Your browser doesn't support javascript.
loading
Influenza viral neuraminidase primes bacterial coinfection through TGF-ß-mediated expression of host cell receptors.
Li, Ning; Ren, Aihui; Wang, Xiaoshuang; Fan, Xin; Zhao, Yong; Gao, George F; Cleary, Patrick; Wang, Beinan.
Afiliação
  • Li N; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and.
  • Ren A; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and.
  • Wang X; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and.
  • Fan X; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and.
  • Zhao Y; State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; and.
  • Gao GF; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and.
  • Cleary P; Department of Microbiology, University of Minnesota, Minneapolis, MN 55455.
  • Wang B; Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, and wangbn@im.ac.cn.
Proc Natl Acad Sci U S A ; 112(1): 238-43, 2015 Jan 06.
Article em En | MEDLINE | ID: mdl-25535343
ABSTRACT
Influenza infection predisposes the host to secondary bacterial pneumonia, which is a major cause of mortality during influenza epidemics. The molecular mechanisms underlying the bacterial coinfection remain elusive. Neuraminidase (NA) of influenza A virus (IAV) enhances bacterial adherence and also activates TGF-ß. Because TGF-ß can up-regulate host adhesion molecules such as fibronectin and integrins for bacterial binding, we hypothesized that activated TGF-ß during IAV infection contributes to secondary bacterial infection by up-regulating these host adhesion molecules. Flow cytometric analyses of a human lung epithelial cell line indicated that the expression of fibronectin and α5 integrin was up-regulated after IAV infection or treatment with recombinant NA and was reversed through the inhibition of TGF-ß signaling. IAV-promoted adherence of group A Streptococcus (GAS) and other coinfective pathogens that require fibronectin for binding was prevented significantly by the inhibition of TGF-ß. However, IAV did not promote the adherence of Lactococcus lactis unless this bacterium expressed the fibronectin-binding protein of GAS. Mouse experiments showed that IAV infection enhanced GAS colonization in the lungs of wild-type animals but not in the lungs of mice deficient in TGF-ß signaling. Taken together, these results reveal a previously unrecognized mechanism IAV NA enhances the expression of cellular adhesins through the activation of TGF-ß, leading to increased bacterial loading in the lungs. Our results suggest that TGF-ß and cellular adhesins may be potential pharmaceutical targets for the prevention of coinfection.
Assuntos
Palavras-chave

Texto completo: 1 Bases de dados: MEDLINE Assunto principal: Vírus da Influenza A / Infecções Estreptocócicas / Fator de Crescimento Transformador beta / Receptores de Superfície Celular / Coinfecção / Neuraminidase Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2015 Tipo de documento: Article

Texto completo: 1 Bases de dados: MEDLINE Assunto principal: Vírus da Influenza A / Infecções Estreptocócicas / Fator de Crescimento Transformador beta / Receptores de Superfície Celular / Coinfecção / Neuraminidase Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2015 Tipo de documento: Article