Abstract

Interferon lambda (IFNλ) signaling is a promising therapeutic target against viral infection in murine models, yet little is known about its molecular regulation and its cognate receptor, interferon lambda receptor 1 (IFNLR1) in human lung. We hypothesized that the IFNλ signaling axis was active in human lung macrophages. In human alveolar macrophages (HAMs), we observed increased IFNLR1 expression and robust increase in interferon-stimulated gene (ISG) expression in response to IFNλ ligand. While human monocytes express minimal IFNLR1, differentiation of monocytes into macrophages with macrophage colony-stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) increased IFNLR1 mRNA, IFNLR1 protein expression, and cellular response to IFNλ ligation. Conversely, in mice, M-CSF or GM-CSF stimulated macrophages failed to produce ISGs in response to related ligands, IFNL2 or IFNL3, suggesting that IFNLR1 signaling in macrophages is species-specific. We next hypothesized that IFNλ signaling was critical in influenza antiviral responses. In primary human airway epithelial cells and precision-cut human lung slices, influenza infection substantially increased IFNλ levels. Pretreatment of both HAMs and differentiated human monocytes with IFNL1 significantly inhibited influenza infection. IFNLR1 knockout in the myeloid cell line, THP-1, exhibited reduced interferon responses to either direct or indirect exposure to influenza infection suggesting the indispensability of IFNLR1 for antiviral responses. These data demonstrate the presence of IFNλ - IFNLR1 signaling axis in human lung macrophages and a critical role of IFNλ signaling in combating influenza infection.

Highlights

  • Influenza type A and B virus infections in humans result in nearly 80,000 deaths in the United States and up to 650,000 deaths worldwide annually [1]

  • We found that interferon lambda receptor 1 (IFNLR1) was enriched in human alveolar macrophages (HAMs) which led to the production of interferonstimulated gene (ISG) in response to IFNl treatment

  • These findings indicate that IFNl is highly induced in the infected human lung environment

Read more

Summary

Introduction

Influenza type A and B virus infections in humans result in nearly 80,000 deaths in the United States and up to 650,000 deaths worldwide annually [1]. One crucial component is the innate immune response, whereby influenza viral RNA is detected by the pattern recognition receptor RIG-I, sequentially enacting MAVS oligomerization, transcriptional activation of IRF and NF-kB, and type I (e.g. IFNa, IFNb) and type III interferon (IFNl) production [6, 7]. While both IFNl and IFNa are antiviral, in some systems, IFNa exclusively leads to the upregulation of pro-inflammatory genes [5]. Cellular tropism of IFNLR1 may mediate this differential response, and it remains to be determined whether these results are reproducible in human studies [9]

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