Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 133(3): 462-74, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18455987

RESUMO

Calcium/calmodulin (Ca2+/CaM)-dependent protein kinase II (CaMKII) couples increases in cellular Ca2+ to fundamental responses in excitable cells. CaMKII was identified over 20 years ago by activation dependence on Ca2+/CaM, but recent evidence shows that CaMKII activity is also enhanced by pro-oxidant conditions. Here we show that oxidation of paired regulatory domain methionine residues sustains CaMKII activity in the absence of Ca2+/CaM. CaMKII is activated by angiotensin II (AngII)-induced oxidation, leading to apoptosis in cardiomyocytes both in vitro and in vivo. CaMKII oxidation is reversed by methionine sulfoxide reductase A (MsrA), and MsrA-/- mice show exaggerated CaMKII oxidation and myocardial apoptosis, impaired cardiac function, and increased mortality after myocardial infarction. Our data demonstrate a dynamic mechanism for CaMKII activation by oxidation and highlight the critical importance of oxidation-dependent CaMKII activation to AngII and ischemic myocardial apoptosis.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cardiopatias/metabolismo , Metionina/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Angiotensina II , Animais , Apoptose , Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Calmodulina/metabolismo , Metionina Sulfóxido Redutases , Camundongos , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Oxirredução , Oxirredutases/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo
2.
Circ Res ; 109(1): 38-46, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21566215

RESUMO

RATIONALE: Although multiple lines of evidence suggest that variable expression of the cardiac sodium channel gene SCN5A plays a role in susceptibility to arrhythmia, little is known about its transcriptional regulation. OBJECTIVE: We used in silico and in vitro experiments to identify possible noncoding sequences important for transcriptional regulation of SCN5A. The results were extended to mice in which a putative regulatory region was deleted. METHODS AND RESULTS: We identified 92 noncoding regions highly conserved (>70%) between human and mouse SCN5A orthologs. Three conserved noncoding sequences (CNS) showed significant (>5-fold) activity in luciferase assays. Further in vitro studies indicated one, CNS28 in intron 1, as a potential regulatory region. Using recombinase-mediated cassette exchange (RMCE), we generated mice in which a 435-base pair region encompassing CNS28 was removed. Animals homozygous for the deletion showed significant increases in SCN5A transcripts, Na(V)1.5 protein abundance, and sodium current measured in isolated ventricular myocytes. ECGs revealed a significantly shorter QRS (10.7±0.2 ms in controls versus 9.7±0.2 ms in knockouts), indicating more rapid ventricular conduction. In vitro analysis of CNS28 identified a short 3' segment within this region required for regulatory activity and including an E-box motif. Deletion of this segment reduced reporter activity to 3.6%±0.3% of baseline in CHO cells and 16%±3% in myocytes (both P<0.05), and mutation of individual sites in the E-box restored activity to 62%±4% and 57%±2% of baseline in CHO cells and myocytes, respectively (both P<0.05). CONCLUSIONS: These findings establish that regulation of cardiac sodium channel expression modulates channel function in vivo, and identify a noncoding region underlying this regulation.


Assuntos
Sequências Reguladoras de Ácido Nucleico , Canais de Sódio/genética , Animais , Sequência de Bases , Células CHO , Sequência Conservada , Cricetinae , Cricetulus , Eletrocardiografia , Regulação da Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.5 , Regiões Promotoras Genéticas , Canais de Sódio/fisiologia , Transcrição Gênica
3.
Circulation ; 124(9): 1001-11, 2011 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-21824921

RESUMO

BACKGROUND: The D1275N SCN5A mutation has been associated with a range of unusual phenotypes, including conduction disease and dilated cardiomyopathy, as well as atrial and ventricular tachyarrhythmias. However, when D1275N is studied in heterologous expression systems, most studies show near-normal sodium channel function. Thus, the relationship of the variant to the clinical phenotypes remains uncertain. METHODS AND RESULTS: We identified D1275N in a patient with atrial flutter, atrial standstill, conduction disease, and sinus node dysfunction. There was no major difference in biophysical properties between wild-type and D1275N channels expressed in Chinese hamster ovary cells or tsA201 cells in the absence or presence of ß1 subunits. To determine D1275N function in vivo, the Scn5a locus was modified to knock out the mouse gene, and the full-length wild-type (H) or D1275N (DN) human SCN5A cDNAs were then inserted at the modified locus by recombinase mediated cassette exchange. Mice carrying the DN allele displayed slow conduction, heart block, atrial fibrillation, ventricular tachycardia, and a dilated cardiomyopathy phenotype, with no significant fibrosis or myocyte disarray on histological examination. The DN allele conferred gene-dose-dependent increases in SCN5A mRNA abundance but reduced sodium channel protein abundance and peak sodium current amplitudes (H/H, 41.0±2.9 pA/pF at -30 mV; DN/H, 19.2±3.1 pA/pF, P<0.001 vs. H/H; DN/DN, 9.3±1.1 pA/pF, P<0.001 versus H/H). CONCLUSIONS: Although D1275N produces near-normal currents in multiple heterologous expression experiments, our data establish this variant as a pathological mutation that generates conduction slowing, arrhythmias, and a dilated cardiomyopathy phenotype by reducing cardiac sodium current.


Assuntos
Flutter Atrial/genética , Cardiomiopatia Dilatada/genética , Mutação de Sentido Incorreto , Síndrome do Nó Sinusal/genética , Canais de Sódio/genética , Animais , Flutter Atrial/terapia , Células CHO , Cricetinae , Cricetulus , Desfibriladores Implantáveis , Eletrocardiografia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Contração Miocárdica/genética , Canal de Sódio Disparado por Voltagem NAV1.5 , Síndrome do Nó Sinusal/terapia , Resultado do Tratamento , Adulto Jovem
4.
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
5.
J Cell Biochem ; 104(4): 1244-53, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18275062

RESUMO

Inositol 1,4,5-trisphosphate (InsP(3)) receptors are calcium-release channels found in the endoplasmic/sarcoplasmic reticulum (ER/SR) membrane of diverse cell types. InsP(3) receptors release Ca(2+) from ER/SR lumenal stores in response to InsP(3) generated from various stimuli. The complex spatial and temporal patterns of InsP(3) receptor-mediated Ca(2+) release regulate many cellular processes, ranging from gene transcription to memory. Ankyrins are adaptor proteins implicated in the targeting of ion channels and transporters to specialized membrane domains. Multiple independent studies have documented in vitro and in vivo interactions between ankyrin polypeptides and the InsP(3) receptor. Moreover, loss of ankyrin-B leads to loss of InsP(3) receptor membrane expression and stability in cardiomyocytes. Despite extensive biochemical and functional data, the validity of in vivo ankyrin-InsP(3) receptor interactions remains controversial. This controversy is based on inconsistencies between a previously identified ankyrin-binding region on the InsP(3) receptor and InsP(3) receptor topology data that demonstrate the inaccessibility of this lumenal binding site on the InsP(3) receptor to cytosolic ankyrin polypeptides. Here we use two methods to revisit the requirements on InsP(3) receptor for ankyrin binding. We demonstrate that ankyrin-B interacts with the cytoplasmic N-terminal domain of InsP(3) receptor. In summary, our findings demonstrate that the ankyrin-binding site is located on the cytoplasmic face of the InsP(3) receptor, thus validating the feasibility of in vivo ankyrin-InsP(3) receptor interactions.


Assuntos
Anquirinas/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Anquirinas/química , Sítios de Ligação , Citoplasma , Humanos , Receptores de Inositol 1,4,5-Trifosfato/química , Miócitos Cardíacos , Ligação Proteica , Transporte Proteico , Técnicas do Sistema de Duplo-Híbrido
6.
PLoS One ; 8(10): e78087, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24155982

RESUMO

Cardiac Na channel remodeling provides a critical substrate for generation of reentrant arrhythmias in border zones of the infarcted canine heart. Recent studies show that Nav1.5 assembly and function are linked to ankyrin-G, gap, and mechanical junction proteins. In this study our objective is to expound the status of the cardiac Na channel, its interacting protein ankyrinG and the mechanical and gap junction proteins at two different times post infarction when arrhythmias are known to occur; that is, 48 hr and 5 day post coronary occlusion. Previous studies have shown the origins of arrhythmic events come from the subendocardial Purkinje and epicardial border zone. Our Purkinje cell (Pcell) voltage clamp study shows that INa and its kinetic parameters do not differ between Pcells from the subendocardium of the 48hr infarcted heart (IZPCs) and control non-infarcted Pcells (NZPCs). Immunostaining studies revealed that disturbances of Nav1.5 protein location with ankyrin-G are modest in 48 hr IZPCs. Therefore, Na current remodeling does not contribute to the abnormal conduction in the subendocardial border zone 48 hr post myocardial infarction as previously defined. In addition, immunohistochemical data show that Cx40/Cx43 co-localize at the intercalated disc (IDs) of control NZPCs but separate in IZPCs. At the same time, Purkinje cell desmoplakin and desmoglein2 immunostaining become diffuse while plakophilin2 and plakoglobin increase in abundance at IDs. In the epicardial border zone 5 days post myocardial infarction, immunoblot and immunocytochemical analyses showed that ankyrin-G protein expression is increased and re-localized to submembrane cell regions at a time when Nav1.5 function is decreased. Thus, Nav1.5 and ankyrin-G remodeling occur later after myocardial infarction compared to that of gap and mechanical junctional proteins. Gap and mechanical junctional proteins remodel in IZPCs early, perhaps to help maintain Nav1.5 subcellular location position and preserve its function soon after myocardial infarction.


Assuntos
Anquirinas/metabolismo , Ativação do Canal Iônico , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Animais , Anquirinas/química , Conexina 43/metabolismo , Conexinas/metabolismo , Cães , Junções Comunicantes/metabolismo , Cinética , Masculino , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/química , Pericárdio/metabolismo , Pericárdio/patologia , Células de Purkinje/metabolismo , Fatores de Tempo , Proteína alfa-5 de Junções Comunicantes
7.
Cardiovasc Res ; 95(3): 300-7, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22562703

RESUMO

AIMS: Female gender is a risk factor for long QT-related arrhythmias, but the underlying mechanisms remain uncertain. Here, we tested the hypothesis that gender-dependent function of the post-depolarization 'late' sodium current (I(Na-L)) contributes. METHODS AND RESULTS: Studies were conducted in mice in which the canonical cardiac sodium channel Scn5a locus was disrupted, and expression of human wild-type SCN5A cDNA substituted. Baseline QT intervals were similar in male and female mice, but exposure to the sodium channel opener anemone toxin ATX-II elicited polymorphic ventricular tachycardia in 0/9 males vs. 6/9 females. Ventricular I(Na-L) and action potential durations were increased in myocytes isolated from female mice compared with those from males before and especially after treatment with ATX-II. Further, ATX-II elicited potentially arrhythmogenic early afterdepolarizations in myocytes from 0/5 male mice and 3/5 female mice. CONCLUSION: These data identify variable late I(Na) as a modulator of gender-dependent arrhythmia susceptibility.


Assuntos
Síndrome do QT Longo/etiologia , Canal de Sódio Disparado por Voltagem NAV1.5/deficiência , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Taquicardia Ventricular/etiologia , Acetanilidas/farmacologia , Potenciais de Ação , Animais , Venenos de Cnidários , Modelos Animais de Doenças , Eletrocardiografia , Feminino , Predisposição Genética para Doença , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Piperazinas/farmacologia , Ranolazina , Fatores de Risco , Fatores Sexuais , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo
8.
J Clin Invest ; 119(4): 986-96, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19273909

RESUMO

Myocardial Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition improves cardiac function following myocardial infarction (MI), but the CaMKII-dependent pathways that participate in myocardial stress responses are incompletely understood. To address this issue, we sought to determine the transcriptional consequences of myocardial CaMKII inhibition after MI. We performed gene expression profiling in mouse hearts with cardiomyocyte-delimited transgenic expression of either a CaMKII inhibitory peptide (AC3-I) or a scrambled control peptide (AC3-C) following MI. Of the 8,600 mRNAs examined, 156 were substantially modulated by MI, and nearly half of these showed markedly altered responses to MI with CaMKII inhibition. CaMKII inhibition substantially reduced the MI-triggered upregulation of a constellation of proinflammatory genes. We studied 1 of these proinflammatory genes, complement factor B (Cfb), in detail, because complement proteins secreted by cells other than cardiomyocytes can induce sarcolemmal injury during MI. CFB protein expression in cardiomyocytes was triggered by CaMKII activation of the NF-kappaB pathway during both MI and exposure to bacterial endotoxin. CaMKII inhibition suppressed NF-kappaB activity in vitro and in vivo and reduced Cfb expression and sarcolemmal injury. The Cfb-/- mice were partially protected from the adverse consequences of MI. Our findings demonstrate what we believe is a novel target for CaMKII in myocardial injury and suggest that CaMKII is broadly important for the genetic effects of MI in cardiomyocytes.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fator B do Complemento/genética , Miocárdio/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Membrana Celular/metabolismo , Fator B do Complemento/deficiência , Expressão Gênica , Perfilação da Expressão Gênica , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/genética , Peptídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
J Cell Biol ; 180(1): 173-86, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18180363

RESUMO

Voltage-gated Na(v) channels are required for normal electrical activity in neurons, skeletal muscle, and cardiomyocytes. In the heart, Na(v)1.5 is the predominant Na(v) channel, and Na(v)1.5-dependent activity regulates rapid upstroke of the cardiac action potential. Na(v)1.5 activity requires precise localization at specialized cardiomyocyte membrane domains. However, the molecular mechanisms underlying Na(v) channel trafficking in the heart are unknown. In this paper, we demonstrate that ankyrin-G is required for Na(v)1.5 targeting in the heart. Cardiomyocytes with reduced ankyrin-G display reduced Na(v)1.5 expression, abnormal Na(v)1.5 membrane targeting, and reduced Na(+) channel current density. We define the structural requirements on ankyrin-G for Na(v)1.5 interactions and demonstrate that loss of Na(v)1.5 targeting is caused by the loss of direct Na(v)1.5-ankyrin-G interaction. These data are the first report of a cellular pathway required for Na(v) channel trafficking in the heart and suggest that ankyrin-G is critical for cardiac depolarization and Na(v) channel organization in multiple excitable tissues.


Assuntos
Anquirinas/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Animais , Anquirinas/química , Sequência de Bases , Células Cultivadas , Humanos , Ativação do Canal Iônico , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Miócitos Cardíacos/química , Canal de Sódio Disparado por Voltagem NAV1.5 , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Interferência de RNA , Ratos , Canais de Sódio/química , Transdução Genética
10.
Future Cardiol ; 1(3): 363-71, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-19804119

RESUMO

Ankyrins are a family of multivalent membrane-adaptor proteins first identified in the erythrocyte over 25 years ago as a link between the anion exchanger and the spectrin-based cytoskeleton. Since their initial discovery, ankyrin function has been linked to protein targeting and membrane domain organization in a variety of cell types including erythrocytes, neurons, epithelial cells, and cardiomyocytes. Recent findings demonstrate that dysfunction in ankyrin-based cellular pathways in the heart leads to human ventricular arrhythmia and sudden cardiac death. This review will present an overview of the ankyrin family with special emphasis on the recently identified roles of ankyrin polypeptides in ion channel and transporter targeting in the heart.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA