Abstract

BackgroundAdministration of propofol, an intravenous anesthetic with antioxidant property, immediately at the onset of post-ischemic reperfusion (propofol postconditioning, P-PostC) has been shown to confer cardioprotection against ischemia–reperfusion injury, while the underlying mechanism remains incompletely understood. The FoxO transcription factors are reported to play critical roles in activating cardiomyocyte survival signaling throughout the process of cellular injuries induced by oxidative stress and are also involved in hypoxic postconditioning mediated neuroprotection, however, the role of FoxO in postconditioning mediated protection in the heart and in particular in high glucose condition is unknown.MethodsRat heart-derived H9c2 cells were exposed to high glucose (HG) for 48 h (h), then subjected to hypoxia/reoxygenation (H/R, composed of 8 h of hypoxia followed by 12 h of reoxygenation) in the absence or presence of postconditioning with various concentrations of propofol (P-PostC) at the onset of reoxygenation. After having identified the optical concentration of propofol, H9c2 cells were subjected to H/R and P-PostC in the absence or presence of FoxO1 or FoxO3a gene silencing to explore their roles in P-PostC mediated protection against apoptotic and autophagic cell deaths under hyperglycemia.ResultsThe results showed that HG with or without H/R decreased cell viability, increased lactate dehydrogenase (LDH) leakage and the production of reactive oxygen species (ROS) in H9c2 cells, all of which were significantly reversed by propofol (P-PostC), especially at the concentration of 25 µmol/L (P25) (all P < 0.05, NC vs. HG; HG vs. HG + HR; HG + HR + P12.5 or HG + HR + P25 or HG + HR + P50 vs. HG + HR). Moreover, we found that propofol (P25) decreased H9c2 cells apoptosis and autophagy that were concomitant with increased FoxO1 and FoxO3a expression (all P < 0.05, HG + HR + P25 vs. HG + HR). The protective effects of propofol (P25) against H/R injury were reversed by silencing FoxO1 or FoxO3a (all P < 0.05, HG + HR + P25 vs. HG + HR + P25 + siRNA-1 or HG + HR + P25 + siRNA-5).ConclusionIt is concluded that propofol postconditioning attenuated H9c2 cardiac cells apoptosis and autophagy induced by H/R injury through upregulating FoxO1 and FoxO3a under hyperglycemia.

Highlights

  • Administration of propofol, an intravenous anesthetic with antioxidant property, immediately at the onset of post-ischemic reperfusion has been shown to confer cardioprotection against ischemia–reperfusion injury, while the underlying mechanism remains incompletely understood

  • Propofol‐postconditioning (P‐PostC) dose dependently alleviated hypoxia/reoxygenation injury in H9c2 cells under hyperglycemia We used the in vitro Hypoxia/Reoxygenation (H/R) model in H9c2 cells exposed to high glucose to simulate the process of ischemia/reperfusion in diabetes

  • We found that propofol postconditioning (P-PostC) attenuated above cell damages in a dose-dependent manner, being effective at concentrations ranging from 12.5 to 50 μmol/L while the most prominent effect was seen at 25 μmol/L

Read more

Summary

Introduction

Administration of propofol, an intravenous anesthetic with antioxidant property, immediately at the onset of post-ischemic reperfusion (propofol postconditioning, P-PostC) has been shown to confer cardioprotection against ischemia–reperfusion injury, while the underlying mechanism remains incompletely understood. Numerous studies have demonstrated that the reestablishment of coronary blood flow to ischemic heart diseases is the most effective treatment to rescue the damage of ischemic myocardium It is worth pointing out, that reperfusion of the ischemic myocardium could result in secondary damage, which is termed ischemic reperfusion (I/R) injury [1,2,3]. Oxidative stress acts as a signal to induce apoptosis, autophagy and necrosis during acute ischemia–reperfusion injury [8,9,10,11,12] Among all these manners of cell death, apoptosis appears to be an inseparable part in the elimination of invalid cells and it’s evidenced that the reduction in the expression of apoptosis-inducing Bax gene is beneficial to myocardial I/R injury [13]. It is debatable regarding whether or not autophagy is a beneficial effector mechanism of cell death, since the study has shown an advantageous effect on the pathological process of I/R injury by inhibiting autophagy via regulating Beclin, while the disadvantageous effect was observed via fading the expression of Atg gene [14, 15]

Methods
Results
Discussion
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