Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Genes Dev ; 25(3): 251-62, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245161

RESUMEN

Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development. We show that, from the fetal period, ERG acts as a direct upstream regulator of Gata2 and Runx1 gene activity. Without ERG, physiological HSC maintenance fails, leading to the rapid exhaustion of definitive hematopoiesis.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas Oncogénicas/metabolismo , Animales , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA2/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas/genética , Factores de Transcripción , Regulador Transcripcional ERG
2.
Blood ; 115(15): 3042-50, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20139099

RESUMEN

The transcription factor Runx1 is a pivotal regulator of definitive hematopoiesis in mouse ontogeny. Vertebrate Runx1 is transcribed from 2 promoters, the distal P1 and proximal P2, which provide a paradigm of the complex transcriptional and translational control of Runx1 function. However, very little is known about the biologic relevance of alternative Runx1 promoter usage in definitive hematopoietic cell emergence. Here we report that both promoters are active at the very onset of definitive hematopoiesis, with a skewing toward the P2. Moreover, functional and morphologic analysis of a novel P1-null and an attenuated P2 mouse model revealed that although both promoters play important nonredundant roles in the emergence of definitive hematopoietic cells, the proximal P2 was most critically required for this. The nature of the observed phenotypes is indicative of a differential contribution of the P1 and P2 promoters to the control of overall Runx1 levels, where and when this is most critically required. In addition, the dynamic expression of P1-Runx1 and P2-Runx1 points at a requirement for Runx1 early in development, when the P2 is still the prevalent promoter in the emerging hemogenic endothelium and/or first committed hematopoietic cells.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Hematopoyesis/genética , Regiones Promotoras Genéticas/genética , Envejecimiento/genética , Alelos , Animales , Aorta/metabolismo , Aorta/patología , Células de la Médula Ósea/metabolismo , Cadherinas/metabolismo , Agregación Celular , Recuento de Células , Ensayo de Unidades Formadoras de Colonias , Subunidad alfa 2 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Pérdida del Embrión/genética , Pérdida del Embrión/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Sitios Genéticos/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones , Modelos Genéticos , Mutación/genética
3.
Blood ; 113(21): 5121-4, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19321859

RESUMEN

The transcription factor Runx1 plays a pivotal role in hematopoietic stem cell (HSC) emergence, and studies into its transcriptional regulation should give insight into the critical steps of HSC specification. Recently, we identified the Runx1 +23 enhancer that targets reporter gene expression to the first emerging HSCs of the mouse embryo when linked to the heterologous hsp68 promoter. Endogenous Runx1 is transcribed from 2 alternative promoters, P1 and P2. Here, we examined the in vivo cis-regulatory potential of these alternative promoters and asked whether they act with and contribute to the spatiotemporal specific expression of the Runx1 +23 enhancer. Our results firmly establish that, in contrast to zebrafish runx1, mouse Runx1 promoter sequences do not confer any hematopoietic specificity in transgenic embryos. Yet, both mouse promoters act with the +23 enhancer to drive reporter gene expression to sites of HSC emergence and colonization, in a +23-specific pattern.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Elementos de Facilitación Genéticos , Células Madre Hematopoyéticas/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Animales , Embrión de Mamíferos , Células Madre Hematopoyéticas/citología , Ratones , Ratones Transgénicos , Especificidad de la Especie
4.
Mol Pharmacol ; 78(4): 685-92, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20610734

RESUMEN

The new immunosuppressant FTY720 (fingolimod), an analog of the endogenous lipid sphingosine, induces transient lymphopenia through the sequestration of lymphocytes in secondary lymphoid organs. Phosphorylation of FTY720 by sphingosine kinase 2 (SphK2) yields the active metabolite FTY720-phosphate (FTY-P), which induces lymphopenia through agonism of the sphingosine 1-phosphate receptor S1P(1) on endothelial cells and lymphocytes. Dephosphorylation of circulating FTY-P creates an equilibrium between FTY720 and its phosphate, and results with human patients indicate that phosphorylation of FTY720 could be rate limiting for efficacy. We report that the FTY720 derivative 2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol [AAL(R)] is phosphorylated much more rapidly than FTY720 in cultured human cells and whole blood. The K(cat) for AAL(R) with recombinant SphK2 is 8-fold higher than for FTY720, whereas the K(m) for the two substrates is very similar, indicating that the increased rate of phosphorylation results from faster turnover by SphK2 rather than a higher binding affinity. Consequently, treating cells with AAL(R), but not FTY720, triggers an apoptotic pathway that is dependent on excessive intracellular accumulation of long-chain base phosphates. In agreement with the in vitro results, phosphorylation of AAL(R) is more complete than that of FTY720 in vivo (mice), and AAL(R) is a more potent inducer of lymphopenia. These differences may be magnified in humans, because phosphorylation of FTY720 is much less efficient in humans compared with rodents. Our results suggest that AAL(R) is a better tool than FTY720 for in vivo studies with S1P analogs and would probably be a more effective immunosuppressant than FTY720.


Asunto(s)
Química Farmacéutica/métodos , Radical Hidroxilo/química , Glicoles de Propileno/química , Glicoles de Propileno/metabolismo , Esfingosina/análogos & derivados , Animales , Línea Celular , Células Cultivadas , Clorhidrato de Fingolimod , Células HeLa , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Fosforilación/fisiología , Ratas , Esfingosina/química , Esfingosina/metabolismo
5.
Blood Cells Mol Dis ; 43(1): 35-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19464215

RESUMEN

The interest in stem cell based therapies has emphasized the importance of understanding the cellular and molecular mechanisms by which stem cells are generated in ontogeny and maintained throughout adult life. Hematopoietic stem cells (HSCs) are first found in clusters of hematopoietic cells budding from the luminal wall of the major arteries in the developing mammalian embryo. The transcription factor Runx1 is critical for their generation and is specifically expressed at sites of HSC generation, prior to their formation. To understand better the transcriptional hierarchies that converge on Runx1 during HSC emergence, we have initiated studies into its transcriptional regulation. Here we systematically analyzed Runx1 P1 and P2 alternative promoter usage in hematopoietic sites and in sorted cell populations during mouse hematopoietic development. Our results indicate that Runx1 expression in primitive erythrocytes is largely P2-derived, whilst in definitive hematopoietic stem and/or progenitor cells from the yolk sac or AGM and vitelline and umbilical arteries both the distal P1 and proximal P2 promoters are active. After cells have migrated to the fetal liver, the P1 gradually becomes the main hematopoietic promoter and remains this into adulthood. In addition, we identified a novel P2-derived Runx1 isoform.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , Regiones Promotoras Genéticas , Animales , Aorta/citología , Aorta/embriología , Aorta/fisiología , Secuencia de Bases , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Humanos , Hígado/citología , Hígado/embriología , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/citología , Placenta/embriología , Placenta/fisiología , Embarazo , Alineación de Secuencia , Transcripción Genética , Saco Vitelino/citología , Saco Vitelino/embriología , Saco Vitelino/fisiología
6.
BMC Cancer ; 9: 383, 2009 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-19874621

RESUMEN

BACKGROUND: Testicular germ cell tumors (TGCTs) are classified as seminonas or non-seminomas of which a major subset is embryonal carcinoma (EC) that can differentiate into diverse tissues. The pluripotent nature of human ECs resembles that of embryonic stem (ES) cells. Many Wnt signalling species are regulated during differentiation of TGCT-derived EC cells. This study comprehensively investigated expression profiles of Wnt signalling components regulated during induced differentiation of EC cells and explored the role of key components in maintaining pluripotency. METHODS: Human embryonal carcinoma cells were stably infected with a lentiviral construct carrying a canonical Wnt responsive reporter to assess Wnt signalling activity following induced differentiation. Cells were differentiated with all-trans retinoic acid (RA) or by targeted repression of pluripotency factor, POU5F1. A Wnt pathway real-time-PCR array was used to evaluate changes in gene expression as cells differentiated. Highlighted Wnt pathway genes were then specifically repressed using siRNA or stable shRNA and transfected EC cells were assessed for proliferation, differentiation status and levels of core pluripotency genes. RESULTS: Canonical Wnt signalling activity was low basally in undifferentiated EC cells, but substantially increased with induced differentiation. Wnt pathway gene expression levels were compared during induced differentiation and many components were altered including ligands (WNT2B), receptors (FZD5, FZD6, FZD10), secreted inhibitors (SFRP4, SFRP1), and other effectors of Wnt signalling (FRAT2, DAAM1, PITX2, Porcupine). Independent repression of FZD5, FZD7 and WNT5A using transient as well as stable methods of RNA interference (RNAi) inhibited cell growth of pluripotent NT2/D1 human EC cells, but did not appreciably induce differentiation or repress key pluripotency genes. Silencing of FZD7 gave the greatest growth suppression in all human EC cell lines tested including NT2/D1, NT2/D1-R1, Tera-1 and 833K cells. CONCLUSION: During induced differentiation of human EC cells, the Wnt signalling pathway is reprogrammed and canonical Wnt signalling induced. Specific species regulating non-canonical Wnt signalling conferred growth inhibition when targeted for repression in these EC cells. Notably, FZD7 repression significantly inhibited growth of human EC cells and is a promising therapeutic target for TGCTs.


Asunto(s)
Carcinoma Embrionario/metabolismo , Diferenciación Celular , Transducción de Señal , Proteínas Wnt/metabolismo , Carcinoma Embrionario/tratamiento farmacológico , Carcinoma Embrionario/genética , Carcinoma Embrionario/fisiopatología , Línea Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética
7.
Mol Biol Cell ; 16(12): 5630-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16195345

RESUMEN

Differentiation of pluripotent embryonic stem (ES) cells through multipotent neural stem (NS) cells into differentiated neurons is accompanied by wholesale changes in transcriptional programs. One factor that is present at all three stages and a key to neuronal differentiation is the RE1-silencing transcription factor (REST/NRSF). Here, we have used a novel chromatin immunoprecipitation-based cloning strategy (SACHI) to identify 89 REST target genes in ES cells, embryonic hippocampal NS cells and mature hippocampus. The gene products are involved in all aspects of neuronal function, especially neuronal differentiation, axonal growth, vesicular transport and release, and ionic conductance. Most target genes are silent or expressed at low levels in ES and NS cells, but are expressed at much higher levels in hippocampus. These data indicate that the REST regulon is specific to each developmental stage and support the notion that REST plays distinct roles in regulating gene expression in pluripotent ES cells, multipotent NS cells, and mature neurons.


Asunto(s)
Neuronas/fisiología , Proteínas Represoras/metabolismo , Células Madre/fisiología , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Cromatina/fisiología , Cromatina/ultraestructura , Clonación Molecular , Secuencia de Consenso , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética
8.
Int J Oncol ; 30(2): 333-40, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17203214

RESUMEN

Metastatic germ cell tumors (GCT) are curable, however GCTs refractory to cisplatin-based chemotherapy have a poor prognosis. This study explores D-type cyclins as molecular targets in GCTs because all-trans-retinoic acid (RA)-mediated differentiation of the human embryonal carcinoma (EC) cell line NT2/D1 is associated with G1 cell cycle arrest and proteasomal degradation of cyclin D1. RA effects on D-type cyclins are compared in human EC cells that are RA sensitive or dually RA and cisplatin resistant (NT2/D1-R1) and in clinical GCTs that have both EC and mature teratoma components. Notably, GCT differentiation was associated with reduced cyclin D1 but increased cyclin D3 expression. RA was shown here to repress cyclin D1 through a transcriptional mechanism in addition to causing its degradation. The siRNA-mediated repression of individual cyclin D species resulted in growth inhibition in both RA sensitive and resistant EC cells. Only repression of cyclin D1 occurred in vitro and when clinical GCTs mature, implicating cyclin D1 as a molecular therapeutic target. To confirm this, the EGFR-tyrosine kinase inhibitor, Erlotinib, was used to repress cyclin D1. This inhibited proliferation in RA and cisplatin sensitive and resistant EC cells. Taken together, these findings implicate cyclin D1 targeting agents for the treatment of GCTs.


Asunto(s)
Antineoplásicos/farmacología , Ciclina D1/antagonistas & inhibidores , Ciclina D1/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Neoplasias de Células Germinales y Embrionarias/metabolismo , Diferenciación Celular , Fragmentación del ADN , Inhibidores Enzimáticos/farmacología , Clorhidrato de Erlotinib , Humanos , Neoplasias de Células Germinales y Embrionarias/patología , Quinazolinas/farmacología , ARN Nuclear Heterogéneo/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Ácido Retinoico/metabolismo , Factores de Tiempo , Tretinoina/metabolismo
9.
Mol Cell Biol ; 35(12): 2165-72, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870111

RESUMEN

The bone morphogenetic protein (BMP)/SMAD signaling pathway is a critical regulator of angiogenic sprouting and is involved in vascular development in the embryo. SMAD1 and SMAD5, the core mediators of BMP signaling, are vital for this activity, yet little is known about their transcriptional regulation in endothelial cells. Here, we have integrated multispecies sequence conservation, tissue-specific chromatin, in vitro reporter assay, and in vivo transgenic data to identify and validate Smad1+63 and the Smad5 promoter as tissue-specific cis-regulatory elements that are active in the developing endothelium. The activity of these elements in the endothelium was dependent on highly conserved ETS, GATA, and E-box motifs, and chromatin immunoprecipitation showed high levels of enrichment of FLI1, GATA2, and SCL at these sites in endothelial cell lines and E11 dorsal aortas in vivo. Knockdown of FLI1 and GATA2 but not SCL reduced the expression of SMAD1 and SMAD5 in endothelial cells in vitro. In contrast, CD31(+) cKit(-) endothelial cells harvested from embryonic day 9 (E9) aorta-gonad-mesonephros (AGM) regions of GATA2 null embryos showed reduced Smad1 but not Smad5 transcript levels. This is suggestive of a degree of in vivo selection where, in the case of reduced SMAD1 levels, endothelial cells with more robust SMAD5 expression have a selective advantage.


Asunto(s)
Endotelio/embriología , Factor de Transcripción GATA2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Smad1/genética , Proteína Smad5/genética , Animales , Secuencia de Bases , Línea Celular , Endotelio/metabolismo , Factor de Transcripción GATA2/genética , Técnicas de Silenciamiento del Gen , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-fli-1/genética
10.
Nat Commun ; 4: 2924, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24326267

RESUMEN

Haematopoietic stem cells (HSCs) are the founding cells of the adult haematopoietic system, born during ontogeny from a specialized subset of endothelium, the haemogenic endothelium (HE) via an endothelial-to-haematopoietic transition (EHT). Although recently imaged in real time, the underlying mechanism of EHT is still poorly understood. We have generated a Runx1 +23 enhancer-reporter transgenic mouse (23GFP) for the prospective isolation of HE throughout embryonic development. Here we perform functional analysis of over 1,800 and transcriptional analysis of 268 single 23GFP(+) HE cells to explore the onset of EHT at the single-cell level. We show that initiation of the haematopoietic programme occurs in cells still embedded in the endothelial layer, and is accompanied by a previously unrecognized early loss of endothelial potential before HSCs emerge. Our data therefore provide important insights on the timeline of early haematopoietic commitment.


Asunto(s)
Embrión de Mamíferos/citología , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos/citología , Hemangioblastos/fisiología , Análisis de la Célula Individual/métodos , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Elementos de Facilitación Genéticos , Femenino , Proteínas Fluorescentes Verdes/genética , Masculino , Ratones , Ratones Transgénicos , Embarazo
11.
Mol Cell Biol ; 31(14): 2817-26, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576367

RESUMEN

The oncogenic transcription factor Runx1 is required for the specification of definitive hematopoietic stem cells (HSC) in the developing embryo. The activity of this master regulator is tightly controlled during development. The transcription factors that upregulate the expression of Runx1 also upregulate the expression of Smad6, the inhibitory Smad, which controls Runx1 activity by targeting it to the proteasome. Here we show that Runx1, in conjunction with Fli1, Gata2, and Scl, directly regulates the expression of Smad6 in the aorta-gonad-mesonephros (AGM) region in the developing embryo, where HSCs originate. Runx1 regulates Smad6 activity via a novel upstream enhancer, and Runx1 null embryos show reduced Smad6 transcripts in the yolk-sac and c-Kit-positive fetal liver cells. By directly regulating the expression of Smad6, Runx1 sets up a functional rheostat to control its own activity. The perturbation of this rheostat, using a proteasomal inhibitor, results in an increase in Runx1 and Smad6 levels that can be directly attributed to increased Runx1 binding to tissue-specific regulatory elements of these genes. Taken together, we describe a scenario in which a key hematopoietic transcription factor controls its own expression levels by transcriptionally controlling its controller.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Proteína smad6/metabolismo , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Embrión de Mamíferos/anatomía & histología , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Células K562 , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos , Proteína smad6/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA