Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Curr Res Neurobiol ; 5: 100114, 2023.
Article in English | MEDLINE | ID: mdl-38020809

ABSTRACT

The chromodomain helicase DNA-binding protein 8 (CHD8) is a chromatin remodeler whose mutation is associated, with high penetrance, with autism. Individuals with CHD8 mutations share common symptoms such as autistic behaviour, cognitive impairment, schizophrenia comorbidity, and phenotypic features such as macrocephaly and facial defects. Chd8-deficient mouse models recapitulate most of the phenotypes seen in the brain and other organs of humans. It is known that CHD8 regulates - directly and indirectly - neuronal, autism spectrum disorder (ASDs)-associated genes and long non-coding RNAs (lncRNAs) genes, which, in turn, regulate fundamental aspects of neuronal differentiation and brain development and function. A major characteristic of CHD8 regulation of gene expression is its non-linear and dosage-sensitive nature. CHD8 mutations appear to affect males predominantly, although the reasons for this observed sex bias remain- unknown. We have recently reported that CHD8 directly regulates X chromosome inactivation (XCI) through the transcriptional control of the Xist long non-coding RNA (lncRNA), the master regulator of mammalian XCI. We identified a role for CHD8 in regulating accessibility at the Xist promoter through competitive binding with transcription factors (TFs) at Xist regulatory regions. We speculate here that CHD8 might also regulate accessibility at neuronal/ASD targets through a similar competitive binding mechanism during neurogenesis and brain development. However, whilst such a model can reconcile the phenotypic differences observed in Chd8 knock-down (KD) vs knock-out (KO) mouse models, explaining the observed CHD8 non-linear dosage-dependent activity, it cannot on its own explain the observed disease sex bias.

2.
Commun Biol ; 4(1): 485, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859315

ABSTRACT

Female mammals achieve dosage compensation by inactivating one of their two X chromosomes during development, a process entirely dependent on Xist, an X-linked long non-coding RNA (lncRNA). At the onset of X chromosome inactivation (XCI), Xist is up-regulated and spreads along the future inactive X chromosome. Contextually, it recruits repressive histone and DNA modifiers that transcriptionally silence the X chromosome. Xist regulation is tightly coupled to differentiation and its expression is under the control of both pluripotency and epigenetic factors. Recent evidence has suggested that chromatin remodelers accumulate at the X Inactivation Center (XIC) and here we demonstrate a new role for Chd8 in Xist regulation in differentiating ES cells, linked to its control and prevention of spurious transcription factor interactions occurring within Xist regulatory regions. Our findings have a broader relevance, in the context of complex, developmentally-regulated gene expression.


Subject(s)
DNA-Binding Proteins/genetics , X Chromosome Inactivation , X Chromosome/genetics , Animals , DNA-Binding Proteins/metabolism , Dosage Compensation, Genetic , Female , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
Mamm Genome ; 28(1-2): 1-12, 2017 02.
Article in English | MEDLINE | ID: mdl-27671790

ABSTRACT

Nonobese diabetic (NOD) mice are a model for type 1 diabetes that displays defects in central immune tolerance, including impairment of thymocyte apoptosis and proliferation. Thymocyte apoptosis is decreased in NOD/Lt mice compared to nondiabetic C3H/HeJ and C57BL/6 mice. Analysis of a set of NOD.C3H and NOD.B6 congenic mouse strains for distal chromosome 6 localizes the phenotype to the 700 kb Idd6.3 interval. Idd6.3 contains the type 1 diabetes candidate gene aryl hydrocarbon receptor nuclear translocator-like 2 (Arntl2), encoding a circadian rhythm-related transcription factor. Newly generated Arntl2 -/- mouse strains reveal that inactivation of the B6 allele of Arntl2 is sufficient to both decrease thymocyte apoptosis and proliferation. When expressed from C3H or B6 alleles, ARNTL2 inhibits the transcription of interleukin 21 (Il21), a major player in the regulation of immune responses. IL-21 injection abolishes the B6 allele-mediated decrease of apoptosis and proliferation. Interestingly, IL-21 also leads to an increase in thymic proinflammatory Th17 helper cells. Our results identify Arntl2 as a gene controlling thymocyte apoptosis and proliferation along with Th17 development through the IL-21 pathway.


Subject(s)
ARNTL Transcription Factors/genetics , Apoptosis/genetics , Circadian Rhythm/genetics , Diabetes Mellitus, Type 1/genetics , Animals , Cell Proliferation/genetics , Chromosomes/genetics , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Inbred NOD/genetics , Mice, Knockout , Th17 Cells/metabolism , Thymocytes/metabolism , Thymocytes/pathology
7.
Article in English | MEDLINE | ID: mdl-26628922

ABSTRACT

BACKGROUND: In female mice, while the presence of two-active X-chromosomes characterises pluripotency, it is not tolerated in most other cellular contexts. In particular, in the trophoblastic lineage, impairment of paternal X (X(P)) inactivation results in placental defects. RESULTS: Here, we show that Trophoblast Stem (TS) cells can undergo a complete reversal of imprinted X-inactivation without detectable change in cell-type identity. This reversal occurs through a reactivation of the X(P) leading to TS clones showing two active Xs. Intriguingly, within such clones, all the cells rapidly and homogeneously either re-inactivate the X(P) or inactivate, de novo, the X(M). CONCLUSION: This secondary non-random inactivation suggests that the two-active-X states in TS and in pluripotent contexts are epigenetically distinct. These observations also reveal a pronounced plasticity of the TS epigenome allowing TS cells to dramatically and accurately reprogram gene expression profiles. This plasticity may serve as a back-up system when X-linked mono-allelic gene expression is perturbed.

8.
Epigenetics ; 10(10): 931-42, 2015.
Article in English | MEDLINE | ID: mdl-26267271

ABSTRACT

During the first divisions of the female mouse embryo, the paternal X-chromosome is coated by Xist non-coding RNA and gradually silenced. This imprinted X-inactivation principally results from the apposition, during oocyte growth, of an imprint on the X-inactivation master control region: the X-inactivation center (Xic). This maternal imprint of yet unknown nature is thought to prevent Xist upregulation from the maternal X (X(M)) during early female development. In order to provide further insight into the X(M) imprinting mechanism, we applied single-cell approaches to oocytes and pre-implantation embryos at different stages of development to analyze the expression of candidate genes within the Xic. We show that, unlike the situation pertaining in most other cellular contexts, in early-growing oocytes, Xist and Tsix sense and antisense transcription occur simultaneously from the same chromosome. Additionally, during early development, Xist appears to be transiently transcribed from the X(M) in some blastomeres of late 2-cell embryos concomitant with the general activation of the genome indicating that X(M) imprinting does not completely suppress maternal Xist transcription during embryo cleavage stages. These unexpected transcriptional regulations of the Xist locus call for a re-evaluation of the early functioning of the maternal imprint on the X-chromosome and suggest that Xist/Tsix antagonist transcriptional activities may participate in imprinting the maternal locus as described at other loci subject to parental imprinting.


Subject(s)
Genomic Imprinting/genetics , Oogenesis/genetics , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics , Animals , Embryo, Mammalian , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Mice , Oocytes/growth & development , Oocytes/metabolism , RNA, Long Noncoding/biosynthesis , X Chromosome/genetics
9.
Genome Biol ; 16: 166, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26282267

ABSTRACT

In female mammals, one of the two X chromosomes in each cell is transcriptionally silenced in order to achieve dosage compensation between the genders in a process called X chromosome inactivation. The master regulator of this process is the long non-coding RNA Xist. During X-inactivation, Xist accumulates in cis on the future inactive X chromosome, triggering a cascade of events that provoke the stable silencing of the entire chromosome, with relatively few genes remaining active. How Xist spreads, what are its binding sites, how it recruits silencing factors and how it induces a specific topological and nuclear organization of the chromatin all remain largely unanswered questions. Recent studies have improved our understanding of Xist localization and the proteins with which it interacts, allowing a reappraisal of ideas about Xist function. We discuss recent advances in our knowledge of Xist-mediated silencing, focusing on Xist spreading, the nuclear organization of the inactive X chromosome, recruitment of the polycomb complex and the role of the nuclear matrix in the process of X chromosome inactivation.


Subject(s)
RNA, Long Noncoding/metabolism , X Chromosome Inactivation , Animals , Cell Nucleus/genetics , Female , Gene Silencing , Mice , Polycomb-Group Proteins/metabolism , RNA, Long Noncoding/analysis
11.
PLoS One ; 9(12): e116109, 2014.
Article in English | MEDLINE | ID: mdl-25546018

ABSTRACT

Random X-chromosome inactivation ensures dosage compensation in mammals through the transcriptional silencing of one of the two X chromosomes present in each female cell. Silencing is initiated in the differentiating epiblast of the mouse female embryos through coating of the nascent inactive X chromosome by the non-coding RNA Xist, which subsequently recruits the Polycomb Complex PRC2 leading to histone H3-K27 methylation. Here we examined in mouse ES cells the early steps of the transition from naive ES cells towards epiblast stem cells as a model for inducing X chromosome inactivation in vitro. We show that these conditions efficiently induce random XCI. Importantly, in a transient phase of this differentiation pathway, both X chromosomes are coated with Xist RNA in up to 15% of the XX cells. In an attempt to determine the dynamics of this process, we designed a strategy aimed at visualizing the nascent inactive X-chromosome in live cells. We generated transgenic female XX ES cells expressing the PRC2 component Ezh2 fused to the fluorescent protein Venus. The fluorescent fusion protein was expressed at sub-physiological levels and located in nuclei of ES cells. Upon differentiation of ES cell towards epiblast stem cell fate, Venus-fluorescent territories appearing in interphase nuclei were identified as nascent inactive X chromosomes by their association with Xist RNA. Imaging of Ezh2-Venus for up to 24 hours during the differentiation process showed survival of some cells with two fluorescent domains and a surprising dynamics of the fluorescent territories across cell division and in the course of the differentiation process. Our data reveal a strategy for visualizing the nascent inactive X chromosome and suggests the possibility for a large plasticity of the nascent inactive X chromosome.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Germ Layers/cytology , Imaging, Three-Dimensional , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Cell Nucleus/metabolism , Cell Survival , Enhancer of Zeste Homolog 2 Protein , Female , Interphase , Mice , Mice, Transgenic , Mitosis , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
12.
Article in English | MEDLINE | ID: mdl-25053977

ABSTRACT

BACKGROUND: Silencing of the paternal X chromosome (Xp), a phenomenon known as imprinted X-chromosome inactivation (I-XCI), characterises, amongst mouse extraembryonic lineages, the primitive endoderm and the extraembryonic endoderm (XEN) stem cells derived from it. RESULTS: Using a combination of chromatin immunoprecipitation characterisation of histone modifications and single-cell expression studies, we show that whilst the Xp in XEN cells, like the inactive X chromosome in other cell types, globally accumulates the repressive histone mark H3K27me3, a large number of Xp genes locally lack H3K27me3 and escape from I-XCI. In most cases this escape is specific to the XEN cell lineage. Importantly, the degree of escape and the genes concerned remain unchanged upon XEN conversion into visceral endoderm, suggesting stringent control of I-XCI in XEN derivatives. Surprisingly, chemical inhibition of EZH2, a member of the Polycomb repressive complex 2 (PRC2), and subsequent loss of H3K27me3 on the Xp, do not drastically perturb the pattern of silencing of Xp genes in XEN cells. CONCLUSIONS: The observations that we report here suggest that the maintenance of gene expression profiles of the inactive Xp in XEN cells involves a tissue-specific mechanism that acts partly independently of PRC2 catalytic activity.

13.
Stem Cells ; 32(2): 377-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24115267

ABSTRACT

Random epigenetic silencing of the X-chromosome in somatic tissues of female mammals equalizes the dosage of X-linked genes between the sexes. Unlike this form of X-inactivation that is essentially irreversible, the imprinted inactivation of the paternal X, which characterizes mouse extra-embryonic tissues, appears highly unstable in the trophoblast giant cells of the placenta. Here, we wished to determine whether such instability is already present in placental progenitor cells prior to differentiation toward lineage-specific cell types. To this end, we analyzed the behavior of a GFP transgene on the paternal X both in vivo and in trophoblast stem (TS) cells derived from the trophectoderm of XX(GFP) blastocysts. Using single-cell studies, we show that not only the GFP transgene but also a large number of endogenous genes on the paternal X are subject to orchestrated cycles of reactivation/de novo inactivation in placental progenitor cells. This reversal of silencing is associated with local losses of histone H3 lysine 27 trimethylation extending over several adjacent genes and with the topological relocation of the hypomethylated loci outside of the nuclear compartment of the inactive X. The "reactivated" state is maintained through several cell divisions. Our study suggests that this type of "metastable epigenetic" states may underlie the plasticity of TS cells and predispose specific genes to relaxed regulation in specific subtypes of placental cells.


Subject(s)
Dosage Compensation, Genetic , Genes, X-Linked , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Blastocyst/metabolism , Epigenesis, Genetic , Female , Humans , Mice , Placenta , Pregnancy , Stem Cells , Trophoblasts/metabolism
14.
Arch Biochem Biophys ; 534(1-2): 20-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23031499

ABSTRACT

Neuronal differentiation from neural stem cells into mature neurons is guided by the concerted action of specific transcription factors that stepwise exercise their role in the context of defined chromatin states. Amongst the classes of proteins that influence chromatin compaction and modification are nucleosome assembly proteins (NAPs). Mammals possess several nucleosome assembly protein 1 like proteins (NAP1L) that show either ubiquitous or neuron-restricted expression. The latter group is presumably involved in the process of neuronal differentiation. Mammalian NAP1Ls can potentially form both homo- and hetero-dimers and octamers, in theory allowing thousands of different combinations to be formed. Detailed studies have been performed on several of the NAP1Ls that point to a range of molecular roles, including transcriptional regulation, nuclear import, and control of cell division. This article aims at summarizing current knowledge of the mammalian NAP1L family and its interactions.


Subject(s)
Neurogenesis , Neurons/cytology , Nucleosome Assembly Protein 1/metabolism , Nucleosomes/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , Gene Expression Regulation , Humans , Mammals/genetics , Mammals/metabolism , Neurons/metabolism , Nucleosome Assembly Protein 1/genetics , Nucleosomes/genetics , Protein Binding , Protein Interaction Mapping/methods , Transcription, Genetic
15.
Annu Rev Cell Dev Biol ; 27: 611-29, 2011.
Article in English | MEDLINE | ID: mdl-21801017

ABSTRACT

X-chromosome inactivation, or the silencing of one X chromosome that occurs initially in the female somatic four-cell-stage embryo, is reversed during embryonic development first at the time of inner cell mass formation and again during formation of germ cell precursors. Such X-chromosome reactivation in the mouse implies the silencing of the Xist gene and the transcription of its antisense partner, Tsix, from both X chromosomes. In murine embryonic stem cells, both genes are under the transcriptional control of a series of critical pluripotency factors, namely, OCT3/4, NANOG, SOX2, KLF4, C-MYC and REX1. Although the inactive/active status of the two X chromosomes present in female human embryonic stem cells remains controversial, the reactivation of X-chromosome inactivation seems to be a signature for the naive pluripotent state.


Subject(s)
Pluripotent Stem Cells/physiology , X Chromosome Inactivation , X Chromosome/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Gene Silencing , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Factor 4 , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Signal Transduction/physiology , X Chromosome/genetics
16.
PLoS Genet ; 7(7): e1002212, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21811421

ABSTRACT

In humans, sexual dimorphism is associated with the presence of two X chromosomes in the female, whereas males possess only one X and a small and largely degenerate Y chromosome. How do men cope with having only a single X chromosome given that virtually all other chromosomal monosomies are lethal? Ironically, or even typically many might say, women and more generally female mammals contribute most to the job by shutting down one of their two X chromosomes at random. This phenomenon, called X-inactivation, was originally described some 50 years ago by Mary Lyon and has captivated an increasing number of scientists ever since. The fascination arose in part from the realisation that the inactive X corresponded to a dense heterochromatin mass called the "Barr body" whose number varied with the number of Xs within the nucleus and from the many intellectual questions that this raised: How does the cell count the X chromosomes in the nucleus and inactivate all Xs except one? What kind of molecular mechanisms are able to trigger such a profound, chromosome-wide metamorphosis? When is X-inactivation initiated? How is it transmitted to daughter cells and how is it reset during gametogenesis? This review retraces some of the crucial findings, which have led to our current understanding of a biological process that was initially considered as an exception completely distinct from conventional regulatory systems but is now viewed as a paradigm "par excellence" for epigenetic regulation.


Subject(s)
Genetics/history , RNA, Untranslated/genetics , Sex Chromatin/genetics , X Chromosome Inactivation , Animals , Genetic Diseases, X-Linked/genetics , Genetics/trends , History, 20th Century , History, 21st Century , Humans , RNA, Long Noncoding
18.
J Mol Biol ; 407(5): 647-60, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21333655

ABSTRACT

Mammals possess five nucleosome assembly protein 1-like (NAP1L) proteins, with three of them being expressed exclusively in the nervous system. The biological importance of the neuron-specific NAP1L2 protein is demonstrated by the neural tube defects occurring during the embryonic development of Nap1l2 mutant mice, which are associated with an overproliferation of neural stem cells and decreased neuronal differentiation. NAP1L2 controls the expression of its target genes, such as the cell cycle regulator Cdkn1c, at least in part via an effect on histone acetylation. Using a two-hybrid analysis, we have identified several proteins interacting with NAP1L2, including the ubiquitously expressed members of the nucleosome assembly protein family, NAP1L1 and NAP1L4. Structural studies further predict that all five NAP1-like proteins are able to interact directly via their highly conserved α-helices. These elements, in conjunction with the coexpression of all the NAP1-like proteins in neurons and the finding that deletion of Nap1l2 affects the cytoplasmic-nuclear distribution patterns of both NAP1L1 and NAP1L4 and their recruitment to target genes, suggest that combinatorial variation within the NAP family may ensure adaptation to the specific requirements for neuronal differentiation such as intercellular repartition, chromatin modification, transcriptional regulation, or the recruitment of specific transcription factors.


Subject(s)
DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Nucleosome Assembly Protein 1/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Mice , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleosome Assembly Protein 1/chemistry , Nucleosome Assembly Protein 1/genetics , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Two-Hybrid System Techniques
19.
J Mol Biol ; 409(1): 62-9, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21329697

ABSTRACT

X-chromosome inactivation (XCI) ensures dosage compensation in mammals. Random XCI is a process where a single X chromosome is silenced in each cell of the epiblast of mouse female embryos. Operating at the level of an entire chromosome, XCI is a major paradigm for epigenetic processes. Here we review the most recent discoveries concerning the role of long noncoding RNAs, pluripotency factors, and chromosome structure in random XCI.


Subject(s)
X Chromosome Inactivation , Animals , Chromosomes/metabolism , Chromosomes/ultrastructure , Mice , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
20.
Hum Mol Genet ; 20(4): 705-18, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21118898

ABSTRACT

X chromosome inactivation (XCI) is an essential epigenetic process which involves several non-coding RNAs (ncRNAs), including Xist, the master regulator of X-inactivation initiation. Xist is flanked in its 5' region by a large heterochromatic hotspot, which contains several transcription units including a gene of unknown function, Ftx (five prime to Xist). In this article, we describe the characterization and functional analysis of murine Ftx. We present evidence that Ftx produces a conserved functional long ncRNA, and additionally hosts microRNAs (miR) in its introns. Strikingly, Ftx partially escapes X-inactivation and is upregulated specifically in female ES cells at the onset of X-inactivation, an expression profile which closely follows that of Xist. We generated Ftx null ES cells to address the function of this gene. In these cells, only local changes in chromatin marks are detected within the hotspot, indicating that Ftx is not involved in the global maintenance of the heterochromatic structure of this region. The Ftx mutation, however, results in widespread alteration of transcript levels within the X-inactivation center (Xic) and particularly important decreases in Xist RNA levels, which were correlated with increased DNA methylation at the Xist CpG island. Altogether our results indicate that Ftx is a positive regulator of Xist and lead us to propose that Ftx is a novel ncRNA involved in XCI.


Subject(s)
Chromatin/chemistry , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Animals , Cattle , Cell Line , Chromatin/metabolism , DNA Methylation , Dosage Compensation, Genetic , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation , Humans , Male , Mice , Mutation , Promoter Regions, Genetic , RNA, Long Noncoding , Sequence Homology , Up-Regulation , X Chromosome Inactivation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...