Sie sind auf Seite 1von 14

REVIEWS

Gene expression regulation


mediated through reversible
m6A RNA methylation
Ye Fu1, Dan Dominissini1,2,3, Gideon Rechavi2,3 and Chuan He1
Abstract | Cellular RNAs carry diverse chemical modifications that used to be regarded as
static and having minor roles in fine-tuning structural and functional properties of RNAs.
In this Review, we focus on reversible methylation through the most prevalent mammalian
mRNA internal modification, N6-methyladenosine (m6A). Recent studies have discovered
protein writers, erasers and readers of this RNA chemical mark, as well as its dynamic
deposition on mRNA and other types of nuclear RNA. These findings strongly indicate
dynamic regulatory roles that are analogous to the well-known reversible epigenetic
modifications of DNA and histone proteins. This reversible RNA methylation adds a new
dimension to the developing picture of post-transcriptional regulation of gene expression.

Epigenetic modifications
In the central dogma of molecular biology, genetic infor- methylation, and emerging functions for m6A in several
Reversible chemical mation flows from DNA to RNA and then to protein. mechanisms of post-transcriptional regulation of gene
modifications on DNA and Reversible epigenetic modifications occur on genomic expression (FIG.1).
histones that regulate gene DNA15 and histone proteins69 to substantially regulate
expression independently of the
genome sequences and that are
gene expression that defines cell status and that affects m6A RNA methylation in eukaryotes
heritable through cell division. cell differentiation and development (FIG.1). Although Discovery and quantification of m6A in mRNAs and
both DNA and proteins are subject to reversible chemi- long non-coding RNAs in eukaryotes. Discovered in the
Writers, erasers and cal tuning, as we pointed out in 2010, a similar process 1970s, m6A is the most prevalent internal modification
readers
on mRNA or other forms of RNA as the third compo- in polyadenylated mRNAs and long non-coding RNAs
Enzymes or proteins that add,
remove or preferentially bind nent of the central dogma had been missing 10. RNA has (lncRNAs) in higher eukaryotes19. m6A is widely con-
to the chemical modifications crucial roles in biological systems not only by passing served among eukaryotic species that range from yeast,
at designated DNA or RNA genetic information from DNA to protein but also by plants, flies to mammals, as well as among viral RNAs
nucleotides and amino acid
regulating various biological processes. The diverse with a nuclear phase2024. The identified sequence content
residues of histones.
functions of RNA are accompanied by more than 100 of m6A obtained from mutational studies and substrate
1
Department of Chemistry
chemical modifications1114, although the functions of preference of the methyltransferase enzyme invitro2527
and Institute for Biophysical most of these RNA modifications have remained a mys- is [G/A/U][G>A]m6AC[U>A>C]. The amount of m6A in
Dynamics, The University tery. Most RNA species were thought to be short lived, isolated RNA was estimated to be 0.10.4% of that of ade-
of Chicago, 929 East 57th and RNA modifications were considered to be static and nines (that is, ~35m6A sites per mRNA) in mammals19,28
Street, Chicago, Illinois
60637, USA.
unalterable after their covalent attachment. The cen- and ~0.25% in meiotic Saccharomyces cerevisiae 29.
2
Cancer Research Center, tral role of RNA in gene expression and the intrinsic The total amount of m 6A in RNA can be probed
Chaim Sheba Medical Center, chemical reversibility of certain types of RNA meth- by several methods, including two-dimensional thin
Tel Hashomer 52621, Israel. ylation prompted us to raise the question of reversible layer chromatography30, dot-blot 15 and high-performance
3
Sackler School of Medicine,
Tel Aviv University, Tel Aviv
RNA modifications in gene expression regulation10. In liquid chromatography coupled with triple-quadrupole
69978, Israel. this Review, we discuss how, only in the past 23years, tandem mass spectrometry (HPLCQqQMS/MS)15,16. The
Correspondence to C.H. N6-methyladenosine (m6A) has been discovered as the femtomole sensitivity achieved by HPLCQqQMS/MS
email: first example of reversible RNA methylation15,16. We makes it a quantitative tool for monitoring m6A dynam-
chuanhe@uchicago.edu
doi:10.1038/nrg3724
describe the transcriptome-wide distribution of m6A ics; the purity of mRNA is extremely important for this
Published online in mammalian systems17,18, the identification of pro- measurement because ribosomal RNA (rRNA), small
25 March 2014 tein writers, erasers and readers for this dynamic RNA nuclear RNA (snRNA) and tRNA also containm6A.

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 293

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Methyltransferase Distribution of m6A in mammalian mRNAs and long Distribution of m 6A in mRNA in meiotic yeast. In
An enzyme that transfers a non-coding RNAs. Before 2012, the genome-wide dis- S.cerevisiae, m6A has an important role in the initiation
methyl group to its substrate. tribution of m6A was unknown, until two independent of meiosis, which is induced by nitrogen starvation31.
Most methyltransferases use studies developed an m 6A RNA immunoprecipitation Although yeast cells lack m6A (or contain very little of
S-adenosyl-l-methionine (SAM)
as the methyl donor.
approach followed by high-throughput sequencing it) in mRNA during the mitotic log phase, they begin to
(MeRIPseq) to map the m6A RNA methylomes with accumulate high levels of m6A during nitrogen starva-
Two-dimensional thin layer a ~100nucleotide resolution17,18. Briefly, the isolated tion29. Genome-wide mapping of m6A has revealed 1,308
chromatography mRNA is fragmented, immunoprecipitated using an methylation sites in 1,183 transcripts in an ndt80deficient
A technique to separate and
m6A-targeted antibody, ligated to sequencing adap- (ndt80/) strain that was arrested during meiotic G2
identify nucleosides on
cellulose plates according to
tors, reverse-transcribed to cDNA, amplified using phase or prophase32. The MeRIPseq approach was fur-
their differential migration PCR and subjected to high-throughput sequencing ther optimized through the use of shorter mRNA frag-
patterns in two different (FIG.2a). The resulting maps have shown that m6A is ments and an ime4/ strain (which lacks the inducer of
solvents. The nucleoside is widely distributed in more than 7,000 mRNA and 300 meiosis4 (Ime4) methyltransferase) as a negative control
typically radiolabelled for
detection.
non-coding RNA (ncRNA) transcripts in human cells, to obtain a map of m6A at higher resolution. The methyl-
and is enriched around stop codons, in 3 untranslated ated transcripts were found to be less structured, and most
High-performance liquid regions (3UTRs) and within internal long exons (FIG.2b). of them encode functions that are particularly related to
chromatography coupled Additionally, the presence of m6A in introns suggests meiosis. These transcripts have a consensus sequence
with triple-quadrupole
that this modification can be added either before or at of ANRGm6ACNNU (where R denotes A or G, and N
tandem mass spectrometry
(HPLCQqQMS/MS). A liquid
the same time as RNA splicing. Many m6A peaks are represents any nucleotide), and their methylation sites
chromatography method well conserved between humans and mice, and dynamic are enriched at the 3 ends, which is similar to those of
coupled with triple-quadrupole changes of certain peaks have been observed under dif- the mammalian systems. Thus, the distribution pattern
tandem mass spectrometry, ferent stress conditions. A potential link between m6A of m6A and the consensus sequence of these sites seem
which can quantitatively and
simultaneously monitor
and microRNA-target sites was also suggested18. Several to be conserved to a large extent from yeast tohumans.
multiple molecular species lncRNAs contain m6A, which indicates that certain
according to their ncRNAs transcribed by RNA polymerase II are also sub- Quantitative detection of the m6A fraction with single-
fragmentation patterns. ject to m6A methylation. This approach has since been nucleotide resolution. The antibody-based profiling of
widely adopted in studies of transcriptome-wide m6A m6A could not provide information at single-nucleotide
distribution. resolution: although this method determines RNA frag-
ments that harbour m6A, the difficulty in distinguishing
Central dogma Chemical modications m6A from unmodified adenines by sequencing hinders
the pinpointing of m6A sites within these fragments.
DNA replication Multiple RRACH (where HdenotesU, A or C) motifs
Reversible m5C hm5C
DNA methylation could be present adjacent to each other, and the modi-
fication may also occur at non-consensus sites. In addi-
DNA
tion, antibody-based profiling cannot reveal the fraction
of cellular RNAs that are modified at each specific site.
Traditionally, radioactive labelling was used to detect
Transcription
the modification site; however, this procedure is long
and tedious. A digestion-based method has recently
? m6A hm6A been developed to determine the percentage of m6A at a
Reversible
RNA methylation specific site with single-nucleotide resolution33. Termed
?
RNA site-specific cleavage and radioactive labelling followed
by ligation-assisted extraction and thin layer chromatog-
raphy (SCARLET), this method uses RNase H guided
Translation by a sequence-specific 2OMe/2-H chimeric oligonu-
cleotide to cleave the 5 end of the candidate site for
Reversible histone
methylation or
Me Ac subsequent labelling and detection. Application of this
acetylation method to two lncRNAs and three mRNAs revealed
Protein genuine m6A sites and quantified the methylation frac-
tions (1177%). These results, together with the 20%
of m6A modification that was previously reported in
bovine prolactin mRNA34, indicate that many m6A sites
in mRNA and lncRNA are incompletely methylated. In
Figure 1 | Reversible chemical modifications that regulate the flow of genetic fact, the majority of m6A consensus sequence sites are
Naturefrom
information. In the central dogma, genetic information is passed Reviews Genetics
DNA |to RNA
not methylated in mammalian mRNA17,18.
and then to protein. Epigenetic DNA modifications (for example, the formation of
5methylcytosine (m5C; also known as 5mC) and 5hydroxymethylcytosine (hm5C;
Methylation on the N6 position of adenosine
also known as 5hmC)) and histone modifications (for example, methylation (me) and slightly reduces the stability of WatsonCrick A:U
acetylation (ac)) are known to have important roles in regulating cell differentiation base pairing 35, but it does not noticeably block the
and development. Reversible RNA modifications (for example, the formation of extension activities of most reverse transcriptases.
N6-methyladenosine (m6A) and N6-hydroxymethyladenosine (hm6A)) add an additional Therefore, methods based on primer extension cannot
layer of dynamic regulation of biological processes. be readily used to map precise modification positions.

294 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Me Me a genome scale is still limited, and a method for the


genome-wide mapping of m6A with single-nucleotide
mRNA A A A A A A An resolution thus remains highly desirable.
Fragmentation m6A writers in eukaryotes
to ~100 nucleotides
METTL3 is an active component of the m6A methyl-
Me A A transferase complex in mammalian cells. m6A mRNA
Me
A methylation is catalysed by a multicomponent methyl
A A A A A transferase complex, which was originally isolated as
~200kDa and ~800kDa subcomplexes from HeLa
Immunoprecipitation
with m6A-specic Input control
nuclear extracts25,38. A 70kDa protein METTL3 (known
antibodies as MTA70 when identified) was the only known com-
ponent characterized38. METTL3 is highly conserved in
eukaryotes from yeast to humans (FIG.3a). Knockdown
Me of METTL3 in HeLa cells led to a ~30% decrease of the
A
Me Me A A total m6A level, and the same knockdown experiment
A
in HepG2 cells induced apoptosis, possibly through the
A

activation of the p53mediated pathway 17,39. A recombi-


A

nant FLAG-tagged human METTL3 protein has recently


been shown to exhibit a low level of activity by itself.
cDNA library construction and Other components are required to achieve optimal
high-throughput sequencing activity invitro40.

m6A RNA immunoprecipitation signal Input signal METTL14 is another active component of the m6A
methyltransferase complex and forms a stable hetero
m6A peak m6A peak complex with METTL3. A phylogenetic analysis of the
METTL3 family of methyltransferases in the human
Signal

Signal

genome identified METTL14 and METTL4 as close


homologues of METTL3 with a conserved motif that
contains either Asp-Pro-Pro-Trp or Glu-Pro-Pro-Leu41
Locus Locus (FIG. 3a) . We found that knockdown of METTL14,
but not METTL4, leads to decreased m6A levels in
HeLa and 293FT cells40. Biochemical characterization
b revealed that these two proteins form a stable complex
with a stoichiometric ratio of 1:1. Although the methyl-
ation activity of METTL14 is slightly higher than that of
m6A abundance METTL3 invitro, the combination of both methyltrans
ferases leads to a substantially enhanced methylation
activity. This heterodimer also shows a strong prefer-
ence for the cognate m6A consensus sequence and a
modest preference for less structured RNA invitro.
mRNA 5 3 METTL3 and METTL14 colocalize in nuclear speckles,
5 transcription Long exon Stop codon and the heterodimer forms the core of the mamma-
start site lian methyltransferase complex. Additional features
of METTL14 include glycinerich sequences in its car-
Figure 2 | Profiling of m6A in RNA by m6A RNA immunoprecipitation.
Nature Reviews
Antibody-based N6-methyladenosine (m6A) RNA immunoprecipitation | Genetics
has been
boxyl terminus and a potential coiled-coil in its amino
developed to profile the transcriptome-wide distribution of m6A. a | Isolated mRNA is terminus (FIG.3a), which may participate in protein
fragmented to ~100nucleotides, immunoprecipitated using m6A-specific antibodies, protein interactions in nuclear speckles. The binding
converted to a cDNA library and subjected to high-throughput sequencing. sites of METTL3 and METTL14 in substrate RNAs,
Comparison between the immunoprecipitated sample and the input sample identifies as shown by a photoactivatable ribonucleoside-enhanced
m6A signal peaks. b | Transcriptome-wide profiling of m6A in mRNA revealed that m6A crosslinking and immunoprecipitation (PARCLIP) assay,
is enriched around stop codons, at 3 untranslated regions and within long exons. contain a similar consensus sequence to that known
The 5 cap contains the N6,2Odimethyladenosine (m6Am) modification, which can for m6A (FIG.3b). Interestingly, silencing of the methyl-
also be enriched using the m6A-specific antibody. Me, methyl group. transferase complex led to an increase in the abundance
and half-lives of their target RNAs, which is consistent
with an emerging role for m6A as a negative regula-
However, Thermus thermophilus DNA polymerase I36 tor of gene expression (see below). A related study in
and HIV reverse transcriptase37 show kinetic differ- mouse embryonic stem cells (mESCs) also indicates that
ences when extending opposite m6A compared with METTL3 and METTL14 work as a complex 42.
unmodified adenines, and they could be used to map The discovery of the second active methyltransferase
m6A positions36,37. The application of this approach at component in the core complex raises the following

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 295

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

m6A RNA a DPAW SAM-binding b Consensus motif


immunoprecipitation S. cerevisiae Ime4 METTL14
An immunoprecipitation
method to selectively enrich DPPW SAM-binding
for N6-methyladenosine Human METTL3
(m6A)-containing RNA using
an m6A-targeted antibody. Coiled-coil EPPL SAM-binding
1 2 3 4 5
Human METTL14
Nuclear speckles
G-rich
Nuclear domains located in
the interchromatin regions METTL3
of the nucleoplasm and
c
enriched with pre-mRNA E. coli AlkB
processing factors.
Coiled-coil
Photoactivatable ribonucle- Human ALKBH5 1 2 3 4 5 6
oside-enhanced crosslinking A-rich
and immunoprecipitation C-terminal domain
(PARCLIP). A biochemical WTAP
Human FTO
method that takes advantage
of incorporated photoreactive
ribonucleoside analogues to
identify the binding sites of AlkB domains Fe(II)-binding Substrate and -KG Extra
motif (HXDXnH) binding (RXXXXXR) loop
RNA-binding proteins in cells. 1 2 3 4 5 6

d METTL3 METTL14

+ other
factors?
H H H
N N H WTAP N N C H m6A reader
H
N N N N
N N
Functions
ALKBH5
A m6A

FTO

H O FTO H OH
N N C N N C H
H H
N N N N
N N

f6A hm6A
Readers?

Figure 3 | Reversible m6A methylation of mRNA and other types of nuclear RNA. The N6-methyladenosine (m6A)
Nature Reviews | Genetics
modification is installed by a hetero complex of two methyltransferases METTL3METTL14, assisted by Wilms
tumour1associating protein (WTAP), and can be demethylated by the -ketoglutarate (-KG)-dependent
dioxygenases FTO and ALKBH5. a | Saccharomyces cerevisiae inducer of meiosis4 (Ime4), and human METTL3 and
METTL14 contain the S-adenosyl-l-methionine (SAM)-dependent methyltransferase domain for m6A methylation. The
(D/E)P(P/A)(W/L) active site and the SAM-binding motif are conserved. b | Photoactivatable ribonucleoside-enhanced
crosslinking and immunoprecipitation (PARCLIP) reveals that the binding sites of METTL14 and METTL3 on mRNA
resemble the consensus sequence of m6A in mammalian mRNA. The sequence bound by WTAP moderately overlaps
with those bound by METTL14 and METTL3. c | Mammalian FTO and ALKBH5 contain the active site motif HXDXnH
(where X denotes any amino acid) for Fe(ii) binding, RXXXXXR for both -KG binding and substrate recognition, and an
extra loop that leads to preferential binding of single-stranded over double-stranded nucleic acids68,121,122. Relative to
Escherichia coli AlkB, mammalian ALKBH5 has an aminoterminal alaninerich sequence and a potential coiled-coil
structure that could be important for its localization. FTO contains an extra carboxyterminal domain with a novel fold,
possibly to engage in additional proteinprotein interactions. d | Methylation and demethylation of m6A on RNA are
shown. Whereas ALKBH5 catalyses the direct removal of m6A, FTO can oxidize m6A to N6-hydroxymethyladenosine
(hm6A) and N6-formyladenosine (f6A) sequentially; hm6A and f6A are moderately stable (with half-lives of ~3hours under
physiological conditions) and can be hydrolysed to adenine.

296 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

question: why is the m6A methyltransferase complex to interact with many proteins and lncRNAs51, could
composed of two active components, both of which bind also recruit other proteins or enzymes to the methyl-
to the methyl donor S-adenosyl-l-methionine (SAM)? transferase complex; these additional factors may affect
The hetero complex may allow the selective tuning the methylation activity and selectivity through direct
of methylation activity through post-translational interactions or post-translational modifications. Future
modification of each component in order to affect dif- research to identify additional factors that interact with
ferent substrate transcripts, thus having an influence or modify the two methyltransferases is crucial for under-
on different biological pathways. A heterodimer of two standing the selectivity of m6A deposition. We may then
methyltransferase components is required for optimal be able to answer questions such as how do cells choose
activities of several other known RNA methyltrans- to methylate certain RNA sites, and how is m6A tar-
ferase complexs4346. Typically, one subunit has a SAM- geted to 3UTRs and long exons. The potential interplay
binding pocket, and the other non-catalytic subunit between RNA methylation, transcriptional regulation
either stabilizes the catalytic subunit or enhances its and splicing could also be further investigated.
activity by forming a continuous substrate-binding sur-
face. However, both METTL3 and METTL14 are active. Mum2Ime4Slz1 (MIS) complex in yeast mediates
A crystal structure of this complex will be helpful in mRNA methylation during meiosis. Ime4 is the homo-
uncovering the synergy between the two enzymes and logue of METTL3 in yeast and is crucial for the induction
the properties associated with each active component. of yeast sporulation. Two other components of the meth-
ylation complex Mum2 and sporulation specific with
WTAP is the third crucial component of the m6A methyl- a leucine zipper motif protein 1 (Slz1) have been iden-
transferase complex invivo. Yeast two-hybrid screens have tified through yeast two-hybrid experiments48. Mum2
identified FKBP12interacting protein of 37kDa (FIP37; is homologous to human WTAP, whereas Slz1 lacks
also known as AT3g54170) in Arabidopsis thaliana47 and mammalian homologues. Interestingly, the increase in
Mum2 in yeast as the partner proteins of the METTL3 m6A levels during meiosis is mainly triggered by Ime1
homologues in these organisms48. These two proteins (a master regulator of meiosis), which transcriptionally
are homologues of the Wilms tumour 1associating pro- induces SLZ1. Ime4 and Mum2 are expressed before the
tein (WTAP) in humans. WTAP was initially identified induction of meiosis, and Slz1 then recruits them from
as a splicing factor that binds to the Wilms tumour1 the cytoplasm to the nucleolus32. This nucleolar localiza-
(WT1) protein49, and it is essential for cell cycle progres- tion of the MIS complex is essential for accumulating the
sion and early mammalian embryonic development. We full level of m6A. In contrast to yeast, mammalian cells
found that knockdown of WTAP leads to a decrease in lack homologues of Slz1, and the mammalian and plant
the total m6A level in HeLa and 293FT cells40. WTAP methyltransferase complex primarily locates in nuclear
interacts with both METTL3 and METTL14, and colo- speckles instead of the nucleolus.
calizes with the METTL3METTL14 heterodimer in METTL3 and WTAP are highly conserved in eukary-
nuclear speckles to participate in m6A RNA methylation otes. In A.thaliana, m6A seems to be mainly found near
(FIG.3d). In fact, knockdown of WTAP leads to the largest 3UTRs52; a mutation in MTA (which is the METTL3
decrease in m6A levels in these cell lines, which indicates homologue in A.thaliana) has been associated with
that WTAP has important roles in cellular m6A depo- cell division defects, arrested seeds, reduced apical
sition. A PARCLIP assay revealed that WTAP shares dominance and organ abnormality 47. In Drosophila
a similar binding sequence of GACU; that is, the sequence melanogaster, the METTL3 homologue Ime4 is essen-
bound by WTAP moderately overlaps with the GGAC tial for viability and regulates Notch signalling during
sequence bound by both METTL3 and METTL14 egg chamber development53. In zebrafish, knockdown
(FIG.3b). As identified by PARCLIP, these targets have of either WTAP or METTL3 leads to multiple develop-
a ~50% overlap with m6A-containing transcripts, which mental defects, and knockdown of both proteins leads
further indicates that METTL3, METTL14 and WTAP to increased apoptosis50.
form the core of the major cellular writer complex of m6A
(REF.40). A large proportion of the binding sites of these m6A erasers in mammals
three proteins are found in introns (2934%), which fur- Demethylation of m6A in RNA by FTO. In 2011, the dis-
ther implies that methylation occurs cotranscriptionally covery of -ketoglutarate-dependent dioxygenase FTO
Yeast two-hybrid screens
either before or at the same time as splicing. As WTAP as the first RNA demethylase was an important break-
A method in which one protein has been thought to be a splicing factor, a recent study through in reigniting investigations of m6A biology 15.
is fused to the GAL4 activation indicates that the knockdown of WTAP or METTL3 The Fto gene was initially discovered in a deletion of
domain and the other to the yields different isoforms of m6A-containing transcripts, four genes that led to a fused-toe phenotype in mutant
GAL4 DNA-binding domain,
which suggests that methylation could affect splicing 50. mice54. In 2007, three independent studies revealed that
and both fusion proteins
are introduced into yeast. How does WTAP enhance the methylation activity a single-nucleotide polymorphism in the first intron of
Expression of a GAL4regulated of METTL3 and METTL14 invivo (FIG.3d)? Potentially, FTO strongly associates with body mass index and the
reporter gene indicates that the WTAP may help to recruit METTL3 and METTL14 to risk of obesity in multiple populations5557. In adult mice,
two proteins physically interact. their target mRNAs. WTAP has also been shown to be Fto has the highest expression level in the brain, particu-
Demethylase
essential for the nuclear speckle localization of METTL3 larly within the hypothalamus58. Deletion or overexpres-
An enzyme that removes a and METTL14, which could affect the methylation sion of Fto in mouse models has been associated with
methyl group from its substrate. efficiency of these proteins50. WTAP, which is known altered body weight or food intake59,60. Fto also affects

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 297

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

development: Fto-knockout mice shows increased post- mRNA synthesis and the rate of splicing 16. Unlike FTO,
natal lethality and growth retardation59, and a homozy- direct immunoprecipitation of ALKBH5 has identified
gous lossoffunction mutation (Arg316Gln) in the FTO bound RNA substrates16, and ALKBH5 has been shown
protein in humans leads to postnatal retardation, as well to be part of the mRNA-bound proteome70, which sug-
as multiple dysmorphisms and malformations61. gests a tight interaction with mRNA and other RNA
FTO is a member of the Fe(ii) and -ketoglutarate- substrates. ALKBH5 also colocalizes well with nuclear
dependent AlkB family of proteins that catalyse oxidative speckles in an RNase Asensitive manner. Alkbh5 has the
demethylation 58 ; close homologues participate in highest expression level in mouse testes. Consistently,
epigenetic regulation, such as oxidative DNA demeth- Alkbh5knockout male mice exhibit aberrant spermato-
ylation6264 and histone demethylation8. FTO was origi- genesis, which is probably a result of altered expression
nally shown to demethylate N3-methylthymidine in of spermatogenesis-related genes16.
single-stranded DNA58 and N3-methyluridine in single-
stranded RNA65 invitro; however, the function of FTO hm6A and f6A modifications on mammalian mRNA.
invivo remained unknown until we discovered that While investigating FTO-catalysed demethyla-
FTO efficiently demethylates m6A in both RNA and tion, we observed two unprecedented intermedi-
DNA invitro15. Further experiments showed that silenc- ates, N 6 -hydroxymethyladenosine (hm 6 A) and
ing of FTO in HeLa and 293FT cells increased total m6A N 6-formyladenosine (f 6A), which were generated
levels in polyadenylated RNA, and overexpression of through the FTO-catalysed oxidation of m6A (REF.71)
FTO decreased m6A levels on RNA15. FTO is expressed in (BOX 1;FIG.3d). The hm6A intermediate is a direct oxi-
dot-like patterns in the nucleoplasm and partially colo- dation product of m6A, and f 6A is the further oxidized
calizes with nuclear speckles. These cell-based results, product of hm6A. Both hm6A and f 6A can decompose in
together with observations that most mammalian cells water to yield unmethylated adenine and formaldehyde
and tissues contain very low levels (a few parts per million) (from hm6A) or formic acid (from f 6A). To our surprise,
of m6A on genomic DNA, led us to conclude that m6A these modifications are metastable under physiological
on nuclear RNA (including mRNA, lncRNA and pos- conditions in neutral buffered solutions at 37C with
sibly other types of RNA) is the main substrate of FTO. half-lives of ~3hours. This observation raised the pos-
Recent work showed that m6A on three mRNA species sibility that both modifications could exist in living
could be demethylated by FTO invivo, and this function cells and could have functional implications, given that
seems to affect neuronal activities66. FTO may also act median mammalian RNA half-lives are ~5hours72,73.
as a nutrient sensor, which could modulate its demeth- Indeed, using a modified protocol to avoid acid, base
ylation activities67. It should be noted that although and heating treatments, we have detected the presence
FTO works preferentially on single-stranded RNA of these modifications in mRNA isolated from mouse
and DNA, it can still exhibit demethylation activity, tissue and human cell lines71. The exact sources of these
albeit low, towards double-stranded RNA andDNA15. modifications invivo remain unknown so far; however,
The crystal structure of the FTO protein reveals these new modifications carry functional groups that
an active domain that is similar to those of other pro- are distinct from m6A and could substantially affect
teins of the AlkB family 68 (FIG.3c). FTO also contains a RNAprotein interactions.
Cterminal domain with a novel fold that is distinct from Although both FTO and ALKBH5 are mainly found
other proteins of this family. This Cterminal domain in the nucleus, the possibility that both proteins could
may engage in additional proteinprotein or protein translocate to the cytoplasm under certain circumstances
RNA interactions to affect the function of FTO. The should not be ruled out. Cytoplasmic RNA may also be
discovery of FTO as an m6A demethylase strongly sug- demethylated by these enzymes or by other currently
gests functional roles for m6A in human developmental unknown demethylases. ALKBH5 is only conserved in
regulation; however, to achieve the end goal of uncover- vertebrates from fish to humans, whereas FTO is con-
ing the underlying mechanism, a considerable amount served in vertebrates and has homologues in marine
of future work is required to identify the physiological algae74. As there are many Fe(ii) and -ketoglutarate-
RNA targets of FTO and to elucidate the functional dependent dioxygenases with unknown functions in var-
consequences of such demethylation. ious organisms75, we should not be surprised to see the
discovery of more m6A demethylases. In addition to its
Demethylation of m6A in RNA by ALKBH5. ALKBH5 occurrence in eukaryotic mRNA, m6A also exists in vari-
is another protein of the AlkB family that shows effi- ous classes of RNA in eukaryotes, bacteria and archaea,
cient demethylation activity towards m 6A in mRNA including rRNA, tRNA and snRNA 14. Furthermore,
and other types of nuclear RNA16,69. ALKBH5 has an chemical modifications can also occur on various nitro-
alanine-rich sequence and a potential coiled-coil struc- gen, carbon and oxygen atoms within the bases and
Oxidative demethylation
A chemical reaction in which ture in its Nterminus (FIG.3c), which may be important backbone of RNA13 (BOX 1). These modifications (for
the CH bond of a methyl for its localization. ALKBH5 knockdown in human cell example, methylation) on the heteroatoms oxygen and
group attached to a nitrogen or lines led not only to increased total m6A levels on poly- nitrogen can, in principle, be enzymatically reversed
an oxygen atom is oxidized to adenylated RNA but also to accelerated export of these through the oxidative demethylation mechanism used
OH by demethylases, and the
intermediate decomposes to
RNAs from the nucleus to the cytoplasm16. However, by FTO and ALKBH5 or through nucleophilic substitu-
release the methyl group as the underlying mechanism is not fully understood. tions (BOX 1). Demethylases that remove these other RNA
formaldehyde. ALKBH5 and its demethylation activity affect nascent methylations could exist and exhibit functionalroles.

298 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | RNA modifications


Cellular RNA species contain more than 100 chemical modifications with -ketoglutarate as a cofactor. When the methyl group is linked to
diverse properties. Chemical modifications of RNA can occur on the N1, a heteroatom such as nitrogen or oxygen, the oxidation of CH to a
N3, N7 and C8 atoms in both adenine and guanine; C2 and N6 in adenine; hemiaminal or hemiacetal intermediate destabilizes the CN or CO
N2 and O6 in guanine; N1, O2, N3 and C5 in cytosine and uracil; N4 in bond, respectively, which leads to the demethylated product with the
cytosine and O4 in uracil; as well as on 2O of the ribose backbone and the release of formaldehyde. The hemiaminal intermediate, such as
OH group of the phosphate backbone (see the figure, part a). These N6-hydroxymethyladenosine (hm6A), may undergo further oxidation to
modifications can modulate hydrophobicity, steric and electrostatic produce a formamide, such as N6-formyladenosine (f 6A), which can
effects, and hydrogen-bonding abilities of RNA bases and backbones. decompose in water to yield the demethylated product with the release of
Methylation or other forms of alkylation on nitrogen or oxygen atoms can formic acid. The demethylation activity could be modulated by the
be removed through either an oxidative or a nucleophilic substitution effective concentrations of Fe(ii), O2 or -ketoglutarate. The bimolecular
mechanism. The oxidative demethylation (see the figure, part b) is best nucleophilic substitution (Sn2) mechanism could also be used to remove
exemplified by Fe(ii) and -ketoglutarate-dependent dioxygenase RNA methylation on heteroatoms; however, such a process has yet to be
enzymes, which use Fe(ii) as a catalytic centre, O2 as an oxidant and shown for RNA demethylation (see the figure, part c).

a
NH2 O NH2 O Ribose O
Base
N N O P O O
N NH N NH
O
N N N N NH2 N O N O O OH

Ribose Ribose Ribose Ribose Phosphate


A G C U RNA backbone

b O
HC H H O
+ C
N N
OH
(e.g. f6A) (e.g. Adenine) (Formic acid)
c
Oxidation
N CH3 N H
OH
CH3 H2C H :Nu + Nu CH3
N N N
CH3 H
(e.g. m6A) (e.g. Adenine) O O
(e.g. hm6A) H O
Oxidation C
+
H
OH
(Formaldyhyde)
CH3 CH2 H
O O O

m6A, N6-methyladenosine; Nu, nucleophile.


Nature Reviews | Genetics

m6A reader proteins and effector functions m6A-containing RNA; second, the presence of m6A in
The discoveries of m 6A RNA demethylation and a specific sequence could weaken the cognate binding
demethylases validate our hypothesis that the ubiqui- interaction of an RNA-binding protein; and third, the
tous m6A modification is dynamic and reversible, which presence of m6A may change the secondary structures
is similar to epigenetic DNA and histone modifications. of RNA and therefore alter proteinRNA interactions.
The noticeable phenotypes of both FTO and Alkbh5
mutations in humans and mice strongly indicate the YTHDF2 preferentially recognizes m 6A-containing
functional importance of this reversible m6A methyla- mRNA and regulates both mRNA stability and localiza-
tion on RNA. For the m6A group to have a biological tion. Using pulldown experiments, we have identified
function, it needs to be recognized through reading three cytoplasmic proteins of the YTH domain fam-
by specific proteins. This process could resemble the ily, YTHDF13, as selective m6A-binding proteins in
roles of proteins that read 5methylcytosine (m5C; also mammalian cell extracts17,76 (FIG. 4a). The YTH domain
known as 5mC) in DNA, or methylated or acetylated family consists of abundant RNA-binding proteins that
amino acid residues of histones in order to exhibit previously had no clear function assigned. We con-
the biological function associated with the modifica- firmed that mammalian YTHDF proteins preferentially
tions and to enable reversible tuning. We can envision bind to RNA that contains m6A at the G[G>A]m6ACU
three types of selective reading mechanisms for m6A on consensus sequence relative to unmethylated RNA of
RNA: first, a reader protein could selectively bind to the the same sequence76. Additionally, RNA probes that

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 299

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

a conserved GAC[U>A] motif; thus, the occupancy of


S. cerevisiae Mrb1 YTH domain YTHDF2 resembles the distribution pattern of m 6A
on mRNA. Notably, the knockdown of YTHDF2 led to
S. pombe Mmi1 YTH domain
decreased half-lives of these RNA targets but had minor
Human YTHDF1 P/Q/N-rich YTH domain effects on the mRNA levels in the actively translating
pool. Ribosome profiling further suggests that YTHDF2
Human YTHDF2 P/Q/N-rich YTH domain alters ribosome occupancy of its mRNA targets. These
Human YTHDF3 P/Q/N-rich YTH domain results suggest that YTHDF2 has a role in RNA decay.
Fluorescence immunostaining of YTHDF2 and fluores-
cence insitu hybridization of its cognate mRNA revealed
b 600 that YTHDF2 binds to m6A through the Cterminal
Non-m6A YTH domain and localizes the cognate mRNA to
500 m6A processing bodies (Pbodies) for accelerated degradation
Number of dierent mRNAs

through its Nterminal Pro/Gln/Asnrich domain (FIG. 4).


400 The exact RNA degradation mechanism needs to be
further elucidated; however, YTHDF2 binds to mRNA
300 with shorter poly(A) tails and does not seem to affect the
deadenylation process76.
200 Several cytoplasmic mRNA decay pathways are
known7786. The YTHDF2mediated mRNA degrada-
100 tion, which affects thousands of mRNA molecules, is
a unique process that is dependent on the methylation
0 of the target mRNA and could therefore be reversibly
0 3 6 9 12 15 18 tuned through m6A methylation and demethylation.
mRNA lifetime (hours) This discovery, together with the negative correlation
of m6A with mRNA stability in general as revealed by
knockdown of methyltransferases40, suggests one main
c Translation function of m6A on RNA: the regulated degradation
of methylated RNA. This process is mediated through
? P/Q/N- selective m6A recognition and subsequent relocaliza-
rich
tion by a reader or effector protein. The control of the
Me YTHDF2 YTH stability of the non-translating pool of mRNA (or other
m7Gppp An
Me RNA species) through the YTHDF2dependent mecha-
RNA nism could be important under various circumstances
Transcription m7Gppp An
for the selective elimination of a group of RNAs77.
?
Interestingly, Mmi1 the homologue of YTHDF pro-
DNA teins in Schizosaccharomyces pombe (FIG.4a) is essen-
Localization Degradation tial for the elimination of meiosis-specific transcripts
(strorage or transport) during meiosis87. However, the presence of m6A has not
been reported in S.pombe, which lacks homologues of
Figure 4 | Functions of the reader (that is, effector) proteins of m6A. a | The METTL3 and METTL14. The potential presence of m6A
characterized YTHDF proteins serve as N6-methyladenosine (m Nature
6 ReviewsHuman
A) readers. | Genetics in mRNA and its functional roles in S.pombe should be
YTHDF13 proteins contain a carboxyterminal YTH RNA-binding domain and an further investigated.
aminoterminal P/Q/N-rich region. The YTH domain protein is conserved in the fission
yeast Schizosaccharomyces pombe and the budding yeast Saccharomyces cerevisiae. hnRNPs could be potential nuclear m6A readers. Besides
b | The m6A modification is enriched in mRNAs with shorter half-lives in general,
the YTH domain family of proteins and other cytoplas-
which supports the proposed main role of m6A in regulating mRNA stability. c | The
m6A-specific RNA-binding proteins are engaged in post-transcriptional regulation mic mRNA-binding proteins, pulldown experiments
of gene expression. YTHDF2 regulates the methylation (me)-dependent RNA have also identified proteins of the heterogeneous
degradation. Other reader proteins may exist and affect RNA splicing, storage, nuclear ribonucleoprotein (hnRNP) type as potential
trafficking and translation. Data in part b courtesy of X.Wang, laboratory of C.H. m6A-selective binding proteins17. Known to form ribo-
nucleoprotein granules that could affect mRNA localiza-
tion and transport, hnRNPs could also block binding of
contain adenine, hm6A or f 6A, or that have m6A in non- splicing factors and affect alternative splicing. Additional
Ribosome profiling
consensus sequences have decreased binding affinity. experiments are required to investigate connections
Qualitative and quantitative
sequencing of the RNA attached The enrichment of m6A in RNA immunoprecipitated between hnRNPs andm6A.
to ribosomes as a signature of with YTHDF13 further supports the role of YTHDF
genes that are expressed. proteins as m6A-specific RNA-binding proteins. Anti-readers of m6A and m6A-derived modifications.
RNA immunoprecipitation and PARCLIP experi- The presence of the methyl group can also disfavour
Processing bodies
(P-bodies). Distinct foci in the
ments revealed mostly mRNA as targets of YTHDF2, binding of an RNA-binding protein to the modified
cytoplasm that are enriched in addition to some lncRNA targets. The binding sites RNA. This mechanism of anti-reading has yet to be
with RNA degradation factors. localize around stop codons and at 3UTRs with a observed for m6A. The m6A modification is widely

300 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

distributed in the 3UTRs of mRNA transcripts a pathway level (for example, the p53-mediated pathway,
region bound by numerous RNA-binding proteins that Notch signalling, nutrient sensing through mammalian
regulate mRNA metabolism and translation. It is possi- target of rapamycin complex1 (mTORC1) and apopto-
ble that certain anti-reading mechanisms exist to regu- sis). Third are roles at the machinery level (for example,
late the fate of methylated mRNA. m6A is also known to the spliceosome and the nuclear export machinery). Last
protect RNA from recognition by cellular innate immu- are functions at the elementary molecular level on which
nity proteins. Toll-like receptor 3 (TLR3) and TLR7 rec- all other levels depend (for example, thermodynamics
ognize unmodified double-stranded and single-stranded and proteinm6A interactions). The reader proteins and
RNA as invasive RNA species and selectively target them their associated recognition mechanisms will be crucial
for degradation88,89. The incorporation of m6A and other in revealing and understanding theseroles.
RNA modifications in transfected exogenous RNA can
reduce the recognition by innate immune systems to Post-transcriptional regulation through methylation-
prevent unnecessary degradation, which increases dependent localization of the target transcript. To our
their expression. An anti-reading mechanism possibly knowledge, the indepth characterization of YTHDF2
operates in this process. as the first m6A reader delineates the first established
molecular pathway mediated by m6A: the binding of
Indirect reading. Certain RNA modifications, such as YTHDF2 to thousands of mRNA transcripts (and also
pseudouridine (), are known to cause secondary and to certain ncRNA transcripts) results in the localization
tertiary structural changes90. The m6A modification of bound mRNA from the translatable pool to decay
reduces the base-pairing energy of A:U only margin- sites, thereby affecting the translation status and half-
ally 35, but this difference may shift the equilibrium of life of mRNA76. This discovery has two fundamental
certain secondary and tertiary structures of RNA. The merits. First, it indicates that a main function of m6A
altered structures could have an effect on the binding methylation as a reversible mark is to affect mRNA
of specific proteins, leading to indirect reading and stability, which fits nicely with the negative correla-
regulation. In a recent study of HuR (also known as tion between m 6A levels and transcript abundance
ELAVL1) a well-known RNA-binding protein that observed upon silencing of methyltransferases. In fact,
affects the stability of many mRNA transcripts in mam- such methylation generally associates with mRNAs that
malian cells9196 the m6A modification affected the have shorter half-lives (FIG.4b), which further supports
ability of HuR to bind to different RNA probes invitro42. this notion. Second, this example illustrates how selec-
In this particular case, the RNA structure altered by tive reading of the m6A mark by a binding protein can
methylation might indirectly contribute to the acces- affect localization of the target RNA, thus providing
sibility of the cognate HuR-binding site. However, a model that applies to other potential readers which
the consensus sequence recognized by HuR invivo is may broadly affect RNA transport, storage, stability,
different from the m6A-containing sequence91,92. The translation and splicing.
extent and details of the cellular connection between Although transcriptional regulation has major
HuR and m6A still need to be further investigated. roles in regulating gene expression, it is protein levels
So far, no cellular example is known for this indirect that mainly determine biological phenotypes. Protein
reading mechanism. production is also subjected to various types of post-
transcriptional regulation such as through mRNA
Biological consequences of m6A secondary structure, microRNAs or mRNA translational
The recent breakthroughs in the discovery and charac- control which probably contributes as much as, if not
terization of m6A writers, erasers and readers, together more than, transcriptional regulation to determine cel-
with the parallel development of high-throughput assays lular protein abundance97103. m6A methylation provides
that profile this methylation on a transcriptome-wide a new dimension of post-transcriptional gene regulation.
scale, set the stage and provide tools for functional inves- The m6A mark on mRNA could affect the abundance,
tigations that aim to identify the mechanisms by which localization and use of mRNA, and potentially splicing;
m6A is translated into biological outcomes. Past studies all of these represent central processes that are connected
that used broad-spectrum methylation inhibitors have to protein expression (FIG.4c). We believe that m6A has
yielded inconclusive results. Now that writer, eraser and a substantial contribution to the post-transcriptional
certain reader proteins have been clearly defined, per- balance that regulates proteinlevels.
turbation of these machineries can lead to more specific
phenotypic outcomes and experimental observations Known examples of RNA methylation in regulating cel-
that will help to elucidate the biological roles of m6A and lular processes. Various circadian RNAs and clock output
the underlying mechanisms. An instrumental aspect of transcripts contain the m6A modification104. Inhibition
this endeavour will be to categorize the phenotypic lev- of m6A formation leads to prolonged nuclear reten-
els influenced by m6A. First are effects at the levels of tion of circadian RNAs and thus delays the nuclear exit
whole organisms or tissues; studies at these levels could of mature period circadian clock 2 (Per2) and aryl
reveal tissue specificity of m6A, as well as its relevance hydrocarbon receptor nuclear translocator-like (Arntl;
to certain diseases and biological processes (for exam- also known as Bmal1) mRNAs104. This observation is
ple, development, infertility, carcinogenesis, stemness, consistent with our discovery that deletion of ALKBH5
meiosis and circadian rhythm). Second are effects at the (which increases m6A levels) in mammalian cells leads

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 301

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

to reduced nuclear retention time16, although it is not Figure 5 | RNA methylation could affect various aspects
clear whether m6A has a direct cis role in modulating the of RNA metabolism and mRNA translation, and
export specifically of m6A-containing RNA molecules or regulate protein expression post-transcrptionally.
whether it is an indirect consequence of perturbation to Whereas N6-methyladenosine (m6A) methyltransferases
the RNA export machinery. and demethylases shape the methylation (me)
landscape, the reader proteins bind to the methylated
The m6A RNA modification is involved in priming
RNA and mediate specific functions. Various cellular
yeast cells to bipotential states and meiosis during nitro- processes could be affected by m6A RNA methylation.
gen starvation. Through carefully monitoring methyla- In the cell nucleus, m6A may affect RNA export, nuclear
tion profiles at different stages, a recent study suggests retention and splicing, possibly through interactions of
that methylation is important for the kinetic control reader proteins with RNA export, retention and splicing
of RNAs during the meiotic prophase32. Although no machineries. After RNAs are exported to the cytoplasm,
marked change in half-lives has been observed for the YTHDF2 can bind to the m6A-containing RNAs and
m6A-containing RNAs, the accessibility of these RNAs direct them to processing bodies (Pbodies) for
to translation may be modulated through interac- accelerated mRNA decay. Pbodies can dynamically
tions with potential reader proteins. Analogous to the form stress granules, in which RNAs could be stored
proposed function of m6A in accelerating both RNA and released back to the translating pool. Besides
YTHDF2, other m6A reader proteins may bind to
export and degradation in mammalian cells, m6A may
m6A-containing RNAs to control their transport
ensure faster turnover of the RNA transcripts that and storage, thereby affecting translation. FTO,
are important during the meiotic prophase but that are -ketoglutarate-dependent dioxygenase FTO; WTAP,
harmful and need to be degraded after the exit from Wilms tumour 1-associating protein.
prophase. This example suggests that m6A could glob-
ally ensure fast kinetic responses by redirecting RNA
to different organelles and by quickly decreasing the
expression of related proteins. A similar mechanism when the response at the DNA level (that is, transcrip-
could also affect eukaryotic mitosis. tion) could be too slow; and when the response at
Similarly, another study in mESCs revealed that the the protein level may require specific interactions or
m6A methylation accelerates transcript decay, which modifications to tens to thousands of proteins, which
is consistent with the main role that we propose for is difficult to achieve. Reversible methylation or other
m6A, and affects stem cell maintenance and differen- forms of modifications on mRNA provides the best
tiation42. Interestingly, when compared with pluripo- option. A specific sequence that can undergo revers-
tency-related genes, developmental regulators were ible modification, and thus be subjected to regulation,
significantly enriched among target genes of METTL3 can be readily included in a group of mRNA transcripts
and METTL14 in mESCs. In particular, m6A destabi- (for example, at their 3UTRs) and lncRNAs in order
lizes developmental regulator transcripts, which may to affect RNA stability, localization and translatability,
suggest that methylation is important for maintenance as shown in the example ofYTHDF2.
and differentiation of mESCs. Temporal and spatial
regulation of mRNA is known to have crucial roles in Perspectives
embryonic development. Post-fertilization, maternal In summary, reversible RNA methylation shares many
mRNA needs to be degraded in a programmed manner. of the same characteristics as epigenetic DNA and his-
The methylation on mRNA could affect this process tone modifications. Expression levels and potentially
through altering the localization and half-lives of tar- post-translational modifications of writers, erasers
get mRNA transcripts. Such methylation could mark and readers can constantly sculpt the RNA methyl-
specific sets of RNA species and therefore differentiate ome (FIG.5), which might in turn affect the eventual
between maternal and zygotic mRNA in a kinetic man- protein expression. Therefore, the reversible chemical
ner. Heritable information could perhaps be passed tagging that dynamically controls the outcome of gene
down to generations of cells through orchestrated RNA expression occurs in all three main components of the
methylation and demethylation activities. central dogma. Whereas epigenetic DNA and histone
modifications affect mostly transcriptional events,
Advantage and specificity of the m6A-based regula- reversible RNA methylation mainly has an impact on
tion. The first m6A reader protein to be characterized is regulation of post-transcriptional gene expression and
known to affect more than 3,000 different mRNA tran- could directly affect protein production. Indeed, recent
scripts76. We propose that the reversible RNA methyla- research indicates that cellular protein levels are not
tion pathway, in general, has evolved to affect processes necessarily correlated with the mRNA levels105,106, which
that involve changes in the expression of large groups of emphasizes the importance of post-transcriptional
genes. This property is intimately related to the poten- regulation of gene expression. Owing to the shared
tial advantages of reversible methylation at the RNA use of reversible chemical tagging for dynamic gene
level. Besides providing increased complexity to the expression control, reversible RNA methylation has
regulatory network, this mechanism may allow rapid been compared with epigenetic DNA and histone
responses to signalling and stimuli when the expres- modifications10,107. To also be a true epigenetic mark,
sion of a group of proteins (which can range from tens m6A would need to be heritable through cell division;
to thousands) needs to be adjusted in a rapid manner; although this has not yet been demonstrated, such

302 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

METTL14 METTL3
WTAP
Histone modication

Other m6A-binding proteins

DNA modication

RNA polymerase II

m7Gppp An
Nuclear retention
Nuclear
speckle ALKBH5

FTO foci Translation


FTO

Me
Me m A
6

m7Gppp An
m7Gppp An
m7Gppp An
m7Gppp An
Storage or
transport
Me m A
6
Me
m7Gppp An

Export

YTHDF2

Translation
Me Me
m7Gppp An m7Gppp An m7Gppp An

Ribosome

Me Me Me Me
m 7Gppp An m7Gppp An m7Gppp An

Translating pool Translatable pool

Me An
Me Me
m 7Gpp Me
p
An
ppp Me
mG
7
m 7G
ppp Me
Me
Me
m 7Gppp Me
An
An
Stress granule mRNA decay
P-body

Nature Reviews | Genetics


NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 303

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

a possibility is conceivable through direct passage Other intriguing chemical modifications exist on
of writer, reader and eraser proteins or methylated mRNA and other types of nuclear RNA, such as m5C,
RNA between generations of cells, which is a research and 2OMe. Some of these modifications are only
direction that needs to be further explored. a few fold less abundant than m6A on mRNA. They
Many challenges lie ahead. It will be important to could also be dynamic and may have important roles
clearly define the spatiotemporal properties of m6A in gene expression regulation, as recently suggested for
in terms of tissue specificity and in response to exter- 108110. Although transcriptome-wide m5C distribution
nal and internal cues. The top priority is to elucidate has been mapped111113, the other two modifications
the involvement of m6A in RNA degradation, trans- have yet to be studied using modern sequencing
port, storage, translation and splicing. The first steps approaches. Both and 2OMe may have connections
to achieve this goal will involve identifying and elu- to human diseases, which suggests functional roles114116.
cidating the functions of m6A reader proteins. Some Modifications on tRNA and rRNA can also be dynamic
additional questions are: what is the interplay between and could affect the outcome of protein expression. Of
methyltransferases and demethylases that orchestrate the nine human homologues of RNA demethylases,
the methylation status of individual sites? How is ALKBH2 and ALKBH3 are DNA repair enzymes that
methylation and demethylation selectivity achieved? use the same oxidative demethylation mechanism to
Is the methylation coupled with transcription, and do remove DNA methyl adducts117,118, and ALKBH8 is a
the two processes have mutual interactions? From yeast tRNA hydroxylase119,120 that seems to affect tRNA codon
to humans, how do the functions of m6A relate to cell usage, whereas ALKBH1, ALKBH4, ALKBH6 and
phenotypes and cell behaviour? Could some of the ALKBH7 still do not have clearly defined functions.
processes be targeted to regulate biological functions Some of these homologues might work on nucleic acids
or to treat human diseases? Further research will answer and act as demethylases for other forms of nucleic acid
some of these questions and reveal fundamental aspects methylations. We are still at the very beginning of this
of m6A biology. new realm of fundamental research.

1. Suzuki,M.M. & Bird,A. DNA methylation landscapes: 18. Meyer,K.D. etal. Comprehensive analysis of mRNA 32. Schwartz,S. etal. High-resolution mapping reveals a
provocative insights from epigenomics. Nature Rev. methylation reveals enrichment in 3 UTRs and near conserved, widespread, dynamic mRNA methylation
Genet. 9, 465476 (2008). stop codons. Cell 149, 16351646 (2012). program in yeast meiosis. Cell 155, 14091421 (2013).
2. Kohli,R.M. & Zhang,Y. TET enzymes, TDG and the References 17 and 18 revealed, for the first time, This study reveals the dynamics of transcriptome-
dynamics of DNA demethylation. Nature 502, the transcriptome-wide distributions of m6A in wide m6A changes during yeast meiosis.
472479 (2013). mammalian genomes. 33. Liu,N. etal. Probing N6methyladenosine RNA
3. Jones,P.A. Functions of DNA methylation: islands, 19. Wei,C.M., Gershowitz,A. & Moss,B. Methylated modification status at single nucleotide resolution
start sites, gene bodies and beyond. Nature Rev. nucleotides block 5 terminus of HeLa-cell messenger- in mRNA and long noncoding RNA. RNA 19,
Genet. 13, 484492 (2012). RNA. Cell 4, 379386 (1975). 18481856 (2013).
4. Branco,M.R., Ficz,G. & Reik,W. Uncovering the role 20. Krug,R.M., Morgan,M.A. & Shatkin,A.J. Influenza 34. Carroll,S.M., Narayan,P. & Rottman,F.M.
of 5hydroxymethylcytosine in the epigenome. Nature viral mRNA contains internal N6methyladenosine and N6methyladenosine residues in an intron-specific
Rev. Genet. 13, 713 (2012). 5terminal 7methylguanosine in cap structures. region of prolactin pre-mRNA. Mol. Cell. Biol. 10,
5. Bhutani,N., Burns,D.M. & Blau,H.M. DNA J.Virol. 20, 4553 (1976). 44564465 (1990).
demethylation dynamics. Cell 146, 866872 (2011). 21. Rottman,F.M., Desrosiers,R.C. & Friderici,K. 35. Kierzek,E. & Kierzek,R. The thermodynamic
6. Strahl,B.D. & Allis,C.D. The language of covalent Nucleotide methylation patterns in eukaryotic mRNA. stability of RNA duplexes and hairpins containing
histone modifications. Nature 403, 4145 (2000). Prog. Nucleic Acid. Res. Mol. Biol. 19, 2138 (1976). N6alkyladenosines and 2methylthio-
7. Shi,Y. Histone lysine demethylases: emerging roles in 22. Beemon,K. & Keith,J. Localization of N6alkyladenosines. Nucleic Acids Res. 31,
development, physiology and disease. Nature Rev. N6methyladenosine in the Rous sarcoma virus 44724480 (2003).
Genet. 8, 829833 (2007). genome. J.Mol. Biol. 113, 165179 (1977). 36. Harcourt,E.M., Ehrenschwender,T., Batista,P.J.,
8. Klose,R.J., Kallin,E.M. & Zhang,Y. JmjC-domain- 23. Schibler,U., Kelley,D.E. & Perry,R.P. Comparison of Chang,H.Y. & Kool,E.T. Identification of a selective
containing proteins and histone demethylation. methylated sequences in messenger RNA and polymerase enables detection of N6-methyladenosine
Nature Rev. Genet. 7, 715727 (2006). heterogeneous nuclear RNA from mouse L cells. in RNA. J.Am. Chem. Soc. 135, 1907919082
9. Bird,A. Molecular biology. Methylation talk between J.Mol. Biol. 115, 695714 (1977). (2013).
histones and DNA. Science. 294, 21132115 (2001). 24. Wei,C.M. & Moss,B. Nucleotide sequences at the 37. Vilfan,I.D. etal. Analysis of RNA base modification
10. He,C. Grand challenge commentary: RNA epigenetics? N6methyladenosine sites of HeLa cell messenger and structural rearrangement by single-molecule real-
Nature Chem. Biol. 6, 863865 (2010). ribonucleic acid. Biochemistry 16, 16721676 (1977). time detection of reverse transcription.
11. Grosjean,H. & Benne,R. Modification and Editing 25. Narayan,P. & Rottman,F.M. An invitro system for J.Nanobiotechnol. 11, 8 (2013).
of RNA (American Society for Microbiology Press, accurate methylation of internal adenosine residues in 38. Bokar,J.A., Shambaugh,M.E., Polayes,D.,
1998). messenger RNA. Science 242, 11591162 (1988). Matera,A.G. & Rottman,F.M. Purification and cDNA
12. Grosjean,H.Fine-Tuning of RNA Functions by 26. Csepany,T., Lin,A., Baldick,C.J.Jr & Beemon,K. cloning of the AdoMet-binding subunit of the human
Modication and Editing (Springer-Verlag, 2005). Sequence specificity of mRNA N6adenosine mRNA (N6adenosine)-methyltransferase. RNA 3,
13. Machnicka,M.A. etal. MODOMICS: a database methyltransferase. J.Biol. Chem. 265, 2011720122 12331247 (1997).
of RNA modification pathways 2013 update. (1990). This pivotal study identifies METTL3 as a
Nucleic Acids Res. 41, D262D267 (2013). 27. Narayan,P., Ludwiczak,R.L., Goodwin,E.C. & key SAM-binding subunit of the RNA
14. Motorin,Y. & Helm,M. RNA nucleotide methylation. Rottman,F.M. Context effects on N6adenosine methyltransferase complex.
Wiley Interdiscip. Rev. RNA 2, 611631 (2011). methylation sites in prolactin mRNA. Nucleic Acids 39. Bokar,J.A. in Fine-Tuning of RNA Functions by
15. Jia,G. etal. N6methyladenosine in nuclear RNA is a Res. 22, 419426 (1994). Modification and Editing 141177 (Springer-Verlag,
major substrate of the obesity-associated FTO. 28. Rottman,F., Shatkin,A.J. & Perry,R.P. Sequences 2005).
Nature Chem. Biol. 7, 885887 (2011). containing methylated nucleotides at 5 termini of 40. Liu,J. etal. A METTL3METTL14 complex mediates
This work describes a major breakthrough of messenger-RNAs possible implications for mammalian nuclear RNA N6-adenosine methylation.
discovering the first m6A RNA demethylase FTO, processing. Cell 3, 197199 (1974). Nature Chem. Biol. 10, 9395 (2014).
which highlights the possible biological function 29. Bodi,Z., Button,J.D., Grierson,D. & Fray,R.G. This paper uncovers the core components of
of m6A. Yeast targets for mRNA methylation. Nucleic Acids the m6A RNA methyltransferase complex and
16. Zheng,G. etal. ALKBH5 is a mammalian RNA Res. 38, 53275335 (2010). reveals an overall negative correlation between
demethylase that impacts RNA metabolism and 30. Keith,G. Mobilities of modified ribonucleotides on the levels of m6A mRNA methylation and gene
mouse fertility. Mol. Cell 49, 1829 (2013). two-dimensional cellulose thin-layer chromatography. expression.
This study discovered the second mammalian m6A Biochimie 77, 142144 (1995). 41. Bujnicki,J.M., Feder,M., Radlinska,M. &
demethylase ALKBH5 that affects mouse 31. Clancy,M.J., Shambaugh,M.E., Timpte,C.S. & Blumenthal,R.M. Structure prediction and
spermatogenesis. Bokar,J.A. Induction of sporulation in Saccharomyces phylogenetic analysis of a functionally diverse family of
17. Dominissini,D. etal. Topology of the human and cerevisiae leads to the formation of N6methyladenosine proteins homologous to the MTA70 subunit of the
mouse m6A RNA methylomes revealed by m6A-seq. in mRNA: a potential mechanism for the activity of the human mRNA:m6A methyltransferase. J.Mol. Evol.
Nature 485, 201206 (2012). IME4 gene. Nucleic Acids Res. 30, 45094518 (2002). 55, 431444 (2002).

304 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

42. Wang,Y. etal. N6-methyladenosine modification 67. Gulati,P. etal. Role for the obesity-related FTO gene 93. Srikantan,S. & Gorospe,M. UneCLIPsing HuR nuclear
destabilizes developmental regulators in embryonic in the cellular sensing of amino acids. Proc. Natl Acad. function. Mol. Cell 43, 319321 (2011).
stem cells. Nature Cell Biol. 16, 191198 (2014). Sci. USA 110, 25572562 (2013). 94. Dormoy-Raclet,V. etal. HuR and miR1192 regulate
This study discovered that the m6A modification 68. Han,Z. etal. Crystal structure of the FTO protein myogenesis by modulating the translation of HMGB1
on mRNA affects embryonic cell differentiation. reveals basis for its substrate specificity. Nature 464, mRNA. Nature Commun. 4, 2388 (2013).
43. Alexandrov,A., Martzen,M.R. & Phizicky,E.M. 12051209 (2010). 95. Barnhart,M.D., Moon,S.L., Emch,A.W.,
Two proteins that form a complex are required for 69. Zheng,G. etal. Sprouts of RNA epigenetics: Wilusz,C.J. & Wilusz,J. Changes in cellular mRNA
7methylguanosine modification of yeast tRNA. the discovery of mammalian RNA demethylases. stability, splicing, and polyadenylation through HuR
RNA 8, 12531266 (2002). RNA Biol. 10, 915918 (2013). protein sequestration by a cytoplasmic RNA virus.
44. Chujo,T. & Suzuki,T. Trmt61B is a methyltransferase 70. Baltz,A.G. etal. The mRNA-bound proteome and its Cell Rep. 5, 909917 (2013).
responsible for 1methyladenosine at position 58 of global occupancy profile on protein-coding transcripts. 96. Abdelmohsen,K. & Gorospe,M. Posttranscriptional
human mitochondrial tRNAs. RNA 18, 22692276 Mol. Cell 46, 674690 (2012). regulation of cancer traits by HuR. Wiley Interdiscip.
(2012). 71. Fu,Y. etal. FTO-mediated formation of Rev. RNA 1, 214229 (2010).
45. Ozanick,S., Krecic,A., Andersland,J. & N6hydroxymethyladenosine and N6formyladenosine in 97. Ambros,V. The functions of animal microRNAs.
Anderson,J.T. The bipartite structure of the mammalian RNA. Nature Commun. 4, 1798 (2013). Nature 431, 350355 (2004).
tRNA m1A58 methyltransferase from S.cerevisiae 72. Schwanhausser,B. etal. Global quantification of 98. Chen,K. & Rajewsky,N. The evolution of gene
is conserved in humans. RNA 11, 12811290 mammalian gene expression control. Nature 473, regulation by transcription factors and microRNAs.
(2005). 337342 (2011). Nature Rev. Genet. 8, 93103 (2007).
46. Leulliot,N. etal. Structure of the yeast tRNA m7G 73. Rabani,M. etal. Metabolic labeling of RNA uncovers 99. Filipowicz,W., Bhattacharyya,S.N. & Sonenberg,N.
methylation complex. Structure 16, 5261 (2008). principles of RNA production and degradation Mechanisms of post-transcriptional regulation by
47. Zhong,S. etal. MTA is an Arabidopsis messenger dynamics in mammalian cells. Nature Biotech. 29, microRNAs: are the answers in sight? Nature Rev.
RNA adenosine methylase and interacts with a 436442 (2011). Genet. 9, 102114 (2008).
homolog of a sex-specific splicing factor. Plant Cell 20, 74. Robbens,S. etal. The FTO gene, implicated in human 100. Parker,R. & Sheth,U. P bodies and the control of
12781288 (2008). obesity, is found only in vertebrates and marine algae. mRNA translation and degradation. Mol. Cell 25,
48. Agarwala,S.D., Blitzblau,H.G., Hochwagen,A. & J.Mol. Evol. 66, 8084 (2008). 635646 (2007).
Fink,G.R. RNA methylation by the MIS complex 75. Iyer,L.M., Tahiliani,M., Rao,A. & Aravind,L. 101. Keene,J.D. RNA regulons: coordination of
regulates a cell fate decision in yeast. PLoS Genet. 8, Prediction of novel families of enzymes involved in post-transcriptional events. Nature Rev. Genet. 8,
e1002732 (2012). oxidative and other complex modifications of bases in 533543 (2007).
49. Little,N.A., Hastie,N.D. & Davies,R.C. Identification nucleic acids. Cell Cycle 8, 16981710 (2009). 102. Gallego,M. & Virshup,D.M. Post-translational
of WTAP, a novel Wilms tumour 1associating protein. 76. Wang,X. etal. N6methyladenosine-dependent modifications regulate the ticking of the circadian
Hum. Mol. Genet. 9, 22312239 (2000). regulation of messenger RNA stability. Nature 505, clock. Nature Rev. Mol. Cell Biol. 8, 139148 (2007).
50. Ping,X.L. etal. Mammalian WTAP is a regulatory 117120 (2014). 103. Eulalio,A., Behm-Ansmant,I. & Izaurralde,E.
subunit of the RNA N6methyladenosine This work presents the first m6A reader protein to P bodies: at the crossroads of post-transcriptional
methyltransferase. Cell Res. 24, 177189 (2014). be characterized, YTHDF2, and a main function of pathways. Nature Rev. Mol. Cell Biol. 8, 922 (2007).
51. Horiuchi,K. etal. Identification of Wilms m6A: YTHDF2 mediates the m6A-dependent RNA 104. Fustin,J.M. etal. RNA-methylation-dependent RNA
Tumor1associating protein complex and its role in decay by targeting RNA substrates to Pbodies. processing controls the speed of the circadian clock.
alternative splicing and the cell cycle. J.Biol. Chem. 77. Schoenberg,D.R. & Maquat,L.E. Regulation of Cell 155, 793806 (2013).
288, 3329233302 (2013). cytoplasmic mRNA decay. Nature Rev. Genet. 13, This study shows that the m6A modification affects
52. Bodi,Z. etal. Adenosine methylation in Arabidopsis 246259 (2012). the export of several mRNAs that are related to
mRNA is associated with the 3 end and reduced 78. Isken,O. & Maquat,L.E. The multiple lives of NMD the circadian cycle.
levels cause developmental defects. Front. Plant Sci. factors: balancing roles in gene and genome 105. Khan,Z. etal. Primate transcript and protein
3, 48 (2012). regulation. Nature Rev. Genet. 9, 699712 (2008). expression levels evolve under compensatory selection
53. Hongay,C.F. & Orr-Weaver,T.L. Drosophila Inducer of 79. Sheth,U. & Parker,R. Decapping and decay of pressures. Science 342, 11001104 (2013).
MEiosis 4 (IME4) is required for Notch signaling messenger RNA occur in cytoplasmic processing 106. Wu,L. etal. Variation and genetic control of protein
during oogenesis. Proc. Natl Acad. Sci. USA 108, bodies. Science 300, 805808 (2003). abundance in humans. Nature 499, 7982 (2013).
1485514860 (2011). 80. Han,D. etal. IRE1 kinase activation modes control 107. Saletore,Y. etal. The birth of the epitranscriptome:
54. Peters,T., Ausmeier,K. & Ruther,U. Cloning of Fatso alternate endoribonuclease outputs to determine deciphering the function of RNA modifications.
(Fto), a novel gene deleted by the Fused toes (Ft) divergent cell fates. Cell 138, 562575 (2009). Genome Biol. 13, 175 (2012).
mouse mutation. Mamm. Genome 10, 983986 81. Marzluff,W.F., Wagner,E.J. & Duronio,R.J. 108. Karijolich,J. & Yu,Y.T. Converting nonsense codons
(1999). Metabolism and regulation of canonical histone into sense codons by targeted pseudouridylation.
55. Dina,C. etal. Variation in FTO contributes to mRNAs: life without a poly(A) tail. Nature Rev. Genet. Nature 474, 395398 (2011).
childhood obesity and severe adult obesity. Nature 9, 843854 (2008). 109. Fernandez,I.S. etal. Unusual base pairing during the
Genet. 39, 724726 (2007). 82. Dasgupta,T. & Ladd,A.N. The importance of CELF decoding of a stop codon by the ribosome. Nature
56. Frayling,T.M. etal. A common variant in the FTO control: molecular and biological roles of the CUGBP, 500, 107110 (2013).
gene is associated with body mass index and Elav-like family of RNA-binding proteins. Wiley 110. Ge,J. & Yu,Y.T. RNA pseudouridylation: new insights
predisposes to childhood and adult obesity. Science Interdiscip. Rev. RNA 3, 104121 (2012). into an old modification. Trends Biochem. Sci. 38,
316, 889894 (2007). 83. Yang,F. & Schoenberg,D.R. Endonuclease-mediated 210218 (2013).
57. Scuteri,A. etal. Genome-wide association scan mRNA decay involves the selective targeting of PMR1 111. Edelheit,S., Schwartz,S., Mumbach,M.R.,
shows genetic variants in the FTO gene are to polyribosome-bound substrate mRNA. Mol. Cell Wurtzel,O. & Sorek,R. Transcriptome-wide mapping
associated with obesity-related traits. PLoS Genet. 3, 14, 435445 (2004). of 5methylcytidine RNA modifications in bacteria,
e115 (2007). 84. Ghosh,S. & Jacobson,A. RNA decay modulates gene archaea, and yeast reveals m5C within archaeal
58. Gerken,T. etal. The obesity-associated FTO gene expression and controls its fidelity. Wiley Interdiscip. mRNAs. PLoS Genet. 9, e1003602 (2013).
encodes a 2oxoglutarate-dependent nucleic acid Rev. RNA 1, 351361 (2010). 112. Hussain,S., Aleksic,J., Blanco,S., Dietmann,S. &
demethylase. Science 318, 14691472 (2007). 85. He,L. & Hannon,G.J. MicroRNAs: small RNAs with a Frye,M. Characterizing 5methylcytosine in the
59. Fischer,J. etal. Inactivation of the Fto gene protects big role in gene regulation. Nature Rev. Genet. 5, mammalian epitranscriptome. Genome Biol. 14, 215
from obesity. Nature 458, 894898 (2009). 522531 (2004). (2013).
60. Church,C. etal. Overexpression of Fto leads to 86. Ameres,S.L. & Zamore,P.D. Diversifying microRNA 113. Squires,J.E. etal. Widespread occurrence of
increased food intake and results in obesity. Nature sequence and function. Nature Rev. Mol. Cell Biol. 14, 5methylcytosine in human coding and non-coding
Genet. 42, 10861092 (2010). 475488 (2013). RNA. Nucleic Acids Res. 40, 50235033 (2012).
61. Boissel,S. etal. Lossoffunction mutation in the 87. Harigaya,Y. etal. Selective elimination of messenger 114. Bykhovskaya,Y., Casas,K., Mengesha,E., Inbal,A. &
dioxygenase-encoding FTO gene causes severe growth RNA prevents an incidence of untimely meiosis. Fischel-Ghodsian,N. Missense mutation in
retardation and multiple malformations. Am. J.Hum. Nature 442, 4550 (2006). pseudouridine synthase 1 (PUS1) causes
Genet. 85, 106111 (2009). 88. Kariko,K., Buckstein,M., Ni,H. & Weissman,D. mitochondrial myopathy and sideroblastic anemia
62. He,Y.F. etal. Tet-mediated formation of Suppression of RNA recognition by Toll-like receptors: (MLASA). Am. J.Hum. Genet. 74, 13031308
5carboxylcytosine and its excision by TDG in the impact of nucleoside modification and the (2004).
mammalian DNA. Science 333, 13031307 evolutionary origin of RNA. Immunity 23, 165175 115. Patton,J.R., Bykhovskaya,Y., Mengesha,E.,
(2011). (2005). Bertolotto,C. & Fischel-Ghodsian,N. Mitochondrial
63. Ito,S. etal. Tet proteins can convert 5methylcytosine 89. Kawai,T. & Akira,S. Toll-like receptor and RIGIlike myopathy and sideroblastic anemia (MLASA):
to 5formylcytosine and 5carboxylcytosine. Science receptor signaling. Ann. NY Acad. Sci. 1143, 120 missense mutation in the pseudouridine synthase 1
333, 13001303 (2011). (2008). (PUS1) gene is associated with the loss of
64. Tahiliani,M. etal. Conversion of 5methylcytosine to 90. Newby,M.I. & Greenbaum,N.L. Sculpting of the tRNA pseudouridylation. J.Biol. Chem. 280,
5hydroxymethylcytosine in mammalian DNA by MLL spliceosomal branch site recognition motif by 1982319828 (2005).
partner TET1. Science 324, 930935 (2009). a conserved pseudouridine. Nature Struct. Biol. 9, 116. Sahoo,T. etal. PraderWilli phenotype caused by
65. Jia,G. etal. Oxidative demethylation of 958965 (2002). paternal deficiency for the HBII85C/D box small
3methylthymine and 3methyluracil in single-stranded 91. Lebedeva,S. etal. Transcriptome-wide analysis of nucleolar RNA cluster. Nature Genet. 40, 719721
DNA and RNA by mouse and human FTO. FEBS Lett. regulatory interactions of the RNA-binding protein (2008).
582, 33133319 (2008). HuR. Mol. Cell 43, 340352 (2011). 117. Sedgwick,B. Repairing DNA-methylation damage.
66. Hess,M.E. etal. The fat mass and obesity associated 92. Mukherjee,N. etal. Integrative regulatory mapping Nature Rev. Mol. Cell Biol. 5, 148157 (2004).
gene (Fto) regulates activity of the dopaminergic indicates that the RNA-binding protein HuR couples 118. Mishina,Y., Duguid,E.M. & He,C. Direct reversal of
midbrain circuitry. Nature Neurosci. 16, 10421048 pre-mRNA processing and mRNA stability. Mol. Cell DNA alkylation damage. Chem. Rev. 106, 215232
(2013). 43, 327339 (2011). (2006).

NATURE REVIEWS | GENETICS VOLUME 15 | MAY 2014 | 305

2014 Macmillan Publishers Limited. All rights reserved


REVIEWS

119. Fu,Y. etal. The AlkB domain of mammalian ABH8 122. Chen, W. et al. Crystal structure of the RNA Competing interests statement
catalyzes hydroxylation of demethylase ALKBH5 from zebrafish. FEBS Lett. 588, The authors declare no competing interests.
5methoxycarbonylmethyluridine at the wobble 892898 (2014).
position of tRNA. Angew. Chem. Int. Ed Engl. 49,
88858888 (2010). Acknowledgements FURTHER INFORMATION
120. van den Born,E. etal. ALKBH8mediated formation The authors apologize to colleagues whose work was not Modomics a database of RNA modification pathways:
of a novel diastereomeric pair of wobble nucleosides cited owing to space limitation. They thank T. Pan, X. Wang, http://modomics.genesilico.pl/
in mammalian tRNA. Nature Commun. 2, 172 Y. Yue and J. Liu for discussion. C.H. is supported by the US Three-dimensional ribosomal modification maps database:
(2011). National Institutes of Health grants GM071440 and the http://people.biochem.umass.edu/fournierlab/3dmodmap/
121. Aik, W. et al. Structure of human RNA EUREKA grant GM088599. This work was also supported main.php
N6-methyladenine demethylase ALKBH5 provides partly by grants from the Israel Science Foundation, the Flight tRNAmod prediction of tRNA modifications: http://crdd.
insights into its mechanisms of nucleic acid recognition Attendant Medical Research Institute (FAMRI) and the Israeli osdd.net/raghava/trnamod/
and demethylation. Nucleic Acids Res. http://dx.doi. Centers of Research Excellence. S.F. Reichard contributed to ALL LINKS ARE ACTIVE IN THE ONLINE PDF
org/10.1093/nar/gku085 (2014). editing of this manuscript.

306 | MAY 2014 | VOLUME 15 www.nature.com/reviews/genetics

2014 Macmillan Publishers Limited. All rights reserved

Das könnte Ihnen auch gefallen