Your browser doesn't support javascript.
loading
A STAT5-Smad3 dyad regulates adipogenic plasticity of visceral adipose mesenchymal stromal cells during chronic inflammation.
Das, Rahul; Giri, Jayeeta; K Paul, Pradyut; Froelich, Nicole; Chinnadurai, Raghavan; McCoy, Sara; Bushman, Wade; Galipeau, Jacques.
Affiliation
  • Das R; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • Giri J; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • K Paul P; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • Froelich N; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • Chinnadurai R; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • McCoy S; School of Medicine, Mercer University, Savannah, GA, 31404, USA.
  • Bushman W; Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
  • Galipeau J; Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
NPJ Regen Med ; 7(1): 41, 2022 Aug 31.
Article in En | MEDLINE | ID: mdl-36045134
ABSTRACT
Adipogenic differentiation of visceral adipose tissue-resident multipotent mesenchymal stromal cells (VA-MSC) into adipocytes is metabolically protective. Under chronic inflammatory stress, this neoadipogenesis process is suppressed by various pro-inflammatory cytokines and growth factors. However, the underlying mechanism(s) regulating VA-MSC plasticity remains largely unexplored. Using an adipogenic differentiation screen, we identified IFNγ and TGFß as key inhibitors of primary human VA-MSC differentiation. Further studies using human and mouse VA-MSCs and a chronic high-fat diet-fed murine model revealed that IFNγ/JAK2-activated STAT5 transcription factor is a central regulator of VA-MSC differentiation under chronic inflammatory conditions. Furthermore, our results indicate that under such conditions, IFNγ-activated STAT5 and TGFß-activated Smad3 physically interact via Smad4. This STAT5-Smad4-Smad3 complex plays a crucial role in preventing the early adipogenic commitment of VA-MSCs by suppressing key pro-adipogenic transcription factors, including CEBPδ, CEBPα, and PPARγ. Genetic or pharmacological disruption of IFNγ-TGFß synergy by inhibiting either STAT5 or Smad3 rescued adipogenesis under chronic inflammatory stress. Overall, our study delineates a central mechanism of MSC plasticity regulation by the convergence of multiple inflammatory signaling pathways.

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: NPJ Regen Med Year: 2022 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: NPJ Regen Med Year: 2022 Document type: Article Affiliation country: United States