Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 153(1): 178-92, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23523135

RESUMEN

The nuclear lamina (NL) interacts with hundreds of large genomic regions termed lamina associated domains (LADs). The dynamics of these interactions and the relation to epigenetic modifications are poorly understood. We visualized the fate of LADs in single cells using a "molecular contact memory" approach. In each nucleus, only ~30% of LADs are positioned at the periphery; these LADs are in intermittent molecular contact with the NL but remain constrained to the periphery. Upon mitosis, LAD positioning is not detectably inherited but instead is stochastically reshuffled. Contact of individual LADs with the NL is linked to transcriptional repression and H3K9 dimethylation in single cells. Furthermore, we identify the H3K9 methyltransferase G9a as a regulator of NL contacts. Collectively, these results highlight principles of the dynamic spatial architecture of chromosomes in relation to gene regulation.


Asunto(s)
Cromosomas/metabolismo , Regulación de la Expresión Génica , Lámina Nuclear/química , Análisis de la Célula Individual/métodos , Adenina/metabolismo , Línea Celular Tumoral , Metilación de ADN , Genoma , Heterocromatina/metabolismo , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Mitosis , Lámina Nuclear/metabolismo
2.
Nature ; 602(7898): 623-631, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35140396

RESUMEN

The mutational landscape is shaped by many processes. Genic regions are vulnerable to mutation but are preferentially protected by transcription-coupled repair1. In microorganisms, transcription has been demonstrated to be mutagenic2,3; however, the impact of transcription-associated mutagenesis remains to be established in higher eukaryotes4. Here we show that ID4-a cancer insertion-deletion (indel) mutation signature of unknown aetiology5 characterized by short (2 to 5 base pair) deletions -is due to a transcription-associated mutagenesis process. We demonstrate that defective ribonucleotide excision repair in mammals is associated with the ID4 signature, with mutations occurring at a TNT sequence motif, implicating topoisomerase 1 (TOP1) activity at sites of genome-embedded ribonucleotides as a mechanistic basis. Such TOP1-mediated deletions occur somatically in cancer, and the ID-TOP1 signature is also found in physiological settings, contributing to genic de novo indel mutations in the germline. Thus, although topoisomerases protect against genome instability by relieving topological stress6, their activity may also be an important source of mutations in the human genome.


Asunto(s)
ADN-Topoisomerasas de Tipo I , Células Germinativas , Mutagénesis , Neoplasias , Animales , Reparación del ADN/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Células Germinativas/metabolismo , Humanos , Mutagénesis/genética , Mutación , Neoplasias/genética , Ribonucleótidos/genética
3.
Genes Dev ; 34(13-14): 931-949, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32439634

RESUMEN

Polycomb group (PcG) proteins silence gene expression by chemically and physically modifying chromatin. A subset of PcG target loci are compacted and cluster in the nucleus; a conformation that is thought to contribute to gene silencing. However, how these interactions influence gross nuclear organization and their relationship with transcription remains poorly understood. Here we examine the role of Polycomb-repressive complex 1 (PRC1) in shaping 3D genome organization in mouse embryonic stem cells (mESCs). Using a combination of imaging and Hi-C analyses, we show that PRC1-mediated long-range interactions are independent of CTCF and can bridge sites at a megabase scale. Impairment of PRC1 enzymatic activity does not directly disrupt these interactions. We demonstrate that PcG targets coalesce in vivo, and that developmentally induced expression of one of the target loci disrupts this spatial arrangement. Finally, we show that transcriptional activation and the loss of PRC1-mediated interactions are separable events. These findings provide important insights into the function of PRC1, while highlighting the complexity of this regulatory system.


Asunto(s)
Núcleo Celular/genética , Genoma/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Animales , Factor de Unión a CCCTC/metabolismo , Embrión de Mamíferos , Ratones , Células Madre Embrionarias de Ratones , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Cell ; 149(5): 1008-22, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22579044

RESUMEN

The presence of ribonucleotides in genomic DNA is undesirable given their increased susceptibility to hydrolysis. Ribonuclease (RNase) H enzymes that recognize and process such embedded ribonucleotides are present in all domains of life. However, in unicellular organisms such as budding yeast, they are not required for viability or even efficient cellular proliferation, while in humans, RNase H2 hypomorphic mutations cause the neuroinflammatory disorder Aicardi-Goutières syndrome. Here, we report that RNase H2 is an essential enzyme in mice, required for embryonic growth from gastrulation onward. RNase H2 null embryos accumulate large numbers of single (or di-) ribonucleotides embedded in their genomic DNA (>1,000,000 per cell), resulting in genome instability and a p53-dependent DNA-damage response. Our findings establish RNase H2 as a key mammalian genome surveillance enzyme required for ribonucleotide removal and demonstrate that ribonucleotides are the most commonly occurring endogenous nucleotide base lesion in replicating cells.


Asunto(s)
Replicación del ADN , Embrión de Mamíferos/metabolismo , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Ribonucleótidos/metabolismo , Animales , Inestabilidad Cromosómica , ADN Polimerasa Dirigida por ADN/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Mol Cell ; 76(3): 473-484.e7, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31494034

RESUMEN

Enhancers can regulate the promoters of their target genes over very large genomic distances. It is widely assumed that mechanisms of enhancer action involve the reorganization of three-dimensional chromatin architecture, but this is poorly understood. The predominant model involves physical enhancer-promoter interaction by looping out the intervening chromatin. However, studying the enhancer-driven activation of the Sonic hedgehog gene (Shh), we have identified a change in chromosome conformation that is incompatible with this simple looping model. Using super-resolution 3D-FISH and chromosome conformation capture, we observe a decreased spatial proximity between Shh and its enhancers during the differentiation of embryonic stem cells to neural progenitors. We show that this can be recapitulated by synthetic enhancer activation, is impeded by chromatin-bound proteins located between the enhancer and the promoter, and appears to involve the catalytic activity of poly (ADP-ribose) polymerase. Our data suggest that models of enhancer-promoter communication need to encompass chromatin conformations other than looping.


Asunto(s)
Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Proteínas Hedgehog/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Neurogénesis , Neuronas/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Ratones , Modelos Genéticos , Neurogénesis/genética , Conformación de Ácido Nucleico , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
6.
PLoS Genet ; 20(5): e1011277, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38781242

RESUMEN

How enhancers regulate their target genes in the context of 3D chromatin organization is extensively studied and models which do not require direct enhancer-promoter contact have recently emerged. Here, we use the activation of estrogen receptor-dependent enhancers in a breast cancer cell line to study enhancer-promoter communication at two loci. This allows high temporal resolution tracking of molecular events from hormone stimulation to efficient gene activation. We examine how both enhancer-promoter spatial proximity assayed by DNA fluorescence in situ hybridization, and contact frequencies resulting from chromatin in situ fragmentation and proximity ligation, change dynamically during enhancer-driven gene activation. These orthogonal methods produce seemingly paradoxical results: upon enhancer activation enhancer-promoter contact frequencies increase while spatial proximity decreases. We explore this apparent discrepancy using different estrogen receptor ligands and transcription inhibitors. Our data demonstrate that enhancer-promoter contact frequencies are transcription independent whereas altered enhancer-promoter proximity depends on transcription. Our results emphasize that the relationship between contact frequencies and physical distance in the nucleus, especially over short genomic distances, is not always a simple one.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Estrógenos , Regiones Promotoras Genéticas , Humanos , Cromatina/genética , Cromatina/metabolismo , Estrógenos/metabolismo , Transcripción Genética , Células MCF-7 , Neoplasias de la Mama/genética , Femenino , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Hibridación Fluorescente in Situ , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Activación Transcripcional , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo
7.
Genome Res ; 33(8): 1269-1283, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37451823

RESUMEN

Contacts between enhancers and promoters are thought to relate to their ability to activate transcription. Investigating factors that contribute to such chromatin interactions is therefore important for understanding gene regulation. Here, we have determined contact frequencies between millions of pairs of cis-regulatory elements from chromosome conformation capture data sets and analyzed a collection of hundreds of DNA-binding factors for binding at regions of enriched contacts. This analysis revealed enriched contacts at sites bound by many factors associated with active transcription. We show that active regulatory elements, independent of cohesin and polycomb, interact with each other across distances of tens of megabases in vertebrate and invertebrate genomes and that interactions correlate and change with activity. However, these ultra-long-range interactions are not dependent on RNA polymerase II transcription or individual transcription cofactors. Using simulations, we show that a model of chromatin and multivalent binding factors can give rise to long-range interactions via bridging-induced clustering. We propose that long-range interactions between cis-regulatory elements are driven by at least three distinct processes: cohesin-mediated loop extrusion, polycomb contacts, and clustering of active regions.


Asunto(s)
Cromatina , Secuencias Reguladoras de Ácidos Nucleicos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Cromatina/genética , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Proteínas del Grupo Polycomb/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Elementos de Facilitación Genéticos , Factor de Unión a CCCTC/metabolismo
8.
Mol Cell ; 72(4): 786-797.e11, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30344096

RESUMEN

Chromatin folded into 3D macromolecular structures is often analyzed by chromosome conformation capture (3C) and fluorescence in situ hybridization (FISH) techniques, but these frequently provide contradictory results. Chromatin can be modeled as a simple polymer composed of a connected chain of units. By embedding data for epigenetic marks (H3K27ac), chromatin accessibility (assay for transposase-accessible chromatin using sequencing [ATAC-seq]), and structural anchors (CCCTC-binding factor [CTCF]), we developed a highly predictive heteromorphic polymer (HiP-HoP) model, where the chromatin fiber varied along its length; combined with diffusing protein bridges and loop extrusion, this model predicted the 3D organization of genomic loci at a population and single-cell level. The model was validated at several gene loci, including the complex Pax6 gene, and was able to determine locus conformations across cell types with varying levels of transcriptional activity and explain different mechanisms of enhancer use. Minimal a priori knowledge of epigenetic marks is sufficient to recapitulate complex genomic loci in 3D and enable predictions of chromatin folding paths.


Asunto(s)
Cromatina/fisiología , Cromosomas/fisiología , Hibridación Fluorescente in Situ/métodos , Animales , Factor de Unión a CCCTC , Línea Celular , Cromatina/genética , Cromosomas/genética , Simulación por Computador , Proteínas de Unión al ADN , Genoma , Genómica/métodos , Humanos , Ratones , Conformación Molecular , Polímeros , Secuencias Reguladoras de Ácidos Nucleicos
9.
Genes Dev ; 30(19): 2173-2186, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27737961

RESUMEN

Chromosomal instability is a hallmark of cancer, but mitotic regulators are rarely mutated in tumors. Mutations in the condensin complexes, which restructure chromosomes to facilitate segregation during mitosis, are significantly enriched in cancer genomes, but experimental evidence implicating condensin dysfunction in tumorigenesis is lacking. We report that mice inheriting missense mutations in a condensin II subunit (Caph2nes) develop T-cell lymphoma. Before tumors develop, we found that the same Caph2 mutation impairs ploidy maintenance to a different extent in different hematopoietic cell types, with ploidy most severely perturbed at the CD4+CD8+ T-cell stage from which tumors initiate. Premalignant CD4+CD8+ T cells show persistent catenations during chromosome segregation, triggering DNA damage in diploid daughter cells and elevated ploidy. Genome sequencing revealed that Caph2 single-mutant tumors are near diploid but carry deletions spanning tumor suppressor genes, whereas P53 inactivation allowed Caph2 mutant cells with whole-chromosome gains and structural rearrangements to form highly aggressive disease. Together, our data challenge the view that mitotic chromosome formation is an invariant process during development and provide evidence that defective mitotic chromosome structure can promote tumorigenesis.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica/genética , Linfoma de Células T/genética , Complejos Multiproteicos/genética , Mutación Missense/genética , Neoplasias del Timo/genética , Adenosina Trifosfatasas/metabolismo , Anafase , Animales , Células Cultivadas , Estructuras Cromosómicas/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Linfoma de Células T/fisiopatología , Masculino , Metafase , Ratones , Complejos Multiproteicos/metabolismo , Timocitos/patología , Neoplasias del Timo/fisiopatología
11.
Genes Dev ; 28(24): 2778-91, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25512564

RESUMEN

Although important for gene regulation, most studies of genome organization use either fluorescence in situ hybridization (FISH) or chromosome conformation capture (3C) methods. FISH directly visualizes the spatial relationship of sequences but is usually applied to a few loci at a time. The frequency at which sequences are ligated together by formaldehyde cross-linking can be measured genome-wide by 3C methods, with higher frequencies thought to reflect shorter distances. FISH and 3C should therefore give the same views of genome organization, but this has not been tested extensively. We investigated the murine HoxD locus with 3C carbon copy (5C) and FISH in different developmental and activity states and in the presence or absence of epigenetic regulators. We identified situations in which the two data sets are concordant but found other conditions under which chromatin topographies extrapolated from 5C or FISH data are not compatible. We suggest that products captured by 3C do not always reflect spatial proximity, with ligation occurring between sequences located hundreds of nanometers apart, influenced by nuclear environment and chromatin composition. We conclude that results obtained at high resolution with either 3C methods or FISH alone must be interpreted with caution and that views about genome organization should be validated by independent methods.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Genoma/genética , Hibridación Fluorescente in Situ/normas , Coloración y Etiquetado/normas , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Técnicas Genéticas/normas , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Mutación , Proteínas del Grupo Polycomb/genética , Estructura Terciaria de Proteína
12.
Neurochem Res ; 45(5): 1168-1179, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32157564

RESUMEN

A number of mutations in a gene encoding RNA-binding protein FUS have been linked to the development of a familial form of amyotrophic lateral sclerosis known as FUS-ALS. C-terminal truncations of FUS by either nonsense or frameshift mutations lead to the development of FUS-ALS with a particularly early onset and fast progression. However, even in patients bearing these highly pathogenic mutations the function of motor neurons is not noticeably compromised for at least a couple of decades, suggesting that until cytoplasmic levels of FUS lacking its C-terminal nuclear localisation signal reaches a critical threshold, motor neurons are able to tolerate its permanent production. In order to identify how the nervous system responds to low levels of pathogenic variants of FUS we produced and characterised a mouse line, L-FUS[1-359], with a low neuronal expression level of a highly aggregation-prone and pathogenic form of C-terminally truncated FUS. In contrast to mice that express substantially higher level of the same FUS variant and develop severe early onset motor neuron pathology, L-FUS[1-359] mice do not develop any clinical or histopathological signs of motor neuron deficiency even at old age. Nevertheless, we detected substantial changes in the spinal cord transcriptome of these mice compared to their wild type littermates. We suggest that at least some of these changes reflect activation of cellular mechanisms compensating for the potentially damaging effect of pathogenic FUS production. Further studies of these mechanism might reveal effective targets for therapy of FUS-ALS and possibly, other forms of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Enfermedades Asintomáticas , Perfilación de la Expresión Génica/métodos , Proteína FUS de Unión a ARN/biosíntesis , Médula Espinal/metabolismo , Transcriptoma/fisiología , Esclerosis Amiotrófica Lateral/genética , Animales , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Proteína FUS de Unión a ARN/genética
13.
Chromosome Res ; 25(3-4): 227-239, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28477268

RESUMEN

In the premature ageing disease Hutchinson-Gilford progeria syndrome (HGPS), the underlying genetic defect in the lamin A gene leads to accumulation at the nuclear lamina of progerin-a mutant form of lamin A that cannot be correctly processed. This has been reported to result in defects in the DNA damage response and in DNA repair, leading to the hypothesis that, as in normal ageing and in other progeroid syndromes caused by mutation of genes of the DNA repair and DNA damage response pathways, increased DNA damage may be responsible for the premature ageing phenotypes in HGPS patients. However, this hypothesis is based upon the study of markers of the DNA damage response, rather than measurement of DNA damage per se or the consequences of unrepaired DNA damage-mutation. Here, using a mutation reporter cell line, we directly compared the inherent and induced mutation rates in cells expressing wild-type lamin A or progerin. We find no evidence for an elevated mutation rate in progerin-expressing cells. We conclude that the cellular defect in HGPS cells does not lie in the repair of DNA damage per se.


Asunto(s)
Expresión Génica , Lamina Tipo A/genética , Tasa de Mutación , Progeria/genética , Animales , Daño del ADN , Modelos Animales de Enfermedad , Heterocromatina/genética , Histonas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Lamina Tipo A/metabolismo , Ratones , Ratones Transgénicos , Mutación , Progeria/metabolismo
14.
Mol Cell ; 38(3): 452-64, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20471950

RESUMEN

How polycomb group proteins repress gene expression in vivo is not known. While histone-modifying activities of the polycomb repressive complexes (PRCs) have been studied extensively, in vitro data have suggested a direct activity of the PRC1 complex in compacting chromatin. Here, we investigate higher-order chromatin compaction of polycomb targets in vivo. We show that PRCs are required to maintain a compact chromatin state at Hox loci in embryonic stem cells (ESCs). There is specific decompaction in the absence of PRC2 or PRC1. This is due to a PRC1-like complex, since decompaction occurs in Ring1B null cells that still have PRC2-mediated H3K27 methylation. Moreover, we show that the ability of Ring1B to restore a compact chromatin state and to repress Hox gene expression is not dependent on its histone ubiquitination activity. We suggest that Ring1B-mediated chromatin compaction acts to directly limit transcription in vivo.


Asunto(s)
Ensamble y Desensamble de Cromatina , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Represoras/metabolismo , Acetilación , Animales , Diferenciación Celular , Línea Celular , Regulación hacia Abajo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Metilación , Ratones , Mutación , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteínas Represoras/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas , Ubiquitinación
15.
Hum Mol Genet ; 22(20): 4180-93, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23760082

RESUMEN

Cornelia de Lange syndrome (CdLS) is a developmental disorder caused by mutations in NIPBL, a protein which has functionally been associated with the cohesin complex. Mutations in core cohesin complex components have also been reported in individuals with CdLS-like phenotypes. In addition to its role in sister chromatid cohesion, cohesin is thought to play a role in regulating gene expression during development. The mechanism of this gene regulation remains unclear, but NIPBL and cohesin have been reported to affect long-range chromosomal interactions, both independently and through interactions with CTCF. We used fluorescence in situ hybridization to investigate whether the disruption of NIPBL affects chromosome architecture. We show that cells from CdLS patients exhibit visible chromatin decompaction, that is most pronounced across gene-rich regions of the genome. Cells carrying mutations predicted to have a more severe effect on NIPBL function show more extensive chromatin decompaction than those carrying milder mutations. This cellular phenotype was reproduced in normal cells depleted for NIPBL with siRNA, but was not seen following the knockdown of either the cohesin component SMC3, or CTCF. We conclude that NIPBL has a function in modulating chromatin architecture, particularly for gene-rich areas of the chromosome, that is not dependent on SMC3/cohesin or CTCF, raising the possibility that the aetiology of disorders associated with the mutation of core cohesin components is distinct from that associated with the disruption of NIPBL itself in classical CdLS.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Cromatina/química , Cromatina/ultraestructura , Proteínas Cromosómicas no Histona/genética , Síndrome de Cornelia de Lange/genética , Proteínas/genética , Proteínas Represoras/genética , Factor de Unión a CCCTC , Línea Celular , Núcleo Celular/ultraestructura , Cromatina/genética , Cromosomas Humanos , Síndrome de Cornelia de Lange/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genoma Humano , Humanos , Hibridación Fluorescente in Situ , Mutación , Fenotipo , Proteínas/metabolismo , Cohesinas
16.
Development ; 139(17): 3157-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22872084

RESUMEN

A late phase of HoxD activation is crucial for the patterning and growth of distal structures across the anterior-posterior (A-P) limb axis of mammals. Polycomb complexes and chromatin compaction have been shown to regulate Hox loci along the main body axis in embryonic development, but the extent to which they have a role in limb-specific HoxD expression, an evolutionary adaptation defined by the activity of distal enhancer elements that drive expression of 5' Hoxd genes, has yet to be fully elucidated. We reveal two levels of chromatin topology that differentiate distal limb A-P HoxD activity. Using both immortalised cell lines derived from posterior and anterior regions of distal E10.5 mouse limb buds, and analysis in E10.5 dissected limb buds themselves, we show that there is a loss of polycomb-catalysed H3K27me3 histone modification and a chromatin decompaction over HoxD in the distal posterior limb compared with anterior. Moreover, we show that the global control region (GCR) long-range enhancer spatially colocalises with the 5' HoxD genomic region specifically in the distal posterior limb. This is consistent with the formation of a chromatin loop between 5' HoxD and the GCR regulatory module at the time and place of distal limb bud development when the GCR participates in initiating Hoxd gene quantitative collinearity and Hoxd13 expression. This is the first example of A-P differences in chromatin compaction and chromatin looping in the development of the mammalian secondary body axis (limb).


Asunto(s)
Tipificación del Cuerpo/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Histonas/metabolismo , Procesamiento de Imagen Asistido por Computador , Hibridación Fluorescente in Situ , Ratones , Microscopía Fluorescente , Proteínas del Grupo Polycomb , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/metabolismo
17.
Proc Natl Acad Sci U S A ; 109(23): 9077-82, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22603795

RESUMEN

Barrett esophagus (BE) is a human metaplastic condition that is the only known precursor to esophageal adenocarcinoma. BE is characterized by a posterior intestinal-like phenotype in an anterior organ and therefore it is reminiscent of homeotic transformations, which can occur in transgenic animal models during embryonic development as a consequence of mutations in HOX genes. In humans, acquired deregulation of HOX genes during adulthood has been linked to carcinogenesis; however, little is known about their role in the pathogenesis of premalignant conditions. We hypothesized that HOX genes may be implicated in the development of BE. We demonstrated that three midcluster HOXB genes (HOXB5, HOXB6, and HOXB7) are overexpressed in BE, compared with the anatomically adjacent normal esophagus and gastric cardia. The midcluster HOXB gene signature in BE is identical to that seen in normal colonic epithelium. Ectopic expression of these three genes in normal squamous esophageal cells in vitro induces markers of intestinal differentiation, such as KRT20, MUC2, and VILLIN. In BE-associated adenocarcinoma, the activation midcluster HOXB gene is associated with loss of H3K27me3 and gain of AcH3, compared with normal esophagus. These changes in histone posttranslational modifications correlate with specific chromatin decompaction at the HOXB locus. We suggest that epigenetically regulated alterations of HOX gene expression can trigger changes in the transcriptional program of adult esophageal cells, with implications for the early stages of carcinogenesis.


Asunto(s)
Adenocarcinoma/metabolismo , Esófago de Barrett/metabolismo , Epigénesis Genética/genética , Neoplasias Esofágicas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Genes Homeobox/genética , Adenocarcinoma/etiología , Adenocarcinoma/genética , Adulto , Esófago de Barrett/complicaciones , Esófago de Barrett/genética , Western Blotting , Inmunoprecipitación de Cromatina , Colon/citología , Colon/metabolismo , Cartilla de ADN/genética , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/genética , Esófago/citología , Esófago/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hibridación Fluorescente in Situ , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa
18.
Brain Tumor Pathol ; 41(1): 4-17, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38097874

RESUMEN

Deletion of CDKN2A occurs in 50% of glioblastomas (GBM), and IFNA locus deletion in 25%. These genes reside closely on chromosome 9. We investigated whether CDKN2A and IFNA were co-deleted within the same heterogeneous tumour and their prognostic implications. We assessed CDKN2A and IFNA14 deletions in 45 glioma samples using an in-house three-colour FISH probe. We examined the correlation between p16INK4a protein expression (via IHC) and CDKN2A deletion along with the impact of these genomic events on patient survival. FISH analyses demonstrated that grades II and III had either wildtype (wt) or amplified CDKN2A/IFNA14, whilst 44% of GBMs harboured homozygous deletions of both genes. Cores with CDKN2A homozygous deletion (n = 11) were negative for p16INK4a. Twenty p16INK4a positive samples lacked CDKN2A deletion with some of cells showing negative p16INK4a. There was heterogeneity in IFNA14/CDKN2A ploidy within each GBM. Survival analyses of primary GBMs suggested a positive association between increased p16INK4a and longer survival; this persisted when considering CDKN2A/IFNA14 status. Furthermore, wt (intact) CDKN2A/IFNA14 were found to be associated with longer survival in recurrent GBMs. Our data suggest that co-deletion of CDKN2A/IFNA14 in GBM negatively correlates with survival and CDKN2A-wt status correlated with longer survival, and with second surgery, itself a marker for improved patient outcomes.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina , Glioblastoma , Humanos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Eliminación de Gen , Glioblastoma/patología , Homocigoto , Eliminación de Secuencia
19.
Genome Res ; 20(5): 554-64, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20212020

RESUMEN

Translocations are known to affect the expression of genes at the breakpoints and, in the case of unbalanced translocations, alter the gene copy number. However, a comprehensive understanding of the functional impact of this class of variation is lacking. Here, we have studied the effect of balanced chromosomal rearrangements on gene expression by comparing the transcriptomes of cell lines from controls and individuals with the t(11;22)(q23;q11) translocation. The number of differentially expressed transcripts between translocation-carrying and control cohorts is significantly higher than that observed between control samples alone, suggesting that balanced rearrangements have a greater effect on gene expression than normal variation. Many of the affected genes are located along the length of the derived chromosome 11. We show that this chromosome is concomitantly altered in its spatial organization, occupying a more central position in the nucleus than its nonrearranged counterpart. Derivative 22-mapping chromosome 22 genes, on the other hand, remain in their usual environment. Our results are consistent with recent studies that experimentally altered nuclear organization, and indicated that nuclear position plays a functional role in regulating the expression of some genes in mammalian cells. Our study suggests that chromosomal translocations can result in hitherto unforeseen, large-scale changes in gene expression that are the consequence of alterations in normal chromosome territory positioning. This has consequences for the patterns of gene expression change seen during tumorigenesis-associated genome instability and during the karyotype changes that lead to speciation.


Asunto(s)
Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 22/genética , Perfilación de la Expresión Génica , Expresión Génica , Translocación Genética , Anomalías Múltiples , Línea Celular Tumoral , Mapeo Cromosómico , Cromosomas Artificiales Bacterianos , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Síndrome
20.
PLoS Genet ; 6(4): e1000922, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20421946

RESUMEN

The spatial organization of the genome in the nucleus plays a role in the regulation of gene expression. Whether co-regulated genes are subject to coordinated repositioning to a shared nuclear space is a matter of considerable interest and debate. We investigated the nuclear organization of estrogen receptor alpha (ERalpha) target genes in human breast epithelial and cancer cell lines, before and after transcriptional activation induced with estradiol. We find that, contrary to another report, the ERalpha target genes TFF1 and GREB1 are distributed in the nucleoplasm with no particular relationship to each other. The nuclear separation between these genes, as well as between the ERalpha target genes PGR and CTSD, was unchanged by hormone addition and transcriptional activation with no evidence for co-localization between alleles. Similarly, while the volume occupied by the chromosomes increased, the relative nuclear position of the respective chromosome territories was unaffected by hormone addition. Our results demonstrate that estradiol-induced ERalpha target genes are not required to co-localize in the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Estrógenos/farmacología , Línea Celular Tumoral , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Factor Trefoil-1 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA