Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Pharmacol Drug Dev ; 10(1): 99-109, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32648303

RESUMEN

Ivosidenib, a small-molecule inhibitor of mutant isocitrate dehydrogenase 1, is primarily cleared by hepatic metabolism. This open-label study investigated the impact of hepatic impairment on ivosidenib pharmacokinetics (ClinicalTrials.gov: NCT03282513). Otherwise healthy participants with mild (n = 9) or moderate (n = 8) hepatic impairment (Child-Pugh score) and matched participants with normal hepatic function (n = 16) received 1 oral dose of 500-mg ivosidenib. Mild hepatic impairment had a negligible effect on total ivosidenib plasma exposure, with geometric mean ratios (90% confidence interval [CI]) of 0.933 (0.715-1.22) for maximum concentration (Cmax ) and 0.847 (0.624-1.15) for area under the plasma concentration-time curve (AUC) in participants with mild hepatic impairment versus matched controls. Moderate hepatic impairment reduced total ivosidenib exposure by 28% to 44%, with geometric mean ratios (90%CI) of 0.565 (0.419-0.763) for Cmax and 0.716 (0.479-1.07) for AUC, although the 90%CI for AUC included 1.00. The ivosidenib unbound fraction was concentration dependent and higher in participants with mild/moderate hepatic impairment compared with matched controls. There was no apparent trend to increasing unbound Cmax with increased hepatic impairment severity. A single 500-mg ivosidenib dose was well tolerated, with no serious or severe adverse events and no adverse events leading to discontinuation. We conclude that mild/moderate hepatic impairment did not lead to clinically relevant changes in ivosidenib exposure following a single 500-mg dose.


Asunto(s)
Antineoplásicos/farmacocinética , Glicina/análogos & derivados , Hepatopatías/metabolismo , Piridinas/farmacocinética , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/sangre , Femenino , Glicina/administración & dosificación , Glicina/efectos adversos , Glicina/sangre , Glicina/farmacocinética , Voluntarios Sanos , Humanos , Hígado/metabolismo , Masculino , Piridinas/administración & dosificación , Piridinas/efectos adversos , Piridinas/sangre
2.
Cell Res ; 24(12): 1420-32, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25367124

RESUMEN

The cardiac progenitor cells (CPCs) in the anterior heart field (AHF) are located in the pharyngeal mesoderm (PM), where they expand, migrate and eventually differentiate into major cell types found in the heart, including cardiomyocytes. The mechanisms by which these progenitors are able to expand within the PM microenvironment without premature differentiation remain largely unknown. Through in silico data mining, genetic loss-of-function studies, and in vivo genetic rescue studies, we identified N-cadherin and interaction with canonical Wnt signals as a critical component of the microenvironment that facilitates the expansion of AHF-CPCs in the PM. CPCs in N-cadherin mutant embryos were observed to be less proliferative and undergo premature differentiation in the PM. Notably, the phenotype of N-cadherin deficiency could be partially rescued by activating Wnt signaling, suggesting a delicate functional interaction between the adhesion role of N-cadherin and Wnt signaling in the early PM microenvironment. This study suggests a new mechanism for the early renewal of AHF progenitors where N-cadherin provides additional adhesion for progenitor cells in the PM, thereby allowing Wnt paracrine signals to expand the cells without premature differentiation.


Asunto(s)
Cadherinas/metabolismo , Diferenciación Celular , Corazón/embriología , Mesodermo/citología , Miocitos Cardíacos/citología , Células Madre/citología , Animales , Cadherinas/genética , Células Cultivadas , Femenino , Eliminación de Gen , Masculino , Mesodermo/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Transducción de Señal , Células Madre/metabolismo , Vía de Señalización Wnt
3.
Nat Biotechnol ; 31(10): 898-907, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24013197

RESUMEN

In a cell-free approach to regenerative therapeutics, transient application of paracrine factors in vivo could be used to alter the behavior and fate of progenitor cells to achieve sustained clinical benefits. Here we show that intramyocardial injection of synthetic modified RNA (modRNA) encoding human vascular endothelial growth factor-A (VEGF-A) results in the expansion and directed differentiation of endogenous heart progenitors in a mouse myocardial infarction model. VEGF-A modRNA markedly improved heart function and enhanced long-term survival of recipients. This improvement was in part due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types. Direct in vivo comparison with DNA vectors and temporal control with VEGF inhibitors revealed the greatly increased efficacy of pulse-like delivery of VEGF-A. Our results suggest that modRNA is a versatile approach for expressing paracrine factors as cell fate switches to control progenitor cell fate and thereby enhance long-term organ repair.


Asunto(s)
Linaje de la Célula , Infarto del Miocardio/terapia , Miocardio/patología , ARN Mensajero/metabolismo , Regeneración , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Técnicas de Transferencia de Gen , Humanos , Cinética , Luciferasas/metabolismo , Ratones , Modelos Biológicos , Músculo Esquelético/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , ARN Mensajero/genética , Trasplante de Células Madre , Análisis de Supervivencia , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Cell Res ; 22(11): 1521-3, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22751090

RESUMEN

The term "lineage reprogramming" is typically used to describe the conversion of one differentiated somatic cell type into another without transit through a pluripotent intermediate. Two recent reports in Nature demonstrate that such a conversion can be achieved in the heart in situ, and suggest a novel, regenerative approach for the development of cardiac therapeutics.


Asunto(s)
Transdiferenciación Celular , Reprogramación Celular , Fibroblastos/citología , Insuficiencia Cardíaca/terapia , Corazón/fisiología , Miocitos Cardíacos/citología , Diferenciación Celular , Linaje de la Célula , Fibroblastos/fisiología , Humanos , Proteína MioD/biosíntesis , Miocitos Cardíacos/fisiología , Regeneración , Medicina Regenerativa/métodos
6.
Cell Res ; 22(1): 142-54, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064699

RESUMEN

Cardiomyocytes derived from pluripotent stem cells can be applied in drug testing, disease modeling and cell-based therapy. However, without procardiogenic growth factors, the efficiency of cardiomyogenesis from pluripotent stem cells is usually low and the resulting cardiomyocyte population is heterogeneous. Here, we demonstrate that induced pluripotent stem cells (iPSCs) can be derived from murine ventricular myocytes (VMs), and consistent with other reports of iPSCs derived from various somatic cell types, VM-derived iPSCs (ViPSCs) exhibit a markedly higher propensity to spontaneously differentiate into beating cardiomyocytes as compared to genetically matched embryonic stem cells (ESCs) or iPSCs derived from tail-tip fibroblasts. Strikingly, the majority of ViPSC-derived cardiomyocytes display a ventricular phenotype. The enhanced ventricular myogenesis in ViPSCs is mediated via increased numbers of cardiovascular progenitors at early stages of differentiation. In order to investigate the mechanism of enhanced ventricular myogenesis from ViPSCs, we performed global gene expression and DNA methylation analysis, which revealed a distinct epigenetic signature that may be involved in specifying the VM fate in pluripotent stem cells.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Alelos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Quimera/embriología , Quimera/genética , Quimera/metabolismo , Metilación de ADN , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Perfilación de la Expresión Génica , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Desarrollo de Músculos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología
7.
Nat Cell Biol ; 13(3): 191-3, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21364566

RESUMEN

The adult human heart lacks sufficient regenerative capacity to recover after a myocardial infarction. Cell-based therapy has emerged as a potential treatment for the failing heart; however, a key issue for the success of future cell-based therapies is the ability to obtain patient-specific high-quality cardiomyocytes in a fast and efficient manner. Recent progress has been made towards this goal using reprogramming-based approaches.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Animales , Técnicas de Cultivo de Célula , Fibroblastos/citología , Fibroblastos/metabolismo , Cardiopatías/patología , Humanos , Modelos Biológicos , Fosforilación , Medicina Regenerativa , Transducción de Señal , Células Madre/citología , Estrés Fisiológico
8.
Dev Biol ; 310(2): 329-40, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17825816

RESUMEN

Tbx1 is required for ear development in humans and mice. Gene manipulation in the mouse has discovered multiple consequences of loss of function on early development of the inner ear, some of which are attributable to a cell autonomous role in maintaining cell proliferation of epithelial progenitors of the cochlear and vestibular apparata. However, ablation of the mesodermal domain of the gene also results in severe but more restricted abnormalities. Here we show that Tbx1 has a dynamic expression during late development of the ear, in particular, is expressed in the sensory epithelium of the vestibular organs but not of the cochlea. Vice versa, it is expressed in the condensed mesenchyme that surrounds the cochlea but not in the one that surrounds the vestibule. Loss of Tbx1 in the mesoderm disrupts this peri-cochlear capsule by strongly reducing the proliferation of mesenchymal cells. The organogenesis of the cochlea, which normally occurs inside the capsule, was dramatically affected in terms of growth of the organ, as well as proliferation, differentiation and survival of its epithelial cells. This model provides a striking demonstration of the essential role played by the periotic mesenchyme in the organogenesis of the cochlea.


Asunto(s)
Cóclea/fisiología , Mesodermo/fisiología , Proteínas de Dominio T Box/biosíntesis , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Cóclea/embriología , Mesodermo/embriología , Ratones , Proteínas de Dominio T Box/genética
9.
Cell Stem Cell ; 1(2): 165-79, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-18371348

RESUMEN

Isl1(+) cardiovascular progenitors and their downstream progeny play a pivotal role in cardiogenesis and lineage diversification of the heart. The mechanisms that control their renewal and differentiation are largely unknown. Herein, we show that the Wnt/beta-catenin pathway is a major component by which cardiac mesenchymal cells modulate the prespecification, renewal, and differentiation of isl1(+) cardiovascular progenitors. This microenvironment can be reconstituted by a Wnt3a-secreting feeder layer with ES cell-derived, embryonic, and postnatal isl1(+) cardiovascular progenitors. In vivo activation of beta-catenin signaling in isl1(+) progenitors of the secondary heart field leads to their massive accumulation, inhibition of differentiation, and outflow tract (OFT) morphogenic defects. In addition, the mitosis rate in OFT myocytes is significantly reduced following beta-catenin deletion in isl1(+) precursors. Agents that manipulate Wnt signals can markedly expand isl1(+) progenitors from human neonatal hearts, a key advance toward the cloning of human isl1(+) heart progenitors.


Asunto(s)
Sistema Cardiovascular/embriología , Proteínas de Homeodominio/fisiología , Células Madre/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Sistema Cardiovascular/citología , Diferenciación Celular/fisiología , Linaje de la Célula , Embrión de Mamíferos/fisiología , Femenino , Corazón/embriología , Corazón/fisiología , Cardiopatías Congénitas/fisiopatología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas con Homeodominio LIM , Masculino , Ratones , Transducción de Señal , Células Madre/citología , Factores de Transcripción , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
Semin Cell Dev Biol ; 18(1): 77-83, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17178242

RESUMEN

Mammalian heart development requires multiple genetic networks, only some of which are becoming known in all their complexity. Substantial new information has become available thanks to an expanding toolkit that offers more and more mouse gene manipulation options, and that is taking the mouse closer to more powerful invertebrate genetic models. We review examples of recent data with a cardiac-lineage-based view of heart development, especially outflow tract and right ventricle. The medical significance of these studies is not only relevant to congenital heart disease, but also to the biology of cardiac cell regeneration.


Asunto(s)
Redes Reguladoras de Genes/fisiología , Corazón/embriología , Mamíferos/embriología , Mamíferos/genética , Morfogénesis/genética , Animales , Linaje de la Célula , Humanos , Ratones , Ratones Transgénicos , Modelos Cardiovasculares
11.
Dev Biol ; 302(2): 670-82, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17074316

RESUMEN

The T-box transcription factor Tbx1 is required for inner ear morphogenesis. Tbx1 null mutants have a small otocyst that fails to grow and remodel and does not give rise to the vestibular and cochlear apparata. Here we show that Tbx1 expression-driven cell tracing identifies a population of otic epithelial cells that contributes to most of the otocyst. Tbx1 is essential for the contribution of this population to the inner ear. Ablation of Tbx1 after this cell population has established itself in the otocyst, restores marker expression lost in germ line mutants, but causes severe reduction in mitotic activity, cell autonomously. Furthermore, timed cell fate mapping demonstrates that loss of Tbx1 switches the fate of some members of the Tbx1-dependent cell population, from non-neurogenic to neurogenic, an event associated with activation of the Delta-Notch pathway. Finally, tissue-specific ablation of Tbx1 demonstrates that, while the abovementioned phenotypic abnormalities are due to loss of epithelial expression of Tbx1, cochlear morphogenesis requires mesodermal Tbx1 expression. We conclude that the main functions of Tbx1 in the inner ear are to control, cell-autonomously, contribution, size and fate of a large population of otic epithelial cells, and, cell non-autonomously, cochlear morphogenesis.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Oído Interno/citología , Células Epiteliales/citología , Proteínas de Dominio T Box/fisiología , Animales , Células Epiteliales/fisiología , Ratones , Ratones Transgénicos , Mutación , Proteínas de Dominio T Box/genética
12.
Development ; 132(23): 5307-15, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16284121

RESUMEN

During embryonic life, the initially paired pharyngeal arch arteries (PAAs) follow a precisely orchestrated program of persistence and regression that leads to the formation of the mature aortic arch and great vessels. When this program fails, specific cardiovascular defects arise that may be life threatening or mild, according to the identity of the affected artery. Fourth PAA-derived cardiovascular defects occur commonly in DiGeorge syndrome and velocardiofacial syndrome (22q11DS), and in Tbx1(+/-) mice that model the 22q11DS cardiovascular phenotype. Tbx1 is expressed in pharyngeal mesoderm, endoderm and ectoderm, and, in addition, we show that it is expressed in precursors of the endothelial cells that line the PAAs, thus expanding the number of tissues in which Tbx1 is potentially required for fourth PAA development. In this study, we have used cell fate mapping and tissue-specific gene deletion, driven by six different Cre lines, to explore Tbx1 gene-dosage requirements in the embryonic pharynx for fourth PAA development. Through this approach, we have resolved the spatial requirements for Tbx1 in this process, and we show pharyngeal epithelia to be a critical tissue. We also thereby demonstrate conclusively that the role of Tbx1 in fourth PAA development is cell non-autonomous.


Asunto(s)
Arterias/crecimiento & desarrollo , Región Branquial/irrigación sanguínea , Epitelio/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Animales , Aorta Torácica , Embrión de Mamíferos , Epitelio/química , Eliminación de Gen , Dosificación de Gen , Ratones , Ratones Endogámicos , Faringe/embriología
13.
Development ; 132(19): 4387-95, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16141220

RESUMEN

The definition of time-specific requirements for a developmental gene can pinpoint the processes within which the gene is involved and can reveal potential late functions in structures and organs that fail to develop in germline mutants. Here, we show the first systematic time-course deletion, in parallel with timed cell fate mapping, of a developmentally crucial gene, Tbx1, during mouse embryogenesis. Tbx1 mouse mutants model DiGeorge syndrome, a disorder of pharyngeal and cardiovascular development. Results revealed different time requirements for the development of individual structures, as well as multiple and time-distinct roles during the development of the same organ or system. We also show that Tbx1 is required throughout pharyngeal segmentation for the regulation of endoderm expansion, thus this is the first gene implicated directly in this process. A genetic-based blueprint of crucial developmental times for organs and systems should be a valuable asset for our understanding of birth defect pathogenesis.


Asunto(s)
Síndrome de DiGeorge/embriología , Síndrome de DiGeorge/metabolismo , Endodermo/citología , Proteínas de Dominio T Box/metabolismo , Animales , Tipificación del Cuerpo , Encéfalo/citología , Encéfalo/embriología , Encéfalo/metabolismo , Región Branquial/embriología , Proliferación Celular , Síndrome de DiGeorge/genética , Desarrollo Embrionario , Endodermo/metabolismo , Integrasas/genética , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas de Dominio T Box/genética
14.
Development ; 131(13): 3217-27, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15175244

RESUMEN

Dysmorphogenesis of the cardiac outflow tract (OFT) causes many congenital heart defects, including those associated with DiGeorge syndrome. Genetic manipulation in the mouse and mutational analysis in patients have shown that Tbx1, a T-box transcription factor, has a key role in the pathogenesis of this syndrome. Here, we have dissected Tbx1 function during OFT development using genetically modified mice and tissue-specific deletion, and have defined a dual role for this protein in OFT morphogenesis. We show that Tbx1 regulates cell contribution to the OFT by supporting cell proliferation in the secondary heart field, a source of cells fated to the OFT. This process might be regulated in part by Fgf10, which we show for the first time to be a direct target of Tbx1 in vitro. We also show that Tbx1 expression is required in cells expressing Nkx2.5 for the formation of the aorto-pulmonary septum, which divides the aorta from the main pulmonary artery. These results explain why aortic arch patterning defects and OFT defects can occur independently in individuals with DiGeorge syndrome. Furthermore, our data link, for the first time, the function of the secondary heart field to congenital heart disease.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Miocardio/metabolismo , Proteínas de Dominio T Box/fisiología , Alelos , Animales , Bromodesoxiuridina/farmacología , Diferenciación Celular , División Celular , Colorantes/farmacología , Análisis Mutacional de ADN , Síndrome de DiGeorge/genética , Células Endoteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Eliminación de Gen , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Luciferasas/metabolismo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Mutación , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...