You are on page 1of 28

Environmental Science and Pollution Research

https://doi.org/10.1007/s11356-020-08991-y

ENVIRONMENTAL POLLUTANTS AND THE RISK OF NEUROLOGICAL DISORDERS

Ochratoxin A–induced genotoxic and epigenetic mechanisms lead


to Alzheimer disease: its modulation with strategies
Kamal Niaz 1 & Syed Zahid Ali Shah 2 & Fazlullah Khan 3,4 & Mohammed Bule 5

Received: 9 December 2019 / Accepted: 22 April 2020


# Springer-Verlag GmbH Germany, part of Springer Nature 2020

Abstract
Ochratoxin A (OTA) is a naturally occurring mycotoxin mostly found in food items including grains and coffee beans. It induces
DNA single-strand breaks and has been considered to be carcinogenic. It is recognized as a serious threat to reproductive health
both in males and females. OTA is highly nephrotoxic and carcinogenic, and its potency changes evidently between species and
sexes. There is a close association between OTA, mutagenicity, carcinogenicity, and genotoxicity, but the underlying mecha-
nisms are not clear. Reports regarding genotoxic effects in relation to OTA which leads to the induction of DNA adduct
formation, protein synthesis inhibition, perturbation of cellular energy production, initiation of oxidative stress, induction of
apoptosis, influences on mitosis, induction of cell cycle arrest, and interference with cytokine pathways. All these mechanisms
are associated with nephrotoxicity, hepatotoxicity, teratotoxicity, immunological toxicity, and neurotoxicity. OTA administration
activates various mechanisms such as p38 MAPK, JNKs, and ERKs dysfunctions, BDNF disruption, TH overexpression,
caspase-3 and 9 activation, and ERK-1/2 phosphorylation which ultimately lead to Alzheimer disease (AD) progression. The
current review will focus on OTA in terms of recent discoveries in the field of molecular biology. The main aim is to investigate
the underlying mechanisms of OTA in regard to genotoxicity and epigenetic modulations that lead to AD. Also, we will highlight
the strategies for the purpose of attenuating the hazards posed by OTA exposure.

Keywords Ochratoxin A . Genotoxicity . Epigenetics . DNA methylation . Histone modifications . Nephrotoxicity .


Therapeutics . Alzheimer disease

Introduction products that are stored for longer periods of time such as
cereals, coffee, cocoa, nuts, and peanuts (Zhu et al. 2017).
Presently ochratoxin A (OTA) is a major threat to public OTA is a mycotoxin that specifically targets kidneys and is
health. Various species of fungi from the Penicillium, known for its ability to cross the placental barrier and expos-
Alternaria, and Aspergillus genera produce the OTA myco- ing a newly formed fetus to its harmful effects (Malir et al.
toxin (Zhu et al. 2017). OTA occurs in nutritional products of 2013a). OTA exposure is recognized as a major risk factor in
both vegetative and animal origin. In general, it affects reproductive health in males and females throughout the

Responsible Editor: Mohamed M. Abdel-Daim

* Kamal Niaz 2
Department of Pathology, Faculty of Veterinary Science, Cholistan
kamalniaz1989@gmail.com; kamalniaz1989@cuvas.edu.pk University of Veterinary and Animal Sciences, Bahawalpur 63100,
Pakistan
Syed Zahid Ali Shah 3
The Institute of Pharmaceutical Sciences (TIPS), School of
zahidvet@cuvas.edu.pk
Pharmacy, International Campus, Tehran University of Medical
Fazlullah Khan Sciences (IC-TUMS), Tehran 1417614411, Iran
f-khan@tums.ac.ir 4
Department of Toxicology and Pharmacology, Faculty of Pharmacy,
1
Tehran University of Medical Science, Tehran 1417614411, Iran
Department of Pharmacology and Toxicology, Faculty of
5
Bio-Sciences, Cholistan University of Veterinary and Animal Department of Pharmacy, College of Medicine and Health Sciences,
Sciences, Bahawalpur 63100, Pakistan Ambo University, Ambo, Oromia, Ethiopia
Environ Sci Pollut Res

world. Besides reproductive and renal damage, its association the oxygen that interacts with R257, although the carboxylic
with developmental malformations, neurotoxicity, hepatotox- acid interacts with the amino acids R218 and/or R222; the
icity, and immunological toxicity has been shown in the lab- customized albumin according to data robustly support the
oratory as well as in dairy farm animals and human beings interaction of the essential functions of R257 and R218 argi-
(Vettorazzi et al. 2013). It has also been observed that organ- nines (Perry et al. 2004). As a result of the deprotonation of
isms with a predisposed susceptibility may develop cancer R257 arginine, a stable ion pair is formed between the pheno-
due to OTA exposure depending on species, sex, and age of lic hydroxyl group of OTA and the human serum albumin
the organisms (Vettorazzi et al. 2013). Humans and animals (HSA).
that consume foods and feedstuffs contaminated with OTA Few other proteins besides albumin are also the targeted
adversely affect cells, tissues, organs, and fluids along with sites of OTA; those protein structures with a molecular weight
blood which is considered an essential indicator of OTA ex- of around 20 kDa have the potential to provide more affinity
posure. OTA contamination in laboratory and farm animals is to attract OTA rather than albumin. These proteins and their
best observed in blood samples due to the difficulty of the mechanism of action are not clearly known as albumin, but
body to excrete it (Malir et al. 2013a, 2013b). The mecha- they could play a major role in the pathogenesis free filtration
nisms responsible for the adverse effects of OTA are mostly through the glomeruli (Kőszegi and Poór 2016). The actions
still under discussion throughout the scientific community. A of phenylalanine-tRNA synthase can be inhibited by OTA, as
variety of toxic effects has been attributed to OTA including the fact is well known that the OTA is protein synthesis in-
DNA damage, perturbation of protein synthesis, cell cycle hibitor both in vitro and in vivo. It has been considered that
arrest, induction of programmed cell death, necrosis, chromo- according to biochemistry, the phenylalanine portion of OTA
somal degradation, and protein damage. All these perturba- plays an important role in the competition between phenylal-
tions are caused due to the reactive oxygen species (ROS)– anine and the toxin (Alshannaq and Yu 2017; Creppy et al.
induced oxidative stress (Costa et al. 2016; Marin-Kuan et al. 1984; Zanicgrubisic et al. 2000). Despite this, a recent study
2011; Zhu et al. 2017). In the year 1965, OTA was isolated by revealed that isocoumarin itself is responsible for such inter-
a chemical process and defined structurally for the first time in actions than the phenylalanine since the modifications on the
South Africa (Van Der Merwe et al. 1965). The name of OTA isocoumarin moiety have resulted in a considerable effect on
was assigned according to the standard naming system and the its action (Kőszegi and Poór 2016). Additional studies need to
Chemical Abstracts Service (CAS) guidelines with a number be conducted in order to emphasize the fact that the phenylal-
303-47-9. Structurally speaking, OTA is a dihydro- anine part is a major part contributing to mycotoxin toxicity
isocoumarin derivative linked to L-phenylalanine N-[(5- (Alshannaq and Yu 2017; Zanicgrubisic et al. 2000).
chloro-3, 4-dihydro-8-hydroxy-3-methyl-1-oxo-1-H-2- Moreover, OTA inhibits phenylalanine hydroxylase and also
benzopyran-7-yl) carbonyl]-(R) via an amide bond. acts as a false substrate of the catalytic reaction. The hydrox-
According to the IUPAC nomenclature, OTA is indicated as ylation of its phenylalanine moiety brings about tyrosine-
(N-[[(3R)-5-chloro-8-hydroxy-3-methyl-1-oxo-7- containing OTA that has been distinguished considerably dur-
isochromanyl] carbonyl]-3-phenylL-alanine). Alternatively, ing in vivo examinations. However, we must underscore the
OTA is sometimes known as -N-[(5-chloro-8-hydroxy-3- impact of OTA with respect to phenylalanine-tRNA synthase,
methyl-1-oxo-7-isochromanyl) carbonyl]-3-phenylalanine which is more toxic than phenylalanine hydroxylase. The cel-
along with the molecular formula C20H18O6ClN (Malir et al. lular production of ATP is influenced negatively by OTA.
2016). Protein synthesis is reduced due to the improper functioning
Before one can grasp the implications of OTA on the health of mitochondria (Kőszegi and Poór 2016). OTA plays a vital
of humans and animals, a basic understanding of the biochem- role in energy metabolism due to its binding with protein
ical mechanisms of action by which OTA works is necessary. moieties found in the building of mitochondrial membranes,
OTA binds to protein moieties in the plasma membrane of blocking and interfering with the phosphate and electron
cells (80–90%). It forms a strong and persistent attachment movement across the membrane.
that results in protein synthesis inhibition and cellular and Benzoquinone and its radicals play an important part in the
mitotic degradations as well as arrest and urinary tract carci- formation of DNA adduct as it is properly known that DNA
nogenicity (Kőszegi and Poór 2016; Malir et al. 2016). adduct is the causative agent of gene mutations and conse-
The target site for the action of OTA is albumin in animals quently results in tumorigenesis, particularly the cancer of
and human beings. In the active site of the target protein, urinary tract (Pfohl-Leszkowicz and Manderville 2007).
OTAs’ isocoumarin structure is mainly situated among amino DNA adducts are the byproducts of the covalent interaction
acids composed of amino acids A291, L238, I260, I264, I290, of OTA generated via radical and benzoquinone intermedi-
R257, and S287, whereas the phenyl group is surrounded by ates. OTAs’ metabolite named as OTA hydroquinone
K199, H242, Y211, L238, and W214 amino acids (Perry et al. (OTHQ) undergoes autoxidation to produce the quinone elec-
2004). The carbonyl and phenolic hydroxyl groups contain trophile OTA quinine (OTQ) that undergoes the reaction with
Environ Sci Pollut Res

DNA. Production of ROS is initially enhanced by the input of that protein synthesis inhibition results from RNA predomi-
OTQ or phenoxy and aryl radicals and its eventually respon- nant downregulation and protein expression. Differentially
sible for cytotoxicity (Pfohl-Leszkowicz and Manderville expressed genes which are directly related to protein inhibi-
2011). C-bound C8-dG adduct is produced by aryl free radical tion have been observed due to OTA treatments such as up-
species while benzoquinone and quinone electrophile forms regulation of prostaglandin F2 receptor negative regulator
benzetheno-type DNA adducts. (PTGFRN), the eukaryotic translation initiation factor 4E
The C-bound C8-OTA adducts play their role in cytotox- binding protein-1 (EIF4EBP1), and downregulation of the
icity at a lower rate. Reductive dehalogenation of OTA also elongation factor 1-alpha 1 (EEF1A1), which promotes the
shows in this manner (via cyclo-oxygenase alternately guanosine triphosphate (GTP)–dependent binding of
lipoxygenase). Those C5-Cl particles are essential to DNA aminoacyl-tRNA to the A-site of ribosomes during protein
adduction (genotoxicity), yet not for cytotoxicity (OTB biosynthesis (Fig. 1). Other related genes to protein synthesis
[Ochratoxin B] is cytotoxic yet all are not genotoxic); a few and proteolysis namely EIF2S1, EIF4E, EIF4G2, and
quinone subsidiaries have been isolated from blood and urine EEF1A1 as well as phosphatases, ligases, transferases, and
exposed to OTA. The carcinogenic effects while co-contact hydrolases are also observed to be differentially expressed
with fumonisin (FB), citrinin (CIT), or both are clear and and downregulated while involving the protein synthesis, pro-
understandable (Mantle et al. 2009). Although both carcino- teolysis, and post-translational modification explaining that
genic and genotoxic effects can be explained by the coopera- the metabolism of proteins is the utmost altered process by
tion of both factors as they are a major culprit for the induction OTA as shown in Fig. 1 (Vettorazzi et al. 2013).
of COX2 leading to genotoxic compounds (Malir et al. 2013a, Mainly produced by different fungi species, the toxin curbs
2013b; Ringot et al. 2006). The professional community the polyuridylic acid (poly-U)–dependent peptide synthesis
agreed with the carcinogenic effect of OTA, and they realized system leading to the accumulation of the MS-nucleotides
that OTA is one of the major mycotoxins that is a great threat ppGpp and pppGpp resulting in phenylalanine inhibition in
as a nephrotoxic, neurotoxic, reprotoxic, teratotoxic, and the aminoacylation reaction, which is strongly dependent on
embryotoxic (Huang and Chan 2016; Lühe et al. 2003; the Mg2+ concentration. This observation has concluded that
Malir et al. 2013a, 2013b; Paradells et al. 2015). It is a threat OTA stands out in non-specific protein synthesis inhibition
to the embryo/fetal development and all other aspects of the such as prohibiting the phenylalanine-tRNA synthase activity
development. by significantly contributing the isocoumarin structure as well
OTA is already an important topic for research and discus- as preventing the phenylalanine hydroxylase (Kőszegi and
sion amongst the scientific community; major milestones have Poór 2016). The toxin could also influence other protein tran-
been achieved in order to understand the molecular mecha- scription showing accurate effects inside the cells. For in-
nisms and epigenetics of OTA. The unique aspect of this stance, it has been reported that OTA induces specific degra-
review is to cover those studies which took place recently. dation of the mRNA that codes for gluconeogenesis key en-
The latest studies about OTA have been summarized in the zyme (phosphoenolpyruvate carboxykinase) resulting in de-
articles that are involved in the molecular mechanisms and creasing the degree of its synthesis. Moreover, upregulation of
epigenetic alterations that cause Alzheimer’s disease (AD). P21 in proximal tubule cells and in mouse skin and downreg-
The article focuses on the key aspects according to molecular ulation in hepatocarcinoma cells were recorded resulting in
and epigenetic alteration caused by OTA which ultimately blocking of the cyclin-dependent kinase activity and thereby
leads to AD that had not been reported previously in a single obstruction of the cell cycle and transcription regulation, DNA
article along with various strategies to lessen OTA toxicity. repair as well as cell motility (Dutto et al. 2015). Studies have
shown that deoxynivalenol (DON) and OTA have toxicolog-
ical effects on the intestinal epithelial protein synthesis and
Molecular mechanisms of OTA barrier integrity as their presence has been revealed in dog
foods, beer, and wines. In fact, the toxin affects Caco-2 cell
Inhibition of protein synthesis permeability by eliminating especial claudin isoforms out of
the tight junctions and the membrane microdomains (Van De
The major toxicities of OTA have been demonstrated in cell Walle et al. 2010; Arrúa et al. 2019; Songsermsakul et al.
culture models including bacteria, yeasts as well as mamma- 2007). Furthermore, in vitro and in vivo lipid peroxidation
lian models. The modulation of transcription factors of differ- improvements following permissible OTA exposure were no-
ent proteins resulting in the inhibition of their synthesis and ticed due to the expression of important cell or mitochondrial
leading to alterations in many specific cell functions. Protein membrane–related effects. These observations have been no-
synthesis repression brings about the blocking of RNA syn- ticed within in vivo mouse liver and cultured hepatoma cells
thesis and peptide elongation inhibition due to the competition and Madin-Darby canine kidney cells and more accentuated
with phenylalanine-tRNA synthetase (PheRS), considering within the kidney and spleen. For instance, lon peptidase 1
Environ Sci Pollut Res

Fig. 1 Mechanisms underlying OTA-induced genotoxicity

(Lonp1) has an essential function in protein metabolism and ATP emission was severely affected, which was noticed as the
modification. Assuming that OTA inhibits the synthesis and first deleterious effect of OTA, followed by morphological
the metabolism of protein and in order to investigate Lonp1 alteration of the organelle (Poór et al. 2014). Studies have
functions in OTA-induced renal cytotoxicity, an approach of demonstrated that this oxidative status of the cells has affected
iTRAQ-based proteomics combined with RNAi was used to respiratory control and oxidative phosphorylation of the iso-
analyze protein expression changes in HEK293 cell. Results lated mitochondria resulting in an alteration within the mem-
have demonstrated that OTA decreases Lonp1 within cells brane structure and the bioenergetics functions as well as ATP
and induces proteomic changes in Lonp1-deficient cells which depletion in isolated mitochondria from rat liver (Poór et al.
improved further knowledge in OTA cytotoxicity and indicat- 2014).
ed that OTA disturbs protein synthesis and modification Lonp1 is an ATP-dependent serine protease that facilitates
(Zhang et al. 2014). some transitory regulatory proteins in the mitochondrial ma-
trix, and it is also a mitochondrial DNA–binding protein
Inhibition of cellular energy production which is mandatory for maintaining mtDNA. By studying
the possible metabolic pathways of Lonp1, which is involved
Adenosine triphosphate (ATP) provides the required energy in OTA-induced renal cytotoxicity, Lonp1 expression in
for chemical reactions, locomotion, and cell division within HEK293 cells was downregulated via RNA interference. It
metabolism or for active transport of chemical species across has been shown that the OTA-subjected cells have expressed
biological membranes (Poór et al. 2014). It has been found Lonp1 deficiency and have demonstrated an undesirable ef-
that OTA represses the phosphate carrier proteins of the inner fect on the mitochondrial protein metabolism resulting in high
mitochondrial membrane resulting in the braking of the elec- oxidative damaged proteins and a significant impact on the
tron transit inside the mitochondria and preventing the mitochondrial functions (Zhang et al. 2014). Genes that are
succinate-supported electron-dependent activities. The mito- associated with the mitochondrial electron transport chain
chondrial respiration chain was found to be blocked and the were upregulated within in vitro HK-2 cells culture.
Environ Sci Pollut Res

Moreover, it has been found that at the mitochondrial and evidence was demonstrated through the repair enzyme
cytosolic levels, genes that are responsible for carbohydrate formamido-pyrimidine-DNA-glycosylase (Fpg) that was
metabolism function were suppressed. These are genes which shown to cause fragmentation of DNA at different oxidized
are mainly associated with glycolysis (phosphofructokinase guanine adducts allowing the detection of oxidized DNA ba-
(PFK) and phosphoglycerate kinase (PGK1)), tricarboxylic ses, namely 8-oxo-7, 8-dihydro-20-deoxyguanosine (dOG),
acid cycle (isocitrate dehydrogenases IDH3B and IDH3G), which is a biomarker for oxidative damage. Moreover, it has
and gluconeogenesis (phosphoenolpyruvate carboxykinase 2 been highlighted that the gene which encodes detoxification,
(PCK2) and C/EBP beta transcription factor) and have been cytoprotective, and antioxidant enzymes was also affected by
highlighted in HepG2 cells, suggesting that OTA significantly OTA exposure which has been found to be engaged in
impairs the cellular energy supply (Vettorazzi et al. 2013). understating the expression of genes regulated by nuclear
Other tests have indicated that exposure of human peripheral factor-erythroid 2 p45-related factor (Nrf2) resulting in the
blood lymphocytes to OTA has generated mitochondrial dys- reduction of the Nrf2 pathway (Pfohl-Leszkowicz and
function, which in turn triggered apoptosis while decreasing Manderville 2011). In the same context, it has been shown
the pro-survival protein Bcl-xL (Fig. 1). In fact, the toxin that OTA-exposed rats have expressed affection for GCLC,
incidences on mitochondrial transmembrane potential have GCLM, glutathione synthetase (GSS), UGT3B5, and multiple
been evaluated and results have explained that the toxin dam- GST isoforms genes and that OTA exposure decreases the
ages the mitochondria and induces disruption of the mitochon- GSTP and GCLC protein levels (Fig. 1). These findings have
drial membrane due to loss of transmembrane potential in H9 shown an mRNA degradation of Nrf2-dependent genes in rat
cell (Darif et al. 2016). liver and kidney as well as in human primary proximal tubule
cells proving that OTA behaves as an Nrf2 inhibitor
Induction of oxidative/nitrosative stress (Limonciel and Jennings 2014). Similarly, it has been reported
that OTA enhances malondialdehyde (MDA) development
Appearance of ROS within the biological system shows the which is a lipid peroxidation final product and a late stage of
rise of oxidative stress which can also trigger the reactive oxidative stress biomarker and cellular damage (Marin-Kuan
nitrogen species and cause nitrosative stress, also referred to et al. 2011). OTA supplementation to in vivo and in vitro rat
as the ROS/RNS pathway (Marin-Kuan et al. 2011). OTA- liver microsomes had increased levels of ethane exhaled and
exposed cells, either in vivo or in vitro, have demonstrated boosts enzyme NADPH-dependent enzyme via chemical
excessive expression of ROS resulting in high oxidative dam- ascorbate/FeS04-dependent pathway. Furthermore, it has
ages. It has been shown that the male Sprague-Dawley rats’ been reported that NADPH metabolizes OTA into two epi-
liver exposed to OTA has significantly decreased expression meric hydroxyl derivatives (4S)-4-0H-OA and (4R)-4-0H-
in the activity of glutathione S-transferase and in the levels of OA, as well as into an irreversible protein reactive species.
glutathione, as well as marked increases in the concentrations Thus, due to the availability of the NADPH-CYP450 reduc-
of thiobarbituric acid reactive substances which had led to tase OTAFe3+ complex, the Fe3+ could be easily reduced to
oxidant and antioxidant imbalanced status and have develop the OTA-Fe2+ complex which gives rise to radical
underlined that oxidative stress is OTA-dependent hepatic species causing DNA damage and lipid peroxidation. Lipid
toxicity (Palabiyik et al. 2012; Reverberi et al. 2012). peroxidation by oxygen uptake has also been indicated as a
Furthermore, it was demonstrated that the expression of in- remarkable stimulator of OTA in a system involving of phos-
ducible nitric oxide synthase (iNOs) and protein nitration are pholipid vesicles, the flavoprotein NADPH-cytochrome P450
marked by exposure to OTA that gives rise to ROS/RNS reductase, Fe3+, EDTA, and NADPH. It has also been shown
production in normal rat kidney cell line and in rat hepatocyte that OTA generates protein carbonyl productions which are
cultures (Fig. 1) (Marin-Kuan et al. 2011). In fact, OTA com- markers of protein oxidation. In fact, protein carbonyl concen-
prises an aryl-ring system that couples with direct redox cy- tration were measured in the plasma, kidney, and liver of
cling reactions that decrease ROS. Besides, this phenol- OTA-treated rats with the help of 2, 4 DNPH reagent
mediated oxidative stress mechanism was suggested for (Sorrenti et al. 2013). Significantly higher concentrations of
OTA as futile thiol pumping, and then it causes oxidation of protein carbonyl due to lipid peroxidation of the kidney and
thiols and depletion of antioxidants, as OTA reduces glutathi- liver cells have been noticed confirming that the mechanism
one reductase levels in mammalian cell lines (Marin-Kuan of OTA-mediated toxicity involves oxidative stress and this is
et al. 2011). These reactions resulted in decreased antioxidant not only by triggering lipid peroxidation but also by affecting
defenses and increased levels of DNA, lipid, and protein ox- proteins (Sorrenti et al. 2013). Likewise, it has been demon-
idation. Furthermore, it has been noticed that oxidative stress strated that the production of ROS namely the superoxide
associated with OTA understates cell defensive ability to an- anion (O2), hydroxyl radical (OH), and peroxide (ROO) stim-
tioxidant and induces cytotoxicity in cells leading to oxidative ulates the proximal tubule cell damage in response to OTA
DNA damages (Table 1) (Marin-Kuan et al. 2011). The exposure, inducing a wide range of lesions in the cell
Environ Sci Pollut Res

Table 1 Different studies showed oxidative stress induced by OTA

Model Exposure Dosage Aim of the study Remarks References


time

Male Sprague-Dawley 3 weeks 3, 140, 280, and Protective role of chitosan COS-NPs enhance the antioxidant (Abdel-Wahhab et al.
rats 50 mg/kg b.w nanoparticles singly effect of quercetin and protect 2017)
(COS-NPs) or plus quercetin against the nephrotoxicity of
against OTA-induced oxida- OTA in high-endemic areas.
tive stress and renal
genotoxicity
Male Sprague-Dawley 21 days 3 and 250 mg/kg Determine total phenolics and Promising protective against OTA (El-Nekeety et al.
rats DPPH radical scavenging oxidative stress and 2017)
activity of papaya fruits and nephrotoxicity
evaluation of the protective
role of these extracts against
OTA-induced oxidative dam-
age and nephrotoxicity
Piglets 30 days 0.05 mg/kg Evaluate OTA oxidative stress Inflammation and chemokines (Marin et al. 2017b)
and immune response were higher in the gut than
those in the kidney.
Male Wistar rats 21 days 0.125 and 0.250 mg/kg Oxidative stress due to OTA OTA reduced GSH in the kidney (Rašić et al. 2018)
and increased MDA in the liver
and kidney.
Genotypes mice 21 days 2.5 mg/kg OTA nephrotoxicity mechanism Downregulation of Nrf2 factor, (Loboda et al. 2017)
during HO-1 deficiency miR-29c, influence in miRNA,
induce p53-regulated miR-34a
and pro-fibrotic miR-2
Broiler chicks 60 days 1.6, 3.2, and 6.4 mg/kg OTA oxidative stress in day-old ↓ SOD, GPx, and TAS indicate (Hameed et al. 2017)
of feed chicks oxidative stress
Pig and PK15 cells – – Evaluate porcine circovirus type In vivo trial illustrated that PCV2 (Gan et al. 2018)
2 (PCV2) infection on infection aggravated
OTA-induced nephrotoxicity OTA-mediated poor growth
performance, nephrotoxicity,
p38 phosphorylation, and au-
tophagy as demonstrated by
Atg5, LC3 II, and p62 protein
expressions in the kidney of
pigs, while in vitro trial elabo-
rated that PCV2 infection sig-
nificantly aggravated
OTA-induced nephrotoxicity
as demonstrated by cell
viabilities, annexin V/PI bind-
ing and caspase 3 activities, and
induced p38 phosphorylation
and autophagy in PK15 cells.
Pig sperm 24 h 10 and 100 μM OTA Evaluate OTA for boar OTA decreased sperm motility (Zhang et al. 2018)
spermatozoa motility and via AMPK and PTEN
pathways.
Piglets 30 day 0.05 mg/kg feed Investigate the low level of OTA OTA activates immune response (Marin et al. 2017a)
has a toxic effect pathways, oxidative stress, and
carcinogenicity.
3D4/21 cell lines 48 h Aflatoxin B1 (0, 0.01, Evaluate aflatoxin B1 and OTA Both induce degradation of IκBa, (Hou et al. 2018)
0.02, 0.04, 0.08, 0.16, induced immunotoxicity the phosphorylation of nuclear
0.32, and 0.64 μg/ml) factor kappa B (p65), and the
and OTA (0, 0.1, 0.5, translocation of activated
1, 1.5, 2, 4, and nuclear factor kappa B (NF-κB)
8 μg/ml) into the nuclei.
Human peripheral blood OTA induced oxidative stress- ROS accumulation–oxidative (Liu et al. 2012)
mononuclear cells DNA damage DNA damage–G1 arrest and
(hPBMC) in vitro apoptosis
Male Sprague-Dawley 14 days 5 mg/kg/day Protective effect of lycopene Lycopene overcomes OTA (Palabiyik et al. 2013)
rats against OTA oxidative stress and apoptosis,
protective against
nephrotoxicity
HepG2 cells Zinc supplementation inhibits Zinc reduces OTA-induced DNA (Zheng et al. 2013)
OTA-induced oxidative stress strand breaks, 8-hydroxy-2′--
and DNA damage deoxyguanosine (8-OHdG)
Environ Sci Pollut Res

Table 1 (continued)

Model Exposure Dosage Aim of the study Remarks References


time

formation, and DNA hypome-


thylation. OTA increased the
mRNA expression of
metallothionein1-A (MT1A),
metallothionein2-A (MT2A),
and Cu/Zn superoxide dismut-
ase (SOD1).
Rat 28 days OTA administrated with OTA facilitates karyomegaly, cell (Taniai et al. 2014)
enzymatically modified cycle aberrations, and
isoquercitrin or α-lipoic acid apoptosis.
HepG2 cells Quercetin modulates OTA-induced calcium, ROS (Ramyaa and Padma
OTA-induced oxidative stress production, activates NF-κB. 2014)
and redox signaling. While quercetin meliorated
ROS and calcium release as
well as NF-κB induction and
expression, Nrf-2 nuclear
translocation and expression.
Quercetin’s anti-inflammatory
COX-2, prevent DNA damage
and micronucleus formation.
Rats 4 or 70 or 210 μg/kg bw Oxidative stress and renal OTA induced mRNA level of (Qi et al. 2014b)
13 wee- carcinogenicity induced by clusterin, vimentin and
ks OTA lipocalin 2
Penicillium verrucosum Penicillium verrucosum produce Citrinin regulates cAMP/PKA (Schmidt-Heydt et al.
citrinin signal transduction pathway 2015)
and compete oxidative stress in
different environmental condi-
tions.
Lymphocytes 20 μM OTA-induced cytotoxicity, OTA-induced NO, TNF-α, IL-6, (Periasamy et al.
genotoxicity, and and IL-8 were significantly re- 2016)
inflammatory response duced in the quercetin, which
revealed quercetin exerts a
cytoprotective effect against
OTA-induced oxidative stress,
genotoxicity, and inflammation
in lymphocytes.
Wistar rat liver 6 mg/kg bw Lipid peroxidation induction by Play a role in the observed toxicity (Rahimtula et al.
microsomes, kidney OTA of OTA 1988)
microsomes
Wistar rat male Gastric Every 48 h/3 weeks Protective effect of superoxide SOD + catalase prevents the (Baudrimont et al.
intuba- 289 μg/kg body dismutase (SOD) and catalase nephrotoxicity induced by 1994)
tion weight OTA in rats.
Swiss mice 48 h 2 mg/kg bw Effects of vitamins on the Vitamins E, A, and C also reduced (Grosse et al. 1997)
genotoxicity of OTA OTA-DNA adduct formation in
mice kidney.
Bacillus brevis 10 min 1 mg/mL Study free radical generation in OTA induces free radical (Hoehler et al. 1996)
bacteria as a model system production, enhancing
permeability of the cellular
membrane to Ca2+.
Sprague-Dawley liver 2.5 mM Free radical generation by OTA Oxidative damage may be one of (Hoehler et al. 1997)
microsomes, liver in hepatocytes, mitochondria, the manifestations of cellular
mitochondria, and and microsomes using electron damage in the toxicity of OTA.
hepatocytes cells paramagnetic resonance (EPR)
Bronchial epithelial 4h 10 μM Roles of cyclooxygenase and OTA is biotransformed into (Pinelli et al. 1999)
cells incubated with lipoxygenase in OTA genotoxic metabolite via a
microsomes of genotoxicity in human lipoxygenase, whereas
seminal vesicles of epithelial lung cells prostaglandin H synthetase
pig (PGHS) decreases OTA
genotoxicity.
Fischer rats 4, 8, 24, 0–2.0 mg/kg bw Chemical and biological markers Oxidative stress may contribute to (Gautier et al. 2001)
48 h induced by OTA exposure the mechanism of OTA renal
associated with oxidative toxicity and carcinogenicity in
stress rats over long term exposure.
2 years
Environ Sci Pollut Res

Table 1 (continued)

Model Exposure Dosage Aim of the study Remarks References


time

Dark Agouti (DA), 0.4 mg/kg Life-time study to evaluate the MESNA decreased karyomegalies (Pfohl-Leszkowicz
Lewis rats bw potential protective effect of 2 in the kidney, but had no et al. 2002)
mercaptoethane sulfonate beneficial effect on renal tumor
(MESNA) and N-acetyl cyste- incidence
ine (NAC)
Dark Agouti (DA), 0.4 mg/kg Life-time Life-time study to evaluate if Lack of effect of MESNA on (Son et al. 2003)
Lewis rats bw MESNA leads to a more OTA-induced urinary tract
effective reduction of damage or renal tumor devel-
OTA-induced tumor develop- opment
ment or urinary tract damage
Proximal tubular cells 24–72 h 5.0 μM, 12.5 μM In vivo and in vitro gene In vitro and in vivo gene (Lühe et al. 2003)
(PTC), Wistar rats (in vivo in vitro; 1 and expression comparative study expression data were
and 10 mg/kg bw comparable. Response to
in vitro) oxidative stress-related genes
hypoxia-inducible factor 1 and
catalase was observed.
Human hepatoma— 1 h, 24 h 0–50 μg/mL Genotoxicity of OTA No inductions of mutations in the (Ehrlich et al. 2002)
derived cell line Ames assay, a dose-dependent
(HepG2) induction of micronuclei in the
MN assay, and DNA migration
(comet assay) were detected.
Wistar rats 10, 30, 120 μg/kg bw Kidney low dose OTA response: Low dose induces oxidative stress, (Petrik et al. 2003)
60 days sequence of events leading to apoptosis in proximal, and
cell death distal tubule kidney cells.
Primary proximal 0–24 h 0–100 μM OTA mediated oxidative stress Oxidative stress contributes to (Schaaf et al. 2002)
tubules renal (PT) response in proximal tubular tubular toxicity. Antioxidants
cells, proximal tubu- cells, oxidative stress (α-tocopherol,
lar cell line protection N-acetyl-L-cysteine (NAC))
(LLC-PK1) treatment prevents OTA toxici-
ty.
Human 24 h 0–40 μg/mL Genotoxicity of OTA Dose-dependent induction of (Knasmuller et al.
hepatoma-derived DNA single-strand breaks 2004)
cell line (HepG2) (comet assay) and micronuclei
(MN)
Fetal rat telencephalon 24–48 h, 0–20 nM Adverse effect of OTA in the Brain inflammatory response (Zurich et al. 2005)
aggregating cells 9 days brain induction of HO-1, iNOs,
PPARγ, cytoskeletal damage
Human fibroblast cells 48–72 h 0–50 μM Oxidative stress protection study Reduction of free radical species (Russo et al. 2005)
production and DNA damage
prevention by cyanin
3-O-β-D-glucoside (C3G)
Sprague-Dawley 15 days 3 mg/kg Oxidative stress protection Preventive effect against (Abdel-Wahhab et al.
in vivo study OTA-induced oxidative stress 2005)
and lipid peroxidation by mel-
atonin
Human hepatoma— 48–72 h 35–10 mM Oxidative damage protection No cytotoxicity protection (Hundhausen et al.
derived cell line study observed with Vitamine E, 2005)
(HepG2) polyphenols
F344 Fischer rats 2 weeks 0–2000 μg/kg bw Genotoxicity of OTA DNA strand breaks in target and (Mally et al. 2005)
nontarget tissues probably
involving oxidative stress
mechanism.
Human 24, 48, 0–100 μM Oxidative stress protection study OTA-induced oxidative stress (Guerra et al. 2005)
hepatoma-derived 72 h damage. Protective effect by
cell lines (HepG2), Cyanidin-3-O-β
human colonic ade- glucopyranoside (C-3-G)
nocarcinoma cell line
(Caco-2)
Rat lymphoid cells 1h 0.5, 2, 20 μM OTA immune function Protein synthesis inhibition, (Alvarez-Erviti et al.
modification oxidative metabolism of OTA, 2005)
prostaglandin synthesis
implicated in NK cells toxicity
V79 (Chinese hamster 1–24 h 2.5, 100 μmol/L OTA Cytotoxicity and oxidative DNA (Kamp et al. 2005b)
lung fibroblasts) damage already at low doses
Environ Sci Pollut Res

Table 1 (continued)

Model Exposure Dosage Aim of the study Remarks References


time

cells, CV-1 (African Relevance of OTA-induced oxi- could be a relevant factor for
green monkey, kid- dative damage in nephrotoxi- the nephrocarcinogenicity
ney) cells, primary rat city and carcinogenicity
kidneycells
F344 rats 0.03, 0.10, 0.30 mg/kg Evaluate relevance of Tumors in rat kidney may be (Kamp et al. 2005a)
bw OTA-induced oxidative dam- attributable to oxidative DNA
age on nephrotoxicity and car- damage in combination with
cinogenicity cell-specific cytotoxic and
proliferation-stimulating effects
as cell-signaling response
Wistar rats 7, 14 and 0.5 mg/kg bw Effect of OTA on protein Increased protein carbonyls in the (Rudeš 2006)
21 days oxidation in kidney and liver kidney and liver
Pig kidney microsomes, Cells: 2, 7, 0.5, 1.0, 2.5 μM Genotoxicity of the hydroquinone OTQ-mediated adduct spots form (Tozlovanu et al.
human bronchial 24 h (OTHQ) metabolite of OTA in a dose- and-time-dependent 2006)
epithelial cells, manner
human kidney cells
Wistar rats 15 days 5 ng; 0.05 mg; Effect of OTA on DNA damage Oxidative stress responsible for (Domijan et al. 2006)
0.5 mg/bw OTA-DNA damage as shown
by Fpg modified comet assay
Wistar rats 7, 14, and 0.5 mg/kg bw/day Genotoxic potential of OTA OTA-induced DNA strand breaks (Zeljezic et al. 2006)
21 days measuring DNA strand breaks were detected, OTA
(comet assay) in the kidney concentration in the kidney and
duration of the treatment
correlated with severity of the
DNA damage.
Swiss mice 24 h 10 mg/kg Immune cell response after acute OTA-induced oxidative stress (Ferrante et al. 2006)
OTA exposure response responsible for its
own toxicity
F344 rats 7 and 300 mg/kg bw OTA mechanism of Oxidative stress and metabolic (Marin-Kuan et al.
21 day- action-microarrays study in response modulated involving 2005)
s; 4, 7, liver and kidney mainly Nrf2 and HNF4α
and pathway disruption
12 mo-
nths
Swiss mice 2 weeks Acute 3.5 mg/kg; Effect of chronic low dose OTA Low doses exposure caused global (Sava et al. 2006b)
chronic 4, 8,16 mg/kg exposure on regional brain oxidative stress.
oxidative stress and stratial DA
metabolism
Swiss ICR 6, 24, 72 h 0–6 mg/kg bw Oxidative stress and OTA Acute depletion of striate DA on a (Sava et al. 2006a)
neurotoxicity background of globally
increased oxidative stress and
transient inhibition of oxidative
DNA repair.
Wistar rats 90 days 289 μg/kg bw Early effects of chronic OTA Reduction in the ability to (Gagliano et al. 2006)
administration in liver counteract oxidative stress in
liver
Eker and wild-type rats 1–14 days 210 μg/kg bw Early carcinogen-specific gene Oxidative DNA damage response (Stemmer et al. 2007)
expression study genes, general stress response,
and cell proliferation
Male Fischer 344; Rats 300 μg–100/kg bw; Demonstration of cellular defense OTA induces depletion of (Cavin et al. 2006)
primary hepatocytes; 2 years; 1.5–6.0 μM reduction by OTA antioxidant defense by
adherent proximal in vitro inhibition of Nrf2 responsible
tubules epithelial culture for oxidative stress response.
NRK cells; rat liver 24–48
RL-34 h
Human renal proximal 50–800 μM Evaluate single-strand DNA Oxidative stress precedes (Arbillaga et al.
tubular epithelial cell breaks and oxidative damage cytotoxicity and genotoxicity. 2007a)
line HK-2 induction by OTA
Human renal cell line 6 and 24 h 50 μM Mechanism of action Significant increase in ROS level (Arbillaga et al.
HK-2 study-microarrays and oxidative DNA damage 2007b)
Rats 4 weeks 289 μg/kg Oxidative stress protection study Melatonin protection against (Sutken et al. 2007)
OTA-induced oxidative dam-
age in the liver and kidney.
CoQ protective in liver
Environ Sci Pollut Res

Table 1 (continued)

Model Exposure Dosage Aim of the study Remarks References


time

Neural stem/progenitor 0.01–100 μg/mL Vulnerability of brains mouse Robust increased in total and (Sava et al. 2007)
cells (NSCs) cells to OTA mitochondrial SOD activity.
OTA impaired hippocampus
neurogenesis.
Human epithelial 100 μM Effect of OTA on barrier function Loss of microdomains associated (Lambert et al. 2007)
colorectal impairment with tight junctions maybe due
adenocarcinoma to oxidative events
Caco-2 cells
Pig kidney cell line 24 h 0, 10, 15, 20 μM Oxidative stress protection study OTA-induced ROS. Scavenging (Costa et al. 2007)
LLC-PK1 by catechins (epigallocatechin
gallate (EGCG) and epicatechin
gallate (ECG))
Rats Sprague-Dawley 4 weeks 200 ppb Oxidative stress protection study OTA-induced oxidative stress and (Di Giacomo et al.
DNA damage chemoprotection 2007)
by cyanidin 3-O-β-D-glucoside
Chinese Hamster lung 0–438 μM OTA mutagenicity OTA is mutagenic at cytotoxic (Palma et al. 2007)
V79 cells; doses in mammalian cells via
Lymphoma mouse oxidative DNA damage
LY5178 cells induction.
Rat Wistar 15 days 5 ng/g bw 50 ng/kg bw Oxidative damage study (proteins Malondialdehyde (MDA) and (Domijan et al. 2007)
and lipids) protein carbonylation (PC) in-
crease in kidney > liver
Human hepatocytes 0–100 μM Decrease GSH No induction of heat shock protein (Hassen et al. 2007)
HepG2; Monkey (HSP)
kidney Vero cells
Rat F344 7 and 0.5 mg/kg bw Mechanism of action Oxidative stress, calcium (Arbillaga et al.
21 days study-microarrays homeostasis, cytoskeleton 2007b)
structure
Rat Sprague-Dawley 15 days 3.0 mg/kg bw Oxidative stress protection study OTA-induced oxidative stress (Abdel-Wahhab et al.
chemoprotection by Inula 2008)
crithmoides
Porcine kidney tubule 24 h 1–100 μmol/L Impact of OTA on Nrf2, AP-1 Enhanced production of ROS, (Boesch-Saadatmandi
cells LL-PK1 activity, antioxidant enzymes, GST impairment. Nrf2 and et al. 2008)
and GST AP-1 disruption by OTA.
Impairment of the detoxifica-
tion machinery
Porcine kidney tubule 6–24 h 1–100 μmol/L Characterization effect of OTA Nrf2 potential signal transduction (Boesch-Saadatmandi
cells LL-PK1 on Nrf2 response pathway by which OTA et al. 2009)
impairs its own detoxification
BALB/c macrophage 24, 48, 30 nM–100 μM OTA immunotoxicity and Induction of iNOs, COX-2, and (Ferrante et al. 2008)
J774a cell line 72 h modulation inflammatory NF-κb expression by OTA.
process OTA is an immunotoxic
compound.

components. Other studies have suggested that OTA toxicity ROS in vitro as well as in vivo. In order to verify the correla-
could be also attributed to its isocoumarin moiety propriety tion between OTA and ROS-induced oxidative brain, liver,
while involving the lactone carbonyl group and engaging Fe2+ and kidney cell damage of OTA-treated rats, they were eval-
and Ca2+ mobilization pathways which leads to uncoupling of uated for non-proteic thiol groups (RSH), lipid hydroperoxide
oxidative phosphorylation and raising the level of hydroxyl (LOOH) levels, and DNA fragmentation. Alterations in
radical through the Fenton-like reaction while describing a LOOH and RSH were observed confirming the oxidative
pro-oxidant behavior of the cell (Sorrenti et al. 2013; Zheng pathway involvement in the OTA-induced damages in all
et al. 2013). The oxidation of OTA phenol by chemical, elec- the examined tissues. It has been pointed out that OTA could
trochemical, and photochemical processes has been found to generate excess NO in kidney and liver inducing iNOS and
generate an OTA hydroquinone/quinone couple. forming the pro-oxidant ONOO− causing an increased nitrite
Hydroquinone is formed through the quinone reductions and nitrate levels and consequently in nitrosative stress as
which leads to the creation of glutathione conjugate of OTA shown in Table 1 (Sorrenti et al. 2013). Other studies have
and consequently causes redox cycling and production of revealed that HepG2 cells exposed to OTA have
Environ Sci Pollut Res

modifications in the transcription of the zinc transporter, ZnT process has been observed in different cell types namely kid-
(SLC30), and Zip (SLC39) genes which are implicated in the ney, liver, neuronal, intestinal, gastric, renal tubule cells, de-
control of the intra- and intercellular zinc translocation. veloping embryos, and lymphocytes and has been associated
Furthermore, OTA can interact directly with Zn2+ and act on with the pathogenesis of many diseases. In fact, notable mor-
the transporter proteins and on the expression of multiple phological modifications could distinguish the apoptotic cells
metallothioneins which results in the depletion of the intracel- and have been recognized in different cell lines following
lular Zn2+ and it induces oxidative damage. Supplementation chromatin aggregation, DNA fragmentation, appearance of
with the zinc chelator N,N,N′,N′-tetrakis (2-pyridylmethyl) apoptotic bodies, apoptosis protein development, proteolysis
ethylenediamine (TPEN) strongly lessened the OTA- of apoptosis specific substrates or exposure of
mediated oxidative stress (Zheng et al. 2013). phosphatidylserine on the outer layer of the cell membrane,
It is well-established that OTA activation is possible due to or some enzyme activation as well as the development of
the production of peroxidase enzymes that adduct C8-OTA chronic tubulointerstitial kidney damage (Kumar et al.
while cytochrome p450 adduct OTA-mediated DNA using 2014). It has been demonstrated that renal tubular lesions,
32
P-post labeling which shows the relationship of OTA struc- nuclear effect, and DNA fragmentation observed in HL-60
ture (Manderville and Pfohl-Leszkowicz 2008). It has shown human promyelotic leukemia cells and the activation of
that OTA causes oxidative DNA damage in the outer medulla caspase-3 and c-Jun amino-terminal kinase (JNK) in
of male rat kidney that leads to mutagenicity. Therefore, it MDCK-C7 cells result from apoptosis significantly stimulated
gives structural evidence of DNA adduction caused by OTA by OTA exposure. Likewise, intracellular apoptotic bodies
which is direct genotoxicity in OTA-mediated renal carcino- have been found in the livers of mice and within myc
genesis (Pfohl-Leszkowicz and Manderville 2011). Among transfected cell lines following OTA intoxication (Atroshi
OTA, OTBr, and OTB toxins, the highest cytotoxicity was et al. 2000). Nephrotoxicity of this mycotoxin was reported
exhibited by OTA due to the presence of C5-Cl atom toward in humans as well as in poultry, swine, mice, rats, and rabbits,
direct genotoxicity in OK and WI26 cell model. These find- and increased apoptotic cell percentage was observed partic-
ings reveal that cytotoxicity occurs due to oxidative DNA ularly over 60 days post-exposure (Kumar et al. 2014). It has
damage while mutagenicity stems from genotoxicity been identified that the inhibition of p53 activation, especially
(Hadjeba-Medjdoub et al. 2012). the suppression of JNK activation as well as the downregula-
On the other hand, there is no significant difference in the tion of Bcl-xL, enhances OTA ability to stimulate apoptosis in
metabolism of OTA in the male and female rat; however, the MARC-145, Vero monkey kidney cells, and HEK293 human
extent of OTA metabolism was greater in male than that in kidney cells significantly (Li et al. 2011). Furthermore, in vivo
female rats which shows that male rats are highly sensitive to and in vitro apoptotic changes in rat and mouse livers have
the metabolism of OTA toxicity (Tozlovanu et al. 2012). It has been observed as OTA hepatotoxic effects which are mediated
been reported that OTA genotoxicity can be lessened with the by the released TNF-α from Kupffer cells. OTA physiological
pretreatment of the model with N-acetylcysteine and 2- pro-apoptotic stimuli of TNF-α essentially involve TNFR I
mercaptoethane sulfonate which act as free thiol diminisher receptor as well as the ActD transcriptional inhibitor as a spe-
in the kidney (Pfohl-Leszkowicz et al. 2002). cific cytotoxic event, although it has been found that OTA
could mediate apoptosis even in the absence of Kupffer cells
Apoptosis and TNF-α following a direct binding to TNFR I, which
mimic cytokine-induced apoptosis. Apoptosis outbreak im-
OTA toxicity, even with relatively low doses, was found to plies binding of TNF-α to the extracellular domain in order
cause either apoptotic or necrotic cell death in accordance with to activate the intracellular domain, referred to as adaptor pro-
the dose and duration of exposure. Lesions’ appearance in the tein which is the cell membrane receptor TNF-α receptor I
plasma membrane characterizes necrosis and apoptosis that (TNFRI) related to the death domain (TRADD). TRADD
could be generated by heat, toxins, free radicals, growth fac- stimulation generates the recruitment and stimulation of the
tors, and cytokines (Zhang et al. 2009). Both processes can initiator aspartate specific cysteine protease caspase-8, as a
occur within different biological systems leading to cell death; stimulator of additional apoptotic actions (Essid et al. 2012).
however, apoptosis is known to be the predominant cell ad- The mitogen-activated protein kinase kinase kinase
aptation system that varies biochemically and morphological- (MAPKKK, MAP 3K) family member apoptotic signal-
ly from necrosis. It has been suggested that the apoptotic cel- regulating kinase 1 (ASK1), which is mainly involved in cell
lular mechanism is mostly trigged by OTA which induces apoptosis, is expressed during OTA-mediated renal cytotox-
diverse toxic effects on several organs and leads to cell death. icity and apoptosis. Investigation on the regulatory mecha-
Therefore, OTA toxicity leads to oxidative damage and calci- nisms of ASK1 in OTA-induced renal cytotoxicity using
um entry which stimulates, in vivo and in vitro, DNA alter- RNA interference of ASK1 in HEK293 cells has shown that
ation and thereby apoptosis (Kőszegi and Poór 2016). This ASK1 significantly promotes OTA-induced apoptosis. It has
Environ Sci Pollut Res

been found that ASK1 downregulates DHRS2 expression, different concentrations causes DNA damages like oxidation
which has a protective role against apoptosis, which is due and strand breakage of the DNA bases that give rise to differ-
to OTA results in partially cell apoptosis (Liang et al. 2015). ent DNA lesions which are permanent cellular alterations
Moreover, it has been demonstrated that ASK1 inhibits Prx4, causing ultimate genomic instability and thereby harmful
which is an antioxidant enzyme, leading to cell apoptosis as a transformations within the cells (Arbillaga et al. 2007a, b).
response to increased free-radicals, via activation of JNK. In OTA-treated kidney epithelial cells of rats have shown
addition, it has been reported that cell apoptosis might be degenerated cells after the use of mitosis marker phospho
induced by OTA via regulation of CDK1 and DHRS2 pro- H3 (Ser10). Gene expressions were analyzed after OTA treat-
teins, which are related to cell death due to ASK1 activation ment at various dose levels that can initiate tumor formation in
by OTA, probably as a result a cascade of activation of JNK the kidney. Many genes were found altered such as the genes
(Table 1). This suggested that the OTA initiates apoptosis involved in G1/S, G2, and M stages of the cell cycle including
through multiple pathways, including mitochondrial mem- cyclins, cyclin-dependent kinase inhibitors, cyclin-dependent
brane potential loss, nucleotide metabolism, cell cycle check- kinases, and genes responsible for the proliferation and sur-
points, DNA repair mechanisms, lipid and lipoprotein metab- vival of cells known as oncogenes and tumor suppressor
olism, and the blockade of RNA synthesis and ASK1 is im- (Gordon et al. 2018). Other genes associated with DNA dam-
plied in these metabolic pathways (Liang et al. 2015). So as to age, chromosomal instability, spindle assembly checkpoints,
further distinguish the impact of OTA on cell viability, cyto- aneuploidy, and poor prognosis along with growth factor
toxic activity of OTA was evaluated separately and along with encoding genes and their receptors were altered. It is well-
citrinin (CTN) in HepG2 cells to induce hepatotoxicity. It has established that the alteration in expression of different mitosis
been found that OTA and CTN act in harmony in mediating regulators that are important in centrosome function, G2/M
the cytotoxic effects in the HepG2 cell line and that OTA was progression, chromosome condensation and segregation,
found to be responsible for nuclear and morphological chang- spindle checkpoint, and cytokinesis was observed indicating
es of apoptosis while the interaction between CTN and OTA that their overexpression in the renal cells of the animal could
was greatly found to lead to apoptotic death of hepatocytes. be the main tumor caused by OTA. Furthermore, OTA-treated
Besides, OTA was shown to alter the mitochondrial trans- immortalized human kidney epithelial cells (IHKE) have
membrane potential, whether tested individually or in combi- shown nuclear enlargement and mitotic aberrations which
nation with CTN, indicating that mitochondria undergo confirm that OTA toxicity generates mitotic disruption
changes in nuclei after which apoptosis becomes morpholog- (Mally 2012). Besides, analyzing the effects of OTA on cell
ically evident through caspase-8-associated death receptor division in IHKE cells has confirmed that OTA generates
pathway or caspase-9-associated mitochondria-mediated in- aberrant condensation in mitotic chromosomes and alters core
trinsic pathway. Therefore, in both pathways, caspase-3 is histones phosphorylation and acetylation of sister chromatid
activated, following the cleavage of procaspase-3, in order to separation which leads to sustain mitotic arrest and exit from
express the cleavage of PARP protein which takes part in the mitosis without nuclear or cellular division. OTAs’ ability to
mechanism of repair of DNA and therefore apoptosis is stim- affect the enzymatic activity of those important for regulating
ulated. OTA and CTN, administered separately or combined, the posttranslational histone modifications was tested, and
have been found to enhance caspase-9 cleavage expression loss of histone H3 phosphorylation at threonine 3 was ob-
while disadvantaging procaspase-8 expression and caspase-8 served as well as a significant blockage of histone acetyltrans-
cleavage indicating that the mycotoxins initiate the ferase (HAT) activity was seen in a nuclear cell extract. These
mitochondria-mediated intrinsic pathway, which includes cas- findings suggest that OTA could inhibit histones acetylation
pase-9. Furthermore, the mycotoxins have been found to re- regulator and non-histone proteins at lysine residues and have
duce procaspase-3 expression and to increase caspase-3 ex- revealed the interference of OTA on the mitotic machinery
pression cleavage while significantly enhancing the plus a mitotic level cell cycle inhibition due to the loss of
proapoptotic protein Bax and PARP cleavage expressions. HAT function (Czakai et al. 2011; Liu et al. 2012). While
These findings have confirmed the mitochondria involvement reviewing the mechanism of action of OTA toxicity for renal
in apoptotic cell death induced by OTA and CTN individually carcinogenicity, a combination of genetic instability and in-
as well as in combination (Gayathri et al. 2015). creased proliferative changes were found as consequences of
OTA-mediated disruption of mitosis due to active trans-
Influence on mitosis porters. In vitro studies conducted on OTA-treated monkey
kidney epithelial cells and IHKE cells have shown abortive
It has been reported that OTA toxicity mediates disruption of mitosis, cell degeneration, the formation of multiple nucleated
mitosis and generates mitotic abnormalities or aberrant exit cells, and abnormal mitotic figures (Mally 2012). Besides,
from mitosis without nuclear division that could generate cell asymmetric spindles, aberrant chromosome alignment, and
death at the level of mitosis. In fact, exposure to OTA at highly condensed metaphases with separated chromatids were
Environ Sci Pollut Res

observed proving further that OTA inhibits sister chromatid expression at messenger RNA (mRNA) and protein levels
separation and genetic material distribution towards the (Cui et al. 2010). This is mandatory in the G2 phase to prog-
daughter cell during mitosis (Mally 2012). Other studies have ress to mitosis, by dephosphorylating Cdc2 and inactivating
shown that OTA treatment triggers breaking of the DNA after the binding between cyclinB1 and Cdc2 which very important
FPG treatment in rat’s liver and kidney and treatment of IHKE for activating cyclinB1–Cdc2 complexes. Hence, inappropri-
cells proposing that the toxin blocks the metaphase/anaphase ate inactivation of Cdc2 during mitosis leads to polyploidy
transition and induces disruption of mitosis by showing aber- and nuclear aberrations. These findings outline that OTA is
rant mitotic figures and giant cells as well as chromosomal able to dramatically obstruct cell proliferation leading to cell
instability through inhibition of microtubule assembly apoptosis (Liu et al. 2012). Moreover, studies have reported
(Rached et al. 2006). These findings of both in vivo and that OTA toxicity results in the formation of DNA adducts and
in vitro suggested that OTA involves instability and tumori- subsequent induction of apurinic/apyrimidinic (AP) endonu-
genesis via counteract with mitosis process during cell divi- clease formation, which itself is the enzyme responsible for
sion. OTA treatment mediated nuclear aberrations which ex- DNA base excision repair pathway. It also inhibits topoisom-
hibited that OTA could influence the functionality of some erase II activity leading to disorders of the normal DNA repair.
mitosis key regulators which brings to block asymmetric cell These findings have elaborated that OTA is causing DNA
division and to increase aneuploidy probability and therefore strand breaks following alterations in Rad18, Brip1, Brcc3,
tumorigenesis and acquisition of a malignant phenotype. and chek1 gene expression. These are DNA double-strand
However, the toxin is categorized as a non-mutagenic and break repair (DSBRs) genes. Analysis of global gene expres-
non-DNA-reactive carcinogen (Pfohl-Leszkowicz and sion similarities under OTA exposure has revealed the molec-
Manderville 2011). Similarly, it has been reported that OTA ular changes of the upregulation of the genes which are related
intoxication leads to the observation of karyomegaly charac- to cell cycle transition and or progression to G1/S (Ccne1),
teristics and disruption of normal mitosis via cyclin-dependent associated to the cell cycle checkpoint at G2/M (Chek1), and
kinase inhibitor expression as well as topoisomerase II expres- involved in cell cycle arrest at G2/M phase (Wee1). Besides,
sion (Adler et al. 2009; Mally 2012). results have supported that the cell proliferation at the S3
segment was enhanced and cycle progression was suppressed
as a consequence of the cell cycle arrest at the G2/M phase.
Induction of cell cycle arrest Another study that was performed in order to highlight
nephrotoxicity-promoting molecular mechanisms of OTA
The processes involved in cell signaling like inhibition of gap has shown that OTA induces DNA damage and S phase arrest
junction intercellular communication (GJIC) interfere with in human embryonic kidney cells (HEK392) following the
microtubule dynamics, and mitotic spindle formation or in- formation of DNA comet tails. The important expression of
creased histone deacetylase (HDAC) enzymatic activity has γ-H2AX and the critical lower expression of cyclin A2, cyclin
been noticed upon OTA treatment. Undesirable effects of E1, and CDK2 are the key cell cycle regulatory factors. In the
OTA treatment on cell cycle arrest were studied in kidney same context, the presumed OTA-induced G2 arrest pathways
cells and in lung fibroblasts, and these changes were assumed were evaluated while exploring the oxidative DNA damage
to occur in order to repair the DNA and to minimize replica- and the ataxia telangiectasia-mutated (ATM) functions in
tion and segregation of the damaged DNA due to the obser- GES-1 cells (Hibi et al. 2013; Yang et al. 2014a). Studies have
vation of the phenomenon prior to DNA replication (through revealed that the intracellular ROS was highly increased due
G1 checkpoint to S), during DNA replication, or before mito- to OTA exposure, leading to DNA damage as well as en-
sis (through G2 checkpoint to mitosis) (Wen et al. 2016). It has hanced levels of 8-OHdG and DNA double-strand breaks.
been reported that OTA decreases CDK4 and cyclinD1 ex- Furthermore, it has been underlined that, in response to
pression in hPBMC leading to arrest the G1 cell cycle phase DNA DSBs, the toxin generates ATM protein phosphoryla-
due to high coordination between the cell cycle and the acti- tion and molecules Chk2 and p53 (Cui et al. 2013). In addi-
vation of cyclin and cyclin-dependent kinase (CDK), especial- tion, investigations on OTA-induced G2 arrest modulation
ly cyclinD1/CDK4/6 kinase complex which are the initiation have involved the MAPK pathway in GES-1 cells, taking into
points to mitogenic signals in G1 phase. Furthermore, it has account the important role MAPK plays in cell cycle progress.
been found that OTA toxicity blocks the G2 phase in immor- Results have demonstrated that cell exposure to OTA pro-
talized human gastric epithelial cells (GES-1) and in Chinese vokes ERK inhibitor PD98059 and p38 inhibitor SB203580
hamster lung fibroblasts (Liu et al. 2012; Chen et al. 2018a, b). activation, which are MAPK family members and remarkably
It might affect proper regulation of the cell cycle and thereby permutes Cdc25C/p-Cdc25C, Cdc2/p-Cdc2, cyclinB1, and
maintain the genomic integrity as well as chromosome insta- cyclinB1–Cdc2 complex repression. Consequently, ERK
bility. In fact, OTA reduces the cyclinB1–Cdc2 complex and p38 MAPK are recognized to have a major role in
which significantly influences Cdc25C, Cdc2, and cyclinB1 OTA-induced G2 phase arrest in human gastric epithelium
Environ Sci Pollut Res

cells and could induce human gastric carcinogenesis (Wang reduces the cells’ responsiveness to mitogens which simulate
et al. 2012). within human T cells and increases the Ca 2+ release from
intracellular stores and Ca 2+ influx across the plasma mem-
Other mechanisms branes giving rise to Ca2+ levels. The toxin has also effects on
the phagocytic function of the reticuloendothelial system by
The harmful effects of OTA have also been associated with reducing macrophages number and phagocytic activity as well
organogenesis, immunosuppression, organ immune decrease, as bacteriolytic capacity and delayed immune response to tu-
antibody response reduction, and immune cell activity distor- berculin (Marin and Taranu 2015). Inflammation is also an-
tion (Ha 2015). The mechanism of toxicity of OTA in the other consequence of the toxin following the increase of vas-
organisms has revealed that low or moderate OTA addition cular permeability and neutrophils (PMN) infiltrating in tis-
decreases the level of lymphocytes and macrophages as well sues and stimulation of cytokine production by immune cells.
as modulates cytokine secretion in order to intensively affect These small peptides are vital inducers of immune and inflam-
the mucosal immunity, which is the first obstacle for pathogen matory responses. It has been observed that OTA eliminates
mucosal immunity and thereby could influence susceptibility bacterial lipopolysaccharides (LPS), which act as a stimulat-
to infectious diseases (Darif et al. 2016). For instance, in ing agent for monocytes/macrophages, production of tissue
zebrafish embryos, OTA treatment has shown improper heart factor (TF), and suppression of plasminogen activator
looping and heart chambers narrowing as well as a damaged inhibitor-2 (PAI-2) response resulting in the impairment of
renal morphology and a decreased rate of glomerular filtra- blood clotting activation, fibrin formation, and cell activation
tion. Diminished expression of prolactin receptor a gene (Rossiello et al. 2008). Thereby, the monocyte/macrophage
(PRLRa) has also been found within these embryos, which role in fibrin accumulation has been found dysfunctional caus-
is indeed involved in the organogenesis and osmoregulation ing a decreased inflammatory response and weakened cell-
within vertebrates. Beside suppressed phosphorylation of the mediated immune response. Moreover, the toxin and its me-
transcription activators (STAT5), promoting cell growth and tabolites diminish the metabolic activity, proliferation, differ-
differentiation during development was observed in addition entiation, and membrane integrity of the immune cells. It pro-
to phosphorylation of the AKT, an essential protein in mam- vokes migration of white blood cells and immune cells to bone
malian cell signaling. Furthermore, microRNA profiling has marrow and hypocellularity resulting in their repression while
demonstrated the upregulation of microRNA-731 (miR-731). minimizing bone marrow macrophage-granulocyte progeni-
All these observations together have suggested that OTA tox- tors and hematopoietic stem cell number as well as erythro-
icity together with miR-731 and PRLR signaling cascade poiesis. Furthermore, it has been noticed that OTA promotes
modulation distorts normal renal development in order to ex- tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6),
press negatively impacted organogenesis within zebrafish which trigger the narrowing of thymus, spleen, bursa, and
(Wu et al. 2016). Apart from that, scientific evidence has lymph node cells resulting in their abnormal functioning and
shown that OTA exhibits immunotoxic properties and mech- depletion (Asrani et al. 2016; Marin and Taranu 2015).
anisms that are underlying immunotoxicity and have indicated Evidence has demonstrated that CD69 and CD25 markers
that the toxin adjusts immune response, even at insignificant have been expressed on the cell surface as a response to low
ranges under the toxic threshold by reducing humoral and OTA concentrations and IL-6 has been produced in lympho-
cellular immunity. Thus, it has been suggested that the result cytes after mitogenic stimulation; however, higher amounts
of the toxin interference with cellular metabolism could lead block the expression of CD69, CD25, CD71, and TNF-α
to immunosuppressive effects, especially after chronic expo- production (Darif et al. 2016). Moreover, it was pointed out
sure. For instance, it has been demonstrated that OTA plasma that MAPK signaling pathway is involved in the mechanism
levels and the number of CD4+T lymphocyte population in of OTA toxicity while including p38 extracellular signal-
the oral mucosa are positively correlated, explaining that OTA regulated kinases (ERK) and c-Jun N-terminal kinase (JNK),
stimulates mucosal inflammation which is believed to be pro- which are proteins that participate in intracellular signaling
moting the HIV transmission to neonates by up to 11-fold in cascades at the time of proliferation, differentiation, cellular
South Africa (Darif et al. 2016). Moreover, it has been found stress responses, and apoptosis (Table 1). A study conducted
that supplementation of food contaminated with OTA to ani- on PK15 cells and porcine primary splenocytes has demon-
mals has suppressed immunoglobulin-containing cells in all strated that OTA induced p38 and ERK1/2 phosphorylation in
the lymphoid tissues resulting in a remarkable reduction in the both types of cells, and the results have indicated that neph-
total serum immunoglobulin levels and thereby an increased rotoxicity due to OTA was mediated by p38 signaling path-
infection frequency (Dwivedi and Burns 1984). Furthermore, way and immunotoxicity by ERK signaling pathway (Gan
it has been proved that OTA suppresses the lymphocytes’ et al. 2017). Other OTA toxicity features have been investi-
blastogenesis by decreasing IL-2 production impairment and gated and have highlighted that OTA significantly enhanced
IL-2 receptor expression of activated T lymphocytes. It also ornithine decarboxylase activity, DNA synthesis, hyperplasia,
Environ Sci Pollut Res

and the expression of cyclin-D1 and COX-2, which are short- the cytosine ring of DNA catalyzed by DNA methyltransfer-
term markers of skin tumor promotion, explaining that the ases (DNMTs) as shown in Fig. 2. Zheng et al. (2013) dem-
toxin could promote skin tumor. In fact, it has been found that onstrated DNA hypomethylation induced by OTA treatment
OTA cytotoxicity decreases DNA synthesis and impacts in HepG2 cells for the first time. Another study reported a
phosphorylation and the resulting activation of epidermal time- and dose-dependent alternation of global DNA methyl-
growth factor receptor (EGFR) and its subsequent signaling ation profile after long-term OTA exposure (Li et al. 2015).
pathways via Akt, ERK1/2, p38, and JNK mitogen-activated High-dose (210 μg/kg·bw) OTA treatment induces DNA
protein kinases (MAPKs) (Fig. 1). Besides, it causes suppres- hypermethylation in promoters of E-cadherin, N-cadherin,
sion of c-Jun, c-Fos, cyclin-D1, and COX-2 as well as de- and cell adhesion–related genes of rat kidneys treated with
creased binding of nuclear factor-kappa B (NF-κB) and AP- OTA for 13 weeks. However, observed deleterious effects
1 transcription factors to the promoter region of cyclin-D1 and disappeared after 26 weeks of OTA treatment. This suggests
COX-2 genes. Thus, it has been suggested that OTA is able to alternate DNA hyper- and hypomethylation occurrence in var-
proliferate cells and to promote skin tumors while involving ious anatomical or genomic loci (Li et al. 2015). Furthermore,
EGFR-mediated MAPKs and Akt pathways along with the mentioned study confirmed observed changes in DNA
NF-κB and AP-1 transcription factors and engaging cyclin- methylation levels by alterations of DNMT enzymes, where
D1 and COX-2 as target genes to promote OTA tumor activity the expression of DNMT1 and DNMT3b mRNAs consider-
(Kumar et al. 2012). Alteration of intracellular calcium ho- ably raised at the end of the13-week OTA exposure (Li et al.
meostasis due to OTA exposure has also been investigated 2015). Conversely, no alterations in global DNA methylation
in relation to genes involved in the calcium homeostasis such status were exhibited in MDCK and BME-UV1 cells after
as chemokine (c-c motif) ligand 14 (CCL1), CD24 antigen, OTA treatment in an in vitro study by Giromini et al.
ATPase AT2A2, and the protein phosphatase PP3CB. (2015). Different contradictory reports on OTA-related DNA
Possible calcium homeostasis deregulation has been noticed methylation levels in kidney cells may be explained by its
through inhibition of regucalcin (RGN) gene and downregu- heterogeneity, as it consists of many cell types that may adopt
lation of senescence marker protein 30 (smp30). Besides, de- different DNA methylation levels (Zhu et al. 2017).
pletion of intracellular calcium stores has been reported in Hep
G2 cells which had a considerable decrease in calcium influx OTA and histone modifications
indicating the OTA has an inhibitory action on store-operated
Ca+2 channels. Furthermore, biotransformation features and Histones are alkaline DNA-binding proteins that facilitate the
involvement of different transporters have been associated packing of DNA into nucleosomes, which are the fundamental
with OTA toxicity. Gene expression assays have associated subunits of chromatin. They are highly conserved and can be
OTA exposure with transporter deregulation as well as genes grouped into 5 classes: H2A, H2B, H3, and H4 which are core
related to biotransformation and detoxification processes and histones and H1/H5 which serves as linkers (Bhasin et al.
have been associated with necrotic lesions observed in the 2006). Histones are involved in the regulation of a number
histological examinations. Enzymes such as alcohol dehydro- of biological processes, such as gene expression, DNA repair,
genase 1, argininosuccinate lyase, glutamine synthetase, mitosis, and meiosis (Alberts et al. 2002; Bannister and
GST’s alpha, P1 and Ya, and sulfotransferase and K2 are Kouzarides 2011). The interaction of histone DNA and nucle-
altered. In addition, downregulation of genes related to trans- ar proteins can be disrupted by post-translational modifica-
porters and xenobiotic metabolism was also observed such as tions, resulting in altered gene expression. Mentioned modifi-
organic anion transporters (OAT) or relevant CYP450s, such cations can be acetylation, phosphorylation, methylation,
as CYP2C11 (Vettorazzi et al. 2013). ubiquitination, SUMOylation, citrullination, and ADP-
ribosylation (Zhu et al. 2017). A recent study on the relation-
ship between ochratoxin toxicity and mitosis in immortalized
Epigenetic alterations caused by OTA human kidney epithelial cells showed that OTA treatment
caused aberrant condensation of mitotic chromosomes and
OTA and DNA methylation premature sister chromatid separation, which has been related
to altered acetylation and phosphorylation of core histones.
The emerging field of epigenetics involving the modulation of Moreover, phosphorylation of histone 3 at threonine 3 posi-
gene functions and/or its regulation without changing the pri- tion (H3T3ph) has been completely lost at OTA concentra-
mary DNA sequence has gained significant interest in recent tions ≥ 10 μM. This is particularly important since histone 3 is
years as one of the most rapidly expanding fields of biology essential in chromatid cohesion and normal chromosome
with massive therapeutic potential. DNA methylation is the alignment (Czakai et al. 2011; Dai et al. 2006). Additionally,
most researched epigenetic mechanism that most commonly OTA showed no effect on the activity of Haspin kinase, which
occurs by the transfer of methyl group to the 5th position on mediates H3T3 phosphorylation, suggesting an indirect
Environ Sci Pollut Res

mechanism of action. This mechanism may involve inhibition intracellular miR-29b with decreased miR-29b amounts
of HAT, as it has been shown in nuclear IHKE cell extract that in the cytoplasm. Overall, these findings suggest that
OTA considerably blocked HAT activity in a dose-dependent OTA may facilitate the incidence of fibrotic kidney disor-
fashion as shown in Fig. 2. Furthermore, since HATs also der through post-transcriptional regulations via miR-29b
regulate posttranslational modifications of non-histone pro- (Hennemeier et al. 2014). A study conducted on porcine
teins, OTA blocks completely the lysine acetylation of nuclear renal proximal tubular cells by Stachurska et al. (2013)
and cytosolic proteins in IHKE cells in a dose-dependent fash- demonstrated that OTA-induced increase in the total pool
ion. Similarly, an in vivo study in rats treated with OTA at 0, of cellular microRNAs along with elevated expression of
21, 70, and 210 μg/kg bodyweight for 90 days revealed a miR-132 and miR-200c (Fig. 2). Also, these 2 miRNA spe-
trend towards a decrease of lysine acetylation levels in kidney cies reversed OTA-mediated reduction of HO-1 expression
isolated proteins (Czakai et al. 2011). and restored ROS production and TGFβ expression. The
inhibition of miR-132 by specific antagomir retrieved
OTA and miRNA (non-coding RNA) OTA-mediated reduced Nrf2 expression. A recent study
(Zhu et al. 2016) on OTA-induced hepatotoxicity and
MicroRNAs are a class of non-coding RNAs of nearly 22- miR-122, the most abundant miRNA in the liver, reports
nucleotide long made of hairpin-shaped 70 to 100 base- findings of both in vitro and in vivo studies. miR-122
pair precursors. They are important regulatory elements levels were found to be raised in the OTA-treated HepG2
that serve via targeting mRNAs for cleavage or translation- cells and reduced in the livers of OTA-exposed rats after
al repression (Bartel 2004). The first report on the relation- 4 weeks but increased back after a 13-week period.
ship between OTA toxicity and miRNAs comes from Dai Additionally, the relationship with apoptosis has been
and his co-workers who analyzed miRNA profiling in rat studied. Briefly, miR-122 was found as the primary effec-
kidney treated with OTA (Dai et al. 2014). They have tor of CCNG1/p53 and Bcl-w/caspase-3 pathway in OTA-
identified 394 miRNAs that were differentially expressed induced hepatocytic apoptosis both in vivo and in vitro.
amongst control and OTA-treated groups. Furthermore,
expression levels of Drosha and Dicer, miRNA processing OTA and acetylation
enzymes, which are required for the maturation of
miRNAs, were reduced in the rats’ kidney upon treatment The relationship between OTA-induced toxicity and acetyla-
with OTA. This indicates that miRNA dysregulation might tion has already been discussed related to histone modifica-
be due to renal carcinogenesis and nephrotoxicity of OTA. tions. In addition to that, here we discuss a brief supplement
Additionally, this research has also identified eight novel focused specifically on the HAT, a group of enzymes that
miRNAs. A study by Qi et al. (2014a) first used multi- catalyze lysine acetylation of histone proteins (Fig. 2). A
omics methods in the investigation of OTA-induced early dose-response trend has been demonstrated in OTA-induced
hepatotoxicity. The miRNA profiling revealed enrichment repression of acetyltransferase activity in IHKE cells. This
of the MAPK signaling pathway, metabolic pathways, and suggests that OTAs’ mechanism of carcinogenesis may in-
pathways in cancer after OTA treatment. Together with volve interfering in the mitotic machinery, with HATs as the
mRNA and protein profiling, five of the most common primary cellular targets of OTA (Czakai et al. 2011).
pathways affected by OTA were identified to be cysteine
and methionine metabolism, PPAR signaling, primary bile OTA and RNA interference
acid biosynthesis, arginine and proline metabolism, and
metabolism of xenobiotics by cytochrome P450 (Qi et al. RNA interference (RNAi) is a mechanism of post-
2014a). In an in vitro study in HEK293 cells and HepG2 transcriptional gene silencing in which double-stranded
cells, OTA-treated cell miRNA profiling revealed that the RNA (dsRNA) molecules neutralize the actions of targeted
same changing miRNAs were the ones of signal transduc- mRNAs in a sequence-specific manner. siRNAs are 21- to
tion pathways, while the different changing miRNAs were 23-base-pair-long dsRNA molecules that recognize their ho-
encompassed in human cancer pathways (Zhao et al. mologous RNA targets, and together with associated protein
2017). OTA treatment also resulted in reduced expression partners, they cause mRNA cleavage (Ozcan et al. 2015).
levels of DGCR8, Dicer1, and Drosha enzymes. Thomas et al. (2018) studied the effect of some designed
Hennemeier et al. (2014) reported increased collagen I, siRNAs on the OTA producing Aspergillus nigri to examine
III, and IV protein concentrations with no variation in if the biosynthesis of OTA could be affected by certain tran-
levels of collagen mRNA expression, suggesting post- scriptional factors. For that purpose, in silico studies were
transcriptional regulated mechanisms that could potentially conducted for pks gene promoter sequences and the influence
involve miRNAs in OTA-exposed HEK293 cells. Apart of RNA interference on OTA silencing was evaluated. It has
from that, OTA treatment affected the distribution of been shown that the treatment of fungal protoplasts with
Environ Sci Pollut Res

Fig. 2 Various epigenetic mechanisms of OTA-induced toxicity

synthetically designed siRNAs directed toward pks gene ef- mostly through adding DNA methylation tags at the alleles of
fectively silenced the production of OTA. The designed imprinted genes in the specific imprinting-controlled regions.
siRNA successfully inhibited OTA production, and no effect Such imprinted expression patterns are established in the pa-
on the transfection efficiency has been observed at concentra- rental germ-line and are further maintained in the offspring
tions between 6.3 and 50 nM. On the contrary, this case was throughout somatic development using histone modifications,
different from OTA inhibition where concentration consider- noncoding RNAs, insulators, and higher-order chromatin
ably influenced the inhibitory action of OTA in Aspergillus structure. Therefore, the gender of the parental germ deter-
carbonarius, except for pks_Ia siRNA that showed a consid- mines the expression levels of each allele (Ferguson-Smith
erable effect at the concentration of 25 nM. However, a study 2011; Wood and Oakey 2006). There are not many studies
by Abdel-Hadi et al. (Abdel-Hadi 2011) reported the concen- available on the subject area that draws a connection between
tration of 25 nM as the most effective siRNA to inhibit AflD OTA and imprinting epigenetic mechanisms. Correlations are
and AflR/AflS genes in Aspergillus flavus and Aspergillus usually referred to as genetic predisposition and the possible
parasiticus. In a study by Leite (2013), 10-nM concentration role of genetic imprinting in mycotoxin sensitivity and subse-
was reported as the optimum for silencing the Otapks PV quent nephropathy, as elaborated in the study on a population
gene, while 25 nM was reported for the Tri5 gene. The differ- from the rural environment of West Tunisia that has been
ences between the studies can be explained as the individual exposed to OTA in their diet (Creppy et al. 2005). This geo-
features of single genes, since different target genes may re- graphic area has been considered as the high OTA contami-
quire different optimal siRNA concentrations. To conclude nation region and even reported as an area of endemic OTA-
with, successful inhibition of pks gene by siRNAs gives prom- related nephropathy. Blood and urine tests along with HLA
ise to its use as a target gene in OTA control using RNA fingerprinting have been used as the relevant method for the
interference technology. evaluation of OTA susceptibility. The results have shown
people with HLA haplotype A3, B27/35, DR7 as the most
sensitive to the onset of chronic interstitial nephropathy, under
Imprinting similar or lower OTA exposure with respect to the other sub-
jects that appeared healthy (Fig. 2). Furthermore, two more
Genomic imprinting or silencing refers to epigenetic marks studies by Yang et al. (2017) and Spoendlin et al. (1995) also
that result in the suppression of the transcriptional activity of reported siblings with similar HLA haplotype patterns related
certain genes depending on the parental origin. It is achieved to OTA renal tubulopathy. It is revealed that the role of OTA
Environ Sci Pollut Res

in contributing to the onset of human nephropathy could be silencing has been demonstrated to contribute to the process
associated with specific genetic imprinting in diseased of tumor development.
individuals.

OTA may lead to Alzheimer’s disease


Nucleosome positioning and modification
It has been recognized that OTA toxicity can lead to AD
Eukaryotic chromatin structure has a flexible and dynamic
progression. Moreover, the neurotoxicity of OTA has been
architecture maintained through the process of nucleosome
confirmed by brain examinations (Belmadani et al. 1998a;
remodeling by helicase ATPase enzymes. These modifica-
Sava et al. 2006a). Different levels of OTA in rodents’ brains
tions involve partial disassembly and repeated assembly of
propose that it can actively cross the blood-brain barrier
nucleosomes, the exchange of histones for variants, or the
(BBB) and accumulates inside the brain following a dose of
movement of histone octamers on DNA. In this way, DNA
OTA in mice. Higher level of OTA was observed in the cer-
sequences are either left accessible to interacting proteins
ebellum, pons, and cerebral cortex regions of the brain
or packed into tightly folded structures (Becker and
(Belmadani et al. 1998b; Sava et al. 2006a, 2006b) as well
Workman 2013). There are very limited numbers of stud-
as in the ventral mesencephalon, striatum, and hippocampus.
ies available about the correlation between OTA and nu-
Nevertheless, moderately high levels of OTA were present in
cleosome remodeling. One study reported the upregulation
the hippocampus, an essential site of neurodegeneration in
of nucleosome assembly protein, which has a major role in
AD (Belmadani et al. 1998b). AD is a neurodegenerative dis-
unpacking chromatin for DNA replication or repair. The
ease that has features indicating its progression such as hered-
observed upregulation could be explained because of the
itary predisposition, gender, hypertension, head injury, and
repair processes subsequent to OTA-induced DNA dam-
chemical exposures which are manifested by changes in the
age (Lühe et al. 2003). Another study reported OTA-
brain regions like entorhinal zone and neocortex and by ex-
induced dysregulation of numerous pathways such as nu-
plicit arrangements of subcortical cores and amygdale hippo-
cleosome regulation in rat renal cortex model (Jennings
campus. Various research works showed the p38 mitogen-
et al. 2012).
initiated protein kinase (MAPK) is responsible for the pro-
gression of AD and inhibition of p38 MAPK has an amelio-
Global or localized epigenetic interaction of OTA rative effect on dementia due to neurodegeneration (Kim and
Choi 2010; Zlokovic 2011). Different investigations discover
There is not much data available on differences between glob- the crucial mechanisms of AD which is correlated with p38
al and localized epigenetic patterns related to OTA. However, MAPK actions, for example, synaptic plasticity,
the results of numerous recent studies have revealed that the excitotoxicity, and tau phosphorylation (Munoz and Ammit
carcinogenic effects induced by OTA do not follow a classical 2010; Wang et al. 2014).
carcinogenesis model but involve a complex network of cel- Brain-derived neurotrophic factor (BDNF) abnormal ex-
lular alterations including epigenetic modifications. It might pression in the brain leads to various disorders such as neuro-
be speculated that OTA exhibits effects on global epigenetic psychiatric and neurodegenerative disorders (Berton et al.
patterns via the RAS gene (Ozden et al. 2015; Wu and 2006). A number of researches have revealed shreds of evi-
Brenner 2014; Gazin et al. 2007; Wajapeyee et al. 2013; dence on neurological disorders such as AD, Huntington’s
Serra et al. 2014). disease, Parkinson’s disease (PD), and schizophrenia brought
about by diminishing in the expression of BDNF which leads
Gene silencing to oxidative stress and apoptotic assault (Murer et al. 2001;
Caccamo et al. 2010). Thus, the effect of OTA on BDNF
Gene silencing refers to epigenetic regulation of the genes to expression revealed that treatment with OTA diminished ex-
prevent its expression, either at the transcriptional or at the pression of BDNF in a dose-dependent way. Likewise, OTAs’
translational level. It is achieved through the aforementioned impact on TH expression was additionally examined and it
mechanisms of DNA methylation, histone modification, and was also considerably reduced in a dose-dependent way.
RNAi (Nephew and Huang 2003). A study by Marin-Kuan The mentioned outcomes reinforce prior research results,
et al. (2007) evaluated the influences of OTA on histone which have also reported treating with OTA ameliorates TH
deacetylases (HDACs), a group of enzymes involved in gene immunoreactivity in the fibers of the striatum, corpus striatum,
silencing. With regard to OTA nephron-carcinogenicity, an and substantia nigra by upregulating the oxidative stress and
increased HDAC enzymatic activity and elevated HDAC3 lipid peroxidation with temporary limitation of oxidative
protein expression have been observed (Fig. 2). These find- DNA repair activity. As a result, a decrease in the level of
ings were particularly relevant since HDAC-induced gene striatal dopamine and its metabolites occurs (Torack and
Environ Sci Pollut Res

Morris 1992; Kastner et al. 1993). Studies have examined at alterations, different adsorbents (yeast-based products, OTA,
all stages in AD the expression of p38-MAPK, JNKs, and T-2 binders, modified zeolite, esterified glucomannans, mixed
ERKs and found that their expression causes the progression organic and inorganic compounds with enzymes, and Bacillus
of neurodegenerative disorders (Liu et al. 2007; Wang et al. amyloliquefaciens ASAG1) are used to protect the gastroin-
2014). testinal tract from the absorbing the OTA. However, long-
After OTA administration, caspase-3 and 9 were activated. term use of these agents leads to specific and non-specific
Moreover, neurotoxicity due to OTA was compelled by cas- problems (Chang et al. 2015; Garcia et al. 2003; Pfohl-
pase inhibitors. Programmed cell death caused by OTA oc- Leszkowicz et al. 2015; Trailović et al. 2013). It is being
curred due to lost mitochondria membrane potential. In gen- well-established that certain physical conditions and chemical
eral, present information showed neurotoxicity of OTA is and biological agents’ applications lead to reduce and/or pre-
demonstrated even in moderately low doses. OTA initiated vent OTA-induced toxicity in human and animal products as
neurotoxicity which is mediated by apoptosis. OTA might shown in Tables 2 and 3. The use of household meat process
also be utilized for the pathogenesis of neurodegenerative dis- methods such as cooking, frying, roasting, and baking reduces
orders (for example AD and PD) where apoptotic progres- OTA level in meat sausage which depends on the processing
sions are mainly included. In general, current information modality used (Pleadin et al. 2014). In another study, it has
shows that OTA prompts apoptosis in both the SH-SY5Y been explored that cholestyramine has the potential to bind
cells and the essential neurons. Numerous neurodegenerative with OTA in addition to resin and bile acid which prevent
disorders like AD and PD are identified with neuronal cell its absorption, leading to eliminate via feces and reduce the
death and low regenerative capability of the CNS tissue chances of enterohepatic circulation of OTA (Kerkadi et al.
(Bhat et al. 2016; Friedlander 2003). It has been well- 1999; Madhyastha et al. 1992). Utilization of sodium bicar-
established that OTA leads to ERK-1/2 phosphorylation in a bonate (NaHCO3) helps in OTA ionization, inhibiting its gas-
MAPK-dependent way while JNK-1/2 was unchanged which trointestinal absorption and enhancing elimination via urine
caused nephropathy in human proximal tubule-derived cells (Yong et al. 1987). However, long-term use of both cholestyr-
(Schulz et al. 2018). It is of significance to limit the potential amine and NaHCO3 is not recommended. As the side effects
harm brought about by OTA and other mycotoxins, due to a are mainly related to gastrointestinal tract disorders, it is dif-
decrease in their event in the food chain. ficult to use cholestyramine as therapy and increase fat-
soluble vitamins timing. Most of the patients have the capacity
to tolerate pure resin instead of market available resin mixed
Strategies to lessen OTA toxicity with sugar, artificial color, and other additives (Kőszegi and
Poór 2016). Kidney disease patients also suffer from hyper-
European food safety authority has listed more prominent lipidemia, so the use of cholestyramine has a role in lowering
compounds that are harmful to humans as well as for agro- the lipid. It has been reported that the long-term usage of
economic sectors including aflatoxins, fumonisins, trichothe- cyanidin 3-O-β-D-glucoside (C3G) has demonstrated to
cene, zearalenone, and OTA (EFSA 2004; Streit et al. 2012; counter the noxious effect of OTA through possible
Wielogórska et al. 2016). It has been revealed that OTA is dimethylarginine dimethylaminohydrolase/nitric oxide syn-
involved in human kidney disease propagation such as thase pathway (Sorrenti et al. 2012). Studies on sweat have
Balkan endemic nephropathy (BEN) which was diagnosed revealed that the presence of sauna leads to an increase in the
with an elevated level of OTA with focal segmental excretion of OTA (Hope and Hope 2012). The use of L-me-
glomerulosclerosis (FSGS). This study illustrated that patients thionine, phenylalanine, and phenobarbital is suggested to
with a history of water-damaged buildings had high levels of protect against the developmental toxicity and reduce the me-
mold and dietary history, since mycotoxins were observed in tabolism of OTA which cause a reduction in OTA toxicity
the urine and other tissue samples that contribute towards the (Kayoko et al. 1985; Abdel-Wahhab et al. 1999). A number
main illness (Hooper et al. 2009; Hope and Hope 2012). It is of studies revealed that aspartame works for the delivery of
evident that the presence of indoor water damage and a higher phenylalanine by cleavage with the peptide and has an effect
level of OTA would be avoided for further exposure to people on the binding property and transport of toxin (Baudrimont
living in that place. Various epigenetic mechanisms of OTA et al. 1997; Creppy et al. 1996, 1998; Creppy et al. 1980;
have a vital role in the progression of oxidative stress, Kayoko et al. 1985). A number of researches indicated that
genotoxicity, carcinogenicity, and nephrotoxicity. clay and zeolite both have the efficacy to bind with mycotoxin
Sometimes higher level of OTA leads to kidney chronic dis- in animals and they are also useful in human disorders caused
ease which should be kept in mind as during kidney transplan- by OTA (Piva and Galvano 1999; Whitlow 2006). The use of
tation as it persists at the time of transplantation (Chen and Wu licorice and melatonin is also safe to apply in cases of ochra-
2017; Pfohl-Leszkowicz and Manderville 2007; Tao et al. toxin toxicity (Abdel-Wahhab et al. 2005; Malekinejad et al.
2018). For the therapeutic approach to reverse OTA 2011; Hope and Hope 2012). The use of superoxide dismutase
Environ Sci Pollut Res

Table 2 OTA reduction via physical conditions and chemical agents

Origin of OTA Conditions Decline in OTA content (%) References

Physical parameters
Inoculation 180 °C, 10 min 31.1 (Nehad et al. 2005)
Inoculation 175–204 °C, 7–9 min > 90 (Romani et al. 2003)
Inoculation 200–220 °C, 10–15 min 22.5–93.9 (Barcelo and Barcelo 2018)
Inoculation 180–240 °C, 5–12 min 8–98 (Ferraz et al. 2010)
Natural Dark to light 69–96 (Van der Stegen et al. 2001)
Natural 200 °C, 3 min 65–100 (La Pera et al. 2008)
Natural Industrial roasting 66.5 (Perez de Obanos et al. 2005)
Chemical agents
Natural Alkaline hydrogen peroxide and calcium hydroxide – (Karlovsky et al. 2016)
Natural Sodium hydroxide and calcium hydroxide – (Milani and Maleki 2014)
Natural Monomethylamine and calcium hydroxide – (Scott 1996)
Natural Ammonium and calcium hydroxide – (Karlovsky et al. 2016)
Natural Ethyl acetate and 2% formic acid 80 (Boone 2017)
Natural Dichloromethane and 2% formic acid 80 (Bortoli and Fabian 2001)
Natural Methylene chloride and 2% formic acid 80 (Bortoli and Fabian 2001)
Natural Alkaline treatment 98 (Amezqueta et al. 2008)
Natural Ozonization (O3), 10 wt.% and 15 s – (Scott 1996)
Inoculation Formic acid, 0.25–1% – (Varga et al. 2010)
Inoculation Propionic acid, 0.25–1% – (Varga et al. 2010)
Inoculation Sorbic acid, 0.25–1% – (Varga et al. 2010)
Inoculation Sodium hypochlorite, 0.25–1% – (Scott 1996)
Inoculation Alkaline hydrogen peroxide, 0.05–0.1% – (Aiko and Mehta 2015)

and catalase enzymes and/or the support of glutathione system rate (Costa et al. 2007). Rosmarinic acid and gallic acid have
by NAC are likely to be potential agents which possess anti- the ability to decrease ROS production, protein, DNA synthe-
oxidants and detoxification effects (Cui et al. 2013; Liu et al. sis, and cell death caused by OTA and aflatoxin B1 (Renzulli
2012; Pfohl-Leszkowicz et al. 2002; Yang et al. 2014b). et al. 2004). Cyanidin-3-0-β-glucopyranoside, which is a nat-
Some of the in vitro and in vivo models illustrated that ural free-radical scavenger, lead to a reduction in DNA frag-
vitamins A, C, and E are also successful candidates for reduc- mentation and inhibition of caspase-3 activation and DNA and
ing OTA-induced cytotoxicity and genotoxicity (Baldi et al. protein synthesis caused by OTA in a human hepatoma cell
2004; Fusi et al. 2010; Fusi et al. 2008; Grosse et al. 1997; line and human colonic adenocarcinoma cell line (Di
Hoehler and Marquardt 1996; Hundhausen et al. 2005; Giacomo et al. 2007; Guerra et al. 2005; Russo et al. 2005).
Kumari and Sinha 1994). The use of flavonoids and polyphe- Ellagic acid and silybin have shown anti-apoptotic, anti-oxi-
nols has a positive effect on OTA-exposed cells. These com- dant, anti-diabetic, and antimicrobial activities in the isolated
pounds reverse the toxic effects through the presence of anti- pancreatic islets. The results showed that dietary polyphenols
oxidant activities that work as superoxide anion scavengers significantly inhibit LPO and DNA protective effect in islet
(Bakr Abdu 2011; Hundhausen et al. 2005; Poór et al. 2012; cells exposed to free radical due to oxidative stress. The anti-
Sorrenti et al. 2013). Utilizing drugs such as piroxicam which apoptotic effect of ellagic acid and silybin is related to Bcl2
is a nonsteroidal anti-inflammatory drug (NSAID) specially independent mechanisms. They both have also a hepatopro-
used for arthritis has also a role in binding with plasma pro- tective and anti-apoptotic role in the OTA toxicity. Also, lactic
teins that prevent OTA binding and transport to target organs; acid–producing bacteria degrade OTA and reduce its cytotox-
however, large doses might lead to nephrotoxicity ic effects (Essid and Petzinger 2011; Luz et al. 2018;
(Baudrimont et al. 1995). Two important catechins— Sepúlveda et al. 2011). Flavonoid such as diosmetin increase
epigallocatechin gallate (EGCG) and epicatechin gallate ATP levels in kidney cells and relieve ATP depletion–induced
(ECG)—have the potential role in inhibiting ROS and DNA OTA toxicity. Polyphenols such as luteolin, chlorogenic acid,
fragmentation along with the enhancement of proliferation and caffeine have shown a protective role through the
Environ Sci Pollut Res

Table 3 OTA reduction via biological agents

Biological agents used to reduce OTA References

Carboxypeptidase A (Hu et al. 2018; Luz et al. 2018)


Commercial proteases (pancreatin from porcine pancreas, protease A, and Prolyve PAC from (Abrunhosa et al. 2006)
Aspergillus niger)
Commercial hydrolases (Amano A, crude lipase preparation from Aspergillus niger) (Zhang et al. 2017)
Aspergillus niger hydrolytic metalloenzyme (Zhang et al. 2017)
Rumen microbes (Cho et al. 2016)
Bacillus subtilis, Bacillus licheniformis (Böhm et al. 2000; Petchkongkaew et al. 2008)
Nocardia corynebacterioides, Rhodococcus erythropolis, Mycobacterium sp. (Holzapfel et al. 2002)
Lactobacillus sp. (Fuchs et al. 2008; Piotrowska and Żakowska
2000)
Eubacterium callenderi, E. ramulus, Streptococcus pleomorphus, Lactobacillus vitullinus, (Schatzmayr et al. 2002; Schatzmayr et al. 2006)
Sphingomonas paucimobilis, S. saccharolytica, Stenotrophomonas nitritreducens, Ralstonia
eutropha, R. basilensis, Ochrobactrum sp., Agrobacterium sp.
Pseudomonas cepacia, Pseudomonas putida, Rhodococcus erythropolis, Agrobacterium (Schatzmayr et al. 2012)
tumefaciens, Comamonas acidovorans
Protozoa (Gallo et al. 2015; Özpinar et al. 2002)
Aspergillus niger, Aspergillus fumigatus (Cho et al. 2016)
Aspergillus niger, Aspergillus versicolor, Aspergillus wentii, Aspergillus ochraceus (De Bellis et al. 2015; Zhang et al. 2016)
Aspergillus niger, Aspergillus japonicus (Bejaoui et al. 2006)
Pleurotus ostreatus (Chen et al. 2018; Engelhardt 2002)
Saccharomyces cerevisiae (Piotrowska and Żakowska 2000)
Saccharomyces cerevisiae, Saccharomyces bayanus (Bizaj et al. 2016)
Rhizopus stolonifer, R. microsporus, R. homothallicus, R. oryzae (Varga et al. 2005)
Trichosporon mycotoxinivorans (Molnar et al. 2004)
Phaffia rhodozyma, Xanthophyllomyces dendrorhous (Péteri et al. 2007)
Saccharomyces cerevisiae, Kloeckera apiculata (Angioni et al. 2007)
Aureobasidium pullulans (De Felice et al. 2008)
Cryptococcus flavus, Cryptococcus laurentii, Cryptococcus curvatus, Cryptococcus humicolus, (Peromingo et al. 2018; Schatzmayr et al. 2012;
Trichosporon ovoides, Trichosporon dulcitum, Trichosporon guehoae, Trichosporon mucoides, Schatzmayr et al. 2003)
Trichosporon coremiiforme, Trichosporon cutaneum, Trichosporon laibachii, Trichosporon
moniliforme, Rhodotorula mucilaginosa, Rhodotorula fujisanensis

amelioration of OTA-induced DNA toxicity of the brain and physiological activities. Both minerals alleviate OTA-
blood cells of BALB/c mice. However, chlorogenic acid had mediated DNA toxicity by improving the expression of sele-
shown the best protective effect (Cariddi et al. 2015; Poór et al. nium involved in enzymatic activity (Gan et al. 2015; Zheng
2014). The most vital role of quercetin is the modulation of et al. 2013). The presence of all data regarding the therapeutic
Nrf2 signaling cascade which inhibits OTA-mediated apopto- remedies for OTA genotoxicity, carcinogenicity, oxidative
sis, DNA fragmentation, inhibition of caspase signals activa- stress, apoptosis, and nephrotoxicity in human cell lines and
tion, and micronucleus formation (Abdel-Wahhab et al. 2017; animal models illustrated that the above-mentioned agents
Abdel-Wahhab et al. 2016; De Oliveira et al. 2016; Nabavi would be useful. So, all possible short- and long-term OTA
et al. 2016; Ramyaa and Padma 2013; Ramyaa and Padma toxicity can be prevented by the use of such agents in the near
2014). Quercetin also downregulates NF-κB and COX-2. future, to reduce and eliminate OTA from target cells or
The use of carotenoid lycopene decreases OTA-mediated ap- bodies.
optosis, DNA damage, and oxidative stress. Several studies
showed that certain plants have beneficial effects on OTA
cytotoxicity and genotoxicity (Abdel-Wahhab et al. 2008; Conclusion
Aydin et al. 2013; Chakraborty and Verma 2010; Palabiyik
et al. 2013). The use of zinc and selenium supplementation It is concluded that OTA has several genotoxic and epigenetic
has a vital role in most of the cofactors and enzymes for normal mechanisms leading to cellular and molecular aberrations. It
Environ Sci Pollut Res

causes oxidative stress, DNA damage, carcinogenicity, and Abdu, S.B., 2011. The protective role of ajwa date against the hepatotox-
icity induced by ochratoxin A Egyp J Natural Toxins 8
nephrotoxicity involving different mechanisms. These mech-
Abrunhosa L, Santos L, Venâncio A (2006) Degradation of ochratoxin A
anisms (p38 MAPK, JNKs, and ERKs dysfunctions, BDNF by proteases and by a crude enzyme of Aspergillus niger. Food
disruption, TH overexpression, caspase-3 and -9 activation, Biotechnol 20:231–242
and ERK-1/2 phosphorylation) of OTA also lead to AD pro- Adler M, Müller K, Rached E, Dekant W, Mally A (2009) Modulation of
gression. On the other hand, the promising objective of this key regulators of mitosis linked to chromosomal instability is an
early event in ochratoxin A carcinogenicity. Carcinogenesis 30(4):
review is to treat and disrupt the normal toxicokinetics of OTA 711–719
such as the prevention of cellular uptake and increasing its Aiko V, Mehta A (2015) Occurrence, detection and detoxification of
elimination from the body. So, the above-mentioned natural, mycotoxins. J Biosci 40:943–954
synthetic, and semisynthetic agents shown in Table 2 are ca- Alberts, B., Johnson, A., Lewis, J., Raff, M., Roberts, K. and Walter, P.,
2002. Chromosomal DNA and its packaging in the chromatin fiber.
pable to reverse and prevent OTA-induced toxicity via differ-
In Molecular Biology of the Cell. 4th edition. Garland Science
ent mechanisms. The use of mentioned phytochemicals, Alshannaq A, Yu JH (2017) Occurrence, toxicity, and analysis of major
drugs, and trace elements are recommended to attenuate mycotoxins in food. Int J Environ Res Public Health 14(6):632
OTA-mediated effects; however, still, it is the need of this Alvarez-Erviti L, Leache C, Gonzalez-Penas E, De Cerain AL (2005)
new era to run in vivo and in vitro studies to understand the Alterations induced in vitro by ochratoxin A in rat lymphoid cells.
Hum Exp Toxicol 24(9):459–466
exact mechanisms in order to know how these physical con- Amezqueta S, Gonzalez-Penas E, Lizarraga T, Murillo-Arbizu M, De
ditions, chemical and biologically active enzymes, microbes, Cerain AL (2008) A simple chemical method reduces ochratoxin
and/or proteins decline OTA-induced toxicity. A in contaminated cocoa shells. J Food Prot 71:1422–1426
Angioni A, Caboni P, Garau A, Farris A, Orro D, Budroni M, Cabras P
Acknowledgments The authors wish to thank and acknowledge their (2007) In vitro interaction between ochratoxin A and different
respective universities and institutes. strains of Saccharomyces cerevisiae and Kloeckera apiculata. J
Agric Food Chem 55:2043–2048
Arbillaga L, Azqueta A, Ezpeleta O, Cerain AL (2007a) Oxidative DNA
Authors’ contributions KN gave the idea and prepared the outlines and
damage induced by ochratoxin A in the HK-2 human kidney cell
draft of the manuscript. SA, SZAS, FK, and MB designed the figures
line: evidence of the relationship with cytotoxicity. Mutagenesis.
along with comprehensive tables along with critical editing and reviewing
22(1):35–42
of the whole manuscript. All the authors have read and approved the final
Arbillaga L, Azqueta A, Van Delft JH, De Cerain AL (2007b) In vitro
version.
gene expression data supporting a DNA non-reactive genotoxic
mechanism for ochratoxin A. Toxicol Appl Pharmacol 220(2):
Compliance with ethical standards 216–224
Arrúa AA, Mendes JM, Arrúa P, Ferreira FP, Caballero G, Cazal C, Kohli
Conflict of interest The authors declare that they have no conflict of MM, Peralta I, Ulke G, Fernández Ríos D (2019) Occurrence of
interest. deoxynivalenol and ochratoxin A in beers and wines commercial-
ized in Paraguay. Toxins 11(6):308
Asrani, R., Patial, V., Thakur, M., 2016. Ochratoxin A: possible mecha-
nisms of toxicity. Biosynt Detec Toxic 57
References Atroshi F, Biese I, Saloniemi H, Ali-Vehmas T, Saari S, Rizzo A,
Veijalainen P (2000) Significance of apoptosis and its relationship
Abdel-Hadi, A., 2011. Molecular ecology of Aspergillus section flavi to antioxidants after ochratoxin A administration in mice. J Pharm
species: approaches to understand the role of aflatoxin genes in Pharm Sci 3(3):281–291
aflatoxin biosynthesis Aydin, S., Palabiyik, Ş.S., Erkekoglu., P., Sahin, G., Başaran, N., Giray,
Abdel-Wahhab MA, Nada SA, Arbid MS (1999) Ochratoxicosis: preven- B.K., 2013. The carotenoid lycopene protects rats against DNA
tion of developmental toxicity by L-methionine in the rats. J Appl damage induced by Ochratoxin A. Toxicon 73, 96–103
Toxicol 19:7–12 Baldi A, Losio M, Cheli F et al (2004) Evaluation of the protective effects
Abdel-Wahhab MA, Abdel-Galil MM, El-Lithey M (2005) Melatonin of α-tocopherol and retinol against ochratoxin A cytotoxicity. Br J
counteracts oxidative stress in rats fed an ochratoxin A contaminated Nutr 91(4):507–512
diet. J Pineal Res 38(2):130–135 Bannister, A.J. and Kouzarides, T., 2011. Regulation of chromatin by
Abdel-Wahhab MA, Abdel-Azim SH, El-Nekeety AA (2008) Inula histone modifications. Cell Res. 21(3), p.381
crithmoides extract protects against ochratoxin A-induced oxidative Barcelo JM, Barcelo RC (2018) Post-harvest practices linked with och-
stress, clastogenic and mutagenic alterations in male rats. Toxicon ratoxin A contamination of coffee in three provinces of Cordillera
52(4):566–573 Administrative Region, Philippines. Food Addit Contam Part A 35:
Abdel-Wahhab MA, Aljawish A, Kenawy AM, Hamed HS, El-Nekeety 328–340
AA, Abdel-Aziem SH (2016) Grafting of gallic acid onto chitosan Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and
enhances antioxidant activities in vitro and protects against ochra- function. Cell 116(2):281–297
toxin A toxicity in catfish (Clarias gariepinus). Environ Toxicol Baudrimont I, Betbeder AM, Gharbi A, Pfohl-Leszkowicz A, Dirheimer
Pharmacol 41:279–288 G, Creppy E (1994) Effect of superoxide dismutase and catalase on
Abdel-Wahhab MA, Aljawish A, El-Nekeety AA, Abdel-Aziem SH, the nephrotoxicity induced by subchronical administration of och-
Hassan NS (2017) Chitosan nanoparticles plus quercetin suppress ratoxin A in rats. Toxicol. 89(2):101–111
the oxidative stress, modulate DNA fragmentation and gene expres- Baudrimont I, Murn M, Betbeder A, Guilcher J, Creppy E (1995) Effect
sion in the kidney of rats fed ochratoxin A-contaminated diet. Food of piroxicam on the nephrotoxicity induced by ochratoxin A in rats.
Chem Toxicol 99:209–221 Toxicol. 95(1–3):147–154
Environ Sci Pollut Res

Baudrimont I, Betbeder A, Creppy E (1997) Reduction of the ochratoxin Chen W, Li C, Zhang B, Zhou Z, Shen Y, Liao X, Yang J, Wang Y, Li X,
A-induced cytotoxicity in Vero cells by aspartame. Arch Toxicol Li Y (2018a) Advances in biodetoxification of ochratoxin AA re-
71(5):290–298 view of the past five decades. Front Microbiol 9
Becker PB, Workman JL (2013) Nucleosome remodeling and epige- Chen L, Gao Y, Zhu L, Song H, Zhao L, Liu A, Zhang G, Shi G (2018b)
netics. Cold Spring Harb Perspect Biol 5(9):a017905 Establishment and characterization of a GES-1 human gastric epi-
Bejaoui H, Mathieu F, Taillandier P, Lebrihi A (2006) Biodegradation of thelial cell line stably expressing miR-23a. Oncol Lett 16(1):977–
ochratoxin A by Aspergillus section Nigri species isolated from 983
French grapes: a potential means of ochratoxin A decontamination Cho SM, Jeong SE, Lee KR, Sudhani HP, Kim M, Hong S-Y, Chung SH
in grape juices and musts. FEMS Microbiol Lett 255:203–208 (2016) Biodegradation of ochratoxin A by Aspergillus tubingensis
Belmadani A, Tramu G, Betbeder AM, Creppy EE (1998a) Subchronic isolated from Meju. J Microbiol Biotechnol 26:1687–1695
effects of ochratoxin A on young adult rat brain and partial preven- Costa S, Utan A, Cervellati R, Speroni E, Guerra M (2007) Catechins:
tion by aspartame, a sweetener. Hum Exp Toxicol 17:380–386. natural free-radical scavengers against ochratoxin A-induced cell
https://doi.org/10.1177/096032719801700704 damage in a pig kidney cell line (LLC-PK1). Food Chem Toxicol
Belmadani A, Tramu G, Betbeder AM, Steyn PS, Creppy EE (1998b) 45(10):1910–1917
Regional selectivity to ochratoxin A, distribution and cytotoxicity in Costa JG, Saraiva N, Guerreiro PS et al (2016) Ochratoxin A-induced
rat brain. Arch Toxicol 72:656–662. https://doi.org/10.1007/ cytotoxicity, genotoxicity and reactive oxygen species in kidney
s002040050557 cells: an integrative approach of complementary endpoints. Food
Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ Chem Toxicol 87:65–76
et al (2006) Essential role of BDNF in the mesolimbic dopamine Creppy EE, Schlegel M, Röschenthaler R, Dirheimer G (1980)
pathway in social defeat stress. Science 311:864–868. https://doi. Phenylalanine prevents acute poisoning by ochratoxin-a in mice.
org/10.1126/science.1120972 Toxicol Lett 6(2):77–80
Bhasin M, Reinherz EL, Reche PA (2006) Recognition and classification Creppy EE, Röschenthaler R, Dirheimer G (1984) Inhibition of protein
of histones using support vector machine. J Comput Biol 13(1):102– synthesis in mice by ochratoxin A and its prevention by phenylala-
112 nine. Food Chem Toxicol 22(11):883–886. https://doi.org/10.1016/
Bhat PV, Pandareesh MD, Khanum F, Tamatam A (2016) Cytotoxic 0278-6915(84)90170-4
effects of ochratoxin A in neuro-2a cells: role of oxidative stress Creppy E, Baudrimont I, Belmadani A, Betbeder A (1996) Aspartame as
evidenced by N-acetylcysteine. Front Microbiol 7:1142 a preventive agent of chronic toxic effects of ochratoxin A in exper-
Bizaj E, Mavri J, Čuš F, Raspor A (2016) Removal of ochratoxin A in imental animals. J Toxicol Toxin Rev 15(3):207–221
Saccharomyces cerevisiae liquid cultures. South African J Enol Creppy EE, Baudrimont I, Marie A (1998) How aspartame prevents the
Viticulture 30:151–155 toxicity of ochratoxin A. J Toxicol Sci 23:165–172
Boesch-Saadatmandi C, Loboda A, Jozkowicz A et al (2008) Effect of Creppy EE, Moukha S, Bacha H, Carratu MR (2005) How much should
ochratoxin A on redox-regulated transcription factors, antioxidant we involve genetic and environmental factors in the risk assessment
enzymes and glutathione-S-transferase in cultured kidney tubulus of mycotoxins in humans? Int J Environ Res Public Health 2(1):
cells. Food Chem Toxicol 46(8):2665–2671 186–193
Boesch-Saadatmandi C, Wagner A, Graeser A, Hundhausen C, Wolffram Cui J, Xing L, Li Z, Wu S, Wang J, Liu J, Wang J, Yan X, Zhang X
S, Rimbach G (2009) Ochratoxin A impairs Nrf2-dependent gene (2010) Ochratoxin A induces G2 phase arrest in human gastric ep-
expression in porcine kidney tubulus cells. J Anim Physiol Anim ithelium GES-1 cells in vitro. Toxicol Lett 193(2):152–158
Nutr 93(5):547–554 Cui J, Liu J, Wu S et al (2013) Oxidative DNA damage is involved in
Böhm J, Grajewski J, Asperger H, Cecon B, Rabus B, Razzazi E (2000) ochratoxin A-induced G2 arrest through ataxia telangiectasia-
Study on biodegradation of some A-and B-trichothecenes and och- mutated (ATM) pathways in human gastric epithelium GES-1 cells
ratoxin A by use of probiotic microorganisms. Mycotoxin Res 16:70 in vitro. Arch Toxicol 87(10):1829–1840
Boone, S., 2017. Improved coffee cherry processing. Google Patents Czakai K, Müller K, Mosesso P et al (2011) Perturbation of mitosis
Bortoli, G., Fabian, M., 2001. A process to remove mycotoxins from through inhibition of histone acetyltransferases: the key to ochratox-
green coffee, ASIC Congress, Trieste in a toxicity and carcinogenicity? Toxicol Sci 122(2):317–329
Caccamo A, Maldonado MA, Bokov AF, Majumder S, Oddo S (2010) Dai J, Sullivan BA, Higgins JM (2006) Regulation of mitotic chromo-
CBP gene transfer increases BDNF levels and ameliorates learning some cohesion by Haspin and Aurora B. Dev Cell 11(5):741–750
and memory deficits in a mouse model of Alzheimer’s disease. Proc Dai Q, Zhao J, Qi X et al (2014) MicroRNA profiling of rats with och-
Natl Acad Sci U S A 107:22687–22692. https://doi.org/10.1073/ ratoxin A nephrotoxicity. BMC Genomics 15(1):333
pnas.1012851108 Darif Y, Mountassif D, Belkebir A et al (2016) Ochratoxin A mediates
Cariddi LN, Sabini MC, Escobar FM et al (2015) Polyphenols as possible MAPK activation, modulates IL-2 and TNF-α mRNA expression
bioprotectors against cytotoxicity and DNA damage induced by and induces apoptosis by mitochondria-dependent and
ochratoxin A. Environ Toxicol Pharmacol 39(3):1008–1018 mitochondria-independent pathways in human H9 T cells. J
Cavin C, Delatour T, Marin-Kuan M et al (2006) Reduction in antioxi- Toxicol Sci 41(3):403–416
dant defenses may contribute to ochratoxin A toxicity and carcino- De Bellis P, Tristezza M, Haidukowski M, Fanelli F, Sisto A, Mulè G,
genicity. Toxicol Sci 96(1):30–39 Grieco F (2015) Biodegradation of ochratoxin A by bacterial strains
Chakraborty D, Verma R (2010) Ameliorative effect of Emblica isolated from vineyard soils. Toxins 7:5079–5093
officinalis aqueous extract on ochratoxin-induced lipid peroxidation De Felice D, Solfrizzo M, De Curtis F, Lima G, Visconti A, Castoria R
in the kidney and liver of mice. International J. Occup Med Environ (2008) Strains of Aureobasidium pullulans can lower ochratoxin A
Health 23(1):63–73 contamination in wine grapes. Phytopathol. 98:1261–1270
Chang X, Wu Z, Wu S, Dai Y, Sun C (2015) Degradation of ochratoxin A De Oliveira MR, Nabavi SM, Braidy N, Setzer WN, Ahmed T, Nabavi
by Bacillus amyloliquefaciens ASAG1. Food Add Contam Part A SF (2016) Quercetin and the mitochondria: a mechanistic view.
32(4):564–571 Biotechnol Adv 34(5):532–549
Chen C, Wu F (2017) The need to revisit ochratoxin A risk in light of Di Giacomo C, Acquaviva R, Piva A et al (2007) Protective effect of
diabetes, obesity, and chronic kidney disease prevalence. Food cyanidin 3-O-β-d-glucoside on ochratoxin A-mediated damage in
Chem Toxicol 103:79–85 the rat. Br J Nutr 98(5):937–943
Environ Sci Pollut Res

Domijan AM, Želježić D, Kopjar N, Peraica M (2006) Standard and Fpg- Gan F, Zhou Y, Hou L, Qian G, Chen X, Huang K (2017) Ochratoxin A
modified comet assay in kidney cells of ochratoxin A-and fumonisin induces nephrotoxicity and immunotoxicity through different
B 1-treated rats. Toxicol. 222(1):53–59 MAPK signaling pathways in PK15 cells and porcine primary
Domijan AM, Peraica M, Vrdoljak AL, Radić B, Žlender V, Fuchs R splenocytes. Chemosphere 182:630–637
(2007) The involvement of oxidative stress in ochratoxin A and Gan F, Zhou Y, Qian G et al (2018) PCV2 infection aggravates ochra-
fumonisin B1 toxicity in rats. Mol Nutr Food Res 51(9):1147–1151 toxin A-induced nephrotoxicity via autophagy involving p38 signal-
Dutto I, Tillhon M, Cazzalini O, Stivala LA, Prosperi E (2015) Biology of ing pathway in vivo and in vitro. Environ Pollut 238:656–662
the cell cycle inhibitor p21CDKN1A: molecular mechanisms and Garcia A, Avila E, Rosiles R, Petrone V (2003) Evaluation of two my-
relevance in chemical toxicology. Arch Toxicol 89(2):155–178 cotoxin binders to reduce toxicity of broiler diets containing ochra-
Dwivedi P, Burns RB (1984) Effect of ochratoxin A on immunoglobulins toxin A and T-2 toxin contaminated grain. Avian Dis 47(3):691–699
in broiler chicks. Res Vet Sci 36(1):117–121 Gautier JC, Holzhaeuser D, Markovic J, Gremaud E, Schilter B, Turesky
EFSA (2004) Opinion of the Scientific Panel on the Contaminants in the RJ (2001) Oxidative damage and stress response from ochratoxin A
Food Chain on a request from the Commission related to ochratoxin exposure in rats. Free Radic Biol Med 30(10):1089–1098
A (OTA) as undesirable substance in animal feed. EFSA J 101:1–36 Gayathri L, Dhivya R, Dhanasekaran D, Periasamy VS, Alshatwi AA,
Ehrlich V, Darroudi F, Uhl M et al (2002) Genotoxic effects of ochratoxin Akbarsha MA (2015) Hepatotoxic effect of ochratoxin A and citri-
A in human-derived hepatoma (HepG2) cells. Food Chem Toxicol nin, alone and in combination, and protective effect of vitamin E:
40(8):1085–1090 In vitro study in HepG2 cell. Food Chem Toxicol 83:151–163
El-Nekeety AA, Abdel-Wahhab KG, Abdel-Aziem SH, Mannaa FA, Gazin C, Wajapeyee N, Gobeil S, Virbasius CM, Green MR (2007) An
Hassan NS, Abdel-Wahhab MA (2017) Papaya fruits extracts en- elaborate pathway required for Ras-mediated epigenetic silencing.
hance the antioxidant capacity and modulate the genotoxicity and Nature. 449:1073–1077
oxidative stress in the kidney of rats fed ochratoxin A-contaminated Giromini C, Rebucci R, Saccone F, Fusi E, Baldi A (2015) Cytotoxicity,
diet. J Appl Pharm Sci 7(07):111–121 DNA integrity and methylation in mammary and kidney epithelial
Engelhardt G (2002) Degradation of ochratoxin a and b by the white rot cell lines exposed to Ochratoxin A. Ital J Anim Sci 14(suppl. 1):44–
funguspleurotus ostreatus. Mycotoxin Res 18:37–43 44
Essid E, Petzinger E (2011) Silibinin pretreatment protects against och- Gordon EM, Ravicz JR, Liu S, Chawla SP, Hall FL (2018) Cell cycle
ratoxin A-mediated apoptosis in primary rat hepatocytes. Mycotoxin checkpoint control: the cyclin G1/Mdm2/p53 axis emerges as a
Res 27(3):167–176 strategic target for broad-spectrum cancer gene therapy-A review
Essid E, Dernawi Y, Petzinger E (2012) Apoptosis induction by OTA and of molecular mechanisms for oncologists. Mol Clin Oncol 9(2):
TNF-α in cultured primary rat hepatocytes and prevention by 115–134
silibinin. Toxins 4(11):1139–1156 Grosse Y, Chekir-Ghedira L, Huc A et al (1997) Retinol, ascorbic acid
Ferguson-Smith AC (2011) Genomic imprinting: the emergence of an and α-tocopherol prevent DNA adduct formation in mice treated
epigenetic paradigm. Nat Rev Genet 12(8):565 with the mycotoxins ochratoxin A and zearalenone. Cancer Lett
Ferrante MC, Bilancione M, Raso GM et al (2006) Expression of COX-2 114(1):225–229
and hsp72 in peritoneal macrophages after an acute ochratoxin A Guerra M, Galvano F, Bonsi L et al (2005) Cyanidin-3-O-β-
treatment in mice. Life Sci 79(13):1242–1247 glucopyranoside, a natural free-radical scavenger against aflatoxin
Ferrante M, Raso GM, Bilancione M, Esposito E, Iacono A, Meli R B1-and ochratoxin A-induced cell damage in a human hepatoma
(2008) Differential modification of inflammatory enzymes in cell line (Hep G2) and a human colonic adenocarcinoma cell line
J774A. 1 macrophages by ochratoxin A alone or in combination (CaCo-2). Br J Nutr 94(2):211–220
with lipopolysaccharide. Toxicol Lett 181(1):40–46 Ha TH (2015) Recent advances for the detection of ochratoxin A. Toxins
Ferraz MB, Farah A, Iamanaka BT, Perrone D, Copetti MV, Marques 7(12):5276–5300
VX, Vitali AA, Taniwaki MH (2010) Kinetics of ochratoxin A Hadjeba-Medjdoub K, Tozlovanu M, Pfohl-Leszkowicz A, Frenette C,
destruction during coffee roasting. Food Control 21:872–877 Paugh RJ, Manderville RA (2012) Structure–activity relationships
Friedlander RM (2003) Apoptosis and caspases in neurodegenerative imply different mechanisms of action for Ochratoxin A-mediated
diseases. N Engl J Med 348:1365–1375 cytotoxicity and genotoxicity. Chem Res Toxicol 25(1):181–190
Fuchs S, Sontag G, Stidl R, Ehrlich V, Kundi M, Knasmüller S (2008) Hameed MR, Khan MZ, Saleemi MK et al (2017) Study of ochratoxin A
Detoxification of patulin and ochratoxin A, two abundant myco- (OTA)-induced oxidative stress markers in broiler chicks. Toxin
toxins, by lactic acid bacteria. Food Chem Toxicol 46:1398–1407 Rev 36(4):270–274
Fusi E, Rebucci R, Pecorini C, Rossi L, D’Ambrosio F, Baldi A (2008) Hassen W, Ayed-Boussema I, Bouslimi A, Bacha H (2007) Heat shock
Evaluation of the damage induced by ochratoxin A and the protec- proteins (Hsp 70) response is not systematic to cell stress: case of the
tive role of α-tocopherol in cultured bovine mammary epithelial mycotoxin ochratoxin A. Toxicology. 242(1):63–70
cells. Vet Res Commun 32(1):343–345 Hennemeier I, Humpf HU, Gekle M, Schwerdt G (2014) Role of
Fusi E, Rebucci R, Pecorini C et al (2010) Alpha-tocopherol counteracts microRNA-29b in the ochratoxin A-induced enhanced collagen for-
the cytotoxicity induced by ochratoxin a in primary porcine fibro- mation in human kidney cells. Toxicology. 324:116–122
blasts. Toxins 2(6):1265–1278 Hibi D, Kijima A, Kuroda K et al (2013) Molecular mechanisms under-
Gagliano N, Dalle Donne I, Torri C et al (2006) Early cytotoxic effects of lying ochratoxin A-induced genotoxicity: global gene expression
ochratoxin A in rat liver: a morphological, biochemical and molec- analysis suggests induction of DNA double-strand breaks and cell
ular study. Toxicol. 225(2):214–224 cycle progression. J Toxicol Sci 38(1):57–69
Gallo A, Giuberti G, Frisvad JC, Bertuzzi T, Nielsen KF (2015) Review Hoehler D, Marquardt RR (1996) Influence of vitamins E and C on the
on mycotoxin issues in ruminants: occurrence in forages, effects of toxic effects of ochratoxin A and T-2 toxin in chicks. Poult Sci
mycotoxin ingestion on health status and animal performance and 75(12):1508–1515
practical strategies to counteract their negative effects. Toxins 7: Hoehler D, Marquardt RR, McIntosh AR, Xiao H (1996) Free radical
3057–3111 generation as induced by ochratoxin A and its analogs in bacteria
Gan F, Xue H, Huang Y, Pan C, Huang K (2015) Selenium alleviates (Bacillus brevis). J Biol Chem 271(44):27388–27394
porcine nephrotoxicity of ochratoxin A by improving selenoenzyme Hoehler D, Marquardt RR, McIntosh AR, Hatch GM (1997) Induction of
expression in vitro. PLoS One 10(3):e0119808 free radicals in hepatocytes, mitochondria and microsomes of rats by
Environ Sci Pollut Res

ochratoxin A and its analogs. Biochim Biophys Acta-Mol Cell Res Kumar R, Alam S, Chaudhari BP et al (2012) Ochratoxin A-induced cell
1357(2):225–233 proliferation and tumor promotion in mouse skin by activating the
Holzapfel, W., Brost, I., Farber, P., Geisen, R., Bresch, H., Jany, K., expression of cyclin-D1 and cyclooxygenase-2 through nuclear
Mengu, M., Jakobsen, M., Steyn, P., Teniola, D., 2002. factor-kappa B and activator protein-1. Carcinogenesis 34(3):647–
Actinomycetes for breaking down aflatoxin B1, ochratoxin A, 657
and/or zearalenon. Patent Number WO02/099142 A3 Kumar M, Dwivedi P, Sharma AK, Sankar M, Patil RD, Singh ND
Hooper DG, Bolton VE, Guilford FT, Straus DC (2009) Mycotoxin de- (2014) Apoptosis and lipid peroxidation in ochratoxin A-and citri-
tection in human samples from patients exposed to environmental nin-induced nephrotoxicity in rabbits. Toxicol Ind Health 30(1):90–
molds. Int J Mol Sci 10(4):1465–1475 98
Hope, J.H., Hope, B.E., 2012. A review of the diagnosis and treatment of Kumari D, Sinha S (1994) Effect of retinol on ochratoxin-produced
ochratoxin A inhalational exposure associated with human illness genotoxicity in mice. Food Chem Toxicol 32(5):471–475
and kidney disease including focal segmental glomerulosclerosis J La Pera L, Avellone G, Lo Turco V, Di Bella G, Agozzino P, Dugo G
Environ Public Health 2012 (2008) Influence of roasting and different brewing processes on the
Hou L, Gan F, Zhou X et al (2018) Immunotoxicity of ochratoxin A and ochratoxin A content in coffee determined by high-performance
aflatoxin B1 in combination is associated with the nuclear factor liquid chromatography-fluorescence detection (HPLC-FLD). Food
kappa B signaling pathway in 3D4/21 cells. Chemosphere 199: Addit Contam 25:1257–1263
718–727 Lambert D, Padfield PJ, McLaughlin J, Cannell S, O’Neill CA (2007)
Hu H, Jia X, Wang Y, Liang Z (2018) Removal of ochratoxin A by a Ochratoxin A displaces claudins from detergent resistant membrane
carboxypeptidase and peptides present in liquid cultures of Bacillus microdomains. Biochem Biophys Res Commun 358(2):632–636
subtilis CW14. World Mycotoxin J:1–12 Leite, G.M., 2013. Potential for control of spoilage and mycotoxigenic
Huang FJ, Chan WH (2016) Effects of ochratoxin a on mouse oocyte species using mixtures of anti-oxidants, aliphatic acids and molecu-
maturation and fertilization, and apoptosis during fetal development. lar approaches using RNAi. http://dspace.lib.cranfield.ac.uk/handle/
Environ Toxicol 31(6):724–735 1826/8632
Hundhausen C, Bösch-Saadatmandi C, Augustin K, Blank R, Wolffram Li J, Yin S, Dong Y, Fan L, Hu H (2011) p53 activation inhibits ochra-
S, Rimbach G (2005) Effect of vitamin E and polyphenols on och- toxin A-induced apoptosis in monkey and human kidney epithelial
ratoxin A-induced cytotoxicity in liver (HepG2) cells. J Plant cells via suppression of JNK activation. Biochem Biophys Res
Physiol 162(7):818–822 Commun 411(2):458–463
Jennings P, Weiland C, Limonciel A et al (2012) Transcriptomic alter- Li X, Gao J, Huang K et al (2015) Dynamic changes of global DNA
ations induced by ochratoxin A in rat and human renal proximal methylation and hypermethylation of cell adhesion-related genes
tubular in vitro models and comparison to a rat in vivo model. in rat kidneys in response to ochratoxin A. World Mycotoxin J
Arch Toxicol 86(4):571–589 8(4):465–476
Kamp HG, Eisenbrand G, Janzowski C et al (2005a) Ochratoxin A in- Liang R, Shen XL, Zhang B et al (2015) Apoptosis signal-regulating
duces oxidative DNA damage in liver and kidney after oral dosing to kinase 1 promotes ochratoxin A-induced renal cytotoxicity. Sci
rats. Mol Nutr Food Res 49(12):1160–1167 Report 5:8078
Kamp HG, Eisenbrand G, Schlatter J, Würth K, Janzowski C (2005b) Limonciel A, Jennings P (2014) A review of the evidence that ochratoxin
Ochratoxin A: induction of (oxidative) DNA damage, cytotoxicity A is an Nrf2 inhibitor: implications for nephrotoxicity and renal
and apoptosis in mammalian cell lines and primary cells. carcinogenicity. Toxins 6(1):371–379
Toxicology 206(3):413–425 Liu Q, Zhang J, Zhu H, Qin C, Chen Q, Zhao B (2007) Dissecting the
Karlovsky P, Suman M, Berthiller F, De Meester J, Eisenbrand G, Perrin signaling pathway of nicotine-mediated neuroprotection in a mouse
I, Oswald IP, Speijers G, Chiodini A, Recker T (2016) Impact of Alzheimer disease model. FASEB J 21:61–73. https://doi.org/10.
food processing and detoxification treatments on mycotoxin con- 1096/fj.06-5841com
tamination. Mycotoxin Res 32:179–205 Liu J, Wang Y, Cui J et al (2012) Ochratoxin A induces oxidative DNA
Kastner A, Hirsch EC, Herrero MT, Javoy-Agid F, Agid Y (1993) damage and G1 phase arrest in human peripheral blood mononucle-
Immunocytochemical quantification of tyrosine hydroxylase at a ar cells in vitro. Toxicol Lett 211(2):164–171
cellular level in the mesencephalon of control subjects and patients Loboda A, Stachurska A, Podkalicka P et al (2017) Effect of heme
with Parkinson’s and Alzheimer’s disease. J Neurochem 61:1024– oxygenase-1 on ochratoxin A-induced nephrotoxicity in mice. Int
1034. https://doi.org/10.1111/j.1471-4159.1993.tb03616.x J Biochem Cell Biol 84:46–57
Kayoko M, Shigetoshi S, Tetsuro K, Mikio Y, Masayoshi K (1985) Lühe A, Hildebrand H, Bach U, Dingermann T, Ahr HJ (2003) A new
Reduction of ochratoxin A toxicity in mice treated with phenylala- approach to studying ochratoxin A (OTA)-induced nephrotoxicity:
nine and phenobarbital. Toxicol Lett 25(1):1–5 expression profiling in vivo and in vitro employing cDNA microar-
Kerkadi A, Barriault C, Marquardt RR et al (1999) Cholestyramine pro- rays. Toxicol Sci 73(2):315–328
tection against ochratoxin A toxicity: role of ochratoxin A sorption Luz C, Ferrer J, Mañes J, Meca G (2018) Toxicity reduction of ochratoxin
by the resin and bile acid enterohepatic circulation. J Food Prot A by lactic acid bacteria. Food Chem Toxicol 112:60–66
62(12):1461–1465 Madhyastha M, Frohlich A, Marquardt R (1992) Effect of dietary chole-
Kim EK, Choi EJ (2010) Pathological roles of MAPK signaling pathways styramine on the elimination pattern of ochratoxin A in rats. Food
in human diseases. Biochim Biophys Acta 1802:396–405. https:// Chem Toxicol 30(8):709–714
doi.org/10.1016/j.bbadis.2009.12.009 Malekinejad H, Mirzakhani N, Razi M, Cheraghi H, Alizadeh A,
Knasmuller S, Cavin C, Chakraborty A et al (2004) Structurally related Dardmeh F (2011) Protective effects of melatonin and Glycyrrhiza
mycotoxins ochratoxin A, ochratoxin B, and citrinin differ in their glabra extract on ochratoxin A–induced damages on testes in ma-
genotoxic activities and in their mode of action in human-derived ture rats. Hum Exp Toxicol 30:110–123
liver (HepG2) cells: implications for risk assessment. Nutr Cancer Malir F, Ostry V, Pfohl-Leszkowicz A, Novotna E (2013a) Ochratoxin A:
50(2):190–197 developmental and reproductive toxicity-An overview. Birth
Kőszegi T, Poór M (2016) Ochratoxin A: molecular interactions, mech- Defects Res. B Dev Reprod Toxicol 98(6):493–502
anisms of toxicity and prevention at the molecular level. Toxins Malir F, Ostry V, Novotna E (2013b) Toxicity of the mycotoxin ochra-
8(4):111 toxin A in the light of recent data. Toxin Rev 32(2):19–33
Environ Sci Pollut Res

Malir F, Ostry V, Pfohl-Leszkowicz A, Malir J, Toman J (2016) Palabiyik S, Erkekoglu P, Zeybek N, Kızılgun M, Sahin G, Giray BK
Ochratoxin A: 50 years of research. Toxins 8(7):191 (2012) Ochratoxin A causes oxidative stress and cell death in rat
Mally A (2012) Ochratoxin A and mitotic disruption: mode of action liver. World Mycotoxin J 5(4):377–384
analysis of renal tumor formation by ochratoxin A. Toxicol Sci Palabiyik SS, Erkekoglu P, Zeybek ND et al (2013) Protective effect of
127(2):315–330 lycopene against ochratoxin A induced renal oxidative stress and
Mally A, Pepe G, Ravoori S et al (2005) Ochratoxin A causes DNA apoptosis in rats. Exp Toxicol Pathol 65(6):853–861
damage and cytogenetic effects but no DNA adducts in rats. Chem Palma N, Cinelli S, Sapora O, Wilson SH, Dogliotti E (2007) Ochratoxin
Res Toxicol 18(8):1253–1261 A-induced mutagenesis in mammalian cells is consistent with the
Manderville R, Pfohl-Leszkowicz A (2008) Bioactivation and DNA ad- production of oxidative stress. Chem Res Toxicol 20(7):1031–1037
duction as a rationale for ochratoxin A carcinogenesis. World Paradells S, Rocamonde B, Llinares C et al (2015) Neurotoxic effects of
Mycotoxin J 1(3):357–367 ochratoxin A on the subventricular zone of adult mouse brain. J
Mantle PG, Faucet-Marquis V, Manderville RA, Squillaci B, Pfohl- Appl Toxicol 35(7):737–751
Leszkowicz A (2009) Structures of covalent adducts between Perez De Obanos A, Gonzalez-Penas E, Lopez de Cerain A (2005)
DNA and ochratoxin A: a new factor in debate about genotoxicity Influence of roasting and brew preparation on the ochratoxin A
and human risk assessment. Chem Res Toxicol 23(1):89–98 content in coffee infusion. Food Addit Contam 22:463–471
Marin DE, Taranu I (2015) Ochratoxin A and its effects on immunity. Periasamy R, Kalal IG, Krishnaswamy R, Viswanadha V (2016)
Toxin Rev 34(1):11–20 Quercetin protects human peripheral blood mononuclear cells from
Marin DE, Braicu C, Gras MA et al (2017a) Low level of ochratoxin A OTA-induced oxidative stress, genotoxicity, and inflammation.
affects genome-wide expression in kidney of pig. Toxicon 136:67– Environ Toxicol 31(7):855–865
77 Peromingo B, Núñez F, Rodríguez A, Alía A, Andrade MJ (2018)
Marin DE, Pistol GC, Gras MA, Palade ML, Taranu I (2017b) Potential of yeasts isolated from dry-cured ham to control ochratoxin
Comparative effect of ochratoxin A on inflammation and oxidative A production in meat models. Int J Food Microbiol 268:73–80
stress parameters in gut and kidney of piglets. Regul Toxicol Perry JL, Goldsmith MR, Peterson MA et al (2004) Structure of the
Pharmacol 89:224–231 ochratoxin A binding site within human serum albumin. J Phys
Marin-Kuan M, Nestler S, Verguet C et al (2005) A toxicogenomics Chem B 108(43):16960–16964
approach to identify new plausible epigenetic mechanisms of och- Petchkongkaew A, Taillandier P, Gasaluck P, Lebrihi A (2008) Isolation
ratoxin a carcinogenicity in rat. Toxicol Sci 89(1):120–134 of Bacillus spp. from Thai fermented soybean (Thua-nao): screening
Marin-Kuan M, Nestler S, Verguet C et al (2007) MAPK-ERK activation for aflatoxin B1 and ochratoxin A detoxification. J Appl Microbiol
in kidney of male rats chronically fed ochratoxin A at a dose causing 104:1495–1502
a significant incidence of renal carcinoma. Toxicol Appl Pharmacol Péteri Z, Téren J, Vágvölgyi C, Varga J (2007) Ochratoxin degradation
224(2):174–181 and adsorption caused by astaxanthin-producing yeasts. Food
Marin-Kuan M, Ehrlich V, Delatour T, Cavin C, Schilter B (2011) Microbiol 24:205–210
Evidence for a role of oxidative stress in the carcinogenicity of Petrik J, Žanić-Grubišić T, Barišić K et al (2003) Apoptosis and oxidative
ochratoxin A. J Toxicol 645361 stress induced by ochratoxin A in rat kidney. Arch Toxicol 77(12):
Milani J, Maleki G (2014) Effects of processing on mycotoxin stability in 685–693
cereals. J Sci Food Agric 94:2372–2375 Pfohl-Leszkowicz A, Manderville RA (2007) Ochratoxin A: an overview
Molnar O, Schatzmayr G, Fuchs E, Prillinger H (2004) Trichosporon on toxicity and carcinogenicity in animals and humans. Mol Nutr
mycotoxinivorans sp. nov., a new yeast species useful in biological Food Res 51(1):61–99
detoxification of various mycotoxins. Syst Appl Microbiol 27:661– Pfohl-Leszkowicz A, Manderville RA (2011) An update on direct
671 genotoxicity as a molecular mechanism of ochratoxin a carcinoge-
Munoz L, Ammit AJ (2010) Targeting p38 MAPK pathway for the treat- nicity. Chem Res Toxicol 25(2):252–262
ment of Alzheimer’s disease. Neuropharmacology 58:561–568. Pfohl-Leszkowicz A, Bartsch H, Azémar B, Mohr U, Estève J,
https://doi.org/10.1016/j.neuropharm.2009 Castegnaro M (2002) MESNA protects rats against nephrotoxicity
Murer MG, Yan Q, Raisman-Vozari R (2001) Brain-derived neurotroph- but not carcinogenicity induced by ochratoxin A, implicating two
ic factor in the control human brain, and in Alzheimer’s disease and separate pathways. FU Med Biol 9:57–63
Parkinson’s disease. Prog Neurobiol 63:71–124. https://doi.org/10. Pfohl-Leszkowicz, A., & Manderville, R.A., 2012. An update on direct
1016/S0301-0082(00)00014-9 genotoxicity as molecular mechanism of ochratoxin A carcinogenic-
Nabavi SF, Barber AJ, Spagnuolo C et al (2016) Nrf2 as molecular target ity. Chem. Res. Toxicol. 25, 252–262.Pfohl-Leszkowicz, A.,
for polyphenols: a novel therapeutic strategy in diabetic retinopathy. Hadjeba-Medjdoub, K., Ballet, N., Schrickx, J., Fink-Gremmels,
Crit Rev Clin Lab Sci 53(5):293–312 J., 2015. Assessment and characterisation of yeast-based products
Nehad E, Farag M, Kawther M, Abdel-Samed A, Naguib K (2005) intended to mitigate ochratoxin exposure using in vitro and in vivo
Stability of ochratoxin A (OTA) during processing and models. Food Addit Contam A 32(4), 604–616
decaffeination in commercial roasted coffee beans. Food Addit Pinelli E, El Adlouni C, Pipy B, Quartulli F, Pfohl-Leszkowicz A (1999)
Contam 22:761–767 Roles of cyclooxygenase and lipoxygenases in ochratoxin A
Nephew KP, Huang THM (2003) Epigenetic gene silencing in cancer genotoxicity in human epithelial lung cells. Environ Toxicol
initiation and progression. Cancer Lett 190(2):125–133 Pharmacol 7(2):95–107
Ozcan G, Ozpolat B, Coleman RL, Sood AK, Lopez-Berestein G (2015) Piotrowska M, Żakowska Z (2000) The biodegradation of ochratoxin A
Preclinical and clinical development of siRNA-based therapeutics. in food products by lactic acid bacteria and baker’s yeast. Prog
Adv Drug Deliv Rev 87:108–119 Biotechnol 307–310
Ozden S, Kara NT, Sezerman OU, Durasi İM, Chen T, Demirel G, Piva, A., Galvano, F., 1999. Nutritional approaches to reduce the impact
Alpertunga B, Chipman JK, Mally A (2015) Assessment of global of mycotoxins. Biotechnol. Feed Ind. 381–399
and gene-specific DNA methylation in rat liver and kidney in re- Pleadin J, Perši N, Kovačević D, Vulić A, Frece J, Markov K (2014)
sponse to non-genotoxic carcinogen exposure. Toxicol Appl Ochratoxin A reduction in meat sausages using processing methods
Pharmacol 289(2):203–212 practiced in households. Food Addit Contam B 7(4):239–246
Özpinar H, Bilal T, Abas I, Kutay C (2002) Degradation of ochratoxin A Poór M, Kunsági-Máté S, Bencsik T, Petrik J, Vladimir-Knežević S,
in rumen fluid in vitro. FU Med Biol 9:66–69 Kőszegi T (2012) Flavonoid aglycones can compete with
Environ Sci Pollut Res

Ochratoxin A for human serum albumin: A new possible mode of Schatzmayr G, Heidler D, Fuchs E, Binder E, Loibner A, Braun R (2002)
action. Int J Biol Macromol 51(3):279–283 Evidence of ochratoxin A-detoxification activity of rumen fluid,
Poór M, Veres B, Jakus PB et al (2014) Flavonoid diosmetin increases intestinal fluid and soil samples as well as isolation of relevant mi-
ATP levels in kidney cells and relieves ATP depleting effect of croorganisms from these environments. Mycotoxin Res 18:183–187
ochratoxin A. J Photochem Photobiol B Biol 132:1–9 Schatzmayr G, Heidler D, Fuchs E, Nitsch S, Mohnl M, Täubel M,
Qi X, Yang X, Chen S et al (2014a) Ochratoxin A induced early hepato- Loibner A, Braun R, Binder E (2003) Investigation of different yeast
toxicity: new mechanistic insights from microRNA, mRNA and strains for the detoxification of ochratoxin A. Mycotoxin Res 19:124
proteomic profiling studies. Sci Report 4:5163 Schatzmayr G, Zehner F, Täubel M, Schatzmayr D, Klimitsch A, Loibner
Qi X, Yu T, Zhu L et al (2014b) Ochratoxin A induces rat renal carcino- AP, Binder EM (2006) Microbiologicals for deactivating myco-
genicity with limited induction of oxidative stress responses. toxins. Mol Nutr Food Res 50:543–551
Toxicol Appl Pharmacol 280(3):543–549 Schatzmayr, G., Heidler, D., Fuchs, E., Binder, E.M., 2012.
Rached E, Pfeiffer E, Dekant W, Mally A (2006) Ochratoxin A: apoptosis Microorganism for biological detoxification of mycotoxins, namely
and aberrant exit from mitosis due to perturbation of microtubule ochratoxins and/or zearalenons, as well as method and use thereof.
dynamics? Toxicol Sci 92(1):78–86 Google Patents
Rahimtula A, Béréziat JC, Bussacchini-Griot V, Bartsch H (1988) Lipid Schmidt-Heydt M, Stoll D, Schütz P, Geisen R (2015) Oxidative stress
peroxidation as a possible cause of ochratoxin A toxicity. Biochem induces the biosynthesis of citrinin by Penicillium verrucosum at the
Pharmacol 37(23):4469–4477 expense of ochratoxin. Int J Food Microbiol 192:1–6
Ramyaa P, Padma VV (2013) Ochratoxin-induced toxicity, oxidative Schulz MC, Schumann L, Rottkord U, Humpf HU, Gekle M, Schwerdt G
stress and apoptosis ameliorated by quercetin–Modulation by (2018) Synergistic action of the nephrotoxic mycotoxins ochratoxin
Nrf2. Food Chem Toxicol 62:205–216 A and citrinin at nanomolar concentrations in human proximal
Ramyaa P, Padma VV (2014) Quercetin modulates OTA-induced oxida- tubule-derived cells. Toxicol Lett 291:149–157
tive stress and redox signalling in HepG2 cells—up regulation of Scott P (1996) Effects of processing and detoxification treatments on
Nrf2 expression and down regulation of NF-κB and COX-2. ochratoxin A: introduction. Food Addit Contam 13:19
Biochim Biophys Acta Gen Subj 1840(1):681–692 Sepúlveda L, Ascacio A, Rodríguez-Herrera R, Aguilera-Carbó A,
Rašić D, Mladinić M, Želježić D et al (2018) Effects of combined treat- Aguilar CN (2011) Ellagic acid: biological properties and biotech-
ment with ochratoxin A and citrinin on oxidative damage in kidneys nological development for production processes. Afr J Biotechnol
and liver of rats. Toxicon. 1646:99–105 10(22):4518–4523
Renzulli C, Galvano F, Pierdomenico L, Speroni E, Guerra M (2004) Serra RW, Fang M, Park SM, Hutchinson L, Green MR (2014) A KRAS-
Effects of rosmarinic acid against aflatoxin B1 and ochratoxin-A- directed transcriptional silencing pathway that mediates the CpG
induced cell damage in a human hepatoma cell line (Hep G2). J island methylator phenotype. eLife. 3:e02313
Appl Toxicol 24(4):289–296 Son WC, Kamino K, Lee YS, Kang KS (2003) Lack of effects of sodium
Reverberi M, Gazzetti K, Punelli F et al (2012) Aoyap1 regulates OTA 2-mercaptoethane sulfonate (mesna) on ochratoxin A induced renal
synthesis by controlling cell redox balance in Aspergillus ochraceus. tumorigenicity following life-time oral administration of Ochratoxin
Appl Microbiol Biotechnol 95(5):1293–1304 A in DA and Lewis rats. Toxicol Lett 142(1):19–27
Ringot D, Chango A, Schneider YJ, Larondelle Y (2006) Toxicokinetics Songsermsakul P, Razzazi-Fazeli E, Böhm J, Zentek J (2007) Occurrence
and toxicodynamics of ochratoxin A, an update. Chem Biol Interact of deoxynivalenol (DON) and ochratoxin A (OTA) in dog foods.
159(1):18–46 Mycotoxin Res 23(2):65–67
Romani S, Pinnavaia GG, Dalla Rosa M (2003) Influence of roasting Sorrenti V, Di Giacomo C, Acquaviva R et al (2012) Dimethylarginine
levels on ochratoxin A content in coffee. J Agric Food Chem 51: dimethylaminohydrolase/nitric oxide synthase pathway in liver and
5168–5171 kidney: protective effect of cyanidin 3-O-β-D-glucoside on
Rossiello MR, Rotunno C, Coluccia A, Carratù MR, Di Santo A, ochratoxin-A toxicity. Toxins 4(5):353–363
Evangelista V, Semeraro N, Colucci M (2008) Ochratoxin A in- Sorrenti V, Di Giacomo C, Acquaviva R, Barbagallo I, Bognanno M,
hibits the production of tissue factor and plasminogen activator Galvano F (2013) Toxicity of ochratoxin a and its modulation by
inhibitor-2 by human blood mononuclear cells: Another potential antioxidants: a review. Toxins 5(10):1742–1766
mechanism of immune-suppression. Toxicol Appl Pharmacol Spoendlin M, Moch H, Brunner F et al (1995) Karyomegalic interstitial
229(2):227–231 nephritis: further support for a distinct entity and evidence for a
Rudeš, K., 2006. The effect of ochratoxin A on the concentration of genetic defect. Am J Kidney Dis 25(2):242–252
protein carbonyls in rats. Farmaceutsko-biokemijski fakultet, Stachurska A, Ciesla M, Kozakowska M et al (2013) Cross-talk between
Sveučilište u Zagrebu microRNAs, nuclear factor E2-related factor 2, and heme
Russo A, La Fauci L, Acquaviva R et al (2005) Ochratoxin A-induced oxygenase-1 in ochratoxin A-induced toxic effects in renal proximal
DNA damage in human fibroblast: protective effect of cyanidin 3-O- tubular epithelial cells. Mol Nutr Food Res 57(3):504–515
β-D-glucoside. J Nutr Biochem 16(1):31–37 Stemmer K, Ellinger-Ziegelbauer H, Ahr HJ, Dietrich DR (2007)
Sava V, Reunova O, Velasquez A, Harbison R, Sanchez-Ramos J Carcinogen-specific gene expression profiles in short-term treated
(2006a) Acute neurotoxic effects of the fungal metabolite ochratox- Eker and wild-type rats indicative of pathways involved in renal
in-A. Neurotoxicology. 27(1):82–92 tumorigenesis. Cancer Res 67(9):4052–4068
Sava V, Reunova O, Velasquez A, Sanchez-Ramos J (2006b) Can low Streit E, Schatzmayr G, Tassis P et al (2012) Current situation of myco-
level exposure to ochratoxin-A cause parkinsonism? J Neurol Sci toxin contamination and co-occurrence in animal feed—Focus on
249(1):68–75 Europe. Toxins 4(10):788–809
Sava V, Velasquez A, Song S, Sanchez-Ramos J (2007) Adult hippocam- Sutken E, Aral E, Ozdemir F, Uslu S, Alatas O, Colak O (2007)
pal neural stem/progenitor cells in vitro are vulnerable to the myco- Protective role of melatonin and coenzyme Q10 in ochratoxin A
toxin ochratoxin-A. Toxicol Sci 98(1):187–197 toxicity in rat liver and kidney. Int J Toxicol 26(1):81–87
Schaaf G, Nijmeijer S, Maas R, Roestenberg P, De Groene E, Fink- Taniai E, Yafune A, Nakajima M et al (2014) Ochratoxin A induces
Gremmels J (2002) The role of oxidative stress in the ochratoxin karyomegaly and cell cycle aberrations in renal tubular cells without
A-mediated toxicity in proximal tubular cells. Biochim Biophys relation to induction of oxidative stress responses in rats. Toxicol
Acta Mol basis Dis 1588(2):149–158 Lett 224(1):64–72
Environ Sci Pollut Res

Tao Y, Xie S, Xu F et al (2018) Ochratoxin A: Toxicity, oxidative stress Wu TS, Yang JJ, Wang YW, Yu FY, Liu BH (2016) Mycotoxin ochra-
and metabolism. Food Chem Toxicol 112:320–331 toxin A disrupts renal development via a miR-731/prolactin receptor
Thomas B, Ogunkanmi L, Iwalokun B, Agu G (2018) Transcriptional axis in zebrafish. Toxicol Res 5(2):519–529
factor influence on OTA production and the quelling attribute of Yang Q, He X, Li X et al (2014a) DNA damage and S phase arrest
Sirna on the OTA producing strains of Aspergillus section Nigri. induced by Ochratoxin A in human embryonic kidney cells (HEK
Afr J Clin Exp Microbiol 18(4):210–217 293). Mutat Res Fundam Mol Mech Mutagen 765:22–31
Torack RM, Morris JC (1992) Tyrosine hydroxylase-like (TH) immuno- Yang Q, Shi L, Huang K, Xu W (2014b) Protective effect of N-
reactivity in Parkinson’s disease and Alzheimer’s disease. J Neural acetylcysteine against DNA damage and S-phase arrest induced by
Transm Park Dis Dement Sect 4:165–171. https://doi.org/10.1007/ ochratoxin A in human embryonic kidney cells (HEK-293). Food
BF02251479 Chem Toxicol 70:40–47
Tozlovanu M, Faucet-Marquis V, Pfohl-Leszkowicz A, Manderville RA Yang X, Liu S, Huang C, Wang H, Luo Y, Xu W, Huang K (2017)
(2006) Genotoxicity of the hydroquinone metabolite of ochratoxin Ochratoxin A induced premature senescence in human renal proxi-
A: structure-activity relationships for covalent DNA adduction. mal tubular cells. Toxicology 382:75–83
Chem Res Toxicol 19(9):1241–1247 Yong S, Albassam M, Prior M (1987) Protective effects of sodium bicar-
Tozlovanu M, Canadas D, Pfohl-Leszkowicz A, Frenette C, Paugh RJ, bonate on murine ochratoxicosis. J Environ Sci Health B 22(4):455–
Manderville RA (2012) Glutathione conjugates of ochratoxin a as 470
biomarkers of exposure/Glutationski Konjugati Okratoksina A Kao
Zanicgrubisic T, Zrinski R, Čepelak I, Petrik J, Radić B, Pepeljnjak S
Biomarkeri Izloženosti. Arch Hig Rada Toksikol 63(4):417–427
(2000) Studies of ochratoxin A-induced inhibition of phenylalanine
Trailović J, Stefanović S, Trailović S (2013) In vitro and in vivo protective
hydroxylase and its reversal by phenylalanine. Toxicol Appl
effects of three mycotoxin adsorbents against ochratoxin A in broiler
Pharmacol 167(2):132–139
chickens. Br Poult Sci 54(4):515–523
Van De Walle J, Sergent T, Piront N, Toussaint O, Schneider YJ, Zeljezic D, Domijan AM, Peraica M (2006) DNA damage by ochratoxin
Larondelle Y (2010) Deoxynivalenol affects in vitro intestinal epi- A in rat kidney assessed by the alkaline comet assay. Br J Med Biol
thelial cell barrier integrity through inhibition of protein synthesis. Res 39(12):1563–1568
Toxicol Appl Pharmacol 245(3):291–298 Zhang X, Boesch-Saadatmandi C, Lou Y, Wolffram S, Huebbe P,
Van Der Merwe KJ, Steyn PS, Fourie L, Scott DB, Theron JJ (1965) Rimbach G (2009) Ochratoxin A induces apoptosis in neuronal
Ochratoxin A, a toxic metabolite produced by Aspergillus cells. Genes Nutr 4(1):41
ochraceus Wilh. Nature 205(4976):1112–1113 Zhang B, Shen XL, Liang R et al (2014) Protective role of the mitochon-
Van Der Stegen GH, Essens PJ, Van der Lijn J (2001) Effect of roasting drial Lon protease 1 in ochratoxin A-induced cytotoxicity in
conditions on reduction of ochratoxin A in coffee. J Agric Food HEK293 cells. J Proteome 101:154–168
Chem 49:4713–4715 Zhang H, Apaliya MT, Mahunu GK, Chen L, Li W (2016) Control of
Varga J, Péteri Z, Tábori K, Téren J, Vágvölgyi C (2005) Degradation of ochratoxin A-producing fungi in grape berry by microbial antago-
ochratoxin A and other mycotoxins by Rhizopus isolates. Int J Food nists: a review. Trends Food Sci Technol 51:88–97
Microbiol 99:321–328 Zhang H, Wang Y, Zhao C, Wang J, Zhang X (2017) Biodegradation of
Varga J, Kocsubé S, Péteri Z, Vágvölgyi C, Tóth B (2010) Chemical, ochratoxin A by Alcaligenes faecalis isolated from soil. J Appl
physical and biological approaches to prevent ochratoxin induced Microbiol 123:661–668
toxicoses in humans and animals. Toxins 2:1718–1750 Zhang TY, Wu RY, Zhao Y et al (2018) Ochratoxin A exposure de-
Vettorazzi A, Van Delft J, De Cerain AL (2013) A review on ochratoxin creased sperm motility via the AMPK and PTEN signaling path-
A transcriptomic studies. Food Chem Toxicol 59:766–783 ways. Toxicol Appl Pharmacol 340:49–57
Wajapeyee N, Malonia SK, Palakurthy RK, Green MR (2013) Oncogenic Zhao J, Qi X, Dai Q et al (2017) Toxicity study of Ochratoxin A using
RAS directs silencing of tumor suppressor genes through ordered HEK293 and HepG2 cell lines based on microRNA profiling. Hum
recruitment of transcriptional repressors. Genes Dev 27:2221–2226 Exp Toxicol 36(1):8–22
Wang Y, Liu J, Cui J et al (2012) ERK and p38 MAPK signaling path- Zheng J, Zhang Y, Xu W et al (2013) Zinc protects HepG2 cells against
ways are involved in ochratoxin A-induced G2 phase arrest in hu- the oxidative damage and DNA damage induced by ochratoxin A.
man gastric epithelium cells. Toxicol Lett 209(2):186–192 Toxicol Appl Pharmacol 268(2):123–131
Wang S, Zhang C, Sheng X, Zhang X, Wang B, Zhang G (2014) Zhu L, Yu T, Qi X et al (2016) miR-122 plays an important role in
Peripheral expression of MAPK pathways in Alzheimer’s and ochratoxin A-induced hepatocyte apoptosis in vitro and in vivo.
Parkinson’s diseases. J Clin Neurosci 21:810–814. https://doi.org/ Toxicol Res 5(1):160–167
10.1016/j.jocn.2013.08.017
Zhu L, Zhang B, Dai Y, Li H, Xu W (2017) A review: epigenetic mech-
Wen J, Mu P, Deng Y (2016) Mycotoxins: cytotoxicity and biotransfor-
anism in ochratoxin a toxicity studies. Toxins 9(4):113
mation in animal cells. Toxicol Res 5(2):377–387
Zlokovic BR (2011) Neurovascular pathways to neurodegeneration in
Whitlow, L.W., 2006. Evaluation of mycotoxin binders. In: Proceedings
Alzheimer’s disease and other disorders. Nat Rev Neurosci 12(12):
4th Mid-Atlantic Nutr. Conf. 132–143
723–738
Wielogórska E, MacDonald S, Elliott C (2016) A review of the efficacy of
mycotoxin detoxifying agents used in feed in light of changing global Zurich MG, Lengacher S, Braissant O, Monnet-Tschudi F, Pellerin L,
environment and legislation. World Mycotoxin J 9(3):419–433 Honegger P (2005) Unusual astrocyte reactivity caused by the food
Wood AJ, Oakey RJ (2006) Genomic imprinting in mammals: emerging mycotoxin ochratoxin A in aggregating rat brain cell cultures.
themes and established theories. PLoS Genet 2(11):e147 Neuroscience 134(3):771–782
Wu BK, Brenner C (2014) Suppression of TET1-dependent DNA de-
methylation is essential for KRAS-mediated transformation. Cell Publisher’s note Springer Nature remains neutral with regard to jurisdic-
Rep 9:1827–1840 tional claims in published maps and institutional affiliations.

You might also like