You are on page 1of 16

REVIEWS

TETonic shift: biological roles of


TETproteins in DNA demethylation
and transcription
William A.Pastor1,3, L. Aravind2 and Anjana Rao1
Abstract | In many organisms, the methylation of cytosine in DNA has a key role in silencing
parasitic DNA elements, regulating transcription and establishing cellular identity.
Therecent discovery that ten-eleven translocation (TET) proteins are 5methylcytosine
oxidases has provided several chemically plausible pathways for the reversal of DNA
methylation, thus triggering a paradigm shift in our understanding of how changes in
DNA methylation are coupled to cell differentiation, embryonic development and cancer.

Dioxygenases
Since the initial description of their enzymatic activity in causative parasite of African sleeping sickness in humans18.
Enzymes that catalyse the 2009 (REFS1,2), proteins of the TET family have become Leishmania species, trypanosomes such as T.brucei and
addition of both oxygen atoms a focus of substantial interest. TET proteins are named other kinetoplastids contain a modified thymine known
from molecular oxygen to one after the ten-eleven translocation (t(10;11)(q22;q23)) that as base J (dglucosyl-hydroxymethyluracil)19. JBP1
or two organic substrates.
is found in rare cases of acute myeloid and lymphocytic and JBP2 generate base J by oxidizing the methyl group
DNA demethylation leukaemia. This translocation fuses the mixed-lineage leu- of thymine to yield 5hydroxymethyluracil, and the result-
Here, defined as replacement kaemia 1 (MLL1) gene located on human chromosome 10 ing hydroxyl group is then glucosylated by an unknown
of 5-methylcytosine, the major with the TET1 gene on human chromosome11 (REFS3,4). glucosyltransferase20 (FIG.1b). JBP enzymes are members
methylated base in The three mammalian TET proteins, namely TET1, TET2 of a large family of Fe2+- and 2oxoglutarate-dependent
mammalian DNA, with
unmodified cytosine, either
and TET3, are Fe2+- and 2oxoglutarate-dependent dioxy- dioxygenases21,22; AlkB enzymes, which remove aberrant
directly or through genases that successively oxidize 5methylcytosine (5mC) methylation from damaged DNA bases by an oxidative
intermediates. to 5hydroxymethylcytosine (5hmC), 5formylcytosine mechanism, are distantly related members of the same
(5fC) and 5carboxylcytosine (5caC) in DNA1,2,5,6 (FIG.1a). superfamily 2123. In T.brucei, base J is present at subtelom-
All three forms of oxidized methylcytosine are now known eric repeats, at inactive copies of the variant surface glyco-
1
Sanford Consortium for to be present in numerous mammalian tissues1,5,79. TET protein that is used by the parasite to evade host immune
Regenerative Medicine and proteins have roles in diverse biological processes, includ- defence and at other silenced regions of the genome24.
La Jolla Institute for Allergy ing epigenetic regulation of gene transcription, embry- InLeishmania species, base J was recently shown to
and Immunology, 9420
onic development, stem cell function and cancer, but the restrain elongation of the unique polycistronic transcripts
Athena Circle, La Jolla,
California 92037, USA. mechanisms underlying these roles are still poorly defined. of kinetoplastids beyond transcription stop sites25.
2
National Center for Here, we review our current understanding of TET Computational screens to identify additional homo-
Biotechnology Information, enzymes and their biological functions, focusing on recent logues of JBP enzymes revealed a large family of pre-
National Library of Medicine, studies not covered in previous reviews1014. We discuss the dicted nucleic acid-modifying dioxygenases from diverse
US National Institutes of
Health, Bethesda, Maryland,
established and controversial roles of TET proteins and eukaryotes and bacteriophages, which included the meta-
USA. methylcytosine oxidation products in DNA demethylation, zoan TET enzymes2,18. A gene encoding an enzyme of
3
Present address: gene regulation and embryonic development. The role the TETJBP family entered the common ancestor of the
Department of Molecular, of TET proteins in haematopoietic differentiation and metazoan lineage and fused with a second gene con-
Celland Developmental
oncogenesis has been reviewed elsewhere10,1517; their role taining a CXXC domain (described below), forming the
Biology, University of
California at Los Angeles, in neurons is only briefly discussed here owing to space TET subfamily 2. TET enzymes are present in all meta-
LosAngeles, California, USA. limitations. zoans that have retained cytosine methylation but are
Correspondence to absent in organisms such as Caenorhabditis elegans
W.A.P and A.R. TET proteins are 5mC oxidases in which methylation has been unambiguously lost 2.
e-mails:
wpastor@ucla.edu;
The prediction that TET proteins might have DNA- The coexistence of TET proteins with DNA methylation,
arao@liai.org modifying activity was based on the analysis of Jbinding their association with CXXC domains which frequently
doi:10.1038/nrm3589 protein 1 (JBP1) and JBP2 from Trypanosoma brucei, the bind unmethylated CpG sequences (see below) and the

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 341

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a
Unknown decarboxylase?

TDG and BER

DNMT enzymes?

NH2 NH2 NH2 OH NH2 O NH2 O


DNMT TET protein TET protein TET protein
N N N N N OH

O N O N O N O N O N
R R R R R
Cytosine 5mC 5hmC 5fC 5caC

AID or APOBEC?
O OH

HN

TDG or SMUG1 and BER O N


R
5hmU c
mC G TET protein hmC G
G Cm G Chm
b Trypansoma brucei, other kinetoplastid protozoa Replication Replication
O O OH
UHRF1
HN HN DNMT1 hmC
UHRF1
mCG G
DNMT1
O N O N GC GC
JBPs m
R R m
Thymine 5hmU CG CG
G Cm G Chm DNMT1
DNMT1
Unknown UHRF1
-glucosyltransferase UHRF1
Impaired
Maintenance maintenance
OH
O methylation methylation?
O OH
HN O mC G hmC
G
OH
HO G Cm GC
O N
mC
R G CG
-glucosylhydroxymethyluracil (base J) G Cm G Chm

Figure 1 | Mechanisms of TET-mediated demethylation. a | Known and putative pathways of DNA demethylation
Nature
that involve oxidized methylcytosine intermediates. Ten-eleven translocation (TET) Reviews
proteins | Molecular
sequentially Cell Biology
oxidize
5methylcytosine (5mC) to 5hydroxymethylcytosine (5hmC), 5formylcytosine (5fC) and 5carboxylcytosine (5caC).
5fC and 5caC can be removed by thymine DNA glycosylase (TDG) and replaced by cytosine via base excision repair
(BER), although the extent to which this mechanism operates in specific cell types during development is unknown.
Other proposed mechanisms of demethylation are less well established, including decarboxylation of 5caC,
DNAmethyltransferase (DNMT)-mediated removal of the hydroxymethyl group of 5hmC and deamination of 5hmC
(and 5mC) (see main text) by the cytidine deaminases AID (activation-induced cytidine deaminase) and APOBEC
(apolipoprotein B mRNA editing enzyme, catalytic polypeptide). AID enzymes deaminate cytosine bases in DNA to
yield uracil. AID and the larger family of APOBEC enzymes have been proposed to effect DNA demethylation by
deaminating 5mC and 5hmC in DNA to yield thymine and 5hmU, respectively. As these are present in mismatched T:G
and 5hmU:G basepairs, they have been proposed to be excised by SMUG1 (single-strand-selective monofunctional
uracil DNA glycosylase) or TDG. This mechanism is controversial, however (see main text). b | The mechanism of base J
(dglucosyl-hydroxymethyluracil) biosynthesis. The thymidine oxidation step mediated by J-binding protein 1 (JBP1)
or JBP2, to produce 5hydroxyuracil (5hmU), is analogous to the 5mC oxidation mediated by TET proteins. JBPs are the
founding members of the TETJBP superfamily: the predicted oxygenase domains of JBP1 and JBP2 were used as the
starting point for the sequence profile searches that recovered the homologous domains of the three mammalian TET
proteins. c | Mechanism by which 5hmC could facilitate replication-dependent DNA demethylation. A symmetrically-
methylated CpG sequence is converted during DNA replication into two asymmetrically methylated DNA strands
(leftpanel). Hemimethylated CpG sites are recognized by UHRFI, the obligate partner of the maintenance DNA
methyltransferase DNMT1, which restores symmetrical methylation. TET proteins act at methylated CpG sites to
generate symmetrically hydroxymethylated CpG sequences. 5hmC and other oxizided methylcytosines may impair
maintenance methylation by inhibiting UHRF1 binding, DNMT1 activity, or both (right panel). As a result, the CpG
sequence progressively loses DNA methylation through successive DNA replication cycles.

342 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

chemical similarity of thymine and 5mC oxidation all led Subfamily2consists of the first two CXXC domains of
to the proposal that TET proteins might function as 5mC MBD1; these CXXC domains have not yet been demon-
oxidases and potentially as DNA demethylases2,18. strated to bind DNA and have no documented function36.
Indeed, ectopic expression of TET proteins in cell Subfamily 3 includes the CXXC domains of TET1, TET3,
lines reduces 5mC levels and causes the appearance of IDAX and RINF (retinoid-inducible nuclear factor; also
5hmC, and this activity is abrogated by mutation of the known as CXXC5)2,39. The IDAX CXXC domain preferen-
signature His-Xaa-Asp motif (where Xaa represents tially binds unmethylated CpG sequences in vitro, and is
any amino acid) of these dioxygenases1,26,27 (see below). found at CpG islands and CpG-rich promoters (which are
Recombinant TET catalytic domains and full-length predominantly unmethylated) in cells (Ref. 41). Curiously,
TET proteins efficiently convert 5mC to 5hmC invitro in however, the TET1 CXXC domain is reported not to bind
the presence of the essential cofactors 2oxoglutarate and DNA33 or alternatively to bind CpG sequences regardless
Fe2+ (REFS1,27). TET proteins also produce the further of whether the cytosine is modified39,40, and the TET3
oxidation products 5fC and 5caC5,6,28 (FIG.1a). Thus, the CXXC domain is reported to bind unmodified cyto-
successive actions of DNA methyltransferases (DNMTs) sine irrespective of whether it is followed by a guanine,
and TET proteins produce four distinct cytosine modi- a finding supported by affinity measurements and Xray
fications, bringing the total number of cytosine species crystallography 42. As subfamily 3CXXC domains have
to five (FIG.1a). a strong positive charge that increases their tendency to
5hmC is found at different levels in mammalian bind nonspecifically to DNA, an analysis of binding sites
cells: it is present at 1% of the total level of 5mC in some invivo is needed to reconcile these invitro findings.
immune cell populations26, ~510% of the level of 5mCin Surprisingly, IDAX (a reported inhibitor of WNT
embryonic stem (ES) cells1 and as high as 40% of 5mC signalling 43) targets TET2, the protein from which it
in Purkinje neurons9. Consistently, the highest reported was separated during evolution, for destruction via a
levels of 5hmC are in the brain5,7,8. An early report caspase-dependent mechanism41. Depletion of IDAX
of 5hmC in mammalian DNA29 is questionable, as in this from ES cells prevented the downregulation of TET2 that
study 5mC was not detected in mouse brain and liver normally occurs upon differentiation, whereas its deple-
DNA, whereas the level of 5hmC was unrealistically high tion from a human myeloid cell line increasedTET2 and
(1517% of all cytosines), suggesting massive oxidation 5hmC levels. IDAX cannot negatively regulate TET2 if
of 5mC during the unconventional DNA extraction pro- key DNA-binding residues in the IDAX CXXC domain
cedure devised by this group29. 5fC and 5caC are present are mutated, suggesting that IDAX recruits TET2 to
in mammalian cells at much lower levelsthan 5hmC DNA before the caspase-dependent degradation mecha-
(0.03% and 0.01%, respectively, of the level of 5mC in nism is activated. The implication is that TET2, which
mouse ES cells)5,6,28, at least partly because there are like other TET proteins is likely to also bind DNA
enzymatic mechanisms for their removal. These include through the cysteine-rich region of its catalytic domain
base excision by thymine DNA glycosylase (TDG)6,30 and (see below), binds different sets of genomic regions
possibly decarboxylation of 5caC by unknown enzymes depending on whether IDAX is present. The CXXC
present in ES cell lysates31 (FIG.1a). domain of TET3 negatively regulates TET3 catalytic
activity perhaps through an autoinhibitory mechanism
Domain structure of metazoan TETs that involves a physical interaction between the CXXC
Most metazoan TET proteins contain an aminoterminal and catalytic domains or, alternatively, by tethering
CXXC domain (~60 amino acids) and a carboxyterminal TET3 to particular DNA elements and thus limiting its
catalytic domain (FIG.2a), the core of which adopts a genome-wideactivity.
double-stranded helix (DSBH) fold1,2. Metazoan TET The catalytic domains of TET proteins are character-
proteins can be distinguished from all other members istic of Fe2+- and 2oxoglutarate-dependent dioxygenases
of the TETJBP family by a Cys-rich domain in the (reviewed in REF.22). The DSBH fold, which comprises
Nterminal region of the DSBH domain. In jawed verte the catalytic domain of all these dioxygenases, contains
brates, the TET gene underwent triplication, giving rise to an His-Xaa-Asp/Glu signature motif, a Cterminal con-
TET1, TET2 and TET3. TET2 then underwent a chromo served His residue that is involved in coordinating Fe2+
somal inversion event in which the exon containing the and a conserved Arg residue that binds 2oxoglutarate
CXXC domain CXXC domain was detached and became a separate via a salt bridge21,22. Because a crystal structure of the
A Zn2+-chelating domain
typified by the signature amino
gene that encodes IDAX (inhibition of the Dvl and axin TET catalytic domain is not yet available, the structure
acid sequence CGXCXXC(X)NC, complex; also known as CXXC4)2,32 (FIG.2a, b). of the AlkB DSBH domain and a detailed view of the
in which X represents any CXXC domains occur in many chromatin-associated active site44 are shown in FIG.2c. As is the case for AlkB,
amino acid. CXXC domains in proteins. Three distinct subfamilies of CXXC domains the substrate 5mC is likely to be flipped out of the DNA
metazoans always contain two
can be identified by their sequence33 (Supplementary double helix into the catalytic cavity of the TET protein,
such sequences.
informationS1 (figure)). Subfamily 1 includes the CXXC where it is brought in close proximity to the catalytic
CpG sequences domains of CXXC1 (also known as CFP1), DNMT1, MLL Fe2+ ion (FIG.2c).
Any instance of a cytosine and Lys-specific demethylase (KDMA) family proteins, as The inserted Cys-rich domain in the catalytic region
followed immediately by a well as the third CXXC domain of methyl CpG-binding of TET proteins is likely to chelate two or more Zn2+ ions
guanine on the same strand of
DNA. Most DNA methylation
protein 1 (MBD1); these CXXC domains specifically via nine conserved Cys residues and one His residue, and
in mammals occurs at recognize unmodified CG sequences and target sub- has been postulated to be part of a DNA-binding surface
CpGsites. family 1 proteins to CpG-rich sequences in DNA3438. that might help in target recognition2. Indeed, the same

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 343

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a TET1 Core catalytic region (1,3482,136) b Ancestral animal TET


CXXC Cys-rich insert
(579637) (14181610) Triplication
1 2,136 (jawed vertebrates)
TET1

IDAX TET2 Core catalytic region (1,0572,002) TET2

CXXC Cys-rich insert TET3


(127185) (11291320)
1 198 1 2,002
Inversion
(jawed vertebrates)
TET1
TET3 Core catalytic region (7331,776)
TET2
CXXC Cys-rich insert IDAX
(46102) (805996) TET3
1 1,776

c Structure of AlkB DSBH domain

Location of Cys-rich
insert in TET
Active site of AlkB
Arg210

C
2OG His187 His131

Fe2+
2OG Methyl
adenine DNA
Arg204
N Flipped out
methyladenine
Asp133
Fe2+ Target methyl
group
Location of
low complexity DNA
insert in TET

Figure 2 | Known protein domains of TET family members. a | Ten-eleven translocation (TET) proteins contain a
DNA-binding CXXC domain towards the amino terminus and a carboxyterminal catalytic core region
Nature Reviews that includes
| Molecular Cell Biology
a Cys-rich insert and a larger double-stranded -helix (DSBH) domain. The number of amino acids is indicated, and the
numbering corresponds to the human proteins. b | Evolutionary changes in the domain structure of TET proteins.
Agenetriplication event that occurred in jawed vertebrates resulted in the generation of three TET family members.
Achromosomal inversion then detached the catalytic domain of TET2 from its CXXC domain, which became a separate
gene (which encodes IDAX (inhibition of the Dvl and axin complex)). c | A cartoon representation of AlkB (Protein
Databank (PDB) identifier: 2FD8), a protein that belongs to the same superfamily as the TET proteins and shares a common
fold with them. AlkB is shown as a complex with its substrate methyladenine and its cofactor 2oxoglutarate (2OG). In the
2OG structure, carbon atoms are shown pink, in the rest of the structure carbons are shown in grey. Nitrogens are shown
in blue, oxygens in red and phosphates in orange (left panel). Astripped-down view of the active site of AlkB in complex
with its substrate methlyadenine. Note the series of interactions, including pipi stacking interactions, between the
Hisresidues and the target base, and cationpi interaction with the active site metal. Such interactions are likely to be
retained in the TET J-binding protein (JBP) family. In the 2OG structure, carbon atoms are shown in cyan and oxygens in
red. In the protein structure, carbons are shown in grey, nitrogens in blue and oxygens in red. In methyladenine, carbons
are orange and nitrogens are blue. Dashed yellow lines represent hydrogen bonds (right panel).

region contains comparable insertions in the distantly the histone-binding PHD finger in SMCX (also known
related Jumonji-like family 45 (for example, the AT-rich as KDM5C or JARID1C)). Metazoan TET proteins
interaction domain (ARID; also known as BRIGHT) in also contain a large low-complexity insert predicted to
Jumonji and ARID domain-containing 1 (JARID1) and be on the exterior surface of the DSBHfold2,32(FIG.2c).

344 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

The sequence and size of this unstructured insert varies intermediates that are removed by TDG. However, even
greatly between TET family members, but its contin- in a TDG-deficient cell, 5fC is rare compared with 5mC
ued presence indicates a function, perhaps as a site of (0.20.3% of the total methylcytosines)5,55,56, and 5caC is
proteinprotein interaction2,32. even less abundant6,56,, indicating that this demethylation
pathway has limited throughput in ES cells.
TET-mediated DNA demethylation: mechanisms The second DNA repair-based mechanism has been
Interest in TET proteins has primarily centred around reported by one laboratory in transfected human cells
the possibility that oxidized methylcytosines could serve and in mouse brain52. They proposed that 5hmC is
as intermediates in one or more DNA demethylation deaminated to 5hydroxyuracil (5hmU) by AID and
pathways. In such a pathway, TET would oxidize 5mC to APOBEC family enzymes, removed by SMUG1 (single-
5hmC, 5fC or 5caC, which would then be replaced with strand-selective monofunctional uracil DNA glyco-
cytosine, the net result thus being demethylation. There sylase 1) or TDG glycosylases and ultimately replaced
are at least four mechanisms by which TET proteins by cytosine (FIG.1a). This sequential deamination and
could mediate DNA demethylation (FIG.1a). removal of 5hmC is similar to the deamination of 5mC
and the removal of the resulting T:G mismatches by
Facilitation of passive DNA demethylation. Because TDG, a mechanism previously proposed to occur in
CG sites are palindromes that in general are symmetri- zebrafish57,58, human cancers58, primordial germ cells59
cally methylated46, DNA replication yields two strands and fused cells undergoing reprogramming 60. However,
with hemimethylated CG sites. Maintenance of DNA there is considerable controversy about the signifi-
methylation patterns requires the DNA methyltrans- cance of deamination-based demethylation mecha-
ferase DNMT1 and its obligate partner UHRF1 (REF.47) nisms in general. In support of such mechanisms, TDG
(FIG.1c). UHRF1 binds to hemimethylated CpG sites can excise T:G mismatches and 5hmU:G mismatches
via its SAD/SRA (SET associated Deinococcus domain invitro54,61, and both PGCs derived from Aid-deficient
(SAD)/SET and RING associated (SRA) domain) domain mice and mouse embryonic fibroblasts (MEFs) derived
and recruits DNMT1. DNMT1 then methylates the from Tdg-deficient mice have modestly increased
CG sites on the nascent DNA strand, thus maintaining levels of methylation at some CpG island promoters61,62.
methylation patterns through cell division47,48. In vitro, Arguing against a deamination mechanism, the AID
UHRF1hemi5hmC binding is tenfold less efficient enzyme primarily acts on single-stranded DNA63, and
than UHRF1hemi5mC binding 49. In addition, the recombinant AID and APOBEC enzymes have their
activity of recombinant DNMT1 is reduced 12-fold50 or strongest activity against unmodified cytosine with
50-fold49 at sites of hemi5hmC invitro. Together, these reduced activity against 5mC and no detectable activity
results imply that the TET-mediated hydroxymethylation against 5hmC54,64. It therefore seems unlikely that AID
of a methylated CG site invivo can block maintenance and APOBEC enzymes have a role in 5hmCdependent
methylation during cell division and eliminate 5mC in demethylation pathways, although their involvement in
a passive, replication-dependent manner. However, specific situations cannot be ruledout.
hydroxymethylated plasmids stably transfected into
transformed human cells retain maintenance methyla- Enzymatic decarboxylation of 5caC. There is some evi-
tion through cycles of plasmid division as efficiently as dence that 5caC may be decarboxylated by protein(s)
a methylated plasmid51, suggesting that 5hmC does not present in ES cell lysates. An oligonucleotide containing
completely block maintenance methylation incells. 5caC that was labelled with 15N at both positions of the
pyrimidine ring was synthesized and incubated with
Active DNA demethylation through DNA repair. Two EScell lysate. Following the recovery and digestion
replication-independent (active) demethylation mecha- of the oligonucleotide, a small but detectable quantity of
nisms have been reported to couple the methylcytosine [15N2]-deoxycytosine was detected, indicating direct
oxidase activity of TET proteins with base excision repair conversion of 5caC to cytosine without BER31 (FIG.1a).
(BER). The first, which involves 5fC, 5caC and TDG, The factors that catalyse this decarboxylation reaction
has been confirmed by multiple laboratories6,30,53. The remain to be identified.
second mechanism, which involves AID (activation-
induced cytidine deaminase) and APOBEC (apolipo Dehydroxymethylation by DNMT enzymes. Remarkably,
protein B mRNA editing enzyme, catalytic polypeptide), DNMT enzymes can remove the hydroxymethyl group
is still controversial52,54. In the first mechanism, TET pro- of 5hmC invitro, directly converting 5hmC to cyto-
teins further oxidize 5hmC to generate 5fC and 5caC5,6,28. sine65,66 (FIG.1a). Reducing conditions favour the methyl
Base excision repair 5fC and 5caC can be excised by TDG6,30; their replace- transferase activity of DNMT3A, whereas oxidizing
(BER). A DNA repair pathway
ment with cytosine results in demethylation (FIG.1a). conditions favour dehydroxymethylation66. Whether
in which a DNA base is
removed by a glycosylase Electrophoretic mobility shift assays (EMSAs) and the this reaction occurs in cells is still unknown. Two papers
enzyme and ultimately structure of TDG in complex with 5caC53 indicate that reported cyclical changes in the DNA methylation status
replaced by a new base. TDG binds 5caC:G mismatches with higher affinity than of several oestrogen-responsive genes upon the addition
T:G mismatches (its conventional substrate). Depletion of oestrogen67,68, and one of these also showed cyclical
Primordial germ cells
(PGCs). Precursors of mature
of TDG causes a 210fold increase in the levels of recruitment of all three DNMTs67. It would be interesting
germ cells (egg in female and 5fC and 5caC in EScells6,55,56, consistent with a model to revisit this system to determine whether reciprocal
sperm in male). in which these bases are short-lived demethylation actions of TET proteins and DNMTs were involved.

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 345

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Detecting and mapping modified cytosines by DNA precipitation


caveats apply: 5fC and 5caC will appear as unmodified
cytosine. Another point to bear in mind is that bind-
5hmC immunoprecipitation ing of transcription factors to a TET-regulated locus may
This is the most straightforward method for detecting 5hydroxymethylcytosine antagonize DNA methylation (REFS 6972), thus if TET
(5hmC) and has been used by many laboratories39,84,93,126,141. However, the efficiency of depletion results in altered transcription factor bind-
precipitation strongly depends on the density of 5hmC in the samples26,76; there are
ing, the resulting changes in cytosine species could lead
notable variations in the results obtained in different laboratories39,84,93,126, and there is
evidence that the 5hmCspecific antibody precipitates polyCA repeats, resulting in
to incorrect interpretations of whether TET is causing
spurious peaks immediately adjacent to such repeats142. demethylation at that locus or not. Methylated DNA
immunoprecipitation (MeDIP) and other methods of
CMS immunoprecipitation
When 5hmC is treated with sodium bisulphite, it is converted into a compound called measuring 5mC abundance are less useful than bisulphite
cytosine methylene sulphonate (CMS)143. CMS is highly immunogenic, and CMS sequencing, as simple conversion of 5mC to 5hmC will
immunoprecipitation recovers 5hmCcontaining DNA with high specificity and produce a drop in measured methylation even if TET
lowbackground76,144. does not mediate demethylation at thelocus.
GLIB (glucosylation, periodate oxidation, biotinylation)
Glucose is attached to the hydroxyl group of 5hmC using the enzyme -glucosyl Genomic distribution of oxidized 5mC
transferase (BGT) from phage T4. Glucose is then oxidized with sodium periodate and Large-scale mapping of 5hmC, and more recently 5fC
treated with aldehyde reactive probe (ARP) to generate an adduct containing two and 5caC, has been performed for mammalian genomes.
biotins at the site of every 5hmC76,145. The hydroxymethylated DNA is then precipitated The available methods for mapping these modified bases
with streptavidin beads. are outlined in BOX1 and BOX 2. Commercial kits are
JBP1 available in many cases.
5hmC is glucosylated by BGT. The glucosylated base is bound by J-binding protein 1
(JBP1), which is conjugated to a bead, allowing for selective precipitation of Oxidized methylcytosine enrichment at promoters.
hydroxymethylated DNA146. The precipitation of 5hmC is highly specific, but not very There is contention as to whether 5hmC is enriched at
efficient. This method has not yet been used for the genome-wide mapping of 5hmC.
promoters, owing largely to how promoter and enrich-
5hmC selective chemical labelling (hMe-Seal) ment are defined. First, the definition of promoter varies
BGT adds a glucoseazide conjugate to 5hmC, which can then be biotinylated under between laboratories. Many promoters that are CpG-rich
gentle conditions using click chemistry73,147.
contain peaks of hydroxymethylation 5002000 bases
5fCDNA pulldown before and after the transcription start site (TSS) but are
An ARP molecule biotinylates the formyl group of 5fC83. This method was used to depleted for 5hmC at the TSS itself, where there is typi-
generate the first published genome-wide map of 5fC83, but it also biotinylates abasic
cally almost no modified cytosine73,74. In this Review, we
sites to some extent55.
classify these promoters as 5hmCenriched. Second, the
5fCSeal definition of enrichment varies depending on whether
BGT is used to protect 5hmC by linking it to an unmodified glucose. 5fC is then reduced
precipitation-based or single-base resolution methods
to 5hmC using sodium borohydride (NaBH4), and the product then reacts with
glucoseazide and is biotinylated as in hMe-Seal. Thus, 5fC is selectively biotinylated55.
are used. Precipitation-based methodologies compare
how many times a given region of DNA is sequenced
5fC and 5caC immunoprecipitation
in a 5hmC pulldown compared with a negative control
Antibodies specific for 5fC and 5caC have been used to precipitate these modifications
for sequencing, although the efficiency of precipitation is fairly low56. sample. As a result, regions that contain a higher than
average number of 5hmC residues in a span of several
hundred bases will be classified as enriched. Incontrast,
single-base resolution studies typically measure the frac-
When a biological role is observed for a TET pro- tion of total reads in which an individual CpG site is
tein, there has been a strong tendency in the literature hydroxymethylated. Thus, promoters with a high CpG
to invoke DNA demethylation as a major mechanism. density, but in which each CpG site has a relatively low
However, an unequivocal demonstration of demethyla- frequency of 5hmC, may be classified as 5hmCenriched
tion can be difficult, especially as the changes observed by a precipitation-based methodology but as 5hmC
in most studies have been subtle. At a given locus, depleted by a single-base methodology. Wewill pre
TET may simply be converting 5mC to 5hmC, or it dominantly discuss results from precipitation-based
may be converting 5mC to cytosine via an oxidized studies, although it is unclear which definition of enrich-
5mC intermediate by one of the pathways described in ment is more biologically relevant. Furthermore, any
FIG.1a. Actual DNA demethylation only occurs when given cytosine modification is either present or absent
5mC is converted to cytosine via an oxidized 5mC at any given CpG site at the level of a single allele, but can
intermediate. TET depletion will necessarily result in seem infrequent when a large, potentially heterogenous
decreased 5hmC, 5fC and 5caC, and a corresponding population of cells is examined.
increase in 5mCfrom which these oxidized methyl- In human73,75 and mouse11,74,76 ES cells, mouse neural
cytosines are derived. Thus, a simple increase in 5mC progenitor cells77, mouse neurons77 and the cerebellum
Click chemistry upon TET depletion is not enough to prove that a TET of 7day-old mice78 (which contains a significant popula-
Chemistry involving high-yield, protein is causing demethylation. Rather, the increase tion of neural progenitor cells), 5hmC is enriched at pro-
highly specific reactions that in 5mC has to be greater than the attendant decrease in moters in absolute terms and especially when compared
are compatible with
physiological conditions and
5hmC, 5fC and 5caC. If TET is demethylating a locus, with 5mC, which is generally depleted at promoters79.
maintain the integrity of TET depletion will result in a decrease in unmodified In these cell types, genes with hydroxymethylated pro-
biological molecules. cytosine as judged by bisulphite sequencing. Even here, moters are expressed at lower levels than other genes,

346 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 2 | Mapping 5hmC at base pair resolution


In ES cells, 5fC is found at largely the same set of pro-
moters as 5hmC83, and levels of 5fC55 and 5caC56 increase at
The simplest and most widespread method for distinguishing cytosine from these promoters upon TDG depletion, indicating that
5methylcytosine (5mC) treatment with sodium bisulphite followed by PCR at least some TET-mediated demethylation occurs at
amplification and bisulphite sequencing (BS-seq)148150 conflates the five cytosine theseloci.
species into two groups. 5mC and 5hydroxymethylcytosine (5hmC) are resistant to
deamination by sodium bisulphite and thus are read as cytosine, whereas cytosine,
5carboxylcytosine (5caC) and 5formylcytosine (5fC) are deaminated and so are read
Oxidized methylcytosine enrichment at gene bodies.
as thymine6,89,151 (see the table). Methods have been developed that distinguish 5hmC or In virtually all mammalian cell types studied, includ-
5fC from each other and from other cytosine species at base resolution, although there ing mouse and human ES cells, mouse liver and brain
is no method yet that allows all five bases to be distinguished simultaneously. and human melanomas, 5hmC is enriched at gene
Single molecule real time (SMRT) sequencing bodies11,73,77,78,81,82. In mouse brain, liver and ES cells (but
Using chemistry similar to 5hmC selective chemical labelling (hMe-Seal) (BOX1), not in human ES cells), gene expression positively corre-
abulkyconjugate is added to sites of 5hmC. The conjugate stalls a specially designed lates with gene body hydroxymethylation78,84,85, although
polymerase during DNA replication, and this stalling is monitored in real time to identify the reason for this correlation is unclear. While promoter
5hmC152. This method, as well as the oxidative bisulphite sequencing (OxBS-seq) and methylation strongly correlates with gene silencing, how
TET-assisted bisulphite sequencing (TAB-seq) methods (see below), provide single-base gene body methylation correlates with gene expression
resolution mapping of 5hmC. 5fC and 5caC produce substantial delays in replication varies with cell type and whether the methylated cyto-
even without the addition of a conjugate, although it is hard to distinguish them from sine exists in a CpG or non-CpG context8688, making it
each other153. SMRT sequencing can handle long DNA fragments (610kb) but does not
unclear how 5mC oxidation and/or DNA demethylation
yet exhibit the throughput required for genome-wide mapping in mammalian cells.
by TETproteins in gene bodies affects gene expression.
OxBS-seq
5hmC is oxidized to 5fC using potassium perruthenate151. Bisulphite sequencing is
Oxidized methylcytosine enrichment at enhancers. In all
performed on the same sample both before and after oxidation. As 5hmC is unaffected by
sodium bisulphite treatment and 5fC is deaminated, the extent of 5hmC at a site can be mammalian cells in which both 5hmC and enhancers
approximated by subtracting the frequency of deamination after perruthenate oxidation have been mapped, 5hmC is strongly enriched at enhanc-
from the frequency of deamination before oxidation. 5hmC is a rare base, so very high ers72,73,75,81,89, which are often regions of low CpG density
sequencing coverage is required to determine the extent of hydroxymethylation. with reduced levels of DNA methylation compared with
TAB-seq neighbouring regions72. Single-base resolution mapping
5hmC is protected from oxidation by glucosyl transferase (BGT)-mediated using TET-assisted bisulphite sequencing (TAB-seq)
glucosylation, and then all 5mC bases are oxidized to 5caC using a recombinant (BOX2) indicates that 5hmC is strongly enriched imme-
ten-eleven translocation (TET) enzyme89. After bisulphite treatment, all cytosine species diately adjacent to transcription factor-binding sites in
are therefore deaminated, except those that were originally 5hmC, so these can be enhancers, but is depleted at the precise site of binding 89.
read specifically. TAB-seq has revealed that 5hmC is often asymmetric with cytosine or Due to the absence of data on the binding of transcrip-
5mC at CpG sites. Like OxBS, this method requires very high sequencing coverage. tion factors to enhancers in TET-deficient cells, it is dif-
5fC chemically assisted bisulphite sequencing (fCAB-seq) ficult to know whether methylcytosine oxidation opens
5fC is normally deaminated by sodium bisulphite treatment, but when it reacts with chromatin structure, allowing transcription factors to
Oethylhydroxylamine, deamination is prevented. The extent of 5fC at a site can be bind, whether transcription factors recruit TET proteins
approximated by subtracting the frequency of deamination after the bisulphite
or whether both mechanisms operate and reinforce one
treatment of unreacted samples from the frequency of deamination after the bisulphite
treatment of Oethylhydroxylamine-treated sample. Although the technique is
another. Moreover, the reduction in 5mC and 5hmC at
selective, 5fC is very rare, so extremely high sequencing coverage (>1000 fold) must transcription factor-binding sites may reflect demethyl
be used to accurately map the base. ation mediated by TET proteins, but could also be because
transcription factors physically block access of DNMT69,90
and TET proteins. Probably, both mechanisms are
Base Read by Read by Read by Read by
BS-seq oxBS-seq TAB-seq fCAB-seq atwork.
Notably, in differentiating ES cells, 5hmC levels
Cytosine T T T T
increase sharply at activated enhancers91. 5hmC levelsrise
5mC C C T C almost immediately with the onset of differentiation and
5hmC C T C C either precede or accompany acetylation of Lys27 of
5fC T T T C histone H3 (H3K27), which is a mark of active enhancers.
Furthermore, 5fC and 5caC are enriched at enhancers in
5caC T T T T
TDG-depleted ES cells, consistent with TET-mediated
demethylation of enhancers55,56. However, the correlation
between the 5fC levels at enhancers and hypomethylation
although this does not prove a silencing role for 5hmC; is weak55. Further studies will be necessary to determine
5hmC can only be formed at promoters that have some the extent to which TET proteins contribute to enhancer
underlying level of methylation, and promoter methyl hypomethylation and function.
ation correlates with silencing. In other cell types,
Sequencing coverage including PGCs80, adult nervous tissue78, liver cells81 and Mapping TETs and TET-interacting proteins
Average number of times that benign nevi82, 5hmC is depleted from TSSs. The discrep- The distributions of TET proteins and oxidized 5mC spe-
a genome or a DNA region
is sequenced using a
ancies in 5hmC distribution among cell types could be cies do not overlap neatly. All three TET proteins and IDAX
next-generation sequencing caused by differences in the underlying distribution of are strongly enriched at promoters, especially promoters
instrument. 5mC and TET proteins. that are CpG-rich39,41,9295. In the case of TET1 (REF.39)

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 347

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

and TET3 (REF.42), this preference is partially determined TET proteins in embryonic development
by the presence of CXXC domains, which most likely bind TET proteins are implicated in several stages of mouse
CpG sequences, as discussed above. development, particularly those in which mass demethyl
The data support a repressive role for TET1 in EScells. ation and denovo methylation take place, namely the
In three studies in mouse ES cells, there is a strong statis- zygote, the inner cell mass of the blastula and PGCs
tical correlation between the physical presence of TET1 (FIG. 3) . The phenotypes of TET-deficient mice are
at a promoter and increased gene expression from this described in BOX3.
promoter upon TET1 knockdown39,92,93. The correla-
tion between the presence of TET1 at a promoter and TET3 in the zygote. Immediately after fertilization,
decreased expression from this promoter upon TET1 before the genetic material from the sperm and egg have
knockdown is much weaker. TET2 shows precisely the fused to form one nucleus, the male pronucleus loses
opposite trend: there is a correlation between the physical almost all methylation as assessed by staining with anti-
presence of TET2 at a promoter and decreased expression bodies specific for 5mC103105 (FIG.3). This process occurs
from this promoter upon TET2 depletion, which indicates before replication and is not inhibited by DNA polymer-
that TET2 is generally a positive regulator of gene expres- ase inhibitors. In contrast, bisulphite sequencing, which
sion95. The likely explanation for the different activities does not distinguish between 5mCand 5hmC106108
of TET1 and TET2 is that, in ES cells, TET1 primarily (BOX2), indicates that this loss of 5mC is much slower
modulates transcription by recruiting the repressive and less complete than the abrupt loss measured by
histone deacetylase-containing MBD3NURD (nucleo- antibody staining 104,109111. These results are reconciled
some remodelling and deacetylase)96 and SIN3A (switch- by the observation that the zygotic demethylation event
independent 3A) complexes13,94,97 to promoters. TET2 is in fact a mass 5mC oxidation event; the apparent
shows no association with these complexes in ES cells93 loss of 5mC stems from the fact that 5mCspecific
and, when exogenously expressed in 293T cells, TET1 and antibodies do not recognize 5hmC or other oxidized
TET3 associate with SIN3A, whereas TET2 does not 94. methylcytosines. Instead, the male pronucleus shows
Furthermore, in contrast to somatic cells in which methyl a selective increase in 5hmC111113 and other oxidized
ation has an impact on the expression of thousands of methylcytosines114, as observed by staining with appro-
genes98, methylation only regulates a handful of genes in priate antibodies. The increase in 5hmC is abrogated by
ES cells and is dispensable for ES cell growth and sur- siRNA-mediated knockdown of Tet3 (REF. 112), and is not
vival99. Hence, the demethylating properties ascribed to observed in fertilizations that involve TET3deficient
TET proteins may be of limited importance in regulating oocytes113. This indicates that TET3 oxidizes 5mC in
transcription in EScells. the male pronucleus.
Recently, three groups have reported a physical asso- The protein DPPA3 (development pluripotency-
ciation between TET proteins and the enzyme OGT associated 3; also known as Stella or PGC7) protects
(Olinked -d-Nacetylglucosamine (OGlcNAc) trans- maternal DNA and a few imprinted loci in the paternal
ferase)94,95,97. Despite some contradictions in these studies, genome from TET3mediated oxidation115. DPPA3 is
it is probable that all three TET proteins associate with reported to be recruited to the protected regions by
OGT100. OGT adds a GlcNAc sugar to Ser and Thr resi- dimethylation of H3 at Lys9 (H3K9me2), a mark that
dues of numerous proteins, including histone H2B and is found selectively at these sites115. In the absence of
other chromatin modifiers. The impact of this sugar DPPA3, both paternal and maternal genomes become
modification on the target protein is highly contextual. hydroxymethylated 112,115 and most show abnorm
For instance, O-GlcNAcylation can serve to antagonize alities by the 2-cell or 4-cell stage 116-118. In contrast,
phosphorylation, direct proteolysis or allow protein Tet3deficient oocytes fertilized with wild-type sperm
protein interaction101. OGT, like TET proteins, is strongly have reduced expression of OCT4 during the morula
enriched at promoters. In ES cells, TET proteins recruit stage but form blastocysts normally. Moreover, about
OGT to promoters, but not vice versa. Depletion of TET half of these animals show serious abnormalities by
proteins diminishes the amount of OGT at promoters, but embryonic day E11.5 and fail to develop113. Thus,
depletion of OGT does not influence the association of methylcytosine oxidation in the male pronucleus
TET1 or TET2 with chromatin, although it does alter the enhances, but is not required for, survival of the embryo.
distribution of 5hmC at certain loci95, 97. There are various It is not clear why some zygotes survive, whereas others
mechanisms by which OGT could influence transcrip- do not, and why it takes 11days for the failure of zygotic
tion. For example, levels of O-GlcNAcylation at Ser112 of oxidation to result in lethality.
Zygote H2B, a mark associated with transcriptional activation102, The bulk of the evidence suggests that cytosine modi-
Cell formed by fertilization drop sharply upon TET2 depletion providing a mecha- fication is lost through a replication-dependent process
ofthe oocyte (egg) with a
nism whereby TET2 increases transcription through OGT. that may be enhanced by, but is not completely dependent
spermcell.
Likewise, OGT glycosylates HCF1 (host cell factor 1), on, TET enzymes or BER. In fact, maintenance methyl
Imprinted locus a component of the H3K4 methyltransferase SET1 complex ation is very inefficient in the early embryo because
In epigenetics this describes (also known as COMPASS) associated with active RNA of the cytoplasmic localization of DNM1O, which is a
agenomic region with a polymeraseII (Pol II), providing a link between H3K4 special splice variant of DNMT1 present in the early
methylation mark that is
present only on the maternally
trimethylation and TET proteins94. Given the many pro- mouse embryo80,103,105. As a result, 5mC on the maternal
or paternally derived copy of tein targets of OGT, it is likely that there will be additional DNA and 5hmC on the paternal DNA are diluted out
an allele. consequences of TET-mediated OGT recruitment. through a passive replication-dependent process (FIG.3).

348 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Whole embryo b PGCs


TET3 high TET1 and TET2 high TET1 and TET2 high

Relative level of cytosine modication


5mC 5mC

Relative level of cytosine modication


5hmC 5hmC
5fC and/or 5caC

(not to scale)
(not to scale)

Fertilization Zygote 216 cell Morula Blastula E9.5 E10.5 E11.5 E12.5 E13.5 Later
stage development
Figure 3 | Methylation dynamics in mammalian development. a | Immediately after fertilization, the male pronucleus
undergoes mass cytosine oxidation111113, mediated by ten-eleven translocationNature Reviews
3 (TET3). | Molecular Cell
5methylcytosine (5mC)Biology
and
oxidized cytosines are then lost from the early embryo in a passive or replication-dependent manner, resulting in the
loss of nearly all modified cytosines by the 16cell stage103,105. Imprinted loci retain methylation183 and some repetitive
element classes184 retain partial methylation. Approximately when the blastula implants into the uterus, the inner cell
mass, which gives rise to the embryo, undergoes mass denovo DNA methylation105,183. TET1 and TET2 are highly
expressed at this stage, potentially fine-tuning methylation patterns. b | Demethylation also occurs in primordial germ
cells (PGCs) between embryonic days E9.5 and E13.5 of embryonic development80,120,128. This event also entails both mass
5mC oxidation by TET1 and TET2 and loss of modified cytosine by passive demethylation, resulting in the loss of imprints.
A similar process of 5mC oxidation and demethylation occurs more slowly in human germ cells185. This demethylation of
imprints is critical because whereas somatic cells of an organism contain male and female imprints, the germ cells of an
organism contain the imprints that correspond exclusively to the gender of the organism. Germ cells are then gradually
re-methylated and imprints placed, starting at E15 in males and after birth in females186.

Substantial levels of 5hmC are retained at least through male pronucleus and that this oxidation event enhances
the 8cell stage111,119, arguing against the possibility that survival of the early embryo, but why this oxidation
it is excised or converted directly to cytosine. However, occurs remains unknown.
some loss of 5mC is observed by bisulphite sequencing
before any cell replication event 113. A likely explana- TET1 and TET2 in ES cells. When pluripotent EScells
tion for this is that TET3 is oxidizing 5mC to 5fC and are injected into immunocompromised mice, they
5caC114, creating the appearance of demethylation by form tumours called teratomas that contain tissues
bisulphite sequencing (BOX2). It is also plausible that from all three germ layers (ectoderm, mesoderm and
some proportion of 5fC and 5caC is removed by TDG endoderm). Tet1/ ES cells123, Tet1/ Tet2/ ES cells124
and replaced by cytosine 30. Evidence showing BER and ES cells stably expressing Tet1 short hairpin RNA
in the male pronucleus supports this possibility110,120. (shRNA)125 form large haemorrhagic teratomas that
Arguing against direct removal of 5fC and 5caC is are enriched for trophoblast cells, suggesting that
evidence demonstrating that the modified bases remain TET1 regulates the first lineage commitment step in
in the embryo at least until the 4cell stage114 and the the embryo by suppressing differentiation towards the
fact that large-scale BER would be expected to cause extra-embryonic lineage. Notably, however, Tet1/
increased rates of mutations in the zygote, which would Tet2/ ES cells contribute efficiently to chimaeras when
be deleterious to the organism and its progeny. injected into blastocysts124, indicating that the skewing
As methylation is lost passively anyway, why are towards trophoblast differentiation observed in Tet1/
methylcytosines oxidized in the mammalian zygote? and Tet1/ Tet2/ teratomas can be overcome by other
The most likely possibility is that methylcytosine oxi- regulatory influences during embryonic development.
dation in the male pronucleus accelerates global DNA TET1 depletion influences gene expression and differ
demethylation, either through 5fC and 5caC formation entiation in mouse ES cells by negatively regulating
and the subsequent removal of these modified bases the expression of key trophectoderm regulators, such as
Trophectoderm
or by ensuring that maintenance methylation is espe- caudal-type homeobox 2 (CDX2)27,125,126, eomesodermin
Cells that give rise to the
placenta and other cially inefficient in the earliest cell divisions. No proof (EOMES)93,123,125 and ELF5 (REF.125), and positively reg-
extra-embryonic tissue. of either mechanism has been demonstrated. It is also ulating expression of the neuroectoderm factors paired
possible that cytosine oxidation promotes broad trans box 6 (PAX6)39,93,123,125,126 and neurogenic differentiation
Embryoid bodies criptional activation previously observed in the male factor 2 (NeuroD2)39,93,123. However, neural progenitor
Aggregates of cells formed by
allowing embryonic stem cells
pronucleus121. However, depletion of TET3 in the early cells can be derived from Tet1/ embryoidbodies123, and
to differentiate without contact zygote is reported to have no effect on global transcrip- Tet1/ and Tet1/ Tet2/ mice develop all three germ
with a solid surface. tional levels122. In short, it is clear that TET3 oxidizes the layers and survive, suggesting that the role of TET1 and

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 349

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 3 | Phenotypes of TET-deficient mice


Generally, the data support a real but limited role for
TET1 and TET2 in demethylation in ES cells. Bisulphite
Ten-eleven translocation (Tet1) / mice on a mixed C57BL/6129 inbred mouse strain sequencing of selected loci in TET1deficient ES cells
background are born at Mendelian ratio and appear healthy despite a low birth does indicate a very modest increase in methylation
weight123. Mouse Tet1 gene-trap mutant animals show embryonic lethality on a levels39. Mass spectrometry indicates that cells deficient
129P2/OlaHsd mouse strain background, with noticeable abnormalities by embryonic
in both TET1 and TET2 show an increase in 5mC lev-
day E8.5, butthey are viable and fertile on a C57BL/6 background129. Female Tet1
gene-trap mutants have smaller litters, similarly to Tet1/ mice. In female Tet1 gene-trap
els (from 5.3% to 5.8% of all cytosines) and a smaller
mutants, which have smaller ovaries, about half of the developing female gametes absolute drop in 5hmC levels (from 0.13% of all cyto-
display a defect in meiotic synapsis and suffer a developmental arrest between E16.5 sine to undetectable) 124. Thus, there is a ~1.05fold
and E18.5 (REF.129). Whether Tet1/ mice have a similar meiotic defect is unknown. increase in the total levels of cytosine modification
Tet2/ mice on C57BL/6 or mixed backgrounds are born at Mendelian ratios and are (5.43% to 5.8%). If the increase in cytosine modifica-
fertile, but display clear haematological abnormalities. They have more haematopoietic tion is concentrated at promoters at which CpG levels
stem cells (HSCs) than normal mice, and their HSCs have enhanced self-renewal and are high and TET proteins are most active, this could
proliferative potential in culture and in experiments in which progenitor cells are serially have a strong impact on transcription and develop-
transferred in mice154157. At 4months old, one strain of Tet2/ mice has been reported to ment. However, caution must be used in extrapolat-
develop a condition similar to human chronic myelomonocytic leukaemia (CMML), a
ing changes in methylation observed in ES cells, which
malignancy typified by a gross abundance of monocytes155,157. A Tet2 gene-trap strain
shows perinatal lethality158, but because this phenotype is not observed in several
undergo many generations of division in culture, to the
conventional Tet2 gene-disrupted strains, it is most likely an artefact. inner cell mass that differentiates quickly.
Tet1/ Tet2/ mice on a mixed C57BL/6129 background are born at near Mendelian Most tissues in Tet1/Tet2/ mice show modest
ratios and have fully developed organs, but roughly half of these animals die perinatally, increases in total cytosine modification levels as meas-
often with visible defects in head development124. Surviving mice have a smaller birth ured by mass spectrometry, although it is unclear
and adult weight, and females have reduced ovary size and fertility, producing litters whether this is due to inadequate demethylation in
of about one fourth the normal litter size. Tet1/ Tet2/ male mice crossed with the blastula, during later embryonic development
wild-type females produced healthy progeny, but more than half of the progeny of or in adult tissue. The incompletely penetrant lethal
Tet1/ Tet2/ females crossed with wild-type males died perinatally, probably due phenotype of Tet1/Tet2/ mice (BOX3) could reflect
to an imprinting defect in the Tet1 Tet2 oocytes (see main text).
a model in which, by stochastic variation, some mice
Roughly half of the embryos that arise from Tet3 oocytes, regardless of the sperm
genotype, arrest around E11.5 and do not survive. The Tet3/ mice that survive
acquire excess methylation at key promoters in early
embryonic development die perinatally for unknown reasons113. development, leading to subsequent lethality, whereas
other mice do not. However, the perinatal lethality of
Tet1/Tet2/ mice cannot be assumed to be caused by
TET2 in differentiation is minor. Overall, the data indi- alterations in DNA methylation. Furthermore, virtually
cate that, despite the abundance of 5hmC in ES cells, all of the Tet1/ Tet2/ mice develop to birth with fully
TET1 and TET2 have modulatory but not essential roles developed organs, so any dysregulation of methylation
in mouse development, ES cell survival or pluripotency. is necessarily fairlymild.
Whether TET3 can compensate for the loss of TET1 and
TET2 has not yet been established. TET proteins in PGCs. PGCs undergo a rapid drop
Nevertheless, the catalytic activity of TET proteins in 5mC levels between E9.5 and E10.5 (FIG.3). As in
may be important in ES cells and the inner cell mass zygotes, this apparent demethylation event corre-
from which they are derived, because of the underlying sponds to mass conversion of 5mC to 5hmC, in this
biology of these cells. EScells approximately recapitulate case mediated by TET1 and TET2 (REFS80,127). There
the stage of development in which large-scale denovo is no detectable formation of 5fC or 5caC as measured
methylation of the genome occurs (FIG. 3). Because tran- by immunocytochemistry 80. Measurement of individ-
scriptional initiation antagonizes methylation71,87, genes ual loci during germ cell development indicates that
with lower levels of transcription are presumably more 5mC is replaced by 5hmC between E9.5 and E11.5,
susceptible to aberrant increases in DNA methylation. and that both marks are subsequently lost at a rate
As methylation is generally maintained through cell consistent with dilution by replication, as opposed to
Inbred mouse strain divisions, aberrant methylation may be inconsequential active removal 80. Thus, if TET proteins are mediat-
Experiments are typically at early embryonic stages but deleterious during later ing demethylation in PGCs, it is by facilitated passive
conducted using inbred mouse development. Thus, the function of TET proteins in demethylation. However, even this mechanism may not
strains, in which all mice are
genetically extremely similar.
the inner cell mass may be to repress lineage-specific be critical, as UHRF1 and DNMT3B levels are quite
C57BL/6 is one of the most genes, while simultaneously antagonizing methylation low in PGCs128 and thus passive demethylation could
frequently used strains. to permit activation of these genes later in develop- occur without TET activity.
129P2/OlaHsd is another ment. Accordingly, in EScells, both TET1 and 5hmC are Germ cells derived from mouse Tet1 gene-trap
inbred mouse strain. The same
enriched at promoters that contain dual H3K4me3 and mutants (BOX 3) have almost normal methylation
mutation can have different
effects in different backgrounds H3K27me3 marks, which are so-called poised promot- levels129. At E13.5, when demethylation is complete,
ers that can be activated later in development 74,76. Loci an increase in methylation corresponding to less than
Gene-trap with high levels of 5fC in ES cells show increased cyto- 1% of the total cytosines at CpG sites is observed in
A mutant in which a gene is sine methylation in TDG-deficient MEFs83, even though TET1deficient cells129. A much greater effect is seen
disrupted by the random
insertion of transgenic DNA
TET1 and TET2 are almost absent from MEFs. Thus, at the level of gene expression: loss of TET1 causes
that contains a splice acceptor a demethylation defect in early development seems to dysregulation of ~1000 genes, 90% of which are posi-
site followed by stop codons. result in excess methylation at a laterstage. tivelyregulated by TET1, indicating that TET1 primarily

350 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

functions to activate transcription in PGCs. A number similar to ES cells130. The classic combination of trans
of critical meiosis-related genes are targets of TET1 cription factors used in these experiments is OCT4 (also
activation, including malate dehydrogenase 1 (Mae1), known as POU5F1), SOX2, Krppel-like factor 4 (KLF4)
synaptonemal complex protein 1 (Sycp1) and Sycp3. and MYC (collectively referred to as OSKM), although
Half of the developing TET1deficient female gametes other combinations have also been used successfully 131.
subsequently display a defect in meiotic synapsis and For efficient reprogramming, endogenous pluripotency
suffer a developmental arrest between E16.5 and E18.5 factors must be activated and their promoters and
(REF. 129), and accordingly Tet1 gene-trap and Tet1/ enhancers demethylated 132. Although TET proteins
mice have reduced litter size. Although it is possible are apparently dispensable for pluripotency in ES cells
that TET1 facilitates critical demethylation events at and invivo, several recent publications indicate roles
key genes, it seems unlikely that such a modest change for TET1 and/or TET2 in the generation of iPScells,
in methylation levels has such a drastic effect on gene although they do not report a common mechanism.
expression. It is more likely that TET1 modulates Specifically, one study showed that the depletion
geneexpression by recruiting other proteins or that of TET2 by shRNA completely ablated reprogramming of
oxidized cytosines are influencing transcription in a fibroblasts by OSKM, and that the Nanog promoter
way that does not involve their removal. was hydroxymethylated and demethylated during the
The simultaneous shRNA-mediated depletion of first 4days after reprogramming 133. These findings,
TET1 and TET2 (REF.80), or the depletion of TET1 from together with the fact that TET1 and TET3 expres-
Tet2/ cells127, seems to partly antagonize demethyl sion was not observed during this time frame, suggest
ation at a few loci in PGC-like cells derived invitro from that TET2 has a role in generating iPS cells. In another
precursors. Furthermore, there is evidence that oocytes study, TET1 and TET2 were shown to physically interact
deficient in TET1 and TET2 may not completely with Nanog in ES cells, and co-transfection of Nanog with
erase all parental imprints during PGC demethyl either TET1 or TET2 greatly enhanced OCT4, KLF4
ation. Although Tet1+/ Tet2+/ mice had no intrinsic and MYC-mediated reprogramming of neural stem
survival defect, half of the progeny of Tet1/ Tet2/ cells134. The authors proposed that Nanog and TET1
females crossed with wild-type males display perinatal or TET2 could co-activate the endogenous Oct4 and
lethality 124. This indicates that some fraction of the Esrrb (oestrogen-related receptor beta) genes via a
Tet1 Tet2 oocytes completed meiosis successfully but mechanism that involves at least some demethylation.
were nevertheless defective, as judged by the perinatal A third study showed that co-transfecting TET1 with
lethal phenotype observed in mice developing from the OSKM factors greatly enhances the reprogramming of
Tet1Tet2 oocytes. Accordingly, some imprinted loci fibroblasts135. These data suggest that TET1 mediates
in the progeny of Tet1/ Tet2/ mice show aberrant the oxidation and demethylation of the Oct4 promoter
methylation. The occurrence and degree of increased and proximal enhancer, and that TET1 can substi-
methylation varies across loci and between individual tute for OCT4 in OSKM-dependent reprogramming
mice. This is consistent with mass genomic demethyl transfection. Inall of the above cases, demethylation
ation proceeding normally in TET-deficient animals, of the target loci is slow enough to be caused by a pas-
but the subsequent erasure of imprints being variably sive mechanism. The requirement for TET proteins in
impaired to cause fatal defects in some, but not all, reprogramming clearly varies depending on the precise
progeny. conditions and status of the starting cell lines. However,
Thus, in the two instances of mass demethylation the ability of TET proteins to generate oxidized methyl-
in mammals, essentially the same pattern occurs. 5mC cytosines and antagonize methylation of select loci may
is oxidized enmasse by TET3 in zygotes and TET1 help to establish the pluripotentstate.
and TET2 in PGCs, predominantly producing 5hmC,
and there is little evidence that the oxidized methyl TET proteins in the nervous system
cytosines are excised. Rather, all modified cytosines The brain consistently has the highest levels of 5hmC
are diluted out during subsequent cell divisions, due to of any mammalian tissue. This may in part reflect
the expression of DNMT1O in zygotes and the down- the abundance of long-lived post-mitotic cells in the
regulation of UHRF1 in PGCs. Insofar as oxidized nervous system, such that 5hmC can accumulate in
methylcytosines are promoting demethylation in these neurons without being depleted through replication.
systems, they probably achieve this by antagonizing It is still largely unknown how TET deficiency influ-
maintenance methylation under conditions in which ences mouse brain development. Increased 5hmC
this process is already ine fficient. Potentially, the and decreased DNA methylation clearly correlate
oxidized methylc ytosines have important functions with higher gene expression levels and a more open
that are not related to their removal as, for example, chromatin state in neural cells85 but causality has not
epigeneticmarks. been established. Depletion of TET2 and TET3 in the
Meiotic synapsis developing mouse cortex, by inutero electroporation of
The event in meiosis prophase I TET proteins in reprogramming shRNA, causes a block in the differentiation of neural
in which homologous The transfection of developmentally-committed cells progenitor cells into neurons77.
chromosomes align to allow
recombination of genetic
with certain combinations of transcription factors can Xenopus laevis embryos injected with morpholinos
material (known as crossing reprogramme them, with low efficiency, into induced targeting tet3 develop a gross defect in early develop-
over). pluripotent stem (iPS) cells that are phenotypically ment, that features loss of expression of critical eye

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 351

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 4 | Effects of hydroxymethylation on methylation-based silencing


Methyl CpG-binding protein 1 (MBD1), MBD2 and a HDAC H3K9MT
Kaisospecifically recognize methylated DNA and recruit MBD MBD
HDAC H3K9MT
histone deacetylases or repressive histone H3 Lys9 MBD MBD H4ac
methyltransferases (H3K9MTs) to 5mC159164.
5hydroxymethylcytosine (5hmC) strongly inhibits the 5mC Oxidized
binding of these MBD proteins to DNA49,108,165 and 5mC
therefore is thought to induce transcriptional activation
(see the figure, part a). However the role of MBD proteins H3K9me3
in regulating gene expression is relatively subtle.
Micedeficient in Kaiso166, MBD1 (REF.167) and MBD2 b HDAC H3K9MT
(REF.168) develop and grow normally, although DNMT1
MBD1- and MBD2deficient mice have mild neurological HDAC H3K9MT UHRF1
DNMT1
impairments. Methyl CpG binding-protein 2 (MeCP2), an UHRF1 H4ac
MBD protein with a more complex regulatory role169171,
may bind 5hmC in addition to 5methylcytosine (5mC) . 49,85
5mC Oxidized
Various other transcriptional modulators bind 5mC 5mC
selectively, and whether and how 5mC oxidation perturbs
their activity is unclear136. H3K9me3
DNA methyltransferases (DNMTs), including DNMT1,
recruit histone deacetylases (HDACs) and H3K9 c
MLL KDMA2A MLL KDMA2A
methyltransferases to DNA. At promoters, this
CXXC CXXC CXXC CXXC
perpetuates a repressive chromatin state through cell
division172 (see the figure, part b). As 5hmC potentially
prevents DNMT1 recruitment, hydroxymethylation could
block this mechanism of methylation-mediated silencing 5mC Oxidized
in dividing cells (FIG. 1c). 5mC
5mC can prevent chromatin remodellers that possess
CXXC domains from binding to DNA (see the figure, H3K36me3
partc)34. These include the H3K4 methyltransferase
mixed-lineage leukaemia (MLL)38, CXXC1, a component
d
of the SET1 H3K4 methyltransferase complex , and 173
5mC ? Oxidized TF
Lys-specific demethylase 2A (KDM2A), which removes 5mC
H3K36 methylation marks37. It is unlikely that these
proteins could bind oxidized 5mCs.
Methylation increases nucleosome compaction (see the e TF TF
figure, part d), and this is likely to reduce the accessibility
of adjacent DNA to transcription factors invitro174.
Howhydroxymethylation affects nucleosome compaction 5mC Oxidized
is unknown, although 5hmCrich Purkinje neurons have 5mC
markedly decondensed nuclei.
In some instances, cytosine methylation physically blocks
the binding of a transcription factor (TF) to its target f RNA Pol mRNA
sequence175,176 (see the figure, part e). Alternatively, by
blocking binding of repressors or insulators, methylation Terminated RNA Pol
mRNA
can actually increase the expression of a locus177180. If the
binding of a transcription factor is inhibited by methylation, 5mC Oxidized
it is unlikely that methylcytosine oxidation would restore 5mC
binding. Furthermore, as 5hmC is bulkier than 5mC, it may
block the binding of proteins that tolerate 5mC.
Nature Reviews
Finally, quantum mechanical calculations indicate an inherent role for 5mC in stabilizing | Molecular
DNA duplexes Cell Biology
by promoting
base stacking interactions181. Recent experiments confirm that methylated DNA has a higher melting temperature
thanunmodified DNA, suggesting that methylation may directly inhibit transcriptional initiation or elongation by
RNApolymerase (RNA Pol) by preventing necessary melting of the template (see the figure, part f)182. By contrast,
5hmCreduces the melting temperature of DNA duplexes to promote transcriptional elongation.

and neural markers and results in small heads and no Interpreters of 5mC oxidation
eyes42. The phenotype was partly rescued by injection As the extent to which 5mC oxidation products are
of mRNA coding for a catalytically inactive TET3, demethylation intermediates is unclear, other mecha-
whereas injection with a TET3 CXXC domain mutant nisms by which they could influence transcription
provided no rescue at all. Thus, TET3 has a role in should be considered.
normal head development in X.laevis. It is unknown Some transcriptional regulators and chromatin-
whether the perinatal lethality observed in Tet3 / associated proteins specifically recognize 5hmC or other
mice113 is linked to a neurological development defect. oxidized methylcytosines. Mass spectrometry has been

352 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

used to identify proteins bound selectively to 5hmC can drastically stall the progression of PolII on a recom-
containing DNA oligonucleotides in ES cells, neural binant substrate140. This phenomenon is functionally and
progenitor cells and mouse adult brain136. Proteins that perhaps also mechanistically analogous to that observed
were found to bind to 5hmC included the neural pro- with base J, which has a crucial role in transcription
genitor cell-specific protein UHRF2, which presumably termination in kinetoplastids25.
recognizes 5hmCcontaining DNA via its SAD/SRA Finally, methylcytosine oxidation could antagonize
domain, transcription factors such as zinc-fingers and 5mCmediated silencing. Six different mechanisms for
homeoboxes protein 1 (ZHX1), ZHX2 and THAP methylation-based silencing have been demonstrated
domain-containing protein 11 (THAP11), as well as sev- (BOX4). The relative importance of each mechanism is
eral uncharacterized proteins. An important limitation of unclear, but their relevance clearly varies across cell types
this study is that it does not distinguish between proteins and loci. The impact of methylcytosine oxidation on
that bind directly and indirectly to 5hmC. Despite the these silencing mechanisms is also likely to be variable.
inability of glycosylases to cleave 5hmC invitro52, sev- Thus, in order to understand how 5hmC influences tran-
eral glycosylases and DNA repair proteins were found scription at a locus, it may first be necessary to determine
to associate with 5hmC in this study, suggesting that how methylation mediates silencing at thatlocus.
5hmC may be a target of DNA repair 136. In addition, two
methyl-binding proteins, methyl CpG binding-protein2 Conclusions and future directions
(MeCP2) and methyl-CpG binding domain protein 4 The discovery of TET proteins has provided a new per-
(MBD4), were shown in this and other studies to bind spective on how DNA modification influences gene
to 5hmC85,136,137. However, an initial finding that MBD3 expression. There are now several potential mechanisms
(REF.96) preferentially binds to 5hmC over 5mC has not by which TET proteins and oxidized methylcytosines
been reproducible49,136. might mediate DNA demethylation, but whether and
TDG specifically binds 5fC and 5caC invitro53 and how these mechanisms contribute to gene regulation
might therefore be a reader of 5fC and 5caC. Indeed, in different cell types remains to be elucidated. It will
5fC and 5caC were found to bind TDG in mouse EScell, be important to determine how proteins that bind oxi-
as well as a substantial population of DNA repair pro- dized methylcytosines contribute to gene regulation and
teins, several transcription factors and a number of mis how the enzymatic activities and biological roles of TET
cellaneous proteins, some of which may simply bind proteins are influenced by interactions with other pro-
formyl or carboxyl groups nonspecifically 136. A sugges- tein complexes. The ongoing development of improved
tion that TDG recruits the histone acetyltransferase p300 methods for mapping oxidized methylcytosines prom-
(REF.138) is countered by the finding that the number of ises to advance the field substantially, especially if it can
p300binding sites in the genome actually increases in be applied at single-base resolution to small numbers of
TDG-deficient ES cells55. However, TDG could neverthe- differentiating cells. A careful analysis of changes in the
less be a transcriptional regulator, as it has been reported distribution of oxidized methylcytosines, transcription
to interact with multiple DNA-binding transcription levels, transcription factor binding and chromatin modi-
factors139. fications in cells from animals lacking one or multiple
Oxidized methylcytosines can affect Pol II processivity TET proteins will also be essential to further our under-
directly. 5mC and 5hmC have a relatively small effect on standing of how TET proteins modulate gene expression,
polymerase processivity invitro, but a single 5fC or 5caC cell differentiation and function.

1. Tahiliani,M. etal. Conversion of 5methylcytosine to Together with references 30 and 53, shows that 13. Williams,K., Christensen,J. & Helin,K. DNA
5hydroxymethylcytosine in mammalian DNA by MLL TDG excises 5fC and 5caC. Demonstrates the methylation: TET proteins-guardians of CpG islands?
partner TET1. Science 324, 930935 (2009). presence of 5caC in mammalian DNA and shows EMBO Rep. 13, 2835 (2012).
Discovery that TET proteins oxidize that 5caC levels increase upon TDG depletion. 14. Tan,L. & Shi,Y.G. Tet family proteins and
5mC to 5hmC. 7. Szwagierczak,A., Bultmann,S., Schmidt,C.S., 5hydroxymethylcytosine in development and
2. Iyer,L.M., Tahiliani,M., Rao,A. & Aravind,L. Spada,F. & Leonhardt,H. Sensitive enzymatic disease. Development 139, 18951902
Prediction of novel families of enzymes involved in quantification of 5hydroxymethylcytosine in (2012).
oxidative and other complex modifications of bases in genomic DNA. Nucleic Acids Res. 38, e181 15. Bejar,R., Levine,R. & Ebert,B.L. Unraveling the
nucleic acids. Cell Cycle 8, 16981710 (2009). (2010). molecular pathophysiology of myelodysplastic
Describes the evolution of TET proteins and the 8. Globisch,D. etal. Tissue distribution of syndromes. J.Clin. Oncol. 29, 504515 (2011).
presence of TET homologues in non-metazoan 5hydroxymethylcytosine and search for active 16. Pronier,E. & Delhommeau,F. Role of TET2
species. demethylation intermediates. PloS one 5, e15367 mutations in myeloproliferative neoplasms.
3. Ono,R. etal. LCX, leukemia-associated protein (2010). Curr. Hematol. Malignancy Rep. 7, 5764
with a CXXC domain, is fused to MLL in acute myeloid 9. Kriaucionis,S. & Heintz,N. The nuclear DNA base (2012).
leukemia with trilineage dysplasia having t(10;11) 5hydroxymethylcytosine is present in Purkinje 17. Mercher,T. etal. TET2, a tumor suppressor in
(q22;q23). Cancer Res. 62, 40754080 (2002). neurons and the brain. Science 324, 929930 hematological disorders. Biochim. Biophys. Acta
4. Lorsbach,R.B. etal. TET1, a member of a novel (2009). 1825, 173177 (2012).
protein family, is fused to MLL in acute myeloid Together with reference 1, convincingly 18. Iyer,L.M., Anantharaman,V., Wolf,M.Y. &
leukemia containing the t(10;11)(q22;q23). Leukemia demonstrates the presence of 5hmC in mammals. Aravind,L. Comparative genomics of transcription
17, 637641 (2003). 10. Cimmino,L., Abdel-Wahab,O., Levine,R.L. & factors and chromatin proteins in parasitic protists
5. Ito,S. etal. Tet proteins can convert 5methylcytosine Aifantis,I. TET family proteins and their role in stem and other eukaryotes. Int. J.Parasitol. 38, 131
to 5formylcytosine and 5carboxylcytosine. Science cell differentiation and transformation. Cell Stem Cell (2008).
333, 13001303 (2011). 9, 193204 (2011). First paper to predict a 5mC oxidase and
Shows that TET proteins can produce 5fC and 5caC 11. Wu,H. & Zhang,Y. Mechanisms and functions of Tet demethylase activity for TET1 (which was then
and quantifies the level of modified cytosines in a protein-mediated 5methylcytosine oxidation. Genes termed CXXC6).
range of cell types. Dev. 25, 24362452 (2011). 19. Gommers-Ampt,J.H. etal. dglucosyl-
6. He,Y.F. etal. Tet-mediated formation of 12. Branco,M.R., Ficz,G. & Reik,W. Uncovering the role hydroxymethyluracil: a novel modified base present in
5carboxylcytosine and its excision by TDG in of 5hydroxymethylcytosine in the epigenome. the DNA of the parasitic protozoan T.brucei. Cell 75,
mammalian DNA. Science 333, 13031307 (2011). Nature Rev. Genet. 13, 713 (2012). 11291136 (1993).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 353

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

20. Yu,Z. etal. The protein that binds to DNA base J in 45. Iyer,L.M., Abhiman,S., de Souza,R.F. & Aravind,L. affects target site demethylation. Mol. Cell. Biol. 20,
trypanosomatids has features of a thymidine Origin and evolution of peptide-modifying 23432349 (2000).
hydroxylase. Nucleic Acids Res. 35, 21072115 dioxygenases and identification of the wybutosine 71. Brandeis,M. etal. Sp1 elements protect a CpG island
(2007). hydroxylase/hydroperoxidase. Nucleic Acids Res. 38, from de novo methylation. Nature 371, 435438
21. Loenarz,C. & Schofield,C.J. Expanding chemical 52615279 (2010). (1994).
biology of 2oxoglutarate oxygenases. Nature Chem. 46. Bird,A. The dinucleotide CG as a genomic signalling 72. Stadler,M.B. etal. DNA-binding factors shape the
Biol. 4, 152156 (2008). module. J.Mol. Biol. 409, 4753 (2011). mouse methylome at distal regulatory regions.
22. Loenarz,C. & Schofield,C.J. Physiological and 47. Bostick,M. etal. UHRF1 plays a role in maintaining Nature 480, 490495 (2011).
biochemical aspects of hydroxylations and DNA methylation in mammalian cells. Science 317, 73. Szulwach,K.E. etal. Integrating
demethylations catalyzed by human 2oxoglutarate 17601764 (2007). 5Hydroxymethylcytosine into the epigenomic
oxygenases. Trends Biochem. Sci. 36, 718 (2011). 48. Sharif,J. etal. The SRA protein Np95 mediates landscape of human embryonic stem cells. PLoS
23. Aravind,L. & Koonin,E.V. The DNA-repair protein epigenetic inheritance by recruiting Dnmt1 to Genet. 7, e1002154 (2011).
AlkB, EGL9, and leprecan define new families of methylated DNA. Nature 450, 908912 (2007). 74. Wu,H. & Zhang,Y. Tet1 and 5hydroxymethylation:
2oxoglutarate- and iron-dependent dioxygenases. 49. Hashimoto,H. etal. Recognition and potential a genome-wide view in mouse embryonic stem cells.
Genome Biol. 2, RESEARCH0007 (2001). mechanisms for replication and erasure of cytosine Cell Cycle 10, 24282436 (2011).
24. Borst,P. & Sabatini,R. Base J: discovery, biosynthesis, hydroxymethylation. Nucleic Acids Res. 40, 75. Stroud,H., Feng,S., Morey Kinney,S., Pradhan,S. &
and possible functions. Annu. Rev. Microbiol. 62, 48414849 (2012). Jacobsen,S.E. 5hydroxymethylcytosine is associated
235251 (2008). 50. Valinluck,V. & Sowers,L.C. Endogenous cytosine with enhancers and gene bodies in human embryonic
25. van Luenen,H.G. etal. Glucosylated damage products alter the site selectivity of human stem cells. Genome Biol. 12, R54 (2011).
hydroxymethyluracil, DNA base j, prevents DNA maintenance methyltransferase DNMT1. 76. Pastor,W.A. etal. Genome-wide mapping of
transcriptional readthrough in leishmania. Cell 150, Cancer Res. 67, 946950 (2007). 5hydroxymethylcytosine in embryonic stem cells.
909921 (2012). 51. Kubosaki,A. etal. CpG site-specific alteration of Nature 473, 394397 (2011).
26. Ko,M. etal. Impaired hydroxylation of hydroxymethylcytosine to methylcytosine beyond DNA 77. Hahn,M.A. etal. Dynamics of
5methylcytosine in myeloid cancers with mutant replication. Biochem. Biophys. Res. Commun. 426, 5hydroxymethylcytosine and chromatin marks in
TET2. Nature 468, 839843 (2010). 141147 (2012). mammalian neurogenesis. Cell Rep. 3, 291300
27. Ito,S. etal. Role of Tet proteins in 5mC to 5hmC 52. Guo,J.U., Su,Y., Zhong,C., Ming,G.L. & Song,H. (2013).
conversion, EScell self-renewal and inner cell mass Hydroxylation of 5methylcytosine by TET1 promotes 78. Szulwach,K.E. etal. 5hmC-mediated epigenetic
specification. Nature 466, 11291133 (2010). active DNA demethylation in the adult brain. Cell 145, dynamics during postnatal neurodevelopment and
28. Pfaffeneder,T. etal. The discovery of 5formylcytosine 423434 (2011). aging. Nature Neurosci. 14, 16071616 (2011).
in embryonic stem cell DNA. Angewandte Chemie 50, 53. Zhang,L. etal. Thymine DNA glycosylase specifically 79. Weber,M. etal. Distribution, silencing potential and
70087012 (2011). recognizes 5carboxylcytosine-modified DNA. evolutionary impact of promoter DNA methylation in
29. Penn,N.W., Suwalski,R., ORiley,C., Bojanowski,K. & Nature Chem. Biol. 8, 328330 (2012). the human genome. Nature Genet. 39, 457466
Yura,R. The presence of 5hydroxymethylcytosine in Together with references 6 and 30, shows that TDG (2007).
animal deoxyribonucleic acid. Biochem. J. 126, excises 5fC and 5caC. This paper also shows a 80. Hackett,J.A. etal. Germline DNA demethylation
781790 (1972). crystal structure of TDG with 5caC. dynamics and imprint erasure through
30. Maiti,A. & Drohat,A.C. Thymine DNA glycosylase 54. Nabel,C.S. etal. AID/APOBEC deaminases disfavor 5hydroxymethylcytosine. Science 339, 448452
can rapidly excise 5formylcytosine and modified cytosines implicated in DNA demethylation. (2013).
5carboxylcytosine: potential implications for active Nature Chem. Biol. 8, 751758 (2012). Demonstrates that TET1 and TET2 oxidize 5mC
demethylation of CpG sites. J. Biol. Chem. 286, 55. Song,C.X. etal. Genome-wide profiling of enmasse in PGCs.
3533435338 (2011). 5formylcytosine reveals its roles in epigenetic 81. Thomson,J.P. etal. Non-genotoxic carcinogen
Together with references 6 and 53, shows that TDG priming. Cell 153, 678691(2013). exposure induces defined changes in the
excises 5fC and 5caC. 56. Shen,L. etal. Genome-wide analysis reveals TET- and 5hydroxymethylome. Genome Biol. 13, R93 (2012).
31. Schiesser,S. etal. Mechanism and stem-cell activity of TDG-dependent 5methylcytosine oxidation dynamics. 82. Lian,C.G. etal. Loss of 5hydroxymethylcytosine is an
5carboxycytosine decarboxylation determined by Cell 153, 692706 (2013). epigenetic hallmark of melanoma. Cell 150,
isotope tracing. Angewandte Chemie 51, 65166520 57. Rai,K. etal. DNA demethylation in zebrafish involves 11351146 (2012).
(2012). the coupling of a deaminase, a glycosylase, and 83. Raiber,E.A. etal. Genome-wide distribution of
32. Iyer,L.M., Abhiman,S. & Aravind,L. Natural history gadd45. Cell 135, 12011212 (2008). 5formylcytosine in ES cells is associated with
of eukaryotic DNA methylation systems. Prog. Mol. 58. Rai,K. etal. DNA demethylase activity maintains transcription and depends on thymine DNA
Biol. Transl. Sci. 101, 25104 (2011). intestinal cells in an undifferentiated state following glycosylase. Genome Biol. 13, R69 (2012).
33. Frauer,C. etal. Different binding properties and loss of APC. Cell 142, 930942 (2010). 84. Wu,H. etal. Genome-wide analysis of
function of CXXC zinc finger domains in Dnmt1 and 59. Popp,C. etal. Genome-wide erasure of DNA 5hydroxymethylcytosine distribution reveals its dual
Tet1. PLoS ONE 6, e16627 (2011). methylation in mouse primordial germ cells is affected function in transcriptional regulation in mouse
34. Lee,J.H., Voo,K.S. & Skalnik,D.G. Identification and by AID deficiency. Nature 463, 11011105 (2010). embryonic stem cells. Genes Dev. 25, 679684
characterization of the DNA binding domain of CpG- 60. Bhutani,N. etal. Reprogramming towards (2011).
binding protein. J.Biol. Chem. 276, 4466944676 pluripotency requires AID-dependent DNA 85. Mellen, M., Ayata, P., Dewell, S., Kriaucionis, S. &
(2001). demethylation. Nature 463, 10421047 (2010). Heintz, N. MeCP2 binds to 5hmC enriched within
35. Birke,M. etal. The MT domain of the proto- 61. Cortellino,S. etal. Thymine DNA glycosylase is active genes and accessible chromatin in the nervous
oncoprotein MLL binds to CpG-containing DNA and essential for active DNA demethylation by linked system. Cell 151, 14171430 (2012).
discriminates against methylation. Nucleic Acids Res. deamination-base excision repair. Cell 146, 6779 86. Lister,R. etal. Human DNA methylomes at base
30, 958965 (2002). (2011). resolution show widespread epigenomic differences.
36. Jorgensen,H.F., Ben-Porath,I. & Bird,A.P. Mbd1 is 62. Cortazar,D. etal. Embryonic lethal phenotype reveals Nature 462, 315322 (2009).
recruited to both methylated and nonmethylated a function of TDG in maintaining epigenetic stability. 87. Straussman,R. etal. Developmental programming of
CpGs via distinct DNA binding domains. Mol. Cell. Nature 470, 419423 (2011). CpG island methylation profiles in the human
Biol. 24, 33873395 (2004). 63. Bransteitter,R., Pham,P., Scharff,M.D. & genome. Nature Struct. Mol. Biol. 16, 564571
37. Blackledge,N.P. etal. CpG islands recruit a histone Goodman,M.F. Activation-induced cytidine (2009).
H3 lysine 36 demethylase. Mol. Cell 38, 179190 deaminase deaminates deoxycytidine on single- 88. Weber,M. etal. Chromosome-wide and promoter-
(2010). stranded DNA but requires the action of RNase. specific analyses identify sites of differential DNA
38. Allen,M.D. etal. Solution structure of the nonmethyl- Proc. Natl Acad. Sci. USA 100, 41024107 (2003). methylation in normal and transformed human cells.
CpG-binding CXXC domain of the leukaemia- 64. Rangam,G., Schmitz,K.M., Cobb,A.J. & Petersen- Nature Genet. 37, 853862 (2005).
associated MLL histone methyltransferase. EMBO J. Mahrt,S.K.AID enzymatic activity is inversely 89. Yu,M. etal. Base-resolution analysis of
25, 45034512 (2006). proportional to the size of cytosine C5 orbital cloud. 5hydroxymethylcytosine in the mammalian genome.
39. Xu,Y. etal. Genome-wide regulation of 5hmC, 5mC, PloS one 7, e43279 (2012). Cell 149, 13681380 (2012).
and gene expression by Tet1 hydroxylase in mouse 65. Liutkeviciute,Z., Lukinavicius,G., Masevicius,V., First genome-wide map of 5hmC at single-molecule
embryonic stem cells. Mol. Cell 42, 451464 (2011). Daujotyte,D. & Klimasauskas,S. Cytosine- resolution.
40. Zhang,H. etal. TET1 is a DNA-binding protein that 5methyltransferases add aldehydes to DNA. 90. Hsieh,C.L. Dependence of transcriptional repression
modulates DNA methylation and gene transcription Nature Chem. Biol. 5, 400402 (2009). on CpG methylation density. Mol. Cell. Biol. 14,
via hydroxylation of 5methylcytosine. Cell Res. 20, 66. Chen,C.C., Wang,K.Y. & Shen,C.K. The mammalian 54875494 (1994).
13901393 (2010). de novo DNA.methyltransferases,DNMT3A and 91. Serandour,A.A. etal. Dynamic hydroxymethylation of
41. Ko,M. etal. Modulation of TET2 expression and DNMT3B are alsoDNA 5hydroxymethylcytosine deoxyribonucleic acid marks differentiation-associated
5methylcytosine oxidation by the CXXC domain dehydroxymethylases. J.Biol. Chem. 287, enhancers. Nucleic Acids Res. 40, 82558265
protein IDAX. Nature 497, 122126 (2013). 3311633121 (2012). (2012).
Describes the role of IDAX in regulating TET2. 67. Metivier,R. etal. Cyclical DNA methylation of a 92. Wu,H. etal. Dual functions of Tet1 in transcriptional
42. Xu,Y. etal. Tet3 CXXC domain and dioxygenase activity transcriptionally active promoter. Nature 452, 4550 regulation in mouse embryonic stem cells. Nature
cooperatively regulate key genes for Xenopus eye and (2008). 473, 389393 (2011).
neural development. Cell 151, 12001213 (2012). 68. Kangaspeska,S. etal. Transient cyclical methylation of 93. Williams,K. etal. TET1 and hydroxymethylcytosine in
43. Hino,S. etal. Inhibition of the Wnt signaling pathway promoter DNA. Nature 452, 112115 (2008). transcription and DNA methylation fidelity. Nature
by Idax, a novel Dvl-binding protein. Mol. Cell. Biol. 69. Hsieh,C.L. Evidence that protein binding specifies 473, 343348 (2011).
21, 330342 (2001). sites of DNA demethylation. Mol. Cell. Biol. 19, Describes the distribution of TET1 and 5hmC
44. Yu,B. etal. Crystal structures of catalytic complexes of 4656 (1999). in EScells and demonstrates, using several
the oxidative DNA/RNA repair enzyme AlkB. Nature 70. Lin,I.G., Tomzynski,T.J., Ou,Q. & Hsieh,C.L. methods, that TET1 recruits SIN3A to target
439, 879884 (2006). Modulation of DNA binding protein affinity directly genes.

354 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

94. Deplus,R. etal. TET2 and TET3 regulate 116. Nakamura,T. etal. PGC7/Stella protects against DNA 141. Jin,S.G., Wu,X., Li,A.X. & Pfeifer,G.P. Genomic
GlcNAcylation and H3K4 methylation through OGT demethylation in early embryogenesis. Nature Cell mapping of 5hydroxymethylcytosine in the human
and SET1/COMPASS. EMBO J. 32, 645655 Biol. 9, 6471 (2007). brain. Nucleic Acids Res. 39, 50155024 (2011).
(2013). 117. Bortvin,A., Goodheart,M., Liao,M. & Page,D.C. 142. Matarese,F., Carrillode Santa Pau,E. &
95. Chen,Q., Chen,Y., Bian,C., Fujiki,R. & Yu,X. TET2 Dppa3 / Pgc7 / stella is a maternal factor and is not Stunnenberg,H.G. 5Hydroxymethylcytosine: a new
promotes histone OGlcNAcylation during gene required for germ cell specification in mice. BMC Dev. kid on the epigenetic block? Mol. Systems Biol. 7,
transcription. Nature 493, 561564 (2013). Biol. 4, 2 (2004). 562 (2011).
96. Yildirim,O. etal. Mbd3/NURD complex regulates 118. Payer,B. etal. Stella is a maternal effect gene required 143. Hayatsu,H. & Shiragami,M. Reaction of bisulfite
expression of 5hydroxymethylcytosine marked genes for normal early development in mice. Curr. Biol. 13, with the 5hydroxymethyl group in pyrimidines and
in embryonic stem cells. Cell 147, 14981510 21102117 (2003). in phage DNAs. Biochemistry 18, 632637
(2011). 119. Inoue, A. & Zhang, Y. Replication-dependent loss of (1979).
97. Vella,P. etal. Tet proteins connect the Olinked 5-hydoxymethylcytosine in mouse preimplantation 144. Huang,Y., Pastor,W.A., Zepeda-Martinez,J.A. &
Nacetylglucosamine transferase Ogt to chromatin in embryos. Science 334, 194 (2011). Rao,A. The anti-CMS technique for genome-wide
embryonic stem cells. Mol. Cell 49, 645656 (2013). 120. Hajkova,P. etal. Genome-wide reprogramming in the mapping of 5hydroxymethylcytosine. Nature Protoc.
98. Jackson-Grusby,L. etal. Loss of genomic methylation mouse germ line entails the base excision repair 7, 18971908 (2012).
causes p53dependent apoptosis and epigenetic pathway. Science 329, 7882 (2010). 145. Pastor,W.A., Huang,Y., Henderson,H.R., Agarwal,S.
deregulation. Nature Genet. 27, 3139 (2001). 121. Aoki,F., Worrad,D.M. & Schultz,R.M. Regulation of & Rao,A. The GLIB technique for genome-wide
99. Fouse,S.D. etal. Promoter CpG methylation transcriptional activity during the first and second cell mapping of 5hydroxymethylcytosine. Nature Protoc.
contributes to ES cell gene regulation in parallel cycles in the preimplantation mouse embryo. 7, 19091917 (2012).
with Oct4/Nanog, PcG complex, and histone H3 Dev. Biol. 181, 296307 (1997). 146. Robertson,A.B. etal. A novel method for the efficient
K4/K27 trimethylation. Cell Stem Cell 2, 160169 122. Inoue,A., Matoba,S. & Zhang,Y. Transcriptional and selective identification of 5hydroxymethylcytosine
(2008). activation of transposable elements in mouse in genomic DNA. Nucleic Acids Res. 39, e55 (2011).
100. Balasubramani,A. & Rao,A. OGlcNAcylation and zygotes is independent of Tet3mediated 147. Song,C.X. etal. Selective chemical labeling reveals the
5methylcytosine oxidation: an unexpected 5methylcytosine oxidation. Cell Res. 22, 16401649 genome-wide distribution of 5hydroxymethylcytosine.
association between OGT and TETs. Mol. Cell 49, (2012). Nature Biotechnol. 29, 6872 (2011).
618619 (2013). 123. Dawlaty,M.M. etal. Tet1 is dispensable for 148. Zilberman,D. & Henikoff,S. Genome-wide analysis of
101. Hart,G.W., Slawson,C., Ramirez-Correa,G. & maintaining pluripotency and its loss is compatible DNA methylation patterns. Development 134,
Lagerlof,O. Cross talk between OGlcNAcylation and with embryonic and postnatal development. Cell Stem 39593965 (2007).
phosphorylation: roles in signaling, transcription, and Cell 9, 166175 (2011). 149. Krueger,F., Kreck,B., Franke,A. & Andrews,S.R. DNA
chronic disease. Annu. Rev. Biochem. 80, 825858 124. Dawlaty,M.M. etal. Combined deficiency of Tet1 and methylome analysis using short bisulfite sequencing
(2011). Tet2 causes epigenetic abnormalities but is data. Nature Methods 9, 145151 (2012).
102. Fujiki,R. etal. GlcNAcylation of histone H2B facilitates compatible with postnatal development. Dev. Cell 24, 150. Rein,T., DePamphilis,M.L. & Zorbas,H. Identifying
its monoubiquitination. Nature 480, 557560 310323 (2013). 5methylcytosine and related modifications in DNA
(2011). Reports the phenotype of Tet1/ Tet2/ mice and genomes. Nucleic Acids Res. 26, 22552264
103. Mayer,W., Niveleau,A., Walter,J., Fundele,R. & cells and demonstrates defective imprinting in (1998).
Haaf,T. Demethylation of the zygotic paternal these mice. 151. Booth,M.J. etal. Quantitative sequencing of
genome. Nature 403, 501502 (2000). 125. Koh,K.P. etal. Tet1 and Tet2 regulate 5methylcytosine and 5hydroxymethylcytosine at
104. Oswald,J. etal. Active demethylation of the paternal 5hydroxymethylcytosine production and cell lineage single-base resolution. Science 336, 934937
genome in the mouse zygote. Curr. Biol. 10, 475478 specification in mouse embryonic stem cells. Cell Stem (2012).
(2000). Cell 8, 200213 (2011). 152. Song,C.X. etal. Sensitive and specific single-molecule
105. Santos,F., Hendrich,B., Reik,W. & Dean,W. 126. Ficz,G. etal. Dynamic regulation of sequencing of 5hydroxymethylcytosine. Nature
Dynamic reprogramming of DNA methylation in the 5hydroxymethylcytosine in mouse ES cells and during Methods 9, 7577 (2012).
early mouse embryo. Dev. Biol. 241, 172182 differentiation. Nature 473, 398402 (2011). 153. Clark,T.A. etal. Enhanced 5methylcytosine detection
(2002). 127. Vincent,J.J. etal. Stage-specific roles for Tet1 and in single-molecule, real-time sequencing via Tet1
106. Huang,Y. etal. The behaviour of Tet2 in DNA demethylation in primordial germ cells. oxidation. BMC Biol. 11, 4 (2013).
5hydroxymethylcytosine in bisulfite sequencing. Cell stem cell 12, 470478 (2013). 154. Quivoron,C. etal. TET2 inactivation results in
PLoS ONE 5, e8888 (2010). 128. Hackett,J.A., Zylicz,J.J. & Surani,M.A. Parallel pleiotropic hematopoietic abnormalities in mouse and
107. Nestor,C., Ruzov,A., Meehan,R. & Dunican,D. mechanisms of epigenetic reprogramming in the is a recurrent event during human lymphomagenesis.
Enzymatic approaches and bisulfite sequencing cannot germline. Trends Genet. 28, 164174 (2012). Cancer Cell 20, 2538 (2011).
distinguish between 5methylcytosine and 129. Yamaguchi,S. etal. Tet1 controls meiosis by 155. Moran-Crusio,K. etal. Tet2 loss leads to increased
5hydroxymethylcytosine in DNA. Biotechniques 48, regulating meiotic gene expression. Nature 492, hematopoietic stem cell self-renewal and myeloid
317319 (2010). 443447 (2012). transformation. Cancer Cell 20, 1124 (2011).
108. Jin,S.G., Kadam,S. & Pfeifer,G.P. Examination of Demonstrates that TET1 positively regulates gene 156. Ko,M. etal. Ten-eleven-translocation 2 (TET2)
the specificity of DNA methylation profiling techniques expression in PGCs and that TET1 deficiency negatively regulates homeostasis and differentiation
towards 5methylcytosine and impairs meiosis. of hematopoietic stem cells in mice. Proc. Natl Acad.
5hydroxymethylcytosine. Nucleic Acids Res. 38, e125 130. Takahashi,K. & Yamanaka,S. Induction of pluripotent Sci. USA 108, 1456614571 (2011).
(2010). stem cells from mouse embryonic and adult fibroblast 157. Li,Z. etal. Deletion of Tet2 in mice leads to
109. Okada,Y., Yamagata,K., Hong,K., Wakayama,T. & cultures by defined factors. Cell 126, 663676 (2006). dysregulated hematopoietic stem cells and
Zhang,Y. A role for the elongator complex in zygotic 131. Cox,J.L. & Rizzino,A. Induced pluripotent stem cells: subsequent development of myeloid malignancies.
paternal genome demethylation. Nature 463, what lies beyond the paradigm shift. Exp. Biol. Med. Blood 118, 45094518 (2011).
554558 (2010). 235, 148158 (2010). 158. Shide,K. etal. TET2 is essential for survival and
110. Wossidlo,M. etal. Dynamic link of DNA 132. Mikkelsen,T.S. etal. Dissecting direct reprogramming hematopoietic stem cell homeostasis. Leukemia 26,
demethylation, DNA strand breaks and repair in through integrative genomic analysis. Nature 454, 22162223 (2012).
mouse zygotes. EMBO J. 29, 18771888 (2010). 4955 (2008). 159. Ng,H.H. etal. MBD2 is a transcriptional repressor
111. Iqbal,K., Jin,S.G., Pfeifer,G.P. & Szabo,P.E. 133. Doege,C.A. etal. Early-stage epigenetic modification belonging to the MeCP1 histone deacetylase complex.
Reprogramming of the paternal genome upon during somatic cell reprogramming by Parp1 and Tet2. Nature Genet. 23, 5861 (1999).
fertilization involves genome-wide oxidation of Nature 488, 652655 (2012). 160. Kass,S.U., Landsberger,N. & Wolffe,A.P. DNA
5methylcytosine. Proc. Natl Acad. Sci. USA 108, 134. Costa,Y. etal. NANOG-dependent function of TET1 methylation directs a time-dependent repression of
36423647 (2011). and TET2 in establishment of pluripotency. Nature transcription initiation. Curr. Biol. 7, 157165
112. Wossidlo,M. etal. 5Hydroxymethylcytosine in the 495, 370374 (2013). (1997).
mammalian zygote is linked with epigenetic 135. Gao,Y. etal. Replacement of Oct4 by Tet1 during iPSC 161. Ng,H.H., Jeppesen,P. & Bird,A. Active repression of
reprogramming. Nature Commun. 2, 241 (2011). induction reveals an important role of DNA methylated genes by the chromosomal protein MBD1.
113. Gu,T.P. etal. The role of Tet3 DNA dioxygenase in methylation and hydroxymethylation in Mol. Cell. Biol. 20, 13941406 (2000).
epigenetic reprogramming by oocytes. Nature 477, reprogramming. Cell Stem Cell 12, 453469 (2013). 162. Fujita,N. etal. MCAF mediates MBD1dependent
606610 (2011). 136. Spruijt,C.G. etal. Dynamic readers for 5-(hydroxy) transcriptional repression. Mol. Cell. Biol. 23,
Together with reference 112, proves that TET3 is methylcytosine and its oxidized derivatives. Cell 152, 28342843 (2003).
responsible for mass methylcytosine oxidation in 11461159 (2013). 163. Sarraf,S.A. & Stancheva,I. Methyl-CpG binding
the male pronucleus. Reference 113 also 137. Otani,J. etal. Structural basis of the versatile DNA protein MBD1 couples histone H3 methylation at
establishes the phenotype of TET3-deficient recognition ability of the methyl-CpG binding domain lysine 9by SETDB1 to DNA replication and chromatin
oocytes. of methyl-CpG binding domain protein 4. J.Biol. assembly. Mol. Cell 15, 595605 (2004).
114. Inoue,A., Shen,L., Dai,Q., He,C. & Zhang,Y. Chem. 288, 63516362 (2013). 164. Prokhortchouk,A. etal. The p120 catenin partner
Generation and replication-dependent dilution of 5fC 138. Tini,M. etal. Association of CBP/p300 acetylase and Kaiso is a DNA methylation-dependent transcriptional
and 5caC during mouse preimplantation development. thymine DNA glycosylase links DNA repair and repressor. Genes Dev. 15, 16131618 (2001).
Cell Res. 21, 16701676 (2011). transcription. Mol. Cell 9, 265277 (2002). 165. Valinluck,V. etal. Oxidative damage to methyl-CpG
115. Nakamura,T. etal. PGC7 binds histone H3K9me2 to 139. Cortazar,D., Kunz,C., Saito,Y., Steinacher,R. & sequences inhibits the binding of the methyl-CpG
protect against conversion of 5mC to 5hmC in early Schar,P. The enigmatic thymine DNA glycosylase. binding domain (MBD) of methyl-CpG binding
embryos. Nature 486, 415419 (2012). DNA Repair 6, 489504 (2007). protein2 (MeCP2). Nucleic Acids Res. 32,
Shows that the maternal pronucleus of the zygote 140. Kellinger,M.W. etal. 5formylcytosine and 41004108 (2004).
and select loci on the paternal pronucleus are 5carboxylcytosine reduce the rate and substrate 166. Prokhortchouk,A. etal. Kaiso-deficient mice show
protected from TET3mediated oxidation by specificity of RNA polymerase II transcription. resistance to intestinal cancer. Mol. Cell. Biol. 26,
DPPA3. Nature Struct. Mol. Biol. 19, 831833 (2012). 199208 (2006).

NATURE REVIEWS | MOLECULAR CELL BIOLOGY VOLUME 14 | JUNE 2013 | 355

2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

167. Zhao,X. etal. Mice lacking methyl-CpG binding 176. Zheng,Y. etal. Role of conserved non-coding DNA human germ line reprogramming, imprint erasure and
protein 1 have deficits in adult neurogenesis and elements in the Foxp3 gene in regulatory Tcell fate. in vitro differentiation. Nature Cell Biol. 15, 113122
hippocampal function. Proc. Natl Acad. Sci. USA 100, Nature 463, 808812 (2010). (2013).
67776782 (2003). 177. Bell,A.C. & Felsenfeld,G. Methylation of a CTCF- 186. Reik,W., Dean,W. & Walter,J. Epigenetic
168. Hendrich,B., Guy,J., Ramsahoye,B., Wilson,V.A. & dependent boundary controls imprinted expression of reprogramming in mammalian development.
Bird,A. Closely related proteins MBD2 and MBD3 the Igf2 gene. Nature 405, 482485 (2000). Science293, 10891093 (2001).
play distinctive but interacting roles in mouse 178. Miranda,T.B. & Jones,P.A. DNA methylation: the
development. Genes Dev. 15, 710723 (2001). nuts and bolts of repression. J.Cell. Physiol. 213, Acknowledgements
169. Jones,P.L. etal. Methylated DNA and MeCP2 recruit 384390 (2007). The authors thank S. Evans and A. Clark for comments on
histone deacetylase to repress transcription. 179. Wu,H. etal. Dnmt3adependent nonpromoter DNA the manuscript. This work was supported by US National
Nature Genet. 19, 187191 (1998). methylation facilitates transcription of neurogenic Institutes of health (NIH) R01 grants AI44432, HD065812
170. Ben-Shachar,S., Chahrour,M., Thaller,C., Shaw,C.A. genes. Science 329, 444448 (2010). and CA151535, grant RM01729 from the California
& Zoghbi,H.Y. Mouse models of MeCP2 disorders 180. Brinkman,A.B. etal. Sequential ChIPbisulfite Institute of Regenerative Medicine, and Translational
share gene expression changes in the cerebellum and sequencing enables direct genome-scale investigation Research grant TRP 618712 from the Leukemia and
hypothalamus. Hum. Mol. Genet. 18, 24312442 of chromatin and DNA methylation cross-talk. Lymphoma Society (to A.R). Work in the laboratory of L.A. is
(2009). Genome Res. 22, 11281138 (2012). supported by intramural funds of the National Library of
171. Skene,P.J. etal. Neuronal MeCP2 is expressed at 181. Acosta-Silva,C., Branchadell,V., Bertran,J. & Medicine, NIH. W.A.P. was supported by a predoctoral grad-
near histone-octamer levels and globally alters the Oliva,A. Mutual relationship between stacking and uate research fellowship from the National Science
chromatin state. Mol. Cell 37, 457468 (2010). hydrogen bonding in DNA. Theoretical study of Foundation and is currently supported by a postdoctoral
172. Klose,R.J. & Bird,A.P. Genomic DNA methylation: guanine-cytosine, guanine-5methylcytosine, and fellowship from the Jane Coffin Childs Memorial Fund for
the mark and its mediators. Trends Biochem. Sci. 31, their dimers. J.Phys. Chem. B 114, 1021710227 Medical Research.
8997 (2006). (2010).
173. Lee,J.H. & Skalnik,D.G. CpG-binding protein 182. Thalhammer,A., Hansen,A.S., ElSagheer,A.H., Competing interests statement
(CXXC finger protein 1) is a component of the Brown,T. & Schofield,C.J. Hydroxylation of The authors declare no competing financial interests.
mammalian Set1 histone H3Lys4 methyltransferase methylated CpG dinucleotides reverses stabilisation of
complex, the analogue of the yeast Set1/COMPASS DNA duplexes by cytosine 5methylation. Chem.
complex. J.Biol. Chem. 280, 4172541731 Commun. (Camb.) 47, 53255327 (2011). FURTHER INFORMATION
(2005). 183. Borgel,J. etal. Targets and dynamics of promoter Anjana Raos homepage:
174. Choy,J.S. etal. DNA methylation increases DNA methylation during early mouse development. http://www.liai.org/pages/faculty-rao
nucleosome compaction and rigidity. J.Am. Chem. Nature Genet. 42, 10931100 (2010). Protein Data Bank: http://www.rcsb.org/pdb/home/home.do
Soc. 132, 17821783 (2010). 184. Lane,N. etal. Resistance of IAPs to methylation 2FD8
175. Watt,F. & Molloy,P.L. Cytosine methylation prevents reprogramming may provide a mechanism for
binding to DNA of a HeLa cell transcription factor epigenetic inheritance in the mouse. Genesis 35, SUPPLEMENTARY INFORMATION
required for optimal expression of the adenovirus 8893 (2003). See online article: S1 (figure)
major late promoter. Genes Dev. 2, 11361143 185. Gkountela,S. etal. The ontogeny of cKIT+ human
ALL LINKS ARE ACTIVE IN THE ONLINE PDF
(1988). primordial germ cells proves to be a resource for

356 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/molcellbio

2013 Macmillan Publishers Limited. All rights reserved

You might also like