Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Biol Chem ; 286(1): 707-16, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-20940310

RESUMO

Voltage-dependent potassium (Kv) channels are tetramers of six transmembrane domain (S1-S6) proteins. Crystallographic data demonstrate that the tetrameric pore (S5-S6) is surrounded by four voltage sensor domains (S1-S4). One key question remains: how do voltage sensors (S4) regulate pore gating? Previous mutagenesis data obtained on the Kv channel KCNQ1 highlighted the critical role of specific residues in both the S4-S5 linker (S4S5(L)) and S6 C terminus (S6(T)). From these data, we hypothesized that S4S5(L) behaves like a ligand specifically interacting with S6(T) and stabilizing the closed state. To test this hypothesis, we designed plasmid-encoded peptides corresponding to portions of S4S5(L) and S6(T) of the voltage-gated potassium channel KCNQ1 and evaluated their effects on the channel activity in the presence and absence of the ancillary subunit KCNE1. We showed that S4S5(L) peptides inhibit KCNQ1, in a reversible and state-dependent manner. S4S5(L) peptides also inhibited a voltage-independent KCNQ1 mutant. This inhibition was competitively prevented by a peptide mimicking S6(T), consistent with S4S5(L) binding to S6(T). Val(254) in S4S5(L) is known to contact Leu(353) in S6(T) when the channel is closed, and mutations of these residues alter the coupling between the two regions. The same mutations introduced in peptides altered their effects, further confirming S4S5(L) binding to S6(T). Our results suggest a mechanistic model in which S4S5(L) acts as a voltage-dependent ligand bound to its receptor on S6 at rest. This interaction locks the channel in a closed state. Upon plasma membrane depolarization, S4 pulls S4S5(L) away from S6(T), allowing channel opening.


Assuntos
Condutividade Elétrica , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Ativação do Canal Iônico , Canal de Potássio KCNQ1/genética , Cinética , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese , Mutação , Fragmentos de Peptídeos/metabolismo , Porosidade , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato
2.
J Mol Cell Cardiol ; 49(4): 639-46, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600101

RESUMO

Gender-related differences in ventricular electrophysiology are known to be important determinants of human arrhythmic risk, but the underlying molecular basis is poorly understood. The present work aims to provide the first detailed analysis of gender-related cardiac ion-channel gene-distribution, based on samples from non-diseased human hearts. By using a high-throughput quantitative approach, we investigated at a genome-scale the expression of 79 genes encoding ion-channel and transporter subunits in epicardial and endocardial tissue samples from non-diseased transplant donors (10 males, 10 females). Gender-related expression differences involved key genes implicated in conduction and repolarization. Female hearts showed reduced expression for a variety of K(+)-channel subunits with potentially important roles in cardiac repolarization, including HERG, minK, Kir2.3, Kv1.4, KChIP2, SUR2 and Kir6.2, as well as lower expression of connexin43 and phospholamban. In addition, they demonstrated an isoform switch in Na(+)/K(+)-ATPase, expressing more of the alpha1 and less of the alpha3 subunit than male hearts, along with increased expression of calmodulin-3. Iroquois transcription factors (IRX3, IRX5) were more strongly expressed in female than male epicardium, but the transmural gradient remained. Protein-expression paralleled transcript patterns for all subunits examined: HERG, minK, Kv1.4, KChIP2, IRX5, Nav1.5 and connexin43. Our results indicate that male and female human hearts have significant differences in ion-channel subunit composition, with female hearts showing decreased expression for a number of repolarizing ion-channels. These findings are important for understanding sex-related differences in the susceptibility to ventricular arrhythmias, particularly for conditions associated with repolarization abnormalities like Brugada and Long QT syndrome.


Assuntos
Canais Iônicos/metabolismo , Miocárdio/metabolismo , Adulto , Síndrome de Brugada/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Immunoblotting , Técnicas In Vitro , Síndrome do QT Longo/metabolismo , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Canais de Potássio/genética , Canais de Potássio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
PLoS One ; 5(2): e9298, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20174578

RESUMO

BACKGROUND: Loss-of-function mutations in SCN5A, the gene encoding Na(v)1.5 Na+ channel, are associated with inherited cardiac conduction defects and Brugada syndrome, which both exhibit variable phenotypic penetrance of conduction defects. We investigated the mechanisms of this heterogeneity in a mouse model with heterozygous targeted disruption of Scn5a (Scn5a(+/-) mice) and compared our results to those obtained in patients with loss-of-function mutations in SCN5A. METHODOLOGY/PRINCIPAL FINDINGS: Based on ECG, 10-week-old Scn5a(+/-) mice were divided into 2 subgroups, one displaying severe ventricular conduction defects (QRS interval>18 ms) and one a mild phenotype (QRS< or = 18 ms; QRS in wild-type littermates: 10-18 ms). Phenotypic difference persisted with aging. At 10 weeks, the Na+ channel blocker ajmaline prolonged QRS interval similarly in both groups of Scn5a(+/-) mice. In contrast, in old mice (>53 weeks), ajmaline effect was larger in the severely affected subgroup. These data matched the clinical observations on patients with SCN5A loss-of-function mutations with either severe or mild conduction defects. Ventricular tachycardia developed in 5/10 old severely affected Scn5a(+/-) mice but not in mildly affected ones. Correspondingly, symptomatic SCN5A-mutated Brugada patients had more severe conduction defects than asymptomatic patients. Old severely affected Scn5a(+/-) mice but not mildly affected ones showed extensive cardiac fibrosis. Mildly affected Scn5a(+/-) mice had similar Na(v)1.5 mRNA but higher Na(v)1.5 protein expression, and moderately larger I(Na) current than severely affected Scn5a(+/-) mice. As a consequence, action potential upstroke velocity was more decreased in severely affected Scn5a(+/-) mice than in mildly affected ones. CONCLUSIONS: Scn5a(+/-) mice show similar phenotypic heterogeneity as SCN5A-mutated patients. In Scn5a(+/-) mice, phenotype severity correlates with wild-type Na(v)1.5 protein expression.


Assuntos
Arritmias Cardíacas/fisiopatologia , Síndrome de Brugada/fisiopatologia , Modelos Animais de Doenças , Canais de Sódio/fisiologia , Adolescente , Adulto , Alelos , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Western Blotting , Síndrome de Brugada/genética , Síndrome de Brugada/patologia , Criança , Eletrocardiografia , Feminino , Expressão Gênica , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Penetrância , Canais de Sódio/genética , Canais de Sódio/metabolismo , Adulto Jovem
4.
Circ Res ; 104(11): 1283-92, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19407241

RESUMO

Conduction slowing of the electric impulse that drives the heartbeat may evoke lethal cardiac arrhythmias. Mutations in SCN5A, which encodes the pore-forming cardiac sodium channel alpha subunit, are associated with familial arrhythmia syndromes based on conduction slowing. However, disease severity among mutation carriers is highly variable. We hypothesized that genetic modifiers underlie the variability in conduction slowing and disease severity. With the aim of identifying such modifiers, we studied the Scn5a(1798insD/+) mutation in 2 distinct mouse strains, FVB/N and 129P2. In 129P2 mice, the mutation resulted in more severe conduction slowing particularly in the right ventricle (RV) compared to FVB/N. Pan-genomic mRNA expression profiling in the 2 mouse strains uncovered a drastic reduction in mRNA encoding the sodium channel auxiliary subunit beta4 (Scn4b) in 129P2 mice compared to FVB/N. This corresponded to low to undetectable beta4 protein levels in 129P2 ventricular tissue, whereas abundant beta4 protein was detected in FVB/N. Sodium current measurements in isolated myocytes from the 2 mouse strains indicated that sodium channel activation in myocytes from 129P2 mice occurred at more positive potentials compared to FVB/N. Using computer simulations, this difference in activation kinetics was predicted to explain the observed differences in conduction disease severity between the 2 strains. In conclusion, genetically determined differences in sodium current characteristics on the myocyte level modulate disease severity in cardiac sodium channelopathies. In particular, the sodium channel subunit beta4 (SCN4B) may constitute a potential genetic modifier of conduction and cardiac sodium channel disease.


Assuntos
Canalopatias/genética , Sistema de Condução Cardíaco/fisiopatologia , Animais , Arritmias Cardíacas/fisiopatologia , Canalopatias/fisiopatologia , Elementos de DNA Transponíveis , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Camundongos , Camundongos Endogâmicos , Células Musculares/citologia , Células Musculares/fisiologia , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5 , RNA Mensageiro/genética , Canais de Sódio/deficiência , Canais de Sódio/genética , Canais de Sódio/fisiologia , Subunidade beta-4 do Canal de Sódio Disparado por Voltagem
5.
Eur Heart J ; 30(4): 487-96, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19029124

RESUMO

AIMS: Brugada syndrome is an inherited sudden-death arrhythmia syndrome. Na(+)-current dysfunction is central, but mutations in the SCN5A gene (encoding the cardiac Na(+)-channel Nav1.5) are present in only 20% of probands. This study addressed the possibility that Brugada patients display specific expression patterns for ion-channels regulating cardiac conduction, excitability, and repolarization. METHODS AND RESULTS: Transcriptional profiling was performed on right-ventricular endomyocardial biopsies from 10 unrelated Brugada probands, 11 non-diseased organ-donors, seven heart-transplant recipients, 10 with arrhythmogenic right-ventricular cardiomyopathy, and nine with idiopathic right-ventricular outflow-tract tachycardia. Brugada patients showed distinct clustering differences vs. the two control and two other ventricular-tachyarrhythmia groups, including 14 of 77 genes encoding important ion-channel/ion-transporter subunits. Nav1.5 and K(+)-channels Kv4.3 and Kir3.4 were more weakly expressed, whereas the Na(+)-channel Nav2.1 and the K(+)-channel TWIK1 were more strongly expressed, in Brugada syndrome. Differences were also seen in Ca(2+)-homeostasis transcripts, including stronger expression of RYR2 and NCX1. The molecular profile of Brugada patients with SCN5A mutations did not differ from Brugada patients without SCN5A mutations. CONCLUSION: Brugada patients exhibit a common ion-channel molecular expression signature, irrespective of the culprit gene. This finding has potentially important implications for our understanding of the pathophysiology of Brugada syndrome, with possible therapeutic and diagnostic consequences.


Assuntos
Displasia Arritmogênica Ventricular Direita/genética , Síndrome de Brugada/genética , Canais Iônicos/genética , Taquicardia Ventricular/genética , Adulto , Displasia Arritmogênica Ventricular Direita/fisiopatologia , Síndrome de Brugada/diagnóstico , Síndrome de Brugada/fisiopatologia , Feminino , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Genótipo , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/genética , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.5 , Fenótipo , Canais de Sódio/genética , Taquicardia Ventricular/fisiopatologia , Transcrição Gênica/genética , Adulto Jovem
6.
Mol Ther ; 16(12): 1937-43, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18813278

RESUMO

We hypothesized that a nonviral gene delivery of the hyperpolarization-activated HCN2 channel combined with the beta(2)-adrenergic receptor (ADRB2) would generate a functional pacemaker in a mouse model of complete atrioventricular block (CAVB) induced by radiofrequency ablation of the His bundle. Plasmids encoding HCN2 and ADRB2 mixed with tetronic 304, a poloxamine block copolymer, were injected in the left ventricular free wall (HCN2-ADRB2 mice). Sham mice received a noncoding plasmid. CAVB was induced 5 days later. Ventricular escape rhythms in HCN2-ADRB2 mice were significantly faster than in sham mice at day 15 after ablation and later. In HCN2-ADRB2 mice, QRS complexes were larger than in sham mice and characterized by abnormal axes. Immunostaining of GFP-HCN2 fusion protein showed an expression of HCN2 channel in left ventricular myocardium for at least 45 days after injection. In the mouse, CAVB induces progressive hypertrophy and heart failure leading to 50% mortality after 110 days. HCN2-ADRB2 mice survived 3 weeks longer than sham mice. Finally, beta-adrenergic input increased ventricular escape rhythms significantly more in HCN2-ADRB2 mice than in sham mice. In conclusion, nonviral gene transfer can produce a functional cardiac biological pacemaker regulated by sympathetic input, which improves life expectancy in a mouse model of CAVB.


Assuntos
Bloqueio Atrioventricular/genética , Bloqueio Atrioventricular/terapia , Relógios Biológicos , Terapia Genética , Transgenes/genética , Animais , Bloqueio Atrioventricular/metabolismo , Bloqueio Atrioventricular/patologia , Doença Crônica , Modelos Animais de Doenças , Eletrocardiografia , Seguimentos , Vetores Genéticos/genética , Masculino , Camundongos , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Taxa de Sobrevida
7.
J Clin Invest ; 118(6): 2260-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18464934

RESUMO

Brugada syndrome is a genetic disease associated with sudden cardiac death that is characterized by ventricular fibrillation and right precordial ST segment elevation on ECG. Loss-of-function mutations in SCN5A, which encodes the predominant cardiac sodium channel alpha subunit NaV1.5, can cause Brugada syndrome and cardiac conduction disease. However, SCN5A mutations are not detected in the majority of patients with these syndromes, suggesting that other genes can cause or modify presentation of these disorders. Here, we investigated SCN1B, which encodes the function-modifying sodium channel beta1 subunit, in 282 probands with Brugada syndrome and in 44 patients with conduction disease, none of whom had SCN5A mutations. We identified 3 mutations segregating with arrhythmia in 3 kindreds. Two of these mutations were located in a newly described alternately processed transcript, beta1B. Both the canonical and alternately processed transcripts were expressed in the human heart and were expressed to a greater degree in Purkinje fibers than in heart muscle, consistent with the clinical presentation of conduction disease. Sodium current was lower when NaV1.5 was coexpressed with mutant beta1 or beta1B subunits than when it was coexpressed with WT subunits. These findings implicate SCN1B as a disease gene for human arrhythmia susceptibility.


Assuntos
Síndrome de Brugada/genética , Canais de Sódio/genética , Adolescente , Adulto , Síndrome de Brugada/patologia , Eletrofisiologia , Feminino , Predisposição Genética para Doença , Cardiopatias/genética , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Mutação , Miocárdio/metabolismo , Ramos Subendocárdicos/metabolismo , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem
8.
Cardiovasc Res ; 79(3): 427-35, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18390900

RESUMO

AIMS: KCNQ1 (alias KvLQT1 or Kv7.1) and KCNE1 (alias IsK or minK) co-assemble to form the voltage-activated K(+) channel responsible for I(Ks)-a major repolarizing current in the human heart-and their dysfunction promotes cardiac arrhythmias. The channel is a component of larger macromolecular complexes containing known and undefined regulatory proteins. Thus, identification of proteins that modulate its biosynthesis, localization, activity, and/or degradation is of great interest from both a physiological and pathological point of view. METHODS AND RESULTS: Using a yeast two-hybrid screening, we detected a direct interaction between beta-tubulin and the KCNQ1 N-terminus. The interaction was confirmed by co-immunoprecipitation of beta-tubulin and KCNQ1 in transfected COS-7 cells and in guinea pig cardiomyocytes. Using immunocytochemistry, we also found that they co-localized in cardiomyocytes. We tested the effects of microtubule-disrupting and -stabilizing agents (colchicine and taxol, respectively) on the KCNQ1-KCNE1 channel activity in COS-7 cells by means of the permeabilized-patch configuration of the patch-clamp technique. None of these agents altered I(Ks). In addition, colchicine did not modify the current response to osmotic challenge. On the other hand, the I(Ks) response to protein kinase A (PKA)-mediated stimulation depended on microtubule polymerization in COS-7 cells and in cardiomyocytes. Strikingly, KCNQ1 channel and Yotiao phosphorylation by PKA-detected by phospho-specific antibodies-was maintained, as was the association of the two partners. CONCLUSION: We propose that the KCNQ1-KCNE1 channel directly interacts with microtubules and that this interaction plays a major role in coupling PKA-dependent phosphorylation of KCNQ1 with I(Ks) activation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Canal de Potássio KCNQ1/metabolismo , Microtúbulos/metabolismo , Miócitos Cardíacos/enzimologia , Tubulina (Proteína)/metabolismo , Proteínas de Ancoragem à Quinase A/metabolismo , Potenciais de Ação , Animais , Células COS , Chlorocebus aethiops , Cobaias , Canal de Potássio KCNQ1/genética , Cinética , Masculino , Camundongos , Microtúbulos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Pressão Osmótica , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Transfecção , Tubulina (Proteína)/genética , Moduladores de Tubulina/farmacologia
9.
J Physiol ; 582(Pt 2): 675-93, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17478540

RESUMO

The various cardiac regions have specific action potential properties appropriate to their electrical specialization, resulting from a specific pattern of ion-channel functional expression. The present study addressed regionally defined differential ion-channel expression in the non-diseased human heart with a genomic approach. High-throughput real-time RT-PCR was used to quantify the expression patterns of 79 ion-channel subunit transcripts and related genes in atria, ventricular epicardium and endocardium, and Purkinje fibres isolated from 15 non-diseased human donor hearts. Two-way non-directed hierarchical clustering separated atria, Purkinje fibre and ventricular compartments, but did not show specific patterns for epicardium versus endocardium, nor left- versus right-sided chambers. Genes that characterized the atria (versus ventricles) included Cx40, Kv1.5 and Kir3.1 as expected, but also Cav1.3, Cav3.1, Cav alpha2 delta2, Nav beta1, TWIK1, TASK1 and HCN4. Only Kir2.1, RyR2, phospholamban and Kv1.4 showed higher expression in the ventricles. The Purkinje fibre expression-portrait (versus ventricle) included stronger expression of Cx40, Kv4.3, Kir3.1, TWIK1, HCN4, ClC6 and CALM1, along with weaker expression of mRNA encoding Cx43, Kir2.1, KChIP2, the pumps/exchangers Na(+),K(+)-ATPase, NCX1, SERCA2, and the Ca(2+)-handling proteins RYR2 and CASQ2. Transcripts that were more strongly expressed in epicardium (versus endocardium) included Cav1.2, KChIP2, SERCA2, CALM3 and calcineurin-alpha. Nav1.5 and Nav beta1 were more strongly expressed in the endocardium. For selected genes, RT-PCR data were confirmed at the protein level. This is the first report of the global portrait of regional ion-channel subunit-gene expression in the non-diseased human heart. Our data point to significant regionally determined ion-channel expression differences, with potentially important implications for understanding regional electrophysiology, arrhythmia mechanisms, and responses to ion-channel blocking drugs. Concordance with previous functional studies suggests that regional regulation of cardiac ion-current expression may be primarily transcriptional.


Assuntos
Canais Iônicos/genética , Miocárdio/metabolismo , Transcrição Gênica , Adulto , Western Blotting , Proteínas de Transporte/genética , Análise por Conglomerados , Endocárdio/metabolismo , Feminino , Átrios do Coração , Ventrículos do Coração , Humanos , Canais Iônicos/metabolismo , Masculino , Pessoa de Meia-Idade , Pericárdio/metabolismo , Isoformas de Proteínas/genética , Ramos Subendocárdicos/metabolismo , RNA Mensageiro/metabolismo , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Distribuição Tecidual
10.
Circulation ; 115(4): 432-41, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17242276

RESUMO

BACKGROUND: Mutations in the ankyrin-B gene (ANK2) cause type 4 long-QT syndrome and have been described in kindreds with other arrhythmias. The frequency of ANK2 variants in large populations and molecular mechanisms underlying the variability in the clinical phenotypes are not established. More importantly, there is no cellular explanation for the range of severity of cardiac phenotypes associated with specific ANK2 variants. METHODS AND RESULTS: We performed a comprehensive screen of ANK2 in populations (control, congenital arrhythmia, drug-induced long-QT syndrome) of different ethnicities to discover unidentified ANK2 variants. We identified 7 novel nonsynonymous ANK2 variants; 4 displayed abnormal activity in cardiomyocytes. Including the 4 new variants, 9 human ANK2 loss-of-function variants have been identified. However, the clinical phenotypes associated with these variants vary strikingly, from no obvious phenotype to manifest long-QT syndrome and sudden death, suggesting that mutants confer a spectrum of cellular phenotypes. We then characterized the relative severity of loss-of-function properties of all 9 nonsynonymous ANK2 variants identified to date in primary cardiomyocytes and identified a range of in vitro phenotypes, including wild-type, simple loss-of-function, and severe loss-of-function activity, seen with the variants causing severe human phenotypes. CONCLUSIONS: We present the first description of differences in cellular phenotypes conferred by specific ANK2 variants. We propose that the various degrees of ankyrin-B loss of function contribute to the range of severity of cardiac dysfunction. These data identify ANK2 variants as modulators of human arrhythmias, provide the first insight into the clinical spectrum of "ankyrin-B syndrome," and reinforce the role of ankyrin-B-dependent protein interactions in regulating cardiac electrogenesis.


Assuntos
Anquirinas/genética , Arritmias Cardíacas/etnologia , Arritmias Cardíacas/genética , Variação Genética , Miócitos Cardíacos/fisiologia , Idoso , Animais , Arritmias Cardíacas/induzido quimicamente , Povo Asiático/genética , População Negra/genética , Citoesqueleto/fisiologia , Feminino , Genótipo , Humanos , Canais Iônicos/fisiologia , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/etnologia , Síndrome do QT Longo/genética , Masculino , Americanos Mexicanos/genética , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miócitos Cardíacos/citologia , Fenótipo , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/etnologia , Taquicardia Ventricular/genética , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/etnologia , Torsades de Pointes/genética , População Branca/genética
11.
Circ Res ; 99(10): 1076-83, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17053194

RESUMO

N-terminal mutations in the KCNQ1 channel are frequently linked to fatal arrhythmias in newborn children and adolescents but the cellular mechanisms involved in this dramatic issue remain, however, to be discovered. Here, we analyzed the trafficking of a series of N-terminal truncation mutants and identified a critical trafficking motif of KCNQ1. This determinant is located in the juxtamembranous region preceding the first transmembrane domain of the protein. Three mutations (Y111C, L114P and P117L) implicated in inherited Romano-Ward LQT1 syndrome, are embedded within this domain. Reexpression studies in both COS-7 cells and cardiomyocytes showed that the mutant proteins fail to exit the endoplasmic reticulum. KCNQ1 subunits harboring Y111C or L114P exert a dominant negative effect on the wild-type KCNQ1 subunit by preventing plasma membrane trafficking of heteromultimeric channels. The P117L mutation had a less pronounced effect on the trafficking of heteromultimeric channels but altered the kinetics of the current. Furthermore, we showed that the trafficking determinant in KCNQ1 is structurally and functionally conserved in other KCNQ channels and constitutes a critical trafficking determinant of the KCNQ channel family. Computed structural predictions correlated the potential structural changes introduced by the mutations with impaired protein trafficking. In conclusion, our studies unveiled a new role of the N-terminus of KCNQ channels in their trafficking and its implication in severe forms of LQT1 syndrome.


Assuntos
Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Membrana Celular/metabolismo , Criança , Chlorocebus aethiops , Cricetinae , Retículo Endoplasmático/metabolismo , Feminino , Hemaglutininas/genética , Hemaglutininas/metabolismo , Humanos , Canal de Potássio KCNQ1/biossíntese , Camundongos , Dados de Sequência Molecular , Mutagênese , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Isoformas de Proteínas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Transfecção
12.
Methods Mol Biol ; 337: 167-83, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16929947

RESUMO

KCNQ1 is a voltage-activated potassium channel alpha-subunit expressed in various cell types, including cardiac myocytes and epithelial cells. KCNQ1 associates with different beta-subunits of the KCNE protein family. In the human heart, KCNQ1 associates with KCNE1 to generate the IKs current characterized by its slow activation and deactivation kinetics. Mutations in either KCNQ1 or KCNE1 are responsible for at least four channelopathies that lead to cardiac dysfunction and one that leads to congenital deafness: the Romano-Ward syndrome, the short QT syndrome, atrial fibrillation, and the Jervell and Lange-Nielsen syndrome (cardioauditory syndrome). To date, nearly 100 different KCNQ1 mutations have been reported as responsible for the cardiac long QT syndrome, characterized by prolonged QT interval, syncopes, and sudden death. Patch clamp and immunofluorescence techniques are instrumental for characterization of the molecular mechanisms responsible for the altered function of KCNQ1 and its partners.


Assuntos
Doenças Cardiovasculares/metabolismo , Canal de Potássio KCNQ1/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Genes Dominantes/genética , Humanos , Mutação/genética , Técnicas de Patch-Clamp , Processamento de Proteína Pós-Traducional
13.
Circ Res ; 98(11): 1422-30, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16690884

RESUMO

The generation of the mammalian heartbeat is a complex and vital function requiring multiple and coordinated ionic channel activities. The functional role of low-voltage activated (LVA) T-type calcium channels in the pacemaker activity of the sinoatrial node (SAN) is, to date, unresolved. Here we show that disruption of the gene coding for CaV3.1/alpha1G T-type calcium channels (cacna1g) abolishes T-type calcium current (I(Ca,T)) in isolated cells from the SAN and the atrioventricular node without affecting the L-type Ca2+ current (I(Ca,L)). By using telemetric electrocardiograms on unrestrained mice and intracardiac recordings, we find that cacna1g inactivation causes bradycardia and delays atrioventricular conduction without affecting the excitability of the right atrium. Consistently, no I(Ca,T) was detected in right atrium myocytes in both wild-type and CaV3.1(-/-) mice. Furthermore, inactivation of cacna1g significantly slowed the intrinsic in vivo heart rate, prolonged the SAN recovery time, and slowed pacemaker activity of individual SAN cells through a reduction of the slope of the diastolic depolarization. Our results demonstrate that CaV3.1/T-type Ca2+ channels contribute to SAN pacemaker activity and atrioventricular conduction.


Assuntos
Nó Atrioventricular/fisiopatologia , Bradicardia/etiologia , Bradicardia/fisiopatologia , Canais de Cálcio Tipo T/deficiência , Animais , Nó Atrioventricular/metabolismo , Nó Atrioventricular/patologia , Bradicardia/metabolismo , Bradicardia/patologia , Condutividade Elétrica , Eletrocardiografia , Eletrofisiologia , Frequência Cardíaca , Hipnóticos e Sedativos/farmacologia , Camundongos , Camundongos Knockout , Isoformas de Proteínas/deficiência , Nó Sinoatrial/fisiopatologia
14.
Circ Res ; 98(12): 1538-46, 2006 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-16728661

RESUMO

The voltage-sensitive Na(+) channel Na(v)1.5 plays a crucial role in generating and propagating the cardiac action potential and its dysfunction promotes cardiac arrhythmias. The channel takes part into a large molecular complex containing regulatory proteins. Thus, factors that modulate its biosynthesis, localization, activity, and/or degradation are of great interest from both a physiological and pathological standpoint. Using a yeast 2-hybrid screen, we unveiled a novel partner, 14-3-3eta, interacting with the Na(v)1.5 cytoplasmic I interdomain. The interaction was confirmed by coimmunoprecipitation of 14-3-3 and full-length Na(v)1.5 both in COS-7 cells expressing recombinant Na(v)1.5 and in mouse cardiac myocytes. Using immunocytochemistry, we also found that 14-3-3 and Na(v)1.5 colocalized at the intercalated discs. We tested the functional link between Na(v)1.5 and 14-3-3eta using the whole-cell patch-clamp configuration. Coexpressing Na(v)1.5, the beta1 subunit and 14-3-3eta induced a negative shift in the inactivation curve of the Na(+) current, a delayed recovery from inactivation, but no changes in the activation curve or in the current density. The negative shift was reversed, and the recovery from inactivation was normalized by overexpressing the Na(v)1.5 cytoplasmic I interdomain interacting with 14-3-3eta. Reversal was also obtained with the dominant negative R56,60A 14-3-3eta mutant, suggesting that dimerization of 14-3-3 is needed for current regulation. Computer simulations suggest that the absence of 14-3-3 could exert proarrhythmic effects on cardiac electrical restitution properties. Based on these findings, we propose that the 14-3-3 protein is a novel component of the cardiac Na(+) channel acting as a cofactor for the regulation of the cardiac Na(+) current.


Assuntos
Proteínas 14-3-3/fisiologia , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Canais de Sódio/metabolismo , Proteínas 14-3-3/química , Potenciais de Ação/fisiologia , Animais , Células COS , Chlorocebus aethiops , Simulação por Computador , Dimerização , Condutividade Elétrica , Eletrofisiologia , Coração/fisiologia , Humanos , Membranas Intracelulares/metabolismo , Modelos Cardiovasculares , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Isoformas de Proteínas/fisiologia , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Canais de Sódio/genética , Canais de Sódio/fisiologia , Transfecção
15.
J Cardiovasc Electrophysiol ; 17(3): 270-5, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16643399

RESUMO

INTRODUCTION: Loss-of-function mutations in the SCN5A gene encoding the cardiac sodium channel are responsible for Brugada syndrome (BS) and also for progressive cardiac conduction disease (inherited Lenègre disease). In an attempt to clarify the frontier between these two entities, we have characterized cardiac conduction defect and its evolution with aging in a cohort of 78 patients carrying a SCN5A mutation linked to Brugada syndrome. METHODS AND RESULTS: Families were included in the study if a SCN5A mutation was identified in a BS proband and if at least two family members were mutation carriers. Sixteen families met the study criteria, representing 78 carriers. Resting ECG showed a spontaneous BS ECG pattern in 28 of 78 (36%) gene carriers. Intraventricular conduction anomalies were identified in 59 of 78 gene carriers including complete (17) or incomplete (24) right bundle branch block, right bundle branch block plus hemiblock (6), left bundle branch block (1), hemiblock (1), and parietal block (10). PR and QRS duration were longer in the gene carrier cohort in comparison with their relatives carrying no mutation. Finally, in the gene carrier cohort conduction defect progressively aggravated with aging leading in five occasions to pacemaker implantations. CONCLUSION: The present study shows that the most common phenotype of gene carriers of a BS-type SCN5A mutation is progressive cardiac conduction defects similar to the Lenègre disease phenotype. In consequence, we propose that carriers of a SCN5A mutation need a clinical and ECG follow-up because of the risk associated with severe conduction defects.


Assuntos
Bloqueio Cardíaco/genética , Bloqueio Cardíaco/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Canais de Sódio/genética , Adolescente , Adulto , Progressão da Doença , Eletrocardiografia , Saúde da Família , Feminino , França , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Síndrome
16.
J Mol Cell Cardiol ; 40(1): 173-84, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16242148

RESUMO

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in humans. The pathophysiology of AF involves electrical, structural and contractile remodeling, which is associated with changes in cardiac gene expression. Previous studies of gene-expression changes in clinical AF have mostly been limited to a small number of candidate genes and have not all been well controlled for underlying heart disease. The present study assessed AF-related gene-expression changes in valve-disease patients with microarrays representing the cardiac transcriptome. Right atrial appendages from 11 patients with chronic AF and underlying valvular heart disease (AF-VHD) and seven patients in sinus rhythm with VHD (SR-VHD) were individually compared to an age-matched sinus-rhythm control group (SR-CTRL, 11 patients) using cardiac-specific microarray analysis. One-class statistical analysis was used to identify genes differentially expressed between SR-VHD and SR-CTRL patients. Two-class statistical analysis was used to identify genes differentially expressed between AF-VHD and SR-VHD patients. Out of 3863 analyzed genes, 832 genes were differentially expressed between SR-VHD and SR-CTRL patients, and 169 genes were differentially expressed between AF-VHD and SR-VHD patients. Striking AF-related changes included altered expression of nine genes pointing towards the development of fibrosis (e.g. upregulation of transforming growth factor beta1), and changes in eight genes potentially related to an increased risk of thromboembolic events (e.g. upregulation of alpha2 macroglobulin). Microarray results were confirmed by quantitative PCR. Our results suggest that AF produces a characteristic profile of gene-expression changes that may be related to the pathophysiology of the arrhythmia.


Assuntos
Fibrilação Atrial/genética , Doenças das Valvas Cardíacas/genética , Análise de Sequência com Séries de Oligonucleotídeos , Idoso , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Reação em Cadeia da Polimerase , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1 , alfa-Macroglobulinas/genética
17.
Physiol Genomics ; 24(1): 4-12, 2005 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-16219869

RESUMO

We investigated the effects of chronic and moderate heart rate (HR) reduction on ion channel expression in the mouse sinoatrial node (SAN) and ventricle. Ten-week-old male C57BL/6 mice were treated twice daily with either vehicle or ivabradine at 5 mg/kg given orally during 3 wk. The effects of HR reduction on cardiac electrical activity were investigated in anesthetized mice with serial ECGs and in freely moving mice with telemetric recordings. With the use of high-throughput real-time RT-PCR, the expression of 68 ion channel subunits was evaluated in the SAN and ventricle at the end of the treatment period. In conscious mice, ivabradine induced a mean 16% HR reduction over a 24-h period that was sustained over the 3-wk administration. Other ECG parameters were not modified. Two-way hierarchical clustering analysis of gene expression revealed a separation of ventricles from SANs but no discrimination between treated and untreated ventricles, indicating that HR reduction per se induced limited remodeling in this tissue. In contrast, SAN samples clustered in two groups depending on the treatment. In the SAN from ivabradine-treated mice, the expression of nine ion channel subunits, including Navbeta1 (-25%), Cav3.1 (-29%), Kir6.1 (-28%), Kvbeta2 (-41%), and Kvbeta3 (-30%), was significantly decreased. Eight genes were significantly upregulated, including K+ channel alpha-subunits (Kv1.1, +30%; Kir2.1, +29%; Kir3.1, +41%), hyperpolarization-activated cation channels (HCN2, +24%; HCN4, +52%), and connexin 43 (+26%). We conclude that reducing HR induces a complex remodeling of ion channel expression in the SAN but has little impact on ion channel transcripts in the ventricle.


Assuntos
Benzazepinas/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Canais Iônicos/genética , Nó Sinoatrial/fisiologia , Transcrição Gênica/efeitos dos fármacos , Função Ventricular , Animais , Benzazepinas/sangue , Eletrocardiografia/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ivabradina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nó Sinoatrial/efeitos dos fármacos
18.
Circulation ; 112(13): 1927-35, 2005 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-16172272

RESUMO

BACKGROUND: The SCN5A sodium channel is a major determinant for cardiac impulse propagation. We used epicardial mapping of the atria, ventricles, and septae to investigate conduction velocity (CV) in Scn5a heterozygous young and old mice. METHODS AND RESULTS: Mice were divided into 4 groups: (1) young (3 to 4 months) wild-type littermates (WT); (2) young heterozygous Scn5a-knockout mice (HZ); (3) old (12 to 17 months) WT; and (4) old HZ. In young HZ hearts, CV in the right but not the left ventricle was reduced in agreement with a rightward rotation in the QRS axes; fibrosis was virtually absent in both ventricles, and the pattern of connexin43 (Cx43) expression was similar to that of WT mice. In old WT animals, the right ventricle transversal CV was slightly reduced and was associated with interstitial fibrosis. In old HZ hearts, right and left ventricle CVs were severely reduced both in the transversal and longitudinal direction; multiple areas of severe reactive fibrosis invaded the myocardium, accompanied by markedly altered Cx43 expression. The right and left bundle-branch CVs were comparable to those of WT animals. The atria showed only mild fibrosis, with heterogeneously disturbed Cx40 and Cx43 expression. CONCLUSIONS: A 50% reduction in Scn5a expression alone or age-related interstitial fibrosis only slightly affects conduction. In aged HZ mice, reduced Scn5a expression is accompanied by the presence of reactive fibrosis and disarrangement of gap junctions, which results in profound conduction impairment.


Assuntos
Envelhecimento , Conexinas/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Canais de Sódio/deficiência , Animais , Fascículo Atrioventricular/metabolismo , Conexina 43/metabolismo , Eletrocardiografia , Fibrose , Átrios do Coração , Ventrículos do Coração , Heterozigoto , Técnicas In Vitro , Camundongos , Camundongos Knockout , Fatores de Tempo , Função Ventricular , Canais de Sódio Disparados por Voltagem , Proteína alfa-5 de Junções Comunicantes
19.
Circulation ; 112(4): 471-81, 2005 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-16027256

RESUMO

BACKGROUND: Valvular heart disease (VHD), which often leads to atrial fibrillation (AF), and AF both cause ion-channel remodeling. We evaluated the ion-channel gene expression profile of VHD patients, in permanent AF (AF-VHD) or in sinus rhythm (SR-VHD), in comparison with patients without AF or VHD, respectively. METHODS AND RESULTS: We used microarrays containing probes for human ion-channel and Ca2+-regulator genes to quantify mRNA expression in atrial tissues from 7 SR-VHD patients and 11 AF-VHD patients relative to 11 control patients in SR without structural heart disease (SR-CAD). From the data set, we selected for detailed analysis 59 transcripts expressed in the human heart. SR-VHD patients differentially expressed 24/59 ion-channel and Ca2+-regulator transcripts. There was significant overlap between VHD groups, with 66% of genes altered in SR-VHD patients being similarly modified in AF-VHD. Statistical differences between the AF- and SR-VHD groups identified the specific molecular portrait of AF, which involved 12 genes that were further confirmed by real-time reverse transcription-polymerase chain reaction. For example, phospholamban, the beta-subunit MinK (KCNE1) and MIRP2 (KCNE3), and the 2-pore potassium channel TWIK-1 were upregulated in AF-VHD compared with SR-VHD, whereas the T-type calcium-channel Cav3.1 and the transient-outward potassium channel Kv4.3 were downregulated. Two-way hierarchical clustering separated SR-VHD from AF-VHD patients. AF-related changes in L-type Ca2+-current and inward-rectifier current were confirmed at protein and functional levels. Finally, for 13 selected genes, SR restoration reversed ion-channel remodeling. CONCLUSIONS: VHD extensively remodels cardiac ion-channel and transporter expression, and AF alters ion-channel expression in VHD patients.


Assuntos
Fibrilação Atrial/metabolismo , Átrios do Coração/metabolismo , Doenças das Valvas Cardíacas/metabolismo , Canais Iônicos/genética , Western Blotting , Canais de Cálcio Tipo L/genética , Conexinas/genética , Perfilação da Expressão Gênica , Humanos , Proteínas de Membrana Transportadoras/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Subunidades Proteicas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Potássio Shal/genética , Proteína alfa-5 de Junções Comunicantes
20.
Cardiovasc Res ; 67(3): 529-38, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16039274

RESUMO

OBJECTIVE: The voltage-gated K+ channel KCNQ1 associates with the small KCNE1 beta subunit to underlie the IKs repolarizing current in the heart. Based on sequence homology, the KCNE family is recognized to comprise five members. Controversial data have indicated their participation in several K+ channel protein complexes, including KCNQ1. The expression level and the putative functions of the different KCNE subunits in the human heart still require further investigation. METHODS: We have carried out a comparative study of all KCNE subunits with KCNQ1 using the patch-clamp technique in mammalian cells. Real-time RT-PCR absolute quantification was performed on human atrial and ventricular tissue. RESULTS: While KCNQ1/KCNE1 heteromultimer reached high current density with slow gating kinetics and pronounced voltage dependence, KCNQ1/KCNE2 and KCNQ1/KCNE3 complexes produced instantaneous voltage-independent currents with low and high current density, respectively. Co-expression of KCNE4 or KCNE5 with KCNQ1 induced small currents in the physiological range of voltages, with kinetics similar to those of the KCNQ1/KCNE1 complex. However, co-expression of these inhibitory subunits with a disease-associated mutation (S140G-KCNQ1) led to currents that were almost undistinguishable from the KCNQ1/KCNE1 canonical complex. Absolute cDNA quantification revealed a relatively homogeneous distribution of each transcript, except for KCNE4, inside left atria and endo- and epicardia of left ventricular wall with the following abundance: KCNQ1 >> KCNE4 > or = KCNE1 > KCNE3 > KCNE2 > KCNE5. KCNE4 expression was twice as high in atrium compared to ventricle. CONCLUSIONS: Our data show that KCNQ1 forms a channel complex with 5 KCNE subunits in a specific manner but only interactions with KCNE1, KCNE2, and KCNE3 may have physiological relevance in the human heart.


Assuntos
Canal de Potássio KCNQ1/genética , Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , RNA Mensageiro/análise , Animais , Células COS , Chlorocebus aethiops , Cricetinae , Átrios do Coração , Humanos , Ativação do Canal Iônico , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA