Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28167635

RESUMO

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Assuntos
Células-Tronco Embrionárias/transplante , Insuficiência Cardíaca/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Miócitos Cardíacos/transplante , Engenharia Tecidual/métodos , Remodelação Ventricular/fisiologia , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Insuficiência Cardíaca/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Miocárdio/citologia , Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Impressão Tridimensional , Ratos , Ratos Nus
2.
Circ Res ; 117(5): 401-12, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26078285

RESUMO

RATIONALE: Monitoring and controlling cardiac myocyte activity with optogenetic tools offer exciting possibilities for fundamental and translational cardiovascular research. Genetically encoded voltage indicators may be particularly attractive for minimal invasive and repeated assessments of cardiac excitation from the cellular to the whole heart level. OBJECTIVE: To test the hypothesis that cardiac myocyte-targeted voltage-sensitive fluorescence protein 2.3 (VSFP2.3) can be exploited as optogenetic tool for the monitoring of electric activity in isolated cardiac myocytes and the whole heart as well as function and maturity in induced pluripotent stem cell-derived cardiac myocytes. METHODS AND RESULTS: We first generated mice with cardiac myocyte-restricted expression of VSFP2.3 and demonstrated distinct localization of VSFP2.3 at the t-tubulus/junctional sarcoplasmic reticulum microdomain without any signs for associated pathologies (assessed by echocardiography, RNA-sequencing, and patch clamping). Optically recorded VSFP2.3 signals correlated well with membrane voltage measured simultaneously by patch clamping. The use of VSFP2.3 for human action potential recordings was confirmed by simulation of immature and mature action potentials in murine VSFP2.3 cardiac myocytes. Optical cardiograms could be monitored in whole hearts ex vivo and minimally invasively in vivo via fiber optics at physiological heart rate (10 Hz) and under pacing-induced arrhythmia. Finally, we reprogrammed tail-tip fibroblasts from transgenic mice and used the VSFP2.3 sensor for benchmarking functional and structural maturation in induced pluripotent stem cell-derived cardiac myocytes. CONCLUSIONS: We introduce a novel transgenic voltage-sensor model as a new method in cardiovascular research and provide proof of concept for its use in optogenetic sensing of physiological and pathological excitation in mature and immature cardiac myocytes in vitro and in vivo.


Assuntos
Potenciais da Membrana/fisiologia , Miócitos Cardíacos/fisiologia , Optogenética/métodos , Animais , Humanos , Camundongos , Camundongos Transgênicos , Imagens com Corantes Sensíveis à Voltagem/métodos
3.
J Physiol ; 591(17): 4087-97, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23732646

RESUMO

New antiarrhythmic drugs for treatment of atrial fibrillation should ideally be atrial selective in order to avoid pro-arrhythmic effects in the ventricles. Currently recognized atrial selective targets include atrial Nav1.5 channels, Kv1.5 channels and constitutively active Kir3.1/3.4 channels, each of which confers atrial selectivity by different mechanisms. Na(+) channel blockers with potential- and frequency-dependent action preferentially suppress atrial fibrillation because of the high excitation rate and less negative atrial resting potential, which promote drug binding in atria. Kv1.5 channels are truly atrial selective because they do not conduct repolarizing current IKur in ventricles. Constitutively active IK,ACh is predominantly observed in remodelled atria from patients in permanent atrial fibrillation (AF). A lot of effort has been invested to detect compounds which will selectively block Kir3.1/Kir3.4 in their remodelled constitutively active form. Novel drugs which have been and are being developed aim at atrial-selective targets. Vernakalant and ranolazine which mainly block atrial Na(+) channels are clinically effective. Newly designed selective IKur blockers and IK,ACh blockers are effective in animal models; however, clinical benefit in converting AF into sinus rhythm (SR) or reducing AF burden remains to be demonstrated. In conclusion, atrial-selective antiarrhythmic agents have a lot of potential, but a long way to go.


Assuntos
Antiarrítmicos/farmacologia , Função Atrial , Miocárdio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/tratamento farmacológico , Humanos , Especificidade de Órgãos , Canais de Potássio/metabolismo , Canais de Sódio/metabolismo
4.
J Physiol ; 591(17): 4189-206, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23878377

RESUMO

The species-specific determinants of repolarization are poorly understood. This study compared the contribution of various currents to cardiac repolarization in canine and human ventricle. Conventional microelectrode, whole-cell patch-clamp, molecular biological and mathematical modelling techniques were used. Selective IKr block (50-100 nmol l(-1) dofetilide) lengthened AP duration at 90% of repolarization (APD90) >3-fold more in human than dog, suggesting smaller repolarization reserve in humans. Selective IK1 block (10 µmol l(-1) BaCl2) and IKs block (1 µmol l(-1) HMR-1556) increased APD90 more in canine than human right ventricular papillary muscle. Ion current measurements in isolated cardiomyocytes showed that IK1 and IKs densities were 3- and 4.5-fold larger in dogs than humans, respectively. IKr density and kinetics were similar in human versus dog. ICa and Ito were respectively ~30% larger and ~29% smaller in human, and Na(+)-Ca(2+) exchange current was comparable. Cardiac mRNA levels for the main IK1 ion channel subunit Kir2.1 and the IKs accessory subunit minK were significantly lower, but mRNA expression of ERG and KvLQT1 (IKr and IKs α-subunits) were not significantly different, in human versus dog. Immunostaining suggested lower Kir2.1 and minK, and higher KvLQT1 protein expression in human versus canine cardiomyocytes. IK1 and IKs inhibition increased the APD-prolonging effect of IKr block more in dog (by 56% and 49%, respectively) than human (34 and 16%), indicating that both currents contribute to increased repolarization reserve in the dog. A mathematical model incorporating observed human-canine ion current differences confirmed the role of IK1 and IKs in repolarization reserve differences. Thus, humans show greater repolarization-delaying effects of IKr block than dogs, because of lower repolarization reserve contributions from IK1 and IKs, emphasizing species-specific determinants of repolarization and the limitations of animal models for human disease.


Assuntos
Potenciais de Ação , Coração/fisiologia , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia , Potássio/metabolismo , Adulto , Animais , Cálcio/metabolismo , Células Cultivadas , Cães , Feminino , Humanos , Transporte de Íons , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Especificidade da Espécie
5.
J Cardiovasc Pharmacol ; 61(5): 408-15, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23364608

RESUMO

The human electrophysiological and pharmacological properties of XEN-D0101 were evaluated to assess its usefulness for treating atrial fibrillation (AF). XEN-D0101 inhibited Kv1.5 with an IC50 of 241 nM and is selective over non-target cardiac ion channels (IC50 Kv4.3, 4.2 µM; hERG, 13 µM; activated Nav1.5, >100 µM; inactivated Nav1.5, 34 µM; Kir3.1/3.4, 17 µM; Kir2.1, >>100 µM). In atrial myocytes from patients in sinus rhythm (SR) and chronic AF, XEN-D0101 inhibited non-inactivating outward currents (Ilate) with IC50 of 410 and 280 nM, respectively, and peak outward currents (Ipeak) with IC50 of 806 and 240 nM, respectively. Whereas Ilate is mainly composed of IKur, Ipeak consists of IKur and Ito. Therefore, the effects on Ito alone were estimated from a double-pulse protocol where IKur was inactivated (3.5 µM IC50 in SR and 1 µM in AF). Thus, inhibition of Ipeak is because of IKur reduction and not Ito. XEN-D0101 significantly prolonged the atrial action potential duration at 20%, 50%, and 90% of repolarization (AF tissue only) and significantly elevated the atrial action potential plateau phase and increased contractility (SR and AF tissues) while having no effect on human ventricular action potentials. In healthy volunteers, XEN-D0101 did not significantly increase baseline- and placebo-adjusted QTc up to a maximum oral dose of 300 mg. XEN-D0101 is a Kv1.5/IKur inhibitor with an attractive atrial-selective profile.


Assuntos
Potenciais de Ação/fisiologia , Função Atrial/efeitos dos fármacos , Átrios do Coração/fisiopatologia , Canal de Potássio Kv1.5/antagonistas & inibidores , Miócitos Cardíacos/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Pirimidinas/farmacologia , Tiofenos/farmacologia , Função Ventricular/fisiologia , Fibrilação Atrial/tratamento farmacológico , Linhagem Celular , Método Duplo-Cego , Eletrocardiografia/efeitos dos fármacos , Átrios do Coração/efeitos dos fármacos , Humanos , Técnicas In Vitro , Masculino , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Potássio/farmacologia
6.
Can J Physiol Pharmacol ; 91(8): 648-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23889090

RESUMO

Dilated cardiomyopathy (DCM) is a multifactorial disease characterized by left ventricular dilation that is associated with systolic dysfunction and increased action potential duration. The Kir2.x K⁺ channels (encoded by KCNJ genes) regulate the inward rectifier current (IK1) contributing to the final repolarization in cardiac muscle. Here, we describe the transitions in the gene expression profiles of 4 KCNJ genes from healthy or dilated cardiomyopathic human hearts. In the healthy adult ventricles, KCNJ2, KCNJ12, and KCNJ4 (Kir2.1-2.3, respectively) genes were expressed at high levels, while expression of the KCNJ14 (Kir2.4) gene was low. In DCM ventricles, the levels of Kir2.1 and Kir2.3 were upregulated, but those of Kir2.2 channels were downregulated. Additionally, the expression of the DLG1 gene coding for the synapse-associated protein 97 (SAP97) anchoring molecule exhibited a 2-fold decline with increasing age in normal hearts, and it was robustly downregulated in young DCM patients. These adaptations could offer a new aspect for the explanation of the generally observed physiological and molecular alterations found in DCM.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Expressão Gênica , Ventrículos do Coração/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Adolescente , Adulto , Envelhecimento/genética , Western Blotting , Cardiomiopatia Dilatada/patologia , Feminino , Ventrículos do Coração/patologia , Humanos , Masculino , Potenciais da Membrana , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Isoformas de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
7.
J Mol Cell Cardiol ; 50(2): 357-66, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20971120

RESUMO

In cardiac regenerative therapy, transplantation of stem cells to form new myocardium is limited by their inability to integrate into host myocardium and conduct cardiac electrical activity. It is now hypothesized that refining cell sorting could upgrade the therapeutic result. Here we characterized a subpopulation of skeletal muscle stem cells with respect to their electrophysiological properties. The aim of our study was to determine whether electrophysiological parameters are compatible with cardiac function and can be influenced by culture conditions. Low-adherent skeletal muscle stem cells were isolated from the hind legs of 12-20 week old mice. After 6 days of culture the cells were analysed using patch-clamp techniques and RT-PCR, and replated in different media for skeletal muscle or cardiac differentiation. The cells generated action potentials (APs) longer than skeletal muscle APs, expressed functional cardiac Na(+) channels (~46% of the total channel fraction), displayed fast activating and inactivating L-type Ca(2+) currents, possibly conducted through cardiac channels and did not show significant Cl(-) conductance. Moreover, a fraction of cells expressed muscarinic acetylcholine receptors. Conditioning the cells for skeletal muscle differentiation resulted in upregulation of skeletal muscle-specific Na(+) and Ca(2+) channel expression, shortening of AP duration and loss of functional cardiac Na(+) channels. Cardiomyogenic conditions however, promoted the participation of cardiac Na(+) channels (57% of the total channel fraction). Nevertheless the cells retained properties of myoblasts such as the expression of nicotinic acetylcholine receptors. We conclude that skeletal muscle stem cells display several electrophysiological properties similar to those of cardiomyocytes. Culture conditions modulated these properties but only partially succeeded in further driving the cells towards a cardiac phenotype. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Assuntos
Fenômenos Eletrofisiológicos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Potenciais de Ação/fisiologia , Animais , Bário/metabolismo , Células Cultivadas , Meios de Cultura/farmacologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Colinérgicos/metabolismo , Canais de Sódio/metabolismo , Fatores de Tempo
8.
J Mol Cell Cardiol ; 51(3): 390-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21740909

RESUMO

Electrophysiological and pharmacological data from the human heart are limited due to the absence of simple but representative experimental model systems of human myocardium. The aim of this study was to establish and characterise adult human myocardial slices from small patients' heart biopsies as a simple, reproducible and relevant preparation suitable for the study of human cardiac tissue at the multicellular level. Vibratome-cut myocardial slices were prepared from left ventricular biopsies obtained from end-stage heart failure patients undergoing heart transplant or ventricular assist device implantation, and from hearts of normal dogs. Multiple slices were prepared from each biopsy. Regular contractility was observed at a range of stimulation frequencies (0.1-2 Hz), and stable electrical activity, monitored using multi-electrode arrays (MEA), was maintained for at least 8 h from slice preparation. ATP/ADP and phosphocreatine/creatine ratios were comparable to intact organ values, and morphology and gap junction distribution were representative of native myocardium. MEA recordings showed that field potential duration (FPD) and conduction velocity (CV) in human and dog slices were similar to the values previously reported for papillary muscles, ventricular wedges and whole hearts. Longitudinal CV was significantly faster than transversal CV, with an anisotropic ratio of 3:1 for human and 2.3:1 for dog slices. Importantly, slices responded to the application of E-4031, chromanol and 4-aminopyridine, three potassium channel blockers known to affect action potential duration, with an increase in FPD. We conclude that viable myocardial slices with preserved structural, biochemical and electrophysiological properties can be prepared from adult human and canine heart biopsies and offer a novel preparation suitable for the study of heart failure and drug screening.


Assuntos
Coração/efeitos dos fármacos , Coração/fisiologia , Adulto , Animais , Cães , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiologia , Humanos , Técnicas In Vitro , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miocárdio/patologia , Bloqueadores dos Canais de Potássio/farmacologia , Sobrevivência de Tecidos/fisiologia
9.
J Mol Cell Cardiol ; 48(1): 161-71, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19747484

RESUMO

The zebrafish has recently emerged as an excellent model for studies of heart development and regeneration. The physiology of the zebrafish heart has been suggested to resemble that of the human heart in many aspects, whereas, in contrast to mammals, the zebrafish has a remarkable ability to regenerate after heart injury. Thus, zebrafish have been proposed as a cost-effective model for genetic and pharmacological screens of factors affecting heart function and repair. However, realizing the full potential of the zebrafish heart as a model will require a better understanding of the electrophysiology of the adult zebrafish myocardium. Here, we characterize action potentials (APs) from intact adult atria and ventricles and find that the overall shape of zebrafish APs is similar to that of humans. We show that zebrafish, like most mammals, display functional acetylcholine-activated K(+) channels in the atrium, but not in the ventricle. Furthermore, the zebrafish AP upstroke is dominated by Na(+) channels, L-type Ca(2+) channels contribute to the plateau phase and I(Kr) channels are involved in repolarization. However, despite these similarities between zebrafish and mammalian electrophysiology, we also identified important differences. In particular, zebrafish display a robust T-type Ca(2+) current in both atrial and ventricular cardiomyocytes. Interestingly, in most mammals T-type Ca(2+) channels are only expressed in the developing heart or under pathophysiological conditions, indicating that adult zebrafish cardiomyocytes display a more immature phenotype.


Assuntos
Coração/fisiologia , Potenciais de Ação/fisiologia , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/metabolismo , Células Cultivadas , Eletrofisiologia , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Humanos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Nifedipino/farmacologia , Tetrodotoxina/farmacologia , Peixe-Zebra
10.
Pflugers Arch ; 460(1): 87-97, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20354865

RESUMO

DPP10 is a transmembrane glycosylated protein belonging to the family of dipeptidyl aminopeptidase-like proteins (DPPLs). DPPLs are auxiliary subunits involved in the regulation of voltage-gated Kv4 channels, key determinants of cardiac and neuronal excitability. Although it is known that DPPLs are needed to generate native-like currents in heterologous expression systems, the molecular basis of this involvement are still poorly defined. In this study, we investigated the functional relevance of DPP10 glycosylation in modulating Kv4.3 channel activities. Using transfected Chinese hamster ovary (CHO) cells to reconstitute Kv4 complex, we show that the pharmacological inhibition of DPP10 glycosylation by tunicamycin and neuraminidase affects transient outward potassium current (I (to)) kinetics. Tunicamycin completely blocked DPP10 glycosylation and reduced DPP10 cell surface expression. The accelerating effects of DPP10 on Kv4.3 current kinetics, i.e. on inactivation and recovery from inactivation, were abolished. Neuraminidase produced different effects on current kinetics than tunicamycin, i.e., shifted the voltage dependence to more negative potentials. The effects of tunicamycin on the native I (to) currents of human atrial myocytes expressing DPP10 were similar to those of the KV4.3/KChIP2/DPP10 complex in CHO cells. Our results suggest that N-linked glycosylation of DPP10 plays an important role in modulating Kv4 channel activities.


Assuntos
Membrana Celular/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Ativação do Canal Iônico , Proteínas Interatuantes com Canais de Kv/metabolismo , Potássio/metabolismo , Processamento de Proteína Pós-Traducional , Canais de Potássio Shal/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Glicosilação , Átrios do Coração/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Proteínas Interatuantes com Canais de Kv/genética , Potenciais da Membrana , Miócitos Cardíacos/metabolismo , Neuraminidase/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Transfecção , Tunicamicina/farmacologia
11.
Pflugers Arch ; 459(1): 11-23, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19768467

RESUMO

The cardiac action potential is primarily shaped by the orchestrated function of several different types of ion channels and transporters. One of the regional differences believed to play a major role in the progression and stability of the action potential is the transmural gradient of electrical activity across the ventricular wall. An altered balance in the ionic currents across the free wall is assumed to be a substrate for arrhythmia. A large fraction of patients with heart failure experience ventricular arrhythmia. However, the underlying substrate of these functional changes is not well-established as expression analyses of human heart failure (HF) are sparse. We have investigated steady-state RNA levels by quantitative polymerase chain reaction of ion channels, transporters, connexin 43, and miR-1 in 11 end-stage HF and seven nonfailing (NF) hearts. The quantifications were performed on endo-, mid-, and epicardium of left ventricle, enabling us to establish changes in the transmural expression gradient. Transcripts encoding Cav1.2, HCN2, Kir2.1, KCNE1, SUR1, and NCX1 were upregulated in HF compared to NF while a downregulation was observed for KChIP2, SERCA2, and miR-1. Additionally, the transmural gradient of KCNE1, KChIP2, Kir6.2, SUR1, Nav1.5, NCX1, and RyR2 found in NF was only preserved for KChiP2 and Nav1.5 in HF. The transmural gradients of NCX1, Nav1.5, and KChIP2 and the downregulation of KChIP2 were confirmed by Western blotting. In conclusion, our results reveal altered expression of several cardiac ion channels and transporters which may in part explain the increased susceptibility to arrhythmia in end-state failing hearts.


Assuntos
Expressão Gênica , Insuficiência Cardíaca/metabolismo , Coração/fisiologia , Canais Iônicos/biossíntese , Potenciais de Ação/fisiologia , Adolescente , Adulto , Western Blotting , Feminino , Humanos , Transporte de Íons , Masculino , Pessoa de Meia-Idade , Miocárdio/metabolismo , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Cell Physiol Biochem ; 24(5-6): 527-36, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19910693

RESUMO

AIM: Isolated papillary muscles and enzymatically dissociated myocytes of guinea-pig hearts are routinely used for experimental cardiac research. The aim of our study is to investigate adult mammalian ventricular slices as an alternative preparation. METHOD: Vibratome cut ventricular slices (350 microm thick) were examined histologically and with 2-photon microscopy for fibre orientation. Intracellular action potentials were recorded with conventional glass microelectrodes, extracellular potentials were measured with tungsten platinum electrodes and multi-electrode arrays (MEA). RESULTS: Dominant direction of fibre orientation was absent in vertical and horizontal transmural slices, but was longitudinal in tangential slices. Control action potential duration (APD(90), 169.9 +/- 4 ms) and drug effects on this parameter were similar to papillary muscles. The L-type Ca-channel blocker nifedipine shortened APD(90) with a half maximal effective concentration (EC(50)) of 4.5 microM. The I(Kr) blocker E4031 and neuroleptic drug risperidone prolonged APD(90) with EC(50) values of 31 nM and 0.67 microM, respectively. Mapping field potentials on multi-electrode arrays showed uniform spread of excitation with a mean conduction velocity of 0.47 m s(-1). CONCLUSION: Slices from adult mammalian hearts could become a useful routine model for electrophysiological and pharmacological research.


Assuntos
Coração/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Antipsicóticos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Avaliação Pré-Clínica de Medicamentos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Cobaias , Coração/anatomia & histologia , Coração/fisiologia , Humanos , Técnicas In Vitro , Masculino , Microeletrodos , Nifedipino/farmacologia , Músculos Papilares/efeitos dos fármacos , Músculos Papilares/fisiologia , Ratos , Ratos Wistar , Risperidona/farmacologia
13.
Int J Cardiol ; 284: 68-73, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30638748

RESUMO

BACKGROUND: Cardiac accessory ß-subunits are part of macromolecular Nav1.5 channel complexes modulating biophysical properties and contributing to arrhythmias. Recent studies demonstrated the structural interaction between ß-subunits of Na+ (Nav1.5) and K+ (Kv4.3) channels. Here, we identified the dipeptidyl peptidase-like protein-10 (DPP10), which is known to modulate Kv4.3-current kinetics, as a new regulator of Nav1.5 channels. METHODS: We assessed DPP10 expression in the healthy and diseased human heart and we studied the functional effects of DPP10 on the Na+ current in isolated rat cardiomyocytes expressing DPP10 after adenoviral gene-transfer (DPP10ad). RESULTS: DPP10 mRNA and proteins were detected in human ventricle, with higher levels in patients with heart failure. In rat cardiomyocytes, DPP10ad significantly reduced upstroke velocity of action potentials indicating reduction in Na+-current density. DPP10 significantly shifted the voltage-dependent Na+ channel activation and inactivation curve to more positive potentials, resulting in greater availability of Na+ channels for activation, along with increasing window Na+ current. In addition, time-to-peak Na+ current was reduced, whereas time course of recovery from inactivation was significantly accelerated by DPP10ad. DPP10 co-immunoprecipitated with Nav1.5 channels in human ventricles, confirming their physical interaction. CONCLUSION: We provide first evidence that DPP10 interacts with Nav1.5 channels, linking Na+- and K+-channel complexes in the heart. Our data suggest that increased ventricular DPP10 expression in heart failure might promote arrhythmias by decreasing peak Na+ current, while increasing window Na+ current and channel re-openings due to accelerated recovery from inactivation.


Assuntos
Arritmias Cardíacas/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Regulação da Expressão Gênica , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , RNA/genética , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Linhagem Celular , Cricetinae , Dipeptidil Peptidases e Tripeptidil Peptidases/biossíntese , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Miocárdio/patologia , Ratos , Ratos Wistar
14.
Naunyn Schmiedebergs Arch Pharmacol ; 376(6): 431-40, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18092154

RESUMO

Drugs that prolong the QT interval by blocking human ether-a-go-go (HERG) channels may enhance the risk of ventricular arrhythmia. The spasmolytic drug propiverine is widely used for the therapy of overactive bladder (OAB). Here, we have investigated the effects of propiverine on cardiac ion channels and action potentials as well as on contractile properties of cardiac tissue, in order to estimate its cardiac safety profile, because other drugs used in this indication had to be withdrawn due to safety reasons. Whole-cell patch clamp technique was used to record the following cardiac ion currents: rapidly and slowly activating delayed rectifier K+ current (I(Kr), I(Ks)), ultra rapidly activating delayed rectifier K+ current (I(Kur)), inwardly rectifying K+ current I(K1), transient outward K+ current (I(to)), and L-type Ca2+ current (I(Ca,L)). Action potentials in cardiac tissue biopsies were recorded with conventional microelectrodes. The torsade de pointes screening assay (TDPScreen) was used for drug scoring. Propiverine blocked in a concentration-dependent manner HERG channels expressed in HEK293 cells, as well as native I(Kr) current in ventricular myocytes of guinea pig (IC50 values: 10 microM and 1.8 microM respectively). At high concentrations (100 microM), propiverine suppressed I(Ks). I(K1) and the transient outward current I(to) and I(Kur) were not affected. In guinea-pig ventricular and human atrial myocytes, propiverine also blocked I(Ca,L) (IC50 values: 34.7 microM and 41.7 microM, respectively) and reduced force of contraction. Despite block of I(Kr), action potential duration was not prolonged in guinea-pig and human ventricular tissue, but decreased progressively until excitation failed altogether. Similar effects were observed in dog Purkinje fibers. Propiverine obtained a low score in the TDPScreen. In conclusion, in vitro and in vivo studies of propiverine do not provide evidence for an enhanced cardiovascular safety risk. We propose that lack of torsadogenic risk of propiverine is related to enhancement of repolarization reserve by block of I(Ca,L).


Assuntos
Benzilatos/efeitos adversos , Canais de Cálcio Tipo L/fisiologia , Contração Miocárdica/efeitos dos fármacos , Parassimpatolíticos/efeitos adversos , Canais de Potássio/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Benzilatos/farmacologia , Linhagem Celular , Bases de Dados Factuais , Cães , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/fisiologia , Cobaias , Humanos , Técnicas In Vitro , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Parassimpatolíticos/farmacologia , Técnicas de Patch-Clamp , Torsades de Pointes/induzido quimicamente
15.
Curr Opin Pharmacol ; 7(2): 214-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17303472

RESUMO

During atrial fibrillation, abnormal high-frequency activation induces 'remodeling' of the atrial myocardium, which includes changes in both electrical and structural properties. Substantial progress in understanding the molecular determinants underlying atrial fibrillation has led to the development of drugs that could enhance the long-term efficacy and safety of treating this common cardiac arrhythmia. New therapeutic approaches aim to prevent atrial remodelling, especially structural remodelling, improve currently used drugs, and design atria- and pathology-specific drugs without concomitant pro-arrhythmic effects in the ventricles. Considerable progress has been made in elucidating the molecular mechanisms of atrial fibrillation. However, a major improvement in the pharmacotherapy is still awaited. The promising efficacy of several new drugs in animal models of atrial fibrillation has yet to be demonstrated in clinical studies.


Assuntos
Antiarrítmicos/uso terapêutico , Fibrilação Atrial/tratamento farmacológico , Átrios do Coração/efeitos dos fármacos , Animais , Antiarrítmicos/efeitos adversos , Antiarrítmicos/farmacologia , Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Eletrofisiologia , Átrios do Coração/fisiopatologia , Humanos , Remodelação Ventricular
16.
Cardiovasc Res ; 74(3): 426-37, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17362895

RESUMO

OBJECTIVE: In chronic atrial fibrillation (cAF) the potassium current IK,ACh develops agonist-independent constitutive activity. We hypothesized that abnormal phosphorylation-dependent regulation underlies the constitutive IK,ACh activity. METHODS: We used voltage-clamp technique and biochemical assays to study IK,ACh regulation in atrial appendages from 61 sinus rhythm (SR), 11 paroxysmal AF (pAF), and 33 cAF patients. RESULTS: Compared to SR basal current was higher in cAF only, whereas the muscarinic receptor (2 micromol/L carbachol)-activated IK,ACh was smaller in pAF and cAF. In pAF the selective IK,ACh blocker tertiapin abolished the muscarinic receptor-activated IK,ACh but excluded agonist-independent constitutive IK,ACh activity. Blockade of type-2A phosphatase and the subsequent shift to increased muscarinic receptor phosphorylation (and inactivation) reduced muscarinic receptor-activated IK,ACh in SR but not in cAF, pointing to an impaired function of G-protein-coupled receptor kinase. Using subtype-selective kinase inhibitors we found that in SR the muscarinic receptor-activated IK,ACh requires phosphorylation by protein kinase G (PKG), protein kinase C (PKC), and calmodulin-dependent protein kinase II (CaMKII), but not by protein kinase A (PKA). In cAF, constitutive IK,ACh activity results from abnormal channel phosphorylation by PKC but not by PKG or CaMKII, whereas the additional muscarinic receptor-mediated IK,ACh activation occurs apparently without involvement of these kinases. In cAF, the higher protein level of PKCepsilon but not PKCalpha, PKCbeta1 or PKCdelta is likely to contribute to the constitutive IK,ACh activity. CONCLUSIONS: The occurrence of constitutive IK,ACh activity in cAF results from abnormal PKC function, whereas the muscarinic receptor-mediated IK,ACh activation does not require the contribution of PKG, PKC or CaMKII. Selective drug targeting of constitutively active IK,ACh channels may be suitable to reduce the ability of AF to become sustained.


Assuntos
Apêndice Atrial/metabolismo , Fibrilação Atrial/metabolismo , Ativação do Canal Iônico , Receptores Muscarínicos/metabolismo , Doença Aguda , Idoso , Análise de Variância , Fibrilação Atrial/genética , Venenos de Abelha/farmacologia , Western Blotting , Estudos de Casos e Controles , Células Cultivadas , Doença Crônica , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Isoenzimas/metabolismo , Masculino , Pessoa de Meia-Idade , Muscarina/metabolismo , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Polimorfismo Genético , Bloqueadores dos Canais de Potássio/farmacologia , Proteína Quinase C-épsilon/metabolismo
17.
Stem Cell Reports ; 10(3): 822-833, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29429959

RESUMO

Human induced pluripotent stem cell (hiPSC) cardiomyocytes (CMs) show less negative resting membrane potential (RMP), which is attributed to small inward rectifier currents (IK1). Here, IK1 was measured in hiPSC-CMs (proprietary and commercial cell line) cultured as monolayer (ML) or 3D engineered heart tissue (EHT) and, for direct comparison, in CMs from human right atrial (RA) and left ventricular (LV) tissue. RMP was measured in isolated cells and intact tissues. IK1 density in ML- and EHT-CMs from the proprietary line was similar to LV and RA, respectively. IK1 density in EHT-CMs from the commercial line was 2-fold smaller than in the proprietary line. RMP in EHT of both lines was similar to RA and LV. Repolarization fraction and IK,ACh response discriminated best between RA and LV and indicated predominantly ventricular phenotype in hiPSC-CMs/EHT. The data indicate that IK1 is not necessarily low in hiPSC-CMs, and technical issues may underlie low RMP in hiPSC-CMs.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Potássio/metabolismo , Átrios do Coração/metabolismo , Átrios do Coração/fisiopatologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Humanos
18.
Eur J Pharmacol ; 566(1-3): 222-5, 2007 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-17477915

RESUMO

We investigated the effects of hyperosmolar solutions on human ether-a-go-go related gene (hERG) potassium currents in chinese hamster ovary (CHO) cells. The addition of d-mannitol to the external solution caused cell shrinkage and reduced current amplitude. The effects were at least partially reversible. Exposure to 108 mM mannitol decreased current amplitude by 57+/-13%. Major effects on current-voltage relations were not observed. Exposure to 308 mM mannitol reduced the current by 89+/-5%, i.e. comparable to the block induced by 1 microM of the selective hERG channel blocker E-4031. We conclude that the investigation of hyperosmolar drug formulations requires control solutions of comparable osmolarity to separate specific drug effects from non-specific effects of hyperosmolarity.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Manitol/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Manitol/química , Concentração Osmolar , Técnicas de Patch-Clamp , Soluções
19.
Anesth Analg ; 105(3): 641-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17717217

RESUMO

BACKGROUND: Levosimendan is a novel drug used for inotropic support in heart failure, but its efficacy in local anesthetic-induced myocardial depression is not known. Therefore, we investigated the effects of levosimendan on the negative inotropic response to ropivacaine in isolated heart preparations of guinea pigs. METHODS: Action potentials and force of contraction were studied with conventional techniques in guinea-pig papillary muscles. Heart rate, systolic pressure, the first derivative of left ventricular pressure (+dP/dt(max)), coronary flow, and PR and QRS intervals were measured in isolated constant-pressure perfused, nonrecirculating Langendorff heart preparations. Single or cumulatively increasing concentrations of levosimendan and ropivacaine were used either alone or in combination. RESULTS: In isolated papillary muscle, ropivacaine reduced force of contraction in a concentration-dependent manner. Exposure to 10 microM levosimendan in the presence of 10 muM ropivacaine almost completely reversed the negative inotropic response. Sensitivity to the positive inotropic effect of levosimendan was not altered by 10 muM ropivacaine (-logEC50 [M] = 7.03 without versus 6.9 with ropivacaine, respectively). Action potential parameters were influenced only at the highest concentration. In the Langendorff heart, levosimendan significantly reversed the ropivacaine-induced reduction in heart rate, systolic pressure, coronary flow, and +dP/dt(max) to baseline values. CONCLUSION: Levosimendan is an effective inotropic drug in ropivacaine-induced myocardial depression and levosimendan myocardial sensitivity, and efficacy was not affected by the local anesthetic. Our results suggest that the calcium-sensitizing action of levosimendan is effective in local anesthetic-induced cardiac depression.


Assuntos
Amidas/toxicidade , Anestésicos Locais/toxicidade , Cardiotônicos/farmacologia , Coração/efeitos dos fármacos , Hidrazonas/farmacologia , Contração Miocárdica/efeitos dos fármacos , Músculos Papilares/efeitos dos fármacos , Piridazinas/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Circulação Coronária/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletrocardiografia , Cobaias , Coração/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Masculino , Músculos Papilares/fisiopatologia , Ropivacaina , Simendana , Fatores de Tempo , Pressão Ventricular/efeitos dos fármacos
20.
Cardiovasc Res ; 71(4): 695-703, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16876774

RESUMO

OBJECTIVES: The function of Kv4.3 (KCND3) channels, which underlie the transient outward current I(to) in human heart, can be modulated by several accessory subunits such as KChIP2 and KCNE1-KCNE5. Here we aimed to determine the regional expression of Kv4.3, KChIP2, and KCNE mRNAs in non-failing and failing human hearts and to investigate the functional consequences of subunit coexpression in heterologous expression systems. METHODS: We quantified mRNA levels for two Kv4.3 isoforms, Kv4.3-S and Kv4.3-L, and for KChIP2 as well as KCNE1-KCNE5 with real-time RT-PCR. We also studied the effects of KCNEs on Kv4.3+KChIP2 current characteristics in CHO cells with the whole-cell voltage-clamp method. RESULTS: In non-failing hearts, low expression was found for KCNE1, KCNE3, and KCNE5, three times higher expression for KCNE2, and 60 times higher for KCNE4. Transmural gradients were detected only for KChIP2 in left and right ventricles. Compared to non-failing tissue, failing hearts showed higher expression of Kv4.3-L and KCNE1 and lower of Kv4.3-S, KChIP2, KCNE4, and KCNE5. In CHO cells, Kv4.3+KChIP2 currents were differentially modified by co-expressed KCNEs: time constants of inactivation were shorter with KCNE1 and KCNE3-5 while time-to-peak was decreased, and V(0.5) of steady-state inactivation was shifted to more negative potentials by all KCNE subunits. Importantly, KCNE2 induced a unique and prominent 'overshoot' of peak current during recovery from inactivation similar to that described for human I(to) while other KCNE subunits induced little (KCNE4,5) or no overshoot. CONCLUSIONS: All KCNEs are expressed in the human heart at the transcript level. Compared to I(to) in native human myocytes, none of the combination of KChIP2 and KCNE produced an ideal congruency in current characteristics, suggesting that additional factors contribute to the regulation of the native I(to) channel.


Assuntos
Insuficiência Cardíaca/metabolismo , Proteínas Interatuantes com Canais de Kv/genética , Miocárdio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , RNA Mensageiro/análise , Canais de Potássio Shal/metabolismo , Animais , Células CHO , Estudos de Casos e Controles , Cricetinae , Cricetulus , Feminino , Regulação da Expressão Gênica , Humanos , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Potenciais da Membrana , Miocárdio/química , Técnicas de Patch-Clamp , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Potássio Shal/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA