SlideShare uma empresa Scribd logo
1 de 11
Baixar para ler offline
Review




Parallel mechanisms of epigenetic
reprogramming in the germline
Jamie A. Hackett, Jan J. Zylicz and M. Azim Surani
Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Physiology, Development and Neuroscience,
University of Cambridge, Cambridge, CB2 1QN, UK



Germ cells possess the extraordinary and unique capac-                                       [5,6]. This process is initiated in response to localised sig-
ity to give rise to a new organism and create an enduring                                    nals, including BMP4 and WNT3, which direct activation of
link between all generations. To acquire this property,                                      the key transcriptional regulators B-lymphocyte-induced
primordial germ cells (PGCs) transit through an unprec-                                      maturation protein 1 (Blimp1) and PR domain containing
edented programme of sequential epigenetic events that                                       14 (Prdm14) in competent epiblast cells [1,7–9]. Lineage-
culminates in an epigenomic basal state that is the                                          restricted PGCs then embark on an orchestrated sequence
foundation of totipotency. This process is underpinned                                       of reprogramming that culminates in a basal epigenetic
by genome-wide DNA demethylation, which may occur                                            state. The number of early PGCs is highly restricted (ap-
through several overlapping pathways, including con-                                         proximately 40 by E7.25) so it is crucial that the complex
version to 5-hydroxymethylcytosine. We propose that                                          series of epigenetic events be robust to ensure that most, if
the epigenetic programme in PGCs operates through                                            not all, cells efficiently transit through the process [10].
multiple parallel mechanisms to ensure robustness at                                         Reprogramming must also proceed rapidly because of strict
the level of individual cells while also being flexible
through functional redundancy to guarantee high fideli-                                         Glossary
ty of the process. Gaining a better understanding of the
                                                                                               5-hydroxymethylcytosine (5hmC): oxidation of methylated cytosines (5mC) by
molecular mechanisms that direct epigenetic repro-                                             TET proteins generates 5hmC, which may be an intermediate during DNA
gramming in PGCs will enhance our ability to manipu-                                           demethylation and can be further converted to 5caC and 5fC. 5hmC is enriched
late epigenetic memory, cell-fate decisions and                                                in pluripotent and some neuronal cell types, but its precise functional
                                                                                               consequences in the genome and its role in DNA demethylation are unclear.
applications in regenerative medicine.                                                         Base excision repair (BER): a cellular mechanism for repair of nonhelix-
                                                                                               distorting base mutations or lesions in the genome. BER is initiated by a DNA
Reprogramming PGCs towards totipotency                                                         glycosylase (e.g. TDG) that removes inappropriate bases and forms an
Development from the zygote to adulthood is characterised                                      apurinic/apyrimidinic (AP) site, which is then cleaved by an AP endonuclease
                                                                                               and repaired by specific lyases and polymerases. BER may function to remove
by a progressive restriction of cellular potential that gives                                  downstream derivatives of 5mC, such as 5caC, and mediate repair to
rise to all the differentiated somatic cell types. A unique                                    unmodified C [60].
                                                                                               Basal epigenetic state: the unique epigenetic state of PGCs following
exception to this unidirectional process occurs in the germ-                                   reprogramming. By E13.5, the PGC epigenome has undergone extensive
line, where an unprecedented reprogramming event in                                            reorganisation of histone modifications and is stripped of genome-wide DNA
PGCs (see Glossary) reverses epigenetic barriers to plas-                                      methylation, rendering it at its most basal level during the mammalian life
                                                                                               cycle.
ticity and resets genomic potential. Reprogramming in                                          Bisulfite sequencing: a technique used to determine the pattern of allelic DNA
PGCs results in chromatin remodelling, erasure of genomic                                      methylation (5mC) at specific genomic regions. Bisulfite sequencing cannot
imprints and extensive DNA demethylation [1]. This pro-                                        distinguish 5mC from 5hmC [33]. Additionally, 5caC is indistinguishable from
                                                                                               unmodified C by bisulfite sequencing [60].
cess represents the most comprehensive erasure of epige-                                       DNA demethylation: the removal of a methyl group from position 5 of a
netic information in the mammalian life cycle and                                              cytosine base (5mC), which usually resides within a CpG genomic context, to
underpins the totipotent state. Therefore, unravelling                                         generate an unmodified C. DNA demethylation may occur through either a
                                                                                               ‘passive’ mechanism that relies on replication-dependent dilution or an ‘active’
the mechanisms that drive reprogramming, particularly                                          process driven by enzymatic replacement independently of DNA replication. As
DNA demethylation, in the unique context of PGCs is of                                         DNA methylation is associated with transcriptional silencing, DNA demethyla-
great interest.                                                                                tion can generate a transcriptionally competent state.
                                                                                               Epigenetic reprogramming: genome-wide reorganisation of epigenetic mod-
   In mice, PGCs are specified from a subset of posterior                                       ifications that overcomes stable epigenetic barriers and enables acquisition of
proximal epiblast cells at approximately embryonic day (E)                                     genomic potential. During the mammalian life cycle, epigenetic reprogram-
                                                                                               ming occurs in PGCs and in early zygotic development.
6.25, resulting in the establishment of a founder population
                                                                                               Genomic imprints: genomic sequences that exhibit differences in CpG methyla-
of PGCs at E7.25 [1,2]. These nascent PGCs subsequently                                        tion according to the parent of origin. These differentially methylated regions
migrate to the genital ridges by approximately E10.5 and,                                      (DMRs) can influence the allele-specific expression of one or more genes.
                                                                                               Primordial germ cell (PGC): the precursors of mature germ cells that are
from E12.5 onwards, they undergo sex-specific development                                       specified during post-implantation development. In vivo, PGCs are restricted
in the gonads [3,4]. Because mammalian PGCs are specified                                       as a unipotent lineage and only give rise to gametes, which generate the
from cells that are already primed towards a somatic fate,                                     totipotent state upon fertilisation. Early PGCs also possess an underlying
                                                                                               genomic plasticity, as evidenced through their capacity to form pluripotent EG
nascent PGCs must both repress the ongoing somatic pro-                                        cells upon in vitro culture.
gramme and activate the germ cell transcriptional network                                      Totipotency: the ability of a cell to give rise to all the cell types of the
                                                                                               embryonic and extra-embryonic lineages. By contrast, pluripotency refers to
                                                                                               the capacity of a cell to generate all the cell types of the embryo.
      Corresponding author: Surani, M.A. (a.surani@gurdon.cam.ac.uk).

164                                0168-9525/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. doi:10.1016/j.tig.2012.01.005 Trends in Genetics, April 2012, Vol. 28, No. 4
Review                                                                              Trends in Genetics April 2012, Vol. 28, No. 4


temporal constraints imposed by the entry of male germ         sperm or oocytes in vivo, during migration (E8.5–E11.5),
cells into mitotic arrest and female germ cells into meiotic   PGCs show multiple transcriptional and epigenetic simi-
arrest at approximately E13.5. To overcome these con-          larities to pluripotent ES cells. Indeed, E8.5–E11.5 PGCs
straints, epigenetic reprogramming in PGCs is probably         can form pluripotent embryonic germ (EG) cells in vitro,
directed by multiple parallel mechanisms that ensure the       which resemble ES cells rather than the post-implantation
fidelity, flexibility and efficiency of this fundamental pro-     epiblast cells from which PGCs were originally specified
cess. As such, events like genome-wide erasure of DNA          [19,20]. It is possible that this epigenetic state of PGCs is
methylation in PGCs may occur through several intercon-        an underlying requirement for the initiation of meiosis
nected mechanisms, including both active and passive path-     and the eventual acquisition of totipotency in the zygote.
ways, that collectively confer redundancy and hence            Interestingly, migrating PGCs additionally exhibit upre-
robustness to PGC reprogramming and development.               gulation of histone H2A/H4 arginine 3 symmetrical meth-
   Here, we discuss the epigenetic events in PGCs and          ylation (H2A/H4R3me2 s), which is catalysed by protein
the mechanisms that may operate to drive epigenetic            arginine methyltransferase 5 (PRMT5) [21]. This modifi-
reprogramming. We suggest that an integrated process           cation may contribute to maintaining PGCs in a unipotent
involving parallel systems is at play and consider the         state in vivo and to repression of the somatic programme
potential pathways of DNA demethylation. We also high-         [22].
light some of the potential roles and developmental               Changes in histone modifications occur in parallel with
processes that epigenetic reprogramming contributes to         a reported reduction in global levels of DNA methylation
in PGCs.                                                       (5mC) in migrating PGCs from approximately E8.0 [11].
                                                               Any loss of 5mC might reflect the effects of BLIMP1 and
Epigenetic events in PGCs                                      PRDM14, which repress both DNA (cytosine-5)-methyl-
At the point at which PGCs are specified from post-im-          transferase 3a and 3b (Dnmt3a and Dnmt3b) and ubiqui-
plantation epiblast cells, they are epigenetically indistin-   tin-like, containing PHD and RING finger domains 1
guishable at the global level from their neighbours, which     (Uhrf1), which are essential components of the de novo
are destined for a somatic fate [9,11]. Therefore, nascent     and maintenance methylation machinery, respectively
PGCs inherit stable epigenetic states, including DNA           (Figure 1) [7,8,23–26]. Additionally, repression of GLP
methylation and X-inactivation, which constitute an epi-       may directly affect DNA methylation through a parallel
genetic barrier against the eventual acquisition of totipo-    mechanism that is both dependent and independent of
tency [1,12]. It is thus an important early step in PGC        H3K9me2 [27,28]. However, most analysed genomic
development to initiate a process of reprogramming that        regions, including transposable elements, imprinted loci
erases these stable epigenetic blocks. The first gross epi-     and single-copy genes, apparently retain DNA methylation
genetic changes in PGCs entail a reciprocal loss of histone    at CpG sites in PGCs until at least E10.5 [29–32], although
H3 lysine 9 dimethylation (H3K9me2) from E7.75 and, in         this does not exclude the possibility that there is conver-
most PGCs, a global increase of H3 lysine 27 trimethyla-       sion of 5mC to 5-hydroxymethylcytosine (5hmC) [33].
tion (H3K27me3) by E9.5 (Figure 1) [11,13,14]. The ge-         Therefore, it is unclear to what extent DNA demethylation
nome-wide depletion of H3K9me2 is potentially a                contributes to the early stages of epigenetic reprogram-
consequence of the downregulation of GLP, a methyltrans-       ming (from E8.0) in PGCs and whether conversion to 5hmC
ferase that forms a heteromeric complex with G9a (also         plays a role. Conversely, it is unclear how 5mC (or 5hmC) is
known as EHMT2) that is required for deposition of H3K9        maintained at analysed genomic regions in migrating
mono- and dimethylation, and the parallel upregulation of      PGCs given that essential components of the de novo
specific lysine demethylases ([15] and unpublished obser-       and maintenance machinery, particularly UHRF1, are
vations). By contrast, the mechanisms responsible for the      absent [7]. One possibility is that UHRF2, which is con-
increased H3K27me3 levels in PGCs remain unclear, al-          served with UHRF1 at the sequence level and preferen-
though notably enhancer of zeste homologue 1 (Ezh1),           tially binds hemi-methylated DNA associated with
which has H3K27me3 methyltransferase activity, is upre-        H3K9me3, can compensate for UHRF1 to maintain DNA
gulated in PGCs [16]. Because H3K9me2 and H3K27me3             methylation in PGCs, either globally or at specific loci [34].
marks are both associated with transcriptional repression,     In support of this, UHRF2 is specifically upregulated
it has been postulated that the loss of H3K9me2 is com-        during PGC specification, at least in vitro [7,35]. Addition-
plemented by the gain of H3K27me3 to maintain a repres-        ally, unlike H3K9me2, global H3K9me3 levels are main-
sive chromatin state in PGCs [17]. However, the precise        tained in migrating PGCs [14]. The in vitro PGC-like cells
genomic location and relationship between these epigenet-      (PGCLC) generated in an elegant recent study may enable
ic changes remains to be determined. Nonetheless, the          further mechanistic insights into epigenetic events in na-
global enrichment of H3K27me3 and loss of H3K9me2              scent PGCs at a higher resolution [36].
establishes a chromatin environment in PGCs that is               The early stages of epigenomic reorganisation in PGCs
grossly similar to that in pluripotent embryonic stem          are followed by, and are probably a prerequisite for, the
(ES) cells and is coupled to upregulation of pluripotency      dramatic genome-wide erasure of DNA methylation and
genes, such as Nanog and SRY Sox2 [1]. Additionally,           extensive chromatin remodelling subsequent to entry into
unlike global H3K27me3 levels, the inactive X-chromo-          the genital ridges at approximately E10.5. Because gross
some (Xi) exhibits a protracted decline in H3K27me3 in         DNA demethylation seems to occur rapidly at a distinct
female PGCs, which is linked to initiation of X-reactivation   time point, while most PGCs are in G2 phase, it is held that
[18]. Thus, while PGCs are unipotent and only form either      this process is an ‘active’ event occurring independently
                                                                                                                             165
Review                                                                                                               Trends in Genetics April 2012, Vol. 28, No. 4



                                 (a) Chromatin dynamics
                                                                    5mC                          H3K27me3




                                      Relative levels




                                                                            H3K9
                                                                                me2
                                                            E5.5   E6.5          E8.0                     E10.5       E11.5

                                 (b) Specification and chromatin-modifying factors
                                                                                                 Blimp1
                                      Relative expression




                                                                                                 Prdm14
                                                                                           G9a
                                                                            Glp




                                                            E5.5   E6.5          E8.0                     E10.5       E11.5

                                 (c) DNA methylation factors

                                                                                                                  Uhrf2
                                      Relative expression




                                                                                                                  Dnmt1
                                                                                                                  Dnmt3a


                                                                   Dnmt3b                                         Uhrf1

                                                            E5.5   E6.5          E8.0                     E10.5       E11.5

                                 (d) Tet dioxygenases
                                      Relative expression




                                                                                                              Tet1

                                                                                                                     Tet2



                                                                                                                          Tet3

                                                            E5.5   E6.5          E8.0                     E10.5       E11.5
                                                                   Spec.              R1                                    R2

                                                                                                                            TRENDS in Genetics

Figure 1. Chromatin and transcriptional changes in primordial germ cells (PGCs). Following specification (Spec.), PGCs transit through two sequential phases of
reprogramming during migration (R1) and subsequent to entry into the genital ridge (R2). (a) Epigenomic reorganisation in PGCs. At R1, PGCs exhibit global erasure of
histone H3 lysine 9 dimethylation (H3K9me2), upregulation of H3 lysine 27 trimethylation (H3K27me3) and some loss of the DNA methylation signal. At R2, there is a further
dramatic loss of DNA methylation, which includes erasure of imprints. This correlates with a transient reorganisation of H3K27me3, loss of heterochromatic chromocentres
and remodelling of other chromatin modifications, including histone H2A/H4 arginine 3 symmetrical methylation (H2A/H4R3me2; not shown). (b–d) Transcriptional changes
during PGC development [13]. (b) Specification and chromatin-modifying factors. Upregulation of B-lymphocyte-induced maturation protein 1 (Blimp1) and PR domain
containing 14 (Prdm14) is necessary for specification of PGC fate, whereas the parallel downregulation of glucagon-like peptide (Glp)/G9a and upregulation of lysine
demethylases (not shown) contribute to erasure of H3K9me2 at R1. (c) DNA methylation proteins. Downregulation of DNA (cytosine-5)-methyltransferase 3b (Dnmt3b) and
ubiquitin-like, containing PHD and RING finger domains 1 (Uhrf1) may account for DNA demethylation at R1. UHRF2 may partially compensate for the absence of UHRF1 to
maintain DNA methylation until R2. (d) Ten-eleven translocation gene Tet dioxygenases. Tet1 and Tet2 are expressed in PGCs by R2, whereas Tet3 cannot be detected [50].
Dashed line indicates putative levels. Abbreviation: E, embryonic day.


from DNA replication, although the precise mechanism is                                      [29,32]. The process of DNA methylation erasure also
yet to be elucidated [14]. Reprogramming in PGCs at this                                     initiates a cascade of chromatin remodelling in PGCs at
stage results in almost full erasure of DNA methylation by                                   approximately E11.5. This includes a transient reorgani-
E13.5 [37], with complete stripping of parental imprints                                     sation of linker histone H1, H3K27me3 and H3K9me3 and
and promoter CpG methylation at germline-specific genes                                       stable remodelling of global H3K9ac and H2A/H4R3me2 s
166
Review                                                                              Trends in Genetics April 2012, Vol. 28, No. 4


[14]. Some sequences, including intracisternal A particle       replace 5mC during global DNA demethylation in PGCs
(IAP) retrotransposons, partially evade DNA demethyla-          [50].
tion in PGCs, although the mechanism that protects these           Mechanistically, conversion of 5mC to 5hmC could lead
elements remains to be determined [31]. Nevertheless, the       to unmodified cytosine through several routes in PGCs,
erasure of DNA methylation and extensive chromatin              including through providing a substrate for base excision
reorganisation at approximately E11.5 renders PGCs in           repair (BER)-mediated active demethylation [40]. Several
a basal epigenetic state that represents an epigenomic          recent studies have linked BER components to demethyl-
nadir during mammalian development. It is of note that          ation during zygotic reprogramming and at specific loci in
the apparent biphasic nature of demethylation in PGCs (at       somatic contexts [50–52]. As BER components are also
approximately E8.0 and approximately E11.5) may poten-          upregulated in PGCs relative to somatic neighbours during
tially represent a continuous process of 5mC erasure. In        epigenetic erasure, genome-wide demethylation in PGCs
any case, reprogramming of the PGC genome as a whole is         may also occur, at least partially through BER. Indeed, the
distinguished from reprogramming during zygotic devel-          presence of chromatin-associated XRCC1 and active poly[-
opment, where imprints, DNA methylation at multiple loci        ADP-ribose] polymerase 1 (PARP1) in PGCs at approxi-
and polycomb-based chromatin modifications remain; this          mately E11.5, which signify single-strand DNA breaks
implies that reprogramming in PGCs represents a more            associated with BER, further support this possibility [50].
extensive process [32]. Although both reprogramming                One potential involvement of BER-mediated DNA de-
events probably share some mechanistic similarities, it         methylation in PGCs is that TET-generated 5hmC is further
is also likely that both employ unique systems to achieve       processed by deamination to 5-hydroxymethyluridine
distinct levels of epigenomic resetting.                        (5hmU) by the activation-induced cytidine deaminase/apo-
                                                                lipoprotein B mRNA-editing, enzyme-catalytic, polypeptide
Erasure of DNA methylation and cellular identity                (AID/APOBEC) family of deaminases, and subsequently
DNA methylation (5mC) within a CpG context is a highly          excised by a glycosylase and repaired to unmodified C.
heritable epigenetic mark that is associated with tran-         Consistent with this, loss of AID impairs global demethyla-
scriptional repression and that contributes to stable line-     tion in PGCs by E13.5, indicating that 5mC erasure in PGCs
age commitment [38–41]. In this respect, global erasure of      depends, at least in part, on AID [37]. Indeed, AID also
DNA methylation during PGC development is a fundamen-           enhances locus-specific active demethylation mediated by
tal event towards the acquisition of totipotency. The ca-       TET1 in somatic cells and has been reported to interact with
pacity of global DNA demethylation to alter cellular            thymine DNA glycosylase (TDG), which can excise the
identity, for example in the course of derivation of induced    deamination product 5hmU [52,53]. Moreover, AID has
pluripotent stem cells (iPS) from somatic cells, makes          been proposed to be required for DNA demethylation of
unravelling the mechanisms that mediate this process of         octamer-binding transcription factor 4 (Oct4) and Nanog
great importance [42,43]. Although extensive studies have       during somatic cell reprogramming induced by heterokary-
established how and where 5mC is and can be introduced,         on formation with ES cells as well as in demethylation in
it remains enigmatic precisely how DNA methylation is           zebrafish embryos [54,55]. However, murine PGCs still
removed from the genome [44]. The global DNA demethyl-          undergo extensive demethylation by E13.5 in the absence
ation during PGC development represents a unique in vivo        of AID; global 5mC is reduced to 22% and 20% in male and
event in that it results in near-complete stripping of CpG      female mutant PGCs, respectively, as opposed to 16% and
methylation at almost all genomic loci, including imprints.     8% in wild-type PGCs and compared with approximately
The magnitude of DNA demethylation makes PGCs an                74% in E13.5 embryonic soma. Moreover, AID-mutant mice
unparalleled system to understand the process of 5mC            are both viable and fertile, as are APOBEC 1- and APOBEC
erasure in an in vivo context.                                  2/3-null mice [56–58]. It is also unclear whether AID is
                                                                expressed to any significant degree in PGCs at the time of
Active demethylation in PGCs                                    gross demethylation (approximately E11.5), as it has only
The recent identification of three 5mC-dioxygenases [ten-        been detected from E12.5 in PGCs, suggesting that AID has
eleven translocation gene 1, 2 and 3 (TET1, TET2 and            a role in PGC demethylation either after global 5mC erasure
TET3)], which can convert 5mC to 5-hydroxymethylcyto-           (approximately E12.5) or perhaps at an earlier stage (ap-
sine, presented a potential solution to the longstanding        proximately E8.5), when the expression of AID is unknown
debate regarding the mechanism of DNA demethylation             [50,59]. Likewise, TDG is not detectable in PGCs between
[45,46]. Conversion of 5mC to 5hmC enables several alter-       E10.5 and E13.5, as judged by immunofluorescence studies
native but partially overlapping routes to generate an          [50]. However, TDG-null PGCs accumulate biallelic CpG
unmodified cytosine (C) residue independently of, or de-         methylation by E11.0 at the insulin-like growth factor 2
pendent on, DNA replication (Figure 2). In the zygote, the      receptor (Igf2r)-imprinted differentially methylated region
rapid loss of 5mC from the male pronucleus correlates with      (DMR), suggesting that TDG has a role in maintaining
a concomitant gain in 5hmC, implying that 5mC is con-           a methylation-free state at this locus, presumably by de-
verted to 5hmC in this context and has a fundamental role       methylation, although this event may occur prior to PGC
in reprogramming [47,48]. Indeed, loss of TET3, which is        specification [52]. Although further clarification of TDG
the only 5mC-dioxigenase significantly expressed in              function and expression in PGCs is necessary, its putative
zygotes, led to a failure to erase 5mC and neonatal lethality   absence at E11.5 argues against a direct AID- and/or TDG-
[49]. Although TET3 is not detectable in PGCs, TET1 and         mediated active demethylation reaction in PGCs, as has
TET2 are present in these cells, suggesting that 5hmC may       been reported in other contexts [52]. Therefore, alternative
                                                                                                                             167
Review                                                                                                                        Trends in Genetics April 2012, Vol. 28, No. 4



                                  (a)




                                                                                        C
                                                                                    OBE           5mC
                                                                              /  AP
                                                                             D
                                                                          AI                            TET1/2




                                                                                   C
                                                                     T           BE              5hmC
                                                                              APO
                                                                            D/                      TET1/2
                                                                          AI




                                                                   5hmU                            5fC
                                                                                                    TET1/2




                                                                                                  5caC
                                                                                                    ?
                                                                                  BE




                                                                                                                  ive
                                                                                    R




                                                                                                                 ss
                                                                                                             Pa




                                  (b)                                                              C
                                                            5mC
                                    Relative level




                                                                                                                              Pa
                                                                                                                                   ss
                                                                                                                                        ive
                                                                                                                      Ac
                                                                                                                         ti
                                                                                                                        ve




                                                            5hmC
                                                                                        No conversion

                                                     E5.5     E6.5          E8.0                          E10.5         E11.5
                                                              Spec                R1                                     R2

                                                                                                                               TRENDS in Genetics

Figure 2. Multiple parallel mechanisms of DNA demethylation. (a) Methylated cytosine (5mC) can be demethylated through several overlapping pathways, including
passive and active mechanisms, which may occur in parallel. Passive demethylation (right) can occur through direct replication-dependent depletion of 5mC owing to an
absence or reduction of DNA methyltransferase activity. Alternatively, TET oncogene (TET) proteins can catalyse oxidation of 5mC to 5-hydroxymethylcytosine (5hmC) or
further conversion to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), which may all lead to passive demethylation. Active erasure of 5mC (left) can occur through
deamination of either 5mC or 5hmC to thymidine (T) or 5-hydroxymethyluridine (5hmU), respectively, which can act as a substrate for base excision repair (BER) to
unmodified C. Further conversion of 5hmC to 5fC or 5caC may also be actively removed by BER or 5caC can putatively be removed via a direct decarboxylation reaction
(centre). (b) Putative temporal pattern of 5mC conversion to 5hmC during primordial germ cell (PGC) development. The depletion of the 5mC signal at R1 may occur as a
result of conversion to 5hmC or through other mechanisms. Similarly, 5mC may be converted to 5hmC at R2, which may subsequently be removed through passive and/or
active mechanisms. Abbreviations: AID/APOBEC, activation-induced cytidine deaminase/apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide; R1 and R2, the two
sequential phases of reprogramming during migration (R1) and subsequent to entry into the genital ridge (R2); Spec. specification.




168
Review                                                                              Trends in Genetics April 2012, Vol. 28, No. 4


deaminase–glycosylase complexes may operate down-               5hmU or further oxidation to 5fC and 5caC, all of which are
stream of 5hmC (or 5mC) in PGCs. Investigation of potential     BER substrates, ultimately leading to repair-driven de-
candidates, such as other members of the APOBEC family,         methylation.
and methyl-CpG-binding domain protein 4 (MBD4), single-
strand selective monofunctional uracil DNA glycosylase          Passive demethylation in PGCs
(SMUG1), nth endonuclease III-like 1 (NTH1) or nei endo-        There is accumulating evidence that genome-wide DNA
nuclease VIII-like 1 (NEIL1) glycosylases, may shed light on    demethylation events include at least a partial ‘passive’
the issue.                                                      component. Conversion of 5mC to 5hmC in the zygote has
   An alternative possibility is that TET-mediated hydro-       recently been shown to lead to replication-dependent pas-
xymethylation has a deamination-independent role in ac-         sive demethylation, rather than to active removal of 5hmC
tive demethylation in PGCs, as TET proteins can further         or its derivatives [68]. Thus, chromosomes originating in
oxidise 5hmC to 5-formylcytosine (5fC) and subsequently to      the paternal pronucleus retain 5hmC through successive
5-carboxylcytosine (5caC) [60,61]. Interestingly, these new     cleavage divisions, whereas newly synthesised sister chro-
C derivatives could also be targets for BER excision [60].      matids are devoid of 5mC and 5hmC, leading to a progres-
Indeed, 5fC and 5caC are substrates for TDG, and poten-         sive dilution of DNA modifications during development.
tially other glycosylases [60,62]. 5caC could also theoreti-    The passive loss of DNA modification is consistent with the
cally be ‘actively’ removed from the genome by a BER-           fact that the human maintenance DNA (cytosine-5)-
independent pathway that would involve decarboxylation          methyltransferase 1 (DNMT1) cannot recognise 5hmC
by an as yet unknown enzyme. It will be important to            and there are no other known 5hmC maintenance mecha-
determine the prevalence of 5fC and 5caC in PGCs during         nisms (although notably UHRF1 does efficiently recognise
the key stages of reprogramming to establish whether they       5hmC) [69,70]. Nevertheless, bisulfite sequencing has in-
contribute to demethylation through any mechanism. A            dicated there may be an additional active mechanism in
further possibility is that 5hmC is directly targeted for       zygotes that is dependent on BER and operates prior to
BER-mediated excision by 5hmC-specific glycosylases with-        DNA replication, which may target specific genomic land-
out the requirement for processing by deamination or fur-       marks [51]. Additionally, the high sensitivity of the 5hmC
ther oxidation. Indeed, 5hmC glycosylase activity has been      antibody may mask partial or locus-specific active erasure
reported in calf thymus extract [63]. Taken together, a         of 5hmC on the paternally derived pronuclear chromatids
mechanism based on BER may play at least a partial role         [71]. However, it seems probable that the bulk of paternal
in PGC demethylation, although it is currently unclear          pronuclear demethylation occurs passively following
which protein complexes direct the process or whether there     5hmC conversion. It remains unclear how the maternal
is a degree of redundancy. Likewise, it is not clear which      pronucleus, which does not undergo 5mC to 5hmC conver-
5hmC or 5mC derivative substrate [5hmC, 5hmU, 5fC, 5caC         sion, undergoes concomitant passive demethylation, as
or thymidine (T)] might be targeted by the BER machinery        DNMT1 is present and sufficient to maintain maternal
for active DNA demethylation in PGCs.                           imprints [68]. Nonetheless, these studies indicate that
   It is also possible that alternative mechanisms, indepen-    conversion of 5mC to 5hmC coupled with passive demeth-
dent of 5hmC, contribute to, or drive, active demethylation     ylation is feasible and could operate in PGCs.
in PGCs. In Arabidopsis thaliana, demethylation occurs via         In murine PGCs, although there is an overall loss of
direct excision of 5mC by the specific bifunctional DNA          DNA methylation over a relatively short period at approx-
glycosylase/lyases, DEMETER (DME) and REPRESSOR                 imately E11.5, it is important to reconsider whether era-
OF SILENCING 1 (ROS1), followed by recruitment of               sure of imprinted genes can occur over a protracted period
BER machinery [64,65]. Although no mammalian orthologs          of 2 days [30], which would not be compatible with a single
of DME–ROS1 or 5mC-specific glycosylases have been de-           genome-wide wave of ‘active’ demethylation model [14].
scribed, it remains a formal possibility that 5mC is directly   Indeed, erasure of DNA methylation from imprinted loci
excised from the genome in PGCs. Another possibility is that    and repeat elements in porcine PGCs occurs over an ex-
5mC is deaminated directly to T followed by excision by a       tended period of up to 20 days, which suggests that ‘active’
glycosylase that recognises the T–G mismatch, such as TDG       DNA demethylation does not apply in all mammalian
or MBD4, and repaired to unmodified cytosine by BER. It is       PGCs, possibly including those in mice [72]. Similarly to
additionally possible that a demethylation route based on       zygotes, conversion of 5mC to 5hmC (or 5fC and 5caC) in
radical sterile alpha motif (SAM) enzymes operates, al-         PGCs would facilitate passive replication-dependent de-
though experimental evidence for this is lacking in PGCs        methylation (Figure 2). Such a conversion of 5mC to 5hmC
[66]. Similarly, active 5mC erasure has been reported to be     would account for the dramatic loss of 5mC staining ob-
linked to the nucleotide excision repair (NER) pathway, and     served in PGCs at E11.5 and the protracted nature of
this could contribute to DNA demethylation during PGC           imprint erasure, given that bisulfite conversion would
migration (at approximately E8.5–E10.5) [67]. However, it       identify hydroxymethylated loci as methylated but 5mC
remains unclear whether such a mechanism could operate          antibodies would not [14,30,33]. Interestingly, conversion
in PGCs at approximately E11.5 owing to the absence of          of 5mC to 5hmC may also skew bisulfite sequencing
several key NER components between E10.5 and E12.5 [50].        results, as polymerases appear to amplify unmethylated
Thus, the consensus of available data points towards a          alleles preferentially over 5hmC-modified alleles after bi-
putative active DNA demethylation mechanism in PGCs             sulfite treatment, which may falsely imply demethylation
at approximately E11.5 being based on initial TET-mediat-       has occurred rather than conversion to 5hmC [73]. Addi-
ed hydroxylation of 5mC and either deamination towards          tionally, TET-mediated conversion to 5hmC may proceed
                                                                                                                             169
Review                                                                             Trends in Genetics April 2012, Vol. 28, No. 4


to 5caC in PGCs, which is read as unmethylated by bisul-       [49]. This suggests that rapid epigenetic reprogramming is
fite sequencing, erroneously indicating the occurrence of       involved in diminishing developmental barriers that some
active demethylation [60]. Although functional conse-          cells can nevertheless still overcome in a stochastic man-
quences of 5hmC, 5caC or other C derivatives are as yet        ner. Similarly, Tet1-null mice sire reduced litter sizes and
unclear, the inherent biases and lack of derivative speci-     mutant pups have a smaller body size at birth, potentially
ficity of bisulfite sequencing mean that observing an            owing to a delay in overcoming epigenetically imposed
unmethylated allele via this technique does not necessarily    barriers [77]. The presence of multiple means of 5mC
signify the presence of unmodified C or the implied active      erasure could compensate for the loss of function of Tet1
demethylation. As such, conversion to 5hmC or down-            through the use of alternative routes in a stochastic man-
stream C derivatives could at least partially account for      ner. For example, given that key maintenance-methylation
the apparent active ‘replication-independent’ demethyla-       enzymes, including UHRF1, are absent in PGCs, it is
tion that has been reported in PGCs.                           possible that direct passive demethylation of 5mC can
   An alternative to passive erasure through 5hmC con-         partly offset the absence of any putative 5hmC-mediated
version is that PGCs undergo direct passive dilution of        mechanisms, a possibility that might also affect interpre-
5mC, based on an absence or exclusion of maintenance           tation of the demethylation that still occurs in AID-defi-
components, such as DNMT1 or UHRF1. As the doubling            cient PGCs by E13.5 [7,37].
time of PGCs at this stage is approximately 16 hr [13,74],        In addition to contributing to a robust system, parallel
direct replication-dependent dilution could account for a      mechanisms of 5mC erasure could function to target spe-
significant reduction in global 5mC in PGCs between E10.5       cific genomic loci in PGCs. For example, passive demeth-
and E11.5. Because the 5mC antibody (33D3) exhibits            ylation could account for the bulk of demethylation,
relatively weak avidity (e.g. as compared to the 5hmC          analogously to the zygote, whereas erasure of imprinted
antibody [71]) and 5mC levels at E10.5 may already be          regions may be targeted by specific active mechanisms that
partially depleted relative to somatic neighbours, this        are absent in the zygote, perhaps based on BER. Interest-
passive reduction of 5mC may appear as a largely complete      ingly, targeting specific loci with distinct demethylation
and, therefore, active erasure of global DNA methylation,      mechanisms may also lead to particular functional out-
while there would also be a significant loss of methylation     comes, perhaps based on the specific C derivative (5hmC,
by bisulfite sequencing. Although it is not clear precisely     5fC, 5caC, etc.) that directed demethylation. This could
how a passive demethylation mechanism might be trig-           have an important functional role during meiosis, similarly
gered, the existence of such a system would circumvent the     to the role of inherited histone modifications at develop-
potential genetic damage that may result from genome-          mental loci in the zygote [81]. Active DNA demethylation
wide BER.                                                      mechanisms could also operate in parallel to a passive
                                                               system to initiate DNA repair-driven chromatin remodel-
Parallel mechanisms of epigenetic reprogramming                ling [14]. Notably, other phases of epigenetic reprogram-
Although cumulative evidence indicates that several mo-        ming in PGCs also utilise parallel systems. For example,
lecular pathways of DNA demethylation may operate,             erasure of H3K9me2 in early PGCs may occur through
the precise mechanism(s) that mediate the comprehen-           both downregulation of the methyltransferase GLP and
sive methylation erasure in PGCs remain to be clarified         upregulation of specific lysine demethylases at approxi-
[44,75,76]. Indeed, DNA demethylation and chromatin            mately E8.5, which potentially contribute to erasure of this
remodelling in PGCs may occur through several comple-          modification in a redundant manner. Similarly, the histone
mentary parallel pathways, including active and passive        replacement that is associated with reorganisation of glob-
systems, which would provide a degree of redundancy            al H3K27me3 and H3K9me3 marks in PGCs at approxi-
and confer robustness and flexibility to the programme          mately E11.5 also occurs in parallel with upregulation of
(Figure 3). This seems necessary because of the funda-         specific lysine demethylases, which may contribute to the
mental importance of epigenetic reprogramming to germ          efficient erasure. Indeed, both Tet1 and Tet2 are also
cell development. Furthermore, there are very limited          upregulated at this stage, indicating a possible parallel
numbers of PGCs and most, if not all, of them must             role [50]. Thus, we propose that the unique importance of
transit through the process efficiently and prior to the        reprogramming in PGCs necessitates a system of multiple
entry of female germ cells into meiosis shortly after          epigenetic erasure mechanisms to confer efficiency, fidelity
reprogramming.                                                 and robustness to the process.
   A robust system based on multiple parallel mechanisms
and inbuilt redundancy may account for the observation         Potential roles of epigenetic reprogramming in PGCs
that Tet1-null and Tet2-null mice are viable and fertile,      The fundamental role of epigenetic reprogramming in
despite a probable role for 5hmC in demethylation in PGCs      PGCs is to overcome multiple epigenomic barriers to the
[77–80]. Although functional redundancy coupled with           eventual acquisition of totipotency acquired by epiblast
genetic background effects may explain this observation,       cells during early development. This is necessary because
it is also possible that impeded DNA demethylation in          mammals utilise an inductive mechanism of germ cell
TET-deficient PGCs would not cause complete loss of the         specification (they specify the germline from cells primed
cells but rather a reduction in their numbers. In support of   towards a somatic fate) rather than acquire germ cell fate
this, impeded demethylation of the paternal genome in          through an inherited germplasm. This essential role of
TET3-deficient zygotes is only associated with a severe         reprogramming in mammals is evident in mice lacking the
developmental phenotype in a subset of mutant embryos          transcriptional regulator PRDM14. Nascent PGCs in
170
Review                                                                                                                                           Trends in Genetics April 2012, Vol. 28, No. 4




                                                                      t 3 a / b , U h r f 1 d o wn r e
                                                              Dnm                                        gu l a
                                                                                                                  t io n
                                                                        T ET 1 / 2 a c t i v i t y

                                                                          Deamination




                                      5mC     5mC     5mC

                                                                                    Me
                               e




                                                                                                                                                       Pa s
                              M




                                                                                                                                                              sive
                         Ac




                                                                                                                                           BE R
                                                                                Ac

                                  e                                            Me
                              M




                                                                                                                           Reprogramming




                                                                                                                                                       DNA demethyla
                                                                  M
                                                                    e
                                      Ac




                                           Primed state




                                                                                                                                                                    tion
                                            H M T/
                                             Histo
                                              HDM




                                                   HA
                                                   ne e
                                                    /HD




                                                     Td
                                                        xch
                                                         AC




                                                         ow
                                                            an


                                                            nr
                                                             up




                                                              eg
                                                               ge
                                                                reg




                                                                ul
                                                                  ati
                                                                    ula




                                                                     on




                                                        on
                                                                        ti




                                                                        His
                                                                           ton
                                                                              em
                                                                                ark r
                                                                                     eprog                                                                                 M
                                                                                                                                     Ac




                                                                                          ramming                                                                              e
                                                                                                                                           Progress
                                                                                                                                                      ion toward
                                                                                                                                             Basal sta                     s
                                                                                                                                                                     te


                                                                                                                                                                               TRENDS in Genetics

Figure 3. Parallel routes towards the basal epigenetic state. Post-implantation epiblast cells acquire epigenetic modifications that constitute a barrier against their reversion
to a pluripotent state but remain primed to respond to signals that specify a primordial germ cell (PGC) fate. Upon specification, PGCs embark on a process of
reprogramming that overcomes the epigenetic barrier and establishes a basal epigenetic environment that underpins totipotency. Multiple parallel mechanisms contribute
to epigenetic reprogramming in PGCs to ensure robustness and redundancy to the process. Global erasure of DNA methylation (upper routes) can occur through passive or
active mechanisms directed through several interconnected processes. Likewise, reorganisation of chromatin architecture (lower routes) can occur through the parallel
upregulation of histone demethylases (HDM)/deacetylases (HDAC), downregulation of chromatin-modifying enzymes, including histone methyltransferases (HMT)/
acetyltransferases (HAT), and dynamic histone exchange. Abbreviations: 5mC, methylated cytosine; Ac, acetylation; BER, base excision repair; Dnmt3a/b, DNA (cytosine-5)-
methyltransferase 3a/b; Me, methylation; TET, ten-eleven translocation gene; Uhrf1, ubiquitin-like, containing PHD and RING finger domains 1.



Prdm14-mutant mice fail to undergo early reprogramming                                                    RNA Xist necessary to complete the process [18,82]. The
of chromatin and DNA methylation or to activate the germ                                                  wave of global DNA demethylation in PGCs at approxi-
cell genetic programme and consequently exhibit severely                                                  mately E11.5 seems to be a trigger for several other
impaired PGC development and are sterile [8]. Evidence                                                    important processes, including the erasure of parental
shows that PGC specification and epigenetic reprogram-                                                     imprints to enable establishment of new methylation
ming are intimately linked. Furthermore, within the broad                                                 marks according to the sex of the embryo [17,29]. The
remit of establishing a state of ‘underlying totipotency’,                                                significant impact that aberrant imprints have on mam-
epigenomic reprogramming has many distinct roles that                                                     malian development, foetal growth and behaviour, and in
collectively contribute to a continuous process of PGC                                                    human disease, has led to the suggestion that the whole
development (Figure 4).                                                                                   process of genome-wide demethylation in PGCs reflects a
   An important event driven by reprogramming is reacti-                                                  byproduct of the necessity to reset locus-specific imprints.
vation of Xi in female PGCs. This requires the integration                                                Indeed, as imprinted loci are resistant to demethylation
of multiple genetic and epigenetic systems; with early                                                    during zygotic reprogramming [82], they may have intrin-
chromatin reorganisation initiating X-reactivation and                                                    sic or epigenetic properties that preclude erasure except
DNA demethylation and repression of the long noncoding                                                    under exceptional cellular conditions, such as exists in
                                                                                                                                                                                                    171
Review                                                                                                              Trends in Genetics April 2012, Vol. 28, No. 4



                   (a)

                                                              Removal of epigenetic barriers to totipotency




                   (b)                                                  X-chromosome reactivation



                   (c)
                                                                             Epimutation erasure




                   (d)                                                                   Germline gene reactivation



                   (e)                                                                           Imprint erasure



                   (f)                                                                  Retrotransposon reactivation



                   (g)                                                                          Setting for meiosis




                                                                      R1                                 R2
                                                                ~E7.75–9.0                         ~E10.5–12.5




                                                                                                                                       TRENDS in Genetics

Figure 4. Potential functions of epigenetic reprogramming in primordial germ cells (PGCs). The distinct roles of epigenetic reprogramming in PGCs and the time point at
which each occurs (rectangles). (a) Epigenetic reprogramming events during PGC migration (R1) and post-migration (R2) overcomes the layers of epigenetic modifications
acquired by epiblast cells, which form a barrier to acquisition of totipotency. (b) In female cells, reprogramming contributes to X-chromosome reactivation. (c) Erasure of
epigenetic marks in PGCs also prevents perpetuation of epimutations through the germline. (d) Global DNA demethylation at R2 triggers activation of stably silenced genes
necessary for germ cell development (e) A fundamental role of reprogramming is to erase parent-of-origin imprints, thereby enabling subsequent sex-specific methylation
marks to be established during gametogenesis (f) Erasure of methylated cytosine (5mC) may permit transient expression of retrotransposons, which are subsequently
targeted for stable transcriptional repression by small RNAs, thereby ensuring that potentially harmful genetic elements are strongly silenced in the germline. (g)
Reprogramming in PGCs contributes to establishing a chromatin and transcriptional environment that is primed for entry into meiosis. Abbreviation: E, embryonic day.



PGCs. Global DNA demethylation in PGCs has other                                        transient activation of TEs during reprogramming
important functions, including the activation of stably                                 enables silencing mechanisms to target repressive chro-
silenced genes that are required for germ cell development                              matin, possibly directed by small RNAs derived from
[32,83]. Genes such as deleted in azoospermia-like (Dazl)                               expressed TEs [86]. This putative mechanism would en-
and synaptonemal complex protein 3 (Sycp3) are robustly                                 sure that harmful genetic elements that may have escaped
silenced by methylated CpG-dense promoters during post-                                 repressive mechanisms are rendered transcriptionally
implantation development, potentially to prevent ectopic                                quiescent throughout germ cell maturation. Notably,
activation that may drive malignant tumour growth, but                                  the transient erasure of the PGC epigenome may also
are activated by demethylation specifically in the germline                              provide an opportunity for epigenetic writers to access
[32,39,84,85]. Another key role of reprogramming in PGCs                                chromatin and establish new modifications that direct
is to erase aberrant epigenetic information or epimuta-                                 events that are crucial for meiosis. Indeed, the absence
tions. Erasure of such modifications in the zygote and                                   of some chromatin-modifying proteins, including G9a,
particularly in PGCs prevents their inheritance and per-                                MLL2, SUV39H1, SUV39H2, LSH and PRDM9, only
petuation through successive generations with potentially                               manifests in germ cells during meiosis [87–91]. One in-
detrimental effects.                                                                    triguing possibility is that epigenetic erasure is necessary
   Global DNA demethylation may generate an epigenetic                                  to promote chiasmata formation at recombination hot-
environment susceptible to expression of harmful trans-                                 spots during meiosis. Here, reprogramming may enable
posable elements (TE), yet paradoxically it is possible that                            the lysine methylase PRDM9 access to target H3K4me3
172
Review                                                                                                     Trends in Genetics April 2012, Vol. 28, No. 4


marks specifically to 13-mer recognition sites, which act as                      16 Shen, X. et al. (2008) EZH1 mediates methylation on histone H3 lysine
                                                                                    27 and complements EZH2 in maintaining stem cell identity and
beacons for recombination [92–95]. Thus, it is probable
                                                                                    executing pluripotency. Mol. Cell 32, 491–502
that functionally, epigenetic reprogramming in PGCs is a                         17 Sasaki, H. and Matsui, Y. (2008) Epigenetic events in mammalian
protracted event (E7.75–E12.5) that contributes to myriad                           germ-cell development: reprogramming and beyond. Nat. Rev. Genet. 9,
interconnected processes that are collectively essential for                        129–140
germ cell potency and development.                                               18 Chuva de Sousa Lopes, S.M. et al. (2008) X chromosome activity in
                                                                                    mouse XX primordial germ cells. PLoS Genet. 4, e30
                                                                                 19 Tada, T. et al. (1998) Epigenotype switching of imprintable loci in
Concluding remarks                                                                  embryonic germ cells. Dev. Genes Evol. 207, 551–561
Considerable advances have been made in understanding                            20 Matsui, Y. et al. (1992) Derivation of pluripotential embryonic stem
how new epigenetic information can be introduced, but less                          cells from murine primordial germ cells in culture. Cell 70, 841–847
is known about the mechanisms that can erase existing                            21 Ancelin, K. et al. (2006) Blimp1 associates with Prmt5 and directs
                                                                                    histone arginine methylation in mouse germ cells. Nat. Cell Biol. 8,
modifications. Studies on PGCs provide an unprecedented                              623–630
opportunity to unravel the role of key factors and their                         22 Tee, W.W. et al. (2010) Prmt5 is essential for early mouse development
combined roles in resetting the epigenome. Fundamental                              and acts in the cytoplasm to maintain ES cell pluripotency. Genes Dev.
knowledge gained from these studies may potentially find                             24, 2772–2777
wider application. Somatic cells are prone to epimutations                       23 Bostick, M. et al. (2007) UHRF1 plays a role in maintaining DNA
                                                                                    methylation in mammalian cells. Science 317, 1760–1764
through ageing and environmental factors, whereas the                            24 Sharif, J. et al. (2007) The SRA protein Np95 mediates epigenetic
germ line can reset the epigenome with the potential to                             inheritance by recruiting Dnmt1 to methylated DNA. Nature 450,
‘rejuvenate’ adult cells. Insights from research on early                           908–912
germ cells may provide knowledge and tools for applica-                          25 Kaneda, M. et al. (2004) Essential role for de novo DNA
                                                                                    methyltransferase Dnmt3a in paternal and maternal imprinting.
tions in ageing-related diseases and generally improve
                                                                                    Nature 429, 900–903
ability to manipulate cell fates for applications in repro-                      26 Kato, Y. et al. (2007) Role of the Dnmt3 family in de novo methylation of
ductive and regenerative medicine.                                                  imprinted and repetitive sequences during male germ cell development
                                                                                    in the mouse. Hum. Mol. Genet. 16, 2272–2280
                                                                                 27 Tachibana, M. et al. (2008) G9a/GLP complexes independently mediate
Acknowledgements
                                                                                    H3K9 and DNA methylation to silence transcription. EMBO J. 27,
We would like to thank Roopsha Sengupta and Harry Leitch for critical
                                                                                    2681–2690
reading of the manuscript and to apologise to the authors whose work
                                                                                 28 Dong, K.B. et al. (2008) DNA methylation in ES cells requires the lysine
could not be cited here owing to space constraints. JAH was funded by a
                                                                                    methyltransferase G9a but not its catalytic activity. EMBO J. 27,
Wellcome Trust Grant and JJZ is the recipient of a Wellcome Trust PhD
                                                                                    2691–2701
Scholarship. MAS is supported by The Wellcome Trust (RG49135).
                                                                                 29 Hajkova, P. et al. (2002) Epigenetic reprogramming in mouse
                                                                                    primordial germ cells. Mech. Dev. 117, 15–23
References                                                                       30 Lee, J. et al. (2002) Erasing genomic imprinting memory in mouse clone
 1 Surani, M.A. et al. (2007) Genetic and epigenetic regulators of                  embryos produced from day 11.5 primordial germ cells. Development
   pluripotency. Cell 128, 747–762                                                  129, 1807–1817
 2 Ginsburg, M. et al. (1990) Primordial germ cells in the mouse embryo          31 Lane, N. et al. (2003) Resistance of IAPs to methylation reprogramming
   during gastrulation. Development 110, 521–528                                    may provide a mechanism for epigenetic inheritance in the mouse.
 3 McLaren, A. (1984) Meiosis and differentiation of mouse germ cells.              Genesis 35, 88–93
   Symp. Soc. Exp. Biol. 38, 7–23                                                32 Maatouk, D.M. et al. (2006) DNA methylation is a primary mechanism
 4 Kocer, A. et al. (2009) Germ cell sex determination in mammals. Mol.             for silencing postmigratory primordial germ cell genes in both germ cell
   Hum. Reprod. 15, 205–213                                                         and somatic cell lineages. Development 133, 3411–3418
 5 Saitou, M. et al. (2002) A molecular programme for the specification of        33 Nestor, C. et al. (2010) Enzymatic approaches and bisulfite
   germ cell fate in mice. Nature 418, 293–300                                      sequencing cannot distinguish between 5-methylcytosine and 5-
 6 Yabuta, Y. et al. (2006) Gene expression dynamics during germline                hydroxymethylcytosine in DNA. Biotechniques 48, 317–319
   specification in mice identified by quantitative single-cell gene               34 Pichler, G. et al. (2011) Cooperative DNA and histone binding by Uhrf2
   expression profiling. Biol. Reprod. 75, 705–716                                   links the two major repressive epigenetic pathways. J. Cell. Biochem.
 7 Kurimoto, K. et al. (2008) Complex genome-wide transcription                     112, 2585–2593
   dynamics orchestrated by Blimp1 for the specification of the germ              35 West, J.A. et al. (2009) A role for Lin28 in primordial germ-cell
   cell lineage in mice. Genes Dev. 22, 1617–1635                                   development and germ-cell malignancy. Nature 460, 909–913
 8 Yamaji, M. et al. (2008) Critical function of Prdm14 for the establishment    36 Hayashi, K. et al. (2011) Reconstitution of the mouse germ cell
   of the germ cell lineage in mice. Nat. Genet. 40, 1016–1022                      specification pathway in culture by pluripotent stem cells. Cell 146,
 9 Ohinata, Y. et al. (2009) A signaling principle for the specification of the      519–532
   germ cell lineage in mice. Cell 137, 571–584                                  37 Popp, C. et al. (2010) Genome-wide erasure of DNA methylation in
10 McLaren, A. and Lawson, K.A. (2005) How is the mouse germ-cell                   mouse primordial germ cells is affected by AID deficiency. Nature 463,
   lineage established? Differentiation 73, 435–437                                 1101–1105
11 Seki, Y. et al. (2005) Extensive and orderly reprogramming of genome-         38 Mohn, F. et al. (2008) Lineage-specific polycomb targets and de novo
   wide chromatin modifications associated with specification and early               DNA methylation define restriction and potential of neuronal
   development of germ cells in mice. Dev. Biol. 278, 440–458                       progenitors. Mol. Cell 30, 755–766
12 Bao, S. et al. (2009) Epigenetic reversion of post-implantation epiblast      39 Borgel, J. et al. (2010) Targets and dynamics of promoter DNA
   to pluripotent embryonic stem cells. Nature 461, 1292–1295                       methylation during early mouse development. Nat. Genet. 42,
13 Seki, Y. et al. (2007) Cellular dynamics associated with the genome-             1093–1100
   wide epigenetic reprogramming in migrating primordial germ cells in           40 Wu, S.C. and Zhang, Y. (2010) Active DNA demethylation: many roads
   mice. Development 134, 2627–2638                                                 lead to Rome. Nat. Rev. Mol. Cell Biol. 11, 607–620
14 Hajkova, P. et al. (2008) Chromatin dynamics during epigenetic                41 Hodges, E. et al. (2011) Directional DNA methylation changes and
   reprogramming in the mouse germ line. Nature 452, 877–881                        complex intermediate states accompany lineage specificity in the adult
15 Tachibana, M. et al. (2005) Histone methyltransferases G9a and GLP               hematopoietic compartment. Mol. Cell 44, 17–28
   form heteromeric complexes and are both crucial for methylation of            42 Mikkelsen, T.S. et al. (2008) Dissecting direct reprogramming through
   euchromatin at H3-K9. Genes Dev. 19, 815–826                                     integrative genomic analysis. Nature 454, 49–55


                                                                                                                                                         173
Review                                                                                                 Trends in Genetics April 2012, Vol. 28, No. 4


43 Surani, M.A. and Hajkova, P. (2010) Epigenetic reprogramming of           69 Valinluck, V. and Sowers, L.C. (2007) Endogenous cytosine damage
   mouse germ cells toward totipotency. Cold Spring Harb. Symp. Quant.          products alter the site selectivity of human DNA maintenance
   Biol. 75, 211–218                                                            methyltransferase DNMT1. Cancer Res. 67, 946–950
44 Ooi, S.K. and Bestor, T.H. (2008) The colorful history of active DNA      70 Frauer, C. et al. (2011) Recognition of 5-hydroxymethylcytosine by the
   demethylation. Cell 133, 1145–1148                                           Uhrf1 SRA domain. PLoS ONE 6, e21306
45 Tahiliani, M. et al. (2009) Conversion of 5-methylcytosine to 5-          71 Ficz, G. et al. (2011) Dynamic regulation of 5-hydroxymethylcytosine in
   hydroxymethylcytosine in mammalian DNA by MLL partner TET1.                  mouse ES cells and during differentiation. Nature 473, 398–402
   Science 324, 930–935                                                      72 Hyldig, S.M. et al. (2011) Epigenetic reprogramming in the porcine
46 Ito, S. et al. (2010) Role of Tet proteins in 5mC to 5hmC conversion,        germ line. BMC Dev. Biol. 11, 11
   ES-cell self-renewal and inner cell mass specification. Nature 466,        73 Huang, Y. et al. (2010) The behaviour of 5-hydroxymethylcytosine in
   1129–1133                                                                    bisulfite sequencing. PLoS ONE 5, e8888
47 Wossidlo, M. et al. (2011) 5-Hydroxymethylcytosine in the mammalian       74 Tam, P.P. and Snow, M.H. (1981) Proliferation and migration of
   zygote is linked with epigenetic reprogramming. Nat. Commun. 2, 241          primordial germ cells during compensatory growth in mouse
48 Iqbal, K. et al. (2011) Reprogramming of the paternal genome upon            embryos. J. Embryol. Exp. Morphol. 64, 133–147
   fertilization involves genome-wide oxidation of 5-methylcytosine. Proc.   75 Bhutani, N. et al. (2011) DNA demethylation dynamics. Cell 146,
   Natl. Acad. Sci. U.S.A. 108, 3642–3647                                       866–872
49 Gu, T.P. et al. (2011) The role of Tet3 DNA dioxygenase in epigenetic     76 Law, J.A. and Jacobsen, S.E. (2010) Establishing, maintaining and
   reprogramming by oocytes. Nature 477, 606–610                                modifying DNA methylation patterns in plants and animals. Nat. Rev.
50 Hajkova, P. et al. (2010) Genome-wide reprogramming in the mouse             Genet. 11, 204–220
   germ line entails the base excision repair pathway. Science 329, 78       77 Dawlaty, M.M. et al. (2011) Tet1 is dispensable for maintaining
51 Wossidlo, M. et al. (2010) Dynamic link of DNA demethylation, DNA            pluripotency and its loss is compatible with embryonic and
   strand breaks and repair in mouse zygotes. EMBO J. 29, 1877–1888             postnatal development. Cell Stem Cell 9, 166–175
52 Cortellino, S. et al. (2011) Thymine DNA glycosylase is essential for     78 Li, Z. et al. (2011) Deletion of Tet2 in mice leads to dysregulated
   active DNA demethylation by linked deamination-base excision repair.         hematopoietic stem cells and subsequent development of myeloid
   Cell 146, 67–79                                                              malignancies. Blood 118, 4509–4518
53 Guo, J.U. et al. (2011) Hydroxylation of 5-methylcytosine by TET1         79 Moran-Crusio, K. et al. (2011) Tet2 loss leads to increased
   promotes active DNA demethylation in the adult brain. Cell 145,              hematopoietic stem cell self-renewal and myeloid transformation.
   423–434                                                                      Cancer Cell 20, 11–24
54 Bhutani, N. et al. (2010) Reprogramming towards pluripotency              80 Quivoron, C. et al. (2011) TET2 inactivation results in pleiotropic
   requires AID-dependent DNA demethylation. Nature 463, 1042–1047              hematopoietic abnormalities in mouse and is a recurrent event
55 Rai, K. et al. (2008) DNA demethylation in zebrafish involves the             during human lymphomagenesis. Cancer Cell 20, 25–38
   coupling of a deaminase, a glycosylase, and gadd45. Cell 135, 1201–1212   81 Hammoud, S.S. et al. (2009) Distinctive chromatin in human sperm
56 Muramatsu, M. et al. (2000) Class switch recombination and                   packages genes for embryo development. Nature 460, 473–478
   hypermutation require activation-induced cytidine deaminase (AID),        82 Morgan, H.D. et al. (2005) Epigenetic reprogramming in mammals.
   a potential RNA editing enzyme. Cell 102, 553–563                            Hum. Mol. Genet. 14, R47–R58 (Spec No 1)
57 Morrison, J.R. et al. (1996) Apolipoprotein B RNA editing enzyme-         83 Rodic, N. et al. (2005) DNA methylation is required for silencing
   deficient mice are viable despite alterations in lipoprotein metabolism.      of ant4, an adenine nucleotide translocase selectively expressed
   Proc. Natl. Acad. Sci. U.S.A. 93, 7154–7159                                  in mouse embryonic stem cells and germ cells. Stem Cells 23,
58 Mikl, M.C. et al. (2005) Mice deficient in APOBEC2 and APOBEC3.               1314–1323
   Mol. Cell. Biol. 25, 7270–7277                                            84 Janic, A. et al. (2010) Ectopic expression of germline genes drives
59 Morgan, H.D. et al. (2004) Activation-induced cytidine deaminase             malignant brain tumor growth in Drosophila. Science 330, 1824–1827
   deaminates 5-methylcytosine in DNA and is expressed in                    85 Simpson, A.J. et al. (2005) Cancer/testis antigens, gametogenesis and
   pluripotent tissues. J. Biol. Chem. 279, 52353–52360                         cancer. Nat. Rev. Cancer 5, 615–625
60 He, Y.F. et al. (2011) Tet-mediated formation of 5-carboxylcytosine and   86 Seisenberger, S. et al. (2010) Retrotransposons and germ cells:
   its excision by TDG in mammalian DNA. Science 333, 1303–1307                 reproduction, death, and diversity. F1000 Biol. Rep. 2
61 Ito, S. et al. (2011) Tet proteins can convert 5-methylcytosine to 5-     87 Tachibana, M. et al. (2007) Functional dynamics of H3K9 methylation
   formylcytosine and 5-carboxylcytosine. Science 333, 1300–1303                during meiotic prophase progression. EMBO J. 26, 3346–3359
62 Maiti, A. and Drohat, A.C. (2011) Thymine DNA glycosylase can             88 Peters, A.H. et al. (2001) Loss of the Suv39 h histone
   rapidly excise 5-formylcytosine and 5–carboxylcytosine: potential            methyltransferases impairs mammalian heterochromatin and
   implications for active demethylation of CpG sites. J. Biol. Chem.           genome stability. Cell 107, 323–337
   286, 35334–35338                                                          89 Glaser, S. et al. (2009) The histone 3 lysine 4 methyltransferase, Mll2,
63 Cannon, S.V. et al. (1988) 5-Hydroxymethylcytosine DNA glycosylase           is only required briefly in development and spermatogenesis.
   activity in mammalian tissue. Biochem. Biophys. Res. Commun. 151,            Epigenetics Chromatin 2, 5
   1173–1179                                                                 90 De La Fuente, R. et al. (2006) Lsh is required for meiotic chromosome
64 Choi, Y. et al. (2002) DEMETER, a DNA glycosylase domain protein, is         synapsis and retrotransposon silencing in female germ cells. Nat. Cell
   required for endosperm gene imprinting and seed viability in                 Biol. 8, 1448–1454
   arabidopsis. Cell 110, 33–42                                              91 Kota, S.K. and Feil, R. (2010) Epigenetic transitions in germ cell
65 Gong, Z. et al. (2002) ROS1, a repressor of transcriptional gene             development and meiosis. Dev. Cell 19, 675–686
   silencing in Arabidopsis, encodes a DNA glycosylase/lyase. Cell 111,      92 Myers, S. et al. (2010) Drive against hotspot motifs in primates
   803–814                                                                      implicates the PRDM9 gene in meiotic recombination. Science 327,
66 Okada, Y. et al. (2010) A role for the elongator complex in zygotic          876–879
   paternal genome demethylation. Nature 463, 554–558                        93 Baudat, F. et al. (2010) prdm9 is a major determinant of meiotic
67 Schmitz, K.M. et al. (2009) TAF12 recruits Gadd45a and the nucleotide        recombination hotspots in humans and mice. Science 327, 836–840
   excision repair complex to the promoter of rRNA genes leading to active   94 Parvanov, E.D. et al. (2010) Prdm9 controls activation of mammalian
   DNA demethylation. Mol. Cell 33, 344–353                                     recombination hotspots. Science 327, 835
68 Inoue, A. and Zhang, Y. (2011) Replication-dependent loss of              95 Hayashi, K. et al. (2005) A histone H3 methyltransferase
   5-Hydroxymethylcytosine in mouse preimplantation embryos.                    controls epigenetic events required for meiotic prophase. Nature
   Science 334, 194                                                             438, 374–378




174

Mais conteúdo relacionado

Mais procurados

Cell cycle (Without Mieosis)
Cell cycle (Without Mieosis)Cell cycle (Without Mieosis)
Cell cycle (Without Mieosis)Dion Orquia
 
E. coli plasmids based vectors
E. coli plasmids based vectorsE. coli plasmids based vectors
E. coli plasmids based vectorsRashmi Rawat
 
Protein engineering
Protein engineeringProtein engineering
Protein engineeringbansalaman80
 
Gene expression introduction
Gene expression introductionGene expression introduction
Gene expression introductionSetia Pramana
 
Selection & Screening of Recombinant cells & expression of recombinant (2) (1)
Selection & Screening of  Recombinant cells & expression of recombinant (2) (1)Selection & Screening of  Recombinant cells & expression of recombinant (2) (1)
Selection & Screening of Recombinant cells & expression of recombinant (2) (1)SunandaArya
 
Dr waheed presentation (1)
Dr waheed presentation (1)Dr waheed presentation (1)
Dr waheed presentation (1)Zahid Hussain
 
Eukaryotic gene Regulation II 2014
Eukaryotic gene Regulation II 2014  Eukaryotic gene Regulation II 2014
Eukaryotic gene Regulation II 2014 Jill Howlin
 
Regulation of gene expression in prokaryotes and viruses
Regulation of gene expression in prokaryotes and virusesRegulation of gene expression in prokaryotes and viruses
Regulation of gene expression in prokaryotes and virusesNOOR ARSHIA
 
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...wangdong2336
 

Mais procurados (18)

P bluescript
P bluescriptP bluescript
P bluescript
 
Thesis - Haluska
Thesis - HaluskaThesis - Haluska
Thesis - Haluska
 
Gene expression
Gene expressionGene expression
Gene expression
 
Cell cycle (Without Mieosis)
Cell cycle (Without Mieosis)Cell cycle (Without Mieosis)
Cell cycle (Without Mieosis)
 
E. coli plasmids based vectors
E. coli plasmids based vectorsE. coli plasmids based vectors
E. coli plasmids based vectors
 
Protein engineering
Protein engineeringProtein engineering
Protein engineering
 
Protein engineering
Protein engineeringProtein engineering
Protein engineering
 
Gene expression introduction
Gene expression introductionGene expression introduction
Gene expression introduction
 
Selection & Screening of Recombinant cells & expression of recombinant (2) (1)
Selection & Screening of  Recombinant cells & expression of recombinant (2) (1)Selection & Screening of  Recombinant cells & expression of recombinant (2) (1)
Selection & Screening of Recombinant cells & expression of recombinant (2) (1)
 
Gene expression in chloroplast
Gene expression in chloroplastGene expression in chloroplast
Gene expression in chloroplast
 
Dr waheed presentation (1)
Dr waheed presentation (1)Dr waheed presentation (1)
Dr waheed presentation (1)
 
Site directed mutagenesis by pcr
Site directed mutagenesis by pcrSite directed mutagenesis by pcr
Site directed mutagenesis by pcr
 
pUC18 vector
pUC18 vector pUC18 vector
pUC18 vector
 
Thesis
ThesisThesis
Thesis
 
P 53 Tumour Biology
P 53 Tumour BiologyP 53 Tumour Biology
P 53 Tumour Biology
 
Eukaryotic gene Regulation II 2014
Eukaryotic gene Regulation II 2014  Eukaryotic gene Regulation II 2014
Eukaryotic gene Regulation II 2014
 
Regulation of gene expression in prokaryotes and viruses
Regulation of gene expression in prokaryotes and virusesRegulation of gene expression in prokaryotes and viruses
Regulation of gene expression in prokaryotes and viruses
 
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...Glucocorticoids modulate micro rna expression and processing during lymphocyt...
Glucocorticoids modulate micro rna expression and processing during lymphocyt...
 

Semelhante a Parallel mechanisms of epigenetic reprogramming in the germline

Cell Cycle
Cell CycleCell Cycle
Cell CycleYapa
 
1768816628DNA replication in eukaryotes.pdf
1768816628DNA replication in eukaryotes.pdf1768816628DNA replication in eukaryotes.pdf
1768816628DNA replication in eukaryotes.pdfAsmaAnaitullah
 
Cell cycle check point By KK Sahu Sir
Cell cycle check point  By KK Sahu SirCell cycle check point  By KK Sahu Sir
Cell cycle check point By KK Sahu SirKAUSHAL SAHU
 
Tumor suppressor
Tumor suppressorTumor suppressor
Tumor suppressorfatmafars
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseGedion Yilma
 
Cupid Peptides presentation wjr
Cupid Peptides presentation wjrCupid Peptides presentation wjr
Cupid Peptides presentation wjrCupid Peptides
 
Review 4.30.2010
Review 4.30.2010Review 4.30.2010
Review 4.30.2010Greg
 
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingDirect Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingAhmed Madni
 
Unit 4-cell cycle and cell division
Unit 4-cell cycle and cell divisionUnit 4-cell cycle and cell division
Unit 4-cell cycle and cell divisionKomal Kp
 
Regulation of cell cycle
Regulation of cell cycleRegulation of cell cycle
Regulation of cell cyclemohit kumar
 
Oncogenesis in Neurosurgery.pptx
Oncogenesis in Neurosurgery.pptxOncogenesis in Neurosurgery.pptx
Oncogenesis in Neurosurgery.pptxGypsyDanger6
 
Gene regulation eukaryote spptx
Gene regulation eukaryote spptxGene regulation eukaryote spptx
Gene regulation eukaryote spptxaljeirou
 
Revised- Gene regulatiion Eukaryotics.pdf
Revised- Gene regulatiion Eukaryotics.pdfRevised- Gene regulatiion Eukaryotics.pdf
Revised- Gene regulatiion Eukaryotics.pdfHemanhuelCTankxes
 
Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Dr Durga Gahlot
 

Semelhante a Parallel mechanisms of epigenetic reprogramming in the germline (20)

Cell Cycle
Cell CycleCell Cycle
Cell Cycle
 
Cancer
CancerCancer
Cancer
 
Seminar 29082022.pptx
Seminar 29082022.pptxSeminar 29082022.pptx
Seminar 29082022.pptx
 
1768816628DNA replication in eukaryotes.pdf
1768816628DNA replication in eukaryotes.pdf1768816628DNA replication in eukaryotes.pdf
1768816628DNA replication in eukaryotes.pdf
 
Cell cycle check point By KK Sahu Sir
Cell cycle check point  By KK Sahu SirCell cycle check point  By KK Sahu Sir
Cell cycle check point By KK Sahu Sir
 
molecular.pdf
molecular.pdfmolecular.pdf
molecular.pdf
 
Tumor suppressor
Tumor suppressorTumor suppressor
Tumor suppressor
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinase
 
Cupid Peptides presentation wjr
Cupid Peptides presentation wjrCupid Peptides presentation wjr
Cupid Peptides presentation wjr
 
Review 4.30.2010
Review 4.30.2010Review 4.30.2010
Review 4.30.2010
 
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingDirect Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
 
Unit 4-cell cycle and cell division
Unit 4-cell cycle and cell divisionUnit 4-cell cycle and cell division
Unit 4-cell cycle and cell division
 
Regulation of cell cycle
Regulation of cell cycleRegulation of cell cycle
Regulation of cell cycle
 
Oncogenes
OncogenesOncogenes
Oncogenes
 
Oncogenesis in Neurosurgery.pptx
Oncogenesis in Neurosurgery.pptxOncogenesis in Neurosurgery.pptx
Oncogenesis in Neurosurgery.pptx
 
Gene regulation eukaryote spptx
Gene regulation eukaryote spptxGene regulation eukaryote spptx
Gene regulation eukaryote spptx
 
Revised- Gene regulatiion Eukaryotics.pdf
Revised- Gene regulatiion Eukaryotics.pdfRevised- Gene regulatiion Eukaryotics.pdf
Revised- Gene regulatiion Eukaryotics.pdf
 
publication 1
publication 1publication 1
publication 1
 
Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.Cell cycle and molecular basis of cancer.
Cell cycle and molecular basis of cancer.
 
Cancer genes
Cancer genesCancer genes
Cancer genes
 

Mais de Loki Stormbringer

Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...
Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...
Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...Loki Stormbringer
 
Meta analysis of genome-wide association studies for personality
Meta analysis of genome-wide association studies for personalityMeta analysis of genome-wide association studies for personality
Meta analysis of genome-wide association studies for personalityLoki Stormbringer
 
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255Loki Stormbringer
 
Primary surface ruptures of the great himalayan
Primary surface ruptures of the great himalayanPrimary surface ruptures of the great himalayan
Primary surface ruptures of the great himalayanLoki Stormbringer
 
Metal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementiaMetal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementiaLoki Stormbringer
 
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...Loki Stormbringer
 
Interventions to Improve Cognitive Functioning After TBI
Interventions to Improve Cognitive Functioning After TBIInterventions to Improve Cognitive Functioning After TBI
Interventions to Improve Cognitive Functioning After TBILoki Stormbringer
 
Matrix rigidity controls endothelial differentiation and morphogenesis of car...
Matrix rigidity controls endothelial differentiation and morphogenesis of car...Matrix rigidity controls endothelial differentiation and morphogenesis of car...
Matrix rigidity controls endothelial differentiation and morphogenesis of car...Loki Stormbringer
 
Mechanisms of white matter changes induced by meditation
Mechanisms of white matter changes induced by meditationMechanisms of white matter changes induced by meditation
Mechanisms of white matter changes induced by meditationLoki Stormbringer
 
Appeal from the united states district court for the eastern district of wisc...
Appeal from the united states district court for the eastern district of wisc...Appeal from the united states district court for the eastern district of wisc...
Appeal from the united states district court for the eastern district of wisc...Loki Stormbringer
 
CTC1 deletion results in defective telomere replication, leading to catastrop...
CTC1 deletion results in defective telomere replication, leading to catastrop...CTC1 deletion results in defective telomere replication, leading to catastrop...
CTC1 deletion results in defective telomere replication, leading to catastrop...Loki Stormbringer
 
Metabolic depression in hibernation and major depression: an explanatory theo...
Metabolic depression in hibernation and major depression: an explanatory theo...Metabolic depression in hibernation and major depression: an explanatory theo...
Metabolic depression in hibernation and major depression: an explanatory theo...Loki Stormbringer
 

Mais de Loki Stormbringer (12)

Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...
Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...
Anatomical plasticity of adult brain is titrated by nogo receptor 1 (06 March...
 
Meta analysis of genome-wide association studies for personality
Meta analysis of genome-wide association studies for personalityMeta analysis of genome-wide association studies for personality
Meta analysis of genome-wide association studies for personality
 
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255
Primary surface ruptures of the great Himalayan earthquakes in 1934 and 1255
 
Primary surface ruptures of the great himalayan
Primary surface ruptures of the great himalayanPrimary surface ruptures of the great himalayan
Primary surface ruptures of the great himalayan
 
Metal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementiaMetal protein attenuating compounds for the treatment of alzheimer's dementia
Metal protein attenuating compounds for the treatment of alzheimer's dementia
 
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...
The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and ...
 
Interventions to Improve Cognitive Functioning After TBI
Interventions to Improve Cognitive Functioning After TBIInterventions to Improve Cognitive Functioning After TBI
Interventions to Improve Cognitive Functioning After TBI
 
Matrix rigidity controls endothelial differentiation and morphogenesis of car...
Matrix rigidity controls endothelial differentiation and morphogenesis of car...Matrix rigidity controls endothelial differentiation and morphogenesis of car...
Matrix rigidity controls endothelial differentiation and morphogenesis of car...
 
Mechanisms of white matter changes induced by meditation
Mechanisms of white matter changes induced by meditationMechanisms of white matter changes induced by meditation
Mechanisms of white matter changes induced by meditation
 
Appeal from the united states district court for the eastern district of wisc...
Appeal from the united states district court for the eastern district of wisc...Appeal from the united states district court for the eastern district of wisc...
Appeal from the united states district court for the eastern district of wisc...
 
CTC1 deletion results in defective telomere replication, leading to catastrop...
CTC1 deletion results in defective telomere replication, leading to catastrop...CTC1 deletion results in defective telomere replication, leading to catastrop...
CTC1 deletion results in defective telomere replication, leading to catastrop...
 
Metabolic depression in hibernation and major depression: an explanatory theo...
Metabolic depression in hibernation and major depression: an explanatory theo...Metabolic depression in hibernation and major depression: an explanatory theo...
Metabolic depression in hibernation and major depression: an explanatory theo...
 

Último

Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...astropune
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableNehru place Escorts
 
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Dipal Arora
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...chandars293
 
Chandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableChandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableDipal Arora
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiAlinaDevecerski
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escortsvidya singh
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Call Girls in Nagpur High Profile
 
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...narwatsonia7
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...narwatsonia7
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...Arohi Goyal
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...astropune
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipur
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls JaipurRussian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipur
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipurparulsinha
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableDipal Arora
 

Último (20)

Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
Best Rate (Hyderabad) Call Girls Jahanuma ⟟ 8250192130 ⟟ High Class Call Girl...
 
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls AvailableVip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
Vip Call Girls Anna Salai Chennai 👉 8250192130 ❣️💯 Top Class Girls Available
 
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
Best Rate (Guwahati ) Call Girls Guwahati ⟟ 8617370543 ⟟ High Class Call Girl...
 
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...Top Rated  Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
Top Rated Hyderabad Call Girls Erragadda ⟟ 6297143586 ⟟ Call Me For Genuine ...
 
Chandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD availableChandrapur Call girls 8617370543 Provides all area service COD available
Chandrapur Call girls 8617370543 Provides all area service COD available
 
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls DelhiRussian Escorts Girls  Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
Russian Escorts Girls Nehru Place ZINATHI 🔝9711199012 ☪ 24/7 Call Girls Delhi
 
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore EscortsCall Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
Call Girls Horamavu WhatsApp Number 7001035870 Meeting With Bangalore Escorts
 
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Coimbatore Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ooty Just Call 9907093804 Top Class Call Girl Service Available
 
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
Book Paid Powai Call Girls Mumbai 𖠋 9930245274 𖠋Low Budget Full Independent H...
 
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 8250192130 ⟟ Call Me For Gen...
 
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...High Profile Call Girls Coimbatore Saanvi☎️  8250192130 Independent Escort Se...
High Profile Call Girls Coimbatore Saanvi☎️ 8250192130 Independent Escort Se...
 
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Dehradun Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Faridabad Just Call 9907093804 Top Class Call Girl Service Available
 
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
All Time Service Available Call Girls Marine Drive 📳 9820252231 For 18+ VIP C...
 
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
♛VVIP Hyderabad Call Girls Chintalkunta🖕7001035870🖕Riya Kappor Top Call Girl ...
 
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Kochi Just Call 9907093804 Top Class Call Girl Service Available
 
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipur
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls JaipurRussian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipur
Russian Call Girls in Jaipur Riya WhatsApp ❤8445551418 VIP Call Girls Jaipur
 
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Cuttack Just Call 9907093804 Top Class Call Girl Service Available
 
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service AvailableCall Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
Call Girls Ludhiana Just Call 9907093804 Top Class Call Girl Service Available
 

Parallel mechanisms of epigenetic reprogramming in the germline

  • 1. Review Parallel mechanisms of epigenetic reprogramming in the germline Jamie A. Hackett, Jan J. Zylicz and M. Azim Surani Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 1QN, UK Germ cells possess the extraordinary and unique capac- [5,6]. This process is initiated in response to localised sig- ity to give rise to a new organism and create an enduring nals, including BMP4 and WNT3, which direct activation of link between all generations. To acquire this property, the key transcriptional regulators B-lymphocyte-induced primordial germ cells (PGCs) transit through an unprec- maturation protein 1 (Blimp1) and PR domain containing edented programme of sequential epigenetic events that 14 (Prdm14) in competent epiblast cells [1,7–9]. Lineage- culminates in an epigenomic basal state that is the restricted PGCs then embark on an orchestrated sequence foundation of totipotency. This process is underpinned of reprogramming that culminates in a basal epigenetic by genome-wide DNA demethylation, which may occur state. The number of early PGCs is highly restricted (ap- through several overlapping pathways, including con- proximately 40 by E7.25) so it is crucial that the complex version to 5-hydroxymethylcytosine. We propose that series of epigenetic events be robust to ensure that most, if the epigenetic programme in PGCs operates through not all, cells efficiently transit through the process [10]. multiple parallel mechanisms to ensure robustness at Reprogramming must also proceed rapidly because of strict the level of individual cells while also being flexible through functional redundancy to guarantee high fideli- Glossary ty of the process. Gaining a better understanding of the 5-hydroxymethylcytosine (5hmC): oxidation of methylated cytosines (5mC) by molecular mechanisms that direct epigenetic repro- TET proteins generates 5hmC, which may be an intermediate during DNA gramming in PGCs will enhance our ability to manipu- demethylation and can be further converted to 5caC and 5fC. 5hmC is enriched late epigenetic memory, cell-fate decisions and in pluripotent and some neuronal cell types, but its precise functional consequences in the genome and its role in DNA demethylation are unclear. applications in regenerative medicine. Base excision repair (BER): a cellular mechanism for repair of nonhelix- distorting base mutations or lesions in the genome. BER is initiated by a DNA Reprogramming PGCs towards totipotency glycosylase (e.g. TDG) that removes inappropriate bases and forms an Development from the zygote to adulthood is characterised apurinic/apyrimidinic (AP) site, which is then cleaved by an AP endonuclease and repaired by specific lyases and polymerases. BER may function to remove by a progressive restriction of cellular potential that gives downstream derivatives of 5mC, such as 5caC, and mediate repair to rise to all the differentiated somatic cell types. A unique unmodified C [60]. Basal epigenetic state: the unique epigenetic state of PGCs following exception to this unidirectional process occurs in the germ- reprogramming. By E13.5, the PGC epigenome has undergone extensive line, where an unprecedented reprogramming event in reorganisation of histone modifications and is stripped of genome-wide DNA PGCs (see Glossary) reverses epigenetic barriers to plas- methylation, rendering it at its most basal level during the mammalian life cycle. ticity and resets genomic potential. Reprogramming in Bisulfite sequencing: a technique used to determine the pattern of allelic DNA PGCs results in chromatin remodelling, erasure of genomic methylation (5mC) at specific genomic regions. Bisulfite sequencing cannot imprints and extensive DNA demethylation [1]. This pro- distinguish 5mC from 5hmC [33]. Additionally, 5caC is indistinguishable from unmodified C by bisulfite sequencing [60]. cess represents the most comprehensive erasure of epige- DNA demethylation: the removal of a methyl group from position 5 of a netic information in the mammalian life cycle and cytosine base (5mC), which usually resides within a CpG genomic context, to underpins the totipotent state. Therefore, unravelling generate an unmodified C. DNA demethylation may occur through either a ‘passive’ mechanism that relies on replication-dependent dilution or an ‘active’ the mechanisms that drive reprogramming, particularly process driven by enzymatic replacement independently of DNA replication. As DNA demethylation, in the unique context of PGCs is of DNA methylation is associated with transcriptional silencing, DNA demethyla- great interest. tion can generate a transcriptionally competent state. Epigenetic reprogramming: genome-wide reorganisation of epigenetic mod- In mice, PGCs are specified from a subset of posterior ifications that overcomes stable epigenetic barriers and enables acquisition of proximal epiblast cells at approximately embryonic day (E) genomic potential. During the mammalian life cycle, epigenetic reprogram- ming occurs in PGCs and in early zygotic development. 6.25, resulting in the establishment of a founder population Genomic imprints: genomic sequences that exhibit differences in CpG methyla- of PGCs at E7.25 [1,2]. These nascent PGCs subsequently tion according to the parent of origin. These differentially methylated regions migrate to the genital ridges by approximately E10.5 and, (DMRs) can influence the allele-specific expression of one or more genes. Primordial germ cell (PGC): the precursors of mature germ cells that are from E12.5 onwards, they undergo sex-specific development specified during post-implantation development. In vivo, PGCs are restricted in the gonads [3,4]. Because mammalian PGCs are specified as a unipotent lineage and only give rise to gametes, which generate the from cells that are already primed towards a somatic fate, totipotent state upon fertilisation. Early PGCs also possess an underlying genomic plasticity, as evidenced through their capacity to form pluripotent EG nascent PGCs must both repress the ongoing somatic pro- cells upon in vitro culture. gramme and activate the germ cell transcriptional network Totipotency: the ability of a cell to give rise to all the cell types of the embryonic and extra-embryonic lineages. By contrast, pluripotency refers to the capacity of a cell to generate all the cell types of the embryo. Corresponding author: Surani, M.A. (a.surani@gurdon.cam.ac.uk). 164 0168-9525/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. doi:10.1016/j.tig.2012.01.005 Trends in Genetics, April 2012, Vol. 28, No. 4
  • 2. Review Trends in Genetics April 2012, Vol. 28, No. 4 temporal constraints imposed by the entry of male germ sperm or oocytes in vivo, during migration (E8.5–E11.5), cells into mitotic arrest and female germ cells into meiotic PGCs show multiple transcriptional and epigenetic simi- arrest at approximately E13.5. To overcome these con- larities to pluripotent ES cells. Indeed, E8.5–E11.5 PGCs straints, epigenetic reprogramming in PGCs is probably can form pluripotent embryonic germ (EG) cells in vitro, directed by multiple parallel mechanisms that ensure the which resemble ES cells rather than the post-implantation fidelity, flexibility and efficiency of this fundamental pro- epiblast cells from which PGCs were originally specified cess. As such, events like genome-wide erasure of DNA [19,20]. It is possible that this epigenetic state of PGCs is methylation in PGCs may occur through several intercon- an underlying requirement for the initiation of meiosis nected mechanisms, including both active and passive path- and the eventual acquisition of totipotency in the zygote. ways, that collectively confer redundancy and hence Interestingly, migrating PGCs additionally exhibit upre- robustness to PGC reprogramming and development. gulation of histone H2A/H4 arginine 3 symmetrical meth- Here, we discuss the epigenetic events in PGCs and ylation (H2A/H4R3me2 s), which is catalysed by protein the mechanisms that may operate to drive epigenetic arginine methyltransferase 5 (PRMT5) [21]. This modifi- reprogramming. We suggest that an integrated process cation may contribute to maintaining PGCs in a unipotent involving parallel systems is at play and consider the state in vivo and to repression of the somatic programme potential pathways of DNA demethylation. We also high- [22]. light some of the potential roles and developmental Changes in histone modifications occur in parallel with processes that epigenetic reprogramming contributes to a reported reduction in global levels of DNA methylation in PGCs. (5mC) in migrating PGCs from approximately E8.0 [11]. Any loss of 5mC might reflect the effects of BLIMP1 and Epigenetic events in PGCs PRDM14, which repress both DNA (cytosine-5)-methyl- At the point at which PGCs are specified from post-im- transferase 3a and 3b (Dnmt3a and Dnmt3b) and ubiqui- plantation epiblast cells, they are epigenetically indistin- tin-like, containing PHD and RING finger domains 1 guishable at the global level from their neighbours, which (Uhrf1), which are essential components of the de novo are destined for a somatic fate [9,11]. Therefore, nascent and maintenance methylation machinery, respectively PGCs inherit stable epigenetic states, including DNA (Figure 1) [7,8,23–26]. Additionally, repression of GLP methylation and X-inactivation, which constitute an epi- may directly affect DNA methylation through a parallel genetic barrier against the eventual acquisition of totipo- mechanism that is both dependent and independent of tency [1,12]. It is thus an important early step in PGC H3K9me2 [27,28]. However, most analysed genomic development to initiate a process of reprogramming that regions, including transposable elements, imprinted loci erases these stable epigenetic blocks. The first gross epi- and single-copy genes, apparently retain DNA methylation genetic changes in PGCs entail a reciprocal loss of histone at CpG sites in PGCs until at least E10.5 [29–32], although H3 lysine 9 dimethylation (H3K9me2) from E7.75 and, in this does not exclude the possibility that there is conver- most PGCs, a global increase of H3 lysine 27 trimethyla- sion of 5mC to 5-hydroxymethylcytosine (5hmC) [33]. tion (H3K27me3) by E9.5 (Figure 1) [11,13,14]. The ge- Therefore, it is unclear to what extent DNA demethylation nome-wide depletion of H3K9me2 is potentially a contributes to the early stages of epigenetic reprogram- consequence of the downregulation of GLP, a methyltrans- ming (from E8.0) in PGCs and whether conversion to 5hmC ferase that forms a heteromeric complex with G9a (also plays a role. Conversely, it is unclear how 5mC (or 5hmC) is known as EHMT2) that is required for deposition of H3K9 maintained at analysed genomic regions in migrating mono- and dimethylation, and the parallel upregulation of PGCs given that essential components of the de novo specific lysine demethylases ([15] and unpublished obser- and maintenance machinery, particularly UHRF1, are vations). By contrast, the mechanisms responsible for the absent [7]. One possibility is that UHRF2, which is con- increased H3K27me3 levels in PGCs remain unclear, al- served with UHRF1 at the sequence level and preferen- though notably enhancer of zeste homologue 1 (Ezh1), tially binds hemi-methylated DNA associated with which has H3K27me3 methyltransferase activity, is upre- H3K9me3, can compensate for UHRF1 to maintain DNA gulated in PGCs [16]. Because H3K9me2 and H3K27me3 methylation in PGCs, either globally or at specific loci [34]. marks are both associated with transcriptional repression, In support of this, UHRF2 is specifically upregulated it has been postulated that the loss of H3K9me2 is com- during PGC specification, at least in vitro [7,35]. Addition- plemented by the gain of H3K27me3 to maintain a repres- ally, unlike H3K9me2, global H3K9me3 levels are main- sive chromatin state in PGCs [17]. However, the precise tained in migrating PGCs [14]. The in vitro PGC-like cells genomic location and relationship between these epigenet- (PGCLC) generated in an elegant recent study may enable ic changes remains to be determined. Nonetheless, the further mechanistic insights into epigenetic events in na- global enrichment of H3K27me3 and loss of H3K9me2 scent PGCs at a higher resolution [36]. establishes a chromatin environment in PGCs that is The early stages of epigenomic reorganisation in PGCs grossly similar to that in pluripotent embryonic stem are followed by, and are probably a prerequisite for, the (ES) cells and is coupled to upregulation of pluripotency dramatic genome-wide erasure of DNA methylation and genes, such as Nanog and SRY Sox2 [1]. Additionally, extensive chromatin remodelling subsequent to entry into unlike global H3K27me3 levels, the inactive X-chromo- the genital ridges at approximately E10.5. Because gross some (Xi) exhibits a protracted decline in H3K27me3 in DNA demethylation seems to occur rapidly at a distinct female PGCs, which is linked to initiation of X-reactivation time point, while most PGCs are in G2 phase, it is held that [18]. Thus, while PGCs are unipotent and only form either this process is an ‘active’ event occurring independently 165
  • 3. Review Trends in Genetics April 2012, Vol. 28, No. 4 (a) Chromatin dynamics 5mC H3K27me3 Relative levels H3K9 me2 E5.5 E6.5 E8.0 E10.5 E11.5 (b) Specification and chromatin-modifying factors Blimp1 Relative expression Prdm14 G9a Glp E5.5 E6.5 E8.0 E10.5 E11.5 (c) DNA methylation factors Uhrf2 Relative expression Dnmt1 Dnmt3a Dnmt3b Uhrf1 E5.5 E6.5 E8.0 E10.5 E11.5 (d) Tet dioxygenases Relative expression Tet1 Tet2 Tet3 E5.5 E6.5 E8.0 E10.5 E11.5 Spec. R1 R2 TRENDS in Genetics Figure 1. Chromatin and transcriptional changes in primordial germ cells (PGCs). Following specification (Spec.), PGCs transit through two sequential phases of reprogramming during migration (R1) and subsequent to entry into the genital ridge (R2). (a) Epigenomic reorganisation in PGCs. At R1, PGCs exhibit global erasure of histone H3 lysine 9 dimethylation (H3K9me2), upregulation of H3 lysine 27 trimethylation (H3K27me3) and some loss of the DNA methylation signal. At R2, there is a further dramatic loss of DNA methylation, which includes erasure of imprints. This correlates with a transient reorganisation of H3K27me3, loss of heterochromatic chromocentres and remodelling of other chromatin modifications, including histone H2A/H4 arginine 3 symmetrical methylation (H2A/H4R3me2; not shown). (b–d) Transcriptional changes during PGC development [13]. (b) Specification and chromatin-modifying factors. Upregulation of B-lymphocyte-induced maturation protein 1 (Blimp1) and PR domain containing 14 (Prdm14) is necessary for specification of PGC fate, whereas the parallel downregulation of glucagon-like peptide (Glp)/G9a and upregulation of lysine demethylases (not shown) contribute to erasure of H3K9me2 at R1. (c) DNA methylation proteins. Downregulation of DNA (cytosine-5)-methyltransferase 3b (Dnmt3b) and ubiquitin-like, containing PHD and RING finger domains 1 (Uhrf1) may account for DNA demethylation at R1. UHRF2 may partially compensate for the absence of UHRF1 to maintain DNA methylation until R2. (d) Ten-eleven translocation gene Tet dioxygenases. Tet1 and Tet2 are expressed in PGCs by R2, whereas Tet3 cannot be detected [50]. Dashed line indicates putative levels. Abbreviation: E, embryonic day. from DNA replication, although the precise mechanism is [29,32]. The process of DNA methylation erasure also yet to be elucidated [14]. Reprogramming in PGCs at this initiates a cascade of chromatin remodelling in PGCs at stage results in almost full erasure of DNA methylation by approximately E11.5. This includes a transient reorgani- E13.5 [37], with complete stripping of parental imprints sation of linker histone H1, H3K27me3 and H3K9me3 and and promoter CpG methylation at germline-specific genes stable remodelling of global H3K9ac and H2A/H4R3me2 s 166
  • 4. Review Trends in Genetics April 2012, Vol. 28, No. 4 [14]. Some sequences, including intracisternal A particle replace 5mC during global DNA demethylation in PGCs (IAP) retrotransposons, partially evade DNA demethyla- [50]. tion in PGCs, although the mechanism that protects these Mechanistically, conversion of 5mC to 5hmC could lead elements remains to be determined [31]. Nevertheless, the to unmodified cytosine through several routes in PGCs, erasure of DNA methylation and extensive chromatin including through providing a substrate for base excision reorganisation at approximately E11.5 renders PGCs in repair (BER)-mediated active demethylation [40]. Several a basal epigenetic state that represents an epigenomic recent studies have linked BER components to demethyl- nadir during mammalian development. It is of note that ation during zygotic reprogramming and at specific loci in the apparent biphasic nature of demethylation in PGCs (at somatic contexts [50–52]. As BER components are also approximately E8.0 and approximately E11.5) may poten- upregulated in PGCs relative to somatic neighbours during tially represent a continuous process of 5mC erasure. In epigenetic erasure, genome-wide demethylation in PGCs any case, reprogramming of the PGC genome as a whole is may also occur, at least partially through BER. Indeed, the distinguished from reprogramming during zygotic devel- presence of chromatin-associated XRCC1 and active poly[- opment, where imprints, DNA methylation at multiple loci ADP-ribose] polymerase 1 (PARP1) in PGCs at approxi- and polycomb-based chromatin modifications remain; this mately E11.5, which signify single-strand DNA breaks implies that reprogramming in PGCs represents a more associated with BER, further support this possibility [50]. extensive process [32]. Although both reprogramming One potential involvement of BER-mediated DNA de- events probably share some mechanistic similarities, it methylation in PGCs is that TET-generated 5hmC is further is also likely that both employ unique systems to achieve processed by deamination to 5-hydroxymethyluridine distinct levels of epigenomic resetting. (5hmU) by the activation-induced cytidine deaminase/apo- lipoprotein B mRNA-editing, enzyme-catalytic, polypeptide Erasure of DNA methylation and cellular identity (AID/APOBEC) family of deaminases, and subsequently DNA methylation (5mC) within a CpG context is a highly excised by a glycosylase and repaired to unmodified C. heritable epigenetic mark that is associated with tran- Consistent with this, loss of AID impairs global demethyla- scriptional repression and that contributes to stable line- tion in PGCs by E13.5, indicating that 5mC erasure in PGCs age commitment [38–41]. In this respect, global erasure of depends, at least in part, on AID [37]. Indeed, AID also DNA methylation during PGC development is a fundamen- enhances locus-specific active demethylation mediated by tal event towards the acquisition of totipotency. The ca- TET1 in somatic cells and has been reported to interact with pacity of global DNA demethylation to alter cellular thymine DNA glycosylase (TDG), which can excise the identity, for example in the course of derivation of induced deamination product 5hmU [52,53]. Moreover, AID has pluripotent stem cells (iPS) from somatic cells, makes been proposed to be required for DNA demethylation of unravelling the mechanisms that mediate this process of octamer-binding transcription factor 4 (Oct4) and Nanog great importance [42,43]. Although extensive studies have during somatic cell reprogramming induced by heterokary- established how and where 5mC is and can be introduced, on formation with ES cells as well as in demethylation in it remains enigmatic precisely how DNA methylation is zebrafish embryos [54,55]. However, murine PGCs still removed from the genome [44]. The global DNA demethyl- undergo extensive demethylation by E13.5 in the absence ation during PGC development represents a unique in vivo of AID; global 5mC is reduced to 22% and 20% in male and event in that it results in near-complete stripping of CpG female mutant PGCs, respectively, as opposed to 16% and methylation at almost all genomic loci, including imprints. 8% in wild-type PGCs and compared with approximately The magnitude of DNA demethylation makes PGCs an 74% in E13.5 embryonic soma. Moreover, AID-mutant mice unparalleled system to understand the process of 5mC are both viable and fertile, as are APOBEC 1- and APOBEC erasure in an in vivo context. 2/3-null mice [56–58]. It is also unclear whether AID is expressed to any significant degree in PGCs at the time of Active demethylation in PGCs gross demethylation (approximately E11.5), as it has only The recent identification of three 5mC-dioxygenases [ten- been detected from E12.5 in PGCs, suggesting that AID has eleven translocation gene 1, 2 and 3 (TET1, TET2 and a role in PGC demethylation either after global 5mC erasure TET3)], which can convert 5mC to 5-hydroxymethylcyto- (approximately E12.5) or perhaps at an earlier stage (ap- sine, presented a potential solution to the longstanding proximately E8.5), when the expression of AID is unknown debate regarding the mechanism of DNA demethylation [50,59]. Likewise, TDG is not detectable in PGCs between [45,46]. Conversion of 5mC to 5hmC enables several alter- E10.5 and E13.5, as judged by immunofluorescence studies native but partially overlapping routes to generate an [50]. However, TDG-null PGCs accumulate biallelic CpG unmodified cytosine (C) residue independently of, or de- methylation by E11.0 at the insulin-like growth factor 2 pendent on, DNA replication (Figure 2). In the zygote, the receptor (Igf2r)-imprinted differentially methylated region rapid loss of 5mC from the male pronucleus correlates with (DMR), suggesting that TDG has a role in maintaining a concomitant gain in 5hmC, implying that 5mC is con- a methylation-free state at this locus, presumably by de- verted to 5hmC in this context and has a fundamental role methylation, although this event may occur prior to PGC in reprogramming [47,48]. Indeed, loss of TET3, which is specification [52]. Although further clarification of TDG the only 5mC-dioxigenase significantly expressed in function and expression in PGCs is necessary, its putative zygotes, led to a failure to erase 5mC and neonatal lethality absence at E11.5 argues against a direct AID- and/or TDG- [49]. Although TET3 is not detectable in PGCs, TET1 and mediated active demethylation reaction in PGCs, as has TET2 are present in these cells, suggesting that 5hmC may been reported in other contexts [52]. Therefore, alternative 167
  • 5. Review Trends in Genetics April 2012, Vol. 28, No. 4 (a) C OBE 5mC / AP D AI TET1/2 C T BE 5hmC APO D/ TET1/2 AI 5hmU 5fC TET1/2 5caC ? BE ive R ss Pa (b) C 5mC Relative level Pa ss ive Ac ti ve 5hmC No conversion E5.5 E6.5 E8.0 E10.5 E11.5 Spec R1 R2 TRENDS in Genetics Figure 2. Multiple parallel mechanisms of DNA demethylation. (a) Methylated cytosine (5mC) can be demethylated through several overlapping pathways, including passive and active mechanisms, which may occur in parallel. Passive demethylation (right) can occur through direct replication-dependent depletion of 5mC owing to an absence or reduction of DNA methyltransferase activity. Alternatively, TET oncogene (TET) proteins can catalyse oxidation of 5mC to 5-hydroxymethylcytosine (5hmC) or further conversion to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC), which may all lead to passive demethylation. Active erasure of 5mC (left) can occur through deamination of either 5mC or 5hmC to thymidine (T) or 5-hydroxymethyluridine (5hmU), respectively, which can act as a substrate for base excision repair (BER) to unmodified C. Further conversion of 5hmC to 5fC or 5caC may also be actively removed by BER or 5caC can putatively be removed via a direct decarboxylation reaction (centre). (b) Putative temporal pattern of 5mC conversion to 5hmC during primordial germ cell (PGC) development. The depletion of the 5mC signal at R1 may occur as a result of conversion to 5hmC or through other mechanisms. Similarly, 5mC may be converted to 5hmC at R2, which may subsequently be removed through passive and/or active mechanisms. Abbreviations: AID/APOBEC, activation-induced cytidine deaminase/apolipoprotein B mRNA-editing, enzyme-catalytic, polypeptide; R1 and R2, the two sequential phases of reprogramming during migration (R1) and subsequent to entry into the genital ridge (R2); Spec. specification. 168
  • 6. Review Trends in Genetics April 2012, Vol. 28, No. 4 deaminase–glycosylase complexes may operate down- 5hmU or further oxidation to 5fC and 5caC, all of which are stream of 5hmC (or 5mC) in PGCs. Investigation of potential BER substrates, ultimately leading to repair-driven de- candidates, such as other members of the APOBEC family, methylation. and methyl-CpG-binding domain protein 4 (MBD4), single- strand selective monofunctional uracil DNA glycosylase Passive demethylation in PGCs (SMUG1), nth endonuclease III-like 1 (NTH1) or nei endo- There is accumulating evidence that genome-wide DNA nuclease VIII-like 1 (NEIL1) glycosylases, may shed light on demethylation events include at least a partial ‘passive’ the issue. component. Conversion of 5mC to 5hmC in the zygote has An alternative possibility is that TET-mediated hydro- recently been shown to lead to replication-dependent pas- xymethylation has a deamination-independent role in ac- sive demethylation, rather than to active removal of 5hmC tive demethylation in PGCs, as TET proteins can further or its derivatives [68]. Thus, chromosomes originating in oxidise 5hmC to 5-formylcytosine (5fC) and subsequently to the paternal pronucleus retain 5hmC through successive 5-carboxylcytosine (5caC) [60,61]. Interestingly, these new cleavage divisions, whereas newly synthesised sister chro- C derivatives could also be targets for BER excision [60]. matids are devoid of 5mC and 5hmC, leading to a progres- Indeed, 5fC and 5caC are substrates for TDG, and poten- sive dilution of DNA modifications during development. tially other glycosylases [60,62]. 5caC could also theoreti- The passive loss of DNA modification is consistent with the cally be ‘actively’ removed from the genome by a BER- fact that the human maintenance DNA (cytosine-5)- independent pathway that would involve decarboxylation methyltransferase 1 (DNMT1) cannot recognise 5hmC by an as yet unknown enzyme. It will be important to and there are no other known 5hmC maintenance mecha- determine the prevalence of 5fC and 5caC in PGCs during nisms (although notably UHRF1 does efficiently recognise the key stages of reprogramming to establish whether they 5hmC) [69,70]. Nevertheless, bisulfite sequencing has in- contribute to demethylation through any mechanism. A dicated there may be an additional active mechanism in further possibility is that 5hmC is directly targeted for zygotes that is dependent on BER and operates prior to BER-mediated excision by 5hmC-specific glycosylases with- DNA replication, which may target specific genomic land- out the requirement for processing by deamination or fur- marks [51]. Additionally, the high sensitivity of the 5hmC ther oxidation. Indeed, 5hmC glycosylase activity has been antibody may mask partial or locus-specific active erasure reported in calf thymus extract [63]. Taken together, a of 5hmC on the paternally derived pronuclear chromatids mechanism based on BER may play at least a partial role [71]. However, it seems probable that the bulk of paternal in PGC demethylation, although it is currently unclear pronuclear demethylation occurs passively following which protein complexes direct the process or whether there 5hmC conversion. It remains unclear how the maternal is a degree of redundancy. Likewise, it is not clear which pronucleus, which does not undergo 5mC to 5hmC conver- 5hmC or 5mC derivative substrate [5hmC, 5hmU, 5fC, 5caC sion, undergoes concomitant passive demethylation, as or thymidine (T)] might be targeted by the BER machinery DNMT1 is present and sufficient to maintain maternal for active DNA demethylation in PGCs. imprints [68]. Nonetheless, these studies indicate that It is also possible that alternative mechanisms, indepen- conversion of 5mC to 5hmC coupled with passive demeth- dent of 5hmC, contribute to, or drive, active demethylation ylation is feasible and could operate in PGCs. in PGCs. In Arabidopsis thaliana, demethylation occurs via In murine PGCs, although there is an overall loss of direct excision of 5mC by the specific bifunctional DNA DNA methylation over a relatively short period at approx- glycosylase/lyases, DEMETER (DME) and REPRESSOR imately E11.5, it is important to reconsider whether era- OF SILENCING 1 (ROS1), followed by recruitment of sure of imprinted genes can occur over a protracted period BER machinery [64,65]. Although no mammalian orthologs of 2 days [30], which would not be compatible with a single of DME–ROS1 or 5mC-specific glycosylases have been de- genome-wide wave of ‘active’ demethylation model [14]. scribed, it remains a formal possibility that 5mC is directly Indeed, erasure of DNA methylation from imprinted loci excised from the genome in PGCs. Another possibility is that and repeat elements in porcine PGCs occurs over an ex- 5mC is deaminated directly to T followed by excision by a tended period of up to 20 days, which suggests that ‘active’ glycosylase that recognises the T–G mismatch, such as TDG DNA demethylation does not apply in all mammalian or MBD4, and repaired to unmodified cytosine by BER. It is PGCs, possibly including those in mice [72]. Similarly to additionally possible that a demethylation route based on zygotes, conversion of 5mC to 5hmC (or 5fC and 5caC) in radical sterile alpha motif (SAM) enzymes operates, al- PGCs would facilitate passive replication-dependent de- though experimental evidence for this is lacking in PGCs methylation (Figure 2). Such a conversion of 5mC to 5hmC [66]. Similarly, active 5mC erasure has been reported to be would account for the dramatic loss of 5mC staining ob- linked to the nucleotide excision repair (NER) pathway, and served in PGCs at E11.5 and the protracted nature of this could contribute to DNA demethylation during PGC imprint erasure, given that bisulfite conversion would migration (at approximately E8.5–E10.5) [67]. However, it identify hydroxymethylated loci as methylated but 5mC remains unclear whether such a mechanism could operate antibodies would not [14,30,33]. Interestingly, conversion in PGCs at approximately E11.5 owing to the absence of of 5mC to 5hmC may also skew bisulfite sequencing several key NER components between E10.5 and E12.5 [50]. results, as polymerases appear to amplify unmethylated Thus, the consensus of available data points towards a alleles preferentially over 5hmC-modified alleles after bi- putative active DNA demethylation mechanism in PGCs sulfite treatment, which may falsely imply demethylation at approximately E11.5 being based on initial TET-mediat- has occurred rather than conversion to 5hmC [73]. Addi- ed hydroxylation of 5mC and either deamination towards tionally, TET-mediated conversion to 5hmC may proceed 169
  • 7. Review Trends in Genetics April 2012, Vol. 28, No. 4 to 5caC in PGCs, which is read as unmethylated by bisul- [49]. This suggests that rapid epigenetic reprogramming is fite sequencing, erroneously indicating the occurrence of involved in diminishing developmental barriers that some active demethylation [60]. Although functional conse- cells can nevertheless still overcome in a stochastic man- quences of 5hmC, 5caC or other C derivatives are as yet ner. Similarly, Tet1-null mice sire reduced litter sizes and unclear, the inherent biases and lack of derivative speci- mutant pups have a smaller body size at birth, potentially ficity of bisulfite sequencing mean that observing an owing to a delay in overcoming epigenetically imposed unmethylated allele via this technique does not necessarily barriers [77]. The presence of multiple means of 5mC signify the presence of unmodified C or the implied active erasure could compensate for the loss of function of Tet1 demethylation. As such, conversion to 5hmC or down- through the use of alternative routes in a stochastic man- stream C derivatives could at least partially account for ner. For example, given that key maintenance-methylation the apparent active ‘replication-independent’ demethyla- enzymes, including UHRF1, are absent in PGCs, it is tion that has been reported in PGCs. possible that direct passive demethylation of 5mC can An alternative to passive erasure through 5hmC con- partly offset the absence of any putative 5hmC-mediated version is that PGCs undergo direct passive dilution of mechanisms, a possibility that might also affect interpre- 5mC, based on an absence or exclusion of maintenance tation of the demethylation that still occurs in AID-defi- components, such as DNMT1 or UHRF1. As the doubling cient PGCs by E13.5 [7,37]. time of PGCs at this stage is approximately 16 hr [13,74], In addition to contributing to a robust system, parallel direct replication-dependent dilution could account for a mechanisms of 5mC erasure could function to target spe- significant reduction in global 5mC in PGCs between E10.5 cific genomic loci in PGCs. For example, passive demeth- and E11.5. Because the 5mC antibody (33D3) exhibits ylation could account for the bulk of demethylation, relatively weak avidity (e.g. as compared to the 5hmC analogously to the zygote, whereas erasure of imprinted antibody [71]) and 5mC levels at E10.5 may already be regions may be targeted by specific active mechanisms that partially depleted relative to somatic neighbours, this are absent in the zygote, perhaps based on BER. Interest- passive reduction of 5mC may appear as a largely complete ingly, targeting specific loci with distinct demethylation and, therefore, active erasure of global DNA methylation, mechanisms may also lead to particular functional out- while there would also be a significant loss of methylation comes, perhaps based on the specific C derivative (5hmC, by bisulfite sequencing. Although it is not clear precisely 5fC, 5caC, etc.) that directed demethylation. This could how a passive demethylation mechanism might be trig- have an important functional role during meiosis, similarly gered, the existence of such a system would circumvent the to the role of inherited histone modifications at develop- potential genetic damage that may result from genome- mental loci in the zygote [81]. Active DNA demethylation wide BER. mechanisms could also operate in parallel to a passive system to initiate DNA repair-driven chromatin remodel- Parallel mechanisms of epigenetic reprogramming ling [14]. Notably, other phases of epigenetic reprogram- Although cumulative evidence indicates that several mo- ming in PGCs also utilise parallel systems. For example, lecular pathways of DNA demethylation may operate, erasure of H3K9me2 in early PGCs may occur through the precise mechanism(s) that mediate the comprehen- both downregulation of the methyltransferase GLP and sive methylation erasure in PGCs remain to be clarified upregulation of specific lysine demethylases at approxi- [44,75,76]. Indeed, DNA demethylation and chromatin mately E8.5, which potentially contribute to erasure of this remodelling in PGCs may occur through several comple- modification in a redundant manner. Similarly, the histone mentary parallel pathways, including active and passive replacement that is associated with reorganisation of glob- systems, which would provide a degree of redundancy al H3K27me3 and H3K9me3 marks in PGCs at approxi- and confer robustness and flexibility to the programme mately E11.5 also occurs in parallel with upregulation of (Figure 3). This seems necessary because of the funda- specific lysine demethylases, which may contribute to the mental importance of epigenetic reprogramming to germ efficient erasure. Indeed, both Tet1 and Tet2 are also cell development. Furthermore, there are very limited upregulated at this stage, indicating a possible parallel numbers of PGCs and most, if not all, of them must role [50]. Thus, we propose that the unique importance of transit through the process efficiently and prior to the reprogramming in PGCs necessitates a system of multiple entry of female germ cells into meiosis shortly after epigenetic erasure mechanisms to confer efficiency, fidelity reprogramming. and robustness to the process. A robust system based on multiple parallel mechanisms and inbuilt redundancy may account for the observation Potential roles of epigenetic reprogramming in PGCs that Tet1-null and Tet2-null mice are viable and fertile, The fundamental role of epigenetic reprogramming in despite a probable role for 5hmC in demethylation in PGCs PGCs is to overcome multiple epigenomic barriers to the [77–80]. Although functional redundancy coupled with eventual acquisition of totipotency acquired by epiblast genetic background effects may explain this observation, cells during early development. This is necessary because it is also possible that impeded DNA demethylation in mammals utilise an inductive mechanism of germ cell TET-deficient PGCs would not cause complete loss of the specification (they specify the germline from cells primed cells but rather a reduction in their numbers. In support of towards a somatic fate) rather than acquire germ cell fate this, impeded demethylation of the paternal genome in through an inherited germplasm. This essential role of TET3-deficient zygotes is only associated with a severe reprogramming in mammals is evident in mice lacking the developmental phenotype in a subset of mutant embryos transcriptional regulator PRDM14. Nascent PGCs in 170
  • 8. Review Trends in Genetics April 2012, Vol. 28, No. 4 t 3 a / b , U h r f 1 d o wn r e Dnm gu l a t io n T ET 1 / 2 a c t i v i t y Deamination 5mC 5mC 5mC Me e Pa s M sive Ac BE R Ac e Me M Reprogramming DNA demethyla M e Ac Primed state tion H M T/ Histo HDM HA ne e /HD Td xch AC ow an nr up eg ge reg ul ati ula on on ti His ton em ark r eprog M Ac ramming e Progress ion toward Basal sta s te TRENDS in Genetics Figure 3. Parallel routes towards the basal epigenetic state. Post-implantation epiblast cells acquire epigenetic modifications that constitute a barrier against their reversion to a pluripotent state but remain primed to respond to signals that specify a primordial germ cell (PGC) fate. Upon specification, PGCs embark on a process of reprogramming that overcomes the epigenetic barrier and establishes a basal epigenetic environment that underpins totipotency. Multiple parallel mechanisms contribute to epigenetic reprogramming in PGCs to ensure robustness and redundancy to the process. Global erasure of DNA methylation (upper routes) can occur through passive or active mechanisms directed through several interconnected processes. Likewise, reorganisation of chromatin architecture (lower routes) can occur through the parallel upregulation of histone demethylases (HDM)/deacetylases (HDAC), downregulation of chromatin-modifying enzymes, including histone methyltransferases (HMT)/ acetyltransferases (HAT), and dynamic histone exchange. Abbreviations: 5mC, methylated cytosine; Ac, acetylation; BER, base excision repair; Dnmt3a/b, DNA (cytosine-5)- methyltransferase 3a/b; Me, methylation; TET, ten-eleven translocation gene; Uhrf1, ubiquitin-like, containing PHD and RING finger domains 1. Prdm14-mutant mice fail to undergo early reprogramming RNA Xist necessary to complete the process [18,82]. The of chromatin and DNA methylation or to activate the germ wave of global DNA demethylation in PGCs at approxi- cell genetic programme and consequently exhibit severely mately E11.5 seems to be a trigger for several other impaired PGC development and are sterile [8]. Evidence important processes, including the erasure of parental shows that PGC specification and epigenetic reprogram- imprints to enable establishment of new methylation ming are intimately linked. Furthermore, within the broad marks according to the sex of the embryo [17,29]. The remit of establishing a state of ‘underlying totipotency’, significant impact that aberrant imprints have on mam- epigenomic reprogramming has many distinct roles that malian development, foetal growth and behaviour, and in collectively contribute to a continuous process of PGC human disease, has led to the suggestion that the whole development (Figure 4). process of genome-wide demethylation in PGCs reflects a An important event driven by reprogramming is reacti- byproduct of the necessity to reset locus-specific imprints. vation of Xi in female PGCs. This requires the integration Indeed, as imprinted loci are resistant to demethylation of multiple genetic and epigenetic systems; with early during zygotic reprogramming [82], they may have intrin- chromatin reorganisation initiating X-reactivation and sic or epigenetic properties that preclude erasure except DNA demethylation and repression of the long noncoding under exceptional cellular conditions, such as exists in 171
  • 9. Review Trends in Genetics April 2012, Vol. 28, No. 4 (a) Removal of epigenetic barriers to totipotency (b) X-chromosome reactivation (c) Epimutation erasure (d) Germline gene reactivation (e) Imprint erasure (f) Retrotransposon reactivation (g) Setting for meiosis R1 R2 ~E7.75–9.0 ~E10.5–12.5 TRENDS in Genetics Figure 4. Potential functions of epigenetic reprogramming in primordial germ cells (PGCs). The distinct roles of epigenetic reprogramming in PGCs and the time point at which each occurs (rectangles). (a) Epigenetic reprogramming events during PGC migration (R1) and post-migration (R2) overcomes the layers of epigenetic modifications acquired by epiblast cells, which form a barrier to acquisition of totipotency. (b) In female cells, reprogramming contributes to X-chromosome reactivation. (c) Erasure of epigenetic marks in PGCs also prevents perpetuation of epimutations through the germline. (d) Global DNA demethylation at R2 triggers activation of stably silenced genes necessary for germ cell development (e) A fundamental role of reprogramming is to erase parent-of-origin imprints, thereby enabling subsequent sex-specific methylation marks to be established during gametogenesis (f) Erasure of methylated cytosine (5mC) may permit transient expression of retrotransposons, which are subsequently targeted for stable transcriptional repression by small RNAs, thereby ensuring that potentially harmful genetic elements are strongly silenced in the germline. (g) Reprogramming in PGCs contributes to establishing a chromatin and transcriptional environment that is primed for entry into meiosis. Abbreviation: E, embryonic day. PGCs. Global DNA demethylation in PGCs has other transient activation of TEs during reprogramming important functions, including the activation of stably enables silencing mechanisms to target repressive chro- silenced genes that are required for germ cell development matin, possibly directed by small RNAs derived from [32,83]. Genes such as deleted in azoospermia-like (Dazl) expressed TEs [86]. This putative mechanism would en- and synaptonemal complex protein 3 (Sycp3) are robustly sure that harmful genetic elements that may have escaped silenced by methylated CpG-dense promoters during post- repressive mechanisms are rendered transcriptionally implantation development, potentially to prevent ectopic quiescent throughout germ cell maturation. Notably, activation that may drive malignant tumour growth, but the transient erasure of the PGC epigenome may also are activated by demethylation specifically in the germline provide an opportunity for epigenetic writers to access [32,39,84,85]. Another key role of reprogramming in PGCs chromatin and establish new modifications that direct is to erase aberrant epigenetic information or epimuta- events that are crucial for meiosis. Indeed, the absence tions. Erasure of such modifications in the zygote and of some chromatin-modifying proteins, including G9a, particularly in PGCs prevents their inheritance and per- MLL2, SUV39H1, SUV39H2, LSH and PRDM9, only petuation through successive generations with potentially manifests in germ cells during meiosis [87–91]. One in- detrimental effects. triguing possibility is that epigenetic erasure is necessary Global DNA demethylation may generate an epigenetic to promote chiasmata formation at recombination hot- environment susceptible to expression of harmful trans- spots during meiosis. Here, reprogramming may enable posable elements (TE), yet paradoxically it is possible that the lysine methylase PRDM9 access to target H3K4me3 172
  • 10. Review Trends in Genetics April 2012, Vol. 28, No. 4 marks specifically to 13-mer recognition sites, which act as 16 Shen, X. et al. (2008) EZH1 mediates methylation on histone H3 lysine 27 and complements EZH2 in maintaining stem cell identity and beacons for recombination [92–95]. Thus, it is probable executing pluripotency. Mol. Cell 32, 491–502 that functionally, epigenetic reprogramming in PGCs is a 17 Sasaki, H. and Matsui, Y. (2008) Epigenetic events in mammalian protracted event (E7.75–E12.5) that contributes to myriad germ-cell development: reprogramming and beyond. Nat. Rev. Genet. 9, interconnected processes that are collectively essential for 129–140 germ cell potency and development. 18 Chuva de Sousa Lopes, S.M. et al. (2008) X chromosome activity in mouse XX primordial germ cells. PLoS Genet. 4, e30 19 Tada, T. et al. (1998) Epigenotype switching of imprintable loci in Concluding remarks embryonic germ cells. Dev. Genes Evol. 207, 551–561 Considerable advances have been made in understanding 20 Matsui, Y. et al. (1992) Derivation of pluripotential embryonic stem how new epigenetic information can be introduced, but less cells from murine primordial germ cells in culture. Cell 70, 841–847 is known about the mechanisms that can erase existing 21 Ancelin, K. et al. (2006) Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nat. Cell Biol. 8, modifications. Studies on PGCs provide an unprecedented 623–630 opportunity to unravel the role of key factors and their 22 Tee, W.W. et al. (2010) Prmt5 is essential for early mouse development combined roles in resetting the epigenome. Fundamental and acts in the cytoplasm to maintain ES cell pluripotency. Genes Dev. knowledge gained from these studies may potentially find 24, 2772–2777 wider application. Somatic cells are prone to epimutations 23 Bostick, M. et al. (2007) UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764 through ageing and environmental factors, whereas the 24 Sharif, J. et al. (2007) The SRA protein Np95 mediates epigenetic germ line can reset the epigenome with the potential to inheritance by recruiting Dnmt1 to methylated DNA. Nature 450, ‘rejuvenate’ adult cells. Insights from research on early 908–912 germ cells may provide knowledge and tools for applica- 25 Kaneda, M. et al. (2004) Essential role for de novo DNA methyltransferase Dnmt3a in paternal and maternal imprinting. tions in ageing-related diseases and generally improve Nature 429, 900–903 ability to manipulate cell fates for applications in repro- 26 Kato, Y. et al. (2007) Role of the Dnmt3 family in de novo methylation of ductive and regenerative medicine. imprinted and repetitive sequences during male germ cell development in the mouse. Hum. Mol. Genet. 16, 2272–2280 27 Tachibana, M. et al. (2008) G9a/GLP complexes independently mediate Acknowledgements H3K9 and DNA methylation to silence transcription. EMBO J. 27, We would like to thank Roopsha Sengupta and Harry Leitch for critical 2681–2690 reading of the manuscript and to apologise to the authors whose work 28 Dong, K.B. et al. (2008) DNA methylation in ES cells requires the lysine could not be cited here owing to space constraints. JAH was funded by a methyltransferase G9a but not its catalytic activity. EMBO J. 27, Wellcome Trust Grant and JJZ is the recipient of a Wellcome Trust PhD 2691–2701 Scholarship. MAS is supported by The Wellcome Trust (RG49135). 29 Hajkova, P. et al. (2002) Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15–23 References 30 Lee, J. et al. (2002) Erasing genomic imprinting memory in mouse clone 1 Surani, M.A. et al. (2007) Genetic and epigenetic regulators of embryos produced from day 11.5 primordial germ cells. Development pluripotency. Cell 128, 747–762 129, 1807–1817 2 Ginsburg, M. et al. (1990) Primordial germ cells in the mouse embryo 31 Lane, N. et al. (2003) Resistance of IAPs to methylation reprogramming during gastrulation. Development 110, 521–528 may provide a mechanism for epigenetic inheritance in the mouse. 3 McLaren, A. (1984) Meiosis and differentiation of mouse germ cells. Genesis 35, 88–93 Symp. Soc. Exp. Biol. 38, 7–23 32 Maatouk, D.M. et al. (2006) DNA methylation is a primary mechanism 4 Kocer, A. et al. (2009) Germ cell sex determination in mammals. Mol. for silencing postmigratory primordial germ cell genes in both germ cell Hum. Reprod. 15, 205–213 and somatic cell lineages. Development 133, 3411–3418 5 Saitou, M. et al. (2002) A molecular programme for the specification of 33 Nestor, C. et al. (2010) Enzymatic approaches and bisulfite germ cell fate in mice. Nature 418, 293–300 sequencing cannot distinguish between 5-methylcytosine and 5- 6 Yabuta, Y. et al. (2006) Gene expression dynamics during germline hydroxymethylcytosine in DNA. Biotechniques 48, 317–319 specification in mice identified by quantitative single-cell gene 34 Pichler, G. et al. (2011) Cooperative DNA and histone binding by Uhrf2 expression profiling. Biol. Reprod. 75, 705–716 links the two major repressive epigenetic pathways. J. Cell. Biochem. 7 Kurimoto, K. et al. (2008) Complex genome-wide transcription 112, 2585–2593 dynamics orchestrated by Blimp1 for the specification of the germ 35 West, J.A. et al. (2009) A role for Lin28 in primordial germ-cell cell lineage in mice. Genes Dev. 22, 1617–1635 development and germ-cell malignancy. Nature 460, 909–913 8 Yamaji, M. et al. (2008) Critical function of Prdm14 for the establishment 36 Hayashi, K. et al. (2011) Reconstitution of the mouse germ cell of the germ cell lineage in mice. Nat. Genet. 40, 1016–1022 specification pathway in culture by pluripotent stem cells. Cell 146, 9 Ohinata, Y. et al. (2009) A signaling principle for the specification of the 519–532 germ cell lineage in mice. Cell 137, 571–584 37 Popp, C. et al. (2010) Genome-wide erasure of DNA methylation in 10 McLaren, A. and Lawson, K.A. (2005) How is the mouse germ-cell mouse primordial germ cells is affected by AID deficiency. Nature 463, lineage established? Differentiation 73, 435–437 1101–1105 11 Seki, Y. et al. (2005) Extensive and orderly reprogramming of genome- 38 Mohn, F. et al. (2008) Lineage-specific polycomb targets and de novo wide chromatin modifications associated with specification and early DNA methylation define restriction and potential of neuronal development of germ cells in mice. Dev. Biol. 278, 440–458 progenitors. Mol. Cell 30, 755–766 12 Bao, S. et al. (2009) Epigenetic reversion of post-implantation epiblast 39 Borgel, J. et al. (2010) Targets and dynamics of promoter DNA to pluripotent embryonic stem cells. Nature 461, 1292–1295 methylation during early mouse development. Nat. Genet. 42, 13 Seki, Y. et al. (2007) Cellular dynamics associated with the genome- 1093–1100 wide epigenetic reprogramming in migrating primordial germ cells in 40 Wu, S.C. and Zhang, Y. (2010) Active DNA demethylation: many roads mice. Development 134, 2627–2638 lead to Rome. Nat. Rev. Mol. Cell Biol. 11, 607–620 14 Hajkova, P. et al. (2008) Chromatin dynamics during epigenetic 41 Hodges, E. et al. (2011) Directional DNA methylation changes and reprogramming in the mouse germ line. Nature 452, 877–881 complex intermediate states accompany lineage specificity in the adult 15 Tachibana, M. et al. (2005) Histone methyltransferases G9a and GLP hematopoietic compartment. Mol. Cell 44, 17–28 form heteromeric complexes and are both crucial for methylation of 42 Mikkelsen, T.S. et al. (2008) Dissecting direct reprogramming through euchromatin at H3-K9. Genes Dev. 19, 815–826 integrative genomic analysis. Nature 454, 49–55 173
  • 11. Review Trends in Genetics April 2012, Vol. 28, No. 4 43 Surani, M.A. and Hajkova, P. (2010) Epigenetic reprogramming of 69 Valinluck, V. and Sowers, L.C. (2007) Endogenous cytosine damage mouse germ cells toward totipotency. Cold Spring Harb. Symp. Quant. products alter the site selectivity of human DNA maintenance Biol. 75, 211–218 methyltransferase DNMT1. Cancer Res. 67, 946–950 44 Ooi, S.K. and Bestor, T.H. (2008) The colorful history of active DNA 70 Frauer, C. et al. (2011) Recognition of 5-hydroxymethylcytosine by the demethylation. Cell 133, 1145–1148 Uhrf1 SRA domain. PLoS ONE 6, e21306 45 Tahiliani, M. et al. (2009) Conversion of 5-methylcytosine to 5- 71 Ficz, G. et al. (2011) Dynamic regulation of 5-hydroxymethylcytosine in hydroxymethylcytosine in mammalian DNA by MLL partner TET1. mouse ES cells and during differentiation. Nature 473, 398–402 Science 324, 930–935 72 Hyldig, S.M. et al. (2011) Epigenetic reprogramming in the porcine 46 Ito, S. et al. (2010) Role of Tet proteins in 5mC to 5hmC conversion, germ line. BMC Dev. Biol. 11, 11 ES-cell self-renewal and inner cell mass specification. Nature 466, 73 Huang, Y. et al. (2010) The behaviour of 5-hydroxymethylcytosine in 1129–1133 bisulfite sequencing. PLoS ONE 5, e8888 47 Wossidlo, M. et al. (2011) 5-Hydroxymethylcytosine in the mammalian 74 Tam, P.P. and Snow, M.H. (1981) Proliferation and migration of zygote is linked with epigenetic reprogramming. Nat. Commun. 2, 241 primordial germ cells during compensatory growth in mouse 48 Iqbal, K. et al. (2011) Reprogramming of the paternal genome upon embryos. J. Embryol. Exp. Morphol. 64, 133–147 fertilization involves genome-wide oxidation of 5-methylcytosine. Proc. 75 Bhutani, N. et al. (2011) DNA demethylation dynamics. Cell 146, Natl. Acad. Sci. U.S.A. 108, 3642–3647 866–872 49 Gu, T.P. et al. (2011) The role of Tet3 DNA dioxygenase in epigenetic 76 Law, J.A. and Jacobsen, S.E. (2010) Establishing, maintaining and reprogramming by oocytes. Nature 477, 606–610 modifying DNA methylation patterns in plants and animals. Nat. Rev. 50 Hajkova, P. et al. (2010) Genome-wide reprogramming in the mouse Genet. 11, 204–220 germ line entails the base excision repair pathway. Science 329, 78 77 Dawlaty, M.M. et al. (2011) Tet1 is dispensable for maintaining 51 Wossidlo, M. et al. (2010) Dynamic link of DNA demethylation, DNA pluripotency and its loss is compatible with embryonic and strand breaks and repair in mouse zygotes. EMBO J. 29, 1877–1888 postnatal development. Cell Stem Cell 9, 166–175 52 Cortellino, S. et al. (2011) Thymine DNA glycosylase is essential for 78 Li, Z. et al. (2011) Deletion of Tet2 in mice leads to dysregulated active DNA demethylation by linked deamination-base excision repair. hematopoietic stem cells and subsequent development of myeloid Cell 146, 67–79 malignancies. Blood 118, 4509–4518 53 Guo, J.U. et al. (2011) Hydroxylation of 5-methylcytosine by TET1 79 Moran-Crusio, K. et al. (2011) Tet2 loss leads to increased promotes active DNA demethylation in the adult brain. Cell 145, hematopoietic stem cell self-renewal and myeloid transformation. 423–434 Cancer Cell 20, 11–24 54 Bhutani, N. et al. (2010) Reprogramming towards pluripotency 80 Quivoron, C. et al. (2011) TET2 inactivation results in pleiotropic requires AID-dependent DNA demethylation. Nature 463, 1042–1047 hematopoietic abnormalities in mouse and is a recurrent event 55 Rai, K. et al. (2008) DNA demethylation in zebrafish involves the during human lymphomagenesis. Cancer Cell 20, 25–38 coupling of a deaminase, a glycosylase, and gadd45. Cell 135, 1201–1212 81 Hammoud, S.S. et al. (2009) Distinctive chromatin in human sperm 56 Muramatsu, M. et al. (2000) Class switch recombination and packages genes for embryo development. Nature 460, 473–478 hypermutation require activation-induced cytidine deaminase (AID), 82 Morgan, H.D. et al. (2005) Epigenetic reprogramming in mammals. a potential RNA editing enzyme. Cell 102, 553–563 Hum. Mol. Genet. 14, R47–R58 (Spec No 1) 57 Morrison, J.R. et al. (1996) Apolipoprotein B RNA editing enzyme- 83 Rodic, N. et al. (2005) DNA methylation is required for silencing deficient mice are viable despite alterations in lipoprotein metabolism. of ant4, an adenine nucleotide translocase selectively expressed Proc. Natl. Acad. Sci. U.S.A. 93, 7154–7159 in mouse embryonic stem cells and germ cells. Stem Cells 23, 58 Mikl, M.C. et al. (2005) Mice deficient in APOBEC2 and APOBEC3. 1314–1323 Mol. Cell. Biol. 25, 7270–7277 84 Janic, A. et al. (2010) Ectopic expression of germline genes drives 59 Morgan, H.D. et al. (2004) Activation-induced cytidine deaminase malignant brain tumor growth in Drosophila. Science 330, 1824–1827 deaminates 5-methylcytosine in DNA and is expressed in 85 Simpson, A.J. et al. (2005) Cancer/testis antigens, gametogenesis and pluripotent tissues. J. Biol. Chem. 279, 52353–52360 cancer. Nat. Rev. Cancer 5, 615–625 60 He, Y.F. et al. (2011) Tet-mediated formation of 5-carboxylcytosine and 86 Seisenberger, S. et al. (2010) Retrotransposons and germ cells: its excision by TDG in mammalian DNA. Science 333, 1303–1307 reproduction, death, and diversity. F1000 Biol. Rep. 2 61 Ito, S. et al. (2011) Tet proteins can convert 5-methylcytosine to 5- 87 Tachibana, M. et al. (2007) Functional dynamics of H3K9 methylation formylcytosine and 5-carboxylcytosine. Science 333, 1300–1303 during meiotic prophase progression. EMBO J. 26, 3346–3359 62 Maiti, A. and Drohat, A.C. (2011) Thymine DNA glycosylase can 88 Peters, A.H. et al. (2001) Loss of the Suv39 h histone rapidly excise 5-formylcytosine and 5–carboxylcytosine: potential methyltransferases impairs mammalian heterochromatin and implications for active demethylation of CpG sites. J. Biol. Chem. genome stability. Cell 107, 323–337 286, 35334–35338 89 Glaser, S. et al. (2009) The histone 3 lysine 4 methyltransferase, Mll2, 63 Cannon, S.V. et al. (1988) 5-Hydroxymethylcytosine DNA glycosylase is only required briefly in development and spermatogenesis. activity in mammalian tissue. Biochem. Biophys. Res. Commun. 151, Epigenetics Chromatin 2, 5 1173–1179 90 De La Fuente, R. et al. (2006) Lsh is required for meiotic chromosome 64 Choi, Y. et al. (2002) DEMETER, a DNA glycosylase domain protein, is synapsis and retrotransposon silencing in female germ cells. Nat. Cell required for endosperm gene imprinting and seed viability in Biol. 8, 1448–1454 arabidopsis. Cell 110, 33–42 91 Kota, S.K. and Feil, R. (2010) Epigenetic transitions in germ cell 65 Gong, Z. et al. (2002) ROS1, a repressor of transcriptional gene development and meiosis. Dev. Cell 19, 675–686 silencing in Arabidopsis, encodes a DNA glycosylase/lyase. Cell 111, 92 Myers, S. et al. (2010) Drive against hotspot motifs in primates 803–814 implicates the PRDM9 gene in meiotic recombination. Science 327, 66 Okada, Y. et al. (2010) A role for the elongator complex in zygotic 876–879 paternal genome demethylation. Nature 463, 554–558 93 Baudat, F. et al. (2010) prdm9 is a major determinant of meiotic 67 Schmitz, K.M. et al. (2009) TAF12 recruits Gadd45a and the nucleotide recombination hotspots in humans and mice. Science 327, 836–840 excision repair complex to the promoter of rRNA genes leading to active 94 Parvanov, E.D. et al. (2010) Prdm9 controls activation of mammalian DNA demethylation. Mol. Cell 33, 344–353 recombination hotspots. Science 327, 835 68 Inoue, A. and Zhang, Y. (2011) Replication-dependent loss of 95 Hayashi, K. et al. (2005) A histone H3 methyltransferase 5-Hydroxymethylcytosine in mouse preimplantation embryos. controls epigenetic events required for meiotic prophase. Nature Science 334, 194 438, 374–378 174