Posted on Authorea 9 Nov 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160495480.00891340/v1 | This a preprint and has not been peer reviewed. Data may be preliminary. ABSTRACT title: Running [email protected] e-mail M.E. or M.B. to *Correspondence 6 Spain. Catalonia, celona, 5 Spain. Catalonia, Barcelona, Badalona, 4 (IJC), Institute Research Leukaemia reras 3 Spain. Catalonia, Barcelona, Spain. Catalonia, 2 Barcelona, Badalona, Institute, Research kaemia 1 in modifications RNA target Mar´ıa Berdasco that Compounds Treatment epitranscriptome: and “druggable” Prevention a Cancer Towards in Avenues New on section Themed benefit. clinical for targeted-RNA of an challenges ARTICLE potential provide principal the REVIEW and the increase cancer discuss to development will in mod- we drug dysregulation Finally, and RNA their biology principal epitranscriptome. of chemical the the evidences of describe target scientific field will to the latest we discovery in the drug review, aberrant state-of-the-art this summarize of the In discovery mRNA), offers of the intervention. on epitrancriptomics overview by molecule whether focus small- see fueled a to by been (with now targeted has post-transcripcional is ifications be of interest challenge to map scientific The mechanims a the cancer. tunable to However, mainly led a diseases, have develoment. human which and with tech- modifications fate the associated RNA by epitranscriptomes quantify boosted normal and is characterize in field to The marks decade RNA. RNA of last modifications the biochemical of studies efforts that nical area emerging exciting an is Epitranscriptomics Abstract 2020 9, November 1 Berdasco Maria cancer target in that modifications Compounds RNA epitranscriptome: “druggable” a Towards nttc´ aaaad eec sui vnct IRA,Breoa aaoi,Spain. Catalonia, Barcelona, Avan¸cats (ICREA), Estudis Bar- i Recerca Barcelona, de of Instituci´o Catalana University Sciences, Health and Medicine of School Department, Sciences Physiological Spain. Madrid, Car- C´ancer (CIBERONC), Investigaci´onRed Josep Biom´edica de en (PEBCL), Program Centro Biology and Leukemia (IDIBELL), and Cancer Institute Group, Research Epigenetics Biomedical Cancer Bellvitge (PEBC), Program Biology and Epigenetics Cancer Leu- Carreras Josep (PHEC), Program Hematology Clinical and Experimental Group, Therapies Epigenetic oe arrsLuamaRsac nttt (IJC) Institute Research Leukaemia Carreras Josep [email protected]; rg httre N oicto nye ncancer. in modification RNA target that Drugs 1,2,* 1 n ae Esteller Manel and n ae Esteller Manel and 1 3,4,5,6* 1 Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. ioeottfrteteteto L-uinluama(ti ta. 08.Opruiishv extended have Opportunities 2018). inhibitor al., DOT1L et the (Stein or leukaemia 2018) treat the MLL-fusion al., to line, of trial et this treatment clinical (Italiano In the amplification II/III 2020). for EZH2 Phase mu- al., pinometostat with a genetic (Coss´ıo et reached lymphoma of therapy tazemotostat non-Hodgkin presence targeted inhibitor refractory Panobinostat) the more EZH2 CML explore a (HMT) development refractory as epidrug in histone enzymes in routine -modifying approaches clinical of New deacetylase for tations of histone 2018). approval Similarly, treatment Popat, FDA- 2016). the and reached al., (Cavenagh et (AML), for also Diesch leukaemia have 2014; practice myeloid al., inhibitors clinical et acute (HDAC) (Prebet into (MDS), (CML) leukaemia implemented syndromes myeloid myelodysplastic chronic been and as have epigenetic such decitabine target malignancies, as that haematological such inhibitors (“ inhibitors small-molecule histones (DNMT) (“ of and modifications range DNA these (“ into The interpret tags groups activity chemical chemical 2019). de- epigenetic add these al., and with move that discovery et enzymes enzymes (Berdasco the Jones of improved include modifiers) pocket 2019; has proteins epigenetic catalytic disease al., the in affecting et targeting alterations mutations (Ganesan epigenetic compounds genetic of germline-related small-molecule deviations. knowledge with of also to Our velopment associated but subject 2018). pathologies) disorders also Esteller, cardiovascular rare is and or setting including diseases orchestrated (e.g. disorders, infectious This human diseases differenti- disorders, multiple cell 2020). and neurological with al., development and associated cancer, et normal replication are Dai guide cell alterations to during 2016; signals Epigenetic Jenuwein, stable environmental is and specific that (Allis control by post- landscape the ation and modulated epigenetic in /demethylation be an role DNA could set accepted that processes. histones an biological of has DNA-related modifications mechanisms other translational epigenetic and by expression mediated gene structure of chromatin of control The regions. lymphoma; untranslated T-cell UTR, peripheral RNA; INTRODUCTION. transfer PTCL, 1. tRNA, xenograft; S-adenosylmethionine; patient-derived SAM, PDX, RNA; RNA; non-coding cancer; ribosomal ncRNA, N6- lung rRNA, microRNA m6A, cell miRNA, RNA; non-small ; messenger acid NSCLC, mRNA, 5-methyl meclofenamic myeloma; of multiple m5C, form MM, ester ethyl N1-methyladenosine; syndrome; MA2, Carcinoma m1A, acid; 5- meclofenamic Squamous hm5C, MA, ; Neck methyladenosine; desacetylases; and protein Head histone lysine HNSCC, HDAC, methyltransferases; KMT, carcinoma; histone DNA HMT, hepatocellular transition; DNMT, cytidine; HCC, epithelial-mesenchymal hydroxylmethyl lymphoma; EMT, multiforme; T-cell carcinoma; glioblastoma cutaneous cell CTCL, GBM, squamous leukaemia; oesophageal myeloid ESCC, chronic methyltransferases; CML, regions; coding CDS, Ψ, ABBREVIATIONS can- therapy, inhibitors, cer. small-molecule A-to-I-editing, , methylation; RNA Epitranscriptomics, and will cancer we potential the in Finally, increase epitranscriptome. dysregulation to KEYWORDS development the drug their benefit. and target clinical of biology chemical to for be evidences of modifications discovery targeted-RNA field to scientific RNA of the drug mechanims in latest principal state-of-the-art challenges tunable principal the the the the a describe discuss of summarize diseases, will offers overview mRNA), human we epitrancriptomics an on with review, provide whether this focus associated see In a epitranscriptomes intervention. scientific to (with aberrant the molecule now However, small- of develoment. is by and discovery challenge fate targeted the The cell by normal cancer. have in fueled which mainly marks modifications is been RNA RNA field quantify has post-transcripcional and The characterize of interest RNA. to map of decade a last modifications to the biochemical of led studies efforts technical that the area by emerging boosted exciting an is Epitranscriptomics suordn;At-,Aeoiet-nsn;AL ct yli ekei;AA 5-azacytidine; AZA, Leukaemia; Myeloid Acute AML, -to-Inosine; A-to-I, pseudouridine; erasers” erasers” n pcfi idn oanpoen htaeal oietf and identify to able are that proteins domain binding specific and ) Gnsn 08 aea ta. 09.DAmethyltransferase DNA 2019). al., et Ganesan 2018; (Ganesan, ) 2 writers enovo de ; D,myelodysplastic MDS, ) rtista re- that proteins ”), pmttos(e.g., epimutations Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. ol euti lee hre aepiigptnil eodr tutr,adpoen N interactions modifications RNA RNA protein- structure, and DNA structure, helix secondary double potential, the base-pairing alter charge, modifications not ( altered RNA does lost. of in is methylation transmission result mark DNA epitranscriptomic a could the whilst far, occurs, degradation so effect: RNA inheritance, tural When mitotic described. show been not pathways. modifications have biological epigenetic multiple 2014). on variable whilst effects al., affecting their and ity: multi- et amplifies action by Wang that of recognized characteristic 2011; mechanisms be a al., different of compartments, can in et aspects modification resulting (Jia wider RNA manner pathways strongly same dependent with biological be context- The associated a to RNA-specie. in be seems the readers function can of ple The modifications specification stability. RNA the and on of dependent distribution, type splicing, all including transcription, expression, gene gene of manner. such regulators a time-dependent aspects: modifications, involves following and as RNA the which type addition, included cell targets high, In differences a pharmacological is a Major binding-sites. in potential modifications have target established are RNA their are not enzymes of or modifications, do activity These number epigenetic still catalytic the as readers. overview their we that general and modify Regrettably, known a to erasers is but guided 2016). writers, machinery, it enzymatic of He, far, conditions associated number and So and external considerable Roundtree modifications changing anticipated. RNA to 2016; identify be of react al., extent could to proteins the et these need of and Dominissini and picture set we function, RNA 2014; full a overview, eraser by Both al., determined or critical et dynamisms reader, (Fu and modifications. a writer, reversibility with epitranscriptome their perform proteins including and To features, of exploitation epigenetic common the share between follow development. modifications will features drug DNA epitranscriptomics divergent of in field and novel epigenetics common the by whether on reached rests plan expectation of of benefit. lot field clinical a the Today, for in targeted-RNA challenges of the EPITRANSCRIPTOME principal potential in THE the the role discuss increase 2. active will to overview an focus development we an a Finally, drug has provide (with evi- and and it efforts. cancer modifications biology latest discovery but in RNA chemical the dysregulation drug principal molecule their from the state-of-the-art effector of describe the clear evidences modifications an will scientific of is RNA as latest we the of What review, act summarize this landscape merely mRNA), involved. In complete on not are expression. a does they gene obtaining of RNA which from epitranscrip- regulation that in far of pathways is still field however, biological are the we of dences, and efforts, progression and all molecular and infancy Despite the onset its and examined. the in recently on still impact been is their has dynamism tomics addition, cancer, RNA range In biogenesis, especially ( that RNA conditions. characterized diseases, for pseudo- sites physiological human and important under 5hmC), modification be identified function to as harbour have RNA known and we are can known that Nowadays, sugar also modifications (m5C; (m1A). ribose posttranslational (hm5C; cytidine N1-methyladenosine mRNA the 5-methyl cytidine and (I), as 5-hydroxylmethyl (m6A) inosine well 5mC), including methyladenosine as as modifications, U) chemical known construct to C, to also processes T, started isomerization we (A, base have bases from decade RNA past four the during The only : is of (Boccaletto abundant it maps identified However, most first been 2017a). the the have al., affecting molecules last et them RNA the (Roundtree of Over in (tRNA) most epitranscriptomes. modifications these 2018), aberrant posttranslational of al., with 140 targeting et diseases than pharmacological human more the in years RNA for strategy the 50 intervention at possibilities an modifications new as post-translational opened of modifications have role “epitranscriptome”) the on (termed discoveries companies level recent of biotech model, volume and epigenetic The the industry Following 2020). pharmaceutical al., market. of et epigenetic-based sector Villanueva meta- the R&D 2019; and boosted Esteller, academia, cardiovascular have is and in diseases, defects Berdasco conducted brain epigenetic 2017; research revert to Li, epigenetic to diseases and able infectious (Ballestar agents from disorders therapeutic ranging bolic as pathologies, drugs other epigenetic to of extending potential the and cancer beyond messenge N mN)mdfiain n oevso hi mato eeregulation. gene on impact their envision to and modifications (mRNA) RNA r versus H EPIGENOME. THE (i) ncnrs ihtepiayrl fDAmodifications DNA of role primary the with contrast in 3 (ii) Nsdmntaemblt ewe cellular between mobility demonstrate RNAs ribosomal N rN)and (rRNA) RNA (iii) iv transfer h heritabil- The h struc- The ) Ψ ,N6- ), RNA Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. elrpormigi lrptny(hne l,21) hsfidn enocstecosakbtenthe between crosstalk the influences reinforces and regulation. finding mechanism gene This of pairing 2015). control sequence the al., in a et 2018). epitranscriptome miRNA (Chen via al., and specific pluripotency epigenome modification et into in incorporated Huang m6A reprogramming be 2017; regulate And could cell al., 2015b). m6A et al., that that (Alarc´on et (Shi suggest miRNA-binding described UTRs decay modulate 3’ to RNA in YTHDC1 sequences and IGF2BP1/2/3, m6A target 2016). YTHDF3, of process al., YTHDF1, deposition translation to et including the addition RNA Strikingly, In proteins, Xiao 2017b). the reader 2015; al., m6A supervise et al., (Roundtree splicing, YTHDC2, et and into Liu hnRNPA2B1 stability RNAs mammalian and RNA 2015a; processed 2020), of hnRNPG, regulating al., export hnRNPC, al., (Alarc´on nuclear et YTHDC1, metabolism mediate et splicing lipid pre-mRNA, also 2017a; (Heck mRNA could in on involved al., modulate cells m6A genes recognizing also et downstream stem By of could (Zhang pluripotency 2018). regulation al., induced circadian specification or et from cell 2017) (Zhong induction al., postranscriptional progenitor et neural of and (Hsu 2018b), spermatogenesis aspects stem the al., of instance, haematopoietic et For almost as translation. Li in such and involved splicing processes stability, controls is YTHDF2 mRNA modulates m6A and including why YTHDF1 cycle, readers explain life future. m6A mRNA near could of of and the readers integration regulation in layer of the gene expected new on range is a focused control wide scenario gene adds The research explain methylation anticipated to 2019a). RNA An signals al., expression. m6A epitranscriptomic et gene and and (Huang of epigenetic H3K36me3 sites control H3K36me3 the histone of with in de- between trimethylation recruiting overlapped complexity studies by that association peaks (ChIP)-sequencing sequences m6A the described immunoprecipitation genomic of codon Altogether, been Chromatin specific 70% stop 2019a). has approximately into the that al., it deposition of monstrated et Interestingly, m6A upstream (Huang influences 2012). just complex (H3K36me3) al., enrichment METTL14 Lys36 et significant regions at Meyer coding a H3 the 2012; with histone in al., mainly (UTR) manner, et regions dependent untranslated (Dominissini sequence- a 3’ in and occurs (CDS) mRNA (hnRNP) on al., ribonucleoproteins deposition et nuclear m6A Generally, (Xiao heterogeneous proteins) some YTHDC and proteins 2018), and identified, 2015a). ALKBH5 al., (YTHDF also al., et family and (Alarc´on been al., et (Huang YTH have FTO et proteins the readers (Jia IGF2BP demethylases. m6A to (FTO) 2016), 2013) belonging acids. protein al., proteins nucleic obesity-associated N-methylated m6A-binding et and modifi- demethylate included (Zheng mass this to (ALKBH5) fat , known splicing 5 dioxygenases the the mRNA cellular are homologue of of the demethylation activity the AlkB in m6A the and of as passive by 2011) regulation well removed from as the actively Apart methylation, is in the 2017). for cation involved as al., donor is et acts methyl (Pendleton METTL16 METTL14 the process (SAM), and methyltransferase. S-adenosylmethionine subunit m6A - of catalytic the snRNA levels ( the at U6 recognition group is a methyl substrate METTL3 METTL16, a for where of scaffold m6A process, addition nowadays, RNA-binding methylation the and of the interest, ( consists in research adenosine It involved intense of an modification. to position RNA led 6 characterized methods best detection the robust is of ( development ) all the of with (0.2%–0.6% mRNAs mammalian in tected N6-Methyladenosine. 3.1. DYNAMISM RNA- DISTRIBUTION, making MODIFICATIONS: for FUNCTION. RNA scenarios AND research CODING major NON-CANONICAL the discovery. introduce 3. drug will knowledge in we of target level sections, current actionable effect next the an and In the complex modifications tremendously though limited. is indirectly somewhat word” “RNA still also the is that but assume targets can we since their together, functionally, by All works directly modification read the change. how be structural influence their could also on modifications change RNA conformational in This changes 2015). al., et (Liu iue1 Figure 6mtyaeoie(6)i h otaudn nenlmdfiainde- modification internal abundant most the is (m6A) N6-methyladenosine .Temtytaseaelk MTL)MTL4htrdmris heterodimer (METTL3)–METTL14 3 methyltransferase-like The ). \ oth RAsaiiydrn tmcl ieetainand differentiation cell stem during stability mRNA soutthe ldzadJnk 06.AohrmAwie rti is protein writer m6A Another 2016). Jinek, Sled´z and ´ 4 ldzadJnk 06.Isaudnetogether abundance Its 2016). Jinek, Sled´z and ´ ieversa vice thsbe recently been has it , Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. eeo h rncito atrMcadisatvto srlvn o h ieetaino rmr human primary of differentiation the for target relevant direct is of a activation consisting as models its identified 2015). Mice and been 2006). al., Watt, Myc has et and (Frye factor It (Xing keratinocytes pathways. transcription cycle biological the division multiple of cell in gene the enrolled during as is p21 act since NSUN2 and cyclin-dependent can Physiologically, event CDK1 of m5C random regulators 5’UTR addition, a cycle at In accumulation cell not context. m5C the is sequence of the deposition CG include 5mC a Examples The inhibitor by translation. in et kinase 2017b). affected protein (Yang and al., strongly of discovered codon et is modulator been start potential (Yang ALYREF/THOC4 a the translational has mRNAs factor about at export target export known accumulates nuclear its nuclear is 5mC of of much the level regulation not of m5C mRNA, the activity in the in the m5C Specifically, m5C of 2017b). for biology al., role the uncover A to consequences. been beginning yet functional not only from have are mRNA species we in RNA overexpres-As from levels only 5mC m5C remove regard, overall that this affected enzymes In , Regrettably, 2017b). NSUN 2017b). identified. al., al., other et et any (Yang (Yang of cells do- mRNA not HeLa methylate so NOL1/NOP2/SUN but However, to the NSUN2 (DNMT2). proved of of 2 been sion/suppression methyltransferase-like members has DNA seven NSUN2 and include only (NSUN1-7), mammals far family in (NSUN) member modifications (Huang family m5C regions main RNA 3’-UTR the of and writers 5’-UTR The the in observed also 2019b). of been has al., 62-70% enrichment et an about incomplete although that CDS, to to estimated due maps is ( mainly come It ( levels discrepancies m5C, methylated treatment. methylation and highly bisulfite of low debate during had transcriptome-mapping strong sites m5C the under cytosine and is establish cytidine mRNA to in of difficulties conversion m5C technical of abundance the al., The et from 2019). (Dominissini Lusser, proposed ( and been (m5C) (Trixl 5-methylcytosine has to stress rise needed; cellular giving are also of biology types RNA could 5-Methylcytosine various in readers 3.3. m1A to m6A of response 2016b). functions of al., the mRNA the family in et in into m1A Li insights role protein to New 2016; a YTH et directly 2018). only (Dominissini bind The al., far, to et tRNA 2020). shown so (Dai and al., were cells mRNA YTHDC1 cancer et in human and modification Esteve-Puig in YTHDF1-3 both m1A 2016b; Specifically, demethylase 2017). signal. al., m1A al., m1A a et et interpret ALKBH3, Safra Li by 2017a; 2016; also al., mRNA al., et does et from activity (Cenik (Li removed its however, cells transcripts be complex, human mitochondrial-encoded TRM6-TRM61 can the the and is in mouse mRNA m1A cytosolic in of Its cover conservation writer 2017). m1A high al., known its in et only The (Cenik by non-canonical function supported facilitating proximal a is by 5’ described regulation of 2016b) devoid 2017). al., of works UTRs al., control et recent 5’ Li the at some 2016; in complex however, al., junction role -exon et elucidation, al., the (Dominissini under et of translation (Roundtree binding is expected. m1A mRNA position be m6A, of modification this could unlike initiation m1A structures that at the GC-rich, secondary/tertiary of is base to RNA interest charged role uniquely and of (about positively maps interactions The aspect m6A a protein-RNA modification An at than carrying m1A 2017). Alterations abundant interface The al., 2017a). its Watson-Crick less 2017). et the Although is al., (Safra in mRNA. m1A transcripts et in occurs that coding Safra consequences in accepted 2016; its positions information al., is and 5’-UTRs little et it very regulation controversial, (Dominissini is m1A is times) recently there was in ten Nowadays, mRNA it involved 2018). cytosolic but players al., in rRNA, key et and frequency the (Boccaletto tRNA frequency, mRNA in in its predominant exists is of also It it 2020). Kouzarides, that and determined ( (Barbieri adenosine RNAs of of position classes nitrogen-1 the at group N1-Methyladenosine. 3.2. p27 I1drn elctv eecne(age l,21) rmCdpsto nte3 UTRs 3’ the in deposition m5C or 2015), al., et (Tang senescence replicative during KIP1 > 0 ehlto)(un ta. 09) h oaino 5 oictosprimarily modifications m5C of location The 2019b). al., et (Huang methylation) 40% ieDA l ye fRAmlclscnb ehltda abn5o cytosine of 5 carbon at methylated be can molecules RNA of types all DNA, Like . h 1mtyaeoiemdfiain(1) rteadto famethyl a of addition the or (m1A), modification N1-methyladenosine The iue1 Figure < 0 ehlto) hl -0 ftestswr oeaeyor moderately were sites the of 8-10% while methylation), 20% iue1 Figure oeigdvrefntosdpnigo h N specie RNA the on depending functions diverse covering ) 5 ,wsdsrbddcdsaot rmrl ffc all affect primarily to ago decades described was ), Nsun2 ncdw xii diinldevelop- additional exhibit knockdown \ soutthe Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. o DR ntecnrlo h icda lc a enrvae Trjm ta. 2017). role al., et A (Terajima 2017). revealed CANCER. been al., for IN has et required clock EPITRANSCRIPTOME (Behm primarily DEREGULATED circadian development is the 4. system and of long brain nervous control of in central the expression editing for in higher promote promiscuous ADAR2 transcripts to a in for Dicer key has with role ADAR2 of complex prominent a editing 2013). forms a al., ADAR1 site-specific plays that et especially indicate (Ota and studies expressed, processing ADAR2 system, Additional widely miRNA and 2016). blood is ADAR1 al., ADAR1 the et Both mammals, may of (Zipeto 2016b). dsRNA In and component (Nishikura, myeloid differentiation. activity activity cellular the editing ADAR2 in lacks in and roles ADAR3 ADAR1 essential whereas of have proteins, regulator proteins negative active a catalytically same as the the act are within ADAR2 2017). other regions by and al., with each ADAR1 catalysed et associated to mostly (Porath is proximity is UTRs edition editing close 3’ A-to-I of and in class regions this adenosines intronic mammals, of sequences, In as repetitive proportion hyper-editing. against understood of for large protecting requisite be A in a sec- could role is A which 2014). transcript A 2018). hyper-editing, al., Levanon, process. is and et editing editing (Eisenberg (Porath that the A-to-I proposed revealed for of been have targets modifications type self-transcripts pro- main ond A-to-I by as the immunity animal well were innate as among of mRNAs and, study activation the or guanine comparative miRNAs of a a of parts like However, function non-coding level and/or cellular 2016b). biogenesis at the (Nishikura, repeats, interpreted alter Alu teins could is ( (e.g., editing regions Inosine editing I non-coding A-to-I 2016a). A-to to consequently, as (Nishikura, mRNAs of known pri-miRNAs) regions or also coding pre-miRNAs process from ranging a sequences, 2014). inosine, genomic al., multiple to et adenosine Schwartz 2014b; of mRNA al., tion pausing inducible et editing. through of (Carlile Adenosine-to-inosine control suggested code the 3.5. also genetic throughout translation been the mRNA has of in localization role adaptation RNA A flexible and 2014b). the al., in et Notably, (Carlile function modifications a 2019). al., suggesting and et cells, (Shaheen Barbieri brain expression human and gene 2014b; muscle of affecting regulators al., diseases as inherited role et ψ cause emerging enzymes (Carlile their PUS reinforced human snRNAs which encoding function genes specific in Mutations modifying of relevance role. by its physiological process The splicing 2020). the Kouzarides, regulating that as known Currently, well well 2017). al., is redundancy et potential It Penzo or 2014a; action al., with of et associated (Carlile mode resolved reader Their 2020). completely or organisms 2017). been eraser other (PUS1, al., yet specific mRNA in not et any on counterparts has (Penzo acting their functions those PUS9) than their including and studied enzymes, in PUS7 PUS less PUS6, of far identification PUS4, are better PUS3, (PUS) enzymes a synthases allow PUS discoveries pseudouridine Human recent by but into humans genes. in transformed 13 represented is by emerged are uridine encoded have Pseudouridylases , (ncRNAs) In pseudouridylases. RNAs as complex. known non-coding very mRNAs, is on Besides pseudouridylation with controversy with targets 2014). serious interesting al., introduce highly ( et limitations as (Schwartz pseudouridine Methodological m6A of of than abundance mapping 2017). abundant the and al., however, distribution et RNA; (Penzo the human addressed total recently in was modification frequent most mechanisms 180 molecular bond, identified. The not 2013). were al., Pseudouridine. differentiation et 3.4. cell (Hussain al., impaired differentiation et and testis (Tuorto deficiencies in others among implicated NSUN2 skeleton, also connecting immature was organization, Nsun2 cortex . cerebral 2012) impaired as such defects, ment otn n3URmN srgltdi epnet niomna inl,sc ssrmsavto in starvation serum as such signals, environmental to response in regulated is mRNA 3’UTR in content ° aertto,adbn eomto HmaadFerr´e-D’Amar´e, and ( (Hamma 2006) reformation bond and base-rotation, suordlto steioeiaino h rdn aevabekg fteglycosidic the of breakage via base uridine the of isomerization the is Pseudouridylation ψ nacstefnto ftN n RAb tblzn h N tutr as structure RNA the stabilizing by rRNA and tRNA of function the enhances \ oth dnsn emns cigo sN aiyo rtis ADAR. proteins, of family dsRNA on acting deaminase adenosine soutthe ψ ψ, ie RnaaDmsyadKte 07.Teezmlg associated enzymology The 2017). Kothe, and (Rintala-Dempsey sites u h eea osnu sthat is consensus general the but nte N oicto nmmasi h reesbedeamina- irreversible the is mammals in modification RNA Another ψ oictoshv enietfid(abeiadKouzarides, and (Barbieri identified been have modifications ψ nmN smr nla ihol e vdne of evidences few a only with unclear more is mRNA in 6 iue1 Figure ψ \ otneiigerce regions enriched editing soutan ie nmN r uhless much are mRNA in sites .At- dtn cusin occurs editing A-to-I ). ψ yacaso enzymes of class a by iue1 Figure ψ nmRNAs in ) .I sthe is It ). Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. ta. 08 ege l,21) ua noera acrcryn oso uain nMTL4 and METTL14, in mutations hotspot Reductions Liu tumorigenicity. carrying 2017; and cancer al., proliferation endometrial increased et m showed Human (Cui in methylation, 2019). On identified m6A al., in 2019b). been et reductions have al., consequently Deng 2017). complex et al., 2018; METTL14 et (Han al., (Ma METTL3- m6 et carcinoma cancer on an hepatocellular tu- bladder in functions in process potential the suppressor of metastatic miRNA126 Tumour proliferation decreased of primary to the the expression leading modulates by manner to positively decreased A-dependent genes interaction leading in DGCR8 suppressor METTL3 and METTL14- resulting tumour example, contrary, PTEN, For the or pri-miR221/222, activity. gene anti-tumoural of suppressor or modulate maturation (Alarc´on pro- mour et dually the with DGCR8 to miRNAs protein promotes able multiple microprocessor overexpression is of the METTL3 METTL3 maturation 2015b). with model, the al., (Alarc´on interacting regulating this et et processing by In Interestingly, miRNA (Lin miRNAs 2019). 2015b). on also patients al., effect of al., been cancer et an maturation has includes liver (Dahal the N-cadherin also expression of action and promotes METTL3 survival of (MMP2) increased overall mode 2 with reader METTL3 adverse its together metallopeptidase the and for cells effectors METTL3 melanoma factor EMT of in the prognosis upregulation observed the of a Moreover, Overexpression as 2019). 2019). al., used al., et has be (Lin metastasis, cells could cancer cancer YTHDF1 liver for AML in in process in m6A mRNA dysregulation. crucial Snail described of METTL3 of a also role with (EMT), been oncogenic associated transition has the also Epithelial-mesenchymal overexpression confirming been 2018). METTL14 2017), al., of al., et et role (Weng (Barbieri oncogenic effect MYC-dependent anti-tumoural A decreased METTL3. and or cells BCL2 Mechanistically, leukemic MYC, 2017). of as al., such involved transcripts et in vivo mRNAs oncogenic Vu demonstrated In of crucial 2017; firstly translation of al., to as increased et maintenance contributes in PTEN, (Barbieri overexpression the results METTL3 cells METTL3 for et that leukemic of (Barbieri essential so overexpression undifferentiated reported is cells, of widely methylation stem/progenitor maintenance been haematopoietic m6A the has of carci- among 2017). model self-renewal 2017), cell al., AML al., the renal et the et in cell (Taketo in cancer clear Vu pancreatic 2017), complex 2017; or al., METTL3-METTL14 2020) al., et of al., (Barbieri et role (Yang AML The cancer in gastric others. proposed 2020), cancer-specific al., been the et has by (Wang METTL3 determined noma for be 2019). role to al., oncogenic seems et An cancer (Zheng the in enzymes Although role m6A-related 2020). dual properties of al., Rossell´o-Tortella this suppressor targets cancer, et 2020; limited, tumour downstream in Kouzarides, still also altered and are but frequently (Barbieri evidences oncogenic is type supporting with methyltransferases tumour compatible m6A the be of on could expression depending consequences The will functional it oncology. the cancer. that in and in foreseeable focus epitran- is contents main it the altered N6-Methyladenosine research. so been of of interest, oncology evidences Alterations for major major possibilities with 4.1. the of area range summarize growth wide a will a is we up This section, infection open cancer. this viral with renewal, In associated tissue scriptome others. responses, immune among human invasion, with angiogenesis, and cell the associated differentiation, or ( In fate are cell to) it diseases. cell epitranscriptomes stem limited 2018), human deregulated including in not to al., pathways, that (but role lead revealed et cancer have epitranscriptome their Morena mainly studies the and 2017a; of pathologies, of number al., cycle expression a et abnormal life years, Roundtree that last RNA surprising 2017; regulating not Pandolfi, therefore in and is (Esteller modifications development RNA human of normal importance the Given eedn ntemArae TD2 oefrFOi h rmto frssac oimmunothera- to resistance of promotion the in for FTO the mechanism for role increases a role oncogenic and in A methylation an SOX10, m6A YTHDF2. and provide CXCR4, decreases reader studies (PDCD1), FTO m6A first PD-1 2019). the as al., the on such et cancer, genes Yang dependent in melanoma 2013; pro-tumorigenic demethylation al., of et RNA stability (Iles m6A melanoma altered in of FTO tumour-suppressor 2019) as case al., acts et the also (Deng METTL3 In pathways 2018). p38/ERK al., through et (Liu cancer mTORC2 colorectal regulator in AKT positive the of pression 6 ehlto edt erae xrsino h eaieATrgltrPLP n nrae ex- increased and PHLPP2 regulator AKT negative the of expression decreased to lead methylation A saspromdi ET3 ncdw muoecetmc euti nrae differentiation increased in result mice immunodeficient knockdown METTL3- in performed assays nvitro in tde efre nteML-3cl ie(ue l,2017). al., et (Vu line cell MOLM-13 the in performed studies al 1 Table 7 hog h eeuaino antumourogenic main of deregulation the through ) Snail D assplsm-eitdtranslation polysome-mediated causes CDS h td fmAdseuainhas dysregulation m6A of study The Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. vra acrptet ihhg SN xrsinadlwIFI xrsinehbthge vrl and overall applications. higher biomarker exhibit potential expression IGF-II has low expression and NSUN2 expression carci- NSUN2 cases, high cell cancer some with breast squamous In and patients oesophageal thyroid, cancer protein bladder, including 2020). Ovarian kidney, of Gray, types, prostate, Gain cervix, cancer and uterine progression. multiple (Chellamuthu pancreas, cancer liver, in human stomach, quantified with (ESCC), was associated noma been NSUN2 has of expression expression cancer. NSUN2 in in in content dysregulation alterations m1A 5-Methylcytosine tion, of of role Alteration pathological breast 4.3. a and anticipates ovarian study in This migration or 2019). species. proliferation Chambers, mRNA colony-stimulating cell and that of (Woo affecting abundance described cells without was transcript invasion cancer it mRNA cell Recently, the promoting increased demethylation. ALKBH3 (CSF1) tRNA by to factor mediated refer contents mainly m1A cancer of in loss dysregulation 2019). m1A cancer. of al., in in role contents et the apoptosis N1-Methyladenosine (Paris avoiding of AML AML transcript Alterations of human Tnfrsf2 4.2. subtypes and receptor expansion. several mouse their factor in in promoting necrosis potential overexpressed propagation cells tumour prediction as is stem of the well leukaemia YTHDF2 half-life results, as the 2019). initiation preliminary decreases al., disease on YTHDF2 et for based required (Liu Although is reader and 2016). HNRNPC to al., extended et metastasis-related (Tanabe is the YTHDF2 of proposed expression been 2019a). of the gain has al., highlighting to et leading abolition, (Han cancer, HIF-1 immunotherapy YTHDF1 colorectal gene metastatic in after in expression overexpressed enhanced is YTHDF1 CD8 is reader of of blockade application checkpoint cross-priming therapeutic PD-L1 potential the of and efficacy antigens therapeutic tumour et (Han of immunity cross-presentation \ neoantigen-specific the stable suppression enhanced facilitating a cells has cells YTHDF1 dendritic reader these in The whether 2019a). presentation elucidated m6A-signal. al., antigen be the to changes of the in needs independently in of result still cancer result it their effect in could not Furthermore, role exert proteins should crucial modification. effector that a readers RNA these have proteins the of readers m6A of levels reader of function expression of molecular the dysregulation the set in the in alterations a Although patterns, by cell m6A interpreted pathways. in stem is changes biological cancer signal multiple breast m6A in a the favours function mentioned, stability previously Nanog as of Finally, overexpression gain the 2016). ALKBH5 The increased, al., Similarly, is Nanog. et (Zhang factor stability 2017c). is phenotype pluripotency its al., expression the et transcript, ALKBH5 of (HIF)-1 (Zhang mRNA Interestingly, transcript factor FOXM1 glioblastoma hypoxia-inducible in diminished. in by differentiation m6A prognosis (stimulated cell of poor properties stem-cells and loss with glioblastoma enhanced of correlated is of glioblastoma is result et maintenance in (Zhang expression a the target model FOXM1 for As ALKBH5 glioblastoma crucial cells. a is self-renewal. stem-like in which glioblastoma and characterized FOXM1, in well factor expressed to been transcription highly contribute has the is that ALKBH5 ALKBH5 on demethylase targets 2017c). depends m6A al., specific strongly involved. the tumorigenesis at are of on that content role effect pathways The m6A FTO down-stream the of the sum, and increase In context an genomic tu- 2017). in the al., AML et results IDH1/2-mutant (Elkashef inhibition suppression in tu- leukaemia FOT accumulates AML This IDH1/2-mutant aberrantly in which effect mours. of suppressive R-2-hydroxyglutarate, promotion a oncometabolite have and could related differentiation it an cell contrast, is In AML FTO transcripts 2019). 2017b). mours. (ATRA)-induced mRNA al., al., RARA acid et et and all-trans-retinoic (Li (Yang ASB2 leukemogenesis of in models levels inhibition mice m6A t(15;17)/PML-RARA, to in demethylase rearrangements, leading FTO t(11q23)/MLL demonstrated mutations. with also NPM1 AML been and/or in FLT3-ITD, progression has tumour therapy promotes melanoma also FTO in anti-PD-1) (i.e., py ot ehnssivligtecnrlo RAtasaino yooa ahpis otiprat the important, Most cathepsins. lysosomal of translation mRNA of control the involving mechanisms souta α, nvivo In ftmu el both cells tumour of α- eoltrt eedn ixgns hti opttvl niie ytestructurally the by inhibited competitively is that dioxygenase dependent ketoglutarate tde sn iemdl eosrtdta oso TD1i lsia dendritic classical in YTHDF1 of loss a that demonstrated models mice using studies nvitro in and nvivo in α- n HIF-2 and 8 oeta imre oefrpeitn metastasis predicting for role biomarker potential A . α ossetwt t oei ellrdifferentia- cellular in role its with Consistent nbes acrdmtyae h mRNA the demethylated cancer breast in ) oto h ffrsfrteeuiainof elucidation the for efforts the of Most + el through cells T Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. euti utpehmndsae,icuigcne.Atrtosi DRatvt a endsrbdfor described been has glioblas- leukaemia, activity myelogenous ADAR chronic in carcinoma, Alterations hepatocellular tran- cancer, mRNAs dysregulation cancer. lung of their including including why properties explains , diseases, pairing proteins) multiple human (and base translation multiple the after in codons both altering result affecting also by but ncRNAs ADARs and of scripts significance biological cancer overall also in The is editing 2020). process inosine DROSHA- al., this to- et microprocessor Mechanistically, Adenosine- (Song the Recently, types. PUS10 with 4.5. of cell caspase-3 uncertain. interacts activity multiple still and amplifies catalytic in active is pre-miRNAs the biogenesis reciprocally it to of involved: miRNA involved PUS10 independent binds promote is early is of PUS10 to synthase the 3 that movement complex pseudouridine in caspase DGCR8 described the the mitochondria and been Whether the whilst PUS10 has 2017). to export it between al., nucleus PUS10 et loop the (Jana for feedback from activity A required exported is apoptosis is TRAIL-induced PUS10 apoptosis. caspase-3 in TRAIL-induced PUS10 2017). synthase of al., pseudouridine stages et the involving (Jana mechanism discovered mechanisms a was molecular hand, the other clarified. Nevertheless, the been receptor- On 2004). yet acid al., not retinoic have et (e.g., phenomenon (Zhao genes SRA1-associated this target complex (SRA1). underlying of pre-initiation 1 pseu- domains transcriptional activator the receptor RNA the through nuclear receptor lish cancer the steroid breast binds cells the and then ncRNA, cancer melanoma target PUS1 with breast its associated on human been activity in has douridylation inactivation PUS1 synthase p53 pseudouridine has and The function Interestingly, the translation DKC1 of 2017). of mRNA stabilization 2010). impairment al., al., the p53 Additionally, et et to aberrant (Montanaro 2015). Penzo linked with al., been 2009; et associated has al., (Penzo cancer carcinomas, been et (TERC) in hepatocellular Sieron component overexpression cancer, RNA 2006; dyskerin telomerase breast al., of been in et consequences have survival (Montanaro molecular levels overall the cancers expression poor prostate DKC1 and and cancers. progression lung, sporadic tumour cancer and for with trigger tumours potential correlated syndrome-associated a as hereditary recognized in been tRNA. been has onset or activity has (DKC1) of rRNA synthase action mainly defects pseudouridine dyskerin the of species, Altered non-mRNA of mode of most and Furthermore, control disease its now. in by to implication mediated likely up its explored is Unfortunately, partially it only contexts. understood, pathological well and being roles, iological not suppressor biogenesis tumor pseudouridylation cancer exert Despite with 2019). NSUN5, associated al., as alterations et such Pseudouridyne (Janin members, (Li tumors 4.4. activation brain familiy ERK1/2 and human NSUN by BPTF, in other inactivated regulator MMP10 chromatin Conversely, epigenetically and to being MMP3 of bind 2018a). metalloproteinase role directly ESCC. a the al., could in described of transcript et (LINC01672) study expression NMR separate NMR reader, stability, pro- the A increased lncRNA promote cancer, 5mC its oncogenic potentially 2019). bladder a of the al., in found result of et increased a authors (Chen stabilization As aberrantly dynamism the addition, 5mC is in In the methylation expression on NSUN2-dependent whose 2019). clues (YBX1), molecular al., 1 re- new et viding (HDGF), protein pathways, (Chen factor binding cancer-related growth are stabilization Y-box well-known binding we mRNAs named in heparin modifications, enhanced enriched the RNA in highly carcinoma and m1A bladder sulting is work ERK–MAPK36, human or elegant mRNAs and in An m6A m5C PI3K–AKT35 resolution including to tumorigenesis. single-nucleotide of governing at manner hypermethylation pathways modifications similar that molecular mRNA showed a the 5mC of In of identifying expression understanding at prediction NSUN2 the 2020). aimed the of from for risk al., association away an biomarker mortality et far by a mediated still (Lu higher as be activation a proposed could Neck T-cell effect been and The and have and survival HNSCC. Head expression in overall immunotherapy in NSUN2 Interestingly, to shorter upregulation response 2018). with NSUN2 al., contrast, associated et In was (Lu 2017a). (HNSCC) al., Carcinoma et Squamous (Yang survival progression-free disease . 9 . Ψ oicto ikdt acrare cancer to linked modification ψ ly oei aiu phys- various in role a plays γ ohl estab- help to ) Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. eo,w ilhglgtteeegn eut n hlegsfrteueo N-mdfiain sactionable as modifications RNA- of field. use promising the models. for this discovery. challenges experimental of drug and specific potential cancer results therapeutic for in emerging targets the found the beginnings, highlight addressing are its will research are therapy we discovery in companies Below, RNA-modifying drug biotech still of several and is benefits important, biology epitranscriptome-targeting of Most the chemical of introduction of for field successful evidences and the model the preclinical new Although Inevitabily, a opens RNA-epitranscriptomics. 2). delineates (Table epitranscriptome on clinic design the also oncology the the in in into defects cancer, discovery drugs revert drug in epigenetic-based potentially for epitranscriptome that opportunities the molecules exciting of small dysregulation of development aforementioned and the consideration into EPITRANSCRIPTOME. Taking THE TARGET TO AIM cell STRATEGIES 2017b). PHARMACOLOGICAL astrocytoma al., 5. et and (Oakes astrocyte patients of transcript in glioblastoma level relative the from ADAR3 potential the of taken to of site a contributes regulate expression Q/R Overexpression revealed ADAR3 subsequently the and in 2017a). glioblastoma at and ADAR2 ADAR2 editing al., transcripts in RNA of et specific ADAR3 inhibits has role target (Oakes lines of GMB to activity A in contribution ADAR3 editing 2013). miR-21) The and and their al., ADAR2 2015). p27, miR222 between et miR221, and al., competition (Galeano miRNAs p21 et binding (e.g., growth Skp2, (Tomaselli astrocytoma suppressors cellular as revealed tumour high-grade such inhibits and In been proteins, oncogenic and tumours. several of cycle brain phase in editing cell aggressive G1/M the ADAR1 key highly the than regulates in control ADAR2 properties described which (GBM), tumour-suppressor widely multiforme was with glioblastoma it associated or as of more editing cancers be A-to-I of the to types through seems enhancing role 2016). ADAR2 protein) suppressor al., Indeed, level tumour ribosomal et a S6 phosphorylation (Chan exerts kinase, transcript the ADAR2 p70S (podocalyxin-like) influences while PODXL mTOR, 2016), expression the (i.e., gene al., ADAR1 pathway suppressor also et signalling cancer, tumour (Dou been mTOR the gastric oncogenesis of has of In players inhibition miR-455-5p) 2015). crucial the (e.g., al., of involving oncogenic miRNAs the mechanism et and a of (Shoshan gene) by as editing CPEB1 repair melanoma proposed DNA mediated metastatic (a been ADAR1- for NEIL1 has 2016b). described as overexpression it such ADAR1 al., Mechanistically, transcripts and (Anad´on et 2016a). target tumours, al., of miR-381 (Anad´on primary frequencies et and outcome and editing amplification poor the lines gene of enhances ADAR1 cell prediction cancer, HNSCC for lung biomarker in In malignant a observed a 2013b). inducing al., been factor) et has transcription (Jiang overexpression (myeloid cells PU.1 stem embryonic of of expression reprogramming with Through correlated carrying mechanism positively CML The 2013a). ADAR1 IFN- In of 2017). al., pathway. of al., et inflammatory expression et the (Jiang protein involved the (Chen activity ADAR2 binding growth CML gene, factor with that tumour fusion of growth associated of demonstrated insulin-like is downregulation BCR-ABL inhibition authors expression of and and work, ADAR1 stabilization apoptosis described which this cell and by been In to edition has leading the et progression. ADAR2 (IGFBP7) for (Zhang ESCC 7 for necessary ESCC to role is in linked activity suppressor JAK/STAT process catalytic been tumour associated editing has a its A-to-I enzyme and contrast, , ADAR2 aberrant In I in in type involved 2017b). Interestingly, resulting also al., is 2014). expression edition, D1 al., ADAR1 A-to-I cyclin et This FLNB upregulates and (Qin with pathway, together ESCC ODC and residue 2013). editing, FLNB of decarboxylase including dependent at pathogenesis al., ornithine proposed, AZIN1- glycine the been the et 2014). has to HCC of al., (Chen in et degradation serine ADAR1 (Chan for properties the COPA of targets invasion of editing substitution A-to-I prevention A-to-I increased Additionally a the al., oncoproteins. and tumorigenesis. by to et mediated of phenotype lead Qin is stages tumorigenic transcripts 2014; effect earlier the (AZIN1) al., the 1 facilitating et in inhibitor Chan 367, fundamental antizyme is 2013a; of ADAR1 al., (HCC), editing et carcinoma Jiang hepatocellular 2013; In al., et (Chen others among 2014). melanoma, and toma nvitro in γ eei sas uhr eosrtdta over-expression that demonstrated authors assays, genetic aha ee rmtsAA1epeso n editing and expression ADAR1 promotes genes pathway 10 GRIA2. GRIA2 otipraty h eainbetween relation the importantly, Most dtn npiaytmu samples tumour primary in editing Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. namuemdlo M,soe htoa diitaino C-5rdcdbt peoeayand splenomegaly both patient-derived-xenografts reduced in SCT-15 reduced studies of was Preclinical administration AML. growth refractory oral tumour for Similarly, that therapy monocytes. showed a circulating AML, as of trial of trials number clinical model clinical the I mouse October Phase I In human a phase Therapeutics. enter on for STORM to by selected ready achieved been phases been has have preclinical results in advanced STORM companies, most inhibitors 2020, the Three METTL3 date, METTL3. To have (NYC, 2019). mainly Therapeutics to (Cully, Gotham methyltransferases, announced and RNA USA) have (MA, targeting Therapeutics USA), Accent discovery UK), CRISPR-Cas9 (Cambridge, drug by Therapeutics early STORM manipulation AML for genetic in invasion race that and showed the proliferation previously cell prevent been importance enzyme and has METTL3 the downregulate it to METTL3-METTL4 to activity. and attention guided methyltransferase technology cancers, drawn m6A in already m6A of upregulated has aberrant manipulation well-defined community is pharmacological the research the with the strengthening together chemical cancers, modification of the RNA with universal for associated most point the patterns starting is given m6A been a that has as Considering data methyltrans- preclinical used of strong observation. be family no however, this fold could methyltransferases; support Rossmann RNA inhibitors to of features, the KMT development unique to drug that and and belong portends design specific methyltransferases similarity has RNA methyltransferases This m6A RNA in- ferases. and specific EZH2 Esteller, m5C by of inhibitor, and DOT1L, followed structure (Berdasco DOT1L therapy, like treatments protein leukaemia The lymphoma for the inhibitors. B-Cell trials Although for KMT clinical 2018). approved as enter tazemetostat) to developed and of inhibitor (GSK2816126 advantage been KMT hibitors taking have first by site pocket was the selective binding activity binding was are its Pimenostat, KMT that for SAM-mimetics SAM inhibit the with potent to in several competes attempt reversibly differences Thereafter, first sinefungin 2015a). The product al., natural 2019). et the (Kaniskan al., that domain et discovery substrate-binding the (Ganesan a on targeted based and successfully mandatory pocket been SAM-binding a have contain is that also (KMT) response methyltransferases therapy lysine cancer cancer. Histone in guides in bottleneck that medicine major precision a mechanisms in improve is molecular sensitivity chemotherapy to NSUN2 to drug multiple concern that resistance hypomethylating showed the acquired et previously increased Since unravelling (Cheng response 2014). in treatments, al., AZA siRNA results et by in knockdown) (Okamoto interference NSUN2 cells genetic of HeLa NSUN1 these (by However, impact and abrogation AZA. the JQ1 METTL1 to on inhibitor resistant and evidences is BRD4 Supporting that the not structure 2018). to (but chromatin al., BRD4 sensitive active factor are an remodelling in cells chromatin results AZA-resistant the two pol-II with NSUN1 AZA RNA of of CDK9/P- and to interaction formation and hnRNPK) the resistance DNMT2 the cells, NSUN3, or AZA-resistant involves recognized In response that directly RNA-pol-II cells. positive hnRNPK action recruit model reader explain to of syndrome the to myelodysplasic TEFb cells, mode and sensitive modifiers a AZA AML RNA In a proposed distinct treatments. in authors including response with Specifically, complexes AZA together 2018). of chromatin a NSUN3) Recently, mediator al., and needed. a et (NSUN1 is not is as investigation NSUN (Cheng does proposed effect further methyltransferases status been and antiproliferative RNA debate methylation has the the under DNMT2 DNA of Whether still that members is the treatment. and involving methylation hypomethylating for DNA mechanism molecule ma- to or setting RNA vast RNA response clinical the the by clinical that a mediated into known the in incorporated is with is it included However, correlate AZA and 2018). methyltrans- of Esteller, approved DNA (e.g., (˜90%) and FDA- methylation current (Berdasco jority DNA that been tumours inhibiting 5-methylcytosine assume haematological have drugs in to of before, decitabine) resulting treatment mentioned unreasonable cytidine or As not of RNA. however (AZA) 5 m5C is che- 5-Azacytidine of position It dynamism similar at molecule. the most group influence RNA The methyl ferases or inhibition. a DNA pharmacological of methylation at addition of RNA both the framework and is the DNA modification between within mical parallelisms exploitation several their are support There that inhibitors. methyltransferase RNA 5.1. nvivo in oes(abeie l,21) osqety rgdvlpr rmboehcmaishv started have companies biotech from developers drug Consequently, 2017). al., et (Barbieri models \ ota none htisfis-ncasdu addt agtn ET3 ae STC-15, named METTL3, targeting candidate drug first-in-class its that announced southas \ otn eutn na ciecnomto fcrmtni estv AML sensitive in chromatin of conformation active an in resulting soutand 11 nvitro in Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. eerhamda eeoigslcieadcl-ciesalmlcl niioso lBsbaiisof subfamilies AlkB of inhibitors molecule small cell-active and the 2015). selective modulate al., and developing et (Wang demethylation at cells FTO living aimed inhibit of with FTO Research derivatives efficiently mRNA inhibited fluorescein the could Two selectively in FL8, derivatives) with 2015). m6A molecules al., its and of fluorescent et of level of FL6 (Wang many design protein permeability, some of FTO the screening (and cell labelled facilitated a improved fluorescein directly has on as that based ALKBH5 well revealed over study as MA demethylation a FTO step, to for later selectivity similar 2018). a MA structures al., In for et improving inhibitors. (Yan basis MERTK) FTO PKC412 the or additional and on BCL-2 nilotinib gained TKI (e.g. knowledge the genes MA The to proliferation or tumour rhein and the to survival exposure of of Mechanistically, sensitivity stability m6A 2018). the mRNA al., targeting and et leukaemia that (Yan m6A Using demonstrated suggests increases been glioblastoma. result has of therapy This treatment (TKI) inhibitor the 2017). and for al., development strategy vivo et tumour promising in (Cui a brain mice be GSC-initiated GSC-engrafted could the methylation of of lifespan suppression the to 2015). prolonged treatment leading al., MA2 (GSC) of et effect cells antiproliferative (Huang glioblastoma-stem the in Furthermore, ALKBH5 tested over 2015). as been al., FTO identified et has (Huang previously inhibit levels (MA), mRNA specifically m m6A acid to increased of 2015). meclofenamic match displacement al., provides the best vitro et screening in In the (Huang This differences as reported the anti-inflammatory, ALKBH5. was of an and ALKBH5 identification FTO than the to rather on 2016a). binding FTO based however, al., was demethylase et respectively; work m6A (Li demethylase, The of inhibition m3C FTO inhibition and or selective m1A ALKB A ALKBH3 for the and involved to ALKBH2 are binds to sites competitively binds binding Rhein also different 2012). years Rhein al., last et 2012). the (Chen al., in rhein spotlight site product the active natural in the FTO been of has optimization dysreg- chemical inhibition m6A an its As disease, RNA inhibitors. and and small-molecule development ( JMJC specific normal by between in modifications. iron targeted impact similarity RNA ulation been and mechanistic of have inhibition 2-oxoglutarate the demethylases the the that RNA for Interestingly, JMJC and of discovery of drug 2018) part inhibitors the current al., are facilitate that et could These given demethylases protein (Hauser interesting available the very family. with is are (JMJC) similarity similarities proteins C This structural family. Jumonji exhibit dioxygenase also the effect (II)-dependent demethylases biological from RNA the demethylases explore inhibitors. lysine to demethylases needed modifications RNA derivatives. still additional adenosine is 5.2. for these models of opportunities preclinical action new in of opening development mode systems, Further high- and ring using 2020). other al., low and by showed et SAM replaced derivative (Bedi of be adenosine moiety can an academia. adenosine adenosine crystallography, Recently, the of in X-ray activity. of METTL3 protein explored derivatives of and and also inhibitor μ METTL3 analogues SAM-competitive are into 4000 a docking inhibitors of is throughput library SAM, METTL3-METTL14 of a of moieties from two discovery starting the examples, Therapeutics of one few Gotham Adenosine, Similarly, these 2019). of Cully, 2019). Out treatment 2019; (Cully, AML therapy al., AML for a et for inhibitors with development (Boriack-Sjodin preclinical 2018 METTL3/METTL14 research October with their in targets, of launched 20 front initial around the an found with at program already discovery drug has its company started has studying now Therapeutics is of Accent company investment tumours. The solid 2019). (Cully, on inhibitor application METTL3 the the with treatment after models (PDX)-AML al 2 Table oec n odlgn ffiiny(eie l,22) neetnl,atossoe htteribose the that showed authors Interestingly, 2020). al., et (Bedi efficiency ligand good and potency M oes oefrtepamclgclihbto fFOt rvn eitnet yoiekinase tyrosine to resistance prevent to FTO of inhibition pharmacological the for role a models, tde eosrtdta ramn fHL acrclswt h ty se omo A(MA2) MA of form ester ethyl the with cells cancer HeLa of treatment that demonstrated studies .Apoersuyo ml-oeueihbtr ftehmnFOdmtyaewsahee by achieved was demethylase FTO human the of inhibitors small-molecule of study pioneer A ). $ 0 ootmz h eeto fRAmdfirihbtr.Te aeanucdta the that announced have They inhibitors. RNA-modifier of selection the optimize to 40M nvitro in nvivo in n lblyicessclua 6 nmN nteB-()cl ie(hnet (Chen line cell BE-2(C) the in mRNA on m6A cellular increases globally and oeso lolsoa(u ta. 07.M2icesdmN 6 eesin levels m6A mRNA increased MA2 2017). al., et (Cui glioblastoma of models $ 4mlinpormi h hr opn ihaMTL niio in inhibitor METTL3 a with company third the is program million 54 12 6 -otiigssDNA A-containing nvitro in and Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. hr r nqeRAmdfiain htcudntb oprdwt N rhsoemdfiain.The modifications. histone or DNA with al., compared et be (Soukarieh not doses could that physiological modifications at RNA activity unique inhibitory are role with transformation, There its in cell compounds Through evaluated in of were implicated 2016). set eIF4E is al., a 2016). of eIF4E initiation et provides inhibitors pathways, translation (Soukarieh and Guanine-based oncogenic eukaryotic proposed assays of reader been angiogenesis. translation m7G have and mRNA the target tumorigenesis, of inhibit actionable regulation be an to contribute the to and needs as Efforts in effect still (eIF4E) It tissue-specific 2019). 4E a homology. (Cully, have structural factor could high specificity inhibitors exhibit increased Pan-YTF proteins of of to inhibitors application family RNA YTF- the same of of development whether the methyl the the family from determined the to to members translated YTF barrier of One YTF be The recognition could that unexplored. readers the Like is and methyl-lysine for uncertain with still 2017). crucial associated is EED discovery cage al., field drug protein methyl-RNA-reader of aromatic et Polycomb success an (He the reader contains Whether H3K27me3 family (Ganesan EED group. the (PRC2) activity protein for 2 their Polycomb identified of readers, complex been inhibitors methyl repressive are have compound marks Polycomb inhibitors epigenetic small the drug for of Potent from readers discovery these the 2019). importantly, to al., Most leading et functions. 2014). diseases, essential al., human with et in proteins (Nakao altered reader cancer of prostate set modifications. for a RNA by models other xenograft Targeting murine and 5.3. lines cell (PCA-1/ALKBH3) antigen-1 cancer 1-(5-methyl-1H-benzimidazol-2-yl)-4- demethy- cancer prostate prostate RNA compound the in additional small inhibit axis However, the to m6A able example, inhibition. was For of (HUHS015) FTO methylpyrazol-5-ol targets. in gain benzyl-3- now “druggable” a relevant is be of attention could in of suppression lases results focus the inhibition primary to the leading FTO Undoubtedly, MYC/CEBPA, 2018). has transcripts al., activity. m6A- also mRNA et FTO the R-2HG (Su the in pathways However, of of of levels proliferation action inhibition stabilization 2018). high the of al., the at and et mode through produced levels (Su the is effect cells of (R-2HG) leukaemia antitumoral more R–2-hydroxyglutarate (IDH1/2) an learn 1/2 oncometabolite we dehydrogenase the as isocitrate example, and mutant design For (BMP6) drug 6 axis. rational FTO protein FTO beyond 2016). morphogenetic for Jaffrey, extend (bone and compounds possibilities (Svensen mRNAs m6A selective The cells target contains several based HEK293C FTO it in identified are at when (UBC) study assays levels C non-fluorescent The Broccoli ubiquitin m6A appear increase These that demethylation. which complex after inhibition fluorogenic 2016). RNA-dye fluorescent the fluorescent Jaffrey, becomes a using and but of screen (Svensen construction high-throughput assay the elegant HTS on an substrate using Broccoli discovered were methylated inhibitors primary FTO (Huang ASB2) inhibits Additional and treatment also RARA, FB23-2 FB23-2 CEBPA, after MYC, importantly, levels 2019c). (e.g., m6A most al., proliferation of but et with gain Mechanistically, lines, associated transcripts cell models. with mRNA mice AML –AML compound modulates of PDX FB23-2 in Based panel cells produced a stem 2019c). leukaemia of and in al., in proliferation FB23 occupied ability inhibition et the of FTO antitumoural fully reduces (Huang to property improved be FB23-2) due physicochemical significantly and could levels the a (FB23 that m6A optimized increased developed pocket showed have selectivity were deeper FB23 MA researchers inhibitors a that keep FTO discovery to to two the benzene acid on ligand, dichloride-substituted carboxylic bulky 12 the benzyl a compound extends by the of but maintains derivative which FTO and ester design, for ethyl subfamilies, rational 2015). an AlkB al., structure-based oxygenases, with et other Using 2OG (Toh Treatment over human cells al., other HeLa FTO demethylases. et against in for protein (Toh m6A discriminates JMJD2A selectivity increases site also and compound 130-fold 2OG-binding the PHD2 to the interesting, as 30-fold of more probably exhibits instead is site 12 what nucleotide-binding Compound the explored 2015). also have demethylases pgntcmdfiain fDAmlclsaeinterpreted are molecules DNA of modifications Epigenetic nvitro in 13 u also but nvivo in odtos B32treatments FB23-2 conditions. nvitro in nvitro in M ellines, cell AML cell-based Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. ffcso agtn h ptasrpoe ntebsso htw aelandfo pgntc based epigenetic- the from Consequently, learned complexes. protein have multimeric we in what in work of obtained modifications from results basis RNA the the that of functional On translation expected the identify epitranscriptome. is to it the essential therapy, are changes targeting assays in cell-based of from result effects apart modifications approaches uncertain. RNA biological still the sophisticated non-easy is More whether is interactions other less, protein-protein phenotype chaperone or even or or resulting binding) structures Or interactions the secondary 2018). ligand protein–protein whether in al., scaffolding, or solve (e.g., et catalysis to interactions (Boriack-Sjodin enzyme protein allow mechanisms) to (e.g., not linked mechanisms do targets biochemical approaches manipulation actionable genetic “druggable” These of involve a gene. to Most modifiers. that and observations. approaches RNA-related RNA due level both applied of full m6A connections between action altered the phenotypic causality of an establish of determine mode (i.e., to cannot observations trying the we two studies of date, between the vision to associations limited but, establishing current effect) are our tumour-suppressive we is words, RNA consider other of to biochemistry In matter the pathways. important about downstream very more the A learn and to pockets need applications. binding still therapeutic ligand we for their approaches, modifiers modifiers, design RNA chemical to molecule of need rational small inhibitors unmet from all an small-molecule Apart is not effective there actuality more Thus, date selectivity. in and To enzyme but selective methods. or develop reported, potency computational target been on acceptable have based demonstrated inhibitors have guided structures inhibitors epitranscriptomic for these of point targeting number starting compounds sizeable good small a a is of This zero ( design well-characterized. the scenario precise currently from are and RNA start proteins not the reader does in and field erasers) discovery the (writers, drug that noticed the be that improve should challenges to It and future limitations near key for the the of modifiers in some RNA addressed discovery highlight ). of we be drug Below, targeting to and 2018). pharmacological biology need al., chemical will of et of (Boriack-Sjodin potential research field the the this infancy, energized in Rossell´o-Tortella its strongly 2020; in Kouzarides, epitranscriptomes and aberrant still and (Barbieri reverting development Although years and last 2020). differentiation the cell al., in in emerged et have modifications cancer RNA in of dysregulation role their the of In evidences accumulating Rapidly 2019). (Cully, therapy showed NSCLC CONCLUSION compound for 8-azaadenosine 6. such target the analogue, companies this adenosine biotech on naturally but working a ADAR1; of vitro be cancer- analogues targeting to studying multiple compound declared by in public have addition, anti-PD1 particularly any involved Therapeutics is to not expression Accent is tumours ADAR1 is be sensitizes as repress ADAR1 profoundly there to could strategy cells Nowadays, before, proteins a tumour Consequently, mentioned of challenging. in 2019). ADAR1 As family al., of ADAR et function (Ishizuka targeting therapy. immunotherapy of loss edition, cancer and A-to-I pathways, for in the related strategy consequences of the promising inhibition so pharmacological a addressed, of in not terms resulting is In determined. inhibition let-7 be mediated Gregory, precursor to TUT4-let-7 and is need of (Lin of the screen inhibitors models It TUTase of uridylation high-throughput preclinical putative automated understanding as the cancer. an selected The Using been in for have 2015). 2015). described compounds inhibitor Gregory, small and been an some (Lin chemicals, has miRNA develop cancer 15,000 a miRNA in to transferase biogenesis, expression Let7 Let-7 miRNA application uridylyl of restored let-7 terminal great Downregulation block modes 3’ selectively of the The to properties. therefore of RNA suppressor characterization manner. let-7 discovery. tumour better precursor context-specific drug to a a with recruited in requires in is precedents area implications (TUT4) this no pathological Zcchc11 in have and discovery editing action drug A-to-I of and or biology pseudouridylation chemical in However, involved modifications RNA nvivo in niiinatvt fAA2(´lze l,2003). al., (V´eliz et ADAR2 of activity inhibition oes nmlmdl arigRAmdfiaindfcs(.. nuil ncoto mutant or knockout inducible (e.g., defects modification RNA carrying models Animal models. nvitro in elbsdasy ontrpiaetepyilgclconditions physiological the replicate not do assays based cell 14 \ oton,snetesrcue fkyenzymes key of structures the since soutpoint, iue2 Figure in Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. em . altd,H,Wdak . rksn . and M., Eriksson, A., Widmark, H., ChemMedChem Wahlstedt, Writer. L., M., Epitranscriptomic Behm, Wiedmer, Human Major S.A., the Eberle, METTL3, of D., Inhibitors Huang, Promoter- Nature R.K., (2017). control. al. translation Bedi, et m6A-dependent S.C., by leukaemia Robson, Mill´an-Zambrano, myeloid Cancer G., J., maintains Rev. Shi, METTL3 Nat. L., bound cancer. Pandolfini, in K., modifications Tzelepis, RNA I., Nat. of Barbieri, diseases. Role (2020). rheumatic T. inflammatory Kouzarides, of and epigenetics I., Barbieri, the into insights lung New (2017). human Rheumatol. T. Rev. enhances Li, ADAR1 and E., enzyme Ballestar, (2016b). editing al. RNA Mart´ınez-Card´us, et the F., Oncogene A., of Setien, tumourigenesis. overexpression lung C., amplification-associated human Moutinho, Sim´o-Riudalbas,Gene L., enhances S., ADAR1 Guil, enzyme Anad´on, C., (2016a). editing al. RNA Mart´ınez-Card´us, et the F., A., Oncogene of Setien, tumourigenesis. overexpression C., Genet. amplification-associated Moutinho, Rev. Sim´o-Riudalbas,Gene L., Nat. S., control. epigenetic Guil, of Anad´on, C., hallmarks molecular The (2016). T. 487–500. marks Jenuwein, N6-methyladenosine and (2015b). C.D., S.F. Allis, Tavazoie, Nature and processing. N., a for Halberg, Is microRNAs H., HNRNPA2B1 primary (2015a). Goodarzi, S.F. H., Cell Tavazoie, Lee, Events. and Alarc´on, C.R., Processing S., RNA Tavazoie, Nuclear X., m6A-Dependent Liu, of H., Mediator Lee, H., Goodarzi, innovativeAlarc´on, C.R., of reality. and development exciting health the an human be Science”, in References will role “epitranscriptome modifications its of RNA clearer and and of pathways overview intervention better signalling comprehensive a therapeutic to have this contribution will its With we et map, barriers, disease. (Morena technical epitranscriptome data and the biological the of chemical, of view other) reliability (and generate, the these ensure to overcoming to In tools priority bioinformatic a modifi- of also development RNA be specific 2018). or should in low-abundance al., controls protocols epitranscriptome of as standardize the quantification and of standards and analyse, precision internal and site-specific identification of quantification (Zhang establishment or the better m6A-REF-seq cations, recognition for a or allow advances antibody 2019) to Technological al., on promises contexts. et NGS dependent held (Garcia-Campos using 2019), methods MAZTER-seq scale al., avoid wide like et at radiolabelling, to draws par- epitranscriptome to Improvements improvement, also the linked 2015). undoubtedly, tremendous studying cleavage a knowledge, for al., shown methods This et have valid effect. of (Linder methods pleiotropic development detection the a drugs modification since prevent associated RNA ticularly of intense to side. toxicity of difficult technical the aspect increase the extremely An could from be 2014). modifications mo- al., would RNA small et of it Wang of nature as complexity 2011; consequences ubiquitous al., the biological the whether et the and is evaluate (Jia modifications debate to epitranscriptome RNA the impediment possible targeting an intervention of still lecules number are involved high pathways the molecular improvements, epigenetic other of seeing on histone are effects and we unforeseen DNA Although induce with can cofactors of epigenetics. modifications most and including RNA 2019a), sites systems on of al., intervention regulation catalytic function et drug-based physiological shared Huang thus, the 2018; enzymes and unravel al., machinery, modifying to et RNA direction (Cheng right actual described the previously the in as move Furthermore, a modifications. are RNA enzymes) RNA-modifying for mice 13 593–605. : 35 35 4407–4413. : 4407–4413. : 519 482–485. : 15 ldz . n aic,A 22) Small-Molecule (2020). A. Caflisch, and P., Sled´z, ´ ha,M 21) cuuaino nuclear of Accumulation (2017). M. Ohman, ¨ 162 1299–1308. : 15 744–748. : 552 20 126–131. : 303–322. : 17 : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. hn . i . u,B-. ag . a,Y-. un . ta.(09.5mtyctsn promotes 5-methylcytosine (2019). al. et X., Biol. Yuan, Cell Is Y.-N., Nat. Methylation Han, mRNAs. RNA Y., stabilizing m6A Yang, through (2015). cancer B.-F., bladder Sun, al. Cell of et A., Stem pathogenesis W., Cell Li, Han, editing Pluripotency. X., M., to RNA Chen, Wang, Reprogramming Recoding M.-M., Promotes 6 (2013). Li, and N Y., MicroRNAs al. Cell-Active by Zhang, et of Regulated Y.-J., Y., Development Med. Hao, Song, T., Nat. R.K.K., (2012). Chen, Chow, carcinoma. T.H.M., hepatocellular al. to Chan, et predisposes C.H., X., AZIN1 Lin, of Zhang, Soc. Y., X., Li, Chem. L., Huang, Am. Chen, J. G., in Inhibitor. Jia, Roles FTO L., Potential Demethylase RNA Yu, RNA Its ADAR-Mediated F., -Methyladenosine and Ye, NSUN2 (2016). B., Methyltransferase Chen, RNA al. The et L., (2020). Cells S.G. Qi, Cancer. Gray, H., Gastroenterology and Yang, Cancer. A., Gastric J., Chellamuthu, of Guo, Prognosis K.T., and Progression balance Tan, editing Predicts A., RNA Editing Gut disrupted Qamra, A carcinoma. T.H.M., hepatocellular (2014). Chan, human al. in et RNA) K.J., on class Yong, act 832–843. Y., common that Li, : DeAminases A -methyladenosine J., (Adenosine Fei, 1 ADARs (2017). L., by N Qi, mediated and al. C.H., features, Lin, et T.H.M., sequence A.F., Chan, coding Palazzo, M.P., early Snyder, distinct RNA A., depletion, Akef, 5’- modification. G., with Singh, transcripts Inhibitor-Related H.N., of Myeloma Deacetylase Chua, Lymphoma. Histone C., Clin. of Cenik, Panobinostat. Management on Focus Optimal A Myeloma: (2014b). Multiple (2018). Leuk. V. With Patients R. W. in Events Popat, Gilbert, Adverse Nature and and cells. J.D., K.M., human Cavenagh, and Bartoli, yeast H., in Shin, pseudouridylation (2014a). mRNA B., 143–146. V. regulated Zinshteyn, reveals W. profiling M.F., Gilbert, Pseudouridine Nature Rojas-Duran, and cells. T.M., K.M., human Carlile, and Bartoli, yeast H., in Shin, pseudouridylation mRNA B., 143–146. regulated Zinshteyn, reveals drug profiling M.F., anticancer Pseudouridine as Rojas-Duran, proteins T.M., RNA-modifying Carlile, Discov. (2018). Drug R.A. Rev. Copeland, Nat. and targets. S., Ribich, (2019). A112–A112. P.A., Drug pp al. In Boriack-Sjodin, Research), et METTL3/METTL14. Cancer E.A., methyltransferase for protein Sickmier, RNA Association M., the (American Laidlaw, on Design, S.M., efforts discovery (2018). Res. Buker, Drug T.A., Acids A112: Wynn, al. Nucleic Abstract A.K., et Gardino, update. P.A., T.K., 2017 Boriack-Sjodin, Wirecki, pathways. B., Bagi´nski, modification P., RNA Piatkowski, of D307. E., database Rev. Purta, a Nat. M.A., MODOMICS: translation. Machnicka, for P., opportunities Boccaletto, seizing epigenetics: Clinical (2019). Rev. M. Nat. Genet. Esteller, translation. and for M., opportunities Berdasco, seizing epigenetics: Clinical (2018). M. Genet. Esteller, Sci. and Cell M., J. Berdasco, development. neuronal during editing 753. RNA adenosine-to-inosine regulates ADAR2 18 20 501–507. : 109–127. : 9 1758. : 23 270–283. : 17 435–453. : 16 19 209–216. : 134 21 17963–17971. : 978–990. : 151 637-650.e10. : 16 289–301. : 46 130 D303– : 745– : 515 515 63 : : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. u . oiisn,D,Rcai . n e .(04.Gn xrsinrglto eitdthrough mediated regulation expression Gene (2014). C. He, Genet. Rev. and Nat. G., methylation. Rechavi, RNA m6A D., reversible Proli- Dominissini, Myc-Induced Mediates Biol. Y., (NSun2) Curr. Fu, Tumours. Misu in Methyltransferase Upregulated RNA Is The and Conferring (2006). feration F.M. Collagen Watt, Targets and Lymphoma M., Hodgkin Frye, 10.1182/blood.2020005823. in doi: ALKBH3 Blood (2020). Demethylase Outcome. al. Clinical RNA et Poor m1A M., Encuentra, of V., Davalos, Loss E., Epigenetic Domingo-Domenech, Pi˜neyro, D., F., Discov. Climent, Cancer R., Cancer. Esteve-Puig, in Com- RNAs Mutation, Noncoding of IDH Epitranscriptome (2017). The al. 7 (2017). P.P. et Cell Pandolfi, P.L.M., and Cancer Dahia, M., Methylation. Esteller, D., RNA Jiang, and di- H., FTO, transcriptome Sill, of J., and Inhibition Myers, petitive protector A.-P., immune Lin, — S.M., editing Elkashef, RNA promotes A-to-I Genet. ADAR1 (2018). Rev. of Nat. E.Y. overexpression versifier. Levanon, Aberrant (2016). and Y. Oncotarget E., Gao, signaling. Eisenberg, and mTOR/p70S6K Y., activating Li, by D., progression Yang, cancer X., gastric Ye, (2016). S., Nature Yu, al. RNA. et N., Nature messenger M.S., Dou, eukaryotic Ben-Haim, m6A-seq. in N., methylome by Kol, E., N1-methyladenosine revealed Peer, dynamic S., methylomes The Moshitch-Moshkovitz, S., RNA Nachtergaele, al. m6A D., et Dominissini, mouse S., Osenberg, and L., human Ungar, the M., 201–206. of Salmon-Divon, Epigenetics S., Topology Clin. Schwartz, (2012). cancers. S., G hematologic Moshitch-Moshkovitz, of and D., treatment M., Dominissini, the Buschbeck, for A., therapy METTL3 Palau, azanucleoside-based methyltransferase A.-K., on m6A Garz, update A., (2019). Zwick, al. Ther. Targets. J., et Onco. Diesch, P., pathways. Li, p38/ERK R., through Huang, migration 12 J., and Volume proliferation Zhang, cancer S., colorectal Ye, and suppresses Y., biology chromatin Cheng, of R., landscape Deng, metabolic evolving The Genet. (2020). Rev. J.W. Nat. Proteins Locasale, Domain-Containing epigenetics. and Chem. V., YTH Anal. Ramesh, of RNA. Z., Identification in Dai, (2018). 1-Methyladenosine Y. N Wang, for of and Readers invasiveness G., the the regulates Gonzalez, as Regulates METTL3 T., Wang, methyltransferase Methylation Res. X., m6A Melanoma RNA Dai, RNA 2. A (2019). metallopeptidase 6 M. matrix by Gupta, m cells and (2017). Discov. melanoma Drug K., al. Rev. Le, Nat. et U., mark. epigenetic G., Dahal, RNA their Sun, make inhibitors Q., Rep. Chemical Qu, Cell (2019). M. Cells. L., Cully, Stem Li, Glioblastoma Exploring P., of Tumourigenesis Ye, cancer: and in H., Self-Renewal therapy Shi, and precise Q., methylation more Cui, cytosine a Biol. Towards RNA Chem. (2020). Opin. (2018). M. Curr. complexity. Berdasco, epigenetic al. and et M., J., leukaemia. Esteller, Coss´ıo, Wei, in F.P., resistance M., and Yue, Oncol. response A., Commun. 5-azacytidine J. Cloe, and Nat. organization Y., functions Int. chromatin ADAR2 Li, mediate carcinoma. L., (2017). methyltransferases cell Chen, al. squamous J.X., et esophageal Cheng, L., in Zhang, IGFBP7 H.-D., editing Qin, via F., suppressor 622–630. Wang, tumour J.-B., a Zhang, as X.-Y., Liao, Y.-B., Chen, 359–368. : 4391–4402. : 9 1163. : 19 473–490. : 57 41–49. : 17 15 16 293–306. : 971–981. : 29 90 31 382–389. : 6380–6384. : 619–620. : te ..(06.Aclinical-molecular A (2016). K.S. ¨ otze, 18 2622–2634. : 7 86161–86173. : 530 441–446. : 18 892–894. : 8 71. : 485 50 : : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. siua .. ags,RT,Ceuyt .. i . ad,A,Iaht-ele . ta.(09.Ls of Loss (2019). Nature al. shows blockade. et FTO checkpoint A., immune Iracheta-Vellve, in variant to A., Panda, resistance A overcomes K., (2013). Bi, tumours al. C.K., in et Cheruiyot, Genet. ADAR1 R., R.T., Nat. Manguso, BMI. Pfeiffer, J.J., to S., Ishizuka, due Fang, not J., risk Han, melanoma S.N., with Stacey, Differentiation. association M.H., Testis Law, for M.M., Required Iles, and RNA Cytosine-5 Body Mouse inhibits Chromatoid The selectively (2013). the Biol. al. acid of et Cell. V., Meclofenamic Component Mol. J. (2015). a Flores, of C., Is al. Cox, Targeting NSun2 S., et Blanco, Small-Molecule Methyltransferase H., S., Menon, Res. (2019c). Zhou, F., Tuorto, Acids S., al. S., Nucleic Hussain, Gong, ALKBH5. et J., over H., Li, m6A of Xu, Q., demethylation Li, Z., FTO 5- J., Dong, Cell Yan, mRNA Cancer Y., L., of Leukemia. Huang, Dong, Myeloid identification Acute Y., Genome-wide in Sheng, (2019b). Demethylase N6- FTO R. R., Oncogenic RNA Zhang, Su, of and Y., at Biol. N., Huang, Recognition trimethylation Mol. Gu, H3 Struct. (2018). J., Histone Nat. (2019a). mammals. Liu, al. al. in W., methylcytosine et et Chen, M., H., Nature T., Sun, co-transcriptionally. B.S., Wu, Huang, modification Zhao, RNA H., T., m6A Wu, guides Shi, K., 36 Zhou, lysine X., H., Biol. Weng, Cell Qin, H., Nat. translation. Huang, W., and stability mRNA Sun, binding enhances N6-methyladenosine proteins H., an IGF2BP is by Weng, methyladenosine Ythdc2 (2017). H., al. et Res. Y., Huang, Cell Liu, spermatogenesis. X., 6 mammalian Shi, m Y., regulates Guo, that destabilizes H., protein Ma, YTHDF2 Y., (2020). Zhu, RNA P.J., C.J. cells. Hsu, stem Wilusz, pluripotent and induced protein– in E.O., EED differentiation Nishimura, restrain The 755. to J., (2017). RNAs Wilusz, al. neural-specific Biol. et J., A-modified Chem. K.M., Russo, Nat. Comess, complex. A.M., H., PRC2 Heck, Zhu, the C.G., inactivates Jakob, A-395 DNA inhibitor M.L., and interaction Curtin, histone protein S., of Selvaraju, modulators Y., molecule small He, Epigenetic Biol. (2018). Chem. M. Opin. Jung, Curr. and methylation. D., Cancer Robaa, Mol. prolifera- A.-T., manner. tumour Hauser, m6A-dependent promote METTL3 in (2019b). maturation al. pri-miR221/222 controlled et immunity accelerating H.-C., by Anti-tumour Lu, 18 R., cancer (2019a). Zhou, bladder al. H., of Yu, et X., tion Nature R., Yang, cells. J., Chang, Wang, dendritic Y., J., in Han, Liu, YTHDF1 L., and Dong, methylation m6A C., mRNA Chen, through J., Liu, D., Deciphering Biol. Han, (2019). Chem. al. Synthases. Ferr´e-D’Amar´e, et Pseudouridine and (2006). S., Cell T., Viukov, A.R. Profiling. Hamma, R., Quantitative Shachar, Antibody-Independent M., via Safra, Code” U., “m6A Toth, the S., epigenetic Edelheit, of Epigenetics timeline M.A., The Clin. Garcia-Campos, (2019). dreams. M. to Berdasco, R. reality and Trans. from C., Philos. discovery: Jeronimo, interference. M.G., drug Rots, cofactor P.B., Arimondo, for A., story Ganesan, success a Sci. discovery: Biol. drug B Soc. Epigenetic (2018). A. Ganesan, Oncogene ADAR2-editing axis. (2013). CDC14B/Skp2/p21/p27 al. the et of M., modulation Pezzullo, the M., 32 through Lezzerini, growth L., glioblastoma Cifaldi, inhibits S., activity Tomaselli, C., Rossetti, F., Galeano, 110. : 998–1009. : 33 373 1561–1570. : 20170069. : 45 73–85. : 18 26 380–388. : 11 174. : 45 27 43 2–2 432e1. 428–32, : 1115–1127. : 373–384. : 567 35 178 414–419. : 566 677-691.e10. : 731-747.e16. : 270–274. : 565 13 13 43–48. : 389–395. : 1125–1135. : 20 285–295. : 26 739– : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. i,X,Ca,G,W,Y,L,J,Ce,F,Lu . ta.(09.RAmAmtyainrgltsthe regulates methylation m6A Single- (2015). RNA Methods S.R. Nat. Jaffrey, (2019). transcriptome. and the al. C.E., throughout Mason, m6Am et C., and m6A Meydan, J., Commun. of A.O., Nat. Liu, mapping Olarerin-George, nucleotide-resolution Snail. V, F., A. of Grozhik, Chen, translation B., and J., Linder, cells activity. Li, cancer TUTase Y., of Zcchc11 transition Wu, of mesenchymal G., epithelial inhibitors Acute Chai, in molecule Role X., small Oncogenic Lin, of an Identification Plays FTO (2015). (2017b). R.I. Biol. Cell al. Gregory, RNA Cancer et and Demethylase. C., Ythdf2 RNA S., Li, reader -Methyladenosine Lin, Z., m6A 6 Zuo, of N X., Suppression a Weng, (2018b). as R., Leukemia al. Su, Myeloid et H., M., Weng, Gogol, Z., Res. X.C., Li, Cell He, expansion. H., cell Shi, promotes stem NMR W., hematopoietic RNA Lett. Shao, noncoding promotes Cancer P., long carcinoma. Novel Qian, cell (2018a). Z., squamous al. esophageal Li, et in W., Yang, BPTF X., and Shi, NSUN2 C., via Reveals Zhou, progression Mapping M., tumour Cell Base-Resolution Luo, Mol. (2017a). J., Transcripts. Li, al. Y., et Mitochondrial-Encoded Li, J., and Zhou, Nuclear- Y., in Chen, reveals Methylome K., mapping Wang, m1A Transcriptome-wide M., (2016b). Distinct Zhang, al. X., et Xiong, Biol. X., S., Chem. Li, Ma, Nat. X., methylome. Shu, N1-methyladenosine L., dynamic Wang, Enzymes Repair and K., Wang, AlkB reversible Chem. Inhibits X., Biol. Rhein Xiong, J. (2016a). X., Damage. C.-G. Li, Yang, DNA and Methylated H., to Chen, J., Cells Gan, Sensitizes X., and Liu, Y., Huang, Q., Li, Chem. H. Kaniskan, immune- in Chem. therapy Epigenetic (2019). H. ADAR1 De Kaniskan, D.D. (2013b). Carvalho, al. Cancer and Sci. Rev. et A., Nat. Acad. Chakravarthy, oncology. J.M., H., Natl. Isquith, Ohtani, Proc. P.A., A.C., leukemia. Jones, Court, myeloid chronic H.-J., ADAR1 in Chun, (2013a). reprogramming C.L., 1041–1046. al. progenitor Barrett, Sci. et malignant L.A., Acad. J.M., promotes Crews, Natl. Isquith, Q., Proc. A.C., leukemia. Jiang, Court, myeloid H.-J., chronic RNA in Chun, nuclear reprogramming C.L., in 1041–1046. N6-Methyladenosine progenitor Barrett, (2011). malignant Biol. L.A., al. Chem. promotes et Crews, Nat. Y., FTO. Q., Yang, obesity-associated G., Jiang, the Zheng, adaptive of Q., substrate stress Dai, major a X., a drive Zhao, is (2019). Y., to Fu, al. G., et targets Jia, M, glioma Soler Neuropathol. in Mart´ınez-Card´us Llin`as-Arias P, Acta NSUN5 MC, program. A, RNA-methyltransferase translational Moura of de loss V, Epigenetic Dis. Ortiz-Barahona Death M, Cell export Janin apoptosis. nuclear TRAIL-induced by during activity caspase-3 PUS10 of protein, amplification RNA-modifying e3093–e3093. Reciprocal human (2017). a putative R. tumours: a Gupta, solid of and advanced A.C., and Hsieh, S., lymphoma Oncol. Jana, non-Hodgkin Lancet Tazemetostat, study. B-cell (2018). 1 refractory al. phase et or A., open-label, relapsed Varga, first-in-human, C., in Lucchesi, inhibitor, J.-M., EZH2 Michot, M., an Toulmonde, J.-C., Soria, A., Italiano, 58 58 1596–629. : 1596–629. : 12 . oz,KD,adJn .(05) eetv niioso rti ehlrnfrss .Med. J. methyltransferases. protein of inhibitors Selective (2015b). J. Jin, and K.D., Konze, U., . oz,KD,adJn .(05) eetv niioso rti ehlrnfrss .Med. J. methyltransferases. protein of inhibitors Selective (2015a). J. Jin, and K.D., Konze, U., ¨ ¨ 792–800. : 19 151–161. : 138 :1053-1074. 19 28 19 904–917. : 649–659. : 291 7 11083–11093. : 885–887. : 12 311–316. : 31 127–141. : 68 10 993-1005.e9. : 2065. : 12 430 767–772. : 57–66. : 110 110 8 : : : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. t,H,Skri . ut,R,Vlne . uff .E,Aioh,K,e l 21) DR Forms ADAR1 Cell (2013). Silencing. al. Gene et RNA-Induced K., and Ariyoshi, Processing B.-E., MicroRNA Wulff, Promote L., to Valente, Dicer 575–589. R., with Gupta, Complex M., Modifying a Genet. Sakurai, tRNA PLoS H., (2014). Cells. HeLa Ota, al. in et 5-Fluorouracil H., to Konishi, Sensitivity F., Determine Yazama, e1004639. METTL1, K., Okada, and M., NSUN2 Glioblas- Hori, Enzymes, in M., Editing Fujiwara, RNA M., Inhibits Okamoto, on Pre-mRNA Acts B Chem. That Subunit Deaminase Biol. Receptor Adenosine J. Glutamate (2017b). to toma. H.A. Binding Hundley, (ADAR3) and 3 A., Glioblas- RNA Cohen-Gadol, in A., Editing Anderson, RNA E., Inhibits on Oakes, Cell Pre-mRNA Acts B That Chem. Mol. Subunit Deaminase Biol. Receptor Adenosine Rev. J. Glutamate (2017a). to toma. H.A. Nat. Binding Hundley, (ADAR3) and ADARs. 3 A., by RNA Cohen-Gadol, Cell A., RNAs Anderson, Mol. non-coding E., and Oakes, Rev. coding Nat. of editing ADARs. A-to-I by Biol. RNAs (2016b). non-coding K. and Nishikura, coding of editing A-to-I Biol. Chem. (2016a). of synthesis Med. K. and Design Nishikura, Bioorg. (2014). drugs. cancer al. anti-prostate et M., as Ueda, inhibitors Lett. Y., (PCA-1/ALKBH3) Takeuchi, Y., antigen-1 Itoh, cancer T., prostate Shimizu, M., Epitranscriptome: (Basel). Mabuchi, the Above S., Genes Nakao, Identity. (2018). S. Cell Dyskerin- Martino, Stem Novel and and C., (2010). Emiliani, Modifications M., al. RNA Bazzucchi, et C., G., Argentati, Res. F., Storci, Cancer Morena, P., Translation. mRNA Sansone, Defective in L., through Inactivation Rocchi, component Dyskerin p53 S., of RNA Mechanism (2006). Bertoni, Mediated telomerase M., al. Calienni, and et L., pseudo-uridylation Montanaro, D., RNA Santini, Pathol. J. C., ribosomal cancer. Ceccarelli, of breast S., level human Barbieri, the J., influences Clohessy, Comprehensive Cell expression M., (2012). Codons. Brigotti, S.R. Stop Jaffrey, near L., and and Montanaro, UTRs C.E., 3’ in Mason, Enrichment O., Reveals Methylation Elemento, mRNA metastatic P., of the Zumbo, Analysis suppresses Y., MicroRNA Saletore, METTL14 primary K.D., (2017). -methyladenosine-dependent Meyer, al. 6 et N F., Wang, modulating J., Hepatology by Yuan, processing. carcinoma F., Invest. hepatocellular Liu, Cancer C., of Carcinoma. Zhou, potential Squamous Expression F., Neck Gene Yang, and NSUN2 J., Transferase Head tRNA Ma, in High (2018). Prognosis K. neck Poor Holzmann, and with and head Q., Associated Xu, gene in Methylation is H., survival methyltransferase Zeng, RNA patient G., RNA Zhu, on m6A L., score Transfer Lu, activation of (2020). cell Value T J. Oncol. Prognostic of Townsend, Oral effect carcinoma. The and the squamous (2019). V.L., modifies expression L. Cannataro, mRNA Wang, S.G., NSUN2 Gaffney, and L., C., Monit. Lu, Sci. Li, Med. L., Adenocarcinoma. Jin, RNA Colon N6-methyladenosine-dependent in C., (2015). Regulators Li, T. Pan, T., methylation and Liu, M., mRNA Nature Parisien, interactions. m6A C., RNA–protein Cell He, (2018). regulate G., Nat. switches al. Zheng, cancer. structural et Q., endometrial Dai, K., of N., Yu, tumourigenicity Liu, Z., and Lu, proliferation the S.-M., promote Liu, Biol. to B.T., activity Harada, AKT M.A., regulates Eckert, J., Liu, 20 17 17 24 1074–1083. : 83–96. : 83–96. : 1071–1074. : 292 292 65 4326–4335. : 4326–4335. : 529–543. : 210 101 10–18. : 104554. : 20 25 9435–9445. : 9 329. : 518 560–564. : 149 36 70 1635–1646. : 4767–4777. : 246–253. : 153 10 : : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. dnsn-oioiemR455 dtn rmtsmlnm rwhadmtsai.Nt elBiol. Reduced Cell (2015). Nat. al. et metastasis. M.E., and Vasquez, growth L., and melanoma Huang, translation T., promotes 311–321. facilitates Kamiya, YTHDF3 editing R.R., miR-455-5p (2017). Braeuer, adenosine-to-inosine al. A.K., et Mobley, P.J., E., Hsu, Shoshan, H., Res. Ma, Cell B.S., RNA. Zhao, N6-methyladenosine-modified microcephaly. Z., of and Lu, decay disability X., Wang, intellectual H., cause Shi, (2019). and al. humans et A.M., in Genet. Alazami, pseudouridylation I.S.M., Hum. Sayed, impair G.M.H., mutations Abdel-Salam, al. S., PUS7 et Maddirevula, M., m1A B.X., Tasak, R., The Leon-Ricardo, Shaheen, R.H., (2017). Herbst, ncRNA of Cell M., al. Pseudouridylation mRNA. Jovanovic, Dynamic-Regulated et and Widespread M.R., Reveals D., Mapping Mumbach, Bar-Yaacov, Transcriptome-wide Nature D.A., (2014). A., resolution. mediates Bernstein, Nachshon, single-base YTHDC1 S., R., at Schwartz, mRNA (2017b). Winkler, mitochondrial R., and al. cytosolic Nir, et on Y., A., landscape Cui, Sas-Chen, T., M., Elife Zhou, gene Safra, mRNAs. X., posttranscriptional methylated Wang, N6-methyladenosine for Z., of messages Zhang, export chemical G.-Z., nuclear — Luo, epigenetics I.A., Roundtree, RNA Biol. Chem. (2016). Opin. C. Curr. He, regulation. Expression and Gene in I.A., Modifications RNA Roundtree, Dynamic (2017a). C. He, Cell and T., Hema- Regulation. Pan, and M.E., Evans, Hematopoiesis I.A., in Roundtree, Epitranscriptomics Discov. RNA Cancer (2020). – Blood M. Esteller, synthases Malignancies. Biol. and tologic pseudouridine RNA G., Ferrer, stand-alone expression? Adenosine-to-Inosine M., gene Eukaryotic Rossello-Tortella, of (2014). regulators (2017). emerging al. Res. and U. et Cancer enzymes Kothe, H., modifying Carcinoma. Liu, and Cell F.-F., Squamous A.C., Li, Esophageal Rintala-Dempsey, in Y.-H., ADARs Zhu, by T.H.M., Mediated Clin. Chan, Editing J. J.-J., RNA E1905. Qiao, myeloid trial Y.-R., acute Intergroup Prolonged Qin, and Leukemia (2014). US syndrome the al. myelodysplastic of et for P.L., results entinostat Oncol. Greenberg, changes: without A., myelodysplasia-related Melnick, or with R., is with leukemia Ketterling, editing azacitidine M.E., RNA Biol. of Figueroa, A-to-I Genome administration Z., Massive reveals abundance. Sun, dsRNA RNA (2017). T., with E.Y. Prebet, hyper-edited correlates Levanon, and of and Metazoa map E., the genome-wide across Eisenberg, common A B.A., Knisbacher, and H.T., (2014). Dyskerin Porath, E.Y. Commun. (2015). Levanon, Nat. al. and sites. et new S., numerous Oncotarget G., Carmi, Bellezza, patients. H.T., cancer J., lung Vannucci, Porath, in A., survival Siggillino, in condition D., Pseudouridylation may Trere, expression RNA A (Basel). TERC V., 6 (2017). Genes Ludovini, m L. Worse. M., snRNA Montanaro, for Penzo, U6 and and The Better D., For Trere, (2017). Medicine: F., al. and Zacchini, et Physiology A.N., B.P., Tu, Guerrieri, Cell Y., M., Retention. Xie, Penzo, Intron V., Synthetase O. SAM Hunter, the Regulates Targeting K., METTL16 Cell Liu, Methyltransferase (2019). Leukemia. B., Myeloid Chen, al. Acute K.E., et in Pendleton, I., Cells Ivanova, Stem A., Cancer Shmakova, Compromises G.J., Cell Selectively Stem Spencer, YTHDF2 J., Reader m6A Campos, RNA M., Morgan, J., Paris, 32 25 1242–8. : 137-148.e6. : 138 169 159 231–239. : 1187–1200. : 148–162. : 5 30 4726. : 46–51. : 1 26–31. : 21 27 315–328. : 6 :. 8 :. 14 6 1185–1196. : 21755–21760. : 169 551 18 824-835.e14. : 185. : 251–255. : 74 840–851. : 17 : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. u .. ikrn,BF,Ceg . acr,S,Nue,D,Mnea . ta.(07.TeN6- The (2017). al. et G., Minuesa, D., Nguyen, hematopoietic S., Med. normal Nat. of Zaccara, cells. differentiation myeloid leukemia Y., controls and METTL3 Cheng, enzyme B.F., (m6A)-forming methyladenosine Pickering, L.P., Vu, Immunol. Trends munotherapy. Soc. Adenosine Chem. RNA-Editing Am. L., an J. cytosine Villanueva, for Design. RNA Analogues Inhibitor Substrate and Biol. (2012). (2003). Investigation P.A. Mol. Mechanistic al. Beal, Deaminase: Struct. and et Nat. L.M., S., synthesis. Easterwood, Kellner, V´eliz, protein E.A., S., and stability Hofmann, tRNA M., promotes Schaefer, of NSun2 19 and T., as Modulation Dnmt2 Musch, role by emerging R., methylation (2015). its Liebers, and RNA 5-methylcytosine F., al. Rev. modification Tuorto, et RNA Interdiscip. V.A., dynamic Wiley The Biol. Polito, mark. Genome (2019). epitranscriptomic S., A. an glioblastoma. Lusser, Galardi, in and deaminase S., L., ADAR2 -methyladenosineTrixl, based Raho, by 6 strategy processing S., and A N Alon, expression of editing, (2015). F., microRNA inhibitor Galeano, al. cell-active et S., and C.W.Q., Tomaselli, subfamily-selective Tang, a J.J.A., Sci. Low, to Chem. J., FTO. leads demethylase Tan, specificity L.Z.M., nucleotide Lau, catalyzes ADARB1 on L., Genet. (2017). Sun, Nat. senescence al. J.D.W., replicative rhythm. et Toh, delays RNA S., Kuroda, regulates NSun2 S., and Shimba, (2015). editing Y., NY). A-to-I al. Suzuki, (Albany. H., circadian et Aging Ozaki, Y., H., translation. Yoshitane, Dou, CDK1 H., B., elevating Terajima, and Jiang, helicase HIF-1 translation Z., RNA (KIP1) which Liu, p27 (2016). J., by repressing al. Xing, by efficiency X., et the Fan, J., H., of Tang, Konno, enhancement H., the Ikeda, epitranscriptome Lett. via Cancer K., The metastasis Takai, Oncol. (2017). translated. J. cancer M., al. Int. promotes et Tsunetomi, cells. T., YTHDC2 cancer K., Tanikawa, pancreatic Satoh, RNA-Modifying in M., A., Study radioresistance Toratani, Tanabe, and to J., chemo- Tool Koseki, promotes A., METTL3 a writer Asai, as m6A M., Aptamers Konno, Anti- RNA K., Exhibits Taketo, Fluorescent R-2HG (2016). (2018). Biol. al. S.R. Chem. et Cell Jaffrey, Enzymes. Y., and Qing, N., M., Wunderlich, Svensen, Cell S., Signaling. FTO/m6A/MYC/CEBPA Nachtergaele, Targeting C., by Li, acute Activity The adult tumour L., (2018). in Dong, al. activity R., et clinical Su, M.R., modest Savona, has J.G., and Berdeja, methylation Blood R., H3K79 leukemia. Tibes, reduces pinometostat D.A., and inhibitor Rizzieri, structural DOT1L G., Synthesis, Garcia-Manero, eIF4E: E.M., factor Stein, Chem. PUS10 initiation Med. human translation J. of the Eur. roles of characterisation. Differential functional inhibitors (2020). non-nucleotide al. and P et Biol. mimetic A., B., Bastide, Chem. M.W., Xie, Nowicki, Nat. F., B., Soukarieh, pseudouridylation. Lu, tRNA H., and Meng, processing C., miRNA Zhu, in Y., Zhuang, J., Song, ldz . n ie,M 21) tutrlisgt notemlclrmcaimo h 6 rtrcomplex. writer m6A the of mechanism molecular overexpression the DKC1 into insights (2009). Elife Structural al. Cancer (2016). et M. J. M., Jinek, Br. Sled´z, and Muller, P., A., progression. Wlazlinski, cancer R., prostate Engers, with J., associated Hatina, C., Hader, P., Sieron, ´ 900–905. : 5 :. lae-rio . n selr .(00.TeCnrbto fEieeist acrIm- Cancer to Epigenetics of Contribution The (2020). M. Esteller, and D., Alvarez-Errico, ´ 131 2661–2669. : 376 23 34–42. : 23 415–425. : 6 41 112–122. : 1369–1376. : 676–691. : yy . oe,C,Ddk . ta.(06.Dsg fnucleotide- of Design (2016). al. et K., Dudek, C., Jones, T., ¨ oyry, 124 200–217. : 22 10 101 e1510. : 1410–1416. : 49 146–151. : 16 172 160–169. : 90-105.e23. : 125 10867–10876. : 7 1143–1155. : α RAis mRNA 16 5. : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. ho . atn .. ai,SL,Foec,B,Ae,SJ,adSajad ..(04.Rglto of Regulation (2004). R.A. Spanjaard, Steroid and of Modification S.J., Single-base Cell Posttranscriptional Mol. Ames, (2019). through Activator. Synthase B., al. RNA Pseudouridine Florence, Receptor a et by S.L., Z., Activity Davis, Adv. Receptor Zhang, J.R., Sci. Nuclear I.A., method. Patton, Roundtree, antibody-independent X., ALKBH5 an C.-G., Demethylase Zhao, by Yang, A A Y.-L., 6 6 m m Zhao, of (2017c). L.-Q., mapping al. I Chen, et type Z., Pro- Z., by Cell Zhang, Lu, induced and Expression S., is FOXM1 Cell Zheng, editing Sustaining Cancer by K., RNA Program. Cells Lin, liferation (2017b). Stem-like A., Glioblastoma J. of Zhou, He, Tumourigenicity B.S., Maintains and Zhao, X., Biol. S., Hu, Tumour Zhang, carcinoma. J., cell Huang, squamous NANOG Z., esophageal of Tang, in A-demethylation Z., interferon breast the 6 Chen, induces m J., Hypoxia ALKBH5-mediated Zhang, (2016). al. and Sci. et Acad. HIF-dependent I., Natl. haematopoietic Chen, by Proc. H., modulates phenotype mRNA. Zhang, m6A J.W., cell (2017a). Bullen, stem al. H., et cancer Lu, D., D., Samanta, Ma, C., Y., Zhang, Nature Yang, specification. L., Res. cell Wang, Cell progenitor B., reader. and m5C Sun, an stem Y., as promotes FTO Chen, ALYREF 5-methylcytosine demethylase (2017b). C., and al. methyltransferase mRNA Zhang, et the m6A W.-Y., as Lai, (2019). NSUN2 J.-W., al. Xu, — Y.-S., export et Chen, mRNA Commun. B.-F., Y., Sun, methyl- Nat. Deng, Y., blockade. Yang, tRNA P., anti-PD-1 X., Yang, of Shah, to G., Association response Park, and (2017a). tumourigenicity Y.-H., L. melanoma Cui, Lu, regulates J., Oncol. and Futur. Wei, H., survival. the S., cancer Huang, Promotes Yang, ovarian C., METTL3 with Huang, (2020). signature M., molecular al. NSUN2/IGF-II Zhang, et transferase Oncol. E., Y., Front. Pathway. Wang, Risch, MYC N6-methyladenosine Q.-N., J., the Wu, dynamic Yang, Targeting via A J.-H., via (2018). Lu, Growth Cancer Gastric al. K., Cell of Yu, et Promotes Res. Progression Z.-H., N., Cell NSun2 Zhao, Chen, inhibitors. (2015). kinase J., D.-D., tyrosine Yang, Pang, al. to J., resistance et acquired Liu, X., and Z., intrinsic Zhang, regulates Zhang, methylome Z., A., Al-Kali, Liu, Biol. F., Reader H., Cell. Yan, A Tang, Mol. 6 Translation. X., m 1 Nuclear Cai, Kinase (2016). Cyclin-Dependent J., al. Elevating et Yi, B.-F., J., Sun, Xing, Mech. Y.-J., Cell Hao, Regul. Mol. Y.-S., Splicing. Gene Chen, mRNA - U., Regulates Dahal, Acta YTHDC1 S., Biophys. Adhikari, Biochim. W., Xiao, cells. cancer CSF-1 ovarian the and increases demethylation breast 35–46. m1A in ALKBH3-induced Human stability Stem (2019). mRNA S.K. Cell Chambers, and Modification. H.-H., Hematopoietic m6A Woo, Inhibits METTL14 mRNA (2018). via al. et Leukemogenesis N6-methyladenosine-dependent L., Dong, Promotes (2014). Cell B.S., and al. Zhao, X., Differentiation et Qin, D., Stem/Progenitor H., Wu, Han, H., Huang, Y., H., Yue, Weng, Nature G.C., stability. Hon, RNA A., messenger Soc. of Gomez, Chem. regulation Z., Am. Lu, J. Protein. X., Wang, FTO Bifunctional of as Derivatives Labeling Fluorescein and (2015). al. Inhibiting et 13739. Y., Simultaneous Chen, the F., Wu, Malignant for H., on Su, Molecules Regulators Oncol. Y., Methylation Front. Huang, RNA T., Carcinoma. Hong, m6A Cell T., of Wang, Clear Effect Renal (2020). in X. Gou, Prognosis and and W., Progression He, C., Zhang, J., Wang, 22 191-205.e9. : 113 31 591-606.e6. : E2047–E2056. : 15 549–558. : 549 505 61 273–276. : 507–519. : 117–120. : 23 39 5 35 101042831770854. : eaax0250. : 4043–4052. : 10 :. 10 :. 10 13 28 2782. : 27 137 1981–1990. : 1062–1076. : 606–625. : 13736– : 1862 : Posted on Authorea 9 Nov 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160495480.00891340/v1 — This a preprint and has not been peer reviewed. Data may be preliminary. druggable-epitranscriptome-compounds-that-target--modifications-in-cancer 2.pdf Table file Hosted druggable-epitranscriptome-compounds-that-target-rna-modifications-in-cancer 1.pdf Table file Hosted druggable-epitranscriptome-compounds-that-target-rna-modifications-in-cancer 2.pptx Figure file Hosted druggable-epitranscriptome-compounds-that-target-rna-modifications-in-cancer 1.pptx Quimatryx. Figure and Ferrer of consultant is file M.E. Hosted interest. of conflicts no discloses MB STATEMENT INTEREST OF RTI2018- ME CONFLICT (LCF/PR/GN18/51140001). no. Foundation Professor. project Banking Research Caixa” ICREA (ERDF) “la an Fund Foundation; is Development Agencia Innovaci´on Cellex Regional (MCI), Funds e the Ciencia and Development European 2017SGR1080 de 094049-B-I00 supported and Regional Ministerio Investigaci´on labis no. (AEI) Catalunya); European M.E. AGAUR-projects de de M.B. at and by (Generalitat Estatal SLT/002/16/00374 Research Government at co-funded Catalan no. PI18/00910). Research the III PERIS-project and of Department Carlos (PI15/00638 Health Europe Salud support. the build de by institutional to Instituto way for a by Catalunya (ERDF/FEDER) supported de is Programme/Generalitat lab CERCA thank We manuscript. the of FUNDING aspects all to contributed authors All CONTRIBUTIONS AUTHOR modification. the m6A, with 2. together cytidine; Figure listed 5-methyl are m5C, modification N1-methyladenosine; writers RNA m1A, known each edition; N6-methyladenosine. The for Adenosine-to-Inosine illustrated. (R) I, are readers pseudouridine; edition and adenosine-to-inosine (E) erasers and (W), pseudouridylation , at tion (2016). 1 al. Figure et H.-J., Chun, LEGENDS L., Cell FIGURE Stem Balaian, Cell Biogenesis. N.P., Let-7 Santos, of Impairing by Regulation Delos 177–191. Self-Renewal Clock : Cell A., Stem Circadian Leukemia and Sadarangani, (2018). Drives Rep. Functions Activation A.C., al. ADAR1 Cell et Multiple Court, Methylation. C.M., mRNA (2019). M.A., Cham, m6A al. Zipeto, Y., of Li, et Modulation X., by M., Weng, Metabolism Zhang, K., Lipid Oncol. Frazier, Hepatic Front. H., J., Cancers. Yu, Jiang, Human X., S., in Zhong, METTL3 Wang, of Y., Role Zhao, the Underlying Mammalian X., Cell Mechanisms a Mol. Dong, Is Fertility. ALKBH5 Mouse W., (2013). and al. Zheng, et Metabolism C.J., RNA Li, Impacts C.-M., Huang, that P., Demethylase Fedorcsak, Y., RNA Niu, J.A., Dahl, G., Zheng, ceai iga ftepeoiatmN oaetmdfiain.Ratosfrmethyla- for Reactions modifications. covalent mRNA predominant the of diagram Schematic . hlegsaddffiute o rgdsoeytreigRNA-modifiers. targeting discovery drug for difficulties and Challenges vial at available at available vial at available at available https://authorea.com/users/374362/articles/491891-towards-a- https://authorea.com/users/374362/articles/491891-towards-a- https://authorea.com/users/374362/articles/491891-towards-a- https://authorea.com/users/374362/articles/491891-towards-a- 24 9 25 49 :. 1816-1828.e4. : 18–29. : Ψ, 19