Your browser doesn't support javascript.
loading
Immature myeloid cells are critical for enhancing bone fracture healing through angiogenic cascade.
Levy, Seth; Feduska, Joseph M; Sawant, Anandi; Gilbert, Shawn R; Hensel, Jonathan A; Ponnazhagan, Selvarangan.
Afiliação
  • Levy S; Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States.
  • Feduska JM; Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States.
  • Sawant A; Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States.
  • Gilbert SR; Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States.
  • Hensel JA; Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States.
  • Ponnazhagan S; Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States. Electronic address: pons@uab.edu.
Bone ; 93: 113-124, 2016 12.
Article em En | MEDLINE | ID: mdl-27664567
ABSTRACT
Bone fractures heal with overlapping phases of inflammation, cell proliferation, and bone remodeling. Osteogenesis and angiogenesis work in concert to control many stages of this process, and when one is impaired it leads to failure of bone healing, termed a nonunion. During fracture repair, there is an infiltration of immune cells at the fracture site that not only mediate the inflammatory responses, but we hypothesize they also exert influence on neovasculature. Thus, further understanding the effects of immune cell participation throughout fracture healing will reveal additional knowledge as to why some fractures heal while others form nonunions, and lead to development of novel therapeutics modulating immune cells, to increase fracture healing and prevent nonunions. Using novel femoral segmental and critical-size defect models in mice, we identified a systemic and significant increase in immature myeloid cell (IMC) infiltration during the initial phase of fracture healing until boney union is complete. Using gemcitabine to specifically ablate the IMC population, we confirmed delayed bone healing. Further, adoptive transfer of IMC increased bone growth in a nonunion model, signifying the role of this unique cell population in fracture healing. We also identified IMC post-fracture have the ability to increase endothelial cell migration, and tube formation, signaling the essential communication between the immune system and angiogenesis as a requirement for proper bone healing. Based on this data we propose that IMC may play a significant role in fracture healing and therapeutic targeting of IMC after fracture would minimize the chances of eventual nonunion pathology.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Consolidação da Fratura / Neovascularização Fisiológica / Células Mieloides / Fraturas Ósseas Limite: Animals / Female / Humans Idioma: En Revista: Bone Assunto da revista: METABOLISMO / ORTOPEDIA Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Estados Unidos País de publicação: EEUU / ESTADOS UNIDOS / ESTADOS UNIDOS DA AMERICA / EUA / UNITED STATES / UNITED STATES OF AMERICA / US / USA

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Consolidação da Fratura / Neovascularização Fisiológica / Células Mieloides / Fraturas Ósseas Limite: Animals / Female / Humans Idioma: En Revista: Bone Assunto da revista: METABOLISMO / ORTOPEDIA Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Estados Unidos País de publicação: EEUU / ESTADOS UNIDOS / ESTADOS UNIDOS DA AMERICA / EUA / UNITED STATES / UNITED STATES OF AMERICA / US / USA