Your browser doesn't support javascript.
loading
Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation.
Amano, Shinya U; Cohen, Jessica L; Vangala, Pranitha; Tencerova, Michaela; Nicoloro, Sarah M; Yawe, Joseph C; Shen, Yuefei; Czech, Michael P; Aouadi, Myriam.
Afiliación
  • Amano SU; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Cohen JL; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Vangala P; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Tencerova M; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Nicoloro SM; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Yawe JC; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Shen Y; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Czech MP; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
  • Aouadi M; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.
Cell Metab ; 19(1): 162-171, 2014 Jan 07.
Article en En | MEDLINE | ID: mdl-24374218
ABSTRACT
Adipose tissue (AT) of obese mice and humans accumulates immune cells, which secrete cytokines that can promote insulin resistance. AT macrophages (ATMs) are thought to originate from bone-marrow-derived monocytes, which infiltrate the tissue from the circulation. Here, we show that a major fraction of macrophages unexpectedly undergo cell division locally within AT, as detected by Ki67 expression and 5-ethynyl-2'-deoxyuridine incorporation. Macrophages within the visceral AT (VAT), but not those in other tissues (including liver and spleen), displayed increased proliferation in obesity. Importantly, depletion of blood monocytes had no impact on ATM content, whereas their proliferation in situ continued. Treatment with monocyte chemotactic protein 1 (MCP-1) induced macrophage cell division in AT explants, whereas mcp-1 deficiency in vivo decreased ATM proliferation. These results reveal that, in addition to blood monocyte recruitment, in situ proliferation driven by MCP-1 is an important process by which macrophages accumulate in the VAT in obesity.
Asunto(s)

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Tejido Adiposo / Inflamación / Macrófagos / Obesidad Tipo de estudio: Risk_factors_studies Límite: Animals / Humans / Male Idioma: En Revista: Cell Metab Asunto de la revista: METABOLISMO Año: 2014 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Tejido Adiposo / Inflamación / Macrófagos / Obesidad Tipo de estudio: Risk_factors_studies Límite: Animals / Humans / Male Idioma: En Revista: Cell Metab Asunto de la revista: METABOLISMO Año: 2014 Tipo del documento: Article País de afiliación: Estados Unidos