Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 173(5): 1150-1164.e14, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29706544

RESUMEN

Tandem repeats (TRs) are generated by DNA replication errors and retain a high level of instability, which in principle would make them unsuitable for integration into gene regulatory networks. However, the appearance of DNA sequence motifs recognized by transcription factors may turn TRs into functional cis-regulatory elements, thus favoring their stabilization in genomes. Here, we show that, in human cells, the transcriptional repressor ZEB1, which promotes the maintenance of mesenchymal features largely by suppressing epithelial genes and microRNAs, occupies TRs harboring dozens of copies of its DNA-binding motif within genomic loci relevant for maintenance of epithelial identity. The deletion of one such TR caused quasi-mesenchymal cancer cells to reacquire epithelial features, partially recapitulating the effects of ZEB1 gene deletion. These data demonstrate that the high density of identical motifs in TRs can make them suitable platforms for recruitment of transcriptional repressors, thus promoting their exaptation into pre-existing cis-regulatory networks.


Asunto(s)
Secuencias Repetidas en Tándem/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Adulto , Animales , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Femenino , Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Mucosa Bucal/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica , Factores de Transcripción/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/deficiencia , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
2.
Mol Cell ; 84(5): 967-980.e10, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38242130

RESUMEN

Histone-modifying enzymes depend on the availability of cofactors, with acetyl-coenzyme A (CoA) being required for histone acetyltransferase (HAT) activity. The discovery that mitochondrial acyl-CoA-producing enzymes translocate to the nucleus suggests that high concentrations of locally synthesized metabolites may impact acylation of histones and other nuclear substrates, thereby controlling gene expression. Here, we show that 2-ketoacid dehydrogenases are stably associated with the Mediator complex, thus providing a local supply of acetyl-CoA and increasing the generation of hyper-acetylated histone tails. Nitric oxide (NO), which is produced in large amounts in lipopolysaccharide-stimulated macrophages, inhibited the activity of Mediator-associated 2-ketoacid dehydrogenases. Elevation of NO levels and the disruption of Mediator complex integrity both affected de novo histone acetylation within a shared set of genomic regions. Our findings indicate that the local supply of acetyl-CoA generated by 2-ketoacid dehydrogenases bound to Mediator is required to maximize acetylation of histone tails at sites of elevated HAT activity.


Asunto(s)
Histonas , Óxido Nítrico , Histonas/genética , Histonas/metabolismo , Acetilcoenzima A/metabolismo , Acetilación , Óxido Nítrico/metabolismo , Complejo Mediador/metabolismo , Oxidorreductasas/metabolismo
3.
Genes Dev ; 37(21-24): 1017-1040, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38092518

RESUMEN

Transcription termination pathways mitigate the detrimental consequences of unscheduled promiscuous initiation occurring at hundreds of thousands of genomic cis-regulatory elements. The Restrictor complex, composed of the Pol II-interacting protein WDR82 and the RNA-binding protein ZC3H4, suppresses processive transcription at thousands of extragenic sites in mammalian genomes. Restrictor-driven termination does not involve nascent RNA cleavage, and its interplay with other termination machineries is unclear. Here we show that efficient termination at Restrictor-controlled extragenic transcription units involves the recruitment of the protein phosphatase 1 (PP1) regulatory subunit PNUTS, a negative regulator of the SPT5 elongation factor, and Symplekin, a protein associated with RNA cleavage complexes but also involved in cleavage-independent and phosphatase-dependent termination of noncoding RNAs in yeast. PNUTS and Symplekin act synergistically with, but independently from, Restrictor to dampen processive extragenic transcription. Moreover, the presence of limiting nuclear levels of Symplekin imposes a competition for its recruitment among multiple transcription termination machineries, resulting in mutual regulatory interactions. Hence, by synergizing with Restrictor, Symplekin and PNUTS enable efficient termination of processive, long-range extragenic transcription.


Asunto(s)
ARN Polimerasa II , Transcripción Genética , Animales , ARN Polimerasa II/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas de Unión al ARN/metabolismo , Procesamiento Proteico-Postraduccional , Mamíferos/genética
4.
Genes Dev ; 36(7-8): 414-432, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35361678

RESUMEN

Six methyltransferases divide labor in establishing genomic profiles of histone H3 lysine 9 methylation (H3K9me), an epigenomic modification controlling constitutive heterochromatin, gene repression, and silencing of retroelements. Among them, SETDB1 is recruited to active chromatin domains to silence the expression of endogenous retroviruses. In the context of experiments aimed at determining the impact of SETDB1 on stimulus-inducible gene expression in macrophages, we found that loss of H3K9me3 caused by SETDB1 depletion was associated with increased recruitment of CTCF to >1600 DNA binding motifs contained within SINE B2 repeats, a previously unidentified target of SETDB1-mediated repression. CTCF is an essential regulator of chromatin folding that restrains DNA looping by cohesin, thus creating boundaries among adjacent topological domains. Increased CTCF binding to SINE B2 repeats enhanced insulation at hundreds of sites and increased loop formation within topological domains containing lipopolysaccharide-inducible genes, which correlated with their impaired regulation in response to stimulation. These data indicate a role of H3K9me3 in restraining genomic distribution and activity of CTCF, with an impact on chromatin organization and gene regulation.


Asunto(s)
Cromatina , Silenciador del Gen , Heterocromatina , Metilación , Retroelementos
5.
Cell ; 152(1-2): 157-71, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23332752

RESUMEN

According to current models, once the cell has reached terminal differentiation, the enhancer repertoire is completely established and maintained by cooperatively acting lineage-specific transcription factors (TFs). TFs activated by extracellular stimuli operate within this predetermined repertoire, landing close to where master regulators are constitutively bound. Here, we describe latent enhancers, defined as regions of the genome that in terminally differentiated cells are unbound by TFs and lack the histone marks characteristic of enhancers but acquire these features in response to stimulation. Macrophage stimulation caused sequential binding of stimulus-activated and lineage-determining TFs to these regions, enabling deposition of enhancer marks. Once unveiled, many of these enhancers did not return to a latent state when stimulation ceased; instead, they persisted and mediated a faster and stronger response upon restimulation. We suggest that stimulus-specific expansion of the cis-regulatory repertoire provides an epigenomic memory of the exposure to environmental agents.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Macrófagos/metabolismo , Animales , Diferenciación Celular , Epigenómica , Código de Histonas , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
6.
Genes Dev ; 31(4): 399-412, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28275002

RESUMEN

Enhancers and promoters that control the transcriptional output of terminally differentiated cells include cell type-specific and broadly active housekeeping elements. Whether the high constitutive activity of these two groups of cis-regulatory elements relies on entirely distinct or instead also on shared regulators is unknown. By dissecting the cis-regulatory repertoire of macrophages, we found that the ELF subfamily of ETS proteins selectively bound within 60 base pairs (bp) from the transcription start sites of highly active housekeeping genes. ELFs also bound constitutively active, but not poised, macrophage-specific enhancers and promoters. The role of ELFs in promoting high-level constitutive transcription was suggested by multiple evidence: ELF sites enabled robust transcriptional activation by endogenous and minimal synthetic promoters, ELF recruitment was stabilized by the transcriptional machinery, and ELF proteins mediated recruitment of transcriptional and chromatin regulators to core promoters. These data suggest that the co-optation of a limited number of highly active transcription factors represents a broadly adopted strategy to equip both cell type-specific and housekeeping cis-regulatory elements with the ability to efficiently promote transcription.


Asunto(s)
Regulación de la Expresión Génica/genética , Genes Esenciales/genética , Macrófagos/fisiología , Factores de Transcripción/metabolismo , Animales , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica , Transporte de Proteínas , Factores de Transcripción/genética
8.
EMBO J ; 38(20): e102161, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31531882

RESUMEN

Differentiation of normal and tumor cells is controlled by regulatory networks enforced by lineage-determining transcription factors (TFs). Among them, TFs such as FOXA1/2 bind naïve chromatin and induce its accessibility, thus establishing new gene regulatory networks. Pancreatic ductal adenocarcinoma (PDAC) is characterized by the coexistence of well- and poorly differentiated cells at all stages of disease. How the transcriptional networks determining such massive cellular heterogeneity are established remains to be determined. We found that FOXA2, a TF controlling pancreas specification, broadly contributed to the cis-regulatory networks of PDACs. Despite being expressed in both well- and poorly differentiated PDAC cells, FOXA2 displayed extensively different genomic distributions and controlled distinct gene expression programs. Grade-specific functions of FOXA2 depended on its partnership with TFs whose expression varied depending on the differentiation grade. These data suggest that FOXA2 contributes to the regulatory networks of heterogeneous PDAC cells via interactions with alternative partner TFs.


Asunto(s)
Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias Pancreáticas/patología , Elementos Reguladores de la Transcripción , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Movimiento Celular , Proliferación Celular , Redes Reguladoras de Genes , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas de Homeodominio/genética , Humanos , Clasificación del Tumor , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Células Tumorales Cultivadas
9.
Mol Cell ; 60(3): 460-74, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593720

RESUMEN

Upon recruitment to active enhancers and promoters, RNA polymerase II (Pol II) generates short non-coding transcripts of unclear function. The mechanisms that control the length and the amount of ncRNAs generated by cis-regulatory elements are largely unknown. Here, we show that the adaptor protein WDR82 and its associated complexes actively limit such non-coding transcription. WDR82 targets the SET1 H3K4 methyltransferases and the nuclear protein phosphatase 1 (PP1) complexes to the initiating Pol II. WDR82 and PP1 also interact with components of the transcriptional termination and RNA processing machineries. Depletion of WDR82, SET1, or the PP1 subunit required for its nuclear import caused distinct but overlapping transcription termination defects at highly expressed genes and active enhancers and promoters, thus enabling the increased synthesis of unusually long ncRNAs. These data indicate that transcription initiated from cis-regulatory elements is tightly coordinated with termination mechanisms that impose the synthesis of short RNAs.


Asunto(s)
Núcleo Celular/metabolismo , Elementos de Facilitación Genéticos/fisiología , Regiones Promotoras Genéticas/fisiología , ARN Polimerasa II/metabolismo , ARN no Traducido/biosíntesis , Terminación de la Transcripción Genética/fisiología , Transporte Activo de Núcleo Celular/fisiología , Animales , Núcleo Celular/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Ratones , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , ARN Polimerasa II/genética , ARN no Traducido/genética
10.
Genes Dev ; 29(4): 394-408, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25637355

RESUMEN

The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8-PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF-AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome. While constitutively expressed IRF8-dependent genes contained only sites mediating basal IRF8/PU.1 recruitment, inducible IRF8-dependent genes contained variable combinations of constitutive and inducible sites. Overall, these data show at the genome scale how the same TF can be linked to constitutive and inducible gene regulation via distinct combinations of alternative DNA-binding sites.


Asunto(s)
Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Macrófagos/metabolismo , Animales , Células Cultivadas , Factores Reguladores del Interferón/genética , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Unión Proteica
11.
Immunity ; 36(4): 572-85, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22483804

RESUMEN

Histone methyltransferases catalyze site-specific deposition of methyl groups, enabling recruitment of transcriptional regulators. In mammals, trimethylation of lysine 4 in histone H3, a modification localized at the transcription start sites of active genes, is catalyzed by six enzymes (SET1a and SET1b, MLL1-MLL4) whose specific functions are largely unknown. By using a genomic approach, we found that in macrophages, MLL4 (also known as Wbp7) was required for the expression of Pigp, an essential component of the GPI-GlcNAc transferase, the enzyme catalyzing the first step of glycosylphosphatidylinositol (GPI) anchor synthesis. Impaired Pigp expression in Wbp7(-/-) macrophages abolished GPI anchor-dependent loading of proteins on the cell membrane. Consistently, loss of GPI-anchored CD14, the coreceptor for lipopolysaccharide (LPS) and other bacterial molecules, markedly attenuated LPS-triggered intracellular signals and gene expression changes. These data link a histone-modifying enzyme to a biosynthetic pathway and indicate a specialized biological role for Wbp7 in macrophage function and antimicrobial response.


Asunto(s)
Glicosilfosfatidilinositoles/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Glicosilfosfatidilinositoles/biosíntesis , Hexosiltransferasas/biosíntesis , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Receptores de Lipopolisacáridos/biosíntesis , Lipopolisacáridos/inmunología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/biosíntesis , Proteína de la Leucemia Mieloide-Linfoide/genética , Transducción de Señal
12.
EMBO J ; 35(6): 595-617, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26769127

RESUMEN

The histological grade of carcinomas describes the ability of tumor cells to organize in differentiated epithelial structures and has prognostic and therapeutic impact. Here, we show that differential usage of the genomic repertoire of transcriptional enhancers leads to grade-specific gene expression programs in human pancreatic ductal adenocarcinoma (PDAC). By integrating gene expression profiling, epigenomic footprinting, and loss-of-function experiments in PDAC cell lines of different grade, we identified the repertoires of enhancers specific to high- and low-grade PDACs and the cognate set of transcription factors acting to maintain their activity. Among the candidate regulators of PDAC differentiation, KLF5 was selectively expressed in pre-neoplastic lesions and low-grade primary PDACs and cell lines, where it maintained the acetylation of grade-specific enhancers, the expression of epithelial genes such as keratins and mucins, and the ability to organize glandular epithelia in xenografts. The identification of the transcription factors controlling differentiation in PDACs will help clarify the molecular bases of its heterogeneity and progression.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Regulación de la Expresión Génica , Línea Celular Tumoral , Epigénesis Genética , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
13.
J Immunol ; 200(7): 2439-2454, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29500242

RESUMEN

The enzymes of the poly-ADP-ribose polymerase (PARP) superfamily control many relevant cellular processes, but a precise understanding of their activities in different physiological or disease contexts is largely incomplete. We found that transcription of several Parp genes was dynamically regulated upon murine macrophage activation by endotoxin. PARP14 was strongly induced by several inflammatory stimuli and translocated into the nucleus of stimulated cells. Quantitative mass spectrometry analysis showed that PARP14 bound to a group of IFN-stimulated gene (ISG)-encoded proteins, most with an unknown function, and it was required for their nuclear accumulation. Moreover, PARP14 depletion attenuated transcription of primary antiviral response genes regulated by the IFN regulatory transcription factor 3, including Ifnb1, thus reducing IFN-ß production and activation of ISGs involved in the secondary antiviral response. In agreement with the above-mentioned data, PARP14 hindered Salmonella typhimurium proliferation in murine macrophages. Overall, these data hint at a role of PARP14 in the control of antimicrobial responses and specifically in nuclear activities of a subgroup of ISG-encoded proteins.


Asunto(s)
Interferón beta/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Poli(ADP-Ribosa) Polimerasas/genética , Salmonella typhimurium/inmunología , Transporte Activo de Núcleo Celular/fisiología , Animales , Sistemas CRISPR-Cas , Línea Celular , Endotoxinas/inmunología , Edición Génica , Activación de Macrófagos/genética , Macrófagos/microbiología , Ratones , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/genética , Salmonella typhimurium/crecimiento & desarrollo
14.
Proc Natl Acad Sci U S A ; 109(42): E2865-74, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22802645

RESUMEN

Histone deacetylases (HDACs) regulate inflammatory gene expression, as indicated by the potent antiinflammatory activity of pan-HDAC inhibitors. However, the specific contribution of each of the 11 HDAC proteins to the inflammatory gene expression program is unknown. Using an integrated genomic approach, we found that Hdac3-deficient macrophages were unable to activate almost half of the inflammatory gene expression program when stimulated with LPS. A large part of the activation defect was attributable to loss of basal and LPS-inducible expression of IFN-ß, which maintains Stat1 protein levels in unstimulated cells and acts in an autocrine/paracrine manner after stimulation to promote a secondary wave of Stat1-dependent gene expression. Loss of Hdac3-mediated repression of nuclear receptors led to hyperacetylation of thousands of genomic sites and associated gene derepression. The up-regulation of the constitutively expressed prostaglandin endoperoxide synthase, Ptgs1 (Cox-1), a nuclear receptor target, had a causative role in the phenotype because its chemical inhibition reverted, albeit partially, the Ifn-ß activation defect. These data indicate a central role for Hdac3 in inflammation and may have relevance for the use of selective Hdac inhibitors as antiinflammatory agents.


Asunto(s)
Regulación de la Expresión Génica/genética , Histona Desacetilasas/metabolismo , Macrófagos/metabolismo , Animales , Secuencia de Bases , Inmunoprecipitación de Cromatina , Ciclooxigenasa 1/metabolismo , Citocinas/análisis , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Genómica , Histona Desacetilasas/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
15.
Nat Struct Mol Biol ; 28(4): 337-346, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33767452

RESUMEN

Interactions between the splicing machinery and RNA polymerase II increase protein-coding gene transcription. Similarly, exons and splicing signals of enhancer-generated long noncoding RNAs (elncRNAs) augment enhancer activity. However, elncRNAs are inefficiently spliced, suggesting that, compared with protein-coding genes, they contain qualitatively different exons with a limited ability to drive splicing. We show here that the inefficiently spliced first exons of elncRNAs as well as promoter-antisense long noncoding RNAs (pa-lncRNAs) in human and mouse cells trigger a transcription termination checkpoint that requires WDR82, an RNA polymerase II-binding protein, and its RNA-binding partner of previously unknown function, ZC3H4. We propose that the first exons of elncRNAs and pa-lncRNAs are an intrinsic component of a regulatory mechanism that, on the one hand, maximizes the activity of these cis-regulatory elements by recruiting the splicing machinery and, on the other, contains elements that suppress pervasive extragenic transcription.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , ARN Polimerasa II/ultraestructura , ARN Largo no Codificante/genética , Transcripción Genética , Empalme Alternativo/genética , Animales , Proteínas Cromosómicas no Histona/ultraestructura , Proteínas de Unión al ADN/ultraestructura , Exones/genética , Humanos , Ratones , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/genética , Empalme del ARN/genética , ARN sin Sentido/genética , ARN sin Sentido/ultraestructura , ARN Largo no Codificante/ultraestructura , ARN Mensajero/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
16.
Dev Cell ; 55(4): 398-412.e7, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32997974

RESUMEN

Many tumors of endodermal origin are composed of highly secretory cancer cells that must adapt endoplasmic reticulum (ER) activity to enable proper folding of secreted proteins and prevent ER stress. We found that pancreatic ductal adenocarcinomas (PDACs) overexpress the myelin regulatory factor (MYRF), an ER membrane-associated transcription factor (TF) released by self-cleavage. MYRF was expressed in the well-differentiated secretory cancer cells, but not in the poorly differentiated quasi-mesenchymal cells that coexist in the same tumor. MYRF expression was controlled by the epithelial identity TF HNF1B, and it acted to fine-tune the expression of genes encoding highly glycosylated, cysteine-rich secretory proteins, thus preventing ER overload. MYRF-deficient PDAC cells showed signs of ER stress, impaired proliferation, and an inability to form spheroids in vitro, while in vivo they generated highly secretory but poorly proliferating and hypocellular tumors. These data indicate a role of MYRF in the control of ER homeostasis in highly secretory PDAC cells.


Asunto(s)
Retículo Endoplásmico/metabolismo , Homeostasis , Proteínas de la Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , Cromatina/metabolismo , ADN de Neoplasias/metabolismo , Retículo Endoplásmico/ultraestructura , Estrés del Retículo Endoplásmico/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Clasificación del Tumor , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/ultraestructura , Unión Proteica , Factores de Transcripción/genética
17.
Cancer Res ; 69(21): 8491-8, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19843847

RESUMEN

The E2F and MYC transcription factors are critical regulators of cell proliferation and contribute to the development of human cancers. Here, we report on the identification of a novel E2F target gene, ATAD2, the predicted protein product of which contains both a bromodomain and an ATPase domain. The pRB-E2F pathway regulates ATAD2 expression, which is limiting for the entry into the S phase of the cell cycle. We show that ATAD2 binds the MYC oncogene and stimulates its transcriptional activity. ATAD2 maps to chromosome 8q24, 4.3 Mb distal to MYC, in a region that is frequently found amplified in cancer. Consistent with this, we show that ATAD2 expression is high in several human tumors and that the expression levels correlate with clinical outcome of breast cancer patients. We suggest that ATAD2 links the E2F and MYC pathways and contributes to the development of aggressive cancer through the enhancement of MYC-dependent transcription.


Asunto(s)
Cromosomas Humanos Par 8/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas , Western Blotting , Ciclo Celular , Proliferación Celular , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Histonas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Metástasis de la Neoplasia , Neoplasias/patología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/farmacología , Proteína de Retinoblastoma/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas
18.
PLoS One ; 3(8): e3034, 2008 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-18716661

RESUMEN

Patterns of methylation at lysine 4 and 27 of histone H3 have been associated with states of gene activation and repression that are developmentally regulated and are thought to underlie the establishment of lineage specific gene expression programs. Recent studies have provided fundamental insight into the problem of lineage specification by comparing global changes in chromatin and transcription between ES and neural stem (NS) cells, points respectively of departure and arrival for neural commitment. With these maps of the differentiated state in place, a central task is now to unravel the chromatin dynamics that enables these differentiation transitions. In particular, the observation that lineage-specific genes repressed in ES cells by Polycomb-mediated H3-K27 trimethylation (H3-K27me3) are demethylated and derepressed in differentiated cells posited the existence of a specific H3-K27 demethylase.In order to gain insight into the epigenetic transitions that enable lineage specification, we investigated the early stages of neural commitment using as model system the monolayer differentiation of mouse ES cells into neural stem (NS) cells. Starting from a comprehensive profiling of JmjC-domain genes, we report here that Jmjd3, recently identified as a H3-K27me3 specific demethylase, controls the expression of key regulators and markers of neurogenesis and is required for commitment to the neural lineage.Our results demonstrate the relevance of an enzymatic activity that antagonizes Polycomb regulation and highlight different modalities through which the dynamics of H3-K27me3 is related to transcriptional output. By showing that the H3-K27 demethylase Jmjd3 is required for commitment to the neural lineage and that it resolves the bivalent domain at the Nestin promoter, our work confirms the functional relevance of bivalent domain resolution that had been posited on the basis of the genome-wide correlation between their controlled resolution and differentiation. In addition, our data indicate that the regulation of H3-K27me3 is highly gene- and context- specific, suggesting that the interplay of methyltransferases and demethylases enables the fine-tuning more than the on/off alternation of methylation states.


Asunto(s)
Células Madre Embrionarias/citología , Neuronas/citología , Neuronas/fisiología , Oxidorreductasas N-Desmetilantes/metabolismo , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Supervivencia Celular , Genes Reporteros , Histona Demetilasas con Dominio de Jumonji , Luciferasas/genética , Ratones , Oxidorreductasas N-Desmetilantes/genética , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Cell ; 130(6): 1083-94, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17825402

RESUMEN

Epigenetic chromatin marks restrict the ability of differentiated cells to change gene expression programs in response to environmental cues and to transdifferentiate. Polycomb group (PcG) proteins mediate gene silencing and repress transdifferentiation in a manner dependent on histone H3 lysine 27 trimethylation (H3K27me3). However, macrophages migrated into inflamed tissues can transdifferentiate, but it is unknown whether inflammation alters PcG-dependent silencing. Here we show that the JmjC-domain protein Jmjd3 is a H3K27me demethylase expressed in macrophages in response to bacterial products and inflammatory cytokines. Jmjd3 binds PcG target genes and regulates their H3K27me3 levels and transcriptional activity. The discovery of an inducible enzyme that erases a histone mark controlling differentiation and cell identity provides a link between inflammation and reprogramming of the epigenome, which could be the basis for macrophage plasticity and might explain the differentiation abnormalities in chronic inflammation.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Silenciador del Gen , Histonas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN , Remoción de Radical Alquila , Inducción Enzimática , Femenino , Silenciador del Gen/efectos de los fármacos , Histona Demetilasas , Proteínas de Homeodominio/metabolismo , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Histona Demetilasas con Dominio de Jumonji , Lipopolisacáridos/farmacología , Lisina/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/patología , Ratones , Datos de Secuencia Molecular , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidorreductasas N-Desmetilantes/biosíntesis , Oxidorreductasas N-Desmetilantes/genética , Proteínas del Grupo Polycomb , ARN Mensajero/biosíntesis , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Células Madre/enzimología , Células Madre/metabolismo , Células Madre/patología , Especificidad por Sustrato , Transcripción Genética , Transducción Genética
20.
J Biol Chem ; 280(2): 1199-208, 2005 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-15536069

RESUMEN

The transcriptional repressor E2F6 has been identified as a component of two distinct polycomb group protein (PcG)-containing complexes, suggesting a mechanism for the recruitment of repressive complexes to target sequences in DNA. Whereas one complex is involved in the repression of classic E2F target genes in G0, a role for E2F6 within the cell cycle has yet to be defined. We searched for novel E2F6-binding proteins using a yeast two-hybrid screen and identified the PcG protein, EPC1. We showed that, both in vitro and in vivo, E2F6, DP1, and EPC1 form a stable core complex with repressive activity. Furthermore, we identified the proliferation-specific PcG, EZH2, as an EPC1-interacting protein. Using affinity purification, we showed that E2F6, DP1, EPC1, EZH2, and Sin3B co-elute, suggesting the identification of a novel E2F6 complex that exists in vivo in both normal and transformed human cell lines. EZH2 is required for cellular proliferation and consistent with this, EZH2 elutes with the E2F6-EPC1 complex only in proliferating cells. Thus we have identified a novel E2F6-PcG complex (E2F6-EPC1) that interacts with EZH2 and may regulate genes required for cell cycle progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F6 , Proteína Potenciadora del Homólogo Zeste 2 , N-Metiltransferasa de Histona-Lisina , Humanos , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/aislamiento & purificación , Complejos Multiproteicos/metabolismo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas/genética , Proteínas Represoras/genética , Especificidad por Sustrato , Factor de Transcripción DP1 , Factores de Transcripción/química , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA