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










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 180, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36635295

RESUMEN

The potential of pluripotent cells to respond to developmental cues and trigger cell differentiation is enhanced during the G1 phase of the cell cycle, but the molecular mechanisms involved are poorly understood. Variations in polycomb activity during interphase progression have been hypothesized to regulate the cell-cycle-phase-dependent transcriptional activation of differentiation genes during lineage transition in pluripotent cells. Here, we show that recruitment of Polycomb Repressive Complex 1 (PRC1) and associated molecular functions, ubiquitination of H2AK119 and three-dimensional chromatin interactions, are enhanced during S and G2 phases compared to the G1 phase. In agreement with the accumulation of PRC1 at target promoters upon G1 phase exit, cells in S and G2 phases show firmer transcriptional repression of developmental regulator genes that is drastically perturbed upon genetic ablation of the PRC1 catalytic subunit RING1B. Importantly, depletion of RING1B during retinoic acid stimulation interferes with the preference of mouse embryonic stem cells (mESCs) to induce the transcriptional activation of differentiation genes in G1 phase. We propose that incremental enrolment of polycomb repressive activity during interphase progression reduces the tendency of cells to respond to developmental cues during S and G2 phases, facilitating activation of cell differentiation in the G1 phase of the pluripotent cell cycle.


Asunto(s)
Histonas , Células Madre Pluripotentes , Complejo Represivo Polycomb 1 , Animales , Ratones , Diferenciación Celular/genética , Cromatina/genética , Histonas/metabolismo , Interfase , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Células Madre Pluripotentes/citología
2.
Oncogene ; 41(28): 3611-3624, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35680984

RESUMEN

Reversible transition between the epithelial and mesenchymal states are key aspects of carcinoma cell dissemination and the metastatic disease, and thus, characterizing the molecular basis of the epithelial to mesenchymal transition (EMT) is crucial to find druggable targets and more effective therapeutic approaches in cancer. Emerging studies suggest that epigenetic regulators might endorse cancer cells with the cell plasticity required to conduct dynamic changes in cell state during EMT. However, epigenetic mechanisms involved remain mostly unknown. Polycomb Repressive Complexes (PRCs) proteins are well-established epigenetic regulators of development and stem cell differentiation, but their role in different cancer systems is inconsistent and sometimes paradoxical. In this study, we have analysed the role of the PRC2 protein EZH2 in lung carcinoma cells. We found that besides its described role in CDKN2A-dependent cell proliferation, EZH2 upholds the epithelial state of cancer cells by repressing the transcription of hundreds of mesenchymal genes. Chemical inhibition or genetic removal of EZH2 promotes the residence of cancer cells in the mesenchymal state during reversible epithelial-mesenchymal transition. In fitting, analysis of human patient samples and tumour xenograft models indicate that EZH2 is required to efficiently repress mesenchymal genes and facilitate tumour colonization in vivo. Overall, this study discloses a novel role of PRC2 as a master regulator of EMT in carcinoma cells. This finding has important implications for the design of therapies based on EZH2 inhibitors in human cancer patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Proteína Potenciadora del Homólogo Zeste 2 , Neoplasias Pulmonares , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Diferenciación Celular , Línea Celular Tumoral , Plasticidad de la Célula/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Transición Epitelial-Mesenquimal/genética , Humanos , Neoplasias Pulmonares/genética , Proteínas del Grupo Polycomb
3.
Nat Genet ; 53(7): 1036-1049, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34183853

RESUMEN

CpG islands (CGIs) represent a widespread feature of vertebrate genomes, being associated with ~70% of all gene promoters. CGIs control transcription initiation by conferring nearby promoters with unique chromatin properties. In addition, there are thousands of distal or orphan CGIs (oCGIs) whose functional relevance is barely known. Here we show that oCGIs are an essential component of poised enhancers that augment their long-range regulatory activity and control the responsiveness of their target genes. Using a knock-in strategy in mouse embryonic stem cells, we introduced poised enhancers with or without oCGIs within topologically associating domains harboring genes with different types of promoters. Analysis of the resulting cell lines revealed that oCGIs act as tethering elements that promote the physical and functional communication between poised enhancers and distally located genes, particularly those with large CGI clusters in their promoters. Therefore, by acting as genetic determinants of gene-enhancer compatibility, CGIs can contribute to gene expression control under both physiological and potentially pathological conditions.


Asunto(s)
Islas de CpG , Metilación de ADN , Elementos de Facilitación Genéticos , Epigénesis Genética , Regulación de la Expresión Génica , Animales , Cromatina/genética , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Técnicas de Sustitución del Gen , Ratones , Regiones Promotoras Genéticas
4.
Life Sci Alliance ; 3(5)2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284355

RESUMEN

Mammals optimize their physiology to the light-dark cycle by synchronization of the master circadian clock in the brain with peripheral clocks in the rest of the tissues of the body. Circadian oscillations rely on a negative feedback loop exerted by the molecular clock that is composed by transcriptional activators Bmal1 and Clock, and their negative regulators Period and Cryptochrome. Components of the molecular clock are expressed during early development, but onset of robust circadian oscillations is only detected later during embryogenesis. Here, we have used naïve pluripotent mouse embryonic stem cells (mESCs) to study the role of Bmal1 during early development. We found that, compared to wild-type cells, Bmal1-/- mESCs express higher levels of Nanog protein and altered expression of pluripotency-associated signalling pathways. Importantly, Bmal1-/- mESCs display deficient multi-lineage cell differentiation capacity during the formation of teratomas and gastrula-like organoids. Overall, we reveal that Bmal1 regulates pluripotent cell differentiation and propose that the molecular clock is an hitherto unrecognized regulator of mammalian development.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Diferenciación Celular/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción ARNTL/fisiología , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Retroalimentación Fisiológica/fisiología , Expresión Génica/genética , Células Madre Pluripotentes Inducidas/citología , Ratones , Células Madre Embrionarias de Ratones/citología , Proteínas Circadianas Period/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Transcripción Genética
5.
Sci Adv ; 6(10): eaay4768, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32181346

RESUMEN

When self-renewing pluripotent cells receive a differentiation signal, ongoing cell duplication needs to be coordinated with entry into a differentiation program. Accordingly, transcriptional activation of lineage specifier genes and cell differentiation is confined to the G1 phase of the cell cycle by unknown mechanisms. We found that Polycomb repressive complex 2 (PRC2) subunits are differentially recruited to lineage specifier gene promoters across cell cycle in mouse embryonic stem cells (mESCs). Jarid2 and the catalytic subunit Ezh2 are markedly accumulated at target promoters during S and G2 phases, while the transcriptionally activating subunits EPOP and EloB are enriched during G1 phase. Fluctuations in the recruitment of PRC2 subunits promote changes in RNA synthesis and RNA polymerase II binding that are compromised in Jarid2 -/- mESCs. Overall, we show that differential recruitment of PRC2 subunits across cell cycle enables the establishment of a chromatin state that facilitates the induction of cell differentiation in G1 phase.


Asunto(s)
Ciclo Celular/genética , Cromatina/química , Proteína Potenciadora del Homólogo Zeste 2/genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 2/genética , Animales , Diferenciación Celular , Línea Celular Transformada , Cromatina/metabolismo , Elonguina/genética , Elonguina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Complejo Represivo Polycomb 2/deficiencia , Regiones Promotoras Genéticas , Unión Proteica , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transducción de Señal , Transcripción Genética
6.
Biomolecules ; 11(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396280

RESUMEN

Gliomas in general and the more advanced glioblastomas (GBM) in particular are the most usual tumors of the central nervous system with poor prognosis. GBM patients develop resistance to distinct therapies, in part due to the existence of tumor cell subpopulations with stem-like properties that participate in trans-differentiation events. Within the complex tumor microenvironment, the involvement of extracellular proteases remains poorly understood. The extracellular protease ADAMTS1 has already been reported to contribute to the plasticity of cancer cells. Accordingly, this basic knowledge and the current availability of massive sequencing data from human gliomas, reinforced the development of this work. We first performed an in silico study of ADAMTS1 and endothelial markers in human gliomas, providing the basis to further assess these molecules in several primary glioblastoma-initiating cells and established GBM cells with the ability to acquire an endothelial-like phenotype. Using a co-culture approach of endothelial and GBM cells, we noticed a relevant function of ADAMTS1 in GBM cells leading the organization of endothelial-like networks and, even more significantly, we found a blockade of the formation of tumor-spheres and a deficient response to hypoxia in the absence of ADAMTS1. Our data support a chief role of this protease modulating the phenotypic plasticity of GBM.


Asunto(s)
Proteína ADAMTS1/genética , Plasticidad de la Célula/genética , Glioblastoma/genética , Glioma/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Glioma/patología , Humanos , Masculino , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Microambiente Tumoral/genética
7.
Sci Rep ; 9(1): 8140, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31148571

RESUMEN

Recent technical advances highlight that to understand mammalian development and human disease we need to consider transcriptional and epigenetic cell-to-cell differences within cell populations. This is particularly important in key areas of biomedicine like stem cell differentiation and intratumor heterogeneity. The recently developed nucleosome occupancy and methylome (NOMe) assay facilitates the simultaneous study of DNA methylation and nucleosome positioning on the same DNA strand. NOMe-treated DNA can be sequenced by sanger (NOMe-PCR) or high throughput approaches (NOMe-seq). NOMe-PCR provides information for a single locus at the single molecule while NOMe-seq delivers genome-wide data that is usually interrogated to obtain population-averaged measures. Here, we have developed a bioinformatic tool that allow us to easily obtain locus-specific information at the single molecule using genome-wide NOMe-seq datasets obtained from bulk populations. We have used NOMePlot to study mouse embryonic stem cells and found that polycomb-repressed bivalent gene promoters coexist in two different epigenetic states, as defined by the nucleosome binding pattern detected around their transcriptional start site.


Asunto(s)
Biología Computacional/métodos , Metilación de ADN , Nucleosomas/genética , Reconocimiento de Normas Patrones Automatizadas , Animales , Islas de CpG , Células Madre Embrionarias/citología , Epigénesis Genética , Genoma Humano , Humanos , Internet , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Programas Informáticos , Sitio de Iniciación de la Transcripción
8.
Cell Rep ; 12(4): 573-86, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26190104

RESUMEN

Jarid2 is part of the Polycomb Repressor complex 2 (PRC2) responsible for genome-wide H3K27me3 deposition. Unlike other PRC2-deficient embryonic stem cells (ESCs), however, Jarid2-deficient ESCs show a severe differentiation block, altered colony morphology, and distinctive patterns of deregulated gene expression. Here, we show that Jarid2(-/-) ESCs express constitutively high levels of Nanog but reduced PCP signaling components Wnt9a, Prickle1, and Fzd2 and lowered ß-catenin activity. Depletion of Wnt9a/Prickle1/Fzd2 from wild-type ESCs or overexpression of Nanog largely phenocopies these cellular defects. Co-culture of Jarid2(-/-) with wild-type ESCs restores variable Nanog expression and ß-catenin activity and can partially rescue the differentiation block of mutant cells. In addition, we show that ESCs lacking Jarid2 or Wnt9a/Prickle1/Fzd2 or overexpressing Nanog induce multiple ICM formation when injected into normal E3.5 blastocysts. These data describe a previously unrecognized role for Jarid2 in regulating a core pluripotency and Wnt/PCP signaling circuit that is important for ESC differentiation and for pre-implantation development.


Asunto(s)
Blastocisto/metabolismo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Proteína Homeótica Nanog , Complejo Represivo Polycomb 2/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...