Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Diabetologia ; 54(7): 1693-701, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21544516

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to investigate the genetic aetiology of permanent diabetes mellitus with onset in the first 12 months of age. METHODS: We studied 46 probands with permanent, insulin-requiring diabetes with onset within the first 6 months of life (permanent neonatal diabetes mellitus [PNDM]/monogenic diabetes of infancy [MDI]) (group 1) and eight participants with diabetes diagnosed between 7 and 12 months of age (group 2). KCNJ11, INS and ABCC8 genes were sequentially sequenced in all patients. For those who were negative in the initial screening, we examined ERN1, CHGA, CHGB and NKX6-1 genes and, in selected probands, CACNA1C, GCK, FOXP3, NEUROG3 and CDK4. The incidence rate for PNDM/MDI was calculated using a database of Italian patients collected from 1995 to 2009. RESULTS: In group 1 we found mutations in KCNJ11, INS and ABCC8 genes in 23 (50%), 9 (19.5%) and 4 (8.6%) patients respectively, and a single homozygous mutation in GCK (2.1%). In group 2, we identified one incidence of a KCNJ11 mutation. No genetic defects were detected in other loci. The incidence rate of PNDM/MDI in Italy is estimated to be 1:210,287. CONCLUSIONS/INTERPRETATION: Genetic mutations were identified in ~75% of non-consanguineous probands with PNDM/MDI, using sequential screening of KCNJ11, INS and ABCC8 genes in infants diagnosed within the first 6 months of age. This percentage decreased to 12% in those with diabetes diagnosed between 7 and 12 months. Patients belonging to the latter group may either carry mutations in genes different from those commonly found in PNDM/MDI or have developed an early-onset form of autoimmune diabetes.


Subject(s)
Diabetes Mellitus/etiology , Diabetes Mellitus/genetics , ATP-Binding Cassette Transporters/genetics , Diabetes Mellitus/epidemiology , Female , Genetic Predisposition to Disease , Germinal Center Kinases , Humans , Infant , Infant, Newborn , Insulin/genetics , Male , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
2.
Pflugers Arch ; 457(6): 1351-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18836740

ABSTRACT

Pancreatic beta-cells of sulfonylurea receptor type 1 knock-out (SUR1(-/-)) mice exhibit an oscillating membrane potential (V (m)) demonstrating that hyper-polarisation occurs despite the lack of K(ATP) channels. We hypothesize that glucose activates the Na(+)/K(+)-ATPase thus increasing a hyper-polarising current. Elevating glucose in SUR1(-/-) beta-cells resulted in a transient fall in V (m) and [Ca(2+)](c) independent of sarcoplasmic and endoplasmic reticulum Ca(2+)-activated ATPase (SERCA) activation. This was not affected by K(+) channel blockade but inhibited by ATP depletion and by ouabain. Increasing glucose also reduced [Na(+)](c), an effect reversed by ouabain. Exogenously applied insulin decreased [Na(+)](c) and hyper-polarised V (m). Inhibiting insulin signalling in SUR1(-/-) beta-cells blunted the glucose-induced decrease of [Ca(2+)](c). Tolbutamide (1 mmol/l) disclosed the SERCA-independent effect of glucose on [Ca(2+)](c) in wild-type beta-cells. The data show that in SUR1(-/-) beta-cells, glucose activates the Na(+)/K(+)-ATPase presumably by increasing [ATP](c). Insulin can also stimulate the pump and potentiate the effect of glucose. Pathways involving the pump may thus serve as potential drug targets in certain metabolic disorders.


Subject(s)
Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphate/physiology , Animals , Calcium/metabolism , Enzyme Activation , Insulin/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Signal Transduction , Sodium-Potassium-Exchanging ATPase/drug effects
3.
Curr Pharm Des ; 11(21): 2699-716, 2005.
Article in English | MEDLINE | ID: mdl-16101450

ABSTRACT

ATP-sensitive K+ channels, termed K(ATP) channels, provide a link between cellular metabolism and membrane electrical activity in a variety of tissues. Channel isoforms have been identified and are targets for compounds that both stimulate and inhibit their activity resulting in membrane hyperpolarization and depolarization, respectively. Examples include relaxation of vascular smooth muscle and stimulation of insulin secretion. This article reviews the cloning, molecular biology, and structure of K(ATP) channels, with particular focus on the SUR1/K(IR)6.2 neuroendocrine channels that are important for the regulation of insulin secretion. We integrate the extensive pharmacologic structure-activity-relationship data on these channels, which defines a bipartite drug binding pocket in the SUR (sulfonylurea receptor), with recent structure-function studies that identify domains of SUR and K(IR)6.2, the channel pore, which are critical for channel assembly, for gating, and for the ligand-receptor interactions that modulate channel activity. The atomic structure of a sulfonylurea in a protein pocket is used to develop insight into the recognition of these compounds. A homology model of K(ATP) channels, based on VC-MsbA, another member of the ABC protein family, is described and used to position amino acids important for the action of channel openers and blockers within the core of SUR. The model has a central chamber which could serve as a multifaceted binding pocket.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Potassium Channels, Inwardly Rectifying/drug effects , Receptors, Drug/drug effects , Sulfonylurea Compounds/metabolism , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Binding Sites/drug effects , Humans , Insulin Secretion , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , KATP Channels , Models, Molecular , Potassium Channels/biosynthesis , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/biosynthesis , Receptors, Drug/genetics , Stimulation, Chemical , Structure-Activity Relationship , Sulfonylurea Receptors
4.
Diabetologia ; 48(5): 913-21, 2005 May.
Article in English | MEDLINE | ID: mdl-15830184

ABSTRACT

AIMS/HYPOTHESIS: Islets or beta cells from Sur1(-/-) mice were used to determine whether changes in plasma membrane potential (V(m)) remain coupled to changes in cytosolic Ca(2+) ([Ca(2+)](i)) in the absence of K(ATP) channels and thus provide a triggering signal for insulin secretion. The study also sought to elucidate whether [Ca(2+)](i) influences oscillations in V(m) in sur1(-/-) beta cells. METHODS: Plasma membrane potential and ion currents were measured with microelectrodes and the patch-clamp technique. [Ca(2+)](i) was monitored with the fluorescent dye fura-2. Insulin secretion from isolated islets was determined by static incubations. RESULTS: Membrane depolarisation of Sur1(-/-) islets by arginine or increased extracellular K(+), elevated [Ca(2+)](i) and augmented insulin secretion. Oligomycin completely abolished glucose-stimulated insulin release from Sur1(-/-) islets. Oscillations in V(m) were influenced by [Ca(2+)](i) as follows: (1) elevation of extracellular Ca(2+) lengthened phases of membrane hyperpolarisation; (2) simulating a burst of action potentials induced a Ca(2+)-dependent outward current that was augmented by increased Ca(2+) influx through L-type Ca(2+) channels; (3) Ca(2+) depletion of intracellular stores by cyclopiazonic acid increased the burst frequency in Sur1(-/-) islets, elevating [Ca(2+)](i) and insulin secretion; (4) store depletion activated a Ca(2+) influx that was not inhibitable by the L-type Ca(2+) channel blocker D600. CONCLUSIONS/INTERPRETATION: Although V(m) is largely uncoupled from glucose metabolism in the absence of K(ATP) channels, increased electrical activity leads to elevations of [Ca(2+)](i) that are sufficient to stimulate insulin secretion. In Sur1(-/-) beta cells, [Ca(2+)](i) exerts feedback mechanisms on V(m) by activating a hyperpolarising outward current and by depolarising V(m) via store-operated ion channels.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Calcium/metabolism , Insulin/metabolism , Islets of Langerhans/physiology , Membrane Potentials/physiology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Receptors, Drug/genetics , Animals , Arginine/pharmacology , Cell Membrane/physiology , Indoles/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels/deficiency , Potassium Channels, Inwardly Rectifying/deficiency , Receptors, Drug/deficiency , Sulfonylurea Receptors
5.
Diabetologia ; 47(3): 488-498, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14872319

ABSTRACT

AIMS/HYPOTHESIS: SUR1(ABCC8)(-/-) mice lacking functional K(ATP) channels are an appropriate model to test the significance of K(ATP) channels in beta-cell function. We examined how this gene deletion interferes with stimulus-secretion coupling. We tested the influence of metabolic inhibition and galanin, whose mode of action is controversial. METHODS: Plasma membrane potential (Vm) and currents were measured with microelectrodes or the patch-clamp technique; cytosolic Ca(2+) concentrations ([Ca(2+)](c)) and mitochondrial membrane potential (DeltaPsi) were measured using fluorescent dyes. RESULTS: In contrast to the controls, SUR1(-/-) beta cells showed electrical activity even at a low glucose concentration. Continuous spike activity was measured with the patch-clamp technique, but with microelectrodes slow oscillations in Vm consisting of bursts of Ca(2+)-dependent action potentials were detected. [Ca(2+)](c) showed various patterns of oscillations or a sustained increase. Sodium azide did not hyperpolarize SUR1(-/-) beta cells. The depolarization of DeltaPsi evoked by sodium azide was significantly lower in SUR1(-/-) than SUR1(+/+) cells. Galanin transiently decreased action potential frequency and [Ca(2+)](c) in cells from both SUR1(-/-) and SUR1(+/+) mice. CONCLUSION/INTERPRETATION: The strong dependence of Vm and [Ca(2+)](c) on glucose concentration observed in SUR1(+/+) beta cells is disrupted in the knock-out cells. This demonstrates that both parameters oscillate in the absence of functional K(ATP) channels. The lack of effect of metabolic inhibition by sodium azide shows that in SUR1(-/-) beta cells changes in ATP/ADP no longer link glucose metabolism and Vm. The results with galanin suggest that this peptide affects beta cells independently of K(ATP) currents and thus could contribute to the regulation of beta-cell function in SUR1(-/-) animals.


Subject(s)
Calcium/physiology , Islets of Langerhans/physiology , Potassium Channels, Inwardly Rectifying/physiology , ATP-Binding Cassette Transporters , Action Potentials/physiology , Animals , Cytosol/metabolism , Glucose/pharmacology , Islets of Langerhans/drug effects , Kinetics , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Proteins , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug , Sulfonylurea Receptors
6.
Diabetes ; 50(2): 322-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272143

ABSTRACT

Mutations in the high-affinity sulfonylurea receptor (SUR)-1 cause one of the severe recessively inherited diffuse forms of congenital hyperinsulinism or, when associated with loss of heterozygosity, focal adenomatosis. We hypothesized that SUR1 mutations would render the beta-cell insensitive to sulfonylureas and to glucose. Stimulated insulin responses were compared among eight patients with diffuse hyperinsulinism (two mutations), six carrier parents, and ten normal adults. In the patients with diffuse hyperinsulinism, the acute insulin response to intravenous tolbutamide was absent and did not overlap with the responses seen in either adult group. There was positive, albeit significantly blunted, acute insulin response to intravenous dextrose in the patients with diffuse hyperinsulinism. Graded infusions of glucose, to raise and then lower plasma glucose concentrations over 4 h, caused similar rises in blood glucose but lower peak insulin levels in the hyperinsulinemic patients. Loss of acute insulin response to tolbutamide can identify children with diffuse SUR1 defects. The greater response to glucose than to tolbutamide indicates that ATP-sensitive potassium (KATP) channel-independent pathways are involved in glucose-mediated insulin release in patients with diffuse SUR1 defects. The diminished glucose responsiveness suggests that SUR1 mutations and lack of KATP channel activity may contribute to the late development of diabetes in patients with hyperinsulinism independently of subtotal pancreatectomy.


Subject(s)
ATP-Binding Cassette Transporters , Hyperinsulinism/congenital , Hyperinsulinism/genetics , Insulin/metabolism , Mutation/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Glucose/pharmacology , Heterozygote , Humans , Hypoglycemic Agents/pharmacology , Injections, Intravenous , Insulin Secretion , Male , Reference Values , Sulfonylurea Receptors , Tolbutamide/pharmacology
7.
Recent Prog Horm Res ; 56: 47-68, 2001.
Article in English | MEDLINE | ID: mdl-11237225

ABSTRACT

K(ATP) channels are a unique, small family of potassium (K+)-selective ion channels assembled from four inward rectifier pore-forming subunits, K(IR)6.x, paired with four sulfonylurea receptors (SURs), members of the adenosine triphosphate (ATP)-binding cassette superfamily. The activity of these channels can be regulated by metabolically driven changes in the ratio of adenosine diphosphate (ADP) to ATP, providing a means to couple membrane electrical activity with metabolism. In pancreatic beta cells in the islets of Langerhans, K(ATP) channels are part of an ionic mechanism that couples glucose metabolism to insulin secretion. This chapter 1) briefly describes the properties of K(ATP) channels; 2) discusses data on a genetically recessive form of persistent hyperinsulinemic hypoglycemia of infancy (PHHI), caused by loss of beta-cell K(ATP) channel activity; and 3) compares the severe impairment of glucose homeostasis that characterizes the human phenotype with the near-normal phenotype observed in K(ATP) channel null mice.


Subject(s)
Hyperinsulinism/genetics , Insulin/metabolism , Pancreatic Diseases/genetics , Potassium Channels/genetics , Potassium Channels/physiology , Animals , Dose-Response Relationship, Drug , Electrophysiology , Genes, Recessive , Homeostasis , Humans , Insulin Secretion , Kinetics , Mice , Mice, Knockout , Multigene Family , Mutation , Phenotype , Time Factors
8.
J Pediatr ; 137(2): 239-46, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10931418

ABSTRACT

OBJECTIVES: To identify infants with hyperinsulinism caused by defects of the beta-cell adenosine triphosphate-dependent potassium channel complex and to distinguish focal and diffuse forms of hyperinsulinism caused by these mutations. STUDY DESIGN: The acute insulin response to intravenous calcium stimulation (CaAIR) was determined in 9 patients <20 years with diffuse hyperinsulinism caused by defective beta-cell sulfonylurea receptor (SUR1(-/-)), 3 patients with focal congenital hyperinsulinism (6 weeks to 18 months), a 10-year-old with insulinoma, 5 with hyperinsulinism/hyperammonemia syndrome caused by defective glutamate dehydrogenase (6 months to 28 years), 4 SUR1(+/-) heterozygotes with no symptoms, and 9 normal adults. Three infants with congenital focal disease, 1 with diffuse hyperinsulinism, and the child with insulinoma underwent selective pancreatic intra-arterial calcium stimulation with hepatic venous sampling. RESULTS: Children with diffuse SUR1(-/-) disease and infants with congenital focal hyperinsulinism responded to CaAIR, whereas the normal control group, patients with hyperinsulinism/hyperammonemia syndrome, and SUR1(+/-) carriers did not. Selective arterial calcium stimulation of the pancreas with hepatic venous sampling revealed selective, significant step-ups in insulin secretion that correlated anatomically with the location of solitary lesions confirmed surgically in 2 of 3 infants with congenital focal disease and in the child with insulinoma. Selective arterial calcium stimulation of the pancreas with hepatic venous sampling demonstrated markedly elevated baseline insulin levels throughout the pancreas of the infant with diffuse hyperinsulinism. CONCLUSIONS: The intravenous CaAIR is a safe and simple test for identifying infants with diffuse SUR1(-/-) hyperinsulinism or with focal congenital hyperinsulinism. Preoperative selective arterial calcium stimulation of the pancreas with hepatic venous sampling can localize focal lesions causing hyperinsulinism in children. The combination of these calcium stimulation tests may help distinguish focal lesions suitable for cure by local surgical resection.


Subject(s)
ATP-Binding Cassette Transporters , Calcium , Hyperinsulinism/congenital , Hyperinsulinism/diagnosis , Potassium Channels, Inwardly Rectifying , Potassium Channels , Receptors, Drug , Sulfonylurea Compounds/metabolism , Adolescent , Adult , Calcium/blood , Case-Control Studies , Child , Child, Preschool , Diagnosis, Differential , Diagnostic Techniques, Endocrine , Female , Humans , Hyperinsulinism/blood , Infant , Injections, Intravenous , Male , Potassium Channels/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
9.
J Biol Chem ; 275(13): 9270-7, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734066

ABSTRACT

Sur1 knockout mouse beta-cells lack K(ATP) channels and show spontaneous Ca(2+) action potentials equivalent to those seen in patients with persistent hyperinsulinemic hypoglycemia of infancy, but the mice are normoglycemic unless stressed. Sur1(-/-) islets lack first phase insulin secretion and exhibit an attenuated glucose-stimulated second phase secretion. Loss of the first phase leads to mild glucose intolerance, whereas reduced insulin output is consistent with observed neonatal hyperglycemia. Loss of K(ATP) channels impairs the rate of return to a basal secretory level after a fall in glucose concentration. This leads to increased hypoglycemia upon fasting and contributes to a very early, transient neonatal hypoglycemia. Whereas persistent hyperinsulinemic hypoglycemia of infancy underscores the importance of the K(ATP)-dependent ionic pathway in control of insulin release, the Sur1(-/-) animals provide a novel model for study of K(ATP)-independent pathways that regulate insulin secretion.


Subject(s)
ATP-Binding Cassette Transporters , Insulin/metabolism , Models, Biological , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Potassium Channels/physiology , Receptors, Drug/genetics , Animals , Hypoglycemia , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Membrane Potentials , Mice , Mice, Knockout , Potassium Channels/metabolism , Receptors, Drug/metabolism , Sulfonylurea Compounds/metabolism , Sulfonylurea Receptors
10.
Kidney Int ; 57(3): 803-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720932

ABSTRACT

Familial hyperinsulinism, also known as persistent hyperinsulinemic hypoglycemia of infancy (PHHI), is a genetic disease characterized by mild to severe hypoglycemia in the presence of inappropriately high levels of insulin. The recessive form is caused by mutations in the adenosine 5'-triphosphate (ATP)-sensitive K+ channel (KATP channel) present in the plasma membrane of pancreatic beta-cells. This channel is formed by two subunits, the high-affinity sulfonylurea receptor, SUR1, and KIR6.2, a member of the inwardly rectifying family of K+ channels. KATP channels regulate insulin secretion by linking membrane excitability with glucose metabolism. Approximately 50 mutations, in both channel subunits, that abolish or alter the regulation of beta-cell KATP channels have been identified in patients with the recessive form of PHHI.


Subject(s)
Adenosine Triphosphate/physiology , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Islets of Langerhans/metabolism , Potassium Channels/metabolism , Humans , Mutation/physiology , Potassium Channels/genetics
11.
Biochim Biophys Acta ; 1461(2): 285-303, 1999 Dec 06.
Article in English | MEDLINE | ID: mdl-10581362

ABSTRACT

The association of sulfonylurea receptors (SURs) with K(IR)6.x subunits to form ATP-sensitive K(+) channels presents perhaps the most unusual function known for members of the transport ATPase family. The integration of these two protein subunits extends well beyond conferring sensitivity to sulfonylureas. Recent studies indicate SUR-K(IR)6.x interactions are critical for all of the properties associated with native K(ATP) channels including quality control over surface expression, channel kinetics, inhibition and stimulation by Mg-nucleotides and response both to channel blockers like sulfonylureas and to potassium channel openers. K(ATP) channels are a unique example of the physiologic and medical importance of a transport ATPase and provide a paradigm for how other members of the family may interact with other ion channels.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Benzamides/pharmacology , Cell Line , Cytoplasm/metabolism , Electrochemistry , Endoplasmic Reticulum/metabolism , Glyburide/pharmacology , Humans , Magnesium/pharmacology , Models, Molecular , Potassium/pharmacology , Potassium Channels/chemistry , Potassium Channels/drug effects , Receptors, Drug/chemistry , Receptors, Drug/drug effects , Sulfonylurea Compounds/metabolism , Sulfonylurea Receptors , Tolbutamide/pharmacology
12.
J Biol Chem ; 274(29): 20628-32, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10400694

ABSTRACT

In beta cells from the pancreas, ATP-sensitive potassium channels, or KATP channels, are composed of two subunits, SUR1 and KIR6.2, assembled in a (SUR1/KIR6.2)4 stoichiometry. The correct stoichiometry of channels at the cell surface is tightly regulated by the presence of novel endoplasmic reticulum (ER) retention signals in SUR1 and KIR6.2; incompletely assembled KATP channels fail to exit the ER/cis-Golgi compartments. In addition to these retrograde signals, we show that the C terminus of SUR1 has an anterograde signal, composed in part of a dileucine motif and downstream phenylalanine, which is required for KATP channels to exit the ER/cis-Golgi compartments and transit to the cell surface. Deletion of as few as seven amino acids, including the phenylalanine, from SUR1 markedly reduces surface expression of KATP channels. Mutations leading to truncation of the C terminus of SUR1 are one cause of a severe, recessive form of persistent hyperinsulinemic hypoglycemia of infancy. We propose that the complete loss of beta cell KATP channel activity seen in this form of hyperinsulinism is a failure of KATP channels to traffic to the plasma membrane.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , COS Cells , DNA Primers , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Membrane Potentials , Molecular Sequence Data , Mutagenesis , Potassium Channels/chemistry , Potassium Channels/genetics , Receptors, Drug/chemistry , Receptors, Drug/genetics , Sulfonylurea Receptors
13.
Endocr Rev ; 20(2): 101-35, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10204114

ABSTRACT

KATP channels are a newly defined class of potassium channels based on the physical association of an ABC protein, the sulfonylurea receptor, and a K+ inward rectifier subunit. The beta-cell KATP channel is composed of SUR1, the high-affinity sulfonylurea receptor with multiple TMDs and two NBFs, and KIR6.2, a weak inward rectifier, in a 1:1 stoichiometry. The pore of the channel is formed by KIR6.2 in a tetrameric arrangement; the overall stoichiometry of active channels is (SUR1/KIR6.2)4. The two subunits form a tightly integrated whole. KIR6.2 can be expressed in the plasma membrane either by deletion of an ER retention signal at its C-terminal end or by high-level expression to overwhelm the retention mechanism. The single-channel conductance of the homomeric KIR6.2 channels is equivalent to SUR/KIR6.2 channels, but they differ in all other respects, including bursting behavior, pharmacological properties, sensitivity to ATP and ADP, and trafficking to the plasma membrane. Coexpression with SUR restores the normal channel properties. The key role KATP channel play in the regulation of insulin secretion in response to changes in glucose metabolism is underscored by the finding that a recessive form of persistent hyperinsulinemic hypoglycemia of infancy (PHHI) is caused by mutations in KATP channel subunits that result in the loss of channel activity. KATP channels set the resting membrane potential of beta-cells, and their loss results in a constitutive depolarization that allows voltage-gated Ca2+ channels to open spontaneously, increasing the cytosolic Ca2+ levels enough to trigger continuous release of insulin. The loss of KATP channels, in effect, uncouples the electrical activity of beta-cells from their metabolic activity. PHHI mutations have been informative on the function of SUR1 and regulation of KATP channels by adenine nucleotides. The results indicate that SUR1 is important in sensing nucleotide changes, as implied by its sequence similarity to other ABC proteins, in addition to being the drug sensor. An unexpected finding is that the inhibitory action of ATP appears to be through a site located on KIR6.2, whose affinity for ATP is modified by SUR1. A PHHI mutation, G1479R, in the second NBF of SUR1 forms active KATP channels that respond normally to ATP, but fail to activate with MgADP. The result implies that ATP tonically inhibits KATP channels, but that the ADP level in a fasting beta-cell antagonizes this inhibition. Decreases in the ADP level as glucose is metabolized result in KATP channel closure. Although KATP channels are the target for sulfonylureas used in the treatment of NIDDM, the available data suggest that the identified KATP channel mutations do not play a major role in diabetes. Understanding how KATP channels fit into the overall scheme of glucose homeostasis, on the other hand, promises insight into diabetes and other disorders of glucose metabolism, while understanding the structure and regulation of these channels offers potential for development of novel compounds to regulate cellular electrical activity.


Subject(s)
Adenosine Triphosphate/pharmacology , Potassium Channels , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 2/genetics , Humans , Hyperinsulinism/genetics , Infant, Newborn , Islets of Langerhans , Molecular Sequence Data , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/physiology
14.
FEBS Lett ; 445(1): 131-6, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-10069387

ABSTRACT

KATP channels are heteromultimers of SUR and KIR6.2. C-terminal truncation of KIR6.2 allows surface expression of the pore. KIR6.2deltaC35 channels display approximately 7-fold lower maximal open probability, approximately 35-fold reduced ATP sensitivity, reduced mean open time, a markedly increased transition rate from a burst into a long-lived closed state, and have no counterpart in vivo. SUR1 and SUR2A restore wild-type bursting, ATP sensitivity and increase channel density in the plasma membrane. The high IC50(ATP) of approximately 4 mM for KIR6.2deltaCK185Q channels results from the additive effects of SUR removal and KIR6.2 modification. The results demonstrate allosteric interaction(s) are essential for normal intrinsic activity, ATP inhibition, and trafficking of KATP channels.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Humans , Potassium Channels/genetics , Potassium Channels/physiology , Sulfonylurea Receptors
16.
Circ Res ; 83(11): 1132-43, 1998 Nov 30.
Article in English | MEDLINE | ID: mdl-9831708

ABSTRACT

ATP-sensitive potassium (KATP) channels in striated myocytes are heteromultimers of KIR6.2, a weak potassium inward rectifier, plus SUR2A, a low-affinity sulfonylurea receptor. We have cloned human KIR6.2 (huKIR6.2) and a huSUR2A that corresponds to the major, full-length splice variant identified by polymerase chain reaction analysis of human cardiac poly A+ mRNA. ATP- and glibenclamide-sensitive K+ channels were produced when both subunits were coexpressed in COSm6 and Chinese hamster ovary cells lacking endogenous KATP channels, but not when huSUR2A or huKIR6.2 were transfected alone. Recombinant channels activated by metabolic inhibition in cell-attached configuration or in inside-out patches with ATP-free internal solution were compared with sarcolemmal KATP channels in human ventricular cells. The single-channel conductance of approximately 80 pS measured at -40 mV in quasi-symmetrical approximately 150 mmol/L K+ solutions, the intraburst kinetics that were dependent on K+ driving force, and the weak inward rectification were indistinguishable for both channels. Similar to the native channels, huSUR2A/huKIR6.2 recombinant channels were inhibited by ATP at quasi-physiological free Mg2+ ( approximately 0. 7 mmol/L) or in the absence of Mg2+, with an apparent IC50 of approximately 20 micromol/L and a pseudo-Hill coefficient of approximately 1. They were "refreshed" by MgATP and stimulated by ADP in the presence of Mg2+ when inhibited by ATP. The huSUR2A/huKIR6.2 channels were stimulated by cromakalim and pinacidil in the presence of ATP and Mg2+ but were insensitive to diazoxide. The results suggest that reconstituted huSUR2A/huKIR6.2 channels represent KATP channels in sarcolemma of human cardiomyocytes and are an adequate experimental model with which to examine structure-function relationships, molecular physiology, and pharmacology of these channels from human heart.


Subject(s)
ATP-Binding Cassette Transporters , Cloning, Molecular , Myocardium/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Potassium Channels/physiology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Adolescent , Adult , Animals , CHO Cells , COS Cells , Child , Cricetinae , Cromakalim/pharmacology , DNA, Complementary/genetics , Diazoxide/pharmacology , Gene Library , Glyburide/pharmacology , Heart Ventricles/cytology , Heart Ventricles/ultrastructure , Humans , Magnesium/pharmacology , Middle Aged , Patch-Clamp Techniques , Pinacidil/pharmacology , Potassium Channel Blockers , Potassium Channels/drug effects , Rats , Receptors, Drug/drug effects , Receptors, Drug/genetics , Sarcolemma/chemistry , Sarcolemma/metabolism , Sulfonylurea Receptors , Tolbutamide/pharmacology , Transfection
17.
Hum Genet ; 103(3): 280-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9799081

ABSTRACT

The high-affinity sulfonylurea receptor (SUR1) gene regulates insulin secretion and may play a role in type 2 diabetes. A silent variant in exon 31 of SUR1 (AGG-->AGA) was detected by single-strand conformational polymorphism and genotypes were determined for 396 Mexican American subjects (289 non-diabetic). The normal and mutant alleles were designated G and A, respectively. Among non-diabetics, those with the AA genotype had higher fasting insulin values than those with the AG and GG genotypes (113.4 pmol/l for AA vs 82.8 pmol/l for AG/GG, P=0.043). Similar results were observed for 2-h insulin (849.6 pmol/l for AA vs 498.6 pmol/l for AG/GG, P=0.0003) and for the proinsulin to specific insulin ratio (0.068 for AA vs 0.056 for AG/GG, P=0.030). Specific insulin levels also differed significantly across the three genotypic classes (P=0.021). No differences in fasting glucose, body mass index, or waist circumference according to genotype were noted. Two-hour glucose was modestly higher in individuals with the AA genotype. Since we have previously reported linkage between SUR1 and hyperglycemia, the present association between a SUR1 variant and hyperinsulinemia in normal individuals from a high diabetes risk ethnic group raises the possibility of primary insulin hypersecretion as an antecedent of type 2 diabetes in at least some individuals from this population.


Subject(s)
ATP-Binding Cassette Transporters , Hyperinsulinism/ethnology , Insulin/blood , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Adult , Blood Glucose/analysis , DNA Mutational Analysis , Diabetes Mellitus, Type 2/etiology , Female , Genotype , Humans , Male , Membrane Proteins/genetics , Polymorphism, Genetic/genetics , Proinsulin/blood , Sulfonylurea Receptors , United States/ethnology
18.
EMBO J ; 17(19): 5529-35, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9755153

ABSTRACT

KATP channels are composed of a small inwardly rectifying K+ channel subunit, either KIR6.1 or KIR6.2, plus a sulfonylurea receptor, SUR1 or SUR2 (A or B), which belong to the ATP-binding cassette superfamily. SUR1/KIR6.2 reconstitute the neuronal/pancreatic beta-cell channel, whereas SUR2A/KIR6.2 and SUR2B/KIR6.1 (or KIR6.2) are proposed to reconstitute the cardiac and the vascular-smooth-muscle-type KATP channels, respectively. We report that potassium channel openers (KCOs) bind to and act through SURs and that binding to SUR1, SUR2A and SUR2B requires ATP. Non-hydrolysable ATP-analogues do not support binding, and Mg2+ or Mn2+ are required. Point mutations in the Walker A motifs or linker regions of both nucleotide-binding folds (NBFs) abolish or weaken [3H]P1075 binding to SUR2B, rendering reconstituted SUR2B/KIR6.2 channels insensitive towards KCOs. The C-terminus of SUR affects KCO affinity with SUR2B approximately SUR1 > SUR2A. KCOs belonging to different structural classes inhibited specific [3H]P1075 binding to SUR2B in a monophasic manner, with the exception of minoxidil sulfate, which induced a biphasic displacement. The affinities of KCO binding to SUR2B were 3.5-8-fold higher than their potencies for activation of SUR2B/KIR6.2 channels. The results establish that SURs are the KCO receptors of KATP channels and suggest that KCO binding requires a conformational change induced by ATP hydrolysis in both NBFs.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Animals , Binding Sites/genetics , Binding, Competitive , Cricetinae , Diazoxide/metabolism , Glyburide/metabolism , Guanidines/metabolism , Humans , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Pinacidil/metabolism , Potassium Channels/agonists , Potassium Channels/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyridines/metabolism , Receptors, Drug/genetics , Recombinant Proteins/metabolism , Sulfonylurea Receptors
20.
Diabetes ; 47(4): 598-605, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9568693

ABSTRACT

The high-affinity sulfonylurea receptor (SUR1) is, as a subunit of the ATP-sensitive potassium channel, an important regulator of insulin secretion in the pancreatic beta-cell. The aim of this study was to examine if genetic variability of the SUR1 gene was associated with NIDDM or altered pancreatic beta-cell function. Mutational analysis of all the 39 SUR1 exons, including intron-exon boundaries, in 63 NIDDM patients revealed two missense variants, five silent variants in the coding region, and four intron variants. The two missense variants (Asp673Asn and Ser1369Ala) and two sequence variants (ACC-->ACT, Thr759Thr and a c-->t intron variant in position -3 of the exon 16 splice acceptor site) were examined for association with NIDDM and for a possible influence on insulin and C-peptide secretion after intravenous glucose and tolbutamide loads in a random sample of unrelated, healthy, young Danish Caucasians. The Asp673Asn variant in exon 14 was only identified in one NIDDM patient, and the allelic frequency of the Ser1369Ala was similar among 247 control subjects (0.38 [95% CI 0.34-0.42]) and 406 NIDDM patients (0.40 [0.37-0.43]). The allelic frequency of the silent exon 18 Thr775Thr variant was 0.051 (0.035-0.067) in NIDDM patients (n=392) and 0.027 (0.013-0.041) in control subjects (n=246; chi2=4.99, P=0.03). The allelic frequency of the intron variant was similar among NIDDM patients (0.45 [0.42-0.48]) and control subjects (0.44 [0.40-0.48]). Of 386 NIDDM patients, 17 had the combined genotype exon 18 C/T and intron -3c/-3t (0.044 [0.024-0.064]), whereas 3 of 243 control subjects had the same combined genotype (0.012 [0-0.026]; chi2=4.87, P=0.03; odds ratio: 3.69 [1.07-12.71]). Of 380 unrelated, healthy, young Danish Caucasians, 10 (0.026 [0.010-0.042]) had the combined at-risk genotype. These subjects had, on average, a 50% reduction in serum C-peptide and a 40% reduction in serum insulin responses upon tolbutamide injection (P=0.002 and P=0.05, respectively) but normal serum C-peptide and insulin responses upon glucose injection. In conclusion, a silent polymorphism in exon 18 of the SUR1 gene is associated with NIDDM in a Danish Caucasian population. In combination with an intron variant, the association is higher, and young, healthy carriers of the intragenic combination have reduced serum C-peptide and insulin responses to a tolbutamide load.


Subject(s)
ATP-Binding Cassette Transporters , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Tolbutamide , Adolescent , Adult , Amino Acid Substitution , Diabetes Mellitus, Type 2/physiopathology , Exons , Female , Genotype , Glucose Tolerance Test , Humans , Insulin Secretion , Introns , Male , Molecular Sequence Data , Point Mutation , Polymorphism, Single-Stranded Conformational , Sulfonylurea Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...