Next Article in Journal
Long-Term Efficacy and Safety of Repeated Intravescial OnabotulinumtoxinA Injections Plus Hydrodistention in the Treatment of Interstitial Cystitis/Bladder Pain Syndrome
Next Article in Special Issue
Teratogenicity of Ochratoxin A and the Degradation Product, Ochratoxin α, in the Zebrafish (Danio rerio) Embryo Model of Vertebrate Development
Previous Article in Journal
UV-B Exposure Affects the Biosynthesis of Microcystin in Toxic Microcystis aeruginosa Cells and Its Degradation in the Extracellular Space
Previous Article in Special Issue
Man-Made Synthetic Receptors for Capture and Analysis of Ochratoxin A
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comparative Ochratoxin Toxicity: A Review of the Available Data

by
Alexandra H. Heussner
1,2,* and
Lewis E. H. Bingle
2
1
Human and Environmental Toxicology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
2
Department of Pharmacy, Health and Well-Being, University of Sunderland, City Campus, Sunderland SR1 3SD, UK
*
Author to whom correspondence should be addressed.
Submission received: 19 August 2015 / Revised: 27 September 2015 / Accepted: 15 October 2015 / Published: 22 October 2015
(This article belongs to the Collection Ochratoxins-Collection)

Abstract

:
Ochratoxins are a group of mycotoxins produced by a variety of moulds. Ochratoxin A (OTA), the most prominent member of this toxin family, was first described by van der Merwe et al. in Nature in 1965. Dietary exposure to OTA represents a serious health issue and has been associated with several human and animal diseases including poultry ochratoxicosis, porcine nephropathy, human endemic nephropathies and urinary tract tumours in humans. More than 30 years ago, OTA was shown to be carcinogenic in rodents and since then extensive research has been performed in order to investigate its mode of action, however, this is still under debate. OTA is regarded as the most toxic family member, however, other ochratoxins or their metabolites and, in particular, ochratoxin mixtures or combinations with other mycotoxins may represent serious threats to human and animal health. This review summarises and evaluates current knowledge about the differential and comparative toxicity of the ochratoxin group.

1. Ochratoxin A—An Important Mycotoxin

Ochratoxin A was first described in a paper published in Nature by van der Merwe and co-workers (1965) after they isolated a new toxic metabolite from Aspergillus ochraceus [1]. Ochratoxins are produced by certain Aspergillus species such as A. ochraceus or A. niger and some Penicillium species, especially P. verrucosum. The main forms are ochratoxin A, B, and C, which differ in that ochratoxin B (OTB) is a non-chlorinated form of ochratoxin A (OTA) and ochratoxin C (OTC) is an ethyl ester of OTA (Table 1). OTA ((R)-N-((5-Chlor-3,4-dihydro-8-hydroxy-3-methyl-1-oxo-1H-benzo[c]-pyran-7-yl)-carbonyl)-3-phenylalanin) is the most prevalent and relevant form of this group, while OTB and OTC are generally assumed to be of lesser importance [2].
Table 1. Overview of main ochratoxin forms.
Table 1. Overview of main ochratoxin forms.
NameOTAOTBOTC
CAS number303-47-94825-86-94865-85-4
Molecular formulaC20H18ClNO6C20H19NO6C22H22ClNO6
Molar mass (g·mol−1)403.8369.4431.9
CAS, Chemical Abstracts Service.
OTA is a highly abundant food and animal feed contaminant and is frequently detected in all types of cereals and cereal products, but also coffee, cacao, grapes, raisins, wine, soy, spices, nuts, pulses, liquorice and beer [3,4,5,6]. A European report estimated the contributions to the total human adult OTA exposure as being 44% cereals, 15% others, 10% wine, 9% coffee, 7% beer, 5% cacao, 4% dried fruits, 3% meat and 3% spices [5]. In general, the mean OTA contamination levels in European food commodities are relatively low (ng kg−1 to µg kg−1), however, elevated concentrations can occur in individual batches. Even though preventive measures are taken to keep the levels of OTA in food low, a certain degree of contamination seems unavoidable [7,8]. Furthermore, in other countries where screening of food is rare and outdated storage and transport conditions are still in use, much higher contamination levels may occur. Many farm animals are fed with cereal-based diets exposing them to ochratoxins (usually in the low to high µg kg−1 range) [9], but again elevated contamination levels may occur depending on storage and feeding practice [10]. The highest contamination level reported was 80 mg OTA kg−1 in mouldy bread intended for animal feeding [11]. OTA has a high affinity for proteins, particularly serum albumin, which promotes bio-accumulation in the organs of animals, leading to carry-over of the contamination [9]. Therefore, muscle and offal products, as well as milk and sometimes eggs, may be contaminated, and the highest level of OTA contamination can be found in porcine blood-based sausages, “black pudding”, and liver products (e.g., pâtés, sausages) [9].
OTA-related diseases of economic importance occur in poultry (poultry ochratoxicosis) [12,13,14,15,16,17] and pigs (mycotoxic porcine nephropathy, MPN) [18,19,20,21]. These diseases are characterised by severe kidney damage, which could be clearly associated with the exposure to ochratoxins, sometimes in combination with other mycotoxins, as shown in several epizootiological and feeding studies [20,21,22,23,24,25]. Similarly, a slow, progressive renal disease (Endemic Nephropathy, EN) occurs in humans which is characterised by cellular interstitial fibrosis, tubular atrophy, and karyomegaly predominately in proximal convoluted tubules [26,27,28,29,30]. The aetiology of EN is still unknown, but based on epidemiological features, researchers agree that the causative agent is of natural origin [26,31]. There are several hypotheses as to the causes of EN that are currently in circulation, the two most important focus on the potential roles of aristolochic acid from the plant birthwort (Aristolochia clematitis) and mycotoxins (OTA and citrinin).
The toxicity of OTA has been investigated intensively in recent decades and numerous comprehensive reviews have been published with respect to all types of toxic effects. The scope of this review is to summarise and evaluate the existing data on the less well-studied members of the ochratoxin family, including the known metabolites, in comparison to OTA.

2. Occurrence of Ochratoxins

2.1. Biosynthesis

The biosynthetic pathway of OTA (an amide formed between phenylalanine and a chlorinated dihydroisocoumarin) is not completely established. However, it is known that the phenylalanine moiety originates from the shikimate pathway and the dihydroisocoumarin moiety from a polyketide (pentaketide) pathway, with the latter differing between Penicillium and Aspergillus species [32,33,34,35,36,37,38,39]. Also, the OTA polyketide synthase genes have been characterised from several fungal species [32,40,41,42,43].
Both ochratoxin production and the primary producing organisms depend on factors that affect fungal physiology such as temperature, moisture and the available substrates [2,20,33,39,44]. Aspergillus species can produce OTA and OTB in parallel [44,45], and experiments with A. ochraceus have revealed growth-associated production of OTA and OTB, in which the yield and the ratio were dependent on the prevailing culture conditions [44,46,47,48]. Often, the amount of OTB produced was considerably lower than that of OTA, but under certain conditions the level of OTB production was comparable to that of OTA [46,47]. The reported production ratios (OTA:OTB) ranged from 2:1 to 34:1 [46,49,50]. A complex interaction of different carbon sources, basal media and nitrogen sources seems to be crucial [44]. An increased OTA production was correlated to an induction of OTA polyketide synthase expression, whereas OTB production does not correlate with transcription of the polyketide synthase gene [44]. Laboratory fermentation experiments with A. ochraceus resulted in very high yields (up to 10 mg/g) of OTA, OTB and transitorily also ochracin (mellein, see Table 2) [46]. The intermediate metabolite OTβ was found to be biotransformed very efficiently into both OTA and OTB (14% and 19%, respectively), whereas OTα was biotransformed only into OTA (4.9%). Also, OTB is poorly converted (1.5%) into OTA, whereas some OTB may be produced by dechlorination of OTA [46].
Table 2. Natural and synthetic forms of ochratoxins. Toxins 07 04253 i001
Table 2. Natural and synthetic forms of ochratoxins. Toxins 07 04253 i001
NameAbbrev.R1 and OthersR2R3R4R5
Naturally occurring ochratoxins, metabolites and conjugates
Ochratoxin AOTAPhenylalanylClHHH
Ochratoxin BOTBPhenylalanylHHHH
Ochratoxin COTCPhenylalanyl, ethyl esterClHHH
Ochratoxin A methyl ester--Phenylalanyl, methyl esterClHHH
Ochratoxin B methyl ester--Phenylalanyl, methyl esterHHHH
Ochratoxin B ethyl ester--Phenylalanyl, ethyl esterHHHH
Ochratoxin αOTαOHClHHH
Ochratoxin βOTβOHHHHH
5′-Hydroxyochratoxin A5′-OH-OTAPhenylalanyl, OH at C-5′ClHHH
7′-Hydroxyochratoxin A7′-OH-OTAPhenylalanyl, OH at C-7′ClHHH
9′-Hydroxyochratoxin A9′-OH-OTAPhenylalanyl, OH at C-9′ClHHH
4R-Hydroxyochratoxin A4R-OH-OTAPhenylalanylClHOHH
4S-Hydroxyochratoxin A4S-OH-OTAPhenylalanylClOHHH
4R-Hydroxyochratoxin B4R-OH-OTBPhenylalanylHHOHH
4S-Hydroxyochratoxin B4S-OH-OTBPhenylalanylHOHHH
10-Hydroxyochratoxin A10-OH-OTAPhenylalanylClHHOH
Open lactone of ochratoxin AOP-OTAPhenylalanylClHHH
Ochratoxin hydroquinoneOTHQPhenylalanylOHHHH
Ochratoxin quinoneOTQPhenylalanylOHHH
Ochratoxin α glucuronide--GlucuronideClHHH
--OH; glucuronide at C-8ClHHH
Ochratoxin A phenol-glucuronide--Phenylalanyl; glucuronide at C-8ClHHH
Ochratoxin A amino-glucuronide--Phenylalanyl; glucuronide at NClHHH
Ochratoxin A acyl-glucuronide--Phenylalanyl; acyl-glucuronide instead of carboxylClHHH
Ochratoxin A acyl-hexose--Phenylalanyl; acyl-hexose instead of carboxylClHHH
Ochratoxin A acyl-pentose--Phenylalanyl; acyl-pentose instead of carboxylClHHH
Synthetic ochratoxins
Ochratoxin A, tyrosine analogue--TyrosineClHHH
Ochratoxin A, serine analogue--SerineClHHH
Ochratoxin A, hydroxyproline analogue--HydroxyprolineClHHH
Ochratoxin A, lysine analogue--LysineClHHH
Ochratoxin A, alanine analogue--AlanineClHHH
Ochratoxin A, leucine analogue--LeucineClHHH
d-Ochratoxin Ad-OTAd-PhenylalanylClHHH
Ochratoxin A, ethylamideOE-OTAPhenylalanyl, ethylamideClHHH
O-methylated ochratoxin AOM-OTAPhenylalanyl, OCH3 at C-8ClHHH
Methyl ester of ochratoxin αM-OTαMethoxyClHHH
Ochratoxin A, decarboxylatedDC-OTAPhenylethylamineClHHH
Other related compounds
Ochracin (=mellein)--H at C-7HHHH
The table contains only those forms that have been detected or synthesised and described until present. Summarised from [39,51,52,53,54,55,56,57].

2.2. Contamination Levels and Human Exposure

Ochratoxins occur ubiquitously and, at least for OTA, contamination levels in food and feed are monitored in many countries. OTB contamination levels of food and feed were assumed to be generally low [58], but based on the data on production described above, it can also be assumed that the contamination levels may sometimes be almost as high as those of OTA. Unfortunately, there is insufficient data on OTB contamination of food and feed, although when tested, OTB is often found together with OTA in both [59]. For example, a survey of Japanese commercial foods containing cereal, fruit, coffee or cacao showed a contamination with OTA at the low µg kg−1 level in more than one third of the samples [60]. Those with high OTA levels also contained OTB (up to 30% of the OTA concentration) [60]. Another survey of 681 samples from more than 50 different spice commodities resulted in positive detection of OTA and OTB in 21% and 10% of samples, respectively [61]. More recently, 57 samples of Sicilian red wines were found to be mostly weakly contaminated (<EU legal limit), ranging 0.02–0.73 μg L−1 and 0.04–0.66 μg L−1 for OTA and OTB, respectively [62]. OTA was found in 71.9% and OTB in 64.9% of samples tested [55]. Similar observations have been published for animal feed. For example, Hamilton and co-workers found a contamination ratio of about 90:8:2 for OTA, OTB and OTC, respectively, in poultry feed [13].
Humans are mainly exposed via food and beverages and the human dietary exposure has been estimated to be 15–60 ng OTA kg−1 b.w. per week for adult consumers in the EU [8,63]. Furthermore, many farm animals are fed with cereal-based diets exposing them to ochratoxins. Based on the available data on ochratoxin occurrence, it can be assumed that animals and humans are mainly exposed to OTA and to a much lesser extent to OTB and OTC. However, varying maximum exposure levels of all three forms as well as mixtures of different ratios may occur.
A recent human biomonitoring study of 33 mycotoxins in the Belgian population revealed that nine of these were detected via LC-MS/MS whereby deoxynivalenol, OTA, citrinin and their metabolites were the most frequently detected [64]. Citrinin and OTA were present in very low concentrations (pg mL−1 urine) and contaminated 59% and 35% of the samples from adults, respectively, with the rates in children being even higher [64]. The estimated daily intake of OTA showed that about 1% of the investigated population possibly exceeded the tolerable daily intake level [64].

2.3. Toxicokinetics of Ochratoxins

For the interpretation of in vivo data from toxicological reports an understanding of the kinetics of ochratoxins is crucial, including the absorption, distribution, metabolism, and excretion of the toxins [65]. The toxicokinetics of OTA and their toxicological implications for animals and humans have been extensively reviewed by various authors [7,39,53,66,67,68,69,70,71,72,73]. For example, the available kinetic data for rats indicate that strain, sex and age differences in OTA kinetics may account for the different sex and species sensitivities towards OTA [69]. Human data is rather limited, however, gender, season and geographic location seem to be most important factors [68].
Toxicokinetic studies employing ochratoxins other than OTA are rare [58,74,75,76]. For example, Mally and co-workers reported that OTB is more extensively metabolised and more rapidly eliminated than OTA in F344 rats due to the lack of specific retention of OTB in the kidneys, which may explain the different toxicological outcomes in animals compared to in vitro approaches [58].
Based on the metabolism of animals and humans which includes hydrolysis, hydroxylation, lactone opening and conjugation [53], various ochratoxin metabolites can occur (Table 2, Figure 1). OTA can be enzymatically hydrolysed (e.g., by carboxypeptidase A, chymotrypsin) to the less toxic OTα by the bacterial microflora in the intestine as shown in ruminants and rodents [53,77,78]. Approximately 25% of ingested OTA is excreted as OTα in the urine of rats after reabsorption from the intestine [79,80]. OTα was also formed in OTA-exposed human bronchial epithelial cells in vitro [81].
Hydroxylated urinary metabolites of OTA include 4-hydroxy (4R- and 4S-) epimers, which are produced in liver [52,80,82] and kidney [80,83] by the action of various cytochromes P450 [80,82,84]. Exposed rats (i.p. or orally) excrete 4R-OH-OTA in their urine, together with the parent compound and OTα [85]. These 4-hydroxy metabolites are also produced by human, pig, goat, chicken and rat liver microsomes or human bronchial epithelial cells in vitro [52,80,81,82]. Furthermore, the 10-hydroxy metabolite is produced by rabbit liver and kidney microsomes and in human bronchial epithelial cells in culture, however, it has not yet been detected in vivo [80,81,84,86,87,88]. A recent study compared the in vitro metabolic profile of OTA in rat, chicken, pig, goat, cow and human liver microsomes [52]. This study resulted in the same six metabolites in all genera, i.e., 4S-OH-OTA, 4R-OH-OTA, 7′-OH-OTA, 9′-OH-OTA, 5′-OH-OTA and OTB [52]. A semi-quantitative analysis showed that most of the OTA remained unchanged, confirming earlier reports that OTA is not primarily metabolised in the liver [52,53,79]. Furthermore, the metabolic capacity of human liver microsomes was higher compared to the other animals tested resulting in larger amounts of the metabolites [52]. Additional in vivo experiments in rats and chicken showed that OTA undergoes extensive metabolism after oral administration resulting in eight and six metabolites in rats and chicken, respectively [52]. These metabolites included 4S-OH-OTB and 4R-OH-OTB (in rats only) in addition to those found in vitro [52]. Again, parent OTA was the major compound in rat urine (64.0% ± 4.0%) and chicken excreta (56.0% ± 2.5%) [77]. The major metabolite was 4R-OH-OTA in rats (>20% vs. <10% in chickens) and 7′-OH-OTA in chickens (20% vs. 0.2% in rats) [52].
Glucuronidation of ochratoxins has been shown in rat liver microsomes [57] and in vivo in [3H]-OTA-exposed mice [89], where a large proportion of the biliary metabolites were ochratoxin glucuronides and sulphate conjugates [89]. Such conjugates were also detected in liver and intestinal tissue, although to a smaller percentage [89]. Also in pigs, glucuronide conjugates were detected in the bile [90]. In contrast, in in vitro experiments using primary rat and human hepatocytes, no ochratoxin glucuronide and sulphate conjugates were observed [91]. Pentose and hexose conjugates of OTA were detected in primary rat and human hepatocytes [91] and in the urine of rats [92]. Glutathione (GSH) conjugation is also an important mechanism in xenobiotic biotransformation and is generally a detoxification step, although it can result in bioactivation, in particular by formation of a thiyl radical, which favours the formation of peroxyl radicals. For example in E. coli, a cysteinyl-OTA conjugate has been detected [93]. Stable GSH-conjugates such as quinone-derived GSH-conjugates can be formed in the liver and can be biotransformed into cysteinyl conjugates in the kidney, which are then N-acetylated and excreted [94]. Most likely, GSH-conjugates are produced to a very low extent (1%) in vivo and, therefore, their detection is difficult and rarely reported [94,95].
Figure 1. Overview of ochratoxin A (OTA) metabolites in animals and humans. Ochratoxins are differentially metabolised in various animals and humans depending on the metabolising enzymes present in liver, kidneys and/or gut. OTA, ochratoxin A; OTB, ochratoxin B; OTC, ochratoxin C; OTα, ochratoxin α; OTβ, ochratoxin β, OTHQ, OT hydroquinone; 4R-OH-OTB, 4R-hydroxyochratoxin B; OP-OTA, lactone-opened OTA; 10-OH-OTA, 10-hydroxyochratoxin A; 4R-OH-OTA, 4R-hydroxyochratoxin A; 4S-OH-OTA, 4S-hydroxyochratoxin A; 4S-OH-OTB, 4S-hydroxyochratoxin B. The respective metabolite is the main metabolite in the underlined taxa (for rodents, data of different authors is contradictory). Genera in brackets represent those where the metabolite has been detected, although it may be present in other animals or humans as well. Summarised from [52,53,92].
Figure 1. Overview of ochratoxin A (OTA) metabolites in animals and humans. Ochratoxins are differentially metabolised in various animals and humans depending on the metabolising enzymes present in liver, kidneys and/or gut. OTA, ochratoxin A; OTB, ochratoxin B; OTC, ochratoxin C; OTα, ochratoxin α; OTβ, ochratoxin β, OTHQ, OT hydroquinone; 4R-OH-OTB, 4R-hydroxyochratoxin B; OP-OTA, lactone-opened OTA; 10-OH-OTA, 10-hydroxyochratoxin A; 4R-OH-OTA, 4R-hydroxyochratoxin A; 4S-OH-OTA, 4S-hydroxyochratoxin A; 4S-OH-OTB, 4S-hydroxyochratoxin B. The respective metabolite is the main metabolite in the underlined taxa (for rodents, data of different authors is contradictory). Genera in brackets represent those where the metabolite has been detected, although it may be present in other animals or humans as well. Summarised from [52,53,92].
Toxins 07 04253 g001
Lactone-opened OTA (OP-OTA) was detected as a metabolite in the bile in OTA-exposed (i.v.) rats, but not in the blood or the urine [86], whereas other researchers found it in bile and urine of OTA-exposed (i.v.) rats [96]. The mechanism of OP-OTA synthesis as well as its potential role in bioactivation of OTA still needs to be elucidated [39].
In all investigations, parent OTA was always the main compound detected followed by species-specific metabolites.

2.4. Detection Methods for Ochratoxins

Knowledge about the different ochratoxin group members begins with the ability to detect and characterise them. OTA was detected first in 1965 [1] and, with improving detection methods, more and more ochratoxin family members are being described with reports of newly detected metabolites as recently as this year [52].
The analytical methods typically used have been reviewed by various authors in recent years [69,97,98,99]. These methods are able to detect and distinguish between the various ochratoxin family members and metabolites, but are technically and financially demanding. In contrast, immunological methods are typically less expensive but can detect only one specific ochratoxin or cannot distinguish between different ochratoxins as comprehensively reviewed by Meulenberg [100]. Most of these assays have been developed for the detection of OTA, however, at least one highly specific monoclonal antibody against OTB has also been successfully produced [101,102,103,104,105]. Although official monitoring programmes preferentially use analytical methods such as HPLC and LC-MS, immunological methods can be equally sensitive and reproducible [100].

3. Differential Toxicity of Ochratoxin Group Members

The toxicological profile of OTA has been investigated in numerous studies and also extensively reviewed [6,7,54,63,69,70,71,72,73,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138]. In summary, these studies showed that OTA is nephrotoxic, hepatotoxic, neurotoxic, teratogenic and immunotoxic in various animals and in vitro, with renal toxicity and carcinogenesis being the key adverse effects. Many studies of OTA from the last decade support a non-DNA-reactive genotoxic mechanism, which involves different epigenetic mechanisms mainly related to oxidative stress, compensatory cell proliferation and disruption of cell signaling and division, and these publications have been thoroughly reviewed by various authors [6,63,70,113,119,139,140]. However, a direct genotoxic mechanism involving OTA bioactivation and DNA adduct formation is also still discussed [115,120] and this mechanism is also consistent with some in vivo gene expressionresults [113]. In summary, the mode(s) of action of OTA, in particular for renal carcinogenesis, are still under debate.
Various public health agencies and (inter)national authorities including the International Agency for Research on Cancer (IARC) [141], the European Food Safety Authority (EFSA) [8,142], the Joint FAO/WHO Expert Committee on Food Additives (JECFA) [128,143,144,145,146], the European Commission [5,147], the U.S. Department of Health and Human Services (National Toxicology Program, NTP) [148,149,150], and Health Canada [151,152] have evaluated the available data. Based on these evaluations, certain maximum limits for OTA in food have been set or proposed by the EU and Canada, ranging from 10 µg OTA kg−1 for dried fruits or instant coffee down to 0.5 µg OTA kg−1 for baby foods [6,147,153]. Egypt, Russia, China and Bosnia and Herzegovina have also established limits similar to that of European regulations [154,155]. OTA limits described in the Codex Alimentarius of the FAO/WHO have been adopted for example in the Arab States of the Gulf, Nigeria, Kenya and India. However, to date, no limits for OTA in foodstuffs have been set in many countries including the USA, Australia, New Zealand, Japan, Mexico or South Africa. Furthermore, there are no regulations for other ochratoxin group members in food and feed worldwide.

3.1. Comparative Toxicity of Ochratoxin Group Members

Comparative studies on more than two ochratoxins are very rare and no recent data are available. For example, Xiao et al. investigated toxicity using three different models [86]. In HeLa S3 cells, a cytotoxicity ranking (LC50 (µM)) of OTA (5) > OTC (9) > OTB (54) > d-OA (163) > OTα (560) > OM-OTA (830) > DC-OTA (7600) > OE-OTA (10,100) was found [86]. Similarly, the minimum toxic dose (nmol disc−1) was determined in an antibacterial assay using Brevibacillus brevis: OE-OTA (1.1) > OTC (2.0) > d-OTA (5.5) > OTA (8.7) > OTB (54) > M-OTα (90) > OTα (390) [86]. Furthermore, the in vivo toxicity was tested in mice (i.p., 72 h) [86]. Here, no toxicity ranking could be established based on the chosen doses. At doses of 500 mg kg−1 b.w. all animals died within 72 h of dosing with OE-OTA, DC-OTA, OM-OTA or M-OTα, whereas there was no lethality at 200 mg kg−1 b.w. [86]. In comparison, OTA showed 30% and 90% lethality at 20 and 50 mg kg−1 b.w., respectively, and d-OTA showed no lethality at 50 and 200 mg kg−1 b.w. [86]. Further structure–activity analysis resulted in the conclusion that the toxicity of OTA is associated with its isocoumarin moiety and most likely with the lactone carbonyl group, but not with the phenyl hydroxyl group nor the iron-chelating properties [86]. In another study, Müller et al. compared the effects of different ochratoxin group members on extracellular radical formation in porcine blood monocytes and granulocytes and presented the following ranking (IC50 in ng mL−1 shown in parenthesis): OTA (89.1) > OTC (154.2) > OTA methylester (285.5) > OTB (>1000) = OTα (>1000) [156].
In summary, based on the limited data from the above studies, no clear general toxicity ranking can be drawn; however, OTA seems to be overall the most toxic, followed by OTC, OTB and OTα.

3.2. Ochratoxin B (OTB)

Most of the published data on ochratoxins other than OTA describes OTB toxicity. In vivo, OTB appears to be much less toxic than OTA as tested in rats, fish and poultry [58,157,158]. The first comparative study was performed in 1-day-old chicks resulting in oral LD50 values of approximately 120 µg OTA (about 3.5 mg kg−1) and 1890 µg OTB (54 mg kg−1) [157]. Another early in vivo study in rainbow trout resulted in an LD50 (i.p.) of 5.53 mg OTA kg−1 b.w., whereas no deaths were observed with doses up to 66.7 mg OTB kg−1 b.w. [158]. Mally and co-workers showed in F344 rats that oral administration of OTB did not induce pronounced adverse effects (minor renal histopathology, no changes in clinical markers for renal damage) [58] whereas the same treatment with OTA resulted in necrosis and degeneration of the renal tubular epithelium [159] and altered renal function [58]. The same authors also provided an explanation for the observed lower in vivo renal OTB toxicity compared to OTA: this is due to an absence of specific retention in the rat kidney which leads to its differential toxicokinetics in vivo, including its more extensive metabolisation combined with a more rapid excretion than OTA [58,74]. In contrast, whole-body autoradiography revealed no difference in OTA and OTB distribution in rats [74,75]. Additional in vivo data was provided by Chattopadhyay and colleagues in a recent hematotoxicity study in Sprague-Dawley rats (250–320 g) [160]. All chosen hematological parameters were significantly changed after i.p. injection of 0.5 μg OTB when compared to vehicle-treated animals [160].
Much more data is available from in vitro studies, where the nephrotoxic, hepatotoxic and the immunotoxic effects have been investigated. Early studies on in vitro toxicity reported OTB to be less toxic than OTA [161,162]. For example, serum-free in vitro cytotoxicity testing in embryonic chick meningeal fibroblasts resulted in significantly higher toxicity (nearly 19-fold) of OTA compared to OTB after eight days of exposure as measured using the MTT or NR assay [162]. Cytotoxicity was also investigated in continuous kidney cell lines (LLC-PK1 and OK) with four cytotoxicity assay endpoints resulting in OTA being more toxic than OTB [161]. The acute toxicity of OTA and OTB was compared in HeLa cells, resulting in LC50 values of 54 µM and 5 µM for OTB and OTA, respectively [86]. More recent investigations have resulted in a more detailed picture, with some endpoints or cell models resulting in a lower or at least different toxicity of OTB compared to OTA [163] while others have suggested an equal toxicity [58]. For example, in the human-derived liver cell line HepG2, OTA but not OTB caused induction of micronuclei and DNA migration, whereas OTB inhibited cell division at concentrations even lower than OTA [163].
Some studies directly compared OTA and OTB toxicity. An exposure to acutely toxic concentrations (µM range) of OTA and OTB typically resulted in a lower toxicity of OTB compared to OTA [58,161], which seemed to support the general belief that OTB is less toxic than OTA. However, this difference is typically only apparent after rather short exposure times, with differences becoming less significant the longer the exposure time. The degree of difference is also dependent on the cell type used. More detailed investigations of the acute effects (24–96 h) in different renal cells of human, porcine and rat origin resulted in a more complicated picture [164,165,166,167] (Table 3).
Table 3. Comparative cytotoxicity of OTA and OTB.
Table 3. Comparative cytotoxicity of OTA and OTB.
Cell Type (Gender)96 h EC5024–96 h EC5048 h EC50
NRMTTCNNRMTTCNCV
HKC (f)OTA ↑OTA ↑OTA ↑n.d.n.d.n.d.OTA ↑
HKC (m)EqualEqualEqualEqualEqualEqualOTA ↑
PKC (f)EqualEqualEqualn.d.n.d.n.d.Equal
PKC (m)EqualEqualEqualEqualEqualEqualEqual
RPTC (f)OTA ↑OTA ↑n.d.n.d.n.d.n.d.n.d.
RPTC (m)OTA ↑OTA ↑n.d.n.d.n.d.n.d.n.d.
LLC-PK1OTA ↑OTA ↑EqualOTA ↑OTA ↑EqualOTA ↑
NRK-52En.d.n.d.n.d.n.d.n.d.n.d.OTA ↑
IHKEn.d.n.d.n.d.EqualEqualEqualn.d.
HKC, primary human kidney cells; PKC, primary porcine kidney cells; RPTC, primary rat kidney cells; LLC-PK1, porcine renal cell line; NRK-52E, rat renal cell line; IHKE, human renal cell line; f, female; m, male; NR, neutral red uptake; MTT, MTT reduction assay; CN, cell number (direct counting); CV, cell number (crystal violet staining); OTA ↑, higher cytotoxicity of OTA compared to OTB; Equal, equal cytotoxicity of OTA and OTB; n.d., not determined; summarised from [166,167,168].
Figure 2. Comparison of cellular uptake of [3H]OTA and [3H]OTB. PKC, primary porcine kidney cells (male); HKC, primary human kidney cells (male). Columns represent means from at least three independent replicates ± SEM; modified from O’Brien et al. with permission [169].
Figure 2. Comparison of cellular uptake of [3H]OTA and [3H]OTB. PKC, primary porcine kidney cells (male); HKC, primary human kidney cells (male). Columns represent means from at least three independent replicates ± SEM; modified from O’Brien et al. with permission [169].
Toxins 07 04253 g002
Equal toxicity of both ochratoxins in exposed LLC-PK1 cells was corroborated by LC-MS/MS measurement of intracellular levels of ochratoxins showing no difference in intracellular concentrations of OTA and OTB, indicating that, provided equal amounts of toxin are taken up by the cells, OTA and OTB may be equally cytotoxic [58]. These authors therefore confirmed the earlier hypothesis, that the differential toxicities observed in some cell types are likely to be due to differences in uptake and cellular protein binding [58], as studied in various renal models. For example, cellular uptake of [3H]OTB was shown to be consistently lower than [3H]OTA uptake [169] (Figure 2). Furthermore, primary renal cells accumulated more toxins than continuous cell lines with the difference being more prominent with OTA [168]. Binding experiments showed that OTA has a binding affinity to a range of renal proteins from humans, pigs, mice and rats with species-specific binding patterns [169,170,171,172] (Table 4). Specific [3H]OTB binding was observed to be consistently lower than that of [3H]OTA, however, the same binding rank-order was determined for both toxins, i.e., human > rat > pig > mouse [169,172]. Furthermore, the specific binding was at least 2.5 times higher with [3H]OTB, when compared with [3H]OTA [169,172].
Table 4. Comparison of [3H]OTA and [3H]OTB protein binding characteristics.
Table 4. Comparison of [3H]OTA and [3H]OTB protein binding characteristics.
Species (Gender)[3H]OTA[3H]OTBp
SB (pmol mg−1 Protein)SB (%)nSB (pmol mg−1 Protein)SB (%)n
Human (m)21.4 ± 4.520.5 ± 4.161.25 ± 0.1552.9 ± 10.730.0002
Human (f)23.1 ± 4.722.0 ± 4.343.79 ± 2.0473.6 ± 4.03<0.0001
Rat (m)10.2 ± 3.710.1 ± 3.4140.44 ± 0.4231.2 ± 15.08<0.0001
Rat (f)14.2 ± 6.311.5 ± 4.7100.73 ± 0.3943.3 ± 11.46<0.0001
Pig (m)3.4 ± 1.03.3 ± 0.980.19 ± 01127.2 ± 13.050.0002
Pig (f)2.1 ± 0.72.0 ± 0.68<LOD<LOD2n.c.
Mouse (m)0.9 ± 020.9 ± 0.260.1614.31n.c.
Mouse (f)1.5 ± 0.61.5 ± 0.59<LOD<LOD2n.c.
Means ± SD; SB, specific binding; n, number of replicates; LOD, limit of detection; p values are shown from two-tailed t-tests and refer to the comparison of SB(%); n.c., not computable; modified from O’Brien et al. with permission [169].
The binding data obtained ex vivo correlated well with the in vivo toxicokinetic profiles of OTA which were studied for example in fish, quail, mouse, rat and monkey [76]. The elimination half-life varied from 0.68 h after oral administration to fish, up to 840 h after intravenous administration to monkey [76]. A comparative study in rats showed that the biological half-life (t1/2) as well as the maximal serum concentration was different with OTA and OTB [70]. OTA showed a t1/2 of 170 h (i.v.) or 120 h (p.o.), whereas that of OTB was much shorter, i.e., 12 h (i.v.) or 18 h (p.o.) [76]. The maximal serum concentrations were 2100 ng mL−1 (i.v.) or 390 ng mL−1 (p.o.) and 760 ng mL−1 (i.v.) or 120 ng mL−1 (p.o.) for OTA and OTB, respectively. Furthermore, the binding parameters were determined by equilibrium dialysis resulting in unbound fractions of ochratoxins in rat (0.02% OTA vs. 0.9% OTB), monkey (0.08% OTA vs. 2.7% OTB), and humans (0.02% OTA vs. 2% OTB) [76]. Plasma protein binding of OTA was studied in various animals after the oral or intravenous administration of 50 ng g−1 b.w. resulting in a free fraction of toxin in plasma of <0.2% in all subjects investigated except fish where the equivalent figure was 22% [76].
Dietrich and colleagues performed further testing with respect to the reversal of toxic effects. After 48 h of exposure (10 µM) with subsequent replacement of the medium, they demonstrated recovery from the cytotoxic effect for OTA in some cell models, i.e., in LLC-PK1, NRK-52E and NRK-49F, but not in human and porcine primary kidney cells [166]. In contrast, only the fibroblast cells (NRK-49F) were able to recover from the toxic effects of OTB [168]. These observations further support the conclusion of differential cytotoxicity of OTA and OTB in different cell models.
Comparative studies were also undertaken with respect to chronic toxicity. For this, an in vitro model of sub-chronic repeated exposure to OTA and OTB was employed to generate ochratoxin-derived subpopulations of human and porcine proximal tubular cells (HKC, IHKE, PKC, LLC-PK1) [167,173,174,175]. Comparison of growth characteristics showed marked changes in almost all cell types, however, a comparison between OTA and OTB showed no difference between the different cell subpopulations [167]. Acute challenge experiments with these cell subpopulations resulted in generally decreased sensitivity towards OTA- and OTB-mediated cytotoxicity [167].
The immunotoxic effects of OTB were studied in isolated human neutrophils where all parameters studied were decreased at µg mL−1 concentrations, indicating a weakened immune defence [176,177]. In porcine monocytes, extracellular radical formation was inhibited by OTA almost completely at 1 µg mL−1, whereas the inhibition by OTB was lower [156]. OTA impaired the IL-2 production and IL-2 receptor expression of activated T lymphocytes, which was prevented when the cells are pre-incubated with OTB prior to OTA exposure [178].
Other comparative toxicity investigations included the examination of mutagenicity and genotoxicity. For example, OTA induced SOS-DNA repair without metabolic activation at concentrations of 0.1–4 mM in E. coli, whereas OTB up to 4 mM was inactive [93,179]. The same was shown for induction of micronuclei and DNA migration assays, whereas both toxins failed to induce mutations in Salmonella/microsome assays [163].
Differential toxicity has also been shown for OTA and OTB in developmental toxicity models. OTB and OTA both caused craniofacial malformations in the FETAX (Frog Embryo Teratogenicity Assay Xenopus) with the effects of OTA being more severe than those of OTB [180]. OTA also caused reduced embryo growth, whereas OTB did not [180]. The authors suggested that this differential embryotoxicity is at least in part due to the higher levels of OTA being accumulated within the embryos as shown by uptake experiments using [3H]OTA and [3H]OTB [180]. Investigations by other authors revealed that OTA and OTB also inhibited differentiation and protein synthesis in prechondrogenic mesenchymal cells from the limb buds of 4-day chick embryos with IC50 values of 1.9 µM and 6.2 µM for OTA and OTB, respectively [181]. Furthermore, OTB had no effect on the aminoacylation of phenylalanine tRNA catalysed by mice liver phenylalanyl-tRNA synthetase, whereas OTA showed a concentration-dependent effect [182]. Furthermore, OTB showed no antagonistic effect on the in vivo protein synthesis inhibition caused by OTA in hepatoma tissue culture cells [182].
In summary, today’s knowledge on comparative toxicity of OTA and OTB must differentiate between in vivo and in vitro effects. OTB is clearly less toxic in vivo compared to OTA as shown in various models. It has been shown that OTB is more readily eliminated and had a lower affinity for plasma proteins, which partly may explain its lower toxicity. In contrast, both toxins can be similarly acutely cytotoxic in vitro provided that similar amounts are taken up and are intracellularly bound, although several results may suggest a different molecular mechanism of chronic toxicity compared to OTA. Furthermore it can be assumed that the small structural difference (chlorine in OTA vs. hydrogen in OTB) although not responsible for the toxicity, may be crucial for the differential uptake and binding in cells.

3.3. Ochratoxin C (OTC) and Other Ethyl or Methyl Esters of Ochratoxins

In an early study, oral LD50 values were reported for OTC (216 mg animal−1) and OTA (166 mg animal−1) in day-old chicks [183]. Later, the acute cytotoxicities of OTA and OTC were compared in HeLa S3 cells, resulting in comparable LC50 values of 9 µM and 5 µM for OTC and OTA, respectively [86]. Furthermore, an antibacterial assay with B. brevis resulted in minimum toxic doses (MTDs, nmol disc−1) of 8.7 ± 1.7 and 2.0 ± 0.5 for OTA and OTC, respectively [86]. OTC was also identified as comparably cytotoxic to OTA in human embryonic (HEK293) and renal carcinoma (A498) cell lines [184]. Based on the above results, OTC is considered to be similarly toxic to OTA.
Further data have shown that OTC and OTA exposure leads to various immunomodulatory effects (e.g., metabolic activity, mitogen-induced proliferation, ROS formation, production of the cytokines IL-6 and TNF-α, phagocytic behaviour, nitrogen oxide synthesis and cell surface markers) in porcine mononuclear cells [185] and in the human monocyte/macrophage line THP-1 [186] at low concentrations (10–1000 ng mL−1). Similar concentrations of OTA and OTC suppressed significantly the formation of free oxygen radicals by porcine monocytes and granulocytes [156]. Long-term exposure of THP-1 cells to OTC and OTA (15 days, 1 ng mL−1) resulted in increased mitochondrial activity and production of IL-6, disturbed cell membrane integrity and inhibited cell proliferation and TNF-α and IL-8 production [187].
An early in vivo study in young rainbow trout with ethyl esters of OTA and OTB showed that these compounds were lethal to trout when administered i.p. over 10 days with LD50 values of 3.0 and 13.0 mg kg−1 b.w. for OTA and OTB ethyl esters, respectively [158]. OTα ethyl ester was non-toxic at a level of 3.9 mg kg−1 b.w., which is equivalent to 13.72 µmol kg−1 (=LD50 of OTA) [158]. In comparison to OTA, the methyl ester of OTA was less toxic than OTA in day-old chicks [183]. Also, OTB methyl and ethyl esters were found to be non-lethal to orally exposed day-old ducklings [188].
In summary, OTC seems to be similarly acutely toxic in vivo and in vitro compared to OTA, however, whether the mode of action of OTC and OTA is the same remains to be elucidated. Other ochratoxin ethyl or methyl esters show lower toxicity compared to OTA.

3.4. Ochratoxin α (OTα) and Ochratoxin β (OTβ)

Unfortunately, there are not many published studies including data on OTα and OTβ and most of the available data considers OTα only. For OTβ, only its occurrence as metabolite has been investigated. To our knowledge, no toxicity data is available. However, based on its structure, it may be assumed that OTβ shows similar behaviour to OTα.
Creppy and co-workers investigated the immunotoxic effect of OTα in BALB/c mice using an immunosuppression assay after exposure to a single dose of OTα of 1 µg kg−1 i.p. [85]. Here, OTA caused an approximately 90% reduction of the IgM and IgG response, whereas OTα showed no effect [85].
The same researchers reported that the growth rate of Morris hepatoma cells (HTC cells) was massively decreased with OTA, whereas OTα had no effect on proliferation [189]. The acute toxicity was also compared in HeLa cells, resulting in EC50 values of 560 µM and 5 µM for OTα and OTA, respectively [86]. A recent report compared the acute cytotoxicity (24 h) in IHKE cells and calculated an EC50 of 0.5 µM for OTA, whereas OTα was non-toxic up to 50 µM [190].
The neurotoxic effects were also investigated with OTA showing pronounced inhibition of various parameters (dehydrogenase activity, lysosomal activity, total cell culture protein, neurofilament 68 kD content) in cultures of embryonic brains of Tetra SL chick stage 29 at low nM concentrations and 8 days of exposure, whereas OTα showed no effect up to 15 µM [55]. In addition, ochracin (also termed mellein) was tested, resulting in weak cytotoxic effects at high concentrations (µM-mM) [55].
Others investigated genotoxic effects, resulting in induction of SOS-DNA repair in the E. coli PQ37 strain in the absence of an exogenous metabolic activation system at concentrations of 0.1–4 mM OTA, whereas OTα was inactive [179]. OTA induced also sister chromatid exchanges in cultured primary porcine urinary bladder epithelial cells (PUBEC) after 5 h of exposure at pM to nM concentrations without apparent cytotoxicity [191]. The same effect was observed with OTα, although higher concentrations were necessary (up to 10 µM) [191].
OTα is much less toxic (approximately 100 fold) than OTA as shown in various investigations [85,86]. Apparently, the isocoumarin moiety alone is not effective but must be bound to phenylalanine to exert toxic effects.

3.5. Hydroxyochratoxins

Also, for hydroxyochratoxins, only a very limited amount of data exists. Some data exist for 4R-OH-OTA: this was shown to be equally cytotoxic compared to OTA in Morris hepatoma cells (HTC cells) and this toxicity could be prevented by phenylalanine [189]. Furthermore, both OTA and 4R-OH-OTA inhibit protein synthesis, which could be prevented by phenylalanine and could be attributed to their binding on the phenylalanine sites of phenylalanyl-tRNA synthetase [189]. The same group also investigated the immunotoxic effects in BALB/c mice using an immunosuppression assay, resulting in similar toxic effects of 4R-OH-OTA and OTA on the IgM and IgG response [85]. 4-OH OTA resulted in no mortality after six days when injected i.p. into male Wistar rats at a dose of 40 mg kg−1 b.w. [192]. 10-OH OTA was found to be non-genotoxic (DNA adduct formation) in bronchial epithelial cells (WI 126 VA) [81,88]. Finally, no toxicity data are available for 4R-hydroxyochratoxin B (4R-OH-OTB) and 4S-hydroxyochratoxin B (4S-OH-OTB) to date. Other hydroxyochratoxins (5′-OH-OTA, 7′-OH-OTA, 9′-OH-OTA), which differ from OTA in the hydroxylation at specific sites of the phenylalanyl group, were detected as metabolites in vitro and in vivo just very recently [52] and no toxicity data are available to present.
Based on some of the results above, it may be concluded that hydroxylation of ochratoxins (in particular 4R-OH-OTA) does not obviously affect their toxicity.

3.6. Ochratoxin Quinone (OTQ)/Hydroquinone (OTHQ)

It has been shown that OTA undergoes an oxidative dechlorination process to generate a quinone (OTQ)/hydroquinone (OTHQ) redox couple like other chlorinated phenols, which may play a role in OTA-mediated toxicity [56,193]. Indeed, OTHQ generated DNA adducts in the absence of metabolic activation in human bronchial epithelial (WI26) and human kidney (HK2) cells. These results were obtained using the [32P]-postlabeling technique and the necessary confirmation by mass spectrometry is still lacking [56]. However, other researchers did not find OTAQ/OTAHQ metabolites in liver and kidney microsomes from rat or mouse [87,194].
In summary, data on OTAQ/OTAHQ metabolites are still too limited to provide meaningful conclusions.

3.7. Lactone-Opened OTA (OP-OTA)

The in vivo toxicity of OP-OTA was tested in rats and mice. OP-OTA was more toxic than OTA in rats when administered i.v. (3/4 rats died within 6 h vs. 2/4 with OTA at 500 µg animal−1 (200 ± 15 g b.w.) [86]. Interestingly, no lethality was observed after i.p. injection [86]. The same effect was observed in mice (6 weeks old, n = 10), where no lethality after 72 h at i.p. doses up to 500 mg kg−1 b.w. occurred, whereas in the same experiments OTA resulted in 90% dead animals after injections of 50 mg kg−1 b.w. [86]. These authors suggested that the more polar OP-OTA is, the less readily it is taken up in the rodents when given i.p. compared to OTA [86]. Whether other factors may be involved such as enhanced metabolism (first pass effect) or elimination was not investigated.
In addition, no toxicity was detected in Brevibacillus brevis at a concentration of 350 nmol/disc, whereas the minimum toxic dose for OTA was 8.7 ± 1.7 nmol disc−1 [86].
Overall, the available data suggests that the toxicity of ochratoxins may be related to the lactone carbonyl group.

3.8. Conjugated Ochratoxin Forms

To date, no toxicity data has been reported for glucuronidated ochratoxins or pentose and hexose conjugates. Only in bacteria (E. coli PQ37), the formation of a cytotoxic thiol-containing derivative was suggested [93]. Conjugation is typically a detoxification step, however, bioactivation might be possible.

3.9. Synthetic OTA Analogues

Investigations with synthetic analogues of OTA mostly included substitutions of phenylalanine with other amino acids. For example, in an early study, the alanine and leucine analogues of OTA were i.p. administered to trout and caused no death at dose levels of 4.49 and 5.06 mg kg−1 b.w., respectively [158]. These doses were equivalent to 13.72 µmol OTA kg−1 b.w. (=LD50 of OTA) [158]. Later, Creppy et al. synthesised eight different analogues of OTA [195]. Their effects were investigated in comparison to OTA, OTB and OTα on yeast tRNA amino acylation and on growth and protein synthesis of HTC cells [195]. All analogues showed inhibitory effects in the three test systems, although to a lesser degree than OTA, whereas OTB and OTα were ineffective [195].
Based on these results, it can again be concluded that phenylalanine is important for the toxicity of OTA, as the toxicity was always reduced if phenylalanine was substituted by another amino acid.

3.10. Structure–Activity Relationship

In summary, current knowledge about the toxicity of ochratoxins suggests that the hydroxyl, carboxyl, chlorine, and lactone groups of ochratoxins substantially affect their toxicities in various models. Furthermore, the biological reactivity of ochratoxins seems to be enhanced by the presence of the phenylalanine moiety, although this group is not essential for toxicity.

4. Toxicity of Mixtures of Mycotoxins

In nature, co-occurrence of mycotoxins is often observed. One mould species may produce many different mycotoxins, and the same mycotoxin may be produced by several species [110,196,197,198,199,200]. Considering this coincident production, it is very likely that humans and animals are always exposed to mixtures rather than to individual compounds. This is particularly true for OTA, OTB, citrinin, penicillic acid and occasionally for patulin as these are all produced by a number of Penicillium and Aspergillus species [110,200]. Furthermore, other mycotoxins such as fumonisins and aflatoxins have been frequently detected together with OTA [62,110,199]. Currently, little is known about mycotoxin interactions although combined exposure is clearly more relevant to real-life situations [2,3,110,114,200]. Further challenges may occur worldwide due to climate change as this may influence the occurrence of mycotoxins due to geographic shifts in the distribution of major cereal cropping systems and potentially favourable climatic conditions for toxin producing organisms [201,202].
Data on combination toxicity is still rather limited and often contradictory, however, the available data on OTA toxicity in combination with other mycotoxins was comprehensively reviewed in 2013 [110]. In summary, most of the studies addressing the combined effect of OTA with other mycotoxins showed additive or synergistic interactions when binary mixtures were tested and data on multiple mixtures is very rare [110]. Since the publication of that review, a few more reports have been published and these are summarised below [203,204,205,206,207].
Rumora et al. investigated the individual and combined toxicity of OTA and citrinin in porcine proximal tubular cells (PK15 cells) and showed that mitogen-activated protein kinases (MAPKs) were differentially activated [203]. Based on their results, the authors suggested that Ca2+ might be an important player in the observed in vitro toxicity [203]. These toxins were also tested by Anninou et al. on human hepatoma cells (Hep3B), in combination with sterigmatocystin, a precursor of aflatoxin biosynthesis, which resulted in additive or antagonistic genotoxic and cytotoxic effects at low concentrations (pM-nM) [208]. Föllmann and colleagues tested OTA in combination with citrinin and its main human metabolite dihydrocitrinone (DH-CIT) in Chinese hamster lung fibroblasts (V79 cells) using the neutral red uptake assay, the in vitro micronucleus assay and flow cytometry [205]. The main outcome of this study, i.e., that mixtures of OTA and citrinin exerted mainly additive effects and that synergism was observed with high micromolar concentrations only [205], confirms some of the earlier results obtained in other cells and/or with other endpoints [209,210,211,212]. Similar results with OTA and citrinin were obtained by Gayathri et al. in human hepatocarcinoma cells (HepG2) [213].
Clarke and co-workers investigated OTA in combination with aflatoxin B1 (AFB1) and fumonisin B1 (FB1) in human intestinal cells (Caco-2), Madin Darby bovine kidney cells (MDBK) and murine macrophage cells (Raw 264.7) using the MTT and neutral red assays [207]. Binary combinations showed additive effects, whereas tertiary combinations of OTA, FB1 and AFB1 resulted in synergistic effects [207]. Further studies employed more sensitive high content analysis (HCA) resulting in synergistic interactions of binary mixtures of OTA and FB1 [204]. In contrast, recent in vivo investigations by Corcuera et al. of the combined toxicity of OTA and AFB1 in orally exposed male F344 rats resulted in an antagonistic genotoxic interaction as tested via the micronucleus and comet assays [214].
Li and co-workers investigated the liver and ovarian toxicity of OTA in combination with the mycotoxin zearalenone in human hepatoma cells (HepG2) and murine ovarian granular cells (KK-1) [206]. Exposure to equi-effective mixtures resulted in additive effects with respect to cell viability (WST-8 assay), whereas differential effects were observed with the endpoint of ROS generation [206]. Further research included also other targets such as intestinal cells or lung cells, for example, Cano-Sancho and co-workers reported an increased cytotoxicity of binary combinations of OTA and deoxynivalenol on Caco-2 cells [215]. Šegvić Klarić et al. investigated subtoxic spore extracts from typically co-occurring grain mill aeromycota in combination with OTA on human lung adenocarcinoma cells (A549) and observed significant DNA damage and cell death with additive interactions [216].
In summary, the combined effect of OTA with other mycotoxins showed mostly additive or synergistic interactions. Depending on the concentrations applied and the in vitro models employed, however, antagonistic interactions have also been detected. Data on multiple mixtures are still incomplete.

5. Summary and Final Remarks

The biosynthesis as well as biotransformation of ochratoxins has not been elucidated in detail so far. Several naturally occurring forms, metabolites and conjugates have been reported and characterised in vitro and/or in vivo, while the toxicity of other detected forms remains to be described. Based on the publication record on ochratoxin metabolism, it can be concluded that humans and animals are mainly exposed to the ingested parent compounds and to a much lesser extent to their (species-specific) metabolites.
The current toxicological database indicates the crucial importance of certain structural components such as the hydroxyl, carboxyl, chlorine, and lactone groups of ochratoxins with respect to cellular uptake and binding. The bioavailability seems to be further enhanced by the presence of the phenylalanine moiety, although this group is not essential for toxicity. The structural differences of the ochratoxin group members may explain most of the experimental outcomes observed in various models. Nevertheless, the contribution of metabolites to OTA toxicity is still rather unclear and further research is needed.
The underlying mechanisms of toxicity of the ochratoxin group members still need to be elucidated. Although recent advances mainly support a non-DNA-reactive, genotoxic mechanism for OTA, there is also evidence of other mechanisms involving OTA bioactivation and OTA-DNA adduct formation. Whether or not other ochratoxin group members show the same mode of action remains to be investigated. However, there are indications that at least OTB may have a different molecular mechanism of chronic toxicity compared to OTA.
Given the potential chronic human exposure to the naturally occurring forms, metabolites, conjugates and mixtures with other mycotoxins, a better understanding of the toxicokinetics and the mechanisms of toxicity of the ochratoxin group is necessary to provide adequate human risk assessments.

Acknowledgments

We would like to thank Evelyn O’Brien for critical reading of the manuscript.

Authors Contributions

Alexandra H. Heussner drafted the manuscript with contributions from Lewis E. H. Bingle. Both authors have read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

b.w., body weight; (B)EN, (Balkan) Endemic Nephropathy; GSH, glutathione; FETAX, Frog Embryo Teratogenicity Assay Xenopus; CAS, Chemical Abstracts Service; EC50, mean effective concentration; EU, European Union; FAO, Food and Agriculture Organization of the United Nations; IgG/M, immunoglobulin G/M; i.p., intraperitoneal; i.v., intravenous; LC50, median lethal concentration; LD50, median lethal dose; MPN, Mycotoxic Porcine Nephropathy; p.o., per os (orally); MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; WHO, World Health Organization.

References

  1. Van der Merwe, K.; Steyn, P.; Fourie, L.; Scott, D.; Theron, J. Ochratoxin A, a toxic metabolite produced by Aspergillus ochraceus Wilh. Nature 1965, 205, 1112–1113. [Google Scholar] [CrossRef] [PubMed]
  2. O’Brien, E.; Dietrich, D. Mycotoxins affecting the kidney. In Toxicology of Kidney; Hook, J., Ed.; CRC Press: Boca Raton, FL, USA, 2004; pp. 895–936. [Google Scholar]
  3. Streit, E.; Schatzmayr, G.; Tassis, P.; Tzika, E.; Marin, D.; Taranu, I.; Tabuc, C.; Nicolau, A.; Aprodu, I.; Puel, O.; et al. Current situation of mycotoxin contamination and co-occurrence in animal feed-focus on Europe. Toxins 2012, 4, 788–809. [Google Scholar] [CrossRef] [PubMed]
  4. Bellver Soto, J.; Fernández-Franzón, M.; Ruiz, M.; 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]
  5. EC. Assessment of Dietary Intake of Ochratoxin A by the Population of EU Member States; Report of the Scientific Cooperation, Task 3.2.7; Directorate-General Health and Consumer Protection, European Commission: Rome, Italy, 2002. [Google Scholar]
  6. Haighton, L.A.; Lynch, B.S.; Magnuson, B.A.; Nestmann, E.R. A reassessment of risk associated with dietary intake of ochratoxin A based on a lifetime exposure model. Crit. Rev. Toxicol. 2012, 42, 147–168. [Google Scholar] [CrossRef] [PubMed]
  7. O’Brien, E.; Dietrich, D.R. Ochratoxin A: The continuing enigma. Crit. Rev. Toxicol. 2005, 35, 33–60. [Google Scholar] [CrossRef] [PubMed]
  8. European Food Safety Authority (EFSA). Opinion of the scientific panel on contaminants in the food chain on a request from the commission related to ochratoxin A in food. EFSA J. 2006, 365, 1–56. [Google Scholar]
  9. Duarte, S.; Lino, C.; Pena, A. Food safety implications of ochratoxin A in animal-derived food products. Vet. J. 2012, 192, 286–292. [Google Scholar] [CrossRef] [PubMed]
  10. FAO. Recommended practices for the prevention of mycotoxins in food, feed and their products. FAO Food Nutr. Pap. 1979, 10, 1–71. [Google Scholar]
  11. Visconti, A.; Bottalico, A. High levels of ochratoxins A and B in moldy bread responsible for mycotoxicosis in farm animals. J. Agric. Food Chem. 1983, 31, 1122–1123. [Google Scholar] [CrossRef] [PubMed]
  12. Huff, W.E.; Wyatt, R.D.; Tucker, T.L.; Hamilton, P.B. Ochratoxicosis in the broiler chicken. Poult. Sci. 1974, 53, 1585–1591. [Google Scholar] [CrossRef] [PubMed]
  13. Hamilton, P.B.; Huff, W.E.; Harris, J.R.; Wyatt, R.D. Natural occurrences of ochratoxicosis in poultry. Poult. Sci. 1982, 61, 1832–1841. [Google Scholar] [CrossRef] [PubMed]
  14. Dwivedi, P.; Burns, R.B.; Maxwell, M.H. Ultrastructural study of the liver and kidney in ochratoxicosis A in young broiler chicks. Res. Vet. Sci. 1984, 36, 104–116. [Google Scholar] [PubMed]
  15. Dwivedi, P.; Burns, R.B. Pathology of ochratoxicosis A in young broiler chicks. Res. Vet. Sci. 1984, 36, 92–103. [Google Scholar] [PubMed]
  16. Kozaczynski, W. Experimental ochratoxicosis A in chickens. Histopathological and histochemical study. Arch. Vet. Pol./Pol. Acad. Sci. Comm. Vet. Sci. 1994, 34, 205–219. [Google Scholar]
  17. Elaroussi, M.A.; Mohamed, F.R.; el Barkouky, E.M.; Atta, A.M.; Abdou, A.M.; Hatab, M.H. Experimental ochratoxicosis in broiler chickens. Avian Pathol. J. WVPA 2006, 35, 263–269. [Google Scholar] [CrossRef] [PubMed]
  18. Stoev, S.; Dutton, M.; Njobeh, P.; Mosonik, J.; Steenkamp, P. Mycotoxic nephropathy in Bulgarian pigs and chickens: Complex aetiology and similarity to Balkan endemic nephropathy. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2010, 27, 72–88. [Google Scholar] [CrossRef] [PubMed]
  19. Elling, F.; Moller, T. Mycotoxic nephropathy in pigs. Bull. World Health Organ. 1973, 49, 411–418. [Google Scholar] [PubMed]
  20. Stoev, S.; Denev, S. Porcine/chicken or human nephropathy as the result of joint mycotoxins interaction. Toxins 2013, 5, 1503–1530. [Google Scholar] [CrossRef] [PubMed]
  21. Stoev, S.; Denev, S.; Dutton, M.; Njobeh, P.; Mosonik, J.; Steenkamp, P.; Petkov, I. Complex etiology and pathology of mycotoxic nephropathy in South African pigs. Mycotoxin Res. 2010, 26, 31–46. [Google Scholar] [CrossRef] [PubMed]
  22. Krogh, P. Epidemiology of mycotoxic porcine nephropathy. Nord. Vet. Med. 1976, 28, 452–458. [Google Scholar] [PubMed]
  23. Stoev, S.; Gundasheva, D.; Zarkov, I.; Mircheva, T.; Zapryanova, D.; Denev, S.; Mitev, Y.; Daskalov, H.; Dutton, M.; Mwanza, M.; et al. Experimental mycotoxic nephropathy in pigs provoked by a mouldy diet containing ochratoxin A and fumonisin B1. Exp. Toxicol. Pathol. 2012, 64, 733–741. [Google Scholar] [CrossRef] [PubMed]
  24. Stoev, S.; Vitanov, S.; Anguelov, G.; Petkova-Bocharova, T.; Creppy, E. Experimental mycotoxic nephropathy in pigs provoked by a diet containing ochratoxin A and penicillic acid. Vet. Res. Commun. 2001, 25, 205–223. [Google Scholar] [CrossRef] [PubMed]
  25. Goliński, P.; Hult, K.; Grabarkiewicz-Szczesna, J.; Chełkowski, J.; Kneblewski, P.; Szebiotko, K. Mycotoxic porcine nephropathy and spontaneous occurrence of ochratoxin A residues in kidneys and blood of Polish swine. Appl. Environ. Microbiol. 1984, 47, 1210–1212. [Google Scholar] [PubMed]
  26. Stoev, S. The role of ochratoxin A as a possible cause of Balkan endemic nephropathy and its risk evaluation. Vet. Hum. Toxicol. 1998, 40, 352–360. [Google Scholar] [PubMed]
  27. Abid, S.; Hassen, W.; Achour, A.; Skhiri, H.; Maaroufi, K.; Ellouz, F.; Creppy, E.; Bacha, H. Ochratoxin A and human chronic nephropathy in Tunisia: Is the situation endemic? Hum. Exp. Toxicol. 2003, 22, 77–84. [Google Scholar] [CrossRef] [PubMed]
  28. Hmaissia Khlifa, K.; Ghali, R.; Mazigh, C.; Aouni, Z.; Machgoul, S.; Hedhili, A. Ochratoxin A levels in human serum and foods from nephropathy patients in Tunisia: Where are you now? Exp. Toxicol. Pathol. 2012, 64, 509–512. [Google Scholar] [CrossRef] [PubMed]
  29. Grollman, A.P.; Jelaković, B. Role of environmental toxins in Endemic (Balkan) Nephropathy. J. Am. Soc. Nephrol. 2007, 18, 2817–2823. [Google Scholar] [CrossRef] [PubMed]
  30. Maaroufi, K.; Pfohl-Leszkowicz, A.; Achour, A.; el May, M.; Grosse, Y.; Hammami, M.; Ellouz, F.; Creppy, E.; Bacha, H. Ochratoxin A genotoxicity, relation to renal tumors. Arch. Inst. Pasteur Tunis 1994, 71, 21–31. [Google Scholar] [PubMed]
  31. Pepeljnjak, S.; Klarić, M. «Suspects» in etiology of endemic nephropathy: Aristolochic acid versus mycotoxins. Toxins 2010, 2, 1414–1427. [Google Scholar] [CrossRef] [PubMed]
  32. Gallo, A.; Knox, B.P.; Bruno, K.S.; Solfrizzo, M.; Baker, S.E.; Perrone, G. Identification and characterization of the polyketide synthase involved in ochratoxin A biosynthesis in Aspergillus carbonarius. Int. J. Food Microbiol. 2014, 179, 10–17. [Google Scholar] [CrossRef] [PubMed]
  33. Rodríguez, A.; Medina, Á.; Córdoba, J.J.; Magan, N. The influence of salt (NaCl) on ochratoxin A biosynthetic genes, growth and ochratoxin A production by three strains of Penicillium nordicum on a dry-cured ham-based medium. Int. J. Food Microbiol. 2014, 178, 113–119. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, L.; Wang, Y.; Wang, Q.; Liu, F.; Selvaraj, J.N.; Liu, L.; Xing, F.; Zhao, Y.; Zhou, L.; Liu, Y. Functional characterization of new polyketide synthase genes involved in ochratoxin A biosynthesis in Aspergillus ochraceus fc-1. Toxins 2015, 7, 2723–2738. [Google Scholar] [CrossRef] [PubMed]
  35. Färber, P.; Geisen, R. Analysis of differentially-expressed ochratoxin A biosynthesis genes of Penicillium nordicum. Eur. J. Plant Pathol. 2004, 110, 661–669. [Google Scholar] [CrossRef]
  36. Geisen, R.; Schmidt-Heydt, M.; Karolewiez, A. A gene cluster of the ochratoxin A biosynthetic genes inPenicillium. Mycotox Res. 2006, 22, 134–141. [Google Scholar] [CrossRef] [PubMed]
  37. Sartori, D.; Massi, F.P.; Ferranti, L.S.; Fungaro, M.H. Identification of genes differentially expressed between ochratoxin-producing and non-producing strains of Aspergillus westerdijkiae. Indian J. Microbiol. 2014, 54, 41–45. [Google Scholar] [CrossRef] [PubMed]
  38. Huffman, J.; Gerber, R.; Du, L. Recent advancements in the biosynthetic mechanisms for polyketide-derived mycotoxins. Biopolymers 2010, 93, 764–776. [Google Scholar] [CrossRef] [PubMed]
  39. 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]
  40. O’Callaghan, J.; Caddick, M.X.; Dobson, A.D. A polyketide synthase gene required for ochratoxin A biosynthesis in Aspergillus ochraceus. Microbiology 2003, 149, 3485–3491. [Google Scholar] [CrossRef] [PubMed]
  41. O’Callaghan, J.; Stapleton, P.C.; Dobson, A.D. Ochratoxin A biosynthetic genes in Aspergillus ochraceus are differentially regulated by pH and nutritional stimuli. Fungal Genet. Biol. 2006, 43, 213–221. [Google Scholar] [CrossRef] [PubMed]
  42. Bacha, N.; Atoui, A.; Mathieu, F.; Liboz, T.; Lebrihi, A. Aspergillus westerdijkiae polyketide synthase gene “aoks1” is involved in the biosynthesis of ochratoxin A. Fungal Genet. Biol. 2009, 46, 77–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Karolewiez, A.; Geisen, R. Cloning a part of the ochratoxin A biosynthetic gene cluster of Penicillium nordicum and characterization of the ochratoxin polyketide synthase gene. Syst. Appl. Microbiol. 2005, 28, 588–595. [Google Scholar] [CrossRef] [PubMed]
  44. Abbas, A.; Valez, H.; Dobson, A.D. Analysis of the effect of nutritional factors on OTA and OTB biosynthesis and polyketide synthase gene expression in Aspergillus ochraceus. Int. J. Food Microbiol. 2009, 135, 22–27. [Google Scholar] [CrossRef] [PubMed]
  45. Varga, J.; Kevei, E.; Rinyu, E.; Téren, J.; Kozakiewicz, Z. Ochratoxin production by Aspergillus species. Appl. Environ. Microbiol. 1996, 62, 4461–4464. [Google Scholar] [PubMed]
  46. Harris, J.; Mantle, P. Biosynthesis of ochratoxins by Aspergillus ochraceus. Phytochemistry 2001, 58, 709–716. [Google Scholar] [CrossRef]
  47. Davis, N.; Searcy, J.; Diener, U. Production of ochratoxin A by Aspergillus ochraceus in a semisynthetic medium. Appl. Microbiol. 1969, 17, 742–744. [Google Scholar] [PubMed]
  48. Trenk, H.; Butz, M.; Chu, F. Production of ochratoxins in different cereal products by Aspergillus ochraceus. Appl. Microbiol. 1971, 21, 1032–1035. [Google Scholar] [PubMed]
  49. Mantle, P.G.; Chow, A.M. Ochratoxin formation in Aspergillus ochraceus with particular reference to spoilage of coffee. Int. J. Food Microbiol. 2000, 56, 105–109. [Google Scholar] [CrossRef]
  50. Hesseltine, C.W. Solid State Fermentations. Biotechnol. Bioeng. 1972, 9, 517–532. [Google Scholar] [CrossRef] [PubMed]
  51. Azpilicueta, C.A.; Arbeloa, M.I.; de Maquirriain, P.F.J.; Moreno, N.J. Study of ochratoxin A in wines from sun-dired grapes and in commercial wines from different geographical origins. In Food Chemistry Research Developments; Papadopoulos, K., Ed.; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2008; pp. 113–139. [Google Scholar]
  52. 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]
  53. Wu, Q.; Dohnal, V.; Huang, L.; Kuča, K.; Wang, X.; Chen, G.; Yuan, Z. Metabolic pathways of ochratoxin A. Curr. Drug Metab. 2011, 12, 1–10. [Google Scholar] [CrossRef] [PubMed]
  54. El Khoury, A.; Atoui, A. Ochratoxin A: General overview and actual molecular status. Toxins 2010, 2, 461–493. [Google Scholar] [CrossRef] [PubMed]
  55. Bruinink, A.; Rasonyi, T.; Sidler, C. Differences in neurotoxic effects of ochratoxin A, ochracin and ochratoxin-α in vitro. Nat. Toxins 1998, 6, 173–177. [Google Scholar] [CrossRef]
  56. 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]
  57. Han, Z.; Tangni, E.K.; di Mavungu, J.D.; Vanhaecke, L.; de Saeger, S.; Wu, A.; Callebaut, A. In vitro glucuronidation of ochratoxin A by rat liver microsomes. Toxins 2013, 5, 2671–2685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Mally, A.; Keim-Heusler, H.; Amberg, A.; Kurz, M.; Zepnik, H.; Mantle, P.; Völkel, W.; Hard, G.C.; Dekant, W. Biotransformation and nephrotoxicity of ochratoxin B in rats. Toxicol. Appl. Pharmacol. 2005, 206, 43–53. [Google Scholar] [CrossRef] [PubMed]
  59. World Health Organization (WHO). Selected Mycotoxins: Ochratoxins, Trichothecenes, Ergot; WHO-IPCS: Geneva, Switzerland, 1990. [Google Scholar]
  60. Tabata, S.; Iida, K.; Kimura, K.; Iwasaki, Y.; Nakazato, M.; Kamata, K.; Hirokado, M. Investigation of ochratoxin A, B and citrinin contamination in various commercial foods. Shokuhin Eiseigaku Zasshi 2008, 49, 111–115. [Google Scholar] [CrossRef] [PubMed]
  61. Scheuer, R.; Gareis, M. Occurrence of ochratoxin A and B in spices. Mycotoxin Res. 2002, 18, 62–66. [Google Scholar] [CrossRef] [PubMed]
  62. Di Stefano, V.; Avellone, G.; Pitonzo, R.; Capocchiano, V.G.; Mazza, A.; Cicero, N.; Dugo, G. Natural co-occurrence of ochratoxin A, ochratoxin B and aflatoxins in Sicilian red wines. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2015, 32, 1343–1351. [Google Scholar] [CrossRef] [PubMed]
  63. 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]
  64. Heyndrickx, E.; Sioen, I.; Huybrechts, B.; Callebaut, A.; de Henauw, S.; de Saeger, S. Human biomonitoring of multiple mycotoxins in the Belgian population: Results of the BIOMYCO study. Environ. Int. 2015, 84, 82–89. [Google Scholar] [CrossRef] [PubMed]
  65. Dixit, R.; Riviere, J.; Krishnan, K.; Andersen, M.E. Toxicokinetics and physiologically based toxicokinetics in toxicology and risk assessment. J. Toxicol. Environ. Health B Crit. Rev. 2003, 6, 1–40. [Google Scholar] [CrossRef] [PubMed]
  66. Fuchs, R.; Hult, K. Ochratoxin A in blood and its pharmacokinetic properties. Food Chem. Toxicol. 1992, 30, 201–204. [Google Scholar] [CrossRef]
  67. Galtier, P. Pharmacokinetics of ochratoxin A in animals. IARC Sci. Publ. 1991, 115, 187–200. [Google Scholar] [PubMed]
  68. Coronel, M.; Sanchis, V.; Ramos, A.; Marin, S. Ochratoxin A: Presence in human plasma and intake estimation. Food Sci. Technol. Int. 2010, 16, 5–18. [Google Scholar] [CrossRef] [PubMed]
  69. Vettorazzi, A.; González-Peñas, E.; de Cerain, A. Ochratoxin A kinetics: A review of analytical methods and studies in rat model. Food Chem. Toxicol. 2014, 72, 273–288. [Google Scholar] [CrossRef] [PubMed]
  70. 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]
  71. Dietrich, D.R.; Heussner, A.H.; O’Brien, E. Ochratoxin A: Comparative pharmacokinetics and toxicological implications (experimental and domestic animals and humans). Food Addit. Contam. 2005, 22, 45–52. [Google Scholar] [CrossRef] [PubMed]
  72. Schlatter, C.; Studer-Rohr, J.; Rásonyi, T. Carcinogenicity and kinetic aspects of ochratoxin A. Food Addit. Contam. 1996, 13 (Suppl.), 43–44. [Google Scholar] [PubMed]
  73. Marquardt, R.R.; Frohlich, A.A. A review of recent advances in understanding ochratoxicosis. J. Anim. Sci. 1992, 70, 3968–3988. [Google Scholar] [PubMed]
  74. Breitholtz Emanuelsson, A.; Fuchs, R.; Hult, K.; Appelgren, L. Distribution of 14C-ochratoxin A and 14C-ochratoxin B in rats: A comparison based on whole-body autoradiography. IARC Sci. Publ. 1991, 115, 201–203. [Google Scholar] [PubMed]
  75. Breitholtz-Emanuelsson, A.; Fuchs, R.; Hult, K.; Appelgren, L. Synthesis of 14C-ochratoxin A and 14C-ochratoxin B and a comparative study of their distribution in rats using whole body autoradiography. Pharmacol. Toxicol. 1992, 70, 255–261. [Google Scholar] [CrossRef] [PubMed]
  76. 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]
  77. Galtier, P.; Charpenteau, J.-L.; Alvinerie, M.; Labouche, C. The pharmacokinetic profile of ochratoxin A in the rat after oral and intravenous administration. Drug Metab. Dispos. 1979, 7, 429–434. [Google Scholar] [PubMed]
  78. Abrunhosa, L.; Santos, L.; Venâncio, A. Degradation of ochratoxin A by proteases and by a crude enzyme of Aspergillus niger. Food Biotechnol. 2006, 20, 231–242. [Google Scholar] [CrossRef] [Green Version]
  79. Støren, O.; Holm, H.; Størmer, F. Metabolism of ochratoxin A by rats. Appl. Environ. Microbiol. 1982, 44, 785–789. [Google Scholar] [PubMed]
  80. World Health Organization (WHO). Safety Evaluation of Certain Mycotoxins in Food; WHO-IPCS: Geneva, Switzerland, 2001. [Google Scholar]
  81. Pinelli, E.; el Adlouni, C.; Pipy, B.; Quartulli, F.; Pfohl-Leszkowicz, A. Roles of cyclooxygenase and lipoxygenases in ochratoxin A genotoxicity in human epithelial lung cells. Environ. Toxicol. Pharmacol. 1999, 7, 95–107. [Google Scholar] [CrossRef]
  82. Størmer, F.; Hansen, C.; Pedersen, J.; Hvistendahl, G.; Aasen, A. Formation of (4R)- and (4S)-4-hydroxyochratoxin A from ochratoxin A by liver microsomes from various species. Appl. Environ. Microbiol. 1981, 42, 1051–1056. [Google Scholar] [PubMed]
  83. Stein, A.; Phillips, T.; Kubena, L.; Harvey, R. Renal tubular secretion and reabsorption as factors inochratoxicosis: Effects of probenecid on nephrotoxicity. J. Toxicol. Environ. Health 1985, 16, 593–605. [Google Scholar] [CrossRef] [PubMed]
  84. Størmer, F.; Støren, O.; Hansen, C.; Pedersen, J.; Aasen, A. 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]
  85. Creppy, E.E.; Størmer, F.C.; Röschenthaler, R.; Dirheimer, G. Effects of two metabolites of ochratoxin A, (4R)-4-hydroxyochratoxin A and ochratoxin alpha, on immune response in mice. Infect. Immun. 1983, 39, 1015–1018. [Google Scholar] [PubMed]
  86. Xiao, H.; Madhyastha, S.; Marquardt, R.R.; Li, S.; Vodela, J.K.; Frohlich, A.A.; Kemppainen, B.W. Toxicity of ochratoxin A, its opened lactone form and several of its analogs: Structure-activity relationships. Toxicol. Appl. Pharmacol. 1996, 137, 182–192. [Google Scholar] [CrossRef] [PubMed]
  87. Zepnik, H.; Paehler, 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]
  88. El Adlouni, C.; Pinelli, E.; Azémar, B.; Zaoui, D.; Beaune, P.; Pfohl-Leszkowicz, A. Phenobarbital increases 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]
  89. 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]
  90. Kühn, I.; Valenta, H.; Rohr, K. Determination of ochratoxin A in bile of swine by high-performance liquid chromatography. J. Chromatogr. B Biomed. Appl. 1995, 668, 333–337. [Google Scholar] [CrossRef]
  91. Gross-Steinmeyer, K.; Weymann, J.; Hege, H.; Metzler, M. Metabolism and lack of DNA reactivity of the mycotoxin ochratoxin a in cultured rat and human primary hepatocytes. J. Agric. Food Chem. 2002, 50, 938–945. [Google Scholar] [CrossRef] [PubMed]
  92. Zepnik, H.; Völkel, W.; Dekant, W. Toxicokinetics of the mycotoxin ochratoxin A in F344 rats after oral administration. Toxicol. Appl. Pharmacol. 2003, 192, 36–44. [Google Scholar] [CrossRef]
  93. 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 cytotoxic thiol derivative as reactive metabolites. Mutat. Res. 1994, 307, 141–147. [Google Scholar] [CrossRef]
  94. 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]
  95. Fink-Gremmels, J.; Jahn, A.; Blom, M.J. Toxicity and metabolism of ochratoxin A. Nat. Toxins 1995, 3, 214–220. [Google Scholar] [CrossRef] [PubMed]
  96. Li, S.; Marquardt, R.R.; Frohlich, A.A. Identification of ochratoxins and some of their metabolites in bile and urine of rats. Food Chem. Toxicol. 2000, 38, 141–152. [Google Scholar] [CrossRef]
  97. Turner, N.W.; Subrahmanyam, S.; Piletsky, S.A. Analytical methods for determination of mycotoxins: A review. Anal. Chim. Acta 2009, 632, 168–180. [Google Scholar] [CrossRef] [PubMed]
  98. Scott, P.M. Methods of analysis for ochratoxin A. Adv. Exp. Med. Biol. 2002, 504, 117–134. [Google Scholar]
  99. Visconti, A.; de Girolamo, A. Fitness for purpose—Ochratoxin A analytical developments. Food Addit. Contam. 2005, 22, 37–44. [Google Scholar] [CrossRef] [PubMed]
  100. Meulenberg, E.P. Immunochemical methods for ochratoxin A detection: A review. Toxins 2012, 4, 244–266. [Google Scholar] [CrossRef] [PubMed]
  101. Moeller, I.; O’Brien, E.; Heussner, A.; Day, B.; Dietrich, D. Monoclonal Antibodies Against Ochratoxin B: Production and Characterisation. In Proceedings of the Joint Speciality Symposium on Renal Toxicology and Toxicologic Pathology, Lindau, Germany, 27 September–1 October 2004.
  102. Heussner, A.H.; Moeller, I.; Day, B.W.; Dietrich, D.R.; O’Brien, E. Production and characterization of monoclonal antibodies against ochratoxin B. Food Chem. Toxicol. 2007, 45, 827–833. [Google Scholar] [CrossRef] [PubMed]
  103. Heussner, A.H.; Ausländer, S.; Dietrich, D.R. Development and characterization of a monoclonal antibody against ochratoxin B and its application in ELISA. Toxins 2010, 2, 1582–1594. [Google Scholar] [CrossRef] [PubMed]
  104. Glaze, T.; Heussner, A.; Dietrich, D.; Rubio, F. ELISA and sample clean-up methods for determination of ochratoxin B. In Proceedings of the 5th International Symposium on recent advances in food analyses (RAFA), Prague, Czech Recpublic, 1–4 November 2011.
  105. Auslaender, S.T.; Heussner, A.H.; Dietrich, D.R. Monoclonal antibodies against OTB: Potential future applications. Toxicol. Sci. (Toxicologist) 2008, 102, 100. Available online: http://www.toxicology.org/pubs/docs/Tox/2008Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  106. Doi, K.; Uetsuka, K. Mechanisms of mycotoxin-induced dermal toxicity and tumorigenesis through oxidative stress-related pathways. J. Toxicol. Pathol. 2014, 27, 1–10. [Google Scholar] [CrossRef] [PubMed]
  107. 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]
  108. Wu, F.; Groopman, J.D.; Pestka, J.J. Public health impacts of foodborne mycotoxins. Annu. Rev. Food Sci. Technol. 2014, 5, 351–372. [Google Scholar] [CrossRef] [PubMed]
  109. Malir, F.; Ostry, V.; Novotna, E. Toxicity of the mycotoxin ochratoxin A in the light of recent data. Toxin Rev. 2013, 32, 19–33. [Google Scholar] [CrossRef]
  110. Klarić, M.S.; Rašić, D.; Peraica, M. Deleterious effects of mycotoxin combinations involving ochratoxin A. Toxins 2013, 5, 1965–1987. [Google Scholar] [CrossRef] [PubMed]
  111. 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 (Basel) 2013, 5, 1742–1766. [Google Scholar] [CrossRef] [PubMed]
  112. Marin, S.; Ramos, A.; Cano-Sancho, G.; Sanchis, V. Mycotoxins: Occurrence, toxicology, and exposure assessment. Food Chem. Toxicol. 2013, 60, 218–237. [Google Scholar] [CrossRef] [PubMed]
  113. Vettorazzi, A.; van Delft, J.; López de Cerain, A. A review on ochratoxin A transcriptomic studies. Food Chem. Toxicol. 2013, 59, 766–783. [Google Scholar] [CrossRef] [PubMed]
  114. Grenier, B.; Oswald, I. Mycotoxin co-contamination of food and feed: Meta-analysis of publications describing toxicological interactions. World Mycotoxin J. 2011, 4, 285–313. [Google Scholar] [CrossRef]
  115. 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]
  116. Doi, K.; Uetsuka, K. Mechanisms of mycotoxin-induced neurotoxicity through oxidative stress-associated pathways. Int. J. Mol. Sci. 2011, 12, 5213–5237. [Google Scholar] [CrossRef] [PubMed]
  117. Paterson, R.R.; Lima, N. Toxicology of mycotoxins. EXS 2010, 100, 31–63. [Google Scholar] [PubMed]
  118. 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]
  119. Marin-Kuan, M.; Cavin, C.; Delatour, T.; Schilter, B. Ochratoxin A carcinogenicity involves a complex network of epigenetic mechanisms. Toxicon 2008, 52, 195–202. [Google Scholar] [CrossRef] [PubMed]
  120. 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]
  121. Clark, H.A.; Snedeker, S.M. Ochratoxin a: Its cancer risk and potential for exposure. J. Toxicol. Environ. Health B Crit. Rev. 2006, 9, 265–296. [Google Scholar] [CrossRef] [PubMed]
  122. Al-Anati, L.; Petzinger, E. Immunotoxic activity of ochratoxin A. J. Vet. Pharmacol. Ther. 2006, 29, 79–90. [Google Scholar] [CrossRef] [PubMed]
  123. Schilter, B.; Marin-Kuan, M.; Delatour, T.; Nestler, S.; Mantle, P.; Cavin, C. Ochratoxin A: Potential epigenetic mechanisms of toxicity and carcinogenicity. Food Addit. Contam. 2005, 22, 88–93. [Google Scholar] [CrossRef] [PubMed]
  124. Mally, A.; Dekant, W. DNA adduct formation by ochratoxin A: Review of the available evidence. Food Addit. Contam. 2005, 22, 65–74. [Google Scholar] [CrossRef] [PubMed]
  125. Turesky, R. Perspective: Ochratoxin A is not a genotoxic carcinogen. Chem. Res. Toxicol. 2005, 18, 1082–1090. [Google Scholar] [CrossRef] [PubMed]
  126. 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]
  127. Gekle, M.; Sauvant, C.; Schwerdt, G. Ochratoxin A at nanomolar concentrations: A signal modulator in renal cells. Mol. Nutr. Food Res. 2005, 49, 118–130. [Google Scholar] [CrossRef] [PubMed]
  128. Walker, R. Risk assessment of ochratoxin: Current views of the European Scientific Committee on Food, the JECFA and the Codex Committee on Food Additives and Contaminants. Adv. Exp. Med. Biol. 2002, 504, 249–255. [Google Scholar] [PubMed]
  129. Gekle, M.; Sauvant, C.; Schwerdt, G.; Silbernagl, S. Tubulotoxic mechanisms of ochratoxin A. Kidney Blood Press. Res. 1998, 21, 277–279. [Google Scholar] [CrossRef] [PubMed]
  130. Fillastre, J.P. Experimental and human nephrotoxicity induced by ochratoxins. Bull. Acad. Natl. Med. 1997, 181, 1447–1460. [Google Scholar] [PubMed]
  131. Godin, M.; Fillastre, J.P.; Simon, P.; Francois, A.; Roy, F.L.; Morin, J.P. Is ochratoxin A nephrotoxic in human beings? Adv. Nephrol. Necker Hosp. 1997, 26, 181–206. [Google Scholar] [PubMed]
  132. Plestina, R. Nephrotoxicity of ochratoxin A. Food Addit. Contam. 1996, 13, 49–50. [Google Scholar] [PubMed]
  133. Dirheimer, G. Mechanistic approaches to ochratoxin toxicity. Food Addit. Contam. 1996, 13, 45–48. [Google Scholar] [PubMed]
  134. Gekle, M.; Silbernagl, S. Renal toxicodynamics of ochratoxin A: A pathophysiological approach. Kidney Blood Press. Res. 1996, 19, 225–235. [Google Scholar] [CrossRef] [PubMed]
  135. Sharma, R.P. Immunotoxicity of mycotoxins. J. Dairy Sci. 1993, 76, 892–897. [Google Scholar] [CrossRef]
  136. Huff, J.E. Carcinogenicity of ochratoxin A in experimental animals. IARC Sci. Publ. 1991, 115, 229–244. [Google Scholar] [PubMed]
  137. Dirheimer, G.; Creppy, E.E. Mechanism of action of ochratoxin A. IARC Sci. Publ. 1991, 115, 171–186. [Google Scholar] [PubMed]
  138. Munro, C.; Scott, P.M.; Moodie, C.A.; Willes, R.F. Ochratoxin A—Occurrence and toxicity. J. Am. Vet. Med. Assoc. 1973, 163, 1269–1273. [Google Scholar] [PubMed]
  139. Rumora, L.; Grubisić, T.Z. A journey through mitogen-activated protein kinase and ochratoxin A interactions. Arh. Hig. Rada Toksikol. 2009, 60, 449–456. [Google Scholar] [CrossRef] [PubMed]
  140. Marin-Kuan, M.; Ehrlich, V.; Delatour, T.; Cavin, C.; Schilter, B. Evidence for a role of oxidative stress in the carcinogenicity of ochratoxin A. J. Toxicol. 2011, 2011. [Google Scholar] [CrossRef] [PubMed]
  141. IARC. Some Naturally Occurring Substances: Food Items and Constituents, Heterocyclic Aromatic Amines and Mycotoxins. In IARC Monographs on the Evaluation of Carcinogenic Risks to Humans; IARC: Lyon, France, 1993. [Google Scholar]
  142. EFSA. Statement on recent scientific information on the toxicity of ochratoxin A. EFSA J. 2010, 8, 1–7. [Google Scholar]
  143. Joint FAO/WHO Expert Committee on Food Additives (JECFA). Evaluation of Certain Food Additives and Contaminants; Technical Report Series No. 806 for WHO: Geneva, Switzerland, 1991. [Google Scholar]
  144. Joint FAO/WHO Expert Committee on Food Additives (JECFA). Evaluation of Certain Food Additives and Contaminants; Technical Report Series No. 855 for WHO: Geneva, Switzerland, 1995. [Google Scholar]
  145. Joint FAO/WHO Expert Committee on Food Additives (JECFA). Evaluation of Certain Mycotoxins in Food; Technical Report for Series No. 896 for WHO: Geneva, Switzerland, 2002. [Google Scholar]
  146. Joint FAO/WHO Expert Committee on Food Additives (JECFA). Safety Evaluation of Certain Food Additives and Contaminants: Ochratoxin A (addendum); WHO: Geneva, Switzerland, 2007. [Google Scholar]
  147. European Commission. Maximum Levels for Certain Contaminants in Foodstuffs. Available online: http://eur-lex.europa.eu/legal-content/EN/TXT/?qid=1445355561269&uri=CELEX:32006R1881 (accessed on 20 October 2015).
  148. NTP (National Toxicology Program). Toxicology and carcinogenesis studies of ochratoxin A (CAS No. 303-47-9) in F344/N rats (gavage studies); U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health: Bethesda, MD, USA, 1989.
  149. NTP (National Toxicology Program). Report on Carcinogens (Ochratoxin A); U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health: Bethesda, MD, USA, 2011.
  150. NTP (National Toxicology Program). Report on Carcinogens (Ochratoxin A); U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health: Bethesda, MD, USA, 2014. Available online: https://ntp.niehs.nih.gov/ntp/roc/content/profiles/ochratoxina.pdf (accessed on 20 October 2015).
  151. Kuiper-Goodman, T.; Scott, P.M. Risk assessment of the mycotoxin ochratoxin A. Biomed. Environ. Sci. 1989, 2, 179–248. [Google Scholar] [PubMed]
  152. Kuiper-Goodman, T.; Hilts, C.; Billiard, S.M.; Kiparissis, Y.; Richard, I.D.; Hayward, S. Health risk assessment of ochratoxin A for all age-sex strata in a market economy. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2010, 27, 212–240. [Google Scholar] [CrossRef] [PubMed]
  153. Health Canada. Information Document on Health Canada’s Proposed Maximum Limits (Standards) for the Presence of the Mycotoxin Ochratoxin A in Foods. Bureau of Chemical Safety, Food Directorate, Health Products and Food Branch: Ottawa, ON, Cannada, 2009. Available online: http://www.hc-sc.gc.ca/fn-an/alt_formats/hpfb-dgpsa/pdf/consultation/myco_consult_ochra-eng.pdf (accessed on 20 October 2015).
  154. Egypt. Organization for Standardazation and Quality: Egyptian Standard 1875-2/2010 Maximum Limits for Mycotoxins in Foods-Ochratoxins; Egyptian Organization for Standardization and Quality Control: Cairo, Egypt, 2010. [Google Scholar]
  155. China’s Maximum Levels for Mycotoxins in Foods. Available online: http://gain.fas.usda.gov/Recent%20GAIN%20Publications/Maximum%20Levels%20of%20Mycotoxins%20in%20Foods_Beijing_China%20-%20Peoples%20Republic%20of_12-29-2014.pdf (accessed on 20 October 2015).
  156. Müller, G.; Burkert, B.; Möller, U.; Diller, R.; Rohrmann, B.; Rosner, H.; Köhler, H. Ochratoxin A and some of its derivatives modulate radical formation of porcine blood monocytes and granulocytes. Toxicology 2004, 199, 251–259. [Google Scholar] [CrossRef] [PubMed]
  157. Peckham, J.; Doupnik, B.J.; Jones, O.J. Acute toxicity of ochratoxins A and B in chicks. Appl. Microbiol. 1971, 21, 492–494. [Google Scholar] [PubMed]
  158. Doster, R.C.; Sinnhuber, R.O.; Pawlowski, N.E. Acute intraperitoneal toxicity of ochratoxin A and B derivatives in rainbow trout (Salmo gairdneri). Food Cosmet. Toxicol. 1974, 12, 499–505. [Google Scholar] [CrossRef]
  159. Luhe, A.; Hildebrand, H.; Bach, U.; Dingermann, T.; Ahr, H. A new approach to studying ochratoxin A (OTA)-induced nephrotoxicity: Expression profiling in vivo and in vitro employing cDNA microarrays. Toxicol. Sci. 2003, 73, 315–328. [Google Scholar] [CrossRef] [PubMed]
  160. Chattopadhyay, P.; Upadhyay, A.; Agnihotri, A.; Karmakar, S.; Ghoyary, D.; Veer, V. Comparative hematoxicity of fusirium mycotoxin in experimental sprague-dawley rats. Toxicol. Int. 2013, 20, 25–29. [Google Scholar] [CrossRef] [PubMed]
  161. Bondy, G.S.; Armstrong, C.L. Cytotoxicity of nephrotoxic fungal toxins to kidney-derived LLC-PK1 and OK cell lines. Cell Biol. Toxicol. 1998, 14, 323–332. [Google Scholar] [CrossRef] [PubMed]
  162. Bruinink, A.; Rasonyi, 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] [PubMed]
  163. Knasmüller, 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: Implications for risk assessment. Nutr. Cancer 2004, 50, 190–197. [Google Scholar] [CrossRef] [PubMed]
  164. Heussner, A.H.; Schwöbel, F.; Dietrich, D.R. Cytotoxicity of ochratoxin A and B in vitro: Comparison of male and female rat primary renal cortex and distal cells and LLC-PK1 cells. Toxicol. Sci. 1998, 42, 288. Available online: http://www.toxicology.org/pubs/docs/Tox/1998Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  165. Heussner, A.H.; Stack, M.E.; Hochberg, K.; Dietrich, D.R. Comparison of cytotoxic effects of ochratoxin A and B on human, rat and porcine renal cells. Toxicol. Sci. 2000, 54, 170. Available online: http://www.toxicology.org/pubs/docs/Tox/2000Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  166. Dietrich, D.R.; O’Brien, E.; Stack, M.E.; Heussner, A.H. Species- and sex-specific renal cytotoxicity of ochratoxin A and B in vitro. Exp. Toxicol. Pathol. 2001, 53, 215–225. [Google Scholar] [CrossRef] [PubMed]
  167. Heussner, A.H.; O’Brien, E.; Dietrich, D.R. Effects of repeated ochratoxin exposure on renal cells in vitro. Toxicol. Vitr. 2007, 21, 72–80. [Google Scholar] [CrossRef] [PubMed]
  168. 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]
  169. O’Brien, E.; Heussner, A.H.; Stack, M.E.; Thiel, R.; Dietrich, D.R. Species comparison of renal [3H]ochratoxin B protein binding and uptake in vitro. Toxicol. Sci. 2002, 66, 82. Available online: http://www.toxicology.org/pubs/docs/Tox/2002Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  170. Stemmer, K.; Heussner, A.; Stack, M.; Thiel, R.; Dietrich, D. Comparison of ochratoxin binding to human, porcine and rat renal proteins in vitro. Toxicol. Sci. 2002, 66, 82. Available online: http://www.toxicology.org/pubs/docs/Tox/2002Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  171. 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]
  172. Heussner, A.H.; O’Brien, E.; Dietrich, D.R. Species- and sex-specific variations in binding of ochratoxin A by renal proteins in vitro. Exp. Toxicol. Pathol. 2002, 54, 151–159. [Google Scholar] [CrossRef] [PubMed]
  173. Heussner, A.H.; O’Brien, E.; Stack, M.E.; Thiel, R.; Dietrich, D.R. Cytotoxicity of OTA and OTB in human and porcine renal cells after repeated exposure. Toxicol. Sci. 2002, 66, 82. Available online: http://www.toxicology.org/pubs/docs/Tox/2002Tox.pdf (accessed on 20 October 2015). [Google Scholar]
  174. Heussner, A.H.; O’Brien, E.; Stack, M.E.; Dietrich, D.R. Alterations in human and porcine renal cells after repeated exposure to OTA and OTB. Toxicol. Sci. (Toxicologist) 2003, 72, 346–347. [Google Scholar]
  175. Grauenhorst, A.; Heussner, A.; Dietrich, D. Differential Expression Analysis of Kidney Cells Repeatedly Exposed to Ochratoxin. In Proceedings of the Joint Speciality Symposium on Renal Toxicology and Toxicologic Pathology, Lindau, Germany, 27 September–1 October 2004.
  176. Richetti, A.; Cavallaro, A.; Ainis, T.; Fimiani, V. Effect of mycotoxins on some activities of isolated human neutrophils. Immunopharmacol. Immunotoxicol. 2005, 27, 433–446. [Google Scholar] [CrossRef] [PubMed]
  177. Richetti, A.; Cavallaro, A.; Ainis, T.; Fimiani, V. Effect of some mycotoxins on superoxide anion production of isolated human neutrophils and in whole blood. Immunopharmacol. Immunotoxicol. 2003, 25, 441–449. [Google Scholar] [CrossRef] [PubMed]
  178. Lea, T.; Steien, K.; Størmer, F.C. Mechanism of ochratoxin A-induced immunosuppression. Mycopathologia 1989, 107, 153–159. [Google Scholar] [CrossRef] [PubMed]
  179. Malaveille, C.; Brun, G.; Bartsch, H. Genotoxicity of ochratoxin A and structurally related compounds in Escherichia coli strains: Studies on their mode of action. IARC Sci. Publ. 1991, 115, 261–266. [Google Scholar] [PubMed]
  180. O’Brien, E.; Prietz, A.; Dietrich, D.R. Investigation of the teratogenic potential of ochratoxin A and B using the FETAX system. Birth Defects Res. B Dev. Reprod. Toxicol. 2005, 74, 417–423. [Google Scholar] [CrossRef] [PubMed]
  181. Wiger, R.; Størmer, F.C. Effects of ochratoxins A and B on prechondrogenic mesenchymal cells from chick embryo limb buds. Toxicol. Lett. 1990, 54, 129–134. [Google Scholar] [CrossRef]
  182. Roth, A.; Creppy, E.E.; Kane, A.; Bacha, H.; Steyn, P.S.; Röschenthaler, R.; Dirheimer, G. Influence of ochratoxin B on the ochratoxin A inhibition of phenylalanyl-tRNA formation in vitro and protein synthesis in hepatoma tissue culture cells. Toxicol. Lett. 1989, 45, 307–313. [Google Scholar] [CrossRef]
  183. Chu, F.S.; Chang, C.C. Sensitivity of chicks to ochratoxins. J. Assoc. Off. Anal. Chem. 1971, 54, 1032–1034. [Google Scholar] [PubMed]
  184. Müller, G.; Burkert, B.; Rosner, H.; Köhler, H. Effects of the mycotoxin ochratoxin A and some of its metabolites on human kidney cell lines. Toxicol. Vitr. 2003, 17, 441–448. [Google Scholar] [CrossRef]
  185. Köhler, H.; Heller, M.; Erler, W.; Müller, G.; Rosner, H.; Gräfe, U. Effect of ochratoxin A and ochratoxin C on the monocyte and lymphocyte function. Mycotoxin Res. 2002, 18, 169–172. [Google Scholar] [CrossRef] [PubMed]
  186. Müller, G.; Rosner, H.; Rohrmann, B.; Erler, W.; Geschwend, G.; Gräfe, U.; Burkert, B.; Möller, U.; Diller, R.; Sachse, K.; et al. Effects of the mycotoxin ochratoxin A and some of its metabolites on the human cell line THP-1. Toxicology 2003, 184, 69–82. [Google Scholar] [CrossRef]
  187. Köhler, H.; Heller, M.; Erler, W.; Müller, G. Effects of a long-term exposure with OTA or OTC on functions of a human monocytic cell line. Mycotoxin Res. 2003, 19, 108–112. [Google Scholar] [CrossRef] [PubMed]
  188. Steyn, P.S.; Holzapfel, C.W. The isolation of methyl and ethyl esters of ochratoxin A and B, metabolites of Aspergillus ochraceus Wilh. J. S. Afr. Chem. Inst. 1967, 20, 186–189. [Google Scholar]
  189. Creppy, E.E.; Størmer, F.C.; Kern, D.; Röschenthaler, R.; Dirheimer, G. Effects of ochratoxin A metabolites on yeast phenylalanyl-tRNA synthetase and on the growth and in vivo protein synthesis of hepatoma cells. Chem. Biol. Interact. 1983, 47, 239–247. [Google Scholar] [CrossRef]
  190. Bittner, A.; Cramer, B.; Harrer, H.; Humpf, H.U. Structure elucidation and in vitro cytotoxicity of ochratoxin α amide, a new degradation product of ochratoxin A. Mycotoxin Res. 2015, 31, 83–90. [Google Scholar] [CrossRef] [PubMed]
  191. Foellmann, W.; Hillebrand, I.; Creppy, E.; Bolt, H. Sister chromatid exchange frequency in cultured isolated porcine urinary bladder epithelial cells (PUBEC) treated with ochratoxin A and α. Arch. Toxicol. 1995, 69, 280–286. [Google Scholar] [CrossRef]
  192. Hutchison, R.D.; Steyn, P.S. The isolation and structure of 4-hydroxyochratoxin and 7-carboxy-3,4-dihydro-8-hydroxy-3-methylisocoumarin from Penicillium viridicatum. Tetrahedron Lett. 1971, 43, 4033–4036. [Google Scholar] [CrossRef]
  193. 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]
  194. Gautier, J.; Richoz, J.; Welti, D.; Markovic, J.; Gremaud, E.; Guengerich, F.; Turesky, R. Metabolism of ochratoxin A: Absence of formation of genotoxic derivatives by human and rat enzymes. Chem. Res. Toxicol. 2001, 14, 34–45. [Google Scholar] [CrossRef] [PubMed]
  195. Creppy, E.E.; Kern, D.; Steyn, P.S.; Vleggaar, R.; Röschenthaler, R.; Dirheimer, G. Comparative study of the effect of ochratoxin A analogues on yeast aminoacyl-tRNA synthetases and on the growth and protein synthesis of hepatoma cells. Toxicol. Lett. 1983, 19, 217–224. [Google Scholar] [CrossRef]
  196. Richard, J. Some major mycotoxins and their mycotoxicoses—An overview. Int. J. Food Microbiol. 2007, 119, 3–10. [Google Scholar] [CrossRef] [PubMed]
  197. Bragulat, M.R.; Martínez, E.; Castellá, G.; Cabañes, F.J. Ochratoxin A and citrinin producing species of the genus Penicillium from feedstuffs. Int. J. Food Microbiol. 2008, 126, 43–48. [Google Scholar] [CrossRef] [PubMed]
  198. Frisvad, J.C.; Frank, J.M.; Houbraken, J.A.M.P.; Kuijpers, A.F.A.; Samson, R.A. New ochratoxin A producing species of Aspergillus section Circumdati. Stud. Mycol. 2004, 50, 23–43. [Google Scholar]
  199. Soares, C.; Calado, T.; Venâncio, A. Mycotoxin production by Aspergillus niger aggregate strains isolated from harvested maize in three Portuguese regions. Rev. Iberoam. Micol. 2013, 30, 9–13. [Google Scholar] [CrossRef] [PubMed]
  200. Speijers, G.J.; Speijers, M.H. Combined toxic effects of mycotoxins. Toxicol. Lett. 2004, 153, 91–98. [Google Scholar] [CrossRef] [PubMed]
  201. Elsgaard, L.; Børgesen, C.; Olesen, J.; Siebert, S.; Ewert, F.; Peltonen-Sainio, P.; Rötter, R.; Skjelvåg, A. Shifts in comparative advantages for maize, oat and wheat cropping under climate change in Europe. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2012, 29, 1514–1526. [Google Scholar] [CrossRef] [PubMed]
  202. Van der Fels-Klerx, H.; Olesen, J.; Naustvoll, L.; Friocourt, Y.; Mengelers, M.; Christensen, J. Climate change impacts on natural toxins in food production systems, exemplified by deoxynivalenol in wheat and diarrhetic shellfish toxins. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2012, 29, 1647–1659. [Google Scholar] [CrossRef] [PubMed]
  203. Rumora, L.; Domijan, A.M.; Zanic Grubisic, T.; Segvic Klaric, M. Differential activation of MAPKs by individual and combined ochratoxin A and citrinin treatments in porcine kidney PK15 cells. Toxicon 2014, 90, 174–183. [Google Scholar] [CrossRef] [PubMed]
  204. Clarke, R.; Connolly, L.; Frizzell, C.; Elliott, C.T. High content analysis: A sensitive tool to detect and quantify the cytotoxic, synergistic and antagonistic effects of chemical contaminants in foods. Toxicol. Lett. 2015, 233, 278–286. [Google Scholar] [CrossRef] [PubMed]
  205. Föllmann, W.; Behm, C.; Degen, G.H. Toxicity of the mycotoxin citrinin and its metabolite dihydrocitrinone and of mixtures of citrinin and ochratoxin A in vitro. Arch. Toxicol. 2014, 88, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
  206. Li, Y.; Zhang, B.; He, X.; Cheng, W.H.; Xu, W.; Luo, Y.; Liang, R.; Luo, H.; Huang, K. Analysis of individual and combined effects of ochratoxin A and zearalenone on HepG2 and KK-1 cells with mathematical models. Toxins 2014, 6, 1177–1192. [Google Scholar] [CrossRef] [PubMed]
  207. Clarke, R.; Connolly, L.; Frizzell, C.; Elliott, C.T. Cytotoxic assessment of the regulated, co-existing mycotoxins aflatoxin B1, fumonisin B1 and ochratoxin, in single, binary and tertiary mixtures. Toxicon 2014, 90, 70–81. [Google Scholar] [CrossRef] [PubMed]
  208. Anninou, N.; Chatzaki, E.; Papachristou, F.; Pitiakoudis, M.; Simopoulos, C. Mycotoxins’ activity at toxic and sub-toxic concentrations: Differential cytotoxic and genotoxic effects of single and combined administration of sterigmatocystin, ochratoxin A and citrinin on the hepatocellular cancer cell line Hep3B. Int. J. Environ. Res. Public Health 2014, 11, 1855–1872. [Google Scholar] [CrossRef] [PubMed]
  209. Heussner, A.H.; Dietrich, D.R.; O’Brien, E. In vitro investigation of individual and combined cytotoxic effects of ochratoxin A and other selected mycotoxins on renal cells. Toxicol. Vitr. 2006, 20, 332–341. [Google Scholar] [CrossRef] [PubMed]
  210. Bouslimi, A.; Bouaziz, C.; Ayed-Boussema, I.; Hassen, W.; Bacha, H. Individual and combined effects of ochratoxin A and citrinin on viability and DNA fragmentation in cultured Vero cells and on chromosome aberrations in mice bone marrow cells. Toxicology 2008, 251, 1–7. [Google Scholar] [CrossRef] [PubMed]
  211. Bouslimi, A.; Ouannes, Z.; Golli, E.E.; Bouaziz, C.; Hassen, W.; Bacha, H. Cytotoxicity and oxidative damage in kidney cells exposed to the mycotoxins ochratoxin a and citrinin: Individual and combined effects. Toxicol. Mech. Methods 2008, 18, 341–349. [Google Scholar] [CrossRef] [PubMed]
  212. Klarić, M.S.; Zelježić, D.; Rumora, L.; Peraica, M.; Pepeljnjak, S.; Domijan, A.M. A potential role of calcium in apoptosis and aberrant chromatin forms in porcine kidney PK15 cells induced by individual and combined ochratoxin A and citrinin. Arch. Toxicol. 2012, 86, 97–107. [Google Scholar] [CrossRef] [PubMed]
  213. Gayathri, L.; Dhivya, R.; Dhanasekaran, D.; Periasamy, V.S.; Alshatwi, A.A.; Akbarsha, M.A. Hepatotoxic effect of ochratoxin A and citrinin, alone and in combination, and protective effect of vitamin E: In vitro study in HepG2 cell. Food Chem. Toxicol. 2015, 83, 151–163. [Google Scholar] [CrossRef] [PubMed]
  214. Corcuera, L.A.; Vettorazzi, A.; Arbillaga, L.; Perez, N.; Gil, A.G.; Azqueta, A.; Gonzalez-Penas, E.; Garcia-Jalon, J.A.; Lopez de Cerain, A. Genotoxicity of Aflatoxin B1 and Ochratoxin A after simultaneous application of the in vivo micronucleus and comet assay. Food Chem. Toxicol. 2015, 76, 116–124. [Google Scholar] [CrossRef] [PubMed]
  215. Cano-Sancho, G.; Gonzalez-Arias, C.A.; Ramos, A.J.; Sanchis, V.; Fernandez-Cruz, M.L. Cytotoxicity of the mycotoxins deoxynivalenol and ochratoxin A on Caco-2 cell line in presence of resveratrol. Toxicol. Vitr. 2015, 29, 1639–1646. [Google Scholar] [CrossRef] [PubMed]
  216. Segvic Klaric, M.; Jaksic Despot, D.; Kopjar, N.; Rasic, D.; Kocsube, S.; Varga, J.; Peraica, M. Cytotoxic and genotoxic potencies of single and combined spore extracts of airborne OTA-producing and OTA-non-producing Aspergilli in Human lung A549 cells. Ecotoxicol. Environ. Saf. 2015, 120, 206–214. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Heussner, A.H.; Bingle, L.E.H. Comparative Ochratoxin Toxicity: A Review of the Available Data. Toxins 2015, 7, 4253-4282. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins7104253

AMA Style

Heussner AH, Bingle LEH. Comparative Ochratoxin Toxicity: A Review of the Available Data. Toxins. 2015; 7(10):4253-4282. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins7104253

Chicago/Turabian Style

Heussner, Alexandra H., and Lewis E. H. Bingle. 2015. "Comparative Ochratoxin Toxicity: A Review of the Available Data" Toxins 7, no. 10: 4253-4282. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins7104253

Article Metrics

Back to TopTop