Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Coll Cardiol ; 73(18): 2310-2324, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31072576

RESUMEN

BACKGROUND: The short QT syndrome (SQTS) is an inherited arrhythmogenic syndrome characterized by abnormal ion channel function, life-threatening arrhythmias, and sudden cardiac death. OBJECTIVES: The purpose of this study was to establish a patient-specific human-induced pluripotent stem cell (hiPSC) model of the SQTS, and to provide mechanistic insights into its pathophysiology and therapy. METHODS: Patient-specific hiPSCs were generated from a symptomatic SQTS patient carrying the N588K mutation in the KCNH2 gene, differentiated into cardiomyocytes, and compared with healthy and isogenic (established by CRISPR/Cas9-based mutation correction) control hiPSC-derived cardiomyocytes (hiPSC-CMs). Patch-clamp was used to evaluate action-potential (AP) and IKr current properties at the cellular level. Conduction and arrhythmogenesis were studied at the tissue level using confluent 2-dimensional hiPSC-derived cardiac cell sheets (hiPSC-CCSs) and optical mapping. RESULTS: Intracellular recordings demonstrated shortened action-potential duration (APD) and abbreviated refractory period in the SQTS-hiPSC-CMs. Similarly, voltage- and AP-clamp recordings revealed increased IKr current density due to attenuated inactivation, primarily in the AP plateau phase. Optical mapping of the SQTS-hiPSC-CCSs revealed shortened APD, impaired APD-rate adaptation, abbreviated wavelength of excitation, and increased inducibility of sustained spiral waves. Phase-mapping analysis revealed accelerated and stabilized rotors manifested by increased rotor rotation frequency, increased rotor curvature, decreased core meandering, and increased rotor complexity. Application of quinidine and disopyramide, but not sotalol, normalized APD and suppressed arrhythmia induction. CONCLUSIONS: A novel hiPSC-based model of the SQTS was established at both the cellular and tissue levels. This model recapitulated the disease phenotype in the culture dish and provided important mechanistic insights into arrhythmia mechanisms in the SQTS and its treatment.


Asunto(s)
Arritmias Cardíacas , Miocitos Cardíacos/metabolismo , Potenciales de Acción , Antiarrítmicos/farmacología , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/prevención & control , Células Cultivadas , Canal de Potasio ERG1/genética , Humanos , Células Madre Pluripotentes Inducidas , Mutación , Técnicas de Placa-Clamp , Modelación Específica para el Paciente
2.
Acta Biomater ; 92: 145-159, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31075518

RESUMEN

Cardiac tissue engineering provides unique opportunities for cardiovascular disease modeling, drug testing, and regenerative medicine applications. To recapitulate human heart tissue, we combined human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with a chitosan-enhanced extracellular-matrix (ECM) hydrogel, derived from decellularized pig hearts. Ultrastructural characterization of the ECM-derived engineered heart tissues (ECM-EHTs) revealed an anisotropic muscle structure, with embedded cardiomyocytes showing more mature properties than 2D-cultured hiPSC-CMs. Force measurements confirmed typical force-length relationships, sensitivity to extracellular calcium, and adequate ionotropic responses to contractility modulators. By combining genetically-encoded calcium and voltage indicators with laser-confocal microscopy and optical mapping, the electrophysiological and calcium-handling properties of the ECM-EHTs could be studied at the cellular and tissue resolutions. This allowed to detect drug-induced changes in contraction rate (isoproterenol, carbamylcholine), optical signal morphology (E-4031, ATX2, isoproterenol, ouabin and quinidine), cellular arrhythmogenicity (E-4031 and ouabin) and alterations in tissue conduction properties (lidocaine, carbenoxolone and quinidine). Similar assays in ECM-EHTs derived from patient-specific hiPSC-CMs recapitulated the abnormal phenotype of the long QT syndrome and catecholaminergic polymorphic ventricular tachycardia. Finally, programmed electrical stimulation and drug-induced pro-arrhythmia led to the development of reentrant arrhythmias in the ECM-EHTs. In conclusion, a novel ECM-EHT model was established, which can be subjected to high-resolution long-term serial functional phenotyping, with important implications for cardiac disease modeling, drug testing and precision medicine. STATEMENT OF SIGNIFICANCE: One of the main objectives of cardiac tissue engineering is to create an in-vitro muscle tissue surrogate of human heart tissue. To this end, we combined a chitosan-enforced cardiac-specific ECM hydrogel derived from decellularized pig hearts with human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from healthy-controls and patients with inherited cardiac disorders. We then utilized genetically-encoded calcium and voltage fluorescent indicators coupled with unique optical imaging techniques and force-measurements to study the functional properties of the generated engineered heart tissues (EHTs). These studies demonstrate the unique potential of the new model for physiological and pathophysiological studies (assessing contractility, conduction and reentrant arrhythmias), novel disease modeling strategies ("disease-in-a-dish" approach) for studying inherited arrhythmogenic disorders, and for drug testing applications (safety pharmacology).


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Matriz Extracelular/metabolismo , Corazón/fisiología , Células Madre Pluripotentes Inducidas/citología , Modelos Cardiovasculares , Miocitos Cardíacos/citología , Ingeniería de Tejidos/métodos , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/patología , Calcio/metabolismo , Fármacos Cardiovasculares/farmacología , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Humanos , Hidrogeles/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Especificidad de Órganos , Porcinos
3.
Stem Cell Reports ; 10(6): 1879-1894, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29754959

RESUMEN

Fulfilling the potential of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes for studying conduction and arrhythmogenesis requires development of multicellular models and methods for long-term repeated tissue phenotyping. We generated confluent hiPSC-derived cardiac cell sheets (hiPSC-CCSs), expressing the genetically encoded voltage indicator ArcLight. ArcLight-based optical mapping allowed generation of activation and action-potential duration (APD) maps, which were validated by mapping the same hiPSC-CCSs with the voltage-sensitive dye, Di-4-ANBDQBS. ArcLight mapping allowed long-term assessment of electrical remodeling in the hiPSC-CCSs and evaluation of drug-induced conduction slowing (carbenoxolone, lidocaine, and quinidine) and APD prolongation (quinidine and dofetilide). The latter studies also enabled step-by-step depiction of drug-induced arrhythmogenesis ("torsades de pointes in the culture dish") and its prevention by MgSO4 and rapid pacing. Phase-mapping analysis allowed biophysical characterization of spiral waves induced in the hiPSC-CCSs and their termination by electrical cardioversion and overdrive pacing. In conclusion, ArcLight mapping of hiPSC-CCSs provides a powerful tool for drug testing and arrhythmia investigation.


Asunto(s)
Biomarcadores , Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Potenciales de Acción , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Humanos , Modelos Biológicos , Imagen Molecular , Miocitos Cardíacos/efectos de los fármacos , Fenetilaminas , Sulfonamidas
4.
Stem Cell Reports ; 5(4): 582-96, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26372632

RESUMEN

The advent of the human-induced pluripotent stem cell (hiPSC) technology has transformed biomedical research, providing new tools for human disease modeling, drug development, and regenerative medicine. To fulfill its unique potential in the cardiovascular field, efficient methods should be developed for high-resolution, large-scale, long-term, and serial functional cellular phenotyping of hiPSC-derived cardiomyocytes (hiPSC-CMs). To achieve this goal, we combined the hiPSC technology with genetically encoded voltage (ArcLight) and calcium (GCaMP5G) fluorescent indicators. Expression of ArcLight and GCaMP5G in hiPSC-CMs permitted to reliably follow changes in transmembrane potential and intracellular calcium levels, respectively. This allowed monitoring short- and long-term changes in action-potential and calcium-handling properties and the development of arrhythmias in response to several pharmaceutical agents and in hiPSC-CMs derived from patients with different inherited arrhythmogenic syndromes. Combining genetically encoded fluorescent reporters with hiPSC-CMs may bring a unique value to the study of inherited disorders, developmental biology, and drug development and testing.


Asunto(s)
Potenciales de Acción , Calcio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Arritmias Cardíacas/metabolismo , Calcio/análisis , Diferenciación Celular , Células Cultivadas , Expresión Génica , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Miocitos Cardíacos/metabolismo , Imagen Óptica/métodos , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Transducción Genética , Transgenes
5.
Curr Treat Options Cardiovasc Med ; 16(9): 331, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25080030

RESUMEN

OPINION STATEMENT: The study of inherited cardiac disorders is hampered by the lack of suitable in vitro human cardiac disease models and relevant functional assays. A potential solution to this cell-sourcing challenge may be the recently described human induced pluripotent stem cell (hiPSC) technology. Pioneering studies were successful in establishing patient-specific, hiPSC-derived cardiomyocyte models of the most common inherited cardiac arrhythmogenic disorders, such as the different long QT syndrome subtypes (types 1, 2, 3, 8), overlap syndrome LQTS3/Brugada syndrome, catecholaminergic polymorphic ventricular tachycardia (CPVT) types 1 and 2, and arrhythmogenic right ventricular cardiomyopathy (ARVC). These studies initially demonstrated the ability of the generated hiPSC models to recapitulate the disease processes in the culture dish. More recently, such studies were also able to provide new mechanistic insights into the disease processes, as well as to derive a unique drug-screening platform to test existing and novel therapeutic treatment options in an environment resembling the human physiological milieu. Moreover, initial evidence suggests that such models can help to optimize drug treatment in a personalized manner in the future. Nevertheless, several hurdles still exist for using hiPSC-based models for the aforementioned tasks, such as the hiPSC-derived cardiomyocytes' relatively immature phenotype, and also the resulting cell heterogeneity. Extensive research work is ongoing to address these challenges, as well as to add new opportunities for the field by utilizing recent advances in gene editing technologies. Here, we discuss the significant findings that hiPSC-based models have provided for each of the inherited cardiac arrhythmia syndromes so far, and the current challenges that this technique is facing.

6.
J Am Coll Cardiol ; 64(5): 451-9, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25082577

RESUMEN

BACKGROUND: Human induced pluripotent stem cells (iPSCs) play an important role in disease modeling and drug testing. However, the current methods are time-consuming and lack an isogenic control. OBJECTIVES: This study sought to establish an efficient technology to generate human PSC-based disease models with isogenic control. METHODS: The ion channel genes KCNQ1 and KCNH2 with dominant negative mutations causing long QT syndrome types 1 and 2, respectively, were stably integrated into a safe harbor AAVS1 locus using zinc finger nuclease technology. RESULTS: Patch-clamp recording revealed that the edited iPSC-derived cardiomyocytes (iPSC-CMs) displayed characteristic long QT syndrome phenotype and significant prolongation of the action potential duration compared with the unedited control cells. Finally, addition of nifedipine (L-type calcium channel blocker) or pinacidil (KATP-channel opener) shortened the action potential duration of iPSC-CMs, confirming the validity of isogenic iPSC lines for drug testing in the future. CONCLUSIONS: Our study demonstrates that iPSC-CM-based disease models can be rapidly generated by overexpression of dominant negative gene mutants.


Asunto(s)
Antiarrítmicos/uso terapéutico , Células Madre Pluripotentes Inducidas/patología , Síndrome de QT Prolongado/genética , Diferenciación Celular , Transdiferenciación Celular , Células Cultivadas , Genoma , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/patología , Técnicas de Placa-Clamp
7.
Cardiovasc Res ; 102(1): 176-87, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24518144

RESUMEN

AIMS: Optogenetics approaches, utilizing light-sensitive proteins, have emerged as unique experimental paradigms to modulate neuronal excitability. We aimed to evaluate whether a similar strategy could be used to control cardiac-tissue excitability. METHODS AND RESULTS: A combined cell and gene therapy strategy was developed in which fibroblasts were transfected to express the light-activated depolarizing channel Channelrhodopsin-2 (ChR2). Patch-clamp studies confirmed the development of a robust inward current in the engineered fibroblasts following monochromatic blue-light exposure. The engineered cells were co-cultured with neonatal rat cardiomyocytes (or human embryonic stem cell-derived cardiomyocytes) and studied using a multielectrode array mapping technique. These studies revealed the ability of the ChR2-fibroblasts to electrically couple and pace the cardiomyocyte cultures at varying frequencies in response to blue-light flashes. Activation mapping pinpointed the source of this electrical activity to the engineered cells. Similarly, diffuse seeding of the ChR2-fibroblasts allowed multisite optogenetics pacing of the co-cultures, significantly shortening their electrical activation time and synchronizing contraction. Next, optogenetics pacing in an in vitro model of conduction block allowed the resynchronization of the tissue's electrical activity. Finally, the ChR2-fibroblasts were transfected to also express the light-sensitive hyperpolarizing proton pump Archaerhodopsin-T (Arch-T). Seeding of the ChR2/ArchT-fibroblasts allowed to either optogentically pace the cultures (in response to blue-light flashes) or completely suppress the cultures' electrical activity (following continuous illumination with 624 nm monochromatic light, activating ArchT). CONCLUSIONS: The results of this proof-of-concept study highlight the unique potential of optogenetics for future biological pacemaking and resynchronization therapy applications and for the development of novel anti-arrhythmic strategies.


Asunto(s)
Luz , Miocitos Cardíacos/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Antiarrítmicos/farmacología , Células Cultivadas , Técnicas de Cocultivo , Fenómenos Electrofisiológicos/fisiología , Corazón , Humanos , Ratones , Miocitos Cardíacos/efectos de los fármacos , Optogenética/métodos , Técnicas de Placa-Clamp/métodos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...