ArticlePDF AvailableLiterature Review

The interplay of histone modifications - Writers that read

Authors:
  • London Institute of Medical Sciences

Abstract

Histones are subject to a vast array of posttranslational modifications including acetylation, methylation, phosphorylation, and ubiquitylation. The writers of these modifications play important roles in normal development and their mutation or misregulation is linked with both genetic disorders and various cancers. Readers of these marks contain protein domains that allow their recruitment to chromatin. Interestingly, writers often contain domains which can read chromatin marks, allowing the reinforcement of modifications through a positive feedback loop or inhibition of their activity by other modifications. We discuss how such positive reinforcement can result in chromatin states that are robust and can be epigenetically maintained through cell division. We describe the implications of these regulatory systems in relation to modifications including H3K4me3, H3K79me3, and H3K36me3 that are associated with active genes and H3K27me3 and H3K9me3 that have been linked to transcriptional repression. We also review the crosstalk between active and repressive modifications, illustrated by the interplay between the Polycomb and Trithorax histone-modifying proteins, and discuss how this may be important in defining gene expression states during development.
Review
Histones and Chromatin Review series
The interplay of histone modifications writers
that read
Tianyi Zhang
, Sarah Cooper
*,
& Neil Brockdorff
Abstract
Histones are subject to a vast array of posttranslational modifi-
cations including acetylation, methylation, phosphorylation, and
ubiquitylation. The writers of these modifications play important
roles in normal development and their mutation or misregulation
is linked with both genetic disorders and various cancers. Readers
of these marks contain protein domains that allow their recruit-
ment to chromatin. Interestingly, writers often contain domains
which can read chromatin marks, allowing the reinforcement of
modifications through a positive feedback loop or inhibition of
their activity by other modifications. We discuss how such positive
reinforcement can result in chromatin states that are robust and
can be epigenetically maintained through cell division. We
describe the implications of these regulatory systems in relation to
modifications including H3K4me3,H3K79me3, and H3K36me3 that
are associated with active genes and H3K27me3 and H3K9me3 that
have been linked to transcriptional repression. We also review the
crosstalk between active and repressive modifications, illustrated
by the interplay between the Polycomb and Trithorax histone-
modifying proteins, and discuss how this may be important in
defining gene expression states during development.
Keywords chromatin; histone modifications; Polycomb; Trithorax
DOI 10.15252/embr.201540945 | Received 29 June 2015 | Revised 4 September
2015 | Accepted 16 September 2015 | Published onl ine 16 October 2015
EMBO Reports (2015) 16: 14671481
See the Glossary for abbreviations used in this article.
Introduction
In eukaryotes, DNA is packaged in the form of chromatin. The basic
unit of chromatin, the nucleosome, is comprised of 147 bp of DNA
wrapped around a histone octamer made of two dimers of H2A and
H2B and a tetramer of H3 and H4 proteins. The N- and C-terminal
histone tails protrude from the nucleosome core and have the poten-
tial to interact with adjacent nucleosomes and the linker DNA. All
histones can be posttranslationally modified, and the sites of modifi-
cation are often on the histone tails. These modifications can
regulate chromatin structure directly and frequently act as binding
sites for the recruitment of other non-histone proteins to chromatin.
The most abundant histone modifications are acetylation, phospho-
rylation, methylation, and ubiquitylation, although many other
modifications have been reported (reviewed recently in [1]).
Transcriptionally active and silent chromatin is characterized by
distinct posttranslational modifications on the histones or combina-
tions thereof. Active genes typically carry high levels of lysine acety-
lation on the H3 and H4 tails, trimethylation of H3 lysine 4,
trimethylation of H3 lysine 79, ubiquitylation of H2B, and trimethy-
lation of H3 lysine 36 (Fig 1). Marks associated with repressed genes
include trimethylation of lysine 27, ubiquitylation of H2A on lysine
119, and trimethylation of H3 lysine 9 (Fig 1). The chromatin-modi-
fying enzymes that catalyze these marks can be recruited to target
sites by sequence-specific DNA-binding transcription factors that
regulate transcriptional states of particular genes. However, other
more general features of the DNA such as its global CG content and
DNA methylation status can be read by the DNA-binding Zn-finger
CxxC domain present in many chromatin-modifying enzymes [2].
Equally, the act of transcription can direct the recruitment of writers
that associate with the transcriptional machinery, leading to the
accumulation of specific marks such as H3K4me3 and H3K36me3.
Given the large number of different histone modifications, the
potential combinatorial complexity is vast. Advances in technology
over the past decade such as ChIP-sequencing have allowed us to
map the distribution and co-localization of histone marks at high-
resolution genome wide, while mass spectrometry, often in combi-
nation with stable isotope labeling, enables the analysis of histone
marks and dynamics at the level of a single histone tail. Interest-
ingly, mass spectrometric data suggest that there are many combina-
tions of modifications that are either more likely to occur together,
or are mutually exclusive, suggesting crosstalk between these
marks. Such crosstalk can occur in cis between distinct modifi-
cations on the same histone tail, or in trans either on neighboring
histones within the same nucleosome or on neighboring nucleo-
somes in a chromatin domain.
The patterns of histone marks associated with distinct transcrip-
tional states are established through a dynamic interplay between
histone readers, writers, and erasers. Importantly, the writers that
place these marks contain chromatin-reading domains that can bind
preexisting histone marks. Studies have shown that such crosstalk
between histone marks can both positively and negatively regulate
binding and catalytic activity of writers, resulting in positive and
Developmental Epigenetics, Department of Biochemistry, University of Oxford, Oxford, UK
*Corresponding author. Tel: +44 1865 613230; E-mail: sarah.cooper@bioch.ox.ac.uk
These authors contributed equally to this work
ª 2015 The Authors. Published under the terms of the CC BY 4.0 license EMBO reports Vol 16 |No 11 | 2015
1467
negative feedback loops. Therefore, writers that can also read the
histone modifications are required for the establishment and mainte-
nance of chromatin states at active and repressed genes and may
play important roles in the memory and switching of gene
expression states.
In this review, we will focus on several examples of the positive
and negative feedback mechanisms that regulate the formation,
reinforcement, and maintenance of the distinct patterns of histone
marks associated with active and repressed transcriptional states
(Fig 2). However, such features are likely to be more general
features of chromatin states, and the principles seen in these exam-
ples are likely to be applicable to the plethora of other chromatin
modifications whose function is still unclear.
Active histone modifications
In eukaryotic organisms, gene expression is regulated through the
synergistic actions of multiple factors, including but not limited to,
transcription factors, the transcriptional machinery, chromatin
remodelers, and the presence of specific histone variants and
histone modifications. Active chromatin domains are characterized
by a distinct array of histone marks. H3K27ac and H3K4me1 are
associated with active enhancers [3], and high levels of H3K4me3
and H3 and H4 acetylation are found at the promoters of active
genes [46]. The bodies of active genes are enriched in H3 and H4
acetylation [7], H3K79me3 [8], and H2BK120u1 [9,10], and increas-
ing H3K36me3 toward the 3
0
end [11]. These histone marks may
regulate transcription by creating an open chromatin structure and
recruit effectors that mediate a transcriptionally competent state.
While the function of many active histone modifications is not fully
understood, there is abundant evidence that their deposition is
required for the proper regulation of gene expression. Positive
crosstalk mechanisms between different histone modifications play
an important role in the recruitment and maintenance of active
histone modifications at active genes.
Establishment and maintenance of H3K4me3
H3K4me3 is a highly conserved histone modification and its associa-
tion with transcription is evolutionarily conserved in eukaryotes.
Glossary
AEPB2 AE-binding protein 2
ASH2L absent, small, or homeotic-like
ATRX5/6 Arabidopsis Trithorax-related protein 5 /6
BEND3 Ben domain containing 3
BLOCS broad local enrichments
BRE1 Brefeldin-A sensitivity protein 1
CBX chromobox
CDYL chromodomain protein, Y-like
CFP1 CXXC finger protein 1
ChIP-sequencing chromatin immunoprecipitation followed by DNA
sequencing
CpG cytosine-phosphate-guanine
CTCF CCCTC-binding factor
CTBP2 C-terminal-binding protein 2
CTD C-terminal domain
DNMT3A/B DNA methyltransferase 3A/B
DOT1 disruptor of telomeric silencing 1
DOT1L DOT1-like
DPY30 dumpy-30 protein homolog
EAF3 Esa1p-associated factor 3
EED embryonic ectoderm development
ESC embryonic stem cell
EZH2/1 enhancer of zeste homolog 2/1
FRAP fluorescence recovery after photobleaching
G9a/GLP G9a and G9
a-like protein
H2AK119u1 histone H2A lysine 119 monoubiqutination
H2BK120u1 histone H2B lysine 120 monoubiqutination
H2BK34u1 histone H2B lysine 34 monoubiqutination
H3K27me1/2/3 histone H3 lysine 27 mono/di/trimethylation
H3K36me1/2/3 histone H3 lysine 36 mono/di/trimethylation
H3K4me1/2/3 histone H3 lysine 4 mono/di/trimethylation
H3K9me1/2/3 histone H3 lysine 9 mono/di/trimethylation
HAT histone acetyltransferase
HBO1 histone acetyltransferase bound to ORC 1
HDAC
histone deactylase complex
HMT histone methyltransferase
Hox gene homeobox-containing gene
HP1 heterochromatin protein 1
JARID2 jumonji, AT-rich interactive domain 2
KDM2A/B lysine demethylase protein 2A/B
MBD methyl binding domain
MES-4 mesoderm expressed 4
MLL1/2/3/4 mixed-lineage leukemia 1/2/3/4 complex
MSL1/2 male-specific lethal 1/2
NO66 nucleolar protein 66
NSD1/2/3 nuclear receptor-binding SET domain
protein 1/2/3
NuA3/4 nucleosomal acetyltransferase of histone H3/H4
NURD nucleosome remodeling and deacetylase
P300/CBP P300- and CREB-binding protein
PAF polymerase-associated factor
PCGF1/2/3/4/5/6 Polycomb group ring finger 1/2
/3/4/5/6
PCL1/2/3 Polycomb-like 1/2/3
PH polyhomeotic
PHD finger plant homeodomain finger
Pol II RNA polymerase II
PRC1 Polycomb repressive complex 1
PRC2 Polycomb repressive complex 2
RAD6 ras-related associated with diabetes protein 6
RBBP5 retinoblastoma-binding protein 5
RING1A/B really interesting new gene 1A/B
RNF20/40 ring finger protein 20/40
RpAb46/48 Rb-associated protein 46/48
RPD3S reduced potassium dependency 3S complex
RYBP RING1- and YY1-binding protein
SAGA Spt-Ada-Gcn5 acetyltransferase
SET domain
Su(var)3-9, enhancer-of-zeste and Trithorax
domain
SETD1A/B SET domain containing 1A/B
SETD2 SET domain containing 2
SETMAR SET domain and mariner transposase fusion
containing protein 2
SMYD2 SET and MYND domain-containing protein 2
SUV3-9 H1/H2 suppressor of variegation 3-9 homolog 1/2
SUZ12 Suppressor of zeste 12 homolog
TrxG Trithorax group
TSS transcription start site
WDR5 WD repeat-containing protein 5
YAF2 YY1-associated factor 2
ZMYND11 zinc finger, MYND-type containing 11
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1468
In mammals, H3K4 methylation is catalyzed by six related homologs
of the yeast SET1SETD1A, SETD1B, MLL1, MLL2, MLL3, and
MLL4 [12]. These complexes are comprised of the catalytic SETD1/
MLL subunits and four core subunits WDR5, RBBP5, ASH2L, and
DPY30, and as well many other complex-specific subunits [1315].
H3K4me3 is a hallmark of active genes and is distributed along the
promoter and TSS regions [6,16,17]. Work in yeast shows that SET1
associates with the PAF complex and the Ser5-phosphorylated initi-
ating form of Pol II and is co-transcriptionally deposited [18] (Fig 3).
Additionally, the recruitment of SETD1 and MLL to specific target
genes is mediated by many cell type-specific transcription factors or
transcriptional coactivators [1923]. However in higher organisms
especially, more general mechanisms of H3K4me3 recruitment and
establishment are also at play.
Notably, the distribution of H3K4me3 is highly coupled to the
presence of CpG islands, regions of CpG- and GC-dense DNA that
are predominately unmethylated and found at 5070% of vertebrate
promoters [24]. A biochemical link between CpGI promoters and
H3K4me3 was eluciated with the discovery of the Zn-finger CxxC
domain which specifically binds nonmethylated CpGs and is present
in MLL1/2 and the CFP1 subunit of SETD1A/B [2] (Fig 3). All CpGI
promoters are marked with H3K4me3, and the level of H3K4me3 is
correlated to gene activity [25,26]. Emerging evidence suggests that
in vivo, MLL2 is responsible for maintaining H3K4me3 at CpGI
promoters with low expression [27,28], while the SETD1-specific
subunit CFP1 is preferentially enriched at active gene promoters
with higher levels of H3K4me3 [29]. In ESCs, CpGI promoters linked
to developmentally regulated genes are bivalent and harbor the
repressive H3K27me3 mark as well as H3K4me3 [30]. Importantly,
it has been suggested that the ability of H3K4 writers to sample
CpGIs genome wide and the presence of H3K4me3 at CpGI promo-
ters may poise silent genes for rapid activation upon differentiation.
SETD1/MLL complexes may reinforce their binding through
recognition of their own mark, H3K4me3. The PHD finger domain
of CFP1 is known to read H3K4me3 and mediates SETD1 interaction
with H3K4me3 [3133]. The third PHD domain in MLL1 is impor-
tant for H3K4me3 binding and MLL1 recruitment to target sites in
the Hox locus [34]. Other PHD domains within SETD1/MLL may
also interact with H3K4me3 but remain to be further characterized
[35]. The ability of SETD1/MLL to sample promoters and bind
H3K4me3 may be involved in the maintenance of this mark at active
genes. These mechanisms of H3K4me3 binding by H3K4 writers
suggest that once established, this mark may positively reinforce its
own deposition.
Crosstalk between H2BK120u1,H3K4me3, and H3K79me3
One of the best-studied pathways of positive histone crosstalk is the
stimulation of H3K4me3 and H3K79me3 by H2BK120u1 (or
H2BK123u1 in yeast). In yeast, H2BK123u1 is established by the
ubiquitin ligase RAD6/BRE1 during transcriptional initiation and
localizes to the TSS and along the bodies of active genes [36]. Deple-
tion of RAD6/BRE1 or mutation of H2BK123 causes severe loss of
H3K4me3 and H3K79me3 [37,38]. This positive crosstalk between
H2BK123u1 and H3K4me3 and H3K79me3 is specific and does not
extend to the regulation of H3K36me3, another mark associated
with transcription [37,38].
H2BK123 lies in close proximity to H3K79 on the exposed
nucleosome surface, and the H3K79 methyltransferase DOT1 in
yeast has been shown to be influenced by deletion and mutation of
residues on the H2B tail [39]. In humans, the situation is more
complex, as H3K79me3 and DOT1L distribution is not solely depen-
dent on H2BK120u1. Human DOT1L localizes at active genes and
peaks around the TSS and moreover has been shown to bind both
Active genes
Repressed genes
Positive
feedback
Negative
feedback
Writers
Active
marks
Positive
feedback
Writers
Repressive
marks
Figure 2. Crosstalk between chromatin writers and histone marks at
active and repressed genes.
Chromatin writers and chromatin marks associated with active genes positively
reinforce each other through various positive feedback mechanisms. The same
holds true for writers and marks associated with repressed genes. Additionally,
negative feedback mechanisms and mutual inhibition between writers and
marks associated with the opposite gene expression state also reinforce distinct
transcriptional states.
Repressed genes
H3K9me3
H2AK119ub1
H3K27me3
Promoter
Active genes
H3/H4 acetylation
H2BK120ub1
H3K79me3
H3K36me3H3K4me3
Promoter
TSS
TSS
Figure 1. The distribution of histone modifications over active and
repressed genes.
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1469
Ser5- and Ser2-phosphorylated forms of the Pol II CTD [40]. As
such, H3K79me3 is a marker of active genes, yet its exact role in
transcriptional regulation remains to be discovered.
The crosstalk between H2BK120u1 and H3K4me3 is conserved in
mammals, as knockdown of the BRE1 homologs RNF20/40 leads to
global reduction in H3K4me3 [41] (Fig 3). More recently, studies on
the MSL1/MSL2 E3 ligase that catalyzes H2BK34u1 have also
revealed a crosstalk between H2BK34u1 and H3K4me3 [42]. Both
H2BK120u1 and H2BK34u1 are now known to allosterically stimu-
late the activity of the MLL complex through binding to the ASH2L
subunit [43]. Sites of ubiquitylation at H2BK120 and H2BK34 reside
on the nucleosome surface and may provide a more favorable
substrate for SET1 or MLL complex binding and activity [43]. As
ASH2L is a core subunit of all writers of H3K4 methylation, H2B
ubiquitylation may be one mechanism of H3K4me3 maintenance at
active promoters through a positive feedback loop whereby tran-
scription results in deposition of H2Bub, which subsequently
activates the H3K4 methyltransferases.
H3K4me3 and histone acetylation
Histone lysine acetylation is a highly abundant mark and is known
to regulate many cellular processes including transcription. Acetyla-
tion of histones H3 and H4 is highly correlated with gene
expression. A unique structural motif, the bromodomain, specifi-
cally recognizes acetylated lysines and is present in many proteins
involved in transcriptional regulation [44]. Besides the direct
recruitment of effectors, histone acetylation has also been proposed
to physically alter chromatin structure by neutralizing the positive
charge of lysines and disrupting intra- and internucleosomal interac-
tions, which lead to an open chromatin environment permissible to
transcription. Lysine acetylation of three residues on the H3 globular
domain H3K56, H3K64, and H3K122, all of which lie at the H3DNA
interface, may disrupt electrostatic interactions within the nucleo-
some and have been linked to gene activation [4547]. H3K122ac
has been shown to directly promote in vitro transcription through
stimulating histone eviction [47]. H3 and H4 histone tail acetyla-
tions enhance DNA unwrapping, while H3 acetylation sensitizes
nucleosomes to salt-induced dissociation [48].
H3K4me3 and H3/H4 acetylation coexist at the promoter and
TSS of active genes, and there are many studies that suggest
H3K4me3 promotes downstream H3/H4 acetylation through recruit-
ment of HATs (Fig 4). H3K4me3 readers have been identified in
many HAT complexes. SGF29, a component of the SAGA HAT
complex, contains a tandem tudor domain that binds H3K4me3 and
overlaps with H3K4me3 at gene promoters. SGF29 deletion causes
loss of H3K9ac and loss of SAGA complex at target sites [49]. Simi-
larly, yeast NuA3 [50] and NuA4 [51], and mammalian HBO1 [52]
provide other examples of HAT complexes that contain PHD fingers
that preferentially bind H3K4me3. Dynamic turnover of H3 lysine
acetylation through the combinatorial action of the HAT p300/CBP
and HDAC has been shown to occur on histone H3 tails with pre-
existing H3K4me3, but not other modifications associated with
active gene expression such as H3K79me3 or H3K36me3 [53]. This
H3K4me3-linked acetylation is conserved in higher eukaryotes
including fly, mouse, and human. Loss of H3K4me3 upon CFP1
deletion leads to loss of CpGI-associated H3K9ac in ESCs [29].
Further work using the Dictyostelium discoideum model shows that
upon knockout of SET1 and loss of H3K4me3, dynamic H3 acetyla-
tion was lost [54]. The dynamic turnover of acetylation rather than
the modification itself may be key in transcriptional activation
(reviewed in [55]). In support of this, many members of the
H3K4me3-binding PHD fingers are associated with HDACs as well
as HATs [56]. As H3K4me3 has been found to be promoter-
associated before transcription initiation, H3K4me3-dependent
co-targeting of both HATS and HDACs may facilitate the dynamic
turnover of histone acetylation. The above examples illustrate that
positive crosstalk between H3K4me3 and histone acetylation is an
evolutionarily conserved pathway and that the cooperativity
Pol II
DPY30
ASH2L
SETD1/
MLL
WDR5
RbBP5
SETD1
CFP1
WDR5
RbBP5
DPY30
ASH2L
ASH2L
MLL1/2
WDR5
RbBP5
DPY30
C
x
x
C
SETD1
CFP1
WDR5
RbBP5
DPY30
ASH2L
CxxC
CxxC
Methylated
CpG site
Unmethylated
CpG site
H3K4me3
H2BK120u1
TSS
Figure 3. Establishment of H3K4me3 and interplay with H2BK120u1.
The SETD1 complex associates with Pol II, and H3K4me3 is deposited co-transcriptionally. CFP1 (associated with SETD1) and MLL1/2 can be recruited to promoters de novo via
CxxC domain binding to CpG islands. H2BK120u1 can recruit H3K4 writers, possibly through recognition of H2BK120u1 by the ASH2L subunit.
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1470
between H3K4me3 and hyperacetylation as well as the dynamic
turnover of acetylation is important in ensuring proper transcrip-
tional regulation.
H3K36me3 and histone deacetylation
Methylation at histone H3K36 is an abundant histone mark highly
conserved in eukaryotes. H3K36 mono-, di-, and trimethylation exist
in yeast and all of these states are catalyzed by SET2. Mammals on
the other hand have multiple writers of H3K36 methylation, includ-
ing the NSD1/2/3 family, ASH1L, SMYD2, SETMAR, and SETD2,
but SETD2 is the sole enzyme responsible for H3K36 trimethylation
in vivo (reviewed in [57]) (Fig 4). Interestingly, the uncoupling of
H3K36me3 activity from H3K36me1/2 over evolution alludes to
specific biologically distinct roles of each methylation state.
H3K36me3 is highly correlated with the transcribed regions of
active genes and levels of H3K36me3 increase toward the 3
0
end of
genes [11]. This particular distribution results from the association
of Set2 with the elongating Ser2-phosphorylated CTD of Pol II,
which is predominant over the bodies and 3
0
ends of active genes
[5860]. Like H3K4me3, H3K36me3 has also been linked to regula-
tion of histone acetylation. H3K36me3 recruits HDACs to sites of
active transcription (Fig 4). In yeast, recognition of H3K36me2/3 by
the bromodomain-containing EAF3 complex recruits the HDAC
RPD3S complex, which deacetylates histones and prevents spurious
transcription initiation from within gene bodies [6163]. H3K4me3
and histone hyperacetylation at gene promoters may regulate tran-
scriptional initiation from the TSS, while H3K36me2/3-mediated
deacetylation is required in the wake of the transcriptional
machinery to prevent initiation from aberrant sites within the gene
body. This mutual exclusivity of H3K4me3 and H3K36me3 may be
important for maintaining transcriptional integrity. This idea is
supported by work showing that promoters lack the H3K36me2/3
mark, and the H3K36me2 demethylases KDM2A/B co-localize with
H3K4me3 at CpGI promoters, ensuring active removal of H3K36me2
from transcriptional start sites [64,65].
H3K36me2/3 is recognized by a protein motif, the PWWP
domain, found in many nuclear chromatin-binding proteins [66
69]. Notably, all three members of the NSD family of H3K36 methyl-
transferases that catalyze H3K36me1/2 each contain two PWWP
domains [70] and have been shown to preferentially bind H3
peptides containing H3K36me3 [69]. This implies that H3K36me2/3
recognition by its writers may be important for the propagation of
H3K36me1 and H3K36me2 at certain sites. Mono-/dimethylation of
H3K36 is more pervasive than H3K36me3 and not restricted to sites
of active transcription or euchromatin domains [71,72]. The biologi-
cal function of mono-/dimethylation is unknown, though an
increase in H3K36me2 levels as a result of mutations in NSD2 has
been linked to upregulation of gene expression profiles in cancers
[7375]. H3K36me2 may have an important biological function in
its own right or may be required to serve as a substrate for subse-
quent SETD2-mediated H3K36 trimethylation. The broad distribu-
tion of H3K36me2 and H3K36me3 over active chromatin may also
prevent the spreading and accumulation of silencing marks such as
H3K27me3 through direct inhibition of the Polycomb complex PRC2
[76,77], which will be discussed below.
Repressive histone modifications
The methylation of residues lysine 27 and lysine 9 of H3 and the
ubiquitinylation of H2A on lysine 119 are hallmarks of repressive
chromatin and are often found at silent gene loci. H3K27me3 and
H2AK119u1 are associated with the formation of facultative hetero-
chromatin, whereas H3K9me2/3, as well as having important roles
in the formation of constitutive heterochromatin, also plays a part
in regulating gene expression during development.
H3K27me3 and H2AK119u1 crosstalk
The Polycomb Repressive Complex 2 (PRC2) is responsible for the
methylation of lysine 27 and contains four core subunits, EZH2/1,
SUZ12, EED, and RBAP46/8 [78]. The catalytic subunit is the SET
domain-containing protein EZH2 (or the related EZH1), although
these enzymes are only functional in the context of the full core
complex [7981]. There are also accessory proteins that can asso-
ciate with the core PRC2 complex to form two types of PRC2:
PRC2.1 which includes a Polycomb-like subunit (PCL1/2/3) and
PRC2.2 which includes the JARID2 and AEBP2 subunits [82]. The
function of these accessory proteins remains unclear, although
HDAC
Pol II
HATS
SETD2
ASH1L
SMYD2
NSD1/2/3
SETMAR
Methylated
CpG site
Unmethylated
CpG site
TSS
H3K4me3
H3/H4ac
H3K36me2
H3K36me3
Figure 4. Interplay between H3K4me3,H3K36me3, and H3/H4 acetylation.
H3K4me3 reinforces H3 and H4 acetylation at the promoters of active genes. Various H3K36 writers catalyze H3K36me1/2 and SETD2 associates with elongating Pol II and
catalyzes H3K36me3 co-transcriptionally. H3K36me2/3 recruits HDACs that deacetylate histones over gene bodies.
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1471
they have been shown to modulate activity of PRC2 and may also
play a role in targeting PRC2 to chromatin. PRC2 is able to mono-,
di-, and trimethylate H3K27, although there is some dispute if
PRC2 is the only H3K27me1 methyltransferase. These different
methylation states have very different roles, and although
H3K27me3 is linked to gene repression, recent studies have
suggested that H3K27me1 may be important for gene activation
and is enriched over the bodies of genes [83]. The H3K27me2
modification is very prevalent in the genome, with MS/MS analy-
sis demonstrating that it accounts for 6080% of all nucleosomes
in mESCs [84], although little is known about its function or bind-
ing proteins. H3K27me3 is the most well-characterized mark in
terms of facultative heterochromatin formation and is critical for
the repression of key transcriptional regulators during develop-
ment. Therefore, in terms of gene silencing, we will focus on the
trimethylation state of H3K27.
In ES cells, H3K27me3 is present at the promoters of several
thousand genes, including the Hox gene clusters, where it is associ-
ated with heritable gene silencing [85]. H3K27me3 modification is
also highly enriched on the inactive X chromosome suggesting a role
in facultative heterochromatin formation [86]. In more differentiated
cell types, larger domains of H3K27me3, termed BLOCS, are often
visualized over silent loci in the genome [87]. As described above,
for many of the enzymes associated with active gene expression,
there are also positive feedback loops important for the establish-
ment and spreading of repressive domains. The PRC2 component
EED contains an aromatic cage that is able to specifically bind to
H3K27me3 [88]. It has been shown that the binding of PRC2 to the
modification it deposits is required for the full establishment of
H3K27me3 domains, and such a positive feedback mechanism could
also be important for the inheritance of the H3K27me3 mark
through cell division [89]. PRC2 has also been shown to be stimu-
lated by dense chromatin via an interaction of the SUZ12 subunit
with the H3 tail (A31-R42) [90]. In this way, positive feedback from
the local chromatin structure will also allow robust domains of
H3K27me3 to be maintained over repressed genes.
The Polycomb repressive complex 1 (PRC1) is an E3 ubiquitin
ligase complex that can modify chromatin by monoubiquitylation of
H2A on lysine 119. All PRC1 complexes contain the catalytic
RING1A/B subunit, and one of six PCGF proteins [91]. The presence
of different PCGF subunits is thought to define the class of PRC1
complex, for example, PCGF2 (MEL18) and PCGF4 (BMI) make up
the canonical PRC1 complexes which also contain CBX (2,4,6,7,8)
and polyhomeotic subunits [92]. Variant complexes include either
the RYBP or YAF2 protein, the presence of which is mutually exclu-
sive with the CBX component [91,93]. These variant complexes,
such as the complex containing RING1B/PCGF1/RYBP/BCOR/
KDM2B, have been implicated in recruitment of PRC1 and have
been shown to have higher H2AK119u1 activity compared with
canonical PRC1 complexes [91,94]. Interestingly, RYBP also
contains a ubiquitin-binding domain and has been shown to bind
H2AK119u1 [95]. This suggests that a positive reinforcement mech-
anism could be important to establish or maintain high levels of
H2AK119u1 at PRC1 target domains, in a similar manner to PRC2
where EED binds to H3K27me3.
Both PRC1 and PRC2, along with their associated chromatin
modifications, H2AK119u1 and H3K27me3, have been shown to co-
localize at many regions of the genome, such as the promoters of
developmentally regulated genes and the inactive X chromosome
[9699]. A hierarchical recruitment model, whereby the H3K27me3
modification placed by PRC2 is read by PRC1, has been proposed to
explain this co-recruitment of both PRC1 and PRC2 to chromatin
[100]. This occurs by a specific interaction of the H3K27me3 modifi-
cation with the chromodomain of the CBX protein found in canoni-
cal PRC1 complexes [101]. Hence, all PRC2 targets would also
become PRC1 targets and a repressive domain would be estab-
lished. However, this hierarchical model is not able to account for
all PRC1 recruitment to chromatin since even in the absence of
PRC2, the variant RYBP-containing complexes still localize to the
correct regions of the genome [93,102]. More recently, data from
three laboratories have shown that the reverse mechanism is also
possible, whereby PRC1 is recruited first, followed by PRC2. In this
model, the H2AK119u1 placed by a variant PRC1 complex is recog-
nized by PRC2 [103105]. At present, we do not know how
H2AK119u1 is recognized by PRC2, but it has been shown that the
PRC2.2 complex (containing the accessory factors AEBP2 and
JARID2) is enriched in chromatin containing the H2AK119u1 modi-
fication [105]. Additionally, in vitro, this PRC2.2 complex is more
active on an H2AK119u1 nucleosome substrate compared with
unmodified nucleosomes [105]. A remaining question is whether
this PRC2 recruitment to H2AK119u1 is via a direct recruitment
mechanism, similar to CBX binding to H3K27me3, or by a change in
chromatin state or structure associated with the large H2AK119u1
modification.
In summary, the establishment of Polycomb repressive domains
may require these enzymes to read not only their own mark, for
example, EED-binding H3K27me3 or RYBP-binding H2AK119u1, but
also the marks placed by their partner complex. In this way,
H3K27me3 can establish or reinforce H2AK119u1 modifications,
and H2AK119u1 can establish or reinforce H3K27me3 deposition
(Fig 5). Which modification or Polycomb complex is initiating this
recruitment is still a matter of debate although recent work has
implicated the variant PRC1-KDM2B-containing complex in initia-
tion [106108]. Polycomb target sites overlap with regions of dense
unmethylated DNA, CpG islands, and the CxxC domain of KDM2B
is able to recognize unmethylated CpGs, providing a plausible
mechanism for PRC1 recruitment. Further work is needed to fully
understand how both PRC1 and PRC2 complexes are initially
recruited to CpG islands. However, once this is established, the posi-
tive feedback mechanisms described above involving the histone
modifications that these enzymes place will be important to
maintain and reinforce their activity at these target sites.
H3K27me3 and H3K9me2/3 crosstalk
Generally, methylation of H3K9 is associated with constitutive
heterochromatin formation and transcriptional silencing. Recently,
there has been some evidence that H3K9 methylation can crosstalk
with the Polycomb H3K27me3 modification to cooperate in gene
repression or as mutually exclusive pathways present at constitutive
heterochromatin.
The heterodimeric complex of G9a and GLP catalyzes H3K9me1
and H3K9me2 modifications [109], which are mainly associated with
transcriptional silencing but also occur in euchromatic regions [110].
Both proteins contain ankyrin repeat domains that can bind to
H3K9me1/2 modifications, allowing the enzymes to read their own
marks and therefore allow spreading of the H3K9me2 modification
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1472
[111]. The enzyme SETDB1 can place both H3K9me2/me3 and has
roles in repression of transposons, in gene silencing and at pericen-
tric heterochromain [112115]. The SUV3-9H1/H2 enzymes deposit
H3K9me2 and H3K9me3 modifications and contain a chromo-
domain which can recognize these marks [116]. A major site of the
H3K9me3 modification is at pericentric heterochromatin, where there
are self-reinforcing feedback loops involving the chromodomain-
containing protein HP1 which can bind to H3K9me3, and recruit de
novo DNA methyltransferases (DNMT3A/B) [117,118]. The resulting
DNA methylation can be recognized by MECP2, a protein containing
a MBD (methyl binding domain) which can also bind to and recruit
SUV3-9 enzymes to pericentric heterochromain [119] (Fig 6A). Inter-
estingly at mitosis, H3 is phosophorylated by the kinase Aurora B at
H3S10, and this modification next to the H3K9me3 mark causes HP1
to be displaced from the pericentric heterochromatin during this
phase of the cell cycle [120].
Several reports have demonstrated that H3K9me3 and H3K27me3
modifications are mutually exclusive [87,103,121,122]. In differenti-
ated cells, H3K27me3 BLOCS, which form over silent gene loci, are
mutually exclusive with H3K9me3 domains over features such as
transposons [87]. In SUV3-9H1/H2 knockout cells, there is a loss of
H3K9me3 at the pericentric heterochromatin, and a subsequent gain
of H3K27me3 [103,121], suggesting that not only can these two
marks compensate for each other, but that normally H3K9me3
prevents H3K27me3 establishment. A recent study isolating proteins
associated with pericentric heterochromatin has shown that a
chromatin-associated protein, BEND3, is recruited to pericentric
chromatin in the absence of H3K9me3 (or DNA methylation) and is
important for recruiting H3K27me3 [122]. Lack of DNA methylation
can also cause H3K27me3 to be recruited to pericentric hetero-
chromatin, but in this case H3K9me3 is still present and forms mutu-
ally exclusive domains with H3K27me3, despite both modifications
now being enriched at DAPI-dense pericentric regions [103]. Recruit-
ment of H3K27me3 to pericentric heterochromatin has also been
shown to occur during early mouse development. In the one-cell
stage embryo, H3K27me3 can be visualized specifically on the male
pericentric heterochromatin, but not the female heterochromatin,
which contains H3K9me3 [123]. In this system, it has recently
been shown that it is not the presence of H3K9me3 on the mater-
nal pericentric heterochromatin itself that prevents H3K27me3
recruitment, but rather the presence of HP1b which binds to
H3K9me3 [124].
Despite reports that H3K27me3 and H3K9me3 are mutually
exclusive, a number of ChIP-sequencing studies in ES cells [115],
extra-embryonic lineages [125], and differentiated cells [126] have
shown that both H3K9me2 and H3K9me3 modifications can coexist
with the PRC2 modification H3K27me3 at developmentally
repressed genes. Given that both marks are associated with gene
repression, it has been suggested that they may cooperate with each
other. A mass spectrometry study, in which H3K27me3-containing
nucleosomes were purified from HeLa cells, showed that H3K9me2
modifications, and to a lesser extent H3K9me3, were also present
with H3K27me3 [127]. Large-scale proteomic screens have identi-
fied several Polycomb proteins as readers of H3K9me3 modifications
[31,69]. However, the authors suggest that this may be due to the
affinity of CBX proteins to H3K27me3 and the high degree of
sequence identity surrounding H3K9 and H3K27 (T
ARKST and
A
ARKSA, respectively). In vitro there is no difference in PRC2 activ-
ity on nucleosomes that contain H3K9 methylation compared to WT
nucleosomes [76]. However, a recent paper has found a direct inter-
action of PRC2 with the G9a/GLP complex and that G9a enzymatic
activity (H3K9me2) modulates PRC2 genomic recruitment [128]. In
addition, studies have reported that PRC2 is necessary for the bind-
ing of HP1 to chromatin [129131]. Both H3K27me3 and H3K9me2
modifications have been shown to be present on the inactive X
chromosome where the two modifications play complementary
roles [132]. Here, the molecular mechanisms of crosstalk have been
elucidated by the discovery of CDYL, a protein that can bind both
H3K9me2 and H3K27me3. CDYL can interact with G9a to propagate
the H3K9me2 modification [133]. Interestingly, the loss of PRC2,
and subsequent loss of H3K27me3, reduces the amount of H3K9me2
present on the Xi, suggesting that CDYL is a link between these two
enzymatic activities allowing the combinatorial reading and writing
of both modifications (Fig 6B).
RbAP48
RYBP
RING1B
PCGF
PRC1
JARID2
SUZ12
EZH2
EED
AEBP2
PRC2.2
PCGF2/4
RING1B
CBX
RbAP48
SUZ12
EZH2
EED
PRC2
PRC1
Unmethylated
CpG site
H2AK119u1 H3K27me3
TSS
Figure 5. Crosstalk between the Polycomb complexes PRC1 and PRC2.
PRC2 reinforces its own mark through binding of EED to H3K27me3. PRC1 may also reinforce its own mark through binding of RYBP to H2AK119u1. Establishment of PRC1 can
be reinforced by the presence of PRC2, through recognition of H3K27me3 by the CBX subunit of PRC1. PRC2 establishment can also be reinforced by PRC1 through the
recognition of H2AK119u1 by the JARID2/AEBP2 PRC2.2 complex.
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1473
The mutually exclusive distribution of H3K27me3 and H3K9me3
described above has predominantly been shown in studies of consti-
tutive heterochromatin loci, and in all cases, this clear separation of
domains has been seen for trimethylation marks. There are also
many examples in which the H3K9me2 modification, and in some
cases H3K9me3, can act in concert with H3K27me3, suggesting posi-
tive crosstalk between these two mechanisms of heterochromatin
formation. In the best-documented examples, this positive crosstalk
appears to be between H3K27me3 and H3K9me2. Crosstalk may
also vary in different cell types or different differentiation states, in
which chromatin structures or the balance of different enzymes may
be altered. What is clear is that at least in some circumstances
through, for example, CDYL, HP1, or DNA methylation, both the
Polycomb system and the H3K9 methylation systems are able to
read the chromatin state placed by each other and to write their
own modifications accordingly.
As discussed above, both H3K27me3 and H3K9me3 repressive
marks can exist as extensive domains within cells. These domains
can spread through positive feedback of the writers and their ability
to recognize and propagate their marks. Given this, the establish-
ment of boundaries is critical to isolate heterochromatin from
euchromatin domains. These boundary elements have been found
in multiple eukaryotic organisms, ranging from yeast to human
(recently reviewed in [134]). Boundaries can be formed by equilib-
rium between heterochromatin-promoting factors (remodelers, Poly-
comb proteins, H3K9me3 machinery) and euchromatin-promoting
factors (remodelers, transcriptional machinery, Trithorax proteins).
Such boundaries could vary in position and this concept forms the
basis of position effect variegation (PEV) in which the spread of
heterochromatin, for example, domains of H3K9me3, results in the
stochastic silencing of a neighboring gene (reviewed in [135]). Addi-
tionally, cis-regulatory elements and the binding of insulator
proteins such as CTCF can also determine boundaries. For instance,
H3K27me3 domain boundaries within the Hox gene cluster are
bound by CTCF; deletion of these CTCF-binding sites results in the
infringement of Pol II and H3K4me3 into adjacent heterochromatin
territories and disruption of Hox gene silencing [136].
Interplay between repressive and active
chromatin modifications
The main theme that emerges from the data we have described is
that chromatin writers, associated with either active or repressed
states, are positively regulated by their own marks or marks associ-
ated with the same transcriptional state. However, there are clear
data that show these chromatin writers can also be negatively influ-
enced by marks associated with the opposite transcriptional state.
These negative feedback loops reinforce the maintenance of distinct
chromatin states, and may play an important role for switching of
gene expression during differentiation and development by creating
and reinforcing a bistable state.
Historically, the best-characterized example of an interplay
between chromatin complexes in regulating gene expression is the
antagonism between Polycomb and Trithorax complexes. Drosophila
genetics first established Polycomb and Trithorax proteins as two
groups having opposing function on Hox gene expression, and sub-
sequently on the regulation of many important developmental
genes. Histone crosstalk is important in this interplay, and some of
the molecular mechanisms that govern this mutual antagonism
between Polycomb and Trithorax proteins and marks have been
elucidated.
Schmitges et al [76] were the first to show a mechanism of direct
inhibition of PRC2 by the TrxG modifications H3K4me2/3 and
H3K36me2/3. The catalytic activity of the PRC2 core complex was
greatly reduced on recombinant nucleosomes carrying trimethyl-
lysine analogs at H3K4 and H3K36 on the H3 tail. This study and
further work demonstrated inhibition of PRC2 activity when the
marks H3K4me2/3 and H3K36me2/3 exist in cis (on the same
histone tail) as the target H3K27. It has been suggested that the
allosteric inhibition of the catalytic subunit EZH2 occurs through
the recognition of the H3K4me2/3 and H3K36me2/3 marks by the
SUZ12 VEFS domain [76]. Inhibition of PRC2 by H3K36me2/3 is
consistent with mass spectrometry data of the histone H3 peptide
fragment K27-R40 isolated from total chromatin from mESCs and
transformed cell lines [73,77,137]. This shows that trimethylation
at H3K27 and H3K36 do not coexist on the same H3 tail or are
DNMT3A/B
HP1
A Positive feedback at repressive pericentric heterochromatin
CDYL
G9A
GLP
G9A
GLP
CDYL
PRC2
B Co-occupancy of H3K9me2 and H3K27me3 on
the inactive X chromosome
SUV39
MECP2
Methylated
CpG site
H3K9me2
H3K9me3
H3K27me3
Figure 6. Interplay between H3K9me3, DNA methylation, and
H3K27me3.
(A) At the pericentric heterochromatin, DNA methylation and H3K9me3
positively reinforce each other. HP1 binds H3K9me3 and recruits the de novo
DNA methyltransferases DNMT3A/B. MECP2 can bind methylated DNA and
recruit the H3K9me3 methyltransferase SUV3-9. (B) H3K27me3 and H3K9me2
coexist on the inactive X chromosome. CDYL may recruit G9a to the inactive
X chromosome through its ability to recognize H3K9me2 and H3K27me3.
CDYL may reinforce the prop agation of H3K9me2 at the Xi.
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1474
present at very low levels. Furthermore, removal of SETD2, the
only HMT capable of placing H3K36me3, leads to an increase
of H3K27me2 over bodies of active genes and reduces levels of
expression [83].
It is important to note that although PRC2 is inhibited in cis by
H3K4me2/3 and H3K36me3, PRC2 is active on nucleosomes harbor-
ing these modifications on only one of the two H3 tails, thereby
allowing the formation of asymmetrically modified nucleosomes
[127]. Such nucleosomes have been identified in vivo and may
represent the nucleosomes present at bivalent promoters in mESCs
(promoters that harbor both active and repressive marks, see later).
This negative feedback mechanism has also been shown to oper-
ate at a chromosomal level in C. elegans. Normally H3K36me3 and
H3K27me3 occupy mutually exclusive domains on the autosomes
[138]. However, removal of the H3K36me1/2 writer MES-4 in the
germ line results in a global loss of H3K36me3 leading to the redis-
tribution of H3K27me3 to exogenous sites at germ line-expressed
genes, which are normally modified by H3K36me3. This redistribu-
tion causes a titration of the H3K27me3 mark from its endogenous
sites, including the X chromosomes, and an inability to maintain
normal gene expression states [138140].
As well as the inhibition of Polycomb activity by Trithorax
marks, Polycomb marks have also been shown to inhibit the activity
of some Trithorax proteins. The best-studied examples are the inhi-
bition of H3K36 methyltransferases by the PRC1 modification
H2AK119u1. Yuan et al [141] show that the catalytic domain of
H3K36 methyltransferases is inhibited by recombinant nucleosomes
containing H2AK119u1. Additionally, there is evidence that the PcG
PRC1 mark H2AK119u1 inhibits H3K4 methyltransferases MLL1 and
possibly MLL3. A study by Endoh et al shows that upon RING1A/B
knockout and subsequent depletion of H2AK119u1, H3K4me3 levels
at several PcG target genes increase [142]. Although not extensively
investigated, it is possible that H3K27me3 may also inhibit the depo-
sition of H3K4 methylation by the SETD1 and MLL3/4 complexes
[143].
These studies provide good evidence that PcG and TrxG marks
mutually inhibit the writers associated with the opposing group. It
was therefore unexpected when several groups showed that the
tudor domains of the PRC2-associated PCL1/2/3 proteins can
specifically bind H3K36me3 [144147]. Structural and biochemical
analyses show that the tudor domain of PCL recognizes
H3K36me2/3 with high specificity; however, PCL co-localizes only
moderately with H3K36me3 in vivo by ChIP-sequencing [144,147].
One interpretation of this observation is that the role of this inter-
action may be important in the spreading of PRC2 and H3K27me3
to bodies of active genes, and perhaps during the switching of tran-
scriptional state by allowing the initial recruitment of PRC2 to
active genes. Consistent with the latter observation, it has been
shown that the H3K36 demethylase NO66 can be recruited by
PCL3, which would allow for the removal of H3K36me3 before the
subsequent acquisition of H3K27me3 [146].
H3K27me3 is associated with gene repression, while H3K27ac is
associated with gene activation and active enhancers. Since they act
on the same lysine residue, these marks are mutually exclusive, and
the switch between methylation and acetylation has been well
established. The removal of H3K27ac by the NURD complex facili-
tates the recruitment of PRC2 and accumulation of H3K27me3 at
promoters leading to gene repression [148]. This process occurs
during differentiation of ESCs, when CTBP2 in combination with NURD
initiates the silencing of genes that were originally active, through
H3K27 deacetylation, allowing deposition of H3K27me3 by PRC2[149].
Upon loss of H3K9 or DNA methylation, PRC2 accumulates at
the pericentric heterochromatin as discussed earlier. In this condi-
tion, BEND3 recruits NURD to the pericentric heterochromatin
[122], and thus, a similar mechanism of deacetylation of H3K27ac
could explain the subsequent PRC2 recruitment and accumulation
of H3K27me3 at these sites.
Activation of Polycomb-repressed genes requires a methylation
to acetylation switch at H3K27. The phosphorylation of H3S28 on
the residue neighboring H3K27 has been shown to mediate this
switch. H3S28p inhibits H3K27me3, allowing an accumulation of
H3K27 acetylation [150,151]. Similarly, the loss of PRC2 subunit
SUZ12 and H3K27me3 leads to the accumulation of H3K27ac at PcG
target genes [84]. It has been suggested that one role of H3K27me3
is to exclude the HATs p300 and CBP, preventing accumulation of
H3K27ac at enhancers that is important for gene activation [152].
H3K27me2 has been suggested to play a similar role to prevent the
firing of non-cell-type-specific enhancers. This idea is supported by
the increase of H3K27ac at these enhancers when H3K27
methylation is lost [83].
Discussion
It is clear that specific histone modifications are associated with the
transcriptional state. For many modifications, it is not well
established whether they directly influence transcription or their
placement is simply a consequence of the transcriptional state
present at a particular gene. There are reports that acetylation can
directly alter chromatin structure to a more accessible state allowing
the recruitment of transcription factors and the transcription
machinery. In addition, ubiquitylation of H2A has been shown to
inhibit the elongating form of Pol II, suggesting direct effects on the
transcriptional state. Conversely, there is substantial evidence to
support the idea that transcription factors determine and initiate
gene expression, and writers recognize this state and aid in the
maintenance of this state through multiple feedback mechanisms.
One such mechanism is the recruitment of the H3K4 HMT SETD1
and the H3K36 HMT SETD2 by Pol II itself to deposit histone modifi-
cations across the promoter and gene body. The emerging idea that
PcG and TrxG can sample CpG islands genome wide and establish
domains of repression or activation depending on the transcriptional
state at the target promoter also supports the idea that the transcrip-
tional state defines the chromatin modification landscape. This
mechanism requires extensive positive and negative crosstalk
between these modifications that we have discussed.
Many writers of chromatin modifications are positively regulated
by the marks that they place, as well as other marks associated with
the same transcriptional state, contributing to the reinforcement of
gene expression or silencing. This mechanism could also account
for the spreading of marks such as the repressive modifications
H3K9me3 and H3K27me3 over large domains in differentiated cells.
These reinforcing mechanisms may also play a role in cellular
memory by faithful propagation of the histone modifications that
allow gene expression profiles to be maintained epigenetically
through cell division. There is evidence to suggest that some writers
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1475
remain associated with chromatin during DNA replication, but the
exact molecular mechanisms of this epigenetic memory have yet to
be fully elucidated.
Negative histone crosstalk plays an equally important role in
dictating distinct chromatin environments. Writers, especially in the
case of PcG and TrxG proteins, are often negatively regulated by
marks associated with the opposing transcriptional state. As we have
seen in the case of H3K27 methylation and acetylation, negative
crosstalk is also involved in the switching of gene expression states.
It is known that in certain circumstances, marks associated with
positive and negative transcription can coexist. In ESCs, bivalent
promoters contain both H3K27me3 and H3K4me3, albeit on different
histone tails of the same nucleosome, and may represent a chro-
matin profile amenable to switching between transcriptional states.
Genes coding for master transcription factors often have bivalent
promoters in ESCs, and their expression is dynamically regulated
through development.
The ability of writers to read and place histone modifications is
important for the maintenance and regulation of specific transcrip-
tional states throughout development. This is evident by the fact
that mice deficient in chromatin-modifying enzymes display severe
developmental defects. Several of these enzymes have been known
to be involved in various genetic disorders such as Sotos syndrome
and WolfHirschhorn syndrome related to translocation of several
members of the NSD family of H3K36 methyltransferases [153155],
and cancers related to translocation or mutation of MLL, NSD2,
PRC2, SETD2, and countless others [156158]. Most recently, an
H3K27M mutation in H3.3 found in pediatric glioma cancer has
been shown to deplete levels of H3K27me3 globally potentially
through a dominant negative mechanism [159].
In conclusion, the ability of chromatin writers to read preexisting
histone modifications contributes in two major ways. First, it allows
the maintenance of distinct and robust transcriptional states, which
could potentially be propagated through cell division, and therefore
act as epigenetically inherited features. Second, crosstalk between
modifications and enzymes of opposing transcriptional states can
allow the establishment of bistable switches that allow the dynamic
regulation of gene expression states. In the future, we hope to
understand the extent to which histone crosstalk plays a role in
defining the epigenetic landscape of a cell (Sidebar A), and address
the role of histone modifications and the crosstalk between them
during the processes of development and disease.
Acknowledgements
We thank David Brown, Vincenzo Di Cerbo, Benoit Moindrot, and Andrew
Bassett for critical reading of the manuscript. Work in the Brockdorff labora-
tory is funded by grants from the European Research Council (340081) and
Wellcome Trust (103768).
Conflict of interest
The authors declare that they have no conflict of interest.
References
1. Arnaudo AM, Garcia BA (2013 ) Proteomic characterization of novel
histone post-translational modifications. Epigenetics Chromatin 6: 24
2. Voo KS, Carlone DL, Jacobsen BM, Flodin A, Skalnik DG (2000) Cloning
of a mammalian transcriptional activator that binds unmethylated
CpG motifs and shares a CXXC domain with DNA methyltransferase,
human trithorax, and methyl-CpG binding domain protein 1. Mol Cell
Biol 20: 2108 2121
3. Creyghton MP, Cheng AW, Welstead GG, Kooistra T, Carey BW, Steine
EJ, Hanna J, Lodato MA, Frampton GM, Sharp PA et al (2010) Histone
H3K27ac separates active from poised enhancers and predicts develop-
mental state. Proc Natl Acad Sci USA 107: 21931 21936
4. Barrera LO, Li Z, Smith AD, Arden KC, Cavenee WK, Zhang MQ,
Green RD, Ren B (2008) Genome-wide mapping and analysis of active
promoters in mouse embryonic stem cells and adult organs. Genome
Res 18: 46 59
5. Deckert J, Struhl K (2001) Histone acetylation at promoters is differen-
tially affected by specific activators and repressors. Mol Cell Biol 21:
2726 2735
Sidebar A: In need of answers
i) How do chromatin modifications regulate gene transcription?
Although there is a clear correlation between chromatin modifi-
cations and gene expression states, it will be important to establish
the role that modifications play in regulating transcription and
indeed whether they are a cause or an effect of transcription.
ii) What is the role of highly prevalent chromatin modifications such
as H3K36me2 or H3K27me2 that mark up to 50%ofH3 in mESCs
and differentiated tissues [ 84,121 ,160]? Is it possible that these
marks are necessary to reduce noise, for example, by blocking inap-
propriate histone modifications?
iii) What is the role of histone variants? H3.3 and H2A.Z are enriched
over active genes and may have more specialized regulatory roles
compared to their more abundant canonical counterparts [161,162].
Histone readers and writers may be sensitive to the histone variant
status. For instance, the putative tumor suppressor ZMYND11 is an
H3.3 variant-specific reader of H3K36me3 [163], while the H3K27
methyltransferase in plants, ATRX5/6, is active on the canonical H3.1
but inhibited by H3.3 [164].
iv) How are epigenetic profiles established within a cell? What are the
relative contributions of direct targeting of histone-modifying activ-
ities and crosstalk between histone modifications or transcriptional
state? For example, are chromatin writers able to sample and read
the preexisting chromatin state to determine their activity or bind-
ing profiles, or are they directly recruited to their sites of action by
sequence-specific DNA-binding factors?
v) To what extent are epigenetic modifications maintained through
cell division, and do self-reinforcing feedback loops provide a model
for the mechanism of such inheritance? Unlike writers and readers,
the genomic location of histone modifications can be easily trans-
mitted through both mitosis and meiosis because they are an inte-
gral part of the packaging of DNA. However, self-reinforcing loops
might become essential after replication to overcome the dilution
of old, modified nucleosomes with new nucleosomes and maintain
an epigenetic code.
vi) Do the reciprocal feedback loops between positively and negatively
acting histone marks provide the basis for a bistable switch, in
which each state is positively reinforced and stable once the initial
decision has been made? In order to properly generate and validate
such mathematical models, it will be critical to obtain quantitative
data on the kinetics and dynamics of the catalytic and binding
processes that are involved. This will involve experiments such as
FRAP and in vitro binding and enzymatic assays, and importantly
determining the changes that occur when the system has been
perturbed.
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1476
6. Liang G, Lin JC, Wei V, Yoo C, Cheng JC, Nguyen CT, Weisenberger DJ,
Egger G, Takai D, Gonzales FA et al (2004) Distinct localization of
histone H3 acetylation and H3-K4 methylation to the transcription start
sites in the human genome. Proc Natl Acad Sci USA 101: 7357 7362
7. Myers FA, Evans DR, Clayton AL, Thorne AW, Crane-Robinson C (2001)
Targeted and extended acetylation of histones H4 and H3 at active
and inactive genes in chicken embryo erythrocytes. J Biol Chem 276:
20197 20205
8. Ng HH, Ciccone DN, Morshead KB, Oettinger MA, Struhl K (2003)
Lysine-79 of histone H3 is hypomethylated at silenced loci in yeast and
mammalian cells: a potential mechanism for position-effect variega-
tion. Proc Natl Acad Sci USA 100: 1820 1825
9. Batta K, Zhang Z, Yen K, Goffman DB, Pugh BF (2011) Genome-wide
function of H2B ubiquitylation in promoter and genic regions. Genes
Dev 25: 2254 2265
10. Ng HH, Dole S, Struhl K (2003) The Rtf1 component of the Paf1 tran-
scriptional elongation complex is required for ubiquitination of histone
H2B. J Biol Chem 278: 33625 33628
11. Pokholok DK, Harbison CT, Levine S, Cole M, Hannett NM, Lee TI, Bell
GW, Walker K, Rolfe PA, Herbolsheimer E et al (2005) Genome-wide
map of nucleosome acetylation and methylation in yeast. Cell 122:
517 527
12. Sims RJ 3
rd, Nishioka K, Reinberg D (2003) Histone lysine methylation:
a signature for chromatin function. Trends Genet 19: 629 639
13. Dehe PM, Dichtl B, Schaft D, Roguev A, Pamblanco M, Lebrun R, Rodri-
guez-Gil A, Mkandawire M, Landsberg K, Shevchenko A et al (2006)
Protein interactions within the Set1 complex and their roles in the
regulation of histone 3 lysine 4 methylation. J Biol Chem 281:
35404 35412
14. Schneider J, Wood A, Lee JS, Schuster R, Dueker J, Maguire C, Swanson
SK, Florens L, Washburn MP, Shilatifard A (2005) Molecular regulation
of histone H3 trimethylation by COMPASS and the regulation of gene
expression. Mol Cell 19: 849 856
15. Miller T, Krogan NJ, Dover J, Erdjument-Bromage H, Tempst P, Johnston
M, Greenblatt JF, Shilatifard A (2001) COMPASS: a complex of proteins
associated with a trithorax-related SET domain protein. Proc Natl Acad
Sci USA 98: 12902 12907
16. Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G,
Alvarez P, Brockman W, Kim TK, Koche RP et al (2007) Genome-wide
maps of chromati n state in pluripotent and lineage-committed cells.
Nature 448: 553 560
17. Schneider R, Bannister AJ, Myers FA, Thorne AW, Crane-Robinson C,
Kouzarides T (2004) Histone H3 lysine 4 methylation patterns in higher
eukaryotic genes. Nat Cell Biol 6: 73 77
18. Ng HH, Robert F, Young RA, Struhl K (2003) Targeted recruitment of
Set1 histone methylase by elongating Pol II provides a localized mark
and memory of recent transcriptional activity. Mol Cell 11: 709
719
19. Song ZT, Sun L, Lu SJ, Tian Y, Ding Y, Liu JX (2015) Transcription factor
interaction with COMPASS-like complex regulates histone H3K4
trimethylation for specific gene expression in plants. Proc Natl Acad Sci
USA 112: 2900 2905
20. Katada S, Sassone-Corsi P (2010) The histone methyltransferase MLL1
permits the oscillation of circadian gene expression. Nat Struct Mol Biol
17: 1414 1421
21. Okuda H, Kawaguchi M, Kanai A, Matsui H, Kawamura T, Inaba T,
Kitabayashi I, Yokoyama A (2014) MLL fusion proteins link transcrip-
tional coactivators to previously active CpG-rich promoters. Nucleic
Acids Res 42: 4241 4256
22. Narayanan A, Ruyechan WT, Kristie TM (2007) The coactivator host cell
factor-1 mediates Set1 and MLL1 H3K4 trimethylation at herpes virus
immediate early promoters for initiation of infection. Proc Natl Acad Sci
USA 104: 10835 10840
23. Yokoyama A, Wang Z, Wysocka J, Sanyal M, Aufiero DJ, Kitabayashi I,
Herr W, Cleary ML (2004) Leukemia proto-oncoprotein MLL forms a
SET1-like histone methyltransferase complex with menin to regulate
Hox gene expression. Mol Cell Biol 24: 5639 5649
24. Deaton AM, Bird A (2011) CpG islands and the regulation of transcrip-
tion. Genes Dev 25: 1010 1022
25. Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G,
Chepelev I, Zhao K (2007) High-resolution profiling of histone methyla-
tions in the human genome. Cell 129: 823
837
26. Guenther MG, Levine SS, Boyer LA, Jaenisch R, Young RA (2007) A chro-
matin landmark and transcription initiation at most promoters in
human cells. Cell 130: 77 88
27. Hu D, Garruss AS, Gao X, Morgan MA, Cook M, Smith ER, Shilatifard A
(2013) The Mll2 branch of the COMPASS family regulates bivalent promot-
ers in mouse embryonic stem cells. Nat Struct Mol Biol 20: 1093 1097
28. Denissov S, Hofemeister H, Marks H, Kranz A, Ciotta G, Singh S, Anas-
tassiadis K, Stunnenberg HG, Stewart AF (2014) Mll2 is required for
H3K4 trimethylation on bivalent promoters in embryonic stem cells,
whereas Mll1 is redundant. Development 141: 526 537
29. Clouaire T, Webb S, Skene P, Illingworth R, Kerr A, Andrews R, Lee JH,
Skalnik D, Bird A (2012) Cfp1 integrates both CpG content and gene
activity for accurate H3K4me3 deposition in embryonic stem cells.
Genes Dev 26: 1714 1728
30. Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B,
Meissner A, Wernig M, Plath K et al (2006) A bivalent chromatin struc-
ture marks key developmental genes in embryonic stem cells. Cell 125:
315 326
31. Eberl HC, Spruijt CG, Kelstrup CD, Vermeulen M, Mann M (2013) A map
of general and specialized chromatin readers in mouse tissues gener-
ated by label-free interaction proteomics. Mol Cell 49 : 368 378
32. Shi X, Kachirskaia I, Walter KL, Kuo JH, Lake A, Davrazou F, Chan SM,
Martin DG, Fingerman IM, Briggs SD et al (2007) Proteome-wide analy-
sis in Saccharomyces cerevisiae identifies several PHD fingers as novel
direct and selective binding modules of histone H3 methylated at
either lysine 4 or lysine 36. J Biol Chem 282: 2450 2455
33. Murton BL, Chin WL, Ponting CP, Itzhaki LS (2010) Characterising the
binding specificities of the subunits associated with the KMT2/Set1
histone lysine methyltransferase. J Mol Biol 398: 481 488
34. Wang Z, Song J, Milne TA, Wang GG, Li H, Allis CD, Patel DJ (2010) Pro
isomerization in MLL1 PHD3-bromo cassette connects H3K4me readout
to CyP33 and HDAC-mediated repression. Cell 141: 1183 1194
35. Ali M, Hom RA, Blakeslee W, Ikenouye L, Kutateladze TG (2014) Diverse
functions of PHD fingers of the MLL/KMT 2 subfamily. Biochim Biophys
Acta 1843: 366 371
36. Schulze JM, Hentrich T, Nakanishi S, Gupta A, Emberly E, Shilatifard A,
Kobor MS (2011) Splitting the task: Ubp8 and Ubp10 deubiquitinate
different cellular pools of H2BK123. Genes Dev 25: 2242 2247
37. Sun ZW, Allis CD (2002) Ubiquitination of histone H2B regulates H3
methylation and gene silencing in yeast. Nature 418: 104 108
38. Ng HH, Xu RM, Zhang Y, Struhl K (
2002) Ubiquitination of histone H2B
by Rad6 is required for efficient Dot1-mediated methylation of histone
H3 lysine 79. J Biol Chem 277: 34655 34657
39. Guan X, Rastogi N, Parthun MR, Freitas MA (2013 ) Discover y of histone
modification crosstalk networks by stable isotope labeling of amino
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1477
acids in cell culture mass spectrometry (SILAC MS). Mol Cell Proteomics
12: 2048 2059
40. Kim SK, Jung I, Lee H, Kang K, Kim M, Jeong K, Kwon CS, Han YM, Kim
YS, Kim D et al (2012) Human histone H3K79 methyltransferase DOT1 L
protein [corrected] binds actively transcribing RNA polymerase II to
regulate gene expression. J Biol Chem 287: 39698 39709
41. Kim J, Hake SB, Roeder RG (2005) The human homolog of yeast BRE1
functions as a transcriptional coactivator through direct activator
interactions. Mol Cell 20: 759 770
42. Wu L, Zee BM, Wang Y, Garcia BA, Dou Y (2011) The RING finger
protein MSL2 in the MOF complex is an E3 ubiquitin ligase for H2B
K34 and is involved in crosstalk with H3K4 and K79 methylation. Mol
Cell 43: 132 144
43. Wu L, Lee SY, Zhou B, Nguyen UT, Muir TW, Tan S, Dou Y (2013) ASH2L
regulates ubiquitylation signaling to MLL: trans-regulation of H3K4
methylation in higher eukaryotes. Mol Cell 49: 1108 1120
44. Zeng L, Zhou MM (2002) Bromodomain: an acetyl-lysine binding
domain. FEBS Lett 513: 124 128
45. Yuan J, Pu M, Zhang Z, Lou Z (2009) Histone H3-K56
acetylation is
important for genomic stability in mammals. Cell Cycle 8: 1747 1753
46. Di Cerbo V, Mohn F, Ryan DP, Montellier E, Kacem S, Tropberger P,
Kallis E, Holzner M, Hoerner L, Feldmann A et al (2014) Acetylation of
histone H3 at lysine 64 regulates nucleosome dynamics and facilitates
transcription. elife 3:e01632
47. Tropberger P, Pott S, Keller C, Kamieniarz-Gdula K, Caron M, Richter F,
Li G, Mittler G, Liu ET, Buhler M et al (2013) Regulation of transcription
through acetylation of H3K122 on the lateral surface of the histone
octamer. Cell 152: 859 872
48. Gansen A, Toth K, Schwarz N, Langowski J (2015) Opposing roles of H3-
and H4-acetylation in the regulation of nucleosome structurea FRET
study. Nucleic Acids Res 43: 1433 1443
49. Bian C, Xu C, Ruan J, Lee KK, Burke TL, Tempel W, Barsyte D, Li J, Wu
M, Zhou BO et al (2011) Sgf29 binds histone H3K4me2/3 and is
required for SAGA complex recruitment and histone H3 acetylation.
EMBO J 30: 2829 2842
50. Taverna SD, Ilin S, Rogers RS, Tanny JC, Lavender H, Li H, Baker L, Boyle J,
Blair LP, Chait BT et al (2006) Yng1 PHD finger binding to H3 trimethy-
lated at K4
promotes NuA3 HAT activity at K14 of H3 and transcription
at a subset of targeted ORFs. Mol Cell 24: 785 796
51. Doyon Y, Selleck W, Lane WS, Tan S, Cote J (2004) Structural and func-
tional conservation of the NuA4 histone acetyltransferase complex
from yeast to humans. Mol Cell Biol 24: 1884 1896
52. Hung T, Binda O, Champagne KS, Kuo AJ, Johnson K, Chang HY, Simon
MD, Kutateladze TG, Gozani O (2009) ING4 mediates crosstalk between
histone H3K4 trimethylation and H3 acetylation to attenuate cellular
transformation. Mol Cell 33: 248 256
53. Crump NT, Hazzalin CA, Bowers EM, Alani RM, Cole PA, Mahadevan LC
(2011) Dynamic acetylation of all lysine-4 trimethylated histone H3 is
evolutionarily conserved and mediated by p300/CBP. Proc Natl Acad Sci
USA 108: 7814 7819
54. Hsu DW, Chubb JR, Muramoto T, Pears CJ, Mahadevan LC (2012)
Dynamic acetylation of lysine-4-trimethylated histone H3 and H3 vari-
ant biology in a simple multicellular eukaryote. Nucleic Acids Res 40:
7247 7256
55. Clayton AL, Hazzalin CA, Mahadevan LC (2006) Enhanced histone acety-
lation and transcription: a dynamic perspective. Mol Cell 23: 289 296
56. Doyon Y, Cayrou C, Ullah M, Landry AJ, Cote V, Selleck W, Lane WS, Tan
S, Yang XJ, Cote J (2006
) ING tumor suppressor proteins are critical
regulators of chromatin acetylation required for genome expression
and perpetuation. Mol Cell 21: 51 64
57. Wagner EJ, Carpenter PB (2012) Understanding the language of Lys36
methylation at histone H3. Nat Rev Mol Cell Biol 13: 115 126
58. Li B, Howe L, Anderson S, Yates JR 3rd, Workman JL (2003) The Set2
histone methyltransferase functions through the phosphorylated
carboxyl-terminal domain of RNA polymerase II. J Biol Chem 278:
8897 8903
59. Kizer KO, Phatnani HP, Shibata Y, Hall H, Greenleaf AL, Strahl BD (2005)
A novel domain in Set2 mediates RNA polymerase II interaction and
couples histone H3K36 methylation with transcript elongation. Mol Cell
Biol 25: 3305 3316
60. Morris SA, Shibata Y, Noma K, Tsukamoto Y, Warren E, Temple B,
Grewal SI, Strahl BD (2005) Histone H3K36 methylation is associated
with transcription elongation in Schizosaccharomyces pombe. Eukaryot
Cell 4: 1446 1454
61. Joshi AA, Struhl K (2005) Eaf3 chromodomain interaction with methy-
lated H3-K36 links histone deacetylation to Pol II elongation. Mol Cell
20: 971 978
62. Carrozza MJ, Li B, Florens L, Suganuma T, Swanson SK, Lee KK, Shia WJ,
Anderson S, Yates J, Washburn MP et al (2005) Histone H3 methylation
by Set2
directs deacetylation of coding regions by Rpd3S to suppress
spurious intragenic transcription. Cell 123: 581 592
63. Keogh MC, Kurdistani SK, Morris SA, Ahn SH, Podolny V, Collins SR,
Schuldiner M, Chin K, Punna T, Thompson NJ et al (2005) Cotranscrip-
tional set2 methylation of histone H3 lysine 36 recruits a repressive
Rpd3 complex. Cell 123: 593 605
64. Blackledge NP, Zhou JC, Tolstorukov MY, Farcas AM, Park PJ, Klose RJ
(2010) CpG islands recruit a histone H3 lysine 36 demethylase. Mol Cell
38: 179 190
65. He J, Kallin EM, Tsukada Y, Zhang Y (2008) The H3K36 demethylase
Jhdm1b/Kdm2b regulates cell proliferation and senescence through p15
(Ink4b). Nat Struct Mol Biol 15: 1169 1175
66. Dhayalan A, Rajavelu A, Rathert P, Tamas R, Jurkowska RZ, Ragozin S,
Jeltsch A (2010) The Dnmt3a PWWP domain reads histone 3 lysine 36
trimethylation and guides DNA methylation. J Biol Chem 285:
26114 26120
67. van Nuland R, van Schaik FM, Simonis M, van Heesch S, Cuppen E,
Boelens R, Timmers HM, van Ingen H (2013) Nucleosomal DNA binding
drives the recognition of H3K36-methylated nucleosomes by the PSIP1-
PWWP domain.
Epigenetics Chromatin 6: 12
68. Maltby VE, Martin BJ, Schulze JM, Johnson I, Hentrich T, Sharma A,
Kobor MS, Howe L (2012) Histone H3 lysine 36 methylation targets the
Isw1b remodeling complex to chromatin. Mol Cell Biol 32: 3479 3485
69. Vermeulen M, Eberl HC, Matarese F, Marks H, Denissov S, Butter F, Lee
KK, Olsen JV, Hyman AA, Stunnenberg HG et al (2010) Quantitative
interaction proteomics and genome-wide profiling of epigenetic
histone marks and their readers. Cell 142: 967 980
70. Stec I, Nagl SB, van Ommen GJ, den Dunnen JT (2000) The PWWP
domain: a potential protein-protein interaction domain in nuclear
proteins influencing differentiation? FEBS Lett 473: 1 5
71. Kim A, Kiefer CM, Dean A (2007) Distinctive signatures of histone
methylation in transcribed coding and noncoding human beta-globin
sequences. Mol Cell Biol 27: 1271 1279
72. Schneider TD, Arteaga-Salas JM, Mentele E, David R, Nicetto D, Imhof A,
Rupp RA (2011) Stage-specific histone modification profiles reveal
global transitions in the Xenopus embryonic epigenome. PLoS ONE 6:
e22548
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1478
73. Zheng Y, Sweet SM, Popovic R, Martinez-Garcia E, Tipton JD, Thomas
PM, Licht JD, Kelleher NL (2012) Total kinetic analysis reveals how
combinatorial methylation patterns are established on lysines 27 and
36 of histone H3. Proc Natl Acad Sci USA 109: 13549 13554
74. Martinez-Garcia E, Popovic R, Min DJ, Sweet SM, Thomas PM,
Zamdborg L, Heffner A, Will C, Lamy L, Staudt LM et al (2011) The
MMSET histone methyl transferase switches global histone methyla-
tion and alters gene expression in t(4;14) multiple myeloma cells. Blood
117: 211 220
75. Jaffe JD, Wang Y, Chan HM, Zhang J, Huether R, Kryukov GV, Bhang HE,
Taylor JE, Hu M, Englund NP et al (2013) Global chromatin profiling
reveals NSD2 mutations in pediatric acute lymphoblastic leukemia. Nat
Genet 45: 1386 1391
76. Schmitges FW, Prusty AB, Faty M, Stutzer A, Lingaraju GM, Aiwazian J,
Sack R, Hess D, Li L, Zhou S et al (2011) Histone methylation by PRC2
is inhibited by active chromatin marks. Mol Cell 42: 330 341
77. Yuan W, Xu M, Huang C, Liu N, Chen S, Zhu B (2011)H3K36 methyla-
tion antagonizes PRC2-mediated H3K27 methylation. J Biol Chem 286:
7983 7989
78. Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D
(2002) Histone methyltransferase activity associated with a human
multiprotein complex containing the Enhancer of Zeste protein. Genes
Dev 16: 2893
2905
79. Cao R, Zhang Y (2004) SUZ12 is required for both the histone methyl-
transferase activity and the silencing function of the EED-EZH2
complex. Mol Cell 15: 57 67
80. Nekrasov M, Wild B, Muller J (2005) Nucleosome binding and histone
methyltransferase activity of Drosophila PRC2. EMBO Rep 6: 348 353
81. Han Z, Xing X, Hu M, Zhang Y, Liu P, Chai J (2007) Structural basis of
EZH2 recognition by EED. Structure 15: 1306 1315
82. Alekseyenko AA, Gorchakov AA, Kharchenko PV, Kuroda MI (2014) Recip-
rocal interactions of human C10orf12 and C17orf96 with PRC2 revealed
by BioTAP-XL cross-linking and affinity purification. Proc Natl Acad Sci
USA 111: 2488 2493
83. Ferrari KJ, Scelfo A, Jammula S, Cuomo A, Barozzi I, Stutzer A, Fischle
W, Bonaldi T, Pasini D (2014) Polycomb-dependent H3K27me 1 and
H3K27me2 regulate active transcription and enhancer fidelity. Mol Cell
53: 49 62
84. Jung HR, Pasini D, Helin K, Jensen ON (2010) Quantitative mass spec-
trometry of histones H3.2 and H3.3
in Suz12-deficient mouse embry-
onic stem cells reveals distinct, dynamic post-translational
modifications at Lys-27 and Lys-36. Mol Cell Proteomics 9: 838 850
85. Bracken AP, Dietrich N, Pasini D, Hansen KH, Helin K (2006) Genome-
wide mapping of Polycomb target genes unravels their roles in cell fate
transitions. Genes Dev 20: 1123 1136
86. Silva J, Mak W, Zvetkova I, Appanah R, Nesterova TB, Webster Z, Peters
AH, Jenuwein T, Otte AP, Brockdorff N (2003) Establishment of histone h3
methylation on the inactive X chromosome requires transient recruit-
ment of Eed-Enx1 polycomb group complexes. Dev Cell 4: 481 495
87. Pauler FM, Sloane MA, Huang R, Regha K, Koerner MV, Tamir I,
Sommer A, Aszodi A, Jenuwein T, Barlow DP (2009)H3K27me3 forms
BLOCs over silent genes and intergenic regions and specifies a histone
banding pattern on a mouse autosomal chromosome. Genome Res 19:
221 233
88. Margueron R, Justin N, Ohno K, Sharpe ML, Son J, Drury WJ 3rd, Voigt
P, Martin SR, Taylor WR, De Marco V et al (2009) Role of the polycomb
protein EED in the propagation of repressive histone marks. Nature
461: 762 767
89. Hansen KH, Bracken AP, Pasini D, Dietrich N, Gehani SS, Monrad A,
Rappsilber J, Lerdrup M, Helin K (2008) A model for transmission of the
H3K27me3 epigenetic mark. Nat Cell Biol 10: 1291 1300
90. Yuan W, Wu T, Fu H, Dai C, Wu H, Liu N, Li X, Xu M, Zhang Z, Niu T
et al (2012) Dense chromatin activates Polycomb repressive complex 2
to regulate H3
lysine 27 methylation. Science 337: 971 975
91. Gao Z, Zhang J, Bonasio R, Strino F, Sawai A, Parisi F, Kluger Y,
Reinberg D (2012) PCGF homologs, CBX proteins, and RYBP define
functionally distinct PRC1 family complexes. Mol Cell 45: 344 356
92. Morey L, Pascual G, Cozzuto L, Roma G, Wutz A, Benitah SA, Di Croce L
(2012) Nonoverlapping functions of the Polycomb group Cbx family of
proteins in embryonic stem cells. Cell Stem Cell 10: 47 62
93. Tavares L, Dimitrova E, Oxley D, Webster J, Poot R, Demmers J, Bezs-
tarosti K, Taylor S, Ura H, Koide H et al (2012) RYBP-PRC1 complexes
mediate H2A ubiquitylation at polycomb target sites independently of
PRC2 and H3K27me3. Cell 148: 664 678
94. Morey L, Aloia L, Cozzuto L, Benitah SA, Di Croce L (2013) RYBP and
Cbx7 define specific biological functions of polycomb complexes in
mouse embryonic stem cells. Cell Rep 3: 60 69
95. Arrigoni R, Alam SL, Wamstad JA, Bardwell VJ, Sundquist WI, Schreiber-
Agus N (2006) The Polycomb-associated protein Rybp is a ubiquitin
binding protein. FEBS Lett 580: 6233 6241
96. Ku M, Koche RP, Rheinbay E, Mendenhall EM, Endoh M, Mikkelsen TS,
Presser A, Nusbaum C, Xie X, Chi AS et al (2008) Genomewide analysis
of PRC1 and PRC2 occupancy identifies two classes of bivalent
domains.
PLoS Genet 4:e1000242
97. Plath K, Fang J, Mlynarczyk-Evans SK, Cao R, Worringer KA, Wang H,
de la Cruz CC, Otte AP, Panning B, Zhang Y (2003) Role of histone H3
lysine 27 methylation in X inactivation. Science 300: 131 135
98. Mak W, Baxter J, Silva J, Newall AE, Otte AP, Brockdorff N (2002) Mitoti-
cally stable association of polycomb group proteins eed and enx1 with
the inactive x chromosome in trophoblast stem cells. Curr Biol 12:
1016 1020
99. de Napoles M, Mermoud JE, Wakao R, Tang YA, Endoh M, Appanah R,
Nesterova TB, Silva J, Otte AP, Vidal M et al (2004) Polycomb group
proteins Ring1A/B link ubiquitylation of histone H2A to heritable gene
silencing and X inactivation. Dev Cell 7: 663 676
100. Wang L, Brown JL, Cao R, Zhang Y, Kassis JA, Jones RS (2004) Hierarchi-
cal recruitment of polycomb group silencing complexes. Mol Cel l 14:
637 646
101. Ren X, Vincenz C, Kerppola TK (2008) Changes in the distributions and
dynamics of polycomb repressive complexes during embryonic stem
cell differentiation. Mol Cell Biol 28: 2884 2895
102. Schoeftner S, Sengupta AK, Kubicek S, Mechtler K, Spahn L, Koseki H,
Jenuwein T, Wutz A (2006) Recruitment of PRC1 function at the initia-
tion of X inactivation independent of PRC2 and silencing. EMBO J 25 :
3110 3122
103. Cooper S, Dienstbier M, Hassan R, Schermelleh L, Sharif J, Blackledge NP,
De Marco V, Elderkin S, Koseki H, Kl ose R et al (2014) Targeting polycomb
to pericentric heterochromatin in embryonic stem cells reveals a role for
H2AK119
u1 in PRC2 recruitment. Cell Rep 7: 1456 1470
104. Blackledge NP, Farcas AM, Kondo T, King HW, McGouran JF, Hanssen
LL, Ito S, Cooper S, Kondo K, Koseki Y et al (2014) Variant PRC1
complex-dependent H2A ubiquitylation drives PRC 2 recruitment and
polycomb domain formation. Cell 157: 1445 1459
105. Kalb R, Latwiel S, Baymaz HI, Jansen PW, Muller CW, Vermeulen M,
Muller J (2014) Histone H2A monoubiquitination promotes histone H3
methylation in Polycomb repression. Nat Struct Mol Biol 21: 569 571
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1479
106. Farcas AM, Blackledge NP, Sudbery I, Long HK, McGouran JF, Rose NR,
Lee S, Sims D, Cerase A, Sheahan TW et al (2012) KDM2B links the
Polycomb Repressive Complex 1 (PRC1) to recognition of CpG islands.
elife 1:e00205
107. Wu X, Johansen JV, Helin K (2013) Fbxl10/Kdm2b recruits polycomb
repressive complex 1 to CpG islands and regulates H2A ubiquitylation.
Mol Cell 49: 1134 1146
108. He J, Shen L, Wan M, Taranova O, Wu H, Zhang Y (2013) Kdm2b main-
tains murine embryonic stem cell status by recruiting PRC1 complex
to CpG islands of developmental genes. Nat Cell Biol 15: 373 384
109. Shinkai Y, Tachibana M (2011)H3K9 methyltransferase G9a and the
related molecule GLP. Genes Dev 25: 781 788
110. Tachibana M, Ueda J, Fukuda M, Takeda N, Ohta T, Iwanari H, Sakihama
T, Kodama T, Hamakubo T, Shinkai Y (2005) Histone methyltransferases
G9a and GLP form heteromeric complexes and are both crucial for
methylation of euchromatin at H3-K9. Genes Dev 19: 815 826
111. Collins RE, Northrop JP, Horton JR, Lee DY, Zhang X, Stallcup MR,
Cheng X (2008) The ankyrin repeats of G9a and GLP histone methyl-
transferases are mono- and dimethyllysine binding modules. Nat Struct
Mol Biol 15:
245 250
112. Karimi MM, Goyal P, Maksakova IA, Bilenky M, Leung D, Tang JX,
Shinkai Y, Mager DL, Jones S, Hirst M et al (2011) DNA methylation
and SETDB1/H3K9me3 regulate predominantly distinct sets of genes,
retroelements, and chimeric transcripts in mESCs. Cell Stem Cell 8:
676 687
113. Wang H, An W, Cao R, Xia L, Erdjument-Bromage H, Chatton B, Tempst
P, Roeder RG, Zhang Y (2003) mAM facilitates conversion by ESET of
dimethyl to trimethyl lysine 9 of histone H3 to cause transcriptional
repression. Mol Cell 12: 475 487
114. Dodge JE, Kang YK, Beppu H, Lei H, Li E (2004) Histone H3-K9 methyl-
transferase ESET is essential for early development. Mol Cell Biol 24:
2478 2486
115. Bilodeau S, Kagey MH, Frampton GM, Rahl PB, Young RA (2009) SetDB1
contributes to repression of genes encoding developmental regulators
and maintenan ce of ES cell state. Genes Dev 23: 2484 2489
116. Schotta G, Ebert A, Reuter G (2003) SU(VAR)3-9 is a conserved key func-
tion in heterochromatic gene silencing. Genetica 117: 149 158
117. Lehnertz B, Ueda Y, Derijck AA, Braunschweig U, Perez-Burgos L, Kubicek
S, Chen T, Li E, Jenuwein T, Peters AH (2003) Suv39 h-mediated histone
H3 lysine 9 methylation directs DNA methylation to major satellite
repeats at pericentric heterochromatin. Curr Biol 13:
1192 1200
118. Fuks F, Hurd PJ, Deplus R, Kouzarides T (2003) The DNA methyltrans-
ferases associate with HP1 and the SUV39H1 histone met hyltrans-
ferase. Nucleic Acids Res 31: 2305 2312
119. Fuks F, Hurd PJ, Wolf D, Nan X, Bird AP, Kouzarides T (2003) The
methyl-CpG-binding protein MeCP2 links DNA methylation to histone
methylation. J Biol Chem 278: 4035 4040
120. Hirota T, Lipp JJ, Toh BH, Peters JM (2005) Histone H3 serine 10 phos-
phorylation by Aurora B causes HP1 dissociation from heterochro-
matin. Nature 438: 1176 1180
121. Peters AH, Kubicek S, Mechtler K, OSullivan RJ, Derijck AA, Perez-Burgos
L, Kohlmaier A, Opravil S, Tachibana M, Shinkai Y et al (2003) Partition-
ing and plasticity of repressive histone methylation states in mamma-
lian chro matin. Mol Cell 12: 1577 1589
122. Saksouk N, Barth TK, Ziegler-Birling C, Olova N, Nowak A, Rey E,
Mateos-Langerak J, Urbach S, Reik W, Torres-Padilla ME et al (2014)
Redundant mechanisms to form silent chromatin at pericentromeric
regions rely on BEND3 and DNA methylation. Mol Cell 56: 580 594
123. Puschendorf M, Terranova R, Boutsma E, Mao X, Isono K, Brykczynska
U, Kolb C, Otte AP, Koseki H, Orkin SH et al (2008) PRC1 and Suv39h
specify parental asymmetry at constitutive heterochromatin in early
mouse embryos. Nat Genet 40: 411
420
124. Tardat M, Albert M, Kunzmann R, Liu Z, Kaustov L, Thierry R,
Duan S, Brykczyns ka U, Arrowsmith CH, Peters AH (2015) Cbx2 targets
PRC1 to constitutive heterochromatin in mouse zygotes in a parent-
of-origin-dependent manner. Mol Cell 58: 157 171
125. Alder O, Lavial F, Helness A, Brookes E, Pinho S, Chandrashekran A,
Arnaud P, Pombo A, ONeill L, Azuara V (2010) Ring1B and Suv39h1
delineate distinct chromatin states at bivalent genes during early
mouse lineage commitment. Development 137: 2483 2492
126. Hawkins RD, Hon GC, Lee LK, Ngo Q, Lister R, Pelizzola M, Edsall LE,
Kuan S, Luu Y, Klugman S et al (2010) Distinct epigenomic landscapes
of pluripotent and lineage-committed human cells. Cell Stem Cell 6:
479 491
127. Voigt P, LeRoy G, Drury WJ 3rd, Zee BM, Son J, Beck DB, Young NL,
Garcia BA, Reinberg D (2012) Asymmetrically modified nucleosomes.
Cell 151: 181 193
128. Mozzetta C, Pontis J, Fritsch L, Robin P, Portoso M, Proux C, Margueron
R, Ait-Si-Ali S (2014) The histone H3 lysine 9 methyltransferases G9a
and GLP regulate polycomb repressive complex 2-mediated gene
silencing. Mol Cell 53: 277 289
129. Boros J, Arnoult N, Stroobant V, Collet JF, Decottignies A (2014) Poly-
comb repressive complex 2 and H3K27me3 cooperate with H3K
9
methylation to maintain heterochromatin protein 1alpha at chromatin.
Mol Cell Biol 34: 3662 3674
130. de la Cruz CC, Kirmizis A, Simon MD, Isono K, Koseki H, Panning B (2007)
The polycomb group protein SUZ12 regulates histone H3 lysine 9
methylation and HP1 alpha distribution. Chromosome Res 15: 299 314
131. Yamamoto K, Sonoda M, Inokuchi J, Shirasawa S, Sasazuki T (2004)
Polycomb group suppressor of zeste 12 links heterochromatin protein
1alpha and enhancer of zeste 2. J Biol Chem 279: 401 406
132. Rougeulle C, Chaumeil J, Sarma K, Allis CD, Reinberg D, Avner P, Heard
E(2004) Differential histone H3 Lys-9 and Lys-27 methylation profiles
on the X chromosome. Mol Cell Biol 24: 5475 5484
133. Escamilla-Del-Arenal M, da Rocha ST, Spruijt CG, Masui O, Renaud O,
Smits AH, Margueron R, Vermeulen M, Heard E (2013) Cdyl, a new
partner of the inactive X chromosome and potential reader of
H3K27me3 and H3K9me2. Mol Cell Biol 33 : 5005 5020
134. Wang J, Lawry ST, Cohen AL, Jia S (2014) Chromosome boundary
elements and regulation of heterochromatin spreading. Cell Mol Life Sci
71: 4841 4852
135. Elgin SC, Reuter G (
2013) Position-effect variegation, heterochromatin
formation, and gene silencing in Drosophila. Cold Spring Harb Perspect
Biol 5:a017780
136. Narendra V, Rocha PP, An D, Raviram R, Skok JA, Mazzoni EO, Reinberg
D(2015) Transcrip tion. CTCF establishes discrete functional chromatin
domains at the Hox clusters during differentiation. Science 347:
1017 1021
137. Jung HR, Sidoli S, Haldbo S, Sprenger RR, Schwammle V, Pasini D, Helin
K, Jensen ON (2013) Precision mapping of coexisting modifications in
histone H3 tails from embryonic stem cells by ETD-MS/MS. Anal Chem
85: 8232 8239
138. Gaydos LJ, Rechtsteiner A, Egelhofer TA, Carroll CR, Strome S (2012)
Antagonism between MES-4 and Polycomb repressive complex 2
promotes appropriate gene expression in C. elegans germ cells. Cell Rep
2: 1169 1177
EMBO reports Vol 16 |No11 | 2015 ª 2015 The Authors
EMBO reports The interplay of histone modifications Tianyi Zhang et al
1480
139. Bender LB, Suh J, Carroll CR, Fong Y, Fingerman IM, Briggs SD, Cao R,
Zhang Y, Reinke V, Strome S (2006) MES-4: an autosome-associated
histone methyltransferase that participates in silencing the X chromo-
somes in the C. elegans germ line. Development 133: 3907 3917
140. Rechtsteiner A, Ercan S, Takasaki T, Phippen TM, Egelhofer TA, Wang
W, Kimura H, Lieb JD, Strome S (2010) The histone H3K36 methyltrans-
ferase MES-4 acts epigenetically to transmit the memory of germline
gene expression to progeny. PLoS Genet 6:e1001091
141. Yuan G, Ma B, Yuan W, Zhang Z, Chen P, Ding X, Feng L, Shen X, Chen S,
Li G et al (2013) Histone H2A ubiquitination inhibits the enzymatic activ-
ity of H3 lysine 36 methyltransferases. J Biol Chem 288: 30832 30842
142. Endoh M, Endo TA, Endoh T, Isono K, Sharif J, Ohara O, Toyoda T, Ito T,
Eskeland R, Bickmore WA et al (2012) Histone H2A mono-ubiquitination
is a crucial step to mediate PRC1-dependent repression of developmen-
tal genes to maintain ES cell identity. PLoS Genet 8:e1002774
143. Kim DH, Tang Z, Shimada M, Fierz B, Houck-Loomis B, Bar-Dagen M,
Lee S, Lee SK, Muir TW, Roede r RG et al (2013) Histone H3K27
trimethylation inhibits H3 binding and function of SET1-like H3K4
methyltransferase complexes. Mol Cell Biol 33: 4936 4946
144. Cai L, Rothbart SB, Lu R, Xu B, Chen WY, Tripathy A, Rockowitz S,
Zheng D, Patel DJ, Allis CD et al (2013
)AnH3K36 methylation-engaging
Tudor motif of polycomb-like proteins mediates PRC2 complex target-
ing. Mol Cell 49: 571 582
145. Musselman CA, Avvakumov N, Watanabe R, Abraham CG, Lalonde ME,
Hong Z, Allen C, Roy S, Nunez JK, Nickoloff J et al (2012) Molecular
basis for H3K36me3 recognition by the Tudor domain of PHF1. Nat
Struct Mol Biol 19: 1266 1272
146. Brien GL, Gambero G, OConnell DJ, Jerman E, Turner SA, Egan CM,
Dunne EJ, Jurgens MC, Wynne K, Piao L et al (2012) Polycomb PHF19
binds H3K36me3 and recruits PRC2 and demethylase NO66 to embry-
onic stem cell genes during differentiation. Nat Struct Mol Biol 19:
1273 1281
147. Ballare C, Lange M, Lapinaite A, Martin GM, Morey L, Pascual G, Liefke
R, Simon B, Shi Y, Gozani O et al (2012) Phf19 links methylated Lys36
of histone H3 to regulation of Polycomb activity. Nat Struct Mol Biol
19: 1257 1265
148. Reynolds N, Salmon-Divon M, Dvinge H, Hynes-Allen A, Balasooriya G,
Leaford D, Behrens A, Bertone P, Hendrich B (2012) NuRD-mediated
deacetylation of H3K27 facilitates recruitment of Polycomb Repressive
Complex 2 to direct gene repression. EMBO J 31:
593 605
149. Kim TW, Kang BH, Jang H, Kwak S, Shin J, Kim H, Lee SE, Lee SM, Lee
JH, Kim JH et al (2015) Ctbp2 modulates NuRD-mediated deacetylat ion
of H3K27 and facilitates PRC2-mediated H3K27me3 in active embryonic
stem cell genes during exit from pluripotency. Stem Cells 33:
2442 2455
150. Lau PN, Cheung P (2011) Histone code pathway involving H3 S28
phosphorylation and K27 acetylation activates transcription and antag-
onizes polycomb silencing. Proc Natl Acad Sci USA 108: 2801 2806
151. Gehani SS, Agrawal-Singh S, Dietrich N, Christophersen NS, Helin K,
Hansen K (2010) Polycomb group protein displacement and gene acti-
vation through MSK-dependent H3K27me3S28 phosphorylation. Mol
Cell 39: 886 900
152. Pasini D, Malatesta M, Jung HR, Walfridsson J, Willer A, Olsson L, Skotte J,
Wutz A, Porse B, Jensen ON et al (2010) Characterization of an antago-
nistic switch between histone H3 lysine 27 methylation and acetylation
in the transcriptional regulation of Polycomb group target genes. Nucleic
Acids Res 38: 4958 4969
153. Turkmen S, Gillessen-Kaesbach G, Meinecke P, Albrecht B, Neumann
LM, Hesse V, Palanduz S, Balg S, Majewski F, Fuchs S et al (2003)
Mutations in NSD1 are responsible for Sotos syndrome, but are not a
frequent finding in other overgrowth phenotypes.
Eur J Hum Genet 11:
858 865
154. Rio M, Clech L, Amiel J, Faivre L, Lyonnet S, Le Merrer M, Odent S,
Lacombe D, Edery P, Brauner R et al (2003) Spectrum of NSD1 muta-
tions in Sotos and Weaver syndromes. J Med Genet 40: 436 440
155. Douglas J, Coleman K, Tatton-Brown K, Hughes HE, Temple IK, Cole TR,
Rahman N, Childhood Overgrowth C (2005) Evaluation of NSD2 and
NSD3 in overgrowth syndromes. Eur J Hum Genet 13: 150 153
156. Morishita M, di Luccio E (2011) Cancers and the NSD family of histone
lysine methyltransferases. Biochim Biophys Acta 1816: 158 163
157. Kanu N, Gronroos E, Martinez P, Burrell RA, Yi Goh X, Bartkova J,
Maya-Mendoza A, Mistrik M, Rowan AJ, Patel H et al (2015) SETD2
loss-of-function promotes renal cancer branched evolution through
replication stress and impaired DNA repair. Oncogene doi: 10 .1038/
onc.2015.24
158. Hock H (2012) A complex Polycomb issue: the two faces of EZH2 in
cancer. Genes Dev 26: 751 755
159. Chan KM, Fang D, Gan H, Hashizume R, Yu C, Schroeder M, Gupta N,
Mueller S, James CD, Jenkins R et al (2013) The histone H3.3K27M
mutation in pediatric glioma reprograms H3K27 methylation and gene
expression. Genes Dev
27: 985 990
160. Garcia BA, Thomas CE, Kelleher NL, Mizzen CA (2008) Tissue-specific
expression and post-translational modification of histone H3 variants.
J Proteome Res 7: 4225 4236
161. Wirbelauer C, Bell O, Schubeler D (2005) Variant histone H3. 3 is depos-
ited at sites of nucleosomal displacement throughout transcribed
genes while active histone modifications show a promoter-proximal
bias. Genes Dev 19: 1761 1766
162. Raisner RM, Hartley PD, Meneghini MD, Bao MZ, Liu CL, Schreiber SL,
Rando OJ, Madhani HD (2005) Histone variant H2A.Z marks the 5
0
ends
of both active and inactive genes in euchromatin. Cell 123: 233 248
163. Wen H, Li Y, Xi Y, Jiang S, Stratton S, Peng D, Tanaka K, Ren Y, Xia Z,
Wu J et al (2014) ZMYND11 links histone H3.3K36me3 to transcription
elongation and tumour suppression. Nature 508: 263 268
164. Jacob Y, Bergamin E, Donoghue MT, Mongeon V, LeBlanc C, Voigt P,
Underwood CJ, Brunzelle JS, Michaels SD, Reinberg D et al (2014)
Selective methylation of histone H3 variant H3.1 regulates hetero-
chromatin replication. Science 343: 1249 1253
License: This is an open access article under the
terms of the Creative Commons Attribution 4.0
License, which permits use, distribution and reproduc-
tion in any medium, provided the original work is
properly cited.
ª 2015 The Authors EMBO reports Vol 16 |No11 | 2015
Tianyi Zhang et al The interplay of histone modifications EMBO reports
1481
... Counts were obtained with HTSeq-count [40,60] using the NCBI Sscrofa11.1 annotation features (GTF) genome file and with the mode option set to intersection-nonempty. Limma [41,61] was used to obtain normalized read count data for each count file using a P value adjusted threshold of 0.05 (Benjamini-Hochberg correction) and trimmed mean of M values (TMM) normalization [42]. Tissuespecific genes were identified as previously described [43,44]. ...
... In both PFFs and PTr2s, several thousand (8,705 in PFFs and 7,416 in PTr2s) H3K4me3 and H3K27ac enrichment sites had overlapping genomic coordinates. The interplay between these PTMs has been investigated [60][61][62] and it has been posited that the upstream presence of H3K27ac may guide the installation of H3K4me3 at promoters by the acetylated histone reader BRD2 [62], though investigations into how H3K4me3 and H3K27ac may cooperate to influence transcription are ongoing. H3K27me3, which is generally associated with transcriptional repression, showed lower enrichment at gene start and end sites compared to H3K4me3 and H3K27ac, mainly appearing in intergenic regions. ...
Article
Full-text available
Background Given their physiological similarities to humans, pigs are increasingly used as model organisms in human-oriented biomedical studies. Additionally, their value to animal agriculture across the globe has led to the development of numerous studies to investigate how to improve livestock welfare and production efficiency. As such, pigs are uniquely poised as compelling models that can yield findings with potential implications in both human and animal contexts. Despite this, many gaps remain in our knowledge about the foundational mechanisms that govern gene expression in swine across different developmental stages, particularly in early development. To address some of these gaps, we profiled the histone marks H3K4me3, H3K27ac, and H3K27me3 and the SWI/SNF central ATPase BRG1 in two porcine cell lines representing discrete early developmental time points and used the resulting information to construct predicted chromatin state maps for these cells. We combined this approach with analysis of publicly available RNA-seq data to examine the relationship between epigenetic status and gene expression in these cell types. Results In porcine fetal fibroblast (PFF) and trophectoderm cells (PTr2), we saw expected patterns of enrichment for each of the profiled epigenetic features relative to specific genomic regions. H3K4me3 was primarily enriched at and around global gene promoters, H3K27ac was enriched in promoter and intergenic regions, H3K27me3 had broad stretches of enrichment across the genome and narrower enrichment patterns in and around the promoter regions of some genes, and BRG1 primarily had detectable enrichment at and around promoter regions and in intergenic stretches, with many instances of H3K27ac co-enrichment. We used this information to perform genome-wide chromatin state predictions for 10 different states using ChromHMM. Using the predicted chromatin state maps, we identified a subset of genomic regions marked by broad H3K4me3 enrichment, and annotation of these regions revealed that they were highly associated with essential developmental processes and consisted largely of expressed genes. We then compared the identities of the genes marked by these regions to genes identified as cell-type-specific using transcriptome data and saw that a subset of broad H3K4me3-marked genes was also specifically expressed in either PFF or PTr2 cells. Conclusions These findings enhance our understanding of the epigenetic landscape present in early swine development and provide insight into how variabilities in chromatin state are linked to cell identity. Furthermore, this data captures foundational epigenetic details in two valuable porcine cell lines and contributes to the growing body of knowledge surrounding the epigenetic landscape in this species.
... This model has the nucleosome with DNA wrapped around it, D, as a basic unit that can be modified either with activating marks, such as H3K4 methylation (H3K4me3) or H3K4 acetylation (H3K4ac), or repressive marks, such as H3K9 methylation (H3K9me3) or DNA methylation. H3K4me3 and H3K4ac are two histone modifications that promote a less compact DNA around the nucleosomes and they are then associated with gene activation (see Chapter 3 of [1] and [33]). In the model, it is assumed that H3K4me3 and H3K4ac co-exist and the nucleosome with either of these modifications is represented by D A . ...
Preprint
Full-text available
Epigenetic cell memory, the inheritance of gene expression patterns across subsequent cell divisions, is a critical property of multi-cellular organisms. In recent work [10], a subset of the authors observed in a simulation study how the stochastic dynamics and time-scale differences between establishment and erasure processes in chromatin modifications (such as histone modifications and DNA methylation) can have a critical effect on epigenetic cell memory. In this paper, we provide a mathematical framework to rigorously validate and extend beyond these computational findings. Viewing our stochastic model of a chromatin modification circuit as a singularly perturbed, finite state, continuous time Markov chain, we extend beyond existing theory in order to characterize the leading coefficients in the series expansions of stationary distributions and mean first passage times. In particular, we characterize the limiting stationary distribution in terms of a reduced Markov chain, provide an algorithm to determine the orders of the poles of mean first passage times, and determine how changing erasure rates affects system behavior. The theoretical tools developed in this paper not only allow us to set a rigorous mathematical basis for the computational findings of our prior work, highlighting the effect of chromatin modification dynamics on epigenetic cell memory, but they can also be applied to other singularly perturbed Markov chains beyond the applications in this paper, especially those associated with chemical reaction networks.
... Histones play important roles in normal development. Mutations or dysregulation of histones are associated with diseases and various cancers [23]. Recent studies have shown that HAT and HDAC can influence acetylated proteins involved in metabolism to coordinate cellular energy metabolism, redox status, and gene expression [24]. ...
Article
Full-text available
Osteoporosis (OP) is a common age-related disease. OP is mainly a decrease in bone density and mass caused by the destruction of bone microstructure, which leads to an increase in bone fragility. SIRT3 is a mitochondrial deacetylase that plays critical roles in mitochondrial homeostasis, metabolic regulation, gene transcription, stress response, and gene stability. Studies have shown that the higher expression levels of SIRT3 are associated with decreased levels of oxidative stress in the body and may play important roles in the prevention of age-related diseases. SIRTs can enhance the osteogenic potential and osteoblastic activity of bone marrow mesenchymal stromal cells not only by enhancing PGC-1α, FOXO3, SOD2, and oxidative phosphorylation, but also by anti-aging and reducing mitochondrial autophagy. SIRT3 is able to upregulate antioxidant enzymes to exert an inhibitory effect on osteoclasts, however, it has been shown that the inflammatory cascade response can in turn increase SIRT3 and inhibit osteoclast differentiation through the AMPK-PGC-1β pathway. SIRT3 plays an important role in different types of osteoporosis by affecting osteoblasts, osteoclasts, and bone marrow mesenchymal cells. In this review, we discuss the classification and physiological functions of SIRTs, the effects of SIRT3 on OCs osteoblasts, and BMSCs, and the roles and mechanisms of SIRT3 in different types of OP, such as diabetic OP, glucocorticoid-induced OP, postmenopausal OP, and senile OP.
... Histone methylation is a modification catalyzed by histone methyltransferases (HMTs), which utilize S-adenosylmethionine (SAM) as a substrate to transfer mono-, di-, or trimethyl groups (me1, me2, and me3, respectively) to the basic residues of histones. Histone methylation patterns such as H3K4me3, H3K79me3, and H3K36me3 are typically associated with transcriptional activation, while H3K27me3 and H3K9me3 have been linked to transcriptional silencing [20]. (Figure 1). ...
Article
Full-text available
Retinal homeostasis, a tightly regulated process maintaining the functional integrity of the retina, is vital for visual function. Emerging research has unveiled the critical role of epigenetic regulation in controlling gene expression patterns during retinal development, maintenance, and response to mutational loads and injuries. Epigenetic switches, including DNA methylation, histone modifications, and non-coding RNAs, play pivotal roles in orchestrating retinal gene expression and cellular responses through various intracellular, extracellular, and environmental modulators. This review compiles the current knowledge on epigenetic switches in retinal homeostasis, providing a deeper understanding of their impact on retinal structural integrity and function and using them as potential targets for therapeutic interventions.
Article
Full-text available
Vitamin B12 (cobalamin) is an essential nutrient for humans and animals. Metabolically active forms of B12-methylcobalamin and 5-deoxyadenosylcobalamin are cofactors for the enzymes methionine synthase and mitochondrial methylmalonyl-CoA mutase. Malfunction of these enzymes due to a scarcity of vitamin B12 leads to disturbance of one-carbon metabolism and impaired mitochondrial function. A significant fraction of the population (up to 20%) is deficient in vitamin B12, with a higher rate of deficiency among elderly people. B12 deficiency is associated with numerous hallmarks of aging at the cellular and organismal levels. Cellular senescence is characterized by high levels of DNA damage by metabolic abnormalities, increased mitochondrial dysfunction, and disturbance of epigenetic regulation. B12 deficiency could be responsible for or play a crucial part in these disorders. In this review, we focus on a comprehensive analysis of molecular mechanisms through which vitamin B12 influences aging. We review new data about how deficiency in vitamin B12 may accelerate cellular aging. Despite indications that vitamin B12 has an important role in health and healthy aging, knowledge of the influence of vitamin B12 on aging is still limited and requires further research.
Article
Full-text available
Histone H2B monoubiquitination (at Lys120 in humans) regulates transcription elongation and DNA repair. In humans, H2B monoubiquitination is catalyzed by the heterodimeric Bre1 complex composed of Bre1A/RNF20 and Bre1B/RNF40. The Bre1 proteins generally function as tumor suppressors, while in certain cancers, they facilitate cancer cell proliferation. To obtain structural insights of H2BK120 ubiquitination and its regulation, we report the cryo-electron microscopy structure of the human Bre1 complex bound to the nucleosome. The two RING domains of Bre1A and Bre1B recognize the acidic patch and the nucleosomal DNA phosphates around SHL 6.0–6.5, which are ideally located to recruit the E2 enzyme and ubiquitin for H2BK120-specific ubiquitination. Mutational experiments suggest that the two RING domains bind in two orientations and that ubiquitination occurs when Bre1A binds to the acidic patch. Our results provide insights into the H2BK120-specific ubiquitination by the Bre1 proteins and suggest that H2B monoubiquitination can be regulated by nuclesomal DNA flexibility.
Article
Histone 2B ubiquitination (H2Bub) and trimethylation of H3 at lysine 4 (H3K4me3) are associated with transcription activation. However, the function of these modifications in transcription in plants remains largely unknown. Here, we report that coordination of H2Bub and H3K4me3 deposition with the binding of the RNA polymerase‐associated factor VERNALIZATION INDEPENDENCE2 (VIP2) to FLOWERING LOCUS C ( FLC ) modulates flowering time in Arabidopsis . We found that RING domain protein HISTONE MONOUBIQUITINATION1 (HUB1) and HUB2 (we refer as HUB1/2), which are responsible for H2Bub, interact with ARABIDOPSIS TRITHORAX1 (ATX1), which is required for H3K4me3 deposition, to promote the transcription of FLC and repress the flowering time. The atx1‐2 hub1‐10 hub2‐2 triple mutant in FRIGIDIA ( FRI ) background displayed early flowering like FRI hub1‐10 hub2‐2 and overexpression of ATX1 failed to rescue the early flowering phenotype of hub1‐10 hub2‐2 . Mutations in HUB1 and HUB2 reduced the ATX1 enrichment at FLC , indicating that HUB1 and HUB2 are required for ATX1 recruitment and H3K4me3 deposition at FLC . We also found that the VIP2 directly binds to HUB1, HUB2, and ATX1 and that loss of VIP2 in FRI hub1‐10 hub2‐2 and FRI atx1‐2 plants resulted in early flowering like that observed in FRI vip2‐10 . Loss of function of HUB2 and ATX1 impaired VIP2 enrichment at FLC , and reduced the transcription initiation and elongation of FLC . In addition, mutations in VIP2 reduced HUB1 and ATX1 enrichment and H2Bub and H3K4me3 levels at FLC . Together, our findings revealed that HUB1/2, ATX1, and VIP2 coordinately modulate H2Bub and H3K4me3 deposition, FLC transcription, and flowering time.
Preprint
Full-text available
Cytosine methylation of genomic DNA contributes to the regulation of gene expression and is involved in normal development including hematopoiesis in mammals. It is catalyzed by the family of DNA methyltransferases (DNMTs) that include DNMT1, DNMT3A, and DNMT3B. Peripheral T-cell lymphomas (PTCLs) represent a diverse group of aggressive mature T-cell malignancies accounting for approximately 10–15% of non-Hodgkin lymphoma cases in the US. PTCLs exhibit a broad spectrum of clinical, histological, and immunophenotypic features with poor prognosis and inadequately understood molecular pathobiology. To better understand the molecular landscape and identify candidate genes involved in disease maintenance, we used high-resolution Whole Genome Bisulfite Sequencing (WGBS) and RNA-seq to profile DNA methylation and gene expression of PTCLs and normal T-cells. We found that the methylation patterns in PTCLs are deregulated and heterogeneous but share 767 hypo- and 567 hypermethylated differentially methylated regions (DMRs) along with 231 genes up- and 91 genes downregulated in all samples suggesting a potential association with tumor development. We further identified 39 hypomethylated promoters associated with increased gene expression in the majority of PTCLs. This putative oncogenic signature included the TRIP13 (thyroid hormone receptor interactor 13) gene whose both genetic and pharmacologic inactivation, inhibited cellular growth of PTCL cell lines by inducing G2-M arrest accompanied by apoptosis suggesting that such an approach might be beneficial in human lymphoma treatment. Altogether we show that human PTCLs are characterized by a large number of recurrent methylation alterations, and demonstrated that TRIP13 is critical for PTCL maintenance in vitro.
Article
Full-text available
CpG islands (CGIs) are associated with most mammalian gene promoters. A subset of CGIs act as polycomb response elements (PREs) and are recognized by the polycomb silencing systems to regulate expression of genes involved in early development. How CGIs function mechanistically as nucleation sites for polycomb repressive complexes remains unknown. Here we discover that KDM2B (FBXL10) specifically recognizes non-methylated DNA in CGIs and recruits the polycomb repressive complex 1 (PRC1). This contributes to histone H2A lysine 119 ubiquitylation (H2AK119ub1) and gene repression. Unexpectedly, we also find that CGIs are occupied by low levels of PRC1 throughout the genome, suggesting that the KDM2B-PRC1 complex may sample CGI-associated genes for susceptibility to polycomb-mediated silencing. These observations demonstrate an unexpected and direct link between recognition of CGIs by KDM2B and targeting of the polycomb repressive system. This provides the basis for a new model describing the functionality of CGIs as mammalian PREs.DOI:http://dx.doi.org/10.7554/eLife.00205.001.
Article
Full-text available
Significance In metazoans, pausing of Pol II during early elongation is a widespread regulatory mechanism for transcription regulation. However, preinitiation complex (PIC) assembly is more important for transcription in plants. Chromatin remodeling and histone modifications are considered important for access of protein factors to the underlying DNA sequences. However, how histone modifications are specifically and timely generated at active promoters is less understood. COMPASS-like complex plays a critical role in PIC assembly and histone H3K4 trimethylation. We found that Arabidopsis transcription factors bZIP28/bZIP60 interact with COMPASS-like components both in vitro and in vivo. We present a general model on how histone H3K4 trimethylation is specifically formed during inducible gene expression by using the endoplasmic reticulum (ER) stress response system in Arabidopsis plants.
Article
Post-translational modifications of proteins have emerged as a major mechanism for regulating gene expression. However, our understanding of how histone modifications directly affect chromatin function remains limited. In this study, we investigate acetylation of histone H3 at lysine 64 (H3K64ac), a previously uncharacterized acetylation on the lateral surface of the histone octamer. We show that H3K64ac regulates nucleosome stability and facilitates nucleosome eviction and hence gene expression in vivo. In line with this, we demonstrate that H3K64ac is enriched in vivo at the transcriptional start sites of active genes and it defines transcriptionally active chromatin. Moreover, we find that the p300 co-activator acetylates H3K64, and consistent with a transcriptional activation function, H3K64ac opposes its repressive counterpart H3K64me3. Our findings reveal an important role for a histone modification within the nucleosome core as a regulator of chromatin function and they demonstrate that lateral surface modifications can define functionally opposing chromatin states. DOI: http://dx.doi.org/10.7554/eLife.01632.001.
Article
Sotos syndrome is an overgrowth syndrome characterised by pre- and postnatal overgrowth, macrocephaly, advanced bone age, and typical facial features. Weaver syndrome is a closely related condition characterised by a distinctive craniofacial appearance, advanced carpal maturation, widened distal long bones, and camptodactyly. Haploinsufficiency of the NSD1 gene has recently been reported as the major cause of Sotos syndrome while point mutations accounted for a minority of cases. We looked for NSD1 deletions or mutations in 39 patients with childhood overgrowth. The series included typical Sotos patients (23/39), Sotos-like patients (lacking one major criteria, 10/39), and Weaver patients (6/39). We identified NSD1 deletions (6/33) and intragenic mutations (16/33) in Sotos syndrome patients. We also identified NSD1 intragenic mutations in 3/6 Weaver patients. We conclude therefore that NSD1 mutations account for most cases of Sotos syndrome and a significant number of Weaver syndrome cases in our series. Interestingly, mental retardation was consistently more severe in patients with NSD1 deletions. Macrocephaly and facial gestalt but not overgrowth and advanced bone age were consistently observed in Sotos syndrome patients. We suggest therefore considering macrocephaly and facial gestalt as mandatory criteria for the diagnosis of Sotos syndrome and overgrowth and advanced bone age as minor criteria.
Article
For cells to exit from pluripotency and commit to a lineage, the circuitry of a core transcription factor (CTF) network must be extinguished in an orderly manner through epigenetic modifications. Yet, how this choreographed epigenetic remodeling at active ESC genes occurs during differentiation is poorly understood. In this study, we demonstrate that C-terminal binding protein 2 (Ctbp2) regulates nucleosome remodeling and deacetylation (NuRD)-mediated deacetylation of H3K27 and facilitates recruitment of polycomb repressive complex 2 (PRC2)-mediated H3K27me3 in active ESC genes for exit from pluripotency during differentiation. By genomewide analysis, we found that Ctbp2 resides in active ESC genes and co-occupies regions with ESC CTFs in undifferentiated ESCs. Further, ablation of Ctbp2 effects inappropriate gene silencing in ESCs by sustaining high levels of H3K27ac and impeding H3K27me3 in active ESC genes, thereby sustaining ESC maintenance during differentiation. Thus, Ctbp2 preoccupies regions in active genes with the NuRD complex in undifferentiated ESCs that are directed toward H3K27me3 by PRC2 to induce stable silencing, which is pivotal for natural lineage commitment. This article is protected by copyright. All rights reserved. © 2015 AlphaMed Press.
Article
Polycomb repressive complexes PRC1 and PRC2 regulate expression of genes involved in proliferation and development. In mouse early embryos, however, canonical PRC1 localizes to paternal pericentric heterochromatin (pat-PCH), where it represses transcription of major satellite repeats. In contrast, maternal PCH (mat-PCH) is enriched for H3 lysine 9 tri-methylation (H3K9me3) and Hp1β. How PRC1 is targeted to pat-PCH, yet excluded from mat-PCH, has remained elusive. Here, we identify a PRC1 targeting mechanism that relies on Cbx2 and Hp1β. Cbx2 directs catalytically active PRC1 to PCH via its chromodomain (CD(Cbx2)) and neighboring AT-hook (AT(Cbx2)) binding to H3K27me3 and AT-rich major satellites, respectively. CD(Cbx2) prevents AT(Cbx2) from interacting with DNA at PCH marked by H3K9me3 and Hp1β. Loss-of-function studies show that Hp1β and not H3K9me3 prevents PRC1 targeting to mat-PCH. Our findings indicate that CD(Cbx2) and AT(Cbx2) separated by a short linker function together to integrate H3K9me3/HP1 and H3K27me3 states. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Polycomb and Trithorax group proteins encode the epigenetic memory of cellular positional identity by establishing inheritable domains of repressive and active chromatin within the Hox clusters. Here we demonstrate that the CCCTC-binding factor (CTCF) functions to insulate these adjacent yet antagonistic chromatin domains during embryonic stem cell differentiation into cervical motor neurons. Deletion of CTCF binding sites within the Hox clusters results in the expansion of active chromatin into the repressive domain. CTCF functions as an insulator by organizing Hox clusters into spatially disjoint domains. Ablation of CTCF binding disrupts topological boundaries such that caudal Hox genes leave the repressed domain and become subject to transcriptional activation. Hence, CTCF is required to insulate facultative heterochromatin from impinging euchromatin to produce discrete positional identities. Copyright © 2015, American Association for the Advancement of Science.