Adventitial fibroblasts induce a distinct proinflammatory/profibrotic macrophage phenotype in pulmonary hypertension

J Immunol. 2014 Jul 15;193(2):597-609. doi: 10.4049/jimmunol.1303048. Epub 2014 Jun 13.

Abstract

Macrophage accumulation is not only a characteristic hallmark but is also a critical component of pulmonary artery remodeling associated with pulmonary hypertension (PH). However, the cellular and molecular mechanisms that drive vascular macrophage activation and their functional phenotype remain poorly defined. Using multiple levels of in vivo (bovine and rat models of hypoxia-induced PH, together with human tissue samples) and in vitro (primary mouse, rat, and bovine macrophages, human monocytes, and primary human and bovine fibroblasts) approaches, we observed that adventitial fibroblasts derived from hypertensive pulmonary arteries (bovine and human) regulate macrophage activation. These fibroblasts activate macrophages through paracrine IL-6 and STAT3, HIF1, and C/EBPβ signaling to drive expression of genes previously implicated in chronic inflammation, tissue remodeling, and PH. This distinct fibroblast-activated macrophage phenotype was independent of IL-4/IL-13-STAT6 and TLR-MyD88 signaling. We found that genetic STAT3 haplodeficiency in macrophages attenuated macrophage activation, complete STAT3 deficiency increased macrophage activation through compensatory upregulation of STAT1 signaling, and deficiency in C/EBPβ or HIF1 attenuated fibroblast-driven macrophage activation. These findings challenge the current paradigm of IL-4/IL-13-STAT6-mediated alternative macrophage activation as the sole driver of vascular remodeling in PH, and uncover a cross-talk between adventitial fibroblasts and macrophages in which paracrine IL-6-activated STAT3, HIF1α, and C/EBPβ signaling are critical for macrophage activation and polarization. Thus, targeting IL-6 signaling in macrophages by completely inhibiting C/EBPβ or HIF1α or by partially inhibiting STAT3 may hold therapeutic value for treatment of PH and other inflammatory conditions characterized by increased IL-6 and absent IL-4/IL-13 signaling.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Animals, Newborn
  • CCAAT-Enhancer-Binding Protein-beta / genetics
  • CCAAT-Enhancer-Binding Protein-beta / immunology
  • CCAAT-Enhancer-Binding Protein-beta / metabolism
  • Cattle
  • Cell Line, Tumor
  • Cells, Cultured
  • Culture Media, Conditioned / metabolism
  • Culture Media, Conditioned / pharmacology
  • Fibroblasts / immunology*
  • Fibroblasts / metabolism
  • Fibrosis / genetics
  • Fibrosis / immunology
  • Fibrosis / metabolism
  • Gene Expression / drug effects
  • Gene Expression / genetics
  • Gene Expression / immunology
  • Humans
  • Hypertension, Pulmonary / genetics
  • Hypertension, Pulmonary / immunology*
  • Hypertension, Pulmonary / metabolism
  • Hypoxia-Inducible Factor 1, alpha Subunit / genetics
  • Hypoxia-Inducible Factor 1, alpha Subunit / immunology
  • Hypoxia-Inducible Factor 1, alpha Subunit / metabolism
  • Immunoblotting
  • Inflammation / genetics
  • Inflammation / immunology
  • Inflammation / metabolism
  • Interleukin-6 / metabolism
  • Interleukin-6 / pharmacology
  • Macrophage Activation / drug effects
  • Macrophage Activation / genetics
  • Macrophage Activation / immunology*
  • Macrophages / immunology*
  • Macrophages / metabolism
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Phenotype
  • Rats, Inbred WKY
  • Reverse Transcriptase Polymerase Chain Reaction
  • STAT3 Transcription Factor / genetics
  • STAT3 Transcription Factor / immunology
  • STAT3 Transcription Factor / metabolism

Substances

  • CCAAT-Enhancer-Binding Protein-beta
  • Culture Media, Conditioned
  • Hypoxia-Inducible Factor 1, alpha Subunit
  • Interleukin-6
  • STAT3 Transcription Factor