C 2014 Wiley Periodicals, Inc.

Birth Defects Research (Part B) 98:493–502 (2013)

Review Article

Ochratoxin A: Developmental and Reproductive Toxicity—An Overview Frantisek Malir,1∗ Vladimir Ostry,2 Annie Pfohl-Leszkowicz,3 and Eva Novotna4 1 Department

of Biology, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic for Health, Nutrition and Food, National Institute of Public Health in Prague, Brno, Czech Republic 3 Department Bioprocess & Microbial Systems, INP/ENSA Toulouse, Laboratory Chemical Engineering, UMR 5503 CNRS/INPT/UPS, University of Toulouse, Auzeville-Tolosane, France 4 Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University Prague, Hradec Kralove, Czech Republic 2 Center

Ochratoxin A (OTA) is nephrotoxic, hepatotoxic, reprotoxic, embryotoxic, teratogenic, neurotoxic, immunotoxic, and carcinogenic for laboratory and farm animals. Male and female reproductive health has deteriorated in many countries during the last few decades. A number of toxins in environment are suspected to affect reproductive system in male and female. OTA is one of them. OTA has been found to be teratogenic in several animal models including rat, mouse, hamster, quail, and chick, with reduced birth weight and craniofacial abnormalities being the most common signs. The presence of OTA also results in congenital defects in the fetus. Neither the potential of OTA to cause malformations in human nor its teratogenic mode of action is known. Exposure to OTA leads to increased embryo lethality manifested as resorptions or dead fetuses. The mechanism of OTA transfer across human placenta (e.g., which transporters are involved in the transfer mechanism) is not fully understood. Some of the toxic effects of OTA are potentiated by other mycotoxins or other contaminants. Therefore, OTA exposure of pregnant women should be minimized. OTA has been shown to be an endocrine disruptor and a reproductive toxicant, with abilities of altering sperm quality. Other studies have shown that OTA is a testicular toxin in animals. Thus, OTA is a biologically plausible cause of testicular cancer in man. Birth Defects Res (Part B) 98:493–502, 2013.

 C 2014 Wiley Periodicals, Inc.

Key words: ochratoxin A; reprotoxicity; developmental toxicity; teratogenicity; pregnant woman; testicular cancer

INTRODUCTION Ochratoxins are one of the important groups of mycotoxins. In this group, many forms of ochratoxins and their metabolites have been described including phenylalanine-based ochratoxin A (OTA), B, and C (Xiao et al., 1995, 1996; Azpilicueta et al., 2008; Wu et al., 2011; Tozlovanu et al., 2012). Some metabolites such as the lactone-opened OTA (OP-OTA) and OTHQ (quinone form) have also been found in blood and urine, and have been described to be as toxic as OTA (OPOTA) or more toxic OTHQ (Xiao et al., 1996; Li et al., 2000; Faucet-Marquis et al., 2006; Tozlovanu et al., 2006; Pfohl-Leszkowicz, 2009). Among ochratoxins, OTA (see Fig. 1) is the prevalent one. OTA is suggested to contribute to human disease and is a cause of farm animal disease and economic losses (Pfohl-Leszkowicz et al., 2002, 2007; Malir et al., 2012, 2013a) to farmers and food processors. Worldwide, OTA is one of the most common mycotoxins found in foodstuffs (SCOOP, 2002; Woo et al., 2012) of both plant and animal origins. It is produced by fungi of genera Aspergillus and Penicillium (Malir et al., 2013a;

Ostry et al., 2013). The consumption of foodstuffs contaminated by OTA represents a major source of exposure for the general population. In agriculture or in food industry or after water damage, exposure to OTA could be due to dermal contact or inhalation (Degen et al., 2007; PfohlLeszkowicz, 2013). OTA has been shown to have many toxic effects in animals and humans (Malir et al., 2013a). The most important are nephrotoxicity, immunotoxicity, and carcinogenicity. IARC (1993) classified OTA as a possible human carcinogen (group 2B). The susceptibility to cancer is species- and sex-dependent (Castegnaro et al., 1998; Pfohl-Leszkowicz et al., 1998; Pfohl-Leszkowicz and Manderville, 2007; Malir et al., 2013a; Hsuuw et al., 2013). Grant sponsor: Czech Ministry of Health (IGA MZ CR); Grant number: NT 12051-3/2011. ∗ Correspondence to: Frantisek Malir, Department of Biology, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic. E-mail: [email protected] Received 2 October 2013; Accepted 10 December 2013 Published online in Wiley Online Library (wileyonlinelibrary.com/journal/ bdrb) DOI: 10.1002/bdrb.21091

494

MALIR ET AL.

Fig. 1. Chemical structure of OTA.

Even though the mechanism of action remains a controversial topic under debate (Malir et al., 2013a), frequent exposure to OTA can cause health problem. OTA ingestion during pregnancy can affect the intrauterine life (Wangikar et al., 2005). Chronic exposure to low OTA doses could be much more damaging than acute exposure to a high dose (Pfohl-Leszkowicz et al., 2007; PfohlLeszkowicz, 2009). While the reproductive toxicity of OTA is well documented in animals, there are currently no data linking OTA exposure to human reproductive toxicity.

OTA BIOMONITORING No data on teratogenic dose and teratogenic OTA effects in humans are available. The fetal and neonatal exposure to OTA is likely determined by the analysis of cord blood samples from pregnant women, human blood, colostrum, milk, and urine (Micco et al., 1991; Kovacs ˜ et al., 1995; Miraglia et al., 1998; Skaug et al., 1998; Munoz et al., 2010; Klapec et al., 2012). It is clear that OTA fetal exposure might pose a potential risk in prenatal and postnatal life for the human infants (Micco et al., 1991; Jonsyn et al., 1995; Kovacs et al., 1995; Skaug et al., 1998). Since OTA is excreted via breast milk, breast-fed children are also exposed to OTA (Gareis et al., 1988). Although OTA amounts in milk are much lower (10 times less) than those of OTA in blood (Breitholtz-Emanuelsson et al., 1993), OTA contamination of human breast milk presents a possible health hazard (Kuiper-Goodman et al., 2010). The determination of OTA in blood remains the basic method for monitoring human exposure to OTA (Malir et al., 2012). Scott (2005) has described OTA in blood serum as a uniquely useful biomarker of OTA exposure due to its high-affinity binding to serum albumin or other small proteins. Serum OTA contents in pregnant women ranged from 0.06 to 3.41 ␮g/l (Zimmerli and Dick, 1995; Rosner et al., 2000; Postupolski et al., 2006, Malir et al., 2013b). Recently, a study on the assessment of OTA intake by pregnant women in the first trimester of pregnancy was conducted. Serum OTA amounts ranged from 0.1 to 0.35 ␮g/l with a mean value of 0.15 ␮g/l. Presence of OTA in serum of pregnant women correlates with OTA dietary exposure assessment (Malir et al., 2013b). In utero, exposure of human fetuses to OTA is proven by the presence of OTA in fetal serum and cord blood samples from 0.13 to 5.42 ␮g/l (Zimmerli and Dick, 1995; Postupolski et al., 2006; Biasucci et al., 2011). This finding

raises concerns about potential health hazards to human fetuses. OTA passing through placenta via active transport (Zimmerli and Dick, 1995; Postupolski et al., 2006) is found in human fetal samples (Jonsyn et al., 1995; Zimmerli and Dick, 1995; Postupolski et al., 2006; Biasucci et al., 2011). The higher amount of OTA in fetal serum than in maternal serum (Zimmerli and Dick, 1995; Postupolski et al., 2006) is due to gradual accumulation over chronic exposure during early gestation (Zimmerli and Dick, 1995). OTA determination in urine is studied to clarify the relationship at the individual level between OTA intake and urinary biomarker (Duarte et al., 2011). OTA in urine is a promising alternative to the measurement of OTA in blood, even though the OTA concentration in urine is much lower than in blood. Despite these difficulties, OTA in urine is thought by some authors to be a better indicator of exposure to OTA than OTA in plasma (Gilbert et al., 2001; Castegnaro et al., 2006; Pfohl-Leszkowicz et al., 2006). Moreover, analysis of OTA metabolites in urine will give additional information concerning toxicokinetic and genetic susceptibility (Pfohl-Leszkowicz, 2009). The first study to determine the exposure of pregnant women to OTA was realized through examination of OTA and ochratoxin alpha in the urine of pregnant women (Klapec et al., 2012).

OTA AND PLACENTA Human placenta plays an important role in the development of the fetus, being the major interface between mother and fetus. Placenta provides a gateway to oxygen and nutrients from the mother to the fetus. It produces hormones to support the pregnancy and serves as an excretion pathway for various metabolites and carbon dioxide. However, it also plays a role in the exposure of the fetus to potential xenobiotics. Transport processes across the placenta (passive diffusion, active transport, facilitated diffusion, filtration, and pinocytosis) and metabolism determine the exposure of the fetus to xenobiotics including OTA (Myllynen and V¨ah¨akangas, 2013). Very little is known about the mechanism of OTA transfer across human placenta. Presence of transporters, immature metabolism, and low elimination capacity of the fetus contribute to the bioaccumulation of OTA in the fetal circulation (Woo et al., 2012). The transfer of OTA is limited by its high-affinity binding to plasma proteins (Woo et al., 2012). It has been estimated that approximately 99% of the circulating OTA is bound to plasma proteins (mainly albumin; Chu, 1971, 1974; Dai et al., 2004) and only a small amount of OTA occurs freely. In human perfused placenta, OTA transfer is not concentrationdependent (Woo et al., 2012). This can be explained by the fact that only unbound molecule is capable of passing through placental barrier (Loebstein and Koren, 2002). OTA administered to pregnant rats is detected in embryos (Mor´e and Galtier, 1978a; Appelgreen and Arora, 1983a; Ballinger et al., 1986; Fukui et al., 1987; Hallen et al., 1998; Minervi et al., 2013). At the beginning of the pregnancy, OTA quickly passes through mouse placenta (Appelgreen and Arora, 1983b). After crossing the maternal placental barrier, OTA reaches the fetus and Birth Defects Research (Part B) 98:493–502, 2013

REPROTOXICITY OF OCHRATOXIN

495

interferes with the development of the organs (Appelgreen and Arora, 1983b). A study on OTA in human perfused placenta shows that OTA is able to cross the placenta, more particularly, at early gestation rather than at late gestation (Woo et al., 2012) Direct genotoxic effect of OTA, reflected by DNA adducts, in the progeny (the fetus and the offspring) through transplacental contamination was demonstrated in a mice study. High amounts of DNA adducts were detected in the kidneys of male progeny, and in liver of female progeny and their mothers exposed to a single-OTA dose during late pregnancy (Petkova-Bocharova et al., 1998b). These data correlate well with the carcinogenicity in mice, for which the target organ for males is the kidney, while for females it is the liver (Petkova-Bocharova et al., 1998b). Similar data were obtained with hamster (Petkova-Bocharova et al., 1998a). Some owners and breeder of cats and dogs observed a decrease of fertility and stillbirth when the animals are fed with petfood. Cereal grain and nuts are often used as ingredients in industrial petfood. All the pet food, leading to reproductive problems, was contaminated by several mycotoxins. The petfood without cereal contains much less mycotoxins and enabling normal birth. Large amount of OTA, zearalenone, fumonisins B, aflatoxin, and their metabolites were found in blood, kidney, liver, intestine, brain, and placenta of monstrous cat. Specific DNA adducts (related to OTA and/or zearalenone (ZEA)) were detected in the tissue of dead little cat. The amounts of individual mycotoxins are low, but their simultaneous occurrence was responsible for the reduced fertility and stillbirth because these mycotoxins have a synergistic effect (Hadjeba-Medjdoub et al., 2013). The continuous exposure to OTA—especially during the organogenesis period—might be responsible for the microscopic changes observed in vital organs of developing fetuses and also for the histopatologic alterations in the fetal organs of rabbits (Wangikar et al., 2005).

genase activity and reduces ovarian steroidogenesis in rats (Gupta et al., 1980). In human placenta, 3␤-HSD1 is expressed at high levels in synctial trophoblast. It catalyzes the conversion of 3␤-hydroxy-5-ene-steroids (dihydroepiandrosterone, pregnolone) to main 3-oxo-4-enesteroids (androstendione, progesterone) to maintain the uterus in a quiescent state (Thomas et al., 1989; Kacsoh, 2000). Androstendione is then converted by aromatase and 17␤-hydroxysteroid dehydrogenase (17␤-HSD) to estradiol. As 3␤-HSD1 is exclusively expressed in human placenta and plays an important role in placental steroidgenesis, modulation of placental 3␤-HSD1 activity may result in disturbance of steroid production and hence affect placental and fetal development (Woo et al., 2013). To study the effects of short- and long-term OTA exposure on the expression of 3␤-HSD1 and progesterone secretions, human placental cells human choriocarcinoma cell line (JEG-3) were exposed to OTA. It was shown that OTA upregulates 3␤-hydroxysteroid dehydrogenase type 1 expression in human placental cells. The induction of 3␤HSD1 mRNA leads to an increase in progesterone production (Woo et al., 2013). OTA also induces the production of estradiol (Frizzell et al., 2013). These data can partially explain the impact of OTA on kidney. Indeed, it has been demonstrated that endogenous sex steroid 17␤-E2 reduces renal basolateral organic cation transport through direct action on renal tubule cells (Pelis et al., 2007). In contrast, OTA inhibits testosterone secretion in vitro (Fenske and Fink-Gremmels, 1990). In rat, OTA inhibits both cortisol and testosterone secretions (Kumar et al., 2011). In another rat study, OTA induces folliclestimulating hormone, testosterone, triiodothyronine 3, and thyroxin, and reduces luteinizing hormone (Hassan et al., 2010). OTA and endosulfan (a pesticide) synergystically alter the hormonal status of male Wistar rats (Kumar et al., 2011).

OTA AND REPROTOXICITY

Male reproductive health is an important component of men’s overall health and wellbeing. Too often, males have been overlooked in discussions on reproductive health, especially when reproductive issues such as contraception and infertility have been perceived as femalerelated. Every day, men, their partners, and healthcare providers can protect their reproductive health by ensuring effective contraception, avoiding sexually transmitted diseases, and preserving fertility. Erectile dysfunction, premature ejaculation, loss of libido, testicular cancer, and prostate disease may cause embarrassment to the patient and, occasionally, the general practitioner (Wijesinha et al., 2013). Male reproductive health has deteriorated in many countries during the last few decades. Several toxins in environment have been suspected to affect reproductive system in male, and OTA is one of them (Verma and Chakraborty, 2008). Male fertility can be affected by impairment of sperm production and/or damage of testis. In mice, OTA exposure significantly decreases sperm count and motility and increases abnormalities in sperm morphology (Bose and Sinha, 1994; Biro´ et al., 2003; Farag et al., 2010). OTA (50 and 100 ␮g/day) given per os to male albino mice alters sperm count, sperm motility,

OTA and Endocrine Disruption All biologic functions in organism are controlled by hormones. Consequences of endocrine disruption are reproductive disorders (decreased libido, disturbed reproductive cycle, anovulation, disturbed spermatogenesis, infertility, neoplasmic lesion, altered behavior, decreased fertility of offspring), osteoporosis, myelofibrosis, adenoma, and skeletal deformations (Anonymous, 2013). Some data show that OTA can act as an endocrine disruptor by interfering with enzymes involved in the synthesis of the steroids or hormones. OTA does not interact directly with the receptor as it is the case for zearalenone, another mycotoxin (Kuiper-Goodman et al., 1987; Zinedine et al., 2007). The enzymatic complex 3␤-hydroxysteroid dehydrogenase (3␤-HSD) is located in the endoplasmic reticulum and mitochondria. Complex 3␤-HSD plays an essential role in the biosynthesis of all classes of steroid hormones. It catalyzes the oxidation and isomerization of 5– 3␤hydroxysteroid precursors into 4 -ketosteroids, first step in the biosynthesis of androgens and estrogens (Thomas et al., 1989). OTA suppresses 3-hydroxysteroid dehydroBirth Defects Research (Part B) 98:493–502, 2013

OTA and Impact on Sperm

496

MALIR ET AL.

sperm viability, and fertility rate in a dose-dependent way (Chakraborty and Verma, 2009). OTA also reduces the fertility of pigs. Indeed, OTA was found in seminal liquid of boar exposed by feed to 20 to 40 ␮g of OTA (Solti et al., 1999; Biro´ et al., 2003). The sperm production and semen quality of boars were decreased by OTA, because OTA provokes instability of the spermatozoal membrane (Ewald and Heer, 1989; Solti et al., 1999). These effects are more pronounced when pigs are simultaneously exposed to OTA and zearalenone (Posea et al., 2013).

OTA and Testicular Cancer In animals, OTA is toxic for testis (Mor´e and Camguilhem, 1979, Gharbi et al., 1993; Bose and Sinha, 1994; Biro´ et al., 2003). Several areas of the Balkans that are known to have high incidence of OTA contamination of food have recently experienced a marked increase in the incidence of testicular cancer. For example, from 1960 to 1985, the incidence of testicular cancer in Vas, Hungary, increased fivefold, from 1.2 to 6.1/100,000 (Huyghe et al., 2007). A similar increase was reported for Slovenia (Huyghe et al., 2007). A possible etiologic role for OTA in testicular cancer in men was hypothesized in 2002 by Schwartz, who noted that the unusual geographic distribution of testis cancer, with especially high rates in Denmark, was correlated with the national per capita consumption of OTAcontaminated foods. Recent toxicologic data in rodents support the concept that dietary exposure to OTA can influence carcinogenesis in the testis. For example, Ueta et al. (2010) administered 2 mg/kg OTA intraperitoneally to Pdn/Pdn mice on day 7.5 of gestation. They observed a significant downregulation of Dmrt-I gene expression in the male conceptuses on day 9 (Ueta et al., 2010). Dmrt-1 (also known as doublesex and mab-3–related transcription factor 1) is essential for the normal development of the mammalian testis (Raymond et al., 2000). DMRT1 is expressed in Sertoli cells and undifferentiated spermatogonia of the postnatal testis (Murphy et al., 2010). DMRT1 is a tumor suppressor gene in the testis; loss of this gene produces germ cell testicular tumors in mice (Krentz et al., 2009) and humans (Turnbull et al., 2010). More direct evidence of a carcinogenic role of OTA in the testis was provided by Mantle and Nolan (2010). These investigators administered 100-␮g OTA as a daily dietary contaminant to 24 male Fischer rats for 35 weeks. Testis tumors developed in six of the rats (25%), an incidence rate equal to that of renal tumors in male Fischer rats chronically exposed to dietary OTA. In a recent study, DNA damage to testis was proved experimentally by intrauterine exposure to OTA (2.5 mg/kg/bw) at day 17 of the gestation. DNA adducts in the testes of newborn mice are similar to the DNA adducts observed in the kidney and testes of adult mice exposed to OTA via the diet (Jennings-Gee et al., 2010). DNA adducts in the testes of mice exposed prenatally to OTA, and the absence of any such adducts in the testes of control mice not exposed prenatally to OTA, are clear evidence of the carcinogenic potential of OTA in the testes. The DNA adducts that we observed are not evidence merely of exposure (as Mantle (2010) suggests), they are markers of biologic effect, as DNA adducts are widely

considered to be markers of an increased risk of cancer (Bonassi et al., 2001; Knudsen and Hansen, 2007; PfohlLeszkowicz, 2008). Schwartz et al. (2010) substantiated the finding reported in Jennings-Gee et al. (2010) by explaining the recent research that has shown that prenatal exposure to OTA in mice significantly depresses the expression of the DMRT1 gene in offspring, particularly in male offspring (Ueta et al., 2010). Hence, significant molecular evidence supports the hypothesis that OTA may be causally related to germ cell testicular tumors in mice and in men. Future epidemiologic and molecular studies in humans are needed to draw a clear conclusion whether OTA is a causative agent in testicular cancer (Srinavasa, 2011).

OTA and Teratogenicity Teratogenicity is the ability to cause developmental anomalies in a fetus. Teratogens include viruses, chemicals, and radiation (Speight and Holford, 1997). The factors that determine the effects of teratogens include dose reaching fetus, step of the development when toxin exposure occurs, duration of exposure, environmental factors, and susceptibility of the fetus (Young and Koda-Kimble, 1995; Di Piro et al., 1997; Speight and Holford, 1997). The most common developmental toxicities are spontaneous abortion, congenital malformations, intrauterine growth retardation, mental retardation, carcinogenesis, and mutagenesis (Behrman, 1996; Di Piro et al., 1997; Speight and Holford, 1997; Hansen and Abbott, 2009). It is known that the gestation period is divided into several periods and that the fetus is more vulnerable during the period of organogenesis (i.e., in humans between 3 and 8 weeks after fertilization; in rats between 6 and 15 days and so on) with the increasing risk of spontaneous abortion and congenital malformations (congenital defects) of the fetus (Porter, 2004; Patil et al., 2006).The teratogenic effects of OTA have been well documented in many animal models (see Table 1). The reduced birth weight and craniofacial abnormalities are the most frequent signs observed (Patil et al., 2006). Most of the available data on OTA teratogenicity (developmental toxicity) come from rodent studies. Several studies have been conducted after intraperitoneal or subcutaneous administration (Hayes et al., 1974; Gilani et al., 1978; Mayura et al., 1982, 1984b; Wei and Sulik, 1993). A single subcutaneous treatment (1.75 mg OTA/kg) given on day 6 of gestation induces visceral and skeletal malformations in rat fetuses (Mayura et al., 1982). Dietary protein deficiency increases the susceptibility of the animal to the teratogenic effects of OTA notably skeletal development (Mayura et al., 1983). A single dose (2.5–20 mg OTA/kg bw) administrated by intraperitoneally (ip) to pregnant golden hamsters increases prenatal mortality between seventh and tenth day of gestation and diminishes fetal growth on the ninth day. Malformations such as micrognathia, hydrocephaly, short tail, oligodactyly, syndactyly, cleft lip, micromelia, and heart defects occurrs (Hood et al., 1976). Harmful effects of OTA are potentiated by other mycotoxins or contaminants. In particular, in mice treated intraperitoneally, OTA enhances the incidence of fetal malformations induced by T-2 toxin (Hood et al., 1978). Similarly subcutaneous Birth Defects Research (Part B) 98:493–502, 2013

REPROTOXICITY OF OCHRATOXIN

497

Table 1 Animal Teratology Studies Animal model Rat

Mice Hamster Chicken embryo Pig Quail Rabbit

Reference Still et al. (1971), Mor´e and Galtier (1974), Mor´e and Galtier (1975), Brown et al. (1976), Mor´e and Galtier (1978a, 1978b), Mayura et al. (1982, 1983, 1984a), Wei and Sulik (1993), Abdel-Wahhab et al. (1999), Wangikar et al. (2004a, 2004b) Hayes et al. (1974), Arora (1982), Petkova-Bocharova et al. (1998a), Katagiri et al. (2007), Jennings-Gee et al. (2010), Ueta et al. (2010) Hood et al. (1975, 1976), Petkova-Bocharova et al. (1998b) Gilani et al. (1978), Wiger and Stormer (1990), Wei and Sulik (1996), Lalitha Kunjamma and Nair (1997) Shreeve et al. (1977) Dwivedi (1984) Wangikar et al. (2004c, 2005)

administration of OTA and citrinin to rats increases prenatal toxicity and teratogenicity (Mayura et al., 1982). Nevertheless, first attention should be given primarily to oral intake, which appears to be natural and realistic way for reliable assessment of developmental disorders induced by food-borne mycotoxins such as OTA (Arora, 1982). Oral administration of OTA (5 mg/kg bw during 2 and 4 days) to the pregnant rats causes a reduced weight of fetus as well as a significant proportion of fetus with hemorrhages. In the F1 generation, there was a reduction in litter size and growth retardation, but no effects were proven in the F2 generation (Mor´e and Galtier, 1974, 1975, 1978b). In another study, oral administration of OTA (0.75 mg/kg bw) to pregnant rats between sixth and fifteenth day of pregnancy induces teratogenicity including visceral anomalies and skeletal anomalies, in addition to embryotoxicity (Brown et al., 1976). Dosing of pregnant rats per os (po) with 0.75 to 1 mg/kg OTA resulted in high incidence of embryo resorption, but also decreased fetal size and increased skeletal defects, primarily in the craniofacial region (Mayura et al., 1982, 1984a, 1984b). External hydrocephaly, omphalocele, and anophthalmia were the major gross anomalies. Internal hydrocephaly and shift in position of esophagus were the main internal soft tissue defects. Major skeletal defects involved sternebrae, vertebrae, and ribs (Mayura et al., 1982). Malformations included growth retardation, hypoplasia of the telencephalon, poor flexion, stunted limb bud development, underdeveloped sensory primordia, and decreased mandibular and maxillary size. Histologic examination demonstrated extensive necrosis of embryonal mesodermal structures and neuroectoderm (Mayura et al., 1989). OTA at different concentrations (2–4 mg/kg bw) and different gestations days (6–15) causes variable developmental defects (external hydrocephaly, incomplete closure of skull, and omphalocele) in fetuses (Patil et al., 2006; van Dorp et al., 2010). A single oral dose of 2.75 mg/kg bw was found as the minimum effective teratogenic dose in pregnant Wistar rats (Patil et al., 2006). The most critical gestational period for the induction of teratogenicity in rats was between the sixth and seventh day (Patil et al., 2006). Craniofacial malformations (exencephaly, midfacial clefting, and cleft lip) were found in

Birth Defects Research (Part B) 98:493–502, 2013

mice exposed to OTA (Wei and Sulik, 1993). Microcephalic brains in mice exposed to OTA are the consequences of a reduced dendritic growth (Fukui et al., 1992). Altogether, OTA is teratogenic in many animals including rat (Brown et al., 1976), mice (Arora et al., 1983), hamster (Hood et al., 1976), rabbit (Wangikar et al., 2004c), and chick (Wiger and Stormer, 1990). In summary, OTA provokes exencephaly, incomplete closure of skull, hypognathia, micromelia, scoliosis, curled tail, skeletal defects involving fused, bifurcated ribs as well soft-tissue anomalies like the ectopic or polycystic kidney, renal agenesis, hydrocephaly, and microphthalmia (Brown et al., 1976; Mayura et al., 1982; Wangikar et al., 2004b, 2004c). The severity and extent of various gross, skeletal, and visceral anomalies were proportional to the doses applied (Wangikar et al., 2004b). Both OTA and OTB (the dechloro analog of OTA) cause craniofacial malformations, while OTA also causes reduced embryo growth. As expected, OTA is by far more potent in inducing these effects than OTB (O´Brien et al., 2005). OTA induces neural-tube defects (NTDs) in rodent embryos (Wei and Sulik, 1993; Wangikar et al., 2004a) due to the inhibition of protein synthesis (Monnet-Tschudi et al., 1997; Wangikar et al., 2007). An antagonism between OTA and aflatoxin B1 (Wangikar et al., 2004a, 2004b) or between OTA and zearalenone is observed in mice exposed by oral route (Arora, 1982). OTA and aflatoxin B1 (AFB1 ) both interfered with the neural-tube development. However, when aflatoxin B1 and OTA are combined together, the effects on the neural tube decreased, but heart defects arose (Wangikar et al., 2007). In ovo inoculation of OTA in chicken embryos and subsequently in the hatching chicks induces teratogenic defects depending on the dose. The symptoms observed were anophthalmia (49% of the embryos), mandibular hypoplasia (45%), micropthalmia (35%), maxillary retrognathism (12%), reduced body size (15%), everted viscera (8%), spina bifida (10%), and exencephaly (4%; Hassan et al., 2012).

OTA and Molecular Mechanisms of Teratogenicity To define the mechanism of action an in vitro teratogen assay using rat embryo midbrain micromass cultures was

498

MALIR ET AL.

conducted (Walum and Flint, 1990). OTA induces cytotoxicity of all brain cells without any selectivity against neurons or astrocytes (Wilk-Zasadna and Minta, 2009). Morphometric assessment using computer imaging revealed that OTA teratogenic potency was correlated with their effects on aggregation of neuronal cells, and formation of neurospheres (Wilk-Zasadna and Minta, 2009). The gene responsible for the polydactyly/arhin encephaly (Pdn/Pdn) mouse, which exhibits polysyndactyly and arhinencephaly and has a 13.2% risk of NTDs, has been identified as Gli3. When Pdn/Pdn embryos were exposed to 2 mg OTA/kg administered intraperitoneally to the Pdn/+ females on day 7.5 and/or 7 of gestation, the incidence of NTD in Pdn/Pdn increased from 51.6% (Katagiri et al., 2007). Downregulation of the Gli3 gene induces the overexpression of the Fgf8 gene at the anterior neural ridge and of Emx2 gene in the dorsal forebrain in the Pdn/Pdn mice (Katagiri et al., 2007). Fgf8 is a growth factor important for the development of the telencephalon (Garel et al., 2003; Storm et al., 2003). Emx2 is homeobox gene (Simeone et al., 1992) playing and important role in the morphogenesis of the telencephalon, diencefalon, and mesencephalon (Suda et al., 1996; Acampora et al., 1997; Yoshida et al., 1997; Suda et al., 2001). OTA treatment accelerated the overexpression of these genes. Pretreatment with folic acid ameliorated the OTA-induced overexpression of Fgf8 in the Pdn/Pdn (Katagiri et al., 2007). Exposure to OTA led to increased embryolethality manifested as resorptions or dead fetuses. OTA had the same effect on all genotypes (Pdn/+; Pdn/Pdn; Katagiri et al., 2007). It was concluded that the increase of NTDs incidence could be consequence of OTA toxicity. More recently, gender-dependent differences in the incidence of NTDs induced by OTA were investigated in Pdn/Pdn mouse (Ueta et al., 2010). In this study, on the basis of real-time polymerase chain reaction analyses, it has been suggested that the manifestation of NTDs in the male OTA-treated Pdn/Pdn might be due to the complicated altered gene expressions among Gli3, Wntb7b, Wntb8b, Fez1, Barx1, Lim1, Dmrt1, Igf1, Fog2, Dax1, and Sox9, and in particular, upregulation and genderdependent difference in Barx 1 and gender-dependent difference in Sox9 gene expressions are noteworthy findings (Ueta et al., 2010). Moreover, prenatal exposure of mice to OTA also significantly downregulates the expression of the DMRT 1 (tumor suppressor gene) in male offspring (Ueta et al., 2010). In mice, it has been also shown that a single-OTA dose (0.5 or 2 mg/kg bw) given by gastric intubation to the mother on the 17th or 18th day of gestation induced OTA-DNA adducts in the kidney and liver of fetus and in newborn several months after birth, and in some of them later cancer tumors occurred (Petkova-Bocharova et al., 1998b). Similar results were obtained with hamster (Petkova-Bocharova et al., 1998a). Recent studies showed that in vitro exposure to OTA triggers apoptosis and retards early postimplantation development after transfer of embryos to host mice (Suda et al., 2001). In addition, OTA induces apoptosismediated injury of mouse blastocysts, via reactive oxygen species (ROS) generation, and promotes mitochondrion-

dependent apoptotic signaling processes that impair subsequent embryonic development (Bouaziz et al., 2011). The risk of spontaneous abortions associated with the exposure to endogenous substances may be modified by the genetic variation in individual metabolic detoxification activities, thus, in the phase I/phase II balance (Wang et al., 1998; Zusterzeel et al., 2000). The occurrence of the glutathione-S-transferase P1b-1b genotype, leading to lower glutathione-S-transferase Pi enzyme activity and, consequently, impairing placental detoxification, may represent a risk factor for recurrent early pregnancy loss (REPL; Zusterzeel et al., 2000). In a recent toxicogenomics study, the predominant effect of OTA was downregulation of RNA and protein expressions (Marin-Kuan et al., 2006). OTA is metabolized by conjugation with glutathione (Pfohl-Leszkowicz et al., 1993; El Adlouni et al., 2000; Pfohl-Leszkowicz et al., 2002; Tozlovanu et al., 2012). Presence of the GSTP 1b-1b genotype, associated with impaired detoxification, causes an imbalance between phase I and phase II of biotransformation, and therefore provokes the development of REPL. Thus, homosigoty for the GSTP 1b allele might be associated with an increased risk for REPL (Zusterzeel et al., 2000). Several data also pointed the implication of glutathione in OTA genotoxicity (Pfohl-Leszkowicz and Manderville, 2007; PfohlLeszkowicz, 2008; Pfohl-Leszkowicz and Manderville, 2012). Andonova et al. (2004) have shown that carriers of at least one glutathioneS-transferase M1 (GSTM1) wildtype allele (positive conjugators) were more prevalent among Balkan endemic nephropathy (BEN) patients compared to controls (OR = 7.92).

CONCLUSIONS AND RECOMMENDATIONS More than 1200 chemical and physical agents are known to cause structural and/or functional malformations in experimental animals (Shepard, 2001). Although the mechanisms by which these agents disrupt the normal development are not well understood, it is known that many teratogens induce cell death in tissues that subsequently develop abnormally causing structural malformations (Knudsen, 1997). These observations apply to OTA teratogenicity as well. In addition to be carcinogenic, OTA has been shown to be an endocrine disruptor and a reproductive toxicant, with abilities of altering sperm quality (Biro´ et al., 2003; Anonymous, 2013; Woo et al., 2013). Taking into account the ubiquitous OTA occurrence in food and feed, its long half-life, and important toxicity of OTA and its metabolites (e.g., the OP-OTA, as toxic as OTA; OTHQ, more toxic than OTA; genotoxicity, carcinogenicity and in animals clearly demonstrated teratogenicity; Xiao et al., 1996; Li et al., 2000; Faucet-Marquis et al., 2006; Tozlovanu et al., 2006; Pfohl-Leszkowicz, 2009), risk management should be implemented. Health Canada claims that OTA should be regulated as a nonthreshold carcinogen and the virtual safety dose (corresponding to negligible cancer risk) should be set above 4 ng/kg bw/day (Kuiper-Goodman et al., 2010). In addition, the harmful effects of OTA can be potentiated by possible synergistic action with other mycotoxins or contaminants. In light of the above-mentioned OTA toxic

Birth Defects Research (Part B) 98:493–502, 2013

REPROTOXICITY OF OCHRATOXIN properties and effects for the unborn child, OTA exposure of pregnant women should be kept as low as possible with an aim of the best protection.

REFERENCES Abdel-Wahhab MA, Nada SA, Arbid MS. 1999. Ochratoxicosis: prevention of developmental toxicity by L-methionone in rats. J Appl Toxicol 19:7–12. Acampora D, Avantaggiato V, Tuorto F, Simeone A. 1997. Genetic control of brain morphogenesis through Otx gene dosage requirement. Development 124:3639–3650. Andonova IE, Sarueva RB, Horvath AD, Simeonov VA, Dimitrov PS, Petropoulos EA, Ganev VS. 2004. Balkan endemic nephropathy and genetic variants of glutathione S-transferases. J Nephrol 17:390–398. Anonymous. 2013. NIEHS. Last reviewed, June 05, 2013. Endocrine Disruptors. Available at: www.niehs.nih.gov/health/topics/ agents/endocrine/. Appelgreen LE, Arora RG. 1983a. Distribution of 14 C- labelled ochratoxin A in pregnant mice. Food Chem Toxicol 21:563–568. Appelgreen LE, Arora RG. 1983b. Distribution studies of 14C-labelled aflatoxin B1 and ochratoxin A in pregnant mice. Food Chem Toxicol 21:563–568. Arora RG. 1982. Mycotoxin induced effects on the prenatal development in mice: a teratopathologic and autoradiographic study of aflatoxin B1, ochratoxin A and zearalenone. Ph.D. thesis, Swedish University of Agricultural Sciences, Uppsala, Sweden. Arora RG, Froelein H, Fellner-Feldegg H. 1983. Inhibition of ochratoxin A teratogenesis by zearalenone and diethylstilboestrol. Fundament Chem Toxicol 21:779–783. Azpilicueta CA, Arbeloa MI, de Maquirriain PFJ. 2008. Natural and syntethic occurring forms of the ochratoxins. In: Papadopoulos KN, editor. Food chemistry research developments. New York: Nova Science Publishers; 116p. Ballinger MB, Phillips TD, Kubena LF. 1986. Assessment of the distribution and elimination of ochratoxin A in the pregnant rat. J Food Safety 8:11–24. Behrman RE. 1996. Nelson texbook of pediatrics. 15th ed. Philadelphia: VB Saunders. p 448–451. Biasucci G, Calabrese G, Di Giuseppe R, Carrara G, Colombo F, Mandelli B, Maj M, Bertuzzi T, Pietri A, Rossi F. 2011. The presence of ochratoxin A in cord serum and in human milk and its correspondence with maternal dietary habits. Eur J Nutr 50:211–218. Biro´ K, Barna-Vetro´ I, P´ecsi T, Szabo´ E, Winkler G, Fink-Gremmels J, Solti L. 2003. Evaluation of spermatological parameters in ochratoxin Achallenged boars. Theriogenol 60:199–207. Bonassi S, Neri M, Puntoni R. 2001. Validation of biomarkers as early predictors of disease. Mutat Res 480–481:349–358. Bose S, Sinha SP. 1994. Modulation of ochratoxin – produced genotoxicity in mice by vitamin C. Food Chem Toxicol 32:533–537. Bouaziz C, Sharaf el dein O, Martel C, Golli EE, Abid-Essefi S, Brenner C, Lemaire C, Bacha H. 2011. Molecular events involved in ochratoxin A induced mitochondrial pathway of apoptosis, modulation by Bcl-2 family members. Environ Toxicol 26:579–590. Breitholtz-Emanuelsson A, Olsen M, Oskarsson A, Palminger I, Hult K. 1993. Ochratoxin A in cow’s milk and in human milk with corresponding human blood samples. J AOAC Int 76:842–846. Brown MH, Szczek GM, Purmalis BP. 1976. Teratogenic and toxic effets of ochratoxin A in rats. Toxicol Appl Pharmacol 37:331–338. Castegnaro M, Mohr U, Pfohl-Leszkowicz A, Esteve J, Steinmann J, Tillmann T, Michelon, J, Bartsch H. 1998. Sex- and strain-specific induction of renal tumors by ochratoxin A in rats correlates with DNA adduction. Inter J Cancer 77:70–75. Castegnaro M, Canadas D, Vrabcheva T, Petkova-Bocharova T, Chernozemsky IN, Pfohl-Leszkowicz A. 2006. Balkan endemic nephropathy: role of ochratoxins A through biomarkers. Mol Nutr Food Res 50:519–529. Chakraborty D, Verma R. 2009. Spermatotoxic effect of ochratoxin and its amelioration by Emblica officinalis aqueous extract. Act Pol Pharm Drug Res 66:689–695. Chu FS. 1971. Interaction of ochratoxin A with bovine serum albumine. Arch Biochem Biophys 147:359–366. Chu FS. 1974. A comparative study of the interaction of ochratoxins with bovine serum albumine. Biochem Pharmacol 23:1105–1107. Dai J, Park G, Perry JL, Ill’ichev YV, Bow DAJ, Pritchard JB, Faucet V, Pfohl-Leszkowicz A, Manderville R, Simon JD. 2004. Molecular as-

Birth Defects Research (Part B) 98:493–502, 2013

499

pects of the transport and toxicity of ochratoxin A. Acc Chem Res 37:874–881. Degen GH, Mayer S, Blaszkevicz M. 2007. Biomonitoring of ochratoxin A in grain workers. Mycotoxin Res 23:88–93. Di Piro JT, Talbert RL, Yee GC, Matzke GR, Wells BG, Posey LM. 1997. Pharmacotherapy: a pathophysiological approach. 3rd ed. Stamford, CT: Appleton Lauge. p 1564–1584. Duarte SC, Lino CM, Pena A. 2011. Ochratoxin A in feed of food-producing animals: an undesirable mycotoxin with health and performance effects. Vet Microbiol 154:1–13. Dwivedi P. 1984. The immunological and pathological changes in poultry induced by ochratoxin A. Ph.D. thesis, University of Edinburgh, UK. El Adlouni C, Pinelli E, Az´emar B, Zaoui D, Beaune P, Pfohl-Leszkowicz A. 2000. Role of CYP 2C and microsomal glutathione-S-transferase in modulating susceptibility to ochratoxin A genotoxicity. Environ Mol Mut 35:123–131. Ewald C, Heer, A. 1989. The sperm quality of 4 boars during the assimilation of mycotoxin-contaminated feed (ochratoxin A) and after changing feed-a case study. Berl Munch Tierarztl Wochenschr 102:261–266. Farag IM, Abdel-Aziz KB, Nada SA, Tawfek NS, Farouk T, Darwish HR. 2010. Modulation of ochratoxin-induced oxidative stress, genotoxicity and spermatotoxic alterations by Lactobacillus rhamnosus GC in male Albino mice. J Am Sci 6:575–587. Faucet-Marquis V, Pont F, Størmer FC, Rizk T, Castegnaro M, PfohlLeszkowicz A. 2006. Evidence of a new dechlorinated OTA derivative formed in opossum kidney cell cultures after pre-treatment by modulators of glutathione pathways. Correlation with DNA adducts formation. Mol Nutr Food Res 50:530–542. Fenske M, Fink-Gremmels J. 1990. Effects of fungal metabolites on testosterone secretion in vitro. Arch Toxicol 64:72–75. Frizzell C, Verhaegen S, Ropstad E, Elliott CT, Connally L. 2013. Endocrine disrupting effects of ochratoxin A at the level of nuclear receptor activation and steroigenesis. Toxicol Lett 217:243–250. Fukui Y, Hoshino K, Kameyama Y, Yasui T, Toda C, Nagano H. 1987. Placental transfer of ochratoxin A and its cytotoxin effect on the mouseembryonic brain. Food Chem Toxicol 25:17–24. Fukui Y, Hyasaka S, Itoh M, Takeuschi Y. 1992. Development of neurons and synapses in ochratoxin A-induced microcephalic mice: a quantitative assessment of somatosensory cortex. Neurotoxicol Teratol 14:191–196. Gareis M, M¨artlbauer E, Bauer J, Gedek B. 1988. Determination of ochratoxin A in human milk. Z Lebensm Unters Forsch 186:114–117. Garel S, Huffman KJ, Rubenstein JLR. 2003. Molecular regionalization of the neocortex is disrupted in Fgf8 hypomorphic mutants. Development 130:1903–1914. Gharbi A, Trillon O, Betbeder AM. 1993. Some efects of ochratoxin A, a mycotoxin contaminating feeds and food, on rat testis. Toxicol 83:9– 18. Gilani SH, Bancroft J, Reily M. 1978. Teratogenicity of ochratoxin A in chick embryos. Tox Appl Pharmacol 46:543–546. Gilbert J, Brereton P, MacDonald S. 2001. Assessment of dietary exposure to ochratoxin A in the UK using a duplicate diet approach and analysis of urine and plasma samples. Food Addit Contam 18:1088–1093. Gupta M, Bandyopadhyay S, Mazumdar SK, Paul B. 1980. Ovarian steroidogenesis in rats following ochratoxin A treatment. Toxicol Appl Pharmacol 53:515–520. Hadjeba-Medjdoub K, Faucet-Marquis V, Monje MC, Dohnal V, Polisenska I, Pfohl-Leszkowicz A. 2013. Is mycotoxin-contaminated pet-food responsible of reduced fertility and stillbirth? Proceeding of 35th Mycotoxin Workshop, Ghent, Belgium, May 2013. 104p. Hallen IP, Breitholtz-Emanuelsson A, Hult K, Olsen M, Oskarsson A. 1998. Placental and lactational transfer of ochratoxin A in rats. Nat Toxins 6:43–49. Hansen DK, Abbot DB. 2009. Developmental toxicology. 3rd ed. New York: Informa Healthcare USA p 1–384. Hassan AA, Rashid MA, Koratum KM. 2010. Effect of aflatoxin B1 , zearalenone and ochratoxin A on some hormones related to fertility in male rats. Life Sci J 7:64–72. Hassan ZU, Khan MZ, Saleemi MK, Khan A, Javed I, Bhatti SA. 2012. Toxico-pathological effects of in ovo inoculation of ochratoxin A (OTA) in chick embryos and subsequently in hatched chicks. Toxicol Pathol 40:33–39. Hayes AW, Hood RD, Lee HL. 1974. Teratogenic effects of ochratoxin A in mice. Teratology 9:93–98. Hood RD, Naughton MJ, Hayes AW. 1975. Teratogenic effects of ochratoxin A in hamsters. Teratology 11:23 A (abst.). Hood RD, Naughton MJ, Hayes AW. 1976. Prenatal effects of ochratoxin A in hamsters. Teratology 13, 11–14.

500

MALIR ET AL.

Hood RD, Kuczuk MH, Szczech GM. 1978. Effects in mice of simultaneous prenatal exposure to ochratoxin A and T-2 toxin. Teratology 17:25–30. Hsuuw YD, Chan WH, Yu JS. 2013. Ochratoxin A inhibits mouse embryonic development by activating a mitochondrion-dependent apoptotic signaling pathway. Int J Mol Sci 14:935–953. Huyghe E, Plante P, Thonneau PF. 2007. Testicular cancer variations in time and space in Europe. Eur Urol 51:621–628. IARC. 1993. Monographs on the evaluation of carcinogenic risks to humans: Some naturally occuring substances: Food items and costituents, heterocyclic aromatic amines and mycotoxins. Lyon, France: IARC 56:523–524. Jennings-Gee JE, Tozlovanu M, Manderville R, Miller MS, PfohlLeszkowicz A, Schwartz GG. 2010. Ochratoxin A: in utero exposure in mice induces adducts in testicular DNA. Toxins 2:1428–1444. Jonsyn FE, Maxwell SM, Hendrickse RG. 1995. Human fetal exposure to ochratoxin A and aflatoxins. Ann Trop Paediatr 15:3–9. Joosten HMLJ, Goetz J, Pittet A, Schellenberg M, Bucheli P. 2001. production of ochratoxin A by Aspergillus carbonarius on coffee cherries. Int J Food Microbiol 65:39–44. Kacsoh, B. 2000. Endocrine physiology. New York, San Francisco, St.Louis, Auckland, Bogota, Caracas, London: The Mc Graw-Hill companies, Inc. Katagiri R-I, Kurome M, Teshima Y, Ueta E, Naruse I. 2007. Prevention of ochratoxin A-induced neural tube defects by folic acid in the genetic polydactyly/arhinencephaly mouse, Pdn/Pdn. Congenit Anom 47:90– 96. ˇ Klapec T, Sarkanj B, Banjari I, Strelec I. 2012. Urinary ochtratoxin A and ochratoxin alpha in pregnant women. Food Chem Toxicol 50:4487– 4492. ˚ 2007. Biomarkers of intermediate endpoints in Knudsen LE, Hansen A. environmental and occupational health. Int J Hyg Environ Health 210:461–470. Knudsen TB. 1997. Cell death. In: Kavlock RJ, Daston GP, editors. Drug toxicity in embryonic development I. Vol 124. Berlin: Springer. p 211– 244. Kovacs F, Sandor G, Vanyi A, Domany S, Zomborsky-Kovacs M. 1995. Detection of ochratoxin A in human blood and colostrum. Acta Vet Hung 43:393–400. Krentz A, Murphy MW, Kim S, Cook MS, Capel B, Zhu R, Matin A, Sarver AL, Parker KL, Griswold MD, Looijenga LHJ, Bardwell VJ, Zarkower D. 2009. The DM domain protein DMRT1 is a dose-sensitive regulator of fetal germ cell proliferation and pluripotency. Proc Natl Acad Sci USA 106:22323–22328. Kuiper-Goodman T, Scott PM, Watanabe H. 1987. Risk assessment of the mycotoxin zearalenone. Regul Toxicol Pharmacol 7: 253–306. Kuiper-Goodman T, Hilts C, Billiard SM, Kiparissis Y, Richard IDK, Hayward S. 2010. Health risk assessment of ochratoxin A for all age-sex strata in a market economy. Food Addit Contam 27:212–240. Kumar SN, Telang AG, Singh KP, Jain AK, Afroz M, Patil RD. 2011. Experimentally induced toxicity of ochratoxin A and endosulfan in male wistar rats: a hormonal disorder in adult male rats. J Anim Vet Adv 10(13):1750–1755. Lalitha Kunjamma CR, Nair MK. 1997. Pathomorphology of the combined effects of ochratoxin A and citrinin in chicken embryos. Indian Vet J 74:22–25 Li S, Marquardt RR, Frohlich AA. 2000. Identification of ochratoxins and some of their metabolites in bile and urine of rats. Food Chem Toxicol 38:141–152. Loebstein R, Koren G. 2002. Clinical relevance of therapeutic drug monitoring during pregnancy. Ther Drug Monit 24:15–22. Malir F, Ostry V, Pfohl-Leszkowicz A, Roubal T. 2012. Ochratoxin A exposure biomarkers in the Czech Republic and comparison with foreign countries. Biomarkers 17:577–589. Malir F, Ostry V, Novotna E. 2013a. Toxicity of the mycotoxin ochratoxin A in the light of recent data. Toxin Rev 32:19–33. Malir F, Ostry V, Dofkova M, Roubal T, Dvorak V, Dohnal V. 2013b. Ochratoxin A levels in blood serum of Czech women in the first trimester of pregnancy and its correspondence with dietary intake of the mycotoxin contaminant. Biomarkers 18:673–678. Mantle PG. 2010. Comments on “Ochratoxin A: In utero Exposure in Mice Induces Adducts in Testicular DNA. Toxins 2010, 2:1428–1444”–Miscitation of rat literature to justify a hypothetical role for ochratoxin A in testicular cancer. Toxins 2:2333–2336. Mantle PG, Nolan CC. 2010. Pathological outcomes in kidney and brain in male fischer rats given dietary ochratoxin A, commencing at one year of age. Toxins 2:1100–1110.

Marin-Kuan M, Nestler SW, Verguet C, Bezencon D, Piguet R, Mansourian R, Holzwarth J, Grigorov M, Delatour T, Mantle P, Cavin C, Schilter B. 2006. A toxicogenomics approach to identify new plausible epigenetic mechanisms of ochratoxin A carcinogenicity in rat. Toxicol Sci 89:120– 134. Mayura K, Reddy RV, Hayes AW, Berndt WO. 1982. Embryocidal, fetotoxic and teratogenic effects of ochratoxin A in rats. Toxicology 25:175–185. Mayura K, Hayes AW, Berndt WO. 1983. Effects of dietary protein on teratogenicity of ochratoxin A in rats. Toxicol 27:147–157. Mayura K, Parker R, Berndt WO, Phillips TD. 1984a. Effects of simultaneous prenatal exposure to ochratoxin A and citrinin in the rat. J Toxicol Environ Health 13:553–561. Mayura K, Stein AF, Berndt WO, Phillips TD. 1984b. Teratogenic effects in rats with impaired renal function. Toxicology 32:277–285. Mayura K, Edwards JF, Maull EA, Phillips. 1989. The effects of ochratoxin A on postimplantation rat embryos in culture. Arch Environ Contam Toxicol 18:411–415. Micco C, Ambruzzi MA, Miraglia M, Brera C, Onori R, Benelli L. 1991. Contamination of human milk with ochratoxin A. IARC Sci Publ 115:105–108. Minervi F, Giannoccaro A, Nicassio M, Panzarini G, Lacalandra GM. 2013. First evidence of placental transfer of ochratoxin A in horses. Toxins 5:84–92. Miraglia M, Brera C, Corneli S, Cava E. 1998. Occurrence of ochratoxin A (OTA) in maternal serum, placenta and funiculum. In: Miraglia M, van Egmond H, Brera C, Gilbert J, Eds. Proceed.of IX International IUPAC Symposium on Mycotoxins and Phycotoxins-Developments in Chemistry, Toxicology and Food Safety. Alaken Inc.: Fort Collins, Co, USA, pp. 165–179. Monnet-Tschudi F, Sorg O, Honegger P, Zurich MG, Huggett AC, Schilter B. 1997. Effects of the naturaly occurring food mycotoxin ochratoxin A on brain cells in culture. Neurotoxicol 18:831–839. Mor´e J, Camguilhem R. 1979. Efects of low doses of ochratoxin A after intratesticular injection in the rat. Experientia 35:890–892. Mor´e J, Galtier P. 1974. Toxicit´e de l´ochratoxine A. I. Effet embryotoxique et teratog`ene chez le rat. Ann Rech Vet 5:167–178. Mor´e J, Galtier P. 1975. Toxicit´e de l´ochratoxine A. II. Effets du traitement sur la descendance (F1 et F2) des rates intoxiqu´ees. Ann Rech Vet 6:379–389. Mor´e J, Galtier P. 1978a. Toxicit´e de l´ochratoxine A. III. Effets pendant les stades initiaux de la gestation Chez le rat. Ann Rech Vet 8: 169–173. Mor´e J, Galtier P. 1978b. Embryotoxic and teratogenic effects of OA in rats. In XIXth morphological congess symposia, Prague, Charles University. p 321–326. ˜ K, Campos V, Blaszkewicz M, Vega M, Alvarez A, Neira J, Degen Munoz GH. 2010. Exposure of neonates to ochratoxin A: first biomonitoring results in human milk (kolostrum) from Chile. Mycotoxin Res 26:59– 67. Murphy MW, Sarver AL, Rice D, Hatzi K, Ye K, Melnick A, Heckert LL, Zarkower D, Bardwell VJ. 2010. Genome-wide analysis of DNA binding and transcriptional regulation by the mammalian Double sex homolog DMRT1 in the juvenile testis. Proc Natl Acad Sci USA 107:13360–13365. Myllynen P, V¨ah¨akangas K. 2013. Placental transfer and metabolism: an overview of the experimental models utilizing human placental tissue. Toxicol In Vitro 27:507–512. O´Brien E, Prietz A, Dietrich DR. 2005. Investigation of the teratogenic potential of ochratoxin A and B using the FETAX system. Birth Defects Res B Dev Reprod Toxicol 74:417–423. Ostry, V., Malir, F., Ruprich, J. 2013. Producers and important dietary sources of ochratoxin A and citrinin. Toxins 5:1574–1586. Patil RD, Dwivedi P, Sharma AK. 2006. Critical period and minimum single oral dose of ochratoxin A for inducing developmental toxicity in pregnant Wistar rats. Reprod Toxicol 22:679–687. Pelis RM, Hartman RC, Wright SH, Wunz TM, Groves CE. 2007. Influence of estrogen and xenoestrogens on basolateral uptake of tetraethylammonium by opossum kidney cells in culture. J Pharmacol Exp Ther 323:555–561. Petkova-Bocharova T, Pfohl-Leszkowicz A, Hadjiolov D, Spirov K. 1998a. Investigation on the genotoxicity of ochratoxin A by measurement of DNA-adducts in different organs of Syrian hamsters. Rev Med Vet 149:658. Petkova-Bocharova T, Stoichev II, Chernozemsky IN, Castegnaro M, Pfohl-Leszkowicz A. 1998b. Formation of DNA adducts in tissues of mice progeny through transplacental contamination and/or

Birth Defects Research (Part B) 98:493–502, 2013

REPROTOXICITY OF OCHRATOXIN lactation after administration of a single dose of ochratoxin A to pregnant mother. Environ Molecular Mutagen 32:155–162. Pfohl-Leszkowicz A. 2008. Formation, persistence and significance of DNA adduct formation in relation to some pollutants from a broad perspective. Adv Mol Toxicol 2:183–239. Pfohl-Leszkowicz A. 2009. Ochratoxin A and aristolochic acid involvement in nephropathies and associated urothelial tract tumours. Arh Hig Rada Toksikol 60:465–483. Pfohl-Leszkowicz A. 2013. Evaluation de risques li´es a` l’exposition, en milieu professionnel et environnemental, aux mycotoxines. Bulletin de veille scientifique ANSES, N◦ 22. Pfohl-Leszkowicz A, Manderville RA. 2007. Ochratoxin A: an overview on toxicity and carcinogenicity in animals and humans. Mol Nutr Food Res 51:61–99. Pfohl-Leszkowicz A, Manderville RA. 2012. An update on direct genotoxicity as a molecular mechanism of ochratoxin A carcinogenicity. Chem Res Toxicol 25:252–262. Pfohl-Leszkowicz A, Grosse Y, Kane A, Gharbi A, Baudrimont I, Obrecht S, Creppy EE, Dirheimer G. 1993. Is the oxydative pathway implicated in the genotoxicity of ochratoxin A? In: Human ochratoxicosis and related pathologies, Montrouge and London: Libbey J, eds, London Colloque INSERM, 231:177–187. Pfohl-Leszkowicz A, Pinelli E, Bartsch H, Mohr U, Castegnaro M. 1998. Sex and Strain differences in ochratoxin A metabolism and DNA adduction in two strains of rats Mol Carcinogenesis 23:76–83. Pfohl-Leszkowicz A, Bartsch H, Az´emar B, Mohr U, Est`eve J, Castegnaro M. 2002. MESNA protects rats against nephrotoxicity but not carcinogenicity induced by ochratoxin A, implicating two separate pathways. Facta Universitatis Series Medicine and Biology 9:57–63. Pfohl-Leszkowicz A, Tozlovanu M, Stepanovic J, Stefanovic V, Manderville R, Castegnaro M. 2006. Comparative genotoxicity of ochratoxin A and aristolochic acid in human kidney cells: interpretation of ongoing analyses of food, blood, urine and kidney tissue from Serbia. Colleg Antropologic Suppl 30:1–17. Pfohl-Leszkowicz A, Tozlovanu M, Manderville R, Peraica M, Castegnaro M, Stefanovic V. 2007. New molecular and field evidences for the implication of mycotoxins but not aristolochic acid in human nephropathy and urinary tract tumor. Mol Nutr Food Res 51:131–146. Porter RS. 2004. The merck manual’s online medical library. Whitehouse Station: Merck Research Lab. Posea C, Sonea A, Birtoiu A, Roman M, Vasile M, Bensce A, Ion C. 2013. The presence of mycotoxins (OTA and ZEA) in feed for pigs and their influence on reproduction. Seria Med Vet 56:322–329. Postupolski J, Karlowski K, Kubik P. 2006. Ochratoxin A in maternal and metal blood and in maternal milk. Rocz Panstw Zakl Hig 57:23–30. Raymond CS, Murphy MW, O’Sullivan MG, Bardwell VJ, Zarkower D. 2000. Dmrt1, a gene related to worm and fly sexual regulators, is required for mammalian testis differentiation. Genes Dev 14: 2587–2595. Rosner H, Rohrmann B, Peiker G. 2000. Ochratoxin A in human serum. Arch Lebensmittelhyg 51:104–107. Schwartz GG, Manderville RA, Pfohl-Leszkowicz A. 2010. Response to comments of Peter G. Mantle. Toxins 2:2337–2339. SCOOP. 2002. Assessment of dietary intake of Ochratoxin A by the population of EU member states. Reports on tasks for scientific cooperation TASK 3.2.7, European Commission, 153p. Scott PM. 2005. Biomarkers of human exposure to ochratoxin A. Food Addit Contam 22(Suppl 1):99–107. Shepard TH. 2001. Catalog of teratogenic agents. 10th ed. Baltimore, MD: Johns Hopkins University Press. Shreeve BJ, Patterson DS, Pepin GA, Roberts BA, Wrathal AE. 1977. Effect of feeding ochratoxin A to pigs during early pregnancy. Br Vet J 133:412–417. Simeone A, Gulisano M, Acampora D, Stornaiuolo A, Rambaldi M, Boncinelli E. 1992. Two vertebrate homeobox genes related to the Drosophila empty spiracles gene are expressed in the embryonic cerebral cortex. EMBO J 11:2541–2550. Skaug MA, Stormer FC, Saugstad OD. 1998. Ochratoxin A: a naturally occurring mycotoxin found in human milk samples from Norway. Acta Paediatr 87:1275–1278. Solti L, P´ecsi T, Barna-Vetro´ I, Sz´asz F, Biro´ K, Szabo E. 1999. Analysis of serum and seminal plasma after feeding ochratoxin A with breeding boars. Anim Reprod Sci 56:123–132. Speight TM, Holford NHG. 1997. Avery´s drug treatment. 4th ed. Auckland: Adis International Limited. p 76–126. Srinavasa J. 2011. Does ochratoxin A (OTA) cause testicular cancer in humans? Act Med Lituan 18:1–3.

Birth Defects Research (Part B) 98:493–502, 2013

501

Still PE, Macklin AW, Ribelin WE, Smalley EB. 1971. Relationship of ochratoxin A to fetal death in laboratory and domestic animals. Nature 234:563–564. Storm EE, Rubenstein JL, Martin GR. 2003. Dosage of Fgf8 determines whether cell survival is positively or negatively regulated in the developing forebrain. Proc Natl Acad Sci USA 100:1757–1762. Suda Y, Matsuo I, Kuratani S, Aizawa S. 1996. Otx1 function overlaps with Otx2 in development of mouse forebrain and midbrain. Genes Cells 1:1031–1044 Suda Y, Hossain ZM, Kobayashi C, Hatano O, Yoshida M, Matsuo I, Aizawa S. 2001. Emx2 directs the development of diencefalon in cooperation with Otx2. Development 128:2433–2450. Thomas JL, Mayers RP, Strickler RC. 1989. Human placental 3[beta]hydroxy-5-enesteroid dehydrogenase and steroid 5–4-ene-isomerase: purification from mitochondria and kinetic profiles, biophysical characterization of the purified mitochondrial and microsomal enzymes. J Steroid Biochem 33:209–217. Tozlovanu M, Faucet-Marquis V, Pfohl-Leszkowicz A, Manderville Richard A. 2006. Genotoxicity of the hydroquinone metabolite of ochratoxin A: structure-activity relationships for covalent DNA adduction. Chem Res Toxicol 19:1241–1247. Tozlovanu M, Canadas D, Pfohl˙Leszkowicz A, Frenette Ch, Paugh RJ, Manderville RA. 2012. Glutathione conjugates of ochratoxin A as biomarkers of exposure. Arh Hig Rada Toksikol 63:417–427. Turnbull C, Rapley EA, Seal S, Pernet D, Renwick A, Hughes D, Ricketts M, Linger M, Nsengimana J, Delouhas P, Huddart RA, Bishop DT, Easton DF, Stratton MR, Rahman N. 2010. Variants near DMRT1, TERT and ATF7IP are associated with testicular germ cell cancer. Nat Genet 42:604–608. Ueta E, Kodama M, Sumino Y, Kurome M, Ohta Ki, Kataigiri Ri, Naruse I. 2010. Gender-dependent differences in the incidence of ochratoxin Ainduced neural tube defects in the Pdn/Pdn mouse. Congenit Anom 50:29–39. van Dorp DR, Malleis JM, Sullivan BP, Klein MD. 2010. Teratogens inducing congenital abdominal wall defects in animal models. Pediatr Surg Int 26:127–139. Verma R, Chakraborty D. 2008. Emblica officinalis aqueous extract ameliorates ochratoxin induced lipid peroxidation in the testes of mice. Act Pol Pharm Drug Res 65(2):187–194. Walum E, Flint OP. 1990. Midbrain micromass culture: a model for studies of teratogenic and subteratogenic effects on CNS development. Acta Physiol Scand 592(suppl 1):61–72. Wang X, Wang M, Niu T, Chen Ch, Xu X. 1998. Microsomal epoxide hydrolase polymorphism and the risk of spontaneous abortion. Epidemiol 9:540–544. Wangikar PB, Dwivedi P, Sharma AK, Sinha N. 2004a. Effect in rats of simultaneous prenatal exposure to ochratoxin A and aflatoxin B1 . II. Histopatological features of teratogenical anomalies induced in fetuses. Birth Defects Res B Dev Reprod Toxicol 71:352–358 Wangikar PB, Dwivedi P, Sinha N. 2004b. Effects in rats of simultaneous prenatal exposure to ochratoxin A and aflatoxin B1 . I. Maternal toxicity and fetal malformations. Birt Def Res B Dev Reprod Toxicol 71:343– 353. Wangikar PB, Dwivedi P, Sinha N. 2004c. Teratogenic effects of ochratoxin A in rabbits. World Rabbit Sci 12:159–171. Wangikar PB, Dwivedi P, Sharma AK, Sinha N, Telang AG. 2005. Teratogenic effects in rabbits of simultaneous exposure to ochratoxin A and aflatoxin B1 with special reference to microscopic effects. Toxicology 215:37–47. Wangikar PB, Sinha N, Dwivedi P, Sharma AK. 2007. Teratogenic effects of ochratoxin A and aflatoxin B1 alone and in combination on postimplantation rat embryos in culture. J Turkish-German Gynecol Assoc 8:357–364. Wei X, Sulik KK. 1993. Pathogenesis of craniofacial and body wall malformations induced by ochratoxin A in mice. Am J Med Gen 47:862–871. Wei X, Sulik KK. 1996. Pathogenesis of caudal dysgenesis/sirenomelia induced by ochratoxin A in chick embryos. Teratology 53:378–391. Wiger R., Stormer FC. 1990. Effects of ochratoxins A and B on prechondrogenic mesenchymal cells from chick embryo limb buds. Toxicol Lett 54:129–134. Wijesinha S, Piterman L, Kirby CN. 2013. The male reproductive system – an overview of common problems. Aust Fam Physician 42:276–278. Wilk-Zasadna I, Minta M. 2009. Developmental toxicity of ochratoxin A in rat embryo midbrain micromass cultures. Int J Mol Sci 10:37–49. Woo CSJ, Partanen H, Myllynen P, V¨ah¨akangas K, El Nezami-H. 2012. Fate of the teratogenic and carcinogenic ochratoxin A in human perfused placenta. Toxicol Lett 208:92–99.

502

MALIR ET AL.

Woo CSJ, Wang LMY, Ahokas J, El Nezami H. 2013. Potential endocrine disrupting effect of ochratoxin A on human placental 3 ␤ hydroxysteroid dehydrogense/isomeras in JEG-3 cells at levels relevant to human exposure. Reprod Toxicol 38:47–52. Wu Q, Dohnal V, Huang L, Kuca K, Wang X, Chen G, Yuan Z. 2011. Metabolic pathways of ochratoxin A. Curr Drug Metab 12:1–10. Xiao H, Marquardt RR, Frohlich AA, Ling YZ. 1995. Synthesis and structural elucidation of analogs of ochratoxin A. J Agric Food Chem 43:524–530. Xiao H, Madhyastha S, Marquardt R, Li S, Vodela J, Frohlich A, Kemppainen B. 1996. Toxicity of ochratoxin A, its opened lactone form and several of its analogs: Structure–activity relationships. Toxicol Appl Pharmacol 137:182–192. Yoshida M, Suda Y, Matsuo I, Miyamoto N, Takeda N, Kuratani S, Aizawa S. 1997. Emx1 and Emx2 functions in development of dorsal telencefalon. Development 124:101–111.

Young LY, Koda-Kimble MA. 1995. Applied theraputics: the clinical use of drugs. 6th ed. Vancouver, WA: Applied Therapeutics. p 12– 14. Zimmerli B, Dick R. 1995. Determination of ochratoxin A at the ppt level in human blood, serum, milk and some foodstuffs by highperformance liquid chromatography with enhanced fluorescence detection and immunoaffinity column cleanup: methodology and Swiss data. J Chromatogr B Biomed Appl 666:85–99. Zinedine A, Soriano JM, Molto JC, Manes J. 2007. Review on the toxicity, occurence, metabolism, detoxification, regulation and intake of zearalenone:an oestrogenic mycotoxin. Food Chem Toxicol 45: 1–18. Zusterzeel PLM, Nelen WLDM, Roelofs HMJ, Petrs WHM, Blom HJ, Steegers EAP. 2000. Polymorphisms in biotransformation enzymes and the risk for recurrent early pregnancy loss. Mol Hum Reprod 6:474–478.

Birth Defects Research (Part B) 98:493–502, 2013

Ochratoxin A: developmental and reproductive toxicity-an overview.

Ochratoxin A (OTA) is nephrotoxic, hepatotoxic, reprotoxic, embryotoxic, teratogenic, neurotoxic, immunotoxic, and carcinogenic for laboratory and far...
165KB Sizes 0 Downloads 0 Views