Basic Study
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Feb 26, 2022; 14(2): 183-199
Published online Feb 26, 2022. doi: 10.4252/wjsc.v14.i2.183
Extracellular vesicles from hypoxia-preconditioned mesenchymal stem cells alleviates myocardial injury by targeting thioredoxin-interacting protein-mediated hypoxia-inducible factor-1α pathway
Cheng-Yu Mao, Tian-Tian Zhang, Dong-Jiu Li, En Zhou, Yu-Qi Fan, Qing He, Chang-Qian Wang, Jun-Feng Zhang
Cheng-Yu Mao, Tian-Tian Zhang, Dong-Jiu Li, En Zhou, Yu-Qi Fan, Qing He, Chang-Qian Wang, Jun-Feng Zhang, Department of Cardiology, Shanghai Ninth People’s Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200010, China
Author contributions: Mao CY, Zhang TT, Li DJ, and Zhou E contributed equally to this work, performed the experiments, and reviewed and edited the manuscript; Fan YQ and He Q wrote the paper; Zhang JF, and Wang CQ conceived of and funded the study; all authors read and approved the final manuscript.
Supported by National Natural Science Foundation of China, No. 81870264 and No. 81470546; the Shanghai Committee of Science and Technology, No. 18411950500; the Major Disease Joint Project of Shanghai Health System, No. 2014ZYJB0501; and Talent Cultivation Project of The Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. JC202005.
Institutional review board statement: The study was reviewed and approved by the Institutional Review Board of Shanghai Ninth People’s Hospital.
Institutional animal care and use committee statement: All animal procedures were approved by the Shanghai Ninth People’s Hospital Institutional Ethics Committee and conducted in accordance with the guidelines of the Directive 2010/63/EU of the European Parliament.
Conflict-of-interest statement: The authors declare that they have no competing interests regarding this study.
Data sharing statement: The data that support the findings of this study are available from the corresponding author upon reasonable request.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Jun-Feng Zhang, MD, PhD, Professor, Department of Cardiology, Shanghai Ninth People’s Hospital affiliated to Shanghai Jiao Tong University School of Medicine, No. 280 Mohe Road, Baoshan District, Shanghai 200010, China. junfengzhang9hos@163.com
Received: October 13, 2021
Peer-review started: October 13, 2021
First decision: November 8, 2021
Revised: November 29, 2021
Accepted: January 25, 2022
Article in press: January 25, 2022
Published online: February 26, 2022
Abstract
BACKGROUND

Extracellular vesicles (EVs) derived from hypoxia-preconditioned (HP) mesenchymal stem cells (MSCs) have better cardioprotective effects against myocardial infarction (MI) in the early stage than EVs isolated from normoxic (NC)-MSCs. However, the cardioprotective mechanisms of HP-EVs are not fully understood.

AIM

To explore the cardioprotective mechanism of EVs derived from HP MSCs.

METHODS

We evaluated the cardioprotective effects of HP-EVs or NC-EVs from mouse adipose-derived MSCs (ADSCs) following hypoxia in vitro or MI in vivo, in order to improve the survival of cardiomyocytes (CMs) and restore cardiac function. The degree of CM apoptosis in each group was assessed by the terminal deoxynucleotidyl transferase dUTP nick end-labeling and Annexin V/PI assays. MicroRNA (miRNA) sequencing was used to investigate the functional RNA diversity between HP-EVs and NC-EVs from mouse ADSCs. The molecular mechanism of EVs in mediating thioredoxin-interacting protein (TXNIP) was verified by the dual-luciferase reporter assay. Co-immunoprecipitation, western blotting, and immunofluorescence were performed to determine if TXNIP is involved in hypoxia-inducible factor-1 alpha (HIF-1α) ubiquitination and degradation via the chromosomal region maintenance-1 (CRM-1)-dependent nuclear transport pathway.

RESULTS

HP-EVs derived from MSCs reduced both infarct size (necrosis area) and apoptotic degree to a greater extent than NC-EVs from CMs subjected to hypoxia in vitro and mice with MI in vivo. Sequencing of EV-associated miRNAs showed the upregulation of 10 miRNAs predicted to bind TXNIP, an oxidative stress-associated protein. We showed miRNA224-5p, the most upregulated miRNA in HP-EVs, directly combined the 3’ untranslated region of TXNIP and demonstrated its critical protective role against hypoxia-mediated CM injury. Our results demonstrated that MI triggered TXNIP-mediated HIF-1α ubiquitination and degradation in the CRM-1-mediated nuclear transport pathway in CMs, which led to aggravated injury and hypoxia tolerance in CMs in the early stage of MI.

CONCLUSION

The anti-apoptotic effects of HP-EVs in alleviating MI and the hypoxic conditions of CMs until reperfusion therapy may partly result from EV miR-224-5p targeting TXNIP.

Keywords: Extracellular vesicles, Myocardial infarction, Mesenchymal stem cells, Hypoxia preconditioning, Thioredoxin-interacting protein, Hypoxia-inducible factor 1 alpha

Core Tip: Extracellular vesicles (EVs) from adipose-derived mesenchymal stem cells treated with hypoxia preconditioning improve tolerance toward myocardial infarction or hypoxic conditions and alleviate the degree of cardiomyocyte apoptosis until reperfusion therapy. The anti-apoptotic effects may result from EV miR-224-5p targeting thioredoxin-interacting protein (TXNIP) and subsequent TXNIP-mediated hypoxia-inducible factor-1 alpha ubiquitination and degradation via the chromosomal region maintenance-1-mediated nuclear transport pathway.