Abstract
N6-Methyladenosine (m6A) is the most abundant modification in eukaryotic mRNA, and plays important biological functions via regulating RNA fate determination. Recent studies have shown that m6A modification plays a key role in hematologic malignancies, including acute myeloid leukemia. The current growth of epitranscriptomic research mainly benefits from technological progress in detecting RNA m6A modification in a transcriptome-wide manner. In this review, we first briefly summarize the latest advances in RNA m6A biology by focusing on writers, readers, and erasers of m6A modification, and describe the development of high-throughput methods for RNA m6A mapping. We further discuss the important roles of m6A modifiers in acute myeloid leukemia, and highlight the identification of potential inhibitors for AML treatment by targeting of m6A modifiers. Overall, this review provides a comprehensive summary of RNA m6A biology in acute myeloid leukemia.
Keywords: Acute myeloid leukemia, Mapping, RNA m6A modification
1. INTRODUCTION
Maintenance of hematopoietic system throughout the lifetime relies on hematopoietic stem cells (HSCs), which possess unique self-renewal capacity and multilineage differentiation potential and replenish all lineages of blood and immune cells.1 The imbalance of hematopoietic homeostasis can lead to the development of various blood diseases such as leukemia, bone marrow failure, myelodysplastic syndrome. Genetic or epigenetic alterations occurred in hematopoietic stem/progenitor cells (HSPCs) transforms them into leukemia stem cells (LSCs), which subsequently initiate the development of leukemia, such as acute myeloid leukemia (AML).2 AML is an aggressive and fatal hematologic malignancy, which is characterized by uncontrolled expansion and impaired differentiation of myeloid progenitor cells.3 In clinic, chemotherapy is still the standard treatment strategy for most newly diagnosed patients with AML. Currently, the 5-year survival rate is around ~30% for AML patients under 60 years old.4,5 Thus, exploring the molecular mechanisms of AML development and investigating potential therapeutic targets for AML treatment remain a big challenge in this field.
Leukemogenesis is a multistage process that is governed by complex regulatory networks.6 Normally, HSCs predominantly reside in a quiescence state that is coupled with different controlled systems such as metabolism, protein synthesis and stress response system.7–9 Dysregulation in these tight and dynamic controls of HSC activity results in malignant transformation. Increasing evidence has suggested that mutations of some epigenetic modifiers such as DNMT3A and TET2 are common early events of leukemogeneis.10,11 These founder mutations result in epigenetic alterations and aberrant transcriptional networks, which subsequently prime HSPCs into a state called pre-leukemic stem cells (pre-LSCs). Secondary or further genetic alterations will fully transform these pre-LSCs into LSCs.12 Histone modification is another aspect of epigenetic regulation. For instance, translocations of MLL, encoding a histone methyltransferase, are recurrently found in chromosomal rearrangements involving 11q23 in AML, highlighting the significance of this epigenetic modifying enzyme in AML development.13 Other histone modification enzymes, such as DOT1L, EZH2, ASXL1, KDM1A, also involve in leukemogenesis by providing a selective advantage to LSCs.14–17
Similar to DNA methylation and histone modification, RNA modification has recently been implicated in AML initiation and progression. N6-methyladenosine (m6A) is the most enriched modification in eukaryotic mRNA and long non-coding RNA, and participates in regulating RNA metabolism by affecting RNA splicing, nuclear export, translation, and degradation.18 Increasing evidence suggests that RNA m6A modification acts as an important adaptive-response mechanism in regulating various biological processes at the epitranscriptomic level. Recent studies have also shown that m6A modification plays an important role in many stages of normal and malignant hematopoiesis. These findings not only uncover a new regulatory layer in leukemogenesis, also provide novel and promising therapeutic strategies for targeting leukemia. Thus, in this review, we first describe m6A machinery, and then focus on AML and discuss recent advances of RNA m6A modification in AML.
2. RNA m6A MODIFICATION
Among over 170 different chemical modifications of RNA that have been identified to date, m6A is the most common one in mammalian mRNA. Comprehensive analysis of mRNA m6A using by establishing high-throughput sequencing reveals enrichment of m6A sites nearby the 5′ untranslated regions (5′-UTR), stop codon, 3′ untranslated regions (3′-UTR), and long internal exons with conserved RRACH motifs (R = G, A, and U; H = U, A, and C).19 Each transcript contains 3 to 5 or more m6A modification sites, accounting for 0.1% to 0.4% of total adenine (m6A/A).20 It is known that m6A modification is reversible and dynamically regulated by methyltransferase complex that catalyzes the installation of m6A, and by demethylases responsible for m6A removal. The methyltransferase complex, also called writer, is a multicomponent nuclear complex composed of 2 methyltransferase-like proteins (METTL3 and METTL14) and several regulatory proteins (WTAP, VIRMA, CBLL1, RBM15/RBM15B, METTL16, and ZC3H13). m6A modification is removed by either ALKBH5 or FTO (also called eraser), both of which belong to the ALKB family of the e(II) and α-ketoglutarate-dependent dioxygenases. In general, m6A methylation sites can be recognized by some specific RNA binding proteins, including YTHDC1/2, YTHDF1-3, and IGF2BP1-3, which play different roles in fine-tuning mRNA metabolism. As these RNA m6A machineries have been widely discussed in many decent literatures, we just briefly describe them below (Fig. 1).
Figure 1.
m6A modification in RNA metabolism. m6A modification is reversible and dynamically regulated by methyltransferase complex (including METTL13, METTL14, METTL16, WTAP, RBM15, KIAA149, and ZC3H13) and demethylase including ALKBH5 and FTO. RNA binding proteins (including YTHDC1/2, YTHDF1-3, IGF2BP1-3, HNRNPA2B1, and HNRNPC) participates in regulating various stages of RNA metabolism including alternative splicing, nuclear export, pri-miRNA processing, translation, stability, and degradation.
2.1. RNA m6A methyltransferases
METTL3 and METTL14 compose the core of the methyltransferase complex, and METTL3 is currently the sole catalytic component in this complex while METTL14 plays an important role in structural stabilization and RNA-substrate binding. This complex catalyzes the transfer of methyl group from S-adenosylmethionine (SAM) to the sixth N atom of RNA adenosine. METTL3 has the leading helix structure (LH) and nuclear localization signal (NLS) domains, which mediate the interaction of METTL3 with METTL14.21–23 CCH-type zinc finger domain (ZFD) of METTL3 serves as the target recognition domain and fulfills the methyltransferase activity of this complex.24 Moreover, both METTL3 and METTL14 have SAM structure binding domain—methyltransferase domain (MTD).
The METTL3–METTL14 complex composes several key subunits that guides the core complex onto specific mRNA regions and are responsible for determining its activity and specificity. WTAP recruits METTL3 and METTL14 to nuclear speckles for m6A methylation.25 VIRMA (KIAA1429) is identified as a scaffold that orchestrates the catalytic core components to guide region-selective methylation. RBM15 is recruited to the methyltransferase complex to promotes RNA methylation.26 ZC3H13 mainly functions to retain the ZC3H13–RBM15–WTAP complex in the nucleus to regulate m6A methylation.27,28 RBM15, a homolog of spen, binds and recruits the m6A methyltransferase complexes to XIST, leading to m6A formation that mediates transcriptional repression.29 VIRMA contains the RBP domain, and facilitates m6A writer complex to the 3′UTR region.30 Post-translational modifications can also affect the activity of METTL3. For instance, TBK1 is a key kinase of antiviral pathways, and a recent study show that TBK1 directly phosphorylates METTL3 at serine 67 and promotes its activation and m6A modification to stabilize IRF3 mRNA.31
Besides METTL3-METTL14 complex, METTL16 is identified as an alternative m6A methyltransferase. Human MAT2A encodes the SAM synthetase expressed in most cells. Recent study found that METTL16 functions as a m6A methyltransferase and catalyzes the methylation of MAT2A hairpin HP1, which regulates MAT2A expression by affecting its intron splicing. Due to the similarity between HP1 and U6 snRNA methylation sites, METTL16 is also responsible for m6A methylation of U6 snRNA.32 METTL16 can also deposit m6A mark on the pre-mRNA. Recent work found that the C. elegans writer METT-10 (the orthology of mammalian METTL16) installs an m6A mark on the 3′ slice site of SAM synthetase pre-mRNA, which inhibits its proper slicing and protein production.33 Splicing inhibition by 3′ splice site m6A is conserved in mammals. METTL16 also binds to other ncRNA as well as numerous lncRNA, suggesting that it might be responsible for m6A methylation of these RNAs.34 In sum, additional m6A methyltransferases remain to be identified.
2.2. RNA m6A demethylases
FTO and ALKBH5 are known as the 2 m6A demethylases, and both belong to the ALKB family. FTO was widely studied as a fat mass and obesity-associated protein associated to metabolic disorders such as diabetes and obesity. Recently, FTO was reported to have efficient oxidative demethylation activity and can sequentially oxidize m6A to hm6A or fm6A, which are unstable and can be hydrolyzed to adenine.35 FTO can also catalyze the demethylation of m6Am on mRNA and snRNAs, and m1A on tRNA. The cellular distribution of FTO affects its access to different RNA substrates, which might play a role in determining FTO’s substrate specificity. FTO in the nucleus has a higher affinity for m6A, while FTO in the cytoplasm has a higher affinity for m6Am.36 Currently, factors that influence the location of FTO remain unknown yet. ALKBH5 is another demethylated enzyme that specifically recognizes RNA m6A and directly remove RNA m6A.37 Although α-KG and Fe2+ are essential for the demethylation activity of FTO and ALKBH5, it remains unclear how the activities and specificity of these m6A erasers are regulated in different contexts.
2.3. RNA m6A readers
RNA m6A readers recognize different m6A sites under distinct contexts and function as the key in regulating the fates of m6A targets. One group of m6A readers contain the YT521-B homology (YTH) domain that is responsible for recognizing m6A, including YTH domain family 1-3 (YTHDF1-3) and YTH domain containing 1-2 (YTHDC1/2). Even belonging to the same class, these readers mediate different fates of m6A-modified mRNAs. YTHDF1 facilitates translation initiation of m6A-modified mRNAs by interacting with eukaryotic initiation factor 3,38 while YTHDF2 mainly destabilizes m6A-modified mRNAs via directly recruiting the CCR4–NOT deadenylase complex in the cytoplasm.39 YTHDF3 facilitates mRNA translational efficiencies of its targets and is dependent on m6A methylation. In addition, a recent study has reported that YTHDF3 cooperates with YTHDF1 to facilitate translation of protein synthesis and affects YTHDF1-mediated decay of methylated mRNA.40 YTHDC1 mainly locates in the nucleus and regulates the export of m6A-tagged mRNAs from nucleus to cytoplasm.41–44 YTHDC1 also mediates mRNA splicing by recruiting 2 competitive mRNA splicing factors serine/arginine-rich splicing factor 3 (SRSF3) and SRSF10.44 YTHDC2 is present both in the cytoplasm and nucleus, where it affects translation and stability of its target mRNAs.45
Another group of m6A readers is IGF2BP1-3, which have 56% identify on their amino acid sequence homology. These readers contain 4 repetitive KH domains, and the third and fourth KH domains are responsible for recognizing m6A sites. IGF2BPs predominantly enhance the stability and translation of m6A-modified mRNAs.46 IGF2BP3 is also required for pre-mRNA splicing.47 Our recent work reveals that YBX1 cooperates with IGF2BPs to promote the stability of m6A-tagged transcripts,48 suggesting YBX1 is 1 component of IGF2BP regulatory machinery. Several heterogeneous nuclear ribonucleoproteins (HNRNPs) including HNRNPC, HNRNPG, and HNRNPA2/B1 also function as m6A reader. HNRNAPC as a nuclear protein can regulate alternative splicing of m6A-modified transcripts. HNRNPA2B1 functions as a m6A nuclear modification reader that can regulate primary microRNA processing and alternative splicing.49 5′UTR m6A modification also can be recognized by HNRNPG and YTHDC1, which prevent the integrator complex from splicing the nascent RNA and promotes the transcription of the nascent RNA.50 Overall, m6A readers are crucial in determining the fate of their target mRNAs via recruiting diverse regulatory machinery to m6A sites.
3. TECHNOLOGIES FOR MAPPING m6A
The field of epitranscriptomics was beginning to revive around 10 years ago, which mostly benefit from the seminal studies that establish a strategy for mapping the transcriptome-wide m6A profiling.19,51 These works have ignited the exponential research about RNA modifications under various physiological and pathological conditions, and the growth of epitranscriptomic research also stimulated the development of new tools or approaches for detecting and mapping RNA marks even in lower-abundance RNA species. Hereby, we briefly discuss the current methods for mapping the transcriptome-wide m6A modification (Table 1). Based on the strategies for detecting or recognizing m6A sites, these methods can be simply divided into 2 groups: antibody-dependent mapping and antibody-independent mapping (Fig. 2).
Table 1.
A summary of advantages and limitations of m6A mapping methods.
| m6A detection methods | Advantages | Limitations | Ref |
|---|---|---|---|
| MeRIP-seq/m6A-seq | First method for global view of m6A | Large amounts of starting RNA Low resolution |
51 |
| miCLIP-seq | Single-nucleotide resolution | Large amounts of input RNA Non-specific binding of antibody |
52 |
| m6A-LAIC-seq | Quantify m6A stoichiometry | Low resolution | 53 |
| m6A-seq2 | Allows quantification across genes and samples | Low resolution | 54 |
| SLIM-seq | Low input material High sensitivity |
Low resolution | 55 |
| MAZTER-seq/m6A-REF-seq | Single-nucleotide resolution Quantitative tracking of m6A |
Only detect 16%–25% of m6A sites | 56 |
| DART-seq | Allow detect m6A accumulation Low amounts of input RNA |
Low sensitivity Only identify partial m6A signal |
57 |
| m6A-label-seq | Single-nucleotide resolution | Low efficiency Lack stoichiometric information |
58 |
| m6A-SEAL-seq | Less false-positive signals | Lack stoichiometric information Unstable efficiency |
59 |
| m6A-SAC-seq | Single-nucleotide resolution Quantitative tracking of m6A |
Low specificity and efficiency | 60 |
Figure 2.
Outlines of different strategies for m6A mapping. (A) Antibody-dependent mapping methods. MeRIP-seq/m6A-seq fragment the RNA to ~100 nt sized fragments following with immunoprecipitation with anti-m6At antibody to enrich RNA fragments containing m6A. miCLIP detect m6A positions at single-nucleotide resolution by inducing mutation or truncation. m6A LAIC-seq using in vitro transcripted m6A spike-ins to quantify methylated versus nonmethylated transcripts. m6A-seq2 add RNA barcodes to fragmented RNA originating from distinct samples, and pooled together followed by a single m6A-IP performed on pooled RNA samples. SLIM-seq combining the advantages of m6A-LAIC-seq and SMART-seq, decoding m6A modification on full-length transcript with high sensitivity for low-input. (B) Antibody-independent mapping methods. MAZTER-seq/m6A-REF-seq use MazF to cleave RNA at unmethylated sites occurring at ACA motifs. DART-seq use YTH domain to recognize m6A and APOBEC1 to mark m6A site. m6A-label-seq metabolically incorporate allyl-group into the adenosine sites (a6A) then transformed (a6A) into N1, N6-cyclized adenosine (cyc-A), the cyc-A induced misincorporation during reverse transcription. m6A-SEAL-seq use FTO’s enzymatic oxidation and a DTT-mediated thiol-addition reaction to convert m6A into dm6A, then labelled by biotin and pulled down by streptavidin. m6A-SAC-seq used Dim1/KsgA family of dimethyltransferases, converting m6A into N6,N6-dimethyladenosine (m62A) in consecutive methylation reactions.
3.1. Antibody-dependent m6A mapping
The first high-throughput method for mapping a transcriptome-wide m6A landscape was developed independently by 2 groups in 2012.19,51 Based on the high sensitivity and selectivity of anti-m6A antibody in detecting m6A, researchers from these 2 groups established the same strategy that combines m6A-specific methylated RNA immunoprecipitation with next-generation sequencing, thus this method is called MeRIP-seq or m6A-seq. The procedure involves randomly fragmenting the RNA to ~100 nt sized fragments following with immunoprecipitation with anti-m6A antibody to enrich RNA fragments containing m6A. This method first identifies more than 7000 mammalian genes that contain m6A in their transcripts, and reveal the distribution of m6A sites that are enriched near stop codons and in 3′ UTRs. These seminal studies provide a tool for investigating m6A profiling in a transcriptome-wide manner, and also indicate that RNA m6A has a fundamental role in regulating gene expression. However, the approach has several limitations, including low resolution and a requirement of large amounts of starting RNA. Since then, different mapping tools or strategies have been developed to reach more effective and accurate m6A profilings.
Although m6A positions could be predicted from MeRIP-seq by searching for DRACH motifs near the point of highest read coverage, precisely identifying m6A residues at high resolution is challenging. Due to the size of fragmented RNA, precise m6A positions cannot be identified. Considering that cross-linked proteins to RNA in living cells would result in mutations or truncations in the cDNA when it is reverse transcribed, researchers introduce UV cross-linking and immunoprecipitation for m6A antibody-enriched RNA fragments, named miCLIP.52 miCLIP enables to detect m6A positions at single-nucleotide resolution, and can also detect m6Am, a modification found at the first nucleotide of certain mRNAs. miCLIP requires large amounts of input materials. In addition, different commercial m6A antibodies might cause distinct mutation or truncation patterns in the cDNA, which would bring a lot of difficulties and noises for decoding the m6A residues.
To quantitatively deconvolute methylated to nonmethylated transcripts in a transcriptome-wide manner, m6A-level and isoform-characterization sequencing (m6A-LAIC-seq) was developed. This approach does not fragment the RNA before anti-m6A RNA immunoprecipitation, instead simultaneously sequences intact full-length transcripts in both m6A-positive and m6A-negative fractions following RNA immunoprecipitation. Importantly, in vitro transcripted m6A spike-ins are introduced to quantify methylated versus nonmethylated transcripts.53 Thus, this method allows to determine the methylation stoichiometry of different transcript isoforms at the full-length levels rather than the m6A position levels.
One of the major limitations of these above methods are the need for high input amounts. To overcome this limitation, 2 new approaches (m6A-seq2 and SLIM-seq) were recently developed. In m6A-seq2, barcoded RNA adapters are added to fragmented RNA originating from distinct samples, and pooled together followed by a single m6A-IP performed on pooled RNA samples instead of on a single sample. Thus, this multiplexed m6A-seq2 reduces the requirements of input materials, and also reducing technical variability which enables quantification of m6A levels across genes and samples.54 Recently, in order to explore m6A landscape of rare cell populations, we developed a highly sensitive and efficient super-low-input m6A sequence (SLIM-seq) by combining the advantages of m6A-LAIC-seq and SMART-seq.55 SLIM-seq focuses on decoding m6A modification on full-length transcript at the expense of the regional information of m6A peaks. Using in vitro-synthesized transcript Luciferase without m6A and GFP containing less than 0.5% m6A-modified adenosine that mimics the physiological level of m6A abundance, we confirm the consistent and comparable high efficiency of this strategy for different amounts of input, as few as 10 ng transcripts. Thus, a strong concordance was also observed using 5000 primary hematopoietic progenitor cells. Importantly, the inherent non-specific binding issue of m6A antibody is also taking into account in SLIM-seq, and adaptions in this approach significantly increase the specificity of SLIM-seq. Thus, SLIM-seq provides us a strategy to map transcriptome-wide m6A-tagged mRNAs for rare cells. As mentioned above, this method cannot provide the regional information of m6A peaks. SLIM-seq intends to decode m6A modification of whole transcript, which endows this strategy with high sensitivity for low-input. Notably, m6A largely determines mRNA abundance by regulating RNA decay and stability, its consequence on mRNA fate can be inferred by integrating expression data.
3.2. Antibody-independent m6A mapping
Antibody-based approach has some limitations including cross-reactivity to other RNA modifications, and limited utility for quantification of m6A stoichiometry. This raises several mapping strategies that are independent on m6A antibody. MAZTER-seq relies on the ability of the bacterial RNase MazF to cleave RNA at unmethylated sites occurring at ACA motifs, but not at the methylated counterparts m6A-CA.56 MAZTER-seq is able to systematically quantify m6A signal at single-nucleotide resolution. A same strategy was also applied in m6A-REF-seq.61 However, due to differential cleavage of by an RNase, this method only detects a small portion (16%–25%) of m6A sites.
APOBEC1 is a cytosine deaminase that targets DNA and RNA to induce cytosine-to-uracil (C-to-U) editing. By fusing APOBEC1 to the m6A-binding YTH domain of YTHDF2, m6A-adjacent cytidines could be induced to uracil by APOBEC1-YTH, which are detected using standard RNA-seq.57 This approach is named DART-seq (deamination adjacent to RNA modification targets), and is antibody-independent. DART-seq can detect about 79% of the edited mRNAs even with as little as 10 ng of total RNA as input. However, this approach only detects partial m6A signal that is recognized by YTH. In addition, the efficiency of APOBEC1-YTH in induing C-to-U transition in vivo or in vitro also need to be considered, and the specificity of YTH domain may also bring some biases.
Although the inert chemical property of RNA m6A poses a big obstacle to detect, many efforts attempt to alter this feature of m6A in developing antibody-free chemical approaches for m6A mapping. Recently, a metabolic labeling method, m6A-label-seq, is established to map m6A transcriptome-wide at base resolution. m6A-label-seq feeds cells with precursors of allyl-SeAM, Se-allyl-l-selenohomocysteine, to metabolically incorporate allyl-group into the adenosine sites (a6A) that are supposed to be m6A-modified.62 After enrichment with a6A antibody, a6A signals were transformed into N1, N6-cyclized adenosine (cyc-A) using the iodination-induced cyclization reaction. During reverse transcription, the cyc-A induced misincorporation. Therefore, the site can be detected through NGS. m6A-label-seq only can be used in cells and relies on the enrichment of a6A antibody. The number of identified m6A sites is limited, possibly due to the RNA a6A labeling yield. The incorporation efficiency of a6A need to be further improved. Considering that FTO oxidize m6A to hm6A in 5 minutes and then slowly oxidizes this hm6A to N6-formyladenosine, another strategy, m6A-SEAL, couples FTO’s enzymatic oxidation of m6A to hm6A with a DTT-mediated thiol-addition reaction to convert unstable hm6A into the more stable N6-dithiolsitolmethyladenosine (dm6A). These dm6A-marked RNA can be labeled by biotin and pulled down by streptavidin.59 Similarly, the oxidation efficiency of FTO need to be considered when using this method. In addition, both m6A-label-seq and m6A-SEAL lack stoichiometric information.
To detect and quantify m6A levels across the transcriptome at single-nucleotide resolution, m6A-SAC-seq is recently established. This method used the Dim1/KsgA family of dimethyltransferases, which transfer the methyl group from SAM to adenosines, converting m6A into N6,N6-dimethyladenosine (m62A) in consecutive methylation reactions.60 This method needs a low abundance of input materials (about ~30 ng of poly(A) or rRNA-depleted RNA), and can provide stoichiometric information of m6A modification. Although it seems that m6A-SAC-seq could overcome the current technological bottleneck for m6A mapping, the specificity and efficiency of Dim1 remains an important issue.
All of these main methods discussed above have their advantages and limitations. As this field continues to burst, we believe new methods for m6A mapping will appear. For instance, although DART-seq is recently applied to single cells,57 a new method that could quantitatively map m6A at single-cell level would be extremely exciting. In addition, it is necessary to establish standard criteria for bioinformatic analysis in this field. Some of controversial findings might be attributed, at least some certain, to differences in the computational strategies used by different studies. Recent analysis of published MeRIP-seq datasets show that, because of high rates of background signals, only about 30% to 60% of m6A peak were reproducible. To improve the reliability in characterizing RNA modification, in vitro transcribed RNA product is introduced as negative control to reduce the false positive resulting from sequencing bias or RNA structure.63 Overall, each method established so far for m6A mapping definitely contributes to the fast advances of this field.
4. RNA m6A IN AML
In recent years, the roles and molecular mechanisms of RNA m6A in different physiological and pathological conditions have been widely uncovered. Numerous studies have shown that methyltransferase complex components (METTL3, METTL14, WTAP), demethylases (FTO, ALKBH5), and the m6A binding protein (YTHDF2, YTHDC1, and IGF2BPs) are all highly expressed in AML. m6A regulators can function as oncogenes by targeting corresponding oncogenes, which are closely related to the occurrence, development, clinical treatment, and prognosis of AML. Hereby, we focus on AML and discuss current main advances in this field (Fig. 3).
Figure 3.
The roles and mechanisms of m6A modification in AML. m6A modification dysregulation can enhance AML leukemogenesis. m6A regulators act as oncogenes affects cell apoptosis, cell cycle, cell differentiation or LSC self-renewal via distinct mechanism. AML = acute myeloid leukemia, LSC = leukemia stem cell.
4.1. Roles and mechanisms of m6A in AML
FTO is the first m6A modifier that is reported to play an oncogenic role in AML.64 High expression of FTO is observed in leukemia cells from different subtypes of AML. Knockout or inhibition of FTO could inhibit the self-renewal of LSCs, and impair the AML development. Mechanistically, FTO controls the degradation of ASB2 and RARA mRNA in an m6A-dependent manner.64 As FTO is a α-ketoglutarate-dependent dioxygenase, its activity can be competitively inhibited by R-2-hydroglutarate (R-2HG), which is structurally closed to α-ketoglutarate. Inhibiting FTO by R-2HG was found increasing the overall level of m6A without affecting FTO expression and decreasing the stability of MYC and CEBPA mRNA in R-2HG-sensitive leukemia cells.65 R-2HG also abrogates FTO/m6A/YTHDF2-mediated upregulation of 2 critical glycolytic genes phosphofructokinase platelet (PFKP) and lactate dehydrogenase B (LDHB), thereby suppressing the glycolysis of leukemia cells.66
Subsequently, the roles of METTL3 and METTL14 in AML were also revealed.67–69 METTL3 deletion inhibits cell growth, promotes differentiation and apoptosis, and significantly prolongs survival in murine AML xenografts. These effects are mainly mediated by altering translation of c-MYC, BCL2, and PTEN mRNA in an m6A-dependent manner.67,68 In addition, mechanistically, METTL3 alone could bind chromatin and localize to transcriptional start site (TSS) of active genes. Promoter-bound METTL3 recruits CEBPZ and regulates the translation of downstream oncogenic drivers SP1 and SP2, subsequently regulating the expression of c-MYC.68 METTL14 exerts its oncogenic role by regulating MYB and MYC mRNA degradation and translation through m6A modification, and also plays a critical role in AML development and maintenance.69 WTAP was found overexpressed in AML patients, which are inversely correlated with overall survival in patients with AML. Depletion of WTAP suppresses cell proliferation, cell cycle, differentiation, and colony formation in AML cells. WTAP has an important role in AML oncogenesis, which is a novel client protein of Hsp90.70 Thus, these works provide rationale for FTO/METTL3/METTL14/WTAP as potential therapeutic targets for AML treatment.
Recently, we reported that ALKBH5 is required for the development and maintenance of AML and self-renewal of LSCs but not essential for normal hematopoiesis. Mechanistically, KDM4C regulates ALKBH5 expression via increasing chromatin accessibility of ALKBH5 locus, by reducing H3K9me3 level and promoting recruitment of MYB and Pol II. Moreover, ALKBH5 affects mRNA stability of receptor tyrosine kinase AXL in an m6A-dependent way.71 Complementary findings also identified the crucial role of another ALKBH5 for the self-renewal and maintenance of LSCs in AML. They found that ALKBH5 may exert tumor-promoting effects in AML by post-transcriptional regulation of a critical target, TACC3, a prognosis-associated oncogene in various cancers.72 Therefore, these 2 studies clearly uncover the selective and important role of ALKBH5 in the pathogenesis of AML.
The roles of m6A readers or related factors in leukemogenesis are also being recognized. Knockout of YTHDC1 severely impedes the development and maintenance of AML as well as LSC self-renewal in mice. Mechanistically, YTHDC1 regulates leukemogenesis through MCM4, which is a critical regulator of DNA replication.73 YTHDF2 deletion extends the half-life of m6A-modified transcripts including Tnfrsf2 to selectively compromise AML initiation and propagation without harming normal hematopoiesis.74 Recently, we reported that YBX1 interacts with IGF2BPs to stabilize m6A-modified transcripts, including MYC and BCL2. Upon YBX1 loss, MYC and BCL2 undergo accelerated decay, thus compromising AML cells.48 The role of IGF2BPs in leukemia is also being uncovered recently. Higher expression of IGF2BP2 predicts poor prognosis in AML patients,75 implying an important role of IGF2BP2 in AML, which need to be investigated. Deletion of IGF2BP1 affects proliferation, promotes myeloid differentiation, and decreases tumorigenic potential of AML cells by regulating HOXB4, MYB, and ALDH1A1.76 IGF2BP3 is also required for AML cell survival in an m6A-dependent manner, and IGF2BP3 loss significantly induces AML cell apoptosis, inhibits proliferation, and attenuates the ability of AML cells to develop leukemia in vitro and in vivo. Mechanistically, IGF2BP3 interacts with RCC2 mRNA and stabilizes the expression of modified m6A RNA.47,77 Taken together, these studies uncover the similar functional roles of different m6A modifiers (writers, erasers, and readers) via totally distinct mechanisms.
4.2. Identifying inhibitors of m6A modifiers for AML treatment
Studies discussed above have provide rationale for targeting some m6A modifiers as a potential therapeutic strategy in AML treatment, which drive seeking for their inhibitors. Several inhibitors targeting FTO and METTL3 are currently being identified. A selective small molecule inhibitor STM2457 of METTL3 is identified by a high throughput screening of 250,000 diverse drug-like compounds. STM2457 is able to bind to the SAM binding site of METTL3, which is structurally different from SAM and other known methyltransferase inhibitors. STM2457 does not recognize the homocysteine binding pocket used by SAM, but binds to the K513 site which is the known structural diversity of the cofactor-binding site of SAM-dependent methyltransferases. STM2457 also shows significant anti-leukemic effects in preclinical AML models.48,78 In addition, FB23 and FB23-2 is found to directly bind to FTO, and selectively inhibit FTO’s m6A demethylase activity. They also effectively inhibit AML progression in AML models.79 Although FB23-2 had a statistically significant effect on inhibiting the development of primary AML in mice, the inhibition rate for the compound was not satisfactory. Another 2 molecules, CS1 and CS2, have been screened out and identified as effective inhibitors of FTO, which can suppress its m6A demethylase activity by occupying the catalytic pocket and interfering with the binding of FTO with m6A-modified RNAs and then exert potent anti-leukemic efficacy in vivo and in vitro.80 The potent anti-tumor efficacy and minimal side effects of CS1 and CS2 suggest that they are highly feasible for clinical application. Thus, these findings highlight the promising potential for AML treatment by targeting of m6A modifiers. However, given the critical role of m6A modification under the physiological conditions, it is critical to carefully assess both the efficacy and safety of these inhibitors in the future.
5. CONCLUSION AND PERSPECTIVES
The biology of RNA modification has attracted tons of interests. It becomes clear that RNA RNA m6A plays critical roles in the pathogenesis of AML. Meanwhile, more and more very interesting scientific questions appear. For instance, given RNA m6A modification seems function as an adaptive-response system, how does this system sense and respond to various environments? How are the activities and expression of m6A modifiers regulated upon different responses? Our recent work found that KDM4C and MYB are upstream regulators of ALKBH5 in AML,71 it is of great interest to investigate how other m6A modifiers are dysregulated in leukemia in the future. Second, what determines the transcript target specificity of m6A modifiers? It remains unknown how the transcripts are selected by m6A modifiers. Our recent work implies that cofactors or RBPs might be recruited to regulatory machineries and play key roles in determining their target specificity. To be more exciting, developing better methods to comprehensively and quantitatively map m6A landscape in a single cell level will be necessary in the future. Finally, it is very hopeful that targeting RNA m6A modification would be a novel and attractive therapeutic approach for AML.
ACKNOWLEDGMENTS
This work is supported by grants to H.Z. from Medical Science Advancement Program (Basic Medical Sciences) of Wuhan University (TFJC2018005) and from the Fundamental Research Funds for the Central Universities (2042021kf0225).
We acknowledge the members of our laboratory for helpful discussion.
Footnotes
R.Y., Y.L. contributed equally to this study.
R.Y., Y.L., and H.Z. wrote the manuscript. W.T. and F.Z. contributed to discussing and revising this manuscript. All authors contributed to the article and approved the submitted version.
The authors declare that they have no conflict of interest.
We acknowledge the members of our laboratory for helpful discussion. This work is supported by grants to H.Z. from Medical Science Advancement Program (Basic Medical Sciences) of Wuhan University (TFJC2018005) and from the Fundamental Research Funds for the Central Universities (2042021kf0225).
REFERENCES
- [1].Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 2008;132(4):631–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Guo W, Lasky JL, Chang CJ, et al. Multi-genetic events collaboratively contribute to Pten-null leukaemia stem-cell formation. Nature 2008;453(7194):529–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Short NJ, Ravandi F. Acute myeloid leukemia: past, present, and prospects for the future. Clin Lymphoma Myeloma Leuk 2016(16 Suppl):S25–S29. [DOI] [PubMed] [Google Scholar]
- [4].Chen X, Glytsou C, Zhou H, et al. Targeting mitochondrial structure sensitizes acute myeloid leukemia to venetoclax treatment. Cancer Discov 2019;9(7):890–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood 2016;127(1):53–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Yamashita M, Dellorusso PV, Olson OC, Passegue E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat Rev Cancer 2020;20(7):365–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Suda T, Takubo K, Semenza GL. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 2011;9(4):298–310. [DOI] [PubMed] [Google Scholar]
- [8].van Velthoven CTJ, Rando TA. Stem cell quiescence: dynamism, restraint, and cellular idling. Cell Stem Cell 2019;24(2):213–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Culp-Hill R, D’Alessandro A, Pietras EM. Extinguishing the embers: targeting AML metabolism. Trends Mol Med 2021;27(4):332–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Challen GA, Sun D, Jeong M, et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat Genet 2011;44(1):23–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Moran-Crusio K, Reavie L, Shih A, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell 2011;20(1):11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Welch JS, Ley TJ, Link DC, et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 2012;150(2):264–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Rao RC, Dou Y. Hijacked in cancer: the KMT2 (MLL) family of methyltransferases. Nat Rev Cancer 2015;15(6):334–346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Harris WJ, Huang X, Lynch JT, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell 2012;21(4):473–487. [DOI] [PubMed] [Google Scholar]
- [15].Chen CW, Koche RP, Sinha AU, et al. DOT1L inhibits SIRT1-mediated epigenetic silencing to maintain leukemic gene expression in MLL-rearranged leukemia. Nat Med 2015;21(4):335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Abdel-Wahab O, Adli M, LaFave LM, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 2012;22(2):180–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Basheer F, Giotopoulos G, Meduri E, et al. Contrasting requirements during disease evolution identify EZH2 as a therapeutic target in AML. J Exp Med 2019;216(4):966–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Yang Y, Hsu PJ, Chen YS, Yang YG. Dynamic transcriptomic m(6)A decoration: writers, erasers, readers and functions in RNA metabolism. Cell Res 2018;28(6):616–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell 2012;149(7):1635–1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Batista PJ, Molinie B, Wang J, et al. m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 2014;15(6):707–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Wang P, Doxtader KA, Nam Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell 2016;63(2):306–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Wang X, Feng J, Xue Y, et al. Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature 2016;534(7608):575–578. [DOI] [PubMed] [Google Scholar]
- [23].Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 2014;10(2):93–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Huang J, Dong X, Gong Z, et al. Solution structure of the RNA recognition domain of METTL3-METTL14 N(6)-methyladenosine methyltransferase. Protein Cell 2019;10(4):272–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Ping XL, Sun BF, Wang L, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res 2014;24(2):177–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Qin T, Cheng Y, Wang X. RNA-binding proteins as drivers of AML and novel therapeutic targets. Leuk Lymphoma 2022:1–13. [DOI] [PubMed] [Google Scholar]
- [27].Knuckles P, Lence T, Haussmann IU, et al. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m(6)A machinery component Wtap/Fl(2)d. Genes Dev 2018;32(5-6):415–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Wen J, Lv R, Ma H, et al. Zc3h13 regulates nuclear RNA m(6)A methylation and mouse embryonic stem cell self-renewal. Mol Cell 2018;69(6):1028–1038.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Patil DP, Chen CK, Pickering BF, et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature 2016;537(7620):369–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Yue Y, Liu J, Cui X, et al. VIRMA mediates preferential m(6)A mRNA methylation in 3’UTR and near stop codon and associates with alternative polyadenylation. Cell Discov 2018;4:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Chen J, Wei X, Wang X, et al. TBK1-METTL3 axis facilitates antiviral immunity. Cell Rep 2022;38(7):110373. [DOI] [PubMed] [Google Scholar]
- [32].Pendleton KE, Chen B, Liu K, et al. The U6 snRNA m(6)A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 2017;169(5):824–835.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Mendel M, Delaney K, Pandey RR, et al. Splice site m(6)A methylation prevents binding of U2AF35 to inhibit RNA splicing. Cell 2021;184(12):3125–3142.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Warda AS, Kretschmer J, Hackert P, et al. Human METTL16 is a N(6)-methyladenosine (m(6)A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep 2017;18(11):2004–2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 2011;7(12):885–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Wei J, Liu F, Lu Z, et al. Differential m(6)A, m(6)Am, and m(1)A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell 2018;71(6):973–985.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2013;49(1):18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Shi H, Zhang X, Weng YL, et al. m(6)A facilitates hippocampus-dependent learning and memory through YTHDF1. Nature 2018;563(7730):249–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Du H, Zhao Y, He J, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun 2016;7:12626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res 2017;27(3):315–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Xu C, Wang X, Liu K, et al. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol 2014;10(11):927–929. [DOI] [PubMed] [Google Scholar]
- [42].Zhang Z, Theler D, Kaminska KH, et al. The YTH domain is a novel RNA binding domain. J Biol Chem 2010;285(19):14701–14710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Roundtree IA, Luo G-Z, Zhang Z, et al. YTHDC1 mediates nuclear export of N-methyladenosine methylated mRNAs. Elife 2017;6:e31311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Xiao W, Adhikari S, Dahal U, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell 2016;61(4):507–519. [DOI] [PubMed] [Google Scholar]
- [45].Hsu PJ, Zhu Y, Ma H, et al. Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res 2017;27(9):1115–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Huang H, Weng H, Sun W, et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol 2018;20(3):285–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Tran TM, Philipp J, Bassi JS, et al. The RNA-binding protein IGF2BP3 is critical for MLL-AF4-mediated leukemogenesis. Leukemia 2022;36(1):68–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Feng M, Xie X, Han G, et al. YBX1 is required for maintaining myeloid leukemia cell survival by regulating BCL2 stability in an m6A-dependent manner. Blood 2021;138(1):71–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Alarcon CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell 2015;162(6):1299–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Xu W, He C, Kaye EG, et al. Dynamic control of chromatin-associated m(6)A methylation regulates nascent RNA synthesis. Mol Cell 2022;82(6):1156–1168.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 2012;485(7397):201–206. [DOI] [PubMed] [Google Scholar]
- [52].Linder B, Grozhik AV, Olarerin-George AO, Meydan C, Mason CE, Jaffrey SR. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods 2015;12(8):767–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Molinie B, Wang J, Lim KS, et al. m(6)A-LAIC-seq reveals the census and complexity of the m(6)A epitranscriptome. Nat Methods 2016;13(8):692–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Dierks D, Garcia-Campos MA, Uzonyi A, et al. Multiplexed profiling facilitates robust m6A quantification at site, gene and sample resolution. Nat Methods 2021;18(9):1060–1067. [DOI] [PubMed] [Google Scholar]
- [55].Yin R, Chang J, Li Y, et al. Differential m(6)A RNA landscapes across hematopoiesis reveal a role for IGF2BP2 in preserving hematopoietic stem cell function. Cell Stem Cell 2022;29(1):149–159.e7. [DOI] [PubMed] [Google Scholar]
- [56].Garcia-Campos MA, Edelheit S, Toth U, et al. Deciphering the “m(6)A Code” via antibody-independent quantitative profiling. Cell 2019;178(3):731–747.e16. [DOI] [PubMed] [Google Scholar]
- [57].Meyer KD. DART-seq: an antibody-free method for global m(6)A detection. Nat Methods 2019;16(12):1275–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Wang Y, Xiao Y, Dong S, Yu Q, Jia G. Antibody-free enzyme-assisted chemical approach for detection of N(6)-methyladenosine. Nat Chem Biol 2020;16(8):896–903. [DOI] [PubMed] [Google Scholar]
- [59].Shu X, Cao J, Cheng M, et al. A metabolic labeling method detects m(6)A transcriptome-wide at single base resolution. Nat Chem Biol 2020;16(8):887–895. [DOI] [PubMed] [Google Scholar]
- [60].Hu L, Liu S, Peng Y, et al. m(6)A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat Biotechnol 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Zhang Z, Chen LQ, Zhao YL, et al. Single-base mapping of m(6)A by an antibody-independent method. Sci Adv 2019;5(7):eaax0250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Kim S, Kim NH, Park JE, et al. PRMT6-mediated H3R2me2a guides Aurora B to chromosome arms for proper chromosome segregation. Nat Commun 2020;11(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Zhang Z, Chen T, Chen HX, et al. Systematic calibration of epitranscriptomic maps using a synthetic modification-free RNA library. Nat Methods 2021;18(10):1213–1222. [DOI] [PubMed] [Google Scholar]
- [64].Li Z, Weng H, Su R, et al. FTO plays an oncogenic role in acute myeloid leukemia as a N(6)-methyladenosine RNA demethylase. Cancer Cell 2017;31(1):127–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Su R, Dong L, Li C, et al. R-2HG exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell 2018;172(1-2):90–105.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Qing Y, Dong L, Gao L, et al. R-2-hydroxyglutarate attenuates aerobic glycolysis in leukemia by targeting the FTO/m(6)A/PFKP/LDHB axis. Mol Cell 2021;81(5):922–939.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Vu LP, Pickering BF, Cheng Y, et al. The N 6-methyladenosine (m 6 A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 2017;23(11):1369–1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Barbieri I, Tzelepis K, Pandolfini L, et al. Promoter-bound METTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature 2017;552(7683):126–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Weng H, Huang H, Wu H, et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m(6)A modification. Cell Stem Cell 2018;22(2):191–205.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Bansal H, Yihua Q, Iyer SP, et al. WTAP is a novel oncogenic protein in acute myeloid leukemia (vol 28, pg 1171, 2014). Leukemia 2014;28(12):11712427–11711174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Wang J, Li Y, Wang P, et al. Leukemogenic chromatin alterations promote AML leukemia stem cells via a KDM4C-ALKBH5-AXL signaling axis. Cell Stem Cell 2020;27(1):81–97.e8. [DOI] [PubMed] [Google Scholar]
- [72].Shen C, Sheng Y, Zhu AC, et al. RNA demethylase ALKBH5 selectively promotes tumorigenesis and cancer stem cell self-renewal in acute myeloid leukemia. Cell Stem Cell 2020;27(1):64–80.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Sheng Y, Wei J, Yu F, et al. A critical role of nuclear m6A reader YTHDC1 in leukemogenesis by regulating MCM complex-mediated DNA replication. Blood 2021;138(26):2838–2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Mapperley C, van de Lagemaat LN, Lawson H, et al. The mRNA m6A reader YTHDF2 suppresses proinflammatory pathways and sustains hematopoietic stem cell function. J Exp Med 2021;218(3):e20200829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].He X, Li W, Liang X, et al. IGF2BP2 overexpression indicates poor survival in patients with acute myelocytic leukemia. Cell Physiol Biochem 2018;51(4):1945–1956. [DOI] [PubMed] [Google Scholar]
- [76].Elcheva IA, Wood T, Chiarolanzio K, et al. RNA-binding protein IGF2BP1 maintains leukemia stem cell properties by regulating HOXB4, MYB, and ALDH1A1. Leukemia 2020;34(5):1354–1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Zhang N, Shen Y, Li H, et al. The m6A reader IGF2BP3 promotes acute myeloid leukemia progression by enhancing RCC2 stability. Exp Mol Med 2022;54(2):194–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Yankova E, Blackaby W, Albertella M, et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 2021;593(7860):597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Huang Y, Su R, Sheng Y, et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 2019;35(4):677–691 e610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Su R, Dong L, Li Y, et al. Targeting FTO suppresses cancer stem cell maintenance and immune evasion. Cancer Cell 2020;38(1):79–96 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]



