Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Rev Mol Cell Biol ; 19(4): 245-261, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29184195

RESUMEN

Multiple cell-signalling pathways converge on chromatin to induce gene expression programmes. The inducible transcriptional programmes that are established as a result of inflammatory or oncogenic signals are controlled by shared chromatin regulators. Therapeutic targeting of such chromatin dependencies has proved effective for controlling tumorigenesis and for preventing immunopathologies that are driven by overt inflammation. In this Review, we discuss how chromatin dependencies are established to regulate the expression of key oncogenes and inflammation-promoting genes and how a better mechanistic understanding of such chromatin dependencies can be leveraged to improve the magnitude, timing, duration and selectivity of cell responses with the aim of minimizing unwanted cellular and systemic effects. Recently, exciting progress has been made in cancer immunotherapy and in the development of drugs that target chromatin regulators. We discuss recent advances in clinical trials and the challenge of combining immune-cell-based therapies and epigenetic therapies to improve human health.


Asunto(s)
Cromatina/genética , Inflamación/genética , Neoplasias/genética , Animales , Carcinogénesis/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Inflamación/metabolismo , Modelos Genéticos , Neoplasias/metabolismo , Neoplasias/terapia , Transducción de Señal/genética , Factores de Transcripción/metabolismo
2.
Genes Dev ; 31(1): 12-17, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115466

RESUMEN

Global DNA demethylation is a hallmark of embryonic epigenetic reprogramming. However, embryos engage noncanonical DNA methylation maintenance mechanisms to ensure inheritance of exceptional epigenetic germline features to the soma. Besides the paradigmatic genomic imprints, these exceptions remain ill-defined, and the mechanisms ensuring demethylation resistance in the light of global reprogramming remain poorly understood. Here we show that the Y-linked gene Rbmy1a1 is highly methylated in mature sperm and resists DNA demethylation post-fertilization. Aberrant hypomethylation of the Rbmy1a1 promoter results in its ectopic activation, causing male-specific peri-implantation lethality. Rbmy1a1 is a novel target of the TRIM28 complex, which is required to protect its repressive epigenetic state during embryonic epigenetic reprogramming.


Asunto(s)
Metilación de ADN/genética , Desarrollo Embrionario/genética , Epigénesis Genética/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Animales , Células Cultivadas , Reprogramación Celular/genética , Implantación del Embrión/genética , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica/genética , Masculino , Mutación , Regiones Promotoras Genéticas/genética , Proteínas de Unión al ARN/genética , Espermatozoides/metabolismo , Proteína 28 que Contiene Motivos Tripartito
3.
Genes Dev ; 29(21): 2312-24, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26545815

RESUMEN

Postnatal spermatogonial stem cells (SSCs) progress through proliferative and developmental stages to populate the testicular niche prior to productive spermatogenesis. To better understand, we conducted extensive genomic profiling at multiple postnatal stages on subpopulations enriched for particular markers (THY1, KIT, OCT4, ID4, or GFRa1). Overall, our profiles suggest three broad populations of spermatogonia in juveniles: (1) epithelial-like spermatogonia (THY1(+); high OCT4, ID4, and GFRa1), (2) more abundant mesenchymal-like spermatogonia (THY1(+); moderate OCT4 and ID4; high mesenchymal markers), and (3) (in older juveniles) abundant spermatogonia committing to gametogenesis (high KIT(+)). Epithelial-like spermatogonia displayed the expected imprinting patterns, but, surprisingly, mesenchymal-like spermatogonia lacked imprinting specifically at paternally imprinted loci but fully restored imprinting prior to puberty. Furthermore, mesenchymal-like spermatogonia also displayed developmentally linked DNA demethylation at meiotic genes and also at certain monoallelic neural genes (e.g., protocadherins and olfactory receptors). We also reveal novel candidate receptor-ligand networks involving SSCs and the developing niche. Taken together, neonates/juveniles contain heterogeneous epithelial-like or mesenchymal-like spermatogonial populations, with the latter displaying extensive DNA methylation/chromatin dynamics. We speculate that this plasticity helps SSCs proliferate and migrate within the developing seminiferous tubule, with proper niche interaction and membrane attachment reverting mesenchymal-like spermatogonial subtype cells back to an epithelial-like state with normal imprinting profiles.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica/genética , Factores de Transcripción/genética , Animales , Cadherinas/genética , Células Cultivadas , Metilación de ADN , Epigenómica , Gametogénesis/genética , Perfilación de la Expresión Génica , Masculino , Ratones , Receptores Odorantes/genética , Transducción de Señal/genética , Antígenos Thy-1/metabolismo
4.
Development ; 146(19)2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30846446

RESUMEN

Global epigenetic reprogramming is vital to purge germ cell-specific epigenetic features to establish the totipotent state of the embryo. This process transpires to be carefully regulated and is not an undirected, radical erasure of parental epigenomes. The TRIM28 complex has been shown to be crucial in embryonic epigenetic reprogramming by regionally opposing DNA demethylation to preserve vital parental information to be inherited from germline to soma. Yet the DNA-binding factors guiding this complex to specific targets are largely unknown. Here, we uncover and characterize a novel, maternally expressed, TRIM28-interacting KRAB zinc-finger protein: ZFP708. It recruits the repressive TRIM28 complex to RMER19B retrotransposons to evoke regional heterochromatin formation. ZFP708 binding to these hitherto unknown TRIM28 targets is DNA methylation and H3K9me3 independent. ZFP708 mutant mice are viable and fertile, yet embryos fail to inherit and maintain DNA methylation at ZFP708 target sites. This can result in activation of RMER19B-adjacent genes, while ectopic expression of ZFP708 results in transcriptional repression. Finally, we describe the evolutionary conservation of ZFP708 in mice and rats, which is linked to the conserved presence of the targeted RMER19B retrotransposons in these species.


Asunto(s)
Represión Epigenética , Proteínas Represoras/metabolismo , Retroelementos/genética , Dedos de Zinc , Animales , Secuencia de Bases , Sitios de Unión/genética , Blastocisto/metabolismo , Metilación de ADN/genética , Embrión de Mamíferos/metabolismo , Evolución Molecular , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/metabolismo , Unión Proteica/genética , Ratas , Transcripción Genética , Proteína 28 que Contiene Motivos Tripartito/metabolismo
5.
Nature ; 523(7558): 96-100, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25970242

RESUMEN

Deregulated expression of the MYC transcription factor occurs in most human cancers and correlates with high proliferation, reprogrammed cellular metabolism and poor prognosis. Overexpressed MYC binds to virtually all active promoters within a cell, although with different binding affinities, and modulates the expression of distinct subsets of genes. However, the critical effectors of MYC in tumorigenesis remain largely unknown. Here we show that during lymphomagenesis in Eµ-myc transgenic mice, MYC directly upregulates the transcription of the core small nuclear ribonucleoprotein particle assembly genes, including Prmt5, an arginine methyltransferase that methylates Sm proteins. This coordinated regulatory effect is critical for the core biogenesis of small nuclear ribonucleoprotein particles, effective pre-messenger-RNA splicing, cell survival and proliferation. Our results demonstrate that MYC maintains the splicing fidelity of exons with a weak 5' donor site. Additionally, we identify pre-messenger-RNAs that are particularly sensitive to the perturbation of the MYC-PRMT5 axis, resulting in either intron retention (for example, Dvl1) or exon skipping (for example, Atr, Ep400). Using antisense oligonucleotides, we demonstrate the contribution of these splicing defects to the anti-proliferative/apoptotic phenotype observed in PRMT5-depleted Eµ-myc B cells. We conclude that, in addition to its well-documented oncogenic functions in transcription and translation, MYC also safeguards proper pre-messenger-RNA splicing as an essential step in lymphomagenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Linfoma/fisiopatología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN/fisiología , Animales , Exones/genética , Células HEK293 , Humanos , Intrones/genética , Ratones , Oligonucleótidos Antisentido/metabolismo , Proteína Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas , Proteínas Proto-Oncogénicas c-myc/genética
6.
Bioinformatics ; 29(19): 2501-2, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23864731

RESUMEN

SUMMARY: Binding free energy calculations obtained through molecular dynamics simulations reflect intermolecular interaction states through a series of independent snapshots. Typically, the free energies of multiple simulated series (each with slightly different starting conditions) need to be estimated. Previous approaches carry out this task by moving averages at certain decorrelation times, assuming that the system comes from a single conformation description of binding events. Here, we discuss a more general approach that uses statistical modeling, wavelets denoising and hierarchical clustering to estimate the significance of multiple statistically distinct subpopulations, reflecting potential macrostates of the system. We present the deltaGseg R package that performs macrostate estimation from multiple replicated series and allows molecular biologists/chemists to gain physical insight into the molecular details that are not easily accessible by experimental techniques. AVAILABILITY: deltaGseg is a Bioconductor R package available at http://bioconductor.org/packages/release/bioc/html/deltaGseg.html.


Asunto(s)
Simulación de Dinámica Molecular , Programas Informáticos , Análisis por Conglomerados , Humanos
7.
Nucleic Acids Res ; 40(19): 9534-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22987071

RESUMEN

p21 is a potent cyclin-dependent kinase inhibitor that plays a role in promoting G1 cell cycle arrest and cellular senescence. Consistent with this role, p21 is a downstream target of several tumour suppressors and oncogenes, and it is downregulated in the majority of tumours, including breast cancer. Here, we report that protein arginine methyltransferase 6 (PRMT6), a type I PRMT known to act as a transcriptional cofactor, directly represses the p21 promoter. PRMT6 knock-down (KD) results in a p21 derepression in breast cancer cells, which is p53-independent, and leads to cell cycle arrest, cellular senescence and reduced growth in soft agar assays and in severe combined immunodeficiency (SCID) mice for all the cancer lines examined. We finally show that bypassing the p21-mediated arrest rescues PRMT6 KD cells from senescence, and it restores their ability to grow on soft agar. We conclude that PRMT6 acts as an oncogene in breast cancer cells, promoting growth and preventing senescence, making it an attractive target for cancer therapy.


Asunto(s)
Neoplasias de la Mama/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Senescencia Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones SCID , Proteínas Nucleares/genética , Fenotipo , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Sci Adv ; 6(2): eaax9852, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31950080

RESUMEN

Holoprosencephaly (HPE) is a congenital forebrain defect often associated with embryonic lethality and lifelong disabilities. Currently, therapeutic and diagnostic options are limited by lack of knowledge of potential disease-causing mutations. We have identified a new mutation in the PRDM15 gene (C844Y) associated with a syndromic form of HPE in multiple families. We demonstrate that C844Y is a loss-of-function mutation impairing PRDM15 transcriptional activity. Genetic deletion of murine Prdm15 causes anterior/posterior (A/P) patterning defects and recapitulates the brain malformations observed in patients. Mechanistically, PRDM15 regulates the transcription of key effectors of the NOTCH and WNT/PCP pathways to preserve early midline structures in the developing embryo. Analysis of a large cohort of patients with HPE revealed potentially damaging mutations in several regulators of both pathways. Our findings uncover an unexpected link between NOTCH and WNT/PCP signaling and A/P patterning and set the stage for the identification of new HPE candidate genes.


Asunto(s)
Polaridad Celular , Proteínas de Unión al ADN/genética , Holoprosencefalia/genética , Mutación con Pérdida de Función/genética , Receptores Notch/metabolismo , Factores de Transcripción/genética , Vía de Señalización Wnt , Animales , Tipificación del Cuerpo/genética , Encéfalo/anomalías , Encéfalo/embriología , Polaridad Celular/genética , Estudios de Cohortes , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Placa Neural/metabolismo , Embarazo , Transcripción Genética , Dedos de Zinc
9.
J Clin Invest ; 126(1): 68-84, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595814

RESUMEN

MDM4 is a promising target for cancer therapy, as it is undetectable in most normal adult tissues but often upregulated in cancer cells to dampen p53 tumor-suppressor function. The mechanisms that underlie MDM4 upregulation in cancer cells are largely unknown. Here, we have shown that this key oncogenic event mainly depends on a specific alternative splicing switch. We determined that while a nonsense-mediated, decay-targeted isoform of MDM4 (MDM4-S) is produced in normal adult tissues as a result of exon 6 skipping, enhanced exon 6 inclusion leads to expression of full-length MDM4 in a large number of human cancers. Although this alternative splicing event is likely regulated by multiple splicing factors, we identified the SRSF3 oncoprotein as a key enhancer of exon 6 inclusion. In multiple human melanoma cell lines and in melanoma patient-derived xenograft (PDX) mouse models, antisense oligonucleotide-mediated (ASO-mediated) skipping of exon 6 decreased MDM4 abundance, inhibited melanoma growth, and enhanced sensitivity to MAPK-targeting therapeutics. Additionally, ASO-based MDM4 targeting reduced diffuse large B cell lymphoma PDX growth. As full-length MDM4 is enhanced in multiple human tumors, our data indicate that this strategy is applicable to a wide range of tumor types. We conclude that enhanced MDM4 exon 6 inclusion is a common oncogenic event and has potential as a clinically compatible therapeutic target.


Asunto(s)
Exones , Melanoma/terapia , Proteínas Nucleares/genética , Oligonucleótidos Antisentido/farmacología , Proteínas Proto-Oncogénicas/genética , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Melanoma/patología , Ratones , Proteínas de Unión al ARN/fisiología , Factores de Empalme Serina-Arginina , Proteína p53 Supresora de Tumor/fisiología
10.
Cell Stem Cell ; 15(2): 239-53, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-24835570

RESUMEN

Adult germline stem cells (AGSCs) self-renew (Thy1(+) enriched) or commit to gametogenesis (Kit(+) enriched). To better understand how chromatin regulates AGSC biology and gametogenesis, we derived stage-specific high-resolution profiles of DNA methylation, 5hmC, histone modifications/variants, and RNA-seq in AGSCs and during spermatogenesis. First, we define striking signaling and transcriptional differences between AGSC types, involving key self-renewal and proliferation pathways. Second, key pluripotency factors (e.g., Nanog) are silent in AGSCs and bear particular chromatin/DNAme attributes that may "poise" them for reactivation after fertilization. Third, AGSCs display chromatin "poising/bivalency" of enhancers and promoters for embryonic transcription factors. Remarkably, gametogenesis occurs without significant changes in DNAme and instead involves transcription of DNA-methylated promoters bearing high RNAPol2, H3K9ac, H3K4me3, low CG content, and (often) 5hmC. Furthermore, key findings were confirmed in human sperm. Here, we reveal AGSC signaling asymmetries and chromatin/DNAme strategies in AGSCs to poise key transcription factors and to activate DNA-methylated promoters during gametogenesis.


Asunto(s)
Células Madre Adultas/citología , Cromatina/fisiología , Espermatogénesis/fisiología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Separación Celular , Cromatina/metabolismo , Metilación de ADN , Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos , Citometría de Flujo , Perfilación de la Expresión Génica , Genómica , Histonas/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Transducción de Señal , Espermatozoides/metabolismo , Factores de Tiempo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA