You are on page 1of 13

Nature Reviews Molecular Cell Biology | AOP, published online 11 March 2015; doi:10.

1038/nrm3965 REVIEWS

Three-dimensional genome
architecture: players and mechanisms
Ana Pombo1 and Niall Dillon2
Abstract | The different cell types of an organism share the same DNA, but during cell
differentiation their genomes undergo diverse structural and organizational changes that
affect gene expression and other cellular functions. These can range from large-scale folding
of whole chromosomes or of smaller genomic regions, to the re-organization of local
interactions between enhancers and promoters, mediated by the binding of transcription
factors and chromatin looping. The higher-order organization of chromatin is also influenced
by the specificity of the contacts that it makes with nuclear structures such as the lamina.
Sophisticated methods for mapping chromatin contacts are generating genome-wide data
that provide deep insights into the formation of chromatin interactions, and into their roles
in the organization and function of the eukaryotic cell nucleus.

Chromatin The 2-metre length of DNA in a mammalian cell is more than 30years ago, it has become clear that this type
immunoprecipitation organized into chromosomes, which are packaged and of genetic element can regulate gene transcription over
A method in which chromatin folded through various mechanisms and occupy discrete large distances. Looping out of the DNA that separates
bound by a protein is
positions in the nucleus. The multiple levels of DNA promoters and distally located enhancers was proposed
immunoprecipitated with an
antibody against that protein,
folding generate extensive contacts between different as a mechanism by which factors that are bound to
to allow the extraction and genomic regions. These contacts are influenced by the enhancers can directly contact their target promoters
analysis of the bound DNA proximity of DNA sequences to one another, by the fold- and influence the composition of transcription initiation
byquantitative PCR or ing architecture of local and long-range chromatin con- complexes (FIG.1a). Evidence supporting looping models
genome-wide sequencing.
tacts and by proteins that associate directly or indirectly was initially obtained by fluorescence insitu hybridiza-
with the DNA. Packing of chromosomes in the nucleus tion (FISH) and by measuring the effects of genomic
also brings them into contact with one another and with distance on enhancer function1,2.
nuclear compartments, such as the nucleolus and the In this Review, we discuss how various cellular fac-
nuclear envelope. As cells progress through the cell cycle tors are thought to contribute to the formation of higher-
and as they differentiate into specialized cell types, their order chromatin structures such as active chromatin
1
Epigenetic Regulation and
chromosomes undergo structural reorganizations that hubs (ACHs) and lamin-associated domains (LADs).
Chromatin Architecture
Group, Berlin Institute for influence gene expression and cell behaviour and func- We also describe the organization of the nucleus into
Medical Systems Biology, tion, which in turn modulate the organization of con- distinct compartments and chromosome territories. We
Max Delbrck Center for tacts between chromosomal regions. Techniques such as then discuss evidence that genome architecture contrib-
Molecular Medicine, chromatin immunoprecipitation followed by deep sequenc- utes to the regulation of cellular functions such as the cell
RobertRoessle Strasse,
13125 Berlin-Buch, Germany.
ing (ChIP-seq) have yielded large amounts of data on cycle, differentiation and senescence.
2
Gene Regulation and transcription factor binding and post-translational his-
Chromatin Group, tone modifications in a range of cell types. Increasingly The organization of chromosomes
MRCClinical Sciences Centre, sophisticated methods of chromatin conformation The development of the 3C3 technique and its subse-
Imperial College London,
capture (3C) are also being used to map genome-wide quent maturation into chromosome conformation cap-
Hammersmith Hospital
Campus, Du Cane Road, chromatin contacts. These approaches are enabling the ture on chip (4C)4, 3C combined with high-throughput
London W12 0NN, UK. study of how different cellular functions are influenced sequencing (4Cseq) 5, multiplexed 3C sequencing
e-mails: by the constraints of chromatin and chromosomal (3Cseq)6, carbon-copy chromosome conformation
ana.pombo@mdc-berlin.de; organization. capture (5C)7, capture-C8, genome conformation cap-
niall.dillon@csc.mrc.ac.uk
doi:10.1038/nrm3965
Contacts between chromatin regions are increasingly ture (GCC)9, Hi-C10, tethered conformation capture
Published online thought to have important roles in gene regulation mech- (TCC)11 and targeted chromatin capture (T2C)12 (BOX1)
11 March 2015 anisms. In particular, since the discovery of enhancers have been critical for the rapid progress in the study of

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 1

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a is located 50kb away, are brought into close contact in


Gene fetal liver cells, where the globin gene is expressed, but
not in fetal brain cells, where it is not. The interaction
TF Promoter sites in the active mouse globin locus are clustered
TF Pol II
Enhancer and are thought to form an ACH13, where multiple con-
tacts occur simultaneously (FIG.1b). ACH-type contacts
have also been described in the human RUNX1 (also
b DNA
known as AML1)17 and CFTR loci18 and in the mouse
Gene Cd8 locus19.
Enhancer Enhancer Clustered enhancers
Formation of the hub structure in the mouse globin
locus precedes activation of the globin genes in
Looping of the DNA
through contact between
erythroid progenitor cells. The core ACH includes the
the promoter and enhancers DNaseI hypersensitive sites of the LCR and additional
ACH hypersensitive sites at the 5 and 3 ends of the locus.
The embryonic and adult globin genes are recruited
to the ACH as they become sequentially activated dur-
c 1.0 ing mouse development 20. Formation of the complete
Polymer
ACH requires the presence of the erythroid-specific
Binder transcription factors GATA1 and Krppel-like factor1
(Rg2/Rg2 (random))

(KLF1; also known as EKLF). The cofactor LIM domain-


Gyration radius

0.6 binding 1 (LDB1) binds to both the globin LCR and


Binding
sites the globin promoter in erythroid cells and promotes
Ctr contact between them. LDB1 is part of a complex con-
taining the transcription factors GATA1 and T cell acute
0.2 lymphocytic leukaemia 1 (TAL1) together with another
5 10 25 cofactor, LIM domain-only 2 (LMO2; also known as
Binder concentration
(cm; nmol l1) rhombotin 2)21. Tethering LDB1 to the globin pro-
Figure 1 | Chromosome organization. a | Direct contact between enhancers and moter using artificial zinc fingers enhanced the for-
promoters occurs through looping out of intervening genomic
Nature Reviews sequences. RNA
| Molecular Cell Biology mation of a chromatin loop between the LCR and the
polymerase II (Pol II) has been shown to bind to enhancers in conjunction with promoter in GATA1deficient cells, with an accompa-
transcription factors (TFs) before activation of transcription. b | Proposed structure of an nying increase in transcription22. The same effect was
active chromatin hub (ACH). The ACH structure is formed through direct contacts achieved by tethering the LDB1 self-association domain,
between multiple enhancers and the promoter of their target gene through looping which promotes the multimerization of the LDB1
outof intervening sequences. Coloured ovals represent factors bound to enhancers. complex 22.
c|Thestrings and binders switch (SBS) model considers a simple scenario in which The ACH hypothesis has considerable implications
chromatin conformations arise by interactions of specific binding sites across the for our understanding of how chromosomal regions
polymer with randomly diffusing binding factors. SBS modelling shows that polymer loop
that contain active genes are organized in the interphase
formation displays a switch-like response to changes in the concentration of binding
factors. Gyration radius is a measure of the average polymer compaction. Cm, nucleus (FIG.1b). ChIP-seq data for histone modifica-
concentration of binding factors; Ctr, concentration of binding factors at threshold tions and transcription factors associated with enhancer
between open and closed polymer states. Figure, part c, is modified from REF.113, PNAS. function indicate that enhancers are present at variable
distances around highly expressed genes in many cell
types23. Clustering of enhancers into ACHs would be
chromatin contacts, allowing high-throughput map- expected to greatly affect the folding of these chroma-
ping of putative contacts across different genomic dis- tin regions, through the formation of large constrained
tances and between chromosomes, with varying levels loops. ACHs would be established initially through the
of resolution. The 3Cbased methods involve crosslink- action of pioneering factors that can access condensed
ing and ligation of interacting regions, after cell frag- chromatin, or facilitated by opening of the chromatin
mentation by restriction enzyme digestion, followed by during the S phase of the cellcycle.
quantification of ligated products, which are interpreted The formation of ACHs seems to be a property of
aschromatin contacts. They have been used to measure LCRs, which are functionally defined as transcription-
chromatin contact frequencies that correlate well with activating sequences that are dominant over chromatin
functional studies of regulatory elements in several loci position effects when randomly inserted into different
and support looping models1316. However, it should be parts of the genome, and often contain multiple elements
noted that important caveats have been raised about the that would be expected to adopt an ACH-type configu-
nature of the contacts that are detected by ligation-based ration2426. This suggests that ACH formation could, at
methods912 (BOX2), which should be kept in mind when least in some cases, create folding structures that are
interpreting results obtained with these techniques. dominant over other, more generic types of chromatin
folding. Arranging single elements from LCRs into tan-
The formation of active chromatin hubs. Early 3Cbased dem repeats in transgenic mice was shown to recreate
analyses indicated that the erythroid-specific mouse the dominant LCR transcriptional effect 25,27, suggesting
globin gene and its locus control region (LCR), which that contacts driven by multiple transcription factors,

2 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Summary of currently used chromosome conformation capture-based techniques


Chromosome conformation capture (3C)-based techniques involve the preparation of chromatin after mild formaldehyde
crosslinking, followed by sonication and/or digestion with restriction enzymes, and ligation of digested DNA fragments
attached to chromatin remnants. The ligation products are captured by a variety of approaches and amplified by PCR or
sequenced using unbiased next-generation sequencing methods. More extensive technical reviews of 3Cbased methods
can be found in REFS105107.

Technique Detection of chromatin contacts Refs


3C PCR amplification using primers of two known (biased) genomic 3
regions of interest
Chromosome conformation capture PCR amplification of genome-wide ligation products using one known 4
on chip (4C) (biased) genomic region of interest, followed by microarray analysis
Chromosome conformation capture Bait-specific library amplification from circularized 3C products using 108
combined with high-throughput adapter-containing primers, followed by high-throughput sequencing
sequencing (4Cseq)
Multiplexed 3C sequencing Bait-specific library amplification from circularized 3C products using 6
(3Cseq) adapter-containing primers; uses restriction enzymes that recognize
4-bp sequences for finer detection of chromatin contacts
Carbon-copy chromosome Ligation and PCR similar to 3C; a collection of PCR primers covering 7
conformation capture (5C) a whole genomic region are annealed to 3C products and ligated to
produce a carbon copy of a section of the 3C library; amplification and
sequencing of the ligated primers generates a high-coverage map of
interactions in a single region
Capture C and targeted chromatin After ligation, DNA fragments are ligated to sequencing primers; 8,12
capture (T2C) genomic regions of interest are captured by bead or array technologies
and subjected to paired-end sequencing
Genome conformation capture The first genome-wide unbiased 3C-based approach: global 9
(GCC) sequencing of the 3C material simultaneously measures the frequency
of ligation events and the presence or absence of each genomic
fragment; achieving good contact frequency signals in larger genomes
is expensive, but the technique allows normalization of contact
frequencies relative to the detectability and copy number of each DNA
fragment
Hi-C Ligation products are tagged with biotinylated nucleotides; after 10
crosslink reversal, biotinylated fragments are pulled down to enrich for
ligation products, prior to paired-end genome-wide sequencing
Tethered conformation capture Biotinylation of chromatin proteins is performed before restriction 11
(TCC) enzyme digestion
Immobilization of the biotinylated chromatin on streptavidin beads
removes non-crosslinked DNA
DNA overhangs are filled with an exonuclease-resistant nucleotide
analogue containing -thio-triphosphate and with a biotinylated
nucleotide analogue to generate blunt ends
After blunt-end ligation, crosslinking is reversed and DNA is purified
Non-ligated DNA ends are digested with Escherichiacoli exonuclease
III, DNA is sheared and biotinylated ligation products are purified as
for Hi-C and subjected to high-throughput sequencing

irrespective of the factors involved, can impose altera- clustering around a promoter. For example, in the KIT
tions in chromatin organization by establishing ACH- locus, distally located sequences contact the promoter
type loops. Results obtained by polymer modelling early in erythropoiesis and enhance transcription16.
support the idea that chromatin looping promoted by Atlater stages of differentiation, the promoter forms
multivalent binders can have a switch-like behaviour (on contacts with downstream intronic sequences, leading to
or off ) that is able to translate a range of transcription repression. In the MYB locus, contact between upstream
factor concentrations or affinities into a single, robust enhancers and an intronic element seems to promote
chromatin folding behaviour, which can be open or inter- transcriptional elongation15.
acting (FIG.1c). A discussion of approaches for character- A high proportion of the genes in vertebrate genomes
izing the functional effects of chromatin contacts can be are located in large gene-dense regions. It is possible that
found in Supplementary informationS1 (Box). genes located in these regions might form different con-
There is still much to be learned about con- tacts from the ACH structure that is observed in loci
tacts between enhancers, promoters and silencers. such as the globin locus, which are relatively distant
Contacts can be highly flexible and diverse rather from other active genes in the cells in which they are
than following a single pattern of groups of enhancers expressed. To address this question, the 4C technique

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 3

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Interpreting 3C data and using them to model chromatin folding


Cohesin has a crucial role in establishing sister
chromatid cohesion during S phase and maintaining it
The precise nature of the chromatin contacts detected by chromosome conformation through G2 and mitosis. The core cohesin complex is
capture (3C)-based techniques is unclear: they were initially thought to quantify made up of a heterodimer of the proteins SMC1 (struc-
contacts between distant DNA sequences brought into close proximity by tural maintenance of chromosomes 1) and SMC3, in
sequence-specific protein bridges. However, recent studies suggest that many of the
combination with SCC1 and SCC3. The complex forms
putative contacts could reflect varying degrees of proximity within larger nuclear
structures (reviewed in REF.109). A significant fraction of interactions detected by
a ring structure and is loaded onto DNA early in G1,
3Cbased techniques may result from ligation events that occur in the insoluble nuclear through a process that depends on the cohesin-loading
remnant of the chromatin preparation110. Electron microscopy of this remnant identifies complex NIPBLMAU2 (reviewed in REF.28).
nuclei that retain their typical morphology and have heterochromatin located in the In addition to its cell cycle functions, cohesin is
periphery. This suggests that many of the ligation events captured by 3Cbased involved in regulating gene expression through the
methods could be happening in the context of the higher-order organization of intact stabilization of long-range contacts between cis-acting
nuclei rather than isolated sonicated fragments that would reflect specific contacts110. regulatory elements. Cornelia de Lange syndrome,
These issues introduce a level of imprecision about exactly what is being measured by which is caused by mutations in NIPBL and the cohesin
3Cbased techniques, which undermines, for example, the unbiased identification from subunits SMC1A and SMC3, is associated with dys-
genome-wide datasets of regulatory regions that are thought to interact specifically
regulation of gene expression in many tissues28. A large
with individual promoters. Ligation events generated between sequences located close
to specific contacts and baseline ligation levels arising from random contacts between
number of cohesin-binding sites, mapped globally in
chromatin105 will add to the background noise. Ligation-based methods are also the mouse genome, colocalize with binding of CTCF29.
affected by inherent biases in restriction enzyme cutting efficiency, ligation frequency Cohesin-binding sites were shown to be important for
and the fact that the contacts detected represent the average of the chromatin states the organization and transcriptional regulation of the
of the individual cells in a population at the moment of crosslinking111. mouse interferon locus30 and for correct rearrange-
The development of effective mathematical models that use 3C data to predict the ment of the Tcell receptor locus31. The mechanisms
folding behaviours of single loci or whole chromosomes is likely to provide an important that allow cohesin to stabilize long-range interactions
complement to the experimental approaches, and should provide insights into the have not yet been elucidated but could involve the same
question of which chromatin contacts are most important for folding, for gene ring structure that maintains sister chromatid cohesion.
regulation and for chromosome architecture. Pioneering efforts to mathematically
CTCF was originally characterized as a factor that
model chromosome organization began by modelling chromatin folding using
approaches that reflect broad-scale organization of chromosomes, but have only
binds to the promoter of the chicken Myc gene32 and
recently started to take advantage of the more detailed information gained from functions as an activator or a repressor of transcription
chromatin contacts measurements by 3Cbased approaches (reviewed in REF.112). depending on the context. A major reason for the inter-
Further developments in the modelling of chromatin contacts will be particularly est in CTCF has been its association with insulators,
important for capturing and shedding light on the mechanisms of folding that underpin which are sequences that block the action of enhanc-
the variability in chromatin folding observed at the single cell level as well as in ers on promoters (reviewed in REF.33). The canonical
different chromatin states, at different stages of the cell cycle, and in different cell example is the insulin growth factor 2 (Igf2)H19 locus,
types and organisms11,113115. in which DNA methylation-sensitive binding of CTCF
is involved in regulating an insulator that controls the
epigenetic imprinting of the locus. However, long-range
was used to measure contacts between Rad23a, a interactions are unimpeded by the presence of CTCF-
housekeeping gene located in a gene-dense region on binding sites in 79% of loci where such sites are present
mouse chromosome 8, and other genomic regions between enhancers and promoters34. This suggests that
and to compare these with contacts made by HS2 of the relationship between CTCF and insulator effects is
the mouse globin LCR4. The analysis showed that more complex than was originally thought.
Rad23a preferentially interacts with many other gene- In addition to its well-established functions in
dense chromosomal regions, both locally and through enhancer blocking, CTCF is involved in promoting
trans interactions with other chromosomes, and that the long-range interactions that stimulate transcription.
interactions are largely conserved between the erythroid These include the interaction between the promoter of
cells in the fetal liver and the fetal brain. By contrast, the insulin gene and synaptotagmin 8 (SYT8), which
the globin gene interacts with other active erythroid- enhances SYT8 transcription in pancreatic islets35, and
specific genes in fetal liver cells, but with silent genes in the enhancerpromoter interactions that generate multi-
the fetalbrain. ple protocadherin isoforms on the surface of neurons36,37.
Overall, the evidence to date suggests that CTCF is
The roles of CTCF and cohesin in chromatin folding. multi-functional and can act as a gene activator or a
Asdiscussed above, current evidence supports a mech- silencer as well as a promoter of looping between distant
anism for action at a distance based on direct contacts regions. These different roles are likely to depend on the
between enhancers and promoters. These contacts bring association of CTCF with different binding partners.
together factors and co-factors that are bound to the It is also possible that the common feature of all three
contacting elements, thereby facilitating the formation functions is the ability of CTCF to promote interactions
and correct function of the transcription pre-initiation between distal sequences.
complex. In addition, another class of proteins has been
identified that promotes interactions between distally Topologically associating domains. Global analyses of
located sequences, the best-characterized being cohesin chromatin contacts revealed genome-wide patterns
and CCCTC-binding factor (CTCF). that suggest that chromatin contacts operate at different

4 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a 100
Normalized
interacting counts
0
Chromosome 6: 50,000,000 51,000,000 52,000,000 53,000,000 54,000,000
TADs:

Region containing predominantly


b Genomic region containing predominantly active tissue-specic genes
active tissue-specic genes Silent region

Housekeeping gene Region of cohesin binding


Tissue-specic gene Region of CTCF binding

Figure 2 | Topologically associating domains. a | HiC profiles reveal that the mammalian genome is organized into
topologically associating domains (TADs): regions that show high levels of interaction within the region and little or no
Nature Reviews | Molecular Cell Biology
interaction with neighbouring regions. The heat map represents normalized HiC interaction frequencies. b | Schematic of
putative TAD structures. The central regions of TADs show high levels of chromatin interaction and coincide with the
presence of tissue-specific genes and their associated enhancers, the interactions of which with their cognate promoters are
facilitated by the presence of cohesin and CCCTC-binding factor (CTCF). The border regions between TADs are enriched for
housekeeping genes, which are often clustered together and generally lack the widely dispersed distal enhancers that are
found around tissue-specific genes. The border regions show high levels of CTCF and cohesin binding, although only CTCF
seems to prevent interactions between TADs. Figure, part a, is reprinted from REF.38, Nature Publishing Group.

levels38,39. HiC analyses of mouse and human embry- cell-type specificities. This makes it unlikely that they
onic stem cells (ESCs) and human fibroblasts38, and of represent a general partitioning of the genome into func-
the chromosomal region around the mouse X inactiva- tional domains. However, there is evidence that bounda-
tion centre39, revealed that the genome contains large ries between TADs can demarcate functionally distinct
regions that are defined by high levels of chromatin regions of the genome in some cases. For example, the
interactions occurring within the region or domain, Hoxa locus is separated into an active and a repressed
interspersed with genomic regions with fewer interac- compartment by an insulator that binds CTCF 43
tions (FIG.2a). These regions are generally referred to as and was found to correspond to a TAD boundary in
topologically associating domains (TADs). A genome- differentiated cells38.
wide HiC analysis identified 2,200 TADs occupying Several recent studies have looked at the roles
91% of the mouse genome, with an average size of ofcohesin and CTCF in TAD organization44,45. In all of
880kb38. Interestingly, the boundaries between TADs these studies, knockout or depletion of cohesin reduced
are enriched for housekeeping genes (FIG.2b). Histone interactions within TADs but the TADs retained their
marks associated with enhancers, such as histone 3Lys4 basic shape. Two studies reported that reducing cohesin
monomethylation (H3K4me1), and with transcrip- levels did not lead to increased interaction between
tion repression, such as histone 3Lys9 trimethylation domains 44,45, whereas a third reported significant
(H3K9me3), were also enriched at TAD boundaries. increases in inter-domain interactions46. Depletion
The structural organization of the genome into TADs of CTCF had the same effect, but it also increased the
is maintained across different cell types and is partially levels of interactions between different TADs44,45. The
conserved between the human and mouse genomes38. different conclusions from these studies could relate
TADs have also been mapped by HiC in the Drosophila to differences between the cell types used. The study
melanogaster genome and have been found to have reporting increased inter-domain interactions46 ana-
many of the characteristics of vertebrate TADs4042. lysed interactions in differentiated astrocytes, which
D.melanogaster TADs are smaller than their vertebrate could have a different chromatin organization from the
counterparts, perhaps reflecting the smaller size ofthe non-cycling thymocytes44 or cycling embryonic kidney
D.melanogaster genome and the closer packing of genes cells45 used in the other two studies. These results suggest
within it. To date, TADs have not been identified in that cohesin and CTCF are primarily involved in pro-
Saccharomyces cerevisiae. moting promoterenhancer interactions but could also
The relatively large size of TADs38,39 means that they have some involvement in delineating the boundaries
mostly contain a large number of genes with a variety of betweenTADs.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 5

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | The organization of chromatin into functional compartments


Different factors can cluster Pol I transcription factories Pol II and Pol III transcription factories
together in the nucleus to form
functional nuclear compartments.
Well-characterized examples of
this include the clustering of
active DNA and RNA polymerases,
as well as chromatin regulators
such as the Polycomb group (PcG)
repressor complexes.
Theclustering of DNA Nucleolus
polymerases during replication in
discrete domains, termed
replication factories, is observed
in yeast, plants and
mammals116,117,72. Strikingly, in
mammalian cells, clustered origins Micrometres Tens of nanometres
of replication that initiate Nucleosomes DNA RNA polymerase
replication simultaneously can Nascent RNA Transcription factory
remain associated during several
rounds of cell divisions72.
RNA polymerases are also found in discrete domains called transcription factories (see the figure). RNA polymerase I
(Pol I) factories transcribe the 45S ribosomal RNA genes, which are clustered in the genome into groups of many tens of
Nature
copies. Pol I factories are found within nucleoli (see the figure, left), which in HeLa Reviewson
cells contain | Molecular
average ofCell
500 Biology
active
enzymes and about four 45S rRNA genes, each of which is transcribed simultaneously by approximately 125enzymes118,119.
Individual nucleoli bring together 45S rRNA genes from clusters present in different chromosomes, constituting one of
the first known examples of gene expression coordinated within a single structure (reviewed in REF.120). By contrast,
PolII and Pol III factories are more abundant but contain far fewer polymerases (see the figure, right). In HeLa cells there
are about 8,000 Pol II and about 2,000 Pol III factories, each containing approximately 6 to 8 active enzymes118,121,122.
Thenumber of factories per nucleus seems to scale with genome size, but their spatial density and size remain broadly
constant. For example, diploid mouse embryonic stem cells (ESCs) have 4,000 transcription factories. By contrast, the
salamander, the genome of which is 11 times larger than that of diploid mouse ESCs, has 33,000 transcription factories123.
The transcription factory model is further supported by observations that active genes associate with one, or a few, active
RNA polymerases124, and that single genes can be found coassociated at shared Pol II sites125,126.
Recent evidence suggests that Pol II transcription factories are functionally specialized according to the activation state
of the polymerase complexes that they contain. Transcription factories in a poised state contain the initiating form of
PolII (in which Ser5, but not Ser2, is phosphorylated in the carboxyterminal domain repeats)63 and are associated with
genes repressed by PcG complexes63,127. In HepG2 cells, UPA (urokinase-type plasminogen activator) is not expressed but
is found associated with poised factories63. Following transcriptional activation, UPA associates with active factories that
contain Pol II phosphorylated on both Ser5 and Ser2. Developmental genes repressed by PcG in ESCs, such as Nkx2-2,
Msx1 and HoxA7, are also found associated with poised factories127.
PcG bodies are another type of functional nuclear compartment, present in the nucleoplasm of Drosophila melanogaster
and mammalian cells. In D.melanogaster, Polycomb response elements (PREs) colocalize with PcG bodies in anterior
nuclei of the syncytial embryo128. In the posterior region of the embryo, inactive PREs such as bxd are also associated with
PcG bodies, whereas active PREs (such as Fab7) lose contact with these domains, supporting the idea that the association
with PcG bodies is important for gene silencing. The pairing of two Fab7 elements from different genomic regions, and
the coassociation of three PREs (Fab7, Mcp and bxd) within the BXC Hox gene locus, also occur within shared
Nuclear lamina PcGbodies128,129. Indeed, analyses of chromatin immunoprecipitation combined with chromosome conformation capture
A protein meshwork made of of a major Polycomb protein, enhancer of zeste 2 (Ezh2), demonstrated its presence at intra- and inter-chromosomal hubs
intermediate filaments (such as that are associated with gene silencing130.
lamins) and membrane-associ-
ated proteins (such as emerin)
that covers the inner nuclear
membrane and is responsible
for maintaining nuclear shape,
Overall, the data suggest that TADs are complex Interaction of chromosomes in the nucleus
organization and function. entities, with structures that are governed by several The specificity and frequency of chromatin contacts are
different parameters and that are potentially gener- affected by the organization of the nucleus into struc-
Heterochromatin ated by heterogeneous mechanisms. ACH formation tural and functional compartments, such as the nuclear
Highly condensed chromatin
through enhancerpromoter contacts is likely to con- lamina and various proteinaceous bodies (BOX3). The
that shows dark staining.
Constitutive heterochromatin tribute strongly to the contacts that are observed within behaviour of whole chromosomes also depends on
remains in this state TADs. The enrichment of housekeeping genes and the their size, the number and state of activity of genes that
throughout the cell cycle. depletion of the H3K4me1 enhancer mark at TAD they contain, and the distribution of heterochromatin.
Facultative heterochromatin is boundaries suggest that some aspects of TAD organi- Chromatin contacts and their functions are also likely
cell-type-specific condensed
chromatin that is often a
zation might be the product of differential distribution to be affected by the biophysical environment in which
feature of terminally of cell-type-specific and housekeeping genes along chromosomes exist. Important parameters include the
differentiated cells. chromosomes. nuclear shape, mechanical stress generated by cellular

6 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Cytoplasm
NPC Nuclear lamina ONM
INM

Chromatin
LADs cLADs
Nucleus
fLADs

Dierentiation

NADs

RNA Pol I
Nucleolus factories

Figure 3 | The nuclear envelope affects genome organization and function. Lamina-associated domains (LADs) are
regions of condensed chromatin that are bound by the nuclear lamina. LADs are also enriched for marks such as
Nature Reviews | Molecular Cell Biology
histone3 Lys9 dimethylation (H3K9me2), which is a mark of heterochromatin and silent genes. Regions of open
chromatin, in which genes are actively transcribed, loop out into the interior of the nucleus. As cells differentiate,
constitutive LADs (cLADs) remain associated with the lamina, whereas facultative LADs (fLADs) become detached as the
genes that they contain become active. After mitosis, some LADs relocate to the periphery of the nucleolus in the
following G1 phase of the cell cycle, although the mechanism underlying this remains unclear. Sequences preferentially
located at the nucleolar periphery (nucleolus-associated chromatin domains; NADs) have been identified independently
by high-throughput sequencing of DNA associated with biochemically isolated nucleoli. NADs are enriched for
pericentric satellite repeats, A- and T-rich sequences and gene-poor regions. INM, inner nuclear membrane;
NPC,nuclear pore complexes; ONM, outer nuclear membrane.

movements and the cell cycle. Below, we discuss some indeed, cells that are depleted of lamins or that express
of these features of nuclear organization. mutant lamins often have misshapen nuclei48. Lamins
also have important roles in many nuclear processes,
Nuclear and nucleolar peripheries organize chromo- including transcription, DNA replication, cell cycle
somes. The nuclear envelope is a membrane bilayer, control and DNA repair (reviewed in REF.49).
made up of the inner and outer nuclear membranes In most cell types, the nuclear periphery is associated
(FIG.3). Embedded in the envelope are the nuclear pores, with heterochromatin, whereas euchromatin is more
which are large and highly organized structures that found centrally located. A remarkable exception to this
span the two membranes and mediate transport in general feature is the central position of heterochroma-
both directions between the cytoplasm and the nucleus. tin and the peripheral position of euchromatin that is
The inner side of the nuclear envelope is coated with observed in the rod cells of the eye in nocturnal animals,
lamins and many other proteins. Together, they form the which is thought to be important for the light trans-
nuclear lamina, which is thought to have key roles in mission properties of rod cells (see below). Chromatin
the spatial organization of chromosomes, as it is a large attachments to the lamina have been mapped at high res-
surface that establishes contacts with chromatin while olution in D.melanogaster, mouse and human cells, using
remaining spatially constrained by its location on the DNAadeninemethyltransferaseidentification (DamID) or
inner wall of the nuclear envelope. ChIP-seq (reviewed in REF.50). Mapping of lamin B
Nuclear lamins are members of the intermediate chromatin interactions revealed that large genomic
filament protein family. They are found in all multi- regions, termed LADs50, are bound to the nuclear lamina
cellular animals but are absent from plants, fungi and (FIG.3). LADs vary in size (0.110Mb) and are strongly

DNAadeninemethyltrans- protozoa. They are divided into two types, lamin B enriched for H3K27me3 and, to a lesser degree, for
feraseidentification and lamins A and C. Type B lamins are present in all cell H3K9me2 (REF.51), which are indicative of Polycomb-
A method based on expression types; type A and C lamins are absent from ESCs, and repressed and heterochromatic sequences, respectively.
of fusion proteins with bacterial their expression increases as cells differentiate, although Two types of LADs have been identified: constitu-
Dam methylase, and detection
of methylated DNA as a
they are expressed at low levels in terminally differenti- tive LADs (cLADs) and facultative LADs (fLADs; FIG.3).
measure of its contact with ated neurons47. A key function of the nuclear lamins is to The genomic distribution of fLADs varies in different
thefusion protein. maintain the shape and mechanical properties of nuclei; cell types, whereas cLADs, which are A-T rich, are

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 7

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

maintained across a wide range of cell types and show a occupy discrete chromosome territories in the inter-
high degree of cross-species conservation (91% between phase nucleus, with preferred positioning that depends
mouse and human, compared with 67% for fLADs)52. on the cell type and is conserved between humans
Ithas been suggested that constitutive interaction of and other primates59,60. Striking changes in chromo-
LADs with the lamina could act as a structural backbone some positioning are rare, but have been reported to
for the organization of interphase chromosomes52. The occur within minutes61. The positioning of many genes
mechanisms that direct the interaction of cLADs and changes in response to environmental stimuli, during
fLADs with the lamina are likely to involve interactions cell commitment and in disease6265.
with DNA-binding factors, although these factors have Many models of chromosome organization have been
yet to be identified. put forward to describe genomic features that are specific
LADs have a low gene density, but owing to their for cell type or species. In mammalian cells, the existence
overall genomic coverage, they still contain thousands of chromosome territories coupled with the observation
of genes, most of which are not expressed. This provides that some genes are repositioned outside these territories
some support for the longstanding hypothesis that periph- following transcriptional activation, led to the formulation
eral nuclear localization promotes gene silencing. Several of a model in which chromosome territories are separated
studies have tested this hypothesis by expressing a lamina by an inter-chromosomal compartment that allows the
protein tagged to lacI and artificially inducing its ectopic formation of rare long-range interchromosomal inter
binding to lacO sites inserted at different chromosomal actions66. Higher-resolution analysis of chromosome ter-
locations. Particularly illuminating results were obtained ritory organization was subsequently carried out using
by repositioning a chromosome to a more peripheral cryo-FISH, a high-resolution method that combines
location in the nucleus, by tethering it to a protein of the FISH with ultrathin sectioning of cryoprotected cells67. This
innernuclearmembrane53. Microarray analysis showed analysis showed that chromosomes intermingle exten-
that several genes on that chromosome were silenced, but sively in primary human lymphocytes, with an estimated
also that expressionof othergeneswas not reduced, which 20% of the nuclear volume containing sequences from
suggests that localization to thenuclearperipheryis not intermingled pairs of chromosomes67; levels are likely to
incompatible with gene expression. These conclusions be even higher in resting lymphocytes68. An important
are supported by the observation that transgenes are functional consequence of chromosome intermingling
expressed even when integrated into heterochromatin54, is that it seems to promote preferential rearrangements
indicating that heterochromatin is not always restrictive between specific chromosomes, depending on their
for transcription. physical proximity59,67. Comparisons of the extent of inter-
Invivo analyses of the dynamics of Dam-methylated mingling in human lymphocytes with the likelihood of
chromatin show that the association of LADs with the chromosomal translocations following ionizing radiation
nuclear periphery is not fully reestablished after each revealed a strong linear correlation with the frequency of
cell cycle;: a proportion of the LADs that appear in the chromosomal translocations. This suggests that sequences
nuclear periphery during one cell cycle can be found at at regions of intermingling are more likely to recombine
the periphery of the nucleolus in the following cell cycle55 than those at the interior of chromosome territories fol-
(FIG.3). Such a dynamic organization suggests that both lowing DNA double-strand break formation67. A direct
the lamina and the nucleolar periphery have the potential correlation between translocation frequencies (meas-
to organize silent chromatin, although it is not yet clear ured using high-throughput genome-wide translocation
whether similar gene silencing mechanisms operate in sequencing (HTGTS)) and inter-chromosomal contacts
both compartments. The mechanisms underlying this (quantified by Hi-C) was also observed in G1arrested
dynamic organization are not known. Inactive ribosomal mouse proBcells69. Analyses of specific loci, such as
RNA gene arrays are found at the periphery of the nucleo- Igh and Myc, also show a direct relationship between
lus, forming a layer that has many of the characteristics of chromatin contacts (measured by 4Cseq) and sporadic
heterochromatin (reviewed in REF.56). High-throughput translocation events (measured by translocation capture
sequencing has been used to characterize the DNA sequencing (TCseq))70.
sequences bound to the nucleolus (nucleolus-associated Quantitative analyses of chromosome territories by
chromatin domains; NADs), and these have been found cryo-FISH revealed additional unexpected features of
to be enriched for pericentric satellite repeats, A- and chromosome organization in human lymphocytes67.
T-rich sequences and gene-poor regions57,58. cLADs are First, the volumes of chromosome territories are not
also enriched for A- and T-rich sequences and gene-poor simply proportional to their DNA content but depend
regions, suggesting that cLAD and NAD sequences share strongly on the linear density of active genes on each
a mechanism of attaching to the nuclear or nucleolar chromosome. The observation that chromosomes with
periphery. Identifying this mechanism is likely to pro- higher transcriptional activity occupy larger nuclear
vide important insights into the systems that govern volumes may help to explain the positive relationship
chromosomal organization in interphasecells. observed between higher RNA polymerase II (Pol II)
occupancy, active histone marks and expression, and
Cryoprotected cells Chromosome territories in the nucleus. Above, we dis- increased translocation frequency 69,70. This is par-
Cells that have been treated
with a cryoproctectant to
cuss the evidence that distinct genomic regions, TADs, ticularly apparent where these parameters are associ-
prevent structural damage are potential units of chromosomal organization. ated with increased chromatin mobility that favours
during freezing. Atthe next level of organization, whole chromosomes chromosomal translocation events71.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

Perhaps surprisingly, regions of chromosome inter- displacement of sequence-specific and basal transcrip-
mingling are not enriched for open chromatin or for tion factors from chromatin, some of which is triggered
active genes. This raises the possibility that both active by protein phosphorylation74. Chromatin is condensed
and repressed gene regions intermingle, and that posi- by up to 50fold, which results in the formation of meta-
tioning at the periphery of chromosome territories is not phase chromosomes75. The details of this process have
generally necessary for gene expression63,67. This suggests not been fully elucidated but it seems likely to have a
that the chromosome specificity, but not the overall level highly disruptive effect on chromatin domain structures,
of chromosome intermingling, depends on transcrip- which can potentially break up loops that bring enhanc-
tion. In addition, nuclear volume expansion during ers and gene promoters together. A recent study using
lymphocyte activation is accompanied by chromosome- 5C and HiC analyses of chromatin contacts showed that
specific changes in chromosome territory decompaction the TAD organization of interphase nuclei largely dis
during gene activation, intermingling and radial posi- appears in metaphase76. Unlike the disruption of loops
tioning 68. These observations confirm the view that gene that occurs during the passage of replication forks, chro-
expression and genome architecture are closely linked, matin condensation in metaphase is not a brief event
and form the basis for the inter-chromosomal network and affects the entire chromosome simultaneously. This
model67. This model proposes that chromatin contacts creates specific challenges for the maintenance of epige-
within (and, to a lesser extent, between) chromosomes netic information. The disruptive effects of mitosis are
and with nuclear compartments such as the nuclear lam- further emphasized by the observation that some LADs
ina or nucleoli, determine the way in which the genome do not reestablish contact with the nuclear lamina fol-
is folded in a dynamic and physiological manner. The lowing exit from mitosis, but instead are repositioned to
effect of locally determined chromatin contacts would the periphery of the nucleoli55 (FIG.3).
be to promote preferential positioning of chromosomes, Genomic organization through mitosis is likely
and of the active and repressed genes within them in to involve epigenetic marks of active genes and open
the nucleus. chromatin, as well as marking by transcription factors,
cofactors and histone modification readers that remain
Nuclear organization and cell function associated with regulatory elements during metaphase.
Chromatin organization and chromosome positioning Immunofluorescence combined with FISH (immuno-
undergo extensive changes during progression through FISH) and ChIP analyses have shown that H3K4 methyl
the cell cycle, during cell differentiation and during ation, H3 and H4 acetylation and the histone variants
senescence. In this section, we consider how these pro- H3.3 and H2A.Z are maintained at active genes during
cesses challenge the maintenance of gene expression pat- mitosis7779, even when the genes are integrated into
terns and cell identity. We also examine the concept that pericentric heterochromatin77.
they constitute an opportunity for chromatin remodel- Transcription factors and cofactors, such as FOXA1
ling, and for changes to gene expression patterns and (REF.80), GATA1 (REF.81), RUNX2 (REF.82), mixed-lineage
nuclear organization. leukaemia protein 1 (MLL1) 83 and bromodomain-
containing protein 4 (BRD4)84, have also been reported
Cell cycle. Progression through the cell cycle is probably to remain associated with mitotic chromosomes. These
the most disruptive challenge to maintaining nuclear factors could act in conjunction with histone modifica-
organization. Replication occurs within defined nuclear tion marks to promote loop formation and facilitate the
domains, called factories, which can be visualized as reestablishment of transcriptionally active and inactive
replication foci and contain stable chromatin associa- chromatin structures as cells exit mitosis. In support of
tions that are maintained through as many as 15 cell this idea, mitosis-specific depletion of GATA1 resulted
divisions51. Replication factories are clusters of synchro- in delayed expression of GATA1binding genes following
nously replicated replicons that extend over regions of exit from mitosis into the G1 phase (REF.81).
about 1Mb of DNA72,73.
DNA replication poses a challenge to the mainte- Quiescence and differentiation. Quiescent cells are non-
nance of gene expression patterns because of the disrup- dividing cells that can still be stimulated to divide and
tion of transcription factor binding and chromatin loop proliferate by appropriate signals. Terminally differenti-
formation during the passage of the replication fork. ated cells exit the cell cycle permanently and cannot, in
Asdiscussed above, current evidence suggests that most general, be induced to divide. Quiescent and terminally
genes transcribed by Pol II are regulated by a combina- differentiated cells have significantly different chromo-
tion of promoters and enhancers, with the latter often somal organization and epigenetic marks compared with
distributed over large genomic regions. It is possible that dividing cells: for example, naive, quiescent lymphocytes
the passage of the replication fork disrupts only one of have small nuclei with a significantly higher fraction of
these elements at a time, allowing contact with other facultative heterochromatin compared with dividing
regulatory elements to act as a form of cellular memory, lymphocytes.
which would promote transcription factor binding and Chromatin condensation in naive, quiescent
reconstitution of the enhancer. Tcells is mediated, at least in part, by the activity of
Mitosis is likely to present the cell with an even condensin 2, which is also involved in condensing
greater challenge to maintaining epigenetic and regula- mitotic chromosomes85. Facultative heterochromatin
tory information. During mitosis, there is widespread in resting Bcells is marked by a combination of the

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 9

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Activated B cell Plasma cell post-mitotic antibody-producing plasma cell, which has
a small nucleus with a substantial amount of facultative
heterochromatin that is organized into a characteristic
cartwheel structure, with radial spokes extending to
the periphery from a central region of heterochroma-
tin (FIG.4a). Plasma cells are highly specialized for the
production and secretion of large amounts of immuno-
globulin and thus have a highly developed endoplasmic
reticulum to cope with this role. Conversion of their
genome into mainly facultative heterochromatin may
10 m 10 m function to reduce nucleus size and free space for the
enlarged endoplasmic reticulum. Unlike constitutive
b heterochromatin in dividing cells, plasma cell hetero-
P0 P6 P14 P28 9m chromatin does not contain heterochromatin protein 1
(HP1) and is marked by the double H3K9me3H3S10ph
modification88. The presence Ser10 phosphorylation has
been shown to prevent HP1 from binding to the adjacent
H3K9me3marked pericentric heterochromatin during
mitosis89, thereby altering the properties of heterochro-
matin and ultimately affecting transcription factor access
Figure 4 | Chromatin structure of post-mitotic cells. a | During differentiation of an and chromatin condensation89.
activated Bcell (left panel) into a plasma cell (rightNature
panel),Reviews
the dense| Molecular Cell Biology
DAPI (4,6diamidino2 Rod photoreceptor cells in the eyes of nocturnal
phenylindole; blue) staining of the heterochromatin forms a characteristic cartwheel mammals (see above) are another cell type in which the
structure as the nucleus shrinks to accommodate the expanded endoplasmic reticulum, organization of heterochromatin has been studied in
which is required for high-level production of immunoglobulins (red; indirect detail, revealing a fascinating link between chromatin
immunodetection of immunoglobulins). b | Rod photoreceptor cells from adult mice show organization and cellular function. Mouse rod cells have
an unusual nuclear organization in which the heterochromatin (red) is located in the an unusual, inverted chromatin organization whereby
centre of the nucleus and the euchromatin (green) at the periphery. Blue staining shows
heterochromatin is concentrated in a dense chromo-
major satellite repeats-containing chromocentres, detected by fluorescence insitu
hybridization (FISH) with a major satellite-specific probe. Heterochromatin was detected centre in the centre of the nucleus and is absent from
with probes for long interspersed elements (LINE1) and euchromatin with probes for B1 the nuclear periphery (FIG.4b). This inversion is caused
short interspersed nuclear elements. The images show rod photoreceptor cells at birth by developmentally controlled silencing of the expres-
(postnatal day 0 (P0)), P6, P14 and P28 and for the fully adult animal aged 9months (9m). sion of lamins A and C, and of the lamin B receptor 90.
The nuclei have a conventional organization at birth, which is inverted into a centrally Incontrast to the genomic distribution observed in most
localized heterochromatin arrangement in the adult. Images in the figure, part a, courtesy cell types, in rod cells both active and inactive genes are
of P. Sabbattini, MRC Clinical Science Centre, London, UK. The figure, part b, is reprinted found mainly at the nuclear periphery 91, which also
with permission from REF. 91, Elsevier. has higher levels of transcription factors92 and nascent
transcripts91. Analysis of 16 species (nine nocturnal and
seven diurnal) revealed a strong correlation between the
doubleH3K9me3H3S10ph (H3S10 phosphorylation) presence of such an inverted nuclear arrangement and
and H3K27me3H3S28ph modifications, which are not a nocturnal lifestyle. The main functional effect of the
present in activated or terminally differentiated Bcells, inverse organization seems to be changes in the optical
and are instead reminiscent of the epigenetic profile of properties of the cell, which have been hypothesized to
metaphase chromosomes86. Quiescent resting lympho- contribute to the much higher sensitivity of the rod cells
cytes have greatly reduced levels of transcription com- of nocturnal mammals to low light levels91.
pared with activated lymphocytes87, and it is tempting to
link this feature with the increase in chromatin condensa- Senescence. In response to exogenous and endogenous
tion found in the resting cells. However, it is important to stress, cells can undergo permanent cell cycle arrest
note that resting lymphocytes maintain gene expression through a process termed cellular senescence. Among
programmes and are among the most mobile cells in the the stimuli that can induce senescence are replica-
body, using amoeboid movement to migrate and seek out tive stress and overexpression of oncogenes. The latter
antigens that will stimulate their activation and prolifera- induces hyper-proliferation and activation of the DNA
tion. Chromatin condensation could have a role in reduc- damage response in S phase, which helps to trigger the
ing global transcription levels, but its main function may senescent phenotype. Senescent cells, which are enlarged
be to reduce the size of the nucleus, thereby reducing the and generally secrete substantial amounts of protein,
overall size of the cell. often have partially replicated DNA and cytoplasmic
Terminally differentiated, post-mitotic cells differ DNA fragments93.
from quiescent cells in that their exit from the cell cycle Human fibroblasts that undergo oncogene-induced
is irreversible. The differentiated post-mitotic cells that senescence have an unusual chromatin organization,
have been studied so far have a variety of chromosomal which is characterized by the presence of senescence-
arrangements that may relate to the different functions associated heterochromatin foci (SAHFs). Each SAHF
of the cells. The end stage of Bcell differentiation is the is formed by the condensation of a single chromosome,

10 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

with telomeric and centromeric chromatin located reentry to the cell cycle by epigenetic mechanisms,
mainly at the periphery of the SAHF94. The formation which could be mediated in part by changes in chromo-
of SAHFs depends on the HIRA histone chaperone com- some organization and the conversion of the genome
plex, which translocates to PML nuclear bodies soon into mainly heterochromatin. Our understanding of
after senescence has been induced and can initiate SAHF these processes is still limited, but it is clear that the cell
formation95. A detailed analysis of the composition of cycle not only poses a challenge for maintaining nuclear
SAHFs revealed that they are organized in concentric organization but also presents opportunities for renew-
non-overlapping layers of chromatin that carry the ing nuclear structures and maintaining cell plasticity
H3K9me3 and H3K27me3 marks. Interestingly, SAHFs during differentiation. Understanding the role of these
were not observed in mouse fibroblasts, which instead processes and the consequences of their absence in dif-
exhibit a more diffuse distribution of heterochromatin, ferent types of quiescent and differentiated cells is likely
suggesting that there are species-specific differences in to be an expanding area of research in thefuture.
the nuclear organization of senescent cells. Lamin B1 is
also depleted in senescent fibroblasts96,97. Knockdown of Summary and perspectives
lamin B1 results in heterochromatin reorganization and 3Cbased techniques have markedly influenced our
facilitates the formation of SAHFs98, emphasizing its role understanding of chromatin contacts in the nucleus
in tethering heterochromatin to the nuclear envelope. and will continue to do so with further methodologi-
Moreover, the binding of lamin B1 to regions of chro- cal developments. However, it is necessary to be aware
matin enriched for H3K27me3 is increased in senescent of their limitations and to integrate the results that
cells, suggesting that redistribution of lamin B1 binding they provide with information obtained using differ-
could contribute to gene silencing in these cells98,99. ent approaches. It will be important to link studies of
The notion that the cell cycle generally has a disrup- chromatin contacts and chromosomal organization with
tive effect on genome organization implies that non- information about the functional characteristics of the
cycling cells should have a stable genome and nuclear sequences that control transcription, and DNA replica-
organization that can last for months or years. The real- tion and repair. Genome-wide analysis of regulatory
ity is likely to be more complex. For example, quiescent sequences presents significant challenges, but ingen-
lymphocytes move rapidly around the secondary lymph ious approaches are being devised that should allow the
oid organs and through the bloodstream and lymphatic effects of gene position on transcription to be studied at
ducts in search of antigens. Migration alters the shape a global level (see Supplementary informationS1 (Box)).
of the nucleus100 and could affect nuclear organization. Efficient generation of large-scale deletions and other
Moreover, a significant proportion of haematopoietic chromosomal rearrangements through homologous
stem cells (HSCs) is maintained in a quiescent state recombination is now possible using genome editing
during the lifetime of the organism, which is thought tools such as the CRISPRCas9 system104. In the future,
to protect them from DNA damage. However, when our understanding of the role of large-scale chroma-
damage does occur in these quiescent cells, it is repaired tin organization in controlling a wide range of cellular
by error-prone, non-homologous end joining, instead functions should be enhanced by combining chromatin
of byhomologous recombination, which is used in the interaction mapping and genomic deletion data with
fraction of HSCs that are proliferating5,101. This exposes experimental manipulation of key cellular factors such
quiescent HSCs to an increased risk of mutagenesis, as cohesins, CTCF and transcription factors that bind
which may contribute to HSC ageing. Other examples to enhancers and promoters. These developments will
are adult mouse neurons and hepatocytes, which dis- be particularly important for understanding cancer and
play substantial levels of polyploidy and aneuploidy 102,103; congenital diseases, in which disruption of genomic
the polyploidy can be reversed by hepatocyte cell divi- organization by chromosomal rearrangements markedly
sions103. Differentiated, post-mitotic cells seem to block affects gene expression.

1. Wijgerde,M., Grosveld,F. & Fraser,P. Transcription mapping interactions between genomic elements. 13. Tolhuis,B., Palstra,R.J., Splinter,E., Grosveld,F. &
complex stability and chromatin dynamics invivo. Genome Res. 16, 12991309 (2006). deLaat,W. Looping and interaction between
Nature 377, 209213 (1995). 8. Hughes,J.R. etal. Analysis of hundreds of cis- hypersensitive sites in the active -globin locus.
2. Dillon,N., Trimborn,T., Strouboulis,J., Fraser,P. & regulatory landscapes at high resolution in a single, Mol.Cell 10, 14531465 (2002).
Grosveld,F. The effect of distance on long-range high-throughput experiment. Nature Genet. 46, 14. Vernimmen,D., De Gobbi,M., Sloane-Stanley,J.A.,
chromatin interactions. Mol. Cell 1, 131139 (1997). 205212 (2014). Wood,W.G. & Higgs,D.R. Long-range
3. Dekker,J., Rippe,K., Dekker,M. & Kleckner,N. 9. Rodley,C.D., Bertels,F., Jones,B. & OSullivan,J.M. chromosomal interactions regulate the timing of
Capturing chromosome conformation. Science 295, Global identification of yeast chromosome interactions the transition between poised and active gene
13061311 (2002). using genome conformation capture. Fungal Genet. expression. EMBO J. 26, 20412051
4. Simonis,M. etal. Nuclear organization of active and Biol. 46, 879886 (2009). (2007).
inactive chromatin domains uncovered by 10. Lieberman-Aiden,E. etal. Comprehensive mapping of 15. Stadhouders,R. etal. Dynamic long-range chromatin
chromosome conformation capture-onchip (4C). long-range interactions reveals folding principles of the interactions control Myb proto-oncogene transcription
Nature Genet. 38, 13481354 (2006). human genome. Science 326, 289293 (2009). during erythroid development. EMBO J. 31, 986999
5. Naka,K. & Hirao,A. Maintenance of genomic integrity 11. Kalhor,R., Tjong,H., Jayathilaka,N., Alber,F. & (2012).
in hematopoietic stem cells. Int. J.Hematol. 93, Chen,L. Genome architectures revealed by tethered 16. Jing,H. etal. Exchange of GATA factors mediates
434439 (2011). chromosome conformation capture and population- transitions in looped chromatin organization at a
6. Stadhouders,R. etal. Multiplexed chromosome based modeling. Nature Biotech. 30, 9098 developmentally regulated gene locus. Mol. Cell 29,
conformation capture sequencing for rapid genome- (2012). 232242 (2008).
scale high-resolution detection of long-range chromatin 12. Kolovos,P. etal. Targeted Chromatin Capture (T2C): a 17. Markova,E.N., Kantidze,O.L. & Razin,S.V.
interactions. Nature Protoc. 8, 509524 (2013). novel high resolution high throughput method to Transcriptional regulation and spatial organisation of
7. Dostie,J. etal. Chromosome Conformation Capture detect genomic interactions and regulatory elements. the human AML1/RUNX1 gene. J.Cell Biochem. 112,
Carbon Copy (5C): a massively parallel solution for Epigenet. Chromatin 7, 10 (2014). 19972005 (2011).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 11

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

18. Blackledge,N.P., Ott,C.J., Gillen,A.E. & Harris,A. 44. Seitan,V.C. etal. Cohesin-based chromatin 70. Hakim,O. etal. DNA damage defines sites of
An insulator element 3 to the CFTR gene binds CTCF interactions enable regulated gene expression within recurrent chromosomal translocations in B
and reveals an active chromatin hub in primary cells. preexisting architectural compartments. Genome Res. lymphocytes. Nature 484, 6974 (2012).
Nucleic Acids Res. 37, 10861094 (2009). 23, 20662077 (2013). 71. Roukos,V. etal. Spatial dynamics of chromosome
19. Ktistaki,E. etal. CD8 locus nuclear dynamics during 45. Zuin,J. etal. Cohesin and CTCF differentially affect translocations in living cells. Science 341, 660664
thymocyte development. J.Immunol. 184, chromatin architecture and gene expression in human (2013).
56865695 (2010). cells. Proc. Natl Acad. Sci. USA 111, 9961001 72. Jackson,D.A. & Pombo,A. Replicon clusters are
20. Palstra,R.J. etal. The -globin nuclear compartment (2014). stable units of chromosome structure: evidence that
in development and erythroid differentiation. 46. Sofueva,S. etal. Cohesin-mediated interactions nuclear organization contributes to the efficient
NatureGenet. 35, 190194 (2003). organize chromosomal domain architecture. EMBO J. activation and propagation of S phase in human cells.
21. Love,P.E., Warzecha,C. & Li,L. Ldb1 complexes: the 32, 31193129 (2013). J.Cell Biol. 140, 12851295 (1998).
new master regulators of erythroid gene transcription. 47. Young,S.G., Jung,H.J., Coffinier,C. & Fong,L.G. 73. Ma,H. etal. Spatial and temporal dynamics of DNA
Trends Genet. 30, 19 (2014). Understanding the roles of nuclear A- and Btype replication sites in mammalian cells. J.Cell Biol. 143,
22. Deng,W. etal. Controlling long-range genomic lamins in brain development. J.Biol. Chem. 287, 14151425 (1998).
interactions at a native locus by targeted tethering of 1610316110 (2012). 74. Gottesfeld,J.M. & Forbes,D.J. Mitotic repression of
a looping factor. Cell 149, 12331244 (2012). 48. Houben,F. etal. Disturbed nuclear orientation and the transcriptional machinery. Trends Biochem. Sci.
23. Andersson,R. etal. An atlas of active enhancers cellular migration in Atype lamin deficient cells. 22, 197202 (1997).
across human cell types and tissues. Nature 507, Biochim. Biophys. Acta 1793, 312324 (2009). 75. Belmont,A.S. Mitotic chromosome structure and
455461 (2014). 49. Dechat,T., Adam,S.A., Taimen,P., Shimi,T. & condensation. Curr. Opin. Cell Biol. 18, 632638
24. Grosveld,F., van Assendelft,G.B., Greaves,D.R. & Goldman,R.D. Nuclear lamins. CSH Persp. Biol. 2, (2006).
Kollias,G. Position-independent, high-level expression a000547 (2010). 76. Naumova,N. etal. Organization of the mitotic
of the human -globin gene in transgenic mice. 50. Amendola,M. & van Steensel,B. Mechanisms and chromosome. Science 342, 948953 (2013).
Cell51, 975985 (1987). dynamics of nuclear lamina-genome interactions. 77. Chow,C.M. etal. Variant histone H3.3 marks
25. Sabbattini,P., Georgiou,A., Sinclair,C. & Dillon,N. Curr.Opin. Cell Biol. 28, 6168 (2014). promoters of transcriptionally active genes during
Analysis of mice with single copies and multiple copies 51. Guelen,L. etal. Domain organization of human mammalian cell division. EMBO Rep. 6, 354360
of transgenes reveals a novel arrangement for the chromosomes revealed by mapping of nuclear lamina (2005).
5VpreB1 locus control region. Mol. Cell. Biol. 19, 671 interactions. Nature 453, 948951 (2008). 78. Kouskouti,A. & Talianidis,I. Histone modifications
679 (1999). 52. Meuleman,W. etal. Constitutive nuclear lamina defining active genes persist after transcriptional
26. Fields,P.E., Lee,G.R., Kim,S.T., Bartsevich,V.V. & genome interactions are highly conserved and andmitotic inactivation. EMBO J. 24, 347357
Flavell,R.A. Th2specific chromatin remodeling and associated with A/Trich sequence. Genome Res. 23, (2004).
enhancer activity in the Th2 cytokine locus control 270280 (2013). 79. Kelly,T.K. etal. H2A.Z maintenance during mitosis
region. Immunity 21, 865876 (2004). 53. Finlan,L.E. etal. Recruitment to the nuclear reveals nucleosome shifting on mitotically silenced
27. Ellis,J., Talbot,D., Dillon,N. & Grosveld,F. Synthetic periphery can alter expression of genes in human genes. Mol. Cell 39, 901911 (2010).
human -globin 5HS2 constructs function as locus cells. PLoS Genet. 4, e1000039 (2008). 80. Caravaca,J.M. etal. Bookmarking by specific and
control regions only in multicopy transgene 54. Lundgren,M. etal. Transcription factor dosage affects nonspecific binding of FoxA1 pioneer factor to
concatamers. EMBO J. 12, 127134 (1993). changes in higher order chromatin structure mitoticchromosomes. Genes Dev. 27, 251260 (2013).
28. Nasmyth,K. & Haering,C.H. Cohesin: its roles and associated with activation of a heterochromatic gene. 81. Kadauke,S. etal. Tissue-specific mitotic bookmarking
mechanisms. Annu. Rev. Genet. 43, 525558 (2009). Cell 103, 733743 (2000). by hematopoietic transcription factor GATA1. Cell
29. Parelho,V. etal. Cohesins functionally associate with 55. Kind,J. etal. Single-cell dynamics of genomenuclear 150, 725737 (2012).
CTCF on mammalian chromosome arms. Cell 132, lamina interactions. Cell 153, 178192 (2013). 82. Young,D.W. etal. Mitotic retention of gene
422433 (2008). 56. Padeken,J. & Heun,P. Nucleolus and nuclear expression patterns by the cell fate-determining
30. Hadjur,S. etal. Cohesins form chromosomal cis- periphery: velcro for heterochromatin. Curr. Opin. Cell transcription factor Runx2. Proc. Natl Acad. Sci. USA
interactions at the developmentally regulated IFNG Biol. 28, 5460 (2014). 104, 31893194 (2007).
locus. Nature 460, 410413 (2009). 57. van Koningsbruggen,S. etal. High-resolution whole- 83. Blobel,G.A. etal. A reconfigured pattern of MLL
31. Seitan,V.C. etal. A role for cohesin in Tcell-receptor genome sequencing reveals that specific chromatin occupancy within mitotic chromatin promotes rapid
rearrangement and thymocyte differentiation. domains from most human chromosomes associate transcriptional reactivation following mitotic exit.
Nature476, 467471 (2011). with nucleoli. Mol. Biol. Cell 21, 37353748 Mol.Cell 36, 970983 (2009).
32. Lobanenkov,V.V. etal. A novel sequence-specific DNA (2010). 84. Zhao,R., Nakamura,T., Fu,Y., Lazar,Z. &
binding protein which interacts with three regularly 58. Nemeth,A. etal. Initial genomics of the human Spector,D.L. Gene bookmarking accelerates the
spaced direct repeats of the CCCTC-motif in the nucleolus. PLoS Genet. 6, e1000889 (2010). kinetics of post-mitotic transcriptional reactivation.
5flanking sequence of the chicken cmyc gene. 59. Parada,L.A., McQueen,P.G. & Misteli,T. Tissue- Nature Cell Biol. 13, 12951304 (2011).
Oncogene 5, 17431753 (1990). specific spatial organization of genomes. Genome Biol. 85. Rawlings,J.S., Gatzka,M., Thomas,P.G. & Ihle,J.N.
33. Ong,C.T. & Corces,V.G. CTCF: an architectural 5, R44 (2004). Chromatin condensation via the condensin II complex
protein bridging genome topology and function. 60. Tanabe,H. etal. Evolutionary conservation of is required for peripheral Tcell quiescence. EMBO J.
Nature Rev. Genet. 15, 234246 (2014). chromosome territory arrangements in cell nuclei from 30, 263276 (2011).
34. Sanyal,A., Lajoie,B.R., Jain,G. & Dekker,J. The long- higher primates. Proc. Natl Acad. Sci. USA 99, 86. Sabbattini,P. etal. An H3K9/S10 methyl-phospho
range interaction landscape of gene promoters. 44244429 (2002). switch modulates Polycomb and Pol II binding at
Nature 489, 109113 (2012). 61. Bridger,J.M. Chromobility: the rapid movement of repressed genes during differentiation. Mol. Biol. Cell
35. Xu,Z., Wei,G., Chepelev,I., Zhao,K. & Felsenfeld,G. chromosomes in interphase nuclei. Biochem. Soc. 25, 904915 (2014).
Mapping of INS promoter interactions reveals its role Trans. 39, 17471751 (2011). 87. Frangini,A. etal. The Aurora B kinase and the
in long-range regulation of SYT8 transcription. 62. Williams,R.R., Broad,S., Sheer,D. & Ragoussis,J. polycomb protein Ring1B combine to regulate active
NatureStruct. Mol. Biol. 18, 372378 (2011). Subchromosomal positioning of the epidermal promoters in quiescent lymphocytes. Mol. Cell 51,
36. Kehayova,P., Monahan,K., Chen,W. & Maniatis,T. differentiation complex (EDC) in keratinocyte and 647661 (2013).
Regulatory elements required for the activation and lymphoblast interphase nuclei. Exp. Cell Res. 272, 88. Sabbattini,P. etal. A novel role for the Aurora B
repression of the protocadherin- gene cluster. 163175 (2002). kinase in epigenetic marking of silent chromatin in
Proc.Natl Acad. Sci. USA 108, 1719517200 63. Ferrai,C. etal. Poised transcription factories prime differentiated postmitotic cells. EMBO J. 26,
(2011). silent uPA gene prior to activation. PLoS Biol. 8, 46574669 (2007).
37. Guo,Y. etal. CTCF/cohesin-mediated DNA looping is e1000270 (2010). 89. Fischle,W. etal. Regulation of HP1chromatin binding
required for protocadherin- promoter choice. 64. Volpi,E. etal. Large-scale chromatin organisation of by histone H3 methylation and phosphorylation.
Proc.Natl Acad. Sci. USA 109, 2108121086 (2012). the major histocompatibility complex and other Nature 438, 11161122 (2005).
38. Dixon,J.R. etal. Topological domains in mammalian regions of human chromosome 6 and its response to 90. Solovei,I. etal. LBR and lamin A/C sequentially tether
genomes identified by analysis of chromatin interferon in interphase nuclei J.Cell Sci. 113, peripheral heterochromatin and inversely regulate
interactions. Nature 485, 376380 (2012). 15651576 (2000). differentiation. Cell 152, 584598 (2013).
39. Nora,E.P. etal. Spatial partitioning of the regulatory 65. Meaburn,K.J., Gudla,P.R., Khan,S., Lockett,S.J. & 91. Solovei,I. etal. Nuclear architecture of rod
landscape of the Xinactivation centre. Nature 485, Misteli,T. Disease-specific gene repositioning in breast photoreceptor cells adapts to vision in mammalian
381385 (2012). cancer. J.Cell Biol. 187, 801812 (2009). evolution. Cell 137, 356368 (2009).
40. Hou,C., Li,L., Qin,Z.S. & Corces,V.G. Gene density, 66. Cremer,T. etal. Chromosome territories a 92. Helmlinger,D. etal. Glutamine-expanded ataxin7
transcription, and insulators contribute to the functional nuclear landscape. Curr. Opin. Cell Biol. 18, alters TFTC/STAGA recruitment and chromatin
partition of the Drosophila genome into physical 307316 (2006). structure leading to photoreceptor dysfunction.
domains. Mol. Cell 48, 471484 (2012). 67. Branco,M.R. & Pombo,A. Intermingling of PLoSBiol. 4, e67 (2006).
41. Sexton,T. etal. Three-dimensional folding and chromosome territories in interphase suggests role in 93. Rai,T.S. & Adams,P.D. Lessons from senescence:
functional organization principles of the Drosophila translocations and transcription-dependent chromatin maintenance in non-proliferating cells.
genome. Cell 148, 458472 (2012). associations. PLoS Biol. 4, e138 (2006). Biochim. Biophys. Acta 1819, 322331 (2013).
42. Van Bortle,K. etal. Insulator function and topological 68. Branco,M.R., Branco,T., Ramirez,F. & Pombo,A. 94. Zhang,R., Chen,W. & Adams,P.D. Molecular
domain border strength scale with architectural Changes in chromosome organization during PHA- dissection of formation of senescence-associated
protein occupancy. Genome Biol. 15, R82 (2014). activation of resting human lymphocytes measured heterochromatin foci. Mol. Cell. Biol. 27, 23432358
43. Kim,Y.J., Cecchini,K.R. & Kim,T.H. Conserved, by cryo-FISH. Chromosome Res. 16, 413426 (2007).
developmentally regulated mechanism couples (2008). 95. Ye,X. etal. Downregulation of Wnt signaling is a
chromosomal looping and heterochromatin barrier 69. Zhang,Y. etal. Spatial organization of the mouse trigger for formation of facultative heterochromatin
activity at the homeobox gene A locus. Proc. Natl genome and its role in recurrent chromosomal and onset of cell senescence in primary human cells.
Acad. Sci. USA 108, 73917396 (2011). translocations. Cell 148, 908921 (2012). Mol. Cell 27, 183196 (2007).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/reviews/molcellbio

2015 Macmillan Publishers Limited. All rights reserved


REVIEWS

96. Freund,A., Laberge,R.M., Demaria,M. & Campisi,J. 110. Gavrilov,A.A. etal. Disclosure of a structural milieu RNA polymerase III. EMBO J. 18, 22412253
Lamin B1 loss is a senescence-associated biomarker. for the proximity ligation reveals the elusive nature of (1999).
Mol. Biol. Cell 23, 20662075 (2012). an active chromatin hub. Nucleic Acids Res. 41, 123. Faro-Trindade,I. & Cook,P.R. A conserved
97. Shimi,T. etal. The role of nuclear lamin B1 in cell 35633575 (2013). organization of transcription during embryonic stem
proliferation and senescence. Genes Dev. 25, 111. OSullivan,J.M., Hendy,M.D., Pichugina,T., cell differentiation and in cells with high C value.
25792593 (2011). Wake,G.C. & Langowski,J. The statistical-mechanics Mol.Biol. Cell 17, 29102920 (2006).
98. Shah,P.P. etal. Lamin B1 depletion in senescent cells of chromosome conformation capture. Nucleus 4, 124. Jackson,D.A. Features of nuclear architecture that
triggers large-scale changes in gene expression and 390398 (2013). influence gene expression in higher eukaryotes:
the chromatin landscape. Genes Dev. 27, 17871799 112. Nicodemi,M. & Pombo,A. Models of chromosome confronting the enigma of epigenetics. J.Cell Biochem.
(2013). structure. Curr. Opin. Cell Biol. 28, 9095 79 (Suppl. 35), 6977 (2000).
99. Sadaie,M. etal. Redistribution of the lamin B1 (2014). 125. Osborne,C. etal. Active genes dynamically colocalize
genomic binding profile affects rearrangement of 113. Barbieri,M. etal. Complexity of chromatin folding is to shared sites of ongoing transcription. Nature Genet.
heterochromatic domains and SAHF formation during captured by the strings and binders switch model. 36, 10651071 (2004).
senescence. Genes Dev. 27, 18001808 (2013). Proc. Natl Acad. Sci. USA 109, 1617316178 126. Schoenfelder,S. etal. Preferential associations
100. Friedl,P., Wolf,K. & Lammerding,J. Nuclear (2012). between coregulated genes reveal a transcriptional
mechanics during cell migration. Curr. Opin. Cell Biol. 114. Bau,D. etal. The three-dimensional folding of the interactome in erythroid cells. Nature Genet. 42,
23, 5564 (2011). -globin gene domain reveals formation of chromatin 5361 (2010).
101. Mohrin,M. etal. Hematopoietic stem cell quiescence globules. Nature Struct. Mol. Biol. 18, 107114 127. Brookes,E. etal. Polycomb associates genome-wide
promotes error-prone DNA repair and mutagenesis. (2011). with a specific RNA polymerase II variant, and
Cell Stem Cell 7, 174185 (2010). 115. Nagano,T. etal. Single-cell HiC reveals celltocell regulates metabolic genes in ESCs. Cell Stem Cell 10,
102. Rehen,S.K. etal. Chromosomal variation in neurons variability in chromosome structure. Nature 502, 157170 (2012).
of the developing and adult mammalian nervous 5964 (2013). 128. Lanzuolo,C., Roure,V., Dekker,J., Bantignies,F. &
system. Proc. Natl Acad. Sci. USA 98, 1336113366 116. Hozak,P. & Cook,P.R. Replication factories. Orlando,V. Polycomb response elements mediate the
(2001). TrendsCell Biol. 4, 4852 (1994). formation of chromosome higher-order structures in
103. Duncan,A.W. etal. The ploidy conveyor of mature 117. Baddeley,D. etal. Measurement of replication the bithorax complex. Nature Cell Biol. 9, 11671174
hepatocytes as a source of genetic variation. structures at the nanometer scale using super- (2007).
Nature467, 707710 (2010). resolution light microscopy. Nucleic Acids Res. 38, e8 129. Grimaud,C. etal. RNAi components are required for
104. Terns,R.M. & Terns,M.P. CRISPR-based (2010). nuclear clustering of Polycomb group response
technologies: prokaryotic defense weapons 118. Jackson,D.A., Iborra,F.J., Manders,E.M. & elements. Cell 124, 957971 (2006).
repurposed. Trends Genet. 30, 111118 Cook,P.R. Numbers and organization of RNA 130. Tiwari,V.K., Cope,L., McGarvey,K.M., Ohm,J.E. &
(2014). polymerases, nascent transcripts, and transcription Baylin,S.B. A novel 6C assay uncovers Polycomb-
105. Dekker,J., Marti-Renom,M.A. & Mirny,L.A. units in HeLa nuclei. Mol. Biol. Cell 9, 15231536 mediated higher order chromatin conformations.
Exploring the three-dimensional organization of (1998). Genome Res. 18, 11711179 (2008).
genomes: interpreting chromatin interaction data. 119. Martin,S. & Pombo,A. Transcription factories:
Nature Rev. Genet. 14, 390403 (2013). quantitative studies of nanostructures in the Acknowledgements
106. Ethier,S.D., Miura,H. & Dostie,J. Discovering mammalian nucleus. Chromosome Res. 11, 461470 The authors thank P. Sabbattini for providing the images
genome regulation with 3C and 3Crelated (2003). shown in FIG.4a. Work in N.D.s laboratory is supported by
technologies. Biochim. Biophys. Acta 1819, 401410 120. Pombo,A. etal. Specialized transcription factories the Medical Research Council, UK. A.P. thanks the Helmholtz
(2012). within mammalian nuclei. Crit. Rev. Eukaryot. Gene Foundation for support.
107. de Wit,E. & de Laat,W. A decade of 3C technologies: Expr 10, 2129 (2000).
insights into nuclear organization. Genes Dev. 26, 121. Kimura,H., Tao,Y., Roeder,R.G. & Cook,P.R. Competing interests statement
1124 (2012). Quantitation of RNA polymerase II and its The authors declare no competing interests.
108. van de Werken,H.J. etal. Robust 4Cseq data transcription factors in an HeLa cell: little soluble
analysis to screen for regulatory DNA interactions. holoenzyme but significant amounts of polymerases
Nature Methods 9, 969972 (2012). attached to the nuclear substructure. Mol. Cell. Biol. SUPPLEMENTARY INFORMATION
109. Belmont,A.S. Large-scale chromatin organization: the 19, 53835392 (1999). See online article: S1 (Box)
good, the surprising, and the still perplexing. 122. Pombo,A. etal. Regional specialization in human ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Curr.Opin. Cell Biol. 26, 6978 (2014). nuclei: visualization of discrete sites of transcription by

NATURE REVIEWS | MOLECULAR CELL BIOLOGY ADVANCE ONLINE PUBLICATION | 13

2015 Macmillan Publishers Limited. All rights reserved

You might also like