Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Clin Exp Pharmacol Physiol ; 42(8): 874-80, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25933122

RESUMO

In critically ill patients regulation of heart-rate is often severely disturbed. Interaction of bacterial endotoxin (lipopolysaccharide, LPS) with hyperpolarization-activated cyclic nucleotide-gated cation-(HCN)-channels may interfere with heart-rate regulation. This study analyzes the effect of LPS, the HCN-channel blocker ivabradine or Ca(2+) -channel blockers (nifedipine, verapamil) on pacemaking in spontaneously beating neonatal rat cardiomyocytes (CM) in vitro. In vivo, the effect of LPS on the heart-rate of adult CD1-mice with and without autonomic blockade is analyzed telemetrically. LPS (100 ng/mL) and ivabradine (5 µg/mL) reduced the beating-rate of CM by 20.1% and 24.6%, respectively. Coincubation of CM with both, LPS and ivabradine, did not further reduce the beating-rate, indicating interaction of both compounds with HCN-channels, while coincubation with Ca(2+) -channel blockers and LPS caused additive beating-rate reduction. In CD1-mice (containing an active autonomic-nervous-system), injection of LPS (0.4 mg/kg) expectedly resulted in increased heart-rate. However, if the autonomic nervous system was blocked by propranolol and atropine, in line with the in vitro data, LPS induced a significant reduction of heart-rate, which was not additive to ivabradine. The in vivo and in vitro results indicate that LPS interacts with HCN-channels of cardiomyocytes. Thus, LPS indirectly sensitizes HCN-channels for sympathetic activation (tachycardic-effect), and in parallel directly inhibits channel activity (bradycardic-effect). Both effects may contribute to the detrimental effects of septic cardiomyopathy and septic autonomic dysfunction.


Assuntos
Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiologia , Animais , Benzazepinas/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Ivabradina , Masculino , Camundongos , Ratos , Sistema Nervoso Simpático/fisiopatologia , Taquicardia/induzido quimicamente , Taquicardia/metabolismo , Taquicardia/fisiopatologia
2.
Basic Res Cardiol ; 108(1): 312, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23184392

RESUMO

Common cardiovascular progenitor cells are characterized and induced by expression of the transcription factor MesP1. To characterize this population we used a 3.4-kb promoter fragment previously described by our group. This served to isolate MesP1-positive cells from differentiating ES stem cells via magnetic cell sorting based on a truncated CD4 surface marker. As this proximal promoter fragment omits a distal non-cardiovasculogenic enhancer region, we were able to achieve a synchronized fraction of highly enriched cardiovascular progenitors. These led to about 90% of cells representing the three cardiovascular lineages: cardiomyocytes, endothelial cells and smooth muscle cells as evident from protein and mRNA analyses. In addition, electrophysiological and pharmacological parameters of the cardiomyocytic fraction show that almost all correspond to the multipotent early/intermediate cardiomyocyte subtype at day 18 of differentiation. Further differentiation of these cells was not impaired as evident from strong and synchronous beating at later stages. Our work contributes to the understanding of the earliest cardiovasculogenic events and may become an important prerequisite for cell therapy, tissue engineering and pharmacological testing in the culture dish using pluripotent stem cell-derived as well as directly reprogrammed cardiovascular cell types. Likewise, these cells provide an ideal source for large-scale transcriptome and proteome analyses.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sistema Cardiovascular/citologia , Células-Tronco Multipotentes/citologia , Regiões Promotoras Genéticas , Animais , Diferenciação Celular , Separação Celular , Células Endoteliais/citologia , Camundongos , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia
3.
Circ Res ; 109(9): 1015-23, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21903939

RESUMO

RATIONALE: The hyperpolarization-activated current I(h) that is generated by hyperpolarization-activated cyclic nucleotide-gated channels (HCNs) plays a key role in the control of pacemaker activity in sinoatrial node cells of the heart. By contrast, it is unclear whether I(h) is also relevant for normal function of cardiac ventricles. OBJECTIVE: To study the role of the HCN3-mediated component of ventricular I(h) in normal ventricular function. METHODS AND RESULTS: To test the hypothesis that HCN3 regulates the ventricular action potential waveform, we have generated and analyzed a HCN3-deficient mouse line. At basal heart rate, mice deficient for HCN3 displayed a profound increase in the T-wave amplitude in telemetric electrocardiographic measurements. Action potential recordings on isolated ventricular myocytes indicate that this effect was caused by an acceleration of the late repolarization phase in epicardial myocytes. Furthermore, the resting membrane potential was shifted to more hyperpolarized potentials in HCN3-deficient mice. Cardiomyocytes of HCN3-deficient mice displayed approximately 30% reduction of total I(h). At physiological ionic conditions, the HCN3-mediated current had a reversal potential of approximately -35 mV and displayed ultraslow deactivation kinetics. CONCLUSIONS: We propose that HCN3 together with other members of the HCN channel family confer a depolarizing background current that regulates ventricular resting potential and counteracts the action of hyperpolarizing potassium currents in late repolarization. In conclusion, our data indicate that HCN3 plays an important role in shaping the cardiac action potential waveform.


Assuntos
Potenciais de Ação/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Função Ventricular/fisiologia , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/deficiência , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Eletrocardiografia , Frequência Cardíaca/fisiologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Miócitos Cardíacos/fisiologia , Canais de Potássio , Nó Sinoatrial/fisiologia
4.
EMBO J ; 26(21): 4423-32, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17914461

RESUMO

Cardiac pacemaking involves a variety of ion channels, but their relative importance is controversial and remains to be determined. Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, which underlie the I(f) current of sinoatrial cells, are thought to be key players in cardiac automaticity. In addition, the increase in heart rate following beta-adrenergic stimulation has been attributed to the cAMP-mediated enhancement of HCN channel activity. We have now studied mice in which the predominant sinoatrial HCN channel isoform HCN4 was deleted in a temporally controlled manner. Here, we show that deletion of HCN4 in adult mice eliminates most of sinoatrial I(f) and results in a cardiac arrhythmia characterized by recurrent sinus pauses. However, the mutants show no impairment in heart rate acceleration during sympathetic stimulation. Our results reveal that unexpectedly the channel does not play a role for the increase of the heart rate; however, HCN4 is necessary for maintaining a stable cardiac rhythm, especially during the transition from stimulated to basal cardiac states.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Regulação da Expressão Gênica , Frequência Cardíaca , Animais , AMP Cíclico/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Isoproterenol/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Isoformas de Proteínas , Nó Sinoatrial/metabolismo , Nó Sinoatrial/patologia
5.
Prog Biophys Mol Biol ; 98(2-3): 179-85, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19351513

RESUMO

Pacemaker activity of the heart is generated by a small group of cells forming the sinoatrial node (SAN). Cells of the SAN are spontaneously active and generate action potentials with remarkable regularity and stability under all physiological conditions. The exact molecular mechanisms underlying pacemaker potentials in the SAN have not yet been fully elucidated. Several voltage-dependent ion channels as well as intracellular calcium cycling processes are thought to contribute to the pacemaker activity. Hyperpolarization-activated cation channels, which generate the I(f) current, have biophysical properties which seem ideally suited for the initiation of spontaneous electrical activity. This review describes recent work on several transgenic mice lacking different cardiac HCN channel subtypes. The role of I(f) for normal pacemaking and sinus node arrhythmia as revealed by these genetic models will be discussed. In addition, a new mouse line is described which enables gene targeting in a temporally-controlled manner selectively in SAN cells. Elucidating the function of HCN and other ion channels in well-controlled mouse models should ultimately lead to a better understanding of the mechanisms underlying human sinoatrial arrhythmias.


Assuntos
Arritmia Sinusal/fisiopatologia , Canais Iônicos/fisiologia , Animais , Arritmia Sinusal/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/deficiência , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/fisiologia , Modelos Animais de Doenças , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/deficiência , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos
6.
J Mol Cell Cardiol ; 45(1): 62-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18538341

RESUMO

Temporally controlled gene deletion provides a powerful technique for examination of gene function in vivo. To permit use of this technology in the study of cardiac pacemaking, we attempted to generate a mouse line expressing an inducible Cre recombinase selectively in cardiac pacemaker cells. The tamoxifen-inducible CreER(T2) construct was 'knocked in' into the pacemaker channel HCN4 locus. In the absence of inducing agent, recombination was undetectable in HCN4-KiT mice. After injection of tamoxifen, highly selective and efficient recombination was observed in the sinoatrial and atrioventricular node. Expression of Cre and tamoxifen per se did not affect cardiac rhythm, basal heart rate and heart rate modulation. By crossing these animals with floxed HCN4 mice, complete deletion of this gene in the sinoatrial node could be achieved. HCN4-KiT mice represent the first tool for the temporally controlled inactivation of floxed target genes selectively in the conduction system of the murine heart.


Assuntos
Antagonistas de Estrogênios/farmacologia , Deleção de Genes , Sistema de Condução Cardíaco/metabolismo , Integrases/biossíntese , Recombinação Genética/efeitos dos fármacos , Tamoxifeno/farmacologia , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Integrases/genética , Camundongos , Camundongos Transgênicos , Locos de Características Quantitativas/genética , Recombinação Genética/genética
7.
Mol Endocrinol ; 21(3): 753-64, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17158221

RESUMO

Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels mediate the pacemaker current (Ih or If) observed in electrically rhythmic cardiac and neuronal cells. Here we describe a hyperpolarization-activated time-dependent cationic current, beta-Ih, in pancreatic beta-cells. Transcripts for HCN1-4 were detected by RT-PCR and quantitative PCR in rat islets and MIN6 mouse insulinoma cells. beta-Ih in rat beta-cells and MIN6 cells displayed biophysical and pharmacological properties similar to those of HCN currents in cardiac and neuronal cells. Stimulation of cAMP production with forskolin/3-isobutyl-1-methylxanthine (50 microM) or dibutyryl-cAMP (1 mM) caused a significant rightward shift in the midpoint activation potential of beta-Ih, whereas expression of either specific small interfering (si)RNA against HCN2 (siHCN2b) or a dominant-negative HCN channel (HCN1-AAA) caused a near-complete inhibition of time-dependent beta-Ih. However, expression of siHCN2b in MIN6 cells had no affect on glucose-stimulated insulin secretion under normal or cAMP-stimulated conditions. Blocking beta-Ih in intact rat islets also did not affect membrane potential behavior at basal glucose concentrations. Taken together, our experiments provide the first evidence for functional expression of HCN channels in the pancreatic beta-cell.


Assuntos
Células Secretoras de Insulina/metabolismo , Bloqueadores dos Canais de Potássio/metabolismo , Canais de Potássio/metabolismo , Animais , Benzazepinas/farmacologia , Células Cultivadas , AMP Cíclico/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Eletrofisiologia , Exocitose/efeitos dos fármacos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Insulinoma/patologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Piperidinas/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos
8.
Methods Find Exp Clin Pharmacol ; 30(8): 633-41, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19088948

RESUMO

Ivabradine has been approved as a heart rate-lowering agent for use in the treatment of chronic stable angina pectoris in case of contraindication or intolerance to beta-blockers. This drug effectively lowers the heart rate by inhibiting the pacemaker current I(f) in the sinoatrial node. It appears to induce fewer adverse reactions than other drugs used for reducing the heart rate, such as calcium channel blockers or beta-blockers. Because of this favorable profile, ivabradine could become the first-choice drug when pure heart rate-lowering is the therapeutic goal. This review evaluates experimental and preclinical data to investigate the possibilities, as well as the limitations, of the clinical use of ivabradine. In experimental studies, it has been shown that ivabradine does have some unfavorable pharmacodynamic properties, such as the block of all four hyperpolarization-activated cyclic nucleotide-gated channels and block of other ion channels at high concentrations. Clinical studies, however, indicate that those properties do not result in clinical consequences as long as ivabradine is given at the recommended dose and contraindications are strictly observed.


Assuntos
Benzazepinas/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Angina Pectoris/tratamento farmacológico , Angina Pectoris/fisiopatologia , Animais , Benzazepinas/efeitos adversos , Benzazepinas/uso terapêutico , Doença Crônica , Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Humanos , Canais Iônicos/antagonistas & inibidores , Ivabradina
9.
Pain ; 158(10): 2012-2024, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28767511

RESUMO

Several studies implicated cyclic adenosine monophosphate (cAMP) as an important second messenger for regulating nociceptor sensitization, but downstream targets of this signaling pathway which contribute to neuronal plasticity are not well understood. We used a Cre/loxP-based strategy to disable the function of either HCN2 or PKA selectively in a subset of peripheral nociceptive neurons and analyzed the nociceptive responses in both transgenic lines. A near-complete lack of sensitization was observed in both mutant strains when peripheral inflammation was induced by an intradermal injection of 8br-cAMP. The lack of HCN2 as well as the inhibition of PKA eliminated the cAMP-mediated increase of calcium transients in dorsal root ganglion neurons. Facilitation of Ih via cAMP, a hallmark of the Ih current, was abolished in neurons without PKA activity. Collectively, these results show a significant contribution of both genes to inflammatory pain and suggest that PKA-dependent activation of HCN2 underlies cAMP-triggered neuronal sensitization.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Canais de Potássio/metabolismo , Células Receptoras Sensoriais/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Bradicinina/farmacologia , Cálcio/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Gânglios Espinais/citologia , Hiperalgesia/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Inflamação/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Limiar da Dor , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Canais de Potássio/genética , Proteínas/genética , Proteínas/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais
10.
Trends Cardiovasc Med ; 14(1): 23-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14720471

RESUMO

Cardiac pacemaker activity is regulated by at least five different classes of ion channels and by the opposing influence of sympathetic and parasympathetic stimulation. Inactivation of several genes, including a subunit coding for the potassium channel activated by the muscarinic receptor, I(KACh); the calcium channel, I(Ca,); and the hyperpolarization-activated channel, I(f), results in sinus node arrhythmia. Inactivation of the gene for the hyperpolarization-activated, cyclic nucleotide-gated channel isoform HCN2 or HCN4 and the use of pacemaker channel blockers show that (a) HCN2 prevents the diastolic membrane potential from becoming too negative, (b) HCN4 is the major channel mediating sympathetic stimulation of the pacemaker activity, and (3) complete blockage of the I(f) current is compatible with slow sinus node rhythm.


Assuntos
Potenciais de Ação , Frequência Cardíaca/genética , Canais Iônicos/metabolismo , Nó Sinoatrial/fisiologia , Animais , Arritmia Sinusal/genética , Arritmia Sinusal/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Eletrofisiologia , Frequência Cardíaca/fisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Canais de Potássio , Coelhos , Fatores de Risco , Sensibilidade e Especificidade
11.
Stem Cell Reports ; 2(5): 592-605, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24936448

RESUMO

Therapeutic approaches for "sick sinus syndrome" rely on electrical pacemakers, which lack hormone responsiveness and bear hazards such as infection and battery failure. These issues may be overcome via "biological pacemakers" derived from pluripotent stem cells (PSCs). Here, we show that forward programming of PSCs with the nodal cell inducer TBX3 plus an additional Myh6-promoter-based antibiotic selection leads to cardiomyocyte aggregates consisting of >80% physiologically and pharmacologically functional pacemaker cells. These induced sinoatrial bodies (iSABs) exhibited highly increased beating rates (300-400 bpm), coming close to those found in mouse hearts, and were able to robustly pace myocardium ex vivo. Our study introduces iSABs as highly pure, functional nodal tissue that is derived from PSCs and may be important for future cell therapies and drug testing in vitro.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes/citologia , Nó Sinoatrial/fisiologia , Animais , Relógios Biológicos , Cálcio/metabolismo , Diferenciação Celular , Linhagem Celular , Técnicas de Cocultura , Técnicas In Vitro , Camundongos , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Técnicas de Patch-Clamp , Células-Tronco Pluripotentes/metabolismo , Síndrome do Nó Sinusal/metabolismo , Síndrome do Nó Sinusal/patologia , Síndrome do Nó Sinusal/veterinária , Nó Sinoatrial/citologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
12.
Cardiovasc Res ; 99(4): 780-8, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23761399

RESUMO

AIMS: The complex molecular mechanisms underlying spontaneous cardiac pacemaking are not fully understood. Recent findings point to a co-ordinated interplay between intracellular Ca(2+) cycling and plasma membrane-localized cation transport determining the origin and periodicity of pacemaker potentials. The sodium-calcium exchanger (NCX1) is a key sarcolemmal protein for the maintenance of calcium homeostasis in the heart. Here, we investigated the contribution of NCX1 to cardiac pacemaking. METHODS AND RESULTS: We used an inducible and sinoatrial node-specific Cre transgene to create micelacking NCX1 selectively in cells of the cardiac pacemaking and conduction system (cpNCX1KO). RT-PCR and immunolabeling experiments confirmed the precise tissue-specific and temporally controlled deletion. Ablation of NCX1 resulted in a progressive slowing of heart rate accompanied by severe arrhythmias. Isolated sinoatrial tissue strips displayed a significantly decreased and irregular contraction rate underpinning a disturbed intrinsic pacemaker activity. Mutant animals displayed a gradual increase in the heart-to-body weight ratio and developed ventricular dilatation; however, their ventricular contractile performance was not significantly affected. Pacemaker cells from cpNCX1KO showed no NCX1 activity in response to caffeine-induced Ca(2+) release, determined by Ca(2+) imaging. Regular spontaneous Ca(2+) discharges were frequently seen in control, but only sporadically in knockout (KO) cells. The majority of NCX1KO cells displayed an irregular and a significantly reduced frequency of spontaneous Ca(2+) signals. Furthermore, Ca(2+) transients measured during electrical field stimulation were of smaller magnitude and decelerated kinetics in KO cells. CONCLUSIONS: Our results establish NCX1 as a critical target for the proper function of cardiac pacemaking.


Assuntos
Frequência Cardíaca , Miocárdio/metabolismo , Trocador de Sódio e Cálcio/fisiologia , Animais , Arritmias Cardíacas/etiologia , Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nó Sinoatrial/metabolismo , Remodelação Ventricular
13.
Cardiovasc Res ; 95(3): 317-26, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22652004

RESUMO

AIMS: Cardiac hypertrophy is accompanied by reprogramming of gene expression, where the altered expression of ion channels decreases electrical stability and increases the risk of life-threatening arrhythmias. However, the underlying mechanisms are not fully understood. Here, we analysed the role of the depolarizing current I(f) which has been hypothesized to contribute to arrhythmogenesis in the hypertrophied ventricle. METHODS AND RESULTS: We used transverse aortic constriction in mice to induce ventricular hypertrophy. This resulted in an increased number of I(f) positive ventricular myocytes as well as a strongly enhanced and accelerated I(f) when compared with controls. Of the four HCN (hyperpolarization-activated cyclic nucleotide-gated channels) isoforms mediating I(f), HCN2 and HCN4 were the predominantly expressed subunits in healthy as well as hypertrophied hearts. Unexpectedly, only the HCN1 transcript was significantly upregulated in response to hypertrophy. However, the combined deletion of HCN2 and HCN4 disrupted ventricular I(f) completely. The lack of I(f) in hypertrophic double-knockouts resulted in a strong attenuation of pro-arrhythmogenic parameters characteristically observed in hypertrophic hearts. In particular, prolongation of the action potential was significantly decreased and lengthening of the QT interval was reduced. CONCLUSIONS: We suggest that the strongly increased HCN channel activity in hypertrophied myocytes prolongs the repolarization of the ventricular action potential and thereby may increase the arrhythmogenic potential. Our results provide for the first time a direct link between an upregulation of ventricular I(f) and a diminished repolarization reserve in cardiac hypertrophy.


Assuntos
Arritmias Cardíacas/etiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/complicações , Miócitos Cardíacos/metabolismo , Canais de Potássio/metabolismo , Remodelação Ventricular , Potenciais de Ação , Animais , Aorta/cirurgia , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/deficiência , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Modelos Animais de Doenças , Eletrocardiografia , Regulação da Expressão Gênica , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Canais Iônicos/genética , Canais Iônicos/metabolismo , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Canais de Potássio/deficiência , Canais de Potássio/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
14.
J Clin Invest ; 122(3): 1119-30, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22354168

RESUMO

Cardiac pacemaker cells create rhythmic pulses that control heart rate; pacemaker dysfunction is a prevalent disorder in the elderly, but little is known about the underlying molecular causes. Popeye domain containing (Popdc) genes encode membrane proteins with high expression levels in cardiac myocytes and specifically in the cardiac pacemaking and conduction system. Here, we report the phenotypic analysis of mice deficient in Popdc1 or Popdc2. ECG analysis revealed severe sinus node dysfunction when freely roaming mutant animals were subjected to physical or mental stress. In both mutants, bradyarrhythmia developed in an age-dependent manner. Furthermore, we found that the conserved Popeye domain functioned as a high-affinity cAMP-binding site. Popdc proteins interacted with the potassium channel TREK-1, which led to increased cell surface expression and enhanced current density, both of which were negatively modulated by cAMP. These data indicate that Popdc proteins have an important regulatory function in heart rate dynamics that is mediated, at least in part, through cAMP binding. Mice with mutant Popdc1 and Popdc2 alleles are therefore useful models for the dissection of the mechanisms causing pacemaker dysfunction and could aid in the development of strategies for therapeutic intervention.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas Musculares/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Relógios Biológicos , Bradicardia/genética , Eletrocardiografia/métodos , Eletrofisiologia/métodos , Frequência Cardíaca , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Estrutura Terciária de Proteína , Telemetria/métodos , Fatores de Tempo
15.
Cardiovasc Res ; 84(2): 263-72, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19564151

RESUMO

AIMS: The proliferative potential of pluripotent stem cell-derived cardiomyocytes is limited, and reasonable yields for novel therapeutic options have yet to be achieved. In addition, various clinical applications will require the generation of specific cardiac cell types. Whereas early cardiovascular precursors appear to be important for novel approaches such as reseeding decellularized hearts, direct cell transplantation may require ventricular cells. Our recent work demonstrated that MesP1 represents a master regulator sufficient to induce cardiovasculogenesis in pluripotent cells. This led to our hypothesis that 'forward programming' towards specific subtypes may be feasible via overexpression of distinct early cardiovascular transcription factors. METHODS AND RESULTS: Here we demonstrate that forced expression of Nkx2.5 similar to MesP1 is sufficient to enhance cardiogenesis in murine embryonic stem cells (mES). In comparison to control transfected mES cells, a five-fold increased appearance of beating foci was observed as well as upregulated mRNA and protein expression levels. In contrast to MesP1, no increase of the endothelial lineage within the cardiovasculogenic mesoderm was observed. Likewise, Flk-1, the earliest known cardiovascular surface marker, was not induced via Nkx2.5 as opposed to MesP1. Detailed patch clamping analyses showed electrophysiological characteristics corresponding to all subtypes of cardiac ES cell differentiation in Nkx2.5 as well as MesP1 programmed embryoid bodies, but fractions of cardiomyocytes had distinct characteristics: MesP1 forced the appearance of early/intermediate type cardiomyocytes in comparison to control transfected ES cells whereas Nkx2.5 led to preferentially differentiated ventricular cells. CONCLUSION: Our findings show proof of principle for cardiovascular subtype-specific programming of pluripotent stem cells and confirm the molecular hierarchy for cardiovascular specification initiated via MesP1 with differentiation factors such as Nkx2.5 further downstream.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo , Potenciais de Ação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Contração Miocárdica/genética , Fenótipo , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/genética , Transfecção
16.
Pflugers Arch ; 454(4): 517-22, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17549513

RESUMO

Hyperpolarization-activated cation currents termed I (f/h) are observed in many neurons and cardiac cells. Four genes (HCN1-4) encode the channels underlying these currents. New insights into the pathophysiological significance of HCN channels have been gained recently from analyses of mice engineered to be deficient in HCN genes. Lack of individual subunits results in markedly different phenotypes. Disruption of HCN1 impairs motor learning but enhances spatial learning and memory. Deletion of HCN2 results in absence epilepsy, ataxia, and sinus node dysfunction. Mice lacking HCN4 die during embryonic development and develop no sinoatrial node-like action potentials. In the present review, we summarize the physiology and pathophysiology of HCN channel family members based primarily on information from the transgenic mouse models and on data from human patients carrying defects in HCN4 channels.


Assuntos
Canais Iônicos/fisiopatologia , Canais de Potássio/fisiologia , Animais , Encéfalo/fisiopatologia , Sistema Cardiovascular/fisiopatologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Regulação da Expressão Gênica , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/genética , Camundongos , Camundongos Transgênicos , Proteínas Musculares/genética , Neurônios/fisiologia , Canais de Potássio/genética , Transmissão Sináptica/fisiologia
17.
Mol Pharmacol ; 69(4): 1328-37, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16387796

RESUMO

Sinus node inhibitors reduce the heart rate presumably by blocking the pacemaker current If in the cardiac conduction system. This pacemaker current is carried by four hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels. We tested the potential subtype-specificity of the sinus node inhibitors cilobradine, ivabradine, and zatebradine using cloned HCN channels. All three substances blocked the slow inward current through human HCN1, HCN2, HCN3, and HCN4 channels. There was no subtype-specificity for the steady-state block, with mean IC50 values of 0.99, 2.25, and 1.96 microM for cilobradine, ivabradine, and zatebradine, respectively. Native If, recorded from mouse sinoatrial node cells, was slightly more efficiently blocked by cilobradine (IC50 value of 0.62 microM) than were the HCN currents. The block of I(f) in sinoatrial node cells resulted in slower and dysrhythmic spontaneous action potentials. The in vivo action of these blockers was analyzed using telemetric ECG recordings in mice. Each compound reduced the heart rate dose-dependently from 600 to 200 bpm with ED50 values of 1.2, 4.7, and 1.8 mg/kg for cilobradine, ivabradine, and zatebradine, respectively. beta-Adrenergic stimulation or forced physical activity only partly reversed this bradycardia. In addition to bradycardia, all three drugs induced increasing arrhythmia at concentrations greater than 5 mg/kg for cilobradine, greater than 10 mg/kg for zatebradine, or greater than 15 mg/kg for ivabradine. This dysrhythmic heart rate is characterized by periodic fluctuations of the duration between the T and P wave, resembling a form of sick sinus syndrome in humans. Hence, all available sinus node inhibitors possess an as-yet-unrecognized proarrhythmic potential.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Bradicardia/induzido quimicamente , Nó Sinoatrial/efeitos dos fármacos , Animais , Arritmias Cardíacas/fisiopatologia , Benzazepinas/farmacologia , Bradicardia/fisiopatologia , Cardiotônicos/farmacologia , Clonagem Molecular , Eletrocardiografia , Humanos , Ivabradina , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/farmacologia , Regulação para Cima
18.
J Biol Chem ; 280(41): 34635-43, 2005 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-16043489

RESUMO

Hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels underlie the inward pacemaker current, termed I(f)/I(h), in a variety of tissues. Many details are known for the HCN subtypes 1, 2, and 4. We now successfully cloned the cDNA for HCN3 from human brain and compared the electrophysiological properties of hHCN3 to the other three HCN subtypes. Overexpression of human HCN3 channels in HEK293 cells resulted in a functional channel protein. Similar to hHCN2 channels, hHCN3 channels are activated with a rather slow time constant of 1244 +/- 526 ms at -100 mV, which is a greater time constant than that of HCN1 but a smaller one than that of HCN4 channels. The membrane potential for half-maximal activation V((1/2)) was -77 +/- 5.4 mV, and the reversal potential E(rev) was -20.5 +/- 4 mV, resulting in a permeability ratio P(Na)/P(K) of 0.3. Like all other HCNs, hHCN3 was inhibited rapidly and reversibly by extracellular cesium and slowly and irreversibly by extracellular applied ZD7288. Surprisingly, the human HCN3 channel was not modulated by intracellular cAMP, a hallmark of the other known HCN channels. Sequence comparison revealed >80% homology of the transmembrane segments, the pore region, and the cyclic nucleotide binding domain of hHCN3 with the other HCN channels. The missing response to cAMP distinguishes human HCN3 from both the well cAMP responding HCN subtypes 2 and 4 and the weak responding subtype 1.


Assuntos
Regulação da Expressão Gênica , Canais de Potássio/biossíntese , Canais de Potássio/fisiologia , Northern Blotting , Western Blotting , Encéfalo/metabolismo , Cardiotônicos/farmacologia , Cátions , Linhagem Celular , Membrana Celular/metabolismo , Césio/química , Césio/farmacologia , Clonagem Molecular , AMP Cíclico/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos , DNA Complementar/metabolismo , Eletrofisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/química , Cinética , Potenciais da Membrana , Modelos Biológicos , Plasmídeos/metabolismo , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/química , Análise de Sequência de DNA , Fatores de Tempo , Transfecção
19.
J Biol Chem ; 278(36): 33672-80, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12813043

RESUMO

The pacemaker channels HCN2 and HCN4 have been identified in cardiac sino-atrial node cells. These channels differ considerably in several kinetic properties including the activation time constant (tau act), which is fast for HCN2 (144 ms at -140 mV) and slow for HCN4 (461 ms at -140 mV). Here, by analyzing HCN2/4 chimeras and mutants we identified single amino acid residues in transmembrane segments 1 and 2 and the connecting loop between S1 and S2 that are major determinants of this difference. Replacement of leucine 272 in S1 of HCN4 by the corresponding phenylalanine present in HCN2 decreased tau act of HCN4 to 149 ms. Conversely, activation of the fast channel HCN2 was decreased 3-fold upon the corresponding mutation of F221L in the S1 segment. Mutation of N291T and T293A in the linker between S1 and S2 of HCN4 shifted tau act to 275 ms. While residues 272, 291, and 293 of HCN4 affected the activation speed at basal conditions they had no obvious influence on the cAMP-dependent acceleration of activation kinetics. In contrast, mutation of I308M in S2 of HCN4 abolished the cAMP-dependent decrease in tau act. Surprisingly, this mutation also prevented the acceleration of channel activation observed after deletion of the C-terminal cAMP binding site. Taken together our results indicate that the speed of activation of the HCN4 channel is determined by structural elements present in the S1, S1-S2 linker, and the S2 segment.


Assuntos
Canais Iônicos/química , Proteínas Musculares/química , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , AMP Cíclico/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos , DNA Complementar/metabolismo , Eletrofisiologia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/metabolismo , Isoleucina/química , Cinética , Leucina/química , Modelos Químicos , Dados de Sequência Molecular , Proteínas Musculares/metabolismo , Mutação , Miocárdio/metabolismo , Fenilalanina/química , Canais de Potássio , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Homologia de Sequência de Aminoácidos , Transfecção , Proteínas tau/química
20.
Proc Natl Acad Sci U S A ; 100(25): 15235-40, 2003 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-14657344

RESUMO

Hyperpolarization-activated, cyclic nucleotide-gated cation currents, termed If or Ih, are generated by four members of the hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channel family. These currents have been proposed to contribute to several functions including pacemaker activity in heart and brain, control of resting potential, and neuronal plasticity. Transcripts of the HCN4 isoform have been found in cardiomyocytes and neurons, but the physiological role of this channel is unknown. Here we show that HCN4 is essential for the proper function of the developing cardiac conduction system. In wild-type embryos, HCN4 is highly expressed in the cardiac region where the early sinoatrial node develops. Mice lacking HCN4 channels globally, as well as mice with a selective deletion of HCN4 in cardiomyocytes, died between embryonic days 9.5 and 11.5. On average, If in cardiomyocytes from mutant embryos is reduced by 85%. Hearts from HCN4-deficient embryos contracted significantly slower compared with wild type and could not be stimulated by cAMP. In both wild-type and HCN4-/- mice, cardiac cells with "primitive" pacemaker action potentials could be found. However, cardiac cells with "mature" pacemaker potentials, observed in wild-type embryos starting at day 9.0, were not detected in HCN4-deficient embryos. Thus, HCN4 channels are essential for the proper generation of pacemaker potentials in the emerging sinoatrial node.


Assuntos
Potenciais de Ação , Canais Iônicos/fisiologia , Proteínas Musculares/fisiologia , Marca-Passo Artificial , Animais , Western Blotting , Fármacos Cardiovasculares/farmacologia , Cátions , AMP Cíclico/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Relação Dose-Resposta a Droga , Eletrofisiologia , Deleção de Genes , Frequência Cardíaca , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Imuno-Histoquímica , Hibridização In Situ , Canais Iônicos/metabolismo , Óperon Lac , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Proteínas Musculares/metabolismo , Miocárdio/citologia , Canais de Potássio , Isoformas de Proteínas , Pirimidinas/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nó Sinoatrial , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA