Progranulin deficiency exacerbates cardiac remodeling after myocardial infarction

IF 2.5 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY FASEB bioAdvances Pub Date : 2023-09-20 DOI:10.1096/fba.2023-00084
Takahiro Sasaki, Yoshiki Kuse, Shinsuke Nakamura, Masamitsu Shimazawa, Hideaki Hara
{"title":"Progranulin deficiency exacerbates cardiac remodeling after myocardial infarction","authors":"Takahiro Sasaki,&nbsp;Yoshiki Kuse,&nbsp;Shinsuke Nakamura,&nbsp;Masamitsu Shimazawa,&nbsp;Hideaki Hara","doi":"10.1096/fba.2023-00084","DOIUrl":null,"url":null,"abstract":"<p>Myocardial infarction (MI) is a lethal disease that causes irreversible cardiomyocyte death and subsequent cardiovascular remodeling. We have previously shown that the administration of recombinant progranulin (PGRN) protects against myocardial ischemia and reperfusion injury. However, the post-MI role of PGRN remains unclear. In the present study, we investigated the effects of PGRN deficiency on cardiac remodeling after MI. Wild-type and PGRN-knockout mice were subjected to MI by ligation of the left coronary artery for histological, electrophysiological, and protein expression analysis. Cardiac macrophage subpopulations were analyzed by flow cytometry. Bone marrow-derived macrophages (BMDMs) were acquired and treated with LPS + IFN-γ and IL-4 to evaluate mRNA levels and phagocytic ability. PGRN expression was gradually increased in the whole heart at 1, 3, and 7 days after MI. Macrophages abundantly expressed PGRN at the border areas at 3 days post-MI. PGRN-knockout mice showed higher mortality, increased LV fibrosis, and severe arrhythmia following MI. PGRN deficiency increased the levels of CD206 and MerTK expression and macrophage infiltration in the infarcted myocardium, which was attributed to a larger subpopulation of cardiac CCR2<sup>+</sup> Ly6C<sup>low</sup> CD11b<sup>+</sup> macrophages. PGRN-deficient BMDMs exhibited higher TGF-β, IL-4R, and lower IL-1β, IL-10 and increased acute phagocytosis following stimulation of LPS and IFN-γ. PGRN deficiency reduced survival and increased cardiac fibrosis following MI with the induction of abnormal subpopulation of cardiac macrophages early after MI, thereby providing insight into the relationship between properly initiating cardiac repair and macrophage polarization after MI.</p>","PeriodicalId":12093,"journal":{"name":"FASEB bioAdvances","volume":null,"pages":null},"PeriodicalIF":2.5000,"publicationDate":"2023-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/69/c5/FBA2-5-395.PMC10551273.pdf","citationCount":"1","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"FASEB bioAdvances","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1096/fba.2023-00084","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 1

Abstract

Myocardial infarction (MI) is a lethal disease that causes irreversible cardiomyocyte death and subsequent cardiovascular remodeling. We have previously shown that the administration of recombinant progranulin (PGRN) protects against myocardial ischemia and reperfusion injury. However, the post-MI role of PGRN remains unclear. In the present study, we investigated the effects of PGRN deficiency on cardiac remodeling after MI. Wild-type and PGRN-knockout mice were subjected to MI by ligation of the left coronary artery for histological, electrophysiological, and protein expression analysis. Cardiac macrophage subpopulations were analyzed by flow cytometry. Bone marrow-derived macrophages (BMDMs) were acquired and treated with LPS + IFN-γ and IL-4 to evaluate mRNA levels and phagocytic ability. PGRN expression was gradually increased in the whole heart at 1, 3, and 7 days after MI. Macrophages abundantly expressed PGRN at the border areas at 3 days post-MI. PGRN-knockout mice showed higher mortality, increased LV fibrosis, and severe arrhythmia following MI. PGRN deficiency increased the levels of CD206 and MerTK expression and macrophage infiltration in the infarcted myocardium, which was attributed to a larger subpopulation of cardiac CCR2+ Ly6Clow CD11b+ macrophages. PGRN-deficient BMDMs exhibited higher TGF-β, IL-4R, and lower IL-1β, IL-10 and increased acute phagocytosis following stimulation of LPS and IFN-γ. PGRN deficiency reduced survival and increased cardiac fibrosis following MI with the induction of abnormal subpopulation of cardiac macrophages early after MI, thereby providing insight into the relationship between properly initiating cardiac repair and macrophage polarization after MI.

Abstract Image

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
孕激素缺乏会加剧心肌梗死后的心脏重塑。
心肌梗死(MI)是一种致命的疾病,可导致不可逆的心肌细胞死亡和随后的心血管重塑。我们之前已经证明,给予重组前颗粒蛋白(PGRN)可以预防心肌缺血和再灌注损伤。然而,PGRN在心肌梗死后的作用尚不清楚。在本研究中,我们研究了PGRN缺乏对MI后心脏重塑的影响。通过结扎左冠状动脉对野生型和PGRN敲除小鼠进行MI,进行组织学、电生理学和蛋白质表达分析。通过流式细胞术分析心脏巨噬细胞亚群。获得骨髓源性巨噬细胞(BMDMs)并用LPS处理 + γ和IL-4来评估mRNA水平和吞噬能力。PGRN在1、3和7时在整个心脏中的表达逐渐增加 MI后几天。巨噬细胞在3时在边境地区大量表达PGRN MI后的天数。PGRN敲除小鼠在MI后表现出更高的死亡率、左心室纤维化增加和严重心律失常。PGRN缺乏增加了梗死心肌中CD206和MerTK的表达水平和巨噬细胞浸润,这归因于心脏CCR2+Ly6Clow CD11b+巨噬细胞的更大亚群。PGRN缺陷型BMDM在LPS和IFN-γ刺激后表现出较高的TGF-β、IL-4R和较低的IL-1β、IL-10,并增加了急性吞噬作用。PGRN缺乏降低了MI后的存活率并增加了心肌纤维化,在MI后早期诱导了异常的心脏巨噬细胞亚群,从而深入了解了MI后正确启动心脏修复和巨噬细胞极化之间的关系。
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 去求助
来源期刊
FASEB bioAdvances
FASEB bioAdvances Multiple-
CiteScore
5.40
自引率
3.70%
发文量
56
审稿时长
10 weeks
期刊最新文献
Issue Information In vivo optical assessment of cerebral and skeletal muscle microvascular response to phenylephrine Human inborn errors of long-chain fatty acid oxidation show impaired inflammatory responses to TLR4-ligand LPS Activation of nociception-sensitive ionotropic glutamate receptor-expressing rostroventrolateral medulla neurons by stimulation of cardiac afferents in rats Osteocalcin binds to a GPRC6A Venus fly trap allosteric site to positively modulate GPRC6A signaling
×
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