The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair

Abstract Background During aging, perturbation of muscle progenitor cell (MPC) constituents leads to progressive loss of muscle mass and accumulation of adipose and fibrotic tissue. Mesenchymal stem cells (MSCs) give rise to adipocytes and fibroblasts that accumulate in injured and pathological skel...

Full description

Bibliographic Details
Main Authors: Aiping Lu, Chieh Tseng, Ping Guo, Zhanguo Gao, Kaitlyn E. Whitney, Mikhail G. Kolonin, Johnny Huard
Format: Article
Language:English
Published: BMC 2022-08-01
Series:Stem Cell Research & Therapy
Subjects:
Online Access:https://doi.org/10.1186/s13287-022-03072-y
_version_ 1811315272534982656
author Aiping Lu
Chieh Tseng
Ping Guo
Zhanguo Gao
Kaitlyn E. Whitney
Mikhail G. Kolonin
Johnny Huard
author_facet Aiping Lu
Chieh Tseng
Ping Guo
Zhanguo Gao
Kaitlyn E. Whitney
Mikhail G. Kolonin
Johnny Huard
author_sort Aiping Lu
collection DOAJ
description Abstract Background During aging, perturbation of muscle progenitor cell (MPC) constituents leads to progressive loss of muscle mass and accumulation of adipose and fibrotic tissue. Mesenchymal stem cells (MSCs) give rise to adipocytes and fibroblasts that accumulate in injured and pathological skeletal muscle through constitutive activation of platelet-derived growth factor receptors (PDGFRs). Although the role of the PDGFRα has been widely explored, there is a paucity of evidence demonstrating the role of PDGFRβ in aged skeletal muscle. Methods In this study, we investigated the role of PDGFRβ lineage cells in skeletal muscle during aging by using Cre/loxP lineage tracing technology. The PDGFR-Cre mice were crossed with global double-fluorescent Cre reporter mice (mTmG) that indelibly marks PDGFRβ lineage cells. Those cells were analyzed and compared at different ages in the skeletal muscle of the mice. Results Our results demonstrated that PDGFRβ lineage cells isolated from the muscles of young mice are MPC-like cells that exhibited satellite cell morphology, expressed Pax7, and undergo myogenic differentiation producing myosin heavy chain expressing myotubes. Conversely, the PDGFRβ lineage cells isolated from muscles of old mice displayed MSC morphology with a reduced myogenic differentiation potential while expressing adipogenic and fibrotic differentiation markers. PDGFRβ lineage cells also gave rise to newly regenerated muscle fibers in young mice after muscle injury, but their muscle regenerative process is reduced in old mice. Conclusions Our data suggest that PDGFRβ lineage cells function as MPCs in young mice, while the same PDGFRβ lineage cells from old mice undergo a fate switch participating in adipose and fibrotic tissue infiltration in aged muscle. The inhibition of fate-switching in PDGFRβ lineage cells may represent a potential approach to prevent fibrosis and fatty infiltration in skeletal muscle during the aging process.
first_indexed 2024-04-13T11:27:19Z
format Article
id doaj.art-3b1b7a739f9844119cb513bb085ef0cf
institution Directory Open Access Journal
issn 1757-6512
language English
last_indexed 2024-04-13T11:27:19Z
publishDate 2022-08-01
publisher BMC
record_format Article
series Stem Cell Research & Therapy
spelling doaj.art-3b1b7a739f9844119cb513bb085ef0cf2022-12-22T02:48:39ZengBMCStem Cell Research & Therapy1757-65122022-08-0113111210.1186/s13287-022-03072-yThe role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repairAiping Lu0Chieh Tseng1Ping Guo2Zhanguo Gao3Kaitlyn E. Whitney4Mikhail G. Kolonin5Johnny Huard6Center for Regenerative and Personalized Medicine, Steadman Philippon Research InstituteM.D. Anderson Cancer Center, The University of Texas Health Science CenterCenter for Regenerative and Personalized Medicine, Steadman Philippon Research InstituteInstitute of Molecular Medicine, The University of Texas Health Science CenterCenter for Regenerative and Personalized Medicine, Steadman Philippon Research InstituteInstitute of Molecular Medicine, The University of Texas Health Science CenterCenter for Regenerative and Personalized Medicine, Steadman Philippon Research InstituteAbstract Background During aging, perturbation of muscle progenitor cell (MPC) constituents leads to progressive loss of muscle mass and accumulation of adipose and fibrotic tissue. Mesenchymal stem cells (MSCs) give rise to adipocytes and fibroblasts that accumulate in injured and pathological skeletal muscle through constitutive activation of platelet-derived growth factor receptors (PDGFRs). Although the role of the PDGFRα has been widely explored, there is a paucity of evidence demonstrating the role of PDGFRβ in aged skeletal muscle. Methods In this study, we investigated the role of PDGFRβ lineage cells in skeletal muscle during aging by using Cre/loxP lineage tracing technology. The PDGFR-Cre mice were crossed with global double-fluorescent Cre reporter mice (mTmG) that indelibly marks PDGFRβ lineage cells. Those cells were analyzed and compared at different ages in the skeletal muscle of the mice. Results Our results demonstrated that PDGFRβ lineage cells isolated from the muscles of young mice are MPC-like cells that exhibited satellite cell morphology, expressed Pax7, and undergo myogenic differentiation producing myosin heavy chain expressing myotubes. Conversely, the PDGFRβ lineage cells isolated from muscles of old mice displayed MSC morphology with a reduced myogenic differentiation potential while expressing adipogenic and fibrotic differentiation markers. PDGFRβ lineage cells also gave rise to newly regenerated muscle fibers in young mice after muscle injury, but their muscle regenerative process is reduced in old mice. Conclusions Our data suggest that PDGFRβ lineage cells function as MPCs in young mice, while the same PDGFRβ lineage cells from old mice undergo a fate switch participating in adipose and fibrotic tissue infiltration in aged muscle. The inhibition of fate-switching in PDGFRβ lineage cells may represent a potential approach to prevent fibrosis and fatty infiltration in skeletal muscle during the aging process.https://doi.org/10.1186/s13287-022-03072-yMesenchymal stem cellsPDGFRβ lineage cellsAgingMuscle progenitor cellSkeletal muscle injuryFibrosis and fatty infiltration
spellingShingle Aiping Lu
Chieh Tseng
Ping Guo
Zhanguo Gao
Kaitlyn E. Whitney
Mikhail G. Kolonin
Johnny Huard
The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
Stem Cell Research & Therapy
Mesenchymal stem cells
PDGFRβ lineage cells
Aging
Muscle progenitor cell
Skeletal muscle injury
Fibrosis and fatty infiltration
title The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
title_full The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
title_fullStr The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
title_full_unstemmed The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
title_short The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair
title_sort role of the aging microenvironment on the fate of pdgfrβ lineage cells in skeletal muscle repair
topic Mesenchymal stem cells
PDGFRβ lineage cells
Aging
Muscle progenitor cell
Skeletal muscle injury
Fibrosis and fatty infiltration
url https://doi.org/10.1186/s13287-022-03072-y
work_keys_str_mv AT aipinglu theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT chiehtseng theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT pingguo theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT zhanguogao theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT kaitlynewhitney theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT mikhailgkolonin theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT johnnyhuard theroleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT aipinglu roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT chiehtseng roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT pingguo roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT zhanguogao roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT kaitlynewhitney roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT mikhailgkolonin roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair
AT johnnyhuard roleoftheagingmicroenvironmentonthefateofpdgfrblineagecellsinskeletalmusclerepair