Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Journal of Hazardous Materials 389 (2020) 122087

Contents lists available at ScienceDirect

Journal of Hazardous Materials


journal homepage: www.elsevier.com/locate/jhazmat

Review

Effects of mycotoxin-contaminated feed on farm animals T


a a, b,
Changwon Yang , Gwonhwa Song *, Whasun Lim **
a
Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of
Korea
b
Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea

GRAPHICAL ABSTRACT

ARTICLE INFO ABSTRACT

Editor: R. Debora Mycotoxins are secondary products produced by fungi in cereals and are frequently found in the livestock industry
Keywords: as contaminants of farm animal feed. Studies analyzing feed mycotoxins have been conducted worldwide and have
Toxin confirmed the presence of mycotoxins with biological activity, including aflatoxin, ochratoxin A, fumonisin, zear-
Farm animal alenone, and deoxynivalenol, in a large proportion of feed samples. Exposure to mycotoxins can cause im-
Pig munotoxicity and impair reproductive function in farm animals. In addition, exposure of tissues, such as the kid-
Poultry neys, liver, and intestines, to mycotoxins can exert histopathological changes that can interfere with animal growth
Aflatoxin and survival. This review describes previous studies regarding the presence of major mycotoxins in the feed of farm
animals, especially pigs and poultry. Moreover, it describes the adverse effects of mycotoxins in farm animals
following exposure, as well as the biological activity of mycotoxins in animal-derived cells. Mycotoxins have been
shown to regulate signaling pathways, oxidative stress, endoplasmic reticulum stress, apoptosis, and proliferation in
porcine and bovine cells. A clear understanding of the effects of mycotoxins on farm animals will help reduce farm
household economic loss and address the health concerns of people who consume these meat and dairy products.

1. Introduction environment at all stages of the food chain. Mycotoxin-contaminated


cereal grains and animal feed are frequently found throughout the
Mycotoxins, which are produced by fungi, are secondary metabo- world (Placinta et al., 1999). Humans and animals are exposed to
lites found in the food, feed, and living organisms of an agricultural mycotoxins through the consumption of contaminated food (Hussein


Corresponding author.
⁎⁎
Corresponding author at: Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul, 02707, Republic of Korea.
E-mail addresses: ghsong@korea.ac.kr (G. Song), wlim@kookmin.ac.kr (W. Lim).

https://doi.org/10.1016/j.jhazmat.2020.122087
Received 8 October 2019; Received in revised form 9 January 2020; Accepted 13 January 2020
Available online 20 January 2020
0304-3894/ © 2020 Elsevier B.V. All rights reserved.
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

and Brasel, 2001). The United States Food and Drug Administration target the toxic mechanisms of mycotoxins and have a beneficial impact
(FDA), the European Union (EU), and countries with advanced agri- on livestock health (Hou et al., 2018). Natural compounds extracted
cultural industries set guidelines for limiting concentrations to prevent from and fed to animals are potential candidates to mitigate mycotoxin
mycotoxin contamination in food and animal feed (Anukul et al., 2013; toxicity, minimizing side effects (Gan et al., 2018a; Qin et al., 2015).
Lee and Ryu, 2017). Many types of mycotoxins are found in human Melatonin supplementation is an emerging method shown to relieve
blood or breast milk samples (Breitholtz-Emanuelsson et al., 1993; Scott mycotoxin toxicity effects while inhibiting oxidative stress (Meki et al.,
et al., 1998). Mycotoxin levels are found to be higher in farm animal 2001; Gesing et al., 2006; Cheng et al., 2019). In this review, we de-
feed than in humans who consume relatively well-refined and pro- scribe the effects of compounds that can prevent the contamination or
cessed foods (Rodrigues and Naehrer, 2012). Therefore, mycotoxins are mitigate the toxicity of mycotoxins as feed additives.
prone to unprotected exposure among many environmental pollutants Among the different types of mycotoxins, only a few are known to
in farm animals. It is difficult to detect the presence of mycotoxins, cause toxicity via exposure to farm animals. These major mycotoxins
especially in farm regions with climates where the fungi that produce are the first to be identified when investigating mycotoxin contamina-
mycotoxins thrive. Pigs and poultry are the most susceptible and sen- tion in animal feeds worldwide, and they cause significant economic
sitive farm animals to the effects of mycotoxins. Therefore, the majority losses to livestock farms. Therefore, several studies examining the
of mycotoxin toxicity studies have been performed on these animals. toxicity of mycotoxins in farm animals have focused on the physiolo-
Since these species are also widely reared by humans for meat, eggs, gical action mechanisms of some mycotoxins. This review examines the
milk, and other processed foods, it is very important to measure the types and features of major mycotoxins that can be contaminants in
residual mycotoxin levels in the animal feed, tissues, and body fluids. farm animal feed, especially for pigs and poultry. It will also cover the
Animals that consume a mycotoxin-contaminated diet are adversely major studies on the types and concentrations of mycotoxins and their
affected by neoplastic, estrogenic, and immunosuppressive effects. metabolites, which are found in livestock tissues and body fluids. Next,
These excessive or persistent properties can result in weight loss and the we will describe how mycotoxins can have adverse immunological,
increased development of diseases resulting from immunosuppression endocrine, reproductive, genetic, and histopathological effects in farm
and reproductive disorders (D’Mello et al., 1999; Lindemann et al., animals. Avian, porcine, and bovine cells treated with different doses of
1993). mycotoxin are used to study apoptosis mechanisms mediated by oxi-
The health risks that mycotoxin exposure pose to livestock cause dative stress, autophagy, and inflammatory responses (Fig. 1). In ad-
economic loss, either by adverse effects on the animals themselves or by dition, in vivo and in vitro studies will be presented to identify which
the secondary cost to humans who eat contaminated animal-derived compounds are useful in mitigating the toxicity of mycotoxins.
foods. The agricultural environment affects the fungal habitat, and thus,
the number of mycotoxins produced. For example, in harvested grains, 2. Mycotoxins identified in animal feed
the presence of fumonisins, a group of mycotoxins, is high when dry
weather is followed by wet weather (Kendra and Dyer, 2007). Some 2.1. Occurrence of aflatoxins in feed
studies have reported that mycotoxin contamination in feed samples
depends on seasonal temperature conditions (Abidin et al., 2017). The most commonly found mycotoxins are aflatoxins, ochratoxin A
However, in Thailand, studies measuring the mycotoxin contamination (OTA), trichothecenes, zearalenone (ZEA), and fumonisins. The con-
in pasteurized milk suggest that mycotoxin contamination in dairy centration range of these mycotoxins in farm animal feeds and their
products are more affected by farm management factors than seasonal intracellular physiological action mechanisms have been reported.
effects (Suriyasathaporn and Nakprasert, 2012). Research has been Therefore, we focused on the concentration range of the five myco-
conducted to measure the concentration of mycotoxins in livestock feed toxins in animal feed and the body, and their toxicity mechanisms.
in numerous agricultural regions worldwide (Sarathchandra and Aflatoxin is produced by Aspergillus flavus or Aspergillus parasiticus and is
Muralimanohar, 2013; Monge et al., 2013; Shar et al., 2014; Sherazi suggested to have carcinogenic and mutagenic effects in humans, in-
et al., 2015). Important indicators in mycotoxin toxicity studies include cluding growth problems in children (Gong et al., 2004; Peters and
the level of mycotoxin contamination in livestock feed and the number Teel, 2003; Abnet, 2007). The FDA limits the aflatoxin levels to 20 ppb
of metabolites present in body tissues and fluids. Additionally, analyses in food and animal feeds and the EU limits the aflatoxin levels to 15
have been performed on residual mycotoxin levels in body fluids, such ppb, which is relatively concentration, although they vary by grain type
as blood and urine, and tissues, such as intestines and kidneys (Vulic and country (Alshannaq and Yu, 2017; Sarma et al., 2017). In poultry,
et al., 2012; Danicke and Winkler, 2015; Danicke and Brezina, 2013). aflatoxin in feed is metabolized into aflatoxin M and aflatoxicol and
Various biological, chemical, and physical methods have been ap- accumulates in organs, such as the gizzard, breast, and liver and also in
plied to prevent mycotoxin contamination or to eliminate toxicity (Yu eggs (Wolzak et al., 1985; Trucksess et al., 1983; Gregory and Manley,
et al., 2018; Zhai et al., 2018; Li et al., 2019; Elliott et al., 2019). As a 1982). Aflatoxin M1 (AFM1) is the hydroxylated metabolite of aflatoxin
feed additive, mycotoxin adsorbents, including phyllosilicate minerals, B1 (AFB1) and is found in dairy products derived from animals that have
activated charcoal, and synthetic resins, have been applied to reduce been fed with contaminated feed (Nakajima et al., 2004). AFM1 is a less
the risk of mycotoxin presence (Rosa et al., 2001; Raju and Devegowda, toxic form that is frequently found in animal milk. The process by
2000; Galvano et al., 1996). The addition of natural zeolite to the diet which AFB1 is metabolized in the body of animals involves the activity
of farm animals allows the consumed feed to adsorb polar mycotoxins of the cytochrome p450 (CYP450) family in the liver, which differs with
and mitigate toxicity (Katsoulos et al., 2016). The addition of sodium species (Deng et al., 2018). CYP1A2 is the major enzyme that converts
carbonate also helps to remove feed-contaminated mycotoxins (Polak AFB1 into AFM1. Another major pathway in which AFB1 is metabolized
et al., 2009). Gamma-irradiation is a major method for reducing my- is during the conversion to highly toxic exo-AFB1-8,9-epoxide (AFBO),
cotoxins in food and feed processing, although there is some debate which is epoxidated by the CYP450 family members such as CYP1A1,
about its effectiveness (Jalili et al., 2012; Herzallah et al., 2008; CYP1A2, and CYP2A6 (Dohnal et al., 2014). AFB1 is frequently found in
Hooshmand and Klopfenstein, 1995; Di Stefano et al., 2014). UV ra- cattle and poultry feed, and the eggs and edible tissues. Measurements
diation has also been suggested as an option for removing mycotoxins conducted in Japan revealed that AFB1 was found in maize fed to dairy
from poultry feed (Murata et al., 2008; Ameer Sumbal et al., 2016). cattle and was subsequently found to be contaminating the milk
Moreover, organic acids, which are frequently used in food and feed (Sugiyama et al., 2008). Analysis of AFB1-contaminated poultry feed
preservation, such as citric acid and lactic acid, also help reduce the samples conducted in Brazil showed concentrations ranging from 1.2 to
concentration of mycotoxins in feed samples (Humer et al., 2016). The 17.5 μg/kg, higher than fumonisins or ZEA (Oliveira et al., 2006). In a
use of chemicals to block signaling pathways or oxidative stress can case study conducted on farms in South Africa, AFB1 was found to

2
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Fig. 1. Physiological properties regulated by major mycotoxins that exert toxicity in exposed cells derived from farm animals. Aflatoxin B1, ochratoxin A, fumonisin
B1, zearalenone, and deoxynivalenol are the most frequently found mycotoxins in the feed of farm animals. Cultured porcine and avian cells have been used to
characterize the cytotoxicity of major mycotoxins. Mycotoxins induce either increased apoptosis or decreased proliferation, or both in animal cells. There are various
physiological changes that underlie mycotoxin-induced cytotoxicity, including oxidative stress, autophagy, ER stress, and signaling pathways.

contaminate various feed materials and it was found in retail milk at OTA is converted into various metabolites by the activity of the CYP450
concentrations of 0.01–3.1 μg/L (Dutton et al., 2012). A survey of milk enzyme in the liver. Metabolites such as 4-hydroxyochratoxin A (4'-OH-
samples from farm animals in Italy found that AFM1 levels below 5.0 OTA), 5'-OH-OTA, 7'-OH-OTA, 9'-OH-OTA, and ochratoxin B (OTB) are
ng/L were observed in 92 % of raw milk samples, while AFM1 was observed in the liver microsomes and feces of farm animals such as pigs,
found in approximately 42 % of various dairy products (Santini et al., cattle, and chickens (Tao et al., 2018). Most metabolites of OTA are
2013). reported to be less toxic, suggesting that the regulation of OTA meta-
Laying hens with AFB1-contaminated diets show high concentra- bolism is important for mitigating toxicity in farm animals (Omar et al.,
tions of AFB1 residues in eggs and liver tissue (Trucksess et al., 1983). 1996). Analysis conducted on animal feed in Argentina showed that
Regional studies conducted in Japan showed that AFM1 was found in OTA was found to be more contained in poultry feed at concentrations
domestic milk samples at a mean of 0.009 μg/kg (Nakajima et al., of 25–30 ng/g than in the feed of other animals, including pigs and
2004). Monitoring conducted in Argentina detected AFM1 in 63.8 % of rabbits (Dalcero et al., 2002). An analysis conducted in Italy found that
raw bulk milk samples from farm cooling tanks, suggesting that it could all feed samples from pig farms were contaminated with OTA, although
pose a threat to milk consumers (Alonso et al., 2010). In Iran, AFM1 the contamination levels were lower than the limits recommended by
levels were found to be 66.7 %, 31.0 %, and 35.4 % in bovine, ovine, the European Commission (Pozzo et al., 2010). Monitoring conducted
and caprine bulk milk samples, respectively (Rahimi and Ameri, 2012). in Pakistan found that the proportion and concentration of OTA con-
Some non-toxic feed additives have a high affinity with AFM1 and are tamination in poultry complete feeds were 38 % and 75 μg/kg, re-
used to prevent contamination in milk samples. Dietary administration spectively (Sherazi et al., 2015).
with clinoptilolite, a natural zeolite, significantly reduces the con- Many countries have confirmed the presence of OTA in porcine
centration of AFM1 in dairy cattle milk, suggesting that it mitigates the blood and edible tissues (Jorgensen, 1998; Morgan et al., 1986). OTA
toxicity caused by AFB1 and AFM1 in livestock (Katsoulos et al., 2016). was observed in the range of 0.05–13.4 ng/ml in 98 % of slaughtered
Hydrated sodium calcium aluminosilicate has also been reported to pig serum samples in Romania, whereas ZEA was found in only 17.3 %
reduce AFM1 levels in cattle milk (Harvey et al., 1991). of serum (Curtui et al., 2001). In addition, OTA levels were highest in
the kidney, followed by the liver and the muscles of slaughtered pigs.
Moreover, 1.0 % of porcine serum samples in Italy were positive for
2.2. Occurrence of ochratoxin A in feed OTA at a concentration of 0.03 to 0.8 ng/ml (Pozzo et al., 2010). In 82
% of swine plasma samples in Norway, the OTA concentrations were
Ochratoxin A (OTA) is a mycotoxin produced by several species of above 0.1 ng/ml (Langseth et al., 1993). Serum samples from slaugh-
the Aspergillus and Penicillium genera and is found in cereal grains and tered pigs in Serbia were found to have OTA concentrations up to 220.8
dairy products. OTA is naturally produced and observed in animal feed ng/ml, and up to 52.5 ng/g with the development of hemorrhages and
worldwide (Birzele et al., 2000; Scudamore and Patel, 2000; Hohler, necrosis in the kidneys (Milicevic et al., 2008).
1998), but its frequent presence in pork products threatens food safety
(Rodriguez et al., 2012; Jorgensen and Petersen, 2002). The FDA has
not set a concentration limit for OTA in foods, but the EU has estab- 2.3. Occurrence of fumonisin in feed
lished a maximum concentration limit of 5 ppb for cereal products and
10 ppb for dried vine fruits (Alshannaq and Yu, 2017; Malir et al., Fusarium species are the most frequently found toxin-producing
2016). OTA has carcinogenic properties when exposed to humans fungi, which are widely distributed in the northern climates of the
(Pfohl-Leszkowicz and Manderville, 2007). It has also been shown to Americas, Europe, and Asia. The most hazardous Fusarium mycotoxins
cause immunosuppressive effects and problems with urinary tract de- in farm animals are fumonisin, deoxynivalenol (DON), and ZEA. In
velopment in humans (Lea et al., 1989; Anon, 1991; Bozic et al., 1995). Korea, fumonisins, DON, and ZEA were found to be contaminated in

3
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

feeds at 93.33, 91.33, and 71.33 %, respectively (Kim et al., 2013). concentration limit of DON is 1000 ppb for processed foods, whereas
Fumonisin is a mycotoxin produced by Fusarium moniliforme and Fu- that of the EU is relatively lower, depending on the type of product (Lee
sarium proliferatum and is a naturally occurring feed contaminant. The and Ryu, 2017; Alshannaq and Yu, 2017). DON is converted into a de-
EU has set a maximum concentration limit of 4000 ppb for fumonisin in epoxidized form, which is less toxic to the intestinal microflora of farm
raw corn, which is relatively higher than that for other mycotoxins, animals (Danicke et al., 2010, 2004). When DON is administered to
although considerably lower limits have been set for processed foods DON, the amount of de-epoxy-DON relative to that of DON is higher in
(Lee and Ryu, 2017; Alshannaq and Yu, 2017). Little is known about the the distal part of the intestine (Danicke and Brezina, 2013). However,
endogenous metabolism of fumonisin in farm animals (Fodor et al., more than half of the ingested DON can be recovered from urine
2008). Veterinary drugs that are converted into low-toxic hydrolyzed (Danicke et al., 2004). In poultry, approximately 10 % of the total
forms using enzymes involved in bacterial fumonisin catabolism are concentration of DON and de-epoxy-DON in feces is observed as de-
used to remove fumonisin from pigs and poultry (Hartinger and Moll, epoxy-DON (Danicke and Brezina, 2013). Feed samples treated with
2011; Hahn et al., 2015). Fumonisin has three subtypes, B1 (FB1), B2 citric acid or lactic acid significantly reduced DON content (Humer
(FB2), and B3 (FB3), which vary in their chemical structure and also to et al., 2016). In randomly harvested wheat samples from Germany,
some extent in their degree of toxicity. Fumonisin is mainly found in the DON was observed at 77–93% of samples, levels higher than other
feed of poultry. In Slovakia, FB1 and FB2 have been observed in poultry mycotoxins including ZEA and nivalenol (NIV) (Muller et al., 2001). In
feed at concentrations from 43 to 798 μg/kg and from 26 to 362 μg/kg, Pakistanian poultry feed, DON and aflatoxins were copresent at con-
respectively (Labuda et al., 2005). Studies confirming mycotoxin con- centrations of 807 μg/kg and 18 μg/kg, respectively (Shar et al., 2014).
centrations performed in whole corn in California have shown that A survey of mycotoxins in pig feed in China revealed that the con-
fumonisin is positive in the highest proportion of samples, with an centration of DON was up to 4000 μg/kg, the highest concentration
average concentration of 1698 μg/kg, higher than other types of my- among mycotoxins (Ma et al., 2018).
cotoxins such as aflatoxins, OTA, ZEA, and DON (Krout-Greenberg
et al., 2013). Research conducted on poultry feed from Nigerian farms 3. Effects of mycotoxin exposure on farm animals
also shows that FB1 is the most frequently found mycotoxin, in 97 % of
samples, with a mean concentration of 1014 μg/kg (Akinmusire et al., 3.1. Effects of aflatoxins on farm animals
2019). Little is known about whether fumonisin can accumulate in the
eggs and edible tissue of poultry. In addition, the metabolites of fu- Studies on the toxicity of mycotoxins in farm animals are based on
monisin in poultry are unclear, requiring further research into the the concentrations reported in feed surveys, confirming changes in
toxicokinetics of fumonisin. various physiological properties after consuming contaminated feed or
dose-dependent treatment of cells with mycotoxins. Reproductive
2.4. Occurrence of zearalenone in feed toxicity occurs in poultry or pigs that are contaminated with aflatoxins.
In addition, aflatoxins contribute to the damage of the immune system
ZEA is an estrogenic mycotoxin produced by Fusarium species. ZEA of animals, making them vulnerable to disease (Oswald et al., 2005).
contains nonsteroidal resorcylic acid lactone, which mimics the activity Diets contaminated with aflatoxins at concentrations of up to 400 μg/kg
of estradiol-17β. The EU’s concentration limit for ZEA is 100 ppb in raw cause functional problems depending on the concentration in the liver
maize and 20 ppb in cereal products (Lee and Ryu, 2017; Alshannaq of wild turkeys (Quist et al., 2000). Moreover, AFB1-contaminated diets
and Yu, 2017). Exposure of the uterus during pregnancy to ZEA causes significantly inhibit the production of eggs in laying hens (Trucksess
reproductive organ disorders in the offspring of rodents, suggesting et al., 1983). RNA sequencing analysis performed on turkey livers re-
transgenerational effects (Nikaido et al., 2004). Plant and fungal me- vealed that AFB1 regulates transcripts involved in cancer, cell cycle
tabolites of ZEA, ZEA-14-O-β-glucoside, ZEA-16-O-β-glucoside, and control, and lipid regulation (Monson et al., 2014). Analysis of tran-
ZEA-14-sulfate are reported to be fully hydrolyzed in porcine gastro- scriptome changes in turkey spleens in response to AFB1 suggests that
intestinal tracks (Binder et al., 2017). α-zearalenol (ZOL) and β-ZOL are AFB1 affects the poultry immune system (Monson et al., 2015). More-
analogs of ZEA that are metabolized and produced in the liver and are over, AFB1 interferes with the maturation of porcine oocytes. In porcine
also found in pig and human excretory products. This metabolism is oocytes exposed to AFB1, DNA and histone methylation are affected in a
similar to the reaction during the synthesis and inactivation of steroid dose-dependent manner, suggesting epigenetic modifications (Liu et al.,
hormones since it is caused by the same dehydrogenase enzyme 2015). In addition, AFB1 increases apoptosis of porcine oocytes, which
(Malekinejad et al., 2006). Because α-ZOL has a higher binding capacity is mediated by increased oxidative stress and autophagy. Splenocytes
to cytoplasmic estrogen receptors than β-ZOL, ZEA is more reactive in are the major target cells of AFB1 in pigs. AFB1 inhibits the production
species where conversion to α-ZOL is preferred (Katzenellenbogen of interleukin (IL)-2 when exposed to porcine splenocytes, leading to
et al., 1979). In a study conducted in China, the maximum concentra- immunotoxicity. The toxicity of AFB1 to porcine splenocytes occurred
tion of ZEA in pig feed was approximately 729 μg/kg (Ma et al., 2018). in a dose-dependent manner. Moreover, AFB1 decreases the level of
A survey conducted in Vietnam revealed that the average concentration reduced glutathione (GSH) and increases lipid peroxidation in porcine
of ZEA in complete feeds for pigs was 101.4 μg/kg (Thieu et al., 2008). splenocytes, which is accompanied by increased phosphorylation of
With oral administration of 10 mg/kg ZEA in pigs, the uptake rate is ERK1/2 (Hao et al., 2015). The effectiveness of some compounds has
80–85% (Biehl et al., 1993). In porcine follicular fluids, ZEA and its been reported to reduce the toxicity of AFB1. They are generally not
metabolite, α-ZOL, are observed at concentrations up to 54.8 pg/ml and toxic on their own and their targeting mechanisms are clear and
26.4 pg/ml, respectively (Sambuu et al., 2011). Analytical studies therefore have potential as feed supplements. In porcine splenocytes,
conducted on feeding supplies and urine from farm animals in Croatia immunotoxicity by AFB1 is alleviated by selenium supplementation,
showed that ZEA and its metabolite, α-ZOL, peaked at concentrations of which improves the antioxidant capacity (Hao et al., 2016). The AFB1-
577 ng/g and 241.1 ng/ml, respectively (Vulic et al., 2012). induced degradation of porcine oocytes is effectively prevented by the
inhibition of oxidative stress caused by melatonin application (Cheng
2.5. Occurrence of deoxynivalenol in feed et al., 2019). Bush sophora root polysaccharide, a traditional herb, also
mitigates AFB1-induced toxicity in chicken liver cells by increasing
DON, which also has an estrogenic function, is another mycotoxin antioxidant activity (Gan et al., 2018a). Additionally, AFB1 induces
produced by a species of Fusarium (Diekman and Green, 1992). DON endoplasmic reticulum (ER) stress-mediated cell death in bovine
belongs to the trichothecene group and is chemically stable, so it is not mammary epithelial cells, which is accompanied by DNA fragmentation
easily removed during food or feed processing. The FDA’s maximum and mitochondrial dysfunction (Park et al., 2019). AFB2 increases

4
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Fig. 2. Physiological functions of afla-


toxin B1, aflatoxin B2, and ochratoxin
A in animal cells. Aflatoxins and
ochratoxin A are mycotoxins produced
primarily by fungi of the Aspergillus
genera. In animal cells, these myco-
toxins share a similar mechanism of
action, including an increase in the
activity of MAP kinases such as ERK1/2
and P38, and they promote oxidative
stress. Transcriptomic analysis suggests
that these mycotoxins can modulate
various properties such as tumor-
igenicity, cell cycle control, and lipid
regulation. These mycotoxins also in-
hibit cell proliferation through mi-
tochondrial disruption, induction of ER
stress, and inhibition of PI3K/AKT sig-
naling pathways. N-acetyl-L-cysteine
and selenomethionine alleviate myco-
toxin-induced toxicity. The effects of
aflatoxin B1, aflatoxin B2, and ochra-
toxin A result in nephrotoxicity, hepa-
totoxicity, and reproductive disorders
in farm animals.

apoptosis and autophagy by inhibiting the activation of the PI3K/AKT/ porcine alveolar macrophages (Xu et al., 2017), causing im-
mTOR signaling pathway in broiler hepatocytes (Chen et al., 2016). munotoxicity through an increase in Toll-like receptor 4 (TLR4)-medi-
AFB2 toxicity is dependent on the production of reactive oxygen species ated inflammatory signaling pathway proteins and elevated in-
(ROS) and the release of cytochrome c in hepatocytes. These studies tracellular ROS production. In vivo studies using feed containing OTA
suggest that aflatoxins can exert toxicity by regulating various physio- also revealed that OTA activates the MAPK signaling pathway and in-
logical properties, such as oxidative stress, ER stress, and signaling duces histopathological modifications by promoting ER stress and au-
pathway regulation, in farm animals. tophagy in the kidneys and spleens of pigs (Gan et al., 2017b). OTA also
promotes mitochondrial ROS-mediated apoptosis in porcine intestinal
3.2. Effects of ochratoxin A on farm animals epithelial cells, which is confirmed by the presence of mitochondria
that target ROS (Wang et al., 2017). In addition, the toxicity of OTA in
OTA is generally more toxic than other ochratoxins because of its porcine intestinal epithelial cells is mediated by increased intracellular
late metabolism (Gautier et al., 2001). OTA-contaminated feed ad- Ca2+ ion activity and myosin light chain kinase, suggesting the de-
versely affects the growth of pigs (Malagutti et al., 2005; Lippold et al., struction of tight junctions (Wang et al., 2018). Recently, various
1992) and has a physiological effect similar to aflatoxin B1 in animal methods have been studied to mitigate the toxicity of OTA. Porcine
cells (Fig. 2). The cytotoxicity of OTA is mostly exerted in a dose-de- circovirus type 2 infection in pigs has an alleviating effect on OTA-
pendent manner, altering various intracellular processes, including induced growth restriction and nephrotoxicity (Gan et al., 2018b).
oxidative stress, signaling pathways, and inflammatory responses. OTA Moreover, selenomethionine and NAC alleviate the toxicity of OTA and
increases chromosomal aberrations and sister chromatid exchanges in AFB1 in porcine alveolar macrophages by regulating the activity of ERK
bovine lymphocytes, causing apoptotic cell death (Lioi et al., 2004). (Hou et al., 2018). In pigs, urinary alkalinization promotes the renal
OTA induces toxicity in piglet kidneys and livers by reducing anti- excretion of OTA, thereby alleviating its toxicity (Blank and Wolffram,
oxidant parameters and causing oxidative stress, with histopathological 2004). In this context, dietary sodium bicarbonate supplementation in
alterations (Zhang et al., 2016). OTA also induces the phosphorylation pigs reduces the systemic availability of OTA (Blank and Wolffram,
of P38 and ERK1/2 in porcine splenocytes, leading to nephrotoxicity 2005). The toxicity of OTA in chicken liver cells has been confirmed to
and immunotoxicity, respectively (Gan et al., 2017a). Moreover, OTA be mitigated by various compounds, such as ammonium glycyrrhizin, L-
induces cytotoxicity by stimulating oxidative stress responses in porcine arginine, silymarin, and glucurolactone (Yu et al., 2018). Identifying a
kidney cells, as evidenced by the reduced toxicity seen with the pre- clear target mechanism for feed additives to mitigate the toxicity of
treatment of N-acetyl-L-cysteine (NAC), a ROS scavenger (Klaric et al., OTA will help prevent economic losses from OTA contamination in li-
2008). OTA also induces the phosphorylation of P38 and ERK1/2 in vestock farms.

5
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

3.3. Effects of fumonisins on farm animals microtubule dynamics. When pig cumulus-oocyte complexes are ex-
posed to DON there is inhibition of cumulus expansion and spindle
In farm animals, fumonisin B1 (FB1) is reported to cause diseases malformations (Schoevers et al., 2010). DON also inhibits barrier
such as equine leukoencephalomalacia and porcine pulmonary edema function in the porcine intestinal epithelium by regulating the expres-
(Marasas, 1995). Moreover, when exposed to pigs, FB1 is associated sion of tight junction proteins (Pinton et al., 2010). DON-mediated in-
with the development of disorders such as pulmonary edema and liver testinal barrier destruction is prevented with TLR2 ligand pre-treatment
failure (Haschek et al., 2001). In vitro studies reported that FB1 ex- by mediating activation of the PI3K/AKT signaling pathway (Gu et al.,
posure to porcine intestinal epithelial cells inhibits cell proliferation 2016). Broilers that consume DON-contaminated feed lose body weight
and barrier function (Bouhet et al., 2004). FB1 also reduces viability and liver weight (Lucke et al., 2017). DON-contaminated feed at a
and promotes lactate dehydrogenase release in porcine renal epithelial concentration of 10 mg/kg reduces feed intake and feed efficiency in
cells, suggesting induction of nephrotoxicity (He et al., 2002). FB1 is broilers (Ghareeb et al., 2012). DON also interferes with biochemical
also reported to affect the porcine immune system by inhibiting lym- indicators, suggesting impaired protein and lipid metabolism in
phocyte proliferation and controlling the Th1/Th2 cytokine balance chickens, along with altered immune responses, such as changes in the
(Harvey et al., 1995; Taranu et al., 2005). Another study reported de- number of lymphocytes and antibodies (Yunus et al., 2012a). Small
creased expression of IL-8 in the gut of pigs following the oral admin- intestinal morphology that is associated with macronutrient retention is
istration of 0.5 mg/kg FB1, although other cytokines were unaffected also altered in broilers (Yunus et al., 2012b). Diets containing DON
(Bouhet et al., 2006). FB1 causes pig intestinal disorders by increasing cause a significant reduction in the length of duodenal and jejunal villi
the level of free sphingolipid bases and reducing the number of glyco- of the chick, and this is accompanied by lipid peroxidation (Awad et al.,
lipids in the plasma membrane (Loiseau et al., 2007). Moreover, FB1 2006, 2014). Dietary DON decreases TNF-α levels in the plasma and IL-
causes a reduction in feed intake, egg production, and body weight in 1β, TGFBR1, and IFN-γ mRNA levels in the jejunum of broiler chicks,
poultry, while also causing disorders such as hepatic necrosis and suggesting the immunomodulatory function of DON in poultry
thymic cortical atrophy (Ledoux et al., 1992; Prathapkumar et al., (Ghareeb et al., 2013). DON-contaminated diets alter the humoral im-
1997). FB1-contaminated chicken feed, at a dose of 200 mg/kg, has mune response to viral vaccines in broiler chicks, a finding that was
immunosuppressive effects and also causes mild hepatic necrosis (Li validated by serum antibody levels (Ghareeb et al., 2016). Moreover,
et al., 1999; Ledoux et al., 2003). Moreover, when FB1 contaminates DON alters lipid profiles, such as increasing serum cholesterol and tri-
broiler diets, it impairs immune parameters by modulating the ex- glycerides in broilers. Feeds contaminated with up to 10 mg/kg of DON
pression of cytokines, including IL-1β and IL-2 in macrophages (Cheng interfere with mineral homeostasis in the bones of chickens (Keci et al.,
et al., 2006). It is reported that toxicity by sphingolipid metabolism, 2019). The adverse effects of DON in chicken are largely mitigated by
which is altered by FB1 in duodenum and jejunum in turkeys and pigs, microbial feed additives (Ghareeb et al., 2012, 2013), which are also
is detoxified by carboxylesterase (Masching et al., 2016). able to mitigate DNA damage in lymphocytes induced by DON in
broilers (Awad et al., 2014).
3.4. Effects of zearalenone on farm animals
3.6. Interaction effect of mycotoxins on farm animals
ZEA is reported to cause diseases related to infertility in pigs. Pigs
exposed to ZEA have vaginal and rectal prolapses, abortions, and lower Farm animals are generally exposed to two or more types of my-
pregnancy rates (Etienne and Dourmad, 1994). Diets containing ex- cotoxins simultaneously, rather than a single mycotoxin (Klaric et al.,
cessive concentrations of ZEA cause hyperestrogenism symptoms in 2009). Therefore, studies of the toxicity of mycotoxins in animals re-
pigs (Kuiper-Goodman et al., 1987). Moreover, the addition of α-ZOL or quire an understanding of the effects of multiple mycotoxins. Different
β-ZOL to porcine granulosa cell cultures inhibits the synthesis of pro- combinations of mycotoxins have a synergistic effect on environmental
gesterone, which is promoted by FSH or forskolin (Tiemann et al., toxicity, while some combinations cause antagonistic effects. Several
2003). ZEA induces caspase-3- and caspase-9-mediated cell death in review papers have described the interaction effects caused by different
porcine granulosa cells (Zhu et al., 2012). In addition, ZEA reduces cell types of mycotoxins (Alassane-Kpembi et al., 2017; Feijo Correa et al.,
proliferation by decreasing the mitochondrial membrane potential and 2018). Exposure to more than one mycotoxin has been demonstrated to
reducing ROS production in porcine granulosa cells. Exposure to ZEA have an interaction effect on hepatotoxicity, gastrointestinal toxicity,
during the prepubertal period causes a deficiency of mature oocytes in and reproductive toxicity in human and rodent cells (Sun et al., 2014;
adulthood. ZEA contained in feeds at doses up to 1 mg/kg decreases Alassane-Kpembi et al., 2013; Li et al., 2014). Studies also revealed that
follicle integrity and increases estrogen receptor expression when ex- combinations of mycotoxins have synergistic or antagonistic effects on
posed to lactating pigs (Schoevers et al., 2012). The increased oxidative cells derived from farm animals. The combination of AFB1, FB1, ZEA,
stress caused by ZEA in porcine granulosa cells is alleviated by cur- and DON in porcine kidney 15 cells (PK-15) causes either synergistic or
cumin, an Asian traditional herb derived from Curcuma longa (Qin et al., antagonistic effects on ROS production and apoptosis (Lei et al., 2013).
2015). ZEA also induces the formation of abnormal chromosomes and The combined treatment of OTA, FB1, and beauvericin also exerts sy-
reduces the mitotic index in bovine lymphocytes (Lioi et al., 2004). nergistic effects on the apoptotic index in PK-15 cells (Klaric et al.,
Bovine oocytes exposed to ZEA are arrested in metaphase 1 and im- 2008). The combination of OTA and FB1 has a synergistic effect on
mature but fertilization is not affected (Takagi et al., 2008). Little is reducing cell viability in porcine lymphocytes (Mwanza et al., 2009).
known about the detailed physiological mechanisms of ZEA toxicity in ZEA and DON are observed together in cereal grains and animal feed
farm animals. Therefore, further research on the toxicokinetics and supplies. Therefore, the physiological effects of their combined pre-
regulatory mechanisms of ZEA is needed to develop effective methods sence have been studied in animal cells (Fig. 3). Diets contaminated
to alleviate the toxicity of ZEA. with ZEA and DON inhibit the proliferative capacity of splenocytes and
cause histopathological modifications associated with spleen hemosi-
3.5. Effects of deoxynivalenol on farm animals derosis in gilts (Tiemann et al., 2006a). Feed contaminated with ZEA
and DON increase the percentage of degenerated meiotic chromatin in
Reports indicate that DON-contaminated pig feed causes feed re- oocytes recovered from the ovaries of gilts and interfere with normal
jection and vomiting (Diekman and Green, 1992). Feeds containing oocyte maturation (Alm et al., 2006). Pregnant sows fed a diet con-
DON at 600 μg/kg lead to an increase in immunoglobulin A (IgA) levels taining 15 % ZEA and DON suffer from splenic disorders, such as he-
in the serum of piglets (Drochner et al., 2004). Evidence suggests that mosiderosis (Tiemann et al., 2008). Diets contaminated with ZEA and
DON causes immaturity of porcine oocytes by interference with DON also cause histopathological disorders in the porcine liver without

6
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Fig. 3. Physiological functions of zearalenone and deoxynivalenol in animal cells. Zearalenone and deoxynivalenol are mycotoxins produced by the Fusarium species.
Zearalenone is metabolized in the liver to form zearalenol, which promotes ROS generation and induces mitochondria-dependent cell death. Deoxynivalenol aug-
ments the effects of zearalenol by inhibiting the PI3K/AKT signaling pathway. In intestinal cells, deoxynivalenol induces epithelial barrier destruction by inhibiting
tight junction proteins. Curcumin and TLR2 ligands alleviate the toxicity of zearalenone and deoxynivalenol, respectively. Exposure to both zearalenone and
deoxynivalenol causes ovarian, splenic, liver, and intestinal dysfunction in farm animals.

causing clinical symptoms (Tiemann et al., 2006b). In porcine granu- Declaration of Competing Interest
losa cells, DON alone promotes the production of P4 and E2, whereas
ZEA alone has not been shown to affect the production of steroid hor- The authors declare that they have no known competing financial
mones (Kolesarova et al., 2017). RNA sequencing was performed on interests or personal relationships that could have appeared to influ-
liver samples from piglets fed with ZEA and DON-contaminated feed, ence the work reported in this paper.
indicating an effect on the expression and network of immune-related
transcripts (Reddy et al., 2018). Acknowledgements

This article was supported by two funding sources as below: 1)


4. Conclusions Korea Health Technology R&D Project through the Korea Health
Industry Development Institute funded by the Ministry of Health &
Mycotoxin contamination of farm animal feed causes significant Welfare (grant number: HI17C0929), Republic of Korea; and 2)
economic loss. Mycotoxins cause fertility disorders and histopatholo- National Research Foundation of Korea (NRF) grant funded by the
gical changes in the kidneys and liver of pigs and poultry. Furthermore, Ministry of Science and ICT (MSIT) (No. 2018R1C1B6009048),
in vitro studies have shown that mycotoxins may regulate signaling Republic of Korea.
pathways in animal cells, promoting oxidative stress and mitochondria-
dependent cell death. Aflatoxin, ochratoxin A, fumonisin, ZEA, and References
DON, which are thought to be the most threatening to farm animals, are
naturally occurring in farm animal feed and frequently identified Abidin, Z., Khatoon, A., Arooj, N., Hussain, S., Ali, S., Manzoor, A.W., Saleemi, M.K.,
during monitoring. Moreover, mycotoxins, which accumulate in animal 2017. Estimation of ochratoxin A in poultry feed and its ingredients with special
reference to temperature conditions. Br. Poult. Sci. 58, 251–255.
tissues and are present in the blood and milk of livestock, can be a Abnet, C.C., 2007. Carcinogenic food contaminants. Cancer Invest. 25, 189–196.
threat to people who consume meat or dairy products. Recently, various Akinmusire, O.O., El-Yuguda, A.D., Musa, J.A., Oyedele, O.A., Sulyok, M., Somorin, Y.M.,
chemical and biological methods for mitigating mycotoxin toxicity have Ezekiel, C.N., Krska, R., 2019. Mycotoxins in poultry feed and feed ingredients in
Nigeria. Mycotoxin Res. 35, 149–155.
been proposed, and through continued research, we hope to reduce the Alassane-Kpembi, I., Kolf-Clauw, M., Gauthier, T., Abrami, R., Abiola, F.A., Oswald, I.P.,
economic loss incurred by farmers and also resolve the risks to human Puel, O., 2013. New insights into mycotoxin mixtures: the toxicity of low doses of
health. Type B trichothecenes on intestinal epithelial cells is synergistic. Toxicol. Appl.
Pharmacol. 272, 191–198.

7
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Alassane-Kpembi, I., Schatzmayr, G., Taranu, I., Marin, D., Puel, O., Oswald, I.P., 2017. Di Stefano, V., Pitonzo, R., Cicero, N., D’Oca, M.C., 2014. Mycotoxin contamination of
Mycotoxins co-contamination: methodological aspects and biological relevance of animal feedingstuff: detoxification by gamma-irradiation and reduction of aflatoxins
combined toxicity studies. Crit. Rev. Food Sci. Nutr. 57, 3489–3507. and ochratoxin A concentrations. Food Addit. Contam. Part A Chem. Anal. Control
Alm, H., Brussow, K.P., Torner, H., Vanselow, J., Tomek, W., Danicke, S., Tiemann, U., Expo. Risk Assess. 31, 2034–2039.
2006. Influence of Fusarium-toxin contaminated feed on initial quality and meiotic Diekman, M.A., Green, M.L., 1992. Mycotoxins and reproduction in domestic livestock. J.
competence of gilt oocytes. Reprod. Toxicol. 22, 44–50. Anim. Sci. 70, 1615–1627.
Alonso, V.A., Monge, M.P., Larriestra, A., Dalcero, A.M., Cavaglieri, L.R., Chiacchiera, Dohnal, V., Wu, Q., Kuca, K., 2014. Metabolism of aflatoxins: key enzymes and inter-
S.M., 2010. Naturally occurring aflatoxin M1 in raw bulk milk from farm cooling individual as well as interspecies differences. Arch. Toxicol. 88, 1635–1644.
tanks in Argentina. Food Addit. Contam. A 27, 373–379. Drochner, W., Schollenberger, M., Piepho, H.P., Gotz, S., Lauber, U., Tafaj, M., Klobasa,
Alshannaq, A., Yu, J.H., 2017. Occurrence, toxicity, and analysis of major mycotoxins in F., Weiler, U., Claus, R., Steffl, M., 2004. Serum IgA-promoting effects induced by
food. Int. J. Environ. Res. Public Health 14. feed loads containing isolated deoxynivalenol (DON) in growing piglets. J. Toxicol.
Ameer Sumbal, G., Hussain Shar, Z., Hussain Sherazi, S.T., Sirajuddin, Nizamani, S.M., Environ. Health A 67, 1051–1067.
Mahesar, S.A., 2016. Decontamination of poultry feed from ochratoxin A by UV and Dutton, M.F., Mwanza, M., de Kock, S., Khilosia, L.D., 2012. Mycotoxins in South African
sunlight radiations. J. Sci. Food Agric. 96, 2668–2673. foods: a case study on aflatoxin M1 in milk. Mycotoxin Res. 28, 17–23.
Anon, 1991. Mycotoxins, Endemic Nephropathy and Urinary Tract Tumours. IARC Sci Elliott, C.T., Connolly, L., Kolawole, O., 2019. Potential adverse effects on animal health
Publ, pp. 1–336. and performance caused by the addition of mineral adsorbents to feeds to reduce
Anukul, N., Vangnai, K., Mahakarnchanakul, W., 2013. Significance of regulation limits in mycotoxin exposure. Mycotoxin Res.
mycotoxin contamination in Asia and risk management programs at the national Etienne, M., Dourmad, J.Y., 1994. Effects of zearalenone or glucosinolates in the diet on
level. J. Food Drug Anal. 21, 227–241. reproduction in sows – a review. Livest Prod Sci 40, 99–113.
Awad, W.A., Bohm, J., Razzazi-Fazeli, E., Ghareeb, K., Zentek, J., 2006. Effect of addition Feijo Correa, J.A., Orso, P.B., Bordin, K., Hara, R.V., Luciano, F.B., 2018. Toxicological
of a probiotic microorganism to broiler diets contaminated with deoxynivalenol on effects of fumonisin B1 in combination with other Fusarium toxins. Food Chem.
performance and histological alterations of intestinal villi of broiler chickens. Poultry Toxicol. 121, 483–494.
Sci 85, 974–979. Fodor, J., Balogh, K., Weber, M., Mezes, M., Kametler, L., Posa, R., Mamet, R., Bauer, J.,
Awad, W.A., Ghareeb, K., Dadak, A., Hess, M., Bohm, J., 2014. Single and combined Horn, P., Kovacs, F., Kovacs, M., 2008. Absorption, distribution and elimination of
effects of deoxynivalenol mycotoxin and a microbial feed additive on lymphocyte fumonisin B(1) metabolites in weaned piglets. Food Addit. Contam. A 25, 88–96.
DNA damage and oxidative stress in broiler chickens. PLoS One 9, e88028. Galvano, F., Pietri, A., Bertuzzi, T., Fusconi, G., Galvano, M., Piva, A., Piva, G., 1996.
Biehl, M.L., Prelusky, D.B., Koritz, G.D., Hartin, K.E., Buck, W.B., Trenholm, H.L., 1993. Reduction of carryover of aflatoxin from cow feed to milk by addition of activated
Biliary excretion and enterohepatic cycling of zearalenone in immature pigs. Toxicol. carbons. J. Food Prot. 59, 551–554.
Appl. Pharmacol. 121, 152–159. Gan, F., Zhou, Y.J., Hou, L.L., Qian, G., Chen, X.X., Huang, K.H., 2017a. Ochratoxin A
Binder, S.B., Schwartz-Zimmermann, H.E., Varga, E., Bichl, G., Michlmayr, H., Adam, G., induces nephrotoxicity and immunotoxicity through different MAPK signaling
Berthiller, F., 2017. Metabolism of Zearalenone and its major modified forms in pigs. pathways in PK15 cells and porcine primary splenocytes. Chemosphere 182,
Toxins (Basel) 9. 630–637.
Birzele, B., Prange, A., Kramer, J., 2000. Deoxynivalenol and ochratoxin A in German Gan, F., Hou, L., Zhou, Y., Liu, Y., Huang, D., Chen, X., Huang, K., 2017b. Effects of
wheat and changes of level in relation to storage parameters. Food Addit. Contam. ochratoxin A on ER stress, MAPK signaling pathway and autophagy of kidney and
17, 1027–1035. spleen in pigs. Environ. Toxicol. 32, 2277–2286.
Blank, R., Wolffram, S., 2004. Alkalinization of urinary pH accelerates renal excretion of Gan, F., Yang, Y., Chen, Y., Che, C., Pan, C., Huang, K., 2018a. Bush sophora root poly-
ochratoxin A in pigs. J. Nutr. 134, 2355–2358. saccharide could help prevent aflatoxin B1-induced hepatotoxicity in the primary
Blank, R., Wolffram, S., 2005. Effect of dietary sodium bicarbonate supplementation on chicken hepatocytes. Toxicon 150, 180–187.
the toxicokinetics of ochratoxin A in pigs. Mycotoxin Res. 21, 147–149. Gan, F., Zhou, Y., Qian, G., Huang, D., Hou, L., Liu, D., Chen, X., Wang, T., Jiang, P., Lei,
Bouhet, S., Hourcade, E., Loiseau, N., Fikry, A., Martinez, S., Roselli, M., Galtier, P., X., Huang, K., 2018b. PCV2 infection aggravates ochratoxin A-induced ne-
Mengheri, E., Oswald, I.P., 2004. The mycotoxin fumonisin B1 alters the proliferation phrotoxicity via autophagy involving p38 signaling pathway in vivo and in vitro.
and the barrier function of porcine intestinal epithelial cells. Toxicol. Sci. 77, Environ. Pollut. 238, 656–662.
165–171. Gautier, J., Richoz, J., Welti, D.H., Markovic, J., Gremaud, E., Guengerich, F.P., Turesky,
Bouhet, S., Le Dorze, E., Peres, S., Fairbrother, J.M., Oswald, I.P., 2006. Mycotoxin fu- R.J., 2001. Metabolism of ochratoxin A: absence of formation of genotoxic deriva-
monisin B1 selectively down-regulates the basal IL-8 expression in pig intestine: in tives by human and rat enzymes. Chem. Res. Toxicol. 14, 34–45.
vivo and in vitro studies. Food Chem. Toxicol. 44, 1768–1773. Gesing, A., Jagiela, J., Lewinski, A., 2006. Effects of melatonin on the process of apoptosis
Bozic, Z., Duancic, V., Belicza, M., Kraus, O., Skljarov, I., 1995. Balkan endemic ne- in rat thyroid follicular cells. Neuro Endocrinol. Lett. 27, 81–84.
phropathy: still a mysterious disease. Eur. J. Epidemiol. 11, 235–238. Ghareeb, K., Awad, W.A., Bohm, J., 2012. Ameliorative effect of a microbial feed additive
Breitholtz-Emanuelsson, A., Olsen, M., Oskarsson, A., Palminger, I., Hult, K., 1993. on infectious bronchitis virus antibody titer and stress index in broiler chicks fed
Ochratoxin A in cow’s milk and in human milk with corresponding human blood deoxynivalenol. Poult. Sci. 91, 800–807.
samples. J. AOAC Int. 76, 842–846. Ghareeb, K., Awad, W.A., Soodoi, C., Sasgary, S., Strasser, A., Bohm, J., 2013. Effects of
Chen, B., Li, D., Li, M., Li, S., Peng, K., Shi, X., Zhou, L., Zhang, P., Xu, Z., Yin, H., Wang, feed contaminant deoxynivalenol on plasma cytokines and mRNA expression of im-
Y., Zhao, X., Zhu, Q., 2016. Induction of mitochondria-mediated apoptosis and PI3K/ mune genes in the intestine of broiler chickens. PLoS One 8, e71492.
Akt/ mTOR-mediated autophagy by aflatoxin B2 in hepatocytes of broilers. Ghareeb, K., Awad, W.A., Zebeli, Q., Bohm, J., 2016. Deoxynivalenol in chicken feed
Oncotarget 7, 84989–84998. alters the vaccinal immune response and clinical biochemical serum parameters but
Cheng, Y.H., Ding, S.T., Chang, M.H., 2006. Effect of fumonisins on macrophage immune not the intestinal and carcass characteristics. J. Anim. Physiol. Anim. Nutr. (Berl)
functions and gene expression of cytokines in broilers. Arch. Anim. Nutr. 60, 100, 53–60.
267–276. Gong, Y., Hounsa, A., Egal, S., Turner, P.C., Sutcliffe, A.E., Hall, A.J., Cardwell, K., Wild,
Cheng, L., Qin, Y., Hu, X., Ren, L., Zhang, C., Wang, X., Wang, W., Zhang, Z., Hao, J., Guo, C.P., 2004. Postweaning exposure to aflatoxin results in impaired child growth: a
M., Wu, Z., Tian, J., An, L., 2019. Melatonin protects in vitro matured porcine oocytes longitudinal study in Benin, West Africa. Environ. Health Perspect. 112, 1334–1338.
from toxicity of Aflatoxin B1. J. Pineal Res. 66, e12543. Gregory, J.F., Manley, D.B., 1982. High-performance liquid-chromatographic quantita-
Curtui, V.G., Gareis, M., Usleber, E., Martlbauer, E., 2001. Survey of Romanian slaugh- tion of aflatoxin metabolites in animal-tissues. J. Assoc. 65, 869–875.
tered pigs for the occurrence of mycotoxins ochratoxins A and B, and zearalenone. Gu, M.J., Song, S.K., Lee, I.K., Ko, S., Han, S.E., Bae, S., Ji, S.Y., Park, B.C., Song, K.D., Lee,
Food Addit. Contam. 18, 730–738. H.K., Han, S.H., Yun, C.H., 2016. Barrier protection via Toll-like receptor 2 signaling
D’Mello, J.P.F., Placinta, C.M., Macdonald, A.M.C., 1999. Fusarium mycotoxins: a review in porcine intestinal epithelial cells damaged by deoxynivalnol. Vet. Res. 47, 25.
of global implications for animal health, welfare and productivity. Anim. Feed Sci. Hahn, I., Nagl, V., Schwartz-Zimmermann, H.E., Varga, E., Schwarz, C., Slavik, V.,
Technol. 80, 183–205. Reisinger, N., Malachova, A., Cirlini, M., Generotti, S., Dall’Asta, C., Krska, R., Moll,
Dalcero, A., Magnoli, C., Hallak, C., Chiacchiera, S.M., Palacio, G., Rosa, C.A., 2002. W.D., Berthiller, F., 2015. Effects of orally administered fumonisin B-1 (FB1), par-
Detection of ochratoxin A in animal feeds and capacity to produce this mycotoxin by tially hydrolysed FB1, hydrolysed FB1 and N-(1-deoxy-D-fructos-1-yl) FB1 on the
Aspergillus section Nigri in Argentina. Food Addit. Contam. 19, 1065–1072. sphingolipid metabolism in rats. Food Chem. Toxicol. 76, 11–18.
Danicke, S., Brezina, U., 2013. Kinetics and metabolism of the Fusarium toxin deox- Hao, S., Pan, S., Hu, J., Qian, G., Gan, F., Huang, K., 2015. Aflatoxin B1 suppressed T-cell
ynivalenol in farm animals: consequences for diagnosis of exposure and intoxication response to Anti-pig-CD3 monoclonal antibody stimulation in primary porcine sple-
and carry over. Food Chem. Toxicol. 60, 58–75. nocytes: a role for the extracellular regulated protein kinase (ERK1/2) MAPK sig-
Danicke, S., Winkler, J., 2015. Invited review: diagnosis of zearalenone (ZEN) exposure of naling pathway. J. Agric. Food Chem. 63, 6094–6101.
farm animals and transfer of its residues into edible tissues (carry over). Food Chem. Hao, S., Hu, J., Song, S., Huang, D., Xu, H., Qian, G., Gan, F., Huang, K., 2016. Selenium
Toxicol. 84, 225–249. alleviates aflatoxin B(1)-induced immune toxicity through improving glutathione
Danicke, S., Valenta, H., Doll, S., 2004. On the toxicokinetics and the metabolism of peroxidase 1 and selenoprotein S expression in primary porcine splenocytes. J. Agric.
deoxynivalenol (DON) in the pig. Arch. Anim. Nutr. 58, 169–180. Food Chem. 64, 1385–1393.
Danicke, S., Hegewald, A.K., Kahlert, S., Kluess, J., Rothkotter, H.J., Breves, G., Doll, S., Hartinger, D., Moll, W.D., 2011. Fumonisin elimination and prospects for detoxification
2010. Studies on the toxicity of deoxynivalenol (DON), sodium metabisulfite, DON- by enzymatic transformation. World Mycotoxin J. 4, 271–283.
sulfonate (DONS) and de-epoxy-DON for porcine peripheral blood mononuclear cells Harvey, R.B., Kubena, L.F., Phillips, T.D., Corrier, D.E., Elissalde, M.H., Huff, W.E., 1991.
and the Intestinal Porcine Epithelial Cell lines IPEC-1 and IPEC-J2, and on effects of Diminution of aflatoxin toxicity to growing lambs by dietary supplementation with
DON and DONS on piglets. Food Chem. Toxicol. 48, 2154–2162. hydrated sodium calcium aluminosilicate. Am. J. Vet. Res. 52, 152–156.
Deng, J., Zhao, L., Zhang, N.Y., Karrow, N.A., Krumm, C.S., Qi, D.S., Sun, L.H., 2018. Harvey, R.B., Edrington, T.S., Kubena, L.F., Elissalde, M.H., Rottinghaus, G.E., 1995.
Aflatoxin B1 metabolism: regulation by phase I and II metabolizing enzymes and Influence of aflatoxin and fumonisin B1-containing culture material on growing
chemoprotective agents. Mutat. Res. 778, 79–89. barrows. Am. J. Vet. Res. 56, 1668–1672.

8
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Haschek, W.M., Gumprecht, L.A., Smith, G., Tumbleson, M.E., Constable, P.D., 2001. ameliorators of aflatoxicosis in weanling growing swine. J. Anim. Sci. 71, 171–178.
Fumonisin toxicosis in swine: an overview of porcine pulmonary edema and current Lioi, M.B., Santoro, A., Barbieri, R., Salzano, S., Ursini, M.V., 2004. Ochratoxin A and
perspectives. Environ. Health Perspect. 109 (Suppl. 2), 251–257. zearalenone: a comparative study on genotoxic effects and cell death induced in
He, Q., Riley, R.T., Sharma, R.P., 2002. Pharmacological antagonism of fumonisin B1 bovine lymphocytes. Mutat. Res. 557, 19–27.
cytotoxicity in porcine renal epithelial cells (LLC-PK1): a model for reducing fumo- Lippold, C.C., Stothers, S.C., Frohlich, A.A., Boila, R.J., Marquardt, R.R., 1992. Effects of
nisin-induced nephrotoxicity in vivo. Pharmacol. Toxicol. 90, 268–277. periodic feeding of diets containing Ochratoxin-a on the performance and clinical-
Herzallah, S., Alshawabkeh, K., Al Fataftah, A., 2008. Aflatoxin decontamination of ar- chemistry of pigs from 15 to 50 kg body-weight. Can. J. Anim. Sci. 72, 135–146.
tificially contaminated feeds by sunlight, gamma-radiation, and microwave heating. Liu, J., Wang, Q.C., Han, J., Xiong, B., Sun, S.C., 2015. Aflatoxin B1 is toxic to porcine
J. Appl. Poultry Res. 17, 515–521. oocyte maturation. Mutagenesis 30, 527–535.
Hohler, D., 1998. Ochratoxin A in food and feed: occurrence, legislation and mode of Loiseau, N., Debrauwer, L., Sambou, T., Bouhet, S., Miller, J.D., Martin, P.G., Viadere,
action. Z. Ernahrungswiss 37, 2–12. J.L., Pinton, P., Puel, O., Pineau, T., Tulliez, J., Galtier, P., Oswald, I.P., 2007.
Hooshmand, H., Klopfenstein, C.F., 1995. Effects of gamma-irradiation on mycotoxin Fumonisin B1 exposure and its selective effect on porcine jejunal segment: sphin-
disappearance and amino-acid contents of corn, wheat, and soybeans with different golipids, glycolipids and trans-epithelial passage disturbance. Biochem. Pharmacol.
moisture contents. Plant Foods Hum. Nutr. 47, 227–238. 74, 144–152.
Hou, L., Zhou, X., Gan, F., Liu, Z., Zhou, Y., Qian, G., Huang, K., 2018. Combination of Lucke, A., Doupovec, B., Paulsen, P., Zebeli, Q., Bohm, J., 2017. Effects of low to mod-
selenomethionine and N-acetylcysteine alleviates the joint toxicities of aflatoxin B1 erate levels of deoxynivalenol on feed and water intake, weight gain, and slaugh-
and ochratoxin a by ERK MAPK signal pathway in porcine alveolar macrophages. J. tering traits of broiler chickens. Mycotoxin Res. 33, 261–271.
Agric. Food Chem. 66, 5913–5923. Ma, R., Zhang, L., Liu, M., Su, Y.T., Xie, W.M., Zhang, N.Y., Dai, J.F., Wang, Y., Rajput,
Humer, E., Lucke, A., Harder, H., Metzler-Zebeli, B.U., Bohm, J., Zebeli, Q., 2016. Effects S.A., Qi, D.S., Karrow, N.A., Sun, L.H., 2018. Individual and combined occurrence of
of citric and lactic acid on the reduction of deoxynivalenol and its derivatives in mycotoxins in feed ingredients and complete feeds in China. Toxins (Basel) 10.
feeds. Toxins (Basel) 8. Malagutti, L., Zannotti, M., Scampini, A., Sciaraffia, F., 2005. Effects of Ochratoxin A on
Hussein, H.S., Brasel, J.M., 2001. Toxicity, metabolism, and impact of mycotoxins on heavy pig production. Anim. Res. 54, 179–184.
humans and animals. Toxicology 167, 101–134. Malekinejad, H., Maas-Bakker, R., Fink-Gremmels, J., 2006. Species differences in the
Jalili, M., Jinap, S., Noranizan, M.A., 2012. Aflatoxins and ochratoxin a reduction in black hepatic biotransformation of zearalenone. Vet. J. 172, 96–102.
and white pepper by gamma radiation. Radiat. Phys. Chem. 81, 1786–1788. Malir, F., Ostry, V., Pfohl-Leszkowicz, A., Malir, J., Toman, J., 2016. Ochratoxin A: 50
Jorgensen, K., 1998. Survey of pork, poultry, coffee, beer and pulses for ochratoxin A. years of research. Toxins 8.
Food Addit. Contam. 15, 550–554. Marasas, W.F.O., 1995. Fumonisins – History, Worldwide Occurrence and Impact. Abstr.
Jorgensen, K., Petersen, A., 2002. Content of ochratoxin A in paired kidney and meat Pap. Am. Chem. S 209 56-Agfd.
samples from healthy Danish slaughter pigs. Food Addit. Contam. 19, 562–567. Masching, S., Naehrer, K., Schwartz-Zimmermann, H.E., Sarandan, M., Schaumberger, S.,
Katsoulos, P.D., Karatzia, M.A., Boscos, C., Wolf, P., Karatzias, H., 2016. In-field eva- Dohnal, I., Nagl, V., Schatzmayr, D., 2016. Gastrointestinal degradation of fumonisin
luation of clinoptilolite feeding efficacy on the reduction of milk aflatoxin M1 con- B(1) by carboxylesterase FumD prevents fumonisin induced alteration of sphingolipid
centration in dairy cattle. J. Anim. Sci. Technol. 58, 24. metabolism in Turkey and swine. Toxins (Basel) 8.
Katzenellenbogen, B.S., Katzenellenbogen, J.A., Mordecai, D., 1979. Zearalenones: Meki, A.R., Abdel-Ghaffar, S.K., El-Gibaly, I., 2001. Aflatoxin B1 induces apoptosis in rat
characterization of the estrogenic potencies and receptor interactions of a series of liver: protective effect of melatonin. Neuro Endocrinol. Lett. 22, 417–426.
fungal beta-resorcylic acid lactones. Endocrinology 105, 33–40. Milicevic, D., Juric, V., Stefanovic, S., Jovanovic, M., Jankovic, S., 2008. Survey of
Keci, M., Lucke, A., Paulsen, P., Zebeli, Q., Bohm, J., Metzler-Zebeli, B.U., 2019. slaughtered pigs for occurrence of ochratoxin A and porcine nephropathy in Serbia.
Deoxynivalenol in the diet impairs bone mineralization in broiler chickens. Toxins Int. J. Mol. Sci. 9, 2169–2183.
(Basel) 11. Monge, M.P., Dalcero, A.M., Magnoli, C.E., Chiacchiera, S.M., 2013. Natural co-occur-
Kendra, D.F., Dyer, R.B., 2007. Opportunities for biotechnology and policy regarding rence of fungi and mycotoxins in poultry feeds from Entre Rios, Argentina. Food
mycotoxin issues in international trade. Int. J. Food Microbiol. 119, 147–151. Addit. Contam. B 6, 168–174.
Kim, D.H., Lee, I.H., Do, W.H., Nam, W.S., Li, H., Jang, H.S., Lee, C., 2013. Incidence and Monson, M.S., Settlage, R.E., McMahon, K.W., Mendoza, K.M., Rawal, S., El-Nezami, H.S.,
levels of deoxynivalenol, fumonisins and zearalenone contaminants in animal feeds Coulombe, R.A., Reed, K.M., 2014. Response of the hepatic transcriptome to aflatoxin
used in Korea in 2012. Toxins (Basel) 6, 20–32. B1 in domestic turkey (Meleagris gallopavo). PLoS One 9, e100930.
Klaric, M.S., Rumora, L., Ljubanovic, D., Pepeljnjak, S., 2008. Cytotoxicity and apoptosis Monson, M.S., Settlage, R.E., Mendoza, K.M., Rawal, S., El-Nezami, H.S., Coulombe, R.A.,
induced by fumonisin B(1), beauvericin and ochratoxin A in porcine kidney PK15 Reed, K.M., 2015. Modulation of the spleen transcriptome in domestic turkey
cells: effects of individual and combined treatment. Arch. Toxicol. 82, 247–255. (Meleagris gallopavo) in response to aflatoxin B1 and probiotics. Immunogenetics 67,
Klaric, M.S., Cvetnic, Z., Pepeljnjak, S., Kosalec, I., 2009. Co-occurrence of aflatoxins, 163–178.
ochratoxin A, fumonisins, and zearalenone in cereals and feed, determined by com- Morgan, M.R.A., Mcnerney, R., Chan, H.W.S., Anderson, P.H., 1986. Ochratoxin-a in Pig-
petitive direct enzyme-linked immunosorbent assay and thin-layer chromatography. Kidney determined by enzyme-linked-immunosorbent-assay (Elisa). J. Sci. Food
Arh. Hig. Rada Toksikol. 60, 427–434. Agric. 37, 475–480.
Kolesarova, A., Medvedova, M., Halenar, M., Sirotkin, A.V., Bulla, J., 2017. The influence Muller, H.M., Reimann, J., Schumacher, U., Schwadorf, K., 2001. Further survey of the
of deoxynivalenol and zearalenone on steroid hormone production by porcine occurrence of Fusarium toxins in wheat grown in southwest Germany. Arch. Anim.
ovarian granulosa cells in vitro. J. Environ. Sci. Health B 52, 823–832. Nutr. 54, 173–182.
Krout-Greenberg, N.D., Puschner, B., Davidson, M.G., DePeters, E.J., 2013. Preliminary Murata, H., Mitsumatsu, M., Shimada, N., 2008. Reduction of feed-contaminating my-
study to assess mycotoxin concentrations in whole corn in the California feed supply. cotoxins by ultraviolet irradiation: an in vitro study. Food Addit. Contam. Part A
J. Dairy Sci. 96, 2705–2712. Chem. Anal. Control Expo. Risk Assess. 25, 1107–1110.
Kuiper-Goodman, T., Scott, P.M., Watanabe, H., 1987. Risk assessment of the mycotoxin Mwanza, M., Kametler, L., Bonai, A., Rajli, V., Kovacs, M., Dutton, M.F., 2009. The cy-
zearalenone. Regul. Toxicol. Pharmacol. 7, 253–306. totoxic effect of fumonisin B1 and ochratoxin A on human and pig lymphocytes using
Labuda, R., Parich, A., Vekiru, E., Tancinova, D., 2005. Incidence of fumonisins, mon- the Methyl Thiazol Tetrazolium (MTT) assay. Mycotoxin Res. 25, 233–238.
iliformin and Fusarium species in poultry feed mixtures from Slovakia. Ann. Agric. Nakajima, M., Tabata, S., Akiyama, H., Itoh, Y., Tanaka, T., Sunagawa, H., Tyonan, T.,
Environ. Med. 12, 81–86. Yoshizawa, T., Kumagai, S., 2004. Occurrence of aflatoxin M1 in domestic milk in
Langseth, W., Nymoen, U., Bergsjo, B., 1993. Ochratoxin A in plasma of Norwegian swine Japan during the winter season. Food Addit. Contam. 21, 472–478.
determined by an HPLC column-switching method. Nat. Toxins 1, 216–221. Nikaido, Y., Yoshizawa, K., Danbara, N., Tsujita-Kyutoku, M., Yuri, T., Uehara, N.,
Lea, T., Steien, K., Stormer, F.C., 1989. Mechanism of ochratoxin A-induced im- Tsubura, A., 2004. Effects of maternal xenoestrogen exposure on development of the
munosuppression. Mycopathologia 107, 153–159. reproductive tract and mammary gland in female CD-1 mouse offspring. Reprod.
Ledoux, D.R., Brown, T.P., Weibking, T.S., Rottinghaus, G.E., 1992. Fumonisin toxicity in Toxicol. 18, 803–811.
broiler chicks. J. Vet. Diagn. Invest. 4, 330–333. Oliveira, G.R., Ribeiro, J.M., Fraga, M.E., Cavaglieri, L.R., Direito, G.M., Keller, K.M.,
Ledoux, D.R., Broomhead, J.N., Bermudez, A.J., Rottinghaus, G.E., 2003. Individual and Dalcero, A.M., Rosa, C.A., 2006. Mycobiota in poultry feeds and natural occurrence of
combined effects of the Fusarium mycotoxins fumonisin B1 and moniliformin in aflatoxins, fumonisins and zearalenone in the Rio de Janeiro State, Brazil.
broiler chicks. Avian Dis. 47, 1368–1375. Mycopathologia 162, 355–362.
Lee, H.J., Ryu, D., 2017. Worldwide occurrence of mycotoxins in cereals and cereal-de- Omar, R.F., Gelboin, H.V., Rahimtula, A.D., 1996. Effect of cytochrome P450 induction
rived food products: public health perspectives of their co-occurrence. J. Agric. Food on the metabolism and toxicity of ochratoxin A. Biochem. Pharmacol. 51, 207–216.
Chem. 65, 7034–7051. Oswald, I.P., Marin, D.E., Bouhet, S., Pinton, P., Taranu, I., Accensi, F., 2005.
Lei, M., Zhang, N., Qi, D., 2013. In vitro investigation of individual and combined cy- Immunotoxicological risk of mycotoxins for domestic animals. Food Addit. Contam.
totoxic effects of aflatoxin B1 and other selected mycotoxins on the cell line porcine 22, 354–360.
kidney 15. Exp. Toxicol. Pathol. 65, 1149–1157. Park, W., Park, M.Y., Song, G., Lim, W., 2019. Exposure to aflatoxin B1 attenuates cell
Li, Y.C., Ledoux, D.R., Bermudez, A.J., Fritsche, K.L., Rottinghaus, G.E., 1999. Effects of viability and induces endoplasmic reticulum-mediated cell death in a bovine mam-
fumonisin B1 on selected immune responses in broiler chicks. Poult. Sci. 78, mary epithelial cell line (MAC-T). Toxicol. In Vitro 61, 104591.
1275–1282. Peters, L.P., Teel, R.W., 2003. Effect of high sucrose diet on liver enzyme content and
Li, Y., Zhang, B., He, X., Cheng, W.H., Xu, W., Luo, Y., Liang, R., Luo, H., Huang, K., 2014. activity and aflatoxin B1-induced mutagenesis. In Vivo 17, 205–210.
Analysis of individual and combined effects of ochratoxin A and zearalenone on Pfohl-Leszkowicz, A., Manderville, R.A., 2007. Ochratoxin A: an overview on toxicity and
HepG2 and KK-1 cells with mathematical models. Toxins (Basel) 6, 1177–1192. carcinogenicity in animals and humans. Mol. Nutr. Food Res. 51, 61–99.
Li, H., Malyar, R.M., Zhai, N., Wang, H., Liu, K., Liu, D., Pan, C., Gan, F., Huang, K., Miao, Pinton, P., Braicu, C., Nougayrede, J.P., Laffitte, J., Taranu, I., Oswald, I.P., 2010.
J., Chen, X., 2019. Zinc supplementation alleviates OTA-induced oxidative stress and Deoxynivalenol impairs porcine intestinal barrier function and decreases the protein
apoptosis in MDCK cells by up-regulating metallothioneins. Life Sci. 234, 116735. expression of Claudin-4 through a mitogen-acitivated protein kinase-dependent me-
Lindemann, M.D., Blodgett, D.J., Kornegay, E.T., Schurig, G.G., 1993. Potential chanism. J. Nutr. 140, 1956–1962.

9
C. Yang, et al. Journal of Hazardous Materials 389 (2020) 122087

Placinta, C.M., D’Mello, J.P.F., Macdonald, A.M.C., 1999. A review of worldwide con- contamination in commercial pasteurised milk from different areas in Thailand. Food
tamination of cereal grains and animal feed with Fusarium mycotoxins. Anim. Feed Addit. Contam. Part B Surveill. 5, 145–149.
Sci. Technol. 78, 21–37. Takagi, M., Mukai, S., Kuriyagawa, T., Takagaki, K., Uno, S., Kokushi, E., Otoi, T.,
Polak, M., Gajecki, M., Kulik, T., Luczynski, M.K., Obremski, K., Gora, M., Gajecka, M., Budiyanto, A., Shirasuna, K., Miyamoto, A., Kawamura, O., Okamoto, K., Deguchi, E.,
Jakimiuk, E., Zielonka, L., 2009. The evaluation of the efficacy of sodium carbonate 2008. Detection of zearalenone and its metabolites in naturally contaminated folli-
as zearalenone destructor in feeding stuffs. Pol. J. Vet. Sci. 12, 103–111. cular fluids by using LC/MS/MS and in vitro effects of zearalenone on oocyte ma-
Pozzo, L., Cavallarin, L., Nucera, D., Antoniazzi, S., Schiavone, A., 2010. A survey of turation in cattle. Reprod. Toxicol. 26, 164–169.
ochratoxin A contamination in feeds and sera from organic and standard swine farms Tao, Y., Xie, S., Xu, F., Liu, A., Wang, Y., Chen, D., Pan, Y., Huang, L., Peng, D., Wang, X.,
in northwest Italy. J. Sci. Food Agric. 90, 1467–1472. Yuan, Z., 2018. Ochratoxin A: toxicity, oxidative stress and metabolism. Food Chem.
Prathapkumar, S.H., Rao, V.S., Paramkishan, R.J., Bhat, R.V., 1997. Disease outbreak in Toxicol. 112, 320–331.
laying hens arising from the consumption of fumonisin-contaminated food. Br. Poult. Taranu, I., Marin, D.E., Bouhet, S., Pascale, F., Bailly, J.D., Miller, J.D., Pinton, P.,
Sci. 38, 475–479. Oswald, I.P., 2005. Mycotoxin fumonisin B1 alters the cytokine profile and decreases
Qin, X., Cao, M., Lai, F., Yang, F., Ge, W., Zhang, X., Cheng, S., Sun, X., Qin, G., Shen, W., the vaccinal antibody titer in pigs. Toxicol. Sci. 84, 301–307.
Li, L., 2015. Oxidative stress induced by zearalenone in porcine granulosa cells and Thieu, N.Q., Ogle, B., Pettersson, H., 2008. Screening of Aflatoxins and Zearalenone in
its rescue by curcumin in vitro. PLoS One 10, e0127551. feedstuffs and complete feeds for pigs in Southern Vietnam. Trop. Anim. Health Prod.
Quist, C.F., Bounous, D.I., Kilburn, J.V., Nettles, V.F., Wyatt, R.D., 2000. The effect of 40, 77–83.
dietary aflatoxin on wild turkey poults. J. Wildl. Dis. 36, 436–444. Tiemann, U., Tomek, W., Schneider, F., Vanselow, J., 2003. Effects of the mycotoxins
Rahimi, E., Ameri, M., 2012. A survey of aflatoxin M1 contamination in bulk milk samples alpha- and beta-zearalenol on regulation of progesterone synthesis in cultured
from dairy bovine, ovine, and caprine herds in Iran. Bull. Environ. Contam. Toxicol. granulosa cells from porcine ovaries. Reprod. Toxicol. 17, 673–681.
89, 158–160. Tiemann, U., Brussow, K.P., Jonas, L., Pohland, R., Schneider, F., Danicke, S., 2006a.
Raju, M.V., Devegowda, G., 2000. Influence of esterified-glucomannan on performance Effects of diets with cereal grains contaminated by graded levels of two Fusarium
and organ morphology, serum biochemistry and haematology in broilers exposed to toxins on selected immunological and histological measurements in the spleen of
individual and combined mycotoxicosis (aflatoxin, ochratoxin and T-2 toxin). Br. gilts. J. Anim. Sci. 84, 236–245.
Poult. Sci. 41, 640–650. Tiemann, U., Brussow, K.P., Kuchenmeister, U., Jonas, L., Kohlschein, P., Pohland, R.,
Reddy, K.E., Jeong, J.Y., Lee, Y., Lee, H.J., Kim, M.S., Kim, D.W., Jung, H.J., Choe, C., Oh, Danicke, S., 2006b. Influence of diets with cereal grains contaminated by graded
Y.K., Lee, S.D., 2018. Deoxynivalenol- and zearalenone-contaminated feeds alter gene levels of two Fusarium toxins on selected enzymatic and histological parameters of
expression profiles in the livers of piglets. Asian-Aust. J. Anim. Sci. 31, 595–606. liver in gilts. Food Chem. Toxicol. 44, 1228–1235.
Rodrigues, I., Naehrer, K., 2012. A three-year survey on the worldwide occurrence of Tiemann, U., Brussow, K.P., Dannenberger, D., Jonas, L., Pohland, R., Jager, K., Danicke,
mycotoxins in feedstuffs and feed. Toxins (Basel) 4, 663–675. S., Hagemann, E., 2008. The effect of feeding a diet naturally contaminated with
Rodriguez, A., Rodriguez, M., Martin, A., Delgado, J., Cordoba, J.J., 2012. Presence of deoxynivalenol (DON) and zearalenone (ZON) on the spleen and liver of sow and
ochratoxin A on the surface of dry-cured Iberian ham after initial fungal growth in the fetus from day 35 to 70 of gestation. Toxicol. Lett. 179, 113–117.
drying stage. Meat Sci. 92, 728–734. Trucksess, M.W., Stoloff, L., Young, K., Wyatt, R.D., Miller, B.L., 1983. Aflatoxicol and
Rosa, C.A., Miazzo, R., Magnoli, C., Salvano, M., Chiacchiera, S.M., Ferrero, S., Saenz, M., Aflatoxin-B1 and Aflatoxin-M1 in eggs and tissues of laying hens consuming afla-
Carvalho, E.C., Dalcero, A., 2001. Evaluation of the efficacy of bentonite from the toxin-contaminated feed. Poultry Sci. 62, 2176–2182.
south of Argentina to ameliorate the toxic effects of aflatoxin in broilers. Poult. Sci. Vulic, A., Pleadin, J., Persi, N., Mitak, M., 2012. Analysis of naturally occurring zear-
80, 139–144. alenone in feeding stuffs and urine of farm animals in Croatia. J. Immunoassay
Sambuu, R., Takagi, M., Shiga, S., Uno, S., Kokushi, E., Namula, Z., Otoi, T., Miyamoto, Immunochem. 33, 369–376.
A., Deguchi, E., Fink-Gremmels, J., 2011. Detection of zearalenone and its metabo- Wang, H., Chen, Y., Zhai, N., Chen, X., Gan, F., Li, H., Huang, K., 2017. Ochratoxin A-
lites in naturally contaminated porcine follicular fluid by using liquid chromato- induced apoptosis of IPEC-J2 cells through ROS-mediated mitochondrial perme-
graphy-tandem mass spectrometry. J. Reprod. Dev. 57, 303–306. ability transition pore opening pathway. J. Agric. Food Chem. 65, 10630–10637.
Santini, A., Raiola, A., Ferrantelli, V., Giangrosso, G., Macaluso, A., Bognanno, M., Wang, H., Zhai, N., Chen, Y., Fu, C., Huang, K., 2018. OTA induces intestinal epithelial
Galvano, F., Ritieni, A., 2013. Aflatoxin M-1 in raw, UHT milk and dairy products in barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca
Sicily (Italy). Food Addit. Contam. B 6, 181–186. (2+)-mediated MLCK activation. Environ. Pollut. 242, 106–112.
Sarathchandra, G., Muralimanohar, B., 2013. Occurrence of mycotoxins in livestock feeds Wolzak, A., Pearson, A.M., Coleman, T.H., Pestka, J.J., Gray, J.I., 1985. Aflatoxin de-
and feed stuffs of Tamil Nadu. J. Environ. Biol. 34, 825–830. position and clearance in the eggs of laying hens. Food Chem. Toxicol. 23,
Sarma, U.P., Bhetaria, P.J., Devi, P., Varma, A., 2017. Aflatoxins: implications on Health. 1057–1061.
Indian J. Clin. Biochem. 32, 124–133. Xu, H., Hao, S., Gan, F., Wang, H., Xu, J., Liu, D., Huang, K., 2017. In vitro immune
Schoevers, E.J., Fink-Gremmels, J., Colenbrander, B., Roelen, B.A.J., 2010. Porcine oo- toxicity of ochratoxin A in porcine alveolar macrophages: a role for the ROS-relative
cytes are most vulnerable to the mycotoxin deoxynivalenol during formation of the TLR4/MyD88 signaling pathway. Chem. Biol. Interact. 272, 107–116.
meiotic spindle. Theriogenology 74, 968–978. Yu, Z., Wu, F., Tian, J., Guo, X., An, R., 2018. Protective effects of compound ammonium
Schoevers, E.J., Santos, R.R., Colenbrander, B., Fink-Gremmels, J., Roelen, B.A., 2012. glycyrrhizin, Larginine, silymarin and glucurolactone against liver damage induced
Transgenerational toxicity of Zearalenone in pigs. Reprod. Toxicol. 34, 110–119. by ochratoxin A in primary chicken hepatocytes. Mol. Med. Rep. 18, 2551–2560.
Scott, P.M., Kanhere, S.R., Lau, B.P., Lewis, D.A., Hayward, S., Ryan, J.J., Kuiper- Yunus, A.W., Ghareeb, K., Twaruzek, M., Grajewski, J., Bohm, J., 2012a. Deoxynivalenol
Goodman, T., 1998. Survey of Canadian human blood plasma for ochratoxin A. Food as a contaminant of broiler feed: effects on bird performance and response to
Addit. Contam. 15, 555–562. common vaccines. Poultry Sci. 91, 844–851.
Scudamore, K.A., Patel, S., 2000. Survey for aflatoxins, ochratoxin A, zearalenone and Yunus, A.W., Blajet-Kosicka, A., Kosicki, R., Khan, M.Z., Rehman, H., Bohm, J., 2012b.
fumonisins in maize imported into the United Kingdom. Food Addit. Contam. 17, Deoxynivalenol as a contaminant of broiler feed: intestinal development, absorptive
407–416. functionality, and metabolism of the mycotoxin. Poultry Sci. 91, 852–861.
Shar, Z.H., Sumbal, G.A., Sherazi, S.T., Bhanger, M.I., Nizamani, S.M., 2014. Natural co- Zhai, N., Wang, H., Chen, Y., Li, H., Viktor, K., Huang, K., Chen, X., 2018. Taurine at-
occurrence of aflatoxins and deoxynivalenol in poultry feed in Pakistan. Food Addit. tenuates OTA-promoted PCV2 replication through blocking ROS-dependent autop-
Contam. Part B Surveill. 7, 162–167. hagy via inhibiting AMPK/mTOR signaling pathway. Chem. Biol. Interact. 296,
Sherazi, S.T., Shar, Z.H., Sumbal, G.A., Tan, E.T., Bhanger, M.I., Kara, H., Nizamani, S.M., 220–228.
2015. Occurrence of ochratoxin A in poultry feeds and feed ingredients from Zhang, Z., Gan, F., Xue, H., Liu, Y., Huang, D., Khan, A.Z., Chen, X., Huang, K., 2016.
Pakistan. Mycotoxin Res. 31, 1–7. Nephropathy and hepatopathy in weaned piglets provoked by natural ochratoxin A
Sugiyama, K., Hiraoka, H., Sugita-Konishi, Y., 2008. Aflatoxin M1 contamination in raw and involved mechanisms. Exp. Toxicol. Pathol. 68, 205–213.
bulk milk and the presence of aflatoxin B1 in corn supplied to dairy cattle in Japan. Zhu, L., Yuan, H., Guo, C., Lu, Y., Deng, S., Yang, Y., Wei, Q., Wen, L., He, Z., 2012.
Shokuhin Eiseigaku Zasshi 49, 352–355. Zearalenone induces apoptosis and necrosis in porcine granulosa cells via a caspase-
Sun, L.H., Lei, M.Y., Zhang, N.Y., Zhao, L., Krumm, C.S., Qi, D.S., 2014. Hepatotoxic 3- and caspase-9-dependent mitochondrial signaling pathway. J. Cell. Physiol. 227,
effects of mycotoxin combinations in mice. Food Chem. Toxicol. 74, 289–293. 1814–1820.
Suriyasathaporn, W., Nakprasert, W., 2012. Seasonal patterns of aflatoxin M1

10

You might also like