Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 4(7): e00677, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29998196

RESUMO

ATP-sensitive potassium channels found in both the sarcolemma (sarcKATP) and mitochondria (mitoKATP) of cardiomyocytes are important mediators of cardioprotection during ischemic heart disease. Sulfonylurea receptor isoforms (SUR2), encoded by Abcc9, an ATP-binding cassette family member, form regulatory subunits of the sarcKATP channel and are also thought to regulate mitoKATP channel activity. A short-form splice variant of SUR2 (SUR2A-55) was previously shown to target mitochondria and display diaxoxide and ATP insensitive KATP activity when co-expressed with the inward rectifier channels Kir6.2 and Kir6.1. We hypothesized that mice with cardiac specific overexpression of SUR2A-55 would mediate cardioprotection from ischemia by altering mitoKATP properties. Mice overexpressing SUR2A-55 (TGSUR2A-55) in cardiomyocytes were generated and showed no significant difference in echocardiographic measured chamber dimension, percent fractional shortening, heart to body weight ratio, or gross histologic features compared to normal mice at 11-14 weeks of age. TGSUR2A-55 had improved hemodynamic functional recovery and smaller infarct size after ischemia reperfusion injury compared to WT mice in an isolated hanging heart model. The mitochondrial membrane potential of TGSUR2A-55 mice was less sensitive to ATP, diazoxide, and Ca2+ loading. These data suggest that the SUR2A-55 splice variant favorably affects mitochondrial function leading to cardioprotection. These data support a role for the regulation of mitoKATP activity by SUR2A-55.

2.
J Am Heart Assoc ; 6(4)2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28373245

RESUMO

BACKGROUND: We have identified the cardiomyopathy-susceptibility gene vinculin (VCL) mutation M94I may account for a sudden unexplained nocturnal death syndrome (SUNDS) case. We addressed whether VCL common variant D841H is associated with SUNDS. METHODS AND RESULTS: In 8 of 120 SUNDS cases, we detected an East Asian common VCL variant p.Asp841His (D841H). Comparing the H841 allele frequency of the general population in the local database (15 of 1818) with SUNDS victims (10 of 240) gives an odds ratio for SUNDS of 5.226 (95% CI, 2.321, 11.769). The VCL-D841H variant was engineered and either coexpressed with cardiac sodium channel (SCN5A) in HEK293 cells or overexpressed in human induced pluripotent stem-cell-derived cardiomyocytes to examine its effects on sodium channel function using the whole-cell patch-clamp method. In HEK293 cells, under physiological pH conditions (pH 7.4), D841H caused a 29% decrease in peak INa amplitude compared to wild type (WT), whereas under acidotic conditions (pH 7.0), D841H decreased further to 43% along with significant negative shift in inactivation compared to WT at pH 7.4. In induced pluripotent stem-cell-derived cardiomyocytes, similar effects of D841H on INa were observed. VCL colocalized with SCN5A at the intercalated disk in human cardiomyocytes. VCL was also confirmed to directly interact with SCN5A, and VCL-D841H did not disrupt the association of VCL and SCN5A. CONCLUSIONS: A VCL common variant was genetically and biophysically associated with Chinese SUNDS. The aggravation of loss of function of SCN5A caused by VCL-D841H under acidosis supports that nocturnal sleep respiratory disorders with acidosis may play a key role in the pathogenesis of SUNDS.


Assuntos
Povo Asiático/genética , Síndrome de Brugada/genética , Variação Genética , Vinculina/genética , Adolescente , Adulto , Síndrome de Brugada/etnologia , Síndrome de Brugada/metabolismo , Síndrome de Brugada/mortalidade , China , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Potenciais da Membrana , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Fenótipo , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Transfecção , Vinculina/metabolismo , Adulto Jovem
3.
Sci Rep ; 7: 42953, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28218286

RESUMO

Sudden unexplained nocturnal death syndrome (SUNDS) remains an autopsy negative disorder with unclear etiology. Vinculin (VCL) was linked to sudden arrhythmia death in VCL knockout mice prior to the appearance of cardiomyopathy. We hypothesized VCL mutations underlie risk for SUNDS. A rare heterozygous variant VCL-M94I was found in a SUNDS victim who suffered sudden nocturnal tachypnea and lacked pathogenic variants in known arrhythmia-causing genes. VCL was identified to interact with SCN5A in vitro/vivo. The VCL-M94I was co-expressed with the cardiac sodium channel in HEK293 cells and also overexpressed in induced pluripotent stem cells derived cardiomyocytes (iPSCs-CM). In HEK293 cells with pH 7.4, VCL-M94I caused ~30% decrease in peak sodium current (INa) amplitude compared to WT; under acidotic conditions (pH 7.0) typically found with hypoxia during sleep apnea, M94I resulted in 37% reduction in peak INa compared to WT and the combination of VCL-M94I and pH 7.0 decreased peak INa by ~56% compared to WT at pH 7.4. In iPSCs-CM, similar effects of M94I on reduction of peak INa were observed. This study initially shows both physical and functional interaction between VCL and cardiac sodium channel, and suggests an important role for respiratory acidosis in triggering the fatal arrhythmia underlying SUNDS.


Assuntos
Síndrome de Brugada/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Vinculina/genética , Adolescente , Adulto , Animais , Síndrome de Brugada/genética , Feminino , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Potenciais da Membrana , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Polimorfismo Genético , Ligação Proteica , Vinculina/deficiência , Vinculina/metabolismo , Adulto Jovem
4.
Mayo Clin Proc ; 91(11): 1503-1514, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27707468

RESUMO

OBJECTIVE: To look for previously unrecognized cardiac structural abnormalities and address the genetic cause for sudden unexplained nocturnal death syndrome (SUNDS). METHODS: Data for 148 SUNDS victims and 444 controls (matched 1:3 on sex, race, and age of death within 1 year) were collected from Sun Yat-sen University from January 1, 1998, to December 31, 2014, to search morphological changes. An additional 17 patients with Brugada syndrome (BrS) collected from January 1, 2006, to December 31, 2014, served as a comparative disease cohort. Target-captured next-generation sequencing for 80 genes associated with arrhythmia/cardiomyopathy was performed in 44 SUNDS victims and 17 patients with BrS to characterize the molecular spectrum. RESULTS: The SUNDS victims had slight but statistically significant increased heart weight and valve circumference compared with controls. Twelve of 44 SUNDS victims (SCN5A, SCN1B, CACNB2, CACNA1C, AKAP9, KCNQ1, KCNH2, KCNJ5, GATA4, NUP155, ABCC9) and 6 of 17 patients with BrS (SCN5A, CACNA1C; P>.05) carried rare variants in primary arrhythmia-susceptibility genes. Only 2 of 44 SUNDS cases compared with 5 of 17 patients with BrS hosted a rare variant in the most common BrS-causing gene, SCN5A (P=.01). Using the strict American College of Medical Genetics guideline-based definition, it was found that only 2 of 44 (KCNQ1) SUNDS and 3 of 17 (SCN5A) patients with BrS hosted a "(likely) pathogenic" variant. Fourteen of 44 SUNDS cases with cardiomyopathy-related variants had a subtle but significantly decreased circumference of cardiac valves, and tended to die on average 5 to 6 years younger compared with the remaining 30 cases (P=.02). CONCLUSION: We present the first comprehensive autopsy evidence that SUNDS victims may have concealed cardiac morphological changes. SUNDS and BrS may result from different molecular pathological underpinnings. The distinct association between cardiomyopathy-related rare variants and SUNDS warrants further investigation.


Assuntos
Arritmias Cardíacas/genética , Cardiomiopatias/genética , Morte Súbita/epidemiologia , Predisposição Genética para Doença , Heterozigoto , Miocárdio/patologia , Adulto , Autopsia , Cardiomiopatias/patologia , Estudos de Casos e Controles , China/epidemiologia , Morte Súbita/etiologia , Feminino , Valvas Cardíacas/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Proteínas de Membrana Transportadoras/genética , Pessoa de Meia-Idade , Mutação , Tamanho do Órgão
6.
Circ Res ; 116(12): 1937-55, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26044249

RESUMO

Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.


Assuntos
Morte Súbita Cardíaca/etiologia , Cardiopatias/metabolismo , Miocárdio/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/metabolismo , Sinalização do Cálcio , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Morte Súbita Cardíaca/prevenção & controle , Junções Comunicantes/fisiologia , Sistema de Condução Cardíaco/fisiopatologia , Homeostase , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Potenciais da Membrana , Doenças Metabólicas/complicações , Doenças Metabólicas/fisiopatologia , Mitocôndrias Cardíacas/metabolismo , Oxirredução , Estresse Oxidativo , Potássio/fisiologia , Espécies Reativas de Oxigênio , Sódio/fisiologia
7.
J Am Heart Assoc ; 3(6): e001491, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25497881

RESUMO

BACKGROUND: The mouse ether-a-go-go-related gene 1a (mERG1a, mKCNH2) encodes mERG K(+) channels in mouse cardiomyocytes. The mERG channels and their human analogue, hERG channels, conduct IKr. Mutations in hERG channels reduce IKr to cause congenital long-QT syndrome type 2, mostly by decreasing surface membrane expression of trafficking-deficient channels. Three cDNA sequences were originally reported for mERG channels that differ by 1 to 4 amino acid residues (mERG-London, mERG-Waterston, and mERG-Nie). We characterized these mERG channels to test the postulation that they would differ in their protein trafficking and biophysical function, based on previous findings in long-QT syndrome type 2. METHODS AND RESULTS: The 3 mERG and hERG channels were expressed in HEK293 cells and neonatal mouse cardiomyocytes and were studied using Western blot and whole-cell patch clamp. We then compared our findings with the recent sequencing results in the Welcome Trust Sanger Institute Mouse Genomes Project (WTSIMGP). CONCLUSIONS: First, the mERG-London channel with amino acid substitutions in regions of highly ordered structure is trafficking deficient and undergoes temperature-dependent and pharmacological correction of its trafficking deficiency. Second, the voltage dependence of channel gating would be different for the 3 mERG channels. Third, compared with the WTSIMGP data set, the mERG-Nie clone is likely to represent the wild-type mouse sequence and physiology. Fourth, the WTSIMGP analysis suggests that substrain-specific sequence differences in mERG are a common finding in mice. These findings with mERG channels support previous findings with hERG channel structure-function analyses in long-QT syndrome type 2, in which sequence changes in regions of highly ordered structure are likely to result in abnormal protein trafficking.


Assuntos
Clonagem Molecular , Canais de Potássio Éter-A-Go-Go/metabolismo , Síndrome do QT Longo/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Animais Recém-Nascidos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Predisposição Genética para Doença , Células HEK293 , Humanos , Ativação do Canal Iônico , Síndrome do QT Longo/genética , Potenciais da Membrana , Camundongos da Linhagem 129 , Mutação , Fenótipo , Transporte Proteico , Análise de Sequência de DNA , Fatores de Tempo , Transfecção
8.
Card Electrophysiol Clin ; 6(4): 797-809, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25395996

RESUMO

Sodium current in the heart flows principally through the pore protein NaV1.5, which is part of a complex of interacting proteins that serve both to target and localize the complex in the membrane, and to modulate function by such post-translational modifications as phosphorylation and nitrosylation. Multiple mutations in seven different NaV1.5 interacting proteins have been associated with dysfunctional sodium current and inherited cardiac diseases, including long QT syndrome, Brugada syndrome, atrial fibrillation, and cardiomyopathy, as well as sudden infant death syndrome (SIDS). Mutations in as yet unidentified interacting proteins may account for cardiac disease for which a genetic basis has not yet been established. Characterizing the mechanisms by which these mutations cause disease may give insight into etiologies and treatments of more common acquired cardiac disease, such as ischemia and heart failure.

9.
J Mol Cell Cardiol ; 61: 34-43, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23557754

RESUMO

The cardiac sodium current underlies excitability in heart, and inherited abnormalities of the proteins regulating and conducting this current cause inherited arrhythmia syndromes. This review focuses on inherited mutations in non-pore forming proteins of sodium channel complexes that cause cardiac arrhythmia, and the deduced mechanisms by which they affect function and dysfunction of the cardiac sodium current. Defining the structure and function of these complexes and how they are regulated will contribute to understanding the possible roles for this complex in normal and abnormal physiology and homeostasis. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".


Assuntos
Canalopatias/genética , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Canalopatias/metabolismo , Canalopatias/fisiopatologia , Homeostase , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Contração Miocárdica , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
10.
Exp Eye Res ; 110: 136-41, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23116563

RESUMO

Autosomal dominant congenital cataracts have been associated with mutations of genes encoding several soluble and membrane proteins. By candidate gene screening, we identified a novel mutation in MIP (c.494 G > A) that segregates with a congenital lamellar cataract within a south Indian family and causes the replacement of a highly conserved glycine by aspartate (G165D) within aquaporin0 (AQP0). Unlike wild type AQP0, expression of AQP0-G165D in Xenopus oocytes did not facilitate swelling in hypotonic medium. In transfected HeLa cells, wild type AQP0 localized at the plasma membrane while AQP0-G165D was retained within the secretory pathway, and localized mainly within the endoplasmic reticulum. These results suggest that mutation of this conserved glycine residue leads to improper trafficking of AQP0-G165D and loss of water channel function. They emphasize the importance of AQP0 for maintenance of lens transparency and identify a critical residue that is conserved among aquaporins, but has not previously been associated with disease-associated replacement.


Assuntos
Aquaporinas/genética , Catarata/congênito , Catarata/genética , Proteínas do Olho/genética , Mutação de Sentido Incorreto , Animais , Povo Asiático/genética , Água Corporal/metabolismo , Catarata/metabolismo , Extração de Catarata , Membrana Celular/metabolismo , Movimento Celular/genética , Análise Mutacional de DNA , Regulação da Expressão Gênica/fisiologia , Genes Dominantes , Humanos , Lactente , Implante de Lente Intraocular , Cristalino/metabolismo , Masculino , Microscopia de Fluorescência , Oócitos/metabolismo , Linhagem , Reação em Cadeia da Polimerase , Estudos Prospectivos , Transporte Proteico/genética , Transfecção , Xenopus laevis
11.
Biochim Biophys Acta ; 1818(8): 1823-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22037495

RESUMO

The amino terminal domain (NT) of the connexins consists of their first 22-23 amino acids. Site-directed mutagenesis studies have demonstrated that NT amino acids are determinants of gap junction channel properties including unitary conductance, permeability/selectivity, and gating in response to transjunctional voltage. The importance of this region has also been emphasized by the identification of multiple disease-associated connexin mutants affecting amino acid residues in the NT region. The first part of the NT is α-helical. The structure of the Cx26 gap junction channel shows that the NT α-helix localizes within the channel, and lines the wall of the pore. Interactions of the amino acid residues in the NT with those in the transmembrane helices may be critical for holding the channel open. The predicted sites of these interactions and the applicability of the Cx26 structure to the NT of other connexins are considered. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Assuntos
Conexinas/química , Conexinas/fisiologia , Junções Comunicantes/metabolismo , Sequência de Aminoácidos , Aminoácidos/química , Animais , Sítios de Ligação , Conexina 26 , Citoplasma/metabolismo , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Conformação Molecular , Dados de Sequência Molecular , Mutação , Permeabilidade , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
12.
J Biol Chem ; 284(30): 20418-27, 2009 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-19478091

RESUMO

The cytoplasmic N-terminal domain of connexins has been implicated in multiple aspects of gap junction function, including connexin trafficking/assembly and channel gating. A synthetic peptide corresponding to the first 23 amino acids of human connexin37 was prepared, and circular dichroism and nuclear magnetic resonance studies showed that this N-terminal peptide was predominantly alpha-helical between glycine 5 and glutamate 16. The importance of this structure for localization of the protein at appositional membranes and channel function was tested by expression of site-directed mutants of connexin37 in which amino acids leucine 10 and glutamine 15 were replaced with prolines or alanines. Wild type connexin37 and both substitution mutants localized to appositional membranes between transfected HeLa cells. The proline mutant did not allow intercellular transfer of microinjected neurobiotin; the alanine mutant allowed transfer, but less extensively than wild type connexin37. When expressed alone in Xenopus oocytes, wild type connexin37 produced hemichannel currents, but neither of the double substitution mutants produced detectable currents. The proline mutant (but not the alanine mutant) inhibited co-expressed wild type connexin37. Taken together, our data suggest that the alpha-helical structure of the connexin37 N terminus may be dispensable for protein localization, but it is required for channel and hemichannel function.


Assuntos
Conexinas/química , Conexinas/metabolismo , Sequência de Aminoácidos , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Dicroísmo Circular , Conexinas/genética , Junções Comunicantes/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Peptídeos/síntese química , Peptídeos/química , Estrutura Secundária de Proteína , Xenopus , Proteína alfa-4 de Junções Comunicantes
13.
J Physiol Sci ; 59(3): 217-25, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19340536

RESUMO

Experimental data accumulated over the past decade show the emerging importance of the late sodium current (I(NaL)) for the function of both normal and, especially, failing myocardium, in which I(NaL) is reportedly increased. While recent molecular studies identified the cardiac Na(+) channel (NaCh) alpha subunit isoform (Na(v)1.5) as a major contributor to I (NaL), the molecular mechanisms underlying alterations of I(NaL) in heart failure (HF) are still unknown. Here we tested the hypothesis that I(NaL) is modulated by the NaCh auxiliary beta subunits. tsA201 cells were transfected simultaneously with human Na(v)1.5 (former hH1a) and cardiac beta(1) or beta(2) subunits, and whole-cell patch-clamp experiments were performed. We found that I(NaL) decay kinetics were significantly slower in cells expressing alpha + beta(1) (time constant tau = 0.73 +/- 0.16 s, n = 14, mean +/- SEM, P < 0.05) but remained unchanged in cells expressing alpha + beta(2) (tau = 0.52 +/- 0.09 s, n = 5), compared with cells expressing Na(v)1.5 alone (tau = 0.54 +/- 0.09 s, n = 20). Also, beta(1), but not beta(2), dramatically increased I(NaL) relative to the maximum peak current, I(NaT) (2.3 +/- 0.48%, n = 14 vs. 0.48 +/- 0.07%, n = 6, P < 0.05, respectively) and produced a rightward shift of the steady-state availability curve. We conclude that the auxiliary beta(1) subunit modulates I(NaL), produced by the human cardiac Na(+) channel Na(v)1.5 by slowing its decay and increasing I(NaL) amplitude relative to I(NaT). Because expression of Na(v)1.5 reportedly decreases but beta(1) remains unchanged in chronic HF, the relatively higher expression of beta(1) may contribute to the known I(NaL) increase in HF via the modulation mechanism found in this study.


Assuntos
Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Subunidades Proteicas/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Células Cultivadas , Insuficiência Cardíaca/metabolismo , Humanos , Rim/citologia , Rim/metabolismo , Modelos Biológicos , Miocárdio/citologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Transfecção
14.
J Cell Sci ; 121(Pt 16): 2744-50, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18664489

RESUMO

The cytoplasmic N-termini of connexins have been implicated in protein trafficking, oligomerization and channel gating. To elucidate the role of the N-terminus in connexin37 (CX37), we studied mutant constructs containing partial deletions of its 23 N-terminal amino acids and a construct with a complete N-terminus in which residues 2-8 were replaced with alanines. All mutants containing nine or more N-terminal amino acids form gap junction plaques in transiently transfected HeLa cells, whereas most of the longer deletions do not. Although wild-type CX37 allowed intercellular transfer of microinjected neurobiotin in HeLa cells and formed conducting hemichannels in Xenopus oocytes, none of the mutant constructs tested show evidence of channel function. However, in coexpression experiments, N-terminal mutants that formed gap junction plaques potently inhibit hemichannel conductance of wild-type CX37 suggesting their co-oligomerization. We conclude that as much as half the length of the connexin N-terminus can be deleted without affecting formation of gap junction plaques, but an intact N-terminus is required for hemichannel gating and intercellular communication.


Assuntos
Conexinas/química , Conexinas/fisiologia , Junções Comunicantes/metabolismo , Alanina/genética , Alanina/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos/fisiologia , Comunicação Celular , Conexinas/genética , Células HeLa , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína/fisiologia , Deleção de Sequência/fisiologia , Transfecção , Proteína alfa-4 de Junções Comunicantes
15.
Am J Physiol Heart Circ Physiol ; 295(2): H667-76, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18552167

RESUMO

Late Na(+) current (I(NaL)) is a major component of the action potential plateau in human and canine myocardium. Since I(NaL) is increased in heart failure and ischemia, it represents a novel potential target for cardioprotection. However, the molecular identity of I(NaL) remains unclear. We tested the hypothesis that the cardiac Na(+) channel isoform (Na(v)1.5) is a major contributor to I(NaL) in adult dog ventricular cardiomyocytes (VCs). Cultured VCs were exposed to an antisense morpholino-based oligonucleotide (Na(v)1.5 asOligo) targeting the region around the start codon of Na(v)1.5 mRNA or a control nonsense oligonucleotide (nsOligo). Densities of both transient Na(+) current (I(NaT)) and I(NaL) (both in pA/pF) were monitored by whole cell patch clamp. In HEK293 cells expressing Na(v)1.5 or Na(v)1.2, Na(v)1.5 asOligo specifically silenced functional expression of Na(v)1.5 (up to 60% of the initial I(NaT)) but not Na(v)1.2. In both nsOligo-treated controls and untreated VCs, I(NaT) and I(NaL) remained unchanged for up to 5 days. However, both I(NaT) and I(NaL) decreased exponentially with similar time courses (tau = 46 and 56 h, respectively) after VCs were treated with Na(v)1.5 asOligo without changes in 1) decay kinetics, 2) steady-state activation and inactivation, and 3) the ratio of I(NaL) to I(NaT). Four days after exposure to Na(v)1.5 asOligo, I(NaT) and I(NaL) amounted to 68 +/- 6% (mean +/- SE; n = 20, P < 0.01) and 60 +/- 7% (n = 11, P < 0.018) of those in VCs treated by nsOligo, respectively. We conclude that in adult dog heart Na(v)1.5 sodium channels have a "functional half-life" of approximately 35 h (0.69tau) and make a major contribution to I(NaL).


Assuntos
Inativação Gênica , Morfolinas/metabolismo , Miócitos Cardíacos/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Animais , Células Cultivadas , Cães , Humanos , Cinética , Potenciais da Membrana , Modelos Cardiovasculares , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Canais de Sódio/genética , Transfecção
16.
Mol Biol Evol ; 25(6): 1016-24, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18258611

RESUMO

Approximately 75% of vertebrate proteins belong to protein families encoded by multiple evolutionarily related genes, a pattern that emerged as a result of gene and genome duplications over the course of vertebrate evolution. In families of genes with similar or related functions, adaptation to a strong selective agent should involve multiple adaptive changes across the entire gene family. However, we know of no evolutionary studies that have explicitly addressed this point. Here, we show how 4 taxonomically diverse species of pufferfishes (Tetraodontidae) each evolved resistance to the guanidinium toxins tetrodotoxin (TTX) and saxitoxin (STX) via parallel amino acid replacements across all 8 sodium channels present in teleost fish genomes. This resulted in diverse suites of coexisting sodium channel types that all confer varying degrees of toxin resistance, yet show remarkable convergence among genes and phylogenetically diverse species. Using site-directed mutagenesis and expression of a vertebrate sodium channel, we also demonstrate that resistance to TTX/STX is enhanced up to 15-fold by single, frequently observed replacements at 2 sites that have not previously been implicated in toxin binding but show similar or identical replacements in pufferfishes and in distantly related vertebrate and nonvertebrate animals. This study presents an example of natural selection acting upon a complete gene family, repeatedly arriving at a diverse but limited number of adaptive changes within the same genome. To be maximally informative, we suggest that future studies of molecular adaptation should consider all functionally similar paralogs of the affected gene family.


Assuntos
Adaptação Biológica/genética , Evolução Molecular , Família Multigênica , Saxitoxina/toxicidade , Canais de Sódio/genética , Tetraodontiformes/genética , Tetrodotoxina/toxicidade , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Dados de Sequência Molecular , Filogenia , Porosidade , Estrutura Terciária de Proteína , Canais de Sódio/classificação , Canais de Sódio/efeitos dos fármacos , Tetraodontiformes/classificação , Tetraodontiformes/fisiologia
17.
Mol Pharmacol ; 70(5): 1514-23, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16885209

RESUMO

Mibefradil is a tetralol derivative once marketed to treat hyper-tension. Its primary target is the T-type Ca(2+) channel (IC(50), approximately 0.1-0.2 microM), but it also blocks Na(+),K(+),Cl(-), and other Ca(2+) channels at higher concentrations. We have recently reported state-dependent mibefradil block of Na(+) channels in which apparent affinity was enhanced when channels were recruited to slow-inactivated conformations. The structural determinants controlling mibefradil block have not been identified, although evidence suggests involvement of regions near or within the inner pore. We tested whether mibefradil interacts with the local anesthetic (LA) binding site, which includes residues in the S6 segments of domains (D) I, III, and IV. Mutagenesis of DIII S6 and DIVS6 did not reveal critical binding determinants. Substitution of Asn406 in DI S6 of cardiac Na(v)1.5, however, altered affinity in a manner dependent on the identity of the substituting residue. Replacing Asn406 with a phenylalanine or a cysteine increased affinity by 4- and 7-fold, respectively, thus conferring T-type Ca(2+) channel-like mibefradil sensitivity to the Na(+) channel. A series of other substitutions that varied in size, charge, and hydrophobicity had minimal effects on mibefradil block, but all mutations dramatically altered the magnitude and voltage-dependence of slow inactivation, consistent with data in other isoforms. Channels did not slow-inactivate, however, at the voltages used to assay mibefradil block, supporting the idea that Asn406 lies within or near the mibefradil binding site.


Assuntos
Asparagina/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Mibefradil/farmacologia , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Canais de Sódio/química , Canais de Sódio/metabolismo , Sequência de Aminoácidos , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.5
18.
Hum Mol Genet ; 15(9): 1497-512, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16565161

RESUMO

Highly alternatively spliced genes may provide complex targets for disease mutations. Structural changes created by missense mutations may differentially affect the activity of alternative gene products, whereas missense, silent and non-coding mutations may alter developmental regulation of splice variant expression. CACNA1H is a human gene encoding Ca(v)3.2 low-voltage-activated, T-type calcium channels associated with bursting behavior in neurons and has been linked to more than 30 mutations apparently predisposing to childhood absence epilepsy (CAE) and other idiopathic generalized epilepsies (IGEs). Biophysical properties, including the effects of missense mutations, have been evaluated previously for a single splice form of Ca(v)3.2 expressed in transformed cell lines. We here show that CACNA1H is alternatively spliced at 12-14 sites, capable of generating both functional and non-functional transcripts. Variable cytoplasmic and extracellular protein domains point to likely differences in gating behavior, sensitivity to neuromodulation and interactions with extracellular matrix. Biophysical profiles of selected physiological Ca(v)3.2 forms reveal variations in kinetics and steady-state gating parameters, most likely to affect membrane firing. These were comparable to or larger than changes reported for previously studied mutations. Missense CAE and IGE mutations were clustered near segments associated with anomalous splicing. Missense and silent mutations were found to destroy, create or change the regulatory specificity of predicted exonic splicing enhancer sequences that may control splicing regulation. We discuss a paradigm for CACNA1H expression of Ca(v)3.2 subunits, which may influence future basic and clinical studies.


Assuntos
Processamento Alternativo/genética , Canais de Cálcio Tipo T/genética , Epilepsia Generalizada/genética , Epilepsia Generalizada/metabolismo , Variação Genética , RNA/genética , Sequência de Aminoácidos , Sequência de Bases , Canais de Cálcio Tipo T/biossíntese , Canais de Cálcio Tipo T/fisiologia , Humanos , Dados de Sequência Molecular , RNA/fisiologia , Transcrição Gênica
19.
Am J Physiol Heart Circ Physiol ; 291(1): H29-37, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16501012

RESUMO

A long QT mutation in the cardiac sodium channel, D1790G (DG), shows enhanced flecainide use-dependent block (UDB). The relative importance of open and inactivated states of the channel in flecainide UDB has been controversial. We used a modifiable, inactivation-deficient mutant channel that contains the F1486C mutation in the IFM motif to investigate the UDB difference between the wild-type (WT-ICM) and DG (DG-ICM) channels. UDB at 5 Hz was greater in DG-ICM than WT-ICM, and IC50 values for steady-state UDB were 7.19 and 18.06 microM, respectively. When [2-(trimethyammonium) ethyl]methanethiosulfonate bromide (MTSET) was included in the pipette and fast inactivation was disabled, IC50 was 5.04 microM for DG-ICM and 12.63 microM for WT-ICM. We measured open-channel block by flecainide directly in MTSET-treated, noninactivating ICM channels. Steady-state block was higher for DG-ICM than WT-ICM (IC50 was 2.34 microM for DG-ICM and 5.87 microM for WT-ICM), suggesting that open-channel block is an important determinant of flecainide UDB. We obtained association (kon) and dissociation (koff) rates for open-channel block by the Langmuir-isotherm model. They were koff = 31.37 s(-1), kon = 5.83 s(-1).microM(-1), and calculated Kd = 5.38 microM for WT-ICM (where Kd = koff/kon); and koff = 24.88 s(-1), kon = 9.54 s(-1).microM(-1), and calculated Kd = 2.61 microM for DG-ICM. These Kd values were similar to IC50 measured from steady-state open-channel block. Furthermore, we modeled UDB mathematically by using these kinetic rates and found that the model predicted experimental UDB accurately. The recovery from UDB had a minor contribution to UDB. Flecainide UDB is predominantly determined by an open-channel blocking mechanism, and DG-ICM channels appeared to have an altered open-channel state with higher flecainide affinity than WT-ICM.


Assuntos
Flecainida/administração & dosagem , Ativação do Canal Iônico/efeitos dos fármacos , Rim/metabolismo , Síndrome do QT Longo/fisiopatologia , Potenciais da Membrana/efeitos dos fármacos , Canais de Sódio/metabolismo , Sódio/metabolismo , Antiarrítmicos/administração & dosagem , Linhagem Celular , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos , Rim/efeitos dos fármacos , Cinética , Modelos Biológicos , Mutagênese Sítio-Dirigida , Mutação , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética , Relação Estrutura-Atividade
20.
J Mol Cell Cardiol ; 34(11): 1477-89, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12431447

RESUMO

We previously reported an ultraslow inactivating late Na+ current (INaL) in left ventricular cardiomyocytes (VC) isolated from normal (NVC) and failing (FVC) human hearts. This current could play a role in heart failure-induced repolarization abnormalities. To identify properties of NaCh contributing to INaL, we examined early and late openings in cell-attached patches of HEK293 cells expressing human cardiac NaCh alpha-subunit (alpha-HEK) and in VC of one normal and three failing human hearts. Two types of the late NaCh openings underlay INaL in all three preparations: scattered late (SLO) and bursts (BO). Amplitude analysis revealed that slope conductance for both SLO and BO was the same compared to the main level of early openings (EO) in both VC (21 vs 22.7pS, NVC; 22.7 vs 22.6pS, FVC) and alpha-HEK (23.2 vs 23pS), respectively. Analysis of SLO latencies revealed voltage-independent ultraslow inactivation in all preparations with tendency to be slower in FVC compared to NCV. EO and SLO render one open voltage-independent state (tau approximately 0.4ms) for NVC and FVC. One open (voltage-dependent) and two closed states (one voltage-dependent and another voltage-independent) were found in BO of both specimens. Burst duration tend to be longer in FVC ( approximately 50ms) than in NVC ( approximately 30ms). In FVC we found both modes SLO and BO at membrane potential of -10mV that is attribute for take-off voltages (from -18 to -2mV) for early afterdepolarizations (EAD's) in FVC. In conclusions, we found a novel gating mode SLO that manifest slow (hundreds of ms), voltage-independent inactivation in both NVC and FVC. We were unable to reliably demonstrate any differences in the properties of the late NaCh in failing vs a normal human heart. Accordingly, the late current appears to be generated by a single population of channels in normal and failing human ventricular myocardium. Both SLO and BO could be implicated in EADs in HF.


Assuntos
Insuficiência Cardíaca/metabolismo , Ativação do Canal Iônico/fisiologia , Miocárdio/metabolismo , Canais de Sódio/fisiologia , Sódio/metabolismo , Potenciais de Ação , Células Cultivadas/fisiologia , Insuficiência Cardíaca/patologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Humanos , Transporte de Íons , Rim , Cinética , Miocárdio/citologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Proteínas Recombinantes de Fusão/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...