figshare
Browse

miR-146a Suppresses SUMO1 Expression and Induces Cardiac Dysfunction in Maladaptive Hypertrophy

Published on by Jae Gyun Oh
ABSTRACT Rationale: Abnormal SUMOylation has emerged as a characteristic of heart failure (HF) pathology. Previously, we found reduced small ubiquitin-like modifier 1 (SUMO1) expression and sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) SUMOylation in human and animal HF models. SUMO1 gene delivery or small-molecule activation of SUMOylation restored SERCA2a SUMOylation and cardiac function in HF models. Despite the critical role of SUMO1 in HF, the regulatory mechanisms underlying SUMO1 expression are largely unknown. Objective: To examine miR-146a-mediated SUMO1 regulation and its consequent effects on cardiac morphology and function. Methods and Results: In this study, miR-146a was identified as a SUMO1-targeting microRNA in the heart. A strong correlation was observed between miR-146a and SUMO1 expression in failing mouse and human hearts. miR-146a was manipulated in cardiomyocytes through adeno-associated virus serotype 9 (AAV9)-mediated gene delivery, and cardiac morphology and function were analyzed by echocardiography and hemodynamics. Overexpression of miR-146a reduced SUMO1 expression, SERCA2a SUMOylation, and cardiac contractility in vitro and in vivo. The effects of miR-146a inhibition on HF pathophysiology were examined by transducing a tough decoy of miR-146a into mice subjected to transverse aortic constriction (TAC). miR-146a inhibition improved cardiac contractile function, and normalized SUMO1 expression. The regulatory mechanisms of miR-146a upregulation were elucidated by examining the major miR-146a-producing cell types and transfer mechanisms. Notably, trans-differentiation of fibroblasts triggered miR-146a overexpression and secretion through extracellular vesicles (EVs), and the EV-associated miR-146a transfer was identified as the causative mechanism of miR-146a upregulation in failing cardiomyocytes. Finally, EVs isolated from failing hearts were shown to contain high levels of miR-146a and exerted negative effects on the SUMO1/SERCA2a signaling axis and hence, cardiomyocyte contractility. Conclusions: Taken together, our results show that miR-146a is a novel regulator of the SUMOylation machinery in the heart, which can be targeted for therapeutic intervention. KEYWORDS Heart failure, SERCA2a, SUMO1, microRNA, extracellular vesicle, miR-146a

Cite items from this project

DataCite
3 Biotech
3D Printing in Medicine
3D Research
3D-Printed Materials and Systems
4OR
AAPG Bulletin
AAPS Open
AAPS PharmSciTech
Abhandlungen aus dem Mathematischen Seminar der Universität Hamburg
ABI Technik (German)
Academic Medicine
Academic Pediatrics
Academic Psychiatry
Academic Questions
Academy of Management Discoveries
Academy of Management Journal
Academy of Management Learning and Education
Academy of Management Perspectives
Academy of Management Proceedings
Academy of Management Review

cite all items

Funding

SOURCES OF FUNDING This work is supported by NIH R00 HL116645 (C. K.). RJH is funded by NIH R01 HL117505, HL 119046, HL129814, 128072, HL131404, R01HL135093, a P50 HL112324, and a Transatlantic Fondation Leducq grant. We would like to acknowledge the Gene Therapy Resource Program (GTRP) of the National Heart, Lung, and Blood Institute, National Institutes of Health for providing the gene vectors used in this study. J.G.O was funded by AHA 17POST33410877.

Share

email