Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cell ; 184(25): 6081-6100.e26, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34861191

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable success in hematological malignancies but remains ineffective in solid tumors, due in part to CAR T cell exhaustion in the solid tumor microenvironment. To study dysfunction of mesothelin-redirected CAR T cells in pancreatic cancer, we establish a robust model of continuous antigen exposure that recapitulates hallmark features of T cell exhaustion and discover, both in vitro and in CAR T cell patients, that CAR dysregulation is associated with a CD8+ T-to-NK-like T cell transition. Furthermore, we identify a gene signature defining CAR and TCR dysregulation and transcription factors, including SOX4 and ID3 as key regulators of CAR T cell exhaustion. Our findings shed light on the plasticity of human CAR T cells and demonstrate that genetic downmodulation of ID3 and SOX4 expression can improve the efficacy of CAR T cell therapy in solid tumors by preventing or delaying CAR T cell dysfunction.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Pancreáticas/terapia , Receptores Quiméricos de Antígenos/inmunología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas Inhibidoras de la Diferenciación/inmunología , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Proteínas de Neoplasias/inmunología , Factores de Transcripción SOXC/inmunología
2.
Cell ; 168(1-2): 135-149.e22, 2017 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-28086087

RESUMEN

CBP/p300 are transcription co-activators whose binding is a signature of enhancers, cis-regulatory elements that control patterns of gene expression in multicellular organisms. Active enhancers produce bi-directional enhancer RNAs (eRNAs) and display CBP/p300-dependent histone acetylation. Here, we demonstrate that CBP binds directly to RNAs in vivo and in vitro. RNAs bound to CBP in vivo include a large number of eRNAs. Using steady-state histone acetyltransferase (HAT) assays, we show that an RNA binding region in the HAT domain of CBP-a regulatory motif unique to CBP/p300-allows RNA to stimulate CBP's HAT activity. At enhancers where CBP interacts with eRNAs, stimulation manifests in RNA-dependent changes in the histone acetylation mediated by CBP, such as H3K27ac, and by corresponding changes in gene expression. By interacting directly with CBP, eRNAs contribute to the unique chromatin structure at active enhancers, which, in turn, is required for regulation of target genes.


Asunto(s)
Histona Acetiltransferasas/metabolismo , ARN no Traducido/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Línea Celular , Elementos de Facilitación Genéticos , Fibroblastos/metabolismo , Histonas/metabolismo , Ratones
3.
Cell ; 151(5): 994-1004, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23159369

RESUMEN

The ectopic expression of transcription factors can reprogram cell fate, yet it is unknown how the initial binding of factors to the genome relates functionally to the binding seen in the minority of cells that become reprogrammed. We report a map of Oct4, Sox2, Klf4, and c-Myc (O, S, K, and M) on the human genome during the first 48 hr of reprogramming fibroblasts to pluripotency. Three striking aspects of the initial chromatin binding events include an unexpected role for c-Myc in facilitating OSK chromatin engagement, the primacy of O, S, and K as pioneer factors at enhancers of genes that promote reprogramming, and megabase-scale chromatin domains spanned by H3K9me3, including many genes required for pluripotency, that prevent initial OSKM binding and impede the efficiency of reprogramming. We find diverse aspects of initial factor binding that must be overcome in the minority of cells that become reprogrammed.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Genoma Humano , Células Madre Pluripotentes/metabolismo , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Fibroblastos/citología , Expresión Génica , Código de Histonas , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Modelos Moleculares , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción SOXB1/metabolismo
4.
Mol Cell ; 73(4): 684-698.e8, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30773298

RESUMEN

Accumulation of senescent cells during aging contributes to chronic inflammation and age-related diseases. While senescence is associated with profound alterations of the epigenome, a systematic view of epigenetic factors in regulating senescence is lacking. Here, we curated a library of short hairpin RNAs for targeted silencing of all known epigenetic proteins and performed a high-throughput screen to identify key candidates whose downregulation can delay replicative senescence of primary human cells. This screen identified multiple new players including the histone acetyltransferase p300 that was found to be a primary driver of the senescent phenotype. p300, but not the paralogous CBP, induces a dynamic hyper-acetylated chromatin state and promotes the formation of active enhancer elements in the non-coding genome, leading to a senescence-specific gene expression program. Our work illustrates a causal role of histone acetyltransferases and acetylation in senescence and suggests p300 as a potential therapeutic target for senescence and age-related diseases.


Asunto(s)
Proliferación Celular , Senescencia Celular , Ensamble y Desensamble de Cromatina , Cromatina/enzimología , Fibroblastos/enzimología , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Proliferación Celular/genética , Senescencia Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Represión Epigenética , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Histonas/genética , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Factores de Transcripción p300-CBP/genética
5.
Genes Dev ; 32(2): 181-193, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440247

RESUMEN

Epithelial tissues rely on a highly coordinated balance between self-renewal, proliferation, and differentiation, disruption of which may drive carcinogenesis. The epigenetic regulator KMT2D (MLL4) is one of the most frequently mutated genes in all cancers, particularly epithelial cancers, yet its normal function in these tissues is unknown. Here, we identify a novel role for KMT2D in coordinating this fine balance, as depletion of KMT2D from undifferentiated epidermal keratinocytes results in reduced proliferation, premature spurious activation of terminal differentiation genes, and disorganized epidermal stratification. Genome-wide, KMT2D interacts with p63 and is enriched at its target enhancers. Depletion of KMT2D results in a broad loss of enhancer histone modifications H3 Lys 4 (H3K4) monomethylation (H3K4me1) and H3K27 acetylation (H3K27ac) as well as reduced expression of p63 target genes, including key genes involved in epithelial development and adhesion. Together, these results reveal a critical role for KMT2D in the control of epithelial enhancers and p63 target gene expression, including the requirement of KMT2D for the maintenance of epithelial progenitor gene expression and the coordination of proper terminal differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos , Queratinocitos/metabolismo , Proteínas de Neoplasias/fisiología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Código de Histonas , Homeostasis , Humanos , Proteínas de Neoplasias/metabolismo
6.
Nature ; 571(7764): 211-218, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31207603

RESUMEN

Exhausted CD8+ T (Tex) cells in chronic infections and cancer have limited effector function, high co-expression of inhibitory receptors and extensive transcriptional changes compared with effector (Teff) or memory (Tmem) CD8+ T cells. Tex cells are important clinical targets of checkpoint blockade and other immunotherapies. Epigenetically, Tex cells are a distinct immune subset, with a unique chromatin landscape compared with Teff and Tmem cells. However, the mechanisms that govern the transcriptional and epigenetic development of Tex cells remain unknown. Here we identify the HMG-box transcription factor TOX as a central regulator of Tex cells in mice. TOX is largely dispensable for the formation of Teff and Tmem cells, but it is critical for exhaustion: in the absence of TOX, Tex cells do not form. TOX is induced by calcineurin and NFAT2, and operates in a feed-forward loop in which it becomes calcineurin-independent and sustained in Tex cells. Robust expression of TOX therefore results in commitment to Tex cells by translating persistent stimulation into a distinct Tex cell transcriptional and epigenetic developmental program.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Epistasis Genética , Proteínas de Homeodominio/metabolismo , Transcripción Genética , Animales , Calcineurina/metabolismo , Señalización del Calcio , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica/inmunología , Genotipo , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Escape del Tumor
7.
Mol Cell ; 68(6): 1023-1037.e15, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29272703

RESUMEN

Heterochromatin is integral to cell identity maintenance by impeding the activation of genes for alternate cell fates. Heterochromatic regions are associated with histone 3 lysine 9 trimethylation (H3K9me3) or H3K27me3, but these modifications are also found in euchromatic regions that permit transcription. We discovered that resistance to sonication is a reliable indicator of the heterochromatin state, and we developed a biophysical method (gradient-seq) to discriminate subtypes of H3K9me3 and H3K27me3 domains in sonication-resistant heterochromatin (srHC) versus euchromatin. These classifications are more accurate than the histone marks alone in predicting transcriptional silence and resistance of alternate fate genes to activation during direct cell conversion. Our proteomics of H3K9me3-marked srHC and functional screens revealed diverse proteins, including RBMX and RBMXL1, that impede gene induction during cellular reprogramming. Isolation of srHC with gradient-seq provides a genome-wide map of chromatin structure, elucidating subtypes of repressed domains that are uniquely predictive of diverse other chromatin properties.


Asunto(s)
Biomarcadores/análisis , Reprogramación Celular , Proteínas Cromosómicas no Histona/metabolismo , Genómica/métodos , Heterocromatina/genética , Heterocromatina/metabolismo , Proteómica/métodos , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Mapeo Cromosómico , Fibroblastos/citología , Fibroblastos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/genética , Histonas/metabolismo , Humanos
8.
Mol Cell ; 62(1): 79-91, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058788

RESUMEN

Nuclear DNA wraps around core histones to form nucleosomes, which restricts the binding of transcription factors to gene regulatory sequences. Pioneer transcription factors can bind DNA sites on nucleosomes and initiate gene regulatory events, often leading to the local opening of chromatin. However, the nucleosomal configuration of open chromatin and the basis for its regulation is unclear. We combined low and high levels of micrococcal nuclease (MNase) digestion along with core histone mapping to assess the nucleosomal configuration at enhancers and promoters in mouse liver. We find that MNase-accessible nucleosomes, bound by transcription factors, are retained more at liver-specific enhancers than at promoters and ubiquitous enhancers. The pioneer factor FoxA displaces linker histone H1, thereby keeping enhancer nucleosomes accessible in chromatin and allowing other liver-specific transcription factors to bind and stimulate transcription. Thus, nucleosomes are not exclusively repressive to gene regulation when they are retained with, and exposed by, pioneer factors.


Asunto(s)
Elementos de Facilitación Genéticos , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Nucleosomas/metabolismo , Animales , Histonas/metabolismo , Hígado/metabolismo , Ratones , Nucleosomas/genética , Especificidad de Órganos , Regiones Promotoras Genéticas , Transcripción Genética
9.
Genes Dev ; 30(3): 321-36, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26833731

RESUMEN

Oncogene-induced senescence (OIS) and therapy-induced senescence (TIS), while tumor-suppressive, also promote procarcinogenic effects by activating the DNA damage response (DDR), which in turn induces inflammation. This inflammatory response prominently includes an array of cytokines known as the senescence-associated secretory phenotype (SASP). Previous observations link the transcription-associated methyltransferase and oncoprotein MLL1 to the DDR, leading us to investigate the role of MLL1 in SASP expression. Our findings reveal direct MLL1 epigenetic control over proproliferative cell cycle genes: MLL1 inhibition represses expression of proproliferative cell cycle regulators required for DNA replication and DDR activation, thus disabling SASP expression. Strikingly, however, these effects of MLL1 inhibition on SASP gene expression do not impair OIS and, furthermore, abolish the ability of the SASP to enhance cancer cell proliferation. More broadly, MLL1 inhibition also reduces "SASP-like" inflammatory gene expression from cancer cells in vitro and in vivo independently of senescence. Taken together, these data demonstrate that MLL1 inhibition may be a powerful and effective strategy for inducing cancerous growth arrest through the direct epigenetic regulation of proliferation-promoting genes and the avoidance of deleterious OIS- or TIS-related tumor secretomes, which can promote both drug resistance and tumor progression.


Asunto(s)
Senescencia Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Transducción de Señal/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Daño del ADN , Técnicas de Silenciamiento del Gen , Células HEK293 , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Inflamación/genética , Células MCF-7 , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores , FN-kappa B/metabolismo , Neoplasias/fisiopatología , Fenotipo
10.
Nature ; 546(7658): 381-386, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28562591

RESUMEN

Metabolic production of acetyl coenzyme A (acetyl-CoA) is linked to histone acetylation and gene regulation, but the precise mechanisms of this process are largely unknown. Here we show that the metabolic enzyme acetyl-CoA synthetase 2 (ACSS2) directly regulates histone acetylation in neurons and spatial memory in mammals. In a neuronal cell culture model, ACSS2 increases in the nuclei of differentiating neurons and localizes to upregulated neuronal genes near sites of elevated histone acetylation. A decrease in ACSS2 lowers nuclear acetyl-CoA levels, histone acetylation, and responsive expression of the cohort of neuronal genes. In adult mice, attenuation of hippocampal ACSS2 expression impairs long-term spatial memory, a cognitive process that relies on histone acetylation. A decrease in ACSS2 in the hippocampus also leads to defective upregulation of memory-related neuronal genes that are pre-bound by ACSS2. These results reveal a connection between cellular metabolism, gene regulation, and neural plasticity and establish a link between acetyl-CoA generation 'on-site' at chromatin for histone acetylation and the transcription of key neuronal genes.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Hipocampo/enzimología , Hipocampo/fisiología , Histonas/metabolismo , Memoria/fisiología , Plasticidad Neuronal/genética , Activación Transcripcional , Acetato CoA Ligasa/deficiencia , Acetato CoA Ligasa/genética , Acetilcoenzima A/metabolismo , Acetilación , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/enzimología , Cromatina/genética , Cromatina/metabolismo , Regulación Enzimológica de la Expresión Génica , Hipocampo/metabolismo , Histonas/química , Consolidación de la Memoria/fisiología , Ratones , Plasticidad Neuronal/fisiología , Neuronas/citología , Neuronas/metabolismo , Regulación hacia Arriba
11.
Genes Dev ; 29(13): 1362-76, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26159996

RESUMEN

Epigenetic mechanisms, including histone post-translational modifications, control longevity in diverse organisms. Relatedly, loss of proper transcriptional regulation on a global scale is an emerging phenomenon of shortened life span, but the specific mechanisms linking these observations remain to be uncovered. Here, we describe a life span screen in Saccharomyces cerevisiae that is designed to identify amino acid residues of histones that regulate yeast replicative aging. Our results reveal that lack of sustained histone H3K36 methylation is commensurate with increased cryptic transcription in a subset of genes in old cells and with shorter life span. In contrast, deletion of the K36me2/3 demethylase Rph1 increases H3K36me3 within these genes, suppresses cryptic transcript initiation, and extends life span. We show that this aging phenomenon is conserved, as cryptic transcription also increases in old worms. We propose that epigenetic misregulation in aging cells leads to loss of transcriptional precision that is detrimental to life span, and, importantly, this acceleration in aging can be reversed by restoring transcriptional fidelity.


Asunto(s)
Epigénesis Genética/fisiología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Longevidad/genética , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Epigénesis Genética/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Metilación , Mutación , Procesamiento Proteico-Postraduccional/genética , Proteínas Represoras/genética , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
12.
Nature ; 527(7576): 105-9, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26524528

RESUMEN

Macroautophagy (hereafter referred to as autophagy) is a catabolic membrane trafficking process that degrades a variety of cellular constituents and is associated with human diseases. Although extensive studies have focused on autophagic turnover of cytoplasmic materials, little is known about the role of autophagy in degrading nuclear components. Here we report that the autophagy machinery mediates degradation of nuclear lamina components in mammals. The autophagy protein LC3/Atg8, which is involved in autophagy membrane trafficking and substrate delivery, is present in the nucleus and directly interacts with the nuclear lamina protein lamin B1, and binds to lamin-associated domains on chromatin. This LC3-lamin B1 interaction does not downregulate lamin B1 during starvation, but mediates its degradation upon oncogenic insults, such as by activated RAS. Lamin B1 degradation is achieved by nucleus-to-cytoplasm transport that delivers lamin B1 to the lysosome. Inhibiting autophagy or the LC3-lamin B1 interaction prevents activated RAS-induced lamin B1 loss and attenuates oncogene-induced senescence in primary human cells. Our study suggests that this new function of autophagy acts as a guarding mechanism protecting cells from tumorigenesis.


Asunto(s)
Autofagia , Lámina Nuclear/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Familia de las Proteínas 8 Relacionadas con la Autofagia , Transformación Celular Neoplásica , Células Cultivadas , Senescencia Celular , Cromatina/química , Cromatina/metabolismo , Citoplasma/metabolismo , Fibroblastos , Células HEK293 , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Lisosomas/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Proteolisis
13.
Nature ; 525(7568): 206-11, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26331536

RESUMEN

TP53 (which encodes p53 protein) is the most frequently mutated gene among all human cancers. Prevalent p53 missense mutations abrogate its tumour suppressive function and lead to a 'gain-of-function' (GOF) that promotes cancer. Here we show that p53 GOF mutants bind to and upregulate chromatin regulatory genes, including the methyltransferases MLL1 (also known as KMT2A), MLL2 (also known as KMT2D), and acetyltransferase MOZ (also known as KAT6A or MYST3), resulting in genome-wide increases of histone methylation and acetylation. Analysis of The Cancer Genome Atlas shows specific upregulation of MLL1, MLL2, and MOZ in p53 GOF patient-derived tumours, but not in wild-type p53 or p53 null tumours. Cancer cell proliferation is markedly lowered by genetic knockdown of MLL1 or by pharmacological inhibition of the MLL1 methyltransferase complex. Our study reveals a novel chromatin mechanism underlying the progression of tumours with GOF p53, and suggests new possibilities for designing combinatorial chromatin-based therapies for treating individual cancers driven by prevalent GOF p53 mutations.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Mutación/genética , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Cromatina/química , Femenino , Genes Supresores de Tumor , Genoma Humano/genética , Histona Acetiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Masculino , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias/metabolismo , Fenotipo , Unión Proteica , Procesamiento Proteico-Postraduccional
14.
Genes Dev ; 27(3): 251-60, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23355396

RESUMEN

While most transcription factors exit the chromatin during mitosis and the genome becomes silent, a subset of factors remains and "bookmarks" genes for rapid reactivation as cells progress through the cell cycle. However, it is unknown whether such bookmarking factors bind to chromatin similarly in mitosis and how different binding capacities among them relate to function. We compared a diverse set of transcription factors involved in liver differentiation and found markedly different extents of mitotic chromosome binding. Among them, the pioneer factor FoxA1 exhibits the greatest extent of mitotic chromosome binding. Genomically, ~15% of the FoxA1 interphase target sites are bound in mitosis, including at genes that are important for liver differentiation. Biophysical, genome mapping, and mutagenesis studies of FoxA1 reveals two different modes of binding to mitotic chromatin. Specific binding in mitosis occurs at sites that continue to be bound from interphase. Nonspecific binding in mitosis occurs across the chromosome due to the intrinsic chromatin affinity of FoxA1. Both specific and nonspecific binding contribute to timely reactivation of target genes post-mitosis. These studies reveal an unexpected diversity in the mechanisms by which transcription factors help retain cell identity during mitosis.


Asunto(s)
Cromatina/metabolismo , Cromosomas/metabolismo , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Mitosis , Línea Celular Tumoral , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Modelos Moleculares , Nucleosomas , Unión Proteica
15.
Genes Dev ; 27(16): 1787-99, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23934658

RESUMEN

Senescence is a stable proliferation arrest, associated with an altered secretory pathway, thought to promote tumor suppression and tissue aging. While chromatin regulation and lamin B1 down-regulation have been implicated as senescence effectors, functional interactions between them are poorly understood. We compared genome-wide Lys4 trimethylation on histone H3 (H3K4me3) and H3K27me3 distributions between proliferating and senescent human cells and found dramatic differences in senescence, including large-scale domains of H3K4me3- and H3K27me3-enriched "mesas" and H3K27me3-depleted "canyons." Mesas form at lamin B1-associated domains (LADs) in replicative senescence and oncogene-induced senescence and overlap DNA hypomethylation regions in cancer, suggesting that pre-malignant senescent chromatin changes foreshadow epigenetic cancer changes. Hutchinson-Gilford progeria syndrome fibroblasts (mutant lamin A) also show evidence of H3K4me3 mesas, suggesting a link between premature chromatin changes and accelerated cell senescence. Canyons mostly form between LADs and are enriched in genes and enhancers. H3K27me3 loss is correlated with up-regulation of key senescence genes, indicating a link between global chromatin changes and local gene expression regulation. Lamin B1 reduction in proliferating cells triggers senescence and formation of mesas and canyons. Our data illustrate profound chromatin reorganization during senescence and suggest that lamin B1 down-regulation in senescence is a key trigger of global and local chromatin changes that impact gene expression, aging, and cancer.


Asunto(s)
Envejecimiento/genética , Envejecimiento/patología , Cromatina/genética , Cromatina/metabolismo , Regulación de la Expresión Génica , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Línea Celular , Proliferación Celular , Células Cultivadas , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/patología , Histonas/metabolismo , Humanos , Metilación , Progeria/patología , Estructura Terciaria de Proteína
16.
Genes Dev ; 27(12): 1406-20, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23756653

RESUMEN

Cellular senescence is accompanied by dramatic changes in chromatin structure and gene expression. Using Saccharomyces cerevisiae mutants lacking telomerase (tlc1Δ) to model senescence, we found that with critical telomere shortening, the telomere-binding protein Rap1 (repressor activator protein 1) relocalizes to the upstream promoter regions of hundreds of new target genes. The set of new Rap1 targets at senescence (NRTS) is preferentially activated at senescence, and experimental manipulations of Rap1 levels indicate that it contributes directly to NRTS activation. A notable subset of NRTS includes the core histone-encoding genes; we found that Rap1 contributes to their repression and that histone protein levels decline at senescence. Rap1 and histones also display a target site-specific antagonism that leads to diminished nucleosome occupancy at the promoters of up-regulated NRTS. This antagonism apparently impacts the rate of senescence because underexpression of Rap1 or overexpression of the core histones delays senescence. Rap1 relocalization is not a simple consequence of lost telomere-binding sites, but rather depends on the Mec1 checkpoint kinase. Rap1 relocalization is thus a novel mechanism connecting DNA damage responses (DDRs) at telomeres to global changes in chromatin and gene expression while driving the pace of senescence.


Asunto(s)
Cromatina/metabolismo , Regulación Fúngica de la Expresión Génica , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Factores de Transcripción/metabolismo , Histonas/genética , Viabilidad Microbiana , Transporte de Proteínas , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/genética , Complejo Shelterina , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Factores de Transcripción/genética , Transcriptoma
17.
Gastroenterology ; 156(6): 1834-1848, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30689973

RESUMEN

BACKGROUND & AIMS: Little is known about mechanisms that underlie postnatal hepatocyte maturation and fibrosis at the chromatin level. We investigated the transcription of genes involved in maturation and fibrosis in postnatal hepatocytes of mice, focusing on the chromatin compaction the roles of the Polycomb repressive complex 2 histone methyltransferases EZH1 and EZH2. METHODS: Hepatocytes were isolated from mixed background C57BL/6J-C3H mice, as well as mice with liver-specific disruption of Ezh1 and/or Ezh2, at postnatal day 14 and 2 months after birth. Liver tissues were collected and analyzed by RNA sequencing, H3K27me3 chromatin immunoprecipitation sequencing, and sonication-resistant heterochromatin sequencing (a method to map heterochromatin and euchromatin). Liver damage was characterized by histologic analysis. RESULTS: We found more than 3000 genes differentially expressed in hepatocytes during liver maturation from postnatal day 14 to month 2 after birth. Disruption of Ezh1 and Ezh2 in livers caused perinatal hepatocytes to differentiate prematurely and to express genes at postnatal day 14 that would normally be induced by month 2 and differentiate prematurely. Loss of Ezh1 and Ezh2 also resulted in liver fibrosis. Genes with H3K27me3-postive and H3K4me3-positive euchromatic promoters were prematurely induced in hepatocytes with loss of Ezh1 and Ezh2-these genes included those that regulate hepatocyte maturation, fibrosis, and genes not specifically associated with the liver lineage. CONCLUSIONS: The Polycomb repressive complex 2 proteins EZH1 and EZH2 regulate genes that control hepatocyte maturation and fibrogenesis and genes not specifically associated with the liver lineage by acting at euchromatic promoter regions. EZH1 and EZH2 thereby promote liver homeostasis and prevent liver damage. Strategies to manipulate Polycomb proteins might be used to improve hepatocyte derivation protocols or developed for treatment of patients with liver fibrosis.


Asunto(s)
Diferenciación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Represión Epigenética , Regulación de la Expresión Génica/genética , Cirrosis Hepática/genética , Complejo Represivo Polycomb 2/genética , Animales , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Eucromatina , Femenino , Expresión Génica , Ontología de Genes , Hepatocitos , Histonas/metabolismo , Cirrosis Hepática/patología , Masculino , Metilación , Ratones , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Factores de Tiempo
18.
EMBO J ; 33(19): 2157-70, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25107471

RESUMEN

Endoderm cells undergo sequential fate choices to generate insulin-secreting beta cells. Ezh2 of the PRC2 complex, which generates H3K27me3, modulates the transition from endoderm to pancreas progenitors, but the role of Ezh2 and H3K27me3 in the next transition to endocrine progenitors is unknown. We isolated endoderm cells, pancreas progenitors, and endocrine progenitors from different staged mouse embryos and analyzed H3K27me3 genome-wide. Unlike the decline in H3K27me3 domains reported during embryonic stem cell differentiation in vitro, we find that H3K27me3 domains increase in number during endocrine progenitor development in vivo. Genes that lose the H3K27me3 mark typically encode transcriptional regulators, including those for pro-endocrine fates, whereas genes that acquire the mark typically are involved in cell biology and morphogenesis. Deletion of Ezh2 at the pancreas progenitor stage enhanced the production of endocrine progenitors and beta cells. Inhibition of EZH2 in embryonic pancreas explants and in human embryonic stem cell cultures increased endocrine progenitors in vitro. Our studies reveal distinct dynamics in H3K27me3 targets in vivo and a means to modulate beta cell development from stem cells.


Asunto(s)
Células Endocrinas/citología , Histonas/metabolismo , Islotes Pancreáticos/citología , Histona Demetilasas con Dominio de Jumonji/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Western Blotting , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Endocrinas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Organogénesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/metabolismo
19.
Genome Res ; 23(8): 1235-47, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23636946

RESUMEN

Genomes of eusocial insects code for dramatic examples of phenotypic plasticity and social organization. We compared the genomes of seven ants, the honeybee, and various solitary insects to examine whether eusocial lineages share distinct features of genomic organization. Each ant lineage contains ∼4000 novel genes, but only 64 of these genes are conserved among all seven ants. Many gene families have been expanded in ants, notably those involved in chemical communication (e.g., desaturases and odorant receptors). Alignment of the ant genomes revealed reduced purifying selection compared with Drosophila without significantly reduced synteny. Correspondingly, ant genomes exhibit dramatic divergence of noncoding regulatory elements; however, extant conserved regions are enriched for novel noncoding RNAs and transcription factor-binding sites. Comparison of orthologous gene promoters between eusocial and solitary species revealed significant regulatory evolution in both cis (e.g., Creb) and trans (e.g., fork head) for nearly 2000 genes, many of which exhibit phenotypic plasticity. Our results emphasize that genomic changes can occur remarkably fast in ants, because two recently diverged leaf-cutter ant species exhibit faster accumulation of species-specific genes and greater divergence in regulatory elements compared with other ants or Drosophila. Thus, while the "socio-genomes" of ants and the honeybee are broadly characterized by a pervasive pattern of divergence in gene composition and regulation, they preserve lineage-specific regulatory features linked to eusociality. We propose that changes in gene regulation played a key role in the origins of insect eusociality, whereas changes in gene composition were more relevant for lineage-specific eusocial adaptations.


Asunto(s)
Hormigas/genética , Genoma de los Insectos , Animales , Conducta Animal , Sitios de Unión , Secuencia Conservada , Metilación de ADN , Evolución Molecular , Regulación de la Expresión Génica , Himenópteros/genética , Proteínas de Insectos/genética , MicroARNs/genética , Modelos Genéticos , Filogenia , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ADN , Conducta Social , Especificidad de la Especie , Sintenía , Factores de Transcripción/genética
20.
Acta Neuropathol Commun ; 12(1): 3, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167174

RESUMEN

Alzheimer's Disease (AD) is a disorder characterized by cognitive decline, neurodegeneration, and accumulation of amyloid plaques and tau neurofibrillary tangles in the brain. Dysregulation of epigenetic histone modifications may lead to expression of transcriptional programs that play a role either in protecting against disease genesis or in worsening of disease pathology. One such histone modification, acetylation of histone H3 lysine residue 27 (H3K27ac), is primarily localized to genomic enhancer regions and promotes active gene transcription. We previously discovered H3K27ac to be more abundant in AD patient brain tissue compared to the brains of age-matched non-demented controls. In this study, we use iPSC-neurons derived from familial AD patients with an amyloid precursor protein (APP) duplication (APPDup neurons) as a model to study the functional effect of lowering CBP/P300 enzymes that catalyze H3K27ac. We found that homeostatic amyloid-reducing genes were upregulated in the APPDup neurons compared to non-demented controls. We lowered CBP/P300 to reduce H3K27ac, which led to decreased expression of numerous of these homeostatic amyloid-reducing genes, along with increased extracellular secretion of a toxic amyloid-ß species, Aß(1-42). Our findings suggest that epigenomic histone acetylation, including H3K27ac, drives expression of compensatory genetic programs in response to AD-associated insults, specifically those resulting from APP duplication, and thus may play a role in mitigating AD pathology in neurons.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/patología , Histonas/genética , Acetilación , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA