Exogenous H2S protects H9c2 cardiac cells against high glucose-induced injury and inflammation by inhibiting the activation of the NF-κB and IL-1β pathways

  • Authors:
    • Wenming Xu
    • Jingfu Chen
    • Jianchong Lin
    • Donghong Liu
    • Liqiu Mo
    • Wanying Pan
    • Jianqiang Feng
    • Wen Wu
    • Dongdan Zheng
  • View Affiliations

  • Published online on: November 19, 2014     https://doi.org/10.3892/ijmm.2014.2007
  • Pages: 177-186
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperglycemia has been reported to activate the nuclear factor‑κB (NF‑κB) pathway. We have previously demonstrated that exogenous hydrogen sulfide (H2S) protects cardiomyocytes against high glucose (HG)‑induced injury by inhibiting the activity of p38 mitogen‑activated protein kinase (MAPK), which can activate the NF‑κB pathway and induce interleukin (IL)‑1β production. In the present study, we aimed to investigate the hypothesis that exogenous H2S protects cardiomyocytes against HG‑induced injury and inflammation through the inhibition of the NF‑κB/IL‑1β pathway. H9c2 cardiac cells were treated with 35 mM glucose (HG) for 24 h to establish a model of HG‑induced damage. Our results demonstrated that treatment of the cells with 400 µM sodium hydrogen sulfide (NaHS, a donor of H2S) or 100 µM pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF‑κB) for 30 min prior to exposure to HG markedly attenuated the HG‑induced increase in the expression levels of the phosphorylated (p)‑NF‑κB p65 subunit. Notably, pre‑treatment of the H9c2 cardiac cells with NaHS or PDTC significantly suppressed the HG‑induced injury, including cytotoxicity, apoptosis, oxidative stress and mitochondrial insults, as evidenced by an increase in cell viability, as well as a decrease in the number of apoptotic cells, the expression of cleaved caspase‑3, the generation of reactive oxygen species (ROS) and the dissipation of mitochondrial membrane potential (MMP). In addition, pre‑treatment of the cells with NaHS or PDTC ameliorated the HG‑induced inflammatory response, leading to a decrease in the levels of IL‑1β, IL‑6 and tumor necrosis factor‑α (TNF‑α). Importantly, co‑treatment of the H9c2 cells with 20 ng/ml IL‑1 receptor antagonist (IL‑1Ra) and HG markedly reduced the HG‑induced increase in p‑NF‑κB p65 expression, cytoto­xicity, the number of apoptotic cells, as well as the production of TNF‑α. In conclusion, the present study presents novel mechanistic evidence that exogenous H2S protects H9c2 cardiac cells against HG‑induced inflammation and injury, including cytotoxicity, apoptosis, overproduction of ROS and the dissipation of MMP, by inhibiting the NF‑κB/IL‑1β pathway. We also provide new data indicating that the positive interaction between the NF‑κB pathway and IL‑1β is critical in HG‑induced injury and inflammation in H9c2 cardiac cells.

Introduction

Hyperglycemia is not only one of the main clinical characteristics of diabetes mellitus (DM), but is also a risk factor for the development of cardiovascular complications associated with chronic diabetes, such as diabetic cardiomyopathy (1,2). Multiple factors have been reported to contribute to hyperglycemia-induced cardiac damage, such as oxidative stress (36), mitochondrial dysfunction (58), apoptosis (59) and activation of several signaling molecules, including mitogen-activated protein kinase (MAPK) (3,5,10,11), leptin (6,12) and p53 (7,9). Another signaling molecule involved in hyperglycemia-induced cardiomyocyte damage may be nuclear factor-κB (NF-κB). NF-κB is a dimeric transcription factor that regulates numerous genes associated with stress responses, including oxidative stress, apoptosis and inflammation. The NF-κB family is composed of 5 subunits, Rel A (p65), Rel B, c-Rel, NF-κB1 (p50) and NF-κB2 (p52), each of which may form hemo- or heterodimers. Among these, the nuclear translocation of the p65 subunit is a key step in the activation of NF-κB (13). NF-κB is known to be activated by p38 MAPK (a member of the MAPK family) in cardiomyocytes (1416). Accumulating evidence indicates that NF-κB plays a significant role in cardiac damage induced by various stimuli (1416). Guo et al demonstrated the involvement of the NF-κB pathway in doxorubicin-induced cardiac cytotoxicity, resulting in a decrease in cell viability (14,15). The NF-κB pathway has also been implicated in G-protein-coupled receptor angonist-elicited cardiomyocyte hypertrophy (16). Recently, the roles of NF-κB in hyperglycemia-induced cardiac insults and inflammation have attracted attention. As shown in a previous study, in mice with streptozotocin-induced DM, a marked increase in cardiac hypertrophy, fibrosis and inflammation was observed with a corresponding upregulation of reactive oxygen species (ROS) generation, interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, cellular adhesion molecules and phosphorylated (p)-p38 MAPK expression, and NF-κB activation (17), suggesting that the NF-κB pathway participates in hyperglycemia-induced myocardial damage and inflammation. However, the authors did not observe the effects of the inhibition of NF-κB activation on cardiac injury and inflammation induced by hyperglycemia. Thus, it is important to further clarify the role of the inhibition of NF-κB activation in the protective effects against hyperglycemia-induced cardiac insults and inflammation.

In a recent study, we demonstrated that exogenous hydrogen sulfide (H2S) protects H9c2 cardiac cells against high glucose (HG)-induced injury, including cytotoxicity, apoptosis, oxidative stress and mitochondrial damage, by inhibiting the activation of the p38 MAPK, extracellular signal-regulated protein kinase 1/2 (ERK1/2) (5) and leptin pathways (18). H2S, previously regarded as a poisonous gas, has been recognized as the third endogenous gasotransmitter signaling molecule alongside nitric oxide (NO) and carbon monoxide (19,20). H2S exerts its cytoprotective effects in various models of cardiac injury. In isoproterenol-treated rat hearts, treatment with H2S has been shown to attenuate myocardial necrosis and rescue contractile activity (21). In an in vivo model of mice subjected to ischemia/reperfusion (I/R), pre-treatment with Na2S markedly reduced the extent of myocardial injury, oxidative stress and apoptosis (22). Recently, it was also demonstrated that exogenous H2S protects H9c2 cardiac cells against chemical hypoxia-induced injury by inhibiting oxidative stress and enhancing heat shock protein 90 (HSP90) expression (23,24). Of note, since lower circulating H2S levels have been measured in animal models of diabetes (2527) and patients with type 2 DM (26,28), the role of H2S in hyperglycemia-induced cardiac insults has attracted considerable attention. Peake et al reported that the exogenous administration of H2S (Na2S) ameliorates myocardial I/R injury in db/db mice (4). Furthermore, H2S has been shown to exert protective effects against myocardial I/R-induced injury in diabetic rats (29). Of note, although H2S has been shown to exert cytoprotective and anti-inflammatory effects by inhibiting the NF-κB pathway in doxorubicin-treated cardiac cells (15) or cobalt chloride (CoCl2, a hypoxia mimetic agent)-treated human skin keratinocytes (HaCaT cells) (30), whether exogenous H2S protects cardiomyocytes against HG-induced injury and inflammation by inhibiting NF-κB activation remains unclear. To investigate this hypothesis, in the current study, H9c2 cardiac cells were exposed to 35 mM glucose (HG) to establish a model of HG-induced cardiac injury and inflammation. We then examined the following: i) the effects of exogenous H2S on the HG-induced increase in NF-κB activation; ii) the roles of NF-κB activation in HG-induced cardiac injury and inflammation; iii) the roles of the inhibition of NF-κB activation in the protective effects of exogenous H2S against HG-induced injury and inflammation; and iv) the interaction between NF-κB and IL-1β in H9c2 cardiac cells.

Materials and methods

Materials

Sodium hydrogen sulfide (NaHS), was obtained from Sigma Chemical Co., (St. Louis, MO, USA) protected from sunlight and stored at 2–4°C. Pyrrolidine dithiocarbamate (PDTC), 2′7′-dichlorodihydrofluorescein diacetate (DCFH-DA), 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethyl-benzimidazol-carbocyanine iodide (JC-1) and Hoechst 33258 were purchased from Sigma-Aldrich (Sigma Chemicals Co.) and stored at −20°C. IL-1 receptor antagonist (IL-1Ra) was purchased from ProSpec (Ness-Ziona, Israel). The enzyme-linked immunosorbent assay (ELISA) kits for IL-1β, IL-6 and TNF-α were purchased from Wuhan Boster Biological Technology, Ltd. (Wuhan, China). The Cell Counting kit-8 (CCK-8) was purchased from Dojindo Laboratories (Kumamoto, Japan). Fetal bovine serum (FBS) and DMEM were obtained from Gibco-BRL (Grand Island, NY, USA). Anti-p-NF-κB p65 antibody, anti-total (t)-NF-κB p65 antibody and anti-cleaved caspase-3 antibody were procured from Cell Signaling Technology, Inc. (Boston, MA, USA); horseradish peroxidase (HRP)-conjugated secondary antibody and the BCA protein assay kit were obtained from KangChen Bio-tech, Inc. (Shanghai, China). Enhanced chemiluminescence (ECL) solution was purchased from Nanjing KeyGen Biotech Co. Ltd., (Nanjing, China). Lysis buffer was purchased from the Beyotime Institute of Biotechnology (Shanghai, China) and Urea sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) was purchased from Hangzhou Fude Chemical Co., Ltd. (Hangzhou, China).

Cell culture and treatment

The H9c2 cardiac cells, a rat cardiac myoblast cell line, were supplied by the Sun Yat-sen University Experimental Animal Center (Guangzhou, Guangdong, China). The cells were cultured in DMEM medium supplemented with 10% fetal bovine serum at 37°C under an atmosphere of 5% CO2 and 95% air. For the treatment procedure, the cells were cultured in DMEM (5.5 mM glucose) medium for 12 h piror to the administration of 35 mM glucose (final concentration) for 24 h. The glucose concentration of the control group was 5.5 mM. To explore the protective effects of H2S against HG (35 mM glucose)-induced injury, the cells were treated with 400 μM NaHS (a well-known H2S donor) for 30 min prior to exposure to 35 mM glucose for 24 h. To further determine whether the protective effects of H2S are associated with the inhibition of the acvitation of the NF-κB/IL-1 pathway, the H9c2 cells were treated with 100 μM PDTC (a specific inhibitor of NF-κB) prior to exposure to 35 mM glucose for 24 h or co-treated with 20 ng/ml IL-1Ra (an antagonist of IL-1) and 35 mM glucose for 24 h.

Cell viability assay

The H9c2 cells were seeded in 96-well plates at a concentration of 1×104 cells/ml and incubated at 37°C. CCK-8 assay was employed to assess the viability of the H9c2 cardiac cells. After the indicated treatments, 10 μl CCK-8 solution at a 1/10 dilution was added to each well and then the plate was incubated for 2 h in an incubator. Absorbance at 450 nm was assayed using a microplate reader (Molecular Devices, Sunnyvale, CA, USA). The means of the optical density (OD) of 3 wells in the indicated groups were used to calculate the percentage of cell viability according to the following formula: cell viability (%) = (ODtreatment group/ODcontrol group) ×100%. The experiment was repeated 5 times.

Hoechst 33258 nuclear staining for the determination of apoptosis

Apoptotic cell death was observed by Hoechst 33258 staining followed by photofluorography. In brief, the H9c2 cells were plated in 35-mm dishes at a density of 1×106 cells/well. After the indicated treatments, the cells were fixed with 4% paraformaldehyde in 0.1 mol/l phosphate-buffered saline (PBS, pH 7.4) for 10 min. The slides were then washed 5 times with PBS. After staining with 5 mg/ml Hoechst 33258 for 15 min, the H9c2 cells were washed 5 times with PBS, and the cells were then visualized under a fluorescence microscope (BX50-FLA; Olympus, Tokyo, Japan). Viable H9c2 cells displayed a uniform blue fluorescence throughout the nucleus and a normal nuclear size. However, apoptotic H9c2 cells showed condensed, distorted or fractured nuclei. The experiment was carried out 3 times.

Examination of intracellular ROS generation

Intracellular ROS generation was determined by the oxidative conversion of the cell-permeable oxidation of DCFH-DA to fluorescent DCF. The H9c2 cells were cultured on a slide with DMEM. After the different treatments, the slides were washed 3 times with PBS. DCFH-DA solution (10 μM) in serum-free medium was added to the slides, and the cells were then incubated at 37°C for a further 30 min in an incubator. The slides were washed 5 times with PBS, and DCF fluorescence was measured over the entire field of vision by using a fluorescence microscope connected to an imaging system (BX50-FLA; Olympus). The mean fluorescence intensity (MFI) from 5 random fields was measured using ImageJ 1.47i software and the MFI was used as an index of the amount of ROS. The experiment was carried out 5 times.

Measurement of mitochondrial membrane potential (MMP)

MMP was assessed using a fluorescent dye, JC-1, a cell-permeable cationic dye that preferentially enters the mitochondria based on the highly negative MMP. The depolarization of MMP results in the loss of MMP from the mitochondria and a decrease in red/green fluorescence. The cells were cultured on a slide with DMEM. After the indicated treatments, the slides were washed 3 times with PBS. The cells were incubated with 1 mg/l JC-1 at 37°C for 30 min in an incubator, washed briefly with PBS 3 times and air dried again. Fluorescence was then measured over the whole field of vision using a fluorescence microscope connected to an imaging system (BX50-FLA, Olympus). The MFI of JC-1 from 5 random fields was analyzed using ImageJ 1.47i software, and the MFI was taken as an index of the levels of MMP. The experiment was carried out 5 times.

Western blot analysis

After the indicated treatments, the H9c2 cells were harvested and lysed with cell lysis solution at 4°C for 30 min. Total protein was quantified using the BCA protein assay kit. Loading buffer was added to cytosolic extracts, followed by boiling for 5 min, and the same amounts of supernatant from each sample were fractionated by 10% SDS-PAGE, and then the total proteins were transferred into polyvinylidene difluoride (PVDF) membranes. The membranes were blocked in 5% fat-free milk for 60 min in fresh blocking buffer [0.1% Tween-20 in Tris-buffered saline (TBS-T)] at room temperature, and incubated with either anti-t-NF-κB p65 (1:1,000 dilution), anti-p-NF-κB p65 (1:1,000 dilution), or anti-cleaved caspase-3 (1:1,000 dilution) in freshly prepared TBS-T with 3% free-fat milk overnight with gentle agitation at 4°C. The membranes were washed for 15 min with TBS-T and incubated with HRP-conjugated goat anti-rabbit secondary antibody (1:2,500 dilution; KangChen Bio-tech, Inc.) in TBS-T with 3% fat-free milk for 1.5 h at room temperature. The membranes were then washed 3 times with TBS-T for 15 min. The immunoreactive signals were visualized by ECL detection. In order to quantify protein expression, the X-ray films were scanned and analyzed using ImageJ 1.47i software. The experiment was carried out 3 times.

ELISA for the detection of IL-1β, IL-6 and TNF-α in the culture supernatant

The H9c2 cells were plated in 96-well plates. After the indicated treatments, the levels of IL-1β, IL-6 and TNF-α in the culture medium were determined by ELISA according to the manufacturer’s instructions (Wuhan Boster Biological Technology, Ltd.). The experiment was performed at least 5 times.

Statistical analysis

All data are presented as the means ± SEM. Differences between groups were analyzed by one-way analysis of variance (ANOVA) using SPSS version 13.0 software (SPSS, Inc., Chicago, IL, USA), followed by the least significant difference (LSD) post hoc comparison test. A value of p<0.05 was considered to indicate a statistically significant difference.

Results

NaHS and NF-κB inhibitor ameliorate the HG-induced increase in the expression of p-NF-κB p65 in H9c2 cardiac cells

To explore the effects of HG (35 mM glucose) on the activation of the NF-κB pathway in H9c2 cells, a time-response experiment to determine the expression level of NF-κB was performed. As shown in Fig. 1A–C, after the cells were exposured to HG for 30, 60, 90, 120, 150, 180 and 210 min, the expression levels of the p-NF-κB p65 subunit were significantly enhanced, reaching a peak at 60 and 90 min. However, the expression levels of the t-NF-κB p65 subunit remained unaltered following exposure of the cells to HG for the indicated periods of time. Based on these data, the p-NF-κB expression level was determined at 90 min following exposure to HG in the following experiments.

Of note, the increased expression of p-NF-κB p65 was reduced by treatment of the cells with 400 μM NaHS (a donor of H2S) for 30 min prior to exposure to HG for 90 min (Fig. 1D–F). Similarly, treatment of the cells with 100 μM PDTC, an inhibitor of NF-κB, for 30 min prior to exposure to HG also attenuated the increased expression of p-NF-κB p65 (Fig. 1D and E). The basal expression levels of p-NF-κB p65 (control) were not altered by the separate treatment with 400 μM NaHS or 100 μM PDTC (Fig. 1D and F).

Role of inhibition of NF-κB activation in the protective effects of H2S against HG-induced cytotoxicity in H9c2 cardiac cells

In agreement with our previous studies (5,18), treatment of the cells with 400 μM NaHS for 30 min prior to exposure to HG for 24 h markedly decreased HG-induced cytotoxicity, as indicated by an increase in cell viability (Fig. 2). Since we demonstrated that the expression level of p-NF-κB p65 was increased by HG treatment (Fig. 1), we then wished to confirm the role of NF-κB activation in the HG-induced cytotoxicity. As shown in Fig. 2, treatment of the H9c2 cardiac cells with 100 μM PDTC (an inhibitor of NF-κB) for 30 min prior to exposure to 35 mM glucose markedly blocked the HG-induced cytotoxicity, leading to an increase in cell viability, indicating the involvement of NF-κB activation in HG-induced cytotoxicity.

Role of inhibition of NF-κB activation in the protective effects of H2S against HG-induced apoptosis in H9c2 cells

As illustrated in Fig. 3, exposure of the cells to 35 mM glucose for 24 h induced significant apoptosis, as evidenced by an increase in the number of apoptotic cells (Fig. 3B and G) and in the expression of cleaved caspase-3 (Fig. 3H and J). However, the increased number of apoptotic cells and the cleaved caspase-3 expression level were markedly diminished by treatment with 400 μM NaHS for 30 min prior to exposure to HG (Fig. 3C and G–I). Similarly, treatment of the cells with 100 μM PDTC for 30 min prior to exposure to HG also considerably blocked HG-induced apoptosis, leading to a decrease in the number of apoptotic cells and the expression level of cleaved caspase-3 (Fig. 3D, G, J and K). Alone, NaHS or PDTC did not significantly alter the percentage of apoptotic cells and the basal expression level of cleaved caspase-3. These results reveal that the inhibition of the activation of NF-κB plays a role in the protective effects of exogenous H2S against HG-induced apoptosis.

Role of inhibition of NF-κB activation in the inhibitory effects of H2S on the HG-induced generation of ROS in H9c2 cardiac cells

Consistent with our previous studies (5,6,10,18), exposure of the cells to 35 mM glucose for 24 h markedly increased the intracellular generation of ROS (Fig. 4B). The elevated ROS generation was reduced by treatment of the cells with 400 μM NaHS for 30 min prior to exposure to HG (Fig. 4C). To determine whether the activation of NF-κB plays a role in HG-induced oxidative stress, the H9c2 cells were treated with 100 μM PDTC for 30 min prior to exposure to HG. It was shown that pre-treatment with PDTC markedly reduced the increased intracellular ROS generation induced by HG (Fig. 4D), indicating the contribution of the activation of the NF-κB pathway to HG-induced oxidative stress in H9c2 cells.

Role of the inhibition of NF-κB activation in the protective effects of H2S against the HG-induced dissipation of MMP in H9c2 cardiac cells

It has been reported that HG induces mitochondrial damage in cardiomyocytes (5,6,10,18). As illustrated in Fig. 5B and G, exposure of the cells to 35 mM glucose for 24 h induced the marked dissipation of MMP. The dissipation of MMP was blocked by pre-treatment with 400 μM NaHS for 30 min prior to exposure to HG (Fig. 5C and G). Additionally, treatment of the H9c2 cells with 100 μM PDTC for 30 min prior to exposure to HG for 24 h also markedly reduced the dissipation of MMP, suggesting that the activation of NF-κB participates in the HG-induced loss of MMP in H9c2 cells.

NaHS and NF-κB inhibitor reduce the HG-induced production of pro-inflammatory cytokines in H9c2 cardiac cells

As indicated in Fig. 6, after the cells were treated with 35 mM glucose (HG) for 24 h, the levels of IL-1β (Fig. 6A), IL-6 (Fig. 6B) and TNF-α (Fig. 6C) were markedly enhanced, compared with the control group (p<0.01). However, the increased levels of IL-1β, IL-6 and TNF-α induced by HG were markedly attenuated by treatment with 400 μM NaHS for 30 min prior to exposure to HG for 24 h, revealing the inhibitory effects of exogenous H2S on the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α induced by HG. Similarly, treatment of the cells with 100 μM PDTC for 30 min prior to exposure to HG also alleviated the increased production of IL-1β, IL-6 and TNF-α (Fig. 6A–C), suggesting that the activation of NF-κB contributes to the HG-induced inflammatory response in H9c2 cardiac cells.

IL-1Ra suppresses the HG-induced upregulation of p-NF-κB p65 expression in H9c2 cardiac cells

Since the above results (Fig. 1) revealed that exposure to HG upregulated the expression of the p-NF-κB p65 subunit and that the activation of NF-κB was involved in the production of IL-1β induced by HG (Fig. 6), and a recent study reported that IL-1Ra reduces hyperglycemia and tissue inflammation in type 2 diabetic GK rats (31), we then wished to explore whether IL-1β contributes to the activation of NF-κB by HG. As shown in Fig. 7, exposure of the H9c2 cells to 35 mM glucose for 90 min significantly enhanced the expression level of p-NF-κB p65. However, co-treatment of the cells with 20 ng/ml IL-1Ra and 35 mM glucose for 90 min significantly attenuated the HG-induced increase in p-NF-κB p65 expression (Fig. 7A and B). Alone, IL-1Ra did not affect the basal expression of p-NF-κB in the H9c2 cells (Fig. 7A and C).

IL-1Ra attenuates HG-induced cytotoxicity, apoptosis and TNF-α production in H9c2 cardiac cells

As shown in Fig. 8, co-treatment of the cells with 20 ng/ml IL-1Ra and 35 mM glucose for 24 h markedly attenuated the HG-induced cytotoxicity, apoptosis and TNF-α production, as evidenced by an increase in cell viability (Fig. 8A), as well as by a decrease in the number of apoptotic cells (Fig. 8B, panels b and e) and TNF-α production (Fig. 8C). Alone, IL-1Ra did not affect cell viability, the number of apoptotic cells and the basal production of TNF-α in the H9c2 cells (Fig. 8A–C).

Discussion

To the best of our knowledge, the present study demonstrates for the first time that exogenous H2S protects H9c2 cardiac cells against HG-induced injury and inflammation by inhibiting the activation of NF-κB and IL-1β and that the positive interaction between NF-κB and IL-1β contributes to the HG-induced injury and inflammation. Several lines of evidence support our conclusion: i) exogenous H2S attenuated the HG-induced increase in p-NF-κB p65 expression, and IL-1β production; ii) exogenous H2S inhibited HG-induced cardiomyocyte injury and inflammation, as evidenced by an increase in cell viability, and a decrease in the number of apoptotic cells and cleaved caspase-3 expression, as well as a decrease in ROS production and the dissipation of MMP and the production of IL-1β, IL-6 and TNF-α; iii) similar to the inhibitory effects of exogenous H2S, pre-treatment of the H9c2 cells with PDTC (an inhibitor of NF-κB) also ameliorated the HG-induced injury and inflammation; and iv) co-treatment of the H9c2 cells with IL-Ra (an inhibitor of IL-1β receptor) and HG alleviated the cytotoxicity, apoptosis and upregulation of p-NF-κB expression induced by HG.

NF-κB is a key transcription factor that regulates inflammatory processes (32). Previous studies have demonstrated that NF-κB is involved in cellular damage and inflammation induced by various stimuli, including chemical hypoxia (30), doxorubicin (14,15) and lipopolysaccharide (LPS) (33). NF-κB may participate in some of the downstream effects of NADH oxide on cardiac hypertrophy and it regulates the expression of inflammatory genes, such as TNF-α and IL-6 (34,35). Recently, the pathophysiological roles of the NF-κB pathway in diatetic complications, such as diabetic cardiomyopathy, have attracted considerable attention (17,31,36). In diabetic rats, NF-κB is activated (17,31,36). However, the roles of the NF-κB pathway in HG-induced cardiac injury and inflammation remain poorly understood. In order to explore this, in this study, we first observed the effects of HG on the activation of NF-κB in H9c2 cardiac cells. In agreement with the results of previous studies (17,31,36), it was shown that exposure to HG significantly induced an increase in p-NF-κB expression, suggesting the activation of the NF-κB pathway by HG. Secondly, we investigated the roles of the NF-κB activation in HG-induced injury. Our results demonstrated that pre-treatment of the H9c2 cardiac cells with PDTC (an inhibitor of NF-κB) markedly suppressed HG-induced multiple injury, including cytotoxicity, apoptosis, oxidative stress and mitochondrial damage, as characterized by an increase in cell viability, as well as a decrease in the number of apoptotic cells, cleaved caspase-3 expression, ROS generation and the dissipation of MMP. These findings suggest that NF-κB activation is involved in HG-induced multiple damage in H9c2 cells. Since our (30), as well as other previous studies (37) have shown that ROS generation induces the activation of NF-κB in other cell types, the combination of the results of this study with those of other studies (30,38) suggests that there may be a crosstalk between the ROS and NF-κB pathway in HG-treated H9c2 cells. To confirm this hypothesis, further studies are required. Moreover, since data from diabetic patients (39,40) and diabetic animals (17,31,36) have demonstrated that hyperglycemia is associated with tissue inflammation, in this study, we examined the effects of NF-κB activation on pro-inflammatory factors. Similar to the results of previous studies (17,31,36,39,40), this study demonstrated that exposure to HG induced significant cardiac inflammatory responses, as indicated by the increased production of IL-1β, IL-6 and TNF-α. However, the increased production of IL-1β, IL-6 and TNF-α was reduced by PDTC, suggesting the involvement of NF-κB activation in the HG-induced production of pro-inflammatory factors. These data provide clear evidence that the activation of the NF-κB pathway contributes to HG-induced injury and inflammation in H9c2 cardiac cells, which confirms and adds to the results of previous studies demonstrating the roles of NF-κB activation in hyperglycemia-induced cardiac damage and inflammation (41).

Another major finding of this study relates to a positive interaction between NF-κB and IL-1β which is critical for the induction of cardiomyocyte injury and inflammation induced by HG. This consideration is based on the results of a previous study demonstrating an interaction between NF-κB and IL-1β in aged murine lungs in response to endotoxic stress (42). IL-1 has been shown to play critical roles in the pathogenesis of heart disease (31,43). IL-1 binds to the transmembrane IL-1 type receptor (IL-1R) and initiates a downstream signal. Its natural antagonist, IL-1Ra, competitively interacts with the same receptor to prevent IL-1 signaling (44). Ehses et al demonstrated that IL-Ra reduces hyperglycemia and tissue inflammation in type 2 diabetic rats (31). Similarly, we observed the inhibitory effects of IL-1 Ra on the HG-induced cardiac cytotoxicity, apoptosis and inflammation. In addition, this study confirmed the contribution of NF-κB activation to the HG-induced cytotoxicity, apoptosis, oxidative stress, loss of MMP and inflammation. Therefore, we speculate that there may be an interaction between NF-κB and IL-1β in the induction of injury and inflammation in HG-exposed H9c2 cardiac cells. Significantly, our data demonstrate that PDTC inhibits the HG-induced production of IL-1β and that IL-1Ra attenuates the increased expression of p-NF-κB p65 subunit expression, which clearly confirms our hypothesis. These results provide novel mechanistic evidence for the roles of both NF-κB and IL-1β in the HG-induced cardiac injury and inflammation.

Another important finding of the present study relates to the roles of the inhibition of the NF-κB and IL-1β pathways in the cardioprotective effects of exogenous H2S against HG-induced multiple cardiac injury and inflammation. Accumulating evidence indicates that H2S exerts cardioprotective effects (4,5,15,18,2124). H2S therapy offers protection against several models of myocardial injury in the setting of type 1 (25,29,45) or type 2 (4) diabetes by attenuating apoptosis and oxidative stress. Recently, we demonstrated that exogenous H2S alleviates cytotoxicity, apoptosis, oxidative stress and the loss of MMP by inhibiting the p38 MAPK, ERK1/2 and leptin pathways in HG-treated H9c2 cardiac cells (5,18). Since we have reported that exogenous H2S suppresses the p38 MAPK/NF-κB-mediated inflammatory response and cytotoxicity in doxorubicin-treated H9c2 cardiac cells (15), this study further explored whether H2S protects the cells against HG-induced injury and inflammation by inhibiting the NF-κB and IL-1β pathways. Our findings demonstrated that exogenous H2S markedly reduced NF-κB activation and IL-1β production. In addition, similar to the inhibitory effects of both PDTC and IL-1Ra as indicated above, pre-treatment with H2S markedly attenuated the HG-induced cardiac injury and inflammation. These results suggest that the inhibition of the NF-κB and IL-1β pathways may be a critical mechanism responsible for the protective effects of exogenous H2S against HG-induced cardiac injury and inflammation. Similarily, several recent studies have indicated that exogenous H2S reduces the chemical hypoxia-induced (30) or doxorubicin-induced (15) cell injury and inflammation through the inhibition of NF-κB and IL-1β. These studies (15,30) support our results.

In conclusion, the present study provides novel evidence that a positive interaction between the NF-κB and IL-1β pathways contributes to the HG-induced cardiac injury and inflammation and that exogenous H2S protects H9c2 cardiac cells against HG-induced injury and inflammation through the inhibition of the NF-κB and IL-1β pathways. These findings may aid in the development of novel therapeutic approaches for the prevention and treatment of hyperglycemia-related injury and inflammation.

Acknowledgements

The present study was supported by grants from the Guangdong Medical Research Foundation (no. A2012172), Science and Techonology Planning Project of Guangdong Province, China (nos. 2012B031800358, 2012B031800365 and 2010B08071044) and the Guangdong Natural Science Foundation (S2011010004381).

References

1 

Brownlee M: Biochemistry and molecular cell biology of diabetic complications. Nature. 414:813–820. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Ren J and Davidoff AJ: Diabetes rapidly induces contractile dysfunctions in isolated ventricular myocytes. Am J Physiol. 272:H148–H158. 1997.PubMed/NCBI

3 

Soetikno V, Sari FR, Sukumaran V, et al: 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

4 

Peake BF, Nicholson CK, Lambert JP, et al: Hydrogen sulfide preconditions the db/db diabetic mouse heart against ischemia-reperfusion injury by activating Nrf2 signaling in an Erk-dependent manner. Am J Physiol Heart Circ Physiol. 304:H1215–H1224. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Xu W, Wu W, Chen J, et al: 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

6 

Chen J, Mo H, Guo R, et al: Inhibition of the leptin-induced activation of the p38 MAPK pathway contributes to the protective effects of naringin against high glucose-induced injury in H9c2 cardiac cells. Int J Mol Med. 33:605–612. 2014.PubMed/NCBI

7 

Huang H, Wu K, You Q, et al: Naringin inhibits high glucose-induced cardiomyocyte apoptosis by attenuating mitochondrial dysfunction and modulating the activation of the p38 signaling pathway. Int J Mol Med. 32:396–402. 2013.PubMed/NCBI

8 

Cai L, Li W, Wang G, et al: Hyperglycemia-induced apoptosis in mouse myocardium: mitochondrial cytochrome C-mediated caspase-3 activation pathway. Diabetes. 51:1938–1948. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Fiordaliso F, Leri A, Cesselli D, et al: Hyperglycemia activates p53 and p53-regulated genes leading to myocyte cell death. Diabetes. 50:2363–2375. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Chen J, Guo R, Yan H, et al: Naringin inhibits ROS-activated MAPK pathway in high glucose-induced injuries in H9c2 cardiac cells. Basic Clin Pharmacol Toxicol. 114:293–304. 2014. View Article : Google Scholar

11 

Yan J, Young ME, Cui L, et al: Increased glucose uptake and oxidation in mouse hearts prevent high fatty acid oxidation but cause cardiac dysfunction in diet-induced obesity. Circulation. 119:2818–2828. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Majumdar P, Chen S, George B, et al: Leptin and endothelin-1 mediated increased extracellular matrix protein production and cardiomyocyte hypertrophy in diabetic heart disease. Diabetes Metab Res Rev. 25:452–463. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Baldwin AS Jr: The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 14:649–683. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Guo RM, Xu WM, Lin JC, et al: Activation of the p38 MAPK/NF-κB pathway contributes to doxorubicin-induced inflammation and cytotoxicity in H9c2 cardiac cells. Mol Med Rep. 8:603–608. 2013.PubMed/NCBI

15 

Guo R, Wu K, Chen J, et al: 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.

16 

Hirotani S, Otsu K, Nishida K, et al: Involvement of nuclear factor-kappaB and apoptosis signal-regulating kinase 1 in G-protein-coupled receptor agonist-induced cardiomyocyte hypertrophy. Circulation. 105:509–515. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Thandavarayan RA, Giridharan VV, Sari FR, et al: Depletion of 14-3-3 protein exacerbates cardiac oxidative stress, inflammation and remodeling process via modulation of MAPK/NF-κB signaling pathways after streptozotocin-induced diabetes mellitus. Cell Physiol Biochem. 28:911–922. 2011. View Article : Google Scholar

18 

Zhuang XD, Hu X, Long M, et al: Exogenous hydrogen sulfide alleviates high glucose-induced cardiotoxicity via inhibition of leptin signaling in H9c2 cells. Mol Cell Biochem. 391:147–155. 2014. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Moore PK, Bhatia M and Moochhala S: Hydrogen sulfide: from the smell of the past to the mediator of the future? Trends Pharmacol Sci. 24:609–611. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Geng B, Chang L, Pan C, et al: Endogenous hydrogen sulfide regulation of myocardial injury induced by isoproterenol. Biochem Biophys Res Commun. 318:756–763. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Calvert JW, Jha S, Gundewar S, Elrod JW, et al: Hydrogen sulfide mediates cardioprotection through Nrf2 signaling. Circ Res. 105:365–374. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Chen SL, Yang CT, Yang ZL, et al: Hydrogen sulphide protects H9c2 cells against chemical hypoxia-induced injury. Clin Exp Pharmacol Physiol. 37:316–321. 2010. View Article : Google Scholar

24 

Yang Z, Yang C, Xiao L, et al: Novel insights into the role of HSP90 in cytoprotection of H2S against chemical hypoxia-induced injury in H9c2 cardiac myocytes. Int J Mol Med. 28:397–403. 2011.PubMed/NCBI

25 

Suzuki K, Olah G, Modis K, et al: Hydrogen sulfide replacement therapy protects the vascular endothelium in hyperglycemia by preserving mitochondrial function. Proc Natl Acad Sci USA. 108:13829–13834. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Jain SK, Bull R, Rains JL, et al: 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

27 

Ahmad FU, Sattar MA, Rathore HA, et al: Exogenous hydrogen sulfide (H2S) reduces blood pressure and prevents the progression of diabetic nephropathy in spontaneously hypertensive rats. Ren Fail. 34:203–210. 2012.

28 

Whiteman M, Gooding KM, Whatmore JL, et al: Adiposity is a major determinant of plasma levels of the novel vasodilator hydrogen sulphide. Diabetologia. 53:1722–1726. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Gao Y, Yao X, Zhang Y, et al: The protective role of hydrogen sulfide in myocardial ischemia-reperfusion-induced injury in diabetic rats. Int J Cardiol. 152:177–183. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Yang C, Yang Z, Zhang M, et al: Hydrogen sulfide protects against chemical hypoxia-induced cytotoxicity and inflammation in HaCaT cells through inhibition of ROS/NF-κB/COX-2 pathway. PLoS One. 6:e219712011. View Article : Google Scholar

31 

Ehses JA, Lacraz G, Giroix MH, et al: IL-1 antagonism reduces hyperglycemia and tissue inflammation in the type 2 diabetic GK rat. Proc Natl Acad Sci USA. 106:13998–14003. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Barnes PJ and Karin M: Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med. 336:1066–1071. 1997. View Article : Google Scholar : PubMed/NCBI

33 

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

34 

Satoh M, Ogita H, Takeshita K, et al: Requirement of Rac1 in the development of cardiac hypertrophy. Proc Natl Acad Sci USA. 103:7432–7437. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Liu SF, Ye X and Malik AB: Inhibition of NF-kappaB activation by pyrrolidine dithiocarbamate prevents in vivo expression of proinflammatory genes. Circulation. 100:1330–1337. 1999. View Article : Google Scholar : PubMed/NCBI

36 

Kowluru RA and Kanwar M: Effects of curcumin on retinal oxidative stress and inflammation in diabetes. Nutr Metab (Lond). 4:82007. View Article : Google Scholar

37 

Cichon MA and Radisky DC: ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-κB-dependent activation of Snail. Oncotarget. 5:2827–2838. 2014.PubMed/NCBI

38 

Goldin A, Beckman JA, Schmidt AM and Creager MA: Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation. 114:597–605. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Varo N, Vicent D, Libby P, et al: Elevated plasma levels of the atherogenic mediator soluble CD40 ligand in diabetic patients: a novel target of thiazolidinediones. Circulation. 107:2664–2669. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Aronson D, Bartha P, Zinder O, et al: Association between fasting glucose and C-reactive protein in middle-aged subjects. Diabet Med. 21:39–44. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Kuo WW, Wang WJ, Tsai CY, et al: Diallyl trisufide (DATS) suppresses high glucose-induced cardiomyocyte apoptosis by inhibiting JNK/NFκB signaling via attenuating ROS generation. Int J Cardiol. 168:270–280. 2013. View Article : Google Scholar

42 

Chang CK and LoCicero J 3rd: Overexpressed nuclear factor kappaB correlates with enhanced expression of interleukin-1beta and inducible nitric oxide synthase in aged murine lungs to endotoxic stress. Ann Thorac Surg. 77:1222–1227. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Bujak M and Frangogiannis NG: The role of IL-1 in the pathogenesis of heart disease. Arch Immunol Ther Exp (Warsz). 57:165–176. 2009. View Article : Google Scholar

44 

Dinarello CA, Cannon JG, Mancilla J, et al: Interleukin-6 as an endogenous pyrogen: induction of prostaglandin E2 in brain but not in peripheral blood mononuclear cells. Brain Res. 562:199–206. 1991. View Article : Google Scholar : PubMed/NCBI

45 

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

Related Articles

Journal Cover

January-2015
Volume 35 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu W, Chen J, Lin J, Liu D, Mo L, Pan W, Feng J, Wu W and Zheng D: Exogenous H2S protects H9c2 cardiac cells against high glucose-induced injury and inflammation by inhibiting the activation of the NF-κB and IL-1β pathways. Int J Mol Med 35: 177-186, 2015
APA
Xu, W., Chen, J., Lin, J., Liu, D., Mo, L., Pan, W. ... Zheng, D. (2015). Exogenous H2S protects H9c2 cardiac cells against high glucose-induced injury and inflammation by inhibiting the activation of the NF-κB and IL-1β pathways. International Journal of Molecular Medicine, 35, 177-186. https://doi.org/10.3892/ijmm.2014.2007
MLA
Xu, W., Chen, J., Lin, J., Liu, D., Mo, L., Pan, W., Feng, J., Wu, W., Zheng, D."Exogenous H2S protects H9c2 cardiac cells against high glucose-induced injury and inflammation by inhibiting the activation of the NF-κB and IL-1β pathways". International Journal of Molecular Medicine 35.1 (2015): 177-186.
Chicago
Xu, W., Chen, J., Lin, J., Liu, D., Mo, L., Pan, W., Feng, J., Wu, W., Zheng, D."Exogenous H2S protects H9c2 cardiac cells against high glucose-induced injury and inflammation by inhibiting the activation of the NF-κB and IL-1β pathways". International Journal of Molecular Medicine 35, no. 1 (2015): 177-186. https://doi.org/10.3892/ijmm.2014.2007