Your browser doesn't support javascript.
loading
Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease.
Singanayagam, Aran; Footitt, Joseph; Marczynski, Matthias; Radicioni, Giorgia; Cross, Michael T; Finney, Lydia J; Trujillo-Torralbo, Maria-Belen; Calderazzo, Maria; Zhu, Jie; Aniscenko, Julia; Clarke, Thomas B; Molyneaux, Philip L; Bartlett, Nathan W; Moffatt, Miriam F; Cookson, William O; Wedzicha, Jadwiga; Evans, Christopher M; Boucher, Richard C; Kesimer, Mehmet; Lieleg, Oliver; Mallia, Patrick; Johnston, Sebastian L.
Afiliação
  • Singanayagam A; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Footitt J; Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom.
  • Marczynski M; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Radicioni G; School of Engineering and Design, Department of Materials Engineering and.
  • Cross MT; Center for Protein Assemblies, Technical University of Munich, Munich, Germany.
  • Finney LJ; Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Trujillo-Torralbo MB; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA.
  • Calderazzo M; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Zhu J; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Aniscenko J; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Clarke TB; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Molyneaux PL; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Bartlett NW; Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom.
  • Moffatt MF; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Cookson WO; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Wedzicha J; College of Health, Medicine and Wellbeing, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia.
  • Evans CM; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Boucher RC; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Kesimer M; National Heart and Lung Institute, Imperial College London, London, United Kingdom.
  • Lieleg O; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA.
  • Mallia P; Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Johnston SL; Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
J Clin Invest ; 132(8)2022 04 15.
Article em En | MEDLINE | ID: mdl-35239513
ABSTRACT
The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases, but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway mucin 5AC (MUC5AC) and MUC5B concentrations during spontaneous and experimentally induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with viral load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation, as Muc5ac-deficient (Muc5ac-/-) mice had attenuated RV-induced (RV-induced) airway inflammation, and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased the release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of RV-induced inflammation in mice. Therapeutic suppression of mucin production using an EGFR antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B expression suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identified a proinflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença Pulmonar Obstrutiva Crônica / Mucina-5AC Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Reino Unido

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Doença Pulmonar Obstrutiva Crônica / Mucina-5AC Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: J Clin Invest Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Reino Unido