Abstract

Background/Aims Circular RNAs (circRNAs) are a class of endogenous noncoding RNAs that regulate gene expression in eukaryotes. Recently, exosomes from cardiomyocytes (CMs) have been found to facilitate cell proliferation and survival by transporting various bioactive molecules, including circRNA. However, the functions of exosomal circRNAs are not clear. The present research is aimed at determining whether circHIPK3 released from hypoxia-pretreated CMs is transferred into cardiac microvascular endothelial cells (CMVECs) by exosomes and becomes functionally active in the CMVECs under oxidative stress conditions. Methods Quantitative polymerase chain reactions were conducted to detect the expression pattern of circHIPK3 in CMVECs under oxidative stress. Annexin V-FITC/propidium iodide (PI) staining assays, TUNEL assays, ROS assays, and Western blot analysis were conducted to detect the role of exosomal circHIPK3 in CMVEC function in vitro. Luciferase activity assays and RNA immunoprecipitation studies were conducted in vitro to reveal the mechanism of circHIPK3-mediated CMVEC function. Results circHIPK3 expression was significantly upregulated in hypoxic exosomes (HPC-exos) compared with normoxic exosomes (Nor-exos). Moreover, HPC-exos induced stronger antioxidant effects than Nor-exos. The silencing or overexpression of circHIPK3 changed CMVEC survival under oxidative conditions in vitro. Furthermore, circHIPK3 silencing in HPC-exos abrogated the protective effects of HPC-exos in CMVECs, as shown by increased levels of apoptosis, ROS, MDA, and proapoptotic proteins. circHIPK3 acted as an endogenous miR-29a sponge to sequester and inhibit miR-29a activity, which led to increased IGF-1 expression. The ectopic expression of miR-29a mimicked the effect of circHIPK3 silencing in CMVECs in vitro. Conclusions circHIPK3 in HPC-exos plays a role in CMVECs under oxidative conditions through miR-29a-mediated IGF-1 expression, leading to a decrease in oxidative stress-induced CMVECs dysfunction. These data suggest that the exosomal circRNA in CMs is a potential target to control CMVECs dysfunction under oxidative conditions.

Highlights

  • Microcirculatory dysfunction is an important etiological component of ischemia-reperfusion injury [1]

  • We investigated the role of exosomal circHIPK3 released from CMs pretreated with hypoxia in maintaining cardiac microvascular endothelial cell function

  • Oxidative stress-related complications in Cardiac microvascular endothelial cells (CMVECs) after myocardial ischemia are the major cause of heart dysfunction [42]

Read more

Summary

Introduction

Microcirculatory dysfunction is an important etiological component of ischemia-reperfusion injury [1]. Oxidative stress caused by a surge in the generation of reactive oxygen species (ROS) during reoxygenation can disrupt microvascular integrity [2], decreasing the oxygen and nutrients supplied to cardiac cells. Cardiac microvascular endothelial cells (CMVECs) play an obligatory role in regulating and maintaining cardiac function by forming connections and constituting the continuous endothelium between the circulation and cardiomyocytes (CMs) [3, 4]. The response of CMVECs to ROS impacts heart function via changes in endothelial barrier function that subsequently disrupt tissue blood flow. To ensure sufficient blood supply to deprived areas [5], it is important to explore powerful strategies to protect CMVECs from oxidative stress. The maintenance of microvascular anatomic and functional integrity

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