Skip to main content
Top
Published in: Diabetologia 3/2014

01-03-2014 | Article

β-Arrestin2 plays a key role in the modulation of the pancreatic beta cell mass in mice

Authors: Magalie A. Ravier, Michele Leduc, Joy Richard, Nathalie Linck, Annie Varrault, Nelly Pirot, Morgane M. Roussel, Joël Bockaert, Stéphane Dalle, Gyslaine Bertrand

Published in: Diabetologia | Issue 3/2014

Login to get access

Abstract

Aims/hypothesis

Beta cell failure due to progressive secretory dysfunction and limited expansion of beta cell mass is a key feature of type 2 diabetes. Beta cell function and mass are controlled by glucose and hormones/neurotransmitters that activate G protein-coupled receptors or receptor tyrosine kinases. We have investigated the role of β-arrestin (ARRB)2, a scaffold protein known to modulate such receptor signalling, in the modulation of beta cell function and mass, with a specific interest in glucagon-like peptide-1 (GLP-1), muscarinic and insulin receptors.

Methods

β-arrestin2-knockout mice and their wild-type littermates were fed a normal or a high-fat diet (HFD). Glucose tolerance, insulin sensitivity and insulin secretion were assessed in vivo. Beta cell mass was evaluated in pancreatic sections. Free cytosolic [Ca2+] and insulin secretion were determined using perifused islets. The insulin signalling pathway was evaluated by western blotting.

Results

Arrb2-knockout mice exhibited impaired glucose tolerance and insulin secretion in vivo, but normal insulin sensitivity compared with wild type. Surprisingly, the absence of ARRB2 did not affect glucose-stimulated insulin secretion or GLP-1- and acetylcholine-mediated amplifications from perifused islets, but it decreased the islet insulin content and beta cell mass. Additionally, there was no compensatory beta cell mass expansion through proliferation in response to the HFD. Furthermore, Arrb2 deletion altered the islet insulin signalling pathway.

Conclusions/interpretation

ARRB2 is unlikely to be involved in the regulation of insulin secretion, but it is required for beta cell mass plasticity. Additionally, we provide new insights into the mechanisms involved in insulin signalling in beta cells.
Appendix
Available only for authorised users
Literature
1.
go back to reference Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110PubMedCrossRef Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110PubMedCrossRef
3.
4.
go back to reference Ahren B (2009) Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov 8:369–385PubMedCrossRef Ahren B (2009) Islet G protein-coupled receptors as potential targets for treatment of type 2 diabetes. Nat Rev Drug Discov 8:369–385PubMedCrossRef
5.
go back to reference Goldfine AB, Kulkarni RN (2012) Modulation of beta-cell function: a translational journey from the bench to the bedside. Diabetes Obes Metab 14(Suppl 3):152–160PubMedCrossRef Goldfine AB, Kulkarni RN (2012) Modulation of beta-cell function: a translational journey from the bench to the bedside. Diabetes Obes Metab 14(Suppl 3):152–160PubMedCrossRef
6.
go back to reference Gilon P, Henquin JC (2001) Mechanisms and physiological significance of the cholinergic control of pancreatic beta-cell function. Endocr Rev 22:565–604PubMed Gilon P, Henquin JC (2001) Mechanisms and physiological significance of the cholinergic control of pancreatic beta-cell function. Endocr Rev 22:565–604PubMed
7.
go back to reference Thorens B (2011) Brain glucose sensing and neural regulation of insulin and glucagon secretion. Diabetes Obes Metab 13(Suppl 1):82–88PubMedCrossRef Thorens B (2011) Brain glucose sensing and neural regulation of insulin and glucagon secretion. Diabetes Obes Metab 13(Suppl 1):82–88PubMedCrossRef
8.
go back to reference Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17:819–837PubMedCrossRef Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17:819–837PubMedCrossRef
9.
go back to reference Gautam D, Han SJ, Duttaroy A et al (2007) Role of the M3 muscarinic acetylcholine receptor in beta-cell function and glucose homeostasis. Diabetes Obes Metab 9(Suppl 2):158–169PubMedCrossRef Gautam D, Han SJ, Duttaroy A et al (2007) Role of the M3 muscarinic acetylcholine receptor in beta-cell function and glucose homeostasis. Diabetes Obes Metab 9(Suppl 2):158–169PubMedCrossRef
10.
go back to reference Leibiger IB, Leibiger B, Berggren PO (2008) Insulin signaling in the pancreatic beta-cell. Annu Rev Nutr 28:233–251PubMedCrossRef Leibiger IB, Leibiger B, Berggren PO (2008) Insulin signaling in the pancreatic beta-cell. Annu Rev Nutr 28:233–251PubMedCrossRef
12.
go back to reference Braun M, Ramracheya R, Rorsman P (2012) Autocrine regulation of insulin secretion. Diabetes Obes Metab 14(Suppl 3):143–151PubMedCrossRef Braun M, Ramracheya R, Rorsman P (2012) Autocrine regulation of insulin secretion. Diabetes Obes Metab 14(Suppl 3):143–151PubMedCrossRef
13.
go back to reference Elghazi L, Bernal-Mizrachi E (2009) Akt and PTEN: beta-cell mass and pancreas plasticity. Trends Endocrinol Metab 20:243–251PubMedCrossRef Elghazi L, Bernal-Mizrachi E (2009) Akt and PTEN: beta-cell mass and pancreas plasticity. Trends Endocrinol Metab 20:243–251PubMedCrossRef
14.
go back to reference Kovacs JJ, Hara MR, Davenport CL, Kim J, Lefkowitz RJ (2009) Arrestin development: emerging roles for beta-arrestins in developmental signaling pathways. Dev Cell 17:443–458PubMedCentralPubMedCrossRef Kovacs JJ, Hara MR, Davenport CL, Kim J, Lefkowitz RJ (2009) Arrestin development: emerging roles for beta-arrestins in developmental signaling pathways. Dev Cell 17:443–458PubMedCentralPubMedCrossRef
15.
go back to reference Lefkowitz RJ, Shenoy SK (2005) Transduction of receptor signals by beta-arrestins. Science 308:512–517PubMedCrossRef Lefkowitz RJ, Shenoy SK (2005) Transduction of receptor signals by beta-arrestins. Science 308:512–517PubMedCrossRef
16.
go back to reference Luttrell LM, Gesty-Palmer D (2010) Beyond desensitization: physiological relevance of arrestin-dependent signaling. Pharmacol Rev 62:305–330PubMedCrossRef Luttrell LM, Gesty-Palmer D (2010) Beyond desensitization: physiological relevance of arrestin-dependent signaling. Pharmacol Rev 62:305–330PubMedCrossRef
17.
go back to reference DeWire SM, Ahn S, Lefkowitz RJ, Shenoy SK (2007) Beta-arrestins and cell signaling. Annu Rev Physiol 69:483–510PubMedCrossRef DeWire SM, Ahn S, Lefkowitz RJ, Shenoy SK (2007) Beta-arrestins and cell signaling. Annu Rev Physiol 69:483–510PubMedCrossRef
18.
19.
go back to reference DeFea KA (2011) Beta-arrestins as regulators of signal termination and transduction: how do they determine what to scaffold? Cell Signal 23:621–629PubMedCrossRef DeFea KA (2011) Beta-arrestins as regulators of signal termination and transduction: how do they determine what to scaffold? Cell Signal 23:621–629PubMedCrossRef
20.
go back to reference Hupfeld CJ, Olefsky JM (2007) Regulation of receptor tyrosine kinase signaling by GRKs and beta-arrestins. Annu Rev Physiol 69:561–577PubMedCrossRef Hupfeld CJ, Olefsky JM (2007) Regulation of receptor tyrosine kinase signaling by GRKs and beta-arrestins. Annu Rev Physiol 69:561–577PubMedCrossRef
21.
go back to reference Luan B, Zhao J, Wu H et al (2009) Deficiency of a beta-arrestin-2 signal complex contributes to insulin resistance. Nature 457:1146–1149PubMedCrossRef Luan B, Zhao J, Wu H et al (2009) Deficiency of a beta-arrestin-2 signal complex contributes to insulin resistance. Nature 457:1146–1149PubMedCrossRef
22.
go back to reference Quoyer J, Longuet C, Broca C et al (2010) GLP-1 mediates antiapoptotic effect by phosphorylating Bad through a beta-arrestin 1-mediated ERK1/2 activation in pancreatic beta-cells. J Biol Chem 285:1989–2002PubMedCrossRef Quoyer J, Longuet C, Broca C et al (2010) GLP-1 mediates antiapoptotic effect by phosphorylating Bad through a beta-arrestin 1-mediated ERK1/2 activation in pancreatic beta-cells. J Biol Chem 285:1989–2002PubMedCrossRef
23.
go back to reference Sonoda N, Imamura T, Yoshizaki T, Babendure JL, Lu JC, Olefsky JM (2008) Beta-Arrestin-1 mediates glucagon-like peptide-1 signaling to insulin secretion in cultured pancreatic beta cells. Proc Natl Acad Sci U S A 105:6614–6619PubMedCentralPubMedCrossRef Sonoda N, Imamura T, Yoshizaki T, Babendure JL, Lu JC, Olefsky JM (2008) Beta-Arrestin-1 mediates glucagon-like peptide-1 signaling to insulin secretion in cultured pancreatic beta cells. Proc Natl Acad Sci U S A 105:6614–6619PubMedCentralPubMedCrossRef
24.
go back to reference Talbot J, Joly E, Prentki M, Buteau J (2012) beta-Arrestin1 mediated recruitment of c-Src underlies the proliferative action of glucagon-like peptide-1 in pancreatic beta INS832/13 cells. Mol Cell Endocrinol 354:65–70CrossRef Talbot J, Joly E, Prentki M, Buteau J (2012) beta-Arrestin1 mediated recruitment of c-Src underlies the proliferative action of glucagon-like peptide-1 in pancreatic beta INS832/13 cells. Mol Cell Endocrinol 354:65–70CrossRef
25.
go back to reference Broca C, Quoyer J, Costes S et al (2009) beta-Arrestin 1 is required for PAC1 receptor-mediated potentiation of long-lasting ERK1/2 activation by glucose in pancreatic beta-cells. J Biol Chem 284:4332–4342PubMedCrossRef Broca C, Quoyer J, Costes S et al (2009) beta-Arrestin 1 is required for PAC1 receptor-mediated potentiation of long-lasting ERK1/2 activation by glucose in pancreatic beta-cells. J Biol Chem 284:4332–4342PubMedCrossRef
26.
go back to reference Kong KC, Butcher AJ, McWilliams P et al (2010) M3-muscarinic receptor promotes insulin release via receptor phosphorylation/arrestin-dependent activation of protein kinase D1. Proc Natl Acad Sci U S A 107:21181–21186PubMedCentralPubMedCrossRef Kong KC, Butcher AJ, McWilliams P et al (2010) M3-muscarinic receptor promotes insulin release via receptor phosphorylation/arrestin-dependent activation of protein kinase D1. Proc Natl Acad Sci U S A 107:21181–21186PubMedCentralPubMedCrossRef
27.
go back to reference Dalle S, Ravier MA, Bertrand G (2011) Emerging roles for beta-arrestin-1 in the control of the pancreatic beta-cell function and mass: new therapeutic strategies and consequences for drug screening. Cell Signal 23:522–528PubMedCrossRef Dalle S, Ravier MA, Bertrand G (2011) Emerging roles for beta-arrestin-1 in the control of the pancreatic beta-cell function and mass: new therapeutic strategies and consequences for drug screening. Cell Signal 23:522–528PubMedCrossRef
28.
go back to reference Zhang M, Zhu Y, Mu K et al (2013) Loss of beta-arrestin2 mediates pancreatic-islet dysfunction in mice. Biochem Biophys Res Commun 435:345–349PubMedCrossRef Zhang M, Zhu Y, Mu K et al (2013) Loss of beta-arrestin2 mediates pancreatic-islet dysfunction in mice. Biochem Biophys Res Commun 435:345–349PubMedCrossRef
29.
go back to reference Bertrand G, Ishiyama N, Nenquin M, Ravier MA, Henquin JC (2002) The elevation of glutamate content and the amplification of insulin secretion in glucose-stimulated pancreatic islets are not causally related. J Biol Chem 277:32883–32891PubMedCrossRef Bertrand G, Ishiyama N, Nenquin M, Ravier MA, Henquin JC (2002) The elevation of glutamate content and the amplification of insulin secretion in glucose-stimulated pancreatic islets are not causally related. J Biol Chem 277:32883–32891PubMedCrossRef
30.
go back to reference Kahn SE, Hull RL, Utzschneider KM (2006) Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444:840–846PubMedCrossRef Kahn SE, Hull RL, Utzschneider KM (2006) Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444:840–846PubMedCrossRef
31.
go back to reference Sachdeva MM, Stoffers DA (2009) Minireview: meeting the demand for insulin: molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol Endocrinol 23:747–758PubMedCrossRef Sachdeva MM, Stoffers DA (2009) Minireview: meeting the demand for insulin: molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol Endocrinol 23:747–758PubMedCrossRef
32.
go back to reference Syme CA, Zhang L, Bisello A (2006) Caveolin-1 regulates cellular trafficking and function of the glucagon-like peptide 1 receptor. Mol Endocrinol 20:3400–3411PubMedCrossRef Syme CA, Zhang L, Bisello A (2006) Caveolin-1 regulates cellular trafficking and function of the glucagon-like peptide 1 receptor. Mol Endocrinol 20:3400–3411PubMedCrossRef
33.
go back to reference Jorgensen R, Martini L, Schwartz TW, Elling CE (2005) Characterization of glucagon-like peptide-1 receptor beta-arrestin 2 interaction: a high-affinity receptor phenotype. Mol Endocrinol 19:812–823PubMedCrossRef Jorgensen R, Martini L, Schwartz TW, Elling CE (2005) Characterization of glucagon-like peptide-1 receptor beta-arrestin 2 interaction: a high-affinity receptor phenotype. Mol Endocrinol 19:812–823PubMedCrossRef
34.
go back to reference Jorgensen R, Kubale V, Vrecl M, Schwartz TW, Elling CE (2007) Oxyntomodulin differentially affects glucagon-like peptide-1 receptor beta-arrestin recruitment and signaling through Galpha(s). J Pharmacol Exp Ther 322:148–154PubMedCrossRef Jorgensen R, Kubale V, Vrecl M, Schwartz TW, Elling CE (2007) Oxyntomodulin differentially affects glucagon-like peptide-1 receptor beta-arrestin recruitment and signaling through Galpha(s). J Pharmacol Exp Ther 322:148–154PubMedCrossRef
35.
go back to reference Schelshorn D, Joly F, Mutel S, Hampe C, Breton B, Mutel V, Lutjens R (2012) Lateral allosterism in the glucagon receptor family: glucagon-like peptide 1 induces G-protein-coupled receptor heteromer formation. Mol Pharmacol 81:309–318PubMedCrossRef Schelshorn D, Joly F, Mutel S, Hampe C, Breton B, Mutel V, Lutjens R (2012) Lateral allosterism in the glucagon receptor family: glucagon-like peptide 1 induces G-protein-coupled receptor heteromer formation. Mol Pharmacol 81:309–318PubMedCrossRef
36.
go back to reference Alejandro EU, Lim GE, Mehran AE et al (2011) Pancreatic beta-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription. FASEB J 25:3884–3895PubMedCrossRef Alejandro EU, Lim GE, Mehran AE et al (2011) Pancreatic beta-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription. FASEB J 25:3884–3895PubMedCrossRef
37.
38.
go back to reference Meur G, Qian Q, da Silva XG et al (2011) Nucleo-cytosolic shuttling of FoxO1 directly regulates mouse Ins2 but not Ins1 gene expression in pancreatic beta cells (MIN6). J Biol Chem 286:13647–13656PubMedCrossRef Meur G, Qian Q, da Silva XG et al (2011) Nucleo-cytosolic shuttling of FoxO1 directly regulates mouse Ins2 but not Ins1 gene expression in pancreatic beta cells (MIN6). J Biol Chem 286:13647–13656PubMedCrossRef
39.
40.
go back to reference Gunasekaran U, Hudgens CW, Wright BT, Maulis MF, Gannon M (2012) Differential regulation of embryonic and adult beta cell replication. Cell Cycle 11:2431–2442PubMedCrossRef Gunasekaran U, Hudgens CW, Wright BT, Maulis MF, Gannon M (2012) Differential regulation of embryonic and adult beta cell replication. Cell Cycle 11:2431–2442PubMedCrossRef
41.
go back to reference Thorens B (2013) The required beta cell research for improving treatment of type 2 diabetes. J Intern Med 274:203–214PubMedCrossRef Thorens B (2013) The required beta cell research for improving treatment of type 2 diabetes. J Intern Med 274:203–214PubMedCrossRef
42.
go back to reference Imai J, Katagiri H, Yamada T et al (2008) Regulation of pancreatic beta cell mass by neuronal signals from the liver. Science 322:1250–1254PubMedCrossRef Imai J, Katagiri H, Yamada T et al (2008) Regulation of pancreatic beta cell mass by neuronal signals from the liver. Science 322:1250–1254PubMedCrossRef
43.
go back to reference El Ouaamari A, Kawamori D, Dirice E et al (2013) Liver-derived systemic factors drive beta cell hyperplasia in insulin-resistant states. Cell Rep 3:401–410PubMedCentralPubMedCrossRef El Ouaamari A, Kawamori D, Dirice E et al (2013) Liver-derived systemic factors drive beta cell hyperplasia in insulin-resistant states. Cell Rep 3:401–410PubMedCentralPubMedCrossRef
44.
go back to reference Yi P, Park JS, Melton DA (2013) Betatrophin: a hormone that controls pancreatic beta cell proliferation. Cell 153:747–758PubMedCrossRef Yi P, Park JS, Melton DA (2013) Betatrophin: a hormone that controls pancreatic beta cell proliferation. Cell 153:747–758PubMedCrossRef
45.
go back to reference Buzzi F, Xu L, Zuellig RA et al (2010) Differential effects of protein kinase B/Akt isoforms on glucose homeostasis and islet mass. Mol Cell Biol 30:601–612PubMedCentralPubMedCrossRef Buzzi F, Xu L, Zuellig RA et al (2010) Differential effects of protein kinase B/Akt isoforms on glucose homeostasis and islet mass. Mol Cell Biol 30:601–612PubMedCentralPubMedCrossRef
46.
go back to reference Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307:1098–1101PubMedCrossRef Sarbassov DD, Guertin DA, Ali SM, Sabatini DM (2005) Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307:1098–1101PubMedCrossRef
47.
go back to reference Kitamura T, Nakae J, Kitamura Y et al (2002) The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. J Clin Invest 110:1839–1847PubMedCentralPubMedCrossRef Kitamura T, Nakae J, Kitamura Y et al (2002) The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. J Clin Invest 110:1839–1847PubMedCentralPubMedCrossRef
48.
go back to reference Liu Y, Tanabe K, Baronnier D, Patel S, Woodgett J, Cras-Meneur C, Permutt MA (2010) Conditional ablation of Gsk-3beta in islet beta cells results in expanded mass and resistance to fat feeding-induced diabetes in mice. Diabetologia 53:2600–2610PubMedCentralPubMedCrossRef Liu Y, Tanabe K, Baronnier D, Patel S, Woodgett J, Cras-Meneur C, Permutt MA (2010) Conditional ablation of Gsk-3beta in islet beta cells results in expanded mass and resistance to fat feeding-induced diabetes in mice. Diabetologia 53:2600–2610PubMedCentralPubMedCrossRef
49.
go back to reference Liu Z, Tanabe K, Bernal-Mizrachi E, Permutt MA (2008) Mice with beta cell overexpression of glycogen synthase kinase-3beta have reduced beta cell mass and proliferation. Diabetologia 51:623–631PubMedCrossRef Liu Z, Tanabe K, Bernal-Mizrachi E, Permutt MA (2008) Mice with beta cell overexpression of glycogen synthase kinase-3beta have reduced beta cell mass and proliferation. Diabetologia 51:623–631PubMedCrossRef
50.
go back to reference Zurita E, Chagoyen M, Cantero M et al (2011) Genetic polymorphisms among C57BL/6 mouse inbred strains. Transgenic Res 20:481–489PubMedCrossRef Zurita E, Chagoyen M, Cantero M et al (2011) Genetic polymorphisms among C57BL/6 mouse inbred strains. Transgenic Res 20:481–489PubMedCrossRef
Metadata
Title
β-Arrestin2 plays a key role in the modulation of the pancreatic beta cell mass in mice
Authors
Magalie A. Ravier
Michele Leduc
Joy Richard
Nathalie Linck
Annie Varrault
Nelly Pirot
Morgane M. Roussel
Joël Bockaert
Stéphane Dalle
Gyslaine Bertrand
Publication date
01-03-2014
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 3/2014
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-013-3130-7

Other articles of this Issue 3/2014

Diabetologia 3/2014 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.