Macrophage-secreted factors promote a profibrotic phenotype in human preadipocytes

Mol Endocrinol. 2009 Jan;23(1):11-24. doi: 10.1210/me.2008-0183. Epub 2008 Oct 22.

Abstract

White adipose tissue (WAT) in obese humans is characterized by macrophage accumulation the effects of which on WAT biology are not fully understood. We previously demonstrated that macrophage-secreted factors impair preadipocyte differentiation and induce inflammation, and we described the excessive fibrotic deposition in WAT from obese individuals. Microarray analysis revealed significant overexpression of extracellular matrix (ECM) genes in inflammatory preadipocytes. We show here an organized deposition of fibronectin, collagen I, and tenascin-C and clustering of the ECM receptor alpha5 integrin, characterizing inflammatory preadipocytes. Anti-alpha5 integrin-neutralizing antibody decreased proliferation of these cells, underlining the importance of the fibronectin/integrin partnership. Fibronectin-cultured preadipocytes exhibited increased proliferation and expression of both nuclear factor-kappaB and cyclin D1. Small interfering RNA deletion of nuclear factor-kappaB and cyclin D1 showed that these factors link preadipocyte proliferation with inflammation and ECM remodeling. Macrophage-secreted molecules increased preadipocyte migration through an increase in active/phosphorylated focal adhesion kinase. Gene expression and neutralizing antibody experiments suggest that inhibin beta A, a TGF-beta family member, is a major fibrotic factor. Interactions between preadipocytes and macrophages were favored in a three-dimensional collagen I matrix mimicking the fibrotic context of WAT. Cell-rich regions were immunostained for preadipocytes, proliferation, and macrophages in the vicinity of fibrotic WAT from obese individuals. In conclusion, an inflammatory environment leads to profound modifications of the human preadipocyte phenotype, producing fibrotic components with increased migration and proliferation. This phenomenon might play a role in facilitating the constitution of quiescent preadipocyte pools and eventually in the maintenance and aggravation of increased fat mass in obesity.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adipocytes, White / cytology*
  • Adipocytes, White / physiology*
  • Base Sequence
  • Cell Adhesion
  • Cell Communication
  • Cell Differentiation
  • Cell Movement
  • Cell Proliferation
  • Cells, Cultured
  • Culture Media, Conditioned
  • Extracellular Matrix Proteins / genetics
  • Fibrosis
  • Gene Expression
  • Genes, bcl-1
  • Humans
  • Inflammation / genetics
  • Inflammation / pathology
  • Inflammation / physiopathology
  • Macrophages / metabolism*
  • Models, Biological
  • Obesity / pathology
  • Obesity / physiopathology
  • Phenotype
  • RNA, Small Interfering / genetics
  • Transcription Factor RelA / antagonists & inhibitors
  • Transcription Factor RelA / genetics

Substances

  • Culture Media, Conditioned
  • Extracellular Matrix Proteins
  • RELA protein, human
  • RNA, Small Interfering
  • Transcription Factor RelA