Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biophys J ; 121(24): 4788-4799, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36325618

RESUMEN

The spatial organization of the eukaryotic genome plays an important role in regulating transcriptional activity. In the nucleus, chromatin forms loops that assemble into fundamental units called topologically associating domains that facilitate or inhibit long-range contacts. These loops are formed and held together by the ring-shaped cohesin protein complex, and this can involve binding of CCCTC-binding factor (CTCF). High-resolution conformation capture experiments provide the frequency at which two DNA fragments physically associate in three-dimensional space. However, technical limitations of this approach, such as low throughput, low resolution, or noise in contact maps, make data interpretation and identification of chromatin intraloop contacts, e.g., between distal regulatory elements and their target genes, challenging. Herein, an existing coarse-grained model of chromatin at single-nucleosome resolution was extended by integrating potentials describing CTCF and cohesin. We performed replica-exchange Monte Carlo simulations with regularly spaced nucleosomes and experimentally determined nucleosome positions in the presence of cohesin-CTCF, as well as depleted systems as controls. In fully extruded loops caused by the presence of cohesin and CTCF, the number of contacts within the formed loops was increased. The number and types of these contacts were impacted by the nucleosome distribution and loop size. Microloops were observed within cohesin-mediated loops due to thermal fluctuations without additional influence of other factors, and the number, size, and shape of microloops were determined by nucleosome distribution and loop size. Nucleosome positions directly affect the spatial structure and contact probability within a loop, with presumed consequences for transcriptional activity.


Asunto(s)
Proteínas de Ciclo Celular , Nucleosomas , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Unión Proteica , Proteínas de Ciclo Celular/metabolismo , Cromatina , Cohesinas
2.
Nat Genet ; 54(2): 202-212, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35145304

RESUMEN

CCCTC-binding factor (CTCF) is critical to three-dimensional genome organization. Upon differentiation, CTCF insulates active and repressed genes within Hox gene clusters. We conducted a genome-wide CRISPR knockout (KO) screen to identify genes required for CTCF-boundary activity at the HoxA cluster, complemented by biochemical approaches. Among the candidates, we identified Myc-associated zinc-finger protein (MAZ) as a cofactor in CTCF insulation. MAZ colocalizes with CTCF at chromatin borders and, similar to CTCF, interacts with the cohesin subunit RAD21. MAZ KO disrupts gene expression and local contacts within topologically associating domains. Similar to CTCF motif deletions, MAZ motif deletions lead to derepression of posterior Hox genes immediately after CTCF boundaries upon differentiation, giving rise to homeotic transformations in mouse. Thus, MAZ is a factor contributing to appropriate insulation, gene expression and genomic architecture during development.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Genes Homeobox , Proteínas de Homeodominio/genética , Factores de Transcripción/metabolismo , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Edición Génica , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Ratones , Factores de Transcripción/química , Factores de Transcripción/genética
3.
Cell Mol Life Sci ; 78(23): 7519-7536, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34657170

RESUMEN

CCCTC-binding factor (CTCF) plays fundamental roles in transcriptional regulation and chromatin architecture maintenance. CTCF is also a tumour suppressor frequently mutated in cancer, however, the structural and functional impact of mutations have not been examined. We performed molecular and structural characterisation of five cancer-specific CTCF missense zinc finger (ZF) mutations occurring within key intra- and inter-ZF residues. Functional characterisation of CTCF ZF mutations revealed a complete (L309P, R339W, R377H) or intermediate (R339Q) abrogation as well as an enhancement (G420D) of the anti-proliferative effects of CTCF. DNA binding at select sites was disrupted and transcriptional regulatory activities abrogated. Molecular docking and molecular dynamics confirmed that mutations in residues specifically contacting DNA bases or backbone exhibited loss of DNA binding. However, R339Q and G420D were stabilised by the formation of new primary DNA bonds, contributing to gain-of-function. Our data confirm that a spectrum of loss-, change- and gain-of-function impacts on CTCF zinc fingers are observed in cell growth regulation and gene regulatory activities. Hence, diverse cellular phenotypes of mutant CTCF are clearly explained by examining structure-function relationships.


Asunto(s)
Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias/patología , Fenotipo , Dedos de Zinc , Apoptosis , Factor de Unión a CCCTC/genética , Proliferación Celular , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
EMBO J ; 40(20): e107795, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34487363

RESUMEN

Somatic mutations in DNA-binding sites for CCCTC-binding factor (CTCF) are significantly elevated in many cancers. Prior analysis has suggested that elevated mutation rates at CTCF-binding sites in skin cancers are a consequence of the CTCF-cohesin complex inhibiting repair of UV damage. Here, we show that CTCF binding modulates the formation of UV damage to induce mutation hot spots. Analysis of genome-wide CPD-seq data in UV-irradiated human cells indicates that formation of UV-induced cyclobutane pyrimidine dimers (CPDs) is primarily suppressed by CTCF binding but elevated at specific locations within the CTCF motif. Locations of CPD hot spots in the CTCF-binding motif coincide with mutation hot spots in melanoma. A similar pattern of damage formation is observed at CTCF-binding sites in vitro, indicating that UV damage modulation is a direct consequence of CTCF binding. We show that CTCF interacts with binding sites containing UV damage and inhibits repair by a model repair enzyme in vitro. Structural analysis and molecular dynamic simulations reveal the molecular mechanism for how CTCF binding modulates CPD formation.


Asunto(s)
Factor de Unión a CCCTC/química , Reparación del ADN , Melanoma/genética , Proteínas Serina-Treonina Quinasas/química , Dímeros de Pirimidina/efectos de la radiación , Neoplasias Cutáneas/genética , Sitios de Unión , Unión Competitiva , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Línea Celular Tumoral , Daño del ADN , Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Dímeros de Pirimidina/biosíntesis , Dímeros de Pirimidina/química , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Rayos Ultravioleta
5.
Dokl Biochem Biophys ; 499(1): 257-259, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34426923

RESUMEN

CTCF belongs to a large family of transcription factors with clusters of C2H2-type zinc finger domains (C2H2 proteins) and is a main architectural protein in mammals. Human CTCF has a homodimerizing unstructured domain at the N-terminus which is involved in long-distance interactions. To test the presence of similar N-terminal domains in other human C2H2 proteins, a yeast two-hybrid system was used. In total, the ability of unstructured N-terminal domains to homodimerize was investigated for six human C2H2 proteins with an expression profile similar to CTCF. The data indicate the lack of the homodimerization ability of these domains. On the other hand, three C2H2 proteins containing the structured domain DUF3669 at the N-terminus demonstrated homo- and heterodimerization activity.


Asunto(s)
Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Dedos de Zinc , Humanos , Dominios Proteicos , Multimerización de Proteína , Estructura Cuaternaria de Proteína
6.
Biochim Biophys Acta Gene Regul Mech ; 1864(10): 194733, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34311130

RESUMEN

CTCF is the most likely ancestor of proteins that contain large clusters of C2H2 zinc finger domains (C2H2) and is conserved among most bilateral organisms. In mammals, CTCF functions as the main architectural protein involved in the organization of topology-associated domains (TADs). In vertebrates and Drosophila, CTCF is involved in the regulation of homeotic genes. Previously, it was found that null mutations in the dCTCF gene died as pharate adults, which failed to eclose from their pupal case, or shortly after hatching of adults. Here, we obtained several new null dCTCF mutations and found that the complete inactivation of dCTCF appears is limited mainly to phenotypic manifestations of the Abd-B gene and fertility of adult flies. Many modifiers that are not associated with an independent phenotypic manifestation can significantly enhance the expressivity of the null dCTCF mutations, indicating that other architectural proteins are able to functionally compensate for dCTCF inactivation in Drosophila. We also mapped the 715-735 aa region of dCTCF as being essential for the interaction with the BTB (Broad-Complex, Tramtrack, and Bric a brac) and microtubule-targeting (M) domains of the CP190 protein, which binds to many architectural proteins. However, the mutational analysis showed that the interaction with CP190 was not important for the functional activity of dCTCF in vivo.


Asunto(s)
Factor de Unión a CCCTC/fisiología , Proteínas de Drosophila/fisiología , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Drosophila/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Infertilidad/genética , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Dominios y Motivos de Interacción de Proteínas
7.
Nat Genet ; 53(7): 1064-1074, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34002095

RESUMEN

Insulators play a critical role in spatiotemporal gene regulation in animals. The evolutionarily conserved CCCTC-binding factor (CTCF) is required for insulator function in mammals, but not all of its binding sites act as insulators. Here we explore the sequence requirements of CTCF-mediated transcriptional insulation using a sensitive insulator reporter in mouse embryonic stem cells. We find that insulation potency depends on the number of CTCF-binding sites in tandem. Furthermore, CTCF-mediated insulation is dependent on upstream flanking sequences at its binding sites. CTCF-binding sites at topologically associating domain boundaries are more likely to function as insulators than those outside topologically associating domain boundaries, independently of binding strength. We demonstrate that insulators form local chromatin domain boundaries and weaken enhancer-promoter contacts. Taken together, our results provide genetic, molecular and structural evidence connecting chromatin topology to the action of insulators in the mammalian genome.


Asunto(s)
Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Animales , Sitios de Unión , Factor de Unión a CCCTC/química , Elementos de Facilitación Genéticos , Humanos , Elementos Aisladores , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Regiones Promotoras Genéticas , Unión Proteica
8.
Cancer Cell ; 39(6): 810-826.e9, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33930311

RESUMEN

STAG2, a cohesin family gene, is among the most recurrently mutated genes in cancer. STAG2 loss of function (LOF) is associated with aggressive behavior in Ewing sarcoma, a childhood cancer driven by aberrant transcription induced by the EWSR1-FLI1 fusion oncogene. Here, using isogenic Ewing cells, we show that, while STAG2 LOF profoundly changes the transcriptome, it does not significantly impact EWSR1-FLI1, CTCF/cohesin, or acetylated H3K27 DNA binding patterns. In contrast, it strongly alters the anchored dynamic loop extrusion process at boundary CTCF sites and dramatically decreases promoter-enhancer interactions, particularly affecting the expression of genes regulated by EWSR1-FLI1 at GGAA microsatellite neo-enhancers. Down-modulation of cis-mediated EWSR1-FLI1 activity, observed in STAG2-LOF conditions, is associated with enhanced migration and invasion properties of Ewing cells previously observed in EWSR1-FLI1low cells. Our study illuminates a process whereby STAG2-LOF fine-tunes the activity of an oncogenic transcription factor through altered CTCF-anchored loop extrusion and cis-mediated enhancer mechanisms.


Asunto(s)
Neoplasias Óseas/genética , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Fusión Oncogénica/genética , Sarcoma de Ewing/genética , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Mutación con Pérdida de Función , Lisina/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/patología , Cohesinas
9.
Dokl Biochem Biophys ; 497(1): 112-115, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33895926

RESUMEN

Human CTCF (hCTCF) is a major architectural protein in mammals. In Drosophila, the CTCF homologue (dCTCF) interacts with the BTB domain of the CP190 protein, which is involved in the establishment of open chromatin and activity of insulators. Previously, it was shown that the BTB protein Kaiso interacts with hCTCF and regulates its activity. We have carried out a detailed study of the interaction between these proteins in the yeast two-hybrid assay. Surprisingly, Kaiso did not interact with hCTCF and its Drosophila homologue. On the other hand, CP190 interacted with the C-terminus of hCTCF. The results obtained demonstrate that the interaction between CTCF and CP190 proteins is highly conserved. It is likely that humans have other BTB proteins that perform the functions described for the Drosophila CP190.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Animales , Factor de Unión a CCCTC/química , Drosophila melanogaster , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos
10.
Biomolecules ; 11(3)2021 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-33673494

RESUMEN

Best known as a transcriptional factor, CCCTC-binding factor (CTCF) is a highly conserved multifunctional DNA-binding protein with 11 zinc fingers. It functions in diverse genomic processes, including transcriptional activation/repression, insulation, genome imprinting and three-dimensional genome organization. A big surprise has recently emerged with the identification of CTCF engaging in the repair of DNA double-strand breaks (DSBs) and in the maintenance of genome fidelity. This discovery now adds a new dimension to the multifaceted attributes of this protein. CTCF facilitates the most accurate DSB repair via homologous recombination (HR) that occurs through an elaborate pathway, which entails a chain of timely assembly/disassembly of various HR-repair complexes and chromatin modifications and coordinates multistep HR processes to faithfully restore the original DNA sequences of broken DNA sites. Understanding the functional crosstalks between CTCF and other HR factors will illuminate the molecular basis of various human diseases that range from developmental disorders to cancer and arise from impaired repair. Such knowledge will also help understand the molecular mechanisms underlying the diverse functions of CTCF in genome biology. In this review, we discuss the recent advances regarding this newly assigned versatile role of CTCF and the mechanism whereby CTCF functions in DSB repair.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Roturas del ADN de Doble Cadena , Animales , Factor de Unión a CCCTC/química , Reparación del ADN/genética , Recombinación Homóloga/genética , Humanos , Modelos Biológicos , Regiones Promotoras Genéticas/genética
11.
Biochemistry ; 60(7): 494-499, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33570402

RESUMEN

The candidate anticancer drug curaxins can insert into DNA base pairs and efficiently inhibit the growth of various cancers. However, how curaxins alter the genomic DNA structure and affect the DNA binding property of key proteins remains to be clarified. Here, we first showed that curaxin CBL0137 strongly stabilizes the interaction between the double strands of DNA and reduces DNA bending and twist rigidity simultaneously, by single-molecule magnetic tweezers. More importantly, we found that CBL0137 greatly impairs the binding of CTCF but facilitates trapping FACT on DNA. We revealed that CBL0137 clamps the DNA double helix that may induce a huge barrier for DNA unzipping during replication and transcription and causes the distinct binding response of CTCF and FACT on DNA. Our work provides a novel mechanical insight into CBL0137's anticancer mechanisms at the nucleic acid level.


Asunto(s)
Carbazoles/farmacología , ADN/efectos de los fármacos , Antineoplásicos/farmacología , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Carbazoles/química , Línea Celular , Línea Celular Tumoral , ADN/metabolismo , Proteínas de Unión al ADN , Humanos , Microscopía de Fuerza Atómica/métodos , Pinzas Ópticas , Unión Proteica , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Brief Bioinform ; 22(1): 589-600, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-32022856

RESUMEN

The CCCTC-binding factor (CTCF) mediates transcriptional regulation and implicates epigenetic modifications in cancers. However, the systematically unveiling inverse regulatory relationship between CTCF and epigenetic modifications still remains unclear, especially the mechanism by which histone modification mediates CTCF binding. Here, we developed a systematic approach to investigate how epigenetic changes affect CTCF binding. Through integration analysis of CTCF binding in 30 cell lines, we concluded that CTCF generally binds with higher intensity in normal cell lines than that in cancers, and higher intensity in genome regions closed to transcription start sites. To facilitate the better understanding of their associations, we constructed linear mixed-effect models to analyze the effects of the epigenetic modifications on CTCF binding in four cancer cell lines and six normal cell lines, and identified seven epigenetic modifications as potential epigenetic patterns that influence CTCF binding intensity in promoter regions and six epigenetic modifications in enhancer regions. Further analysis of the effects in different locations revealed that the epigenetic regulation of CTCF binding was location-specific and cancer cell line-specific. Moreover, H3K4me2 and H3K9ac showed the potential association with immune regulation of disease. Taken together, our method can contribute to improve the understanding of the epigenetic regulation of CTCF binding and provide potential therapeutic targets for treating tumors associated with CTCF.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Epigénesis Genética , Código de Histonas , Factor de Unión a CCCTC/química , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Humanos , Especificidad de Órganos , Unión Proteica
13.
J Comput Biol ; 28(2): 133-145, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33232622

RESUMEN

The three-dimensional (3D) organization of the human genome is of crucial importance for gene regulation, and the CCCTC-binding factor (CTCF) plays an important role in chromatin interactions. However, it is still unclear what sequence patterns in addition to CTCF motif pairs determine chromatin loop formation. To discover the underlying sequence patterns, we have developed a deep learning model, called DeepCTCFLoop, to predict whether a chromatin loop can be formed between a pair of convergent or tandem CTCF motifs using only the DNA sequences of the motifs and their flanking regions. Our results suggest that DeepCTCFLoop can accurately distinguish the CTCF motif pairs forming chromatin loops from the ones not forming loops. It significantly outperforms CTCF-MP, a machine learning model based on word2vec and boosted trees, when using DNA sequences only. Furthermore, we show that DNA motifs binding to several transcription factors, including ZNF384, ZNF263, ASCL1, SP1, and ZEB1, may constitute the complex sequence patterns for CTCF-mediated chromatin loop formation. DeepCTCFLoop has also been applied to disease-associated sequence variants to identify candidates that may disrupt chromatin loop formation. Therefore, our results provide useful information for understanding the mechanism of 3D genome organization and may also help annotate and prioritize the noncoding sequence variants associated with human diseases.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Biología Computacional/métodos , ADN/química , ADN/metabolismo , Sitios de Unión , Factor de Unión a CCCTC/química , Línea Celular , Cromatina/metabolismo , Aprendizaje Profundo , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Células K562 , Motivos de Nucleótidos , Análisis de Secuencia de ADN , Factores de Transcripción/química , Factores de Transcripción/metabolismo
14.
Nat Commun ; 11(1): 5612, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154377

RESUMEN

Current models propose that boundaries of mammalian topologically associating domains (TADs) arise from the ability of the CTCF protein to stop extrusion of chromatin loops by cohesin. While the orientation of CTCF motifs determines which pairs of CTCF sites preferentially stabilize loops, the molecular basis of this polarity remains unclear. By combining ChIP-seq and single molecule live imaging we report that CTCF positions cohesin, but does not control its overall binding dynamics on chromatin. Using an inducible complementation system, we find that CTCF mutants lacking the N-terminus cannot insulate TADs properly. Cohesin remains at CTCF sites in this mutant, albeit with reduced enrichment. Given the orientation of CTCF motifs presents the N-terminus towards cohesin as it translocates from the interior of TADs, these observations explain how the orientation of CTCF binding sites translates into genome folding patterns.


Asunto(s)
Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Cromosomas de los Mamíferos/química , Secuencias de Aminoácidos , Animales , Sitios de Unión , Factor de Unión a CCCTC/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Cricetinae , Drosophila , Ratones , Mutación , Motivos de Nucleótidos , Unión Proteica , Relación Estructura-Actividad , Cohesinas
15.
Nat Commun ; 11(1): 3289, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32620890

RESUMEN

The spatial organization of chromosomes has key functional roles, yet how chromosomes fold remains poorly understood at the single-molecule level. Here, we employ models of polymer physics to investigate DNA loci in human HCT116 and IMR90 wild-type and cohesin depleted cells. Model predictions on single-molecule structures are validated against single-cell imaging data, providing evidence that chromosomal architecture is controlled by a thermodynamics mechanism of polymer phase separation whereby chromatin self-assembles in segregated globules by combinatorial interactions of chromatin factors that include CTCF and cohesin. The thermodynamics degeneracy of single-molecule conformations results in broad structural and temporal variability of TAD-like contact patterns. Globules establish stable environments where specific contacts are highly favored over stochastic encounters. Cohesin depletion reverses phase separation into randomly folded states, erasing average interaction patterns. Overall, globule phase separation appears to be a robust yet reversible mechanism of chromatin organization where stochasticity and specificity coexist.


Asunto(s)
Cromatina/química , Conformación Molecular , Fenómenos Físicos , Polímeros/química , Análisis de la Célula Individual/métodos , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Células HCT116 , Humanos , Unión Proteica , Procesos Estocásticos , Termodinámica , Cohesinas
16.
Epigenetics Chromatin ; 13(1): 30, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680543

RESUMEN

Several thousand sex-differential distal enhancers have been identified in mouse liver; however, their links to sex-biased genes and the impact of any sex-differences in nuclear organization and chromatin interactions are unknown. To address these issues, we first characterized 1847 mouse liver genomic regions showing significant sex differential occupancy by cohesin and CTCF, two key 3D nuclear organizing factors. These sex-differential binding sites were primarily distal to sex-biased genes but rarely generated sex-differential TAD (topologically associating domain) or intra-TAD loop anchors, and were sometimes found in TADs without sex-biased genes. A substantial subset of sex-biased cohesin-non-CTCF binding sites, but not sex-biased cohesin-and-CTCF binding sites, overlapped sex-biased enhancers. Cohesin depletion reduced the expression of male-biased genes with distal, but not proximal, sex-biased enhancers by >10-fold, implicating cohesin in long-range enhancer interactions regulating sex-biased genes. Using circularized chromosome conformation capture-based sequencing (4C-seq), we showed that sex differences in distal sex-biased enhancer-promoter interactions are common. Intra-TAD loops with sex-independent cohesin-and-CTCF anchors conferred sex specificity to chromatin interactions indirectly, by insulating sex-biased enhancer-promoter contacts and by bringing sex-biased genes into closer proximity to sex-biased enhancers. Furthermore, sex-differential chromatin interactions involving sex-biased gene promoters, enhancers, and lncRNAs were associated with sex-biased binding of cohesin and/or CTCF. These studies elucidate how 3D genome organization impacts sex-biased gene expression in a non-reproductive tissue through both direct and indirect effects of cohesin and CTCF looping on distal enhancer interactions with sex-differentially expressed genes.


Asunto(s)
Cromatina/metabolismo , Genoma , Hígado/metabolismo , Sexo , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Elementos de Facilitación Genéticos , Femenino , Masculino , Ratones , Cohesinas
17.
Nat Commun ; 11(1): 2606, 2020 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-32451376

RESUMEN

Nucleoporin proteins (Nups) have been proposed to mediate spatial and temporal chromatin organization during gene regulation. Nevertheless, the molecular mechanisms in mammalian cells are not well understood. Here, we report that Nucleoporin 153 (NUP153) interacts with the chromatin architectural proteins, CTCF and cohesin, and mediates their binding across cis-regulatory elements and TAD boundaries in mouse embryonic stem (ES) cells. NUP153 depletion results in altered CTCF and cohesin binding and differential gene expression - specifically at the bivalent developmental genes. To investigate the molecular mechanism, we utilize epidermal growth factor (EGF)-inducible immediate early genes (IEGs). We find that NUP153 controls CTCF and cohesin binding at the cis-regulatory elements and POL II pausing during the basal state. Furthermore, efficient IEG transcription relies on NUP153. We propose that NUP153 links the nuclear pore complex (NPC) to chromatin architecture allowing genes that are poised to respond rapidly to developmental cues to be properly modulated.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Animales , Factor de Unión a CCCTC/química , Proteínas de Ciclo Celular/química , Línea Celular , Cromatina/química , Cromatina/genética , Proteínas Cromosómicas no Histona/química , Genes Inmediatos-Precoces , Células HeLa , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/deficiencia , Proteínas de Complejo Poro Nuclear/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Polimerasa II/metabolismo , Elementos Reguladores de la Transcripción , Cohesinas
18.
Sci Adv ; 6(8): eaaw4651, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32128389

RESUMEN

Chromatin topological organization is instrumental in gene transcription. Gene-enhancer interactions are accommodated in the same CTCF-mediated insulated neighborhoods. However, it remains poorly understood whether and how the 3D genome architecture is dynamically restructured by external signals. Here, we report that LATS kinases phosphorylated CTCF in the zinc finger (ZF) linkers and disabled its DNA-binding activity. Cellular stress induced LATS nuclear translocation and CTCF ZF linker phosphorylation, and altered the landscape of CTCF genomic binding partly by dissociating it selectively from a small subset of its genomic binding sites. These sites were highly enriched for the boundaries of chromatin domains containing LATS signaling target genes. The stress-induced CTCF phosphorylation and locus-specific dissociation from DNA were LATS-dependent. Loss of CTCF binding disrupted local chromatin domains and down-regulated genes located within them. The study suggests that external signals may rapidly modulate the 3D genome by affecting CTCF genomic binding through ZF linker phosphorylation.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Proteínas Quinasas/metabolismo , Sitios de Unión , Factor de Unión a CCCTC/química , Cromatina/genética , Cromatina/metabolismo , Genómica/métodos , Humanos , Lipoproteínas/metabolismo , Modelos Biológicos , Fosforilación , Unión Proteica , Transducción de Señal , Estrés Fisiológico , Dedos de Zinc
19.
Soft Matter ; 16(9): 2406-2414, 2020 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-32067018

RESUMEN

We use Brownian dynamics simulations to study the formation of chromatin loops through diffusive sliding of slip-link-like proteins, mimicking the behaviour of cohesin molecules. We recently proposed that diffusive sliding is sufficient to explain the extrusion of chromatin loops of hundreds of kilo-base-pairs (kbp), which may then be stabilised by interactions between cohesin and CTCF proteins. Here we show that the flexibility of the chromatin fibre strongly affects this dynamical process, and find that diffusive loop extrusion is more efficient on stiffer chromatin regions. We also show that the dynamics of loop formation are faster in confined and collapsed chromatin conformations but that this enhancement is counteracted by the increased crowding. We provide a simple theoretical argument explaining why stiffness and collapsed conformations favour diffusive extrusion. In light of the heterogeneous physical and conformational properties of eukaryotic chromatin, we suggest that our results are relevant to understand the looping and organisation of interphase chromosomes in vivo.


Asunto(s)
Cromatina/química , Cromosomas/química , Eucariontes/genética , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/genética , Cromosomas/metabolismo , Difusión , Eucariontes/química , Eucariontes/metabolismo , Humanos , Modelos Biológicos , Cohesinas
20.
Epigenetics Chromatin ; 13(1): 2, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31937365

RESUMEN

BACKGROUND: Paternal allele-specific DNA methylation of the H19 imprinting control region (ICR) regulates imprinted expression of the Igf2/H19 genes. The molecular mechanism by which differential methylation of the H19 ICR is established during gametogenesis and maintained after fertilization, however, is not fully understood. We previously showed that a 2.9-kb H19 ICR fragment in transgenic mice was differentially methylated only after fertilization, demonstrating that two separable events, gametic and post-fertilization methylation, occur at the H19 ICR. We then determined that CTCF/Sox-Oct motifs and the 478-bp sequence of the H19 ICR are essential for maintaining its maternal hypomethylation status and for acquisition of paternal methylation, respectively, during the post-fertilization period. RESULTS: Using a series of 5'-truncated H19 ICR transgenes to dissect the 478-bp sequence, we identified a 118-bp region required for post-fertilization methylation activity. Deletion of the sequence from the paternal endogenous H19 ICR caused loss of methylation after fertilization, indicating that methylation activity of the sequence is required to protect endogenous H19 ICR from genome-wide reprogramming. We then reconstructed a synthetic DNA fragment in which the CTCF binding sites, Sox-Oct motifs, as well as the 118-bp sequence, were inserted into lambda DNA, and used it to replace the endogenous H19 ICR. The fragment was methylated during spermatogenesis; moreover, its allele-specific methylation status was faithfully maintained after fertilization, and imprinted expression of the both Igf2 and H19 genes was recapitulated. CONCLUSIONS: Our results identified a 118-bp region within the H19 ICR that is required for de novo DNA methylation of the paternally inherited H19 ICR during pre-implantation period. A lambda DNA-based artificial fragment that contains the 118-bp sequence, in addition to the previously identified cis elements, could fully replace the function of the H19 ICR in the mouse genome.


Asunto(s)
Metilación de ADN , Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , ARN Largo no Codificante/genética , Espermatogonias/metabolismo , Animales , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/genética , Femenino , Fertilización , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Largo no Codificante/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...