Your browser doesn't support javascript.
loading
Identification of Periostin as a critical niche for myofibroblast dynamics and fibrosis during tendon healing.
Ackerman, Jessica E; Adjei-Sowah, Emmanuela; Korcari, Antonion; Muscat, Samantha N; Nichols, Anne E C; Buckley, Mark R; Loiselle, Alayna E.
Afiliação
  • Ackerman JE; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY.
  • Adjei-Sowah E; Current affiliation: NDORMS, University of Oxford, Oxford, United Kingdom.
  • Korcari A; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY.
  • Muscat SN; Department of Biomedical Engineering, University of Rochester, Rochester, NY.
  • Nichols AEC; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY.
  • Buckley MR; Department of Biomedical Engineering, University of Rochester, Rochester, NY.
  • Loiselle AE; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY.
bioRxiv ; 2023 Jul 21.
Article em En | MEDLINE | ID: mdl-37502924
ABSTRACT
Tendon injuries are a major clinical problem, with poor patient outcomes caused by abundant scar tissue deposition during healing. Myofibroblasts play a critical role in the initial restoration of structural integrity after injury. However, persistent myofibroblast activity drives the transition to fibrotic scar tissue formation. As such, disrupting myofibroblast persistence is a key therapeutic target. While myofibroblasts are typically defined by the presence of αSMA+ stress fibers, αSMA is expressed in other cell types including the vasculature. As such, modulation of myofibroblast dynamics via disruption of αSMA expression is not a translationally tenable approach. Recent work has demonstrated that Periostin-lineage (PostnLin) cells are a precursor for cardiac fibrosis-associated myofibroblasts. In contrast to this, here we show that PostnLin cells contribute to a transient αSMA+ myofibroblast population that is required for functional tendon healing, and that Periostin forms a supportive matrix niche that facilitates myofibroblast differentiation and persistence. Collectively, these data identify the Periostin matrix niche as a critical regulator of myofibroblast fate and persistence that could be targeted for therapeutic manipulation to facilitate regenerative tendon healing.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Diagnostic_studies Idioma: En Revista: BioRxiv Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Diagnostic_studies Idioma: En Revista: BioRxiv Ano de publicação: 2023 Tipo de documento: Article