Download X inactivation Xplained

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Biology and consumer behaviour wikipedia , lookup

Genetic engineering wikipedia , lookup

Transposable element wikipedia , lookup

RNA interference wikipedia , lookup

Short interspersed nuclear elements (SINEs) wikipedia , lookup

Epigenomics wikipedia , lookup

Gene desert wikipedia , lookup

Gene therapy wikipedia , lookup

Oncogenomics wikipedia , lookup

Ridge (biology) wikipedia , lookup

Non-coding DNA wikipedia , lookup

Point mutation wikipedia , lookup

Epigenetics wikipedia , lookup

Cancer epigenetics wikipedia , lookup

Primary transcript wikipedia , lookup

Human genome wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

Minimal genome wikipedia , lookup

Epigenetics of neurodegenerative diseases wikipedia , lookup

History of genetic engineering wikipedia , lookup

Non-coding RNA wikipedia , lookup

RNA silencing wikipedia , lookup

Genome evolution wikipedia , lookup

Epigenetics of diabetes Type 2 wikipedia , lookup

Epigenetics in learning and memory wikipedia , lookup

Chromosome wikipedia , lookup

Vectors in gene therapy wikipedia , lookup

Gene expression profiling wikipedia , lookup

Mir-92 microRNA precursor family wikipedia , lookup

Epigenetics in stem-cell differentiation wikipedia , lookup

Gene expression programming wikipedia , lookup

Gene wikipedia , lookup

Therapeutic gene modulation wikipedia , lookup

Site-specific recombinase technology wikipedia , lookup

Polyploid wikipedia , lookup

Y chromosome wikipedia , lookup

Karyotype wikipedia , lookup

Microevolution wikipedia , lookup

RNA-Seq wikipedia , lookup

NEDD9 wikipedia , lookup

Long non-coding RNA wikipedia , lookup

Genomic imprinting wikipedia , lookup

Nutriepigenomics wikipedia , lookup

Designer baby wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Neocentromere wikipedia , lookup

Genome (book) wikipedia , lookup

Epigenetics of human development wikipedia , lookup

Skewed X-inactivation wikipedia , lookup

Polycomb Group Proteins and Cancer wikipedia , lookup

X-inactivation wikipedia , lookup

Transcript
X inactivation Xplained
Anton Wutz1 and Joost Gribnau2
Random inactivation of one of the two female X chromosomes
establishes dosage compensation between XY males and XX
females in placental mammals. X inactivation is controlled by
the X inactivation center (Xic). Recent advances in genome
sequencing show that the Xic has evolved from an ancestral
vertebrate gene cluster in placental mammals and has
undergone separate rearrangements in marsupials. The Xic
ensures that all but one X chromosome per diploid genome are
inactivated. Which chromosome remains active is randomly
chosen. Pairing of Xic loci on the two X chromosomes and
alternate states of the X chromosomes before inactivation have
recently been implicated in the mechanism of random choice.
Chromosome-wide silencing is then initiated by the noncoding
Xist RNA, which evolved with the mammalian Xic and covers
the inactive X chromosome.
Addresses
1
Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7,
1030 Vienna, Austria
2
Department of Reproduction and Development, Erasmus University
Medical Center, Rotterdam, The Netherlands
Corresponding author: Wutz, Anton ([email protected]) and
Gribnau, Joost ([email protected])
Current Opinion in Genetics & Development 2007, 17:387–393
This review comes from a themed issue on
Differentiation and gene regulation
Edited by Denis Duboule and Frank Grosveld
Available online 14th September 2007
0959-437X/$ – see front matter
# 2007 Elsevier Ltd. All rights reserved.
DOI 10.1016/j.gde.2007.08.001
Introduction
Mammals have evolved a complex set of functions for
gene regulation allowing elegant control of expression
during development. One of the most intriguing forms of
gene regulation leads to exclusive gene expression from
one single parental allele. Different mechanisms are used
to achieve monoallelic gene expression ranging from
parental imprinting [1] to odorant receptor choice in
the olfactory system [2]. These processes are tailored
to suit the requirements of different biological systems.
For instance, the immunoglobulin heavy chain rearrangements in B-lymphopoiesis generate a pool of cells each
expressing one specific antibody, which allows for clonal
selection in the immune system [3]. This process is
random with respect to the parental origin of the locus.
In contrast, imprinted genes show exclusive expression
from one parental allele [4]. In mammals dosage
www.sciencedirect.com
compensation of X-linked genes between the sexes is
established by inactivation of one of the two female Xs
(reviewed in [5]). In mice X chromosome inactivation
(XCI) is random in the embryo, and imprinted in extraembryonic tissues, where the paternally inherited X
chromosome is inactivated [6]. X inactivation, thus,
exemplifies the functions of the mammalian cell nucleus
in domain regulation and monoallelic expression. Here,
we review recent progress in understanding of the evolution of the mammalian dosage compensation system, the
mechanism of random choice and chromosome-wide
inactivation.
Evolution does dosage compensation
Dosage compensation systems adjust the gene doses
of sex chromosome linked genes between the sexes
and are observed throughout the animal kingdom.
Different strategies to accomplish dosage compensation
are used by fruit flies, worms, and mammals (reviewed
in [7]). Interestingly, no evidence for efficient dosage
compensation has been found by genome-wide expression analysis in chicken and finches [8]. Differences in
Z gene dosis between ZZ males and ZW females
result in elevated Z gene levels in males and might
contribute to sexual differentiation in birds. The observation of dosis compensation systems in other organisms must therefore be explained by an additional
advantage rather than an absolute necessity during
evolution. This hypothesis is highlighted by the observation that many genes escape XCI in human [9], and
supported by the intriguing finding that monoallelic
expression of the entire X chromosome has facilitated
the evolution of trichromatic color vision in monkeys
[10].
The mammalian dosage compensation system is controlled to a large extent by the X inactivation center
(Xic) located on the X chromosome. The Xic regulates
counting of the number of X chromosomes and contains
the noncoding Xist RNA gene, which localizes specifically
to the inactive X chromosome (Xi) and triggers chromosome-wide gene repression. Recent findings show that
this locus has apparently evolved exclusively in placental
mammals [11,12,13]. In marsupials and the more
distant vertebrates, such as frog or fish, the genomic
region around the Xic has an entirely different structure
with evidence for multiple rearrangements (Figure 1).
Interestingly, a different study indicates that the Xist
gene has newly emerged from an ancestral protein-coding
gene Lnx3 during mammalian evolution [14], indicating
that XCI in marsupials requires an alternative mechanism
for dosage compensation.
Current Opinion in Genetics & Development 2007, 17:387–393
388 Differentiation and gene regulation
Figure 1
Evolution of the X inactivation center. (a) A scheme of the Xic locus in placental mammals is shown. The Xist and regulatory Tsix transcript are
indicated. Genes that are derived from vertebrate protein-coding genes and have lost protein-coding potential in eutheria are indicated in blue.
Boxed genes serve as anchors for identifying the genomic region in vertebrates and marsupials. (b) The ancestral vertebrate gene cluster
corresponding to the mammalian Xic region has undergone an expansion during placental mammal evolution. The protein-coding LNX3 gene
shown in green shares sequence homology in a few exons with the noncoding Xist and is believed to be the ancestral Xist gene. (c) In the
marsupial lineage the ancestral vertebrate gene cluster has been rearranged and split in two clusters of genes, which map to distant regions on
the X. Xist has exclusively evolved in placental mammals and in marsupial genomes the LNX3 protein-coding gene is found.
Regulation of randomness
How random X inactivation is established is unclear at
present. Recent progress in the search for mechanisms
driving the XCI counting and choice process, which
mark the initiation of XCI, has produced evidence that
multiple regulatory systems may be involved. Different
observations made with aneuploid, and tetraploid cells
indicated that one X chromosome remains active per
diploid genome after completion of XCI. These and other
findings led to the hypothesis that counting and choice in
XCI can be explained by the action of an autosomal
blocking factor (BF), which protects one X chromosome
per diploid genome from inactivation (Figure 2a). Studies
aimed to identify the blocking factor binding site have
resulted in the identification of a 1.2 kb candidate
element, DXPas34, which is located 30 of Xist [15].
Deletion of this element resulted in ectopic XCI in male
cells, indicating a role for this element in the counting
process [16]. Surprisingly, a different study describing a
similar deletion of DXPas34 reported no ectopic XCI in
Current Opinion in Genetics & Development 2007, 17:387–393
male cells, indicating that more work has to be done to
clarify this topic [17].
The real nature of the blocking factor remains elusive so
far, and could be a nuclear episome, or a limited protein or
protein complex. Based on the blocking factor model
using theoretical considerations a symmetry-breaking
model has been proposed, in which many diffusible
molecules are quantitatively sequestered on the active
X chromosome by a mechanism that involves intermolecular BF interactions [18] (Figure 2b). Close proximity of
the Xic loci would benefit this model to allow binding
equilibration to occur fast enough to be able to discriminate between an active blocked and an inactive nonblocked X chromosome. Indeed, trans-interaction of X
chromosomes by Xic–Xic pairing has been observed
[19,20]. Based on this observation a mutual exclusive
choice model has been proposed that posits coordinate
trans-regulation of the interacting Xics [21] (Figure 2d).
Trans-vection is known from the fruit fly [22] and may aid
www.sciencedirect.com
X inactivation Xplained Wutz and Gribnau 389
Figure 2
Different models for the initiation of random XCI. (a) An autosomally encoded blocking factor (BF) protects one X chromosome from inactivation.
(b) Breaking of the symmetry in particle binding leads to accumulation of many diffusible molecules on one X chromosome protecting it from
inactivation. (c) Alternate epigenetic states mark the future Xa and Xi before the initiation of XCI. (d) Both X chromosomes meet in space and
determine the future Xa and Xi. (e) In a stochastic model each X chromosome has a certain probability to be inactivated.
blocking factor binding. The precise function of the Xic
interaction remains to be determined. Notably, XCI
counting is not affected in female cells with a 65 kb
deletion 30 of Xist which shows a loss of Xic pairing [19].
In several models, the XCI counting and choice process is
anticipated to be triggered by developmental or differentiation cues. Nevertheless, X chromosomes might be
distinguished before inactivation by assuming alternative
states (Figure 2c). Recently, it has been reported that
cohesion of sister chromatids is regulated differentially
www.sciencedirect.com
between the two female X chromosomes [23]. This
could highlight an inherent difference between the two
genetically identical chromosomes in at least part of the
cell population. The different states may represent blocking factor binding to one chromosome, or accessibility or
transcriptional differences between the two Xics,
although more work is required to establish the molecular
basis of this phenomenon.
The potential risk of female lethality dictates a need for a
precise counting process. This assumption has spurred a
Current Opinion in Genetics & Development 2007, 17:387–393
390 Differentiation and gene regulation
hunt for regulatory mechanisms. Nevertheless, any given
mechanism of XCI counting and choice requires a stochastic element. In principle, this could be achieved
simply by inactivating each X chromosome with a certain
fixed and fine-tuned probability. It has been shown that
chaotic choice results if Tsix is disrupted on both Xic
alleles [24]. Surprisingly, this can result in viable female
mice, albeit with a low frequency [24] (Figure 2e). Therefore, chaotic choice may represent the basic mechanism
underlying the XCI counting and choice process. Currently, the molecular identity for factors involved in the
counting process remains unknown [5]. Suggestions that
beside an autosomally encoded blocking factor an
X-linked competence factor might stimulate X inactivation further complicate the scenarios [21]. The molecular underpinning is clearly complex and it is hard to
see how to discriminate between different models if not
by analysis of molecular components.
XCI starts with the accumulation of Xist along the future
inactive X chromosome. Recent work indicates that Xist
up-regulation is the consequence of sex-specific induction of Xist transcription on the future Xi and not stabilization of the Xist RNA [25]. In differentiated cells Xist
repression on the Xa is mediated by DNA methylation
[26]. A recent study shows that the methyl-DNA binding
protein Mbd2 is required for Xist repression and, thus,
provides a link to repressor complexes [27]. During the
early stages of XCI, Xist expression is regulated by the
Tsix gene. Tsix is a long untranslated RNA, which acts
predominantly in the nucleus and is transcribed in antisense direction over the Xist gene. A recent report shows
that Tsix does not require splicing for its function [28],
leaving open the question whether the act of transcription
or the Tsix RNA itself are required for Xist repression. Tsix
function requires the intronic DXPas34 element [16,17].
Deletion of this element results in nonrandom XCI of the
mutated X chromosome and a defect in imprinted XCI
[17]. DXPas34 harbors many putative CTCF binding
sites, frequently flanked by putative YY1 sites [29]. Several of these CTCF sites show differential methylation in
gametes and somatic cells, implying a direct role for this
element in imprinted and random XCI [30]. Moreover,
analysis of YY1 mutant embryos has indicated a role for
this protein in the regulation of Tsix and Xist [29], but it
remains to be established if YY1 and CTCF act as bona
fide blocking factors, or are required to perform structural
roles for Xic regulation.
A conundrum is the lack of conservation of Tsix and
DXPas34 between the mouse and human XIC. It has
been observed in human and bovine cells that the antisense TSIX RNA is coexpressed with XIST and, thus,
apparently does not act to repress XIST in all mammals
[31,32]. This raises the possibility that evolution has
selected different molecular components for regulating
randomness in different mammals.
Current Opinion in Genetics & Development 2007, 17:387–393
Building a cis-limited repressive nuclear
compartment
The human Xi is comprised of a heterogeneous chromatin
landscape [33]. Not all genes are repressed on the Xi and a
relatively large set of genes escape from inactivation in
human. Recently, a gene activity profile has been derived
for the human Xi [9]. The availability of the human
X chromosome sequence [34] allowed the identification
of X enriched sequence elements [35,36]. Importantly, a
prediction for gene inactivation and escape of inactivation
was reported [35]. The identified sequences originate in
part from genomic repeat elements of the L1 long interspersed elements (LINE) class, which were already
implicated in spreading of X inactivation [37]. Experimental support for this hypothesis comes from analyses
of mice carrying the T(X;4)37H X;autosome translocation
chromosome. This study shows that spreading of Xist
RNA is attenuated at the autosomal boundary of the
translocation, which has a low abundance of LINE
elements [38].
Albeit L1 elements are not exclusive to the X chromosome,
they might be part of the features on the X chromosome
that facilitate spreading of the dosage compensation complex. Recent studies indicate that in the interphase nucleus
the human Xi is organized into an outer rim of genes and a
repeat-rich core, to which Xist localizes [39]. In differentiated mouse ES cells, Xist forms a repressive compartment
from which the transcription machinery is excluded as a
first step in X inactivation [40]. This compartment
initially does not contain genes and is also not sufficient
for gene silencing [40]. Genes become repressed and
associate with the repressive compartment upon silencing,
which requires the repeat-A sequences on the 50 -end of Xist
RNA [41] (Figure 3a). How silencing is established and
how it is restricted to one specific chromosome requires
further studies. The function of Xist in gene silencing has
been reported to be restricted to early development and
specific somatic cell types such as precursors in the blood
system [42] (Figure 3b). This suggests that the pathways
for gene silencing are regulated in a cell type-specific
manner.
Once X inactivation has been established the silent
X chromosome is stably maintained in somatic cells.
Chromosome-wide chromatin analysis of X-linked epigenetic modifications confirmed many cytological observations, including hypoacetylation of histone H4,
accumulation of histone H3, K9, and K27 tri-methylation
and accumulation of histone variant macroH2A on the Xi
[43,44] (Figure 3b). These results suggest that a wide
variety of chromatin modifiers including Polycomb group
complexes, histone deacetylases, and DNA methylases act
in concert to maintain repression of the Xi [45]. Beside
profiles of histone modification marks along the X chromosome, a DNA methylation profile of the X chromosome
has also been established [46]. This study shows that in
www.sciencedirect.com
X inactivation Xplained Wutz and Gribnau 391
Figure 3
Silencing of one X chromosome by Xist. (a) The two X chromosome territories are depicted. Xist RNA produced from the Xic locus accumulates
on the future Xi. This leads to the formation of a nuclear compartment that is devoid of the transcription machinery, such as RNA polymerase II
(Pol II). Polycomb group complexes are recruited to the Xist-covered chromosome and establish chromosome-wide histone modifications. The
genes localize to the outside this compartment and are silenced in a manner that requires the Xist repeat-A sequence. Silencing of genes
coincides with their association with the Xist compartment. (b) A summary of chromatin modifications during initiation of X inactivation. Xist RNA
accumulation is followed by recruitment of Polycomb repressor complex 1 (PRC1), which mediates histone H2A lysine 119 ubiquitinylation
(H2AK119ub1), and PRC2, which mediates histone H3 lysine 27 tri-methylation (H3K27me3). Accumulation of histone H4 lysine 20 monomethylation can also be observed on the X. In this phase Xist triggers gene silencing. Initially X inactivation is reversible and genes become
reactivated if Xist is lost. If cell differentiation progresses histone macroH2A becomes enriched on the Xi and histone H4 is hypoacetylated. Later
during the XCI process DNA cytosine methylation marks the promoters of genes on the Xi. In this phase X inactivation is irreversible and Xist
is not required for maintenance of the silent state. In somatic cells Xist does not have the capacity to initiate chromosome-wide silencing. Thus,
the silencing function of Xist depends on the Xist repeat-A sequence and a defined cellular context.
contrast to CpG island methylation found on the Xi, DNA
methylation is surprisingly more abundant on the Xa and
appears to be confined to gene body sequences on the Xa.
This finding may be related to the doubling of gene
expression of the single Xa relative to autosomes [47].
Intriguingly, in female XX ES cells global DNA hypomethylation is observed [48]. Could this possibly indicate a
potential function of demethylation in the counting process? The same epigenetic marks appear to be used at
different places for different regulatory functions in X
inactivation. This highlights a crucial role for spatial organization in complex gene regulatory mechanisms.
The Barr body has been originally observed to locate
sometimes close to the nucleolus [27]. A recent investigation has quantified the location of the Xi in differentiating ES cells and demonstrated that the association of the Xi
with the nucleolus occurs in S phase [49]. This has led to
www.sciencedirect.com
speculation about a role for the subnuclear localization in
providing an environment for stable replication of heterochromatin. It has been shown before that the Xi replicates
out of synchrony with other chromosomes either early in
S-phase in preimplantation embryos and late in S-phase in
embryonic cells after implantation [6]. This highlights a
spatial and temporal separation of the facultative heterochromatin of the Xi. Deletion of the Xist gene has been
shown to result in loss of perinucleolar position of the Xi,
but reactivation of the chromosome is limited to single
genes in occasional cells [49]. Moreover late replication and
chromosome-wide histone H4 hypoacetylation are maintained independent of Xist, yet, histone methylation marks
and macroH2A are lost without Xist [50–52]. These
results indicate that many superimposed epigenetic mechanisms maintain silencing of the Xi, and the exact role of
the temporal association of the Xi with the nucleolus in
maintaining the silent state remains to be determined.
Current Opinion in Genetics & Development 2007, 17:387–393
392 Differentiation and gene regulation
Conclusion and outlook
There is increasing evidence that indexing of the information stored in the genome is aided by chromatin
modifications such as histone methylation and acetylation. A combination of these signals has been proposed
to constitute a histone code [53]. Recently, it has become
clear that this is only one side of the full story and that
nuclear positioning and compartmentalization carry an
equal if not greater weight [54]. The chromosome-wide
phenomenon of X inactivation might hold promise to
further unravel the interplay between chromatin modifications and compartmentalization and will shed light on
the gene regulatory repertoire of mammals.
References and recommended reading
Papers of particular interest, published within the annual period of
review, have been highlighted as:
of special interest
of outstanding interest
1.
Yang PK, Kuroda MI: Noncoding RNAs and intranuclear
positioning in monoallelic gene expression. Cell 2007,
128:777-786.
2.
Lomvardas S, Barnea G, Pisapia DJ, Mendelsohn M, Kirkland J,
Axel R: Interchromosomal interactions and olfactory receptor
choice. Cell 2006, 126:403-413.
3.
Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Busslinger M:
Pax5 induces V-to-DJ rearrangements and locus contraction
of the immunoglobulin heavy-chain gene. Genes Dev 2004,
18:411-422.
4.
Pauler FM, Koerner MV, Barlow DP: Silencing by imprinted
noncoding RNAs: is transcription the answer? Trends Genet
2007, 23:284-292.
5.
Heard E, Disteche CM: Dosage compensation in mammals:
fine-tuning the expression of the X chromosome. Genes Dev
2006, 20:1848-1867.
6.
Takagi N, Sasaki M: Preferential inactivation of the paternally
derived X chromosome in the extraembryonic membranes of
the mouse. Nature 1975, 256:640-642.
7.
Lucchesi JC, Kelly WG, Panning B: Chromatin remodeling in
dosage compensation. Annu Rev Genet 2005, 39:615-651.
8.
Itoh Y, Melamed E, Yang X, Kampf K, Wang S, Yehya N, Van Nas A,
Replogle K, Band MR, Clayton DF et al.: Dosage compensation is
less effective in birds than in mammals. J Biol 2007, 6:2.
9.
Carrel L, Willard HF: X-inactivation profile reveals extensive
variability in X-linked gene expression in females. Nature 2005,
434:400-404.
This paper contains a comprehensive X inactivation profile of the human X
chromosome in fibroblast-like cells. About 15% of X-linked genes escape
inactivation to some degree. Variable patterns of escaping genes are
found between different cell lines suggesting a potential expression
heterogeneity among females.
10. Jacobs GH, Williams GA, Cahill H, Nathans J: Emergence of
novel color vision in mice engineered to express a human cone
photopigment. Science 2007, 315:1723-1725.
11. Davidow LS, Breen M, Duke SE, Samollow PB, McCarrey JR,
Lee JT: The search for a marsupial XIC reveals a break with
vertebrate synteny. Chromosome Res 2007, 15:137-146.
In this report, the authors fail to identify an ortholog of Xist in the marsupial
Monodelphis domestica. Cytological mapping of the area homologs to
the eutherian XIC indicates that this area is rearranged in marsupials.
12. Hore TA, Koina E, Wakefield MJ, Marshall Graves JA: The region
homologous to the X-chromosome inactivation centre has
been disrupted in marsupial and monotreme mammals.
Chromosome Res 2007, 15:147-161.
Current Opinion in Genetics & Development 2007, 17:387–393
This study describes the localization of the genes flanking the XIST gene
in eutherians along the marsupial X chromosome. The authors report that
XIST is absent in marsupials and monotremes and originated in a
genomic region that expanded in early mammals. The region homologous
to the eutherian XIC was disrupted in marsupials and monotreme
lineages.
13. Shevchenko AI, Zakharova IS, Elisaphenko EA, Kolesnikov NN,
Whitehead S, Bird C, Ross M, Weidman JR, Jirtle RL,
Karamysheva TV et al.: Genes flanking Xist in mouse and human
are separated on the X chromosome in American marsupials.
Chromosome Res 2007, 15:127-136.
The localization of the sequence interval between the eutherian genes
flanking XIST is analyzed in two different marsupial species Monodelphis
domestica and Didelphis virginia. The study indicates a breakpoint in this
region in both marsupial species.
14. Duret L, Chureau C, Samain S, Weissenbach J, Avner P: The Xist
RNA gene evolved in eutherians by pseudogenization of a
protein-coding gene. Science 2006, 312:1653-1655.
Sequence analysis of XIST and flanking regions in different eutherian
species and in opposum, chicken and frog reveals that XIST evolved at
least in part from an ancestral protein-coding gene Lnx3. This study
indicates that XIST is absent in marsupials suggesting the presence of
different mechanisms of XCI in eutherians and marsupials.
15. Debrand E, Chureau C, Arnaud D, Avner P, Heard E: Functional
analysis of the DXPas34 locus, a 30 regulator of Xist
expression. Mol Cell Biol 1999, 19:8513-8525.
16. Vigneau S, Augui S, Navarro P, Avner P, Clerc P: An essential role
for the DXPas34 tandem repeat and Tsix transcription in the
counting process of X chromosome inactivation. Proc Natl
Acad Sci USA 2006, 103:7390-7395.
17. Cohen DE, Davidow LS, Erwin JA, Xu N, Warshawsky D, Lee JT:
The DXPas34 repeat regulates random and imprinted X
inactivation. Dev Cell 2007, 12:57-71.
18. Nicodemi M, Prisco A: Symmetry-breaking model for
X-chromosome inactivation. Phys Rev Lett 2007, 98:108104.
19. Bacher CP, Guggiari M, Brors B, Augui S, Clerc P, Avner P, Eils R,
Heard E: Transient colocalization of X-inactivation centres
accompanies the initiation of X inactivation. Nat Cell Biol 2006,
8:293-299.
This study and the study by Xu et al. [20] indicate that both Xics
transiently pair at the onset of XCI in female cells. Analysis of Xic–Xic
pairing in Tsix mutant cell lines and cell lines with single copy and
multicopy Xic transgenes indicates that colocalization may be necessary
but not sufficient for correct counting and choice.
20. Xu N, Tsai CL, Lee JT: Transient homologous chromosome
pairing marks the onset of X inactivation. Science 2006,
311:1149-1152.
Similar to Bacher et al. [19] Xic pairing is reported during the early stages
of XCI. Also in this study Tsix and Xite mutant cell lines and cell lines with
autosomal Xic transgenes have been analyzed with respect to Xic pairing.
Results with Xite and Tsix mutant cell lines indicate that these sequences
are required for Xic–Xic pairing. Analysis of cell lines with small Tsix or Xite
fragments capable of inducing Xic-autosome pairing, indicates that Xic
pairing is an intrinsic part of the counting and choice process.
21. Anguera MC, Sun BK, Xu N, Lee JT: X-chromosome kiss and tell:
how the xs go their separate ways. Cold Spring Harb Symp
Quant Biol 2006, 71:429-437.
22. Vazquez J, Muller M, Pirrotta V, Sedat JW: The Mcp element
mediates stable long-range chromosome–chromosome
interactions in Drosophila. Mol Biol Cell 2006, 17:2158-2165.
23. Mlynarczyk-Evans S, Royce-Tolland M, Alexander MK,
Andersen AA, Kalantry S, Gribnau J, Panning B: X chromosomes
alternate between two states prior to random X-inactivation.
PLoS Biol 2006, 4:e159.
This study describes a difference in sister chromatid cohesion in at least
part of the cell population which is present already before the onset of
XCI. The difference in sister chromatid cohesion, as determined by DNA
FISH, correlates with the outcome of the XCI choice process, and
indicates that alternate states present before the XCI process may direct
the choice process.
24. Lee JT: Homozygous Tsix mutant mice reveal a sex-ratio
distortion and revert to random X-inactivation. Nat Genet 2002,
32:195-200.
www.sciencedirect.com
X inactivation Xplained Wutz and Gribnau 393
25. Sun BK, Deaton AM, Lee JT: A transient heterochromatic state
in Xist preempts X inactivation choice without RNA
stabilization. Mol Cell 2006, 21:617-628.
26. Beard C, Li E, Jaenisch R: Loss of methylation activates Xist
in somatic but not in embryonic cells. Genes Dev 1995,
9:2325-2334.
27. Barr H, Hermann A, Berger J, Tsai HH, Adie K, Prokhortchouk A,
Hendrich B, Bird A: Mbd2 contributes to DNA methylationdirected repression of the xist gene. Mol Cell Biol 2007,
27:3750-3757.
28. Sado T, Hoki Y, Sasaki H: Tsix defective in splicing is competent
to establish Xist silencing. Development 2006, 133:4925-4931.
29. Donohoe ME, Zhang LF, Xu N, Shi Y, Lee JT: Identification of a
Ctcf cofactor, Yy1, for the X chromosome binary switch. Mol
Cell 2007, 25:43-56.
30. Prissette M, El-Maarri O, Arnaud D, Walter J, Avner P: Methylation
profiles of DXPas34 during the onset of X-inactivation. Hum
Mol Genet 2001, 10:31-38.
31. Migeon BR, Lee CH, Chowdhury AK, Carpenter H: Species
differences in TSIX/Tsix reveal the roles of these genes in
X-chromosome inactivation. Am J Hum Genet 2002, 71:286-293.
32. Farazmand A, Basrur PK, Stranzinger G, Graphodatskaya D,
Reyes ER, King WA: Expression of XIST sense and antisense in
bovine fetal organs and cell cultures. Chromosome Res 2004,
12:275-283.
33. Chadwick BP, Willard HF: Multiple spatially distinct types of
facultative heterochromatin on the human inactive X
chromosome. Proc Natl Acad Sci USA 2004, 101:17450-17455.
34. Ross MT, Grafham DV, Coffey AJ, Scherer S, McLay K, Muzny D,
Platzer M, Howell GR, Burrows C, Bird CP et al.: The DNA
sequence of the human X chromosome. Nature 2005,
434:325-337.
35. Carrel L, Park C, Tyekucheva S, Dunn J, Chiaromonte F,
Makova KD: Genomic environment predicts expression
patterns on the human inactive X chromosome. PLoS Genet
2006, 2:e151.
40. Chaumeil J, Le Baccon P, Wutz A, Heard E: A novel role for Xist
RNA in the formation of a repressive nuclear compartment into
which genes are recruited when silenced. Genes Dev 2006,
20:2223-2237.
This study shows that exclusion of components of the transcription
machinery from the Xi represents the earliest event following Xist RNA
accumulation. X-linked genes are initially found outside the Xist domain.
Upon silencing, genes associate with the Xist RNA compartment in a
manner that depends on repeat-A sequences of Xist.
41. Wutz A, Rasmussen TP, Jaenisch R: Chromosomal silencing
and localization are mediated by different domains of Xist
RNA. Nat Genet 2002, 30:167-174.
42. Savarese F, Flahndorfer K, Jaenisch R, Busslinger M, Wutz A:
Hematopoietic precursor cells transiently reestablish
permissiveness for X inactivation. Mol Cell Biol 2006,
26:7167-7177.
43. Brinkman AB, Roelofsen T, Pennings SW, Martens JH,
Jenuwein T, Stunnenberg HG: Histone modification patterns
associated with the human X chromosome. EMBO Rep 2006,
7:628-634.
44. Valley CM, Pertz LM, Balakumaran BS, Willard HF: Chromosomewide, allele-specific analysis of the histone code on the human
X chromosome. Hum Mol Genet 2006, 15:2335-2347.
45. Masui O, Heard E: RNA and protein actors in x-chromosome
inactivation. Cold Spring Harb Symp Quant Biol 2006,
71:419-428.
46. Hellman A, Chess A: Gene body-specific methylation on the
active X chromosome. Science 2007, 315:1141-1143.
47. Nguyen DK, Disteche CM: Dosage compensation of the active X
chromosome in mammals. Nat Genet 2006, 38:47-53.
48. Zvetkova I, Apedaile A, Ramsahoye B, Mermoud JE, Crompton LA,
John R, Feil R, Brockdorff N: Global hypomethylation of the
genome in XX embryonic stem cells. Nat Genet 2005,
37:1274-1279.
49. Zhang LF, Huynh KD, Lee JT: Perinucleolar targeting of the
inactive X during S phase: evidence for a role in the
maintenance of silencing. Cell 2007, 129:693-706.
36. McNeil JA, Smith KP, Hall LL, Lawrence JB: Word frequency
analysis reveals enrichment of dinucleotide repeats on the
human X chromosome and [GATA]n in the X escape region.
Genome Res 2006, 16:477-484.
50. Csankovszki G, Panning B, Bates B, Pehrson JR, Jaenisch R:
Conditional deletion of Xist disrupts histone macroH2A
localization but not maintenance of X inactivation. Nat Genet
1999, 22:323-324.
37. Lyon MF: The Lyon and the LINE hypothesis. Semin Cell Dev Biol
2003, 14:313-318.
51. Kohlmaier A, Savarese F, Lachner M, Martens J, Jenuwein T,
Wutz A: A chromosomal memory triggered by Xist regulates
histone methylation in X inactivation. PLoS Biol 2004, 2:E171.
38. Popova BC, Tada T, Takagi N, Brockdorff N, Nesterova TB:
Attenuated spread of X-inactivation in an X;autosome
translocation. Proc Natl Acad Sci USA 2006, 103:7706-7711.
39. Clemson CM, Hall LL, Byron M, McNeil J, Lawrence JB: The X
chromosome is organized into a gene-rich outer rim and an
internal core containing silenced nongenic sequences. Proc
Natl Acad Sci USA 2006, 103:7688-7693.
This paper describes the higher order organization of the human Xi.
Genes are positioned on the outer rim of the Xi and active X chromosome.
The core of the X chromosome excludes genes and contains noncoding
sequences and genomic repeats.
www.sciencedirect.com
52. Schoeftner S, Sengupta AK, Kubicek S, Mechtler K, Spahn L,
Koseki H, Jenuwein T, Wutz A: Recruitment of PRC1 function at
the initiation of X inactivation independent of PRC2 and
silencing. EMBO J 2006, 25:3110-3122.
53. Turner BM: Defining an epigenetic code. Nat Cell Biol 2007,
9:2-6.
54. Cremer T, Cremer M, Dietzel S, Muller S, Solovei I, Fakan S:
Chromosome territories — a functional nuclear landscape.
Curr Opin Cell Biol 2006, 18:307-316.
Current Opinion in Genetics & Development 2007, 17:387–393