Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 23(4): 555-64, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25526092

RESUMEN

Epigenetic changes on DNA and chromatin are implicated in cell differentiation and organogenesis. For the heart, distinct histone methylation profiles were recently linked to stage-specific gene expression programs during cardiac differentiation in vitro. However, the enzymes catalyzing these modifications and the genes regulated by them remain poorly defined. We therefore decided to identify the epigenetic enzymes that are potentially involved in cardiomyogenesis by analyzing the expression profile of the 85 genes encoding the epigenetic-related proteins in mouse cardiomyocytes (CMs), and then study how they affect gene expression during differentiation and maturation of this cell type. We show here with gene expression screening of epigenetic enzymes that the highly expressed H3 methyltransferase disruptor of telomeric silencing 1-like (DOT1L) drives a transitional pattern of di-methylation on H3 lysine 79 (H3K79) in CMs at different stages of differentiation in vitro and in vivo. Through a genome-wide chromatin-immunoprecipitation DNA-sequencing approach, we found H3K79me2 enriched at genes expressed during cardiac differentiation. Moreover, knockdown of Dot1L affected the expression of H3K79me2-enriched genes. Our results demonstrate that histone methylation, and in particular DOT1L-mediated H3K79me2 modification, drives cardiomyogenesis through the definition of a specific transcriptional landscape.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Histonas/metabolismo , Metiltransferasas/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Línea Celular , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Metiltransferasas/genética , Ratones
2.
Cell Death Dis ; 5: e1053, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24525729

RESUMEN

Cell-based regenerative therapies are significantly improved by engineering allografts to express factors that increase vascularization and engraftment, such as placental growth factor (PlGF) and matrix metalloproteinase 9 (MMP9). Moreover, the seeding of therapeutic cells onto a suitable scaffold is of utmost importance for tissue regeneration. On these premises, we sought to assess the reparative potential of induced pluripotent stem (iPS) cells bioengineered to secrete PlGF or MMP9 and delivered to infarcted myocardium upon a poly(ethylene glycol)-fibrinogen scaffold. When assessing optimal stiffness of the PEG-fibrinogen (PF) scaffold, we found that the appearance of contracting cells after cardiogenic induction was accelerated on the support designed with an intermediate stiffness. Revascularization and hemodynamic parameters of infarcted mouse heart were significantly improved by injection into the infarct of this optimized PF scaffold seeded with both MiPS (iPS cells engineered to secrete MMP9) and PiPS (iPS cells engineered to secrete PlGF) cells as compared with nonengineered cells or PF alone. Importantly, allograft-derived cells and host myocardium were functionally integrated. Therefore, survival and integration of allografts in the ischemic heart can be significantly improved with the use of therapeutic cells bioengineered to secrete MMP9 and PlGF and encapsulated within an injectable PF hydrogel having an optimized stiffness.


Asunto(s)
Fibrinógeno/química , Ingeniería Genética , Células Madre Pluripotentes Inducidas/trasplante , Metaloproteinasa 9 de la Matriz/metabolismo , Infarto del Miocardio/prevención & control , Miocardio/enzimología , Miocitos Cardíacos/trasplante , Polietilenglicoles/química , Proteínas Gestacionales/metabolismo , Regeneración , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hemodinámica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Contracción Miocárdica , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocitos Cardíacos/enzimología , Neovascularización Fisiológica , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Recuperación de la Función , Factores de Tiempo , Transducción Genética , Transfección
3.
Gene Ther ; 19(6): 642-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22378345

RESUMEN

Engineered recombinant viral vectors are a powerful tool for vehiculating genetic information into mammalian cells. Because of their ability to infect both dividing and non-dividing cells with high efficiency, lentiviral vectors have gained particular interest for basic research and preclinical studies in the cardiovascular field. We review here the major applications for lentiviral-vector technology in the cardiovascular field: we will discuss their use in trailing gene expression during the induction of differentiation, in protocols for the isolation of cardiac cells and in the tracking of cardiac cells after transplantation in vivo; we will also describe lentivirally-mediated gene delivery uses, such as the induction of a phenotype of interest in a target cell or the treatment of cardiovascular diseases. In addition, a section of the review will be dedicated to reprogramming approaches, focusing attention on the generation of pluripotent stem cells and on transdifferentiation, two emerging strategies for the production of cardiac myocytes from human cells and for the investigation of human diseases. Finally, in order to give a perspective on their future clinical use we will critically discuss advantages and disadvantages of lentivirus-based strategies for the treatment of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Lentivirus/genética , Miocitos Cardíacos/fisiología , Diferenciación Celular , Transdiferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/trasplante
4.
Cell Death Differ ; 19(7): 1162-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22261617

RESUMEN

Adult mammalian cells can be reprogrammed to a pluripotent state by forcing the expression of a few embryonic transcription factors. The resulting induced pluripotent stem (iPS) cells can differentiate into cells of all three germ layers. It is well known that post-natal cardiomyocytes (CMs) lack the capacity to proliferate. Here, we report that neonatal CMs can be reprogrammed to generate iPS cells that express embryonic-specific markers and feature gene-expression profiles similar to those of mouse embryonic stem (mES) cell and cardiac fibroblast (CF)-derived iPS cell populations. CM-derived iPS cells are able to generate chimeric mice and, moreover, re-differentiate toward CMs more efficiently then either CF-derived iPS cells or mES cells. The increased differentiation capacity is possibly related to CM-derived iPS cells retaining an epigenetic memory of the phenotype of their founder cell. CM-derived iPS cells may thus lead to new information on differentiation processes underlying cardiac differentiation and proliferation.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Animales , Proteína Morfogenética Ósea 2/farmacología , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Reprogramación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo
5.
Curr Drug Targets ; 11(8): 907-12, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20415655

RESUMEN

Heart ion-channel function and expression are continuously being regulated on the basis of the hemodynamic state of the cardiovascular system, the neurohumoral milieu and the properties of the ongoing ionic fluxes. These homeostatic forces act through multiple mechanisms at transcriptional, translational and post-translational levels. Of clinical importance is the fact that with adverse stress these regulatory mechanisms can produce arrhythmogenic channel remodelling. Although a great deal is known about the functionality of ion channels and the generation of the action potential, much less is known about the underlying controlling mechanisms and how these become derailed during disease. microRNA-mediated posttranscriptional control is a very recent addition to cardiovascular biology. Here, we outline discoveries pertaining to these new regulators and how they might be involved in cardiac electrophysiology and pathology.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Sistema de Conducción Cardíaco/metabolismo , MicroARNs/metabolismo , Potenciales de Acción , Animales , Arritmias Cardíacas/fisiopatología , Electrofisiología Cardíaca , Humanos , Canales Iónicos/metabolismo
6.
Cell Death Differ ; 16(12): 1590-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19816508

RESUMEN

Mechanisms controlling vascular smooth muscle cell (VSMC) plasticity and renewal still remain to be elucidated completely. A class of small RNAs called microRNAs (miRs) regulate gene expression at the post-transcriptional level. Here, we show a critical role of the miR-143/145 cluster in SMC differentiation and vascular pathogenesis, also through the generation of a mouse model of miR-143 and -145 knockout (KO). We determined that the expression of miR-143 and -145 is decreased in acute and chronic vascular stress (transverse aortic constriction and in aortas of the ApoE KO mouse). In human aortic aneurysms, the expression of miR-143 and -145 was significantly decreased compared with control aortas. In addition, overexpression of miR-143 and -145 decreased neointimal formation in a rat model of acute vascular injury. An in-depth analysis of the miR-143/145 KO mouse model showed that this miR cluster is expressed mostly in the SMC compartment, both during development and postnatally, in vessels and SMC-containing organs. Loss of miR-143 and miR-145 expression induces structural modifications of the aorta, because of an incomplete differentiation of VSMCs. In conclusion, our results show that the miR-143/145 gene cluster has a critical role during SMC differentiation and strongly suggest its involvement in the reversion of the VSMC differentiation phenotype that occurs during vascular disease.


Asunto(s)
Diferenciación Celular , Homeostasis , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Secuencia de Bases , Línea Celular , Proliferación Celular , Humanos , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Ratas
7.
Gene Ther ; 15(3): 161-70, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18033312

RESUMEN

Human embryonic stem cells (hESCs) may become important for cardiac repair due to their potentially unlimited ability to generate cardiomyocytes (CMCs). Moreover, genetic manipulation of hESC-derived CMCs would be a very promising technique for curing myocardial disorders. At the present time, however, inducing the differentiation of hESCs into CMCs is extremely difficult and, therefore, an easy and standardizable technique is needed to evaluate differentiation strategies. Vectors driving cardiac-specific expression may represent an important tool not only for monitoring new cardiac-differentiation strategies, but also for the manipulation of cardiac differentiation of ESCs. To this aim, we generated cardiac-specific lentiviral vectors (LVVs) in which expression is driven by a short fragment of the cardiac troponin-I proximal promoter (TNNI3) with a human cardiac alpha-actin enhancer, and tested its suitability in inducing tissue-specific gene expression and ability to track the CMC lineage during differentiation of ESCs. We determined that (1) TNNI3-LVVs efficiently drive cardiac-specific gene expression and mark the cardiomyogenic lineage in human and mouse ESC differentiation systems (2) the cardiac alpha-actin enhancer confers a further increase in gene-expression specificity of TNNI3-LVVs in hESCs. Although this technique may not be useful in tracking small numbers of cells, data suggested that TNNI3-based LVVs are a powerful tool for manipulating human ESCs and modifying hESC-derived CMCs.


Asunto(s)
Células Madre Embrionarias/citología , Terapia Genética/métodos , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/citología , Actinas/genética , Animales , Diferenciación Celular , Línea Celular , Elementos de Facilitación Genéticos , Citometría de Flujo , Ingeniería Genética , Vectores Genéticos/farmacología , Humanos , Lentivirus/genética , Ratones , Regiones Promotoras Genéticas , Transducción Genética/métodos , Troponina I/genética
9.
Gene Ther ; 10(8): 630-6, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12692591

RESUMEN

Efficient gene transduction in cardiomyocytes is a task that can be accomplished only by viral vectors. Up to now, the most commonly used vectors for this purpose have been adenoviral-derived ones. Recently, it has been demonstrated that lentiviral vectors can transduce growth-arrested cells, such as hematopoietic stem cells. Moreover, a modified form of lentiviral vector (the 'advanced' generation), containing an mRNA-stabilizer sequence and a nuclear import sequence, has been shown to significantly improve gene transduction in growth-arrested cells as compared to the third-generation vector. Therefore, we tested whether the 'advanced' generation lentivirus is capable of infecting and transducing cardiomyocytes both in vitro and in vivo, comparing efficacy in vitro against the third-generation of the same vector. Here we report that 'advanced' generation lentiviral vectors infected most (>80%) cardiomyocytes in culture, as demonstrated by immunofluorescence and FACS analyses: in contrast the percentage of cardiomyocytes infected by third-generation lentivirus was three- to four-fold lower. Moreover, 'advanced' generation lentivirus was also capable of infecting and inducing stable gene expression in adult myocardium in vivo. Thus, 'advanced' generation lentiviral vectors can be used for both in vitro and in vivo gene expression studies in the cardiomyocyte.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Terapia Genética/métodos , Vectores Genéticos/farmacología , Lentivirus/genética , Miocitos Cardíacos/metabolismo , Transducción Genética/métodos , Animales , Línea Celular , Citometría de Flujo , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...