Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO J ; 18(10): 2897-907, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10329635

ABSTRACT

We have investigated the role of histone acetylation in X chromosome inactivation, focusing on its possible involvement in the regulation of Xist, an essential gene expressed only from the inactive X (Xi). We have identified a region of H4 hyperacetylation extending up to 120 kb upstream from the Xist somatic promoter P1. This domain includes the promoter P0, which gives rise to the unstable Xist transcript in undifferentiated cells. The hyperacetylated domain was not seen in male cells or in female XT67E1 cells, a mutant cell line heterozygous for a partially deleted Xist allele and in which an increased number of cells fail to undergo X inactivation. The hyperacetylation upstream of Xist was lost by day 7 of differentiation, when X inactivation was essentially complete. Wild-type cells differentiated in the presence of the histone deacetylase inhibitor Trichostatin A were prevented from forming a normally inactivated X, as judged by the frequency of underacetylated X chromosomes detected by immunofluorescence microscopy. Mutant XT67E1 cells, lacking hyperacetylation upstream of Xist, were less affected. We propose that (i) hyperacetylation of chromatin upstream of Xist facilitates the promoter switch that leads to stabilization of the Xist transcript and (ii) that the subsequent deacetylation of this region is essential for the further progression of X inactivation.


Subject(s)
Dosage Compensation, Genetic , Histones/metabolism , RNA, Untranslated , Transcription Factors/genetics , X Chromosome/genetics , Acetylation , Animals , Cell Differentiation , Cell Division , Cell Line , Chromatin/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Mice , Mutation , Promoter Regions, Genetic , RNA, Long Noncoding , Stem Cells , Time Factors
2.
Hum Mol Genet ; 8(2): 195-204, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9931327

ABSTRACT

The propagation of X chromosome inactivation is thought to be mediated by the cis- limited spreading of the non-protein coding Xist transcript. In this report we have investigated the localization of Xist RNA on rodent metaphase chromosomes. We show that Xist RNA exhibits a banded pattern on the inactive X and is excluded from regions of constitutive heterochromatin. The banding pattern suggests a preferential association with gene-rich, G-light regions. Analysis of X:autosome rearrangements revealed that restricted propagation of X inactivation into cis -linked autosomal material is reflected by a corresponding limited spread of Xist RNA. We discuss these results in the context of models for the function of Xist RNA in the propagation of X inactivation.


Subject(s)
Dosage Compensation, Genetic , RNA, Untranslated , Transcription Factors/genetics , X Chromosome/genetics , Animals , Arvicolinae , Cells, Cultured , Chromosome Mapping , Female , In Situ Hybridization, Fluorescence , Male , Metaphase/genetics , Mice , RNA/genetics , RNA, Long Noncoding , Translocation, Genetic
3.
Hum Mol Genet ; 8(2): 377-83, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9931347

ABSTRACT

The inactive X (Xi) differs from its active homologue (Xa) in a number of ways, including increased methylation of CpG islands, replication late in S phase, underacetylation of histone H4 and association with XIST RNA. Global changes in DNA methylation occur relatively late in development, but the other properties all change during or shortly after the establishment of Xi and may play a role in the mechanism by which an inactive chromatin conformation spreads across most of the chromosome. In the present report, we use two human X;autosome translocation chromosomes to study the spreading of inactive X chromatin across X;autosome boundaries. In one of these chromosomes, t(X;6), Xp distal to p11.2 is replaced by 6p21.1-6pter and, in the other, ins(X;16), a small fragment derived from 16p13 is inserted into the distal third of Xq. In lymphoid cells from patients carrying these translocations in an unbalanced form, Xi was shown by HUMARA assay to be derived exclusively [t(X:6)] or predominantly [ins (X;16)] from the derived X chromosome. We used a combination of immunolabelling and RNA/DNA fluorescence in situ hybridization to define the distribution of XIST RNA, deacetylated H4 and late-replicating DNA across the two derived X chromosomes in inactive form. Within the limits of the cytogenetic techniques employed, the results show complete coincidence of these three parameters, with all three being excluded from the autosomal component of the derived X chromosome.


Subject(s)
Chromosome Aberrations/genetics , DNA Replication/genetics , Histones/metabolism , RNA, Untranslated , RNA/metabolism , Transcription Factors/genetics , X Chromosome/genetics , Acetylation , Cell Line, Transformed , Chromosome Aberrations/pathology , Chromosome Banding , Chromosome Disorders , Chromosome Painting , Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 6/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , RNA/genetics , RNA, Long Noncoding , Time Factors , Translocation, Genetic
4.
Dev Genet ; 22(1): 65-73, 1998.
Article in English | MEDLINE | ID: mdl-9499581

ABSTRACT

In mammals, the levels of X-linked gene products in males and females are equalised by the silencing, early in development, of most of the genes on one of the two female X chromosomes. Once established, the silent state is stable from one cell generation to the next. In eutherian mammals, the inactive X chromosome (Xi) differs from its active homologue (Xa) in a number of ways, including increased methylation of selected CpGs, replication late in S-phase, expression of the Xist gene with binding of Xist RNA and underacetylation of core histones. The latter is a common property of genetically inactive chromatin but, in the case of Xi, it is not clear whether it is an integral part of the silencing process or simply a consequence of some other property of Xi, such as late replication. The present review describes two approaches that address this problem. The first shows that Xi in marsupial mammals also contains underacetylated H4, even though its properties differ widely from those of the eutherian Xi. The continued presence of histone underacetylation on Xi in these evolutionarily distant mammals argues for its fundamental importance. The second approach uses mouse embryonic stem cells and places H4 deacetylation in a sequence of events leading to complete X inactivation. The results argue that histone underacetylation plays a role in the stabilisation of the inactive state, rather than in its initiation.


Subject(s)
Dosage Compensation, Genetic , Histones/metabolism , Mammals/genetics , Acetylation , Animals , Blastocyst/cytology , Cell Differentiation/physiology , Female , Humans , Male , Marsupialia/genetics , Stem Cells/cytology
5.
Proc Natl Acad Sci U S A ; 94(18): 9665-8, 1997 Sep 02.
Article in English | MEDLINE | ID: mdl-9275180

ABSTRACT

Underacetylation of histone H4 is thought to be involved in the molecular mechanism of mammalian X chromosome inactivation, which is an important model system for large-scale genetic control in eukaryotes. However, it has not been established whether histone underacetylation plays a critical role in the multistep inactivation pathway. Here we demonstrate differential histone H4 acetylation between the X chromosomes of a female marsupial, Macropus eugenii. Histone underacetylation is the only molecular aspect of X inactivation known to be shared by marsupial and eutherian mammals. Its strong evolutionary conservation implies that, unlike DNA methylation, histone underacetylation was a feature of dosage compensation in a common mammalian ancestor, and is therefore likely to play a central role in X chromosome inactivation in all mammals.


Subject(s)
Histones/genetics , X Chromosome , Acetylation , Animals , Female , Histones/metabolism , Marsupialia
6.
Exp Cell Res ; 230(2): 399-402, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9024800

ABSTRACT

It has previously been shown by immunocytochemistry that the inactive X chromosome (Xi) in somatic cells of human and mouse females is marked by underacetylation of histone H4. It has been suggested that this may be important for transcriptional silencing of genes on Xi. We have now investigated X-inactivation in meiotic cells of the male germline. In these cells the single X chromosome is transcriptionally inactive and expresses XIST, a gene that in somatic cells is transcribed only from Xi. By immunostaining with antibodies to H4 acetylated at lysines 5, 8, 12, or 16, we demonstrate that histone H4 on the male X is not underacetylated. We conclude that there is a differential germline strategy for maintenance of X-inactivation and that H4 underacetylation, though associated with the long-term marking of inactive X chromosomes in the female soma, is not always essential for the transcriptional down-regulation of X-linked genes.


Subject(s)
Histones/metabolism , RNA, Untranslated , Transcription Factors/metabolism , X Chromosome , Acetylation , Animals , Dosage Compensation, Genetic , Germ Cells , Heterochromatin/metabolism , Male , Meiosis , Mice , Mice, Inbred BALB C , RNA, Long Noncoding , Rabbits , Sertoli Cells/metabolism , Spermatids/metabolism , Testis/cytology , Testis/metabolism
7.
Dev Biol ; 180(2): 618-30, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8954732

ABSTRACT

In female mammalian cells, dosage compensation for X-linked genes is achieved by the transcriptional silencing, early in development, of many genes on just one of the two X chromosomes. Several properties distinguish the inactive X (Xi) from its active counterpart (Xa). These include expression of Xist, a gene located in the X-inactivation center (Xic), late replication, differential methylation of selected CpG islands and underacetylation of histone H4. The relationship between these properties and transcriptional silencing remains unclear. Female mouse embryonic stem (ES) cells have two active X chromosomes, one of which is inactivated as cells differentiate in culture. We describe here the use of these cells in studying the sequence of events leading to X-inactivation. By immunofluorescent labeling of metaphase chromosome spreads from ES cells with antibodies to acetylated H4, we show that an underacetylated X chromosome appears only after 4 days of differentiation, and only in female cells. The frequency of cells with an underacetylated X reaches a maximum by Day 6. In undifferentiated cells, H4 in centric heterochromatin is acetylated to the same extent as that in euchromatin but has become relatively underacetylated, as in adult cells, by Day 4 of differentiation (i.e. , when deacetylation of Xi is first seen). The overall deacetylation of Xi follows Xist expression and the first appearance of a single, late-replicating X, both of which occur on Day 2. It also follows the silencing of X-linked genes. Levels of mRNA from four such genes, Hprt, G6pd, Rps4, and Pgk-1, had all fallen by approximately 50% (relative to the autosomal gene Aprt) by Days 2-4. The results show that properties that characterize Xi are put in place in a set order over several days. H4 deacetylation occupies a defined place within this sequence, suggesting that it is an intrinsic part of the X-inactivation process. The stage at which a completely deacetylated Xi is first seen suggests that deacetylation may be necessary for the maintenance of silencing but is not required for its initiation. Nor is it required for, or an immediate consequence of, late replication. However, we note that selective deacetylation of H4 on specific genes would not be detected by the microscopical approach we have used and that such selective deacetylation may still be part of the silencing process.


Subject(s)
Histones/metabolism , RNA, Untranslated , Stem Cells/physiology , Transcription Factors/genetics , X Chromosome , Acetylation , Adenine Phosphoribosyltransferase/biosynthesis , Adenine Phosphoribosyltransferase/genetics , Animals , Cell Differentiation , Cell Line , Chromatin/physiology , Female , Genetic Markers , Glucosephosphate Dehydrogenase/genetics , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mammals , Mice , Phosphoglycerate Kinase/genetics , Polymerase Chain Reaction , RNA, Long Noncoding , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription, Genetic
8.
Exp Cell Res ; 225(2): 277-85, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8660915

ABSTRACT

The distribution of acetylated isoforms of histone H4 along Chinese hamster chromosomes has been studied by immunostaining with antibodies recognizing H4 acetylated at defined lysines in its N-terminal domain. The heterochromatic long arm of the X chromosome in both female (CHO) and male (DON) cell lines is underacetylated at three out of four lysines (5, 8, and 12). In contrast, the level of acetylation at lysine 16, which is the first to be acetylated in mammals, was similar in X chromosomes and autosomes. Labeling of the cells with bromodeoxyuridine (BrdU) to mark late-replicating chromosome domains, followed by double immunostaining with antibodies to BrdU and acetylated H4, showed a close, though not perfect, correlation between late replication and low levels of H4 acetylation. The results show that levels of histone acetylation are associated with the replication timing of defined domains on both the X chromosome and autosomes, but the exceptions we observe suggest that this link is not absolute or essential.


Subject(s)
CHO Cells/cytology , Chromosomes/physiology , Histones/genetics , Histones/metabolism , Acetylation , Animals , CHO Cells/metabolism , Cell Division/physiology , Cricetinae , DNA Replication , Female , Fluorescent Antibody Technique , Male , Metaphase/physiology , Time Factors
9.
Hum Genet ; 97(5): 573-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8655133

ABSTRACT

It has previously been shown that the acetylated forms of histone H4 are depleted or absent in both constitutive, centric heterochromatin and in the facultative heterochromatin of the inactive X chromosome (Xi) in female cells. By immunostaining of metaphase chromosomes from human lymphocytes with antibodies to the acetylated isoforms of histones H2A and H3, we now show that these histones too are underacetylated in both Xi and centric heterochromatin. Xi shows two prominent regions of residual H3 acetylation, one encompassing the pseudoautosomal region at the end of the short arm and one at about Xq22. Both these regions have been shown previously to be sites of residual H4 acetylation. H2A acetylation on Xi is higher overall than that of H3 or H4 and is particularly high around the pseudoautosomal region, but not at Xq22. The results suggest that the acetylated isoforms of H3 and H4 have at least some effects on chromosomal structure and function that are not shared by acetylated H2A.


Subject(s)
Histones/chemistry , Histones/metabolism , X Chromosome , Acetylation , Amino Acid Sequence , Chromosome Mapping , Enzyme-Linked Immunosorbent Assay , Female , Histones/isolation & purification , Humans , Karyotyping , Lymphocytes/cytology , Lymphocytes/physiology , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/immunology
SELECTION OF CITATIONS
SEARCH DETAIL