GET THE APP

Individual and Combined Effects of Subchronic Exposure of Three F
Journal of Clinical Toxicology

Journal of Clinical Toxicology
Open Access

ISSN: 2161-0495

+44 1478 350008

Research Article - (2015) Volume 5, Issue 4

Individual and Combined Effects of Subchronic Exposure of Three Fusarium Toxins (Fumonisin B, Deoxynivalenol and Zearalenone) in Rabbit Bucks

Judit Szabó-Fodor1*, Mariam Kachlek1, Sándor Cseh3, Bence Somoskői3, András Szabó1,2, Zsófia Blochné Bodnár2, Gábor Tornyos2, Miklós Mézes4, Miklós Mézes4, Krisztián Balogh4, Róbert Glávits4, Dóra Hafner2 and Melinda Kovács1,2
1Faculty of Agricultural and Environmental Sciences, MTA-KE Mycotoxins in the Food Chain Research Group, Kaposvár University, Hungary
2Faculty of Agricultural and Environmental Sciences, Kaposvár University, Hungary
3SZIU Faculty of Veterinary Science, István, Hungary
4SZIU Faculty of Agricultural and Environmental Sciences, Hungary
*Corresponding Author: Judit Szabó-Fodor, Faculty of Agricultural and Environmental Sciences, MTA-KE Mycotoxins in the Food Chain Research Group, Kaposvár University, Hungary, Tel: +36304303345 Email:

Abstract

Objective of the study was to determine reproductive toxicity of Fusarium toxins orally at three subchronic doses on adult Pannon White male rabbits. The four treatments were: control (C, toxin-free diet), F (5 mg/kg FB1), DZ (1 mg/kg DON+0.25 mg/kg ZEA), FDZ (5 mg/kg FB1+1 mg/kg DON+0.25 mg/kg ZEA) for 65 days (n=15/treatment). The doses were pre-determined according the EU limits in finished feed for young pig (in the absence of limits for rabbits’ feed; based on the European Commission Recommendation 2006/576/EC and the European Commission Directive 2003/100/EC). The most pronounced effects of the toxins were exerted on the reproductive processes. The ratio of spermatozoa showing progressive forward motility decreased (P<0.05) from 80% to 67% in the FDZ group by day 60. Differences were found between the groups DZ (66.3% ± 23.7) and C (80.2% ± 11.2) in spermatozoa morphology. GnRH treated animals produced less testosterone in FDZ animals, compared to the other three groups (P<0.05). In the comet assay the individual fumonisin treatment resulted in significantly less 0 comets (intact cells), compared to all others. Based on the prevalence of score, lower (P<0.0001) damage was observed in FDZ group, as compared to F and DZ. Among the mycotoxins studied, additive or less than additive effect was found in case of spermatogenesis and sperm cell morphology, synergism in testosterone production, while FB1 acted antagonistically against DON+ZEA in comet assay. All mycotoxins provoked moderate lipid-peroxidation, based on the changes of glutathione concentration, glutathione peroxidase activity and formation of malondialdehyde and conjugated dienes and trienes, and exerted slight genotoxicity based on comet assay, FB1 being antagonistic towards DON+ZEA. In F, DZ and FDZ animals the intensity of spermatogenesis decreased by 43, 31 and 64%, respectively, which was reflected by lack of differentiated spermatozoa, thinning of the germinal epithelium, the appearance of multinuclear giant cells, indicative of the disturbance of meiosis and mitosis of the germinal epithelial cells and in some cases the lack of spermatogonia.

Keywords: Fumonisin; Deoxynivalenol; Zearalenon; Spermatogenesis; Testosterone production; Rabbit

Introduction

The worldwide contamination of foods and feeds with mycotoxins is a significant problem. Studies have shown extensive mycotoxin contamination in both developing and developed countries [1-3], as effect of global climate change [4].

The toxicity of mycotoxins differs depending on the kind of toxin, and in animals it is related to the species, the dose ingested, the duration of the exposure, and their gender and age. The main classes of Fusarium mycotoxins with respect to animal health and production are the non-oestrogenic trichothecenes such as deoxynivalenol (DON) and T-2 toxin (T-2), the myco-oestrogens including zearalenone (ZEA) and its zearalenol metabolites [5] and fumonisins.

Fumonisins (B1 and B2) are cancer-promoting metabolites of Fusarium proliferatum and Fusarium verticillioides that have a long-chain hydrocarbon unit (similar to that of sphingosine and sphinganine), which plays a role in their toxicity. Fumonisin B1 (FB1) is the most toxic and has been shown to promote tumour in rats and cause equine leukoencephalomalacia and porcine pulmonary oedema. FB, the most abundant of the numerous fumonisin analogues, was classified by the IARC as a Group 2B carcinogen (possibly carcinogenic in humans) [6].

Although DON is the least toxic type of trichothecene, it can cause significant harmful effects in animals and humans [7]. DON causes a broad variety of toxic effects in animals, and its toxicity is well recognized in mammals. The main effect at the cellular level is the inhibition of protein synthesis via binding to the ribosomal subunit [8]. Chronic oral exposure induces anorexia, decreased weight gain, reduction in feed conversion, gastrointestinal hemorrhaging, inflammation and immune system alterations.

ZEA competes with the naturally produced hormone estradiol-17β for binding sites (estradiol receptors) in various organs in the body of both genders. ZEA can obstruct normal steroid hormone (estradiol, testosterone, progesterone) synthesis in the ovaries and testicles of livestock.

ZEA is found, especially, as a contaminant in corn. ZEA, which is produced mainly by F. graminearum and F. culmorum and commonly co-occurs with DON and its derivatives. It is among one of the most frequently encountered mycotoxins in grain from FHB (fusarium head blight)-diseased small-grain cereals throughout the Mediterranean countries. It may co-occur with DON in grains such as wheat, barley, oats and corn and fumonisins in corn [9]. Generally, DON is found in higher doses than ZEA when this occurs [10]. Most studies indicate that in artificial environment high moisture favours the production of both classes of mycotoxins, but the optimum temperatures for trichothecene and ZEA production in Fusarium-infected grain appears to be specific to the substrate, species and individual metabolites. The accumulated data revealed definite geographical differences in the level and frequency of DON, NIV and ZEA in wheat and barley. Streit et al. [11] reviewed mycotoxin co-occurrences in animal feed in Europe since 2004. Since Fusarium species are the most frequent fungal pathogens on field crops, it was not surprising that B-trichothecenes (DON), ZEA, and FBs were the major co-contaminants.

Reproductive efficiency is a very important economic factor in animal production. Exposure to several Fusarium mycotoxins have been linked to reproductive disorders as it was reviewed by Cortinovis et al [12]. The effect of these toxins on reproduction has been widely studied in female animals, while knowledge about their effect on reproduction processes in males is limited. As far as it is known they can affect spermatogenesis, and sperm quality, Leydig cell function (testosterone production), gonadotropin secretion, and fertility [13-18].

Humans and animals are generally exposed to multiple mycotoxins in parallel, as some mycotoxins typically co-occur in cereals. However, toxicological data, risk assessments are based on, are provided by studies in which only the individual effects of certain toxins are investigated. The simultaneous exposure of animals to more than one toxin is of concern and requires more study [19]. Synergistic effects may explain why animals sometimes respond negatively to mycotoxin levels much lower than those reported in scientific studies as able to cause mycotoxicoses.

In a survey conducted over a period of 4.5 years in countries of Southern Europe (Portugal, Spain, Italy, Greece and Cyprus) the Fusarium mycotoxins were found to be the major contaminants (fumonisins, type B trichothecenes and ZEA) of feed material and compounded feed samples [20]. There are several studies about mycotoxins’ interaction (mainly in vitro), but only a few report the combined effects of three specific Fusarium mycotoxins which are more likely to co-occur; i.e. FB, DON and ZEA. Although there are studies about the three aforementioned mycotoxins very few focus on tertiary mixtures [21-24]. In rabbit bucks, ZEN impairs spermatogenesis and decreases libido, although only at high doses (117.3 mg/kg feed) [25]. Pregnant rabbit does fed on a DON-contaminated diet (0.3 and 0.6 mg/kg) showed marked bodyweight loss, but teratogenic effect was not proven [26]. FB1 was found to be nephron- and hepatotoxic in rabbits [27], and it has also been shown to negatively influence haematopoiesis by impairing bone marrow function [28]. Lung oedema was found only in a small number of rabbits fed a FB1-contaminated diet [29]. The teratogenic effect of FB1 was also described using an oral dose of 300 mg/d for 14 d [18] but a lack of embryotoxicity was found at lower (0.1, 0.5 or 1.00 mg/d) oral doses [30].

Grenier and Oswald [31] performed a meta-analysis of published raw data on mycotoxin interactions in vivo which varies according to the animal species, the dose of toxins, the length of exposure, but also the parameters measured; and classified the interaction into the following categories: synergistic, additive, less than additive, and antagonistic. The authors also differentiated between three types of synergistic effects and two types of antagonisms. Such characterization of mycotoxin interactions is helpful in experimental designs and interpretations of combined toxicity outcomes and should be included into further investigations on mycotoxin interactions.

The aim of this experiment was to study the dietary, low-dose of FB, DON and ZEA mycotoxins’ individual and combined effects on breeding rabbit bucks, in particular with regards to reproductive toxicity.

Materials and Methods

Experimental animals, housing and feeding

The experimental animals were Pannon White rabbit breeding bucks (24 weeks of age, 4.0 ± 0.5 kg mean bodyweight, n=60). They were individually housed in wire mesh cages (42 × 50 cm) in a closed building, with 16 light h/day. Average temperature ranged from 16°C to 18°C and the farm had overpressure ventilation.

The animals received a commercial diet containing 10.3 MJ digestible energy/kg, 15.5% crude protein, 4.0% crude fat and 14.7% crude fibre for a total of 65 feeding days. The feedstuffs provided were available ad libitum, and the rabbits also had free access to drinking water.

Toxin production

Fusarium verticillioides (for FB1) and Fusarium graminearum (for DON and ZEA) (NRRL 20960 [MRC 826] and NRRL 5883, respectively) fungal culture (7 days old) was grown on 0.5 strength potato dextrose agar (PDA; Chemika-Biochemica, Basle, Switzerland). Agar discs (5 mm) were prepared with cork borer (Boekel Scientifica, Pennsylvania, USA), which were then stored at 10°C in darkness in test tubes containing sterile distilled water (10 discs/10 ml).

For toxin production, maize (40 g) was soaked in distilled water (40 ml) at room temperature for 1 hour in Erlenmeyer flasks (500 ml), closed with cotton wool plugs. This was followed by the addition of the inoculated agar discs (10 agar discs per flask) to the two-times autoclaved (20 min.) matrix. The cultures were then stored and incubated at 24°C (FB1), 28°C (DON) and 18°C (ZEA) for 3 weeks, respectively. The flasks were shaken twice every day during the first week of incubation. When the incubation time was complete the fungus-infected cereal was dried at room temperature and ground.

The homogenized fungal cultures contained FB, DON and ZEA at concentrations of 3300, 2010 and 1298 mg/kg, respectively. The European Commission has made recommendations (2006/576/EC) regarding the maximum level of several mycotoxins in complete diets (European Commission, 2006) and introduced regulations (2003/100/EC) regarding aflatoxins (European Commission, 2003). However, these only apply to certain cases, particularly with regard to rabbit feed. Thus in this study doses were pre-determined according the EU limits (based on the European Commission Recommendation 2006/576/EC and the European Commission Directive 2003/100/EC) in finished feed for young pig as the most sensitive animal towards these fusarium mycotoxins among livestock. Fungal cultures were mixed into the feed of experimental animals, based on the presented dose in Table 1.

Group Mycotoxin concentration (mg/kg) of the feed n
C (control) 0 15
F (FB1) 5 15
DZ (DON+ZEA) 1+0.25 15
FDZ (FB1+DON+ZEA) 5+1+0.25 15

Table 1: Experimental groups.

LC-MS analysis

LC-MS analysis was performed by a Shimadzu Prominence UFLC separation system equipped with a LC-MS-2020 single quadrupole (ultra-fast) liquid chromatograph mass spectrometer (Shimadzu, Kyoto, Japan) comprised a vacuum degasser, a binary pump (20AD), a column oven (CTO 20A) autosampler (SIL 20ACHT), and mass analyser (MS 2020) with both atmospheric-pressure chemical ionization (APCI) and electrospray ionization (ESI) systems. Optimized mass spectra were obtained with an interface voltage of 4.5 kV, a detector voltage of 1.05 kV in negative mode, 1.25 kV in positive mode. Heat block temperature was 200°C and a desolvation gas temperature was 200°C. For nebulizing and drying gas, nitrogen was used (1.5 L/min and 15 L/min flow rate, respectively). Chromatographic separation was performed at 50°C and achieved on an RP-18 (2.1 × 100 mm, 2.6 µm, Kinetex™ Phenomenex USA) stationary phase applying gradient elution 0.3 mL/min eluent total flow rate for mycotoxins and 0.4 mL/min for silymarin flavonoids, with A: 0.1% AcOH and B: 0.1% AcOH in methanol as eluent. With optimum method performance characteristics analytes were quantified using external calibration.

Rabbit-feed samples were milled and extracted using 1% AcOH containing 75:25=MeOH:H2O (V/V) for F-2 CH3CN:water 1:1 (V/V) for FB1 and water for DON and ZEA as extraction solvent. The extracts were shaken at room temperature for an hour then decanted and the supernatant was collected. 1 mL of clean water extract was applied to the immunoaffinity column (IAC; Vicam, DON test) which contains specific antibodies for DON for purification. The IAC was washed with 5 mL water, and DON was slowly eluted in 2 mL methanol. ZEA and FB1 were measured by dilute and shot method. Romer Mix 4 (containing trichotecenes+zearalenon at 10 mg/L) and Romer MIX 3 (containing FB1-2 at 50 mg/L) primary stock solution used as reference. 1 µL of each samples were analysed with a gradient: (0 min) 5% B, (3 min) 60% B, (8 min) 100% B, followed by a holding-time of 3 min at 100% eluent B and 3 min column re-equilibration at eluent A pumped at a flow rate of 0.3 mL/min. DON is detected as [M+AcO]– at m/z=355, ZEA at m/z=317 as [M-H]–, FB1 at m/z=722[M+H]+. The limit of detection (LOD) for FB, ZEA and DON was 3.0, 5.0 and 5.0 μg/kg, while the limit of quantification was (LOQ) 10, 1.0 and 2.0 μg/kg, respectively.

Experimental design

Bucks were divided into 4 experimental groups. One group of the experimental animals served as control, while into the feed of the other three groups fungal culture was mixed in a pre-defined concentration (Table 1).

The daily feed intake was registered by measuring back the left-over feed amount (in the first 3 weeks daily, and weekly thereafter), while bodyweight was recorded once every week.

The health status of the animals was observed throughout the experiment, morbidity and mortality was logged daily.

On days 30 and 60 blood and sperm was sampled (n=15/group), and a gonadotropin-releasing hormone (GnRH) test was performed (blood samplings related to GnRH test: n=6/group). Blood was sampled from the marginal ear vein, while sperm was collected after a training period into artificial vaginas and seminal plasma was separated with centrifugation.

At the end of the study (on day 65) animals were exsanguinated after stunning. The weight of liver, kidneys, testicles and spleen was measured and macroscopic changes were analyzed and recorded. After dissection (n=15/group) samples were taken for histopathological analysis; the testis, liver and kidney was fixed in 10% neutrally buffered formalin. For the analysis of antioxidant status 2-2 g samples were taken from the liver.

The experimental protocol was authorized by the Food Chain Safety and Animal Health Directorate of the Somogy County Agricultural Office, under allowance number SOI/31/1679-11/2014.

Clinical chemical parameters

The plasma total protein (TP), albumin (ALB), globulin (GLOB), total cholesterol (tCHOL), triglyceride (TG), glucose (GLU), fructosamine (FA), creatinine (CREA) and bilirubin (BIL) concentrations, and the activity of aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT), lactate dehydrogenase (LDH) and alkaline phosphatase (ALKP) were determined in a veterinary laboratory (Vet-med Laboratory, Budapest, Hungary), using Roche Hitachi 912 Chemistry Analyzer (Hitachi, Tokyo, Japan) with commercial diagnostic kits (Diagnosticum LTD., Budapest, Hungary).

Antioxidant status

For the determination of lipid peroxidation, the samples of blood plasma, red blood cell (1:9) hemolysate (RBCH) and liver were stored at -70°C until analysis. Lipid peroxidation was determined by the quantification of malondialdehyde (MDA) levels with 2-thiobarbituric acid method in blood plasma and RBCH [32] and liver homogenate [33], and determination of conjugated dienes (CD) and trienes (CT) according to the AOAC [34] method in the liver. Among the components of the antioxidant system some parameters of the glutathione redox system was determined in blood plasma, RBCH and liver. The amount of reduced glutathione (GSH) measured by the method of Sedlak and Lindsay [35] and the activity of glutathione peroxidase (GPx) according to Lawrence and Burk [36].

GnRH test and determination of testosterone concentration

Experimental and control bucks were treated i.m. with 0.2 ml GnRH analogue (Receptal; Intervet, Boxmeer, The Netherlands) for the analysis of the toxin effect on the Leydig cell function. The levels of testosterone hormone were determined from blood samples taken just prior to GnRH analogue injection (0 min) and thereafter in the subsequent 2 hours in every 25 minutes (a total of 6 blood samplings).

The testosterone concentration was determined with a direct 3H-radioimmunoassay method [37] adopted and validated for rodents’ (chinchilla rabbit and Angora rabbit) plasma as described previously [38].

Spermatology

Spermatological analyses covered the following parameters: pH, sperm cell concentration (improved Neubauer cell counting chamber), motility, morphology (native and stained) and acrosomal integrity (Spermac™ staining, Beernem, Belgium) of the spermatozoa. Motility was evaluated with a computer assisted sperm analyzer (Medealab™ CASA System, Erlangen, Germany). Moreover, vital test, hyposmotic and peroxidase tests were carried out. A minimum number of 200 spermatozoa were examined for morphology and 500 for motility evaluation [39].

Comet assay

For comet assay sperm was sampled on day 60 of the experiment (n=15/group). The method was adapted from human spermium examination protocols [40,41], with the following modifications. The semen was washed three times in PBS and re-suspended in PBS to a final of 106 cell/ml number. Onto the microscope slides pre-coated with 1% NMP agarose 10 µl cell suspensions and 75 µl 1% LMP agarose were loaded. The decondensation was performed in two steps, first the slides were soaked for 1 hour at 4°C in a lysis buffer with dithiothreitol, and second the slides were soaked for 1 hour at 37°C in a lysis buffer with proteinase K. After lysis the slides were washed in sterile redistilled water to eliminate the salt adhered to the gel. Electrophoresis was performed at 300 mA and 25 V for 30 minutes at 4°C. After washing and drying the slides were stained with ethidium bromide. All chemicals used in this study were obtained from Sigma-Aldrich Ltd. (Budapest, Hungary). Specialized chemicals used were: Histopaque-1077 and RPMI-1640 medium (Sigma-Aldrich Ltd., Budapest, Hungary).

The fluorescence images were generated using an Alpha-Optika B-600TiFL fluorescence microscope (Optika Microscopes, Bergamo, Italy). Scoring was carried out according to Singh et al. [42] and Collins et al. [43], in which comets are classified into scores of ‘0’, ‘1’,‘2’, ‘3’ and ‘4’ according to DNA damage and head/tail migration. Each single comet was scored visually and assigned into an arbitrary unit from 0 to 4, depending on the relative intensity of DNA fluorescence in the tail; 800 cells/group were counted.

Histopathological analysis

After registering the macroscopic pathological signs on the internal and external organs, testicles, liver, kidneys and spleen were stored in 10% neutrally buffered formalin and were embedded into paraffin. For light microscopic analysis microtome slides of 5 micrometer were prepared and stained with hematoxylin-eosin.

The histopathological analysis was performed according to the Act/2011 (03.30) of the Hungarian Ministry of Agriculture and Rural Development and was in accordance with the ethical guidelines of the OECD Good Laboratory Practice for Chemicals [44].

Statistical analyses

Statistical analyses were performed using IBM SPSS 20.0 [45] software. Data processing and the mathematical-statistical calculations were performed using the compare means (independent-samples-t-test, oneway ANOVA with Tukey post-hoc test), correlate and descriptive statistics modules. In case of comet assay crosstabs options were used for chi-square test.

Results

Feed consumption and body weight

The feed intake of the rabbit bucks was not different among the groups. No significant difference in body weight among groups was detected at any of the 12 time points (data not shown), average body weight of the groups was between 4252 and 4442 g by the end of the experiment.

Clinico-chemical parameters

No inter-group differences (P ≥ 0.05) were found for ALB, TG, GLU, FA, CREA and BIL concentrations, and AST, ALT, GGT, LDH and ALKP activities.

Differences in TP, GLOB and tCHOL at the two sampling dates (day 30 and 60) are summarized in Table 2. In spite of the slight significant differences between treatments all data were within the reference ranges, which are 54-75 g/L for TP, 15-27 g/L for GLOB and 0.3-3.00 mmol/L for tCHOL [46]. Only GLOB concentration exceeded the upper limit of 27 g/L in the C and FDZ groups on day 30 (29 and 28 g/L, respectively).

    Day 30 Day 60
    mean SD mean SD
TP (g/L) C 69.85b 5.85 62.16a 3.02
F 68.57b 1.72 64.73ab 3.97
DZ 61.33a 4.28 65.81ab 7.34
FDZ 69.57b 1.92 67.74b 3.40
GLOB (g/L) C 28.65b 3.54 22.38a 2.23
F 27.13b 1.61 22.96a 2.09
DZ 20.23a 3.39 24.10ab 5.95
FDZ 27.98b 1.63 26.67b 3.12
tCHOL (mmol/L) C 1.45ab 0.12 1.51b 0.35
F 1.62b 0.19 1.21a 0.09
DZ 1.33a 0.16 1.29a 0.14
FDZ 1.58ab 0.17 1.44ab 0.14
a,bnumbers with different superscripts indicate significant differences (P≤0.05) between groups (C, F, DZ and FDZ).

Table 2: Clinico-chemical parameters of rabbits on day 30 and 60.

Antioxidant status

On day 30 only MDA concentration in the RBCH showed significant difference between treatments F (35.7 ± 4.5 mmol/l) and DZ (34.2 ± 1.6 mmol/l), as compared to C (43.4 ± 3.5 mmol/l) and FDZ (43.7 ± 7.6 mmol/l), but other antioxidant parameters did not change significantly (data not shown).

Parameters showing significant treatment effect on day 60 are summarised in Table 3. At day 60, DZ treatment resulted in significantly increased GPx activity in the red blood cells and MDA formation both in RBCH and plasma, while less GSH concentration in the blood plasma. As a result of peroxidation of dienoic and trienoic fatty acids, the concentration of conjugated dienes and trienes increased as a result of the DZ exposure, as compared to the combined effect of three mycotoxins (FDZ).

Parameter Groups
C F DZ FDZ
GPx-RBCH1 2.7 ± 1.0a 3.5 ± 1.2a 4.9 ± 1.1b 3.1 ± 0.8a
MDA-RBCH2 30.3 ± 3.0ab 28.3 ± 3.2a 33.9 ± 7.5b 28.8 ± 3.2a
GSH-plasma3 2.2 ± 0.2a 2.2 ± 0.5a 2.0 ± 0.2a 2.6 ± 0.5b
MDA-plasma2 18.9 ± 2.8a 18.7 ± 3.0a 24.5 ± 8.0b 16.8 ± 2.9a
CD liver4 0.51 ± 0.03ab 0.51 ± 0.02ab 0.53 ± 0.05b 0.48 ± 0.05a
CT liver4 0.19 ± 0.01a 0.20 ± 0.01ab 0.21 ± 0.04b 0.18 ± 0.01a
1U/g protein, 2μmol/ml, 3μmol/g protein, 4Abs 232 nm a,bnumbers with different superscripts indicate significant differences (P≤0.05) between groups (C, F, DZ and FDZ)

Table 3: Antioxidant parameters measured on day 60.

Testosterone concentration

The testosterone concentration was significantly different due to mycotoxin exposure only at day 60 at the sampling minutes of 75, 90 and 115 (Figure 1). Figure 1 clearly demonstrates that Leydig cells of the animals intoxicated with the three mycotoxins in combination (FDZ) for a total of 60 days synthesize significantly less testosterone as a response to exogenous GnRH. In the F group blood testosterone level was slightly lowered, while the two toxin combination (DZ) induced a more expressed decline in the concentration, although the differences were not statistically significant.

clinical-toxicology-Gonadotropin-releasing

Figure 1: Gonadotropin-releasing hormone (GnRH; mean and standard deviation) induced testosterone production at day 60. a,bnumbers with different superscripts indicate significant differences (P ≤ 0.05) between groups (C, F, DZ and FDZ).

Spermatological analysis

No effect of toxin treatment was detected on semen pH (ranging from 6.4 to 8.2 in each group), quantity of the semen (on average 1 ml in each group) and concentration of spermatozoa (ranging from 2.4 to 2.6 × 107/ml). Comparing the sperm motility between groups, significant effect could also not be proven. The ratio of spermatozoa showing progressive forward motility was around 80% at the beginning of the experiment and decreased from 80 ± 1.7% to 67 ± 4% in the semen of FDZ animals showing significant difference when compared to the other three treatment groups. Between the normal morphology of the spermatozoa of the DZ and C animals, statistically significant differences were found (Table 4).

Group Spermatozoa with normal morphology
C 80.2 ± 11.2b
F 76.0 ± 9.0ab
DZ 66.3 ± 23.7a
FDZ 68.9 ± 14.1ab
a,bdifferent superscripts in the column indicate significant between-group differences (P≤0.05)

Table 4: The ratio of spermatozoa with normal morphology in the semen after 60 days of mycotoxin exposure (%, mean ± SD).

The most frequent morphological abnormalities were: abnormality of the tail, retention of cytoplasmic drop, absence of the acrosome and altered head. A cell was considered altered if at least one defect was present.

Comet assay

All treatments caused DNA damage and 98.6, 91.6 and 91.8% of the treated cells could be categorized as having 1 to 3 scores in the F, DZ and FDZ group, respectively (Figure 2). Cells with 0 values showed the highest prevalence in group C, while cells with the slightest damage (score 1) were dominant in all toxin treated groups. FB1 resulted significantly less 0 comets (intact cells) compared to the other treatments.

clinical-toxicology-comet-scores

Figure 2: Number of cells with the respective comet values (values of comet scores: 0 to 4). a,bcolumns with different superscripts indicate significant differences (P<0.0001).

Cells, with score 1 (Figure 3a) occurred in nearly similar proportions in case of the three toxin treatments. The comet value 2 (Figure 3b) showed the highest prevalence in samples of F and DZ animals, FDZ treatment resulted in significantly less cells of this type. The number of cells with score 3 (Figure 3c) occurred only in 0.6% of the categorized cells. No cells were found with a comet assay score of 4 in the experiment.

clinical-toxicology-visual-scoring

Figure 3: Comet assay (DNA damage) scoring: images of comets from bucks’ spermatozoa stained with EtBr. Classes 0 (a), 1 (b) and 2 (c) were used for visual scoring.

Histopathology

No sign of mycotoxicosis was detected. In animals consumed FB1, DZ and FDZ contaminated diet the intensity of spermatogensis decreased by 43, 31 and 64%, respectively (Table 5), which was reflected by the lack of differentiated spermatozoa, thinning of the germinal epithelium, the appearance of multinuclear giant cells indicative of the disturbance of meiosis and mitosis of the germinal epithelial cells and in some cases the lack of spermatogonia.

Group NO Number of animals showing alteration
1 2 3 All together %
C 15 0 0 0 0 0
F 8 6 0 0 6 43
DZ 9 4 0 0 4 31
FDZ 5 7 1 1 9 64
NO=number of animals without alteration, 1=slight/small area/low occurrence frequency, 2=intermediate severity/intermediate area/intermediate occurrence frequency, 3=marked/extensive/high occurrence frequency

Table 5: Decreased intensity of spermatogenesis as revealed by the histopathology of the testicles.

These histological findings were observed to different severities in the seminiferous tubules. In case of slight alteration (grade 1) disturbance of sperm cell formation was observed in 20-30% of the seminiferous tubules, while in case of grade 2 and 3, 30-70 or 100 % of the tubules, respectively, showed decreased spermatogenesis. Decreased spermatogenesis was manifested in lack of well differentiated sperm cells and less primary, secondary spermatocytes and spermatids, and thinning of the seminiferous epithelium as a consequence (grade 1). In case of intermediate severity (grade 2) these alterations were observed in higher ratio of the tubules, the disturbance of meiosis and mitosis was indicated by the appearance of multinuclear cells. Cases, when all tubules were attacked and moreover, in some tubules even the initial spermiogenetic cells, spermatogonia were also absent, were classified as marked damage (grade 3) (Figures 4 and 5).

clinical-toxicology-Active-spermatogenesis

Figure 4: Active spermatogenesis in a healthy control animal’s seminiferous tubules. The cells at different differentiation levels in the epithelium germinativum (spermatogonia, stage I and II spermatocytes, spermatides and mature spermia (↑) are as well visible. Haematoxylin-eosin staining, 400X.

clinical-toxicology-Haematoxylin-eosin

Figure 5: A part of the seminiferous tubule after FDZ treatment. The marked lack of germinal epithelialcells is visible. Haematoxylin-eosin staining, 400X.

In the testicles of the toxin treated animals the cytomorphology and the proportion of the Leydig interstitial cells was not significantly different from the controls. In the Malpighian bodies of the spleen of every DZ and FDZ animal, slight lymphocyte depletion (slight thinning of the T and B dependent zones of the Malpighian tubules) was observed. However, the cytomorphology of lymphoblast and lymphocyte cells was not different from the control. In the livers and kidneys no detectable alteration attributable to mycotoxins was found.

Organ masses

Among all organs which were measured for weight (testicles, liver, kidneys, spleen) only the weight of the spleen was significantly different between groups. It was the most (1.84 ± 0.49b) in DZ animals, and least (1.46 ± 0.38a) in C, while 1.65 ± 0.30ab and 1.65 ± 0.37ab in F and FDZ, respectively. However the relative organ mass values (data not shown) of the different groups were not significantly different.

Discussion

The present study intended to determine the negative effect of a combined, 65-day oral Fusarium mycotoxin administration on the reproductive traits of 24-week old breeding Pannon White rabbit bucks. The low toxin exposure levels were chosen taking the limit values given by the 2006/576/EC recommendation into consideration. The in feed concentrations of FB, DON and ZEA (5, 1 and 0.25 mg/kg feed, respectively) revealed 169-193 μg FB1/kg BW, 33.7-38.7 μg DON/kg BW, and 8.5-9.7 μg ZEA/kg bw exposure. Results indicate that a prolonged low-dose mycotoxin exposure may adversely affect male reproduction as it has been previously demonstrated in case of T-2 toxin. The combined toxin effect indicates highly complex interactions among the mycotoxins, appearing differently in the divergent physiological processes. Among the mycotoxins studied here additive, synergistic, as well as antagonistic effects were detected (Table 6).

Parameter examined Type of interaction
feed intake antagonism (not significant)
lipid peroxidation (MDA, GPx, CD, CT) antagonism
testosterone production synergism
spermatogenesis by histology additive
sperm cells with normal morphology less than additive
weight of the spleen antagonism
genotoxicity antagonism

Table 6: Summary of the interactions between FB, ZEA and DON experienced in the experiment (classified according to Grenier and Oswald, 2011).

Feed consumption and body weight

The only observation to mention concerning feed consumption and body weight (BW) was that the mean feed intake (FI) of DZ and FDZ animals was somewhat lower, as compared to F and C (significance was not proven). This result may mostly be attributed to DON, since this is the most well-known characteristic effect of trichothecene mycotoxins [47]. In the study of Hewit et al. [48] using naturally contaminated (4.2 mg/kg DON) corn for Fusarium mycotoxin exposure in growing rabbits, no feed refusal was observed and the toxin treated animals had slightly increased BW gain, suggesting that rabbits are less sensitive to this mycotoxin. Deep discussion of a mere tendency is void, but observing data at weeks 4 and 5 again indicate the “likewise positive effect” of low-dose FB, since the lowest FI was found for DZ, but FDZ and F were less suppressed. This is supported by the result on non-human primates of Gelderblom et al. [49], where long-term feeding of FB1 alone had no negative effect on FI and BW, even at higher doses. In that study apparent no-effect threshold in terms of kidney and liver damage was estimated at 8.21 and 13.25 mg total fumonisin/kg diet, being about the double exposure, as compared to the present study. Also no effect on final live weight and weight gain was observed when male rabbits were fed diets containing 12.3 and 24.5 mg/kg FB1 for 5 weeks [50]. Summarized, our study indicates that DON provides its characteristic feed-intake lowering effect in a slight, but insignificant rate at the dose according to Commission Recommendation (2006/576/EC), while fumonisin shows no determinative effect on BW and FI at these concentrations, in breeding rabbit bucks.

Clinical chemical parameters and antioxidant status

The health status of the animals can be monitored by regular determination of certain clinical chemical parameters. Despite that the liver and the kidney is considered to be the primary target organs of many mycotoxins (like trichothecenes and FB1) in all species examined, gross hepatic lesions and disorder was not induced by any of the treatments, as underscored by the unaltered ALT, AST, GGT and CREA values, the identical liver and kidney mass values and the lack of histopathological alterations in all groups. These organs were likewise tolerant towards the exposure of low dose of these mycotoxins, without providing degenerative processes. Sprando et al. [14] described an analogous observation, namely few lesions of various tissues in control and treated animals, either incidental or related to experimental techniques (gavage) and unrelated to the test substance. The effects of low (10 mg/kgBW) and high (100 mg/kgBW) oral doses of purified ZEN were studied by Čonková et al [47] in rabbits. Low ZEN doses resulted in a significant increase in ALP activity, while high ZEN doses showed significant increases of AST, ALT, AP, GGT, and LDH activity, indicating possible liver toxicity due to the chronic effects of the toxin.

The alteration found in the TP was attributed to the concentration changes of globulin, since albumin concentration was constant. According to Kim et al. [51], DON stress lowers immunoglobulin expression in mice, but ZEA was found to have no influence on it. This could be the reason of the slight temporary decrease in GLOB and TP of DZ animals at day 30. The results at day 60 are difficult to explain, because difference between groups was rather attributable to the lowered GLOB level in control animals than toxin treatments. However fumonisin treatment decreased GLOB concentration at day 60, compared to day 30. Results of the FDZ group on day 60 are in accordance with those of Tessari et al. [52], describing that FB1 (added to an aflatoxin B1 containing diet) has primary additive effect on the immunological response in birds. It is interesting to highlight that FB1 alone and in higher doses (8 mg/kg feed) had a sex-dependent immunosuppressive effect, male pigs being more prone [53]. The FDZ treatment evokes the question, whether the low dose of FB1 or the interaction between these toxins augmented the immune response. Some kind of anomaly in the immune system was supported by histology, in that in both DZ and FDZ animals, slight lymphocyte depletion (slight thinning of the T and B dependent zones of the Malpighian tubules) was observed in the spleen, without any change in the cytomorphology of lymphoblasts and lymphocytes. In the experiment Sprando et al. [14] similar finding was described when rats orally exposed with 5 mg/kg DON.

The pathophysiological significance of the changes in tCHOL level, which was around 5-20%, is questionable, taking the broad physiological ranges (0.3-3.00 mmol/L) into consideration.

The increased GPx activity in the red blood cells of DZ animals by day 60 reflects more lipid hydroperoxide production, as the role of the enzyme is to reduce lipid hydroperoxides to their corresponding alcohols and this way to reduce free hydrogen peroxide to water. This is supported by the highest MDA concentrations both in the red blood cells and the blood plasma, referring to augmented in vivo lipid peroxidation [54], and also by the decreased GSH, as co-substrate of GPx, level in plasma. One of the mechanisms by which DON exerts its toxicity is inducing oxidative stress within the cells [55]. ZEA is also known to induce generation of reactive oxygen species (ROS) and consequently lipidperoxidation, while fumonisin was characterised as moderate oxidant mycotoxin [56]. Our results show general agreement with those of Minervini et al. [57], reporting that in vitro FB1 dose at the level of the EC Regulation is not inducing oxidative stress in intestinal cells. On the other hand FB1 has proven to induce ROS production in broiler chicks, but only at extreme high (100 mg/kg feed) dose [58]. Our findings suggest that FB1 in low dose may act antagonistically towards Z and D, which has not been reported yet. Though there is no literature on the mitigating property of FB1 on lipid peroxidation. We thus summarize DZ combination was the most harmful in terms of initial (CD and CT) and terminal (MDA) phases of lipid peroxidation after long-term exposure (60 days); meanwhile FB1 at the EC Regulation level did not cause significant oxidative stress, even more relieved those effects triggered by ZEA and DON.

Gn-RH induced testosterone production

The testosterone concentration was not significantly influenced by one (F) and two toxins (DZ), but the FDZ treatment dramatically lowered its synthesis and/or release from the Leydig cells by the end of the sampling intervals (minutes 75, 90 and 115), suggesting synergistic effect. As far as the authors are aware, there exists only a single study on male reproduction traits as affected by orally administered DON, which was performed in rats [14]. In this study 0.5, 1, 2.5 and 5 mg/kg DON was applied, and the authors found a dose-dependent down-regulation of testosterone synthesis after 28 days of exposure. Trichothecene toxins may decrease the testosterone production of the Leydig cells by inhibiting early steps of the steroidogenic pathway, i.e. the conversion of pregnenolone to progesterone [25], as it was also observed in our previous study in case of T-2 exposure [38]. These Fusarium toxins may act indirectly as well, on the pituitary gland, or affect Sertoli cell inhibin production, as shown in case of DON [14]. In present study a direct effect of the mycotoxin on Leydig cells was not supported by the histopathological findings.

In fact not DON, but zearalenone is the most potent factor, directly inhibiting testosterone biosynthesis in the Leydig cells, adding that this inhibition is concentration- and time-dependent, happening via changing the nuclear estrogen receptor signaling [59], however the exact molecular mechanism is not known yet. For FB1 direct toxicological studies are lacking, but according to Lu et al. [60], elevated ceramide levels are directly apoptotic towards Leydig cells, which is inhibited by FB, a ceramide-synthase blocking substance. Thus FB1 had no deleterious effect on the rat testicular Leydig cell function in vitro. In summary the main, testosterone synthesis blocking effect in our study was primarily attributed to ZEA. Some kind of synergistic effect occurred between FB1 and DON+ZEA. Taking the common cellular modes of actions of these toxins into consideration, this might be explained with respect to the DNA-synthesis inhibiting effect. FB1 affected the DNA synthesis together with ZEA in a more than additive, rather synergistic way in a DNA inhibition assay, although this effect was not fully confirmed when it was further analysed in a second stage [61].

Spermatology, histopathology and comet assay

Toxin effect and interaction between the Fusarium toxins applied, manifested in lower sperm motility (FDZ), less spermatozoa with normal morphology (DZ), additive effect in the decreased intensity of spermatogenesis as shown by the histology of the testes, while antagonism concerning genotoxicity measured by comet assay.

Concerning the individual effects of these mycotoxins on sperm motility, only a few data are available. FB1 fed with male rabbits at 7.5 and 10.0 mg/kg dietary levels decreased sperm motility by approximately 18%, the possible explanation was the inhibition of cyclic 3’5’AMP activity and calcium ion transport, or, the decreased formation of the acidic epididymidal glycoprotein synthesis required to maintain motility [50]. Changes, after increasing oral exposure of DON were not consequent, and considered to be random [14], while in an in vitro study no significant effect of DON on motility was found using rabbit spermatozoa (Medvedová et al., 2012). Boeira et al. [62] reported compromised spermatozoa motility after feeding mice with ZEA, while 12 mg daily ZEA exposure for 8 weeks did not alter sperm motility in adult rams [63]. Further data from in vitro studies support no effect of ZEA on boar spermatozoa [64], while on the contrary Rajkovic et al. [65] and Tsakmakidis et al. [66] described impairment of progressive motility. Interestingly in an in vitro study with stallion sperm ZEA caused hyperactivation in motility [67].

As the cellular level effects by which the above described mycotoxins may adversely influence motility are not well described yet, the combined effect is also difficult to explain. Decreased sperm motility caused by the exposure of the three mycotoxins (FDZ) by day 60 might be attributable also to the decreased testosterone concentration as a result of synergistic effect between FB, DON and ZEA observed also by day 60. The male reproductive organs are strongly androgen-dependent in respect of structure, as well as function. Testosterone supports spermatogenesis, sperm maturation, seminal plasma production and sexual functions [68]. So the decreased testosterone secretion may give an explanation also for the more morphologically abnormal cells, and the decreased intensity of spermatogenesis shown by histology. However, direct effect of the particular toxins is also presumable. Interestingly FB1 had only a slight adverse effect on the cell morphology and spermatogenesis. The characteristic toxic effect of FB1 is the inhibition of the sphingolipid synthesis, thus causing cellular ceramide depletion and elevation of shinganine, which is cytotoxic, and has growth inhibitory and pre-apoptotic effects [69]. This results in disturbances of cell growth, differentiation and morphology. In rabbits orally administered FB1 caused similar morphological and structural abnormalities in sperm morphology and spermatogenesis, and 7.5 mg/kg in the diet for FB1 as LOAEL (lowest observed adverse effect level) was suggested [50]. In our study even lower dose, 5 mg/kg had exerted adverse effect; however sensitivity to toxic substance is influenced by several factors. Compared to F group, DZ was similarly compromised. In case F and DZ, disturbance of sperm cell formation was observed only in 20-30% of the seminiferous tubules, however the combined treatment of DON and ZEA resulted in more cells of abnormal morphology. After exposure of the three toxin (FDZ) in 1 animal 30-70, while in another 100% of the seminiferous tubules were concerned, indicating additive effect. The effect of ZEA on spermatogenesis is more thoroughly studied compared to FB1 and DON. In orally exposed mice Zatecka et al. [70] detected decreased sperm concentration, and increased number of morphologically abnormal and also apoptotic spermatozoa. No pathological patterns in the seminiferous tubules were observed and the spermatogenesis was not interrupted, only the number of spermatogonia and spermatocytes were lower (not significantly) in exposed animals (25 μg/kg BW). Kim et al. [71] highlighted that apoptosis is the main effect by which ZEA causes germ cell depletion and atrophy of the testes. Germ cell apoptosis and thus impaired spermatogenesis was described also by Cho et al [72].

The in vitro exposure to ZEA induced cytotoxicity in boar sperm cells [64]. DON in 2.5 and 5 mg/kg concentration caused germ cell degeneration, decreased sperm cell release and abnormal cell development [14]. As the authors reported, DON may inhibit protein, RNA and DNA synthesis, and be cytotoxic to certain cells, which could cause the harmful effect in the most sensitive pre-leptotene spermatocytes.

Genotoxicity of Fusarium mycotoxins on sperm cells has been reported previously in case of FB1 [17] and ZEA [17,64,66,67], using sperm chromatin structure assay (SCSA), while no data on genotoxicity of DON related to sperm cells are available. A recent study showed that DON decreased cell viability, caused damage to the membrane, the chromosomes and the DNA in human lymphocytes, and potentially induced genotoxicity by the depletion of the antioxidant capacity. On the other hand according to Golli-Bennour and Bacha [56] DON is a non-oxidant mycotoxin, and its genotoxicity is the result of a direct effect on DNA fragmentation and caspade-dependent apoptosis.

In case of FB, Minervini et al. [17] found high degree of individual variability, when chromatin structure stability was checked in vitro, using equine spermatozoa. Interestingly no increase in ROS production was observed even in those cases when genotoxicity was detected, so the damage was attributed to mitochondrial dysfunction related to the altered ceramide metabolism. This mode of action might influence motility as well, knowing that spermatozoa are used as biosensors when testing mitochondrial toxicity, because of the strong influence of mitochondrial function on motility [17].

When we examined the single and combined genotoxic effect of FB1 and DON+ZEA on rabbit sperm cells, an antagonistic effect was observed, highly in accordance with the antioxidant parameters of the same experiment. It is also worth to mention, that none of the toxins had strong genotoxic effect as shown by the total lack of highly damaged cells (score 4). To our knowledge this is the first study analyzing the DNA damage on sperm cells by comet assay in orally mycotoxin exposed animals. There is only one in vitro study using sperm cells for comet assay in mycotoxin genotoxicity studies [73-75], in which human spermatozoa and lymphocytes were assessed for their sensitivity to the genotoxic effect of DON. It was described that spermatozoa are more sensitive towards DON (in terms of DNA damage) as compared to lymphocytes. The reason may be that spermatozoa are a highly specialized cell types with no cytoplasm and extremely condensed chromatin, which seems to be prone towards toxic stimuli. Thus the moderate genotoxic effect of DON has been proven and spermatozoa comet assay as a rapid test for genotoxicity recommended.

Conclusions

In this study three orally administered fusariotoxins (FB, DON and ZEA) were tested in adult male rabbits, focusing primarily on their reproduction endpoints. The dietary levels were relatively low according to the EU limits in finished feed for young pig, in the absence of limits for rabbits’ feed (based on the European Commission Recommendation 2006/576/EC and the European Commission Directive 2003/100/EC). Results indicate that a prolonged low-dose mycotoxin exposure may adversely affect male reproduction. Reproductive processes show higher sensitivity to toxic effects, shown by the different parameters examined. Among the mycotoxins studied additive or less than additive effect was found in case of spermatogenesis and sperm cell morphology, synergism in testosterone production, while FB1 acted antagonistic against DON+ZEA on feed intake, lipid-peroxidation, and genotoxicity. All mycotoxins provoked moderate lipid-peroxidation and exerted slight genotoxicity.

Acknowledgements

The research was supported by the Hungarian Academy of Sciences (within the frame of the MTA-KE “Mycotoxins in the food chain” Research Group), by the National Scientific Research Fund (OTKA PD 104823) to KB and János Bolyai Research Grants of the Hungarian Academy of Sciences (BO/261/13 and BO/499/13) to KB and JSzF.

References

  1. Jelinek CF, Pohland AE, Wood GE (1989) Worldwide occurrence of mycotoxins in foods and feeds-an update. J Assoc Off Anal Chem 72: 223-230.
  2. Placinta CM, D’Mello JF, Macdonald AC (1999) A review of worldwide contamination of cereal grains and animal feed with Fusariummycotoxins. Animal Feed Science and Technology 78: 21-37.
  3. Marroquín-Cardona AG, Johnson NM, Phillips TD, Hayes AW (2014) Mycotoxins in a changing global environment-a review. Food ChemToxicol 69: 220-230.
  4. Glenn AE (2007) MycotoxigenicFusarium species in animal feed. Animal Feed Science and Technology 137: 213-24.
  5. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans (2002) Some traditional herbal medicines, some mycotoxins, naphthalene and styrene. IARC MonogrEvalCarcinog Risks Hum 82: 1-556.
  6. Wegulo SN1 (2012) Factors influencing deoxynivalenol accumulation in small grain cereals. Toxins (Basel) 4: 1157-1180.
  7. Pestka JJ, Zhou HR, Moon Y, Chung YJ (2004) Cellular and molecular mechanisms for immune modulation by deoxynivalenol and other trichothecenes: unraveling a paradox. ToxicolLett 153: 61-73.
  8. González HH, Martínez EJ, Pacin AM, Resnik SL, Sydenham EW (1999) Natural co-occurrence of fumonisins, deoxynivalenol, zearalenone and aflatoxins in field trial corn in Argentina. Food AdditContam 16: 565-569.
  9. Osweiler GD (1986) Diagnosis of mycotoxicoses Occurrence and clinical manifestations of trichothecenetoxicoses and zearalenonetoxicoses. Richard JL, Thurston JR, editors. National Animal Disease Center; Ames, Iowa, USA.
  10. Streit E, Schwab C, Sulyok M, Naehrer K, Krska R, et al. (2013) Multi-mycotoxin screening reveals the occurrence of 139 different secondary metabolites in feed and feed ingredients. Toxins (Basel) 5: 504-523.
  11. Cortinovis C, Pizzo F, Spicer LJ, Caloni F (2013) Fusariummycotoxins: effects on reproductive function in domestic animals-a review. Theriogenology 80: 557-564.
  12. Young LG, King GJ (1983) Prolonged feeding of low levels of zearalenone to young boars. Journal of Animal Science 57: 313-314.
  13. Sprando RL, Collins TF, Black TN, Olejnik N, Rorie JI, et al. (2005) Characterization of the effect of deoxynivalenol on selected male reproductive endpoints. Food ChemToxicol 43: 623-635.
  14. Gbore FA, Egbunike GN (2008) Testicular and epididymal sperm reserves and sperm production of pubertal boars fed dietary fumonisin B(1). AnimReprodSci 105: 392-397.
  15. Gbore FA1 (2009) Reproductive organ weights and semen quality of pubertal boars fed dietary fumonisin B1. Animal 3: 1133-1137.
  16. Minervini F, Lacalandra GM, Filannino A, Nicassio M, Visconti A, et al. (2010) Effects of in vitro exposure to natural levels of zearalenone and its derivatives on chromatin structure stability in equine spermatozoa. Theriogenology 73: 392-403.
  17. Kovács M, Tornyos G, Matics Z, Mézes M, Balogh K, et al. (2013) Effect of chronic T-2 toxin exposure in rabbit bucks, determination of the No Observed Adverse Effect Level (NOAEL). AnimReprodSci 137: 245-252.
  18. Speijers GJ, Speijers MH (2004) Combined toxic effects of mycotoxins. ToxicolLett 153: 91-98.
  19. Griessler K, Rodrigues I, Handl J, Hofstetter U (2010) Occurrence of mycotoxins in Southern Europe. World Mycotoxin Journal 3: 301-309.
  20. Kouadio JH, Dano SD, Moukha S, Mobio TA, Creppy EE (2007) Effects of combinations of Fusariummycotoxins on the inhibition of macromolecular synthesis, malondialdehyde levels, DNA methylation and fragmentation, and viability in Caco-2 cells. Toxicon 49: 306-317.
  21. Lei M, Zhang N, Qi D (2013) In vitro investigation of individual and combined cytotoxic effects of aflatoxin B1 and other selected mycotoxins on the cell line porcine kidney 15. ExpToxicolPathol 65: 1149-1157.
  22. Wan LY, Turner PC, El-Nezami H (2013) Individual and combined cytotoxic effects of Fusarium toxins (deoxynivalenol, nivalenol, zearalenone and fumonisins B1) on swine jejunal epithelial cells. Food ChemToxicol 57: 276-283.
  23. Wan LY, Woo CS, Turner PC, Wan JM, El-Nezami H (2013) Individual and combined effects of Fusarium toxins on the mRNA expression of pro-inflammatory cytokines in swine jejunal epithelial cells. ToxicolLett 220: 238-246.
  24. Fenske M, Fink-Gremmels J (1990) Effects of fungal metabolites on testosterone secretion in vitro. Arch Toxicol 64: 72-75.
  25. Khera KS, Whalen C, Angers G (1986) A teratology study on vomitoxin (4-deoxynivalenol) in rabbits. Food ChemToxicol 24: 421-424.
  26. Gumprecht LA, Marcucci A, Weigel RM, Vesonder RF, Riley RT, et al. (1995) Effects of intravenous fumonisin B1 in rabbits: nephrotoxicity and sphingolipid alterations. Nat Toxins 3: 395-403.
  27. Mariscal-Quintanar MG, Garcia-Escamilla RM, Garcia-Escamilla N, Torres-Lopez J, Bautista-Ordonez JA, et al.(1997) Effect of ingesting Aspergillusflavus and Fusariummoniliforme on the cytology of bone marrow and blood and on serum albumin and globulin concentrations. Veterinaria Mexico 28: 75-81.
  28. Orova Z (2003) Investigations on the teratogenic effects of fumonisin B1 in swine and rabbit. Thesis, University of Kaposvár, Kaposvár.
  29. LaBorde JB, Terry KK, Howard PC, Chen JJ, Collins TF, et al. (1997) Lack of embryotoxicity of fumonisin B1 in New Zealand white rabbits. FundamApplToxicol 40: 120-128.
  30. Grenier B, Oswald I (2011) Mycotoxin co-contamination of food and feed: meta-analysis of publications describing toxicological interactions. World Mycotoxin Journal 4: 285-313.
  31. Placer ZA, Cushman LL, Johnson BC (1966) Estimation of product of lipid peroxidation (malonyldialdehyde) in biochemical systems. Anal Biochem 16: 359-364.
  32. Botsoglou NA, Fletouris DJ, Papageorgiou GE, Vassilopoulos VN, Mantis AJ, et al. (1994) Rapid, sensitive and specific thiobarbituric acid method for measuring lipid peroxidation in animal tissue, food and feedstuff samples. Journal Agricultural and Food Chemistry 42: 1931-1937.
  33. AOAC (1984) Official Methods of Analysis. (14thedn), Association of Official Analytical Chemists, Arlington, VA, USA.
  34. Sedlak J, Lindsay RH (1968) Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman's reagent. Anal Biochem 25: 192-205.
  35. Lawrence RA, Burk RF (1978) Species, tissue and subcellular distribution of non Se-dependent glutathione peroxidase activity. J Nutr 108: 211-215.
  36. Csernus V (1982) Antibodies of high affinity and specificity for RIA determination of progesterone, testosterone, estradiol-17ß and cortisol. In: Görög S (eds) Advances in steroid analysis. Academic Press, Budapest, Hungary.
  37. Kovács M, Tornyos G, Matics Z, Kametler L, Rajli V, et al. (2011) Subsequent effect of subacute T-2 toxicosis on spermatozoa, seminal plasma and testosterone production in rabbits. Animal 5: 1563-1569.
  38. World Health Organisation (1999) WHO laboratory manual for the examination of human semen and sperm-cervical mucus interaction. (4thedn), Cambridge University Press, UK.
  39. Simon L, Carrell DT (2013) Sperm DNA damage measured by comet assay. Methods MolBiol 927: 137-146.
  40. Gopalan RC, Emerce E, Wright CW, Karahalil B, Karakaya AE, et al. (2011) Effects of the anti-malarial compound cryptolepine and its analogues in human lymphocytes and sperm in the Comet assay. ToxicolLett 207: 322-325.
  41. Singh NP, McCoy MT, Tice RR, Schneider EL (1988) A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 175: 184-191.
  42. Collins A, Dusinská M, Franklin M, Somorovská M, Petrovská H, Duthie S, et al. (1997) Comet assay in human biomonitoring studies: reliability, validation, and applications. Environ Mol Mutagen 30: 139-146.
  43. OECD Principles of Good Laboratory Practice (as revised in 1997). OECD Environmental Health and Safety Publications (OECD).
  44. Rotter BA, Prelusky DB, Pestka JJ (1996) Toxicology of deoxynivalenol (vomitoxin). J Toxicol Environ Health 48: 1-34.
  45. Harcourt-Brown F (2002) Textbook of rabbit medicine. (1stedn), Elsevier Science. New York, USA.
  46. Hewitt MA, Girgis GN, Brash M, Smith TK (2012) Effects of feed-borne Fusariummycotoxins on performance, serum chemistry, and intestinal histology of New Zealand White fryer rabbits. J AnimSci 90: 4833-4838.
  47. Gelderblom WC, Seier JV, Snijman PW, Van Schalkwyk DJ, Shephard GS, et al. (2001) Toxicity of culture material of Fusariumverticillioides strain MRC 826 to nonhuman primates. Environ Health Perspect 109 Suppl 2: 267-276.
  48. Ewuola EO, Egbunike GN (2010) Effects of dietary fumonisin B1 on the onset of puberty, semen quality, fertility rates and testicular morphology in male rabbits. Reproduction 139: 439-445.
  49. Kim EJ, Jeong SH, Cho JH, Ku HO, Pyo HM, et al. (2008) Plasma haptoglobin and immunoglobulins as diagnostic indicators of deoxynivalenol intoxication. J Vet Sci 9: 257-266.
  50. Tessari EN, Oliveira CA, Cardoso AL, Ledoux DR, Rottinghaus GE (2006) Effects of aflatoxin B1 and fumonisin B1 on body weight, antibody titres and histology of broiler chicks. Br PoultSci 47: 357-364.
  51. Marin DE, Taranu I, Pascale F, Lionide A, Burlacu R, et al. (2006) Sex-related differences in the immune response of weanling piglets exposed to low doses of fumonisin extract. Br J Nutr 95: 1185-1192.
  52. Mead JF, Alfin-Slater RB, Howton DR, Popják G (1986) Lipids: Chemistry, biochemistry and nutrition. Plenum Press, NY, USA.
  53. Mishra S, Tripathi A, Chaudhari BP, Dwivedi PD, Pandey HP, et al. (2014) Deoxynivalenol induced mouse skin cell proliferation and inflammation via MAPK pathway. ToxicolApplPharmacol 279: 186-197.
  54. El Golli-Bennour E, Bacha H (2011) Hsp70 expression as biomarkers of oxidative stress: mycotoxins' exploration. Toxicology 287: 1-7.
  55. Minervini F, Garbetta A, D'Antuono I, Cardinali A, Martino NA, et al. (2014) Toxic mechanisms induced by fumonisin b1 mycotoxin on human intestinal cell line. Arch Environ ContamToxicol 67: 115-123.
  56. Poersch AB, Trombetta F, Braga AC, Boeira SP, Oliveira MS, et al. (2014) Involvement of oxidative stress in subacute toxicity induced by fumonisin B1 in broiler chicks. Vet Microbiol 174: 180-185.
  57. Liu Q, Wang Y, Gu J, Yuan Y, Liu X, et al. (2014) Zearalenone inhibits testosterone biosynthesis in mouse Leydig cells via the crosstalk of estrogen receptor signaling and orphan nuclear receptor Nur77 expression. Toxicology In Vitro 28: 647-656.
  58. Lu ZH, Mu YM, Wang BA, Li XL, Lu JM, et al. (2003) Saturated free fatty acids, palmitic acid and stearic acid, induce apoptosis by stimulation of ceramide generation in rat testicular Leydig cell. BiochemBiophys Res Commun 303: 1002-1007.
  59. Tajima O, Schoen ED, Feron VJ, Groten JP (2002) Statistically designed experiments in a tiered approach to screen mixtures of Fusariummycotoxins for possible interactions. Food ChemToxicol 40: 685-695.
  60. Boeira SP, Funck VR, Borges Filho C, Del'Fabbro L, de Gomes MG, et al . (2015) Lycopene protects against acute zearalenone-induced oxidative, endocrine, inflammatory and reproductive damages in male mice. Chemico-Biological Interaction 25: 50-57.
  61. Milano GD, Odriozola E, Lopez TA (1991) Lack of effect of a diet containing zearalenone on spermatogenesis in rams. Vet Rec 129: 33-35.
  62. Benzoni E, Minervini F, Giannoccaro A, Fornelli F, Vigo D, et al. (2008) Influence of in vitro exposure to mycotoxinzearalenone and its derivatives on swine sperm quality. ReprodToxicol 25: 461-467.
  63. Rajkovic A, Uyttendaele M, Debevere J (2007) Computer aided boar semen motility analysis for cereulide detection in different food matrices. Int J Food Microbiol 114: 92-99.
  64. Tsakmakidis IA, Lymberopoulos AG, Alexopoulos C, Boscos CM, Kyriakis SC (2006) In vitro effect of zearalenone and alpha-zearalenol on boar sperm characteristics and acrosome reaction. ReprodDomestAnim 41: 394-401.
  65. Filannino A, Stout TA, Gadella BM, Sostaric E, Pizzi F, et al. (2011) Dose-response effects of estrogenic mycotoxins (zearalenone, alpha- and beta-zearalenol) on motility, hyperactivation and the acrosome reaction of stallion sperm. Reproductive Biology and Endocrinology 9: 134.
  66. de Kretser DM, Kerr JB (1994) The cytology of testis. In: E Knobil and JD Neill (eds) The physiology of reproduction. Raven Press Ltd, New York, USA.
  67. Müller S, Dekant W, Mally A (2012) Fumonisin B1 and the kidney: modes of action for renal tumor formation by fumonisin B1 in rodents. Food ChemToxicol 50: 3833-3846.
  68. Zatecka E, Ded L, Elzeinova F, Kubatova A, Dorosh A, et al. (2014) Effect of zearalenone on reproductive parameters and expression of selected testicular genes in mice. Reproductive Toxicology 45: 20-30.
  69. Kim IH, Son HY, Cho SW, Ha CS, Kang BH (2003) Zearalenone induces male germ cell apoptosis in rats. ToxicolLett 138: 185-192.
  70. Cho ES, Ryu SY, Jung JY, Park BK, Son HY (2011) Effects of red ginseng extract on zearalenone induced spermatogenesis impairment in rat. J Ginseng Res 35: 294-300.
  71. Baumgartner A, Kurzawa-Zegota M, Laubenthal J, Cemeli E, Anderson D (2012) Comet-assay parameters as rapid biomarkers of exposure to dietary/environmental compounds - an in vitro feasibility study on spermatozoa and lymphocytes. Mutation Research 743: 25-35.
  72. Conková E, Laciaková A, Pástorová B, Seidel H, Kovác G (2001) The effect of zearalenone on some enzymatic parameters in rabbits. ToxicolLett 121: 145-149.
  73. Marína MA, Kalafová M, Schneidgenová N, Maruniaková N (2012) The effect of deoxynivalenol on rabbit spermatozoa motility in vitro. Journal of Microbiology, Biotechnology and Food Sciences 2: 368-377.
Citation: Szabó-Fodor J, Kachlek K, Cseh S, Somoskoi B, Szabó A, et al. (2015) Individual and Combined Effects of Subchronic Exposure of Three Fusarium Toxins (Fumonisin B, Deoxynivalenol and Zearalenone) in Rabbit Bucks. J Clin Toxicol 5:264.

Copyright: © 2015 Szabó-Fodor J, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top