Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell ; 81(15): 3082-3095.e6, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34197738

RESUMEN

To understand how chromatin domains coordinate gene expression, we dissected select genetic elements organizing topology and transcription around the Prdm14 super enhancer in mouse embryonic stem cells. Taking advantage of allelic polymorphisms, we developed methods to sensitively analyze changes in chromatin topology, gene expression, and protein recruitment. We show that enhancer insulation does not rely strictly on loop formation between its flanking boundaries, that the enhancer activates the Slco5a1 gene beyond its prominent domain boundary, and that it recruits cohesin for loop extrusion. Upon boundary inversion, we find that oppositely oriented CTCF terminates extrusion trajectories but does not stall cohesin, while deleted or mutated CTCF sites allow cohesin to extend its trajectory. Enhancer-mediated gene activation occurs independent of paused loop extrusion near the gene promoter. We expand upon the loop extrusion model to propose that cohesin loading and extrusion trajectories originating at an enhancer contribute to gene activation.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Elementos de Facilitación Genéticos , Animales , Factor de Unión a CCCTC/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Ratones , Células Madre Embrionarias de Ratones , Coactivador 2 del Receptor Nuclear/genética , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Cohesinas
2.
Mol Cell ; 61(3): 461-473, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26833089

RESUMEN

Detailed genomic contact maps have revealed that chromosomes are structurally organized in megabase-sized topologically associated domains (TADs) that encompass smaller subTADs. These domains segregate in the nuclear space to form active and inactive nuclear compartments, but cause and consequence of compartmentalization are largely unknown. Here, we combined lacO/lacR binding platforms with allele-specific 4C technologies to track their precise position in the three-dimensional genome upon recruitment of NANOG, SUV39H1, or EZH2. We observed locked genomic loci resistant to spatial repositioning and unlocked loci that could be repositioned to different nuclear subcompartments with distinct chromatin signatures. Focal protein recruitment caused the entire subTAD, but not surrounding regions, to engage in new genomic contacts. Compartment switching was found uncoupled from transcription changes, and the enzymatic modification of histones per se was insufficient for repositioning. Collectively, this suggests that trans-associated factors influence three-dimensional compartmentalization independent of their cis effect on local chromatin composition and activity.


Asunto(s)
Núcleo Celular/metabolismo , Segregación Cromosómica , Células Madre Embrionarias/metabolismo , Sitios Genéticos , Operón Lac , Represoras Lac/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Proteína Potenciadora del Homólogo Zeste 2 , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Represoras Lac/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteína Homeótica Nanog , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transfección
3.
Mol Cell ; 60(4): 676-84, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26527277

RESUMEN

CCCTC-binding factor (CTCF) is an architectural protein involved in the three-dimensional (3D) organization of chromatin. In this study, we assayed the 3D genomic contact profiles of a large number of CTCF binding sites with high-resolution 4C-seq. As recently reported, our data also suggest that chromatin loops preferentially form between CTCF binding sites oriented in a convergent manner. To directly test this, we used CRISPR/Cas9 genome editing to delete core CTCF binding sites in three loci, including the CTCF site in the Sox2 super-enhancer. In all instances, CTCF and cohesin recruitment were lost, and chromatin loops with distal, convergent CTCF sites were disrupted or destabilized. Re-insertion of oppositely oriented CTCF recognition sequences restored CTCF and cohesin recruitment, but did not re-establish chromatin loops. We conclude that CTCF binding polarity plays a functional role in the formation of higher-order chromatin structure.


Asunto(s)
Cromatina/química , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Factor de Unión a CCCTC , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/citología , Ratones , Unión Proteica , Cohesinas
4.
Am J Hum Genet ; 101(3): 326-339, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28844486

RESUMEN

During pregnancy, cell-free DNA (cfDNA) in maternal blood encompasses a small percentage of cell-free fetal DNA (cffDNA), an easily accessible source for determination of fetal disease status in risk families through non-invasive procedures. In case of monogenic heritable disease, background maternal cfDNA prohibits direct observation of the maternally inherited allele. Non-invasive prenatal diagnostics (NIPD) of monogenic diseases therefore relies on parental haplotyping and statistical assessment of inherited alleles from cffDNA, techniques currently unavailable for routine clinical practice. Here, we present monogenic NIPD (MG-NIPD), which requires a blood sample from both parents, for targeted locus amplification (TLA)-based phasing of heterozygous variants selectively at a gene of interest. Capture probes-based targeted sequencing of cfDNA from the pregnant mother and a tailored statistical analysis enables predicting fetal gene inheritance. MG-NIPD was validated for 18 pregnancies, focusing on CFTR, CYP21A2, and HBB. In all cases we could predict the inherited alleles with >98% confidence, even at relatively early stages (8 weeks) of pregnancy. This prediction and the accuracy of parental haplotyping was confirmed by sequencing of fetal material obtained by parallel invasive procedures. MG-NIPD is a robust method that requires standard instrumentation and can be implemented in any clinic to provide families carrying a severe monogenic disease with a prenatal diagnostic test based on a simple blood draw.


Asunto(s)
Hiperplasia Suprarrenal Congénita/diagnóstico , Biomarcadores/sangre , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/diagnóstico , Polimorfismo de Nucleótido Simple , Diagnóstico Prenatal/métodos , Esteroide 21-Hidroxilasa/genética , Hiperplasia Suprarrenal Congénita/sangre , Hiperplasia Suprarrenal Congénita/genética , Células Cultivadas , Fibrosis Quística/sangre , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/sangre , ADN/sangre , ADN/genética , Femenino , Haplotipos , Humanos , Embarazo , Esteroide 21-Hidroxilasa/sangre
5.
Nature ; 501(7466): 227-31, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-23883933

RESUMEN

It is becoming increasingly clear that the shape of the genome importantly influences transcription regulation. Pluripotent stem cells such as embryonic stem cells were recently shown to organize their chromosomes into topological domains that are largely invariant between cell types. Here we combine chromatin conformation capture technologies with chromatin factor binding data to demonstrate that inactive chromatin is unusually disorganized in pluripotent stem-cell nuclei. We show that gene promoters engage in contacts between topological domains in a largely tissue-independent manner, whereas enhancers have a more tissue-restricted interaction profile. Notably, genomic clusters of pluripotency factor binding sites find each other very efficiently, in a manner that is strictly pluripotent-stem-cell-specific, dependent on the presence of Oct4 and Nanog protein and inducible after artificial recruitment of Nanog to a selected chromosomal site. We conclude that pluripotent stem cells have a unique higher-order genome structure shaped by pluripotency factors. We speculate that this interactome enhances the robustness of the pluripotent state.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Posicionamiento de Cromosoma , Genoma/genética , Imagenología Tridimensional , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Sitios de Unión , Línea Celular , Cromatina/genética , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Imagen Molecular , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/metabolismo
6.
Genome Res ; 25(7): 958-69, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25883320

RESUMEN

Despite recent progress in genome topology knowledge, the role of repeats, which make up the majority of mammalian genomes, remains elusive. Satellite repeats are highly abundant sequences that cluster around centromeres, attract pericentromeric heterochromatin, and aggregate into nuclear chromocenters. These nuclear landmark structures are assumed to form a repressive compartment in the nucleus to which genes are recruited for silencing. We have designed a strategy for genome-wide identification of pericentromere-associated domains (PADs) in different mouse cell types. The ∼1000 PADs and non-PADs have similar chromatin states in embryonic stem cells, but during lineage commitment, chromocenters progressively associate with constitutively inactive genomic regions at the nuclear periphery. This suggests that PADs are not actively recruited to chromocenters, but that chromocenters are themselves attracted to inactive chromatin compartments. However, we also found that experimentally induced proximity of an active locus to chromocenters was sufficient to cause gene repression. Collectively, our data suggest that rather than driving nuclear organization, pericentromeric satellite repeats mostly co-segregate with inactive genomic regions into nuclear compartments where they can contribute to stable maintenance of the repressed status of proximal chromosomal regions.


Asunto(s)
Centrómero/genética , Genómica , Repeticiones de Minisatélite , Animales , Eucromatina , Regulación de la Expresión Génica , Genómica/métodos , Heterocromatina , Ratones , Activación Transcripcional
7.
Nat Methods ; 9(10): 969-72, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22961246

RESUMEN

Regulatory DNA elements can control the expression of distant genes via physical interactions. Here we present a cost-effective methodology and computational analysis pipeline for robust characterization of the physical organization around selected promoters and other functional elements using chromosome conformation capture combined with high-throughput sequencing (4C-seq). Our approach can be multiplexed and routinely integrated with other functional genomics assays to facilitate physical characterization of gene regulation.


Asunto(s)
ADN/química , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Regulación de la Expresión Génica , Región de Control de Posición , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas
8.
Nucleic Acids Res ; 41(14): 6905-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23748562

RESUMEN

In developing B cells, the immunoglobulin heavy chain (IgH) locus is thought to move from repressive to permissive chromatin compartments to facilitate its scheduled rearrangement. In mature B cells, maintenance of allelic exclusion has been proposed to involve recruitment of the non-productive IgH allele to pericentromeric heterochromatin. Here, we used an allele-specific chromosome conformation capture combined with sequencing (4C-seq) approach to unambigously follow the individual IgH alleles in mature B lymphocytes. Despite their physical and functional difference, productive and non-productive IgH alleles in B cells and unrearranged IgH alleles in T cells share many chromosomal contacts and largely reside in active chromatin. In brain, however, the locus resides in a different repressive environment. We conclude that IgH adopts a lymphoid-specific nuclear location that is, however, unrelated to maintenance of allelic exclusion. We additionally find that in mature B cells-but not in T cells-the distal VH regions of both IgH alleles position themselves away from active chromatin. This, we speculate, may help to restrict enhancer activity to the productively rearranged VH promoter element.


Asunto(s)
Alelos , Linfocitos B/inmunología , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Animales , Núcleo Celular/química , Cromatina/química , Cromosomas de los Mamíferos , Sitios Genéticos , Cadenas Pesadas de Inmunoglobulina/análisis , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Región Variable de Inmunoglobulina/genética , Ratones , Recombinación Genética , Análisis de Secuencia de ADN , Bazo/inmunología , Linfocitos T/inmunología , Transcripción Genética
9.
Cell Rep ; 42(4): 112373, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37060567

RESUMEN

Monoallelic inactivation of CCCTC-binding factor (CTCF) in human cancer drives altered methylated genomic states, altered CTCF occupancy at promoter and enhancer regions, and deregulated global gene expression. In patients with T cell acute lymphoblastic leukemia (T-ALL), we find that acquired monoallelic CTCF-inactivating events drive subtle and local genomic effects in nearly half of t(5; 14) (q35; q32.2) rearranged patients, especially when CTCF-binding sites are preserved in between the BCL11B enhancer and the TLX3 oncogene. These solitary intervening sites insulate TLX3 from the enhancer by inducing competitive looping to multiple binding sites near the TLX3 promoter. Reduced CTCF levels or deletion of the intervening CTCF site abrogates enhancer insulation by weakening competitive looping while favoring TLX3 promoter to BCL11B enhancer looping, which elevates oncogene expression levels and leukemia burden.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina , Elementos de Facilitación Genéticos/genética , Mutación , Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
10.
Nat Struct Mol Biol ; 29(6): 563-574, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35710842

RESUMEN

Developmental gene expression is often controlled by distal regulatory DNA elements called enhancers. Distant enhancer action is restricted to structural chromosomal domains that are flanked by CTCF-associated boundaries and formed through cohesin chromatin loop extrusion. To better understand how enhancers, genes and CTCF boundaries together form structural domains and control expression, we used a bottom-up approach, building series of active regulatory landscapes in inactive chromatin. We demonstrate here that gene transcription levels and activity over time reduce with increased enhancer distance. The enhancer recruits cohesin to stimulate domain formation and engage flanking CTCF sites in loop formation. It requires cohesin exclusively for the activation of distant genes, not of proximal genes, with nearby CTCF boundaries supporting efficient long-range enhancer action. Our work supports a dual activity model for enhancers: its classic role of stimulating transcription initiation and elongation from target gene promoters and a role of recruiting cohesin for the creation of chromosomal domains, the engagement of CTCF sites in chromatin looping and the activation of distal target genes.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Sitios de Unión , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos/genética , Cohesinas
11.
Nat Commun ; 12(1): 3361, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099699

RESUMEN

In routine diagnostic pathology, cancer biopsies are preserved by formalin-fixed, paraffin-embedding (FFPE) procedures for examination of (intra-) cellular morphology. Such procedures inadvertently induce DNA fragmentation, which compromises sequencing-based analyses of chromosomal rearrangements. Yet, rearrangements drive many types of hematolymphoid malignancies and solid tumors, and their manifestation is instructive for diagnosis, prognosis, and treatment. Here, we present FFPE-targeted locus capture (FFPE-TLC) for targeted sequencing of proximity-ligation products formed in FFPE tissue blocks, and PLIER, a computational framework that allows automated identification and characterization of rearrangements involving selected, clinically relevant, loci. FFPE-TLC, blindly applied to 149 lymphoma and control FFPE samples, identifies the known and previously uncharacterized rearrangement partners. It outperforms fluorescence in situ hybridization (FISH) in sensitivity and specificity, and shows clear advantages over standard capture-NGS methods, finding rearrangements involving repetitive sequences which they typically miss. FFPE-TLC is therefore a powerful clinical diagnostics tool for accurate targeted rearrangement detection in FFPE specimens.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Linfoma de Células B/genética , Linfoma no Hodgkin/genética , Adhesión en Parafina/métodos , Fijación del Tejido/métodos , Translocación Genética , Biología Computacional/métodos , Reordenamiento Génico , Genes bcl-2/genética , Genes myc/genética , Humanos , Hibridación Fluorescente in Situ/métodos , Linfoma de Células B/diagnóstico , Linfoma no Hodgkin/diagnóstico , Proteínas Proto-Oncogénicas c-bcl-6/genética , Reproducibilidad de los Resultados , Estudios Retrospectivos , Sensibilidad y Especificidad
12.
Nat Protoc ; 15(2): 364-397, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932773

RESUMEN

We present the experimental protocol and data analysis toolbox for multi-contact 4C (MC-4C), a new proximity ligation method tailored to study the higher-order chromatin contact patterns of selected genomic sites. Conventional chromatin conformation capture (3C) methods fragment proximity ligation products for efficient analysis of pairwise DNA contacts. By contrast, MC-4C is designed to preserve and collect large concatemers of proximity ligated fragments for long-molecule sequencing on an Oxford Nanopore or Pacific Biosciences platform. Each concatemer of proximity ligation products represents a snapshot topology of a different individual allele, revealing its multi-way chromatin interactions. By inverse PCR with primers specific for a fragment of interest (the viewpoint) and DNA size selection, sequencing is selectively targeted to thousands of different complex interactions containing this viewpoint. A tailored statistical analysis toolbox is able to generate background models and three-way interaction profiles from the same dataset. These profiles can be used to distinguish whether contacts between more than two regulatory sequences are mutually exclusive or, conversely, simultaneously occurring at chromatin hubs. The entire procedure can be completed in 2 w, and requires standard molecular biology and data analysis skills and equipment, plus access to a third-generation sequencing platform.


Asunto(s)
Cromatina/química , Cromatina/genética , Análisis de Secuencia de ADN/métodos , Humanos , Células K562 , Conformación Molecular
13.
Nat Genet ; 50(8): 1151-1160, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988121

RESUMEN

Chromatin folding contributes to the regulation of genomic processes such as gene activity. Existing conformation capture methods characterize genome topology through analysis of pairwise chromatin contacts in populations of cells but cannot discern whether individual interactions occur simultaneously or competitively. Here we present multi-contact 4C (MC-4C), which applies Nanopore sequencing to study multi-way DNA conformations of individual alleles. MC-4C distinguishes cooperative from random and competing interactions and identifies previously missed structures in subpopulations of cells. We show that individual elements of the ß-globin superenhancer can aggregate into an enhancer hub that can simultaneously accommodate two genes. Neighboring chromatin domain loops can form rosette-like structures through collision of their CTCF-bound anchors, as seen most prominently in cells lacking the cohesin-unloading factor WAPL. Here, massive collision of CTCF-anchored chromatin loops is believed to reflect 'cohesin traffic jams'. Single-allele topology studies thus help us understand the mechanisms underlying genome folding and functioning.


Asunto(s)
Cromatina/genética , Elementos de Facilitación Genéticos/genética , Alelos , Animales , Factor de Unión a CCCTC/genética , Ratones , Conformación de Ácido Nucleico , Secuencias Reguladoras de Ácidos Nucleicos/genética , Globinas beta/genética
15.
Genome Biol ; 16: 289, 2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26700097

RESUMEN

BACKGROUND: Linker histone H1 is a core chromatin component that binds to nucleosome core particles and the linker DNA between nucleosomes. It has been implicated in chromatin compaction and gene regulation and is anticipated to play a role in higher-order genome structure. Here we have used a combination of genome-wide approaches including DNA methylation, histone modification and DNase I hypersensitivity profiling as well as Hi-C to investigate the impact of reduced cellular levels of histone H1 in embryonic stem cells on chromatin folding and function. RESULTS: We find that depletion of histone H1 changes the epigenetic signature of thousands of potential regulatory sites across the genome. Many of them show cooperative loss or gain of multiple chromatin marks. Epigenetic alterations cluster to gene-dense topologically associating domains (TADs) that already showed a high density of corresponding chromatin features. Genome organization at the three-dimensional level is largely intact, but we find changes in the structural segmentation of chromosomes specifically for the epigenetically most modified TADs. CONCLUSIONS: Our data show that cells require normal histone H1 levels to expose their proper regulatory landscape. Reducing the levels of histone H1 results in massive epigenetic changes and altered topological organization particularly at the most active chromosomal domains. Changes in TAD configuration coincide with epigenetic landscape changes but not with transcriptional output changes, supporting the emerging concept that transcriptional control and nuclear positioning of TADs are not causally related but independently controlled by the locally associated trans-acting factors.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epigénesis Genética , Histonas/metabolismo , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Histonas/genética , Ratones
16.
Nat Biotechnol ; 32(10): 1019-25, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25129690

RESUMEN

Despite developments in targeted gene sequencing and whole-genome analysis techniques, the robust detection of all genetic variation, including structural variants, in and around genes of interest and in an allele-specific manner remains a challenge. Here we present targeted locus amplification (TLA), a strategy to selectively amplify and sequence entire genes on the basis of the crosslinking of physically proximal sequences. We show that, unlike other targeted re-sequencing methods, TLA works without detailed prior locus information, as one or a few primer pairs are sufficient for sequencing tens to hundreds of kilobases of surrounding DNA. This enables robust detection of single nucleotide variants, structural variants and gene fusions in clinically relevant genes, including BRCA1 and BRCA2, and enables haplotyping. We show that TLA can also be used to uncover insertion sites and sequences of integrated transgenes and viruses. TLA therefore promises to be a useful method in genetic research and diagnostics when comprehensive or allele-specific genetic information is needed.


Asunto(s)
Genómica/métodos , Haplotipos/genética , Modelos Genéticos , Técnicas de Amplificación de Ácido Nucleico/métodos , Análisis de Secuencia de ADN/métodos , Fusión Génica/genética , Genes BRCA1 , Genes BRCA2 , Sitios Genéticos/genética , Humanos , Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA