4.6 Article

Clearance of Senescent Cells From Injured Muscle Abrogates Heterotopic Ossification in Mouse Models of Fibrodysplasia Ossificans Progressiva

Journal

JOURNAL OF BONE AND MINERAL RESEARCH
Volume 37, Issue 1, Pages 95-107

Publisher

WILEY
DOI: 10.1002/jbmr.4458

Keywords

CELLULAR SENESCENCE; FIBRODYSPLASIA OSSIFICANS PROGRESSIVA; HETEROTOPIC OSSIFICATION; MUSCLE INJURY; SENOLYTICS

Funding

  1. Radiant Hope Foundation
  2. Robert and Arlene Kogod Professorship in Geriatric Medicine
  3. Center for Research in FOP and Related Disorders
  4. Ian Cali Endowment for FOP Research
  5. Isaac and Rose Nassau Professorship of Orthopaedic Molecular Medicine
  6. International Fibrodysplasia Ossificans Progressiva Association
  7. [P01 AG062413]

Ask authors/readers for more resources

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease caused by mutations in ACVR1/ALK2, leading to soft tissue injuries resulting in muscles transforming into ectopic bone, preventing normal movement.
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disease caused by mutations in activin A receptor type I/activin-like kinase 2 (ACVR1/ALK2), a bone morphogenetic protein (BMP) type I receptor, resulting in the formation of extraskeletal or heterotopic ossification (HO) and other features consistent with premature aging. During the first decade of life, episodic bouts of inflammatory swellings (flare-ups) occur, which are typically triggered by soft tissue trauma. Through an endochondral process, these exacerbations ultimately result in skeletal muscles, tendons, ligaments, fascia, and aponeuroses transforming into ectopic bone, rendering movement impossible. We have previously shown that soft tissue injury causes early FOP lesions characterized by cellular hypoxia, cellular damage, and local inflammation. Here we show that muscle injury in FOP also results in senescent cell accumulation, and that senescence promotes tissue reprogramming toward a chondrogenic fate in FOP muscle but not wild-type (WT) muscle. Using a combination of senolytic drugs we show that senescent cell clearance and reduction in the senescence associated secretory phenotype (SASP) ameliorate HO in mouse models of FOP. We conclude that injury-induced senescent cell burden and the SASP contribute to FOP lesion formation and that tissue reprogramming in FOP is mediated by cellular senescence, altering myogenic cell fate toward a chondrogenic cell fate. Furthermore, pharmacological removal of senescent cells abrogates tissue reprogramming and HO formation. Here we provide proof-of-principle evidence for senolytic drugs as a future therapeutic strategy in FOP. (c) 2021 American Society for Bone and Mineral Research (ASBMR).

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available