Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Development ; 149(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35831950

RESUMEN

Vsx2 is a transcription factor essential for retinal proliferation and bipolar cell differentiation, but the molecular mechanisms underlying its developmental roles are unclear. Here, we have profiled VSX2 genomic occupancy during mouse retinogenesis, revealing extensive retinal genetic programs associated with VSX2 during development. VSX2 binds and transactivates its enhancer in association with the transcription factor PAX6. Mice harboring deletions in the Vsx2 regulatory landscape exhibit specific abnormalities in retinal proliferation and in bipolar cell differentiation. In one of those deletions, a complete loss of bipolar cells is associated with a bias towards photoreceptor production. VSX2 occupies cis-regulatory elements nearby genes associated with photoreceptor differentiation and homeostasis in the adult mouse and human retina, including a conserved region nearby Prdm1, a factor implicated in the specification of rod photoreceptors and suppression of bipolar cell fate. VSX2 interacts with the transcription factor OTX2 and can act to suppress OTX2-dependent enhancer transactivation of the Prdm1 enhancer. Taken together, our analyses indicate that Vsx2 expression can be temporally and spatially uncoupled at the enhancer level, and they illuminate important mechanistic insights into how VSX2 is engaged with gene regulatory networks that are essential for retinal proliferation and cell fate acquisition.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Adulto , Animales , Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Factores de Transcripción/metabolismo
2.
Front Genet ; 12: 775205, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34764989

RESUMEN

Regulation of gene expression by chromatin structure has been under intensive investigation, establishing nuclear organization and genome architecture as a potent and effective means of regulating developmental processes. The substantial growth in our knowledge of the molecular mechanisms underlying retinogenesis has been powered by several genome-wide based tools that mapped chromatin organization at multiple cellular and biochemical levels. Studies profiling the retinal epigenome and transcriptome have allowed the systematic annotation of putative cis-regulatory elements associated with transcriptional programs that drive retinal neural differentiation, laying the groundwork to understand spatiotemporal retinal gene regulation at a mechanistic level. In this review, we outline recent advances in our understanding of the chromatin architecture in the mammalian retina during development and disease. We focus on the emerging roles of non-coding regulatory elements in controlling retinal cell-type specific transcriptional programs, and discuss potential implications in untangling the etiology of eye-related disorders.

3.
Epigenetics Chromatin ; 14(1): 11, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33563331

RESUMEN

In the developing vertebrate retina, retinal progenitor cells (RPCs) proliferate and give rise to terminally differentiated neurons with exquisite spatio-temporal precision. Lineage commitment, fate determination and terminal differentiation are controlled by intricate crosstalk between the genome and epigenome. Indeed, epigenetic regulation plays pivotal roles in numerous cell fate specification and differentiation events in the retina. Moreover, aberrant chromatin structure can contribute to developmental disorders and retinal pathologies. In this review, we highlight recent advances in our understanding of epigenetic regulation in the retina. We also provide insight into several aspects of epigenetic-related regulation that should be investigated in future studies of retinal development and disease. Importantly, focusing on these mechanisms could contribute to the development of novel treatment strategies targeting a variety of retinal disorders.


Asunto(s)
Epigénesis Genética , Células Madre , Diferenciación Celular , Neuronas , Retina
4.
Cell Rep ; 22(10): 2601-2614, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514090

RESUMEN

Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.


Asunto(s)
Reprogramación Celular , Metilación de ADN , Histonas/metabolismo , Organogénesis , Organoides/crecimiento & desarrollo , Procesamiento Proteico-Postraduccional , Retina/citología , Retina/metabolismo , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Núcleo Celular/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Regiones Promotoras Genéticas/genética
5.
Dev Biol ; 437(1): 27-40, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29518376

RESUMEN

Neural basic helix-loop helix (bHLH) transcription factors promote progenitor cell differentiation by activation of downstream target genes that coordinate neuronal differentiation. Here we characterize a neural bHLH target gene in Xenopus laevis, vexin (vxn; previously sbt1), that is homologous to human c8orf46 and is conserved across vertebrate species. C8orf46 has been implicated in cancer progression, but its function is unknown. Vxn is transiently expressed in differentiating progenitors in the developing central nervous system (CNS), and is required for neurogenesis in the neural plate and retina. Its function is conserved, since overexpression of either Xenopus or mouse vxn expands primary neurogenesis and promotes early retinal cell differentiation in cooperation with neural bHLH factors. Vxn protein is localized to the cell membrane and the nucleus, but functions in the nucleus to promote neural differentiation. Vxn inhibits cell proliferation, and works with the cyclin-dependent kinase inhibitor p27Xic1 (cdkn1b) to enhance neurogenesis and increase levels of the proneural protein Neurog2. We propose that vxn provides a key link between neural bHLH activity and execution of the neurogenic program.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neurogénesis/genética , Proteínas de Xenopus/genética , Animales , Western Blotting , Diferenciación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Proteínas del Tejido Nervioso/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Retina/embriología , Retina/metabolismo , Xenopus laevis
6.
Neuron ; 94(3): 550-568.e10, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28472656

RESUMEN

In the developing retina, multipotent neural progenitors undergo unidirectional differentiation in a precise spatiotemporal order. Here we profile the epigenetic and transcriptional changes that occur during retinogenesis in mice and humans. Although some progenitor genes and cell cycle genes were epigenetically silenced during retinogenesis, the most dramatic change was derepression of cell-type-specific differentiation programs. We identified developmental-stage-specific super-enhancers and showed that most epigenetic changes are conserved in humans and mice. To determine how the epigenome changes during tumorigenesis and reprogramming, we performed integrated epigenetic analysis of murine and human retinoblastomas and induced pluripotent stem cells (iPSCs) derived from murine rod photoreceptors. The retinoblastoma epigenome mapped to the developmental stage when retinal progenitors switch from neurogenic to terminal patterns of cell division. The epigenome of retinoblastomas was more similar to that of the normal retina than that of retina-derived iPSCs, and we identified retina-specific epigenetic memory.


Asunto(s)
Carcinogénesis/genética , Diferenciación Celular/genética , Reprogramación Celular/genética , Metilación de ADN/genética , Epigénesis Genética , Código de Histonas/genética , Retina/metabolismo , Retinoblastoma/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Retina/embriología , Células Fotorreceptoras Retinianas Bastones/citología , Proteína de Retinoblastoma/genética
7.
Development ; 142(23): 4092-106, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26628093

RESUMEN

Retinal development requires precise temporal and spatial coordination of cell cycle exit, cell fate specification, cell migration and differentiation. When this process is disrupted, retinoblastoma, a developmental tumor of the retina, can form. Epigenetic modulators are central to precisely coordinating developmental events, and many epigenetic processes have been implicated in cancer. Studying epigenetic mechanisms in development is challenging because they often regulate multiple cellular processes; therefore, elucidating the primary molecular mechanisms involved can be difficult. Here we explore the role of Brg1 (Smarca4) in retinal development and retinoblastoma in mice using molecular and cellular approaches. Brg1 was found to regulate retinal size by controlling cell cycle length, cell cycle exit and cell survival during development. Brg1 was not required for cell fate specification but was required for photoreceptor differentiation and cell adhesion/polarity programs that contribute to proper retinal lamination during development. The combination of defective cell differentiation and lamination led to retinal degeneration in Brg1-deficient retinae. Despite the hypocellularity, premature cell cycle exit, increased cell death and extended cell cycle length, retinal progenitor cells persisted in Brg1-deficient retinae, making them more susceptible to retinoblastoma. ChIP-Seq analysis suggests that Brg1 might regulate gene expression through multiple mechanisms.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Retina/metabolismo , Retinoblastoma/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Apoptosis , Tipificación del Cuerpo , Adhesión Celular , Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Ratones , Microftalmía/genética , Retina/patología , Factores de Tiempo , Transgenes
8.
Development ; 140(14): 2867-78, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23739135

RESUMEN

The histone methyltransferase complex PRC2 controls key steps in developmental transitions and cell fate choices; however, its roles in vertebrate eye development remain unknown. Here, we report that in Xenopus, PRC2 regulates the progression of retinal progenitors from proliferation to differentiation. We show that the PRC2 core components are enriched in retinal progenitors and downregulated in differentiated cells. Knockdown of the PRC2 core component Ezh2 leads to reduced retinal progenitor proliferation, in part due to upregulation of the Cdk inhibitor p15(Ink4b). In addition, although PRC2 knockdown does not alter eye patterning, retinal progenitor gene expression or expression of the neural competence factor Sox2, it does cause suppression of proneural bHLH gene expression, indicating that PRC2 is crucial for the initiation of neural differentiation in the retina. Consistent with this, knocking down or blocking PRC2 function constrains the generation of most retinal neural cell types and promotes a Müller glial cell fate decision. We also show that Wnt/ß-catenin signaling acting through the receptor Frizzled 5, but independent of Sox2, regulates expression of key PRC2 subunits in the developing retina. This is consistent with a role for this pathway in coordinating proliferation and the transition to neurogenesis in the Xenopus retina. Our data establish PRC2 as a regulator of proliferation and differentiation during eye development.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Retina/embriología , Vía de Señalización Wnt , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Diferenciación Celular , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Receptores Frizzled/metabolismo , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Metilación , Complejo Represivo Polycomb 2/genética , Proteínas Represoras/metabolismo , Retina/citología , Retina/metabolismo , Proteínas de Xenopus/genética
9.
Dev Biol ; 367(2): 91-9, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22565092

RESUMEN

During organogenesis, tissues expand in size and eventually acquire consistent ratios of cells with dazzling diversity in morphology and function. During this process progenitor cells exit the cell cycle and execute differentiation programs through extensive genetic reprogramming that involves the silencing of proliferation genes and the activation of differentiation genes in a step-wise temporal manner. Recent years have witnessed expansion in our understanding of the epigenetic mechanisms that contribute to cellular differentiation and maturation during organ development, as this is a crucial step toward advancing regenerative therapy research for many intractable disorders. Among such epigenetic programs, the developmental roles of the polycomb repressive complex 2 (PRC2), a chromatin remodeling complex that mediates silencing of gene expression, have been under intensive examination. This review summarizes recent findings of how PRC2 functions to regulate the transition from proliferation to differentiation during organogenesis and discusses some aspects of the remaining questions associated with its regulation and mechanisms of action.


Asunto(s)
Organogénesis/fisiología , Proteínas Represoras/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Desarrollo de Músculos , Neurogénesis , Organogénesis/genética , Proteínas del Grupo Polycomb , Procesamiento Proteico-Postraduccional , Proteínas Represoras/genética
10.
Dev Dyn ; 238(12): 3185-92, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19877271

RESUMEN

The Polycomb repressive complex 2 is a multimeric aggregate that mediates silencing of a broad range of genes, and is associated with important biological contexts such as stem cell maintenance and cancer progression. PRC2 mainly trimethylates lysine 27 of histone H3 and is composed of three essential core subunits: EZH2, EED, and SUZ12. The Xenopus orthologs of PRC2 subunits Ezh2 and Eed have been described but Suz12 remained unidentified. Here, we report the cloning of the Xenopus Suz12, and determine its spatiotemporal expression during development. Xsuz12 transcript is provided maternally and continues to be expressed throughout development, particularly in the anterior part of the developing central nervous system. Importantly, comparative analysis of the expression of the PRC2 subunits Xez, Xeed, and Xrbbp4 indicates that their expression largely coincides with Xsuz12 in the nervous system, suggesting that PRC2 may have unexplored functions in the development of the frog central nervous system.


Asunto(s)
Desarrollo Embrionario/genética , Proteínas Represoras/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Xenopus laevis/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Embrión no Mamífero , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Datos de Secuencia Molecular , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Filogenia , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Proteína 4 de Unión a Retinoblastoma/genética , Proteína 4 de Unión a Retinoblastoma/metabolismo , Homología de Secuencia de Aminoácido , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...