Blocking the TRAIL-DR5 Pathway Reduces Cardiac Ischemia-Reperfusion Injury by Decreasing Neutrophil Infiltration and Neutrophil Extracellular Traps Formation.

IF 3.1 3区 医学 Q2 CARDIAC & CARDIOVASCULAR SYSTEMS
Xuance Wang, Ran Xie, Dan Zhao, Guiling Wang, Lijie Zhang, Wei Shi, Yanyan Chen, Tingting Mo, Yuxin Du, Xuefei Tian, Wanjun Wang, Run Cao, Yuanfang Ma, Yinxiang Wei, Yaohui Wang
{"title":"Blocking the TRAIL-DR5 Pathway Reduces Cardiac Ischemia-Reperfusion Injury by Decreasing Neutrophil Infiltration and Neutrophil Extracellular Traps Formation.","authors":"Xuance Wang, Ran Xie, Dan Zhao, Guiling Wang, Lijie Zhang, Wei Shi, Yanyan Chen, Tingting Mo, Yuxin Du, Xuefei Tian, Wanjun Wang, Run Cao, Yuanfang Ma, Yinxiang Wei, Yaohui Wang","doi":"10.1007/s10557-024-07591-z","DOIUrl":null,"url":null,"abstract":"<p><strong>Purpose: </strong>Acute myocardial infarction (AMI) is a leading cause of mortality. Neutrophils penetrate injured heart tissue during AMI or ischemia-reperfusion (I/R) injury and produce inflammatory factors, chemokines, and extracellular traps that exacerbate heart injury. Inhibition of the TRAIL-DR5 pathway has been demonstrated to alleviate cardiac ischemia-reperfusion injury in a leukocyte-dependent manner. However, it remains unknown whether TRAIL-DR5 signaling is involved in regulating neutrophil extracellular traps (NETs) release.</p><p><strong>Methods: </strong>This study used various models to examine the effects of activating the TRAIL-DR5 pathway with soluble mouse TRAIL protein and inhibiting the TRAIL-DR5 signaling pathway using DR5 knockout mice or mDR5-Fc fusion protein on NETs formation and cardiac injury. The models used included a co-culture model involving bone marrow-derived neutrophils and primary cardiomyocytes and a model of myocardial I/R in mice.</p><p><strong>Results: </strong>NETs formation is suppressed by TRAIL-DR5 signaling pathway inhibition, which can lessen cardiac I/R injury. This intervention reduces the release of adhesion molecules and chemokines, resulting in decreased neutrophil infiltration and inhibiting NETs production by downregulating PAD4 in neutrophils.</p><p><strong>Conclusion: </strong>This work clarifies how the TRAIL-DR5 signaling pathway regulates the neutrophil response during myocardial I/R damage, thereby providing a scientific basis for therapeutic intervention targeting the TRAIL-DR5 signaling pathway in myocardial infarction.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":null,"pages":null},"PeriodicalIF":3.1000,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cardiovascular Drugs and Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s10557-024-07591-z","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CARDIAC & CARDIOVASCULAR SYSTEMS","Score":null,"Total":0}
引用次数: 0

Abstract

Purpose: Acute myocardial infarction (AMI) is a leading cause of mortality. Neutrophils penetrate injured heart tissue during AMI or ischemia-reperfusion (I/R) injury and produce inflammatory factors, chemokines, and extracellular traps that exacerbate heart injury. Inhibition of the TRAIL-DR5 pathway has been demonstrated to alleviate cardiac ischemia-reperfusion injury in a leukocyte-dependent manner. However, it remains unknown whether TRAIL-DR5 signaling is involved in regulating neutrophil extracellular traps (NETs) release.

Methods: This study used various models to examine the effects of activating the TRAIL-DR5 pathway with soluble mouse TRAIL protein and inhibiting the TRAIL-DR5 signaling pathway using DR5 knockout mice or mDR5-Fc fusion protein on NETs formation and cardiac injury. The models used included a co-culture model involving bone marrow-derived neutrophils and primary cardiomyocytes and a model of myocardial I/R in mice.

Results: NETs formation is suppressed by TRAIL-DR5 signaling pathway inhibition, which can lessen cardiac I/R injury. This intervention reduces the release of adhesion molecules and chemokines, resulting in decreased neutrophil infiltration and inhibiting NETs production by downregulating PAD4 in neutrophils.

Conclusion: This work clarifies how the TRAIL-DR5 signaling pathway regulates the neutrophil response during myocardial I/R damage, thereby providing a scientific basis for therapeutic intervention targeting the TRAIL-DR5 signaling pathway in myocardial infarction.

Abstract Image

阻断 TRAIL-DR5 通路可通过减少中性粒细胞浸润和中性粒细胞胞外陷阱的形成减轻心脏缺血再灌注损伤
目的:急性心肌梗死(AMI)是导致死亡的主要原因。在急性心肌梗死或缺血再灌注(I/R)损伤期间,中性粒细胞会穿透受伤的心脏组织,并产生炎症因子、趋化因子和细胞外捕获物,从而加剧心脏损伤。抑制 TRAIL-DR5 通路已被证实能以白细胞依赖的方式减轻心脏缺血再灌注损伤。然而,TRAIL-DR5 信号是否参与调节中性粒细胞胞外捕获物(NETs)的释放仍是未知数:本研究使用多种模型来研究用可溶性小鼠 TRAIL 蛋白激活 TRAIL-DR5 通路和用 DR5 基因敲除小鼠或 mDR5-Fc 融合蛋白抑制 TRAIL-DR5 信号通路对 NETs 形成和心脏损伤的影响。使用的模型包括骨髓中性粒细胞和原代心肌细胞共培养模型和小鼠心肌I/R模型:结果:抑制 TRAIL-DR5 信号通路可抑制 NETs 的形成,从而减轻心脏 I/R 损伤。这一干预措施可减少粘附分子和趋化因子的释放,从而减少中性粒细胞的浸润,并通过下调中性粒细胞中的 PAD4 抑制 NETs 的生成:这项研究阐明了 TRAIL-DR5 信号通路如何调控心肌 I/R 损伤过程中的中性粒细胞反应,从而为针对 TRAIL-DR5 信号通路的心肌梗死治疗干预提供了科学依据。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Cardiovascular Drugs and Therapy
Cardiovascular Drugs and Therapy 医学-心血管系统
CiteScore
8.30
自引率
0.00%
发文量
110
审稿时长
4.5 months
期刊介绍: Designed to objectively cover the process of bench to bedside development of cardiovascular drug, device and cell therapy, and to bring you the information you need most in a timely and useful format, Cardiovascular Drugs and Therapy takes a fresh and energetic look at advances in this dynamic field. Homing in on the most exciting work being done on new therapeutic agents, Cardiovascular Drugs and Therapy focusses on developments in atherosclerosis, hyperlipidemia, diabetes, ischemic syndromes and arrhythmias. The Journal is an authoritative source of current and relevant information that is indispensable for basic and clinical investigators aiming for novel, breakthrough research as well as for cardiologists seeking to best serve their patients. Providing you with a single, concise reference tool acknowledged to be among the finest in the world, Cardiovascular Drugs and Therapy is listed in Web of Science and PubMed/Medline among other abstracting and indexing services. The regular articles and frequent special topical issues equip you with an up-to-date source defined by the need for accurate information on an ever-evolving field. Cardiovascular Drugs and Therapy is a careful and accurate guide through the maze of new products and therapies which furnishes you with the details on cardiovascular pharmacology that you will refer to time and time again.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信