• Aucun résultat trouvé

Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells

N/A
N/A
Protected

Academic year: 2022

Partager "Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells"

Copied!
6
0
0

Texte intégral

(1)

Article

Reference

Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells

AROUS, Caroline, et al.

Abstract

Elevated levels of fasting insulin release and insufficient glucose-stimulated insulin secretion (GSIS) are hallmarks of diabetes. Studies have established cross-talk between integrin signaling and insulin activity, but more details of how integrin-dependent signaling impacts the pathophysiology of diabetes are needed. Here, we dissected integrin-dependent signaling pathways involved in the regulation of insulin secretion in β-cells and studied their link to the still debated autocrine regulation of insulin secretion by insulin/insulin-like growth factor (IGF) 2-AKT signaling. We observed for the first time a cooperation between different AKT isoforms and focal adhesion kinase (FAK)-dependent adhesion signaling, which either controlled GSIS or prevented insulin secretion under fasting conditions. Indeed, β-cells form integrin-containing adhesions, which provide anchorage to the pancreatic extracellular matrix and are the origin of intracellular signaling via FAK and paxillin. Under low-glucose conditions, β-cells adopt a starved adhesion phenotype consisting of actin stress fibers and large peripheral focal adhesion. [...]

AROUS, Caroline, et al. Integrin and autocrine IGF2 pathways control fasting insulin secretion in β-cells. Journal of Biological Chemistry, 2020, vol. 295, no. 49, p. 16510-16528

DOI : 10.1074/jbc.RA120.012957 PMID : 32934005

Available at:

http://archive-ouverte.unige.ch/unige:153888

Disclaimer: layout of this document may differ from the published version.

(2)

Integrin and autocrine IGF2-pathways control fasting insulin secretion in β-cells

Caroline Arous1, Maria Luisa Mizgier2, Katharina Rickenbach1, Michel Pinget2, Karim Bouzakri2* and Bernhard Wehrle-Haller1*

From the 1 Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Switzerland

2UMR DIATHEC, EA 7294, Centre Européen d’Etude du Diabète, Université de Strasbourg, France

Running title : Integrin and IGF2-pathways control fasting insulin secretion

(3)

S-1

Supp info 1: AG1024 did not induced cell death in low glucose condition. Rat primary β- cells were cultured in 2.8 mM glucose medium for 5h, containing none (CTL) or AG1024. After cell fixation, TUNEL assay were performed according to the commercial instruction.

(4)

S-2

Supp Info 2: Inhibition of PI3K activity induced remodeling in low glucose condition but blunted glucose-induced remodeling in MIN6B1 cell. MIN6B1 were cultured in 2.8 mM glucose for 4h in presence of LY294002 (10 µM) or not, and then stimulated or not for 20 min with 16.7 mM glucose. Cell were subsequently fixed and stained for actin (phalloidin in cyan) and paxillin (green). All images are fully representative of 3 independent experiments. Scale bar: 10µm.

(5)

S-3

Supp info 3: Dasatinib and saracatinib effect on phospho-c-Src and adhesion remodeling.

MIN6B1 cells were cultured in 2.8 mM glucose for 3h in presence or not (CTL) of dasatinib (dasat) or saracatinib (sara) and then stimulated or not with 16.7 mM glucose for 20 min in the continued presence of the inhibitors. Cell were subsequently fixed and stained for actin (phalloidin), paxillin (green) and phospho (Y418) c-Src (red). All images are fully representative of 3 independent experiments. Scale bar: 10µm.

(6)

S-4

Supp info 4: Effect of inhibitors on adhesion remodeling in rat primary β-cell. Rat primary β-cells were cultured in 2.8 mM glucose for 4h in presence or not (CTL) of inhibitors and then stimulated or not for 20 min with 16.7 mM glucose. PF562271 (PF56: 0.1 µM), Saracatinib (Sara:0.1 µM), ROCK inhibitor (Y27632: 50 µM), Rac1 inhibitor (Rac1: 0.2 µM) and Rho activator (RhoAc: 0.25 µg/ml). Cell were subsequently fixed and stained for actin (phalloidin) and paxillin (green). All images are fully representative of 3 independent experiments. Scale bar: 10µm.

Références

Documents relatifs

It was shown that EM66 is generated early during the ontogenesis of the fetal human adrenal gland [5] and that the peptide occurs in the secretory granules of adrenergic cells in

ATP content and release from isolated mouse or human islets or from INS-1 cells after transfection with mitochondrial targeted VDAC1 (mtVDAC1) or plasma membrane targeted

KO of IL-1Ra in b cells of normal and obese mice led to diminished glucose- stimulated insulin secretion in vivo and in vitro, to impaired glucose tolerance in obese mice, to

Our results show that the defec- tive glucose-induced insulin secretion observed after H 2 O 2 treatment correlates with altered mitochondrial activation seen as a loss of

SGLT2 is not expressed in pancreatic α- and β-cells, and its inhibition does not directly affect glucagon and insulin secretion in rodents and humans CHAE, Heeyoung, et al...

In contrast, under low-glucose conditions (fasting phase, starving conditions), IGF2 autocrine binding to the IGF1 receptor and downstream AKT2 activation led to the reinforcement of

Glucose stimulation leads to increased phorbol ester binding without causing membrane insertion of the enzyme which, however, occurs with receptor agonists.. It is suggested that

The present study reveals the critical role of the IP3R1/STIM1/Orai1 pathway in GPR40-mediated GIIS poten- tiation in pancreatic β -cells by showing that: (1) knockdown of IP3R1,