Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 111(27): 9989-94, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24951510

RESUMO

For an excitable cell to function properly, a precise number of ion channel proteins need to be trafficked to distinct locations on the cell surface membrane, through a network and anchoring activity of cytoskeletal proteins. Not surprisingly, mutations in anchoring proteins have profound effects on membrane excitability. Ca(2+)-activated K(+) channels (KCa2 or SK) have been shown to play critical roles in shaping the cardiac atrial action potential profile. Here, we demonstrate that filamin A, a cytoskeletal protein, augments the trafficking of SK2 channels in cardiac myocytes. The trafficking of SK2 channel is Ca(2+)-dependent. Further, the Ca(2+) dependence relies on another channel-interacting protein, α-actinin2, revealing a tight, yet intriguing, assembly of cytoskeletal proteins that orchestrate membrane expression of SK2 channels in cardiac myocytes. We assert that changes in SK channel trafficking would significantly alter atrial action potential and consequently atrial excitability. Identification of therapeutic targets to manipulate the subcellular localization of SK channels is likely to be clinically efficacious. The findings here may transcend the area of SK2 channel studies and may have implications not only in cardiac myocytes but in other types of excitable cells.


Assuntos
Cálcio/metabolismo , Filaminas/metabolismo , Proteínas de Membrana/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Potenciais de Ação , Animais , Animais Recém-Nascidos , Filaminas/genética , Células HEK293 , Átrios do Coração/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética
2.
Circ Res ; 107(7): 851-9, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20689065

RESUMO

RATIONALE: Ca(2+)-activated K(+) channels are present in a wide variety of cells. We have previously reported the presence of small conductance Ca(2+)-activated K(+) (SK or K(Ca)) channels in human and mouse cardiac myocytes that contribute functionally toward the shape and duration of cardiac action potentials. Three isoforms of SK channel subunits (SK1, SK2, and SK3) are found to be expressed. Moreover, there is differential expression with more abundant SK channels in the atria and pacemaking tissues compared with the ventricles. SK channels are proposed to be assembled as tetramers similar to other K(+) channels, but the molecular determinants driving their subunit interaction and assembly are not defined in cardiac tissues. OBJECTIVE: To investigate the heteromultimeric formation and the domain necessary for the assembly of 3 SK channel subunits (SK1, SK2, and SK3) into complexes in human and mouse hearts. METHODS AND RESULTS: Here, we provide evidence to support the formation of heteromultimeric complexes among different SK channel subunits in native cardiac tissues. SK1, SK2, and SK3 subunits contain coiled-coil domains (CCDs) in the C termini. In vitro interaction assay supports the direct interaction between CCDs of the channel subunits. Moreover, specific inhibitory peptides derived from CCDs block the Ca(2+)-activated K(+) current in atrial myocytes, which is important for cardiac repolarization. CONCLUSIONS: The data provide evidence for the formation of heteromultimeric complexes among different SK channel subunits in atrial myocytes. Because SK channels are predominantly expressed in atrial myocytes, specific ligands of the different isoforms of SK channel subunits may offer a unique therapeutic opportunity to directly modify atrial cells without interfering with ventricular myocytes.


Assuntos
Potenciais de Ação/fisiologia , Miócitos Cardíacos/fisiologia , Potássio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Sequência de Aminoácidos , Animais , Células Cultivadas , Átrios do Coração/citologia , Humanos , Camundongos , Dados de Sequência Molecular , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Multimerização Proteica , Estrutura Terciária de Proteína , Canais de Potássio Ativados por Cálcio de Condutância Baixa/química , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Transfecção
3.
Am J Physiol Heart Circ Physiol ; 299(5): H1459-67, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20709863

RESUMO

Adenylyl cyclase (AC) is the principal effector molecule in the ß-adrenergic receptor pathway. AC(V) and AC(VI) are the two predominant isoforms in mammalian cardiac myocytes. The disparate roles among AC isoforms in cardiac hypertrophy and progression to heart failure have been under intense investigation. Specifically, the salutary effects resulting from the disruption of AC(V) have been established in multiple models of cardiomyopathy. It has been proposed that a continual activation of AC(V) through elevated levels of protein kinase C could play an integral role in mediating a hypertrophic response leading to progressive heart failure. Elevated protein kinase C is a common finding in heart failure and was demonstrated in murine cardiomyopathy from cardiac-specific overexpression of G(αq) protein. Here we assessed whether the disruption of AC(V) expression can improve cardiac function, limit electrophysiological remodeling, or improve survival in the G(αq) mouse model of heart failure. We directly tested the effects of gene-targeted disruption of AC(V) in transgenic mice with cardiac-specific overexpression of G(αq) protein using multiple techniques to assess the survival, cardiac function, as well as structural and electrical remodeling. Surprisingly, in contrast to other models of cardiomyopathy, AC(V) disruption did not improve survival or cardiac function, limit cardiac chamber dilation, halt hypertrophy, or prevent electrical remodeling in G(αq) transgenic mice. In conclusion, unlike other established models of cardiomyopathy, disrupting AC(V) expression in the G(αq) mouse model is insufficient to overcome several parallel pathophysiological processes leading to progressive heart failure.


Assuntos
Adenilil Ciclases/metabolismo , Cardiomiopatias/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , Adenilil Ciclases/genética , Animais , Bradicardia/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Técnicas Eletrofisiológicas Cardíacas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Proteína Quinase C/metabolismo
4.
Cardiovasc Pathol ; 19(2): 83-93, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19217318

RESUMO

BACKGROUND: Murine cytomegalovirus (MCMV) is an etiologic agent of acute and chronic myocarditis in BALB/c mice. Immunologic host responses appear to play a key role in pathogenesis but have been incompletely defined. METHODS: BALB/c mice were infected with a sublethal dose of MCMV. Cytokine transcription and viral load (measured by quantitative real-time polymerase chain reaction) and histopathological analyses were performed at specified time points. RESULTS: Increased tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, and interferon (IFN)-gamma, as well as IL-10 mRNA transcripts, were detected in the hearts of infected mice starting at Day 1 post-infection (p.i.), with peak levels occurring at Day 8 p.i. (7-fold, 14-fold, 41-fold, and 16-fold higher than background, respectively). Peak cytokine transcription significantly correlated with a 10-fold increase in viral load (P<.001) at Day 8 p.i. Myocarditis-related pathological changes, measured by infiltration foci, were greatest at Day 8 p.i., corresponding with peak cytokine transcription and significantly correlated with IFN-gamma levels (P<.0001). Infiltration foci were predominantly composed of CD3(+) T cells. Cardiac calcification was observed in most infected mice predominantly over the right ventricle. Histological analysis of heart sections from mice infected with recombinant enhanced green fluorescence protein-MCMV revealed a localized and sporadic pattern of virus throughout all heart layers. CONCLUSIONS: MCMV-induced myocarditis in BALB/c mice is characterized by in vivo production of proinflammatory cytokines in a pattern correlating with MCMV viral load. The infection pattern and inflammatory response is highly localized, sporadic, and involves endocardium, epicardium, as well as the myocardium, with greatest amounts of virus detected in areas of pathologic calcification.


Assuntos
Citocinas/análise , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Miocardite/virologia , Carga Viral , Doença Aguda , Animais , Apoptose , Calcinose/imunologia , Calcinose/patologia , Calcinose/virologia , Citocinas/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , Microdissecção , Miocardite/imunologia , Miocardite/patologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia , RNA Mensageiro/metabolismo
5.
J Mol Cell Cardiol ; 47(6): 835-45, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19716829

RESUMO

Myocardial infarction (MI) leading to myocardial cell loss represents one of the common causes leading to cardiac failure. We have previously demonstrated the beneficial effects of several potent soluble epoxide hydrolase (sEH) inhibitors in cardiac hypertrophy. sEH catalizes the conversion of epoxyeicosatrienoic acids (EETs) to form the corresponding dihydroxyeicosatrienoic acids (DHETs). EETs are products of cytochrome P450 epoxygenases that have vasodilatory properties. Additionally, EETs inhibit the activation of nuclear factor (NF)-kappaB-mediated gene transcription. Motivated by the potential to uncover a new class of therapeutic agents for cardiovascular diseases which can be effectively used in clinical setting, we directly tested the biological effects of sEH inhibitors (sEHIs) on the progression of cardiac remodeling using a clinically relevant murine model of MI. We demonstrated that sEHIs were highly effective in the prevention of progressive cardiac remodeling post MI. Using metabolomic profiling of the inflammatory lipid mediators, we documented a significant decrease in EETs/DHETs ratio in MI model predicting a heightened inflammatory state. Treatment with sEHIs resulted in a change in the pattern of lipid mediators from one of inflammation towards resolution. Moreover, the oxylipin profiling showed a striking parallel to the changes in inflammatory cytokines in this model. Our study provides evidence for a possible new therapeutic strategy to improve cardiac function post MI.


Assuntos
Benzoatos/farmacologia , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Metabolômica , Infarto do Miocárdio/metabolismo , Ureia/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Arritmias Cardíacas/sangue , Arritmias Cardíacas/complicações , Arritmias Cardíacas/prevenção & controle , Cromatografia Líquida , Citocinas/sangue , Modelos Animais de Doenças , Testes de Função Cardíaca , Marcação In Situ das Extremidades Cortadas , Mediadores da Inflamação/sangue , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Miocárdio/patologia , Solubilidade , Espectrometria de Massas em Tandem , Ultrassonografia , Ureia/farmacologia , Remodelação Ventricular/efeitos dos fármacos
6.
J Physiol ; 587(Pt 5): 1087-100, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19139040

RESUMO

Small conductance Ca(2+)-activated K(+) channels (SK channels) have been reported in excitable cells, where they aid in integrating changes in intracellular Ca(2+) (Ca(2+)(i)) with membrane potential. We have recently reported the functional existence of SK2 channels in human and mouse cardiac myocytes. Moreover, we have found that the channel is predominantly expressed in atria compared to the ventricular myocytes. We hypothesize that knockout of SK2 channels may be sufficient to disrupt the intricate balance of the inward and outward currents during repolarization in atrial myocytes. We further predict that knockout of SK2 channels may predispose the atria to tachy-arrhythmias due to the fact that the late phase of the cardiac action potential is highly susceptible to aberrant excitation. We take advantage of a mouse model with genetic knockout of the SK2 channel gene. In vivo and in vitro electrophysiological studies were performed to probe the functional roles of SK2 channels in the heart. Whole-cell patch-clamp techniques show a significant prolongation of the action potential duration prominently in late cardiac repolarization in atrial myocytes from the heterozygous and homozygous null mutant animals. Moreover, in vivo electrophysiological recordings show inducible atrial fibrillation in the null mutant mice but not wild-type animals. No ventricular arrhythmias are detected in the null mutant mice or wild-type animals. In summary, our data support the important functional roles of SK2 channels in cardiac repolarization in atrial myocytes. Genetic knockout of the SK2 channels results in the delay in cardiac repolarization and atrial arrhythmias.


Assuntos
Potenciais de Ação/genética , Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Miócitos Cardíacos/patologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/deficiência , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
7.
Proc Natl Acad Sci U S A ; 104(48): 19108-13, 2007 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-18025474

RESUMO

The structural phenotype of neural connections in the auditory brainstem is sculpted by spontaneous and stimulus-induced neural activities during development. However, functional and molecular mechanisms of spontaneous action potentials (SAPs) in the developing cochlea are unknown. Additionally, it is unclear how regenerating hair cells establish their neural ranking in the constellation of neurons in the brainstem. We have demonstrated that a transient Ca(2+) current produced by the Ca(v)3.1 channel is expressed early in development to initiate spontaneous Ca(2+) spikes. Ca(v)1.3 currents, typical of mature hair cells, appeared later in development. Moreover, there is a surprising disappearance of the Ca(v)3.1 current that coincides with the attenuation of the transient Ca(2+) current as the electrical properties of hair cells transition to the mature phenotype. Remarkably, this process is recapitulated during hair-cell regeneration, suggesting that the transient expression of Ca(v)3.1 and the ensuing SAPs are signatures of hair cell development and regeneration.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo T/fisiologia , Cóclea/fisiologia , Células Ciliadas Auditivas/citologia , Regeneração , Potenciais de Ação/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Embrião de Galinha , Galinhas , Cóclea/efeitos dos fármacos , Cóclea/embriologia , Cóclea/crescimento & desenvolvimento , Gentamicinas/toxicidade , Células Ciliadas Auditivas/efeitos dos fármacos , Mibefradil/farmacologia , Níquel/farmacologia , Técnicas de Patch-Clamp , Venenos de Escorpião/farmacologia
8.
Proc Natl Acad Sci U S A ; 103(49): 18733-8, 2006 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-17130447

RESUMO

Sustained cardiac hypertrophy represents one of the most common causes leading to cardiac failure. There is emerging evidence to implicate the involvement of NF-kappaB in the development of cardiac hypertrophy. However, several critical questions remain unanswered. We tested the use of soluble epoxide hydrolase (sEH) inhibitors as a means to enhance the biological activities of epoxyeicosatrienoic acids (EETs) to treat cardiac hypertrophy. sEH catalyzes the conversion of EETs to form the corresponding dihydroxyeicosatrienoic acids. Previous data have suggested that EETs may inhibit the activation of NF-kappaB-mediated gene transcription. We directly demonstrate the beneficial effects of several potent sEH inhibitors (sEHIs) in cardiac hypertrophy. Specifically, we show that sEHIs can prevent the development of cardiac hypertrophy using a murine model of pressure-induced cardiac hypertrophy. In addition, sEHIs reverse the preestablished cardiac hypertrophy caused by chronic pressure overload. We further demonstrate that these compounds potently block the NF-kappaB activation in cardiac myocytes. Moreover, by using in vivo electrophysiologic recordings, our study shows a beneficial effect of the compounds in the prevention of cardiac arrhythmias that occur in association with cardiac hypertrophy. We conclude that the use of sEHIs to increase the level of the endogenous lipid epoxides such as EETs may represent a viable and completely unexplored avenue to reduce cardiac hypertrophy by blocking NF-kappaB activation.


Assuntos
Cardiomegalia/tratamento farmacológico , Cardiomegalia/prevenção & controle , Inibidores Enzimáticos/uso terapêutico , Epóxido Hidrolases/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Solubilidade
9.
J Mol Cell Cardiol ; 41(1): 170-81, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16750219

RESUMO

Adenylyl cyclase (AC) is an effector molecule in beta-adrenergic receptor signaling, catalyzing conversion of ATP to cAMP. Agents that increase intracellular levels of cAMP have been used previously to treat clinical heart failure. Recently, Roth et al. have shown that long-term cardiac-directed expression of ACVI, a dominant AC isoform in mammalian cardiac myocytes, increases survival and abrogates myocardial hypertrophy in transgenic (TG) mice with Galphaq-associated cardiomyopathy. Indeed, it has been proposed that increasing the cardiac content of ACVI is fundamentally different than other strategies used to increase cAMP function. However, one important but unexplored issue is its effects on electrical remodeling. Electrophysiological properties of Galphaq mice have been characterized. Similar to other models of cardiac hypertrophy and failure, cardiac myocytes isolated from Galphaq mice show prolonged action potential with reduced transient outward K+ current (Ito) and inward rectifier K+ current (IK1) density compare to wild-type (WT) animals. We directly examined the electrical remodeling of cardiac-directed ACVI over-expression in Galphaq mice using ECG recordings and whole-cell patch-clamp recordings. Four groups of animals were used: WT (double negative), ACVI, Galphaq and double positive TG mice (Galphaq/ACVI). Cardiac-directed expression of ACVI results in the reversal of adverse electrical remodeling in the Galphaq mice and is associated with significant improvement in the delay of cardiac repolarization and arrhythmias. Specifically, there is a normalization of Ito, IK1 and action potential duration in Galphaq/ACVI compared to Galphaq mice. In summary, our data provide evidence that increased cardiac ACVI content has a salutary effect in cardiomyopathy and cardiac electrical remodeling.


Assuntos
Adenilil Ciclases/metabolismo , Cardiomiopatias/fisiopatologia , Remodelação Ventricular , Adenilil Ciclases/genética , Animais , Cardiomegalia/genética , Cardiomegalia/prevenção & controle , Ecocardiografia , Estimulação Elétrica , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Canais de Potássio/metabolismo , Regulação para Cima
10.
Circulation ; 112(13): 1936-44, 2005 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-16172271

RESUMO

BACKGROUND: Previous data suggest that L-type Ca2+ channels containing the Cav1.3(alpha(1D)) subunit are expressed mainly in neurons and neuroendocrine cells, whereas those containing the Cav1.2(alpha1C) subunit are found in the brain, vascular smooth muscle, and cardiac tissue. However, our previous report as well as others have shown that Cav1.3 Ca2+ channel-deficient mice (Cav1.3(-/-)) demonstrate sinus bradycardia with a prolonged PR interval. In the present study, we extended our study to examine the role of the Cav1.3(alpha1D) Ca2+ channel in the atria of Cav1.3(-/-) mice. METHODS AND RESULTS: We obtained new evidence to demonstrate that there is significant expression of Cav1.3 Ca2+ channels predominantly in the atria compared with ventricular tissues. Whole-cell L-type Ca2+ currents (I(Ca,L)) recorded from single, isolated atrial myocytes from Cav1.3(-/-) mice showed a significant depolarizing shift in voltage-dependent activation. In contrast, there were no significant differences in the I(Ca,L) recorded from ventricular myocytes from wild-type and null mutant mice. We previously documented the hyperpolarizing shift in the voltage-dependent activation of Cav1.3 compared with Cav1.2 Ca2+ channel subunits in a heterologous expression system. The lack of Cav1.3 Ca2+ channels in null mutant mice would result in a depolarizing shift in the voltage-dependent activation of I(Ca,L) in atrial myocytes. In addition, the Cav1.3-null mutant mice showed evidence of atrial arrhythmias, with inducible atrial flutter and fibrillation. We further confirmed the isoform-specific differential expression of Cav1.3 versus Cav1.2 by in situ hybridization and immunofluorescence confocal microscopy. CONCLUSIONS: Using gene-targeted deletion of the Cav1.3 Ca2+ channel, we established the differential distribution of Cav1.3 Ca2+ channels in atrial myocytes compared with ventricles. Our data represent the first report demonstrating important functional roles for Cav1.3 Ca2+ channel in atrial tissues.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Miocárdio/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Eletrofisiologia , Imunofluorescência , Marcação de Genes , Átrios do Coração , Ventrículos do Coração , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Physiol Heart Circ Physiol ; 289(6): H2714-23, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16055520

RESUMO

Small-conductance Ca2+-activated K+ channels (SK channels, KCa channels) have been reported in excitable cells, where they aid in integrating changes in intracellular Ca2+ with membrane potential. We recently reported for the first time the functional existence of SK2 (KCa2.2) channels in human and mouse cardiac myocytes. Here, we report cloning of SK1 (KCa2.1) and SK3 (KCa2.3) channels from mouse atria and ventricles using RT-PCR. Full-length transcripts and their variants were detected for both SK1 and SK3 channels. Variants of mouse SK1 channel (mSK1) differ mainly in the COOH-terminal structure, affecting a portion of the sixth transmembrane segment (S6) and the calmodulin binding domain (CaMBD). Mouse SK3 channel (mSK3) differs not only in the number of polyglutamine repeats in the NH2 terminus but also in the intervening sequences between the polyglutamine repeats. Full-length cardiac mSK1 and mSK3 show 99 and 91% nucleotide identity with those of mouse colon SK1 and SK3, respectively. Quantification of SK1, SK2, and SK3 transcripts between atria and ventricles was performed using real-time quantitative RT-PCR from single, isolated cardiomyocytes. SK1 transcript was found to be more abundant in atria compared with ventricles, similar to the previously reported finding for SK2 channel. In contrast, SK3 showed similar levels of expression in atria and ventricles. Together, our data are the first to indicate the presence of the three different isoforms of SK channels in heart and the differential expression of SK1 and SK2 in mouse atria and ventricles. Because of the marked differential expression of SK channel isoforms in heart, specific ligands for Ca2+-activated K+ currents may offer a unique therapeutic opportunity to modify atrial cells without interfering with ventricular myocytes.


Assuntos
Regulação da Expressão Gênica/fisiologia , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Ventrículos do Coração/citologia , Masculino , Camundongos , Dados de Sequência Molecular , Canais de Potássio Cálcio-Ativados/química
13.
Pacing Clin Electrophysiol ; 28(6): 591-3, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15955196

RESUMO

INTRODUCTION: Postural orthostatic tachycardia syndrome (POTS) is a rare disease characterized by syncope, sinus tachycardia, and orthostasis due to autonomic dysfunction. METHODS AND RESULTS: Two women aged 26 and 24 years with severe POTS became pregnant. Both women experienced hyperemesis gravidarum with subsequent marked improvement in their POTS symptoms until 6 months gestation, when their syncope and sinus tachycardia caused clinical decompensation. Both patients delivered healthy babies at 37 weeks by elective cesarean section. CONCLUSION: In long-term follow-up, both women reported improvement in their prepartum symptoms. We describe the first report, to our knowledge, of two successful pregnancy outcomes in severe POTS, including the first report of midodrine use in pregnant women.


Assuntos
Tontura/terapia , Complicações Cardiovasculares na Gravidez/terapia , Síncope/terapia , Taquicardia/terapia , Adulto , Feminino , Humanos , Postura , Gravidez , Resultado da Gravidez , Síndrome
14.
J Physiol ; 562(Pt 3): 745-58, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15564282

RESUMO

We tested the hypothesis that chronic changes in intracellular Ca(2+) (Ca(2+)(i)) can result in changes in ion channel expression; this represents a novel mechanism of crosstalk between changes in Ca(2+) cycling proteins and the cardiac action potential (AP) profile. We used a transgenic mouse with cardiac-specific overexpression of sarcoplasmic reticulum Ca(2+) ATPase (SERCA) isoform 1a (SERCA1a OE) with a significant alteration of SERCA protein levels without cardiac hypertrophy or failure. Here, we report significant changes in the expression of a transient outward K(+) current (I(to,f)), a slowly inactivating K(+) current (I(K,slow)) and the steady state current (I(SS)) in the transgenic mice with resultant prolongation in cardiac action potential duration (APD) compared with the wild-type littermates. In addition, there was a significant prolongation of the QT interval on surface electrocardiograms in SERCA1a OE mice. The electrophysiological changes, which correlated with changes in Ca(2+)(i), were further corroborated by measuring the levels of ion channel protein expression. To recapitulate the in vivo experiments, the effects of changes in Ca(2+)(i) on ion channel expression were further tested in cultured adult and neonatal mouse cardiac myocytes. We conclude that a primary defect in Ca(2+) handling proteins without cardiac hypertrophy or failure may produce profound changes in K(+) channel expression and activity as well as cardiac AP.


Assuntos
Potenciais de Ação/fisiologia , ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Ativação do Canal Iônico/fisiologia , Camundongos Transgênicos/metabolismo , Células Musculares/fisiologia , Animais , ATPases Transportadoras de Cálcio/genética , Células Cultivadas , Eletrocardiografia , Ativação Enzimática , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Líquido Intracelular/metabolismo , Camundongos , Proteínas Recombinantes/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
15.
J Mol Cell Cardiol ; 37(1): 79-89, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15242738

RESUMO

Congenital long QT syndrome (LQTS) is a genetic disease that predisposes affected individuals to arrhythmias, syncope, and sudden death. Mutations in several ion channel genes have been discovered in different families with LQTS: KCNQ1 (KVLQT1, LQT1), KCNH2 (HERG, LQT2), SCN5A (LQT3), KCNE1 (minK, LQT5), and KCNE2 (MiRP1, LQT6). Previously, the P448R-KVLQT1 missense mutation has been reported as an LQT1-causing mutation. In this report, we demonstrate the presence of the P448R polymorphism in two, unrelated Chinese LQTS families. Although absent from 500 reference alleles derived from 150 white and 100 African-American subjects, P448R was present in 14% of healthy Chinese volunteers. Given the inconsistencies between the genotype (LQT1) and clinical phenotype (LQT2) in our two LQTS families, together with the finding that the P448R appears to be a common, ethnic-specific polymorphism, mutational analysis was extended to the other LQTS-causing genes resulting in the identification of distinct HERG missense mutations in each of these two families. Heterologous expression of P448R-KVLQT1 yielded normal, wild-type (WT) currents. In contrast, the two unique HERG mutations resulted in dominant-negative suppression of the WT HERG channel. Our study has profound implications for those engaged in genetic research. Importantly, one child of the original proband was initially diagnosed with LQT1 based upon the presence of P448R-KVLQT1 and was treated with beta-blockers. However, he did not possess the subsequently determined LQT2-causing mutation. On the other hand, his untreated P448R-negative brother harbored the true, disease-causing HERG mutation. These findings underscore the importance of distinguishing channel polymorphisms from mutations pathogenic for LQTS and emphasize the importance of using appropriate ethnically matched controls in the genotypic analysis of LQTS.


Assuntos
Testes Genéticos , Síndrome do QT Longo/genética , Mutação , Polimorfismo Genético , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Alelos , Povo Asiático , Sequência de Bases , População Negra , China , Análise Mutacional de DNA , Eletrofisiologia , Saúde da Família , Feminino , Genes Dominantes , Genótipo , Humanos , Íons , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Síndrome do QT Longo/etnologia , Masculino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Canais de Potássio/genética , Transfecção , População Branca
16.
J Biol Chem ; 278(49): 49085-94, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-13679367

RESUMO

The repolarization phase of cardiac action potential is prone to aberrant excitation that is common in cardiac patients. Here, we demonstrate that this phase is markedly sensitive to Ca2+ because of the surprising existence of a Ca2+-activated K+ currents in cardiac cells. The current was revealed using recording conditions that preserved endogenous Ca2+ buffers. The Ca2+-activated K+ current is expressed differentially in atria compared with ventricles. Because of the significant contribution of the current toward membrane repolarization in cardiac myocytes, alterations of the current magnitude precipitate abnormal action potential profiles. We confirmed the presence of a small conductance Ca2+-activated K+ channel subtype (SK2) in human and mouse cardiac myocytes using Western blot analysis and reverse transcription-polymerase chain reaction and have cloned SK2 channels from human atria, mouse atria, and ventricles. Because of the marked differential expression of SK2 channels in the heart, specific ligands for Ca2+-activated K+ currents may offer a unique therapeutic opportunity to modify atrial cells without interfering with ventricular myocytes.


Assuntos
Coração/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Sequência de Aminoácidos , Animais , Apamina/farmacologia , Sequência de Bases , Western Blotting , Clonagem Molecular , Primers do DNA , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Canais de Potássio Cálcio-Ativados/genética , Homologia de Sequência de Aminoácidos
17.
Circ Res ; 90(9): 981-7, 2002 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-12016264

RESUMO

We directly examined the role of the Ca(v)1.3 (alpha(1D)) Ca(2+) channel in the sinoatrial (SA) node by using Ca(v)1.3 Ca(2+) channel-deficient mice. A previous report has shown that the null mutant (Ca(v)1.3(-/-)) mice have sinus bradycardia with a prolonged PR interval. In the present study, we show that spontaneous action potentials recorded from the SA nodes show a significant decrease in the beating frequency and rate of diastolic depolarization in Ca(v)1.3(-/-) mice compared with their heterozygous (Ca(v)1.3(+/-)) or wild-type (WT, Ca(v)1.3(+/+)) littermates, suggesting that the deficit is intrinsic to the SA node. Whole-cell L-type Ca(2+) currents (I(Ca,L)s) recorded in single isolated SA node cells from Ca(v)1.3(-/-) mice show a significant depolarization shift in the activation threshold. The voltage-dependent activation of Ca(v)1.2 (alpha(1C)) versus Ca(v)1.3 Ca(2+) channel subunits was directly compared by using a heterologous expression system without beta coexpression. Similar to the I(Ca,L) recorded in the SA node of Ca(v)1.3(-/-) mutant mice, the Ca(v)1.2 Ca(2+) channel shows a depolarization shift in the voltage-dependent activation compared with that in the Ca(v)1.3 Ca(2+) channel. In summary, using gene-targeted deletion of the Ca(v)1.3 Ca(2+) channel, we were able to establish a role for Ca(v)1.3 Ca(2+) channels in the generation of the spontaneous action potential in SA node cells.


Assuntos
Canais de Cálcio/fisiologia , Nó Sinoatrial/fisiopatologia , Animais , Antiarrítmicos/farmacologia , Nó Atrioventricular/fisiologia , Nó Atrioventricular/fisiopatologia , Atropina/farmacologia , Bradicardia/fisiopatologia , Canais de Cálcio/genética , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/fisiologia , Linhagem Celular , Eletrocardiografia/efeitos dos fármacos , Genótipo , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Mutagênese , Mutação , Técnicas de Patch-Clamp , Propranolol/farmacologia , Nó Sinoatrial/citologia , Nó Sinoatrial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...