Abstract

Interferon-inducible protein (IFI204) (p204, the murine homolog of human IFI16) is known as a cytosolic DNA sensor to recognize DNA viruses and intracellular bacteria. However, little is known about its role during extracellular bacterial infection. Here we show that IFI204 is required for host defense against the infection of Staphylococcus aureus, an extracellular bacterial pathogen. IFI204 deficiency results in decreased survival, increased bacterial loads, severe organs damage, and decreased recruitment of neutrophils and macrophages. Production of several inflammatory cytokines/chemokines including IFN-β and KC is markedly decreased, as well as the related STING-IRF3 and NF-κB pathways are impaired. However, exogenous administration of recombinant KC or IFN-β is unable to rescue the susceptibility of IFI204-deficient mice, suggesting that other mechanisms rather than KC and IFN-β account for IFI204-mediated host defense. IFI204 deficiency leads to a defect in extracellular bacterial killing in macrophages and neutrophils, although bacterial engulf, and intracellular killing activity are normal. Moreover, the defect of bactericidal activity is mediated by decreased extracellular trap formation in the absence of IFI204. Adoptively transferred WT bone marrow cells significantly protect WT and IFI204-deficient recipients against Staphylococcus infection compared with transferred IFI204-deficient bone marrow cells. Hence, this study suggests that IFI204 is essential for the host defense against Staphylococcus infection.

Highlights

  • The pathogen Staphylococcus aureus persistently colonizes a large proportion of the human population and is a frequent cause of skin and soft tissue infections, pneumonia, and sepsis

  • More bacteria were detected in the lungs, Bronchoalveolar lavage fluid (BALF) and blood of interferon-inducible protein 204 (IFI204)−/− mice compared with WT mice (Figures 1A–F)

  • Several studies including ours showed that murine IFI204 or human ortholog interferon gamma inducible protein 16 (IFI16) detects cytosolic bacterial DNA for the type I IFN response or cell death in vitro [5,6,7,8]

Read more

Summary

Introduction

The pathogen Staphylococcus aureus persistently colonizes a large proportion of the human population and is a frequent cause of skin and soft tissue infections, pneumonia, and sepsis. Despite intense research in understanding the pathogenesis and host-pathogen interaction, the mechanisms by which Staphylococcus is cleared from the host are largely unclarified, thereby impeding the development of novel strategies for control of this infection. The innate immune system plays a key role in the early recognition and elimination of invading pathogens. Sensing bacteria through pattern recognition receptors (PRRs) enables innate immune cells to categorize microbial invaders and to initiate appropriate signaling cascades that mobilize. Characterized as a DNA sensor, interferon-inducible protein 204 (IFI204) (its human ortholog IFI16) is one member of PRRs that detects cytosolic DNA for the type I IFN response [2]. In response to cytosolic DNA stimulation or virus infection, IFI204/IFI16 interacts with STING to induce TBK1-dependent IFN-β production. Several studies reported that IFI204/IFI16 recognizes DNA viral genomes in the nucleus and activates the inflammasome pathway through ASC and caspase-1, leading to IL-1β, and IL-18 production [3, 4]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call