Skip to main content
Top
Published in: Diabetologia 7/2016

Open Access 01-07-2016 | Article

Neonatal diabetes caused by a homozygous KCNJ11 mutation demonstrates that tiny changes in ATP sensitivity markedly affect diabetes risk

Authors: Natascia Vedovato, Edward Cliff, Peter Proks, Varadarajan Poovazhagi, Sarah E. Flanagan, Sian Ellard, Andrew T. Hattersley, Frances M. Ashcroft

Published in: Diabetologia | Issue 7/2016

Login to get access

Abstract

Aims/hypothesis

The pancreatic ATP-sensitive potassium (KATP) channel plays a pivotal role in linking beta cell metabolism to insulin secretion. Mutations in KATP channel genes can result in hypo- or hypersecretion of insulin, as in neonatal diabetes mellitus and congenital hyperinsulinism, respectively. To date, all patients affected by neonatal diabetes due to a mutation in the pore-forming subunit of the channel (Kir6.2, KCNJ11) are heterozygous for the mutation. Here, we report the first clinical case of neonatal diabetes caused by a homozygous KCNJ11 mutation.

Methods

A male patient was diagnosed with diabetes shortly after birth. At 5 months of age, genetic testing revealed he carried a homozygous KCNJ11 mutation, G324R, (Kir6.2-G324R) and he was successfully transferred to sulfonylurea therapy (0.2 mg kg−1 day−1). Neither heterozygous parent was affected. Functional properties of wild-type, heterozygous and homozygous mutant KATP channels were examined after heterologous expression in Xenopus oocytes.

Results

Functional studies indicated that the Kir6.2-G324R mutation reduces the channel ATP sensitivity but that the difference in ATP inhibition between homozygous and heterozygous channels is remarkably small. Nevertheless, the homozygous patient developed neonatal diabetes, whereas the heterozygous parents were, and remain, unaffected. Kir6.2-G324R channels were fully shut by the sulfonylurea tolbutamide, which explains why the patient’s diabetes was well controlled by sulfonylurea therapy.

Conclusions/interpretation

The data demonstrate that tiny changes in KATP channel activity can alter beta cell electrical activity and insulin secretion sufficiently to cause diabetes. They also aid our understanding of how the Kir6.2-E23K variant predisposes to type 2 diabetes.
Appendix
Available only for authorised users
Literature
1.
go back to reference Gloyn AL, Pearson ER, Antcliff JF et al (2004) Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 350:1838–1849CrossRefPubMed Gloyn AL, Pearson ER, Antcliff JF et al (2004) Activating mutations in the gene encoding the ATP-sensitive potassium-channel subunit Kir6.2 and permanent neonatal diabetes. N Engl J Med 350:1838–1849CrossRefPubMed
2.
go back to reference Hattersley AT, Ashcroft FM (2005) Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes 54:2503–2513CrossRefPubMed Hattersley AT, Ashcroft FM (2005) Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes 54:2503–2513CrossRefPubMed
3.
go back to reference Flanagan SE, Clauin S, Bellanné-Chantelot C et al (2009) 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 30:170–180CrossRefPubMed Flanagan SE, Clauin S, Bellanné-Chantelot C et al (2009) 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 30:170–180CrossRefPubMed
4.
go back to reference Yorifuji T, Nagashima K, Kurokawa K et al (2005) The C42R mutation in the Kir6.2 (KCNJ11) gene as a cause of transient neonatal diabetes, childhood diabetes, or later-onset, apparently type 2 diabetes mellitus. J Clin Endocrinol Metab 90:3174–3178CrossRefPubMed Yorifuji T, Nagashima K, Kurokawa K et al (2005) The C42R mutation in the Kir6.2 (KCNJ11) gene as a cause of transient neonatal diabetes, childhood diabetes, or later-onset, apparently type 2 diabetes mellitus. J Clin Endocrinol Metab 90:3174–3178CrossRefPubMed
5.
go back to reference D'Amato E, Tammaro P, Craig TJ et al (2008) Variable phenotypic spectrum of diabetes mellitus in a family carrying a novel KCNJ11 gene mutation. Diabet Med 25:651–656CrossRefPubMed D'Amato E, Tammaro P, Craig TJ et al (2008) Variable phenotypic spectrum of diabetes mellitus in a family carrying a novel KCNJ11 gene mutation. Diabet Med 25:651–656CrossRefPubMed
6.
go back to reference Liu L, Nagashima K, Yasuda T et al (2013) Mutations in KCNJ11 are associated with the development of autosomal dominant, early-onset type 2 diabetes. Diabetologia 56:2609–2618CrossRefPubMed Liu L, Nagashima K, Yasuda T et al (2013) Mutations in KCNJ11 are associated with the development of autosomal dominant, early-onset type 2 diabetes. Diabetologia 56:2609–2618CrossRefPubMed
7.
go back to reference Sakura H, Wat N, Horton V, Millns H, Turner RC, Ashcroft FM (1996) Sequence variations in the human Kir6.2 gene, a subunit of the ß-cell ATP-sensitive K-channel: no association with NIDDM in white Caucasian subjects or evidence of abnormal function when expressed in vitro. Diabetologia 39:1233–1237CrossRefPubMed Sakura H, Wat N, Horton V, Millns H, Turner RC, Ashcroft FM (1996) Sequence variations in the human Kir6.2 gene, a subunit of the ß-cell ATP-sensitive K-channel: no association with NIDDM in white Caucasian subjects or evidence of abnormal function when expressed in vitro. Diabetologia 39:1233–1237CrossRefPubMed
8.
go back to reference Gloyn AL, Weedon MN, Owen KR et al (2003) Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes 52:568–572CrossRefPubMed Gloyn AL, Weedon MN, Owen KR et al (2003) Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes 52:568–572CrossRefPubMed
9.
go back to reference Villareal DT, Koster JC, Robertson H et al (2009) Kir6.2 variant E23K increases ATP-sensitive K+ channel activity and is associated with impaired insulin release and enhanced insulin sensitivity in adults with normal glucose tolerance. Diabetes 58:1869–1878CrossRefPubMedPubMedCentral Villareal DT, Koster JC, Robertson H et al (2009) Kir6.2 variant E23K increases ATP-sensitive K+ channel activity and is associated with impaired insulin release and enhanced insulin sensitivity in adults with normal glucose tolerance. Diabetes 58:1869–1878CrossRefPubMedPubMedCentral
10.
go back to reference Ashcroft FM (2007) The Walter B. Cannon physiology in perspective lecture, 2007. ATP-sensitive K+ channels and disease: from molecule to malady. Am J Physiol Endocrinol Metab 293:E880–E889CrossRefPubMed Ashcroft FM (2007) The Walter B. Cannon physiology in perspective lecture, 2007. ATP-sensitive K+ channels and disease: from molecule to malady. Am J Physiol Endocrinol Metab 293:E880–E889CrossRefPubMed
11.
go back to reference Cook DL, Hales CN (1984) Intracellular ATP directly blocks K+ channels in pancreatic B cells. Nature 311:271–273CrossRefPubMed Cook DL, Hales CN (1984) Intracellular ATP directly blocks K+ channels in pancreatic B cells. Nature 311:271–273CrossRefPubMed
12.
go back to reference Nichols CG, Shyng SL, Nestorowicz A (1996) Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272:1785–1787CrossRefPubMed Nichols CG, Shyng SL, Nestorowicz A (1996) Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272:1785–1787CrossRefPubMed
13.
go back to reference Tucker SJ, Gribble FM, Zhao C, Trapp S, Ashcroft FM (1997) Truncation of Kir6.2 produces ATP-sensitive K+ channels in the absence of the sulphonylurea receptor. Nature 387:179–183CrossRefPubMed Tucker SJ, Gribble FM, Zhao C, Trapp S, Ashcroft FM (1997) Truncation of Kir6.2 produces ATP-sensitive K+ channels in the absence of the sulphonylurea receptor. Nature 387:179–183CrossRefPubMed
14.
go back to reference Schwanstecher C, Meyer U, Schwanstecher M (2002) KIR6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic beta-cell ATP-sensitive K+ channels. Diabetes 51:875–879CrossRefPubMed Schwanstecher C, Meyer U, Schwanstecher M (2002) KIR6.2 polymorphism predisposes to type 2 diabetes by inducing overactivity of pancreatic beta-cell ATP-sensitive K+ channels. Diabetes 51:875–879CrossRefPubMed
15.
go back to reference Riedel MJ, Steckley DC, Light PE (2005) Current status of the E23K Kir6.2 polymorphism: implications for type-2 diabetes. Hum Genet 116:133–145CrossRefPubMed Riedel MJ, Steckley DC, Light PE (2005) Current status of the E23K Kir6.2 polymorphism: implications for type-2 diabetes. Hum Genet 116:133–145CrossRefPubMed
16.
go back to reference Hamming KSC, Soliman D, Matemisz LC et al (2009) Coexpression of the type 2 diabetes susceptibility gene variants KCNJ11 E23K and ABCC8 S1369A alter the ATP and sulfonylurea sensitivities of the ATP-sensitive K+ channel. Diabetes 58:2419–2424CrossRefPubMedPubMedCentral Hamming KSC, Soliman D, Matemisz LC et al (2009) Coexpression of the type 2 diabetes susceptibility gene variants KCNJ11 E23K and ABCC8 S1369A alter the ATP and sulfonylurea sensitivities of the ATP-sensitive K+ channel. Diabetes 58:2419–2424CrossRefPubMedPubMedCentral
17.
go back to reference Gribble FM, Ashfield R, Ammälä C, Ashcroft FM (1997) Properties of cloned ATP-sensitive K+ currents expressed in Xenopus oocytes. J Physiol Lond 498:87–98CrossRefPubMedPubMedCentral Gribble FM, Ashfield R, Ammälä C, Ashcroft FM (1997) Properties of cloned ATP-sensitive K+ currents expressed in Xenopus oocytes. J Physiol Lond 498:87–98CrossRefPubMedPubMedCentral
18.
go back to reference Proks P, Antcliff JF, Lippiat J, Gloyn AL, Hattersley AT, Ashcroft FM (2004) Molecular basis of Kir6.2 mutations associated with neonatal diabetes or neonatal diabetes plus neurological features. Proc Natl Acad Sci U S A 101:17539–17544CrossRefPubMedPubMedCentral Proks P, Antcliff JF, Lippiat J, Gloyn AL, Hattersley AT, Ashcroft FM (2004) Molecular basis of Kir6.2 mutations associated with neonatal diabetes or neonatal diabetes plus neurological features. Proc Natl Acad Sci U S A 101:17539–17544CrossRefPubMedPubMedCentral
19.
go back to reference Tarasov AI, Welters HJ, Senkel S et al (2006) A Kir6.2 mutation causing neonatal diabetes impairs electrical activity and insulin secretion from INS-1 beta-cells. Diabetes 55:3075–3082CrossRefPubMed Tarasov AI, Welters HJ, Senkel S et al (2006) A Kir6.2 mutation causing neonatal diabetes impairs electrical activity and insulin secretion from INS-1 beta-cells. Diabetes 55:3075–3082CrossRefPubMed
20.
go back to reference Ashcroft FM, Rorsman P (2004) Type 2 diabetes mellitus: not quite exciting enough? Hum Mol Genet 13:R21–R31CrossRefPubMed Ashcroft FM, Rorsman P (2004) Type 2 diabetes mellitus: not quite exciting enough? Hum Mol Genet 13:R21–R31CrossRefPubMed
21.
go back to reference Antcliff JF, Haider S, Proks P, Sansom MSP, Ashcroft FM (2005) Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit. EMBO J 24:229–239CrossRefPubMedPubMedCentral Antcliff JF, Haider S, Proks P, Sansom MSP, Ashcroft FM (2005) Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit. EMBO J 24:229–239CrossRefPubMedPubMedCentral
22.
go back to reference Lodwick D, Rainbow RD, Rubaiy HN, Al Johi M, Vuister GW, Norman RI (2014) Sulfonylurea receptors regulate the channel pore in ATP-sensitive potassium channels via an intersubunit salt bridge. Biochem J 464:343–354CrossRefPubMed Lodwick D, Rainbow RD, Rubaiy HN, Al Johi M, Vuister GW, Norman RI (2014) Sulfonylurea receptors regulate the channel pore in ATP-sensitive potassium channels via an intersubunit salt bridge. Biochem J 464:343–354CrossRefPubMed
23.
go back to reference Tarasov AI, Girard CA, Larkin B et al (2007) Functional analysis of two Kir6.2 (KCNJ11) mutations, K170T and E322K, causing neonatal diabetes. Diabetes Obes Metab 9(Suppl 2):S46–S55CrossRef Tarasov AI, Girard CA, Larkin B et al (2007) Functional analysis of two Kir6.2 (KCNJ11) mutations, K170T and E322K, causing neonatal diabetes. Diabetes Obes Metab 9(Suppl 2):S46–S55CrossRef
24.
go back to reference Riedel MJ, Boora P, Steckley D, de Vries G, Light PE (2003) Kir6.2 polymorphisms sensitize beta-cell ATP-sensitive potassium channels to activation by acyl CoAs: a possible cellular mechanism for increased susceptibility to type 2 diabetes? Diabetes 52:2630–2635CrossRefPubMed Riedel MJ, Boora P, Steckley D, de Vries G, Light PE (2003) Kir6.2 polymorphisms sensitize beta-cell ATP-sensitive potassium channels to activation by acyl CoAs: a possible cellular mechanism for increased susceptibility to type 2 diabetes? Diabetes 52:2630–2635CrossRefPubMed
25.
go back to reference Schwanstecher C, Neugebauer B, Schulz M, Schwanstecher M (2002) The common single nucleotide polymorphism E23K in KIR6.2 sensitizes pancreatic beta-cell ATP-sensitive potassium channels toward activation through nucleoside diphosphates. Diabetes 51(Suppl 3):S363–S367CrossRefPubMed Schwanstecher C, Neugebauer B, Schulz M, Schwanstecher M (2002) The common single nucleotide polymorphism E23K in KIR6.2 sensitizes pancreatic beta-cell ATP-sensitive potassium channels toward activation through nucleoside diphosphates. Diabetes 51(Suppl 3):S363–S367CrossRefPubMed
26.
go back to reference Florez JC, Burtt N, de Bakker PIW et al (2004) Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53:1360–1368CrossRefPubMed Florez JC, Burtt N, de Bakker PIW et al (2004) Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53:1360–1368CrossRefPubMed
27.
go back to reference Fatehi M, Raja M, Carter C, Soliman D, Holt A, Light PE (2012) The ATP-sensitive K+ channel ABCC8 S1369A type 2 diabetes risk variant increases MgATPase activity. Diabetes 61:241–249CrossRefPubMed Fatehi M, Raja M, Carter C, Soliman D, Holt A, Light PE (2012) The ATP-sensitive K+ channel ABCC8 S1369A type 2 diabetes risk variant increases MgATPase activity. Diabetes 61:241–249CrossRefPubMed
Metadata
Title
Neonatal diabetes caused by a homozygous KCNJ11 mutation demonstrates that tiny changes in ATP sensitivity markedly affect diabetes risk
Authors
Natascia Vedovato
Edward Cliff
Peter Proks
Varadarajan Poovazhagi
Sarah E. Flanagan
Sian Ellard
Andrew T. Hattersley
Frances M. Ashcroft
Publication date
01-07-2016
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 7/2016
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-016-3964-x

Other articles of this Issue 7/2016

Diabetologia 7/2016 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.