Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Mol Cell Cardiol ; 168: 13-23, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35405106

RESUMEN

A key therapeutic target for heart failure and arrhythmia is the deleterious leak through sarcoplasmic reticulum (SR) ryanodine receptor 2 (RyR2) calcium release channels. We have previously developed methods to detect the pathologically leaky state of RyR2 in adult cardiomyocytes by monitoring RyR2 binding to either calmodulin (CaM) or a biosensor peptide (DPc10). Here, we test whether these complementary binding measurements are effective as high-throughput screening (HTS) assays to discover small molecules that target leaky RyR2. Using FRET, we developed and validated HTS procedures under conditions that mimic a pathological state, to screen the library of 1280 pharmaceutically active compounds (LOPAC) for modulators of RyR2 in cardiac SR membrane preparations. Complementary FRET assays with acceptor-labeled CaM and DPc10 were used for Hit prioritization based on the opposing binding properties of CaM vs. DPc10. This approach narrowed the Hit list to one compound, Ro 90-7501, which altered FRET to suggest increased RyR2-CaM binding and decreased DPc10 binding. Follow-up studies revealed that Ro 90-7501 does not detrimentally affect myocyte Ca2+ transients. Moreover, Ro 90-7501 partially inhibits overall Ca2+ leak, as assessed by Ca2+ sparks in permeabilized rat cardiomyocytes. Together, these results demonstrate (1) the effectiveness of our HTS approach where two complementary assays synergize for Hit ranking and (2) a drug discovery process that combines high-throughput, high-precision in vitro structural assays with in situ myocyte assays of the pathologic RyR2 leak. These provide a drug discovery platform compatible with large-scale HTS campaigns, to identify agents that inhibit RyR2 for therapeutic development.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Canal Liberador de Calcio Receptor de Rianodina , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Descubrimiento de Drogas , Transferencia Resonante de Energía de Fluorescencia/métodos , Miocitos Cardíacos/metabolismo , Ratas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
2.
Circulation ; 128(11 Suppl 1): S3-13, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24030418

RESUMEN

BACKGROUND: Drug-induced arrhythmia is one of the most common causes of drug development failure and withdrawal from market. This study tested whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with a low-impedance microelectrode array (MEA) system could improve on industry-standard preclinical cardiotoxicity screening methods, identify the effects of well-characterized drugs, and elucidate underlying risk factors for drug-induced arrhythmia. hiPSC-CMs may be advantageous over immortalized cell lines because they possess similar functional characteristics as primary human cardiomyocytes and can be generated in unlimited quantities. METHODS AND RESULTS: Pharmacological responses of beating embryoid bodies exposed to a comprehensive panel of drugs at 65 to 95 days postinduction were determined. Responses of hiPSC-CMs to drugs were qualitatively and quantitatively consistent with the reported drug effects in literature. Torsadogenic hERG blockers, such as sotalol and quinidine, produced statistically and physiologically significant effects, consistent with patch-clamp studies, on human embryonic stem cell-derived cardiomyocytes hESC-CMs. False-negative and false-positive hERG blockers were identified accurately. Consistent with published studies using animal models, early afterdepolarizations and ectopic beats were observed in 33% and 40% of embryoid bodies treated with sotalol and quinidine, respectively, compared with negligible early afterdepolarizations and ectopic beats in untreated controls. CONCLUSIONS: We found that drug-induced arrhythmias can be recapitulated in hiPSC-CMs and documented with low impedance MEA. Our data indicate that the MEA/hiPSC-CM assay is a sensitive, robust, and efficient platform for testing drug effectiveness and for arrhythmia screening. This system may hold great potential for reducing drug development costs and may provide significant advantages over current industry standard assays that use immortalized cell lines or animal models.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Fármacos Cardiovasculares/efectos adversos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Análisis de Matrices Tisulares/métodos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adolescente , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Impedancia Eléctrica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Microelectrodos , Miocitos Cardíacos/fisiología
3.
Cold Spring Harb Protoc ; 2013(4): 305-9, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23547145

RESUMEN

It is widely agreed that the best method for measuring the ionized free calcium concentration ([Ca(2+)]) in large volumes of biological solutions is to use Ca(2+)-sensitive macroelectrodes. These are commercially available. To measure [Ca(2+)] in small volumes of solution, minielectrodes with 1-2-mm tips can easily be made and used, and may also be commercially available. Ca(2+)-sensitive microelectrodes (CaSMs, with 0.5-2-µm tips) can also be made and used extracellularly or intracellularly in robust cells, but interest in their use has recently been largely eclipsed. This is because of practical difficulties and the introduction of a large number of fluorescent and other optical calcium probes with calcium sensitivities varying from the nanomolar to the millimolar range, such as Fura-2, Indo-1, Fluo-4, and many others. In this article, we emphasize the utility of Ca(2+)-selective electrodes and show that their use is complementary to use of fluorescent and other optical methods. Each method has advantages and disadvantages. Because numerous reviews and books have been dedicated to the theoretical aspects of ion-selective electrode principles and technology, this article is mainly intended for investigators who have some degree of electrophysiological experience with ion-selective electrodes or microelectrodes.


Asunto(s)
Calcio/análisis , Electrodos de Iones Selectos , Electrofisiología/métodos , Fluorometría/métodos
4.
Circulation ; 127(16): 1677-91, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23519760

RESUMEN

BACKGROUND: Cardiotoxicity is a leading cause for drug attrition during pharmaceutical development and has resulted in numerous preventable patient deaths. Incidents of adverse cardiac drug reactions are more common in patients with preexisting heart disease than the general population. Here we generated a library of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients with various hereditary cardiac disorders to model differences in cardiac drug toxicity susceptibility for patients of different genetic backgrounds. METHODS AND RESULTS: Action potential duration and drug-induced arrhythmia were measured at the single cell level in hiPSC-CMs derived from healthy subjects and patients with hereditary long QT syndrome, familial hypertrophic cardiomyopathy, and familial dilated cardiomyopathy. Disease phenotypes were verified in long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy hiPSC-CMs by immunostaining and single cell patch clamp. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and the human ether-a-go-go-related gene expressing human embryonic kidney cells were used as controls. Single cell PCR confirmed expression of all cardiac ion channels in patient-specific hiPSC-CMs as well as hESC-CMs, but not in human embryonic kidney cells. Disease-specific hiPSC-CMs demonstrated increased susceptibility to known cardiotoxic drugs as measured by action potential duration and quantification of drug-induced arrhythmias such as early afterdepolarizations and delayed afterdepolarizations. CONCLUSIONS: We have recapitulated drug-induced cardiotoxicity profiles for healthy subjects, long QT syndrome, hypertrophic cardiomyopathy, and dilated cardiomyopathy patients at the single cell level for the first time. Our data indicate that healthy and diseased individuals exhibit different susceptibilities to cardiotoxic drugs and that use of disease-specific hiPSC-CMs may predict adverse drug responses more accurately than the standard human ether-a-go-go-related gene test or healthy control hiPSC-CM/hESC-CM screening assays.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Predisposición Genética a la Enfermedad , Células Madre Pluripotentes Inducidas/citología , Síndrome de QT Prolongado/genética , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica Familiar/patología , Diferenciación Celular , Línea Celular/efectos de los fármacos , Línea Celular/fisiología , Tamaño de la Célula , Cisaprida/toxicidad , Cuerpos Embrioides/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Perfilación de la Expresión Génica , Células HEK293/efectos de los fármacos , Células HEK293/fisiología , Humanos , Técnicas In Vitro , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Riñón/citología , Riñón/embriología , Síndrome de QT Prolongado/patología , Miocitos Cardíacos/fisiología , Miofibrillas/ultraestructura , Nicorandil/toxicidad , Técnicas de Placa-Clamp , Quinazolinas/toxicidad , Verapamilo/toxicidad
5.
Circ Res ; 110(11): 1454-64, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22539768

RESUMEN

RATIONALE: ß-Adrenergic receptor stimulation produces sarcoplasmic reticulum Ca(2+) overload and delayed afterdepolarizations in isolated ventricular myocytes. How delayed afterdepolarizations are synchronized to overcome the source-sink mismatch and produce focal arrhythmia in the intact heart remains unknown. OBJECTIVE: To determine whether local ß-adrenergic receptor stimulation produces spatiotemporal synchronization of delayed afterdepolarizations and to examine the effects of tissue geometry and cell-cell coupling on the induction of focal arrhythmia. METHODS AND RESULTS: Simultaneous optical mapping of transmembrane potential and Ca(2+) transients was performed in normal rabbit hearts during subepicardial injections (50 µL) of norepinephrine (NE) or control (normal Tyrode's solution). Local NE produced premature ventricular complexes (PVCs) from the injection site that were dose-dependent (low-dose [30-60 µmol/L], 0.45±0.62 PVCs per injection; high-dose [125-250 µmol/L], 1.33±1.46 PVCs per injection; P<0.0001) and were inhibited by propranolol. NE-induced PVCs exhibited abnormal voltage-Ca(2+) delay at the initiation site and were inhibited by either sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase inhibition or reduced perfusate [Ca(2+)], which indicates a Ca(2+)-mediated mechanism. NE-induced PVCs were more common at right ventricular than at left ventricular sites (1.48±1.50 versus 0.55±0.89, P<0.01), and this was unchanged after chemical ablation of endocardial Purkinje fibers, which suggests that source-sink interactions may contribute to the greater propensity to right ventricular PVCs. Partial gap junction uncoupling with carbenoxolone (25 µmol/L) increased focal activity (2.18±1.43 versus 1.33±1.46 PVCs per injection, P<0.05), which further supports source-sink balance as a critical mediator of Ca(2+)-induced PVCs. CONCLUSIONS: These data provide the first experimental demonstration that localized ß-adrenergic receptor stimulation produces spatiotemporal synchronization of sarcoplasmic reticulum Ca(2+) overload and release in the intact heart and highlight the critical nature of source-sink balance in initiating focal arrhythmias.


Asunto(s)
Agonistas Adrenérgicos beta , Comunicación Celular , Miocitos Cardíacos/metabolismo , Norepinefrina , Receptores Adrenérgicos beta/metabolismo , Complejos Prematuros Ventriculares/inducido químicamente , Potenciales de Acción , Agonistas Adrenérgicos beta/administración & dosificación , Antagonistas Adrenérgicos beta/administración & dosificación , Animales , Señalización del Calcio , Ablación por Catéter , Comunicación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Uniones Comunicantes/metabolismo , Inyecciones , Masculino , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/administración & dosificación , Perfusión , Propranolol/administración & dosificación , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/cirugía , Conejos , Receptores Adrenérgicos beta/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Factores de Tiempo , Complejos Prematuros Ventriculares/metabolismo , Complejos Prematuros Ventriculares/fisiopatología , Complejos Prematuros Ventriculares/prevención & control , Imagen de Colorante Sensible al Voltaje
6.
J Mol Cell Cardiol ; 39(6): 972-81, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16242149

RESUMEN

Cardiac Na/Ca exchange (NCX, NCX1.1) is critical in cardiac myocyte Ca regulation, and its altered function contributes to inotropic state, systolic dysfunction in heart failure and arrhythmogenesis. Regulation of NCX is multifaceted, but protein kinase A (PKA) effects on NCX function are controversial. Here, we use three different and complementary approaches to compare NCX function +/-1 microM isoproterenol (ISO) in intact rabbit cardiac myocytes (in paired comparisons). First, in field-stimulated intact cells we inferred the cytosolic [Ca] ([Ca](i)) dependence of NCX function from the decay rate of caffeine-induced [Ca](i) transients. Second, we measured caffeine-induced [Ca](i) and inward I(NCX) simultaneously (perforated patch voltage clamp), to measure directly the [Ca](i) dependence of NCX rate. Third, using whole cell ruptured patch with [Ca](i) heavily buffered to 100 nM, [Na](i)=10 mM, and I(Ca), SR Ca release and Na/K pump all blocked, we recorded I(NCX) ramps at 37 degrees C. We find that NCX function is not altered by PKA activation under any of these three protocols, where intracellular conditions ranged from near-physiological to highly controlled. This does not rule out NCX modulation by PKA under all conditions, or in species other than rabbit. However, such effects are likely to be either minor (vs. other PKA actions on myocyte Ca handling) or indirect, such as secondary effects dependent on altered local [Ca](i) and [Na](i).


Asunto(s)
Cardiotónicos/farmacología , Isoproterenol/farmacología , Miocitos Cardíacos/enzimología , Potasio/metabolismo , Sodio/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Cafeína/farmacología , Células Cultivadas , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Transporte Iónico/efectos de los fármacos , Miocitos Cardíacos/citología , Inhibidores de Fosfodiesterasa/farmacología , Conejos , ATPasa Intercambiadora de Sodio-Potasio
7.
Sci STKE ; 2003(177): PE13, 2003 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-12684526

RESUMEN

Highly localized changes in intracellular calcium concentration [Ca2+]i play a critical role in regulating numerous cellular functions, ranging from muscle contraction to neurotransmitter and hormone secretion to gene transcription. Fluorescent Ca2+ indicators have been invaluable tools in elucidating the role of localized changes in [Ca2+]i in regulating ion channels and other key proteins in various signaling pathways. Other techniques used to investigate localized changes in [Ca2+]i include approaches based on fluorescence resonance energy transfer, and electrophysiological measurements of ionic flux through Ca2+-sensitive channels. This Perspective discusses research using fluorescent Ca2+ indicators to study excitation-contraction coupling in cardiac myocytes, presenting both key findings and limitations of this approach. Complementary approaches useful in studying localized changes in Ca2+ and other second messengers (such as cyclic adenosine monophosphate) are also discussed.


Asunto(s)
Señalización del Calcio/fisiología , Fenómenos Fisiológicos Celulares , Imagenología Tridimensional/métodos , Animales , Ecocardiografía Tetradimensional/métodos , Microscopía Confocal/métodos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA