Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 594(21): 6175-6187, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27296897

RESUMO

KEY POINTS: Channelopathies of autoimmune origin are novel and are associated with corrected QT (QTc) prolongation and complex ventricular arrhythmias. We have recently demonstrated that anti-SSA/Ro antibodies from patients with autoimmune diseases and with QTc prolongation on the ECG target the human ether-à-go-go-related gene (HERG) K+ channel by inhibiting the corresponding current, IKr , at the pore region. Immunization of guinea-pigs with a peptide (E-pore peptide) corresponding to the extracellular loop region connecting the S5 and S6 segments of the HERG channel induces high titres of antibodies that inhibit IKr , lengthen the action potential and cause QTc prolongation on the surface ECG. In addition, anti-SSA/Ro-positive sera from patients with connective tissue diseases showed high reactivity to the E-pore peptide. The translational impact is the development of a peptide-based approach for the diagnosis and treatment of autoimmune-associated long QT syndrome. ABSTRACT: We recently demonstrated that anti-SSA/52 kDa Ro antibodies (Abs) from patients with autoimmune diseases and corrected QT (QTc) prolongation directly target and inhibit the human ether-à-go-go-related gene (HERG) K+ channel at the extracellular pore (E-pore) region, where homology with SSA/52 kDa Ro antigen was demonstrated. We tested the hypothesis that immunization of guinea-pigs with a peptide corresponding to the E-pore region (E-pore peptide) will generate pathogenic inhibitory Abs and cause QTc prolongation. Guinea-pigs were immunized with a 31-amino-acid peptide corresponding to the E-pore region of HERG. On days 10-62 after immunization, ECGs were recorded and blood was sampled for the detection of E-pore peptide Abs. Serum samples from patients with autoimmune diseases were evaluated for reactivity to E-pore peptide by enzyme-linked immunosorbent assay (ELISA), and histology was performed on hearts using Masson's Trichrome. Inhibition of the HERG channel was assessed by electrophysiology and by computational modelling of the human ventricular action potential. The ELISA results revealed the presence of high titres of E-pore peptide Abs and significant QTc prolongation after immunization. High reactivity to E-pore peptide was found using anti-SSA/Ro Ab-positive sera from patients with QTc prolongation. Histological data showed no evidence of fibrosis in immunized hearts. Simulations of simultaneous inhibition of repolarizing currents by anti-SSA/Ro Ab-positive sera showed the predominance of the HERG channel in controlling action potential duration and the QT interval. These results are the first to demonstrate that inhibitory Abs to the HERG E-pore region induce QTc prolongation in immunized guinea-pigs by targeting the HERG channel independently from fibrosis. The reactivity of anti-SSA/Ro Ab-positive sera from patients with connective tissue diseases with the E-pore peptide opens novel pharmacotherapeutic avenues in the diagnosis and management of autoimmune-associated QTc prolongation.


Assuntos
Autoimunidade , Canais de Potássio Éter-A-Go-Go/imunologia , Síndrome do QT Longo/imunologia , Animais , Anticorpos/imunologia , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/química , Cobaias , Células HEK293 , Humanos , Fragmentos de Peptídeos/imunologia
2.
Circulation ; 132(4): 230-40, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25995318

RESUMO

BACKGROUND: Emerging clinical evidence demonstrates high prevalence of QTc prolongation and complex ventricular arrhythmias in patients with anti-Ro antibody (anti-Ro Ab)-positive autoimmune diseases. We tested the hypothesis that anti-Ro Abs target the HERG (human ether-a-go-go-related gene) K(+) channel, which conducts the rapidly activating delayed K(+) current, IKr, thereby causing delayed repolarization seen as QT interval prolongation on the ECG. METHODS AND RESULTS: Anti-Ro Ab-positive sera, purified IgG, and affinity-purified anti-52kDa Ro Abs from patients with autoimmune diseases and QTc prolongation were tested on IKr using HEK293 cells expressing HERG channel and native cardiac myocytes. Electrophysiological and biochemical data demonstrate that anti-Ro Abs inhibit IKr to prolong action potential duration by directly binding to the HERG channel protein. The 52-kDa Ro antigen-immunized guinea pigs showed QTc prolongation on ECG after developing high titers of anti-Ro Abs, which inhibited native IKr and cross-reacted with guinea pig ERG channel. CONCLUSIONS: The data establish that anti-Ro Abs from patients with autoimmune diseases inhibit IKr by cross-reacting with the HERG channel likely at the pore region where homology between anti-52-kDa Ro antigen and HERG channel is present. The animal model of autoimmune-associated QTc prolongation is the first to provide strong evidence for a pathogenic role of anti-Ro Abs in the development of QTc prolongation. It is proposed that adult patients with anti-Ro Abs may benefit from routine ECG screening and that those with QTc prolongation should receive counseling about drugs that may increase the risk for life-threatening arrhythmias.


Assuntos
Anticorpos Anti-Idiotípicos/fisiologia , Doenças Autoimunes/etiologia , Doenças Autoimunes/fisiopatologia , Síndrome do QT Longo/etiologia , Síndrome do QT Longo/fisiopatologia , Ribonucleoproteínas/imunologia , Adulto , Idoso , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/farmacologia , Arritmias Cardíacas/epidemiologia , Arritmias Cardíacas/fisiopatologia , Doenças Autoimunes/imunologia , Células Cultivadas , Modelos Animais de Doenças , Canal de Potássio ERG1 , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Cobaias , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Síndrome do QT Longo/imunologia , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fatores de Risco
3.
Biochem Biophys Res Commun ; 426(2): 237-41, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22935420

RESUMO

UNLABELLED: Pervious biochemical and hemodynamic studies have highlighted the important role of εPKC in cardioprotection during ischemic preconditioning. However, little is known about the electrophysiological consequences of εPKC modulation in ischemic hearts. Membrane permeable peptide εPKC selective activator and inhibitor were used to investigate the role of εPKC modulation in reperfusion arrhythmias. METHODS: Protein transduction domain from HIV-TAT was used as a carrier for peptide delivery into intact Langendorff perfused guinea pig hearts. Action potentials were imaged and mapped (124 sites) using optical techniques and surface ECG was continuously recorded. Hearts were exposed to 30 min stabilization period, 15 min of no-flow ischemia, followed by 20 min reperfusion. Peptides (0.5 µM) were infused as follows: (a) control (vehicle-TAT peptide; TAT-scrambled ψεRACK peptide); (b) εPKC agonist (TAT-ψεRACK); (c) εPKC antagonist (TAT-εV1). RESULTS: Hearts treated with εPKC agonist ψεRACK had reduced incidence of ventricular tachycardia (VT, 64%) and fibrillation (VF, 50%) compared to control (VT, 80%, P<0.05) and (VF, 70%, P < 0.05). However, the highest incidence of VT (100%, P < 0.05) and VF (80%) occurred in hearts treated with εPKC antagonist peptide εV1 compared to control and to εPKC agonist ψεRACK. Interestingly, at 20 min reperfusion, 100% of hearts treated with εPKC agonist ψεRACK exhibited complete recovery of action potentials compared to 40% (P < 0.05) of hearts treated with εPKC antagonist peptide, εV1 and 65% (P < 0.5) of hearts in control. At 20 min reperfusion, maps of action potential duration from εPKC agonist ψεRACK showed minimal dispersion (48.2 ± 9 ms) compared to exacerbated dispersion (115.4 ± 42 ms, P < 0.05) in εPKC antagonist and control (67 ± 20 ms, P<0.05). VT/VF and dispersion from hearts treated with scrambled agonist or antagonist peptides were similar to control. CONCLUSION: The results demonstrate that εPKC activation by ψεRACK peptide protects intact hearts from reperfusion arrhythmias and affords better recovery. On the other hand, inhibition of εPKC increased the incidence of arrhythmias and worsened recovery compared to controls. The results carry significant therapeutic implications for the treatment of acute ischemic heart disease by preconditioning-mimicking agents.


Assuntos
Arritmias Cardíacas/enzimologia , Traumatismo por Reperfusão Miocárdica/complicações , Proteína Quinase C-épsilon/biossíntese , Animais , Arritmias Cardíacas/etiologia , Ativação Enzimática , Cobaias , Técnicas In Vitro
4.
Pediatr Res ; 69(6): 479-84, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21378599

RESUMO

The novel Cav1.3 (α1D) L-type Ca²âº channel plays a significant role in sinoatrial (SA) and atrioventricular (AV) nodes function and in atrial fibrillation. However, the characterization of α1D Ca²âº channel during heart development is very limited. We used real-time RT-PCR, Western blotting, and indirect immunostaining to characterize the developmental expression and localization of α1D Ca²âº channel in rat hearts. Both protein and mRNA levels of α1D Ca²âº channel decreased postnatally. Two forms of α1D Ca²âº channel protein (250 and 190 kD) were observed, with the full-length (250 kD) channel protein being predominant in the prenatal stages. Both Western blots and confocal imaging demonstrated that α1D Ca²âº channel protein was expressed in both atria and ventricles at fetal and neonatal stages but was absent in the adult ventricles. Interestingly, α1D Ca²âº channel was also found at the nucleus/perinucleus of immature but not adult atrial cells. Furthermore, the nuclear staining was reproduced in adult atrial cell line, HL-1 cells, which possess immature properties. The data are first to show that α1D Ca²âº channel has unique age-dependent expression profile and subcellular localization in the heart, suggesting a developmental stage-dependent specific function.


Assuntos
Canais de Cálcio/metabolismo , Átrios do Coração/metabolismo , Coração/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Canais de Cálcio/genética , Células Cultivadas , Coração/fisiologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
5.
J Autoimmun ; 34(2): 80-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19640679

RESUMO

Congenital heart block (CHB) is an autoimmune disease associated with autoantibodies against intracellular ribonucleoproteins SSB/La and SSA/Ro. The hallmark of CHB is complete atrioventricular block. We have recently established that anti-SSA/Ro -SSB/La autoantibodies inhibit alpha(1D) L-type Ca current, I(Ca-L), and cross-react with the alpha(1D) Ca channel protein. This study aims at identifying the possible binding sites on alpha(1D) protein for autoantibodies from sera of mothers with CHB children. GST fusion proteins of the extracellular regions between the transmembrane segments (S5-S6) of each of the four alpha(1D) Ca channel protein domains I-IV were prepared and tested for reactivity with sera from mothers with CHB children and controls using ELISA. Sera containing anti-Ro/La autoantibodies from 118 mothers with CHB children and from 15 mothers with anti-Ro/La autoantibodies but have healthy children, and from 28 healthy mothers without anti-Ro/La autoantibodies and healthy children were evaluated. Seventeen of 118 (14.4%) sera from mothers with CHB children reacted with the extracellular loop of domain I S5-S6 region (E1). In contrast, only 2 of 28 (7%) of sera from healthy mothers (-anti-Ro/La) and healthy children reacted with E1 loop and none (0 of 15) of sera from healthy mothers (+anti-Ro/La) and healthy children reacted with the E1 loop. Preincubation of E1 loop with the positive sera decreased the O.D reading establishing the specificity of the response. Electrophysiological characterization of the ELISA positive sera and purified IgG showed inhibition (44.1% and 49.8%, respectively) of the alpha(1D) I(Ca-L) expressed in tsA201 cells. The inhibition was abolished when the sera were pre-incubated with E1 fusion protein. The results identified the extracellular loop of domain I S5-S6 of L-type Ca channel alpha(1D) subunit as a target for autoantibodies from a subset of mothers with CHB children. This novel finding provides insights into the potential development of therapeutic peptides that could bind to the pathogenic antibodies and prevent CHB.


Assuntos
Autoanticorpos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Bloqueio Cardíaco/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Autoanticorpos/sangue , Autoantígenos/imunologia , Bloqueadores dos Canais de Cálcio/sangue , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/imunologia , Desenho de Fármacos , Mapeamento de Epitopos , Feminino , Bloqueio Cardíaco/sangue , Bloqueio Cardíaco/congênito , Bloqueio Cardíaco/prevenção & controle , Humanos , Gravidez , Conformação Proteica , Engenharia de Proteínas , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Ribonucleoproteínas/imunologia , Antígeno SS-B
6.
Biochem Biophys Res Commun ; 384(4): 409-14, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19422800

RESUMO

Cav1.2 (alpha1C) and Cav1.3 (alpha1D) L-type Ca channels are co-expressed in the heart. To date, there are no pharmacological or biophysical tools to separate alpha1D from alpha1C Ca currents (I(Ca-L)) in cardiomyocytes. Here, we established a physiological model to study alpha1D I(Ca-L) in native myocytes using RNA interference. Transfection of rat neonatal cardiomyocytes (RNC) with alpha1C specific siRNA resulted in low silencing efficiency (50-60%) at the mRNA and protein levels. The use of lentivirus shRNA resulted in 100% transfection efficiency and 92% silencing of the alpha1C gene by real-time PCR and Western blot. Electrophysiological experiments showed that the total I(Ca-L) was similarly reduced by 80% in lentivirus transfected cells. Both biochemical and functional data demonstrated high transfection and silencing efficiency in the cardiomyocytes using lentiviral shRNA. This novel approach allows for the assessments of the roles of alpha1C and alpha1D Ca channels in native myocytes and could be used to examine their roles in physiological and pathological settings.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Canais de Cálcio/genética , Canais de Cálcio Tipo L/genética , Lentivirus/genética , Modelos Cardiovasculares , Miócitos Cardíacos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Transfecção
7.
Circulation ; 111(23): 3034-41, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15939813

RESUMO

BACKGROUND: Congenital heart block (CHB) is an autoimmune disease that affects fetuses/infants born to mothers with anti-Ro/La antibodies (positive IgG). Although the hallmark of CHB is complete atrioventricular block, sinus bradycardia has been reported recently in animal models of CHB. Interestingly, knockout of the neuroendocrine alpha1D Ca channel in mice results in significant sinus bradycardia and atrioventricular block, a phenotype reminiscent to that seen in CHB. Here, we tested the hypothesis that the alpha1D Ca channel is a novel target for positive IgG. METHODS AND RESULTS: Reverse transcription-polymerase chain reaction, confocal indirect immunostaining, and Western blot data established the expression of the alpha1D Ca channel in the human fetal heart. The effect of positive IgG on alpha1D Ca current (I(Ca-L)) was characterized in heterologous expression systems (tsA201 cells and Xenopus oocytes) because of the unavailability of alpha1D-specific modulators. alpha1D I(Ca-L) activated at negative potentials (between -60 and -50 mV). Positive IgG inhibited alpha1D I(Ca-L) in both expression systems. This inhibition was rescued by a Ca channel activator, Bay K8644. No effect on alpha1D I(Ca-L) was observed with negative IgG and denatured positive IgG. Western blot data showed that positive IgG binds directly to alpha1D Ca channel protein. CONCLUSIONS: The data are the first to demonstrate (1) expression of the alpha1D Ca channel in human fetal heart, (2) inhibition of alpha1D I(Ca-L) by positive IgG, and (3) direct cross-reactivity of positive IgG with the alpha1D Ca channel protein. Given that alpha1D I(Ca-L) activates at voltages within the pacemaker's diastolic depolarization, inhibition of alpha1D I(Ca-L) in part may account for autoimmune-associated sinus bradycardia. In addition, Bay K8644 rescue of alpha1D I(Ca-L) inhibition opens new directions in the development of pharmacotherapeutic approaches in the management of CHB.


Assuntos
Doenças Autoimunes/complicações , Bradicardia/etiologia , Canais de Cálcio Tipo L/fisiologia , Animais , Autoanticorpos , Bradicardia/imunologia , Cálcio/metabolismo , Canais de Cálcio Tipo L/análise , Canais de Cálcio Tipo L/genética , Linhagem Celular , Eletrofisiologia , Feto/citologia , Coração/fisiopatologia , Humanos , Imunoglobulina G/imunologia , Miócitos Cardíacos/química , Miócitos Cardíacos/fisiologia , RNA Mensageiro/análise , Coelhos , Nó Sinoatrial/fisiopatologia , Transdução Genética
8.
Circ Res ; 94(4): e32-8, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-14963005

RESUMO

Congenital heart block (CHB) is a conduction abnormality characterized by complete atrioventricular (AV) block. CHB affects fetuses and/or newborn of mothers with autoantibodies reactive with ribonucleoproteins 48-kDa SSB/La, 52-kDa SSA/Ro, and 60-kDa SSA/Ro. We recently established animal models of CHB and reported, for the first time, significant sinus bradycardia preceding AV block. This unexpected observation implies that the spectrum of conduction abnormalities extends beyond the AV node to also affect the SA node. To test this hypothesis, we investigated the functional basis of this sinus bradycardia by characterizing the effects of antibodies from mothers with CHB children (positive IgG) on ionic currents that are known to significantly contribute to spontaneous pacing in SA node cells. We recorded L- (I(Ca.L)) and T- (I(Ca.T)) type Ca2+, delayed rectifier K+ (I(K)), hyperpolarization-activated (I(f)) currents, and action potentials (APs) from young rabbit SA node cells. We demonstrated that positive IgG significantly inhibited both I(Ca.T) and I(Ca.L) and induced sinus bradycardia but did not affect I(f) and I(K). Normal IgG from mothers with healthy children did not affect all the currents studied and APs. These results establish that IgG from mothers with CHB children causes substantial inhibition of I(Ca.T) and I(Ca.L), two important pacemaker currents in rabbit SA node cells and point to both I(Ca.T) and I(Ca.L) as major players in the ionic mechanism by which maternal antibodies induce sinus bradycardia in CHB. These novel findings have important clinical significance and suggest that sinus bradycardia may be a potential marker in the detection and prevention of CHB. The full text of this article is available online at http://circres.ahajournals.org


Assuntos
Anticorpos Antinucleares/farmacologia , Arritmia Sinusal/fisiopatologia , Autoantígenos , Bradicardia/fisiopatologia , Bloqueio Cardíaco/fisiopatologia , Imunoglobulina G/farmacologia , RNA Citoplasmático Pequeno , Nó Sinoatrial/fisiopatologia , Potenciais de Ação , Adulto , Animais , Anticorpos Antinucleares/imunologia , Arritmia Sinusal/etiologia , Arritmia Sinusal/imunologia , Bradicardia/etiologia , Bradicardia/imunologia , Cálcio/metabolismo , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/fisiologia , Feminino , Bloqueio Cardíaco/complicações , Bloqueio Cardíaco/congênito , Bloqueio Cardíaco/imunologia , Humanos , Imunidade Materno-Adquirida , Imunoglobulina G/imunologia , Transporte de Íons/efeitos dos fármacos , Gravidez , Coelhos , Ribonucleoproteínas/imunologia , Nó Sinoatrial/patologia , Antígeno SS-B
9.
Circ Arrhythm Electrophysiol ; 9(4): e003419, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27030700

RESUMO

BACKGROUND: In patients with autoimmune disease, anti-Ro/SSA antibodies (anti-Ro/SSA) are responsible for a novel autoimmune-associated long-QT syndrome by targeting the hERG potassium channel and inhibiting the related current (IKr). Because anti-Ro/SSA are also present in a significant proportion of healthy subjects and may be associated with torsades de pointes (TdP) arrhythmia, we tested the hypothesis that anti-Ro/SSA may represent a silent risk factor in patients developing TdP. METHODS AND RESULTS: Twenty-five consecutive patients who experienced TdP were prospectively collected independent of ongoing therapies and concomitant diseases. Anti-Ro/SSA were detected by fluoroenzyme immunoassay, immuno-Western blotting, and line-blot immunoassay. Purified IgGs from anti-Ro/SSA-positive and anti-Ro/SSA-negative patients were tested on IKr using HEK293 cells stably expressing the hERG channel. As expected, in TdP patients, many known corrected QT interval-prolonging risk factors were simultaneously present, including hypokalemia that was the most common (52%). Anti-Ro/SSA were present in 60% of the subjects, mostly the anti-Ro/SSA-52-kD subtype detected by immuno-Western blotting only. A history of autoimmune disease was found in only 2 of anti-Ro/SSA-positive patients. Experimental data demonstrated that purified anti-Ro/SSA-positive IgGs significantly inhibited IKr and cross reacted with hERG-channel proteins. Moreover, anti-Ro/SSA-positive sera exhibited high reactivity with a peptide corresponding to the hERG-channel pore-forming region. CONCLUSIONS: Anti-Ro/SSA may represent a clinically silent novel risk factor for TdP development via an autoimmune-mediated electrophysiological interference with the hERG channel. We propose that TdP patients may benefit from specific anti-Ro/SSA testing even in the absence of autoimmune diseases as immunomodulating therapies may be effective in shortening corrected QT interval and reducing TdP recurrence risk.


Assuntos
Anticorpos Antinucleares/imunologia , Autoimunidade , Eletrocardiografia , Torsades de Pointes/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antinucleares/sangue , Western Blotting , Canal de Potássio ERG1 , Ensaio de Imunoadsorção Enzimática , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Seguimentos , Células HEK293/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Torsades de Pointes/sangue , Torsades de Pointes/fisiopatologia
10.
Cardiovasc Res ; 55(3): 681-9, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12160965

RESUMO

OBJECTIVES: Ischemic preconditioning (PC) reduces myocardial infarction by a mechanism that involves opening of mitochondrial ATP-dependent potassium channels (mK(ATP)), reactive oxygen species (ROS), and possibly activation of p38 mitogen-activated protein kinase (p38 MAPK). The actual order of these steps, however, is a matter of current debate. This study examined whether protection afforded by menadione, which protects by causing mitochondria to produce ROS, requires mK(ATP) opening. In addition, we tested whether protection from anisomycin, a p38 MAPK activator, is dependent on ROS production. METHODS AND RESULTS: Isolated, buffer-perfused rat hearts were pretreated with menadione, and infarction was assessed after 30 min of regional ischemia and 120 min of reperfusion. Menadione reduced infarction in a dose-dependent manner with an EC(50) of 270 nM. Menadione's infarct-limiting effect was insensitive to 200 microM 5-hydroxydecanoate (5HD), an mK(ATP) channel blocker, whereas protection by diazoxide and PC were blocked by 5HD. Anisomycin caused hearts to resist infarction and this protective effect was abrogated by SB203580, a p38 MAPK inhibitor, and 2-mercaptopropionylglycine (MPG), a free radical scavenger. CONCLUSIONS: These results indicate that mK(ATP) opening occurs upstream of mitochondrial ROS generation in the protective pathway. Furthermore, protection afforded by anisomycin was p38 MAPK- and ROS-dependent.


Assuntos
Precondicionamento Isquêmico Miocárdico/métodos , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/prevenção & controle , Canais de Potássio/metabolismo , Transdução de Sinais/fisiologia , Animais , Anisomicina/farmacologia , Ácidos Decanoicos/farmacologia , Diazóxido/farmacologia , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Radicais Livres/metabolismo , Hidroxiácidos/farmacologia , Imidazóis/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infarto do Miocárdio/metabolismo , Perfusão , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia , Ratos , Ratos Wistar , Tiopronina/farmacologia , Vitamina K 3/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
11.
J Huazhong Univ Sci Technolog Med Sci ; 24(2): 132-3, 137, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15315161

RESUMO

This study evaluated the effects of adenovirus vector mediated human vascular endothelial growth factor-165 (hVEGF165) gene on prevention of restenosis after angioplasty. Rabbit models of bilateral carotid artery injury were established by balloon denudation. The recombinant adenoviruses containing hVEGF165 cDNA was directly injected into left side of the injured carotid arteries. On day 3 and week 3 after transfection the expression of VEGF was observed by RT-PCR and immunohistochemistry. The thrombokinesis, reendothelialization (rET) and intimal hyperplasia in carotid arteries were evaluated by computerized image analysis system 3 weeks after gene transfer. The changes in the VEGF gene-treated side were compared with the control side. Our results showed that 3 days and 3 weeks after hVEGF165 gene transfer the VEGF mRNA and antigen expression were detected in vivo. 3 weeks after the transfer, the carotid artery rET was markedly better in the VEGF gene-treated group compared with the control. The thrombokinesis, intima area/media area (I/M), maximal intimal and medial thicknesses (ITmax and MTmax) demonstrated a statistically significant decrease in arteries treated with VEGF gene as compared with the control group. It is concluded that VEGF gene transfer could be achieved by intra-arterial injection of recombinant adenoviruses. It might accelerate the restoration of endothelial integrity, inhibit thrombokinesis and attenuate intimal hyperplasia in the injured arteries after VEGF gene transfer. This procedure could be useful in preventing restenosis after angioplasty.


Assuntos
Angioplastia com Balão/efeitos adversos , Estenose das Carótidas/prevenção & controle , Endotélio Vascular/lesões , Terapia Genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Lesões das Artérias Carótidas/patologia , Estenose das Carótidas/fisiopatologia , Divisão Celular/efeitos dos fármacos , Endotélio Vascular/patologia , Hiperplasia/prevenção & controle , Masculino , Músculo Liso Vascular/citologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Coelhos , Recombinação Genética , Transfecção/métodos , Fator A de Crescimento do Endotélio Vascular/genética
12.
Heart Rhythm ; 6(9): 1359-65, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19716092

RESUMO

BACKGROUND: Abnormal intracellular calcium (Ca(i)) kinetics during ischemia/reperfusion (I/R) can alter membrane voltage (V(m)) and destabilize wavefront propagation. OBJECTIVE: We used optical mapping to investigate the hypothesis that early V(m)/Ca(i) uncoupling during a ventricular tachyarrhythmia (VT) can play a primary role in perpetuation of VT episodes. METHODS: Seventeen Langendorff-perfused guinea pig hearts were subjected to 15 min I/15 min R. Simultaneous optical recordings of V(m) and Ca(i) signals were obtained using a dual-photodiode array. Spatiotemporal entropy (E) was used to quantify differences in V(m)/Ca(i) kinetics during VT and compare wavefront topology during the first 500 ms of a VT episode. RESULTS: A total of 39 episodes of VT were analyzed; VT was classified as self-terminating (ST, n = 28) and non-self-terminating (NST, n = 11). The ST/VTs were further classified into short ST/VT (1 to 5 s in duration; n = 16) and long ST/VT (>5 s, n = 12). E values for NST/VTs were significantly higher than E values for both short and long ST/VTs separately as well as E values for ST/VTs as a group. Further, E values for long ST/VTs were significantly higher than E values for short ST/VTs. Wave breaks were consistently identified during periods of high E. CONCLUSION: High E during the first 500 ms of the onset of VT (the first 2 to 3 beats) is significantly correlated with long ST or NST episodes. This may be related to destabilization of wave propagation that helps to perpetuate VT. Early V(m)/Ca(i) uncoupling can predestinate the development of a malignant NST/VT.


Assuntos
Canais de Cálcio , Isquemia Miocárdica , Reperfusão Miocárdica/efeitos adversos , Taquicardia Ventricular/fisiopatologia , Animais , Sinalização do Cálcio , Análise por Conglomerados , Eletrofisiologia , Feminino , Cobaias , Comportamento de Redução do Risco , Fatores de Tempo
13.
Am J Physiol Heart Circ Physiol ; 295(5): H2017-24, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18790836

RESUMO

The novel alpha1D Ca2+ channel together with alpha1C Ca2+ channel contribute to the L-type Ca2+ current (I(Ca-L)) in the mouse supraventricular tissue. However, its functional role in the heart is just emerging. We used the alpha1D gene knockout (KO) mouse to investigate the electrophysiological features, the relative contribution of the alpha1D Ca2+ channel to the global I(Ca-L), the intracellular Ca2+ transient, the Ca2+ handling by the sarcoplasmic reticulum (SR), and the inducibility of atrial fibrillation (AF). In vivo and ex vivo ECG recordings from alpha1D KO mice demonstrated significant sinus bradycardia, atrioventricular block, and vulnerability to AF. The wild-type mice showed no ECG abnormalities and no AF. Patch-clamp recordings from isolated alpha1D KO atrial myocytes revealed a significant reduction of I(Ca-L) (24.5%; P < 0.05). However, there were no changes in other currents such as I(Na), I(Ca-T), I(K), I(f), and I(to) and no changes in alpha1C mRNA levels of alpha1D KO atria. Fura 2-loaded atrial myocytes showed reduced intracellular Ca2+ transient (approximately 40%; P < 0.05) and rapid caffeine application caused a 17% reduction of the SR Ca2+ content (P < 0.05) and a 28% reduction (P < 0.05) of fractional SR Ca2+ release in alpha1D KO atria. In conclusion, genetic deletion of alpha1D Ca2+ channel in mice results in atrial electrocardiographic abnormalities and AF vulnerability. The electrical abnormalities in the alpha1D KO mice were associated with a decrease in the total I(Ca-L) density, a reduction in intracellular Ca2+ transient, and impaired intracellular Ca2+ handling. These findings provide new insights into the mechanism leading to atrial electrical dysfunction in the alpha1D KO mice.


Assuntos
Fibrilação Atrial/metabolismo , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Miócitos Cardíacos/metabolismo , Potenciais de Ação , Animais , Fibrilação Atrial/genética , Bloqueio Atrioventricular/metabolismo , Bradicardia/metabolismo , Cafeína/farmacologia , Canais de Cálcio Tipo L/deficiência , Canais de Cálcio Tipo L/genética , Sinalização do Cálcio/efeitos dos fármacos , Eletrocardiografia , Átrios do Coração/metabolismo , Homeostase , Camundongos , Camundongos Knockout , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
14.
J Pharmacol Exp Ther ; 321(2): 517-25, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17322024

RESUMO

The recent discovery of zinc signals and their essential role in the redox signaling network implies that zinc homeostasis and the function of zinc-containing proteins are probably altered as a result of oxidative stress, suggesting new targets for pharmacological intervention. We hypothesized that the level of intracellular labile zinc is changed in hearts subjected to ischemia/reperfusion (I/R) and investigated whether the maintenance of myocardial zinc status protected heart functions. Using fluorescent imaging, we demonstrated decreased levels of labile zinc in the I/R hearts. Phorbol 12-myristate 13-acetate, a known trigger of zinc release, liberated zinc ions in control hearts but failed to produce any increase in zinc levels in the I/R rat hearts. Adding the zinc ionophore pyrithione at reperfusion improved myocardial recovery up to 100% and reduced the incidence of arrhythmias more than 2-fold. This effect was dose-dependent, and high concentrations of zinc were toxic. Adding membrane-impermeable zinc chloride was ineffective. Hearts from rats receiving zinc pyrithione supplements in their diet fully recovered from I/R. The recovery was associated with the prevention of degradation of the two protein kinase C isoforms, delta and epsilon, during I/R. In conclusion, our results suggest a protective role of intracellular zinc in myocardial recovery from oxidative stress imposed by I/R. The data support the potential clinical use of zinc ionophores in the settings of acute redox stress in the heart.


Assuntos
Traumatismo por Reperfusão Miocárdica/prevenção & controle , Proteína Quinase C-delta/fisiologia , Proteína Quinase C-épsilon/fisiologia , Zinco/fisiologia , Animais , Masculino , Metalotioneína/biossíntese , Miocárdio/química , Ratos , Ratos Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacologia , Zinco/análise
15.
Biochem Biophys Res Commun ; 349(1): 432-8, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16945341

RESUMO

PURPOSE: Ischemic heart disease carries an increased risk of malignant ventricular tachycardia (VT), fibrillation (VF), and sudden cardiac death. Protein kinase C (PKC) epsilon activation has been shown to improve the hemodynamics in hearts subjected to ischemia/reperfusion. However, very little is known about the role of epsilon PKC in reperfusion arrhythmias. Here we show that epsilon PKC activation is anti-arrhythmic and its inhibition is pro-arrhythmic. METHOD: Langendorff-perfused isolated hearts from epsilonPKC agonist (epsilonPKC activation), antagonist (epsilonPKC inhibition) transgenic (TG), and wild-type control mice were subjected to 30 min stabilization period, 10 min global ischemia, and 30 min reperfusion. Action potentials (APs) and calcium transients (CaiT) were recorded simultaneously at 37 degrees C using optical mapping techniques. The incidence of VT and VF was assessed during reperfusion. RESULTS: No VT/VF was seen in any group during the stabilization period in which hearts were perfused with Tyrode's solution. Upon reperfusion, 3 out of the 16 (19%) wild-type mice developed VT but no VF. In epsilonPKC antagonist group, in which epsilonPKC activity was downregulated, 10 out of 13 (76.9%) TG mice developed VT, of which six (46.2%) degenerated into sustained VF upon reperfusion. Interestingly, in epsilonPKC agonist mice, in which the activity of epsilonPKC was upregulated, no VF was observed and only 1 out of 12 mice showed only transient VT during reperfusion. During ischemia and reperfusion, CaiT decay was exceedingly slower in the antagonist mice compared to the other two groups. CONCLUSION: Moderate in vivo activation of epsilonPKC exerts beneficial antiarrhythmic effect vis-a-vis the lethal reperfusion arrhythmias. Abnormal CaiT decay may, in part, contribute to the high incidence of reperfusion arrhythmias in the antagonist mice. These findings have important implications for the development of PKC isozyme targeted therapeutics and subsequently for the treatment of ischemic heart diseases.


Assuntos
Arritmias Cardíacas/patologia , Proteína Quinase C-épsilon/metabolismo , Traumatismo por Reperfusão , Potenciais de Ação , Animais , Ativação Enzimática , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Perfusão , Proteína Quinase C-épsilon/antagonistas & inibidores , Fatores de Tempo
16.
Am J Physiol Heart Circ Physiol ; 288(5): H2123-30, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15615842

RESUMO

Alpha1D L-type Ca channel was assumed to be of neuroendocrine origin only; however, alpha1D L-type Ca channel knockout mice exhibit sinus bradycardia and atrioventricular block, indicating a distinct role of alpha1D in the heart. The presence and distribution of alpha1D Ca channel in the heart and its regulation by protein kinase A (PKA) are just emerging. Our objective was to examine the localization of alpha1D L-type Ca channel in rabbit and rat hearts and its modulation by PKA. Here, we show the exclusive presence of alpha1D Ca channel transcript in the sinoatrial node, atrioventricular node, and atria but not in the ventricle by RT-PCR and the expression of alpha1D Ca channel protein in atrial myocytes' sarcolemma by indirect immunostaining and Western blot. There is no significant difference in the expression level of alpha1D Ca channel in the left versus right atrium. Superfusion of membrane-permeable 8-bromo-cAMP resulted in a significant increase of the peak current density of alpha1D Ca current expressed in tsA201 cells. This increase was inhibited by the PKA inhibitor (PKI). Application of 8-bromo-cAMP also readily phosphorylated the alpha1D Ca channel protein. The results are first to demonstrate that PKA phosphorylation of L-type Ca channel alpha1D-subunit resulted in an increase of the alpha1D Ca channel activity. Together with the observation that alpha1D Ca channel is exclusively present in the sinoatrial node and atria, the findings suggest that alpha1D Ca channel plays a unique role in the sinoatrial tissue and is a target for sympathetic control of heart rhythm.


Assuntos
Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nó Sinoatrial/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , AMP Cíclico/metabolismo , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Fosforilação , RNA Mensageiro/análise , Coelhos , Sistema Nervoso Simpático/fisiologia
17.
Biochem Biophys Res Commun ; 314(3): 749-54, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14741699

RESUMO

It is well established that beta-adrenoceptor stimulation activates PKA and alpha(1)-adrenoceptor stimulation activates PKC. In normal ventricular myocytes, acute activation of alpha(1)-adrenoceptors inhibits beta-adrenoceptor stimulated L-type Ca current (I(Ca-L)) and direct activation of epsilonPKC leads to I(Ca-L) inhibition. Because increased PKC activity has been observed chronically in in vivo setting such as failing human heart, we hypothesized that chronic in vivo activation of epsilonPKC alters I(Ca-L) and its response to adrenergic stimulation. Therefore, we investigated the interaction between beta- and alpha(1)-adrenoceptors vis-à-vis I(Ca-L) in myocytes from transgenic mice (TG) with cardiac specific constitutive activation of epsilonPKC (epsilonPKC agonist). Whole-cell I(Ca-L) was recorded from epsilonPKC agonist TG mice and age-matched non-TG (NTG) littermates under: (1) basal condition, (2) beta-adrenoceptor agonist, isoproterenol (ISO), and (3) ISO plus alpha(1)-adrenoceptor agonist, methoxamine. The present results are the first to demonstrate that chronic in vivo activation of epsilonPKC leads to reduced basal I(Ca-L) density. beta-adrenoceptor activation of I(Ca-L) is blunted in epsilonPKC agonist TG mice. alpha-adrenoceptor cross-talk with beta-adrenoceptor signaling pathways vis-à-vis L-type Ca channels is impaired in epsilonPKC agonist TG mice. The diminished response to ISO and methoxamine suggests a protective feedback regulatory mechanism in epsilonPKC agonist TG mice and could be vital in the settings of excessive release of catecholamines during heart failure.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Proteína Quinase C/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1 , Animais , Eletrofisiologia , Ativação Enzimática/efeitos dos fármacos , Isoproterenol/farmacologia , Metoxamina/farmacologia , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Proteína Quinase C-épsilon , Transdução de Sinais
18.
J Autoimmun ; 19(1-2): 79-86, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12367562

RESUMO

It was recently reported that sera from patients with systemic lupus erythematosus contain antibodies reactive with the second extracellular loop of the serotoninergic 5-HT(4) receptor expressed in the human heart. This antibody response was associated with antibodies to 52kD SSA/Ro, a reactivity prevalent in mothers of children with congenital heart block (CHB). The current study was undertaken to determine whether the 5-HT(4) receptor is a target of the immune response in these mothers. Initial experiments demonstrated mRNA expression of the 5-HT(4) receptor in the human foetal atrium. Electrophysiologic studies established that human foetal atrial cells express functional 5-HT(4) receptors. Sera from 116 mothers enrolled in the Research Registry for Neonatal Lupus, whose children have CHB, were evaluated. Ninety-nine (85%) of these maternal sera contained antibodies to SSA/Ro, 84% of which were reactive with the 52kD SSA/Ro component by immunoblot. None of the 116 sera were reactive with the peptide spanning aa165-185 of the serotoninergic receptor. Rabbit antisera which recognized this peptide did not react with 52kD SSA/Ro or peptide aa365-382 in the C terminus. Although 5-HT(4) receptors are present and functional in the human foetal heart, maternal antibodies to the 5-HT(4) receptor are not associated with the development of CHB.


Assuntos
Autoantígenos , Autoimunidade/imunologia , Bloqueio Cardíaco/imunologia , RNA Citoplasmático Pequeno , Receptores de Serotonina/imunologia , Ribonucleoproteínas/imunologia , Anticorpos/imunologia , Feminino , Feto/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Miocárdio/imunologia , Receptores de Serotonina/metabolismo , Síndrome de Sjogren/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA