Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 333
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(4): 928-943.e22, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30712874

RESUMEN

Understanding the molecular programs that guide differentiation during development is a major challenge. Here, we introduce Waddington-OT, an approach for studying developmental time courses to infer ancestor-descendant fates and model the regulatory programs that underlie them. We apply the method to reconstruct the landscape of reprogramming from 315,000 single-cell RNA sequencing (scRNA-seq) profiles, collected at half-day intervals across 18 days. The results reveal a wider range of developmental programs than previously characterized. Cells gradually adopt either a terminal stromal state or a mesenchymal-to-epithelial transition state. The latter gives rise to populations related to pluripotent, extra-embryonic, and neural cells, with each harboring multiple finer subpopulations. The analysis predicts transcription factors and paracrine signals that affect fates and experiments validate that the TF Obox6 and the cytokine GDF9 enhance reprogramming efficiency. Our approach sheds light on the process and outcome of reprogramming and provides a framework applicable to diverse temporal processes in biology.


Asunto(s)
Reprogramación Celular/genética , Perfilación de la Expresión Génica/métodos , Análisis de la Célula Individual/métodos , Animales , Diferenciación Celular/genética , Células Cultivadas , Células Madre Embrionarias/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Análisis de Secuencia de ARN/métodos , Factores de Transcripción/metabolismo
2.
Cell ; 175(3): 615-632, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340033

RESUMEN

The derivation of human embryonic stem cells (hESCs) and the stunning discovery that somatic cells can be reprogrammed into human induced pluripotent stem cells (hiPSCs) holds the promise to revolutionize biomedical research and regenerative medicine. In this Review, we focus on disorders of the central nervous system and explore how advances in human pluripotent stem cells (hPSCs) coincide with evolutions in genome engineering and genomic technologies to provide realistic opportunities to tackle some of the most devastating complex disorders.


Asunto(s)
Enfermedades del Sistema Nervioso Central/terapia , Edición Génica/métodos , Trasplante de Células Madre/métodos , Investigación Biomédica Traslacional/métodos , Animales , Enfermedades del Sistema Nervioso Central/genética , Humanos
3.
Cell ; 172(5): 979-992.e6, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29456084

RESUMEN

Fragile X syndrome (FXS), the most common genetic form of intellectual disability in males, is caused by silencing of the FMR1 gene associated with hypermethylation of the CGG expansion mutation in the 5' UTR of FMR1 in FXS patients. Here, we applied recently developed DNA methylation editing tools to reverse this hypermethylation event. Targeted demethylation of the CGG expansion by dCas9-Tet1/single guide RNA (sgRNA) switched the heterochromatin status of the upstream FMR1 promoter to an active chromatin state, restoring a persistent expression of FMR1 in FXS iPSCs. Neurons derived from methylation-edited FXS iPSCs rescued the electrophysiological abnormalities and restored a wild-type phenotype upon the mutant neurons. FMR1 expression in edited neurons was maintained in vivo after engrafting into the mouse brain. Finally, demethylation of the CGG repeats in post-mitotic FXS neurons also reactivated FMR1. Our data establish that demethylation of the CGG expansion is sufficient for FMR1 reactivation, suggesting potential therapeutic strategies for FXS.


Asunto(s)
Metilación de ADN/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Edición Génica , Neuronas/patología , Animales , Proteína 9 Asociada a CRISPR/metabolismo , Epigénesis Genética , Células HEK293 , Heterocromatina/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Masculino , Ratones , Neuronas/metabolismo , Fenotipo , Regiones Promotoras Genéticas , ARN Guía de Kinetoplastida/metabolismo , Expansión de Repetición de Trinucleótido/genética
4.
Cell ; 171(7): 1573-1588.e28, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29224777

RESUMEN

There is considerable evidence that chromosome structure plays important roles in gene control, but we have limited understanding of the proteins that contribute to structural interactions between gene promoters and their enhancer elements. Large DNA loops that encompass genes and their regulatory elements depend on CTCF-CTCF interactions, but most enhancer-promoter interactions do not employ this structural protein. Here, we show that the ubiquitously expressed transcription factor Yin Yang 1 (YY1) contributes to enhancer-promoter structural interactions in a manner analogous to DNA interactions mediated by CTCF. YY1 binds to active enhancers and promoter-proximal elements and forms dimers that facilitate the interaction of these DNA elements. Deletion of YY1 binding sites or depletion of YY1 protein disrupts enhancer-promoter looping and gene expression. We propose that YY1-mediated enhancer-promoter interactions are a general feature of mammalian gene control.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Factor de Transcripción YY1/metabolismo , Animales , Factor de Unión a CCCTC/metabolismo , Células Madre Embrionarias/metabolismo , Humanos , Ratones
5.
Cell ; 167(1): 233-247.e17, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27662091

RESUMEN

Mammalian DNA methylation is a critical epigenetic mechanism orchestrating gene expression networks in many biological processes. However, investigation of the functions of specific methylation events remains challenging. Here, we demonstrate that fusion of Tet1 or Dnmt3a with a catalytically inactive Cas9 (dCas9) enables targeted DNA methylation editing. Targeting of the dCas9-Tet1 or -Dnmt3a fusion protein to methylated or unmethylated promoter sequences caused activation or silencing, respectively, of an endogenous reporter. Targeted demethylation of the BDNF promoter IV or the MyoD distal enhancer by dCas9-Tet1 induced BDNF expression in post-mitotic neurons or activated MyoD facilitating reprogramming of fibroblasts into myoblasts, respectively. Targeted de novo methylation of a CTCF loop anchor site by dCas9-Dnmt3a blocked CTCF binding and interfered with DNA looping, causing altered gene expression in the neighboring loop. Finally, we show that these tools can edit DNA methylation in mice, demonstrating their wide utility for functional studies of epigenetic regulation.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Edición Génica/métodos , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor de Unión a CCCTC , Proteína 9 Asociada a CRISPR , Línea Celular , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Endonucleasas/metabolismo , Elementos de Facilitación Genéticos , Genoma , Ratones , Proteína MioD/metabolismo , Neuronas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/metabolismo
6.
Cell ; 163(1): 218-29, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26406378

RESUMEN

Mammalian DNA methylation plays an essential role in development. To date, only snapshots of different mouse and human cell types have been generated, providing a static view on DNA methylation. To enable monitoring of methylation status as it changes over time, we establish a reporter of genomic methylation (RGM) that relies on a minimal imprinted gene promoter driving a fluorescent protein. We show that insertion of RGM proximal to promoter-associated CpG islands reports the gain or loss of DNA methylation. We further utilized RGM to report endogenous methylation dynamics of non-coding regulatory elements, such as the pluripotency-specific super enhancers of Sox2 and miR290. Loci-specific DNA methylation changes and their correlation with transcription were visualized during cell-state transition following differentiation of mouse embryonic stem cells and during reprogramming of somatic cells to pluripotency. RGM will allow the investigation of dynamic methylation changes during development and disease at single-cell resolution.


Asunto(s)
Metilación de ADN , Análisis de la Célula Individual , Animales , Islas de CpG , Metilasas de Modificación del ADN/metabolismo , Células Madre Embrionarias , Elementos de Facilitación Genéticos , Humanos , Ratones , MicroARNs/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/metabolismo
8.
Cell ; 154(6): 1370-9, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-23992847

RESUMEN

The type II bacterial CRISPR/Cas system is a novel genome-engineering technology with the ease of multiplexed gene targeting. Here, we created reporter and conditional mutant mice by coinjection of zygotes with Cas9 mRNA and different guide RNAs (sgRNAs) as well as DNA vectors of different sizes. Using this one-step procedure we generated mice carrying a tag or a fluorescent reporter construct in the Nanog, the Sox2, and the Oct4 gene as well as Mecp2 conditional mutant mice. In addition, using sgRNAs targeting two separate sites in the Mecp2 gene, we produced mice harboring the predicted deletions of about 700 bps. Finally, we analyzed potential off-targets of five sgRNAs in gene-modified mice and ESC lines and identified off-target mutations in only rare instances.


Asunto(s)
Marcación de Gen/métodos , Ratones/genética , Animales , Secuencia de Bases , Ingeniería Genética , Mutación
9.
Cell ; 153(4): 910-8, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23643243

RESUMEN

Mice carrying mutations in multiple genes are traditionally generated by sequential recombination in embryonic stem cells and/or time-consuming intercrossing of mice with a single mutation. The CRISPR/Cas system has been adapted as an efficient gene-targeting technology with the potential for multiplexed genome editing. We demonstrate that CRISPR/Cas-mediated gene editing allows the simultaneous disruption of five genes (Tet1, 2, 3, Sry, Uty--8 alleles) in mouse embryonic stem (ES) cells with high efficiency. Coinjection of Cas9 mRNA and single-guide RNAs (sgRNAs) targeting Tet1 and Tet2 into zygotes generated mice with biallelic mutations in both genes with an efficiency of 80%. Finally, we show that coinjection of Cas9 mRNA/sgRNAs with mutant oligos generated precise point mutations simultaneously in two target genes. Thus, the CRISPR/Cas system allows the one-step generation of animals carrying mutations in multiple genes, an approach that will greatly accelerate the in vivo study of functionally redundant genes and of epistatic gene interactions.


Asunto(s)
Marcación de Gen/métodos , Ratones/genética , Animales , Secuencia de Bases , Células Madre Embrionarias/metabolismo , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , ARN Pequeño no Traducido
10.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24290359

RESUMEN

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Asunto(s)
Interacción Gen-Ambiente , Mitocondrias/efectos de los fármacos , Paraquat/toxicidad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factores de Transcripción MEF2 , Mutación/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Especies de Nitrógeno Reactivo/metabolismo , Sustancia Negra/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
11.
Nat Immunol ; 16(6): 653-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25867473

RESUMEN

The methylcytosine dioxygenase TET1 ('ten-eleven translocation 1') is an important regulator of 5-hydroxymethylcytosine (5hmC) in embryonic stem cells. The diminished expression of TET proteins and loss of 5hmC in many tumors suggests a critical role for the maintenance of this epigenetic modification. Here we found that deletion of Tet1 promoted the development of B cell lymphoma in mice. TET1 was required for maintenance of the normal abundance and distribution of 5hmC, which prevented hypermethylation of DNA, and for regulation of the B cell lineage and of genes encoding molecules involved in chromosome maintenance and DNA repair. Whole-exome sequencing of TET1-deficient tumors revealed mutations frequently found in non-Hodgkin B cell lymphoma (B-NHL), in which TET1 was hypermethylated and transcriptionally silenced. Our findings provide in vivo evidence of a function for TET1 as a tumor suppressor of hematopoietic malignancy.


Asunto(s)
Linfocitos B/fisiología , Citosina/análogos & derivados , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/fisiología , Linfoma de Células B/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Inestabilidad Cromosómica , Citosina/metabolismo , Metilación de ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Epigénesis Genética , Exoma/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética
12.
Cell ; 150(6): 1209-22, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980981

RESUMEN

During cellular reprogramming, only a small fraction of cells become induced pluripotent stem cells (iPSCs). Previous analyses of gene expression during reprogramming were based on populations of cells, impeding single-cell level identification of reprogramming events. We utilized two gene expression technologies to profile 48 genes in single cells at various stages during the reprogramming process. Analysis of early stages revealed considerable variation in gene expression between cells in contrast to late stages. Expression of Esrrb, Utf1, Lin28, and Dppa2 is a better predictor for cells to progress into iPSCs than expression of the previously suggested reprogramming markers Fbxo15, Fgf4, and Oct4. Stochastic gene expression early in reprogramming is followed by a late hierarchical phase with Sox2 being the upstream factor in a gene expression hierarchy. Finally, downstream factors derived from the late phase, which do not include Oct4, Sox2, Klf4, c-Myc, and Nanog, can activate the pluripotency circuitry.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Línea Celular , Embrión de Mamíferos/citología , Células Madre Embrionarias , Fibroblastos/citología , Fibroblastos/metabolismo , Marcadores Genéticos , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Ratones , Técnicas Analíticas Microfluídicas , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/metabolismo
13.
Mol Cell ; 75(5): 905-920.e6, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31422875

RESUMEN

Variable levels of DNA methylation have been reported at tissue-specific differential methylation regions (DMRs) overlapping enhancers, including super-enhancers (SEs) associated with key cell identity genes, but the mechanisms responsible for this intriguing behavior are not well understood. We used allele-specific reporters at the endogenous Sox2 and Mir290 SEs in embryonic stem cells and found that the allelic DNA methylation state is dynamically switching, resulting in cell-to-cell heterogeneity. Dynamic DNA methylation is driven by the balance between DNA methyltransferases and transcription factor binding on one side and co-regulated with the Mediator complex recruitment and H3K27ac level changes at regulatory elements on the other side. DNA methylation at the Sox2 and the Mir290 SEs is independently regulated and has distinct consequences on the cellular differentiation state. Dynamic allele-specific DNA methylation at the two SEs was also seen at different stages in preimplantation embryos, revealing that methylation heterogeneity occurs in vivo.


Asunto(s)
Diferenciación Celular/fisiología , Metilación de ADN/fisiología , Elementos de Facilitación Genéticos/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Transcripción Genética/fisiología , Animales , Línea Celular , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
14.
Mol Cell ; 73(5): 1001-1014.e8, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30527540

RESUMEN

In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.


Asunto(s)
Antiparkinsonianos/farmacología , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Metabolómica/métodos , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Estearoil-CoA Desaturasa/antagonistas & inhibidores , alfa-Sinucleína/toxicidad , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/enzimología , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Degeneración Nerviosa , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/enzimología , Células-Madre Neurales/patología , Neuronas/enzimología , Neuronas/patología , Ácido Oléico/metabolismo , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Ratas Sprague-Dawley , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Estearoil-CoA Desaturasa/metabolismo , Triglicéridos/metabolismo , alfa-Sinucleína/genética
15.
Nat Rev Neurosci ; 22(5): 290-307, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33772226

RESUMEN

GABAergic inhibition shapes the connectivity, activity and plasticity of the brain. A series of exciting new discoveries provides compelling evidence that disruptions in a number of key facets of GABAergic inhibition have critical roles in the aetiology of neurodevelopmental disorders (NDDs). These facets include the generation, migration and survival of GABAergic neurons, the formation of GABAergic synapses and circuit connectivity, and the dynamic regulation of the efficacy of GABAergic signalling through neuronal chloride transporters. In this Review, we discuss recent work that elucidates the functions and dysfunctions of GABAergic signalling in health and disease, that uncovers the contribution of GABAergic neural circuit dysfunction to NDD aetiology and that leverages such mechanistic insights to advance precision medicine for the treatment of NDDs.


Asunto(s)
Trastornos del Neurodesarrollo/fisiopatología , Transducción de Señal , Ácido gamma-Aminobutírico , Neuronas GABAérgicas , Humanos , Red Nerviosa/fisiopatología , Trastornos del Neurodesarrollo/terapia , Medicina de Precisión
16.
Cell ; 146(2): 318-31, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21757228

RESUMEN

Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinson's disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies.


Asunto(s)
Enfermedad de Parkinson/patología , Células Madre Pluripotentes , Mutación Puntual , Línea Celular , Células Madre Embrionarias , Ingeniería Genética , Estudio de Asociación del Genoma Completo , Humanos , Mutagénesis , Oligonucleótidos/metabolismo , alfa-Sinucleína/genética
17.
Nature ; 586(7829): 440-444, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32698189

RESUMEN

Methyl CpG binding protein 2 (MeCP2) is a key component of constitutive heterochromatin, which is crucial for chromosome maintenance and transcriptional silencing1-3. Mutations in the MECP2 gene cause the progressive neurodevelopmental disorder Rett syndrome3-5, which is associated with severe mental disability and autism-like symptoms that affect girls during early childhood. Although previously thought to be a dense and relatively static structure1,2, heterochromatin is now understood to exhibit properties consistent with a liquid-like condensate6,7. Here we show that MeCP2 is a dynamic component of heterochromatin condensates in cells, and is stimulated by DNA to form liquid-like condensates. MeCP2 contains several domains that contribute to the formation of condensates, and mutations in MECP2 that lead to Rett syndrome disrupt the ability of MeCP2 to form condensates. Condensates formed by MeCP2 selectively incorporate and concentrate heterochromatin cofactors rather than components of euchromatic transcriptionally active condensates. We propose that MeCP2 enhances the separation of heterochromatin and euchromatin through its condensate partitioning properties, and that disruption of condensates may be a common consequence of mutations in MeCP2 that cause Rett syndrome.


Asunto(s)
Heterocromatina/metabolismo , Discapacidad Intelectual/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Mutación , Inmunidad Adaptativa , Animales , Femenino , Inmunidad Innata , Discapacidad Intelectual/patología , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Síndrome de Rett/genética
18.
Rev Med Virol ; 34(1): e2488, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37921610

RESUMEN

Bat-borne viruses have attracted considerable research, especially in relation to the Covid-19 pandemic. Although bats can carry multiple zoonotic viruses that are lethal to many mammalian species, they appear to be asymptomatic to viral infection despite the high viral loads contained in their bodies. There are several differences between bats and other mammals. One of the major differences between bats and other mammals is the bats' ability to fly, which is believed to have induced evolutionary changes. It may have also favoured them as suitable hosts for viruses. This is related to their tolerance to viral infection. Innate immunity is the first line of defence against viral infection, but bats have metamorphosed the type of responses induced by innate immunity factors such as interferons. The expression patterns of interferons differ, as do those of interferon-related genes such as interferon regulatory factors and interferon-stimulated genes that contribute to the antiviral response of infected cells. In addition, the signalling pathways related to viral infection and immune responses have been subject to evolutionary changes, including mutations compared to their homologues in other mammals and gene selection. This article discusses the differences in the interferon-mediated antiviral response in bats compared to that of other mammals and how these differences are correlated to viral tolerance in bats. The effect of bat interferons related genes on human antiviral response against bat-borne viruses is also discussed.


Asunto(s)
Quirópteros , Virosis , Virus , Animales , Humanos , Línea Celular , Pandemias , Interferones/genética , Virosis/tratamiento farmacológico , Virosis/genética , Antivirales/farmacología , Antivirales/uso terapéutico , Antivirales/metabolismo , Genómica
19.
Cell ; 143(4): 508-25, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-21074044

RESUMEN

Direct reprogramming of somatic cells to induced pluripotent stem cells by ectopic expression of defined transcription factors has raised fundamental questions regarding the epigenetic stability of the differentiated cell state. In addition, evidence has accumulated that distinct states of pluripotency can interconvert through the modulation of both cell-intrinsic and exogenous factors. To fully realize the potential of in vitro reprogrammed cells, we need to understand the molecular and epigenetic determinants that convert one cell type into another. Here we review recent advances in this rapidly moving field and emphasize unresolved and controversial questions.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Células Madre Pluripotentes/citología , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes/metabolismo
20.
Cell ; 141(5): 872-83, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20471072

RESUMEN

The presence of two active X chromosomes (XaXa) is a hallmark of the ground state of pluripotency specific to murine embryonic stem cells (ESCs). Human ESCs (hESCs) invariably exhibit signs of X chromosome inactivation (XCI) and are considered developmentally more advanced than their murine counterparts. We describe the establishment of XaXa hESCs derived under physiological oxygen concentrations. Using these cell lines, we demonstrate that (1) differentiation of hESCs induces random XCI in a manner similar to murine ESCs, (2) chronic exposure to atmospheric oxygen is sufficient to induce irreversible XCI with minor changes of the transcriptome, (3) the Xa exhibits heavy methylation of the XIST promoter region, and (4) XCI is associated with demethylation and transcriptional activation of XIST along with H3K27-me3 deposition across the Xi. These findings indicate that the human blastocyst contains pre-X-inactivation cells and that this state is preserved in vitro through culture under physiological oxygen.


Asunto(s)
Cromosomas Humanos X/metabolismo , Células Madre Embrionarias/metabolismo , Oxígeno/metabolismo , Inactivación del Cromosoma X , Animales , Diferenciación Celular , Femenino , Histonas/metabolismo , Humanos , Cariotipificación , Masculino , Ratones , Estrés Oxidativo , Células Madre Pluripotentes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA