IRF-8 regulates expansion of myeloid-derived suppressor cells and Foxp3+ regulatory T cells and modulates Th2 immune responses to gastrointestinal nematode infection

PLoS Pathog. 2017 Oct 2;13(10):e1006647. doi: 10.1371/journal.ppat.1006647. eCollection 2017 Oct.

Abstract

Interferon regulatory factor-8 (IRF-8) is critical for Th1 cell differentiation and negatively regulates myeloid cell development including myeloid-derived suppressor cells (MDSC). MDSC expand during infection with various pathogens including the gastrointestinal (GI) nematode Heligmosomoides polygyrus bakeri (Hpb). We investigated if IRF-8 contributes to Th2 immunity to Hpb infection. Irf8 expression was down-regulated in MDSC from Hpb-infected C57BL/6 (B6) mice. IRF-8 deficient Irf8-/- and BXH-2 mice had significantly higher adult worm burdens than B6 mice after primary or challenge Hpb infection. During primary infection, MDSC expanded to a significantly greater extent in mesenteric lymph nodes (MLN) and spleens of Irf8-/- and BXH-2 than B6 mice. CD4+GATA3+ T cells numbers were comparable in MLN of infected B6 and IRF-8 deficient mice, but MLN cells from infected IRF-8 deficient mice secreted significantly less parasite-specific IL-4 ex vivo. The numbers of alternatively activated macrophages in MLN and serum levels of Hpb-specific IgG1 and IgE were also significantly less in infected Irf8-/- than B6 mice. The frequencies of antigen-experienced CD4+CD11ahiCD49dhi cells that were CD44hiCD62L- were similar in MLN of infected Irf8-/- and B6 mice, but the proportions of CD4+GATA3+ and CD4+IL-4+ T cells were lower in infected Irf8-/- mice. CD11b+Gr1+ cells from naïve or infected Irf8-/- mice suppressed CD4+ T cell proliferation and parasite-specific IL-4 secretion in vitro albeit less efficiently than B6 mice. Surprisingly, there were significantly more CD4+ T cells in infected Irf8-/- mice, with a higher frequency of CD4+CD25+Foxp3+ T (Tregs) cells and significantly higher numbers of Tregs than B6 mice. In vivo depletion of MDSC and/or Tregs in Irf8-/- mice did not affect adult worm burdens, but Treg depletion resulted in higher egg production and enhanced parasite-specific IL-5, IL-13, and IL-6 secretion ex vivo. Our data thus provide a previously unrecognized role for IRF-8 in Th2 immunity to a GI nematode.

MeSH terms

  • Animals
  • Cells, Cultured
  • Forkhead Transcription Factors / genetics
  • Forkhead Transcription Factors / immunology
  • Gastrointestinal Diseases / immunology*
  • Interferon Regulatory Factors / drug effects
  • Interferon Regulatory Factors / genetics
  • Interferon Regulatory Factors / immunology*
  • Interleukin-4 / metabolism
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Myeloid-Derived Suppressor Cells / immunology*
  • Nematode Infections / immunology*
  • Nematospiroides dubius / immunology*
  • T-Lymphocytes, Regulatory / immunology
  • Th2 Cells / immunology*

Substances

  • Forkhead Transcription Factors
  • Foxp3 protein, mouse
  • Interferon Regulatory Factors
  • interferon regulatory factor-8
  • Interleukin-4

Grants and funding

This work was supported by grants from the Natural Sciences and Engineering Research Council of Canada (http://www.nserc-crsng.gc.ca/index_eng) (#342150 to MS, #326946 to TGG, and #238294 to AJ), National Institute of Health (https://www.niaid.nih.gov) RO1-AI035327-19 to PG, Intramural Program of the National Institute of Child Health and Development, National Institutes of Health (https://www.nichd.nih.gov) to KO, and the Canadian Institutes of Health Research (http://www.cihr-irsc.gc.ca/e) MOP-81169 and MOP-130369 to MMS. RMV was supported by a Studentship from the Research Institute of the McGill University Health Centre (http://www.rimuhc.ca/). The Centre for Host-Parasite Interactions is supported by Fonds de Québec de recherche sur la nature et les technologies (http://www.frqnt.gouv.qc.ca/en). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.