Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 596(24): 6141-6155, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30259525

RESUMO

KEY POINTS: The sinoatrial node (SAN) is the primary pacemaker of the heart. SAN dysfunction, or 'sick sinus syndrome', can cause excessively slow heart rates and pauses, leading to exercise limitation and syncope, currently treated by implantation of an electronic pacemaker. 'Biopacemaking' utilises gene therapy to restore pacemaker activity by manipulating gene expression. Overexpressing the HCN pacemaker ion channel has been widely used with limited success. We utilised bradycardic rat subsidiary atrial pacemaker tissue to evaluate alternative gene targets: the Na+ /Ca2+ exchanger NCX1, and the transcription factors TBX3 and TBX18 known to be involved in SAN embryonic development. TBX18 overexpression restored normal SAN function, as assessed by increased rate, improved heart rate stability and restoration of isoprenaline response. TBX3 and NCX1 were not effective in accelerating the rate of subsidiary atrial pacemaker tissue. Gene therapy targeting TBX18 could therefore have the potential to restore pacemaker function in human sick sinus syndrome obviating electronic pacemakers. ABSTRACT: The sinoatrial node (SAN) is the primary pacemaker of the heart. Disease of the SAN, sick sinus syndrome, causes heart rate instability in the form of bradycardia and pauses, leading to exercise limitation and syncope. Biopacemaking aims to restore pacemaker activity by manipulating gene expression, and approaches utilising HCN channel overexpression have been widely used. We evaluated alternative gene targets for biopacemaking to restore normal SAN pacemaker physiology within bradycardic subsidiary atrial pacemaker (SAP) tissue, using the Na+ /Ca2+ exchanger NCX1, and the transcription factors TBX3 and TBX18. TBX18 expression in SAP tissue restored normal SAN function, as assessed by increased rate (SAN 267.5 ± 13.6 bpm, SAP 144.1 ± 8.6 bpm, SAP-TBX18 214.4 ± 14.4 bpm; P < 0.001), improved heart rate stability (standard deviation of RR intervals fell from 39.3 ± 7.2 ms to 6.9 ± 0.8 ms, P < 0.01; root mean square of successive differences of RR intervals fell from 41.7 ± 8.2 ms to 6.1 ± 1.2 ms, P < 0.01; standard deviation of points perpendicular to the line of identity of Poincaré plots (SD1) fell from 29.5 ± 5.8 ms to 7.9 ± 2.0 ms, P < 0.05) and restoration of isoprenaline response (increases in rates of SAN 65.5 ± 1.3%, SAP 28.4 ± 3.4% and SAP-TBX18 103.3 ± 10.2%; P < 0.001). These changes were driven by a TBX18-induced switch in the dominant HCN isoform in SAP tissue, with a significant upregulation of HCN2 (from 1.01 × 10-5  ± 2.2 × 10-6 to 2.8 × 10-5  ± 4.3 × 10-6 arbitrary units, P < 0.001). Biophysically detailed computer modelling incorporating isoform-specific HCN channel electrophysiology confirmed that the measured changes in HCN abundance could account for the observed changes in beating rates. TBX3 and NCX1 were not effective in accelerating the rate of SAP tissue.


Assuntos
Sistema de Condução Cardíaco/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Síndrome do Nó Sinusal/terapia , Nó Sinoatrial/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Simulação por Computador , Regulação da Expressão Gênica , Átrios do Coração , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Masculino , Modelos Biológicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratos , Trocador de Sódio e Cálcio/metabolismo , Proteínas com Domínio T/genética , Técnicas de Cultura de Tecidos
2.
Exp Physiol ; 102(11): 1424-1434, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28804970

RESUMO

NEW FINDINGS: What is the central question of this study? Type 2 diabetes is associated with a higher rate of ventricular arrhythmias compared with the non-diabetic population, but the associated myocardial gene expression changes are unknown; furthermore, it is also unknown whether any changes are attributable to chronic hyperglycaemia or are a consequence of structural changes. What is the main finding and its importance? We found downregulation of left ventricular ERG gene expression and increased NCX1 gene expression in humans with type 2 diabetes compared with control patients with comparable left ventricular hypertrophy and possible myocardial fibrosis. This was associated with QT interval prolongation. Diabetes and associated chronic hyperglycaemia may therefore promote ventricular arrhythmogenesis independently of structural changes. Type 2 diabetes is associated with a higher rate of ventricular arrhythmias, and this is hypothesized to be independent of coronary artery disease or hypertension. To investigate further, we compared changes in left ventricular myocardial gene expression in type 2 diabetes patients with patients in a control group with left ventricular hypertrophy. Nine control patients and seven patients with type 2 diabetes with aortic stenosis undergoing aortic valve replacement had standard ECGs, signal-averaged ECGs and echocardiograms before surgery. During surgery, a left ventricular biopsy was taken, and mRNA expressions for genes relevant to the cardiac action potential were estimated by RT-PCR. Mathematical modelling of the action potential and calcium transient was undertaken using the O'Hara-Rudy model using scaled changes in gene expression. Echocardiography revealed similar values for left ventricular size, filling pressures and ejection fraction between groups. No difference was seen in positive signal-averaged ECGs between groups, but the standard ECG demonstrated a prolonged QT interval in the diabetes group. Gene expression of KCNH2 and KCNJ3 were lower in the diabetes group, whereas KCNJ2, KCNJ5 and SLC8A1 expression were higher. Modelling suggested that these changes would lead to prolongation of the action potential duration with generation of early after-depolarizations secondary to a reduction in density of the rapid delayed rectifier K+ current and increased Na+ -Ca2+ exchange current. These data suggest that diabetes leads to pro-arrythmogenic changes in myocardial gene expression independently of left ventricular hypertrophy or fibrosis in an elderly population.


Assuntos
Estenose da Valva Aórtica/genética , Arritmias Cardíacas/genética , Diabetes Mellitus Tipo 2/genética , Hipertrofia Ventricular Esquerda/genética , Volume Sistólico , Função Ventricular Esquerda , Remodelação Ventricular , Potenciais de Ação , Idoso , Idoso de 80 Anos ou mais , Estenose da Valva Aórtica/complicações , Estenose da Valva Aórtica/diagnóstico , Estenose da Valva Aórtica/fisiopatologia , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Estudos de Casos e Controles , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/fisiopatologia , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Feminino , Fibrose , Regulação da Expressão Gênica , Frequência Cardíaca , Humanos , Hipertrofia Ventricular Esquerda/diagnóstico , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Modelos Cardiovasculares , Modelos Genéticos , Miocárdio/metabolismo , Miocárdio/patologia , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo
3.
Am J Physiol Cell Physiol ; 311(2): C255-68, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27281482

RESUMO

Polyunsaturated fatty acids (PUFAs) modulate voltage-gated K(+) channel inactivation by an unknown site and mechanism. The effects of ω-6 and ω-3 PUFAs were investigated on the heterologously expressed Kv1.4 channel. PUFAs inhibited wild-type Kv1.4 during repetitive pulsing as a result of slowing of recovery from inactivation. In a mutant Kv1.4 channel lacking N-type inactivation, PUFAs reversibly enhanced C-type inactivation (Kd, 15-43 µM). C-type inactivation was affected by extracellular H(+) and K(+) as well as PUFAs and there was an interaction among the three: the effect of PUFAs was reversed during acidosis and abolished on raising K(+) Replacement of two positively charged residues in the extracellular pore (H508 and K532) abolished the effects of the PUFAs (and extracellular H(+) and K(+)) on C-type inactivation but had no effect on the lipoelectric modulation of voltage sensor activation, suggesting two separable interaction sites/mechanisms of action of PUFAs. Charge calculations suggest that the acidic head group of the PUFAs raises the pKa of H508 and this reduces the K(+) occupancy of the selectivity filter, stabilizing the C-type inactivated state.


Assuntos
Ácidos Graxos Insaturados/metabolismo , Canal de Potássio Kv1.4/metabolismo , Animais , Hidrogênio/metabolismo , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Xenopus laevis/metabolismo , Xenopus laevis/fisiologia
4.
Folia Morphol (Warsz) ; 74(2): 258-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26050816

RESUMO

In this study, a fixation protocol using a 10% neutral buffered formalin (FA) solution and another protocol using a methanol (MeOH) solution were compared for detection of ion channels, Kv1.5, Kv4.2, Cav1.2, Kir6.2, Nav1.5 and Nav1.1 in rat myocytes by immunolabelling. Kv1.5 and Kv4.2 at intercalated discs and Cav1.2 at transverse tubules were not detected by FA but were detected by MeOH. Kir6.2 at transverse tubules and Nav1.5 at sarcolemma were detected by FA but not by MeOH. It is suggested that both FA and MeOH fixation protocols should be used for the detection of cardiac ion channels by immunolabelling.

5.
J Mol Cell Cardiol ; 83: 88-100, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25668431

RESUMO

Sick sinus syndrome remains a highly relevant clinical entity, being responsible for the implantation of the majority of electronic pacemakers worldwide. It is an infinitely more complex disease than it was believed when first described in the mid part of the 20th century. It not only involves the innate leading pacemaker region of the heart, the sinoatrial node, but also the atrial myocardium, predisposing to atrial tachydysrhythmias. It remains controversial as to whether the dysfunction of the sinoatrial node directly causes the dysfunction of the atrial myocardium, or vice versa, or indeed whether these two aspects of the condition arise through some related underlying pathological mechanism, such as extracellular matrix remodeling, i.e., fibrosis. This review aims to shed new light on the myriad possible contributing factors in the development of sick sinus syndrome, with a particular focus on the sinoatrial nodal myocyte. This article is part of a Special Issue entitled CV Aging.


Assuntos
Envelhecimento/metabolismo , Fibrilação Atrial/metabolismo , Bradicardia/metabolismo , Átrios do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Nó Sinoatrial/metabolismo , Idoso , Envelhecimento/patologia , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/patologia , Bradicardia/genética , Bradicardia/patologia , Conexinas/genética , Conexinas/metabolismo , Regulação da Expressão Gênica , Átrios do Coração/patologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Transporte de Íons , MicroRNAs/genética , MicroRNAs/metabolismo , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Sistema Renina-Angiotensina/genética , Nó Sinoatrial/patologia
6.
Int J Cardiol ; 177(2): 423-8, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25305681

RESUMO

BACKGROUND: Macitentan is a new endothelin receptor antagonist that is used to treat pulmonary arterial hypertension in humans. Treatment of established pulmonary hypertension with macitentan was studied using the monocrotaline model of pulmonary hypertension. METHODS: Three groups of rats were created (n=12): control (CON: macitentan only), monocrotaline (MCT: monocrotaline only) and macitentan (MACI: macitentan and monocrotaline). Monocrotaline (60 mg/kg) was injected in the MCT and MACI groups on day 0; volume matched saline was injected in the CON groups. Macitentan therapy (30 mg/kg/day) was commenced on day 11 in the CON and MACI groups. Serial echocardiography and ECGs were performed. The rats were sacrificed if they showed clinical deterioration. RESULTS: The MCT and MACI rats showed signs of pulmonary hypertension by day 7 (maximum pulmonary velocity, CON 1.15 ± 0.15m/s vs MCT 1.04 ± 0.10 m/s vs MACI 0.99 ± 0.18 m/s; p<0.05). Both the MCT and MACI groups developed pulmonary hypertension, but this was less severe in the MACI group (day 21 pulmonary artery acceleration time, MCT 17.55 ± 1.56 ms vs MACI 22.55 ± 1.00 ms; pulmonary artery deceleration, MCT 34.72 ± 3.72 m/s(2) vs MACI 17.30 ± 1.89 m/s(2); p<0.05). Right ventricular hypertrophy and QT interval increases were more pronounced in MCT than MACI (right ventricle wall thickness, MCT 0.13 ± 0.1cm vs MACI 0.10 ± 0.1cm; QT interval, MCT 85 ± 13 ms vs MACI 71 ± 14 ms; p<0.05). Survival benefit was not seen in the MACI group (p=0.50). CONCLUSIONS: Macitentan treatment improves haemodynamic parameters in established pulmonary hypertension. Further research is required to see if earlier introduction of macitentan has greater effects.


Assuntos
Modelos Animais de Doenças , Progressão da Doença , Antagonistas dos Receptores de Endotelina/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/patologia , Pirimidinas/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Masculino , Ratos , Ratos Wistar , Resultado do Tratamento
7.
Front Physiol ; 2: 67, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022314

RESUMO

Strong emotion and mental stress are now recognized as playing a significant role in severe and fatal ventricular arrhythmias. The mechanisms, although incompletely understood, include central processing at the cortical and brain stem level, the autonomic nerves and the electrophysiology of the myocardium. Each of these is usually studied separately by investigators from different disciplines. However, many are regulatory processes which incorporate interactive feedforward and feedback mechanisms. In this review we consider the whole as an integrated interactive brain-heart system.

8.
Exp Physiol ; 96(11): 1163-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21724736

RESUMO

The function of the sino-atrial node (SAN), the pacemaker of the heart, is known to decline with age, resulting in pacemaker disease in the elderly. The aim of the study was to investigate the effects of ageing on the SAN by characterizing electrophysiological changes and determining whether changes in gene expression are involved. In young and old rats, SAN function was characterized in the anaesthetized animal, isolated heart and isolated right atrium using ECG and action potential recordings; gene expression was characterized using quantitative PCR. The SAN function declined with age as follows: the intrinsic heart rate declined by 18 ± 3%; the corrected SAN recovery time increased by 43 ± 13%; and the SAN action potential duration increased by 11 ± 3% (at 75% repolarization). Gene expression in the SAN changed considerably with age, e.g. there was an age-dependent decrease in the Ca(2+) clock gene, RYR2, and changes in many ion channels (e.g. increases in Na(v)1.5, Na(v)ß1 and Ca(v)1.2 and decreases in K(v)1.5 and HCN1). In conclusion, with age, there are changes in the expression of ion channel and Ca(2+) clock genes in the SAN, and the changes may provide a partial explanation for the age-dependent decline in pacemaker function.


Assuntos
Envelhecimento/fisiologia , Canais Iônicos/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Nó Sinoatrial/fisiologia , Potenciais de Ação , Animais , Função do Átrio Direito/fisiologia , Canais de Cálcio/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Ecocardiografia , Frequência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Técnicas In Vitro , Perfusão , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Ratos , Nó Sinoatrial/fisiopatologia , Canais de Sódio/metabolismo , Canais de Cátion TRPC/fisiologia
9.
Circ Heart Fail ; 4(4): 496-508, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21565973

RESUMO

BACKGROUND: Heart failure (HF) causes a decline in the function of the pacemaker of the heart-the sinoatrial node (SAN). The aim of the study was to investigate HF-induced changes in the expression of the ion channels and related proteins underlying the pacemaker activity of the SAN. METHODS AND RESULTS: HF was induced in rats by the ligation of the proximal left coronary artery. HF animals showed an increase in the left ventricular (LV) diastolic pressure (317%) and a decrease in the LV systolic pressure (19%) compared with sham-operated animals. They also showed SAN dysfunction wherein the intrinsic heart rate was reduced (16%) and the corrected SAN recovery time was increased (56%). Quantitative polymerase chain reaction was used to measure gene expression. Of the 91 genes studied during HF, 58% changed in the SAN, although only 1% changed in the atrial muscle. For example, there was an increase in the expression of ERG, K(v)LQT1, K(ir)2.4, TASK1, TWIK1, TWIK2, calsequestrin 2, and the A1 adenosine receptor in the SAN that could explain the slowing of the intrinsic heart rate. In addition, there was an increase in Na(+)-H(+) exchanger, and this could be the stimulus for the remodeling of the SAN. CONCLUSIONS: SAN dysfunction is associated with HF and is the result of an extensive remodeling of ion channels; gap junction channels; Ca(2+)-, Na(+)-, and H(+)-handling proteins; and receptors in the SAN.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Canais Iônicos/genética , Canais Iônicos/fisiologia , Nó Sinoatrial/fisiopatologia , Animais , Canais de Cálcio/genética , Canais de Cálcio/fisiologia , Conexinas/genética , Conexinas/fisiologia , Modelos Animais de Doenças , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Insuficiência Cardíaca/patologia , Frequência Cardíaca/fisiologia , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/fisiologia , Canais de Potássio/genética , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Canais de Sódio/genética , Canais de Sódio/fisiologia , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/fisiologia
10.
J Mol Cell Cardiol ; 50(4): 642-51, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21256850

RESUMO

The atrioventricular conduction axis, located in the septal component of the atrioventricular junctions, is arguably the most complex structure in the heart. It fulfils a multitude of functions, including the introduction of a delay between atrial and ventricular systole and backup pacemaking. Like any other multifunctional tissue, complexity is a key feature of this specialised tissue in the heart, and this complexity is both anatomical and electrophysiological, with the two being inextricably linked. We used quantitative PCR, histology and immunohistochemistry to analyse the axis from six human subjects. mRNAs for ~50 ion and gap junction channels, Ca(2+)-handling proteins and markers were measured in the atrial muscle (AM), a transitional area (TA), inferior nodal extension (INE), compact node (CN), penetrating bundle (PB) and ventricular muscle (VM). When compared to the AM, we found a lower expression of Na(v)1.5, K(ir)2.1, Cx43 and ANP mRNAs in the CN for example, but a higher expression of HCN1, HCN4, Ca(v)1.3, Ca(v)3.1, K(ir)3.4, Cx40 and Tbx3 mRNAs. Expression of some related proteins was in agreement with the expression of the corresponding mRNAs. There is a complex and heterogeneous pattern of expression of ion and gap junction channels and Ca(2+)-handling proteins in the human atrioventricular conduction axis that explains the function of this crucial pathway.


Assuntos
Nó Atrioventricular/citologia , Nó Atrioventricular/metabolismo , Sistema de Condução Cardíaco/citologia , Sistema de Condução Cardíaco/metabolismo , Arritmias Cardíacas/metabolismo , Canais de Cálcio Tipo T/metabolismo , Caveolina 3/metabolismo , Conexina 43/metabolismo , Conexinas/metabolismo , Eletrofisiologia , Junções Comunicantes/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Canais Iônicos/metabolismo , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Sódio/metabolismo
11.
J Mol Cell Cardiol ; 48(4): 653-62, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19729016

RESUMO

During ageing, the function of sinoatrial node (SAN), the pacemaker of the heart, declines, and the incidence of sick sinus syndrome increases markedly. The aim of the study was to investigate structural and functional remodelling of the SAN during ageing. Rats, 3 and 24 months old (equivalent to young adult and approximately 69-year-old humans), were studied. Extracellular potential recording from right atrial preparations showed that (as expected) the intrinsic heart rate was slower in the old animals. It also showed a shift of the leading pacemaker site towards the inferior vena cava in the old animals. Consistent with this, intracellular potential recording showed that slow pacemaker action potentials were more widespread and extended further towards the inferior vena cava in old animals. Immunohistochemistry demonstrated that SAN tissue expressing HCN4, but lacking the expression of Na(v)1.5 (lack of Na(v)1.5 explains why pacemaker action potential is slow), was also more widespread and extended further towards the inferior vena cava in the old animals. Immunolabelling of caveolin3 (expressed in cell membrane of cardiac myocytes) demonstrated that there was a hypertrophy of the SAN cells in the old animals. Histology, quantitative PCR, and immunohistochemistry revealed evidence of a substantial age-dependent remodelling of the extracellular matrix (e.g. approximately 79% downregulation of genes responsible for collagens 1 and 3 and approximately 52% downregulation of gene responsible for elastin). It is concluded that the age- (and/or obesity-) dependent decline in SAN function is associated with a structural remodelling of the SAN: an enlargement of the SAN, a hypertrophy of the SAN cells, and a remodelling of the extracellular matrix.


Assuntos
Obesidade/fisiopatologia , Nó Sinoatrial/patologia , Envelhecimento , Animais , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Imuno-Histoquímica/métodos , Masculino , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Potássio/metabolismo , Ratos , Ratos Wistar , Canais de Sódio/metabolismo , Fatores de Tempo , Veia Cava Inferior/patologia
12.
Biophys J ; 97(8): 2117-27, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19843444

RESUMO

Mathematical models are a repository of knowledge as well as research and teaching tools. Although action potential models have been developed for most regions of the heart, there is no model for the atrioventricular node (AVN). We have developed action potential models for single atrio-nodal, nodal, and nodal-His cells. The models have the same action potential shapes and refractoriness as observed in experiments. Using these models, together with models for the sinoatrial node (SAN) and atrial muscle, we have developed a one-dimensional (1D) multicellular model including the SAN and AVN. The multicellular model has slow and fast pathways into the AVN and using it we have analyzed the rich behavior of the AVN. Under normal conditions, action potentials were initiated in the SAN center and then propagated through the atrium and AVN. The relationship between the AVN conduction time and the timing of a premature stimulus (conduction curve) is consistent with experimental data. After premature stimulation, atrioventricular nodal reentry could occur. After slow pathway ablation or block of the L-type Ca(2+) current, atrioventricular nodal reentry was abolished. During atrial fibrillation, the AVN limited the number of action potentials transmitted to the ventricle. In the absence of SAN pacemaking, the inferior nodal extension acted as the pacemaker. In conclusion, we have developed what we believe is the first detailed mathematical model of the AVN and it shows the typical physiological and pathophysiological characteristics of the tissue. The model can be used as a tool to analyze the complex structure and behavior of the AVN.


Assuntos
Potenciais de Ação , Nó Atrioventricular/fisiologia , Fascículo Atrioventricular/fisiologia , Modelos Cardiovasculares , Neurônios/fisiologia , Animais , Fibrilação Atrial/fisiopatologia , Nó Atrioventricular/fisiopatologia , Relógios Biológicos/fisiologia , Fascículo Atrioventricular/fisiopatologia , Canais de Cálcio Tipo L/metabolismo , Potenciais da Membrana , Condução Nervosa , Vias Neurais/fisiologia , Vias Neurais/fisiopatologia , Coelhos , Fatores de Tempo
13.
Chaos ; 19(2): 028509, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19566284

RESUMO

Fluctuations in the time interval between two consecutive R-waves of electrocardiogram during normal sinus rhythm may result from irregularities in the autonomic drive of the pacemaking sinoatrial node (SAN). We use a biophysically detailed mathematical model of the action potentials of rabbit SAN to quantify the effects of fluctuations in acetylcholine (ACh) on the pacemaker activity of the SAN and its variability. Fluctuations in ACh concentration model the effect of stochastic activity in the vagal parasympathetic fibers that innervate the SAN and produce varying rates of depolarization during the pacemaker potential, leading to fluctuations in cycle length (CL). Both the estimated maximal Lyapunov exponent and the noise limit of the resultant sequence of fluctuating CLs suggest chaotic dynamics. Apparently chaotic heart rate variability (HRV) seen in sinus rhythm can be produced by stochastic modulation of the SAN. The identification of HRV data as chaotic by use of time series measures such as a positive maximal Lyapunov exponent or positive noise limit requires both caution and a quantitative, predictive mechanistic model that is fully deterministic.


Assuntos
Frequência Cardíaca/fisiologia , Modelos Cardiovasculares , Nó Sinoatrial/fisiologia , Nervo Vago/fisiologia , Acetilcolina/fisiologia , Potenciais de Ação , Animais , Fenômenos Biofísicos , Dinâmica não Linear , Coelhos , Processos Estocásticos
15.
Mol Cell Biochem ; 319(1-2): 105-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18629610

RESUMO

Abnormal QT prolongation with the associated arrhythmias is a significant predictor of mortality in diabetic patients. Gap junctional intercellular communication allows electrical coupling between heart muscle cells. The effects of streptozotocin (STZ)-induced diabetes mellitus on the expression and distribution of connexin 43 (Cx43) in ventricular muscle have been investigated. Cx43 mRNA expression was measured in ventricular muscle by quantitative PCR. The distribution of total Cx43, phosphorylated Cx43 (at serine 368) and non-phosphorylated Cx43 was measured in ventricular myocytes and ventricular muscle by immunocytochemistry and confocal microscopy. There was no significant difference in Cx43 mRNA between diabetic rat ventricle and controls. Total and phosphorylated Cx43 were significantly increased in ventricular myocytes and ventricular muscle and dephosphorylated Cx43 was not significantly altered in ventricular muscle from diabetic rat hearts compared to controls. Disturbances in gap junctional intercellular communication, which in turn may be attributed to alterations in balance between total, phosphorylated and dephosporylated Cx43, might partly underlie prolongation of QRS and QT intervals in diabetic heart.


Assuntos
Conexina 43/biossíntese , Diabetes Mellitus Experimental/metabolismo , Regulação da Expressão Gênica , Proteínas Musculares/biossíntese , Miocárdio/metabolismo , RNA Mensageiro/biossíntese , Animais , Diabetes Mellitus Experimental/patologia , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Masculino , Miocárdio/patologia , Fosforilação , Ratos , Ratos Wistar
18.
Am J Physiol Heart Circ Physiol ; 292(4): H1737-46, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17012364

RESUMO

In the heart, ACh activates the ACh-activated K(+) current (I(K,ACh)) via the M(2) muscarinic receptor. The relationship between desensitization of I(K,ACh) and internalization of the M(2) receptor has been studied in rat atrial cells. On application of the stable muscarinic agonist carbachol for 2 h, I(K,ACh) declined by approximately 62% with time constants of 1.5 and 26.9 min, whereas approximately 83% of the M(2) receptor was internalized from the cell membrane with time constants of 2.9 and 51.6 min. Transfection of the cells with beta-adrenergic receptor kinase 1 (G protein-receptor kinase 2) and beta-arrestin 2 significantly increased I(K,ACh) desensitization and M(2) receptor internalization during a 3-min application of agonist. Internalized M(2) receptor in cells exposed to carbachol for 2 h was colocalized with clathrin and not caveolin. It is concluded that a G protein-receptor kinase 2- and beta-arrestin 2-dependent internalization of the M(2) receptor into clathrin-coated vesicles could play a major role in I(K,ACh) desensitization.


Assuntos
Vesículas Revestidas por Clatrina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Coração/fisiologia , Potássio/metabolismo , Receptor Muscarínico M2/metabolismo , Acetilcolina/farmacologia , Animais , Arrestinas/genética , Arrestinas/metabolismo , Carbacol/farmacologia , Caveolina 3/genética , Membrana Celular/metabolismo , Colinérgicos/farmacologia , Agonistas Colinérgicos/farmacologia , Endocitose/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G , Coração/inervação , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Ratos , Receptor Muscarínico M2/fisiologia , Transfecção , Nervo Vago/fisiologia , Quinases de Receptores Adrenérgicos beta/genética , Quinases de Receptores Adrenérgicos beta/metabolismo , beta-Arrestina 2 , beta-Arrestinas
19.
Am J Physiol Heart Circ Physiol ; 292(1): H165-74, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16877551

RESUMO

Voltage-dependent sodium (Na(+)) channels are heterogeneously distributed through the pacemaker of the heart, the sinoatrial node (SA node). The measured sodium channel current (i(Na)) density is higher in the periphery but low or zero in the center of the SA node. The functional roles of i(Na) in initiation and conduction of cardiac pacemaker activity remain uncertain. We evaluated the functional roles of i(Na) by computer modeling. A gradient model of the intact SA node and atrium of the rabbit heart was developed that incorporates both heterogeneities of the SA node electrophysiology and histological structure. Our computations show that a large i(Na) in the periphery helps the SA node to drive the atrial muscle. Removal i(Na) from the SA node slows down the pacemaking rate and increases the sinoatrial node-atrium conduction time. In some cases, reduction of the SA node i(Na) results in impairment of impulse initiation and conduction that leads to the SA node-atrium conduction exit block. Decrease in active SA node cell population has similar effects. Combined actions of reduced cell population and removal of i(Na) from the SA node have greater impacts on weakening the ability of the SA node to pace and drive the atrium.


Assuntos
Potenciais de Ação/fisiologia , Relógios Biológicos/fisiologia , Ativação do Canal Iônico/fisiologia , Modelos Cardiovasculares , Nó Sinoatrial/fisiologia , Canais de Sódio/fisiologia , Sódio/metabolismo , Animais , Simulação por Computador , Humanos
20.
J Mol Cell Cardiol ; 41(5): 855-67, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16996082

RESUMO

Kir2.1 and Kir6.2 are ion channel subunits partly responsible for the background inward rectifier and ATP-sensitive K(+) currents (I(K1) and I(KATP)) in the heart. Very little is known about how the distribution of ion channel subunits is controlled. In this study, we have investigated the expression (at protein and mRNA levels) of GFP-tagged Kir2.1 and Kir6.2 transgenes under the control of the alpha-MHC promoter in the sinoatrial node (SAN), atrioventricular node (AVN), His bundle and working myocardium of transgenic mice. After dissection, serial 10-microm cryosections were cut. Histological staining was carried out to identify tissues, confocal microscopy was carried out to map the distribution of the GFP-tagged ion channel subunits and in situ hybridization was carried out to map the distribution of corresponding mRNAs. We demonstrate heterologous expression of the ion channel subunits in the working myocardium, but not necessarily in the SAN, AVN or His bundle; the distribution of the subunits does not correspond to the expected distribution of alpha-MHC. Both protein and mRNA expression does, however, correspond to the expected distributions of native Kir6.2 and Kir2.1 in the SAN, AVN, His bundle and working myocardium. The data demonstrate novel transcriptional and/or post-transcriptional control of ion channel subunit expression and raise important questions about the control of regional expression of ion channels.


Assuntos
Nó Atrioventricular/metabolismo , Cadeias Pesadas de Miosina/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Nó Sinoatrial/metabolismo , Animais , Relógios Biológicos , Regulação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Cadeias Pesadas de Miosina/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Regiões Promotoras Genéticas , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...