Cargando…

Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling

Proper mechanical stimulation can improve rotator cuff enthesis injury repair. However, the underlying mechanism of mechanical stimulation promoting injury repair is still unknown. In this study, we found that Prrx1(+) cell was essential for murine rotator cuff enthesis development identified by sin...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiao, Han, Zhang, Tao, Li, Changjun, Cao, Yong, Wang, Linfeng, Chen, Huabin, Li, Shengcan, Guan, Changbiao, Hu, Jianzhong, Chen, Di, Chen, Can, Lu, Hongbin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: eLife Sciences Publications, Ltd 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9094755/
https://www.ncbi.nlm.nih.gov/pubmed/35475783
http://dx.doi.org/10.7554/eLife.73614
_version_ 1784705610214801408
author Xiao, Han
Zhang, Tao
Li, Changjun
Cao, Yong
Wang, Linfeng
Chen, Huabin
Li, Shengcan
Guan, Changbiao
Hu, Jianzhong
Chen, Di
Chen, Can
Lu, Hongbin
author_facet Xiao, Han
Zhang, Tao
Li, Changjun
Cao, Yong
Wang, Linfeng
Chen, Huabin
Li, Shengcan
Guan, Changbiao
Hu, Jianzhong
Chen, Di
Chen, Can
Lu, Hongbin
author_sort Xiao, Han
collection PubMed
description Proper mechanical stimulation can improve rotator cuff enthesis injury repair. However, the underlying mechanism of mechanical stimulation promoting injury repair is still unknown. In this study, we found that Prrx1(+) cell was essential for murine rotator cuff enthesis development identified by single-cell RNA sequence and involved in the injury repair. Proper mechanical stimulation could promote the migration of Prrx1(+) cells to enhance enthesis injury repair. Meantime, TGF-β signaling and primary cilia played an essential role in mediating mechanical stimulation signaling transmission. Proper mechanical stimulation enhanced the release of active TGF-β1 to promote migration of Prrx1(+) cells. Inhibition of TGF-β signaling eliminated the stimulatory effect of mechanical stimulation on Prrx1(+) cell migration and enthesis injury repair. In addition, knockdown of Pallidin to inhibit TGF-βR2 translocation to the primary cilia or deletion of Ift88 in Prrx1(+) cells also restrained the mechanics-induced Prrx1(+) cells migration. These findings suggested that mechanical stimulation could increase the release of active TGF-β1 and enhance the mobilization of Prrx1(+) cells to promote enthesis injury repair via ciliary TGF-β signaling.
format Online
Article
Text
id pubmed-9094755
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher eLife Sciences Publications, Ltd
record_format MEDLINE/PubMed
spelling pubmed-90947552022-05-12 Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling Xiao, Han Zhang, Tao Li, Changjun Cao, Yong Wang, Linfeng Chen, Huabin Li, Shengcan Guan, Changbiao Hu, Jianzhong Chen, Di Chen, Can Lu, Hongbin eLife Stem Cells and Regenerative Medicine Proper mechanical stimulation can improve rotator cuff enthesis injury repair. However, the underlying mechanism of mechanical stimulation promoting injury repair is still unknown. In this study, we found that Prrx1(+) cell was essential for murine rotator cuff enthesis development identified by single-cell RNA sequence and involved in the injury repair. Proper mechanical stimulation could promote the migration of Prrx1(+) cells to enhance enthesis injury repair. Meantime, TGF-β signaling and primary cilia played an essential role in mediating mechanical stimulation signaling transmission. Proper mechanical stimulation enhanced the release of active TGF-β1 to promote migration of Prrx1(+) cells. Inhibition of TGF-β signaling eliminated the stimulatory effect of mechanical stimulation on Prrx1(+) cell migration and enthesis injury repair. In addition, knockdown of Pallidin to inhibit TGF-βR2 translocation to the primary cilia or deletion of Ift88 in Prrx1(+) cells also restrained the mechanics-induced Prrx1(+) cells migration. These findings suggested that mechanical stimulation could increase the release of active TGF-β1 and enhance the mobilization of Prrx1(+) cells to promote enthesis injury repair via ciliary TGF-β signaling. eLife Sciences Publications, Ltd 2022-04-27 /pmc/articles/PMC9094755/ /pubmed/35475783 http://dx.doi.org/10.7554/eLife.73614 Text en © 2022, Xiao et al https://creativecommons.org/licenses/by/4.0/This article is distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use and redistribution provided that the original author and source are credited.
spellingShingle Stem Cells and Regenerative Medicine
Xiao, Han
Zhang, Tao
Li, Changjun
Cao, Yong
Wang, Linfeng
Chen, Huabin
Li, Shengcan
Guan, Changbiao
Hu, Jianzhong
Chen, Di
Chen, Can
Lu, Hongbin
Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title_full Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title_fullStr Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title_full_unstemmed Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title_short Mechanical stimulation promotes enthesis injury repair by mobilizing Prrx1(+) cells via ciliary TGF-β signaling
title_sort mechanical stimulation promotes enthesis injury repair by mobilizing prrx1(+) cells via ciliary tgf-β signaling
topic Stem Cells and Regenerative Medicine
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9094755/
https://www.ncbi.nlm.nih.gov/pubmed/35475783
http://dx.doi.org/10.7554/eLife.73614
work_keys_str_mv AT xiaohan mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT zhangtao mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT lichangjun mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT caoyong mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT wanglinfeng mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT chenhuabin mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT lishengcan mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT guanchangbiao mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT hujianzhong mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT chendi mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT chencan mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling
AT luhongbin mechanicalstimulationpromotesenthesisinjuryrepairbymobilizingprrx1cellsviaciliarytgfbsignaling