Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochem Biophys Res Commun ; 449(3): 278-83, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24857987

RESUMO

Atrial fibrillation (AF) is progressive and is the most common clinical arrhythmia. It is associated with inflammatory changes characterized by signal transducer and activator of transcription 3 (STAT3) signaling. A zinc finger homeobox 3 (ZFHX3, also named AT-motif binding factor 1, ATBF1) gene variant has been found in patients with AF. However, the mechanism by which the ATBF1 leads to inflammation in AF remains unknown. The aim of this study was to investigate whether tachypacing induces a decrease in ATBF1 expression and then activates STAT3 signaling via protein inhibitor of activated STAT3 (PIAS3). Atrial (HL-1 myocytes) cells were cultured in the presence of rapid electrical stimulations. In tachypaced HL-1 cells, we found that ATBF1 and PIAS3 protein levels were decreased, while the level of phosphorylated STAT3 (p-STAT3) was highly up-regulated compared with that of total STAT3. Knockdown of ATBF1 enhanced this trend, while the overexpression of ATBF1 had the opposite effect. A binary complex of ATBF1 and PIAS3 was formed and then the DNA-binding ability of activated STAT3 was enhanced in tachypaced HL-1 cells. These data indicate that tachypacing decreased ATBF1, leading to enhanced STAT3 DNA-binding activity due to the reduced formation of a binary complex of ATBF1 and PIAS3.


Assuntos
Fibrilação Atrial/fisiopatologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/fisiologia , Proteínas Inibidoras de STAT Ativados/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Fibrilação Atrial/metabolismo , Linhagem Celular , DNA/metabolismo , Regulação para Baixo , Estimulação Elétrica , Técnicas de Silenciamento de Genes , Átrios do Coração/citologia , Proteínas de Homeodomínio/genética , Inflamação/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção
2.
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
3.
Am J Physiol Heart Circ Physiol ; 302(3): H782-9, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22101522

RESUMO

Na(+) current derived from expression of the cardiac isoform SCN5A is reduced by receptor-mediated or direct activation of protein kinase C (PKC). Previous work has suggested a possible role for loss of Na(+) channels at the plasma membrane in this effect, but the results are controversial. In this study, we tested the hypothesis that PKC activation acutely modulates the intracellular distribution of SCN5A channels and that this effect can be visualized in living cells. In human embryonic kidney cells that stably expressed SCN5A with green fluorescent protein (GFP) fused to the channel COOH-terminus (SCN5A-GFP), Na(+) currents were suppressed by an exposure to PKC activation. Using confocal microscopy, colocalization of SCN5A-GFP channels with the plasma membrane under control and stimulated conditions was quantified. A separate population of SCN5A channels containing an extracellular epitope was immunolabeled to permit temporally stable labeling of the plasma membrane. Our results demonstrated that Na(+) channels were preferentially trafficked away from the plasma membrane by PKC activation, with a major contribution by Ca(2+)-sensitive or conventional PKC isoforms, whereas stimulation of protein kinase A (PKA) had the opposite effect. Removal of the conserved PKC site Ser(1503) or exposure to the NADPH oxidase inhibitor apocynin eliminated the PKC-mediated effect to alter channel trafficking, indicating that both channel phosphorylation and ROS were required. Experiments using fluorescence recovery after photobleaching demonstrated that both PKC and PKA also modified channel mobility in a manner consistent with the dynamics of channel distribution. These results demonstrate that the activation of protein kinases can acutely regulate the intracellular distribution and molecular mobility of cardiac Na(+) channels in living cells.


Assuntos
Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Transporte Proteico/fisiologia , Canais de Sódio/metabolismo , Cálcio/metabolismo , Membrana Celular/enzimologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/fisiologia , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Potenciais da Membrana/fisiologia , Contração Miocárdica/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Sódio/genética
4.
J Cardiovasc Pharmacol ; 59(3): 249-53, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22030895

RESUMO

Beta-adrenergic receptor antagonists (ß-blockers) are the therapy of choice for the long QT syndrome but their efficacy is not homogeneous: propranolol and nadolol are the most effective, whereas metoprolol is associated with more treatment failures. Propranolol has a blocking effect on the sodium current ("membrane-stabilizing" effect), and it has been hypothesized that the efficacy of nadolol might be due to a similar effect. Accordingly, we used whole-cell patch-clamp recording to assess propranolol, nadolol, and metoprolol block of wild-type or mutant cardiac sodium channels (Nav1.5) coexpressed with ß1 subunit in tsA201 cells. Nadolol had a ∼20% non-use-dependent blocking effect on peak sodium current and no effect on the persistent current evoked by the LQT3 mutant A1330D, whereas propranolol blocked Nav1.5 in a use-dependent manner and reduced A1330D persistent current. Metoprolol had no effect on either the peak or persistent current. Analysis of the biophysical properties of the channel revealed that both nadolol and propranolol cause hyperpolarizing shifts on voltage dependence of activation and steady-state inactivation, whereas metoprolol shifts only the activation curve. These results provide partial explanation for the differences between nadolol and metoprolol but do not explain the similar clinical efficacy of nadolol and propranolol.


Assuntos
Metoprolol/farmacologia , Nadolol/farmacologia , Propranolol/farmacologia , Canais de Sódio/efeitos dos fármacos , Antagonistas Adrenérgicos beta/farmacologia , Linhagem Celular , Humanos , Síndrome do QT Longo/tratamento farmacológico , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/metabolismo
5.
Circ Res ; 102(3): 364-71, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18048769

RESUMO

Mutations in SCN5A encoding the principal Na+ channel alpha-subunit expressed in human heart (Na(V)1.5) have recently been linked to an inherited form of dilated cardiomyopathy with atrial and ventricular arrhythmia. We compared the biophysical properties of 2 novel Na(V)1.5 mutations associated with this syndrome (D2/S4--R814W; D4/S3--D1595H) with the wild-type (WT) channel using heterologous expression in cultured tsA201 cells and whole-cell patch-clamp recording. Expression levels were similar among WT and mutant channels, and neither mutation affected persistent sodium current. R814W channels exhibited prominent and novel defects in the kinetics and voltage dependence of activation characterized by slower rise times and a hyperpolarized conductance-voltage relationship resulting in an increased "window current." This mutant also displayed enhanced slow inactivation and greater use-dependent reduction in peak current at fast pulsing frequencies. By contrast, D1595H channels exhibited impaired fast inactivation characterized by slower entry into the inactivated state and a hyperpolarized steady-state inactivation curve. Our findings illustrate the divergent biophysical defects caused by 2 different SCN5A mutations associated with familial dilated cardiomyopathy. Retrospective review of the published clinical data suggested that cardiomyopathy was not common in the family with D1595H, but rather sinus bradycardia was the predominant clinical finding. However, for R814W, we speculate that an increased window current coupled with enhanced slow inactivation and rate-dependent loss of channel availability provided a unique substrate predisposing myocytes to disordered Na+ and Ca2+ homeostasis leading to myocardial dysfunction.


Assuntos
Substituição de Aminoácidos , Arritmias Cardíacas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Doenças Genéticas Inatas/metabolismo , Proteínas Musculares/metabolismo , Mutação de Sentido Incorreto , Canais de Sódio/metabolismo , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Cálcio/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Linhagem Celular , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Homeostase/genética , Humanos , Transporte de Íons/genética , Proteínas Musculares/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Sódio/metabolismo , Canais de Sódio/genética
6.
J Am Soc Nephrol ; 20(12): 2556-69, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19939939

RESUMO

Loss of polycystin-2 (PC2) in mice (Pkd2(-/-)) results in total body edema, focal hemorrhage, structural cardiac defects, abnormal left-right axis, hepatorenal and pancreatic cysts, and embryonic lethality. The molecular mechanisms by which loss of PC2 leads to these phenotypes remain unknown. We generated a model to allow targeted Pkd2 inactivation using the Cre-loxP system. Global inactivation of Pkd2 produced a phenotype identical to Pkd2(-/-) mice with undetectable PC2 protein and perinatal lethality. Using various Cre mouse lines, we found that kidney, pancreas, or time-specific deletion of Pkd2 led to cyst formation. In addition, we developed an immortalized renal collecting duct cell line with inactive Pkd2; these cells had aberrant cell-cell contact, ciliogenesis, and tubulomorphogenesis. They also significantly upregulated beta-catenin, axin2, and cMyc. Our results suggest that loss of PC2 disrupts normal behavior of renal epithelial cells through dysregulation of beta-catenin-dependent signaling, revealing a potential role for this signaling pathway in PC2-associated ADPKD.


Assuntos
Mutação , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , beta Catenina/metabolismo , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Cistos/genética , Cistos/patologia , Modelos Animais de Doenças , Feminino , Túbulos Renais Coletores/anormalidades , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/patologia , Hepatopatias/genética , Hepatopatias/patologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pancreatopatias/genética , Pancreatopatias/patologia , Fenótipo , Rim Policístico Autossômico Dominante/etiologia , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Gravidez , Transdução de Sinais , Canais de Cátion TRPP/deficiência , Canais de Cátion TRPP/metabolismo , Regulação para Cima
7.
Neuron ; 34(6): 877-84, 2002 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12086636

RESUMO

Epilepsy is a common neurological condition that reflects neuronal hyperexcitability arising from largely unknown cellular and molecular mechanisms. In generalized epilepsy with febrile seizures plus, an autosomal dominant epilepsy syndrome, mutations in three genes coding for voltage-gated sodium channel alpha or beta1 subunits (SCN1A, SCN2A, SCN1B) and one GABA receptor subunit gene (GABRG2) have been identified. Here, we characterize the functional effects of three mutations in the human neuronal sodium channel alpha subunit SCN1A by heterologous expression with its known accessory subunits, beta1 and beta2, in cultured mammalian cells. SCN1A mutations alter channel inactivation, resulting in persistent inward sodium current. This gain-of-function abnormality will likely enhance excitability of neuronal membranes by causing prolonged membrane depolarization, a plausible underlying biophysical mechanism responsible for this inherited human epilepsy.


Assuntos
Epilepsia Generalizada/genética , Proteínas do Tecido Nervoso/genética , Canais de Sódio/genética , Células Cultivadas , Clonagem Molecular , Humanos , Ativação do Canal Iônico/genética , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1 , Proteínas Recombinantes/genética
8.
Circulation ; 115(3): 361-7, 2007 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-17210839

RESUMO

BACKGROUND: The hypothesis that some cases of sudden infant death syndrome (SIDS) could be caused by long-QT syndrome (LQTS) has been supported by molecular studies. However, there are inadequate data regarding the true prevalence of mutations in arrhythmia-susceptibility genes among SIDS cases. Given the importance and potential implications of these observations, we performed a study to more accurately quantify the contribution to SIDS of LQTS gene mutations and rare variants. METHODS AND RESULTS: Molecular screening of 7 genes (KCNQ1, KCNH2, SCN5A, KCNE1, KCNE2, KCNJ2, CAV3) associated with LQTS was performed with denaturing high-performance liquid chromatography and nucleotide sequencing of genomic DNA from 201 cases diagnosed as SIDS according to the Nordic Criteria, and from 182 infant and adult controls. All SIDS and control cases originated from the same regions in Norway. Genetic analysis was blinded to diagnosis. Mutations and rare variants were found in 26 of 201 cases (12.9%). On the basis of their functional effect, however, we considered 8 mutations and 7 rare variants found in 19 of 201 cases as likely contributors to sudden death (9.5%; 95% CI, 5.8 to 14.4%). CONCLUSIONS: We demonstrated that 9.5% of cases diagnosed as SIDS carry functionally significant genetic variants in LQTS genes. The present study demonstrates that sudden arrhythmic death is an important contributor to SIDS. As these variants likely modify ventricular repolarization and QT interval duration, our results support the debated concept that an ECG would probably identify most infants at risk for sudden death due to LQTS either in infancy or later on in life.


Assuntos
Eletrocardiografia , Variação Genética , Síndrome do QT Longo/complicações , Síndrome do QT Longo/genética , Morte Súbita do Lactente/etiologia , Morte Súbita do Lactente/genética , Adulto , Arritmias Cardíacas/complicações , Arritmias Cardíacas/genética , Estudos de Casos e Controles , Caveolina 3/genética , Pré-Escolar , Feminino , Testes Genéticos/métodos , Humanos , Lactente , Síndrome do QT Longo/diagnóstico , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/genética , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.5 , Noruega , Canais de Potássio/genética , Fatores de Risco , Método Simples-Cego , Canais de Sódio/genética
9.
Circulation ; 115(3): 368-76, 2007 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-17210841

RESUMO

BACKGROUND: Mutations in genes responsible for the congenital long-QT syndrome, especially SCN5A, have been identified in some cases of sudden infant death syndrome. In a large-scale collaborative genetic screen, several SCN5A variants were identified in a Norwegian sudden infant death syndrome cohort (n=201). We present functional characterization of 7 missense variants (S216L, R680H, T1304M, F1486L, V1951L, F2004L, and P2006A) and 1 in-frame deletion allele (delAL586-587) identified by these efforts. METHODS AND RESULTS: Whole-cell sodium currents were measured in tsA201 cells transiently transfected with recombinant wild-type or mutant SCN5A cDNA (hH1) coexpressed with the human beta1 subunit. All variants exhibited defects in the kinetics and voltage dependence of inactivation. Five variants (S216L, T1304M, F1486L, F2004L, and P2006A) exhibited significantly increased persistent sodium currents (range, 0.5% to 1.7% of peak current) typical of SCN5A mutations associated with long-QT syndrome. These same 5 variants also displayed significant depolarizing shifts in voltage dependence of inactivation (range, 5 to 14 mV) and faster recovery from inactivation, but F1486L uniquely exhibits a depolarizing shift in the conductance-voltage relationship. Three alleles (delAL586-587, R680H, and V1951L) exhibited increased persistent current only under conditions of internal acidosis (R680H) or when expressed in the context of a common splice variant (delQ1077), indicating that they have a latent dysfunctional phenotype. CONCLUSIONS: Our present results greatly expand the spectrum of functionally characterized SCN5A variants associated with sudden infant death syndrome and provide further biophysical correlates of arrhythmia susceptibility in this syndrome.


Assuntos
Eletrocardiografia , Síndrome do QT Longo/complicações , Síndrome do QT Longo/genética , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Mutação de Sentido Incorreto/genética , Canais de Sódio/genética , Canais de Sódio/fisiologia , Morte Súbita do Lactente/etiologia , Morte Súbita do Lactente/genética , Alelos , Arritmias Cardíacas/complicações , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Estudos de Coortes , DNA Complementar/genética , Eletrofisiologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Testes Genéticos/métodos , Variação Genética/genética , Humanos , Lactente , Síndrome do QT Longo/fisiopatologia , Matemática , Canal de Sódio Disparado por Voltagem NAV1.5 , Noruega , Fenótipo , Fatores de Risco
10.
J Clin Invest ; 112(7): 1019-28, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14523039

RESUMO

Sick sinus syndrome (SSS) describes an arrhythmia phenotype attributed to sinus node dysfunction and diagnosed by electrocardiographic demonstration of sinus bradycardia or sinus arrest. Although frequently associated with underlying heart disease and seen most often in the elderly, SSS may occur in the fetus, infant, and child without apparent cause. In this setting, SSS is presumed to be congenital. Based on prior associations with disorders of cardiac rhythm and conduction, we screened the alpha subunit of the cardiac sodium channel (SCN5A) as a candidate gene in ten pediatric patients from seven families who were diagnosed with congenital SSS during the first decade of life. Probands from three kindreds exhibited compound heterozygosity for six distinct SCN5A alleles, including two mutations previously associated with dominant disorders of cardiac excitability. Biophysical characterization of the mutants using heterologously expressed recombinant human heart sodium channels demonstrate loss of function or significant impairments in channel gating (inactivation) that predict reduced myocardial excitability. Our findings reveal a molecular basis for some forms of congenital SSS and define a recessive disorder of a human heart voltage-gated sodium channel.


Assuntos
Mutação , Síndrome do Nó Sinusal/congênito , Síndrome do Nó Sinusal/genética , Canais de Sódio/genética , Eletrocardiografia , Feminino , Heterozigoto , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5 , Subunidades Proteicas , Síndrome do Nó Sinusal/fisiopatologia
11.
Cardiovasc Res ; 72(2): 250-61, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16973141

RESUMO

OBJECTIVE: Na(+) current derived from expression of the principal cardiac Na(+) channel, Na(v)1.5, is increased by activation of protein kinase A (PKA). This effect is blocked by inhibitors of cell membrane recycling, or removal of a cytoplasmic endoplasmic reticulum (ER) retention motif, suggesting that PKA stimulation increases trafficking of cardiac Na(+) channels to the plasma membrane. METHODS: To test this hypothesis, green fluorescent protein (GFP) was fused to Na(v)1.5 (Na(v)1.5-GFP), and the effects of PKA activation were investigated in intact, living cells that stably expressed the fusion protein. Using confocal microscopy, the spatial relationship of GFP-tagged channels relative to the plasma membrane was quantitated using a measurement that could control for variables present during live-cell imaging, and permit an unbiased analysis for all cells in a given field. RESULTS: In the absence of kinase stimulation, intracellular fluorescence representing Na(v)1.5-GFP channels was greatest in the perinuclear area, with additional concentration of channels beneath the cell surface. Activation of PKA promoted trafficking of Na(+) channels from both regions to the plasma membrane. Experimental results using a chemiluminescence-based assay further confirmed that PKA stimulation increased expression of Na(v)1.5 channels at the cell membrane. CONCLUSIONS: Our results provide direct evidence for PKA-mediated trafficking of cardiac Na(+) channels into the plasma membrane in living, mammalian cells, and they support the existence of multiple intracellular storage pools of channel protein that can be mobilized following a physiologic stimulus.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Rim/metabolismo , Canais de Sódio/metabolismo , Animais , Western Blotting/métodos , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Ativação Enzimática , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luminescência , Microscopia Confocal , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Oócitos/metabolismo , Técnicas de Patch-Clamp , Transporte Proteico , Canais de Sódio/genética , Transfecção/métodos , Xenopus
12.
Medicine (Baltimore) ; 95(15): e3323, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27082579

RESUMO

Mitochondrial DNA (mtDNA) copy number variation (CNV), which reflects the oxidant-induced cell damage, has been observed in a wide range of human diseases. However, whether it correlates with heart failure, which is closely related to oxidative stress, has never been elucidated before. We aimed to systematically investigate the associations between leukocyte mtDNA CNV and heart failure risk and prognosis. A total of 1700 hospitalized patients with heart failure and 1700 age- and sex-matched community population were consecutively enrolled in this observational study, as well as 1638 (96.4%) patients were followed prospectively for a median of 17 months (12-24 months). The relative mtDNA copy number of leukocyte of peripheral blood or cardiac tissue was measured in triplicate by quantitative real-time PCR method. Patients with heart failure possessed much lower relative mtDNA copy number compared with control subjects (median 0.83, interquartile range [IQR] 0.60-1.16 vs median 1.00, IQR 0.47-2.20; P < 0.001), especially for the patients with ischemic etiology (median, 0.77 for ischemic and 0.91 for non-ischemic, P < 0.001). Patients with lower mtDNA copy number exhibited 1.7 times higher risk of heart failure (odds ratio 1.71, 95% confidence interval [CI] 1.48-1.97, P < 0.001). Long-term follow-up (median of 17 months) showed that decreased mtDNA copy number was significant associated with both increased cardiovascular deaths (hazard ratio [HR] 1.58, 95% CI 1.16-2.16, P = 0.004) and cardiovascular rehospitalization (HR 1.48, 95% CI 1.21-1.82, P < 0.001). After adjusting for the conventional risk factors and medications, lower mtDNA copy numbers were still significantly associated with 50% higher cardiovascular mortality (P = 0.035). In conclusion, mtDNA copy number depletion is an independent risk factor for heart failure and predicts higher cardiovascular mortality in patients with heart failure.


Assuntos
Variações do Número de Cópias de DNA , DNA Mitocondrial/análise , Insuficiência Cardíaca , Leucócitos/metabolismo , Idoso , Estudos de Casos e Controles , China , Feminino , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Estresse Oxidativo , Prognóstico , Modelos de Riscos Proporcionais , Fatores de Risco
13.
Eur J Med Genet ; 59(8): 396-400, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27373676

RESUMO

Genetic factor plays an important role in cardiac arrhythmias. Several loci have been identified associated with this disease. However, they only explained parts of it and more genes and loci remain to be identified. In present study, we recruited a four generation family from the north of China. Four members of this family were diagnosed with atrial fibrillation by electrocardiogram (ECG). We used Exome Sequencing and Sanger sequencing to explore the candidate mutation for cardiac arrhythmia in this family. A nonsense mutation (c.G1494A, p.Trp498Ter) in the LMNA gene were identified as the candidate mutation. This variant is a novel mutation and has not yet been reported for any actual databases. This novel mutation co-segregated exactly with the disease in this family. Meanwhile, it was not detected in 524 control subjects of matched ancestry. According to structural model prediction, the mutation is expected to affect the Lamin Tail Domain (LTD) of lamin A/C protein. So the nonsense mutation discovered in the family probably was a novel mutation associated with familial atrial fibrillation. This discovery expands the mutation spectrum of LMNA and indicates the importance of LMNA in AF.


Assuntos
Fibrilação Atrial/diagnóstico , Fibrilação Atrial/genética , Códon sem Sentido , Lamina Tipo A/genética , Sequência de Aminoácidos , Biologia Computacional/métodos , Análise Mutacional de DNA , Eletrocardiografia , Exoma , Família , Feminino , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lamina Tipo A/química , Masculino , Modelos Moleculares , Linhagem , Conformação Proteica , Relação Estrutura-Atividade
14.
Heart Rhythm ; 13(7): 1515-20, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27033637

RESUMO

BACKGROUND: In Brugada syndrome (BrS), spontaneous type 1 electrocardiogram (ECG) is an established risk marker for fatal arrhythmias whereas drug-induced type 1 ECG shows a relatively benign prognosis. No study has analyzed the prognosis of fever-induced type 1 ECG (F-type1) in a large BrS cohort. OBJECTIVES: The objectives of this study were to assess the prognosis of F-type1 in asymptomatic BrS and to compare the effects of fever and drugs on ECG parameters. METHODS: One hundred twelve patients with BrS who developed F-type1 were retrospectively enrolled. Prognosis was evaluated in 88 asymptomatic patients. In a subgroup (n = 52), ECG parameters of multiple ECGs (at baseline, during fever, and after drug challenge) were analyzed. RESULTS: Eighty-eight asymptomatic patients had a mean age of 45.8 ± 18.7 years, and 71.6% (67 of 88) were men. Twenty-one percent (18 of 88) had a family history of sudden cardiac death, and 26.4% (14 of 53) carried a pathogenic SCN5A mutation. Drug challenge was positive in 29 of 36 patients tested (80.6%). The risk of ventricular fibrillation in asymptomatic patients was 0.9%/y (3 of 88; 43.6 ± 37.4 months). ST-segment elevation in lead V2 during fever and after drug challenge was not significantly different (0.41 ± 0.21 ms during fever and 0.40 ± 0.30 ms after drug challenge; P > .05). Fever shortened the PR interval compared to baseline, whereas drug challenge resulted in prolonged PR interval and QRS duration (PR interval: 169 ± 29 ms at baseline, 148 ± 45 ms during fever, and 202 ± 35 ms after drug challenge; QRS duration: 97 ± 18 ms at baseline, 92 ± 28 ms during fever, and 117 ± 21 ms after drug challenge). CONCLUSION: Patients with BrS who develop F-type1 are at risk of arrhythmic events. F-type1 appears to develop through a more complex mechanism as compared with drug-induced type 1 ECG.


Assuntos
Síndrome de Brugada , Eletrocardiografia/métodos , Febre/complicações , Sistema de Condução Cardíaco , Bloqueadores dos Canais de Sódio/efeitos adversos , Fibrilação Ventricular , Adulto , Doenças Assintomáticas , Síndrome de Brugada/diagnóstico , Síndrome de Brugada/fisiopatologia , Feminino , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Pacientes Ambulatoriais , Prognóstico , Estudos Retrospectivos , Medição de Risco , Fibrilação Ventricular/diagnóstico , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/fisiopatologia
15.
Circulation ; 105(3): 341-6, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11804990

RESUMO

BACKGROUND: Three distinct cardiac arrhythmia disorders, the long-QT syndrome, Brugada syndrome, and conduction system disease, have been associated with heterozygous mutations in the cardiac voltage-gated sodium channel alpha-subunit gene (SCN5A). We present clinical, genetic, and biophysical features of 2 new SCN5A mutations that result in atrioventricular (AV) conduction block. Methods and Results- SCN5A was used as a candidate gene in 2 children with AV block. Molecular genetic studies revealed G to A transition mutations that resulted in the substitution of serine for glycine (G298S) in the domain I S5-S6 loop and asparagine for aspartic acid (D1595N) within the S3 segment of domain IV. The functional consequences of G298S and D1595N were assessed by whole-cell patch clamp recording of recombinant mutant channels coexpressed with the beta1 subunit in a cultured cell line (tsA201). Both mutations impair fast inactivation but do not exhibit sustained non-inactivating currents. The mutations also reduce sodium current density and enhance slower inactivation components. Action potential simulations predict that this combination of biophysical abnormalities will significantly slow myocardial conduction velocity. CONCLUSIONS: A distinct pattern of biophysical abnormalities not previously observed for any other SCN5A mutant have been recognized in association with AV block. These data provide insight into the distinct clinical phenotypes resulting from mutation of a single ion channel.


Assuntos
Nó Atrioventricular , Bloqueio Cardíaco/genética , Mutação , Canais de Sódio/genética , Potenciais de Ação , Adulto , Sequência de Aminoácidos , Nó Atrioventricular/fisiopatologia , Linhagem Celular , Criança , Simulação por Computador , Eletrocardiografia , Feminino , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/fisiopatologia , Humanos , Cinética , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Canais de Sódio/química , Canais de Sódio/metabolismo
16.
Int J Clin Exp Med ; 8(4): 5485-91, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26131127

RESUMO

The involvement of the AGC protein kinase family in regulating arrhythmia has drawn considerable attention, but the underlying mechanisms are still not clear. The aim of this study is to explore the role of 3-phosphoinositide-dependent protein kinase-1 (PDK1), one of upstream protein kinases of the AGC protein kinase family, in the pathogenesis of dysregulated electrophysiological basis. PDK1(F/F) αMHC-Cre mice and PDK1(F/F) mice were divided into experiment group and control group. Using patch clamping technology, we explored action potential duration in both groups, and investigated the functions of transient outward potassium channel and L-type Ca(2+) channel to explain the abnormal action potential duration. Significant prolongation action potential duration was found in mice with PDK1 deletion. Further, the peak current of transient outward potassium current and L-type Ca(2+) current were decreased by 84% and 49% respectively. In addition, dysregulation of channel kinetics lead to action potential duration prolongation further. In conclusion, we have demonstrated that PDK1 participates in action potential prolongation in cardiac ablation of PDK1 mice. This effect is likely to be mediated largely through downregulation of transient outward potassium current. These findings indicate the modulation of the PDK1 pathway could provide a new mechanism for abnormal electrophysiological basis.

17.
Eur J Pharmacol ; 754: 52-60, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25701724

RESUMO

A-803467 is a selective Nav1.8 blocker, but its mechanism of action at cardiac sodium channels is uncertain. Thus, we investigated the mechanistic effects of A-803467 on cardiac sodium channels in isolated mouse ventricular myocytes and in human embryonic kidney 293 (HEK293) cell lines that transiently expressed Nav1.5/SCN5A, the predominant cardiac sodium channel. At 0.3µM and greater, A-803467 blocked cardiac sodium currents in a dose-dependent manner in both ventricular myocytes and in SCN5A-expressing HEK293 cell lines. In both models, the drug caused significant depolarizing shifts at the conductance voltage relationship midpoint, hyperpolarizing shifts in voltage-dependent channel inactivation, and slower recovery from inactivation. Also, the drug reduced sodium current amplitude in a frequency-dependent manner, and blocked late sodium currents, accelerated inactivation, and enhanced the intermediate inactivation state. Our results provide strong evidence that A-803467 affects multiple biophysical characteristics of the canonical cardiac Nav1.5 channel and our data can be used to study potential applications of A-803467 as an antiarrhythmic drug.


Assuntos
Compostos de Anilina/farmacologia , Furanos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Condutividade Elétrica , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Bloqueadores dos Canais de Sódio/farmacologia
18.
PLoS One ; 10(3): e0122436, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25781322

RESUMO

BACKGROUND: The AGC protein kinase family regulates multiple cellular functions. 3-phosphoinositide-dependent protein kinase-1 (PDK1) is involved in the pathogenesis of arrhythmia, and its downstream factor, Forkhead box O1 (Foxo1), negatively regulates the expression of the cardiac sodium channel, Nav1.5. Mice are known to die suddenly after PDK1 deletion within 11 weeks, but the underlying electrophysiological bases are unclear. Thus, the aim of this study was to investigate the potential mechanisms between PDK1 signaling pathway and cardiac sodium current. METHODS AND RESULTS: Using patch clamp and western blotting techniques, we investigated the role of the PDK1-Foxo1 pathway in PDK1 knockout mice and cultured cardiomyocytes. We found that PDK1 knockout mice undergo slower heart rate, prolonged QRS and QTc intervals and abnormal conduction within the first few weeks of birth. Furthermore, the peak sodium current is decreased by 33% in cells lacking PDK1. The phosphorylation of Akt (308T) and Foxo1 (24T) and the expression of Nav1.5 in the myocardium of PDK1-knockout mice are decreased, while the nuclear localization of Foxo1 is increased. The role of the PDK1-Foxo1 pathway in regulating Nav1.5 levels and sodium current density was verified using selective PDK1, Akt and Foxo1 inhibitors and isolated neonatal rat cardiomyocytes. CONCLUSION: These results indicate that PDK1 participates in the dysregulation of electrophysiological basis by regulating the PDK1-Foxo1 pathway, which in turn regulates the expression of Nav1.5 and cardiac sodium channel function.


Assuntos
Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sódio/metabolismo , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Deleção de Genes , Transporte de Íons/fisiologia , Camundongos , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Ratos , Ratos Sprague-Dawley
19.
PLoS One ; 10(2): e0117567, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25689866

RESUMO

BACKGROUND AND OBJECT: Cell apoptosis is a contributing factor in the initiation, progression and relapse of atrial fibrillation (AF), a life-threatening illness accompanied with stroke and heart failure. However, the regulatory cascade of apoptosis is intricate and remains unidentified, especially in the setting of AF. The aim of this study was to explore the roles of endoplasmic reticulum (ER) stress, mitochondrial apoptotic pathway (MAP), mitogen-activated protein kinases (MAPKs), and their cross-talking in tachypacing-induced apoptosis. METHODS AND RESULTS: HL-1 cells were cultured in the presence of tachypacing for 24 h to simulate atrial tachycardia remodeling. Results showed that tachypacing reduced cell viability measured by the cell counting kit-8, dissipated mitochondrial membrane potential detected by JC-1 staining and resulted in approximately 50% apoptosis examined by Hoechst staining and annexin V/propidium iodide staining. In addition, the proteins involved in ER stress, MAP and MAPKs were universally up-regulated or activated via phosphorylation, as confirmed by western blotting; and reversely silencing of ER stress, caspase-3 (the ultimate executor of MAP) and MAPKs with specific inhibitors prior to pacing partially alleviated apoptosis. An inhibitor of ER stress was applied to further investigate the responses of mitochondria and MAPKs to ER stress, and results indicated that suppression of ER stress comprehensively but incompletely attenuated the activation of MAP and MAPKs aroused by tachypacing, with the exception of ERK1/2, one branch of MAPKs. CONCLUSIONS: Our study suggested tachypacing-induced apoptosis is regulated by ER stress-mediated MAP and MAPKs. Thus, the above three components are all promising anti-apoptotic targets in AF patients and ER stress appears to play a dominant role due to its comprehensive effects.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Átrios do Coração/patologia , Mitocôndrias/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/patologia , Taquicardia/patologia , Animais , Remodelamento Atrial , Linhagem Celular Tumoral , Sobrevivência Celular , Sistema de Sinalização das MAP Quinases , Camundongos , Taquicardia/fisiopatologia , Fatores de Tempo
20.
Eur J Med Genet ; 58(4): 258-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25765472

RESUMO

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a cardiomyopathy that primarily involves the right ventricle. Mutations in desmosomal genes have been associated with ARVC. But its prevalence and spectrum are much less defined in the Chinese population, especially Han Chinese, a majority ethnic group in China; also the genotype-phenotype correlation regarding left ventricular involvement is still poorly understood. The aim of this study was to elucidate the genotype in Han Chinese patients with ARVC and the phenotype regarding cardiac left ventricle involvement in mutation carriers of ARVC. 48 Han Chinese patients were recruited into the present study based on the Original International Task Force Criteria of ARVC. Clinical data were reassessed according to the modified criteria published in 2010. A total of 36 subjects were diagnosed with ARVC; 12 patients were diagnosed with suspected ARVC. Five desmosomal genes (PKP2, DSG2, DSP, DSC2 and JUP) were sequenced directly from genomic DNA. Among the 36 patients, 21 mutations, 12 of which novel, were discovered in 19 individuals (19 of 36, 53%). The distribution of the mutations was 25% in PKP2, 14% in DSP, 11% in DSG2, 6% in JUP, and 3% in DSC2. Multiple mutations were identified in 2 subjects (2 of 36, 6%); both had digenic heterozygosity. Eight mutations, of which six were novel, were located in highly conserved regions. Seven mutations introduced a stop codon prematurely, which would result in premature termination of the protein synthesis. Two-dimensional echocardiography showed that LDVd and LDVs parameters were significantly larger in nonsense mutation carriers than in carriers of other mutations. In this comprehensive desmosome genetic analysis, 21 mutations were identified in five desmosomal genes in a group of 48 local Han Chinese subjects with ARVC, 12 of which were novel. PKP2 mutations were the most common variants. Left ventricular involvement could be a sign that the patient is a carrier of a nonsense cardiac desmosomal gene mutation.


Assuntos
Arritmias Cardíacas/genética , Displasia Arritmogênica Ventricular Direita/genética , Desmossomos/genética , Ventrículos do Coração/patologia , Adulto , Arritmias Cardíacas/patologia , Displasia Arritmogênica Ventricular Direita/mortalidade , Povo Asiático/genética , Sequência de Bases , China , Códon sem Sentido/genética , Análise Mutacional de DNA , Ecocardiografia , Etnicidade/genética , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Testes Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Placofilinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA