N-acetylcysteine ameliorates repetitive/stereotypic behavior due to its antioxidant properties without activation of the canonical Wnt pathway in a valproic acid-induced rat model of autism

  • Authors:
    • Yinghua Zhang
    • Weigang Cui
    • Qianqian Zhai
    • Tianran Zhang
    • Xiaojun Wen
  • View Affiliations

  • Published online on: June 15, 2017     https://doi.org/10.3892/mmr.2017.6787
  • Pages: 2233-2240
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

N-acetylcysteine (NAC) is widely used as an antioxidant, and previous studies have suggested that it may have potential as an alternative therapeutic strategy for the treatment of patients with autism. However, the exact effects of NAC administration on the development of autism, as well as the molecular mechanisms underlying its actions, have yet to be fully elucidated. The present study aimed to investigate the effects of NAC on the oxidative status of rats in a valproic acid (VPA)‑induced model of autism, and to examine the involvement of the canonical Wnt signaling pathway in the actions of NAC. Rats exposed to VPA were monitored for behavioral changes, and oxidative stress indicators and key molecules of the canonical Wnt pathway were investigated using colorimetric and western blot analysis, respectively. The present results demonstrated that NAC ameliorated repetitive and stereotypic activity in autism model rats. Furthermore, NAC was revealed to relieve oxidative stress, as demonstrated by the increased glutathione and reduced malondialdehyde levels compared with VPA‑treated rats. However, NAC did not appear to affect the activity of the canonical Wnt signaling pathway. The present findings suggested that the beneficial effects of NAC in autism may be associated with its antioxidative properties, and may not be mediated by the canonical Wnt pathway. However, it may be hypothesized that the canonical Wnt pathway can be indirectly regulated by NAC through the activation of other signaling pathways or upstream factors. Taken together, the present study has contributed to the elucidation of the molecular mechanisms that underlie the actions of NAC in autism, suggesting its potential for the development of novel therapeutic strategies for the treatment of patients with autism.

Introduction

Autism is a complex neurodevelopmental disorder characterized by three central features: Social interaction deficits, language retardation and aberrant stereotypic behavior (1,2). In 2016, the Centers for Disease Control and Prevention reported that 1 in 68 children is diagnosed with autism spectrum disorder in the United States (3). Currently, no effective therapeutic strategies are available to ameliorate the key deficits associated with autism Therefore, the need for the development of novel targeted treatments is urgent for the management of autism, and for improvement of the patients' quality of life.

Previous studies have suggested that oxidative stress may be involved in the development of autism (4,5). N-acetylcysteine (NAC) is a source of the amino acid L-cysteine, and is a precursor in the formation of glutathione (GSH) (6), which is a major intracellular antioxidant within the central nervous system (CNS) (7). In addition to providing cysteine for GSH production, NAC has been revealed to directly scavenge oxygen free radicals through its thiol-reducing group, and has been suggested as a promising neuroprotectant in the CNS (8,9). NAC has been reported to cross the blood brain barrier (10,11), decrease oxidative stress and inflammatory cytokine production, replenish GSH levels, and thus ameliorate autism-associated irritability, including self-injurious behavior (12). Previous placebo-controlled human trials demonstrated that, following administration of NAC, autism-associated irritability was attenuated in pediatric patients with autism (12,13). In addition, two clinical trials demonstrated that the combination of NAC with risperidone significantly alleviated irritability in young patients with autism (14,15). Furthermore, NAC has been suggested to ameliorate additional features of autism, as it was demonstrated to improve social interaction (7,16) and reduce anxiety-like behaviors (16). However, to the best of our knowledge, the effects of NAC on repetitive/stereotypic activity and oxidative stress in autism models, and the molecular mechanisms underlying the effects of NAC in the treatment of autism, have yet to be elucidated.

The canonical Wnt pathway includes the major effector β-catenin and glycogen synthase kinase (GSK)-3β. In the absence of Wnt ligands, β-catenin associates with a cytoplasmic degradation protein complex, leading to proteasomal degradation (17). With secreted Wnt ligands, the canonical Wnt pathway is activated, and the cytoplasmic degradation protein complex is prevented from forming. As a result, β-catenin accumulates in cytoplasm and translocates to the nucleus (18). GSK-3β destabilizes β-catenin by phosphorylating its inhibitory sites, such as Ser33, Ser37, and Thr41 (19). Previous studies have reported that oxidative stress activated the canonical Wnt intracellular signaling pathway during the pathogenesis of various diseases, including preeclampsia (20), diabetic nephropathy (21) and gastric cancer (22). The canonical Wnt pathway has also been reported to be involved in the development of autism (2325). Therefore, elucidating the molecular mechanisms that are involved in the modulation of oxidative processes, and the roles of the Wnt pathway in the pathogenesis of autism, is of critical importance.

The present study aimed to investigate the effects of NAC on autism-like behavioral phenotypes, and to examine the molecular mechanisms underlying its actions in a valproic acid (VPA)-induced animal model of autism. NAC was demonstrated to reverse behavioral abnormalities, including stereotypic behaviors, and its therapeutic effects were associated with a reduction in oxidative stress. However, the molecular mechanisms involved in the actions of NAC did not appear to be associated with the canonical Wnt signaling pathway. The Wnt/β-catenin pathway may be indirectly regulated by NAC through the activation of alternative signaling pathways or upstream factors, and further studies are required to elucidate the mechanisms that may be involved in its actions.

Materials and methods

Animals and experimental groups

Experimental procedures were approved by the Laboratory Animal Care and Use Committee of Xinxiang Medical University, and were in accordance with the guidelines for animal experimentation of the Institutes of Health of Xinxiang Medical University (Xinxiang, China). A total of 60 Sprague-Dawley rats (age, 12 weeks; 40 female, 220–250 g; 20 male, 350–390 g) were purchased from Vital River Laboratories Co., Ltd. (Beijing, China) and were allowed to breed. The rats were kept in a cage with 40–80% relative humidity and a controlled temperature of 24±1°C on a 12-h light/dark cycle (lights off at 19:00). Rats had free access to food and water. The presence of vaginal plugs was used to indicate gestation. For the establishment of the autism model (26,27), female rats were intraperitoneally injected with 600 mg/kg VPA (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) on gestation day 12.5, vehicle-treated rats received physiological saline. The pups were weaned on postnatal day 23 and housed separately. Offspring (23 days old) of VPA-treated mothers were randomly assigned into the following 2 groups: VPA and VPA + NAC (Sigma-Aldrich; Merck KGaA) groups. The 23-day-old-offspring of saline-treated mothers were randomly divided into the following 2 groups: Control and NAC groups. NAC (150 mg/kg) was intraperitoneally administered to rats in the NAC and VPA + NAC groups once daily for 4 weeks. Subsequently, male offspring were used for repetitive/stereotypic-like behavioral testing and then sacrificed by decapitation. Prefrontal cortex (PFC) and hippocampus (HC) samples were isolated and frozen at −80°C for further analysis.

Behavioral testing

Behavioral testing was conducted using an open field test, as previously described (28). Briefly, rats were placed individually in a plywood apparatus (100×100×40 cm). Testing was performed between 9:00 am and 3:00 pm. Two rats were observed simultaneously, with one animal per chamber. The animals were individually placed in the field and allowed to explore it freely for 60 min. Movements were recorded using a digital video camera linked to a computer (Ethovision 3.0; Noldus Information Technology BV, Wageningen, The Netherlands); the mean activity during the 60-min testing session, divided into 6 time bins (10 min each), was then analyzed (EthoVision 3.0; Noldus). Repetitive/stereotypic-like movements (for example run and rotate, nose picking, lip biting, repeated sucking) were measured and analyzed by an observer blind to the treatments.

Oxidative stress assays
Malondialdehyde (MDA) assay

As a marker of lipid peroxidation, the MDA contents were evaluated in brain tissue samples isolated from rats using a commercially available MDA assay kit (cat no. S0131; Beyotime Institute of Biotechnology, Haimen, China), according to the manufacturer's protocol. Briefly, brain tissue sections were homogenized in 0.15 ml thiobarbituric acid (TBA) diluent, 0.05 ml TBA and 3 µl antioxidant then heated and boiled for 15 min. Following centrifugation at 1,000 × g for 10 min at room temperature, the supernatants were collected, and the absorbance of the samples was measured at 532 nm using a microplate reader. MDA contents were expressed as µmol/mg protein.

GSH assay

5,5′-Dithiobis-(2-nitrobenzoic acid) was used to detect intracellular GSH levels with a Total GSH assay kit (cat no. S0052; Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Briefly, brain tissue sections were homogenized with glass homogenizer and incubated with the assay solution for 5 min at 25°C. The concentration of GSH was measured spectrophotometrically at 412 nm using a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA). GSH contents were expressed as µmol/mg protein.

Western blot analysis

Cytoplasmic and nuclear proteins were isolated from brain tissue samples using a Nuclear and Cytoplasmic Protein Extraction kit (cat no. P0028; Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Tissue samples (60 mg) were lysed in a 200 µl solution containing cytoplasmic protein extraction agent A and B (20:1) supplemented with phenylmethylsulfonyl fluoride (PMSF). Following tissue homogenization, the samples were incubated for 15 min on ice. Then the lysates were centrifuged for 5 min at 1,500 × g at 4°C, and the supernatants, consisting of the cytoplasmic fraction, were immediately frozen at −80°C for subsequent experiments. The pellets were resuspended in nuclear protein extraction agent B supplemented with PMSF. Following vortexing for 5 sec, the mixture was incubated on ice for 1 min and centrifuged for 10 min at 12,000 × g at 4°C to obtain supernatants containing nuclear proteins. Proteins were quantified with a bicinchoninic acid assay kit (cat no. P0010; Beyotime Institute of Biotechnology). Equal amounts of extracted protein samples (20–50 µg) were separated by 10% SDS-PAGE and transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% bovine serum albumin (cat no. A7030; Sigma-Aldrich; Merck KGaA) or fat-free milk in TBS containing 0.1% Tween-20 for 1 h at room temperature. Membranes were then incubated with primary antibodies overnight at 4°C. Following incubation with horseradish peroxidase-conjugated goat anti-rabbit antibodies (cat no. 111035003; Jackson Immunoresearch Laboratories, Inc., West Grove, PA, USA) at a 1:5,000 dilution for 1 h at room temperature, the protein bands were visualized using an enhanced chemiluminescence kit (EMD Millipore). Blots were semi-quantified using densitometric analysis with ImageJ software (version 1.44p; National Institutes of Health, Bethesda, MD, USA). GAPDH (1:5,000; cat no. HRP-60004; ProteinTech Group, Inc., Chicago, IL, USA) or histone H3 (1:1,000; cat no. AF0009; Beyotime Institute of Biotechnology,) were used as the internal control for cytoplasmic and nuclear proteins, respectively. The following primary antibodies were used in the present study: Anti-glycogen synthase kinase (GSK)-3β (1:1,000; cat no. 9315), anti-phosphorylated (p)-GSK-3β (Ser-9; 1:500; cat no. 9336) (both from Cell Signaling Technology, Inc., Danvers, MA, USA), anti-β-catenin (1:1,000; cat no. sc-7199; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) and anti-p-β-catenin (Ser-33/37/Thr41; 1:500; cat no. 9561; Cell Signaling Technology, Inc.).

Statistical analysis

Data are expressed as the mean ± standard error of the mean, for 5 repeated experiments. The statistical significance of the differences between groups was assessed using a one-way analysis of variance (ANOVA) followed by Fisher's least-significant difference post hoc correction for multiple comparisons. The statistical significance of the differences between correlated samples was assessed using a repeated measures two-way ANOVA followed by Fisher's least-significant difference post hoc test for multiple comparisons. Statistical analyses were performed using SPSS software version 16.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of NAC administration on repetitive/stereotypic behavior in VPA-exposed rats

Previous studies have reported that treatment with NAC ameliorated behavioral disorders in children with autism (13,15,29,30). Therefore, the present study investigated the effects of NAC on autism-associated behavioral disorders in a VPA-induced rat model of autism.

An open field test was used to assess repetitive/stereotypic behavior in VPA-exposed male rats. The present results demonstrated that the distance traveled by rats in the VPA group was significantly increased compared with the control group between 0 and 40 min of testing (Fig. 1A). The distance traveled by rats in the NAC and NAC + VPA groups was comparable to that of rats in the control group, and was significantly decreased compared with the VPA group across the 0–20, 30–40 and 50–60 min testing durations (Fig. 1A). In addition, VPA-treated rats spent significantly more time engaged in repetitive/stereotypic activities, and their number of repetitive/stereotypic movements was increased compared with the control group. VPA-treated rats were overactive between 0 and 40 min, and between 50 and 60 min of testing (Fig. 1B), and they spent more time occupied in repetitive/stereotypic movements between 0 and 40 min of testing compared with the control rats (Fig. 1C). Conversely, rats in the VPA + NAC group were not overactive and spent significantly less time occupied in repetitive/stereotypic movements between 0 and 40 min of testing (Fig. 1C). Furthermore, they exhibited significantly fewer repetitive/stereotypic movements throughout the duration of the experiment compared with rats in the VPA group (Fig. 1B). VPA + NAC-treated rats exhibited a similar behavior to rats in the control group throughout the 60-min duration of the experiment.

Effects of NAC on oxidative stress in VPA-exposed rats

NAC has previously been demonstrated to ameliorate autism-associated social interaction deficits and anxiety-related behaviors (16), and the present results suggested it could also attenuate repetitive/stereotypic movements. Therefore, the molecular mechanisms underlying the effects of NAC on autism-like phenotypes were further investigated, and its restorative actions on abnormal oxidative homeostasis were revealed. MDA and GSH contents were measured in brain tissue samples isolated from VPA-exposed rats, in order to examine the effects of NAC on lipid peroxidation and total antioxidative capacity in the brain. As demonstrated in Fig. 2, MDA levels were significantly increased, whereas GSH levels were significantly decreased, in the prefrontal cortex and hippocampus of VPA-treated rats compared with controls. Notably, treatment with NAC was revealed to significantly downregulate cortical and hippocampal MDA levels, and upregulate GSH levels compared with VPA-exposed rats (Fig. 2). No significant differences were detected in MDA and GSH levels between rats in the VPA + NAC and control groups (Fig. 2).

Effects of NAC on the canonical Wnt signaling pathway

Oxidative stress has been suggested to be involved in the pathogenesis of autism (4,5), and the canonical Wnt signaling pathway has been implicated in oxidative processes (2022). Therefore, the present study investigated the involvement of the Wnt pathway in the beneficial effects of NAC in VPA-exposed rats.

β-catenin is a key signaling molecule involved in the canonical Wnt pathway, whereas GSK-3β negatively regulates β-catenin via phosphorylation of its inhibitory sites, including Ser-33, Ser-37 and Thr-41 (19). The present results demonstrated that GSK-3β Ser-9 phosphorylation was enhanced in brain tissue samples isolated from VPA-exposed rats compared with control rats (Fig. 3A and B), thus suggesting that GSK-3β activity may be suppressed in VPA-induced autism. In addition, VPA-exposed rats exhibited significantly decreased β-catenin phosphorylation levels (Ser-33/37/Thr-41) compared with the controls (Fig. 3C and D), thus suggesting that VPA may inhibit GSK-3β to stabilize β-catenin. However, no statistically significant differences were detected in GSK-3β and β-catenin phosphorylation levels between rats in the VPA + NAC and VPA groups (Fig. 3).

Active β-catenin is translocated into the nucleus to activate the transcription of target genes of the Wnt/β-catenin pathway (31). Therefore, the protein expression levels of β-catenin were further investigated in the cytoplasm and nucleus. VPA-exposed rats exhibited significantly increased nuclear and cytoplasmic protein expression levels of β-catenin compared with rats in the control group (Fig. 4). Compared with the VPA group, NAC alone decreased the protein levels of β-catenin in the cytoplasm and nucleus (Fig. 4). However, no statistically significant differences were detected in nuclear and cytoplasmic β-catenin levels between rats in the VPA + NAC and VPA groups (Fig. 4).

Discussion

The present study demonstrated that NAC exerted beneficial effects in a VPA-induced rat model of autism. Treatment with NAC was demonstrated to reverse abnormal locomotor behaviors, including repetitive/stereotypic activity in rats prenatally exposed to VPA, compared with untreated VPA-exposed offspring. The beneficial effects of NAC in autism-induced deficits were associated with its antioxidative properties, resulting in the restoration of redox homeostasis in the brain. However, its actions did not appear to be associated with the canonical Wnt signaling pathway, as demonstrated by the unaltered GSK-3β and β-catenin phosphorylation levels, and the β-catenin protein levels in the cytoplasm and nucleus. These results suggested that the Wnt/β-catenin signaling pathway may be indirectly regulated by NAC through the activation of alternative signaling pathways or upstream factors. NAC alters glutathione metabolism and improves mitochondrial dysfunction in a subset of autistic lymphoblastoid cell lines (32). In accordance with these results, NAC has been reported to ameliorate autism phenotypes, associated with altered GSH levels (7,30).

Autism is a complex neurodevelopmental disorder, and its etiology has yet to be fully elucidated. Previous studies have suggested that oxidative stress may be involved in the pathogenesis of autism (30,3336). NAC is a potent antioxidant that is widely used for the treatment of an acetaminophen overdose (37), as a renal protectant in contrast-induced nephropathy (38), and as a preventive agent for atrial fibrillation (39). NAC is a membrane-permeable L-cysteine precursor, and is intracellularly reduced to L-cysteine, a precursor of the endogenous antioxidant, GSH (40). In the brain, the oxidation of NAC-derived L-cysteine to cystine has been reported, which can subsequently modulate extracellular glutamate levels (41). The regulatory effects of NAC on extracellular glutamate levels, and its restorative actions on intracellular GSH levels, in addition to its well-established safety profile, suggest a potential for NAC as an alternative therapeutic strategy for the treatment of patients with autism. Clinical studies have reported that NAC alone, or in combination with risperidone improved some of the core symptoms of autism, including irritability or aggressive behavior (7,12,14,15), self-injurious behavior (13), social interaction impairments (7,16), and anxiety-like behavior (16). In the present study, NAC was demonstrated to ameliorate stereotypic/repetitive locomotor activity in a VPA-induced rat model of autism.

During open field behavioral testing, NAC was revealed to attenuate repetitive/stereotypic activity in VPA-exposed rats, possibly through a decrease in hyper-reactivity to stress. A previous study from our laboratory demonstrated that VPA-treated rats have reduced sensitivity to stress and exhibited repetitive/stereotypic activity (28). Conversely, when stress sensitivity is restored to physiological levels, repetitive/stereotypic activities are abolished (28). Therefore, it may be hypothesized that the frequent repetitive/stereotypic movements in VPA-exposed rats are a result of hyper-reactivity to stress. However, a previous study reported contradictory results, demonstrating that the distance traveled in the open field was similar between VPA + NAC-treated and VPA-treated rats (16). This discrepancy may be due to the different doses of VPA used and the variation in NAC treatment duration. However, further studies are required in order to examine the complex interactions between neuropeptides and neuroendocrine factors that modulate stress reactions, and to determine their effects on movement impairments in VPA-treated rats.

The present study investigated the involvement of the antioxidative properties of NAC in the amelioration of autism-like behavioral abnormalities. In order to assess the antioxidative effects of NAC, the levels of MDA, a marker of lipid peroxidation, and thus of oxidative stress, were assessed in rat brain tissue samples. GSH is an intracellular antioxidant that participates in the endogenous mechanisms of defense against reactive oxygen species (42). The present results demonstrated that prenatal exposure to VPA induced oxidative stress in rats, as demonstrated by the increased MDA and decreased GSH levels in brain tissue samples. Notably, following treatment with NAC, MDA levels were suppressed, whereas GSH levels were enhanced in VPA-exposed rats compared with the untreated VPA group. These findings are in accordance with a previous study reporting that NAC significantly enhanced GSH levels in diabetic rats (43).

Further studies are required to elucidate the molecular mechanisms underlying the effects of NAC in the prevention of abnormal locomotor behaviors in autism. NAC is a GSH precursor and augments intracellular GSH levels. Wnt and its associated signaling proteins have been identified as targets for GSH (44), whereas NAC has been demonstrated to inhibit the activation of the canonical Wnt pathway in the retinas of diabetic rats (43). Furthermore, previous studies have reported a critical role for the Wnt pathway in the pathogenesis of autism (23,28,45). In a previous study by the present authors, VPA was revealed to inhibit the phosphorylation, and thus the inactivation, of β-catenin, which is a key effector during canonical Wnt signaling, possibly through the upregulation of WNT1 and WNT2 expression, thereby activating the canonical Wnt pathway (46). Notably, following treatment with the specific Wnt pathway inhibitor sulindac, Wnt/β-catenin signaling was suppressed and autism-like behavioral abnormalities in a VPA-induced rat model of autism were attenuated (28). These results suggested that increased Wnt-mediated signaling may contribute to the susceptibility to autism.

Oxidative stress and the Wnt signaling pathway have been implicated in numerous pathological conditions (21,47,48); however, further studies are required to elucidate the modulatory mechanisms that affect oxidative processes and implicate the Wnt pathway in the pathogenesis of autism. As a critical effector of Wnt signaling, β-catenin can activate the transcription of various target genes, following its cytoplasmic accumulation and subsequent nuclear translocation (18,49). In the present study, VPA was revealed to induce oxidative stress, as demonstrated by the increased levels of the oxidative stress marker, MDA, and the downregulation in the levels of the endogenous antioxidant, GSH, in VPA-exposed rats. Furthermore, prenatal exposure to VPA enhanced β-catenin levels in the cytoplasm and nucleus, whereas it was revealed to inhibit the phosphorylation of β-catenin, and promote the phosphorylation of GSK-3β. These results indicated that VPA activated the canonical Wnt signaling pathway in the rat autism model. However, treatment with NAC did not appear to affect the nuclear and cytoplasmic β-catenin levels, or the phosphorylation of β-catenin and GSK-3β, thus suggesting that the Wnt pathway may not be involved in NAC-mediated amelioration of autism-like behavior in VPA-treated rats. These results are in accordance with our previous in vitro study, which demonstrated that NAC reduced oxidative stress in primary neuronal cultures following pre-treatment with VPA without affecting the activity of the Wnt pathway (45). These findings suggested that the unaltered activity of the canonical Wnt pathway may be an indirect consequence, of reduced oxidative stress in VPA-exposed offspring followed by NAC administration.

The association between the Wnt signaling pathway and oxidative stress under various pathological conditions remains controversial. A previous study from our group reported that treatment with sulindac, a small-molecule inhibitor of the canonical Wnt pathway, reduced the levels of 4-hydroxynonenal, a marker of lipid peroxidation, in a VPA-induced rat model of autism, thus suggesting that oxidative stress may be involved in the regulation of Wnt signaling (28). However, it may be hypothesized that sulindac directly suppressed oxidative stress due to its antioxidative properties, and these effects may not be mediated by the Wnt pathway. Additionally, sulindac, a nonsteroidal anti-inflammatory drug, which has also been examined in cancer, neurodegenerative disease, and age-related macular degeneration, is able to reduce the levels of lipid peroxidation in an N-nitrosamine-induced mouse model of lung tumorigenesis (50), to reduce the generation of superoxide anions in Alzheimer's disease (51), and to lower ROS levels in a retinal pigmented epithelial cell line (52). VPA and sulindac reduce the activity of the canonical Wnt signaling pathway, compared with VPA exposure alone (28), however, sulindac has anti-inflammatory properties. Therefore, VPA may induce inflammatory status by possibly, though not exclusively, affecting the Wnt pathway. Further studies are required to investigate the association between oxidative stress and inflammation in the pathogenesis of autism. The molecular mechanisms underlying the direct and indirect implication of the canonical Wnt pathway in oxidative stress during the pathogenesis of autism require further investigation.

In conclusion, the results of the present study demonstrated that NAC reversed abnormal locomotor behaviors, including repetitive/stereotypic activity, in a VPA-induced rat model of autism, possibly due to its antioxidative properties. Notably, the beneficial effects of NAC in autism-like behavior did not appear to be associated with the canonical Wnt signaling pathway, suggesting that Wnt/β-catenin signaling may be indirectly regulated by NAC through alternative signaling pathways or upstream mediators. However, the results of the present study should be interpreted with caution, as autism is a complex and highly heterogeneous group of pathologies, and a VPA-induced rat model is not representative of the wide spectrum of disorders that autism encompasses.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant no. 81301174), the support project for the Disciplinary group of Psychology and Neuroscience, Xinxiang Medical University (grant no. 2016PN-KFKT-18), the Doctoral Scientific Research Foundation of Xinxiang Medical University (grant no. DOC2012-07) and the National College Student Innovation Project (grant no. 201310472070).

References

1 

Baird G, Simonoff E, Pickles A, Chandler S, Loucas T, Meldrum D and Charman T: Prevalence of disorders of the autism spectrum in a population cohort of children in South Thames: The special needs and autism project (SNAP). Lancet. 368:210–215. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Levitt P and Campbell DB: The genetic and neurobiologic compass points toward common signaling dysfunctions in autism spectrum disorders. J Clin Invest. 119:747–754. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Christensen DL, Baio J, Van Naarden Braun K, Bilder D, Charles J, Constantino JN, Daniels J, Durkin MS, Fitzgerald RT, Kurzius-Spencer M, et al: Prevalence and characteristics of autism spectrum disorder among children aged 8 years-autism and developmental disabilities monitoring network, 11 sites, United States, 2012. MMWR Surveill Summ. 65:1–23. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Ghezzo A, Visconti P, Abruzzo PM, Bolotta A, Ferreri C, Gobbi G, Malisardi G, Manfredini S, Marini M, Nanetti L, et al: Oxidative stress and erythrocyte membrane alterations in children with autism: Correlation with clinical features. PLoS One. 8:e664182013. View Article : Google Scholar : PubMed/NCBI

5 

Ghanizadeh A, Akhondzadeh S, Hormozi M, Makarem A, Abotorabi-Zarchi M and Firoozabadi A: Glutathione-related factors and oxidative stress in autism, a review. Curr Med Chem. 19:4000–4005. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Dean O, Giorlando F and Berk M: N-acetylcysteine in psychiatry: Current therapeutic evidence and potential mechanisms of action. J Psychiatry Neurosci. 36:78–86. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Ghanizadeh A and Derakhshan N: N-acetylcysteine for treatment of autism, a case report. J Res Med Sci. 17:985–987. 2012.PubMed/NCBI

8 

Paintlia MK, Paintlia AS, Barbosa E, Singh I and Singh AK: N-acetylcysteine prevents endotoxin-induced degeneration of oligodendrocyte progenitors and hypomyelination in developing rat brain. J Neurosci Res. 78:347–361. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Wang X, Svedin P, Nie C, Lapatto R, Zhu C, Gustavsson M, Sandberg M, Karlsson JO, Romero R, Hagberg H and Mallard C: N-acetylcysteine reduces lipopolysaccharide-sensitized hypoxic-ischemic brain injury. Ann Neurol. 61:263–271. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Farr SA, Poon HF, Dogrukol-Ak D, Drake J, Banks WA, Eyerman E, Butterfield DA and Morley JE: The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. J Neurochem. 84:1173–1183. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Holmay MJ, Terpstra M, Coles LD, Mishra U, Ahlskog M, Öz G, Cloyd JC and Tuite PJ: N-acetylcysteine boosts brain and blood glutathione in gaucher and parkinson diseases. Clin Neuropharmacol. 36:103–106. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Hardan AY, Fung LK, Libove RA, Obukhanych TV, Nair S, Herzenberg LA, Frazier TW and Tirouvanziam R: A randomized controlled pilot trial of oral N-acetylcysteine in children with autism. Biol Psychiatry. 71:956–961. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Marler S, Sanders KB and Veenstra-VanderWeele J: N-acetylcysteine as treatment for self-injurious behavior in a child with autism. J Child Adolesc Psychopharmacol. 24:231–234. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Ghanizadeh A and Moghimi-Sarani E: A randomized double blind placebo controlled clinical trial of N-Acetylcysteine added to risperidone for treating autistic disorders. BMC Psychiatry. 13:1962013. View Article : Google Scholar : PubMed/NCBI

15 

Nikoo M, Radnia H, Farokhnia M, Mohammadi MR and Akhondzadeh S: N-acetylcysteine as an adjunctive therapy to risperidone for treatment of irritability in autism: A randomized, double-blind, placebo-controlled clinical trial of efficacy and safety. Clin Neuropharmacol. 38:11–17. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Chen YW, Lin HC, Ng MC, Hsiao YH, Wang CC, Gean PW and Chen PS: Activation of mGluR2/3 underlies the effects of N-acetylcystein on amygdala-associated autism-like phenotypes in a valproate-induced rat model of autism. Front Behav Neurosci. 8:2192014. View Article : Google Scholar : PubMed/NCBI

17 

Kimelman D and Xu W: Beta-catenin destruction complex: Insights and questions from a structural perspective. Oncogene. 25:7482–7491. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Sierra J, Yoshida T, Joazeiro CA and Jones KA: The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev. 20:586–600. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Toledo EM, Colombres M and Inestrosa NC: Wnt signaling in neuroprotection and stem cell differentiation. Prog Neurobiol. 86:281–296. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Zhuang B, Luo X, Rao H, Li Q, Shan N, Liu X and Qi H: Oxidative stress-induced C/EBPβ inhibits β-catenin signaling molecule involving in the pathology of preeclampsia. Placenta. 36:839–846. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Hsu YC, Lee PH, Lei CC, Ho C, Shih YH and Lin CL: Nitric oxide donors rescue diabetic nephropathy through oxidative-stress-and nitrosative-stress-mediated Wnt signaling pathways. J Diabetes Investig. 6:24–34. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Yoshida GJ and Saya H: Inversed relationship between CD44 variant and c-Myc due to oxidative stress-induced canonical Wnt activation. Biochem Biophys Res Commun. 443:622–627. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Hormozdiari F, Penn O, Borenstein E and Eichler EE: The discovery of integrated gene networks for autism and related disorders. Genome Res. 25:142–154. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zhang Y, Yuan X, Wang Z and Li R: The canonical Wnt signaling pathway in autism. CNS Neurol Disord Drug Targets. 13:765–770. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Marui T, Funatogawa I, Koishi S, Yamamoto K, Matsumoto H, Hashimoto O, Jinde S, Nishida H, Sugiyama T, Kasai K, et al: Association between autism and variants in the wingless-type MMTV integration site family member 2 (WNT2) gene. Int J Neuropsychopharmacol. 13:443–449. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Schneider T, Turczak J and Przewłocki R: Environmental enrichment reverses behavioral alterations in rats prenatally exposed to valproic acid: Issues for a therapeutic approach in autism. Neuropsychopharmacology. 31:36–46. 2006.PubMed/NCBI

27 

Silvestrin R Bristot, Bambini-Junior V, Galland F, Bobermim L Daniele, Quincozes-Santos A, Abib R Torres, Zanotto C, Batassini C, Brolese G, Gonçalves CA, et al: Animal model of autism induced by prenatal exposure to valproate: Altered glutamate metabolism in the hippocampus. Brain Res. 1495:52–60. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Y, Sun Y, Wang F, Wang Z, Peng Y and Li R: Downregulating the canonical Wnt/β-catenin signaling pathway attenuates the susceptibility to autism-like phenotypes by decreasing oxidative stress. Neurochem Res. 37:1409–1419. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Slattery J Deepmala, Kumar N, Delhey L, Berk M, Dean O, Spielholz C and Frye R: Clinical trials of N-acetylcysteine in psychiatry and neurology: A systematic review. Neurosci Biobehav Rev. 55:294–321. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Wink LK, Adams R, Wang Z, Klaunig JE, Plawecki MH, Posey DJ, McDougle CJ and Erickson CA: A randomized placebo-controlled pilot study of N-acetylcysteine in youth with autism spectrum disorder. Mol Autism. 7:262016. View Article : Google Scholar : PubMed/NCBI

31 

Nelson WJ and Nusse R: Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 303:1483–1487. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Rose S, Frye RE, Slattery J, Wynne R, Tippett M, Melnyk S and James SJ: Oxidative stress induces mitochondrial dysfunction in a subset of autistic lymphoblastoid cell lines. Transl Psychiatry. 4:e3772014. View Article : Google Scholar : PubMed/NCBI

33 

Gvozdjáková A, Kucharská J, Ostatníková D, Babinská K, Nakládal D and Crane FL: Ubiquinol improves symptoms in children with autism. Oxid Med Cell Longev. 2014:7989572014. View Article : Google Scholar : PubMed/NCBI

34 

Gu F, Chauhan V and Chauhan A: Glutathione redox imbalance in brain disorders. Curr Opin Clin Nutr Metab Care. 18:89–95. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Raymond LJ, Deth RC and Ralston NV: Potential role of selenoenzymes and antioxidant metabolism in relation to autism etiology and pathology. Autism Res Treat. 2014:1649382014.PubMed/NCBI

36 

James SJ, Rose S, Melnyk S, Jernigan S, Blossom S, Pavliv O and Gaylor DW: Cellular and mitochondrial glutathione redox imbalance in lymphoblastoid cells derived from children with autism. FASEB J. 23:2374–2383. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Green JL, Heard KJ, Reynolds KM and Albert D: Oral and intravenous acetylcysteine for treatment of acetaminophen toxicity: A systematic review and meta-analysis. West J Emerg Med. 14:218–226. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Quintavalle C, Donnarumma E, Fiore D, Briguori C and Condorelli G: Therapeutic strategies to prevent contrast-induced acute kidney injury. Curr Opin Cardiol. 28:676–682. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Liu XH, Xu CY and Fan GH: Efficacy of N-acetylcysteine in preventing atrial fibrillation after cardiac surgery: A meta-analysis of published randomized controlled trials. BMC Cardiovasc Disord. 14:522014. View Article : Google Scholar : PubMed/NCBI

40 

Shahripour R Bavarsad, Harrigan MR and Alexandrov AV: N-acetylcysteine (NAC) in neurological disorders: Mechanisms of action and therapeutic opportunities. Brain Behav. 4:108–122. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Berk M, Malhi GS, Gray LJ and Dean OM: The promise of N-acetylcysteine in neuropsychiatry. Trends Pharmacol Sci. 34:167–177. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Baker MS, Feigan J and Lowther DA: Chondrocyte antioxidant defences: The roles of catalase and glutathione peroxidase in protection against H2O2 dependent inhibition of proteoglycan biosynthesis. J Rheumatol. 15:670–677. 1988.PubMed/NCBI

43 

Zhou T, Zhou KK, Lee K, Gao G, Lyons TJ, Kowluru R and Ma JX: The role of lipid peroxidation products and oxidative stress in activation of the canonical wingless-type MMTV integration site (WNT) pathway in a rat model of diabetic retinopathy. Diabetologia. 54:459–468. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Gong SP, Lee EJ, Lee ST, Kim H, Lee SH, Han HJ and Lim JM: Improved establishment of autologous stem cells derived from preantral follicle culture and oocyte parthenogenesis. Stem Cells Dev. 17:695–712. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Zhang Y, Yang C, Yuan G, Wang Z, Cui W and Li R: Sulindac attenuates valproic acid-induced oxidative stress levels in primary cultured cortical neurons and ameliorates repetitive/stereotypic-like movement disorders in Wistar rats prenatally exposed to valproic acid. Int J Mol Med. 35:263–270. 2015.PubMed/NCBI

46 

Wang Z, Xu L, Zhu X, Cui W, Sun Y, Nishijo H, Peng Y and Li R: Demethylation of specific Wnt/β-catenin pathway genes and its upregulation in rat brain induced by prenatal valproate exposure. Anat Rec (Hoboken). 293:1947–1953. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Strakovsky RS and Pan YX: A decrease in DKK1, a WNT inhibitor, contributes to placental lipid accumulation in an obesity-prone rat model. Biol Reprod. 86:812012. View Article : Google Scholar : PubMed/NCBI

48 

Chen JR, Lazarenko OP, Shankar K, Blackburn ML, Badger TM and Ronis MJ: A role for ethanol-induced oxidative stress in controlling lineage commitment of mesenchymal stromal cells through inhibition of Wnt/beta-catenin signaling. J Bone Miner Res. 25:1117–1127. 2010. View Article : Google Scholar : PubMed/NCBI

49 

McGrew LL, Takemaru K, Bates R and Moon RT: Direct regulation of the Xenopus engrailed-2 promoter by the Wnt signaling pathway, and a molecular screen for Wnt-responsive genes, confirm a role for Wnt signaling during neural patterning in Xenopus. Mech Dev. 87:21–32. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Bilodeau JF, Wang M, Chung FL and Castonguay A: Effects of nonsteroidal antiinflammatory drugs on oxidative pathways in A/J mice. Free Radic Biol Med. 18:47–54. 1995. View Article : Google Scholar : PubMed/NCBI

51 

Dairam A, Müller AC and Daya S: Non-steroidal anti-inflammatory agents, tolmetin and sulindac attenuate quinolinic acid (QA)-induced oxidative stress in primary hippocampal neurons and reduce QA-induced spatial reference memory deficits in male Wistar rats. Life Sci. 80:1431–1438. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Sur A, Kesaraju S, Prentice H, Ayyanathan K, Baronas-Lowell D, Zhu D, Hinton DR, Blanks J and Weissbach H: Pharmacological protection of retinal pigmented epithelial cells by sulindac involves PPAR-α. Proc Natl Acad Sci USA. 111:16754–16759. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 16 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Cui W, Zhai Q, Zhang T and Wen X: N-acetylcysteine ameliorates repetitive/stereotypic behavior due to its antioxidant properties without activation of the canonical Wnt pathway in a valproic acid-induced rat model of autism. Mol Med Rep 16: 2233-2240, 2017
APA
Zhang, Y., Cui, W., Zhai, Q., Zhang, T., & Wen, X. (2017). N-acetylcysteine ameliorates repetitive/stereotypic behavior due to its antioxidant properties without activation of the canonical Wnt pathway in a valproic acid-induced rat model of autism. Molecular Medicine Reports, 16, 2233-2240. https://doi.org/10.3892/mmr.2017.6787
MLA
Zhang, Y., Cui, W., Zhai, Q., Zhang, T., Wen, X."N-acetylcysteine ameliorates repetitive/stereotypic behavior due to its antioxidant properties without activation of the canonical Wnt pathway in a valproic acid-induced rat model of autism". Molecular Medicine Reports 16.2 (2017): 2233-2240.
Chicago
Zhang, Y., Cui, W., Zhai, Q., Zhang, T., Wen, X."N-acetylcysteine ameliorates repetitive/stereotypic behavior due to its antioxidant properties without activation of the canonical Wnt pathway in a valproic acid-induced rat model of autism". Molecular Medicine Reports 16, no. 2 (2017): 2233-2240. https://doi.org/10.3892/mmr.2017.6787