Your browser doesn't support javascript.
loading
Osteochondroprogenitor cells and neutrophils expressing p21 and senescence markers modulate fracture repair.
Saul, Dominik; Doolittle, Madison L; Rowsey, Jennifer L; Froemming, Mitchell N; Kosinsky, Robyn L; Vos, Stephanie J; Ruan, Ming; LeBrasseur, Nathan K; Chandra, Abhishek; Pignolo, Robert J; Passos, João F; Farr, Joshua N; Monroe, David G; Khosla, Sundeep.
Affiliation
  • Saul D; Division of Endocrinology and.
  • Doolittle ML; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA.
  • Rowsey JL; Department of Trauma and Reconstructive Surgery, BG Clinic, University of Tübingen, Tübingen, Germany.
  • Froemming MN; Division of Endocrinology and.
  • Kosinsky RL; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA.
  • Vos SJ; Division of Endocrinology and.
  • Ruan M; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA.
  • LeBrasseur NK; Division of Endocrinology and.
  • Chandra A; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA.
  • Pignolo RJ; Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA.
  • Passos JF; Robert Bosch Center for Tumor Diseases, Stuttgart, Germany.
  • Farr JN; Division of Endocrinology and.
  • Monroe DG; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA.
  • Khosla S; Division of Endocrinology and.
J Clin Invest ; 134(12)2024 May 16.
Article in En | MEDLINE | ID: mdl-38753433
ABSTRACT
Cells expressing features of senescence, including upregulation of p21 and p16, appear transiently following tissue injury, yet the properties of these cells or how they contrast with age-induced senescent cells remains unclear. Here, we used skeletal injury as a model and identified the rapid appearance following fracture of p21+ cells expressing senescence markers, mainly as osteochondroprogenitors (OCHs) and neutrophils. Targeted genetic clearance of p21+ cells suppressed senescence-associated signatures within the fracture callus and accelerated fracture healing. By contrast, p21+ cell clearance did not alter bone loss due to aging; conversely, p16+ cell clearance, known to alleviate skeletal aging, did not affect fracture healing. Following fracture, p21+ neutrophils were enriched in signaling pathways known to induce paracrine stromal senescence, while p21+ OCHs were highly enriched in senescence-associated secretory phenotype factors known to impair bone formation. Further analysis revealed an injury-specific stem cell-like OCH subset that was p21+ and highly inflammatory, with a similar inflammatory mesenchymal population (fibro-adipogenic progenitors) evident following muscle injury. Thus, intercommunicating senescent-like neutrophils and mesenchymal progenitor cells were key regulators of tissue repair in bone and potentially across tissues. Moreover, our findings established contextual roles of p21+ versus p16+ senescent/senescent-like cells that may be leveraged for therapeutic opportunities.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cellular Senescence / Fracture Healing / Cyclin-Dependent Kinase Inhibitor p21 / Neutrophils Limits: Animals Language: En Journal: J Clin Invest Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cellular Senescence / Fracture Healing / Cyclin-Dependent Kinase Inhibitor p21 / Neutrophils Limits: Animals Language: En Journal: J Clin Invest Year: 2024 Document type: Article