[1] ROUNDTREE I A,EVANS M E,PAN T,et al. Dynamic RNA modifications in gene expression regulation[J]. Cell,2017,169(7):1187-1200. [2] ZHAO B S,ROUNDTREE I A,HE C.Post-transcriptional gene regulation by mRNA modifications[J]. Nat Rev Mol Cell Biol, 2017,18(1):31-42. [3] FU Y,DOMINISSINI D,RECHAVI G,et al. Gene expression regulation mediated through reversible m6A RNA methylation[J]. Nat Rev Genet,2014,15(5):293-306. [4] LIU J,YUE Y, HAN D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation[J]. Nat Chem Biol, 2014,10(2):93-95. [5] JIA G, FU Y, ZHAO X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO[J]. Nat Chem Biol,2011,7(12):885-887. [6] ZHENG G,DAHL J A,NIU Y,et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility[J]. Mol Cell,2013,49(1):18-29. [7] WANG X,LU Z,GOMEZ A,et al. N6-methyladenosine-dependent regulation of messenger RNA stability[J]. Nature, 2014,505(7481):117-120. [8] WANG X,ZHAO B S,ROUNDTREE I A,et al. N6-methyladenosine Modulates Messenger RNA Translation Efficiency[J]. Cell,2015,161(6):1388-1399. [9] TAKABA H, TAKAYANAGI H.The mechanisms of T cell selection in the thymus[J]. Trends Immunol,2017,38(11):805-816. [10] LI H B,TONG J,ZHU S,et al. m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways[J]. Nature,2017,548(7667):338-342. [11] CROTTY S.T Follicular helper cell biology: A decade of discovery and diseases[J]. Immunity,2019,50(5):1132-1148. [12] YAO Y,YANG Y,GUO W,et al. METTL3-dependent m6A modification programs T follicular helper cell differentiation[J]. Nat Commun,2021,12(1):1333. [13] TONG J,CAO G,ZHANG T,et al. m6A mRNA methylation sustains Treg suppressive functions[J]. Cell Res,2018,28(2):253-256. [14] LIU Z,WANG T,SHE Y,et al. N6-methyladenosine-modified circIGF2BP3 inhibits CD8+ T-cell responses to facilitate tumor immune evasion by promoting the deubiquitination of PD-L1 in non-small cell lung cancer[J]. Mol Cancer,2021,20(1):105. [15] DONG L,CHEN C,ZHANG Y,et al. The loss of RNA N6-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8+ T cell dysfunction and tumor growth[J]. Cancer Cell,2021,39(7):945-957. [16] LU T X, ZHENG Z, ZHANG L, et al. A new model of spontaneous colitis in mice induced by deletion of an RNA m6A methyltransferase component METTL14 in T cells[J]. Cell Mol Gastroenterol Hepatol,2020,10(4):747-761. [17] ZHOU J,ZHANG X,HU J,et al. m6A demethylase ALKBH5 controls CD4+ T cell pathogenicity and promotes autoimmunity[J]. Sci Adv,2021,7(25):eabg0470. [18] DING C, XU H, YU Z, et al. RNA m6A demethylase ALKBH5 regulates the development of γδ T cells[J]. Proc Natl Acad Sci U S A, 2022,119(33):e2203318119. [19] HUANG H,ZHANG G,RUAN G X,et al. Mettl14-Mediated m6A modification is essential for germinal center B cell response[J]. J Immunol, 2022,208(8):1924-1936. [20] ZHENG Z, ZHANG L, CUI X L, et al. Control of early B cell development by the RNA N6-Methyladenosine methylation[J]. Cell Rep,2020,31(13):107819. [21] KANG X, CHEN S, PAN L, et al. Deletion of mettl3 at the pro-B stage marginally affects B cell development and profibrogenic activity of B cells in liver fibrosis[J]. J Immunol Res,2022(2022):8118577. [22] NOY R,POLLARD J W.Tumor-associated macrophages: from mechanisms to therapy[J]. Immunity,2014,41(1):49-61. [23] TONG J,WANG X,LIU Y,et al. Pooled CRISPR screening identifies m6A as a positive regulator of macrophage activation[J]. Sci Adv,2021,7(18):eabd4742. [24] WANG X,JI Y,FENG P,et al. The m6A reader IGF2BP2 regulates macrophage phenotypic activation and inflammatory diseases by stabilizing TSC1 and PPARγ[J]. Adv Sci (Weinh),2021,8(13):2100209. [25] GUO M,YAN R,JI Q,et al. IFN regulatory Factor-1 induced macrophage pyroptosis by modulating m6A modification of circ_0029589 in patients with acute coronary syndrome[J]. Int Immunopharmacol,2020(86):106800. [26] SUN Z, CHEN W, WANG Z, et al. Matr3 reshapes m6A modification complex to alleviate macrophage inflammation during atherosclerosis[J]. Clin Immunol,2022(245):109176. [27] ZHOU K, CHENG T, ZHAN J, et al. Targeting tumor-associated macrophages in the tumor microenvironment[J]. Oncol Lett,2020,20(5):234. [28] LIU Y,LIU Z,TANG H,et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 facilitates M1 macrophage polarization through the methylation of STAT1 mRNA[J]. Am J Physiol Cell Physiol, 2019,317(4):C762-C775. [29] YIN H,ZHANG X,YANG P,et al. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming[J]. Nat Commun,2021,12(1):1394. [30] SHI B,LIU W W,YANG K,et al. The role, mechanism, and application of RNA methyltransferase METTL14 in gastrointestinal cancer[J]. Mol Cancer,2022,21(1):163. [31] LIU Y,SHI M,HE X,et al. LncRNA-PACERR induces pro-tumour macrophages via interacting with miR-671-3p and m6A-reader IGF2BP2 in pancreatic ductal adenocarcinoma[J]. J Hematol Oncol, 2022,15(1):52. [32] MORVAN M G, LANIER L L.NK cells and cancer: you can teach innate cells new tricks[J]. Nat Rev Cancer,2016,16(1):7-19. [33] SPITS H, BERNINK J H, LANIER L. NK cells and type 1 innate lymphoid cells: partners in host defense[J]. Nat Immunol,2016,17(7):758-764. [34] MA S,YAN J,BARR T, et al. The RNA m6A reader YTHDF2 controls NK cell antitumor and antiviral immunity[J]. J Exp Med,2021,218(8):e20210279. [35] SONG H,SONG J,CHENG M,et al. METTL3-mediated m6A RNA methylation promotes the anti-tumour immunity of natural killer cells[J]. Nat Commun,2021,12(1):5522. [36] STEINMAN R M,BANCHEREAU J.Taking dendritic cells into medicine[J]. Nature,2007,449(7161):419-426. [37] WANG H,HU X,HUANG M,et al. Mettl3-mediated mRNA m6A methylation promotes dendritic cell activation[J]. Nat Commun,2019,10(1):1898. [38] WU H, XU Z, WANG Z,et al. Dendritic cells with METTL3 gene knockdown exhibit immature properties and prolong allograft survival[J]. Genes Immun,2020,21(3):193-202. [39] HAN D,LIU J,CHEN C,et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells[J]. Nature,2019,566(7743):270-274. [40] BINNEWIES M, ROBERTS E W, KERSTEN K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy[J]. Nat Med,2018,24(5):541-550. [41] TANG H,QIAO J,FU Y X.Immunotherapy and tumor microenvironment[J]. Cancer Lett, 2016, 370(1):85-90. [42] LU M, ZHANG Z, XUE M, et al. N6-methyladenosine modification enables viral RNA to escape recognition by RNA sensor RIG-I[J]. Nat Microbiol,2020,5(4):584-598. [43] SHULMAN Z,STERN-GINOSSAR N.The RNA modification N6-methyladenosine as a novel regulator of the immune system[J]. Nat Immunol,2020,21(5):501-512. [44] TSIAMBAS E,CHRYSOVERGIS A,PAPANIKOLAOU V, et al. Impact of Ribosome Activity on SARS-CoV-2 LNP-Based mRNA Vaccines[J]. Front Mol Biosci,2021(8):654866. [45] LAN H,LIU Y,LIU J,et al. Tumor-associated macrophages promote oxaliplatin resistance via METTL3-mediated m6A of TRAF5 and necroptosis in colorectal cancer[J]. Mol Pharm,2021,18(3):1026-1037. [46] SHEN S, YAN J, ZHANG Y,et al. N6-methyladenosine (m6A)-mediated messenger RNA signatures and the tumor immune microenvironment can predict the prognosis of hepatocellular carcinoma[J]. Ann Transl Med,2021,9(1):59. [47] LUO Y,SUN Y,LI L,et al. METTL3 may regulate testicular germ cell tumors through EMT and immune pathways[J]. Cell Transplant, 2020(29):963689720946653. [48] WANG L, HUI H, AGRAWAL K,et al. m6 A RNA methyltransferases METTL3/14 regulate immune responses to anti-PD-1 therapy[J]. EMBO J,2020,39(20):e104514. [49] NI H H,ZHANG L,HUANG H,et al. Connecting METTL3 and intratumoural CD33+ MDSCs in predicting clinical outcome in cervical cancer[J]. J Transl Med,2020,18(1):393. [50] HE X, TAN L, NI J, et al. Expression pattern of m6A regulators is significantly correlated with malignancy and antitumor immune response of breast cancer[J]. Cancer Gene Ther,2021,28(3-4):188-196. [51] YI L,WU G,GUO L,et al. Comprehensive analysis of the PD-L1 and immune infiltrates of m6A RNA methylation regulators in head and neck squamous cell carcinoma[J]. Mol Ther Nucleic Acids, 2020(21):299-314. [52] GONG P J,SHAO Y C,YANG Y,et al. Analysis of N6-Methyladenosine methyltransferase reveals METTL14 and ZC3H13 as tumor suppressor genes in breast cancer[J]. Front Oncol,2020(10):578963. [53] LIU Y,LIANG G,XU H,et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance[J]. Cell Metab,2021,33(6):1221-1233.e11. [54] YANKOVA E,BLACKABY W,ALBERTELLA M,et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia[J]. Nature,2021,593(7860):597-601. [55] CHEN B,YE F,YU L,et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor[J]. J Am Chem Soc,2012,134(43):17963-17971. [56] HUANG Y, YAN J, LI Q, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5[J]. Nucleic Acids Res,2015,43(1):373-384. [57] WANG R, HAN Z, LIU B, et al. Identification of natural compound radicicol as a potent FTO inhibitor[J]. Mol Pharm,2018,15(9):4092-4098. [58] HE W, ZHOU B, LIU W, et al. Identification of a novel small-molecule binding site of the fat mass and obesity associated protein (FTO)[J]. J Med Chem,2015,58(18):7341-7348. [59] PENG S, XIAO W, JU D, et al. Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1[J]. Sci Transl Med,2019,11(488):eaau7116. [60] SU R,DONG L,LI C,et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling[J]. Cell,2018,172(1-2):90-105.e23. [61] HUANG Y,SU R,SHENG Y,et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia[J]. Cancer Cell,2019,35(4):677-691.e10. [62] SU R, DONG L, LI Y, et al. Targeting FTO suppresses cancer stem cell maintenance and immune evasion[J]. Cancer Cell,2020,38(1):79-96.e11. [63] HAN X,WANG N,LI J,et al. Identification of nafamostat mesilate as an inhibitor of the fat mass and obesity-associated protein (FTO) demethylase activity[J]. Chem Biol Interact,2019(297):80-84. [64] XIE L J,YANG X T,WANG R L, et al. Identification of flavin mononucleotide as a cell-Active artificial N6 -methyladenosine RNA demethylase[J]. Angew Chem Int Ed Engl,2019,58(15):5028-5032. [65] KZHYSHKOWSKA J,LARIONOVA I,LIU T.YKL-39 as a potential new target for anti-Angiogenic therapy in cancer[J]. Front Immunol, 2020(10):2930. [66] WANG W,MARINIS J M,BEAL A M,et al. RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer[J]. Cancer Cell,2018,34(5):757-774.e7. |