Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 12: 1357576, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38476259

RESUMO

Transposable elements (TEs) are mobile genetic elements that constitute on average 45% of mammalian genomes. Their presence and activity in genomes represent a major source of genetic variability. While this is an important driver of genome evolution, TEs can also have deleterious effects on their hosts. A growing number of studies have focused on the role of TEs in the brain, both in physiological and pathological contexts. In the brain, their activity is believed to be important for neuronal plasticity. In neurological and age-related disorders, aberrant activity of TEs may contribute to disease etiology, although this remains unclear. After providing a comprehensive overview of transposable elements and their interactions with the host, this review summarizes the current understanding of TE activity within the brain, during the aging process, and in the context of neurological and age-related conditions.

2.
Nat Commun ; 13(1): 5432, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114205

RESUMO

Reprogramming of somatic cells into induced Pluripotent Stem Cells (iPSCs) is a major leap towards personalised approaches to disease modelling and cell-replacement therapies. However, we still lack the ability to fully control the epigenetic status of iPSCs, which is a major hurdle for their downstream applications. Epigenetic fidelity can be tracked by genomic imprinting, a phenomenon dependent on DNA methylation, which is frequently perturbed in iPSCs by yet unknown reasons. To try to understand the causes underlying these defects, we conducted a thorough imprinting analysis using IMPLICON, a high-throughput method measuring DNA methylation levels, in multiple female and male murine iPSC lines generated under different experimental conditions. Our results show that imprinting defects are remarkably common in iPSCs, but their nature depends on the sex of donor cells and their response to culture conditions. Imprints in female iPSCs resist the initial genome-wide DNA demethylation wave during reprogramming, but ultimately cells accumulate hypomethylation defects irrespective of culture medium formulations. In contrast, imprinting defects on male iPSCs depends on the experimental conditions and arise during reprogramming, being mitigated by the addition of vitamin C (VitC). Our findings are fundamental to further optimise reprogramming strategies and generate iPSCs with a stable epigenome.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Ácido Ascórbico/metabolismo , Metilação de DNA , Feminino , Genoma , Impressão Genômica , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos
3.
Biochem Soc Trans ; 49(6): 2549-2560, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34882219

RESUMO

X-inactive-specific transcript (Xist) is a long non-coding RNA (lncRNA) essential for X-chromosome inactivation (XCI) in female placental mammals. Thirty years after its discovery, it is still puzzling how this lncRNA triggers major structural and transcriptional changes leading to the stable silencing of an entire chromosome. Recently, a series of studies in mouse cells have uncovered domains of functional specialization within Xist mapping to conserved tandem repeat regions, known as Repeats A-to-F. These functional domains interact with various RNA binding proteins (RBPs) and fold into distinct RNA structures to execute specific tasks in a synergistic and coordinated manner during the inactivation process. This modular organization of Xist is mostly conserved in humans, but recent data point towards differences regarding functional specialization of the tandem repeats between the two species. In this review, we summarize the recent progress on understanding the role of Xist repetitive blocks and their involvement in the molecular mechanisms underlying XCI. We also discuss these findings in the light of the similarities and differences between mouse and human Xist.


Assuntos
RNA Longo não Codificante/genética , Sequências de Repetição em Tandem , Animais , Inativação Gênica , Humanos , Camundongos , Proteínas do Grupo Polycomb/metabolismo , Transcrição Gênica , Inativação do Cromossomo X
4.
Front Cell Dev Biol ; 9: 740937, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631717

RESUMO

X-chromosome inactivation (XCI) and random monoallelic expression of autosomal genes (RMAE) are two paradigms of gene expression regulation where, at the single cell level, genes can be expressed from either the maternal or paternal alleles. X-chromosome inactivation takes place in female marsupial and placental mammals, while RMAE has been described in mammals and also other species. Although the outcome of both processes results in random monoallelic expression and mosaicism at the cellular level, there are many important differences. We provide here a brief sketch of the history behind the discovery of XCI and RMAE. Moreover, we review some of the distinctive features of these two phenomena, with respect to when in development they are established, their roles in dosage compensation and cellular phenotypic diversity, and the molecular mechanisms underlying their initiation and stability.

5.
Nat Commun ; 12(1): 5330, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504093

RESUMO

Most autosomal genes are thought to be expressed from both alleles, with some notable exceptions, including imprinted genes and genes showing random monoallelic expression (RME). The extent and nature of RME has been the subject of debate. Here we investigate the expression of several candidate RME genes in F1 hybrid mouse cells before and after differentiation, to define how they become persistently, monoallelically expressed. Clonal monoallelic expression is not present in embryonic stem cells, but we observe high frequencies of monoallelism in neuronal progenitor cells by assessing expression status in more than 200 clones. We uncover unforeseen modes of allelic expression that appear to be gene-specific and epigenetically regulated. This non-canonical allelic regulation has important implications for development and disease, including autosomal dominant disorders and opens up therapeutic perspectives.


Assuntos
Alelos , Desequilíbrio Alélico , Epigênese Genética , Doenças Musculares/genética , Células-Tronco Neurais/metabolismo , Doenças Neurodegenerativas/genética , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Diferenciação Celular , Quimera , Células Clonais , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Feminino , Dosagem de Genes , Frequência do Gene , Loci Gênicos , Impressão Genômica , Masculino , Camundongos , Doenças Musculares/metabolismo , Doenças Musculares/patologia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neurais/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Receptor de GluK2 Cainato
6.
Essays Biochem ; 63(6): 663-676, 2019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31782494

RESUMO

Monoallelic gene expression occurs in diploid cells when only one of the two alleles of a gene is active. There are three main classes of genes that display monoallelic expression in mammalian genomes: (1) imprinted genes that are monoallelically expressed in a parent-of-origin dependent manner; (2) X-linked genes that undergo random X-chromosome inactivation in female cells; (3) random monoallelically expressed single and clustered genes located on autosomes. The heritability of monoallelic expression patterns during cell divisions implies that epigenetic mechanisms are involved in the cellular memory of these expression states. Among these, methylation of CpG sites on DNA is one of the best described modification to explain somatic inheritance. Here, we discuss the relevance of DNA methylation for the establishment and maintenance of monoallelic expression patterns among these three groups of genes, and how this is intrinsically linked to development and cellular states.


Assuntos
Alelos , Metilação de DNA , DNA/metabolismo , Epigênese Genética/fisiologia , Expressão Gênica/fisiologia , Animais , DNA/genética , Metilação de DNA/fisiologia , Feminino , Impressão Genômica/fisiologia , Humanos , Inativação do Cromossomo X/fisiologia
8.
Nat Genet ; 49(3): 377-386, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28112738

RESUMO

We developed an allele-specific assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) to genotype and profile active regulatory DNA across the genome. Using a mouse hybrid F1 system, we found that monoallelic DNA accessibility across autosomes was pervasive, developmentally programmed and composed of several patterns. Genetically determined accessibility was enriched at distal enhancers, but random monoallelically accessible (RAMA) elements were enriched at promoters and may act as gatekeepers of monoallelic mRNA expression. Allelic choice at RAMA elements was stable across cell generations and bookmarked through mitosis. RAMA elements in neural progenitor cells were biallelically accessible in embryonic stem cells but premarked with bivalent histone modifications; one allele was silenced during differentiation. Quantitative analysis indicated that allelic choice at the majority of RAMA elements is consistent with a stochastic process; however, up to 30% of RAMA elements may deviate from the expected pattern, suggesting a regulated or counting mechanism.


Assuntos
DNA/genética , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco/fisiologia , Alelos , Animais , Diferenciação Celular/genética , Linhagem Celular , Cromatina/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Masculino , Camundongos , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética
9.
Semin Cell Dev Biol ; 56: 100-110, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27101886

RESUMO

Genes are generally expressed from their two alleles, except in some particular cases such as random inactivation of one of the two X chromosomes in female mammals or imprinted genes which are expressed only from the maternal or the paternal allele. A lesser-known phenomenon is random monoallelic expression (RME) of autosomal genes, where genes can be stably expressed in a monoallelic manner, from either one of the parental alleles. Studies on autosomal RME face several challenges. First, RME that is based on epigenetic mechanisms has to be distinguished from biased expression of one allele caused by a DNA sequence polymorphism in a regulatory element. Second, RME should not be confused with transient monoallelic expression often observed in single cell analyses, and that often corresponds to dynamic bursting of expression. Thanks to analyses on clonal cell populations, the existence of RME in cultured cells is now well established. Future studies of RME in vivo will have to overcome tissue heterogeneity and certain technical limitations. Here, we discuss current knowledge on autosomal RME, as well as possible mechanisms controlling these expression patterns and potential implications for development and disease, drawing parallels with what is known for X-chromosome inactivation, a paradigm of random monoallelic expression.


Assuntos
Alelos , Cromossomos/genética , Inativação do Cromossomo X/genética , Animais , Doença/genética , Humanos , Análise de Célula Única
10.
Mol Cell Biol ; 35(23): 4053-68, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26391951

RESUMO

The chromosomal protein SMCHD1 plays an important role in epigenetic silencing at diverse loci, including the inactive X chromosome, imprinted genes, and the facioscapulohumeral muscular dystrophy locus. Although homology with canonical SMC family proteins suggests a role in chromosome organization, the mechanisms underlying SMCHD1 function and target site selection remain poorly understood. Here we show that SMCHD1 forms an active GHKL-ATPase homodimer, contrasting with canonical SMC complexes, which exist as tripartite ring structures. Electron microscopy analysis demonstrates that SMCHD1 homodimers structurally resemble prokaryotic condensins. We further show that the principal mechanism for chromatin loading of SMCHD1 involves an LRIF1-mediated interaction with HP1γ at trimethylated histone H3 lysine 9 (H3K9me3)-modified chromatin sites on the chromosome arms. A parallel pathway accounts for chromatin loading at a minority of sites, notably the inactive X chromosome. Together, our results provide key insights into SMCHD1 function and target site selection.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Histonas/metabolismo , Cromossomo X/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Cromatina/química , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/química , Células HEK293 , Histonas/química , Humanos , Lisina/análise , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mapas de Interação de Proteínas , Multimerização Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência
11.
Annu Rev Cell Dev Biol ; 30: 561-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000994

RESUMO

In mammals, the process of X-chromosome inactivation ensures equivalent levels of X-linked gene expression between males and females through the silencing of one of the two X chromosomes in female cells. The process is established early in development and is initiated by a unique locus, which produces a long noncoding RNA, Xist. The Xist transcript triggers gene silencing in cis by coating the future inactive X chromosome. It also induces a cascade of chromatin changes, including posttranslational histone modifications and DNA methylation, and leads to the stable repression of all X-linked genes throughout development and adult life. We review here recent progress in our understanding of the molecular mechanisms involved in the initiation of Xist expression, the propagation of the Xist RNA along the chromosome, and the cis-elements and trans-acting factors involved in the maintenance of the repressed state. We also describe the diverse strategies used by nonplacental mammals for X-chromosome dosage compensation and highlight the common features and differences between eutherians and metatherians, in particular regarding the involvement of long noncoding RNAs.


Assuntos
Inativação Gênica , RNA Longo não Codificante/genética , Inativação do Cromossomo X/genética , Animais , Cromatina/genética , Cromatina/ultraestrutura , Mapeamento Cromossômico , Cromossomos Humanos X/genética , Células-Tronco Embrionárias/ultraestrutura , Evolução Molecular , Feminino , Impressão Genômica , Humanos , Elementos Nucleotídeos Longos e Dispersos , Masculino , Marsupiais/genética , Camundongos , Processos de Determinação Sexual , Fatores de Transcrição/genética , Cromossomo X/genética , Cromossomo X/ultraestrutura
12.
Elife ; 3: e02024, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24842875

RESUMO

Histone acetyl transferases (HATs) play distinct roles in many cellular processes and are frequently misregulated in cancers. Here, we study the regulatory potential of MYST1-(MOF)-containing MSL and NSL complexes in mouse embryonic stem cells (ESCs) and neuronal progenitors. We find that both complexes influence transcription by targeting promoters and TSS-distal enhancers. In contrast to flies, the MSL complex is not exclusively enriched on the X chromosome, yet it is crucial for mammalian X chromosome regulation as it specifically regulates Tsix, the major repressor of Xist lncRNA. MSL depletion leads to decreased Tsix expression, reduced REX1 recruitment, and consequently, enhanced accumulation of Xist and variable numbers of inactivated X chromosomes during early differentiation. The NSL complex provides additional, Tsix-independent repression of Xist by maintaining pluripotency. MSL and NSL complexes therefore act synergistically by using distinct pathways to ensure a fail-safe mechanism for the repression of X inactivation in ESCs.DOI: http://dx.doi.org/10.7554/eLife.02024.001.


Assuntos
Células-Tronco Embrionárias/citologia , Histona Acetiltransferases/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Diferenciação Celular , Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Camundongos , Ligação Proteica , RNA Longo não Codificante/genética , Inativação do Cromossomo X
13.
Dev Cell ; 28(4): 366-80, 2014 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-24576422

RESUMO

X chromosome inactivation (XCI) and allelic exclusion of olfactory receptors or immunoglobulin loci represent classic examples of random monoallelic expression (RME). RME of some single copy genes has also been reported, but the in vivo relevance of this remains unclear. Here we identify several hundred RME genes in clonal neural progenitor cell lines derived from embryonic stem cells. RME occurs during differentiation, and, once established, the monoallelic state can be highly stable. We show that monoallelic expression also occurs in vivo, in the absence of DNA sequence polymorphism. Several of the RME genes identified play important roles in development and have been implicated in human autosomal-dominant disorders. We propose that monoallelic expression of such genes contributes to the fine-tuning of the developmental regulatory pathways they control, and, in the context of a mutation, RME can predispose to loss of function in a proportion of cells and thus contribute to disease.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Expressão Gênica , Alelos , Animais , Sequência de Bases/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Metilação de DNA/fisiologia , Genótipo , Humanos , Inativação do Cromossomo X
14.
Mol Cell Biol ; 33(16): 3150-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23754746

RESUMO

The Smchd1 gene encodes a large protein with homology to the SMC family of proteins involved in chromosome condensation and cohesion. Previous studies have found that Smchd1 has an important role in CpG island (CGI) methylation on the inactive X chromosome (Xi) and in stable silencing of some Xi genes. In this study, using genome-wide expression analysis, we showed that Smchd1 is required for the silencing of around 10% of the genes on Xi, apparently independent of CGI hypomethylation, and, moreover, that these genes nonrandomly occur in clusters. Additionally, we found that Smchd1 is required for CpG island methylation and silencing at a cluster of four imprinted genes in the Prader-Willi syndrome (PWS) locus on chromosome 7 and genes from the protocadherin-alpha and -beta clusters. All of the affected autosomal loci display developmentally regulated brain-specific methylation patterns which are lost in Smchd1 homozygous mutants. We discuss the implications of these findings for understanding the function of Smchd1 in epigenetic regulation of gene expression.


Assuntos
Proteínas Cromossômicas não Histona/genética , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Família Multigênica , Cromossomo X/genética , Animais , Caderinas/genética , Proteínas Cromossômicas não Histona/metabolismo , Ilhas de CpG , Metilação de DNA , Embrião de Mamíferos/metabolismo , Feminino , Deleção de Genes , Impressão Genômica , Masculino , Camundongos , Síndrome de Prader-Willi/genética , Receptores de Superfície Celular/genética
15.
Development ; 140(4): 861-72, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23362347

RESUMO

Mammalian X-chromosome inactivation (XCI) enables dosage compensation between XX females and XY males. It is an essential process and its absence in XX individuals results in early lethality due primarily to extra-embryonic defects. This sensitivity to X-linked gene dosage in extra-embryonic tissues is difficult to reconcile with the reported tendency of escape from XCI in these tissues. The precise transcriptional status of the inactive X chromosome in different lineages has mainly been examined using transgenes or in in vitro differentiated stem cells and the degree to which endogenous X-linked genes are silenced in embryonic and extra-embryonic lineages during early postimplantation stages is unclear. Here we investigate the precise temporal and lineage-specific X-inactivation status of several genes in postimplantation mouse embryos. We find stable gene silencing in most lineages, with significant levels of escape from XCI mainly in one extra-embryonic cell type: trophoblast giant cells (TGCs). To investigate the basis of this epigenetic instability, we examined the chromatin structure and organization of the inactive X chromosome in TGCs obtained from ectoplacental cone explants. We find that the Xist RNA-coated X chromosome has a highly unusual chromatin content in TGCs, presenting both heterochromatic marks such as H3K27me3 and euchromatic marks such as histone H4 acetylation and H3K4 methylation. Strikingly, Xist RNA does not form an overt silent nuclear compartment or Cot1 hole in these cells. This unusual combination of silent and active features is likely to reflect, and might underlie, the partial activity of the X chromosome in TGCs.


Assuntos
Cromatina/genética , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário/fisiologia , Células Gigantes/metabolismo , Trofoblastos/citologia , Inativação do Cromossomo X/fisiologia , Cromossomo X/genética , Acetilação , Animais , Cromatina/metabolismo , Metilação de DNA , Feminino , Imunofluorescência , Inativação Gênica/fisiologia , Hibridização in Situ Fluorescente , Masculino , Camundongos , Cromossomo X/metabolismo
16.
Dev Cell ; 23(2): 265-79, 2012 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-22841499

RESUMO

X chromosome inactivation involves multiple levels of chromatin modification, established progressively and in a stepwise manner during early development. The chromosomal protein Smchd1 was recently shown to play an important role in DNA methylation of CpG islands (CGIs), a late step in the X inactivation pathway that is required for long-term maintenance of gene silencing. Here we show that inactive X chromosome (Xi) CGI methylation can occur via either Smchd1-dependent or -independent pathways. Smchd1-dependent CGI methylation, the primary pathway, is acquired gradually over an extended period, whereas Smchd1-independent CGI methylation occurs rapidly after the onset of X inactivation. The de novo methyltransferase Dnmt3b is required for methylation of both classes of CGI, whereas Dnmt3a and Dnmt3L are dispensable. Xi CGIs methylated by these distinct pathways differ with respect to their sequence characteristics and immediate chromosomal environment. We discuss the implications of these results for understanding CGI methylation during development.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Ilhas de CpG , Metilação de DNA , Inativação do Cromossomo X , Alelos , Animais , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Camundongos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
17.
Development ; 138(23): 5049-55, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22069183

RESUMO

The third X-inactivation meeting 'Fifty years of X-inactivation research', which celebrated the fiftieth anniversary of Mary Lyon's formulation of the X-inactivation hypothesis, was an EMBO workshop held in Oxford, UK, in July 2011. This conference brought together the usual suspects from the field, as well as younger researchers, to discuss recent advances in X-inactivation research. Here, we review the results presented at the meeting and highlight some of the exciting progress that has been made. We also discuss the future challenges for the field, which aim to further our understanding of the developmental regulation of X inactivation, the randomness (or skewing) of X inactivation, and the diverse strategies used by mammalian species to mediate X inactivation.


Assuntos
Genes Ligados ao Cromossomo X/genética , RNA não Traduzido/metabolismo , Pesquisa/história , Pesquisa/tendências , Inativação do Cromossomo X/fisiologia , Animais , História do Século XX , História do Século XXI , Humanos , Camundongos , RNA Longo não Codificante , Especificidade da Espécie
18.
Nat Genet ; 40(5): 663-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18425126

RESUMO

X-chromosome inactivation is the mammalian dosage compensation mechanism by which transcription of X-linked genes is equalized between females and males. In an N-ethyl-N-nitrosourea (ENU) mutagenesis screen on mice for modifiers of epigenetic reprogramming, we identified the MommeD1 (modifier of murine metastable epialleles) mutation as a semidominant suppressor of variegation. MommeD1 shows homozygous female-specific mid-gestation lethality and hypomethylation of the X-linked gene Hprt1, suggestive of a defect in X inactivation. Here we report that the causative point mutation lies in a previously uncharacterized gene, Smchd1 (structural maintenance of chromosomes hinge domain containing 1). We find that SmcHD1 is not required for correct Xist expression, but localizes to the inactive X and has a role in the maintenance of X inactivation and the hypermethylation of CpG islands associated with the inactive X. This finding links a group of proteins normally associated with structural aspects of chromosome biology with epigenetic gene silencing.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Inativação Gênica , Inativação do Cromossomo X , Cromossomo X/metabolismo , Animais , Proteínas Cromossômicas não Histona/análise , Proteínas Cromossômicas não Histona/genética , Ilhas de CpG , Metilação de DNA , Fibroblastos/ultraestrutura , Camundongos , Mutação Puntual , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Cromossomo X/química , Cromossomo X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...