Abstract

Interleukin-36γ (IL-36γ), a member of the IL-1 cytokine superfamily, amplifies lung inflammation and impairs host defense during acute pulmonary Pseudomonas aeruginosa infection. To be fully active, IL-36γ is cleaved at its N-terminal region by proteases such as neutrophil elastase (NE) and cathepsin S (CatS). However, it remains unclear whether limiting extracellular proteolysis restrains the inflammatory cascade triggered by IL-36γ during P. aeruginosa infection. Thrombospondin-1 (TSP-1) is a matricellular protein with inhibitory activity against NE and the pathogen-secreted Pseudomonas elastase LasB-both proteases implicated in amplifying inflammation. We hypothesized that TSP-1 tempers the inflammatory response during lung P. aeruginosa infection by inhibiting the proteolytic environment required for IL-36γ activation. Compared to wild-type (WT) mice, TSP-1-deficient (Thbs1-/-) mice exhibited a hyperinflammatory response in the lungs during P. aeruginosa infection, with increased cytokine production and an unrestrained extracellular proteolytic environment characterized by higher free NE and LasB, but not CatS activity. LasB cleaved IL-36γ proximally to M19 at a cleavage site distinct from those generated by NE and CatS, which cleave IL-36γ proximally to Y16 and S18, respectively. N-terminal truncation experiments in silico predicted that the M19 and the S18 isoforms bind the IL-36R complex almost identically. IL-36γ neutralization ameliorated the hyperinflammatory response and improved lung immunity in Thbs1-/- mice during P. aeruginosa infection. Moreover, administration of cleaved IL-36γ induced cytokine production and neutrophil recruitment and activation that was accentuated in Thbs1-/- mice lungs. Collectively, our data show that TSP-1 regulates lung neutrophilic inflammation and facilitates host defense by restraining the extracellular proteolytic environment required for IL-36γ activation.IMPORTANCEPseudomonas aeruginosa pulmonary infection can lead to exaggerated neutrophilic inflammation and tissue destruction, yet host factors that regulate the neutrophilic response are not fully known. IL-36γ is a proinflammatory cytokine that dramatically increases in bioactivity following N-terminal processing by proteases. Here, we demonstrate that thrombospondin-1, a host matricellular protein, limits N-terminal processing of IL-36γ by neutrophil elastase and the Pseudomonas aeruginosa-secreted protease LasB. Thrombospondin-1-deficient mice (Thbs1-/-) exhibit a hyperinflammatory response following infection. Whereas IL-36γ neutralization reduces inflammatory cytokine production, limits neutrophil activation, and improves host defense in Thbs1-/- mice, cleaved IL-36γ administration amplifies neutrophilic inflammation in Thbs1-/- mice. Our findings indicate that thrombospondin-1 guards against feed-forward neutrophilic inflammation mediated by IL-36γ in the lung by restraining the extracellular proteolytic environment.

Highlights

  • Interleukin-36g (IL-36g), a member of the IL-1 cytokine superfamily, amplifies lung inflammation and impairs host defense during acute pulmonary Pseudomonas aeruginosa infection

  • As N-terminal processing of IL-36g is required for full bioactivity and the triggering of proinflammatory cytokines IL-6 and CXCL-1 by IL36R in bone marrow-derived dendritic cells (BMDCs) and human keratinocytes [39, 40], we show that cleaved IL-36g just proximal to S18 but not full-length IL-36g leads to a robust production of IL-6 and CXCL-1 by BMDCs in vitro (Fig. 2F)

  • Our findings indicate TSP-1 regulates the inflammatory response in the lungs mediated by IL-36g during P. aeruginosa lower respiratory tract infection by restraining the extracellular proteolytic environment

Read more

Summary

Introduction

Interleukin-36g (IL-36g), a member of the IL-1 cytokine superfamily, amplifies lung inflammation and impairs host defense during acute pulmonary Pseudomonas aeruginosa infection. IL-36g is cleaved at its N-terminal region by proteases such as neutrophil elastase (NE) and cathepsin S (CatS) It remains unclear whether limiting extracellular proteolysis restrains the inflammatory cascade triggered by IL-36g during P. aeruginosa infection. Compared to wildtype (WT) mice, TSP-1-deficient (Thbs12/2) mice exhibited a hyperinflammatory response in the lungs during P. aeruginosa infection, with increased cytokine production and an unrestrained extracellular proteolytic environment characterized by higher free NE and LasB, but not CatS activity. IL-36g appears to be protective during lung infection caused by different pathogenic bacteria such as Streptococcus pneumoniae, Klebsiella pneumoniae, and Legionella pneumophila [30, 34] In contrast to these infections, IL-36g is paradoxically harmful following P. aeruginosa infection-induced injury, as IL36R-deficient (IL-36R2/2) and IL-36g2/2 mice are protected from excessive host inflammatory response and show improved lung bacterial clearance [31]. These findings suggest that while IL-36g is important in early host defense, excessive inflammation mediated by IL-36g may be a rational target against P. aeruginosa-induced lung tissue damage

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