Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1

  • Authors:
    • Chun‑Yan Wang
    • Wei Zou
    • Xiao‑Yu Liang
    • Zhi‑Sheng Jiang
    • Xiang Li
    • Hai‑Jun Wei
    • Yi‑Yun Tang
    • Ping Zhang
    • Xiao‑Qing Tang
  • View Affiliations

  • Published online on: July 15, 2017     https://doi.org/10.3892/mmr.2017.7004
  • Pages: 3587-3593
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It was previously confirmed that hydrogen sulfide (H2S) has a neuroprotective effect, preventing homocysteine‑induced neurotoxicity. However, the exact molecular mechanisms underlying this protective effect remain to be fully elucidated. Endoplasmic reticulum (ER) stress contributes to homocysteine‑induced neurotoxicity. Silent mating type information regulator 2 homolog 1 (SIRT‑1) can attenuate ER stress, exerting its neuroprotective effect. Therefore, the present study aimed to investigate whether H2S protects PC12 cells against homocysteine‑induced ER stress and whether SIRT‑1 mediates this protective effect of H2S. Western blotting was used to detect the expression of SIRT‑1, glucose‑regulated protein 78 (GRP78), and cleaved caspase‑12 in PC12 cells. It was observed that sodium hydrosulfide (NaHS), an exogenous H2S donor, significantly attenuated the homocysteine‑induced ER stress responses, including increases in the protein expression levels of GRP78 and cleaved caspase‑12. Simultaneously, NaHS upregulated the expression of SIRT‑1 and reversed the homocysteine‑induced downregulation of SIRT‑1 in PC12 cells. Sirtinol, a specific inhibitor of SIRT‑1, eliminated the protective effects of H2S in homocysteine‑induced ER stress. These data indicated that H2S prevented homocysteine‑induced ER stress via enhancing the expression of SIRT‑1. These findings offer novel insight into the protective mechanisms of H2S against homocysteine‑induced neurotoxicity.

Introduction

Homocysteine, a thiol-containing amino acid, is generated by the demethylation of methionine (1,2). It has been established that an elevated level of circulating homocysteine is an independent risk factor for Alzheimer's disease (AD) (38), and there is increasing evidence that homocysteine directly causes neurotoxicity in multiple neuronal types (911). In addition, it is known that endoplasmic reticulum (ER) stress is closely associated with the development and pathology of AD, which has the typical characteristics of inclusion bodies, abnormal formation and misfolded protein aggregation (1214). It has also been reported that homocysteine leads to ER stress in neuronal cells (1518), which suggests that ER stress-mediated homocysteine-induced neurotoxicity may be vital in the pathogenesis of AD. Therefore, the suppression of ER stress may provide a promising approach for the treatment of homocysteine-dependent neurodegenerative diseases.

Hydrogen sulfide (H2S) is considered to be a novel endogenous neuroprotectant (1923). Of note, data from our previous study demonstrated that the disturbance of endogenous H2S generation was involved in the neurotoxicity of homocysteine (24), and that H2S ameliorated homocysteine induced-neurotoxicity (25), indicating the potential of H2S-based prevention and treatment for neuronal injury induced by homocysteine exposure. Based on the importance of ER stress in the neurotoxicity of homocysteine, the present study aimed to expand on current understanding of the protective effects of H2S in homocysteine-elicited neurotoxicity by examining the effects of H2S on homocysteine-induced ER stress and the underlying mechanisms.

Sirtuins are nicotinamide adenine dinucleotide-dependent histone deacetylases, which counter aging, having a broad spectrum of metabolic and stress-tolerance functions. Emerging evidence has confirmed that silent mating type information regulator 2 homolog 1 (SIRT-1), one of the seven mammalian sirtuins, is directly involved in the neuronal protective effect against cellular damage and stressful perturbations in neurological diseases, including AD (2629), amyotrophic lateral sclerosis (28), Huntington's disease (30,31) and Parkinson's disease (32). Furthermore, it has been reported that SIRT-1 mediates the neuroprotective effect of paliperidone against MK-801-induced neuronal damage (33) and hyperbaric oxygen preconditioning-induced ischemic tolerance in the rat brain (34). Of note, previous studies have suggested that SIRT-1 exhibits its beneficial effects in neuroprotection via alleviation of the ER stress response (35,36). Therefore, the present study investigated whether SIRT-1 contributes to the protective effects of H2S against homocysteine-induced ER stress.

The results of the present study revealed that H2S prevented homocysteine-induced ER stress and increased the protein expression of SIRT-1 in PC12 cells. Sirtinol, a specific inhibitor of SIRT-1, eliminated the inhibitory effects of H2S against homocysteine-induced ER stress in the PC12 cells. These findings indicated that H2S protects PC12 cells against homocysteine-induced ER stress via upregulating the expression of SIRT-1.

Materials and methods

Materials

Sodium hydrosulfide (NaHS), an exogenous donor of H2S, homocysteine and sirtinol, a specific inhibitor of SIRT-1, were supplied by Sigma-Aldrich (cat. no. S7942; Merck KGaA, Darmstadt, Germany). Specific antibody against SIRT-1 (cat. no. ab110304) was purchased from Abcam (Cambridge, UK). Specific antibody against glucose-regulated protein 78 (GRP78; cat. no. S1931) was obtained from Epitomics (Burlingame, CA, USA). Specific antibody against cleaved caspase-12 (cat. no. C7611) was supplied by Sigma-Aldrich (Merck KGaA); β-actin polyclonal antibody (cat. no. 20536-1-AP) and goat anti-rat immunoglobulin (Ig)G (cat. no. SA00001-2) or goat anti-mouse IgG (cat. no. SA00001-1) antibody were obtained from ProteinTech Group, Inc. (Chicago, IL, USA). RPMI-1640 medium, fetal bovine serum (FBS) and horse serum were obtained from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA).

Cell culture

The PC12 cells (American Type Culture Collection; CRL-1721), provided by Sun Yat-sen University Experimental Animal Center (Guangzhou, China), were cultured in RPMI-1640 medium supplemented with 10% (v/v) heat-inactivated horse serum and 5% FBS (v/v) at 37°C, in an atmosphere containing 5% CO2 and 95% air. The culture medium was replaced every 2–3 days.

Western blot analysis

The PC12 cells, treated as described above, were homogenized in radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Shanghai, China) containing phenylmethylsulphonyl fluoride (1 mM) for 30 min 4°C and the supernatants was obtained by centrifugation at 5,000 × g for 10 min at 4°C. Protein concentrations were determined using a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology). Equivalent quantities of protein (50 µg) were separated by SDS-PAGE on a 12% gel. The proteins were then transferred onto polyvinylidene fluoride membranes, and the membranes were blocked with 5% skim milk in Tris-buffered saline containing 0.1% Tween-20 (TBST) for 2 h at room temperature. The membranes were then incubated with primary antibodies specific for blocking solution, containing primary antibodies against SIRT-1 (1:2,000), GRP78 (1:2,000), cleaved caspase-12 (1:2,000) and β-actin (1:5,000) overnight at 4°C. Following washing with TBST three times, the membranes with SIRT-1 were incubated in peroxidase-conjugated affinipure goat anti-mouse IgG (1:5,000) and others were incubated in anti-rabbit secondary antibodies (1:5,000) in blocking solution for 2 h at 25°C and then washed in TBST buffer. The bands of protein were visualized using an enhanced chemiluminescence reaction solution (solution 1:0.1 M Tris-HCl, luminol and p-coumaric acid; solution 2:0.1 M Tris-HCl and hydrogen peroxide) for 2 min, and quantified using an image analysis system equipped withBIO-1D software (v4.62; VilberLourmat, Marne-la-Vallée, France).

Statistical analysis

Data are expressed as the mean ± standard error of the mean. The significance of differences in different groups was assessed using one-way analyses of variance followed by the least significant difference test using SPSS version 19.0 (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Homocysteine induces ER stress in PC12 cells

To investigate whether homocysteine induces ER stress in PC12 cells, the present study measured the expression levels of ER stress-related proteins, including GRP78 and cleaved caspase-12, in homocysteine-treated PC12 cells using western blot analysis. It was found that treatment with homocysteine (1.25, 2.5 or 5 mM for 24 h) significantly increased the expression levels of GRP78 (Fig. 1A) and cleaved caspase-12 (Fig. 1B) in the PC12 cells, which indicated that homocysteine-induced ER stress in the PC12 cells.

H2S protects PC12 cells from homocysteine-induced ER stress

To determine whether H2S protects PC12 cells against homocysteine-induced ER stress, the present study examined the effects of H2S on the protein levels of GRP78 and cleaved caspase-12 in homocysteine-exposed PC12 cells. As shown in Fig. 2, cotreatment of the PC12 cells with NaHS (200 or 400 µM) significantly downregulated the expression levels of GRP78 (Fig. 2A) and cleaved caspase-12 (Fig. 2B), compared with the cells treated with 5 mM of homocysteine for 24 h. This indicated that H2S had a protective effect in homocysteine-induced ER stress.

H2S upregulates the expression of SIRT-1 in PC12 cells

As shown in Fig. 3A, treatment with homocysteine (1.25, 2.5 or 5 mM, for 24 h) significantly decreased the expression levels of SIRT-1 in the PC12 cells, which indicated that homocysteine downregulated the protein expression of SIRT-1 in PC12 cells. Treatment with NaHS at concentrations of 200 and 400 µM for 24 h dose-dependently increased the expression of SIRT-1 in the PC12 cells (Fig. 3B), and also significantly increased the expression of SIRT-1 in the homocysteine-exposed (5 mM for 24 h) PC12 cells (Fig. 3C), which indicated the promoting effect of H2S on the expression of SIRT-1 in PC12 cells.

Inhibition of SIRT-1 eliminates the beneficent effect of H2S against homocysteine-induced ER stress in PC12 cells

To further investigate whether the protective effect of H2S on homocysteine-induced ER stress in PC12 cells was through the upregulation of SIRT-1, the present study used sirtinol, a specific inhibitor of SIRT-1, to examine the effect of H2S on ER stress under homocysteine treatment. The PC12 cells were pretreated with sirtinol (15 µM) for 30 min prior to the administration of NaHS (200 µM). The results demonstrated that sirtinol (15 µM) treatment inhibited the NaHS (200 µM)-induced suppression of GRP78 (Fig. 4A) and cleaved caspase-12 (Fig. 4B) in the homocysteine-exposed PC12 cells. Treatment with sirtinol alone did not affect the expression of these two proteins. These results indicated that the inhibition of SIRT-1 eliminated the protective effect of H2S against homocysteine-induced ER stress.

Discussion

In our previous study, it was demonstrated that H2S had a protective effect against homocysteine-induced neurotoxicity (25). In addition, the involvement of abnormal ER stress has been shown to be prominent in the neurotoxicity of homocysteine (15,16). Therefore, the present study was designed to investigate whether the protective role of H2S in the neurotoxicity of homocysteine was associated with regulating neuronal ER stress, and the underlying mechanisms were investigated. The main findings of the present study were as follows: i) H2S markedly inhibited homocysteine-induced ER stress in the PC12 cells; ii) H2S enhanced the protein level of SIRT-1 in the presence or absence of homocysteine treatment; and iii) sirtinol, an inhibitor of SIRT-1, eliminated the inhibitory effect of H2S on homocysteine-induced ER stress. These findings suggested the protective role of H2S against homocysteine-induced ER stress by enhancing the expression of SIRT-1.

Increasing evidence has confirmed the neurotoxic effects of homocysteine (3740), which are associated with ER stress (15,16). It is known that GRP78 is an ER-chaperone protein involved in the modulation of ER dynamic homeostasis (41,42). Pro-caspase-12 is located on the cytoplasmic region of ER and is proteolytically activated during excess ER stress (4345). GRP78 and caspase-12 are two important markers of ER stress. In the present study, the effects of homocysteine on the protein expression levels of GRP78 and cleaved caspase-12 in PC12 cells were examined. It was demonstrated that homocysteine upregulated the protein levels of GRP78 and cleaved caspase-12 in the PC12 cells. These results indicated that homocysteine was able to elevate ER stress in the PC12 cells. It is known that ER stress is involved in the pathogenic effects of homocysteine in several diseases, including cardiovascular disease (46), apoptosis of osteoblastic cells (47), insulin resistance of adipose tissue (48), and type 2 diabetes mellitus (49). Therefore, ER stress may be a common intermediate pathway in the homocysteine-induced pathogenic effects in tissues and cells.

H2S is a protective gaseous signaling molecule. In our previous study, it was demonstrated that H2S prevented homocysteine-induced neurotoxicity in PC12 cells (25). To improve current understanding of the protective role of H2S in the neurotoxicity of homocysteine, the present study investigated whether H2S suppresses the homocysteine-induced upregulatory effect on the expression levels of GRP78 and cleaved caspase-12. The results showed that NaHS (the donor of H2S) downregulated the expression levels of GRP78 and cleaved caspase-12 in the homocysteine-exposed PC12 cells, which indicated that H2S was able to suppress homocysteine-induced ER stress. Previous studies have demonstrated that H2S prevents ER stress in doxorubicin-induced cardiotoxicity (50) and 6-hydroxydopamine-induced neurotoxicity (51). These previous findings offer a reasonable explanation for the results obtained in the present study. Furthermore, Wei et al (46) revealed the protective effect of H2S against homocysteine-induced cardiomyocytic ER stress. In the present study, the inhibitory role of H2S in homocysteine-induced ER stress was further confirmed in the PC12 cells. Therefore, the regulation of ER stress offers insights into the protective effect of H2S against homocysteine neurotoxicity.

The present study also examined the possible underlying signaling mechanisms for the protective effect of H2S against homocysteine-induced ER stress. SIRT-1is involved in lifespan modulation (5254) and orchestrates diverse biological processes, including cell survival, differentiation and metabolism (55,56). SIRT-1 is considered to be a vital modulator of cellular defenses and survival in response to stress (57,58). SIRT-1 is also expressed in the brain. Accumulating evidence has indicated that the upregulation of SIRT-1 rescues neurons in acute and chronic neurological diseases (28,31,59). It has also been found that impaired SIRT1-deacetylation induces ER stress (60) and that the overexpression of SIRT-1 attenuates ER stress (35,36). This suggests that SIRT-1 is important in counteracting ER stress, therefore, the present study focused on the effect of homocysteine on the expression of SIRT-1, and the role of H2S in the expression of SIRT-1 in PC12 cells treated with or without homocysteine. The results showed that the expression of SIRT-1 was downregulated in the homocysteine-exposed PC12 cells, which indicated the involvement of downregulated SIRT-1 in the increase of ER stress induced by homocysteine. In addition, NaHS was found to increase the protein expression of SIRT-1 in PC12 cells. It was also found that NaHS inhibited the homocysteine-induced decrease in the protein expression of SIRT-1 in PC12 cells. These results suggested that the upregulation of SIRT-1 contributed to the beneficent effects of H2S on homocysteine-induced ER stress. To confirm whether SIRT-1 mediates the protective effect of H2S against homocysteine-induced ER stress, the present study examined whether the inhibition of SIRT-1 eliminates the protective effect of H2S against ER stress induced by homocysteine. The results confirmed that the inhibition of SIRT-1 inhibited the reversal effect of H2S on the homocysteine-increased protein expression of GRP78 and cleaved caspase-12 in PC12 cells. Taken together, these results suggested that the upregulation of SIRT-1 mediated the H2S-induced protective effects against homocysteine-induced ER stress.

In conclusion, the present study demonstrated that H2S was able to overcome homocysteine-induced ER stress and increases in the protein expression of SIRT-1 in PC12 cells. Inhibiting SIRT-1 reversed the protective effect of H2S against ER stress elicited by homocysteine in PC12 cells. These results suggested that H2S had the ability to inhibit homocysteine-induced ER stress and that the upregulation of SIRT-1 mediated this protective effect of H2S. These findings shed light on the molecular mechanism underlying the protective role of H2S in the neurotoxicity of homocysteine. Homocysteine is an independent risk factor for AD (37) and ER stress is a crucial process in the pathogenesis of AD (1214). Therefore, the results of the present study suggested that positive intervention of SIRT-1 may have profound therapeutic benefits against homocysteine-dependent neurodegenerative diseases.

Acknowledgements

This study was supported by the Natural Science Foundation of China (grant nos. 81400881 and 81471310) and the Zhengxiang Scholar Program of University of South China (grant no. 2014-004).

References

1 

Prudova A, Bauman Z, Braun A, Vitvitsky V, Lu SC and Banerjee R: S-adenosylmethionine stabilizes cystathionine beta-synthase and modulates redox capacity. Proc Natl Acad Sci USA. 103:6489–6494. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Selhub J: Homocysteine metabolism. Annu Rev Nutr. 19:217–246. 1999. View Article : Google Scholar : PubMed/NCBI

3 

Van Dam F and Van Gool WA: Hyperhomocysteinemia and Alzheimer's disease: A systematic review. Arch Gerontol Geriatr. 48:425–430. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D'Agostino RB, Wilson PW and Wolf PA: Plasma homocysteine as a risk factor for dementia and Alzheimer's disease. N Engl J Med. 346:476–483. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Miller JW: Homocysteine and Alzheimer's disease. Nutr Rev. 57:126–129. 1999.PubMed/NCBI

6 

Clarke R, Smith AD, Jobst KA, Refsum H, Sutton L and Ueland PM: Folate, vitamin B12, and serum total homocysteine levels in confirmed Alzheimer disease. Arch Neurol. 55:1449–1455. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Dwyer BE, Raina AK, Perry G and Smith MA: Homocysteine and Alzheimer's disease: A modifiable risk? Free Radic Biol Med. 36:1471–1475. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Sharma GS, Kumar T, Dar TA and Singh LR: Protein N-homocysteinylation: From cellular toxicity to neurodegeneration. Biochim Biophys Acta. 1850:2239–2245. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Lin N, Qin S, Luo S, Cui S, Huang G and Zhang X: Homocysteine induces cytotoxicity and proliferation inhibition in neural stem cells via DNA methylation in vitro. FEBS J. 281:2088–2096. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Abushik PA, Niittykoski M, Giniatullina R, Shakirzyanova A, Bart G, Fayuk D, Sibarov DA, Antonov SM and Giniatullin R: The role of NMDA and mGluR5 receptors in calcium mobilization and neurotoxicity of homocysteine in trigeminal and cortical neurons and glial cells. J Neurochem. 129:264–274. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Park YJ, Ko JW, Jang Y and Kwon YH: Activation of AMP-activated protein kinase alleviates homocysteine-mediated neurotoxicity in SH-SY5Y cells. Neurochem Res. 38:1561–1571. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Hoozemans JJ, Veerhuis R, Van Haastert ES, Rozemuller JM, Baas F, Eikelenboom P and Scheper W: The unfolded protein response is activated in Alzheimer's disease. Acta Neuropathol. 110:165–172. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Salminen A, Kauppinen A, Suuronen T, Kaarniranta K and Ojala J: ER stress in Alzheimer's disease: A novel neuronal trigger for inflammation and Alzheimer's pathology. J Neuroinflammation. 6:412009. View Article : Google Scholar : PubMed/NCBI

14 

Cornejo VH and Hetz C: The unfolded protein response in Alzheimer's disease. Semin Immunopathol. 35:277–292. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Althausen S and Paschen W: Homocysteine-induced changes in mRNA levels of genes coding for cytoplasmic- and endoplasmic reticulum-resident stress proteins in neuronal cell cultures. Brain Res Mol Brain Res. 84:32–40. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Chigurupati S, Wei Z, Belal C, Vandermey M, Kyriazis GA, Thiruma V, Arumugam TV and Chan SL: The homocysteine-inducible endoplasmic reticulum stress protein counteracts calcium store depletion and induction of CCAAT enhancer-binding protein homologous protein in a neurotoxin model of Parkinson disease. J Biol Chem. 284:18323–18333. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Kim HJ, Cho HK and Kwon YH: Synergistic induction of ER stress by homocysteine and beta-amyloid in SH-SY5Y cells. J Nutr Biochem. 19:754–761. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Park YJ, Jang Y and Kwon YH: Protective effect of isoflavones against homocysteine-mediated neuronal degeneration in SH-SY5Y cells. Amino Acids. 39:785–794. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Zhou CF and Tang XQ: Hydrogen sulfide and nervous system regulation. Chin Med J (Engl). 124:3576–3582. 2011.PubMed/NCBI

20 

Łowicka E and Beltowski J: Hydrogen sulfide (H2S)-the third gas of interest for pharmacologists. Pharmacol Rep. 59:4–24. 2007.PubMed/NCBI

21 

Kimura H, Shibuya N and Kimura Y: Hydrogen sulfide is a signaling molecule and a cytoprotectant. Antioxid Redox Signal. 17:45–57. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Hu LF, Lu M, Wong PT Hon and Bian JS: Hydrogen sulfide: Neurophysiology and neuropathology. Antioxid Redox Signal. 15:405–419. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Qu K, Lee SW, Bian JS, Low CM and Wong PT: Hydrogen sulfide: Neurochemistry and neurobiology. Neurochem Int. 52:155–165. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Tang XQ, Shen XT, Huang YE, Chen RQ, Ren YK, Fang HR, Zhuang YY and Wang CY: Inhibition of endogenous hydrogen sulfide generation is associated with homocysteine-induced neurotoxicity: Role of ERK1/2 activation. J Mol Neurosci. 45:60–67. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Tang XQ, Shen XT, Huang YE, Ren YK, Chen RQ, Hu B, He JQ, Yin WL, Xu JH and Jiang ZS: Hydrogen sulfide antagonizes homocysteine-induced neurotoxicity in PC12 cells. Neurosci Res. 68:241–249. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi S, Mucke L and Gan L: SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem. 280:40364–40374. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Qin W, Yang T, Ho L, Zhao Z, Wang J, Chen L, Zhao W, Thiyagarajan M, MacGrogan D, Rodgers JT, et al: Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J Biol Chem. 281:21745–21754. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, Delalle I, Baur JA, Sui G, Armour SM, et al: SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO J. 26:3169–3179. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Donmez G, Wang D, Cohen DE and Guarente L: SIRT1 suppresses beta-amyloid production by activating the alpha-secretase gene ADAM10. Cell. 142:320–332. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Jeong H, Cohen DE, Cui L, Supinski A, Savas JN, Mazzulli JR, Yates JR III, Bordone L, Guarente L and Krainc D: Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat Med. 18:159–165. 2012. View Article : Google Scholar

31 

Jiang M, Wang J, Fu J, Du L, Jeong H, West T, Xiang L, Peng Q, Hou Z, Cai H, et al: Neuroprotective role of Sirt1 in mammalian models of Huntington's disease through activation of multiple Sirt1 targets. Nat Med. 18:153–158. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Donmez G, Arun A, Chung CY, McLean PJ, Lindquist S and Guarente L: SIRT1 protects against α-synuclein aggregation by activating molecular chaperones. J Neurosci. 32:124–132. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Zhu D, Zhang J, Wu J, Li G, Yao W, Hao J and Sun J: Paliperidone protects SH-SY5Y cells against MK-801-induced neuronal damage through inhibition of Ca(2+) influx and regulation of SIRT1/miR-134 signal pathway. Mol Neurobiol. 53:2498–2509. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yan W, Fang Z, Yang Q, Dong H, Lu Y, Lei C and Xiong L: SirT1 mediates hyperbaric oxygen preconditioning-induced ischemic tolerance in rat brain. J Cereb Blood Flow Metab. 33:396–406. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Li Y, Xu S, Giles A, Nakamura K, Lee JW, Hou X, Donmez G, Li J, Luo Z, Walsh K, et al: Hepatic overexpression of SIRT1 in mice attenuates endoplasmic reticulum stress and insulin resistance in the liver. FASEB J. 25:1664–1679. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Jung TW, Lee KT, Lee MW and Ka KH: SIRT1 attenuates palmitate-induced endoplasmic reticulum stress and insulin resistance in HepG2 cells via induction of oxygen-regulated protein 150. Biochem Biophys Res Commun. 422:229–232. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Kuszczyk M, Gordon-Krajcer W and Lazarewicz JW: Homo-cysteine-induced acute excitotoxicity in cerebellar granule cells in vitro is accompanied by PP2A-mediated dephosphorylation of tau. Neurochem Int. 55:174–180. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Oldreive CE and Doherty GH: Neurotoxic effects of homocysteine on cerebellar Purkinje neurons in vitro. Neurosci Lett. 413:52–57. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Lipton SA, Kim WK, Choi YB, Kumar S, D'Emilia DM, Rayudu PV, Arnelle DR and Stamler JS: Neurotoxicity associated with dual actions of homocysteine at the N-methyl-D-aspartate receptor. Proc Natl Acad Sci USA. 94:5923–5928. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Bhatia P and Singh N: Homocysteine excess: Delineating the possible mechanism of neurotoxicity and depression. Fundam Clin Pharmacol. 29:522–528. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Ni M, Zhang Y and Lee AS: Beyond the endoplasmic reticulum: Atypical GRP78 in cell viability, signalling and therapeutic targeting. Biochem J. 434:181–188. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Lee AS: The ER chaperone and signaling regulator GRP78/BiP as a monitor of endoplasmic reticulum stress. Methods. 35:373–381. 2005. View Article : Google Scholar : PubMed/NCBI

43 

Szegezdi E, Fitzgerald U and Samali A: Caspase-12 and ER-stress-mediated apoptosis: The story so far. Ann N Y Acad Sci. 1010:186–194. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA and Yuan J: Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature. 403:98–103. 2000. View Article : Google Scholar : PubMed/NCBI

45 

de la Cadena SG, Hernandez-Fonseca K, Camacho-Arroyo I and Massieu L: Glucose deprivation induces reticulum stress by the PERK pathway and caspase-7- and calpain-mediated caspase-12 activation. Apoptosis. 19:414–427. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Wei H, Zhang R, Jin H, Liu D, Tang X, Tang C and Du J: Hydrogen sulfide attenuates hyperhomocysteinemia-induced cardiomyocytic endoplasmic reticulum stress in rats. Antioxid Redox Signal. 12:1079–1091. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Li Y, Zhang H, Jiang C, Xu M, Pang Y, Feng J, Xiang X, Kong W, Xu G, Li Y and Wang X: Hyperhomocysteinemia promotes insulin resistance by inducing endoplasmic reticulum stress in adipose tissue. J Biol Chem. 288:9583–9592. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Zbidi H, Redondo PC and López JJ, Bartegi A, Salido GM and López JJ: Homocysteine induces caspase activation by endoplasmic reticulum stress in platelets from type 2 diabetics and healthy donors. Thromb Haemost. 103:1022–1032. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Park SJ, Kim KJ, Kim WU, Oh IH and Cho CS: Involvement of endoplasmic reticulum stress in homocysteine-induced apoptosis of osteoblastic cells. J Bone Miner Metab. 30:474–484. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Wang XY, Yang CT, Zheng DD, Mo LQ, Lan AP, Yang ZL, Hu F, Chen PX, Liao XX and Feng JQ: Hydrogen sulfide protects H9c2 cells against doxorubicin-induced cardiotoxicity through inhibition of endoplasmic reticulum stress. Mol Cell Biochem. 363:419–426. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Xie L, Tiong CX and Bian JS: Hydrogen sulfide protects SH-SY5Y cells against 6-hydroxydopamine-induced endoplasmic reticulum stress. Am J Physiol Cell Physiol. 303:C81–C91. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Canto C and Auwerx J: Caloric restriction, SIRT1 and longevity. Trends Endocrinol Metab. 20:325–331. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Bordone L and Guarente L: Calorie restriction, SIRT1 and metabolism: Understanding longevity. Nat Rev Mol Cell Biol. 6:298–305. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Satoh A, Brace CS, Rensing N, Cliften P, Wozniak DF, Herzog ED, Yamada KA and Imai S: Sirt1 extends life span and delays aging in mice through the regulation of Nk2 homeobox 1 in the DMH and LH. Cell Metab. 18:416–430. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Tang BL: SIRT1, neuronal cell survival and the insulin/IGF-1 aging paradox. Neurobiol Aging. 27:501–505. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Michan S and Sinclair D: Sirtuins in mammals: Insights into their biological function. Biochem J. 404:1–13. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, et al: Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 303:2011–2015. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Milner J: Cellular regulation of SIRT1. Curr Pharm Des. 15:39–44. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Herskovits AZ and Guarente L: SIRT1 in neurodevelopment and brain senescence. Neuron. 81:471–483. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Ghemrawi R, Pooya S, Lorentz S, Gauchotte G, Arnold C, Gueant JL and Battaglia-Hsu SF: Decreased vitamin B12 availability induces ER stress through impaired SIRT1-deacetylation of HSF1. Cell Death Dis. 4:e5532013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 16 Issue 3

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
Wang CY, Zou W, Liang XY, Jiang ZS, Li X, Wei HJ, Tang YY, Zhang P and Tang XQ: Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1. Mol Med Rep 16: 3587-3593, 2017
APA
Wang, C., Zou, W., Liang, X., Jiang, Z., Li, X., Wei, H. ... Tang, X. (2017). Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1. Molecular Medicine Reports, 16, 3587-3593. https://doi.org/10.3892/mmr.2017.7004
MLA
Wang, C., Zou, W., Liang, X., Jiang, Z., Li, X., Wei, H., Tang, Y., Zhang, P., Tang, X."Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1". Molecular Medicine Reports 16.3 (2017): 3587-3593.
Chicago
Wang, C., Zou, W., Liang, X., Jiang, Z., Li, X., Wei, H., Tang, Y., Zhang, P., Tang, X."Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1". Molecular Medicine Reports 16, no. 3 (2017): 3587-3593. https://doi.org/10.3892/mmr.2017.7004