Your browser doesn't support javascript.
loading
The signaling protein GIV/Girdin mediates the Nephrin-dependent insulin secretion of pancreatic islet ß cells in response to high glucose.
Wang, Hao; Yuan, Ying-Chao; Chang, Cong; Izumi, Tetsuro; Wang, Hong-Hui; Yang, Jin-Kui.
Afiliación
  • Wang H; Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China. Electronic address: hwang2002@126.com.
  • Yuan YC; Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
  • Chang C; College of Biology, Hunan University, Changsha, Hunan, China; Hunan Food and Drug Vocational College, Changsha, Hunan, China.
  • Izumi T; Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan.
  • Wang HH; College of Biology, Hunan University, Changsha, Hunan, China. Electronic address: wanghonghui@hnu.edu.cn.
  • Yang JK; Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China. Electronic address: jkyang@ccmu.edu.cn.
J Biol Chem ; 299(4): 103045, 2023 04.
Article en En | MEDLINE | ID: mdl-36822326
ABSTRACT
Glucose-stimulated insulin secretion of pancreatic ß cells is essential in maintaining glucose homeostasis. Recent evidence suggests that the Nephrin-mediated intercellular junction between ß cells is implicated in the regulation of insulin secretion. However, the underlying mechanisms are only partially characterized. Herein we report that GIV is a signaling mediator coordinating glucose-stimulated Nephrin phosphorylation and endocytosis with insulin secretion. We demonstrate that GIV is expressed in mouse islets and cultured ß cells. The loss of function study suggests that GIV is essential for the second phase of glucose-stimulated insulin secretion. Next, we demonstrate that GIV mediates the high glucose-stimulated tyrosine phosphorylation of GIV and Nephrin by recruiting Src kinase, which leads to the endocytosis of Nephrin. Subsequently, the glucose-induced GIV/Nephrin/Src signaling events trigger downstream Akt phosphorylation, which activates Rac1-mediated cytoskeleton reorganization, allowing insulin secretory granules to access the plasma membrane for the second-phase secretion. Finally, we found that GIV is downregulated in the islets isolated from diabetic mice, and rescue of GIV ameliorates the ß-cell dysfunction to restore the glucose-stimulated insulin secretion. We conclude that the GIV/Nephrin/Akt signaling axis is vital to regulate glucose-stimulated insulin secretion. This mechanism might be further targeted for therapeutic intervention of diabetic mellitus.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Islotes Pancreáticos / Diabetes Mellitus Experimental / Células Secretoras de Insulina Límite: Animals Idioma: En Revista: J Biol Chem Año: 2023 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Islotes Pancreáticos / Diabetes Mellitus Experimental / Células Secretoras de Insulina Límite: Animals Idioma: En Revista: J Biol Chem Año: 2023 Tipo del documento: Article