Basic Study
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Nov 26, 2023; 15(11): 1017-1034
Published online Nov 26, 2023. doi: 10.4252/wjsc.v15.i11.1017
Dissecting molecular mechanisms underlying ferroptosis in human umbilical cord mesenchymal stem cells: Role of cystathionine γ-lyase/hydrogen sulfide pathway
Bin Hu, Xiang-Xi Zhang, Tao Zhang, Wan-Cheng Yu
Bin Hu, Xiang-Xi Zhang, Tao Zhang, Wan-Cheng Yu, Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250062, Shandong Province, China
Co-first authors: Bin Hu and Xiang-Xi Zhang.
Author contributions: Hu B and Zhang XX contributed equally to this work; Yu WC conceived the research; Hu B and Yu WC participated in the design of the study, performed the statistical analysis, and helped to draft the manuscript; Hu B, Zhang XX, Zhang T, and Yu WC performed the experiments; and all authors participated in discussing and revising the manuscript, and approving the final manuscript.
Supported by the Natural Science Foundation of Shandong Province of China, No. ZR2021QH179 and ZR2020MH014.
Institutional animal care and use committee statement: Animal procedures were performed in compliance with the Institutional Animal Care and Use Committee of Shandong Provincial Hospital Affiliated to Shandong First Medical University (No. 2022-333).
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No additional unpublished data are available.
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: Wan-Cheng Yu, Doctor, MD, Postdoc, Surgeon, Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan 250062, Shandong Province, China. yuwancheng123@126.com
Received: September 2, 2023
Peer-review started: September 2, 2023
First decision: October 19, 2023
Revised: October 25, 2023
Accepted: November 17, 2023
Article in press: November 17, 2023
Published online: November 26, 2023
ARTICLE HIGHLIGHTS
Research background

Ferroptosis can decrease retention and engraftment after mesenchymal stem cell (MSC) delivery, which is considered a major challenge to the effectiveness of MSC-based therapy for pulmonary arterial hypertension (PAH). Interestingly, the cystathionine γ-lyase (CSE)/hydrogen sulfide (H2S) pathway may contribute to mediating ferroptosis.

Research motivation

We aimed to investigate the influence of the CSE/H2S pathway on ferroptosis in human umbilical cord MSCs (HUCMSCs).

Research objectives

We aimed to clarify whether the effect of HUCMSCs on vascular remodelling in mice with PAH is by CSE/H2S pathway-mediated ferroptosis. Furthermore, the effect of the CSE/H2S pathway on ferroptosis in HUCMSCs and the underlying mechanisms were investigated.

Research methods

Erastin and ferrostatin-1 (Fer-1) were used to induce and inhibit ferroptosis, respectively. HUCMSCs were transfected with a vector that overexpressed or inhibited CSE. A PAH mouse model was established using 4-wk-old male BALB/c nude mice under hypoxic conditions, and pulmonary pressure and vascular remodelling were measured. The survival of HUCMSCs after delivery was observed by in vivo bioluminescence imaging. Cell viability, iron accumulation, reactive oxygen species (ROS) production, cystine uptake, and lipid peroxidation in HUCMSCs were tested. Ferroptosis-related proteins and S-sulfhydrated Kelch-like ECH-associating protein 1 (Keap1) were detected by western blot analysis.

Research results

In vivo, CSE overexpression improved cell survival after erastin-treated HUCMSCs were delivered to mice with hypoxia-induced PAH. In vitro, CSE overexpression improved H2S production and ferroptosis-related indexes in erastin-treated HUCMSCs, such as cell viability, the iron level, ROS production, cystine uptake, lipid peroxidation, mitochondrial membrane density, and ferroptosis-related protein expression. In contrast, in vivo, CSE inhibition decreased cell survival after Fer-1-treated HUCMSC delivery and aggravated vascular remodelling in the PAH mice. In vitro, CSE inhibition decreased H2S levels and restored ferroptosis in Fer-1-treated HUCMSCs. Interestingly, we found that upregulation of the CSE/H2S pathway induced the S-sulfhydration of Keap1, which contributed to the inhibition of ferroptosis.

Research conclusions

Regulation of the CSE/H2S pathway in HUCMSCs contributes to the inhibition of ferroptosis and improves the effect of vascular remodelling suppression in hypoxia-induced PAH mice. Moreover, the protective effect of the CSE/H2S pathway on ferroptosis in HUCMSCs is mediated via S-sulfhydrated Keap1/nuclear factor erythroid 2-related factor 2 (Nrf2) signalling. The present study may provide a novel therapeutic avenue for improving the protective capacity of transplanted MSCs in PAH.

Research perspectives

Regulation of the CSE/H2S pathway in HUCMSCs contributes to the inhibition of ferroptosis and improves the ability of HUCMSCs to suppress vascular remodelling in mice with hypoxia-induced PAH. Moreover, the protective effect of the CSE/H2S pathway on ferroptosis in HUCMSCs is mediated via S-sulfhydrated Keap1/Nrf2 signalling. The present study may provide a novel therapeutic avenue for improving the protective capacity of transplanted MSCs in PAH.