Cargando…

miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging

Satellite cell‐dependent skeletal muscle regeneration declines during aging. Disruptions within the satellite cells and their niche, together with alterations in the myofibrillar environment, contribute to age‐related dysfunction and defective muscle regeneration. In this study, we demonstrated an a...

Descripción completa

Detalles Bibliográficos
Autores principales: Soriano‐Arroquia, Ana, Gostage, John, Xia, Qin, Bardell, David, McCormick, Rachel, McCloskey, Eugene, Bellantuono, Ilaria, Clegg, Peter, McDonagh, Brian, Goljanek‐Whysall, Katarzyna
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8520721/
https://www.ncbi.nlm.nih.gov/pubmed/34560818
http://dx.doi.org/10.1111/acel.13475
_version_ 1784584730233012224
author Soriano‐Arroquia, Ana
Gostage, John
Xia, Qin
Bardell, David
McCormick, Rachel
McCloskey, Eugene
Bellantuono, Ilaria
Clegg, Peter
McDonagh, Brian
Goljanek‐Whysall, Katarzyna
author_facet Soriano‐Arroquia, Ana
Gostage, John
Xia, Qin
Bardell, David
McCormick, Rachel
McCloskey, Eugene
Bellantuono, Ilaria
Clegg, Peter
McDonagh, Brian
Goljanek‐Whysall, Katarzyna
author_sort Soriano‐Arroquia, Ana
collection PubMed
description Satellite cell‐dependent skeletal muscle regeneration declines during aging. Disruptions within the satellite cells and their niche, together with alterations in the myofibrillar environment, contribute to age‐related dysfunction and defective muscle regeneration. In this study, we demonstrated an age‐related decline in satellite cell viability and myogenic potential and an increase in ROS and cellular senescence. We detected a transient upregulation of miR‐24 in regenerating muscle from adult mice and downregulation of miR‐24 during muscle regeneration in old mice. FACS‐sorted satellite cells were characterized by decreased levels of miR‐24 and a concomitant increase in expression of its target: Prdx6. Using GFP reporter constructs, we demonstrated that miR‐24 directly binds to its predicted site within Prdx6 mRNA. Subtle changes in Prdx6 levels following changes in miR‐24 expression indicate miR‐24 plays a role in fine‐tuning Prdx6 expression. Changes in miR‐24 and Prdx6 levels were associated with altered mitochondrial ROS generation, increase in the DNA damage marker: phosphorylated‐H2Ax and changes in viability, senescence, and myogenic potential of myogenic progenitors from mice and humans. The effects of miR‐24 were more pronounced in myogenic progenitors from old mice, suggesting a context‐dependent role of miR‐24 in these cells, with miR‐24 downregulation likely a part of a compensatory response to declining satellite cell function during aging. We propose that downregulation of miR‐24 and subsequent upregulation of Prdx6 in muscle of old mice following injury are an adaptive response to aging, to maintain satellite cell viability and myogenic potential through regulation of mitochondrial ROS and DNA damage pathways.
format Online
Article
Text
id pubmed-8520721
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-85207212021-10-25 miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging Soriano‐Arroquia, Ana Gostage, John Xia, Qin Bardell, David McCormick, Rachel McCloskey, Eugene Bellantuono, Ilaria Clegg, Peter McDonagh, Brian Goljanek‐Whysall, Katarzyna Aging Cell Original Papers Satellite cell‐dependent skeletal muscle regeneration declines during aging. Disruptions within the satellite cells and their niche, together with alterations in the myofibrillar environment, contribute to age‐related dysfunction and defective muscle regeneration. In this study, we demonstrated an age‐related decline in satellite cell viability and myogenic potential and an increase in ROS and cellular senescence. We detected a transient upregulation of miR‐24 in regenerating muscle from adult mice and downregulation of miR‐24 during muscle regeneration in old mice. FACS‐sorted satellite cells were characterized by decreased levels of miR‐24 and a concomitant increase in expression of its target: Prdx6. Using GFP reporter constructs, we demonstrated that miR‐24 directly binds to its predicted site within Prdx6 mRNA. Subtle changes in Prdx6 levels following changes in miR‐24 expression indicate miR‐24 plays a role in fine‐tuning Prdx6 expression. Changes in miR‐24 and Prdx6 levels were associated with altered mitochondrial ROS generation, increase in the DNA damage marker: phosphorylated‐H2Ax and changes in viability, senescence, and myogenic potential of myogenic progenitors from mice and humans. The effects of miR‐24 were more pronounced in myogenic progenitors from old mice, suggesting a context‐dependent role of miR‐24 in these cells, with miR‐24 downregulation likely a part of a compensatory response to declining satellite cell function during aging. We propose that downregulation of miR‐24 and subsequent upregulation of Prdx6 in muscle of old mice following injury are an adaptive response to aging, to maintain satellite cell viability and myogenic potential through regulation of mitochondrial ROS and DNA damage pathways. John Wiley and Sons Inc. 2021-09-24 2021-10 /pmc/articles/PMC8520721/ /pubmed/34560818 http://dx.doi.org/10.1111/acel.13475 Text en © 2021 The Authors. Aging Cell published by Anatomical Society and John Wiley & Sons Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Papers
Soriano‐Arroquia, Ana
Gostage, John
Xia, Qin
Bardell, David
McCormick, Rachel
McCloskey, Eugene
Bellantuono, Ilaria
Clegg, Peter
McDonagh, Brian
Goljanek‐Whysall, Katarzyna
miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title_full miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title_fullStr miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title_full_unstemmed miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title_short miR‐24 and its target gene Prdx6 regulate viability and senescence of myogenic progenitors during aging
title_sort mir‐24 and its target gene prdx6 regulate viability and senescence of myogenic progenitors during aging
topic Original Papers
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8520721/
https://www.ncbi.nlm.nih.gov/pubmed/34560818
http://dx.doi.org/10.1111/acel.13475
work_keys_str_mv AT sorianoarroquiaana mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT gostagejohn mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT xiaqin mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT bardelldavid mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT mccormickrachel mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT mccloskeyeugene mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT bellantuonoilaria mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT cleggpeter mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT mcdonaghbrian mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging
AT goljanekwhysallkatarzyna mir24anditstargetgeneprdx6regulateviabilityandsenescenceofmyogenicprogenitorsduringaging