Your browser doesn't support javascript.
loading
Hematopoietic-to-mesenchymal transition of adipose tissue macrophages is regulated by integrin ß1 and fabricated fibrin matrices.
Gavin, Kathleen M; Majka, Susan M; Kohrt, Wendy M; Miller, Heidi L; Sullivan, Timothy M; Klemm, Dwight J.
Afiliación
  • Gavin KM; a Geriatric Research, Education and Clinical Center , VA Eastern Colorado Health Care System , Denver , CO.
  • Majka SM; b Division of Geriatric Medicine, Department of Medicine , University of Colorado Anschutz Medical Center , Aurora , CO.
  • Kohrt WM; c Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine , Vanderbilt University School of Medicine , Nashville , TN.
  • Miller HL; a Geriatric Research, Education and Clinical Center , VA Eastern Colorado Health Care System , Denver , CO.
  • Sullivan TM; b Division of Geriatric Medicine, Department of Medicine , University of Colorado Anschutz Medical Center , Aurora , CO.
  • Klemm DJ; d Division of Pulmonary and Critical Care Medicine , University of Colorado Anschutz Medical Center , Aurora , CO.
Adipocyte ; 6(3): 234-249, 2017 07 03.
Article en En | MEDLINE | ID: mdl-28441086
ABSTRACT
Some bona fide adult adipocytes arise de novo from a bone marrow-derived myeloid lineage. These studies further demonstrate that adipose tissue stroma contains a resident population of myeloid cells capable of adipocyte and multilineage mesenchymal differentiation. These resident myeloid cells lack hematopoietic markers and express mesenchymal and progenitor cell markers. Because bone marrow mesenchymal progenitor cells have not been shown to enter the circulation, we hypothesized that myeloid cells acquire mesenchymal differentiation capacity in adipose tissue. We fabricated a 3-dimensional fibrin matrix culture system to define the adipose differentiation potential of adipose tissue-resident myeloid subpopulations, including macrophages, granulocytes and dendritic cells. Our data show that multilineage mesenchymal potential was limited to adipose tissue macrophages, characterized by the acquisition of adipocyte, osteoblast, chondrocyte and skeletal muscle myocyte phenotypes. Fibrin hydrogel matrices stimulated macrophage loss of hematopoietic cell lineage determinants and the expression of mesenchymal and progenitor cell markers, including integrin ß1. Ablation of integrin ß1 in macrophages inhibited adipocyte specification. Therefore, some bona fide adipocytes are specifically derived from adipose tissue-resident macrophages via an integrin ß1-dependent hematopoietic-to-mesenchymal transition, whereby they become capable of multipotent mesenchymal differentiation. The requirement for integrin ß1 highlights this molecule as a potential target for controlling the production of marrow-derived adipocytes and their contribution to adipose tissue development and function.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Integrina beta1 / Células Madre Mesenquimatosas Límite: Animals / Humans Idioma: En Revista: Adipocyte Año: 2017 Tipo del documento: Article País de afiliación: Colombia

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Integrina beta1 / Células Madre Mesenquimatosas Límite: Animals / Humans Idioma: En Revista: Adipocyte Año: 2017 Tipo del documento: Article País de afiliación: Colombia