Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 35(17-18): 1209-1228, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34413137

RESUMEN

The generation of myotubes from fibroblasts upon forced MyoD expression is a classic example of transcription factor-induced reprogramming. We recently discovered that additional modulation of signaling pathways with small molecules facilitates reprogramming to more primitive induced myogenic progenitor cells (iMPCs). Here, we dissected the transcriptional and epigenetic dynamics of mouse fibroblasts undergoing reprogramming to either myotubes or iMPCs using a MyoD-inducible transgenic model. Induction of MyoD in fibroblasts combined with small molecules generated Pax7+ iMPCs with high similarity to primary muscle stem cells. Analysis of intermediate stages of iMPC induction revealed that extinction of the fibroblast program preceded induction of the stem cell program. Moreover, key stem cell genes gained chromatin accessibility prior to their transcriptional activation, and these regions exhibited a marked loss of DNA methylation dependent on the Tet enzymes. In contrast, myotube generation was associated with few methylation changes, incomplete and unstable reprogramming, and an insensitivity to Tet depletion. Finally, we showed that MyoD's ability to bind to unique bHLH targets was crucial for generating iMPCs but dispensable for generating myotubes. Collectively, our analyses elucidate the role of MyoD in myogenic reprogramming and derive general principles by which transcription factors and signaling pathways cooperate to rewire cell identity.


Asunto(s)
Desarrollo de Músculos , Proteína MioD , Animales , Diferenciación Celular/genética , Ratones , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas , Músculo Esquelético , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/metabolismo , Células Madre/metabolismo
2.
Genes Dev ; 29(5): 467-82, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25737276

RESUMEN

Wilms' tumor, or nephroblastoma, is the most common pediatric renal cancer. The tumors morphologically resemble embryonic kidneys with a disrupted architecture and are associated with undifferentiated metanephric precursors. Here, we discuss genetic and epigenetic findings in Wilms' tumor in the context of renal development. Many of the genes implicated in Wilms' tumorigenesis are involved in the control of nephron progenitors or the microRNA (miRNA) processing pathway. Whereas the first group of genes has been extensively studied in normal development, the second finding suggests important roles for miRNAs in general-and specific miRNAs in particular-in normal kidney development that still await further analysis. The recent identification of Wilms' tumor cancer stem cells could provide a framework to integrate these pathways and translate them into new or improved therapeutic interventions.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Neoplasias Renales/genética , Riñón/embriología , Organogénesis/genética , Tumor de Wilms/genética , Animales , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Riñón/patología , MicroARNs/genética
3.
Development ; 146(19)2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515224

RESUMEN

Maintenance of pluripotency and specification towards a new cell fate are both dependent on precise interactions between extrinsic signals and transcriptional and epigenetic regulators. Directed methylation of cytosines by the de novo methyltransferases DNMT3A and DNMT3B plays an important role in facilitating proper differentiation, whereas DNMT1 is essential for maintaining global methylation levels in all cell types. Here, we generated single-cell mRNA expression data from wild-type, DNMT3A, DNMT3A/3B and DNMT1 knockout human embryonic stem cells and observed a widespread increase in cellular and transcriptional variability, even with limited changes in global methylation levels in the de novo knockouts. Furthermore, we found unexpected transcriptional repression upon either loss of the de novo methyltransferase DNMT3A or the double knockout of DNMT3A/3B that is further propagated upon differentiation to mesoderm and ectoderm. Taken together, our single-cell RNA-sequencing data provide a high-resolution view into the consequences of depleting the three catalytically active DNMTs in human pluripotent stem cells.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Ciclo Celular/genética , Diferenciación Celular/genética , Metilación de ADN/genética , ADN Metiltransferasa 3A , Elementos de Facilitación Genéticos/genética , Entropía , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , ADN Metiltransferasa 3B
4.
Genes Dev ; 28(9): 971-82, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24732380

RESUMEN

Wilms Tumor, the most common pediatric kidney cancer, evolves from the failure of terminal differentiation of the embryonic kidney. Here we show that overexpression of the heterochronic regulator Lin28 during kidney development in mice markedly expands nephrogenic progenitors by blocking their final wave of differentiation, ultimately resulting in a pathology highly reminiscent of Wilms tumor. Using lineage-specific promoters to target Lin28 to specific cell types, we observed Wilms tumor only when Lin28 is aberrantly expressed in multiple derivatives of the intermediate mesoderm, implicating the cell of origin as a multipotential renal progenitor. We show that withdrawal of Lin28 expression reverts tumorigenesis and markedly expands the numbers of glomerulus-like structures and that tumor formation is suppressed by enforced expression of Let-7 microRNA. Finally, we demonstrate overexpression of the LIN28B paralog in a significant percentage of human Wilms tumor. Our data thus implicate the Lin28/Let-7 pathway in kidney development and tumorigenesis.


Asunto(s)
Diferenciación Celular , Neoplasias Renales/genética , Neoplasias Renales/fisiopatología , Proteínas de Unión al ARN/genética , Células Madre/citología , Tumor de Wilms/genética , Tumor de Wilms/fisiopatología , Animales , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/embriología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo
5.
Expert Rev Mol Med ; 19: e8, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28716159

RESUMEN

Wilms tumour (WT) is the most common paediatric kidney cancer and affects approximately one in 10 000 children. The tumour is associated with undifferentiated embryonic lesions called nephrogenic rests (NRs) or, when diffuse, nephroblastomatosis. WT or NRs can occur in both kidneys, termed bilateral disease, found in only 5-8% of cases. Management of bilateral WT presents a major clinical challenge in terms of maximising survival, preserving renal function and understanding underlying genetic risk. In this review, we compile clinical data from 545 published cases of bilateral WT and discuss recent progress in understanding the molecular basis of bilateral WT and its associated precursor NRs in the context of the latest radiological, surgical and epidemiological features.


Asunto(s)
Tumor de Wilms/diagnóstico , Tumor de Wilms/etiología , Terapia Combinada/métodos , Diagnóstico por Imagen/métodos , Susceptibilidad a Enfermedades , Epigénesis Genética , Predisposición Genética a la Enfermedad , Humanos , Fallo Renal Crónico/etiología , Fenotipo , Resultado del Tratamiento , Tumor de Wilms/complicaciones , Tumor de Wilms/terapia
6.
Future Oncol ; 11(15): 2221-34, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26235184

RESUMEN

Wilms tumor (WT) is the most common pediatric renal tumor. Survival rates are high, whether treated according to the European protocols (SIOP-RTSG) that use prenephrectomy chemotherapy or the Children's Oncology Group (COG) protocols, with immediate nephrectomy. However, the more intensive treatment given to higher risk subgroups may result in late effects. Current risk stratification does not identify all tumors that relapse and loss of heterozygosity of 16q and 1p are the only molecular biomarkers used in risk stratification. In this review we describe recent new genetic and epigenetic findings in WT and discuss their potential use as biomarkers. We discuss approaches to ensure representative sampling of WTs including the potential for 'liquid biopsy' to circumvent intratumoral heterogeneity.


Asunto(s)
Biomarcadores de Tumor/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Tumor de Wilms/genética , Adolescente , Niño , Preescolar , Epigénesis Genética/genética , Humanos , Pronóstico , Tumor de Wilms/patología
7.
Nat Cell Biol ; 25(8): 1121-1134, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37460697

RESUMEN

The epigenetic mechanisms that maintain differentiated cell states remain incompletely understood. Here we employed histone mutants to uncover a crucial role for H3K36 methylation in the maintenance of cell identities across diverse developmental contexts. Focusing on the experimental induction of pluripotency, we show that H3K36M-mediated depletion of H3K36 methylation endows fibroblasts with a plastic state poised to acquire pluripotency in nearly all cells. At a cellular level, H3K36M facilitates epithelial plasticity by rendering fibroblasts insensitive to TGFß signals. At a molecular level, H3K36M enables the decommissioning of mesenchymal enhancers and the parallel activation of epithelial/stem cell enhancers. This enhancer rewiring is Tet dependent and redirects Sox2 from promiscuous somatic to pluripotency targets. Our findings reveal a previously unappreciated dual role for H3K36 methylation in the maintenance of cell identity by integrating a crucial developmental pathway into sustained expression of cell-type-specific programmes, and by opposing the expression of alternative lineage programmes through enhancer methylation.


Asunto(s)
Epigénesis Genética , Histonas , Metilación , Histonas/genética , Histonas/metabolismo , Diferenciación Celular/genética , Fibroblastos/metabolismo , Linaje de la Célula/genética
8.
Am J Med Genet A ; 152A(12): 3124-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21082660

RESUMEN

We describe a patient with striking generalized symmetrical enchondromatosis of the tubular bones and a de novo duplication of chromosome 12p11.23 to 12p11.22. The PTHLH gene within this region encodes a ligand for PTHR1: mutations in the gene encoding this receptor are associated with some cases of Ollier disease, several skeletal dysplasias including Blomstrand, Eiken, and Jansen and down-regulation of PTHLH expression in brachydactyly type E. Our findings suggest that abnormal PTHLH-PTHR1 signaling may underly this unusual form of enchondromatosis and indicate that unlike most cases of Ollier disease it is dominantly inherited.


Asunto(s)
Duplicación Cromosómica , Cromosomas Humanos Par 12 , Encondromatosis/genética , Proteína Relacionada con la Hormona Paratiroidea/genética , Adolescente , Enfermedades del Desarrollo Óseo/diagnóstico por imagen , Enfermedades del Desarrollo Óseo/genética , Huesos/diagnóstico por imagen , Huesos/metabolismo , Hibridación Genómica Comparativa , ADN/genética , Regulación hacia Abajo , Encondromatosis/diagnóstico por imagen , Encondromatosis/metabolismo , Femenino , Genes Dominantes , Humanos , Hibridación Fluorescente in Situ , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Radiografía , Receptor de Hormona Paratiroídea Tipo 1/genética , Transducción de Señal/genética
9.
Cell Stem Cell ; 26(1): 108-122.e10, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31839570

RESUMEN

Stem-cell-derived tissues could transform disease research and therapy, yet most methods generate functionally immature products. We investigate how human pluripotent stem cells (hPSCs) differentiate into pancreatic islets in vitro by profiling DNA methylation, chromatin accessibility, and histone modification changes. We find that enhancer potential is reset upon lineage commitment and show how pervasive epigenetic priming steers endocrine cell fates. Modeling islet differentiation and maturation regulatory circuits reveals genes critical for generating endocrine cells and identifies circadian control as limiting for in vitro islet function. Entrainment to circadian feeding/fasting cycles triggers islet metabolic maturation by inducing cyclic synthesis of energy metabolism and insulin secretion effectors, including antiphasic insulin and glucagon pulses. Following entrainment, hPSC-derived islets gain persistent chromatin changes and rhythmic insulin responses with a raised glucose threshold, a hallmark of functional maturity, and function within days of transplantation. Thus, hPSC-derived tissues are amenable to functional improvement by circadian modulation.


Asunto(s)
Diferenciación Celular , Ritmo Circadiano , Islotes Pancreáticos/citología , Células Madre Pluripotentes/citología , Glucagón/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo
10.
Nat Genet ; 52(8): 819-827, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32514123

RESUMEN

Mammalian cells stably maintain high levels of DNA methylation despite expressing both positive (DNMT3A/B) and negative (TET1-3) regulators. Here, we analyzed the independent and combined effects of these regulators on the DNA methylation landscape using a panel of knockout human embryonic stem cell (ESC) lines. The greatest impact on global methylation levels was observed in DNMT3-deficient cells, including reproducible focal demethylation at thousands of normally methylated loci. Demethylation depends on TET expression and occurs only when both DNMT3s are absent. Dynamic loci are enriched for hydroxymethylcytosine and overlap with subsets of putative somatic enhancers that are methylated in ESCs and can be activated upon differentiation. We observe similar dynamics in mouse ESCs that were less frequent in epiblast stem cells (EpiSCs) and scarce in somatic tissues, suggesting a conserved pluripotency-linked mechanism. Taken together, our data reveal tightly regulated competition between DNMT3s and TETs at thousands of somatic regulatory sequences within pluripotent cells.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Elementos de Facilitación Genéticos/genética , Oxigenasas de Función Mixta/genética , Células Madre Pluripotentes/fisiología , Proteínas Proto-Oncogénicas/genética , Animales , Diferenciación Celular/genética , Línea Celular , ADN Metiltransferasa 3A , Células Madre Embrionarias/fisiología , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica/genética , Estratos Germinativos/fisiología , Humanos , Ratones , Ratones Noqueados
11.
Nat Cell Biol ; 22(4): 389-400, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32231305

RESUMEN

In mouse embryonic stem cells (mESCs), chemical blockade of Gsk3α/ß and Mek1/2 (2i) instructs a self-renewing ground state whose endogenous inducers are unknown. Here we show that the axon guidance cue Netrin-1 promotes naive pluripotency by triggering profound signalling, transcriptomic and epigenetic changes in mESCs. Furthermore, we demonstrate that Netrin-1 can substitute for blockade of Gsk3α/ß and Mek1/2 to sustain self-renewal of mESCs in combination with leukaemia inhibitory factor and regulates the formation of the mouse pluripotent blastocyst. Mechanistically, we reveal how Netrin-1 and the balance of its receptors Neo1 and Unc5B co-regulate Wnt and MAPK pathways in both mouse and human ESCs. Netrin-1 induces Fak kinase to inactivate Gsk3α/ß and stabilize ß-catenin while increasing the phosphatase activity of a Ppp2r2c-containing Pp2a complex to reduce Erk1/2 activity. Collectively, this work identifies Netrin-1 as a regulator of pluripotency and reveals that it mediates different effects in mESCs depending on its receptor dosage, opening perspectives for balancing self-renewal and lineage commitment.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Receptores de Netrina/genética , Netrina-1/genética , Receptores de Superficie Celular/genética , Vía de Señalización Wnt/genética , Animales , Línea Celular , Embrión de Mamíferos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones SCID , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Receptores de Superficie Celular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
12.
Nat Cell Biol ; 21(11): 1449-1461, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31659274

RESUMEN

Development and differentiation are associated with profound changes to histone modifications, yet their in vivo function remains incompletely understood. Here, we generated mouse models expressing inducible histone H3 lysine-to-methionine (K-to-M) mutants, which globally inhibit methylation at specific sites. Mice expressing H3K36M developed severe anaemia with arrested erythropoiesis, a marked haematopoietic stem cell defect, and rapid lethality. By contrast, mice expressing H3K9M survived up to a year and showed expansion of multipotent progenitors, aberrant lymphopoiesis and thrombocytosis. Additionally, some H3K9M mice succumbed to aggressive T cell leukaemia/lymphoma, while H3K36M mice exhibited differentiation defects in testis and intestine. Mechanistically, induction of either mutant reduced corresponding histone trimethylation patterns genome-wide and altered chromatin accessibility as well as gene expression landscapes. Strikingly, discontinuation of transgene expression largely restored differentiation programmes. Our work shows that individual chromatin modifications are required at several specific stages of differentiation and introduces powerful tools to interrogate their roles in vivo.


Asunto(s)
Epigénesis Genética , Histonas/metabolismo , Leucemia de Células T/genética , Lisina/metabolismo , Metionina/metabolismo , Teratoma/genética , Animales , Trasplante de Médula Ósea , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Células Eritroides/metabolismo , Células Eritroides/patología , Femenino , Granulocitos/metabolismo , Granulocitos/patología , Histonas/genética , Leucemia de Células T/inducido químicamente , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Masculino , Metilación , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/patología , Mutación , Transducción de Señal , Análisis de Supervivencia , Linfocitos T/metabolismo , Linfocitos T/patología , Teratoma/inducido químicamente , Teratoma/metabolismo , Teratoma/patología
13.
Cell Stem Cell ; 25(5): 622-638.e13, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31588046

RESUMEN

Post-transcriptional mechanisms have the potential to influence complex changes in gene expression, yet their role in cell fate transitions remains largely unexplored. Here, we show that suppression of the RNA helicase DDX6 endows human and mouse primed embryonic stem cells (ESCs) with a differentiation-resistant, "hyper-pluripotent" state, which readily reprograms to a naive state resembling the preimplantation embryo. We further demonstrate that DDX6 plays a key role in adult progenitors where it controls the balance between self-renewal and differentiation in a context-dependent manner. Mechanistically, DDX6 mediates the translational suppression of target mRNAs in P-bodies. Upon loss of DDX6 activity, P-bodies dissolve and release mRNAs encoding fate-instructive transcription and chromatin factors that re-enter the ribosome pool. Increased translation of these targets impacts cell fate by rewiring the enhancer, heterochromatin, and DNA methylation landscapes of undifferentiated cell types. Collectively, our data establish a link between P-body homeostasis, chromatin organization, and stem cell potency.


Asunto(s)
Diferenciación Celular/genética , Plasticidad de la Célula/genética , ARN Helicasas DEAD-box/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ribonucleoproteínas/metabolismo , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/genética , ARN Helicasas DEAD-box/genética , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/genética , Ontología de Genes , Homeostasis/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/enzimología , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Homeótica Nanog/metabolismo , Organoides/citología , Organoides/diagnóstico por imagen , Organoides/metabolismo , Biosíntesis de Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , RNA-Seq , Ribonucleoproteínas/genética , Ribosomas/metabolismo
14.
Elife ; 72018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30468428

RESUMEN

DNA methylation plays an essential role in mammalian genomes and expression of the responsible enzymes is tightly controlled. Deregulation of the de novo DNA methyltransferase DNMT3B is frequently observed across cancer types, yet little is known about its ectopic genomic targets. Here, we used an inducible transgenic mouse model to delineate rules for abnormal DNMT3B targeting, as well as the constraints of its activity across different cell types. Our results explain the preferential susceptibility of certain CpG islands to aberrant methylation and point to transcriptional state and the associated chromatin landscape as the strongest predictors. Although DNA methylation and H3K27me3 are usually non-overlapping at CpG islands, H3K27me3 can transiently co-occur with DNMT3B-induced DNA methylation. Our genome-wide data combined with ultra-deep locus-specific bisulfite sequencing suggest a distributive activity of ectopically expressed Dnmt3b that leads to discordant CpG island hypermethylation and provides new insights for interpreting the cancer methylome.


Asunto(s)
Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Metilación de ADN , Expresión Génica , Proteínas Recombinantes/biosíntesis , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias/fisiología , Regulación de la Expresión Génica , Humanos , Ratones Transgénicos , Neoplasias/patología , Proteínas Recombinantes/genética , ADN Metiltransferasa 3B
15.
Nat Struct Mol Biol ; 25(4): 327-332, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29531288

RESUMEN

Cytosine methylation is widespread among organisms and essential for mammalian development. In line with early postulations of an epigenetic role in gene regulation, symmetric CpG methylation can be mitotically propagated over many generations with extraordinarily high fidelity. Here, we combine BrdU labeling and immunoprecipitation with genome-wide bisulfite sequencing to explore the inheritance of cytosine methylation onto newly replicated DNA in human cells. Globally, we observe a pronounced lag between the copying of genetic and epigenetic information in embryonic stem cells that is reconsolidated within hours to accomplish faithful mitotic transmission. Populations of arrested cells show a global reduction of lag-induced intermediate CpG methylation when compared to proliferating cells, whereas sites of transcription factor engagement appear cell-cycle invariant. Alternatively, the cancer cell line HCT116 preserves global epigenetic heterogeneity independently of cell-cycle arrest. Taken together, our data suggest that heterogeneous methylation largely reflects asynchronous proliferation, but is intrinsic to actively engaged cis-regulatory elements and cancer.


Asunto(s)
Citosina/química , Metilación de ADN , Ciclo Celular , Proliferación Celular , Islas de CpG , ADN/química , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Replicación del ADN , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación de la Expresión Génica , Genoma Humano , Células HCT116 , Humanos , Masculino , Metilación , Mitosis , Neuronas Motoras/metabolismo , Neoplasias/genética , Análisis de Secuencia de ARN , Factores de Transcripción/metabolismo , ADN Metiltransferasa 3B
16.
Nat Commun ; 9(1): 597, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29426832

RESUMEN

In normal mammalian development cytosine methylation is essential and is directed to specific regions of the genome. Despite notable advances through mapping its genome-wide distribution, studying the direct contribution of DNA methylation to gene and genome regulation has been limited by the lack of tools for its precise manipulation. Thus, combining the targeting capability of the CRISPR-Cas9 system with an epigenetic modifier has attracted interest in the scientific community. In contrast to profiling the genome-wide cleavage of a nuclease competent Cas9, tracing the global activity of a dead Cas9 (dCas9) methyltransferase fusion protein is challenging within a highly methylated genome. Here, we report the generation and use of an engineered, methylation depleted but maintenance competent mouse ES cell line and find surprisingly ubiquitous nuclear activity of dCas9-methyltransferases. Subsequent experiments in human somatic cells refine these observations and point to an important difference between genetic and epigenetic editing tools that require unique experimental considerations.


Asunto(s)
Línea Celular , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Embrionarias/enzimología , Animales , Proteínas Bacterianas , Proteína 9 Asociada a CRISPR , Endonucleasas , Edición Génica , Humanos , Ratones
17.
Nat Struct Mol Biol ; 25(4): 355, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29581568

RESUMEN

Following online publication of this article, the Gene Expression Omnibus records corresponding to accession codes GSM2406773, MN-d6, and GSM2406772, MN-d14, listed in the data availability statement were deleted. The data are now available under accession codes GSM3039355, WGBS_hESC_WT_D6_R4 (MN day 6), and GSM3039351, WGBS_hESC_WT_D14_R4 (MN day 14), and the data availability statement has been updated with the new accession codes in the HTML and PDF versions of the article.

18.
Nat Genet ; 50(2): 250-258, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29358654

RESUMEN

Transcription factors (TFs) direct developmental transitions by binding to target DNA sequences, influencing gene expression and establishing complex gene-regultory networks. To systematically determine the molecular components that enable or constrain TF activity, we investigated the genomic occupancy of FOXA2, GATA4 and OCT4 in several cell types. Despite their classification as pioneer factors, all three TFs exhibit cell-type-specific binding, even when supraphysiologically and ectopically expressed. However, FOXA2 and GATA4 can be distinguished by low enrichment at loci that are highly occupied by these factors in alternative cell types. We find that expression of additional cofactors increases enrichment at a subset of these sites. Finally, FOXA2 occupancy and changes to DNA accessibility can occur in G1-arrested cells, but subsequent loss of DNA methylation requires DNA replication.


Asunto(s)
ADN/metabolismo , Epigénesis Genética/fisiología , Redes Reguladoras de Genes/fisiología , Factores de Transcripción/metabolismo , Células A549 , Sitios de Unión/genética , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Cultivadas , Biología Computacional , ADN/genética , Epistasis Genética/fisiología , Factor de Transcripción GATA4/metabolismo , Regulación de la Expresión Génica , Genes de Cambio , Células HEK293 , Células Hep G2 , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Unión Proteica
19.
Methods Mol Biol ; 1467: 1-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27417955

RESUMEN

In this chapter, the role of WT1 in childhood cancer is discussed, using the key examples Wilms' tumor, desmoplastic small round cell of childhood, and leukemia. The role of WT1 in each disease is described and mirrored to the role of WT1 in normal development.


Asunto(s)
Tumor Desmoplásico de Células Pequeñas Redondas/genética , Leucemia/genética , Proteínas WT1/genética , Tumor de Wilms/genética , Adolescente , Niño , Preescolar , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Riñón/crecimiento & desarrollo , Mutación
20.
Genome Med ; 7(1): 11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25763109

RESUMEN

BACKGROUND: Wilms tumours (WTs) are characterised by several hallmarks that suggest epimutations such as aberrant DNA methylation are involved in tumour progression: loss of imprinting at 11p15, lack of recurrent mutations and formation of nephrogenic rests (NRs), which are lesions of retained undifferentiated embryonic tissue that can give rise to WTs. METHODS: To identify such epimutations, we performed a comprehensive methylome analysis on 20 matched trios of micro-dissected WTs, NRs and surrounding normal kidneys (NKs) using Illumina Infinium HumanMethylation450 Bead Chips and functionally validated findings using RNA sequencing. RESULTS: Comparison of NRs with NK revealed prominent tissue biomarkers: 629 differentially methylated regions, of which 55% were hypermethylated and enriched for domains that are bivalent in embryonic stem cells and for genes expressed during development (P = 2.49 × 10(-5)). Comparison of WTs with NRs revealed two WT subgroups; group-2 WTs and NRs were epigenetically indistinguishable whereas group-1 WTs showed an increase in methylation variability, hypomethylation of renal development genes, hypermethylation and relative loss of expression of cell adhesion genes and known and potential new WT tumour suppressor genes (CASP8, H19, MIR195, RB1 and TSPAN32) and was strongly associated with bilateral disease (P = 0.032). Comparison of WTs and NRs to embryonic kidney highlighted the significance of polycomb target methylation in Wilms tumourigenesis. CONCLUSIONS: Methylation levels vary during cancer evolution. We have described biomarkers related to WT evolution from its precursor NRs which may be useful to differentiate between these tissues for patients with bilateral disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA