Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Am Heart Assoc ; 2(4): e000365, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23974906

ABSTRACT

BACKGROUND: KATP channels, assembled from pore-forming (Kir6.1 or Kir6.2) and regulatory (SUR1 or SUR2) subunits, link metabolism to excitability. Loss of Kir6.2 results in hypoglycemia and hyperinsulinemia, whereas loss of Kir6.1 causes Prinzmetal angina-like symptoms in mice. Conversely, overactivity of Kir6.2 induces neonatal diabetes in mice and humans, but consequences of Kir6.1 overactivity are unknown. METHODS AND RESULTS: We generated transgenic mice expressing wild-type (WT), ATP-insensitive Kir6.1 [Gly343Asp] (GD), and ATP-insensitive Kir6.1 [Gly343Asp,Gln53Arg] (GD-QR) subunits, under Cre-recombinase control. Expression was induced in smooth muscle cells by crossing with smooth muscle myosin heavy chain promoter-driven tamoxifen-inducible Cre-recombinase (SMMHC-Cre-ER) mice. Three weeks after tamoxifen induction, we assessed blood pressure in anesthetized and conscious animals, as well as contractility of mesenteric artery smooth muscle and KATP currents in isolated mesenteric artery myocytes. Both systolic and diastolic blood pressures were significantly reduced in GD and GD-QR mice but normal in mice expressing the WT transgene and elevated in Kir6.1 knockout mice as well as in mice expressing dominant-negative Kir6.1 [AAA] in smooth muscle. Contractile response of isolated GD-QR mesenteric arteries was blunted relative to WT controls, but nitroprusside relaxation was unaffected. Basal KATP conductance and pinacidil-activated conductance were elevated in GD but not in WT myocytes. CONCLUSIONS: KATP overactivity in vascular muscle can lead directly to reduced vascular contractility and lower blood pressure. We predict that gain of vascular KATP function in humans would lead to a chronic vasodilatory phenotype, as indeed has recently been demonstrated in Cantu syndrome.


Subject(s)
Blood Pressure , Hypotension/metabolism , KATP Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Dose-Response Relationship, Drug , Genetic Predisposition to Disease , Hypotension/genetics , Hypotension/physiopathology , KATP Channels/genetics , Membrane Potentials , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Mutation , Phenotype , Potassium/metabolism , Vasoconstriction , Vasodilation , Vasodilator Agents/pharmacology
2.
Antimicrob Agents Chemother ; 55(4): 1377-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245443

ABSTRACT

Adverse effects induced by HIV protease inhibitors (PIs) are a significant factor in limiting their clinical success. PIs directly contribute to peripheral insulin resistance and alterations in lipid metabolism. GS-8374 is a novel PI with potent antiretroviral activity and a favorable resistance profile. Here we report on the potential of GS-8374 to adversely affect glucose and lipid homeostasis. Acute effects of GS-8374 and control PIs on glucose uptake and lipid accumulation were assessed in vitro in mouse OP9 and primary human adipocytes, respectively. GS-8374 and atazanavir showed no effect on insulin-stimulated deoxyglucose uptake, whereas ritonavir and lopinavir caused significant reductions. Similarly, in vitro lipid accumulation was not significantly affected in adipocytes treated with either GS-8374 or atazanavir. In euglycemic-hyperinsulinemic clamp experiments performed in rats during acute infusion of therapeutic levels of PIs, sustained serum GS-8374 levels of 8 µM had no effect on peripheral glucose disposal (similar to the findings for atazanavir). Comparable serum levels of lopinavir and ritonavir produced acute 19% and 53% reductions in in vivo glucose disposal, respectively. In conclusion, similar to atazanavir, but unlike ritonavir and lopinavir, GS-8374 neither affects insulin-stimulated glucose uptake in adipocytes in culture nor acutely alters peripheral glucose disposal in a rodent model system. These results dissociate the antiretroviral activity of GS-8374 from adverse effects on insulin sensitivity observed with some of the first-generation PIs and provide further support for the use of these experimental systems in the preclinical evaluation of novel PIs.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Glucose/metabolism , HIV Protease Inhibitors/pharmacology , Animals , Atazanavir Sulfate , Biological Transport/drug effects , Cells, Cultured , Glucose Clamp Technique , Homeostasis/drug effects , Humans , Insulin/pharmacology , Lopinavir , Male , Mice , Molecular Structure , Oligopeptides/pharmacology , Pyridines/pharmacology , Pyrimidinones/pharmacology , Rats , Rats, Wistar , Ritonavir/pharmacology
3.
Diabetes ; 60(1): 209-17, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980454

ABSTRACT

OBJECTIVE: The ATP-sensitive K(+) channel (K(ATP)) controls insulin secretion from the islet. Gain- or loss-of-function mutations in channel subunits underlie human neonatal diabetes and congenital hyperinsulinism (HI), respectively. In this study, we sought to identify the mechanistic basis of K(ATP)-induced HI in two probands and to characterize the clinical course. RESEARCH DESIGN AND METHODS: We analyzed HI in two probands and characterized the course of clinical treatment in each, as well as properties of mutant K(ATP) channels expressed in COSm6 cells using Rb efflux and patch-clamp methods. RESULTS: We identified mutation V290M in the pore-forming Kir6.2 subunit in each proband. In vitro expression in COSm6 cells supports the mutation resulting in an inactivating phenotype, which leads to significantly reduced activity in intact cells when expressed homomerically, and to a lesser extent when expressed heteromerically with wild-type subunits. In one heterozygous proband, a fluoro-DOPA scan revealed a causal focal lesion, indicating uniparental disomy with loss of heterozygosity. In a second family, the proband, homozygous for the mutation, was diagnosed with severe diazoxide-unresponsive hypersinsulinism at 2 weeks of age. The patient continues to be treated successfully with octreotide and amlodipine. The parents and a male sibling are heterozygous carriers without overt clinical HI. Interestingly, both the mother and the sibling exhibit evidence of abnormally enhanced glucose tolerance. CONCLUSIONS: V290M results in inactivating K(ATP) channels that underlie HI. Homozygous individuals may be managed medically, without pancreatectomy. Heterozygous carriers also show evidence of enhanced glucose sensitivity, consistent with incomplete loss of K(ATP) channel activity.


Subject(s)
Hyperinsulinism/congenital , Polymorphism, Single Nucleotide , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Adenosine Triphosphate/antagonists & inhibitors , Carrier State , Child , DNA/blood , DNA/genetics , DNA/isolation & purification , Gas Chromatography-Mass Spectrometry , Gene Amplification , Gene Silencing , Glucose Tolerance Test , Heterozygote , Homozygote , Humans , Hyperinsulinism/therapy , Immunoblotting , Insulin/metabolism , Insulin Secretion , Ion Channel Gating/genetics , Islets of Langerhans/metabolism , Male , Mutation , Phenotype , Polymerase Chain Reaction , Potassium Channels/physiology , Rubidium/metabolism
4.
J Biol Chem ; 285(47): 36395-400, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20864532

ABSTRACT

HIV protease inhibitors acutely block glucose transporters (GLUTs) in vitro, and this may contribute to altered glucose homeostasis in vivo. However, several GLUT-independent mechanisms have been postulated. To determine the contribution of GLUT blockade to protease inhibitor-mediated glucose dysregulation, the effects of ritonavir were investigated in mice lacking the insulin-sensitive glucose transporter GLUT4 (G4KO). G4KO and control C57BL/6J mice were administered ritonavir or vehicle at the start of an intraperitoneal glucose tolerance test and during hyperinsulinemic-euglycemic clamps. G4KO mice exhibited elevated fasting blood glucose compared with C57BL/6J mice. Ritonavir impaired glucose tolerance in control mice but did not exacerbate glucose intolerance in G4KO mice. Similarly, ritonavir reduced peripheral insulin sensitivity in control mice but not in G4KO mice. Serum insulin levels were reduced in vivo in ritonavir-treated mice. Ritonavir reduced serum leptin levels in C57BL/6J mice but had no effect on serum adiponectin. No change in these adipokines was observed following ritonavir treatment of G4KO mice. These data confirm that a primary effect of ritonavir on peripheral glucose disposal is mediated through direct inhibition of GLUT4 activity in vivo. The ability of GLUT4 blockade to contribute to derangements in the other molecular pathways that influence insulin sensitivity remains to be determined.


Subject(s)
Adipose Tissue/drug effects , Glucose Transporter Type 4/physiology , HIV Protease Inhibitors/pharmacology , Muscle, Skeletal/drug effects , Ritonavir/pharmacology , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Glucose Intolerance , Glucose Tolerance Test , Insulin/metabolism , Insulin Resistance , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism
5.
Physiol Rev ; 90(3): 799-829, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20664073

ABSTRACT

ATP-sensitive potassium (K(ATP)) channels are present in the surface and internal membranes of cardiac, skeletal, and smooth muscle cells and provide a unique feedback between muscle cell metabolism and electrical activity. In so doing, they can play an important role in the control of contractility, particularly when cellular energetics are compromised, protecting the tissue against calcium overload and fiber damage, but the cost of this protection may be enhanced arrhythmic activity. Generated as complexes of Kir6.1 or Kir6.2 pore-forming subunits with regulatory sulfonylurea receptor subunits, SUR1 or SUR2, the differential assembly of K(ATP) channels in different tissues gives rise to tissue-specific physiological and pharmacological regulation, and hence to the tissue-specific pharmacological control of contractility. The last 10 years have provided insights into the regulation and role of muscle K(ATP) channels, in large part driven by studies of mice in which the protein determinants of channel activity have been deleted or modified. As yet, few human diseases have been correlated with altered muscle K(ATP) activity, but genetically modified animals give important insights to likely pathological roles of aberrant channel activity in different muscle types.


Subject(s)
KATP Channels/metabolism , Muscle, Skeletal/metabolism , Muscle, Smooth/metabolism , Myocardium/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Heart/physiology , Heart/physiopathology , Humans , KATP Channels/chemistry , KATP Channels/genetics , Molecular Structure , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Muscle, Smooth, Vascular/physiology , Mutation , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , Sulfonylurea Receptors , Vasomotor System/physiology , Viscera/metabolism
6.
Dev Biol ; 346(1): 39-53, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20643119

ABSTRACT

Consistent left-right asymmetry requires specific ion currents. We characterize a novel laterality determinant in Xenopus laevis: the ATP-sensitive K(+)-channel (K(ATP)). Expression of specific dominant-negative mutants of the Xenopus Kir6.1 pore subunit of the K(ATP) channel induced randomization of asymmetric organ positioning. Spatio-temporally controlled loss-of-function experiments revealed that the K(ATP) channel functions asymmetrically in LR patterning during very early cleavage stages, and also symmetrically during the early blastula stages, a period when heretofore largely unknown events transmit LR patterning cues. Blocking K(ATP) channel activity randomizes the expression of the left-sided transcription of Nodal. Immunofluorescence analysis revealed that XKir6.1 is localized to basal membranes on the blastocoel roof and cell-cell junctions. A tight junction integrity assay showed that K(ATP) channels are required for proper tight junction function in early Xenopus embryos. We also present evidence that this function may be conserved to the chick, as inhibition of K(ATP) in the primitive streak of chick embryos randomizes the expression of the left-sided gene Sonic hedgehog. We propose a model by which K(ATP) channels control LR patterning via regulation of tight junctions.


Subject(s)
Body Patterning , Chick Embryo/growth & development , KATP Channels/physiology , Xenopus laevis/embryology , Animals , Blastomeres/metabolism , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Tight Junctions/physiology
7.
Pflugers Arch ; 460(2): 307-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19921246

ABSTRACT

Adenosine-triphosphate-sensitive potassium channels (KATP) are regulated by adenosine nucleotides, and, thereby, couple cellular metabolism with electrical activity in multiple tissues including the pancreatic beta-cell. The critical involvement of KATP in insulin secretion is confirmed by the demonstration that inactivating and activating mutations in KATP underlie persistent hyperinsulinemia and neonatal diabetes mellitus, respectively, in both animal models and humans. In addition, a common variant in KATP represents a risk factor in the etiology of type 2 diabetes. This review focuses on the mechanistic basis by which KATP mutations underlie insulin secretory disorders and the implications of these findings for successful clinical intervention.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Channelopathies/physiopathology , Congenital Hyperinsulinism/genetics , Potassium Channels, Inwardly Rectifying/physiology , Receptors, Drug/physiology , ATP-Binding Cassette Transporters/genetics , Animals , Channelopathies/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
8.
Pediatr Diabetes ; 11(4): 286-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19656320

ABSTRACT

Activating mutations in the K(ATP)-channel cause neonatal diabetes mellitus (NDM), and patients have been safely transitioned from insulin to sulfonylureas. We report a male infant with permanent NDM (PNDM), born to a PNDM mother. Blood glucose began to rise on day of life (DOL) 2, and sulfonylurea (glyburide) therapy was initiated on DOL 5. Glucose was subsequently well controlled and normal at 3 months. A K(ATP) mutation (R201H; KCNJ11) was detected in the infant, the mother, and 6-yr-old sister with PNDM; both were also subsequently transitioned off insulin onto glyburide. To our knowledge, this is the youngest NDM patient to receive oral glyburide and, importantly, the only one deliberately initiated on sulfonylureas. Strikingly, the current dose (0.017 mg/kg/d) is below the reported therapeutic range and approximately 75-fold lower than doses required by the affected sister and mother. Pancreatic insulin disappears in an animal model of K(ATP)-induced NDM, unless glycemia is well controlled, thus, a dramatically lower glyburide requirement in the infant may reflect preserved insulin content because of early sulfonylurea intervention. Safe and effective initiation of glyburide in an insulin-naïve neonatal patient with K(ATP)-dependent PNDM argues for early detection and sulfonylurea intervention.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Glyburide/therapeutic use , Hypoglycemic Agents/therapeutic use , Potassium Channels, Inwardly Rectifying/genetics , Blood Glucose/drug effects , Blood Glucose/genetics , Child , Diabetes Mellitus/diagnosis , Early Diagnosis , Female , Humans , Infant, Newborn , Insulin/therapeutic use , Male , Mutation , Treatment Outcome , Young Adult
9.
Diabetes ; 58(8): 1869-78, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19491206

ABSTRACT

OBJECTIVE: The E23K variant in the Kir6.2 subunit of the ATP-sensitive K(+) channel (K(ATP) channel) is associated with increased risk of type 2 diabetes. The present study was undertaken to increase our understanding of the mechanisms responsible. To avoid confounding effects of hyperglycemia, insulin secretion and action were studied in subjects with the variant who had normal glucose tolerance. RESEARCH DESIGN AND METHODS: Nine subjects with the E23K genotype K/K and nine matched subjects with the E/E genotype underwent 5-h oral glucose tolerance tests (OGTTs), graded glucose infusion, and hyperinsulinemic-euglycemic clamp with stable-isotope-labeled tracer infusions to assess insulin secretion, action, and clearance. A total of 461 volunteers consecutively genotyped for the E23K variant also underwent OGTTs. Functional studies of the wild-type and E23K variant potassium channels were conducted. RESULTS: Insulin secretory responses to oral and intravenous glucose were reduced by approximately 40% in glucose-tolerant subjects homozygous for E23K. Normal glucose tolerance with reduced insulin secretion suggests a change in insulin sensitivity. The hyperinsulinemic-euglycemic clamp revealed that hepatic insulin sensitivity is approximately 40% greater in subjects with the E23K variant, and these subjects demonstrate increased insulin sensitivity after oral glucose. The reconstituted E23K channels confirm reduced sensitivity to inhibitory ATP and increase in open probability, a direct molecular explanation for reduced insulin secretion. CONCLUSIONS: The E23K variant leads to overactivity of the K(ATP) channel, resulting in reduced insulin secretion. Initially, insulin sensitivity is enhanced, thereby maintaining normal glucose tolerance. Presumably, over time, as insulin secretion falls further or insulin resistance develops, glucose levels rise resulting in type 2 diabetes.


Subject(s)
Insulin/metabolism , KATP Channels/physiology , Potassium Channels, Inwardly Rectifying/genetics , ATP-Binding Cassette Transporters/genetics , Adult , Amino Acid Substitution , Cross-Sectional Studies , Female , Genetic Variation , Genotype , Glucose Clamp Technique , Glucose Tolerance Test , Humans , Insulin Secretion , Male , Middle Aged , Receptors, Drug/genetics , Reference Values , Sulfonylurea Receptors
10.
Cell Metab ; 9(2): 140-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187772

ABSTRACT

ATP-insensitive K(ATP) channel mutations cause neonatal diabetes mellitus (NDM). To explore the mechanistic etiology, we generated transgenic mice carrying an ATP-insensitive mutant K(ATP) channel subunit. Constitutive expression in pancreatic beta cells caused neonatal hyperglycemia and progression to severe diabetes and growth retardation, with loss of islet insulin content and beta cell architecture. Tamoxifen-induced expression in adult beta cells led to diabetes within 2 weeks, with similar secondary consequences. Diabetes was prevented by transplantation of normal islets under the kidney capsule. Moreover, the endogenous islets maintained normal insulin content and secretion in response to sulfonylureas, but not glucose, consistent with reduced ATP sensitivity of beta cell K(ATP) channels. In NDM, transfer to sulfonylurea therapy is less effective in older patients. This may stem from poor glycemic control or lack of insulin because glibenclamide treatment prior to tamoxifen induction prevented diabetes and secondary complications in mice but failed to halt disease progression after diabetes had developed.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Insulin-Secreting Cells/physiology , KATP Channels/genetics , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/physiopathology , Glyburide/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Transgenic , Models, Animal , Sulfonylurea Compounds/pharmacology , Tamoxifen/pharmacology
11.
J Cardiovasc Electrophysiol ; 20(1): 93-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19120683

ABSTRACT

BACKGROUND: Early repolarization in the inferolateral leads has been recently recognized as a frequent syndrome associated with idiopathic ventricular fibrillation (VF). We report the case of a patient presenting dramatic changes in the ECG in association with recurrent VF in whom a novel genetic variant has been identified. CASE REPORT: This young female (14 years) was resuscitated in 2001 following an episode of sudden death due to VF. All examinations including coronary angiogram with ergonovine injection, MRI, and flecainide or isoproterenol infusion were normal. The patient had multiple (>100) recurrences of VF unresponsive to beta-blockers, lidocaine/mexiletine, verapamil, and amiodarone. Recurrences of VF were associated with massive accentuation of the early repolarization pattern at times mimicking acute myocardial ischemia. Coronary angiography during an episode with 1.2 mV J/ST elevation was normal. Isoproterenol infusion acutely suppressed electrical storms, while quinidine eliminated all recurrences of VF and restored a normal ECG over a follow-up of 65 months. Genomic DNA sequencing of K(ATP) channel genes showed missense variant in exon 3 (NC_000012) of the KCNJ8 gene, a subunit of the K(ATP) channel, conferring predisposition to dramatic repolarization changes and ventricular vulnerability.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/genetics , Adolescent , Electrocardiography , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Humans , KATP Channels
12.
Biophys J ; 95(10): 4689-97, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18708460

ABSTRACT

ATP-sensitive K(+)-channels link metabolism and excitability in neurons, myocytes, and pancreatic islets. Mutations in the pore-forming subunit (Kir6.2; KCNJ11) cause neonatal diabetes, developmental delay, and epilepsy by decreasing sensitivity to ATP inhibition and suppressing electrical activity. Mutations of residue G53 underlie both mild (G53R,S) and severe (G53D) forms of the disease. All examined substitutions (G53D,R,S,A,C,F) reduced ATP-sensitivity, indicating an intolerance of any amino acid other than glycine. Surprisingly, each mutation reduces ATP affinity, rather than intrinsic gating, although structural modeling places G53 at a significant distance from the ATP-binding pocket. We propose that glycine is required in this location for flexibility of the distal N-terminus, and for an induced fit of ATP at the binding site. Consistent with this hypothesis, glycine substitution of the adjacent residue (Q52G) partially rescues ATP affinity of reconstituted Q52G/G53D channels. The results reveal an important feature of the noncanonical ATP-sensing mechanism of K(ATP) channels.


Subject(s)
Adenosine Triphosphate/metabolism , KATP Channels/metabolism , Membrane Potentials/physiology , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/chemistry , Animals , COS Cells , Chlorocebus aethiops , KATP Channels/chemistry , Mutagenesis, Site-Directed , Structure-Activity Relationship
13.
J Clin Endocrinol Metab ; 93(3): 1054-61, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18073297

ABSTRACT

CONTEXT: Mutations in the Kir6.2 subunit (KCNJ11) of the ATP-sensitive potassium channel (KATP) underlie neonatal diabetes mellitus. In severe cases, Kir6.2 mutations underlie developmental delay, epilepsy, and neonatal diabetes (DEND). All Kir6.2 mutations examined decrease the ATP inhibition of KATP, which is predicted to suppress electrical activity in neurons (peripheral and central), muscle, and pancreas. Inhibitory sulfonylureas (SUs) have been used successfully to treat diabetes in patients with activating Kir6.2 mutations. There are two reports of improved neurological features in SU-treated DEND patients but no report of such improvement in adulthood. OBJECTIVE: The objective of the study was to determine the molecular basis of intermediate DEND in a 27-yr-old patient with a KCNJ11 mutation (G53D) and the patient's response to SU therapy. DESIGN: The G53D patient was transferred from insulin to gliclazide and then to glibenclamide over a 160-d period. Motor function was assessed throughout. Electrophysiology assessed the effect of the G53D mutation on KATP activity. RESULTS: The G53D patient demonstrated improved glycemic control and motor coordination with SU treatment, although glibenclamide was more effective than gliclazide. Reconstituted G53D channels exhibit reduced ATP sensitivity, which is predicted to suppress electrical activity in vivo. G53D channels coexpressed with SUR1 (the pancreatic and neuronal isoform) exhibit high-affinity block by gliclazide but are insensitive to block when coexpressed with SUR2A (the skeletal muscle isoform). High-affinity block by glibenclamide is present in G53D channels coexpressed with either SUR1 or SUR2A. CONCLUSION: The results demonstrate that SUs can resolve motor dysfunction in an adult with intermediate DEND and that this improvement is due to inhibition of the neuronal but not skeletal muscle KATP.


Subject(s)
Diabetes Mellitus/genetics , Potassium Channels, Inwardly Rectifying/genetics , Sulfonylurea Compounds/therapeutic use , Adenosine Triphosphate/pharmacology , Adult , Diabetes Mellitus/drug therapy , Diabetes Mellitus/physiopathology , Genotype , Humans , Infant, Newborn , Motor Activity , Mutation , Potassium Channels, Inwardly Rectifying/physiology
14.
Diabetes ; 56(2): 328-36, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17259376

ABSTRACT

Mutations in the pancreatic ATP-sensitive K(+) channel (K(ATP) channel) cause permanent neonatal diabetes mellitus (PNDM) in humans. All of the K(ATP) channel mutations examined result in decreased ATP inhibition, which in turn is predicted to suppress insulin secretion. Here we describe a patient with severe PNDM, which includes developmental delay and epilepsy, in addition to neonatal diabetes (developmental delay, epilepsy, and neonatal diabetes [DEND]), due to a G334D mutation in the Kir6.2 subunit of K(ATP) channel. The patient was wholly unresponsive to sulfonylurea therapy (up to 1.14 mg . kg(-1) . day(-1)) and remained insulin dependent. Consistent with the putative role of G334 as an ATP-binding residue, reconstituted homomeric and mixed WT+G334D channels exhibit absent or reduced ATP sensitivity but normal gating behavior in the absence of ATP. In disagreement with the sulfonylurea insensitivity of the affected patient, the G334D mutation has no effect on the sulfonylurea inhibition of reconstituted channels in excised patches. However, in macroscopic rubidium-efflux assays in intact cells, reconstituted mutant channels do exhibit a decreased, but still present, sulfonylurea response. The results demonstrate that ATP-binding site mutations can indeed cause DEND and suggest the possibility that sulfonylurea insensitivity of such patients may be a secondary reflection of the presence of DEND rather than a simple reflection of the underlying molecular basis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/genetics , Developmental Disabilities/genetics , Diabetes Mellitus, Type 1/genetics , Epilepsy/genetics , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Receptors, Drug/genetics , Adenosine Triphosphate/metabolism , Adolescent , Alleles , Binding Sites/genetics , Diabetes Mellitus, Type 1/congenital , Diabetes Mellitus, Type 1/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Infant, Newborn , Male , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Compounds/therapeutic use , Sulfonylurea Receptors , Syndrome , Tolbutamide/metabolism
15.
Diabetes ; 55(11): 2957-64, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065331

ABSTRACT

Glucose metabolism in pancreatic beta-cells elevates cytoplasmic [ATP]/[ADP], causing closure of ATP-sensitive K(+) channels (K(ATP) channels), Ca(2+) entry through voltage-dependent Ca(2+) channels, and insulin release. Decreased responsiveness of K(ATP) channels to the [ATP]/[ADP] ratio should lead to decreased insulin secretion and diabetes. We generated mice expressing K(ATP) channels with reduced ATP sensitivity in their beta-cells. Previously, we described a severe diabetes, with nearly complete neonatal lethality, in four lines (A-C and E) of these mice. We have now analyzed an additional three lines (D, F, and G) in which the transgene is expressed at relatively low levels. These animals survive past weaning but are glucose intolerant and can develop severe diabetes. Despite normal islet morphology and insulin content, islets from glucose-intolerant animals exhibit reduced glucose-stimulated insulin secretion. The data demonstrate that a range of phenotypes can be expected for a reduction in ATP sensitivity of beta-cell K(ATP) channels and provide models for the corollary neonatal diabetes in humans.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Insulin-Secreting Cells/physiology , Potassium Channels, Inwardly Rectifying/genetics , Animals , Animals, Newborn , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Insulin/blood , Islets of Langerhans/physiology , Mice , Mice, Transgenic , Patch-Clamp Techniques
16.
PLoS Biol ; 4(2): e26, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16402858

ABSTRACT

Pancreatic beta-cells secrete insulin in response to closure of ATP-sensitive K+ (KATP) channels, which causes membrane depolarization and a concomitant rise in intracellular Ca2+ (Cai). In intact islets, beta-cells are coupled by gap junctions, which are proposed to synchronize electrical activity and Cai oscillations after exposure to stimulatory glucose (>7 mM). To determine the significance of this coupling in regulating insulin secretion, we examined islets and beta-cells from transgenic mice that express zero functional KATP channels in approximately 70% of their beta-cells, but normal KATP channel density in the remainder. We found that KATP channel activity from approximately 30% of the beta-cells is sufficient to maintain strong glucose dependence of metabolism, Cai, membrane potential, and insulin secretion from intact islets, but that glucose dependence is lost in isolated transgenic cells. Further, inhibition of gap junctions caused loss of glucose sensitivity specifically in transgenic islets. These data demonstrate a critical role of gap junctional coupling of KATP channel activity in control of membrane potential across the islet. Control via coupling lessens the effects of cell-cell variation and provides resistance to defects in excitability that would otherwise lead to a profound diabetic state, such as occurs in persistent neonatal diabetes mellitus.


Subject(s)
Gap Junctions/metabolism , Insulin/metabolism , Potassium Channels/metabolism , Animals , Calcium/chemistry , Calcium/metabolism , Cations, Divalent/chemistry , Cells, Cultured , Glucose , Insulin Secretion , Islets of Langerhans/metabolism , Membrane Potentials , Models, Biological , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Tissue Culture Techniques
17.
Diabetes ; 54(11): 3065-72, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16249427

ABSTRACT

The ATP-sensitive K+ channel (K ATP channel) senses metabolic changes in the pancreatic beta-cell, thereby coupling metabolism to electrical activity and ultimately to insulin secretion. When K ATP channels open, beta-cells hyperpolarize and insulin secretion is suppressed. The prediction that K ATP channel "overactivity" should cause a diabetic state due to undersecretion of insulin has been dramatically borne out by recent genetic studies implicating "activating" mutations in the Kir6.2 subunit of K ATP channel as causal in human diabetes. This article summarizes the emerging picture of K ATP channel as a major cause of neonatal diabetes and of a polymorphism in K ATP channel (E23K) as a type 2 diabetes risk factor. The degree of K ATP channel "overactivity" correlates with the severity of the diabetic phenotype. At one end of the spectrum, polymorphisms that result in a modest increase in K ATP channel activity represent a risk factor for development of late-onset diabetes. At the other end, severe "activating" mutations underlie syndromic neonatal diabetes, with multiple organ involvement and complete failure of glucose-dependent insulin secretion, reflecting K ATP channel "overactivity" in both pancreatic and extrapancreatic tissues.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Insulin/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans , Insulin Secretion , Mutation/genetics
18.
Diabetes ; 54(10): 2925-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186394

ABSTRACT

As the rate-limiting controller of glucose metabolism, glucokinase represents the primary beta-cell "glucose sensor." Inactivation of both glucokinase (GK) alleles results in permanent neonatal diabetes; inactivation of a single allele causes maturity-onset diabetes of the young type 2 (MODY-2). Similarly, mice lacking both alleles (GK(-/-)) exhibit severe neonatal diabetes and die within a week, whereas heterozygous GK(+/-) mice exhibit markedly impaired glucose tolerance and diabetes, resembling MODY-2. Glucose metabolism increases the cytosolic [ATP]-to-[ADP] ratio, which closes ATP-sensitive K(+) channels (K(ATP) channels), leading to membrane depolarization, Ca(2+) entry, and insulin exocytosis. Glucokinase insufficiency causes defective K(ATP) channel regulation, which may underlie the impaired secretion. To test this prediction, we crossed mice lacking neuroendocrine glucokinase (nGK(+/-)) with mice lacking K(ATP) channels (Kir6.2(-/-)). Kir6.2 knockout rescues perinatal lethality of nGK(-/-), although nGK(-/-)Kir6.2(-/-) animals are postnatally diabetic and still die prematurely. nGK(+/-) animals are diabetic on the Kir6.2(+/+) background but only mildly glucose intolerant on the Kir6.2(-/-) background. In the presence of glutamine, isolated nGK(+/-)Kir6.2(-/-) islets show improved insulin secretion compared with nGK(+/-)Kir6.2(+/+). The significant abrogation of nGK(-/-) and nGK(+/-) phenotypes in the absence of K(ATP) demonstrate that a major factor in glucokinase deficiency is indeed altered K(ATP) signaling. The results have implications for understanding and therapy of glucokinase-related diabetes.


Subject(s)
Diabetes Mellitus/enzymology , Glucokinase/deficiency , Potassium Channels, Inwardly Rectifying/physiology , Signal Transduction , Animals , Animals, Newborn , Blood Glucose/analysis , Crosses, Genetic , Diabetes Mellitus/genetics , Diabetes Mellitus/mortality , Genotype , Glucokinase/physiology , Glutamine/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/enzymology , Islets of Langerhans/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Inwardly Rectifying/deficiency
19.
Diabetes ; 54(9): 2645-54, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123353

ABSTRACT

The prediction that overactivity of the pancreatic ATP-sensitive K(+) channel (K(ATP) channel) underlies reduced insulin secretion and causes a diabetic phenotype in humans has recently been borne out by genetic studies implicating "activating" mutations in the Kir6.2 subunit of K(ATP) as causal in both permanent and transient neonatal diabetes. Here we characterize the channel properties of Kir6.2 mutations that underlie transient neonatal diabetes (I182V) or more severe forms of permanent neonatal diabetes (V59M, Q52R, and I296L). In all cases, the mutations result in a significant decrease in sensitivity to inhibitory ATP, which correlates with channel "overactivity" in intact cells. Mutations can be separated into those that directly affect ATP affinity (I182V) and those that stabilize the open conformation of the channel and indirectly reduce ATP sensitivity (V59M, Q52R, and I296L). With respect to the latter group, alterations in channel gating are also reflected in a functional "uncoupling" of sulfonylurea (SU) block: SU sensitivity of I182V is similar to that of wild-type mutants, but the SU sensitivity of all gating mutants is reduced, with the I296L mutant being resistant to block by tolbutamide (

Subject(s)
Adenosine Triphosphate/physiology , Glyburide/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Tolbutamide/pharmacology , Animals , Cell Line , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Humans , Hypoglycemic Agents/pharmacology , Mutation , Pharmacogenetics
20.
Diabetes ; 53(12): 3159-67, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15561946

ABSTRACT

ATP-sensitive K+ channels (K(ATP) channels) control electrical activity in beta-cells and therefore are key players in excitation-secretion coupling. Partial suppression of beta-cell K(ATP) channels in transgenic (AAA) mice causes hypersecretion of insulin and enhanced glucose tolerance, whereas complete suppression of these channels in Kir6.2 knockout (KO) mice leads to hyperexcitability, but mild glucose intolerance. To test the interplay of hyperexcitability and dietary stress, we subjected AAA and KO mice to a high-fat diet. After 3 months on the diet, both AAA and KO mice converted to an undersecreting and markedly glucose-intolerant phenotype. Although Kir6.2 is expressed in multiple tissues, its primary functional consequence in both AAA and KO mice is enhanced beta-cell electrical activity. The results of our study provide evidence that, when combined with dietary stress, this hyperexcitability is a causal diabetic factor. We propose an "inverse U" model for the response to enhanced beta-cell excitability: the expected initial hypersecretion can progress to undersecretion and glucose-intolerance, either spontaneously or in response to dietary stress.


Subject(s)
Glucose Intolerance/physiopathology , Islets of Langerhans/physiopathology , Potassium Channels, Inwardly Rectifying/physiology , Animals , Blood Glucose/metabolism , Glucose Intolerance/drug therapy , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...