Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Pacing Clin Electrophysiol ; 41(5): 459-471, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29436714

RESUMO

BACKGROUND/AIMS: Cardiac autonomic nerve remodeling (ANR) is an important mechanism of atrial fibrillation (AF). GTP cyclohydrolase I, encoded by GCH1, is the rate-limiting enzyme in de novo synthesis of tetrahydrobiopterin (BH4), an essential cofactor for nitric oxide (NO) synthesis. Previous studies reported that increased BH4 and NO content negatively regulated nerve regeneration. This study investigated the effects of GCH1 on ANR via BH4 pathway, regulated by microRNA-206 (miR-206). METHODS AND RESULTS: In canines, atrial tachypacing (A-TP), together with miR-206 overexpression, increased PGP9.5 level and inhibited GCH1 expression by quantitative real-time polymerase chain reaction and western blot analysis. GCH1 was validated to be a direct target of miR-206 by luciferase assays. Meanwhile, miR-206 overexpression by lentiviruses infection into right superior pulmonary vein fat pad decreased GCH1 expression to ∼40% and further reduced BH4 and NO content compared with the control canines. After infection of GCH1 overexpression lentiviruses for two weeks, atrial effective refractory period was increased compared with the control group (105.8 ± 1.537 ms vs 99.17 ± 2.007 ms, P < 0.05). Moreover, GCH1 overexpression attenuated canines' atrial PGP9.5 level to ∼56% of the controls. In myocardial cells, transfection of GCH1 overexpression lentiviruses also decreased PGP9.5 expression to 26% of the control group. In patients, plasma was collected and miR-206 expression was upregulated in AF patients (n = 18) than the controls (n = 12). CONCLUSIONS: Our findings suggested that GCH1 downregulation exacerbated ANR by decreasing atrial BH4 and NO content modulated by miR-206 in A-TP canines. This indicates that GCH1 may prevent the initiation of AF through inhibiting ANR.


Assuntos
Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/veterinária , Vias Autônomas/enzimologia , Vias Autônomas/fisiopatologia , Biopterinas/análogos & derivados , GTP Cicloidrolase/metabolismo , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/fisiopatologia , MicroRNAs/metabolismo , Animais , Biopterinas/metabolismo , Western Blotting , Estimulação Cardíaca Artificial , Cães , Óxido Nítrico/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
2.
Trends Cardiovasc Med ; 27(7): 451-459, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28687226

RESUMO

While it is well known that mutation of several different ion channels can cause congenital long QT syndrome, block of IKr is widely thought to be responsible for most cases of drug-induced acquired long QT syndrome (aLQTS). In this article, we review evidence supporting another cause of aLQTS due to inhibition of phosphoinositide 3-kinase (PI3K) signaling. Inhibition of PI3K affects multiple plateau currents, reducing IKr, IKs, and ICaL while increasing the persistent sodium current (INaP). The effects of PI3K inhibitors develop slowly, requiring hours to days to reach steady state. Dofetilide and terfenadine, an antihistamine on which much of the original IKr hypothesis was based, are among the many drugs that inhibit the PI3K pathway. Reduced PI3K signaling may also play a role in aLQTS associated with diabetes. Drug safety testing to identify aLQTS risk may be improved by examining PI3K-dependent effects that develop over time.


Assuntos
Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Síndrome do QT Longo/induzido quimicamente , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/efeitos adversos , Potenciais de Ação , Animais , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Síndrome do QT Longo/enzimologia , Síndrome do QT Longo/fisiopatologia , Fosfatidilinositol 3-Quinase/metabolismo , Bloqueadores dos Canais de Potássio/efeitos adversos , Fatores de Risco , Bloqueadores dos Canais de Sódio/efeitos adversos
3.
Mech Ageing Dev ; 146-148: 72-80, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25956603

RESUMO

Connexin43 (Cx43) is critical for maintaining electrical conduction across atrial muscle. During progressive ageing atrial conduction slows associating with increasing susceptibility to arrhythmias. Changes in Cx43 protein expression, or its phosphorylation status, can instigate changes in the conduction of the cardiac action potential. This study investigated whether increased levels of activated c-jun N-terminal kinase (JNK) is responsible for the decline of Cx43 during ageing. Right atria from guinea pigs aged between 1 day and 38 months of age were examined. The area of the intercalated disc increased with age concurrent with a 75% decline in C43 protein expression. An age-dependent increase in activated-JNK correlated with a rise in phosphorylated Cx43, but also slowing of action potential conduction velocity across the atria from 0.38±0.01 m/s at 1 month of age to 0.30±0.01 m/s at 38 months. The JNK activator anisomycin increased activated JNK in myocytes and reduced Cx43 protein expression simulating ageing. The JNK inhibitor SP600125, was found to eradicate almost all trace of Cx43 protein. We conclude that in vivo activation of JNK increases with age leading to the loss of Cx43 protein resulting in impaired conduction and contributing to the increasing risk of atrial arrhythmias with advancing age.


Assuntos
Envelhecimento/metabolismo , Arritmias Cardíacas/enzimologia , Regulação da Expressão Gênica , Sistema de Condução Cardíaco/enzimologia , MAP Quinase Quinase 4/metabolismo , Envelhecimento/patologia , Animais , Arritmias Cardíacas/patologia , Conexina 43/biossíntese , Feminino , Cobaias , Átrios do Coração/enzimologia , Átrios do Coração/patologia , Sistema de Condução Cardíaco/patologia
4.
Hypertension ; 66(1): 190-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916722

RESUMO

Elevated B-type natriuretic peptide (BNP) regulates cGMP-phosphodiesterase activity. Its elevation is regarded as an early compensatory response to cardiac failure where it can facilitate sympathovagal balance and cardiorenal homeostasis. However, recent reports suggest a paradoxical proadrenergic action of BNP. Because phosphodiesterase activity is altered in cardiovascular disease, we tested the hypothesis that BNP might lose its efficacy by minimizing the action of cGMP on downstream pathways coupled to neurotransmission. BNP decreased norepinephrine release from atrial preparations in response to field stimulation and also significantly reduced the heart rate responses to sympathetic nerve stimulation in vitro. Using electrophysiological recording and fluorescence imaging, BNP also reduced the depolarization evoked calcium current and intracellular calcium transient in isolated cardiac sympathetic neurons. Pharmacological manipulations suggested that the reduction in the calcium transient was regulated by a cGMP/protein kinase G pathway. Fluorescence resonance energy transfer measurements for cAMP, and an immunoassay for cGMP, showed that BNP increased cGMP, but not cAMP. In addition, overexpression of phosphodiesterase 2A after adenoviral gene transfer markedly decreased BNP stimulation of cGMP and abrogated the BNP responses to the calcium current, intracellular calcium transient, and neurotransmitter release. These effects were reversed on inhibition of phosphodiesterase 2A. Moreover, phosphodiesterase 2A activity was significantly elevated in stellate neurons from the prohypertensive rat compared with the normotensive control. Our data suggest that abnormally high levels of phosphodiesterase 2A may provide a brake against the inhibitory action of BNP on sympathetic transmission.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/fisiologia , Sistema de Condução Cardíaco/enzimologia , Hipertensão/enzimologia , Peptídeo Natriurético Encefálico/farmacologia , Sistema Nervoso Simpático/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiologia , Frequência Cardíaca , Hipertensão/genética , Hipertensão/fisiopatologia , Isatina/farmacologia , Masculino , Peptídeo Natriurético Encefálico/fisiologia , Neurônios/enzimologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores do Fator Natriurético Atrial/efeitos dos fármacos , Receptores do Fator Natriurético Atrial/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Gânglio Estrelado/citologia , Gânglio Estrelado/efeitos dos fármacos , Gânglio Estrelado/fisiologia , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica/fisiologia
6.
Europace ; 16(11): 1689-96, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24741027

RESUMO

AIMS: Diabetic patients present a high level of cardiac arrhythmias and risk of cardiac sudden death. The renin-angiotensin system (RAS) plays a key role in diabetes and cardiac diseases. The present study aimed to evaluate whether an angiotensin-converting enzyme 2 (ACE2) activator, diminazene aceturate (DIZE), could improve the streptozotocin (STZ)-induced electrical changes in ventricular repolarization in hyperglycaemic rats. METHODS AND RESULTS: Hyperglycaemia was induced in Wistar male rats with STZ (60 mg/kg/iv). After 4 weeks of STZ injection, rats were daily treated with saline (control) or DIZE (1 mg/kg/gavage) for four consecutive weeks. The cardiac electrical function was evaluated in vivo by electrocardiogram and in vitro by cardiac action potential records in different pacing frequencies. Treatment with DIZE was not able to reverse hyperglycaemia nor body weight loss. However, DIZE reversed hyperglycaemia-induced cardiac electrical changes in ventricular repolarization. Specifically, animals treated with DIZE showed shorter QT and QTc intervals. In addition, ACE2 activation was capable to shorten the cardiac action potential and also reverse the arrhythmic markers. Diminazene aceturate treatment did not induce arrhythmic events in normal, as well as in hyperglycaemic animals. CONCLUSION: Our data indicate that activation of ACE2 has a beneficial effect in hyperglycaemic rats, improving the cardiac electrical function. Thus, DIZE represents a promising new therapeutic agent to treat hyperglycaemia-induced cardiac electrical changes in ventricular repolarization.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/prevenção & controle , Diminazena/análogos & derivados , Ativadores de Enzimas/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Hiperglicemia/tratamento farmacológico , Peptidil Dipeptidase A/metabolismo , Estreptozocina , Potenciais de Ação , Enzima de Conversão de Angiotensina 2 , Animais , Arritmias Cardíacas/sangue , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/fisiopatologia , Diminazena/farmacologia , Ativação Enzimática , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Hiperglicemia/sangue , Hiperglicemia/induzido quimicamente , Hiperglicemia/enzimologia , Hiperglicemia/fisiopatologia , Masculino , Ratos Wistar , Fatores de Tempo
7.
Bull Exp Biol Med ; 155(5): 618-21, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24288723

RESUMO

Histoenzymological changes, indicating inhibition of the main metabolic processes, were found in the conduction cardiomyocytes of the left ventricle and ventricular septum in experimental stenosis of the aorta. The histoenzymological changes in the conduction system of both ventricles and ventricular septum were similar in experimental stenosis of the pulmonary artery and indicated primarily activation of glycolysis. The histoenzymological profile of conduction cardiomyocytes differed little in cases when the increase of the pressure load was complicated or not complicated by the development of heart failure, particularly in pulmonary artery stenosis. The histoenzymological changes in the conduction system in response to increased afterload differed significantly from those in the contractile myocardium and correlated with the level of cellular functional activity and sensitivity to the regulatory and alterative exposure. These data attest to minor role of metabolic shifts in conduction cell injuries with increasing afterload, primarily, of the right ventricle.


Assuntos
Sistema de Condução Cardíaco/enzimologia , Insuficiência Cardíaca/enzimologia , Ventrículos do Coração/enzimologia , Miócitos Cardíacos/enzimologia , Animais , Estenose da Valva Aórtica/enzimologia , Estenose da Valva Aórtica/patologia , Glicólise , Cobaias , Sistema de Condução Cardíaco/patologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/patologia , Contração Miocárdica , Miocárdio/enzimologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Circulação Pulmonar , Estenose da Valva Pulmonar/enzimologia , Estenose da Valva Pulmonar/patologia , Septo Interventricular/enzimologia , Septo Interventricular/patologia
8.
Drug Saf ; 36(5): 295-316, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23620167

RESUMO

The development of tyrosine kinase inhibitors (TKI) represents a major milestone in oncology. However, their use has been found to be associated with serious toxicities that impinge on various vital organs including the heart. Sixteen TKIs have been approved for use in oncology as of 30 September 2012, and a large number of others are in development or under regulatory review. Cardiovascular safety of medicinal products is a major public health issue that has concerned all the stakeholders. This review focuses on three specific cardiovascular safety aspects of TKIs, namely their propensity to induce QT interval prolongation, left ventricular (LV) dysfunction and hypertension (both systemic and pulmonary). Analyses of information in drug labels, the data submitted to the regulatory authorities and the published literature show that a number of TKIs are associated with these undesirable effects. Whereas LV dysfunction and systemic hypertension are on-target effects related to the inhibition of ligand-related signalling pathways, QT interval prolongation appears to be an off-target class III electrophysiologic effect, possibly related to the presence of a fluorine-based pharmacophore. If not adequately managed, these cardiovascular effects significantly increase the morbidity and mortality in a population already at high risk. Hitherto, the QT effect of most QT-prolonging TKIs (except lapatinib, nilotinib, sunitinib and vandetanib) is relatively mild at clinical doses and has not led to appreciable morbidity clinically. In contrast, LV dysfunction and untreated hypertension have resulted in significant morbidity. Inevitably, dilemmas arise in determining the risk/benefit of a TKI therapy in an individual patient who develops any of these effects following the treatment of the TKI-sensitive cancer. QT interval prolongation, hypertension and LV dysfunction can be managed effectively by using reliable methods of measurement and careful monitoring of patients whose clinical management requires optimisation by a close collaboration between an oncologist and a cardiologist, an evolving subspecialty referred to as cardio-oncology. Despite their potential adverse clinical impact, the effects of TKIs on hypertension and LV function are generally inadequately characterised during their development. As has been the case with QT liability of drugs, there is now a persuasive case for a regulatory requirement to study TKIs systematically for these effects. Furthermore, since most of these novel drugs are studied in trials with relatively small sample sizes and approved on an expedited basis, there is also a compelling case for their effective pharmacovigilance and on-going reassessment of their risk/benefit after approval.


Assuntos
Cardiotoxinas/efeitos adversos , Doenças Cardiovasculares/induzido quimicamente , Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Inibidores de Proteínas Quinases/efeitos adversos , Animais , Cardiotoxinas/farmacocinética , Doenças Cardiovasculares/enzimologia , Sistema de Condução Cardíaco/enzimologia , Frequência Cardíaca/fisiologia , Humanos , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/enzimologia , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 304(4): H600-9, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23241322

RESUMO

Mesenchymal stem cells (MSCs) were shown to improve cell survival and alleviate cardiac arrhythmias when transplanted into cardiac tissue; however, little is known about the mechanism by which MSCs modify the electrophysiological properties of cardiac tissue. We aimed to distinguish the influence of cell-cell coupling between myocytes and MSCs from that of MSC-derived paracrine factors on the spontaneous activity and conduction velocity (θ) of multicellular cardiomyocyte preparations. HL-1 cells were plated on microelectrode arrays and their spontaneous activity and θ was determined from field potential recordings. In heterocellular cultures of MSCs and HL-1 cells the beating frequency was attenuated (t(0h): 2.26 ± 0.18 Hz; t(4h): 1.98 ± 0.26 Hz; P < 0.01) concomitant to the intercellular coupling between MSCs and cardiomyocytes. In HL-1 monolayers supplemented with MSC conditioned media (ConM) or tyrode (ConT) θ significantly increased in a time-dependent manner (ConT: t(0h): 2.4 cm/s ± 0.2; t(4h): 3.1 ± 0.4 cm/s), whereas the beating frequency remained constant. Connexin (Cx)43 mRNA and protein expression levels also increased after ConM or ConT treatment over the same time period. Enhanced low-density lipoprotein receptor-related protein 6 (LRP6) phosphorylation after ConT treatment implicates the Wnt signaling pathway. Suppression of Wnt secretion from MSCs (IWP-2; 5 µmol/l) reduced the efficacy of ConT to induce phospho-LRP6 and to increase θ. Inhibition of ß-catenin (cardamonin; 10 µmol/l) or GSK3-α/ß (LiCl; 5 mmol/l) also suppressed changes in θ, further supporting the hypothesis that MSC-mediated Cx43 upregulation occurs in part through secreted Wnt ligands and activation of the canonical Wnt signaling pathway.


Assuntos
Conexina 43/biossíntese , Sistema de Condução Cardíaco/fisiologia , Transplante de Células-Tronco Mesenquimais , Comunicação Parácrina/fisiologia , Regulação para Cima/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular , Chalconas/farmacologia , Meios de Cultivo Condicionados , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/enzimologia , Cloreto de Lítio/farmacologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/fisiologia , Comunicação Parácrina/efeitos dos fármacos , Fosforilação , Regulação para Cima/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/antagonistas & inibidores
11.
Am J Physiol Heart Circ Physiol ; 301(3): H975-83, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21666110

RESUMO

Proline-rich tyrosine kinase 2 (Pyk2) is a nonreceptor protein kinase regulated by intracellular Ca(2+), CaMK, and PKC and can be activated by different stress signals involved in heart failure. However, Pyk2 has not been investigated in the human heart, and the functional role of Pyk2 signaling at the whole heart level has not been elucidated. We hypothesize that Ca(2+)-dependent activation of Pyk2 is involved in cardiac electrophysiology. We examined the expression of Pyk2 in nonfailing versus ischemic and nonischemic failing human hearts (n = 6 hearts/group). To investigate Pyk2 function, we optically mapped perfused hearts from wild-type (WT; n = 7) and knockout (Pyk2(-/-); n = 8) mice during autonomic stimulation. Experiments were done in control mice and after 1 wk of transverse aortic constriction. We used the Illumina beadarray approach for transcriptional profiling of WT and Pyk2(-/-) mouse ventricles. Western blot analysis revealed a doubling of Pyk2 activation in nonischemic failing versus nonfailing human hearts. In mouse hearts, we observed a much higher probability of ventricular tachyarrhythmia during ACh perfusion in Pyk2(-/-) versus WT mice. Parasympathetic stimulation resulted in a dose-dependent decrease of atrial action potential duration (APD) in both WT and Pyk2(-/-) mice, whereas in ventricles it induced APD shortening in Pyk2(-/-) mice but not in WT mice. Deficiency of Pyk2 abolished ACh-induced prolongation of atrioventricular delay in Pyk2(-/-) mouse hearts but did not affect heart rate. Lower mRNA and protein levels of sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 and higher mRNA levels of Na(+)/Ca(2+) exchanger 1 were detected in Pyk2(-/-) hearts compared with WT hearts. The transverse aortic constriction protocol did not change the phenotype. In conclusion, our results indicate a protective role of Pyk2 with respect to ventricular tachyarrhythmia during parasympathetic stimulation by regulation of gene expression related to Ca(2+) handling. We hypothesize that activation of Pyk2 in the human heart during heart failure may contribute to protection against arrhythmia.


Assuntos
Arritmias Cardíacas/enzimologia , Quinase 2 de Adesão Focal/metabolismo , Sistema de Condução Cardíaco/enzimologia , Frequência Cardíaca , Acetilcolina , Potenciais de Ação , Adulto , Idoso , Análise de Variância , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Sistema Nervoso Autônomo/fisiopatologia , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Estimulação Cardíaca Artificial , Estudos de Casos e Controles , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Quinase 2 de Adesão Focal/deficiência , Quinase 2 de Adesão Focal/genética , Regulação da Expressão Gênica , Genótipo , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Humanos , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Perfusão , Fenótipo , Fosforilação , RNA Mensageiro/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo , Imagens com Corantes Sensíveis à Voltagem
12.
J Physiol Pharmacol ; 61(1): 53-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20228415

RESUMO

Leukotrienes are lipid mediators produced via 5-lipooxygenase pathway of arachidonic acid. At least two cysteinyl-leukotrienes receptors are highly expressed in the heart, including the conduction system. Coronary angiography or angioplasty is accompanied by release of cysteinyl leukotrienes into coronary circulation and into urine. We tested the hypothesis that inhibition of leukotrienes biosynthesis would affect the conductance system function. In a double-blind placebo controlled study, patients with stable angina undergoing elective coronary catheterization or angioplasty were randomly assigned to 48 hrs treatment with a 5-lipoxgenase inhibitor (n=54) or placebo (n=49). ECG Holter recording was carried out for 24 hrs before and after the procedure and urinary leukotriene E(4) measurements were done. Inhibition of 5-lipoxygenase caused 26% reduction of urinary leukotriene E(4), associated with: 1) decrease in heart rate by about 7%, 2) enhanced heart rate variability; 3) protection against depressions in atrioventricular conductance and ventricular repolarization induced by the procedure. No effects on either arrhythmias, or ECG patterns of ischemia were noted. We conclude that pharmacological inhibition of 5-lipoxygenase, shortly before percutaneous coronary intervention, reveals specific actions of leukotrienes on the heart rhythm. Inhibitors of 5-lipoxygenase might be of interest as a novel class of cardiac drugs affecting the conductive system.


Assuntos
Sistema de Condução Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Antagonistas de Leucotrienos/farmacologia , Leucotrienos , Idoso , Angina Instável/tratamento farmacológico , Angina Instável/enzimologia , Angina Instável/urina , Araquidonato 5-Lipoxigenase/biossíntese , Método Duplo-Cego , Feminino , Sistema de Condução Cardíaco/enzimologia , Humanos , Leucotrieno E4/antagonistas & inibidores , Leucotrieno E4/biossíntese , Leucotrieno E4/urina , Leucotrienos/biossíntese , Leucotrienos/metabolismo , Leucotrienos/fisiologia , Inibidores de Lipoxigenase/farmacologia , Masculino , Pessoa de Meia-Idade
13.
Cardiovasc Res ; 87(3): 485-93, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20211865

RESUMO

AIMS: The small GTPase Rac1 seems to play a role in the pathogenesis of atrial fibrillation (AF). The aim of the present study was to characterize the effects of Rac1 overexpression on atrial electrophysiology. METHODS AND RESULTS: In mice with cardiac overexpression of constitutively active Rac1 (RacET), statin-treated RacET, and wild-type controls (age 6 months), conduction in the right and left atrium (RA and LA) was mapped epicardially. The atrial effective refractory period (AERP) was determined and inducibility of atrial arrhythmias was tested. Action potentials were recorded in isolated cells. Left ventricular function was measured by pressure-volume analysis. Five of 11 RacET hearts showed spontaneous or inducible atrial tachyarrhythmias vs. 0 of 9 controls (P < 0.05). In RacET, the P-wave duration was significantly longer (26.8 +/- 2.1 vs. 16.7 +/- 1.1 ms, P = 0.001) as was total atrial activation time (RA: 13.6 +/- 4.4 vs. 3.2 +/- 0.5 ms; LA: 7.1 +/- 1.2 vs. 2.2 +/- 0.3 ms, P < 0.01). Prolonged local conduction times occurred more often in RacET (RA: 24.4 +/- 3.8 vs. 2.7 +/- 2.1%; LA: 19.1 +/- 6.3 vs. 1.2 +/- 0.7%, P < 0.01). The AERP and action potential duration did not differ significantly between both groups. RacET demonstrated significant atrial fibrosis but only moderate systolic heart failure. RacET and statin-treated RacET were not significantly different regarding atrial electrophysiology. CONCLUSION: The substrate for atrial arrhythmias in mice with Rac1 overexpression is characterized by conduction disturbances and atrial fibrosis. Electrical remodelling (i.e. a shortening of AERP) does not play a role. Statin treatment cannot prevent the structural and electrophysiological effects of pronounced Rac1 overexpression in this model.


Assuntos
Arritmias Cardíacas/enzimologia , Função Atrial , Átrios do Coração/enzimologia , Sistema de Condução Cardíaco/enzimologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Função Atrial/efeitos dos fármacos , Eletrocardiografia , Fibrose , Regulação da Expressão Gênica , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/patologia , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Camundongos , Camundongos Transgênicos , Fatores de Tempo , Regulação para Cima , Função Ventricular Esquerda , Pressão Ventricular , Proteínas rac1 de Ligação ao GTP/genética
14.
J Mol Cell Cardiol ; 44(2): 293-303, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18182166

RESUMO

Mutations in the lamin A/C (LMNA) gene, which encodes nuclear membrane proteins, cause a variety of human conditions including dilated cardiomyopathy (DCM) with associated cardiac conduction system disease. To investigate mechanisms responsible for electrophysiologic and myocardial phenotypes caused by dominant human LMNA mutations, we performed longitudinal evaluations in heterozygous Lmna(+/-) mice. Despite one normal allele, Lmna(+/-) mice had 50% of normal cardiac lamin A/C levels and developed cardiac abnormalities. Conduction system function was normal in neonatal Lmna(+/-) mice but, by 4 weeks of age, atrioventricular (AV) nodal myocytes had abnormally shaped nuclei and active apoptosis. Telemetric and in vivo electrophysiologic studies in 10-week-old Lmna(+/-) mice showed AV conduction defects and both atrial and ventricular arrhythmias, analogous to those observed in humans with heterozygous LMNA mutations. Isolated myocytes from 12-month-old Lmna(+/-) mice exhibited impaired contractility. In vivo cardiac studies of aged Lmna(+/-) mice revealed DCM; in some mice this occurred without overt conduction system disease. However, neither histopathology nor serum CK levels indicated skeletal muscle pathology. These data demonstrate cardiac pathology due to heterozygous Lmna mutations reflecting a 50% reduction in lamin protein levels. Lamin haploinsufficiency caused early-onset programmed cell death of AV nodal myocytes and progressive electrophysiologic disease. While lamin haploinsufficiency was better tolerated by non-conducting myocytes, ultimately, these too succumbed to diminished lamin levels leading to dilated cardiomyopathy, which presumably arose independently from conduction system disease.


Assuntos
Apoptose , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Sistema de Condução Cardíaco/patologia , Lamina Tipo A/genética , Idade de Início , Animais , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/enzimologia , Nó Atrioventricular/patologia , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/enzimologia , Núcleo Celular/patologia , Separação Celular , Eletrofisiologia , Sistema de Condução Cardíaco/enzimologia , Heterozigoto , Marcação In Situ das Extremidades Cortadas , Lamina Tipo A/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Doenças Musculares/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Telemetria , Ultrassonografia
15.
Heart ; 94(2): 153-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17483124

RESUMO

BACKGROUND: Anderson-Fabry disease is an X-linked glycosphingolipid storage disorder caused by deficient activity of the lysosomal enzyme alpha-galactosidase A. This leads to a progressive accumulation of globotriaosylceramide (Gb(3)) in the lysosomes of cells throughout the body that ultimately results in premature death from renal, cardiac or cerebrovascular complications. Until recently, there was no effective therapy available for this disease. The present study was designed to assess the safety and efficacy of enzyme replacement therapy with agalsidase alfa on the cardiac manifestations of Anderson-Fabry disease. METHOD: The effects of therapy with agalsidase alfa on cardiac structure and function were assessed in a randomised, double-blind, placebo-controlled study of 15 adult male patients with Anderson-Fabry disease. The following parameters were measured at baseline and 6 months: left ventricular mass, QRS duration and levels of Gb(3) in cardiac tissue, urine sediment and plasma. After 6 months of the randomised trial patients were enrolled in a 2-year open-label extension study. RESULTS: Left ventricular mass, as measured by MRI, was significantly reduced following 6 months of treatment with agalsidase alfa compared with placebo (p = 0.041). A mean 20% reduction in myocardial Gb(3) content as assessed by serial transvenous endomyocardial biopsies was demonstrated over the 6 months of enzyme replacement compared to a mean 10% increase in patients receiving placebo (p = 0.42) CONCLUSION: Enzyme replacement therapy with agalsidase alfa resulted in regression of the hypertrophic cardiomyopathy associated with Anderson-Fabry disease.


Assuntos
Cardiomiopatias/tratamento farmacológico , Doença de Fabry/tratamento farmacológico , alfa-Galactosidase/uso terapêutico , Adulto , Idoso , Cardiomiopatias/metabolismo , Cromatografia Líquida de Alta Pressão , Método Duplo-Cego , Ecocardiografia , Eletrocardiografia , Sistema de Condução Cardíaco/enzimologia , Humanos , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Angiografia por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Miocárdio/química , Triexosilceramidas/metabolismo , Disfunção Ventricular Esquerda/tratamento farmacológico , alfa-Galactosidase/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 292(6): H2854-66, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17277017

RESUMO

Alternans of cardiac repolarization is associated with arrhythmias and sudden death. At the cellular level, alternans involves beat-to-beat oscillation of the action potential (AP) and possibly Ca(2+) transient (CaT). Because of experimental difficulty in independently controlling the Ca(2+) and electrical subsystems, mathematical modeling provides additional insights into mechanisms and causality. Pacing protocols were conducted in a canine ventricular myocyte model with the following results: 1) CaT alternans results from refractoriness of the sarcoplasmic reticulum Ca(2+) release system; alternation of the L-type calcium current has a negligible effect; 2) CaT-AP coupling during late AP occurs through the sodium-calcium exchanger and underlies AP duration (APD) alternans; 3) increased Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activity extends the range of CaT and APD alternans to slower frequencies and increases alternans magnitude; its decrease suppresses CaT and APD alternans, exerting an antiarrhythmic effect; and 4) increase of the rapid delayed rectifier current (I(Kr)) also suppresses APD alternans but without suppressing CaT alternans. Thus CaMKII inhibition eliminates APD alternans by eliminating its cause (CaT alternans) while I(Kr) enhancement does so by weakening CaT-APD coupling. The simulations identify combined CaMKII inhibition and I(Kr) enhancement as a possible antiarrhythmic intervention.


Assuntos
Arritmias Cardíacas/fisiopatologia , Sinalização do Cálcio , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Simulação por Computador , Sistema de Condução Cardíaco/fisiopatologia , Modelos Cardiovasculares , Miócitos Cardíacos/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Canais de Potássio de Retificação Tardia/metabolismo , Cães , Cobaias , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/metabolismo , Cinética , Miócitos Cardíacos/enzimologia , Potássio/metabolismo , Reprodutibilidade dos Testes , Projetos de Pesquisa , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Trocador de Sódio e Cálcio/metabolismo
17.
J Mol Cell Cardiol ; 41(2): 364-70, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16806265

RESUMO

Gene transfer of neuronal nitric oxide synthase (nNOS) with nonspecific adenoviral vectors can cause promiscuous transduction. We provide direct evidence that nNOS targeted only to cardiac sympathetic neurons inhibits sympathetic neurotransmission. An adenovirus constructed with a noradrenergic neuron-specific promoter (PRSx8), driving nNOS or enhanced green fluorescence protein (eGFP) gene expression caused exclusive expression in tyrosine hydroxylase (TH) positive rat cardiac sympathetic neurons. There was no detectable leakage of transgene expression in other cell types in the preparation nor did the transgene express in choline acetyltransferase (CHAT)-positive intracardiac cholinergic ganglia. Functionally, Ad.PRS-nNOS gene transfer increased nNOS activity and significantly reduced norephinephrine release evoked by field stimulation of isolated right atria. These effects were reversed by the NOS inhibitor N(omega)-Nitro-L-arginine. Our results demonstrate that noradrenergic cell-specific gene transfer with nNOS can inhibit cardiac sympathetic neurotransmission. This targeted technique may provide a novel method for reducing presynaptic sympathetic hyperactivity.


Assuntos
Sistema de Condução Cardíaco/enzimologia , Neurônios/enzimologia , Óxido Nítrico Sintase Tipo I/biossíntese , Norepinefrina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sistema Nervoso Simpático/enzimologia , Simpatomiméticos/farmacologia , Adenoviridae , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Expressão Gênica/efeitos dos fármacos , Átrios do Coração/citologia , Átrios do Coração/enzimologia , Óxido Nítrico Sintase Tipo I/genética , Ratos , Ratos Sprague-Dawley , Transdução Genética
18.
Ann Anat ; 184(2): 125-36, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11936191

RESUMO

This study was conducted to determine the overall number of intrinsic neurons distributed through-out the entire heart, in which most neurons are located inside of intramural ganglia and are hidden to observers. For this reason, we attempted to ascertain: (1) how the number of neurons located inside of intrinsic cardiac ganglion is related to its area, and (2) whether this relationship is dependent on age and species of animals. Hearts of rats, guinea pigs, dogs and humans were used to examine intramural ganglia stained histochemically for acetylcholinesterase (AChE). The number and parameters of neurons located inside of 104 ganglia were estimated in serial sections. Although the revealed intrinsic cardiac ganglia varied extremely in shape and size, two different types were identified: the globular and plain ones. In the plain ganglia, perikarya of side by side situated neurons were always intensely stained for AChE and, being clearly discernible, they could be reliably counted in any plain ganglia on total heart preparations using a contact microscope. Contrarily, neuron somata in the globular ganglia were densely packed above one another and their perikarya were almost indiscernible for the observer. Counting of neurons located inside of globular ganglia was possible in serial sections only. The largest cardiac ganglia were revealed in dogs, in which some globular ganglia containing up to 2000 neurons occupied more than 1 mm2. In spite of evident species-dependent differences with respect to frequency of large ganglia, the majority of intrinsic cardiac ganglia both in humans and animals were comparatively small, involved approximately 100-200 nerve cells and occupied an area ranging from 0.01 to 0.17 mm2. Overall, the number of neurons located inside of globular ganglion was related to its area (correlation coefficient = 0.82). However, the correlation coefficients between the globular ganglion area and its neuron number were unequal in different species (0.92 in guinea pig; 0.80 in dog; 0.72 in human; and 0.44 in rat) as well as dependent on (1) ganglion size (0.8 for ganglia equal to or larger than 0.17 mm2 and 0.6 for ganglia smaller than 0.17 mm2) and (2) age of specimens (respectively, 0.98 for juvenile and 0.87 for adult dogs; 0.71 for infants and 0.54 for aged human). In all examined animals and humans, the mean measurements of neuron perikarya were similar (on average, 23 microm in width, 32 microm in length, and 615 microm2 in area) and differences between them were statistically insignificant. However, neuron perikarya of adult dogs and aged humans were significantly larger than those revealed in the juvenile dogs and infants, respectively. Based on the data of this study, we concluded that the number of intrinsic cardiac neurons may be approximated in the total heart preparation via counting and measuring of intramural ganglia, contours of which are well-discernible following a histochemical reaction for AChE.


Assuntos
Acetilcolinesterase/análise , Gânglios/enzimologia , Sistema de Condução Cardíaco/citologia , Neurônios/citologia , Idoso , Animais , Cães , Feminino , Gânglios/citologia , Cobaias , Sistema de Condução Cardíaco/enzimologia , Sistema de Condução Cardíaco/crescimento & desenvolvimento , Histocitoquímica , Humanos , Lactente , Masculino , Neurônios/enzimologia , Ratos , Ratos Wistar , Análise de Regressão
19.
Neurosci Lett ; 305(2): 111-4, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11376896

RESUMO

We previously reported three kinds of nitric oxide synthase-immunoreactive (NOS-ir) axons in the guinea pig heart: the sparse fiber network covering the right atrium, the basket-like endings around intracardiac neurons, and the axons in the septal region. The sparse NOS-ir nerve fiber network in the right atrium remained after vagotomy and has been suggested to be originated from intrinsic cardiac ganglia. Using Chorera toxin B as a retrograde tracer, we determined a part of them were derived from cardiac ganglionic neurons located in the area near the vena cavae.


Assuntos
Sistema de Condução Cardíaco/enzimologia , Fibras Nervosas/enzimologia , Óxido Nítrico Sintase/metabolismo , Animais , Toxina da Cólera , Feminino , Gânglios/enzimologia , Cobaias , Átrios do Coração , Imuno-Histoquímica , Rede Nervosa/enzimologia , Vagotomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA