Exogenous hydrogen sulfide protects against high glucose‑induced inflammation and cytotoxicity in H9c2 cardiac cells

  • Authors:
    • Zena Huang
    • Xiaobian Dong
    • Xiaodong Zhuang
    • Xun Hu
    • Lichun Wang
    • Xinxue Liao
  • View Affiliations

  • Published online on: October 13, 2016     https://doi.org/10.3892/mmr.2016.5846
  • Pages: 4911-4917
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperglycemia serves an important role in the pathogenesis of diabetic cardiomyopathy. The aim of the present study was to investigate whether exogenous hydrogen sulfide (H2S) protects against high glucose‑induced inflammation and cytotoxicity in cardiac cells by inhibiting the p38 mitogen‑activated protein kinase (MAPK)/nuclear factor‑κB (NF‑κB), cyclooxygenase‑2 (COX‑2) and inducible nitric oxide synthase (iNOS) signaling pathways. Rat H9c2 myocardium cells were exposed to 33 mM glucose (high glucose, HG) for 24 h to stimulate HG‑induced cytotoxicity. One group of cells was pretreated with NaHS (a donor of H2S) prior to HG exposure, and cell viability was determined using the Cell Counting Kit‑8 assay. The protein expression levels of p38MAPK, the phosphorylated p65 subunit of NF‑κB, iNOS, COX‑2 and caspase‑3 were analyzed by western blotting, and the protein expression levels of interleukin (IL)‑1β and IL‑6 were detected by enzyme‑linked immunosorbent assay (ELISA). Pretreatment of H9c2 cells with NaHS for 30 min prior to exposure to HG significantly ameliorated the expression of p38MAPK and NF‑κB. In addition, pretreatment with NaHS markedly attenuated p38MAPK/NF‑κB‑mediated cytotoxicity and inflammation, as evidenced by the significant increase in cell viability and decrease in iNOS, COX‑2, IL‑1β and IL‑6 expression levels. Furthermore, treatment of cells with NaHS significantly decreased the expression of caspase‑3, which suggested that NaHS attenuated HG‑induced apoptosis. In conclusion, the results of the present study provided evidence to suggest that exogenous H2S protects against HG‑induced cytotoxicity and inflammation in H9c2 cardiac cells. H2S may exert these cytoprotective effects via inhibition of the p38MAPK/NF‑κB, COX‑2 and iNOS signaling pathways.

Introduction

Over the last 30 years, the number of patients with diabetes has increased >2-fold, which renders it a major threat to human health (1). Among the complications associated with diabetes, diabetic cardiomyopathy (DCM) is characterized by structural and functional alterations in the myocardium, and is one of the leading causes of morbidity and mortality in patients with diabetes worldwide (2,3). Hyperglycemia-induced cardiac inflammation and cytotoxicity are the major pathological causes of DCM. In the hearts of leptin receptor-deficient db/db and streptozotocin (STZ)-induced diabetic mice, hyperglycemia was demonstrated to induce cytotoxicity of cardiac myocytes (4).

Recently, an increasing number of studies have focused on elucidating the signal transduction pathways associated with high glucose (HG)-induced inflammation and cytotoxicity in cardiac tissues (2,5,6). Hyperglycemia may activate the p38 mitogen-activated protein kinase (MAPK) pathway (7), which serves a critical role in the activation of nuclear factor-κB (NF-κB). NF-κB is an essential transcription factor that regulates the expression of proinflammatory genes, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin (IL)-1β and IL-6 (8). In addition, activation of NF-κB may upregulate the transcription of specific genes involved in the inflammatory response (9). iNOS mediates the nitrosylation of caspase-3, which may lead to cardiomyocyte cell death (10). COX-2 has been demonstrated to induce cardiomyocyte apoptosis in several mouse models of cardiomyopathy (11,12). Soetikno et al (13) demonstrated that curcumin prevents DCM in STZ-induced diabetic rats by inhibiting the activation of the p38MAPK/NF-κB signaling pathway. Therefore, molecules that function to inhibit p38MAPK/NF-κB, COX-2 and iNOS activation may protect against HG-induced cardiomyocyte injury.

Hydrogen sulfide (H2S) is synthesized from cysteine by cystathionine gamma lyase, as well as additional naturally occurring enzymes. Along with nitric oxide and carbon monoxide, H2S forms part of a group of biologically active gases termed gasotransmitters or gasomediators (1416). An increasing number of previous studies have demonstrated that H2S is an important cardioprotective agent (17,18). One such study indicated that exogenous H2S exhibits cardioprotective effects by inhibiting oxidative stress and enhancing heat shock protein 90 expression levels (17). Guo et al (19) reported that H2S may protect against doxorubicin-induced inflammation and cytotoxicity in cardiomyocytes by inhibiting the p38MAPK/NF-κB signaling pathway. In diabetic rats and in vitro models, H2S was reported to protect cardiomyocytes from inflammation and cell death (2022). However, the mechanisms underlying these protective effects of H2S in diabetic cardiomyocytes remain unclear. Therefore, the aim of the present study was to investigate the cardioprotective effects of H2S against HG-induced injury in cardiomyocytes, and to determine whether this may involve the p38MAPK/NF-κB, COX-2 and iNOS signaling pathways.

Materials and methods

Reagents

The sodium H2S donor (NaHS), the selective inhibitor of p38MAPK (SB203580), the selective inhibitor of NF-κB (pyrrolidine dithiocarbamate, PDTC), and glucose were purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). The Cell Counting Kit-8 (CCK-8) assay was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Primary antibodies specific to the phosphorylated (p)-p65 as a measure of NF-κB protein expression levels, COX-2, caspase-3, and p38 were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA; cat. nos. 3033, 12282, 9665 and 4511, respectively). The horseradish peroxidase-conjugated goat anti-rabbit IgG secondary antibody (cat. no. KC-RB-035), and the enzyme linked immunosorbent assay (ELISA) kits for assessing IL-1β and IL-6 expression levels were purchased from Zhejiang Kangchen Biotech Co., Inc. (Hangzhou, China). The primary antibody against iNOS (cat no. sc650) was purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). The primary glyceraldehyde 3-phosphate dehydrogenase (GAPDH) antibody (cat. no. HJTW0125) was purchased from Guangzhou Jetway Biotech Co., Ltd. (Guangzhou, China). The interleukin-1 receptor antagonist (IL-1Ra) was purchased from Prospec-Tany TechnoGene, Ltd. (East Brunswick, NJ, USA). The bicinchoninic acid (BCA) protein assay kit was purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). The radioimmunoprecipitation assay (RIPA) buffer was purchased from Beyotime Institute of Biotechnology (Shanghai, China). Low-glucose Dulbecco's modified Eagle's medium-Ham's F12 (DMEM-F12), phosphate-buffered saline (PBS) and fetal bovine serum (FBS) were purchased from Gibco; Thermo Fisher Scientific, Inc. H9c2 cells were obtained from the Sun Yat-sen University Experimental Animal Center (Guangzhou, China).

Cell culture and treatments

H9c2 cardiac cells were cultured in DMEM-F12 medium supplemented with 10% FBS at 37°C and 5% CO2. In order to investigate the cytotoxic effects of HG, H9c2 cells were treated with 11, 22, 33 or 44 mM glucose for 24 h. To investigate the cardioprotective effects of H2S on HG-induced injury, the cells were pretreated with 400 µM NaHS for 30 min prior to HG treatment. To further determine whether the anti-inflammatory effects of H2S were associated with inhibition of p38MAPK/NF-κB pathway, H9c2 cells were pretreated with 3 µM SB203580 (a selective inhibitor of p38MAPK) for 60 min or 100 µM PDTC (a selective inhibitor of NF-κB) for 30 min prior to HG treatment, or were co-treated with HG plus 20 ng/ml IL-1Ra (a selective antagonist of the IL-1β receptor) for 24 h.

Cell viability assay

H9c2 cells were cultured in 96-well plates (1×104 cells/well) and were divided into the following groups: HG, HG+NaHS, HG+SB203580, HG+PDTC, HG+IL-Ra, NaHS, SB203580, PDTC or IL-1Ra) before 10 µl CCK-8 solution was added to each well at a dilution of 1:10, and the cells were incubated for 2 h. The absorbance was measured at 450 nm using a Multiskan MK3 microplate reader (Thermo Fisher Scientific, Inc.). The mean optical density (OD) of five wells for each treatment group was used to calculate the percentage cell viability relative to untreated control cells, according to the following formula: Cell viability (%) = (ODtreatment/ODcontrol) ×100. The experiments were performed in triplicate.

Detection of IL-1β and IL-6 production in the cell culture media using ELISA

H9c2 cells were plated in 60 mm dishes at a density of 1×106 cells/well. Cells were divided into the following treatment groups: HG, HG+NaHS, HG+SB203580, HG+PDTC, HG+IL-Ra, NaHS, SB203580, PDTC or IL-1Ra. The level of IL-1β and IL-6 in the culture media was determined using ELISA, according to the manufacturer's instructions. The absorbance was read at 450 nm, and experiments were performed >5 times.

Extraction of cytoplasmic and nuclear proteins

H9c2 cells were plated in 60 mm dishes at a density of 1×106 cells/well and divided into the following treatment groups: HG, HG+NaHS, HG+SB203580, HG+PDTC, HG+IL-Ra, NaHS, SB203580, PDTC or IL-1Ra. Cytoplasmic and nuclear proteins from H9c2 cells were isolated using the NE-PER Nuclear and Cytoplasmic Extraction Reagent kit, according to the manufacturer's instructions (Thermo Fisher Scientific, Inc.). Briefly, after washing three times with cold PBS, the cells were treated with cytoplasmic protein extraction buffer to isolate cytoplasmic protein fraction proteins. The nuclear proteins were subsequently extracted by adding the nuclear protein extraction buffer. Cytoplasmic and nuclear protein extracts were subject to western blot analysis.

Western blot analysis

H9c2 cells were seeded in 60-mm dishes at a density of 1×106 cells/well and divided into the following treatment groups: HG, HG+NaHS, HG+SB203580, HG+PDTC, HG+IL-Ra, NaHS, SB203580, PDTC or IL-1Ra, Cells were washed in PBS and lysed in RIPA buffer for 30 min, and the homogenate was centrifuged at 21,380 × g for 10 min at 4°C. The total protein concentration of the sample supernatant was determined using a BCA protein assay kit. Total protein (30 µg) was separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The proteins were then transferred onto a polyvinylidene difluoride membrane and the membrane was blocked with 5% non-fat milk diluted in Tris-buffered saline-0.1% Tween 20 (TBS-T) for 1 h at room temperature. The membranes were subsequently incubated with primary antibodies specific to phosphorylated (p)-p65 (dilution, 1:500), iNOS (dilution, 1:1,000), COX-2 (dilution, 1:1,000), caspase-3 (dilution, 1:1,000), p-p38 (dilution, 1:1,000) or GAPDH (dilution, 1:10,000) with gentle agitation at 4°C overnight. The following day, the membranes were incubated with secondary antibodies (dilution, 1:5,000) for 1.5 h at room temperature. Following three washes with TBS-T, the membranes were developed using enhanced chemiluminescence and exposed to X-ray films. To quantify protein expression levels, the X-ray films were visualized and analyzed using ImageJ software (version, 1.41; National Institutes of Health, Bethesda, MA, USA).

Statistical analysis

The data are presented as the mean ± standard error. Differences among treatment groups were analyzed using one-way analysis of variance with the least significant difference test. Statistical analyses were performed using the SPSS software program (version, 13.0; SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

HG decreased cell viability in a concentration-dependent and time-dependent manner

In order to investigate the cytotoxic effects of HG, H9c2 cells were treated with 11, 22, 33 or 44 mM glucose for 24 h As shown in Fig. 1, a significant reduction in cell viability was observed following treatment with 33 mM HG when compared with untreated controls (58.10±2.24% reduction; P<0.01). Based on these results, together with those of a previous study (21), treatment of cells with 33 mM HG for 24 h was selected to model the effects of hyperglycemia on cardiac cells in vitro.

Exogenous H2S attenuates HG-induced cytotoxicity in H9c2 cells

As shown in Fig. 2A, exposure of H9c2 cells to HG for 24 h induced cytotoxicity, as evidenced by the significant decrease in cell viability (P<0.01). However, this effect was significantly diminished when the cells were pretreated with 400 µM NaHS for 30 min prior to HG treatment (P<0.01), which suggests that H2S may attenuate HG-induced cytotoxicity. Similarly, pretreatment of cells with SB203580 or PDTC prior to HG treatment, significantly attenuated the cytotoxic effects of HG treatment (SB203580 and PDTC, P<0.01; Fig. 2A). These results demonstrate that the p38MAPK/NF-κB signaling pathway may be involved in HG-induced cytotoxicity. Notably, treatment of H9c2 cells with NaHS alone did not have a significant effect on cell viability (Fig. 2A).

Exogenous H2S inhibits the p38MAPK/NF-κB signaling pathway in HG-treated H9c2 cells

The next aim of the present study was to investigate the effect of H2S on the p38MAPK/NF-κB signaling pathway. As shown in Fig. 2B, exposure of H9c2 cells to HG for 24 h significantly enhanced the protein expression levels of p-p38MAPK when compared with untreated controls (P<0.01). In contrast, pretreatment of cells with 400 µM NaHS for 30 min prior to HG treatment was associated with a significant reduction in p-p38 expression when compared with HG-treated cells (P<0.01; Fig. 2B).

Exposure of H9c2 cells to HG for 24 h significantly increased the expression of NF-κB (P<0.01; Fig. 2C). However, pretreatment of cells with 400 µM NaHS for 30 min or 3 µM SB203580 for 60 min prior to HG treatment was associated with a significant reduction in p-p65 expression levels (NaHS, P<0.01; SB203580, P<0.01; Fig. 2C). These findings suggest that NaHS may inhibit the expression of NF-κB and the p38MAPK/NF-κB signaling pathway following exposure to HG.

Exogenous H2S suppresses the HG-induced production of proinflammatory cytokines by inhibiting the p38MAPK/NF-κB pathway in H9c2 cells

Following exposure to HG for 24 h, the production of COX-2 (P<0.01; Fig. 3A), iNOS (P<0.01; Fig. 3B), IL-1β (P<0.01; Fig. 3C) and IL-6 (P<0.01; Fig. 3D) were significantly increased when compared with the untreated control group. However, pretreatment of cells with 400 µM NaHS for 30 min prior to HG exposure, significantly ameliorated the HG-induced increase in COX-2, iNOS, IL-1β and IL-6 expression levels (COX-2, iNOS, IL-1β and IL-6, P<0.01; Fig. 3A-D). These results suggest that H2S may suppress the production of proinflammatory cytokines in HG-treated H9c2 cells.

Pretreatment of H9c2 cells with 100 µM PDTC for 30 min prior to HG exposure attenuated the HG-induced production of COX-2, iNOS, IL-1β and IL-6 (COX-2, iNOS, IL-1β and IL-6, P<0.01; Fig. 3A-D). In addition, H9c2 cells pretreated with 3 µM SB203580 for 60 min prior to HG exposure demonstrated a significant reduction in IL-1β and IL-6 expression (IL-1β and IL-6, P<0.01; Fig. 3C and D) when compared with HG-treated controls. This suggests that H2S may suppress the HG-induced production of COX-2, iNOS, IL-1β and IL-6 by inhibiting the p38MAPK/NF-κB signaling pathway in H9c2 cells.

Exogenous H2S exhibits anti-inflammatory effects by ameliorating the HG-induced increase in caspase-3 expression

The results presented so far suggest that exogenous H2S may protect against HG-induced cytotoxicity (Fig. 2A) and reduce the production of proinflammatory cytokines (Fig. 3) in H9c2 cells. Therefore, the final aim of the present study was to investigate whether the anti-inflammatory effects of H2S may attenuate HG-induced apoptosis. Following exposure of H9c2 cells to HG for 24 h, the expression levels of caspase-3, which is an important inducer of apoptosis (23), were significantly increased (Fig. 4). However, pretreatment of cells with 400 µM NaHS prior to HG exposure was associated with a significant reduction in caspase-3 expression levels when compared with cells treated with HG alone (P<0.01). Similarly, treatment of cells with HG plus 20 ng/ml IL-1Ra for 24 h significantly attenuated the production of caspase-3 when compared with cells exposed to HG alone (P<0.01; Fig. 4). This suggests that the IL-1 receptor may be involved in mediating the HG-induced increase in caspase-3 expression. These results indicate that exogenous H2S may exhibit anti-inflammatory effects by ameliorating the HG-induced increase in caspase-3 expression, which protects against HG-induced apoptosis.

Discussion

The results of the present study suggest that exogenous H2S protects cardiac cells against HG-induced inflammation and cytotoxicity, and inhibition of the p38MAPK/NF-κB, COX-2 and iNOS signaling pathways may be involved in mediating these cardioprotective effects of H2S in cardiac cells in vitro. In addition, H2S exhibited anti-inflammatory effects by significantly attenuating the HG-induced increase in caspase-3 expression.

Several factors have been implicated in the development of DCM, including metabolic disturbances, myocardial fibrosis, small vessel disease, autonomic dysfunction and insulin resistance (2). Metabolic disturbances, particularly hyperglycemia, initiate the development of DCM (24). Consistent with previous studies (20,25), exposure to 33 mM glucose for 24 h in the present study, was associated with cytotoxicity and the induction of inflammatory responses in H9c2 cells, as evidenced by a decrease in cell viability, increased expression of the apoptosis inducing factor-caspase 3, and an increase in the expression of multiple pro-inflammatory cytokines, including COX-2, iNOS, IL-1β and IL-6. In addition, HG-treated cardiac cells demonstrated an increase in p38MAPK and p-p65 expression, which suggests that the p38MAPK/NF-κB signaling pathway may have been involved in mediating the pro-apoptotic and cytotoxic effects of HG in H9c2 cells.

The physiological importance of H2S was recognized in the last 15 years. Previous studies have demonstrated that H2S influences a wide range of physiological and pathological processes, including blood vessel dilation (2628), arterial contraction (27,29,30), neurotransmission (31), the regulation of inflammation (32,33) and cardioprotection (34,35). In patients with diabetes (36) and in rats with STZ-induced diabetes (36,37), the levels of H2S in the circulation were significantly reduced. Therefore, a significant amount of attention has focused on investigating whether exogenous H2S supplementation may protect cardiac cells from diabetes-induced cardiac injury. Several studies have revealed that exogenous H2S protects against HG-induced cytotoxicity and inflammation in cardiac cells (20,21). A previous in vivo study demonstrated that intraperitoneal or oral administration of H2S reduced myocardial hypertrophy, as well as the degree of fibrosis (18). Consistent with previous studies, the results of the present study demonstrate that exogenous H2S may protect against cytotoxicity and inflammation in HG-treated H9c2 cells.

The present study investigated the mechanisms underlying the cardioprotective effects of H2S against HG-induced cytotoxicity in H9c2 cells. Exogenous H2S inhibited the HG-induced activation of the p38MAPK/NF-κB signaling pathway, and significantly attenuated the HG-induced cytotoxicity of HG-treated H9c2 cells. Consistent with these observations, H2S has been demonstrated to inhibit p38MAPK activity in various cell types, including rat aortic vascular smooth muscle cells (38), microglia (32) and neuroblastoma cells (39). In addition, H2S has been demonstrated to inhibit NF-κB activation and nuclear translocation in macrophages (40), kidney cells (41), pancreatic acinar cells (42) and heart cells (43). It is possible that activation of the p38MAPK signaling pathway may be a common mechanism involved in the pathogenesis of chronic complications associated with diabetes, as the elevation of p-p38MAPK in mouse diabetic cardiomyocytes activates the production of several cytokines (44). NF-κB is a key factor involved in inflammation, which regulates the transcription of >100 genes associated with immune and inflammatory responses (16). The functional consequence of reduced NF-κB activation in cells is a downregulation in the number of activated proinflammatory genes and cytokines, including COX-2, iNOS, IL-1β and IL-6. Zheng et al (45) observed that downregulation of the NF-κB pathway reduced the downstream expression of proinflammatory genes, including IL-1β, IL-6 and tumor necrosis factor-α, which led to attenuation of inflammation-mediated injury of the vascular wall. These data support the role of H2S as a cardioprotective agent against HG-induced inflammation and cytotoxicity through modulating the activity of the p38MAPK/NF-κB signaling pathway in H9c2 cells.

iNOS is an enzyme and can be induced in all cells and tissues through the action of cytokines (46). Activation of iNOS has been associated with the development of cardiovascular complications in rat models of diabetes. Bardell et al (47) demonstrated that the expression of iNOS in the mesenteric arteries is increased in rat models of diabetes. In the present study, the expression levels of iNOS were significantly elevated in cardiac cells following exposure to HG, which were significantly attenuated by H2S treatment or following inhibition of NF-κB. A previous study, involving STZ-induced diabetic rats, demonstrated that iNOS mediated the nitrosylation of GAPDH and caspase-3, which led to cardiomyocyte death (5). Therefore, the p38MAPK/NF-κB and iNOS signaling pathways are likely to be involved in mediating the cardioprotective effects of exogenous H2S against HG-induced inflammation and cytotoxicity in H9c2 cells.

COX is an enzyme involved in the metabolism of arachidonic acid (AA), and the COX-2 isoform is a key enzyme in the conversion of AA to prostaglandins (48). A previous study demonstrated that overexpression of COX-2 is correlated with induction of inflammatory processes (49). In the present study, the level of COX-2 expression was significantly elevated in cardiac cells following HG exposure. In addition, exposure of cardiac H9c2 cells to H2S or a specific inhibitor to NF-κB, abolished the HG-induced increase in COX-2 expression. In a rat model of heart ischemia/reperfusion injury (50), inhibition of COX-2 was associated with a reduction in cellular apoptosis. Consistent with these observations, the results of the present study suggest that H2S may attenuate apoptosis of H9c2 cells following HG exposure by reducing caspase-3 expression levels. Therefore, H2S may protect cardiac cells by inhibiting COX-2 expression potentially through modulating the p38MAPK/NF-κB signaling pathway.

In the present study, pretreatment of H9c2 cells with an IL-1Ra prior to HG exposure significantly reduced caspase-3 expression. This suggests that inhibition of inflammation may protect against HG-induced apoptosis in H9c2 cells. Consistent with this observation, inhibition of the inflammatory response significantly reduced doxorubicin-induced cytotoxicity of H9c2 cells (19). In addition, p38MAPK was demonstrated to promote apoptosis in a number of in vitro models, such as pulmonary microvascular endothelial cells (51) and breast cancer cells (52), while H2S prevented apoptosis by inhibition of p38MAPK (53). Therefore, the p38MAPK/NF-κB pathway-mediated activation of the anti-inflammatory response may be associated with the cardioprotective effects of exogenous H2S treatment against HG-induced cytotoxicity in H9c2 cells.

In conclusion, the results of the present study demonstrated that exogenous H2S exhibits cardioprotective effects against HG-induced cytotoxicity and inflammation in rat cardiac cells in vitro. H2S exerted cytoprotective effects potentially via the p38MAPK/NF-κB-mediated anti-inflammatory and antiapoptotic signaling pathways. The results provide evidence to suggest that H2S may have a potential therapeutic value in hyperglycemia-induced cardiac lesions, which are predominant in DCM.

Acknowledgements

The present study was supported by the National Nature Science Foundation of China (grant no. 81270296) and the Social Development Project of Guangdong Province in China (grant no. 2014SC107).

References

1 

Chen L, Magliano DJ and Zimmet PZ: The worldwide epidemiology of type 2 diabetes mellitus-present and future perspectives. Nat Rev Endocrinol. 8:228–236. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Goyal BR and Mehta AA: Diabetic cardiomyopathy: Pathophysiological mechanisms and cardiac dysfuntion. Hum Exp Toxicol. 32:571–590. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Poornima IG, Parikh P and Shannon RP: Diabetic cardiomyopathy: The search for a unifying hypothesis. Circ Res. 98:596–605. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Shen E, Li Y, Li Y, Shan L, Zhu H, Feng Q, Arnold JM and Peng T: Rac1 is required for cardiomyocyte apoptosis during hyperglycemia. Diabetes. 58:2386–2395. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Puthanveetil P, Zhang D, Wang Y, Wang F, Wan A, Abrahani A and Rodrigues B: Diabetes triggers a PARP1 mediated death pathway in the heart through participation of FoxO1. J Mol Cell Cardiol. 53:677–686. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Evans JL, Goldfine ID, Maddux BA and Grodsky GM: Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocr Rev. 23:599–622. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Igarashi M, Wakasaki H, Takahara N, Ishii H, Jiang ZY, Yamauchi T, Kuboki K, Meier M, Rhodes CJ and King GL: Glucose or diabetes activates p38 mitogen-activated protein kinase via different pathways. J Clin Invest. 103:185–195. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Crowell JA, Steele VE, Sigman CC and Fay JR: Is inducible nitric oxide synthase a target for chemoprevention? Mol Cancer Ther. 2:815–823. 2003.PubMed/NCBI

9 

Cho SY, Park SJ, Kwon MJ, Jeong TS, Bok SH, Choi WY, Jeong WI, Ryu SY, Do SH, Lee CS, et al: Quercetin suppresses proinflammatory cytokines production through MAP kinases and NF-kappaB pathway in lipopolysaccharide-stimulated macrophage. Mol Cell Biochem. 243:153–160. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Puthanveetil P, Zhang D, Wang Y, Wang F, Wan A, Abrahani A and Rodrigues B: Diabetes triggers a PARP1 mediated death pathway in the heart through participation of FoxO1. J Mol Cell Cardiol. 53:677–686. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Jenke A, Wilk S, Poller W, Eriksson U, Valaperti A, Rauch BH, Stroux A, Liu P, Schultheiss HP, Scheibenbogen C and Skurk C: Adiponectin protects against Toll-like receptor 4-mediated cardiac inflammation and injury. Cardiovasc Res. 99:422–431. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Streicher JM, Kamei K, Ishikawa TO, Herschman H and Wang Y: Compensatory hypertrophy induced by ventricular cardiomyocyte-specific COX-2 expression in mice. J Mol Cell Cardiol. 49:88–94. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Soetikno V, Sari FR, Sukumaran V, Lakshmanan AP, Mito S, Harima M, Thandavarayan RA, Suzuki K, Nagata M, Takagi R and Watanabe K: Curcumin prevents diabetic cardiomyopathy in streptozotocin-induced diabetic rats: Possible involvement of PKC-MAPK signaling pathway. Eur J Pharm Sci. 47:604–614. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Liu YH, Lu M, Hu LF, Wong PT, Webb GD and Bian JS: Hydrogen sulfide in the mammalian cardiovascular system. Antioxid Redox Signal. 17:141–185. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Holwerda KM, Karumanchi SA and Lely AT: Hydrogen sulfide: Role in vascular physiology and pathology. Curr Opin Nephrol Hypertens. 24:170–176. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Li L, Bhatia M and Moore PK: Hydrogen sulphide-A novel mediator of inflammation? Curr Opin Pharmacol. 6:125–129. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Chen SL, Yang CT, Yang ZL, Guo RX, Meng JL, Cui Y, Lan AP, Chen PX and Feng JQ: Hydrogen sulphide protects H9c2 cells against chemical hypoxia-induced injury. Clin Exp Pharmacol Physiol. 37:316–321. 2010. View Article : Google Scholar : PubMed/NCBI

18 

El-Seweidy MM, Sadik NA and Shaker OG: Role of sulfurous mineral water and sodium hydrosulfide as potent inhibitors of fibrosis in the heart of diabetic rats. Arch Biochem Biophys. 506:48–57. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Guo R, Wu K, Chen J, Mo L, Hua X, Zheng D, Chen P, Chen G, Xu W and Feng J: Exogenous hydrogen sulfide protects against doxorubicin-induced inflammation and cytotoxicity by inhibiting p38MAPK/NFκB pathway in H9c2 cardiac cells. Cell Physiol Biochem. 32:1668–1680. 2013.PubMed/NCBI

20 

Xu W, Wu W, Chen J, Guo R, Lin J, Liao X and Feng J: Exogenous hydrogen sulfide protects H9c2 cardiac cells against high glucose-induced injury by inhibiting the activities of the p38 MAPK and ERK1/2 pathways. Int J Mol Med. 32:917–925. 2013.PubMed/NCBI

21 

Wei WB, Hu X, Zhuang XD, Liao LZ and Li WD: GYY4137, a novel hydrogen sulfide-releasing molecule, likely protects against high glucose-induced cytotoxicity by activation of the AMPK/mTOR signal pathway in H9c2 cells. Mol Cell Biochem. 389:249–256. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Zhou X and Lu X: Hydrogen sulfide inhibits high-glucose-induced apoptosis in neonatal rat cardiomyocytes. Exp Biol Med (Maywood). 238:370–374. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Fulda S: Targeting apoptosis for anticancer therapy. Semin Cancer Biol. 31:84–88. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Mortuza R and Chakrabarti S: Glucose-induced cell signaling in the pathogenesis of diabetic cardiomyopathy. Heart Fail Rev. 19:75–86. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Rodrigues B, Cam MC and McNeill JH: Metabolic disturbances in diabetic cardiomyopathy. Mol Cell Biochem. 180:53–57. 1998. View Article : Google Scholar : PubMed/NCBI

26 

Cheng Y, Ndisang JF, Tang G, Cao K and Wang R: Hydrogen sulfide-induced relaxation of resistance mesenteric artery beds of rats. Am J Physiol Heart Circ Physiol. 287:H2316–H2323. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Ali MY, Ping CY, Mok YY, Ling L, Whiteman M, Bhatia M and Moore PK: Regulation of vascular nitric oxide in vitro and in vivo; a new role for endogenous hydrogen sulphide? Br J Pharmacol. 149:625–634. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Hosoki R, Matsuki N and Kimura H: The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem Biophys Res Commun. 237:527–531. 1997. View Article : Google Scholar : PubMed/NCBI

29 

Kiss L, Deitch EA and Szabo C: Hydrogen sulfide decreases adenosine triphosphate levels in aortic rings and leads to vasorelaxation via metabolic inhibition. Life Sci. 83:589–594. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Lim JJ, Liu YH, Khin ES and Bian JS: Vasoconstrictive effect of hydrogen sulfide involves downregulation of cAMP in vascular smooth muscle cells. Am J Physiol Cell Physiol. 295:C1261–C1270. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Abe K and Kimura H: The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci. 16:1066–1071. 1996.PubMed/NCBI

32 

Hu LF, Wong PT, Moore PK and Bian JS: Hydrogen sulfide attenuates lipopolysaccharide-induced inflammation by inhibition of p38 mitogen-activated protein kinase in microglia. J Neurochem. 100:1121–1128. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Li L, Bhatia M and Moore PK: Hydrogen sulphide-a novel mediator of inflammation? Curr Opin Pharmacol. 6:125–129. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Johansen D, Ytrehus K and Baxter GF: Exogenous hydrogen sulfide (H2S) protects against regional myocardial ischemia-reperfusion injury-Evidence for a role of K ATP channels. Basic Res Cardiol. 101:53–60. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Elrod JW, Calvert JW, Morrison J, Doeller JE, Kraus DW, Tao L, Jiao X, Scalia R, Kiss L, Szabo C, et al: Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc Natl Acad Sci USA. 104:15560–15565. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Jain SK, Bull R, Rains JL, Bass PF, Levine SN, Reddy S, McVie R and Bocchini JA: Low levels of hydrogen sulfide in the blood of diabetes patients and streptozotocin-treated rats causes vascular inflammation? Antioxid Redox Signal. 12:1333–1337. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Yusuf M, Huat BT Kwong, Hsu A, Whiteman M, Bhatia M and Moore PK: Streptozotocin-induced diabetes in the rat is associated with enhanced tissue hydrogen sulfide biosynthesis. Biochem Biophys Res Commun. 333:1146–1152. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Du J, Hui Y, Cheung Y, Bin G, Jiang H, Chen X and Tang C: The possible role of hydrogen sulfide as a smooth muscle cell proliferation inhibitor in rat cultured cells. Heart Vessels. 19:75–80. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Hu LF, Lu M, Wu ZY, Wong PT and Bian JS: Hydrogen sulfide inhibits rotenone-induced apoptosis via preservation of mitochondrial function. Mol Pharmacol. 75:27–34. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Oh GS, Pae HO, Lee BS, Kim BN, Kim JM, Kim HR, Jeon SB, Jeon WK, Chae HJ and Chung HT: Hydrogen sulfide inhibits nitric oxide production and nuclear factor-kappaB via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide. Free Radic Biol Med. 41:106–119. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Tripatara P, Patel NS, Collino M, Gallicchio M, Kieswich J, Castiglia S, Benetti E, Stewart KN, Brown PA, Yaqoob MM, et al: Generation of endogenous hydrogen sulfide by cystathionine gamma-lyase limits renal ischemia/reperfusion injury and dysfunction. Lab Invest. 88:1038–1048. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Tamizhselvi R, Moore PK and Bhatia M: Inhibition of hydrogen sulfide synthesis attenuates chemokine production and protects mice against acute pancreatitis and associated lung injury. Pancreas. 36:e24–e31. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Sivarajah A, McDonald MC and Thiemermann C: The production of hydrogen sulfide limits myocardial ischemia and reperfusion injury and contributes to the cardioprotective effects of preconditioning with endotoxin, but not ischemia in the rat. Shock. 26:154–161. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Boudina S and Abel ED: Diabetic cardiomyopathy, causes and effects. Rev Endocr Metab Disord. 11:31–39. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Zheng X, Zhu S, Chang S, Cao Y, Dong J, Li J, Long R and Zhou Y: Protective effects of chronic resveratrol treatment on vascular inflammatory injury in steptozotocin-induced type 2 diabetic rats: Role of NF-kappaB signaling. Eur J Pharmacol. 720:147–157. 2013. View Article : Google Scholar : PubMed/NCBI

46 

MacMicking J, Xie QW and Nathan C: Nitric oxide and macrophage function. Annu Rev Immunol. 15:323–350. 1997. View Article : Google Scholar : PubMed/NCBI

47 

Bardell AL and MacLeod KM: Evidence for inducible nitric-oxide synthase expression and activity in vascular smooth muscle of streptozotocin-diabetic rats. J Pharmacol Exp Ther. 296:252–259. 2001.PubMed/NCBI

48 

Alhouayek M and Muccioli GG: COX-2-derived endocannabinoid metabolites as novel inflammatory mediators. Trends Pharmacol Sci. 35:284–292. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Zamorano B and Carmona MT: Prostaglandin-E2 and cyclic adenosine 3′-5′ monophosphate levels in the hypertrophied rat heart. Biol Res. 25:85–89. 1992.PubMed/NCBI

50 

Song ZF, Chen DY, DU B and Ji XP: Poly (ADP-ribose) polymerase inhibitor reduces heart ischaemia/reperfusion injury via inflammation and Akt signalling in rats. Chin Med J (Engl). 126:1913–1917. 2013.PubMed/NCBI

51 

Liu ZF, Zheng D, Fan GC, Peng T and Su L: Heat stress prevents lipopolysaccharide-induced apoptosis in pulmonary microvascular endothelial cells by blocking calpain/p38 MAPK signalling. Apoptosis. 21:896–904. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Dong Y, Yin S, Song X, Huo Y, Fan L, Ye M and Hu H: Involvement of ROS-p38-H2AX axis in novel curcumin analogues-induced apoptosis in breast cancer cells. Mol Carcinog. 55:323–334. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Han OJ, Joe KH, Kim SW, Lee HS, Kwon NS, Baek KJ and Yun HY: Involvement of p38 mitogen-activated protein kinase and apoptosis signal-regulating kinase-1 in nitric oxide-induced cell death in PC12 cells. Neurochem Res. 26:525–532. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

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
Huang Z, Dong X, Zhuang X, Hu X, Wang L and Liao X: Exogenous hydrogen sulfide protects against high glucose‑induced inflammation and cytotoxicity in H9c2 cardiac cells. Mol Med Rep 14: 4911-4917, 2016
APA
Huang, Z., Dong, X., Zhuang, X., Hu, X., Wang, L., & Liao, X. (2016). Exogenous hydrogen sulfide protects against high glucose‑induced inflammation and cytotoxicity in H9c2 cardiac cells. Molecular Medicine Reports, 14, 4911-4917. https://doi.org/10.3892/mmr.2016.5846
MLA
Huang, Z., Dong, X., Zhuang, X., Hu, X., Wang, L., Liao, X."Exogenous hydrogen sulfide protects against high glucose‑induced inflammation and cytotoxicity in H9c2 cardiac cells". Molecular Medicine Reports 14.5 (2016): 4911-4917.
Chicago
Huang, Z., Dong, X., Zhuang, X., Hu, X., Wang, L., Liao, X."Exogenous hydrogen sulfide protects against high glucose‑induced inflammation and cytotoxicity in H9c2 cardiac cells". Molecular Medicine Reports 14, no. 5 (2016): 4911-4917. https://doi.org/10.3892/mmr.2016.5846