Skip to main content
Top
Published in: Reviews in Endocrine and Metabolic Disorders 3/2010

01-09-2010

Permanent neonatal diabetes due to activating mutations in ABCC8 and KCNJ11

Authors: Emma L. Edghill, Sarah E. Flanagan, Sian Ellard

Published in: Reviews in Endocrine and Metabolic Disorders | Issue 3/2010

Login to get access

Abstract

The ATP-sensitive potassium (KATP) channel is composed of two subunits SUR1 and Kir6.2. The channel is key for glucose stimulated insulin release from the pancreatic beta cell. Activating mutations have been identified in the genes encoding these subunits, ABCC8 and KCNJ11, and account for approximately 40% of permanent neonatal diabetes cases. The majority of patients with a KATP mutation present with isolated diabetes however some have presented with the Developmental delay, Epilepsy and Neonatal Diabetes syndrome. This review focuses on mutations in the KATP channel which result in permanent neonatal diabetes, we review the clinical and functional effects as well as the implications for treatment.
Literature
1.
go back to reference Iafusco D et al. Permanent diabetes mellitus in the first year of life. Diabetologia. 2002;456:798–804. Iafusco D et al. Permanent diabetes mellitus in the first year of life. Diabetologia. 2002;456:798–804.
2.
go back to reference Edghill EL et al. HLA genotyping supports a nonautoimmune etiology in patients diagnosed with diabetes under the age of 6 months. Diabetes. 2006;556:1895–8.CrossRef Edghill EL et al. HLA genotyping supports a nonautoimmune etiology in patients diagnosed with diabetes under the age of 6 months. Diabetes. 2006;556:1895–8.CrossRef
3.
go back to reference Gardner RJ et al. An imprinted locus associated with transient neonatal diabetes mellitus. Hum Mol Genet. 2000;94:589–96.CrossRef Gardner RJ et al. An imprinted locus associated with transient neonatal diabetes mellitus. Hum Mol Genet. 2000;94:589–96.CrossRef
4.
go back to reference Flanagan SE et al. Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes. 2007;567:1930–7.CrossRef Flanagan SE et al. Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes. 2007;567:1930–7.CrossRef
5.
go back to reference Wiedemann B et al. Incidence of neonatal diabetes in Austria-calculation based on the Austrian Diabetes Register. Pediatr Diabetes. 2010;111:18–23.CrossRef Wiedemann B et al. Incidence of neonatal diabetes in Austria-calculation based on the Austrian Diabetes Register. Pediatr Diabetes. 2010;111:18–23.CrossRef
6.
go back to reference Slingerland AS et al. Referral rates for diagnostic testing support an incidence of permanent neonatal diabetes in three European countries of at least 1 in 260,000 live births. Diabetologia. 2009;528:1683–5.CrossRef Slingerland AS et al. Referral rates for diagnostic testing support an incidence of permanent neonatal diabetes in three European countries of at least 1 in 260,000 live births. Diabetologia. 2009;528:1683–5.CrossRef
7.
go back to reference Stanik J et al. Prevalence of permanent neonatal diabetes in Slovakia and successful replacement of insulin with sulfonylurea therapy in KCNJ11 and ABCC8 mutation carriers. J Clin Endocrinol Metab. 2007;924:1276–82.CrossRef Stanik J et al. Prevalence of permanent neonatal diabetes in Slovakia and successful replacement of insulin with sulfonylurea therapy in KCNJ11 and ABCC8 mutation carriers. J Clin Endocrinol Metab. 2007;924:1276–82.CrossRef
8.
go back to reference Greeley SA et al. Update in neonatal diabetes. Curr Opin Endocrinol Diabetes Obes. 2010;171:13–9. Greeley SA et al. Update in neonatal diabetes. Curr Opin Endocrinol Diabetes Obes. 2010;171:13–9.
9.
go back to reference Sellick GS et al. Mutations in PTF1A cause pancreatic and cerebellar agenesis. Nat Genet. 2004;3612:1301–5.CrossRef Sellick GS et al. Mutations in PTF1A cause pancreatic and cerebellar agenesis. Nat Genet. 2004;3612:1301–5.CrossRef
10.
go back to reference Senee V et al. Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism. Nat Genet. 2006;386:682–7.CrossRef Senee V et al. Mutations in GLIS3 are responsible for a rare syndrome with neonatal diabetes mellitus and congenital hypothyroidism. Nat Genet. 2006;386:682–7.CrossRef
11.
go back to reference Rubio-Cabezas O, et al. Homozygous mutations in NEUROD1 are responsible for a novel syndrome of permanent neonatal diabetes and neurological abnormalities. Diabetes. 2010;59:2326–31. Rubio-Cabezas O, et al. Homozygous mutations in NEUROD1 are responsible for a novel syndrome of permanent neonatal diabetes and neurological abnormalities. Diabetes. 2010;59:2326–31.
12.
go back to reference Stoffers DA et al. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet. 1997;151:106–10.CrossRef Stoffers DA et al. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet. 1997;151:106–10.CrossRef
13.
go back to reference Smith SB, et al. Rfx6 directs islet formation and insulin production in mice and humans. Nature. 2010;463:775–80. Smith SB, et al. Rfx6 directs islet formation and insulin production in mice and humans. Nature. 2010;463:775–80.
14.
go back to reference Stoy J et al. Insulin gene mutations as a cause of permanent neonatal diabetes. Proc Natl Acad Sci USA. 2007;10438:15040–4.CrossRef Stoy J et al. Insulin gene mutations as a cause of permanent neonatal diabetes. Proc Natl Acad Sci USA. 2007;10438:15040–4.CrossRef
15.
go back to reference Edghill EL et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;574:1034–42.CrossRef Edghill EL et al. Insulin mutation screening in 1,044 patients with diabetes: mutations in the INS gene are a common cause of neonatal diabetes but a rare cause of diabetes diagnosed in childhood or adulthood. Diabetes. 2008;574:1034–42.CrossRef
16.
go back to reference Garin I et al. Recessive mutations in the INS gene result in neonatal diabetes through reduced insulin biosynthesis. Proc Natl Acad Sci USA. 2010;1077:3105–10.CrossRef Garin I et al. Recessive mutations in the INS gene result in neonatal diabetes through reduced insulin biosynthesis. Proc Natl Acad Sci USA. 2010;1077:3105–10.CrossRef
17.
go back to reference Ashcroft FM et al. Stimulus-secretion coupling in pancreatic beta cells. J Cell Biochem. 1994;55(Suppl):54–65.CrossRefPubMed Ashcroft FM et al. Stimulus-secretion coupling in pancreatic beta cells. J Cell Biochem. 1994;55(Suppl):54–65.CrossRefPubMed
18.
go back to reference Thomas PM et al. Mutations in the sulfonylurea receptor gene in familial persistent hyperinsulinemic hypoglycemia of infancy. Science. 1995;2685209:426–9.CrossRef Thomas PM et al. Mutations in the sulfonylurea receptor gene in familial persistent hyperinsulinemic hypoglycemia of infancy. Science. 1995;2685209:426–9.CrossRef
19.
go back to reference Thomas P, Ye Y, Lightner E. Mutation of the pancreatic islet inward rectifier Kir6.2 also leads to familial persistent hyperinsulinemic hypoglycemia of infancy. Hum Mol Genet. 1996;511:1809–12.CrossRef Thomas P, Ye Y, Lightner E. Mutation of the pancreatic islet inward rectifier Kir6.2 also leads to familial persistent hyperinsulinemic hypoglycemia of infancy. Hum Mol Genet. 1996;511:1809–12.CrossRef
20.
go back to reference Koster JC et al. Targeted overactivity of beta cell KATP channels induces profound neonatal diabetes. Cell. 2000;1006:645–54.CrossRef Koster JC et al. Targeted overactivity of beta cell KATP channels induces profound neonatal diabetes. Cell. 2000;1006:645–54.CrossRef
21.
go back to reference Gloyn AL et al. Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med. 2004;35018:1838–49.CrossRef Gloyn AL et al. Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med. 2004;35018:1838–49.CrossRef
22.
go back to reference Proks P et al. A heterozygous activating mutation in the sulphonylurea receptor SUR1 ABCC8 causes neonatal diabetes. Hum Mol Genet. 2006;1511:1793–800.CrossRef Proks P et al. A heterozygous activating mutation in the sulphonylurea receptor SUR1 ABCC8 causes neonatal diabetes. Hum Mol Genet. 2006;1511:1793–800.CrossRef
23.
go back to reference Babenko AP et al. Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med. 2006;3555:456–66.CrossRef Babenko AP et al. Activating mutations in the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med. 2006;3555:456–66.CrossRef
24.
go back to reference Remedi MS, Koster JC. KATP channelopathies in the pancreas. Pflugers Arch. 2010;460:307–20. Remedi MS, Koster JC. KATP channelopathies in the pancreas. Pflugers Arch. 2010;460:307–20.
25.
go back to reference Flanagan SE et al. Update of mutations in the genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 KCNJ11 and sulfonylurea receptor 1 ABCC8 in diabetes mellitus and hyperinsulinism. Hum Mutat. 2009;302:170–80.CrossRef Flanagan SE et al. Update of mutations in the genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 KCNJ11 and sulfonylurea receptor 1 ABCC8 in diabetes mellitus and hyperinsulinism. Hum Mutat. 2009;302:170–80.CrossRef
26.
go back to reference Craig TJ, et al. An in-frame deletion in Kir6.2 KCNJ11 causing neonatal diabetes reveals a site of interaction between Kir6.2 and SUR1. J Clin Endocrinol Metab. 2009;26:2551–7. Craig TJ, et al. An in-frame deletion in Kir6.2 KCNJ11 causing neonatal diabetes reveals a site of interaction between Kir6.2 and SUR1. J Clin Endocrinol Metab. 2009;26:2551–7.
27.
go back to reference Ellard S et al. Permanent neonatal diabetes caused by dominant, recessive, or compound heterozygous SUR1 mutations with opposite functional effects. Am J Hum Genet. 2007;812:375–82.CrossRef Ellard S et al. Permanent neonatal diabetes caused by dominant, recessive, or compound heterozygous SUR1 mutations with opposite functional effects. Am J Hum Genet. 2007;812:375–82.CrossRef
28.
go back to reference Sagen JV et al. Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes. 2004;5310:2713–8.CrossRef Sagen JV et al. Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes. 2004;5310:2713–8.CrossRef
29.
go back to reference Vaxillaire M et al. Kir6.2 mutations are a common cause of permanent neonatal diabetes in a large cohort of French patients. Diabetes. 2004;5310:2719–22.CrossRef Vaxillaire M et al. Kir6.2 mutations are a common cause of permanent neonatal diabetes in a large cohort of French patients. Diabetes. 2004;5310:2719–22.CrossRef
30.
go back to reference Massa O et al. KCNJ11 activating mutations in Italian patients with permanent neonatal diabetes. Hum Mutat. 2005;251:22–7.CrossRef Massa O et al. KCNJ11 activating mutations in Italian patients with permanent neonatal diabetes. Hum Mutat. 2005;251:22–7.CrossRef
31.
go back to reference Flanagan SE et al. Mutations in KCNJ11, which encodes Kir6.2, are a common cause of diabetes diagnosed in the first 6 months of life, with the phenotype determined by genotype. Diabetologia. 2006;496:1190–7.CrossRef Flanagan SE et al. Mutations in KCNJ11, which encodes Kir6.2, are a common cause of diabetes diagnosed in the first 6 months of life, with the phenotype determined by genotype. Diabetologia. 2006;496:1190–7.CrossRef
32.
go back to reference Gloyn AL et al. Permanent neonatal diabetes due to paternal germline mosaicism for an activating mutation of the KCNJ11 Gene encoding the Kir6.2 subunit of the beta-cell potassium adenosine triphosphate channel. J Clin Endocrinol Metab. 2004;898:3932–5.CrossRef Gloyn AL et al. Permanent neonatal diabetes due to paternal germline mosaicism for an activating mutation of the KCNJ11 Gene encoding the Kir6.2 subunit of the beta-cell potassium adenosine triphosphate channel. J Clin Endocrinol Metab. 2004;898:3932–5.CrossRef
33.
go back to reference Edghill EL et al. Origin of de novo KCNJ11 mutations and risk of neonatal diabetes for subsequent siblings. J Clin Endocrinol Metab. 2007;925:1773–7.CrossRef Edghill EL et al. Origin of de novo KCNJ11 mutations and risk of neonatal diabetes for subsequent siblings. J Clin Endocrinol Metab. 2007;925:1773–7.CrossRef
34.
go back to reference Pearson ER et al. Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med. 2006;3555:467–77.CrossRef Pearson ER et al. Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med. 2006;3555:467–77.CrossRef
35.
go back to reference Mohamadi A, et al. Medical and developmental impact of transition from subcutaneous insulin to oral glyburide in a 15-yr-old boy with neonatal diabetes mellitus and intermediate DEND syndrome: extending the age of KCNJ11 mutation testing in neonatal DM. Pediatr Diabetes. 2010;11:203-11. Mohamadi A, et al. Medical and developmental impact of transition from subcutaneous insulin to oral glyburide in a 15-yr-old boy with neonatal diabetes mellitus and intermediate DEND syndrome: extending the age of KCNJ11 mutation testing in neonatal DM. Pediatr Diabetes. 2010;11:203-11.
36.
go back to reference Masia R et al. An ATP-binding mutation G334D in KCNJ11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes. 2007;562:328–36.CrossRef Masia R et al. An ATP-binding mutation G334D in KCNJ11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes. 2007;562:328–36.CrossRef
37.
go back to reference Sakura H et al. Cloning and functional expression of the cDNA encoding a novel ATP-sensitive potassium channel subunit expressed in pancreatic beta-cells, brain, heart and skeletal muscle. FEBS Lett. 1995;3773:338–44.CrossRef Sakura H et al. Cloning and functional expression of the cDNA encoding a novel ATP-sensitive potassium channel subunit expressed in pancreatic beta-cells, brain, heart and skeletal muscle. FEBS Lett. 1995;3773:338–44.CrossRef
38.
go back to reference Hattersley AT, Ashcroft FM. Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes. 2005;549:2503–13.CrossRef Hattersley AT, Ashcroft FM. Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes. 2005;549:2503–13.CrossRef
39.
go back to reference Patch AM et al. Mutations in the ABCC8 gene encoding the SUR1 subunit of the KATP channel cause transient neonatal diabetes, permanent neonatal diabetes or permanent diabetes diagnosed outside the neonatal period. Diabetes Obes Metab. 2007;9 Suppl 2:28–39.CrossRefPubMed Patch AM et al. Mutations in the ABCC8 gene encoding the SUR1 subunit of the KATP channel cause transient neonatal diabetes, permanent neonatal diabetes or permanent diabetes diagnosed outside the neonatal period. Diabetes Obes Metab. 2007;9 Suppl 2:28–39.CrossRefPubMed
40.
go back to reference Shimomura K et al. Mutations at the same residue R50 of Kir6.2 KCNJ11 that cause neonatal diabetes produce different functional effects. Diabetes. 2006;556:1705–12.CrossRef Shimomura K et al. Mutations at the same residue R50 of Kir6.2 KCNJ11 that cause neonatal diabetes produce different functional effects. Diabetes. 2006;556:1705–12.CrossRef
41.
go back to reference Tammaro P et al. A Kir6.2 mutation causing severe functional effects in vitro produces neonatal diabetes without the expected neurological complications. Diabetologia. 2008;515:802–10.CrossRef Tammaro P et al. A Kir6.2 mutation causing severe functional effects in vitro produces neonatal diabetes without the expected neurological complications. Diabetologia. 2008;515:802–10.CrossRef
42.
go back to reference Shimomura K et al. Adjacent mutations in the gating loop of Kir6.2 produce neonatal diabetes and hyperinsulinism. EMBO. Mol Med. 2009;13:166–77. Shimomura K et al. Adjacent mutations in the gating loop of Kir6.2 produce neonatal diabetes and hyperinsulinism. EMBO. Mol Med. 2009;13:166–77.
43.
go back to reference Proks P et al. Molecular basis of Kir6.2 mutations associated with neonatal diabetes or neonatal diabetes plus neurological features. Proc Natl Acad Sci USA. 2004;10150:17539–44.CrossRef Proks P et al. Molecular basis of Kir6.2 mutations associated with neonatal diabetes or neonatal diabetes plus neurological features. Proc Natl Acad Sci USA. 2004;10150:17539–44.CrossRef
44.
go back to reference Proks P et al. A gating mutation at the internal mouth of the Kir6.2 pore is associated with DEND syndrome. EMBO Rep. 2005;65:470–5.CrossRef Proks P et al. A gating mutation at the internal mouth of the Kir6.2 pore is associated with DEND syndrome. EMBO Rep. 2005;65:470–5.CrossRef
45.
go back to reference Zingman LV et al. Tandem function of nucleotide binding domains confers competence to sulfonylurea receptor in gating ATP-sensitive K+ channels. J Biol Chem. 2002;27716:14206–10.CrossRef Zingman LV et al. Tandem function of nucleotide binding domains confers competence to sulfonylurea receptor in gating ATP-sensitive K+ channels. J Biol Chem. 2002;27716:14206–10.CrossRef
46.
go back to reference Aittoniemi J et al. Review. SUR1: a unique ATP-binding cassette protein that functions as an ion channel regulator. Philos Trans R Soc Lond B Biol Sci. 2009;3641514:257–67. Aittoniemi J et al. Review. SUR1: a unique ATP-binding cassette protein that functions as an ion channel regulator. Philos Trans R Soc Lond B Biol Sci. 2009;3641514:257–67.
47.
go back to reference de Wet H et al. Increased ATPase activity produced by mutations at arginine-1380 in nucleotide-binding domain 2 of ABCC8 causes neonatal diabetes. Proc Natl Acad Sci USA. 2007;10448:18988–92.CrossRef de Wet H et al. Increased ATPase activity produced by mutations at arginine-1380 in nucleotide-binding domain 2 of ABCC8 causes neonatal diabetes. Proc Natl Acad Sci USA. 2007;10448:18988–92.CrossRef
48.
go back to reference de Wet H et al. A mutation R826W in nucleotide-binding domain 1 of ABCC8 reduces ATPase activity and causes transient neonatal diabetes. EMBO Rep. 2008;97:648–54.CrossRef de Wet H et al. A mutation R826W in nucleotide-binding domain 1 of ABCC8 reduces ATPase activity and causes transient neonatal diabetes. EMBO Rep. 2008;97:648–54.CrossRef
49.
go back to reference Proks P et al. Mechanism of action of a sulphonylurea receptor SUR1 mutation F132L that causes DEND syndrome. Hum Mol Genet. 2007;1616:2011–9.CrossRef Proks P et al. Mechanism of action of a sulphonylurea receptor SUR1 mutation F132L that causes DEND syndrome. Hum Mol Genet. 2007;1616:2011–9.CrossRef
50.
go back to reference Girard CA et al. Expression of an activating mutation in the gene encoding the KATP channel subunit Kir6.2 in mouse pancreatic beta cells recapitulates neonatal diabetes. J Clin Invest. 2009;1191:80–90. Girard CA et al. Expression of an activating mutation in the gene encoding the KATP channel subunit Kir6.2 in mouse pancreatic beta cells recapitulates neonatal diabetes. J Clin Invest. 2009;1191:80–90.
51.
go back to reference Clark RH, et al. Muscle dysfunction caused by a KATP channel mutation in neonatal diabetes is neuronal in origin. Science. 2010;329:458–61. Clark RH, et al. Muscle dysfunction caused by a KATP channel mutation in neonatal diabetes is neuronal in origin. Science. 2010;329:458–61.
52.
go back to reference Rafiq M et al. Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 SUR1 mutations. Diab Care. 2008;312:204–9. Rafiq M et al. Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 SUR1 mutations. Diab Care. 2008;312:204–9.
53.
go back to reference Gribble FM, Reimann F. Sulphonylurea action revisited: the post-cloning era. Diabetologia. 2003;467:875–91.CrossRef Gribble FM, Reimann F. Sulphonylurea action revisited: the post-cloning era. Diabetologia. 2003;467:875–91.CrossRef
54.
go back to reference Codner E et al. High-dose glibenclamide can replace insulin therapy despite transitory diarrhea in early-onset diabetes caused by a novel R201L Kir6.2 mutation. Diab Care. 2005;283:758–9.CrossRef Codner E et al. High-dose glibenclamide can replace insulin therapy despite transitory diarrhea in early-onset diabetes caused by a novel R201L Kir6.2 mutation. Diab Care. 2005;283:758–9.CrossRef
55.
go back to reference Kumaraguru J et al. Tooth discoloration in patients with neonatal diabetes after transfer onto glibenclamide: a previously unreported side effect. Diab Care. 2009;328:1428–30.CrossRef Kumaraguru J et al. Tooth discoloration in patients with neonatal diabetes after transfer onto glibenclamide: a previously unreported side effect. Diab Care. 2009;328:1428–30.CrossRef
56.
go back to reference Zung A et al. Glibenclamide treatment in permanent neonatal diabetes mellitus due to an activating mutation in Kir6.2. J Clin Endocrinol Metab. 2004;8911:5504–7.CrossRef Zung A et al. Glibenclamide treatment in permanent neonatal diabetes mellitus due to an activating mutation in Kir6.2. J Clin Endocrinol Metab. 2004;8911:5504–7.CrossRef
57.
go back to reference Slingerland AS et al. Improved motor development and good long-term glycaemic control with sulfonylurea treatment in a patient with the syndrome of intermediate developmental delay, early-onset generalised epilepsy and neonatal diabetes associated with the V59M mutation in the KCNJ11 gene. Diabetologia. 2006;4911:2559–63.CrossRef Slingerland AS et al. Improved motor development and good long-term glycaemic control with sulfonylurea treatment in a patient with the syndrome of intermediate developmental delay, early-onset generalised epilepsy and neonatal diabetes associated with the V59M mutation in the KCNJ11 gene. Diabetologia. 2006;4911:2559–63.CrossRef
58.
go back to reference Slingerland AS et al. Sulphonylurea therapy improves cognition in a patient with the V59M KCNJ11 mutation. Diabet Med. 2008;253:277–81.CrossRef Slingerland AS et al. Sulphonylurea therapy improves cognition in a patient with the V59M KCNJ11 mutation. Diabet Med. 2008;253:277–81.CrossRef
59.
go back to reference Shimomura K et al. A novel mutation causing DEND syndrome: a treatable channelopathy of pancreas and brain. Neurology. 2007;6913:1342–9.CrossRef Shimomura K et al. A novel mutation causing DEND syndrome: a treatable channelopathy of pancreas and brain. Neurology. 2007;6913:1342–9.CrossRef
60.
go back to reference Gurgel LC et al. Sulfonylrea treatment in permanent neonatal diabetes due to G53D mutation in the KCNJ11 gene: improvement in glycemic control and neurological function. Diab Care. 2007;3011:e108.CrossRef Gurgel LC et al. Sulfonylrea treatment in permanent neonatal diabetes due to G53D mutation in the KCNJ11 gene: improvement in glycemic control and neurological function. Diab Care. 2007;3011:e108.CrossRef
Metadata
Title
Permanent neonatal diabetes due to activating mutations in ABCC8 and KCNJ11
Authors
Emma L. Edghill
Sarah E. Flanagan
Sian Ellard
Publication date
01-09-2010
Publisher
Springer US
Published in
Reviews in Endocrine and Metabolic Disorders / Issue 3/2010
Print ISSN: 1389-9155
Electronic ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-010-9149-x

Other articles of this Issue 3/2010

Reviews in Endocrine and Metabolic Disorders 3/2010 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