Your browser doesn't support javascript.
loading
Adipogenic role of alternatively activated macrophages in ß-adrenergic remodeling of white adipose tissue.
Lee, Yun-Hee; Kim, Sang-Nam; Kwon, Hyun-Jung; Maddipati, Krishna Rao; Granneman, James G.
Affiliation
  • Lee YH; College of Pharmacy, Yonsei University, Incheon, South Korea;
  • Kim SN; College of Pharmacy, Yonsei University, Incheon, South Korea;
  • Kwon HJ; College of Pharmacy, Yonsei University, Incheon, South Korea;
  • Maddipati KR; Lipidomics Core Facility and Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan; and.
  • Granneman JG; Center for Integrative Metabolic and Endocrine Research, Wayne State University School of Medicine, Detroit, Michigan jgranne@med.wayne.edu.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R55-65, 2016 Jan 01.
Article in En | MEDLINE | ID: mdl-26538237
ABSTRACT
De novo brown adipogenesis involves the proliferation and differentiation of progenitors, yet the mechanisms that guide these events in vivo are poorly understood. We previously demonstrated that treatment with a ß3-adrenergic receptor (ADRB3) agonist triggers brown/beige adipogenesis in gonadal white adipose tissue following adipocyte death and clearance by tissue macrophages. The close physical relationship between adipocyte progenitors and tissue macrophages suggested that the macrophages that clear dying adipocytes might generate proadipogenic factors. Flow cytometric analysis of macrophages from mice treated with CL 316,243 identified a subpopulation that contained elevated lipid and expressed CD44. Lipidomic analysis of fluorescence-activated cell sorting-isolated macrophages demonstrated that CD44+ macrophages contained four- to five-fold higher levels of the endogenous peroxisome-proliferator activated receptor gamma (PPARγ) ligands 9-hydroxyoctadecadienoic acid (HODE), and 13-HODE compared with CD44- macrophages. Gene expression profiling and immunohistochemistry demonstrated that ADRB3 agonist treatment upregulated expression of ALOX15, the lipoxygenase responsible for generating 9-HODE and 13-HODE. Using an in vitro model of adipocyte efferocytosis, we found that IL-4-primed tissue macrophages accumulated lipid from dying fat cells and upregulated expression of Alox15. Furthermore, treatment of differentiating adipocytes with 9-HODE and 13-HODE potentiated brown/beige adipogenesis. Collectively, these data indicate that noninflammatory removal of adipocyte remnants and coordinated generation of PPARγ ligands by M2 macrophages provides localized adipogenic signals to support de novo brown/beige adipogenesis.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cell Communication / Dioxoles / Adipogenesis / Adipose Tissue, White / Adipocytes, White / Adrenergic beta-3 Receptor Agonists / Macrophage Activation / Macrophages Type of study: Prognostic_studies Language: En Journal: Am J Physiol Regul Integr Comp Physiol Journal subject: FISIOLOGIA Year: 2016 Type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cell Communication / Dioxoles / Adipogenesis / Adipose Tissue, White / Adipocytes, White / Adrenergic beta-3 Receptor Agonists / Macrophage Activation / Macrophages Type of study: Prognostic_studies Language: En Journal: Am J Physiol Regul Integr Comp Physiol Journal subject: FISIOLOGIA Year: 2016 Type: Article