Abstract
N6-methyladenosine (m6A) modification is found the most prevalent and abundant post-transcriptional mRNA modification in eukaryotic cells. It regulates almost all stages of RNA life cycle including splicing, translocation, stability, decay and translation. As a dynamic and reversible process, m6A modification is catalyzed by the RNA methyltransferases (‘writers’), removed by the demethylases (‘erasers’), and interacts with m6A-binding proteins (‘readers’). Recent studies have revealed that these m6A modification regulators are frequently expressed aberrantly in various types of cancer, and involved in cell proliferation, differentiation, metabolism, particularly, in tumorigenesis and tumor progression through diverse mechanisms. In this review, the m6A modification process and its regulatory functions in lung cancer are summarized. Furthermore, the research progress in the inhibitor development of m6A modification, and the potential of targeting m6A modifying proteins for clinical application are discussed.
Keywords: Eraser, Lung cancer, N6-methyladenosine, Reader, Writer
Introduction
Lung cancer is the most common cancer in the world (∼11.6% of total cases), with the highest mortality (18.4% of all cancer deaths). Lung cancer has become one of the most serious threats to human health.1,2 According to the histological characteristics, lung cancer can be classified into two major subtypes: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Among them, NSCLC accounts for 80%–85% of total lung cancer cases. NSCLC can be further classified into three types: adenocarcinoma, squamous cell carcinoma and large cell carcinoma. The 5-year survival rate of NSCLC patients is only 15%. The occurrence and progression of lung cancer are complicated and consist of several steps. Understanding of molecular mechanisms of carcinogenesis, identification of biomarkers for diagnosis and prognosis, and the development of novel therapy strategies for lung cancer are urgently needed.
The post-transcriptional modifications have been proved to play critical roles in a variety of physiological and pathological processes. Among numerous types of RNA modifications, N6-methyladenosine (m6A), which was first identified in the 1970s,3, 4, 5 is the most prevalent and abundant internal mRNA modification in eukaryotic cells.6,7 Similar to the methylation modification in DNA, the m6A RNA methylation regulates the post-transcriptional expression of genes without changing the base sequence. It has been found that m6A mainly occurring within the “RRACH” consensus sequence (R = A or G, H = A, C, or U), which is enriched in the stop codon, 3’ untranslated region (UTR) and long internal exon.8, 9, 10
Studies have revealed that m6A modification participates in almost all stages of RNA life cycle including mRNA splicing, exportation, stabilization and translation.11, 12, 13, 14 Besides mRNA, the m6A methylation also regulates the generation and function of ribosomal RNA (rRNA), transfer RNA (tRNA), and non-coding RNAs (ncRNA) including long non-coding RNA (lncRNA), microRNA (miRNA) and circular RNAs (circRNA). Recently, m6A has been reported to be present on some special regulatory RNAs. Liu et al revealed that METTL3 may deposit m6A modifications on chromosome-associated regulatory RNAs (carRNAs), such as enhancer RNAs, promoter-associated RNAs, and repeat RNAs. Depletion of METTL3 reduces m6A modification and promotes open chromatin state and downstream RNA transcription.15
In nucleus, the m6A modification of RNA is dynamically and reversibly regulated by two groups of catalytic proteins: the methyltransferases (also called “writers”), and the demethylases (also called “erasers”).16, 17, 18 A group of m6A binding proteins (also called “readers”) subsequently recognize and bind to the m6A-rich domain in RNA, and perform corresponding downstream functional processes.
Numerous studies have demonstrated that aberrant m6A methylation closely correlate with tumorigenesis and progression of human cancers through diverse mechanisms.19, 20, 21 The dysregulations of writers, erasers and readers are proved to regulate these processes by activating oncogenes or inhibiting tumor suppressors.22,23 Recent studies have also revealed that dysregulated m6A methylation plays critical roles in lung cancer.24,25 In this review, we summarized the latest research progress in the function and underlying mechanism of m6A methylation in lung cancer, and discussed the potential of targeting m6A modifying proteins for cancer therapy.
Regulators of RNA m6A methylation
There are mainly 3 classes of m6A methylation regulators: writers, erasers and readers. The cross-talks among them are involved in tumorigenesis and tumor progression (Fig. 1).
Figure 1.
Mechanism of RNA m6A modification.
Writers
The identified m6A writers consist of methyltransferase-like protein 3 (METTL3), METTL14, METTL16, METTL5 and their cofactors Wilms tumor 1associated protein (WTAP), RNA-binding motif protein 15 (RBM15/15B), Vir-like m6A methyltransferase associated (VIRMA; also known as KIAA1429), zinc finger CCCH-type containing 13 (ZC3H13) and Cbl proto-oncogene-like 1 (CBLL1; also known as HAKAI).
m6A is introduced co-transcriptionally by the methyltransferase complex (MTC) which consists of METTL3 and other accessory components including METTL14, WTAP, RBM15, VIRMA and ZC3H13.26 METTL3 is the first identified component of the m6A MTC. It serves as the primary methyltransferase critical for m6A methylation and is highly conserved in eukaryotic cells from yeast to human. METTL3 is a sadenosyl methionine (SAM)-binding protein which catalyzes the transfer of methyl groups in SAM to adenine bases in RNA and produces sadenosyl homocysteine (SAH). Aberrant expression of METTL3 affects the total level of m6A methylation.10,27
METTL14 is another active component in the m6A MTC. Studies have revealed that METTL3 and METTL14 are co-localized in nuclear speckles and form stable heterodimer.28 In m6A MTC, only METTL3 acts as a catalyst, whereas METTL14 primarily acts to form a stable structure with METTL3 and plays a key role in recognizing specific sequences in catalytic substrates.28, 29, 30 METTL3-METTL14 heterodimer forms the core MTC inducing m6A modification synergistically.
Similar to METTL14, WTAP has no methyltransferase activity. Its major function is to ensure the METTL3-METTL14 heterodimer could local to the nuclear speckle.31,32 RBM15 and RBM15B also have no catalytic function. They bind to METTL3 and WTAP, and recruit the MTC to specific RNA sites for m6A modification.33,34 VIRMA/KIAA1429 mediates region-selective m6A methylation of adenine bases in 3′UTR and near stop codon by recruiting the MTC, and also interacts with cleavage and polyadenylation specificity factor subunit 5 and 6 (CPSF5 and CPSF6).35 ZC3H13 contains abundant low-complexity domains. By bridging WTAP to the mRNA-binding factor Nito, ZC3H13 promotes the m6A MTC retained in nuclear speckles and improves its catalytic function.34,36 ZCCHC4, another CCHC zinc-finger-containing protein, is found to involve in the modification of the 28S rRNA, mediates rRNA ribosome subunit distribution and global translation.37
METTL16 is a newly discovered m6A writer which possesses methyltransferase activity. Shima et al showed that METTL16 regulates mRNA stability and splicing, and the binding sites of METTL16 do not overlap with those of METTL3/METTL14 methylation complexes, suggesting its independent functions.38 Pendleton et al found that METTL16 directly binds to pre-RNA and regulates the splicing of SAM synthetase.39 In addition, METTL16 could function alone and catalyze U6-snRNA m6A methylation on A43 box (“ACAGAGA”) and regulate tumorigenesis by targeting pre-mRNAs and ncRNAs.40
Another new methyltransferase responsible for 18S rRNA m6A modification, METTL5, is identified in 2019.41,42 By forming a heterodimer with a co-activator TRMT112, the metabolic stability of METTL5 and modification area on precursor and mature forms of 18S rRNA is increased. The structure of METTL5-TRMT112 heterodimer suggests its different RNA binding mode from other m6A writers. Furthermore, co-immunoprecipitation studies revealed that there are more than 100 proteins may bind to METTL3 or METTL14,43 suggesting there may be other components of the m6A methyltransferase complexes exist.
Erasers
The demethylases can remove m6A in RNA and called as ‘erasers’. To date, there are only two m6A erasers have been identified, the fat mass and obesity-associated protein (FTO) and the AlkB homolog 5 (ALKBH5). These two proteins are all belong to the alpha-ketoglutarate-dependent dioxygenase family and catalyze m6A demethylation in a Fe (II) and α-ketoglutaric acid-dependent manner. However, the demethylases activities of them are independent of each other. These two proteins are mainly localized in the nucleus where the removal of m6A mainly occurs. Whether there is any other m6A demethylase located in the cytoplasm and how the m6A is modified in the cytoplasm is still not clear.
FTO is the first molecule identified to catalyze m6A demethylation, and highly expressed in muscle and brain. It mediates demethylation with its oxidative activity by targeting the m6A-rich region in RNA.44,45 Rau et al investigated the mechanism of sequence-specific m6A demethylation of FTO by fusing FTO and RCas9 together as an RNA targeting module. This ingeniously designed RCas9-FTO retained the demethylation activity and bound to RNA in a sequence-specific manner depending on the single-guide RNA (sgRNA) and PAMmer.46 The study of FTO function confirms that the m6A modification is a dynamic and reversible process. Accumulating evidences show that FTO interacts with different RNA modifications, such as m6A, m6Am, m1A, 6 mA, 3 mT. Zhang et al investigated the role of FTO structure in FTO function in different RNA substrates. Their study showed that the catalytic activity of FTO depends on the interaction of residues in the catalytic pocket with the nucleobase, also depends on the sequence and the tertiary structure of RNA. They found that m6A is the most favorable substrate of FTO.47
ALKBH5 is the second identified m6A eraser and its expression is particularly abundant in the testes.48 ALKBH5 catalyzes m6A demethylation in RNA, also takes part in splicing and the formation of longer 3’ UTR mRNAs.49,50 Interestingly, the activity of ALKBH5 and FTO are similar but also specific. Zou et al illustrated that an m6A-induced conformational change which is not in the consensus sequence (GG (m6A)CU) in RNA may account for the specificity.51 Furthermore, FTO could mediate m6Am (N6, 2′-O-dimethyladenosine) demethylation while ALKBH5 is an m6A-specific demethylase.52 Recent studies reported that ALKBH3 is a novel m6A eraser, and it preferentially modifies tRNA than mRNA or rRNA.53
Readers
For different downstream biological functions, the m6A modification must be identified by interacting factors such as YTH domain-containing proteins (YTHDC1-2), YTH-family proteins (YTHDF1-3), heterogeneous nuclear ribonucleoproteins (including hnRNPC, hnRNPG and hnRNPA2B1) and insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs). These proteins are defined as ‘reader’.
The YT521-B homology (YTH) domain family members, including YTH domain family proteins (YTHDF1-3) and YTH domain containing proteins (YTHDC1-2), are the most important readers and have conserved m6A binding domains.54 In cytoplasm, YTHDF1-3 proteins work synergistically to influence RNA metabolism.45,54,55 YTHDF2 is the first identified m6A reader. It selectively binds to m6A modified mRNA and regulates RNA degradation. The C-terminal region of YTHDF2 recognizes specific m6A sites, and the N-terminal region binds to the SH domain of CCR4-NOT transcription complex subunit 1 (CNOT1), thus it could recruit the CCR4-NOT deadenylase complex and transport RNA to the processing body (P-body) to accelerate the degradation of m6A modified RNA.56 Different to YTHDF2, YTHDF1 binds to m6A sites close to stop codon in mRNA, and recruits the translation initiation complex (including eIF3, eIF4E, poly(A) binding protein (PABP)) and the 40S ribosomal subunit to promote translation of target RNA.45 YTHDF3 promotes RNA translation by cooperating with YTHDF1 and initiation factor eIF4A3, and mediates mRNA decay through direct interaction with YTHDF2.54,55,57
The main role of YTHDC1 is to regulate mRNA export. YTHDC1 exports m6A-containing mRNA from the nucleus to the cytoplasm through interacting with serine- and arginine-rich splicing factor 3 (SRSF3) and the splicing factor. When YTHDC1 is repressed, methylated mRNAs are accumulated in the nucleus.58 Lesbirel et al reported that YTHDC1 plays a synergistic role with the three prime repair exonuclease (TREX) mRNA export complex by interacting with SRSF3, and promotes the exportation of m6A methylated mRNA from the nucleus to cytoplasm.59 YTHDC1 also participates in the regulation of m6A methylation by regulating intracellular SAM synthesis.38 YTHDC2 enhances the translation efficiency of target mRNA. Studies have shown that YTHDC2 knockdown causes the upregulation of the m6A-modified transcripts, and this function of YTHDC2 is essential for fertility in mammals.60,61
The proteins of hnRNPs superfamily related to m6A modification consist of HNRNPA2B1, HNRNPC and HNRNPG. HNRNPA2B1 participates in primary RNA (pri-miRNA) processing and alternative splicing by interacting with drosha ribonuclease III (DROSHA) and DiGeorge syndrome critical region 8 (DGCR8)62; whereas HNRNPC and HNRNPG selectively recognize m6A-induced mRNA secondary structures and regulate mRNA abundance and splicing.63,64
IGF2BP1/2/3 (insulin-like growth factor 2 mRNA-binding proteins 1, 2, and 3) is a new class of m6A reader. Different to the functions of YTHDF2, IGF2BPs enhance the stability and translation of their target mRNAs by recognizing the consensus GG (m6A)C sequence under normal and stressed conditions.65
Notably, because the interactions between m6A modifications and RNA-binding proteins are complicated, mRNA expressions are regulated at multiple levels.
Regulators of RNA m6A methylation in lung cancer
m6A methylation regulates RNA processing and metabolism and participates in carcinogenesis. On one hand, m6A-modification itself may lead to alterations of mRNA translation, acts oncogenic role and accelerate tumor progression. On the other hand, the dysregulation of m6A writers, erasers and readers also facilitates tumor development.21 Several studies also showed that m6A regulators influence the prognosis of lung cancer patients.66,67
Writers
m6A writers deposit m6A on mRNA and non-coding RNAs, regulates the expression of oncogene and tumor suppressor gene, and play critical role in lung cancer initiation and progression.
Numerous studies showed that METTL3 acts as an oncogene in lung cancer; it promotes lung cancer progression via diverse mechanisms. (a) Regulation on gene translation. METTL3 stimulates the translation of epidermal growth factor receptor (EGFR) and tafazzin (TAZ), promotes the growth, survival and invasion of lung cancer cells.24 By direct interaction with the eukaryotic translation initiation factor 3 subunit h (eIF3h), METTL3 promotes translation of many oncogenic mRNAs such as bromodomain-containing protein 4 (BRD4).68 Jin et al reported that METTL3 promotes yes-associated protein (YAP) translation and increases YAP activity which leads to NSCLC drug resistance and metastasis.69 Studies also demonstrate that METTL3 may repress gene expression in lung cancer cells. In cigarette smoke extract (CSE) treated human bronchial epithelial (HBE) cells, METTL3 level is elevated. METTL3 introduces m6A modification to Zinc finger and BTB domain-containing 4 (ZBTB4) to reduce ZBTB4 expression, which is responsible to CSE induced epithelial–mesenchymal transition (EMT) in lung cancer cells.70 (b) Regulation on miRNA biogenesis. METTL3 increases the splicing of precursor miR-143–3p to stimulate its biogenesis. Increased miR-143–3p accelerates bone metastasis of lung cancer by targeting vasohibin-1 (VASH1).71 (c) Regulation on mRNA and lncRNA stability. METTL3 enhances the mRNA stability of JUNB, a transcriptional regulator of EMT, and contributes to TGF-β induced EMT in lung cancer cells.72 LncRNA ABHD11-AS1 is upregulated in NSCLC cells; it stimulates the proliferation and Warburg effect of NSCLC cells. Further study illustrates that METTL3 enhances the transcript stability of ABHD11-AS1 to increase its expression.73 (d) Regulation on signaling pathway. MiR-600 inhibits migration and proliferation of lung cancer cells. The mechanistic study revealed that miR-600 targets METTL3. The reduced METTL3 alters the expression and phosphorylation of members of PI3K/Akt signaling pathway.74
Interestingly, METTL3 itself is also modulated in lung cancer which affects its function. Du et al reported that METTL3 is SUMOylated at lysine residues K177, K211, K212 and K215. The SUMOylation represses METTL3 activity and reduces the m6A level of mRNA, resulting in enhanced tumor growth.75
For the role of other m6A writers in lung cancer, Shen et al reported that METTL14 modulates m6A level of NOTCH1 mRNA, increases the translation level of NOTCH1 and promotes NSCLC cell growth.76
Erasers
m6A erasers are involved in lung cancer through regulating m6A levels and mRNA stability of certain genes. FTO is identified as a prognostic factor in lung squamous cell carcinoma (LUSC). It facilitates proliferation and development of LUSC cells by decreasing the m6A levels in myeloid zinc finger 1 (MZF1) mRNA and enhancing its stability.77 In addition, Li et al revealed that high level of FTO promotes proliferation and colony formation of NSCLC cells by improving ubiquitin-specific peptidase 7 (USP7) mRNA stability and expression.78
ALKBH5 is another m6A eraser, it was found to promote the proliferation and invasion of lung adenocarcinoma cells under intermittent hypoxia (IH) conditions by down-regulating m6A modification of forkhead box M1 (FOXM1) mRNA and promoting FOXM1 expression.79 In NSCLC, upregulated ALKBH5 decreases the m6A level and promotes the expression of ubiquitin-conjugating enzyme E2C (UBE2C) leading to NSCLC progression.80 Although several studies demonstrated that ALKBH5 promotes lung cancer progression, ALKBH5 is also reported to suppress lung cancer. Jin et al revealed that ALKBH5 inhibits tumor growth and metastasis of NSCLC by reducing YTHDFs mediated YAP expression and inhibiting YAP activity.81 The opposite roles of ALKBH5 reported in lung cancer by different groups maybe due to the cell and animal models, the pathological stages, and study methods. Further investigation is needed to explore the complicity of lung cancer.
Readers
m6A readers play opposite roles in lung cancer. YTHDF2 was found up-regulated and promotes proliferation in lung cancer cells. By directly binding to the m6A-modified site in 6-phosphogluconate dehydrogenase (6-PGD), YTHDF2 promotes the translation of 6PGD mRNA and enhances the pentose phosphate pathway (PPP).82 IGF2BP1 promotes growth and invasion of lung cancer cells and associated with poor prognosis by impairing the miRNA directed decay of the serum response factor (SRF) mRNA.83 In contrast, readers also act as tumor suppressors. YTHDC2 inhibits lung adenocarcinoma tumorigenesis by suppressing cystine uptake and blocking downstream SLC7A11-dependent antioxidant program.84 Wang et al revealed that YTHDC2 was down-regulated in lung cancer. The overexpression of YTHDC2 inhibited the proliferation of lung cancer cells as well as tumor growth in nude mice. The mechanistic study showed that cylindromatosis (CYLD)/NF-κB pathways mediated YTHDC2's inhibitory effect.85
So far, most m6A modifications in lung cancer were found on mRNA, miRNA and lncRNA. Interestingly, recent study reported that in mouse embryonic stem cells, METTL3 introduces m6A modifications to regulatory RNAs such as enhancer RNAs, repeat RNAs. Since enhancer RNAs play important roles in cancer,86 whether m6A modification on enhancer RNAs is involved in lung cancer warrants further investigation.
The target genes, potential mechanisms and functions of related m6A regulators in lung cancer are summarized in Table 1.
Table 1.
Target genes, mechanisms and functions of m6A regulators in lung cancer.
Regulator | Target | Molecular mechanism | Function | Refs |
---|---|---|---|---|
METTL3 | EGFR and TAZ | Promotes the translation of EGFR and TAZ | Promotes growth, survival, and invasion of human lung cancer cells | 24 |
METTL3 | eIF3h and BRD4 | Interacts with eIF3h, and promotes the translation of BRD4 | METTL3-eIF3h interaction is required for enhanced translation, formation of densely packed polyribosomes and oncogenic transformation in human lung cancer cells. | 68 |
METTL3 | YAP | Promotes YAP translation via recruiting YTHDF1/3 and eIF3b to translation initiation complex, and increases YAP mRNA stability through regulating MALAT1-miR-1914-3p-YAP axis | Induces treatment resistance and metastasis in NSCLC cells. | 69 |
METTL3 | ZBTB4 | Reduce the expression of ZBTB4 | Promotes the cigarette smoke extract induced EMT in lung cancer cells | 70 |
METTL3 | VASH1 | Promotes the biogenesis of miR-143–3p, and inhibits the expression of VASH1 | Induces brain metastasis and angiogenesis of lung cancer cells | 71 |
METTL3 | JUNB | Enhances the mRNA stability of JUNB | Contributes to the TGF-β-induced EMT in lung cancer cells | 72 |
METTL3 | lncRNA ABHD11-AS1 | Enhances the transcript stability and expression of lncRNA ABHD11-AS1 | Promotes the proliferation and Warburg effect in NSCLC cells | 73 |
METTL3 | Bax/Bcl-2, PI3K/Akt pathway | Promotes PI3K/Akt signaling pathway | Knockdown of METTL3 inhibits the survival, proliferation and migration of NSCLC cells | 74 |
METTL14 | NOTCH1 | Reduces the stability of NOTCH1 mRNA, and regulates NOTCH1 expression with GPER | GPER could promote NSCLC cell growth via regulating the YAP1/circNOTCH1/m6A methylated NOTCH1 pathway | 76 |
FTO | MZF1 | Increases the mRNA stability of MZF1, leading to enhanced MZF1 expression | Facilitates the proliferation, invasion and the malignant phenotypes of lung squamous cells | 77 |
FTO | USP7 | Increases the mRNA stability of USP7 | Promotes the proliferation and the colony formation ability of lung cancer cells | 78 |
ALKBH5 | FOXM1 | Increases the translation efficiency of FOXM1 mRNA, and promote the expression | Promotes the proliferation and invasion of lung adenocarcinoma cells under intermittent hypoxia (IH) conditions | 79 |
ALKBH5 | UBE2C | Increases the mRNA stability of UBE2C and promotes the expression | Represses autophagy and aggravates cell proliferation, colonigenicity and invasive growth of NSCLC | 80 |
ALKBH5 | YAP | Reduces YTHDFs-mediated YAP expression and inhibits miR-107/LATS2-mediated YAP activity | Inhibits cellular proliferation, invasion, migration, and EMT of NSCLC cells in vitro and inhibits tumor growth and metastasis in vivo | 81 |
YTHDF2 | 6PGD | Promotes the translation of 6PGD mRNA and enhances the PPP | Promotes the growth of lung cancer cells | 82 |
IGF2BP1 | SRF | Increases the stability of SRF mRNA, and promotes the expression of SRF | Promotes the cell growth and invasion of lung cancer cells. | 83 |
YTHDC2 | SLC7A11 | Reduces the stability of SLC7A11 mRNA | Decreases tumorigenesis and exhibits antitumor activity in human LUAD | 84 |
YTHDC2 | CYLD | Inhibits the degradation of CYLD mRNA and promotes the expression of CYLD, then inhibits the NF-κB pathway activity | Promotes the proliferation and migration of lung cancer cells | 85 |
Abbreviations: ALKBH5: AlkB homolog 5; BRD4: bromodomain-containing protein 4; CYLD: cylindromatosis; EGFR: epidermal growth factor receptor; eIF3h: eukaryotic translation initiation factor 3h; EMT: epithelial–mesenchymal transition; FOXM1: forkhead box M1; FTO: fat mass and obesity-associated protein; GPER: G protein-couples oestrogen receptor; IGF2BPs: insulin-like growth factor 2 mRNA-binding proteins; LATS2: large tumor suppressor kinase 2; LUAD: human lung adenocarcinoma; MALAT 1: metastasis associated in lung adenocarcinoma transcript 1; METTL3: methyltransferase-like protein 3; METTL14: methyltransferase-like protein 14; MZF1: myeloid zinc finger 1; NSCLC: non-small cell lung cancer; PPP: pentose phosphate pathway; SRF: serum response factor; TAZ: tafazzin; TGF-β: transforming growth factor-β; UBE2C: ubiquitin-conjugating enzyme E2C; USP7: ubiquitin specific protease 7; VASH1: vasohibin-1; YAP: yes associated protein; YTHDC: YTH domain-containing proteins; YTHDF: YTH-family proteins; ZBTB4: Zinc finger and BTB domain-containing 4; 6PGD: 6-phosphogluconate dehydrogenase.
Gene expression profiles of m6A methylation regulator for diagnosis and prognosis
The dysregulated expression of m6A regulators can be used for potential clinical purpose. Li et al study showed that the expression of 19 m6A regulators was significantly different in lung cancer tissues, and identified a three-m6A-regulator signature (KIAA1429, METTL3, and IGF2BP1) as an independent prognostic model for patient stratification, prognostic assessment, and personalized treatment.87 Based on TCGA dataset analysis, Li et al found that a profile of ten m6A-associated regulators is significantly related to advanced lung cancer stage, they also showed that the loss of FTO and YTHDC2 is associated with poor overall survival (OS) rate.88 Shi et al reported that in NSCLC tissues, high level of YTHDF1 links to hypoxia adaptation and cancer progression.89 Similar researches were conducted in lung adenocarcinoma, risk scoring signatures contain 3, 6, or 11 m6A methylated genes were built for clinical diagnosis and prognosis by different research groups.90, 91, 92 These studies suggest that m6A regulators could be potential diagnostic markers for diagnosis and prognosis of lung cancer.
Therapeutic potential of targeting m6A methylation regulators
Since it is proved that the m6A methylation of RNA plays important roles in tumorigenesis and tumor progression, m6A methylation regulators could serve as potential therapeutic targets for drug development.
Several FTO inhibitors have been discovered, and FTO inhibitors suppress cell growth in various types of cancer. FTO inhibitors were firstly identified by structure based virtual screening and biochemical analyses. Among these inhibitors, the natural compound Rhein competitively binds to the FTO catalytic domain and exhibits effective inhibitory activity on m6A demethylation both in vivo and in vitro.93 Meanwhile, Yang et al reported that Rhein significantly reduces the viability, induces apoptosis and cell cycle arrest in NSCLC cells,94 however, the inhibitory effect of Rhein on FTO in NSCLC still needs further investigation. Meclofenamic acid (MA) can selectively inhibit FTO by competing binding to the surface of FTO active site, and increases the level of m6A modification in cells.95 As an ethyl ester derivative of MA, MA2 suppresses glioblastoma stem cell growth and self-renewal in vitro and reduces tumor growth in vivo.96 4-chloro-6-(6′-chloro-7′-hydroxy-2′,4′,4′-trimethyl-chroman-2′-yl) benzene-1,3-diol (CHTB) and N-(5-Chloro-2,4-dihydroxyphenyl)-1- phenylcyclobutanecarboxamide (N-CDPCB) have been identified as FTO inhibitors by virtual screening. Structural studies proved that CHTB binds to the surface of FTO active site, while N-CDPCB is sandwiched in FTO.97,98 A metabolite of mutant IDH1/2 enzymes, R-2-hydroxyglutarate (R-2HG), inhibits FTO activity and elevates m6A level by binding to FTO directly. It enhances the YTHDF2 mediated degradation of MYC and CEBPA, and causes cell growth inhibition, cell-cycle arrest and apoptosis of acute myelocytic leukemia (AML).99 In addition, R-2HG also exerts synergistic effects with other chemotherapy drugs against leukemia and glioma. Huang et al reported that another inhibitor of FTO, FB23-2 suppresses the proliferation and promotes differentiation of AML cells.100 Both FTO and ALKBH5 are α-ketoglutarate (α-KG) dependent dioxygenases. They are highly expressed in isocitrate dehydrogenases (IDH) mutant cancers and could be inhibited by D2-hydorxyglutarate (D2-HG) by competitive binding.101,102
Besides FTO inhibitors, Weng et al reported that hematopoietic transcription factor SPI1 inhibits METTL14 expression and malignant hematopoiesis by regulating the METTL14-MYB/MYC signaling axis.103 Zhang et al showed that a member of the carbonic anhydrases, carbonic anhydrase IV inhibits the tumorigenicity of colon cancer by inducing WTAP degradation and promoting the transcriptional activity of the Wilms' tumor 1 (WT1), which is an antagonist of WNT pathway.104 S-adenosylmethionine is a cofactor substrate in METTL3-METTL14 complex. By competing with adenosylmethionine, a derivate of S-adenosylmethionine, S-adenosylhomocysteine inhibits the activity of some methyltransferases.105
The development of inhibitors of m6A methylation regulators is still in its early stages. The improvement on specificity and efficacy of inhibitors are key issues, and this depends on further understanding of the mechanism of m6A modification.
Conclusions and future perspectives
Numerous studies have revealed that m6A RNA methylation controls RNA life including RNA transcription, splicing, processing, translation and decay. It also plays important roles in a broad range of biological processes such as cell proliferation, differentiation and embryonic development. In particular, m6A RNA methylation is often dysregulated in tumors, and plays crucial roles in tumorigenesis and tumor progression through regulating oncogenes and tumor suppressor genes.
Although great progress has been achieved, there are still many fields warrant further investigation: (a) the discovery of novel ‘writers’, ‘erasers’ and ‘readers’; (b) the study on the mechanism of RNA modification; (c) the development of quantitative technologies to detect RNA modification precisely; (d) the establishment of the profile of modified RNAs for cancer diagnosis and prognosis; (e) the development of novel anti-cancer drugs via targeting RNA modification. We hope the exploration of the mechanism of m6A modification and the development of inhibitor of m6A regulators will pave a new path for cancer therapy.
Author contributions
Qicheng Zhang selected literature, drafted the manuscript, and prepared the figure and table. Ke Xu designed this review, discussed and revised the manuscript. All authors contributed to this manuscript. All authors read and approved the final manuscript.
Conflict of interests
Authors declare no conflict of interests.
Funding
This work was supported by the National Natural Science Foundation of China (No. 81372519), the Project of Health Commission of Tianjin (No. TJWJ2021MS006), the Key Project of Natural Science Foundation of Tianjin (No. 18JCZDJC98500) and the Specialized Research Fund for the Doctoral Program of Higher Education of China (No. 20131202110005).
Footnotes
Peer review under responsibility of Chongqing Medical University.
References
- 1.Bray F., Ferlay J., Soerjomataram I., et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Chen W., Zheng R., Baade P.D., et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–132. doi: 10.3322/caac.21338. [DOI] [PubMed] [Google Scholar]
- 3.Desrosiers R., Friderici K., Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci U S A. 1974;71(10):3971–3975. doi: 10.1073/pnas.71.10.3971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Adams J.M., Cory S. Modified nucleosides and bizarre 5'-termini in mouse myeloma mRNA. Nature. 1975;255(5503):28–33. doi: 10.1038/255028a0. [DOI] [PubMed] [Google Scholar]
- 5.Perry R.P., Kelley D.E., Friderici K., et al. The methylated constituents of L cell messenger RNA: evidence for an unusual cluster at the 5' terminus. Cell. 1975;4(4):387–394. doi: 10.1016/0092-8674(75)90159-2. [DOI] [PubMed] [Google Scholar]
- 6.Jia G., Fu Y., He C. Reversible RNA adenosine methylation in biological regulation. Trends Genet. 2013;29(2):108–115. doi: 10.1016/j.tig.2012.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Yue Y., Liu J., He C. RNA N6-methyladenosine methylation in post-transcriptional gene expression regulation. Genes Dev. 2015;29(13):1343–1355. doi: 10.1101/gad.262766.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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: 10.1038/nature11112. [DOI] [PubMed] [Google Scholar]
- 9.Meyer K.D., Saletore Y., Zumbo P., et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell. 2012;149(7):1635–1646. doi: 10.1016/j.cell.2012.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bokar J.A., Shambaugh M.E., Polayes D., et al. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA. 1997;3(11):1233–1247. [PMC free article] [PubMed] [Google Scholar]
- 11.Liu Q., Gregory R.I. RNAmod: an integrated system for the annotation of mRNA modifications. Nucleic Acids Res. 2019;47(W1):W548–W555. doi: 10.1093/nar/gkz479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Genenncher B., Durdevic Z., Hanna K., et al. Mutations in cytosine-5 tRNA methyltransferases impact mobile element expression and genome stability at specific DNA repeats. Cell Rep. 2018;22(7):1861–1874. doi: 10.1016/j.celrep.2018.01.061. [DOI] [PubMed] [Google Scholar]
- 13.Ke S., Alemu E.A., Mertens C., et al. A majority of m6A residues are in the last exons, allowing the potential for 3' UTR regulation. Genes Dev. 2015;29(19):2037–2053. doi: 10.1101/gad.269415.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ke S., Pandya-Jones A., Saito Y., et al. m(6)A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev. 2017;31(10):990–1006. doi: 10.1101/gad.301036.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Liu J., Dou X., Chen C., et al. N (6)-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science. 2020;367(6477):580–586. doi: 10.1126/science.aay6018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dai D., Wang H., Zhu L., et al. N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 2018;9(2):124. doi: 10.1038/s41419-017-0129-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Batista P.J. The RNA modification N(6)-methyladenosine and its implications in human disease. Dev Reprod Biol. 2017;15(3):154–163. doi: 10.1016/j.gpb.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wang Y., Li Y., Toth J.I., et al. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 2014;16(2):191–198. doi: 10.1038/ncb2902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jaffrey S.R., Kharas M.G. Emerging links between m(6)A and misregulated mRNA methylation in cancer. Genome Med. 2017;9(1):2. doi: 10.1186/s13073-016-0395-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wang S., Sun C., Li J., et al. Roles of RNA methylation by means of N(6)-methyladenosine (m(6)A) in human cancers. Cancer Lett. 2017;408:112–120. doi: 10.1016/j.canlet.2017.08.030. [DOI] [PubMed] [Google Scholar]
- 21.Chen X.Y., Zhang J., Zhu J.S. The role of m(6)A RNA methylation in human cancer. Mol Cancer. 2019;18(1):103. doi: 10.1186/s12943-019-1033-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tuncel G., Kalkan R. Importance of m N(6)-methyladenosine (m(6)A) RNA modification in cancer. Med Oncol. 2019;36(4):36. doi: 10.1007/s12032-019-1260-6. [DOI] [PubMed] [Google Scholar]
- 23.Chen B., Li Y., Song R., et al. Functions of RNA N6-methyladenosine modification in cancer progression. Mol Biol Rep. 2019;46(1):1383–1391. doi: 10.1007/s11033-018-4471-6. [DOI] [PubMed] [Google Scholar]
- 24.Lin S., Choe J., Du P., et al. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–345. doi: 10.1016/j.molcel.2016.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu Y., Guo X., Zhao M., et al. Contributions and prognostic values of m(6) A RNA methylation regulators in non-small-cell lung cancer. J Cell Physiol. 2020;235(9):6043–6057. doi: 10.1002/jcp.29531. [DOI] [PubMed] [Google Scholar]
- 26.Roundtree I.A., Evans M.E., Pan T., et al. Dynamic RNA modifications in gene expression regulation. Cell. 2017;169(7):1187–1200. doi: 10.1016/j.cell.2017.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bokar J.A., Rath-Shambaugh M.E., Ludwiczak R., et al. Characterization and partial purification of mRNA N6-adenosine methyltransferase from HeLa cell nuclei. Internal mRNA methylation requires a multisubunit complex. J Biol Chem. 1994;269(26):17697–17704. [PubMed] [Google Scholar]
- 28.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: 10.1038/nchembio.1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wang P., Doxtader K.A., Nam Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell. 2016;63(2):306–317. doi: 10.1016/j.molcel.2016.05.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.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: 10.1038/nature18298. [DOI] [PubMed] [Google Scholar]
- 31.Schwartz S., Mumbach M.R., Jovanovic M., et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites. Cell Rep. 2014;8(1):284–296. doi: 10.1016/j.celrep.2014.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ping X.L., Sun B.F., Wang L., et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–189. doi: 10.1038/cr.2014.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Patil D.P., Chen C.K., Pickering B.F., et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 2016;537(7620):369–373. doi: 10.1038/nature19342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Knuckles P., Lence T., Haussmann I.U., 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: 10.1101/gad.309146.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.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: 10.1038/s41421-018-0019-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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. doi: 10.1016/j.molcel.2018.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ma H., Wang X., Cai J., et al. N(6-)Methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat Chem Biol. 2019;15(1):88–94. doi: 10.1038/s41589-018-0184-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Shima H., Matsumoto M., Ishigami Y., et al. S-Adenosylmethionine synthesis is regulated by selective N(6)-adenosine methylation and mRNA degradation involving METTL16 and YTHDC1. Cell Rep. 2017;21(12):3354–3363. doi: 10.1016/j.celrep.2017.11.092. [DOI] [PubMed] [Google Scholar]
- 39.Pendleton K.E., 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. doi: 10.1016/j.cell.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Warda A.S., 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: 10.15252/embr.201744940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.van Tran N., Ernst F.G.M., Hawley B.R., et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 2019;47(15):7719–7733. doi: 10.1093/nar/gkz619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Richard E.M., Polla D.L., Assir M.Z., et al. Bi-allelic variants in METTL5 cause autosomal-recessive intellectual disability and microcephaly. Am J Hum Genet. 2019;105(4):869–878. doi: 10.1016/j.ajhg.2019.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schöller E., Weichmann F., Treiber T., et al. Interactions, localization, and phosphorylation of the m(6)A generating METTL3-METTL14-WTAP complex. RNA. 2018;24(4):499–512. doi: 10.1261/rna.064063.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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: 10.1038/nchembio.687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang X., Zhao B.S., Roundtree I.A., et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161(6):1388–1399. doi: 10.1016/j.cell.2015.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rau K., Rösner L., Rentmeister A. Sequence-specific m(6)A demethylation in RNA by FTO fused to RCas9. RNA. 2019;25(10):1311–1323. doi: 10.1261/rna.070706.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang X., Wei L.H., Wang Y., et al. Structural insights into FTO's catalytic mechanism for the demethylation of multiple RNA substrates. Proc Natl Acad Sci U S A. 2019;116(8):2919–2924. doi: 10.1073/pnas.1820574116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Aik W., Scotti J.S., Choi H., et al. Structure of human RNA N(6)-methyladenine demethylase ALKBH5 provides insights into its mechanisms of nucleic acid recognition and demethylation. Nucleic Acids Res. 2014;42(7):4741–4754. doi: 10.1093/nar/gku085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zheng G., Dahl J.A., 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: 10.1016/j.molcel.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tang C., Klukovich R., Peng H., et al. ALKBH5-dependent m6A demethylation controls splicing and stability of long 3'-UTR mRNAs in male germ cells. Proc Natl Acad Sci U S A. 2018;115(2):E325–E333. doi: 10.1073/pnas.1717794115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zou S., Toh J.D., Wong K.H., et al. N(6)-Methyladenosine: a conformational marker that regulates the substrate specificity of human demethylases FTO and ALKBH5. Sci Rep. 2016;6:25677. doi: 10.1038/srep25677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Huang H., Weng H., Chen J. m(6)A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer. Cancer Cell. 2020;37(3):270–288. doi: 10.1016/j.ccell.2020.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ueda Y., Ooshio I., Fusamae Y., et al. AlkB homolog 3-mediated tRNA demethylation promotes protein synthesis in cancer cells. Sci Rep. 2017;7:42271. doi: 10.1038/srep42271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang X., Lu Z., Gomez A., et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117–120. doi: 10.1038/nature12730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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: 10.1038/cr.2017.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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: 10.1038/ncomms12626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Li A., Chen Y.S., Ping X.L., et al. Cytoplasmic m(6)A reader YTHDF3 promotes mRNA translation. Cell Res. 2017;27(3):444–447. doi: 10.1038/cr.2017.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Roundtree I.A., Luo G.Z., Zhang Z., et al. YTHDC1 mediates nuclear export of N(6)-methyladenosine methylated mRNAs. Elife. 2017;6 doi: 10.7554/eLife.31311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lesbirel S., Viphakone N., Parker M., et al. The m(6)A-methylase complex recruits TREX and regulates mRNA export. Sci Rep. 2018;8(1):13827. doi: 10.1038/s41598-018-32310-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hsu P.J., 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: 10.1038/cr.2017.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wojtas M.N., Pandey R.R., Mendel M., et al. Regulation of m(6)A transcripts by the 3'→5' RNA helicase YTHDC2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68(2):374–387. doi: 10.1016/j.molcel.2017.09.021. [DOI] [PubMed] [Google Scholar]
- 62.Alarcón C.R., Goodarzi H., Lee H., et al. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015;162(6):1299–1308. doi: 10.1016/j.cell.2015.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Liu N., Dai Q., Zheng G., et al. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518(7540):560–564. doi: 10.1038/nature14234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Liu N., Zhou K.I., Parisien M., et al. N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 2017;45(10):6051–6063. doi: 10.1093/nar/gkx141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.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: 10.1038/s41556-018-0045-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Xu R., Pang G., Zhao Q., et al. The momentous role of N6-methyladenosine in lung cancer. J Cell Physiol. 2021;236(5):3244–3256. doi: 10.1002/jcp.30136. [DOI] [PubMed] [Google Scholar]
- 67.Pan H., Li X., Chen C., et al. Research advances of m(6)A RNA methylation in non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 2020;23(11):961–969. doi: 10.3779/j.issn.1009-3419.2020.102.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Choe J., Lin S., Zhang W., et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature. 2018;561(7724):556–560. doi: 10.1038/s41586-018-0538-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Jin D., Guo J., Wu Y., et al. m(6)A mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. J Hematol Oncol. 2019;12(1):135. doi: 10.1186/s13045-019-0830-6. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 70.Cheng C., Wu Y., Xiao T., et al. METTL3-mediated m(6)A modification of ZBTB4 mRNA is involved in the smoking-induced EMT in cancer of the lung. Mol Ther Nucleic Acids. 2021;23:487–500. doi: 10.1016/j.omtn.2020.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 71.Wang H., Deng Q., Lv Z., et al. N6-methyladenosine induced miR-143-3p promotes the brain metastasis of lung cancer via regulation of VASH1. Mol Cancer. 2019;18(1):181. doi: 10.1186/s12943-019-1108-x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 72.Wanna-Udom S., Terashima M., Lyu H., et al. The m6A methyltransferase METTL3 contributes to Transforming Growth Factor-beta-induced epithelial-mesenchymal transition of lung cancer cells through the regulation of JUNB. Biochem Biophys Res Commun. 2020;524(1):150–155. doi: 10.1016/j.bbrc.2020.01.042. [DOI] [PubMed] [Google Scholar]
- 73.Xue L., Li J., Lin Y., et al. m(6) A transferase METTL3-induced lncRNA ABHD11-AS1 promotes the Warburg effect of non-small-cell lung cancer. J Cell Physiol. 2021;236(4):2649–2658. doi: 10.1002/jcp.30023. [DOI] [PubMed] [Google Scholar]
- 74.Wei W., Huo B., Shi X. miR-600 inhibits lung cancer via downregulating the expression of METTL3. Cancer Manag Res. 2019;11:1177–1187. doi: 10.2147/CMAR.S181058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Du Y., Hou G., Zhang H., et al. SUMOylation of the m6A-RNA methyltransferase METTL3 modulates its function. Nucleic Acids Res. 2018;46(10):5195–5208. doi: 10.1093/nar/gky156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Shen Y., Li C., Zhou L., et al. G protein-coupled oestrogen receptor promotes cell growth of non-small cell lung cancer cells via YAP1/QKI/circNOTCH1/m6A methylated NOTCH1 signalling. J Cell Mol Med. 2021;25(1):284–296. doi: 10.1111/jcmm.15997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liu J., Ren D., Du Z., et al. m(6)A demethylase FTO facilitates tumor progression in lung squamous cell carcinoma by regulating MZF1 expression. Biochem Biophys Res Commun. 2018;502(4):456–464. doi: 10.1016/j.bbrc.2018.05.175. [DOI] [PubMed] [Google Scholar]
- 78.Li J., Han Y., Zhang H., et al. The m6A demethylase FTO promotes the growth of lung cancer cells by regulating the m6A level of USP7 mRNA. Biochem Biophys Res Commun. 2019;512(3):479–485. doi: 10.1016/j.bbrc.2019.03.093. [DOI] [PubMed] [Google Scholar]
- 79.Chao Y., Shang J., Ji W. ALKBH5-m(6)A-FOXM1 signaling axis promotes proliferation and invasion of lung adenocarcinoma cells under intermittent hypoxia. Biochem Biophys Res Commun. 2020;521(2):499–506. doi: 10.1016/j.bbrc.2019.10.145. [DOI] [PubMed] [Google Scholar]
- 80.Guo J., Wu Y., Du J., et al. Deregulation of UBE2C-mediated autophagy repression aggravates NSCLC progression. Oncogenesis. 2018;7(6):49. doi: 10.1038/s41389-018-0054-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Jin D., Guo J., Wu Y., et al. m(6)A demethylase ALKBH5 inhibits tumor growth and metastasis by reducing YTHDFs-mediated YAP expression and inhibiting miR-107/LATS2-mediated YAP activity in NSCLC. Mol Cancer. 2020;19(1):40. doi: 10.1186/s12943-020-01161-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sheng H., Li Z., Su S., et al. YTH domain family 2 promotes lung cancer cell growth by facilitating 6-phosphogluconate dehydrogenase mRNA translation. Carcinogenesis. 2020;41(5):541–550. doi: 10.1093/carcin/bgz152. [DOI] [PubMed] [Google Scholar]
- 83.Müller S., Glaß M., Singh A.K., et al. IGF2BP1 promotes SRF-dependent transcription in cancer in a m6A- and miRNA-dependent manner. Nucleic Acids Res. 2019;47(1):375–390. doi: 10.1093/nar/gky1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ma L., Chen T., Zhang X., et al. The m(6)A reader YTHDC2 inhibits lung adenocarcinoma tumorigenesis by suppressing SLC7A11-dependent antioxidant function. Redox Biol. 2021;38:101801. doi: 10.1016/j.redox.2020.101801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wang J., Tan L., Jia B., et al. Downregulation of m(6)A reader YTHDC2 promotes the proliferation and migration of malignant lung cells via CYLD/NF-κB pathway. Int J Biol Sci. 2021;17(10):2633–2651. doi: 10.7150/ijbs.58514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lee J.H., Xiong F., Li W. Enhancer RNAs in cancer: regulation, mechanisms and therapeutic potential. RNA Biol. 2020;17(11):1550–1559. doi: 10.1080/15476286.2020.1712895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Li N., Zhan X. Identification of pathology-specific regulators of m(6)A RNA modification to optimize lung cancer management in the context of predictive, preventive, and personalized medicine. EPMA J. 2020;11(3):485–504. doi: 10.1007/s13167-020-00220-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Li N., Chen X., Liu Y., et al. Gene characteristics and prognostic values of m(6)A RNA methylation regulators in nonsmall cell lung cancer. J Healthc Eng. 2021;2021:2257066. doi: 10.1155/2021/2257066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Shi Y., Fan S., Wu M., et al. YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression. Nat Commun. 2019;10(1):4892. doi: 10.1038/s41467-019-12801-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zhu J., Wang M., Hu D. Deciphering N(6)-methyladenosine-related genes signature to predict survival in lung adenocarcinoma. BioMed Res Int. 2020;2020:2514230. doi: 10.1155/2020/2514230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhuang Z., Chen L., Mao Y., et al. Diagnostic, progressive and prognostic performance of m(6)A methylation RNA regulators in lung adenocarcinoma. Int J Biol Sci. 2020;16(11):1785–1797. doi: 10.7150/ijbs.39046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Li F., Wang H., Huang H., et al. m6A RNA methylation regulators participate in the malignant progression and have clinical prognostic value in lung adenocarcinoma. Front Genet. 2020;11:994. doi: 10.3389/fgene.2020.00994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Chen B., Ye F., Yu L., et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc. 2012;134(43):17963–17971. doi: 10.1021/ja3064149. [DOI] [PubMed] [Google Scholar]
- 94.Yang L., Li J., Xu L., et al. Rhein shows potent efficacy against non-small-cell lung cancer through inhibiting the STAT3 pathway. Cancer Manag Res. 2019;11:1167–1176. doi: 10.2147/CMAR.S171517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Huang Y., Yan J., Li Q., et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015;43(1):373–384. doi: 10.1093/nar/gku1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Cui Q., Shi H., Ye P., et al. m(6)A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 2017;18(11):2622–2634. doi: 10.1016/j.celrep.2017.02.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.He W., Zhou B., Liu W., et al. Identification of A novel small-molecule binding site of the fat mass and obesity associated protein (FTO) J Med Chem. 2015;58(18):7341–7348. doi: 10.1021/acs.jmedchem.5b00702. [DOI] [PubMed] [Google Scholar]
- 98.Qiao Y., Zhou B., Zhang M., et al. A novel inhibitor of the obesity-related protein FTO. Biochemistry. 2016;55(10):1516–1522. doi: 10.1021/acs.biochem.6b00023. [DOI] [PubMed] [Google Scholar]
- 99.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. doi: 10.1016/j.cell.2017.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.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. doi: 10.1016/j.ccell.2019.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Fedeles B.I., Singh V., Delaney J.C., et al. The AlkB family of Fe(II)/alpha-ketoglutarate-dependent dioxygenases: repairing nucleic acid alkylation damage and beyond. J Biol Chem. 2015;290(34):20734–20742. doi: 10.1074/jbc.R115.656462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lin A.P., Abbas S., Kim S.W., et al. D2HGDH regulates alpha-ketoglutarate levels and dioxygenase function by modulating IDH2. Nat Commun. 2015;6:7768. doi: 10.1038/ncomms8768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.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. doi: 10.1016/j.stem.2017.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhang J., Tsoi H., Li X., et al. Carbonic anhydrase IV inhibits colon cancer development by inhibiting the Wnt signalling pathway through targeting the WTAP-WT1-TBL1 axis. Gut. 2016;65(9):1482–1493. doi: 10.1136/gutjnl-2014-308614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kloor D., Osswald H. S-Adenosylhomocysteine hydrolase as a target for intracellular adenosine action. Trends Pharmacol Sci. 2004;25(6):294–297. doi: 10.1016/j.tips.2004.04.004. [DOI] [PubMed] [Google Scholar]