Upregulation of SOCS3 in lung CD4+ T cells in a mouse model of chronic PA lung infection and suppression of Th17‑mediated neutrophil recruitment in exogenous SOCS3 transfer in vitro

Mol Med Rep. 2017 Jul;16(1):778-786. doi: 10.3892/mmr.2017.6630. Epub 2017 May 25.

Abstract

Neutrophilic airway inflammation in chronic lung infections caused by Pseudomonas aeruginosa (PA) is associated with T helper (Th)17 responses. Suppressor of cytokine signaling 3 (SOCS3) is the major negative modulator of Th17 function through the suppression of signal transducer and activator of transcription (STAT)3 activation. The aim of the present study was to investigate the expression of SOCS3 in lung CD4+ T cells in a mouse model of chronic PA lung infection and the effect of exogenous SOCS3 on Th17‑mediated neutrophil recruitment in vitro. A mouse model of chronic PA lung infection was established and the activation of STAT3 and Th17 response in lung tissues and lung CD4+ T cells was assessed. The protein and mRNA expression of SOCS3 in lung CD4+ T cells was analyzed by western blotting and reverse transcription‑quantitative polymerase chain reaction. The authors constructed a recombinant lentivirus carrying the SOCS3 gene and transferred it into lung CD4+ T cells isolated from a mouse model. These transfected cells were stimulated with interleukin (IL)‑23 in vitro and the protein level of p‑STAT3 and retinoid‑related orphan receptor (ROR)γt was determined by western blotting. The expression of IL‑17A+ cells was analyzed by flow cytometry and the level of IL‑17A in cell culture supernatant was measured by ELISA. The mouse lung epithelial cell line, MLE‑12, was cocultured with lung CD4+ T cells that overexpressed the SOCS3 gene and the culture supernatant was harvested and used for a chemotaxis assay. Compared with control mice, mice with chronic PA lung infection had significantly higher level of p‑STAT3 and Th17 response in both lung tissues and lung CD4+ T cells. The protein and mRNA level of SOCS3 in lung CD4+ T cells increased as the chronic PA lung infection developed. Exogenous SOCS3 gene transfer in PA‑infected lung CD4+ T cells decreased p‑STAT3 and RORγt expression and suppressed the level of IL‑17A+ cells in vitro. MLE‑12 cells cocultured with SOCS3‑overexpressing lung CD4+ T cells expressed a significantly lower level of neutrophil chemoattractants chemokine (C‑X‑C motif) ligand (CXCL) 1 and CXCL5, and recruited significantly smaller numbers of migrating neutrophils than those cocultured with control cells. SOCS3 was upregulated in lung CD4+ T cells following the activation of STAT3/Th17 axis in a mouse model of chronic PA lung infection. Exogenous SOCS3 transfer in PA‑infected lung CD4+ T cells suppresses Th17‑mediated neutrophil recruitment in vitro.

MeSH terms

  • Animals
  • CD4-Positive T-Lymphocytes / immunology*
  • CD4-Positive T-Lymphocytes / metabolism*
  • Chronic Disease
  • Disease Models, Animal
  • Female
  • Gene Expression Regulation*
  • Immunohistochemistry
  • Interleukin-17 / metabolism
  • Mice
  • Neutrophil Infiltration / genetics
  • Neutrophil Infiltration / immunology*
  • Nuclear Receptor Subfamily 1, Group F, Member 3 / genetics
  • Nuclear Receptor Subfamily 1, Group F, Member 3 / metabolism
  • Pneumonia / genetics
  • Pneumonia / immunology
  • Pneumonia / metabolism
  • Pneumonia / pathology
  • Pseudomonas Infections / genetics
  • Pseudomonas Infections / immunology
  • Pseudomonas Infections / metabolism
  • Pseudomonas Infections / pathology
  • STAT3 Transcription Factor / metabolism
  • Signal Transduction
  • Suppressor of Cytokine Signaling 3 Protein / genetics*
  • Th17 Cells / immunology*
  • Th17 Cells / metabolism*
  • Transduction, Genetic

Substances

  • Interleukin-17
  • Nuclear Receptor Subfamily 1, Group F, Member 3
  • STAT3 Transcription Factor
  • Suppressor of Cytokine Signaling 3 Protein