SlideShare a Scribd company logo
1 of 10
Download to read offline
Erase–Maintain–Establish: Natural Reprogramming
of the Mammalian Epigenome
MILENA LESEVA,1
BARBARA B. KNOWLES,2
DANIEL M. MESSERSCHMIDT,1
AND DAVOR SOLTER
3
1
Department for Developmental Epigenetics and Disease, Institute of Molecular and Cell Biology,
AÃ
STAR, 138673 Singapore
2
Emerita, The Jackson Laboratory, Bar Harbor, Maine 04609
3
Emeritus Member and Director, Max-Planck Institute of Immunobiology and Epigenetics,
79180 Freiburg, Germany
Correspondence: Danielm@imcb.a-star.edu.sg; davorsolter@mac.com
The genetic information is largely identical across most cell types in a given organism but the epigenome, which controls
expression of the genome,is cell type– and context-dependent. Although most mature mammalian cells appear to have a stable,
heritable epigenome, a dynamic intricate process reshapes it as these cells transition from soma to germline and back again.
During normal embryogenesis, primordial germ cells, of somatic origin, are set aside to become gametes. In doing so their
genome is reprogrammed—that is, the epigenome of specific regions is replaced in a sex-specific fashion as they terminally
differentiate into oocytes or spermatocytes in the gonads. Upon union of these gametes, reprogramming of the new organism’s
epigenome is initiated, which eventually leads, through pluripotent cells, to the cell lineages required for proper embryonic
development to a sexually mature adult. This never-ending cycle of birth and rebirth is accomplished through methylation and
demethylation of specific genomic sites within the gametes and pluripotent cells of an organism. This enigmatic process of
natural epigenomic reprogramming is now being dissected in vivo, focusing on specific genomic regions—that is, imprinted
genes and retrotransposons, where TRIM28 molecular complexes appear to guide the transition from gamete to embryo.
Mammalian reproduction requires union of two highly
specialized cells, the egg and sperm, at fertilization, an
event that marks the beginning of new life. Epigenetic
marks of this newly formed genome are immediately
required to establish the cell type–specific gene expres-
sion programs required for embryonic growth and devel-
opment. DNA methylation is an epigenetic modification
that leads to repression of specific gene expression, X-
chromosome inactivation, and silencing of mobile DNA
elements (for review, see Messerschmidt et al. 2014) and
affects alternative splicing (Yearim et al. 2015).
Establishment and maintenance of stable methylation
patterns are thus crucial to execution of a normal devel-
opmental program (Okano et al. 1999). The DNA meth-
ylome should be, and is, extremely plastic during very
early embryogenesis. Its dynamic control is initiated at
fertilization during the switch from germ cell to embryo-
specific methylation and a reciprocal change manifests
during germ cell specification with the switch from a
somatic to a germline epigenomic signature (Fig. 1).
Fertilization triggers extensive reprogramming where-
by the global DNA hypermethylation of the germ cell–
specific epigenomes become, on the whole, demethylat-
ed. The paternal genome in the pronucleus of the zygote
is rapidly demethylated and reorganized. The methyla-
tion of the maternal genome is gradually depleted in a
replication-dependent manner over consecutive mitotic
divisions (Santos and Dean 2004). Although the precise
mechanics of the active process are not completely un-
derstood, TET3-mediated oxidation of 5-methylcytosine
(5mC) (Gu et al. 2011; Iqbal et al. 2011; Wossidlo et al.
2011) and restoration of unmethylated cytosines by com-
ponents of the base excision repair pathway (Hajko-
va et al. 2010) are involved. These changes to the em-
bryonic genome lead to an open chromatin state, essential
for the pluripotency of some cells in the mouse blasto-
cyst’s inner cell mass (ICM). Reacquisition of de novo
promoter methylation occurs between the time of estab-
lishment of the ICM of the blastocyst and the develop-
ment of the postimplantation epiblast when the cells of
the embryo differentiate into the somatic lineages (Borgel
et al. 2010).
Resetting the epigenome is also mandatory during pri-
mordial germ cell (PGC) development when the methyl-
ation marks of the parental epigenome are erased and new
gender-specific imprinting patterns are established (Sei-
senberger et al. 2012). PGCs are specified at E6.5 from a
small group of BLIMP1-positive cells located in the
somatic proximal postimplantation epiblast (for review,
see Magnu´sdo´ttir and Surani 2014). In mouse PGCs 5mC
levels gradually decline in a partially TET1/2-dependent
manner, as the cells migrate through the hindgut endo-
derm (E9.5), reach the gonadal anlagen (E11.5), and un-
dergo sexual determination (E13.5) (Fig. 1).
Given the size and complexity of the mammalian ge-
nome, it is fascinating that demethylation and remethyl-
ation events are intricately controlled both temporally
and in a genome context-specific manner. How are cer-
Copyright # 2015 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/sqb.2015.80.027441
Cold Spring Harbor Symposia on Quantitative Biology, Volume LXXX 155
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
tain loci identified to escape the loss of DNA methylation
in an environment that favors genome-wide demethyla-
tion events? Recent studies of the molecular interactions
at specific sites in imprinted genes and in endogenous
retroelements (EREs) have afforded a glimpse of this
complex process. Because methylation protection of spe-
cific DNA regions initiates immediately after fertiliza-
tion, and during a time of complete transcriptional
silence, the molecular agents, either RNA and/or protein,
must be in the oocyte—that is, maternally inherited. The
role of one such maternal factor, TRIM28, will be used to
illustrate the complexity of this process in the mouse.
TRIM28—A MULTIFACETED WORKER
IN THE CHROMATIN FIELD
Tripartite motif containing 28/KRAB-domain-asso-
ciated protein1/transcription intermediary factor-1b
(TRIM28/KAP1/TIF1b) is a multifunctional protein
implicated in processes as diverse as embryonic de-
velopment, maintenance of pluripotency, imprinting,
endogenous and exogenous retroviral silencing, and dou-
ble-strand break DNA damage response, to name a few.
The structure of TRIM28, with its amino-terminal RING,
B-box, and coiled-coil (RBCC) domain, central HP1-
binding domain, and carboxy-terminal PHD-bromodo-
mains, provides it with the means to interact with multiple
transcription factors (TFs) and a variety of chromatin re-
modelers and epigenetic modifiers (Iyengar and Farnham
2011; Cheng 2014).
TRIM28 itself does not bind to DNA directly but rather
relies on DNA-binding TF-mediated recruitment. These
are largely members of the Kru¨ppel-associated box
(KRAB) domain zinc-finger proteins (ZFPs), a family
of TFs containing more than 200 genes in mice and hu-
mans that encode distinct proteins with highly variable
DNA-binding domains. KRAB-ZFPs may have evolved
from Prdm9 (Meisetz) (Birtle and Ponting 2006) and ex-
panded rapidly by duplication events during evolution.
Loss of the original SET domain, accumulation of se-
quence changes to the ancestral KRAB domain, and ap-
pearance of corepressors are responsible for establishing
the KRAB-ZFPs as transcriptional repressors. One expla-
nation for the huge expansion and diversification of these
TFs lies in their presumed coevolution with EREs to
prevent their transcriptional reactivation (Thomas and
Schneider 2011).
KRAB-ZFPs contain tandem arrays of C2H2 zinc fin-
gers, each engaging three consecutive nucleotides on one
DNA strand and one nucleotide on the opposite strand.
The combination of multiple ZNFs within one protein
allows KRAB-ZFPs to bind with high specificity and to
recognize long stretches of DNA. The amino-terminally
encoded KRAB domain is highly conserved throughout
the KRAB-ZFP family and mediates interaction with
TRIM28.
Numerous other TFs have been reported to interact with
and potentiallyattractTRIM28 to their DNA-binding sites
(Fig. 2; Satou 2001; Li et al. 2003; Satou et al. 2004; Wang
et al. 2005, 2007; Jin et al. 2007; Tsuruma et al. 2007;
Kamitani et al. 2008, 2011; Peng et al. 2009; Seki et al.
2010; Liang et al. 2011; Maruyama et al. 2011; Huang
et al. 2013). For most of these, the KRAB-ZFPs included,
TRIM28functionsasaco-repressordown-regulatinggene
transcription by its heterochromatin-inducing activity at
its target sites (Iyengar and Farnham 2011; Cheng 2014).
1
2
3
5
4
6
7
8
Figure 1. Epigenetic reprogramming during the mammalian reproductive cycle. (1) Both male and female germ cells are differentiated
cell types and have highly specialized epigenomes. Fertilization of the oocyte by sperm initiates embryonic development. (2) The
paternal genome (blue) is rapidly demethylated at early pronuclear stages. (3) The maternal genome (pink) is passively demethylated
in a replication-dependent manner. (4) Erasure of germ cell–specific DNA methylation patterns allows the establishment of the
pluripotent inner cell mass of the blastocyst. (5) Pluripotent cells begin the acquisition of DNA methylation marks as they begin to
differentiate into somatic lineages. (6) PGCs are specified in the proximal postimplantation epiblast. They carry somatic epigenetic
features, such as genomic imprints, inactive X-chromosomes, and silenced germ cell/pluripotency genes. (7) These features and
potential epimutations are erased during primordial germ cell reprogramming. (8) Male- and female-specific germ cell differentiation
includes establishment of paternal and maternal imprints.
LESEVA ET AL.156
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
TRIM28 acts as a scaffolding protein associating with
the H3K9me3-writer SETDB1/ESET, the H3K9me3-
reader HP1, and the nucleosome remodeling and histone
deacetylation complex NuRD (Schultz et al. 2002, 2001).
This multisubunit, heterochromatin-inducing molecular
complex also serves as a platform for recruiting DNA
methyltransferases, which can then induce and maintain
DNA methylation in proximity to their target sites (Fig. 2;
Quenneville et al. 2011; Zuo et al. 2012).
From a developmental biologist’s point of view, it was
known for some time that Trim28 was highly expressed in
the preimplantation embryo and that its zygotic knockout
is embryonic lethal in early postimplantation stages
(Cammas et al. 2000). Trim28 was also identified in in-
dependent screens for factors required for pluripotency
and self-renewal of embryonic stem cells (ESCs) (Fazzio
et al. 2008; Hu et al. 2009). However, the precise role of
Trim28 in mammalian cells remained unclear, although it
was found to relocalize to centromeric heterochromatin
upon cell differentiation (Cammas et al. 2002).
Additional insights to TRIM28 function in vivo
were gained from studies in preimplantation embryos
Trim28
STAT3
STAT1
SRY/KRAB-O
HP1α
HP1γ
HP1β
Histonedeacetylation
N-CoR
NuRD
HDAC1
SMARCAD1
DNMT1
DNMT3b
DNMT3a
DNA methylation
SETDB1
Histone methylation
Pou5f1
Myc
E2F1
p53
IRF7
HIF1α
NRF2
FOXP3
NFκB
KRAB-ZFPs
TranscriptionFactors
B Interaction Partners of TRIM28
C KRAB-ZFPs & Function
Genomic imprinting maintenance
exo- and endogenous ERE silencing
ZFP57
ZFP809
ZFP819
Gm6871
MuLV
IAP,LINE
LINE
Mouse
Mouse/
Human gDMRs
Human
ZNF350
ZNF175
ZNF10
HIV
ZNF93 L1
ZNF91 SVA
ZNF282 HTLV
Chromatin readers
Chromatin writers
Chromatinmodifiers
A The TRIM28-complex
G9a
DNMTs
G9a
K9K9K9 K9
NuRD
SETDB1
H3 H3 H3 H3
HP1
NH2
N
NH2
N
R
NH2
OOOOOO
N
NH2NH22
NNNNNN
R
222
CH3
TF
TRIM28
1 2
3
others
ZNF160Human
TLR4
ZBRKMouse p21
ZNF689 miRNAs
O
R
NNNNOO
Figure 2. The TRIM28 complex and interactome. (A) TRIM28 acts as a central scaffolding protein for a multifunctional, large protein
complex. It is guided to DNA by interacting transcription factors and attracts in turn: (1) The NuRD histone deacetylation complex
(deacetylates H3K9Ac); (2) SETDB1 and G9a histone methyltransfereases (trimethylate H3K9me3); (3) DNA methyltransferases
(DNMTs) (methylate cytosines). Heterochromatin protein 1 (HP1) recognizes H3K9me3 chromatin and further physically interacts
with TRIM28. (B) TRIM28 interaction partners: KRAB-ZFPs and other TFs can target TRIM28 to DNA (left) (Satou 2001; Li et al.
2003; Satou et al. 2004; Wang et al. 2005, 2007; Jin et al. 2007; Tsuruma et al. 2007; Kamitani et al. 2008, 2011; Peng et al. 2009; Seki
et al. 2010; Liang et al. 2011; Maruyama et al. 2011; Huang et al. 2013). TRIM28 attracts chromatin writers and readers (right)
(Nielsen et al. 1999; Ryan et al. 1999; Schultz et al. 2001, 2002; Quenneville et al. 2011; Zuo et al. 2012). (C) Few KRAB-ZFP
proteins have been studied in detail. ZFP57 plays a conserved role in imprinting maintenance in humans and mice. Diverse ZFPs/ZFNs
(mouse/human) are involved in retrovirus silencing and other physiological functions (Okumura et al. 1997; Zheng et al. 2000; Li et al.
2008; Takahashi et al. 2009; Wolf and Goff 2009; Quenneville et al. 2011; Nishitsuji et al. 2012, 2015; Tan et al. 2013; Castro-Diaz
et al. 2014; Jacobs et al. 2014; Wolf et al. 2015b).
EPIGENETIC REPROGRAMMING 157
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
(Messerschmidt 2012; Messerschmidt et al. 2012). Dele-
tion of maternal Trim28 alone causes embryonic lethality,
despite its reexpression from the paternal allele follow-
ing embryonic genome activation at the two-cell stage.
Intriguingly, the maternal-null embryos show highly
variable phenotypes and die at various times throughout
gestation, albeit at a much later time than when the
molecular deficiency of TRIM28 first occurs. The pleio-
tropic defects displayed by these embryos are indicative
of accumulated, random combinations of epimutations
incompatible with life (Messerschmidt et al. 2012; Lor-
thongpanich et al. 2013).
We now know that TRIM28 is essential for proper and
successful epigenetic reprogramming through at least
two fundamental functions—that is, maintaining imprint-
ing and silencing retrotransposons.
THE TRIM28-COMPLEX FUNCTION
IN GENOMIC IMPRINTING
Genomic imprinting, or the establishment and mainte-
nance of monoallelic expression of particular genes in a
parent-of-origin-specific manner, is at the heart of the
developmental failure observed in genetically bimaternal
(gynogenote) and bipaternal (androgenote) embryos. The
observation that the incomplete development of embryos
derived from gynogenotes is largelyembryo-like, whereas
that of androgenotes is mostly of extraembryonic-like tis-
sues, showed that the maternal and paternal genomes are
functionally nonequivalent (McGrath and Solter 1984;
Surani et al. 1984). Imprinting implies that the maternal
and paternal genomes are differentially marked at specif-
ic genetic loci but does not involve changes in the DNA
sequence. These epigenetic marks regulate gene expres-
sion, are mitotically heritable, and are set during female
and male gametogenesis, respectively. Imprinted genes
are often clustered in specific sites of the genome and
within these clusters they can be coordinately regulated
by differentially methylated regions at the so-called im-
printing control regions (ICRs). Methylation at ICRs ul-
timately controls gene expression in cis by multiple
mechanisms. Many imprinting clusters express noncod-
ing RNAs (ncRNAs) that can further regulate genes
within the cluster in trans. Collectively, epigenetic mod-
ification ensures that maternally imprinted genes are
expressed from the maternally transmitted allele and si-
lenced on the paternal allele, and vice versa (Hanna and
Kelsey 2014; Peters 2014).
Parent-specific DNA methylation patterns at the ICRs
are instituted during gametogenesis, part of a process
requiring erasure of the inherited parental marks, so that
imprints can be established sex-specifically. These paren-
tal imprints are protected and preserved postfertilization
and stabilized throughout embryonic and adult life, as
proper levels of the product of imprinted genes are essen-
tial to embryonic development, or are involved in adult
cell and tissue homeostasis. For this reason, specialized
mechanisms must be put in place to protect such sites
from erasure during the wave of demethylation in the
early embryo. Genomic imprints are therefore paradig-
matic of the subtle epigenetic fine-tuning required during
genomic reprogramming. Although the genomic imprint-
ing cycle of “erasure–establishment–maintenance and
erasure again” has been postulated and shown for some
time, the molecular mechanisms behind imprint mainte-
nance involving the TRIM28 complex were only recently
unraveled.
Demethylation in the preimplantation embryos is
achieved by a combination of active and passive erasure
of the methylation from 5mC (for review, see Messer-
schmidt et al. 2014). To allow for efficient methylation
removal, the maintenance DNA methyltransferase-1
(DNMT1) is largely excluded from the nuclei of the
cleavage-stage embryos (Howell et al. 2001; Ratnam
et al. 2002; Branco et al. 2008; Hirasawa et al. 2008).
However, this compartmentalization and nuclear exclu-
sion cannot be complete, as genetic deletion of both ma-
ternal and zygotic Dnmt1 results in imprinting defects,
the result of complete ICR demethylation (Hirasawa et al.
2008). DNMT1 is canonically targeted to the replication
fork in mitotic cells by its cofactor NP95. Because the
drastically reduced DNMT1 levels and the global DNA
methylation maintenance function of this complex make
it unlikely to maintain imprints in embryos, a noncanon-
ical targeting mechanism of DNMT1 had been proposed.
Insights into how DNMT1 might be targeted specifi-
cally to ICRs came from studies in vivo and in vitro.
We were able to show that loss of maternal Trim28
alone can result in dramatic misexpression of imprinted
genes in midgestation embryos (Messerschmidt 2012;
Messerschmidt et al. 2012, 2014; Lorthongpanich et al.
2013). This observed deregulation was observed across
multiple imprinting clusters with the deregulation fol-
lowing a pattern predicated on a loss-of-ICR-methylation
scenario. Indeed, partial, severe ICR demethylation, pre-
dominantly at the paternally imprinted H19/IGF2 locus,
was found in these embryos. All tested imprinted loci,
however, displayed variable degrees of methylation loss,
arguing for (1) a global mechanism and (2) a partial res-
cue of the defect by paternal Trim28 expression (Messer-
schmidt et al. 2012; Lorthongpanich et al. 2013). Indeed,
single-cell analysis of DNA methylation in eight-cell-
stage embryos revealed that not all blastomeres are
affected equally; hence, the pleiotropic defects observed
are likely a consequence of epigenetic mosaicism in the
mutant embryos. Interestingly, maternal/zygotic-null
Trim28 embryos develop to apparently normal blasto-
cysts and implant (DM Messerschmidt, unpubl.); thus,
the complete knockout does not show a phenotype during
preimplantation embryogenesis. However, it remains to
be seen whether loss of both maternal and paternal
Trim28 will result in complete eradication of ICR meth-
ylation in preimplantation embryos.
TRIM28 is physically bound to known ICRs in mid-
gestation embryos, arguing for a direct recognition and
maintenance mechanism (Messerschmidt et al. 2012).
ICRs in these embryos are co-occupied by ZFP57 and
as a consequence of the binding of the TRIM28 complex,
histones were H3K9me3 modified (Messerschmidt et al.
LESEVA ET AL.158
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
2012). ZFP57 is a TRIM28-interacting KRAB-ZFP and
both maternal and zygotic Zfp57 are necessary for the
proper maintenance of parental imprints in blastocysts
(Li et al. 2008). Investigations in ESCs showed ZFP57
recognizes a CpG-containing consensus TGCCGC se-
quence, highly conserved at ICRs (Quenneville et al.
2011). However, TRIM28 binding to hypomethylated
sites was lost in embryos (Messerschmidt et al. 2012)
leading us to suggest that ZFP57 recognizes the methyl-
ated cytosine. Indeed, ZFP57 shows much higher affinity
to its methylated target sequence in vitro (Quenneville
et al. 2011; Liu et al. 2012). Thus, sequence-specific
and methylation-dependent binding to ICRs offers an
elegant solution to the convoluted undertaking of (1) rec-
ognizing specialized regions within the genome and
(2) maintaining differential DNA methylation at a given
locus. It also explains the lack of paternal rescue in
the maternal Trim28-null embryo because preexisting
DNA-methylated sites, inherited from the gametes, are
fundamental to the ZFP57-mediated TRIM28-complex
binding. If a particular CpG is demethylated under ma-
ternal-null circumstances, reexpressed paternal TRIM28
cannot be targeted to it and cannot restore DNA methyl-
ation. Similarly, reexpression of ZFP57 in Zfp57-deleted
ES cells cannot restore methylation to sites that have lost
it (Zuo et al. 2012). Finally, this mode of interaction
offers a plausible model for the noncanonical targeting
of the dramatically reduced levels of DNMT1 to ICRs
in preimplantation embryos; both maintenance DNA-
methyltransferase and the de novo DNA-methyl-
transferases (DNMT3A/B) are bona fide interaction part-
ners of TRIM28 (Quenneville et al. 2011; Zuo et al.
2012).
The highly specialized role and mode of chromatin
interaction of ZFP57 with specific genomic sequences,
be they methylated or not, is an exquisite example of the
functional diversity acquired by the KRAB-ZFPs. Fur-
ther, the multiple interactions made possible through
the TRIM28 scaffolding complex not only clarify the
specificity of epigenetic reprogramming but also accen-
tuate the importance of the balance between erasure of
most parental epigenetic features versus the careful main-
tenance of a few stable epigenetic sites.
THE TRIM28 COMPLEX IN
RETROTRANSPOSON REGULATION
Mammalian cells face a formidable challenge in deal-
ing with potentially damaging endogenous retroviruses
(ERVs) and retrotransposons, collectively termed endog-
enous retroviral elements (EREs). These elements make
up 40% of the mammalian genome, highlighting the
magnitude of this task (Lander et al. 2001; Mouse Ge-
nome Sequencing Consortium et al. 2002).
Aberrant activation of EREs can impact the host on two
major levels. For one, certain retrotransposons remain
capable of active retrotransposition that can lead to ran-
dom insertional mutagenesis following replicative trans-
position into the host’s genome. This must be prevented
at all costs, particularly in the germline. Most EREs,
however, have lost their ability to replicate but still their
remnants are important targets of the host’s silencing
machinery. Loss of silencing at such sites can result in
drastic, localized chromatin changes and impact on cer-
tain cellular genes whose expression may be under the
influence of neighboring, ERE-derived promoters/en-
hancers. In certain contexts, ERE control over gene
regulation and mobility is of beneficial effect to the
host. A case in point is the developmentally regulated
expression of nonconventional chimeric transcripts driv-
en by transposon-derived 50
sequences that can encode
new proteins with potentially novel functions (Peaston
et al. 2004). Temporary loss of a silencing mechanism
can also explain the stage-specific expression of different
long terminal repeat (LTR) retrotransposons during the
oocyte-to-embryo transition (Peaston et al. 2004). As it
now stands, most EREs are continuously repressed and
the first line of defense against their expression is DNA
methylation–based silencing, substantially contributing
to the overall hypermethylated state of the mammalian
genome.
One insight into the molecular mechanism control-
ling retrotransposon silencing during embryonic develop-
ment came from the discovery that Dnmt1-deficient
embryos reactivate all major classes of the intracisternal
A-type particles (IAPs) (Walsh et al. 1998). Later,
DNMT3L was shown to protect against spurious tran-
scriptional reactivation of long interspersed nuclear ele-
ments 1 (L1NE1) and ID1 IAP retrotransposons in male
germ cells (Bourc’his and Bestor 2004). Extensive re-
search has shown that during mammalian reproduction,
when DNA methylation patterns are globally modified
and removed, other mechanisms serve as a barrier to
ERE activation. The small RNAs (piRNAs) seem to pre-
dominant in the germline but in the embryo piRNAs play
a minor role to that of the KRAB-ZFP TFs, which medi-
ate formation of heterochromatin.
One transacting repressor complex specific to pluripo-
tent cells, and present in embryos, was shown to be im-
portant for ERV silencing. Initially termed binding factor
A, this 1.3-MDa multisubunit complex contains TRIM28
as a repressor subunit (Reik 2007) and the KRAB-
ZFP809 that serves as its sequence-specific DNA-bind-
ing partner (Wolf and Goff 2009). TRIM28 links the
DNA methylation and KRAB-ZFP repressive machiner-
ies as shown by tethering a doxycycline-controllable
KRAB domain to a lentiviral construct in vivo (Wizner-
owicz et al. 2007). Here, a green fluorescent protein
(GFP) reporter gene was found to be expressed only if
recruitment of the repressive KRAB-ZFP was prevented
by adding doxycycline in a narrow time window right
after fertilization (between E0.5 and E3.5). Conversely,
GFP silencing was permanently established if the KRAB-
containing fusion protein bound its target sequence very
early in embryogenesis.
ERVs are classified based on their sequence similari-
ties. Class I and particularly class II ERVs such as IAPs
and MusD are drastically deregulated in mouse ESCs
lacking TRIM28. Also, treatment of Trim28 knockout-
EPIGENETIC REPROGRAMMING 159
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
derived ESCs with 5-azacytidine (a DNMT inhibitor) has
a synergistic effect on ERV activation. However, con-
current reactivation is accompanied by depletion of the
H3K9me3 histone modification of the IAP and MusD
sequence. DNA methylation–independent silencing in
ESC has also been described, which requires TRIM28
and SETDB1 but does not rely on de novo or maintenance
methylation because Dnmt1/3a/3b triple-knockout ESCs
do not display the massive derepression of murine leuke-
mia virus (MLV), IAP, and MusD observed in Setdb1-
or Trim28-deficient cells (Matsui et al. 2010; Rowe et al.
2010).
It appears that certain redundant mechanisms func-
tion to ensure efficient ERV silencing; the preferred
mechanism depends on the class and evolutionary history
of the particular element (ancient vs. young). It has
been suggested that the repressive KRAB-ZFP machinery
likely coevolved together with the ERVs as a host mech-
anism to restrict their horizontal and vertical transmission
(Wolf et al. 2015a,b). Surprisingly, only a limited number
of KRAB-ZFPs have been shown to regulate specific
ERV classes in mice. Such examples include the regula-
tion of MLV by ZFP809 described earlier, of IAPs and
LINE1 by ZFP819, and of LINE1 by Gm6871 (Fig. 2). It
remains to be seen whether and which KRAB-ZFPs are
still actively involved exclusively in retrotransposon si-
lencing and whether many have evolved to undertake
“endogenous” host gene regulatory functions, such as
ZFP57.
ERV repression by the TRIM28 complex has been
scantily studied in vivo. Class II ERVs, particularly IAP
elements, are known to be massively activated in zygotic
Trim28 knockout blastocysts and periimplantation em-
bryos (Rowe et al. 2010). During oocyte growth, these
elements are generally repressed (Peaston et al. 2004)
but ZP3-cre-mediated removal of maternal Trim28 in
the growing oocyte does not result in IAP or MusD
derepression nor in the ectopic activation of any other
retroviral element tested here (Fig. 3). Thus, Trim28-
independent mechanisms, in collaboration with DNA
methylation and possibly piRNAs, may act to silence
ERVs in the mouse egg.
As early as the two-cell stage, when the embryonic
genome is activated, a veritable explosion of ERV tran-
scription takes place. Predominantly class III mouse tran-
script (MT) and ERV-like elements (ERVL) and class II
elements (IAPs) are transcribed (Kigami et al. 2003;
Peaston et al. 2004). This massive activation of ERVs
gives the two-cell-stage mouse embryo a characteristic
transcriptome that is recapitulated in a small subfraction
(1%) of ESCs (Kigami et al. 2003; Macfarlan et al. 2012).
It is unclear whether this wave of transcriptional activa-
tion is required for proper development or is only a by-
product of epigenetic reprogramming in the embryo.
Mouse (Mu) ERVL transcription is regulated by chro-
matin modifications, particularly H3K4me3. Removal of
LSD1/KDM1, an H3K4me3 demethylase, results in
MuERVL activation and embryonic arrest at gastrulation
(Macfarlan et al. 2012). We find loss of maternal Trim28
does not impact MuERVL expression or that of any other
class III or class I ERV tested. Also LINE and short
interspersed nuclear element (SINE) expression re-
mained unchanged (Fig. 3). Remarkably, despite the mas-
sive endogenous activation of class II element observed at
the two-cell stage, loss of maternal Trim28 causes a
hyperactivation of IAPs and MusD (Fig. 3). The dynam-
ics of this regulation and to what extent this overexpres-
sion, with all its repercussions, contributes to the
maternal-deletion phenotype of Trim28 are unclear and
warrant further investigation.
MII OocyteA Two-Cell StageB
IAP
100 2000
Trim28
100 2000
Oct4
100 2000
Line1
100 2000
Sine (ALU)
100 2000
MusD
100 2000
RLTR1b
100 2000
LTR class II
LTR class I
ORRA1A
2000 100 400300
MuERVL
100 2000
MT
100 2000
LTR class IIInon-LTR elements
Trim28
100 2000
Oct4
100 2000
Line1
100 2000
Sine (ALU)
100 2000
MusD
2000 100 400300
IAP
2000 400 600
RLTR1b
100 2000
MuERVL
100 2000
MT
100 2000
ORRA1A
100 2000
LTR class II
LTR class III
LTR class I
non-LTR elements
Trim28 maternal-null Trim28 wild type
Figure 3. ERV expression in maternal-null Trim28 and wild-type oocytes and embryos. Long terminal repeat (LTR) and non-LTR
retrotransposon expression was tested in (A) MII oocytes and (B) two-cell-stage embryos in the presence and absence of maternal
Trim28 (white and black bars, respectively). Although oocytes show no changes in ERV expression, two-cell-stage maternal-null
Trim28 embryos show elevated expression of the LTR class II elements, IAP and MusD.
LESEVA ET AL.160
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
CONCLUSION
Establishment of a unique epigenome is the basis for
correct functioning of each cell in a multicellular organ-
ism. Mechanistically we can envision the existence of two
types of epigenome. The dynamic or transitory epige-
nome is unique to each cell and tissue type, and it
is driven by classical TFs. It is malleable and repro-
grammable as showed by the phenomena of cloning,
transdifferentiation, and induced pluripotent stem (iPS)
cell derivation. In the stable epigenome, as represented
by imprinted genes and retroelements, silencing is based
predominantly on stable genomic modifications, which
are mitotically inherited and change only at the specific
stages of the life cycle of the organism. Establishment and
control of the stable epigenome has likelyevolved through
an evolutionary process we are just beginning to decipher.
Considering the importance of a stable epigenome to nor-
mal development, significant efforts should be undertak-
en to understand its establishment and maintenance.
ACKNOWLEDGMENTS
We thank the members of the Developmental Epige-
netics and Disease Laboratory and Heike Wollmann for
their comments and input to this work; Mimi DeVries,
The Jackson Laboratory, USA and Anne Peaston, Uni-
versity of Adelaide, Roseworthy, Australia for their early
contributions. M.L. and D.M.M. are supported by the
National Research Foundation (Singapore), AÃ
STAR’s
Bio-Medical Research Council, and AÃ
STAR’s Institute
of Molecular and Cell Biology, Singapore. B.B.K., D.S.,
and D.M.M. were previously supported by AÃ
STAR’s
Institute of Medical Biology, Singapore.
REFERENCES
Birtle Z, Ponting CP. 2006. Meisetz and the birth of the KRAB
motif. Bioinformatics 22: 2841–2845.
Borgel J, Guibert S, Li Y, Chiba H, Schu¨beler D, Sasaki H,
Forne´ T, Weber M. 2010. Targets and dynamics of promot-
er DNA methylation during early mouse development. Nat
Genet 42: 1093–1100.
Bourc’his D, Bestor TH. 2004. Meiotic catastrophe and retro-
transposon reactivation in male germ cells lacking Dnmt3L.
Nature 431: 96–99.
Branco MR, Oda M, Reik W. 2008. Safeguarding parental
identity: Dnmt1 maintains imprints during epigenetic repro-
gramming in early embryogenesis. Genes Dev 22: 1567–
1571.
Cammas F, Mark M, Dolle´ P, Dierich A, Chambon P, Losson R.
2000. Mice lacking the transcriptional corepressor TIF1b are
defective in early postimplantation development. Develop-
ment 127: 2955–2963.
Cammas F, Oulad-Abdelghani M, Vonesch J-L, Huss-Garcia Y,
Chambon P, Losson R. 2002. Cell differentiation induces
TIF1b association with centromeric heterochromatin via an
HP1 interaction. J Cell Sci 115: 3439–3448.
Castro-Diaz N, Ecco G, Coluccio A, Kapopoulou A, Yazdanpa-
nah B, Friedli M, Duc J, Jang SM, Turelli P, Trono D. 2014.
Evolutionally dynamic L1 regulation in embryonic stem cells.
Genes Dev 28: 1397–1409.
Cheng C-T. 2014. KAPtain in charge of multiple missions:
Emerging roles of KAP1. World J Biol Chem 5: 308–320.
Fazzio TG, Huff JT, Panning B. 2008. An RNAi screen of chro-
matin proteins identifies Tip60-p400 as a regulator of embry-
onic stem cell identity. Cell 134: 162–174.
Gu T-P, Guo F, Yang H, Wu H-P, Xu G-F, Liu W, Xie Z-G, Shi
L, He X, Jin S-G, et al. 2011. The role of Tet3 DNA dioxy-
genase in epigenetic reprogramming by oocytes. Nature 477:
606–610.
Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani
MA. 2010. Genome-wide reprogramming in the mouse
germ line entails the base excision repair pathway. Science
329: 78–82.
Hanna CW, Kelsey G. 2014. The specification of imprints in
mammals. Heredity 113: 176–183.
Hirasawa R, Chiba H, Kaneda M, Tajima S, Li E, Jaenisch R,
Sasaki H. 2008. Maternal and zygotic Dnmt1 are necessary
and sufficient for the maintenance of DNA methylation im-
prints during preimplantation development. Genes Dev 22:
1607–1616.
Howell CY, Bestor TH, Ding F, Latham KE, Mertineit C, Trasler
JM, Chaillet JR. 2001. Genomic imprinting disrupted by
a maternal effect mutation in the Dnmt1 gene. Cell 104:
829–838.
Hu G, Kim J, Xu Q, Leng Y, Orkin SH, Elledge SJ. 2009. A
genome-wide RNAi screen identifies a new transcriptional
module required for self-renewal. Genes Dev 23: 837–848.
Huang C, Martin S, Pfleger C, Du J, Buckner JH, Bluestone JA,
Riley JL, Ziegler SF. 2013. Cutting Edge: A novel, human-
specific interacting protein couples FOXP3 to a chromatin-
remodeling complex that contains KAP1/TRIM28. J Immu-
nol 190: 4470–4473.
Iqbal K, Jin S-G, Pfeifer GP, Szabo´ PE. 2011. Reprogramming
of the paternal genome upon fertilization involves genome-
wide oxidation of 5-methylcytosine. Proc Natl Acad Sci 108:
3642–3647.
Iyengar S, Farnham PJ. 2011. KAP1 protein: An enigmatic
master regulator of the genome. J Biol Chem 286: 26267–
26276.
Jacobs FMJ, Greenberg D, Nguyen N, Haeussler M, Ewing
AD, Katzman S, Paten B, Salama SR, Haussler D. 2014.
An evolutionary arms race between KRAB zinc-finger genes
ZNF91/93 and SVA/L1 retrotransposons. Nature 516: 242–
245.
Jin VX, O’Geen H, Iyengar S, Green R, Farnham PJ. 2007.
Identification of an OCT4 and SRY regulatory module using
integrated computational and experimental genomics ap-
proaches. Genome Res 17: 807–817.
Kamitani S, Ohbayashi N, Ikeda O, Togi S, Muromoto R, Sekine
Y, Ohta K, Ishiyama H, Matsuda T. 2008. KAP1 regulates
type I interferon/STAT1-mediated IRF-1 gene expression.
Biochem Biophys Res Commun 370: 366–370.
Kamitani S, Togi S, Ikeda O, Nakasuji M, Sakauchi A, Sekine Y,
Muromoto R, Oritani K, Matsuda T. 2011. Kru¨ppel-associat-
ed box-associated protein 1 negatively regulates TNF-a-
induced NF-kB transcriptional activity by influencing the
interactions among STAT3, p300, and NF-kB/p65. J Immu-
nol 187: 2476–2483.
Kigami D, Minami N, Takayama H, Imai H. 2003. MuERV-L
is one of the earliest transcribed genes in mouse one-cell
embryos. Biol Reprod 68: 651–654.
Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Bald-
win J, Devon K, Dewar K, Doyle M, FitzHugh W, et al. 2001.
Initial sequencing and analysis of the human genome. Nature
409: 860–921.
Li Z, Wang D, Na X, Schoen SR, Messing EM, Wu G. 2003. The
VHL protein recruits a novel KRAB-A domain protein
to repress HIF-1a transcriptional activity. EMBO J 22:
1857–1867.
Li X, Ito M, Zhou F, Youngson N, Zuo X, Leder P, Ferguson-
Smith AC. 2008. A maternal-zygotic effect gene, Zfp57,
maintains both maternal and paternal imprints. Dev Cell 15:
547–557.
Liang Q, Deng H, Li X, Wu X, Tang Q, Chang T-H, Peng H,
Rauscher FJ, Ozato K, Zhu F. 2011. Tripartite motif-contain-
EPIGENETIC REPROGRAMMING 161
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
ing protein 28 is a small ubiquitin-related modifier E3 ligase
and negative regulator of IFN regulatory factor 7. J Immunol
187: 4754–4763.
Liu Y, Toh H, Sasaki H, Zhang X, Cheng X. 2012. An atomic
model of Zfp57 recognition of CpG methylation within a
specific DNA sequence. Genes Dev 26: 2374–2379.
Lorthongpanich C, Cheow LF, Balu S, Quake SR, Knowles BB,
Burkholder WF, Solter D, Messerschmidt DM. 2013. Single-
cell DNA-methylation analysis reveals epigenetic chimerism
in preimplantation embryos. Science 341: 1110–1112.
Macfarlan TS, Gifford WD, Driscoll S, Lettieri K, Rowe HM,
Bonanomi D, Firth A, Singer O, Trono D, Pfaff SL. 2012.
Embryonic stem cell potency fluctuates with endogenous ret-
rovirus activity. Nature 487: 57–63.
Magnu´sdo´ttir E, Surani MA. 2014. How to make a primordial
germ cell. Development 141: 245–252.
Maruyama A, Nishikawa K, Kawatani Y, Mimura J, Hosoya T,
Harada N, Yamamato M, Itoh K. 2011. The novel Nrf2-inter-
acting factor KAP1 regulates susceptibility to oxidative stress
by promoting the Nrf2-mediated cytoprotective response.
Biochem J 436: 387–397.
Matsui T, Leung D, Miyashita H, Maksakova IA, Miyachi
H, Kimura H, Tachibana M, Lorincz MC, Shinkai Y. 2010.
Proviral silencing in embryonic stem cells requires the histone
methyltransferase ESET. Nature 464: 927–931.
McGrath J, Solter D. 1984. Completion of mouse embryogenesis
requires both the maternal and paternal genomes. Cell 37:
179–183.
Messerschmidt DM. 2012. Should I stay or should I go: Protec-
tion and maintenance of DNA methylation at imprinted
genes. Epigenetics 7: 969–975.
Messerschmidt DM, de Vries W, Ito M, Solter D, Ferguson-
Smith A, Knowles BB. 2012. Trim28 is required for epige-
netic stability during mouse oocyte to embryo transition. Sci-
ence 335: 1499–1502.
Messerschmidt DM, Knowles BB, Solter D. 2014. DNA meth-
ylation dynamics during epigenetic reprogramming in the
germline and preimplantation embryos. Genes Dev 28:
812–828.
Mouse Genome Sequencing Consortium, Waterston RH, Lind-
blad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agar-
wala R, Ainscough R, Alexandersson M, et al. 2002. Initial
sequencing and comparative analysis of the mouse genome.
Nature 420: 520–562.
Nielsen AL, Ortiz JA, You J, Oulad-Abdelghani M, Khechu-
mian R, Gansmuller A, Chambon P, Losson R. 1999. Inter-
action with members of the heterochromatin protein 1 (HP1)
family and histone deacetylation are differentially involved in
transcriptional silencing by members of the TIF1 family.
EMBO J 18: 6385–6395.
Nishitsuji H, Abe M, Sawada R, Takaku H. 2012. ZBRK1 re-
presses HIV-1 LTR-mediated transcription. FEBS Lett 586:
3562–3568.
Nishitsuji H, Sawada L, Sugiyama R, Takaku H. 2015. ZNF10
inhibits HIV-1 LTR activity through interaction with NF-kB
and Sp1 binding motifs. FEBS Lett 589: 2019–2025.
Okano M, Bell DW, Haber DA, Li E. 1999. DNA methyltrans-
ferases Dnmt3a and Dnmt3b are essential for de novo meth-
ylation and mammalian development. Cell 99: 247–257.
Okumura K, Sakaguchi G, Naito K, Tamura T, Igarashi H. 1997.
HUB1, a novel Kru¨ppel type zinc finger protein, represses the
human T cell leukemia virus type I long terminal repeat-me-
diated expression. Nucleic Acids Res 25: 5025–5032.
Peaston AE, Evsikov AV, Graber JH, de Vries WN, Holbrook
AE, Solter D, Knowles BB. 2004. Retrotransposons regulate
host genes in mouse oocytes and preimplantation embryos.
Dev Cell 7: 597–606.
Peng H, Ivanov AV, Oh HJ, Lau YFC, Rauscher FJ. 2009. Epi-
genetic gene silencing by the SRY protein is mediated by a
KRAB-O protein that recruits the KAP1 co-repressor machin-
ery. J Biol Chem 284: 35670–35680.
Peters J. 2014. The role of genomic imprinting in biology and
disease: An expanding view. Nat Rev Genet 15: 517–530.
Quenneville S, Verde G, Corsinotti A, Kapopoulou A, Jakobs-
son J, Offner S, Baglivo I, Pedone PV, Grimaldi G, Riccio A,
et al. 2011. In embryonic stem cells, ZFP57/KAP1 recognize
a methylated hexanucleotide to affect chromatin and DNA
methylation of imprinting control regions. Mol Cell 44:
361–372.
Ratnam S, Mertineit C, Ding F, Howell CY, Clarke HJ, Bestor
TH, Chaillet JR, Trasler JM. 2002. Dynamics of Dnmt1 meth-
yltransferase expression and intracellular localization during
oogenesis and preimplantation development. Dev Biol 245:
304–314.
Reik W. 2007. Stability and flexibility of epigenetic gene
regulation in mammalian development. Nature 447: 425–
432.
Rowe HM, Jakobsson J, Mesnard D, Rougemont J, Reynard S,
Aktas T, Maillard PV, Layard-Liesching H, Verp S, Marquis
J, et al. 2010. KAP1 controls endogenous retroviruses in em-
bryonic stem cells. Nature 463: 237–240.
Ryan RF, Schultz DC, Ayyanathan K, Singh PB, Friedman JR,
Fredericks WJ, Rauscher FJ. 1999. KAP-1 corepressor pro-
tein interacts and colocalizes with heterochromatic and
euchromatic HP1 proteins: A potential role for Kru¨ppel-asso-
ciated box-zinc finger proteins in heterochromatin-mediated
gene silencing. Mol Cell Biol 19: 4366–4378.
Santos F, Dean W. 2004. Epigenetic reprogramming during ear-
ly development in mammals. Reproduction 127: 643–651.
Satou A. 2001. A novel transrepression pathway of c-Myc. Re-
cruitment of a transcriptional corepressor complex to c-Myc
by MM-1, a c-Myc-binding protein. J Biol Chem 276:
46562–46567.
Satou A, Hagio Y, Taira T, Iguchi-Ariga SMM, Ariga H. 2004.
Repression of the c-fms gene in fibroblast cells by c-Myc-
MM-1-TIF1b complex. FEBS Lett 572: 211–215.
Schultz DC, Friedman JR, Rauscher FJ. 2001. Targeting histone
deacetylase complexes via KRAB-zinc finger proteins: The
PHD and bromodomains of KAP-1 form a cooperative unit
that recruits a novel isoform of the Mi-2a subunit of NuRD.
Genes Dev 15: 428–443.
Schultz DC, Ayyanathan K, Negorev D, Maul GG, Rauscher FJ.
2002. SETDB1: A novel KAP-1-associated histone H3, lysine
9-specific methyltransferase that contributes to HP1-mediat-
ed silencing of euchromatic genes by KRAB zinc-finger pro-
teins. Genes Dev 16: 919–932.
Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos
F, Popp C, Thienpont B, Dean W, Reik W. 2012. The dynam-
ics of genome-wide DNA methylation reprogramming in
mouse primordial germ cells. Mol Cell 48: 849–862.
Seki Y, Kurisaki A, Watanabe-Susaki K, Nakajima Y, Nakanishi
M, Arai Y, Shiota K, Sugino H, Asashima M. 2010. TIF1b
regulates the pluripotency of embryonic stem cells in a phos-
phorylation-dependent manner. Proc Natl Acad Sci 107:
10926–10931.
Surani MAH, Barton SC, Norris ML. 1984. Development of
reconstituted mouse eggs suggests imprinting of the genome
during gametogenesis. Nature 308: 548–550.
Takahashi K, Sugi Y, Hosono A, Kaminogawa S. 2009. Epige-
netic regulation of TLR4 gene expression in intestinal epithe-
lial cells for the maintenance of intestinal homeostasis. J
Immunol 183: 6522–6529.
Tan X, Xu X, Elkenani M, Smorag L, Zechner U, Nolte J, Engel
W, Pantakani DVK. 2013. Zfp819, a novel KRAB-zinc finger
protein, interacts with KAP1 and functions in genomic integ-
rity maintenance of mouse embryonic stem cells. Stem Cell
Res 11: 1045–1059.
Thomas JH, Schneider S. 2011. Coevolution of retroelements
and tandem zinc finger genes. Genome Res 21: 1800–1812.
Tsuruma R, Ohbayashi N, Kamitani S, Ikeda O, Sato N, Mur-
omoto R, Sekine Y, Oritani K, Matsuda T. 2007. Physical and
functional interactions between STAT3 and KAP1. Oncogene
27: 3054–3059.
Walsh CP, Chaillet JR, Bestor TH. 1998. Transcription of IAP
endogenous retroviruses is constrained by cytosine methyla-
tion. Nat Genet 20: 116–117.
LESEVA ET AL.162
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
Wang C, Ivanov A, Chen L, Fredericks WJ, Seto E, Rauscher FJ,
Chen J. 2005. MDM2 interaction with nuclear corepressor
KAP1 contributes to p53 inactivation. EMBO J 24: 3279–
3290.
Wang C, Rauscher FJ, Cress WD, Chen J. 2007. Regulation of
E2F1 function by the nuclear corepressor KAP1. J Biol Chem
282: 29902–29909.
Wiznerowicz M, Jakobsson J, Szulc J, Liao S, Quazzola A,
Beermann F, Aebischer P, Trono D. 2007. The Kru¨ppel-as-
sociated box repressor domain can trigger de novo promoter
methylation during mouse early embryogenesis. J Biol Chem
282: 34535–34541.
Wolf D, Goff SP. 2009. Embryonic stem cells use ZFP809 to
silence retroviral DNAs. Nature 458: 1201–1204.
Wolf G, Greenberg D, Macfarlan TS. 2015a. Spotting the enemy
within: Targeted silencing of foreign DNA in mammalian
genomes by the Kru¨ppel-associated box zinc finger protein
family. Mobile DNA 6: 17.
Wolf G, Yang P, Fu¨chtbauer AC, Fu¨chtbauer E-M, Silva AM,
Park C, Wu W, Nielsen AL, Pedersen FS, Macfarlan TS.
2015b. The KRAB zinc finger protein ZFP809 is required
to initiate epigenetic silencing of endogenous retroviruses.
Genes Dev 29: 538–554.
Wossidlo M, Nakamura T, Lepikhov K, Marques CJ, Zakhartch-
enko V, Boiani M, Arand J, Nakano T, Reik W, Walter J. 2011.
5-Hydroxymethylcytosine in the mammalian zygote is linked
with epigenetic reprogramming. Nat Commun 2: 241.
Yearim A, Gelfman S, Shayevitch R, Melcer S, Glaich O, Mallm
J-P, Nissim-Rafinia M, Cohen A-HS, Rippe K, Meshorer E,
et al. 2015. HP1 is involved in regulating the global impact
of DNA methylation on alternative splicing. Cell Rep 10:
1122–1134.
Zheng L, Pan H, Li S, Flesken-Nikitin A, Chen PL, Boyer TG,
Lee WH. 2000. Sequence-specific transcriptional corepressor
function for BRCA1 through a novel zinc finger protein,
ZBRK1. Mol Cell 6: 757–768.
Zuo X, Sheng J, Lau H-T, McDonald CM, Andrade M, Cullen
DE, Bell FT, Iacovino M, Kyba M, Xu G, et al. 2012. Zinc
finger protein ZFP57 requires its co-factor to recruit DNA
methyltransferases and maintains DNA methylation imprint
in embryonic stem cells via its transcriptional repression
domain. J Biol Chem 287: 2107–2118.
EPIGENETIC REPROGRAMMING 163
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
10.1101/sqb.2015.80.027441Access the most recent version at doi:
2016
2015 80: 155-163 originally published online January 13,Cold Spring Harb Symp Quant Biol
Milena Leseva, Barbara B. Knowles, Daniel M. Messerschmidt, et al.
Mammalian Epigenome
Establish: Natural Reprogramming of the−Maintain−Erase
References
http://symposium.cshlp.org/content/80/155.full.html#ref-list-1
This article cites 74 articles, 38 of which can be accessed free at:
License
Commons
Creative
Service
Email Alerting
click here.the box at the top right corner of the article or
Receive free email alerts when new articles cite this article - sign up in
http://symposium.cshlp.org/subscriptions
go to:Cold Spring Harbor Symposia on Quantitative BiologyTo subscribe to
Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved
Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from

More Related Content

What's hot

Epigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plantsEpigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plantsSachin Ekatpure
 
Dna methyltransferase screening
Dna methyltransferase screeningDna methyltransferase screening
Dna methyltransferase screeningBennie George
 
002 & 003 Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14002 & 003   Dna Methyl And Human Disease 14
002 & 003 Dna Methyl And Human Disease 14guest0fa715
 
Epigenetic regulation in higher plants
Epigenetic regulation in higher plantsEpigenetic regulation in higher plants
Epigenetic regulation in higher plantsFOODCROPS
 
Epigenetic regulation in plants
Epigenetic regulation in plantsEpigenetic regulation in plants
Epigenetic regulation in plantsRyza Priatama
 
Introduction to CpG island power point presentation
Introduction to CpG island power point presentationIntroduction to CpG island power point presentation
Introduction to CpG island power point presentationjkhdfhk
 
Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2mugdha-30
 
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium Poster
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium PosterSite-Directed Mutagenesis of β-2 Microglobulin Research Symposium Poster
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium PosterTyler Liang
 
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...wujunbo1015
 
Dna methylation ppt
Dna methylation pptDna methylation ppt
Dna methylation pptIbad khan
 
Endometriosis gene-expression, meta-analysis
Endometriosis gene-expression, meta-analysisEndometriosis gene-expression, meta-analysis
Endometriosis gene-expression, meta-analysisMehis Pold
 
Epigenetic regulation ataee
Epigenetic regulation ataeeEpigenetic regulation ataee
Epigenetic regulation ataeeelham ataie
 

What's hot (20)

Epigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plantsEpigenetics mediated gene regulation in plants
Epigenetics mediated gene regulation in plants
 
Dna methyltransferase screening
Dna methyltransferase screeningDna methyltransferase screening
Dna methyltransferase screening
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
002 & 003 Dna Methyl And Human Disease 14
002 & 003   Dna Methyl And Human Disease 14002 & 003   Dna Methyl And Human Disease 14
002 & 003 Dna Methyl And Human Disease 14
 
Epigenetic regulation in higher plants
Epigenetic regulation in higher plantsEpigenetic regulation in higher plants
Epigenetic regulation in higher plants
 
Epigenetic regulation in plants
Epigenetic regulation in plantsEpigenetic regulation in plants
Epigenetic regulation in plants
 
Dna methylation
Dna methylationDna methylation
Dna methylation
 
Introduction to CpG island power point presentation
Introduction to CpG island power point presentationIntroduction to CpG island power point presentation
Introduction to CpG island power point presentation
 
IME 1
IME 1IME 1
IME 1
 
Definition of epigenetics
Definition of epigeneticsDefinition of epigenetics
Definition of epigenetics
 
J Cell Sci 2005
J Cell Sci 2005J Cell Sci 2005
J Cell Sci 2005
 
Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2Mugdha's seminar msc sem 2
Mugdha's seminar msc sem 2
 
Epigenetics and Addiction Research - Michael Kobor
Epigenetics and Addiction Research - Michael Kobor Epigenetics and Addiction Research - Michael Kobor
Epigenetics and Addiction Research - Michael Kobor
 
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium Poster
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium PosterSite-Directed Mutagenesis of β-2 Microglobulin Research Symposium Poster
Site-Directed Mutagenesis of β-2 Microglobulin Research Symposium Poster
 
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
Mouse zar1 like (xm 359149) colocalizes with m-rna processing components and ...
 
Epigenetics
Epigenetics Epigenetics
Epigenetics
 
Dna methylation ppt
Dna methylation pptDna methylation ppt
Dna methylation ppt
 
Endometriosis gene-expression, meta-analysis
Endometriosis gene-expression, meta-analysisEndometriosis gene-expression, meta-analysis
Endometriosis gene-expression, meta-analysis
 
Epigenetic regulation ataee
Epigenetic regulation ataeeEpigenetic regulation ataee
Epigenetic regulation ataee
 

Viewers also liked (20)

Ekvationer
EkvationerEkvationer
Ekvationer
 
Estudio de caso
Estudio de casoEstudio de caso
Estudio de caso
 
Sinqueb
SinquebSinqueb
Sinqueb
 
Nerdfotainment
NerdfotainmentNerdfotainment
Nerdfotainment
 
Copia de pablo 10 años
Copia de pablo 10 añosCopia de pablo 10 años
Copia de pablo 10 años
 
I.e. técnica agropecuaría la granja
I.e. técnica agropecuaría la granjaI.e. técnica agropecuaría la granja
I.e. técnica agropecuaría la granja
 
2012 07-13-politica-nacional-de-extensao
2012 07-13-politica-nacional-de-extensao2012 07-13-politica-nacional-de-extensao
2012 07-13-politica-nacional-de-extensao
 
Architecture_Portfolio1
Architecture_Portfolio1Architecture_Portfolio1
Architecture_Portfolio1
 
Manual do sistema sem fio XD V35 Line 6 (PORTUGUÊS)
Manual do sistema sem fio XD V35 Line 6 (PORTUGUÊS)Manual do sistema sem fio XD V35 Line 6 (PORTUGUÊS)
Manual do sistema sem fio XD V35 Line 6 (PORTUGUÊS)
 
Clasificacion de las necesidades
Clasificacion de las necesidadesClasificacion de las necesidades
Clasificacion de las necesidades
 
DfoodFactory
DfoodFactoryDfoodFactory
DfoodFactory
 
NOSSO
NOSSONOSSO
NOSSO
 
Presentación1
Presentación1Presentación1
Presentación1
 
Procent, promille,ppm
Procent, promille,ppm Procent, promille,ppm
Procent, promille,ppm
 
Gina Indelicada - Marketing Digital
Gina Indelicada - Marketing DigitalGina Indelicada - Marketing Digital
Gina Indelicada - Marketing Digital
 
Jornades "Comunicació 3.0 i accessibilitat total". Ponència de Joan Gil: "Com...
Jornades "Comunicació 3.0 i accessibilitat total". Ponència de Joan Gil: "Com...Jornades "Comunicació 3.0 i accessibilitat total". Ponència de Joan Gil: "Com...
Jornades "Comunicació 3.0 i accessibilitat total". Ponència de Joan Gil: "Com...
 
Fasesolida
FasesolidaFasesolida
Fasesolida
 
ISMATDR : Coordinate Graphing Project
ISMATDR : Coordinate Graphing ProjectISMATDR : Coordinate Graphing Project
ISMATDR : Coordinate Graphing Project
 
The Incredible Shrinking CPM
The Incredible Shrinking CPMThe Incredible Shrinking CPM
The Incredible Shrinking CPM
 
RCP_2010_FR_111006_Def
RCP_2010_FR_111006_DefRCP_2010_FR_111006_Def
RCP_2010_FR_111006_Def
 

Similar to Cold Spring Harb Symp Quant Biol-2015-Leseva-sqb.2015.80.027441

A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docx
A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docxA RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docx
A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docxransayo
 
Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Jacob Wong
 
Nop2 is required for mammalian preimplantation development
Nop2 is required for mammalian preimplantation developmentNop2 is required for mammalian preimplantation development
Nop2 is required for mammalian preimplantation developmentWei Cui
 
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...Yara Tarabay
 
Fabrizio 2003 dev biol
Fabrizio 2003 dev biolFabrizio 2003 dev biol
Fabrizio 2003 dev biolJames Fabrizio
 
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFb
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFbCharacterizing Mouse Mammary Epithelial Cell Response to Activin and TGFb
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFbNatalia Trikoz
 
Epithelial to Mesenchymal Transition Epithelial vs.docx
Epithelial to Mesenchymal Transition Epithelial vs.docxEpithelial to Mesenchymal Transition Epithelial vs.docx
Epithelial to Mesenchymal Transition Epithelial vs.docxSALU18
 
Molecular embryology part (2)
Molecular embryology  part (2)Molecular embryology  part (2)
Molecular embryology part (2)Khaled Elmasry
 
Molecular embryology part (2) slideshow
Molecular embryology  part (2) slideshowMolecular embryology  part (2) slideshow
Molecular embryology part (2) slideshowKhaled Elmasry
 
Models of Human Diseases Conference (2010) Tetrahymena model by Dr. R. Pearl...
Models of Human Diseases Conference (2010)  Tetrahymena model by Dr. R. Pearl...Models of Human Diseases Conference (2010)  Tetrahymena model by Dr. R. Pearl...
Models of Human Diseases Conference (2010) Tetrahymena model by Dr. R. Pearl...Medical Education Advising
 
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription Elongation
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription ElongationTIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription Elongation
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription ElongationJoe Lee
 
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...Joe Lee
 
Hinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsHinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsAndrew Hinton
 

Similar to Cold Spring Harb Symp Quant Biol-2015-Leseva-sqb.2015.80.027441 (20)

A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docx
A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docxA RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docx
A RESEARCH ON GENOMIC IMPRINTINGGenomic imprinting is the epig.docx
 
Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013Wong J et al. - PLoS ONE - 2013
Wong J et al. - PLoS ONE - 2013
 
Nop2 is required for mammalian preimplantation development
Nop2 is required for mammalian preimplantation developmentNop2 is required for mammalian preimplantation development
Nop2 is required for mammalian preimplantation development
 
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
The mammalian-specific Tex19.1 gene plays an essential role in spermatogenesi...
 
Fabrizio 2003 dev biol
Fabrizio 2003 dev biolFabrizio 2003 dev biol
Fabrizio 2003 dev biol
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFb
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFbCharacterizing Mouse Mammary Epithelial Cell Response to Activin and TGFb
Characterizing Mouse Mammary Epithelial Cell Response to Activin and TGFb
 
Epithelial to Mesenchymal Transition Epithelial vs.docx
Epithelial to Mesenchymal Transition Epithelial vs.docxEpithelial to Mesenchymal Transition Epithelial vs.docx
Epithelial to Mesenchymal Transition Epithelial vs.docx
 
Molecular embryology part (2)
Molecular embryology  part (2)Molecular embryology  part (2)
Molecular embryology part (2)
 
Molecular embryology part (2) slideshow
Molecular embryology  part (2) slideshowMolecular embryology  part (2) slideshow
Molecular embryology part (2) slideshow
 
Models of Human Diseases Conference (2010) Tetrahymena model by Dr. R. Pearl...
Models of Human Diseases Conference (2010)  Tetrahymena model by Dr. R. Pearl...Models of Human Diseases Conference (2010)  Tetrahymena model by Dr. R. Pearl...
Models of Human Diseases Conference (2010) Tetrahymena model by Dr. R. Pearl...
 
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription Elongation
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription ElongationTIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription Elongation
TIF1-gamma Controls Erythroid Cell Fate by Regulating Transcription Elongation
 
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...
TiF1-gamma Plays an Essential Role in Murine Hematopoiesis and Regulates Tran...
 
3ab Recovery_epichr
3ab Recovery_epichr3ab Recovery_epichr
3ab Recovery_epichr
 
GenesDev_2008
GenesDev_2008GenesDev_2008
GenesDev_2008
 
Transgenic.pptx
Transgenic.pptxTransgenic.pptx
Transgenic.pptx
 
Hinton et al 2012 Adv Genetics
Hinton et al 2012 Adv GeneticsHinton et al 2012 Adv Genetics
Hinton et al 2012 Adv Genetics
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
CTR 2016 april ALW & H
CTR 2016 april ALW & HCTR 2016 april ALW & H
CTR 2016 april ALW & H
 
replicación 3.pdf
replicación 3.pdfreplicación 3.pdf
replicación 3.pdf
 

Cold Spring Harb Symp Quant Biol-2015-Leseva-sqb.2015.80.027441

  • 1. Erase–Maintain–Establish: Natural Reprogramming of the Mammalian Epigenome MILENA LESEVA,1 BARBARA B. KNOWLES,2 DANIEL M. MESSERSCHMIDT,1 AND DAVOR SOLTER 3 1 Department for Developmental Epigenetics and Disease, Institute of Molecular and Cell Biology, AÃ STAR, 138673 Singapore 2 Emerita, The Jackson Laboratory, Bar Harbor, Maine 04609 3 Emeritus Member and Director, Max-Planck Institute of Immunobiology and Epigenetics, 79180 Freiburg, Germany Correspondence: Danielm@imcb.a-star.edu.sg; davorsolter@mac.com The genetic information is largely identical across most cell types in a given organism but the epigenome, which controls expression of the genome,is cell type– and context-dependent. Although most mature mammalian cells appear to have a stable, heritable epigenome, a dynamic intricate process reshapes it as these cells transition from soma to germline and back again. During normal embryogenesis, primordial germ cells, of somatic origin, are set aside to become gametes. In doing so their genome is reprogrammed—that is, the epigenome of specific regions is replaced in a sex-specific fashion as they terminally differentiate into oocytes or spermatocytes in the gonads. Upon union of these gametes, reprogramming of the new organism’s epigenome is initiated, which eventually leads, through pluripotent cells, to the cell lineages required for proper embryonic development to a sexually mature adult. This never-ending cycle of birth and rebirth is accomplished through methylation and demethylation of specific genomic sites within the gametes and pluripotent cells of an organism. This enigmatic process of natural epigenomic reprogramming is now being dissected in vivo, focusing on specific genomic regions—that is, imprinted genes and retrotransposons, where TRIM28 molecular complexes appear to guide the transition from gamete to embryo. Mammalian reproduction requires union of two highly specialized cells, the egg and sperm, at fertilization, an event that marks the beginning of new life. Epigenetic marks of this newly formed genome are immediately required to establish the cell type–specific gene expres- sion programs required for embryonic growth and devel- opment. DNA methylation is an epigenetic modification that leads to repression of specific gene expression, X- chromosome inactivation, and silencing of mobile DNA elements (for review, see Messerschmidt et al. 2014) and affects alternative splicing (Yearim et al. 2015). Establishment and maintenance of stable methylation patterns are thus crucial to execution of a normal devel- opmental program (Okano et al. 1999). The DNA meth- ylome should be, and is, extremely plastic during very early embryogenesis. Its dynamic control is initiated at fertilization during the switch from germ cell to embryo- specific methylation and a reciprocal change manifests during germ cell specification with the switch from a somatic to a germline epigenomic signature (Fig. 1). Fertilization triggers extensive reprogramming where- by the global DNA hypermethylation of the germ cell– specific epigenomes become, on the whole, demethylat- ed. The paternal genome in the pronucleus of the zygote is rapidly demethylated and reorganized. The methyla- tion of the maternal genome is gradually depleted in a replication-dependent manner over consecutive mitotic divisions (Santos and Dean 2004). Although the precise mechanics of the active process are not completely un- derstood, TET3-mediated oxidation of 5-methylcytosine (5mC) (Gu et al. 2011; Iqbal et al. 2011; Wossidlo et al. 2011) and restoration of unmethylated cytosines by com- ponents of the base excision repair pathway (Hajko- va et al. 2010) are involved. These changes to the em- bryonic genome lead to an open chromatin state, essential for the pluripotency of some cells in the mouse blasto- cyst’s inner cell mass (ICM). Reacquisition of de novo promoter methylation occurs between the time of estab- lishment of the ICM of the blastocyst and the develop- ment of the postimplantation epiblast when the cells of the embryo differentiate into the somatic lineages (Borgel et al. 2010). Resetting the epigenome is also mandatory during pri- mordial germ cell (PGC) development when the methyl- ation marks of the parental epigenome are erased and new gender-specific imprinting patterns are established (Sei- senberger et al. 2012). PGCs are specified at E6.5 from a small group of BLIMP1-positive cells located in the somatic proximal postimplantation epiblast (for review, see Magnu´sdo´ttir and Surani 2014). In mouse PGCs 5mC levels gradually decline in a partially TET1/2-dependent manner, as the cells migrate through the hindgut endo- derm (E9.5), reach the gonadal anlagen (E11.5), and un- dergo sexual determination (E13.5) (Fig. 1). Given the size and complexity of the mammalian ge- nome, it is fascinating that demethylation and remethyl- ation events are intricately controlled both temporally and in a genome context-specific manner. How are cer- Copyright # 2015 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/sqb.2015.80.027441 Cold Spring Harbor Symposia on Quantitative Biology, Volume LXXX 155 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 2. tain loci identified to escape the loss of DNA methylation in an environment that favors genome-wide demethyla- tion events? Recent studies of the molecular interactions at specific sites in imprinted genes and in endogenous retroelements (EREs) have afforded a glimpse of this complex process. Because methylation protection of spe- cific DNA regions initiates immediately after fertiliza- tion, and during a time of complete transcriptional silence, the molecular agents, either RNA and/or protein, must be in the oocyte—that is, maternally inherited. The role of one such maternal factor, TRIM28, will be used to illustrate the complexity of this process in the mouse. TRIM28—A MULTIFACETED WORKER IN THE CHROMATIN FIELD Tripartite motif containing 28/KRAB-domain-asso- ciated protein1/transcription intermediary factor-1b (TRIM28/KAP1/TIF1b) is a multifunctional protein implicated in processes as diverse as embryonic de- velopment, maintenance of pluripotency, imprinting, endogenous and exogenous retroviral silencing, and dou- ble-strand break DNA damage response, to name a few. The structure of TRIM28, with its amino-terminal RING, B-box, and coiled-coil (RBCC) domain, central HP1- binding domain, and carboxy-terminal PHD-bromodo- mains, provides it with the means to interact with multiple transcription factors (TFs) and a variety of chromatin re- modelers and epigenetic modifiers (Iyengar and Farnham 2011; Cheng 2014). TRIM28 itself does not bind to DNA directly but rather relies on DNA-binding TF-mediated recruitment. These are largely members of the Kru¨ppel-associated box (KRAB) domain zinc-finger proteins (ZFPs), a family of TFs containing more than 200 genes in mice and hu- mans that encode distinct proteins with highly variable DNA-binding domains. KRAB-ZFPs may have evolved from Prdm9 (Meisetz) (Birtle and Ponting 2006) and ex- panded rapidly by duplication events during evolution. Loss of the original SET domain, accumulation of se- quence changes to the ancestral KRAB domain, and ap- pearance of corepressors are responsible for establishing the KRAB-ZFPs as transcriptional repressors. One expla- nation for the huge expansion and diversification of these TFs lies in their presumed coevolution with EREs to prevent their transcriptional reactivation (Thomas and Schneider 2011). KRAB-ZFPs contain tandem arrays of C2H2 zinc fin- gers, each engaging three consecutive nucleotides on one DNA strand and one nucleotide on the opposite strand. The combination of multiple ZNFs within one protein allows KRAB-ZFPs to bind with high specificity and to recognize long stretches of DNA. The amino-terminally encoded KRAB domain is highly conserved throughout the KRAB-ZFP family and mediates interaction with TRIM28. Numerous other TFs have been reported to interact with and potentiallyattractTRIM28 to their DNA-binding sites (Fig. 2; Satou 2001; Li et al. 2003; Satou et al. 2004; Wang et al. 2005, 2007; Jin et al. 2007; Tsuruma et al. 2007; Kamitani et al. 2008, 2011; Peng et al. 2009; Seki et al. 2010; Liang et al. 2011; Maruyama et al. 2011; Huang et al. 2013). For most of these, the KRAB-ZFPs included, TRIM28functionsasaco-repressordown-regulatinggene transcription by its heterochromatin-inducing activity at its target sites (Iyengar and Farnham 2011; Cheng 2014). 1 2 3 5 4 6 7 8 Figure 1. Epigenetic reprogramming during the mammalian reproductive cycle. (1) Both male and female germ cells are differentiated cell types and have highly specialized epigenomes. Fertilization of the oocyte by sperm initiates embryonic development. (2) The paternal genome (blue) is rapidly demethylated at early pronuclear stages. (3) The maternal genome (pink) is passively demethylated in a replication-dependent manner. (4) Erasure of germ cell–specific DNA methylation patterns allows the establishment of the pluripotent inner cell mass of the blastocyst. (5) Pluripotent cells begin the acquisition of DNA methylation marks as they begin to differentiate into somatic lineages. (6) PGCs are specified in the proximal postimplantation epiblast. They carry somatic epigenetic features, such as genomic imprints, inactive X-chromosomes, and silenced germ cell/pluripotency genes. (7) These features and potential epimutations are erased during primordial germ cell reprogramming. (8) Male- and female-specific germ cell differentiation includes establishment of paternal and maternal imprints. LESEVA ET AL.156 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 3. TRIM28 acts as a scaffolding protein associating with the H3K9me3-writer SETDB1/ESET, the H3K9me3- reader HP1, and the nucleosome remodeling and histone deacetylation complex NuRD (Schultz et al. 2002, 2001). This multisubunit, heterochromatin-inducing molecular complex also serves as a platform for recruiting DNA methyltransferases, which can then induce and maintain DNA methylation in proximity to their target sites (Fig. 2; Quenneville et al. 2011; Zuo et al. 2012). From a developmental biologist’s point of view, it was known for some time that Trim28 was highly expressed in the preimplantation embryo and that its zygotic knockout is embryonic lethal in early postimplantation stages (Cammas et al. 2000). Trim28 was also identified in in- dependent screens for factors required for pluripotency and self-renewal of embryonic stem cells (ESCs) (Fazzio et al. 2008; Hu et al. 2009). However, the precise role of Trim28 in mammalian cells remained unclear, although it was found to relocalize to centromeric heterochromatin upon cell differentiation (Cammas et al. 2002). Additional insights to TRIM28 function in vivo were gained from studies in preimplantation embryos Trim28 STAT3 STAT1 SRY/KRAB-O HP1α HP1γ HP1β Histonedeacetylation N-CoR NuRD HDAC1 SMARCAD1 DNMT1 DNMT3b DNMT3a DNA methylation SETDB1 Histone methylation Pou5f1 Myc E2F1 p53 IRF7 HIF1α NRF2 FOXP3 NFκB KRAB-ZFPs TranscriptionFactors B Interaction Partners of TRIM28 C KRAB-ZFPs & Function Genomic imprinting maintenance exo- and endogenous ERE silencing ZFP57 ZFP809 ZFP819 Gm6871 MuLV IAP,LINE LINE Mouse Mouse/ Human gDMRs Human ZNF350 ZNF175 ZNF10 HIV ZNF93 L1 ZNF91 SVA ZNF282 HTLV Chromatin readers Chromatin writers Chromatinmodifiers A The TRIM28-complex G9a DNMTs G9a K9K9K9 K9 NuRD SETDB1 H3 H3 H3 H3 HP1 NH2 N NH2 N R NH2 OOOOOO N NH2NH22 NNNNNN R 222 CH3 TF TRIM28 1 2 3 others ZNF160Human TLR4 ZBRKMouse p21 ZNF689 miRNAs O R NNNNOO Figure 2. The TRIM28 complex and interactome. (A) TRIM28 acts as a central scaffolding protein for a multifunctional, large protein complex. It is guided to DNA by interacting transcription factors and attracts in turn: (1) The NuRD histone deacetylation complex (deacetylates H3K9Ac); (2) SETDB1 and G9a histone methyltransfereases (trimethylate H3K9me3); (3) DNA methyltransferases (DNMTs) (methylate cytosines). Heterochromatin protein 1 (HP1) recognizes H3K9me3 chromatin and further physically interacts with TRIM28. (B) TRIM28 interaction partners: KRAB-ZFPs and other TFs can target TRIM28 to DNA (left) (Satou 2001; Li et al. 2003; Satou et al. 2004; Wang et al. 2005, 2007; Jin et al. 2007; Tsuruma et al. 2007; Kamitani et al. 2008, 2011; Peng et al. 2009; Seki et al. 2010; Liang et al. 2011; Maruyama et al. 2011; Huang et al. 2013). TRIM28 attracts chromatin writers and readers (right) (Nielsen et al. 1999; Ryan et al. 1999; Schultz et al. 2001, 2002; Quenneville et al. 2011; Zuo et al. 2012). (C) Few KRAB-ZFP proteins have been studied in detail. ZFP57 plays a conserved role in imprinting maintenance in humans and mice. Diverse ZFPs/ZFNs (mouse/human) are involved in retrovirus silencing and other physiological functions (Okumura et al. 1997; Zheng et al. 2000; Li et al. 2008; Takahashi et al. 2009; Wolf and Goff 2009; Quenneville et al. 2011; Nishitsuji et al. 2012, 2015; Tan et al. 2013; Castro-Diaz et al. 2014; Jacobs et al. 2014; Wolf et al. 2015b). EPIGENETIC REPROGRAMMING 157 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 4. (Messerschmidt 2012; Messerschmidt et al. 2012). Dele- tion of maternal Trim28 alone causes embryonic lethality, despite its reexpression from the paternal allele follow- ing embryonic genome activation at the two-cell stage. Intriguingly, the maternal-null embryos show highly variable phenotypes and die at various times throughout gestation, albeit at a much later time than when the molecular deficiency of TRIM28 first occurs. The pleio- tropic defects displayed by these embryos are indicative of accumulated, random combinations of epimutations incompatible with life (Messerschmidt et al. 2012; Lor- thongpanich et al. 2013). We now know that TRIM28 is essential for proper and successful epigenetic reprogramming through at least two fundamental functions—that is, maintaining imprint- ing and silencing retrotransposons. THE TRIM28-COMPLEX FUNCTION IN GENOMIC IMPRINTING Genomic imprinting, or the establishment and mainte- nance of monoallelic expression of particular genes in a parent-of-origin-specific manner, is at the heart of the developmental failure observed in genetically bimaternal (gynogenote) and bipaternal (androgenote) embryos. The observation that the incomplete development of embryos derived from gynogenotes is largelyembryo-like, whereas that of androgenotes is mostly of extraembryonic-like tis- sues, showed that the maternal and paternal genomes are functionally nonequivalent (McGrath and Solter 1984; Surani et al. 1984). Imprinting implies that the maternal and paternal genomes are differentially marked at specif- ic genetic loci but does not involve changes in the DNA sequence. These epigenetic marks regulate gene expres- sion, are mitotically heritable, and are set during female and male gametogenesis, respectively. Imprinted genes are often clustered in specific sites of the genome and within these clusters they can be coordinately regulated by differentially methylated regions at the so-called im- printing control regions (ICRs). Methylation at ICRs ul- timately controls gene expression in cis by multiple mechanisms. Many imprinting clusters express noncod- ing RNAs (ncRNAs) that can further regulate genes within the cluster in trans. Collectively, epigenetic mod- ification ensures that maternally imprinted genes are expressed from the maternally transmitted allele and si- lenced on the paternal allele, and vice versa (Hanna and Kelsey 2014; Peters 2014). Parent-specific DNA methylation patterns at the ICRs are instituted during gametogenesis, part of a process requiring erasure of the inherited parental marks, so that imprints can be established sex-specifically. These paren- tal imprints are protected and preserved postfertilization and stabilized throughout embryonic and adult life, as proper levels of the product of imprinted genes are essen- tial to embryonic development, or are involved in adult cell and tissue homeostasis. For this reason, specialized mechanisms must be put in place to protect such sites from erasure during the wave of demethylation in the early embryo. Genomic imprints are therefore paradig- matic of the subtle epigenetic fine-tuning required during genomic reprogramming. Although the genomic imprint- ing cycle of “erasure–establishment–maintenance and erasure again” has been postulated and shown for some time, the molecular mechanisms behind imprint mainte- nance involving the TRIM28 complex were only recently unraveled. Demethylation in the preimplantation embryos is achieved by a combination of active and passive erasure of the methylation from 5mC (for review, see Messer- schmidt et al. 2014). To allow for efficient methylation removal, the maintenance DNA methyltransferase-1 (DNMT1) is largely excluded from the nuclei of the cleavage-stage embryos (Howell et al. 2001; Ratnam et al. 2002; Branco et al. 2008; Hirasawa et al. 2008). However, this compartmentalization and nuclear exclu- sion cannot be complete, as genetic deletion of both ma- ternal and zygotic Dnmt1 results in imprinting defects, the result of complete ICR demethylation (Hirasawa et al. 2008). DNMT1 is canonically targeted to the replication fork in mitotic cells by its cofactor NP95. Because the drastically reduced DNMT1 levels and the global DNA methylation maintenance function of this complex make it unlikely to maintain imprints in embryos, a noncanon- ical targeting mechanism of DNMT1 had been proposed. Insights into how DNMT1 might be targeted specifi- cally to ICRs came from studies in vivo and in vitro. We were able to show that loss of maternal Trim28 alone can result in dramatic misexpression of imprinted genes in midgestation embryos (Messerschmidt 2012; Messerschmidt et al. 2012, 2014; Lorthongpanich et al. 2013). This observed deregulation was observed across multiple imprinting clusters with the deregulation fol- lowing a pattern predicated on a loss-of-ICR-methylation scenario. Indeed, partial, severe ICR demethylation, pre- dominantly at the paternally imprinted H19/IGF2 locus, was found in these embryos. All tested imprinted loci, however, displayed variable degrees of methylation loss, arguing for (1) a global mechanism and (2) a partial res- cue of the defect by paternal Trim28 expression (Messer- schmidt et al. 2012; Lorthongpanich et al. 2013). Indeed, single-cell analysis of DNA methylation in eight-cell- stage embryos revealed that not all blastomeres are affected equally; hence, the pleiotropic defects observed are likely a consequence of epigenetic mosaicism in the mutant embryos. Interestingly, maternal/zygotic-null Trim28 embryos develop to apparently normal blasto- cysts and implant (DM Messerschmidt, unpubl.); thus, the complete knockout does not show a phenotype during preimplantation embryogenesis. However, it remains to be seen whether loss of both maternal and paternal Trim28 will result in complete eradication of ICR meth- ylation in preimplantation embryos. TRIM28 is physically bound to known ICRs in mid- gestation embryos, arguing for a direct recognition and maintenance mechanism (Messerschmidt et al. 2012). ICRs in these embryos are co-occupied by ZFP57 and as a consequence of the binding of the TRIM28 complex, histones were H3K9me3 modified (Messerschmidt et al. LESEVA ET AL.158 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 5. 2012). ZFP57 is a TRIM28-interacting KRAB-ZFP and both maternal and zygotic Zfp57 are necessary for the proper maintenance of parental imprints in blastocysts (Li et al. 2008). Investigations in ESCs showed ZFP57 recognizes a CpG-containing consensus TGCCGC se- quence, highly conserved at ICRs (Quenneville et al. 2011). However, TRIM28 binding to hypomethylated sites was lost in embryos (Messerschmidt et al. 2012) leading us to suggest that ZFP57 recognizes the methyl- ated cytosine. Indeed, ZFP57 shows much higher affinity to its methylated target sequence in vitro (Quenneville et al. 2011; Liu et al. 2012). Thus, sequence-specific and methylation-dependent binding to ICRs offers an elegant solution to the convoluted undertaking of (1) rec- ognizing specialized regions within the genome and (2) maintaining differential DNA methylation at a given locus. It also explains the lack of paternal rescue in the maternal Trim28-null embryo because preexisting DNA-methylated sites, inherited from the gametes, are fundamental to the ZFP57-mediated TRIM28-complex binding. If a particular CpG is demethylated under ma- ternal-null circumstances, reexpressed paternal TRIM28 cannot be targeted to it and cannot restore DNA methyl- ation. Similarly, reexpression of ZFP57 in Zfp57-deleted ES cells cannot restore methylation to sites that have lost it (Zuo et al. 2012). Finally, this mode of interaction offers a plausible model for the noncanonical targeting of the dramatically reduced levels of DNMT1 to ICRs in preimplantation embryos; both maintenance DNA- methyltransferase and the de novo DNA-methyl- transferases (DNMT3A/B) are bona fide interaction part- ners of TRIM28 (Quenneville et al. 2011; Zuo et al. 2012). The highly specialized role and mode of chromatin interaction of ZFP57 with specific genomic sequences, be they methylated or not, is an exquisite example of the functional diversity acquired by the KRAB-ZFPs. Fur- ther, the multiple interactions made possible through the TRIM28 scaffolding complex not only clarify the specificity of epigenetic reprogramming but also accen- tuate the importance of the balance between erasure of most parental epigenetic features versus the careful main- tenance of a few stable epigenetic sites. THE TRIM28 COMPLEX IN RETROTRANSPOSON REGULATION Mammalian cells face a formidable challenge in deal- ing with potentially damaging endogenous retroviruses (ERVs) and retrotransposons, collectively termed endog- enous retroviral elements (EREs). These elements make up 40% of the mammalian genome, highlighting the magnitude of this task (Lander et al. 2001; Mouse Ge- nome Sequencing Consortium et al. 2002). Aberrant activation of EREs can impact the host on two major levels. For one, certain retrotransposons remain capable of active retrotransposition that can lead to ran- dom insertional mutagenesis following replicative trans- position into the host’s genome. This must be prevented at all costs, particularly in the germline. Most EREs, however, have lost their ability to replicate but still their remnants are important targets of the host’s silencing machinery. Loss of silencing at such sites can result in drastic, localized chromatin changes and impact on cer- tain cellular genes whose expression may be under the influence of neighboring, ERE-derived promoters/en- hancers. In certain contexts, ERE control over gene regulation and mobility is of beneficial effect to the host. A case in point is the developmentally regulated expression of nonconventional chimeric transcripts driv- en by transposon-derived 50 sequences that can encode new proteins with potentially novel functions (Peaston et al. 2004). Temporary loss of a silencing mechanism can also explain the stage-specific expression of different long terminal repeat (LTR) retrotransposons during the oocyte-to-embryo transition (Peaston et al. 2004). As it now stands, most EREs are continuously repressed and the first line of defense against their expression is DNA methylation–based silencing, substantially contributing to the overall hypermethylated state of the mammalian genome. One insight into the molecular mechanism control- ling retrotransposon silencing during embryonic develop- ment came from the discovery that Dnmt1-deficient embryos reactivate all major classes of the intracisternal A-type particles (IAPs) (Walsh et al. 1998). Later, DNMT3L was shown to protect against spurious tran- scriptional reactivation of long interspersed nuclear ele- ments 1 (L1NE1) and ID1 IAP retrotransposons in male germ cells (Bourc’his and Bestor 2004). Extensive re- search has shown that during mammalian reproduction, when DNA methylation patterns are globally modified and removed, other mechanisms serve as a barrier to ERE activation. The small RNAs (piRNAs) seem to pre- dominant in the germline but in the embryo piRNAs play a minor role to that of the KRAB-ZFP TFs, which medi- ate formation of heterochromatin. One transacting repressor complex specific to pluripo- tent cells, and present in embryos, was shown to be im- portant for ERV silencing. Initially termed binding factor A, this 1.3-MDa multisubunit complex contains TRIM28 as a repressor subunit (Reik 2007) and the KRAB- ZFP809 that serves as its sequence-specific DNA-bind- ing partner (Wolf and Goff 2009). TRIM28 links the DNA methylation and KRAB-ZFP repressive machiner- ies as shown by tethering a doxycycline-controllable KRAB domain to a lentiviral construct in vivo (Wizner- owicz et al. 2007). Here, a green fluorescent protein (GFP) reporter gene was found to be expressed only if recruitment of the repressive KRAB-ZFP was prevented by adding doxycycline in a narrow time window right after fertilization (between E0.5 and E3.5). Conversely, GFP silencing was permanently established if the KRAB- containing fusion protein bound its target sequence very early in embryogenesis. ERVs are classified based on their sequence similari- ties. Class I and particularly class II ERVs such as IAPs and MusD are drastically deregulated in mouse ESCs lacking TRIM28. Also, treatment of Trim28 knockout- EPIGENETIC REPROGRAMMING 159 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 6. derived ESCs with 5-azacytidine (a DNMT inhibitor) has a synergistic effect on ERV activation. However, con- current reactivation is accompanied by depletion of the H3K9me3 histone modification of the IAP and MusD sequence. DNA methylation–independent silencing in ESC has also been described, which requires TRIM28 and SETDB1 but does not rely on de novo or maintenance methylation because Dnmt1/3a/3b triple-knockout ESCs do not display the massive derepression of murine leuke- mia virus (MLV), IAP, and MusD observed in Setdb1- or Trim28-deficient cells (Matsui et al. 2010; Rowe et al. 2010). It appears that certain redundant mechanisms func- tion to ensure efficient ERV silencing; the preferred mechanism depends on the class and evolutionary history of the particular element (ancient vs. young). It has been suggested that the repressive KRAB-ZFP machinery likely coevolved together with the ERVs as a host mech- anism to restrict their horizontal and vertical transmission (Wolf et al. 2015a,b). Surprisingly, only a limited number of KRAB-ZFPs have been shown to regulate specific ERV classes in mice. Such examples include the regula- tion of MLV by ZFP809 described earlier, of IAPs and LINE1 by ZFP819, and of LINE1 by Gm6871 (Fig. 2). It remains to be seen whether and which KRAB-ZFPs are still actively involved exclusively in retrotransposon si- lencing and whether many have evolved to undertake “endogenous” host gene regulatory functions, such as ZFP57. ERV repression by the TRIM28 complex has been scantily studied in vivo. Class II ERVs, particularly IAP elements, are known to be massively activated in zygotic Trim28 knockout blastocysts and periimplantation em- bryos (Rowe et al. 2010). During oocyte growth, these elements are generally repressed (Peaston et al. 2004) but ZP3-cre-mediated removal of maternal Trim28 in the growing oocyte does not result in IAP or MusD derepression nor in the ectopic activation of any other retroviral element tested here (Fig. 3). Thus, Trim28- independent mechanisms, in collaboration with DNA methylation and possibly piRNAs, may act to silence ERVs in the mouse egg. As early as the two-cell stage, when the embryonic genome is activated, a veritable explosion of ERV tran- scription takes place. Predominantly class III mouse tran- script (MT) and ERV-like elements (ERVL) and class II elements (IAPs) are transcribed (Kigami et al. 2003; Peaston et al. 2004). This massive activation of ERVs gives the two-cell-stage mouse embryo a characteristic transcriptome that is recapitulated in a small subfraction (1%) of ESCs (Kigami et al. 2003; Macfarlan et al. 2012). It is unclear whether this wave of transcriptional activa- tion is required for proper development or is only a by- product of epigenetic reprogramming in the embryo. Mouse (Mu) ERVL transcription is regulated by chro- matin modifications, particularly H3K4me3. Removal of LSD1/KDM1, an H3K4me3 demethylase, results in MuERVL activation and embryonic arrest at gastrulation (Macfarlan et al. 2012). We find loss of maternal Trim28 does not impact MuERVL expression or that of any other class III or class I ERV tested. Also LINE and short interspersed nuclear element (SINE) expression re- mained unchanged (Fig. 3). Remarkably, despite the mas- sive endogenous activation of class II element observed at the two-cell stage, loss of maternal Trim28 causes a hyperactivation of IAPs and MusD (Fig. 3). The dynam- ics of this regulation and to what extent this overexpres- sion, with all its repercussions, contributes to the maternal-deletion phenotype of Trim28 are unclear and warrant further investigation. MII OocyteA Two-Cell StageB IAP 100 2000 Trim28 100 2000 Oct4 100 2000 Line1 100 2000 Sine (ALU) 100 2000 MusD 100 2000 RLTR1b 100 2000 LTR class II LTR class I ORRA1A 2000 100 400300 MuERVL 100 2000 MT 100 2000 LTR class IIInon-LTR elements Trim28 100 2000 Oct4 100 2000 Line1 100 2000 Sine (ALU) 100 2000 MusD 2000 100 400300 IAP 2000 400 600 RLTR1b 100 2000 MuERVL 100 2000 MT 100 2000 ORRA1A 100 2000 LTR class II LTR class III LTR class I non-LTR elements Trim28 maternal-null Trim28 wild type Figure 3. ERV expression in maternal-null Trim28 and wild-type oocytes and embryos. Long terminal repeat (LTR) and non-LTR retrotransposon expression was tested in (A) MII oocytes and (B) two-cell-stage embryos in the presence and absence of maternal Trim28 (white and black bars, respectively). Although oocytes show no changes in ERV expression, two-cell-stage maternal-null Trim28 embryos show elevated expression of the LTR class II elements, IAP and MusD. LESEVA ET AL.160 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 7. CONCLUSION Establishment of a unique epigenome is the basis for correct functioning of each cell in a multicellular organ- ism. Mechanistically we can envision the existence of two types of epigenome. The dynamic or transitory epige- nome is unique to each cell and tissue type, and it is driven by classical TFs. It is malleable and repro- grammable as showed by the phenomena of cloning, transdifferentiation, and induced pluripotent stem (iPS) cell derivation. In the stable epigenome, as represented by imprinted genes and retroelements, silencing is based predominantly on stable genomic modifications, which are mitotically inherited and change only at the specific stages of the life cycle of the organism. Establishment and control of the stable epigenome has likelyevolved through an evolutionary process we are just beginning to decipher. Considering the importance of a stable epigenome to nor- mal development, significant efforts should be undertak- en to understand its establishment and maintenance. ACKNOWLEDGMENTS We thank the members of the Developmental Epige- netics and Disease Laboratory and Heike Wollmann for their comments and input to this work; Mimi DeVries, The Jackson Laboratory, USA and Anne Peaston, Uni- versity of Adelaide, Roseworthy, Australia for their early contributions. M.L. and D.M.M. are supported by the National Research Foundation (Singapore), AÃ STAR’s Bio-Medical Research Council, and AÃ STAR’s Institute of Molecular and Cell Biology, Singapore. B.B.K., D.S., and D.M.M. were previously supported by AÃ STAR’s Institute of Medical Biology, Singapore. REFERENCES Birtle Z, Ponting CP. 2006. Meisetz and the birth of the KRAB motif. Bioinformatics 22: 2841–2845. Borgel J, Guibert S, Li Y, Chiba H, Schu¨beler D, Sasaki H, Forne´ T, Weber M. 2010. Targets and dynamics of promot- er DNA methylation during early mouse development. Nat Genet 42: 1093–1100. Bourc’his D, Bestor TH. 2004. Meiotic catastrophe and retro- transposon reactivation in male germ cells lacking Dnmt3L. Nature 431: 96–99. Branco MR, Oda M, Reik W. 2008. Safeguarding parental identity: Dnmt1 maintains imprints during epigenetic repro- gramming in early embryogenesis. Genes Dev 22: 1567– 1571. Cammas F, Mark M, Dolle´ P, Dierich A, Chambon P, Losson R. 2000. Mice lacking the transcriptional corepressor TIF1b are defective in early postimplantation development. Develop- ment 127: 2955–2963. Cammas F, Oulad-Abdelghani M, Vonesch J-L, Huss-Garcia Y, Chambon P, Losson R. 2002. Cell differentiation induces TIF1b association with centromeric heterochromatin via an HP1 interaction. J Cell Sci 115: 3439–3448. Castro-Diaz N, Ecco G, Coluccio A, Kapopoulou A, Yazdanpa- nah B, Friedli M, Duc J, Jang SM, Turelli P, Trono D. 2014. Evolutionally dynamic L1 regulation in embryonic stem cells. Genes Dev 28: 1397–1409. Cheng C-T. 2014. KAPtain in charge of multiple missions: Emerging roles of KAP1. World J Biol Chem 5: 308–320. Fazzio TG, Huff JT, Panning B. 2008. An RNAi screen of chro- matin proteins identifies Tip60-p400 as a regulator of embry- onic stem cell identity. Cell 134: 162–174. Gu T-P, Guo F, Yang H, Wu H-P, Xu G-F, Liu W, Xie Z-G, Shi L, He X, Jin S-G, et al. 2011. The role of Tet3 DNA dioxy- genase in epigenetic reprogramming by oocytes. Nature 477: 606–610. Hajkova P, Jeffries SJ, Lee C, Miller N, Jackson SP, Surani MA. 2010. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 329: 78–82. Hanna CW, Kelsey G. 2014. The specification of imprints in mammals. Heredity 113: 176–183. Hirasawa R, Chiba H, Kaneda M, Tajima S, Li E, Jaenisch R, Sasaki H. 2008. Maternal and zygotic Dnmt1 are necessary and sufficient for the maintenance of DNA methylation im- prints during preimplantation development. Genes Dev 22: 1607–1616. Howell CY, Bestor TH, Ding F, Latham KE, Mertineit C, Trasler JM, Chaillet JR. 2001. Genomic imprinting disrupted by a maternal effect mutation in the Dnmt1 gene. Cell 104: 829–838. Hu G, Kim J, Xu Q, Leng Y, Orkin SH, Elledge SJ. 2009. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev 23: 837–848. Huang C, Martin S, Pfleger C, Du J, Buckner JH, Bluestone JA, Riley JL, Ziegler SF. 2013. Cutting Edge: A novel, human- specific interacting protein couples FOXP3 to a chromatin- remodeling complex that contains KAP1/TRIM28. J Immu- nol 190: 4470–4473. Iqbal K, Jin S-G, Pfeifer GP, Szabo´ PE. 2011. Reprogramming of the paternal genome upon fertilization involves genome- wide oxidation of 5-methylcytosine. Proc Natl Acad Sci 108: 3642–3647. Iyengar S, Farnham PJ. 2011. KAP1 protein: An enigmatic master regulator of the genome. J Biol Chem 286: 26267– 26276. Jacobs FMJ, Greenberg D, Nguyen N, Haeussler M, Ewing AD, Katzman S, Paten B, Salama SR, Haussler D. 2014. An evolutionary arms race between KRAB zinc-finger genes ZNF91/93 and SVA/L1 retrotransposons. Nature 516: 242– 245. Jin VX, O’Geen H, Iyengar S, Green R, Farnham PJ. 2007. Identification of an OCT4 and SRY regulatory module using integrated computational and experimental genomics ap- proaches. Genome Res 17: 807–817. Kamitani S, Ohbayashi N, Ikeda O, Togi S, Muromoto R, Sekine Y, Ohta K, Ishiyama H, Matsuda T. 2008. KAP1 regulates type I interferon/STAT1-mediated IRF-1 gene expression. Biochem Biophys Res Commun 370: 366–370. Kamitani S, Togi S, Ikeda O, Nakasuji M, Sakauchi A, Sekine Y, Muromoto R, Oritani K, Matsuda T. 2011. Kru¨ppel-associat- ed box-associated protein 1 negatively regulates TNF-a- induced NF-kB transcriptional activity by influencing the interactions among STAT3, p300, and NF-kB/p65. J Immu- nol 187: 2476–2483. Kigami D, Minami N, Takayama H, Imai H. 2003. MuERV-L is one of the earliest transcribed genes in mouse one-cell embryos. Biol Reprod 68: 651–654. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Bald- win J, Devon K, Dewar K, Doyle M, FitzHugh W, et al. 2001. Initial sequencing and analysis of the human genome. Nature 409: 860–921. Li Z, Wang D, Na X, Schoen SR, Messing EM, Wu G. 2003. The VHL protein recruits a novel KRAB-A domain protein to repress HIF-1a transcriptional activity. EMBO J 22: 1857–1867. Li X, Ito M, Zhou F, Youngson N, Zuo X, Leder P, Ferguson- Smith AC. 2008. A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints. Dev Cell 15: 547–557. Liang Q, Deng H, Li X, Wu X, Tang Q, Chang T-H, Peng H, Rauscher FJ, Ozato K, Zhu F. 2011. Tripartite motif-contain- EPIGENETIC REPROGRAMMING 161 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 8. ing protein 28 is a small ubiquitin-related modifier E3 ligase and negative regulator of IFN regulatory factor 7. J Immunol 187: 4754–4763. Liu Y, Toh H, Sasaki H, Zhang X, Cheng X. 2012. An atomic model of Zfp57 recognition of CpG methylation within a specific DNA sequence. Genes Dev 26: 2374–2379. Lorthongpanich C, Cheow LF, Balu S, Quake SR, Knowles BB, Burkholder WF, Solter D, Messerschmidt DM. 2013. Single- cell DNA-methylation analysis reveals epigenetic chimerism in preimplantation embryos. Science 341: 1110–1112. Macfarlan TS, Gifford WD, Driscoll S, Lettieri K, Rowe HM, Bonanomi D, Firth A, Singer O, Trono D, Pfaff SL. 2012. Embryonic stem cell potency fluctuates with endogenous ret- rovirus activity. Nature 487: 57–63. Magnu´sdo´ttir E, Surani MA. 2014. How to make a primordial germ cell. Development 141: 245–252. Maruyama A, Nishikawa K, Kawatani Y, Mimura J, Hosoya T, Harada N, Yamamato M, Itoh K. 2011. The novel Nrf2-inter- acting factor KAP1 regulates susceptibility to oxidative stress by promoting the Nrf2-mediated cytoprotective response. Biochem J 436: 387–397. Matsui T, Leung D, Miyashita H, Maksakova IA, Miyachi H, Kimura H, Tachibana M, Lorincz MC, Shinkai Y. 2010. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464: 927–931. McGrath J, Solter D. 1984. Completion of mouse embryogenesis requires both the maternal and paternal genomes. Cell 37: 179–183. Messerschmidt DM. 2012. Should I stay or should I go: Protec- tion and maintenance of DNA methylation at imprinted genes. Epigenetics 7: 969–975. Messerschmidt DM, de Vries W, Ito M, Solter D, Ferguson- Smith A, Knowles BB. 2012. Trim28 is required for epige- netic stability during mouse oocyte to embryo transition. Sci- ence 335: 1499–1502. Messerschmidt DM, Knowles BB, Solter D. 2014. DNA meth- ylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev 28: 812–828. Mouse Genome Sequencing Consortium, Waterston RH, Lind- blad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agar- wala R, Ainscough R, Alexandersson M, et al. 2002. Initial sequencing and comparative analysis of the mouse genome. Nature 420: 520–562. Nielsen AL, Ortiz JA, You J, Oulad-Abdelghani M, Khechu- mian R, Gansmuller A, Chambon P, Losson R. 1999. Inter- action with members of the heterochromatin protein 1 (HP1) family and histone deacetylation are differentially involved in transcriptional silencing by members of the TIF1 family. EMBO J 18: 6385–6395. Nishitsuji H, Abe M, Sawada R, Takaku H. 2012. ZBRK1 re- presses HIV-1 LTR-mediated transcription. FEBS Lett 586: 3562–3568. Nishitsuji H, Sawada L, Sugiyama R, Takaku H. 2015. ZNF10 inhibits HIV-1 LTR activity through interaction with NF-kB and Sp1 binding motifs. FEBS Lett 589: 2019–2025. Okano M, Bell DW, Haber DA, Li E. 1999. DNA methyltrans- ferases Dnmt3a and Dnmt3b are essential for de novo meth- ylation and mammalian development. Cell 99: 247–257. Okumura K, Sakaguchi G, Naito K, Tamura T, Igarashi H. 1997. HUB1, a novel Kru¨ppel type zinc finger protein, represses the human T cell leukemia virus type I long terminal repeat-me- diated expression. Nucleic Acids Res 25: 5025–5032. Peaston AE, Evsikov AV, Graber JH, de Vries WN, Holbrook AE, Solter D, Knowles BB. 2004. Retrotransposons regulate host genes in mouse oocytes and preimplantation embryos. Dev Cell 7: 597–606. Peng H, Ivanov AV, Oh HJ, Lau YFC, Rauscher FJ. 2009. Epi- genetic gene silencing by the SRY protein is mediated by a KRAB-O protein that recruits the KAP1 co-repressor machin- ery. J Biol Chem 284: 35670–35680. Peters J. 2014. The role of genomic imprinting in biology and disease: An expanding view. Nat Rev Genet 15: 517–530. Quenneville S, Verde G, Corsinotti A, Kapopoulou A, Jakobs- son J, Offner S, Baglivo I, Pedone PV, Grimaldi G, Riccio A, et al. 2011. In embryonic stem cells, ZFP57/KAP1 recognize a methylated hexanucleotide to affect chromatin and DNA methylation of imprinting control regions. Mol Cell 44: 361–372. Ratnam S, Mertineit C, Ding F, Howell CY, Clarke HJ, Bestor TH, Chaillet JR, Trasler JM. 2002. Dynamics of Dnmt1 meth- yltransferase expression and intracellular localization during oogenesis and preimplantation development. Dev Biol 245: 304–314. Reik W. 2007. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447: 425– 432. Rowe HM, Jakobsson J, Mesnard D, Rougemont J, Reynard S, Aktas T, Maillard PV, Layard-Liesching H, Verp S, Marquis J, et al. 2010. KAP1 controls endogenous retroviruses in em- bryonic stem cells. Nature 463: 237–240. Ryan RF, Schultz DC, Ayyanathan K, Singh PB, Friedman JR, Fredericks WJ, Rauscher FJ. 1999. KAP-1 corepressor pro- tein interacts and colocalizes with heterochromatic and euchromatic HP1 proteins: A potential role for Kru¨ppel-asso- ciated box-zinc finger proteins in heterochromatin-mediated gene silencing. Mol Cell Biol 19: 4366–4378. Santos F, Dean W. 2004. Epigenetic reprogramming during ear- ly development in mammals. Reproduction 127: 643–651. Satou A. 2001. A novel transrepression pathway of c-Myc. Re- cruitment of a transcriptional corepressor complex to c-Myc by MM-1, a c-Myc-binding protein. J Biol Chem 276: 46562–46567. Satou A, Hagio Y, Taira T, Iguchi-Ariga SMM, Ariga H. 2004. Repression of the c-fms gene in fibroblast cells by c-Myc- MM-1-TIF1b complex. FEBS Lett 572: 211–215. Schultz DC, Friedman JR, Rauscher FJ. 2001. Targeting histone deacetylase complexes via KRAB-zinc finger proteins: The PHD and bromodomains of KAP-1 form a cooperative unit that recruits a novel isoform of the Mi-2a subunit of NuRD. Genes Dev 15: 428–443. Schultz DC, Ayyanathan K, Negorev D, Maul GG, Rauscher FJ. 2002. SETDB1: A novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediat- ed silencing of euchromatic genes by KRAB zinc-finger pro- teins. Genes Dev 16: 919–932. Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, Popp C, Thienpont B, Dean W, Reik W. 2012. The dynam- ics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol Cell 48: 849–862. Seki Y, Kurisaki A, Watanabe-Susaki K, Nakajima Y, Nakanishi M, Arai Y, Shiota K, Sugino H, Asashima M. 2010. TIF1b regulates the pluripotency of embryonic stem cells in a phos- phorylation-dependent manner. Proc Natl Acad Sci 107: 10926–10931. Surani MAH, Barton SC, Norris ML. 1984. Development of reconstituted mouse eggs suggests imprinting of the genome during gametogenesis. Nature 308: 548–550. Takahashi K, Sugi Y, Hosono A, Kaminogawa S. 2009. Epige- netic regulation of TLR4 gene expression in intestinal epithe- lial cells for the maintenance of intestinal homeostasis. J Immunol 183: 6522–6529. Tan X, Xu X, Elkenani M, Smorag L, Zechner U, Nolte J, Engel W, Pantakani DVK. 2013. Zfp819, a novel KRAB-zinc finger protein, interacts with KAP1 and functions in genomic integ- rity maintenance of mouse embryonic stem cells. Stem Cell Res 11: 1045–1059. Thomas JH, Schneider S. 2011. Coevolution of retroelements and tandem zinc finger genes. Genome Res 21: 1800–1812. Tsuruma R, Ohbayashi N, Kamitani S, Ikeda O, Sato N, Mur- omoto R, Sekine Y, Oritani K, Matsuda T. 2007. Physical and functional interactions between STAT3 and KAP1. Oncogene 27: 3054–3059. Walsh CP, Chaillet JR, Bestor TH. 1998. Transcription of IAP endogenous retroviruses is constrained by cytosine methyla- tion. Nat Genet 20: 116–117. LESEVA ET AL.162 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 9. Wang C, Ivanov A, Chen L, Fredericks WJ, Seto E, Rauscher FJ, Chen J. 2005. MDM2 interaction with nuclear corepressor KAP1 contributes to p53 inactivation. EMBO J 24: 3279– 3290. Wang C, Rauscher FJ, Cress WD, Chen J. 2007. Regulation of E2F1 function by the nuclear corepressor KAP1. J Biol Chem 282: 29902–29909. Wiznerowicz M, Jakobsson J, Szulc J, Liao S, Quazzola A, Beermann F, Aebischer P, Trono D. 2007. The Kru¨ppel-as- sociated box repressor domain can trigger de novo promoter methylation during mouse early embryogenesis. J Biol Chem 282: 34535–34541. Wolf D, Goff SP. 2009. Embryonic stem cells use ZFP809 to silence retroviral DNAs. Nature 458: 1201–1204. Wolf G, Greenberg D, Macfarlan TS. 2015a. Spotting the enemy within: Targeted silencing of foreign DNA in mammalian genomes by the Kru¨ppel-associated box zinc finger protein family. Mobile DNA 6: 17. Wolf G, Yang P, Fu¨chtbauer AC, Fu¨chtbauer E-M, Silva AM, Park C, Wu W, Nielsen AL, Pedersen FS, Macfarlan TS. 2015b. The KRAB zinc finger protein ZFP809 is required to initiate epigenetic silencing of endogenous retroviruses. Genes Dev 29: 538–554. Wossidlo M, Nakamura T, Lepikhov K, Marques CJ, Zakhartch- enko V, Boiani M, Arand J, Nakano T, Reik W, Walter J. 2011. 5-Hydroxymethylcytosine in the mammalian zygote is linked with epigenetic reprogramming. Nat Commun 2: 241. Yearim A, Gelfman S, Shayevitch R, Melcer S, Glaich O, Mallm J-P, Nissim-Rafinia M, Cohen A-HS, Rippe K, Meshorer E, et al. 2015. HP1 is involved in regulating the global impact of DNA methylation on alternative splicing. Cell Rep 10: 1122–1134. Zheng L, Pan H, Li S, Flesken-Nikitin A, Chen PL, Boyer TG, Lee WH. 2000. Sequence-specific transcriptional corepressor function for BRCA1 through a novel zinc finger protein, ZBRK1. Mol Cell 6: 757–768. Zuo X, Sheng J, Lau H-T, McDonald CM, Andrade M, Cullen DE, Bell FT, Iacovino M, Kyba M, Xu G, et al. 2012. Zinc finger protein ZFP57 requires its co-factor to recruit DNA methyltransferases and maintains DNA methylation imprint in embryonic stem cells via its transcriptional repression domain. J Biol Chem 287: 2107–2118. EPIGENETIC REPROGRAMMING 163 Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from
  • 10. 10.1101/sqb.2015.80.027441Access the most recent version at doi: 2016 2015 80: 155-163 originally published online January 13,Cold Spring Harb Symp Quant Biol Milena Leseva, Barbara B. Knowles, Daniel M. Messerschmidt, et al. Mammalian Epigenome Establish: Natural Reprogramming of the−Maintain−Erase References http://symposium.cshlp.org/content/80/155.full.html#ref-list-1 This article cites 74 articles, 38 of which can be accessed free at: License Commons Creative Service Email Alerting click here.the box at the top right corner of the article or Receive free email alerts when new articles cite this article - sign up in http://symposium.cshlp.org/subscriptions go to:Cold Spring Harbor Symposia on Quantitative BiologyTo subscribe to Copyright © 2015 Cold Spring Harbor Laboratory Press; all rights reserved Cold Spring Harbor Laboratory Presson October 4, 2016 - Published bysymposium.cshlp.orgDownloaded from