Skip to main content

The molecular mechanism of METTL3 promoting the malignant progression of lung cancer

Abstract

Lung cancer remains one of the major causes of cancer-related death globally. Recent studies have shown that aberrant m6A levels caused by METTL3 are involved in the malignant progression of various tumors, including lung cancer. The m6A modification, the most abundant RNA chemical modification, regulates RNA stabilization, splicing, translation, decay, and nuclear export. The methyltransferase complex plays a key role in the occurrence and development of many tumors by installing m6A modification. In this complex, METTL3 is the first identified methyltransferase, which is also the major catalytic enzyme. Recent findings have revealed that METTL3 is remarkably associated with different aspects of lung cancer progression, influencing the prognosis of patients. In this review, we will focus on the underlying mechanism of METT3 in lung cancer and predict the future work and potential clinical application of targeting METTL3 for lung cancer therapy.

Introduction

Lung cancer is one of the most common malignant tumors with the highest mortality rate worldwide [1,2,3]. According to histological appearance, lung cancer is classified into small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) [4, 5]. Unfortunately, due to the lack of effective means of early diagnosis, most patients with lung cancer are found to be in an advanced stage and have a poor prognosis [6]. Even with the development of multidisciplinary comprehensive management of lung cancer, the overall survival (OS) of patients with lung cancer is still very low, about 15% [7].

As we all know, tumorigenesis is an extremely complex biological process involving genomics and epigenetics [8, 9]. There is strong evidence that epigenetic modification has a profound impact on the occurrence and development of tumors without DNA sequence changes [10]. The epigenetic modifications include DNA methylation, histone modification, RNA modification, and noncoding RNA [11, 12]. However, with the studies of DNA methylation, histone modification and noncoding RNA in full swing, we know little about RNA modification.

At present, more than 170 RNA modifications have been found in mammals [13], while m6A modification is the most abundant RNA chemical modification, accounting for approximately 60% of all RNA modifications [14, 15]. RNA m6A modification was reported as early as 1970 [16, 17], but it was not until the emergence of methylated RNA immunoprecipitation sequencing (MeRIP-seq) that m6A modification became the focus of epigenetic modification. In addition, aberrant m6A levels mediated by METTL3 have been reported to be involved in the malignant progression of various tumors, including proliferation, invasion, metastasis, and drug resistance.

However, RNA m6A modifications account for about 0.1–0.4% of the isolated RNA from eukaryotic cells [16]. Studies have shown that the sites modified by m6A are highly conservative and tend to be present in the consensus sequence Pu[G > A] m6AC[U > A > C](Pu, purine) [18]. Of note, m6A sites are mainly enriched near the 3' untranslated region (UTR), stop codon and long internal exon [18, 19], and play an important role in regulating RNA metabolism. In mammals, m6A modification is dynamically reversible, installed by the methyltransferase complex (writers) and removed by demethylases (erasers) [20, 21]. At least eight methyltransferases are found in the methyltransferase complex, including METTL3, METTL14, WTAP, KIAA1429(VIRMA), RBM15, HAK AI, ZC3H13(KIA A0853), and METTL16. In the methyltransferase complex, METTL3 is the first identified and the sole catalytic subunit that can catalyze the transfer of a methyl group from S-adenosylmethionine (SAM) to the N6-position of adenosine [13]. In addition, RNA binding proteins, also called readers, can selectively recognize and bind to the m6A modification sites of target RNAs, thereby participating in the RNA metabolism process, including RNA splicing, maturation, decay, translation, stabilization, Pri-mRNA processing, nuclear export [22, 23] (Fig. 1). With the deepening of the research, more and more novel m6A-related regulators will be identified.

Fig. 1
figure 1

Molecular components involved in the dynamic and reversible process of m6A methylation include methyltransferases (METTL3, METTL14, METTL16, WTAP, KIAA1429, ZC3H13, RBM15/15B), demethylases (FTO, ALKBH5) and RNA-binding proteins (YTHDF1, YTHDF2, YTHDF3, YTHDC1, YTHDC2, IGF2BP1/2/3, HNRNPC, HNRNPA2B1). METTL3 has an influence on the expression of key oncogenes by regulating RNA metabolism, such as Pri-mRNA processing, RNA splicing, maturation, stability, translation, decay, and nuclear export

Recent findings have revealed that METTL3 is closely related to the progression of a variety of tumors, including lung cancer. In the present review, we will discuss the underlying molecular mechanism of METTL3 in the occurrence and development of lung cancer and predict the future research direction, as well as the potential clinical application of targeting METTL3 in lung cancer treatment.

Effects of methyltransferase METTL3 on lung cancer

Accumulating evidence has shown that abnormal m6A levels mediated by METTL3 are involved in the malignant progression of lung cancer, including cell proliferation, invasion, metastasis, angiogenesis, drug resistance, glycolysis, cancer stem cells, and tumor environment [24,25,26]. Therefore, we summarize the recent findings of METTL3 in the tumorigenesis of lung cancer (Table 1).

Table 1 Effect of METTL3 on lung cancer

Effects of METTL3 on the proliferation of lung cancer

Lung cancer is a malignant tumor originating from bronchial mucosal epithelial cells that is closely associated with excessive cell division, cycle disturbance, and apoptosis dysregulation [27,28,29]. It has been reported that long noncoding RNA (LncRNA) ABHD11-AS1 was highly expressed in NSCLC, closely related to poor prognosis. Mechanistically, METTL3 increases the transcript stability of ABHD11-AS1 and reduces the expression of its downstream gene KIF4, promoting cell proliferation [30]. In contrast, RNA pumilio RNA binding family member 1 (circPUM1), a functional circRNA, promotes NSCLC cell growth by activating the miR-590-5p/METTL3 axis [31]. Thus, METTL3 participates in the initiation and development of lung cancer by modifying noncoding RNA. Interestingly, miR-33a can inhibit NSCLC cell proliferation by downregulating METTL3 expression and its downstream genes, such as epidermal growth factor receptor (EGFR) and transcriptional coactivator with PDZ-binding motif (TAZ), via directly targeting the 3'UTR of METL3 mRNA [32].

In addition, METTL3 can also act as a tumor suppressor in lung cancer. Wu et al. found that numerous m6A methylation sites on FBXW7 mRNA in lung adenocarcinoma (LUAD) by using MeRIP-qPCR analysis. Further study showed that METTL3 upregulates FBXW7 expression to regulate proliferation or apoptosis-related genes such as Bax, c-Myc, Mcl-1, in an m6A manner [33]. Similarly, METTL3 represses LUAD tumorigenesis by regulating SLC7A11 mRNA expression. Mechanistically, YTHDC2 (reader) preferentially binds to m6A-modified SLC7A11 mRNA, which makes it more likely to be degraded, preventing cystine uptake and antioxidant function [34].

Effects of METTL3 on the invasion, migration and metastasis of lung cancer

Accumulating studies have shown that METTL3 overexpression in lung cancer tissues was significantly higher than that in normal cells and was strongly associated with tumor invasion, migration, and metastasis [35,36,37]. In NSCLC, METTL3-mediated Yes-associated protein (YAP) overexpression leads to tumor metastasis. By analyzing the upstream regulatory mechanism of YAP, researchers found that METTL3 not only upregulates the m6A level of YAP transcript to enhance its translation but also activates the MALAT1-miR-1914-3p-YAP axis to increase the stability of YAP transcript [38]. In lung cancer epithelial-mesenchymal transition (EMT) models mediated by TGF-β, METTL3 markedly accelerates the EMT process [35]. Mechanistically, METTL3 significantly enhances JUNB mRNA stability dependent of m6A methylation. In contrast, E-cadherin expression was upregulated when METTL3 knockdown was performed.

Moreover, CTNNB1 gene  encodes β-catenin protein. The m6A level of CTNNB1 mRNA is abnormally increased, closely related to the EMT process in HeLa cell line. Mechanistically, METTL3 modifies the 5'UTR region of CTNNB1 mRNA and negatively regulates CTNNB1 mRNA stability and translation [39]. In addition, METTL3 upregulates the expression of transcription factor E2F1, thereby indirectly downregulating β-catenin protein. Intriguingly, this study also revealed that YTHDF1 selectively binds eIF4E1 or eIF4E3 to regulate β-catenin expression in a noncanonical pathway, according to the level of METTL3 in the cell. For example, in METTL3 knockdown cells, YTHDF1 tends to bind the oncoprotein eIF4E1 to upregulate β-catenin.  Eventually, METTL3 can also decrease c-Met kinase expression to repress membrane localization of β-catenin, inhibiting cell migration.

The m6A levels were reported to be affected by environmental factors, including particulate matter (PM2.5) [40] and cigarette smoke [41]. Recent studies have shown that METTL3 overexpression induced by smoke exposure downregulates E-cadherin to accelerate the malignant transformation of normal lung tissue in mice by activating the ZBTB4/EZH2/ H3K27me3 axis [41]. It has been reported that METTL3 participates in m6A-modified mRNA translation independently of its catalytic activity or m6A readers in lung cancer lines [42]. In METTL3 mutation assays, its N-terminal domain (1–200 amino acids) did not exhibit methyltransferase catalytic activity but is sufficient to increase the translation efficiency of target mRNAs. Mechanistically, METTL3 interacts with eIF3h to form the RNA looping to enhance significantly the translation efficiency of polyribosomes, leading to an increase in the expression of key oncogenes such as EGFR and TAZ [43]. However, METTL3 approximately selectively binds only 22% of RNA containing the m6A methylation site [44], so it is vital to explore selection mechanisms of METTL3 for target mRNAs to elucidate its biological function.

Effects of METTL3 on drug resistance of lung cancer

Drug resistance is the major reason for the failure of most solid tumor treatments [8, 45, 46], its mechanism is quite complicated, including cancer stemness, the ABC transporter family, noncoding RNA regulation, tumor microenvironment, hypoxia, autophagy, DNA damage and repair, and epigenetic modification. However, there is a lot of evidence that METTL3 is linked to drug resistance in many different types of tumors, including lung cancer [27].

In a murine cisplatin-resistant lung cancer model, METTL3 upregulates YAP expression to promote drug resistance and metastasis by YTHDF1/3, eIF3b and the MALAT1-miR-1914-3p-YAP axis [38]. In contrast, METTL3 knockdown increased the sensitivity of lung cancer cells to Cisplatin by downregulating YAP expression. C-Met overexpression in NSCLC was positively correlated with Crizotinib resistance [47, 48]. NSCLC cells treated with Chidamide increased sensitivity to Crizotinib in vivo and vitro. Mechanistically, Chidamide reduced m6A level of c-Met and its expression by downregulating METTL3 and WTAP [49]. Liu et al. found that METTL3-mediated autophagy is involved in NSCLC resistance to gefitinib, and further mechanistic studies revealed that METTL3 in NSCLC regulates autophagy-related gene expression such as ATG5, ATG7, LC3B, and SQSTM1 to promote cell survival in an m6A manner [50]. Similarly, METTL3 overexpression was positively correlated with MET in gefitinib-resistant LUAD. Further studies revealed that METTL3 regulates MET expression, thereby synergistically activating the downstream PI3K/AKT pathway and reducing the sensitivity of LUAD to gefitinib [51]. Conversely, METTL3 knockdown has also been reported to increase NSCLC resistance to Cisplatin [52].

Effects of METTL3 on the glycolysis of lung cancer

Abnormal glucose metabolism has been reported to facilitate malignant tumor initiation and development, which is one of the key energy metabolism features of tumors [8, 53]. Recent findings have revealed that circPUM1 upregulated the expression levels of glucose transporter 1(GLUT1) and hexokinase-2 (HK2) to promote the glycolysis of lung cancer by upregulating METTL3 [31].

Different from the oxidative phosphorylation of mitochondria in normal cells, tumor cells mainly rely on aerobic glycolysis for energy supply, a phenomenon called "Warburg effect" [54, 55], which provides a beneficial environment for tumor cell growth. The study has shown that METTL3 could activate ABHD11-AS1/EZH2/KLF4 axis to downregulate the expression of transcription factor Kruppel-like factor4 (KLF4), enhancing the Warburg effect [30]. Besides, METTL3 might also be involved in other energy mechanism pathways, such as lipid metabolism, amino acid metabolism, which needs to be further explored.

Effects of METTL3 on angiogenesis of lung cancer

Angiogenesis, a typical feature of the malignant progression of tumor cells, is a complex biological process by which new capillaries grow from preexisting vessels, providing oxygen and nutrients for the malignant progression of tumors [8, 56, 57]. METTL3 was reported to regulate the expression of let-7e-5p and miR-18a-5p to significantly improve endothelial cells (ECs) biological functions that facilitate neovascularization in limb ischemia and myocardial infarction mouse models [58]. Thus, METTL3 can serve as an important regulator of angiogenesis. Similarly, METTL3 also regulated miR-143-3p/VASH1 axis to enhance the angiogenesis ability of lung cancer cells in an m6A manner [59].

Effects of METTL3 on the tumor microenvironment of lung cancer

The tumor microenvironment (TME) is composed of cancer cells, cancer stem cells, endothelial cells, pericytes, cancer-associated fibroblasts, immune and inflammatory cells, as well as extracellular components such as vascular endothelial-derived growth factor (VEGF), ERGF etc., participating in tumor growth, invasion, metastasis, and drug resistance [8, 27].

METTL3 depletion in macrophages reshaped the TME by increasing M1- and M2-like tumor-associated macrophages (TAMs) and regulatory T (Treg) cell infiltration in vivo, resulting in tumor growth, metastasis, and drug resistance. Mechanistically, ablation of METTL3 in macrophages inhibits the YTHDF1-mediated SPRED2 translation to upregulate ERK expression to activate NF-κB and STAT3 signaling [60]. Thus, METTL3 plays a key role in the TME. However, the molecular mechanisms involved in the TME are quite complex, and METTL3-mediated remodelling of the TME also only reveals the tip of the iceberg where m6A methylation participates in the formation of the TME (Fig. 2).

Fig. 2
figure 2

The establishment of the protein–protein interaction network for m6A regulators and its associated genes in lung cancer on the basis of String database

Effects of METTL3 on the prognosis of lung cancer

More and more studies have demonstrated that METTL3 is strongly linked to the prognosis of lung cancer. However, whether METTL3 can precisely reflect clinical outcomes is controversial. The retrospective study conducted by Liu et al. evaluated the relationship between METTL3 and the prognosis of lung cancer through meta-analysis and bioinformatic analysis [61]. The result has shown that METTL3 overexpression is closely related to the prognosis of various tumors and could serve as a potential tumor biomarker [61].

By analyzing 22 immune cell types, Xu et al. identified that T follicular helper cell signature (risk core) could serve as an independent prognostic factor in patients with lung squamous cell carcinoma (LUSC) [62]. LUSC patients were separated into low-risk and high-risk groups based on this risk core. Interestingly, low-risk groups exhibited a worse OS, in which the expression of METTL3, HNRNPC, ALKBH5, and KIAA1429 was upregulated. While high-risk groups in which m6A-related regulators is downregulated better respond to chemotherapies and immunotherapies, suggesting METTL3 overexpression may predict a poor prognosis in LUSC patients [62].

The study by Zhang et al. has shown that METTL3 overexpression in LUAD was strongly related to better OS and progression free survival (PFS) [63]. Hence, METTL3 could serve as the protective gene in LUAD. However, another study suggested the opposite conclusion that METTL3 overexpression was negatively associated with LUAD prognosis [64].

Construction of a risk score model and lung cancer prognosis

METTL3, together with other m6A-related regulators, participates synergistically in an m6A methylation. Therefore, it is less reliable to estimate the prognostic value of METTL3 in lung cancer. Hence, based on multiple-gene signatures, the risk score model is constructed to reflect the prognosis of patients with lung cancer more reasonably. Zhu et al. found that nothing in the six m6A-related regulators is a prognostic risk factor for lung cancer [65]. The risk score model of six genes was built through bioinformatics analysis, which is significantly associated with clinicopathological features and survival outcomes, serving as an independent predictor of prognosis in LUAD [65]. Similarly, based on eight m6A regulators, an optimal prognostic gene risk score model was constructed by Liu et al., which could serve as an independent prognostic factor in LUAD [66]. Additionally, the risk score model constructed by three risk genes (METTL3, YTHDC1, and HNRNPC) also did well in predicting the prognosis of LUSC patients [66]. Moreover, Zhuang et al. constructed the risk score model by using ten m6A regulators, which were strongly related to clinicopathological characteristics and could be used to be an independent risk factor in LUAD [67]. Unfortunately, this model was not applicable to LUSC.

Gene alternative splicing (GAS) can be explained by the production of multiple mRNA isoforms from a single gene, which regulates gene expression at the posttranscriptional level and plays a crucial role in the development of diverse diseases, including cancer [68,69,70,71]. The risk signature of the m6A-associated GAS events was constructed by Zhao et al., which could serve as an independent prognostic risk factor in LUAD and LUSC [72]. Of note, METTL3, HNRNPC, and RBM15, as the splicing factors, can also be directly involved in GAS events in NSCLC [72].

Relationship between METTL3 and noncoding RNA in lung cancer

Noncoding RNA is functional RNA that is not translated into proteins but can regulate gene expression [73, 74]. According to length, noncoding RNA can be divided into short-chain noncoding RNA (siRNA, miRNA, piRNA) and long-chain noncoding RNA (LncRNA) [75, 76].

In mammals, miRNA can regulate the expression of target genes at the posttranscriptional level through incomplete complementary pairing with mRNA 3'UTR. Interestingly, 3'UTR is also where m6A modification is enriched [75]. Studies have shown that approximately 67% of transcripts in the 3'UTR with m6A modification contain at least one miRNA binding site [18]. Hence, METTL3-mediated m6A methylation modification has a high link with miRNA. Functionally, both can regulate critical oncogene expression and influence tumor progression. Additionally, it has been reported that nine m6A-mediated miRNA were identified in human bronchial epithelial cells (HBEs) treated with arsenite by using the Venn diagram and KEGG analysis [77], which might regulate crucial pathways related to cell proliferation and apoptosis, including the P53 pathway, mTOR pathway, and MAPK pathway, suggesting that miRNA could serve as the pivotal bridge by which METTL3-mediated m6A facilitates to cell proliferation or apoptosis in HBEs treated with arsenite. Table 2 lists the relationships between METTL3 and noncoding RNA in lung cancer.

Table 2 The relationships between METTL3 and noncoding RNA

In addition, the METTL3-YTHDC1 participates in the back-splicing of some circRNAs and affects their biogenesis. For example, YTHDF3/eIF4G2 regulates its translation by recognizing a specific m6A site on circ-ZNF609 [78]. It has been reported that circPUM1 knockdown inhibits NSCLC cell growth and glycolysis in vivo and vitro. Further study has shown that circPUM1 sponges miR-590-5p, which can directly target METTL3 and downregulate its expression. Liu et al. identified a novel circIGF2BP3 that is overexpressed in NSCLC and inhibits CD8+ T cell infiltration [79]. Mechanistically, METTL3 promotes PKP3 mRNA to form a protein-RNA complex with FXR1 to stabilize OTUB1 mRNA by regulating the circIGF2BP3/PKP3 axis. OTUB1 upregulates its expression by inhibiting PD-L1 ubiquitination in NSCLC cells to induce immune escape and resistance to PD-1 inhibitors.

Recent advances in targeting METTL3

Accumulating evidence has shown that METTL3 plays a crucial role in the tumorigenesis of lung cancer, dependent or independent of m6A modification. It could act as a potential therapeutic target (Table 3).

Table 3 METTL3 activators and inhibitors

METTL3 inhibitors

As the sole catalytic subunit in the methyltransferase complex, METTL3 is involved in different aspects of tumor progression, such as cell proliferation, invasion, migration, metastasis, tumor environment, cancer stem cells, and drug resistance [43, 80, 81].

Because adenosine could serve as a SAM-competitive inhibitor of METTL3, Bedi et al. identified seven compounds from among 4000 adenosine analogs and derivatives using high-throughput docking into METTL3, two of which (compounds 2 and 7) showed good ligand efficiency [82]. Additionally, simvastatin has been reported to exert anti-tumor activity in various cancers, including lung cancer. The study by Chen et al. has indicated that simvastatin suppresses cell proliferation, migration, invasion, metastasis and EMT by reducing EZH2 expression via downregulating METTL3 in lung cancer [83].

Drug combination

In order to overcome multiple drug resistance and prolong patient survival, drug combination is becoming the mainstream of lung cancer therapy [84, 85]. Recent evidence has shown that two different targeted agents for lung cancer could block numerous targets on the signal transduction pathway to prevent the malignant progression of lung cancer, thereby achieving better clinical efficacy [86,87,88]. Chidamide is a novel small molecular inhibitor targeting HDAC1/2/3/10 [89]. Interestingly, histone deacetylase inhibitors (HDACIs) combined with other agents strongly improved anti-tumor activities [90,91,92,93]. Recently, Ding et al. revealed that Chidamide could make NSCLC cells more sensitive to Crizotinib in vivo. Mechanistically, Chidamide can decrease the stability and translational efficiency of METTL3 and WTAP transcripts to downregulate the m6A level and expression of c-Met [49].

Others

METTL3 overexpression is common in NSCLC and dramatically accelerates the transcriptional efficiency of key oncogenes, resulting in NSCLC malignancy [42, 43]. However, it has also been shown that METTL3 overexpression is associated with a better prognosis in NSCLC patients. Based on bioinformatics analysis, Liu et al. constructed a risk score model with eight m6A methylation regulators, including METTL3, which could better respond to the clinical outcomes of LUAD and LUSC patients. METTL3 acted as a protective gene in this model and was commonly enriched in the low-risk group [66]. Similarly, Zhang et al. found that METTL3 overexpression was positively correlated with OS in LUAD samples from The Cancer Genome Atlas (TCGA) [63]. Zhu et al. constructed a risk score model based on six m6A methylation regulators that could better predict the clinicopathological characteristics of LUAD patients, and METTL3 acted as a tumor suppressor in this model [65].

It has been reported that Interleukin 37 (IL-37) increases METTL3 expression to upregulate the total m6A level, inhibiting A549 cell proliferation. Furthermore, IL-37 has anti-tumor activity by targeting the Wnt5a/5b pathway [94]. Therefore, Selberg et al. performed a virtual screening based on the crystal structure of the methyltransferase complex. It turned out that four small molecule compounds enhance its methyltransferase activity by specifically binding METTL3, upregulating the total m6A level in RNA in cells [95]. Mechanically, the compound increases SAM affinity for METTL3 and lowers the energy barrier of the m6A methylation reaction by interaction with SAM in the active center of METTL3 [95].

METTL3-METTL14-WTAP complex in lung cancer

METTL3 is the first identified methyltransferase and has sole enzymatic activity. However, METTL3 interacts with METTL14 to form a heterodimer with the highest enzymatic activity and a better preference for substrate RNAs [44]. However, WTAP binds to the METTL3-METTL14 heterodimer to form a METTL3-METTL14-WTAP complex localized at the nuclear speckle, thereby affecting the total m6A level [96]. Interestingly, METTL3, as the only catalytically active subunit of the methyltransferase complex, promotes the development of lung cancer dependent or independent of catalytic activity. On the one hand, METTL3 affects m6A levels of oncogenes or tumor suppressors, thereby regulating their expression to promote tumor progression [39, 81]. On the other hand, METTL3 interacts with eIF3h to form RNA looping independent on catalytic activity, which greatly accelerates the translation efficiency of polyribosomes and upregulates the expression of key oncogenes, such as EGFR and TAZ [42, 43] (Fig. 3).

Fig. 3
figure 3

METTL3 regulates tumor suppressors and oncogenes expression to promote lung cancer progression in an m6A manner (left). In addition, METTL3 also accelerate the translation efficiency of key oncogenes to affect lung cancer progression independent of its methyltransferase activity (right)

It has been reported that METTL14 depletion significantly downregulates m6A levels in tumor stromal cells, thereby decreasing CD8+ T cell infiltration and increasing dysfunctional T cells, leading to tumor growth [79]. Further study revealed that the METTL14-YTHDF2 axis maintained the balance between cytotoxic CD8+ T cells and dysfunctional T cells, and METTL14-depleted TAMs overexpressing Epstein-Barr virus-induced protein 3 (Ebi3) transcripts inhibited the anti-tumor activity of CD8+ T cells, which resulted in the conversion of CD8+ T cells to dysfunctional T cells. Furthermore, METTL14 is overexpressed in NSCLC cell lines and induces the EMT process [97]. Mechanistically, METTL14 knockdown downregulates Twist expression to inhibit the AKT pathway and upregulate E-cadherin, thereby inhibiting NSCLC cell migration. Of note, Li et al., based on TCGA and GEO databases, found that METTL14 was downregulated in lung cancer tissues compared to normal tissues. And METTL14 overexpression inhibited lung cancer growth and metastasis in vivo and in vitro through the miR-30c-1-3p/MARCKSL1 axis [98].

In 2000, Little et al. first identified WTAP using a yeast two-hybrid assay [99]. WTAP, a widespread nuclear protein, can specifically bind to the WT1 protein to co-localize at the nuclear speckle [99]. In addition, WTAP can also act as a splicing factor and participate in alternative splicing of specific a subset of m6A-modified RNAs. It has been reported that LncRNA PCGEM1 is highly expressed in NSCLC and promotes cell growth [100]. Mechanistically, LncRNA PCGEM1 sponges miR-433-3p to upregulate WTAP expression and accelerate NSCLC progression. Intriguingly, METTL3 levels are critical for WTAP homeostasis, and METTL3 knockdown or overexpression both upregulate WTAP expression [101]. Mechanistically, METTL3 overexpression increases WTAP mRNA translation and protein stability independent of its catalytic activity. In contrast, METTL3 knockdown increases WTAP mRNA levels and eventually upregulates the expression of WTAP. Notably, WTAP overexpression is insufficient to promote tumor progression when functional METTL3 is absent, implying that WTAP must depend on the METTL3-METTL14 complex to exert oncogenic activity [101].

Conclusions

Accumulating studies have shown that METTL3 plays a crucial role in the occurrence and development of lung cancer. METTL3 participates in cell proliferation, invasion, migration, metastasis, angiogenesis, glycolysis, drug resistance, and tumor microenvironment, dependent or not on catalytic activity. Thus, METTL3 could act as a potential therapeutic target for lung cancer. Notably, m6A regulators include methyltransferases and demethylases, while METTL3, as the sole methyltransferase with enzymatic activity, does not ultimately determine the global m6A level, suggesting that m6A levels may be regulated by specific patterns that require further investigation.

On the one hand, METTL3-mediated m6A methylation regulates the expression of oncogenes or tumor suppressors, influencing the malignant progression of lung cancer. On the other hand, METTL3 interacts with eIF3h to form RNA looping, significantly increasing the translation efficiency of key oncogenes. The combined effect of the two may determine the development of lung cancer. Intriguingly, either METTL3 knockdown or overexpression can regulate WTAP expression, suggesting that they can interact with each other, and act as upstream regulators of each other. Therefore, the regulatory network with METTL3 as the core in lung cancer is extremely complex and involves many molecular mechanisms.

A large number of studies have shown that METTL3 interacts with noncoding RNA to regulate the expression of downstream genes, thereby affecting the progression of lung cancer. Among noncoding RNA, especially circRNAs, the METTL3-YTHDC1 participates in circRNAs biogenesis and regulates the translation of specific m6A-modified circRNAs.

Notably, there is solid evidence that METTL3 can act as an oncogene in lung cancer cell lines but as a tumor suppressor in tumor stromal cells. For example, METTL3 deletion in macrophages can promote tumor growth, metastasis, and drug resistance by increasing M1- and M2-TAM and Treg infiltration in tumors and reshaping the tumor microenvironment. In addition, METTL14-deficient macrophages significantly downregulated the global m6A level to reduce CD8+ T cells and increase dysfunctional T cells, promoting tumor growth. In tumor stromal cells, especially TAM, METTL3 or METTL14 can serve as a protective genes for lung cancer. To our knowledge, this is the first time to propose that METTL3 and METTL14 play opposite roles in lung cancer cells and tumor stromal cells, precisely explaining why high levels of m6A  methylation  can be predictive of better prognosis in NSCLC patients.

Availability of data and materials

Not applicable.

Abbreviations

ALKBH5:

Alkb homologue 5

ATTM:

Ammonium tetrathiomolybdate

BRD4:

Bromodomain-containing protein

CSCs:

Cancer stem cells

EGFR:

Epidermal growth factor receptor

EMT:

Epithelial-mesenchymal transition

ECs:

Endothelial cells

FTO:

Fat mass and obesity-associated protein

GEO:

Gene Expression Omnibus

GAS:

Gene alternative splicing

GLUT1:

Glucose transporter 1

KLF4:

Kruppel-like factor4

HBEs:

Human bronchial epithelial cells

HGF:

Hepatocyte growth factor

HBXIP:

Hepatitis B X-interacting protein

HK2:

Hexokinase-2

HDGF:

Hepatoma-derived growth factor

IL-37:

Interleukin 37

LUAD:

Lung adenocarcinoma

LUSC:

Lung squamous cell carcinoma

Lnc RNA:

Long noncoding RNA

METTL3:

Methyltransferase-like 3

NSCLC:

Non-small cell lung cancer

OS:

Overall survival

RFS:

Progression free survival

LASSO:

Least absolute shrinkage and selection operator

PM2.5 :

Particulate matter 2.5

SCLC:

Small cell lung cancer

SNAI1:

Snail family transcriptional repressor 1

SAM:

S-Adenosylmethionine

SOCS2:

Suppressor of cytokine signaling 2

TCGA:

The Cancer Genome Atlas

TME:

Tumor environment

TAZ:

Transcriptional coactivator with PDZ-binding motif

VEGF:

Vascular endothelial-derived growth factor

YAP:

Yes-associated protein

3'UTR:

3' Untranslated region

References

  1. Sheikh M, Mukeriya A, Shangina O, Brennan P, Zaridze D. Postdiagnosis smoking cessation and reduced risk for lung cancer progression and mortality: a prospective cohort study. Ann Intern Med. 2021;174(9):1232–9.

    PubMed  Google Scholar 

  2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.

    PubMed  Google Scholar 

  3. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet. 2021;398(10299):535–54.

    PubMed  Google Scholar 

  4. Zhang Y, Wang DC, Shi L, Zhu B, Min Z, Jin J. Genome analyses identify the genetic modification of lung cancer subtypes. Semin Cancer Biol. 2017;42:20–30.

    CAS  PubMed  Google Scholar 

  5. Morgan E, Arnold M, Rutherford MJ, Bardot A, Ferlay J, De P, Engholm G, Jackson C, Little A, Saint-Jacques N, et al. The impact of reclassifying cancers of unspecified histology on international differences in survival for small cell and non-small cell lung cancer (ICBP SurvMark-2 project). Int J Cancer. 2021;149(5):1013–20.

    CAS  PubMed  Google Scholar 

  6. Oudkerk M, Liu S, Heuvelmans MA, Walter JE, Field JK. Lung cancer LDCT screening and mortality reduction—evidence, pitfalls and future perspectives. Nat Rev Clin Oncol. 2021;18(3):135–51.

    PubMed  Google Scholar 

  7. Leon G, MacDonagh L, Finn SP, Cuffe S, Barr MP. Cancer stem cells in drug resistant lung cancer: targeting cell surface markers and signaling pathways. Pharmacol Ther. 2016;158:71–90.

    CAS  PubMed  Google Scholar 

  8. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    CAS  PubMed  Google Scholar 

  9. Wang T, Kong S, Tao M, Ju S. The potential role of RNA N6-methyladenosine in cancer progression. Mol Cancer. 2020;19(1):88.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Hogg SJ, Beavis PA, Dawson MA, Johnstone RW. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800.

    CAS  PubMed  Google Scholar 

  11. Wątroba M, Dudek I, Skoda M, Stangret A, Rzodkiewicz P, Szukiewicz D. Sirtuins, epigenetics and longevity. Ageing Res Rev. 2017;40:11–9.

    PubMed  Google Scholar 

  12. Skvortsova K, Iovino N, Bogdanović O. Functions and mechanisms of epigenetic inheritance in animals. Nat Rev Mol Cell Biol. 2018;19(12):774–90.

    CAS  PubMed  Google Scholar 

  13. Zeng C, Huang W, Li Y, Weng H. Roles of METTL3 in cancer: mechanisms and therapeutic targeting. J Hematol Oncol. 2020;13(1):117.

    PubMed  PubMed Central  Google Scholar 

  14. Li J, Yang X, Qi Z, Sang Y, Liu Y, Xu B, Liu W, Xu Z, Deng Y. The role of mRNA m(6)A methylation in the nervous system. Cell Biosci. 2019;9:66.

    PubMed  PubMed Central  Google Scholar 

  15. Xu J, Liu Y, Liu J, Xu T, Cheng G, Shou Y, Tong J, Liu L, Zhou L, Xiao W, et al. The identification of critical m(6)A RNA methylation regulators as malignant prognosis factors in prostate adenocarcinoma. Front Genet. 2020;11:602485.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci USA. 1974;71(10):3971–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wei CM, Gershowitz A, Moss B. Methylated nucleotides block 5’ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379–86.

    CAS  PubMed  Google Scholar 

  18. 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):1635–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Ke S, Alemu EA, Mertens C, Gantman EC, Fak JJ, Mele A, Haripal B, Zucker-Scharff I, Moore MJ, Park CY, et al. A majority of m6A residues are in the last exons, allowing the potential for 3’ UTR regulation. Genes Dev. 2015;29(19):2037–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang Y-G, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang C-M, Li CJ, Vågbø CB, Shi Y, Wang W-L, Song S-H, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.

    CAS  PubMed  Google Scholar 

  22. Chen X-Y, Zhang J, Zhu J-S. The role of m6A RNA methylation in human cancer. Mol Cancer. 2019;18(1):103.

    PubMed  PubMed Central  Google Scholar 

  23. Meyer KD, Jaffrey SR. Rethinking m(6)A readers, writers, and erasers. Annu Rev Cell Dev Biol. 2017;33:319–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Lan Q, Liu PY, Haase J, Bell JL, Hüttelmaier S, Liu T. The critical role of RNA m(6)A methylation in cancer. Cancer Res. 2019;79(7):1285–92.

    CAS  PubMed  Google Scholar 

  25. 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):270–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang Q, Geng W, Guo H, Wang Z, Xu K, Chen C, Wang S. Emerging role of RNA methyltransferase METTL3 in gastrointestinal cancer. J Hematol Oncol. 2020;13(1):57.

    PubMed  PubMed Central  Google Scholar 

  27. Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12(1):31–46.

    CAS  PubMed  Google Scholar 

  28. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.

    PubMed  Google Scholar 

  29. Liu Z, Wang T, She Y, Wu K, Gu S, Li L, Dong C, Chen C, Zhou Y. N(6)-methyladenosine-modified circIGF2BP3 inhibits CD8(+) T-cell responses to facilitate tumor immune evasion by promoting the deubiquitination of PD-L1 in non-small cell lung cancer. Mol Cancer. 2021;20(1):105.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Xue L, Li J, Lin Y, Liu D, Yang Q, Jian J, Peng J. 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):2649–58.

    CAS  PubMed  Google Scholar 

  31. Li M, Wang Q, Zhang X, Yan N, Li X. CircPUM1 promotes cell growth and glycolysis in NSCLC via up-regulating METTL3 expression through miR-590–5p. Cell Cycle. 2021;20(13):1279–94.

    CAS  PubMed  Google Scholar 

  32. Du M, Zhang Y, Mao Y, Mou J, Zhao J, Xue Q, Wang D, Huang J, Gao S, Gao Y. MiR-33a suppresses proliferation of NSCLC cells via targeting METTL3 mRNA. Biochem Biophys Res Commun. 2017;482(4):582–9.

    CAS  PubMed  Google Scholar 

  33. Wu Y, Chang N, Zhang Y, Zhang X, Xu L, Che Y, Qiao T, Wu B, Zhou Y, Jiang J, et al. METTL3-mediated m(6)A mRNA modification of FBXW7 suppresses lung adenocarcinoma. J Exp Clin Cancer Res. 2021;40(1):90.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Ma L, Chen T, Zhang X, Miao Y, Tian X, Yu K, Xu X, Niu Y, Guo S, Zhang C, et al. The m(6)A reader YTHDC2 inhibits lung adenocarcinoma tumorigenesis by suppressing SLC7A11-dependent antioxidant function. Redox Biol. 2021;38:101801.

    CAS  PubMed  Google Scholar 

  35. Wanna-Udom S, Terashima M, Lyu H, Ishimura A, Takino T, Sakari M, Tsukahara T, Suzuki T. 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):150–5.

    CAS  PubMed  Google Scholar 

  36. Deng X, Su R, Feng X, Wei M, Chen J. Role of N(6)-methyladenosine modification in cancer. Curr Opin Genet Dev. 2018;48:1–7.

    CAS  PubMed  Google Scholar 

  37. Teng PC, Liang Y, Yarmishyn AA, Hsiao YJ, Lin TY, Lin TW, Teng YC, Yang YP, Wang ML, Chien CS, et al. RNA modifications and epigenetics in modulation of lung cancer and pulmonary diseases. Int J Mol Sci. 2021;22(19):10592.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, Di W, Hu B, An J, Kong L, 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Li J, Xie G, Tian Y, Li W, Wu Y, Chen F, Lin Y, Lin X, Wing-Ngor AuS, Cao J, et al. RNA m(6)A methylation regulates the dissemination of cancer cells via modulating expression and membrane localization of β-catenin. Mol Ther. 2022. https://doi.org/10.1016/j.ymthe.2022.01.019.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Cayir A, Barrow TM, Guo L, Byun H-M. Exposure to environmental toxicants reduces global N6-methyladenosine RNA methylation and alters expression of RNA methylation modulator genes. Environ Res. 2019;175:228–34.

    CAS  PubMed  Google Scholar 

  41. Cheng C, Wu Y, Xiao T, Xue J, Sun J, Xia H, Ma H, Lu L, Li J, Shi A, 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.

    CAS  PubMed  Google Scholar 

  42. Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 2016;62(3):335–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Choe J, Lin S, Zhang W, Liu Q, Wang L, Ramirez-Moya J, Du P, Kim W, Tang S, Sliz P, et al. mRNA circularization by METTL3-eIF3h enhances translation and promotes oncogenesis. Nature. 2018;561(7724):556–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10(2):93–5.

    CAS  PubMed  Google Scholar 

  45. Szakács G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5(3):219–34.

    PubMed  Google Scholar 

  46. Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019;575(7782):299–309.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol. 2018;29(suppl_1):i10–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale C-M, Zhao X, Christensen J, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316(5827):1039–43.

    CAS  PubMed  Google Scholar 

  49. Ding N, You A, Tian W, Gu L, Deng D. Chidamide increases the sensitivity of non-small cell lung cancer to crizotinib by decreasing c-MET mRNA methylation. Int J Biol Sci. 2020;16(14):2595–611.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu S, Li Q, Li G, Zhang Q, Zhuo L, Han X, Zhang M, Chen X, Pan T, Yan L, et al. The mechanism of m(6)A methyltransferase METTL3-mediated autophagy in reversing gefitinib resistance in NSCLC cells by β-elemene. Cell Death Dis. 2020;11(11):969.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Gao F, Wang Q, Zhang C, Zhang C, Qu T, Zhang J, Wei J, Guo R. RNA methyltransferase METTL3 induces intrinsic resistance to gefitinib by combining with MET to regulate PI3K/AKT pathway in lung adenocarcinoma. J Cell Mol Med. 2021;25(5):2418–25.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Song Z, Jia G, Ma P, Cang S. Exosomal miR-4443 promotes cisplatin resistance in non-small cell lung carcinoma by regulating FSP1 m6A modification-mediated ferroptosis. Life Sci. 2021;276:119399.

    CAS  PubMed  Google Scholar 

  53. Ganapathy-Kanniappan S, Geschwind J-FH. Tumor glycolysis as a target for cancer therapy: progress and prospects. Mol Cancer. 2013;12:152.

    PubMed  PubMed Central  Google Scholar 

  54. Icard P, Shulman S, Farhat D, Steyaert JM, Alifano M, Lincet H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist Updat. 2018;38:1–11.

    PubMed  Google Scholar 

  55. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis. 2017;20(4):409–26.

    CAS  PubMed  Google Scholar 

  57. Yetkin-Arik B, Kastelein AW, Klaassen I, Jansen CHJR, Latul YP, Vittori M, Biri A, Kahraman K, Griffioen AW, Amant F, et al. Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta Rev Cancer. 2021;1875(1):188446.

    CAS  PubMed  Google Scholar 

  58. Chamorro-Jorganes A, Sweaad WK, Katare R, Besnier M, Anwar M, Beazley-Long N, Sala-Newby G, Ruiz-Polo I, Chandrasekera D, Ritchie AA, et al. METTL3 regulates angiogenesis by modulating let-7e-5p and miRNA-18a-5p expression in endothelial cells. Arterioscler Thromb Vasc Biol. 2021;41(6):e325–37.

    CAS  PubMed  Google Scholar 

  59. Wang H, Deng Q, Lv Z, Ling Y, Hou X, Chen Z, Dinglin X, Ma S, Li D, Wu Y, 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.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Yin H, Zhang X, Yang P, Zhang X, Peng Y, Li D, Yu Y, Wu Y, Wang Y, Zhang J, et al. RNA m6A methylation orchestrates cancer growth and metastasis via macrophage reprogramming. Nat Commun. 2021;12(1):1394.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Liu K, Gao Y, Gan K, Wu Y, Xu B, Zhang L, Chen M. Prognostic roles of N6-methyladenosine METTL3 in different cancers: a system review and meta-analysis. Cancer Control. 2021;28:1073274821997455.

    PubMed  PubMed Central  Google Scholar 

  62. Xu F, Zhang H, Chen J, Lin L, Chen Y. Immune signature of T follicular helper cells predicts clinical prognostic and therapeutic impact in lung squamous cell carcinoma. Int Immunopharmacol. 2020;81:105932.

    CAS  PubMed  Google Scholar 

  63. Zhang Y, Liu X, Liu L, Li J, Hu Q, Sun R. Expression and prognostic significance of m6A-related genes in lung adenocarcinoma. Med Sci Monit. 2020;26:e919644.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang H, Zhao X, Lu Z. m(6)A RNA methylation regulators act as potential prognostic biomarkers in lung adenocarcinoma. Front Genet. 2021;12:622233.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. 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.

    PubMed  PubMed Central  Google Scholar 

  66. Liu Y, Guo X, Zhao M, Ao H, Leng X, Liu M, Wu C, Ma J, Zhu J. Contributions and prognostic values of m6 A RNA methylation regulators in non-small-cell lung cancer. J Cell Physiol. 2020;235(9):6043–57.

    CAS  PubMed  Google Scholar 

  67. Zhuang Z, Chen L, Mao Y, Zheng Q, Li H, Huang Y, Hu Z, Jin Y. Diagnostic, progressive and prognostic performance of m(6)A methylation RNA regulators in lung adenocarcinoma. Int J Biol Sci. 2020;16(11):1785–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Coomer AO, Black F, Greystoke A, Munkley J, Elliott DJ. Alternative splicing in lung cancer. Biochim Biophys Acta Gene Regul Mech. 2019;1862(11–12):194388.

    CAS  PubMed  Google Scholar 

  69. Urbanski LM, Leclair N, Anczuków O. Alternative-splicing defects in cancer: splicing regulators and their downstream targets, guiding the way to novel cancer therapeutics. Wiley Interdiscip Rev RNA. 2018;9(4):e1476.

    PubMed  PubMed Central  Google Scholar 

  70. Wu L, Wu D, Ning J, Liu W, Zhang D. Changes of N6-methyladenosine modulators promote breast cancer progression. BMC Cancer. 2019;19(1):326.

    PubMed  PubMed Central  Google Scholar 

  71. Baralle FE, Giudice J. Alternative splicing as a regulator of development and tissue identity. Nat Rev Mol Cell Biol. 2017;18(7):437–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Qin F, Cai B, Zhao J, Zhang L, Zheng Y, Liu B, Gao C. Methyltransferase-like protein 14 attenuates mitochondrial antiviral signaling protein expression to negatively regulate antiviral immunity via N(6) -methyladenosine modification. Adv Sci. 2021;8:e2100606.

    Google Scholar 

  73. Yang F, Yi F, Zheng Z, Ling Z, Ding J, Guo J, Mao W, Wang X, Wang X, Ding X, et al. Characterization of a carcinogenesis-associated long non-coding RNA. RNA Biol. 2012;9(1):110–6.

    CAS  PubMed  Google Scholar 

  74. St Laurent G, Wahlestedt C, Kapranov P. The landscape of long noncoding RNA classification. Trends Genet. 2015;31(5):239–51.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen Y, Lin Y, Shu Y, He J, Gao W. Interaction between N(6)-methyladenosine (m(6)A) modification and noncoding RNAs in cancer. Mol Cancer. 2020;19(1):94.

    PubMed  PubMed Central  Google Scholar 

  76. Yi YC, Chen XY, Zhang J, Zhu JS. Novel insights into the interplay between m(6)A modification and noncoding RNAs in cancer. Mol Cancer. 2020;19(1):121.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Gu S, Sun D, Dai H, Zhang Z. N(6)-methyladenosine mediates the cellular proliferation and apoptosis via microRNAs in arsenite-transformed cells. Toxicol Lett. 2018;292:1–11.

    CAS  PubMed  Google Scholar 

  78. Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen L-L, Wang Y, et al. Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 2017;27(5):626–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Dong L, Chen C, Zhang Y, Guo P, Wang Z, Li J, Liu Y, Liu J, Chang R, Li Y, et al. The loss of RNA N(6)-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8(+) T cell dysfunction and tumor growth. Cancer Cell. 2021;39(7):945–57.

    CAS  PubMed  Google Scholar 

  80. Lin X, Chai G, Wu Y, Li J, Chen F, Liu J, Luo G, Tauler J, Du J, Lin S, et al. RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nat Commun. 2019;10(1):2065.

    PubMed  PubMed Central  Google Scholar 

  81. Zhang Y, Liu S, Zhao T, Dang C. METTL3-mediated m6A modification of Bcl-2 mRNA promotes non-small cell lung cancer progression. Oncol Rep. 2021;46(2):163.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bedi RK, Huang D, Eberle SA, Wiedmer L, Śledź P, Caflisch A. Small-molecule inhibitors of METTL3, the major human epitranscriptomic writer. ChemMedChem. 2020;15(9):744–8.

    CAS  PubMed  Google Scholar 

  83. Chen W-W, Qi J-W, Hang Y, Wu J-X, Zhou X-X, Chen J-Z, Wang J, Wang H-H. Simvastatin is beneficial to lung cancer progression by inducing METTL3-induced m6A modification on EZH2 mRNA. Eur Rev Med Pharmacol Sci. 2020;24(8):4263–70.

    PubMed  Google Scholar 

  84. Li W, Zhang H, Assaraf YG, Zhao K, Xu X, Xie J, Yang DH, Chen ZS. Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies. Drug Resist Updat. 2016;27:14–29.

    CAS  PubMed  Google Scholar 

  85. Li F, Wang H, Huang H, Zhang L, Wang D, Wan Y. m6A RNA methylation regulators participate in the malignant progression and have clinical prognostic value in lung adenocarcinoma. Front Genet. 2020;11:994.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Losanno T, Rossi A, Maione P, Napolitano A, Gridelli C. Anti-EGFR and antiangiogenic monoclonal antibodies in metastatic non-small-cell lung cancer. Expert Opin Biol Ther. 2016;16(6):747–58.

    CAS  PubMed  Google Scholar 

  87. Tang H, Liu Y, Wang C, Zheng H, Chen Y, Liu W, Chen X, Zhang J, Chen H, Yang Y, et al. Inhibition of COX-2 and EGFR by melafolone improves anti-PD-1 therapy through vascular normalization and PD-L1 downregulation in lung cancer. J Pharmacol Exp Ther. 2019;368(3):401–13.

    CAS  PubMed  Google Scholar 

  88. Zhang Z, Zhang Y, Luo F, Ma Y, Fang W, Zhan J, Li S, Yang Y, Zhao Y, Hong S, et al. Dual blockade of EGFR and VEGFR pathways: results from a pilot study evaluating apatinib plus gefitinib as a first-line treatment for advanced EGFR-mutant non-small cell lung cancer. Clin Transl Med. 2020;10(2):e33.

    PubMed  PubMed Central  Google Scholar 

  89. Qiu T, Zhou L, Zhu W, Wang T, Wang J, Shu Y, Liu P. Effects of treatment with histone deacetylase inhibitors in solid tumors: a review based on 30 clinical trials. Future Oncol. 2013;9(2):255–69.

    CAS  PubMed  Google Scholar 

  90. Tang S-W, Thomas A, Murai J, Trepel JB, Bates SE, Rajapakse VN, Pommier Y. Overcoming resistance to DNA-targeted agents by epigenetic activation of schlafen 11 ( expression with class I histone deacetylase inhibitors. Clin Cancer Res. 2018;24(8):1944–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Guan W, Jing Y, Dou L, Wang M, Xiao Y, Yu L. Chidamide in combination with chemotherapy in refractory and relapsed T lymphoblastic lymphoma/leukemia. Leuk Lymphoma. 2020;61(4):855–61.

    CAS  PubMed  Google Scholar 

  92. Mao J, Li S, Zhao H, Zhu Y, Hong M, Zhu H, Qian S, Li J. Effects of chidamide and its combination with decitabine on proliferation and apoptosis of leukemia cell lines. Am J Transl Res. 2018;10(8):2567–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Guan XW, Wang HQ, Ban WW, Chang Z, Chen HZ, Jia L, Liu FT. Novel HDAC inhibitor Chidamide synergizes with Rituximab to inhibit diffuse large B-cell lymphoma tumour growth by upregulating CD20. Cell Death Dis. 2020;11(1):20.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Mu X, Zhao Q, Chen W, Zhao Y, Yan Q, Peng R, Zhu J, Yang C, Lan K, Gu X, et al. IL-37 confers anti-tumor activity by regulation of m6A methylation. Front Oncol. 2021;10:526866.

    PubMed  PubMed Central  Google Scholar 

  95. Selberg S, Blokhina D, Aatonen M, Koivisto P, Siltanen A, Mervaala E, Kankuri E, Karelson M. Discovery of small molecules that activate RNA methylation through cooperative binding to the METTL3-14-WTAP complex active site. Cell Rep. 2019;26(13):3762-3771.e3765.

    CAS  PubMed  Google Scholar 

  96. Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Yang F, Yuan W-Q, Li J, Luo Y-Q. Knockdown of METTL14 suppresses the malignant progression of non-small cell lung cancer by reducing Twist expression. Oncol Lett. 2021;22(6):847.

    PubMed  PubMed Central  Google Scholar 

  98. Li F, Zhao J, Wang L, Chi Y, Huang X, Liu W. METTL14-mediated miR-30c-1-3p maturation represses the progression of lung cancer via regulation of MARCKSL1 expression. Mol Biotechnol. 2022;64(2):199–212.

    CAS  PubMed  Google Scholar 

  99. Little NA, Hastie ND, Davies RC. Identification of WTAP, a novel Wilms’ tumour 1-associating protein. Hum Mol Genet. 2000;9(15):2231–9.

    CAS  PubMed  Google Scholar 

  100. Weng L, Qiu K, Gao W, Shi C, Shu F. LncRNA PCGEM1 accelerates non-small cell lung cancer progression via sponging miR-433-3p to upregulate WTAP. BMC Pulm Med. 2020;20(1):213.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Sorci M, Ianniello Z, Cruciani S, Larivera S, Ginistrelli LC, Capuano E, Marchioni M, Fazi F, Fatica A. METTL3 regulates WTAP protein homeostasis. Cell Death Dis. 2018;9(8):796.

    PubMed  PubMed Central  Google Scholar 

  102. Wei W, Huo B, Shi X. miR-600 inhibits lung cancer via downregulating the expression of METTL3. Cancer Manag Res. 2019;11:1177–87.

    PubMed  PubMed Central  Google Scholar 

  103. Li X, Li N, Huang L, Xu S, Zheng X, Hamsath A, Zhang M, Dai L, Zhang H, Wong JJ-L, et al. Is hydrogen sulfide a concern during treatment of lung adenocarcinoma with ammonium tetrathiomolybdate? Front Oncol. 2020;10:234.

    PubMed  PubMed Central  Google Scholar 

  104. Liu P, Li F, Lin J, Fukumoto T, Nacarelli T, Hao X, Kossenkov AV, Simon MC, Zhang R. m(6)A-independent genome-wide METTL3 and METTL14 redistribution drives the senescence-associated secretory phenotype. Nat Cell Biol. 2021;23(4):355–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Wu H, Li F, Zhu R. miR-338-5p inhibits cell growth and migration via inhibition of the METTL3/m6A/c-Myc pathway in lung cancer. Acta Biochim Biophys Sin. 2021;53(3):304–16.

    CAS  PubMed  Google Scholar 

  106. Gong C, Fan Y, Liu J. METTL14 mediated m6A modification to LncRNA ZFAS1/ RAB22A: a novel therapeutic target for atherosclerosis. Int J Cardiol. 2021;328:177.

    PubMed  Google Scholar 

  107. Yankova E, Blackaby W, Albertella M, Rak J, De Braekeleer E, Tsagkogeorga G, Pilka ES, Aspris D, Leggate D, Hendrick AG, et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature. 2021;593(7860):597–601.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Shandong Provincial Natural Science Foundation (Grant No. ZR2020MH204), the 19th batch of science and technology innovation development plan of Jinan in 2020 (Clinical medicine science and technology innovation plan, Grant No. 202019032), and the second group of science and technology projects of Jinan Health Committee (Grant No. 2020-3-15).

Author information

Authors and Affiliations

Authors

Contributions

Z-GS and NZ designed the work. CM wrote the manuscript. KH and R-JM prepared the figures and tables. Q-MZ and Y-PW drafted and revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Nan Zhang or Zhi-Gang Sun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, C., Ma, RJ., Hu, K. et al. The molecular mechanism of METTL3 promoting the malignant progression of lung cancer. Cancer Cell Int 22, 133 (2022). https://doi.org/10.1186/s12935-022-02539-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-022-02539-5

Keywords