Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Cell ; 169(4): 693-707.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475897

RESUMEN

The spatial organization of chromosomes influences many nuclear processes including gene expression. The cohesin complex shapes the 3D genome by looping together CTCF sites along chromosomes. We show here that chromatin loop size can be increased and that the duration with which cohesin embraces DNA determines the degree to which loops are enlarged. Cohesin's DNA release factor WAPL restricts this loop extension and also prevents looping between incorrectly oriented CTCF sites. We reveal that the SCC2/SCC4 complex promotes the extension of chromatin loops and the formation of topologically associated domains (TADs). Our data support the model that cohesin structures chromosomes through the processive enlargement of loops and that TADs reflect polyclonal collections of loops in the making. Finally, we find that whereas cohesin promotes chromosomal looping, it rather limits nuclear compartmentalization. We conclude that the balanced activity of SCC2/SCC4 and WAPL enables cohesin to correctly structure chromosomes.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Acetiltransferasas/metabolismo , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN , Elongasas de Ácidos Grasos , Edición Génica , Humanos , Complejos Multiproteicos/metabolismo , Proteínas Represoras/metabolismo , Cohesinas
2.
EMBO J ; 42(20): e113150, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37691488

RESUMEN

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


Asunto(s)
Cromatina , Factores de Transcripción SOXB1 , Factores de Transcripción , Sitios de Unión , Cromatina/genética , Regulación de la Expresión Génica , Unión Proteica , Factores de Transcripción/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Ratones
3.
Mol Cell ; 76(5): 724-737.e5, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31629658

RESUMEN

Condensin is a conserved SMC complex that uses its ATPase machinery to structure genomes, but how it does so is largely unknown. We show that condensin's ATPase has a dual role in chromosome condensation. Mutation of one ATPase site impairs condensation, while mutating the second site results in hyperactive condensin that compacts DNA faster than wild-type, both in vivo and in vitro. Whereas one site drives loop formation, the second site is involved in the formation of more stable higher-order Z loop structures. Using hyperactive condensin I, we reveal that condensin II is not intrinsically needed for the shortening of mitotic chromosomes. Condensin II rather is required for a straight chromosomal axis and enables faithful chromosome segregation by counteracting the formation of ultrafine DNA bridges. SMC complexes with distinct roles for each ATPase site likely reflect a universal principle that enables these molecular machines to intricately control chromosome architecture.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Adenosina Trifosfato/química , Sitios de Unión/genética , Sitios de Unión/fisiología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , Cromosomas/fisiología , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Humanos , Complejos Multiproteicos/fisiología , Unión Proteica/fisiología , Subunidades de Proteína/metabolismo , Cohesinas
4.
Nature ; 578(7795): 472-476, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31905366

RESUMEN

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


Asunto(s)
Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Cromatina/química , Cromatina/metabolismo , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Humanos , Ligandos , Modelos Moleculares , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estabilidad Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Cohesinas
5.
EMBO Rep ; 23(2): e53902, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-34927791

RESUMEN

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


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Activación Transcripcional
6.
EMBO Rep ; 23(12): e55782, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36245428

RESUMEN

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


Asunto(s)
Genómica , Heterocromatina , Humanos , Heterocromatina/genética , Antígeno Ki-67/genética
7.
Mol Cell ; 61(3): 461-473, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26833089

RESUMEN

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


Asunto(s)
Núcleo Celular/metabolismo , Segregación Cromosómica , Células Madre Embrionarias/metabolismo , Sitios Genéticos , Operón Lac , Represoras Lac/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Proteína Potenciadora del Homólogo Zeste 2 , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Represoras Lac/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteína Homeótica Nanog , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transfección
8.
Mol Cell ; 60(4): 676-84, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26527277

RESUMEN

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


Asunto(s)
Cromatina/química , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Factor de Unión a CCCTC , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/citología , Ratones , Unión Proteica , Cohesinas
9.
Nucleic Acids Res ; 47(18): 9557-9572, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31372638

RESUMEN

Estrogen receptor α (ERα) is an enhancer activating transcription factor, a key driver of breast cancer and a main target for cancer therapy. ERα-mediated gene regulation requires proper chromatin-conformation to facilitate interactions between ERα-bound enhancers and their target promoters. A major determinant of chromatin structure is the CCCTC-binding factor (CTCF), that dimerizes and together with cohesin stabilizes chromatin loops and forms the boundaries of topologically associated domains. However, whether CTCF-binding elements (CBEs) are essential for ERα-driven cell proliferation is unknown. To address this question in a global manner, we implemented a CRISPR-based functional genetic screen targeting CBEs located in the vicinity of ERα-bound enhancers. We identified four functional CBEs and demonstrated the role of one of them in inducing chromatin conformation changes in favor of activation of PREX1, a key ERα target gene in breast cancer. Indeed, high PREX1 expression is a bona-fide marker of ERα-dependency in cell lines, and is associated with good outcome after anti-hormonal treatment. Altogether, our data show that distinct CTCF-mediated chromatin structures are required for ERα- driven breast cancer cell proliferation.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Unión a CCCTC/genética , Proliferación Celular/genética , Receptor alfa de Estrógeno/genética , Sitios de Unión/genética , Neoplasias de la Mama/patología , Sistemas CRISPR-Cas/genética , Cromatina/genética , Elementos de Facilitación Genéticos/genética , Femenino , Humanos , Células MCF-7 , Unión Proteica/genética
11.
Nucleic Acids Res ; 46(15): e91, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29800273

RESUMEN

It is becoming increasingly clear that chromosome organization plays an important role in gene regulation. High-resolution methods such as 4C, Capture-C and promoter capture Hi-C (PCHiC) enable the study of chromatin loops such as those formed between promoters and enhancers or CTCF/cohesin binding sites. An important aspect of 4C/Capture-C/PCHiC analyses is the reliable identification of chromatin loops, preferably not based on visual inspection of a DNA contact profile, but on reproducible statistical analysis that robustly scores interaction peaks in the non-uniform contact background. Here, we present peakC, an R package for the analysis of 4C/Capture-C/PCHiC data. We generated 4C data for 13 viewpoints in two tissues in at least triplicate to test our methods. We developed a non-parametric peak caller based on rank-products. Sampling analysis shows that not read depth but template quality is the most important determinant of success in 4C experiments. By performing peak calling on single experiments we show that the peak calling results are similar to the replicate experiments, but that false positive rates are significantly reduced by performing replicates. Our software is user-friendly and enables robust peak calling for one-vs-all chromosome capture experiments. peakC is available at: https://github.com/deWitLab/peakC.


Asunto(s)
Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Animales , Sitios de Unión/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Cromatina/metabolismo , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Hígado/embriología , Hígado/metabolismo , Ratones , Reproducibilidad de los Resultados
12.
Nucleic Acids Res ; 46(8): 4213-4227, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29481642

RESUMEN

Oncogene-induced senescence (OIS), provoked in response to oncogenic activation, is considered an important tumor suppressor mechanism. Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nt without a protein-coding capacity. Functional studies showed that deregulated lncRNA expression promote tumorigenesis and metastasis and that lncRNAs may exhibit tumor-suppressive and oncogenic function. Here, we first identified lncRNAs that were differentially expressed between senescent and non-senescent human fibroblast cells. Using RNA interference, we performed a loss-function screen targeting the differentially expressed lncRNAs, and identified lncRNA-OIS1 (lncRNA#32, AC008063.3 or ENSG00000233397) as a lncRNA required for OIS. Knockdown of lncRNA-OIS1 triggered bypass of senescence, higher proliferation rate, lower abundance of the cell-cycle inhibitor CDKN1A and high expression of cell-cycle-associated genes. Subcellular inspection of lncRNA-OIS1 indicated nuclear and cytosolic localization in both normal culture conditions as well as following oncogene induction. Interestingly, silencing lncRNA-OIS1 diminished the senescent-associated induction of a nearby gene (Dipeptidyl Peptidase 4, DPP4) with established role in tumor suppression. Intriguingly, similar to lncRNA-OIS1, silencing DPP4 caused senescence bypass, and ectopic expression of DPP4 in lncRNA-OIS1 knockdown cells restored the senescent phenotype. Thus, our data indicate that lncRNA-OIS1 links oncogenic induction and senescence with the activation of the tumor suppressor DPP4.


Asunto(s)
Senescencia Celular/genética , Dipeptidil Peptidasa 4/genética , ARN Largo no Codificante/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Expresión Génica , Genes ras , Genoma , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/metabolismo
13.
Proc Natl Acad Sci U S A ; 112(40): 12408-13, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26392529

RESUMEN

Microvillus inclusion disease (MVID) is a rare intestinal enteropathy with an onset within a few days to months after birth, resulting in persistent watery diarrhea. Mutations in the myosin Vb gene (MYO5B) have been identified in the majority of MVID patients. However, the exact pathophysiology of MVID still remains unclear. To address the specific role of MYO5B in the intestine, we generated an intestine-specific conditional Myo5b-deficient (Myo5bfl/fl;Vil-CreERT2) mouse model. We analyzed intestinal tissues and cultured organoids of Myo5bfl/fl;Vil-CreERT2 mice by electron microscopy, immunofluorescence, and immunohistochemistry. Our data showed that Myo5bfl/fl;Vil-CreERT2 mice developed severe diarrhea within 4 d after tamoxifen induction. Periodic Acid Schiff and alkaline phosphatase staining revealed subapical accumulation of intracellular vesicles in villus enterocytes. Analysis by electron microscopy confirmed an almost complete absence of apical microvilli, the appearance of microvillus inclusions, and enlarged intercellular spaces in induced Myo5bfl/fl;Vil-CreERT2 intestines. In addition, we determined that MYO5B is involved not only in apical but also basolateral trafficking of proteins. The analysis of the intestine during the early onset of the disease revealed that subapical accumulation of secretory granules precedes occurrence of microvillus inclusions, indicating involvement of MYO5B in early differentiation of epithelial cells. By comparing our data with a novel MVID patient, we conclude that our mouse model completely recapitulates the intestinal phenotype of human MVID. This includes severe diarrhea, loss of microvilli, occurrence of microvillus inclusions, and subapical secretory granules. Thus, loss of MYO5B disturbs both apical and basolateral trafficking of proteins and causes MVID in mice.


Asunto(s)
Síndromes de Malabsorción/metabolismo , Microvellosidades/patología , Mucolipidosis/metabolismo , Miosina Tipo V/metabolismo , Animales , Modelos Animales de Enfermedad , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/ultraestructura , Femenino , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/ultraestructura , Síndromes de Malabsorción/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Mucolipidosis/inducido químicamente , Miosina Tipo V/genética , Técnicas de Cultivo de Órganos , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Tamoxifeno
14.
Nature ; 476(7360): 293-7, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21727895

RESUMEN

The adult stem cell marker Lgr5 and its relative Lgr4 are often co-expressed in Wnt-driven proliferative compartments. We find that conditional deletion of both genes in the mouse gut impairs Wnt target gene expression and results in the rapid demise of intestinal crypts, thus phenocopying Wnt pathway inhibition. Mass spectrometry demonstrates that Lgr4 and Lgr5 associate with the Frizzled/Lrp Wnt receptor complex. Each of the four R-spondins, secreted Wnt pathway agonists, can bind to Lgr4, -5 and -6. In HEK293 cells, RSPO1 enhances canonical WNT signals initiated by WNT3A. Removal of LGR4 does not affect WNT3A signalling, but abrogates the RSPO1-mediated signal enhancement, a phenomenon rescued by re-expression of LGR4, -5 or -6. Genetic deletion of Lgr4/5 in mouse intestinal crypt cultures phenocopies withdrawal of Rspo1 and can be rescued by Wnt pathway activation. Lgr5 homologues are facultative Wnt receptor components that mediate Wnt signal enhancement by soluble R-spondin proteins. These results will guide future studies towards the application of R-spondins for regenerative purposes of tissues expressing Lgr5 homologues.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Células Madre Adultas/metabolismo , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptores Frizzled/metabolismo , Eliminación de Gen , Células HEK293 , Humanos , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Regeneración , Transducción de Señal/genética , Proteínas Wnt/genética , Proteína Wnt3 , Proteína Wnt3A
15.
Gastroenterology ; 146(4): 1040-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24333384

RESUMEN

BACKGROUND & AIMS: The contribution of genetic factors to the pathogenesis of inflammatory bowel disease (IBD) has been established by twin, targeted-sequencing, and genome-wide association studies. These studies identified many risk loci, and research is underway to identify causal variants. These studies have focused mainly on protein-coding genes. We investigated other functional elements in the human genome, such as regulatory regions. METHODS: Using acetylated histone 3 lysine 27 chromatin immunoprecipitation and sequencing, we identified tens of thousands of potential regulatory regions that are active in intestinal epithelium (primary intestinal crypts and cultured organoids) isolated from resected material and from biopsies collected during ileo-colonoscopies and immune cells (monocytes, macrophages, CD34(+), CD4(+), and CD8(+)). We correlated these regions with susceptibility loci for IBD. RESULTS: We have generated acetylated histone 3 lysine 27 profiles from primary intestinal epithelium and cultured organoids, which we have made publically available. We found that 45 of 163 single nucleotide polymorphisms (SNPs) associated with IBD overlap specifically with active regulatory elements. In addition, by taking strong linkage disequilibrium into account, another 47 IBD-associated SNPs colocalized with active regulatory elements through other SNPs in their vicinity. Altogether, 92 of 163 IBD-associated SNPs correlated with distinct active regulatory elements-a frequency 2.5- to 3.5-fold greater than that expected from random sampling. The variations in these SNPs often create or disrupt known binding motifs; they might affect the binding of transcriptional regulators to alter expression of regulated genes. CONCLUSIONS: In addition to variants in protein coding genes, variants in noncoding DNA regulatory regions that are active in intestinal epithelium and immune cells are potentially involved in the pathogenesis of IBD.


Asunto(s)
Sitios Genéticos , Inmunidad Mucosa/genética , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/inmunología , Secuencias Reguladoras de Ácidos Nucleicos , Acetilación , Animales , Animales Modificados Genéticamente , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Histonas/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Desequilibrio de Ligamiento , Fenotipo , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Factores de Riesgo , Análisis de Secuencia de ADN , Pez Cebra/genética
16.
Nat Genet ; 38(6): 694-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16628213

RESUMEN

Polycomb group (PcG) proteins maintain transcriptional repression of developmentally important genes and have been implicated in cell proliferation and stem cell self-renewal. We used a genome-wide approach to map binding patterns of PcG proteins (Pc, esc and Sce) in Drosophila melanogaster Kc cells. We found that Pc associates with large genomic regions of up to approximately 150 kb in size, hereafter referred to as 'Pc domains'. Sce and esc accompany Pc in most of these domains. PcG-bound chromatin is trimethylated at histone H3 Lys27 and is generally transcriptionally silent. Furthermore, PcG proteins preferentially bind to developmental genes. Many of these encode transcriptional regulators and key components of signal transduction pathways, including Wingless, Hedgehog, Notch and Delta. We also identify several new putative functions of PcG proteins, such as in steroid hormone biosynthesis. These results highlight the extensive involvement of PcG proteins in the coordination of development through the formation of large repressive chromatin domains.


Asunto(s)
Cromatina/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Genoma , Isoformas de Proteínas/genética , Animales , Línea Celular , ADN Complementario , N-Metiltransferasa de Histona-Lisina , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas
17.
PLoS Genet ; 7(3): e1001343, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21455484

RESUMEN

Polycomb group (PcG) proteins bind and regulate hundreds of genes. Previous evidence has suggested that long-range chromatin interactions may contribute to the regulation of PcG target genes. Here, we adapted the Chromosome Conformation Capture on Chip (4C) assay to systematically map chromosomal interactions in Drosophila melanogaster larval brain tissue. Our results demonstrate that PcG target genes interact extensively with each other in nuclear space. These interactions are highly specific for PcG target genes, because non-target genes with either low or high expression show distinct interactions. Notably, interactions are mostly limited to genes on the same chromosome arm, and we demonstrate that a topological rather than a sequence-based mechanism is responsible for this constraint. Our results demonstrate that many interactions among PcG target genes exist and that these interactions are guided by overall chromosome architecture.


Asunto(s)
Cromosomas/química , Cromosomas/metabolismo , Drosophila melanogaster/genética , Proteínas Represoras/metabolismo , Animales , Encéfalo/metabolismo , Cromatina/metabolismo , Cromosomas de Insectos/química , Cromosomas de Insectos/genética , Biología Computacional , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Homeobox/genética , Histonas/metabolismo , Larva , Proteínas del Grupo Polycomb , Unión Proteica , Proteínas Represoras/química
18.
PLoS Biol ; 8(11): e1000539, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21103407

RESUMEN

Wnt signaling maintains the undifferentiated state of intestinal crypt progenitor cells by inducing the formation of nuclear TCF4/ß-catenin complexes. In colorectal cancer, activating mutations in Wnt pathway components cause inappropriate activation of TCF4/ß-catenin-driven transcription. Despite the passage of a decade after the discovery of TCF4 and ß-catenin as the molecular effectors of the Wnt signal, few transcriptional activators essential and unique to the regulation of this transcription program have been found. Using proteomics, we identified the leukemia-associated Mllt10/Af10 and the methyltransferase Dot1l as Tcf4/ß-catenin interactors in mouse small intestinal crypts. Mllt10/Af10-Dot1l, essential for transcription elongation, are recruited to Wnt target genes in a ß-catenin-dependent manner, resulting in H3K79 methylation over their coding regions in vivo in proliferative crypts of mouse small intestine in colorectal cancer and Wnt-inducible HEK293T cells. Depletion of MLLT10/AF10 in colorectal cancer and Wnt-inducible HEK293T cells followed by expression array analysis identifies MLLT10/AF10 and DOT1L as essential activators to a large extent dedicated to Wnt target gene regulation. In contrast, previously published ß-catenin coactivators p300 and BRG1 displayed a more pleiotropic target gene expression profile controlling Wnt and other pathways. tcf4, mllt10/af10, and dot1l are co-expressed in Wnt-driven tissues in zebrafish and essential for Wnt-reporter activity. Intestinal differentiation defects in apc-mutant zebrafish can be rescued by depletion of Mllt10 and Dot1l, establishing these genes as activators downstream of Apc in Wnt target gene activation in vivo. Morpholino-depletion of mllt10/af10-dot1l in zebrafish results in defects in intestinal homeostasis and a significant reduction in the in vivo expression of direct Wnt target genes and in the number of proliferative intestinal epithelial cells. We conclude that Mllt10/Af10-Dot1l are essential, largely dedicated activators of Wnt-dependent transcription, critical for maintenance of intestinal proliferation and homeostasis. The methyltransferase DOT1L may present an attractive candidate for drug targeting in colorectal cancer.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Homeostasis/fisiología , Intestino Delgado/fisiología , Leucemia/fisiopatología , Metiltransferasas/fisiología , Factores de Transcripción/fisiología , beta Catenina/metabolismo , Animales , Línea Celular , Proliferación Celular , Metilación de ADN , N-Metiltransferasa de Histona-Lisina , Humanos , Intestino Delgado/patología , Metiltransferasas/genética , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción 4 , Factores de Transcripción/genética , Transcripción Genética , Pez Cebra/genética
19.
Nat Commun ; 14(1): 7762, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040699

RESUMEN

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


Asunto(s)
Tumor Rabdoide , Humanos , Niño , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Proteína SMARCB1/genética , Factores de Transcripción/genética , Mutación , Regiones Promotoras Genéticas/genética , Carcinogénesis/genética
20.
Cell Rep ; 42(10): 113124, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37733591

RESUMEN

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


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias , Humanos , Resistencia a Antineoplásicos/genética , Paclitaxel/farmacología , Resistencia a Múltiples Medicamentos/genética , Neoplasias/genética , Metilación de ADN/genética , Línea Celular Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA