Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(5): 1179-1181, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445037

RESUMEN

In this issue, Kazachenka, Bertozzi, and colleagues identify elements in the mouse genome with epigenetic variability between littermates, a phenomenon linked to transmission of phenotypes over generations. This addresses two questions that remained unanswered despite intense speculation: how prevalent are these alleles, and what is their effect, within and across generations?


Asunto(s)
Epigénesis Genética , Mamíferos/genética , Alelos , Animales , Epigenómica , Ratones , Fenotipo
2.
Nature ; 619(7969): 385-393, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37407816

RESUMEN

The basic helix-loop-helix (bHLH) family of transcription factors recognizes DNA motifs known as E-boxes (CANNTG) and includes 108 members1. Here we investigate how chromatinized E-boxes are engaged by two structurally diverse bHLH proteins: the proto-oncogene MYC-MAX and the circadian transcription factor CLOCK-BMAL1 (refs. 2,3). Both transcription factors bind to E-boxes preferentially near the nucleosomal entry-exit sites. Structural studies with engineered or native nucleosome sequences show that MYC-MAX or CLOCK-BMAL1 triggers the release of DNA from histones to gain access. Atop the H2A-H2B acidic patch4, the CLOCK-BMAL1 Per-Arnt-Sim (PAS) dimerization domains engage the histone octamer disc. Binding of tandem E-boxes5-7 at endogenous DNA sequences occurs through direct interactions between two CLOCK-BMAL1 protomers and histones and is important for circadian cycling. At internal E-boxes, the MYC-MAX leucine zipper can also interact with histones H2B and H3, and its binding is indirectly enhanced by OCT4 elsewhere on the nucleosome. The nucleosomal E-box position and the type of bHLH dimerization domain jointly determine the histone contact, the affinity and the degree of competition and cooperativity with other nucleosome-bound factors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , ADN , Histonas , Factores de Transcripción ARNTL/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN/genética , ADN/metabolismo , Secuencias Hélice-Asa-Hélice/genética , Histonas/química , Histonas/metabolismo , Nucleosomas/química , Nucleosomas/genética , Nucleosomas/metabolismo , Unión Proteica , Proteínas CLOCK/química , Proteínas CLOCK/metabolismo , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/metabolismo , Regulación Alostérica , Leucina Zippers , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Multimerización de Proteína
3.
Mol Cell ; 81(2): 255-267.e6, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33290745

RESUMEN

Gene activation requires the cooperative activity of multiple transcription factors at cis-regulatory elements (CREs). Yet, most transcription factors have short residence time, questioning the requirement of their physical co-occupancy on DNA to achieve cooperativity. Here, we present a DNA footprinting method that detects individual molecular interactions of transcription factors and nucleosomes with DNA in vivo. We apply this strategy to quantify the simultaneous binding of multiple transcription factors on single DNA molecules at mouse CREs. Analysis of the binary occupancy patterns at thousands of motif combinations reveals that high DNA co-occupancy occurs for most types of transcription factors, in the absence of direct physical interaction, at sites of competition with nucleosomes. Perturbation of pairwise interactions demonstrates the function of molecular co-occupancy in binding cooperativity. Our results reveal the interactions regulating CREs at molecular resolution and identify DNA co-occupancy as a widespread cooperativity mechanism used by transcription factors to remodel chromatin.


Asunto(s)
Huella de ADN/métodos , ADN/genética , Nucleosomas/química , Elementos Reguladores de la Transcripción , Factores de Transcripción/genética , Animales , Sitios de Unión , ADN/química , ADN/metabolismo , Masculino , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Nucleosomas/metabolismo , Unión Proteica , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Transcripción Genética
4.
Nat Rev Genet ; 23(12): 728-740, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35831531

RESUMEN

Cell type-specific gene expression relies on transcription factors (TFs) binding DNA sequence motifs embedded in chromatin. Understanding how motifs are accessed in chromatin is crucial to comprehend differential transcriptional responses and the phenotypic impact of sequence variation. Chromatin obstacles to TF binding range from DNA methylation to restriction of DNA access by nucleosomes depending on their position, composition and modification. In vivo and in vitro approaches now enable the study of TF binding in chromatin at unprecedented resolution. Emerging insights suggest that TFs vary in their ability to navigate chromatin states. However, it remains challenging to link binding and transcriptional outcomes to molecular characteristics of TFs or the local chromatin substrate. Here, we discuss our current understanding of how TFs access DNA in chromatin and novel techniques and directions towards a better understanding of this critical step in genome regulation.


Asunto(s)
Cromatina , Factores de Transcripción , Cromatina/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sitios de Unión/genética , Nucleosomas/genética , Unión Proteica , ADN/genética
5.
Cell ; 153(2): 480-92, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582333

RESUMEN

To gain insight into the cellular readout of DNA methylation, we established a strategy for systematically profiling the genome-wide distribution of chromatin-interacting factors. This enabled us to create genomic maps for the methyl-CpG-binding domain (MBD) family of proteins, including disease-relevant mutants, deletions, and isoforms. In vivo binding of MBD proteins occurs predominantly as a linear function of local methylation density, requiring functional MBD domains and methyl-CPGs. This interaction directs specificity of MBD proteins to methylated, CpG-dense, and inactive regulatory regions. In contrast, binding to unmethylated sites varies between MBD proteins and is mediated via alternative domains or protein-protein interactions. Such targeting is exemplified by NuRD-complex-mediated tethering of MBD2 to a subset of unmethylated, active regulatory regions. Interestingly, MBD3 also occupies these sites, but like MBD2, binding is independent of the presence of hydroxymethylation. These functional binding maps reveal methylation-dependent and -independent binding modes and revise current models of DNA methylation readout through MBD proteins.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Secuencia de Aminoácidos , Animales , Biotina/metabolismo , Cromatina/metabolismo , Islas de CpG , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células Madre Embrionarias , Estudio de Asociación del Genoma Completo , Humanos , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína , Alineación de Secuencia
6.
Nature ; 596(7870): 133-137, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34234345

RESUMEN

The majority of gene transcripts generated by RNA polymerase II in mammalian genomes initiate at CpG island (CGI) promoters1,2, yet our understanding of their regulation remains limited. This is in part due to the incomplete information that we have on transcription factors, their DNA-binding motifs and which genomic binding sites are functional in any given cell type3-5. In addition, there are orphan motifs without known binders, such as the CGCG element, which is associated with highly expressed genes across human tissues and enriched near the transcription start site of a subset of CGI promoters6-8. Here we combine single-molecule footprinting with interaction proteomics to identify BTG3-associated nuclear protein (BANP) as the transcription factor that binds this element in the mouse and human genome. We show that BANP is a strong CGI activator that controls essential metabolic genes in pluripotent stem and terminally differentiated neuronal cells. BANP binding is repelled by DNA methylation of its motif in vitro and in vivo, which epigenetically restricts most binding to CGIs and accounts for differential binding at aberrantly methylated CGI promoters in cancer cells. Upon binding to an unmethylated motif, BANP opens chromatin and phases nucleosomes. These findings establish BANP as a critical activator of a set of essential genes and suggest a model in which the activity of CGI promoters relies on methylation-sensitive transcription factors that are capable of chromatin opening.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Cromatina/genética , Cromatina/metabolismo , Islas de CpG/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Cromatina/química , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN , Regulación de la Expresión Génica , Genes Esenciales , Humanos , Ratones , Imagen Individual de Molécula
7.
Immunity ; 44(3): 527-541, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26982363

RESUMEN

Lymphopoiesis requires the activation of lineage-specific genes embedded in naive, inaccessible chromatin or in primed, accessible chromatin. The mechanisms responsible for de novo gain of chromatin accessibility, known as "pioneer" function, remain poorly defined. Here, we showed that the EBF1 C-terminal domain (CTD) is required for the regulation of a specific gene set involved in B cell fate decision and differentiation, independently of activation and repression functions. Using genome-wide analysis of DNaseI hypersensitivity and DNA methylation in multipotent Ebf1(-/-) progenitors and derivative EBF1wt- or EBF1ΔC-expressing cells, we found that the CTD promoted chromatin accessibility and DNA demethylation in previously naive chromatin. The CTD allowed EBF1 to bind at inaccessible genomic regions that offer limited co-occupancy by other transcription factors, whereas the CTD was dispensable for EBF1 binding at regions that are occupied by multiple transcription factors. Thus, the CTD enables EBF1 to confer permissive lineage-specific changes in progenitor chromatin landscape.


Asunto(s)
Linfocitos B/fisiología , Cromatina/metabolismo , Células Progenitoras Linfoides/fisiología , Transactivadores/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Metilación de ADN/genética , Redes Reguladoras de Genes/genética , Linfopoyesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Estructura Terciaria de Proteína/genética , Transactivadores/genética
8.
Cell ; 143(2): 183-4, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20946976

RESUMEN

Chromatin consists of DNA and a large number of associated proteins. Filion et al. (2010) provide a genome-wide analysis of the location of 53 chromatin proteins in Drosophila, revealing important principles underlying chromatin regulation and providing colorful insights into their organization.

9.
Cell ; 141(4): 618-31, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20478254

RESUMEN

Adaptation to different levels of illumination is central to the function of the retina. Here, we demonstrate that levels of the miR-183/96/182 cluster, miR-204, and miR-211 are regulated by different light levels in the mouse retina. Concentrations of these microRNAs were downregulated during dark adaptation and upregulated in light-adapted retinas, with rapid decay and increased transcription being responsible for the respective changes. We identified the voltage-dependent glutamate transporter Slc1a1 as one of the miR-183/96/182 targets in photoreceptor cells. We found that microRNAs in retinal neurons decay much faster than microRNAs in nonneuronal cells. The high turnover is also characteristic of microRNAs in hippocampal and cortical neurons, and neurons differentiated from ES cells in vitro. Blocking activity reduced turnover of microRNAs in neuronal cells while stimulation with glutamate accelerated it. Our results demonstrate that microRNA metabolism in neurons is higher than in most other cells types and linked to neuronal activity.


Asunto(s)
MicroARNs/metabolismo , Neuronas/metabolismo , Animales , Adaptación a la Oscuridad , Regulación hacia Abajo , Células Madre Embrionarias , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ratones , Células Fotorreceptoras de Vertebrados/metabolismo , Neuronas Retinianas/metabolismo , Regulación hacia Arriba
10.
Nature ; 569(7754): 136-140, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30996347

RESUMEN

Chromatin remodelling complexes evict, slide, insert or replace nucleosomes, which represent an intrinsic barrier for access to DNA. These remodellers function in most aspects of genome utilization including transcription-factor binding, DNA replication and repair1,2. Although they are frequently mutated in cancer3, it remains largely unclear how the four mammalian remodeller families (SWI/SNF, ISWI, CHD and INO80) orchestrate the global organization of nucleosomes. Here we generated viable embryonic stem cells that lack SNF2H, the ATPase of ISWI complexes, enabling study of SNF2H cellular function, and contrast it to BRG1, the ATPase of SWI/SNF. Loss of SNF2H decreases nucleosomal phasing and increases linker lengths, providing in vivo evidence for an ISWI function in ruling nucleosomal spacing in mammals. Systematic analysis of transcription-factor binding reveals that these remodelling activities have specific effects on binding of different transcription factors. One group critically depends on BRG1 and contains the transcriptional repressor REST, whereas a non-overlapping set of transcription factors, including the insulator protein CTCF, relies on SNF2H. This selectivity readily explains why chromosomal folding and insulation of topologically associated domains requires SNF2H, but not BRG1. Collectively, this study shows that mammalian ISWI is critical for nucleosomal periodicity and nuclear organization and that transcription factors rely on specific remodelling pathways for correct genomic binding.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/genética , Animales , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/metabolismo , Células Madre Embrionarias/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Unión Proteica
11.
Nature ; 571(7764): E6, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31239520

RESUMEN

In this Article, in Fig. 1a, the 5' and 3' labels were reversed in the DNA sequence, and Fig. 4 was missing panel labels a-e. These errors have been corrected online.

12.
Nature ; 571(7763): 79-84, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31142837

RESUMEN

Access to DNA packaged in nucleosomes is critical for gene regulation, DNA replication and DNA repair. In humans, the UV-damaged DNA-binding protein (UV-DDB) complex detects UV-light-induced pyrimidine dimers throughout the genome; however, it remains unknown how these lesions are recognized in chromatin, in which nucleosomes restrict access to DNA. Here we report cryo-electron microscopy structures of UV-DDB bound to nucleosomes bearing a 6-4 pyrimidine-pyrimidone dimer or a DNA-damage mimic in various positions. We find that UV-DDB binds UV-damaged nucleosomes at lesions located in the solvent-facing minor groove without affecting the overall nucleosome architecture. In the case of buried lesions that face the histone core, UV-DDB changes the predominant translational register of the nucleosome and selectively binds the lesion in an accessible, exposed position. Our findings explain how UV-DDB detects occluded lesions in strongly positioned nucleosomes, and identify slide-assisted site exposure as a mechanism by which high-affinity DNA-binding proteins can access otherwise occluded sites in nucleosomal DNA.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , ADN/ultraestructura , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Dímeros de Pirimidina/análisis , Microscopía por Crioelectrón , ADN/química , ADN/efectos de la radiación , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/ultraestructura , Histonas/química , Histonas/metabolismo , Histonas/ultraestructura , Humanos , Modelos Moleculares , Nucleosomas/genética , Nucleosomas/efectos de la radiación , Dímeros de Pirimidina/química , Dímeros de Pirimidina/genética , Termodinámica , Rayos Ultravioleta/efectos adversos
13.
Mol Cell ; 67(3): 411-422.e4, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28735898

RESUMEN

Transcription initiation entails chromatin opening followed by pre-initiation complex formation and RNA polymerase II recruitment. Subsequent polymerase elongation requires additional signals, resulting in increased residence time downstream of the start site, a phenomenon referred to as pausing. Here, we harnessed single-molecule footprinting to quantify distinct steps of initiation in vivo throughout the Drosophila genome. This identifies the impact of promoter structure on initiation dynamics in relation to nucleosomal occupancy. Additionally, perturbation of transcriptional initiation reveals an unexpectedly high turnover of polymerases at paused promoters-an observation confirmed at the level of nascent RNAs. These observations argue that absence of elongation is largely caused by premature termination rather than by stable polymerase stalling. In support of this non-processive model, we observe that induction of the paused heat shock promoter depends on continuous initiation. Our study provides a framework to quantify protein binding at single-molecule resolution and refines concepts of transcriptional pausing.


Asunto(s)
ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , ARN/biosíntesis , Imagen Individual de Molécula , Transcripción Genética , Animales , Sitios de Unión , ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Estudio de Asociación del Genoma Completo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Semivida , Cinética , Unión Proteica , Estabilidad Proteica , Proteolisis , ARN/genética , ARN Polimerasa II/genética , TATA Box , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética , Terminación de la Transcripción Genética
14.
Bioinformatics ; 38(9): 2624-2625, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35199152

RESUMEN

SUMMARY: Proteins binding to specific nucleotide sequences, such as transcription factors, play key roles in the regulation of gene expression. Their binding can be indirectly observed via associated changes in transcription, chromatin accessibility, DNA methylation and histone modifications. Identifying candidate factors that are responsible for these observed experimental changes is critical to understand the underlying biological processes. Here, we present monaLisa, an R/Bioconductor package that implements approaches to identify relevant transcription factors from experimental data. The package can be easily integrated with other Bioconductor packages and enables seamless motif analyses without any software dependencies outside of R. AVAILABILITY AND IMPLEMENTATION: monaLisa is implemented in R and available on Bioconductor at https://bioconductor.org/packages/monaLisa with the development version hosted on GitHub at https://github.com/fmicompbio/monaLisa. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Programas Informáticos , Factores de Transcripción
15.
Genome Res ; 29(4): 554-563, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30709850

RESUMEN

Most mammalian RNA polymerase II initiation events occur at CpG islands, which are rich in CpGs and devoid of DNA methylation. Despite their relevance for gene regulation, it is unknown to what extent the CpG dinucleotide itself actually contributes to promoter activity. To address this question, we determined the transcriptional activity of a large number of chromosomally integrated promoter constructs and monitored binding of transcription factors assumed to play a role in CpG island activity. This revealed that CpG density significantly improves motif-based prediction of transcription factor binding. Our experiments also show that high CpG density alone is insufficient for transcriptional activity, yet results in increased transcriptional output when combined with particular transcription factor motifs. However, this CpG contribution to promoter activity is independent of DNA methyltransferase activity. Together, this refines our understanding of mammalian promoter regulation as it shows that high CpG density within CpG islands directly contributes to an environment permissive for full transcriptional activity.


Asunto(s)
Islas de CpG , Metilación de ADN , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Línea Celular , Células Cultivadas , Ratones , Unión Proteica , Factores de Transcripción/metabolismo
16.
Genes Dev ; 28(6): 652-64, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24637118

RESUMEN

The mammalian intestinal epithelium has a unique organization in which crypts harboring stem cells produce progenitors and finally clonal populations of differentiated cells. Remarkably, the epithelium is replaced every 3-5 d throughout adult life. Disrupted maintenance of the intricate balance of proliferation and differentiation leads to loss of epithelial integrity or barrier function or to cancer. There is a tight correlation between the epigenetic status of genes and expression changes during differentiation; however, the mechanism of how changes in DNA methylation direct gene expression and the progression from stem cells to their differentiated descendants is unclear. Using conditional gene ablation of the maintenance methyltransferase Dnmt1, we demonstrate that reducing DNA methylation causes intestinal crypt expansion in vivo. Determination of the base-resolution DNA methylome in intestinal stem cells and their differentiated descendants shows that DNA methylation is dynamic at enhancers, which are often associated with genes important for both stem cell maintenance and differentiation. We establish that the loss of DNA methylation at intestinal stem cell gene enhancers causes inappropriate gene expression and delayed differentiation.


Asunto(s)
Diferenciación Celular , Metilación de ADN , Intestino Delgado/citología , Células Madre/citología , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Ratones
17.
Nature ; 517(7534): 321-6, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25592537

RESUMEN

Cytosine methylation is a DNA modification generally associated with transcriptional silencing. Factors that regulate methylation have been linked to human disease, yet how they contribute to malignances remains largely unknown. Genomic maps of DNA methylation have revealed unexpected dynamics at gene regulatory regions, including active demethylation by TET proteins at binding sites for transcription factors. These observations indicate that the underlying DNA sequence largely accounts for local patterns of methylation. As a result, this mark is highly informative when studying gene regulation in normal and diseased cells, and it can potentially function as a biomarker. Although these findings challenge the view that methylation is generally instructive for gene silencing, several open questions remain, including how methylation is targeted and recognized and in what context it affects genome readout.


Asunto(s)
Metilación de ADN , Animales , Islas de CpG/genética , Citosina/química , Citosina/metabolismo , Enfermedad , Genoma/genética , Humanos , Invertebrados/genética , Factores de Transcripción/metabolismo , Vertebrados/genética
18.
Nature ; 528(7583): 575-9, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26675734

RESUMEN

Eukaryotic transcription factors (TFs) are key determinants of gene activity, yet they bind only a fraction of their corresponding DNA sequence motifs in any given cell type. Chromatin has the potential to restrict accessibility of binding sites; however, in which context chromatin states are instructive for TF binding remains mainly unknown. To explore the contribution of DNA methylation to constrained TF binding, we mapped DNase-I-hypersensitive sites in murine stem cells in the presence and absence of DNA methylation. Methylation-restricted sites are enriched for TF motifs containing CpGs, especially for those of NRF1. In fact, the TF NRF1 occupies several thousand additional sites in the unmethylated genome, resulting in increased transcription. Restoring de novo methyltransferase activity initiates remethylation at these sites and outcompetes NRF1 binding. This suggests that binding of DNA-methylation-sensitive TFs relies on additional determinants to induce local hypomethylation. In support of this model, removal of neighbouring motifs in cis or of a TF in trans causes local hypermethylation and subsequent loss of NRF1 binding. This competition between DNA methylation and TFs in vivo reveals a case of cooperativity between TFs that acts indirectly via DNA methylation. Methylation removal by methylation-insensitive factors enables occupancy of methylation-sensitive factors, a principle that rationalizes hypomethylation of regulatory regions.


Asunto(s)
Unión Competitiva , Cromatina/metabolismo , Metilación de ADN , Factor Nuclear 1 de Respiración/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Cromatina/química , Cromatina/genética , Desoxirribonucleasa I/metabolismo , Genoma/genética , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica
19.
Nature ; 520(7546): 243-7, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25607372

RESUMEN

DNA methylation is an epigenetic modification associated with transcriptional repression of promoters and is essential for mammalian development. Establishment of DNA methylation is mediated by the de novo DNA methyltransferases DNMT3A and DNMT3B, whereas DNMT1 ensures maintenance of methylation through replication. Absence of these enzymes is lethal, and somatic mutations in these genes have been associated with several human diseases. How genomic DNA methylation patterns are regulated remains poorly understood, as the mechanisms that guide recruitment and activity of DNMTs in vivo are largely unknown. To gain insights into this matter we determined genomic binding and site-specific activity of the mammalian de novo DNA methyltransferases DNMT3A and DNMT3B. We show that both enzymes localize to methylated, CpG-dense regions in mouse stem cells, yet are excluded from active promoters and enhancers. By specifically measuring sites of de novo methylation, we observe that enzymatic activity reflects binding. De novo methylation increases with CpG density, yet is excluded from nucleosomes. Notably, we observed selective binding of DNMT3B to the bodies of transcribed genes, which leads to their preferential methylation. This targeting to transcribed sequences requires SETD2-mediated methylation of lysine 36 on histone H3 and a functional PWWP domain of DNMT3B. Together these findings reveal how sequence and chromatin cues guide de novo methyltransferase activity to ensure methylome integrity.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Epigénesis Genética/genética , Genoma/genética , Animales , Línea Celular , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/química , ADN Metiltransferasa 3A , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos/genética , Genómica , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Transcripción Genética/genética , ADN Metiltransferasa 3B
20.
PLoS Genet ; 13(12): e1007102, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29267285

RESUMEN

Genomic location can inform on potential function and recruitment signals for chromatin-associated proteins. High mobility group (Hmg) proteins are of similar size as histones with Hmga1 and Hmga2 being particularly abundant in replicating normal tissues and in cancerous cells. While several roles for Hmga proteins have been proposed we lack a comprehensive description of their genomic location as a function of chromatin, DNA sequence and functional domains. Here we report such a characterization in mouse embryonic stem cells in which we introduce biotin-tagged constructs of wild-type and DNA-binding domain mutants. Comparative analysis of the genome-wide distribution of Hmga proteins reveals pervasive binding, a feature that critically depends on a functional DNA-binding domain and which is shared by both Hmga proteins. Assessment of the underlying queues instructive for this binding modality identifies AT richness, defined as high frequency of A or T bases, as the major criterion for local binding. Additionally, we show that other chromatin states such as those linked to cis-regulatory regions have little impact on Hmga binding both in stem and differentiated cells. As a consequence, Hmga proteins are preferentially found at AT-rich regions such as constitutively heterochromatic regions but are absent from enhancers and promoters arguing for a limited role in regulating individual genes. In line with this model, we show that genetic deletion of Hmga proteins in stem cells causes limited transcriptional effects and that binding is conserved in neuronal progenitors. Overall our comparative study describing the in vivo binding modality of Hmga1 and Hmga2 identifies the proteins' preference for AT-rich DNA genome-wide and argues against a suggested function of Hmga at regulatory regions. Instead we discover pervasive binding with enrichment at regions of higher AT content irrespective of local variation in chromatin modifications.


Asunto(s)
Secuencia Rica en At , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Animales , Composición de Base , Secuencia de Bases , Cromatina/genética , Cromatina/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Histonas/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA