Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 20(11): 715, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31506602

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Immunity ; 52(1): 151-166.e6, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31924474

RESUMEN

In addition to helper and regulatory potential, CD4+ T cells also acquire cytotoxic activity marked by granzyme B (GzmB) expression and the ability to promote rejection of established tumors. Here, we examined the molecular and cellular mechanisms underpinning the differentiation of cytotoxic CD4+ T cells following immunotherapy. CD4+ transfer into lymphodepleted animals or regulatory T (Treg) cell depletion promoted GzmB expression by tumor-infiltrating CD4+, and this was prevented by interleukin-2 (IL-2) neutralization. Transcriptional analysis revealed a polyfunctional helper and cytotoxic phenotype characterized by the expression of the transcription factors T-bet and Blimp-1. While T-bet ablation restricted interferon-γ (IFN-γ) production, loss of Blimp-1 prevented GzmB expression in response to IL-2, suggesting two independent programs required for polyfunctionality of tumor-reactive CD4+ T cells. Our findings underscore the role of Treg cells, IL-2, and Blimp-1 in controlling the differentiation of cytotoxic CD4+ T cells and offer a pathway to enhancement of anti-tumor activity through their manipulation.


Asunto(s)
Granzimas/inmunología , Neoplasias/inmunología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Traslado Adoptivo , Animales , Línea Celular Tumoral , Humanos , Interferón gamma/inmunología , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/citología , Microambiente Tumoral/inmunología
3.
Nat Rev Mol Cell Biol ; 18(5): 331-337, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28270684

RESUMEN

Transcription and chromatin function are regulated by proteins that bind to DNA, nucleosomes or RNA polymerase II, with specific non-coding RNAs (ncRNAs) functioning to modulate their recruitment or activity. Unlike ncRNAs, nascent pre-mRNA was considered to be primarily a passive player in these processes. In this Opinion article, we describe recently identified interactions between nascent pre-mRNAs and regulatory proteins, highlight commonalities between the functions of nascent pre-mRNA and nascent ncRNA, and propose that both types of RNA have an active role in transcription and chromatin regulation.


Asunto(s)
Cromatina/metabolismo , Transcripción Genética , Animales , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Humanos , Empalme del ARN , Proteínas Represoras/metabolismo , Elongación de la Transcripción Genética , Factores de Transcripción
4.
Mol Cell ; 81(14): 2944-2959.e10, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34166609

RESUMEN

A number of regulatory factors are recruited to chromatin by specialized RNAs. Whether RNA has a more general role in regulating the interaction of proteins with chromatin has not been determined. We used proteomics methods to measure the global impact of nascent RNA on chromatin in embryonic stem cells. Surprisingly, we found that nascent RNA primarily antagonized the interaction of chromatin modifiers and transcriptional regulators with chromatin. Transcriptional inhibition and RNA degradation induced recruitment of a set of transcriptional regulators, chromatin modifiers, nucleosome remodelers, and regulators of higher-order structure. RNA directly bound to factors, including BAF, NuRD, EHMT1, and INO80 and inhibited their interaction with nucleosomes. The transcriptional elongation factor P-TEFb directly bound pre-mRNA, and its recruitment to chromatin upon Pol II inhibition was regulated by the 7SK ribonucleoprotein complex. We postulate that by antagonizing the interaction of regulatory proteins with chromatin, nascent RNA links transcriptional output with chromatin composition.


Asunto(s)
Cromatina/metabolismo , ARN/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Nucleosomas/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Unión Proteica/fisiología , Proteómica/métodos , ARN Polimerasa II/metabolismo , Transcripción Genética/fisiología , Factores de Elongación Transcripcional/metabolismo
5.
Nat Immunol ; 16(2): 207-213, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25531831

RESUMEN

Lymphocyte recruitment maintains intestinal immune homeostasis but also contributes to inflammation. The orphan chemoattractant receptor GPR15 mediates regulatory T cell homing and immunosuppression in the mouse colon. We show that GPR15 is also expressed by mouse TH17 and TH1 effector cells and is required for colitis in a model that depends on the trafficking of these cells to the colon. In humans GPR15 is expressed by effector cells, including pathogenic TH2 cells in ulcerative colitis, but is expressed poorly or not at all by colon regulatory T (Treg) cells. The TH2 transcriptional activator GATA-3 and the Treg-associated transcriptional repressor FOXP3 robustly bind human, but not mouse, GPR15 enhancer sequences, correlating with receptor expression. Our results highlight species differences in GPR15 regulation and suggest it as a potential therapeutic target for colitis.


Asunto(s)
Colitis/fisiopatología , Colon/fisiopatología , Regulación de la Expresión Génica , Receptores Acoplados a Proteínas G/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Receptores de Péptidos/metabolismo , Animales , Células Cultivadas , Colitis/inmunología , Colon/inmunología , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Factores de Transcripción Forkhead/metabolismo , Técnicas de Inactivación de Genes , Humanos , Ratones , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Especificidad de la Especie
6.
Nucleic Acids Res ; 50(8): 4557-4573, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35438764

RESUMEN

Lineage-determining transcription factors (LD-TFs) drive the differentiation of progenitor cells into a specific lineage. In CD4+ T cells, T-bet dictates differentiation of the TH1 lineage, whereas GATA3 drives differentiation of the alternative TH2 lineage. However, LD-TFs, including T-bet and GATA3, are frequently co-expressed but how this affects LD-TF function is not known. By expressing T-bet and GATA3 separately or together in mouse T cells, we show that T-bet sequesters GATA3 at its target sites, thereby removing GATA3 from TH2 genes. This redistribution of GATA3 is independent of GATA3 DNA binding activity and is instead mediated by the T-bet DNA binding domain, which interacts with the GATA3 DNA binding domain and changes GATA3's sequence binding preference. This mechanism allows T-bet to drive the TH1 gene expression program in the presence of GATA3. We propose that redistribution of one LD-TF by another may be a common mechanism that could explain how specific cell fate choices can be made even in the presence of other transcription factors driving alternative differentiation pathways.


Asunto(s)
Factor de Transcripción GATA3 , Proteínas de Dominio T Box/metabolismo , Células Th2 , Animales , Linaje de la Célula , ADN/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Expresión Génica , Ratones , Proteínas de Dominio T Box/genética , Células Th2/citología , Células Th2/metabolismo
7.
Eur J Immunol ; 52(4): 566-581, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35092032

RESUMEN

T-bet is the lineage-specifying transcription factor for CD4+ TH 1 cells. T-bet has also been found in other CD4+ T cell subsets, including TH 17 cells and Treg, where it modulates their functional characteristics. However, we lack information on when and where T-bet is expressed during T cell differentiation and how this impacts T cell differentiation and function. To address this, we traced the ontogeny of T-bet-expressing cells using a fluorescent fate-mapping mouse line. We demonstrate that T-bet is expressed in a subset of CD4+ T cells that have naïve cell surface markers and transcriptional profile and that this novel cell population is phenotypically and functionally distinct from previously described populations of naïve and memory CD4+ T cells. Naïve-like T-bet-experienced cells are polarized to the TH 1 lineage, predisposed to produce IFN-γ upon cell activation, and resist repolarization to other lineages in vitro and in vivo. These results demonstrate that lineage-specifying factors can polarize T cells in the absence of canonical markers of T cell activation and that this has an impact on the subsequent T-helper response.


Asunto(s)
Proteínas de Dominio T Box , Células TH1 , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo , Células Th2
8.
J Immunol ; 205(6): 1608-1619, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32817333

RESUMEN

CD4+ T cell functional inhibition (exhaustion) is a hallmark of malaria and correlates with impaired parasite control and infection chronicity. However, the mechanisms of CD4+ T cell exhaustion are still poorly understood. In this study, we show that Ag-experienced (Ag-exp) CD4+ T cell exhaustion during Plasmodium yoelii nonlethal infection occurs alongside the reduction in mammalian target of rapamycin (mTOR) activity and restriction in CD4+ T cell glycolytic capacity. We demonstrate that the loss of glycolytic metabolism and mTOR activity within the exhausted Ag-expCD4+ T cell population during infection coincides with reduction in T-bet expression. T-bet was found to directly bind to and control the transcription of various mTOR and metabolism-related genes within effector CD4+ T cells. Consistent with this, Ag-expTh1 cells exhibited significantly higher and sustained mTOR activity than effector T-bet- (non-Th1) Ag-expT cells throughout the course of malaria. We identified mTOR to be redundant for sustaining T-bet expression in activated Th1 cells, whereas mTOR was necessary but not sufficient for maintaining IFN-γ production by Th1 cells. Immunotherapy targeting PD-1, CTLA-4, and IL-27 blocked CD4+ T cell exhaustion during malaria infection and was associated with elevated T-bet expression and a concomitant increased CD4+ T cell glycolytic metabolism. Collectively, our data suggest that mTOR activity is linked to T-bet in Ag-expCD4+ T cells but that reduction in mTOR activity may not directly underpin Ag-expTh1 cell loss and exhaustion during malaria infection. These data have implications for therapeutic reactivation of exhausted CD4+ T cells during malaria infection and other chronic conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Malaria/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Plasmodium yoelii/fisiología , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Animales , Senescencia Celular , Regulación de la Expresión Génica , Glucólisis , Humanos , Tolerancia Inmunológica , Memoria Inmunológica , Interferón gamma/metabolismo , Interleucina-27/metabolismo , Activación de Linfocitos , Malaria/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética
9.
Immunity ; 37(4): 674-84, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23063332

RESUMEN

Mice lacking the transcription factor T-bet in the innate immune system develop microbiota-dependent colitis. Here, we show that interleukin-17A (IL-17A)-producing IL-7Rα(+) innate lymphoid cells (ILCs) were potent promoters of disease in Tbx21(-/-)Rag2(-/-) ulcerative colitis (TRUC) mice. TNF-α produced by CD103(-)CD11b(+) dendritic cells synergized with IL-23 to drive IL-17A production by ILCs, demonstrating a previously unrecognized layer of cellular crosstalk between dendritic cells and ILCs. We have identified Helicobacter typhlonius as a key disease trigger driving excess TNF-α production and promoting colitis in TRUC mice. Crucially, T-bet also suppressed the expression of IL-7R, a key molecule involved in controlling intestinal ILC homeostasis. The importance of IL-7R signaling in TRUC disease was highlighted by the dramatic reduction in intestinal ILCs and attenuated colitis following IL-7R blockade. Taken together, these data demonstrate the mechanism by which T-bet regulates the complex interplay between mucosal dendritic cells, ILCs, and the intestinal microbiota.


Asunto(s)
Colitis Ulcerosa/inmunología , Proteínas de Unión al ADN/inmunología , Inmunidad Innata , Linfocitos/inmunología , Receptores de Interleucina-7/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Células Cultivadas , Enfermedad Crónica , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Proteínas de Unión al ADN/deficiencia , Helicobacter/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal , Proteínas de Dominio T Box/deficiencia
10.
Am J Transplant ; 20(10): 2715-2727, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32277570

RESUMEN

Organ transplantation is often lifesaving, but the long-term deleterious effects of combinatorial immunosuppression regimens and allograft failure cause significant morbidity and mortality. Long-term graft survival in the absence of continuing immunosuppression, defined as operational tolerance, has never been described in the context of multiple major histocompatibility complex (MHC) mismatches. Here, we show that miR-142 deficiency leads to indefinite allograft survival in a fully MHC mismatched murine cardiac transplant model in the absence of exogenous immunosuppression. We demonstrate that the cause of indefinite allograft survival in the absence of miR-142 maps specifically to the T cell compartment. Of therapeutic relevance, temporal deletion of miR-142 in adult mice prior to transplantation of a fully MHC mismatched skin allograft resulted in prolonged allograft survival. Mechanistically, miR-142 directly targets Tgfbr1 for repression in regulatory T cells (TREG ). This leads to increased TREG sensitivity to transforming growth factor - beta and promotes transplant tolerance via an augmented peripheral TREG response in the absence of miR-142. These data identify manipulation of miR-142 as a promising approach for the induction of tolerance in human transplantation.


Asunto(s)
Rechazo de Injerto , MicroARNs , Aloinjertos , Animales , Rechazo de Injerto/etiología , Supervivencia de Injerto , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , MicroARNs/genética , Linfocitos T Reguladores , Tolerancia al Trasplante , Trasplante Homólogo
11.
PLoS Genet ; 13(2): e1006587, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28187197

RESUMEN

The polarization of CD4+ T cells into distinct T helper cell lineages is essential for protective immunity against infection, but aberrant T cell polarization can cause autoimmunity. The transcription factor T-bet (TBX21) specifies the Th1 lineage and represses alternative T cell fates. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) that may be causative for autoimmune diseases. The majority of these polymorphisms are located within non-coding distal regulatory elements. It is considered that these genetic variants contribute to disease by altering the binding of regulatory proteins and thus gene expression, but whether these variants alter the binding of lineage-specifying transcription factors has not been determined. Here, we show that SNPs associated with the mucosal inflammatory diseases Crohn's disease, ulcerative colitis (UC) and celiac disease, but not rheumatoid arthritis or psoriasis, are enriched at T-bet binding sites. Furthermore, we identify disease-associated variants that alter T-bet binding in vitro and in vivo. ChIP-seq for T-bet in individuals heterozygous for the celiac disease-associated SNPs rs1465321 and rs2058622 and the IBD-associated SNPs rs1551398 and rs1551399, reveals decreased binding to the minor disease-associated alleles. Furthermore, we show that rs1465321 is an expression quantitative trait locus (eQTL) for the neighboring gene IL18RAP, with decreased T-bet binding associated with decreased expression of this gene. These results suggest that genetic polymorphisms may predispose individuals to mucosal autoimmune disease through alterations in T-bet binding. Other disease-associated variants may similarly act by modulating the binding of lineage-specifying transcription factors in a tissue-selective and disease-specific manner.


Asunto(s)
Enfermedad Celíaca/genética , Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple , Proteínas de Dominio T Box/genética , Animales , Sitios de Unión/genética , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Celíaca/metabolismo , Células Cultivadas , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Humanos , Subunidad beta del Receptor de Interleucina-18/genética , Subunidad beta del Receptor de Interleucina-18/metabolismo , Ratones Noqueados , Unión Proteica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo
12.
Genome Res ; 26(7): 896-907, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27197219

RESUMEN

Polycomb repressive complex 2 (PRC2) modifies chromatin to maintain genes in a repressed state during development. PRC2 is primarily associated with CpG islands at repressed genes and also possesses RNA binding activity. However, the RNAs that bind PRC2 in cells, the subunits that mediate these interactions, and the role of RNA in PRC2 recruitment to chromatin all remain unclear. By performing iCLIP for PRC2 in comparison with other RNA binding proteins, we show here that PRC2 binds nascent RNA at essentially all active genes. Although interacting with RNA promiscuously, PRC2 binding is enriched at specific locations within RNAs, primarily exon-intron boundaries and the 3' UTR. Deletion of other PRC2 subunits reveals that SUZ12 is sufficient to establish this RNA binding profile. Contrary to prevailing models, we also demonstrate that the interaction of PRC2 with RNA or chromatin is mutually antagonistic in cells and in vitro. RNA degradation in cells triggers PRC2 recruitment to CpG islands at active genes. Correspondingly, the release of PRC2 from chromatin in cells increases RNA binding. Consistent with this, RNA and nucleosomes compete for PRC2 binding in vitro. We propose that RNA prevents PRC2 recruitment to chromatin at active genes and that mutual antagonism between RNA and chromatin underlies the pattern of PRC2 chromatin association across the genome.


Asunto(s)
Cromatina/metabolismo , Complejo Represivo Polycomb 2/fisiología , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Animales , Células Cultivadas , Exones , Regulación de la Expresión Génica , Intrones , Ratones , Células Madre Embrionarias de Ratones/fisiología , Nucleosomas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Unión Proteica , Estabilidad del ARN
13.
Mol Cell ; 38(5): 675-88, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20542000

RESUMEN

Polycomb proteins maintain cell identity by repressing the expression of developmental regulators specific for other cell types. Polycomb repressive complex-2 (PRC2) catalyzes trimethylation of histone H3 lysine-27 (H3K27me3). Although repressed, PRC2 targets are generally associated with the transcriptional initiation marker H3K4me3, but the significance of this remains unclear. Here, we identify a class of short RNAs, approximately 50-200 nucleotides in length, transcribed from the 5' end of polycomb target genes in primary T cells and embryonic stem cells. Short RNA transcription is associated with RNA polymerase II and H3K4me3, occurs in the absence of mRNA transcription, and is independent of polycomb activity. Short RNAs form stem-loop structures resembling PRC2 binding sites in Xist, interact with PRC2 through SUZ12, cause gene repression in cis, and are depleted from polycomb target genes activated during cell differentiation. We propose that short RNAs play a role in the association of PRC2 with its target genes.


Asunto(s)
ARN/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Ratones , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/fisiología , Conformación de Ácido Nucleico , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , ARN/química , ARN/genética , Proteínas Represoras/genética , Linfocitos T/citología , Linfocitos T/fisiología
14.
Nucleic Acids Res ; 44(10): 4636-50, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26883634

RESUMEN

In B cells infected by the cancer-associated Epstein-Barr virus (EBV), RUNX3 and RUNX1 transcription is manipulated to control cell growth. The EBV-encoded EBNA2 transcription factor (TF) activates RUNX3 transcription leading to RUNX3-mediated repression of the RUNX1 promoter and the relief of RUNX1-directed growth repression. We show that EBNA2 activates RUNX3 through a specific element within a -97 kb super-enhancer in a manner dependent on the expression of the Notch DNA-binding partner RBP-J. We also reveal that the EBV TFs EBNA3B and EBNA3C contribute to RUNX3 activation in EBV-infected cells by targeting the same element. Uncovering a counter-regulatory feed-forward step, we demonstrate EBNA2 activation of a RUNX1 super-enhancer (-139 to -250 kb) that results in low-level RUNX1 expression in cells refractory to RUNX1-mediated growth inhibition. EBNA2 activation of the RUNX1 super-enhancer is also dependent on RBP-J. Consistent with the context-dependent roles of EBNA3B and EBNA3C as activators or repressors, we find that these proteins negatively regulate the RUNX1 super-enhancer, curbing EBNA2 activation. Taken together our results reveal cell-type-specific exploitation of RUNX gene super-enhancers by multiple EBV TFs via the Notch pathway to fine tune RUNX3 and RUNX1 expression and manipulate B-cell growth.


Asunto(s)
Linfocitos B/virología , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Elementos de Facilitación Genéticos , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Linfocitos B/metabolismo , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptores Notch/metabolismo
15.
Nucleic Acids Res ; 43(12): 5785-97, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-25990740

RESUMEN

In this report we have analyzed the role of antisense transcription in the control of LEF1 transcription factor expression. A natural antisense transcript (NAT) is transcribed from a promoter present in the first intron of LEF1 gene and undergoes splicing in mesenchymal cells. Although this locus is silent in epithelial cells, and neither NAT transcript nor LEF1 mRNA are expressed, in cell lines with an intermediate epithelial-mesenchymal phenotype presenting low LEF1 expression, the NAT is synthesized and remains unprocessed. Contrarily to the spliced NAT, this unspliced NAT down-regulates the main LEF1 promoter activity and attenuates LEF1 mRNA transcription. Unspliced LEF1 NAT interacts with LEF1 promoter and facilitates PRC2 binding to the LEF1 promoter and trimethylation of lysine 27 in histone 3. Expression of the spliced form of LEF1 NAT in trans prevents the action of unspliced NAT by competing for interaction with the promoter. Thus, these results indicate that LEF1 gene expression is attenuated by an antisense non-coding RNA and that this NAT function is regulated by the balance between its spliced and unspliced forms.


Asunto(s)
Regulación de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/genética , Empalme del ARN , ARN sin Sentido/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética
16.
Nucleic Acids Res ; 43(7): 3563-77, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25779048

RESUMEN

Lytic replication of the human gamma herpes virus Epstein-Barr virus (EBV) is an essential prerequisite for the spread of the virus. Differential regulation of a limited number of cellular genes has been reported in B-cells during the viral lytic replication cycle. We asked whether a viral bZIP transcription factor, Zta (BZLF1, ZEBRA, EB1), drives some of these changes. Using genome-wide chromatin immunoprecipitation coupled to next-generation DNA sequencing (ChIP-seq) we established a map of Zta interactions across the human genome. Using sensitive transcriptome analyses we identified 2263 cellular genes whose expression is significantly changed during the EBV lytic replication cycle. Zta binds 278 of the regulated genes and the distribution of binding sites shows that Zta binds mostly to sites that are distal to transcription start sites. This differs from the prevailing view that Zta activates viral genes by binding exclusively at promoter elements. We show that a synthetic Zta binding element confers Zta regulation at a distance and that distal Zta binding sites from cellular genes can confer Zta-mediated regulation on a heterologous promoter. This leads us to propose that Zta directly reprograms the expression of cellular genes through distal elements.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 4/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Transactivadores/fisiología , Secuencia de Bases , Línea Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa , Transcriptoma
17.
PLoS Pathog ; 9(9): e1003636, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068937

RESUMEN

Epstein-Barr virus (EBV) epigenetically reprogrammes B-lymphocytes to drive immortalization and facilitate viral persistence. Host-cell transcription is perturbed principally through the actions of EBV EBNA 2, 3A, 3B and 3C, with cellular genes deregulated by specific combinations of these EBNAs through unknown mechanisms. Comparing human genome binding by these viral transcription factors, we discovered that 25% of binding sites were shared by EBNA 2 and the EBNA 3s and were located predominantly in enhancers. Moreover, 80% of potential EBNA 3A, 3B or 3C target genes were also targeted by EBNA 2, implicating extensive interplay between EBNA 2 and 3 proteins in cellular reprogramming. Investigating shared enhancer sites neighbouring two new targets (WEE1 and CTBP2) we discovered that EBNA 3 proteins repress transcription by modulating enhancer-promoter loop formation to establish repressive chromatin hubs or prevent assembly of active hubs. Re-ChIP analysis revealed that EBNA 2 and 3 proteins do not bind simultaneously at shared sites but compete for binding thereby modulating enhancer-promoter interactions. At an EBNA 3-only intergenic enhancer site between ADAM28 and ADAMDEC1 EBNA 3C was also able to independently direct epigenetic repression of both genes through enhancer-promoter looping. Significantly, studying shared or unique EBNA 3 binding sites at WEE1, CTBP2, ITGAL (LFA-1 alpha chain), BCL2L11 (Bim) and the ADAMs, we also discovered that different sets of EBNA 3 proteins bind regulatory elements in a gene and cell-type specific manner. Binding profiles correlated with the effects of individual EBNA 3 proteins on the expression of these genes, providing a molecular basis for the targeting of different sets of cellular genes by the EBNA 3s. Our results therefore highlight the influence of the genomic and cellular context in determining the specificity of gene deregulation by EBV and provide a paradigm for host-cell reprogramming through modulation of enhancer-promoter interactions by viral transcription factors.


Asunto(s)
Reprogramación Celular , Elementos de Facilitación Genéticos , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Marcación de Gen , Herpesvirus Humano 4/metabolismo , Modelos Biológicos , Proteínas Represoras/metabolismo , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Sitios de Unión , Unión Competitiva , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Co-Represoras , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Antígenos Nucleares del Virus de Epstein-Barr/química , Antígenos Nucleares del Virus de Epstein-Barr/genética , Interacciones Huésped-Patógeno , Humanos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
J Immunol ; 191(12): 5925-32, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24249732

RESUMEN

The complex relationship between Th1 and Th17 cells is incompletely understood. The transcription factor T-bet is best known as the master regulator of Th1 lineage commitment. However, attention is now focused on the repression of alternate T cell subsets mediated by T-bet, particularly the Th17 lineage. It has recently been suggested that pathogenic Th17 cells express T-bet and are dependent on IL-23. However, T-bet has previously been shown to be a negative regulator of Th17 cells. We have taken an unbiased approach to determine the functional impact of T-bet on Th17 lineage commitment. Genome-wide analysis of functional T-bet binding sites provides an improved understanding of the transcriptional regulation mediated by T-bet, and suggests novel mechanisms by which T-bet regulates Th cell differentiation. Specifically, we show that T-bet negatively regulates Th17 lineage commitment via direct repression of the transcription factor IFN regulatory factor-4 (IRF4). An in vivo analysis of the pathogenicity of T-bet-deficient T cells demonstrated that mucosal Th17 responses were augmented in the absence of T-bet, and we have demonstrated that the roles of T-bet in enforcing Th1 responses and suppressing Th17 responses are separable. The interplay of the two key transcription factors T-bet and IRF4 during the determination of T cell fate choice significantly advances our understanding of the mechanisms underlying the development of pathogenic T cells.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/antagonistas & inhibidores , Linfopoyesis/genética , Proteínas de Dominio T Box/fisiología , Células Th17/citología , Transcripción Genética , Traslado Adoptivo , Animales , Sitios de Unión , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Células Cultivadas , Quimera , Colitis/inmunología , Proteínas de Unión al ADN/deficiencia , Femenino , Genes Reporteros , Vectores Genéticos , Estudio de Asociación del Genoma Completo , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Dominio T Box/genética
19.
Retrovirology ; 11: 53, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24990269

RESUMEN

BACKGROUND: Human Immunodeficiency Virus 1 (HIV-1) exhibits a wide range of interactions with the host cell but whether viral proteins interact with cellular RNA is not clear. A candidate interacting factor is the trans-activator of transcription (Tat) protein. Tat is required for expression of virus genes but activates transcription through an unusual mechanism; binding to an RNA stem-loop, the transactivation response element (TAR), with the host elongation factor P-TEFb. HIV-1 Tat has also been shown to alter the expression of host genes during infection, contributing to viral pathogenesis but, whether Tat also interacts with cellular RNAs is unknown. RESULTS: Using RNA immunoprecipitation coupled with microarray analysis, we have discovered that HIV-1 Tat is associated with a specific set of human mRNAs in T cells. mRNAs bound by Tat share a stem-loop structural element and encode proteins with common biological roles. In contrast, we do not find evidence that Tat associates with microRNAs or the RNA-induced silencing complex (RISC). The interaction of Tat with cellular RNA requires an intact RNA binding domain and Tat RNA binding is linked to an increase in RNA abundance in cell lines and during infection of primary CD4+ T cells by HIV. CONCLUSIONS: We conclude that Tat interacts with a specific set of human mRNAs in T cells, many of which show changes in abundance in response to Tat and HIV infection. This work uncovers a previously unrecognised interaction between HIV and its host that may contribute to viral alteration of the host cellular environment.


Asunto(s)
Infecciones por VIH/virología , VIH-1/genética , ARN Mensajero/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Secuencia de Bases , Linfocitos T CD4-Positivos/virología , Línea Celular , Regulación Viral de la Expresión Génica , Células HEK293 , Humanos , MicroARNs/genética , Datos de Secuencia Molecular , Unión Proteica/genética , Proteínas de Unión al ARN/genética , Transcripción Genética , Activación Transcripcional/genética
20.
Cell Rep ; 43(3): 113856, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38416641

RESUMEN

Polycomb repressive complex 2 (PRC2) modifies chromatin to maintain repression of genes specific for other cell lineages. In vitro, RNA inhibits PRC2 activity, but the effect of RNA on PRC2 in cells is less clear, with studies concluding that RNA either antagonizes or promotes PRC2 chromatin association. The addition of RNase A to chromatin immunoprecipitation reactions has been reported to reduce detection of PRC2 target sites, suggesting the existence of RNA bridges connecting PRC2 to chromatin. Here, we show that the apparent loss of PRC2 chromatin association after RNase A treatment is due to non-specific chromatin precipitation. RNA degradation precipitates chromatin out of solution, thereby masking enrichment of specific DNA sequences in chromatin immunoprecipitation reactions. Maintaining chromatin solubility by the addition of poly-L-glutamic acid rescues detection of PRC2 chromatin occupancy upon RNA degradation. These findings undermine support for the model that RNA bridges PRC2 and chromatin in cells.


Asunto(s)
Cromatina , Complejo Represivo Polycomb 2 , Complejo Represivo Polycomb 2/metabolismo , ARN/metabolismo , Artefactos , Ribonucleasa Pancreática/metabolismo , Estabilidad del ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA