Skip to main content
Top
Published in: Diabetologia 5/2012

01-05-2012 | Article

Ectopic expression of glucagon receptor in skeletal muscles improves glucose homeostasis in a mouse model of diabetes

Authors: A. Maharaj, L. Zhu, F. Huang, H. Qiu, H. Li, C. Y. Zhang, T. Jin, Q. Wang

Published in: Diabetologia | Issue 5/2012

Login to get access

Abstract

Aims/hypothesis

Excessive secretion of glucagon partially contributes to the development of diabetic hyperglycaemia. However, complete blocking of glucagon action will lead to adverse effects, since glucagon exerts certain beneficial effects via its receptor in many organs. We aimed to study the effects of a ‘decoy receptor’ for circulating glucagon on modulating beta cell function and glucose homeostasis in mice by over-producing the glucagon receptor (GCGR) in skeletal muscles.

Methods

We generated transgenic mice in which the expression of Gcgr is driven by the muscle specific creatine kinase (Mck) promoter, and assessed the effects of glucagon on the modulation of glucose homeostasis under conditions of extremes of glucose influx or efflux.

Results

Mck/Gcgr mice showed increased circulating levels of glucagon and insulin, resulting in an unchanged ratio of glucagon-to-insulin. The levels of hepatic glucose-6-phosphatase (G6PC) and fructose-1,6-bisphosphatase (F1,6P2ase) were significantly decreased, whereas the phosphorylation level of pancreatic cAMP-response-element-binding-protein (CREB) was significantly increased in these transgenic mice. Under basal conditions, the mice displayed normal blood glucose levels and unchanged glucose tolerance and insulin sensitivity when compared with their age-matched wild-type (WT) littermates. However, following multiple low-dose streptozotocin injections, Mck/Gcgr mice exhibited a delay in the onset of hyperglycaemia compared with the WT controls. This was associated with preserved beta cell mass and beta cell secretory capacity in response to glucose challenge.

Conclusions/interpretation

We suggest that mild and chronic hyperglucagonaemia, through a strategy involving neutralising peripheral glucagon action, provides beneficial effects on beta cell function and glucose homeostasis. Mck/Gcgr mice thus represent a novel mouse model for studying the physiological effects of glucagon.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bansal P, Wang Q (2008) Insulin as a physiological modulator of glucagon secretion. Am J Physiol Endocrinol Metab 295:E751–E761PubMedCrossRef Bansal P, Wang Q (2008) Insulin as a physiological modulator of glucagon secretion. Am J Physiol Endocrinol Metab 295:E751–E761PubMedCrossRef
2.
go back to reference Jiang G, Zhang BB (2003) Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol Metab 284:E671–E678PubMed Jiang G, Zhang BB (2003) Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol Metab 284:E671–E678PubMed
3.
go back to reference Gromada J, Franklin I, Wollheim CB (2007) Alpha-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 28:84–116PubMedCrossRef Gromada J, Franklin I, Wollheim CB (2007) Alpha-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocr Rev 28:84–116PubMedCrossRef
4.
go back to reference Gastaldelli A, Baldi S, Pettiti M et al (2000) Influence of obesity and type 2 diabetes on gluconeogenesis and glucose output in humans: a quantitative study. Diabetes 49:1367–1373PubMedCrossRef Gastaldelli A, Baldi S, Pettiti M et al (2000) Influence of obesity and type 2 diabetes on gluconeogenesis and glucose output in humans: a quantitative study. Diabetes 49:1367–1373PubMedCrossRef
5.
go back to reference Sherwin RS, Fisher M, Hendler R, Felig P (1976) Hyperglucagonemia and blood glucose regulation in normal, obese and diabetic subjects. N Engl J Med 294:455–461PubMedCrossRef Sherwin RS, Fisher M, Hendler R, Felig P (1976) Hyperglucagonemia and blood glucose regulation in normal, obese and diabetic subjects. N Engl J Med 294:455–461PubMedCrossRef
6.
go back to reference Unger RH (1978) Role of glucagon in the pathogenesis of diabetes: the status of the controversy. Metabolism 27:1691–1709PubMedCrossRef Unger RH (1978) Role of glucagon in the pathogenesis of diabetes: the status of the controversy. Metabolism 27:1691–1709PubMedCrossRef
7.
go back to reference Li XC, Liao TD, Zhuo JL (2008) Long-term hyperglucagonaemia induces early metabolic and renal phenotypes of type 2 diabetes in mice. Clin Sci (Lond) 114:591–601CrossRef Li XC, Liao TD, Zhuo JL (2008) Long-term hyperglucagonaemia induces early metabolic and renal phenotypes of type 2 diabetes in mice. Clin Sci (Lond) 114:591–601CrossRef
8.
go back to reference Rizza R, Verdonk C, Miles J, Service FJ, Gerich J (1979) Effect of intermittent endogenous hyperglucagonemia on glucose homeostasis in normal and diabetic man. J Clin Invest 63:1119–1123PubMedCrossRef Rizza R, Verdonk C, Miles J, Service FJ, Gerich J (1979) Effect of intermittent endogenous hyperglucagonemia on glucose homeostasis in normal and diabetic man. J Clin Invest 63:1119–1123PubMedCrossRef
9.
go back to reference Unger RH, Orci L (2010) Paracrinology of islets and the paracrinopathy of diabetes. Proc Natl Acad Sci U S A 107:16009–16012PubMedCrossRef Unger RH, Orci L (2010) Paracrinology of islets and the paracrinopathy of diabetes. Proc Natl Acad Sci U S A 107:16009–16012PubMedCrossRef
10.
go back to reference Gelling RW, Du XQ, Dichmann DS et al (2003) Lower blood glucose, hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100:1438–1443PubMedCrossRef Gelling RW, Du XQ, Dichmann DS et al (2003) Lower blood glucose, hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 100:1438–1443PubMedCrossRef
11.
go back to reference Conarello SL, Jiang G, Mu J et al (2007) Glucagon receptor knockout mice are resistant to diet-induced obesity and streptozotocin-mediated beta cell loss and hyperglycaemia. Diabetologia 50:142–150PubMedCrossRef Conarello SL, Jiang G, Mu J et al (2007) Glucagon receptor knockout mice are resistant to diet-induced obesity and streptozotocin-mediated beta cell loss and hyperglycaemia. Diabetologia 50:142–150PubMedCrossRef
12.
go back to reference Petersen KF, Sullivan JT (2001) Effects of a novel glucagon receptor antagonist (Bay 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia 44:2018–2024PubMedCrossRef Petersen KF, Sullivan JT (2001) Effects of a novel glucagon receptor antagonist (Bay 27-9955) on glucagon-stimulated glucose production in humans. Diabetologia 44:2018–2024PubMedCrossRef
13.
go back to reference Liang Y, Osborne MC, Monia BP et al (2004) Reduction in glucagon receptor expression by an antisense oligonucleotide ameliorates diabetic syndrome in db/db mice. Diabetes 53:410–417PubMedCrossRef Liang Y, Osborne MC, Monia BP et al (2004) Reduction in glucagon receptor expression by an antisense oligonucleotide ameliorates diabetic syndrome in db/db mice. Diabetes 53:410–417PubMedCrossRef
14.
go back to reference Gu W, Yan H, Winters KA et al (2009) Long-term inhibition of the glucagon receptor with a monoclonal antibody in mice causes sustained improvement in glycemic control, with reversible alpha-cell hyperplasia and hyperglucagonemia. J Pharmacol Exp Ther 331:871–881PubMedCrossRef Gu W, Yan H, Winters KA et al (2009) Long-term inhibition of the glucagon receptor with a monoclonal antibody in mice causes sustained improvement in glycemic control, with reversible alpha-cell hyperplasia and hyperglucagonemia. J Pharmacol Exp Ther 331:871–881PubMedCrossRef
15.
go back to reference Yan H, Gu W, Yang J et al (2009) Fully human monoclonal antibodies antagonizing the glucagon receptor improve glucose homeostasis in mice and monkeys. J Pharmacol Exp Ther 329:102–111PubMedCrossRef Yan H, Gu W, Yang J et al (2009) Fully human monoclonal antibodies antagonizing the glucagon receptor improve glucose homeostasis in mice and monkeys. J Pharmacol Exp Ther 329:102–111PubMedCrossRef
16.
go back to reference Vuguin PM, Kedees MH, Cui L et al (2006) Ablation of the glucagon receptor gene increases fetal lethality and produces alterations in islet development and maturation. Endocrinology 147:3995–4006PubMedCrossRef Vuguin PM, Kedees MH, Cui L et al (2006) Ablation of the glucagon receptor gene increases fetal lethality and produces alterations in islet development and maturation. Endocrinology 147:3995–4006PubMedCrossRef
17.
go back to reference Schertzer JD, Antonescu CN, Bilan PJ et al (2009) A transgenic mouse model to study glucose transporter 4myc regulation in skeletal muscle. Endocrinology 150:1935–1940PubMedCrossRef Schertzer JD, Antonescu CN, Bilan PJ et al (2009) A transgenic mouse model to study glucose transporter 4myc regulation in skeletal muscle. Endocrinology 150:1935–1940PubMedCrossRef
18.
go back to reference Hansen LH, Abrahamsen N, Nishimura E (1995) Glucagon receptor mRNA distribution in rat tissues. Peptides 16:1163–1166PubMedCrossRef Hansen LH, Abrahamsen N, Nishimura E (1995) Glucagon receptor mRNA distribution in rat tissues. Peptides 16:1163–1166PubMedCrossRef
19.
go back to reference Burcelin R, Li J, Charron MJ (1995) Cloning and sequence analysis of the murine glucagon receptor-encoding gene. Gene 164:305–310PubMedCrossRef Burcelin R, Li J, Charron MJ (1995) Cloning and sequence analysis of the murine glucagon receptor-encoding gene. Gene 164:305–310PubMedCrossRef
20.
go back to reference Soltani N, Kumar M, Glinka Y, Prud'homme GJ, Wang Q (2007) In vivo expression of GLP-1/IgG-Fc fusion protein enhances beta-cell mass and protects against streptozotocin-induced diabetes. Gene Ther 14:981–988PubMedCrossRef Soltani N, Kumar M, Glinka Y, Prud'homme GJ, Wang Q (2007) In vivo expression of GLP-1/IgG-Fc fusion protein enhances beta-cell mass and protects against streptozotocin-induced diabetes. Gene Ther 14:981–988PubMedCrossRef
21.
go back to reference Wang Q, Khayat Z, Kishi K, Ebina Y, Klip A (1998) GLUT4 translocation by insulin in intact muscle cells: detection by a fast and quantitative assay. FEBS Lett 427:193–197PubMedCrossRef Wang Q, Khayat Z, Kishi K, Ebina Y, Klip A (1998) GLUT4 translocation by insulin in intact muscle cells: detection by a fast and quantitative assay. FEBS Lett 427:193–197PubMedCrossRef
22.
go back to reference Jin T, Drucker DJ (1995) The proglucagon gene upstream enhancer contains positive and negative domains important for tissue-specific proglucagon gene transcription. Mol Endocrinol 9:1306–1320PubMedCrossRef Jin T, Drucker DJ (1995) The proglucagon gene upstream enhancer contains positive and negative domains important for tissue-specific proglucagon gene transcription. Mol Endocrinol 9:1306–1320PubMedCrossRef
23.
go back to reference Wang Q, Somwar R, Bilan PJ et al (1999) Protein kinase B/Akt participates in GLUT4 translocation by insulin in L6 myoblasts. Mol Cell Biol 19:4008–4018PubMed Wang Q, Somwar R, Bilan PJ et al (1999) Protein kinase B/Akt participates in GLUT4 translocation by insulin in L6 myoblasts. Mol Cell Biol 19:4008–4018PubMed
24.
go back to reference Unson CG, Cypess AM, Kim HN et al (1995) Characterization of deletion and truncation mutants of the rat glucagon receptor. Seven transmembrane segments are necessary for receptor transport to the plasma membrane and glucagon binding. J Biol Chem 270:27720–27727PubMedCrossRef Unson CG, Cypess AM, Kim HN et al (1995) Characterization of deletion and truncation mutants of the rat glucagon receptor. Seven transmembrane segments are necessary for receptor transport to the plasma membrane and glucagon binding. J Biol Chem 270:27720–27727PubMedCrossRef
25.
go back to reference Zhang N, Kumar M, Xu G et al (2006) Activin receptor-like kinase 7 induces apoptosis of pancreatic beta cells and beta cell lines. Diabetologia 49:506–518PubMedCrossRef Zhang N, Kumar M, Xu G et al (2006) Activin receptor-like kinase 7 induces apoptosis of pancreatic beta cells and beta cell lines. Diabetologia 49:506–518PubMedCrossRef
26.
go back to reference Wang Q, Brubaker PL (2002) Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice. Diabetologia 45:1263–1273PubMedCrossRef Wang Q, Brubaker PL (2002) Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice. Diabetologia 45:1263–1273PubMedCrossRef
27.
go back to reference Kim SJ, Nian C, Widenmaier S, McIntosh CH (2008) Glucose-dependent insulinotropic polypeptide-mediated up-regulation of beta-cell antiapoptotic Bcl-2 gene expression is coordinated by cyclic AMP (cAMP) response element binding protein (CREB) and cAMP-responsive CREB coactivator 2. Mol Cell Biol 28:1644–1656PubMedCrossRef Kim SJ, Nian C, Widenmaier S, McIntosh CH (2008) Glucose-dependent insulinotropic polypeptide-mediated up-regulation of beta-cell antiapoptotic Bcl-2 gene expression is coordinated by cyclic AMP (cAMP) response element binding protein (CREB) and cAMP-responsive CREB coactivator 2. Mol Cell Biol 28:1644–1656PubMedCrossRef
28.
go back to reference Jhala US, Canettieri G, Screaton RA et al (2003) cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17:1575–1580PubMedCrossRef Jhala US, Canettieri G, Screaton RA et al (2003) cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17:1575–1580PubMedCrossRef
29.
go back to reference Balage M, Grizard J (1986) Relationship between plasma glucagon disappearance and tissue uptake in rats. Reprod Nutr Dev 26:31–38PubMedCrossRef Balage M, Grizard J (1986) Relationship between plasma glucagon disappearance and tissue uptake in rats. Reprod Nutr Dev 26:31–38PubMedCrossRef
30.
go back to reference Kuwajima M, Golden S, Katz J, Unger RH, Foster DW, McGarry JD (1986) Active hepatic glycogen synthesis from gluconeogenic precursors despite high tissue levels of fructose 2,6-bisphosphate. J Biol Chem 261:2632–2637PubMed Kuwajima M, Golden S, Katz J, Unger RH, Foster DW, McGarry JD (1986) Active hepatic glycogen synthesis from gluconeogenic precursors despite high tissue levels of fructose 2,6-bisphosphate. J Biol Chem 261:2632–2637PubMed
32.
go back to reference Huypens P, Ling Z, Pipeleers D, Schuit F (2000) Glucagon receptors on human islet cells contribute to glucose competence of insulin release. Diabetologia 43:1012–1019PubMedCrossRef Huypens P, Ling Z, Pipeleers D, Schuit F (2000) Glucagon receptors on human islet cells contribute to glucose competence of insulin release. Diabetologia 43:1012–1019PubMedCrossRef
33.
go back to reference Kawai K, Yokota C, Ohashi S, Watanabe Y, Yamashita K (1995) Evidence that glucagon stimulates insulin secretion through its own receptor in rats. Diabetologia 38:274–276PubMedCrossRef Kawai K, Yokota C, Ohashi S, Watanabe Y, Yamashita K (1995) Evidence that glucagon stimulates insulin secretion through its own receptor in rats. Diabetologia 38:274–276PubMedCrossRef
34.
go back to reference Sorensen H, Winzell MS, Brand CL et al (2006) Glucagon receptor knockout mice display increased insulin sensitivity and impaired beta-cell function. Diabetes 55:3463–3469PubMedCrossRef Sorensen H, Winzell MS, Brand CL et al (2006) Glucagon receptor knockout mice display increased insulin sensitivity and impaired beta-cell function. Diabetes 55:3463–3469PubMedCrossRef
35.
go back to reference Gelling RW, Vuguin PM, Du XQ et al (2009) Pancreatic beta-cell overexpression of the glucagon receptor gene results in enhanced beta-cell function and mass. Am J Physiol Endocrinol Metab 297:E695–E707PubMedCrossRef Gelling RW, Vuguin PM, Du XQ et al (2009) Pancreatic beta-cell overexpression of the glucagon receptor gene results in enhanced beta-cell function and mass. Am J Physiol Endocrinol Metab 297:E695–E707PubMedCrossRef
36.
go back to reference Xu E, Kumar M, Zhang Y et al (2006) Intra-islet insulin suppresses glucagon release via GABA-GABAA receptor system. Cell Metab 3:47–58PubMedCrossRef Xu E, Kumar M, Zhang Y et al (2006) Intra-islet insulin suppresses glucagon release via GABA-GABAA receptor system. Cell Metab 3:47–58PubMedCrossRef
37.
go back to reference Tsuchiyama N, Takamura T, Ando H et al (2007) Possible role of alpha-cell insulin resistance in exaggerated glucagon responses to arginine in type 2 diabetes. Diabetes Care 30:2583–2587PubMedCrossRef Tsuchiyama N, Takamura T, Ando H et al (2007) Possible role of alpha-cell insulin resistance in exaggerated glucagon responses to arginine in type 2 diabetes. Diabetes Care 30:2583–2587PubMedCrossRef
38.
go back to reference Dalle S, Longuet C, Costes S et al (2004) Glucagon promotes cAMP-response element-binding protein phosphorylation via activation of ERK1/2 in MIN6 cell line and isolated islets of Langerhans. J Biol Chem 279:20345–20355PubMedCrossRef Dalle S, Longuet C, Costes S et al (2004) Glucagon promotes cAMP-response element-binding protein phosphorylation via activation of ERK1/2 in MIN6 cell line and isolated islets of Langerhans. J Biol Chem 279:20345–20355PubMedCrossRef
39.
go back to reference Aspinwall CA, Lakey JR, Kennedy RT (1999) Insulin-stimulated insulin secretion in single pancreatic beta cells. J Biol Chem 274:6360–6365PubMedCrossRef Aspinwall CA, Lakey JR, Kennedy RT (1999) Insulin-stimulated insulin secretion in single pancreatic beta cells. J Biol Chem 274:6360–6365PubMedCrossRef
40.
go back to reference Wang Q, Jin T (2009) The role of insulin signaling in the development of beta-cell dysfunction and diabetes. Islets 1:95–101PubMedCrossRef Wang Q, Jin T (2009) The role of insulin signaling in the development of beta-cell dysfunction and diabetes. Islets 1:95–101PubMedCrossRef
41.
go back to reference Johnson JD, Bernal-Mizrachi E, Alejandro EU et al (2006) Insulin protects islets from apoptosis via Pdx1 and specific changes in the human islet proteome. Proc Natl Acad Sci U S A 103:19575–19580PubMedCrossRef Johnson JD, Bernal-Mizrachi E, Alejandro EU et al (2006) Insulin protects islets from apoptosis via Pdx1 and specific changes in the human islet proteome. Proc Natl Acad Sci U S A 103:19575–19580PubMedCrossRef
42.
go back to reference Paraskevas S, Aikin R, Maysinger D et al (2001) Modulation of JNK and p38 stress activated protein kinases in isolated islets of Langerhans: insulin as an autocrine survival signal. Ann Surg 233:124–133PubMedCrossRef Paraskevas S, Aikin R, Maysinger D et al (2001) Modulation of JNK and p38 stress activated protein kinases in isolated islets of Langerhans: insulin as an autocrine survival signal. Ann Surg 233:124–133PubMedCrossRef
43.
go back to reference Jetton TL, Lausier J, LaRock K et al (2005) Mechanisms of compensatory beta-cell growth in insulin-resistant rats: roles of Akt kinase. Diabetes 54:2294–2304PubMedCrossRef Jetton TL, Lausier J, LaRock K et al (2005) Mechanisms of compensatory beta-cell growth in insulin-resistant rats: roles of Akt kinase. Diabetes 54:2294–2304PubMedCrossRef
44.
go back to reference Maeda H, Rajesh KG, Maeda H, Suzuki R, Sasaguri S (2004) Epidermal growth factor and insulin inhibit cell death in pancreatic beta cells by activation of PI3-kinase/AKT signaling pathway under oxidative stress. Transplant Proc 36:1163–1165PubMedCrossRef Maeda H, Rajesh KG, Maeda H, Suzuki R, Sasaguri S (2004) Epidermal growth factor and insulin inhibit cell death in pancreatic beta cells by activation of PI3-kinase/AKT signaling pathway under oxidative stress. Transplant Proc 36:1163–1165PubMedCrossRef
45.
go back to reference Zhao J, Zhang N, He M et al (2008) Increased beta-cell apoptosis and impaired insulin signaling pathway contributes to the onset of diabetes in OLETF rats. Cell Physiol Biochem 21:445–454PubMedCrossRef Zhao J, Zhang N, He M et al (2008) Increased beta-cell apoptosis and impaired insulin signaling pathway contributes to the onset of diabetes in OLETF rats. Cell Physiol Biochem 21:445–454PubMedCrossRef
46.
go back to reference Peltier J, O'Neill A, Schaffer DV (2007) PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation. Dev Neurobiol 67:1348–1361PubMedCrossRef Peltier J, O'Neill A, Schaffer DV (2007) PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation. Dev Neurobiol 67:1348–1361PubMedCrossRef
47.
go back to reference Ramnanan CJ, Edgerton DS, Kraft G, Cherrington AD (2011) Physiologic action of glucagon on liver glucose metabolism. Diabetes Obes Metab 13(Suppl 1):S118–S125CrossRef Ramnanan CJ, Edgerton DS, Kraft G, Cherrington AD (2011) Physiologic action of glucagon on liver glucose metabolism. Diabetes Obes Metab 13(Suppl 1):S118–S125CrossRef
48.
go back to reference Rivera N, Ramnanan CJ, An Z et al (2010) Insulin-induced hypoglycemia increases hepatic sensitivity to glucagon in dogs. J Clin Invest 120:4425–4435PubMedCrossRef Rivera N, Ramnanan CJ, An Z et al (2010) Insulin-induced hypoglycemia increases hepatic sensitivity to glucagon in dogs. J Clin Invest 120:4425–4435PubMedCrossRef
49.
go back to reference Taniguchi CM, Tran TT, Kondo T et al (2006) Phosphoinositide 3-kinase regulatory subunit p85alpha suppresses insulin action via positive regulation of PTEN. Proc Natl Acad Sci U S A 103:12093–12097PubMedCrossRef Taniguchi CM, Tran TT, Kondo T et al (2006) Phosphoinositide 3-kinase regulatory subunit p85alpha suppresses insulin action via positive regulation of PTEN. Proc Natl Acad Sci U S A 103:12093–12097PubMedCrossRef
50.
go back to reference Fisher SJ, Kahn CR (2003) Insulin signaling is required for insulin’s direct and indirect action on hepatic glucose production. J Clin Invest 111:463–468PubMed Fisher SJ, Kahn CR (2003) Insulin signaling is required for insulin’s direct and indirect action on hepatic glucose production. J Clin Invest 111:463–468PubMed
Metadata
Title
Ectopic expression of glucagon receptor in skeletal muscles improves glucose homeostasis in a mouse model of diabetes
Authors
A. Maharaj
L. Zhu
F. Huang
H. Qiu
H. Li
C. Y. Zhang
T. Jin
Q. Wang
Publication date
01-05-2012
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 5/2012
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-012-2464-x

Other articles of this Issue 5/2012

Diabetologia 5/2012 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.