Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Mol Cell Cardiol ; 52(2): 410-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21888913

ABSTRACT

AMPK is an important sensor of cellular energy levels. The aim of these studies was to investigate whether cardiac K(ATP) channels, which couple cellular energy metabolism to membrane excitability, are regulated by AMPK activity. We investigated effects of AMPK on rat ventricular K(ATP) channels using electrophysiological and biochemical approaches. Whole-cell K(ATP) channel current was activated by metabolic inhibition; this occurred more rapidly in the presence of AICAR (an AMPK activator). AICAR had no effects on K(ATP) channel activity recorded in the inside-out patch clamp configuration, but ZMP (the intracellular intermediate of AICAR) strongly activated K(ATP) channels. An AMPK-mediated effect is demonstrated by the finding that ZMP had no effect on K(ATP) channels in the presence of Compound C (an AMPK inhibitor). Recombinant AMPK activated Kir6.2/SUR2A channels in a manner that was dependent on the AMP concentration, whereas heat-inactivated AMPK was without effect. Using mass-spectrometry and co-immunoprecipitation approaches, we demonstrate that the AMPK α-subunit physically associates with K(ATP) channel subunits. Our data demonstrate that the cardiac K(ATP) channel function is directly regulated by AMPK activation. During metabolic stress, a small change in cellular AMP that activates AMPK can be a potential trigger for K(ATP) channel opening. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism/physiology , KATP Channels/metabolism , Adenosine Monophosphate/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , COS Cells , Chlorocebus aethiops , KATP Channels/agonists , KATP Channels/genetics , Male , Mice , Myocytes, Cardiac/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Signal Transduction , Stress, Physiological
2.
Heart Rhythm ; 6(7): 1038-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19419905

ABSTRACT

BACKGROUND: KCNH2 gene mutations disrupting rapid component of I(K) (I(Kr)) underlie type 2 congenital long QT syndrome (LQT2). Startled auditory stimuli are specific symptomatic triggers in LQT2, thus suggesting fast arrhythmogenic mechanism. OBJECTIVE: We investigated acute alpha(1A)- and cyclic adenosine monophosphate (cAMP)-related beta-adrenergic modulation of I(Kr) in HL-1 cardiomyocytes, wild type (WT)- and 2 LQT2-associated mutant Kv11.1 channels (Y43D- and K595E-Kv11.1) reconstituted in Chinese hamster ovary (CHO) cells. METHODS: I(Kr) and Kv11.1 currents were recorded using the whole-cell patch-clamp technique and confocal microscopy of HL-1 cardiomyocytes transfected with green fluorescent protein (GFP)-tagged pleckstrin homology domain of phospholipase C-delta(1) visualized fluctuations of membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) content. RESULTS: In HL-1 cardiomyocytes expressing human alpha(1A)-adrenoceptor, superfusion with phenylephrine significantly reduced I(Kr) amplitude, shifted current activation to more positive potentials, and accelerated kinetics of deactivation. Confocal images showed a decline of membrane PIP(2) content during phenylephrine exposure. Simultaneous application of adenylyl cyclase activator forskolin and phosphodiesterase inhibitor 3-isobutyl-1-methylxantine (IBMX) shifted I(Kr) activation to more negative potentials and decreased tail current amplitudes after depolarizations between +10 and +50 mV. In CHO cells, alpha(1A)-adrenoceptor activation downregulated WT-Kv11.1 channels and forskolin/IBMX produced a dual effect. Expressed alone, the Y43D-Kv11.1 or K595E-Kv11.1 channel had no measurable function. However, co-expression of WT-Kv11.1 and each mutant protein evoked currents with loss-of-function alterations but identical to WT-Kv11.1 alpha(1A)- and forskolin/IBMX-induced regulation. CONCLUSION: Acute adrenergic regulation of at least 2 Kv11.1 mutant channels is preserved as in WT-Kv11.1 and native I(Kr). Suppression of alpha(1A)-adrenoceptor-related transduction might have therapeutic implications in some cases of LQT2.


Subject(s)
Long QT Syndrome/physiopathology , Myocytes, Cardiac/physiology , Potassium Channels, Voltage-Gated/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adult , Animals , Cells, Cultured , Cricetinae , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Female , Humans , Long QT Syndrome/congenital , Long QT Syndrome/genetics , Microscopy, Confocal , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/genetics
3.
Hum Mutat ; 30(4): 557-63, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19306396

ABSTRACT

Long QT syndrome (LQTS) is an inherited disease involving mutations in the genes encoding a number of cardiac ion channels and a membrane adaptor protein. Among the genes that are responsible for LQTS, KCNE1 and KCNE2 are members of the KCNE family of genes, and function as ancillary subunits of Kv channels. The third KCNE gene, KCNE3, is expressed in cardiac myocytes and interacts with KCNQ1 to change the channel properties. However, KCNE3 has never been linked to LQTS. To investigate the association between KCNE3 and LQTS, we conducted a genetic screening of KCNE3 mutations and single nucleotide polymorphisms (SNPs) in 485 Japanese LQTS probands using DHPLC-WAVE system and direct sequencing. Consequently, we identified two KCNE3 missense mutations, located in the N- and C-terminal domains. The functional effects of these mutations were examined by heterologous expression systems using CHO cells stably expressing KCNQ1. One mutation, p.R99lambdaH was identified in a 76-year-old woman who suffered torsades de pointes (TdP) after administration of disopyramide. Another mutation, p.T4A was identified in a 16-year-old boy and 67-year-old woman. Although the boy carried another KCNH2 mutation, he was asymptomatic. On the other hand, the woman suffered from hypokalemia-induced TdP. In a series of electrophysiological analyses, the KCNQ1(Q1)+KCNE3(E3)-R99lambdaH channel significantly reduced outward current compared to Q1+E3-WT, though the current density of the Q1+E3-T4A channel displayed no statistical significance. This is the first report of KCNE3 mutations associated with LQTS. Screening for variants in the KCNE3 gene is of clinical importance for LQTS patients.


Subject(s)
Long QT Syndrome/genetics , Mutation, Missense , Potassium Channels, Voltage-Gated/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Female , Humans , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Long QT Syndrome/physiopathology , Male , Membrane Potentials , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/physiology , Protein Binding , Transfection
4.
Circ J ; 72(5): 694-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18441445

ABSTRACT

BACKGROUND: In the LQT2 form of long QT syndrome (LQTS), mutation sites are reported to correlate with clinical phenotypes in Caucasians, but the relationship in Asian patients remains unknown. The present study was designed to determine whether the location of KCNH2 mutations would influence the arrhythmic risk in LQT2 patients. METHODS AND RESULTS: In 118 genetically-confirmed LQT2 patients (69 families, 62 KCNH2 mutations), the ECG parameters, Schwartz scores, and the incidence of cardiac events, defined as syncope, aborted cardiac arrest, and sudden cardiac death, were evaluated. To examine the effect of mutation sites, the participants were divided accordingly: pore (n=56) and non-pore (n=62) groups. The corrected QTend interval was significantly greater in the pore than in the non-pore group (QTc; 522+/-63 ms vs 490+/-49 ms, p=0.002). In this study, the clinical course of each of the probands did not differ according to the mutation sites, whereas non-probands carrying the pore site mutation experienced their first cardiac events at significantly younger age than those with the non-pore site mutation (log-rank, p=0.0005). CONCLUSIONS: In a Japanese LQT2 cohort, family members with the pore site mutation were at higher arrhythmic risk than those with the non-pore site mutation.


Subject(s)
Asian People/genetics , Asian People/statistics & numerical data , Long QT Syndrome/ethnology , Long QT Syndrome/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Codon, Nonsense , Cohort Studies , Disease-Free Survival , Family Health , Female , Gene Deletion , Genetic Variation , Humans , Male , Middle Aged , Mutation, Missense , Risk Factors , Torsades de Pointes/ethnology , Torsades de Pointes/genetics
5.
Circ J ; 72(5): 687-93, 2008 May.
Article in English | MEDLINE | ID: mdl-18441444

ABSTRACT

BACKGROUND: The Jervell and Lange-Nielsen (JLN) syndrome is a variant of long QT syndromes (LQTS) and is associated with congenital deafness. The syndrome is caused by homozygous or compound heterozygous mutations in genes KCNQ1 and KCNE1, which are responsible for encoding the delayed rectifier repolarizing current, I(Ks). METHODS AND RESULTS: A novel and homozygous KCNQ1 mutation in a 23-year-old deaf woman with a prolonged QT interval and recurrent syncope in a Japanese family was identified. Genetic analyses revealed that the proband harbored a KCNQ1 missense mutation (W248F) located in the intracellular S4-S5 linker on both alleles. The same mutation was identified in both maternal and paternal families in a heterozygous manner. However, the family members of both sides had no clinical evidence of LQTS or hearing defects. Functional assays using a heterologous expression system revealed that W248F KCNQ1 plus KCNE1 channels reconstitute hardly measurable I(Ks) currents. In contrast, heterozygous wild-type/W248F KCNQ1 plus KCNE1 channels displayed biophysical properties similar to those of the wild-type KCNQ1 plus KCNE1 channels with a weak dominant-negative effect. CONCLUSION: In this study, we present a family with JLN syndrome. The electrophysiological properties of the mutant I(Ks) channels explain the pathophysiology underlying JLNS.


Subject(s)
Asian People/genetics , Jervell-Lange Nielsen Syndrome/genetics , KCNQ1 Potassium Channel/genetics , Mutation, Missense , Potassium Channels, Voltage-Gated/genetics , Adult , Animals , CHO Cells , Cricetinae , Cricetulus , Deafness/genetics , Family Health , Female , Genetic Testing , Heterozygote , Humans , Jervell-Lange Nielsen Syndrome/physiopathology , KCNQ1 Potassium Channel/physiology , Male , Membrane Potentials/physiology , Pedigree , Polymorphism, Single-Stranded Conformational , Potassium Channels, Voltage-Gated/physiology
6.
Heart Rhythm ; 4(3): 332-40, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17341399

ABSTRACT

BACKGROUND: Long QT syndromes (LQTS) are inherited diseases involving mutations to genes encoding a number of cardiac ion channels and a membrane adaptor protein. The MinK protein is a cardiac K-channel accessory subunit encoded by the KCNE1 gene, mutations of which are associated with the LQT5 form of LQTS. OBJECTIVE: The purpose of this study was to search for the KCNE1 mutations and clarify the function of those mutations. METHODS: We conducted a genetic screen of KCNE1 mutations in 151 Japanese LQTS patients using the denaturing high-performance liquid chromatography-WAVE system and direct sequencing. In two LQTS patients, we identified two KCNE1 missense mutations, located in the MinK N- and C-terminal domains. The functional effects of these mutations were examined by heterologous coexpression with KCNQ1 and KCNH2. RESULTS: One mutation, which was identified in a 67-year-old woman, A8V, was novel. Her electrocardiogram (ECG) revealed marked bradycardia and QT interval prolongation. Another mutation, R98W, was identified in a 19-year-old woman. She experienced syncope followed by palpitation in exercise. At rest, her ECG showed bradycardia with mild QT prolongation, which became more prominent during exercise. In electrophysiological analyses, R98W produced reduced I(Ks) currents with a positive shift in the half activation voltages. In addition, when the A8V mutation was coexpressed with KCNH2, this reduced current magnitude, which is suggestive of a modifier effect by the A8V KCNE1 mutation on I(Kr). CONCLUSION: KCNE1 mutations may be associated with mild LQTS phenotypes, and KCNE1 gene screening is of clinical importance for asymptomatic and mild LQTS patients.


Subject(s)
Asian People/genetics , Long QT Syndrome/genetics , Mutation, Missense , Phenotype , Potassium Channels, Voltage-Gated/genetics , Adult , Aged , Chromatography, High Pressure Liquid , ERG1 Potassium Channel , Electrocardiography , Electrophysiologic Techniques, Cardiac , Ether-A-Go-Go Potassium Channels/genetics , Female , Gene Expression/genetics , Genetic Testing , Genotype , Humans , Japan , KCNQ1 Potassium Channel/genetics , Polymorphism, Single Nucleotide , Research Design
7.
J Mol Cell Cardiol ; 42(3): 662-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17292394

ABSTRACT

Mutations in KCNQ1, the gene encoding the delayed rectifier K(+) channel in cardiac muscle, cause long QT syndrome (LQTS). We studied 3 families with LQTS, in whom a guanine to adenine change in the last base of exon 7 (c.1032G>A), previously reported as a common splice-site mutation, was identified. We performed quantitative measurements of exon-skipping KCNQ1 mRNAs caused by this mutation using real-time reverse transcription polymerase chain reaction. Compared with normal individuals who have minor fractions of splicing variants (Delta7-8: 0.1%, Delta8: 6.9%, of total KCNQ1 transcripts), the affected individuals showed remarkable increases of exon-skipping mRNAs (Delta7: 23.5%, Delta7-8: 16.8%, Delta8: 4.5%). Current recordings from Xenopus laevis oocytes heterologously expressing channels of wild-type (WT) or exon-skipping KCNQ1 (Delta7, Delta7-8, or Delta8) revealed that none of the mutants produced any measurable currents, and moreover they displayed mutant-specific degree of dominant-negative effects on WT currents, when co-expressed with WT. Confocal microscopy analysis showed that fluorescent protein-tagged WT was predominantly expressed on the plasma membrane, whereas the mutants showed intracellular distribution. When WT was co-expressed with mutants, the majority of WT co-localized with the mutants in the intracellular space. Finally, we provide evidence showing direct protein-protein interactions between WT and the mutants, by using fluorescence resonance energy transfer. Thus, the mutants may exert their dominant-negative effects by trapping WT intracellularly and thereby interfering its translocation to the plasma membrane. In conclusion, our data provide a mechanistic basis for the pathogenesis of LQTS caused by a splicing mutation in KCNQ1.


Subject(s)
KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Long QT Syndrome/metabolism , RNA Splicing/genetics , Animals , Base Sequence , Biophysical Phenomena , Biophysics , COS Cells , Chlorocebus aethiops , Electrophysiology , Exons/genetics , Long QT Syndrome/genetics , Mutation/genetics , Patch-Clamp Techniques , RNA, Messenger/genetics , Xenopus laevis
8.
Hum Mutat ; 28(2): 208, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17221872

ABSTRACT

Andersen-Tawil syndrome (ATS) is a rare inherited disorder characterized by periodic paralysis, mild dysmorphic features, and QT or QU prolongation with ventricular arrhythmias in electrocardiograms (ECGs). Mutations of KCNJ2, encoding the human inward rectifying potassium channel Kir 2.1, have been identified in patients with ATS. We aimed to clarify the genotype-phenotype correlations in ATS patients. We screened 23 clinically diagnosed ATS patients from 13 unrelated Japanese families. Ten different forms of KCNJ2 mutations were identified in the 23 ATS patients included in this study. Their ECGs showed normal QTc intervals and abnormal U waves with QUc prolongation and a variety of ventricular arrhythmias. Especially, bidirectional ventricular tachycardia (VT) was observed in 13 of 23 patients (57%). Periodic paralysis was seen in 13 of 23 carriers (57%), dysmorphic features in 17 (74%), and seizures during infancy in 4 (17%). Functional assays for the two novel KCNJ2 mutations (c. 200G>A (p. R67Q) and c. 436G>A (p. G146S)) displayed no functional inward rectifying currents in a heterologous expression system and showed strong dominant negative effects when co-expressed with wild-type KCNJ2 channels (91% and 84% reduction at -50 mV respectively compared to wild-type alone). Immunocytochemistry and confocal imaging revealed normal trafficking for mutant channels. In our study, all of the clinically diagnosed ATS patients had KCNJ2 mutations and showed a high penetrance with regard to the typical cardiac phenotypes: predominant U wave and ventricular arrhythmias, typically bidirectional VT.


Subject(s)
Andersen Syndrome/diagnosis , Genotype , Mutation , Phenotype , Potassium Channels, Inwardly Rectifying/genetics , Adolescent , Adult , Aged , Andersen Syndrome/diagnostic imaging , Andersen Syndrome/genetics , Animals , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , DNA Mutational Analysis , Echocardiography , Female , Humans , Male , Middle Aged , Paralyses, Familial Periodic/diagnostic imaging , Paralyses, Familial Periodic/genetics , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/physiology , Protein Structure, Tertiary , Tachycardia, Ventricular/diagnostic imaging , Tachycardia, Ventricular/genetics
9.
Am J Physiol Heart Circ Physiol ; 291(2): H543-51, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16501027

ABSTRACT

Cardiac ATP-sensitive K+ (K(ATP)) channels are formed by Kir6.2 and SUR2A subunits. We produced transgenic mice that express dominant negative Kir6.x pore-forming subunits (Kir6.1-AAA or Kir6.2-AAA) in cardiac myocytes by driving their expression with the alpha-myosin heavy chain promoter. Weight gain and development after birth of these mice were similar to nontransgenic mice, but an increased mortality was noted after the age of 4-5 mo. Transgenic mice lacked cardiac K(ATP) channel activity as assessed with patch clamp techniques. Consistent with a decreased current density observed at positive voltages, the action potential duration was increased in these mice. Some myocytes developed EADs after isoproterenol treatment. Hemodynamic measurements revealed no significant effects on ventricular function (apart from a slightly elevated heart rate), whereas in vivo electrophysiological recordings revealed a prolonged ventricular effective refractory period in transgenic mice. The transgenic mice tolerated stress less well as evident from treadmill stress tests. The proarrhythmogenic features and lack of adaptation to a stress response in transgenic mice suggest that these features are intrinsic to the myocardium and that K(ATP) channels in the myocardium have an important role in protecting the heart from lethal arrhythmias and adaptation to stress situations.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/physiology , Animals , Blotting, Western , Electrocardiography , Electrophysiology , Heart Ventricles/cytology , Hemodynamics/physiology , KATP Channels , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Pericardium/physiology , Physical Exertion/physiology , Promoter Regions, Genetic/genetics , RNA/biosynthesis , RNA/genetics , Refractory Period, Electrophysiological/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sarcolemma/metabolism , Subcellular Fractions/metabolism , Ventricular Function
10.
J Am Coll Cardiol ; 46(11): 2100-6, 2005 Dec 06.
Article in English | MEDLINE | ID: mdl-16325048

ABSTRACT

OBJECTIVES: We carried out a complete screening of the SCN5A gene in 38 Japanese patients with Brugada syndrome to investigate the genotype-phenotype relationship. BACKGROUND: The gene SCN5A encodes the pore-forming alpha-subunit of voltage-gated cardiac sodium (Na) channel, which plays an important role in heart excitation/contraction. Mutations of SCN5A have been identified in 15% of patients with Brugada syndrome. METHODS: In 38 unrelated patients with clinically diagnosed Brugada syndrome, we screened for SCN5A gene mutations using denaturing high-performance liquid chromatography and direct sequencing, and conducted a functional assay for identified mutations using whole-cell patch-clamp in heterologous expression system. RESULTS: Four heterozygous mutations were identified (T187I, D356N, K1578fs/52, and R1623X) in 4 of the 38 patients. All of them had bradyarrhythmic complications: three with sick sinus syndrome (SSS) and the other (D356N) with paroxysmal complete atrioventricular block. SCN5A-linked Brugada patients were associated with a higher incidence of bradyarrhythmia (4 of 4) than non-SCN5A-linked Brugada patients (2 of 34). Families with T187I and K1578fs/52 had widespread penetrance of SSS. Notably, the patient with K1578fs/52, who had been diagnosed as having familial SSS without any clinical signs of Brugada syndrome, showed a Brugada-type ST-segment elevation after intravenous administration of pilsicainide and programmed electrical stimulation-induced ventricular tachycardia. All of the mutations encoded non-functional Na channels, and thus were suggested to cause impulse propagation defect underlying bradyarrhythmias. CONCLUSIONS: Our findings suggest that loss-of-function SCN5A mutations resulting in Brugada syndrome are distinguished by profound bradyarrhythmias.


Subject(s)
Bradycardia/complications , Bradycardia/genetics , Bundle-Branch Block/complications , Bundle-Branch Block/genetics , Sodium Channels/genetics , Adult , Aged , Asian People/genetics , Chromatography, High Pressure Liquid , Codon, Nonsense , DNA Mutational Analysis , Female , Frameshift Mutation , Humans , Male , Middle Aged , Mutagenesis, Site-Directed , NAV1.5 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Syndrome
11.
J Biol Chem ; 280(46): 38464-70, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16170200

ABSTRACT

The regulation of ATP-sensitive potassium (K(ATP)) channel activity is complex and a multitude of factors determine their open probability. Physiologically and pathophysiologically, the most important of these are intracellular nucleotides, with a long-recognized role for glycolytically derived ATP in regulating channel activity. To identify novel regulatory subunits of the K(ATP) channel complex, we performed a two-hybrid protein-protein interaction screen, using as bait the mouse Kir6.2 C terminus. Screening a rat heart cDNA library, we identified two potential interacting proteins to be the glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and triose-phosphate isomerase. The veracity of interaction was verified by co-immunoprecipitation techniques in transfected mammalian cells. We additionally demonstrated that pyruvate kinase also interacts with Kir6.2 subunits. The physiological relevance of these interactions is illustrated by the demonstration that native Kir6.2 protein similarly interact with GAPDH and pyruvate kinase in rat heart membrane fractions and that Kir6.2 protein co-localize with these glycolytic enzymes in rat ventricular myocytes. The functional relevance of our findings is demonstrated by the ability of GAPDH or pyruvate kinase substrates to directly block the K(ATP) channel under patch clamp recording conditions. Taken together, our data provide direct evidence for the concept that key enzymes involved in glycolytic ATP production are part of a multisubunit K(ATP) channel protein complex. Our data are consistent with the concept that the activity of these enzymes (possibly by ATP formation in the immediate intracellular microenvironment of this macromolecular K(ATP) channel complex) causes channel closure.


Subject(s)
Gene Expression Regulation , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/chemistry , Pyruvate Kinase/chemistry , Triose-Phosphate Isomerase/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Bacteria/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , DNA/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Glycolysis , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Heart Ventricles/metabolism , Hypoxia , Immunoglobulin G/chemistry , Immunoprecipitation , Kinetics , Mice , Microscopy, Fluorescence , Muscle Cells/metabolism , Mutation , Myocardium/metabolism , Patch-Clamp Techniques , Potassium/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Protein Structure, Tertiary , Pyruvate Kinase/metabolism , Rats , Rats, Sprague-Dawley , Transfection , Two-Hybrid System Techniques
12.
Pediatr Res ; 58(2): 185-92, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16085792

ABSTRACT

Prevailing data suggest that sarcolemmal ATP-sensitive (K(ATP)) channels in the adult heart consist of Kir6.2 and SUR2A subunits, but the expression of other K(ATP) channel subunits (including SUR1, SUR2B, and Kir6.1) is poorly defined. The situation is even less clear for the immature heart, which shows a remarkable resistance to hypoxia and metabolic stress. The hypoxia-induced action potential shortening and opening of sarcolemmal K(ATP) channels that occurs in adults is less prominent in the immature heart. This might be due in part to the different biophysical and pharmacological properties of K(ATP) channels of immature and adult K(ATP) channels. Because these properties are largely conferred by subunit composition, it is important to examine the relative expression levels of the various K(ATP) channel subunits during maturation. We therefore used RNAse protection assays, reverse transcription-PCR approaches, and Western blotting to characterize the mRNA and protein expression profiles of K(ATP) channel subunits in fetal, neonatal, and adult mouse heart. Our data indicate that each of the K(ATP) channel subunits (Kir6.1, Kir6.2, SUR1, SUR2A, and SUR2B) is expressed in the mouse heart at all of the developmental time points studied. However, the expression level of each of the subunits is low in the fetal heart and progressively increases with maturation. Each of the subunits seems to be expressed in ventricular myocytes with a subcellular expression pattern matching that found in the adult. Our data suggest that the K(ATP) channel composition may change during maturation, which has important implications for K(ATP) channel function in the developing heart.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Gene Expression Regulation, Developmental , Heart/embryology , Multidrug Resistance-Associated Proteins/biosynthesis , Myocardium/metabolism , Potassium Channels, Inwardly Rectifying/biosynthesis , Adenosine Triphosphate/metabolism , Alternative Splicing , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , DNA Primers/chemistry , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , KATP Channels , Mice , Muscle Cells/metabolism , Protein Isoforms , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Drug , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Sulfonylurea Receptors , Tissue Distribution , Transfection , Up-Regulation
13.
BMC Physiol ; 5(1): 1, 2005 Jan 12.
Article in English | MEDLINE | ID: mdl-15647111

ABSTRACT

BACKGROUND: Electrophysiological data suggest that cardiac KATP channels consist of Kir6.2 and SUR2A subunits, but the distribution of these (and other KATP channel subunits) is poorly defined. We examined the localization of each of the KATP channel subunits in the mouse and rat heart. RESULTS: Immunohistochemistry of cardiac cryosections demonstrate Kir6.1 protein to be expressed in ventricular myocytes, as well as in the smooth muscle and endothelial cells of coronary resistance vessels. Endothelial capillaries also stained positive for Kir6.1 protein. Kir6.2 protein expression was found predominantly in ventricular myocytes and also in endothelial cells, but not in smooth muscle cells. SUR1 subunits are strongly expressed at the sarcolemmal surface of ventricular myocytes (but not in the coronary vasculature), whereas SUR2 protein was found to be localized predominantly in cardiac myocytes and coronary vessels (mostly in smaller vessels). Immunocytochemistry of isolated ventricular myocytes shows co-localization of Kir6.2 and SUR2 proteins in a striated sarcomeric pattern, suggesting t-tubular expression of these proteins. Both Kir6.1 and SUR1 subunits were found to express strongly at the sarcolemma. The role(s) of these subunits in cardiomyocytes remain to be defined and may require a reassessment of the molecular nature of ventricular KATP channels. CONCLUSIONS: Collectively, our data demonstrate unique cellular and subcellular KATP channel subunit expression patterns in the heart. These results suggest distinct roles for KATP channel subunits in diverse cardiac structures.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Coronary Vessels/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Endothelium, Vascular/metabolism , Heart Ventricles , Immunohistochemistry , In Vitro Techniques , KATP Channels , Mice , Mitochondria, Heart/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rats , Receptors, Drug , Subcellular Fractions/metabolism , Sulfonylurea Receptors , Tissue Distribution
14.
J Mol Cell Cardiol ; 37(4): 857-69, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15380676

ABSTRACT

Functional ATP-sensitive potassium (K(ATP)) channels can be reconstituted by expression of various combinations of different pore-forming subunits (Kir6.1 and Kir6.2) and sulfonylurea receptor (SUR) subunits. Using dominant negative and gene knockout approaches, Kir6.2 subunits have been identified as required pore-forming components of plasmalemmal K(ATP) channels in ventricular myocytes. Previous data obtained in heterologous expression systems suggest that Kir6.1 and Kir6.2 subunits are capable of forming a functional heteromultimeric channel complex. However, until now the existence of such heteromultimeric Kir6.1/Kir6.2 complexes has not been demonstrated for native K(ATP) channels. The primary aim of this study was to identify the molecular composition of native K(ATP) channels in primary human coronary artery endothelial cells (HCAEC) and smooth muscle cells (HCASMC) from human origin. We specifically investigated the potential that heteromultimeric Kir6.1/Kir6.2 channels exist in these cells. Using reverse transcriptase-polymerase chain reaction, we detected the expression of Kir6.1, Kir6.2, and SUR2B in both cell types. Western blotting and immunoprecipitation assays demonstrated the presence of Kir6.1 protein in both HCAEC and HCASMC; however, Kir6.2 protein was only expressed in HCAEC. Interaction between Kir6.1 and Kir6.2 subunits was demonstrated by reciprocal co-immunoprecipitation of these two subunits in HCAEC. Furthermore, Kir6.1 and Kir6.2 were detected in the immunoprecipitate when using an anti-SUR2 antibody. Confocal microscopy imaging demonstrated Kir6.1 and Kir6.2 subunits to co-localize at the cell surface membrane in HCAEC. In conclusion, our data characterize the molecular composition of primary human coronary smooth muscle and endothelial cells. We demonstrate that human coronary endothelial K(ATP) channels consist of a heteromultimeric complex of Kir6.1, Kir6.2, and SUR2B subunits.


Subject(s)
ATP-Binding Cassette Transporters/analysis , Coronary Vessels/cytology , Endothelium, Vascular/chemistry , Potassium Channels, Inwardly Rectifying/analysis , Potassium Channels/analysis , Receptors, Drug/analysis , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cells, Cultured , Coronary Vessels/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Gene Expression , Humans , Immunoprecipitation , KATP Channels , Membrane Potentials , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Subunits/analysis , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Drug/genetics , Receptors, Drug/metabolism , Sulfonylurea Receptors
15.
Circ Res ; 91(3): 232-9, 2002 Aug 09.
Article in English | MEDLINE | ID: mdl-12169649

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) stimulates ATP-sensitive K+ (K(ATP)) channel activity. Because phospholipase C (PLC) hydrolyzes membrane-bound PIP2, which in turn may potentially decrease K(ATP) channel activity, we investigated the effects of the alpha1-adrenoceptor-G(q)-PLC signal transduction axis on pinacidil-activated K(ATP) channel activity in adult rat and neonatal mouse ventricular myocytes. The alpha1-adrenoceptor agonist methoxamine (MTX) reversibly inhibited the pinacidil-activated K(ATP) current in a concentration-dependent manner (IC50 20.9+/-6.6 micromol/L). This inhibition did not occur when the specific alpha1-adrenoceptor antagonist, prazosin, was present. An involvement of G proteins is suggested by the ability of GDPbetaS to prevent this response. Blockade of PLC by U-73122 (2 micromol/L) or neomycin (2 mmol/L) attenuated the MTX-induced inhibition of K(ATP) channel activity. In contrast, the MTX response was unaffected by protein kinase C inhibition or stimulation by H-7 (100 micro mol/L) or phorbol 12,13-didecanoate. The MTX-induced inhibition became irreversible in the presence of wortmannin (20 micro mol/L), an inhibitor of phosphatidylinositol-4 kinase, which is expected to prevent membrane PIP2 replenishment. In excised inside-out patch membranes, pinacidil induced a significantly rightward shift of ATP sensitivity of the channel. This phenomenon was reversed by pretreatment of myocytes with MTX. Direct visualization of PIP2 subcellular distribution using a PLCdelta pleckstrin homology domain-green fluorescent protein fusion constructs revealed reversible translocation of green fluorescent protein fluorescence from the membrane to the cytosol after alpha1-adrenoceptor stimulation. Our data demonstrate that alpha1-adrenoceptor stimulation reduces the membrane PIP2 level, which in turn inhibits pinacidil-activated K(ATP) channels.


Subject(s)
Adenosine Triphosphate/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Pinacidil/antagonists & inhibitors , Potassium Channels/physiology , Receptors, Adrenergic, alpha-1/physiology , Ventricular Function , Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-Agonists/pharmacology , Androstadienes/pharmacology , Animals , Animals, Newborn , Cell Membrane/metabolism , Cells, Cultured , Electric Conductivity , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/physiology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Methoxamine/pharmacology , Mice , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Type C Phospholipases/physiology , Wortmannin
16.
Jpn J Physiol ; 52(1): 31-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12047800

ABSTRACT

Cell swelling enhances a slowly activating delayed rectifier K(+) current (I(Ks)) in cardiac cells. This investigation was undertaken to determine which of the two structural units reconstituting the I(Ks) channel, KCNQ1 (KvLQT1) and KCNE1 (minK/IsK), plays a key role in the cell swelling-induced I(Ks) enhancement and to dissect a possible involvement of tyrosine phosphorylation therein. KCNQ1 was transiently expressed alone or together with KCNE1 in a heterologous mammalian cell line. Two distinct whole-cell membrane currents were separately observed during the exposure of transfected cells to various degrees of hyposmotic solutions. A hyposmotic challenge (0.7 times control osmolarity) resulted in about a twofold increase not only in the heteromeric KCNQ1/KCNE1, but also in the homomeric KCNQ1 channel currents. There was no significant difference in the incremental ratio of current amplitude in response to hyposmotic stress between the two KCNQ1-related currents, and the cells expressing the heteromeric channels swelled less than those with the homomeric channels or without the exogenous ones. The cell swelling-induced I(Ks) enhancement was not affected by a protein tyrosine kinase (PTK) inhibitor, by genistein (50 microM), or by an inhibitor of phosphotyrosine phosphatase (PTP), orthovanadate (500 microM), or a nonhydrolyzable ATP analogue, AMP-PNP (5 mM). Taken together, it is very likely that KCNQ1 might primarily participate in the I(Ks) enhancement by osmotic cell swelling. The obligatory dependence of the I(Ks) augmentation on PTK activity remained to be demonstrated, at least, in this expression system.


Subject(s)
Cell Physiological Phenomena , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , Animals , COS Cells , Electric Conductivity , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Phosphorylation , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...