Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(2): 294-306, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38238608

RESUMEN

Antigen-experienced CD8+ T cells form effector and central memory T cells (TEM and TCM cells, respectively); however, the mechanism(s) controlling their lineage plasticity remains incompletely understood. Here we show that the transcription cofactor Tle3 critically regulates TEM and TCM cell fates and lineage stability through dynamic redistribution in antigen-responding CD8+ T cell genome. Genetic ablation of Tle3 promoted CD8+ TCM cell formation at the expense of CD8+ TEM cells. Lineage tracing showed that Tle3-deficient CD8+ TEM cells underwent accelerated conversion into CD8+ TCM cells while retaining robust recall capacity. Tle3 acted as a coactivator for Tbet to increase chromatin opening at CD8+ TEM cell-characteristic sites and to activate CD8+ TEM cell signature gene transcription, while engaging Runx3 and Tcf1 to limit CD8+ TCM cell-characteristic molecular features. Thus, Tle3 integrated functions of multiple transcription factors to guard lineage fidelity of CD8+ TEM cells, and manipulation of Tle3 activity could favor CD8+ TCM cell production.


Asunto(s)
Linfocitos T CD8-positivos , Células T de Memoria , Factores de Transcripción/genética , Diferenciación Celular , Memoria Inmunológica/genética
2.
Nat Immunol ; 23(8): 1222-1235, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35882936

RESUMEN

CD8+ T cell homeostasis is maintained by the cytokines IL-7 and IL-15. Here we show that transcription factors Tcf1 and Lef1 were intrinsically required for homeostatic proliferation of CD8+ T cells. Multiomics analyses showed that Tcf1 recruited the genome organizer CTCF and that homeostatic cytokines induced Tcf1-dependent CTCF redistribution in the CD8+ T cell genome. Hi-C coupled with network analyses indicated that Tcf1 and CTCF acted cooperatively to promote chromatin interactions and form highly connected, dynamic interaction hubs in CD8+ T cells before and after cytokine stimulation. Ablating CTCF phenocopied the proliferative defects caused by Tcf1 and Lef1 deficiency. Tcf1 and CTCF controlled a similar set of genes that regulated cell cycle progression and promoted CD8+ T cell homeostatic proliferation in vivo. These findings identified CTCF as a Tcf1 cofactor and uncovered an intricate interplay between Tcf1 and CTCF that modulates the genomic architecture of CD8+ T cells to preserve homeostasis.


Asunto(s)
Linfocitos T CD8-positivos , Transducción de Señal , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Genómica , Homeostasis
3.
Nat Immunol ; 23(3): 386-398, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35190717

RESUMEN

The mechanisms underlying the heightened protection mediated by central memory CD8+ T (TCM) cells remain unclear. Here we show that the transcription factor Tcf1 was required in resting TCM cells to generate secondary effector CD8+ T cells and to clear pathogens during recall responses. Recall stimulation of CD8+ TCM cells caused extensive reprogramming of the transcriptome and chromatin accessibility, leading to rapid induction of glycolytic enzymes, cell cycle regulators and transcriptional regulators, including Id3. This cluster of genes did not require Tcf1 in resting CD8+ TCM cells, but depended on Tcf1 for optimal induction and chromatin opening in recall-stimulated CD8+ TCM cells. Tcf1 bound extensively to these recall-induced gene loci in resting CD8+ TCM cells and mediated chromatin interactions that positioned these genes in architectural proximity with poised enhancers. Thus, Tcf1 preprogramed a transcriptional program that supported the bioenergetic and proliferative needs of CD8+ TCM cells in case of a secondary challenge.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Glucólisis/genética , Memoria Inmunológica/genética , Ratones , Ratones Endogámicos C57BL
6.
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
7.
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
8.
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
9.
Proc Natl Acad Sci U S A ; 120(51): e2313476120, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38085779

RESUMEN

CD62L+ central memory CD8+ T (TCM) cells provide enhanced protection than naive cells; however, the underlying mechanism, especially the contribution of higher-order genomic organization, remains unclear. Systematic Hi-C analyses reveal that antigen-experienced CD8+ T cells undergo extensive rewiring of chromatin interactions (ChrInt), with TCM cells harboring specific interaction hubs compared with naive CD8+ T cells, as observed at cytotoxic effector genes such as Ifng and Tbx21. TCM cells also acquire de novo CTCF (CCCTC-binding factor) binding sites, which are not only strongly associated with TCM-specific hubs but also linked to increased activities of local gene promoters and enhancers. Specific ablation of CTCF in TCM cells impairs rapid induction of genes in cytotoxic program, energy supplies, transcription, and translation by recall stimulation. Therefore, acquisition of CTCF binding and ChrInt hubs by TCM cells serves as a chromatin architectural basis for their transcriptomic dynamics in primary response and for imprinting the code of "recall readiness" against secondary challenge.


Asunto(s)
Linfocitos T CD8-positivos , Cromatina , Cromatina/genética , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Sitios de Unión , Genómica
10.
Nucleic Acids Res ; 51(12): 6020-6038, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37125636

RESUMEN

Cell identity genes are distinct from other genes with respect to the epigenetic mechanisms to activate their transcription, e.g. by super-enhancers and broad H3K4me3 domains. However, it remains unclear whether their post-transcriptional regulation is also unique. We performed a systematic analysis of transcriptome-wide RNA stability in nine cell types and found that unstable transcripts were enriched in cell identity-related pathways while stable transcripts were enriched in housekeeping pathways. Joint analyses of RNA stability and chromatin state revealed significant enrichment of super-enhancers and broad H3K4me3 domains at the gene loci of unstable transcripts. Intriguingly, the RNA m6A methyltransferase, METTL3, preferentially binds to chromatin at super-enhancers, broad H3K4me3 domains and their associated genes. METTL3 binding intensity is positively correlated with RNA m6A methylation and negatively correlated with RNA stability of cell identity genes, probably due to co-transcriptional m6A modifications promoting RNA decay. Nanopore direct RNA-sequencing showed that METTL3 knockdown has a stronger effect on RNA m6A and mRNA stability for cell identity genes. Our data suggest a run-and-brake model, where cell identity genes undergo both frequent transcription and fast RNA decay to achieve precise regulation of RNA expression.


Asunto(s)
Cromatina , Regulación de la Expresión Génica , Estabilidad del ARN , Cromatina/genética , Epigénesis Genética , Metiltransferasas/metabolismo , ARN/química
11.
J Immunol ; 209(12): 2269-2278, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36469845

RESUMEN

T cells are essential for mounting defense against various pathogens and malignantly transformed cells. Thymic development and peripheral T cell differentiation are highly orchestrated biological processes that require precise gene regulation. Higher-order genome organization on multiple scales, in the form of chromatin loops, topologically associating domains and compartments, provides pivotal control of T cell gene expression. CTCF and the cohesin machinery are ubiquitously expressed architectural proteins responsible for establishing chromatin structures. Recent studies indicate that transcription factors, such as T lineage-defining Tcf1 and TCR-induced Batf, may have intrinsic ability and/or engage CTCF to shape chromatin architecture. In this article, we summarize current knowledge on the dynamic changes in genome topology that underlie normal or leukemic T cell development, CD4+ helper T cell differentiation, and CD8+ cytotoxic T cell functions. The knowledge lays a solid foundation for elucidating the causative link of spatial chromatin configuration to transcriptional and functional output in T cells.


Asunto(s)
Proteínas de Ciclo Celular , Cromatina , Factor de Unión a CCCTC/genética , Cromatina/genética , Proteínas de Ciclo Celular/metabolismo , Genoma , Cromosomas , Diferenciación Celular/genética
12.
Cell ; 138(5): 1019-31, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19698979

RESUMEN

Histone acetyltransferases (HATs) and deacetylases (HDACs) function antagonistically to control histone acetylation. As acetylation is a histone mark for active transcription, HATs have been associated with active and HDACs with inactive genes. We describe here genome-wide mapping of HATs and HDACs binding on chromatin and find that both are found at active genes with acetylated histones. Our data provide evidence that HATs and HDACs are both targeted to transcribed regions of active genes by phosphorylated RNA Pol II. Furthermore, the majority of HDACs in the human genome function to reset chromatin by removing acetylation at active genes. Inactive genes that are primed by MLL-mediated histone H3K4 methylation are subject to a dynamic cycle of acetylation and deacetylation by transient HAT/HDAC binding, preventing Pol II from binding to these genes but poising them for future activation. Silent genes without any H3K4 methylation signal show no evidence of being bound by HDACs.


Asunto(s)
Histona Acetiltransferasas/genética , Histona Desacetilasas/genética , Acetilación , Línea Celular , Expresión Génica , Genoma Humano , Histona Acetiltransferasas/metabolismo , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Metilación , Fosforilación , ARN Polimerasa II/metabolismo
13.
Genes Dev ; 30(2): 149-63, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26744420

RESUMEN

Class switch recombination (CSR) diversifies antibodies for productive immune responses while maintaining stability of the B-cell genome. Transcription at the immunoglobulin heavy chain (Igh) locus targets CSR-associated DNA damage and is promoted by the BRCT domain-containing PTIP (Pax transactivation domain-interacting protein). Although PTIP is a unique component of the mixed-lineage leukemia 3 (MLL3)/MLL4 chromatin-modifying complex, the mechanisms for how PTIP promotes transcription remain unclear. Here we dissected the minimal structural requirements of PTIP and its different protein complexes using quantitative proteomics in primary lymphocytes. We found that PTIP functions in transcription and CSR separately from its association with the MLL3/MLL4 complex and from its localization to sites of DNA damage. We identified a tandem BRCT domain of PTIP that is sufficient for CSR and identified PA1 as its main functional protein partner. Collectively, we provide genetic and biochemical evidence that a PTIP-PA1 subcomplex functions independently from the MLL3/MLL4 complex to mediate transcription during CSR. These results further our understanding of how multifunctional chromatin-modifying complexes are organized by subcomplexes that harbor unique and distinct activities.


Asunto(s)
Proteínas Portadoras/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Proteínas Nucleares/metabolismo , Daño del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica/inmunología , Estructura Molecular , Estructura Terciaria de Proteína , Transporte de Proteínas
14.
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
15.
Immunity ; 37(6): 971-85, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23159437

RESUMEN

Histone 3 lysine 4 trimethylation (H3K4me3) is associated with promoters of active genes and found at hot spots for DNA recombination. Here we have shown that PAXIP1 (also known as PTIP), a protein associated with MLL3 and MLL4 methyltransferase and the DNA damage response, regulates RAG-mediated cleavage and repair during V(D)J recombination in CD4(+) CD8(+) DP thymocytes. Loss of PAXIP1 in developing thymocytes diminished Jα H3K4me3 and germline transcription, suppressed double strand break formation at 3' Jα segments, but resulted in accumulation of unresolved T cell receptor α-chain gene (Tcra) breaks. Moreover, PAXIP1 was essential for release of mature single positive (SP) αß T cells from the thymus through transcriptional activation of sphingosine-1-phosphate receptor S1pr1 as well as for natural killer T cell development. Thus, in addition to maintaining genome integrity during Tcra rearrangements, PAXIP1 controls distinct transcriptional programs during DP differentiation necessary for Tcra locus accessibility, licensing mature thymocytes for trafficking and natural killer T cell development.


Asunto(s)
Proteínas Portadoras/genética , Diferenciación Celular , Movimiento Celular , Daño del ADN , Regulación de la Expresión Génica , Proteínas Nucleares/genética , Timocitos/citología , Timocitos/inmunología , Animales , Proteínas Portadoras/metabolismo , Linaje de la Célula/genética , Movimiento Celular/genética , Proteínas de Unión al ADN , Histonas/metabolismo , Ratones , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Lisoesfingolípidos/genética , Recombinación Genética , Receptores de Esfingosina-1-Fosfato , Linfocitos T/citología , Linfocitos T/metabolismo , Timocitos/metabolismo , Transcripción Genética
16.
Immunity ; 32(6): 840-51, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20620946

RESUMEN

Signal transducer and activator of transcription 4 (STAT4) and STAT6 are key factors in the specification of helper T cells; however, their direct roles in driving differentiation are not well understood. Using chromatin immunoprecipitation and massive parallel sequencing, we quantitated the full complement of STAT-bound genes, concurrently assessing global STAT-dependent epigenetic modifications and gene transcription by using cells from cognate STAT-deficient mice. STAT4 and STAT6 each bound over 4000 genes with distinct binding motifs. Both played critical roles in maintaining chromatin configuration and transcription of a core subset of genes through the combination of different epigenetic patterns. Globally, STAT4 had a more dominant role in promoting active epigenetic marks, whereas STAT6 had a more prominent role in antagonizing repressive marks. Clusters of genes negatively regulated by STATs were also identified, highlighting previously unappreciated repressive roles of STATs. Therefore, STAT4 and STAT6 play wide regulatory roles in T helper cell specification.


Asunto(s)
Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Factor de Transcripción STAT4/inmunología , Factor de Transcripción STAT6/inmunología , Linfocitos T Colaboradores-Inductores/citología , Animales , Inmunoprecipitación de Cromatina , Epigénesis Genética , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT6/genética , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Transcripción Genética
17.
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
18.
Cell Mol Biol (Noisy-le-grand) ; 65(7): 123-126, 2019 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31880529

RESUMEN

The aim of this study is to investigate  the effect of mesenchymal stem cell (MSC)-derived exosomes on diabetes mellitus-induced myocardial injury, and the underlying mechanism. Thirty adult male Sprague Dawley rats were randomly assigned to three groups of ten rats each: normal control group, diabetic control group and MSC exosomes group. Exosomes were isolated from MSCs through gradient ultracentrifugation. With the exception of normal control, diabetes mellitus (DM) was induced in the rats with a single intraperitoneal injection of 30 mg/kg body weight streptozotocin (STZ) in 0.1 mol/L sodium citrate buffer. Rats in MSC exosomes group were intravenously injected with MSC-derived exosomes once a week for 12 weeks. Left ventricular collagen (LVC) level was measured using acid hydrolysis method. Fatty acid transporters (FATPs) and fatty acid beta oxidase (FA-ß-oxidase) were determined using enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions of TGF-ß and Smad2 were determined using real-time quantitative polymerase chain reaction (qRT-PCR) and Western blotting. Flow cytometric analysis and Western blotting revealed positive expression of exosomal specific marker, CD63. The level of LVC was significantly higher in diabetic control group than in normal control group, but was significantly reduced after treatment with MSC-derived exosomes (p < 0.05). The levels of FATPs and FA-ß-oxidase were significantly lower in diabetic control group than in normal control group (p < 0.05). However, treatment with MSC-derived exosomes significantly increased the levels of these proteins (p < 0.05). The levels of expression of TGF-ß1 and Smad2 mRNAs were significantly higher in the diabetic control group than in normal control group, but were significantly reduced after treatment with MSC-derived exosomes (p < 0.05). The expressions of TGF-ß1 and Smad2 proteins were also significantly upregulated in diabetic control group, when compared with normal control group (p < 0.05). However, treatment with MSC-derived exosomes significantly down-regulated the expression of these proteins (p < 0.05). The results obtained in this study indicate that MSC-derived exosomes improve DM-induced myocardial injury and fibrosis via inhibition of TGF-ß1/Smad2 signaling pathway.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Exosomas/metabolismo , Fibrosis/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Exosomas/fisiología , Fibrosis/etiología , Masculino , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Nature ; 499(7457): 223-7, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23823717

RESUMEN

The variant antigen Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), which is expressed on the surface of P. falciparum-infected red blood cells, is a critical virulence factor for malaria. Each parasite has 60 antigenically distinct var genes that each code for a different PfEMP1 protein. During infection the clonal parasite population expresses only one gene at a time before switching to the expression of a new variant antigen as an immune-evasion mechanism to avoid the host antibody response. The mechanism by which 59 of the 60 var genes are silenced remains largely unknown. Here we show that knocking out the P. falciparum variant-silencing SET gene (here termed PfSETvs), which encodes an orthologue of Drosophila melanogaster ASH1 and controls histone H3 lysine 36 trimethylation (H3K36me3) on var genes, results in the transcription of virtually all var genes in the single parasite nuclei and their expression as proteins on the surface of individual infected red blood cells. PfSETvs-dependent H3K36me3 is present along the entire gene body, including the transcription start site, to silence var genes. With low occupancy of PfSETvs at both the transcription start site of var genes and the intronic promoter, expression of var genes coincides with transcription of their corresponding antisense long noncoding RNA. These results uncover a previously unknown role of PfSETvs-dependent H3K36me3 in silencing var genes in P. falciparum that might provide a general mechanism by which orthologues of PfSETvs repress gene expression in other eukaryotes. PfSETvs knockout parasites expressing all PfEMP1 proteins may also be applied to the development of a malaria vaccine.


Asunto(s)
Silenciador del Gen , Histonas/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Factores de Virulencia/genética , Proteínas de Unión al ADN , Proteínas de Drosophila , Eritrocitos/citología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Genes Protozoarios/genética , Histonas/química , Intrones/genética , Lisina/metabolismo , Vacunas contra la Malaria/genética , Metilación , Plasmodium falciparum/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/genética , ARN Largo no Codificante/genética , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Virulencia/genética
20.
Nucleic Acids Res ; 45(11): 6388-6403, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28398509

RESUMEN

Histone H3K4me1/2 methyltransferases MLL3/MLL4 and H3K27 acetyltransferases CBP/p300 are major enhancer epigenomic writers. To understand how these epigenomic writers orchestrate enhancer landscapes in cell differentiation, we have profiled genomic binding of MLL4, CBP, lineage-determining transcription factors (EBF2, C/EBPß, C/EBPα, PPARγ), coactivator MED1, RNA polymerase II, as well as epigenome (H3K4me1/2/3, H3K9me2, H3K27me3, H3K36me3, H3K27ac), transcriptome and chromatin opening during adipogenesis of immortalized preadipocytes derived from mouse brown adipose tissue (BAT). We show that MLL4 and CBP drive the dynamic enhancer epigenome, which correlates with the dynamic transcriptome. MLL3/MLL4 are required for CBP/p300 binding on enhancers activated during adipogenesis. Further, MLL4 and CBP identify super-enhancers (SEs) of adipogenesis and that MLL3/MLL4 are required for SE formation. Finally, in brown adipocytes differentiated in culture, MLL4 identifies primed SEs of genes fully activated in BAT such as Ucp1. Comparison of MLL4-defined SEs in brown and white adipogenesis identifies brown-specific SE-associated genes that could be involved in BAT functions. These results establish MLL3/MLL4 and CBP/p300 as master enhancer epigenomic writers and suggest that enhancer-priming by MLL3/MLL4 followed by enhancer-activation by CBP/p300 sequentially shape dynamic enhancer landscapes during cell differentiation. Our data also provide a rich resource for understanding epigenomic regulation of brown adipogenesis.


Asunto(s)
Adipogénesis , Proteína de Unión a CREB/metabolismo , Proteína p300 Asociada a E1A/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Adipocitos Marrones/fisiología , Tejido Adiposo Pardo/citología , Animales , Células Cultivadas , Elementos de Facilitación Genéticos , Ratones Transgénicos , Unión Proteica , Activación Transcripcional , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA