Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nat Immunol ; 18(8): 931-939, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28604718

RESUMEN

Activated CD8+ T cells differentiate into cytotoxic effector (TEFF) cells that eliminate target cells. How TEFF cell identity is established and maintained is not fully understood. We found that Runx3 deficiency limited clonal expansion and impaired upregulation of cytotoxic molecules in TEFF cells. Runx3-deficient CD8+ TEFF cells aberrantly upregulated genes characteristic of follicular helper T (TFH) cell lineage, including Bcl6, Tcf7 and Cxcr5. Mechanistically, the Runx3-CBFß transcription factor complex deployed H3K27me3 to Bcl6 and Tcf7 genes to suppress the TFH program. Ablating Tcf7 in Runx3-deficient CD8+ TEFF cells prevented the upregulation of TFH genes and ameliorated their defective induction of cytotoxic genes. As such, Runx3-mediated Tcf7 repression coordinately enforced acquisition of cytotoxic functions and protected the cytotoxic lineage integrity by preventing TFH-lineage deviation.


Asunto(s)
Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Linfopoyesis/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Linaje de la Célula , Ensayo de Inmunoadsorción Enzimática , Epigénesis Genética , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Inmunohistoquímica , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores CXCR5/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Regulación hacia Arriba
2.
Nat Immunol ; 17(6): 695-703, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111144

RESUMEN

The CD4(+) and CD8(+) T cell dichotomy is essential for effective cellular immunity. How individual T cell identity is established remains poorly understood. Here we show that the high-mobility group (HMG) transcription factors Tcf1 and Lef1 are essential for repressing CD4(+) lineage-associated genes including Cd4, Foxp3 and Rorc in CD8(+) T cells. Tcf1- and Lef1-deficient CD8(+) T cells exhibit histone hyperacetylation, which can be ascribed to intrinsic histone deacetylase (HDAC) activity in Tcf1 and Lef1. Mutation of five conserved amino acids in the Tcf1 HDAC domain diminishes HDAC activity and the ability to suppress CD4(+) lineage genes in CD8(+) T cells. These findings reveal that sequence-specific transcription factors can utilize intrinsic HDAC activity to guard cell identity by repressing lineage-inappropriate genes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Histona Desacetilasas/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Acetilación , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Histona Desacetilasas/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Dominios Proteicos/genética
3.
Nat Immunol ; 16(9): 980-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26214741

RESUMEN

Follicular helper T cells (T(FH) cells) are specialized effector CD4(+) T cells that help B cells develop germinal centers (GCs) and memory. However, the transcription factors that regulate the differentiation of T(FH) cells remain incompletely understood. Here we report that selective loss of Lef1 or Tcf7 (which encode the transcription factor LEF-1 or TCF-1, respectively) resulted in T(FH) cell defects, while deletion of both Lef1 and Tcf7 severely impaired the differentiation of T(FH) cells and the formation of GCs. Forced expression of LEF-1 enhanced T(FH) differentiation. LEF-1 and TCF-1 coordinated such differentiation by two general mechanisms. First, they established the responsiveness of naive CD4(+) T cells to T(FH) cell signals. Second, they promoted early T(FH) differentiation via the multipronged approach of sustaining expression of the cytokine receptors IL-6Rα and gp130, enhancing expression of the costimulatory receptor ICOS and promoting expression of the transcriptional repressor Bcl6.


Asunto(s)
Diferenciación Celular/inmunología , Receptor gp130 de Citocinas/inmunología , Proteínas de Unión al ADN/inmunología , Centro Germinal/inmunología , Factor Nuclear 1-alfa del Hepatocito/inmunología , Factor de Unión 1 al Potenciador Linfoide/inmunología , Receptores de Interleucina-6/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Diferenciación Celular/genética , Receptor gp130 de Citocinas/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Centro Germinal/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Proteínas Proto-Oncogénicas c-bcl-6 , Receptores de Interleucina-6/genética , Linfocitos T Colaboradores-Inductores/metabolismo
4.
Nucleic Acids Res ; 48(15): 8724-8739, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32735645

RESUMEN

T cell activation is a well-established model for studying cellular responses to exogenous stimulation. Motivated by our previous finding that intron retention (IR) could lead to transcript instability, in this study, we performed BruChase-Seq to experimentally monitor the expression dynamics of nascent transcripts in resting and activated CD4+ T cells. Computational modeling was then applied to quantify the stability of spliced and intron-retained transcripts on a genome-wide scale. Beyond substantiating that intron-retained transcripts were considerably less stable than spliced transcripts, we found a global stabilization of spliced mRNAs upon T cell activation, although the stability of intron-retained transcripts remained relatively constant. In addition, we identified that La-related protein 4 (LARP4), an RNA-binding protein (RBP) known to enhance mRNA stability, was involved in T cell activation-dependent mRNA stabilization. Knocking out Larp4 in mice destabilized Nfκb1 mRNAs and reduced secretion of interleukin-2 (IL2) and interferon-gamma (IFNγ), two factors critical for T cell proliferation and function. We propose that coordination between splicing regulation and mRNA stability may provide a novel paradigm to control spatiotemporal gene expression during T cell activation.


Asunto(s)
Interferón gamma/genética , Interleucina-2/genética , Proteínas/genética , Estabilidad del ARN/genética , Transcriptoma/genética , Empalme Alternativo/genética , Animales , Humanos , Intrones/genética , Activación de Linfocitos/genética , Ratones , FN-kappa B/genética , Unión Proteica/genética , ARN Mensajero/genética , Linfocitos T/metabolismo
5.
J Immunol ; 199(3): 911-919, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28652395

RESUMEN

In response to acute viral infection, activated naive T cells give rise to effector T cells that clear the pathogen and memory T cells that persist long-term and provide heightened protection. T cell factor 1 (Tcf1) is essential for several of these differentiation processes. Tcf1 is expressed in multiple isoforms, with all isoforms sharing the same HDAC and DNA-binding domains and the long isoforms containing a unique N-terminal ß-catenin-interacting domain. In this study, we specifically ablated Tcf1 long isoforms in mice, while retaining expression of Tcf1 short isoforms. During CD8+ T cell responses, Tcf1 long isoforms were dispensable for generating cytotoxic CD8+ effector T cells and maintaining memory CD8+ T cell pool size, but they contributed to optimal maturation of central memory CD8+ T cells and their optimal secondary expansion in a recall response. In contrast, Tcf1 long isoforms were required for differentiation of T follicular helper (TFH) cells, but not TH1 effectors, elicited by viral infection. Although Tcf1 short isoforms adequately supported Bcl6 and ICOS expression in TFH cells, Tcf1 long isoforms remained important for suppressing the expression of Blimp1 and TH1-associated genes and for positively regulating Id3 to restrain germinal center TFH cell differentiation. Furthermore, formation of memory TH1 and memory TFH cells strongly depended on Tcf1 long isoforms. These data reveal that Tcf1 long and short isoforms have distinct, yet complementary, functions and may represent an evolutionarily conserved means to ensure proper programming of CD8+ and CD4+ T cell responses to viral infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Factor 1 de Transcripción de Linfocitos T/química , Factor 1 de Transcripción de Linfocitos T/inmunología , Animales , Diferenciación Celular , Pruebas Inmunológicas de Citotoxicidad , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Memoria Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Isoformas de Proteínas , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Factor 1 de Transcripción de Linfocitos T/deficiencia , Factor 1 de Transcripción de Linfocitos T/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Nucleic Acids Res ; 44(14): 6817-29, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27369383

RESUMEN

T cell activation is a well-established model for studying cellular responses to exogenous stimulation. Using strand-specific RNA-seq, we observed that intron retention is prevalent in polyadenylated transcripts in resting CD4(+) T cells and is significantly reduced upon T cell activation. Several lines of evidence suggest that intron-retained transcripts are less stable than fully spliced transcripts. Strikingly, the decrease in intron retention (IR) levels correlate with the increase in steady-state mRNA levels. Further, the majority of the genes upregulated in activated T cells are accompanied by a significant reduction in IR. Of these 1583 genes, 185 genes are predominantly regulated at the IR level, and highly enriched in the proteasome pathway, which is essential for proper T cell proliferation and cytokine release. These observations were corroborated in both human and mouse CD4(+) T cells. Our study revealed a novel post-transcriptional regulatory mechanism that may potentially contribute to coordinated and/or quick cellular responses to extracellular stimuli such as an acute infection.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica , Intrones/genética , Activación de Linfocitos/genética , Animales , Secuencia Conservada/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética
7.
Sci Rep ; 13(1): 5141, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36991079

RESUMEN

Regulation of intron retention (IR), a form of alternative splicing, is a newly recognized checkpoint in gene expression. Since there are numerous abnormalities in gene expression in the prototypic autoimmune disease systemic lupus erythematosus (SLE), we sought to determine whether IR was intact in patients with this disease. We, therefore, studied global gene expression and IR patterns of lymphocytes in SLE patients. We analyzed RNA-seq data from peripheral blood T cell samples from 14 patients suffering from systemic lupus erythematosus (SLE) and 4 healthy controls and a second, independent data set of RNA-seq data from B cells from16 SLE patients and 4 healthy controls. We identified intron retention levels from 26,372 well annotated genes as well as differential gene expression and tested for differences between cases and controls using unbiased hierarchical clustering and principal component analysis. We followed with gene-disease enrichment analysis and gene-ontology enrichment analysis. Finally, we then tested for significant differences in intron retention between cases and controls both globally and with respect to specific genes. Overall decreased IR was found in T cells from one cohort and B cells from another cohort of patients with SLE and was associated with increased expression of numerous genes, including those encoding spliceosome components. Different introns within the same gene displayed both up- and down-regulated retention profiles indicating a complex regulatory mechanism. These results indicate that decreased IR in immune cells is characteristic of patients with active SLE and may contribute to the abnormal expression of specific genes in this autoimmune disease.


Asunto(s)
Lupus Eritematoso Sistémico , Linfocitos T , Humanos , Intrones/genética , Linfocitos T/metabolismo , Linfocitos B
8.
Nat Commun ; 12(1): 5863, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34615872

RESUMEN

T cell identity is established during thymic development, but how it is maintained in the periphery remains unknown. Here we show that ablating Tcf1 and Lef1 transcription factors in mature CD8+ T cells aberrantly induces genes from non-T cell lineages. Using high-throughput chromosome-conformation-capture sequencing, we demonstrate that Tcf1/Lef1 are important for maintaining three-dimensional genome organization at multiple scales in CD8+ T cells. Comprehensive network analyses coupled with genome-wide profiling of chromatin accessibility and Tcf1 occupancy show the direct impact of Tcf1/Lef1 on the T cell genome is to promote formation of extensively interconnected hubs through enforcing chromatin interaction and accessibility. The integrative mechanisms utilized by Tcf1/Lef1 underlie activation of T cell identity genes and repression of non-T lineage genes, conferring fine control of various T cell functionalities. These findings suggest that Tcf1/Lef1 control global genome organization and help form intricate chromatin-interacting hubs to facilitate promoter-enhancer/silencer contact, hence providing constant supervision of CD8+ T cell identity and function.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Genómica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Animales , Diferenciación Celular , Factor Nuclear 1-alfa del Hepatocito/genética , Inmunogenética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Transcriptoma
9.
J Exp Med ; 216(4): 847-866, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30837262

RESUMEN

Tcf1 and Lef1 have versatile functions in regulating T cell development and differentiation, but intrinsic requirements for these factors in regulatory T (T reg) cells remain to be unequivocally defined. Specific ablation of Tcf1 and Lef1 in T reg cells resulted in spontaneous multi-organ autoimmunity that became more evident with age. Tcf1/Lef1-deficient T regs showed reduced protection against experimentally induced colitis, indicative of diminished immuno-suppressive capacity. Transcriptomic analysis revealed that Tcf1 and Lef1 were responsible for positive regulation of a subset of T reg-overrepresented signature genes such as Ikzf4 and Izumo1r Unexpectedly, Tcf1 and Lef1 were necessary for restraining expression of cytotoxic CD8+ effector T cell-associated genes in T reg cells, including Prdm1 and Ifng Tcf1 ChIP-seq revealed substantial overlap between Tcf1 and Foxp3 binding peaks in the T reg cell genome, with Tcf1-Foxp3 cooccupancy observed at key T reg signature and cytotoxic effector genes. Our data collectively indicate that Tcf1 and Lef1 are critical for sustaining T reg suppressive functions and preventing loss of self-tolerance.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , Tolerancia Inmunológica , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transcriptoma/inmunología
10.
Nat Commun ; 9(1): 4221, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30310060

RESUMEN

The CCCTC-binding zinc-finger protein (CTCF)-mediated network of long-range chromatin interactions is important for genome organization and function. Although this network has been considered largely invariant, we find that it exhibits extensive cell-type-specific interactions that contribute to cell identity. Here, we present Lollipop, a machine-learning framework, which predicts CTCF-mediated long-range interactions using genomic and epigenomic features. Using ChIA-PET data as benchmark, we demonstrate that Lollipop accurately predicts CTCF-mediated chromatin interactions both within and across cell types, and outperforms other methods based only on CTCF motif orientation. Predictions are confirmed computationally and experimentally by Chromatin Conformation Capture (3C). Moreover, our approach identifies other determinants of CTCF-mediated chromatin wiring, such as gene expression within the loops. Our study contributes to a better understanding about the underlying principles of CTCF-mediated chromatin interactions and their impact on gene expression.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Genoma , Línea Celular , Redes Reguladoras de Genes , Humanos , Curva ROC
11.
Nat Commun ; 9(1): 5452, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30575739

RESUMEN

Ezh2 is an histone methyltransferase (HMT) that catalyzes H3K27me3 and functions in TH1, TH2, and Treg cells primarily via HMT activity. Here we show that Ezh2 ablation impairs T follicular helper (TFH) cell differentiation and activation of the TFH transcription program. In TFH cells, most Ezh2-occupied genomic sites, including the Bcl6 promoter, are associated with H3K27ac rather than H3K27me3. Mechanistically, Ezh2 is recruited by Tcf1 to directly activate Bcl6 transcription, with this function requiring Ezh2 phosphorylation at Ser21. Meanwhile, Ezh2 deploys H3K27me3 to repress Cdkn2a expression in TFH cells, where aberrantly upregulated p19Arf, a Cdkn2a protein product, triggers TFH cell apoptosis and antagonizes Bcl6 function via protein-protein interaction. Either forced expression of Bcl6 or genetic ablation of p19Arf in Ezh2-deficient cells improves TFH cell differentiation and helper function. Thus, Ezh2 orchestrates TFH-lineage specification and function maturation by integrating phosphorylation-dependent transcriptional activation and HMT-dependent gene repression.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Diferenciación Celular , Supervivencia Celular , Epigénesis Genética , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA