Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3452, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658543

RESUMEN

Mutations in chromatin regulators are widespread in cancer. Among them, the histone H3 lysine 27 methyltransferase Polycomb Repressive Complex 2 (PRC2) shows distinct alterations according to tumor type. This specificity is poorly understood. Here, we model several PRC2 alterations in one isogenic system to reveal their comparative effects. Focusing then on lymphoma-associated EZH2 mutations, we show that Ezh2Y641F induces aberrant H3K27 methylation patterns even without wild-type Ezh2, which are alleviated by partial PRC2 inhibition. Remarkably, Ezh2Y641F rewires the response to PRC2 inhibition, leading to induction of antigen presentation genes. Using a unique longitudinal follicular lymphoma cohort, we further link EZH2 status to abnormal H3K27 methylation. We also uncover unexpected variability in the mutational landscape of successive biopsies, pointing to frequent co-existence of different clones and cautioning against stratifying patients based on single sampling. Our results clarify how oncogenic PRC2 mutations disrupt chromatin and transcription, and the therapeutic vulnerabilities this creates.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Histonas , Linfoma Folicular , Mutación , Complejo Represivo Polycomb 2 , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Humanos , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Histonas/metabolismo , Histonas/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Metilación , Cromatina/metabolismo , Cromatina/genética , Transcripción Genética
2.
Nat Commun ; 15(1): 3016, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589367

RESUMEN

Myelodysplastic syndromes (MDS) with mutated SF3B1 gene present features including a favourable outcome distinct from MDS with mutations in other splicing factor genes SRSF2 or U2AF1. Molecular bases of these divergences are poorly understood. Here we find that SF3B1-mutated MDS show reduced R-loop formation predominating in gene bodies associated with intron retention reduction, not found in U2AF1- or SRSF2-mutated MDS. Compared to erythroblasts from SRSF2- or U2AF1-mutated patients, SF3B1-mutated erythroblasts exhibit augmented DNA synthesis, accelerated replication forks, and single-stranded DNA exposure upon differentiation. Importantly, histone deacetylase inhibition using vorinostat restores R-loop formation, slows down DNA replication forks and improves SF3B1-mutated erythroblast differentiation. In conclusion, loss of R-loops with associated DNA replication stress represents a hallmark of SF3B1-mutated MDS ineffective erythropoiesis, which could be used as a therapeutic target.


Asunto(s)
Síndromes Mielodisplásicos , Estructuras R-Loop , Humanos , Factor de Empalme U2AF/genética , Factores de Empalme Serina-Arginina/genética , Factores de Empalme de ARN/genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Mutación , Factores de Transcripción/genética , Fosfoproteínas/genética
3.
Br J Dermatol ; 190(2): 226-243, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37831592

RESUMEN

BACKGROUND: Neurofibromatosis type 1 (NF1) is characterized by the highly variable and unpredictable development of benign peripheral nerve sheath tumours: cutaneous (cNFs), subcutaneous (scNFs) and plexiform (pNFs) neurofibromas. OBJECTIVES: To identify neurofibroma modifier genes, in order to develop a database of patients with NF1. METHODS: All patients were phenotypically evaluated by a medical practitioner using a standardized questionnaire and the causal NF1 variant identified. We enrolled 1333 patients with NF1 who were genotyped for > 7 million common variants. RESULTS: A genome-wide association case-only study identified a significant association with 9q21.33 in the pNF phenotype in the discovery cohort. Twelve, three and four regions suggestive of association at the P ≤ 1 × 10-6 threshold were identified for pNFs, cNFs and scNFs, respectively. Evidence of replication was observed for 4, 2 and 6 loci, including 168 candidate modifier protein-coding genes. Among the candidate modifier genes, some were implicated in the RAS-mitogen-activated protein kinase pathway, cell-cycle control and myelination. Using an original CRISPR/Cas9-based functional assay, we confirmed GAS1 and SPRED2 as pNF and scNF candidate modifiers, as their inactivation specifically affected NF1-mutant Schwann cell growth. CONCLUSIONS: Our study may shed new light on the pathogenesis of NF1-associated neurofibromas and will, hopefully, contribute to the development of personalized care for patients with this deleterious and life-threatening condition.


Asunto(s)
Neurofibroma Plexiforme , Neurofibroma , Neurofibromatosis 1 , Humanos , Neurofibromatosis 1/genética , Neurofibroma Plexiforme/complicaciones , Neurofibroma Plexiforme/genética , Estudio de Asociación del Genoma Completo , Neurofibroma/complicaciones , Neurofibroma/genética , Genotipo , Proteínas Represoras/genética
4.
Cell Rep ; 42(9): 113132, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37708024

RESUMEN

Uveal melanoma (UM) is a rare cancer resulting from the transformation of melanocytes in the uveal tract. Integrative analysis has identified four molecular and clinical subsets of UM. To improve our molecular understanding of UM, we performed extensive multi-omics characterization comparing two aggressive UM patient-derived xenograft models with normal choroidal melanocytes, including DNA optical mapping, specific histone modifications, and DNA topology analysis using Hi-C. Our gene expression and cytogenetic analyses suggest that genomic instability is a hallmark of UM. We also identified a recurrent deletion in the BAP1 promoter resulting in loss of expression and associated with high risk of metastases in UM patients. Hi-C revealed chromatin topology changes associated with the upregulation of PRAME, an independent prognostic biomarker in UM, and a potential therapeutic target. Our findings illustrate how multi-omics approaches can improve our understanding of tumorigenesis and reveal two distinct mechanisms of gene expression dysregulation in UM.


Asunto(s)
Melanoma , Multiómica , Humanos , Melanoma/patología , Melanocitos/metabolismo , ADN , Antígenos de Neoplasias/genética
5.
Life Sci Alliance ; 6(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37684044

RESUMEN

Cell type-specific barcoding of genomes requires the establishment of hundreds of heterochromatin domains where heterochromatin-associated repressive complexes hinder chromatin accessibility thereby silencing genes. At heterochromatin-euchromatin borders, regulation of accessibility not only depends on the delimitation of heterochromatin but may also involve interplays with nearby genes and their transcriptional activity, or alternatively on histone modifiers, chromatin barrier insulators, and more global demarcation of chromosomes into 3D compartmentalized domains and topological-associating domain (TADs). Here, we show that depletion of H3K36 di- or tri-methyl histone methyltransferases dMes-4/NSD or Hypb/dSet2 induces reproducible increasing levels of H3K27me3 at heterochromatin borders including in nearby promoters, thereby repressing hundreds of genes. Furthermore, dMes-4/NSD influences genes demarcated by insulators and TAD borders, within chromatin hubs, unlike transcription-coupled action of Hypb/dSet2 that protects genes independently of TADs. Insulator mutants recapitulate the increase of H3K27me3 upon dMes-4/NSD depletion unlike Hypb/dSet2. Hi-C data demonstrate how dMes-4/NSD blocks propagation of long-range interactions onto active regions. Our data highlight distinct mechanisms protecting genes from H3K27me3 silencing, highlighting a direct influence of H3K36me on repressive TADs.


Asunto(s)
Cromatina , Histonas , Cromatina/genética , Histonas/genética , Heterocromatina/genética , Ensamble y Desensamble de Cromatina
6.
Trends Cell Biol ; 33(10): 887-897, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37061355

RESUMEN

Increase in lineage infidelity and/or imbalance is frequently observed around the earliest stage of breast tumor initiation. In response to disruption of homeostasis, differentiated cells can partially lose their identity and gain cellular plasticity, a process involving epigenome landscape remodeling. This increase of cellular plasticity may promote the malignant transformation of breast tumors and fuel their heterogeneity. Here, we review recent studies that have yield insights into important regulators of lineage integrity and mechanisms that trigger mammary epithelial lineage derail, and evaluate their impacts on breast tumor development.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Diferenciación Celular , Neoplasias de la Mama/genética , Células Epiteliales , Linaje de la Célula/fisiología
7.
Am J Hum Genet ; 109(12): 2196-2209, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36459980

RESUMEN

The TERT/CLPTM1L risk locus on chromosome 5p15.33 is a pleiotropic cancer risk locus in which multiple independent risk alleles have been identified, across well over ten cancer types. We previously conducted a genome-wide association study in uveal melanoma (UM), which uncovered a role for the TERT/CLPTM1L risk locus in this intraocular tumor and identified multiple highly correlated risk alleles. Aiming to unravel the biological mechanisms in UM of this locus, which contains a domain enriched in active chromatin marks and enhancer elements, we demonstrated the allele-specific enhancer activity of this risk region using reporter assays. In UM, we identified the functional variant rs452384, of which the C risk allele is associated with higher gene expression, increased CLPTM1L expression in UM tumors, and a longer telomere length in peripheral blood mononuclear cells. Electrophoretic mobility shift assays and quantitative mass spectrometry identified NKX2.4 as an rs452384-T-specific binding protein, whereas GATA4 preferentially interacted with rs452384-C. Knockdown of NKX2.4 but not GATA4 resulted in increased TERT and CLPTM1L expression. In summary, the UM risk conferred by the 5p locus is at least partly due to rs452384, for which NKX2.4 presents strong differential binding activity and regulates CLPTM1L and TERT expression. Altogether, our work unraveled some of the complex regulatory mechanisms at the 5p15.33 susceptibility region in UM, and this might also shed light on shared mechanisms with other tumor types affected by this susceptibility region.


Asunto(s)
Estudio de Asociación del Genoma Completo , Neoplasias de la Úvea , Humanos , Alelos , Leucocitos Mononucleares , Neoplasias de la Úvea/genética
8.
Methods Mol Biol ; 2529: 253-265, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733019

RESUMEN

Elucidating the biological function of histone methyltransferases requires knowledge of the genomic sites at which they act. CUT&RUN represents a valuable alternative to chromatin immunoprecipitation for the mapping of histone methylation patterns, generally producing results of equivalent quality while requiring less sequencing depth, less starting material and less effort. Automated CUT&RUN procedures have been developed to further facilitate chromatin profiling. Here we describe our automated CUT&RUN protocol using the Thermo Fisher KingFisher Duo Prime system.


Asunto(s)
Cromatina , Genoma , Cromatina/genética , Inmunoprecipitación de Cromatina/métodos , Histona Metiltransferasas/genética , Metilación
9.
Methods Mol Biol ; 2529: 297-311, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35733021

RESUMEN

Identification of histone lysine methyltransferase (HKMT) substrates has recently benefited from chemical-biology-based strategies in which artificial S-adenosyl-L-methionine (SAM) cofactors are engineered to allow substrate labeling using either the wild-type target enzyme or designed mutants. Once labeled, substrates can be selectively functionalized with an affinity tag, using a bioorthogonal ligation reaction, to allow their recovery from cell extracts and subsequent identification. In this chapter, we describe steps on how to proceed to set up such an approach to characterize substrates of specific HKMTs of the SET domain superfamily, from the characterization of the HKMT able to accommodate a SAM surrogate containing a bioorthogonal moiety, to the proteomic analysis conducted on a cell extract. We focus in particular on the controls that are necessary to ensure reliable proteomic data analysis. The example of PR-Set7 on which we have implemented this approach is shown.


Asunto(s)
Metionina , S-Adenosilmetionina , N-Metiltransferasa de Histona-Lisina/química , Dominios PR-SET , Proteómica , S-Adenosilmetionina/química
10.
Cell ; 185(12): 2164-2183.e25, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35597241

RESUMEN

X inactivation (XCI) is triggered by upregulation of XIST, which coats the chromosome in cis, promoting formation of a heterochromatic domain (Xi). XIST role beyond initiation of XCI is only beginning to be elucidated. Here, we demonstrate that XIST loss impairs differentiation of human mammary stem cells (MaSCs) and promotes emergence of highly tumorigenic and metastatic carcinomas. On the Xi, XIST deficiency triggers epigenetic changes and reactivation of genes overlapping Polycomb domains, including Mediator subunit MED14. MED14 overdosage results in increased Mediator levels and hyperactivation of the MaSC enhancer landscape and transcriptional program, making differentiation less favorable. We further demonstrate that loss of XIST and Xi transcriptional instability is common among human breast tumors of poor prognosis. We conclude that XIST is a gatekeeper of human mammary epithelium homeostasis, thus unveiling a paradigm in the control of somatic cell identity with potential consequences for our understanding of gender-specific malignancies.


Asunto(s)
Complejo Mediador/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Epigénesis Genética , Humanos , ARN Largo no Codificante/genética , Inactivación del Cromosoma X
11.
Dev Cell ; 57(8): 1037-1052.e8, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35429435

RESUMEN

Polycomb repressive complex 2 (PRC2) maintains transcriptionally silent genes in a repressed state via deposition of histone H3K27-trimethyl (me3) marks. PRC2 has also been implicated in silencing transposable elements (TEs), yet how PRC2 is targeted to TEs remains unclear. To address this question, we identified proteins that physically interact with the Paramecium enhancer-of-zeste Ezl1 enzyme, which catalyzes H3K9me3 and H3K27me3 deposition at TEs. We show that the Paramecium PRC2 core complex comprises four subunits, each required in vivo for catalytic activity. We also identify PRC2 cofactors, including the RNA interference (RNAi) effector Ptiwi09, which are necessary to target H3K9me3 and H3K27me3 to TEs. We find that the physical interaction between PRC2 and the RNAi pathway is mediated by a RING finger protein and that small RNA recruitment of PRC2 to TEs is analogous to the small RNA recruitment of H3K9 methylation SU(VAR)3-9 enzymes.


Asunto(s)
Paramecium , Complejo Represivo Polycomb 2 , Elementos Transponibles de ADN/genética , Histonas/metabolismo , Paramecium/genética , Paramecium/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , ARN
12.
Nat Genet ; 53(12): 1686-1697, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34782763

RESUMEN

Epigenetic inheritance of gene expression states enables a single genome to maintain distinct cellular identities. How histone modifications contribute to this process remains unclear. Using global chromatin perturbations and local, time-controlled modulation of transcription, we establish the existence of epigenetic memory of transcriptional activation for genes that can be silenced by the Polycomb group. This property emerges during cell differentiation and allows genes to be stably switched after a transient transcriptional stimulus. This transcriptional memory state at Polycomb targets operates in cis; however, rather than relying solely on read-and-write propagation of histone modifications, the memory is also linked to the strength of activating inputs opposing Polycomb proteins, and therefore varies with the cellular context. Our data and computational simulations suggest a model whereby transcriptional memory arises from double-negative feedback between Polycomb-mediated silencing and active transcription. Transcriptional memory at Polycomb targets thus depends not only on histone modifications but also on the gene-regulatory network and underlying identity of a cell.


Asunto(s)
Epigénesis Genética , Mamíferos/genética , Proteínas del Grupo Polycomb/genética , Activación Transcripcional , Animales , Femenino , Código de Histonas , Humanos , Masculino , Ratones , Complejo Represivo Polycomb 2/genética
13.
Oncogene ; 40(20): 3475-3491, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33934105

RESUMEN

Enhancers are cis-regulatory sequences that fine-tune expression of their target genes in a spatiotemporal manner. They are recognized by sequence-specific transcription factors, which in turn recruit transcriptional coactivators that facilitate transcription by promoting assembly and activation of the basal transcriptional machinery. Their functional importance is underscored by the fact that they are often the target of genetic and nongenetic events in human disease that disrupt their sequence, interactome, activation potential, and/or chromatin environment. Dysregulation of transcription and addiction to transcriptional effectors that interact with and modulate enhancer activity are common features of cancer cells and are amenable to therapeutic intervention. Here, we discuss the current knowledge on enhancer biology, the broad spectrum of mechanisms that lead to their malfunction in tumor cells, and recent progress in developing drugs that efficaciously target their dependencies.


Asunto(s)
Elementos de Facilitación Genéticos , Neoplasias/genética , Neoplasias/terapia , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Neoplasias/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Cancer Res ; 81(11): 2888-2902, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33888468

RESUMEN

Inactivation of Polybromo 1 (PBRM1), a specific subunit of the PBAF chromatin remodeling complex, occurs frequently in cancer, including 40% of clear cell renal cell carcinomas (ccRCC). To identify novel therapeutic approaches to targeting PBRM1-defective cancers, we used a series of orthogonal functional genomic screens that identified PARP and ATR inhibitors as being synthetic lethal with PBRM1 deficiency. The PBRM1/PARP inhibitor synthetic lethality was recapitulated using several clinical PARP inhibitors in a series of in vitro model systems and in vivo in a xenograft model of ccRCC. In the absence of exogenous DNA damage, PBRM1-defective cells exhibited elevated levels of replication stress, micronuclei, and R-loops. PARP inhibitor exposure exacerbated these phenotypes. Quantitative mass spectrometry revealed that multiple R-loop processing factors were downregulated in PBRM1-defective tumor cells. Exogenous expression of the R-loop resolution enzyme RNase H1 reversed the sensitivity of PBRM1-deficient cells to PARP inhibitors, suggesting that excessive levels of R-loops could be a cause of this synthetic lethality. PARP and ATR inhibitors also induced cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) innate immune signaling in PBRM1-defective tumor cells. Overall, these findings provide the preclinical basis for using PARP inhibitors in PBRM1-defective cancers. SIGNIFICANCE: This study demonstrates that PARP and ATR inhibitors are synthetic lethal with the loss of PBRM1, a PBAF-specific subunit, thus providing the rationale for assessing these inhibitors in patients with PBRM1-defective cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2888/F1.large.jpg.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/deficiencia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Renales/patología , Neoplasias Pulmonares/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Mutaciones Letales Sintéticas , Factores de Transcripción/deficiencia , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Proliferación Celular , Femenino , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
EMBO Rep ; 22(3): e51009, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33512761

RESUMEN

Histone post-translational modifications (PTMs) are key players in chromatin regulation. The identification of novel histone acylations raises important questions regarding their role in transcription. In this study, we characterize the role of an acylation on the lateral surface of the histone octamer, H3K122 succinylation (H3K122succ), in chromatin function and transcription. Using chromatin succinylated at H3K122 in in vitro transcription assays, we show that the presence of H3K122succ is sufficient to stimulate transcription. In line with this, we found in our ChIP assays H3K122succ enriched on promoters of active genes and H3K122succ enrichment scaling with gene expression levels. Furthermore, we show that the co-activators p300/CBP can succinylate H3K122 and identify sirtuin 5 (SIRT5) as a new desuccinylase. By applying single molecule FRET assays, we demonstrate a direct effect of H3K122succ on nucleosome stability, indicating an important role for histone succinylation in modulating chromatin dynamics. Together, these data provide the first insights into the mechanisms underlying transcriptional regulation by H3K122succ.


Asunto(s)
Histonas , Nucleosomas , Cromatina/genética , Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Nucleosomas/genética , Procesamiento Proteico-Postraduccional
16.
Curr Opin Chem Biol ; 56: 51-62, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31981999

RESUMEN

Targeting chromatin-modifying enzymes is a promising strategy for cancer treatment. The antitumor effectivity of compounds inhibiting histone methyltransferases - mainly EZH2 - is currently being tested in phase I/II clinical trials, some of them showing positive results in hematological malignancies and solid tumors of specific mutational background. In this review, we aim at highlighting the recent advances in the field of histone methyltransferase inhibitors and describing the challenges that need to be addressed for their successful implementation in the clinics.


Asunto(s)
Antineoplásicos/química , Inhibidores Enzimáticos/química , Histona Metiltransferasas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Carcinogénesis/metabolismo , Cromatina/genética , Cromatina/metabolismo , Diseño de Fármacos , Quimioterapia Combinada , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Humanos , Metilación , Mutación
17.
Nature ; 578(7793): E8, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932733

RESUMEN

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

18.
Nature ; 577(7791): 576-581, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31875854

RESUMEN

DNA replication is a tightly regulated process that ensures the precise duplication of the genome during the cell cycle1. In eukaryotes, the licensing and activation of replication origins are regulated by both DNA sequence and chromatin features2. However, the chromatin-based regulatory mechanisms remain largely uncharacterized. Here we show that, in HeLa cells, nucleosomes containing the histone variant H2A.Z are enriched with histone H4 that is dimethylated on its lysine 20 residue (H4K20me2) and with bound origin-recognition complex (ORC). In vitro studies show that H2A.Z-containing nucleosomes bind directly to the histone lysine methyltransferase enzyme SUV420H1, promoting H4K20me2 deposition, which is in turn required for ORC1 binding. Genome-wide studies show that signals from H4K20me2, ORC1 and nascent DNA strands co-localize with H2A.Z, and that depletion of H2A.Z results in decreased H4K20me2, ORC1 and nascent-strand signals throughout the genome. H2A.Z-regulated replication origins have a higher firing efficiency and early replication timing compared with other origins. Our results suggest that the histone variant H2A.Z epigenetically regulates the licensing and activation of early replication origins and maintains replication timing through the SUV420H1-H4K20me2-ORC1 axis.


Asunto(s)
Momento de Replicación del ADN , Replicación del ADN , Histonas/metabolismo , Origen de Réplica/genética , ADN/metabolismo , Replicación del ADN/genética , Epigénesis Genética , Células HeLa , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Humanos , Lisina/metabolismo , Metilación , Nucleosomas/química , Nucleosomas/metabolismo , Complejo de Reconocimiento del Origen/metabolismo
20.
Nat Commun ; 10(1): 3858, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31451685

RESUMEN

The Polycomb group of proteins is required for the proper orchestration of gene expression due to its role in maintaining transcriptional silencing. It is composed of several chromatin modifying complexes, including Polycomb Repressive Complex 2 (PRC2), which deposits H3K27me2/3. Here, we report the identification of a cofactor of PRC2, EZHIP (EZH1/2 Inhibitory Protein), expressed predominantly in the gonads. EZHIP limits the enzymatic activity of PRC2 and lessens the interaction between the core complex and its accessory subunits, but does not interfere with PRC2 recruitment to chromatin. Deletion of Ezhip in mice leads to a global increase in H3K27me2/3 deposition both during spermatogenesis and at late stages of oocyte maturation. This does not affect the initial number of follicles but is associated with a reduction of follicles in aging. Our results suggest that mature oocytes Ezhip-/- might not be fully functional and indicate that fertility is strongly impaired in Ezhip-/- females. Altogether, our study uncovers EZHIP as a regulator of chromatin landscape in gametes.


Asunto(s)
Proteínas Oncogénicas/metabolismo , Óvulo/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Espermatozoides/metabolismo , Adulto , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Femenino , Células HEK293 , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/aislamiento & purificación , Oogénesis , Ovario/citología , Ovario/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Células Sf9 , Espermatogénesis , Testículo/citología , Testículo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...