Skip to main content
Top
Published in: Diabetologia 2/2008

01-02-2008 | Article

Inhibition of c-jun N terminal kinase (JNK) improves functional beta cell mass in human islets and leads to AKT and glycogen synthase kinase-3 (GSK-3) phosphorylation

Authors: A. Fornoni, A. Pileggi, R. D. Molano, N. Y. Sanabria, T. Tejada, J. Gonzalez-Quintana, H. Ichii, L. Inverardi, C. Ricordi, R. L. Pastori

Published in: Diabetologia | Issue 2/2008

Login to get access

Abstract

Aims/hypothesis

Activation of c-jun N-terminal kinase (JNK) has been described in islet isolation and engraftment, making JNK a key target in islet transplantation. The objective of this study was to investigate if JNK inhibition with a cell-permeable TAT peptide inhibitor (L-JNKI) protects functional beta cell mass in human islets and affects AKT and its substrates in islet cells.

Methods

The effect of L-JNKI (10 μmol/l) on islet count, mitochondrial membrane potential, glucose-stimulated insulin release and phosphorylation of both AKT and its substrates, as well as on reversal of diabetes in immunodeficient diabetic Nu/Nu mice was studied.

Results

In vitro, L-JNKI reduced the islet loss in culture and protected from cell death caused by acute cytokine exposure. In vivo, treatment of freshly isolated human islets and diabetic Nu/Nu mice recipients of such islets resulted in improved functional beta cell mass. We showed that L-JNKI activates AKT and downregulates glycogen synthase kinase-3 beta (GSK-3B) in human islets exposed to cytokines, while other AKT substrates were unaffected, suggesting that a specific AKT/GSK-3B regulation by L-JNKI may represent one of its mechanisms of cytoprotection.

Conclusions/interpretation

In conclusion, we have demonstrated that targeting JNK in human pancreatic islets results in improved functional beta cell mass and in the regulation of AKT/GSK3B activity.
Appendix
Available only for authorised users
Literature
1.
go back to reference Abdelli S, Ansite J, Roduit R et al (2004) Intracellular stress signaling pathways activated during human islet preparation and following acute cytokine exposure. Diabetes 53:2815–2823PubMedCrossRef Abdelli S, Ansite J, Roduit R et al (2004) Intracellular stress signaling pathways activated during human islet preparation and following acute cytokine exposure. Diabetes 53:2815–2823PubMedCrossRef
2.
go back to reference Ammendrup A, Maillard A, Nielsen K et al (2000) The c-Jun amino-terminal kinase pathway is preferentially activated by interleukin-1 and controls apoptosis in differentiating pancreatic beta-cells. Diabetes 49:1468–1476PubMedCrossRef Ammendrup A, Maillard A, Nielsen K et al (2000) The c-Jun amino-terminal kinase pathway is preferentially activated by interleukin-1 and controls apoptosis in differentiating pancreatic beta-cells. Diabetes 49:1468–1476PubMedCrossRef
3.
go back to reference Rosenberg L, Wang R, Paraskevas S, Maysinger D (1999) Structural and functional changes resulting from islet isolation lead to islet cell death. Surgery 126:393–398PubMed Rosenberg L, Wang R, Paraskevas S, Maysinger D (1999) Structural and functional changes resulting from islet isolation lead to islet cell death. Surgery 126:393–398PubMed
4.
go back to reference Aikin R, Maysinger D, Rosenberg L (2004) Cross-talk between phosphatidylinositol 3-kinase/AKT and c-jun NH2-terminal kinase mediates survival of isolated human islets. Endocrinology 145:4522–4531PubMedCrossRef Aikin R, Maysinger D, Rosenberg L (2004) Cross-talk between phosphatidylinositol 3-kinase/AKT and c-jun NH2-terminal kinase mediates survival of isolated human islets. Endocrinology 145:4522–4531PubMedCrossRef
5.
go back to reference Noguchi H, Nakai Y, Ueda M et al (2007) Activation of c-Jun NH(2)-terminal kinase (JNK) pathway during islet transplantation and prevention of islet graft loss by intraportal injection of JNK inhibitor. Diabetologia 50:612–619PubMedCrossRef Noguchi H, Nakai Y, Ueda M et al (2007) Activation of c-Jun NH(2)-terminal kinase (JNK) pathway during islet transplantation and prevention of islet graft loss by intraportal injection of JNK inhibitor. Diabetologia 50:612–619PubMedCrossRef
6.
go back to reference Abdelli S, Abderrahmani A, Hering BJ, Beckmann JS, Bonny C (2007) The c-Jun N-terminal kinase JNK participates in cytokine- and isolation stress-induced rat pancreatic islet apoptosis. Diabetologia 50:1660–1669PubMedCrossRef Abdelli S, Abderrahmani A, Hering BJ, Beckmann JS, Bonny C (2007) The c-Jun N-terminal kinase JNK participates in cytokine- and isolation stress-induced rat pancreatic islet apoptosis. Diabetologia 50:1660–1669PubMedCrossRef
7.
go back to reference Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF (2001) Cell-permeable peptide inhibitors of JNK: novel blockers of beta-cell death. Diabetes 50:77–82PubMedCrossRef Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF (2001) Cell-permeable peptide inhibitors of JNK: novel blockers of beta-cell death. Diabetes 50:77–82PubMedCrossRef
8.
go back to reference Fornoni A, Cobianchi L, Sanabria NY et al (2007) The l-isoform but not d-isoforms of a JNK inhibitory peptide protects pancreatic beta-cells. Biochem Biophys Res Commun 354:227–233PubMedCrossRef Fornoni A, Cobianchi L, Sanabria NY et al (2007) The l-isoform but not d-isoforms of a JNK inhibitory peptide protects pancreatic beta-cells. Biochem Biophys Res Commun 354:227–233PubMedCrossRef
9.
go back to reference Noguchi H, Nakai Y, Matsumoto S et al (2005) Cell permeable peptide of JNK inhibitor prevents islet apoptosis immediately after isolation and improves islet graft function. Am J Transplant 5:1848–1855PubMedCrossRef Noguchi H, Nakai Y, Matsumoto S et al (2005) Cell permeable peptide of JNK inhibitor prevents islet apoptosis immediately after isolation and improves islet graft function. Am J Transplant 5:1848–1855PubMedCrossRef
10.
go back to reference Ribeiro MM, Klein D, Pileggi A et al (2003) Heme oxygenase-1 fused to a TAT peptide transduces and protects pancreatic beta-cells. Biochem Biophys Res Commun 305:876–881PubMedCrossRef Ribeiro MM, Klein D, Pileggi A et al (2003) Heme oxygenase-1 fused to a TAT peptide transduces and protects pancreatic beta-cells. Biochem Biophys Res Commun 305:876–881PubMedCrossRef
11.
go back to reference Klein D, Ribeiro MM, Mendoza V et al (2004) Delivery of Bcl-XL or its BH4 domain by protein transduction inhibits apoptosis in human islets. Biochem Biophys Res Commun 323:473–478PubMedCrossRef Klein D, Ribeiro MM, Mendoza V et al (2004) Delivery of Bcl-XL or its BH4 domain by protein transduction inhibits apoptosis in human islets. Biochem Biophys Res Commun 323:473–478PubMedCrossRef
12.
go back to reference Cardozo AK, Buchillier V, Mathieu M et al (2007) Cell-permeable peptides induce dose- and length-dependent cytotoxic effects. Biochim Biophys Acta 1768:2222–2234PubMedCrossRef Cardozo AK, Buchillier V, Mathieu M et al (2007) Cell-permeable peptides induce dose- and length-dependent cytotoxic effects. Biochim Biophys Acta 1768:2222–2234PubMedCrossRef
13.
go back to reference Hirosumi J, Tuncman G, Chang L et al (2002) A central role for JNK in obesity and insulin resistance. Nature 420:333–336PubMedCrossRef Hirosumi J, Tuncman G, Chang L et al (2002) A central role for JNK in obesity and insulin resistance. Nature 420:333–336PubMedCrossRef
14.
go back to reference Kaneto H, Nakatani Y, Miyatsuka T et al (2004) Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide. Nat Med 10:1128–1132PubMedCrossRef Kaneto H, Nakatani Y, Miyatsuka T et al (2004) Possible novel therapy for diabetes with cell-permeable JNK-inhibitory peptide. Nat Med 10:1128–1132PubMedCrossRef
15.
go back to reference Lee YH, Giraud J, Davis RJ, White MF (2003) c-Jun N-terminal kinase (JNK) mediates feedback inhibition of the insulin signaling cascade. J Biol Chem 278:2896–2902PubMedCrossRef Lee YH, Giraud J, Davis RJ, White MF (2003) c-Jun N-terminal kinase (JNK) mediates feedback inhibition of the insulin signaling cascade. J Biol Chem 278:2896–2902PubMedCrossRef
16.
go back to reference Bernal-Mizrachi E, Wen W, Stahlhut S, Welling CM, Permutt MA (2001) Islet beta cell expression of constitutively active Akt1/PKB alpha induces striking hypertrophy, hyperplasia, and hyperinsulinemia. J Clin Invest 108:1631–1638PubMedCrossRef Bernal-Mizrachi E, Wen W, Stahlhut S, Welling CM, Permutt MA (2001) Islet beta cell expression of constitutively active Akt1/PKB alpha induces striking hypertrophy, hyperplasia, and hyperinsulinemia. J Clin Invest 108:1631–1638PubMedCrossRef
17.
go back to reference Tuttle RL, Gill NS, Pugh W et al (2001) Regulation of pancreatic beta-cell growth and survival by the serine/threonine protein kinase Akt1/PKBalpha. Nat Med 7:1133–1137PubMedCrossRef Tuttle RL, Gill NS, Pugh W et al (2001) Regulation of pancreatic beta-cell growth and survival by the serine/threonine protein kinase Akt1/PKBalpha. Nat Med 7:1133–1137PubMedCrossRef
18.
go back to reference Aikin R, Rosenberg L, Maysinger D (2000) Phosphatidylinositol 3-kinase signaling to Akt mediates survival in isolated canine islets of Langerhans. Biochem Biophys Res Commun 277:455–461PubMedCrossRef Aikin R, Rosenberg L, Maysinger D (2000) Phosphatidylinositol 3-kinase signaling to Akt mediates survival in isolated canine islets of Langerhans. Biochem Biophys Res Commun 277:455–461PubMedCrossRef
19.
go back to reference Ricordi C, Strom TB (2004) Clinical islet transplantation: advances and immunological challenges. Nat Rev Immunol 4:259–268PubMedCrossRef Ricordi C, Strom TB (2004) Clinical islet transplantation: advances and immunological challenges. Nat Rev Immunol 4:259–268PubMedCrossRef
20.
go back to reference Ichii H, Pileggi A, Molano RD et al (2005) Rescue purification maximizes the use of human islet preparations for transplantation. Am J Transplant 5:21–30PubMedCrossRef Ichii H, Pileggi A, Molano RD et al (2005) Rescue purification maximizes the use of human islet preparations for transplantation. Am J Transplant 5:21–30PubMedCrossRef
21.
go back to reference Latif ZA, Noel J, Alejandro R (1988) A simple method of staining fresh and cultured islets. Transplantation 45:827–830PubMedCrossRef Latif ZA, Noel J, Alejandro R (1988) A simple method of staining fresh and cultured islets. Transplantation 45:827–830PubMedCrossRef
22.
go back to reference Ricordi C, Gray DW, Hering BJ et al (1990) Islet isolation assessment in man and large animals. Acta Diabetol Lat 27:185–195PubMedCrossRef Ricordi C, Gray DW, Hering BJ et al (1990) Islet isolation assessment in man and large animals. Acta Diabetol Lat 27:185–195PubMedCrossRef
23.
go back to reference Ichii H, Inverardi L, Pileggi A et al (2005) A novel method for the assessment of cellular composition and beta-cell viability in human islet preparations. Am J Transplant 5:1635–1645PubMedCrossRef Ichii H, Inverardi L, Pileggi A et al (2005) A novel method for the assessment of cellular composition and beta-cell viability in human islet preparations. Am J Transplant 5:1635–1645PubMedCrossRef
24.
go back to reference Lukowiak B, Vandewalle B, Riachy R et al (2001) Identification and purification of functional human beta-cells by a new specific zinc-fluorescent probe. J Histochem Cytochem 49:519–528PubMed Lukowiak B, Vandewalle B, Riachy R et al (2001) Identification and purification of functional human beta-cells by a new specific zinc-fluorescent probe. J Histochem Cytochem 49:519–528PubMed
25.
go back to reference Brendel MD, Kong SS, Alejandro R, Mintz DH (1994) Improved functional survival of human islets of Langerhans in three-dimensional matrix culture. Cell Transplant 3:427–435PubMed Brendel MD, Kong SS, Alejandro R, Mintz DH (1994) Improved functional survival of human islets of Langerhans in three-dimensional matrix culture. Cell Transplant 3:427–435PubMed
26.
go back to reference Berney T, Molano RD, Cattan P et al (2001) Endotoxin-mediated delayed islet graft function is associated with increased intra-islet cytokine production and islet cell apoptosis. Transplantation 71:125–132PubMedCrossRef Berney T, Molano RD, Cattan P et al (2001) Endotoxin-mediated delayed islet graft function is associated with increased intra-islet cytokine production and islet cell apoptosis. Transplantation 71:125–132PubMedCrossRef
27.
go back to reference Pileggi A, Molano RD, Berney T et al (2001) Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation. Diabetes 50:1983–1991PubMedCrossRef Pileggi A, Molano RD, Berney T et al (2001) Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation. Diabetes 50:1983–1991PubMedCrossRef
28.
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
29.
go back to reference Aikin R, Hanley S, Maysinger D et al (2006) Autocrine insulin action activates Akt and increases survival of isolated human islets. Diabetologia 49:2900–2909PubMedCrossRef Aikin R, Hanley S, Maysinger D et al (2006) Autocrine insulin action activates Akt and increases survival of isolated human islets. Diabetologia 49:2900–2909PubMedCrossRef
30.
go back to reference Lakey JR, Burridge PW, Shapiro AM (2003) Technical aspects of islet preparation and transplantation. Transpl Int 16:613–632PubMedCrossRef Lakey JR, Burridge PW, Shapiro AM (2003) Technical aspects of islet preparation and transplantation. Transpl Int 16:613–632PubMedCrossRef
31.
go back to reference Toyama H, Takada M, Suzuki Y, Kuroda Y (2003) Activation of macrophage-associated molecules after brain death in islets. Cell Transplant 12:27–32PubMed Toyama H, Takada M, Suzuki Y, Kuroda Y (2003) Activation of macrophage-associated molecules after brain death in islets. Cell Transplant 12:27–32PubMed
32.
go back to reference Noguchi H, Matsumoto S (2006) Protein transduction technology: a novel therapeutic perspective. Acta Med Okayama 60:1–11PubMed Noguchi H, Matsumoto S (2006) Protein transduction technology: a novel therapeutic perspective. Acta Med Okayama 60:1–11PubMed
33.
go back to reference Eckhoff DE, Smyth CA, Eckstein C et al (2003) Suppression of the c-Jun N-terminal kinase pathway by 17beta-estradiol can preserve human islet functional mass from proinflammatory cytokine-induced destruction. Surgery 134:169–179PubMedCrossRef Eckhoff DE, Smyth CA, Eckstein C et al (2003) Suppression of the c-Jun N-terminal kinase pathway by 17beta-estradiol can preserve human islet functional mass from proinflammatory cytokine-induced destruction. Surgery 134:169–179PubMedCrossRef
34.
go back to reference Paraskevas S, Aikin R, Maysinger D et al (1999) Activation and expression of ERK, JNK, and p38 MAP-kinases in isolated islets of Langerhans: implications for cultured islet survival. FEBS Lett 455:203–208PubMedCrossRef Paraskevas S, Aikin R, Maysinger D et al (1999) Activation and expression of ERK, JNK, and p38 MAP-kinases in isolated islets of Langerhans: implications for cultured islet survival. FEBS Lett 455:203–208PubMedCrossRef
35.
go back to reference Kutlu B, Cardozo AK, Darville MI et al (2003) Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells. Diabetes 52:2701–2719PubMedCrossRef Kutlu B, Cardozo AK, Darville MI et al (2003) Discovery of gene networks regulating cytokine-induced dysfunction and apoptosis in insulin-producing INS-1 cells. Diabetes 52:2701–2719PubMedCrossRef
36.
go back to reference Scarim AL, Heitmeier MR, Corbett JA (1997) Irreversible inhibition of metabolic function and islet destruction after a 36-hour exposure to interleukin-1beta. Endocrinology 138:5301–5307PubMedCrossRef Scarim AL, Heitmeier MR, Corbett JA (1997) Irreversible inhibition of metabolic function and islet destruction after a 36-hour exposure to interleukin-1beta. Endocrinology 138:5301–5307PubMedCrossRef
37.
go back to reference Eizirik DL, Mandrup-Poulsen T (2001) A choice of death—the signal-transduction of immune-mediated beta-cell apoptosis. Diabetologia 44:2115–2133PubMedCrossRef Eizirik DL, Mandrup-Poulsen T (2001) A choice of death—the signal-transduction of immune-mediated beta-cell apoptosis. Diabetologia 44:2115–2133PubMedCrossRef
38.
go back to reference Montolio M, Biarnes M, Tellez N, Escoriza J, Soler J, Montanya E (2007) Interleukin-1beta and inducible form of nitric oxide synthase expression in early syngeneic islet transplantation. J Endocrinol 192:169–177PubMedCrossRef Montolio M, Biarnes M, Tellez N, Escoriza J, Soler J, Montanya E (2007) Interleukin-1beta and inducible form of nitric oxide synthase expression in early syngeneic islet transplantation. J Endocrinol 192:169–177PubMedCrossRef
39.
go back to reference Cardozo AK, Heimberg H, Heremans Y et al (2001) A comprehensive analysis of cytokine-induced and nuclear factor-kappa B-dependent genes in primary rat pancreatic beta-cells. J Biol Chem 276:48879–48886PubMedCrossRef Cardozo AK, Heimberg H, Heremans Y et al (2001) A comprehensive analysis of cytokine-induced and nuclear factor-kappa B-dependent genes in primary rat pancreatic beta-cells. J Biol Chem 276:48879–48886PubMedCrossRef
40.
go back to reference Sarti P, Arese M, Bacchi A et al (2003) Nitric oxide and mitochondrial complex IV. IUBMB Life 55:605–611PubMedCrossRef Sarti P, Arese M, Bacchi A et al (2003) Nitric oxide and mitochondrial complex IV. IUBMB Life 55:605–611PubMedCrossRef
41.
go back to reference Storling J, Binzer J, Andersson AK et al (2005) Nitric oxide contributes to cytokine-induced apoptosis in pancreatic beta cells via potentiation of JNK activity and inhibition of Akt. Diabetologia 48:2039–2050PubMedCrossRef Storling J, Binzer J, Andersson AK et al (2005) Nitric oxide contributes to cytokine-induced apoptosis in pancreatic beta cells via potentiation of JNK activity and inhibition of Akt. Diabetologia 48:2039–2050PubMedCrossRef
42.
go back to reference Muller D, Huang GC, Amiel S, Jones PM, Persaud SJ (2006) Identification of insulin signaling elements in human β-cells: autocrine regulation of insulin gene expression. Diabetes 55:2835–2842PubMedCrossRef Muller D, Huang GC, Amiel S, Jones PM, Persaud SJ (2006) Identification of insulin signaling elements in human β-cells: autocrine regulation of insulin gene expression. Diabetes 55:2835–2842PubMedCrossRef
43.
go back to reference Leibiger IB, Leibiger B, Berggren PO (2002) Insulin feedback action on pancreatic beta-cell function. FEBS Lett 532:1–6PubMedCrossRef Leibiger IB, Leibiger B, Berggren PO (2002) Insulin feedback action on pancreatic beta-cell function. FEBS Lett 532:1–6PubMedCrossRef
44.
go back to reference Mussmann R, Geese M, Harder F et al (2007) Inhibition of GSK3 promotes replication and survival of pancreatic beta cells. J Biol Chem 282:12030–12037PubMedCrossRef Mussmann R, Geese M, Harder F et al (2007) Inhibition of GSK3 promotes replication and survival of pancreatic beta cells. J Biol Chem 282:12030–12037PubMedCrossRef
45.
go back to reference Amaral ME, Cunha DA, Anhe GF et al (2004) Participation of prolactin receptors and phosphatidylinositol 3-kinase and MAP kinase pathways in the increase in pancreatic islet mass and sensitivity to glucose during pregnancy. J Endocrinol 183:469–476PubMedCrossRef Amaral ME, Cunha DA, Anhe GF et al (2004) Participation of prolactin receptors and phosphatidylinositol 3-kinase and MAP kinase pathways in the increase in pancreatic islet mass and sensitivity to glucose during pregnancy. J Endocrinol 183:469–476PubMedCrossRef
46.
go back to reference Nascimento EB, Fodor M, van der Zon GC et al (2006) Insulin-mediated phosphorylation of the proline-rich Akt substrate PRAS40 is impaired in insulin target tissues of high-fat diet-fed rats. Diabetes 55:3221–3228PubMedCrossRef Nascimento EB, Fodor M, van der Zon GC et al (2006) Insulin-mediated phosphorylation of the proline-rich Akt substrate PRAS40 is impaired in insulin target tissues of high-fat diet-fed rats. Diabetes 55:3221–3228PubMedCrossRef
47.
go back to reference Saito A, Narasimhan P, Hayashi T, Okuno S, Ferrand-Drake M, Chan PH (2004) Neuroprotective role of a proline-rich Akt substrate in apoptotic neuronal cell death after stroke: relationships with nerve growth factor. J Neurosci 24:1584–1593PubMedCrossRef Saito A, Narasimhan P, Hayashi T, Okuno S, Ferrand-Drake M, Chan PH (2004) Neuroprotective role of a proline-rich Akt substrate in apoptotic neuronal cell death after stroke: relationships with nerve growth factor. J Neurosci 24:1584–1593PubMedCrossRef
48.
go back to reference Vilimek D, Duronio V (2006) Cytokine-stimulated phosphorylation of GSK-3 is primarily dependent upon PKCs, not PKB. Biochem Cell Biol 84:20–29PubMedCrossRef Vilimek D, Duronio V (2006) Cytokine-stimulated phosphorylation of GSK-3 is primarily dependent upon PKCs, not PKB. Biochem Cell Biol 84:20–29PubMedCrossRef
49.
go back to reference He J, Usui I, Ishizuka K et al (2006) Interleukin-1alpha inhibits insulin signaling with phosphorylating insulin receptor substrate-1 on serine residues in 3T3-L1 adipocytes. Mol Endocrinol 20:114–124PubMedCrossRef He J, Usui I, Ishizuka K et al (2006) Interleukin-1alpha inhibits insulin signaling with phosphorylating insulin receptor substrate-1 on serine residues in 3T3-L1 adipocytes. Mol Endocrinol 20:114–124PubMedCrossRef
50.
go back to reference Fahmy RG, Waldman A, Zhang G et al (2006) Suppression of vascular permeability and inflammation by targeting of the transcription factor c-Jun. Nat Biotechnol 24:856–863PubMedCrossRef Fahmy RG, Waldman A, Zhang G et al (2006) Suppression of vascular permeability and inflammation by targeting of the transcription factor c-Jun. Nat Biotechnol 24:856–863PubMedCrossRef
Metadata
Title
Inhibition of c-jun N terminal kinase (JNK) improves functional beta cell mass in human islets and leads to AKT and glycogen synthase kinase-3 (GSK-3) phosphorylation
Authors
A. Fornoni
A. Pileggi
R. D. Molano
N. Y. Sanabria
T. Tejada
J. Gonzalez-Quintana
H. Ichii
L. Inverardi
C. Ricordi
R. L. Pastori
Publication date
01-02-2008
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 2/2008
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-007-0889-4

Other articles of this Issue 2/2008

Diabetologia 2/2008 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