Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Clin Genet ; 87(5): 448-54, 2015 May.
Article in English | MEDLINE | ID: mdl-24814349

ABSTRACT

ABCC8 encodes a subunit of the ß-cell potassium channel (KATP ) whose loss of function is responsible for congenital hyperinsulinism (CHI). Patients with two recessive mutations of ABCC8 typically have severe diffuse forms of CHI unresponsive to diazoxide. Some dominant ABCC8 mutations are responsible for a subset of diffuse diazoxide-unresponsive forms of CHI. We report the analysis of 21 different ABCC8 mutations identified in 25 probands with diazoxide-unresponsive diffuse CHI and carrying a single mutation in ABCC8. Nine missense ABCC8 mutations were subjected to in vitro expression studies testing traffic efficiency and responses of mutant channels to activation by MgADP and diazoxide. Eight of the 9 missense mutations exhibited normal trafficking. Seven of the 8 mutants reaching the plasma membrane had dramatically reduced response to MgADP or to diazoxide (<10% of wild-type response). In our cohort, dominant KATP mutations account for 22% of the children with diffuse unresponsive-diazoxide CHI. Their clinical phenotype being indistinguishable from that of children with focal CHI and diffuse CHI forms due to two recessive KATP mutations, we show that functional testing is essential to make the most reliable diagnosis and offer appropriate genetic counseling.


Subject(s)
Alleles , Congenital Hyperinsulinism/drug therapy , Congenital Hyperinsulinism/genetics , Diazoxide/therapeutic use , Drug Resistance/genetics , Mutation , Sulfonylurea Receptors/genetics , Amino Acid Substitution , Congenital Hyperinsulinism/diagnosis , DNA Mutational Analysis , Female , Genes, Recessive , Humans , Infant , Infant, Newborn , Male , Phenotype , Sulfonylurea Receptors/metabolism , Treatment Outcome
2.
J Clin Endocrinol Metab ; 98(2): E355-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23275527

ABSTRACT

CONTEXT: Hypoglycemia due to congenital hyperinsulinism (HI) is caused by mutations in 9 genes. OBJECTIVE: Our objective was to correlate genotype with phenotype in 417 children with HI. METHODS: Mutation analysis was carried out for the ATP-sensitive potassium (KATP) channel genes (ABCC8 and KCNJ11), GLUD1, and GCK with supplemental screening of rarer genes, HADH, UCP2, HNF4A, HNF1A, and SLC16A1. RESULTS: Mutations were identified in 91% (272 of 298) of diazoxide-unresponsive probands (ABCC8, KCNJ11, and GCK), and in 47% (56 of 118) of diazoxide-responsive probands (ABCC8, KCNJ11, GLUD1, HADH, UCP2, HNF4A, and HNF1A). In diazoxide-unresponsive diffuse probands, 89% (109 of 122) carried KATP mutations; 2% (2 of 122) had GCK mutations. In mutation-positive diazoxide-responsive probands, 42% were GLUD1, 41% were dominant KATP mutations, and 16% were in rare genes (HADH, UCP2, HNF4A, and HNF1A). Of the 183 unique KATP mutations, 70% were novel at the time of identification. Focal HI accounted for 53% (149 of 282) of diazoxide-unresponsive probands; monoallelic recessive KATP mutations were detectable in 97% (145 of 149) of these cases (maternal transmission excluded in all cases tested). The presence of a monoallelic recessive KATP mutation predicted focal HI with 97% sensitivity and 90% specificity. CONCLUSIONS: Genotype to phenotype correlations were most successful in children with GLUD1, GCK, and recessive KATP mutations. Correlations were complicated by the high frequency of novel missense KATP mutations that were uncharacterized, because such defects might be either recessive or dominant and, if dominant, be either responsive or unresponsive to diazoxide. Accurate and timely prediction of phenotype based on genotype is critical to limit exposure to persistent hypoglycemia in infants and children with congenital HI.


Subject(s)
Congenital Hyperinsulinism/genetics , Genetic Association Studies , Glutamate Dehydrogenase/genetics , Potassium Channels/genetics , Protein Serine-Threonine Kinases/genetics , Child , Child, Preschool , Congenital Hyperinsulinism/diagnosis , Female , Genotype , Germinal Center Kinases , Humans , Infant , Male , Mutation , Phenotype
3.
Am J Physiol Endocrinol Metab ; 283(2): E207-16, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12110524

ABSTRACT

ATP-sensitive potassium (K(ATP)) channels are inhibited by intracellular ATP and activated by ADP. Nutrient oxidation in beta-cells leads to a rise in [ATP]-to-[ADP] ratios, which in turn leads to reduced K(ATP) channel activity, depolarization, voltage-dependent Ca(2+) channel activation, Ca(2+) entry, and exocytosis. Persistent hyperinsulinemic hypoglycemia of infancy (HI) is a genetic disorder characterized by dysregulated insulin secretion and, although rare, causes severe mental retardation and epilepsy if left untreated. The last five or six years have seen rapid advance in understanding the molecular basis of K(ATP) channel activity and the molecular genetics of HI. In the majority of cases for which a genotype has been uncovered, causal HI mutations are found in one or the other of the two genes, SUR1 and Kir6.2, that encode the K(ATP) channel. This article will review studies that have defined the link between channel activity and defective insulin release and will consider implications for future understanding of the mechanisms of control of insulin secretion in normal and diseased states.


Subject(s)
Adenosine Triphosphate/physiology , Endocrine System Diseases/metabolism , Insulin/metabolism , Potassium Channels/metabolism , Animals , Finland/epidemiology , Humans , Hyperinsulinism/complications , Hyperinsulinism/congenital , Hyperinsulinism/epidemiology , Hyperinsulinism/genetics , Hypoglycemia/etiology , Incidence , Infant, Newborn , Insulin Secretion , Mutation , Potassium Channels/genetics
4.
J Biol Chem ; 276(44): 41270-8, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11546780

ABSTRACT

Sulfonylurea receptors (SURx) are multi-spanning transmembrane proteins of the ATP-binding cassette (ABC) family, which associate with Kir6.x to form ATP-sensitive potassium channels. Two models, with 13-17 transmembrane segments, have been proposed for SURx topologies. Recently, we demonstrated that the amino-terminal region of SUR1 contains 5 transmembrane segments, supporting the 17-transmembrane model. To investigate the topology of the complete full-length SUR1, two strategies were employed. Topology was probed by accessibility of introduced cysteines to a membrane-impermeable biotinylating reagent, biotin maleimide. Amino acid positions 6/26, 99, 159, 337, 567, 1051, and 1274 were accessible, therefore extracellular, whereas many endogenous and some introduced cysteines were inaccessible, thus likely cytoplasmic or intramembrane. These sites correspond to extracellular loops 1-3, 5-6, and 8 and the NH2 terminus, and intracellular loops 3-8 and COOH terminus in the 17-transmembrane model. Immunofluorescence was used to determine accessibility of epitope-tagged SUR1 in intact and permeabilized cells. Epitopes at positions 337 and 1050 (putative external loops 3 and 6) were labeled in intact cells, therefore external, whereas positions 485 and 1119 (putative internal loops 5 and 7) only were accessible after permeabilization and therefore internal. These results are compatible with the 17-transmembrane model with two pairs of transmembrane segments as possible reentrant loops.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Animals , Biotin/metabolism , COS Cells , Cysteine/metabolism , Models, Molecular , Mutagenesis , Patch-Clamp Techniques , Potassium Channels/chemistry , Potassium Channels/genetics , Receptors, Drug/chemistry , Receptors, Drug/genetics , Sulfonylurea Receptors
5.
Proc Natl Acad Sci U S A ; 98(5): 2882-7, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226335

ABSTRACT

The ATP-sensitive potassium channel (K(ATP)) regulates insulin secretion in pancreatic beta cells. Loss of functional K(ATP) channels because of mutations in either the SUR1 or Kir6.2 channel subunit causes persistent hyperinsulinemic hypoglycemia of infancy (PHHI). We investigated the molecular mechanism by which a single phenylalanine deletion in SUR1 (DeltaF1388) causes PHHI. Previous studies have shown that coexpression of DeltaF1388 SUR1 with Kir6.2 results in no channel activity. We demonstrate here that the lack of functional expression is due to failure of the mutant channel to traffic to the cell surface. Trafficking of K(ATP) channels requires that the endoplasmic reticulum-retention signal, RKR, present in both SUR1 and Kir6.2, be shielded during channel assembly. To ask whether DeltaF1388 SUR1 forms functional channels with Kir6.2, we inactivated the RKR signal in DeltaF1388 SUR1 by mutation to AAA (DeltaF1388 SUR1(AAA)). Inactivation of similar endoplasmic reticulum-retention signals in the cystic fibrosis transmembrane conductance regulator has been shown to partially overcome the trafficking defect of a cystic fibrosis transmembrane conductance regulator mutation, DeltaF508. We found that coexpression of DeltaF1388 SUR1(AAA) with Kir6.2 led to partial surface expression of the mutant channel. Moreover, mutant channels were active. Compared with wild-type channels, the mutant channels have reduced ATP sensitivity and do not respond to stimulation by MgADP or diazoxide. The RKR --> AAA mutation alone has no effect on channel properties. Our results establish defective trafficking of K(ATP) channels as a molecular basis of PHHI and show that F1388 in SUR1 is critical for normal trafficking and function of K(ATP) channels.


Subject(s)
ATP-Binding Cassette Transporters , Hyperinsulinism/genetics , Hypoglycemia/genetics , Mutation , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Amino Acid Motifs , Animals , COS Cells , Hyperinsulinism/complications , Hypoglycemia/complications , Membrane Potentials , Potassium Channels/genetics , Potassium Channels/physiology , Protein Transport , Receptors, Drug/genetics , Sulfonylurea Receptors
6.
J Gen Physiol ; 116(5): 599-608, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11055989

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP(2)) activates K(ATP) and other inward rectifier (Kir) channels. To determine residues important for PIP(2) regulation, we have systematically mutated each positive charge in the COOH terminus of Kir6.2 to alanine. The effects of these mutations on channel function were examined using (86)Rb efflux assays on intact cells and inside-out patch-clamp methods. Both methods identify essentially the same basic residues in two narrow regions (176-222 and 301-314) in the COOH terminus that are important for the maintenance of channel function and interaction with PIP(2). Only one residue (R201A) simultaneously affected ATP and PIP(2) sensitivity, which is consistent with the notion that these ligands, while functionally competitive, are unlikely to bind to identical sites. Strikingly, none of 13 basic residues in the terminal portion (residues 315-390) of the COOH terminus affected channel function when neutralized. The data help to define the structural requirements for PIP(2) sensitivity of K(ATP) channels. Moreover, the regions and residues defined in this study parallel those uncovered in recent studies of PIP(2) sensitivity in other inward rectifier channels, indicating a common structural basis for PIP(2) regulation.


Subject(s)
Adenosine Triphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Cell Culture Techniques , Humans , Ligands , Phosphatidylinositol 4,5-Diphosphate/pharmacokinetics , Point Mutation , Potassium Channels/drug effects , Potassium Channels/genetics , Structure-Activity Relationship
7.
Biophys J ; 78(5): 2334-48, 2000 May.
Article in English | MEDLINE | ID: mdl-10777731

ABSTRACT

K(ATP) channels can be formed from Kir6.2 subunits with or without SUR1. The open-state stability of K(ATP) channels can be increased or reduced by mutations throughout the Kir6.2 subunit, and is increased by application of PIP(2) to the cytoplasmic membrane. Increase of open-state stability is manifested as an increase in the channel open probability in the absence of ATP (Po(zero)) and a correlated decrease in sensitivity to inhibition by ATP. Single channel lifetime analyses were performed on wild-type and I154C mutant channels expressed with, and without, SUR1. Channel kinetics include a single, invariant, open duration; an invariant, brief, closed duration; and longer closed events consisting of a "mixture of exponentials," which are prolonged in ATP and shortened after PIP(2) treatment. The steady-state and kinetic data cannot be accounted for by assuming that ATP binds to the channel and causes a gate to close. Rather, we show that they can be explained by models that assume the following regarding the gating behavior: 1) the channel undergoes ATP-insensitive transitions from the open state to a short closed state (C(f)) and to a longer-lived closed state (C(0)); 2) the C(0) state is destabilized in the presence of SUR1; and 3) ATP can access this C(0) state, stabilizing it and thereby inhibiting macroscopic currents. The effect of PIP(2) and mutations that stabilize the open state is then to shift the equilibrium of the "critical transition" from the open state to the ATP-accessible C(0) state toward the O state, reducing accessibility of the C(0) state, and hence reducing ATP sensitivity.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Animals , Biophysical Phenomena , Biophysics , COS Cells , Ion Channel Gating , Kinetics , Models, Biological , Mutagenesis, Site-Directed , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Structure, Quaternary , Receptors, Drug/chemistry , Receptors, Drug/genetics , Receptors, Drug/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfonylurea Receptors
8.
Proc Natl Acad Sci U S A ; 97(2): 937-41, 2000 Jan 18.
Article in English | MEDLINE | ID: mdl-10639183

ABSTRACT

ATP-sensitive potassium channels (K(ATP) channels) regulate cell excitability in response to metabolic changes. K(ATP) channels are formed as a complex of a sulfonylurea receptor (SURx), a member of the ATP-binding cassette protein family, and an inward rectifier K(+) channel subunit (Kir6.x). Membrane phospholipids, in particular phosphatidylinositol (PI) 4,5-bisphosphate (PIP(2)), activate K(ATP) channels and antagonize ATP inhibition of K(ATP) channels when applied to inside-out membrane patches. To examine the physiological relevance of this regulatory mechanism, we manipulated membrane PIP(2) levels by expressing either the wild-type or an inactive form of PI-4-phosphate 5-kinase (PIP5K) in COSm6 cells and examined the ATP sensitivity of coexpressed K(ATP) channels. Channels from cells expressing the wild-type PIP5K have a 6-fold lower ATP sensitivity (K(1/2), the half maximal inhibitory concentration, approximately 60 microM) than the sensitivities from control cells (K(1/2) approximately 10 microM). An inactive form of the PIP5K had little effect on the K(1/2) of wild-type channels but increased the ATP-sensitivity of a mutant K(ATP) channel that has an intrinsically lower ATP sensitivity (from K(1/2) approximately 450 microM to K(1/2) approximately 100 microM), suggesting a decrease in membrane PIP(2) levels as a consequence of a dominant-negative effect of the inactive PIP5K. These results show that PIP5K activity, which regulates PIP(2) and PI-3,4,5-P(3) levels, is a significant determinant of the physiological nucleotide sensitivity of K(ATP) channels.


Subject(s)
Adenosine Triphosphate/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Potassium Channels/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/physiology , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/drug effects , Mutation , Patch-Clamp Techniques , Phosphatidylinositols/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Potassium Channel Blockers , Potassium Channels/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
10.
J Physiol ; 515 ( Pt 1): 19-30, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9925874

ABSTRACT

1. To gain insight into the role of the cytoplasmic regions of the Kir6.2 subunit in regulating channel activity, we have expressed the sulphonylurea receptor SUR1 with Kir6.2 subunits containing systematic truncations of the N- and C-termini. Up to 30 amino acids could be truncated from the N-terminus, and up to 36 amino acids from the C-terminus without loss of functional channels in co-expression with SUR1. Furthermore, Kir6.2DeltaC25 and Kir6. 2DeltaC36 subunits expressed functional channels in the absence of SUR1. 2. In co-expression with SUR1, N-terminal truncations increased Ki,ATP ([ATP] causing half-maximal inhibition of channel activity) by as much as 10-fold, accompanied by an increase in the ATP-insensitive open probability, whereas the C-terminal truncations did not affect the ATP sensitivity of co-expressed channels. 3. A mutation in the near C-terminal region, K185Q, reduced ATP sensitivity of co-expressed channels by approximately 30-fold, and on the Kir6.2DeltaN2-30 background, this mutation decreased ATP sensitivity of co-expressed channels by approximately 400-fold. 4. Each of these mutations also reduced the sensitivity to inhibition by ADP, AMP and adenosine tetraphosphate. 5. The results can be quantitatively explained by assuming that the N-terminal deletions stabilize the ATP-independent open state, whereas the Kir6.2K185Q mutation may alter the stability of ATP binding. These two effects are energetically additive, causing the large reduction of ATP sensitivity in the double mutant channels.


Subject(s)
Adenosine Triphosphate/pharmacology , Membrane Proteins , Potassium Channel Blockers , Potassium Channels, Inwardly Rectifying , Saccharomyces cerevisiae Proteins , ATP-Binding Cassette Transporters , Adenine Nucleotides/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Animals , Cell Line , Energy Metabolism/drug effects , Energy Metabolism/genetics , Glycosyltransferases , Ion Channel Gating/genetics , Ion Channel Gating/physiology , KATP Channels , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium Channels/biosynthesis , Potassium Channels/genetics , Rats , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/biosynthesis , Rubidium/metabolism , Rubidium Radioisotopes , Sequence Deletion/physiology
11.
Science ; 282(5391): 1138-41, 1998 Nov 06.
Article in English | MEDLINE | ID: mdl-9804554

ABSTRACT

Adenosine triphosphate (ATP)-sensitive potassium (KATP) channels couple cell metabolism to electrical activity. Phosphatidylinositol phosphates (PIPs) profoundly antagonized ATP inhibition of KATP channels when applied to inside-out membrane patches. It is proposed that membrane-incorporated PIPs can bind to positive charges in the cytoplasmic region of the channel's Kir6.2 subunit, stabilizing the open state of the channel and antagonizing the inhibitory effect of ATP. The tremendous effect of PIPs on ATP sensitivity suggests that in vivo alterations of membrane PIP levels will have substantial effects on KATP channel activity and hence on the gain of metabolism-excitation coupling.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , COS Cells , Cell Line , Islets of Langerhans/metabolism , Mutation , Myocardium/cytology , Myocardium/metabolism , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphatidylinositol Phosphates/pharmacology , Potassium Channels/chemistry , Potassium Channels/genetics , Receptors, Drug/metabolism , Recombinant Fusion Proteins/metabolism , Sulfonylurea Receptors
12.
Diabetes ; 47(7): 1145-51, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9648840

ABSTRACT

The ATP-sensitive potassium channel, K(ATP) channel, a functional complex of the sulfonylurea receptor 1, SUR1, and an inward rectifier potassium channel subunit, Kir6.2, regulates insulin secretion in the pancreas. Mutations in both the Kir6.2 and SUR1 genes are associated with persistent hyperinsulinemic hypoglycemia of infancy (PHHI), a disorder of pancreatic beta-cell function characterized by excess insulin secretion and hypoglycemia. We have studied the functional properties of novel SUR1 mutations identified in PHHI patients, including H125Q, N188S, F591L, T1139M, R1215Q, G1382S, and R1394H. R1394H and deltaF1388 SUR1, a previously identified PHHI mutation, resulted in no functional channels when coexpressed with Kir6.2 in COS cells, while H125Q, N188S, F591L, T1139M, R1215Q, and G1382S SUR1 generated functional channels in the absence of ATP. With the exception of N188S and H125Q, all mutants had reduced response to stimulation by MgADP. These results indicate that lack of, or reduction of, K(ATP) channel sensitivity to MgADP is a common molecular defect associated with the disease. The mutant channels also showed varied response to activation by the potassium channel opener diazoxide. Because these mutations are distributed throughout the molecule, our data have new implications for structure-function relationships of the K(ATP) channel, suggesting that structural elements in SUR1 outside of the two nucleotide-binding folds are also important in regulating channel activity.


Subject(s)
ATP-Binding Cassette Transporters , Hyperinsulinism/complications , Hyperinsulinism/genetics , Hypoglycemia/genetics , Mutagenesis, Site-Directed , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Alleles , Animals , COS Cells , Cricetinae , Diazoxide/pharmacology , Humans , Infant , Infant, Newborn , Insulin/metabolism , Insulin Secretion , Mice , Pancreas/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Rubidium Radioisotopes/metabolism , Sulfonylurea Receptors , Transfection
13.
Hum Mol Genet ; 7(7): 1119-28, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9618169

ABSTRACT

Familial hyperinsulinism (HI) is a disorder characterized by dysregulation of insulin secretion and profound hypoglycemia. Mutations in both the Kir6.2 and sulfonylurea receptor (SUR1) genes have been associated with the autosomal recessive form of this disorder. In this study, the spectrum and frequency of SUR1 mutations in HI and their significance to clinical manifestations of the disease were investigated by screening 45 HI probands of various ethnic origins for mutations in the SUR1 gene. Single-strand conformation polymorphism (SSCP) and nucleotide sequence analyses of genomic DNA revealed a total of 17 novel and three previously described mutations in SUR1 . The novel mutations comprised one nonsense and 10 missense mutations, two deletions, three mutations in consensus splice-site sequences and an in-frame insertion of six nucleotides. One mutation occurred in the first nucleotide binding domain (NBF-1) of the SUR1 molecule and another eight mutations were located in the second nucleotide binding domain (NBF-2), including two at highly conserved amino acid residues within the Walker A sequence motif. The majority of the remaining mutations was distributed throughout the three putative transmembrane domains of the SUR1 protein. With the exception of the 3993-9G-->A mutation, which was detected on 4.5% (4/88) disease chromosomes, allelic frequencies for the identified mutations varied between 1.1 and 2.3% for HI chromosomes, indicating that each mutation was rare within the patient cohort. The clinical manifestations of HI in those patients homozygous for mutations in the SUR1 gene are described. In contrast with the allelic homogeneity of HI previously described in Ashkenazi Jewish patients, these findings suggest that a large degree of allelic heterogeneity at the SUR1 locus exists in non-Ashkenazi HI patients. These data have important implications for genetic counseling and prenatal diagnosis of HI, and also provide a basis to further elucidate the molecular mechanisms underlying the pathophysiology of this disease.


Subject(s)
ATP-Binding Cassette Transporters , Genetic Carrier Screening , Hyperinsulinism/genetics , Potassium Channels, Inwardly Rectifying , Alternative Splicing , Amino Acid Sequence/genetics , Codon, Nonsense/genetics , Cohort Studies , DNA Mutational Analysis , Humans , Mutagenesis, Insertional , Mutation/genetics , Mutation, Missense , Potassium Channels/genetics , Receptors, Drug/genetics , Sequence Deletion , Sulfonylurea Receptors
14.
J Gen Physiol ; 110(2): 141-53, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9236207

ABSTRACT

KATP channels are a functional complex of sulphonylurea receptor (SUR1, SUR2) and inward rectifier K+ (Kir6.1, Kir6.2) channel subunits. We have studied the role of the putative pore forming subunit (Kir6.2) in regulation of rectification and gating of KATP channels generated by transfection of SUR1 and Kir6.2 cDNAs in COSm6 cells. In the absence of internal polyvalent cations, the current-voltage relationship is sigmoidal. Mg2+ or spermine4+ (spm) each induces a mild inward rectification. Mutation of the asparagine at position 160 in Kir6.2 to aspartate (N160D) or glutamate (N160E) increases the degree of rectification induced by Mg2+ or spermine4+, whereas wild-type rectification is still observed after mutation to other neutral residues (alanine-N160A, glutamine-N160Q). These results are consistent with this residue lining the pore of the channel and contributing to the binding of these cations, as demonstrated for the equivalent site in homomeric ROMK1 (Kir1.1) channels. Since Kir6.2 contains no consensus ATP binding site, whereas SUR1 does, inhibition by ATP has been assumed to depend on interactions with SUR1. However, we found that the [ATP] causing half-maximal inhibition of current (Ki) was affected by mutation of N160. Channels formed from N160D or N160Q mutant subunits had lower apparent sensitivity to ATP (Ki,N160D = 46.1 microM; Ki,N160Q = 62.9 microM) than wild-type, N160E, or N160A channels (Ki = 10.4, 17.7, 6.4 microM, respectively). This might suggest that ATP binding to the channel complex was altered, although examination of channel open probabilities indicates instead that the residue at position 160 alters the ATP-independent open probability, i.e., it controls the free energy of the open state, thereby affecting the "coupling" of ATP binding to channel inhibition. The results can be interpreted in terms of a kinetic scheme whereby the residue at Kir6.2 position 160 controls the rate constants governing transitions to and from the open state, without directly affecting ATP binding or unbinding transitions.


Subject(s)
Adenosine Triphosphate/physiology , Ion Channel Gating/physiology , Potassium Channels/physiology , Adenosine Triphosphate/metabolism , Asparagine/metabolism , Cell Line , Cloning, Molecular , Electric Stimulation , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Kinetics , Membrane Potentials/physiology , Mutation , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/genetics , Rubidium Radioisotopes , Spermine/pharmacology
15.
J Gen Physiol ; 110(6): 643-54, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9382893

ABSTRACT

KATP channels were reconstituted in COSm6 cells by coexpression of the sulfonylurea receptor SUR1 and the inward rectifier potassium channel Kir6.2. The role of the two nucleotide binding folds of SUR1 in regulation of KATP channel activity by nucleotides and diazoxide was investigated. Mutations in the linker region and the Walker B motif (Walker, J.E., M.J. Saraste, M.J. Runswick, and N.J. Gay. 1982. EMBO [Eur. Mol. Biol. Organ.] J. 1:945-951) of the second nucleotide binding fold, including G1479D, G1479R, G1485D, G1485R, Q1486H, and D1506A, all abolished stimulation by MgADP and diazoxide, with the exception of G1479R, which showed a small stimulatory response to diazoxide. Analogous mutations in the first nucleotide binding fold, including G827D, G827R, and Q834H, were still stimulated by diazoxide and MgADP, but with altered kinetics compared with the wild-type channel. None of the mutations altered the sensitivity of the channel to inhibition by ATP4-. We propose a model in which SUR1 sensitizes the KATP channel to ATP inhibition, and nucleotide hydrolysis at the nucleotide binding folds blocks this effect. MgADP and diazoxide are proposed to stabilize this desensitized state of the channel, and mutations at the nucleotide binding folds alter the response of channels to MgADP and diazoxide by altering nucleotide hydrolysis rates or the coupling of hydrolysis to channel activation.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Diphosphate/pharmacology , Antihypertensive Agents/pharmacology , Diazoxide/pharmacology , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Receptors, Drug/genetics , Adenosine Triphosphate/pharmacology , Animals , COS Cells/chemistry , COS Cells/physiology , Hydrolysis , Ion Channel Gating/drug effects , Kinetics , Magnesium/pharmacology , Mutagenesis, Site-Directed/physiology , Patch-Clamp Techniques , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Folding , Receptors, Drug/chemistry , Receptors, Drug/metabolism , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/metabolism , Sulfonylurea Receptors
16.
J Gen Physiol ; 110(6): 655-64, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9382894

ABSTRACT

ATP-sensitive potassium (KATP) channels link cellular metabolism to electrical activity in nerve, muscle, and endocrine tissues. They are formed as a functional complex of two unrelated subunits-a member of the Kir inward rectifier potassium channel family, and a sulfonylurea receptor (SUR), a member of the ATP-binding cassette transporter family, which includes cystic fibrosis transmembrane conductance regulators and multidrug resistance protein, regulators of chloride channel activity. This recent discovery has brought together proteins from two very distinct superfamilies in a novel functional complex. The pancreatic KATP channel is probably formed specifically of Kir6.2 and SUR1 isoforms. The relationship between SUR1 and Kir6.2 must be determined to understand how SUR1 and Kir6.2 interact to form this unique channel. We have used mutant Kir6.2 subunits and dimeric (SUR1-Kir6.2) constructs to examine the functional stoichiometry of the KATP channel. The data indicate that the KATP channel pore is lined by four Kir6.2 subunits, and that each Kir6.2 subunit requires one SUR1 subunit to generate a functional channel in an octameric or tetradimeric structure.


Subject(s)
ATP-Binding Cassette Transporters , Potassium Channels, Inwardly Rectifying , Potassium Channels/chemistry , Potassium Channels/physiology , Receptors, Drug/chemistry , Receptors, Drug/physiology , Adenosine Triphosphate/metabolism , Animals , COS Cells/chemistry , COS Cells/physiology , Patch-Clamp Techniques , Protein Structure, Tertiary , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/metabolism , Sulfonylurea Receptors
17.
Proc Natl Acad Sci U S A ; 93(21): 12014-9, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8876254

ABSTRACT

Two different approaches were used to examine the in vivo role of polyamines in causing inward rectification of potassium channels. In two-microelectrode voltage-clamp experiments, 24-hr incubation of Xenopus oocytes injected with 50 nl of difluoromethylornithine (5 mM) and methylglyoxal bis(guanylhydrazone) (1 mM) caused an approximate doubling of expressed Kir2.1 currents and relieved rectification by causing an approximately +10-mV shift of the voltage at which currents are half-maximally inhibited. Second, a putrescine auxotrophic, ornithine decarboxylase-deficient Chinese hamster ovary (O-CHO) cell line was stably transfected with the cDNA encoding Kir2.3. Withdrawal of putrescine from the medium led to rapid (1-day) loss of the instantaneous phase of Kir2.3 channel activation, consistent with a decline of intracellular putrescine levels. Four days after putrescine withdrawal, macroscopic conductance, assessed using an 86Rb+ flux assay, was approximately doubled, and this corresponded to a +30-mV shift of V1/2 of rectification. With increasing time after putrescine withdrawal, there was an increase in the slowest phase of current activation, corresponding to an increase in the spermine-to-spermidine ratio over time. These results provide direct evidence for a role of each polyamine in induction of rectification, and they further demonstrate that in vivo modulation of rectification is possible by manipulation of polyamine levels using genetic and pharmacological approaches.


Subject(s)
Polyamines/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Animals , CHO Cells , Cricetinae , Female , Kinetics , Oocytes/physiology , Ornithine Decarboxylase/deficiency , Patch-Clamp Techniques , Potassium Channels/biosynthesis , Putrescine/pharmacology , Recombinant Proteins/biosynthesis , Rubidium/metabolism , Transfection , Xenopus laevis
18.
Science ; 272(5269): 1785-7, 1996 Jun 21.
Article in English | MEDLINE | ID: mdl-8650576

ABSTRACT

Adenosine triphosphate (ATP)-sensitive potassium (KATP) channels couple the cellular metabolic state to electrical activity and are a critical link between blood glucose concentration and pancreatic insulin secretion. A mutation in the second nucleotide-binding fold (NBF2) of the sulfonylurea receptor (SUR) of an individual diagnosed with persistent hyperinsulinemic hypoglycemia of infancy generated KATP channels that could be opened by diazoxide but not in response to metabolic inhibition. The hamster SUR, containing the analogous mutation, had normal ATP sensitivity, but unlike wild-type channels, inhibition by ATP was not antagonized by adenosine diphosphate (ADP). Additional mutations in NBF2 resulted in the same phenotype, whereas an equivalent mutation in NBF1 showed normal sensitivity to MgADP. Thus, by binding to SUR NBF2 and antagonizing ATP inhibition of KATP++ channels, intracellular MgADP may regulate insulin secretion.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Insulin/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Cell Line, Transformed , Chlorocebus aethiops , Cricetinae , Diazoxide/pharmacology , Humans , Hyperinsulinism/genetics , Hypoglycemia/genetics , Insulin Secretion , Islets of Langerhans/metabolism , Molecular Sequence Data , Patch-Clamp Techniques , Point Mutation , Potassium Channels/drug effects , Potassium Channels/genetics , Receptors, Drug/drug effects , Receptors, Drug/genetics , Rubidium/metabolism , Sulfonylurea Receptors , Transfection
20.
J Biol Chem ; 270(50): 30221-9, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8530433

ABSTRACT

There is currently no effective therapy for human prion diseases. However, several polyanionic glycans, including pentosan sulfate and dextran sulfate, prolong the incubation time of scrapie in rodents, and inhibit the production of the scrapie isoform of the prion protein (PrPSc), the major component of infectious prions, in cultured neuroblastoma cells. We report here that pentosan sulfate and related compounds rapidly and dramatically reduce the amount of PrPC, the non-infectious precursor of PrPSc, present on the cell surface. This effect results primarily from the ability of these agents to stimulate endocytosis of PrPC, thereby causing a redistribution of the protein from the plasma membrane to the cell interior. Pentosan sulfate also causes a change in the ultrastructural localization of PrPC, such that a portion of the protein molecules are shifted into late endosomes and/or lysosomes. In addition, we demonstrate, using PrP-containing bacterial fusion proteins, that cultured cells express saturable and specific surface binding sites for PrP, many of which are glycosaminoglycan molecules. Our results raise the possibility that sulfated glycans inhibit prion production by altering the cellular localization of PrPC precursor, and they indicate that endogenous proteoglycans are likely to play an important role in the cellular metabolism of both PrPC and PrPSc.


Subject(s)
Endocytosis/drug effects , Polysaccharides/pharmacology , PrPC Proteins/metabolism , Animals , Base Sequence , CHO Cells , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , DNA Primers , Dextran Sulfate/pharmacology , Humans , Kinetics , Mice , Molecular Sequence Data , Neuroblastoma , Pentosan Sulfuric Polyester/pharmacology , Pentosyltransferases/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoric Diester Hydrolases/metabolism , Polymerase Chain Reaction , PrPC Proteins/analysis , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Sulfuric Acids/pharmacology , Transfection , Tumor Cells, Cultured , UDP Xylose-Protein Xylosyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...