ATP-sensitive K+ channels contribute to the protective effects of exogenous hydrogen sulfide against high glucose-induced injury in H9c2 cardiac cells

  • Authors:
    • Weijie Liang
    • Jingfu Chen
    • Liqiu Mo
    • Xiao Ke
    • Wenzhu Zhang
    • Dongdan Zheng
    • Wanying Pan
    • Shaoyun Wu
    • Jianqiang Feng
    • Mingcai Song
    • Xinxue Liao
  • View Affiliations

  • Published online on: January 25, 2016     https://doi.org/10.3892/ijmm.2016.2467
  • Pages: 763-772
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hyperglycemia, as well as diabetes mellitus, has been shown to impair ATP-sensitive K+ (KATP) channels in human vascular smooth muscle cells. Hydrogen sulfide (H2S) is also known to be an opener of KATP channels. We previously demonstrated the cardioprotective effects exerted by H2S against high-glucose (HG, 35 mM glucose)-induced injury in H9c2 cardiac cells. As such, we hypothesized that KATP channels play a role in the cardioprotective effects of H2S against HG-induced injury. In this study, to examine this hypothesis, H9c2 cardiac cells were treated with HG for 24 h to establish a model of HG-induced insults. Our findings revealed that treatment of the cells with HG markedly decreased the expression level of KATP channels. However, the decreased expression of KATP channels was reversed by the treatment of the cells with 400 µM sodium hydrogen sulfide (NaHS, a donor of H2S) for 30 min prior to exposure to HG. Additionally, the HG-induced cardiomyocyte injuries, including cytotoxicity, apoptosis, oxidative stress and mitochondrial damage, were ameliorated by treatment with NaHS or 100 µM diazoxide (a mitochondrial KATP channel opener) or 50 µM pinacidil (a non-selective KATP channel opener) for 30 min prior to exposure to HG, as indicated 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). Notably, treatment of the H9c2 cardiac cells with 100 µM 5-hydroxydecanoic acid (5-HD, a mitochondrial KATP channel blocker) or 1 mM glibenclamide (Gli, a non-selective KATP channel blocker) for 30 min prior to treatment with NaHS and exposure to HG significantly attenuated the above-mentioned cardioprotective effects exerted by NaHS. Notably, treatment of the cells with 500 µM N-acetyl‑L‑cysteine (NAC, a scavenger of ROS) for 60 min prior to exposure to HG markedly reduced the HG-induced inhibitory effect on the expression of KATP channels. Taken together, our results suggest that KATP channels play an important role in the cardioprotective effects of exogenous H2S against HG-induced injury. This study also provides novel data demonstraring that there is an antagonistic interaction between ROS and KATP channels in HG-exposed H9c2 cardiac cells.

Introduction

Hydrogen sulfide (H2S), a well-known toxic gas with a characteristic smell of rotten eggs, has been previously described as an endogenously produced labile diffusible gasotransmitter which plays multiple roles in the cardiovascular system in general health and also in diseases (16). For example, in murine models of ischemia-induced heart failure, endogenous and exogenous H2S clearly were shown to exert protective effects against left ventricular structural and functional impairment caused by ischemia-induced heart failure (7). Wang et al reported that H2S attenuated ventricular dysfunction and arrested the progression of heart failure following myocardial infarction (MI) in a rat model (8). In addition, in relation to plasma H2S levels in patients with coronary heart disease (CHD), a significant inverse correlation with the severity of CHD and changes in the coronary artery has been noted (9). Previously, we demonstrated that exogenous H2S protects H9c2 cardiomyocytes against chemical hypoxia- (10,11) or doxorubicin-induced (1214) injury. The roles of H2S in diabetes-related cardiovascular complications have attracted considerable attention, due to the following findings. First, lower circulating H2S concentrations have been noted in animal models of diabetes (5,15,16) and patients with type 2 diabetes mellitus (DM) (5,6). Second, low blood H2S levels may be associated with the vascular inflammation observed in diabetes since the supplementation of H2S prevents the secretion of inflammatory factors by monocytes cultured in high-glucose (HG) medium (5). Third, exogenous H2S protects against the development of HG-induced endothelial dysfunction (15). Fourth, exogenous H2S alleviates myocardial ischemia/reperfusion (IR) injury in db/db mice (17). Fifth, H2S has been shown to exert protective effects against myocardial I/R-induced damage in diabetic rats (18). Furthermore, our recent studies demonstrated that exogenous H2S protects H9c2 cardiac cells against HG-induced injury and inflammation (1921). Although we have reported that several factors, including antioxidant, anti-apoptotic and anti-inflammatory effects, mitochondrial protection, and the inhibition of certain intracellular signaling pathways, such as mitogen-activated protein kinase (MAPK) (19), leptin (20) and nuclear factor-κB (NF-κB) (21), contribute to the protective effects of H2S against HG-induced cardiomyocyte injury, the mechanisms responsible for these cardioprotective effects of H2S remain unclear. Since previous studies have indicated that H2S activates ATP-sensitive K+ (KATP) channels in both the heart (22,23) and vascular tissues (24) and that KATP channels are cardioprotective (23,2529), we thus hypothesized that KATP channels are involved in the protective effects which H2S exerts against HG-induced injury in H9c2 cardiomyocytes.

KATP channels are abundant in cardiac tissue (30). Cardiomyocytes contain KATP channels in both the sarcolemma [surface membrane (31)] and mitochondria (32,33). The opening of sarcolemmal KATP channels is associated with the shortening of cardiac action potential, and a decrease in intracellular Ca2+ loading and cardioprotection during ischemia (3436). The opening of mitochondrial KATP channels contributes to the regulation of cardiac mitochondrial function (37) and cardio-protection induced by ischemic preconditioning (23,37,39). In addition, mitochondrial KATP channels ameliorate the apoptosis induced by oxidative stress in cardiac cells (26). Notably, previous research has revealed that DM is associated with the dysfunction of the cardiovascular KATP channels (40). Hyperglycemia, as well as DM, is harmful to the vasodilation mediated by KATP channels in human vascular smooth muscle cells (4143). However, the roles of both sarcolemmal KATP channels and mitochondrial KATP channels in HG-induced cardiomyocyte injury, in particular, in relation to the cardio-protective effects of exogenous H2S, remain unclear.

Based on our recent studies (1921) and other previous studies (5,6,15,17,18,2229), we hypothesized that H2S exerts cardioprotective effects by modulating the activation of KATP channels in HG-treated cardiomyocytes. Therefore, the present study was designed to examine the following points: i) the effect of HG on the expression of cardiac KATP channels; ii) the roles of both sarcolemmal KATP channels and mitochondrial KATP channels in HG-induced cardiomyocyte injury; iii) whether exogenous H2S protects cardiomyocytes against HG-induced injury by modulating KATP channel activity; and iv) the role of reactive oxygen species (ROS) in the inhibitory effect of HG on the expression of cardiac KATP channels.

Materials and methods

Materials and reagents

Anti-kir6.1 (R-14) antibody (sc-11224) was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA); anti-cleaved caspase-3 antibody (#9662) was purchased from Cell Signaling Technology, Inc. (Boston, MA, USA); anti-GAPDH antibody (10494-1-AP) was purchased from Proteintech Group, Inc. (Wuhan, China); horseradish peroxidase (HRP) conjugated secondary antibody and the BCA Protein assay kit were obtained from KangChen Biotech (Shanghai, China). Diazoxide (DZ), pinacidil (Pin), 5-hydroxydecanoic acid (5-HD) and glibenclamide (Gli) were all purchased from Cayman Chemical Co. (Ann Arbor, MI, USA). Sodium hydrosulfide (NaHS; a donor of H2S) was obtained from Sigma-Aldrich (St. Louis, MO, USA), and was protected from sunlight and stored at 2–4°C. The cell counting kit-8 (CCK-8) was supplied by Dojindo Laboratories (Kumamoto, Japan). 2′,7′-Dichlorofluorescein diacetate (DCFH-DA), 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraeth-ylbenzimidazolylcarbocyanine iodide (JC-1), Hoechst 33258 and N-acetyl-L-cysteine (NAC) were all purchased from Sigma-Aldrich. Fetal bovine serum (FBS) and Dulbecco's modified Eagle's medium (DMEM) medium were obtained from Gibco-BRL (Grand Island, NY, USA). The enhanced chemiluminescence (ECL) solution was purchased from KeyGen Biotech (Nanjing, China). The H9c2 cardiac cells were supplied by the Sun Yat-sen University Experimental Animal Center (Guangzhou, China).

Cell culture and treatment

The H9c2 cardiac cells, a rat cardiac myoblast cell line, were cultured in DMEM, supplemented with 10% FBS in a humidified atmosphere of 95% air and 5% CO2 at 37°C. The culture medium was replaced with fresh medium every 2–3 days and expanded to new culture plates when the cells reached approximately 80% confluence.

To investigate the role of KATP channels in HG-induced cardiomyocyte injury, the H9c2 cardiac cells were treated with 100 µM DZ (a mitochondrial KATP channel opener) or 50 µM Pin (a non-selective KATP channel opener) for 30 min prior to exposure to 35 mM glucose for 24 h. To explore the protective effects of H2S on HG-induced injury, the H9c2 cells were conditioned with 400 µM NaHS for 30 min prior to exposure to HG for 24 h. To further determine whether the protective effects of NaHS were associated with the activation of KATP channels, the cells were conditioned with 100 µM 5-HD (a mitochondrial KATP channel blocker) or 1 mM Gli (a non-selective KATP channel blocker) for 30 min prior to treatment with NaHS and 35 mM glucose for 24 h. To confirm whether there is an antagonistic interaction between ROS and KATP channels, the H9c2 cells were treated with 500 µM NAC (a scavenger of ROS) for 60 min prior to exposure to HG.

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 cells. After being subjected to the above-mentioned treatments, the cells were washed with phosphate-buffered saline (PBS), and 10 µl CCK-8 solution at 10% dilution was added to each well, and the plate was then incubated for approximately 2 h in an incubator. The 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 assessment of apoptosis

Apoptotic cell death was assessed using the Hoechst 33258 staining method followed by photofluorography. The H9c2 cells were plated in 35-mm dishes at a density of 1×106 cells/well. After being subjected to the indicated treatments, the cells were harvested and fixed with paraformaldehyde in 0.1 mol/l PBS (pH 7.4) for 10 min. The slides were then washed 5 times with PBS. After rinsing with PBS, the nuclear DNA was stained with 5 mg/ml Hoechst 33258 for 10 min before being rinsed briefly with PBS and then visualized under a fluorescence microscope (BX50-FLA; Olympus, Tokyo, Japan). The viable H9c2 cells exhibited a uniform blue fluorescence throughout the nucleus, whereas the apoptotic cells had fragmented and condensed nuclei. The experiment was carried out 3 times.

Measurement of intracellular ROS levels

The determination of intracellular ROS levels was performed by measuring the fluorescence product formed by the oxidation of DCFH-DA, as prevoiusly described (19). Briefly, the culture medium was removed and the cells were then washed 3 times with PBS. Following the addition of fresh culture medium, the cells were incubated with DCFH-DA at a final concentration of 10 µM, for 30 min at 37°C. The cells were then washed 5 times with PBS, and the relative amount of fluorescence product was assessed 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 for the amount of ROS. The experiment was carried out 5 times.

Measurement of mitochondrial membrane potential (MMP)

As previousy described (19), MMP was assessed using a fluorescent dye, JC-1, which is a cell-permeable cationic dye that enters the mitochondria based on a highly negative MMP. The depolarization of MMP results in the loss of JC-1 from the mitochondria and a decrease in intracellular green fluorescence. The H9c2 cardiac cells were cultured on a slide with Eagle's minimal essential medium (EMEM). After being subjected to the indicated treatments, the slides were washed 3 times with PBS. The H9c2 cells were incubated with 1 mg/l JC-1 at 37°C for 30 min in an incubator and washed 3 times with PBS. JC-1 fluorescence was then measured over the entire 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 was taken as an index of the levels of MMP. The experiment was carried out 5 times.

Western blot analysis

After being subjected to the indicated treatments, the H9c2 cardiac cells were harvested and lysed with cell lysis solution at 4°C for 30 min. Total proteins in the cell lysates were quantified using a BCA protein assay kit. Loading buffer was added to the cytosolic extracts and, after boiling for approximately 5 min, equal amounts of supernatant from each sample were fractionated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Total proteins in the gel were transferred onto polyvinylidene difluoride (PVDF) membranes. The membranes were blocked for approximately 90 min at room temperature in fresh blocking buffer [0.1% Tween-20 in Tris-buffered saline (TBS-T) containing 5% fat-free milk] and then incubated with either anti-KATP (1:1,000 dilution), or anti-cleaved caspase-3 antibody (1:1,000 dilution) in freshly prepared TBS-T with 3% fat-free milk overnight with slow agitation at 4°C. Following 3 washes with TBS-T, the membranes were incubated with HRP-conjugated goat anti-rabbit secondary antibody (1:2,500 dilution; KangChen Bio-tech) in TBS-T with 3% fat-free milk for 90 min at room temperature. GAPDH was used as an internal control. The membranes were then washed 3 times with TBS-T solution for 15 min. The immunoreative 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. Each experiment was repeated 3 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 13.0 software (SPSS, Inc., Chicago, IL, USA) followed by the least significant difference (LSD) post hoc comparison test. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

NaHS attenuates the HG-induced decrease in protein expression of KATP channels in H9c2 cardiac cells

In order to investigate the influence of HG (35 mM glucose) on the protein expression of KATP channels in H9c2 cardiac cells, a time-response experiment to determine the protein expression levels of KATP channels was performed. As shown in Fig. 1A and B, the cells were exposed to HG for 1, 3, 6, 9, 12 and 24 h, respectively. Following exposure to HG for 6 h, the protein expression level of KATP channels began to decrease, and the maximum decrease in expression levels was observed after the cells were exposed to HG for 12 and 24 h. Based on these results, the expression levels of KATP channels were detected at 12 h following exposure to HG in the subsequent experiments.

It is important to note that the decrease in the KATP channel levels was ameliorated by treatment with 400 µM NaHS (a donor of H2S) for 30 min prior to exposure to HG for 12 h (Fig. 1C and D). However, the basal expression level of KATP channels was not markedly altered by treatment with 400 µM NaHS alone for 30 min. These data indicate that exogenous H2S alleviates the decrease in the protein expression levels of KATP channels induced by HG in H9c2 cardiac cells.

KATP channels are involved in the protective effects which H2S exerts against HG-induced cytotoxicity to H9c2 cardiac cells

Consistent with our recent studies (1921), treatment of the cells with 400 µM NaHS for 30 min prior to exposure to HG for 24 h significantly inhibited HG-induced cytotoxicity, leading to an increase in cell viability (Fig. 2). To explore the role of KATP channels in HG-induced cytotoxicity, the cells were treated with 100 µM DZ (a mitochondrial KATP channel opener) or 50 µM Pin (a non-selective KATP channel opener) for 30 min prior to exposure to HG. As shown in Fig. 2, pre-treatment of the H9c2 cardiac cells with DZ or Pin considerably reduced HG-induced cytotoxicity, as evidenced by an increase in cell viability. To further investigate the role which KATP channels play in the protective effects exerted by H2S against HG-induced cytotoxicity, the cells were treated with 100 µM 5-HD (a mitochondrial KATP channel blocker) or 1 mM Gli (a non-selective KATP channel blocker) for 30 min prior to treatment with NaHS and exposure to HG. It was demonstrated that the blockade of KATP channels with 5-HD or Gli markedly reduced the protective effects of NaHS against HG-induced cytotoxicity, resulting in a decrease in cell viability (Fig. 2). Alone, 5-HD and Gli did not significantly alter cell viability. These data suggest that KATP channels mediate the protective effects of H2S against cytotoxicity to H9c2 cardiac cells induced by HG.

KATP channels are involved in the protective effects which H2S exerts against HG-induced apoptosis in H9c2 cardiac cells

In agreement with our recent studies (1921), exposure of the cells to HG for 24 h markedly increased the number of apoptotic cells (Fig. 3A, panel b), leading to an increase in the percentage of apoptotic cells (Fig. 3A, panel m). The increased number of apoptotic cells was decreased by pre-treatment with NaHS, Pin or DZ (Fig. 3A, panels c, d and e). Similarly, exposure of the cells to 35 mM glucose for 24 h markedly enhanced the expression level of cleaved caspase-3 (Fig. 3B). However, the increased expression level of cleaved caspase-3 was attenuated by treatment with 400 µM NaHS or 50 µM Pin or 100 µM DZ for 30 min prior to exposure to HG for 24 h (Fig. 3B, panels a to d). Furthermore, treatment of the H9c2 cardiac cells with 100 µM 5-HD (Fig. 3A, panel f and B, panels c and d) or 1 mM Gli (Fig. 3A, panel g and B, panels c and d) for 30 min prior to treatment with NaHS and exposure to HG blocked the above-mentioned anti-apoptotic effects of NaHS, as evidenced by the increase in the percentage of apoptotic cells (Fig. 3A, panels f and g) and the increase in cleaved caspase-3 expression (Fig. 3B, panels c and d). Alone, NaHS, Pin, DZ, 5-HD and Gli did not significantly affect the percentage of apoptotic cells or the basal expression level of cleaved caspase-3.

KATP channels are implicated in the protective effects exerted by H2S against HG-induced oxidative stress in H9c2 cardiac cells

In agreement with our recent findings (1921), treatment of the cells with 35 mM glucose for 24 h significantly increased the intracellular production of ROS (Fig. 4B and M). The increased ROS production was ameliorated by treatment with 400 µM NaHS for 30 min prior to exposure to HG for 24 h (Fig. 4C and M). Similarly, treatment with 50 µM Pin (Fig. 4D and M) or 100 µM DZ (Fig. 4E and M) for 30 min prior to exposure to HG for 24 h also attenuated the generation of ROS. To confirm the role played by KATP channels in the protective effects of NaHS against HG-induced oxidative stress, the cells were treated with 100 µM 5-HD or 1 mM Gli for 30 min prior to exposure to NaHS and HG. Our data indicated that pre-treatment with 5-HD (Fig. 4F and M) or Gli (Fig. 4G and M) blocked the inhibitory effects which NaHS exerted on the generation of ROS induced by HG, suggesting that the KATP channels contribute to the protective effects which H2S exerts against the HG-induced overproduction of ROS.

KATP channels are linked to the protective effects of H2S against HG-induced mitochondrial insults in H9c2 cardiac cells

Consistent with our recent studies (1921), treatment of the cells with 35 mM glucose for 24 h markedly induced mitochondrial damage, as evidenced by the loss of MMP (Fig. 5A, panel b and 5M). In addition, we noted that the HG-induced decrease in MMP was reversed by treatment of the cells with 400 µM NaHS for 30 min prior to exposure to HG (Fig. 5A, panel c and 5M). Notably, treatment of the cells with 50 µM Pin (Fig. 5A, panel d and 5M) or 100 µM DZ (Fig. 5A, panel e and 5M) for 30 min prior to exposure to HG also blocked the HG-induced loss of MMP. Subsequently, we further explored the role of KATP channels in the protective effects of NaHS against the dissipation of MMP induced by HG. Our findings revealed that treatment with 100 µM 5-HD or 1 mM Gli for 30 min prior to treatment with NaHS and exposure to HG considerably reduced the inhibitory effects of NaHS on the HG-induced dissipation of MMP (Fig. 5A, panels f and g and 5M). These results demonstrate that KATP channels play a critical role in the protective effects of H2S against the HG-induced mitochondrial damage.

ROS scavenger blocks the HG-induced downregulation of the expression of KATP channels in H9c2 cardiac cells

Since hyperglycemia, as well as DM, has been reported to impair KATP channels in human vascular smooth muscle cells via ROS (4143), we investigated the role of ROS in the HG-induced decrease in KATP channel expression in the H9c2 cardiac cells. As shown in Fig. 6, treatment of the cells with 500 µM NAC (a scavenger of ROS and NAC) for 60 min prior to exposure to 35 mM glucose for 12 h blocked the inhibitory effects of HG on the expression levels of KATP channels. Alone, NAC did not significantly alter the basal expression level of KATP channels. These results suggest that ROS participates in the HG-induced downregulation of the expression of KATP channels.

Discussion

Cardiac KATP channels are key sensors and effectors of the metabolic status of cardiomyocytes, and their roles in HG-induced cardiomyocyte injury and in the cardioprotective effects of H2S are noteworthy. The major findings of this study can be summarized as follows: i) HG markedly downregulated the expression levels of cardiac KATP channels; ii) exogenous H2S attenuated the inhibitory effects of HG on the expression of cardiac KATP channels; iii) the KATP channel openers, DZ (a mitochondrial KATP channel opener) and Pin (a non-selective KATP channel openner), ameliorated the HG-induced cardiomyocyte injury, including cytotoxicity, apoptosis, ROS generation and the dissipation of MMP; iv) the KATP channel antagonists, namely 5-HD (a mitochondrial KATP channel antagonist) and Gli (a non-selective KATP channel antagonist), blocked the cardio-protective effects of exogenous H2S against the HG-induced cardiomyocyte injury; v) the ROS scavenger, NAC, reduced the inhibition of cardiac KATP channel expression induced by HG. These results strongly suggest that the impairment of KATP channels is implicated in HG-induced cardiomyocyte injury and that the activation of KATP channels is linked to the protective effects which exogenous H2S exerts against HG-induced cardiomyocyte injury.

KATP channels have the unique ability to regulate membrane excitability in response to changes in the energetic status of cells (31,44,45). This ability serves to decrease myocardial energy consumption and vulnerability to stress (44,45). Previous research has indicated that the ability of cardiac KATP channels to affect cellular excitability and function depends on their abundance at the membrane surface (46,47). For example, an increase in cardiac sarcolemmal KATP channels enhances the speed and degree of shortening of action potentials and reduces cardiac energy consumption in response to escalating workloads (46). Several studies have indicated a correlation between dysfunction in KATP channel gating and insulin secretory disorders (40,48), such as neonatal diabetes (48). Furthermore, in human vascular smooth muscle cells, HG impairs vasorelaxation by inhibiting the activity of KATP channels (4143). However, our knowledge of the roles of KATP channels in HG-induced cardiac cells remains incomplete.

In order to explore this issue, in the present study, we first investigated the effects of HG on the expression levels of KATP channels in H9c2 cardiac cells. Our data demonstrated that the exposure of the cells to HG markedly reduced the expression levels of KATP channels. This reduced expression level of KATP channels indirectively suggests a decrease in their presence in the HG-treated cardiac cells. Since a previous study indicated that a reduction in the number of sarcolemmal KATP channels slows cardiac action potential during shortening under hypoxia (47),. Thus, we hypothesized that the inhibition of cardiac KATP channels is a critical mechanism which underlies HG-induced cardiomyocyte injury. To confirm this hypothesis, we observed the influence of KATP channel activation on HG-induced injury. In agreement with our recent studies (1921), the findings of this study demonstrated that treatment of the H9c2 cardiac cells with HG induced considerable injuries, including a decrease in cell viability, an increase in apoptotic cells, cleaved caspase-3 expression and ROS generation, as well as the loss of MMP. However, treatment of the H9c2 cardiac cells with DZ (a mitochondrial KATP channel opener) or Pin (a non-selective KATP channel opener) markedly attenuated the above-mentioned HG-induced injuries, as evidenced by an increase in cell viability, and a decrease in the number of apoptotic cells, decreased cleaved caspase-3 expression and ROS generation, as well as the loss of MMP. The above-mentioned results suggest that HG impairs the function of cardiac KATP channels, which contributes to HG-induced injuries. In support of our results are experimental studies which demonstrate that hyperglycemia damages the functionality of the human ether-ago-go-related gene (HERG) K+ channels, reduces the transient outward K+ current, enhances the intracellular concentration of Ca2+, and impairs the excitation-contraction coupling in the heart (49,50). It has also been noted that the opening of the mitochondrial KATP channels plays an important role in the regulation of cardiac mitochondrial function (37) and attenuates the oxidative stress-induced apoptosis of cardiac cells.

Of note, sulfonylurea drugs have been shown to stimulate insulin secretion by closing KATP channels (51,52). Despite the fact that their target is not completely clear, these drugs have been used to treat type 2 DM since the 1950s and they are still used today. Thus, the findings of this study drive us to explore the effects of sulfonylurea drugs on HG-induced cardiomyocyte injury in future experiments.

Another important result of this study relates to the role of the activation of KATP channels in the cardioprotective effects of exogenous H2S against HG-induced cardiac injuries. Previously, the protective effects of H2S on DM-related cardiovascular insults have received much attention. Exogenous H2S attenuates I/R-induced injury in db/db mice (17) and diabetic rats (18). Our recent studies have also examined the protective effects of exogenous H2S against HG-induced injury and inflammation in H9c2 cardiac cells (1921). The mechanisms responsible for these cardioprotective effects of H2S are associated with the inhibition of the MAPK (19), leptin (20) and NF-κB (21) pathways. However, the protective mechanisms of H2S are complex, and other factors are likely involved in these cardioprotective effects of H2S. Since the KATP channels in the heart (22,23) have been reported to be activated by H2S, this study further investigated that roles KATP channels play in the protective effects which H2S exerts against HG-induced cardiomyocyte injury. In agreement with our recent studies, the findings of this study demonstrated that exogenous H2S exerted protective effects against HG-induced cardiomyocyte injuries, as indicated by an increase in cell viability, and a decrease in the number of apoptotic cells, decreased cleaved caspase-3 expression and ROS generation, as well as the loss of MMP. Notably, our results demonstrated that exogenous H2S markedly reduced the downregulation of KATP channel expression by HG and that both 5-HD (a mitochondrial KATP channel blocker) and Gli (a non-selective KATP channel blocker) blocked the cardioprotective effects of H2S mentioned above. These results suggest that KATP channels, in particular mitochondrial KATP channels, play a critical role in the cardioprotective effects of H2S. Similarly, Zhao et al demonstrated the protective effects which H2S exerts against arrhythmia by opening mitochondrial KATP channels (24). However, Bian et al (23) revealed that sarcolemmal KATP channels, but not mitochondrial KATP channels, mediate the cardioprotective effects of H2S in isolated cardiac myocytes exposed to simulated ischemia solution. Therefore, the roles of subtypes of KATP channels involved in the cardioprotective effects of H2S in the present study differ from the ones reported by Bian et al (23), although we did not use a sarcolemmal KATP channels blocker or MCC-134 that opens sarcolemmal KATP channels and blocks minochondrial KATP channels. One possible explanation for this difference in the results may lie in the use of different experimental models.

In the present study, we also investigated the interaction between ROS and cardiac KATP channel activation. As aforementioned, both DZ and Pin ameliorated HG-induced ROS generation, suggesting that KATP channel activity had an inhibitory effect on ROS generation. A previous study reporting that KATP channel opener (DZ) reduces mitochondrial ROS production at reoxygenation (53) supports our results. On the other hand, we noted that NAC, a ROS scavenger, blocked the decrease in the expression of KATP channel protein induced by HG. Collectively, these results suggest that there is an interaction between ROS and KATP channels in HG-treated cardiac cells. The elucidation of the molecular mechanism underlying this interaction may be significant for the treatment and prevention of DM-related cardiovascular complications.

In conclusion, the present study provides novel evidence that the impairment of KATP channels is associated with HG-induced multiple cardiac injuries, including cytotoxicity, apoptosis, oxidative stress and mitochondrial damage. Therefore, the inhibition of cardiac KATP channels by insulin secretagogues should be considered to increase cardiac risk. Importantly, the present study has demonstrated that exogenous H2S protects cardiomyocytes against HG-induced injuries by activating KATP channels. Based on the results of the present study and previous studies (5,6,15,16), we suggest that low levels of endogenous H2S and the impairment of KATP channels are an important pathophysiological mechanism underlying huperglycemia-induced cardiovascular complications. Therefore, supplementation with H2S and modulation of the H2S-KATP channel pathway should be considered a potential approach to attenuating hyperglycemia-induced cardiomyocyte injury.

Acknowledgments

The present study was supported by grants from the National Natural Science Foundation of China (no. 81270296) and the Guangdong Province Finance Science and Technical Fund (no. 2014Sc107).

References

1 

Wang R: Two's company, three's a crowd: can H2S be the third endogenous gaseous transmitter? FASEB J. 16:1792–1798. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Fiorucci S, Distrutti E, Cirino G and Wallace JL: The emerging roles of hydrogen sulfide in the gastrointestinal tract and liver. Gastroenterology. 131:259–271. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Szabo C: Hydrogen sulphide and its therapeutic potential. Nat Rev Drug Discov. 6:917–935. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Li L and Moore PK: Putative biological roles of hydrogen sulfide in health and disease: a breath of not so fresh air? Trends Pharmacol Sci. 29:84–90. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Jain SK, Bull R, Rains JL, Bass PF, Levine SN, Reddy S, McVie R and Bocchini JA Jr: 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

6 

Whiteman M, Gooding KM, Whatmore JL, Ball CI, Mawson D, Skinner K, Tooke JE and Shore AC: 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

7 

Calvert JW, Elston M, Nicholson CK, Gundewar S, Jha S, Elrod JW, Ramachandran A and Lefer DJ: Genetic and pharmacologic hydrogen sulfide therapy attenuates ischemia-induced heart failure in mice. Circulation. 122:11–19. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Wang X, Wang Q, Guo W and Zhu YZ: Hydrogen sulfide attenuates cardiac dysfunction in a rat model of heart failure: a mechanism through cardiac mitochondrial protection. Biosci Rep. 31:87–98. 2011. View Article : Google Scholar

9 

Zhang Y, Tang ZH, Ren Z, Qu SL, Liu MH, Liu LS and Jiang ZS: Hydrogen sulfide, the next potent preventive and therapeutic agent in aging and age-associated diseases. Mol Cell Biol. 33:1104–1113. 2013. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Yang Z, Yang C, Xiao L, Liao X, Lan A, Wang X, Guo R, Chen P, Hu C and Feng J: 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

12 

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

13 

Guo R, Lin J, Xu W, Shen N, Mo L, Zhang C and Feng J: Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibition of the p38 MAPK pathway in H9c2 cells. Int J Mol Med. 31:644–650. 2013.PubMed/NCBI

14 

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/NF-κB pathway in H9c2 cardiac cells. Cell Physiol Biochem. 32:1668–1680. 2013.

15 

Suzuki K, Olah G, Modis K, Coletta C, Kulp G, Gero D, Szoleczky P, Chang T, Zhou Z, Wu L, 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

16 

Ahmad FU, Sattar MA, Rathore HA, Abdullah MH, Tan S, Abdullah NA and Johns EJ: Exogenous hydrogen sulfide (H2S) reduces blood pressure and prevents the progression of diabetic nephropathy in spontaneously hypertensive rats. Ren Fail. 34:203–210. 2012. View Article : Google Scholar

17 

Peake BF, Nicholson CK, Lambert JP, Hood RL, Amin H, Amin S and Calvert JW: 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

18 

Gao Y, Yao X, Zhang Y, Li W, Kang K, Sun L and Sun X: 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

19 

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

20 

Zhuang XD, Hu X, Long M, Dong XB, Liu DH and Liao XX: 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

21 

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.

22 

Geng B, Yang J, Qi Y, Zhao J, Pang Y, Du J and Tang C: H2S generated by heart in rat and its effects on cardiac function. Biochem Biophys Res Commun. 313:362–368. 2004. View Article : Google Scholar

23 

Bian JS, Yong QC, Pan TT, Feng ZN, Ali MY, Zhou S and Moore PK: Role of hydrogen sulfide in the cardioprotection caused by ischemic preconditioning in the rat heart and cardiac myocytes. J Pharmacol Exp Ther. 316:670–678. 2006. View Article : Google Scholar

24 

Zhao W, Zhang J, Lu Y and Wang R: The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener. EMBO J. 20:6008–6016. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Eisen A, Fisman EZ, Rubenfire M, Freimark D, McKechnie R, Tenenbaum A, Motro M and Adler Y: Ischemic preconditioning: nearly two decades of research. A comprehensive review. Atherosclerosis. 172:201–210. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Akao M, Ohler A, O'Rourke B and Marban E: Mitochondrial ATP-sensitive potassium channels inhibit apoptosis induced by oxidative stress in cardiac cells. Circ Res. 88:1267–1275. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Yamada S, Kane GC, Behfar A, Liu XK, Dyer RB, Faustino RS, Miki T, Seino S and Terzic A: Protection conferred by myocardial ATP-sensitive K+ channels in pressure overload-induced congestive heart failure revealed in KCNJ11 Kir6.2-null mutant. J Physiol. 577:1053–1065. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Zingman LV, Alekseev AE, Hodgson-Zingman DM and Terzic A: ATP-sensitive potassium channels: Metabolic sensing and cardioprotection. J Appl Physiol. 103:1888–1893. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Sierra A, Zhu Z, Sapay N, Sharotri V, Kline CF, Luczak ED, Subbotina E, Sivaprasadarao A, Snyder PM, Mohler PJ, et al: Regulation of cardiac ATP-sensitive potassium channel surface expression by calcium/calmodulin-dependent protein kinase II. J Biol Chem. 288:1568–1581. 2013. View Article : Google Scholar :

30 

Nichols CG and Lederer WJ: Adenosine triphosphate-sensitive potassium channels in the cardiovascular system. Am J Physiol. 261:H1675–H1686. 1991.PubMed/NCBI

31 

Noma A: ATP-regulated K+ channels in cardiac muscle. Nature. 305:147–148. 1983. View Article : Google Scholar : PubMed/NCBI

32 

Garlid KD, Paucek P, Yarov-Yarovoy V, Sun X and Schindler PA: The mitochondrial KATP channel as a receptor for potassium channel openers. Biol Chem. 271:8796–8799. 1996. View Article : Google Scholar

33 

Liu Y, Sato T, O'Rourke B and Marban E: Mitochondrial ATP-dependent potassium channels: novel effectors of cardioprotection? Circulation. 97:2463–2469. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Findlay I: Interactive regulation of the ATP-sensitive potassium channel of cardiac muscle. J Cardiovasc Pharmacol. 24:S6–S11. 1994.PubMed/NCBI

35 

López JR, Ghanbari RA and Terzic A: A KATP channel opener protects cardiomyocytes from Ca2 waves: a laser confocal microscopy study. Am J Physiol. 270:H1384–H1389. 1996.

36 

Grover GJ: Pharmacology of ATP-sensitive potassium channel (KATP) openers in models of myocardial ischemia and reperfusion. Can J Physiol Pharmacol. 75:309–315. 1997. View Article : Google Scholar : PubMed/NCBI

37 

Holmuhamedov EL, Jovanović S, Dzeja PP, Jovanović A and Terzic A: Mitochondrial ATP-sensitive K+ channels modulate cardiac mitochondrial function. Am J Physiol. 275:H1567–H1576. 1998.PubMed/NCBI

38 

Yellon DM and Downey JM: Preconditioning the myocardium: from cellular physiology to clinical cardiology. Physiol Rev. 83:1113–1151. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Gross GJ and Peart JN: KATP channels and myocardial preconditioning: an update. Am J Physiol Heart Circ Physiol. 285:H921–H930. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Weintraub NL: Impaired hypoxic coronary vasodilation and ATP-sensitive potassium channel function: a manifestation of diabetic microangiopathy in humans? Circ Res. 92:127–129. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Miura H, Wachtel RE, Loberiza FR Jr, Saito T, Miura M, Nicolosi AC and Gutterman DD: Diabetes mellitus impairs vasodilation to hypoxia in human coronary arterioles: reduced activity of ATP-sensitive potassium channels. Circ Res. 92:151–158. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Kinoshita H, Azma T, Nakahata K, Iranami H, Kimoto Y, Dojo M, Yuge O and Hatano Y: Inhibitory effect of high concentration of glucose on relaxations to activation of ATP-sensitive K+ channels in human omental artery. Arterioscler Thromb Vasc Biol. 24:2290–2295. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Kinoshita H, Azma T, Iranami H, Nakahata K, Kimoto Y, Dojo M, Yuge O and Hatano Y: Synthetic peroxisome proliferator-activated receptor-gamma agonists restore impaired vasorelaxation via ATP-sensitive K+ channels by high glucose. J Pharmacol Exp Ther. 318:312–318. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Seino S and Miki T: Physiological and pathophysiological roles of ATP-sensitive K+ channels. Prog Biophys Mol Biol. 81:133–176. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Flagg TP, Enkvetchakul D, Koster JC and Nichols CG: Muscle KATP channels: recent insights to energy sensing and myoprotection. Physiol Rev. 90:799–829. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Zingman LV, Zhu Z, Sierra A, Stepniak E, Burnett CM, Maksymov G, Anderson ME, Coetzee WA and Hodgson-Zingman DM: Exercise-induced expression of cardiac ATP-sensitive potassium channels promotes action potential shortening and energy conservation. J Mol Cell Cardiol. 51:72–81. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Zhu Z, Burnett CM, Maksymov G, Stepniak E, Sierra A, Subbotina E, Anderson ME, Coetzee WA, Hodgson-Zingman DM and Zingman LV: Reduction in number of sarcolemmal KATP channels slows cardiac action potential duration shortening under hypoxia. Biochem Biophys Res Commun. 415:637–641. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Pearson ER, Flechtner I, Njølstad PR, Malecki MT, Flanagan SE, Larkin B, Ashcroft FM, Klimes I, Codner E, Iotova V, et al: Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med. 355:467–477. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Zhang Y, Han H, Wang J, Wang H, Yang B and Wang Z: Impairment of human ether-à-go-go-related gene (HERG) K+ channel function by hypoglycemia and hyperglycemia. Similar phenotypes but different mechanisms. J Biol Chem. 278:10417–10426. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Smogorzewski M, Galfayan V and Massry SG: High glucose concentration causes a rise in [Ca2+]i of cardiac myocytes. Kidney Int. 53:1237–1243. 1998. View Article : Google Scholar : PubMed/NCBI

51 

Gribble FM and Reimann F: Sulphonylurea action revisited: the post-cloning era. Diabetologia. 46:875–891. 2003. View Article : Google Scholar : PubMed/NCBI

52 

Trube G, Rorsman P and Ohno-Shosaku T: Opposite effects of tolbutamide and diazoxide on the ATP-dependent K+ channel in mouse pancreatic beta-cells. Pflugers Arch. 407:493–499. 1986. View Article : Google Scholar : PubMed/NCBI

53 

Ozcan C, Bienengraeber M, Dzeja PP and Terzic A: Potassium channel openers protect cardiac mitochondria by attenuating oxidant stress at reoxygenation. Am J Physiol Heart Circ Physiol. 282:H531–H539. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 37 Issue 3

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
Liang W, Chen J, Mo L, Ke X, Zhang W, Zheng D, Pan W, Wu S, Feng J, Song M, Song M, et al: ATP-sensitive K+ channels contribute to the protective effects of exogenous hydrogen sulfide against high glucose-induced injury in H9c2 cardiac cells. Int J Mol Med 37: 763-772, 2016
APA
Liang, W., Chen, J., Mo, L., Ke, X., Zhang, W., Zheng, D. ... Liao, X. (2016). ATP-sensitive K+ channels contribute to the protective effects of exogenous hydrogen sulfide against high glucose-induced injury in H9c2 cardiac cells. International Journal of Molecular Medicine, 37, 763-772. https://doi.org/10.3892/ijmm.2016.2467
MLA
Liang, W., Chen, J., Mo, L., Ke, X., Zhang, W., Zheng, D., Pan, W., Wu, S., Feng, J., Song, M., Liao, X."ATP-sensitive K+ channels contribute to the protective effects of exogenous hydrogen sulfide against high glucose-induced injury in H9c2 cardiac cells". International Journal of Molecular Medicine 37.3 (2016): 763-772.
Chicago
Liang, W., Chen, J., Mo, L., Ke, X., Zhang, W., Zheng, D., Pan, W., Wu, S., Feng, J., Song, M., Liao, X."ATP-sensitive K+ channels contribute to the protective effects of exogenous hydrogen sulfide against high glucose-induced injury in H9c2 cardiac cells". International Journal of Molecular Medicine 37, no. 3 (2016): 763-772. https://doi.org/10.3892/ijmm.2016.2467