Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Toxicol Sci ; 147(1): 39-54, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26092927

RESUMEN

Drug-induced liver injury (DILI) remains a great challenge and a major concern during late-stage drug development. Induced pluripotent stem cells (iPSC) represent an exciting alternative in vitro model system to explore the role of genetic diversity in DILI, especially when derived from patients who have experienced drug-induced hepatotoxicity. The development and validation of the iPSC-derived hepatocytes as an in vitro cell-based model of DILI is an essential first step in creating more predictive tools for understanding patient-specific hepatotoxic responses to drug treatment. In this study, we performed extensive morphological and functional analyses on iPSC-derived hepatocytes from a commercial source. iPSC-derived hepatocytes exhibit many of the key morphological and functional features of primary hepatocytes, including membrane polarity and production of glycogen, lipids, and key hepatic proteins, such as albumin, asialoglycoprotein receptor and α1-antitrypsin. They maintain functional activity for many drug-metabolizing enzyme pathways and possess active efflux capacity of marker substrates into bile canalicular compartments. Whole genome-wide array analysis of multiple batches of iPSC-derived cells showed that their transcriptional profiles are more similar to those from neonatal and adult hepatocytes than those from fetal liver. Results from experiments using prototype DILI compounds, such as acetaminophen and trovafloxacin, indicate that these cells are able to reproduce key characteristic metabolic and adaptive responses attributed to the drug-induced hepatotoxic effects in vivo. Overall, this novel system represents a promising new tool for understanding the underlying mechanisms of idiosyncratic DILI and for screening new compounds for DILI-related liabilities.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatocitos/ultraestructura , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/ultraestructura , Pruebas de Toxicidad/métodos , Acetaminofén/toxicidad , Adulto , Bilis/metabolismo , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Fluoroquinolonas/toxicidad , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Recién Nacido , Metabolismo de los Lípidos/efectos de los fármacos , Lipidosis/inducido químicamente , Lipidosis/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Naftiridinas/toxicidad , Embarazo , Reproducibilidad de los Resultados , Transcripción Genética
2.
J Biomol Screen ; 18(10): 1203-11, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24071917

RESUMEN

A major hurdle for cardiovascular disease researchers has been the lack of robust and physiologically relevant cell-based assays for drug discovery. Derivation of cardiomyocytes from human-induced pluripotent stem (iPS) cells at high purity, quality, and quantity enables the development of relevant models of human cardiac disease with source material that meets the demands of high-throughput screening (HTS). Here we demonstrate the utility of iPS cell-derived cardiomyocytes as an in vitro model of cardiac hypertrophy. Exposure of cardiomyocytes to endothelin 1 (ET-1) leads to reactivation of fetal genes, increased cell size, and robust expression of B-type natriuretic peptide (BNP). Using this system, we developed a suite of assays focused on BNP detection, most notably a high-content imaging-based assay designed for phenotypic screening. Miniaturization of this assay to a 384-well format enabled the profiling of a small set of tool compounds known to modulate the hypertrophic response. The assays described here provide consistent and reliable results and have the potential to increase our understanding of the many mechanisms underlying this complex cardiac condition. Moreover, the HTS-compatible workflow allows for the incorporation of human biology into early phases of drug discovery and development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/efectos de los fármacos , Biomarcadores/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Cardiomegalia/tratamiento farmacológico , Diferenciación Celular , Tamaño de la Célula , Células Cultivadas , Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Imidazoles/farmacología , Concentración 50 Inhibidora , Miocitos Cardíacos/metabolismo , Fenotipo , Quinolinas/farmacología , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo , Verapamilo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA