Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cardiovasc Res ; 118(2): 517-530, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33705529

RESUMO

AIMS: Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS: Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION: This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.


Assuntos
Cardiomiopatia Hipertrófica/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Peroxidase/antagonistas & inibidores , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomiopatia Hipertrófica/enzimologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/fisiopatologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Humanos , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/fisiopatologia , Células-Tronco Pluripotentes Induzidas/enzimologia , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Peroxidase/metabolismo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
3.
JACC Basic Transl Sci ; 1(7): 590-602, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30167544

RESUMO

Cardiovascular disease remains the number one global cause of death and presents as multiple phenotypes in which the interplay between cardiomyocytes and cardiac fibroblasts (CFs) has become increasingly highlighted. Fetal and adult CFs influence neighboring cardiomyocytes in different ways. Thus far, a detailed comparison between the two is lacking. Using a genome-wide approach, we identified and validated 2 crucial players for maintaining the adult primary human CF phenotype. Knockdown of these factors induced significant phenotypical changes, including senescence and reduced collagen gene expression. These may now represent novel therapeutic targets against deleterious functions of CFs in adult cardiovascular disease.

4.
J Mol Cell Cardiol ; 67: 12-25, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24370890

RESUMO

The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.


Assuntos
Diferenciação Celular , Fenômenos Eletrofisiológicos , Miócitos Cardíacos/citologia , Técnicas de Cultura , Epigenômica , Coração/embriologia , Coração/crescimento & desenvolvimento , Humanos , Células-Tronco Pluripotentes/citologia , Transdução de Sinais
5.
Brain ; 136(Pt 12): 3775-86, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24240197

RESUMO

We studied a two-generation family presenting with conditions that included progressive permanent weakness, myopathic myopathy, exercise-induced contracture before normokalaemic periodic paralysis or, if localized to the tibial anterior muscle group, transient compartment-like syndrome (painful acute oedema with neuronal compression and drop foot). 23Na and 1H magnetic resonance imaging displayed myoplasmic sodium overload, and oedema. We identified a novel familial Ca(v)1.1 calcium channel mutation, R1242G, localized to the third positive charge of the domain IV voltage sensor. Functional expression of R1242G in the muscular dysgenesis mouse cell line GLT revealed a 28% reduced central pore inward current and a -20 mV shift of the steady-state inactivation curve. Both changes may be at least partially explained by an outward omega (gating pore) current at positive potentials. Moreover, this outward omega current of 27.5 nS/nF may cause the reduction of the overshoot by 13 mV and slowing of the upstroke of action potentials by 36% that are associated with muscle hypoexcitability (permanent weakness and myopathic myopathy). In addition to the outward omega current, we identified an inward omega pore current of 95 nS/nF at negative membrane potentials after long depolarizing pulses that shifts the R1242G residue above the omega pore constriction. A simulation reveals that the inward current might depolarize the fibre sufficiently to trigger calcium release in the absence of an action potential and therefore cause an electrically silent depolarization-induced muscle contracture. Additionally, evidence of the inward current can be found in 23Na magnetic resonance imaging-detected sodium accumulation and 1H magnetic resonance imaging-detected oedema. We hypothesize that the episodes are normokalaemic because of depolarization-induced compensatory outward potassium flux through both delayed rectifiers and omega pore. We conclude that the position of the R1242G residue before elicitation of the omega current is decisive for its conductance: if the residue is located below the gating pore as in the resting state then outward currents are observed; if the residue is above the gating pore because of depolarization, as in the inactivated state, then inward currents are observed. This study shows for the first time that functional characterization of omega pore currents is possible using a cultured cell line expressing mutant Ca(v)1.1 channels. Likewise, it is the first calcium channel mutation for complicated normokalaemic periodic paralysis.


Assuntos
Canais de Cálcio Tipo L/genética , Mutação/genética , Paralisias Periódicas Familiares/genética , Paralisias Periódicas Familiares/fisiopatologia , Potenciais de Ação/genética , Cálcio/metabolismo , Células Cultivadas , Estimulação Elétrica , Saúde da Família , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/fisiopatologia , Paralisias Periódicas Familiares/diagnóstico por imagem , Técnicas de Patch-Clamp , Cintilografia , Isótopos de Sódio , Trítio
7.
J Mol Cell Cardiol ; 53(3): 369-74, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22742967

RESUMO

The rapid component of the delayed rectifier potassium current (I(Kr)), encoded by the ether-a-go-go-related gene (ERG1, officially denominated as KCNH2), is a major contributor to repolarization in the mammalian heart. Acute (e.g. drug-induced) and chronic (e.g. inherited genetic disorder) disruptions of this current can lead to prolongation of the action potential and potentiate occurrence of lethal arrhythmias. Many cardiac and non-cardiac drugs show high affinity for the I(Kr) channel and it is therefore extensively studied during safety pharmacology. The unique biophysical and pharmacological properties of the I(Kr) channel are largely recapitulated by expressing the human variant (hERG1a) in overexpressing systems. hERG1a channels are tetramers consisting of four 1159 amino acid long proteins and have electrophysiological properties similar, but not identical, to native I(Kr). In the search for an explanation to the discrepancies between I(Kr) and hERG1a channels, two alternative hERG1 proteins have been found. Alternative transcription of hERG1 leads to a protein with a 56 amino acid shorter N-terminus, known as hERG1b. hERG1b can form channels alone or coassemble with hERG1a. Alternative splicing leads to an alternate C-terminus and a protein known as hERGuso. hERGuso and hERG1b regulate hERG1a channel trafficking, functional expression and channel kinetics. Expression of hERGuso leads to a reduced number of channels at the plasma membrane and thereby reduces current density. On the contrary, co-assembly with hERG1b alters channel kinetics resulting in more available channels and a larger current. These findings have implication for understanding mechanisms of disease, acute and chronic drug effects, and potential gender differences.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Coração/fisiologia , Miocárdio/metabolismo , Potássio/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Coração/efeitos dos fármacos , Humanos , Bloqueadores dos Canais de Potássio/farmacologia , Fatores Sexuais
8.
J Mol Cell Cardiol ; 52(5): 998-1008, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22353256

RESUMO

Human embryonic stem cell-derived cardiomyocytes (hESC-CM) have been proposed as a new model for safety pharmacology. So far, a thorough description of their basic electrophysiology and extensive testing, and mechanistic explanations, of their overall pro-arrhythmic ability is lacking. Under standardized conditions, we have evaluated the sensitivity of hESC-CM to proarrhythmic provocations by blockade of hERG and other channels. Using voltage patch clamp, some ion current densities (pA/pF) in hESC-CM were comparable to adult CM: I(Kr) (-12.5 ± 6.9), I(Ks) (0.65 ± 0.12), I(Na,peak) (-72 ± 21), I(Na,late) (-1.10 ± 0.36), and I(Ca,L) (-4.3 ± 0.6). I(f) density was larger (-10 ± 1.1) and I(K1) not existent or very small (-2.67 ± 0.3). The low I(K1) density was corroborated by low KCNJ2 mRNA levels. Effects of pro-arrhythmic compounds on action potential (AP) parameters and provocation of early afterdepolarizations (EADs) revealed that Chromanol293B (100 µmol/l) and Bay K8644 (1 µmol/l) both significantly prolonged APD(90). ATX-II (<1 µmol/l ) and BaCl(2) (10 µmol/l ) had no effect on APD. The only compound that triggered EADs was hERG blocker Cisapride. Computer simulations and AP clamp showed that the immature AP of hESC-CM prevents proper functioning of I(Na)-channels, and result in lower peak/maximal currents of several other channels, compared to the adult situation. Lack of functional I(K1) channels and shifted I(Na) channel activation cause a rather immature electrophysiological phenotype in hESC-CM, and thereby limits the potential of this model to respond accurately to pro-arrhythmic triggers other than hERG block. Maturation of the electrical phenotype is a prerequiste for future implementation of the model in arrhythmogenic safety testing.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Células-Tronco Embrionárias/fisiologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Potenciais de Ação , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/patologia , Benzazepinas/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Células Cultivadas , Cisaprida/farmacologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Canais de Sódio/metabolismo
9.
Assay Drug Dev Technol ; 9(6): 589-99, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22085047

RESUMO

The xCELLigence real time cell analyzer Cardio system offers a new system for real-time cell analysis that measures impedance-based signals in a label-free noninvasive manner. The aim of this study was to test whether impedance readings are a useful tool to detect compound effects on beating frequency (beats per minute, bpm) and arrhythmias of human induced pluripotent stem cell- and a mouse embryonic stem cell-derived cardiomyocyte line (hiPSC-CM and mESC-CM, respectively). Baseline values for control wells were 45±3 and 179±6 bpm, respectively (n=6). Correspondingly, isoproterenol increased beating frequency by 77% and 71%, whereas carbachol decreased frequency by 11% and 100% (stopped in 5/6 mESC-CM wells). E-4031 decreased beating rate and caused arrhythmias in both cell types, however, more pronounced in the human iPSC-CMs. Amlodipine inhibited contractions in both models, and T-type calcium channel block strongly reduced beating rate and eventually stopped beating in mESC-CM but caused a smaller effect in hiPSC-CM. The results of this initial study show that, under the right conditions, the beating frequency of a monolayer of cells can be stably recorded over several days. Additionally, the system detects changes in beating frequency and amplitude caused by added reference compounds. This assay system has the potential to enable medium-throughput screening, but for implementation into routine daily work, extended validation, testing of additional batches of cardiomyocytes, and further assay optimization (e.g., frequency of media exchange, growth matrix, seeding density, age of cells after plating, and temperature control) will be needed.


Assuntos
Arritmias Cardíacas , Cardiotônicos/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Animais , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Impedância Elétrica , Células-Tronco Embrionárias/fisiologia , Humanos , Camundongos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/fisiologia
10.
FASEB J ; 25(11): 3861-72, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21795504

RESUMO

Mesenchymal progenitor cells can be differentiated in vitro into myotubes that exhibit many characteristic features of primary mammalian skeletal muscle fibers. However, in general, they do not show the functional excitation-contraction coupling or the striated sarcomere arrangement typical of mature myofibers. Epigenetic modifications have been shown to play a key role in regulating the progressional changes in transcription necessary for muscle differentiation. In this study, we demonstrate that treatment of murine C2C12 mesenchymal progenitor cells with 10 µM of the DNA methylation inhibitor 5-azacytidine (5AC) promotes myogenesis, resulting in myotubes with enhanced maturity as compared to untreated myotubes. Specifically, 5AC treatment resulted in the up-regulation of muscle genes at the myoblast stage, while at later stages nearly 50% of the 5AC-treated myotubes displayed a mature, well-defined sarcomere organization, as well as spontaneous contractions that coincided with action potentials and intracellular calcium transients. Both the percentage of striated myotubes and their contractile activity could be inhibited by 20 nM TTX, 10 µM ryanodine, and 100 µM nifedipine, suggesting that action potential-induced calcium transients are responsible for these characteristics. Our data suggest that genomic demethylation induced by 5AC overcomes an epigenetic barrier that prevents untreated C2C12 myotubes from reaching full maturity.


Assuntos
Azacitidina/farmacologia , Metilação de DNA/fisiologia , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/citologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Quelantes/farmacologia , Metilação de DNA/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Epigênese Genética , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais , Camundongos , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas , Nifedipino/farmacologia , Rianodina/farmacologia , Sarcômeros/efeitos dos fármacos , Sarcômeros/ultraestrutura , Tetrodotoxina/farmacologia
11.
Stem Cell Res ; 4(3): 189-200, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20303332

RESUMO

To improve proarrhythmic predictability of preclinical models, we assessed whether human ventricular-like embryonic stem cell-derived cardiomyocytes (hESC-CMs) can be selected following a standardized protocol. Also, we quantified their arrhythmogenic response and compared this to a contemporary used rabbit Purkinje fiber (PF) model. Multiple transmembrane action potentials (AP) were recorded from 164 hESC-CM clusters (9 different batches), and 12 isolated PFs from New Zealand White rabbits. AP duration (APD), early afterdepolarizations (EADs), triangulation (T), and short-term variability of repolarization (STV) were determined on application of the I(Kr) blocker E-4031 (0.03/0.1/0.3/1 muM). Isoproterenol (0.1 muM) was used to assess adrenergic response. To validate the phenotype, RNA isolated from atrial- and ventricular-like clusters (n=8) was analyzed using low-density Taqman arrays. Based on initial experiments, slow beating rate (<50 bpm) and long APD (>200 ms) were used to select 31 ventricular-like clusters. E-4031 (1 muM) prolonged APD (31/31) and induced EADs only in clusters with APD90>300 ms (11/16). EADs were associated with increased T (1.6+/-0.2 vs 2.0+/-0.3) and STV (2.7+/-1.5 vs 6.9+/-1.9). Rabbit PF reacted in a similar way with regards to EADs (5/12), increased T (1.3+/-0.1 vs 1.9+/-0.4), and STV (1.2+/-0.9 vs 7.1+/-5.6). According to ROC values, hESC-CMs (STV 0.91) could predict EADs at least equivalent to PF (STV 0.69). Isoproterenol shortened APD and completely suppressed EADs. Gene expression analysis revealed that HCN1/2, KCNA5, and GJA5 were higher in atrial/nodal-like cells, whereas KCNJ2 and SCN1B were higher in ventricular-like cells (P<0.05). Selection of hESC-CM clusters with a ventricular-like phenotype can be standardized. The proarrhythmic results are qualitatively and quantitatively comparable between hESC-CMs and rabbit PF. Our results indicate that additional validation of this new safety pharmacology model is warranted.


Assuntos
Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/fisiologia , Potenciais de Ação/fisiologia , Animais , Linhagem Celular , Conexinas/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Eletrocardiografia , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Canais Iônicos/metabolismo , Isoproterenol/farmacologia , Canal de Potássio Kv1.5/metabolismo , Modelos Biológicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fenótipo , Canais de Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ramos Subendocárdicos/citologia , Ramos Subendocárdicos/fisiologia , RNA Mensageiro/metabolismo , Coelhos , Canais de Sódio/metabolismo , Proteína alfa-5 de Junções Comunicantes
12.
J Mol Cell Cardiol ; 48(1): 254-60, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19460390

RESUMO

Cardiomyocyte progenitor cells (CMPCs) can be isolated from the human heart and differentiated into cardiomyocytes in vitro. A comprehensive assessment of their electrical phenotype upon differentiation is essential to predict potential future applications of this cell source. CMPCs isolated from human fetal heart were differentiated in vitro and examined using immunohistochemistry, Western blotting, RT-PCR, voltage clamp and current clamp techniques. Differentiated cultures presented up to 95% alpha-actinin positive cardiomyocytes. Adherens junction and desmosomal proteins beta-catenin, N-cadherin, desmin and plakophilin2 were upregulated. Expression levels of cardiac connexins were not affected by differentiation, however Cx43 phosphorylation was increased upon differentiation, accompanied by translocation of connexins to the cell border. RT-PCR analysis demonstrated upregulation of all major cardiac ion channel constituents during differentiation. Patch clamp experiments showed that cardiomyocytes had a stable resting membrane potential of -73.4+/-1.8 mV. Infusion of 1 mM BaCl(2) resulted in depolarization to -59.9+/-2.8 mV, indicating I(K1) channel activity. Subsequent voltage clamp experiments confirmed presence of near mature I(Na), I(Ca,L) and I(K1) current densities. Infusion of the I(Kr) blocker Almokalant caused prolongation of the action potential by 40%. Differentiated monolayers were not spontaneously contracting in the absence of serum, but responded to field stimulation, displaying adult ventricular-like action potentials. Human fetal CMPC-derived cardiomyocytes have a homogenous and rather mature electrical phenotype that benefits to in vitro physiology and pharmacology. In the context of cardiac repair, their properties may translate into a reduced pro-arrhythmic risk and enhanced electrical integration upon transplantation.


Assuntos
Eletrofisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco/citologia , Antiarrítmicos/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Canais Iônicos/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Propanolaminas/farmacologia
13.
Curr Stem Cell Res Ther ; 4(1): 23-33, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19149627

RESUMO

The regenerative capacity of the mammalian heart is insufficient to recover from myocardial infarction. Stem cells are currently considered as a promising and valuable tool to replace the, often large, loss of contractile tissue. One of the bottlenecks hampering fast clinical application is the large amount of cells required to replace a single damaged region combined with an appropriate strategy to succeed in homogeneous repair. A second class of major cardiac disorders for which stem cell therapy might be fruitful and would require less cells for repair, are chronic rhythm disorders. In this area, most research has been focused on stem-cell based biological pacemakers, but increasing amounts of data on AV nodal repair appear in literature. Both therapies, in principle, could eventually replace current instrumentation with electronic pacemakers. Finally, an emerging field of interest explores transplantation of stem cells expressing specific ion channels aiming at suppression of focal arrhythmias, providing an alternative strategy for surgical and catheter-mediated ablation. Since in this second class of applications the number of transplanted cells required may be relatively low, effective clinical therapy may be within close range. Here, we will review recent achievements in the fields of stem-cell based biological pacemakers, AV nodal repair and biological ablation.


Assuntos
Relógios Biológicos/fisiologia , Sistema de Condução Cardíaco/fisiopatologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Recuperação de Função Fisiológica , Transplante de Células-Tronco , Animais , Arritmias Cardíacas , Movimento Celular , Engenharia Genética , Sistema de Condução Cardíaco/fisiologia , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA