LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 35

Search options

  1. Article ; Online: Sex-biased and parental allele-specific gene regulation by KDM6A.

    Ma, Wenxiu / Fang, He / Pease, Nicolas / Filippova, Galina N / Disteche, Christine M / Berletch, Joel B

    Biology of sex differences

    2022  Volume 13, Issue 1, Page(s) 40

    Abstract: Background: KDM6A is a demethylase encoded by a gene with female-biased expression due to escape from X inactivation. Its main role is to facilitate gene expression through removal of the repressive H3K27me3 mark, with evidence of some additional ... ...

    Abstract Background: KDM6A is a demethylase encoded by a gene with female-biased expression due to escape from X inactivation. Its main role is to facilitate gene expression through removal of the repressive H3K27me3 mark, with evidence of some additional histone demethylase-independent functions. KDM6A mutations have been implicated in congenital disorders such as Kabuki Syndrome, as well as in sex differences in cancer.
    Methods: Kdm6a was knocked out using CRISPR/Cas9 gene editing in F1 male and female mouse embryonic stem cells (ES) derived from reciprocal crosses between C57BL6 x Mus castaneus. Diploid and allelic RNA-seq analyses were done to compare gene expression between wild-type and Kdm6a knockout (KO) clones. The effects of Kdm6a KO on sex-biased gene expression were investigated by comparing gene expression between male and female ES cells. Changes in H3K27me3 enrichment and chromatin accessibility at promoter regions of genes with expression changes were characterized by ChIP-seq and ATAC-seq followed by diploid and allelic analyses.
    Results: We report that Kdm6a KO in male and female embryonic stem (ES) cells derived from F1 hybrid mice cause extensive gene dysregulation, disruption of sex biases, and specific parental allele effects. Among the dysregulated genes are candidate genes that may explain abnormal developmental features of Kabuki syndrome caused by KDM6A mutations in human. Strikingly, Kdm6a knockouts result in a decrease in sex-biased expression and in preferential downregulation of the maternal alleles of a number of genes. Most promoters of dysregulated genes show concordant epigenetic changes including gain of H3K27me3 and loss of chromatin accessibility, but there was less concordance when considering allelic changes.
    Conclusions: Our study reveals new sex-related roles of KDM6A in the regulation of developmental genes, the maintenance of sex-biased gene expression, and the differential expression of parental alleles.
    MeSH term(s) Abnormalities, Multiple ; Alleles ; Animals ; Chromatin ; Face/abnormalities ; Female ; Hematologic Diseases ; Histone Demethylases/genetics ; Histone Demethylases/metabolism ; Histones/genetics ; Histones/metabolism ; Humans ; Male ; Mice ; Vestibular Diseases
    Chemical Substances Chromatin ; Histones ; Histone Demethylases (EC 1.14.11.-)
    Language English
    Publishing date 2022-07-23
    Publishing country England
    Document type Journal Article ; Research Support, U.S. Gov't, Non-P.H.S. ; Research Support, N.I.H., Extramural
    ZDB-ID 2587352-0
    ISSN 2042-6410 ; 2042-6410
    ISSN (online) 2042-6410
    ISSN 2042-6410
    DOI 10.1186/s13293-022-00452-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: X Inactivation and Escape: Epigenetic and Structural Features.

    Fang, He / Disteche, Christine M / Berletch, Joel B

    Frontiers in cell and developmental biology

    2019  Volume 7, Page(s) 219

    Abstract: X inactivation represents a complex multi-layer epigenetic mechanism that profoundly modifies chromatin composition and structure of one X chromosome in females. The heterochromatic inactive X chromosome adopts a unique 3D bipartite structure and a ... ...

    Abstract X inactivation represents a complex multi-layer epigenetic mechanism that profoundly modifies chromatin composition and structure of one X chromosome in females. The heterochromatic inactive X chromosome adopts a unique 3D bipartite structure and a location close to the nuclear periphery or the nucleolus. X-linked lncRNA loci and their transcripts play important roles in the recruitment of proteins that catalyze chromatin and DNA modifications for silencing, as well as in the control of chromatin condensation and location of the inactive X chromosome. A subset of genes escapes X inactivation, raising questions about mechanisms that preserve their expression despite being embedded within heterochromatin. Escape gene expression differs between males and females, which can lead to physiological sex differences. We review recent studies that emphasize challenges in understanding the role of lncRNAs in the control of epigenetic modifications, structural features and nuclear positioning of the inactive X chromosome. Second, we highlight new findings about the distribution of genes that escape X inactivation based on single cell studies, and discuss the roles of escape genes in eliciting sex differences in health and disease.
    Language English
    Publishing date 2019-10-01
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2737824-X
    ISSN 2296-634X
    ISSN 2296-634X
    DOI 10.3389/fcell.2019.00219
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: KDM6A facilitates Xist upregulation at the onset of X inactivation.

    Lin, Josephine / Zhang, Jinli / Ma, Li / Fang, He / Ma, Rui / Groneck, Camille / Filippova, Galina N / Deng, Xinxian / Ma, Wenxiu / Disteche, Christine M / Berletch, Joel B

    bioRxiv : the preprint server for biology

    2023  

    Abstract: X chromosome inactivation (XCI) is a female-specific process in which one X chromosome is silenced to balance X-linked gene expression between the sexes. XCI is initiated in early development by upregulation of the ... ...

    Abstract X chromosome inactivation (XCI) is a female-specific process in which one X chromosome is silenced to balance X-linked gene expression between the sexes. XCI is initiated in early development by upregulation of the lncRNA
    Language English
    Publishing date 2023-08-17
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.08.16.553617
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: X-chromosome inactivation and escape.

    Disteche, Christine M / Berletch, Joel B

    Journal of genetics

    2015  Volume 94, Issue 4, Page(s) 591–599

    Abstract: X-chromosome inactivation, which was discovered by Mary Lyon in 1961 results in random silencing of one X chromosome in female mammals. This review is dedicated to Mary Lyon, who passed away last year. She predicted many of the features of X inactivation, ...

    Abstract X-chromosome inactivation, which was discovered by Mary Lyon in 1961 results in random silencing of one X chromosome in female mammals. This review is dedicated to Mary Lyon, who passed away last year. She predicted many of the features of X inactivation, for e.g., the existence of an X inactivation center, the role of L1 elements in spreading of silencing and the existence of genes that escape X inactivation. Starting from her published work here we summarize advances in the field.
    MeSH term(s) Animals ; Gene Silencing/physiology ; Humans ; X Chromosome/genetics ; X Chromosome Inactivation/genetics
    Language English
    Publishing date 2015-12-04
    Publishing country India
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Review
    ZDB-ID 3039-9
    ISSN 0973-7731 ; 0958-8361 ; 0022-1333
    ISSN (online) 0973-7731
    ISSN 0958-8361 ; 0022-1333
    DOI 10.1007/s12041-015-0574-1
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: X Inactivation and Escape

    He Fang / Christine M. Disteche / Joel B. Berletch

    Frontiers in Cell and Developmental Biology, Vol

    Epigenetic and Structural Features

    2019  Volume 7

    Abstract: X inactivation represents a complex multi-layer epigenetic mechanism that profoundly modifies chromatin composition and structure of one X chromosome in females. The heterochromatic inactive X chromosome adopts a unique 3D bipartite structure and a ... ...

    Abstract X inactivation represents a complex multi-layer epigenetic mechanism that profoundly modifies chromatin composition and structure of one X chromosome in females. The heterochromatic inactive X chromosome adopts a unique 3D bipartite structure and a location close to the nuclear periphery or the nucleolus. X-linked lncRNA loci and their transcripts play important roles in the recruitment of proteins that catalyze chromatin and DNA modifications for silencing, as well as in the control of chromatin condensation and location of the inactive X chromosome. A subset of genes escapes X inactivation, raising questions about mechanisms that preserve their expression despite being embedded within heterochromatin. Escape gene expression differs between males and females, which can lead to physiological sex differences. We review recent studies that emphasize challenges in understanding the role of lncRNAs in the control of epigenetic modifications, structural features and nuclear positioning of the inactive X chromosome. Second, we highlight new findings about the distribution of genes that escape X inactivation based on single cell studies, and discuss the roles of escape genes in eliciting sex differences in health and disease.
    Keywords X chromosome ; X inactivation ; 3D-structure ; LncRNAs ; escape gene ; epigenetics ; Biology (General) ; QH301-705.5
    Language English
    Publishing date 2019-10-01T00:00:00Z
    Publisher Frontiers Media S.A.
    Document type Article ; Online
    Database BASE - Bielefeld Academic Search Engine (life sciences selection)

    More links

    Kategorien

  6. Article: CTCF-mediated insulation and chromatin environment modulate

    Fang, He / Tronco, Ana R / Bonora, Giancarlo / Nguyen, Truong / Thakur, Jitendra / Berletch, Joel B / Filippova, Galina N / Henikoff, Steven / Shendure, Jay / Noble, William S / Disteche, Christine M / Deng, Xinxian

    bioRxiv : the preprint server for biology

    2023  

    Abstract: Background: The number and escape levels of genes that escape X chromosome inactivation (XCI) in female somatic cells vary among tissues and cell types, potentially contributing to specific sex differences. Here we investigate the role of CTCF, a master ...

    Abstract Background: The number and escape levels of genes that escape X chromosome inactivation (XCI) in female somatic cells vary among tissues and cell types, potentially contributing to specific sex differences. Here we investigate the role of CTCF, a master chromatin conformation regulator, in regulating escape from XCI. CTCF binding profiles and epigenetic features were systematically examined at constitutive and facultative escape genes using mouse allelic systems to distinguish the inactive X (Xi) and active X (Xa) chromosomes.
    Results: We found that escape genes are located inside domains flanked by convergent arrays of CTCF binding sites, consistent with the formation of loops. In addition, strong and divergent CTCF binding sites often located at the boundaries between escape genes and adjacent neighbors subject to XCI would help insulate domains. Facultative escapees show clear differences in CTCF binding dependent on their XCI status in specific cell types/tissues. Concordantly, deletion but not inversion of a CTCF binding site at the boundary between the facultative escape gene
    Conclusion: Our findings indicate that escape from XCI is modulated both by looping and insulation of chromatin via convergent arrays of CTCF binding sites and by compaction and epigenetic features of the surrounding heterochromatin.
    Language English
    Publishing date 2023-05-04
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.05.04.539469
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: X-Chromosome Inactivation and Escape from X Inactivation in Mouse.

    Ma, Wenxiu / Bonora, Giancarlo / Berletch, Joel B / Deng, Xinxian / Noble, William S / Disteche, Christine M

    Methods in molecular biology (Clifton, N.J.)

    2018  Volume 1861, Page(s) 205–219

    Abstract: X chromosome inactivation silences one X chromosome in female mammals. However, this silencing is incomplete, and some genes escape X inactivation. We describe methods to determine the chromosome-wide X inactivation status of genes in tissues or cell ... ...

    Abstract X chromosome inactivation silences one X chromosome in female mammals. However, this silencing is incomplete, and some genes escape X inactivation. We describe methods to determine the chromosome-wide X inactivation status of genes in tissues or cell lines derived from mice using a combination of skewing of X inactivation and allele-specific analyses of gene expression based on RNA-seq.
    MeSH term(s) Alleles ; Animals ; Cell Line ; Embryo, Mammalian/metabolism ; Epigenomics/methods ; Female ; Gene Expression Profiling/methods ; Gene Expression Regulation, Developmental ; Mice ; Mice, Inbred C57BL ; Polymorphism, Single Nucleotide ; Sequence Analysis, RNA/methods ; X Chromosome Inactivation
    Language English
    Publishing date 2018-09-14
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ISSN 1940-6029
    ISSN (online) 1940-6029
    DOI 10.1007/978-1-4939-8766-5_15
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: X chromosome regulation: diverse patterns in development, tissues and disease.

    Deng, Xinxian / Berletch, Joel B / Nguyen, Di K / Disteche, Christine M

    Nature reviews. Genetics

    2014  Volume 15, Issue 6, Page(s) 367–378

    Abstract: Genes on the mammalian X chromosome are present in one copy in males and two copies in females. The complex mechanisms that regulate the X chromosome lead to evolutionary and physiological variability in gene expression between species, the sexes, ... ...

    Abstract Genes on the mammalian X chromosome are present in one copy in males and two copies in females. The complex mechanisms that regulate the X chromosome lead to evolutionary and physiological variability in gene expression between species, the sexes, individuals, developmental stages, tissues and cell types. In early development, delayed and incomplete X chromosome inactivation (XCI) in some species causes variability in gene expression. Additional diversity stems from escape from XCI and from mosaicism or XCI skewing in females. This causes sex-specific differences that manifest as differential gene expression and associated phenotypes. Furthermore, the complexity and diversity of X dosage regulation affect the severity of diseases caused by X-linked mutations.
    MeSH term(s) Animals ; Chromosome Disorders/genetics ; Chromosome Disorders/metabolism ; Chromosomes, Human, X/genetics ; Chromosomes, Human, X/metabolism ; Female ; Gene Expression Regulation ; Genetic Diseases, X-Linked/genetics ; Genetic Diseases, X-Linked/metabolism ; Humans ; Male ; Mosaicism ; Sex Characteristics ; X Chromosome Inactivation
    Language English
    Publishing date 2014-04-15
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Review
    ZDB-ID 2035157-4
    ISSN 1471-0064 ; 1471-0056
    ISSN (online) 1471-0064
    ISSN 1471-0056
    DOI 10.1038/nrg3687
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article: X-chromosome inactivation and escape

    DISTECHE, CHRISTINE M / BERLETCH, JOEL B

    Journal of genetics. 2015 Dec., v. 94, no. 4

    2015  

    Abstract: X-chromosome inactivation, which was discovered by Mary Lyon in 1961 results in random silencing of one X chromosome in female mammals. This review is dedicated to Mary Lyon, who passed away last year. She predicted many of the features of X inactivation, ...

    Abstract X-chromosome inactivation, which was discovered by Mary Lyon in 1961 results in random silencing of one X chromosome in female mammals. This review is dedicated to Mary Lyon, who passed away last year. She predicted many of the features of X inactivation, for e.g., the existence of an X inactivation center, the role of L1 elements in spreading of silencing and the existence of genes that escape X inactivation. Starting from her published work here we summarize advances in the field.
    Keywords X chromosome ; females ; genes ; mammals
    Language English
    Dates of publication 2015-12
    Size p. 591-599.
    Publishing place Springer India
    Document type Article
    ZDB-ID 3039-9
    ISSN 0973-7731 ; 0958-8361 ; 0022-1333
    ISSN (online) 0973-7731
    ISSN 0958-8361 ; 0022-1333
    DOI 10.1007/s12041-015-0574-1
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  10. Article ; Online: Female bias in Rhox6 and 9 regulation by the histone demethylase KDM6A.

    Berletch, Joel B / Deng, Xinxian / Nguyen, Di Kim / Disteche, Christine M

    PLoS genetics

    2013  Volume 9, Issue 5, Page(s) e1003489

    Abstract: The Rhox cluster on the mouse X chromosome contains reproduction-related homeobox genes expressed in a sexually dimorphic manner. We report that two members of the Rhox cluster, Rhox6 and 9, are regulated by de-methylation of histone H3 at lysine 27 by ... ...

    Abstract The Rhox cluster on the mouse X chromosome contains reproduction-related homeobox genes expressed in a sexually dimorphic manner. We report that two members of the Rhox cluster, Rhox6 and 9, are regulated by de-methylation of histone H3 at lysine 27 by KDM6A, a histone demethylase with female-biased expression. Consistent with other homeobox genes, Rhox6 and 9 are in bivalent domains prior to embryonic stem cell differentiation and thus poised for activation. In female mouse ES cells, KDM6A is specifically recruited to Rhox6 and 9 for gene activation, a process inhibited by Kdm6a knockdown in a dose-dependent manner. In contrast, KDM6A occupancy at Rhox6 and 9 is low in male ES cells and knockdown has no effect on expression. In mouse ovary where Rhox6 and 9 remain highly expressed, KDM6A occupancy strongly correlates with expression. Our study implicates Kdm6a, a gene that escapes X inactivation, in the regulation of genes important in reproduction, suggesting that KDM6A may play a role in the etiology of developmental and reproduction-related effects of X chromosome anomalies.
    MeSH term(s) Animals ; DNA Methylation ; Embryonic Stem Cells/cytology ; Embryonic Stem Cells/metabolism ; Female ; Gene Expression Regulation, Developmental ; Histone Demethylases/genetics ; Histone Demethylases/metabolism ; Homeodomain Proteins/genetics ; Homeodomain Proteins/metabolism ; Jumonji Domain-Containing Histone Demethylases/genetics ; Mice ; Reproduction/genetics ; Reproduction/physiology ; Sex Characteristics ; X Chromosome Inactivation/genetics
    Chemical Substances Homeodomain Proteins ; Rhox6 protein, mouse ; Histone Demethylases (EC 1.14.11.-) ; Jumonji Domain-Containing Histone Demethylases (EC 1.14.11.-) ; Utx protein, mouse (EC 1.14.11.-) ; Kdm6b protein, mouse (EC 1.5.-)
    Language English
    Publishing date 2013-05-02
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2186725-2
    ISSN 1553-7404 ; 1553-7390
    ISSN (online) 1553-7404
    ISSN 1553-7390
    DOI 10.1371/journal.pgen.1003489
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top