Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 4109, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37433783

RESUMEN

Genetic variants in chromatin regulators are frequently found in neurodevelopmental disorders, but their effect in disease etiology is rarely determined. Here, we uncover and functionally define pathogenic variants in the chromatin modifier EZH1 as the cause of dominant and recessive neurodevelopmental disorders in 19 individuals. EZH1 encodes one of the two alternative histone H3 lysine 27 methyltransferases of the PRC2 complex. Unlike the other PRC2 subunits, which are involved in cancers and developmental syndromes, the implication of EZH1 in human development and disease is largely unknown. Using cellular and biochemical studies, we demonstrate that recessive variants impair EZH1 expression causing loss of function effects, while dominant variants are missense mutations that affect evolutionarily conserved aminoacids, likely impacting EZH1 structure or function. Accordingly, we found increased methyltransferase activity leading to gain of function of two EZH1 missense variants. Furthermore, we show that EZH1 is necessary and sufficient for differentiation of neural progenitor cells in the developing chick embryo neural tube. Finally, using human pluripotent stem cell-derived neural cultures and forebrain organoids, we demonstrate that EZH1 variants perturb cortical neuron differentiation. Overall, our work reveals a critical role of EZH1 in neurogenesis regulation and provides molecular diagnosis for previously undefined neurodevelopmental disorders.


Asunto(s)
Trastornos del Neurodesarrollo , Neurogénesis , Complejo Represivo Polycomb 2 , Animales , Embrión de Pollo , Humanos , Diferenciación Celular/genética , Núcleo Celular , Cromatina/genética , Metiltransferasas , Trastornos del Neurodesarrollo/genética , Neurogénesis/genética , Complejo Represivo Polycomb 2/genética
2.
Nat Commun ; 13(1): 3263, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672304

RESUMEN

Enhancers are key regulatory elements that govern gene expression programs in response to developmental signals. However, how multiple enhancers arrange in the 3D-space to control the activation of a specific promoter remains unclear. To address this question, we exploited our previously characterized TGFß-response model, the neural stem cells, focusing on a ~374 kb locus where enhancers abound. Our 4C-seq experiments reveal that the TGFß pathway drives the assembly of an enhancer-cluster and precise gene activation. We discover that the TGFß pathway coactivator JMJD3 is essential to maintain these structures. Using live-cell imaging techniques, we demonstrate that an intrinsically disordered region contained in JMJD3 is involved in the formation of phase-separated biomolecular condensates, which are found in the enhancer-cluster. Overall, in this work we uncover novel functions for the coactivator JMJD3, and we shed light on the relationships between the 3D-conformation of the chromatin and the TGFß-driven response during mammalian neurogenesis.


Asunto(s)
Células-Madre Neurales , Factor de Crecimiento Transformador beta , Animales , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Expresión Génica , Genoma , Mamíferos/genética , Células-Madre Neurales/metabolismo , Activación Transcripcional/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
3.
Nat Rev Mol Cell Biol ; 23(7): 481-497, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35228718

RESUMEN

Transposable elements (TEs) comprise about half of the mammalian genome. TEs often contain sequences capable of recruiting the host transcription machinery, which they use to express their own products and promote transposition. However, the regulatory sequences carried by TEs may affect host transcription long after the TEs have lost the ability to transpose. Recent advances in genome analysis and engineering have facilitated systematic interrogation of the regulatory activities of TEs. In this Review, we discuss diverse mechanisms by which TEs contribute to transcription regulation. Notably, TEs can donate enhancer and promoter sequences that influence the expression of host genes, modify 3D chromatin architecture and give rise to novel regulatory genes, including non-coding RNAs and transcription factors. We discuss how TEs spur regulatory evolution and facilitate the emergence of genetic novelties in mammalian physiology and development. By virtue of their repetitive and interspersed nature, TEs offer unique opportunities to dissect the effects of mutation and genomic context on the function and evolution of cis-regulatory elements. We argue that TE-centric studies hold the key to unlocking general principles of transcription regulation and evolution.


Asunto(s)
Elementos Transponibles de ADN , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Elementos Transponibles de ADN/genética , Evolución Molecular , Regulación de la Expresión Génica/genética , Mamíferos/genética , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética
4.
Elife ; 72018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-30095408

RESUMEN

Class II HLH proteins heterodimerize with class I HLH/E proteins to regulate transcription. Here, we show that E proteins sharpen neurogenesis by adjusting the neurogenic strength of the distinct proneural proteins. We find that inhibiting BMP signaling or its target ID2 in the chick embryo spinal cord, impairs the neuronal production from progenitors expressing ATOH1/ASCL1, but less severely that from progenitors expressing NEUROG1/2/PTF1a. We show this context-dependent response to result from the differential modulation of proneural proteins' activity by E proteins. E proteins synergize with proneural proteins when acting on CAGSTG motifs, thereby facilitating the activity of ASCL1/ATOH1 which preferentially bind to such motifs. Conversely, E proteins restrict the neurogenic strength of NEUROG1/2 by directly inhibiting their preferential binding to CADATG motifs. Since we find this mechanism to be conserved in corticogenesis, we propose this differential co-operation of E proteins with proneural proteins as a novel though general feature of their mechanism of action.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neurogénesis , Animales , Sitios de Unión , Embrión de Pollo , Unión Proteica
5.
Nucleic Acids Res ; 46(7): 3351-3365, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29438503

RESUMEN

During neurogenesis, dynamic developmental cues, transcription factors and histone modifying enzymes regulate the gene expression programs by modulating the activity of neural-specific enhancers. How transient developmental signals coordinate transcription factor recruitment to enhancers and to which extent chromatin modifiers contribute to enhancer activity is starting to be uncovered. Here, we take advantage of neural stem cells as a model to unravel the mechanisms underlying neural enhancer activation in response to the TGFß signaling. Genome-wide experiments demonstrate that the proneural factor ASCL1 assists SMAD3 in the binding to a subset of enhancers. Once located at the enhancers, SMAD3 recruits the histone demethylase JMJD3 and the remodeling factor CHD8, creating the appropriate chromatin landscape to allow enhancer transcription and posterior gene activation. Finally, to analyze the phenotypical traits owed to cis-regulatory regions, we use CRISPR-Cas9 technology to demonstrate that the TGFß-responsive Neurog2 enhancer is essential for proper neuronal polarization.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Elementos de Facilitación Genéticos/genética , Neurogénesis/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Animales , Sistemas CRISPR-Cas/genética , Linaje de la Célula/genética , Polaridad Celular/genética , Proteínas de Unión al ADN/genética , Epigénesis Genética , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal/genética , Factores de Transcripción/genética
6.
Epigenetics ; 13(3): 331-341, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29384431

RESUMEN

The Barcelona Conference on Epigenetics and Cancer (BCEC) entitled "Epigenetic Mechanisms in Health and Disease" was held in Barcelona, October 26-26, 2017. The 2017 BCEC was the fifth and last edition of a series of annual conferences organized as a joint effort of five leading Barcelona research institutes together with B-Debate. This edition was organized by Albert Jordan from the Molecular Biology Institute of Barcelona (IBMB-CSIC) and Marcus Bushbeck from the Josep Carreras Leukaemia Research Institute (IJC). Jordi Bernués, Marian Martínez-Balbás, and Ferran Azorín were also part of the scientific committee. In 22 talks and 51 posters, researchers presented their latest results in the fields of histone variants, epigenetic regulation, and chromatin 3D organization to an audience of around 250 participants from 16 countries. This year, a broad number of talks focused on the epigenetic causes and possible related treatments of complex diseases such as cancer. Participants at the 2017 BCEC elegantly closed the series, discussing progress made in the field of epigenetics and highlighting its role in human health and disease.


Asunto(s)
Cromatina/genética , Metilación de ADN/genética , Epigénesis Genética , Humanos , Neoplasias/genética
7.
Oncotarget ; 8(48): 83384-83406, 2017 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-29137351

RESUMEN

MicroRNAs are critical regulators of gene networks in normal and abnormal biological processes. Focusing on invasive ductal breast cancer (IDC), we have found dysregulated expression in tumor samples of several microRNAs, including the miR-200 family, along progression from primary tumors to distant metastases, further reflected in higher blood levels of miR-200b and miR-7 in IDC patients with regional or distant metastases relative to patients with primary node-negative tumors. Forced expression of miR-200s in MCF10CA1h mammary cells induced an enhanced epithelial program, aldehyde dehydrogenase (ALDH) activity, mammosphere growth and ability to form branched tubuloalveolar structures while promoting orthotopic tumor growth and lung colonization in vivo. MiR-200s also induced the constitutive activation of the PI3K-Akt signaling through downregulation of PTEN, and the enhanced mammosphere growth and ALDH activity induced in MCF10CA1h cells by miR-200s required the activation of this signaling pathway. Interestingly, the morphology of tumors formed in vivo by cells expressing miR-200s was reminiscent of metaplastic breast cancer (MBC). Indeed, the epithelial components of MBC samples expressed significantly higher levels of miR-200s than their mesenchymal components and displayed a marker profile compatible with luminal progenitor cells. We propose that microRNAs of the miR-200 family promote traits of highly proliferative breast luminal progenitor cells, thereby exacerbating the growth and metastatic properties of transformed mammary epithelial cells.

8.
Cell Stem Cell ; 20(5): 689-705.e9, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28285903

RESUMEN

Poised enhancers marked by H3K27me3 in pluripotent stem cells have been implicated in the establishment of somatic expression programs during embryonic stem cell (ESC) differentiation. However, the functional relevance and mechanism of action of poised enhancers remain unknown. Using CRISPR/Cas9 technology to engineer precise genetic deletions, we demonstrate that poised enhancers are necessary for the induction of major anterior neural regulators. Interestingly, circularized chromosome conformation capture sequencing (4C-seq) shows that poised enhancers already establish physical interactions with their target genes in ESCs in a polycomb repressive complex 2 (PRC2)-dependent manner. Loss of PRC2 does not activate poised enhancers or induce their putative target genes in undifferentiated ESCs; however, loss of PRC2 in differentiating ESCs severely and specifically compromises the induction of major anterior neural genes representing poised enhancer targets. Overall, our work illuminates an unexpected function for polycomb proteins in facilitating neural induction by endowing major anterior neural loci with a permissive regulatory topology.


Asunto(s)
Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Ratones , Complejo Represivo Polycomb 2/genética , Reacción en Cadena de la Polimerasa
9.
Nucleic Acids Res ; 45(7): 3800-3811, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28100697

RESUMEN

A precise immune response is essential for cellular homeostasis and animal survival. The paramount importance of its control is reflected by the fact that its non-specific activation leads to inflammatory events that ultimately contribute to the appearance of many chronic diseases. However, the molecular mechanisms preventing non-specific activation and allowing a quick response upon signal activation are not yet fully understood. In this paper we uncover a new function of PHF8 blocking signal independent activation of immune gene promoters. Affinity purifications coupled with mass spectrometry analysis identified SIN3A and HDAC1 corepressors as new PHF8 interacting partners. Further molecular analysis demonstrated that prior to interferon gamma (IFNγ) stimulation, PHF8 is bound to a subset of IFNγ-responsive promoters. Through the association with HDAC1 and SIN3A, PHF8 keeps the promoters in a silent state, maintaining low levels of H4K20me1. Upon IFNγ treatment, PHF8 is phosphorylated by ERK2 and evicted from the promoters, correlating with an increase in H4K20me1 and transcriptional activation. Our data strongly indicate that in addition to its well-characterized function as a coactivator, PHF8 safeguards transcription to allow an accurate immune response.


Asunto(s)
Histona Demetilasas/metabolismo , Interferón gamma/farmacología , Factores de Transcripción/metabolismo , Activación Transcripcional , Línea Celular , Cromatina/metabolismo , Silenciador del Gen , Histona Desacetilasa 1/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3
10.
Open Biol ; 6(4): 150227, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27248655

RESUMEN

The function of EZH2 as a transcription repressor is well characterized. However, its role during vertebrate development is still poorly understood, particularly in neurogenesis. Here, we uncover the role of EZH2 in controlling the integrity of the neural tube and allowing proper progenitor proliferation. We demonstrate that knocking down the EZH2 in chick embryo neural tubes unexpectedly disrupts the neuroepithelium (NE) structure, correlating with alteration of the Rho pathway, and reduces neural progenitor proliferation. Moreover, we use transcriptional profiling and functional assays to show that EZH2-mediated repression of p21(WAF1/CIP1) contributes to both processes. Accordingly, overexpression of cytoplasmic p21(WAF1/CIP1) induces NE structural alterations and p21(WAF1/CIP1) suppression rescues proliferation defects and partially compensates for the structural alterations and the Rho activity. Overall, our findings describe a new role of EZH2 in controlling the NE integrity in the neural tube to allow proper progenitor proliferation.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Células Neuroepiteliales/citología , Células Neuroepiteliales/metabolismo , Proteínas Represoras/metabolismo , Animales , Polaridad Celular , Proliferación Celular , Embrión de Pollo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Tubo Neural/citología , Tubo Neural/metabolismo , Regiones Promotoras Genéticas/genética
11.
Cell Tissue Res ; 359(1): 87-98, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24950624

RESUMEN

Central nervous system (CNS) development is driven by coordinated actions of developmental signals and chromatin regulators that precisely regulate gene expression patterns. Histone methylation is a regulatory mechanism that controls transcriptional programs. In the last 10 years, several histone demethylases (HDM) have been identified as important players in neural development, and their implication in cell fate decisions is beginning to be recognized. Identification of the physiological roles of these enzymes and their molecular mechanisms of action will be necessary for completely understanding the process that ultimately generates different neural cells in the CNS. In this review, we provide an overview of the Jumonji family of HDMs involved in neurodevelopment, and we discuss their roles during neural fate establishment and neuronal differentiation.


Asunto(s)
Histona Demetilasas/metabolismo , Sistema Nervioso/crecimiento & desarrollo , Neurogénesis , Animales , Humanos , Modelos Biológicos , Familia de Multigenes , Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/enzimología , Enfermedades del Sistema Nervioso/patología
12.
Mol Biol Cell ; 24(3): 351-60, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23243002

RESUMEN

JMJD3 H3K27me3 demethylase plays an important role in the transcriptional response to different signaling pathways; however, the mechanism by which it facilitates transcription has been unclear. Here we show that JMJD3 regulates transcription of transforming growth factor ß (TGFß)-responsive genes by promoting RNA polymerase II (RNAPII) progression along the gene bodies. Using chromatin immunoprecipitation followed by sequencing experiments, we show that, upon TGFß treatment, JMJD3 and elongating RNAPII colocalize extensively along the intragenic regions of TGFß target genes. According to these data, genome-wide analysis shows that JMJD3-dependent TGFß target genes are enriched in H3K27me3 before TGFß signaling pathway activation. Further molecular analyses demonstrate that JMJD3 demethylates H3K27me3 along the gene bodies, paving the way for the RNAPII progression. Overall these findings uncover the mechanism by which JMJD3 facilitates transcriptional activation.


Asunto(s)
Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Procesamiento Proteico-Postraduccional , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Inmunoprecipitación de Cromatina , Quinasa 9 Dependiente de la Ciclina/metabolismo , Genoma , Células HEK293 , Humanos , Metilación , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Análisis de Secuencia de ADN , Proteína smad3/metabolismo , Sitio de Iniciación de la Transcripción , Factor de Crecimiento Transformador beta/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...