NEUROGENIC HETEROTOPIC OSSIFICATIONS ARE DERIVED FROM FIBROADIPOGENIC PROGENITORS IN THE SKELETAL MUSCLE NOT FROM SATELLITE CELLS

التفاصيل البيبلوغرافية
العنوان: NEUROGENIC HETEROTOPIC OSSIFICATIONS ARE DERIVED FROM FIBROADIPOGENIC PROGENITORS IN THE SKELETAL MUSCLE NOT FROM SATELLITE CELLS
المؤلفون: Kylie A. Alexander, Irina Kulina, Bianca Nowlan, Susan M. Millard, François Genêt, Marjorie Salga, Hsu-Wen Tseng, Allison R. Pettit, Whitney Fleming, Beulah Jose, Jean-Pierre Levesque
المصدر: Experimental Hematology. 76:S89
بيانات النشر: Elsevier BV, 2019.
سنة النشر: 2019
مصطلحات موضوعية: 0301 basic medicine, Cancer Research, biology, Mesenchymal stem cell, Skeletal muscle, Cell Biology, Hematology, Cell biology, 03 medical and health sciences, 030104 developmental biology, 0302 clinical medicine, medicine.anatomical_structure, 030220 oncology & carcinogenesis, Genetics, biology.protein, medicine, Myocyte, PAX7, Progenitor cell, Stem cell, Molecular Biology, Platelet-derived growth factor receptor, Progenitor
الوصف: Neurological heterotopic ossifications (NHO) are frequent complications of traumatic brain and spinal cord injuries (SCI). NHO are abnormal ossifications within periarticular muscles resulting in joint ankylosis, vessel and nerve compression. To elucidate NHO pathogenesis, we have developed the first murine model of SCI-NHO, in which NHO is induced in cardiotoxin-injured muscle only following a SCI. However, what stem cell populations within muscle differentiate into osteoblasts and directly contribute to NHO is unclear. We hypothesized that injured muscles do not repair normally after SCI, and instead osteogenic differentiation of skeletal muscle-resident stem cells takes place. We focused on the two progenitor populations known to reside in muscles: satellite cells (SCs) and fibro- adipogenic progenitors (FAPs). SCs, which specifically express the transcription factor Pax7, differentiate into myoblasts, whereas FAPs, which express are of mesenchymal origin and express Prrx1+ instead, can differentiate into fibroblasts and adipocytes in vitro. We had shown that both populations can deposit calcium under osteogenic condition in vitro. We have now generated Pax7CreER x R26-ZsGreen (Pax7ZsG) and Prrx1Cre x R26-ZsGreen (Prrx1ZsG) to specifically trace the fate of SCs and FAPs respectively in our NHO model in vivo. We demonstrate that after SCI, Pax7+ SCs fail to proliferate and regenerate myofibers with instead persistent proliferation of Prrx1+ FAPs differentiating into osteocalcin+ osteoblasts in SCI-NHO at 21 days post-surgery. The above suggest that SCI causes muscle repair failure with instead proliferation and osteogenic differentiation of FAPs residing in the muscle. In addition, SCI significantly upregulated platelet-derived growth factor receptor (PDGFR) expression in FAPs indicating dysregulation of PDGFR signalling may be involved in the NHO development.
تدمد: 0301-472X
الوصول الحر: https://explore.openaire.eu/search/publication?articleId=doi_________::89bcdde3dacdb105eb870dd41f02637dTest
https://doi.org/10.1016/j.exphem.2019.06.449Test
حقوق: CLOSED
رقم الانضمام: edsair.doi...........89bcdde3dacdb105eb870dd41f02637d
قاعدة البيانات: OpenAIRE