aflatoxins B1B1, B2, G1, G2M1
Cereal products24-165/2010
Maize and rice to be subjected to sorting or other physical treatment before human consumption or use as an ingredient in foodstuffs510 165/2010
Raw milk, heat-treated milk and milk for the manufacture of milk-based products--0.05165/2010
Feed raw materials (concerns feed with a moisture content of 12%)0.02--574/2011
Complementary and complete feeding stuffs, except: (A) compound feeds for dairy cattle and calves, dairy sheep and lambs, dairy goats and kids, piglets and young poultry, and (B) compound feeding stuffs for bovine animals (excluding dairy cattle and calves), sheep (excluding dairy sheep and lambs), goats (excluding dairy goats and goats) and pigs (excluding piglets) and poultry (except chickens) (concerns feed with a moisture content of 12%)0.01--574/2011
Compound feed for dairy cattle and calves, dairy sheep and lambs, piglets, dairy goats and kids and young poultry (concerns feed with a moisture content of 12%)0.005--574/2011
Compound feeding stuffs for bovine animals (excluding dairy cattle and calves), sheep (excluding dairy sheep and lambs), goats (excluding dairy goats and young goats) and pigs (excluding piglets) and poultry (excluding from young) (concerns feed with a moisture content of 12%)0.02--574/2011
Ochratoxin AUnprocessed cereals5594/2012
Feed raw materials Cereal products (concerns feed with a moisture content of 12%)0.252016/1319 *
Complementary and complete feed for pigs (concerns feed with a moisture content of 12%)0.052016/1319 *
Complementary and complete feed for poultry (concerns feed with a moisture content of 12%)0.12016/1319 *
Complementary and complete feed for dog and cats0.012016/1319 *
ZearalenoneUnprocessed cereals (not maize)1001126/2007
Unprocessed maize with the exception of unprocessed maize intended to be processed by wet milling3501126/2007
Feed raw materials Cereal products (concerns feed with a moisture content of 12%)22016/1319 *
Feed raw materials maize by-products (concerns feed with a moisture content of 12%)32016/1319 *
Compound feed for piglets, gilts (young sows), puppies, kittens, dogs and cats for reproduction (concerns feed with a moisture content of 12%)0.12016/1319 *
Compound feed adult dogs and cats other than for reproduction (concerns feed with a moisture content of 12%)0.22016/1319 *
Compound feed sows and fattening pigs (concerns feed with a moisture content of 12%)0.252016/1319 *
Compound feed calves, dairy cattle, sheep (including lamb) and goats (including kids) (concerns feed with a moisture content of 12%)0.52016/1319 *
FumonisinsUnprocessed maize, with the exception of unprocessed maize intended to be processed by wet milling40001126/2007
Raw materials: maize products (concerns feed with a moisture content of 12%)602016/1319 *
Compound feed for pigs, horses (Equidae), rabbits and pets (concerns feed with a moisture content of 12%)52016/1319 *
Compound feed for fish (concerns feed with a moisture content of 12%)102016/1319 *
Compound feed for poultry, calves (<4 months) and lambs and young goats (concerns feed with a moisture content of 12%)202016/1319 *
Complementary and complete feed for adult ruminants (>4 months) and mink animals (concerns feed with a moisture content of 12%)502016/1319 *
T-2 and HT-2 toxinCompound feed for cats0.052016/1319 *
DeoxynivalenolFeed materials, cereal products with the exception of maize (concerns feed with a moisture content of 12%) by-products82016/1319 *
Feed materials—Maize by-products (concerns feed with a moisture content of 12%)122016/1319 *
Compound feed for pigs (concerns feed with a moisture content of 12%)0.92016/1319 *
Compound feed for calves (<4 months), lambs, kids and dogs (concerns feed with a moisture content of 12%)22016/1319 *
Compound feed (concerns feed with a moisture content of 12%)52016/1319 *
CitrininFood supplements based on rice fermented with red yeast Monascus purpureus2000212/2014
* denotes Commission Recommendation (EU).
Table 3. Selected studies presenting the effects of mycotoxins on poultry and swine’s oxidative indices and antioxidant enzymes.
Table 3. Selected studies presenting the effects of mycotoxins on poultry and swine’s oxidative indices and antioxidant enzymes.
Animal SpeciesMycotoxin TestedLevelsOxidative IndicesAntioxidant EnzymesOther IndicesReferences
BroilersAflatoxin B1(1) 0.15 mg AFB1/kg
(2) 0.3 mg AFB1/kg
(3) 0.6 mg AFB1/kg
↑ MDA
↑ GSH
Spleen:
↓GSH-Px
↓GR
↓CAT
[51]
BroilersAflatoxin B11 mg AFB1/kg↑ MDALiver and serum:
↓CAT
↓ GSH-Px
↓ T-SOD
↓ GR
↓ GSTs
[52]
BroilersAflatoxin B11 ppm↑ MDA
↓ TAC
Serum:
↑ SOD
↓ CAT
↑ AST
↑ ALT
↓ Glucose
↑ Cholesterol
↑ Triglyceride
[54]
BroilersAflatoxin B1(1) 0.05 mg/kg
(2) 0.1 mg/kg
(3) 0.5 mg/kg
(4) 1.0 mg/kg
↑ MDA↓ SOD
↓ CAT
↓ G6PD
↓ GSH-Px
[55]
BroilersOchratoxin50 μg/kg OTAKidneys:
↑ MDA
↓ GSH
↓ TAC
↓ CAT
↓ SOD
↓ CAT (mRNA expression)
↓ SOD(mRNA expression)
↓ GSH-Px(mRNA expression)
[56]
Broilers (and broilers hepatocytes cells in vitro)T-2 toxin
HT-2 toxin
(1) 1 mg/kg T-2 + 0.167 mg/kg HT-2
(2) 2 mg/kg T-2 + 0.333 mg/kg HT-2
(3) 4 mg/kg T-2 + 0.667 mg/kg HT-2
Hepatocytes treated for 24 h with 10, 20, 50 and 100 nM of T-2 and HT-2 toxins
↑ MDA (Relative mRNA expression of in vivo and in vitro trials)
↑ GSH-Px
↑ CAT
↑ SOD
↑ALT
↑AST
[33]
BroilersT-2 toxin8.1 mg/kg↓ reduced glutathione↓ Se-GSH-Px [58]
Chicken (hepatocytes cells in vitro)Aflatoxin B15 μM↑ MDA↓SOD
↓CAT
↓ GR
↑IL1β
↑NFkB
↑TNF-α
[57]
Pigs (weaned)Deoxynivalenol
Zearaleone
(1) 0.8 mg DON/kg
(2) 3.1 mg DON/kg + 1.8 mg ZEA/kg
Plasma:
↑ MDA
Liver and plasma:
−GSH
↑ SOD in liver
↓ GPX2 gene expression in jejunum
[61]
PigsAflatoxins20 μg AF/kg−MDA ↑TNF-α[62]
PigsFumonisin B1
Deoxynivalenol
(1) 10 μM DON
(2) 70 μM FB1
(3) 10 μM DON + 70 μM FB1
↓ GSH
↑ MDA
↓ TAC (ABTS)
[63]
Pigs (weaned)Aflatoxins320 ppb pure AFB1↓ TACPlasma and organs:
↓ CAT
↓ SOD
↓ GSH-Px
[64]
Pigs (porcine splenic lymphocytes cells in vitro)Deoxynivalenol
Zearaleone
(1) 0.06, 0.3, 1.5,
and 7.5 μg/mL DON
(2) 0.08, 0.4, 2, and 10 μg/mL ZEA
(3) DON + ZEA at 0.06 and 0.08 μg/mL, 0.3 and 0.4 μg/mL, and 1.5 and 2 μg/mL respectively
↑ MDA
↓ GSH
↓ SOD
↓ CAT
↓ GSH-Px
[65]
↓ = significant decrease; ↑ = significant increase; − = no significant alternations.
Table 4. Selected studies presenting the effects of mycotoxins on the ruminants’ oxidative indices, antioxidant enzymes, and cellular function.
Table 4. Selected studies presenting the effects of mycotoxins on the ruminants’ oxidative indices, antioxidant enzymes, and cellular function.
Animal SpeciesMycotoxin TestedLevelsOxidative IndicesAntioxidant EnzymesOther IndicesNotesReferences
Dairy goatsAflatoxin B1 (AFB1)
Ochratoxin (OTA)
Zearaleone (ZEA)
50 μg AFB1/kg DMI
50 μg AFB1 + 100 μg OTA/kg DMI
50 μg AFB1 + 500 μg ZEA/kg DMI
50 μg AFB1 + 100 μg OTA + 500 μg ZEA/kg DMI
↓ TAC
↑ MDA
↓ SOD
↓ GSH-PX
↑ ALT
↑ ALP
↑ TBIL
↑ IL-6
↓ IgA
OTA + AFB1 more detrimental than ZEA + AFB1[77]
Goats (kids)T-2 toxin10 and 20 ppm↑ MDA
(Lipid peroxidation)
Liver, Intestines, Kidneys: ↑ CAT
↑ SOD
2–3 months old[84]
Sheep (Peripartum period)Aflatoxin B1 (AFB1)
Ochratoxin (OTA)
50 μg AFB1 + 100 μg OTA/kg DMI↓ TAC
↑ MDA
↓ CAT
↓ SOD
↓ GSH-PX
↓ TP
↓ ALB
↓ Chol
↑ ALT
↑ AST
↑ Urea
Lambs’ mortality[81]
LambsAflatoxin B1 (AFB1)100 μg AFB1/kg DMI↓ GSH Liver
↓ GSH Duodenal
↓ GSTs Liver and Duodenal
↓ GR Liver and Duodenal
2 months old[82]
CowsAflatoxin B1 (AFB1)
Zearaleone (ZEA)
Level 1: 20.08 μg
AFB1 + 80.13 μg ZEA/daily/cow
Level 2: 40.16 μg
AFB1 + 160.26 μg ZEA/daily/cow
-MDA-GSH-PX
-SOD
↓ GGT-14 days interval
-Late lactation
[87]
CowsAflatoxin B1 (AFB1)20 or 40 μg AFB1/kg DMI
(app. 20 kg DMI/day)
↑ TAC
↑ MDA (40 μg)
↓ SOD (40 μg) -7 days contamination interval
-Late lactation
[89]
CowsAflatoxin B1 (AFB1)20 or 40 μg AFB1/kg DMI
(app. 17 kg DMI/day)
↑ MDA↓ GSH-Px -7 days contamination interval
-Late lactation
[90]
CowsAflatoxin B1 (AFB1)20 μg AFB1/kg DMI
(app. 24 kg DMI/day)
↓ TAC
↑ MDA
↓ SOD
↓ GSH-Px
↓ IgG
↓ IgA
-Early lactation
-9 Weeks contamination interval
[36]
CowsAflatoxin B1 (AFB1)5–20 ng/mL
(TLC assay)
↑ MDA↑ CAT
↓ GSH-Px
↓ Total protein
↑ ALT
↑ AST
↑ ALP
↑ Creatine
Naturally contaminated feeds[92]
CowsAflatoxin B1 (AFB1)100 μg AFB1/kg DMI
(21.9–23.4 kg DMI/day)
↑ SOD↑ Glucose-7 days contamination interval
-Mid-lactation
[34]
CowsAflatoxin B1 (AFB1)100 μg AFB1/kg DMI
(21.4–22.8 kg DMI/day)
↑ SOD -3 days contamination interval
-Mid-lactation
-ingested through rumen canula
[35]
CowsAflatoxin B1 (AFB1)100 μg AFB1/kg DMI
(24.9 kg DMI/day)
-SOD
-GPX
↑ GPX1 (1)
-Chol
-Albumin
-BUN
↑ NFkB (1)
-Gene expression in Liver
-Mid-late lactation
-3 days contamination interval
[99]
Cows
(bovine fetal hepatocytes cells in vitro)
Aflatoxin B1 (AFB1)3.6 μM AFB1 in 6 × 103 hepatocytes↑ MDA-GSH-Px
-CAT
-SOD
transcriptional profiles using RNA -seq[93]
Cows
(bovine mammary epithelia cells in vitro)
Deoxynivalenol (DON)Cells were treated with DON (0.25 μg/mL) for 24 h↓ TAC
↑ MDA
↓ GSH
↓ SOD↑ NFkB
↑ MyD88
↑ TNF-α
↑ IL-1b
↑ IL-6
↑ IL-8
-Higher cells’ apoptotic rate[94]
Cows
(bovine mammary epithelia cells in vitro)
Deoxynivalenol (DON)Cells were treated with DON (0.25 μg/mL) for 24 h↓ TAC
↑ MDA
↓ GSH
↓ SOD1 (expres.)
↓ SOD2 (expres.)
↑ NFkB
↑ COX-2
↑ iNOS
↑ IL-1b
↑ IL-6
↑ IL-8
↑ TNF-α (Protein)
-Incubated for 9 h.
-Decreased cell viability and proliferation
[95]
Cows in vitro
(Peripheral blood mononuclear cells)
Aflatoxin B1 (AFB1)
Fumonisin B1 (FB1)
0, 5, 20 μg/mL AFB1
0, 35, 70 μg/mL FB1
↑ MDA↓ SOD (expres.)
↓ GPX1 (expres.) AFB1 5 μg
↓ GPX1 (expres.) FB1
2- and 7-days incubation[96]
↓ = significant decrease; ↑ = significant increase; − = no significant alternations.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mavrommatis, A.; Giamouri, E.; Tavrizelou, S.; Zacharioudaki, M.; Danezis, G.; Simitzis, P.E.; Zoidis, E.; Tsiplakou, E.; Pappas, A.C.; Georgiou, C.A.; et al. Impact of Mycotoxins on Animals’ Oxidative Status. Antioxidants 2021, 10, 214. https://doi.org/10.3390/antiox10020214

AMA Style

Mavrommatis A, Giamouri E, Tavrizelou S, Zacharioudaki M, Danezis G, Simitzis PE, Zoidis E, Tsiplakou E, Pappas AC, Georgiou CA, et al. Impact of Mycotoxins on Animals’ Oxidative Status. Antioxidants. 2021; 10(2):214. https://doi.org/10.3390/antiox10020214

Chicago/Turabian Style

Mavrommatis, Alexandros, Elisavet Giamouri, Savvina Tavrizelou, Maria Zacharioudaki, George Danezis, Panagiotis E. Simitzis, Evangelos Zoidis, Eleni Tsiplakou, Athanasios C. Pappas, Constantinos A. Georgiou, and et al. 2021. "Impact of Mycotoxins on Animals’ Oxidative Status" Antioxidants 10, no. 2: 214. https://doi.org/10.3390/antiox10020214

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop