Your browser doesn't support javascript.
loading
Adipocyte-specific FAK deletion promotes pancreatic ß-cell apoptosis via adipose inflammatory response to exacerbate diabetes mellitus.
Ding, Fei; Zheng, Peng; Fang, Hong-Ting; Luo, Yuan-Yuan; Yan, Xi-Yue; Chen, Hui-Jian; Yan, You-E.
Affiliation
  • Ding F; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Zheng P; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Fang HT; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Luo YY; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Yan XY; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Chen HJ; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
  • Yan YE; Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China.
Clin Transl Med ; 14(7): e1742, 2024 Jul.
Article in En | MEDLINE | ID: mdl-38925910
ABSTRACT

BACKGROUND:

White adipose tissue (WAT) has a key role in maintaining energy balance throughout the body, and their dysfunction take part in the regulation of diabetes mellitus. However, the internal regulatory mechanisms underlying are still unknown. METHODS AND

RESULTS:

We generated adipocyte-specific FAK KO (FAK-AKO) mice and investigated their phenotype. The cascade of adipocyte, macrophage in adipocyte tissues, and pancreatic ß-cells were proposed in FAK-AKO mice and validated by cell line studies using 3T3-L1, Raw264.7 and Min6. The FAK-AKO mice exhibited glucose intolerance, reduced adipose tissue mass and increased apoptosis, lipolysis and inflammatory response in adipose tissue. We further demonstrate that adipocyte FAK deletion increases ß cell apoptosis and inflammatory infiltrates into islets, which is potentiated if mice were treated with STZ. In the STZ-induced diabetes model, FAK AKO mice exhibit less serum insulin content and pancreatic ß cell area. Moreover, serum pro-inflammatory factors increased and insulin levels decreased after glucose stimulation in FAK AKO mice. In a parallel vitro experiment, knockdown or inhibition of FAK during differentiation also increased apoptosis, lipolysis and inflammatory in 3T3-L1 adipocytes, whereas the opposite was observed upon overexpression of FAK. Moreover, coculturing LPS-treated RAW264.7 macrophages with knockdown FAK of 3T3-L1 adipocytes increased macrophage pro-inflammatory response. Furthermore, conditioned medium from above stimulated Min6 cells apoptosis (with or without STZ), whereas the opposite was observed upon overexpression of FAK. Mechanistically, FAK protein interact with TRAF6 in adipocytes and knockdown or inhibition of FAK activated TRAF6/TAK1/NF-κB signaling, which exacerbates inflammation of adipocytes themselves.

CONCLUSION:

Adipocyte FAK deletion promotes both adipocyte apoptosis and adipose tissue inflammation. Pro-inflammatory factors released by the FAK-null adipose tissue further trigger apoptosis in pancreatic islets induced by the administration of STZ, thereby exacerbating the diabetes mellitus. This study reveals a link between FAK-mediated adipose inflammation and diabetes mellitus, a mechanism that has not been previously recognized.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Apoptosis / Mice, Knockout / Adipocytes / Diabetes Mellitus, Experimental / Insulin-Secreting Cells / Focal Adhesion Kinase 1 Limits: Animals Language: En Journal: Clin Transl Med Year: 2024 Document type: Article Affiliation country: China

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Apoptosis / Mice, Knockout / Adipocytes / Diabetes Mellitus, Experimental / Insulin-Secreting Cells / Focal Adhesion Kinase 1 Limits: Animals Language: En Journal: Clin Transl Med Year: 2024 Document type: Article Affiliation country: China