Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(9): 1393-1411.e7, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37030288

RESUMEN

Polycomb repressive complex 2 (PRC2) mediates H3K27me3 deposition, which is thought to recruit canonical PRC1 (cPRC1) via chromodomain-containing CBX proteins to promote stable repression of developmental genes. PRC2 forms two major subcomplexes, PRC2.1 and PRC2.2, but their specific roles remain unclear. Through genetic knockout (KO) and replacement of PRC2 subcomplex-specific subunits in naïve and primed pluripotent cells, we uncover distinct roles for PRC2.1 and PRC2.2 in mediating the recruitment of different forms of cPRC1. PRC2.1 catalyzes the majority of H3K27me3 at Polycomb target genes and is sufficient to promote recruitment of CBX2/4-cPRC1 but not CBX7-cPRC1. Conversely, while PRC2.2 is poor at catalyzing H3K27me3, we find that its accessory protein JARID2 is essential for recruitment of CBX7-cPRC1 and the consequent 3D chromatin interactions at Polycomb target genes. We therefore define distinct contributions of PRC2.1- and PRC2.2-specific accessory proteins to Polycomb-mediated repression and uncover a new mechanism for cPRC1 recruitment.


Asunto(s)
Histonas , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Histonas/genética , Histonas/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Cromatina/genética
2.
Mol Cell ; 81(17): 3526-3541.e8, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34186021

RESUMEN

BAP1 is mutated or deleted in many cancer types, including mesothelioma, uveal melanoma, and cholangiocarcinoma. It is the catalytic subunit of the PR-DUB complex, which removes PRC1-mediated H2AK119ub1, essential for maintaining transcriptional repression. However, the precise relationship between BAP1 and Polycombs remains elusive. Using embryonic stem cells, we show that BAP1 restricts H2AK119ub1 deposition to Polycomb target sites. This increases the stability of Polycomb with their targets and prevents diffuse accumulation of H2AK119ub1 and H3K27me3. Loss of BAP1 results in a broad increase in H2AK119ub1 levels that is primarily dependent on PCGF3/5-PRC1 complexes. This titrates PRC2 away from its targets and stimulates H3K27me3 accumulation across the genome, leading to a general chromatin compaction. This provides evidence for a unifying model that resolves the apparent contradiction between BAP1 catalytic activity and its role in vivo, uncovering molecular vulnerabilities that could be useful for BAP1-related pathologies.


Asunto(s)
Cromatina/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular/metabolismo , Cromatina/genética , Cromatina/fisiología , Células Madre Embrionarias/metabolismo , Heterocromatina , Histonas/metabolismo , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/fisiología , Ubiquitinación
3.
Mol Cell ; 77(4): 840-856.e5, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31883952

RESUMEN

Polycomb group proteins (PcGs) maintain transcriptional repression to preserve cellular identity in two distinct repressive complexes, PRC1 and PRC2, that modify histones by depositing H2AK119ub1 and H3K27me3, respectively. PRC1 and PRC2 exist in different variants and show a complex regulatory cross-talk. However, the contribution that H2AK119ub1 plays in mediating PcG repressive functions remains largely controversial. Using a fully catalytic inactive RING1B mutant, we demonstrated that H2AK119ub1 deposition is essential to maintain PcG-target gene repression in embryonic stem cells (ESCs). Loss of H2AK119ub1 induced a rapid displacement of PRC2 activity and a loss of H3K27me3 deposition. This preferentially affected PRC2.2 variant with respect to PRC2.1, destabilizing canonical PRC1 activity. Finally, we found that variant PRC1 forms can sense H2AK119ub1 deposition, which contributes to their stabilization specifically at sites where this modification is highly enriched. Overall, our data place H2AK119ub1 deposition as a central hub that mounts PcG repressive machineries to preserve cell transcriptional identity.


Asunto(s)
Regulación de la Expresión Génica , Histonas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Transcripción Genética , Ubiquitinación , Línea Celular , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Mutación Missense , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Mol Cell ; 74(5): 1037-1052.e7, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31029542

RESUMEN

Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) control cell identity by establishing facultative heterochromatin repressive domains at common sets of target genes. PRC1, which deposits H2Aub1 through the E3 ligases RING1A/B, forms six biochemically distinct subcomplexes depending on the assembled PCGF protein (PCGF1-PCGF6); however, it is yet unclear whether these subcomplexes have also specific activities. Here we show that PCGF1 and PCGF2 largely compensate for each other, while other PCGF proteins have high levels of specificity for distinct target genes. PCGF2 associates with transcription repression, whereas PCGF3 and PCGF6 associate with actively transcribed genes. Notably, PCGF3 and PCGF6 complexes can assemble and be recruited to several active sites independently of RING1A/B activity (therefore, of PRC1). For chromatin recruitment, the PCGF6 complex requires the combinatorial activities of its MGA-MAX and E2F6-DP1 subunits, while PCGF3 requires an interaction with the USF1 DNA binding transcription factor.


Asunto(s)
Complejo Represivo Polycomb 1/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cromatina/genética , Proteínas de Unión al ADN/genética , Factor de Transcripción E2F6/genética , Heterocromatina/genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Proteínas Represoras/genética , Factor de Transcripción DP1/genética , Factores de Transcripción/genética , Factores Estimuladores hacia 5'/genética
5.
Mol Cell ; 70(2): 371-379.e5, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29606589

RESUMEN

The Polycomb repressor complex 2 (PRC2) is composed of the core subunits Ezh1/2, Suz12, and Eed, and it mediates all di- and tri-methylation of histone H3 at lysine 27 in higher eukaryotes. However, little is known about how the catalytic activity of PRC2 is regulated to demarcate H3K27me2 and H3K27me3 domains across the genome. To address this, we mapped the endogenous interactomes of Ezh2 and Suz12 in embryonic stem cells (ESCs), and we combined this with a functional screen for H3K27 methylation marks. We found that Nsd1-mediated H3K36me2 co-locates with H3K27me2, and its loss leads to genome-wide expansion of H3K27me3. These increases in H3K27me3 occurred at PRC2/PRC1 target genes and as de novo accumulation within what were previously broad H3K27me2 domains. Our data support a model in which Nsd1 is a key modulator of PRC2 function required for regulating the demarcation of genome-wide H3K27me2 and H3K27me3 domains in ESCs.


Asunto(s)
Proteínas Portadoras/metabolismo , Ensamble y Desensamble de Cromatina , Histonas/metabolismo , Células Madre Embrionarias de Ratones/enzimología , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Proteínas Portadoras/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Metilación , Ratones , Proteínas Nucleares/genética , Complejo Represivo Polycomb 2/genética , Procesamiento Proteico-Postraduccional
7.
Trends Genet ; 38(4): 333-352, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34426021

RESUMEN

Cell identity is tightly controlled by specific transcriptional programs which require post-translational modifications of histones. These histone modifications allow the establishment and maintenance of active and repressed chromatin domains. Histone H2A lysine 119 ubiquitination (H2AK119ub1) has an essential role in building repressive chromatin domains during development. It is regulated by the counteracting activities of the Polycomb repressive complex 1 (PRC1) and the Polycomb repressive-deubiquitinase (PR-DUB) complexes, two multi-subunit ensembles that write and erase this modification, respectively. We have catalogued the recurrent genetic alterations in subunits of the PRC1 and PR-DUB complexes in both neurodevelopmental disorders and cancer. These genetic lesions are often shared across disorders, and we highlight common mechanisms of H2AK119ub1 dysregulation and how they affect development in multiple disease contexts.


Asunto(s)
Discapacidades del Desarrollo , Histonas , Neoplasias , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Ubiquitinación , Niño , Cromatina/genética , Discapacidades del Desarrollo/genética , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
8.
Gastroenterology ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38971196

RESUMEN

BACKGROUND AND AIMS: WNT signaling is central to spatial tissue arrangement, regulating stem cell activity, and represents the hallmark of gastrointestinal cancers. While its role in driving intestinal tumors is well characterized, WNT's role in gastric tumorigenesis remains elusive. METHODS: We have developed mouse models to control the specific expression of an oncogenic form of B-CATENIN in combination with MYC activation in Lgr5+ cells of the gastric antrum. We used multi-omics approaches applied in vivo and in organoid models to characterize their cooperation in driving gastric tumorigenesis. RESULTS: We report that constitutive B-CATENIN stabilization in the stomach has negligible oncogenic effects and requires MYC activation to induce gastric tumour formation. While physiologically low MYC levels in gastric glands limit B-CATENIN transcriptional activity, increased MYC expression unleashes the WNT oncogenic transcriptional program, promoting B-CATENIN enhancer invasion without a direct transcriptional cooperation. MYC activation induces a metabolic rewiring that suppresses lysosomal biogenesis through mTOR and ERK activation and MiT/TFE inhibition. This prevents EPCAM degradation by macropinocytosis, promoting B-CATENIN chromatin accumulation and activation of WNT oncogenic transcription. CONCLUSION: Our results uncovered a new signaling framework with important implications for the control of gastric epithelial architecture and WNT-dependent oncogenic transformation.

9.
Mol Cell ; 64(4): 637-638, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863221

RESUMEN

Two studies published in this issue of Molecular Cell (Beringer et al., 2016; Liefke et al., 2016) characterize the novel interaction of EPOP with Elongin BC in regulating gene transcription at both H3K4me3-broad active and H3K27me3 Polycomb-repressed chromatin domains.

11.
Nucleic Acids Res ; 49(2): 791-804, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33398338

RESUMEN

The proteolytic cleavage of histone tails, also termed histone clipping, has been described as a mechanism for permanent removal of post-translational modifications (PTMs) from histone proteins. Such activity has been ascribed to ensure regulatory function in key cellular processes such as differentiation, senescence and transcriptional control, for which different histone-specific proteases have been described. However, all these studies were exclusively performed using cell lines cultured in vitro and no clear evidence that histone clipping is regulated in vivo has been reported. Here we show that histone H3 N-terminal tails undergo extensive cleavage in the differentiated cells of the villi in mouse intestinal epithelium. Combining biochemical methods, 3D organoid cultures and in vivo approaches, we demonstrate that intestinal H3 clipping is the result of multiple proteolytic activities. We identified Trypsins and Cathepsin L as specific H3 tail proteases active in small intestinal differentiated cells and showed that their proteolytic activity is differentially affected by the PTM pattern of histone H3 tails. Together, our findings provide in vivo evidence of H3 tail proteolysis in mammalian tissues, directly linking H3 clipping to cell differentiation.


Asunto(s)
Enterocitos/metabolismo , Histonas/metabolismo , Intestino Delgado/citología , Células de Paneth/metabolismo , Péptido Hidrolasas/metabolismo , Procesamiento Proteico-Postraduccional , Células Madre/metabolismo , Animales , Catepsina L/metabolismo , Diferenciación Celular , Homeostasis , Mucosa Intestinal/citología , Ratones , Microvellosidades/ultraestructura , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Organoides , Dominios Proteicos , Tripsina/metabolismo
12.
Mol Cell ; 53(1): 49-62, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24289921

RESUMEN

H3K27me3 is deposited at promoters by the preferential association of Polycomb repressive complex 2 (PRC2) with CpG-rich DNA elements regulating development by repressing gene transcription. H3K27 is also present in mono- and dimethylated states; however, the functional roles of H3K27me1 and H3K27me2 deposition remain poorly characterized. Here, we show that PRC2 activity is not only associated with H3K27me3 but also regulates all forms of H3K27 methylation in a spatially defined manner, contributing to different genomic functions in mouse embryonic stem cells. H3K27me1 accumulates within transcribed genes, promotes transcription, and is regulated by Setd2-dependent H3K36me3 deposition. Contrarily, H3K27me2 is present on approximately 70% of total histone H3 and is distributed in large chromatin domains, exerting protective functions by preventing firing of non-cell-type-specific enhancers. Considering that only 5%-10% of deregulated genes in PRC2-deficient cells are direct H3K27me3 targets, our data support an active role for all H3K27 methylated forms in regulating transcription and determining cell identity.


Asunto(s)
Células Madre Embrionarias/enzimología , Células Madre Embrionarias/inmunología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Transcripción Genética/fisiología , Animales , Línea Celular , Células Madre Embrionarias/citología , Histonas/genética , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Metilación , Ratones , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo
13.
EMBO J ; 36(15): 2216-2232, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28554894

RESUMEN

Sin3a is the central scaffold protein of the prototypical Hdac1/2 chromatin repressor complex, crucially required during early embryonic development for the growth of pluripotent cells of the inner cell mass. Here, we compare the composition of the Sin3a-Hdac complex between pluripotent embryonic stem (ES) and differentiated cells by establishing a method that couples two independent endogenous immunoprecipitations with quantitative mass spectrometry. We define the precise composition of the Sin3a complex in multiple cell types and identify the Fam60a subunit as a key defining feature of a variant Sin3a complex present in ES cells, which also contains Ogt and Tet1. Fam60a binds on H3K4me3-positive promoters in ES cells, together with Ogt, Tet1 and Sin3a, and is essential to maintain the complex on chromatin. Finally, we show that depletion of Fam60a phenocopies the loss of Sin3a, leading to reduced proliferation, an extended G1-phase and the deregulation of lineage genes. Taken together, Fam60a is an essential core subunit of a variant Sin3a complex in ES cells that is required to promote rapid proliferation and prevent unscheduled differentiation.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/fisiología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Animales , Diferenciación Celular , Inmunoprecipitación , Espectrometría de Masas , Ratones , Unión Proteica
14.
Hepatology ; 72(4): 1430-1443, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31965581

RESUMEN

BACKGROUND AND AIMS: Activation of MYC and catenin beta-1 (CTNNB1, encoding ß-catenin) can co-occur in liver cancer, but how these oncogenes cooperate in tumorigenesis remains unclear. APPROACH AND RESULTS: We generated a mouse model allowing conditional activation of MYC and WNT/ß-catenin signaling (through either ß-catenin activation or loss of APC - adenomatous polyposis coli) upon expression of CRE recombinase in the liver and monitored their effects on hepatocyte proliferation, apoptosis, gene expression profiles, and tumorigenesis. Activation of WNT/ß-catenin signaling strongly accelerated MYC-driven carcinogenesis in the liver. Both pathways also cooperated in promoting cellular transformation in vitro, demonstrating their cell-autonomous action. Short-term induction of MYC and ß-catenin in hepatocytes, followed by RNA-sequencing profiling, allowed the identification of a "Myc/ß-catenin signature," composed of a discrete set of Myc-activated genes whose expression increased in the presence of active ß-catenin. Notably, this signature enriched for targets of Yes-associated protein (Yap) and transcriptional coactivator with PDZ-binding motif (Taz), two transcriptional coactivators known to be activated by WNT/ß-catenin signaling and to cooperate with MYC in mitogenic activation and liver transformation. Consistent with these regulatory connections, Yap/Taz accumulated upon Myc/ß-catenin activation and were required not only for the ensuing proliferative response, but also for tumor cell growth and survival. Finally, the Myc/ß-catenin signature was enriched in a subset of human hepatocellular carcinomas characterized by comparatively poor prognosis. CONCLUSIONS: Myc and ß-catenin show a strong cooperative action in liver carcinogenesis, with Yap and Taz serving as mediators of this effect. These findings warrant efforts toward therapeutic targeting of Yap/Taz in aggressive liver tumors marked by elevated Myc/ß-catenin activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Ciclo Celular/fisiología , Neoplasias Hepáticas Experimentales/etiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Transactivadores/fisiología , beta Catenina/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Vía de Señalización Wnt/fisiología , Proteínas Señalizadoras YAP
15.
Mol Cell ; 49(4): 645-56, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23352454

RESUMEN

O-linked N-acetylglucosamine (O-GlcNAc) transferase (Ogt) activity is essential for embryonic stem cell (ESC) viability and mouse development. Ogt is present both in the cytoplasm and the nucleus of different cell types and catalyzes serine and threonine glycosylation. We have characterized the biochemical features of nuclear Ogt and identified the ten-eleven translocation (TET) proteins Tet1 and Tet2 as stable partners of Ogt in the nucleus of ESCs. We show at a genome-wide level that Ogt preferentially associates with Tet1 to genes promoters in close proximity of CpG-rich transcription start sites. These regions are characterized by low levels of DNA modification, suggesting a link between Tet1 and Ogt activities in regulating CpG island methylation. Finally, we show that Tet1 is required for binding of Ogt to chromatin affecting Tet1 activity. Taken together, our data characterize how O-GlcNAcylation is recruited to chromatin and interacts with the activity of 5-methylcytosine hydroxylases.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina , Islas de CpG , Proteínas de Unión al ADN/aislamiento & purificación , Dioxigenasas , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Inmunoprecipitación , Redes y Vías Metabólicas/genética , Ratones , N-Acetilglucosaminiltransferasas/aislamiento & purificación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas/aislamiento & purificación , Transducción de Señal/genética , Sitio de Iniciación de la Transcripción
16.
EMBO J ; 35(21): 2301-2314, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27585866

RESUMEN

Chromatin modifications shape cell heterogeneity by activating and repressing defined sets of genes involved in cell proliferation, differentiation and development. Polycomb-repressive complexes (PRCs) act synergistically during development and differentiation by maintaining transcriptional repression of common genes. PRC2 exerts this activity by catalysing H3K27 trimethylation. Here, we show that in the intestinal epithelium PRC2 is required to sustain progenitor cell proliferation and the correct balance between secretory and absorptive lineage differentiation programs. Using genetic models, we show that PRC2 activity is largely dispensable for intestinal stem cell maintenance but is strictly required for radiation-induced regeneration by preventing Cdkn2a transcription. Combining these models with genomewide molecular analysis, we further demonstrate that preferential accumulation of secretory cells does not result from impaired proliferation of progenitor cells induced by Cdkn2a activation but rather from direct regulation of transcription factors responsible for secretory lineage commitment. Overall, our data uncover a dual role of PRC2 in intestinal homeostasis highlighting the importance of this repressive layer in controlling cell plasticity and lineage choices in adult tissues.


Asunto(s)
Mucosa Intestinal/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Ratones Transgénicos
17.
Circ Res ; 122(2): 231-245, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29233846

RESUMEN

RATIONALE: The mechanistic foundation of vascular maturation is still largely unknown. Several human pathologies are characterized by deregulated angiogenesis and unstable blood vessels. Solid tumors, for instance, get their nourishment from newly formed structurally abnormal vessels which present wide and irregular interendothelial junctions. Expression and clustering of the main endothelial-specific adherens junction protein, VEC (vascular endothelial cadherin), upregulate genes with key roles in endothelial differentiation and stability. OBJECTIVE: We aim at understanding the molecular mechanisms through which VEC triggers the expression of a set of genes involved in endothelial differentiation and vascular stabilization. METHODS AND RESULTS: We compared a VEC-null cell line with the same line reconstituted with VEC wild-type cDNA. VEC expression and clustering upregulated endothelial-specific genes with key roles in vascular stabilization including claudin-5, vascular endothelial-protein tyrosine phosphatase (VE-PTP), and von Willebrand factor (vWf). Mechanistically, VEC exerts this effect by inhibiting polycomb protein activity on the specific gene promoters. This is achieved by preventing nuclear translocation of FoxO1 (Forkhead box protein O1) and ß-catenin, which contribute to PRC2 (polycomb repressive complex-2) binding to promoter regions of claudin-5, VE-PTP, and vWf. VEC/ß-catenin complex also sequesters a core subunit of PRC2 (Ezh2 [enhancer of zeste homolog 2]) at the cell membrane, preventing its nuclear translocation. Inhibition of Ezh2/VEC association increases Ezh2 recruitment to claudin-5, VE-PTP, and vWf promoters, causing gene downregulation. RNA sequencing comparison of VEC-null and VEC-positive cells suggested a more general role of VEC in activating endothelial genes and triggering a vascular stability-related gene expression program. In pathological angiogenesis of human ovarian carcinomas, reduced VEC expression paralleled decreased levels of claudin-5 and VE-PTP. CONCLUSIONS: These data extend the knowledge of polycomb-mediated regulation of gene expression to endothelial cell differentiation and vessel maturation. The identified mechanism opens novel therapeutic opportunities to modulate endothelial gene expression and induce vascular normalization through pharmacological inhibition of the polycomb-mediated repression system.


Asunto(s)
Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Endotelio Vascular/metabolismo , Epigénesis Genética/fisiología , Animales , Antígenos CD/genética , Cadherinas/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Endotelio Vascular/ultraestructura , Expresión Génica , Células HEK293 , Humanos , Ratones , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica/fisiología
19.
Genes Dev ; 24(3): 265-76, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20123906

RESUMEN

Polycomb complexes establish chromatin modifications for maintaining gene repression and are essential for embryonic development in mice. Here we use pluripotent embryonic stem (ES) cells to demonstrate an unexpected redundancy between Polycomb-repressive complex 1 (PRC1) and PRC2 during the formation of differentiated cells. ES cells lacking the function of either PRC1 or PRC2 can differentiate into cells of the three germ layers, whereas simultaneous loss of PRC1 and PRC2 abrogates differentiation. On the molecular level, the differentiation defect is caused by the derepression of a set of genes that is redundantly repressed by PRC1 and PRC2 in ES cells. Furthermore, we find that genomic repeats are Polycomb targets and show that, in the absence of Polycomb complexes, endogenous murine leukemia virus elements can mobilize. This indicates a contribution of the Polycomb group system to the defense against parasitic DNA, and a potential role of genomic repeats in Polycomb-mediated gene regulation.


Asunto(s)
Proteínas Represoras/genética , Animales , Diferenciación Celular , Línea Celular , Embrión de Mamíferos/metabolismo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Ratones , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas del Grupo Polycomb , Proteínas Represoras/metabolismo , Secuencias Repetidas Terminales
20.
Nature ; 464(7286): 306-10, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20075857

RESUMEN

The Polycomb group (PcG) proteins have an important role in controlling the expression of genes essential for development, differentiation and maintenance of cell fates. The Polycomb repressive complex 2 (PRC2) is believed to regulate transcriptional repression by catalysing the di- and tri-methylation of lysine 27 on histone H3 (H3K27me2/3). At present, it is unknown how the PcG proteins are recruited to their target promoters in mammalian cells. Here we show that PRC2 forms a stable complex with the Jumonji- and ARID-domain-containing protein, JARID2 (ref. 4). Using genome-wide location analysis, we show that JARID2 binds to more than 90% of previously mapped PcG target genes. Notably, we show that JARID2 is sufficient to recruit PcG proteins to a heterologous promoter, and that inhibition of JARID2 expression leads to a major loss of PcG binding and to a reduction of H3K27me3 levels on target genes. Consistent with an essential role for PcG proteins in early development, we demonstrate that JARID2 is required for the differentiation of mouse embryonic stem cells. Thus, these results demonstrate that JARID2 is essential for the binding of PcG proteins to target genes and, consistent with this, for the proper differentiation of embryonic stem cells and normal development.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Células HeLa , Humanos , Ratones , Proteínas del Tejido Nervioso/genética , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA