Your browser doesn't support javascript.
loading
Mitochondrial dysfunction in macrophages promotes inflammation and suppresses repair after myocardial infarction.
Cai, Shanshan; Zhao, Mingyue; Zhou, Bo; Yoshii, Akira; Bugg, Darrian; Villet, Outi; Sahu, Anita; Olson, Gregory S; Davis, Jennifer; Tian, Rong.
Afiliação
  • Cai S; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Zhao M; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Zhou B; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Yoshii A; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Bugg D; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA.
  • Villet O; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Sahu A; Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and.
  • Olson GS; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA.
  • Davis J; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, Washington, USA.
  • Tian R; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA.
J Clin Invest ; 133(4)2023 02 15.
Article em En | MEDLINE | ID: mdl-36480284
ABSTRACT
Innate immune cells play important roles in tissue injury and repair following acute myocardial infarction (MI). Although reprogramming of macrophage metabolism has been observed during inflammation and resolution phases, the mechanistic link to macrophage phenotype is not fully understood. In this study, we found that myeloid-specific deletion (mKO) of mitochondrial complex I protein, encoded by Ndufs4, reproduced the proinflammatory metabolic profile in macrophages and exaggerated the response to LPS. Moreover, mKO mice showed increased mortality, poor scar formation, and worsened cardiac function 30 days after MI. We observed a greater inflammatory response in mKO mice on day 1 followed by increased cell death of infiltrating macrophages and blunted transition to the reparative phase during post-MI days 3-7. Efferocytosis was impaired in mKO macrophages, leading to lower expression of antiinflammatory cytokines and tissue repair factors, which suppressed the proliferation and activation of myofibroblasts in the infarcted area. Mitochondria-targeted ROS scavenging rescued these impairments, improved myofibroblast function in vivo, and reduced post-MI mortality in mKO mice. Together these results reveal a critical role of mitochondria in inflammation resolution and tissue repair via modulation of efferocytosis and crosstalk with fibroblasts. These findings have potential significance for post-MI recovery as well as for other inflammatory conditions.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Macrófagos / Mitocôndrias / Infarto do Miocárdio Limite: Animals Idioma: En Revista: J Clin Invest Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Macrófagos / Mitocôndrias / Infarto do Miocárdio Limite: Animals Idioma: En Revista: J Clin Invest Ano de publicação: 2023 Tipo de documento: Article