人脐带间充质干细胞铁下垂的解剖分子机制:半胱硫氨酸γ-裂解酶/硫化氢途径的作用。

IF 3.6 3区 医学 Q3 CELL & TISSUE ENGINEERING World journal of stem cells Pub Date : 2023-11-26 DOI:10.4252/wjsc.v15.i11.1017
Bin Hu, Xiang-Xi Zhang, Tao Zhang, Wan-Cheng Yu
{"title":"人脐带间充质干细胞铁下垂的解剖分子机制:半胱硫氨酸γ-裂解酶/硫化氢途径的作用。","authors":"Bin Hu, Xiang-Xi Zhang, Tao Zhang, Wan-Cheng Yu","doi":"10.4252/wjsc.v15.i11.1017","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis can induce low retention and engraftment after mesenchymal stem cell (MSC) delivery, which is considered a major challenge to the effectiveness of MSC-based pulmonary arterial hypertension (PAH) therapy. Interestingly, the cystathionine γ-lyase (CSE)/hydrogen sulfide (H<sub>2</sub>S) pathway may contribute to mediating ferroptosis. However, the influence of the CSE/H<sub>2</sub>S pathway on ferroptosis in human umbilical cord MSCs (HUCMSCs) remains unclear.</p><p><strong>Aim: </strong>To clarify whether the effect of HUCMSCs on vascular remodelling in PAH mice is affected by CSE/H<sub>2</sub>S pathway-mediated ferroptosis, and to investigate the functions of the CSE/H<sub>2</sub>S pathway in ferroptosis in HUCMSCs and the underlying mechanisms.</p><p><strong>Methods: </strong>Erastin and ferrostatin-1 (Fer-1) were used to induce and inhibit ferroptosis, respectively. HUCMSCs were transfected with a vector to overexpress or inhibit expression of 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 <i>in vivo</i> bioluminescence imaging. Cell viability, iron accumulation, reactive oxygen species 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.</p><p><strong>Results: </strong><i>In vivo</i>, CSE overexpression improved cell survival after erastin-treated HUCMSC delivery in mice with hypoxia-induced PAH. <i>In vitro</i>, CSE overexpression improved H<sub>2</sub>S production and ferroptosis-related indexes, such as cell viability, iron level, reactive oxygen species production, cystine uptake, lipid peroxidation, mitochondrial membrane density, and ferroptosis-related protein expression, in erastin-treated HUCMSCs. In contrast, <i>in vivo</i>, CSE inhibition decreased cell survival after Fer-1-treated HUCMSC delivery and aggravated vascular remodelling in PAH mice. <i>In vitro</i>, CSE inhibition decreased H<sub>2</sub>S levels and restored ferroptosis in Fer-1-treated HUCMSCs. Interestingly, upregulation of the CSE/H<sub>2</sub>S pathway induced Keap1 S-sulfhydration, which contributed to the inhibition of ferroptosis.</p><p><strong>Conclusion: </strong>Regulation of the CSE/H<sub>2</sub>S pathway in HUCMSCs contributes to the inhibition of ferroptosis and improves the suppressive effect on vascular remodelling in mice with hypoxia-induced PAH. Moreover, the protective effect of the CSE/H<sub>2</sub>S pathway against ferroptosis in HUCMSCs is mediated <i>via</i> S-sulfhydrated Keap1/nuclear factor erythroid 2-related factor 2 signalling. The present study may provide a novel therapeutic avenue for improving the protective capacity of transplanted MSCs in PAH.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":null,"pages":null},"PeriodicalIF":3.6000,"publicationDate":"2023-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10696191/pdf/","citationCount":"0","resultStr":"{\"title\":\"Dissecting molecular mechanisms underlying ferroptosis in human umbilical cord mesenchymal stem cells: Role of cystathionine γ-lyase/hydrogen sulfide pathway.\",\"authors\":\"Bin Hu, Xiang-Xi Zhang, Tao Zhang, Wan-Cheng Yu\",\"doi\":\"10.4252/wjsc.v15.i11.1017\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Ferroptosis can induce low retention and engraftment after mesenchymal stem cell (MSC) delivery, which is considered a major challenge to the effectiveness of MSC-based pulmonary arterial hypertension (PAH) therapy. Interestingly, the cystathionine γ-lyase (CSE)/hydrogen sulfide (H<sub>2</sub>S) pathway may contribute to mediating ferroptosis. However, the influence of the CSE/H<sub>2</sub>S pathway on ferroptosis in human umbilical cord MSCs (HUCMSCs) remains unclear.</p><p><strong>Aim: </strong>To clarify whether the effect of HUCMSCs on vascular remodelling in PAH mice is affected by CSE/H<sub>2</sub>S pathway-mediated ferroptosis, and to investigate the functions of the CSE/H<sub>2</sub>S pathway in ferroptosis in HUCMSCs and the underlying mechanisms.</p><p><strong>Methods: </strong>Erastin and ferrostatin-1 (Fer-1) were used to induce and inhibit ferroptosis, respectively. HUCMSCs were transfected with a vector to overexpress or inhibit expression of 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 <i>in vivo</i> bioluminescence imaging. Cell viability, iron accumulation, reactive oxygen species 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.</p><p><strong>Results: </strong><i>In vivo</i>, CSE overexpression improved cell survival after erastin-treated HUCMSC delivery in mice with hypoxia-induced PAH. <i>In vitro</i>, CSE overexpression improved H<sub>2</sub>S production and ferroptosis-related indexes, such as cell viability, iron level, reactive oxygen species production, cystine uptake, lipid peroxidation, mitochondrial membrane density, and ferroptosis-related protein expression, in erastin-treated HUCMSCs. In contrast, <i>in vivo</i>, CSE inhibition decreased cell survival after Fer-1-treated HUCMSC delivery and aggravated vascular remodelling in PAH mice. <i>In vitro</i>, CSE inhibition decreased H<sub>2</sub>S levels and restored ferroptosis in Fer-1-treated HUCMSCs. Interestingly, upregulation of the CSE/H<sub>2</sub>S pathway induced Keap1 S-sulfhydration, which contributed to the inhibition of ferroptosis.</p><p><strong>Conclusion: </strong>Regulation of the CSE/H<sub>2</sub>S pathway in HUCMSCs contributes to the inhibition of ferroptosis and improves the suppressive effect on vascular remodelling in mice with hypoxia-induced PAH. Moreover, the protective effect of the CSE/H<sub>2</sub>S pathway against ferroptosis in HUCMSCs is mediated <i>via</i> S-sulfhydrated Keap1/nuclear factor erythroid 2-related factor 2 signalling. The present study may provide a novel therapeutic avenue for improving the protective capacity of transplanted MSCs in PAH.</p>\",\"PeriodicalId\":23775,\"journal\":{\"name\":\"World journal of stem cells\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.6000,\"publicationDate\":\"2023-11-26\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10696191/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"World journal of stem cells\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.4252/wjsc.v15.i11.1017\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"CELL & TISSUE ENGINEERING\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"World journal of stem cells","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.4252/wjsc.v15.i11.1017","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"CELL & TISSUE ENGINEERING","Score":null,"Total":0}
引用次数: 0

摘要

背景:间充质干细胞(MSC)输注后,铁下沉可诱导低潴留和植入,这被认为是对基于间充质干细胞的肺动脉高压(PAH)治疗有效性的主要挑战。有趣的是,胱硫氨酸γ-裂解酶(CSE)/硫化氢(H2S)途径可能参与了铁下垂的介导。然而,CSE/H2S通路对人脐带间充质干细胞(HUCMSCs)铁下垂的影响尚不清楚。目的:阐明HUCMSCs对PAH小鼠血管重构的影响是否受CSE/H2S途径介导的铁凋亡的影响,并探讨CSE/H2S途径在HUCMSCs铁凋亡中的作用及其机制。方法:采用Erastin和fero -1分别诱导和抑制大鼠铁下垂。用载体转染HUCMSCs,过表达或抑制CSE的表达。采用4周龄雄性BALB/c裸鼠在缺氧条件下建立PAH小鼠模型,测量肺压和血管重构。采用体内生物发光成像技术观察HUCMSCs分娩后的存活情况。我们测试了HUCMSCs的细胞活力、铁积累、活性氧产生、胱氨酸摄取和脂质过氧化。western blot检测凋亡相关蛋白和s-巯基kelch样ech - associated protein 1 (Keap1)。结果:在体内,CSE过表达提高了缺氧诱导的PAH小鼠经erastin处理的HUCMSC递送后的细胞存活率。在体外,CSE过表达提高了erastin处理的HUCMSCs中H2S的产生和铁中毒相关指标,如细胞活力、铁水平、活性氧产生、胱氨酸摄取、脂质过氧化、线粒体膜密度和铁中毒相关蛋白的表达。相比之下,在体内,CSE抑制降低了fe -1处理的HUCMSC递送后的细胞存活率,并加剧了PAH小鼠的血管重构。在体外,CSE抑制降低了fe -1处理的HUCMSCs中H2S水平并恢复了铁下垂。有趣的是,CSE/H2S通路的上调诱导Keap1 s -巯基化,这有助于抑制铁下垂。结论:调节HUCMSCs的CSE/H2S通路有助于抑制缺氧诱导的PAH小鼠的铁凋亡,提高对血管重构的抑制作用。此外,CSE/H2S通路对HUCMSCs铁凋亡的保护作用是通过s -巯基化Keap1/核因子红系2相关因子2信号传导介导的。本研究可能为提高移植间充质干细胞对PAH的保护能力提供新的治疗途径。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Dissecting molecular mechanisms underlying ferroptosis in human umbilical cord mesenchymal stem cells: Role of cystathionine γ-lyase/hydrogen sulfide pathway.

Background: Ferroptosis can induce low retention and engraftment after mesenchymal stem cell (MSC) delivery, which is considered a major challenge to the effectiveness of MSC-based pulmonary arterial hypertension (PAH) therapy. Interestingly, the cystathionine γ-lyase (CSE)/hydrogen sulfide (H2S) pathway may contribute to mediating ferroptosis. However, the influence of the CSE/H2S pathway on ferroptosis in human umbilical cord MSCs (HUCMSCs) remains unclear.

Aim: To clarify whether the effect of HUCMSCs on vascular remodelling in PAH mice is affected by CSE/H2S pathway-mediated ferroptosis, and to investigate the functions of the CSE/H2S pathway in ferroptosis in HUCMSCs and the underlying mechanisms.

Methods: Erastin and ferrostatin-1 (Fer-1) were used to induce and inhibit ferroptosis, respectively. HUCMSCs were transfected with a vector to overexpress or inhibit expression of 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 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.

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

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

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
World journal of stem cells
World journal of stem cells Biochemistry, Genetics and Molecular Biology-Molecular Biology
CiteScore
7.80
自引率
4.90%
发文量
750
期刊介绍: The World Journal of Stem Cells (WJSC) is a leading academic journal devoted to reporting the latest, cutting-edge research progress and findings of basic research and clinical practice in the field of stem cells. It was launched on December 31, 2009 and is published monthly (12 issues annually) by BPG, the world''s leading professional clinical medical journal publishing company.
期刊最新文献
Innovative mesenchymal stem cell treatments for fatty liver disease. Mechanism of mesenchymal stem cells in liver regeneration: Insights and future directions. Mesenchymal stem cells in wound healing: A bibliometric analysis as a powerful research tool. Stem cell transplantation in cerebrovascular accidents: A global bibliometric analysis (2000-2023). Bone marrow mesenchymal stem cells in treatment of peripheral nerve injury.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1