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

01-03-2014 | Article

Islet infiltration, cytokine expression and beta cell death in the NOD mouse, BB rat, Komeda rat, LEW.1AR1-iddm rat and humans with type 1 diabetes

Authors: Anne Jörns, Tanja Arndt, Andreas Meyer zu Vilsendorf, Jürgen Klempnauer, Dirk Wedekind, Hans-Jürgen Hedrich, Lorella Marselli, Piero Marchetti, Nagakatsu Harada, Yutaka Nakaya, Gen-Sheng Wang, Fraser W. Scott, Conny Gysemans, Chantal Mathieu, Sigurd Lenzen

Published in: Diabetologia | Issue 3/2014

Login to get access

Abstract

Aims/hypothesis

Research on the pathogenesis of type 1 diabetes relies heavily on good animal models. The aim of this work was to study the translational value of animal models of type 1 diabetes to the human situation.

Methods

We compared the four major animal models of spontaneous type 1 diabetes, namely the NOD mouse, BioBreeding (BB) rat, Komeda rat and LEW.1AR1-iddm rat, by examining the immunohistochemistry and in situ RT-PCR of immune cell infiltrate and cytokine pattern in pancreatic islets, and by comparing findings with human data.

Results

After type 1 diabetes manifestation CD8+ T cells, CD68+ macrophages and CD4+ T cells were observed as the main immune cell types with declining frequency, in infiltrated islets of all diabetic pancreases. IL-1β and TNF-α were the main proinflammatory cytokines in the immune cell infiltrate in NOD mice, BB rats and LEW.1AR1-iddm rats, as well as in humans. The Komeda rat was the exception, with IFN-γ and TNF-α being the main cytokines. In addition, IL-17 and IL-6 and the anti-inflammatory cytokines IL-4, IL-10 and IL-13 were found in some infiltrating immune cells. Apoptotic as well as proliferating beta cells were observed in infiltrated islets. In healthy pancreases no proinflammatory cytokine expression was observed.

Conclusions/interpretation

With the exception of the Komeda rat, the animal models mirror very well the situation in humans with type 1 diabetes. Thus animal models of type 1 diabetes can provide meaningful information on the disease processes in the pancreas of patients with type 1 diabetes.
Appendix
Available only for authorised users
Literature
1.
go back to reference Imagawa A, Hanafusa T, Itoh N et al (1996) Islet-infiltrating T lymphocytes in insulin-dependent diabetic patients express CD80 (B7-1) and CD86 (B7-2). J Autoimmun 9:391–396PubMedCrossRef Imagawa A, Hanafusa T, Itoh N et al (1996) Islet-infiltrating T lymphocytes in insulin-dependent diabetic patients express CD80 (B7-1) and CD86 (B7-2). J Autoimmun 9:391–396PubMedCrossRef
2.
go back to reference Imagawa A, Hanafusa T, Tamura S et al (2001) Pancreatic biopsy as a procedure for detecting in situ autoimmune phenomena in type 1 diabetes: close correlation between serological markers and histological evidence of cellular autoimmunity. Diabetes 50:1269–1273PubMedCrossRef Imagawa A, Hanafusa T, Tamura S et al (2001) Pancreatic biopsy as a procedure for detecting in situ autoimmune phenomena in type 1 diabetes: close correlation between serological markers and histological evidence of cellular autoimmunity. Diabetes 50:1269–1273PubMedCrossRef
3.
go back to reference Uno S, Imagawa A, Okita K et al (2007) Macrophages and dendritic cells infiltrating islets with or without beta cells produce tumour necrosis factor-alpha in patients with recent-onset type 1 diabetes. Diabetologia 50:596–601PubMedCrossRef Uno S, Imagawa A, Okita K et al (2007) Macrophages and dendritic cells infiltrating islets with or without beta cells produce tumour necrosis factor-alpha in patients with recent-onset type 1 diabetes. Diabetologia 50:596–601PubMedCrossRef
4.
go back to reference Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y (1980) Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29:1–13PubMed Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y (1980) Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29:1–13PubMed
5.
go back to reference Anderson MS, Bluestone JA (2005) The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 23:447–485PubMedCrossRef Anderson MS, Bluestone JA (2005) The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 23:447–485PubMedCrossRef
6.
go back to reference Gysemans C, Callewaert H, Moore F et al (2009) Interferon regulatory factor-1 is a key transcription factor in murine beta cells under immune attack. Diabetologia 52:2374–2384PubMedCrossRef Gysemans C, Callewaert H, Moore F et al (2009) Interferon regulatory factor-1 is a key transcription factor in murine beta cells under immune attack. Diabetologia 52:2374–2384PubMedCrossRef
8.
go back to reference Nakhooda AF, Like AA, Chappel CI, Murray FT, Marliss EB (1977) The spontaneously diabetic Wistar rat. Metabolic and morphologic studies. Diabetes 26:100–112PubMedCrossRef Nakhooda AF, Like AA, Chappel CI, Murray FT, Marliss EB (1977) The spontaneously diabetic Wistar rat. Metabolic and morphologic studies. Diabetes 26:100–112PubMedCrossRef
9.
go back to reference Mordes JP, Bortell R, Blankenhorn EP, Rossini AA, Greiner DL (2004) Rat models of type 1 diabetes: genetics, environment, and autoimmunity. ILAR J 45:278–291PubMedCrossRef Mordes JP, Bortell R, Blankenhorn EP, Rossini AA, Greiner DL (2004) Rat models of type 1 diabetes: genetics, environment, and autoimmunity. ILAR J 45:278–291PubMedCrossRef
10.
go back to reference Wang GS, Kauri LM, Patrick C, Bareggi M, Rosenberg L, Scott FW (2010) Enhanced islet expansion by beta-cell proliferation in young diabetes-prone rats fed a protective diet. J Cell Physiol 224:501–508PubMedCrossRef Wang GS, Kauri LM, Patrick C, Bareggi M, Rosenberg L, Scott FW (2010) Enhanced islet expansion by beta-cell proliferation in young diabetes-prone rats fed a protective diet. J Cell Physiol 224:501–508PubMedCrossRef
11.
go back to reference Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, Natori T (1991) New inbred strain of Long-Evans Tokushima lean rats with IDDM without lymphopenia. Diabetes 40:1375–1381PubMedCrossRef Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, Natori T (1991) New inbred strain of Long-Evans Tokushima lean rats with IDDM without lymphopenia. Diabetes 40:1375–1381PubMedCrossRef
12.
go back to reference Komeda K, Noda M, Terao K, Kuzuya N, Kanazawa M, Kanazawa Y (1998) Establishment of two substrains, diabetes-prone and non-diabetic, from Long-Evans Tokushima Lean (LETL) rats. Endocr J 45:737–744PubMedCrossRef Komeda K, Noda M, Terao K, Kuzuya N, Kanazawa M, Kanazawa Y (1998) Establishment of two substrains, diabetes-prone and non-diabetic, from Long-Evans Tokushima Lean (LETL) rats. Endocr J 45:737–744PubMedCrossRef
13.
go back to reference Lenzen S, Tiedge M, Elsner M et al (2001) The LEW.1AR1/Ztm-iddm rat: a new model of spontaneous insulin-dependent diabetes mellitus. Diabetologia 44:1189–1196PubMedCrossRef Lenzen S, Tiedge M, Elsner M et al (2001) The LEW.1AR1/Ztm-iddm rat: a new model of spontaneous insulin-dependent diabetes mellitus. Diabetologia 44:1189–1196PubMedCrossRef
14.
go back to reference Jörns A, Günther A, Hedrich HJ, Wedekind D, Tiedge M, Lenzen S (2005) Immune cell infiltration, cytokine expression, and beta-cell apoptosis during the development of type 1 diabetes in the spontaneously diabetic LEW.1AR1/Ztm-iddm rat. Diabetes 54:2041–2052PubMedCrossRef Jörns A, Günther A, Hedrich HJ, Wedekind D, Tiedge M, Lenzen S (2005) Immune cell infiltration, cytokine expression, and beta-cell apoptosis during the development of type 1 diabetes in the spontaneously diabetic LEW.1AR1/Ztm-iddm rat. Diabetes 54:2041–2052PubMedCrossRef
15.
go back to reference Arndt T, Jörns A, Weiss H et al (2013) A variable CD3(+) T cell frequency in peripheral blood lymphocytes associated with type 1 diabetes mellitus development in the LEW.1AR1-iddm rat. PLoS One 8:e64305PubMedCentralPubMedCrossRef Arndt T, Jörns A, Weiss H et al (2013) A variable CD3(+) T cell frequency in peripheral blood lymphocytes associated with type 1 diabetes mellitus development in the LEW.1AR1-iddm rat. PLoS One 8:e64305PubMedCentralPubMedCrossRef
16.
17.
go back to reference Buschard K (2011) What causes type 1 diabetes? Lessons from animal models. APMIS, Suppl 132:1–19CrossRef Buschard K (2011) What causes type 1 diabetes? Lessons from animal models. APMIS, Suppl 132:1–19CrossRef
18.
go back to reference Roep BO, Atkinson M, von Herrath M (2004) Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol 4:989–997PubMedCrossRef Roep BO, Atkinson M, von Herrath M (2004) Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol 4:989–997PubMedCrossRef
19.
go back to reference Roep BO, Buckner J, Sawcer S, Toes R, Zipp F (2012) The problems and promises of research into human immunology and autoimmune disease. Nat Med 18:48–53PubMedCrossRef Roep BO, Buckner J, Sawcer S, Toes R, Zipp F (2012) The problems and promises of research into human immunology and autoimmune disease. Nat Med 18:48–53PubMedCrossRef
20.
go back to reference Pearson T, Greiner DL, Shultz LD (2008) Creation of “humanized” mice to study human immunity. Curr Protoc Immunol Chapter 15:Unit 15.21 Pearson T, Greiner DL, Shultz LD (2008) Creation of “humanized” mice to study human immunity. Curr Protoc Immunol Chapter 15:Unit 15.21
21.
go back to reference Jörns A, Rath KJ, Terbish T et al (2010) Diabetes prevention by immunomodulatory FTY720 treatment in the LEW.1AR1-iddm rat despite immune cell activation. Endocrinology 151:3555–3565PubMedCrossRef Jörns A, Rath KJ, Terbish T et al (2010) Diabetes prevention by immunomodulatory FTY720 treatment in the LEW.1AR1-iddm rat despite immune cell activation. Endocrinology 151:3555–3565PubMedCrossRef
22.
go back to reference MacMurray AJ, Moralejo DH, Kwitek AE et al (2002) Lymphopenia in the BB rat model of type 1 diabetes is due to a mutation in a novel immune-associated nucleotide (Ian)-related gene. Genome Res 12:1029–1039PubMedCrossRef MacMurray AJ, Moralejo DH, Kwitek AE et al (2002) Lymphopenia in the BB rat model of type 1 diabetes is due to a mutation in a novel immune-associated nucleotide (Ian)-related gene. Genome Res 12:1029–1039PubMedCrossRef
23.
go back to reference Sommandas V, Rutledge EA, van Yserloo B, Fuller J, Lernmark A, Drexhage HA (2007) Low-density cells isolated from the rat thymus resemble branched cortical macrophages and have a reduced capability of rescuing double-positive thymocytes from apoptosis in the BB-DP rat. J Leukoc Biol 82:869–876PubMedCrossRef Sommandas V, Rutledge EA, van Yserloo B, Fuller J, Lernmark A, Drexhage HA (2007) Low-density cells isolated from the rat thymus resemble branched cortical macrophages and have a reduced capability of rescuing double-positive thymocytes from apoptosis in the BB-DP rat. J Leukoc Biol 82:869–876PubMedCrossRef
24.
go back to reference Atkinson MA, Bluestone JA, Eisenbarth GS et al (2011) How does type 1 diabetes develop? the notion of homicide or beta-cell suicide revisited. Diabetes 60:1370–1379PubMedCrossRef Atkinson MA, Bluestone JA, Eisenbarth GS et al (2011) How does type 1 diabetes develop? the notion of homicide or beta-cell suicide revisited. Diabetes 60:1370–1379PubMedCrossRef
25.
go back to reference Reddy S, Chai RC, Rodrigues JA, Hsu TH, Robinson E (2008) Presence of residual beta cells and co-existing islet autoimmunity in the NOD mouse during longstanding diabetes: a combined histochemical and immunohistochemical study. J Mol Histol 39:25–36PubMedCrossRef Reddy S, Chai RC, Rodrigues JA, Hsu TH, Robinson E (2008) Presence of residual beta cells and co-existing islet autoimmunity in the NOD mouse during longstanding diabetes: a combined histochemical and immunohistochemical study. J Mol Histol 39:25–36PubMedCrossRef
26.
go back to reference Coppieters KT, Dotta F, Amirian N et al (2012) Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J Exp Med 209:51–60PubMedCentralPubMedCrossRef Coppieters KT, Dotta F, Amirian N et al (2012) Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J Exp Med 209:51–60PubMedCentralPubMedCrossRef
27.
go back to reference Lally FJ, Ratcliff H, Bone AJ (2001) Apoptosis and disease progression in the spontaneously diabetic BB/S rat. Diabetologia 44:320–324PubMedCrossRef Lally FJ, Ratcliff H, Bone AJ (2001) Apoptosis and disease progression in the spontaneously diabetic BB/S rat. Diabetologia 44:320–324PubMedCrossRef
29.
go back to reference Richardson SJ, Willcox A, Bone AJ, Morgan NG, Foulis AK (2011) Immunopathology of the human pancreas in type-I diabetes. Semin Immunopathol 33:9–21PubMedCrossRef Richardson SJ, Willcox A, Bone AJ, Morgan NG, Foulis AK (2011) Immunopathology of the human pancreas in type-I diabetes. Semin Immunopathol 33:9–21PubMedCrossRef
31.
go back to reference Carrero JA, Calderon B, Towfic F, Artyomov MN, Unanue ER (2013) Defining the transcriptional and cellular landscape of type 1 diabetes in the NOD mouse. PLoS One 8:e59701PubMedCentralPubMedCrossRef Carrero JA, Calderon B, Towfic F, Artyomov MN, Unanue ER (2013) Defining the transcriptional and cellular landscape of type 1 diabetes in the NOD mouse. PLoS One 8:e59701PubMedCentralPubMedCrossRef
32.
go back to reference Voorbij HA, Jeucken PH, Kabel PJ, de Haan M, Drexhage HA (1989) Dendritic cells and scavenger macrophages in pancreatic islets of prediabetic BB rats. Diabetes 38:1623–1629PubMedCrossRef Voorbij HA, Jeucken PH, Kabel PJ, de Haan M, Drexhage HA (1989) Dendritic cells and scavenger macrophages in pancreatic islets of prediabetic BB rats. Diabetes 38:1623–1629PubMedCrossRef
33.
go back to reference Akirav EM, Baquero MT, Opare-Addo LW et al (2011) Glucose and inflammation control islet vascular density and beta-cell function in NOD mice: control of islet vasculature and vascular endothelial growth factor by glucose. Diabetes 60:876–883PubMedCrossRef Akirav EM, Baquero MT, Opare-Addo LW et al (2011) Glucose and inflammation control islet vascular density and beta-cell function in NOD mice: control of islet vasculature and vascular endothelial growth factor by glucose. Diabetes 60:876–883PubMedCrossRef
34.
go back to reference Kleemann R, Scott FW, Worz-Pagenstert U, Nimal Ratnayake WM, Kolb H (1998) Impact of dietary fat on Th1/Th2 cytokine gene expression in the pancreas and gut of diabetes-prone BB rats. J Autoimmun 11:97–103PubMedCrossRef Kleemann R, Scott FW, Worz-Pagenstert U, Nimal Ratnayake WM, Kolb H (1998) Impact of dietary fat on Th1/Th2 cytokine gene expression in the pancreas and gut of diabetes-prone BB rats. J Autoimmun 11:97–103PubMedCrossRef
35.
go back to reference Mastrandrea L, Yu J, Behrens T et al (2009) Etanercept treatment in children with new-onset type 1 diabetes: pilot randomized, placebo-controlled, double-blind study. Diabetes Care 32:1244–1249PubMedCrossRef Mastrandrea L, Yu J, Behrens T et al (2009) Etanercept treatment in children with new-onset type 1 diabetes: pilot randomized, placebo-controlled, double-blind study. Diabetes Care 32:1244–1249PubMedCrossRef
36.
go back to reference Arif S, Cox P, Afzali B et al (2010) Anti-TNFalpha therapy–killing two birds with one stone? Lancet 375:2278PubMedCrossRef Arif S, Cox P, Afzali B et al (2010) Anti-TNFalpha therapy–killing two birds with one stone? Lancet 375:2278PubMedCrossRef
37.
go back to reference Kalden JR (2011) Anti-TNF therapy: what have we learned in 12 years? Arthritis Res Ther 13(Suppl. 1): S1 Kalden JR (2011) Anti-TNF therapy: what have we learned in 12 years? Arthritis Res Ther 13(Suppl. 1): S1
38.
go back to reference Wang F, Smith N, Maier L et al (2012) Etanercept suppresses regenerative hyperplasia in psoriasis by acutely downregulating epidermal expression of IL-19, IL-20 and IL-24. Br J Dermatol 167:92–102PubMedCrossRef Wang F, Smith N, Maier L et al (2012) Etanercept suppresses regenerative hyperplasia in psoriasis by acutely downregulating epidermal expression of IL-19, IL-20 and IL-24. Br J Dermatol 167:92–102PubMedCrossRef
39.
go back to reference Goldbach-Mansky R (2012) Immunology in clinic review series; focus on autoinflammatory diseases: update on monogenic autoinflammatory diseases: the role of interleukin (IL)-1 and an emerging role for cytokines beyond IL-1. Clin Exp Immunol 167:391–404PubMedCentralPubMedCrossRef Goldbach-Mansky R (2012) Immunology in clinic review series; focus on autoinflammatory diseases: update on monogenic autoinflammatory diseases: the role of interleukin (IL)-1 and an emerging role for cytokines beyond IL-1. Clin Exp Immunol 167:391–404PubMedCentralPubMedCrossRef
40.
go back to reference Ordas I, Mould DR, Feagan BG, Sandborn WJ (2012) Anti-TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics-based dosing paradigms. Clin Pharmacol Ther 91:635–646PubMedCrossRef Ordas I, Mould DR, Feagan BG, Sandborn WJ (2012) Anti-TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics-based dosing paradigms. Clin Pharmacol Ther 91:635–646PubMedCrossRef
41.
go back to reference Yokoi N, Komeda K, Wang HY et al (2002) Cblb is a major susceptibility gene for rat type 1 diabetes mellitus. Nat Genet 31:391–394PubMed Yokoi N, Komeda K, Wang HY et al (2002) Cblb is a major susceptibility gene for rat type 1 diabetes mellitus. Nat Genet 31:391–394PubMed
42.
go back to reference Bonner-Weir S, Li WC, Ouziel-Yahalom L, Guo L, Weir GC, Sharma A (2010) Beta-cell growth and regeneration: replication is only part of the story. Diabetes 59:2340–2348PubMedCrossRef Bonner-Weir S, Li WC, Ouziel-Yahalom L, Guo L, Weir GC, Sharma A (2010) Beta-cell growth and regeneration: replication is only part of the story. Diabetes 59:2340–2348PubMedCrossRef
43.
go back to reference Salpeter SJ, Klein AM, Huangfu D, Grimsby J, Dor Y (2010) Glucose and aging control the quiescence period that follows pancreatic beta cell replication. Development 137:3205–3213PubMedCrossRef Salpeter SJ, Klein AM, Huangfu D, Grimsby J, Dor Y (2010) Glucose and aging control the quiescence period that follows pancreatic beta cell replication. Development 137:3205–3213PubMedCrossRef
44.
go back to reference Rabinovitch A, Suarez-Pinzon WL (1998) Cytokines and their roles in pancreatic islet beta-cell destruction and insulin-dependent diabetes mellitus. Biochem Pharmacol 55:1139–1149PubMedCrossRef Rabinovitch A, Suarez-Pinzon WL (1998) Cytokines and their roles in pancreatic islet beta-cell destruction and insulin-dependent diabetes mellitus. Biochem Pharmacol 55:1139–1149PubMedCrossRef
45.
go back to reference Bergmann L, Kröncke KD, Suschek C, Kolb H, Kolb-Bachofen V (1992) Cytotoxic action of IL-1 beta against pancreatic islets is mediated via nitric oxide formation and is inhibited by NG-monomethyl-L-arginine. FEBS Lett 299:103–106PubMedCrossRef Bergmann L, Kröncke KD, Suschek C, Kolb H, Kolb-Bachofen V (1992) Cytotoxic action of IL-1 beta against pancreatic islets is mediated via nitric oxide formation and is inhibited by NG-monomethyl-L-arginine. FEBS Lett 299:103–106PubMedCrossRef
46.
go back to reference Kacheva S, Lenzen S, Gurgul-Convey E (2011) Differential effects of proinflammatory cytokines on cell death and ER stress in insulin-secreting INS1E cells and the involvement of nitric oxide. Cytokine 55:195–201PubMedCrossRef Kacheva S, Lenzen S, Gurgul-Convey E (2011) Differential effects of proinflammatory cytokines on cell death and ER stress in insulin-secreting INS1E cells and the involvement of nitric oxide. Cytokine 55:195–201PubMedCrossRef
47.
go back to reference Ortis F, Pirot P, Naamane N et al (2008) Induction of nuclear factor-kappaB and its downstream genes by TNF-alpha and IL-1beta has a pro-apoptotic role in pancreatic beta cells. Diabetologia 51:1213–1225PubMedCrossRef Ortis F, Pirot P, Naamane N et al (2008) Induction of nuclear factor-kappaB and its downstream genes by TNF-alpha and IL-1beta has a pro-apoptotic role in pancreatic beta cells. Diabetologia 51:1213–1225PubMedCrossRef
48.
go back to reference Sleater M, Diamond AS, Gill RG (2007) Islet allograft rejection by contact-dependent CD8+ T cells: perforin and FasL play alternate but obligatory roles. Am J Transplant 7:1927–1933PubMedCrossRef Sleater M, Diamond AS, Gill RG (2007) Islet allograft rejection by contact-dependent CD8+ T cells: perforin and FasL play alternate but obligatory roles. Am J Transplant 7:1927–1933PubMedCrossRef
49.
go back to reference Barthson J, Germano CM, Moore F et al (2011) Cytokines tumor necrosis factor-alpha and interferon-gamma induce pancreatic beta-cell apoptosis through STAT1-mediated Bim protein activation. J Biol Chem 286:39632–39643PubMedCrossRef Barthson J, Germano CM, Moore F et al (2011) Cytokines tumor necrosis factor-alpha and interferon-gamma induce pancreatic beta-cell apoptosis through STAT1-mediated Bim protein activation. J Biol Chem 286:39632–39643PubMedCrossRef
50.
go back to reference Walz M, Overbergh L, Mathieu C, Kolb H, Martin S (2002) A murine interleukin-4-Ig fusion protein regulates the expression of Th1- and Th2-specific cytokines in the pancreas of NOD mice. Horm Metab Res 34:561–569PubMedCrossRef Walz M, Overbergh L, Mathieu C, Kolb H, Martin S (2002) A murine interleukin-4-Ig fusion protein regulates the expression of Th1- and Th2-specific cytokines in the pancreas of NOD mice. Horm Metab Res 34:561–569PubMedCrossRef
Metadata
Title
Islet infiltration, cytokine expression and beta cell death in the NOD mouse, BB rat, Komeda rat, LEW.1AR1-iddm rat and humans with type 1 diabetes
Authors
Anne Jörns
Tanja Arndt
Andreas Meyer zu Vilsendorf
Jürgen Klempnauer
Dirk Wedekind
Hans-Jürgen Hedrich
Lorella Marselli
Piero Marchetti
Nagakatsu Harada
Yutaka Nakaya
Gen-Sheng Wang
Fraser W. Scott
Conny Gysemans
Chantal Mathieu
Sigurd Lenzen
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/s00125-013-3125-4

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.