Skip to main content
Top
Published in: Diabetologia 5/2008

Open Access 01-05-2008 | Article

A Kir6.2 mutation causing severe functional effects in vitro produces neonatal diabetes without the expected neurological complications

Authors: P. Tammaro, S. E. Flanagan, B. Zadek, S. Srinivasan, H. Woodhead, S. Hameed, I. Klimes, A. T. Hattersley, S. Ellard, F. M. Ashcroft

Published in: Diabetologia | Issue 5/2008

Login to get access

Abstract

Aims/hypothesis

Heterozygous activating mutations in the pancreatic ATP-sensitive K+ channel cause permanent neonatal diabetes mellitus (PNDM). This results from a decrease in the ability of ATP to close the channel, which thereby suppresses insulin secretion. PNDM mutations that cause a severe reduction in ATP inhibition may produce additional symptoms such as developmental delay and epilepsy. We identified a heterozygous mutation (L164P) in the pore-forming (Kir6.2) subunit of the channel in three unrelated patients and examined its functional effects.

Methods

The patients (currently aged 2, 8 and 20 years) developed diabetes shortly after birth. The two younger patients attempted transfer to sulfonylurea therapy but were unsuccessful (up to 1.1 mg kg−1 day−1). They remain insulin dependent. None of the patients displayed neurological symptoms. Functional properties of wild-type and mutant channels were examined by electrophysiology in Xenopus oocytes.

Results

Heterozygous (het) and homozygous L164P KATP channels showed a marked reduction in channel inhibition by ATP. Consistent with its predicted location within the pore, L164P enhanced the channel open state, which explains the reduction in ATP sensitivity. HetL164P currents exhibited greatly increased whole-cell currents that were unaffected by sulfonylureas. This explains the inability of sulfonylureas to ameliorate the diabetes of affected patients.

Conclusions/interpretation

Our results provide the first demonstration that mutations such as L164P, which produce a severe reduction in ATP sensitivity, do not inevitably cause developmental delay or neurological problems. However, the neonatal diabetes of these patients is unresponsive to sulfonylurea therapy. Functional analysis of PNDM mutations can predict the sulfonylurea response.
Literature
1.
go back to reference Seino S, Miki T (2003) Physiological and pathophysiological roles of ATP-sensitive K+ channels. Prog Biophys Mol Biol 81:133–176PubMedCrossRef Seino S, Miki T (2003) Physiological and pathophysiological roles of ATP-sensitive K+ channels. Prog Biophys Mol Biol 81:133–176PubMedCrossRef
2.
go back to reference Ashcroft FM, Harrison DE, Ashcroft SJ (1984) Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature 312:446–448PubMedCrossRef Ashcroft FM, Harrison DE, Ashcroft SJ (1984) Glucose induces closure of single potassium channels in isolated rat pancreatic beta-cells. Nature 312:446–448PubMedCrossRef
3.
go back to reference Ashcroft F, Rorsman P (2004) Type 2 diabetes mellitus: not quite exciting enough? Hum Mol Genet 13(Spec No. 1):R21–R31PubMedCrossRef Ashcroft F, Rorsman P (2004) Type 2 diabetes mellitus: not quite exciting enough? Hum Mol Genet 13(Spec No. 1):R21–R31PubMedCrossRef
4.
go back to reference Inagaki N, Gonoi T, Clement JPT et al (1995) Reconstitution of IKATP: an inward rectifier subunit plus the sulfonylurea receptor. Science 270:1166–1170PubMedCrossRef Inagaki N, Gonoi T, Clement JPT et al (1995) Reconstitution of IKATP: an inward rectifier subunit plus the sulfonylurea receptor. Science 270:1166–1170PubMedCrossRef
5.
go back to reference Clement JPT, Kunjilwar K, Gonzalez G et al (1997) Association and stoichiometry of KATP channel subunits. Neuron 18:827–838PubMedCrossRef Clement JPT, Kunjilwar K, Gonzalez G et al (1997) Association and stoichiometry of KATP channel subunits. Neuron 18:827–838PubMedCrossRef
6.
go back to reference Shyng S, Nichols CG (1997) Octameric stoichiometry of the KATP channel complex. J Gen Physiol 110:655–664PubMedCrossRef Shyng S, Nichols CG (1997) Octameric stoichiometry of the KATP channel complex. J Gen Physiol 110:655–664PubMedCrossRef
7.
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 387:179–183PubMedCrossRef 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 387:179–183PubMedCrossRef
8.
go back to reference Drain P, Li L, Wang J (1998) KATP channel inhibition by ATP requires distinct functional domains of the cytoplasmic C terminus of the pore-forming subunit. Proc Natl Acad Sci U S A 95:13953–13958PubMedCrossRef Drain P, Li L, Wang J (1998) KATP channel inhibition by ATP requires distinct functional domains of the cytoplasmic C terminus of the pore-forming subunit. Proc Natl Acad Sci U S A 95:13953–13958PubMedCrossRef
9.
go back to reference Haider S, Tarasov AI, Craig TJ, Sansom MS, Ashcroft FM (2007) Identification of the PIP(2)-binding site on Kir6.2 by molecular modelling and functional analysis. EMBO J 26:3749–3759PubMedCrossRef Haider S, Tarasov AI, Craig TJ, Sansom MS, Ashcroft FM (2007) Identification of the PIP(2)-binding site on Kir6.2 by molecular modelling and functional analysis. EMBO J 26:3749–3759PubMedCrossRef
10.
go back to reference Nichols CG, Shyng SL, Nestorowicz A et al (1996) Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272:1785–1787PubMedCrossRef Nichols CG, Shyng SL, Nestorowicz A et al (1996) Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272:1785–1787PubMedCrossRef
11.
go back to reference Gribble FM, Tucker SJ, Ashcroft FM (1997) The essential role of the Walker A motifs of SUR1 in KATP channel activation by Mg-ADP and diazoxide. EMBO J 16:1145–1152PubMedCrossRef Gribble FM, Tucker SJ, Ashcroft FM (1997) The essential role of the Walker A motifs of SUR1 in KATP channel activation by Mg-ADP and diazoxide. EMBO J 16:1145–1152PubMedCrossRef
12.
go back to reference Shyng S, Ferrigni T, Nichols CG (1997) Regulation of KATP channel activity by diazoxide and MgADP. Distinct functions of the two nucleotide binding folds of the sulfonylurea receptor. J Gen Physiol 110:643–654PubMedCrossRef Shyng S, Ferrigni T, Nichols CG (1997) Regulation of KATP channel activity by diazoxide and MgADP. Distinct functions of the two nucleotide binding folds of the sulfonylurea receptor. J Gen Physiol 110:643–654PubMedCrossRef
13.
go back to reference Zingman LV, Alekseev AE, Bienengraeber M et al (2001) Signaling in channel/enzyme multimers: ATPase transitions in SUR module gate ATP-sensitive K+ conductance. Neuron 31:233–245PubMedCrossRef Zingman LV, Alekseev AE, Bienengraeber M et al (2001) Signaling in channel/enzyme multimers: ATPase transitions in SUR module gate ATP-sensitive K+ conductance. Neuron 31:233–245PubMedCrossRef
14.
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 350:1838–1849PubMedCrossRef 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 350:1838–1849PubMedCrossRef
15.
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 101:17539–17544PubMedCrossRef 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 101:17539–17544PubMedCrossRef
16.
go back to reference Proks P, Girard C, Haider S et al (2005) A gating mutation at the internal mouth of the Kir6.2 pore is associated with DEND syndrome. EMBO Rep 6:470–475PubMedCrossRef Proks P, Girard C, Haider S et al (2005) A gating mutation at the internal mouth of the Kir6.2 pore is associated with DEND syndrome. EMBO Rep 6:470–475PubMedCrossRef
17.
go back to reference Flanagan SE, Patch AM, Mackay DJ et al (2007) Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes 56:1930–1937PubMedCrossRef Flanagan SE, Patch AM, Mackay DJ et al (2007) Mutations in ATP-sensitive K+ channel genes cause transient neonatal diabetes and permanent diabetes in childhood or adulthood. Diabetes 56:1930–1937PubMedCrossRef
18.
go back to reference Gloyn AL, Reimann F, Girard C et al (2005) Relapsing diabetes can result from moderately activating mutations in KCNJ11. Hum Mol Genet 14:925–934PubMedCrossRef Gloyn AL, Reimann F, Girard C et al (2005) Relapsing diabetes can result from moderately activating mutations in KCNJ11. Hum Mol Genet 14:925–934PubMedCrossRef
19.
go back to reference Massa O, Iafusco D, D, Amato E et al (2005) KCNJ11 activating mutations in Italian patients with permanent neonatal diabetes. Hum Mutat 25:22–27PubMedCrossRef Massa O, Iafusco D, D, Amato E et al (2005) KCNJ11 activating mutations in Italian patients with permanent neonatal diabetes. Hum Mutat 25:22–27PubMedCrossRef
20.
go back to reference Sagen JV, Raeder H, Hathout E et al (2004) Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 53:2713–2718PubMedCrossRef Sagen JV, Raeder H, Hathout E et al (2004) Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 53:2713–2718PubMedCrossRef
21.
go back to reference Ashcroft FM (2005) ATP-sensitive potassium channelopathies: focus on insulin secretion. J Clin Invest 115:2047–2058PubMedCrossRef Ashcroft FM (2005) ATP-sensitive potassium channelopathies: focus on insulin secretion. J Clin Invest 115:2047–2058PubMedCrossRef
22.
go back to reference Zung A, Glaser B, Nimri R, Zadik Z (2004) Glibenclamide treatment in permanent neonatal diabetes mellitus due to an activating mutation in Kir6.2. J Clin Endocrinol Metab 89:5504–5507PubMedCrossRef Zung A, Glaser B, Nimri R, Zadik Z (2004) Glibenclamide treatment in permanent neonatal diabetes mellitus due to an activating mutation in Kir6.2. J Clin Endocrinol Metab 89:5504–5507PubMedCrossRef
23.
go back to reference Gribble FM, Reimann F (2003) Sulphonylurea action revisited: the post-cloning era. Diabetologia 46:875–891PubMedCrossRef Gribble FM, Reimann F (2003) Sulphonylurea action revisited: the post-cloning era. Diabetologia 46:875–891PubMedCrossRef
24.
go back to reference Proks P, Girard C, Ashcroft FM (2005) Functional effects of KCNJ11 mutations causing neonatal diabetes: enhanced activation by MgATP. Hum Mol Genet 14:2717–2726PubMedCrossRef Proks P, Girard C, Ashcroft FM (2005) Functional effects of KCNJ11 mutations causing neonatal diabetes: enhanced activation by MgATP. Hum Mol Genet 14:2717–2726PubMedCrossRef
25.
go back to reference Flanagan SE, Edghill EL, Gloyn AL, Ellard S, Hattersley AT (2006) 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 49:1190–1197PubMedCrossRef Flanagan SE, Edghill EL, Gloyn AL, Ellard S, Hattersley AT (2006) 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 49:1190–1197PubMedCrossRef
26.
go back to reference Tammaro P, Girard C, Molnes J, Njolstad PR, Ashcroft FM (2005) Kir6.2 mutations causing neonatal diabetes provide new insights into Kir6.2-SUR1 interactions. EMBO J 24:2318–2330PubMedCrossRef Tammaro P, Girard C, Molnes J, Njolstad PR, Ashcroft FM (2005) Kir6.2 mutations causing neonatal diabetes provide new insights into Kir6.2-SUR1 interactions. EMBO J 24:2318–2330PubMedCrossRef
27.
go back to reference Stanik J, Gasperikova D, Paskova M et al (2007) 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 92:1276–1282PubMedCrossRef Stanik J, Gasperikova D, Paskova M et al (2007) 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 92:1276–1282PubMedCrossRef
28.
go back to reference Pearson ER, Flechtner I, Njolstad PR et al (2006) Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 355:467–477PubMedCrossRef Pearson ER, Flechtner I, Njolstad PR et al (2006) Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 355:467–477PubMedCrossRef
29.
go back to reference Gribble FM, Ashfield R, Ammala C, Ashcroft FM (1997) Properties of cloned ATP-sensitive K+ currents expressed in Xenopus oocytes. J Physiol 498:87–98PubMed Gribble FM, Ashfield R, Ammala C, Ashcroft FM (1997) Properties of cloned ATP-sensitive K+ currents expressed in Xenopus oocytes. J Physiol 498:87–98PubMed
30.
go back to reference Enkvetchakul D, Loussouarn G, Makhina E, Shyng SL, Nichols CG (2000) The kinetic and physical basis of KATP channel gating: toward a unified molecular understanding. Biophys J 78:2334–2348PubMed Enkvetchakul D, Loussouarn G, Makhina E, Shyng SL, Nichols CG (2000) The kinetic and physical basis of KATP channel gating: toward a unified molecular understanding. Biophys J 78:2334–2348PubMed
31.
go back to reference Trapp S, Proks P, Tucker SJ, Ashcroft FM (1998) Molecular analysis of ATP-sensitive K channel gating and implications for channel inhibition by ATP. J Gen Physiol 112:333–349PubMedCrossRef Trapp S, Proks P, Tucker SJ, Ashcroft FM (1998) Molecular analysis of ATP-sensitive K channel gating and implications for channel inhibition by ATP. J Gen Physiol 112:333–349PubMedCrossRef
32.
go back to reference Antcliff JF, Haider S, Proks P, Sansom MS, Ashcroft FM (2005) Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit. EMBO J 24:229–239PubMedCrossRef Antcliff JF, Haider S, Proks P, Sansom MS, Ashcroft FM (2005) Functional analysis of a structural model of the ATP-binding site of the KATP channel Kir6.2 subunit. EMBO J 24:229–239PubMedCrossRef
33.
go back to reference Shimomura K, Girard CA, Proks P et al (2006) Mutations at the same residue (R50) of Kir6.2 (KCNJ11) that cause neonatal diabetes produce different functional effects. Diabetes 55:1705–1712PubMedCrossRef Shimomura K, Girard CA, Proks P et al (2006) Mutations at the same residue (R50) of Kir6.2 (KCNJ11) that cause neonatal diabetes produce different functional effects. Diabetes 55:1705–1712PubMedCrossRef
34.
go back to reference Masia R, Koster JC, Tumini S et al (2007) An ATP-binding mutation (G334D) in KCNJ11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes 56:328–336PubMedCrossRef Masia R, Koster JC, Tumini S et al (2007) An ATP-binding mutation (G334D) in KCNJ11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes 56:328–336PubMedCrossRef
35.
go back to reference Haider S, Antcliff JF, Proks P, Sansom MS, Ashcroft FM (2005) Focus on Kir6.2: a key component of the ATP-sensitive potassium channel. J Mol Cell Cardiol 38:927–936PubMedCrossRef Haider S, Antcliff JF, Proks P, Sansom MS, Ashcroft FM (2005) Focus on Kir6.2: a key component of the ATP-sensitive potassium channel. J Mol Cell Cardiol 38:927–936PubMedCrossRef
36.
go back to reference Loussouarn G, Makhina EN, Rose T, Nichols CG (2000) Structure and dynamics of the pore of inwardly rectifying KATP channels. J Biol Chem 275:1137–1144PubMedCrossRef Loussouarn G, Makhina EN, Rose T, Nichols CG (2000) Structure and dynamics of the pore of inwardly rectifying KATP channels. J Biol Chem 275:1137–1144PubMedCrossRef
37.
go back to reference Loussouarn G, Phillips LR, Masia R, Rose T, Nichols CG (2001) Flexibility of the Kir6.2 inward rectifier K+ channel pore. Proc Natl Acad Sci U S A 98:4227–4232PubMedCrossRef Loussouarn G, Phillips LR, Masia R, Rose T, Nichols CG (2001) Flexibility of the Kir6.2 inward rectifier K+ channel pore. Proc Natl Acad Sci U S A 98:4227–4232PubMedCrossRef
38.
go back to reference Cordes FS, Bright JN, Sansom MS (2002) Proline-induced distortions of transmembrane helices. J Mol Biol 323:951–960PubMedCrossRef Cordes FS, Bright JN, Sansom MS (2002) Proline-induced distortions of transmembrane helices. J Mol Biol 323:951–960PubMedCrossRef
39.
go back to reference Enkvetchakul D, Loussouarn G, Makhina E, Nichols CG (2001) ATP interaction with the open state of the KATP channel. Biophys J 80:719–728PubMedCrossRef Enkvetchakul D, Loussouarn G, Makhina E, Nichols CG (2001) ATP interaction with the open state of the KATP channel. Biophys J 80:719–728PubMedCrossRef
40.
go back to reference Reimann F, Tucker SJ, Proks P, Ashcroft FM (1999) Involvement of the n-terminus of Kir6.2 in coupling to the sulphonylurea receptor. J Physiol 518:325–336PubMedCrossRef Reimann F, Tucker SJ, Proks P, Ashcroft FM (1999) Involvement of the n-terminus of Kir6.2 in coupling to the sulphonylurea receptor. J Physiol 518:325–336PubMedCrossRef
Metadata
Title
A Kir6.2 mutation causing severe functional effects in vitro produces neonatal diabetes without the expected neurological complications
Authors
P. Tammaro
S. E. Flanagan
B. Zadek
S. Srinivasan
H. Woodhead
S. Hameed
I. Klimes
A. T. Hattersley
S. Ellard
F. M. Ashcroft
Publication date
01-05-2008
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 5/2008
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-008-0923-1

Other articles of this Issue 5/2008

Diabetologia 5/2008 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

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.