scholarly journals GABABreceptor activation inhibits exocytosis in rat pancreatic β-cells by G-protein-dependent activation of calcineurin

2004 ◽  
Vol 559 (2) ◽  
pp. 397-409 ◽  
Author(s):  
Matthias Braun ◽  
Anna Wendt ◽  
Karsten Buschard ◽  
Albert Salehi ◽  
Sabine Sewing ◽  
...  
2018 ◽  
Vol 10 (1) ◽  
pp. 84-93 ◽  
Author(s):  
Hirotaka Watada ◽  
Masanari Shiramoto ◽  
Shin Irie ◽  
Yasuo Terauchi ◽  
Yuichiro Yamada ◽  
...  

2002 ◽  
Vol 277 (41) ◽  
pp. 38373-38380 ◽  
Author(s):  
Jean-François Rolland ◽  
Jean-Claude Henquin ◽  
Patrick Gilon

2014 ◽  
Vol 34 (10) ◽  
pp. 2261-2267 ◽  
Author(s):  
Blake J. Cochran ◽  
Radjesh J. Bisoendial ◽  
Liming Hou ◽  
Elias N. Glaros ◽  
Jérémie Rossy ◽  
...  

2020 ◽  
Vol 11 (4) ◽  
pp. 814-822
Author(s):  
Haruhide Udagawa ◽  
Masaki Hiramoto ◽  
Miho Kawaguchi ◽  
Takashi Uebanso ◽  
Mica Ohara‐Imaizumi ◽  
...  

2005 ◽  
Vol 338 (3) ◽  
pp. 1455-1459 ◽  
Author(s):  
Vladimir V. Sharoyko ◽  
Irina I. Zaitseva ◽  
Mark Varsanyi ◽  
Neil Portwood ◽  
Barbara Leibiger ◽  
...  

2007 ◽  
Vol 293 (6) ◽  
pp. C1924-C1933 ◽  
Author(s):  
Leonid E. Fridlyand ◽  
Mark C. Harbeck ◽  
Michael W. Roe ◽  
Louis H. Philipson

In this report we describe a mathematical model for the regulation of cAMP dynamics in pancreatic β-cells. Incretin hormones such as glucagon-like peptide 1 (GLP-1) increase cAMP and augment insulin secretion in pancreatic β-cells. Imaging experiments performed in MIN6 insulinoma cells expressing a genetically encoded cAMP biosensor and loaded with fura-2, a calcium indicator, showed that cAMP oscillations are differentially regulated by periodic changes in membrane potential and GLP-1. We modeled the interplay of intracellular calcium (Ca2+) and its interaction with calmodulin, G protein-coupled receptor activation, adenylyl cyclases (AC), and phosphodiesterases (PDE). Simulations with the model demonstrate that cAMP oscillations are coupled to cytoplasmic Ca2+ oscillations in the β-cell. Slow Ca2+ oscillations (<1 min−1) produce low-frequency cAMP oscillations, and faster Ca2+ oscillations (>3–4 min−1) entrain high-frequency, low-amplitude cAMP oscillations. The model predicts that GLP-1 receptor agonists induce cAMP oscillations in phase with cytoplasmic Ca2+ oscillations. In contrast, observed antiphasic Ca2+ and cAMP oscillations can be simulated following combined glucose and tetraethylammonium-induced changes in membrane potential. The model provides additional evidence for a pivotal role for Ca2+-dependent AC and PDE activation in coupling of Ca2+ and cAMP signals. Our results reveal important differences in the effects of glucose/TEA and GLP-1 on cAMP dynamics in MIN6 β-cells.


2018 ◽  
Vol 293 (47) ◽  
pp. 18086-18098 ◽  
Author(s):  
Rajesh Gupta ◽  
Dan C. Nguyen ◽  
Michael D. Schaid ◽  
Xia Lei ◽  
Appakalai N. Balamurugan ◽  
...  

Secreted proteins are important metabolic regulators in both healthy and disease states. Here, we sought to investigate the mechanism by which the secreted protein complement 1q-like-3 (C1ql3) regulates insulin secretion from pancreatic β-cells, a key process affecting whole-body glucose metabolism. We found that C1ql3 predominantly inhibits exendin-4– and cAMP-stimulated insulin secretion from mouse and human islets. However, to a lesser extent, C1ql3 also reduced insulin secretion in response to KCl, the potassium channel blocker tolbutamide, and high glucose. Strikingly, C1ql3 did not affect insulin secretion stimulated by fatty acids, amino acids, or mitochondrial metabolites, either at low or submaximal glucose concentrations. Additionally, C1ql3 inhibited glucose-stimulated cAMP levels, and insulin secretion stimulated by exchange protein directly activated by cAMP-2 and protein kinase A. These results suggest that C1ql3 inhibits insulin secretion primarily by regulating cAMP signaling. The cell adhesion G protein–coupled receptor, brain angiogenesis inhibitor-3 (BAI3), is a C1ql3 receptor and is expressed in β-cells and in mouse and human islets, but its function in β-cells remained unknown. We found that siRNA-mediated Bai3 knockdown in INS1(832/13) cells increased glucose-stimulated insulin secretion. Furthermore, incubating the soluble C1ql3-binding fragment of the BAI3 protein completely blocked the inhibitory effects of C1ql3 on insulin secretion in response to cAMP. This suggests that BAI3 mediates the inhibitory effects of C1ql3 on insulin secretion from pancreatic β-cells. These findings demonstrate a novel regulatory mechanism by which C1ql3/BAI3 signaling causes an impairment of insulin secretion from β-cells, possibly contributing to the progression of type 2 diabetes in obesity.


Sign in / Sign up

Export Citation Format

Share Document