Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Opin Genet Dev ; 86: 102194, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636335

RESUMEN

Chromatin loops, which bring two distal loci of the same chromosome into close physical proximity, are the ubiquitous units of the three-dimensional genome. Recent advances in understanding the spatial organisation of chromatin suggest that several distinct mechanisms control chromatin interactions, such as loop extrusion by cohesin complexes, compartmentalisation by phase separation, direct protein-protein interactions and others. Here, we review different types of chromatin loops and highlight the factors and processes involved in their regulation. We discuss how loop extrusion and compartmentalisation shape chromatin interactions and how these two processes can either positively or negatively influence each other.


Asunto(s)
Proteínas de Ciclo Celular , Cromatina , Proteínas Cromosómicas no Histona , Cohesinas , Genoma , Cromatina/genética , Genoma/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Núcleo Celular/genética , Cromosomas/genética , Animales
2.
Nat Commun ; 14(1): 7762, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040699

RESUMEN

Malignant rhabdoid tumor (MRT) is a highly malignant and often lethal childhood cancer. MRTs are genetically defined by bi-allelic inactivating mutations in SMARCB1, a member of the BRG1/BRM-associated factors (BAF) chromatin remodeling complex. Mutations in BAF complex members are common in human cancer, yet their contribution to tumorigenesis remains in many cases poorly understood. Here, we study derailed regulatory landscapes as a consequence of SMARCB1 loss in the context of MRT. Our multi-omics approach on patient-derived MRT organoids reveals a dramatic reshaping of the regulatory landscape upon SMARCB1 reconstitution. Chromosome conformation capture experiments subsequently reveal patient-specific looping of distal enhancer regions with the promoter of the MYC oncogene. This intertumoral heterogeneity in MYC enhancer utilization is also present in patient MRT tissues as shown by combined single-cell RNA-seq and ATAC-seq. We show that loss of SMARCB1 activates patient-specific epigenetic reprogramming underlying MRT tumorigenesis.


Asunto(s)
Tumor Rabdoide , Humanos , Niño , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Proteína SMARCB1/genética , Factores de Transcripción/genética , Mutación , Regiones Promotoras Genéticas/genética , Carcinogénesis/genética
3.
Cell Rep ; 42(10): 113124, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37733591

RESUMEN

Acquired drug resistance is a major problem in the treatment of cancer. hTERT-immortalized, untransformed RPE-1 cells can acquire resistance to Taxol by derepressing the ABCB1 gene, encoding for the multidrug transporter P-gP. Here, we investigate how the ABCB1 gene is derepressed. ABCB1 activation is associated with reduced H3K9 trimethylation, increased H3K27 acetylation, and ABCB1 displacement from the nuclear lamina. While altering DNA methylation and H3K27 methylation had no major impact on ABCB1 expression, nor did it promote resistance, disrupting the nuclear lamina component Lamin B Receptor did promote the acquisition of a Taxol-resistant phenotype in a subset of cells. CRISPRa-mediated gene activation supported the notion that lamina dissociation influences ABCB1 derepression. We propose a model in which nuclear lamina dissociation of a repressed gene allows for its activation, implying that deregulation of the 3D genome topology could play an important role in tumor evolution and the acquisition of drug resistance.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias , Humanos , Resistencia a Antineoplásicos/genética , Paclitaxel/farmacología , Resistencia a Múltiples Medicamentos/genética , Neoplasias/genética , Metilación de ADN/genética , Línea Celular Tumoral
4.
EMBO J ; 42(20): e113150, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37691488

RESUMEN

Genome-wide transcriptional activity involves the binding of many transcription factors (TFs) to thousands of sites in the genome. Pioneer TFs are a class of TFs that maintain open chromatin and allow non-pioneer TFs access to their target sites. Determining which TF binding sites directly drive transcription remains a challenge. Here, we use acute protein depletion of the pioneer TF SOX2 to establish its functionality in maintaining chromatin accessibility. We show that thousands of accessible sites are lost within an hour of protein depletion, indicating rapid turnover of these sites in the absence of the pioneer factor. To understand the relationship with transcription, we performed nascent transcription analysis and found that open chromatin sites that are maintained by SOX2 are highly predictive of gene expression, in contrast to all other SOX2 binding sites. We use CRISPR-Cas9 genome editing in the Klf2 locus to functionally validate a predicted regulatory element. We conclude that the regulatory activity of SOX2 is exerted mainly at sites where it maintains accessibility and that other binding sites are largely dispensable for gene regulation.


Asunto(s)
Cromatina , Factores de Transcripción SOXB1 , Factores de Transcripción , Sitios de Unión , Cromatina/genética , Regulación de la Expresión Génica , Unión Proteica , Factores de Transcripción/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Ratones
5.
Mol Cell ; 83(16): 2834-2836, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37595552

RESUMEN

In this issue, Zhang et al.1 show that CTCF blocks cohesin-mediated loop extrusion in an orientation-dependent manner. Using single-molecule imaging assays, the authors find that dCas9 and R-loops can also stall extrusion.


Asunto(s)
Bioensayo , Elevación , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Cohesinas
6.
Nat Rev Genet ; 24(2): 73-85, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36180596

RESUMEN

Chromatin folds into dynamic loops that often span hundreds of kilobases and physically wire distant loci together for gene regulation. These loops are continuously created, extended and positioned by structural maintenance of chromosomes (SMC) protein complexes, such as condensin and cohesin, and their regulators, including CTCF, in a highly dynamic process known as loop extrusion. Genetic loss of extrusion factors is lethal, complicating their study. Inducible protein degradation technologies enable the depletion of loop extrusion factors within hours, leading to the rapid reconfiguration of chromatin folding. Here, we review how these technologies have changed our understanding of genome organization, upsetting long-held beliefs on its role in transcription. Finally, we examine recent models that attempt to reconcile observations after chronic versus acute perturbations, and discuss future developments in this rapidly developing field of research.


Asunto(s)
Cromatina , Cromosomas , Cromosomas/genética , Regulación de la Expresión Génica , Genoma , Proteínas de Ciclo Celular/genética
7.
EMBO Rep ; 23(12): e55782, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36245428

RESUMEN

Ki-67 is a chromatin-associated protein with a dynamic distribution pattern throughout the cell cycle and is thought to be involved in chromatin organization. The lack of genomic interaction maps has hampered a detailed understanding of its roles, particularly during interphase. By pA-DamID mapping in human cell lines, we find that Ki-67 associates with large genomic domains that overlap mostly with late-replicating regions. Early in interphase, when Ki-67 is present in pre-nucleolar bodies, it interacts with these domains on all chromosomes. However, later in interphase, when Ki-67 is confined to nucleoli, it shows a striking shift toward small chromosomes. Nucleolar perturbations indicate that these cell cycle dynamics correspond to nucleolar maturation during interphase, and suggest that nucleolar sequestration of Ki-67 limits its interactions with larger chromosomes. Furthermore, we demonstrate that Ki-67 does not detectably control chromatin-chromatin interactions during interphase, but it competes with the nuclear lamina for interaction with late-replicating DNA, and it controls replication timing of (peri)centromeric regions. Together, these results reveal a highly dynamic choreography of genome interactions and roles for Ki-67 in heterochromatin organization.


Asunto(s)
Genómica , Heterocromatina , Humanos , Heterocromatina/genética , Antígeno Ki-67/genética
8.
Nat Struct Mol Biol ; 29(10): 1000-1010, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36220895

RESUMEN

Precise control of gene expression underpins normal development. This relies on mechanisms that enable communication between gene promoters and other regulatory elements. In embryonic stem cells (ESCs), the cyclin-dependent kinase module Mediator complex (CKM-Mediator) has been reported to physically link gene regulatory elements to enable gene expression and also prime genes for induction during differentiation. Here, we show that CKM-Mediator contributes little to three-dimensional genome organization in ESCs, but it has a specific and essential role in controlling interactions between inactive gene regulatory elements bound by Polycomb repressive complexes (PRCs). These interactions are established by the canonical PRC1 (cPRC1) complex but rely on CKM-Mediator, which facilitates binding of cPRC1 to its target sites. Importantly, through separation-of-function experiments, we reveal that this collaboration between CKM-Mediator and cPRC1 in creating long-range interactions does not function to prime genes for induction during differentiation. Instead, we discover that priming relies on an interaction-independent mechanism whereby the CKM supports core Mediator engagement with gene promoters during differentiation to enable gene activation.


Asunto(s)
Complejo Mediador , Complejo Represivo Polycomb 1 , Diferenciación Celular/genética , Quinasas Ciclina-Dependientes/metabolismo , Complejo Mediador/genética , Complejo Represivo Polycomb 1/genética , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
9.
Genome Biol ; 23(1): 185, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36050765

RESUMEN

BACKGROUND: Lamina-associated domains (LADs) are large genomic regions that are positioned at the nuclear lamina. It has remained largely unclear what drives the positioning and demarcation of LADs. Because the insulator protein CTCF is enriched at LAD borders, it was postulated that CTCF binding could position some LAD boundaries, possibly through its function in stalling cohesin and hence preventing cohesin invading into the LAD. To test this, we mapped genome-nuclear lamina interactions in mouse embryonic stem cells after rapid depletion of CTCF and other perturbations of cohesin dynamics. RESULTS: CTCF and cohesin contribute to a sharp transition in lamina interactions at LAD borders, while LADs are maintained after depletion of these proteins, also at borders marked by CTCF. CTCF and cohesin may thus reinforce LAD borders, but do not position these. CTCF binding sites within LADs are locally detached from the lamina and enriched for accessible DNA and active histone modifications. Remarkably, despite lamina positioning being strongly correlated with genome inactivity, this DNA remains accessible after the local detachment is lost following CTCF depletion. At a chromosomal scale, cohesin depletion and cohesin stabilization by depletion of the unloading factor WAPL quantitatively affect lamina interactions, indicative of perturbed chromosomal positioning in the nucleus. Finally, while H3K27me3 is locally enriched at CTCF-marked LAD borders, we find no evidence for an interplay between CTCF and H3K27me3 on lamina interactions. CONCLUSIONS: These findings illustrate that CTCF and cohesin are not primary determinants of LAD patterns. Rather, these proteins locally modulate NL interactions.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Histonas , Lámina Nuclear , Animales , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Ratones , Lámina Nuclear/química , Cohesinas
10.
Methods Mol Biol ; 2532: 311-331, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35867256

RESUMEN

The organization of the genome inside the nucleus facilitates many nuclear processes. Because the nuclear genome is highly dynamic and often regulated by essential proteins, rapid depletion strategies are necessary to perform loss-of-function analyses. Fortunately, in recent years, various methods have been developed to manipulate the cellular levels of a protein directly and acutely. Here, we describe different methods that have been developed to rapidly deplete proteins from cells, with a focus on auxin inducible degron and dTAG methods, as these are most commonly used in 3D genome organization studies. We outline best practices for designing a knockin strategy, as well as generation and validation of knockin cell lines. Acute depletion strategies have been transformative for the study of the 3D genome and will be important tools for delineating the processes and factors that determine organization of the genome inside the nucleus.


Asunto(s)
Ácidos Indolacéticos , Proteínas , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo , Genoma , Ácidos Indolacéticos/metabolismo , Proteínas/metabolismo , Proteolisis
11.
Nat Struct Mol Biol ; 29(6): 586-591, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35710836

RESUMEN

Cohesin structures the genome through the formation of chromatin loops and by holding together the sister chromatids. The acetylation of cohesin's SMC3 subunit is a dynamic process that involves the acetyltransferase ESCO1 and deacetylase HDAC8. Here we show that this cohesin acetylation cycle controls the three-dimensional genome in human cells. ESCO1 restricts the length of chromatin loops, and of architectural stripes emanating from CTCF sites. HDAC8 conversely promotes the extension of such loops and stripes. This role in controlling loop length turns out to be distinct from the canonical role of cohesin acetylation that protects against WAPL-mediated DNA release. We reveal that acetylation controls the interaction of cohesin with PDS5A to restrict chromatin loop length. Our data support a model in which this PDS5A-bound state acts as a brake that enables the pausing and restart of loop enlargement. The cohesin acetylation cycle hereby provides punctuation in the process of genome folding.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Acetilación , Proteínas de Ciclo Celular/metabolismo , Cromátides/metabolismo , Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Histona Desacetilasas/genética , Humanos , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Cohesinas
12.
Nat Commun ; 13(1): 754, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136067

RESUMEN

The genome consists of regions of transcriptionally active euchromatin and more silent heterochromatin. We reveal that the formation of heterochromatin domains requires cohesin turnover on DNA. Stabilization of cohesin on DNA through depletion of its release factor WAPL leads to a near-complete loss of heterochromatin domains. We observe the opposite phenotype in cells deficient for subunits of the Mediator-CDK module, with an almost binary partition of the genome into dense H3K9me3 domains, and regions devoid of H3K9me3 spanning the rest of the genome. We suggest that the Mediator-CDK module might contribute to gene expression by limiting the formation of dense heterochromatin domains. WAPL deficiency prevents the formation of heterochromatin domains, and allows for gene expression even in the absence of the Mediator-CDK subunit MED12. We propose that cohesin and Mediator affect heterochromatin in different ways to enable the correct distribution of epigenetic marks, and thus to ensure proper gene expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Complejo Mediador/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Portadoras/genética , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Epigénesis Genética , Técnicas de Inactivación de Genes , Humanos , Complejo Mediador/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , RNA-Seq , Cohesinas
13.
EMBO Rep ; 23(2): e53902, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34927791

RESUMEN

The discovery of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and its development as a genome editing tool has revolutionized the field of molecular biology. In the DNA damage field, CRISPR has brought an alternative to induce endogenous double-strand breaks (DSBs) at desired genomic locations and study the DNA damage response and its consequences. Many systems for sgRNA delivery have been reported in order to efficiently generate this DSB, including lentiviral vectors. However, some of the consequences of these systems are not yet well understood. Here, we report that lentiviral-based sgRNA vectors can integrate into the endogenous genomic target location, leading to undesired activation of the target gene. By generating a DSB in the regulatory region of the ABCB1 gene using a lentiviral sgRNA vector, we can induce the formation of Taxol-resistant colonies. We show that these colonies upregulate ABCB1 via integration of the EEF1A1 and the U6 promoters from the sgRNA vector. We believe that this is an unreported CRISPR/Cas9 on-target effect that researchers need to be aware of when using lentiviral vectors for genome editing.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Activación Transcripcional
14.
Nat Commun ; 12(1): 4360, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272384

RESUMEN

The glucocorticoid receptor (GR) regulates gene expression, governing aspects of homeostasis, but is also involved in cancer. Pharmacological GR activation is frequently used to alleviate therapy-related side-effects. While prior studies have shown GR activation might also have anti-proliferative action on tumours, the underpinnings of glucocorticoid action and its direct effectors in non-lymphoid solid cancers remain elusive. Here, we study the mechanisms of glucocorticoid response, focusing on lung cancer. We show that GR activation induces reversible cancer cell dormancy characterised by anticancer drug tolerance, and activation of growth factor survival signalling accompanied by vulnerability to inhibitors. GR-induced dormancy is dependent on a single GR-target gene, CDKN1C, regulated through chromatin looping of a GR-occupied upstream distal enhancer in a SWI/SNF-dependent fashion. These insights illustrate the importance of GR signalling in non-lymphoid solid cancer biology, particularly in lung cancer, and warrant caution for use of glucocorticoids in treatment of anticancer therapy related side-effects.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Cromatina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Glucocorticoides/farmacología , Neoplasias Pulmonares/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Imidazoles/farmacología , Inmunohistoquímica , Neoplasias Pulmonares/genética , Ratones , Proteómica , Pirazinas/farmacología , ARN Interferente Pequeño , RNA-Seq , Receptor IGF Tipo 1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
NAR Genom Bioinform ; 3(2): lqab040, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34046591

RESUMEN

Conformation capture-approaches like Hi-C can elucidate chromosome structure at a genome-wide scale. Hi-C datasets are large and require specialised software. Here, we present GENOVA: a user-friendly software package to analyse and visualise chromosome conformation capture (3C) data. GENOVA is an R-package that includes the most common Hi-C analyses, such as compartment and insulation score analysis. It can create annotated heatmaps to visualise the contact frequency at a specific locus and aggregate Hi-C signal over user-specified genomic regions such as ChIP-seq data. Finally, our package supports output from the major mapping-pipelines. We demonstrate the capabilities of GENOVA by analysing Hi-C data from HAP1 cell lines in which the cohesin-subunits SA1 and SA2 were knocked out. We find that ΔSA1 cells gain intra-TAD interactions and increase compartmentalisation. ΔSA2 cells have longer loops and a less compartmentalised genome. These results suggest that cohesinSA1 forms longer loops, while cohesinSA2 plays a role in forming and maintaining intra-TAD interactions. Our data supports the model that the genome is provided structure in 3D by the counter-balancing of loop formation on one hand, and compartmentalization on the other hand. By differentially controlling loops, cohesinSA1 and cohesinSA2 therefore also affect nuclear compartmentalization. We show that GENOVA is an easy to use R-package, that allows researchers to explore Hi-C data in great detail.

16.
EMBO Rep ; 22(2): e51184, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33410591

RESUMEN

Differentiation of naïve peripheral B cells into terminally differentiated plasma cells is characterized by epigenetic alterations, yet the epigenetic mechanisms that control B-cell fate remain unclear. Here, we identified a role for the histone H3K79 methyltransferase DOT1L in controlling B-cell differentiation. Mouse B cells lacking Dot1L failed to establish germinal centers (GC) and normal humoral immune responses in vivo. In vitro, activated B cells in which Dot1L was deleted showed aberrant differentiation and prematurely acquired plasma cell characteristics. Similar results were obtained when DOT1L was chemically inhibited in mature B cells in vitro. Mechanistically, combined epigenomics and transcriptomics analysis revealed that DOT1L promotes expression of a pro-proliferative, pro-GC program. In addition, DOT1L indirectly supports the repression of an anti-proliferative plasma cell differentiation program by maintaining the repression of Polycomb Repressor Complex 2 (PRC2) targets. Our findings show that DOT1L is a key modulator of the core transcriptional and epigenetic landscape in B cells, establishing an epigenetic barrier that warrants B-cell naivety and GC B-cell differentiation.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular , N-Metiltransferasa de Histona-Lisina , Histonas , Metiltransferasas , Animales , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones
17.
Nat Genet ; 53(1): 100-109, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33318687

RESUMEN

The cohesin complex has an essential role in maintaining genome organization. However, its role in gene regulation remains largely unresolved. Here we report that the cohesin release factor WAPL creates a pool of free cohesin, in a process known as cohesin turnover, which reloads it to cell-type-specific binding sites. Paradoxically, stabilization of cohesin binding, following WAPL ablation, results in depletion of cohesin from these cell-type-specific regions, loss of gene expression and differentiation. Chromosome conformation capture experiments show that cohesin turnover is important for maintaining promoter-enhancer loops. Binding of cohesin to cell-type-specific sites is dependent on the pioneer transcription factors OCT4 (POU5F1) and SOX2, but not NANOG. We show the importance of cohesin turnover in controlling transcription and propose that a cycle of cohesin loading and off-loading, instead of static cohesin binding, mediates promoter and enhancer interactions critical for gene regulation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación de la Expresión Génica , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteínas/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/genética , Línea Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Ratones , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Cohesinas
18.
Proc Natl Acad Sci U S A ; 117(34): 20706-20716, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32764145

RESUMEN

Cytotoxic T cell differentiation is guided by epigenome adaptations, but how epigenetic mechanisms control lymphocyte development has not been well defined. Here we show that the histone methyltransferase DOT1L, which marks the nucleosome core on active genes, safeguards normal differentiation of CD8+ T cells. T cell-specific ablation of Dot1L resulted in loss of naïve CD8+ T cells and premature differentiation toward a memory-like state, independent of antigen exposure and in a cell-intrinsic manner. Mechanistically, DOT1L controlled CD8+ T cell differentiation by ensuring normal T cell receptor density and signaling. DOT1L also maintained epigenetic identity, in part by indirectly supporting the repression of developmentally regulated genes. Finally, deletion of Dot1L in T cells resulted in an impaired immune response. Through our study, DOT1L is emerging as a central player in physiology of CD8+ T cells, acting as a barrier to prevent premature differentiation and controlling epigenetic integrity.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Diferenciación Celular/genética , Epigénesis Genética/genética , Epigenómica , Femenino , Histona Metiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/metabolismo , Masculino , Metiltransferasas/metabolismo , Ratones
19.
Nature ; 578(7795): 472-476, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31905366

RESUMEN

Cohesin catalyses the folding of the genome into loops that are anchored by CTCF1. The molecular mechanism of how cohesin and CTCF structure the 3D genome has remained unclear. Here we show that a segment within the CTCF N terminus interacts with the SA2-SCC1 subunits of human cohesin. We report a crystal structure of SA2-SCC1 in complex with CTCF at a resolution of 2.7 Å, which reveals the molecular basis of the interaction. We demonstrate that this interaction is specifically required for CTCF-anchored loops and contributes to the positioning of cohesin at CTCF binding sites. A similar motif is present in a number of established and newly identified cohesin ligands, including the cohesin release factor WAPL2,3. Our data suggest that CTCF enables the formation of chromatin loops by protecting cohesin against loop release. These results provide fundamental insights into the molecular mechanism that enables the dynamic regulation of chromatin folding by cohesin and CTCF.


Asunto(s)
Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Cromatina/química , Cromatina/metabolismo , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Humanos , Ligandos , Modelos Moleculares , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estabilidad Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Cohesinas
20.
J Mol Biol ; 432(3): 638-642, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31654669

RESUMEN

Developments in proximity ligation methods and sequencing technologies have provided high-resolution views of the organization of the genome inside the nucleus. A prominent feature of Hi-C maps is regions of increased self-interaction called topologically associating domains (TADs). Despite the strong evolutionary conservation and clear link with gene expression, the exact role of TADs and even their definition remains debatable. Here, I review the discovery of TADs, how they are commonly identified, and the mechanisms that lead to their formation. Furthermore, I discuss recent results that have created a more nuanced view of the role of TADs in the regulation of genes. In light of this, I propose that when we define TADs, we also consider the mechanisms that shape them.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Biología Molecular/historia , Conformación Molecular , Regulación de la Expresión Génica , Historia del Siglo XXI , Biología Molecular/tendencias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...