FoxO1 regulates allergic asthmatic inflammation through regulating polarization of the macrophage inflammatory phenotype

Oncotarget. 2016 Apr 5;7(14):17532-46. doi: 10.18632/oncotarget.8162.

Abstract

Inflammatory monocyte and tissue macrophages influence the initiation, progression, and resolution of type 2 immune responses, and alveolar macrophages are the most prevalent immune-effector cells in the lung. While we were characterizing the M1- or M2-like macrophages in type 2 allergic inflammation, we discovered that FoxO1 is highly expressed in alternatively activated macrophages. Although several studies have been focused on the fundamental role of FoxOs in hematopoietic and immune cells, the exact role that FoxO1 plays in allergic asthmatic inflammation in activated macrophages has not been investigated. Growing evidences indicate that FoxO1 acts as an upstream regulator of IRF4 and could have a role in a specific inflammatory phenotype of macrophages. Therefore, we hypothesized that IRF4 expression regulated by FoxO1 in alveolar macrophages is required for established type 2 immune mediates allergic lung inflammation. Our data indicate that targeted deletion of FoxO1 using FoxO1-selective inhibitor AS1842856 and genetic ablation of FoxO1 in macrophages significantly decreases IRF4 and various M2 macrophage-associated genes, suggesting a mechanism that involves FoxO1-IRF4 signaling in alveolar macrophages that works to polarize macrophages toward established type 2 immune responses. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, macrophage specific FoxO1 overexpression is associated with an accentuation of asthmatic lung inflammation, whereas pharmacologic inhibition of FoxO1 by AS1842856 attenuates the development of asthmatic lung inflammation. Thus, our study identifies a role for FoxO1-IRF4 signaling in the development of alternatively activated alveolar macrophages that contribute to type 2 allergic airway inflammation.

Keywords: FoxO1; Immune response; Immunity; Immunology and Microbiology Section; M2 macrophage phenotype; asthma; eosinophilic lung inflammation.

Publication types

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

MeSH terms

  • Animals
  • Asthma / immunology*
  • Cell Polarity / immunology
  • Forkhead Box Protein O1 / immunology*
  • Inflammation / immunology
  • Interferon Regulatory Factors / immunology
  • Macrophages, Alveolar / immunology*
  • Mice
  • Mice, Knockout
  • Phenotype

Substances

  • Forkhead Box Protein O1
  • Foxo1 protein, mouse
  • Interferon Regulatory Factors
  • interferon regulatory factor-4