You are on page 1of 6

Analytical Biochemistry 578 (2019) 45–50

Contents lists available at ScienceDirect

Analytical Biochemistry
journal homepage: www.elsevier.com/locate/yabio

Role of identified RNA N6-methyladenosine methylation in liver T


a,1 c,1 c d c b,*
Kechen Xu , Yuchao Sun , Baixiang Sheng , Yiwen Zheng , Xiaoli Wu , Keyang Xu
a
Clinical Laboratory Center, Wuyi First People's Hospital, Wuyi, 321200, Zhejiang, China
b
The Fourth Clinical Medicine School of Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
c
The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
d
Hangzhou Medical College, Hangzhou, 310053, Zhejiang, China

A R T I C LE I N FO A B S T R A C T

Keywords: N6-Methyladenosine (m6A) is the most abundant and important internal modification site of RNA methylation in
N6-methyladenosine viruses and eukaryotic. m6A RNA methylation plays key roles in the regulation of post-transcriptional gene
Methylation expression, including messenger RNA (mRNA), microRNA (miRNA) and long noncoding RNA (lncRNA). And
Messenger RNA m6A methylation regulates the various aspects of RNA metabolism, including structure, maturation, stability,
Noncoding RNA
splicing, export, translation and decay. Liver is a vital metabolic and digestive organ in the pathophysiological
Liver
processes. Recent studies suggested that m6A RNA modification highly regulates hepatic function and devel-
opment of liver diseases. Here, we aim to summarize the biological and clinical significance of m6A modification
in hepatic growth and hepatic disease including viral hepatitis, non-alcoholic fatty liver disease (NAFLD), and
liver cancer.

1. Introduction Furthermore, some researchers have found that YTHDF2 can selectively
bind m6A-methylated mRNA and control RNA decay in a methylation-
Methylated RNA nucleotides widely exist in all kingdoms of life, dependent manner [10]. As a splicing modulator, m6A affects mRNA
which play crucial roles in pathophysiological processes. Methylated maturation by recruiting m6A regulators to pre-mRNA [11]. And m6A
nucleosides in messenger RNA (mRNA) were firstly discovered from methylation of mRNA can inhibit its tRNA accommodation and trans-
Novikoff hepatoma cells in 1974 [1] and then several methylation lation elongation rate [12]. Furthermore, m6A modification can reg-
modifications in eukaryotic messenger RNA (mRNA) were identified, ulate the structure and function of noncoding RNA, including tRNA,
including N7-methylguanosine at the cap, N6-methyl-2′-O -methyla- rRNA, small nuclear RNA, miRNA and long non-coding RNA [6]. The
denosine, 2′-O-methylation within the cap and the internal positions, expression of m6A demethylase fat mass and obesity-associated protein
and internal N6-methyladenosine (m6A) and 5-methylcytosine [2]. (FTO) can affect the steady state level of certain miRNAs, including up-
Among them, m6A as a reversible modification site can be methylated regulation of hsa-miR-6505-5p, hsa-miR-651-5p and hsa-miR-493-5p,
by “writers”, demethylated by “erasers”, and recognized by “readers” and down-regulation of hsa-miR-7-5p, hsa-miR-92a-1-5p and hsa-miR-
[3]. Over 12,000 m6A sites are identified with a typical consensus in the 6769a-3p [13]. In reverse, miRNA145 can also modulate m6A levels by
transcripts of more than 7000 human genes [4]. The sites of m6A targeting the 3′-untranslated mRNA region of the m6A binding protein
modification are identified to preferentially concentrate on two distinct YTHDF2 [14]. In addition, m6A modification of lncRNA can induce the
coordinates: around stop codons and within long internal exons [4]. proliferation, migration and apoptosis of cancer cells, including pan-
Furthermore, m6A methylation has been widely reported in regulation creatic cancer, ovarian cancer and hepatoma [15]. It was also reported
of mRNA [5] and non-coding RNA (ncRNA) [6]. that the m6A methyltransferase-like 16 (METTL16) can methylate di-
m6A methylation can regulate mRNA at various stages, including verse cellular RNAs in HEK293 cells, including 8 pre-mRNAs, 355
structure, maturation, stability, splicing, export, translation and decay mRNAs, 68 lncRNAs and others [16]. And RNA m6A methylation is
[7]. The dynamic m6A modification is recognized by YTH domain fa- highly involved in hepatic cell differentiation, metabolic processes and
mily 2 (YTHDF2) to affect the stability of mRNA [8]. Human YTHDF1 pathologic development [17]. m6A may be a potential therapeutic and
recognizes m6A-modified mRNAs and then enhances mRNA translation diagnostic target for hepatic diseases.
efficiency by interacting with initiation factors and ribosomes [9]. Here, we aim to summarize the functions and clinical implications

*
Corresponding author.
E-mail address: keyangxu@qq.com (K. Xu).
1
These authors contributed equally to this paper.

https://doi.org/10.1016/j.ab.2019.05.005
Received 26 December 2018; Received in revised form 1 May 2019; Accepted 6 May 2019
Available online 07 May 2019
0003-2697/ © 2019 Elsevier Inc. All rights reserved.
K. Xu, et al. Analytical Biochemistry 578 (2019) 45–50

of RNA m6A modification in liver diseases.

2. m6A writers, erasers, readers

The m6A “writers”, a multicomponent protein complex, are con-


sisted of methyltransferase-like 3 (METTL3) [18], methyltransferase-
like 14 (METTL14) [19], Wilms’ tumor 1 associating protein (WTAP)
[20], KIAA1429 21 and RNA binding motif protein 15/15B (RBM15/
15B) [22]. METTL3, a S-adenosyl methionine (SAM)-binding subunit, is
associated with mRNA methylation [18], and the knockdown of
METTL3 can lead to the near-complete loss of m6A [23]. Interaction
between the METTL3 enzymatic subunit of the MTC and retarded
RNAPII has suggested that m6A modification is at least in part co-
transcriptional [24]. METTL3 and METTL14 can form a stable hetero-
Fig. 1. The dynamic and reversible processes and consequences of m6A me-
dimer as the core of the methylase complex, and the knockdown of
thylation. RNA, the transcript of DNA, can be modified by m6A methylation.
METTL3 or METTL14 both reduces the m6A level due to existing me- The processes of m6A methylation are dynamic and reversible, methylated by
thyltransferase domains [25]. However, only METTL3 has catalytic “writers”-METTL3, METTL14, WTAP, RBM, and demethylated by “erasers”-FTO
activity, and METTL14 just stabilizes METTL3 and its interaction with and ALKBH5, as well as recognized by “readers”-YTHDF1, YTHDF2, YTHDF3,
the RNA molecule [26]. WTAP as a regulatory subunit is required for YTHDC1 and YTHDC2. m6A methylation plays critical roles on the regulation of
the formation METTL3-METTL14 m6A methyltransferase complex, RNA metabolism, including splicing, export, translation, and stability.
which plays a crucial role in the regulation of gene expression and al-
ternative splicing [27]. And KIAA1429 is also required for the intact 3. m6A methylation and hepatic growth
catalytic activity of the methyltransferase complex [21]. Furthermore,
RMB15 and RMB15B, two other WTAP interactors, can recruit me- The dynamic m6A modification affects tremendous genes expres-
thyltransferase complex to DRACH (the consensus sequence sites for sion, alternative splicing patterns and their related signaling pathways,
m6A methylation) [28]. which may highly involve the functions of cells and tissues [34,45]. The
The demethylated process of m6A “erasers” is mainly achieved correlation between m6A and hepatic growth has been shown by pro-
through two enzymes, including FTO [29] and alkB homolog 5 filing transcriptome-wide m6A in porcine liver at three developmental
(ALKBH5) [30]. FTO can catalyze the demethylation of m6A residues stages: newborn (0 day), suckling (21 days) and adult (2 years) [17]. In
both in vitro and in vivo [29]. FTO-mediated demethylation is involved liver tissue, about 33% post-transcripts of genes can occur as m6A
in m6A modification in different cell contexts such as ultraviolet-in- modification, with 1.33–1.42 m6A peaks per modified gene [17]. Me-
duced DNA damage response [31], adipogenesis [32], acute myeloid thylated post-transcripts of genes [5] can regulate several hepatic vital
leukemia cell transformation and leukemogenesis [33]. However, some functions such as growth, development, metabolism and protein cata-
researchers believe that FTO exhibits weak demethylase activities to- bolic [17]. Furthermore, the various levels of m6A methylation are
ward m6A in controlling mRNA stability and snRNA biogenesis, and N6, required for liver biological processes at any particular stage [17]
2′-O-dimethyladenosine (m6Am) as a related nucleotide of m6A might (Table 1). Therefore, the methylation of RNA m6A has profound im-
be real substrate for FTO [34,35]. Therefore, the effects of FTO on m6A pacts on hepatic growth.
and m6Am are further analyzed, and FTO is indeed the demethylase of
m6Am. But the abundance of m6A is much higher than that of m6Am,
and the change of m6A is more obvious in FTO knockout cell lines [33]. 4. m6A methylation and viral hepatitis
Recent study have founded that FTO can demethylate internal m6A and
cap m6Am in mRNA, internal m6A in U6 RNA, internal and cap m6Am m6A modification plays a pro-viral or anti-viral role in the genomes
in snRNAs, and N1-methyladenosine (m1A) in tRNA [36]. In addition, of RNA viruses and transcripts of a DNA virus [46]. For example, m6A
the overexpression of ALKBH5 can reduce the modification of m6A in methylation can strongly enhance HIV-1 protein and mRNA expression,
mRNAs [30] and small nucleolar RNAs [37]. Moreover, ALKBH5 has and virus replication by recruiting the cellular YTHDF m6A “reader”
been shown to promote the growth of breast cancer stem cells and proteins [47,48]. HIV replication and release can be suppressed by
glioblastoma stem cells [6] though increasing the demethylation of 3′ depletion of the METTL3 or METTL14 [48]. However, other study has
untranslated region of m6A in NANOG [38] and FOXM1 mRNAs [39], found that YTHDF1–3 proteins inhibit HIV-1 infection by blocking viral
respectively. reverse transcription and promoting degradation of viral RNA. There-
m6A “readers” contain YTH domain family proteins, heterogeneous fore, the functions of m6A in promoting/suppressing HIV infections are
nuclear ribonucleoproteins and on forth [40]. As the most important still controversial.
members of m6A “readers”, YTH domain family proteins including Hepatitis C virus (HCV) replication is negatively regulated though
YTHDF1, YTHDF2, YTHDF3, YTHDC1 and YTHDC2, regulate various m6A methylation mediated viral packaging inhibition [46]. YTHDF
gene expression through recognizing m6A modification [41]. The proteins can inhibit HCV replication by re-localizing to lipid droplets
translation of m6A-modified mRNA is promoted by YTHDF1 and and sites of viral assembly, and its depletion significantly enhances the
YTHDF3, the decay of m6A-modified mRNA is accelerated by YTHDF2 production of HCV infectious viral particles [49]. Furthermore, the
and YTHDF3, and the metabolism of m6A-modified mRNA in the cy- infectious HCV particle production can also be suppressed by methyl-
toplasm is enhanced by YTHDF1, YTHDF2 and YTHDF3 [41]. The nu- transferases METTL3 or METTL14, and enhanced by de-methyl-
clear export and splicing of m6A-modified mRNA are mediated by transferase FTO [49]. Interestingly, the presence of m6A modifications
YTHDC1 [42,43]. YTHDC2 enhances the translation efficiency of its in HCV RNA can be recognized to silence innate immune signaling
targets and also decreases their mRNA abundance by selectively caused by retinoic acid-inducible gene I (RIG-I-like) receptor [50]. And
binding m6A at its consensus motif [44]. previous studies have suggested that invading pathogen nucleic acids
In summary, the functions of m6A writers, erasers, readers were can be recognized and bound by cytoplasmic RIG-I-like receptors to
presented in Fig. 1. activate innate immune signaling [51].
HBV, a DNA virus, completes its life cycle by an RNA intermediate

46
K. Xu, et al. Analytical Biochemistry 578 (2019) 45–50

Table 1
The functions of RNA m6A methylation in liver.
m6A Regulators Functions Refs

Hepatic growth Unclear Methylation of post-transcripts of specific genes, including HOGA1, DPYS, FOLR1, GATM, CYP1A2, ALDH4A1, CYB5R3, SARDH, [17]
GSTZ1, BAAT, GNMT, EDEM2, MAFK, UBALD2, etc.
Hepatitis B METTL3 Enhancing reverse transcription of the HBV and inhibiting the expression of HBc and HBs proteins [52]
METTL14
Hepatitis C YTHDF Inhibiting HCV replication [49]
METTL3 Inhibiting infectious HCV particle production
METTL14 Inhibiting infectious HCV particle production
FTO Enhancing infectious HCV particle production
NAFLD METTL3 Suppressing adipogenesis [56]
FTO Promoting adipogenesis [55]
HCC METTL3 Promoting growth, migration and colony formation of HCC cells [63]
METTL14 Suppressing metastasis of HCC [64]
YTHDF1 Promoting cell cycle progression and metabolism of HCC [65]
YTHDF2 Promoting proliferation of HCC cells [14]
CCA WTAP Promoting TNM stage, lymph node metastasis and vascular invasion of HCC [67]

m6A methylation plays critical roles on the regulation of hepatic diseases, including virus replication, adipogenesis, growth and migration of HCC. Refs: references.

termed pregenomic RNA (pgRNA) [52]. The site of m6A methylation is 6. m6A methylation and liver cancer
located within the epsilon stem loop structure of the 5′and 3′ termini of
the pgRNA as well as the 3′ terminus of all HBV mRNAs [52]. These Liver cancer is the sixth common cancer worldwide, mainly in-
modifications are required for reverse transcription of pgRNA and ef- cluding hepatocellular carcinoma (HCC) and cholangiocarcinoma
ficient destabilization of all HBV transcripts [52]. Depletion of METTL3 (CCA) [62]. Recent studies have suggested that HCC is associated with
or METTL14 can reduce reverse transcription of the pgRNA, but lead to m6A “writers”, such as METTL3 and METTL14. METTL3 is up-regulated
up-regulation of HBc and HBs proteins, suggesting that m6A methyla- in HCC, and it can promote the growth, migration and colony formation
tion of HBV transcripts suppresses HBV proteins expression [52]. De- of HCC cells in vitro and facilitate tumorigenicity, growth and lung
pletion of either YTHDF2 or YTHDF3 significantly promoted the ex- metastasis of HCC in vivo through repressing suppressor of cytokine
pression of the HBV proteins HBs and HBc, suggesting that the YTHDF signaling 2 (SOCS2) mRNA [63]. SOCS2 mRNA stability can be
proteins also suppress HBV protein expression [52]. Furthermore, m6A downregulated through m6A-YTHDF2-dependent pathway [63]. In ad-
modification in HBV RNA is critical to modulate the viral life cycle and dition, overexpression of METTL3 can predict a poor prognosis of pa-
its related hepatitis by regulating gene expression and reverse tran- tients with HCC [63]. Down-regulation of METTL14 plays an important
scription [52]. role in tumor metastasis, and it is regarded as a poor prognostic in-
Apparently, present studies show that m6A modification negatively dicator for recurrence-free survival of HCC [64]. METTL14-dependent
regulates HBV/HCV infections. Therefore, m6A modification has a ne- m6A methylation positively regulates the primary microRNA 126 pro-
gative impact on hepatic virus infection but its roles in regulating other cess by the microprocessor protein DGCR8, whereas microRNA 126 can
hepatitis viruses need to be further clarified. Especially, because the suppress the inhibiting effect of METTL14 in tumor metastasis [64].
functions of m6A in promoting/suppressing HIV infections are still Moreover, m6A “readers”, mainly including YTHDF1 and YTHDF2, are
controversial, its roles in regulating other hepatitis viruses maybe are also associated with HCC. YTHDF1 is remarkably overexpressed in
different. HCC, and has been shown to positively correlate with pathology stage
by promoting HCC cell cycle progression and metabolism [65].
YTHDF2 can enhance mRNA degradation cells by identifying mRNA
5. m6A methylation and non-alcoholic fatty liver disease (NAFLD) m6A sites, resulting in promoting proliferation of HCC [14]. However,
miR-145 can suppress proliferation of HCC cells by down-regulating
Non-alcoholic fatty liver disease (NAFLD) is characterized by he- YTHDF2 through targeting its mRNA 3′UTR [14]. Therefore, WTAP
patic steatosis with no history of significant alcohol use or other known may be a promising target for the treatment of CCA.
liver disease [53], and hepatic steatosis is caused by metabolic dysre- WTAP is associated with cell proliferation and apoptosis in glio-
gulation of de novo lipogenesis, fatty acid uptake, fatty acid oxidation, blastoma [66]. WTAP is also overexpressed in CCA, which plays a po-
and triglycerides export [54]. Currently, m6A demethylation has been sitive role in CCA TNM stage, lymph node metastasis and vascular in-
shown to play a vital role in the regulation of adipogenesis. FTO posi- vasion [67]. The migration, invasion and tumorigenicity of CCA cells
tively regulated adipogenesis by m6A demethylation, but METTL3 ne- can be inhibited by WTAP siRNA, but the proliferation is insignificantly
gatively correlated with adipogenesis by m6A methylation [55,56]. affected by WTAP siRNA or overexpression [67]. Moreover, WTAP
More importantly, FTO can promote adipogenesis by inhibiting the could promote the expression of metastasis-related genes, including
Wnt/β-catenin signaling pathway in porcine intramuscular pre- MMP7, MMP28, and Muc1 [67]. Therefore, WTAP may be a promising
adipocytes [57], and enhancing RUNX1T1 mediated adipocyte pro- target for the treatment of CCA.
liferation [58], as well as promoting Cyclin A2 (CCNA2) and cyclin
dependent kinase 2 (CDK2) mediated mitotic clonal expansion at the
early stage of adipocyte differentiation [59]. And YTHDF2 can inverse 7. Clinical significance of m6A
FTO-mediated adipogenesis by m6A methylation [59]. In humans, a
significant increase in FTO mRNA and protein levels has been found in The m6A methylation of RNAs plays important roles in maintenance
the liver of NAFLD patients [60]. Elevated levels of FTO mRNA and and progression of liver diseases, and its invention may be a potentially
protein can be also found in a NAFLD rat, which are involved in oxi- effective treatment. For example, the curcumin has protective effects in
dative stress and lipid deposition [61]. All in all, FTO may be a pro- the LPS-induced liver injury and disruption of hepatic lipid metabolism,
misingly therapeutic target to improve hepatic steatosis. probably owning to the increasing level of m6A RNA methylation [68].
Furthermore, recent studies have suggested that betaine has the hepa-
toprotective effects in NAFLD through improving hepatic m6A

47
K. Xu, et al. Analytical Biochemistry 578 (2019) 45–50

methylation status by decreasing FTO expression [69,70], and recti- Acknowledgments


fying the decreased m6A methylation in adipose tissue by promoting
FTO expression [71]. However, natural products like curcumin and None.
betaine, have various effects such as antioxidant, anti-inflammatory,
and anticancer properties [72,73]. Therefore, it is difficult to evaluate Appendix A. Supplementary data
the role and effect of m6A modification in treatment of related liver
diseases by curcumin or betaine. Supplementary data to this article can be found online at https://
Moreover, inhibition of the RNA demethylase, FTO, can suppress doi.org/10.1016/j.ab.2019.05.005.
the vital activity of HBV/HCV and their hepatitis progression [49,52].
Therefore, FTO inhibitors may provide potential therapeutic agents for References
viral hepatitis such as rhein, αKG analogues, CHTB, meclofenamic acid,
4-chloro-6-(6′-chloro-7′-hydroxy-2′,4′,4′-trimethyl- [1] R. Desrosiers, K. Friderici, F. Rottman, Identification of methylated nucleosides in
Chroman-2′-yl), benzene-1,3-diol (CHTB), N-(5-Chloro-2,4-dihy- messenger RNA from Novikoff hepatoma cells, Proc. Natl. Acad. Sci. U. S. A. 71 (10)
(1974) 3971–3975.
droxyphenyl)-1-phenylcyclobutanecarboxamide (N-CDPCB), nafamo- [2] Guifang. Methylation modifications in eukaryotic messenger RNA, J. Genet.
stat mesylate, and Radicicol and on forth [74–82]. However, the clin- Genom. 41 (1) (2014) 21–33.
ical application or off target effects of these inhibitors may be further [3] A. Li, Y.S. Chen, X.L. Ping, X. Yang, W. Xiao, Y. Yang, et al., Cytoplasmic m6A
reader YTHDF3 promotes mRNA translation, Cell Res. 27 (3) (2017) 444–447.
identified. [4] D. Dan, M.M. Sharon, S. Schraga, S.D. Mali, U. Lior, O. Sivan, et al., Topology of the
Besides, S-adenosylhomocysteine (SAH) can be hydrolyzed by SAH human and mouse m6A RNA methylomes revealed by m6A-seq, Nature 485 (7397)
hydrolase into adenosine and homocysteine, and it is reported as a (2012) 201.
[5] K. Meyer, Y. Saletore, P. Zumbo, O. Elemento, C. Mason, S. Jaffrey, Comprehensive
competitive inhibitor of adenosylmethionine-dependent methyl-
analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons,
transferases [83]. 3-Deazaadenosine (DAA), a SAH hydrolysis inhibitor, Cell 149 (7) (2012) 1635–1646.
can inhibit m6A modification of mRNA substrates [84]. Furthermore, [6] T. Pan, N 6-methyl-adenosine modification in messenger and long non-coding RNA,
Trends Biochem. Sci. 38 (4) (2013) 204–209.
DAA and its analogues can inhibit virus replication by editing m6A-
[7] Z. Liu, J. Zhang, Human C-to-U coding RNA editing is largely nonadaptive, Mol.
containing mRNA [85,86]. In addition, IGF-1 can direct nuclear 26 S Biol. Evol. 35 (3) (2018).
proteasome mediated WTAP degradation by IGF-1 receptor-phospha- [8] X. Wang, Z. Lu, A. Gomez, G.C. Hon, Y. Yue, D. Han, et al., N6-methyladenosine-
tidylinositol 3-kinase-Akt signaling [87], and it may be a novel ther- dependent regulation of messenger RNA stability, Nature 505 (7481) (2014)
117–120.
apeutic agent for treatment of CCA. [9] X. Wang, B.S. Zhao, I.A. Roundtree, Z. Lu, D. Han, H. Ma, et al., N(6)-methylade-
nosine modulates messenger RNA translation efficiency, Cell 161 (6) (2015)
1388–1399.
[10] D. Dominissini, S. Moshitch-Moshkovitz, S. Schwartz, M. Salmon-Divon, L. Ungar,
8. Conclusions and perspectives S. Osenberg, et al., Topology of the human and mouse m6A RNA methylomes re-
vealed by m6A-seq, Nature 485 (7397) (2012) 201–206.
In conclusion, m6A modification is highly involved in the regulation [11] B.S. Zhao, I.A. Roundtree, C. He, Post-transcriptional gene regulation by mRNA
modifications, Nat. Rev. Mol. Cell Biol. 18 (1) (2017) 31–42.
of hepatic growth and hepatic diseases by modifying mRNAs and [12] J. Choi, K.W. Ieong, H. Demirci, J. Chen, A. Petrov, A. Prabhakar, et al., N(6)-
ncRNAs (Table 1). m6A-mediated gene post-transcriptional modifica- methyladenosine in mRNA disrupts tRNA selection and translation-elongation dy-
tion plays physiological roles in regulation of proteins production, cir- namics, Nat. Struct. Mol. Biol. 23 (2) (2016) 110–115.
[13] T. B, S. R, K. R, L. K-H, B. H, N6-adenosine methylation in MiRNAs, PLoS One 10 (2)
cadian rhythms, cell cycle, reprogramming, cell differentiation, state (2015) e0118438.
transitions, stress responses and cancer [88]. And the expressions of [14] Z. Yang, J. Li, G. Feng, S. Gao, Y. Wang, S. Zhang, et al., MicroRNA-145 modulates
ncRNAs (miRNAs, lncRNAs and circRNAs) are closely associated with N6-methyladenosine levels by targeting the 3'-untranslated mRNA region of the N6-
methyladenosine binding YTH domain family 2 protein, J. Biol. Chem. 292 (9)
the development of liver diseases, and they are regarded as potential
(2017) 3614–3623.
targets for early diagnosis and therapeutic intervention [89]. In addi- [15] R. Jacob, S. Zander, T. Gutschner, The dark side of the epitranscriptome: chemical
tion, RNA m6A methylation has multi-functions in regulation of hepatic modifications in long non-coding RNAs, Int. J. Mol. Sci. 18 (11) (2017) 2387.
growth, HBV and HCV replication, adipogenesis, as well as HCC and [16] A.S. Warda, J. Kretschmer, P. Hackert, C. Lenz, H. Urlaub, C. Hã¶Bartner, et al.,
Human METTL16 is a N6-methyladenosine (m6A) methyltransferase that targets
CCA progression. Many promising agents can be used to target m6A in pre-mRNAs and various non-coding RNAs, EMBO Rep. 18 (11) (2017) 2004.
the treatment of liver diseases such as FTO inhibitors, SAH hydrolysis [17] S. He, H. Wang, R. Liu, M. He, T. Che, L. Jin, et al., mRNA N6-methyladenosine
inhibitors, WTAP inhibitors and other natural productions. methylation of postnatal liver development in pig, PLoS One 12 (3) (2017)
e0173421.
In summary, methylated m6A level may be potential indicator for [18] J.A. Bokar, M.E. Shambaugh, D. Polayes, A.G. Matera, F.M. Rottman, Purification
early diagnosis of hepatitis and liver cancer, and m6A may be a pro- and cDNA Cloning of the AdoMet-binding subunit of the human mrna (n6-adeno-
mising target for the treatment of liver diseases. In addition, the func- sine)-methyltransferase, Rna-a Publication of the Rna Society 3 (11) (1997) 1233.
[19] J. Liu, Y. Yue, D. Han, X. Wang, Y. Fu, L. Zhang, et al., A METTL3–METTL14
tion of m6A in promoting/suppressing viral infections exists different complex mediates mammalian nuclear RNA N6-adenosine methylation, Nat. Chem.
views. Therefore, the pro/antivirus effects of m6A methylation on he- Biol. 10 (2) (2014) 93–95.
patic viruses need to be identified. Recent reports have suggested that [20] X.L. Ping, B.F. Sun, L. Wang, W. Xiao, X. Yang, W.J. Wang, et al., Mammalian WTAP
is a regulatory subunit of the RNA N6-methyladenosine methyltransferase, Cell Res.
m6A RNA methylation plays important roles in self-renewal, tumor- 24 (2) (2014) 177–189.
igenesis and radio-resistance of glioblastoma stem cells by METTL3 [21] S. Schwartz, M. Mumbach, M. Jovanovic, T. Wang, K. Maciag, G.G. Bushkin, et al.,
[90,91]. The effect of m6A on liver cancer stem cells needs to further be Perturbation of m6A writers reveals two distinct classes of mRNA methylation at
internal and 5′ sites, Cell Rep. 8 (1) (2014) 284–296.
identified. Furthermore, the effects of ncRNAs by methylated m6A on
[22] D.P. Patil, C.K. Chen, B.F. Pickering, A. Chow, C. Jackson, M. Guttman, et al., m(6)A
liver diseases remain unclear. The further studies on methylated RNA methylation promotes XIST-mediated transcriptional repression, Nature 537
ncRNAs would help us to better understand their roles in hepatic dis- (7620) (2016) 369.
eases or other diseases. [23] S. Geula, S. Moshitchmoshkovitz, D. Dan, A.F. Mansour, N. Kol, M. Salmondivon,
et al., m6A mRNA methylation facilitates resolution of naïve pluripotency toward
differentiation, Science 347 (6225) (2015) 1002–1006.
[24] B. Slobodin, R. Han, V. Calderone, J.A.F.O. Vrielink, F. Loayza-Puch, E. Ran, et al.,
Authors contribution Transcription impacts the efficiency of mRNA translation via Co-transcriptional N6-
adenosine methylation, Cell 169 (2) (2017) 326–337.
[25] X. Wang, J. Feng, Y. Xue, Z. Guan, D. Zhang, Z. Liu, et al., Corrigendum: structural
Writing-Original Draft preparation, Kechen Xu and Yuchao Sun, basis of N6-adenosine methylation by the METTL3-METTL14 complex, Nature 542
Baixiang Sheng, Yiwen Zheng, Xiaoli Wu; Writing-Review & Editing, (7640) (2017) 260.
[26] X. Wang, J. Feng, Y. Xue, Z. Guan, D. Zhang, Z. Liu, et al., Structural basis of N(6)-
Keyang Xu. adenosine methylation by the METTL3-METTL14 complex, Nature 534 (7608)

48
K. Xu, et al. Analytical Biochemistry 578 (2019) 45–50

(2016) 575–578. preadipocytes, Anim. Biotechnol. 28 (4) (2017) 1–7.


[27] X.-L. Ping, B.-F. Sun, L. Wang, W. Xiao, X. Yang, W.-J. Wang, et al., Mammalian [58] M. Merkestein, S. Laber, F. Mcmurray, D. Andrew, G. Sachse, J. Sanderson, et al.,
WTAP is a Regulatory subunit of the RNA N6-methyladenosine methyltransferase FTO influences adipogenesis by regulating mitotic clonal expansion, Nat. Commun.
24 (2014), pp. 177–189 2. 6 (2015) 6792.
[28] D.P. Patil, C.K. Chen, B.F. Pickering, A. Chow, C. Jackson, M. Guttman, et al., m(6)A [59] R. Wu, Y. Liu, Y. Yao, Y. Zhao, Z. Bi, Q. Jiang, et al., FTO regulates adipogenesis by
RNA methylation promotes XIST-mediated transcriptional repression, Nature 537 controlling cell cycle progression via m(6)A-YTHDF2 dependent mechanism,
(7620) (2016) 369–373. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1863 (10) (2018) 1323–1330.
[29] G. Jia, Y. Fu, X. Zhao, Q. Dai, G. Zheng, Y. Yang, et al., N6-methyladenosine in [60] J.R.W. Guo, X. Li, G. Xi, Y. Li, L. Gao, J. Liu, D. Su, Altering of FTO in the serum and
nuclear RNA is a major substrate of the obesity-associated FTO, Nat. Chem. Biol. 7 livers of NAFLD patients: a correlation analysis, Int. J. Clin. Exp. Med. 2018 (11)
(12) (2011) 885–887. (2018) 6046–6053.
[30] G. Zheng, J.A. Dahl, Y. Niu, P. Fedorcsak, C.M. Huang, C. Li, et al., ALKBH5 is a [61] J. Guo, W. Ren, A. Li, Y. Ding, W. Guo, D. Su, et al., Fat mass and obesity-associated
mammalian RNA demethylase that impacts RNA metabolism and mouse fertility, gene enhances oxidative stress and lipogenesis in nonalcoholic fatty liver disease,
RNA Biol. 49 (6) (2013) 18–29. Dig. Dis. Sci. 58 (4) (2013) 1004–1009.
[31] Y. Xiang, B. Laurent, C.H. Hsu, S. Nachtergaele, Z. Lu, W. Sheng, et al., RNA m6A [62] L. Gravitz, Liver cancer, Nature 516 (7529) (2014) S1.
methylation regulates the ultraviolet-induced DNA damage response, Nature 552 [63] M. Chen, L. Wei, C.T. Law, F.H. Tsang, J. Shen, C.L. Cheng, et al., RNA N6-me-
(7685) (2017) 573–576. thyladenosine methyltransferase-like 3 promotes liver cancer progression through
[32] D. Christian, M. David, G. Sophie, D. Emmanuelle, Kr Antje, J. Peter, et al., YTHDF2-dependent posttranscriptional silencing of SOCS2, Hepatology 67 (6)
Variation in FTO contributes to childhood obesity and severe adult obesity, Nat. (2018) 2254.
Genet. 39 (6) (2007) 724–726. [64] J.Z. Ma, F. Yang, C.C. Zhou, F. Liu, J.H. Yuan, F. Wang, et al., METTL14 suppresses
[33] R. Su, L. Dong, C. Li, S. Nachtergaele, M. Wunderlich, Y. Qing, et al., R-2HG exhibits the metastatic potential of hepatocellular carcinoma by modulating
anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling, Cell 172 (1–2) N6‐methyladenosine‐dependent primary MicroRNA processing, Hepatology 65 (2)
(2018) 90–105 e123. (2017) 529.
[34] J. Mauer, M. Sindelar, V. Despic, T. Guez, B.R. Hawley, J.-J. Vasseur, et al., FTO [65] X. Zhao, Y. Chen, Q. Mao, X. Jiang, W. Jiang, J. Chen, et al., Overexpression of
controls reversible m6Am RNA methylation during snRNA biogenesis, Nat. Chem. YTHDF1 is associated with poor prognosis in patients with hepatocellular carci-
Biol. 15 (4) (2019) 340–347. noma, Cancer Biomark. 21 (4) (2018) 859.
[35] J. Mauer, X. Luo, A. Blanjoie, X. Jiao, A.V. Grozhik, D.P. Patil, et al., Reversible [66] J. Du-Il, L.S. Weon, H. Myoung-Eun, K. Hyun-Jung, S. Seon-Ae, H. Gi-Yeong, et al.,
methylation of m6Am in the 5' cap controls mRNA stability, Nature 541 (7637) Expression and roles of Wilms' tumor 1-associating protein in glioblastoma, Cancer
(2017) 371–375. Sci. 103 (12) (2012) 2102–2109.
[36] J. Wei, F. Liu, Z. Lu, Q. Fei, Y. Ai, P.C. He, et al., Differential m(6)A, m(6)Am, and m [67] H.J. Jo, H.E. Shim, M.E. Han, H.J. Kim, K.S. Kim, S. Baek, et al., WTAP regulates
(1)A demethylation mediated by FTO in the cell nucleus and cytoplasm, Mol. Cell migration and invasion of cholangiocarcinoma cells, J. Gastroenterol. 48 (11)
71 (6) (2018) 973–985 e975. (2013) 1271–1282.
[37] B. Linder, A.V. Grozhik, A.O. Olarerin-George, C. Meydan, C.E. Mason, S.R. Jaffrey, [68] N. Lu, X. Li, J. Yu, Y. Li, C. Wang, L. Zhang, et al., Curcumin attenuates lipopoly-
Single-nucleotide-resolution mapping of m6A and m6Am throughout the tran- saccharide-induced hepatic lipid metabolism disorder by modification of m 6 a RNA
scriptome, Nat. Methods 12 (8) (2015) 767–772. methylation in piglets, Lipids 53 (1) (2018) 53–63.
[38] C. Zhang, D. Samanta, H. Lu, J.W. Bullen, H. Zhang, I. Chen, et al., Hypoxia induces [69] K.K. Kharbanda, D.D.R. Ii, M.E. Mailliard, G.L. Siford, A.J. Barak,
the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated H.C. Beckenhauer, et al., Role of elevated S-adenosylhomocysteine in rat hepatocyte
m6A-demethylation of NANOG mRNA, Proc. Natl. Acad. Sci. U. S. A. 113 (14) apoptosis: protection by betaine, Biochem. Pharmacol. 70 (12) (2005) 1883–1890.
(2016) E2047. [70] L-j Wang, H-w Zhang, J-y Zhou, Y. Liu, Y. Yang, X-l Chen, et al., Betaine attenuates
[39] S. Zhang, B.S. Zhao, A. Zhou, K. Lin, S. Zheng, Z. Lu, et al., m 6 a demethylase hepatic steatosis by reducing methylation of the MTTP promoter and elevating
ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining genomic methylation in mice fed a high-fat diet ☆, J. Nutr. Biochem. 25 (3) (2014)
FOXM1 expression and cell proliferation program, Cancer Cell 31 (4) (2017) 591. 329–336.
[40] B.S. Zhao, I.A. Roundtree, C. He, Post-transcriptional gene regulation by mRNA [71] X. Zhou, J. Chen, J. Chen, W. Wu, X. Wang, Y. Wang, The beneficial effects of
modifications, Nat. Rev. Mol. Cell Biol. 18 (1) (2017) 31. betaine on dysfunctional adipose tissue and N6-methyladenosine mRNA methyla-
[41] H. Shi, W. Xiao, Z. Lu, B.S. Zhao, H. Ma, P.J. Hsu, et al., YTHDF3 facilitates tion requires the AMP-activated protein kinase α1 subunit, JNB (J. Nutr. Biochem.)
translation and decay of N6-methyladenosine-modified RNA, Cell Res. 27 (3) 26 (12) (2015) 1678–1684.
(2017) 315–328. [72] T. Tsuda, Curcumin as a functional food-derived factor: degradation products,
[42] I.A. Roundtree, G.Z. Luo, Z. Zhang, X. Wang, T. Zhou, Y. Cui, et al., YTHDC1 metabolites, bioactivity, and future perspectives, Food & function 9 (2) (2018)
mediates nuclear export of N6-methyladenosine methylated mRNAs, 2017-10-04, 705–714.
eLife 6 (2017) 6:e31311. [73] J.M. Cholewa, L. Guimaraes-Ferreira, N.E. Zanchi, Effects of betaine on perfor-
[43] W. Xiao, S. Adhikari, U. Dahal, Y.-S. Chen, Y.-J. Hao, B.-F. Sun, et al., Nuclear m 6 a mance and body composition: a review of recent findings and potential mechan-
reader YTHDC1 regulates mRNA splicing, Mol. Cell 61 (4) (2016) 507–519. isms, Amino Acids 46 (8) (2014) 1785–1793.
[44] P.J. H, Y. Z, H. M, Y. G, X. S, Y. L, et al., Ythdc2 is an N(6)-methyladenosine binding [74] Q. Yan, B. Zhou, M. Zhang, W. Liu, Z. Han, C. Song, et al., A novel inhibitor of the
protein that regulates mammalian spermatogenesis, Cell Res. 27 (9) (2017) 1115. obesity-related protein FTO, Biochemistry 55 (10) (2016) 1516.
[45] I.A. Roundtree, M.E. Evans, T. Pan, C. He, Dynamic RNA modifications in gene [75] B. Chen, F. Ye, L. Yu, G. Jia, X. Huang, X. Zhang, et al., Development of cell-active
expression regulation, Cell 169 (7) (2017) 1187–1200. N6-methyladenosine RNA demethylase FTO inhibitor, J. Am. Chem. Soc. 134 (43)
[46] B. Tan, S.J. Gao, RNA epitranscriptomics: regulation of infection of RNA and DNA (2012) 17963–17971.
viruses by N6 -methyladenosine (m6 A), Quaest. Math. 29 (1) (2018) 125–131. [76] H. Yue, Y. Jingli, L. Qi, L. Jiafei, G. Shouzhe, Z. Hu, et al., Meclofenamic acid
[47] E.M. Kennedy, H.P. Bogerd, A.V. Kornepati, D. Kang, D. Ghoshal, J.B. Marshall, selectively inhibits FTO demethylation of m6A over ALKBH5, Nucleic Acids Res. 43
et al., Posttranscriptional m(6)A editing of HIV-1 mRNAs enhances viral gene ex- (1) (2015) 373–384.
pression, Cell Host Microbe 19 (5) (2016) 675–685. [77] W. Aik, M. Demetriades, M.K. Hamdan, E.A. Bagg, K.K. Yeoh, C. Lejeune, et al.,
[48] S. Nachtergaele, C. He, Chemical modifications in the life of an mRNA transcript, Structural basis for inhibition of the fat mass and obesity associated protein (FTO),
Annu. Rev. Genet. 52 (2018) 349–372. J. Med. Chem. 56 (9) (2013) 3680–3688.
[49] N. Gokhale, A.R. Mcintyre, M. Mcfadden, A. Roder, E. Kennedy, J. Gandara, et al., [78] G. Zheng, T. Cox, L. Tribbey, G.Z. Wang, P. Iacoban, M.E. Booher, et al., Synthesis of
N6 -methyladenosine in flaviviridae viral RNA genomes regulates infection, Cell a FTO inhibitor with anticonvulsant activity, ACS Chem. Neurosci. 5 (8) (2014)
Host Microbe 20 (5) (2016) 654–665. 658–665.
[50] A.F. Durbin, C. Wang, J. Marcotrigiano, L. Gehrke, RNAs containing modified nu- [79] J.D.W. Toh, L. Sun, L.Z.M. Lau, J. Tan, J.J.A. Low, C.W.Q. Tang, et al., A strategy
cleotides fail to trigger RIG-I conformational changes for innate immune signaling, based on nucleotide specificity leads to a subfamily-selective and cell-active in-
mBio 7 (5) (2016). hibitor of N(6)-methyladenosine demethylase FTO, Chem. Sci. 6 (1) (2015)
[51] M.U. Gack, Mechanisms of RIG-I-like receptor activation and manipulation by viral 112–122.
pathogens, J. Virol. 88 (10) (2014) 5213. [80] Y. Qiao, B. Zhou, M. Zhang, W. Liu, Z. Han, C. Song, et al., A novel inhibitor of the
[52] H. Imam, M. Khan, N.S. Gokhale, A.B.R. McIntyre, G.W. Kim, J.Y. Jang, et al., N6- obesity-related protein FTO, Biochemistry 55 (10) (2016) 1516–1522.
methyladenosine modification of hepatitis B virus RNA differentially regulates the [81] R. Wang, Z. Han, B. Liu, B. Zhou, N. Wang, Q. Jiang, et al., Identification of natural
viral life cycle, Proc. Natl. Acad. Sci. U. S. A. 115 (35) (2018) 8829–8834. compound Radicicol as a potent FTO inhibitor, Mol. Pharm. 15 (9) (2018)
[53] D.E. Kleiner, E.M. Brunt, M. Van Natta, C. Behling, M.J. Contos, O.W. Cummings, 4092–4098.
et al., Design and validation of a histological scoring system for nonalcoholic fatty [82] X. Han, N. Wang, J. Li, Y. Wang, R. Wang, J. Chang, Identification of nafamostat
liver disease, Hepatology 41 (6) (2005) 1313–1321. mesilate as an inhibitor of the fat mass and obesity-associated protein (FTO) de-
[54] W. Liu, H. Cao, J. Yan, R. Huang, H. Ying, Micro-managers' of hepatic lipid meta- methylase activity, Chem. Biol. Interact. 297 (2019) 80–84.
bolism and NAFLD, Wiley interdisciplinary reviews RNA 6 (5) (2015) 581–593. [83] D. Kloor, H. Osswald, S-Adenosylhomocysteine hydrolase as a target for in-
[55] X. Zhao, Y. Yang, B.F. Sun, Y. Shi, X. Yang, W. Xiao, et al., FTO-dependent de- tracellular adenosine action, Trends Pharmacol. Sci. 25 (6) (2004) 294–297.
methylation of N6-methyladenosine regulates mRNA splicing and is required for [84] J.P. Bader, N.R. Brown, P.K. Chiang, G.L. Cantoni, 3-Deazaadenosine, an inhibitor
adipogenesis, Cell Res. 24 (12) (2014) 1403–1419. of adenosylhomocysteine hydrolase, inhibits reproduction of rous sarcoma virus
[56] X. Wang, L. Zhu, J. Chen, Y. Wang, mRNA m(6)A methylation downregulates and transformation of chick embryo cells, Virology 89 (2) (1978) 494–505.
adipogenesis in porcine adipocytes, Biochem. Biophys. Res. Commun. 459 (2) [85] R.K. Gordon, G. Krzysztof, W.R. Rudnicki, R. Leszek, M.C. Pankaskie, J.M. Bujnicki,
(2015) 201–207. et al., Anti-HIV-1 activity of 3-deaza-adenosine analogs. Inhibition of S-adeno-
[57] X. Chen, Y. Luo, J. Gang, G. Liu, Z. Hua, Z. Huang, FTO promotes adipogenesis sylhomocysteine hydrolase and nucleotide congeners, FEBS J. 270 (17) (2010)
through inhibition of the wnt/β-catenin signaling pathway in porcine intramuscular 3507–3517.

49
K. Xu, et al. Analytical Biochemistry 578 (2019) 45–50

[86] W.Y. Gao, D.G. Johns, H. Mitsuya, Anti-human immunodeficiency virus type 1 [89] L. Xu, M. Zhang, X. Zheng, P. Yi, C. Lan, M. Xu, The circular RNA ciRS-7 (Cdr1as)
activity of hydroxyurea in combination with 2',3'-dideoxynucleosides, Mol. acts as a risk factor of hepatic microvascular invasion in hepatocellular carcinoma,
Pharmacol. 46 (4) (1994) 767–772. J. Cancer Res. Clin. Oncol. 143 (1) (2017) 17–27.
[87] T.W. Small, P. J Geoffrey, Nuclear degradation of Wilms tumor 1-associating pro- [90] Q. Cui, H. Shi, P. Ye, L. Li, Q. Qu, G. Sun, et al., m(6)A RNA methylation regulates
tein and survivin splice variant switching underlie IGF-1-mediated survival, J. Biol. the self-renewal and tumorigenesis of glioblastoma stem cells, Cell Rep. 18 (11)
Chem. 284 (37) (2009) 24684–24695. (2017) 2622–2634.
[88] J.M. Fustin, M. Doi, Y. Yamaguchi, H. Hida, S. Nishimura, M. Yoshida, et al., RNA- [91] A. Visvanathan, V. Patil, A. Arora, A.S. Hegde, A. Arivazhagan, V. Santosh, et al.,
Methylation-Dependent RNA processing controls the speed of the circadian clock, Essential role of METTL3-mediated m(6)A modification in glioma stem-like cells
Cell 155 (4) (2013) 793–806. maintenance and radioresistance, Oncogene 37 (4) (2018) 522–533.

50

You might also like