Your browser doesn't support javascript.
loading
Respiratory SARS-CoV-2 Infection Causes Skeletal Muscle Atrophy and Long-Lasting Energy Metabolism Suppression.
Homma, Sachiko T; Wang, Xingyu; Frere, Justin J; Gower, Adam C; Zhou, Jingsong; Lim, Jean K; tenOever, Benjamin R; Zhou, Lan.
Afiliação
  • Homma ST; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA.
  • Wang X; Department of Neurology, Hospital for Special Surgery, New York, NY 10021, USA.
  • Frere JJ; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
  • Gower AC; Clinical and Translational Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA.
  • Zhou J; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA.
  • Lim JK; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
  • tenOever BR; Department of Microbiology, Grossman School of Medicine, New York University, New York, NY 10016, USA.
  • Zhou L; Department of Neurology, Hospital for Special Surgery, New York, NY 10021, USA.
Biomedicines ; 12(7)2024 Jun 28.
Article em En | MEDLINE | ID: mdl-39062017
ABSTRACT
Muscle fatigue represents the most prevalent symptom of long-term COVID, with elusive pathogenic mechanisms. We performed a longitudinal study to characterize histopathological and transcriptional changes in skeletal muscle in a hamster model of respiratory SARS-CoV-2 infection and compared them with influenza A virus (IAV) and mock infections. Histopathological and bulk RNA sequencing analyses of leg muscles derived from infected animals at days 3, 30, and 60 post-infection showed no direct viral invasion but myofiber atrophy in the SARS-CoV-2 group, which was accompanied by persistent downregulation of the genes related to myofibers, ribosomal proteins, fatty acid ß-oxidation, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation complexes. While both SARS-CoV-2 and IAV infections induced acute and transient type I and II interferon responses in muscle, only the SARS-CoV-2 infection upregulated TNF-α/NF-κB but not IL-6 signaling in muscle. Treatment of C2C12 myotubes, a skeletal muscle cell line, with combined IFN-γ and TNF-α but not with IFN-γ or TNF-α alone markedly impaired mitochondrial function. We conclude that a respiratory SARS-CoV-2 infection can cause myofiber atrophy and persistent energy metabolism suppression without direct viral invasion. The effects may be induced by the combined systemic interferon and TNF-α responses at the acute phase and may contribute to post-COVID-19 persistent muscle fatigue.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Biomedicines Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Biomedicines Ano de publicação: 2024 Tipo de documento: Article