TADS y Xi

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

REVIEWS

M O D E S O F T R A N S C R I P T I O N A L R E G U L AT I O N

The X chromosome in space


Teddy Jégu1,2, Eric Aeby1,2 and Jeannie T. Lee1,2
Abstract | Extensive 3D folding is required to package a genome into the tiny nuclear space, and
this packaging must be compatible with proper gene expression. Thus, in the well-hierarchized
nucleus, chromosomes occupy discrete territories and adopt specific 3D organizational structures
that facilitate interactions between regulatory elements for gene expression. The mammalian
X chromosome exemplifies this structure–function relationship. Recent studies have shown that,
upon X‑chromosome inactivation, active and inactive X chromosomes localize to different
subnuclear positions and adopt distinct chromosomal architectures that reflect their activity
states. Here, we review the roles of long non-coding RNAs, chromosomal organizational structures
and the subnuclear localization of chromosomes as they relate to X‑linked gene expression.

X‑chromosome inactivation In mammals, sex is determined by a pair of sexually Recent investigations in mice have focused inten‑
(XCI). A process to dimorphic chromosomes (the X and Y chromosomes), sively on the functional roles of lncRNAs. For example,
epigenetically silence one with males being the heterogametic sex (XY) and the Jpx RNA has been proposed as a numerator element
X chromosome in the somatic females being the homogametic partner (XX). This in controlling X‑chromosome counting 17. Tsix and Xite
cells of female mammals to
achieve similar expression
context induces an imbalance in the dosage of X‑linked together control allelic choice and also designate the
levels between the sexes. genes between the two sexes. In females, dosage com‑ active X chromosome by preventing Xist expression on
pensation between the sexes is achieved by transcrip‑ this chromosome18–20. By contrast, Jpx and RepA RNAs
Long non-coding RNAs tionally silencing most genes on one X chromosome, a have been suggested to serve as positive regulators of
(lncRNAs). Transcripts of more
process called X‑chromosome inactivation (XCI)1–5. The Xist 9,21. Once expressed, Xist RNA binds to a critical
than 200 nucleotides that do
not encode proteins. choice of which X chromosome to silence takes place nucleation site located in the Xist gene body 22 and spreads
early in development in a random manner, generating from that site to the rest of the X chromosome to establish
Xist an active X chromo­some (Xa) and an inactive X chromo‑ chromosome-wide silencing 23–25. Xist RNA interacts with
(X-inactive specific transcript). some (Xi), which are maintained thus throughout adult a large proteome26–28 and collaborates with that proteome
A long non-coding RNA that is
transcribed from the
life. XCI induces exclusion of RNA polymerase II and to form heterochromatin on the X chromosome.
X-inactivation centre and is the transcription factors from the Xi. This is accompanied In the past few years, it has become clear that l­ nc­RNAs
key regulator of mouse by the recruitment of histone and DNA modification of the Xic and beyond do more than target silencing pro‑
X‑chromosome inactivation. complexes to deposit repressive marks on the Xi such teins to the chromosome. In fact, the epigenetic regula‑
as histone H3 lysine 27 trim­ethylation (H3K27me3), tion of XCI is about much more than getting chromatin
H3K9me2, the H2A ­histone variant macroH2A and DNA complexes and transcription factors to the right place at
methylation6–10. the right time. Recent work suggests that the Xic also con‑
XCI is initiated by a single key locus, the X‑inactivation trols chromosome movement and the overall architecture
centre (Xic), which in the mouse X chromosome is a of the X chromosome in 3D nuclear space26,29–33. Here,
100–500 kb region that contains all required sequences for we discuss these recent advances as well as the essential
this process11 (FIG. 1a). The core region of the Xic — the roles of chromatin architecture (BOX 1) and lncRNAs in
1
Howard Hughes Medical region sufficient to direct silencing in cis when placed on the XCI process. Most of the discussed studies focus on
Institute; Department of
Molecular Biology,
an autosome — is only 100 kb in size and, intriguingly, mice, although we also provide an overview of human
Massachusetts General contains no protein-coding genes. Instead, this region har‑ studies where appropriate, in particular when reviewing
Hospital, Boston, bours elements that produce long non-coding RNAs (lncR‑ the implications of both the 3D structure and subnuclear
Massachusetts 02114, USA. NAs), including Xist, RepA, Tsix, Xite and Jpx (also known localization of X chromosomes during XCI.
2
Department of Genetics,
as Enox (expressed neighbour of Xist))11,12. Outside the core
Harvard Medical School,
Boston, Massachusetts region lie genes encoding other lncRNAs (for example, Counting in time
02115, USA. Ftx andTsx)13,14, as well as one protein-coding gene, Rnf12 XCI occurs very early in development. In mice, XCI is
Correspondence to J.T.L. (also known as Rlim)15. The minimal critical region of the observed in the late blastocyst stage, specifically in the
lee@molbio.mgh.harvard.edu human XIC has yet to be defined, but the region is highly epiblast lineage at the time of uterine im­­plantation4.
doi:10.1038/nrg.2017.17 conserved around the 2.3 Mb syntenic region, which At this time, all epiblast cells make a numerical
Published online 8 May 2017 contains many of the same genes as in mice16. ­determination of their X‑chromosome content.

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a 10 kb
Xic Pairing
Rnf12 Xpct Ftx Xist RepA centre

Jpx Tsix Tsx


Xpr Xite
Nucleation
centre

b
Pre-XCI X–X pairing XCI onset Xist
Xi
Xist
CTCF Xist Xite Jpx Tsix
Jpx Tsix Xite CTCF Xite
OCT4 CTCF CTCF CTCF
Jpx Tsix
OCT4 CTCF CTCF
Xist CTCF Xa Xist
CTCF Xist Xite CTCF
Jpx Tsix Xite Jpx
Tsix Xite

Xist activation Nucleation Xist spreading


YY1
Xist
Xist Xist
RNA YY1 RNA
Xi Pol II Xi Pol II Xi
Nucleation centre

Figure 1 | The potential roles of long non-coding RNAs in the initiation of X‑chromosome inactivation.
Nature Reviews | Genetics
a | The X‑inactivation centre (Xic) in mice. Non-coding genes with established functions in regulating X‑chromosome
inactivation (XCI) are shown in colour, whereas those in grey have been proposed as candidate regulators. The role of
the X‑pairing region (Xpr) in pairing is under debate. Rnf12 is a protein-coding gene located ~500 kb upstream of Xist.
Arrows indicate regions involved in the different steps of XCI. b | A proposed model that integrates counting,
X‑chromosome pairing, nucleation and Xist spreading mechanisms. Upon initiation of XCI, X–X pairing allows for
random expression of Xist from one allele only. X–X pairing involves the long non-coding RNA (lncRNA) Tsix, CTCF and
octamer-binding transcription factor 4 (OCT4). However, Xist expression also requires relief of CTCF negative
regulation at its promoter. This might be accomplished by the lncRNA Jpx (also known as Enox (expressed neighbour of
Xist)), which has been proposed to function as a X‑chromosome numerator in the counting process17. An alternative
model has also been reported39. Once activated, Xist is captured by the nucleation centre, where yin and yang 1 (YY1)
serves as receptor, and from here, Xist spreads in a cis-limited fashion to inactivate the X chromosome. CTCF- and
YY1-binding sites relevant to each XCI step are represented. RNA Pol II, RNA polymerase II; Xa, active X chromosome;
Xi, inactive X chromosome.

How does a cell know whether it has one, two named the ‘competence factor’ (REFS 18,19,39) — a com‑
or three X chromosomes? It has long been established plex of X‑linked and autosomal factors that would titrate
that counting involves not only determining each other and effectuate the X:A ratio.
the X‑chromosome number but also the ratio With its existence supported by genetic evidence
of X chromo­s omes to autosomes (X:A). In diploid only, the competence factor remained elusive until
cells, one X chromo­some is retained as the Xa and all recently, when two candidates came to light. The E3
remaining X chromo­somes are inactivated. In tetraploid ubiquitin ligase RNF12 has been proposed to acti‑
cells, two X chromosomes are retained as Xas and all oth‑ vate Xist in trans by targeting the pluripotency factor
ers are inactivated. Initially, the counting mechanism was reduced expression protein 1 (REX1; also known as
proposed to involve only a ‘blocking factor’ — defined ZFP42) for degradation; REX1 has been shown to acti‑
as a complex of autosomal factors that binds to one Xic vate Tsix and negatively regulate Xist expression15,41,42.
and prevents inactivation of the X chromosome in cis34–37. Interestingly, studies recently reported that RNF12
Previously, the blocking factor was thought to bind and is dispensable for random XCI in vivo, but demon‑
transactivate Tsix 38,39, which in turn prevents Xist induc‑ strated that RNF12 is required for imprinted XCI43,44,
tion and thereby keeps the X chromosome on in cis. a form of XCI that occurs in preimplantation embryos
However, deleting Tsix in male cells does not result in Xist and placental tissue of mice. Notably, imprinted XCI
upregulation and silencing of the only X chromosome does not involve a counting mechanism. Furthermore,
present18,40. This observation suggested that an additional although Xist upregulation is affected, a significant frac‑
factor (or factors) is required to render female cells com‑ tion of Rnf12+/− female embryonic stem (ES) cells and
petent for XCI. The additional hypothetical factor was embryos can still undergo XCI15,44,45. Therefore, RNF12

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

CTCF may not strictly be required for the counting process, It should be noted that there is an ongoing debate
(CCCTC-binding factor). although it clearly affects X‑chromosome silencing in regarding the relative contributions of Jpx and RNF12 to
A highly conserved other ways. the counting process. Barakat et al.45 created a Xic dele‑
DNA-binding protein that tion that removed Jpx along with nearly all sequences at
regulates chromatin
organization and is involved in
The role of Jpx in counting. The exact molecular nature of the Xic and showed that mouse female ES cells with a
transcriptional regulation and the X‑chromosome counting mechanism remains largely heterozygous deletion could still undergo XCI using the
insulation. unknown. One factor that has recently been implicated wild-type Xist allele. The female cells showed no impair‑
as a numerator element of the X:A ratio in mice is the Jpx ment of XCI on differentiation. However, in a separate
Insulator
lncRNA (FIG. 1b). This lncRNA is encoded in the Xic locus genetic experiment, the authors showed that a dele‑
A DNA sequence element that
blocks heterochromatin
just upstream of Xist and is conserved in placental mam‑ tion containing both Jpx and Rnf12 worsened the XCI
spreading and interaction mals, including mice and humans16,46,47. Genetic analysis defect when the deletion was restricted to Rnf12 (REF. 45),
between enhancers and showed that Jpx RNA serves as an activator of Xist in ES supporting a role for Jpx as a positive regulator of Xist
promoters. cells and, furthermore, that Jpx is haploinsufficient 17,21. expression. In vivo experiments in mice have also argued
Chromosome territories
When a single allele of Jpx was deleted, female mouse against RNF12 as a counting factor during random XCI,
The domains occupied by ES cells were no longer able to upregulate Xist RNA on as the RNF12 protein is downregulated in epiblast cells
chromosomes in the differentiation21. By contrast, when extra copies of Jpx of wild-type embryos before random XCI occurs, and
interphase cell nucleus. were introduced into male ES cells, Xist turned on17, sug‑ females bearing a null mutation targeted to epiblast cells
gesting that XCI is sensitive to the dosage of Jpx RNA. In exhibited no XCI defects44. Moreover, a re‑analysis of the
fact, Jpx seems to function as a diffusible RNA, as the Jpx RNF12‑knockout made in ES cells15 suggests that it yields
gene could be placed on a different chromosome and still a 45 kDa truncated protein that lacks the RING finger
function as an activator, and short hairpin RNAs directed domain (I. Bach, personal communication). Because a
against Jpx abolished its activity 21. Additional genetic and RING-finger-mutated RNF12 has dominant-negative
biochemical analyses showed that Jpx RNA functions by activity 48, it is possible that the mutated RNF12 in ES
evicting CTCF, an architectural protein and chromatin cells resulted in a gain‑of‑function mutation that could
insulator, from the Xist promoter 17 (FIG. 1b). The same study indirectly influence XCI. Thus, although RNF12 influ‑
showed that the transcriptional activation of Xist is regu‑ ences imprinted XCI43,49, its role in random XCI remains
lated by a dynamic balance between levels of Jpx RNA and unclear. In summary, further analyses will be necessary to
CTCF protein, which is suggestive of a model in which Jpx address the contributions of both Jpx and RNF12. Other
RNA serves as a numerator of the X:A ratio and CTCF factors are likely to participate in the counting process.
serves as a denominator 17 (FIG. 1b). For example, depletion of octamer-­binding transcription
factor 4 (OCT4; also known as POU5F1) was shown to
impair X‑chromosome pairing and resulted in increased
Box 1 | Chromatin: a key structure for looping Xist upregulation from both X chromosomes in female
mouse ES cells50, which is indicative of a role for Oct4 as
Although DNA in eukaryotic cells allows the storage of genetic information, it is its a trans factor that regulates counting.
packaging into chromatin that enables the fine organization of this genetic information
throughout the development of organisms. Chromatin is not organized randomly in the
nucleus, and each chromosome in the interphase occupies a spatially well-defined X–X pairing: a rendezvous in space
territory called chromosome territory (see the figure, part a). Although occupying a More than 10 years ago, it was discovered that two
defined territory, chromosomes can be in contact with each other121. Indeed, chromatin female X chromosomes undergo an unusual homo­
loops from one chromosome territory can intermingle with adjacent chromosome logous chromo­some pairing event just before the ini‑
territories. In addition to these interchromosomal interactions, chromosomes can form tiation of XCI51,52 (FIG. 1b). This pairing event has been
multiple intrachromosomal interactions, which are regrouped into different topologically studied extensively in the ex vivo model, mouse ES cells.
associated domains (TADs) with an average size of 1 Mb (REF. 81) (see the figure, part b). Undifferentiated female ES cells harbour two active
Chromosomes are partitioned into several TADs, which are spatially separated from each X chromosomes, and the process of cell differentiation
other. TADs seem to regulate gene expression by facilitating or preventing interactions induces the female cells to transition through the differ‑
between regulatory elements and, interestingly, gene expression within the same TAD is
ent steps of XCI. The two X chromosomes occupy dis‑
more correlated than between two different TADs. Boundaries between each TAD
constitute key points of TAD organization. The TAD boundaries are enriched for binding tinct chromosome territories in undifferentiated female
sites of the architectural proteins CTCF and cohesin122 (see the figure, part c). It has been ES cells, but they become colocalized when the cells are
observed that cohesins can organize interactions within domains and that CTCF is induced to differentiation (FIG. 1b). This phenomenon has
required to separate neighbouring folding domains123. Although CTCF and cohesin are been called ‘X–X pairing’ (REFS 51,52) and occurs only
known to shape genome topology, the mechanisms by which they regulate 3D transiently, with a measured half-life of <30 minutes53,54.
chromosome organization remain to be elucidated in more detail. When the X chromosomes separate, they adopt opposite
a b c transcriptional fates, with one becoming the Xa chromo­
some and the other becoming the Xi chromosome.
TADs
This physical proximity is directed by the Xic and
occurs only at the Xic 51. Genetic analyses have shown
Cohesin
that Tsix and Xite, a known positive regulator of Tsix, are
CTCF both necessary and sufficient to direct the pairing inter‑
action51 (FIG. 1b). Indeed, deleting Tsix and Xite impairs
Chromosome the X–X interactions and results in a chaotic choice of
territories
Xa and Xi19,51,55. Under chaotic choice, Xa and Xi would
Nature Reviews | Genetics

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

no longer be chosen in a mutually exclusive manner by activator 39. In this model, each X chromosome inde‑
putative blocking and competence factors. By contrast, pendently decides whether it will be inactivated. Thus,
inserting 1–3 kb sequences taken from within Tsix and/ some cells would inactivate no X chromosome, others
or Xite into autosomes results in a remarkable ectopic would inactivate both X chromosomes, and the remain‑
pairing between the X chromosome and autosome — an ing cells would inactivate only one of the two X chromo‑
action that precludes X–X pairing and results in a block somes. Only those cells that make the stochastic choice
to the XCI process51. These genetic studies argue for a of inactivating one X chromosome would survive the
biological role of X–X pairing in XCI. developmental process and go on to contribute to the
Characterization of the genetic elements required for embryo, supporting a mechanism that is less than robust
X–X pairing point to an essential function of the Xic, in a biological sense (that is, a mechanism that inher‑
although the mechanisms remain mostly uncharac‑ ently includes errors). Hence, one would expect female
terized. It is, however, known that active transcription embryos undergoing XCI to demonstrate excessive cell
and CTCF are both required for X–X pairing 53. Acutely death in the epiblast lineage, a phenomenon that has not
inhibiting RNA polymerase II transcription blocks the been observed39.
colocalization of the Xics during cell differen­tiation. With regard to allelic choice, recent evidence suggests
Interestingly, this requirement for transcription may that the counting and choice mechanisms underlying
reflect the necessity of Tsix and Xite RNAs for the dosage compensation might differ in some respects
recruitment of CTCF protein to the ~15 kb ‘pairing cen‑ between mice and humans, despite a strong conserva‑
tre’ (REF. 56) (FIG. 1b). Indeed, disrupting the RNA–CTCF tion of the Xic and XIC elements between these species.
interaction prevented X–X pairing 56. The pluripotency Indeed, a number of studies have suggested biallelic XIST
factor OCT4 also influences the timing of the pairing expression in human preimplantation embryos63,64, and
event during cell differentiation by binding to the 5′ end that biallelic expression is accompanied by a reduction of
of Tsix 50 (FIG. 1b). Another locus, Xpr (X‑pairing region), gene expression from both chromosomes64,65. XCI does
has been proposed to influence pairing 57 (FIG. 1a) . not formally begin until one of the XIST loci is silenced.
However, a later study argued against a role for Xpr on The XACT lncRNA has been proposed to be a regulator
the basis of its inability to induce pairing on a transgene in this regard66,67. XACT expression is associated with the
array 58. A knockout of Xpr has, so far, not shed light on Xa, and a recent study used transgenic and knockdown
its necessity for X–X interactions. analyses to propose that it may affect the choice of XCI
Although Xic–Xic pairing is unequivocally one of by opposing XIST silencing 67. Genetic deletion of XACT
the first observable events during female cell differen‑ will be required to determine whether it directly affects
tiation and precedes XCI, the molecular consequences XIST expression.
of pairing have been the subject of active debate. One
idea is that pairing is part of an allelic choice mecha‑ The Xi and its spatial transformation
nism. That is, pairing could be a mechanism by which Nucleation: a required step for Xist spreading and
the two X chromosomes adopt mutually exclusive and Xi‑silencing launch. Once expressed from the Xi, Xist
opposite transcriptional states51. Indeed, inactivation coats the chromosome, and its spreading along the Xi
of one X chromosome must preclude silencing of the induces chromosome-wide silencing 23–25. Before this
second X chromosome, and there would need to be a cis spreading and during its transcription, Xist RNA
robust mechanism to render the decision mutually exclu‑ must be captured by a nucleation site located in the Xist
sive. Live-cell imaging that tracks the movement of two gene body in order to be retained on the Xi22 (FIG. 1b).
X chromosomes supports the idea that Tsix expression This capture is mediated by the transcription factor yin
becomes monoallelic after X–X pairing 54. Interestingly, and yang 1 (YY1), which binds to the nucleation site
allelic pairing in mammalian somatic cells has been in the Xist exon 1. The bivalent property of YY1 allows
observed in other cases of allelism59–62 and could poten‑ the protein to bridge and tether Xist RNA to the nuclea‑
tially be generalized to other allelic regulatory situations. tion site DNA on the Xi22. Without the nucleation event,
Alternatively, pairing may not be required in the case Xist RNA is released from the site of transcription
of XCI. One study observed XCI occurring in the two and cannot spread in cis along the X chromosome22.
nuclei of heterokaryons in spite of the X chromosomes Although Xist RNA diffuses through the nucleus, it
being separated by a nuclear envelope45. However, it was does not bind to ectopic sites, nor does it bind to the
not ruled out that the heterokaryons continue to divide (future) Xa22. It is currently not clear what blocks the
and the nuclear envelope breaks downs during cell divi‑ Xa site and other YY1-binding sites from Xist binding,
sion, permitting the mixing of chromosomes between although differential DNA methylation was proposed
two nuclei. to contribute to this68,69. Jeon and Lee22 observed that
As with the counting mechanism, a number introducing a Xist transgene into the genome of dif‑
of questions regarding the choice mechanism still ferentiated cells enables endogenous Xist RNA to dif‑
await answers. Although we favour a specific, robust fuse throughout the nucleus and migrate (in trans) to
mechanism for the determination of allelic choice, a the transgenic Xist DNA locus, as the newly introduced
stochastic model has been proposed in which the choice transgene lacks the blocking signal.
Heterokaryons
of X chromo­somes for inactivation is made by chance, In contrast to these observations, one study sug‑
Cells containing genetically with every X chromosome having an equal prob­ability gested that the YY1 element could be involved in the
different nuclei. of becoming inactivated on titration of an X‑linked direct transcriptional upregulation of Xist in ES cells68.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

It is entirely possible that the YY1 element has both tran‑ trans sites. Xist RNA would be expected to bind YY1
scriptional and nucleating functions. Another study did targets in cis with greater probability owing to the
not observe any diffusion capacity of Xist in mouse ES co‑transcriptional nature of the Xist–YY1 interaction22.
cells70, although the lack of diffusion might be explained The ability to observe trans diffusion would be increased
by stoichiometry and competition between cis and when the stoichiometry of the trans site is favoured over
the cis site. Indeed, the trans diffusion observed by Jeon
and Lee occurred in the context of multi-copy trans‑
A
genes22. Finally, how Xist RNA spreads from the nucle‑
ation centre to the rest of the X chromosome in cis is
currently not known. One idea is that its spreading is
guided by the 3D spatial organization of the Xi24,25, but
specific factors for spreading have yet to be identified.
Xi
Two distinct 3D X‑chromosome structures. Profound
Xa changes in overall chromosome structure during the
process of XCI perfectly exemplify the link between
the 3D chromosome organization and gene expression.
Differences in shape between Xa and Xi territories have
long been noted. The Xi territory is characterized by a
Ba Xa chromosome territory Bb Xi chromosome territory spherical shape and smooth surface, whereas the Xa ter‑
ritory displays a flatter shape and a larger and more irreg‑
IC TF CD or TAD ular surface71,72 (FIG. 2A). Although the Xi is significantly
Xist
more compacted than the Xa, the differences between
PR RNA Pol II these two homologues are surprisingly small; the overall
compaction of the Xi is 1.2‑fold higher than that of the
Speckles IC Collapsed Xa30,73,74. Except for local differences on the promoters
PR ANC
CDC of genes that are expressed from the Xa and silent on
INC the Xi, both chromosomes have a similar global 30 nm
Inactive chromatin structure. However, the two X chromosomes
gene display significant differences beyond this 30 nm fibre.
INC Indeed, large-scale chromatin structures on gene-rich
ANC PR regions are more condensed on the Xi than on the Xa74.
These observations were corroborated by 3D fluores‑
cence in situ hybridization (FISH) analysis showing that
Ca Xa TADs Cb Xi megadomains chromatin domains of approximately 1 Mb are similarly
compacted between the Xa and Xi, but a higher compac‑
tion in Xi territory is observed for specific chromosomal
segments of 20 Mb (REF. 73). Moreover, light and electron
microscopy have revealed that the condensed region of
the Xi has a distinctive ultrastructure and that the Xi
is not solid but rather has a sponge-like structure that is
organized in large, tightly packed chromatin domains
Dxz4 separated by interchromatin channels, which are contigu‑
ous with nuclear pores75. These observations are consist‑
ent with the concept of an active nuclear compartment
(ANC) and an inactive nuclear compartment (INC)76,
500 kb resolution 500 kb resolution a network model in which the chromosome territories
Figure 2 | The 3D organization of X chromosomes. A | The inactive X chromosome (Xi) are composed of two interacting networks with their
adopts a spherical shape with a smooth surface and is organizedNatureinto two megadomains,
Reviews | Genetics own dynamic organization. The INC is formed by the
whereas the active X chromosome (Xa) adopts a flatter shape with a more irregular transcriptionally silent and compacted core of chromatin
surface area. B | The active nuclear compartment (ANC) and inactive nuclear domain clusters (CDCs), whereas the ANC is formed by
compartment (INC)77 network model of chromosome territory structure proposes that the perichromatin region and the interchromatin channel
basic structural features of chromosome territory organization are present in both the Xa and is where transcription occurs77 (FIG. 2B).
(part Ba) and the Xi (part Bb). The INC is transcriptionally silent and contains compacted Recent super-resolution microscopy studies have
chromatin domain clusters (CDCs), whereas transcription occurs in the ANC, which is revealed that the Xi territory displays major differences
composed of the perichromatin region (PR) and the interchromatin channel (IC)77. In the Xi
from transcriptionally active chromosome territories,
chromosome territory, the ANC is partially collapsed and the CDCs are located closer to
each other than in the Xa chromosome territory. C | Allele-specific Hi‑C maps of the Xa although the basic structural features of chromosome
(part Ca) and Xi (part Cb) chromosomes, showing that the Xa is organized into territory organization seem to be maintained76. Indeed,
topologically associated domains (TADs) and the Xi is organized into two megadomains although the CDCs and interchromatin channels are
separated by the Dxz4 locus. CD, chromatin domain; RNA Pol II, RNA polymerase II. observable, the Xi chromosome territory displays par‑
Part C is reproduced with permission from REF. 30, Macmillan Publishers Ltd. tially collapsed ANCs with CDCs closer to each other

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

(FIG. 2B) .Altogether, these observations argue for a the Xi by repelling their binding in cis, preventing the
‘non-monotonous compaction’ model of the Xi in which ­establishment of an Xa‑like binding pattern.
Sponge-like structure differences observed between the two X chromosomes Previously, allele-specific chromosome conforma‑
(Also called a porous structure). result from reorganization of the higher-order structures tion capture-on-chip (4C) experiments showed that
A model of chromosome
territory structure in which
rather than a modulation of ­chromatin compaction at silenced genes on the Xi lack 3D contacts that are seen
chromolome territories are the nucleosome level. on the Xa, and deletion of Xist from the Xi causes con‑
composed of interconnected The development of chromosome conformation tacts to re‑form in the same genomic locations as found
chromatin domains permeated capture methods has enabled the fine-mapping of on the Xa31. Hi‑C experiments performed in an Xist
by an interchromatin channel.
X‑chromosome topology 78,79. At whole-chromosome ablation mutant (XiΔXist) extended this observation and
Interchromatin channels scale, recent studies have shed light on significant demonstrated a crucial role for Xist in shaping the Xi 3D
Channels that pervade differences in the folding of the Xa and Xi in human conformation26. Indeed, deletion of Xist results in a mod‑
chromatin domain clusters and mouse cells 26,30,31,80–84. Hi‑C experiments have ulation of Xi folding, forming an Xa‑like chromosome
both between and within revealed that the Xa is organized into >100 topologically conformation through the restoration of TAD struc‑
chromosome territories. The
associated domains (TADs) on the whole chromosome, tures on the Xi. This TAD restoration correlated with
channels are a chromatin-free
compartment containing RNA whereas the Xi seems devoid of such megabase-scale the re‑establishment of Xi–cohesin binding, suggesting
polymerase II, transcription structures across its full length and is instead organized that Xist-mediated repulsion of cohesins from the Xi
factories, splicing speckles and into two megadomains of frequent intrachromosomal prevents an Xa conformation. More recently, it has been
architectural proteins.
contacts26,30,80,82,84 (FIG. 2C). The boundary region between observed that induction of Xist RNA expression on the
Chromatin domain clusters these two megadomains is conserved between human X chromosome in undifferentiated male mouse ES cells
(CDCs). Groups of chromatin and mouse and is located near the macrosatellite repeat generates the formation of the two megadomains sepa‑
domains that comprise the DXZ4 or Dxz4, respectively26,30,80,82,84. rated by the boundary at the Dxz4 locus; the same study
inactive nuclear compartment. Significant differences in loop structures between the found that the role of Xist in regulation of Xi topology
Perichromatin region
two X chromosomes can be observed. Indeed, in human seems to be dependent on the Xist A‑repeat region30. Xist
The decondensed and cells, there is a network of long-range superloops that is shapes the topology of the Xi, while the 3D organiza‑
transcriptionally competent present only on the Xi, and four of the main anchor tion of the Xi in turn guides the binding of Xist lncRNA
chromatin region residing at regions involved in the superloop network contain the during the establishment of XCI24,25. In the future, the
the periphery of chromatin
lncRNA genes XIST, DXZ4, FIRRE and LOC550643 integration of a temporal dimension to the existing 3D
domain clusters.
(REFS 82,84,85). Interestingly, YY1 seems to bind these analysis using time course analysis during cell differen‑
Chromosome conformation regions only on the Xi, suggesting a potential role of YY1 tiation will enable understanding of the reciprocal regu‑
capture in the regulation of Xi‑specific superloops86. Moreover, lation between Xist and Xi topology during the different
A technique to detect physical the three genes DXZ4, FIRRE and LOC550643 are bound steps of XCI.
interactions between distant
DNA sequences.
by CTCF, and the superloops between these three loci The idea that lncRNAs may generally regulate CTCF
can occur simultaneously in the same cell, specifically on and cohesin localization could be generalized to include
Topologically associated the Xi84. Chromatin interaction analysis by paired-end domains outside the X chromosome26,56,91–94. Thus, it is
domains tag sequencing (ChIA-PET) experiments have supported tempting to speculate on the possibility that the inter‑
(TADs). Units of chromatin that
the direct role of CTCF in anchoring the super- actions between the chromatin architectural proteins,
display a high frequency of
DNA long-range interactions long-range interactions between these loci only on the CTCF and cohesins, and specific lncRNAs constitute a
between loci located in the Xi87. This finding suggests a crucial role for the allelic key regulatory aspect of 3D nuclear organization.
same unit and a low frequency binding of CTCF in X‑chromosome topology regula‑
of association between loci tion. CTCF therefore seems to be a recurrent player in Different spaces and destinations for silent and escapee
located in adjacent units.
the regulation of XCI, from counting, to choice, to the genes on the Xi. Genes on both the mouse and human
Macrosatellite spread of silencing. X chromosomes have been observed to escape from XCI.
A specific class of large tandem In humans, 15% of X‑linked genes are not fully subject to
repeat DNA that is Xist RNA shapes the Xi conformation. Evidence indi‑ XCI and are apparently less susceptible to the repressive
characterized by the large size
cates that asymmetric binding of CTCF and cohesins on effect of XIST RNA95. In mice, a smaller number of genes
of individual repeats, which can
each be several kilobases long. the Xa versus Xi could account for their distinct topol‑ escape from XCI. Although escapee genes vary between
Macrosatellites can span ogies26,56,80,88–90. Interestingly, cohesins bind the X chro‑ mouse tissues90,96–100, most are positioned close to each
hundreds of kilobases of DNA. mosomes in an allele-specific manner that favours the other along the chromosome90, which suggests that
Xa. There are 500–600 Xa‑specific cohesin sites and only local rearrangements between tissues expose escapee
Superloops
Extremely long-range
~20 Xi‑specific cohesin sites, and the rest of the ~200 genes to the transcriptionally competent compartments.
intrachromosomal contacts sites are shared by the Xa and Xi26. In this same study, This idea is corroborated by recent discoveries showing
between two loci, up to 80 Mb it was shown that an Xi conditionally deleted for Xist that escapee genes that are close to each other can be
long. (XiΔXist) in mouse fibroblasts is sufficient to restore the associated with the same individual TAD in mouse ES
binding of a large number of cohesins on the Xi, sug‑ cells100. Interestingly, although the Xi has two megad‑
Cohesins
Protein complexes composed gesting that cohesin binding on the Xi is influenced omains in both Patski and F1 brain cells, the Xi mega­
of the core subunits SMC3, by Xist RNA. In fact, the anti-correlation between Xist domains in brain seem to be more condensed than
SMC1, RAD21 and STAG1 and cohesins implies that Xist has a repelling effect on in Patski cells, which have more escapee genes80,90.
(also known as SA1) or STAG2 cohesins. Proteomic purification for Xist RNA showed Although the Xi is devoid of TAD structures, escapee
(also known as SA2). Cohesin
participates with CTCF to
that Xist can directly interact with CTCF and co­­ genes have been proposed to form mini-TADs30 in
mediate long-range chromatin hesins26. Thus, in this new model, Xist interaction with neural progenitor clones (NPCs). This study also sug‑
interactions. co­­hesins is proposed to antagonize a set of cohesins on gested an unusually large number of escapee genes in

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a b Escapee of the Xi — the so‑called core, which is composed of


Escapee
repeat-dense regions and inactivated genes bound by
Xist lncRNA. This relocalization seems to be dependent
on the Xist A‑repeat region and may be associated with
long interspersed nuclear elements (LINEs)103,104. Taken
together, these studies suggest that the inside of the Xi
Xist territory forms a transcriptionally silent compartment,
Xist marked by Xist, whereas the periphery of the Xi creates
an environ­ment that is more permissive for transcrip‑
Inactive tion, with specific interactions involved in gene escape
gene (FIG. 3). A potential concern for this model, which argues
Inactive
gene for a compacted central core and a sharp geographical
division between repressed (interior) and expressed
(exterior) genes, is that repressed and expressed genes
(such as escapees) are often separated only by a few
c d kilobases, and it may be difficult to envision how two
Xist neighbouring genes can be partitioned so distantly and
ANC sharply in space.
Escapee Indeed, additional studies have reached somewhat
Escapee different conclusions with regard to how the Xi terri‑
tory is organized at the cytological level. Clemson and
collaborators agree that the internal core of the Xi terri‑
Xist tory is composed of non-genic sequences, but found that
the outer rim is formed by gene-rich regions regardless
Inactive
Inactive gene of their transcriptional activity 105 (FIG. 3). Another study
gene has shown that Xi‑silencing genes can be located and
Collapsed ANC maintained outside the Xi territory and that transcrip‑
tion can occur inside the Xi99 (FIG. 3). In separate studies,
Figure 3 | Models for the position of silent and escapee genes on the Xi. The ‘core’ the Cremer and Belmont groups also argue against the
model103,104 (part a) posits that inactive genes are in a transcriptionally silent compartment
model whereby inactive genes are inside the Xi territory
marked by Xist, and escapees are exterior to this core. The ‘outerNature
rim’ model
Reviews(part
105
b)
| Genetics
proposes that non-genic sequences form a core marked by Xist, whereas inactive and and escapee genes are localized at the periphery, instead
escapee genes are localized to the outer rim of the inactive X chromosome (Xi). A model proposing that genes escaping XCI are distributed
proposed by Calabrese et al.99 (part c) suggests that X‑silencing can be located and throughout the Xi territory 73,75,76 (FIG. 3). In this model,
maintained outside the Xist core and that transcription can occur inside this core. In the silenced chromatin is closer to transcriptionally compe‑
‘ANC-INC’ model (part d) proposed by Smeets et al.76, the Xi chromosome territory is tent compartments than in the central core model, and
pervaded by a collapsed ANC, which is enriched in Xist and inactive genes. Escapees are therefore allows for a close proximity between active
located both inside and outside the Xi chromosome territory. escapees and adjacent silenced genes. This model, in
which active and silent genes can ‘intermix’ in space,
would circumvent the physical problem associated with
the central core model.
one NPC clone and a high variability between inde‑ How escapees, which often reside next to silent
pendent NPC clones. For this reason, further investiga‑ genes, can be immune to the silencing effect of Xist is
tions are required to better understand the link between currently not known. However, studies have shown that
escapee gene expression and 3D chromosome structure. escapees often have CTCF-binding sites located near
A 3D model of the Xi with 1 Mb resolution showed their promoters30, and CTCF seems to separate inactive
that CTCF-enriched regions and active escapee genes and escape domains by binding to the boundary regions
Xist A‑repeat region are located preferentially at the periphery of the Xi ter‑ between escapees and closely juxtaposed inactive
A domain of Xist RNA that has ritory in mouse F1 brain80. Another study also proposed genes90,106,107. Interestingly, CTCF may be enriched in the
been identified as necessary that whereas silenced genes on the Xi are engaged in perichromatin regions77, suggesting that CTCF could
for Xist-mediated silencing but
not for localization of Xist to
very few interchromosomal contacts and reside in the bind to these boundary regions in the peri­chromatin
the X chromosome. chromosomal territory’s interior 31, escapee genes are regions to sequester inactive genes in the INC and
located at the periphery of the Xi. Escapee genes dis‑ expose adjacent escapees in the ANC. Notably, studies
Escapee genes play more specific contacts compared with genes that using allele-specific chromatin immunoprecipitation
X‑Linked genes that are
are subject to XCI and are engaged in long-range con‑ followed by sequencing in mouse cells have shown
biallelically expressed in
female somatic cells. tacts with each other and in many interchromosomal that CTCF binds to the Xa and Xi in a distinct man‑
interactions31,80. ner, helping to organize the Xa and Xi into functional
Long interspersed nuclear These studies confirm previous observations show‑ compartments for gene expression and repression26,56,99.
elements ing that silenced genes have a more internal position Recently, it has been hypothesized that YY1 may have
(LINEs). A type of repetitive
DNA in the mammalian
in the Xi101–103. After cell differentiation and the initi‑ a role in facilitating escape from XCI in human cells,
genome that is derived from ation of XCI, X‑linked genes are re‑localized from the potentially by binding near the transcription start site
transposons. periphery of the Xi territory to a more internal location of escapee genes86.

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Human Mouse Dxz4 macrosatellite, the DsTR minisatellite, their asso‑


ciated lncRNAs and CTCF-binding sites. In mice, the
DXZ4 Dxz4 DsTR Firre locus and its corresponding lncRNA also seem to
Xi Xi be involved in the regulation of Xi topology 111.
CTCF CTCF CTCF CTCF CTCF

TADs in Xic and the role of RS14. In the X chromo‑


some, the Xic can be partitioned into two distinct TADs,
DXZ4 Dxz4 DsTR which have opposite effects on Xist expression (FIG. 5).
Xa Xa
CTCF CTCF CTCF The Xist TAD contains the Xist promoter, its 5′ sequence
and its positive regulators, including Jpx, Ftx, Xpr and
Figure 4 | Organization of the DXZ4 and Dxz4 loci in human and mouse models, Rnf12 loci in a domain of ~550 kb. The Tsix TAD con‑
respectively. The human DXZ4 locus consists of 50–100 copies ofNature
a CpG-rich 3 kb repeat, tains the Tsix promoter, the 5′ sequence of Tsix and its
Reviews | Genetics
which is hypomethylated, organized in euchromatin (green circles) and bound by CTCF on associated regulators, including Xite, Tsx, Dxpas34,
the inactive X chromosome (Xi). On the active X chromosome (Xa), the locus is Chic1 and Linx (also known as Islr2), spanning a total
hypermethylated (red dashed line) and packaged into heterochromatin (red circles) without of 260 kb. The 3D organization of the Xic into the Xist
CTCF binding109. In mice, Dxz4 consists of approximately six repeats and is organized in TAD and the Tsix TAD could facilitate the interactions
euchromatin only on the Xa, but is biallelically bound by CTCF26,108. The minisatellite DsTR is of regulatory elements in each TAD in order to activate
found 50 kb downstream of mouse Dxz4, but does not occur in humans. or repress Xist, respectively 81,112. Expression profiles of
genes located in the same TAD might be correlated dur‑
The DXZ4 locus: a platform for Xi folding. DXZ4 is an ing differentiation. During XCI, genes in the Xist TAD
X‑linked locus that is localized at the boundary between become upregulated, whereas genes located in the Tsix
the two megadomains of the Xi and has a fundamental TAD decrease in expression81. It has been proposed that
role in Xi topology in mouse and human cells. In both before XCI, Tsix and its enhancer Xite interact to pro‑
species, deletion of the DXZ4 locus disrupts mega­domain mote Tsix expression and prevent the activation of Xist,
formation but does not seem to affect the establishment which seems to make a looping contact with Jpx in a
of XCI30,84. The structure and organization of this macro­ ‘poised’ interaction112 (FIG. 5).
satellite are somewhat different between the two species108 On the Xa, the Tsix–Xite interaction is maintained,
(FIG. 4). The human DXZ4 consists of 50–100 copies of a favouring Tsix expression and Xist repression. At the
CpG-rich 3 kb repeat, which is hypo­methylated, organ‑ same time, the interaction between Xist and Jpx is lost
ized in euchromatin and bound by CTCF on the Xi, but on the Xa. By contrast, on the future Xi, the interaction
is hypermethylated, packaged into hetero­chromatin and between Tsix and Xite is lost 112. This leads to repres‑
not bound by CTCF on the Xa109. In mice, Dxz4 con‑ sion of Tsix, which in turn enables the activation of
sists of approximately six repeats that are not particu‑ Xist expression, and the Xist–Jpx interaction becomes
larly GC‑rich, and it is organized in euchromatin only activated112 (FIG. 5). Similar interactions are observed
on Xa and biallelically bound by CTCF26,108. The mini‑ in human cells, in which the interaction between XIST
satellite DsTR lies 50 kb downstream of the mouse Dxz4 and its putative regulator JPX is anchored by RNA poly­
locus, and could be expressed biallelically but seems to merase II87 and is only detected on the Xi, supporting
be mainly bound by CTCF on the Xa80,108. There is no the role of this chromatin loop in the regulation of XIST
evidence for the presence of this minisatellite in humans. expression from one of the two X chromosomes only.
In human cells, the Xi is organized into two, The Xist and Tsix TADs are separated by a region
non-overlapping types of heterochromatin, which are called RS14, which is located at the 3′ end of Xist. RS14
sequestered into distinct territories, with macroH2A, contains a strong DNase I hypersensitive site (DHS),
H3K27me3 and Xist representing one type, and can bind CTCF and forms a border between the Xist
H3K9me3 and HP1 marking the second type110. DXZ4, and Tsix TADs81,112–114 (FIG. 5). RS14 plays a part in XCI
FIRRE and LOC550643 loci form a euchromatin domain initiation, as deleting it results in compromised Xist
on the Xi that is bound by CTCF and localized at the induction114. From a molecular point of view, the dis‑
boundary of the two distinct heterochromatin territo‑ ruption of this boundary leads to an altered organization
ries85. Deletion of the DXZ4 locus in somatic human of the Xist and Tsix TADs, and induces ectopic contacts
cells leads to a partial loss of heterochromatin, as the between sequences from previously separated TADs81,113.
H3K27me3 mark seems to recede84. By contrast, deletion Disorganization of the Xist and Tsix TADs in turn causes
of the Dxz4 locus on the mouse Xi is proposed to cause transcriptional misregulation of the genes contained in
more silencing on the Xi, especially for escapee genes30. them81. Interestingly, although the RS14 boundary can
However, a caveat is that this effect was observed pre‑ control the organization of the Xist and Tsix TADs, a
dominantly in one cell line, and there are variable effects reciprocal regulation also seems possible because inter‑
in other cell lines. Therefore, apart from partitioning the actions within the TADs could contribute to defining
Xi into two megadomains, the true function of DXZ4 the boundary between them. Indeed, using a physical
and Dxz4 is not currently known, nor is it known which polymer model-based deconvolution of chromosome
specific element (or elements) in the Dxz4/DXZ4 locus conformation capture carbon copy (5C) data, a virtual
is responsible for the cis effects observed in the two spe‑ disruption of Tsix and its regulators Linx and Chic1 was
cies. Indeed, published deletions are large (~200 kb) and shown to be able to affect the sharpness of the boundary
encompass multiple potential elements, including the between the Xist and Tsix TADs113.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Tsix TAD Xist TAD more specifically during mid‑to‑late S phase when the Xi
is undergoing DNA replication along with other blocks
of heterochromatin across the genome (euchromatin is
Linx
replicated in early S phase). It has been proposed that
Xpr the separation of DNA replication in time and in space
Pre-XCI
Chic1 + Tsix enables the faithful replication and maintenance of the
Xite Xist Xi‑heterochromatin state29. Notably, the perinucleolar
Rnf12
Tsx Jpx space is enriched for sucrose non-fermenting protein 2
homologue (SNF2H; also known as SMARCA5), an
Ftx ISWI chromatin remodelling subunit that has a key role
in heterochromatin replication29.
RS14
Regulation and role of the Xi perinuclear localization.
CTCF CTCF
Although the perinuclear localization of the Xi has been
observed for many decades, the role of this subnuclear
localization during XCI and the molecular mechanisms
DHS
involved are poorly understood. During cell differen‑
tiation, some TADs on the X chromosomes become
Linx Xite lamina-associated domains81. Recent studies showed that
Xpr Xist can interact with membrane attachment nuclear
After XCI
(on Xi) envelope proteins (lamin B receptor (LBR), lamina-
Chic1 associated proteins (LAPs) and SUN domain-containing
Xist
Tsix + Rnf12 protein 2 (SUN2)) (FIG. 5). Among these proteins, LBR
Jpx
Tsx may have a role in Xist-mediated transcriptional silenc‑
ing26,27, which suggests a role of the Xist protein part‑
Xi ners in Xi perinuclear targeting. Although the nuclear
Ftx
periphery is often associated with transcriptional silenc‑
ing 118, the Xa can also be observed at the nuclear enve‑
Figure 5 | The X-inactivation centre is partitioned into two topologically associated lope73, which suggests that the perinuclear localization
domains. Before X-chromosome inactivation (XCI), the X‑inactivationNaturecentre (Xic)
Reviews is
| Genetics of the Xi at the nuclear periphery is not sufficient to
divided into two topologically associated domains (TADs), the Tsix TAD and the Xist TAD,
which are separated by the RS14 boundary. RS14 contains a strong DNase I hypersensitive explain, by itself, the opposite epigenetic states between
site (DHS) and is bound by CTCF. Each TAD contains promoter and 5′ sequences of Tsix or the Xa and Xi. A possible mechanism is that the nuclear
Xist with their respective positive regulators. Before XCI, Tsix and its enhancer Xite interact periphery has two chromatin compartments. One com‑
to promote Tsix expression, whereas the interaction between Xist and Jpx is poised. After partment harbours active chromatin and is associated
differentiation, the interaction between Tsix and Xite is lost, whereas the Xist–Jpx with the nuclear pores that facilitate splicing and the
interaction becomes active. Xi, inactive X chromosome; Xpr, X-pairing region. export of mRNAs for translation in the cytoplasm, and
another compartment harbours heterochromatin and is
associated with lamin B119.
The final journey
XCI is a multistage journey through space. The X chromo­ Regulation and role of the Xi perinucleolar localization.
some not only undergoes regional and local changes Several studies in the past decade have suggested that
in chromosome structure but also travels through the Xic plays a major part in targeting the Xi to the peri‑
nucleus to find its final destination. nucleolar space29,32,33. Zhang and colleagues29 showed
that inserting Xic into an autosome will cause locali‑
Perinuclear and perinucleolar Xi localizations. After zation of the autosome to the perinucleolar space in a
XCI takes place, the Xa and Xi move to separate sub­ cell-cycle-dependent manner. Thus, the Xic is sufficient
nuclear positions as they adopt opposite epigenetic to target chromosomes to the perinucleolar ring during
states (FIG. 6). The Xi (historically known as the hetero‑ mid‑to‑late S phase. It is specifically Xist that is respon‑
chromatic Barr body) was observed to be a ‘nucleolar sible for this localization29 (FIG. 6), as an Xi conditionally
satellite’ present in female, but not male, cells of the deleted for Xist (XiΔXist) in mouse fibroblasts loses the
cat 115. Like other non-sex chromosomes, the Xa does ability to localize to the perinucleolar space. This role of
not have a specified position in the nucleus, but the Xi is Xist is conserved between mammalian species because
distinct in having two favoured subnuclear localization ectopic expression of XIST on an autosome in human
Nuclear periphery
domains — one at the nuclear periphery (perinuclear) and cells is also sufficient to promote the relocalization of
A subcompartment of the
nucleus that is composed of the other next to the nucleolus (perinucleolar)29,116,117 the XIST-bearing chromosome near to the nucleolus32,33.
the nuclear-membrane bilayer, (FIG. 6). Analysis by both electron microscopy and in situ An interesting open question is whether it is the Xist
its associated proteins and the hybridization showed that the Xi is located at the nuclear lncRNA or the Xist locus that is responsible for the
nuclear-pore complexes. envelope in 75–80% of interphase cells116,117. At a spe‑ ­targeting mechanism.
Lamina-associated domains
cific time during the cell cycle, however, the Xi moves More recently, it was observed that Firre, Dxz4 and
Genomic regions that interact to a perinucleolar location29. Indeed, 80–90% of the Xi DsTR are also associated with the nucleolus and repre‑
with the nuclear lamina. migrates to the perinucleolar region during S phase, sent nucleolus-associated domains that can be bound

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Xa TAD
Xa

Nucleolus

XCI initiation

Firre
Nucleolus Xite Xite
Tsix Tsix Dxz4
CTCF CTCF
CTCF Xist
CTCF
X–X pairing
Xic
SNF2H

XCI establishment

Dxz4/DXZ4
LBR
Xi

Xi
Xa Nucleolus Xa Nucleolus
Xist
Two megadomains

Mid-to-late S phase

Figure 6 | The X chromosome in space. Before X-chromosome inactivation (XCI), the two active X chromosomes (Xas)
assume random positions relative to each other. At the onset of XCI, the homologous X chromosomes come into contact
Nature Reviews | Genetics
transiently to choose the future inactive X chromosome (Xi). After X–X pairing, the Xi is located at the nuclear periphery;
this localization might be regulated by the interaction between Xist and the membrane attachment nuclear envelope
protein lamin B receptor (LBR). The Xa can also be located at the nuclear periphery. During mid‑to‑late S phase, the Xi is
located at the nucleolar periphery, where Xist, Dxz4 and Firre loci represent nucleolus-associated domains. This Xi
perinucleolar localization is regulated by the long non-coding RNAs (lncRNAs) Xist and Firre and by CTCF, and seems to be
involved in heterochromatin maintenance. SNF2H, sucrose non-fermenting protein 2 homologue; TAD, topologically
associated domain. Xic, X-inactivation centre.

by the nucleolar protein nucleophosmin80,88. However, the perinucleolar region88 Given the previous observa‑
the mechanism by which these loci are targeted to the tion that CTCF can interact with nucleophosmin and
nucleoli is poorly understood, and it is also unknown tether an insulator region to the nucleolus120, CTCF
whether these sequences are involved in the establish‑ could have a similar role in Xi perinucleolar localiza‑
ment or maintenance phase of silencing. Yang and col‑ tion, that is, tethering Xist, Firre, Dxz4 and DsTR loci
leagues have proposed that Firre and Dxz4 loci can serve to the nucleolus.
as additional attachment points to target the Xi to the The precise mechanistic relationships remain to be
nucleolus88 (FIG. 6). A reduction of Firre lncRNA expres‑ investigated. It is known that disrupting the Xi peri­
sion using short interfering RNAs decreased the asso‑ nucleolar localization observed in Firre lncRNA and
ciation frequency between Firre and Dxz4 loci with the Ctcf knockdowns leads to a decrease of the repressive
nucleolus88. Interestingly, Firre and Dxz4 loci are bound mark H3K27me3 on the Xi, but this reduction does not
by CTCF26,56,80,88,108, and knockdown of CTCF signifi‑ result in the reactivation of X‑linked genes88. Conversely,
cantly reduces the association of Firre and Dxz4 with the loss of Xi nucleolar localization in Xist-deleted cell

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

lines (XiΔXist) is accompanied by a decrease in H3K27me3 knowledge of XCI as well as other epigenetic processes.
enrichment and an increase in Xi gene expression29. However, many questions remain: for example, how can
Furthermore, ectopic localization of an Xist-bearing escapees be immune to the Xi silencing during XCI?
autosome to the perinucleolar region is correlated with And what is the relationship between their 3D organi‑
deposition of the repressive H3K27me3 mark and gene zation and their transcription? What specific factors and
repression33. Taken together, these results suggest that molecular mechanisms are involved in the regulatory
the targeting of the Xi to the nucleolus could play a part processes? It is predicted that lncRNAs play a key part
in heterochromatin maintenance and gene silencing. in the regulation of architectural protein factors such
as CTCF and cohesins. We therefore anticipate that, in
Conclusions the next few years, the development of new approaches
Technological and methodological developments over to study protein–RNA interactions will pave the
the past decade have improved our ability to study 3D way to a better understanding of the molecular linkage
genome organization and have enabled a better under‑ between gene expression, conformational changes in
standing of the relationship between structure and higher-order chromosome structures and nuclear ­spatial
function. This understanding is helping to shape our compartments.

1. Lyon, M. F. Gene action in the X‑chromosome of the 20. Ogawa, Y. & Lee, J. T. Xite, X‑inactivation intergenic 36. Lyon, M. F. Possible mechanisms of X chromosome
mouse (Mus musculus L.). Nature 190, 372–373 transcription elements that regulate the probability of inactivation. Nat. New Biol. 232, 229–232 (1971).
(1961). choice. Mol. Cell 11, 731–743 (2003). 37. Riggs, A. D. X inactivation, differentiation, and DNA
2. Starmer, J. & Magnuson, T. A new model for random 21. Tian, D., Sun, S. & Lee, J. T. The long noncoding RNA, methylation. Cytogenet. Cell Genet. 14, 9–25
X chromosome inactivation. Development 136, 1–10 Jpx, is a molecular switch for X chromosome (1975).
(2009). inactivation. Cell 143, 390–403 (2010). 38. Chao, W., Huynh, K. D., Spencer, R. J., Davidow, L. S.
3. Disteche, C. M. Dosage compensation of the sex 22. Jeon, Y. & Lee, J. T. YY1 tethers Xist RNA to the & Lee, J. T. CTCF, a candidate trans-acting factor for
chromosomes. Annu. Rev. Genet. 46, 537–560 inactive X nucleation center. Cell 146, 119–133 X‑inactivation choice. Science 295, 345–347
(2012). (2011). (2002).
4. Lee, J. T. Gracefully ageing at 50, X‑chromosome 23. Clemson, C. M., McNeil, J. A., Willard, H. F. & 39. Monkhorst, K., Jonkers, I., Rentmeester, E.,
inactivation becomes a paradigm for RNA and Lawrence, J. B. XIST RNA paints the inactive Grosveld, F. & Gribnau, J. X inactivation counting
chromatin control. Nat. Rev. Mol. Cell Biol. 12, X chromosome at interphase: evidence for a novel RNA and choice is a stochastic process: evidence for
815–826 (2011). involved in nuclear/chromosome structure. J. Cell Biol. involvement of an X‑linked activator. Cell 132,
5. Wutz, A. Gene silencing in X‑chromosome inactivation: 132, 259–275 (1996). 410–421 (2008).
advances in understanding facultative 24. Simon, M. D. et al. High-resolution Xist binding maps 40. Sado, T., Wang, Z., Sasaki, H. & Li, E. Regulation of
heterochromatin formation. Nat. Rev. Genet. 12, reveal two-step spreading during X‑chromosome imprinted X‑chromosome inactivation in mice by Tsix.
542–553 (2011). inactivation. Nature 504, 465–469 (2013). Development 128, 1275–1286 (2001).
6. Costanzi, C. & Pehrson, J. R. Histone macroH2A1 is 25. Engreitz, J. M. et al. The Xist lncRNA exploits three- 41. Navarro, P. et al. Molecular coupling of Xist regulation
concentrated in the inactive X chromosome of female dimensional genome architecture to spread across the and pluripotency. Science 321, 1693–1695 (2008).
mammals. Nature 393, 599–601 (1998). X chromosome. Science 341, 1237973 (2013). 42. Gontan, C. et al. RNF12 initiates X‑chromosome
7. Heard, E. et al. Methylation of histone H3 at Lys‑9 26. Minajigi, A. et al. Chromosomes. A comprehensive Xist inactivation by targeting REX1 for degradation.
is an early mark on the X chromosome during X interactome reveals cohesin repulsion and an RNA- Nature 485, 386–390 (2012).
inactivation. Cell 107, 727–738 (2001). directed chromosome conformation. Science 349, 43. Shin, J. et al. Maternal Rnf12/RLIM is required for
8. Plath, K. et al. Role of histone H3 lysine 27 aab2276 (2015). imprinted X‑chromosome inactivation in mice. Nature
methylation in X inactivation. Science 300, 131–135 This work shows that in mice, the Xi has two 467, 977–981 (2010).
(2003). megadomains that are separated by the Dxz4 44. Shin, J. et al. RLIM is dispensable for X‑chromosome
9. Zhao, J., Sun, B. K., Erwin, J. A., Song, J. J. & Lee, J. T. boundary, and that Xist lncRNA shapes the 3D inactivation in the mouse embryonic epiblast. Nature
Polycomb proteins targeted by a short repeat RNA to organization of the Xi. The authors also suggest 511, 86–89 (2014).
the mouse X chromosome. Science 322, 750–756 that Xist interacts directly with cohesins and evicts 45. Barakat, T. S. et al. The trans-activator RNF12 and cis-
(2008). them from the Xi. acting elements effectuate X chromosome inactivation
10. Mira-Bontenbal, H. & Gribnau, J. New Xist-interacting 27. McHugh, C. A. et al. The Xist lncRNA interacts directly independent of X‑pairing. Mol. Cell 53, 965–978
proteins in X‑chromosome inactivation. Curr. Biol. 26, with SHARP to silence transcription through HDAC3. (2014).
R338–R342 (2016). Nature 521, 232–236 (2015). 46. Chow, J. C., Hall, L. L., Clemson, C. M., Lawrence, J. B.
11. Lee, J. T., Strauss, W. M., Dausman, J. A. & 28. Chu, C. et al. Systematic discovery of Xist RNA binding & Brown, C. J. Characterization of expression at the
Jaenisch, R. A. 450 kb transgene displays properties proteins. Cell 161, 404–416 (2015). human XIST locus in somatic, embryonal carcinoma,
of the mammalian X‑inactivation center. Cell 86, 29. Zhang, L. F., Huynh, K. D. & Lee, J. T. Perinucleolar and transgenic cell lines. Genomics 82, 309–322
83–94 (1996). targeting of the inactive X during S phase: evidence (2003).
12. Lee, J. T., Lu, N. & Han, Y. Genetic analysis of the for a role in the maintenance of silencing. Cell 129, 47. Johnston, C. M., Newall, A. E., Brockdorff, N. &
mouse X inactivation center defines an 80‑kb 693–706 (2007). Nesterova, T. B. Enox, a novel gene that maps 10 kb
multifunction domain. Proc. Natl Acad. Sci. USA 96, This paper describes the perinucleolar targeting of upstream of Xist and partially escapes X inactivation.
3836–3841 (1999). the Xi during S phase and shows that is dependent Genomics 80, 236–244 (2002).
13. Simmler, M. C. et al. A 94 kb genomic sequence on Xist. 48. Ostendorff, H. P. et al. Ubiquitination-dependent
3′ to the murine Xist gene reveals an AT rich region 30. Giorgetti, L. et al. Structural organization of the cofactor exchange on LIM homeodomain transcription
containing a new testis specific gene Tsx. Hum. Mol. inactive X chromosome in the mouse. Nature 535, factors. Nature 416, 99–103 (2002).
Genet. 5, 1713–1726 (1996). 575–579 (2016). 49. Wang, F. et al. Regulation of X‑linked gene expression
14. Chureau, C. et al. Ftx is a non-coding RNA which These authors find that Dxz4 deletion on the Xi in during early mouse development by Rlim. eLife 5,
affects Xist expression and chromatin structure within mouse cells induces a disruption in the formation of e19127 (2016).
the X‑inactivation center region. Hum. Mol. Genet. 20, the two megadomains. 50. Donohoe, M. E., Silva, S. S., Pinter, S. F., Xu, N. &
705–718 (2011). 31. Splinter, E. et al. The inactive X chromosome adopts a Lee, J. T. The pluripotency factor Oct4 interacts with
15. Jonkers, I. et al. RNF12 is an X‑encoded dose- unique three-dimensional conformation that is Ctcf and also controls X‑chromosome pairing and
dependent activator of X chromosome inactivation. dependent on Xist RNA. Genes Dev. 25, 1371–1383 counting. Nature 460, 128–132 (2009).
Cell 139, 999–1011 (2009). (2011). 51. Xu, N., Tsai, C.‑l. & Lee, J. T. Transient homologous
16. Chureau, C. et al. Comparative sequence analysis of 32. Hall, L. L. et al. An ectopic human XIST gene can chromosome pairing marks the onset of X inactivation.
the X‑inactivation center region in mouse, human, induce chromosome inactivation in postdifferentiation Science 311, 1149–1152 (2006).
and bovine. Genome Res. 12, 894–908 (2002). human HT‑1080 cells. Proc. Natl Acad. Sci. USA 99, This paper describes the importance of X–X pairing
17. Sun, S. et al. Jpx RNA activates Xist by evicting CTCF. 8677–8682 (2002). for XCI initiation.
Cell 153, 1537–1551 (2013). 33. Kelsey, A. D. et al. Impact of flanking chromosomal 52. Bacher, C. P. et al. Transient colocalization of
Sun et al. show that Jpx allows Xist expression by sequences on localization and silencing by the human X‑inactivation centres accompanies the initiation of
alleviating the repressive action of CTCF on its non-coding RNA XIST. Genome Biol. 16, 208 (2015). X inactivation. Nat. Cell Biol. 8, 293–299 (2006).
promoter. 34. Gartler, S. M. & Riggs, A. D. Mammalian In this publication, the authors observed that X–X
18. Lee, J. T. & Lu, N. Targeted mutagenesis of Tsix leads X‑chromosome inactivation. Annu. Rev. Genet. 17, pairing occurs at the initiation of XCI.
to nonrandom X inactivation. Cell 99, 47–57 (1999). 155–190 (1983). 53. Xu, N., Donohoe, M. E., Silva, S. S. & Lee, J. T.
19. Lee, J. T. Regulation of X‑chromosome counting by 35. Lyon, M. F. X‑Chromosome inactivation and Evidence that homologous X‑chromosome pairing
Tsix and Xite sequences. Science 309, 768–771 developmental patterns in mammals. Biol. Rev. Camb. requires transcription and Ctcf protein. Nat. Genet.
(2005). Philos. Soc. 47, 1–35 (1972). 39, 1390–1396 (2007).

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

54. Masui, O. et al. Live-cell chromosome dynamics 75. Rego, A., Sinclair, P. B., Tao, W., Kireev, I. & sequencing in mouse. Genome Res. 20, 614–622
and outcome of X chromosome pairing events Belmont, A. S. The facultative heterochromatin of the (2010).
during ES cell differentiation. Cell 145, 447–458 inactive X chromosome has a distinctive condensed 98. Li, S. M. et al. Transcriptome-wide survey of mouse
(2011). ultrastructure. J. Cell Sci. 121, 1119–1127 (2008). CNS-derived cells reveals monoallelic expression
55. Lee, J. T. Homozygous Tsix mutant mice reveal a 76. Smeets, D. et al. Three-dimensional super-resolution within novel gene families. PLoS ONE 7, e31751
sex-ratio distortion and revert to random microscopy of the inactive X chromosome territory (2012).
X‑inactivation. Nat. Genet. 32, 195–200 (2002). reveals a collapse of its active nuclear compartment 99. Calabrese, J. M. et al. Site-specific silencing of
56. Kung, J. T. et al. Locus-specific targeting to the harboring distinct Xist RNA foci. Epigenetics Chromatin regulatory elements as a mechanism of X inactivation.
X chromosome revealed by the RNA interactome of 7, 8 (2014). Cell 151, 951–963 (2012).
CTCF. Mol. Cell 57, 361–375 (2015). In this work, the authors show that the Xi displays These authors find that Xi‑silencing genes can be
In this publication, CTCF is shown to interact with a sponge-like structure, in which escapee genes are located and maintained outside the Xi territory and
Tsix RNA and to be a key player in X–X pairing. distributed throughout the Xi territory. that transcription can occur inside the Xi.
57. Augui, S. et al. Sensing X chromosome pairs before 77. Cremer, T. et al. The 4D nucleome: evidence for a 100. Marks, H. et al. Dynamics of gene silencing during X
X inactivation via a novel X‑pairing region of the Xic. dynamic nuclear landscape based on co‑aligned active inactivation using allele-specific RNA-seq. Genome Biol.
Science 318, 1632–1636 (2007). and inactive nuclear compartments. FEBS Lett. 589, 16, 149 (2015).
58. Sun, S., Fukue, Y., Nolen, L., Sadreyev, R. & Lee, J. T. 2931–2943 (2015). 101. Dietzel, S. et al. The 3D positioning of ANT2 and
Characterization of Xpr (Xpct) reveals instability but no 78. Nagano, T. et al. Single-cell Hi‑C reveals cell‑to‑cell ANT3 genes within female X chromosome territories
effects on X‑chromosome pairing or Xist expression. variability in chromosome structure. Nature 502, correlates with gene activity. Exp. Cell Res. 252,
Transcription 1, 46–56 (2010). 59–64 (2013). 363–375 (1999).
59. Thatcher, K. N., Peddada, S., Yasui, D. H. & 79. Lieberman-Aiden, E. et al. Comprehensive mapping 102. Namekawa, S. H., Payer, B., Huynh, K. D., Jaenisch, R.
Lasalle, J. M. Homologous pairing of 15q11–13 of long-range interactions reveals folding principles of & Lee, J. T. Two-step imprinted X inactivation: repeat
imprinted domains in brain is developmentally the human genome. Science 326, 289–293 (2009). versus genic silencing in the mouse. Mol. Cell. Biol.
regulated but deficient in Rett and autism samples. 80. Deng, X. et al. Bipartite structure of the inactive 30, 3187–3205 (2010).
Hum. Mol. Genet. 14, 785–797 (2005). mouse X chromosome. Genome Biol. 16, 152 (2015). 103. Chaumeil, J., Le Baccon, P., Wutz, A. & Heard, E.
60. Hewitt, S. L. et al. RAG‑1 and ATM coordinate 81. Nora, E. P. et al. Spatial partitioning of the regulatory A novel role for Xist RNA in the formation of a
monoallelic recombination and nuclear positioning of landscape of the X‑inactivation centre. Nature 485, repressive nuclear compartment into which genes are
immunoglobulin loci. Nat. Immunol. 10, 655–664 381–385 (2012). recruited when silenced. Genes Dev. 20, 2223–2237
(2009). 82. Rao, S. S. P. et al. A 3D map of the human genome at (2006).
61. Hogan, M. S., Parfitt, D. E., Zepeda-Mendoza, C. J., kilobase resolution reveals principles of chromatin 104. Chow, J. C. et al. LINE‑1 activity in facultative
Shen, M. M. & Spector, D. L. Transient pairing of looping. Cell 159, 1665–1680 (2014). heterochromatin formation during X chromosome
homologous Oct4 alleles accompanies the onset of In this paper, the authors show that in human cells, inactivation. Cell 141, 956–969 (2010).
embryonic stem cell differentiation. Cell Stem Cell 16, the Xi is folded into two megadomains separated These authors show that the Xi is formed by a
275–288 (2015). by the DXZ4 boundary. This work also finds that Xi transcriptionally silent core, marked by Xist, and
62. Zorca, C. E. et al. Myosin VI regulates gene pairing and exhibits superloops, among which DXZ4 and FIRRE that escapees are located exterior to this core.
transcriptional pause release in T cells. Proc. Natl interact. 105. Clemson, C. M., Hall, L. L., Byron, M., McNeil, J. &
Acad. Sci. USA 112, E1587–E1593 (2015). 83. Wang, S. et al. Spatial organization of chromatin Lawrence, J. B. The X chromosome is organized into a
63. Okamoto, I. et al. Eutherian mammals use diverse domains and compartments in single chromosomes. gene-rich outer rim and an internal core containing
strategies to initiate X‑chromosome inactivation Science 353, 598–602 (2016). silenced nongenic sequences. Proc. Natl Acad. Sci.
during development. Nature 472, 370–374 84. Darrow, E. M. et al. Deletion of DXZ4 on the human USA 103, 7688–7693 (2006).
(2011). inactive X chromosome alters higher-order genome These authors show that the internal core of the Xi
64. Petropoulos, S. et al. Single-cell RNA-seq reveals architecture. Proc. Natl Acad. Sci. USA 113, is composed of non-genic sequences and that the
lineage and X chromosome dynamics in human E45504–E45512 (2016). outer rim is formed by gene-rich regions regardless
preimplantation embryos. Cell 165, 1012–1026 This paper shows that deletion of DXZ4 in human of their transcriptional activity.
(2016). cells leads to the disappearance of the two 106. Filippova, G. N. et al. Boundaries between
65. Theunissen, T. W. et al. Molecular criteria for defining megadomains of the Xi. chromosomal domains of X inactivation and escape
the naive human pluripotent state. Cell Stem Cell 19, 85. Horakova, A. H., Moseley, S. C., Mclaughlin, C. R., bind CTCF and lack CpG methylation during early
502–515 (2016). Tremblay, D. C. & Chadwick, B. P. The macrosatellite development. Dev. Cell 8, 31–42 (2005).
66. Vallot, C. et al. XACT, a long noncoding transcript DXZ4 mediates CTCF-dependent long-range 107. Goto, Y. & Kimura, H. Inactive X chromosome-specific
coating the active X chromosome in human intrachromosomal interactions on the human inactive histone H3 modifications and CpG hypomethylation
pluripotent cells. Nat. Genet. 45, 239–241 (2013). X chromosome. Hum. Mol. Genet. 21, 4367–4377 flank a chromatin boundary between an X‑inactivated
67. Vallot, C. et al. XACT noncoding RNA competes with (2012). and an escape gene. Nucleic Acids Res. 37,
XIST in the control of X chromosome activity during 86. Chen, C. Y. et al. YY1 binding association with sex- 7416–7428 (2009).
human early development. Cell Stem Cell 20, biased transcription revealed through X‑linked 108. Horakova, A. H. et al. The mouse DXZ4 homolog
102–111 (2017). transcript levels and allelic binding analyses. Sci. Rep. retains Ctcf binding and proximity to Pls3 despite
68. Makhlouf, M. et al. A prominent and conserved role 6, 37324 (2016). substantial organizational differences compared to
for YY1 in Xist transcriptional activation. Nat. 87. Tang, Z. et al. CTCF-mediated human 3D genome the primate macrosatellite. Genome Biol. 13, R70
Commun. 5, 4878 (2014). architecture reveals chromatin topology for (2012).
69. Chapman, A. G., Cotton, A. M., Kelsey, A. D. & transcription. Cell 163, 1611–1627 (2015). In this work, the authors show that the structure
Brown, C. J. Differentially methylated CpG island 88. Yang, F. et al. The lncRNA Firre anchors the inactive and organization of Dxz4 in mice and DXZ4 in
within human XIST mediates alternative P2 X chromosome to the nucleolus by binding CTCF and humans are different.
transcription and YY1 binding. BMC Genet. 15, 89 maintains H3K27me3 methylation. Genome Biol. 16, 109. Chadwick, B. P. DXZ4 chromatin adopts an opposing
(2014). 52 (2015). conformation to that of the surrounding chromosome
70. Jonkers, I. et al. Xist RNA is confined to the nuclear 89. Ding, Z. et al. Quantitative genetics of CTCF binding and acquires a novel inactive X‑specific role involving
territory of the silenced X chromosome throughout reveal local sequence effects and different modes of CTCF and antisense transcripts. Genome Res. 18,
the cell cycle. Mol. Cell. Biol. 28, 5583–5594 X‑chromosome association. PLoS Genet. 10, 1259–1269 (2008).
(2008). e1004798 (2014). 110. Chadwick, B. P. & Willard, H. F. Multiple spatially
71. Bischoff, A. et al. Differences of size and shape of 90. Berletch, J. B. et al. Escape from X inactivation distinct types of facultative heterochromatin on the
active and inactive X‑chromosome domains in human varies in mouse tissues. PLoS Genet. 11, e1005079 human inactive X chromosome. Proc. Natl Acad. Sci.
amniotic fluid cell nuclei. Microsc. Res. Tech. 25, (2015). USA 101, 17450–17455 (2004).
68–77 (1993). 91. Yao, H. et al. Mediation of CTCF transcriptional 111. Hacisuleyman, E. et al. Topological organization of
72. Eils, R. et al. Three-dimensional reconstruction of insulation by DEAD-box RNA-binding protein p68 and multichromosomal regions by the long intergenic
painted human interphase chromosomes: active and steroid receptor RNA activator SRA. Genes Dev. 24, noncoding RNA Firre. Nat. Struct. Mol. Biol. 21,
inactive X chromosome territories have similar 2543–2555 (2010). 198–206 (2014).
volumes but differ in shape and surface structure. 92. Li, W. et al. Functional roles of enhancer RNAs for 112. Tsai, C. L., Rowntree, R. K., Cohen, D. E. & Lee, J. T.
J. Cell Biol. 135, 1427–1440 (1996). oestrogen-dependent transcriptional activation. Higher order chromatin structure at the X‑inactivation
This paper shows that the Xi territory is Nature 498, 516–520 (2013). center via looping DNA. Dev. Biol. 319, 416–425
characterized by a spherical shape and smooth 93. Zhao, X. et al. CTCF cooperates with noncoding RNA (2008).
surface. MYCNOS to promote neuroblastoma progression This paper describes that the Xic is partitioned into
73. Teller, K. et al. A top-down analysis of Xa‑ and through facilitating MYCN expression. Oncogene 35, two distinct TADs.
Xi‑territories reveals differences of higher order 3565–3576 (2016). 113. Giorgetti, L. et al. Predictive polymer modeling reveals
structure at ≥20 Mb genomic length scales. Nucleus 94. Saldaña-Meyer, R. et al. CTCF regulates the human coupled fluctuations in chromosome conformation and
2, 465–477 (2011). p53 gene through direct interaction with its natural transcription. Cell 157, 950–963 (2014).
74. Naughton, C., Sproul, D., Hamilton, C. & Gilbert, N. antisense transcript, Wrap53. Genes Dev. 28, 114. Spencer, R. J. et al. A boundary element between Tsix
Analysis of active and inactive X chromosome 723–734 (2014). and Xist binds the chromatin insulator Ctcf and
architecture reveals the independent organization of 95. Balaton, B. P. & Brown, C. J. Escape artists of the contributes to initiation of X‑chromosome inactivation.
30 nm and large-scale chromatin structures. Mol. Cell X chromosome. Trends Genet. 32, 348–359 (2016). Genetics 189, 441–454 (2011).
40, 397–409 (2010). 96. Berletch, J. B., Yang, F. & Disteche, C. M. Escape from 115. Barr, M. L. & Bertram, E. G. A morphological
These authors find that the compaction of the Xi is X inactivation in mice and humans. Genome Biol. 11, distinction between neurones of the male and female,
only 1.2‑fold higher than that of the Xa. This paper 213 (2010). and the behaviour of the nucleolar satellite during
also argues for a non-monotonous compaction of 97. Yang, F., Babak, T., Shendure, J. & Disteche, C. M. accelerated nucleoprotein synthesis. Nature 163, 676
the Xi. Global survey of escape from X inactivation by RNA- (1949).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrg


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

116. Bourgeois, C. A., Laquerriere, F., Hemon, D., Nat. Rev. Mol. Cell Biol. 11, 317–328 123. Zuin, J. et al. Cohesin and CTCF differentially affect
Hubert, J. & Bouteille, M. New data on the in situ (2010). chromatin architecture and gene expression in human
position of the inactive X chromosome in the 120. Yusufzai, T. M., Tagami, H., Nakatani, Y. & cells. Proc. Natl Acad. Sci. USA 111, 996–1001 (2014).
interphase nucleus of human fibroblasts. Hum. Genet. Felsenfeld, G. CTCF tethers an insulator to
69, 122–129 (1985). subnuclear sites, suggesting shared insulator Acknowledgements
117. Dyer, K. A., Canfield, T. K. & Gartler, S. M. Molecular mechanisms across species. Mol. Cell 13, The authors thank M.-E. Gilles, Y. Jeon and F. Ariel for many
cytological differentiation of active from inactive 291–298 (2004). inspirational discussions. T.J. is a European Molecular Biology
X domains in interphase: implications for 121. Branco, M. R. & Pombo, A. Intermingling of Organization (EMBO) postdoctoral fellow (EMBO ALTF 1313–
X chromosome inactivation. Cytogenet. Cell Genet. 50, chromosome territories in interphase suggests 2015); E.A. is funded by the Swiss National Science Foundation.
116–120 (1989). role in translocations and transcription-dependent J.T.L. is funded by the US National Institutes of Health
118. Padeken, J. & Heun, P. Nucleolus and nuclear associations. PLoS Biol. 4, 780–788 (2006). (R37‑GM58839) and is an investigator of the Howard Hughes
periphery: Velcro for heterochromatin. Curr. Opin. Cell 122. Dixon, J. R. et al. Topological domains in Medical Institute.
Biol. 28, 54–60 (2014). mammalian genomes identified by analysis of
119. Mekhail, K. & Moazed, D. The nuclear envelope chromatin interactions. Nature 485, 376–380 Competing interests statement
in genome organization, expression and stability. (2012). The authors declare no competing interests.

NATURE REVIEWS | GENETICS ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like