Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(4): 914-930.e20, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38280375

RESUMEN

The gut and liver are recognized to mutually communicate through the biliary tract, portal vein, and systemic circulation. However, it remains unclear how this gut-liver axis regulates intestinal physiology. Through hepatectomy and transcriptomic and proteomic profiling, we identified pigment epithelium-derived factor (PEDF), a liver-derived soluble Wnt inhibitor, which restrains intestinal stem cell (ISC) hyperproliferation to maintain gut homeostasis by suppressing the Wnt/ß-catenin signaling pathway. Furthermore, we found that microbial danger signals resulting from intestinal inflammation can be sensed by the liver, leading to the repression of PEDF production through peroxisome proliferator-activated receptor-α (PPARα). This repression liberates ISC proliferation to accelerate tissue repair in the gut. Additionally, treating mice with fenofibrate, a clinical PPARα agonist used for hypolipidemia, enhances colitis susceptibility due to PEDF activity. Therefore, we have identified a distinct role for PEDF in calibrating ISC expansion for intestinal homeostasis through reciprocal interactions between the gut and liver.


Asunto(s)
Intestinos , Hígado , Animales , Ratones , Proliferación Celular , Hígado/metabolismo , PPAR alfa/metabolismo , Proteómica , Células Madre/metabolismo , Vía de Señalización Wnt , Intestinos/citología , Intestinos/metabolismo
2.
Cell ; 176(1-2): 213-226.e18, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30554876

RESUMEN

Transcriptional regulation in metazoans occurs through long-range genomic contacts between enhancers and promoters, and most genes are transcribed in episodic "bursts" of RNA synthesis. To understand the relationship between these two phenomena and the dynamic regulation of genes in response to upstream signals, we describe the use of live-cell RNA imaging coupled with Hi-C measurements and dissect the endogenous regulation of the estrogen-responsive TFF1 gene. Although TFF1 is highly induced, we observe short active periods and variable inactive periods ranging from minutes to days. The heterogeneity in inactive times gives rise to the widely observed "noise" in human gene expression and explains the distribution of protein levels in human tissue. We derive a mathematical model of regulation that relates transcription, chromosome structure, and the cell's ability to sense changes in estrogen and predicts that hypervariability is largely dynamic and does not reflect a stable biological state.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Transcripción Genética/fisiología , Receptor alfa de Estrógeno/metabolismo , Estrógenos , Expresión Génica/genética , Humanos , Modelos Teóricos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Análisis de la Célula Individual/métodos , Transcripción Genética/genética , Activación Transcripcional/fisiología , Factor Trefoil-1/genética
3.
Immunity ; 57(5): 987-1004.e5, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38614090

RESUMEN

The development and function of the immune system are controlled by temporospatial gene expression programs, which are regulated by cis-regulatory elements, chromatin structure, and trans-acting factors. In this study, we cataloged the dynamic histone modifications and chromatin interactions at regulatory regions during T helper (Th) cell differentiation. Our data revealed that the H3K4me1 landscape established by MLL4 in naive CD4+ T cells is critical for restructuring the regulatory interaction network and orchestrating gene expression during the early phase of Th differentiation. GATA3 plays a crucial role in further configuring H3K4me1 modification and the chromatin interaction network during Th2 differentiation. Furthermore, we demonstrated that HSS3-anchored chromatin loops function to restrict the activity of the Th2 locus control region (LCR), thus coordinating the expression of Th2 cytokines. Our results provide insights into the mechanisms of how the interplay between histone modifications, chromatin looping, and trans-acting factors contributes to the differentiation of Th cells.


Asunto(s)
Diferenciación Celular , Cromatina , Código de Histonas , Histonas , Células Th2 , Diferenciación Celular/inmunología , Animales , Cromatina/metabolismo , Ratones , Células Th2/inmunología , Histonas/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Región de Control de Posición , Citocinas/metabolismo
4.
Immunity ; 56(5): 944-958.e6, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37040761

RESUMEN

Interferon-γ (IFN-γ) is a key cytokine in response to viral or intracellular bacterial infection in mammals. While a number of enhancers are described to promote IFN-γ responses, to the best of our knowledge, no silencers for the Ifng gene have been identified. By examining H3K4me1 histone modification in naive CD4+ T cells within Ifng locus, we identified a silencer (CNS-28) that restrains Ifng expression. Mechanistically, CNS-28 maintains Ifng silence by diminishing enhancer-promoter interactions within Ifng locus in a GATA3-dependent but T-bet-independent manner. Functionally, CNS-28 restrains Ifng transcription in NK cells, CD4+ cells, and CD8+ T cells during both innate and adaptive immune responses. Moreover, CNS-28 deficiency resulted in repressed type 2 responses due to elevated IFN-γ expression, shifting Th1 and Th2 paradigm. Thus, CNS-28 activity ensures immune cell quiescence by cooperating with other regulatory cis elements within the Ifng gene locus to minimize autoimmunity.


Asunto(s)
Linfocitos T CD8-positivos , Interferón gamma , Animales , Interferón gamma/genética , Interferón gamma/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Secuencias Reguladoras de Ácidos Nucleicos , Homeostasis , Células TH1 , Mamíferos
5.
Nat Immunol ; 25(3): 390-391, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38356060
6.
Nat Immunol ; 20(9): 1150-1160, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31358996

RESUMEN

Innate lymphoid cells (ILCs) play important functions in immunity and tissue homeostasis, but their development is poorly understood. Through the use of single-cell approaches, we examined the transcriptional and functional heterogeneity of ILC progenitors, and studied the precursor-product relationships that link the subsets identified. This analysis identified two successive stages of ILC development within T cell factor 1-positive (TCF-1+) early innate lymphoid progenitors (EILPs), which we named 'specified EILPs' and 'committed EILPs'. Specified EILPs generated dendritic cells, whereas this potential was greatly decreased in committed EILPs. TCF-1 was dispensable for the generation of specified EILPs, but required for the generation of committed EILPs. TCF-1 used a pre-existing regulatory landscape established in upstream lymphoid precursors to bind chromatin in EILPs. Our results provide insight into the mechanisms by which TCF-1 promotes developmental progression of ILC precursors, while constraining their dendritic cell lineage potential and enforcing commitment to ILC fate.


Asunto(s)
Linaje de la Célula/inmunología , Células Dendríticas/citología , Factor Nuclear 1-alfa del Hepatocito/inmunología , Células Progenitoras Linfoides/citología , Linfocitos T/citología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Regulación de la Expresión Génica/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética
7.
Immunity ; 55(4): 639-655.e7, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35381213

RESUMEN

Adaptive CD4+ T helper cells and their innate counterparts, innate lymphoid cells, utilize an identical set of transcription factors (TFs) for their differentiation and functions. However, similarities and differences in the induction of these TFs in related lymphocytes are still elusive. Here, we show that T helper-1 (Th1) cells and natural killer (NK) cells displayed distinct epigenomes at the Tbx21 locus, which encodes T-bet, a critical TF for regulating type 1 immune responses. The initial induction of T-bet in NK precursors was dependent on the NK-specific DNase I hypersensitive site Tbx21-CNS-3, and the expression of the interleukin-18 (IL-18) receptor; IL-18 induced T-bet expression through the transcription factor RUNX3, which bound to Tbx21-CNS-3. By contrast, signal transducer and activator of transcription (STAT)-binding motifs within Tbx21-CNS-12 were critical for IL-12-induced T-bet expression during Th1 cell differentiation both in vitro and in vivo. Thus, type 1 innate and adaptive lymphocytes utilize distinct enhancer elements for their development and differentiation.


Asunto(s)
Inmunidad Innata , Interleucina-18 , Células Asesinas Naturales , Células TH1 , Diferenciación Celular , Interleucina-18/metabolismo , Células Asesinas Naturales/inmunología , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Factores de Transcripción/metabolismo
8.
Immunity ; 55(8): 1402-1413.e4, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35882235

RESUMEN

The differentiation of innate lymphoid cells (ILCs) from hematopoietic stem cells needs to go through several multipotent progenitor stages. However, it remains unclear whether the fates of multipotent progenitors are predefined by epigenetic states. Here, we report the identification of distinct accessible chromatin regions in all lymphoid progenitors (ALPs), EILPs, and ILC precursors (ILCPs). Single-cell MNase-seq analyses revealed that EILPs contained distinct subpopulations epigenetically primed toward either dendritic cell lineages or ILC lineages. We found that TCF-1 and GATA3 co-bound to the lineage-defining sites for ILCs (LDS-Is), whereas PU.1 binding was enriched in the LDSs for alternative dendritic cells (LDS-As). TCF-1 and GATA3 were indispensable for the epigenetic priming of LDSs at the EILP stage. Our results suggest that the multipotency of progenitor cells is defined by the existence of a heterogeneous population of cells epigenetically primed for distinct downstream lineages, which are regulated by key transcription factors.


Asunto(s)
Inmunidad Innata , Linfocitos , Diferenciación Celular , Linaje de la Célula , Epigénesis Genética , Células Madre Hematopoyéticas
9.
Cell ; 165(6): 1375-1388, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259149

RESUMEN

How the chromatin regulatory landscape in the inner cell mass cells is established from differentially packaged sperm and egg genomes during preimplantation development is unknown. Here, we develop a low-input DNase I sequencing (liDNase-seq) method that allows us to generate maps of DNase I-hypersensitive site (DHS) of mouse preimplantation embryos from 1-cell to morula stage. The DHS landscape is progressively established with a drastic increase at the 8-cell stage. Paternal chromatin accessibility is quickly reprogrammed after fertilization to the level similar to maternal chromatin, while imprinted genes exhibit allelic accessibility bias. We demonstrate that transcription factor Nfya contributes to zygotic genome activation and DHS formation at the 2-cell stage and that Oct4 contributes to the DHSs gained at the 8-cell stage. Our study reveals the dynamic chromatin regulatory landscape during early development and identifies key transcription factors important for DHS establishment in mammalian embryos.


Asunto(s)
Blastocisto , Cromatina/metabolismo , Animales , Sitios de Unión , Blastocisto/citología , Masa Celular Interna del Blastocisto/metabolismo , Factor de Unión a CCAAT/metabolismo , Mapeo Cromosómico , ADN/metabolismo , Desoxirribonucleasa I/metabolismo , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas
10.
Cell ; 165(2): 357-71, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058666

RESUMEN

We report a mechanism through which the transcription machinery directly controls topoisomerase 1 (TOP1) activity to adjust DNA topology throughout the transcription cycle. By comparing TOP1 occupancy using chromatin immunoprecipitation sequencing (ChIP-seq) versus TOP1 activity using topoisomerase 1 sequencing (TOP1-seq), a method reported here to map catalytically engaged TOP1, TOP1 bound at promoters was discovered to become fully active only after pause-release. This transition coupled the phosphorylation of the carboxyl-terminal-domain (CTD) of RNA polymerase II (RNAPII) with stimulation of TOP1 above its basal rate, enhancing its processivity. TOP1 stimulation is strongly dependent on the kinase activity of BRD4, a protein that phosphorylates Ser2-CTD and regulates RNAPII pause-release. Thus the coordinated action of BRD4 and TOP1 overcame the torsional stress opposing transcription as RNAPII commenced elongation but preserved negative supercoiling that assists promoter melting at start sites. This nexus between transcription and DNA topology promises to elicit new strategies to intercept pathological gene expression.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , ADN/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , ADN/química , ADN-Topoisomerasas de Tipo I/genética , Técnicas de Silenciamiento del Gen , Humanos , Regiones Promotoras Genéticas , ARN Polimerasa II/química , ARN Polimerasa II/aislamiento & purificación , Elongación de la Transcripción Genética , Factores de Transcripción/aislamiento & purificación , Sitio de Iniciación de la Transcripción
11.
Nat Immunol ; 24(10): 1602-1603, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37709987
12.
Nat Immunol ; 18(9): 1035-1045, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28759003

RESUMEN

MLL4 is an essential subunit of the histone H3 Lys4 (H3K4)-methylation complexes. We found that MLL4 deficiency compromised the development of regulatory T cells (Treg cells) and resulted in a substantial decrease in monomethylated H3K4 (H3K4me1) and chromatin interaction at putative gene enhancers, a considerable portion of which were not direct targets of MLL4 but were enhancers that interacted with MLL4-bound sites. The decrease in H3K4me1 and chromatin interaction at the enhancers not bound by MLL4 correlated with MLL4 binding at distant interacting regions. Deletion of an upstream MLL4-binding site diminished the abundance of H3K4me1 at the regulatory elements of the gene encoding the transcription factor Foxp3 that were looped to the MLL4-binding site and compromised both the thymic differentiation and the inducible differentiation of Treg cells. We found that MLL4 catalyzed methylation of H3K4 at distant unbound enhancers via chromatin looping, which identifies a previously unknown mechanism for regulating the T cell enhancer landscape and affecting Treg cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Cromatina/metabolismo , Factores de Transcripción Forkhead/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Linfocitos T Reguladores , Animales , Sistemas CRISPR-Cas , Citocinas/inmunología , Citometría de Flujo , Regulación de la Expresión Génica , Immunoblotting , Técnicas In Vitro , Metilación , Ratones
13.
Immunity ; 52(1): 83-95.e4, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31882362

RESUMEN

Lymphoid tissue inducer (LTi) cells are regarded as a subset of innate lymphoid cells (ILCs). However, these cells are not derived from the ILC common progenitor, which generates other ILC subsets and is defined by the expression of the transcription factor PLZF. Here, we examined transcription factor(s) determining the fate of LTi progenitors versus non-LTi ILC progenitors. Conditional deletion of Gata3 resulted in the loss of PLZF+ non-LTi progenitors but not the LTi progenitors that expressed the transcription factor RORγt. Consistently, PLZF+ non-LTi progenitors expressed high amounts of GATA3, whereas GATA3 expression was low in RORγt+ LTi progenitors. The generation of both progenitors required the transcriptional regulator Id2, which defines the common helper-like innate lymphoid progenitor (ChILP), but not cytokine signaling. Nevertheless, low GATA3 expression was necessary for the generation of functionally mature LTi cells. Thus, differential expression of GATA3 determines the fates and functions of distinct ILC progenitors.


Asunto(s)
Factor de Transcripción GATA3/biosíntesis , Células Madre/citología , Subgrupos de Linfocitos T/citología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linaje de la Célula/inmunología , Células Cultivadas , Factor de Transcripción GATA3/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Receptor de Muerte Celular Programada 1/biosíntesis , Proteína de la Leucemia Promielocítica con Dedos de Zinc/biosíntesis , Células Madre/inmunología , Subgrupos de Linfocitos T/inmunología
14.
Cell ; 159(2): 374-387, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303531

RESUMEN

The pluripotent state of embryonic stem cells (ESCs) is produced by active transcription of genes that control cell identity and repression of genes encoding lineage-specifying developmental regulators. Here, we use ESC cohesin ChIA-PET data to identify the local chromosomal structures at both active and repressed genes across the genome. The results produce a map of enhancer-promoter interactions and reveal that super-enhancer-driven genes generally occur within chromosome structures that are formed by the looping of two interacting CTCF sites co-occupied by cohesin. These looped structures form insulated neighborhoods whose integrity is important for proper expression of local genes. We also find that repressed genes encoding lineage-specifying developmental regulators occur within insulated neighborhoods. These results provide insights into the relationship between transcriptional control of cell identity genes and control of local chromosome structure.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Animales , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/metabolismo , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/citología , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Especificidad de Órganos , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Análisis de Secuencia de ADN , Cohesinas
15.
Nat Immunol ; 17(2): 169-78, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26595886

RESUMEN

The transcription factor GATA-3 is indispensable for the development of all innate lymphoid cells (ILCs) that express the interleukin 7 receptor α-chain (IL-7Rα). However, the function of low GATA-3 expression in committed group 3 ILCs (ILC3 cells) has not been identified. We found that GATA-3 regulated the homeostasis of ILC3 cells by controlling IL-7Rα expression. In addition, GATA-3 served a critical function in the development of the NKp46(+) ILC3 subset by regulating the balance between the transcription factors T-bet and RORγt. Among NKp46(+) ILC3 cells, although GATA-3 positively regulated genes specific to the NKp46(+) ILC3 subset, it negatively regulated genes specific to lymphoid tissue-inducer (LTi) or LTi-like ILC3 cells. Furthermore, GATA-3 was required for IL-22 production in both ILC3 subsets. Thus, despite its low expression, GATA-3 was critical for the homeostasis, development and function of ILC3 subsets.


Asunto(s)
Diferenciación Celular , Factor de Transcripción GATA3/metabolismo , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Análisis por Conglomerados , Factor de Transcripción GATA3/deficiencia , Factor de Transcripción GATA3/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Homeostasis , Inmunidad Innata/genética , Inmunofenotipificación , Interleucinas/biosíntesis , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fenotipo , Unión Proteica , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Proteínas de Dominio T Box/metabolismo , Interleucina-22
16.
Development ; 151(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39007366

RESUMEN

Many tissue-specific adult stem cell lineages maintain a balance between proliferation and differentiation. Here, we study how the H3K4me3 methyltransferase Set1 regulates early-stage male germ cells in Drosophila. Early-stage germline-specific knockdown of Set1 results in temporally progressive defects, arising as germ cell loss and developing into overpopulated early-stage germ cells. These germline defects also impact the niche architecture and cyst stem cell lineage non-cell-autonomously. Additionally, wild-type Set1, but not the catalytically inactive Set1, rescues the Set1 knockdown phenotypes, highlighting the functional importance of the methyltransferase activity of Set1. Further, RNA-sequencing experiments reveal key signaling pathway components, such as the JAK-STAT pathway gene Stat92E and the BMP pathway gene Mad, which are upregulated upon Set1 knockdown. Genetic interaction assays support the functional relationships between Set1 and JAK-STAT or BMP pathways, as both Stat92E and Mad mutations suppress the Set1 knockdown phenotypes. These findings enhance our understanding of the balance between proliferation and differentiation in an adult stem cell lineage. The phenotype of germ cell loss followed by over-proliferation when inhibiting a histone methyltransferase also raises concerns about using their inhibitors in cancer therapy.


Asunto(s)
Diferenciación Celular , Proteínas de Drosophila , Drosophila melanogaster , Células Germinativas , N-Metiltransferasa de Histona-Lisina , Transducción de Señal , Animales , Masculino , Diferenciación Celular/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Transducción de Señal/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Células Germinativas/metabolismo , Células Germinativas/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Células Madre/metabolismo , Células Madre/citología , Factores de Transcripción STAT/metabolismo , Factores de Transcripción STAT/genética , Quinasas Janus/metabolismo , Quinasas Janus/genética , Proliferación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica
17.
Nat Immunol ; 16(10): 1077-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26322481

RESUMEN

The molecular mechanisms by which signaling via transforming growth factor-ß (TGF-ß) and interleukin 4 (IL-4) control the differentiation of CD4(+) IL-9-producing helper T cells (TH9 cells) remain incompletely understood. We found here that the DNA-binding inhibitor Id3 regulated TH9 differentiation, as deletion of Id3 increased IL-9 production from CD4(+) T cells. Mechanistically, TGF-ß1 and IL-4 downregulated Id3 expression, and this process required the kinase TAK1. A reduction in Id3 expression enhanced binding of the transcription factors E2A and GATA-3 to the Il9 promoter region, which promoted Il9 transcription. Notably, Id3-mediated control of TH9 differentiation regulated anti-tumor immunity in an experimental melanoma-bearing model in vivo and also in human CD4(+) T cells in vitro. Thus, our study reveals a previously unrecognized TAK1-Id3-E2A-GATA-3 pathway that regulates TH9 differentiation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteínas Inhibidoras de la Diferenciación/inmunología , Interleucina-9/biosíntesis , Proteínas de Neoplasias/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Citometría de Flujo , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , Interleucina-9/inmunología , Ratones , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa , Transducción de Señal/inmunología
18.
Immunity ; 48(2): 227-242.e8, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466755

RESUMEN

How chromatin reorganization coordinates differentiation and lineage commitment from hematopoietic stem and progenitor cells (HSPCs) to mature immune cells has not been well understood. Here, we carried out an integrative analysis of chromatin accessibility, topologically associating domains, AB compartments, and gene expression from HSPCs to CD4+CD8+ T cells. We found that abrupt genome-wide changes at all three levels of chromatin organization occur during the transition from double-negative stage 2 (DN2) to DN3, accompanying the T lineage commitment. The transcription factor BCL11B, a critical regulator of T cell commitment, is associated with increased chromatin interaction, and Bcl11b deletion compromised chromatin interaction at its target genes. We propose that these large-scale and concerted changes in chromatin organization present an energy barrier to prevent the cell from reversing its fate to earlier stages or redirecting to alternatives and thus lock the cell fate into the T lineages.


Asunto(s)
Linaje de la Célula , Núcleo Celular/fisiología , Cromatina/fisiología , Linfocitos T/fisiología , Animales , Diferenciación Celular , Humanos , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/fisiología
19.
Nat Rev Genet ; 22(4): 235-250, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33244170

RESUMEN

Single-cell sequencing-based methods for profiling gene transcript levels have revealed substantial heterogeneity in expression levels among morphologically indistinguishable cells. This variability has important functional implications for tissue biology and disease states such as cancer. Mapping of epigenomic information such as chromatin accessibility, nucleosome positioning, histone tail modifications and enhancer-promoter interactions in both bulk-cell and single-cell samples has shown that these characteristics of chromatin state contribute to expression or repression of associated genes. Advances in single-cell epigenomic profiling methods are enabling high-resolution mapping of chromatin states in individual cells. Recent studies using these techniques provide evidence that variations in different aspects of chromatin organization collectively define gene expression heterogeneity among otherwise highly similar cells.


Asunto(s)
Cromatina/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Heterogeneidad Genética , Linaje de la Célula/genética , Biología Computacional , Histonas/genética , Humanos , Regiones Promotoras Genéticas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Secuencia de ADN
20.
Cell ; 151(3): 576-89, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23101626

RESUMEN

Embryonic stem cell (ESC) pluripotency requires bivalent epigenetic modifications of key developmental genes regulated by various transcription factors and chromatin-modifying enzymes. How these factors coordinate with one another to maintain the bivalent chromatin state so that ESCs can undergo rapid self-renewal while retaining pluripotency is poorly understood. We report that Utf1, a target of Oct4 and Sox2, is a bivalent chromatin component that buffers poised states of bivalent genes. By limiting PRC2 loading and histone 3 lysine-27 trimethylation, Utf1 sets proper activation thresholds for bivalent genes. It also promotes nuclear tagging of messenger RNAs (mRNAs) transcribed from insufficiently silenced bivalent genes for cytoplasmic degradation through mRNA decapping. These opposing functions of Utf1 promote coordinated differentiation. The mRNA degradation function also ensures rapid cell proliferation by blocking the Myc-Arf feedback control. Thus, Utf1 couples the core pluripotency factors with Myc and PRC2 networks to promote the pluripotency and proliferation of ESCs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Mensajero/metabolismo , Transactivadores/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Epigénesis Genética , Humanos , Células Madre Pluripotentes/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA