You are on page 1of 17

Food and Chemical Toxicology 111 (2018) 27–43

Contents lists available at ScienceDirect

Food and Chemical Toxicology


journal homepage: www.elsevier.com/locate/foodchemtox

Individual and combined effects of Fusarium toxins on apoptosis in PK15 T


cells and the protective role of N-acetylcysteine
Wei Zhanga, Shihua Zhanga, Meiling Zhanga, Lige Yanga, Baojing Chenga, Jianping Lia,b,∗,
Anshan Shana,∗∗
a
Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, PR China
b
Key Laboratory of Animal Common Disease Prevention, Northeast Agricultural University, Harbin, 150030, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Deoxynivalenol (DON), zearalenone (ZEN) and fumonisin B1 (FB1) are among the most toxicologically important
Deoxynivalenol Fusarium toxins commonly found in nature that lead to nephrotoxicity in animals. The present study investigated
Zearalenone that the individual and combined effects of subcytotoxic DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) on
Fumonisin B1 oxidative stress and apoptosis in porcine kidney cells (PK15). In addition, the protective effect of N-acet-
N-acetylcysteine
ylcysteine (NAC) against the toxicity of Fusarium toxins was also evaluated. Our results showed that the activities
PK15 cells
Apoptosis
of glutathione reductase (GR) and total superoxide dismutase (SOD) were affected by DON, ZEN and FB1, and
this change in activity induced reactive oxygen species (ROS) and malondialdehyde (MDA) production, in-
creased apoptosis and regulated the mRNA expression of Bax, Bcl-2, caspase-3, caspase-9, cytochrome c (cyto c)
and P53. This study demonstrated the complexity of combined mycotoxin infection since the combination of
toxins exhibited more profound defects in the oxidative stress responses and apoptosis. Moreover, NAC reduced
the oxidative damage and inhibited the apoptosis induced by Fusarium toxins. It was concluded that oxidative
damage and apoptosis through the mitochondria-dependent channel were the mechanisms of Fusarium toxin
mediated toxicity, and NAC reversed these damages to some extent.

1. Introduction of the growth hormone axis, and altered gut integrity (Pestka, 2010).
Moreover, DON significantly affects fetal development and the innate
Mycotoxins are toxic secondary metabolites produced by a range of immune system (Kinser et al., 2005; Collins et al., 2006; Pestka and
fungi that can lead to numerous adverse health effects in animals and Amuzie, 2008).
humans (Wentzel et al., 2016). The Food and Agriculture Organization ZEN is a Fusarium fungal metabolite with oestrogenic properties
of the United Nations (FAO) estimated that up to 25% of the world's (Diaz, 2005). Currently, studies not only focused on ZEN-induced le-
food crop is contaminated with mycotoxins. Aflatoxins, ochratoxins, sions of the reproductive organs but also demonstrated the histo-
trichothecenes, zearalenone, fumonisins, tremorgenic toxins, and ergot pathological damage, oxidative stress and inflammatory cytokines
alkaloids are the mycotoxins of greatest agro-economic importance production in pregnant animals and their offspring (Jia et al., 2015; Yin
(Hussein and Brasel, 2001). Among these mycotoxins, DON, ZEN and et al., 2015; Zhang et al., 2015). In addition, intake of ZEN could induce
FB1 are the most toxicologically important toxins that occur frequently different degrees of hematotoxicity and negatively affects immune
in food and animal feeds (Martins et al., 2006; Monbaliu et al., 2010; function (Yang et al., 2016). In vitro studies showed that ZEN induced
Garrido et al., 2012). obvious apoptosis in endometrial stromal cells (ESCs) via the Bcl-2 fa-
DON is one of the most common Fusarium trichothecene mycotoxin. mily and caspases-dependent signaling pathway (Hu et al., 2016).
This large family of toxins is generally associated with feed refusal, Fumonisins (FUM) can inhibit lipid synthesis in biological mem-
vomiting, and lesions of the gastrointestinal tract in animals (Diaz, branes and cause a lethal condition, equine leucoencephalomalacia,
2005). Pathophysiologic effects associated with DON include altered that is characterized by massive atrophy of the brain and sudden death
neuroendocrine signaling, proinflammatory gene induction, disruption (Diaz, 2005). Swine are less sensitive to FUM, which is characterized by


Corresponding author. Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, PR China.
∗∗
Corresponding author. Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, PR China.
E-mail addresses: dianjini2012@163.com (W. Zhang), 931048522@qq.com (S. Zhang), 1768385490@qq.com (M. Zhang), ylgpy123456@163.com (L. Yang),
63300674@qq.com (B. Cheng), ljpneau@163.com (J. Li), asshan@neau.edu.cn (A. Shan).

http://dx.doi.org/10.1016/j.fct.2017.10.057
Received 24 April 2017; Received in revised form 19 October 2017; Accepted 30 October 2017
Available online 07 November 2017
0278-6915/ © 2017 Elsevier Ltd. All rights reserved.
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

pulmonary oedema. FB1 is a potent toxin which causes disorders in lipid Table 1
metabolism, renal filtration perturb, rhabdomyolysis and blood lym- Design matrix from three Fusarium toxins and NAC.
phocyte cell death (Kouadio et al., 2013). Moreover, FB1 causes brain
Group Treatment
hyperexcitability in vivo and mitochondrial dysfunction may represent
a potential underlying mechanism (Poersch et al., 2015). DON ZEN FB1 NAC
NAC, a well-known thiol antioxidant that acts as a glutathione 0.25 μM 20 μM 10 μM 0.5 mM
precursor and reacts with OH, NO2, CO3 (−), and thiyl radicals, is
1 – – – –
widely used in clinical practice (Samuni et al., 2013). It has anti-in- 2 + – – –
flammatory effects, impacts apoptosis and neurogenesis, and reverses 3 – + – –
models of mitochondrial toxicity (Asevedo et al., 2014). Supple- 4 – – + –
mentation with NAC (50 μM) resulted in significant cytoprotection, a 5 + + – –
6 + – + –
reduction in ROS generation, higher antioxidant levels that were similar 7 – + + –
to that of control cells and the inhibition of DNA strand breaks induced 8 + + + –
by hypoxia (Jayalakshmi et al., 2005). Moreover, NAC has protective 9 – – – +
effects against DNA damage and carcinogenesis (Flora et al., 2001). 10 + – – +
11 – + – +
DON, ZEN and FB1 are likely to damage the host simultaneously
12 – – + +
because of their co-occurrence in contaminated cereal grains. As the 13 + + – +
main excretory organ, the kidney may be exposed to high concentra- 14 + – + +
tions of the toxin after the ingestion of mycotoxin-contaminated food. 15 – + + +
Although some researchers have discussed the individual effects of 16 + + + +

DON, ZEN or FB1 in different in vitro models including intestinal porcine


+: Mycotoxin/NAC treatment.
epithelial cell lines (IPEC) (Dänicke et al., 2010), the Chinese Hamster −: No mycotoxin/NAC treatment.
Ovary (CHO-K1) cell line (Ferrer et al., 2009) and both human and pig
lymphocytes (Mwanza et al., 2009). However, the reports on the then, 100 μl (0.5 mg/ml) of MTT solution was added to each well. After
combined and interactive effects of DON, ZEN and FB1 on kidney cell a 4 h incubation at 37 °C, the MTT solution from each well was dis-
are limited. Therefore, this research aimed to investigate the individual carded and 100 μl of DMSO was added to each well and shaken for
and combined effects of DON, ZEN and FB1 on the activity of anti- 5 min to solubilize the formazan formed in the viable cells. The ab-
oxidant enzymes, the level of MDA and ROS and the mRNA expression sorbance was measured at 570 nm using a GENios microplate reader
of apoptosis-related genes in PK15 cells. In addition, the protective role (Tecan Austria GmbH, Austria). The viability of the cells treated with
of NAC was investigated. Fusarium toxins was expressed as a percentage compared to the control
(non-mycotoxin treated). From these data, subcytotoxic concentrations
2. Materials and methods were used for subsequent study. In addition, to assess the protective
role of NAC in Fusarium mycotoxins-induced cytotoxicity, NAC (0.5,
2.1. Chemicals 1.0, and 2.0 mM) was co-administered with DON, ZEN or FB1 for 24 h.

DON, ZEN, FB1, NAC, [3-(4,5-dimethylthiazol-2-yl)-2,5- diphe-


2.4. Inscribed central composite design for complete mixtures
nyltetrazolium bromide] (MTT) and dimethyl sulfoxide (DMSO) were
obtained from Sigma Aldrich (St. Louis, MO, USA). DON, ZEN and FB1
An inscribed central composite design including a fractional fac-
were dissolved in DMSO and NAC was dissolved in Dulbecco's modified
torial part was applied with four factors (DON, ZEN, FB1 and NAC) to
Eagle's medium (DMEM) containing high glucose (4.5 g/L) obtained
minimize the number of possible toxin combinations from 44 (all pos-
from GE Healthcare Life Sciences (Beijing, China). Fetal bovine serum
sible combinations of every concentration of each toxin/NAC) to 16.
(FBS) was obtained from Gibco-Life Technology (Eggenstein,
The design matrix is shown in Table 1. The cell viability was de-
Germany). The final concentration of the DMSO used as a solvent in the
termined by MTT assay as described above.
culture medium was 0.5%. All other chemicals were purchased from
Beyotime Biotechnology (Nantong, China).
2.5. Antioxidant enzymes activity assays
2.2. Cell culture
Cells were seeded at a density of 2.4 × 105 cells/well in 6-well
The porcine kidney cell line (PK15) was obtained from the Harbin plates and incubated for 24 h. Subsequently, cells were rinsed with PBS
veterinary research institute (Harbin, China). Cells were cultured in and treated as described in Table 1. After 24 h incubation, protein
DMEM supplemented with 10% FBS and 1% penicillin-streptomycin concentration and the activity of glutathione reductase (GR) and total
solution at 37 °C in a humid atmosphere of 5% CO2. Cells (2 × 105/ml) superoxide dismutase (SOD) were assessed using kits (Beyotime Bio-
were seeded in 5 ml of medium in a 25 cm2 flasks and cultured for 36 h. technology) according to the manufacturer's instructions. The results
Thereafter, the cultured cells were either passaged or used in experi- were expressed as U per mg protein.
ments. Cell monolayers were washed with phosphate-buffered saline
(PBS) and trypsinized with 1 × trypsin/EDTA. 2.6. Lipid peroxidation assay

2.3. Preliminary concentration–response experiment: cell viability assay by The level of lipid peroxidation was measured via the 2-thiobarbi-
MTT turic acid (TBA) color reaction for malondialdehyde (MDA) by using the
kit (Beyotime Biotechnology) according to the manufacturer's instruc-
The MTT assay was performed to assess cell viability (Mosmann, tions. The results were expressed as nmol per mg protein.
1983). Cells were seeded at a density of 2000–4000 cells/well in 96-
well plates and incubated for 24 h. Subsequently, the cells were rinsed 2.7. Evaluation of ROS level
with PBS and treated with Fusarium toxins (0.25–8 μM of DON,
5–160 μM ZEN and 5–160 μM FB1) in serum-free media for 24 h. At the Cells were seeded at a density of 2.4 × 105 cells/well in 6-well
end of the incubation, the cells in each well were washed with PBS, and plates and incubated for 24 h. After treatment with or without Fusarium

28
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

toxins or/and NAC for 24 h, the cells were rinsed 2 times using PBS and least three independent experiments. The relative expressions of the
incubated with 10 μM DCFH-DA at 37 °C for 20 min according to the apoptosis-related gene mRNAs were determined with the 2-△△Ct
manufacturer's instructions. DCF fluorescence was detected by a method (Livak and Schmittgen, 2001).
fluorescence microplate reader (Tecan Austria GmbH, Austria) at the
excitation wavelength of 488 nm and emission wavelength of 525 nm.
The level of ROS was expressed as a percentage of the controls. 2.10. Western blotting

PK15 cells were seeded in 6-well plates and incubated for 24 h. After
2.8. Apoptosis status
the indicated treatment, cells were harvested, washed twice with ice-
cold PBS (pH 7.2) and lysed in ice-cold RIPA buffer containing complete
To analyze the cell apoptosis status, a double-staining assay with
protease inhibitors (PMSF). The lysate was centrifuged at 10,000 rpm
Annexin V-fluorescein sothiocyanate (FITC) was performed. After 24 h
for 10 min at 4 °C, and the supernatant was stored at −80 °C until
incubation with or without Fusarium toxins or/and NAC, the cells were
further analysis. Then equal amounts of lysate (protein samples) were
trypsinized and collected and then rinsed 2 times using PBS. Finally,
loaded onto polyacrylamide gels and transferred onto polyvinylidene
cells were resuspended in 195 μl of binding buffer. Then, 5 μl of
fluoride (PVDF) membranes (Millipore, USA). After blocking with 5%
Annexin V- FITC and 10 μl of propidium iodide (PI) were added to the
milk in TBS containing 0.05% Tween 20 at 37 °C for 2 h, the mem-
tubes that were then gently vortexed. Subsequently, the cells were in-
branes were incubated with the first antibody against targeted protein
cubated for 20 min at room temperature in the dark, and then 1 ml of
at 4 °C overnight. Followed by incubation with the corresponding sec-
binding buffer was added to each tube. Thereafter, the cells were
ondary antibodies (dilution, 1: 5000) for 1 h at room temperature.
analyzed by flow cytometry (Becton, Dickinson and Company,
Membranes were then exposed to a super enhanced chemiluminescence
America) at an excitation wavelength of 488 nm and emission wave-
(ECL) Plus detection system (P1010, Applygen, Beijing, China) and
length of 530 nm.
developed using a Kodak film cartridge and film. The expression of each
protein was normalized to that of β-actin. Antibodies against Bcl-2
2.9. Quantitative real-time PCR (D160117) and caspase-3 (D120074) were purchased from Sangong
Biotech Co., Ltd. (Shanghai, China).
The cells were seeded at a density of 1.2 × 105 cells/well in 12-well
plates and incubated for 24 h. After treatment with or without myco-
toxin or/and NAC for 24 h, the cells were rinsed 2 times using PBS and 2.11. Statistical analysis
collected. Total RNA was extracted from fresh cells by using TRIzol
(Invitrogen China, Shanghai, China) following the manufacturer's in- All experiments were performed at least three independent experi-
structions. The concentration of RNA was measured by using a ments. The statistical analysis was conducted using the Statistical
NanoPhotometer P-Class (IMPLEN, Germany). Total RNA was con- Product and Service Solutions software (SPSS Inc., Chicago, IL, USA),
verted into cDNA by the SYBR Premix Ex Taq kit (Takara, Dalian, and the data were expressed as mean ± SD. The significance of dif-
China) according to the manufacturer's instructions and used for RT- ference between two groups was analyzed by Student's t-test with
PCR. SYBR Green I RT-PCR kit (Takara, Dalian, China) was used to subsequent Bonferroni correction. IC50 and IC10 values were calcu-
measure the mRNA expression of apoptosis-related genes (P53, Bax, lated by using probit regression in SPSS 19.
cytochrome c, caspase-9, caspase-3 and Bcl-2), and the expression level
was expressed relative to the quantity of the β-actin endogenous con-
trol. As shown in Table 2, the primers were designed from published 3. Results
GenBank sequences and were synthesized by Sangon (Shanghai, China).
For analyses on an Applied Biosystems 7500 Real-Tine PCR thermal 3.1. Preliminary concentration–response experiment on cell viability
cycler apparatus (Applied Biosystems, Foster City, CA, USA), PCR re-
actions were performed with 1.0 μl of cDNA and 0.2 μl of each of the To determine the suitable experimental concentrations of Fusarium
primers in a final volume of 10 μl. PCR reactions were initialed at 95 °C toxins for subsequent interaction experiments, the cytotoxic effects of
for 30 s, followed by 40 cycles of denaturing at 95 °C for 5 s and an- the individual toxins on the PK15 cells was first determined by mea-
nealing at 60 °C for 34 s. All of the PCR reactions were performed at suring the cell viability (MTT assay) after incubation for 24 h in the
absence or presence of DON, ZEN or FB1. The results are shown in
Table 2 Fig. 1. DON, ZEN and FB1 reduced the cell viability in a dose-dependent
Primers used for qRT-PCR. manner with statistically significant effects observed at 0.25 μM, 20 μM
and 10 μM, respectively. The IC50 values by the MTT assay were
Genes Sequences (5′→3′) Fragments Accession
size (bp) number 2.08 μM, 210.7 μM and 196.1 μM for DON, ZEN and FB1, respectively.
IC10 values were 0.157 μM, 27.583 μM and 9.882 μM for DON, ZEN
β-actin FP:ATGCTTCTAGGCGGACTGT 211 AY550069 and FB1, respectively. Based on these results, subcytotoxic concentra-
RP:CCATCCAACCGACTGCT
tions close to IC10 of Fusarium toxins [DON (0.25 μM), ZEN (20 μM)
Bcl-2 FP: GCGACTTTGCCGAGATGT 116 AB271960.1
RP: CACAATCCTCCCCCAGTTC and FB1 (10 μM)] were chosen for the subsequent experiments. Cyto-
Bax FP: TTTGCTTCAGGGTTTCATCC 113 XM_ protective actions of NAC on cell death induced by Fusarium toxins were
003127290.3 investigated in the present study. As shown in Fig. 1D, when NAC (0.5,
RP: GACACTCGCTCAACTTCTTGG 1.0, and 2.0 mM) was co-administered with DON (0.25 μM), ZEN
Caspase-3 FP: TTGGACTGTGGGATTGAGAC 121 NM_214131.1
RP: TTCGCCAGGAATAGTAACCAG
(20 μM) or FB1 (10 μM) for 24 h, the cell death induced by Fusarium
Caspase-9 FP: GGACATTGGTTCTGGAGGATT 116 XM_ toxins was prevented in part by NAC (p < 0.05). However, the highest
RP: TGTTGATGATGAGGCAGTGG 013998997.1 NAC concentration (2.0 mM) did not show a greater protective role
P53 FP: CGAACTGGCTGGATGAAAAT 124 AF098067.1 when compared with 0.5 mM NAC. Therefore, 0.5 mM NAC was chosen
RP: GAAGGGACAAAGGACGACAG
to explore its protective role in PK15 exposed to Fusarium toxins in the
Cyto C FP: CTCTTACACAGATGCCAACAA 139 NM_
RP: TTCCCTTTCTCCCTTCTTCT 001129970.1 further study.

FP forward primer, RP reverse primer.

29
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 1. Effects of DON (A), ZEN (B) and FB1 (C) on cell viability and cytoprotective action of NAC (D). (A–C) The cells were exposed to DON (0.25–8 μM), ZEN (5–160 μM) or FB1
(5–160 μM) for 24 h. (D) The cells were incubated with DON (0.25 μM), ZEN (20 μM), FB1 (10 μM) and/or NAC (0.5, 1.0 and 2.0 mM) for 24 h. Cellular viability was determined by the
MTT test. The results are expressed as a percentage of the control. Bars represent mean ± SD of three parallel measurements. #, ##, ### means significantly different from controls at
p < 0.05, p < 0.01 and p < 0.001, respectively. * means statistical significance between cells cultured in the absence and presence of NAC. *p < 0.05.

3.2. Influences of Fusarium toxins and NAC on antioxidant enzyme 3.3. Influences of Fusarium toxins and NAC on lipid peroxidation
activities
The results showed that the MDA level of 0.25 μM DON, 10 μM FB1
3.2.1. GR activity and all mycotoxin mixture treatments was significantly higher than that
As shown in Fig. 2A, GR activity of DON treatment and mixture of in the control (p < 0.05), and the highest MDA level was observed in
DON-ZEN, DON-FB1 and DON-ZEN-FB1 significantly decreased DON-ZEN-FB1 mix (2.67 ± 0.12 nmol/mg protein) (Fig. 4A). For the
(p < 0.01), and the lowest GR activity was observed in DON-ZEN-FB1 mixtures of two Fusarium toxins, the MDA levels were higher than in the
mix (3.69 ± 0.30 U/mg protein). The GR activity was significantly individual mycotoxin treatments (p < 0.05) (Fig. 4B–D). The MDA
lower in the mixtures of DON-ZEN and DON-FB1 relative to mycotoxins level of DON-ZEN-FB1 was higher than mixtures of ZEN and DON or FB1
individually (p < 0.05) (Fig. 2B and C). The results showed that the (p < 0.001). Treatment with 0.5 mM NAC significantly decreased the
GR activity of DON-ZEN-FB1 mix was significantly lower than that of MDA level in cells treated with 0.25 μM DON and DON-FB1
the mixtures of two Fusarium toxins (p < 0.05) (Fig. 2E). The reduction (p < 0.001) (Fig. 4F).
in GR activity induced by DON-FB1 and DON-ZEN-FB1 was restored by
the treatment of cells with 0.5 mM NAC (Fig. 2F).
3.4. Influences of Fusarium toxins and NAC on ROS

3.2.2. SOD activity In the present study, ROS production was monitored as an indicator
As shown in Fig. 3A, the SOD activity of DON treatment and all of oxidative stress. Intracellular ROS production was significantly in-
mixtures significantly decreased (p < 0.05), and the lowest activity creased after 0.25 μM DON, 10 μM FB1 and the mixture of DON-ZEN,
was observed in DON-ZEN-FB1 mix (14.34 ± 0.56 U/mg protein). It DON-FB1 and DON-ZEN-FB1 treatments (p < 0.01) (Fig. 5A). The ROS
has been noted that SOD activity was significantly lower in cells treated level of DON-ZEN was significantly higher in the groups treated with
with mixtures of DON-ZEN and DON-FB1 relative to the individual mixtures of two Fusarium toxins than that of any of the individual
mycotoxins treatments (p < 0.05) (Fig. 3B and C). The result showed mycotoxins alone (p < 0.05) (Fig. 5B) and the ROS level of DON-FB1
that the SOD activity of DON-ZEN-FB1 mix was significantly lower than was significantly higher than FB1 alone (p < 0.01) (Fig. 5C). In ad-
that of DON-FB1 and ZEN-FB1 mix (p < 0.001) (Fig. 3E). As shown in dition, in the mixture of three Fusarium toxins, the ROS level was sig-
Fig. 3F, the recovery of the reduced SOD activity by NAC supple- nificantly higher than DON-FB1 mix (p < 0.01) (Fig. 5E). The increase
mentation was only observed in the DON-ZEN-FB1 mixture (Fig. 3F). in ROS induced by 0.25 μM DON, 10 μM FB1 and DON-FB1 was restored
by the treating the cells with 0.5 mM NAC (p < 0.05) (Fig. 5F).

30
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 2. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on GR activity in PK15 cells. Cells were exposed to individual or combinations of DON
(0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as U/mg protein. Each data point represents the mean ± SD of three measurements. #,
##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between two connected treatments
at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

3.5. Influences of Fusarium toxins and NAC on apoptosis mixture of these three Fusarium toxins, cell apoptosis was significantly
higher than in the DON-ZEN and DON-FB1 mix (p < 0.01) (Fig. 6E). As
Flow cytometry results showed that all treatments with mycotoxin shown in Fig. 6F, NAC could decrease the apoptosis induced by Fu-
significantly promoted cell apoptosis compared to the control sarium toxins to some extent.
(p < 0.05), and the highest cell apoptosis was observed in the DON-
ZEN-FB1 treatment (20.97 ± 0.40%) (Fig. 6A). Cell apoptosis in all
mixtures of two Fusarium toxins was significantly higher than that in the
individual mycotoxin treatments (p < 0.01) (Fig. 6B–D). And in the

31
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 3. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on SOD activity in PK15 cells. Cells were exposed to individual or combinations treatments of
DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as U/mg protein. Bars represent mean ± SD obtained from measurements
conducted in triplicate. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between
two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

3.6. Influences of Fusarium toxins and NAC on the apoptosis-related mRNA decreased the mRNA expression of Bax in the mycotoxin treatments to
expression some extent (Fig. 7F).

3.6.1. Bax 3.6.2. Bcl-2


Bax levels were significantly up-regulated in 0.25 μM DON, 20 μM Contrary to Bax mRNA expression, Bcl-2 mRNA expression was
ZEN and all mycotoxin mixture treatments (p < 0.01) (Fig. 7A). In the significantly lower in treatments of 0.25 μM DON, 20 μM ZEN and any
DON-ZEN and ZEN-FB1 treatments, the expression of Bax was sig- mixtures of Fusarium toxins compared to the control (p < 0.001)
nificantly higher than in the mycotoxin alone (p < 0.01) (Fig. 7B and (Fig. 8A). The expression of Bcl-2 was significantly lower in ZEN-FB1
C). The expression of Bax was significantly increased in DON-FB1 mix mix relative to the groups treated with either of these two mycotoxins
compared to FB1 alone (p < 0.001) (Fig. 7D). While, the Bax expres- alone (p < 0.001) (Fig. 8D). The expression of Bcl-2 was significantly
sion of DON-ZEN-FB1 was significantly higher than that of mixtures of decreased in DON-FB1 mix compared to FB1 alone (p < 0.001)
two Fusarium toxins (p < 0.05) (Fig. 7E). Treatment with 0.5 mM NAC (Fig. 8C). However, in the mixture of DON and ZEN, Bcl-2 expression

32
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 4. Individual and combined effects of DON, ZEN and


FB1 and the protective role of NAC on MDA production in
PK15 cells. Cells were exposed to individual or combina-
tions of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and
NAC (0.5 mM) for 24 h. The results are expressed as nmol/
mg protein. Each data point represents the mean ± SD of
three measurements. #, ##, ### means significantly dif-
ferent from control at p < 0.05, p < 0.01 and p < 0.001,
respectively (A). *, **, *** means significant differences
between two connected treatments at p < 0.05, p < 0.01
and p < 0.001, respectively (B–F).

was significantly higher than DON alone (Fig. 8B). And the Bcl-2 ex- caspase-9 was observed in the DON-ZEN-FB1 treatment (1.83 ± 0.11),
pression noted in DON-ZEN-FB1 mix was significantly lower than DON- and it was significantly higher than groups treated with mixture of any
ZEN, while higher than ZEN-FB1 (p < 0.05) (Fig. 8E). The reduction of two Fusarium toxins (p < 0.01) (Fig. 10E). The expression of caspase-9
Bcl-2 expression induced by mixtures of DON-FB1 and ZEN-FB1 was was significantly increased in DON-FB1 mix compared to DON alone
restored in the cells treated with 0.5 mM NAC (Fig. 8F). (p < 0.01) (Fig. 10C). Treatment with 0.5 mM NAC could significantly
decrease the mRNA expression of caspase-9 in 10 μM FB1 and DON-FB1
mix (p < 0.05) (Fig. 10F).
3.6.3. Caspase-3
Treatments with any Fusarium toxins except FB1 alone also induced
a significant up-regulation in caspase-3 (p < 0.05) (Fig. 9A). The
3.6.5. Cyto c
highest expression of caspase-3 was observed in DON-ZEN-FB1
Compared to the control, cyto c mRNA levels were significantly up-
(1.75 ± 0.06) treated cells, and it was significantly higher than that of
regulated in the 20 μM ZEN treatment and any mixture of Fusarium
any mixtures of two Fusarium toxins (p < 0.001) (Fig. 9E). In DON-
toxins (p < 0.05) (Fig. 11A). The highest expression of cyto c was
ZEN mix, the expression of caspase-3 was significantly higher than that
observed in DON-ZEN-FB1 (2.41 ± 0.07), and it was significantly
these two mycotoxin alone (p < 0.01) (Fig. 9B). The expression of
higher than the expression in any of the mixture of two Fusarium toxins
caspase-3 was significantly increased in DON-FB1 mix compared to FB1
(p < 0.01) (Fig. 11E). The expression of cyto c in DON-ZEN mix was
alone (p < 0.05) (Fig. 9C). Treatment with 0.5 mM NAC significantly
significantly higher than in the individual mycotoxin treatment
decreased the mRNA expression of caspase-3 in DON-ZEN and DON-
(p < 0.01) (Fig. 11B). The expression of cyto c significantly increased
ZEN-FB1 (p < 0.05) (Fig. 9F).
in DON-FB1 mix compared to DON alone (p < 0.05) (Fig. 11C). The
expression of cyto c significantly increased in ZEN-FB1 mix compared to
3.6.4. Caspase-9 FB1 alone (p < 0.01) (Fig. 11D). Treatment with 0.5 mM NAC sig-
Similar to caspase-3 mRNA expression, caspase-9 mRNA levels were nificantly decreased the mRNA expression of cyto c in DON-ZEN and
significantly higher after treatment with any individual or mixture of DON-ZEN-FB1 treatments (p < 0.05) (Fig. 11F).
Fusarium toxins (p < 0.05) (Fig. 10A). The highest expression of

33
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 5. Individual and combined effects of DON, ZEN and


FB1 and the protective role of NAC on ROS production in
PK15 cells. Cells were exposed to individual or combina-
tions of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and
NAC (0.5 mM) for 24 h. The results are expressed as a
percentage of the control. Bars represent mean ± SD ob-
tained from measurements conducted in triplicate. #, ##,
### means significantly different from control at
p < 0.05, p < 0.01 and p < 0.001, respectively (A). *,
**, *** means significant differences between two con-
nected treatments at p < 0.05, p < 0.01 and p < 0.001,
respectively (B–F).

3.6.6. P53 difference of Bcl-2 and Caspase-3 between individual and combined
Treatments with 20 μM ZEN, 10 μM FB1 and any mixtures of toxin treatment were observed in DON-FB1 mix and DON-ZEN, re-
Fusarium toxins also induced a significant up-regulation of P53 spectively (Fig. 13C–E and I–K). Treatment with 0.5 mM NAC could
(p < 0.01) (Fig. 12A). The P53 mRNA was significantly lower in DON- significantly increase Bcl-2 expression in all mycotoxin treatments ex-
ZEN mix relative to ZEN treatment alone (p < 0.001) (Fig. 12B). In cept DON-ZEN-FB1 and decrease Caspase-3 expression in all mycotoxin
addition, the expression of P53 in the DON-FB1 treatment was sig- treatments except 10 μM FB1 (p < 0.05) (Fig. 13G and M).
nificantly higher relative to DON treatment alone (p < 0.001)
(Fig. 12C). Compared to the individual mycotoxin treatments, P53 4. Discussion
mRNA expression in the ZEN-FB1 treatment was significantly up-regu-
lated (p < 0.001) (Fig. 12D). While, the P53 expression in the DON- Fusarium toxins are extremely toxic to rapidly dividing cells leading
ZEN-FB1 treatment was significantly higher than DON-ZEN and DON- to hepatotoxicity, nephrotoxicity, neurotoxicity, immunotoxicity, and
FB1 (p < 0.01) (Fig. 12E). Treatment with 0.5 mM NAC could sig- reproductive toxicity in the hosts. Several researchers have discussed
nificantly decrease the mRNA expression of P53 in DON-ZEN and ZEN- the individual effects of DON, ZEN or FB1 using different in vitro models
FB1 (p < 0.05) (Fig. 12F). including intestinal porcine epithelial cell lines (IPEC) (Dänicke et al.,
2010), Chinese Hamster Ovary (CHO-K1) cell line (Ferrer et al., 2009)
3.7. Influences of Fusarium toxins and NAC on the Bcl-2 and Caspase-3 and human and pig lymphocytes (Mwanza et al., 2009). However, the
expression reports on the combined effects of DON, ZEN and FB1 on kidney cells
are limited. In the present study, we have shown the nephrotoxic ac-
To analyze the effect of Fusarium toxins and NAC on the expression tivity of DON, ZEN and FB1 using PK15 cell line.
of Bcl-2 and Caspase-3, PK15 cells were treated with individual or
combined Fusarium toxins or/and NAC for 24 h followed by Western 4.1. Effects of Fusarium toxins and NAC on cell viability
blot analysis. Compare to control, all combined treatments and 0.25 μM
DON significantly increased Caspase-3 levels, while only 0.25 μM DON Mycotoxin has been proven to be cytotoxic. Similar to those re-
and DON-ZEN-FB1 mix significantly decreased Bcl-2 levels (Fig. 13B ported by Kouadio et al. (2005), DON, ZEN and FB1 decreased the
and H). And for the mixture of two Fusarium toxins, significant viability of PK15 cells in a dose dependent manner. However, the

34
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 6. Individual and combined effects of DON, ZEN and FB1 and
the protective role of NAC on apoptosis in PK15 cells. Cells were
exposed to individual or combinations of DON (0.25 μM), ZEN
(20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are
expressed as percentage. Bars represent mean ± SD of three parallel
measurements. #, ##, ### means significantly different from
control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *,
**, *** means significant differences between two connected treat-
ments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

35
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 7. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on relative expression of Bax mRNA of PK15 cells. Cells were exposed to individual or
combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as a percentage of the control. Each experimental point represents
the means ( ± SD) obtained with four parallel cultures. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means
significant differences between two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

sensitivity of different cells to the same mycotoxin was not the same. A et al., 2010). After ZEN treatment, cell proliferation was reduced in a
significant decrease in cell viability was observed after 24 h treatment dose-dependent manner as attested by the MTT assay with different
at DON concentrations of 0.25 μM, ZEN concentrations of 20 μM and IC50 values (100 μM on human hepatoma cells (HepG2), 79.40 μM on
FB1 concentrations of 10 μM. In addition, the IC50 values by the MTT CHO-K1) (Ayed-Boussema et al., 2008; Ferrer et al., 2009). After a 48 h
assay were 2.08 μM, 210.7 μM and 196.1 μM for DON, ZEN and FB1, treatment of FB1, the viability of rat spleen mononuclear cells was
respectively. These values were not completely consistent with previous significantly decreased at 40 μM, and had an IC50 value of 230 μM
reports discussed below. One in vitro study indicated that the applica- (Mary et al., 2012). The reason for these differences insensitivity to
tion of DON concentrations of 500 ng/ml or higher decreased the via- DON, ZEN and FB1 among various animal species may be explained by
bility of intestinal porcine epithelial cells in a dose dependent manner differences in the modes of action and the metabolic biotransformation
after 48 h and 72 h incubations (Diesing et al., 2011). The IC50 values of these toxins (Wan et al., 2013). To study the effects of subcytotoxic
were estimated at 1.2, 1.3 and 3.0 μM for porcine peripheral blood DON, ZEN and FB1 on cell injury, concentrations close to the IC10 of the
mononuclear cells (PBMC), IPEC-1 and IPEC-J2, respectively (Dänicke Fusarium toxins [DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM)] were

36
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 8. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on the relative expression in Bcl-2 mRNA of PK15 cells. Cells were exposed to individual or
combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as a percentage of the control. Bars represent mean ± SD of four
parallel measurements. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between
two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

chosen for subsequent experiments. 4.2. Effects of individual and combined Fusarium toxins and NAC on
Simultaneously, the cells were incubated with DON (0.25 μM), ZEN oxidative stress
(20 μM) or FB1 (10 μM) and NAC (0.5, 1.0 and 2.0 mM) for 24 h to
assess the cytoprotective effect of antioxidant NAC. The decreased cell The extent of the oxidative damage on biomolecules reflects the
viability induced Fusarium toxins was significantly prevented by NAC actual balance between pro- and antioxidant activities along with the
(Fig. 1D). Also, the 2.0 mM NAC did not shown a higher cell viability effectiveness of cell-damage repair mechanisms (Mary et al., 2012). To
than 0.5 mM NAC (p > 0.05), demonstrating that NAC should be at a the best of our knowledge, enzymes, comprising of GR and SOD, etc, are
proper concentration for its positive role (He et al., 2012). a source of protection against oxidative stress. The GR enzyme works to
reduce GSSG to GSH to scavenge excess hydrogen peroxide and SOD
enzymes catalyze the dismutation of superoxide radical into hydrogen
peroxide (H2O2) and molecular oxygen (O2) and consequently present
an important defense mechanism against superoxide radical toxicity

37
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 9. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on the relative expression of caspase-3 mRNA in PK15 cells. Cells were exposed to individual
or combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as a percentage of the control. Bars represent mean ± SD of four
parallel measurements. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between
two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

(Assady et al., 2011). Our results showed that the treatments with in- 2011; Li et al., 2014).
dividual DON and most mixtures could decrease the activities of GR and The generation of ROS is involved in important physiological
SOD to different degrees, while the lowest observed in DON-ZEN-FB1. functions of aerobic organisms. However, the formation of ROS in ex-
Moreover, the GR and SOD activity of the combined toxin treatment cess, can lead to a disturbance in metabolic pathways, the depletion of
was lower than that of the individual toxin, except for ZEN-FB1, sug- cellular antioxidants and damage to macromolecules, including DNA,
gesting that they had worse O2− radical scavenging ability. Similar proteins and lipids, creating oxidative injury that results in the reduc-
results correlating to antioxidative system activity induced by in- tion of differentiation and proliferation or the promotion of the cell
dividual mycotoxin were also reported by other researchers (Dinu et al., death process (Ferrer et al., 2009). ROS include both free radicals, such

38
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 10. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on relative expression of Caspase-9 mRNA in PK15 cells. Cells were exposed to individual or
combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as a percentage of the control. Bars represent mean ± SD of four
parallel measurements. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between
two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

as superoxide (O2−), nitric oxide (NO) and hydroxyl radical (OH) and to decreased antioxidant enzyme activities, which increased H2O2 le-
other molecular species, such as hydrogen peroxide (H2O2) and per- vels and membrane damage. In addition, the increases of MDA levels
oxynitrite (ONOO-). ROS can induce peroxidation of membrane lipids. might be due to the elevation of ROS level observed in our study. 10 μM
The degree of oxidative damage to lipids is often estimated by mea- FB1 resulted in almost unchanged activities of GR and SOD but sig-
suring the MDA levels, one of the peroxidation end-products. Our re- nificantly increased ROS and MDA generation, which was perhaps due
sults demonstrated that individual DON and FB1 and almost all mix- to the action of other enzymes in antioxidant system.
tures could significantly increase ROS and MDA production, and a NAC is a well-established thiol antioxidant which can be used as
greater enhancement was observed in the combination treatments. alleviate glutathione (GSH), which functions as a reactive oxygen in-
These are in agreement with some previous studies (Yang et al., 2014; termediate scavenger (Nazıroğlu et al., 2014). High glucose levels
Stockmann-Juvala et al., 2004a, 2004b; Ferrer et al., 2009; Li et al., promoted apoptosis by affecting mitochondria-dependent caspase ac-
2014). These results strongly suggest that the ROS generation was due tivity through elevation of ROS production could also be reversed by

39
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 11. Individual and combined effects of DON, ZEN and FB1 and the protective role of NAC on relative expression of cyto c mRNA in PK15 cells. Cells were exposed to individual or
combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC (0.5 mM) for 24 h. The results are expressed as a percentage of the control. Bars represent mean ± SD of four
parallel measurements. #, ##, ### means significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively (A). *, **, *** means significant differences between
two connected treatments at p < 0.05, p < 0.01 and p < 0.001, respectively (B–F).

the antioxidant NAC (Park et al., 2015). Our study showed that ad- 4.3. Effects of individual and combined Fusarium toxins and NAC on
ministration of NAC tends to restore the porcine kidney cell peroxides apoptosis
and ROS induced by partial Fusarium toxin, primarily in treatments of
larger difference. These results suggested that the combination of sub- To investigate whether DON, ZEN and FB1 impact PK15 cells death
cytotoxic Fusarium toxin induced stronger oxidative stress, and some of process, we detected the level of apoptosis and the expression of genes
them could be relieved by NAC. associated with apoptosis. Apoptotic cell death is a genetically pro-
grammed mechanism that allows the cells to commit suicide. It has
been reported that DON was able to induce apoptosis, which is ac-
companied by the up-regulation of apoptosis-related genes including
caspase-3, caspase-8, caspase-9, and AIFM1 in DF-1 cells (Li et al.,

40
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 12. Individual and combined effects of DON, ZEN and


FB1 and the protective role of NAC on relative expression of
P53 mRNA in PK15 cells. Cells were exposed to individual
or combinations of DON (0.25 μM), ZEN (20 μM) and FB1
(10 μM) and NAC (0.5 mM) for 24 h. The results are ex-
pressed as a percentage of the control. Bars represent
mean ± SD of four parallel measurements. #, ##, ###
means significantly different from control at p < 0.05,
p < 0.01 and p < 0.001, respectively (A). *, **, ***
means significant differences between two connected
treatments at p < 0.05, p < 0.01 and p < 0.001, re-
spectively (B–F).

2014). Moreover, it has been demonstrated previously that DON re- P53, Bax, Cyto c, Caspase-3 and -9 and down-regulated Bcl-2 mRNA
duced the mitochondrial transmembrane potential, up-regulated the expression except FB1, although some differences were not significant.
concentrations of p53, Bax, and Bak-1, and down-regulated the con- The mRNA expression of Bax and Bcl-2 didn't change after treated with
centrations of Bcl-2 (Bensassi et al., 2009, 2012; Ren et al., 2015). ZEN 10 μM FB1. Moreover, the protein expression of Bcl-2 in treatments of
was found to induce apoptosis in human leukemic HL-60 and U937 cells DON, ZEN, DON-ZEN and DON-ZEN-FB1 decreased and Caspase-3
via the endoplasmic stress and mitochondrial pathway such as the ac- protein expression of all mycotoxin treatments increased to some ex-
tivation of mitochondrial release of cyto c, transmembrane potential tent. As Bcl-2 family member, Bcl-2 acts to inhibit cyto c translocation
reduction, activation of caspase-3 and -8, production of reactive oxygen and Bax is a potent proapoptotic molecule that, on translocation from
species and an altered expression level of 22 membrane proteins such as cytosol to mitochondria, triggers the activation of terminal caspases by
apoptosis inducing factor and the endoplasmic reticulum (ER) stress increasing mitochondrial membrane permeability and resulting in the
proteins including endoplasmic reticulum protein 29 (ERp29), 78 kDa release of apoptosis-promoting factors, including cyto c. The release of
glucose-regulated protein, heat shock protein 90 and calreticulin cyto c may trigger the interaction of Apaf1, a mammalian CED-4
(Banjerdpongchai et al., 2010). Furthermore, p53 and JNK/p38 played homolog, and Caspase-9, which in turn results in the activation of
a key role in the ZEN induced cytotoxicity in mouse macrophage Caspase-9. Activated Casp9 then cleaves and activates pro-Caspase-3,
RAW264.7 cells via apoptosis-inducing factor- (AIF) and ROS-mediated an event that leads to the cleavage of other death substrates, cellular
signaling (Yu et al., 2011). For FB1, the increase in caspase-3 activity and nuclear morphological changes, and ultimately causes cell death (Li
preceding changes in the apoptotic index showed that the cytotoxic and et al., 1998). The tumor suppressor protein p53 is known to function as
apoptotic effects on PK15 cells were in a concentration- and time-de- a key player in mediating apoptosis through modulating the Bax to Bcl-
pendent manner (Klarić et al., 2008). The involvement of the TNF 2 ratio and the regulation of mitochondrial ROS generation (Gosslau
signal transduction pathway was another pathway of FB1 induced et al., 2005). Taken together, our results suggested that Fusarium toxins
apoptosis, and the tumor suppressor gene p53 was not required (Jones induced apoptosis through the mitochondria-dependent channel, and
et al., 2001). In this study, after treatments with Fusarium toxins, alone Bcl-2 and Bax were not essential pathways for FB1 to induce apoptosis.
or combined, significant increase in apoptosis were observed. In gen- It has been suggested that NAC could inhibit apoptosis in some
eral, all mycotoxin treatments up-regulated the mRNA expression of systems. Anuradha et al. have reported that antioxidants NAC protected

41
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Fig. 13. Individual and combined effects of DON, ZEN and FB1 and the protective role of
NAC on protein expression of Bcl-2 and Caspase-3 in PK15 cells. Cells were exposed to
individual or combinations of DON (0.25 μM), ZEN (20 μM) and FB1 (10 μM) and NAC
(0.5 mM) for 24 h. This graphical representation also includes the relative density of
protein levels in PK15 cells, which were normalized to those of β-actin. Bars represent
mean ± SD obtained from measurements conducted in triplicate. #, ##, ### means
significantly different from control at p < 0.05, p < 0.01 and p < 0.001, respectively
(B and H). *, **, *** means significant differences between two connected treatments at
p < 0.05, p < 0.01 and p < 0.001, respectively (C-G and I-M).

the cells from loss of mitochondrial membrane potential and there was
no cytochrome c exit or Bcl-2 downregulation (Anuradha et al., 2001).
In the present study, NAC supplement reversed the Fusarium toxins-
induced apoptosis to a certain extent, particularly in treatments of
DON, FB1, ZEN-FB1 and DON-ZEN-FB1, although the difference was not
significant. For expression of genes associated with mitochondrial
apoptotic pathways, NAC partially regulated the change induced by
Fusarium toxin, most of which were combined toxins. These results were
consistent to those of oxidative stress.

5. Conclusion

In conclusion, the results of this study confirmed that individual and


combined Fusarium toxins induced different degrees of oxidative stress
response and apoptosis, with greater change in combinations and most
potent toxic effect in DON-ZEN-FB1 mix. The expression of mitochon-
drial apoptotic pathway-related genes species were regulated differen-
tially by Fusarium toxins, and these findings suggest that mitochondrial
pathway played an important role in Fusarium toxins induced apoptosis.
Moreover, NAC reversed the oxidative damage and apoptosis of
Fusarium toxins to some extent, especially in combinations. Overall,
these findings suggest that antioxidant such as NAC are important for
treating and preventing the toxicity of Fusarium toxins.

Conflict of interest

The authors declare no competing financial interests.

Acknowledgments

This work was supported by the National Key R & D Program


(2016YFD0501207), the China Agriculture Research System (CARS-35)
and the Key Laboratory of Animal Common Disease Prevention Open
Project of Heilongjiang Province (120103).

Transparency document

Transparency document related to this article can be found online at


http://dx.doi.org/10.1016/j.fct.2017.10.057.

References

Anuradha, C.D., Kanno, S., Hirano, S., 2001. Oxidative damage to mitochondria is a
preliminary step to caspase-3 activation in fluoride-induced apoptosis in HL-60 cells.
Free Radic. Biol. Med. 31, 367.
Asevedo, E., Mendes, A.C., Berk, M., Brietzke, E., 2014. Systematic review of N-acet-
ylcysteine in the treatment of addictions. Rev. Bras. Psiquiatr. 36, 168–175.
Assady, M., Farahnak, A., Golestani, A., Esharghian, M., 2011. Superoxide dismutase
(SOD) enzyme activity assay in Fasciola spp. parasites and liver tissue extract. Iran. J.
Parasitol. 6, 17.
Ayed-Boussema, I., Bouaziz, C., Rjiba, K., Valenti, K., Laporte, F., Bacha, H., Hassen, W.,
2008. The mycotoxin Zearalenone induces apoptosis in human hepatocytes (HepG2)
via p53-dependent mitochondrial signaling pathway. Toxicol. In Vitro 22,
1671–1680.
Banjerdpongchai, R., Kongtawelert, P., Khantamat, O., Srisomsap, C.,
Chokchaichamnankit, D., Subhasitanont, P., Svasti, J., 2010. Mitochondrial and en-
doplasmic reticulum stress pathways cooperate in zearalenone-induced apoptosis of
human leukemic cells. J. Hematol. Oncol. 3, 50.
Bensassi, F., El Golli-Bennour, E., Abid-Essefi, S., Bouaziz, C., Hajlaoui, M.R., Bacha, H.,
2009. Pathway of deoxynivalenol-induced apoptosis in human colon carcinoma cells.

42
W. Zhang et al. Food and Chemical Toxicology 111 (2018) 27–43

Toxicology 264, 104–109. Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-
Bensassi, F., Gallerne, C., El Dein, O.S., Lemaire, C., Hajlaoui, M.R., Bacha, H., 2012. time quantitative PCR and the 2− ΔΔCT method. Methods 25, 402–408.
Involvement of mitochondria-mediated apoptosis in deoxynivalenol cytotoxicity. Martins, H.M., Guerra, M.M., Bernardo, F., 2006. Zearalenone, deoxynivalenol and fu-
Food Chem. Toxicol. 50, 1680–1689. monisins in mixed-feed for laying hens. Mycotoxin Res. 22, 206.
Collins, T.F., Sprando, R.L., Black, T.N., Olejnik, N., Eppley, R.M., Hines, F.A., Rorie, J., Mary, V.S., Theumer, M.G., Arias, S.L., Rubinstein, H.R., 2012. Reactive oxygen species
Ruggles, D.I., 2006. Effects of deoxynivalenol (DON, vomitoxin) on in utero devel- sources and biomolecular oxidative damage induced by aflatoxin B1 and fumonisin
opment in rats. Food Chem. Toxicol. 44, 747–757. B1 in rat spleen mononuclear cells. Toxicology 302, 299–307.
Dänicke, S., Hegewald, A.-K., Kahlert, S., Kluess, J., Rothkötter, H.-J., Breves, G., Döll, S., Monbaliu, S., Poucke, C.V., Detavernier, C., Dumoulin, F., Velde, M.V.D., Schoeters, E.,
2010. Studies on the toxicity of deoxynivalenol (DON), sodium metabisulfite, DON- Dyck, S.V., Averkieva, O., Peteghem, C.V., Saeger, S.D., 2010. Occurrence of myco-
sulfonate (DONS) and de-epoxy-DON for porcine peripheral blood mononuclear cells toxins in feed as analyzed by a multi-mycotoxin LC-MS/MS method. J. Agric. Food
and the Intestinal Porcine Epithelial Cell lines IPEC-1 and IPEC-J2, and on effects of Chem. 58, 66–71.
DON and DONS on piglets. Food Chem. Toxicol. 48, 2154–2162. Mosmann, T., 1983. Rapid colorimetric assay for cellular growth and survival: application
Diaz, D.E., 2005. The Mycotoxin Blue Book. Nottingham University Press. to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63.
Diesing, A.-K., Nossol, C., Panther, P., Walk, N., Post, A., Kluess, J., Kreutzmann, P., Mwanza, M., Kametler, L., Bonai, A., Rajli, V., Kovacs, M., Dutton, M.F., 2009. The cy-
Dänicke, S., Rothkötter, H.-J., Kahlert, S., 2011. Mycotoxin deoxynivalenol (DON) totoxic effect of fumonisin B1 and ochratoxin A on human and pig lymphocytes using
mediates biphasic cellular response in intestinal porcine epithelial cell lines IPEC-1 the Methyl Thiazol Tetrazolium (MTT) assay. Mycotoxin Res. 25, 233–238.
and IPEC-J2. Toxicol. Lett. 200, 8–18. Nazıroğlu, M., Şenol, N., Ghazizadeh, V., Yürüker, V., 2014. Neuroprotection induced by
Dinu, D., Bodea, G.O., Ceapa, C.D., Munteanu, M.C., Roming, F.I., Serban, A.I., N-acetylcysteine and selenium against traumatic brain injury-induced apoptosis and
Hermenean, A., Costache, M., Zarnescu, O., Dinischiotu, A., 2011. Adapted response calcium entry in hippocampus of rat. Cell. Mol. Neurobiol. 34, 895–903.
of the antioxidant defense system to oxidative stress induced by deoxynivalenol in Park, J.H., Kang, S.-S., Kim, J.Y., Tchah, H., 2015. The antioxidant N-acetylcysteine in-
Hek-293 cells. Toxicon 57, 1023–1032. hibits inflammatory and apoptotic processes in human conjunctival epithelial cells in
Ferrer, E., Juan-García, A., Font, G., Ruiz, M., 2009. Reactive oxygen species induced by a high-glucose environment effect of NAC on conjunctiva in a high-glucose en-
beauvericin, patulin and zearalenone in CHO-K1 cells. Toxicol. In Vitro 23, vironment. Invest. Ophthalmol. Vis. Sci. 56, 5614–5621.
1504–1509. Pestka, J.J., 2010. Deoxynivalenol: mechanisms of action, human exposure, and tox-
Flora, S. De, Izzotti, A., D'Agostini, F., Balansky, R.M., 2001. Mechanisms of N-acet- icological relevance. Arch. Toxicol. 84, 663–679.
ylcysteine in the prevention of DNA damage and cancer, with special reference to Pestka, J.J., Amuzie, C.J., 2008. Tissue distribution and proinflammatory cytokine gene
smoking-related end-points. Carcinogenesis 22, 999–1013. expression following acute oral exposure to deoxynivalenol: comparison of weanling
Garrido, C.E., Pezzani, C.H., Pacin, A., 2012. Mycotoxins occurrence in Argentina's maize and adult mice. Food Chem. Toxicol. 46, 2826–2831.
( Zea mays L.), from 1999 to 2010. Food Control 25, 660–665. Poersch, A.B., Trombetta, F., Souto, N.S., de Oliveira Lima, C., Braga, A.C.M.,
Gosslau, A., Chen, M., Ho, C., Chen, K.Y., 2005. A methoxy derivative of resveratrol Dobrachinski, F., Ribeiro, L.R., Soares, F.A.A., Fighera, M.R., Royes, L.F.F., 2015.
analogue selectively induced activation of the mitochondrial apoptotic pathway in Fumonisin B1 facilitates seizures induced by pentylenetetrazol in mice. Neurotoxicol.
transformed fibroblasts. Br. J. Cancer 92, 513. Teratol. 51, 61–67.
He, S.J., Hou, J.F., Dai, Y.Y., Zhou, Z.L., Deng, Y.F., 2012. N-acetyl-cysteine protects Ren, Z., Wang, Y., Deng, H., Deng, Y., Deng, J., Zuo, Z., Wang, Y., Peng, X., Cui, H., Shen,
chicken growth plate chondrocytes from T-2 toxin-induced oxidative stress. J. Appl. L., 2015. Deoxynivalenol induces apoptosis in chicken splenic lymphocytes via the
Toxicol. 32, 980–985. reactive oxygen species-mediated mitochondrial pathway. Environ. Toxicol.
Hu, J., Xu, M., Dai, Y., Ding, X., Xiao, C., Ji, H., Xu, Y., 2016. Exploration of Bcl-2 family Pharmacol. 39, 339–346.
and caspases-dependent apoptotic signaling pathway in Zearalenone-treated mouse Samuni, Y., Goldstein, S., Dean, O.M., Berk, M., 2013. The chemistry and biological ac-
endometrial stromal cells. Biochem. Biophys. Res. Commun. 476, 553–559. tivities of N-acetylcysteine. Biochim. Biophys. Acta Gen. Subj. 1830, 4117–4129.
Hussein, H.S., Brasel, J.M., 2001. Toxicity, metabolism, and impact of mycotoxins on Stockmann-Juvala, H., Mikkola, J., Naarala, J., Loikkanen, J., Elovaara, E., Savolainen,
humans and animals. Toxicology 167, 101–134. K., 2004a. Fumonisin B 1-induced toxicity and oxidative damage in U-118MG glio-
Jayalakshmi, K., Sairam, M., Singh, S., Sharma, S., Ilavazhagan, G., Banerjee, P., 2005. blastoma cells. Toxicology 202, 173–183.
Neuroprotective effect of N-acetyl cysteine on hypoxia-induced oxidative stress in Stockmann-Juvala, H., Mikkola, J., Naarala, J., Loikkanen, J., Elovaara, E., Savolainen,
primary hippocampal culture. Brain Res. 1046, 97–104. K., 2004b. Oxidative stress induced by fumonisin B1 in continuous human and rodent
Jia, Z., Yin, S., Liu, M., Zhang, Y., Gao, R., Shi, B., Shan, A., Chen, Z., 2015. Modified neural cell cultures. Free Radic. Res. 38, 933–942.
halloysite nanotubes and the alleviation of kidney damage induced by dietary zear- Wan, L.Y.M., Turner, P.C., El-Nezami, H., 2013. Individual and combined cytotoxic ef-
alenone in swine. Food Addit. Contam. Part A 32, 1312–1321. fects of Fusarium toxins (deoxynivalenol, nivalenol, zearalenone and fumonisins B1)
Jones, C., Ciacci-Zanella, J.R., Zhang, Y., Henderson, G., Dickman, M., 2001. Analysis of on swine jejunal epithelial cells. Food Chem. Toxicol. 57, 276–283.
fumonisin B1-induced apoptosis. Environ. Health Perspect. 109, 315. Wentzel, J.F., Lombard, M.J., Du, P.L., Zandberg, L., 2016. Evaluation of the cytotoxic
Kinser, S., Li, M., Jia, Q., Pestka, J.J., 2005. Truncated deoxynivalenol-induced splenic properties, gene expression profiles and secondary signalling responses of cultured
immediate early gene response in mice consuming (n-3) polyunsaturated fatty acids. cells exposed to fumonisin B1, deoxynivalenol and zearalenone mycotoxins. Arch.
J. Nutr. Biochem. 16, 88–95. Toxicol. 1–18.
Klarić, M.Š., Rumora, L., Ljubanović, D., Pepeljnjak, S., 2008. Cytotoxicity and apoptosis Yang, L., Yang, W., Feng, Q., Huang, L., Zhang, G., Liu, F., Jiang, S., Yang, Z., 2016.
induced by fumonisin B1, beauvericin and ochratoxin A in porcine kidney PK15 cells: Effects of purified zearalenone on selected immunological measurements of blood in
effects of individual and combined treatment. Arch. Toxicol. 82, 247–255. post-weaning gilts. Anim. Nutr. 2, 142–148.
Kouadio, J., Moukha, S., Brou, K., Gnakri, D., 2013. Lipid metabolism disorders, lym- Yang, W., Yu, M., Fu, J., Bao, W., Wang, D., Hao, L., Yao, P., Nüssler, A.K., Yan, H., Liu, L.,
phocytes cells death, and renal toxicity induced by very low levels of deoxynivalenol 2014. Deoxynivalenol induced oxidative stress and genotoxicity in human peripheral
and fumonisin b1 alone or in combination following 7 days oral administration to blood lymphocytes. Food Chem. Toxicol. 64, 383–396.
mice. Toxicol. Int. 20, 218. Yin, S., Meng, Q., Zhang, B., Shi, B., Shan, A., Li, Z., 2015. Alleviation of zearalenone
Kouadio, J.H., Mobio, T.A., Baudrimont, I., Moukha, S., Dano, S.D., Creppy, E.E., 2005. toxicity by modified halloysite nanotubes in the immune response of swine. Food
Comparative study of cytotoxicity and oxidative stress induced by deoxynivalenol, Addit. Contam. Part A 32, 87–99.
zearalenone or fumonisin B1 in human intestinal cell line Caco-2. Toxicology 213, Yu, J.-Y., Zheng, Z.-H., Son, Y.-O., Shi, X., Jang, Y.-O., Lee, J.-C., 2011. Mycotoxin
56–65. zearalenone induces AIF-and ROS-mediated cell death through p53-and MAPK-de-
Li, Honglin, Zhu, Hong, Xu, Chijie, Yuan, Junying, 1998. Cleavage of BID by caspase 8 pendent signaling pathways in RAW264. 7 macrophages. Toxicol. In Vitro 25,
mediates the mitochondrial damage in the fas pathway of apoptosis. Cell 94, 491. 1654–1663.
Li, D., Ye, Y., Lin, S., Deng, L., Fan, X., Zhang, Y., Deng, X., Li, Y., Yan, H., Ma, Y., 2014. Zhang, Y., Gao, R., Liu, M., Shi, B., Shan, A., Cheng, B., 2015. Use of modified halloysite
Evaluation of deoxynivalenol-induced toxic effects on DF-1 cells in vitro: cell-cycle nanotubes in the feed reduces the toxic effects of zearalenone on sow reproduction
arrest, oxidative stress, and apoptosis. Environ. Toxicol. Pharmacol. 37, 141–149. and piglet development. Theriogenology 83, 932–941.

43

You might also like