Abstract

BackgroundChronic granulomatous disease (CGD), an inherited disorder of the NADPH oxidase in which phagocytes are defective in generating superoxide anion and downstream reactive oxidant intermediates (ROIs), is characterized by recurrent bacterial and fungal infections and by excessive inflammation (e.g., inflammatory bowel disease). The mechanisms by which NADPH oxidase regulates inflammation are not well understood.Methodology/Principal FindingsWe found that NADPH oxidase restrains inflammation by modulating redox-sensitive innate immune pathways. When challenged with either intratracheal zymosan or LPS, NADPH oxidase-deficient p47phox−/− mice and gp91phox-deficient mice developed exaggerated and progressive lung inflammation, augmented NF-κB activation, and elevated downstream pro-inflammatory cytokines (TNF-α, IL-17, and G-CSF) compared to wildtype mice. Replacement of functional NADPH oxidase in bone marrow-derived cells restored the normal lung inflammatory response. Studies in vivo and in isolated macrophages demonstrated that in the absence of functional NADPH oxidase, zymosan failed to activate Nrf2, a key redox-sensitive anti-inflammatory regulator. The triterpenoid, CDDO-Im, activated Nrf2 independently of NADPH oxidase and reduced zymosan-induced lung inflammation in CGD mice. Consistent with these findings, zymosan-treated peripheral blood mononuclear cells from X-linked CGD patients showed impaired Nrf2 activity and increased NF-κB activation.Conclusions/SignificanceThese studies support a model in which NADPH oxidase-dependent, redox-mediated signaling is critical for termination of lung inflammation and suggest new potential therapeutic targets for CGD.

Highlights

  • The lung is an interface where inhaled microbes and antigens interact with host defense cells

  • We evaluated whether NADPH oxidase activity would counterbalance the immediate pro-inflammatory events that follow Pathogen recognition receptors (PRRs) signaling by interacting with redox-sensitive pathways to dampen inflammation

  • We asked whether NADPH oxidase, which is activated by bacterial and fungal pathogens, would have a role in restraining lung inflammation induced by microbial motifs

Read more

Summary

Introduction

The lung is an interface where inhaled microbes and antigens interact with host defense cells. Consistent with these in vivo findings, zymosan treatment resulted in increased NF-kB activation in isolated p47phox2/2 bone marrow-derived macrophages (BMDMs) compared to WT Since it is possible that p47phox can modulate inflammation independently of NADPH oxidase, we evaluated zymosan-induced lung inflammation in X-linked gp91phox2/ CGD mice [21].

Results
Conclusion
Full Text
Paper version not known

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