Rhinovirus attenuates non-typeable Hemophilus influenzae-stimulated IL-8 responses via TLR2-dependent degradation of IRAK-1

PLoS Pathog. 2012;8(10):e1002969. doi: 10.1371/journal.ppat.1002969. Epub 2012 Oct 4.

Abstract

Bacterial infections following rhinovirus (RV), a common cold virus, are well documented, but pathogenic mechanisms are poorly understood. We developed animal and cell culture models to examine the effects of RV on subsequent infection with non-typeable Hemophilus influenzae (NTHi). We focused on NTHI-induced neutrophil chemoattractants expression that is essential for bacterial clearance. Mice infected with RV1B were superinfected with NTHi and lung bacterial density, chemokines and neutrophil counts determined. Human bronchial epithelial cells (BEAS-2B) or mouse alveolar macrophages (MH-S) were infected with RV and challenged with NHTi, TLR2 or TLR5 agonists. Chemokine levels were measured by ELISA and expression of IRAK-1, a component of MyD88-dependent TLR signaling, assessed by immunoblotting. While sham-infected mice cleared all NTHi from the lungs, RV-infected mice showed bacteria up to 72 h post-infection. However, animals in RV/NTHi cleared bacteria by day 7. Delayed bacterial clearance in RV/NTHi animals was associated with suppressed chemokine levels and neutrophil recruitment. RV-infected BEAS-2B and MH-S cells showed attenuated chemokine production after challenge with either NTHi or TLR agonists. Attenuated chemokine responses were associated with IRAK-1 protein degradation. Inhibition of RV-induced IRAK-1 degradation restored NTHi-stimulated IL-8 expression. Knockdown of TLR2, but not other MyD88-dependent TLRs, also restored IRAK-1, suggesting that TLR2 is required for RV-induced IRAK-1 degradation.In conclusion, we demonstrate for the first time that RV infection delays bacterial clearance in vivo and suppresses NTHi-stimulated chemokine responses via degradation of IRAK-1. Based on these observations, we speculate that modulation of TLR-dependent innate immune responses by RV may predispose the host to secondary bacterial infection, particularly in patients with underlying chronic respiratory disorders.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Bacterial Load
  • Chemokines / blood
  • Epithelial Cells / immunology
  • Epithelial Cells / microbiology
  • Epithelial Cells / virology
  • Haemophilus Infections / complications*
  • Haemophilus Infections / immunology*
  • Haemophilus Infections / microbiology
  • Haemophilus influenzae / immunology*
  • Humans
  • Interleukin-1 Receptor-Associated Kinases / metabolism*
  • Interleukin-8 / immunology*
  • Leukocyte Count
  • Lung / microbiology
  • Macrophages, Alveolar / immunology
  • Macrophages, Alveolar / metabolism
  • Mice
  • Mice, Inbred BALB C
  • Neutrophils / immunology
  • Picornaviridae Infections / complications*
  • Picornaviridae Infections / virology
  • Rhinovirus / pathogenicity*
  • Toll-Like Receptor 2 / genetics
  • Toll-Like Receptor 2 / metabolism*
  • Toll-Like Receptor 5 / metabolism

Substances

  • Chemokines
  • Interleukin-8
  • Tlr2 protein, mouse
  • Toll-Like Receptor 2
  • Toll-Like Receptor 5
  • IRAK1 protein, human
  • Interleukin-1 Receptor-Associated Kinases
  • Irak1 protein, mouse