Você está na página 1de 9

FEBS Letters 588 (2014) 25142522

journal homepage: www.FEBSLetters.org

Review

X-chromosome inactivation in development and cancer


Ronan Chalign, Edith Heard
Mammalian Developmental Epigenetics Group, Genetics and Developmental Biology Unit, Institut Curie, CNRS UMR3215, INSERM U934, 75248 Paris, France

a r t i c l e i n f o a b s t r a c t

Article history: X-chromosome inactivation represents an epigenetics paradigm and a powerful model system of
Received 5 June 2014 facultative heterochromatin formation triggered by a non-coding RNA, Xist, during development.
Accepted 6 June 2014 Once established, the inactive state of the Xi is highly stable in somatic cells, thanks to a combina-
Available online 14 June 2014
tion of chromatin associated proteins, DNA methylation and nuclear organization. However,
sporadic reactivation of X-linked genes has been reported during ageing and in transformed cells
Edited by Wilhelm Just
and disappearance of the Barr body is frequently observed in cancer cells. In this review we summa-
rise current knowledge on the epigenetic changes that accompany X inactivation and discuss the
Keywords:
X-chromosome inactivation
extent to which the inactive X chromosome may be epigenetically or genetically perturbed in breast
Epigenetics cancer.
Cancer 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.
Development
Chromatin

1. Introduction some cases in cancer cells resulting in a double dose of X-linked


genes. However, another mechanism for Barr body loss in cancer
In mammals, dosage compensation for X-linked gene products that has been proposed involves the decompaction of its hetero-
between the sexes is achieved by X-chromosome inactivation chromatic structure, which may be accompanied by X-linked gene
(XCI) in females [1]. This process leads to the highly regulated tran- reactivation.
scriptional silencing of one of the two X-chromosome during early Here we provide a brief overview of the current knowledge
development, leading to the formation of the heterochromatic Barr pertaining to the establishment and maintenance of X inactivation,
body. X inactivation is an outstanding example of chromosome- as well as highlights of some of the known perturbations of the
wide epigenetic regulation involving the developmental silencing inactive X chromosomes state in a cancer context. This review
of approximately one thousand genes. XCI shares many of the fea- does not provide an in depth analysis, for this, the reader is
tures of others epigenetic mechanisms such as a mosaic cellular referred to other recent reviews [46].
phenotype; mitotic heritability but developmental reversibility of
X-chromosome inactivity; asynchronous DNA replication timing 2. Setting up inactivation of the X chromosome in mammals
compared to the rest of the genome; and nally a combination of
several epigenetic mechanisms including DNA promoter methyl- The X-inactivation center (Xic) is a key locus that is required for
ation, histones post-translational modications, and an unusual the initiation of X inactivation. Studies involving X-chromosome
nuclear organization. These various features are believed to act deletions and X-autosome translocations in mice and humans
synergistically to maintain the X inactive state. Thus, the inactive mapped the precise region which contains the essential gene
X chromosome represents a remarkable illustration of the numer- for triggering the silencing process, the non-coding RNA Xist
ous epigenetic mechanisms that can underlie the formation and (X-inactive-specic-transcript) (for review [7]). Xist is a 17 000
maintenance of facultative heterochromatin throughout the nucleotide (19 kb in Human), spliced, untranslated regulatory tran-
lifetime of mammals. Pathologists have long noted that the hetero- script that coats the X chromosome from which it is expressed in
chromatic structure of the Barr body was frequently absent in cis [8,9]. Perhaps surprisingly, Xist is present only in eutherian
breast cancer cells, particularly in the most aggressive tumors mammals, with no orthologous described in marsupials or mono-
([2,3]). This observation was later found to be due to the frequent tremes to date. However in marsupials, another non-coding RNA
genetic loss of the Xi, with reduplication of the Xa also occurring in Rsx (RNA on the silent X) was recently discovered, that appears
to have similar properties to Xist even though its sequence is unre-
Corresponding author. lated to Xist [10]. In the mouse, deletions and transgenes have
E-mail address: Edith.Heard@curie.fr (E. Heard). demonstrated that Xist is required for both the imprinted and

http://dx.doi.org/10.1016/j.febslet.2014.06.023
0014-5793/ 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.
R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522 2515

random forms of X inactivation [1113]. Nevertheless, Xist alone regulating Xist, such as an early maternal imprint to prevent pre-
cannot recapitulate all the roles of the Xic which include sens- mature XCI, as well as reactivation of the Xp to allow for random
ing/competence, whereby a cell initiates XCI only when more XCI (see [21,24] for discussion).
than one Xic is present; counting, where only one X chromosome The mechanisms underlying the early events during XCI are still
stays active per diploid autosome set; and choice, whereby one not fully understood. Nevertheless, gene silencing is clearly trig-
of the two X chromosomes is chosen for inactivation while the gered by Xist RNA coating of the future inactive X chromosome.
other remains active. Some of these functions seem to be ensured It has been proposed that the Xist transcript might bind to high
by other elements of the Xic, in the neighborhood of Xist. For exam- afnity sites on the chromosome, thereby inducing local hetero-
ple, in mice the cis-acting Xist antisense transcription unit, Tsix chromatinisation, which can then facilitate the spread of XCI.
[14], plays a key role in the choice of which chromosome will be LINE1 (Long Interpersed Nuclear Elements 1) repeat elements,
inactivated. However, several additional cis-acting elements and which are highly enriched on the X chromosome compared to
trans-acting factors are involved in the dosage-sensitive, monoall- autosomes, have been proposed to facilitate the spread of XCI, by
elic regulation of Xist expression during early differentiation. For participating in the local propagation of inactivation and facilitat-
example, the Rnf12 gene lies upstream of Xist and its protein prod- ing the recruitment of genes into the silent nuclear compartment
uct, an ubiquitin ligase that leads to the degradation of the Xist formed by Xist RNA [26]. However, direct genetic evidence proving
repressor protein Rex1, has been shown to have a key role in the that L1s are required for efcient spreading or gene silencing is still
XX-dosage dependent activation of Xist [15,16]. A recent study also lacking. More recent ndings, based on mapping of Xist RNA along
shed light on the precise chromosomal folding of the Xic region in the X chromosome and of the nuclear organization of the Xist RNA
ESCs, revealing that the Xist and Tsix promoters lie in separate coated inactive X, suggest that Xist propagation and coating are
Topologically Associated Domains (TADs) [17]. The Xist promoter highly related to the three-dimensional conformation of the inac-
and all of Xists known positive regulators, including Rnf12, lie in tive X chromosome, and possibly also to the binding of Polycomb
a single 500 kb TAD, while the Xist repressor Tsix and its regulators Repressive Complex 2 (PRC2) protein complex [2729]. However,
lie in the neighboring 220 kb TAD, with the Xist/Tsix overlapping although studies comparing PRC2/H3K27me3 density and Xist
gene bodies across the boundary between the two TADs. The spa- RNA mapping suggest co-localisation [27,28], this is not in agree-
tial separation between the Xist and Tsix promoters is likely to be ment with super-resolution Xist RNA FISH/immunouorescence
essential for the nely tuned reciprocal regulation and expression studies, where PRC2 and Xist RNA are found to be spatially segre-
of these two keys players in the XCI process [18,19]. gated [30]. Furthermore, these studies do not demonstrate that
So far, most studies of XCI during early development have been the regions of the X associated with Xist RNA are directly involved
carried out in mice, which are more amenable to a genetic in its spreading or silencing functions. Indeed, the mechanisms
approach and where access to embryonic material is easier. In this underlying Xists cis-limited chromosome coating capacity, as well
species, XCI occurs in two phases during embryogenesis, an initial as its ability to inactivate genes, remain mysterious.
imprinted form and a later random form. In the imprinted form, Several studies have focused on the early changes in chromatin
Xist itself is imprinted such that only the paternal allele is structure that occur during the initiation of XCI with the hope of
expressed from two cell stage, whereas the maternal allele is providing insight into the role of Xist and the establishment of
repressed by an as yet undened maternal imprint that is estab- the silent state of the inactive X. Loss of euchromatin-associated
lished during oogenesis. The paternal X chromosome (Xp) becomes histone modications (such as H3K9Ac, H3K4me2 and H3K4me3)
silenced during pre-implantation development and remains inac- are amongst the earliest chromatin changes that occur following
tive in extra-embryonic tissues. However the Xp is reactivated in Xist RNA coating [20,31,32] (Fig. 1). Global H4 hypoacetylation
the inner cell mass (ICM) of the mid-stage blastocyst, that will give occurs shortly afterwards [33]. Passive histone-loss during replica-
rise to the embryo proper [20]. XCI is then reinitiated and occurs tion or else active removal (either by enzymatic activity, proteo-
randomly by up-regulation of Xist from either the paternal or the lytic activity or histone exchange) may underlie the early
maternal X chromosome. This second wave of XCI, can be recapit- disappearance of those histones modications. In addition to these
ulated in vitro in differentiating mouse ESCs which provide a pow- early chromatin changes, the disappearance of factors associated
erful model system for the study of the molecular mechanisms with transcription, such as RNA polymerase II and loss of nascent
underlying random XCI. Remarkably, the differentiation process transcripts are observed on the Xi immediately after Xist RNA coat-
is delayed or paused in female ES cells until one of the two ing. One or two cell cycles later, several new histone modications
X chromosomes has become silenced, suggesting that a double appear on the Xist-coated chromosome. These include H3K27me3,
dose of X-linked genes interferes directly with differentiation, thus H3K9me2, H4k20me1 and H2Ak119ub1 (for review [22]) which all
creating a coupling between XCI and development [21]. Unexpect- become enriched with rather similar kinetics of Xi enrichment dur-
edly, in other mammals recent studies have revealed that the tim- ing random XCI in differentiating ESCs. In pre-implantation
ing and pattern of XCI initiation observed in rodents is rather embryos, H3K27 tri-methylation precedes H3K9 di-methylation
different to that seen in other eutherian mammals such as humans on the X chromosome undergoing inactivation [20]. Intriguingly,
and rabbits [22,23], where XIST is not imprinted but is expressed H3K27me3 and H3K9me3 seem to be enriched in different regions
from all X chromosomes in males and females from the 4 or 8 cells of the human inactive X chromosome in somatic cells, suggesting
stages [24]. In rabbits, a signicant proportion of blastomeres dis- the existence of two different types of heterochromatin [34]. In
play XIST RNA coating of both X chromosomes in early blastocysts. murine differentiating female ESCs and somatic cells, H3K27me3
This situation is rapidly resolved at slightly later stages, where and H3K9me2 are enriched rather uniformly along the Xi, though
most cells display only one XIST RNA-coated chromosome. In slightly more in gene-rich regions [35]. The Xi prole of H2Aub1
humans, the onset of XCI appears to be much slower than in mice has not yet been described.
and rabbits, as the two X chromosome remain active both in the The factors that lay down or bind to the different chromatin
ICM and the trophectoderm even though XIST coating is present. marks present on the Xi are being unraveled in mice. Polycomb
By contrast, early XCI appears to be absolutely essential in mice, repressive complex 1 (PRC1) mediates mono-ubiquitination of his-
in order to ensure proper dosage compensation given their fast tone H2A lysine 119 (H2Aub1) [36,37] and PRC2 catalyzes the tri-
embryonic development [25]. The exact reasons for this evolution- methylation of H3K27 [38,39]. Xist RNA is believed to have a role in
ary diversity are still not clear but the rapid evolution of rodent the targeting of both PRC1 and PRC2 to the Xi, though whether this
genomes may mean that they have acquired mechanisms for nely is direct or indirect is still unclear. In the case of PRC2, the cofactor
2516 R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522

A
Initiation Propagation Maintenance
Transcriptionally active chromatin state Reversibly silenced chromatin state Stable silenced chromatin state

PRC2 CpG islands methylation


SUZ12
XIST RNA ?
PCL2
XIST RNA JARID2 Dnmt
EED HMTase ?
EZH
G9a Smchd1
?
CH3 +
? ?

H3K4me3 H3K9ac H3K27me3 CDYL H3K9me2 H3K27me3 CDYL H3K9me2


CH3
CH3
CH3

CH3
CH3
CH3 CH3
CH3
CH3
CBX7
BMI1
DNA RNAP II PHC
H4Ac H3K4me3 RING1 H2AUb H3K20me1 H2AUb H3K20me1
PRC1
RNA
canonical
RYBP RING1 PR Set7 macroH2A
BMI1
?

PRC1-like

B Formation of a Xi silent compartment


Xist RNA coats the Xi and Genes escaping XCI remain outside the Xi domain and
forms a nuclear silent compartment are engaged in long-range contacts with each other

Xa Xi Xi

Xa Xa Xa
Nucleus

Two active X-chromosomes


Active X-linked gene Inactive X-linked gene Xist RNA

Fig. 1. The kinetics of X-chromosome inactivation. Schematic view of the kinetics of events on the X chromosome based on ndings of many laboratories, performed mainly
in differentiating female embryonic stem cells. (A) During the initiation of XCI, Xist is expressed in cis from the future inactive X chromosome. Then Xist RNA exerts its
repressive effect through unknown factors. One of the earliest events following Xist RNA coating is loss of euchromatic histone marks, H3K4me2/3, H3K9Ac and H4Ac. During
this same time window, X-linked gene-silencing initiates. Several histone modications also become enriched on the Xist RNA-coated chromosome at this time. These include
H3K27me3, H3K9me2, H2Aub1 and H4K20me1. The PRC2 complex proteins Eed, Pcl2, Suz12, Ezh2 and Jarid2 are also detectable on the X at this stage. Ezh2 is responsible for
the appearance of H3K27me3 on the X. Jarid2 plays a role in the targeting of PRC2 to the chromatin. The histone methyl-transferases responsible for H4K20 mono-
methylation is PRSet7 but that for H3K9 di-methylation is not yet clear. By binding H3K27me3/H3K9me2 enriched Xi, Cdyl may also participate in propagating
heterochromatin marks such as H3K9me2 through G9a recruitment. PRC1 canonical complex may be recruited through binding of the chromodomain region of the Cbx7
protein to H3K27me3 enriched chromatin; while the non-canonical (Rybp containing) PRC1 complex appears to be Xist RNA dependent. The mono-ubiquitination of histone
H2A is induced thanks to the Ring1b protein, although the exact role of this mark in X inactivation is not clear. At later stages of differentiation, in the maintenance phase, the
PRC2 and PRC1 complexes no longer appear to be present on the Xi. However, macroH2A becomes associated with the inactive X at the nal differentiation stage. The latest
mark to appear is DNA methylation of promoters of X-linked genes. DNA methylation is deposit thanks to Dnmt proteins. Smchd1 may play a role in deposition or
maintenance of DNA methylation. (B) During the XCI process, Xist RNA coating on the inactive X chromosome induces the formation of a nuclear silent compartment. The
genes escaping silencing are excluded from this nuclear domain and then preferentially interact with each other in a long-range manner.

Jarid2 was recently found to be essential for the targeting of PRC2 Xi. Cdyl also associates with other factors such as MGA and the
to Xist RNA coated chromatin [40] (Fig. 1). In the case of PRC1, the H3K9 histone methyltransferase (HMTase) G9a. Although the pre-
canonical complex may be recruited through binding of the chrom- cise mechanisms are still not clear, it has been proposed that the
odomain region of the Cbx7 protein to H3K27me3 enriched chro- combination of H3K9me2 and H3K27me3 on the Xi help to recruit
matin; while the non-canonical (Rybp cofactor containing) PRC1 Cdyl, which in turn facilitates propagation of the H3K9me2 mark
complex appears to be Xist RNA dependent but H3K27me3 inde- by anchoring the G9a HMTase [43].
pendent [41]. Intriguingly, both PRC2 and PRC1 complexes are The chromatin changes so far described do not appear to be
enriched on the Xi early on in differentiation but become depleted sufcient for the initiation of gene silencing. For example, the dis-
at later stages of differentiation, suggesting that their association ruption of PRC2 and PRC1 components does not impede the onset
with the Xi is linked more to early than to late maintenance events of XCI and Xist RNA lacking the 50 conserved region of the transcript
[37,42]. The Cdyl protein, another chromodomain protein and tran- (known as the A repeats) can still induce these chromatin
scriptional corepressor, was recently identied as a new Xi partner changes and yet does not induce gene silencing [4345]. Thus
and potential reader of the H3K9me2 and H3K27me3 marks [43]. repression of gene transcription that is triggered by Xist repeat A
Cdyl relies on H3K9me2 for its general association to chromatin must involve different pathways, for example through interaction
in vivo, but requires both the presence of H3K9me2 and with proteins such as SATB1 and other factors [46]. The A-repeat
H3K27me3 for its differentiation-dependent enrichment on the region of Xist has also been proposed to play a role in interacting
R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522 2517

with PRC complexes, in the context of an independent transcript, giant cells (TGC) that are derived from this extraembryonic lineage,
RepA [47,48]. However, the exact role of RepA remains unclear high rates of escape from XCI are observed for some, but not all,
and the A-repeat region of Xist is clearly dispensable for Polycomb X-linked genes [56]. The current hypothesis is that there is a need
factor recruitment to the Xi, whereas another Xist region contain- for longer-term, more stable silencing in embryonic lineages as
ing the conserved B and F repeats was recently identied as being these give rise to the embryo-proper, where cellular memory must
critical for inducing PRC2 recruitment, via Jarid2 [40]. The multiple be maintained throughout the lifetime of the animal. On the other
roles of Xist RNA in X inactivation are still poorly understood and hand, there is only a short-term requirement for XCI in extra-
seem to be at several levels, involving chromatin, nuclear compart- embryonic tissues which are dispensable after birth. In particular,
mentalization, as well as chromosomal organization. Although the DNA (cytosine) methylation of X-linked CpG islands appears to
formation of a silent nuclear compartment by Xist RNA and the loss be present in embryonic but not extra-embryonic lineages in
of euchromatic marks correlate well with the onset of transcrip- eutherians mammals and its absence correlates with reactivation
tional inactivity [44], whether these changes are cause or conse- [57]. Indeed, epigenetic marks involved in maintaining the inactive
quence of XCI remains to be seen. state appear to vary considerably in both embryonic and extraem-
In addition to the recruitment of protein complexes such as bryonic lineages [58]. In TGCs, the Xi has an highly unusual chro-
PRC2, PRC1, as well as Cdyl and its partners, some further changes matin content, not only lacking DNA methylation at CpG islands
occur at later stages during the onset of XCI in differentiating ESC. but presenting both heterochromatic marks such as H3K27me3
These include enrichment of histone variant macroH2A [49] which and euchromatic marks such as histone H4 acetylation and H3K4
has been shown to interfere with transcription factor afnity and methylation. Moreover, Xist RNA does not form an overt silent
SWI-SNF nucleosome-remodeling in vitro [64,65]; the Trithorax nuclear compartment and reactivation of Xi-linked has been
group protein Ash2L [45]; and the scaffold factor SAF-A [50], which observed [56]. This may be partly due to the fact the trophecto-
is thought to participate in restricting or recruiting Xist RNA to the derm cells undergo endoreplication resulting in an unusual
inactive X. Most X-linked genes on the Xi also acquire DNA meth- chromosome organization and possibly less stable propagation of
ylation at their CpG islands, with different genes having rather dif- the silent chromatin state. Nevertheless, although the Xi shows a
ferent kinetics of hypermethylation [51]. The mechanism through substantial degree of gene reactivation in these cells it remains
which DNA methylation is recruited to X-linked genes remains globally silent. The presence of PRC2 and H3K27me3 on the Xi
unclear, although the Smchd1 and Dnmt1 DNA methyltransferase may be sufcient to maintain silence in this context. Indeed mouse
proteins have been proposed to participate [52,53] (Fig. 1). Smchd1 embryos mutant for the PRC2 component, Eed, show much higher
was originally identied in a genetic screen for epigenetic modi- reactivation rates of an X-linked GFP transgene specically in the
ers, as a factor involved in the maintenance of X inactivation and trophectoderm [59]. In the same mutants, no impact was found
the hypermethylation of CpG islands associated with the inactive in the epiblast, presumably due to the presence of other epigenetic
X [51,52]. Smchd1 has also been shown to participate in the com- factors, including PRC1 complexes and DNA methylation. Although
paction of the Xi in somatic human cells, through an interaction a direct role of PRC1 complex proteins in the maintenance of X
with HBiX1 [66]. These two proteins act in a PRC2-independent inactivity has not so far been demonstrated, two different com-
manner. plexes have been found associated with the Xi [41]. The timing
The inactive X chromosome also displays a shift to asynchro- of recruitment of PRC1 complexes to the Xi during the same time
nous replication timing during development. Asynchronous window as PRC2 in developing embryos [36,37,60] suggests that
replication is in fact one of the most conserved features of the inac- this complex is likely involved in the maintenance of inactivity.
tive X (see review [22]) and may provide a temporal segregation This is potentially by SWI-SNF complexes inhibition, blocking tran-
that minimizes the exposure of the inactive X to transcription fac- scriptional activity or by recruiting silencing components, as
tors, thus improving the maintenance of transcriptional silencing. described in other systems [6163].
This change in replication timing of the Xi occurs downstream of The extent that different epigenetic marks participate in the
chromatin changes such as loss of euchromatic marks and PRC maintenance of the inactive state has been addressed in experi-
recruitment [31]. It is still not known whether asynchronous ments aiming to prevent or reverse them. Studies have disrupted
replication has a role in XCI, or is simply a consequence of the several marks on the Xi simultaneously such as DNA methylation
heterochromatinisation of the Xi. Thus several independent path- (by 5-azacytidine treatment or in DNA methyltransferase mutant
ways seem to participate in the heterochromatinisation of the Xi. cells, Dnmt1/), histone hypoacetylation (by pan-histone deace-
Establishing the mechanisms of recruitment and action and the tylase inhibitor treatment, TSA) and Xist RNA (by a conditional
possible links between them represents an important challenge KO) can lead to a signicant increase of Xi-linked genes expres-
for the future. sion [54]. However disruption of each of these marks alone has lit-
tle effect. This clearly demonstrates that several epigenetic
3. Developmental plasticity of the inactive state and synergy of modications lock in the inactive state of the Xi. Nevertheless,
epigenetic marks in the maintenance of X-chromosome given the relatively low reactivation frequencies even after the
inactivity combined removal of these three marks, other epigenetic players
are likely to be involved in the maintenance of silencing. Indeed,
In somatic cells, the inactive X chromosome is in a highly stable so far, full reactivation of the Xi has never been achieved experi-
state of transcriptional silence, with sporadic reactivation of mentally other than via fusion with ES cells, which have the repro-
X-linked genes estimated to occur at frequencies of less than gramming potential of the inner cell mass [20,37], or by nuclear
108 [54]. However, during mouse development different lineages transfer where the reprogramming potential of the oocyte is
display rather different degrees of X-linked gene silencing, and this involved [67,68], or through the expression of the Yamanaka cock-
may be associated with slightly different epigenetic signatures. tail pluripotency factors (Oct4, Klf4, c-Myc and Sox2) that can
During early development, the paternal X becomes associated with drive somatic cells into an induced pluripotent state resembling
Xist RNA, macroH2A, PRC2 and the histone modication it deposits, ES cells [67,6971]. The reactivation of the Xi is thought to be
H3K27me3. At the blastocyst stage, these marks are reversed in the one of the later events during reprogramming, presumably due
inner cell mass where the inactive paternal X (Xp) is reactivated to the need to reverse several layers of epigenetic marking, some
[20,55]. However these marks are maintained on the inactive Xp of which may require active mechanisms (specic transcription
in extra-embryonic cells of the trophoblast lineage. In trophoblast factors or chromatin enzymatic activities) and others may require
2518 R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522

passive DNA replication to enable gradual removal. Further studies accurately quantifying allelic expression of X-linked genes. Never-
on the key players involved in the X-chromosome reactivation pro- theless, comparative analysis of CpG island DNA methylation has
cess during reprogramming, both during development (in the ICM been successfully used to determine indirectly the XCI status of
and in the germ line) and also experimentally (via nuclear transfer X-linked genes between different tissues, supporting the idea of
or IPS formation) should provide insights into the nature and substantial tissue-specic escape from XCI [84]. There is also
extent of the maintenance mechanisms that are at play in different evidence for human individual-specic genes that escape from X
cellular states and for different parts of the X chromosome (for inactivation. TIMP1 is good example of this phenomenon where it
review [5]). has been shown that H3 acetylation status correlates well with
females in which escape of this gene is found [85].
4. Variability in X-inactivation status and escape from XCI Apart from a small number of genes for which escape from XCI
appears to be evolutionarily conserved (such as KDM5C/JARID1C),
In eutherian mammals, such as humans and mice, the majority for most escapees on the human X, escape from XCI is not neces-
of X-linked genes are subject to transcriptional repression during sarily consistent between individuals or between tissues or even
XCI, although a variable subset of X-linked genes can escape silenc- between cells within the same individual. Nevertheless, the exis-
ing (escapees) and are bi-allelically expressed showing higher tence of escape from X inactivation, particularly in human remains
expression in females when compared to males [7275]. Only Xist largely unexplained [78]. Escape genes may play essential roles for
appears to be expressed exclusively from inactive X in female normal development. Indeed, Turner syndrome is very deleterious
somatic cells. Two main groups of escapees are present on the X to patient with a single X chromosome, perhaps due to haploinsuf-
chromosome. One group comprises escapees that lie within the ciency of escape genes, even though XO mice are largely not
pseudo autosomal region (PAR) and have an exact homolog on affected [8688].
the Y chromosome, and show equal expression levels between A comprehensive study of X-linked allelic expression in hun-
the sexes [73]. A second group consists of genes that are located dreds of human lymphoblastoid cell lines has revealed that 5% of
outside the PARs. These include genes such as Kdm5c (Smcx or Jar- X-linked genes show increased expression level in females when
id1c) and Utx, which have Y-chromosomal homologs lying outside compared to male. Based on this in vitro system, it seems that only
of the PAR and presumably represent evolutionary remnants from a few escape genes have a role in compensating sex biases even
the proto-sex chromosomes. Some other escapees have no obvious though this 5% could have a strong impact when they are lost
Y-linked homologs. The actual degree of escape from XCI for such [83]. A recent in vivo study using RNA sequencing done on post-
genes can vary considerably and depend on the tissue or the spe- mortem human brain tissues revealed that X-linked genes show
cies [74,76,77]. Such escapees usually display a lower level of gene expression dimorphisms. Sex-biased gene expression was
expression from the inactive X compared to the active X, thus lead- widespread in terms of gene number, chromosomes and range of
ing to female expression that is less than twice the dose compared brain regions involved. However, the percent of X- and Y-linked
to males [78]. The lower expression may be due to the gene only genes involved in the sex-bias seem highly related to particular
being expressed from the Xi in a proportion of cells and/or to lower regions of the brain [89]. For a more comprehensive view of the
transcription levels from the Xi. Allele-specic analyses of genes biological roles of genes that escape X inactivation in mammals,
that escape X inactivation reveal that they occupy an unique chro- and particularly in the context of human health and disease, more
matin conguration, with no/low enrichment of heterochromatin- primary tissue-specic and developmental analyses of XCI status
associated marks and no coating by Xist RNA [27,29,35,74,76]. RNA will be required. Further investigation of the DNA sequence con-
FISH studies have shown that escapees tend to be located outside text, chromatin environment, nuclear organization and chromo-
the Xist RNA coated territory [28,44]. More recently, chromosome some conformation, as well as developmental regulation will be
conformation capture analyses have revealed that Xi escapees tend needed to assess the mechanisms underlying escape from XCI.
to interact with each other preferentially in the nucleus [29]. The More detailed investigation of some of the known factors that
ability to escape may require a specic genomic context, as it has ensure the maintenance of Xi gene silencing, for example Smchd1
been shown that Kdm5c gene has a capacity to escape from which results in partial escape from XCI when deleted in the
XCI that is independent of its location on the inactive X chromo- mouse [93], will also be needed to understand the molecular basis
some [79]. This raises the question of how genes that are subject for developmental and tissue-specic escape from XCI. Finally,
to XCI, can resist the inuence of nearby escapees. Recent studies there are some examples of X-linked gene reactivation from the
suggested that regions of the X chromosome that are subject to Xi that have been associated with the aging, for example as shown
XCI appear to be protected from the spreading of transcriptionally for the Otc gene in mice [94]. The mechanism for this age-related
active escapee domains thanks to CTCF insulator elements escape is unclear although it may be due to epigenetic "erosion"
[80,81] and the presence of young active LINE-1 elements has also due to inefcient maintenance of chromatin states over extended
been speculated to be involved in facilitating XCI of genes in cell division or in adult stem cells. Although loss of the Barr body
regions that are otherwise prone to escape [28]. in ageing females has been reported, the degree to which
Surprisingly, many more escapees have been found in humans age-related epigenetic relaxation of the Xi occurs in humans and
when compared to mice, with an estimation of up to 15% of mice has not so far been investigated systematically.
X-linked genes escaping XCI to some extent in humans. An addi-
tional 10% of human X-linked genes show variable degree of 5. Unstable X-chromosome inactivation in cancer?
escape, based on investigation in cell lines [82,83]. Many of these
human escapee genes lie on the more recently added short arm As described above, Xi reactivation is observed in a develop-
of the X chromosome. Indeed, it has been hypothesized that their mental context in mouse extra-embryonic lineages and in the germ
incomplete silencing could be due to a barrier effect caused by line, and escape from XCI is observed for a subset of X-linked genes
the centromeric heterochromatin that separates the XIC on the in somatic cells particularly in humans. However, the Xi is globally
long arm (Xq13) from the short arm where most escapees are stably silent in somatic tissues suggesting that dosage compensa-
located (for review [78]). It should be noted however, that to date, tion is important, although the impact that failure to maintain Xi
only a few of these human Xi escapees have been conrmed in vivo inactivity in adult tissues would have, has not so far been assessed
given the difculties in obtaining human tissue samples and in in any systematic way. The synergy and partial redundancy of
R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522 2519

heterochromatin-associated marks that cooperatively maintain the [99101]. In some cases, XIST RNA mislocalisation and sporadic
integrity of heterochromatic inactive X were discussed above. The Xi reactivation has been observed [102,103]. For example, one
loss of Xist from the Xi in terminally differentiated cells does not study on an ovarian cancer cell line, showed a disruption of XIST
lead to global reactivation of the inactive X, as genes remain expression and potential reactivation of the MPP1 (p55) gene
repressed presumably via the above-mentioned synergistic epige- [97]. In the context of breast cancer, bi-allelic expression of a single
netic modications [54]. Thus although Xist is required for initia- X-linked gene, VBP1, associated with promoter DNA hypomethyla-
tion of X inactivation, it was thought not to be necessary for the tion was reported in one primary breast tumor sample [96]. How-
maintenance of transcriptional silencing on the Xi up until recently ever, without global analyses of expression status and chromatin
[90,91]. However, a recent study showed that the conditional structure of the inactive X chromosome, it is difcult to assess
knock-out of Xist in the hematopoietic compartment of the mouse, the precise extent of Xi instability in a cancer context. On the other
led to an aggressive form of hematologic cancer in females [92]. hand, Barr body disruption can be due to physical loss of the Xi in
Although allele-specic analyses were not performed in this study female cancers [103,104]. Indeed, recent evidences show that the
and thus Xi reactivation could not be demonstrated directly, the inactive X chromosome is genetically unstable in cancer as this
authors hypothesized that X-linked gene over-expression could study reporting an higher mutations rate on the inactive X com-
be the result of X inactivation relaxation in tumor cells and some- pare to rest of the genome [105]. The consequence of both models
how trigger cancer. would result with double dose of some or all X-linked genes. In
In humans, there is still only limited evidence available for the early embryos, the presence of two active X chromosomes is
status of the Xi in cancer to date, even though it was described ultimately lethal [12]. However in somatic cells the increased dose
more than 50 years ago that the Barr body is frequently lost in of a subset of genes on the X chromosome could potentially
breast cancers [2,95] (Fig. 2), leading to the proposal that Xi provide a selective advantage and promote cancer development,
reactivation may be a common event in some cancers. More recent considering that even a rare cell expressing a gene with a prolifer-
studies suggest that the disappearance of the Barr body is ative advantage could contribute to cancer progression. Future
sometimes associated with over-expression of X-linked genes, sug- studies on the causes or consequences of Xi reactivation in differ-
gesting a potential role of the X chromosome in cancer progression ent situations, during normal development and in cancer, will be
[96]. In some cases, the tumors lacking an inactive X chromosome, needed. Indeed, the emerging role of aberrant gene dosage in dis-
also present a duplication of the active X chromosome [9698]. eases, whether of the X chromosome or for autosomes, brings with
Thus, at least two mechanisms could explain the loss of Barr body it the possible application of drugs that impact on epigenetic reg-
in cancer cells (Fig. 2). On one hand, epigenetic instability of Xi may ulators in potential therapeutic strategies [106108].
occur, with decondensation of heterochromatic Barr body, leading A direct role for BRCA1, a tumor suppressor protein that is fre-
to Xi-linked gene reactivation. Although this mechanism has been quently mutated in familial cases of breast and ovarian cancer, was
frequently hypothesized, so far no evidence supports a model of proposed some years ago to explain loss of the Xi [109]. This study
global Xi reactivation and heterochromatin loss in cancer proposed a direct action of BRCA1 on XIST RNA coating of the Xi

Fig. 2. Genetic and epigenetic instability of the Xi in cancer. Schematic view of genetic and epigenetic alterations that can affect the X chromosome in breast cancer cells. The
left panel lists the different genetic situations that have been be observed concerning the X chromosomes in cancer cells i.e. Xi loss and Xa reduplication (for example [2]); Xi
hypermutation [105]; and no Xi loss (for example [104]). On the right, we represent potential epigenetic instability that has been hypothesized to affect the Xi in some cancer
cell. So far, few studies have evaluated X-linked gene reactivation in breast cancer (for example [96]). Further studies are needed to explore to what extent epigenetic instabily
is present on the inactive X chromosome in cancer cells and what could be their impact in carcinogenesis.
2520 R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522

and suggested that in BRCA1 mutated tumors, the Xi might be epi- [6] Pollex, T. and Heard, E. (2012) Recent advances in X-chromosome
inactivation research. Curr. Opin. Cell Biol. 24, 825832.
genetically relaxed. However subsequent studies [102,104]
[7] Avner, P. and Heard, E. (2001) X-chromosome inactivation: counting, choice
revealed that cells of BRCA1-mutated primary breast tumors can and initiation. Nat. Rev. Genet. 2, 5967.
contain one or several XIST RNA coated chromosomes. Neverthe- [8] Brown, C.J., Hendrich, B.D., Rupert, J.L., Lafreniere, R.G., Xing, Y., Lawrence, J.
less, this does not invalidate a role for BRCA1 in XIST expression and Willard, H.F. (1992) The human XIST gene: analysis of a 17 kb inactive
X-specic RNA that contains conserved repeats and is highly localized within
[102], as, among its many roles it may act as a transcription the nucleus. Cell 71, 527542.
regulator [110]. Another chromatin regulator frequently over- [9] Brown, S.D. (1991) XIST and the mapping of the X chromosome inactivation
expressed in cancer, Aurora B Kinase (AURKB), has also been centre. BioEssays 13, 607612.
[10] Grant, J., Mahadevaiah, S.K., Khil, P., Sangrithi, M.N., Royo, H., Duckworth, J.,
proposed to regulate the association of XIST to the Xi [111,112]. McCarrey, J.R., VandeBerg, J.L., Renfree, M.B., Taylor, W., Elgar, G., Camerini-
However, what the exact consequences are on Xi status are not Otero, R.D., Gilchrist, M.J. and Turner, J.M. (2012) Rsx is a metatherian RNA
clear. Thus, the disappearance of the Barr body in aggressive breast with Xist-like properties in X-chromosome inactivation. Nature 487,
254258.
tumors noted more than 50 years ago, still remains a fascinating [11] Penny, G.D., Kay, G.F., Sheardown, S.A., Rastan, S. and Brockdorff, N. (1996)
but poorly understood phenomenon. Further investigation will be Requirement for Xist in X chromosome inactivation. Nature 379, 131137.
required to elucidate the nature and extent of the inactive X [12] Marahrens, Y., Panning, B., Dausman, J., Strauss, W. and Jaenisch, R. (1997)
Xist-decient mice are defective in dosage compensation but not
chromosomes genetic and epigenetic instability in cancer, with spermatogenesis. Genes Dev. 11, 156166.
obvious clinical interest. [13] Wutz, A. and Jaenisch, R. (2000) A shift from reversible to irreversible X
inactivation is triggered during ES cell differentiation. Mol. Cell 5, 695705.
[14] Sado, T., Wang, Z., Sasaki, H. and Li, E. (2001) Regulation of imprinted
6. Conclusion X-chromosome inactivation in mice by Tsix. Development 128, 12751286.
[15] Gontan, C., Achame, E.M., Demmers, J., Barakat, T.S., Rentmeester, E., van
The inactive X chromosome provides a remarkable example of IJcken, W., Grootegoed, J.A. and Gribnau, J. (2012) RNF12 initiates
X-chromosome inactivation by targeting REX1 for degradation. Nature 485,
chromosome-wide gene repression that reveals the diversity of 386390.
processes that can contribute to the formation of facultative [16] Barakat, T.S., Gunhanlar, N., Pardo, C.G., Achame, E.M., Ghazvini, M., Boers, R.,
heterochromatin. XCI also highlights the complex issue of gene Kenter, A., Rentmeester, E., Grootegoed, J.A. and Gribnau, J. (2011) RNF12
activates Xist and is essential for X chromosome inactivation. PLoS Genet. 7,
dosage. The repeated inactivation and reactivation of the X chro- e1002001.
mosome during development illustrates the importance of a ne [17] Nora, E.P., Lajoie, B.R., Schulz, E.G., Giorgetti, L., Okamoto, I., Servant, N.,
regulation of all the mechanisms involved. Recent studies have Piolot, T., van Berkum, N.L., Meisig, J., Sedat, J., Gribnau, J., Barillot, E.,
Bluthgen, N., Dekker, J. and Heard, E. (2012) Spatial partitioning of the
revealed that a combination of chromatin factors, chromosome
regulatory landscape of the X-inactivation centre. Nature 485, 381385.
conformation and nuclear compartmentalization together ensure [18] Nora, E.P., Dekker, J. and Heard, E. (2013) Segmental folding of chromosomes:
maintenance of the inactive state. The mechanism that triggers a basis for structural and regulatory chromosomal neighborhoods? BioEssays
35, 818828.
gene silencing in the rst place, via Xist RNA remains tantalizingly
[19] Giorgetti, L., Galupa, R., Nora, E.P., Piolot, T., Lam, F., Dekker, J., Tiana, G. and
obscure. Once XCI is established, silencing appears to be rather sta- Heard, E. (2014) Predictive polymer modeling reveals coupled uctuations in
ble in terminally differentiated cells, although XCI is less complete chromosome conformation and transcription. Cell 157, 950963.
in humans than in mice and some genes can escape this silencing [20] Okamoto, I., Otte, A.P., Allis, C.D., Reinberg, D. and Heard, E. (2004) Epigenetic
dynamics of imprinted X inactivation during early mouse development.
process, either for a purpose or accidentally. Increasing evidence Science 303, 644649.
suggests that silencing efciency of X-linked genes is dependent [21] Schulz, E.G., Meisig, J., Nakamura, T., Okamoto, I., Sieber, A., Picard, C.,
on tissue types and individuals and points to a complex interplay Borensztein, M., Saitou, M., Bluthgen, N. and Heard, E. (2014) The two active
X chromosomes in female ESCs block exit from the pluripotent state by
between genetic variation and epigenetic status. The growing real- modulating the ESC signaling network. Cell Stem Cell 14, 203216.
ization that both genetic and epigenetic defects may affect the [22] Escamilla-Del-Arenal, M., da Rocha, S.T. and Heard, E. (2011) Evolutionary
inactive X chromosome in tumors opens up new avenues of diversity and developmental regulation of X-chromosome inactivation. Hum.
Genet. 130, 307327.
research to understand how Xi epigenetic instability in cancer [23] Okamoto, I. and Heard, E. (2009) Lessons from comparative analysis of
arises and whether this instability can actually contribute to cancer X-chromosome inactivation in mammals. Chromosome Res. 17, 659669.
progression and would thus be of therapeutic interest. [24] Okamoto, I., Patrat, C., Thepot, D., Peynot, N., Fauque, P., Daniel, N.,
Diabangouaya, P., Wolf, J.P., Renard, J.P., Duranthon, V. and Heard, E. (2011)
Eutherian mammals use diverse strategies to initiate X-chromosome
Acknowledgments inactivation during development. Nature 472, 370374.
[25] Mugford, J.W., Yee, D. and Magnuson, T. (2012) Failure of extra-embryonic
progenitor maintenance in the absence of dosage compensation.
We would like to thank Simao Teixeira da Rocha for critical Development 139, 21302138.
reading of the manuscript. Support is gratefully acknowledged [26] Chow, J.C., Ciaudo, C., Fazzari, M.J., Mise, N., Servant, N., Glass, J.L., Attreed, M.,
from lAssociation pour la Recherche sur le Cancer (ARC) Avner, P., Wutz, A., Barillot, E., Greally, J.M., Voinnet, O. and Heard, E. (2010)
LINE-1 activity in facultative heterochromatin formation during X
(post-doc fellowship to R.C.); FRM (Equipe FRM to E.H); Equipe chromosome inactivation. Cell 141, 956969.
labellise La Ligue Contre Le Cancer (Equipe Labllis to E.H) [27] Engreitz, J.M., Pandya-Jones, A., McDonel, P., Shishkin, A., Sirokman, K., Surka,
and EU FP7 MODHEP EU grant no. 259743 (to E.H.) and Labex DEEP C., Kadri, S., Xing, J., Goren, A., Lander, E.S., Plath, K. and Guttman, M. (2013)
The Xist lncRNA exploits three-dimensional genome architecture to spread
(ANR-11-LBX-0044) part of the IDEX Idex PSL (ANR-10-IDEX-0001-
across the X chromosome. Science 341, 1237973.
02 PSL). [28] Simon, M.D., Pinter, S.F., Fang, R., Sarma, K., Rutenberg-Schoenberg, M.,
Bowman, S.K., Kesner, B.A., Maier, V.K., Kingston, R.E. and Lee, J.T. (2013)
High-resolution Xist binding maps reveal two-step spreading during
References X-chromosome inactivation. Nature 504, 465469.
[29] Splinter, E., de Wit, E., Nora, E.P., Klous, P., van de Werken, H.J., Zhu, Y., Kaaij,
[1] Lyon, M.F. (1961) Gene action in the X-chromosome of the mouse (Mus L.J., van Ijcken, W., Gribnau, J., Heard, E. and de Laat, W. (2011) The inactive X
musculus L.). Nature 190, 372373. chromosome adopts a unique three-dimensional conformation that is
[2] Barr, M.L. and Moore, K.L. (1957) Chromosomes, sex chromatin, and cancer. dependent on Xist RNA. Genes Dev. 25, 13711383.
Proc. Can. Cancer Conf. 2, 316. [30] Cerase, A., Smeets, D., Tang, Y.A., Gdula, M., Kraus, F., Spivakov, M., Moindrot,
[3] Perry, M. (1972) Evaluation of breast tumour sex chromatin (Barr body) as an B., Leleu, M., Tattermusch, A., Demmerle, J., Nesterova, T.B., Green, C., Otte,
index of survival and response to pituitary ablation. Br. J. Surg. 59, 731734. A.P., Schermelleh, L. and Brockdorff, N. (2014) Spatial separation of Xist RNA
[4] Carone, D.M. and Lawrence, J.B. (2013) Heterochromatin instability in cancer: and polycomb proteins revealed by superresolution microscopy. Proc. Natl.
from the Barr body to satellites and the nuclear periphery. Semin. Cancer Biol. Acad. Sci. U.S.A. 111, 22352240.
23, 99108. [31] Chaumeil, J., Okamoto, I., Guggiari, M. and Heard, E. (2002) Integrated
[5] Ohhata, T. and Wutz, A. (2013) Reactivation of the inactive X chromosome in kinetics of X chromosome inactivation in differentiating embryonic stem
development and reprogramming. Cell. Mol. Life Sci. 70, 24432461. cells. Cytogenet. Genome Res. 99, 7584.
R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522 2521

[32] Heard, E., Rougeulle, C., Arnaud, D., Avner, P., Allis, C.D. and Spector, D.L. [56] Corbel, C., Diabangouaya, P., Gendrel, A.V., Chow, J.C. and Heard, E. (2013)
(2001) Methylation of histone H3 at Lys-9 is an early mark on the X Unusual chromatin status and organization of the inactive X chromosome in
chromosome during X inactivation. Cell 107, 727738. murine trophoblast giant cells. Development 140, 861872.
[33] Keohane, A.M., ONeill, L.P., Belyaev, N.D., Lavender, J.S. and Turner, B.M. [57] Kratzer, P.G., Chapman, V.M., Lambert, H., Evans, R.E. and Liskay, R.M. (1983)
(1996) X-Inactivation and histone H4 acetylation in embryonic stem cells. Differences in the DNA of the inactive X chromosomes of fetal and
Dev. Biol. 180, 618630. extraembryonic tissues of mice. Cell 33, 3742.
[34] Chadwick, B.P. and Willard, H.F. (2004) Multiple spatially distinct types of [58] Kunath, T., Arnaud, D., Uy, G.D., Okamoto, I., Chureau, C., Yamanaka, Y., Heard,
facultative heterochromatin on the human inactive X chromosome. Proc. E., Gardner, R.L., Avner, P. and Rossant, J. (2005) Imprinted X-inactivation in
Natl. Acad. Sci. U.S.A. 101, 1745017455. extra-embryonic endoderm cell lines from mouse blastocysts. Development
[35] Marks, H., Chow, J.C., Denissov, S., Francoijs, K.J., Brockdorff, N., Heard, E. and 132, 16491661.
Stunnenberg, H.G. (2009) High-resolution analysis of epigenetic changes [59] Mak, W., Baxter, J., Silva, J., Newall, A.E., Otte, A.P. and Brockdorff, N. (2002)
associated with X inactivation. Genome Res. 19, 13611373. Mitotically stable association of polycomb group proteins eed and enx1 with
[36] de Napoles, M., Mermoud, J.E., Wakao, R., Tang, Y.A., Endoh, M., Appanah, R., the inactive X chromosome in trophoblast stem cells. Curr. Biol. 12,
Nesterova, T.B., Silva, J., Otte, A.P., Vidal, M., Koseki, H. and Brockdorff, N. 10161020.
(2004) Polycomb group proteins Ring1A/B link ubiquitylation of histone H2A [60] Bernstein, E., Duncan, E.M., Masui, O., Gil, J., Heard, E. and Allis, C.D. (2006)
to heritable gene silencing and X inactivation. Dev. Cell 7, 663676. Mouse polycomb proteins bind differentially to methylated histone H3 and
[37] Plath, K., Talbot, D., Hamer, K.M., Otte, A.P., Yang, T.P., Jaenisch, R. and RNA and are enriched in facultative heterochromatin. Mol. Cell. Biol. 26,
Panning, B. (2004) Developmentally regulated alterations in Polycomb 25602569.
repressive complex 1 proteins on the inactive X chromosome. J. Cell Biol. [61] Simon, J.A. and Tamkun, J.W. (2002) Programming off and on states in
167, 10251035. chromatin: mechanisms of Polycomb and trithorax group complexes. Curr.
[38] Plath, K., Fang, J., Mlynarczyk-Evans, S.K., Cao, R., Worringer, K.A., Wang, H., Opin. Genet. Dev. 12, 210218.
de la Cruz, C.C., Otte, A.P., Panning, B. and Zhang, Y. (2003) Role of histone H3 [62] Otte, A.P. and Kwaks, T.H. (2003) Gene repression by Polycomb group protein
lysine 27 methylation in X inactivation. Science 300, 131135. complexes: a distinct complex for every occasion? Curr. Opin. Genet. Dev. 13,
[39] Silva, J., Mak, W., Zvetkova, I., Appanah, R., Nesterova, T.B., Webster, Z., Peters, 448454.
A.H., Jenuwein, T., Otte, A.P. and Brockdorff, N. (2003) Establishment of [63] Lund, A.H. and van Lohuizen, M. (2004) Polycomb complexes and silencing
histone h3 methylation on the inactive X chromosome requires transient mechanisms. Curr. Opin. Cell Biol. 16, 239246.
recruitment of Eed-Enx1 polycomb group complexes. Dev. Cell 4, 481495. [64] Gomez, M. and Brockdorff, N. (2004) Heterochromatin on the inactive X
[40] da Rocha, S.T., Boeva, V., Escamilla-Del-Arenal, M., Ancelin, K., Granier, C., chromosome delays replication timing without affecting origin usage. Proc.
Matias, N.R., Sanulli, S., Chow, J., Schulz, E., Picard, C., Kaneko, S., Helin, K., Natl. Acad. Sci. U.S.A. 101, 69236928.
Reinberg, D., Stewart, A.F., Wutz, A., Margueron, R. and Heard, E. (2014) Jarid2 [65] Costanzi, C., Stein, P., Worrad, D.M., Schultz, R.M. and Pehrson, J.R. (2000)
Is Implicated in the Initial Xist-Induced Targeting of PRC2 to the Inactive X Histone macroH2A1 is concentrated in the inactive X chromosome of female
Chromosome. Mol. Cell 53, 301316. preimplantation mouse embryos. Development 127, 22832289.
[41] Tavares, L., Dimitrova, E., Oxley, D., Webster, J., Poot, R., Demmers, J., [66] Nozawa, R.S., Nagao, K., Igami, K.T., Shibata, S., Shirai, N., Nozaki, N., Sado, T.,
Bezstarosti, K., Taylor, S., Ura, H., Koide, H., Wutz, A., Vidal, M., Elderkin, S. and Kimura, H. and Obuse, C. (2013) Human inactive X chromosome is
Brockdorff, N. (2012) RYBP-PRC1 complexes mediate H2A ubiquitylation at compacted through a PRC2-independent SMCHD1-HBiX1 pathway. Nat.
polycomb target sites independently of PRC2 and H3K27me3. Cell 148, Struct. Mol. Biol. 20, 566573.
664678. [67] Eggan, K., Akutsu, H., Hochedlinger, K., Rideout 3rd, W., Yanagimachi, R. and
[42] Kohlmaier, A., Savarese, F., Lachner, M., Martens, J., Jenuwein, T. and Wutz, A. Jaenisch, R. (2000) X-chromosome inactivation in cloned mouse embryos.
(2004) A chromosomal memory triggered by Xist regulates histone Science 290, 15781581.
methylation in X inactivation. PLoS Biol. 2, E171. [68] Pasque, V., Gillich, A., Garrett, N. and Gurdon, J.B. (2011) Histone variant
[43] Escamilla-Del-Arenal, M., da Rocha, S.T., Spruijt, C.G., Masui, O., Renaud, O., macroH2A confers resistance to nuclear reprogramming. EMBO J. 30,
Smits, A.H., Margueron, R., Vermeulen, M. and Heard, E. (2013) Cdyl, a new 23732387.
partner of the inactive X chromosome and potential reader of H3K27me3 and [69] Bao, S., Miyoshi, N., Okamoto, I., Jenuwein, T., Heard, E. and Azim Surani, M.
H3K9me2. Mol. Cell. Biol. 33, 50055020. (2005) Initiation of epigenetic reprogramming of the X chromosome in
[44] Chaumeil, J., Le Baccon, P., Wutz, A. and Heard, E. (2006) A novel role for Xist somatic nuclei transplanted to a mouse oocyte. EMBO Rep. 6, 748754.
RNA in the formation of a repressive nuclear compartment into which genes [70] Wang, W., Yang, J., Liu, H., Lu, D., Chen, X., Zenonos, Z., Campos, L.S., Rad, R.,
are recruited when silenced. Genes Dev. 20, 22232237. Guo, G., Zhang, S., Bradley, A. and Liu, P. (2011) Rapid and efcient
[45] Pullirsch, D., Hartel, R., Kishimoto, H., Leeb, M., Steiner, G. and Wutz, A. (2010) reprogramming of somatic cells to induced pluripotent stem cells by
The trithorax group protein Ash2l and Saf-A are recruited to the inactive X retinoic acid receptor gamma and liver receptor homolog 1. Proc. Natl.
chromosome at the onset of stable X inactivation. Development 137, Acad. Sci. U.S.A. 108, 1828318288.
935943. [71] Gillich, A., Bao, S., Grabole, N., Hayashi, K., Trotter, M.W., Pasque, V.,
[46] Agrelo, R., Souabni, A., Novatchkova, M., Haslinger, C., Leeb, M., Komnenovic, Magnusdottir, E. and Surani, M.A. (2012) Epiblast stem cell-based system
V., Kishimoto, H., Gresh, L., Kohwi-Shigematsu, T., Kenner, L. and Wutz, A. reveals reprogramming synergy of germline factors. Cell Stem Cell 10,
(2009) SATB1 denes the developmental context for gene silencing by Xist in 425439.
lymphoma and embryonic cells. Dev. Cell 16, 507516. [72] Talebizadeh, Z., Simon, S.D. and Butler, M.G. (2006) X chromosome gene
[47] Maenner, S., Blaud, M., Fouillen, L., Savoye, A., Marchand, V., Dubois, A., expression in human tissues: male and female comparisons. Genomics 88,
Sanglier-Cianferani, S., Van Dorsselaer, A., Clerc, P., Avner, P., Visvikis, A. and 675681.
Branlant, C. (2010) 2-D structure of the A region of Xist RNA and its [73] Johnston, C.M., Lovell, F.L., Leongamornlert, D.A., Stranger, B.E.,
implication for PRC2 association. PLoS Biol. 8, e1000276. Dermitzakis, E.T. and Ross, M.T. (2008) Large-scale population study of
[48] Zhao, J., Sun, B.K., Erwin, J.A., Song, J.J. and Lee, J.T. (2008) Polycomb proteins human cell lines indicates that dosage compensation is virtually complete.
targeted by a short repeat RNA to the mouse X chromosome. Science 322, PLoS Genet. 4, e9.
750756. [74] Yang, F., Babak, T., Shendure, J. and Disteche, C.M. (2010) Global survey of
[49] Costanzi, C. and Pehrson, J.R. (1998) Histone macroH2A1 is concentrated in escape from X inactivation by RNA-sequencing in mouse. Genome Res. 20,
the inactive X chromosome of female mammals. Nature 393, 599601. 614622.
[50] Helbig, R. and Fackelmayer, F.O. (2003) Scaffold attachment factor A (SAF-A) [75] Sudbrak, R., Wieczorek, G., Nuber, U.A., Mann, W., Kirchner, R., Erdogan, F.,
is concentrated in inactive X chromosome territories through its RGG Brown, C.J., Wohrle, D., Sterk, P., Kalscheuer, V.M., Berger, W., Lehrach, H. and
domain. Chromosoma 112, 173182. Ropers, H.H. (2001) X chromosome-specic cDNA arrays: identication of
[51] Gendrel, A.V., Tang, Y.A., Suzuki, M., Godwin, J., Nesterova, T.B., Greally, J.M., genes that escape from X-inactivation and other applications. Hum. Mol.
Heard, E. and Brockdorff, N. (2013) Epigenetic functions of smchd1 repress Genet. 10, 7783.
gene clusters on the inactive X chromosome and on autosomes. Mol. Cell. [76] Calabrese, J.M., Sun, W., Song, L., Mugford, J.W., Williams, L., Yee, D., Starmer,
Biol. 33, 31503165. J., Mieczkowski, P., Crawford, G.E. and Magnuson, T. (2012) Site-specic
[52] Blewitt, M.E., Gendrel, A.V., Pang, Z., Sparrow, D.B., Whitelaw, N., Craig, J.M., silencing of regulatory elements as a mechanism of X inactivation. Cell 151,
Apedaile, A., Hilton, D.J., Dunwoodie, S.L., Brockdorff, N., Kay, G.F. and 951963.
Whitelaw, E. (2008) SmcHD1, containing a structural-maintenance-of- [77] Wu, H., Luo, J., Yu, H., Rattner, A., Mo, A., Wang, Y., Smallwood, P.M., Erlanger,
chromosomes hinge domain, has a critical role in X inactivation. Nat. B., Wheelan, S.J. and Nathans, J. (2014) Cellular resolution maps of X
Genet. 40, 663669. chromosome inactivation: implications for neural development, function,
[53] Sado, T., Fenner, M.H., Tan, S.S., Tam, P., Shioda, T. and Li, E. (2000) X and disease. Neuron 81, 103119.
inactivation in the mouse embryo decient for Dnmt1: distinct effect of [78] Berletch, J.B., Yang, F., Xu, J., Carrel, L. and Disteche, C.M. (2011) Genes that
hypomethylation on imprinted and random X inactivation. Dev. Biol. 225, escape from X inactivation. Hum. Genet. 130, 237245.
294303. [79] Li, N. and Carrel, L. (2008) Escape from X chromosome inactivation is an
[54] Csankovszki, G., Nagy, A. and Jaenisch, R. (2001) Synergism of Xist RNA, DNA intrinsic property of the Jarid1c locus. Proc. Natl. Acad. Sci. U.S.A. 105,
methylation, and histone hypoacetylation in maintaining X chromosome 1705517060.
inactivation. J. Cell Biol. 153, 773784. [80] Filippova, G.N., Cheng, M.K., Moore, J.M., Truong, J.P., Hu, Y.J., Nguyen, D.K.,
[55] Mak, W., Nesterova, T.B., de Napoles, M., Appanah, R., Yamanaka, S., Otte, A.P. Tsuchiya, K.D. and Disteche, C.M. (2005) Boundaries between chromosomal
and Brockdorff, N. (2004) Reactivation of the paternal X chromosome in early domains of X inactivation and escape bind CTCF and lack CpG methylation
mouse embryos. Science 303, 666669. during early development. Dev. Cell 8, 3142.
2522 R. Chalign, E. Heard / FEBS Letters 588 (2014) 25142522

[81] Horvath, L.M., Li, N. and Carrel, L. (2013) Deletion of an X-inactivation Loss of the inactive X chromosome and replication of the active X in BRCA1-
boundary disrupts adjacent gene silencing. PLoS Genet. 9, e1003952. defective and wild-type breast cancer cells. Cancer Res. 65, 21392146.
[82] Carrel, L. and Willard, H.F. (2005) X-inactivation prole reveals extensive [99] Spatz, A., Borg, C. and Feunteun, J. (2004) X-chromosome genetics and human
variability in X-linked gene expression in females. Nature 434, 400404. cancer. Nat. Rev. Cancer 4, 617629.
[83] Cotton, A.M., Ge, B., Light, N., Adoue, V., Pastinen, T. and Brown, C.J. (2013) [100] Pageau, G.J., Hall, L.L., Ganesan, S., Livingston, D.M. and Lawrence, J.B. (2007)
Analysis of expressed SNPs identies variable extents of expression from the The disappearing Barr body in breast and ovarian cancers. Nat. Rev. Cancer 7,
human inactive X chromosome. Genome Biol. 14, R122. 628633.
[84] Cotton, A.M., Lam, L., Afeck, J.G., Wilson, I.M., Penaherrera, M.S., McFadden, [101] Agrelo, R. and Wutz, A. (2010) ConteXt of change X inactivation and
D.E., Kobor, M.S., Lam, W.L., Robinson, W.P. and Brown, C.J. (2011) disease. EMBO Mol. Med. 2, 615.
Chromosome-wide DNA methylation analysis predicts human tissue- [102] Pageau, G.J., Hall, L.L. and Lawrence, J.B. (2007) BRCA1 does not paint the
specic X inactivation. Hum. Genet. 130, 187201. inactive X to localize XIST RNA but may contribute to broad changes in
[85] Anderson, C.L. and Brown, C.J. (2005) Epigenetic predisposition to expression cancer that impact XIST and Xi heterochromatin. J. Cell. Biochem. 100,
of TIMP1 from the human inactive X chromosome. BMC Genet. 6, 48. 835850.
[86] Adler, D.A., Bressler, S.L., Chapman, V.M., Page, D.C. and Disteche, C.M. (1991) [103] Sirchia, S.M., Tabano, S., Monti, L., Recalcati, M.P., Gariboldi, M., Grati, F.R.,
Inactivation of the Zfx gene on the mouse X chromosome. Proc. Natl. Acad. Porta, G., Finelli, P., Radice, P. and Miozzo, M. (2009) Misbehaviour of XIST
Sci. U.S.A. 88, 45924595. RNA in breast cancer cells. PLoS One 4, e5559.
[87] Ashworth, A., Rastan, S., Lovell-Badge, R. and Kay, G. (1991) X-chromosome [104] Vincent-Salomon, A., Ganem-Elbaz, C., Manie, E., Raynal, V., Sastre-Garau, X.,
inactivation may explain the difference in viability of XO humans and mice. Stoppa-Lyonnet, D., Stern, M.H. and Heard, E. (2007) X inactive-specic
Nature 351, 406408. transcript RNA coating and genetic instability of the X chromosome in BRCA1
[88] Heard, E. and Turner, J. (2011) Function of the sex chromosomes in breast tumors. Cancer Res. 67, 51345140.
mammalian fertility. Cold Spring Harb. Perspect. Biol. 3, a002675. [105] Jager, N., Schlesner, M., Jones, D.T., Raffel, S., Mallm, J.P., Junge, K.M.,
[89] Trabzuni, D., Ramasamy, A., Imran, S., Walker, R., Smith, C., Weale, M.E., Weichenhan, D., Bauer, T., Ishaque, N., Kool, M., Northcott, P.A., Korshunov,
Hardy, J. and Ryten, M. (2013) Widespread sex differences in gene expression A., Drews, R.M., Koster, J., Versteeg, R., Richter, J., Hummel, M., Mack, S.C.,
and splicing in the adult human brain. Nat. Commun. 4, 2771. Taylor, M.D., Witt, H., Swartman, B., Schulte-Bockholt, D., Sultan, M., Yaspo,
[90] Hall, L.L., Clemson, C.M., Byron, M., Wydner, K. and Lawrence, J.B. (2002) M.L., Lehrach, H., Hutter, B., Brors, B., Wolf, S., Plass, C., Siebert, R., Trumpp, A.,
Unbalanced X;autosome translocations provide evidence for sequence Rippe, K., Lehmann, I., Lichter, P., Pster, S.M. and Eils, R. (2013)
specicity in the association of XIST RNA with chromatin. Hum. Mol. Hypermutation of the inactive X chromosome is a frequent event in
Genet. 11, 31573165. cancer. Cell 155, 567581.
[91] Brown, C.J. and Willard, H.F. (1994) The human X-inactivation centre is not [106] Gendrel, A.V., Attia, M., Chen, C.J., Diabangouaya, P., Servant, N., Barillot, E.
required for maintenance of X-chromosome inactivation. Nature 368, and Heard, E. (2014) Developmental dynamics and disease potential of
154156. random monoallelic gene expression. Dev. Cell 28, 366380.
[92] Yildirim, E., Kirby, J.E., Brown, D.E., Mercier, F.E., Sadreyev, R.I., Scadden, D.T. [107] Eckersley-Maslin, M.A., Thybert, D., Bergmann, J.H., Marioni, J.C., Flicek, P.
and Lee, J.T. (2013) Xist RNA is a potent suppressor of hematologic cancer in and Spector, D.L. (2014) Random monoallelic gene expression increases upon
mice. Cell 152, 727742. embryonic stem cell differentiation. Dev. Cell 28, 351365.
[93] Leong, H.S., Chen, K., Hu, Y., Lee, S., Corbin, J., Pakusch, M., Murphy, J.M., [108] Huang, H.S., Allen, J.A., Mabb, A.M., King, I.F., Miriyala, J., Taylor-Blake, B.,
Majewski, I.J., Smyth, G.K., Alexander, W.S., Hilton, D.J. and Blewitt, M.E. Sciaky, N., Dutton Jr., J.W., Lee, H.M., Chen, X., Jin, J., Bridges, A.S., Zylka, M.J.,
(2013) Epigenetic regulator Smchd1 functions as a tumor suppressor. Cancer Roth, B.L. and Philpot, B.D. (2012) Topoisomerase inhibitors unsilence the
Res. 73, 15911599. dormant allele of Ube3a in neurons. Nature 481, 185189.
[94] Wareham, K.A., Lyon, M.F., Glenister, P.H. and Williams, E.D. (1987) Age [109] Ganesan, S., Silver, D.P., Greenberg, R.A., Avni, D., Drapkin, R., Miron, A., Mok,
related reactivation of an X-linked gene. Nature 327, 725727. S.C., Randrianarison, V., Brodie, S., Salstrom, J., Rasmussen, T.P., Klimke, A.,
[95] Borah, V., Shah, P.N., Ghosh, S.N., Sampat, M.B. and Jussawalla, D.J. (1980) Marrese, C., Marahrens, Y., Deng, C.X., Feunteun, J. and Livingston, D.M.
Further studies on the prognostic importance of Barr body frequency in (2002) BRCA1 supports XIST RNA concentration on the inactive X
human breast cancer: with discussion on its probable mechanism. J. Surg. chromosome. Cell 111, 393405.
Oncol. 13, 17. [110] Gorski, J.J., Savage, K.I., Mulligan, J.M., McDade, S.S., Blayney, J.K., Ge, Z. and
[96] Richardson, A.L., Wang, Z.C., De Nicolo, A., Lu, X., Brown, M., Miron, A., Liao, X., Harkin, D.P. (2011) Proling of the BRCA1 transcriptome through microarray
Iglehart, J.D., Livingston, D.M. and Ganesan, S. (2006) X chromosomal and ChIP-chip analysis. Nucleic Acids Res. 39, 95369548.
abnormalities in basal-like human breast cancer. Cancer Cell 9, 121132. [111] Meraldi, P., Honda, R. and Nigg, E.A. (2004) Aurora kinases link chromosome
[97] Kawakami, T., Zhang, C., Taniguchi, T., Kim, C.J., Okada, Y., Sugihara, H., segregation and cell division to cancer susceptibility. Curr. Opin. Genet. Dev.
Hattori, T., Reeve, A.E., Ogawa, O. and Okamoto, K. (2004) Characterization of 14, 2936.
loss-of-inactive X in Klinefelter syndrome and female-derived cancer cells. [112] Hall, L.L., Byron, M., Pageau, G. and Lawrence, J.B. (2009) AURKB-mediated
Oncogene 23, 61636169. effects on chromatin regulate binding versus release of XIST RNA to the
[98] Sirchia, S.M., Ramoscelli, L., Grati, F.R., Barbera, F., Coradini, D., Rossella, F., inactive chromosome. J. Cell Biol. 186, 491507.
Porta, G., Lesma, E., Ruggeri, A., Radice, P., Simoni, G. and Miozzo, M. (2005)

Você também pode gostar