GP96 Drives Exacerbation of Secondary Bacterial Pneumonia following Influenza A Virus Infection

mBio. 2021 Jun 29;12(3):e0326920. doi: 10.1128/mBio.03269-20. Epub 2021 Jun 1.

Abstract

Influenza A virus (IAV) infection predisposes the host to secondary bacterial pneumonia, known as a major cause of morbidity and mortality during influenza virus epidemics. Analysis of interactions between IAV-infected human epithelial cells and Streptococcus pneumoniae revealed that infected cells ectopically exhibited the endoplasmic reticulum chaperone glycoprotein 96 (GP96) on the surface. Importantly, efficient pneumococcal adherence to epithelial cells was imparted by interactions with extracellular GP96 and integrin αV, with the surface expression mediated by GP96 chaperone activity. Furthermore, abrogation of adherence was gained by chemical inhibition or genetic knockout of GP96 as well as addition of RGD peptide, an inhibitor of integrin-ligand interactions. Direct binding of extracellular GP96 and pneumococci was shown to be mediated by pneumococcal oligopeptide permease components. Additionally, IAV infection induced activation of calpains and Snail1, which are responsible for degradation and transcriptional repression of junctional proteins in the host, respectively, indicating increased bacterial translocation across the epithelial barrier. Notably, treatment of IAV-infected mice with the GP96 inhibitor enhanced pneumococcal clearance from lung tissues and ameliorated lung pathology. Taken together, the present findings indicate a viral-bacterial synergy in relation to disease progression and suggest a paradigm for developing novel therapeutic strategies tailored to inhibit pneumococcal colonization in an IAV-infected respiratory tract. IMPORTANCE Secondary bacterial pneumonia following an influenza A virus (IAV) infection is a major cause of morbidity and mortality. Although it is generally accepted that preceding IAV infection leads to increased susceptibility to secondary bacterial infection, details regarding the pathogenic mechanism during the early stage of superinfection remain elusive. Here, we focused on the interaction of IAV-infected cells and Streptococcus pneumoniae, which revealed that human epithelial cells infected with IAV exhibit a cell surface display of GP96, an endoplasmic reticulum chaperon. Notably, extracellular GP96 was shown to impart efficient adherence for secondary infection by S. pneumoniae, and GP96 inhibition ameliorated lung pathology of superinfected mice, indicating it to be a useful target for development of therapeutic strategies for patients with superinfection.

Keywords: Streptococcus pneumoniae; influenza virus; pneumonia; superinfection.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • A549 Cells
  • Animals
  • Bacterial Adhesion
  • Coinfection / complications
  • Coinfection / microbiology
  • Coinfection / virology
  • Epithelial Cells / microbiology
  • Female
  • Humans
  • Influenza A virus / pathogenicity*
  • Influenza, Human / complications*
  • Influenza, Human / virology
  • Lung / microbiology
  • Membrane Glycoproteins / genetics*
  • Mice
  • Mice, Inbred BALB C
  • Orthomyxoviridae Infections / microbiology
  • Orthomyxoviridae Infections / virology
  • Pneumonia, Bacterial / etiology
  • Pneumonia, Bacterial / pathology
  • Pneumonia, Bacterial / virology*
  • Streptococcus pneumoniae / pathogenicity*
  • Symptom Flare Up*

Substances

  • Membrane Glycoproteins
  • endoplasmin