Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2018, Vol. 12 Issue (4) : 481-489    https://doi.org/10.1007/s11684-018-0654-8
REVIEW
RNA m6A modification and its function in diseases
Jiyu Tong1, Richard A. Flavell2,3(), Hua-Bing Li1()
1. Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
2. Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
3. Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
 Download: PDF(215 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

N6-methyladenosine (m6A) is the most common post-transcriptional RNA modification throughout the transcriptome, affecting fundamental aspects of RNA metabolism. m6A modification could be installed by m6A “writers” composed of core catalytic components (METTL3/METTL14/WTAP) and newly defined regulators and removed by m6A “erasers” (FTO and ALKBH5). The function of m6A is executed by m6A “readers” that bind to m6A directly (YTH domain-containing proteins, eIF3 and IGF2BPs) or indirectly (HNRNPA2B1). In the past few years, advances in m6A modulators (“writers,” “erasers,” and “readers”) have remarkably renewed our understanding of the function and regulation of m6A in different cells under normal or disease conditions. However, the mechanism and the regulatory network of m6A are still largely unknown. Moreover, investigations of the m6A physiological roles in human diseases are limited. In this review, we summarize the recent advances in m6A research and highlight the functional relevance and importance of m6A modification in in vitro cell lines, in physiological contexts, and in cancers.

Keywords RNA modification      m6A      immunity      cancer      epigenetics     
Corresponding Author(s): Richard A. Flavell,Hua-Bing Li   
Just Accepted Date: 10 July 2018   Online First Date: 10 August 2018    Issue Date: 03 September 2018
 Cite this article:   
Jiyu Tong,Richard A. Flavell,Hua-Bing Li. RNA m6A modification and its function in diseases[J]. Front. Med., 2018, 12(4): 481-489.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-018-0654-8
https://academic.hep.com.cn/fmd/EN/Y2018/V12/I4/481
Fig.1  Dynamic m6A system. m6A marker is installed at the 3′-UTR by m6A “writers” co-transcriptionally and could be reversed by m6A “erasers.” The marker is recognized in nucleus and cytoplasm by different m6A “readers” and executes different functions involved in RNA metabolism, including splicing, export, translation, and RNA decay.
Fig.2  Does “RNA code” exist? How over 100 histone modifications coordinate to regulate the transcription prompted the well-known concepts of histone code hypothesis. Similarly, over 100 different RNA modifications are known, and how those different RNA modifications may regulate the RNA metabolism may constitute the “RNA code.”
1 He C. Grand challenge commentary: RNA epigenetics? Nat Chem Biol 2010; 6(12): 863–865
https://doi.org/10.1038/nchembio.482 pmid: 21079590
2 Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci USA 1974; 71(10): 3971–3975
https://doi.org/10.1073/pnas.71.10.3971 pmid: 4372599
3 Munns TW, Sims HF. Methylation and processing of transfer ribonucleic acid in mammalian and bacterial cells. J Biol Chem 1975; 250(6): 2143–2149
pmid: 1090617
4 Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 2012; 149(7): 1635–1646
https://doi.org/10.1016/j.cell.2012.05.003 pmid: 22608085
5 Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, Sorek R, Rechavi G. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 2012; 485(7397): 201–206
https://doi.org/10.1038/nature11112 pmid: 22575960
6 Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang YG, He C. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 2011; 7(12): 885–887
https://doi.org/10.1038/nchembio.687 pmid: 22002720
7 Bokar JA, Rath-Shambaugh ME, Ludwiczak R, Narayan P, Rottman F. Characterization and partial purification of mRNA N6-adenosine methyltransferase from HeLa cell nuclei. Internal mRNA methylation requires a multisubunit complex. J Biol Chem 1994; 269(26): 17697–17704
pmid: 8021282
8 Bokar JA, Shambaugh ME, Polayes D, Matera AG, Rottman FM. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA 1997; 3(11): 1233–1247
pmid: 9409616
9 Batista PJ, Molinie B, Wang J, Qu K, Zhang J, Li L, Bouley DM, Lujan E, Haddad B, Daneshvar K, Carter AC, Flynn RA, Zhou C, Lim KS, Dedon P, Wernig M, Mullen AC, Xing Y, Giallourakis CC, Chang HY. m6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 2014; 15(6): 707–719
https://doi.org/10.1016/j.stem.2014.09.019 pmid: 25456834
10 Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z, Zhao JC. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol 2014; 16(2): 191–198
https://doi.org/10.1038/ncb2902 pmid: 24394384
11 Chen T, Hao YJ, Zhang Y, Li MM, Wang M, Han W, Wu Y, Lv Y, Hao J, Wang L, Li A, Yang Y, Jin KX, Zhao X, Li Y, Ping XL, Lai WY, Wu LG, Jiang G, Wang HL, Sang L, Wang XJ, Yang YG, Zhou Q. m6A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell 2015; 16(3): 289–301
https://doi.org/10.1016/j.stem.2015.01.016 pmid: 25683224
12 Zhong S, Li H, Bodi Z, Button J, Vespa L, Herzog M, Fray RG. MTA is an Arabidopsis messenger RNA adenosine methylase and interacts with a homolog of a sex-specific splicing factor. Plant Cell 2008; 20(5): 1278–1288
https://doi.org/10.1105/tpc.108.058883 pmid: 18505803
13 Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AA, Kol N, Salmon-Divon M, Hershkovitz V, Peer E, Mor N, Manor YS, Ben-Haim MS, Eyal E, Yunger S, Pinto Y, Jaitin DA, Viukov S, Rais Y, Krupalnik V, Chomsky E, Zerbib M, Maza I, Rechavi Y, Massarwa R, Hanna S, Amit I, Levanon EY, Amariglio N, Stern-Ginossar N, Novershtern N, Rechavi G, Hanna JH. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 2015; 347(6225): 1002–1006
https://doi.org/10.1126/science.1261417 pmid: 25569111
14 Schwartz S, Agarwala SD, Mumbach MR, Jovanovic M, Mertins P, Shishkin A, Tabach Y, Mikkelsen TS, Satija R, Ruvkun G, Carr SA, Lander ES, Fink GR, Regev A. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis. Cell 2013; 155(6): 1409–1421
https://doi.org/10.1016/j.cell.2013.10.047 pmid: 24269006
15 Licatalosi DD, Mele A, Fak JJ, Ule J, Kayikci M, Chi SW, Clark TA, Schweitzer AC, Blume JE, Wang X, Darnell JC, Darnell RB. HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature 2008; 456(7221): 464–469
https://doi.org/10.1038/nature07488 pmid: 18978773
16 Ke S, Pandya-Jones A, Saito Y, Fak JJ, Vågbø CB, Geula S, Hanna JH, Black DL, Darnell JE Jr, Darnell RB. m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev 2017; 31(10): 990–1006
https://doi.org/10.1101/gad.301036.117 pmid: 28637692
17 Knuckles P, Carl SH, Musheev M, Niehrs C, Wenger A, Bühler M. RNA fate determination through cotranscriptional adenosine methylation and microprocessor binding. Nat Struct Mol Biol 2017; 24(7): 561–569
https://doi.org/10.1038/nsmb.3419 pmid: 28581511
18 Slobodin B, Han R, Calderone V, Vrielink JA, Loayza-Puch F, Elkon R, Agami R. Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 2017; 169(2):326–337.e12
https://doi.org/10.1016/j.cell.2017.03.03 pmid: 28388414
19 Xiang Y, Laurent B, Hsu CH, Nachtergaele S, Lu Z, Sheng W, Xu C, Chen H, Ouyang J, Wang S, Ling D, Hsu PH, Zou L, Jambhekar A, He C, Shi Y. RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 2017; 543(7646): 573–576
https://doi.org/10.1038/nature21671 pmid: 28297716
20 Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, Dai Q, Chen W, He C. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 2014; 10(2): 93–95
https://doi.org/10.1038/nchembio.1432 pmid: 24316715
21 Schwartz S, Mumbach MR, Jovanovic M, Wang T, Maciag K, Bushkin GG, Mertins P, Ter-Ovanesyan D, Habib N, Cacchiarelli D, Sanjana NE, Freinkman E, Pacold ME, Satija R, Mikkelsen TS, Hacohen N, Zhang F, Carr SA, Lander ES, Regev A. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5′ sites. Cell Reports 2014; 8(1): 284–296
https://doi.org/10.1016/j.celrep.2014.05.048 pmid: 24981863
22 Śledź P, Jinek M. Structural insights into the molecular mechanism of the m6A writer complex. eLife 2016; 5: e1843
https://doi.org/10.7554/eLife.18434 pmid: 27627798
23 Wang P, Doxtader KA, Nam Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell 2016; 63(2): 306–317
https://doi.org/10.1016/j.molcel.2016.05.041 pmid: 27373337
24 Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, Gong Z, Wang Q, Huang J, Tang C, Zou T, Yin P. Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature 2016; 534(7608): 575–578
https://doi.org/10.1038/nature18298 pmid: 27281194
25 Agarwala SD, Blitzblau HG, Hochwagen A, Fink GR. RNA methylation by the MIS complex regulates a cell fate decision in yeast. PLoS Genet 2012; 8(6): e1002732
https://doi.org/10.1371/journal.pgen.1002732 pmid: 22685417
26 Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, Zhao X, Li A, Yang Y, Dahal U, Lou XM, Liu X, Huang J, Yuan WP, Zhu XF, Cheng T, Zhao YL, Wang X, Rendtlew Danielsen JM, Liu F, Yang YG. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 2014; 24(2): 177–189
https://doi.org/10.1038/cr.2014.3 pmid: 24407421
27 Hilfiker A, Amrein H, Dübendorfer A, Schneiter R, Nöthiger R. The gene virilizer is required for female-specific splicing controlled by Sxl, the master gene for sexual development in Drosophila. Development 1995; 121(12): 4017–4026
pmid: 8575302
28 Granadino B, Penalva LO, Sánchez L. The gene fl(2)d is needed for the sex-specific splicing of transformer pre-mRNA but not for double-sex pre-mRNA in Drosophila melanogaster. Mol Gen Genet 1996; 253(1-2): 26–31
https://doi.org/10.1007/s004380050292 pmid: 9003283
29 Patil DP, Chen CK, Pickering BF, Chow A, Jackson C, Guttman M, Jaffrey SR. m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature 2016; 537(7620): 369–373
https://doi.org/10.1038/nature19342 pmid: 27602518
30 Pendleton KE, Chen B, Liu K, Hunter OV, Xie Y, Tu BP, Conrad NK. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 2017; 169(5): 824–835.e14
https://doi.org/10.1016/j.cell.2017.05.003 pmid: 28525753
31 Jia G, Yang CG, Yang S, Jian X, Yi C, Zhou Z, He C. Oxidative demethylation of 3-methylthymine and 3-methyluracil in single-stranded DNA and RNA by mouse and human FTO. FEBS Lett 2008; 582(23-24): 3313–3319
https://doi.org/10.1016/j.febslet.2008.08.019 pmid: 18775698
32 Dina C, Meyre D, Gallina S, Durand E, Körner A, Jacobson P, Carlsson LM, Kiess W, Vatin V, Lecoeur C, Delplanque J, Vaillant E, Pattou F, Ruiz J, Weill J, Levy-Marchal C, Horber F, Potoczna N, Hercberg S, Le Stunff C, Bougnères P, Kovacs P, Marre M, Balkau B, Cauchi S, Chèvre JC, Froguel P. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet 2007; 39(6): 724–726
https://doi.org/10.1038/ng2048 pmid: 17496892
33 Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, Perry JR, Elliott KS, Lango H, Rayner NW, Shields B, Harries LW, Barrett JC, Ellard S, Groves CJ, Knight B, Patch AM, Ness AR, Ebrahim S, Lawlor DA, Ring SM, Ben-Shlomo Y, Jarvelin MR, Sovio U, Bennett AJ, Melzer D, Ferrucci L, Loos RJ, Barroso I, Wareham NJ, Karpe F, Owen KR, Cardon LR, Walker M, Hitman GA, Palmer CN, Doney AS, Morris AD, Smith GD, Hattersley AT, McCarthy MI. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 2007; 316(5826): 889–894
https://doi.org/10.1126/science.1141634 pmid: 17434869
34 Scuteri A, Sanna S, Chen WM, Uda M, Albai G, Strait J, Najjar S, Nagaraja R, Orrú M, Usala G, Dei M, Lai S, Maschio A, Busonero F, Mulas A, Ehret GB, Fink AA, Weder AB, Cooper RS, Galan P, Chakravarti A, Schlessinger D, Cao A, Lakatta E, Abecasis GR. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet 2007; 3(7): e115
https://doi.org/10.1371/journal.pgen.0030115 pmid: 17658951
35 Li Z, Weng H, Su R, Weng X, Zuo Z, Li C, Huang H, Nachtergaele S, Dong L, Hu C, Qin X, Tang L, Wang Y, Hong GM, Huang H, Wang X, Chen P, Gurbuxani S, Arnovitz S, Li Y, Li S, Strong J, Neilly MB, Larson RA, Jiang X, Zhang P, Jin J, He C, Chen J. FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell 2017; 31(1): 127–141
https://doi.org/10.1016/j.ccell.2016.11.017 pmid: 28017614
36 Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, Deng X, Wang Y, Weng X, Hu C, Yu M, Skibbe J, Dai Q, Zou D, Wu T, Yu K, Weng H, Huang H, Ferchen K, Qin X, Zhang B, Qi J, Sasaki AT, Plas DR, Bradner JE, Wei M, Marcucci G, Jiang X, Mulloy JC, Jin J, He C, Chen J. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell 2018; 172(1-2): 90–105.e23
https://doi.org/10.1016/j.cell.2017.11.031 pmid: 29249359
37 Hess ME, Hess S, Meyer KD, Verhagen LA, Koch L, Brönneke HS, Dietrich MO, Jordan SD, Saletore Y, Elemento O, Belgardt BF, Franz T, Horvath TL, Rüther U, Jaffrey SR, Kloppenburg P, Brüning JC. The fat mass and obesity associated gene (Fto) regulates activity of the dopaminergic midbrain circuitry. Nat Neurosci 2013; 16(8): 1042–1048
https://doi.org/10.1038/nn.3449 pmid: 23817550
38 Mauer J, Luo X, Blanjoie A, Jiao X, Grozhik AV, Patil DP, Linder B, Pickering BF, Vasseur JJ, Chen Q, Gross SS, Elemento O, Debart F, Kiledjian M, Jaffrey SR. Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 2017; 541(7637): 371–375
https://doi.org/10.1038/nature21022 pmid: 28002401
39 Zhao B, Nachtergaele S, Roundtree IA, He C. Our views of dynamic N6-methyladenosine RNA methylation. RNA 2018; 24(3): 268–272
https://doi.org/10.1261/rna.064295.117 pmid: 29222116
40 Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, Vågbø CB, Shi Y, Wang WL, Song SH, Lu Z, Bosmans RP, Dai Q, Hao YJ, Yang X, Zhao WM, Tong WM, Wang XJ, Bogdan F, Furu K, Fu Y, Jia G, Zhao X, Liu J, Krokan HE, Klungland A, Yang YG, He C. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2013; 49(1): 18–29
https://doi.org/10.1016/j.molcel.2012.10.015 pmid: 23177736
41 Linder B, Grozhik AV, Olarerin-George AO, Meydan C, Mason CE, Jaffrey SR. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods 2015; 12(8): 767–772
https://doi.org/10.1038/nmeth.3453 pmid: 26121403
42 Zhang S, Zhao BS, Zhou A, Lin K, Zheng S, Lu Z, Chen Y, Sulman EP, Xie K, Bogler O, Majumder S, He C, Huang S. m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell, 2017; 31(4):591–606.e6
https://doi.org/10.1016/j.ccell.2017.02.013 pmid: 28344040
43 Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, Pestova TV, Qian SB, Jaffrey SR. 5′ UTR m6A promotes cap-independent translation. Cell 2015; 163(4): 999–1010
https://doi.org/10.1016/j.cell.2015.10.012 pmid: 26593424
44 Zhou J, Wan J, Gao X, Zhang X, Jaffrey SR, Qian SB. Dynamic m6A mRNA methylation directs translational control of heat shock response. Nature 2015; 526(7574): 591–594
https://doi.org/10.1038/nature15377 pmid: 26458103
45 Luo S, Tong L. Molecular basis for the recognition of methylated adenines in RNA by the eukaryotic YTH domain. Proc Natl Acad Sci USA 2014; 111(38): 13834–13839
https://doi.org/10.1073/pnas.1412742111 pmid: 25201973
46 Theler D, Dominguez C, Blatter M, Boudet J, Allain FH. Solution structure of the YTH domain in complex with N6-methyladenosine RNA: a reader of methylated RNA. Nucleic Acids Res 2014; 42(22): 13911–13919
https://doi.org/10.1093/nar/gku1116 pmid: 25389274
47 Xu C, Wang X, Liu K, Roundtree IA, Tempel W, Li Y, Lu Z, He C, Min J. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol 2014; 10(11): 927–929
https://doi.org/10.1038/nchembio.1654 pmid: 25242552
48 König J, Zarnack K, Rot G, Curk T, Kayikci M, Zupan B, Turner DJ, Luscombe NM, Ule J. iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat Struct Mol Biol 2010; 17(7): 909–915
https://doi.org/10.1038/nsmb.1838 pmid: 20601959
49 Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H, He C. N6-methyladenosine modulates messenger RNA translation efficiency. Cell 2015; 161(6): 1388–1399
https://doi.org/10.1016/j.cell.2015.05.014 pmid: 26046440
50 Kennedy EM, Bogerd HP, Kornepati AV, Kang D, Ghoshal D, Marshall JB, Poling BC, Tsai K, Gokhale NS, Horner SM, Cullen BR. Posttranscriptional m6A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe 2016; 19(5): 675–685
https://doi.org/10.1016/j.chom.2016.04.002 pmid: 27117054
51 Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J, Wu L. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun 2016; 7: 12626
https://doi.org/10.1038/ncomms12626 pmid: 27558897
52 Gokhale NS, McIntyre ABR, McFadden MJ, Roder AE, Kennedy EM, Gandara JA, Hopcraft SE, Quicke KM, Vazquez C, Willer J, Ilkayeva OR, Law BA, Holley CL, Garcia-Blanco MA, Evans MJ, Suthar MS, Bradrick SS, Mason CE, Horner SM. N6-methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe 2016; 20(5): 654–665
https://doi.org/10.1016/j.chom.2016.09.015 pmid: 27773535
53 Lichinchi G, Zhao BS, Wu Y, Lu Z, Qin Y, He C, Rana TM. Dynamics of human and viral RNA methylation during Zika virus infection. Cell Host Microbe 2016; 20(5): 666–673
https://doi.org/10.1016/j.chom.2016.10.002 pmid: 27773536
54 Tirumuru N, Zhao BS, Lu W, Lu Z, He C, Wu L. N6-methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag protein expression. eLife 2016; 5: e15528
https://doi.org/10.7554/eLife.15528 pmid: 27371828
55 Tanabe A, Tanikawa K, Tsunetomi M, Takai K, Ikeda H, Konno J, Torigoe T, Maeda H, Kutomi G, Okita K, Mori M, Sahara H. RNA helicase YTHDC2 promotes cancer metastasis via the enhancement of the efficiency by which HIF-1α mRNA is translated. Cancer Lett 2016; 376(1): 34–42
https://doi.org/10.1016/j.canlet.2016.02.022 pmid: 26996300
56 Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m6A-dependent nuclear RNA processing events. Cell 2015; 162(6): 1299–1308
https://doi.org/10.1016/j.cell.2015.08.011 pmid: 26321680
57 Spitale RC, Flynn RA, Zhang QC, Crisalli P, Lee B, Jung JW, Kuchelmeister HY, Batista PJ, Torre EA, Kool ET, Chang HY. Structural imprints in vivo decode RNA regulatory mechanisms. Nature 2015; 519(7544): 486–490
https://doi.org/10.1038/nature14263 pmid: 25799993
58 Degrauwe N, Suvà ML, Janiszewska M, Riggi N, Stamenkovic I. IMPs: an RNA-binding protein family that provides a link between stem cell maintenance in normal development and cancer. Genes Dev 2016; 30(22): 2459–2474
https://doi.org/10.1101/gad.287540.116 pmid: 27940961
59 Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, Hu YC, Hüttelmaier S, Skibbe JR, Su R, Deng X, Dong L, Sun M, Li C, Nachtergaele S, Wang Y, Hu C, Ferchen K, Greis KD, Jiang X, Wei M, Qu L, Guan JL, He C, Yang J, Chen J. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 2018; 20(3): 285–295
https://doi.org/10.1038/s41556-018-0045-z pmid: 29476152
60 Sommer S, Lavi U, Darnell JE Jr. The absolute frequency of labeled N6-methyladenosine in HeLa cell messenger RNA decreases with label time. J Mol Biol 1978; 124(3): 487–499
https://doi.org/10.1016/0022-2836(78)90183-3 pmid: 712844
61 Li HB, Tong J, Zhu S, Batista PJ, Duffy EE, Zhao J, Bailis W, Cao G, Kroehling L, Chen Y, Wang G, Broughton JP, Chen YG, Kluger Y, Simon MD, Chang HY, Yin Z, Flavell RA. m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 2017; 548(7667): 338–342
https://doi.org/10.1038/nature23450 pmid: 28792938
62 Tong J, Cao G, Zhang T, Sefik E, Amezcua Vesely MC, Broughton JP, Zhu S, Li H, Li B, Chen L, Chang HY, Su B, Flavell RA, Li HB. m6A mRNA methylation sustains Treg suppressive functions. Cell Res 2018; 28(2): 253–256
https://doi.org/10.1038/cr.2018.7 pmid: 29303144
63 Liu N, Pan T. Probing RNA modification status at single-nucleotide resolution in total RNA. Methods Enzymol 2015; 560: 149–159
https://doi.org/10.1016/bs.mie.2015.03.005 pmid: 26253970
64 Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, Sun HY, Li A, Ping XL, Lai WY, Wang X, Ma HL, Huang CM, Yang Y, Huang N, Jiang GB, Wang HL, Zhou Q, Wang XJ, Zhao YL, Yang YG. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol Cell 2016; 61(4): 507–519
https://doi.org/10.1016/j.molcel.2016.01.012 pmid: 26876937
65 Haussmann IU, Bodi Z, Sanchez-Moran E, Mongan NP, Archer N, Fray RG, Soller M. m6A potentiates Sxl alternative pre-mRNA splicing for robust Drosophila sex determination. Nature 2016; 540(7632): 301–304
https://doi.org/10.1038/nature20577 pmid: 27919081
66 Lence T, Akhtar J, Bayer M, Schmid K, Spindler L, Ho CH, Kreim N, Andrade-Navarro MA, Poeck B, Helm M, Roignant JY. m6A modulates neuronal functions and sex determination in Drosophila. Nature 2016; 540(7632): 242–247
https://doi.org/10.1038/nature20568 pmid: 27919077
67 Kan L, Grozhik AV, Vedanayagam J, Patil DP, Pang N, Lim KS, Huang YC, Joseph B, Lin CJ, Despic V, Guo J, Yan D, Kondo S, Deng WM, Dedon PC, Jaffrey SR, Lai EC. The m6A pathway facilitates sex determination in Drosophila. Nat Commun 2017; 8: 15737
https://doi.org/10.1038/ncomms15737 pmid: 28675155
68 Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, Chou T, Chow A, Saletore Y, MacKay M, Schulman J, Famulare C, Patel M, Klimek VM, Garrett-Bakelman FE, Melnick A, Carroll M, Mason CE, Jaffrey SR, Kharas MG. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 2017; 23(11): 1369–1376
https://doi.org/10.1038/nm.4416 pmid: 28920958
69 Barbieri I, Tzelepis K, Pandolfini L, Shi J, Millán-Zambrano G, Robson SC, Aspris D, Migliori V, Bannister AJ, Han N, De Braekeleer E, Ponstingl H, Hendrick A, Vakoc CR, Vassiliou GS, Kouzarides T. Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature 2017; 552(7683): 126–131
https://doi.org/10.1038/nature24678 pmid: 29186125
70 Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee CC JM, Wong IO, Ng CM, Wong . RNA N6-methyladenosine methyltransferase METTL3 promotes liver cancer progression through YTHDF2 dependent post-transcriptional silencing of SOCS2. Hepatology 2018; 67(6):2254–2270
https://doi.org/10.1002/hep.29683 pmid: 29171881
71 Cui Q, Shi H, Ye P, Li L, Qu Q, Sun G, Sun G, Lu Z, Huang Y, Yang CG, Riggs AD, He C, Shi Y. m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Reports 2017; 18(11): 2622–2634
https://doi.org/10.1016/j.celrep.2017.02.059 pmid: 28297667
72 Zhao BS, Wang X, Beadell AV, Lu Z, Shi H, Kuuspalu A, Ho RK, He C. m6A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature 2017; 542(7642): 475–478
https://doi.org/10.1038/nature21355 pmid: 28192787
[1] Yong Fan, Yan Geng, Lin Shen, Zhuoli Zhang. Advances on immune-related adverse events associated with immune checkpoint inhibitors[J]. Front. Med., 2021, 15(1): 33-42.
[2] Solmaz Ohadian Moghadam, Seyed Ali Momeni. Human microbiome and prostate cancer development: current insights into the prevention and treatment[J]. Front. Med., 2021, 15(1): 11-32.
[3] Hongnan Mo, Binghe Xu. Progress in systemic therapy for triple-negative breast cancer[J]. Front. Med., 2021, 15(1): 1-10.
[4] Jiahui Xu, Qianqian Wang, Elaine Lai Han Leung, Ying Li, Xingxing Fan, Qibiao Wu, Xiaojun Yao, Liang Liu. Compound C620-0696, a new potent inhibitor targeting BPTF, the chromatin-remodeling factor in non-small-cell lung cancer[J]. Front. Med., 2020, 14(1): 60-67.
[5] Jiajia Hu, Wenbin Shen, Qian Qu, Xiaochun Fei, Ying Miao, Xinyun Huang, Jiajun Liu, Yingli Wu, Biao Li. NES1/KLK10 and hNIS gene therapy enhanced iodine-131 internal radiation in PC3 proliferation inhibition[J]. Front. Med., 2019, 13(6): 646-657.
[6] Rui Zhou, Yuanshu Liu, Wenjun Huang, Xitong Dang. Potential functions of esophageal cancer-related gene-4 in the cardiovascular system[J]. Front. Med., 2019, 13(6): 639-645.
[7] Hong Zhang, Ying Chang, Qingqing Zheng, Rong Zhang, Cheng Hu, Weiping Jia. Altered intestinal microbiota associated with colorectal cancer[J]. Front. Med., 2019, 13(4): 461-470.
[8] Yumeng Wang, Guiling Li. PD-1/PD-L1 blockade in cervical cancer: current studies and perspectives[J]. Front. Med., 2019, 13(4): 438-450.
[9] Nikolay V. Tsygan, Alexandr P. Trashkov, Igor V. Litvinenko, Viktoriya A. Yakovleva, Alexandr V. Ryabtsev, Andrey G. Vasiliev, Leonid P. Churilov. Autoimmunity in acute ischemic stroke and the role of blood--brain barrier: the dark side or the light one?[J]. Front. Med., 2019, 13(4): 420-426.
[10] Tao Wang, Florent Chuffart, Ekaterina Bourova-Flin, Jin Wang, Jianqing Mi, Sophie Rousseaux, Saadi Khochbin. Histone variants: critical determinants in tumour heterogeneity[J]. Front. Med., 2019, 13(3): 289-297.
[11] Qiongna Dong, Bizhi Shi, Min Zhou, Huiping Gao, Xiaoying Luo, Zonghai Li, Hua Jiang. Growth suppression of colorectal cancer expressing S492R EGFR by monoclonal antibody CH12[J]. Front. Med., 2019, 13(1): 83-93.
[12] Synat Kang, Yanyan Li, Yifeng Bao, Yi Li. High-affinity T cell receptors redirect cytokine-activated T cells (CAT) to kill cancer cells[J]. Front. Med., 2019, 13(1): 69-82.
[13] Zhao Zhang, Jun Jiang, Xiaodong Wu, Mengyao Zhang, Dan Luo, Renyu Zhang, Shiyou Li, Youwen He, Huijie Bian, Zhinan Chen. Chimeric antigen receptor T cell targeting EGFRvIII for metastatic lung cancer therapy[J]. Front. Med., 2019, 13(1): 57-68.
[14] Chenfei Zhou, Jun Zhang. Immunotherapy-based combination strategies for treatment of gastrointestinal cancers: current status and future prospects[J]. Front. Med., 2019, 13(1): 12-23.
[15] Yinlong Zhang, Guangna Liu, Jingyan Wei, Guangjun Nie. Platelet membrane-based and tumor-associated platelet- targeted drug delivery systems for cancer therapy[J]. Front. Med., 2018, 12(6): 667-677.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed