Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
EMBO Rep ; 13(4): 355-62, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22402664

RESUMEN

The transcription factor Eomesodermin (Eomes) is involved in early embryonic patterning, but the range of cell fates that it controls as well as its mechanisms of action remain unclear. Here we show that transient expression of Eomes promotes cardiovascular fate during embryonic stem cell differentiation. Eomes also rapidly induces the expression of Mesp1, a key regulator of cardiovascular differentiation, and directly binds to regulatory sequences of Mesp1. Eomes effects are strikingly modulated by Activin signalling: high levels of Activin inhibit the promotion of cardiac mesoderm by Eomes, while they enhance Eomes-dependent endodermal specification. These results place Eomes upstream of the Mesp1-dependent programme of cardiogenesis, and at the intersection of mesodermal and endodermal specification, depending on the levels of Activin/Nodal signalling.


Asunto(s)
Activinas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Regulación de la Expresión Génica/efectos de los fármacos , Miocardio/citología , Proteínas de Dominio T Box/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Organogénesis/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Dominio T Box/genética
2.
Nat Cell Biol ; 24(7): 1114-1128, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35817961

RESUMEN

The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP-seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its transcription-factor activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cromatina , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Corazón , Proteínas de Homeodominio/metabolismo , Mamíferos/metabolismo , Mesodermo , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Sci Adv ; 6(2): eaax5849, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31934626

RESUMEN

Psoriasis is a common chronic skin disorder characterized by keratinocyte hyperproliferation with altered differentiation accompanied by inflammation and increased angiogenesis. It remains unclear whether the first events that initiate psoriasis development occur in keratinocytes or inflammatory cells. Here, using different psoriasis mouse models, we showed that conditional deletion of Flt1 or Nrp1 in epidermal cells inhibited psoriasis mediated by Vegfa overexpression or c-Jun/JunB deletion. Administration of anti-Nrp1 antibody reverted the psoriasis phenotype. Using transcriptional and chromatin profiling of epidermal cells following Vegfa overexpression together with Flt1 or Nrp1 deletion, we identified the gene regulatory network regulated by Vegfa/Nrp1/Flt1 during psoriasis development and uncovered a key role of Fosl1 in regulating the chromatin remodeling mediated by Vegfa overexpression in keratinocytes. In conclusion, our study identifies an epidermal autonomous function of Vegfa/Nrp1/Flt1 that mediates psoriatic-like disease and demonstrates the clinical relevance of blocking Vegfa/Nrp1/Flt1 axis in psoriasis.


Asunto(s)
Células Epidérmicas/metabolismo , Neuropilina-1/metabolismo , Psoriasis/metabolismo , Psoriasis/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Células Epidérmicas/efectos de los fármacos , Células Epidérmicas/patología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones Noqueados , Fenotipo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Psoriasis/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
4.
Science ; 359(6380): 1177-1181, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29371425

RESUMEN

Mouse heart development arises from Mesp1-expressing cardiovascular progenitors (CPs) that are specified during gastrulation. The molecular processes that control early regional and lineage segregation of CPs have been unclear. We performed single-cell RNA sequencing of wild-type and Mesp1-null CPs in mice. We showed that populations of Mesp1 CPs are molecularly distinct and span the continuum between epiblast and later mesodermal cells, including hematopoietic progenitors. Single-cell transcriptome analysis of Mesp1-deficient CPs showed that Mesp1 is required for the exit from the pluripotent state and the induction of the cardiovascular gene expression program. We identified distinct populations of Mesp1 CPs that correspond to progenitors committed to different cell lineages and regions of the heart, identifying the molecular features associated with early lineage restriction and regional segregation of the heart at the early stage of mouse gastrulation.


Asunto(s)
Corazón/embriología , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , División Celular , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mesodermo/citología , Ratones , Ratones Mutantes , ARN/genética , Análisis de Secuencia de ARN , Células Madre/metabolismo
5.
J Cell Biol ; 213(4): 463-77, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27185833

RESUMEN

During embryonic development, Mesp1 marks the earliest cardiovascular progenitors (CPs) and promotes their specification, epithelial-mesenchymal transition (EMT), and cardiovascular differentiation. However, Mesp1 deletion in mice does not impair initial CP specification and early cardiac differentiation but induces cardiac malformations thought to arise from a defect of CP migration. Using inducible gain-of-function experiments during embryonic stem cell differentiation, we found that Mesp2, its closest homolog, was as efficient as Mesp1 at promoting CP specification, EMT, and cardiovascular differentiation. However, only Mesp1 stimulated polarity and directional cell migration through a cell-autonomous mechanism. Transcriptional analysis and chromatin immunoprecipitation experiments revealed that Mesp1 and Mesp2 activate common target genes that promote CP specification and differentiation. We identified two direct Mesp1 target genes, Prickle1 and RasGRP3, that are strongly induced by Mesp1 and not by Mesp2 and that control the polarity and the speed of cell migration. Altogether, our results identify the molecular interface controlled by Mesp1 that links CP specification and cell migration.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular/fisiología , Corazón/crecimiento & desarrollo , Células Madre/metabolismo , Células Madre/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Inmunoprecipitación de Cromatina/métodos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Transición Epitelial-Mesenquimal/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Mesodermo/metabolismo , Mesodermo/fisiología , Ratones , Miocardio/metabolismo
6.
Nat Cell Biol ; 16(9): 829-40, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25150979

RESUMEN

Cardiac development arises from two sources of mesoderm progenitors, the first heart field (FHF) and the second (SHF). Mesp1 has been proposed to mark the most primitive multipotent cardiac progenitors common for both heart fields. Here, using clonal analysis of the earliest prospective cardiovascular progenitors in a temporally controlled manner during early gastrulation, we found that Mesp1 progenitors consist of two temporally distinct pools of progenitors restricted to either the FHF or the SHF. FHF progenitors were unipotent, whereas SHF progenitors were either unipotent or bipotent. Microarray and single-cell PCR with reverse transcription analysis of Mesp1 progenitors revealed the existence of molecularly distinct populations of Mesp1 progenitors, consistent with their lineage and regional contribution. Together, these results provide evidence that heart development arises from distinct populations of unipotent and bipotent cardiac progenitors that independently express Mesp1 at different time points during their specification, revealing that the regional segregation and lineage restriction of cardiac progenitors occur very early during gastrulation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Atrios Cardíacos/embriología , Ventrículos Cardíacos/embriología , Células Madre/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Tipificación del Cuerpo , Diferenciación Celular , Linaje de la Célula , Femenino , Corazón Fetal/citología , Corazón Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Ratones , Ratones Transgénicos , Especificidad de Órganos , Transcriptoma
7.
J Cell Biol ; 192(5): 751-65, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21383076

RESUMEN

During embryonic development and embryonic stem cell (ESC) differentiation, the different cell lineages of the mature heart arise from two types of multipotent cardiovascular progenitors (MCPs), the first and second heart fields. A key question is whether these two MCP populations arise from differentiation of a common progenitor. In this paper, we engineered Mesp1-green fluorescent protein (GFP) ESCs to isolate early MCPs during ESC differentiation. Mesp1-GFP cells are strongly enriched for MCPs, presenting the ability to differentiate into multiple cardiovascular lineages from both heart fields in vitro and in vivo. Transcriptional profiling of Mesp1-GFP cells uncovered cell surface markers expressed by MCPs allowing their prospective isolation. Mesp1 is required for MCP specification and the expression of key cardiovascular transcription factors. Isl1 is expressed in a subset of early Mesp1-expressing cells independently of Mesp1 and acts together with Mesp1 to promote cardiovascular differentiation. Our study identifies the early MCPs residing at the top of the cellular hierarchy of cardiovascular lineages during ESC differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Miocardio/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/análisis , Corazón/embriología , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM , Ratones , Proteínas Recombinantes de Fusión/análisis , Factores de Transcripción
8.
Cell Stem Cell ; 3(1): 69-84, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18593560

RESUMEN

During embryonic development, multipotent cardiovascular progenitor cells are specified from early mesoderm. Using mouse ESCs in which gene expression can be temporally regulated, we have found that transient expression of Mesp1 dramatically accelerates and enhances multipotent cardiovascular progenitor specification through an intrinsic and cell autonomous mechanism. Genome-wide transcriptional analysis indicates that Mesp1 rapidly activates and represses a discrete set of genes, and chromatin immunoprecipitation shows that Mesp1 directly binds to regulatory DNA sequences located in the promoter of many key genes in the core cardiac transcriptional machinery, resulting in their rapid upregulation. Mesp1 also directly represses the expression of key genes regulating other early mesoderm and endoderm cell fates. Our results demonstrate that Mesp1 acts as a key regulatory switch during cardiovascular specification, residing at the top of the hierarchy of the gene network responsible for cardiovascular cell-fate determination.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Sistema Cardiovascular/citología , Diferenciación Celular/fisiología , Células Madre Pluripotentes/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/fisiología , Secuencias Hélice-Asa-Hélice/fisiología , Homeostasis , Ratones , Miocardio/citología , Células Madre Pluripotentes/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA