Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 8: 103, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32195249

RESUMEN

Global heterochromatin reduction, which is one of the hallmarks of senescent cells, is associated with reduced transposable element repression and increased risk of chromatin instability. To ensure genomic integrity, the irreparable cells in a population exit permanently from the cell cycle, and this process is termed "senescence." However, senescence only blocks the expansion of unwanted cells, and the aberrant chromatin of senescent cells remains unstable. Serendipitously, we found that the transient ectopic expression of a repressive epigenetic modulator, DNA methyltransferase 3-like (DNMT3L) was sufficient to delay the premature senescence progression of late-passage mouse embryonic fibroblasts (MEFs) associated with a tightened global chromatin structure. DNMT3L induces more repressive H3K9 methylation on endogenous retroviruses and downregulates the derepressed transposons in aging MEFs. In addition, we found that a pulse of ectopic DNMT3L resulted in the reestablishment of H3K27me3 on polycomb repressive complex 2 (PRC2)-target genes that were derepressed in old MEFs. We demonstrated that ectopic DNMT3L interacted with PRC2 in MEFs. Our data also suggested that ectopic DNMT3L might guide PRC2 to redress deregulated chromatin regions in cells undergoing senescence. This study might lead to an epigenetic reinforcement strategy for overcoming aging-associated epimutation and senescence.

2.
BMC Genomics ; 19(1): 480, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29921224

RESUMEN

Following publication of the original article [1], the authors reported that one of the authors' names is spelled incorrectly.

3.
BMC Genomics ; 19(1): 425, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859049

RESUMEN

BACKGROUND: The PIWI/piRNA pathway is a conserved machinery important for germ cell development and fertility. This piRNA-guided molecular machinery is best known for repressing derepressed transposable elements (TE) during epigenomic reprogramming. The extent to which piRNAs are involved in modulating transcripts beyond TEs still need to be clarified, and it may be a stage-dependent event. We chose chicken germline as a study model because of the significantly lower TE complexity in the chicken genome compared to mammalian species. RESULTS: We generated high-confidence piRNA candidates in various stages across chicken germline development by 3'-end-methylation-enriched small RNA sequencing and in-house bioinformatics analysis. We observed a significant developmental stage-dependent loss of TE association and a shifting of the ping-pong cycle signatures. Moreover, the stage-dependent reciprocal abundance of LINE retrotransposons, CR1-C, and its associated piRNAs implicated the developmental stage-dependent role of piRNA machinery. The stage dependency of piRNA expression and its potential functions can be better addressed by analyzing the piRNA precursors/clusters. Interestingly, the new piRNA clusters identified from embryonic chicken testes revealed evolutionary conservation between chickens and mammals, which was previously thought to not exist. CONCLUSIONS: In this report, we provided an original chicken RNA resource and proposed an analytical methodology that can be used to investigate stage-dependent changes in piRNA compositions and their potential roles in TE regulation and beyond, and also revealed possible conserved functions of piRNAs in developing germ cells.


Asunto(s)
Pollos/genética , ARN Interferente Pequeño/genética , Espermatozoides/citología , Animales , Linaje de la Célula/genética , Elementos Transponibles de ADN/genética , Masculino , Espermatozoides/metabolismo
4.
Reproduction ; 150(4): 245-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26159833

RESUMEN

Nuclear transfer (NT) is a technique used to investigate the development and reprogramming potential of a single cell. DNA methyltransferase-3-like, which has been characterized as a repressive transcriptional regulator, is expressed in naturally fertilized egg and morula/blastocyst at pre-implantation stages. In this study, we demonstrate that the use of Dnmt3l-knockout (Dnmt3l-KO) donor cells in combination with Trichostatin A treatment improved the developmental efficiency and quality of the cloned embryos. Compared with the WT group, Dnmt3l-KO donor cell-derived cloned embryos exhibited increased cell numbers as well as restricted OCT4 expression in the inner cell mass (ICM) and silencing of transposable elements at the blastocyst stage. In addition, our results indicate that zygotic Dnmt3l is dispensable for cloned embryo development at pre-implantation stages. In Dnmt3l-KO mouse embryonic fibroblasts, we observed reduced nuclear localization of HDAC1, increased levels of the active histone mark H3K27ac and decreased accumulation of the repressive histone marks H3K27me3 and H3K9me3, suggesting that Dnmt3l-KO donor cells may offer a more permissive epigenetic state that is beneficial for NT reprogramming.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Células Híbridas , Técnicas de Transferencia Nuclear , Animales , Blastocisto , Reprogramación Celular , Clonación de Organismos , Elementos Transponibles de ADN , Epigénesis Genética , Femenino , Fibroblastos , Silenciador del Gen , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Embarazo , Inhibidores de la Síntesis de la Proteína/farmacología
5.
Reproduction ; 150(3): R77-91, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26116003

RESUMEN

Appropriate regulation of epigenome within cells is crucial for the determination of cell fate and contributes to the lifelong maintenance of tissue homeostasis. Epigenomic re-establishment during embryonic prospermatogonia development and fine-tune of the epigenetic landscape in postnatal spermatogonial stem cells (SSCs) are two key processes required for functional male germ cell formation. Repression of re-activated transposons and male germline-specific epigenome establishment occur in prospermatogonia, whereas modulations of the epigenetic landscape is important for SSC self-renewal and differentiation to maintain the stem cell pool and support long-term sperm production. Here, we describe the impact of epigenome-related regulators and small non-coding RNAs as well as the influence of epigenome modifications that result from extrinsic signaling for controlling the decision between self-renewal, differentiation and survival in mouse prospermatogonia and SSCs. This article provides a review of epigenome-related molecules involved in cell fate determination in male germ cells and discusses the intriguing questions that arise from these studies.


Asunto(s)
Células Madre Adultas/fisiología , Epigénesis Genética , Espermatogonias/fisiología , Animales , Linaje de la Célula , Autorrenovación de las Células , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Humanos , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Transducción de Señal , Espermatogénesis , Nicho de Células Madre
6.
J Virol ; 88(18): 10680-95, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24991018

RESUMEN

UNLABELLED: Mammalian genomes are replete with retrotransposable elements, including endogenous retroviruses. DNA methyltransferase 3-like (DNMT3L) is an epigenetic regulator expressed in prospermatogonia, growing oocytes, and embryonic stem (ES) cells. Here, we demonstrate that DNMT3L enhances the interaction of repressive epigenetic modifiers, including histone deacetylase 1 (HDAC1), SET domain, bifurcated 1 (SETDB1), DNA methyltransferase 3A (DNMT3A), and tripartite motif-containing protein 28 (TRIM28; also known as TIF1ß and KAP1) in ES cells and orchestrates retroviral silencing activity with TRIM28 through mechanisms including, but not limited to, de novo DNA methylation. Ectopic expression of DNMT3L in somatic cells causes methylation-independent retroviral silencing activity by recruitment of the TRIM28/HDAC1/SETDB1/DNMT3A/DNMT3L complex to newly integrated Moloney murine leukemia virus (Mo-MuLV) proviral DNA. Concurrent with this recruitment, we also observed the accumulation of histone H3 lysine 9 trimethylation (H3K9me3) and heterochromatin protein 1 gamma (HP1γ), as well as reduced H3K9 and H3K27 acetylation at Mo-MuLV proviral sequences. Ectopic expression of DNMT3L in late-passage mouse embryonic fibroblasts (MEFs) recruited cytoplasmically localized HDAC1 to the nucleus. The formation of this epigenetic modifying complex requires interaction of DNMT3L with DNMT3A as well as with histone H3. In fetal testes at embryonic day 17.5, endogenous DNMT3L also enhanced the binding among TRIM28, DNMT3A, SETDB1, and HDAC1. We propose that DNMT3L may be involved in initiating a cascade of repressive epigenetic modifications by assisting in the preparation of a chromatin context that further attracts DNMT3A-DNMT3L binding and installs longer-term DNA methylation marks at newly integrated retroviruses. IMPORTANCE: Almost half of the mammalian genome is composed of endogenous retroviruses and other retrotransposable elements that threaten genomic integrity. These elements are usually subject to epigenetic silencing. We discovered that two epigenetic regulators that lack enzymatic activity, DNA methyltransferase 3-like (DNMT3L) and tripartite motif-containing protein 28 (TRIM28), collaborate with each other to impose retroviral silencing. In addition to modulating de novo DNA methylation, we found that by interacting with TRIM28, DNMT3L can attract various enzymes to form a DNMT3L-induced repressive complex to remove active marks and add repressive marks to histone proteins. Collectively, these results reveal a novel and pivotal function of DNMT3L in shaping the chromatin modifications necessary for retroviral and retrotransposon silencing.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Silenciador del Gen , Leucemia Experimental/enzimología , Leucemia Experimental/genética , Virus de la Leucemia Murina de Moloney/fisiología , Proteínas Represoras/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/metabolismo , Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histonas/metabolismo , Humanos , Leucemia Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Leucemia Murina de Moloney/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito
7.
Development ; 141(12): 2402-13, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24850856

RESUMEN

The ability of adult stem cells to reside in a quiescent state is crucial for preventing premature exhaustion of the stem cell pool. However, the intrinsic epigenetic factors that regulate spermatogonial stem cell quiescence are largely unknown. Here, we investigate in mice how DNA methyltransferase 3-like (DNMT3L), an epigenetic regulator important for interpreting chromatin context and facilitating de novo DNA methylation, sustains the long-term male germ cell pool. We demonstrated that stem cell-enriched THY1(+) spermatogonial stem/progenitor cells (SPCs) constituted a DNMT3L-expressing population in postnatal testes. DNMT3L influenced the stability of promyelocytic leukemia zinc finger (PLZF), potentially by downregulating Cdk2/CDK2 expression, which sequestered CDK2-mediated PLZF degradation. Reduced PLZF in Dnmt3l KO THY1(+) cells released its antagonist, Sal-like protein 4A (SALL4A), which is associated with overactivated ERK and AKT signaling cascades. Furthermore, DNMT3L was required to suppress the cell proliferation-promoting factor SALL4B in THY1(+) SPCs and to prevent premature stem cell exhaustion. Our results indicate that DNMT3L is required to delicately balance the cycling and quiescence of SPCs. These findings reveal a novel role for DNMT3L in modulating postnatal SPC cell fate decisions.


Asunto(s)
Células Madre Adultas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/fisiología , Regulación del Desarrollo de la Expresión Génica , Espermatogonias/metabolismo , Alelos , Animales , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Heterocigoto , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Testículo/metabolismo , Factores de Transcripción/metabolismo , Dedos de Zinc
8.
Biol Cell ; 104(10): 571-87, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22671959

RESUMEN

DNA methyltransferase 3-like (DNMT3L) is one of the key players in de novo DNA methylation of imprinting control elements and retrotransposons, which occurs after genome-wide epigenetic erasure during germ cell development. In this review, we summarise the biochemical properties of DNMT3L and discuss the possible mechanisms behind DNMT3L-mediated imprinting establishment and retrotransposon silencing in germ cells. We also discuss possible connections between DNMT3L and non-coding RNA-mediated epigenetic remodelling, the roles of DNMT3L in germ cell development and the implications in stem cell and cancer research.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Epigénesis Genética , Genoma , Células Germinativas/enzimología , Retroelementos/genética , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Impresión Genómica , Células Germinativas/citología , Células Germinativas/crecimiento & desarrollo , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Neoplasias/enzimología , Neoplasias/patología , Células Madre/citología , Células Madre/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...