Abstract

Hydrogen sulfide (H2S), an endogenous gasotransmitter, possesses multiple physiological and pharmacological properties including anti-apoptotic, anti-oxidative stress and cardiac protective activities in diabetic cardiomyopathy. An increasing body of evidence has suggested that signal transducer and activator of transcription 3 (STAT3) has beneficial effects in the heart. However, the effect of diabetes on the phosphorylation or activation of cardiac STAT3 appears to be controversial. The present study was designed to investigate the precise function of the STAT3/hypoxia-inducible factor-1α (HIF-1α) signaling pathway in high glucose (HG)-induced H9c2 cardiomyocyte injury and the function of the STAT3/HIF-1α pathway in the cardioprotective action of H2S. The results revealed that GYY4137 pretreatment substantially ameliorated the HG-induced decrease in cell viability and the increase in lactate dehydrogenase (LDH) release in H9c2 cells. Additionally, HG treatment resulted in the upregulation of the phosphorylated (p)-STAT3/STAT3 ratio and HIF-1α protein expression in H9c2 cells, indicating that the activation of the STAT3/HIF-1α pathway was induced by HG. STAT3/HIF-1α pathway inhibition induced by transfection with STAT3 small interfering (si)-RNA attenuated the HG-induced downregulation of cell viability and the upregulation of LDH release. Furthermore, STAT3 siRNA transfection and GYY4137 pretreatment combined attenuated HG-induced apoptosis as illustrated by the decrease in the number of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells, caspase-3 activity, apoptosis ratio and BCL2 associated X, apoptosis regulator/BCL2 apoptosis regulator ratio in H9c2 cells. In addition, STAT3 siRNA transfection and GYY4137 blocked HG-induced oxidative stress as evidenced by the decrease in reactive oxygen species generation, malondialdehyde content and NADPH oxidase 2 expression, and the increase in superoxide dismutase activity and glutathione level. Notably, GYY4137 pretreatment was revealed to reduce the p-STAT3/STAT3 ratio and HIF-1α protein expression, resulting in the inhibition of the STAT3/HIF-1α signaling pathway in HG-treated H9c2 cells. Altogether, the present results demonstrated that H2S mitigates HG-induced H9c2 cell damage, and reduces apoptosis and oxidative stress by suppressing the STAT3/HIF-1α signaling pathway.

Highlights

  • Diabetes mellitus is a common metabolic disorder disease that is characterized by impaired glucose tolerance and is closely associated with excess cardiovascular morbidity and mortality [1]

  • The Cell Counting Kit‐8 (CCK‐8) results revealed that the cell viability was significantly decreased following 33 and 44 mM glucose treatment for 48 h when compared with the normal glucose group (5.5 mM; P

  • GYY4137 treatment alone produced no such effect. These results indicated that exogenous H2S prevents H9c2 cells against HG‐induced H9c2 cell injury

Read more

Summary

Introduction

Diabetes mellitus is a common metabolic disorder disease that is characterized by impaired glucose tolerance and is closely associated with excess cardiovascular morbidity and mortality [1]. Diabetic cardiomyopathy (DCM), a diabetes‐specific complication, is characterized by systolic and autonomic dysfunction independent of hypertension, hyperlipidemia or coronary artery disease [2,3]. A number of studies have revealed that a number of mechanisms are involved in the pathogenesis of DCM, including myocardial insulin resistance, oxidative stress, mitochondrion dysfunction, inflammation and cardiomyocyte apoptosis [3,4]. Among all these events, persistent hyperglycemia in diabetes provokes the excessive production of reactive oxygen species (ROS), resulting in oxidative stress which contributes to the development and pathogenesis of DCM [5,6].

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call