Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Commun ; 13(1): 4464, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35915095

ABSTRACT

X chromosome inactivation (XCI) is a dosage compensation phenomenon that occurs in females. Initiation of XCI depends on Xist RNA, which triggers silencing of one of the two X chromosomes, except for XCI escape genes that continue to be biallelically expressed. In the soma XCI is stably maintained with continuous Xist expression. How Xist impacts XCI maintenance remains an open question. Here we conditionally delete Xist in hematopoietic system of mice and report differentiation and cell cycle defects in female hematopoietic stem and progenitor cells (HSPCs). By utilizing female HSPCs and mouse embryonic fibroblasts, we find that X-linked genes show variable tolerance to Xist loss. Specifically, XCI escape genes exhibit preferential transcriptional upregulation, which associates with low H3K27me3 occupancy and high chromatin accessibility that accommodates preexisting binding of transcription factors such as Yin Yang 1 (YY1) at the basal state. We conclude that Xist is necessary for gene-specific silencing during XCI maintenance and impacts lineage-specific cell differentiation and proliferation during hematopoiesis.


Subject(s)
RNA, Long Noncoding , X Chromosome Inactivation , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Female , Fibroblasts/metabolism , Hematopoiesis/genetics , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , X Chromosome/metabolism , X Chromosome Inactivation/genetics
2.
Proc Natl Acad Sci U S A ; 115(52): 13336-13341, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30530664

ABSTRACT

Acetylation of histone H4 at lysine 16 (H4K16) modulates nucleosome-nucleosome interactions and directly affects nucleosome binding by certain proteins. In Drosophila, H4K16 acetylation by the dosage compensation complex subunit Mof is linked to increased transcription of genes on the single X chromosome in males. Here, we analyzed Drosophila containing different H4K16 mutations or lacking Mof protein. An H4K16A mutation causes embryonic lethality in both sexes, whereas an H4K16R mutation permits females to develop into adults but causes lethality in males. The acetyl-mimic mutation H4K16Q permits both females and males to develop into adults. Complementary analyses reveal that males lacking maternally deposited and zygotically expressed Mof protein arrest development during gastrulation, whereas females of the same genotype develop into adults. Together, this demonstrates the causative role of H4K16 acetylation by Mof for dosage compensation in Drosophila and uncovers a previously unrecognized requirement for this process already during the onset of zygotic gene transcription.


Subject(s)
Dosage Compensation, Genetic/genetics , Histones/genetics , Acetylation , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Histone Acetyltransferases/metabolism , Histones/metabolism , Lysine/genetics , Male , Nuclear Proteins/metabolism , Nucleosomes/metabolism , Phenotype , Point Mutation/genetics , Protein Processing, Post-Translational/genetics , Sex , Sex Factors , Transcription Factors/metabolism , X Chromosome/metabolism
3.
G3 (Bethesda) ; 4(1): 143-53, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24281426

ABSTRACT

During animal development, gene transcription is tuned to tissue-appropriate levels. Here we uncover antagonistic regulation of transcript levels in the germline of Caenorhabditis elegans hermaphrodites. The histone methyltransferase MES-4 (Maternal Effect Sterile-4) marks genes expressed in the germline with methylated lysine on histone H3 (H3K36me) and promotes their transcription; MES-4 also represses genes normally expressed in somatic cells and genes on the X chromosome. The DRM transcription factor complex, named for its Dp/E2F, Retinoblastoma-like, and MuvB subunits, affects germline gene expression and prevents excessive repression of X-chromosome genes. Using genome-scale analyses of germline tissue, we show that common germline-expressed genes are activated by MES-4 and repressed by DRM, and that MES-4 and DRM co-bind many germline-expressed genes. Reciprocally, MES-4 represses and DRM activates a set of autosomal soma-expressed genes and overall X-chromosome gene expression. Mutations in mes-4 and the DRM subunit lin-54 oppositely skew the transcript levels of their common targets and cause sterility. A double mutant restores target gene transcript levels closer to wild type, and the concomitant loss of lin-54 suppresses the severe germline proliferation defect observed in mes-4 single mutants. Together, "yin-yang" regulation by MES-4 and DRM ensures transcript levels appropriate for germ-cell function, elicits robust but not excessive dampening of X-chromosome-wide transcription, and may poise genes for future expression changes. Our study reveals that conserved transcriptional regulators implicated in development and cancer counteract each other to fine-tune transcript dosage.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Germ Cells/metabolism , X Chromosome/metabolism , Animals , Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation, Developmental , Microarray Analysis , Trans-Activators/genetics , Trans-Activators/metabolism , X Chromosome/genetics
4.
Genetika ; 47(7): 874-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21938950

ABSTRACT

The genotoxic and antigenotoxic effects of Cotinus coggygria Scop. methanol extract was investigated using the Drosophila sex-linked recessive lethal (or SLRL) test. The results presented here show that the methanol extract of Cotinus coggygria in a concentration of 5% and artificial chemical agent ethyl methanesulfonate EMS (0.75 ppm) induce recessive lethal mutations on X-chromosome on Drosophila melanogaster in all broods (I, II and III). Post-treatment with lower concentration of the methanol extract of Cotinus coggygria (2%) was effective in reducing genotoxicity ofmutagen.


Subject(s)
Anacardiaceae/chemistry , DNA Damage/drug effects , Methanol/chemistry , Mutagens/pharmacology , Plant Extracts/pharmacology , Animals , Drosophila melanogaster , Mutagens/chemistry , Plant Extracts/chemistry , X Chromosome/genetics , X Chromosome/metabolism
5.
Nature ; 460(7251): 128-32, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19536159

ABSTRACT

Pluripotency of embryonic stem (ES) cells is controlled by defined transcription factors. During differentiation, mouse ES cells undergo global epigenetic reprogramming, as exemplified by X-chromosome inactivation (XCI) in which one female X chromosome is silenced to achieve gene dosage parity between the sexes. Somatic XCI is regulated by homologous X-chromosome pairing and counting, and by the random choice of future active and inactive X chromosomes. XCI and cell differentiation are tightly coupled, as blocking one process compromises the other and dedifferentiation of somatic cells to induced pluripotent stem cells is accompanied by X chromosome reactivation. Recent evidence suggests coupling of Xist expression to pluripotency factors occurs, but how the two are interconnected remains unknown. Here we show that Oct4 (also known as Pou5f1) lies at the top of the XCI hierarchy, and regulates XCI by triggering X-chromosome pairing and counting. Oct4 directly binds Tsix and Xite, two regulatory noncoding RNA genes of the X-inactivation centre, and also complexes with XCI trans-factors, Ctcf and Yy1 (ref. 17), through protein-protein interactions. Depletion of Oct4 blocks homologous X-chromosome pairing and results in the inactivation of both X chromosomes in female cells. Thus, we have identified the first trans-factor that regulates counting, and ascribed new functions to Oct4 during X-chromosome reprogramming.


Subject(s)
Chromosome Pairing , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , X Chromosome Inactivation/genetics , X Chromosome/genetics , X Chromosome/metabolism , Animals , CCCTC-Binding Factor , Cell Line , Female , Humans , Male , Mice , Octamer Transcription Factor-3/deficiency , Octamer Transcription Factor-3/genetics , Protein Binding , RNA, Long Noncoding , RNA, Untranslated/genetics , SOXB1 Transcription Factors , Transcriptional Activation , YY1 Transcription Factor/metabolism
6.
Tsitologiia ; 50(3): 256-60, 2008.
Article in Russian | MEDLINE | ID: mdl-18664128

ABSTRACT

Using computer-based methods we determined the global distribution of short interspersed nuclear elements (SINEs) in the human and mouse X chromosomes. It has been shown that this distributions is similar to the distributions of CpG islands and genes but is different from the distribution of LINE1 elements. Since SINEs (human Alu and mouse B2) may have binding sites for Polycomb protein YY1, we suggest that these repeats can serve as additional signals ("boosters") in Polycomb-dependent silencing of gene rich segments during X inactivation.


Subject(s)
Short Interspersed Nucleotide Elements/genetics , X Chromosome/genetics , Alu Elements/genetics , Animals , Electronic Data Processing , Female , Heterochromatin/metabolism , Humans , Mammals/genetics , Mice , Short Interspersed Nucleotide Elements/physiology , X Chromosome/metabolism , X Chromosome Inactivation , YY1 Transcription Factor/metabolism
7.
Mol Cell Biol ; 27(24): 8760-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923683

ABSTRACT

In organisms with sex chromosomes, dosage compensation equalizes gene expression between the sexes. In Drosophila melanogaster males, the male-specific lethal (MSL) complex of proteins and two noncoding roX RNAs coat the X chromosome, resulting in a twofold transcriptional upregulation to equalize gene expression with that of females. How MSL complex enrichment on the X chromosome is regulated is not well understood. We performed an RNA interference screen to identify new factors required for dosage compensation. Using a Drosophila Schneider S2 cell line in which green fluorescent protein (GFP)-tagged MSL2 localizes to the X chromosome, we assayed approximately 7,200 knockdowns for their effects on GFP-MSL2 distribution. One factor identified is the zinc finger protein Zn72D. In its absence, the MSL complex no longer coats the X chromosome. We demonstrate that Zn72D is required for productive splicing of the transcript for the MSL protein Maleless, explaining the dosage compensation defect. However, Zn72D is required for the viability of both sexes, indicating its functions are not sex specific. Consistent with this, Zn72D colocalizes with elongating RNA polymerase II, implicating it as a more general factor involved in RNA metabolism.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , RNA Splicing/genetics , Transcription Factors/genetics , Zinc Fingers , Animals , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA, Complementary/metabolism , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Female , Genes, X-Linked , Introns , Male , Multiprotein Complexes/metabolism , Protein Transport , RNA Interference , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transgenes , X Chromosome/metabolism
8.
EMBO Rep ; 6(8): 748-54, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007070

ABSTRACT

The active and inactive X chromosomes have distinct epigenetic marks in somatic nuclei, which undergo reprogramming after transplantation into oocytes. We show that, despite the disappearance of Xist RNA coating in 30 min, the epigenetic memory of the inactive X persists with the precocious appearance of histone H3 trimethylation of lysine 27 (H3-3meK27), without the expected colocalization with Eed/Ezh2. Subsequently, Xist re-appears on the original inactive X, and the silent Xist on the active X undergoes re-activation, resulting in unusual biallelic Xist RNA domains. Despite this abnormal Xist expression pattern, colocalization of H3-3meK27 and Eed is thereafter confined to a single Xist domain, which is presumably on the original inactive X. These epigenetic events differ markedly from the kinetics of preferential paternal X inactivation in normal embryos. All the epigenetic marks on the X are apparently erased in the epiblast, suggesting that the oocyte and epiblast may have distinct properties for stepwise programming of the genome.


Subject(s)
Cell Nucleus/metabolism , Cloning, Organism/methods , Epigenesis, Genetic , Oocytes/metabolism , X Chromosome , Alleles , Animals , DNA Methylation , DNA, Complementary/metabolism , Female , Fibroblasts/metabolism , Gene Silencing , Histones/chemistry , In Situ Hybridization, Fluorescence , Kinetics , Lysine/chemistry , Mice , Mice, Inbred C57BL , Models, Biological , RNA/chemistry , RNA/metabolism , RNA, Long Noncoding , RNA, Untranslated/metabolism , Time Factors , Up-Regulation , X Chromosome/metabolism , X Chromosome Inactivation
9.
Mol Cell Biol ; 24(12): 5475-84, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169908

ABSTRACT

Histone H3 tail modifications are among the earliest chromatin changes in the X-chromosome inactivation process. In this study we investigated the relative profiles of two important repressive marks on the X chromosome: methylation of H3 lysine 9 (K9) and 27 (K27). We found that both H3K9 dimethylation and K27 trimethylation characterize the inactive X in somatic cells and that their relative kinetics of enrichment on the X chromosome as it undergoes inactivation are similar. However, dynamic changes of H3K9 and H3K27 methylation on the inactivating X chromosome compared to the rest of the genome are distinct, suggesting that these two modifications play complementary and perhaps nonredundant roles in the establishment and/or maintenance of X inactivation. Furthermore, we show that a hotspot of H3K9 dimethylation 5' to Xist also displays high levels of H3 tri-meK27. However, analysis of this region in G9a mutant embryonic stem cells shows that these two methyl marks are dependent on different histone methyltransferases.


Subject(s)
Histones/chemistry , X Chromosome/chemistry , Animals , Cells, Cultured , Dosage Compensation, Genetic , Female , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Kinetics , Lysine/chemistry , Male , Methylation , Mice , Mutation , Protein Methyltransferases , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , X Chromosome/genetics , X Chromosome/metabolism
10.
Curr Biol ; 10(24): R899-903, 2000.
Article in English | MEDLINE | ID: mdl-11137025

ABSTRACT

A new study shows that expression of Tsix, an antisense Xist gene, can be controlled by imprinting, and that high Tsix activity during X inactivation can protect the future active X chromosome from silencing by Xist. Tsix and Xist seem to have a yin and yang relationship, with opposite effects on X inactivation.


Subject(s)
Antisense Elements (Genetics) , Dosage Compensation, Genetic , Genomic Imprinting/physiology , RNA, Untranslated/genetics , Transcription Factors/genetics , X Chromosome/metabolism , Zygote/physiology , Animals , Gene Expression Regulation , Humans , RNA, Long Noncoding , X Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL