Mitochondria-encoded peptide MOTS-c participates in plasma membrane repair by facilitating the translocation of TRIM72 to membrane

IF 12.4 1区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL Theranostics Pub Date : 2024-08-19 DOI:10.7150/thno.100321
Hong Jia, Lyu-Chen Zhou, Yong-Feng Chen, Wei Zhang, Wei Qi, Peng Wang, Xiao Huang, Jian-Wei Guo, Wai-Fang Hou, Ran-Ran Zhang, Jing-Jun Zhou, Da-Wei Zhang
{"title":"Mitochondria-encoded peptide MOTS-c participates in plasma membrane repair by facilitating the translocation of TRIM72 to membrane","authors":"Hong Jia, Lyu-Chen Zhou, Yong-Feng Chen, Wei Zhang, Wei Qi, Peng Wang, Xiao Huang, Jian-Wei Guo, Wai-Fang Hou, Ran-Ran Zhang, Jing-Jun Zhou, Da-Wei Zhang","doi":"10.7150/thno.100321","DOIUrl":null,"url":null,"abstract":"<b>Rationale:</b> An impairment of plasma membrane repair has been implicated in various diseases such as muscular dystrophy and ischemia/reperfusion injury. MOTS-c, a short peptide encoded by mitochondria, has been shown to pass through the plasma membrane into the bloodstream. This study determined whether this biological behavior was involved in membrane repair and its underlying mechanism./n<b>Methods and Results:</b> In human participants, the level of MOTS-c was positively correlated with the abundance of mitochondria, and the membrane repair molecule TRIM72. In contrast to high-intensity eccentric exercise, moderate-intensity exercise improved sarcolemma integrity and physical performance, accompanied by an increase of mitochondria beneath the damaged sarcolemma and secretion of MOTS-c. Furthermore, moderate-intensity exercise increased the interaction between MOTS-c and TRIM72, and MOTS-c facilitated the trafficking of TRIM72 to the sarcolemma. <i>In vitro</i> studies demonstrated that MOTS-c attenuated membrane damage induced by hypotonic solution, which could be blocked by siRNA-TRIM72, but not AMPK inhibitor. Co-immunoprecipitation study showed that MOTS-c interacted with TRIM72 C-terminus, but not N-terminus. The dynamic membrane repair assay revealed that MOTS-c boosted the trafficking of TRIM72 to the injured membrane. However, MOTS-c itself had negligible effects on membrane repair, which was recapitulated in TRIM72<sup>-/-</sup> mice. Unexpectedly, MOTS-c still increased the fusion of vesicles with the membrane in TRIM72<sup>-/-</sup> mice, and dot blot analysis revealed an interaction between MOTS-c and phosphatidylinositol (4,5) bisphosphate [PtdIns (4,5) P<sub>2</sub>]. Finally, MOTS-c blunted ischemia/reperfusion-induced membrane disruption, and preserved heart function./n<b>Conclusions:</b> MOTS-c/TRIM72-mediated membrane integrity improvement participates in mitochondria-triggered membrane repair. An interaction between MOTS-c and plasma lipid contributes to the fusion of vesicles with membrane. Our data provide a novel therapeutic strategy for rescuing organ function by facilitating membrane repair with MOTS-c.","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":null,"pages":null},"PeriodicalIF":12.4000,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Theranostics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/thno.100321","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Rationale: An impairment of plasma membrane repair has been implicated in various diseases such as muscular dystrophy and ischemia/reperfusion injury. MOTS-c, a short peptide encoded by mitochondria, has been shown to pass through the plasma membrane into the bloodstream. This study determined whether this biological behavior was involved in membrane repair and its underlying mechanism./nMethods and Results: In human participants, the level of MOTS-c was positively correlated with the abundance of mitochondria, and the membrane repair molecule TRIM72. In contrast to high-intensity eccentric exercise, moderate-intensity exercise improved sarcolemma integrity and physical performance, accompanied by an increase of mitochondria beneath the damaged sarcolemma and secretion of MOTS-c. Furthermore, moderate-intensity exercise increased the interaction between MOTS-c and TRIM72, and MOTS-c facilitated the trafficking of TRIM72 to the sarcolemma. In vitro studies demonstrated that MOTS-c attenuated membrane damage induced by hypotonic solution, which could be blocked by siRNA-TRIM72, but not AMPK inhibitor. Co-immunoprecipitation study showed that MOTS-c interacted with TRIM72 C-terminus, but not N-terminus. The dynamic membrane repair assay revealed that MOTS-c boosted the trafficking of TRIM72 to the injured membrane. However, MOTS-c itself had negligible effects on membrane repair, which was recapitulated in TRIM72-/- mice. Unexpectedly, MOTS-c still increased the fusion of vesicles with the membrane in TRIM72-/- mice, and dot blot analysis revealed an interaction between MOTS-c and phosphatidylinositol (4,5) bisphosphate [PtdIns (4,5) P2]. Finally, MOTS-c blunted ischemia/reperfusion-induced membrane disruption, and preserved heart function./nConclusions: MOTS-c/TRIM72-mediated membrane integrity improvement participates in mitochondria-triggered membrane repair. An interaction between MOTS-c and plasma lipid contributes to the fusion of vesicles with membrane. Our data provide a novel therapeutic strategy for rescuing organ function by facilitating membrane repair with MOTS-c.
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
线粒体编码的肽 MOTS-c 通过促进 TRIM72 向膜的转位参与质膜修复
理由肌肉萎缩症和缺血/再灌注损伤等多种疾病都与质膜修复功能受损有关。线粒体编码的短肽 MOTS-c 已被证明能穿过质膜进入血液。本研究确定了这种生物行为是否参与了膜修复及其内在机制:在人类参与者中,MOTS-c 的水平与线粒体的丰度和膜修复分子 TRIM72 呈正相关。与高强度偏心运动相比,中等强度运动能改善肌浆完整性和体能,同时增加受损肌浆下的线粒体和MOTS-c的分泌。此外,中等强度运动增加了MOTS-c和TRIM72之间的相互作用,MOTS-c促进了TRIM72向肌浆的迁移。体外研究表明,MOTS-c可减轻低渗溶液引起的膜损伤,这种损伤可被siRNA-TRIM72阻断,但不能被AMPK抑制剂阻断。共免疫沉淀研究表明,MOTS-c与TRIM72的C端有相互作用,但与N端没有相互作用。动态膜修复试验显示,MOTS-c能促进TRIM72向损伤膜的迁移。然而,MOTS-c本身对膜修复的影响微乎其微,这在TRIM72-/-小鼠中得到了再现。意想不到的是,在 TRIM72-/- 小鼠体内,MOTS-c 仍然增加了囊泡与膜的融合,点印迹分析显示 MOTS-c 与磷脂酰肌醇(4,5)二磷酸[PtdIns (4,5) P2]之间存在相互作用。最后,MOTS-c 可减轻缺血/再灌注引起的膜破坏,并保护心脏功能:MOTS-c/TRIM72 介导的膜完整性改善参与了线粒体触发的膜修复。MOTS-c 与血浆脂质之间的相互作用有助于囊泡与膜的融合。我们的数据为通过 MOTS-c 促进膜修复来挽救器官功能提供了一种新的治疗策略。
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
Theranostics
Theranostics MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
25.40
自引率
1.60%
发文量
433
审稿时长
1 months
期刊介绍: Theranostics serves as a pivotal platform for the exchange of clinical and scientific insights within the diagnostic and therapeutic molecular and nanomedicine community, along with allied professions engaged in integrating molecular imaging and therapy. As a multidisciplinary journal, Theranostics showcases innovative research articles spanning fields such as in vitro diagnostics and prognostics, in vivo molecular imaging, molecular therapeutics, image-guided therapy, biosensor technology, nanobiosensors, bioelectronics, system biology, translational medicine, point-of-care applications, and personalized medicine. Encouraging a broad spectrum of biomedical research with potential theranostic applications, the journal rigorously peer-reviews primary research, alongside publishing reviews, news, and commentary that aim to bridge the gap between the laboratory, clinic, and biotechnology industries.
期刊最新文献
A hierarchically acidity-unlocking nanoSTING stimulant enables cascaded STING activation for potent innate and adaptive antitumor immunity. Hypoxanthine is a metabolic biomarker for inducing GSDME-dependent pyroptosis of endothelial cells during ischemic stroke. Multifunctionally disordered TiO2 nanoneedles prevent periprosthetic infection and enhance osteointegration by killing bacteria and modulating the osteoimmune microenvironment. Nicotinamide mononucleotide enhances fracture healing by promoting skeletal stem cell proliferation. p-STAT3-elevated E3 ubiquitin ligase DTX4 confers the stability of HBV cccDNA by ubiquitinating APOBEC3B in liver.
×
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