Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Cell ; 74(3): 542-554.e5, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30905509

RESUMEN

Developing and adult tissues use different cis-regulatory elements. Although DNA at some decommissioned embryonic enhancers is hypomethylated in adult cells, it is unknown whether this putative epigenetic memory is complete and recoverable. We find that, in adult mouse cells, hypomethylated CpG dinucleotides preserve a nearly complete archive of tissue-specific developmental enhancers. Sites that carry the active histone mark H3K4me1, and are therefore considered "primed," are mainly cis elements that act late in organogenesis. In contrast, sites decommissioned early in development retain hypomethylated DNA as a singular property. In adult intestinal and blood cells, sustained absence of polycomb repressive complex 2 indirectly reactivates most-and only-hypomethylated developmental enhancers. Embryonic and fetal transcriptional programs re-emerge as a result, in reverse chronology to cis element inactivation during development. Thus, hypomethylated DNA in adult cells preserves a "fossil record" of tissue-specific developmental enhancers, stably marking decommissioned sites and enabling recovery of this epigenetic memory.


Asunto(s)
Metilación de ADN/genética , Elementos de Facilitación Genéticos/genética , Epigenómica , Histonas/genética , Animales , Regulación del Desarrollo de la Expresión Génica/genética , Ratones
2.
Clin Immunol ; 263: 110223, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636890

RESUMEN

Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.


Asunto(s)
Anemia Aplásica , Linfocitos T CD8-positivos , Proteínas Ligadas a GPI , Células Madre Hematopoyéticas , Interleucina-15 , Monocitos , Receptores de IgG , Humanos , Anemia Aplásica/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Interleucina-15/farmacología , Interleucina-15/inmunología , Receptores de IgG/metabolismo , Receptores de IgG/inmunología , Monocitos/inmunología , Monocitos/efectos de los fármacos , Femenino , Masculino , Adulto , Células Madre Hematopoyéticas/inmunología , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/inmunología , Persona de Mediana Edad , Proteína Ligando Fas/metabolismo , Proteína Ligando Fas/inmunología , Adulto Joven , Adolescente , Interferón gamma/inmunología , Interferón gamma/metabolismo , Receptores de Interleucina-15/metabolismo , Receptores de Interleucina-15/inmunología , Apoptosis/efectos de los fármacos , Diferenciación Celular/inmunología
3.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34373913

RESUMEN

Neutrophils are the most abundant vertebrate leukocytes and they are essential to host defense. Despite extensive investigation, the molecular network controlling neutrophil differentiation remains incompletely understood. GFI1 is associated with several myeloid disorders, but its role and the role of its co-regulators in granulopoiesis and pathogenesis are far from clear. Here, we demonstrate that zebrafish gfi1aa deficiency induces excessive neutrophil progenitor proliferation, accumulation of immature neutrophils from the embryonic stage, and some phenotypes similar to myelodysplasia syndrome in adulthood. Both genetic and epigenetic analyses demonstrate that immature neutrophil accumulation in gfi1aa-deficient mutants is due to upregulation of cebpa transcription. Increased transcription was associated with Lsd1-altered H3K4 methylation of the cebpa regulatory region. Taken together, our results demonstrate that Gfi1aa, Lsd1 and cebpa form a regulatory network that controls neutrophil development, providing a disease progression-traceable model for myelodysplasia syndrome. Use of this model could provide new insights into the molecular mechanisms underlying GFI1-related myeloid disorders as well as a means by which to develop targeted therapeutic approaches for treatment.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Hematopoyesis/genética , Histona Demetilasas/metabolismo , Neutrófilos/citología , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Embrión no Mamífero , Epigénesis Genética , Células Precursoras de Granulocitos/citología , Células Precursoras de Granulocitos/metabolismo , Histona Demetilasas/genética , Neutrófilos/metabolismo , Pez Cebra , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
4.
Nature ; 543(7644): 270-274, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28241139

RESUMEN

Recurrent chromosomal translocations producing a chimaeric MLL oncogene give rise to a highly aggressive acute leukaemia associated with poor clinical outcome. The preferential involvement of chromatin-associated factors as MLL fusion partners belies a dependency on transcription control. Despite recent progress made in targeting chromatin regulators in cancer, available therapies for this well-characterized disease remain inadequate, prompting the need to identify new targets for therapeutic intervention. Here, using unbiased CRISPR-Cas9 technology to perform a genome-scale loss-of-function screen in an MLL-AF4-positive acute leukaemia cell line, we identify ENL as an unrecognized gene that is specifically required for proliferation in vitro and in vivo. To explain the mechanistic role of ENL in leukaemia pathogenesis and dynamic transcription control, a chemical genetic strategy was developed to achieve targeted protein degradation. Acute loss of ENL suppressed the initiation and elongation of RNA polymerase II at active genes genome-wide, with pronounced effects at genes featuring a disproportionate ENL load. Notably, an intact YEATS chromatin-reader domain was essential for ENL-dependent leukaemic growth. Overall, these findings identify a dependency factor in acute leukaemia and suggest a mechanistic rationale for disrupting the YEATS domain in disease.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Leucemia/genética , Leucemia/metabolismo , Dominios Proteicos , Transcripción Genética , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Edición Génica , Genoma/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Leucemia/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteolisis , ARN Polimerasa II/metabolismo , Elongación de la Transcripción Genética , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Elongación Transcripcional/genética
5.
Mol Cell ; 57(2): 304-316, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25578878

RESUMEN

Polycomb repressive complex 2 (PRC2) plays crucial roles in transcriptional regulation and stem cell development. However, the context-specific functions associated with alternative subunits remain largely unexplored. Here we show that the related enzymatic subunits EZH1 and EZH2 undergo an expression switch during blood cell development. An erythroid-specific enhancer mediates transcriptional activation of EZH1, and a switch from GATA2 to GATA1 controls the developmental EZH1/2 switch by differential association with EZH1 enhancers. We further examine the in vivo stoichiometry of the PRC2 complexes by quantitative proteomics and reveal the existence of an EZH1-SUZ12 subcomplex lacking EED. EZH1 together with SUZ12 form a non-canonical PRC2 complex, occupy active chromatin, and positively regulate gene expression. Loss of EZH2 expression leads to repositioning of EZH1 to EZH2 targets. Thus, the lineage- and developmental stage-specific regulation of PRC2 subunit composition leads to a switch from canonical silencing to non-canonical functions during blood stem cell specification.


Asunto(s)
Factores de Transcripción GATA/fisiología , Complejo Represivo Polycomb 2/metabolismo , Secuencia de Bases , Carcinogénesis , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Células Eritroides/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas , Histonas/metabolismo , Humanos , Células K562 , Metilación , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Subunidades de Proteína
6.
Proc Natl Acad Sci U S A ; 117(35): 21450-21458, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817427

RESUMEN

How overall principles of cell-type-specific gene regulation (the "logic") may change during ontogeny is largely unexplored. We compared transcriptomic, epigenomic, and three-dimensional (3D) genomic profiles in embryonic (EryP) and adult (EryD) erythroblasts. Despite reduced chromatin accessibility compared to EryP, distal chromatin of EryD is enriched in H3K27ac, Gata1, and Myb occupancy. EryP-/EryD-shared enhancers are highly correlated with red blood cell identity genes, whereas cell-type-specific regulation employs different cis elements in EryP and EryD cells. In contrast to EryP-specific genes, which exhibit promoter-centric regulation through Gata1, EryD-specific genes rely more on distal enhancers for regulation involving Myb-mediated enhancer activation. Gata1 HiChIP demonstrated an overall increased enhancer-promoter interactions at EryD-specific genes, whereas genome editing in selected loci confirmed distal enhancers are required for gene expression in EryD but not in EryP. Applying a metric for enhancer dependence of transcription, we observed a progressive reliance on cell-specific enhancers with increasing ontogenetic age among diverse tissues of mouse and human origin. Our findings highlight fundamental and conserved differences at distinct developmental stages, characterized by simpler promoter-centric regulation of cell-type-specific genes in embryonic cells and increased combinatorial enhancer-driven control in adult cells.


Asunto(s)
Factores de Edad , Factor de Transcripción GATA1/genética , Regulación del Desarrollo de la Expresión Génica/genética , Animales , Cromatina , Elementos de Facilitación Genéticos/genética , Eritroblastos , Eritropoyesis/fisiología , Femenino , Expresión Génica , Genómica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética
7.
Stem Cells ; 38(7): 818-821, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32159910

RESUMEN

The four transcription factors of the Yamanaka cocktail (Oct4, Sox2, Klf4, and Myc, termed OSKM) are famously capable of reprogramming somatic cells into induced pluripotent stem cells (iPSCs). In an article recently published in Nature Communications, Wang et al describe the unexpected discovery that short-term activation of OSKM expression in acute myeloid leukemia cells in vivo induces apoptosis while negligibly affecting normal hematopoietic stem and progenitor cells (Nat Commun 2019;10:5594). These findings have potential implications for novel anticancer strategies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Leucemia , Reprogramación Celular/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Leucemia/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/metabolismo
8.
Mol Cell ; 45(3): 330-43, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22325351

RESUMEN

Polycomb repressive complexes (PRCs) play key roles in developmental epigenetic regulation. Yet the mechanisms that target PRCs to specific loci in mammalian cells remain incompletely understood. In this study we show that Bmi1, a core component of Polycomb Repressive Complex 1 (PRC1), binds directly to the Runx1/CBFß transcription factor complex. Genome-wide studies in megakaryocytic cells demonstrate significant chromatin occupancy overlap between the PRC1 core component Ring1b and Runx1/CBFß and functional regulation of a considerable fraction of commonly bound genes. Bmi1/Ring1b and Runx1/CBFß deficiencies generate partial phenocopies of one another in vivo. We also show that Ring1b occupies key Runx1 binding sites in primary murine thymocytes and that this occurs via PRC2-independent mechanisms. Genetic depletion of Runx1 results in reduced Ring1b binding at these sites in vivo. These findings provide evidence for site-specific PRC1 chromatin recruitment by core binding transcription factors in mammalian cells.


Asunto(s)
Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Cromatografía de Afinidad , Análisis por Conglomerados , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/fisiología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/aislamiento & purificación , Linfocitos T/metabolismo , Timocitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
9.
Stem Cells ; 35(7): 1773-1785, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28436588

RESUMEN

ERG, an ETS family transcription factor frequently overexpressed in human leukemia, has been implicated as a key regulator of hematopoietic stem cells. However, how ERG controls normal hematopoiesis, particularly at the stem and progenitor cell level, and how it contributes to leukemogenesis remain incompletely understood. Using homologous recombination, we generated an Erg knockdown allele (Ergkd ) in which Erg expression can be conditionally restored by Cre recombinase. Ergkd/kd animals die at E10.5-E11.5 due to defects in endothelial and hematopoietic cells, but can be completely rescued by Tie2-Cre-mediated restoration of Erg in these cells. In Ergkd/+ mice, ∼40% reduction in Erg dosage perturbs both fetal liver and bone marrow hematopoiesis by reducing the numbers of Lin- Sca-1+ c-Kit+ (LSK) hematopoietic stem and progenitor cells (HSPCs) and megakaryocytic progenitors. By genetic mosaic analysis, we find that Erg-restored HSPCs outcompete Ergkd/+ HSPCs for contribution to adult hematopoiesis in vivo. This defect is in part due to increased apoptosis of HSPCs with reduced Erg dosage, a phenotype that becomes more drastic during 5-FU-induced stress hematopoiesis. Expression analysis reveals that reduced Erg expression leads to changes in expression of a subset of ERG target genes involved in regulating survival of HSPCs, including increased expression of a pro-apoptotic regulator Bcl2l11 (Bim) and reduced expression of Jun. Collectively, our data demonstrate that ERG controls survival of HSPCs, a property that may be used by leukemic cells. Stem Cells 2017;35:1773-1785.


Asunto(s)
Apoptosis/genética , Dosificación de Gen , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Oncogénicas/genética , Regulador Transcripcional ERG/genética , Animales , Antimetabolitos/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Femenino , Fluorouracilo/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Prueba de Complementación Genética , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Integrasas/genética , Integrasas/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Regulador Transcripcional ERG/deficiencia
10.
Proc Natl Acad Sci U S A ; 112(52): 15988-93, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26668377

RESUMEN

Changes of histone modification status at critical lineage-specifying gene loci in multipotent precursors can influence cell fate commitment. The contribution of these epigenetic mechanisms to natural killer (NK) cell lineage determination from common lymphoid precursors is not understood. Here we investigate the impact of histone methylation repressive marks (H3 Lys27 trimethylation; H3K27(me3)) on early NK cell differentiation. We demonstrate that selective loss of the histone-lysine N-methyltransferase Ezh2 (enhancer of zeste homolog 2) or inhibition of its enzymatic activity with small molecules unexpectedly increased generation of the IL-15 receptor (IL-15R) CD122(+) NK precursors and mature NK progeny from both mouse and human hematopoietic stem and progenitor cells. Mechanistic studies revealed that enhanced NK cell expansion and cytotoxicity against tumor cells were associated with up-regulation of CD122 and the C-type lectin receptor NKG2D. Moreover, NKG2D deficiency diminished the positive effects of Ezh2 inhibitors on NK cell commitment. Identification of the contribution of Ezh2 to NK lineage specification and function reveals an epigenetic-based mechanism that regulates NK cell development and provides insight into the clinical application of Ezh2 inhibitors in NK-based cancer immunotherapies.


Asunto(s)
Diferenciación Celular/genética , N-Metiltransferasa de Histona-Lisina/genética , Células Asesinas Naturales/metabolismo , Complejo Represivo Polycomb 2/genética , Animales , Línea Celular Tumoral , Linaje de la Célula/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Proteína Potenciadora del Homólogo Zeste 2 , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Immunoblotting , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células K562 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Mol Cell ; 36(4): 682-95, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19941827

RESUMEN

The transcription factor GATA-1 is required for terminal erythroid maturation and functions as an activator or repressor depending on gene context. Yet its in vivo site selectivity and ability to distinguish between activated versus repressed genes remain incompletely understood. In this study, we performed GATA-1 ChIP-seq in erythroid cells and compared it to GATA-1-induced gene expression changes. Bound and differentially expressed genes contain a greater number of GATA-binding motifs, a higher frequency of palindromic GATA sites, and closer occupancy to the transcriptional start site versus nondifferentially expressed genes. Moreover, we show that the transcription factor Zbtb7a occupies GATA-1-bound regions of some direct GATA-1 target genes, that the presence of SCL/TAL1 helps distinguish transcriptional activation versus repression, and that polycomb repressive complex 2 (PRC2) is involved in epigenetic silencing of a subset of GATA-1-repressed genes. These data provide insights into GATA-1-mediated gene regulation in vivo.


Asunto(s)
Cromatina/metabolismo , Factor de Transcripción GATA1/metabolismo , Genoma/genética , Proteínas Represoras/metabolismo , Activación Transcripcional/genética , Animales , Secuencia de Bases , Sitios de Unión , Biotina/metabolismo , Biotinilación , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Biología Computacional , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Silenciador del Gen , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Proteínas del Grupo Polycomb , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Secuencia de ADN , Estreptavidina/metabolismo
12.
J Neurosci ; 35(22): 8640-52, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041929

RESUMEN

Myelination of peripheral nerves by Schwann cells requires coordinate regulation of gene repression as well as gene activation. Several chromatin remodeling pathways critical for peripheral nerve myelination have been identified, but the functions of histone methylation in the peripheral nerve have not been elucidated. To determine the role of histone H3 Lys27 methylation, we have generated mice with a Schwann cell-specific knock-out of Eed, which is an essential subunit of the polycomb repressive complex 2 (PRC2) that catalyzes methylation of histone H3 Lys27. Analysis of this mutant revealed no significant effects on early postnatal development of myelin. However, its loss eventually causes progressive hypermyelination of small-diameter axons and apparent fragmentation of Remak bundles. These data identify the PRC2 complex as an epigenomic modulator of mature myelin thickness, which is associated with changes in Akt phosphorylation. Interestingly, we found that Eed inactivation causes derepression of several genes, e.g., Sonic hedgehog (Shh) and Insulin-like growth factor-binding protein 2 (Igfbp2), that become activated after nerve injury, but without activation of a primary regulator of the injury program, c-Jun. Analysis of the activated genes in cultured Schwann cells showed that Igfbp2 regulates Akt activation. Our results identify an epigenomic pathway required for establishing thickness of mature myelin and repressing genes that respond to nerve injury.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Vaina de Mielina/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Células de Schwann/metabolismo , Nervio Ciático/citología , Animales , Animales Recién Nacidos , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas Hedgehog/metabolismo , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Proteína P0 de la Mielina/genética , Proteína Oncogénica v-akt/metabolismo , Complejo Represivo Polycomb 2/genética , Ratas , Nervio Ciático/ultraestructura , Transducción de Señal/fisiología , Transfección
13.
Proc Natl Acad Sci U S A ; 109(13): 5028-33, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22396593

RESUMEN

A growing body of data suggests the importance of epigenetic mechanisms in cancer. Polycomb repressive complex 2 (PRC2) has been implicated in self-renewal and cancer progression, and its components are overexpressed in many cancers. However, its role in cancer development and progression remains unclear. We used conditional alleles for the PRC2 components enhancer of zeste 2 (Ezh2) and embryonic ectoderm development (Eed) to characterize the role of PRC2 function in leukemia development and progression. Compared with wild-type leukemia, Ezh2-null MLL-AF9-mediated acute myeloid leukemia (AML) failed to accelerate upon secondary transplantation. However, Ezh2-null leukemias maintained self-renewal up to the third round of transplantation, indicating that Ezh2 is not strictly required for MLL-AF9 AML, but plays a role in leukemia progression. Genome-wide analyses of PRC2-mediated trimethylation of histone 3 demonstrated locus-specific persistence of H3K27me3 despite inactivation of Ezh2, suggesting partial compensation by Ezh1. In contrast, inactivation of the essential PRC2 gene, Eed, led to complete ablation of PRC2 function, which was incompatible with leukemia growth. Gene expression array analyses indicated more profound gene expression changes in Eed-null compared with Ezh2-null leukemic cells, including down-regulation of Myc target genes and up-regulation of PRC2 targets. Manipulating PRC2 function may be of therapeutic benefit in AML.


Asunto(s)
Leucemia/patología , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Represoras/metabolismo , Animales , Proliferación Celular , Inmunoprecipitación de Cromatina , Citoprotección , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Silenciador del Gen , Genes Relacionados con las Neoplasias/genética , Sitios Genéticos/genética , Genoma/genética , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Leucemia/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Fenotipo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo
14.
Circ Res ; 110(3): 406-15, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22158708

RESUMEN

RATIONALE: Epigenetic marks are crucial for organogenesis, but their role in heart development is poorly understood. Polycomb repressive complex 2 (PRC2) trimethylates histone H3 at lysine 27, which establishes H3K27me3 repressive epigenetic marks that promote tissue-specific differentiation by silencing ectopic gene programs. OBJECTIVE: We studied the function of PRC2 in murine heart development using a tissue-restricted conditional inactivation strategy. METHODS AND RESULTS: Inactivation of the PRC2 subunit Ezh2 by Nkx2-5(Cre) (Ezh2(NK)) caused lethal congenital heart malformations, namely, compact myocardial hypoplasia, hypertrabeculation, and ventricular septal defect. Candidate and genome-wide RNA expression profiling and chromatin immunoprecipitation analyses of Ezh2(NK) heart identified genes directly repressed by EZH2. Among these were the potent cell cycle inhibitors Ink4a/b (inhibitors of cyclin-dependent kinase 4 A and B), the upregulation of which was associated with decreased cardiomyocyte proliferation in Ezh2(NK). EZH2-repressed genes were enriched for transcriptional regulators of noncardiomyocyte expression programs such as Pax6, Isl1, and Six1. EZH2 was also required for proper spatiotemporal regulation of cardiac gene expression, because Hcn4, Mlc2a, and Bmp10 were inappropriately upregulated in ventricular RNA. PRC2 was also required later in heart development, as indicated by cardiomyocyte-restricted TNT-Cre inactivation of the PRC2 subunit Eed. However, Ezh2 inactivation by TNT-Cre did not cause an overt phenotype, likely because of functional redundancy with Ezh1. Thus, early Ezh2 inactivation by Nk2-5(Cre) caused later disruption of cardiomyocyte gene expression and heart development. CONCLUSIONS: Our study reveals a previously undescribed role of EZH2 in regulating heart formation and shows that perturbation of the epigenetic landscape early in cardiogenesis has sustained disruptive effects at later developmental stages.


Asunto(s)
Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Corazón/embriología , Corazón/fisiología , Proteínas Represoras/fisiología , Animales , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Estudio de Asociación del Genoma Completo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Modelos Animales , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Proteínas Represoras/genética , Factores de Transcripción/fisiología
15.
Proc Natl Acad Sci U S A ; 105(16): 6057-62, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18424555

RESUMEN

Earlier work has shown that the transcription factor C/EBPalpha induced a transdifferentiation of committed lymphoid precursors into macrophages in a process requiring endogenous PU.1. Here we have examined the effects of PU.1 and C/EBPalpha on fibroblasts, a cell type distantly related to blood cells and akin to myoblasts, adipocytes, osteoblasts, and chondroblasts. The combination of the two factors, as well as PU.1 and C/EBPbeta, induced the up-regulation of macrophage/hematopoietic cell surface markers in a large proportion of NIH 3T3 cells. They also up-regulated these markers in mouse embryo- and adult skin-derived fibroblasts. Based on cell morphology, activation of macrophage-associated genes, and extinction of fibroblast-associated genes, cell lines containing an attenuated form of PU.1 and C/EBPalpha acquired a macrophage-like phenotype. The lines also display macrophage functions: They phagocytose small particles and bacteria, mount a partial inflammatory response, and exhibit strict CSF-1 dependence for growth. The myeloid conversion is primarily induced by PU.1, with C/EBPalpha acting as a modulator of macrophage-specific gene expression. Our data suggest that it might become possible to induce the transdifferentiation of skin-derived fibroblasts into cell types desirable for tissue regeneration.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Transdiferenciación Celular , Fibroblastos/citología , Macrófagos/citología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Transdiferenciación Celular/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Antígenos Comunes de Leucocito/análisis , Antígenos Comunes de Leucocito/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Antígeno de Macrófago-1/análisis , Antígeno de Macrófago-1/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células 3T3 NIH , Fagocitosis/genética , Proteínas Proto-Oncogénicas/genética , Retroviridae/genética , Transactivadores/genética , Transfección , Regulación hacia Arriba
17.
Sci Rep ; 8(1): 14335, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254371

RESUMEN

Thymic epithelial cells (TEC) are essential for thymocyte differentiation and repertoire selection. Despite their indispensable role in generating functional T cells, the molecular mechanisms that orchestrate TEC development from endodermal progenitors in the third pharyngeal pouch (3rd PP) are not fully understood. We recently reported that the T-box transcription factor TBX1 negatively regulates TEC development. Although initially expressed throughout the 3rd PP, Tbx1 becomes downregulated in thymus-fated progenitors and when ectopically expressed impairs TEC progenitor proliferation and differentiation. Here we show that ectopic Tbx1 expression in thymus fated endoderm increases expression of Polycomb repressive complex 2 (PRC2) target genes in TEC. PRC2 is an epigenetic modifier that represses gene expression by catalyzing trimethylation of lysine 27 on histone H3. The increased expression of PRC2 target genes suggests that ectopic Tbx1 interferes with PRC2 activity and implicates PRC2 as an important regulator of TEC development. To test this hypothesis, we used Foxn1Cre to delete Eed, a PRC2 component required for complex stability and function in thymus fated 3rd PP endoderm. Proliferation and differentiation of fetal and newborn TEC were disrupted in the conditional knockout (EedCKO) mutants leading to severely dysplastic adult thymi. Consistent with PRC2-mediated transcriptional silencing, the majority of differentially expressed genes (DEG) were upregulated in EedCKO TEC. Moreover, a high frequency of EedCKO DEG overlapped with DEG in TEC that ectopically expressed Tbx1. These findings demonstrate that PRC2 plays a critical role in TEC development and suggest that Tbx1 expression must be downregulated in thymus fated 3rd PP endoderm to ensure optimal PRC2 function.


Asunto(s)
Células Epiteliales/citología , Complejo Represivo Polycomb 2/metabolismo , Timo/citología , Animales , Diferenciación Celular , Linaje de la Célula , Eliminación de Gen , Regulación de la Expresión Génica , Histonas/metabolismo , Metilación , Ratones , Complejo Represivo Polycomb 2/deficiencia , Complejo Represivo Polycomb 2/genética , Proteínas de Dominio T Box/genética
18.
Cell Metab ; 27(6): 1294-1308.e7, 2018 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-29754954

RESUMEN

To date, it remains largely unclear to what extent chromatin machinery contributes to the susceptibility and progression of complex diseases. Here, we combine deep epigenome mapping with single-cell transcriptomics to mine for evidence of chromatin dysregulation in type 2 diabetes. We find two chromatin-state signatures that track ß cell dysfunction in mice and humans: ectopic activation of bivalent Polycomb-silenced domains and loss of expression at an epigenomically unique class of lineage-defining genes. ß cell-specific Polycomb (Eed/PRC2) loss of function in mice triggers diabetes-mimicking transcriptional signatures and highly penetrant, hyperglycemia-independent dedifferentiation, indicating that PRC2 dysregulation contributes to disease. The work provides novel resources for exploring ß cell transcriptional regulation and identifies PRC2 as necessary for long-term maintenance of ß cell identity. Importantly, the data suggest a two-hit (chromatin and hyperglycemia) model for loss of ß cell identity in diabetes.


Asunto(s)
Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Silenciador del Gen , Células Secretoras de Insulina/metabolismo , Complejo Represivo Polycomb 2/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Mapeo Cromosómico , Diabetes Mellitus Tipo 2/genética , Epigenómica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hiperglucemia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Complejo Represivo Polycomb 2/genética , Análisis de la Célula Individual
19.
Mol Biol Cell ; 14(5): 2005-15, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12802071

RESUMEN

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the retinoblastoma protein, thereby promoting cell-cycle progression. Cyclin D1 is overexpressed in hematopoetic and epithelial malignancies correlating with poor prognosis and metastasis in several cancer types. Because tumor-associated macrophages have been shown to enhance malignant progression and metastasis, and cyclin D1-deficient mice are resistant to oncogene-induced malignancies, we investigated the function of cyclin D1-/- bone marrow-derived macrophages. Cyclin D1 deficiency increased focal complex formation at the site of substratum contact, and enhanced macrophage adhesion, yielding a flattened, circular morphology with reduced membrane ruffles. Migration in response to wounding, cytokine-mediated chemotaxis, and transendothelial cell migration of cyclin D1-/- bone marrow-derived macrophages were all substantially reduced. Thus, apart from proliferative and possible motility defects in the tumor cells themselves, the reduced motility and invasiveness of cyclin D1-/- tumor-associated macrophages may contribute to the tumor resistance of these mice.


Asunto(s)
Movimiento Celular/fisiología , Ciclina D1/metabolismo , Macrófagos/metabolismo , Animales , Adhesión Celular/fisiología , Quimiotaxis/fisiología , Ciclina D1/deficiencia , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones
20.
Elife ; 62017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28394251

RESUMEN

In proliferating cells, where most Polycomb repressive complex 2 (PRC2) studies have been performed, gene repression is associated with PRC2 trimethylation of H3K27 (H3K27me3). However, it is uncertain whether PRC2 writing of H3K27me3 is mechanistically required for gene silencing. Here, we studied PRC2 function in postnatal mouse cardiomyocytes, where the paucity of cell division obviates bulk H3K27me3 rewriting after each cell cycle. EED (embryonic ectoderm development) inactivation in the postnatal heart (EedCKO) caused lethal dilated cardiomyopathy. Surprisingly, gene upregulation in EedCKO was not coupled with loss of H3K27me3. Rather, the activating histone mark H3K27ac increased. EED interacted with histone deacetylases (HDACs) and enhanced their catalytic activity. HDAC overexpression normalized EedCKO heart function and expression of derepressed genes. Our results uncovered a non-canonical, H3K27me3-independent EED repressive mechanism that is essential for normal heart function. Our results further illustrate that organ dysfunction due to epigenetic dysregulation can be corrected by epigenetic rewiring.


Asunto(s)
Represión Epigenética , Corazón/embriología , Histona Desacetilasas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Células Cultivadas , Metilación , Ratones , Ratones Noqueados , Miocitos Cardíacos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA