Next Article in Journal
Identification of Fungi in Flaxseed (L. usitatissimum L.) Using the ITS1 and ITS2 Intergenic Regions
Previous Article in Journal
An Assessment of the Lactic Acid-Producing Potential of Bacterial Strains Isolated from Food Waste
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Deoxynivalenol: An Overview on Occurrence, Chemistry, Biosynthesis, Health Effects and Its Detection, Management, and Control Strategies in Food and Feed

1
Applied Microbiology Laboratory, Department of Forestry, North-Eastern Regional Institute of Science and Technology, Nirjuli 791109, India
2
CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC 3125, Australia
3
Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi 221005, India
4
Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara 144411, India
5
Faculty of Agricultural Sciences, GLA University, Mathura 281406, India
6
Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, Mumbai 400019, India
7
Centre for Protected Cultivation Technology, ICAR-Indian Agricultural Research Institute, Pusa Campus, New Delhi 110012, India
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microbiol. Res. 2022, 13(2), 292-314; https://doi.org/10.3390/microbiolres13020023
Submission received: 8 April 2022 / Revised: 21 May 2022 / Accepted: 27 May 2022 / Published: 1 June 2022
(This article belongs to the Topic Microbiology Metabolomics)

Abstract

:
Mycotoxins are fungi-produced secondary metabolites that can contaminate many foods eaten by humans and animals. Deoxynivalenol (DON), which is formed by Fusarium, is one of the most common occurring predominantly in cereal grains and thus poses a significant health risk. When DON is ingested, it can cause both acute and chronic toxicity. Acute signs include abdominal pain, anorexia, diarrhea, increased salivation, vomiting, and malaise. The most common effects of chronic DON exposure include changes in dietary efficacy, weight loss, and anorexia. This review provides a succinct overview of various sources, biosynthetic mechanisms, and genes governing DON production, along with its consequences on human and animal health. It also covers the effect of environmental factors on its production with potential detection, management, and control strategies.

1. Introduction

Natural contaminants in food and feed are significant sources of human and animal health concerns [1,2,3]. Mycotoxins are deadly substances produced by fungi that flourish in food or feed, and they pose a major health risk to humans and animals [4,5,6,7,8,9]. When mycotoxins enter the bodily system, they are well-known for causing serious health problems in humans and animals, and they exhibit their effects in a variety of ways. Ingesting mycotoxin-contaminated food or animal feed has been shown to cause carcinogenic, mutagenic, teratogenic, and immunosuppressive consequences [10]. Aspergillus, Fusarium, and Penicillium are the most common mycotoxin-producing fungal genera. Contamination with mycotoxin is a global problem, but it is exacerbated in warm, humid areas that promote fungal growth and mycotoxin synthesis. Contamination with mycotoxins has a financial impact on agriculture and the food industry. Aside from the financial costs of mycotoxin contamination of crops and food products, additional significant mycotoxin concerns include human and animal health difficulties, reduced agricultural production, and the recall and disposal of mycotoxin-infected commodities [11].
Deoxynivalenol (DON) is a prevalent mycotoxin identified in cereals around the world. It is a type-B trichothecene that Fusarium graminearum and Fusarium culmorum typically produce [12,13,14]. It is also known as vomitoxin because of its emetic effects in pigs and gastrointestinal complaints in people [15]. DON can also be found in pastures and silages, as well as cereals such as wheat, maize, barley, rye, oat, and safflower seed. Asia, Africa, America, Europe, and the Middle East are all affected by DON pollution [16]. DON is a naturally occurring foodborne mycotoxin that is easily caused by environmental changes and is typically present in grains during the preharvest, processing, drying, and storage processes (e.g., temperature, humidity). DON is also extremely thermostable, withstanding temperatures ranging from 170 to 350 °C (no reduction detected after 30 min at 170 °C treatment), rendering it a common dietary pollutant in mycotoxins pollution [15].
This article provides an overview of the main sources, chemistry and biosynthetic pathway, genes responsible for DON occurrence in food and feed, as well as the mechanism of toxicity and health effects on humans and animals, considering the numerous publications describing the toxicological consequences of mycotoxins. It also highlights the effect of environmental factors on DON production as well as discusses the management and control strategies with special emphasis on masked mycotoxins.

2. Major Source of Deoxynivalenol

DON is a naturally occurring metabolite produced by the fungi of the Fusarium genus, especially Fusarium graminearum, Fusarium crookwellense, and Fusarium culmorum, which contaminate the food and feed globally [17,18]. The fungi growth is enhanced by mild temperatures and high humidity during flowering and maturation periods [19]. Other environmental factors that affect growth and toxin accumulation are water activity (aw), pH, and nutrient composition [20]. The most commonly infected food groups are cereals like wheat [21], maize [22], barley [23], rice [22], oats [24], and their products viz, breakfast cereals [25], infant cereals, meals, feed, and baby mix. Fusarium head blight (FHB) infection and DON contamination are dependent on agricultural practices such as crop rotation, tillage, fungicide application, FHB resistant varieties, and climatic factors such as spring rainfalls and warm temperatures that promote the infection and DON formation on cereal crops [26]. The most common disease caused by Fusarium graminearum in cereal crops like wheat and barley is FHB or scab, which causes not only economic loss but also safety concerns related to mycotoxin (DON) accumulation in products due to its stability [27]. The fungus grows as mycelia which develop into perithecia that release ascospores which are then carried by wind or rain to the crop plants [28]. DON suppresses protein synthesis by binding to the 60S ribosomal subunit’s peptidyl transferase protein RPL3 and two F. graminearum strains can be distinguished by their production of 3-ADON or 15-ADON, which differ in the position of the acetyl group [29]. Fusarium culmorum is the major species that causes FHB in cereal crops, such as durum wheat, triticale, rye, and bread wheat in Europe, thus contaminating the grains with DON [30]. Fusarium crookwellense was responsible for causing infection and producing DON in oat kernels [31]. Fusarium cerealis has been known to cause FHB and DON accumulation in durum wheat [32]. Another strain causing the FHB and DON accumulation is Fusarium equiseti, which has also been studied to cause infection in wheat in South Africa [33]. Some of the species that have been recently reported to cause infection are F. verticillioides, F. poae, F. proliferatum, F. subglutinans, and F. temperatum [34].

3. Chemistry and Biosynthesis of Deoxynivalenol

DON (3,7,15-trihydroxy-12,13-epoxytrichothec-9-en-8-one) is a trichothecene mycotoxin present mainly in cereals and cereal-based products [35,36]. DON has a double bond with oxygen at carbon number 8 in its molecular structure, making it a type B trichothecene [37]. In eukaryotic cells, DON is a potent inhibitor of protein synthesis [38]. It is hazardous to human and animal wellbeing, as well as playing a key role in plant infection. DON inhibits protein synthesis by binding to peptidyl transferase and mediates oxidative stress-induced DNA damage and apoptosis. DON toxicity in humans and livestock causes anorexia, malnutrition, gastroenteritis, endotoxemia, and even shock-like death [36].
DON is made by combining three mevalonate units derived from trans-farnesyl pyrophosphate (FPP), which are then cyclized to form tricodiene, a precursor to trichothecenes. DON biosynthesis is governed by fifteen genes distributed over three chromosomes that code for the enzymes and regulatory proteins involved [39]. The remaining loci include TRI1-TRI16 and TRI101, while one locus contains a set of 12 TRI genes. Trichothecene synthase, which is encoded by the TRI5 gene, catalyzes the reaction that produces trichothecene, which is followed by nine reactions catalyzed by enzymes encoded by the TRI4, TRI101, TRI11, and TRI3 genes, which produce various DON precursors. An acidic pH is a significant environmental element that encourages DON biosynthesis. The accumulation of ammonium is caused by the absorption of available nitrogen-containing compounds in food or the culture medium, which causes the pH to decrease. The TRI5 gene product, which is responsible for FPP cyclization and the production of tricodiene, a precursor to DON, is activated in this state. DON biosynthesis is also induced by plant defense mechanisms in response to fungi infection. DON is produced by F. graminearum, which causes fungi to develop hyphae and spread from an infected branch to a healthy branch. In addition, the toxin inhibits the thickening of plant cell walls, which will serve as a deterrent to fungal invasion [39].

4. Genes Responsible for Deoxynivalenol Production

All the TRI genes involved in trichothecene biosynthesis have been identified in Fusarium graminearum and Fusarium sporotrichioides. Except for TRI1, TRI16, and TRI101, the other TRI genes are included in the largest TRI gene cluster [40]. The biosynthesis of the mycotoxin DON in Fusarium graminearum is affected by several host and environmental factors and is controlled by two pathway-specific transcription factors, TRI6 and TRI10. The TRI6 binding site in the promoters of TRI genes has formerly been recognized and characterized. TRI10 deletion in F. graminearum suppresses the expression of the TRI gene, despite the fact that its exact function is unclear. However, how the transcription factors Tri6 and TRI10 are designed to regulate the expression of other TRI genes is unknown (Jiang et al., 2016). The cyclic adenosine monophosphate (cAMP) signaling pathway, as well as all three MAP kinase pathways, is required for DON biosynthesis in F. graminearum [41,42]. Extracellular signals are likely to activate TRI6 and TRI10, which regulate TRI gene expression, by any of these key signaling pathways. Extracellular signals are likely to activate TRI6 and TRI10, which regulate TRI gene expression, by any of these key signaling pathways [38]. In addition, the sequenced F. graminearum strain PH-1 lacks functional TRI13 and TRI7, preventing DON development and Nivalenol production (NIV). TRI5 is a key cluster gene that catalyzes the formation of trichodiene from farnesyl pyrophosphate, the first step in trichothecene biosynthesis [40].

5. Effects of Environmental Factors on Deoxynivalenol Production

The environmental factors that primarily affect mycotoxin production are temperature, moisture, water activity (aw), substrate oxygen levels, physical damage, competition, and the presence of fungal spores [35,43]. The growth of the fungi, Fusarium, and its mycotoxin, DON, is dependent on weather conditions, such as high intensity warm and wet conditions and heavy precipitation, which makes the transfer of macroconidia to the upper parts of the plant easier through raindrops. The optimum temperature conditions are 10–25 °C during flowering and humidity greater than 85–90% [44]. High to extreme humidity during the growth and harvesting period enhances the growth of mold and hence the production of mycotoxin [35]. It has been shown by Pascari et al. [26] that warm temperatures and rainfall promote the growth and consequently mycotoxin formation in cereals. The other factors that affect mycotoxin production are aw and incubation time, and as reported by Han et al. [45], these factors affected the DON production through TRI5 gene expression under optimum conditions of 20–30 °C, 0.95–0.98 aw, and 7–28 days of incubation. Rainfall positively affected the growth of mold and DON production as temperature and humidity are important factors for growth and flowering [46]. Ramirez et al. [47] studied the effect of temperature, aw, and incubation time and observed that growth was optimum at 0.99 aw and 25 °C, and the growth was negatively affected when the aw of the medium was reduced. A temperature of 22 °C after 35 days of incubation time also favored the DON production, as shown by Martins and Martins [48].
The environmental factors affect not only the growth of mold but also other physiological processes that are important for survival and competition, such as the expression of hydrolytic enzymes playing a crucial role in a fungal establishment on a substrate [49]. DON production was maximum at 0.98 aw, 30 °C, and 400–800 ppm of CO2 [50]. It was also observed that the DON concentration was maximum at 0.97 aw and 30 °C [51]. The favorable condition for DON production was 25 °C and 0.98–0.99 aw, as observed by Rybecky et al. [52]. Further, the optimum growth of the fungus was observed at 0.99 aw and the growth decreased as aw was lowered from 0.97 to 0.94 [53]

6. Occurrence in Food and Feed, including Masked Mycotoxins

The most infected foodstuffs are cereals such as wheat, maize, barley, rice, oats, and their products viz; breakfast cereals, infant cereals, meals, feed, and baby mix. FHB infection and DON contamination are dependent on agricultural practices, such as crop rotation, tillage, fungicide application, FHB resistant varieties, and climatic factors, as spring rainfalls and warm temperatures promote the infection and DON formation on cereal crops [26]. In the real world, DON is always co-occurring with related metabolites, such as modified or masked versions and related molecules like Culmorin (CUL). CUL is classified as an “emerging mycotoxin” as it has yet to be controlled by food safety authorities, despite the fact that it has been found in a variety of foods, sometimes in significant amounts [54]. CUL contamination levels and natural occurrence have been linked to DON occurrence and levels, with CUL concentrations up to three times greater than DON in naturally contaminated samples [55,56]. Although CUL is mildly hazardous; there have been few studies on the possible health risks of CUL in food products; whether alone or in combination with DON [57,58]. As a result, the high frequencies and amounts of CUL and related compounds in cereal-based raise concerns regarding their co-occurrence with disguised DON mycotoxins. Contamination of DON in wheat and its by-products poses a significant danger to public health. The occurrence of DON in foods and feeds (Table 1) is a major concern for the food industry around the world.

7. Mechanism of Toxicity

Consumption of DON contaminated food and feed is a potential risk to the health of both humans and animals. DON is the most reported mycotoxin in cereals and is linked with gastroenteritis and immune disorders [119,120]. Exposure to the toxin can result in numerous problems such as digestive problems, feed refusal, diarrhea, reproductive problems, nutrient malabsorption, increased incidence of diseases, and endocrine disruption [121,122,123]. DON also causes oxidative stress due to the production of free radicals, which then damage DNA and the cell membrane. It also causes degradation of the ribosome, inducing ribotoxic stress, inhibiting protein synthesis, and ultimately apoptosis [121,124,125]. Studies have proven that DON is associated with the rise of reactive oxygen species, triggering lipid peroxidation and hepatoxicity as the liver is the main organ to get affected by oxidative stress [122,126,127,128]. Exposure to a high dose of DON, even for a short period, causes gastrointestinal problems in both humans and animals.
At doses as low as 25 ng/mL, DON-induced cytokine and chemokine production can be generated in human blood monocytes [129]. Induction of these mediators, like endotoxin, may play a role in DON-induced anorexia. DON inhibits intestinal cell development and is absorbed via the intestinal epithelium via simple diffusion [130]. DON activated p38 ERK and JNK as well as disrupted intestinal permeability at levels equivalent to those reported in nature. DON suppresses the activity of numerous intestinal transporters [131].

8. Effects on Human Health

DON causes hematopoietic progenitor cells less harm in vitro than T-2 toxin [132]. Human platelet progenitors (CFU-MK) are cytotoxic to DON; however, human red blood cell progenitors (CFU-GM) are not (BFU-E). In CD34+ cells, DON does not cause DNA fragmentation or annexin-V binding, both of which are apoptotic markers [133]. In addition, Bensassi et al. [134] studied DON toxicity in human colon cancer cells (HT 29), finding that the toxin caused DNA breakage as well as p53 and caspase-3 activation. DON causes direct DNA damage, according to scientists, and might be categorized as a genotoxic agent capable of causing apoptotic cell death.
DON has been linked to gastroenteritis in humans, but its long-term effects are unknown [135]. DON directly suppresses protein synthesis, but it also has an indirect effect on DNA and RNA synthesis, inflammatory responses (ribotic stress response), and neurological functioning [136]. Two species typically used to evaluate DON toxicity include the pig, a highly sensitive food-producing animal with economic ramifications, and the mouse, a model widely used in human disease and safety investigations [136].

9. Effects on Animal Health

The sensitivity of the DON varies in different species, age groups, and sex of the animals. Hogs are the most sensitive to DON contamination, along with the ruminants, poultry, cats, dogs, and rodents [137,138]. The qualitative and quantitative production rates of the egg, meat, and milk get reduced by the DON contamination. The DON concentration in the range of 0.1–2 µg/mL can inhibit the protein synthesis related to the lymphocytes and fibroblasts due to cytotoxicity [139]. In addition, DON contamination results in weak offspring, dead birth, stiff hog, female pig’s abortion, reduced quality and quantity of the produced eggs in poultry, as well as decreased production performance in cattle [140]. Further, the increased rate of embryo deformity in chicken and chromosomal aberration due to DON contamination has been reported [141]. In cows, a slight reduction in the consumption of the feed was reported when the DON quantity increased from 1.5 to 6.4 mg/kg [142]. In hogs, the absolute feed refusal was found at 12 mg/kg of DON, and at 20 mg/kg of DON, they started vomiting. Compared to hogs, chickens are less sensitive to DON and the reduced weight gain and feed refusal were observed when the feed concentration of DON increased up to 16–20 mg/kg [143].
The estimated mean dietary concentration of the DON and its derivatives range between 64.2–996 µg/kg for lactating cows and beef sheep. Ruminants are less sensitive to DON as compared to poultry. In cows, poultry, and hogs, the transmission of the DON from their feed to foodstuffs was reported. The concentration of DON at 9 mg/kg of feed can negatively affect the chickens [142]. Even exposure to a lower dose for a long time poses a potential risk. A study by Huang et al. [144] administered different doses of the toxin via oral gavage (0, 0.03, 0.1, 0.3, 1, and 3 mg/kg/day) to female rats (F0 generation) during gestation and then to offspring (F1 generation) up to 27 days postnatal. The results showed that a dose of up to 3 mg/kg/day has no effect on the body weight or survival of the F0 generation, but a decrease in body weight was observed in offspring (F1 generation) at a dose of 3 mg/kg/day. As there was no significant data on maternal toxicity either during gestation or lactation, the decrease in the bodyweight of F1 rats could be considered a direct effect of DON.
Further, DON is known to cause neurotoxicity. In a study by Wang et al. [145], piglets were fed on a DON-based diet of different concentrations (1.3 and 2.2 mg/kg) for 60 days and observed for its effect on their hippocampus, cerebral cortex, and cerebellum. The increasing concentration of the toxin was found to be linked with oxidative damage in brain parts, where the damage in the cell structure of the hippocampus was revealed using scanning electron microscopy. Moreover, an increase in the concentration of calcium and Ca2+ calmodulin-dependent kinase II (CaMKII) was observed. Other studies have suggested that the calcium ions are linked with neurotransmitter release and cell proliferation, and the disruption in Ca2+ homeostasis can negatively affect the neuronal circuits of the hippocampus [146,147]. Wang et al. [145] further suggested that DON toxicity involves Ca2+/CaM/CaMKII signaling pathways in neurotransmission and lipid peroxidation. The cytotoxic effect of DON has also been studied in mammary epithelial cells. Research indicated a lower yield of milk when cows were fed contaminated feed [148,149]. Lee et al. [150] investigated the effect of DON on mammary epithelial cells (MAC-T) in bovine animals. Different concentrations of the toxin (1–10 μM) were used for their impact on MAC-T cells. Results showed a significant reduction in proliferation activity of the cells with the increase in toxin level with the highest effect at 10 μM. The phosphorylation of phosphoinositide-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling molecules increased in MAC-T cells, demonstrating that DON regulates these signaling pathways.

10. Detection Techniques including Masked Mycotoxins

The conventional analytical methods for detecting DON includes gas chromatography-mass spectrometry (GCMS), high-performance liquid chromatography (HPLC) [151,152] and liquid chromatography-tandem mass spectrometry (LCMS/MS) [153,154]. Although these methods benefit from high sensitivity and specificity, the downsides are that they are time-consuming for sample preparation, require expensive instrumentation, and highly qualified personnel to operate [151]. Other methods for convenient, cheap and high sensitivity detection include enzyme-linked immunosorbent assay (ELISA) [155], lateral-flow immunochromatographic assay [156], lateral flow immunoassay [84], fluorescence [157], surface-enhanced Raman scattering (SERS) [158,159], and electrochemical detection [160].
To meet the need for real-time monitoring of mycotoxin, methods like lateral flow immunoassays (LFA) [161,162] and biosensing assays [163,164,165] were developed. LFA is supposed to be the cheapest, fastest and simplest way of detecting mycotoxins [166]. Jin et al. [167] developed a new dual near-infrared fluorescence-based LFA for detecting DON in maize. 5 g chopped maize sample was combined with 25 mL 70% methanol and vortexed for 2 min for sample preparation. The extracted solution was centrifuged at 5000× g for 5 min, and the supernatant was diluted 10 times with phosphate buffer saline, PBS (0.01 M, pH 7.4), followed by Tween 20 added to get a final 0.5% (v/v), and 200 μL of the diluted sample was evaluated by the NIR-based LFA. Further, Zhao et al. [168] developed a fast lateral flow fluorescence microsphere immunoassay test strip (FM-ICTS) for detecting DON residues in a variety of agricultural goods. 5 g of material was shaken for 8 min with 25 mL of extraction solution, then centrifuged (8000× g for 12 min). The supernatant was diluted 10-fold in 10 mM PBS + 3% Rhodasurf® On-870 before being detected with FM-ICTS. For identifying DON residues in agricultural goods, this approach is easy, reliable, and sensitive.
In addition, electrochemical and bio-sensors are powerful tools with high transmission and easy and low-cost operation [169,170]. Ong et al. [171] developed a selective biosensing device based on iron nanoflorets graphene nickel (INFGN) for selective detection of DON. Furthermore, Li et al. [172] developed a sensitive and selective antibody-like sensor based on molecularly imprinted poly(L-arginine) on carboxylic acid-functionalized carbon nanotubes for detecting DON in agri-food products. The sample was made by mixing wheat flour with acetonitrile/deionized water (84:16, v/v), ultrasonic extraction for 30 min, centrifugation at 15,000 rpm for 10 min, and collecting the supernatant. Finally, the supernatants were filtered and incubated at 4 °C through a 0.22 μm sterile membrane. This method provides a realistic biomimetic sensing platform for identifying mycotoxins in food and agriculture.
Besides these, multiplex immunochromatographic assays (mICAs) are employed for screening mycotoxins with rapid and excellent selectivity [173]. Recently gold nanoparticles (AuNPs) and quantum dots (QDs) [174,175] have been used as signal tags for detecting multiple mycotoxins in foodstuffs. Semiconductor QDs with a unique luminescent property are utilized for developing highly sensitive biosensors based on Forster resonance energy transfer (FRET). Goryacheva et al. [176] developed a FRET-based fluorescent immunoassay for detecting DON. 25 g of wheat samples were mixed with 100 mL of methanol/water (70/30, v/v) for 15 min on a horizontal shaker at room temperature. The wheat particles were precipitated via centrifugation (4000× g, 20 °C, 15 min). The supernatant was used for analysis after dilution of 1:5 with PBS.
As conventional immunochromatographic assays (ICAs) based on AuNPs have low sensitivity, Li et al. [177] developed a highly sensitive ICA based on polydopamine coated zirconium metal-organic frameworks labeled antibodies for visual determination of DON. Still, AuNPs-based mICAs show low sensitivity while QDs-based mICAs have interference between multiple analytes and recognition molecules [178,179]. Thereby, a multiplex immunochromatographic assay (mICA) was developed by Zhao et al. [168] based on novel α-Fe2O3 nanocubes (FNCs) for the simultaneous detection of DON. For 15 min, 5 g of samples were mixed with 10 mL methanol–water (70:30, v/v) and then sonicated for 10 min, followed by centrifugation (5439× g, 10 min) and dilution to 10-fold with 10 mM PBS (pH 7.4) solution to get the supernatant. The FNCs-mICA method provided sensitive, quick, and repeatable detection of multiple mycotoxins on-site.
Similarly, Huang et al. [180] developed an immunochromatographic test strip for rapid and simultaneous detection of fumonisin B1 and DON in grains. 1 g sieved sample was combined with 5 mL ultrapure water in a vortex mixer for 5 min, allowed to stand for 10 min, and the supernatant was filtered through 0.45 and 0.22 μm filter membranes to get clear solutions for the analysis. The developed technique was suitable for the on-site screening of large-scale samples. Furthermore, Subak et al. [181] developed a voltametric aptasensor for detecting DON in food and feed samples. A high-speed blender was used to mix 25 g of maize flour with 50 mL of 50% ethanol/water (v/v) for 5 min. The supernatant was collected and evaporated to dryness under a nitrogen flow after centrifugation at 5000 rpm for 5 min. The dried sample obtained was suspended in affinity buffer and differential pulse voltammetry measurements were used to record the response.
DON can bind with proteins or carbohydrates to form a varied structure known as “modified mycotoxins”, which include both “biologically and chemically modified” forms [182,183,184]. The term “masked mycotoxins” especially refers to “biologically modified” forms conjugated by plants [184]. Masked form of DON forms Deoxynivalenol-3-β-D-glucoside (D3G) by enzymatic interaction with glucose [39,185,186] while 3-Acetyl-deoxynivalenol (3Ac-DON) and 15-Acetyl-deoxynivalenol (15Ac-DON) [187,188] are formed during deacetylation of DON. DON and its masked forms, namely D3G, 3Ac-DON, and 15Ac-DON, have been reported in cereal and cereal-based products [189]. There is limited study on the toxicodynamics of these forms; however, their absorption, bioavailability, and toxicity have been studied [186,190].
These modified or masked forms of mycotoxins remain undetected by conventional analytical techniques leading to underreporting [184,185]. Hence, there are serious health concerns associated with masked/modified DON. They can even re-convert to their native form [187], which raises further toxicity concerns in mammals [3,4,5,6,191]. Therefore, Fan et al. [112] developed and validated an Ultra-High-Performance Liquid Chromatography-tandem Mass Spectrometry Method (UHPLC-MS) for simultaneous detection of masked DON in various feed samples. Further, Olopade et al. [192] used LC-MS/MS procedure to quantify DON and its masked forms, 3Ac-DON and 15Ac- DON. Despite this, their combined effects are unknown [193], necessitating additional toxicokinetic studies of masked versions [194]. To address these concerns, modified DON could be transformed using a hydrolytic process involving alkaline, acidic, or enzymatic methods [195,196] as well as an integrated strategy for qualitative and quantitative analysis of modified mycotoxins, as suggested by Lu et al. [194].

11. Legislation

Due to its widespread occurrence and severe impacts on human and animal health, DON has attracted international attention. As a result, measures for preventing, excluding, or inactivating DON in meals and feeds must be developed. DON contamination might be difficult to avoid from preharvest to manufacture. DON management strategies have been developed to (a) minimize DON formation or contamination prior to harvest, (b) degrade or eliminate DON from polluted foods and feeds, and (c) reduce DON bioavailability by reducing gastric absorption [16]. Table 2 summarizes the permissible limits of DON in various foods and feeds set by various regulatory organizations. The permissible limits of DON in various foods and feeds are higher in Europe than that set by US-FDA and FAO. Further, among the cereal category, the limits for maize and maize-based products are higher. In a real-world situation of an eco-environmental system, DON contamination occurs in combination with other mycotoxins, as well as its masked forms in food and feeds. This could be the reason behind the variation in permissible limits depending on the type of food and feed and their geographical location, which poses new challenges for government organizations with setting scientific legislation, regulations, and standards [15].

12. Management and Control Strategies, including Pre-Harvest Preventive Strategies and Post-Harvest Treatments and Detoxification Strategies

The development and implementation of efficient management and control strategies are necessary to reduce the DON contamination in food and feed when pre- and post-harvesting strategies cannot control them [198]. This detoxification technique is classified into physical, chemical, and biological methods [139]. Physical methods include washing, cleaning, dehulling, segregation, sieving, heat treatments, gamma, UV, and visible light radiations [199]. This method is most suitable for food compared to feed. Thermal treatment in food processing techniques has been divided into dry methods (baking, roasting, and frying) and wet methods (steaming and cooking) [200] and is considered to be an important tool for DON mitigation in foods, but the degradation products formed can have different toxic behavior than the parent mycotoxin [201].
Stadler et al. [202] concluded that baking of food could cause a partial degradation of DON because a considerable reduction in DON was observed with increased baking time and temperature, and the degradation of by-products was less toxic. The reduction during baking is either due to binding to matrix compounds or due to the transformation into other toxins but not the actual destruction of toxins [203]. During roasting, an effective reduction in DON was observed between 180 °C to 220 °C for 30 min and the reduction increased with increasing temperature [35]. However, frying had no significant effect on DON levels due to the high thermal stability and release of bound forms [200]. Further, Kalagatur et al. [204] observed that high pressure (1000–5500 bar) between 30–60 °C for 10–30 min during cooking significantly reduced DON levels in foods. More than a 40% reduction in DON was observed in the boiled product due to the leaching of DON into the broth [205]. In addition, gamma radiations have successfully reduced DON levels in wheat [198]. Several works have also reported degradation of DON using UV [206,207], where significant was observed without any changes in color and protein content of the treated food samples [208].
Chemical and biological methods are suitable for both food and feed samples. The chemical method uses chemicals such as calcium hydroxide monomethylamine and sodium bisulfite [209,210,211], moist and dry ozone [210,211], ascorbic acid and ammonium hydroxide [139], acid and alkaline electrolyzed water [212], and ammonia [213]. However, the physical and chemical methods have several disadvantages, such as limited efficacy, high cost, harmful chemical residues, loss of nutritional value, sophisticated equipment requirements coupled with safety issues; hence, environmentally-friendly techniques using microorganisms and enzymes have been developed [214].
Biological detoxification involves the use of bacteria, fungi, and actinomycetes to reduce or eliminate DON from products either through adsorption or enzymatic degradation [215,216]. Li et al. [217] observed that Bacillus subtilis had the highest detoxification rate out of 16 strains of bacteria and showed a synergistic effect with Lactobacillus plantarum, B. velezensis RC 218, and Streptomyces albidoflavus RC 87B showing up to 51% DON reduction in durum wheat, hence a potential biocontrol agent [218]. B. subtilis ASAG 216 degraded 81.1% DON under optimum temperature (35–50 °C), time period (8 hr), and pH (6.5–9.0) conditions because of degradation ability inherent in extracellular enzymes or proteins [219]. Bacillus licheniformis strain YB9 also reduced 82.6% DON [220]. Further, the bacterial consortium C20 was able to degrade DON at 30 °C and pH 8 [221]. Devosia insulae A16 was also able to degrade 88% DON within 48 hrs at 35 °C at pH 7 [222]. Slackia sp. D-G6 reduced DON by deep oxidation between 37–47 °C and pH 6–10 with non-toxic DOM-1 as a degradation by-product [223]. In addition, Pseudomonas sp. Y1 and Lysobacter sp. S1, soil-derived bacteria, had a significant DON degrading ability by an enzymatic transformation of DON into less toxic, 3-epi-DON [224]. Pelagibacterium halotolerans ANSP101 degraded DON by transforming it into less toxic 3-keto-deoxynivalenol at 40 °C and pH 8 [225]. Devosia strain D6-9 completely removed DON in wheat by catabolizing it into 3-keto-DON and 3-epi-DON [226]. Several works listed by Gao et al. [223] have shown the biodegradation of DON by fungi. DON detoxification can also be achieved by enzymatic reactions such as deep oxidation, oxidation, epimerization, and glycosylation and the formation of by-products like DOM-1, 3-keto DON, 3-epi DON, and DON-3-glucoside (D3G) [188].
However, due to certain limitations of physical, chemical, and biological methods such as low efficacy, the requirement of expensive chemicals, sophisticated equipment, and the formation of harmful chemical residues [227], a novel technology such as photocatalytic degradation has been developed that is easy to use, inexpensive, environmentally friendly, reusable, and has high stability [228]. This technique degrades the target toxin using a suitable photocatalyst such as the TiON@PdO nanoparticle [229], carbon-supported TiO2 [230], ZnO@graphene hybrids [231], upconversion nanoparticles@ TiO2 composite [215], and dendritic-like-Fe2O3 [232]. Another technique is electrochemical oxidation (ECO), which has emerged as a new oxidation technology that destroys DON by several mechanisms; in an experiment using graphite as an electrode, DON was significantly reduced due to the high potential and acidic conditions of the technique [233]. Atmospheric cold plasma technology (ACP) is another novel non-thermal approach for detoxification of DON, which utilizes plasma under low atmospheric pressure conditions with the main advantage of high chemical reactivity and efficacy compared to ozone or UV treatments and resulting in decontamination of DON within seconds [227,234].
With food safety becoming a global concern, significant focus on the control and prevention of accumulation of DON and its entry into the food supply chain is achievable through scientific agricultural interventions at pre-and post-harvest conditions [235]. These interventions involve good agricultural practices (GAPs), good storage practices (GSPs), and good management practices (GMPs) [236].
The mycotoxin contamination occurs during the growing period of crops, so it becomes necessary to identify appropriate pre- and post-agronomic practices to avoid grain contamination [237]. Pre-harvest strategies include the use of resistant varieties, removal of weed and damaged kernels, soil analysis, use of herbicide, fungicide and insecticide, seedbed treatment, crop rotation, tillage and plowing, the use of fertilizers, and genetically modified plants for suppression of mycotoxins [238,239,240]. The most effective practice to control the disease is the use of genetic resistant varieties, pedigree selection, and identification of primary infection pathways, such as silk channels, during the flowering time [46]. An attractive technology is to employ gene editing to resist fungus growth or to prevent mycotoxin formation in plants through kernel-specific RNA interference (RNAi) gene cassette targeting the aflC gene involved in the mycotoxin biosynthetic pathway [241]. Crop rotation or tillage is also suggested as a control measure of the disease as it decreases and manages the crop residue where most of the fungi survive [242]. The use of fertilizers, particularly nitrogen, boosts Fusarium development together with a favorable effect on plant growth, and, therefore, mycotoxin contamination rises [243].
During harvesting, it is important to consider the time of harvesting as a greater number of over mature or immature seeds can increase the mycotoxin levels in final products [244]. Crop stress during harvesting should also be minimized by avoiding early harvesting, damaged kernel collection, mechanical damage, and kernel contact with soil [245]. While transporting the harvested grain, the containers and vehicles should be clean, dry, and free of insect and fungal growth [246]. After harvesting and before storage, it is important to dry the grains to reduce the moisture level, which is responsible for fungal growth, and it can be done by the use of solar dryers instead of sun drying as slow drying increases the mycotoxin concentration [247]. During storage, it is necessary to control moisture, temperature, and relative humidity of the grain to prevent deterioration through fungal growth and mycotoxin production, with the ideal temperature being 1–4 °C in winter and 10–15 °C in summer, the water activity of less than or equal to 0.70 and relative humidity less than 70% [248]. Along with these storage conditions, the storage rooms should be clean, well ventilated, and protected from rain, drainage, insects, rodents, and birds and aeration of place with the circulation of air to maintain a uniform temperature [246]. For best management practices of stored grains, the most important principle is SLAM (sanitation, loading, aeration, and monitoring) [249].
To lower mycotoxin levels, post-harvest management strategies include adequate drying, shipping, and packaging, sorting and cleaning, drying, smoking, insect control, and pesticide use in storage rooms. These interventions can also involve best-practices teaching and awareness initiatives. It is critical to maintaining appropriate hygiene and the application of natural and chemical agents after harvesting the crop during storage [250]. From farm to table, a successful application of the Hazard Analysis Critical Control Point (HACCP)-based postharvest strategy should include measures for prevention, control, and quality [248].

13. Conclusions

DON mycotoxin is found in agricultural products worldwide, presenting a health risk to humans and animals. The extensive prevalence of DON in small grain cereals worldwide necessitates the application of fast and effective techniques. This is particularly important in developing countries, where DON and its transformed forms are underappreciated. Relevant food safety initiatives should be prioritized, with an emphasis on the unknown health effects of these mycotoxins co-occurring in vulnerable populations. The most effective way to monitor the occurrence of total mycotoxins in cereals and cereal-based products is to take preventive steps before and after harvest. Future studies should concentrate on elucidating previously unknown pathways of both individual and cumulative toxic impacts, as well as being more objective in determining the risk level of chronic DON toxicity in both humans and animals, which is essential for designing more precise policies and regulations to ensure of food and feed safety and security.

Author Contributions

P.K. and M.K. (Madhu Kamle) conceptualized and designed the manuscript; D.K.M., A.G., S.P., B.S., K.D., V., S.M. and R.S. wrote the manuscript; P.R., A.D.T., M.K. (Manoj Kumar) and A.K. assisted in manuscript editing; P.K., M.K. (Madhu Kamle) and S.G. critically evaluated and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

No external funding was received to support this work.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

All the authors would like to thank their respective departments for their support and cooperation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pleadin, J.; Frece, J.; Markov, K. Mycotoxins in food and feed. Adv. Food Nutr. Res. 2019, 89, 297–345. [Google Scholar] [PubMed]
  2. Kumar, P.; Mahato, D.K.; Kamle, M.; Mohanta, T.K.; Kang, S.G. Aflatoxins: A global concern for food safety, human health and their management. Front. Microbiol. 2017, 7, 2170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mahato, D.K.; Devi, S.; Pandhi, S.; Sharma, B.; Maurya, K.K.; Mishra, S.; Dhawan, K.; Selvakumar, R.; Kamle, M.; Mishra, A.K. Occurrence, Impact on Agriculture, Human Health, and Management Strategies of Zearalenone in Food and Feed: A Review. Toxins 2021, 13, 92. [Google Scholar] [CrossRef] [PubMed]
  4. Kamle, M.; Mahato, D.K.; Devi, S.; Lee, K.E.; Kang, S.G.; Kumar, P. Fumonisins: Impact on agriculture, food, and human health and their management strategies. Toxins 2019, 11, 328. [Google Scholar] [CrossRef] [Green Version]
  5. Kumar, P.; Mahato, D.K.; Sharma, B.; Borah, R.; Haque, S.; Mahmud, M.M.C.; Shah, A.K.; Rawal, D.; Bora, H.; Bui, S. Ochratoxins in food and feed: Occurrence and its impact on human health and management strategies. Toxicon 2020, 187, 151–162. [Google Scholar] [CrossRef]
  6. Mahato, D.K.; Lee, K.E.; Kamle, M.; Devi, S.; Dewangan, K.N.; Kumar, P.; Kang, S.G. Aflatoxins in food and feed: An overview on prevalence, detection and control strategies. Front. Microbiol. 2019, 10, 2266. [Google Scholar] [CrossRef]
  7. Xu, H.; Wang, L.; Sun, J.; Wang, L.; Guo, H.; Ye, Y.; Sun, X. Microbial detoxification of mycotoxins in food and feed. Crit. Rev. Food Sci. Nutr. 2021, 1–19. [Google Scholar] [CrossRef]
  8. Mahato, D.K.; Kamle, M.; Sharma, B.; Pandhi, S.; Devi, S.; Dhawan, K.; Selvakumar, R.; Mishra, D.; Kumar, A.; Arora, S. Patulin in food: A mycotoxin concern for human health and its management strategies. Toxicon 2021, 198, 12–23. [Google Scholar] [CrossRef]
  9. Kamle, M.; Mahato, D.K.; Gupta, A.; Pandhi, S.; Sharma, N.; Sharma, B.; Mishra, S.; Arora, S.; Selvakumar, R.; Saurabh, V. Citrinin Mycotoxin Contamination in Food and Feed: Impact on Agriculture, Human Health, and Detection and Management Strategies. Toxins 2022, 14, 85. [Google Scholar] [CrossRef]
  10. Kebede, H.; Liu, X.; Jin, J.; Xing, F. Current status of major mycotoxins contamination in food and feed in Africa. Food Control 2020, 110, 106975. [Google Scholar] [CrossRef]
  11. Afsah-Hejri, L.; Hajeb, P.; Ehsani, R.J. Application of ozone for degradation of mycotoxins in food: A review. Compr. Rev. Food Sci. Food Saf. 2020, 19, 1777–1808. [Google Scholar] [CrossRef] [PubMed]
  12. Nagl, V.; Schatzmayr, G. Deoxynivalenol and its masked forms in food and feed. Curr. Opin. Food Sci. 2015, 5, 43–49. [Google Scholar] [CrossRef]
  13. Yuan, J.; Sun, C.; Guo, X.; Yang, T.; Wang, H.; Fu, S.; Li, C.; Yang, H. A rapid Raman detection of deoxynivalenol in agricultural products. Food Chem. 2017, 221, 797–802. [Google Scholar] [CrossRef]
  14. Mahato, D.K.; Pandhi, S.; Kamle, M.; Gupta, A.; Sharma, B.; Panda, B.K.; Srivastava, S.; Kumar, M.; Selvakumar, R.; Pandey, A.K. Trichothecenes in food and feed: Occurrence, impact on human health and their detection and management strategies. Toxicon 2022, 208, 62–77. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, H.; Guog, T.; Dai, H.; Yu, Y.; Zhang, Y.; Ma, L. Deoxynivalenol: Toxicological profiles and perspective views for future research. World Mycotoxin J. 2020, 13, 179–188. [Google Scholar] [CrossRef]
  16. Guo, H.; Ji, J.; Wang, J.S.; Sun, X. Deoxynivalenol: Masked forms, fate during food processing, and potential biological remedies. Compr. Rev. Food Sci. Food Saf. 2020, 19, 895–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Holanda, D.M.; Kim, S.W. Mycotoxin occurrence, toxicity, and detoxifying agents in pig production with an emphasis on deoxynivalenol. Toxins 2021, 13, 171. [Google Scholar] [CrossRef]
  18. Bretz, M.; Beyer, M.; Cramer, B.; Knecht, A.; Humpf, H.-U. Thermal degradation of the Fusarium mycotoxin deoxynivalenol. J. Agric. Food Chem. 2006, 54, 6445–6451. [Google Scholar] [CrossRef]
  19. Gruber-Dorninger, C.; Jenkins, T.; Schatzmayr, G. Global mycotoxin occurrence in feed: A ten-year survey. Toxins 2019, 11, 375. [Google Scholar] [CrossRef] [Green Version]
  20. Femenias, A.; Gatius, F.; Ramos, A.J.; Sanchis, V.; Marín, S. Use of hyperspectral imaging as a tool for Fusarium and deoxynivalenol risk management in cereals: A review. Food Control 2020, 108, 106819. [Google Scholar] [CrossRef]
  21. Petcu, C.D.; Georgescu, I.M.; Zvorișteanu, O.V.; Negreanu, C.N. Study referring to the appearance of contamination with deoxynivalenol in grains, grain flour and bakery products on the Romanian market. Sci. Papers Ser. D. Anim. Sci. 2019, 62, 214–245. [Google Scholar]
  22. Juan, C.; Raiola, A.; Mañes, J.; Ritieni, A. Presence of mycotoxin in commercial infant formulas and baby foods from Italian market. Food Control 2014, 39, 227–236. [Google Scholar] [CrossRef]
  23. Hietaniemi, V.; Rämö, S.; Yli-Mattila, T.; Jestoi, M.; Peltonen, S.; Kartio, M.; Sieviläinen, E.; Koivisto, T.; Parikka, P. Updated survey of Fusarium species and toxins in Finnish cereal grains. Food Addit. Contam. Part A 2016, 33, 831–848. [Google Scholar] [CrossRef] [PubMed]
  24. Lindblad, M.; Börjesson, T.; Hietaniemi, V.; Elen, O. Statistical analysis of agronomical factors and weather conditions influencing deoxynivalenol levels in oats in Scandinavia. Food Addit. Contam. Part A 2012, 29, 1566–1571. [Google Scholar] [CrossRef]
  25. Oueslati, S.; Berrada, H.; Mañes, J.; Juan, C. Presence of mycotoxins in Tunisian infant foods samples and subsequent risk assessment. Food Control 2018, 84, 362–369. [Google Scholar] [CrossRef]
  26. Pascari, X.; Marín, S.; Ramos, A.J.; Molino, F.; Sanchis, V. Deoxynivalenol in cereal-based baby food production process. A review. Food Control 2019, 99, 11–20. [Google Scholar] [CrossRef] [Green Version]
  27. Nakagawa, H.; He, X.; Matsuo, Y.; Singh, P.K.; Kushiro, M. Analysis of the masked metabolite of deoxynivalenol and Fusarium resistance in CIMMYT wheat germplasm. Toxins 2017, 9, 238. [Google Scholar] [CrossRef] [Green Version]
  28. Xia, R.; Schaafsma, A.W.; Wu, F.; Hooker, D.C. The change in winter wheat response to deoxynivalenol and Fusarium Head Blight through technological and agronomic progress. Plant Dis. 2021, 105, 840–850. [Google Scholar] [CrossRef]
  29. Brauer, E.K.; Balcerzak, M.; Rocheleau, H.; Leung, W.; Schernthaner, J.; Subramaniam, R.; Ouellet, T. Genome editing of a deoxynivalenol-induced transcription factor confers resistance to Fusarium graminearum in wheat. Mol. Plant Microbe Interact. 2020, 33, 553–560. [Google Scholar] [CrossRef]
  30. Gaikpa, D.S.; Lieberherr, B.; Maurer, H.P.; Longin, C.F.H.; Miedaner, T. Comparison of rye, triticale, durum wheat and bread wheat genotypes for Fusarium head blight resistance and deoxynivalenol contamination. Plant Breed. 2020, 139, 251–262. [Google Scholar] [CrossRef]
  31. Mielniczuk, E.; Cegiełko, M.; Kiecan, I.; Perkowski, J.; Pastucha, A. The pathogenicity and toxigenic properties of Fusarium crookwellense LW Burgess, PE Nelson & Toussoun depending on weather conditions. Acta Sci. Pol. Hortorum Cultus 2020, 19, 17–24. [Google Scholar]
  32. Palacios, S.A.; Del Canto, A.; Erazo, J.; Torres, A.M. Fusarium cerealis causing Fusarium head blight of durum wheat and its associated mycotoxins. Int. J. Food Microbiol. 2021, 346, 109161. [Google Scholar] [CrossRef] [PubMed]
  33. Minnaar-Ontong, A.; Herselman, L.; Kriel, W.-M.; Leslie, J.F. Morphological characterization and trichothecene genotype analysis of a Fusarium Head Blight population in South Africa. Eur. J. Plant Pathol. 2017, 148, 261–269. [Google Scholar] [CrossRef]
  34. Pfordt, A.; Schiwek, S.; Rathgeb, A.; Rodemann, C.; Bollmann, N.; Buchholz, M.; Karlovsky, P.; von Tiedemann, A. Occurrence, Pathogenicity, and Mycotoxin Production of Fusarium temperatum in Relation to Other Fusarium Species on Maize in Germany. Pathogens 2020, 9, 864. [Google Scholar] [CrossRef]
  35. Pleadin, J.; Babić, J.; Vulić, A.; Kudumija, N.; Aladić, K.; Kiš, M.; Jaki Tkalec, V.; Škrivanko, M.; Lolić, M.; Šubarić, D. The effect of thermal processing on the reduction of deoxynivalenol and zearalenone cereal content. Croat. J. Food Sci. Technol. 2019, 11, 44–51. [Google Scholar] [CrossRef]
  36. Deng, Y.; You, L.; Nepovimova, E.; Wang, X.; Musilek, K.; Wu, Q.; Wu, W.; Kuca, K. Biomarkers of deoxynivalenol (DON) and its modified form DON-3-glucoside (DON-3G) in humans. Trends Food Sci. Technol. 2021, 110, 551–558. [Google Scholar] [CrossRef]
  37. Park, J.; Chang, H.; Kim, D.; Chung, S.; Lee, C. Long-term occurrence of deoxynivalenol in feed and feed raw materials with a special focus on South Korea. Toxins 2018, 10, 127. [Google Scholar] [CrossRef] [Green Version]
  38. Jiang, C.; Zhang, C.; Wu, C.; Sun, P.; Hou, R.; Liu, H.; Wang, C.; Xu, J.R. TRI6 and TRI10 play different roles in the regulation of deoxynivalenol (DON) production by cAMP signalling in Fusarium graminearum. Environ. Microbiol. 2016, 18, 3689–3701. [Google Scholar] [CrossRef]
  39. Khaneghah, A.M.; Martins, L.M.; von Hertwig, A.M.; Bertoldo, R.; Sant’Ana, A.S. Deoxynivalenol and its masked forms: Characteristics, incidence, control and fate during wheat and wheat based products processing—A review. Trends Food Sci. Technol. 2018, 71, 13–24. [Google Scholar] [CrossRef]
  40. Hallen-Adams, H.E.; Wenner, N.; Kuldau, G.A.; Trail, F. Deoxynivalenol biosynthesis-related gene expression during wheat kernel colonization by Fusarium graminearum. Phytopathology 2011, 101, 1091–1096. [Google Scholar] [CrossRef] [Green Version]
  41. Zheng, D.; Zhang, S.; Zhou, X.; Wang, C.; Xiang, P.; Zheng, Q.; Xu, J.-R. The FgHOG1 pathway regulates hyphal growth, stress responses, and plant infection in Fusarium graminearum. PLoS ONE 2012, 7, e49495. [Google Scholar] [CrossRef]
  42. Hu, S.; Zhou, X.; Gu, X.; Cao, S.; Wang, C.; Xu, J.-R. The cAMP-PKA pathway regulates growth, sexual and asexual differentiation, and pathogenesis in Fusarium graminearum. Mol. Plant Microbe Interact. 2014, 27, 557–566. [Google Scholar] [CrossRef] [Green Version]
  43. Fan, K.; Liu, X.; Guo, W.; Zhang, Z.; Meng, J.; Du, Y.; Nie, D. Production of free and modified forms of deoxynivalenol in wheat grain. J. Food Saf. Qual. 2019, 10, 2545–2554. [Google Scholar]
  44. Kotowicz, N.K.; Frąc, M.; Lipiec, J. The importance of Fusarium fungi in wheat cultivation–pathogenicity and mycotoxins production: A review. J. Anim. Plant Sci. 2014, 21, 3326–3343. [Google Scholar]
  45. Han, Z.; Shen, Y.; Di Mavungu, J.D.; Zhang, D.; Nie, D.; Jiang, K.; De Saeger, S.; Zhao, Z. Relationship between environmental conditions, TRI5 gene expression and deoxynivalenol production in stored Lentinula edodes infected with Fusarium graminearum. World Mycotoxin J. 2018, 11, 177–186. [Google Scholar] [CrossRef]
  46. Czembor, E.; Stępień, Ł.; Waśkiewicz, A. Effect of environmental factors on Fusarium species and associated mycotoxins in maize grain grown in Poland. PLoS ONE 2015, 10, e0133644. [Google Scholar] [CrossRef]
  47. Ramirez, M.L.; Chulze, S.; Magan, N. Impact of environmental factors and fungicides on growth and deoxinivalenol production by Fusarium graminearum isolates from Argentinian wheat. Crop Prot. 2004, 23, 117–125. [Google Scholar] [CrossRef]
  48. Martins, M.L.G.; Martins, H.M. Influence of water activity, temperature and incubation time on the simultaneous production of deoxynivalenol and zearalenone in corn (Zea mays) by Fusarium graminearum. Food Chem. 2002, 79, 315–318. [Google Scholar] [CrossRef]
  49. 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]
  50. Peter Mshelia, L.; Selamat, J.; Iskandar Putra Samsudin, N.; Rafii, M.Y.; Abdul Mutalib, N.-A.; Nordin, N.; Berthiller, F. Effect of temperature, water activity and carbon dioxide on fungal growth and mycotoxin production of acclimatised isolates of Fusarium verticillioides and F. graminearum. Toxins 2020, 12, 478. [Google Scholar] [CrossRef]
  51. Belizán, M.M.E.; Gomez, A.D.L.A.; Baptista, Z.P.T.; Jimenez, C.M.; Matías, M.d.H.S.; Catalán, C.A.N.; Sampietro, D.A. Influence of water activity and temperature on growth and production of trichothecenes by Fusarium graminearum sensu stricto and related species in maize grains. Int. J. Food Microbiol. 2019, 305, 108242. [Google Scholar] [CrossRef]
  52. Rybecky, A.I.; Chulze, S.N.; Chiotta, M.L. Effect of water activity and temperature on growth and trichothecene production by Fusarium meridionale. Int. J. Food Microbiol. 2018, 285, 69–73. [Google Scholar] [CrossRef]
  53. Cambaza, E.; Koseki, S.; Kawamura, S. Fusarium graminearum colors and Deoxynivalenol synthesis at different water activity. Foods. 2019, 8, 7. [Google Scholar] [CrossRef] [Green Version]
  54. Gruber-Dorninger, C.; Novak, B.; Nagl, V.; Berthiller, F. Emerging mycotoxins: Beyond traditionally determined food contaminants. J. Agric. Food Chem. 2017, 65, 7052–7070. [Google Scholar] [CrossRef]
  55. Ghebremeskel, M.; Langseth, W. The occurrence of culmorin and hydroxy-culmorins in cereals. Mycopathologia 2001, 152, 103–108. [Google Scholar] [CrossRef]
  56. Uhlig, S.; Eriksen, G.S.; Hofgaard, I.S.; Krska, R.; Beltrán, E.; Sulyok, M. Faces of a changing climate: Semi-quantitative multi-mycotoxin analysis of grain grown in exceptional climatic conditions in Norway. Toxins 2013, 5, 1682–1697. [Google Scholar] [CrossRef]
  57. Flynn, C.; Tang, G.; Boenisch, M.; Blum, A.; Broz, K.; Chen, Y.; Schmidt-Dannert, C.; Ma, Z.; Kistler, C. Fusarium toxisomes may be necessary for synthesis of high levels of deoxynivalenol and production of the distinct sesquiterpene mycotoxin, culmorin. In Proceedings of the International Congress of Plant Pathology (ICPP) 2018: Plant Health in A Global Economy, Boston, MA, USA, 29 July–3 August 2018. [Google Scholar]
  58. Generotti, S.; Cirlini, M.; Šarkanj, B.; Sulyok, M.; Berthiller, F.; Dall’Asta, C.; Suman, M. Formulation and processing factors affecting trichothecene mycotoxins within industrial biscuit-making. Food Chem. 2017, 229, 597–603. [Google Scholar] [CrossRef]
  59. Nogueira, M.S.; Decundo, J.; Martinez, M.; Dieguez, S.N.; Moreyra, F.; Moreno, M.V.; Stenglein, S.A. Natural contamination with mycotoxins produced by Fusarium graminearum and Fusarium poae in malting barley in Argentina. Toxins 2018, 10, 78. [Google Scholar] [CrossRef] [Green Version]
  60. Piacentini, K.C.; Rocha, L.O.; Savi, G.D.; Carnielli-Queiroz, L.; Almeida, F.G.; Minella, E.; Corrêa, B. Occurrence of deoxynivalenol and zearalenone in brewing barley grains from Brazil. Mycotoxin Res. 2018, 34, 173–178. [Google Scholar] [CrossRef]
  61. Tabuc, C.; Marin, D.; Guerre, P.; Sesan, T.; Bailly, J.D. Molds and mycotoxin content of cereals in southeastern Romania. J. Food Prot. 2009, 72, 662–665. [Google Scholar] [CrossRef]
  62. Bensassi, F.; Rjiba, I.; Zarrouk, A.; Rhouma, A.; Hajlaoui, M.R.; Bacha, H. Deoxynivalenol contamination in Tunisian barley in the 2009 harvest. Food Addit. Contam. Part B 2011, 4, 205–211. [Google Scholar] [CrossRef]
  63. Ambrus, Á.; Szeitznė-Szabó, M.; Zentai, A.; Sali, J.; Szabo, I.J. Exposure of consumers to deoxynivalenol from consumption of white bread in Hungary. Food Addit. Contam. 2011, 28, 209–217. [Google Scholar] [CrossRef] [Green Version]
  64. Kim, D.-H.; Hong, S.-Y.; Jeon, M.-H.; An, J.-M.; Kim, S.-Y.; Kim, H.-Y.; Yoon, B.R.; Chung, S.H. Simultaneous determination of the levels of deoxynivalenol, 3-acetyldeoxynivalenol, and nivalenol in grain and feed samples from South Korea using a high-performance liquid chromatography–photodiode array detector. Appl. Biol. Chem. 2016, 59, 881–887. [Google Scholar] [CrossRef]
  65. Wu, L.; Li, J.; Li, Y.; Li, T.; He, Q.; Tang, Y.; Liu, H.; Su, Y.; Yin, Y.; Liao, P. Aflatoxin B1, zearalenone and deoxynivalenol in feed ingredients and complete feed from different Province in China. J. Anim. Sci. Biotechnol. 2016, 7, 63. [Google Scholar] [CrossRef] [Green Version]
  66. Torović, L. Fusarium toxins in corn food products: A survey of the Serbian retail market. Food Addit. Contam. Part A 2018, 35, 1596–1609. [Google Scholar] [CrossRef]
  67. Liu, J.; Sun, L.; Zhang, J.; Guo, J.; Chen, L.; Qi, D.; Zhang, N. Aflatoxin B1, zearalenone and deoxynivalenol in feed ingredients and complete feed from central China. Food Addit. Contam. Part B 2016, 9, 91–97. [Google Scholar] [CrossRef]
  68. Abdallah, M.F.; Girgin, G.; Baydar, T.; Krska, R.; Sulyok, M. Occurrence of multiple mycotoxins and other fungal metabolites in animal feed and maize samples from Egypt using LC-MS/MS. J. Sci. Food Agric. 2017, 97, 4419–4428. [Google Scholar] [CrossRef]
  69. Tima, H.; Rácz, A.; Guld, Z.; Mohácsi-Farkas, C.; Kiskó, G. Deoxynivalenol, zearalenone and T-2 in grain based swine feed in Hungary Food Addit. Contam. Part B 2016, 9, 275–280. [Google Scholar] [CrossRef]
  70. Kosicki, R.; Błajet-Kosicka, A.; Grajewski, J.; Twarużek, M. Multiannual mycotoxin survey in feed materials and feedingstuffs. Anim. Feed Sci. Technol. 2016, 215, 165–180. [Google Scholar] [CrossRef]
  71. Kos, J.; Hajnal, E.J.; Šarić, B.; Jovanov, P.; Nedeljković, N.; Milovanović, I.; Krulj, J. The influence of climate conditions on the occurrence of deoxynivalenol in maize harvested in Serbia during 2013–2015. Food Control 2017, 73, 734–740. [Google Scholar] [CrossRef]
  72. Gruber-Dorninger, C.; Jenkins, T.; Schatzmayr, G. Multi-mycotoxin screening of feed and feed raw materials from Africa. World Mycotoxin J. 2018, 11, 369–383. [Google Scholar] [CrossRef]
  73. Desjardins, A.E.; Manandhar, G.; Plattner, R.D.; Maragos, C.M.; Shrestha, K.; McCormick, S.P. Occurrence of Fusarium species and mycotoxins in Nepalese maize and wheat and the effect of traditional processing methods on mycotoxin levels. J. Agric. Food Chem. 2000, 48, 1377–1383. [Google Scholar] [CrossRef] [PubMed]
  74. Tittlemier, S.A.; Roscoe, M.; Trelka, R.; Gaba, D.; Chan, J.M.; Patrick, S.K.; Sulyok, M.; Krska, R.; McKendry, T.; Gräfenhan, T. Fusarium damage in small cereal grains from Western Canada. 2. Occurrence of Fusarium toxins and their source organisms in durum wheat harvested in 2010. J. Agric. Food Chem. 2013, 61, 5438–5448. [Google Scholar] [CrossRef] [PubMed]
  75. Marques, M.F.; Martins, H.M.; Costa, J.M.; Bernardo, F. Co-occurrence of deoxynivalenol and zearalenone in crops marketed in Portugal. Food Addit. Contam. 2008, 1, 130–133. [Google Scholar] [CrossRef] [PubMed]
  76. Tutelyan, V.A.; Zakharova, L.P.; Sedova, I.B.; Perederyaev, O.I.; Aristarkhova, T.V.; Eller, K.I. Fusariotoxins in Russian Federation 2005–2010 grain harvests. Food Addit. Contam. Part B 2013, 6, 139–145. [Google Scholar] [CrossRef]
  77. Fredlund, E.; Gidlund, A.; Sulyok, M.; Börjesson, T.; Krska, R.; Olsen, M.; Lindblad, M. Deoxynivalenol and other selected Fusarium toxins in Swedish oats—Occurrence and correlation to specific Fusarium species. Int. J. Food Microbiol. 2013, 167, 276–283. [Google Scholar] [CrossRef]
  78. Edwards, S.G. Impact of agronomic and climatic factors on the mycotoxin content of harvested oats in the United Kingdom. Food Addit. Contam. Part A 2017, 34, 2230–2241. [Google Scholar] [CrossRef]
  79. Raiola, A.; Meca, G.; Mañes, J.; Ritieni, A. Bioaccessibility of deoxynivalenol and its natural co-occurrence with ochratoxin A and aflatoxin B1 in Italian commercial pasta. Food Chem. Toxicol. 2012, 50, 280–287. [Google Scholar] [CrossRef] [Green Version]
  80. Majeed, S.; De Boevre, M.; De Saeger, S.; Rauf, W.; Tawab, A.; Rahman, M.; Iqbal, M. Multiple mycotoxins in rice: Occurrence and health risk assessment in children and adults of Punjab, Pakistan. Toxins 2018, 10, 77. [Google Scholar] [CrossRef] [Green Version]
  81. Hoogenboom, L.A.P.; Bokhorst, J.G.; Northolt, M.D.; Van de Vijver, L.P.L.; Broex, N.J.G.; Mevius, D.J.; Meijs, J.A.C.; Van der Roest, J. Contaminants and microorganisms in Dutch organic food products: A comparison with conventional products. Food Addit. Contam. 2008, 25, 1195–1207. [Google Scholar] [CrossRef]
  82. Jajić, I.; Jurić, V.; Glamočić, D.; Abramović, B. Occurrence of deoxynivalenol in maize and wheat in Serbia. Int. J. Mol. Sci. 2008, 9, 2114–2126. [Google Scholar] [CrossRef] [Green Version]
  83. Darsanaki, R.K.; Issazadeh, K.; Aliabadi, M.A.; Chakoosari, M.M.D. Occurrence of Deoxynivalenol (DON) in wheat flours in Guilan Province, northern Iran. Ann. Agric. Environ. Med. 2015, 22, 35–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Urusov, A.E.; Gubaidullina, M.K.; Petrakova, A.V.; Zherdev, A.V.; Dzantiev, B.B. A new kind of highly sensitive competitive lateral flow immunoassay displaying direct analyte-signal dependence. Application to the determination of the mycotoxin deoxynivalenol. Microchim. Acta 2018, 185, 29. [Google Scholar] [CrossRef] [PubMed]
  85. Pan, D.; Graneri, J.; Bettucci, L. Correlation of rainfall and levels of deoxynivalenol in wheat from Uruguay, 1997–2003. Food Addit. Contam. 2009, 2, 162–165. [Google Scholar] [CrossRef]
  86. Bernhoft, A.; Christensen, E.; Sandvik, M. The surveillance Programme for Mycotoxins and Fungi in Feed Materials, and Complete and Complementary Feed in Norway 2015; Norwegian Veterinary Institute: Oslo, Norway, 2016. [Google Scholar]
  87. Lindblad, M.; Gidlund, A.; Sulyok, M.; Börjesson, T.; Krska, R.; Olsen, M.; Fredlund, E. Deoxynivalenol and other selected Fusarium toxins in Swedish wheat—Occurrence and correlation to specific Fusarium species. Int. J. Food Microbiol. 2013, 167, 284–291. [Google Scholar] [CrossRef]
  88. Tima, H.; Brückner, A.; Mohácsi-Farkas, C.; Kiskó, G. Fusarium mycotoxins in cereals harvested from Hungarian fields. Food Addit. Contam. Part B 2016, 9, 127–131. [Google Scholar] [CrossRef]
  89. Palacios, S.A.; Erazo, J.G.; Ciasca, B.; Lattanzio, V.M.T.; Reynoso, M.M.; Farnochi, M.C.; Torres, A.M. Occurrence of deoxynivalenol and deoxynivalenol-3-glucoside in durum wheat from Argentina. Food Chem. 2017, 230, 728–734. [Google Scholar] [CrossRef]
  90. Vogelgsang, S.; Musa, T.; Bänziger, I.; Kägi, A.; Bucheli, T.D.; Wettstein, F.E.; Pasquali, M.; Forrer, H.-R. Fusarium mycotoxins in Swiss wheat: A survey of growers’ samples between 2007 and 2014 shows strong year and minor geographic effects. Toxins 2017, 9, 246. [Google Scholar] [CrossRef] [Green Version]
  91. Egbontan, A.O.; Afolabi, C.G.; Kehinde, I.A.; Enikuomehin, O.A.; Ezekiel, C.N.; Sulyok, M.; Warth, B.; Krska, R. A mini-survey of moulds and mycotoxins in locally grown and imported wheat grains in Nigeria. Mycotoxin Res. 2017, 33, 59–64. [Google Scholar] [CrossRef]
  92. Silva, M.V.; Pante, G.C.; Romoli, J.C.Z.; de Souza, A.P.M.; Rocha, G.H.O.D.; Ferreira, F.D.; Feijó, A.L.R.; Moscardi, S.M.P.; de Paula, K.R.; Bando, E. Occurrence and risk assessment of population exposed to deoxynivalenol in foods derived from wheat flour in Brazil. Food Addit. Contam. Part A 2018, 35, 546–554. [Google Scholar] [CrossRef]
  93. Bryła, M.; Waśkiewicz, A.; Podolska, G.; Szymczyk, K.; Jędrzejczak, R.; Damaziak, K.; Sułek, A. Occurrence of 26 mycotoxins in the grain of cereals cultivated in Poland. Toxins 2016, 8, 160. [Google Scholar] [CrossRef] [Green Version]
  94. Zhao, Y.; Guan, X.; Zong, Y.; Hua, X.; Xing, F.; Wang, Y.; Wang, F.; Liu, Y. Deoxynivalenol in wheat from the Northwestern region in China. Food Addit. Contam. Part B 2018, 11, 281–285. [Google Scholar] [CrossRef]
  95. Mishra, S.; Ansari, K.M.; Dwivedi, P.D.; Pandey, H.P.; Das, M. Occurrence of deoxynivalenol in cereals and exposure risk assessment in Indian population. Food Control 2013, 30, 549–555. [Google Scholar] [CrossRef]
  96. Šliková, S.; Gavurníková, S.; Šudyová, V.; Gregová, E. Occurrence of deoxynivalenol in wheat in Slovakia during 2010 and 2011. Toxins 2013, 5, 1353–1361. [Google Scholar] [CrossRef]
  97. Vidal, A.; Marín, S.; Ramos, A.J.; Cano-Sancho, G.; Sanchis, V. Determination of aflatoxins, deoxynivalenol, ochratoxin A and zearalenone in wheat and oat based bran supplements sold in the Spanish market. Food Chem. Toxicol. 2013, 53, 133–138. [Google Scholar] [CrossRef] [Green Version]
  98. Bertuzzi, T.; Camardo Leggieri, M.; Battilani, P.; Pietri, A. Co-occurrence of type A and B trichothecenes and zearalenone in wheat grown in northern Italy over the years 2009–2011. Food Addit. Contam. Part B 2014, 7, 273–281. [Google Scholar] [CrossRef]
  99. Jajić, I.; Krstović, S.; Kos, J.; Abramović, B. Incidence of deoxynivalenol in Serbian wheat and barley. J. Food Prot. 2014, 77, 853–858. [Google Scholar] [CrossRef]
  100. Nathanail, A.V.; Syvähuoko, J.; Malachová, A.; Jestoi, M.; Varga, E.; Michlmayr, H.; Adam, G.; Sieviläinen, E.; Berthiller, F.; Peltonen, K. Simultaneous determination of major type A and B trichothecenes, zearalenone and certain modified metabolites in Finnish cereal grains with a novel liquid chromatography-tandem mass spectrometric method. Anal. Bioanal. Chem. 2015, 407, 4745–4755. [Google Scholar] [CrossRef] [Green Version]
  101. Jakšić, S.; Abramović, B.; Jajić, I.; Baloš, M.Ž.; Mihaljev, Ž.; Despotović, V.; Šojić, D. Co-occurrence of fumonisins and deoxynivalenol in wheat and maize harvested in Serbia. Bull. Environ. Contam. Toxicol. 2012, 89, 615–619. [Google Scholar] [CrossRef]
  102. Topi, D.; Babič, J.; Pavšič-Vrtač, K.; Tavčar-Kalcher, G.; Jakovac-Strajn, B. Incidence of Fusarium Mycotoxins in Wheat and Maize from Albania. Molecules 2021, 26, 172. [Google Scholar] [CrossRef]
  103. Supronienė, S.; Sakalauskas, S.; Mankevičienė, A.; Barčauskaitė, K.; Jonavičienė, A. Distribution of B type trichothecene producing Fusarium species in wheat grain and relation to mycotoxins DON and NIV concentrations. Zemdirbyste-Agriculture 2016, 103, 281–288. [Google Scholar] [CrossRef] [Green Version]
  104. Sanders, M.; McPartlin, D.; Moran, K.; Guo, Y.; Eeckhout, M.; O’Kennedy, R.; De Saeger, S.; Maragos, C. Comparison of enzyme-linked immunosorbent assay, surface plasmon resonance and biolayer interferometry for screening of deoxynivalenol in wheat and wheat dust. Toxins 2016, 8, 103. [Google Scholar] [CrossRef] [Green Version]
  105. Remža, J.; Lacko-Bartošová, M.; Kosík, T. Official control of wheat mycotoxins contamination in the Slovak Republic. J. Microbiol. Biotechnol. Food Sci. 2021, 2021, 270–272. [Google Scholar]
  106. Femenias, A.; Gatius, F.; Ramos, A.J.; Sanchis, V.; Marín, S. Near-infrared hyperspectral imaging for deoxynivalenol and ergosterol estimation in wheat samples. Food Chem. 2021, 341, 128206. [Google Scholar] [CrossRef]
  107. Gholampour Azizi, I.; Arjmandi, J.; Ahmadi, S.; Rouhi, S. A Comparative Study on Deoxynivalenol Mycotoxin Level in Wheat Flour and Bread Samples. J. Qazvin Univ. Med. Sci. 2020, 24, 366–373. [Google Scholar] [CrossRef]
  108. Dombrink-Kurtzman, M.A.; Poling, S.M.; Kendra, D.F. Determination of deoxynivalenol in infant cereal by immunoaffinity column cleanup and high-pressure liquid chromatography–UV detection. J. Food Prot. 2010, 73, 1073–1076. [Google Scholar] [CrossRef]
  109. Gummadidala, P.M.; Omebeyinje, M.H.; Burch, J.A.; Chakraborty, P.; Biswas, P.K.; Banerjee, K.; Wang, Q.; Jesmin, R.; Mitra, C.; Moeller, P.D.R. Complementary feeding may pose a risk of simultaneous exposures to aflatoxin M1 and deoxynivalenol in Indian infants and toddlers: Lessons from a mini-survey of food samples obtained from Kolkata, India. Food Chem. Toxicol. 2019, 123, 9–15. [Google Scholar] [CrossRef]
  110. Kongkapan, J.; Poapolathep, S.; Isariyodom, S.; Kumagai, S.; Poapolathep, A. Simultaneous detection of multiple mycotoxins in broiler feeds using a liquid chromatography tandem-mass spectrometry. J. Vet. Med. Sci. 2015, 78, 259–264. [Google Scholar] [CrossRef] [Green Version]
  111. Romera, D.; Mateo, E.M.; Mateo-Castro, R.; Gomez, J.V.; Gimeno-Adelantado, J.V.; Jimenez, M. Determination of multiple mycotoxins in feedstuffs by combined use of UPLC–MS/MS and UPLC–QTOF–MS. Food Chem. 2018, 267, 140–148. [Google Scholar] [CrossRef]
  112. Fan, Z.; Bai, B.; Jin, P.; Fan, K.; Guo, W.; Zhao, Z.; Han, Z. Development and validation of an ultra-high performance liquid chromatography-tandem mass spectrometry method for simultaneous determination of four type B trichothecenes and masked deoxynivalenol in various feed products. Molecules 2016, 21, 747. [Google Scholar] [CrossRef]
  113. Makau, C.M.; Matofari, J.W.; Muliro, P.S.; Bebe, B.O. Aflatoxin B1 and Deoxynivalenol contamination of dairy feeds and presence of Aflatoxin M 1 contamination in milk from smallholder dairy systems in Nakuru, Kenya. Int. J. Food Contam. 2016, 3, 6. [Google Scholar] [CrossRef] [Green Version]
  114. Birr, T.; Jensen, T.; Preußke, N.; Sönnichsen, F.D.; De Boevre, M.; De Saeger, S.; Hasler, M.; Verreet, J.-A.; Klink, H. Occurrence of Fusarium Mycotoxins and Their Modified Forms in Forage Maize Cultivars. Toxins 2021, 13, 110. [Google Scholar] [CrossRef]
  115. Changwa, R.; Abia, W.; Msagati, T.; Nyoni, H.; Ndleve, K.; Njobeh, P. Multi-mycotoxin occurrence in dairy cattle feeds from the gauteng province of South Africa: A pilot study using UHPLC-QTOF-MS/MS. Toxins 2018, 10, 294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Carvalho, B.F.; Ávila, C.L.S.; Krempser, P.M.; Batista, L.R.; Pereira, M.N.; Schwan, R.F. Occurrence of mycotoxins and yeasts and moulds identification in corn silages in tropical climate. J. Appl. Microbiol. 2016, 120, 1181–1192. [Google Scholar] [CrossRef] [PubMed]
  117. Dagnac, T.; Latorre, A.; Fernández Lorenzo, B.; Llompart, M. Validation and application of a liquid chromatography-tandem mass spectrometry based method for the assessment of the co-occurrence of mycotoxins in maize silages from dairy farms in NW Spain. Food Addit. Contam. Part A 2016, 33, 1850–1863. [Google Scholar] [CrossRef]
  118. Cogan, T.; Hawkey, R.; Higgie, E.; Lee, M.R.F.; Mee, E.; Parfitt, D.; Raj, J.; Roderick, S.; Walker, N.; Ward, P. Silage and total mixed ration hygienic quality on commercial farms: Implications for animal production. Grass Forage Sci. 2017, 72, 601–613. [Google Scholar] [CrossRef] [Green Version]
  119. Vidal, A.; Marín, S.; Morales, H.; Ramos, A.J.; Sanchis, V. The fate of deoxynivalenol and ochratoxin A during the breadmaking process, effects of sourdough use and bran content. Food Chem. Toxicol. 2014, 68, 53–60. [Google Scholar] [CrossRef] [Green Version]
  120. Vidal, A.; Morales, H.; Sanchis, V.; Ramos, A.J.; Marín, S. Stability of DON and OTA during the breadmaking process and determination of process and performance criteria. Food Control 2014, 40, 234–242. [Google Scholar] [CrossRef] [Green Version]
  121. Berthiller, F.; Krska, R.; Domig, K.J.; Kneifel, W.; Juge, N.; Schuhmacher, R.; Adam, G. Hydrolytic fate of deoxynivalenol-3-glucoside during digestion. Toxicol. Lett. 2011, 206, 264–267. [Google Scholar] [CrossRef] [Green Version]
  122. Li, D.; Ye, Y.; Lin, S.; Deng, L.; Fan, X.; Zhang, Y.; Deng, X.; Li, Y.; Yan, H.; Ma, Y. Evaluation of deoxynivalenol-induced toxic effects on DF-1 cells in vitro: Cell-cycle arrest, oxidative stress, and apoptosis. Environ. Toxicol. Pharmacol. 2014, 37, 141–149. [Google Scholar] [CrossRef]
  123. Urbanek, K.A.; Habrowska-Górczyńska, D.E.; Kowalska, K.; Stańczyk, A.; Domińska, K.; Piastowska-Ciesielska, A.W. Deoxynivalenol as potential modulator of human steroidogenesis. J. Appl. Toxicol. 2018, 38, 1450–1459. [Google Scholar] [CrossRef] [PubMed]
  124. Payros, D.; Alassane-Kpembi, I.; Pierron, A.; Loiseau, N.; Pinton, P.; Oswald, I.P. Toxicology of deoxynivalenol and its acetylated and modified forms. Arch. Toxicol. 2016, 90, 2931–2957. [Google Scholar] [CrossRef] [PubMed]
  125. Schmeits, P.C.J.; Katika, M.R.; Peijnenburg, A.A.C.M.; van Loveren, H.; Hendriksen, P.J.M. DON shares a similar mode of action as the ribotoxic stress inducer anisomycin while TBTO shares ER stress patterns with the ER stress inducer thapsigargin based on comparative gene expression profiling in Jurkat T cells. Toxicol. Lett. 2014, 224, 395–406. [Google Scholar] [CrossRef]
  126. Mikami, O.; Yamaguchi, H.; Murata, H.; Nakajima, Y.; Miyazaki, S. Induction of apoptotic lesions in liver and lymphoid tissues and modulation of cytokine mRNA expression by acute exposure to deoxynivalenol in piglets. J. Vet. Sci. 2010, 11, 107–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. 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] [PubMed]
  128. Wu, Q.-H.; Wang, X.; Yang, W.; Nüssler, A.K.; Xiong, L.-Y.; Kuča, 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] [PubMed]
  129. Islam, Z.; Gray, J.S.; Pestka, J.J. p38 Mitogen-activated protein kinase mediates IL-8 induction by the ribotoxin deoxynivalenol in human monocytes. Toxicol. Appl. Pharmacol. 2006, 213, 235–244. [Google Scholar] [CrossRef]
  130. Sergent, T.; Parys, M.; Garsou, S.; Pussemier, L.; Schneider, Y.-J.; Larondelle, Y. Deoxynivalenol transport across human intestinal Caco-2 cells and its effects on cellular metabolism at realistic intestinal concentrations. Toxicol. Lett. 2006, 164, 167–176. [Google Scholar] [CrossRef]
  131. Maresca, M.; Mahfoud, R.; Garmy, N.; Fantini, J. The mycotoxin deoxynivalenol affects nutrient absorption in human intestinal epithelial cells. J. Nutr. 2002, 132, 2723–2731. [Google Scholar] [CrossRef] [Green Version]
  132. Froquet, R.; Sibiril, Y.; Parent-Massin, D. Trichothecene toxicity on human megakaryocyte progenitors (CFU-MK). Hum. Exp. Toxicol. 2001, 20, 84–89. [Google Scholar] [CrossRef]
  133. Le Drean, G.; Auffret, M.; Batina, P.; Arnold, F.; Sibiril, Y.; Arzur, D.; Parent-Massin, D. Myelotoxicity of trichothecenes and apoptosis: An in vitro study on human cord blood CD34+ hematopoietic progenitor. Toxicol. In Vitro 2005, 19, 1015–1024. [Google Scholar] [CrossRef] [PubMed]
  134. Bensassi, F.; El Golli-Bennour, E.; Abid-Essefi, S.; Bouaziz, C.; Hajlaoui, M.R.; Bacha, H. Pathway of deoxynivalenol-induced apoptosis in human colon carcinoma cells. Toxicology 2009, 264, 104–109. [Google Scholar] [CrossRef] [PubMed]
  135. Warth, B.; Sulyok, M.; Fruhmann, P.; Berthiller, F.; Schuhmacher, R.; Hametner, C.; Adam, G.; Fröhlich, J.; Krska, R. Assessment of human deoxynivalenol exposure using an LC–MS/MS based biomarker method. Toxicol. Lett. 2012, 211, 85–90. [Google Scholar] [CrossRef] [PubMed]
  136. Pestka, J.J. Deoxynivalenol: Mechanisms of action, human exposure, and toxicological relevance. Arch. Toxicol. 2010, 84, 663–679. [Google Scholar] [CrossRef]
  137. Mishra, S.; Srivastava, S.; Dewangan, J.; Divakar, A.; Kumar Rath, S. Global occurrence of deoxynivalenol in food commodities and exposure risk assessment in humans in the last decade: A survey. Crit. Rev. Food Sci. Nutr. 2020, 60, 1346–1374. [Google Scholar] [CrossRef]
  138. Yao, Y.; Long, M. The biological detoxification of deoxynivalenol: A review. Food Chem. Toxicol. 2020, 145, 111649. [Google Scholar] [CrossRef]
  139. Feizollahi, E.; Roopesh, M.S. Mechanisms of deoxynivalenol (DON) degradation during different treatments: A review. Crit. Rev. Food Sci. Nutr. 2021, 1–22. [Google Scholar] [CrossRef]
  140. Bracarense, A.-P.F.; Lucioli, J.; Grenier, B.; Pacheco, G.D.; Moll, W.-D.; Schatzmayr, G.; Oswald, I.P. Chronic ingestion of deoxynivalenol and fumonisin, alone or in interaction, induces morphological and immunological changes in the intestine of piglets. Br. J. Nutr. 2012, 107, 1776–1786. [Google Scholar] [CrossRef] [Green Version]
  141. Ostry, V.; Malir, F.; Toman, J.; Grosse, Y. Mycotoxins as human carcinogens—The IARC Monographs classification. Mycotoxin Res. 2017, 33, 65–73. [Google Scholar] [CrossRef]
  142. Knutsen, H.K.; Alexander, J.; Barregård, L.; Bignami, M.; Brüschweiler, B.; Ceccatelli, S.; Cottrill, B.; Dinovi, M.; Grasl-Kraupp, B. EFSA Panel on Contaminants in the Food Chain. Risks to human and animal health related to the presence of deoxynivalenol and its acetylated and modified forms in food and feed. EFSA J. 2017, 15, e04718. [Google Scholar]
  143. Eriksen, G.S.; Pettersson, H. Toxicological evaluation of trichothecenes in animal feed. Anim. Feed Sci. Technol. 2004, 114, 205–239. [Google Scholar] [CrossRef]
  144. Huang, M.C.; Furr, J.R.; Robinson, V.G.; Betz, L.; Shockley, K.; Cunny, H.; Witt, K.; Waidyanatha, S.; Germolec, D. Oral deoxynivalenol toxicity in Harlan Sprague Dawley (Hsd: Sprague Dawley® SD®) rat dams and their offspring. Food Chem. Toxicol. 2021, 148, 111963. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, X.; Chen, X.; Cao, L.; Zhu, L.; Zhang, Y.; Chu, X.; Zhu, D.; ur Rahman, S.; Peng, C.; Feng, S. Mechanism of deoxynivalenol-induced neurotoxicity in weaned piglets is linked to lipid peroxidation, dampened neurotransmitter levels, and interference with calcium signaling. Ecotoxicol. Environ. Saf. 2020, 194, 110382. [Google Scholar] [CrossRef] [PubMed]
  146. Tsien, R.W.; Tsien, R.Y. Calcium channels, stores, and oscillations. Annu. Rev. Cell Biol. 1990, 6, 715–760. [Google Scholar] [CrossRef] [PubMed]
  147. Adelsberger, H.; Garaschuk, O.; Konnerth, A. Cortical calcium waves in resting newborn mice. Nat. Neurosci. 2005, 8, 988–990. [Google Scholar] [CrossRef] [PubMed]
  148. Mayer, E.; Novak, B.; Springler, A.; Schwartz-Zimmermann, H.E.; Nagl, V.; Reisinger, N.; Hessenberger, S.; Schatzmayr, G. Effects of deoxynivalenol (DON) and its microbial biotransformation product deepoxy-deoxynivalenol (DOM-1) on a trout, pig, mouse, and human cell line. Mycotoxin Res. 2017, 33, 297–308. [Google Scholar] [CrossRef] [Green Version]
  149. Seeling, K.; Lebzien, P.; Dänicke, S.; Spilke, J.; Südekum, K.H.; Flachowsky, G. Effects of level of feed intake and Fusarium toxin-contaminated wheat on rumen fermentation as well as on blood and milk parameters in cows. J. Anim. Physiol. Anim. Nutr. 2006, 90, 103–115. [Google Scholar] [CrossRef]
  150. Lee, J.-Y.; Lim, W.; Park, S.; Kim, J.; You, S.; Song, G. Deoxynivalenol induces apoptosis and disrupts cellular homeostasis through MAPK signaling pathways in bovine mammary epithelial cells. Environ. Pollut. 2019, 252, 879–887. [Google Scholar] [CrossRef]
  151. Ok, H.E.; Lee, S.Y.; Chun, H.S. Occurrence and simultaneous determination of nivalenol and deoxynivalenol in rice and bran by HPLC-UV detection and immunoaffinity cleanup. Food Control 2018, 87, 53–59. [Google Scholar] [CrossRef]
  152. Ji, J.; Zhu, P.; Cui, F.; Pi, F.; Zhang, Y.; Sun, X. The disorder metabolic profiling in kidney and spleen of mice induced by mycotoxins deoxynivalenol through gas chromatography mass spectrometry. Chemosphere 2017, 180, 267–274. [Google Scholar] [CrossRef]
  153. Flores-Flores, M.E.; González-Peñas, E. An LC–MS/MS method for multi-mycotoxin quantification in cow milk. Food Chem. 2017, 218, 378–385. [Google Scholar] [CrossRef] [PubMed]
  154. Schlegel, K.M.; Elsinghorst, P.W. Myco-DES: Enabling remote extraction of mycotoxins for robust and reliable quantification by stable isotope dilution LC–MS/MS. Anal. Chem. 2020, 92, 5387–5395. [Google Scholar] [CrossRef]
  155. Ran, L.; Ze-Jia, L.; Jin-Yi, Y.; Zhen-Lin, X.; Hong, W.; Hong-Tao, L.; Yuan-Ming, S.; Yu-Dong, S. An indirect competitive enzyme-linked immunosorbent assay for simultaneous determination of florfenicol and thiamphenicol in animal meat and urine. Chin. J. Anal. Chem. 2018, 46, 1321–1328. [Google Scholar]
  156. Kong, D.; Wu, X.; Li, Y.; Liu, L.; Song, S.; Zheng, Q.; Kuang, H.; Xu, C. Ultrasensitive and eco-friendly immunoassays based monoclonal antibody for detection of deoxynivalenol in cereal and feed samples. Food Chem. 2019, 270, 130–137. [Google Scholar] [CrossRef] [PubMed]
  157. Guo, L.; Shao, Y.; Duan, H.; Ma, W.; Leng, Y.; Huang, X.; Xiong, Y. Magnetic quantum dot nanobead-based fluorescent immunochromatographic assay for the highly sensitive detection of aflatoxin B1 in dark soy sauce. Anal. Chem. 2019, 91, 4727–4734. [Google Scholar] [CrossRef]
  158. Li, J.; Yan, H.; Tan, X.; Lu, Z.; Han, H. Cauliflower-inspired 3D SERS substrate for multiple mycotoxins detection. Anal. Chem. 2019, 91, 3885–3892. [Google Scholar] [CrossRef]
  159. Zhang, W.; Tang, S.; Jin, Y.; Yang, C.; He, L.; Wang, J.; Chen, Y. Multiplex SERS-based lateral flow immunosensor for the detection of major mycotoxins in maize utilizing dual Raman labels and triple test lines. J. Hazard. Mater. 2020, 393, 122348. [Google Scholar] [CrossRef]
  160. Chen, Y.; Yang, Y.; Wang, Y.; Peng, Y.; Nie, J.; Gao, G.; Zhi, J. Development of an Escherichia coli-based electrochemical biosensor for mycotoxin toxicity detection. Bioelectrochemistry 2020, 133, 107453. [Google Scholar] [CrossRef]
  161. Urusov, A.E.; Petrakova, A.V.; Zherdev, A.V.; Dzantiev, B.B. “Multistage in one touch” design with a universal labelling conjugate for high-sensitive lateral flow immunoassays. Biosens. Bioelectron. 2016, 86, 575–579. [Google Scholar] [CrossRef]
  162. Xu, S.; Zhang, G.; Fang, B.; Xiong, Q.; Duan, H.; Lai, W. Lateral flow immunoassay based on polydopamine-coated gold nanoparticles for the sensitive detection of zearalenone in maize. ACS Appl. Mater. Interfaces 2019, 11, 31283–31290. [Google Scholar] [CrossRef]
  163. Goud, K.Y.; Kailasa, S.K.; Kumar, V.; Tsang, Y.F.; Gobi, K.V.; Kim, K.-H. Progress on nanostructured electrochemical sensors and their recognition elements for detection of mycotoxins: A review. Biosens. Bioelectron. 2018, 121, 205–222. [Google Scholar] [CrossRef] [PubMed]
  164. Hossain, M.Z.; Maragos, C.M. Gold nanoparticle-enhanced multiplexed imaging surface plasmon resonance (iSPR) detection of Fusarium mycotoxins in wheat. Biosens. Bioelectron. 2018, 101, 245–252. [Google Scholar] [CrossRef] [PubMed]
  165. Machado, J.M.D.; Soares, R.R.G.; Chu, V.; Conde, J.P. Multiplexed capillary microfluidic immunoassay with smartphone data acquisition for parallel mycotoxin detection. Biosens. Bioelectron. 2018, 99, 40–46. [Google Scholar] [CrossRef] [PubMed]
  166. Raeisossadati, M.J.; Danesh, N.M.; Borna, F.; Gholamzad, M.; Ramezani, M.; Abnous, K.; Taghdisi, S.M. Lateral flow based immunobiosensors for detection of food contaminants. Biosens. Bioelectron. 2016, 86, 235–246. [Google Scholar] [CrossRef] [PubMed]
  167. Jin, Y.; Chen, Q.; Luo, S.; He, L.; Fan, R.; Zhang, S.; Yang, C.; Chen, Y. Dual near-infrared fluorescence-based lateral flow immunosensor for the detection of zearalenone and deoxynivalenol in maize. Food Chem. 2021, 336, 127718. [Google Scholar] [CrossRef] [PubMed]
  168. Zhao, S.; Bu, T.; He, K.; Bai, F.; Zhang, M.; Tian, Y.; Sun, X.; Wang, X.; Zhangsun, H.; Wang, L. A novel α-Fe2O3 nanocubes-based multiplex immunochromatographic assay for simultaneous detection of deoxynivalenol and aflatoxin B1 in food samples. Food Control 2021, 123, 107811. [Google Scholar] [CrossRef]
  169. Seo, S.E.; Tabei, F.; Park, S.J.; Askarian, B.; Kim, K.H.; Moallem, G.; Chong, J.W.; Kwon, O.S. Smartphone with optical, physical, and electrochemical nanobiosensors. J. Ind. Eng. Chem. 2019, 77, 1–11. [Google Scholar] [CrossRef]
  170. Zhang, K.; Ge, Y.; He, S.; Ge, F.; Huang, Q.; Huang, Z.; Wang, X.; Wen, Y.; Wang, B. Development of New Electrochemical Sensor based on Kudzu Vine Biochar Modified Flexible Carbon Electrode for Portable Wireless Intelligent Analysis of Clenbuterol. Int. J. Electrochem. Sci. 2020, 15, 7326–7336. [Google Scholar] [CrossRef]
  171. Ong, C.C.; Sangu, S.S.; Illias, N.M.; Gopinath, S.C.B.; Saheed, M.S.M. Iron nanoflorets on 3D-graphene-nickel: A ‘dandelion’nanostructure for selective deoxynivalenol detection. Biosens. Bioelectron. 2020, 154, 112088. [Google Scholar] [CrossRef]
  172. Li, W.; Diao, K.; Qiu, D.; Zeng, Y.; Tang, K.; Zhu, Y.; Sheng, Y.; Wen, Y.; Li, M. A highly-sensitive and selective antibody-like sensor based on molecularly imprinted poly (L-arginine) on COOH-MWCNTs for electrochemical recognition and detection of deoxynivalenol. Food Chem. 2021, 350, 129229. [Google Scholar] [CrossRef]
  173. Hristov, D.R.; Rodriguez-Quijada, C.; Gomez-Marquez, J.; Hamad-Schifferli, K. Designing paper-based immunoassays for biomedical applications. Sensors 2019, 19, 554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Huang, X.; Huang, X.; Xie, J.; Li, X.; Huang, Z. Rapid simultaneous detection of fumonisin B1 and deoxynivalenol in grain by immunochromatographic test strip. Anal. Biochem. 2020, 606, 113878. [Google Scholar] [CrossRef] [PubMed]
  175. Shao, Y.; Duan, H.; Zhou, S.; Ma, T.; Guo, L.; Huang, X.; Xiong, Y. Biotin–streptavidin system-mediated ratiometric multiplex immunochromatographic assay for simultaneous and accurate quantification of three mycotoxins. J. Agric. Food Chem. 2019, 67, 9022–9031. [Google Scholar] [CrossRef] [PubMed]
  176. Goryacheva, O.A.; Beloglazova, N.V.; Goryacheva, I.Y.; De Saeger, S. Homogenous FRET-based fluorescent immunoassay for deoxynivalenol detection by controlling the distance of donor-acceptor couple. Talanta 2021, 225, 121973. [Google Scholar] [CrossRef] [PubMed]
  177. Li, R.; Bu, T.; Zhao, Y.; Sun, X.; Wang, Q.; Tian, Y.; Bai, F.; Wang, L. Polydopamine coated zirconium metal-organic frameworks-based immunochromatographic assay for highly sensitive detection of deoxynivalenol. Anal. Chim. Acta 2020, 1131, 109–117. [Google Scholar] [CrossRef]
  178. Chang, X.; Zhang, Y.; Liu, H.; Tao, X. A quadruple-label time-resolved fluorescence immunochromatographic assay for simultaneous quantitative determination of three mycotoxins in grains. Anal. Methods 2020, 12, 247–254. [Google Scholar] [CrossRef]
  179. Cheng, N.; Shi, Q.; Zhu, C.; Li, S.; Lin, Y.; Du, D. Pt–Ni(OH)2 nanosheets amplified two-way lateral flow immunoassays with smartphone readout for quantification of pesticides. Biosens. Bioelectron. 2019, 142, 111498. [Google Scholar] [CrossRef]
  180. Huang, X.; Huang, T.; Li, X.; Huang, Z. Flower-like gold nanoparticles-based immunochromatographic test strip for rapid simultaneous detection of fumonisin B1 and deoxynivalenol in Chinese traditional medicine. J. Pharm. Biomed. Anal. 2020, 177, 112895. [Google Scholar] [CrossRef]
  181. Subak, H.; Selvolini, G.; Macchiagodena, M.; Ozkan-Ariksoysal, D.; Pagliai, M.; Procacci, P.; Marrazza, G. Mycotoxins aptasensing: From molecular docking to electrochemical detection of deoxynivalenol. Bioelectrochemistry 2021, 138, 107691. [Google Scholar] [CrossRef]
  182. Berthiller, F.; Dall’Asta, C.; Schuhmacher, R.; Lemmens, M.; Adam, G.; Krska, R. Masked mycotoxins: Determination of a deoxynivalenol glucoside in artificially and naturally contaminated wheat by liquid chromatography− tandem mass spectrometry. J. Agric. Food Chem. 2005, 53, 3421–3425. [Google Scholar] [CrossRef]
  183. De Angelis, E.; Monaci, L.; Pascale, M.; Visconti, A. Fate of deoxynivalenol, T-2 and HT-2 toxins and their glucoside conjugates from flour to bread: An investigation by high-performance liquid chromatography high-resolution mass spectrometry. Food Addit. Contam. Part A 2013, 30, 345–355. [Google Scholar] [CrossRef] [PubMed]
  184. Rychlik, M.; Humpf, H.-U.; Marko, D.; Dänicke, S.; Mally, A.; Berthiller, F.; Klaffke, H.; Lorenz, N. Proposal of a comprehensive definition of modified and other forms of mycotoxins including “masked” mycotoxins. Mycotoxin Res. 2014, 30, 197–205. [Google Scholar] [CrossRef] [Green Version]
  185. Berthiller, F.; Crews, C.; Dall’Asta, C.; Saeger, S.D.; Haesaert, G.; Karlovsky, P.; Oswald, I.P.; Seefelder, W.; Speijers, G.; Stroka, J. Masked mycotoxins: A review. Mol. Nutr. Food Res. 2013, 57, 165–186. [Google Scholar] [CrossRef]
  186. Nagl, V.; Woechtl, B.; Schwartz-Zimmermann, H.E.; Hennig-Pauka, I.; Moll, W.-D.; Adam, G.; Berthiller, F. Metabolism of the masked mycotoxin deoxynivalenol-3-glucoside in pigs. Toxicol. Lett. 2014, 229, 190–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Freire, L.; Sant’Ana, A.S. Modified mycotoxins: An updated review on their formation, detection, occurrence, and toxic effects. Food Chem. Toxicol. 2018, 111, 189–205. [Google Scholar] [CrossRef] [PubMed]
  188. Tian, Y.; Tan, Y.; Liu, N.; Yan, Z.; Liao, Y.; Chen, J.; De Saeger, S.; Yang, H.; Zhang, Q.; Wu, A. Detoxification of deoxynivalenol via glycosylation represents novel insights on antagonistic activities of Trichoderma when confronted with Fusarium graminearum. Toxins 2016, 8, 335. [Google Scholar] [CrossRef] [Green Version]
  189. Khaneghah, A.M.; Fakhri, Y.; Raeisi, S.; Armoon, B.; Sant’Ana, A.S. Prevalence and concentration of ochratoxin A, zearalenone, deoxynivalenol and total aflatoxin in cereal-based products: A systematic review and meta-analysis. Food Chem. Toxicol. 2018, 118, 830–848. [Google Scholar] [CrossRef]
  190. Broekaert, N.; Devreese, M.; De Mil, T.; Fraeyman, S.; Antonissen, G.; De Baere, S.; De Backer, P.; Vermeulen, A.; Croubels, S. Oral bioavailability, hydrolysis, and comparative toxicokinetics of 3-acetyldeoxynivalenol and 15-acetyldeoxynivalenol in broiler chickens and pigs. J. Agric. Food Chem. 2015, 63, 8734–8742. [Google Scholar] [CrossRef]
  191. Berthiller, F.; Schuhmacher, R.; Adam, G.; Krska, R. Formation, determination and significance of masked and other conjugated mycotoxins. Anal. Bioanal. Chem. 2009, 395, 1243–1252. [Google Scholar] [CrossRef]
  192. Olopade, B.K.; Oranusi, S.U.; Nwinyi, O.C.; Gbashi, S.; Njobeh, P.B. Occurrences of Deoxynivalenol, Zearalenone and some of their masked forms in selected cereals from Southwest Nigeria. NFS J. 2021, 23, 24–29. [Google Scholar] [CrossRef]
  193. Lorenz, N.; Dänicke, S.; Edler, L.; Gottschalk, C.; Lassek, E.; Marko, D.; Rychlik, M.; Mally, A. A critical evaluation of health risk assessment of modified mycotoxins with a special focus on zearalenone. Mycotoxin Res. 2019, 35, 27–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Lu, Q.; Qin, J.-A.; Fu, Y.-W.; Luo, J.-Y.; Lu, J.-H.; Logrieco, A.F.; Yang, M.-H. Modified mycotoxins in foodstuffs, animal feed, and herbal medicine: A systematic review on global occurrence, transformation mechanism and analysis methods. Trends Analyt. Chem. 2020, 133, 116088. [Google Scholar] [CrossRef]
  195. Vidal, A.; Marín, S.; Sanchis, V.; De Saeger, S.; De Boevre, M. Hydrolysers of modified mycotoxins in maize: α-Amylase and cellulase induce an underestimation of the total aflatoxin content. Food Chem. 2018, 248, 86–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Beloglazova, N.V.; De Boevre, M.; Goryacheva, I.Y.; Werbrouck, S.; Guo, Y.; De Saeger, S. Immunochemical approach for zearalenone-4-glucoside determination. Talanta. 2013, 106, 422–430. [Google Scholar] [CrossRef]
  197. European Commission. Recommendation of 17 August 2006 on the presence of deoxynivalenol, zearalenone, ochratoxin A, T-2 and HT-2 and fumonisins in products intended for animal feeding. Off. J. Eur. Union 2006, 229, 7–9. Available online: https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2006:229:0007:0009:EN:PDF (accessed on 23 August 2006).
  198. Awad, W.A.; Ghareeb, K.; Böhm, J.; Zentek, J. Decontamination and detoxification strategies for the Fusarium mycotoxin deoxynivalenol in animal feed and the effectiveness of microbial biodegradation. Food Addit. Contam. 2010, 27, 510–520. [Google Scholar] [CrossRef]
  199. Zhang, H.; Dong, M.; Yang, Q.; Apaliya, M.T.; Li, J.; Zhang, X. Biodegradation of zearalenone by Saccharomyces cerevisiae: Possible involvement of ZEN responsive proteins of the yeast. J. Proteom. 2016, 143, 416–423. [Google Scholar] [CrossRef]
  200. Schaarschmidt, S.; Fauhl-Hassek, C. The fate of mycotoxins during secondary food processing of maize for human consumption. Compr. Rev. Food Sci. Food Saf. 2021, 20, 91–148. [Google Scholar] [CrossRef]
  201. Stadler, D.; Berthiller, F.; Suman, M.; Schuhmacher, R.; Krska, R. Novel analytical methods to study the fate of mycotoxins during thermal food processing. Anal. Bioanal. Chem. 2020, 412, 9–16. [Google Scholar] [CrossRef] [Green Version]
  202. Stadler, D.; Lambertini, F.; Woelflingseder, L.; Schwartz-Zimmermann, H.; Marko, D.; Suman, M.; Berthiller, F.; Krska, R. The Influence of processing parameters on the mitigation of deoxynivalenol during industrial baking. Toxins 2019, 11, 317. [Google Scholar] [CrossRef] [Green Version]
  203. Vidal, A.; Sanchis, V.; Ramos, A.J.; Marín, S. Thermal stability and kinetics of degradation of deoxynivalenol, deoxynivalenol conjugates and ochratoxin A during baking of wheat bakery products. Food Chem. 2015, 178, 276–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Kalagatur, N.K.; Kamasani, J.R.; Mudili, V.; Krishna, K.; Chauhan, O.P.; Sreepathi, M.H. Effect of high pressure processing on growth and mycotoxin production of Fusarium graminearum in maize. Food Biosci. 2018, 21, 53–59. [Google Scholar] [CrossRef]
  205. Vidal, A.; Bendicho, J.; Sanchis, V.; Ramos, A.J.; Marín, S. Stability and kinetics of leaching of deoxynivalenol, deoxynivalenol-3-glucoside and ochratoxin A during boiling of wheat spaghettis. Food Res. Int. 2016, 85, 182–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Jajić, I.; Jakšić, S.; Krstović, S.; Abramović, B. Preliminary results on deoxynivalenol degradation in maize by UVA and UVC irradiation. Contemp. Agric. 2016, 65, 7–12. [Google Scholar] [CrossRef] [Green Version]
  207. Popović, V.; Fairbanks, N.; Pierscianowski, J.; Biancaniello, M.; Zhou, T.; Koutchma, T. Feasibility of 3D UV-C treatment to reduce fungal growth and mycotoxin loads on maize and wheat kernels. Mycotoxin Res. 2018, 34, 211–221. [Google Scholar] [CrossRef]
  208. Yi-gang, Y.; Han-ruo, M.; Rui, H.; Yu-qian, T.; Xing-long, X. Degradation of Deoxynivalenol in Flour by Ozone and Ultraviolet Light and their Effects on Flour Quality. Mod. Food Sci. Technol. 2016, 32, 196–202. [Google Scholar]
  209. Rempe, I.; Kersten, S.; Valenta, H.; Dänicke, S. Hydrothermal treatment of naturally contaminated maize in the presence of sodium metabisulfite, methylamine and calcium hydroxide; effects on the concentration of zearalenone and deoxynivalenol. Mycotoxin Res. 2013, 29, 169–175. [Google Scholar] [CrossRef]
  210. Santos Alexandre, A.P.; Vela-Paredes, R.S.; Santos, A.S.; Costa, N.S.; Canniatti-Brazaca, S.G.; Calori-Domingues, M.A.; Augusto, P.E.D. Ozone treatment to reduce deoxynivalenol (DON) and zearalenone (ZEN) contamination in wheat bran and its impact on nutritional quality. Food Addit. Contam. Part A 2018, 35, 1189–1199. [Google Scholar] [CrossRef]
  211. Sun, X.; Ji, J.; Gao, Y.; Zhang, Y.; Zhao, G.; Sun, C. Fate of deoxynivalenol and degradation products degraded by aqueous ozone in contaminated wheat. Food Res. Int. 2020, 137, 109357. [Google Scholar] [CrossRef]
  212. Lyu, F.; Gao, F.; Zhou, X.; Zhang, J.; Ding, Y. Using acid and alkaline electrolyzed water to reduce deoxynivalenol and mycological contaminations in wheat grains. Food Control 2018, 88, 98–104. [Google Scholar] [CrossRef]
  213. Borràs-Vallverdú, B.; Ramos, A.J.; Marín, S.; Sanchis, V.; Rodríguez-Bencomo, J.J. Deoxynivalenol degradation in wheat kernels by exposition to ammonia vapours: A tentative strategy for detoxification. Food Control 2020, 118, 107444. [Google Scholar] [CrossRef]
  214. Ji, C.; Fan, Y.; Zhao, L. Review on biological degradation of mycotoxins. Anim. Nutr. 2016, 2, 127–133. [Google Scholar] [CrossRef] [PubMed]
  215. Wu, S.; Wang, F.; Li, Q.; Wang, J.; Zhou, Y.; Duan, N.; Niazi, S.; Wang, Z. Photocatalysis and degradation products identification of deoxynivalenol in wheat using upconversion nanoparticles@TiO2 composite. Food Chem. 2020, 323, 126823. [Google Scholar] [CrossRef] [PubMed]
  216. Hathout, A.S.; Aly, S.E. Biological detoxification of mycotoxins: A review. Ann. Microbiol. 2014, 64, 905–919. [Google Scholar] [CrossRef]
  217. Li, W.; Yu, S.Y.; Cheng, P.; Zhang, J.B. Screening, identification, and application of deoxynivalenol-detoxifying bacteria from mildewed alfalfa silage. Int. Food Res. J. 2020, 27, 350–356. [Google Scholar]
  218. Palazzini, J.; Roncallo, P.; Cantoro, R.; Chiotta, M.; Yerkovich, N.; Palacios, S.; Echenique, V.; Torres, A.; Ramirez, M.; Karlovsky, P. Biocontrol of Fusarium graminearum sensu stricto, reduction of deoxynivalenol accumulation and phytohormone induction by two selected antagonists. Toxins 2018, 10, 88. [Google Scholar] [CrossRef] [Green Version]
  219. Jia, R.; Cao, L.; Liu, W.; Shen, Z. Detoxification of deoxynivalenol by Bacillus subtilis ASAG 216 and characterization the degradation process. Eur. Food Res. Technol. 2020, 247, 67–76. [Google Scholar] [CrossRef]
  220. Wang, S.; Hou, Q.; Guo, Q.; Zhang, J.; Sun, Y.; Wei, H.; Shen, L. Isolation and characterization of a deoxynivalenol-degrading bacterium Bacillus licheniformis YB9 with the capability of modulating intestinal microbial flora of mice. Toxins 2020, 12, 184. [Google Scholar] [CrossRef] [Green Version]
  221. Wang, Y.; Wang, G.; Dai, Y.; Wang, Y.; Lee, Y.-W.; Shi, J.; Xu, J. Biodegradation of deoxynivalenol by a novel microbial consortium. Front. Microbiol. 2020, 10, 2964. [Google Scholar] [CrossRef] [Green Version]
  222. Wang, G.; Wang, Y.; Ji, F.; Xu, L.; Yu, M.; Shi, J.; Xu, J. Biodegradation of deoxynivalenol and its derivatives by Devosia insulae A16. Food Chem. 2019, 276, 436–442. [Google Scholar] [CrossRef]
  223. Gao, X.; Mu, P.; Zhu, X.; Chen, X.; Tang, S.; Wu, Y.; Miao, X.; Wang, X.; Wen, J.; Deng, Y. Dual Function of a Novel Bacterium, Slackia sp. D-G6: Detoxifying Deoxynivalenol and Producing the Natural Estrogen Analogue, Equol. Toxins 2020, 12, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Zhai, Y.; Zhong, L.; Gao, H.; Lu, Z.; Bie, X.; Zhao, H.; Zhang, C.; Lu, F. Detoxification of deoxynivalenol by a mixed culture of soil bacteria with 3-epi-deoxynivalenol as the main intermediate. Front. Microbiol. 2019, 10, 2172. [Google Scholar] [CrossRef] [PubMed]
  225. Zhang, J.; Qin, X.; Guo, Y.; Zhang, Q.; Ma, Q.; Ji, C.; Zhao, L. Enzymatic degradation of deoxynivalenol by a novel bacterium, Pelagibacterium halotolerans ANSP101. Food Chem. Toxicol. 2020, 140, 111276. [Google Scholar] [CrossRef] [PubMed]
  226. He, W.-J.; Shi, M.-M.; Yang, P.; Huang, T.; Zhao, Y.; Wu, A.-B.; Dong, W.-B.; Li, H.-P.; Zhang, J.-B.; Liao, Y.-C. A quinone-dependent dehydrogenase and two NADPH-dependent aldo/keto reductases detoxify deoxynivalenol in wheat via epimerization in a Devosia strain. Food Chem. 2020, 321, 126703. [Google Scholar] [CrossRef] [PubMed]
  227. 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]
  228. Li, Q.; Deng, Y.; Dai, S.; Wu, Y.; Li, W.; Zhuo, S.; Jiao, S.; Wang, S.; Jin, Y.; Li, J. Microfluidic Assembly Synthesis of Magnetic TiO2@SiO2 Hybrid Photonic Crystal Microspheres for Photocatalytic Degradation of Deoxynivalenol. J. Inorg. Organomet. Polym. Mater. 2020, 31, 2360–2367. [Google Scholar] [CrossRef]
  229. Zhang, J.; Liu, Y.; Li, Q.; Zhang, X.; Shang, J.K. Antifungal activity and mechanism of palladium-modified nitrogen-doped titanium oxide photocatalyst on agricultural pathogenic fungi Fusarium graminearum. ACS Appl. Mater. Interfaces 2013, 5, 10953–10959. [Google Scholar] [CrossRef]
  230. Sun, S.; Zhao, R.; Xie, Y.; Liu, Y. Photocatalytic degradation of aflatoxin B1 by activated carbon supported TiO2 catalyst. Food Control 2019, 100, 183–188. [Google Scholar] [CrossRef]
  231. Bai, X.; Sun, C.; Liu, D.; Luo, X.; Li, D.; Wang, J.; Wang, N.; Chang, X.; Zong, R.; Zhu, Y. Photocatalytic degradation of deoxynivalenol using graphene/ZnO hybrids in aqueous suspension. Appl. Catal. B Environ. 2017, 204, 11–20. [Google Scholar] [CrossRef] [Green Version]
  232. Wang, H.; Mao, J.; Zhang, Z.; Zhang, Q.; Zhang, L.; Zhang, W.; Li, P. Photocatalytic degradation of deoxynivalenol over dendritic-like α-Fe2O3 under visible light irradiation. Toxins 2019, 11, 105. [Google Scholar] [CrossRef] [Green Version]
  233. Xiong, S.; Li, X.; Zhao, C.; Gao, J.; Yuan, W.; Zhang, J. The degradation of deoxynivalenol by using electrochemical oxidation with graphite electrodes and the toxicity assessment of degradation products. Toxins 2019, 11, 478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Feizollahi, E.; Iqdiam, B.; Vasanthan, T.; Thilakarathna, M.S.; Roopesh, M.S. Effects of atmospheric-pressure cold plasma treatment on deoxynivalenol degradation, quality parameters, and germination of barley grains. Appl. Sci. 2020, 10, 3530. [Google Scholar] [CrossRef]
  235. Murugesan, P.; Brunda, D.K.; Moses, J.; Anandharamakrishnan, C. Photolytic and photocatalytic detoxification of mycotoxins in foods. Food Control 2021, 123, 107748. [Google Scholar] [CrossRef]
  236. 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] [PubMed]
  237. Gromadzka, K.; Górna, K.; Chełkowski, J.; Waśkiewicz, A. Mycotoxins and related Fusarium species in preharvest maize ear rot in Poland. Plant Soil Environ. 2016, 62, 348–354. [Google Scholar] [CrossRef] [Green Version]
  238. Adebiyi, J.A.; Kayitesi, E.; Adebo, O.A.; Changwa, R.; Njobeh, P.B. Food fermentation and mycotoxin detoxification: An African perspective. Food Control 2019, 106, 106731. [Google Scholar] [CrossRef]
  239. Mandappa, I.M.; Basavaraj, K.; Manonmani, H.K. Analysis of mycotoxins in fruit juices. In Fruit Juices; Elsevier: Amsterdam, The Netherlands, 2018; pp. 763–777. [Google Scholar]
  240. Degraeve, S.; Madege, R.; Audenaert, K.; Kamala, A.; Ortiz, J.; Kimanya, M.; Tiisekwa, B.; De Meulenaer, B.; Haesaert, G. Impact of local pre-harvest management practices in maize on the occurrence of Fusarium species and associated mycotoxins in two agro-ecosystems in Tanzania. Food Control 2016, 59, 225–233. [Google Scholar] [CrossRef]
  241. Thakare, D.; Zhang, J.; Wing, R.A.; Cotty, P.J.; Schmidt, M.A. Aflatoxin-free transgenic maize using host-induced gene silencing. Sci. Adv. 2017, 3, e1602382. [Google Scholar] [CrossRef] [Green Version]
  242. Munkvold, G.P.; Arias, S.; Taschl, I.; Gruber-Dorninger, C. Mycotoxins in corn: Occurrence, impacts, and management. In Corn; Serna-Saldivar, S.O., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 235–287. [Google Scholar]
  243. Hofer, K.; Barmeier, G.; Schmidhalter, U.; Habler, K.; Rychlik, M.; Hückelhoven, R.; Hess, M. Effect of nitrogen fertilization on Fusarium head blight in spring barley. Crop Prot. 2016, 88, 18–27. [Google Scholar] [CrossRef]
  244. Torres, A.M.; Barros, G.G.; Palacios, S.A.; Chulze, S.N.; Battilani, P. Review on pre-and post-harvest management of peanuts to minimize aflatoxin contamination. Food Res. Int. 2014, 62, 11–19. [Google Scholar] [CrossRef]
  245. Munkvold, G.P. Crop management practices to minimize the risk of mycotoxins contamination in temperate-zone maize. In Mycotoxin Reduction in grain Chains; Leslie, J.F., Logrieco, A.F., Eds.; Wiley: New York, NY, USA, 2014; pp. 59–75. [Google Scholar]
  246. World Health Organization (WHO). Prevention and Reduction of Food and Feed Contamination. 2012. Available online: http://www.fao.org/home/en/ (accessed on 10 October 2012).
  247. Pitt, J.I.; Taniwaki, M.H.; Cole, M.B. Mycotoxin production in major crops as influenced by growing, harvesting, storage and processing, with emphasis on the achievement of Food Safety Objectives. Food Control 2013, 32, 205–215. [Google Scholar] [CrossRef]
  248. 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]
  249. Channaiah, L.H.; Maier, D.E. Best stored maize management practices for the prevention of mycotoxin contamination. In Mycotoxin Reduction in Grain Chains; Leslie, J.F., Logrieco, A.F., Eds.; Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2014; p. 78. [Google Scholar] [CrossRef]
  250. Adegoke, G.O.; Letuma, P. Strategies for the prevention and reduction of mycotoxins in developing countries. In Mycotoxin and Food Safety in Developing Countries; Makun, H.A., Ed.; IntechOpen: London, UK, 2013; pp. 123–136. [Google Scholar]
Table 1. Occurrence of Deoxynivalenol in food and feed around the world.
Table 1. Occurrence of Deoxynivalenol in food and feed around the world.
Food/Feed MatrixCountryRange (μg/kg)Detection TechniqueReference
Food
Barley/Bakery productsArgentina2360HPLC-UV[59]
Brazil310–15,500LC-MS/MS[60]
Romania0–4000ELISA[61]
Tunisia500–3600HPLC[62]
Hungary97–3065HPLC[63]
CornSouth Korea3.3–232.56HPLC[64]
Corn/Corn germ mealChina100–4320.9/100–4402.7HPLC[65]
Corn flour/CornflakesSerbia931/878HPLC[66]
MaizeChina100–19,811HPLC-UV[67]
Egypt26–807LC-MS/MS[68]
Hungary225–2963ELISA[69]
Poland1–6688HPLC[70]
Serbia260.4–9050ELISA[71]
South Africa9176LC-MS/MS[72]
Nepal>1HPLC[73]
OatsCanada50–2340HPLC-PDA[74]
Finland21,608GC-MS[23]
Portugal17,900HPLC[75]
Russia50–1030HPLC-MS[76]
Sweden99–5544HPLC/ESI-MS/MS[77]
UK1866LC-MS/MS[78]
Noodles and PastaItaly35–450LC-MS/MS[79]
RicePakistan6.99LC-MS/MS[80]
WheatNetherlands100–11,000LC-MS/MS[81]
Serbia630–1840HPLC[82]
Iran23–1270ELISA[83]
Romania110–1787LC-MS/MS[84]
Uruguay1400–3400HPLC/UV[85]
Norway5–94HPLC[86]
Sweden1189HPLC/ESI-MS/MS[87]
Hungary1880ELISA[88]
Argentina9480LC-MS/MS[89]
Switzerland10,600LC-MS/MS[90]
Israel1.2–1746LC-MS/MS[90]
Nigeria119–2560LC-MS[91]
Brazil73–2794HPLC[92]
Poland10–1265HPLC[93]
China33–3030HPLC[94]
Sweden1189HPLC/ESI-MS/MS[87]
India70–4730HPLC[95]
Canada4700HPLC-PDA[74]
Slovakia788ELISA[96]
Spain6178HPLC[97]
Italy56–27,088GC-MS[98]
Serbia64–4808HPLC/ELISA[99]
Iran23–1270ELISA[83]
Finland5510LC-MS/MS[100]
Serbia154–16,528ELISA[101]
Albania1916LCMS[102]
Spring wheatLithuania100–10,644UPLC/MS[103]
Wheat dustBelgium607–14,043UPLC/MS[104]
Winter wheatLithuania100–1393UPLC/MS[103]
Winter wheatSlovak Republic20–2651.79HPLC-DAD[105]
Wheat flourSpain501HPLC[106]
Wheat flour and breadIran0.78ELISA[107]
Infant FoodUSA10–224HPLC-UV[108]
Infant FoodIndia5–228ELISA[109]
Barley/PastaRomania21.52–721.88/28.23–173.55ELISA and HPLC[21]
Flour and breakfast cerealsRomania31.56–172.37ELISA and HPLC[21]
Feed
Broiler feedsThailand33.58–60.81LC-MS[110]
Cattle compound feedSpain289.9UPLC–MS/MS and UPLC–QTOF–MS[111]
Cattle/Chicken/Pig feedSouth Korea91.65–950.25/3.3–603.10/32.38–932.48HPLC[64]
Concentrated feed/Formula feed/Premixed feedChina11.6–277.6/47.1–864.5/97.4–776.3UPHLC-MS[112]
Dairy concentrate feedKenya18.53–179.89ELISA[113]
Duck complete feedChina100–2613.7HPLC-UV[67]
Finished feedSouth Africa9805LC-MS[72]
Forage maizeNorthern Germany2237–3038LC-HRMS[114]
Compound feedsSouth Africa3.22–56.52UHPLC-MS/MS[115]
FeedEgypt1516LC-MS/MS[68]
Pig complete feed (powder)/(pellet)China100–2767.6/100–3346.0HPLC[65]
SilageBrazil300HPLC[116]
Spain43.1–6685.6LC-MS[117]
England10–7111UPLC[118]
Poland1–7, 860HPLC[70]
Poultry/Sheep/Swine compound feedSpain250/250/254.9UPLC–MS/MS and UPLC–QTOF–MS[111]
Swine feedHungary137–997ELISA[69]
Table 2. Permissible limits of DON in various foods and feeds established by different governing bodies.
Table 2. Permissible limits of DON in various foods and feeds established by different governing bodies.
Governing BodyFood/Feed TypePermissible Limit (mg/kg)Reference
European CommissionCereals and cereal-based products (except for maize by-products)8[197]
Maize by-products12
Complete feedingstuffs and complementary with the exception of:5
Complete feedingstuffs and complementary for pigs0.9
Complete feedingstuffs and complementary for calves (<4 months), lambs, and kids2
United States Food Drug & Administration (US-FDA)Wheat products (e.g., bran, flour, and germ) for human consumption1[137]
Food and Agricultural Organization (FAO) of the United States/Codex AlimentariusMaize, barley, and raw wheat2[137]
Maize or barley derived flour, semolina, meal, and flakes, wheat1
Cereal-based infant foods0.5
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kamle, M.; Mahato, D.K.; Gupta, A.; Pandhi, S.; Sharma, B.; Dhawan, K.; Vasundhara; Mishra, S.; Kumar, M.; Tripathi, A.D.; et al. Deoxynivalenol: An Overview on Occurrence, Chemistry, Biosynthesis, Health Effects and Its Detection, Management, and Control Strategies in Food and Feed. Microbiol. Res. 2022, 13, 292-314. https://doi.org/10.3390/microbiolres13020023

AMA Style

Kamle M, Mahato DK, Gupta A, Pandhi S, Sharma B, Dhawan K, Vasundhara, Mishra S, Kumar M, Tripathi AD, et al. Deoxynivalenol: An Overview on Occurrence, Chemistry, Biosynthesis, Health Effects and Its Detection, Management, and Control Strategies in Food and Feed. Microbiology Research. 2022; 13(2):292-314. https://doi.org/10.3390/microbiolres13020023

Chicago/Turabian Style

Kamle, Madhu, Dipendra Kumar Mahato, Akansha Gupta, Shikha Pandhi, Bharti Sharma, Kajal Dhawan, Vasundhara, Sadhna Mishra, Manoj Kumar, Abhishek Dutt Tripathi, and et al. 2022. "Deoxynivalenol: An Overview on Occurrence, Chemistry, Biosynthesis, Health Effects and Its Detection, Management, and Control Strategies in Food and Feed" Microbiology Research 13, no. 2: 292-314. https://doi.org/10.3390/microbiolres13020023

Article Metrics

Back to TopTop