Next Article in Journal
Detection of Enterotoxigenic Potential and Determination of Clonal Profile in Staphylococcus aureus and Coagulase-Negative Staphylococci Isolated from Bovine Subclinical Mastitis in Different Brazilian States
Next Article in Special Issue
Ochratoxin A: 50 Years of Research
Previous Article in Journal
The Kunitz-Type Protein ShPI-1 Inhibits Serine Proteases and Voltage-Gated Potassium Channels
Previous Article in Special Issue
Maternal-Fetal Cancer Risk Assessment of Ochratoxin A during Pregnancy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ochratoxin A: Molecular Interactions, Mechanisms of Toxicity and Prevention at the Molecular Level

1
Department of Laboratory Medicine, University of Pécs, H-7624 Pécs, Hungary
2
János Szentágothai Research Center, Lab-on-a-chip Research Group, H-7624 Pécs, Hungary
3
Department of Pharmacology and Pharmacotherapy, Toxicology Section, University of Pécs, H-7624 Pécs, Hungary
*
Author to whom correspondence should be addressed.
Toxins 2016, 8(4), 111; https://doi.org/10.3390/toxins8040111
Submission received: 23 February 2016 / Revised: 31 March 2016 / Accepted: 6 April 2016 / Published: 15 April 2016
(This article belongs to the Collection Ochratoxins-Collection)

Abstract

:
Ochratoxin A (OTA) is a widely-spread mycotoxin all over the world causing major health risks. The focus of the present review is on the molecular and cellular interactions of OTA. In order to get better insight into the mechanism of its toxicity and on the several attempts made for prevention or attenuation of its toxic action, a detailed description is given on chemistry and toxicokinetics of this mycotoxin. The mode of action of OTA is not clearly understood yet, and seems to be very complex. Inhibition of protein synthesis and energy production, induction of oxidative stress, DNA adduct formation, as well as apoptosis/necrosis and cell cycle arrest are possibly involved in its toxic action. Since OTA binds very strongly to human and animal albumin, a major emphasis is done regarding OTA-albumin interaction. Displacement of OTA from albumin by drugs and by natural flavonoids are discussed in detail, hypothesizing their potentially beneficial effect in order to prevent or attenuate the OTA-induced toxic consequences.

Graphical Abstract

1. Introduction

Ochratoxin A (OTA) is a well-known and widely-spread mycotoxin all over the world [1]. Ochratoxins (A, B, and C) are secondary metabolites of Pencillium and Aspergillus micro fungi of which mainly the A form exerts hazardous effects in animals and in humans, as well [1,2,3]. OTA was first found in the Balkan region; however, it can be detected practically in all territories, it is accumulated in animal feed and in human food due to the favorable weather conditions and microclimate, and/or to improper storage of food components [4]. OTA is present at all stages of the food chain (cereals, meat, fruits, wine, beer, coffee, etc.) [5,6,7,8,9,10], and based on previous studies its presence may be associated with the chronic tubulo-interstitial kidney disease called Balkan Endemic Nephropathy (BEN) [11,12,13,14]. BEN is a chronic progressive disease with a period of 6–10 years leading to irreversible kidney failure. However, apart from long-term OTA exposition in the endemic areas, some other potential causative factors are postulated in the development of BEN: aristolochic acid, heavy metal intoxication, selenium deficient diet, and genetic predisposition [11,12,15]. Due to its high heat stability, complete removal of OTA from food is practically impossible [16], although several approaches exist for reducing OTA contamination [17,18,19].
After the absorption of OTA from the gastrointestinal tract, it binds primarily to albumin with high affinity, which results in its very long half-life (from a few days to one month, depending on species). In healthy human populations the plasma concentration of OTA ranges from some hundreds of pmol/L to a few nmol/L, while in endemic areas it can exceed 100 nmol/L [20,21,22,23]. Due to the pKa values of OTA (4.2–4.4 and 7.0–7.3, the carboxyl group and the phenolic hydroxyl group, respectively) [24], at physiological pH the toxin is present in monoanionic (OTA) and in dianionic (OTA2−) forms [25,26]. In the circulatory system OTA is almost completely bound to albumin. The extent of albumin binding determines the plasma half-life of OTA, being the longest in humans (compared to other species) of approximately one month [27,28]. Albumin binding strongly affects toxicokinetics of OTA: in albumin deficient rats the toxin excretion through the kidneys was 20–70-fold faster than in normal rats [29]. The increased elimination rate might reduce the chronic exposure of the target cells (mainly kidney tubule and liver cells). Since albumin is the key transport protein of the toxin in the circulatory system, and because of the fact that the association constant of the OTA-albumin complex is extremely high (more than 107 L/mol) [25,30,31,32], extensive studies are known characterizing the albumin binding of OTA. Although glomerular filtration of OTA is strongly limited due to its albumin binding, the small filtrated and secreted fraction is partially reabsorbed [1], which might help the accumulation of the toxin in the kidney tubule cells.
OTA toxicity is strongly correlated with the occurrence of BEN [9,33,34]; however, its mechanism of action is very complex [35,36,37,38,39]. It is thought to be carcinogenic, teratogenic, hepatotoxic, neurotoxic, and immunotoxic, based on in vitro and on animal studies [1,40]. IARC (International Agency for Research on Cancer) categorizes OTA as a member of 2B subgroup which means that, based on animal studies, OTA is a potential human carcinogen [34,41]. The NCI/NTP (National Cancer Institute/National Toxicological Program) renders OTA to be the most potent renal carcinogen in rodents ever studied [42]. In fact, the incidence of upper urinary tract tumors in endemic regions of Bulgaria is 90-fold higher compared to that of non-endemic regions [43]. In addition to BEN, some studies make emphasis on the role of OTA in the development of Tunisian Nephropathy [44], gastric and esophageal tumors in some regions of China [45,46], as well as testicular cancer [47].
In spite of the several known hypotheses, the mode of action and prevention, or at least attenuation of OTA toxicity, are only partially understood. The present review focuses on the basic knowledge related to Ochratoxin A and on the current information on its molecular and cellular interactions, and also on the attempts to prevent/attenuate its toxicity at the molecular and cellular level.

2. Chemistry of Ochratoxins

Ochratoxins (Ochratoxin A: OTA, Ochratoxin B: OTB and Ochratoxin C: OTC) are toxic metabolites of different fungi; their structure consists of a dihydro-isocoumarin moiety linked with a phenylalanine through an amide bond (Figure 1). Furthermore, OTA and OTC contain a parachlorophenol part as well. OTA (C20H18ClNO6; IUPAC name: N-{[(3R)-5-chloro-8-hydroxy-3-methyl-1-oxo-3,4-dihydro-1H-isochromen-7-yl]carbonyl}-L-phenylalanine; molecular weight: 403.8) is a white, odorless, heat stable, crystalline solid agent (melting point: 168–173 °C) with poor aqueous solubility [3,48]. OTA does not completely disappear during baking [49]; furthermore, OTA resists against three hours of high pressure steam sterilization at 121 °C [50], and even at 250 °C it is only partially degraded [51]. Even during coffee roasting OTA is only partially decomposed [52,53]; one of the identified products is 14-(R)-ochratoxin A (about 25%) that shows slightly lower cytotoxicity in vitro [54]. Due to the structure of OTA, the mycotoxin exhibits strong fluorescence property [25,26,30]. Depending on the microenvironment, OTA exists in non-ionic, monoanionic (OTA), and dianionic (OTA2−) forms.

3. Dietary Sources of OTA

Ochratoxin A occurs in wheat, fruits, oilseeds, and animal feed resulting in its presence in milk, meat, and even in eggs [1,3]. Therefore, many drinks (e.g., wine, beer, coffee, tea, milk, etc.) [55,56,57], as well as common meals (bakery, meat, and dairy products) [1,40] contain more or less amounts of OTA. Furthermore, recent studies also highlighted its presence in herbal medicines [58,59,60], food coloring agents [61], spices [62,63,64], and even in bottled water [65]. The wide occurrence of OTA and its high thermal stability makes the eradication of OTA from the food chain very difficult.

4. Toxicokinetics of OTA

4.1. Absorption

The amount of absorbed toxin is species-dependent; in pigs it is about 60%, while in rodents it is much lower [27,66]. The oral bioavailability of OTA is the highest in humans, approximately 93% [27,28]. Based on data of animal experiments, the non-ionic and monoanionic forms of OTA are absorbed from the stomach and the jejunum without known specific transport mechanisms [67,68,69]. On the other hand, the MRP2 multidrug resistance efflux transporter might slow down its absorption from the small intestine due to the transport of OTA back to the intestinal lumen [70]. Furthermore, OTA is also a substrate for BCRP (ABCG2) but not for P-gp (P-glycoprotein), suggesting the potential role of BCRP similar to that of MRP2 [71]. Furthermore, in vitro studies suggest that OTA alters the intestinal barrier and absorption functions [72].

4.2. Distribution

4.2.1. Binding to Plasma Proteins

Albumin is the most abundant plasma protein in the circulatory system. Albumin binds OTA with unusually high affinity; therefore, 99.8% of OTA is in albumin-bound form in the human circulatory system [28]. Erythrocytes contain only traces of OTA [67]. It has been shown that the primary binding site of OTA on human serum albumin (HSA) is located on subdomain IIA (Sudlow’s Site I) [24,73,74,75]. There is a secondary binding site on subdomain IIIA (Sudlow’s Site II) but with much less affinity, suggesting its negligible relevance regarding the toxicokinetics of OTA [76]. The primary binding site of OTA is almost identical to that of warfarin (also presenting a coumarin backbone) [77]. The isocoumarin moiety of OTA is localized in an apolar cavity among amino acids A291, L238, I260, I264, I290, R257, and S287, while the phenyl group is surrounded by K199, H242, Y211, L238, and W214 amino acids [75]. The oxygen atoms of the carbonyl and phenolic hydroxyl groups orientate towards R257, while the carboxylic group towards R218 and/or R222 amino acids; data obtained for modified albumin strongly support the pivotal role of R257 and R218 arginines in the interaction [75]. The R257 arginine can deprotonate the phenolic hydroxyl group of OTA resulting in the formation of a very stable ion pair with HSA; this explains why the dianionic form of OTA is bound to HSA (even if the monoanionic form binds, it is rapidly deprotonated).
Apart from albumin, some other proteins of a molecular mass of about 20 kDa have been described which exert even higher binding affinity than albumin does [78,79]. These proteins are of much less concentration than albumin; however, they might have a potential role in the pathogenesis of BEN because of their free filtration through the glomeruli.

4.2.2. Tissue Distribution

The tissue distribution of OTA is species-dependent and is also largely influenced by many factors including the amount of toxin, the way of ingestion, the composition of the diet, and the overall health status of the body. However, the major targets are the kidneys and the liver [1,3], skeletal muscle, fat tissue, and the brain also contain lesser amounts of the toxin, as well [80]. A possible explanation for the vulnerability of the kidneys and the liver might be explained by their special transport mechanisms. In the kidneys organic anion transporters (OATs), while in the liver organic anion-transporting polypeptides (OATPs), are the major molecular structures responsible for the active cellular uptake of OTA (OATs and OATPs are membrane transport proteins, belonging to the solute carrier transporter family) [81,82,83]. While basolateral OATs are mainly responsible for the uptake of OTA from blood into the tubule kidney cells, the apical OAT4 transporter may be involved in the urinary reabsorption of OTA resulting again its accumulation in tubule kidney cells [83]. Some of the kidney-specific OATPs can take part in the cellular uptake of OTA, as well [84]. Furthermore, low doses of OTA led to the increased expression of more organic anion transporter isotypes in rat kidneys [85]. In the proximal tubule cells a 62 kDa protein was identified with strong toxin-binding characteristics which might facilitate the tissue accumulation of the toxin [79].
Since under physiological circumstances OTA is present as a charged molecule, it crosses the placenta most probably by active transport mechanisms. Unfortunately, OTA levels are higher in the placenta and also two-fold higher in the fetus’s blood than those of the mother [86]. In vitro experiments suggest the possible involvement of OAT4 transporter [87].

4.3. Biotransformation

Previous studies suggested that most of OTA remains unchanged, and that liver is not the sole organ to metabolize OTA [88]. Nevertheless, OTA can be biotransformed by both phase I and phase II enzymes. The majority of the metabolites (Figure 2) show low or no toxicity. In the gut, part of OTA is hydrolyzed to Ochratoxin α (OTα) by the action of proteolytic enzymes and by enzymes of the bacterial microflora [89,90,91,92]. Another possibility of the hydrolysis of OTA is opening the lactone ring under alkaline conditions that results in the formation of a highly toxic compound called lactone-opened OTA (OP-OA) [93]. 4-hyroxyochratoxin A (4-OH-OTA) is a product of microsomal oxidation of the toxin with low toxicity [94,95,96,97,98], while another less toxic product is 10-hydroxyochratoxin A (10-OH-OTA) [99,100,101,102]. Moreover, the formation of further hydroxyl metabolites of OTA was also observed in different species [103]. These phase I-type reactions are most probably related to the action of the CYP450 enzyme family [95,104,105,106,107,108,109]. Some studies suggest that after dechlorination OTA is transformed to Ochratoxin B with less genotoxicity [39,110,111,112,113]. Among phase II reactions sulfate, glucuronide, hexose/pentose (hex/pen-OTA), and glutathione (OT-GSH) conjugations of OTA are described as well [114,115,116,117,118]. The above listed metabolites have been observed in tissues, blood, and urine of animals and/or humans. Furthermore, in vivo studies suggest that there are sex differences regarding the carcinogenicity of OTA originating from the variation of OTA biotransformation in animals [15,33,37,119,120,121].

4.4. Excretion

4.4.1. Renal Excretion

Due to the strong albumin binding of OTA, its elimination by glomerular filtration is negligible. Excretion of OTA is primarily done through tubular secretion. The tubular reabsorption of the toxin might be considered to be partially responsible for the intracellular accumulation of OTA [1]. Both in vitro and in vivo experiments suggest the importance of organic anion transporters [122,123,124]. Human OAT1 in the kidneys, while OAT3 transporter in the liver and brain, are considered as active members in the uptake of OTA from blood into tissues [81]. MRP2 transporters may play a role in the transport of OTA from kidney tubule cells into the urine [125,126]. Unfortunately, in vivo studies verify that the toxin is able to be reabsorbed from practically any part of the nephron both by active transport and by passive diffusion in a pH-dependent fashion [127].

4.4.2. Fecal Excretion and Entero-Hepatic Circulation

Biliary excretion of OTA and its metabolites is the major route in its fecal excretion; however, secretion into the small intestine is also present. Intestinal mechanisms might involve MRP2 and BCRP transporters [70,71,128,129,130,131].
In animals, especially in rodents, the role of entero-hepatic circulation of OTA has been demonstrated [68,69,128,132]. In mice, the biliary excretion of the conjugated form of the toxin was observed; OTA possibly absorbs again after the hydrolysis of its conjugate by the bacterial microflora [69]. In addition to the strong albumin binding property, entero-hepatic circulation might enhance the slow elimination of OTA from the body.

4.4.3. Excretion through Breast Milk

Although a great fluctuation is described for the toxin concentration in milk [133,134], there is a direct relationship between the ingestion of OTA and its concentration in the milk [135]. In a human study it was observed that the highest OTA level was found in breast milk during the first few days after delivery [136].

5. Mode of Action of OTA

5.1. Inhibition of Protein Synthesis

OTA is an inhibitor of protein synthesis in both in vivo and in vitro models. It was verified that OTA can inhibit the activity of phenylalanine t-RNA synthase [137,138]. First it was hypothesized that the phenylalanine moiety of OTA has a major role as a competitor between phenylalanine and the toxin. However, further studies showed that the isocoumarin structure is more important in this interaction than the phenylalanine moiety because modification of the isocoumarin structure has a significant impact on this action [139,140]. Docking studies have also supported the slight importance of the phenylalanine part [141]. Furthermore, OTA is also an inhibitor of phenylalanine hydroxylase; the toxin behaves as a false substrate of the enzyme because the hydroxylation of its phenylalanine moiety results in tyrosine-containing OTA which was detected even in in vivo experiments [142]. However, we have to note that the effects of OTA on phenylalanine t-RNA synthase and phenylalanine hydroxylase, in vivo, were observed after the treatment with relatively high OTA doses [137,142]. In addition to these non-specific ways of protein synthesis inhibition, OTA may influence the transcription of many proteins resulting in specific intracellular effects [143].

5.2. Inhibition of Cellular Energy Production

OTA has a strong negative effect on cellular energy (ATP) production [144]. Mitochondrial dysfunction is an early sign of toxicity [145] resulting in an overall decrease in protein synthesis. There are some key enzymes in gluconeogenesis e.g., phosphoenolpyruvate-carboxykinase (PEPCK) which shows decreased activity due to OTA exposure [146,147]. It was also proven that OTA interferes with the expression of PEPCK at the mRNA level [148,149]. Furthermore, the toxin can penetrate into the mitochondria and most probably binds to proteins involved in maintenance of the membrane potential and the oxidative phosphorylation by interfering with phosphate transport and by inhibition of electron transport as well [150,151,152].

5.3. Genotoxic Effect

Several experiments suggest that OTA has genotoxic effects [38]. Following bioactivation, electrophilic products are formed from the toxin which can covalently bind to DNA causing mutations and subsequent formation of malignant tumors. Figure 3 summarizes the mechanisms of adduct formations detailed below.
A common principle for the different theories based on the parachlorophenol (PCP) structure found in OTA [153] is that PCP can undergo CYP450-catalized oxidative dechlorination, resulting in a quinoidal structure that can bind covalently to thiol groups as well as 2′-deoxyguanosine (dG), or other adducts may form [154,155,156]. Oxidation of OTA by CYP450 enzymes also produces a reactive electrophilic product called OTA-quinone (OTQ). OTQ can be partially detoxified by conjugation with GSH or it possibly forms OTA-hydroquinone (OTHQ) after reduction [117,157,158]. OTHQ was detected both in rat and in human urine samples [15,37,121,159].
Another theory considers the activation by peroxidases as an important step to produce the phenoxyl radical from OTA [160,161]. Then, in the presence of glutathione, the phenoxyl radical may be converted to OTA again, but at the expense of the formation of a superoxide anion radical (O2¯) [162,163]. O2¯ forms H2O2 which can induce a Fenton reaction resulting in the appearance of a hydroxyl radical (OH), causing oxidative damage again. This mechanism might partly explain the observation that OTA depletes GSH in cellular models [164]. Furthermore, the phenoxyl radical may directly form C8-deoxyguanosine-adducts (C8-dG) [165,166].
Reductive dechlorination of OTA may produce reactive aryl radicals [167] resulting in the formation of C8 purine nucleotide adducts [168,169,170].
Figure 4 demonstrates the chemical structure of the potentially-occurring dG adducts. Photoreaction of OTA in the presence of dG and further reactions in the presence of OTA, dG, Fe2+, or horseradish peroxidase/H2O2 resulted in C-coupled C8-dG-adducts (C-OTB-dG) [171]. C-OTB-dG adduct may be produced through phenoxyl and aryl radical pathways; however, the phenoxyl radical of OTA may also lead to the formation of O-coupled C8-dG-adducts (O-OTA-dG) [172,173]. Finally, the third potential compound is the OTHQ-dG-adduct [37,117]. In spite of much in vitro and in vivo experimental data obtained by isotope and LC-MS techniques, there are controversies about the presence of DNA adducts [174,175]. C-OTB-dG adduct formation might occur with the highest probability [176] but several authors seriously query the presence of DNA adducts due to OTA exposure [177,178,179]. A recent study suggests that OTA alone does not lead to DNA adduct formation in rats; however, the co-treatment of aristolochic acid with OTA results in higher adduct formation than the treatment with aristolochic acid alone [180].

5.4. Induction of Oxidative/Nitrosative Stress

Several studies suggest that both in vitro and in vivo OTA exposure results in the overproduction of free radicals. Increased ROS production, as well as oxidative damage (lipids, proteins, and DNA) are described by many authors [1,181]. The formation of each OTQ or phenoxyl and aryl radicals can lead to increased ROS production as discussed previously (Figure 3). Furthermore, OTA can possibly induce lipid peroxidation using Fe3+ as cofactor. In the presence of NADPH-CYP450 reductase OTA-Fe3+ complex facilitates the reduction of Fe3+ and the formed OTA-Fe2+ complex initiates the appearance of free radicals leading to lipid peroxidation and DNA damage [182,183].
Recent data indicate that intracellular Zn2+ levels might also be in connection with OTA toxicity. OTA can deplete Zn2+ most probably by its action on transporter proteins and metallothioneins [184]. It was shown in liver cell cultures that OTA significantly increases ROS concentration and expression of several metallothioneins, while reducing superoxide dismutase (SOD) activity and catalase mRNA levels [185]. Furthermore, OTA is able to directly interact with Zn2+; the complex formation can also be responsible for the decreased presence of free intracellular zinc levels [186].
Moreover, OTA treatment causes not only increased ROS production but also reduces the antioxidant defense of cells by lowering activator protein 1 (AP-1) and nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) activation, which molecules regulate the transcription of glutathione, glutathione S-transferase (GST), and further cytoprotective enzymes [187,188,189]. Recent studies highlighted that OTA can also inhibit the expression of Nrf2 protein, its translocation into the cell nucleus, as well as its binding to DNA [190].
It is important to note that the reactive nitrogen species levels may also increase in OTA-treated cells. OTA facilitates the expression of inducible nitrogen oxide synthase (iNOS) enzyme and also increases the expression and activity of dimethylarginine dimethylaminohydrolase (DDAH) with subsequent elevation of nitrogen monoxide (NO) synthesis and increased nitrite/nitrate concentrations [181,191]. High levels of NO may cause nitrosative stress because it can react with O2¯ resulting in the formation of peroxynitrite (ONOO¯), which in turn gives nitrogen dioxide (NO2) and hydroxyl radicals (OH).
Nevertheless, some studies suggest that the carcinogenic effect of OTA is independent from oxidative damage. In these studies, OTA-induced increased cell proliferation, cell cycle aberration, and apoptosis were observed in rats [192,193].

5.5. Apoptosis

OTA can cause both apoptotic and necrotic cell death [1,181]. Even at nanomolar concentration apoptosis markers could be observed, e.g., DNA fragmentation, chromatin condensation and increased caspase-3 activity [194]. Altered expression of different genes could be a potential cause of apoptosis: marked transcriptional changes of many genes were observed which are involved in DNA damage response and apoptosis (e.g., GADD153, GADD45, clusterin, and p53) [192,195]. The important protective role of p53 regarding the carcinogenic effect of OTA-treated mice was demonstrated by Kuroda et al. [196]. Regulation of signal transduction pathways can also be a possible reason of apoptosis induction; activation of MAPK-, ERK-, p38, and JNK were described in kidney and/or liver cells [194,197,198]. Interestingly, ERK is activated only weakly, and p38 is not activated at all, by OTA in human kidney cells; despite OTA being a strong activator of MAPK, ERK, and p38 in rat liver cells [197]. In a recent study, the sustained activation of c-MET/PI3K/Akt and MEK/ERK1-2 signaling pathways were reported in human kidney cells [199]. Another study suggests the role of apoptosis signal-regulating kinase 1 (ASK1) which can activate JNK and p38 pathways, as well as it has a pivotal role in oxidative stress- and in endoplasmic reticulum stress-induced cell death [200]. The probability of the type of OTA-induced cell injury (apoptosis or necrosis) most likely depends on the applied concentration of the toxin [198]. Oxidative stress may also play a role in cell death [201,202]; however, its relevance is still unclear. In other experiments, increased pro-inflammatory mediator levels (TNFα- and IL-6) were observed in rat liver perfusates of OTA-treated rats [203,204], where the NF-κB pathway might also be involved [205].

5.6. Influence on Mitosis

Several researchers consider OTA to be a non-mutagenic agent with no direct effect on DNA. They explain the OTA-induced carcinogenesis by causing disruptions of mitosis and chromosomal instability [179,206,207,208,209]. In human kidney cells, the block of the metaphase/anaphase transition, aberrant mitotic formations, and giant cells were observed [206,208]. Disorganization of the microtubular system and inhibition of histone acetyl transferase (HAT) enzymes were also detected [209]. Since HATs are responsible for the regulation of gene expression, DNA repair, and cell cycle control, their inhibition can lead to disruption of mitosis, cell proliferation and genetic instability [179].

5.7. Induction of Cell Cycle Arrest

Several studies have shown the negative effects of OTA on cell cycle in kidney cells and in lung fibroblasts [164,210,211]. Selective G2 phase arrest was observed in OTA-treated gastric epithelial cells [45]. Negative effects of OTA on cyclin-CDK (cyclin-dependent kinase) system were established: decreased expression of CDK25, CDK2, and cyclin B1 were observed both at the protein and at the mRNA levels, and the amount of the cyclin B1-CDK2 complex was reduced as well. It appeared that activation of ERK and p38 plays an important role in the above mechanism [212]. The role of free radicals cannot be underestimated because N-acetyl cysteine (NAC) supplementation effectively abolished OTA-induced cell cycle arrest [213]; the authors suggest the involvement of ATM-Chk2 and ATM-p53-p21 signaling pathways. Downregulation of Cdc2 and cyclin B1, as well as consequential G2 phase arrest, were also observed in esophageal cells [46]. In another study performed with human peripheral blood mononuclear cells, OTA caused G1 phase cell cycle arrest due to the decreased CDK4 and cyclin D1 expression; NAC alleviated again this unpleasant effect [214]. Furthermore, decreases of both mRNA and protein levels of cyclin A2, cyclin E1, and CDK2 were observed in human embryonic kidney cells after treatment with OTA, resulting in S-phase cell cycle arrest [215]. These negative consequences were alleviated with the pretreatment of cells with NAC [216].

5.8. Other Mechanisms

Few previous studies suggest that, due to lipid peroxidation in OTA-treated cells, the permeability towards Ca2+ increases and also the intracellular calcium stores may be depleted [217,218]. Hoehler et al. [219,220] found in kidney cells that increased intracellular Ca2+ in the presence of OTA causes uncoupling of oxidative phosphorylation and decreases ATP synthesis. OTA, even in nanomolar concentrations, negatively affects Ca2+- and cAMP homeostasis interfering with calcium signaling which, in turn, results in abnormal cell proliferation [221]. Another possible explanation of the increase in total intracellular calcium is the characteristic shrinkage of toxin-treated cells. It has been postulated by Dopp et al. [218] that OTA binds irreversibly to actin filaments causing their shortening and aggregation.
OTA is able to directly interact with different alkali and alkaline earth metal ions; low stability complexes are formed with K+, Na+, and Li+ ions while the presence of more stable complexes of OTA were observed with Ba2+, Ca2+, and Mg2+, in this order [26]. Each ion showed higher preference towards the dianionic form of OTA. Similarly with OTA-Zn2+ interaction [186], the biological importance of these complex formations are yet unclear.

6. Protective Agents—Overview

There have been some attempts to inhibit the absorption of OTA from the gastrointestinal tract by using adsorbents [19,222,223]. However, these methods are not selective and cause problems in long-term usage. The anion exchange resin cholestyramine can bind OTA in the gut, therefore inhibiting its absorption, enhancing its fecal elimination, and disrupting the enterohepatic circulation of OTA [224,225]. Application of NaHCO3 favors ionization of the toxin, decreasing its gastrointestinal absorption and increasing its elimination through the urine [226]. However, long-term usage of both cholestyramine and NaHCO3 is not recommended. Piroxicam is a substrate of OAT and may compete with OTA for its uptake into kidney cells [227]. However, larger doses of piroxicam also lead to nephrotoxic consequences. Application of the enzyme inductor phenobarbital resulted in controversial outcomes in different studies [102,228,229,230]. Phenylalanine and aspartame may counteract with the nonspecific effects of OTA in reducing protein synthesis due to the competitive action of the phenylalanine moiety [137,231,232,233]. However, contradictory results regarding phenylalanine exist as well [234].
There have been many trials to reduce the effects of OTA by administration of antioxidants prior, simultaneously, and after toxin exposure in cellular and in animal models. Application of SOD and catalase enzymes or the support of the glutathione system by NAC attenuated some negative effects of OTA (e.g., cytotoxicity, cell cycle arrest, or DNA damage) in several cases [213,214,216,235]. In other studies NAC only poorly alleviated the OTA-induced toxicity [113,236]. In cellular models vitamin C, α-tocopherol (vitamin E), and retinol (vitamin A) supplementation proved to be successful as well [237,238,239,240]. Pretreatment or co-treatment of OTA-exposed cells with different polyphenolic and other natural compounds showed positive effects on cell viability [181]. Pretreatment of kidney cells with epigallocatechin gallate (EGCG) increased the proliferation rate and decreased ROS levels and DNA fragmentation [241]; however, simultaneous exposure with the toxin or exposure after OTA treatment did not cause attenuation of toxicity. Co-exposure with rosmarinic acid of liver cells resulted in decreased ROS production and improvement in viability with less inhibition of protein and DNA synthesis [242]. Similar beneficial effects were found during the pretreatment or co-treatment of gingival fibroblasts, colon epithelial and liver cells with cyanidin 3-O-beta-d-glucoside (C3G) [243,244]. Animal experiments showed that C3G could attenuate OTA toxicity by influencing increased DDAH- and iNOS activation in rats, and in this way counterbalancing nitrosative stress [245]. Silibinin effectively decreased OTA-induced apoptosis in hepatocytes [246]. Luteolin, chlorogenic acid, and caffeic acid attenuated the OTA-mediated viability loss in kidney cells and lymphocytes while decreasing OTA-induced DNA damage of the blood cells of BALB/c mice [247]. Quercetin pretreatment suppressed OTA-induced cytotoxicity and oxidative stress and prevented OTA-induced apoptosis by lowering the activation of caspase cascade, DNA fragmentation, and micronucleus formation in kidney and in liver cells; these positive effects were attributed to the activation of Nrf2 pathway by quercetin causing the protection against OTA-induced alteration of the antioxidant defense [248,249]. Furthermore, diosmetin completely abolished the OTA-induced ATP depletion in kidney cells [144]. Carotenoid lycopene alleviated OTA-induced DNA damage, oxidative stress and apoptosis in rats [250,251]. Treatment with certain plant extracts also resulted in beneficial impacts in animal experiments [181,252,253]. Since OTA depletes intracellular zinc levels, zinc supplementation might be beneficial [185]. Furthermore, selenomethionine alleviated OTA-induced toxicity in kidney cells, the positive impact is possibly caused by the improvement of the expression of some selenoenzymes [254].
Despite of the presence of several data from in vitro and from animal studies, we have no strict evidence regarding the protective effect of the above-listed agents. Many hypotheses exist on the main toxic mechanism of OTA; however, even nowadays our knowledge is limited. Since we do not know the exact target of OTA in human kidney cells responsible for the toxic consequences after the long-term exposition with the mycotoxin, both the prevention and the treatment of OTA-induced toxicity are very difficult. Furthermore, the application of these protective agents can be problematic, in general, because of poor gastrointestinal absorption, high presystemic elimination, or insufficient tissue distribution. On the other hand, enhancement of the elimination of OTA from the body may be a promising tool to reduce the long-term exposition of target cells.

7. Displacement of OTA from Albumin

In our previous experiments three different models were applied for the calculation of the association constant (K) of OTA-HSA complex which showed logK values at about 7.3–7.6 [25,31,32]. However, OTA-albumin binding of different species is not uniform. Human serum albumin (HSA) seems to exert a very strong binding while, e.g., bovine (BSA) or rat serum albumin (RSA), show less affinity for the toxin [24,31]. In our previous study, the association constants of OTA-HSA, OTA-BSA, and OTA-RSA were determined (logKOTA-HSA = 7.65 ± 0.36, logKOTA-BSA = 6.48 ± 0.22 and logKOTA-RSA = 6.17 ± 0.12) [31]. Thus, the stability of OTA-HSA complex is 15-fold and 30-fold higher compared to those of OTA-BSA and OTA-RSA complexes, respectively. This observation perfectly explains why we notice, by far, the longest elimination half-life of OTA in humans [28]. A previous study with albumin-deficient rats showed the very high importance of albumin binding regarding the slow elimination of OTA [29]. Furthermore, a recent study with rats also proved that the accumulation of OTA in the kidney is due to strong binding of OTA to plasma proteins and its long half-life in plasma [255]. It is very likely that, in humans, this impact could be more dominant because of the substantially stronger interaction of OTA with human albumin.
If we displace a considerable amount of OTA from albumin, its increased elimination rate might reduce the chronic exposure of the target cells (mainly kidney tubule and liver cells). In this case, the free (not albumin-bound) OTA concentration will increase both in blood and in urine. Since the total concentration of OTA in the blood is not so high, it is very unlikely that the displacement will cause acute toxicity. OTA is transported to kidney and liver cells through active transport proteins. Due to the half-maximal transport rate of OTA regarding organic anion transporters is relatively low [81], the cellular uptake of OTA may become capacity-limited in the presence of other endogenous and exogenous substrates of these transporters. On the other hand, the half-maximal transport rate of OTA through OATP is high (approximately 20 µM) [82]; therefore, it is very unlikely that the uptake of OTA by OATP will become capacity-limited. Protective effects of nonsteroidal anti-inflammatory drugs piroxicam and indomethacin against the toxic impacts of OTA could be partly caused by the competitive displacement of OTA from albumin [227,256,257]. Co-treatment with phenylbutazone caused negative result in some studies [258]; however, our previous investigation revealed that phenylbutazone is a poorly effective competitor in this context [30]. In addition to piroxicam, aspartam is also able to effectively displace OTA from HSA [259]. After six-week pretreatment of rats with OTA, administration of aspartame for 10 days resulted in the strong decrease of OTA in the blood and kidneys [235]. Furthermore, lower levels of OTA in the brain, liver, and testicles were observed, as well as relatively higher rates of poorly-toxic metabolites of OTA (Ochratoxin α and hydroxylated OTA derivatives) were found. These results also strongly suggest the protective effect of displacement of OTA from albumin. In our previous study, we demonstrated that besides warfarin and the above-listed drugs glipizide, simvastatin, and mainly furosemide, are also able to compete with OTA for binding to HSA [30]. In our opinion, it would be highly interesting to examine the occurrence of BEN and urinary tract tumors in patients who have been continuously treated with these drugs and live in the endemic areas.
Application of naturally-occurring flavonoids for the same purpose could also be very promising. As we demonstrated, flavonoid aglycones are able to displace OTA from HSA very effectively; they are much stronger competitors than the drug molecules tested [25,30]. Most of the flavonoids are therapeutically safe and, besides the displacement of OTA from albumin, they may have further positive effects (see in Section 6). Furthermore, some flavonoids are able to inhibit different OAT and OATP transporters [260,261,262] which take part in the cellular uptake of OTA. In contrast, we also need to mention that certain flavonoids can inhibit the MRP2 transporter as well, which can decrease the excretion of OTA [70,263,264]. The first-pass metabolism of flavonoids is high, and it results in their limited pharmacological effects [265]. On the other hand, previous studies highlighted that many flavonoid metabolites also show (even similar or higher) albumin binding properties compared to the parent compound [266].

8. Conclusions

As we discussed above, the mechanism of action of OTA is still unclear; therefore, it is difficult to develop effective strategies in order to alleviate the OTA-induced toxicity at the cellular level. On the other hand, disruption of the normal toxicokinetics of OTA, e.g., inhibition of its cellular uptake or enhancement of its elimination from the body, could be promising techniques to prevent the cellular accumulation of OTA as well as its toxic action. Obviously, animal experiments are needed to decide if displacement of OTA from albumin and its increased elimination have a positive impact on attenuating OTA toxicity. However, it should be kept in mind that binding of OTA to albumin is strongly species-dependent; therefore, to find a proper animal model is essential. Based on these considerations, we suggest that further investigations of agents which are able to positively influence the toxicokinetics of OTA is a very important issue, and it might provide a more suitable way to prevent and/or treat the OTA-induced unpleasant consequences, especially in the endemic areas of increased OTA exposure.

Acknowledgments

The present scientific contribution is dedicated to the 650th anniversary of the foundation of the University of Pécs, Hungary.

Author Contributions

In this review article both authors contributed equally in the work and in writing the paper.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ATP
Adenosine triphosphate
BCRP
Breast Cancer Resistance Protein
BEN
Balkan Endemic Nephropathy
C8-dG
C8-deoxyguanosine DNA-adduct
CYP450
Cytochrom P450 enzyme family
DDAH
Dimethylarginine dimethylaminohydrolase
GSH
Glutathione (reduced)
GSSG
Glutathione (oxidized)
HSA
Human serum albumin
iNOS
Inducible Nitrogen Oxide Synthase
MRP2
Multidrug Resistance-associated Protein 2
NAC
N-acetyl cysteine
Nrf2
Nuclear factor-erythroid 2 p45-related factor 2
OAT
Organic Anion Transporter
OATP
Organic Anion-transporting Polypeptide
OTα
Ochratoxin α
OTA
Ochratoxin A
OTB
Ochratoxin B
OTC
Ochratoxin C
OTHQ
OTA-hydroquinone
OTQ
OTA-quinone
PEPCK
Phosphoenolpyruvate carboxykinase
ROS
Reactive oxygen species
SOD
Superoxide dismutase

References

  1. Ringot, D.; Chango, A.; Schneider, Y.J.; Larondelle, Y. Toxicokinetics and toxicodynamics of Ochratoxin A, an update. Chem. Biol. Interact. 2006, 159, 18–46. [Google Scholar] [CrossRef] [PubMed]
  2. Van Der Merwe, K.J.; Steyn, P.S.; Fourie, L. Part II. The constitution of Ochratoxins A, B, and C, metabolites of Aspergillus ochraceus wilh. J. Chem. Soc. 1965, 7083–7088. [Google Scholar] [CrossRef]
  3. Pohland, A.E.; Nesheim, S.; Friedman, L. Ochratoxin A: A review. Pure Appl. Chem. 1992, 64, 1029–1046. [Google Scholar] [CrossRef]
  4. Speijers, G.J.A.; van Egmond, H.P. Worldwide Ochratoxin A levels in food and feeds. In Human Ochratoxicosis and its Pathologies; Creppy, E.E., Castegnaro, M., Dirheimer, G., Eds.; John Libbey Eurotext: London, UK, 1993; Vol. 231, pp. 85–100. [Google Scholar]
  5. Krogh, P.; Hald, B.; Plestina, R.; Ceović, S. Balkan (endemic) nephropathy and foodborn Ochratoxin A: Preliminary results of a survey of foodstuffs. Acta Pathol. Microbiol. Scand. B 1977, 85, 238–240. [Google Scholar] [CrossRef] [PubMed]
  6. Pavlovic, M.; Plestina, R.; Krogh, P. Ochratoxin A contamination of foodstuffs in an area with Balkan (endemic) nephropathy. Acta Pathol. Microbiol. Scand. B 1979, 87, 243–246. [Google Scholar] [CrossRef] [PubMed]
  7. Petkova-Bocharova, T.; Castegnaro, M. Ochratoxin A contamination of cereals in an area of high incidence of Balkan endemic nephropathy in Bulgaria. Food Addit. Contam. 1985, 2, 267–270. [Google Scholar] [CrossRef] [PubMed]
  8. Petkova-Bocharova, T.; Castegnaro, M.; Michelon, J.; Maru, V. Ochratoxin A and other mycotoxins in cereals from an area of Balkan endemic nephropathy and urinary tract tumours in Bulgaria. In Mycotoxins, Endemic Nephropathy and Urinary Tract Tumours; Castegnaro, M., Plestina, R., Dirheimer, G., Chernozemsky, I.N., Bartsch, H., Eds.; IARC Scientific Publications: Lyon, France, 1991; pp. 83–87. [Google Scholar]
  9. Pfohl-Leszkowicz, A.; Petkova-Bocharova, T.; Chernozemsky, I.N.; Castegnaro, M. Balkan endemic nephropathy and associated urinary tract tumours: A review on aetiological causes and the potential role of mycotoxins. Food Addit. Contam. 2002, 19, 282–302. [Google Scholar] [CrossRef] [PubMed]
  10. Vrabcheva, T.; Usleber, E.; Dietrich, R.; Martlbauer, E. Co-occurrence of Ochratoxin A and citrinin in cereals from Bulgarian villages with a history of Balkan endemic nephropathy. J. Agric. Food Chem. 2000, 48, 2483–2488. [Google Scholar] [CrossRef] [PubMed]
  11. Bui-Klimke, T.; Wu, F. Evaluating weight of evidence in the mystery of balkan endemic nephropathy. Risk Anal. 2014, 34, 1688–1705. [Google Scholar] [CrossRef] [PubMed]
  12. Pavlović, N.M. Balkan endemic nephropathy—Current status and future perspectives. Clin. Kidney J. 2013, 6, 257–265. [Google Scholar] [CrossRef] [PubMed]
  13. Markovic, B.; Lebedev, S.; Djordjevic, M.; Arambasic, M. Endemic urinary tract cancer in Bulgaria, Yugoslavia and Romania: Etiology and pathogenesis. Med. Biol. Environ. 1976, 1–2. [Google Scholar]
  14. Chernozemsky, I.N.; Stoyanov, I.S.; Petkova-Bocharova, T.K.; Nicolov, I.G.; Draganov, I.V.; Stoichev, I.I.; Tanchev, Y.; Naidenov, D.; Kalcheva, N.D. Geographic correlation between the occurrence of endemic nephropathy and urinary tract tumours in vratza district, Bulgaria. Int. J. Cancer 1977, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  15. Pfohl-Leszkowicz, A. Ochratoxin A and aristolochic acid involvement in nephropathies and associated urothelial tract tumours. Arh. Hig. Rada Toksikol. 2009, 60, 465–483. [Google Scholar] [CrossRef] [PubMed]
  16. Duarte, S.C.; Pena, A.; Lino, C.M. Ochratoxin A non-conventional exposure sources—A review. Microchem. J. 2009, 93, 115–120. [Google Scholar] [CrossRef]
  17. Amézqueta, S.; González-Peñas, E.; Murillo-Arbizu, M.; López de Cerain, A. Ochratoxin A decontamination: A review. Food Control 2009, 20, 326–333. [Google Scholar] [CrossRef]
  18. Ciconová, P.; Laciková, A.; Máté, D. Prevention of Ochratoxin A contamination of food and Ochratoxin A detoxification by microorganisms—A review. Czech J. Food Sci. 2010, 28, 465–474. [Google Scholar]
  19. Pfohl-Leszkowicz, A.; Hadjeba-Medjdoub, K.; Ballet, N.; Schrickx, J.; Fink-Gremmels, J. Assessment and characterization of yeast-based products intended to mitigate ochratoxin exposure using in vitro and in vivo models. Food Addit. Contam. A 2015, 32, 604–616. [Google Scholar] [CrossRef] [PubMed]
  20. Radic, B.; Fuchs, R.; Peraica, M.; Lucic, A. Ochratloxin A in human sera in the area with endemic nephropathy in Croatia. Toxicol. Lett. 1997, 91, 105–109. [Google Scholar] [CrossRef]
  21. Peraica, M.; Domijan, A.M.; Fuchs, R.; Lucic, A.; Radic, B. The occurrence of Ochratoxin A in blood in general population of Croatia. Toxicol. Lett. 1999, 110, 105–112. [Google Scholar] [CrossRef]
  22. Özcelik, N.; Kosar, A.; Soysal, D. Ochratoxin A in human serum samples collected in Isparta-Turkey from healthy individuals and individuals suffering from different urinary disorders. Toxicol. Lett. 2001, 121, 9–13. [Google Scholar] [CrossRef]
  23. Lino, C.M.; Baeta, M.L.; Henri, M.; Dinis, A.M.P.; Pena, A.S.; Silveira, M.I.N. Levels of Ochratoxin A in serum from urban and rural Portuguese populations and estimation of exposure degree. Food Chem. Toxicol. 2008, 46, 879–885. [Google Scholar] [CrossRef] [PubMed]
  24. Perry, J.L.; Christensen, T.; Goldsmith, M.R.; Toone, E.J.; Beratan, D.N.; Simon, J.D. Binding of Ochratoxin A to human serum albumin stabilized by a protein-ligand ion pair. J. Phys. Chem. B 2003, 107, 7884–7888. [Google Scholar] [CrossRef]
  25. Poór, M.; Kunsági-Máté, S.; Bencsik, T.; Petrik, J.; Vladimir-Knežević, S.; Kőszegi, T. Flavonoid aglycones can compete with Ochratoxin A for human serum albumin: A new possible mode of action. Int. J. Biol. Macromol. 2012, 51, 279–283. [Google Scholar] [CrossRef] [PubMed]
  26. Poór, M.; Kunsági-Máté, S.; Matisz, G.; Li, Y.; Czibulya, Z.; Peles-Lemli, B.; Kőszegi, T. Interaction of alkali and alkaline earth ions with Ochratoxin A. J. Lumin. 2013, 135, 276–280. [Google Scholar]
  27. Hagelberg, S.; Hult, K.; Fuchs, R. Toxicokinetics of Ochratoxin A in several species and its plasma-binding properties. J. Appl. Toxicol. 1989, 9, 91–96. [Google Scholar] [CrossRef] [PubMed]
  28. Studer-Rohr, I.; Schlatter, J.; Dietrich, D.R. Kinetic parameters and intraindividual fluctuations of Ochratoxin A plasma levels in human. Arch. Toxicol. 2000, 74, 499–510. [Google Scholar] [CrossRef] [PubMed]
  29. Kumagai, S. Ochratoxin A: Plasma concentration and excretion into bile and urine in albumin-deficient rats. Food Chem. Toxicol. 1985, 23, 941–943. [Google Scholar] [CrossRef]
  30. Poór, M.; Kunsági-Máté, S.; Czibulya, Z.; Li, Y.; Peles-Lemli, B.; Petrik, J.; Vladimir-Knežević, S.; Kőszegi, T. Fluorescence spectroscopic investigation of competitive interactions between Ochratoxin A and 13 drug molecules for binding to human serum albumin. Luminescence 2013, 28, 726–733. [Google Scholar] [CrossRef] [PubMed]
  31. Poór, M.; Li, Y.; Matisz, G.; Kiss, L.; Kunsági-Máté, S.; Kőszegi, T. Quantitation of species differences in albumin-ligand interactions for bovine, human and rat serum albumins using fluorescence spectroscopy: A test case with some Sudlow's site I ligands. J. Lumin. 2014, 145, 767–773. [Google Scholar]
  32. Li, Y.; Czibulya, Z.; Poór, M.; Lecomte, S.; Kiss, L.; Harte, E.; Kőszegi, T.; Kunsági-Máté, S. Thermodynamic study of the effects of ethanol on the interaction of Ochratoxin A with human serum albumin. J. Lumin. 2014, 148, 18–25. [Google Scholar] [CrossRef]
  33. Castegnaro, M.; Canadas, D.; Vrabcheva, T.; Petkova-Bocharova, T.; Chernozemsky, I.N.; Pfohl-Leszkowicz, A. Balkan endemic nephropathy: Role of Ochratoxins A through biomarkers. Mol. Nutr. Food Res. 2006, 50, 519–529. [Google Scholar] [CrossRef] [PubMed]
  34. Pfohl-Leszkowicz, A.; Manderville, R.A. Ochratoxin A: An overview on toxicity and carcinogenicity in animals and humans. Mol. Nutr. Food Res. 2007, 51, 61–99. [Google Scholar] [CrossRef] [PubMed]
  35. Manderville, R.A. A case for the genotoxicity of Ochratoxin A by bioactivation and covalent DNA adduction. Chem. Res. Toxicol. 2005, 18, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  36. Turesky, R.J. Perspective: Ochratoxin A is not a genotoxic carcinogen. Chem. Res. Toxicol. 2005, 18, 1082–1090. [Google Scholar] [CrossRef] [PubMed]
  37. Manderville, R.; Pfohl-Leszkowicz, A. Bioactivation and DNA adduction as a rationale for Ochratoxin A carcinogenesis. World Mycotoxin J. 2008, 1, 357–367. [Google Scholar] [CrossRef]
  38. Pfohl-Leszkowicz, A.; Manderville, R.A. An update on direct genotoxicity as a molecular mechanism of Ochratoxin A carcinogenicity. Chem. Res. Toxicol. 2012, 25, 252–262. [Google Scholar] [CrossRef] [PubMed]
  39. Hadjeba-Medjdoub, K.; Tozlovanu, M.; Pfohl-Leszkowicz, A.; Frenette, C.; Paugh, R.J.; Manderville, R.A. Structure-activity relationships imply different mechanisms of action for Ochratoxin A-mediated cytotoxicity and genotoxicity. Chem. Res. Toxicol. 2012, 25, 181–190. [Google Scholar] [CrossRef] [PubMed]
  40. El Khoury, A.; Atoui, A. Ochratoxin A: General overview and actual molecular status. Toxins 2010, 2, 461–493. [Google Scholar] [CrossRef] [PubMed]
  41. IARC. Ochratoxin A. Monographs on the Evaluation of Carcinogenic Risks to Humans. In Some Naturally Occurring Substances: Food Items and Constituents, Heterocyclic Aromatic Amines and Mycotoxins; International Agency for Research on Cancer: Lyon, France, 1993; pp. 489–521. [Google Scholar]
  42. Boorman, G. NTP Technical Report on the Toxicology and Carcinogenesis Studies of Ochratoxin A (CAS No. 303-47-9) in F344/N Rats (Gavage Studies); NIH Publication No. 89-2813; U.S. Department of Health and Human Services, National Institutes of Health: Research Triangle Park, NC, USA, 1989.
  43. Chernozemsky, I.N. Balkan endemic nephropathy and the associated tumours of the urinary system: A summary of epidemiological features in Bulgaria. IARC Sci. Publ. 1991, 115, 3–4. [Google Scholar] [PubMed]
  44. Hassen, W.; Abid-Essafi, S.; Achour, A.; Guezzah, N.; Zakhama, A.; Ellouz, F.; Creppy, E.E.; Bacha, H. Karyomegaly of tubular kidney cells in human chronic interstitial nephropathy in Tunisia: Respective role of Ochratoxin A and possible genetic predisposition. Hum. Exp. Toxicol. 2004, 23, 339–346. [Google Scholar] [CrossRef] [PubMed]
  45. Cui, J.; Xing, L.; Li, Z.; Wu, S.; Wang, J.; Liu, J.; Wang, J.; Yan, X.; Zhang, X. Ochratoxin A induces G(2) phase arrest in human gastric epithelium GES-1 cells in vitro. Toxicol. Lett. 2010, 193, 152–158. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, J.; Wu, S.; Shen, H.; Cui, J.; Wang, Y.; Xing, L.; Wang, J.; Yan, X.; Zhang, X. Ochratoxin A induces DNA damage and G2 phase arrest in human esophageal epithelium Het-1A cells in vitro. J. Toxicol. Sci. 2015, 40, 657–665. [Google Scholar] [CrossRef] [PubMed]
  47. Schwartz, G.G. Hypothesis: Does Ochratoxin A cause testicular cancer? Cancer Causes Control 2002, 13, 91–100. [Google Scholar] [CrossRef] [PubMed]
  48. Pohland, A.E.; Schuller, P.L.; Steyn, P.S. Physicochemical data for some selected mycotoxins. Pure Appl. Chem. 1982, 54, 2219–2284. [Google Scholar] [CrossRef]
  49. Vidal, A.; Sanchis, V.; Ramos, A.J.; Marín, S. Thermal stability and kinetics of degradation of deoxynivalenol, deoxynivalenol conjugates and Ochratoxin A during baking of wheat bakery products. Food Chem. 2015, 178, 276–286. [Google Scholar] [CrossRef] [PubMed]
  50. Trivedi, A.B.; Doi, E.; Kitabatake, N. Detoxification of Ochratoxin A on heating under acidic and alkaline conditions. Biosci. Biotech. Biochem. 1992, 56, 741–745. [Google Scholar] [CrossRef]
  51. Boudra, H.; Le Bars, P.; Le Bars, J. Thermostability of Ochratoxin A in wheat under two moisture conditions. Appl. Environ. Microbiol. 1995, 61, 1156–1158. [Google Scholar] [PubMed]
  52. Blanc, M.; Pittet, A.; Munoz-Box, R.; Viani, R. Behavior of Ochratoxin A during green coffee roasting and soluble coffee manufacture. J. Agric. Food Chem. 1998, 46, 673–675. [Google Scholar] [CrossRef] [PubMed]
  53. Van der Stegen, G.H.; Essens, P.J.; van der Lijn, J. Effect of roasting conditions on reduction of Ochratoxin A in coffee. J. Agric. Food Chem. 2001, 49, 4713–4715. [Google Scholar] [CrossRef] [PubMed]
  54. Cramer, B.; Königs, M.; Humpf, H.U. Identification and in vitro cytotoxicity of Ochratoxin A degradation products formed during coffee roasting. J. Agric. Food Chem. 2008, 56, 5673–5681. [Google Scholar] [CrossRef] [PubMed]
  55. Bellver Soto, J.; Fernández-Franzón, M.; Ruiz, M.J.; Juan-García, A. Presence of Ochratoxin A (OTA) mycotoxin in alcoholic drinks from southern European countries: Wine and beer. J. Agric. Food Chem. 2014, 62, 7643–7651. [Google Scholar] [CrossRef] [PubMed]
  56. Malir, F.; Ostry, V.; Pfohl-Leszkowicz, A.; Toman, J.; Bazin, I.; Roubal, T. Transfer of Ochratoxin A into tea and coffee beverages. Toxins 2014, 6, 3438–3453. [Google Scholar] [CrossRef] [PubMed]
  57. Flores-Flores, M.E.; Lizarraga, E.; López de Cerain, A.; González-Penas, E. Presence of mycotoxins in animal milk: A review. Food Control 2015, 53, 163–176. [Google Scholar] [CrossRef]
  58. Shim, W.B.; Ha, K.S.; Kim, M.G.; Kim, J.S.; Chung, D.H. Evaluation of the transfer rate of Ochratoxin A to decoctions of herbal medicines. Food Sci. Biotechnol. 2014, 23, 2103–2108. [Google Scholar] [CrossRef]
  59. Chen, A.J.; Jiao, X.; Hu, Y.; Lu, X.; Gao, W. Mycobiota and Mycotoxins in traditional medicinal seeds from China. Toxins 2015, 7, 3858–3875. [Google Scholar] [CrossRef] [PubMed]
  60. Veprikova, Z.; Zachariasova, M.; Dzuman, Z.; Zachariasova, A.; Fenclova, M.; Slavikova, P.; Vaclavikova, M.; Mastovska, K.; Hengst, D.; Hajslova, J. Mycotoxins in plant-based dietary supplements: Hidden health risk for consumers. J. Agric. Food Chem. 2015, 63, 6633–6643. [Google Scholar] [CrossRef] [PubMed]
  61. Solfrizzo, M.; Piemontese, L.; Gambacorta, L.; Zivoli, R.; Longobardi, F. Food coloring agents and plant food supplements derived from Vitis vinifera: A new source of human exposure to Ochratoxin A. J. Agric. Food Chem. 2015, 63, 3609–3614. [Google Scholar] [CrossRef] [PubMed]
  62. Ostry, V.; Malir, F.; Ruprich, J. Producers and important dietary sources of Ochratoxin A and citrinin. Toxins 2013, 5, 1574–1586. [Google Scholar] [CrossRef] [PubMed]
  63. Jeswal, P.; Kumar, D. Mycobiota and natural incidence of Aflatoxins, Ochratoxin A, and Citrinin in Indian spices confirmed by LC-MS/MS. Int. J. Microbiol. 2015, 2015, 242486. [Google Scholar] [CrossRef] [PubMed]
  64. Do, K.H.; An, T.J.; Oh, S.K.; Moon, Y. Nation-based occurrence and endogenous biological reduction of mycotoxins in medicinal herbs and spices. Toxins 2015, 7, 4111–4130. [Google Scholar] [CrossRef] [PubMed]
  65. Mata, A.T.; Ferreira, J.P.; Oliveira, B.R.; Batoréu, M.C.; Barreto Crespo, M.T.; Pereira, V.J.; Bronze, M.R. Bottled water: Analysis of mycotoxins by LC-MS/MS. Food Chem. 2015, 176, 455–464. [Google Scholar] [CrossRef] [PubMed]
  66. Galtier, P.; Alvinerie, M.; Charpenteau, J.L. The pharmacokinetic profiles of Ochratoxin A in pigs, rabbits and chickens. Food Cosmet. Toxicol. 1981, 19, 735–738. [Google Scholar] [CrossRef]
  67. Galtier, P. Contribution of pharmacokinetic studies to mycotoxicology-Ochratoxin A. Vet. Sci. Commun. 1978, 1, 349–358. [Google Scholar] [CrossRef]
  68. Kumagai, S.; Aibara, K. Intestinal absorption and secretion of Ochratoxin A in the rat. Toxicol. Appl. Pharmacol. 1982, 64, 94–102. [Google Scholar] [CrossRef]
  69. Roth, A.; Chakor, K.; Creppy, E.E.; Kane, A.; Roschenthaler, R.; Dirheimer, G. Evidence for an enterohepatic circulation of Ochratoxin A in mice. Toxicology 1988, 48, 293–308. [Google Scholar] [CrossRef]
  70. Berger, V.; Gabriel, A.F.; Sergent, T.; Trouet, A.; Larondelle, Y.; Schneider, Y.J. Interaction of Ochratoxin A with human intestinal Caco-2 cells: Possible implication of a multidrug resistance-associated protein (MRP2). Toxicol. Lett. 2003, 140–141, 465–476. [Google Scholar] [CrossRef]
  71. Schrickx, J.; Lektarau, Y.; Fink-Gremmels, J. Ochratoxin A secretion by ATP-dependent membrane transporters in Caco-2 cells. Arch. Toxicol. 2006, 80, 243–249. [Google Scholar] [CrossRef] [PubMed]
  72. Maresca, M.; Mahfoud, R.; Pfohl-Leszkowicz, A.; Fantini, J. The mycotoxin Ochratoxin A alters intestinal barrier and absorption functions but has no effect on chloride secretion. Toxicol. Appl. Pharmacol. 2001, 176, 54–63. [Google Scholar] [CrossRef] [PubMed]
  73. Il’ichev, Y.V.; Perry, J.L.; Simon, J.D. Interaction of Ochratoxin A with human serum albumin. Preferential binding of the dianion and pH effects. J. Phys. Chem. B 2002, 106, 452–459. [Google Scholar] [CrossRef]
  74. Il’ichev, Y.V.; Perry, J.L.; Rüker, F.; Dockal, M.; Simon, J.D. Interaction of Ochratoxin A with human serum albumin. Binding sites localized by competitive interactions with the native protein and its recombinant fragments. Chem. Biol. Interact. 2002, 141, 275–293. [Google Scholar] [CrossRef]
  75. Perry, J.L.; Goldsmith, M.R.; Peterson, M.A.; Beratan, D.N. Structure of the Ochratoxin A binding site within human serum albumin. J. Phys. Chem. B 2004, 108, 16960–16964. [Google Scholar] [CrossRef]
  76. Dai, J.; Park, G.; Perry, J.L.; Il'ichev, Y.V.; Bow, D.A.; Pritchard, J.B.; Faucet, V.; Pfohl-Leszkowicz, A.; Manderville, R.A.; Simon, J.D. Molecular aspects of the transport and toxicity of Ochratoxin A. Acc. Chem. Res. 2004, 37, 874–881. [Google Scholar] [CrossRef] [PubMed]
  77. Il’ichev, Y.V.; Perry, J.L.; Simon, J.D. Interaction of Ochratoxin A with human serum albumin. A common binding site of Ochratoxin A and warfarin in subdomain IIA. J. Phys. Chem. B 2002, 106, 460–465. [Google Scholar] [CrossRef]
  78. Stojković, R.; Hult, K.; Gamulin, S.; Plestina, R. High affinity binding of Ochratoxin A to plasma constituents. Biochem. Int. 1984, 9, 33–38. [Google Scholar] [PubMed]
  79. Schwerdt, G.; Freudinger, R.; Silbernagl, S.; Gekle, M. Ochratoxin A—binding proteins in rat organs and plasma and in different cell lines of the kidney. Toxicology 1999, 135, 1–10. [Google Scholar] [CrossRef]
  80. Belmadani, A.; Tramu, G.; Betbeder, A.M.; Steyn, P.S.; Creppy, E.E. Regional selectivity to Ochratoxin A, distribution and cytotoxicity in rat brain. Arch. Toxicol. 1998, 72, 656–662. [Google Scholar] [CrossRef] [PubMed]
  81. Jung, K.Y.; Takeda, M.; Kim, D.K.; Tojo, A.; Narikawa, S.; Yoo, B.S.; Hosoyamada, M.; Cha, S.H.; Sekine, T.; Endou, H. Characterization of Ochratoxin A transport by human organic anion transporters. Life Sci. 2001, 69, 2123–2135. [Google Scholar] [CrossRef]
  82. Kontaxi, M.; Echkardt, U.; Hagenbuch, B.; Stieger, B.; Meier, P.J.; Petzinger, E. Uptake of the mycotoxin Ochratoxin A in liver cells occurs via the cloned organic anion transporting polypeptide. J. Pharmacol. Exp. Ther. 1996, 279, 1507–1513. [Google Scholar] [PubMed]
  83. Anzai, N.; Jutabha, P.; Endou, H. Molecular mechanism of Ochratoxin A transport in the kidney. Toxins 2010, 2, 1381–1398. [Google Scholar] [CrossRef] [PubMed]
  84. Takeuchi, A.; Masuda, S.; Saito, H.; Abe, T.; Inui, K. Multispecific substrate recognition of kidney-specific organic anion transporters OAT-K1 and OAT-K2. J. Pharmacol. Exp. Ther. 2001, 299, 261–267. [Google Scholar] [PubMed]
  85. Zlender, V.; Breljak, D.; Ljubojević, M.; Flajs, D.; Balen, D.; Brzica, H.; Domijan, A.M.; Peraica, M.; Fuchs, R.; Anzai, N.; et al. Low doses of Ochratoxin A upregulate the protein expression of organic anion transporters Oat1, Oat2, Oat3 and Oat5 in rat kidney cortex. Toxicol. Appl. Pharmacol. 2009, 239, 284–296. [Google Scholar] [CrossRef] [PubMed]
  86. Miraglia, M.; Brera, C.; Corneli, S.; Cava, E.; Montanino, G.; Miraglia, E. Occurence of Ochratoxin A (OTA) in Maternal Serum, Placenta and Funiculum. In Proceedings of the IX International IUPAC Symposium on Mycotoxins and Phycotoxins—Developments in Chemistry, Toxicology and Food Safety, Rome, Italy, 27–31 May 1996; Miraglia, M., van Egmond, H., Brera, C., Gilbert, J., Eds.; Alaken Inc.: Fort Collins, CO, USA; pp. 165–179.
  87. Cha, S.H.; Sekine, T.; Kusushura, H.; Yu, E.; Kim, J.Y.; Kim, D.K.; Sugiyama, Y.; Kanai, Y.; Endou, H. Molecular cloning and characterization of multispecific organic anion transporter 4 expressed in the placenta. J. Biol. Chem. 2000, 275, 4507–4512. [Google Scholar] [CrossRef] [PubMed]
  88. Heussner, A.H.; Bingle, L.E. Comparative Ochratoxin toxicity: A review of the available data. Toxins 2015, 7, 4253–4282. [Google Scholar] [CrossRef] [PubMed]
  89. Pitout, M.J. The hydrolysis of Ochratoxin A by some proteolytic enzymes. Biochem. Pharmacol. 1969, 18, 485–491. [Google Scholar] [CrossRef]
  90. Doster, R.C.; Sinnhuber, R.O. Comparative rate of hydrolysis of Ochratoxin A and B in vitro. Food Cosmet. Toxicol. 1972, 10, 389–394. [Google Scholar] [CrossRef]
  91. Hoehler, D.; Sudekum, K.H.; Wolfram, S.; Frohlich, A.A.; Marquardt, R.R. Metabolism and excretion of Ochratoxin A fed to sheep. J. Anim. Sci. 1999, 77, 1217–1223. [Google Scholar]
  92. Madhyastha, M.S.; Marquardt, R.R.; Frohlich, A.A. Hydrolysis of Ochratoxin A by the microbial activity of digesta in the gastrointestinal tract. Arch. Environ. Contam. Toxicol. 1992, 23, 468–472. [Google Scholar] [CrossRef] [PubMed]
  93. Xiao, H.; Madhyastha, S.; Marquardt, R.R.; Li, S.; Vodela, J.K.; Frohlich, A.A.; Kemppainen, W. Toxicity of OTA, its opened lactone form and several of its analogs: Structure-activity relationships. Toxicol. Appl. Pharmacol. 1996, 137, 182–192. [Google Scholar] [CrossRef] [PubMed]
  94. Stormer, F.C.; Pederson, J.I. Formation of (4R)- and (4S)-hydroxyochratoxin A from Ochratoxin A by rat liver microsomes. Appl. Environ. Microbiol. 1980, 39, 971–975. [Google Scholar] [PubMed]
  95. Stormer, F.C.; Storen, O.; Hansen, C.E.; Pedersen, J.I.; Hvistendahl, G.; Aarsen, A.J. Formation of (4R)- and (4S)-hydroxyochratoxin A from Ochratoxin A by liver microsomes from various species. Appl. Environ. Microbiol. 1981, 42, 1051–1056. [Google Scholar] [PubMed]
  96. Oster, T.; Jayyosi, Z.; Creppy, E.E.; El Amri, H.S.; Batt, A.M. Characterization of pig liver purified cytochrome P-450 isoenzymes for Ochratoxin A metabolism studies. Toxicol. Lett. 1991, 57, 203–214. [Google Scholar] [CrossRef]
  97. Hutchinson, R.D.; Steyn, P.S.; Thompson, D.L. The isolation and structure of 4-hydroxyochratoxin A and 7-carboxy-3,4-dihydro-8-hydroxy-3-methylisocoumarin from Penicillium viridicatum. Tetrahedron Lett. 1971, 43, 4033–4036. [Google Scholar] [CrossRef]
  98. Creppy, E.E.; Kern, D.; Steyn, P.S.; Vleggaar, R.; Roschenthaler, R.; Dirheimer, G. Comparative study on the effect of Ochratoxin A analogues on yeast aminoacyl-t-RNA synthetases and on the growth and protein synthesis of hepatoma cells. Toxicol. Lett. 1983, 19, 217–224. [Google Scholar] [CrossRef]
  99. Xiao, H.; Marquardt, R.R.; Abramson, R.R.; Frohlich, A.A. Metabolites of ochratoxins in rat urine and in a culture of Aspergillus ochraceus. Appl. Environ. Microbiol. 1996, 62, 648–655. [Google Scholar] [PubMed]
  100. Zepnik, H.; Pahler, A.; Schauer, U.; Dekant, W. Ochratoxin A-Induced tumor formation: Is there a role of reactive Ochratoxin A metabolites? Toxicol. Sci. 2001, 59, 59–67. [Google Scholar] [CrossRef] [PubMed]
  101. Pinelli, E.; El Adlouni, C.; Pipy, B.; Quartulli, F.; Pfohl-Leskzowicz, A. Roles of cyclooxygenase and lipoygenases in Ochratoxin A genotoxicity in human epithelial lung cells. Environ. Toxicol. Pharmacol. 1999, 7, 95–107. [Google Scholar] [CrossRef]
  102. El Adlouni, C.; Pinelli, E.; Azemar, B.; Zaoui, D.; Beaune, P.; Leszkowicz, A.P. Phenobarbital increases of DNA adduct and metabolites formed by Ochratoxin A: Role of CYP 2C9 and microsomal glutathione-S-transferase. Environ. Mol. Mutagen. 2000, 35, 123–131. [Google Scholar] [CrossRef]
  103. Yang, S.; Zhang, H.; de Saeger, S.; de Boevre, M.; Sun, F.; Zhang, S.; Cao, X.; Wang, Z. In vitro and in vivo metabolism of Ochratoxin A: A comparative study using ultra-performance liquid chromatography-quadrupole/time-of-flight hybrid mass spectrometry. Anal. Bioanal. Chem. 2015, 407, 3579–3589. [Google Scholar] [CrossRef] [PubMed]
  104. Ueno, Y. Biotransformation of mycotoxins in the reconstituted cytochrome P-450 system. Proc. Jpn. Assoc. Mycotoxicol. 1985, 22, 28–30. [Google Scholar] [CrossRef]
  105. Stormer, F.C.; Storen, O.; Hansen, C.E.; Pedersen, J.I.; Aasen, A.J. Formation of (4R)- and (4S)-4-hydroxyochratoxin A and 10-hydroxyochratoxin A from Ochratoxin A by rabbit liver microsomes. Appl. Environ. Microbiol. 1983, 45, 1183–1187. [Google Scholar] [PubMed]
  106. Fink-Gremmels, J.; Jahn, A.; Blom, M.J. Toxicity and metabolism of Ochratoxin A. Nat. Toxins 1995, 3, 214–220. [Google Scholar] [CrossRef]
  107. Simarro Doorten, A.Y.; Bull, S.; van der Doelen, M.A.; Fink-Gremmels, J. Metabolism-mediated cytotoxicity of Ochratoxin A. Toxicol. In Vitro 2004, 18, 271–277. [Google Scholar] [CrossRef] [PubMed]
  108. Omar, R.F.; Gelboin, H.V.; Rahimtula, A.D. Effect of cytochrome P450 induction on the metabolism and toxicity of Ochratoxin A. Biochem. Pharmacol. 1996, 51, 207–216. [Google Scholar] [CrossRef]
  109. Grosse, Y.; Monje, M.C.; Mace, K.; Pfeifer, A.; Pfohl-Leszkowicz, A. Use of bronchial epithelial cells expressing human cytochrome P450 for study on metabolism and genotoxicity of Ochratoxin A. In Vitro Toxicol. 1997, 10, 93–102. [Google Scholar]
  110. Chu, F.S.; Noh, I.; Chang, C.C. Structure requirements for Ochratoxin A intoxication. Life Sci. 1972, 11, 503–508. [Google Scholar]
  111. Grosse, Y.; Baudrimont, I.; Castegnaro, M.; Creppy, E.E.; Dirheimer, G.; Pfohl-Leskowicz, A. Ochratoxin A metabolites and DNA-adducts formation in monkey kidney cell. Chem. Biol. Interact. 1995, 95, 175–187. [Google Scholar] [CrossRef]
  112. Kasmuller, S.; Cavin, C.; Chakraborty, A.; Darroudi, F.; Majer, B.J.; Huber, W.W.; Ehrlich, V.A. Structurally related mycotoxins Ochratoxin A, Ochratoxin B, and citrinin differ in their genotoxic activities and in their mode of action in human-derived liver (HepG2) cells: Implication for Risk Assessment. Nutr. Cancer 2004, 50, 190–197. [Google Scholar] [CrossRef] [PubMed]
  113. Faucet-Marquis, V.; Pont, F.; Størmer, F.C.; Rizk, T.; Castegnaro, M.; Pfohl-Leszkowicz, A. Evidence of a new dechlorinated Ochratoxin A derivative formed in opossum kidney cell cultures after pretreatment by modulators of glutathione pathways: Correlation with DNA-adduct formation. Mol. Nutr. Food Res. 2006, 50, 530–542. [Google Scholar] [CrossRef] [PubMed]
  114. Gross-Steinmeyer, K.; Weymann, J.; Hege, H.G.; Metzler, M. Metabolism and lack of DNA reactivity of the mycotoxin Ochratoxin A in cultured and human primary hepatocytes. J. Agric. Food Chem. 2002, 50, 935–938. [Google Scholar] [CrossRef]
  115. Zepnik, H.; Volkel, W.; Dekant, W. Toxicokinetics of the mycotoxin Ochratoxin A in F 344 rats after oral administration. Toxicol. Appl. Pharmacol. 2003, 192, 36–44. [Google Scholar] [CrossRef]
  116. Malaveille, C.; Brun, G.; Bartsch, H. Structure-activity studies in E. coli strains on Ochratoxin A (OTA) and its analogues implicate a genotoxic free radical and a cytosolic thiol derivative as reactive metabolites. Mutat. Res. 1994, 307, 141–147. [Google Scholar] [CrossRef]
  117. Dai, J.; Park, G.; Wright, M.W.; Adams, M.; Akman, S.A.; Manderville, R.A. Detection and characterization of a glutathione conjugate of Ochratoxin A. Chem. Res. Toxicol. 2002, 15, 1581–1588. [Google Scholar] [CrossRef] [PubMed]
  118. Tozlovanu, M.; Canadas, D.; Pfohl-Leszkowicz, A.; Frenette, C.; Paugh, R.J.; Manderville, R.A. Glutathione conjugates of Ochratoxin A as biomarkers of exposure. Arh. Hig. Rada Toksikol. 2012, 63, 417–427. [Google Scholar] [CrossRef] [PubMed]
  119. Pfohl-Leszkowicz, A.; Pinelli, E.; Bartsch, H.; Mohr, U.; Castegnaro, M. Sex and strain differences in Ochratoxin A metabolism and DNA adduction in two strains of rats. Mol. Carcinog. 1998, 23, 76–83. [Google Scholar] [CrossRef]
  120. Petkova-Bocharova, T.; El Adlouni, C.; Faucet, V.; Pfohl-Leszkowicz, A.; Mantle, P. Analysis for DNA adducts, Ochratoxin A content and enzymes expression in kidneys of pigs exposed to mild experimental chronic ochratoxicosis. Facta Univ. Ser. Med. Biol. 2003, 10, 111–115. [Google Scholar]
  121. Pfohl-Leszkowicz, A.; Tozlovanu, M.; Manderville, R.; Peraica, M.; Castegnaro, M.; Stefanovic, V. New molecular and field evidences for the implication of mycotoxins but not aristolochic acid in human nephropathy and urinary tract tumor. Mol. Nutr. Food Res. 2007, 51, 1131–1146. [Google Scholar] [CrossRef] [PubMed]
  122. Gekle, M.; Silbernagl, S. The role of the proximal tubule in Ochratoxin A nephrotoxicity in vivo. Toxicodynamics and toxicokinetics aspects. Ren. Physiol. Biochem. 1994, 17, 40–49. [Google Scholar] [PubMed]
  123. Groves, C.E.; Morales, M.; Wright, S.H. Peritubular transport of Ochratoxin A in rabbit renal proximal tubules. J. Pharmacol. Exp. Ther. 1998, 284, 943–948. [Google Scholar] [PubMed]
  124. Kusushura, H.; Sekine, T.; Utsunomiya-Tate, N.M.; Tsuda, M.; Kojima, R.; Cha, S.H.; Sugiyama, Y.; Kanai, Y.; Endou, H. Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain. J. Biol. Chem. 1999, 274, 13675–13680. [Google Scholar] [CrossRef]
  125. Bahnemann, E.; Kerling, H.P.; Ensminger, S.; Scwerdt, G.; Silbernagl, S.; Gekle, M. Renal transepithelial secretion of Ochratoxin A in the non-filtering toad kidney. Toxicology 1997, 120, 11–17. [Google Scholar] [CrossRef]
  126. Leier, I.; Hummel-Eisenbeiss, J.; Cui, Y.; Keppler, D. ATP-dependent para-aminohippurate transport by apical multidrug resistance protein MRP2. Kidney Int. 2000, 57, 1636–1642. [Google Scholar] [CrossRef] [PubMed]
  127. Dahlman, A.; Dantzker, W.H.; Silbernagl, S.; Gekle, M. Detailed mapping of Ochratoxin A reabsorption along the rate nephron in vivo: The nephrotoxin can be reabsorbed in all nephron segments by different mechanisms. J. Pharmacol. Exp. Ther. 1998, 286, 157–162. [Google Scholar]
  128. Sreemannarayana, O.; Frohlich, A.A.; Vitti, T.G.; Marquardt, R.R.; Abramson, D.; Phillips, G.D. Studies of the tolerance and disposition of Ochratoxin A in young calves. J. Anim. Sci. 1988, 66, 1703–1711. [Google Scholar] [PubMed]
  129. Suzuki, S.; Satoh, T.; Yamakazi, M. The pharmacokinetics of Ochratoxin A in rats. Jpn. J. Pharmacol. 1977, 27, 735–744. [Google Scholar] [CrossRef] [PubMed]
  130. Nip, W.K.; Chu, F.S. The fate of Ochratoxin A in goats. J. Environ. Sci. Health B 1979, 14, 319–333. [Google Scholar] [CrossRef] [PubMed]
  131. Xiao, H.; Marquardt, R.R.; Frohlich, A.A.; Philips, G.D.; Vitti, T.G. Effect of hay and a grain diet on the rate of hydrolysis of Ochratoxin A in the rumen sheep. J. Anim. Sci. 1991, 69, 3706–3714. [Google Scholar] [PubMed]
  132. Fuchs, R.; Radic, B.; Peraica, M.; Hult, K.; Plestina, R. Enterohepatic circulation of Ochratoxin A in rats. Period. Biol. 1988, 90, 39–42. [Google Scholar]
  133. Gareis, M.; Martlbauer, E.; Bauer, J.; Gedek, B. Determination of Ochratoxin A in breast milk. Zeitschr. Lebensm. Undersch. Forsch. 1988, 186, 114–117. [Google Scholar] [CrossRef]
  134. Micco, C.; Miraglia, M.; Brera, C.; Corneli, S.; Ambruzi, A. Evaluation of Ochratoxin A level in human milk in Italy. Food Addit. Contam. 1995, 12, 351–354. [Google Scholar] [CrossRef] [PubMed]
  135. Skaug, M.A.; Helland, I.; Solvoll, K.; Saugstad, O.D. Presence of Ochratoxin A in human milk in relation to dietary intake. Food Addit. Contam. 2001, 18, 321–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Muñoz, K.; Blaszkewicz, M.; Campos, V.; Vega, M.; Degen, G.H. Exposure of infants to Ochratoxin A with breast milk. Arch. Toxicol. 2014, 88, 837–846. [Google Scholar] [CrossRef] [PubMed]
  137. Creppy, E.E.; Röschenthaler, R.; Dirheimer, G. Inhibition of protein synthesis in mice by Ochratoxin A and its prevention by phenylalanine. Food Chem. Toxicol. 1984, 22, 883–886. [Google Scholar] [CrossRef]
  138. Dirheimer, G.; Creppy, E.E. Mechanism of action of Ochratoxin A. IARC Sci. Publ. 1991, 115, 171–186. [Google Scholar] [PubMed]
  139. Bruinink, A.; Rasony, T.; Sidler, C. Reduction of Ochratoxin A toxicity by heat-induced epimerization. In vitro effects of ochratoxins on embryonic chick meningeal and other cell cultures. Toxicology 1997, 118, 205–210. [Google Scholar] [CrossRef]
  140. Bruinink, A.; Sidler, C. The neurotoxic effect of Ochratoxin A are reduced by protein binding but not affected byl-phenylalanine. Toxicol. Appl. Pharmacol. 1997, 146, 173–179. [Google Scholar] [CrossRef] [PubMed]
  141. McMaster, D.R.; Vedani, A. Ochratoxin A binding to phenlylalanine-tRNA synthetatse: Computational approach to the mechanism of ochratoxicoses and its antagonism. J. Med. Chem. 1999, 42, 3075–3086. [Google Scholar] [CrossRef] [PubMed]
  142. Creppy, E.E.; Chakor, K.; Fisher, M.; Dirheimer, G. The mycotoxin Ochratoxin A is a substrate for phenylalanine hydroxylase in isolated rat hepatocytes and in vivo. Arch. Toxicol. 1990, 64, 279–284. [Google Scholar] [CrossRef] [PubMed]
  143. Vettorazzi, A.; van Delft, J.; de Cerain, A.L. A review on Ochratoxin A transcriptomic studies. Food Chem. Toxicol. 2013, 59, 766–783. [Google Scholar] [CrossRef] [PubMed]
  144. Poór, M.; Veres, B.; Jakus, P.B.; Antus, C.; Montskó, G.; Zrínyi, Z.; Vladimir-Knežević, S.; Petrik, J.; Kőszegi, T. Flavonoid diosmetin increases ATP levels in kidney cells and relieves ATP depleting effect of Ochratoxin A. J. Photochem. Photobiol. B 2014, 132, 1–9. [Google Scholar] [CrossRef] [PubMed]
  145. Aleo, M.D.; Wyat, R.D.; Schnellmann, R.G. Mitochondrial dysfunction is an early event in Ochratoxin A but not oosporein toxicity to rat renal proximal tubules. Toxicol. Appl. Pharmacol. 1991, 107, 73–80. [Google Scholar] [CrossRef]
  146. Meisner, H.; Meisner, P. Ochratoxin A an in vivo inhibitor of renal phosphoenolpyruvate carboxykinase. Arch. Biochem. Biophys. 1981, 208, 146–153. [Google Scholar] [CrossRef]
  147. Meisner, H.; Krogh, P. Phosphoenolpyruvate carboxykinase as a selective indicator of Ochratoxin A induced nephropathy. Dev. Toxicol. Environ. Sci. 1986, 14, 199–206. [Google Scholar] [PubMed]
  148. Meisner, H.; Polsinelli, P. Changes of renal mRNA species abundance by Ochratoxin A. Biochem. Pharmacol. 1986, 35, 661–665. [Google Scholar] [CrossRef]
  149. Thekkumkarra, T.J.; Patel, M.S. Ochratoxin A decrease the activity of phosphoenolpyruvate carboxykinase and its mRNA content in primary cultures of rat kidney proximal convoluted tubular cells. Biochem. Biophys. Res. Commun. 1989, 162, 916–920. [Google Scholar] [CrossRef]
  150. Suzuki, S.; Kozuka, Y.; Satoh, T.; Yamazaki, M. Studies on the nephrotoxicity of Ochratoxin A in rats. Toxicol. Appl. Pharmacol. 1975, 34, 479–490. [Google Scholar] [CrossRef]
  151. Meisner, H.; Chan, S. Ochratoxin A, an inhibitor of mitochondrial transport systems. Biochemistry 1974, 13, 2795–2800. [Google Scholar] [CrossRef] [PubMed]
  152. Wei, Y.H.; Lu, T.N.; Wei, R.D. Effect of Ochratoxin A on rat liver mitochondrial respiration and oxidative phosphorilation. Toxicology 1985, 36, 119–123. [Google Scholar] [CrossRef]
  153. Manderville, R.A.; Pfohl-Leszkowicz, A. Genotoxicity of chlorophenols and Ochratoxin A. Adv. Mol. Toxicol. 2006, 1, 73–118. [Google Scholar]
  154. Waidyanatha, S.; Lin, P.H.; Rappaport, S.M. Characterization of chlorinated adducts of hemoglobin and albumin following administration of pentachlorophenol to rats. Chem. Res. Toxicol. 1996, 9, 647–653. [Google Scholar] [CrossRef] [PubMed]
  155. Vaidyanathan, V.G.; Villalta, P.W.; Sturla, S.J. Nucleobase-dependent reactivity of a quinone metabolite of pentachlorophenol. Chem. Res. Toxicol. 2007, 20, 913–919. [Google Scholar] [CrossRef] [PubMed]
  156. Tozlovanu, M.; Faucet-Marquis, V.; Pfohl-Leszkowicz, A.; Manderville, R.A. Genotoxicity of the hydroquinone metabolite of Ochratoxin A: Structure-activity relationships for covalent DNA adduction. Chem. Res. Toxicol. 2006, 19, 1241–1247. [Google Scholar] [CrossRef] [PubMed]
  157. Gillman, I.G.; Clark, T.N.; Manderville, R.A. Oxidation of Ochratoxin A by an Fe-porphyrin system: Model for enzymatic activation and DNA cleavage. Chem. Res. Toxicol. 1999, 12, 1066–1076. [Google Scholar] [CrossRef] [PubMed]
  158. Reljic, Z.; Zlatovic, M.; Savic-Radojevic, A.; Pekmezovic, T.; Djukanovic, L.; Matic, M.; Pljesa-Ercegovac, M.; Mimic-Oka, J.; Opsenica, D.; Simic, T. Is increased susceptibility to Balkan endemic nephropathy in carriers of common GSTA1 (*A/*B) polymorphism linked with the catalytic role of GSTA1 in Ochratoxin A biotransformation? Serbian case control study and in silico analysis. Toxins 2014, 6, 2348–2362. [Google Scholar] [CrossRef] [PubMed]
  159. Mally, A.; Zepnik, H.; Wanek, P.; Eder, E.; Dingley, K.; Ihmels, H.; Völkel, W.; Dekant, W. Ochratoxin A: Lack of formation of covalent DNA adducts. Chem. Res. Toxicol. 2004, 17, 234–242. [Google Scholar] [CrossRef] [PubMed]
  160. Samokyszyn, V.M.; Freeman, J.P.; Maddipati, K.R.; Lloyd, R.V. Peroxidase-catalyzed oxidation of pentachlorophenol. Chem. Res. Toxicol. 1995, 8, 349–355. [Google Scholar] [CrossRef] [PubMed]
  161. El Adlouni, C.; Pinelli, E.; Azémar, B.; Zaoui, D.; Beaune, P.; Pfohl-Leszkowicz, A. Role of CYP 2C and microsomal glutathione-S-transferase in modulating susceptibility to Ochratoxin A genotoxicity. Environ. Mol. Mutagen. 2000, 35, 123–131. [Google Scholar] [CrossRef]
  162. Stoyanovosky, D.A.; Goldman, R.; Jonnalagadda, S.S.; Day, B.W.; Claycamp, H.G.; Kagan, V.E. Detection and characterization of the electron paramagnetic resonance-silent glutathionyl-5,5-dimethyl-1-pyrroline N-oxide adduct derived from redox cycling of phenoxyl radicals in model systems and HL-60 cells. Arch. Biochem. Biophys. 1996, 330, 3–11. [Google Scholar] [CrossRef] [PubMed]
  163. Murray, A.R.; Kisin, E.; Castranova, V.; Kommineni, C.; Gunther, M.R.; Shvedova, A.A. Phenol-induced in vivo oxidative stress in skin: Evidence for enhanced free radical generation, thiol oxidation, and antioxidant depletion. Chem. Res. Toxicol. 2007, 20, 1769–1777. [Google Scholar] [CrossRef] [PubMed]
  164. Kamp, H.G.; Eisenbrand, G.; Schlatter, J.; Würth, K.; Janzowski, C. Ochratoxin A: Induction of (oxidative) DNA damage, cytotoxicity and apoptosis in mammalian cell lines and primary cells. Toxicology 2005, 206, 413–425. [Google Scholar] [CrossRef] [PubMed]
  165. Dai, J.; Wright, M.W.; Manderville, R.A. An oxygen-bonded C8-deoxyguanosine nucleoside adduct of pentachlorophenol by peroxidase activation: Evidence for ambident C8 reactivity by phenoxyl radicals. Chem. Res. Toxicol. 2003, 16, 817–821. [Google Scholar] [CrossRef] [PubMed]
  166. Dai, J.; Sloat, A.L.; Wright, M.W.; Manderville, R.A. Role of phenoxyl radicals in DNA adduction by chlorophenol xenobiotics following peroxidase activation. Chem. Res. Toxicol. 2005, 18, 771–779. [Google Scholar] [CrossRef] [PubMed]
  167. Mohn, W.W.; Kennedy, K.J. Reductive dehalogenation of chlorophenols by Desulfomonile tiedjei DCB-1. Appl. Environ. Microbiol. 1992, 58, 1367–1370. [Google Scholar] [PubMed]
  168. Lawson, T.; Gannett, P.M.; Yau, W.M.; Dalal, N.S.; Toth, B. Different patterns of mutagenicity of arenediazonium ions in V79 cells and Salmonella typhimurium TA102: Evidence for different mechanisms of action. J. Agric. Food Chem. 1995, 43, 2627–2635. [Google Scholar] [CrossRef]
  169. Hiramoto, K.; Kaku, M.; Sueyoshi, A.; Fujise, M.; Kikugawa, K. DNA base and deoxyribose modification by the carbon-centered radical generated from 4-(hydroxymethyl)-benzenediazonium salt, a carcinogen in mushroom. Chem. Res. Toxicol. 1995, 8, 356–362. [Google Scholar] [CrossRef] [PubMed]
  170. Gannett, P.M.; Powell, J.H.; Rao, R.; Shi, X.; Lawson, T.; Kolar, C.; Toth, B. C8-Arylguanine and C8-aryladenine formation in calf thymus DNA from arenediazonium ions. Chem. Res. Toxicol. 1999, 12, 297–304. [Google Scholar] [CrossRef] [PubMed]
  171. Dai, J.; Wright, M.W.; Manderville, R.A. Ochratoxin a forms a carbon-bonded C8-deoxyguanosine nucleoside adduct: Implications for C8 reactivity by a phenolic radical. J. Am. Chem. Soc. 2003, 125, 3716–3717. [Google Scholar] [CrossRef] [PubMed]
  172. Il'ichev, Y.V.; Perry, J.L.; Manderville, R.A.; Chignell, C.F.; Simon, J.D. The pH-dependent primary photoreactions of Ochratoxin A. J. Phys. Chem. B 2001, 105, 11369–11376. [Google Scholar] [CrossRef]
  173. Faucet, V.; Pfohl-Leszkowicz, A.; Dai, J.; Castegnaro, M.; Manderville, R.A. Evidence for covalent DNA adduction by Ochratoxin A following chronic exposure to rat and subacute exposure to pig. Chem. Res. Toxicol. 2004, 17, 1289–1296. [Google Scholar] [CrossRef] [PubMed]
  174. Pfohl-Leszkowicz, A.; Gabryelski, W.; Manderville, R.A. Formation of 2′-deoxyguanosine-carbon 8-bound Ochratoxin A adduct in rat kidney DNA. Mol. Nutr. Food Res. 2009, 53, 154–155. [Google Scholar] [CrossRef] [PubMed]
  175. Pfohl-Leszkowicz, A.; Castegnaro, M. Further arguments in favour of direct covalent binding of Ochratoxin A (OTA) after metabolic biotransformation. Food Addit. Contam. 2005, 22 (Suppl. 1), 75–87. [Google Scholar] [CrossRef] [PubMed]
  176. Mantle, P.G.; Faucet-Marquis, V.; Manderville, R.A.; Squillaci, B.; Pfohl-Leszkowicz, A. Structures of covalent adducts between DNA and Ochratoxin A: A new factor in debate about genotoxicity and human risk assessment. Chem. Res. Toxicol. 2010, 23, 89–98. [Google Scholar] [CrossRef] [PubMed]
  177. Mally, A.; Pepe, G.; Ravoori, S.; Fiore, M.; Gupta, R.C.; Dekant, W.; Mosesso, P. Ochratoxin A causes DNA damage and cytogenetic effects but no DNA adducts in rats. Chem. Res. Toxicol. 2005, 18, 1253–1261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Delatour, T.; Mally, A.; Richoz, J.; Ozden, S.; Dekant, W.; Ihmels, H.; Otto, D.; Gasparutto, D.; Marin-Kuan, M.; Schilter, B.; et al. Absence of 2′-deoxyguanosine-carbon 8-bound Ochratoxin A adduct in rat kidney DNA monitored by isotope dilution LC-MS/MS. Mol. Nutr. Food Res. 2008, 52, 472–482. [Google Scholar] [CrossRef] [PubMed]
  179. Mally, A. Ochratoxin A and mitotic disruption: Mode of action analysis of renal tumor formation by Ochratoxin A. Toxicol. Sci. 2012, 127, 315–330. [Google Scholar] [CrossRef] [PubMed]
  180. Stiborová, M.; Bárta, F.; Levová, K.; Hodek, P.; Frei, E.; Arlt, V.M.; Schmeiser, H.H. The influence of Ochratoxin A on DNA adduct formation by the carcinogen aristolochic acid in rats. Arch. Toxicol. 2015, 89, 2141–2158. [Google Scholar] [CrossRef] [PubMed]
  181. Sorrenti, V.; di Giacomo, C.; Acquaviva, R.; Barbagallo, I.; Bognanno, M.; Galvano, F. Toxicity of Ochratoxin A and its modulation by antioxidants: A review. Toxins 2013, 5, 1742–1766. [Google Scholar] [CrossRef] [PubMed]
  182. Rahimtula, A.D.; Béréziat, J.C.; Bussacchini-Griot, V.; Bartsch, H. Lipid peroxidation as a possible cause of Ochratoxin A toxicity. Biochem. Pharmacol. 1988, 37, 4469–4477. [Google Scholar] [CrossRef]
  183. Omar, R.F.; Hasinoff, B.B.; Mejilla, F.; Rahimtula, A.D. Mechanism of Ochratoxin A stimulated lipid peroxidation. Biochem. Pharmacol. 1990, 40, 1183–1191. [Google Scholar] [CrossRef]
  184. Ranaldi, G.; Caprini, V.; Sambuy, Y.; Perozzi, G.; Murgia, C. Intracellular zinc stores protect the intestinal epithelium from Ochratoxin A toxicity. Toxicol. In Vitro 2009, 23, 1516–1521. [Google Scholar] [CrossRef] [PubMed]
  185. Zheng, J.; Zhang, Y.; Xu, W.; Luo, Y.; Hao, J.; Shen, X.L.; Yang, X.; Li, X.; Huang, K. Zinc protects HepG2 cells against the oxidative damage and DNA damage induced by Ochratoxin A. Toxicol. Appl. Pharmacol. 2013, 268, 123–131. [Google Scholar] [CrossRef] [PubMed]
  186. Poór, M.; Kuzma, M.; Matisz, G.; Li, Y.; Perjési, P.; Kunsági-Máté, S.; Kőszegi, T. Further aspects of Ochratoxin A-cation interactions: Complex formation with zinc ions and a novel analytical application of Ochratoxin A-magnesium interaction in the HPLC-FLD system. Toxins 2014, 6, 1295–1307. [Google Scholar] [CrossRef] [PubMed]
  187. Marin-Kuan, M.; Nestler, S.; Verguet, C.; Bezençon, C.; Piguet, D.; Mansourian, R.; Holzwarth, J.; Grigorov, M.; Delatour, T.; Mantle, P.; et al. A toxicogenomics approach to identify new plausible epigenetic mechanisms of Ochratoxin A carcinogenicity in rat. Toxicol. Sci. 2006, 89, 120–134. [Google Scholar] [CrossRef] [PubMed]
  188. Cavin, C.; Delatour, T.; Marin-Kuan, M.; Holzhäuser, D.; Higgins, L.; Bezençon, C.; Guignard, G.; Junod, S.; Richoz-Payot, J.; Gremaud, E.; et al. Reduction in antioxidant defenses may contribute to Ochratoxin A toxicity and carcinogenicity. Toxicol. Sci. 2007, 96, 30–39. [Google Scholar] [CrossRef] [PubMed]
  189. Boesch-Saadatmandi, C.; Loboda, A.; Jozkowicz, A.; Huebbe, P.; Blank, R.; Wolffram, S.; Dulak, J.; Rimbach, G. Effect of Ochratoxin A on redox-regulated transcription factors, antioxidant enzymes and glutathione-S-transferase in cultured kidney tubulus cells. Food Chem. Toxicol. 2008, 46, 2665–2671. [Google Scholar] [CrossRef] [PubMed]
  190. Limonciel, A.; Jennings, P. A review of the evidence that Ochratoxin A is an Nrf2 inhibitor: Implications for nephrotoxicity and renal carcinogenicity. Toxins 2014, 6, 371–379. [Google Scholar] [CrossRef] [PubMed]
  191. Cavin, C.; Delatour, T.; Marin-Kuan, M.; Fenaille, F.; Holzhäuser, D.; Guignard, G.; Bezençon, C.; Piguet, D.; Parisod, V.; Richoz-Payot, J.; et al. Ochratoxin A-mediated DNA and protein damage: Roles of nitrosative and oxidative stresses. Toxicol. Sci. 2009, 110, 84–94. [Google Scholar] [CrossRef] [PubMed]
  192. Qi, X.; Yu, T.; Zhu, L.; Gao, J.; He, X.; Huang, K.; Luo, Y.; Xu, W. Ochratoxin A induces rat renal carcinogenicity with limited induction of oxidative stress responses. Toxicol. Appl. Pharmacol. 2014, 280, 543–549. [Google Scholar] [CrossRef] [PubMed]
  193. Taniai, E.; Yafune, A.; Nakajima, M.; Hayashi, S.M.; Nakane, F.; Itahashi, M.; Shibutani, M. Ochratoxin A induces karyomegaly and cell cycle aberrations in renal tubular cells without relation to induction of oxidative stress responses in rats. Toxicol. Lett. 2014, 224, 64–72. [Google Scholar] [CrossRef] [PubMed]
  194. Gekle, M.; Schwerdt, G.; Freudinger, R.; Mildenberg, S.; Wilflingseder, D.; Pollack, V.; Dander, M.; Schramek, H. Ochratoxin A induces JNK activation and apoptosis in MDKC-C7 cells at nanomolar concentrations. J. Pharmacol. Exp. Ther. 2000, 293, 837–844. [Google Scholar] [PubMed]
  195. Lühe, A.; Hildebrandt, H.; Bach, U.; Dingerman, T.; Ahr, H.J. A new approach to studying Ochratoxin A (OTA)-induced nephrotoxicity: Expressing profiling in vivo and in vitro employing cDNA microarrays. Toxicol. Sci. 2003, 73, 315–328. [Google Scholar] [CrossRef] [PubMed]
  196. Kuroda, K.; Hibi, D.; Ishii, Y.; Yokoo, Y.; Takasu, S.; Kijima, A.; Matsushita, K.; Masumura, K.; Kodama, Y.; Yanai, T.; et al. Role of p53 in the progression from ochratoxin A-induced DNA damage to gene mutations in the kidneys of mice. Toxicol. Sci. 2015, 144, 65–76. [Google Scholar] [CrossRef] [PubMed]
  197. Horvath, A.; Upham, B.L.; Ganev, V.; Trosko, J.E. Determination of the epigenetic effects of ochratoxin in a human kidney and a rat liver epithelial cell line. Toxicon 2002, 40, 273–282. [Google Scholar] [CrossRef]
  198. Sauvant, C.; Holzinger, H.; Gekle, M. The nephrotoxin Ochratoxin A induces key parameters of chronic interstitial nephropathy in renal proximal tubular cells. Cell Physiol. Biochem. 2005, 15, 125–134. [Google Scholar] [CrossRef] [PubMed]
  199. Özcan, Z.; Gül, G.; Yaman, I. Ochratoxin A activates opposing c-MET/PI3K/Akt and MAPK/ERK 1–2 pathways in human proximal tubule HK-2 cells. Arch. Toxicol. 2015, 89, 1313–1327. [Google Scholar] [CrossRef] [PubMed]
  200. Liang, R.; Shen, X.L.; Zhang, B.; Li, Y.; Xu, W.; Zhao, C.; Luo, Y.; Huang, K. Apoptosis signal-regulating kinase 1 promotes Ochratoxin A-induced renal cytotoxicity. Sci. Rep. 2015, 5, 8078. [Google Scholar] [CrossRef] [PubMed]
  201. Atroshi, F.; Biese, I.; Saloniemi, H. Significance of apoptosis and its relationship to antioxidants after Ochratoxin A administration in mice. J. Pharm. Pharm. Sci. 2000, 3, 281–291. [Google Scholar] [PubMed]
  202. Petrik, J.; Zanic-Grubisic, T.; Barisic, K.; Pepeljnjak, S.; Radic, B.; Ferencic, Z.; Cepelak, I. Apoptosis and oxidative stress induced by Ochratoxin A in rat kidney. Arch. Toxicol. 2003, 77, 685–693. [Google Scholar] [CrossRef] [PubMed]
  203. Weidenbach, A.; Schuh, K.; Failing, K.; Petzinger, E. Ochratoxin A induced TNF-release from the isolated and blood-free perfused rat liver. Mycotoxin Res. 2000, 16, 189–193. [Google Scholar] [CrossRef] [PubMed]
  204. Al-Anati, L.; Katz, N.; Petzinger, E. Interference of arachidonic acid and its metabolites with TNFα release by Ochratoxin A from rat liver. Toxicology 2005, 208, 335–346. [Google Scholar] [CrossRef] [PubMed]
  205. Petzinger, E.; Weidenbach, A. Mycotoxins in the food chain: The role of ochratoxins. Livest. Prod. Sci. 2002, 76, 245–250. [Google Scholar] [CrossRef]
  206. Rached, E.; Pfeiffer, E.; Dekant, W.; Mally, A. Ochratoxin A: Apoptosis and aberrant exit from mitosis due to perturbation of microtubule dynamics? Toxicol. Sci. 2006, 92, 78–86. [Google Scholar] [CrossRef] [PubMed]
  207. Mally, A.; Dekant, W. Mycotoxins and the kidney: Modes of action for renal tumor formation by Ochratoxin A in rodents. Mol. Nutr. Food Res. 2009, 53, 467–478. [Google Scholar] [CrossRef] [PubMed]
  208. Adler, M.; Müller, K.; Rached, E.; Dekant, W.; Mally, A. Modulation of key regulators of mitosis linked to chromosomal instability is an early event in Ochratoxin A carcinogenicity. Carcinogenesis 2009, 30, 711–719. [Google Scholar] [CrossRef] [PubMed]
  209. Czakai, K.; Müller, K.; Mosesso, P.; Pepe, G.; Schulze, M.; Gohla, A.; Patnaik, D.; Dekant, W.; Higgins, J.M.G.; Mally, A. Perturbation of mitosis through inhibition of histone acetyltransferases: The key to Ochratoxin A toxicity and carcinogenicity? Toxicol. Sci. 2011, 122, 317–329. [Google Scholar] [CrossRef] [PubMed]
  210. O'Brien, E.; Heussner, A.H.; Dietrich, D.R. Species-, sex-, and cell type-specific effects of Ochratoxin A and B. Toxicol. Sci. 2001, 63, 256–264. [Google Scholar] [CrossRef] [PubMed]
  211. Palma, N.; Cinelli, S.; Sapora, O.; Wilson, S.H.; Dogliotti, E. Ochratoxin A-induced mutagenesis in mammalian cells is consistent with the production of oxidative stress. Chem. Res. Toxicol. 2007, 20, 1031–1037. [Google Scholar] [CrossRef] [PubMed]
  212. Wang, Y.; Liu, J.; Cui, J.; Xing, L.; Wang, J.; Yan, X.; Zhang, X. ERK and p38 MAPK signaling pathways are involved in Ochratoxin A-induced G2 phase arrest in human gastric epithelium cells. Toxicol. Lett. 2012, 209, 186–192. [Google Scholar] [CrossRef] [PubMed]
  213. Cui, J.; Liu, J.; Wu, S.; Wang, Y.; Shen, H.; Xing, L.; Wang, J.; Yan, X.; Zhang, X. Oxidative DNA damage is involved in Ochratoxin A-induced G2 arrest through ataxia telangiectasia-mutated (ATM) pathways in human gastric epithelium GES-1 cells in vitro. Arch. Toxicol. 2013, 87, 1829–1840. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, J.; Wang, Y.; Cui, J.; Xing, L.; Shen, H.; Wu, S.; Lian, H.; Wang, J.; Yan, X.; Zhang, X. Ochratoxin A induces oxidative DNA damage and G1 phase arrest in human peripheral blood mononuclear cells in vitro. Toxicol. Lett. 2012, 211, 164–171. [Google Scholar] [CrossRef] [PubMed]
  215. Yang, Q.; He, X.; Li, X.; Xu, W.; Luo, Y.; Yang, X.; Wang, Y.; Li, Y.; Huang, K. DNA damage and S phase arrest induced by Ochratoxin A in human embryonic kidney cells (HEK 293). Mutat. Res. 2014, 765, 22–31. [Google Scholar] [CrossRef] [PubMed]
  216. Yang, Q.; Shi, L.; Huang, K.; Xu, W. Protective effect of N-acetylcysteine against DNA damage and S-phase arrest induced by Ochratoxin A in human embryonic kidney cells (HEK-293). Food Chem Toxicol. 2014, 70, 40–47. [Google Scholar] [CrossRef] [PubMed]
  217. Khan, S.; Martin, M.; Bartsch, H.; Rahimtula, A.D. Perturbation of liver microsomal calcium homeostasis by Ochratoxin A. Biochem. Pharmacol. 1989, 38, 67–72. [Google Scholar] [CrossRef]
  218. Dopp, E.; Müller, J.; Hahnel, C.; Schiffmann, D. Induction of genotoxic effects and modulation of the intracellular calcium level in Syrian Hamster Embryo (SHE) fibroblasts caused by Ochratoxin A. Food Chem. Toxicol. 1999, 37, 713–721. [Google Scholar] [CrossRef]
  219. Hoehler, D.; Marquardt, R.R.; McIntosh, A.R.; Xiao, H. Free radical generation as induced by Ochratoxin A and its analogs in bacteria (Bacillus brevis). J. Biol. Chem. 1996, 271, 27388–27394. [Google Scholar] [CrossRef] [PubMed]
  220. Hoehler, D.; Marquardt, R.R.; McIntosh, A.R.; Hatch, G.M. Induction of free radicals in hepatocytes, mitochondria and microsomes of rats by Ochratoxin A and its analogs. Biochim. Biophys. Acta 1997, 1357, 225–233. [Google Scholar] [CrossRef]
  221. Benesic, A.; Mildenberger, S.; Gekle, M. Nephritogenic Ochratoxin A interferes with hormonal signalling in immortalized human kidney epithelial cells. Pflügers Arch. 2000, 439, 278–287. [Google Scholar] [CrossRef] [PubMed]
  222. García, A.R.; Avila, E.; Rosiles, R.; Petrone, V.M. Evaluation of two mycotoxin binders to reduce toxicity of broiler diets containing Ochratoxin A and T-2 toxin contaminated grain. Avian Dis. 2003, 47, 691–699. [Google Scholar] [CrossRef] [PubMed]
  223. Trailović, J.N.; Stefanović, S.; Trailović, S.M. In vitro and in vivo protective effects of three mycotoxin adsorbents against Ochratoxin A in broiler chickens. Br. Poult. Sci. 2013, 54, 515–523. [Google Scholar] [CrossRef] [PubMed]
  224. Madhyastha, M.S.; Frohlich, A.A.; Marquardt, R.R. Effect of dietary cholestyramine on the elimination pattern of Ochratoxin A in rats. Food Chem. Toxicol. 1992, 30, 709–714. [Google Scholar] [CrossRef]
  225. Kerkadi, A.; Barriault, C.; Marquardt, R.R.; Frohlich, A.A.; Yousef, I.M.; Zhu, X.X.; Tuchweber, B. Cholestyramine protection against Ochratoxin A toxicity: Role of Ochratoxin A sorption by the resin and bile acid enterohepatic circulation. J. Food Prot. 1999, 62, 1461–1465. [Google Scholar] [PubMed]
  226. Yong, S.; Albassam, M.; Prior, M. Protective effects of sodium bicarbonate on murine ochratoxicosis. J. Environ. Sci. Health B 1987, 22, 455–470. [Google Scholar] [CrossRef] [PubMed]
  227. Baudrimont, I.; Murn, M.; Betbeder, A.M.; Guilcher, J.; Creppy, E.E. Effect of piroxicam on the nephrotoxicity induced by Ochratoxin A in rats. Toxicology 1995, 95, 147–154. [Google Scholar] [CrossRef]
  228. Moroi, K.; Suzuki, S.; Kuga, T.; Yamazaki, M.; Kanisawa, M. Reduction of Ochratoxin A toxicity in mice treated with phenylalanine and phenobarbital. Toxicol. Lett. 1985, 25, 1–5. [Google Scholar] [PubMed]
  229. Chakor, K.; Creppy, E.E.; Dirheimer, G. In vivo Studies on the relationship between hepatic metabolism and toxicity of Ochratoxin A. Arch. Toxicol. 1988, 12, 201–204. [Google Scholar]
  230. Suzuki, S.; Moroi, K.; Kanisawa, M.; Satoh, T. Effect of drug metabolizing enzyme inducers on carcinogenesis and toxicity of Ochratoxin A in mice. Toxicol. Lett. 1986, 31, 206. [Google Scholar]
  231. Creppy, E.E.; Schlegel, M.; Röschenthaler, R.; Dirheimer, G. Phenylalanine prevents acute poisoning by ochratoxina in mice. Toxicol. Lett. 1980, 6, 77–80. [Google Scholar] [CrossRef]
  232. Baudrimont, I.; Betbeder, A.M.; Creppy, E.E. Reduction of the Ochratoxin A-induced cytotoxicity in Vero cells by aspartame. Arch. Toxicol. 1997, 71, 290–298. [Google Scholar] [CrossRef] [PubMed]
  233. Baudrimont, I.; Sostaric, B.; Yenot, C.; Betbeder, A.M.; Dano-Djedje, S.; Sanni, A.; Steyn, P.S.; Creppy, E.E. Aspartame prevents the karyomegaly induced by Ochratoxin A in rat kidney. Arch. Toxicol. 2001, 75, 176–183. [Google Scholar] [PubMed]
  234. Stoev, S.D. Studies on carcinogenic and toxic effects of Ochratoxin A in chicks. Toxins 2010, 2, 649–664. [Google Scholar] [CrossRef] [PubMed]
  235. Creppy, E.E.; Baudrimont, I.; Belmadani, A.; Betbeder, A.M. Aspartame as a preventive agent of chronic toxic effects of Ochratoxin A in experimental animals. Toxin Rev. 1996, 15, 207–221. [Google Scholar] [CrossRef]
  236. Pfohl-Leszkowicz, A.; Bartsch, H.; Azémar, B.; Mohr, U.; Estève, J.; Castegnaro, M. MESNA protects rats against nephrotoxicity but not carcinogenicity induced by Ochratoxin A, implicating two separate pathways. Facta Univ. Ser. Med. Biol. 2002, 9, 57–63. [Google Scholar]
  237. Kumari, D.; Sinha, S.P. Effect of retinol on ochratoxin-produced genotoxicity in mice. Food Chem. Toxicol. 1994, 32, 471–475. [Google Scholar] [CrossRef]
  238. Hoehler, D.; Marquardt, R.R. Influence of vitamins E and C on the toxic effects of Ochratoxin A and T-2 toxin in chicks. Poult. Sci. 1996, 75, 1508–1515. [Google Scholar] [CrossRef] [PubMed]
  239. Grosse, Y.; Chekir-Ghedira, L.; Huc, A.; Obrecht-Pflumio, S.; Dirheimer, G.; Bacha, H.; Pfohl-Leszkowicz, A. Retinol, ascorbic acid and alpha-tocopherol prevent DNA adduct formation in mice treated with the mycotoxins Ochratoxin A and zearalenone. Cancer Lett. 1997, 114, 225–229. [Google Scholar] [CrossRef]
  240. Baldi, A.; Losio, M.N.; Cheli, F.; Rebucci, R.; Sangalli, L.; Fusi, E.; Bertasi, B.; Pavoni, E.; Carli, S.; Politis, I. Evaluation of the protective effects of alpha-tocopherol and retinol against Ochratoxin A cytotoxicity. Br. J. Nutr. 2004, 91, 507–512. [Google Scholar] [CrossRef] [PubMed]
  241. Costa, S.; Utan, A.; Cervellati, R.; Speroni, E.; Guerra, M.C. Catechins: Natural free-radical scavengers against Ochratoxin A-induced cell damage in a pig kidney cell line (LLC-PK1). Food Chem. Toxicol. 2007, 45, 1910–1917. [Google Scholar] [CrossRef] [PubMed]
  242. Renzulli, C.; Galvano, F.; Pierdomenico, L.; Speroni, E.; Guerra, M.C. Effects of rosmarinic acid against aflatoxin B1 and Ochratoxin-A-induced cell damage in a human hepatoma cell line (Hep G2). J. Appl. Toxicol. 2004, 24, 289–296. [Google Scholar] [CrossRef] [PubMed]
  243. Russo, A.; la Fauci, L.; Acquaviva, R.; Campisi, A.; Raciti, G.; Scifo, C.; Renis, M.; Galvano, G.; Vanella, A.; Galvano, F. Ochratoxin A-induced DNA damage in human fibroblast: Protective effect of cyanidin 3-O-beta-d-glucoside. J. Nutr. Biochem. 2005, 16, 31–37. [Google Scholar] [CrossRef] [PubMed]
  244. Guerra, M.C.; Galvano, F.; Bonsi, L.; Speroni, E.; Costa, S.; Renzulli, C.; Cervellati, R. Cyanidin-3-O-beta-glucopyranoside, a natural free-radical scavenger against aflatoxin B1- and Ochratoxin A-induced cell damage in a human hepatoma cell line (Hep G2) and a human colonic adenocarcinoma cell line (CaCo-2). Br. J. Nutr. 2005, 94, 211–220. [Google Scholar] [CrossRef] [PubMed]
  245. Sorrenti, V.; di Giacomo, C.; Acquaviva, R.; Bognanno, M.; Grilli, E.; D'Orazio, N.; Galvano, F. Dimethylarginine dimethylaminohydrolase/nitric oxide synthase pathway in liver and kidney: Protective effect of cyanidin 3-O-β-d-glucoside on Ochratoxin-A toxicity. Toxins 2012, 4, 353–363. [Google Scholar] [CrossRef] [PubMed]
  246. Essid, E.; Dernawi, Y.; Petzinger, E. Apoptosis induction by OTA and TNF-α in cultured primary rat hepatocytes and prevention by silibinin. Toxins 2012, 4, 1139–1156. [Google Scholar] [CrossRef] [PubMed]
  247. Cariddi, L.N.; Sabini, M.C.; Escobar, F.M.; Montironi, I.; Manas, F.; Iglesias, D.; Comini, L.R.; Sabini, L.I.; Dalcero, A.M. Polyphenols as possible bioprotectors against cytotoxicity and DNA damage induced by Ochratoxin A. Environ. Toxicol. Pharmacol. 2015, 39, 1008–1018. [Google Scholar] [CrossRef] [PubMed]
  248. Ramyaa, P.; Padma, V.V. Ochratoxin-induced toxicity, oxidative stress and apoptosis ameliorated by quercetin—Modulation by Nrf2. Food Chem. Toxicol. 2013, 62, 205–216. [Google Scholar] [CrossRef] [PubMed]
  249. Ramyaa, P.; Krishnaswamy, R.; Padma, V.V. Quercetin modulates OTA-induced oxidative stress and redox signalling in HepG2 cells—up regulation of Nrf2 expression and down regulation of NF-κB and COX-2. Biochim. Biophys. Acta 2014, 1840, 681–692. [Google Scholar] [CrossRef] [PubMed]
  250. Aydin, S.; Palabiyik, S.S.; Erkekoglu, P.; Sahin, G.; Başaran, N.; Giray, B.K. The carotenoid lycopene protects rats against DNA damage induced by Ochratoxin A. Toxicon 2013, 73, 96–103. [Google Scholar] [CrossRef] [PubMed]
  251. Palabiyik, S.S.; Erkekoglu, P.; Zeybek, N.D.; Kizilgun, M.; Baydar, D.E.; Sahin, G.; Giray, B.K. Protective effect of lycopene against Ochratoxin A induced renal oxidative stress and apoptosis in rats. Exp. Toxicol. Pathol. 2013, 65, 853–861. [Google Scholar] [CrossRef] [PubMed]
  252. Abdel-Wahhab, M.A.; Abdel-Azim, S.H.; El-Nekeety, A.A. Inula crithmoides extract protects against Ochratoxin A-induced oxidative stress, clastogenic and mutagenic alterations in male rats. Toxicon 2008, 52, 566–573. [Google Scholar] [CrossRef] [PubMed]
  253. Chakraborty, D.; Verma, R. Ameliorative effect of Emblica officinalis aqueous extract on ochratoxin-induced lipid peroxidation in the kidney and liver of mice. Int. J. Occup. Med. Environ. Health 2010, 23, 63–73. [Google Scholar] [CrossRef] [PubMed]
  254. Gan, F.; Xue, H.; Huang, Y.; Pan, C.; Huang, K. Selenium alleviates porcine nephrotoxicity of Ochratoxin A by improving selenoenzyme expression in vitro. PLoS ONE 2015, 10, e119808. [Google Scholar]
  255. Mantle, P.; Kilic, M.A.; Mor, F.; Ozmen, O. Contribution of organ vasculature in rat renal analysis for Ochratoxin A: Relevance to toxicology of nephrotoxins. Toxins 2015, 7, 1005–1017. [Google Scholar] [CrossRef] [PubMed]
  256. Obrecht-Pflumio, S.; Grosse, Y.; Pfohl-Leszkowicz, A.; Dirheimer, G. Protection by indomethacin and aspirin against genotoxicity of Ochratoxin A, particularly in the urinary bladder and kidney. Arch. Toxicol. 1996, 70, 244–248. [Google Scholar] [CrossRef] [PubMed]
  257. Baudrimont, I.; Ahouandjivo, R.; Creppy, E.E. Prevention of lipid peroxidation induced by Ochratoxin A in Vero cells in culture by several agents. Chem. Biol. Interact. 1997, 104, 29–40. [Google Scholar] [CrossRef]
  258. Galtier, P.; Camguilhem, R.; Bodin, G. Evidence for in vitro and in vivo interaction between Ochratoxin A and three acidic drugs. Food Cosmet. Toxicol. 1980, 18, 493–496. [Google Scholar] [CrossRef]
  259. Creppy, E.E.; Baudrimont, I.; Betbeder, A.M. Prevention of nephrotoxicity of Ochratoxin A, a food contaminant. Toxicol. Lett. 1995, 82–83, 869–877. [Google Scholar] [CrossRef]
  260. Hong, S.S.; Seo, K.; Lim, S.C.; Han, H.K. Interaction characteristics of flavonoids with human organic anion transporter 1 (hOAT1) and 3 (hOAT3). Pharmacol. Res. 2007, 56, 468–473. [Google Scholar] [CrossRef] [PubMed]
  261. Wong, C.C.; Botting, N.P.; Orfila, C.; Al-Maharik, N.; Williamson, G. Flavonoid conjugates interact with organic anion transporters (OATs) and attenuate cytotoxicity of adefovir mediated by organic anion transporter 1 (OAT1/SLC22A6). Biochem. Pharmacol. 2011, 81, 942–949. [Google Scholar] [CrossRef] [PubMed]
  262. Wang, X.; Wolkoff, A.W.; Morris, M.E. Flavonoids as a novel class of human organic anion-transporting polypeptide OATP1B1 (OATP-C) modulators. Drug Metab. Dispos. 2005, 33, 1666–1672. [Google Scholar] [CrossRef] [PubMed]
  263. Sergent, T.; Garsou, S.; Schaut, A.; Saeger, S.D.; Pussemier, L.; Peteghem, C.V.; Larondelle, Y.; Schneider, Y.J. Differential modulation of Ochratoxin A absorption across Caco-2 cells by dietary polyphenols, used at realistic intestinal concentrations. Toxicol. Lett. 2005, 159, 60–70. [Google Scholar] [CrossRef] [PubMed]
  264. Brand, W.; Schutte, M.E.; Williamson, G.; van Zanden, J.J.; Cnubben, N.H.P.; Groten, J.P.; van Bladeren, P.J.; Rietjens, I.M.C.M. Flavonoid-mediated inhibition of intestinal ABC transporters may affect the oral bioavailability of drugs, food-borne toxic compounds and bioactive ingredients. Biomed. Pharmacother. 2006, 60, 508–519. [Google Scholar] [CrossRef] [PubMed]
  265. Havsteen, B.H. The biochemistry and medical significance of the flavonoids. Pharmacol. Ther. 2002, 96, 67–202. [Google Scholar] [CrossRef]
  266. Dangles, O.; Dufour, C.; Manach, C.; Morand, C.; Remesy, C. Binding of flavonoids to plasma proteins. Methods Enzymol. 2001, 335, 319–333. [Google Scholar] [PubMed]
Figure 1. Chemical structures of Ochratoxin A (dark blue: phenylalanine part, red: dihydro-isocoumarin ring, green: acidic hydrogens), B, and C. The highlighted structures are characteristic to the three different ochratoxin molecules (light blue).
Figure 1. Chemical structures of Ochratoxin A (dark blue: phenylalanine part, red: dihydro-isocoumarin ring, green: acidic hydrogens), B, and C. The highlighted structures are characteristic to the three different ochratoxin molecules (light blue).
Toxins 08 00111 g001
Figure 2. Biotransformation of Ochratoxin A.
Figure 2. Biotransformation of Ochratoxin A.
Toxins 08 00111 g002
Figure 3. Bioactivation of Ochratoxin A producing genotoxic metabolites based on Pfohl-Leszkowicz and Manderville [38] (OTA: Ochratoxin A, OT-GSH: OTA-glutathione conjugate, OTQ: OTA-quinone, OTHQ: OTA-hydroquinone, and OTB: Ochratoxin B).
Figure 3. Bioactivation of Ochratoxin A producing genotoxic metabolites based on Pfohl-Leszkowicz and Manderville [38] (OTA: Ochratoxin A, OT-GSH: OTA-glutathione conjugate, OTQ: OTA-quinone, OTHQ: OTA-hydroquinone, and OTB: Ochratoxin B).
Toxins 08 00111 g003
Figure 4. Chemical structures of OTA nucleoside adducts based on Pfohl-Leszkowicz and Manderville [38] (C-OTB-dG: C-coupled C8-deoxyguanosine-adduct, O-OTA-dG: O-coupled C8-deoxyguanosine-adduct, and OTHQ-dG: OTA-hydroquinone C8-deoxyguanosine-adduct).
Figure 4. Chemical structures of OTA nucleoside adducts based on Pfohl-Leszkowicz and Manderville [38] (C-OTB-dG: C-coupled C8-deoxyguanosine-adduct, O-OTA-dG: O-coupled C8-deoxyguanosine-adduct, and OTHQ-dG: OTA-hydroquinone C8-deoxyguanosine-adduct).
Toxins 08 00111 g004

Share and Cite

MDPI and ACS Style

Kőszegi, T.; Poór, M. Ochratoxin A: Molecular Interactions, Mechanisms of Toxicity and Prevention at the Molecular Level. Toxins 2016, 8, 111. https://doi.org/10.3390/toxins8040111

AMA Style

Kőszegi T, Poór M. Ochratoxin A: Molecular Interactions, Mechanisms of Toxicity and Prevention at the Molecular Level. Toxins. 2016; 8(4):111. https://doi.org/10.3390/toxins8040111

Chicago/Turabian Style

Kőszegi, Tamás, and Miklós Poór. 2016. "Ochratoxin A: Molecular Interactions, Mechanisms of Toxicity and Prevention at the Molecular Level" Toxins 8, no. 4: 111. https://doi.org/10.3390/toxins8040111

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

Article Metrics

Back to TopTop