Cargando…

Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics

Aging causes degenerative changes such as epigenetic changes and mitochondrial dysfunction in skeletal muscle. Exercise can upregulate muscle mitochondrial homeostasis and enhance antioxidant capacity and represents an effective treatment to prevent muscle aging. Epigenetic changes such as DNA methy...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Jialin, Wang, Zhe, Li, Can, Song, Yu, Wang, Yan, Bo, Hai, Zhang, Yong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9266156/
https://www.ncbi.nlm.nih.gov/pubmed/35805170
http://dx.doi.org/10.3390/cells11132086
_version_ 1784743394345484288
author Li, Jialin
Wang, Zhe
Li, Can
Song, Yu
Wang, Yan
Bo, Hai
Zhang, Yong
author_facet Li, Jialin
Wang, Zhe
Li, Can
Song, Yu
Wang, Yan
Bo, Hai
Zhang, Yong
author_sort Li, Jialin
collection PubMed
description Aging causes degenerative changes such as epigenetic changes and mitochondrial dysfunction in skeletal muscle. Exercise can upregulate muscle mitochondrial homeostasis and enhance antioxidant capacity and represents an effective treatment to prevent muscle aging. Epigenetic changes such as DNA methylation, histone posttranslational modifications, and microRNA expression are involved in the regulation of exercise-induced adaptive changes in muscle mitochondria. Reactive oxygen species (ROS) play an important role in signaling molecules in exercise-induced muscle mitochondrial health benefits, and strong evidence emphasizes that exercise-induced ROS can regulate gene expression via epigenetic mechanisms. The majority of mitochondrial proteins are imported into mitochondria from the cytosol, so mitochondrial homeostasis is regulated by nuclear epigenetic mechanisms. Exercise can reverse aging-induced changes in myokine expression by modulating epigenetic mechanisms. In this review, we provide an overview of the role of exercise-generated ROS in the regulation of mitochondrial homeostasis mediated by epigenetic mechanisms. In addition, the potential epigenetic mechanisms involved in exercise-induced myokine expression are reviewed.
format Online
Article
Text
id pubmed-9266156
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-92661562022-07-09 Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics Li, Jialin Wang, Zhe Li, Can Song, Yu Wang, Yan Bo, Hai Zhang, Yong Cells Review Aging causes degenerative changes such as epigenetic changes and mitochondrial dysfunction in skeletal muscle. Exercise can upregulate muscle mitochondrial homeostasis and enhance antioxidant capacity and represents an effective treatment to prevent muscle aging. Epigenetic changes such as DNA methylation, histone posttranslational modifications, and microRNA expression are involved in the regulation of exercise-induced adaptive changes in muscle mitochondria. Reactive oxygen species (ROS) play an important role in signaling molecules in exercise-induced muscle mitochondrial health benefits, and strong evidence emphasizes that exercise-induced ROS can regulate gene expression via epigenetic mechanisms. The majority of mitochondrial proteins are imported into mitochondria from the cytosol, so mitochondrial homeostasis is regulated by nuclear epigenetic mechanisms. Exercise can reverse aging-induced changes in myokine expression by modulating epigenetic mechanisms. In this review, we provide an overview of the role of exercise-generated ROS in the regulation of mitochondrial homeostasis mediated by epigenetic mechanisms. In addition, the potential epigenetic mechanisms involved in exercise-induced myokine expression are reviewed. MDPI 2022-06-30 /pmc/articles/PMC9266156/ /pubmed/35805170 http://dx.doi.org/10.3390/cells11132086 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Review
Li, Jialin
Wang, Zhe
Li, Can
Song, Yu
Wang, Yan
Bo, Hai
Zhang, Yong
Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title_full Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title_fullStr Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title_full_unstemmed Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title_short Impact of Exercise and Aging on Mitochondrial Homeostasis in Skeletal Muscle: Roles of ROS and Epigenetics
title_sort impact of exercise and aging on mitochondrial homeostasis in skeletal muscle: roles of ros and epigenetics
topic Review
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9266156/
https://www.ncbi.nlm.nih.gov/pubmed/35805170
http://dx.doi.org/10.3390/cells11132086
work_keys_str_mv AT lijialin impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT wangzhe impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT lican impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT songyu impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT wangyan impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT bohai impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics
AT zhangyong impactofexerciseandagingonmitochondrialhomeostasisinskeletalmusclerolesofrosandepigenetics