Open Access

METTL14‑mediated RNA methylation in digestive system tumors

  • Authors:
    • Jiexuan Hu
    • Haishan Lin
    • Cong Wang
    • Qiang Su
    • Bangwei Cao
  • View Affiliations

  • Published online on: August 4, 2023     https://doi.org/10.3892/ijmm.2023.5289
  • Article Number: 86
  • Copyright: © Hu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

N6‑methyladenosine (m6A) RNA methylation is one of the most common post‑transcriptional modification mechanism in eukaryotes. m6A is involved in almost all stages of the mRNA life cycle, specifically regulating its stability, splicing, export and translation. Methyltransferase‑like 14 (METTL14) is a particularly important m6A methylation ‘writer’ that can recognize RNA substrates. METTL14 has been documented to improve the activity and catalytic efficiency of METTL3. However, as individual proteins they can also regulate different biological processes. Malignancies in the digestive system are some of the most common malignancies found in humans, which are typically associated with poor prognoses with limited clinical solutions. METTL14‑mediated methylation has been implicated in both the potentiation and inhibition of digestive system tumor growth, cell invasion and metastasis, in addition to drug resistance. In the present review, the research progress and regulatory mechanisms of METTL14‑mediated methylation in digestive system malignancies were summarized. In addition, future research directions and the potential for its clinical application were examined.

1. Introduction

Epigenetics is the study of heritable phenotypes that do not involve changes in the actual nuclear DNA sequence (1-4). Examples of known epigenetic modulations include DNA methylation, expression interference by non-coding RNAs (ncRNA) and histone modifications (5-8). Genetic information is encoded by DNA, which is transcribed into coding mRNA or non-coding RNA. mRNA is then translated into functional proteins (9). Epigenetic inheritance not only serves a role in transcription and translation, but is also involved in post-transcriptional mRNA regulation (10).

Some of the most commonly studied RNA methylation modification processes include N6-methyladenosine (m6A), 5-methylcytosine (m5C) and 7-methylguanosine (m7G) (11). m6A is the most common epigenetic modification mechanism of RNA molecules, which was first identified in eukaryotic mRNAs in the early 1970s (12). In 2011, it was then discovered that the m6A process is reversible, when fat mass and obesity-associated (FTO) protein was shown to function as an m6A mRNA demethylase (13).

m6A methylation was subsequently found to occur in all species of eukaryotes, prokaryotes and viruses (14,15). m6A modifications can occur on mRNAs, ribosomal (rRNAs), small nuclear (snRNAs) and microRNAs (miRNAs) (16). To date, m6A sites have been identified in >7,000 coding RNAs and 300 non-coding RNAs (17,18). Functionally, m6A can regulate almost all processes during the mRNA life cycle, including stability, splicing, export and translation (19,20). m6A modifications generally occur on 'RRACH' RNA sequences, where R can be A or G bases and H can be A, C or U bases (21). Qualcomm test sequencing previously revealed that the m6A signals typically clustered near the 3'-untranslated region (UTR) and stop codons (22). This likely aids in maintaining the structural stability of the mRNA (22). By contrast, clustering near the 5'-UTR or long internal exons is considered to be involved in mRNA splicing, translation and degradation (23-25). Specifically, YTH domain-family proteins (YTHD) 2 can bind m6A in the 5'-UTR of mRNAs, promoting their cap-independent translation. mRNAs containing the m6A modification in the 5'-UTR can be identified to promote cap-independent translation initiation by direct binding to eukaryotic initiation factor (eIF)3 (23).

At present, m6A regulators can be classified into methyltransferases ('writers'), demethylases ('erasers) and RNA-binding proteins ('readers') (26). Specifically, 'Writers' catalyze m6A methylation and include methyltransferase-like (METTL) proteins, such as METTL3 (27-29), METTL14 (30,31), METTL16 (32,33), Wilms tumor 1-associated protein (WTAP) (26,34,35), RNA-binding motif protein 15 (26) and Vir-like m6A methyltransferase (36-38). Alternatively, 'erasers', such as FTO, AlkB homolog (ALKBH)3 and ALKBH5, generally catalyze the demethylation of m6A residues in an iron (II) and α-ketoglutarate-dependent manner (39-42). The renders m6A methylation to be a dynamically reversible process (10). The role of 'readers' are to identify the methylation modifications and transmit regulatory signals to downstream functional proteins to facilitate various biological processes, such as RNA splicing, stabilization, export and mRNA translation (21,43). Such 'readers' include YTHDC1/2 and YTHDF1/2/3 (44,45), heterogenous nuclear ribonucleoproteins (hnRNP)C, hnRNPG and hnRNPA2B1 (46), eIF3 (38) and insulin-like growth factor-2 mRNA-binding proteins (IGF2BP; Fig. 1) (44,47).

2. METTL3/METTL14-mediated RNA methylation

The m6A methylation 'writer' METTL14 has been characterized to be a weak methyltransferase that cannot catalyze methylation independently (6). Complete catalysis is achieved by the METTL3/METTL14 complex, a stoichiometrically 1:1 heterodimer (48). These two methylase components were initially discovered by isolating nuclear extracts from HeLa cells, such that the key 70-kDa METTL3 subunit was extracted from the 200-kDa mRNA adenosine methylase complex (49). Through its open methyltransferase domain (MTD), the internal DPPW motif on METTL3 binds to the free methyl donor S-adenosylmethionine methionine (SAM) to generate the metabolite S-adenosine homocysteine (SAH). SAH then competitively regulates methylation homeostasis, by promoting the association of the tyrosine residue Y406 on the METTL3 with the mRNA (50,51). By contrast, METTL14 also contains an MTD, but its EPPL motif that confers catalytic cavity adopts a closed conformation, obstructing its access to SAM (52,53). Additionally, METTL3 has a unique Cys-Cys-Cys-His (CCCH)-type zinc-binding motif, which has been reported to bind nucleic acids and be required for RNA methyltransferase activity (50,54,55). The current working model of this METTL3/METTL14 complex is that the main function of METTL14 is to recognize the RNA substrate whilst enhancing the activity and catalytic efficiency of METTL3 (52). However, METTL3 and METTL14 have also been found to serve opposing regulatory roles in cancer. One previous study found the opposite expression and prognostic value of METTL3 and METTL14 in hepatocellular carcinoma (HCC) (56). In addition, separately downregulating the expression of METTL3 and METTL14 resulted in the alteration of distinct mRNA signaling pathways and biological processes in HCC (56). Apart from METTL3/METTL14 dimerization, WTAP can contribute to METTL3/METTL14 complex stabilization and localization to the nucleus (57,58).

3. METTL3/METTL14 and digestive system cancer

Lin et al (59) previously found that in gallbladder cancer, deoxycholic acid can bind to the METTL3/METTL14 complex, promoting METTL3 separation to enhance the expression of miR-92b-3p (59). Furthermore, another previous study on liver cancer found that thiamine treatment enhanced METTL3 acetylation, which weakened its binding to METTL14 to reduce the expression of metal regulatory transcription factor (MTF1) mRNA (60). However, it also extended the half-life of this mRNA transcript, which in turn promoted MTF1 protein expression (60). Overexpression of MTF1 was therefore proposed to be a marker of HCC proliferation and consequently poor prognosis.

4. Roles of METTL14 in digestive system cancer

During neoplastic transformation, m6A modification can regulate tumor development and progression by modulating the structures of oncogene and suppressor gene mRNAs (4,61-63). Recently, various studies have reported that METTL14 can serve a diverse array of roles in digestive system malignancies, including regulation of proliferation, invasion, metastasis, angiogenesis and drug resistance. Therefore, current research progress (until December 2022) on the functions of METTL14 in digestive system tumors was summarized in this section and Table I.

Table I

Roles of METTL14 in digestive system tumors.

Table I

Roles of METTL14 in digestive system tumors.

CancerTargetRoleImpact on the targetMechanismFunctions in cancer(Refs.)
Gastric cancerWnt/PI3K/AktTumor suppressorKnockdown of METTL14 activates Wnt and PI3K-Akt signalingRegulate Wnt and PI3K/Akt signalingProliferation and invasion(67)
PI3K/AKT/mTORTumor suppressorKnockdown of METTL14 increases the phosphorylation levels of PI3K, AKT and mTORRegulate the PI3K/AKT/mTOR pathwayProliferation, migration, and invasion(68)
PTENTumor suppressorOverexpression of METTL14 increases the expression of PTENStabilized PTEN mRNAGrowth and metastasis(69)
CircORC5Tumor suppressorKnockdown of METTL14 increases the protein expression of circORC5Regulate miR-30c-2-3p/AKT1 substrate 1 axisGrowth and invasion(70)
LHPPTumor suppressorKnockdown of METTL14 decreases the expression of LHPPMediate HIF1A to inhibit glycolysisGlycolysis, proliferation, invasion, and metastasis(71)
p75NTRTumor suppressorOverexpression of METTL14 increases the expression of p75NTRIbuprofen increased m6A-p53 expressionGrowth(72)
LINC01320OncogeneOverexpression of METTL14 increases the expression of LINC01320Regulate the miR-495-5p/RAB19 axisProliferation, migration, and invasion(73)
Liver cancermiR-126Tumor suppressorOverexpression of METTL14 increases the expression of miR-126Interact with the microprocessor protein DiGeorge syndrome critical region 8Metastasis(79)
p53Tumor suppressorKnockdown of METTL14 decreases the p53 translationFusaric acid decreased p53 expression-(81)
EGFRTumor suppressorKnockdown of METTL14 increases the phosphorylation of EGFRModulate EGFR/PI3K/AKT signaling pathwayMigration, invasion, and EMT(82)
HNF3γTumor suppressorKnockdown of METTL14 decreases the expression of HNF3γTransactivation of OATP1B1 and OATP1B3 expressionDedifferentiation and sorafenib resistance(83)
USP48Tumor suppressorOverexpression of METTL14 increases the expression of USP48Regulate the Sirtuin 6 signaling pathwayGlycolysis and malignancy(84)
SLC7A11Tumor suppressorKnockdown of METTL14 increases the expression of SLC7A11HIF-1α/METTL14/YTH DF2/SLC7A11 axisFerroptosis of hepatocellular carcinoma(85)
ACLY/SCD1OncogeneOverexpression of METTL14 increases the expression of ACLY/SCD1N/AFatty acid synthase expression(86)
MIR155HGOncogeneOverexpression of METTL14 increases the expression of MIR155HGRegulate programmed death-ligand 1 expression via miR-223-3p/STAT1 axisImmune escape(87)
ARHGAP5-AS1OncogeneKnockdown of METTL14 decreases the expression of ARHGAP5-AS1Promote translation of vimentin and RAC1 activate the ERK pathwayInvasion and metastasis(88)
CircFUT8OncogeneOverexpression of METTL14 increases the m6A level of CircFUT8Regulate the circFUT8/miR-552-3p/charged multivesicular body protein 4B pathwayGrowth(89)
Colorectal cancerANKLE1Tumor suppressorKnockdown of METTL14 decreases the expression of ANKLE1Recognized by YTHDF1Proliferation and colony formation(92)
LncRNA XISTTumor suppressorKnockdown of METTL14 increases the expression of LncRNA XISTYTHDF2 mediate the degradation of XISTProliferation and metastasis(93)
SOX4Tumor suppressorOverexpression of METTL14 decreases the expression of SOX4SOX4-mediated EMT process and PI3K/Akt signalsMigration, invasion, and metastasis(94)
KLF4Tumor suppressorKnockdown of METTL14 decreases the expression of KLF4MethylCpG-binding protein 2 interacted with METTL14Migration and invasion(95)
ARRDC4Tumor suppressorOverexpression of METTL14 decreases the expression of ARRDC4Transcription factor 4/Hu antigen receptor/METTL14/YHTDF2/ARRDC4/zinc finger E-box binding homeobox 1 axisMetastasis(96)
STAT1/IRF1Tumor suppressorKnockdown of METTL14 promotes the STAT1/IRF1 signalingPromote IFN-γ/STAT1/IRF1 signaling via YTHDF2Immune responses to anti-PD-1 therapy(97)
EbI3Tumor suppressorKnockdown of METTL14 increases the expression of EbI3Trigger a shift of intratumoral CD8+ T cells toward a dysfunctional stateGrowth and impedes CD8+ T cell infiltration(98)
miR-149-3pTumor suppressorKnockdown of METTL14 decreases the expression of miR-149-3pPHD finger protein 5A transactivated superoxide dismutase 2 through regulating lysine acetyltransferase 2A mRNA alternative splicing by enterotoxigenic bacteroides fragilisInflammation and malignancy(99)
PHLDB2OncogeneOverexpression of METTL14 increases the expression of PHLDB2Stabilize EGFR and promotes its nuclear translocationCetuximab resistance(100)
Pancreatic cancerPERPOncogeneOverexpression of METTL14 decreases the expression of PERPIncrease PERP mRNA turnoverProliferation and migration(104)
miR-380-3pOncogeneKnockdown of METTL14 decreases the expression of miR-380-3pmiR-380-3p/PTEN/Akt pathwayProliferation, EMT and tumorigenesis(105)
AMPKα/ERK/mTOROncogeneKnockdown of METTL14 activates AMPKα/ERK/mTOR signalingEnhance apoptosis and autophagyCisplatin resistance(106)
CDAOncogeneOverexpression of METTL14 increases the expression of CDAp65 targeted the promoter region of METTL14Gemcitabine resistance(108)
PIK3CBTumor suppressorOverexpression of PIK3CB decreases the expression of METTL14Activate AKT signaling pathwayProliferation and invasion(109)
CLK1/Serine/arginine-rich splicing factor 5Tumor suppressorOverexpression of CLK1 increases the m6A level of METTL14Control the exon skipping of METTL14 and Cyclin L2Proliferation and metastasis(110)
CholangiocarcinomaSTIM2Tumor suppressorKnockdown of METTL14 decreases the expression of STIM2STIM2-keratin 8 regulatory paradigmMetastasis(111)

[i] METTL, methyltransferase-like; circ, circular RNA; Orc, origin recognition complex 5; LHPP, phospholysine phosphohistidine inorganic pyrophosphate phosphatase; HIF1α, Hypoxia-inducible factor 1α; p75 NTR, p75 neurotrophin receptor; miR, microRNA; LINC, long intergenic non-coding RNA; HNF3γ, hepatocyte nuclear factor 3γ; OATP, solute carrier organic anion transporter family member; USP48, ubiquitin-specific peptidase 48; SLC7A11, solute carrier family 7, member 11; YTHD, YTH domain-containing/family protein; ACLY, ATP citrate lyase; SCD1, Stearoyl-CoA 9-desaturase; MIR155HG, miR-155 host gene; ARHGAP5-AS1, rho GTPase-activating protein 5-antisense 1; FUT8, fucosyltransferase 8; ANKLE1, LEM domain-containing protein 1; XIST, X-inactive specific transcript; EMT, epithelial-mesenchymal transition; IRF1, IFN regulatory factor 1; KLF4, Kruppel-like factor 4; EbI3, Epstein-Barr virus-induced protein 3; PHLDB2, pleckstrin homology-like domain family B member 2; ARRDC4, arrestin Domain-Containing 4; PERP, p53 effector related to peripheral myelin protein 22; CDA, Cytidine Deaminase; PIK3B, PI3K catalytic subunit; CLK1, Cell division cycle 2-like kinase 1; STIM2, stromal interaction molecule 2.

Gastric cancer

To date, great progress has been made in the development of treatment methods for gastric cancer (GC). Immuno- and targeted therapies have contributed favorably to the prognosis of patients with GC. However, the mortality rate from GC remains to rank highly in China, ranking second after lung cancer in 2022. This is due in part to its high propensity for metastasis and invasion (64,65). m6A RNA methylation has been previously associated with the occurrence and aggressiveness of GC (66).

Zhang et al (67) found that knocking down METTL14 expression can activate Wnt and PI3K/Akt signaling, which in turn promoted GC cell proliferation and invasion (67). METTL14 downregulation also resulted in the increased levels of secreted IFN-α/β/γ proteins in the HGC27 cell line. As such, it was speculated that RNA m6A methylation can regulate the immune response to GC by inhibiting IFN production. In addition, another study previously reported that in GC tissues, METTL14 expression and m6A levels were lower compared with those in adjacent tissues (68). By contrast, METTL14 overexpression was found to inhibit GC cell proliferation and invasion in vitro (68). Mechanistically, METTL14 downregulation increased the phosphorylation levels of PI3K, AKT and mTOR, whilst increasing the expression of vimentin, N-cadherin and MMP9, but decreased the expression of E-cadherin (68). Taken together, these findings suggest that METTL14 overexpression may inhibit the progression and aggressiveness of GC cells by inactivating the PI3K/AKT/mTOR and epithelial-mesenchymal transition (EMT) pathways. Yao et al (69) previously demonstrated that significantly lower METTL14 expression levels in stomach adenocarcinoma (STAD) tissues are associated with poorer prognosis (69). Overexpression of METTL14 was observed to enhance the stability of PTEN mRNA in a m6A modification-dependent manner, which prolonged its half-life (69). Further in vivo and in vitro experiments revealed that this METTL14-enhanced PTEN expression also inhibited the proliferative and invasive capabilities of STAD cells (69). IGF2BP2 and IGF2BP3 were also shown to be 'readers' that can interact with the PTEN mRNA (69). Fan et al (70) previously found that METTL14 expression is decreased in GC tissue samples, which is associated with poorer survival (70). Using methylation-RNA immunoprecipitation, Fan et al (70) showed that METTL14 can mediate the m6A modification of hsa_circ_0007612, also known as circular RNA (circ) origin recognition complex 5 (ORC5). Further investigations revealed that the downregulation of METTL14 lead to the upregulation of circORC5 expression. This increase in circORC5 expression then counteracted the inhibitory effects of miR-30c-23p on AKT1 substrate 1 and eIF4B, which increased the proliferation and invasion of GC cells (70). In addition, another study previously found that m6A modification of the phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) mRNA can be regulated by METTL14 in GC cells, where METTL14 correlated positively with LHPP with significance. Increasing the expression of LHPP was found to effectively inhibit the proliferation, invasion and migration of GC both in vitro and in vivo (71). LHPP overexpression can also inhibit the Wnt signaling pathway, which consequently inhibits the phosphorylation of GSK-3β and aerobic glycolysis in GC cells (71). Jin et al (72) reported that ibuprofen treatment can promote the expression of METTL3 and METTL14 in GC cells. METTL3 and METTL14 can also promote p53 translation, which in turn epigenetically increases p75 neurotrophin receptor (p75NTR) expression (72). p75NTR is considered to be a potential tumor suppressor that is associated with proliferation inhibition (72). Therefore, p75NTR may prove to be promising novel therapeutic target in the future.

The majority of research findings to date appear to suggest METTL14 to be a tumor suppressor in GC (67-72). However, data suggesting that METTL14 can function as an oncogene also exist. Hu and Ji (73) reported that METTL14-mediated m6A modification can upregulate long intergenic non-coding RNA 01320 expression, which promoted the proliferation, migration and invasion of GC cells by increasing the expression of miR-495-5p whilst decreasing that of RAB19 (73).

Liver cancer

HCC is the fourth leading cause of cancer-related mortality worldwide, accounting for >80% of all cases of primary liver cancer (74,75). In total, 6% of China's population is affected by hepatitis B infection and/or HCC (76). The low 5-year survival and high mortality rates for HCC are largely due to metastasis, poor response to chemotherapy and frequent postoperative recurrence (77). Epigenetic studies are therefore important for the discovery of novel therapeutic targets for HCC.

Zhou et al (78) previously found that METTL14 expression is significantly lower in HCC tissues, which is an independent prognostic factor for overall postoperative survival and tumor-free survival (78). Ma et al (79) reported that METTL14 can regulate the m6A modification of miR-126 by interacting with the microprocessor protein, DiGeorge syndrome critical region 8 (79). Functionally, miR-126 expression is significantly reduced in HCC cells, the overexpression of which was found to inhibit the metastatic effect of METTL14-deficiency in vitro (79). In HCC, expression of the tumor suppressor p53 can also be regulated by METTL14 (80). A study previously found that fusaric acid (FA) can reduce the expression of METTL14 to epigenetically suppress p53 expression in the HepG2 cell line (81). In addition, using RNA-sequencing and m6A-sequencing, Shi et al (82) found that EGFR is a downstream target of METTL14 in HCC. METTL14 expression is downregulated in HCC, which can activate the phosphorylation of EGFR and subsequent PI3K/AKT signaling to promote migration, invasion and EMT (82). METTL14 can indirectly promote sorafenib resistance by decreasing hepatocyte nuclear factor 3γ (HNF3γ) mRNA expression through m6A. HNF3γ can promote hepatic cancer stem cell differentiation into HCC cells, inhibit the proliferation of HCC and sensitize HCC cells to sorafenib-induced and apoptosis by activating the expression of sorafenib membrane transporters solute carrier organic anion transporter family member (OATP)1B1 and OATP1B3 (83). Mechanistically, the increased HNF3γ mRNA stability were likely to be mediated through IGF2BPs (83). In addition, Du et al (84) demonstrated that m6A modification induced by METTL14 increased the stability of ubiquitin-specific peptidase 48 (USP48) mRNA to upregulate its expression. USP48 then stabilized sirtuin 6 proteins to prevent metabolic reprogramming from mitochondrial respiration toward aerobic glycolysis, then enhanced glycolytic homeostasis and inhibited tumor growth (84). Another study previously found that ferroptosis was blocked in HCC cells under hypoxic conditions due to the inhibition of METTL14 by hypoxia-inducible factor 1-alpha (HIF-1α) (85). Further investigation revealed that METTL14 can negatively regulate the expression of solute carrier family 7, member 11 (SLC7A11) through the YTHDF2-dependent pathway. Downregulation of SLC7A11 resulted in ferroptosis, inhibiting the progression of HCC (85). Therefore, the HIF-1α/METTL14/YTHDF2/SLC7A11 axis may provide a basis for the treatment design of HCC.

However, at present there is no unanimous consensus on the role of METTL14 in liver cancer. A number of studies have suggested that METTL14 can serve as an oncogene in liver cancer. Yang et al (86) found that the levels of METTL14 and METTL3 expression are higher in HCC cell lines and human HCC tissue samples (86). Overexpression of METTL14 directly increased m6A levels on the ATP citrate lyase and stearyl-CoA desaturase 1 mRNA transcripts in HCC cells. This enhanced the production of triglycerides and cholesterol, in addition to the accumulation of lipid droplets in HCC, leading to HCC proliferation, angiogenesis and the HCC progression (86). The pathogenesis of HCC is associated with pathogen-associated molecular patterns, where lipopolysaccharides (LPS) can enter the liver through the entero-liver axis to promote the progression of HCC. LPS may be one of the regulators upregulated by METTL14 in HCC (87). LPS can upregulate METTL14 to enhance the m6A methylation level of miR-155 host gene (MIR155HG), which then stabilizes the MIR155HG mRNA through the 'reader' protein abnormal vision-like 1. In addition, LPS can increase the expression of programmed death-ligand 1 (PD-L1) by upregulating the expression of miR-155HG (87). In addition, overexpression of MIR155HG can inhibit the expression of miR-223-3p, which then increases the expression of STAT1 and the expression of PD-L1 in HCC. Based on the m6A-sequencing data from HCC cells, Liu et al (88) previously observed that rho GTPase-activating protein 5 (ARHGAP5)-antisense (AS)1 is the long non-coding RNA (lncRNA) showing the highest level of m6A modification. Silencing METTL14 expression was found to reduce m6A and ARHGAP5-AS1 expression (88). Furthermore, inhibition of cold shock domain-containing E1 degradation by ARHGAP5-AS1 was found to promote the translation of vimentin and Ras-related C3 botulinum toxin substrate 1 (RAC1) to activate the ERK pathway in HCC. ERK activation then in turn promoted HCC cell proliferation, migration and invasion (88). Another previous study revealed that miR-628-5p in the exosomes secreted by M1 macrophages can inhibit expression of METTL14 in HCCs following transportation (89). The m6A modification of Circ-fucosyltransferase 8 (circFUT8) is positively regulated by METTL14, where CircFUT8 is upregulated in HCC cells. CircFUT8 overexpression can inhibit HCC apoptosis whilst promoting cell proliferation through the miR-552-3p/charged multivesicular body protein 4B pathway (89).

Colorectal cancer (CRC)

CRC is one of the most common malignancies in adults, ranking third in the cases of cancer-related mortality worldwide (90) in 2020. The mortality rate in patients with CRC remains high largely due to high rates of metastasis and recurrence (65,91). Studying epigenetics can provide insight into the mechanisms regulating the progression of colorectal cancer.

LEM domain-containing protein 1 (ANKLE1) is a CRC susceptibility gene, where ANKLE1 m6A methylation is catalyzed by the METTL3/METTL14/WTAP complex and then recognized by YTHDF1 (92). Presence of the rs8100241(A) allele has been documented to increase ANKLE1 m6A methylation (92). Methylated ANKLE1 inhibits tumor growth, maintains genomic stability and reduces CRC risk (92). Yang et al (93) found that the downregulation of METTL14 expression is associated with poorer prognosis in patients with CRC (93). LncRNA X-inactive specific transcript (XIST) is a downstream target of METTL14, where knocking down METTL14 expression was found to reduce the level of m6A modification and enhance XIST expression in CRC cells (93). METTL14 can also mediate XIST degradation through the YTHDF2-dependent m6A modification pathway. XIST degradation suppresses the proliferation, invasion and migration of CRC cells (93). Another study found that the demethylation of histone H3 in the METTL14 gene promoter region mediated by lysine-specific histone demethylase 5C (KDM5C) can inhibit METTL14 transcription (94). In addition, METTL14 has been shown to promote the degradation of SOX4 mRNA through a YTHDF2-dependent pathway (94). This suppression of SOX4 in turn inhibits the migration and invasion of CRC cells, since SOX4 can normally activate the EMT process and PI3K/Akt signaling in CRC cells (94). MethylCpG-binding protein 2 (MeCP 2) was also found to inhibit METTL14 in CRC cells (95). Specifically, MeCP2 expression was found to be upregulated in CRC, whilst that of METTL14 was downregulated. MeCP2 and METTL14 can interact and specifically recognize Kruppel-like factor 4 (KLF4) mRNA through the 'reader' protein IGF2BP2 (95). The tumor suppressor KLF4 has been previously demonstrated to inhibit the migration and invasion of CRC. Binding of MeCP2 and METTL14 can inhibit the translation of the KLF4 mRNA to promote the metastasis of CRC (95). Wang et al (96) found that Hu antigen receptor (HuR) can inhibit METTL14 mRNA expression by binding to its promoter in CRC. Transcription factor 4 (TCF4) deficiency was also found to increase the ubiquitination of METTL14, thereby promoting its degradation. METTL14 can itself inhibit downstream arrestin domain-containing 4/zinc finger E-box binding homeobox 1 (ZEB1) signaling through YTHDF2-dependent m6A modification, which prevents colorectal cancer metastasis (96).

In addition to demonstrating its function in CRC proliferation, invasion and migration, a number of studies have also reported METTL14 to regulate other biological processes. Wang et al (97) found that decreased expression of METTL3 and METTL14 can increase IFN-γ/STAT1/IFN regulatory factor 1 (IRF1) signaling by stabilizing IFN-γ, C-X-C motif chemokine ligand (CXCL)9 and CXCL10 secreted by tumor-infiltrating CD8+ cytotoxic T cells in the tumor microenvironment. This then increases the IFN content in the tumor microenvironment and increase the recruitment of immune cells (97). IFN-γ/STAT1/IRF1 signaling may provide insight into the mechanism of poor response to immune checkpoint inhibitor (ICIs) therapy in mismatch-repair-proficient or microsatellite low instability CRC (97). Another recent study found that the downregulation of METTL14 m6A methylation in C1q+ tumor-associated macrophages can increase the levels of the cytokine IL-35 subunit Epstein-Barr virus-induced protein 3 (EBI3) mRNA transcript. This EBI3 elevation in turn promoted CD8+ T cell differentiation into a dysfunctional state, which have weaker proliferative capabilities and reduced response potency to tumors, leading to CRC progression (98). Consistent with the findings in liver cancer, METTL14 was also found to regulate miRNA expression by modifying pri-miRNA splicing in CRC (99). Enterotoxigenic bacteroides fragilis in the gut flora was shown to positively regulate the expression of pri-miR-149 through METTL14-mediated m6A methylation in CRC (99). Reducing miR-149-3p expression in the plasma exosomes of patients with CRC was found to increase proliferation and induce an inflammatory response in CRC cells (99). Functionally, miR-149-3p increases the expression of the PHD finger protein 5A gene downstream to stabilize the splicing factor 3b complex and transactivate superoxide dismutase 2 (SOD2), by regulating lysine acetyltransferase 2A mRNA alternative splicing and increase the expression of SOD2 mRNA, to induce colorectal carcinogenesis (99). Another previous study documented that oxidative stress induced by chemotherapeutic drugs can upregulate the methylation of pleckstrin homology-like domain family B member 2 (PHLDB2) by METTL14 in CRC, which increases protein expression (100). Nuclear EGFR is a transcriptional cofactor that can function to regulate gene expression and promote DNA repair (100). Increased PHLDB2 expression can stabilize EGFR and promotes its nuclear translocation, which promotes cetuximab resistance (100). Rab11A is a common EGFR recycling protein that facilitate EGFR translocation back to the cell membrane, the mechanism by which PHLDB2 can promote the nuclear translocation of EGFR may be mediated by reducing the binding affinity between EGFR and Rab11A (100).

Pancreatic cancer (PC)

PC is one of the most aggressive malignancies, with a 5-year mortality rate of ~95% after diagnosis (65). Additionally, majority of patients with PC do not survive for >7 years following surgical treatment (101,102). For patients with advanced pancreatic cancer, chemotherapy or chemo-radiotherapy is typically applied, but chemotherapy resistance frequently leads to worse prognosis (103). Unlike other digestive system malignancies, existing evidence actually suggest METTL14 to be an oncogene in PC.

A previous study showed that the overexpression of METTL14 in PC cells can increase methylation of p53 effector related to peripheral myelin protein 22 (PERP) downstream. PERP mRNA methylation reduces PERP protein expression, which promotes PC cell proliferation and migration (104). Decreased m6A modification of METTL3 and METTL14 inhibits the expression of miR-380-3p in PC cells, which was demonstrated by Jiang et al (105). By contrast, the expression level of miR-380-3p was previously found to be significantly higher in PC tissues and cells. Further experiments found that miR-380-3p can promote PC cell proliferation and tumorigenesis through Akt signaling, EMT by suppressing PTEN expression (105). In addition, drug-resistant PC tissues tend to express higher levels of METTL14 according to Kong et al (106). Accordingly, knocking down METTL14 expression was found to enhance both apoptosis and autophagy in PC cells through activating 5'AMP-activated protein kinase (AMPKα) and ERK1/2 signaling to restrain mTOR signaling, allowing for increased sensitivity to cisplatin. Cisplatin is essential therapeutic agent for the treatment of patients with breast cancer gene (BRCA)1/2 or partner and localizer of BRCA2 mutations (107). Therefore, these findings may provide guidance for chemotherapy regimen selection in patients with PC. Furthermore, another study found that METTL14 can promote resistance to gemcitabine in PC cells, which can be reversed with METTL14 knockdown (108). METTL14 expression was observed to be upregulated in gemcitabine-resistant human PC cells, where increased METTL14 expression was also found after gemcitabine treatment. Further studies then revealed that the NF-κB transcription factor p65 can target the promoter region of METTL14 to positively its mRNA and protein expression. This in turn upregulated the expression of cytidine deaminase to inactivate gemcitabine.

Contrary to the aforementioned findings, certain studies have also reported that METTL14 can function as a tumor suppressor in PC. PI3K catalytic subunit (PIK3CB) expression is frequently increased in PTEN-deficient PC cells (109). PIK3CB overexpression was found to reduce the expression of components in the METTL3/METTL14/WTAP complex and activate AKT signaling, which enhanced the proliferative and migratory capacity of PC cells. Cell division cycle 2-like kinase 1 (CLK1) was also found to be upregulated in PC tissues, which enhances the phosphorylation of serine/arginine-rich splicing factor 5 (SRSF5) to suppress METTL14 exon 10 skipping. METTL14 exon 10 skipping enhances the degree of m6A modification on its gene. CLK1 can also enhance PC cell proliferation, migration and invasion by decreasing the m6A level of METTL14, in addition to promoting cyclin L2 exon 6.3 skipping (110). However, the downstream regulatory mechanism of METTL14 remains unclear.

Other cancers

In cholangiocarcinoma (CCA), lower expression levels of stromal interaction molecule 2 (STIM2) is associated with poor prognosis (111). m6A modification mediated by METTL14 and YTHDC2 was found to decrease the expression of STIM2 mRNA (111). Consequently, knockdown of STIM2 can increase keratin 8 (KRT8) expression to facilitate extrahepatic CCA metastasis (111).

5. Research advances in bioinformatics

The Cancer Genome Atlas (TCGA) is a useful tool for determining DNA, RNA and protein expression levels, in addition to exploring gene function, in a variety of cancers (112). Preliminary understanding can first be obtained, which can then be verified using experimental data (112). This section summarizes the results of m6A-related studies obtained by bioinformatics analysis alone.

The majority of the bioinformatics studies performed focused on the expression levels of METTL14 and their prognostic value. Zhou et al (78) found that METTL14 expression in HCC tissues is significantly lower compared with those in adjacent tissues. Furthermore, METTL14 expression in HCC tissues is associated with tumor size and tumor-node-metastasis (TNM) stage in patients with HCC. Lower expression levels of METTL14 in HCC tissues were significantly associated with poorer prognosis, with the overall survival and tumor-free survival rates being significantly shortened. Liu et al (56) also showed that METTL14 expression is lower in HCC tissues and associated with poorer prognosis. By contrast, the opposite trend was observed in terms of the prognostic values of METTL3 in HCC (56). Kong et al (113) found that the expression of METTL14 is upregulated in HCC tissues, but the downregulation of METTL14 is associated with poor prognosis (113), possibly due to lower levels of immune cell infiltration (113). Another study revealed that METTL14 may participate in the malignant progression of HCC by regulating the expression levels of cysteine sulfinic acid decarboxylase, glutamate-oxaloacetate transaminase 2 and suppressors of cytokine signaling 2 (114). The author used LASSO analysis to analyze a total of 124 prognostic genes in 307 TCGA samples to find real hub genes that are associated with patient prognosis, but the specific mechanism needs to be verified by further experiments (114). The results of the aforementioned analyses suggest that METTL14 is upregulated in HCC and inversely associated with clinical prognosis. Xu et al (115) previously suggested that METTL14 expression is upregulated in PC, which displayed a U-shaped association with clinical stage. Specifically, expression in stages I and IV was higher compared with that at stages II and III. Additionally, METTL14 expression is negatively associated with the T phase (115). Similarly, another previous analysis by Zhang et al (116) revealed that METTL14 is highly expressed in PC tissues. Additionally, two studies on rectal cancer found that METTL14 expression is associated with worse prognosis (117,118). In addition, METTL14 expression was positively correlated with the degree of immune cell infiltration, as found by Cai et al (117). In GC, Xu et al (119) found that lower METTL14 expression significantly correlated with the expression levels of PD-L1 and PD-1 (119). Using LASSO regression analysis, a study on esophageal cancer (ESCA) found that METTL14 expression can be used to predict overall survival in patients with ESCA (120).

6. Potential research direction for METTL14 and digestive system cancers

As a methyltransferase, METTL14 can mediate m6A methylation to regulate various physiological processes in malignancies, such as proliferation, invasion, metastasis and drug resistance (10). In addition, METTL14 expression can be used as a marker for the diagnosis and prognosis of digestive system tumors. Using bioinformatics, several studies have analyzed the expression profile of METTL14 in digestive system cancers and developed various prognostic risk models (114,115). Further validation is required to explore the specificity and sensitivity of METTL14 as a marker. METTL14 expression may also confer benefits for predicting efficacy. The role of METTL14 in cisplatin and gemcitabine resistance in PC suggests that it can be used for predicting chemotherapy outcome (108). Wang et al (97) previously found that METTL14 can regulate the response of CRC cells to immunotherapy, suggesting that METTL14 can enhance the effectiveness of immunotherapy, either when combined with other drugs or as a potential independent therapeutic target (97).

METTL14 mediate different regulatory roles in different tumors. In GC and CRC, the majority of the evidence suggest that METTL14 serves as a tumor suppressor, whereas in PC they suggest METTL14 to be an oncogene. In HCC, the role of METTL14 remains controversial. Whether METTL14 itself is an oncogene or tumor suppressor remains a matter of debate. In addition, the mechanism underlying the regulation of METTL14 remain elusive. A study did previously find that the SUMOylation of METTL3 significantly inhibited its m6A methyltransferase activity in NSCLC, resulting in reduced m6A levels. However, this did not affect METTL14 stability, localization or its interaction with WTAP (121).

In terms of the possible upstream mechanisms that can regulate METTL14 expression, binding to its promoters and subsequent inhibition of METT14 transcription or direct regulation of METTL14 mRNA expression has been considered. Upstream regulators include RBPs, transcription factors, exosomes from M1 macrophages and endotoxins produced by the gut flora (Table II). In liver cancer, FA can reduce the m6A of METTL14 in HepG2 cells to subsequently inhibit p53 gene expression (81). The transcription factor HIF-1α can also inhibit METTL14 expression and promote the development of HCC under hypoxic conditions (85). In addition, LPS treatment was found to increase the expression of METTL14 in Huh7 cells whilst increasing the m6A methylation level of MIR155HG (87). Exosomes of M1 macrophages inhibited METTL14 expression by transferring miR-628-5p to HCC cells, which inhibited the m6A modification of circFUT8 (89). In CRC, KDM5C-mediated demethylation of histone H3K4me3 in the promoter region inhibited METTL14 transcription, which may account for the reduction of METTL14 expression found in CRC (94). MeCP2 was also found to inhibit the m6A methylation of METTL14 in CRC cells, mechanistically due to MeCP2 competitively disrupting the interaction between METTL3 and METTL14 (95). In addition, the RBP HuR can inhibit METTL14 mRNA expression by binding to its promoter (96), whereas TCF4 deficiency significantly increased the ubiquitination level of METTL14 and promoted its protein degradation (96). In PC, CLK1 enhanced the phosphorylation of SRSF5 on Ser-250, which inhibited METTL14 exon 10 skipping and increased the level of m6A modification (110). Therefore, the regulatory mechanism of METTL14 function in the individual tumor types must be strictly defined. In addition, the mechanism of action of the METTL3/METTL14 complex remains unclear. In particularly, whether the two function allosterically or synergistically remain unknown. A previous study in HCC reported that the expression and function profiles of the two are distinct and do not share common pathways downstream (56).

Table II

Upstream mechanism of METTL14.

Table II

Upstream mechanism of METTL14.

CancerUpstream regulatorFunctionRegulation mechanism(Refs.)
Liver cancerFusaric acidInhibit the m6A level of METTL14N/A(81)
Hypoxia-inducible factor 1αInhibit the expression of METTL14Under hypoxic conditions(85)
LipopolysacharideEnhance the expression of METTL14N/A(87)
miR-628-5pInhibit the expression of METTL14M1-exo transfer miR-628-5p to HCC cell(89)
Colorectal cancerLysine-specific histone demethylase 5CInhibit the transcription of METTL14Demethylation of histone H3K4me3 in the promoter region(94)
MethylCpG-binding protein 2Inhibit the m6A level of METTL14Affect the interaction between METTL3 and METTL14(95)
Hu antigen receptorInhibit the expression of METTL14Binding to the promoter(96)
Transcription factor 4Enhance the expression of METTL14Inhibits ubiquitination of METTL14(96)
Pancreatic cancerCell division cycle 2-like kinase 1Enhance the m6A level of METTL14Inhibited METTL14exon 10 skipping(110)

[i] METTL, methyltransferase-like; miR, microRNA.

The PI3K/AKT signaling pathway is frequently found to be associated with cancer development, metastasis and EMT. Previous studies in LC, GC and CRC have confirmed that the PI3K/AKT signaling pathway is a downstream regulatory pathway of METTL14 (67,68,82,94). HIF-1α can inhibit the expression of METTL14 in HCC, but HIF-1α can be regulated by METTL14 in GC as one of its downstream targets (71,85). The EGFR signaling pathway was found to serve downstream of METTL14 in LC and CRC (82,100), whereas the ERK signaling pathway was found to serve downstream of METTL14 in HCC and PC (88,107). In conclusion, the mechanism of regulation by METTL14 in digestive system malignancies is one of the most important directions for future research.

Anti-angiogenic therapy is an important modality for digestive system tumor treatment (122). Due to the low mutation rate of anti-angiogenic genes in CRC (122), epigenetic modifications may provide a more appropriate target for blocking tumor angiogenesis. The role of m6A regulation in tumor angiogenesis is currently a topic of intense research. Chen et al (123) previously found that METTL14-mediated m6A methylation can increase TNF receptor-associated factor RNA stability and inhibit sunitinib-mediated anti-angiogenic effects in renal cancer cells (123). In addition, Wen et al (124) revealed that basic leucine zipper activating transcription factors-like transcription factor 2 (BATF2) expression is downregulated in tongue squamous cell carcinoma (TSCC). Downregulated BATF2 serves a role in promoting tumor growth, metastasis and angiogenesis (124). METTL14-mediated m6A modification was found to inhibit BATF2 mRNA expression and increase the expression of vascular endothelial growth factor A (124). The role of METTL14 in angiogenesis in digestive system tumors remains uncharacterized, exploring it would be of great clinical significance for patients with digestive system tumors in addition to providing a potential research direction.

In conclusion, the expression and role of METTL14 in different digestive system cancers remain controversial. In particular, the regulatory mechanism of METTL14 require further study. Although METTL14 has been demonstrated to serve an important role in digestive system tumors, its implications for clinical application also requires further investigation.

7. Conclusion

In summary, METTL14 serves diverse roles in digestive system cancers and has diagnostic and therapeutic potential. Further exploration of the regulatory mechanisms of METTL14 is anticipated, which is expected to accelerate their incorporation into clinical treatment.

Availability of data and materials

Not applicable.

Authors' contributions

JXH and HSL contributed to conception and design. JXH and CW contributed to literature search. JXH contributed to manuscript writing. QS and BWC approved the final version of the manuscript, and agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

Supported by Beijing Natural Science Foundation Program (grant no. 7222032), the National Natural Science Foundation of China (grant no. 82173056) and the Beijing Natural Science Foundation (grant no. 7214220 to HL), the Friendship Seed Project (grant no. YYZZ202034 to HL).

References

1 

Li S, Kuo HC, Yin R, Wu R, Liu X, Wang L, Hudlikar R, Peter RM and Kong AN: Epigenetics/epigenomics of triterpenoids in cancer prevention and in health. Biochem Pharmacol. 175:1138902020. View Article : Google Scholar : PubMed/NCBI

2 

Goel A and Boland CR: Epigenetics of colorectal cancer. Gastroenterology. 143:1442–1460.e1. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Zhou S, Treloar AE and Lupien M: Emergence of the noncoding cancer genome: A target of genetic and epigenetic alterationsthe noncoding cancer genome. Cancer Discov. 6:1215–1229. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Zhao Y, Shi Y, Shen H and Xie W: m6A-binding proteins: The emerging crucial performers in epigenetics. J Hematol Oncol. 13:352020. View Article : Google Scholar

5 

Ghavami S, Zamani M, Ahmadi M, Erfani M, Dastghaib S, Darbandi M, Darbandi S, Vakili O, Siri M, Grabarek BO, et al: Epigenetic regulation of autophagy in gastrointestinal cancers. Biochim Biophys Acta Mol Basis Dis. 1868:1665122022. View Article : Google Scholar : PubMed/NCBI

6 

Jiang X, Liu B, Nie Z, Duan L, Xiong Q, Jin Z, Yang C and Chen Y: The role of m6A modification in the biological functions and diseases. Signal Transduct Target Ther. 6:742021. View Article : Google Scholar : PubMed/NCBI

7 

Jones PA, Issa JP and Baylin S: Targeting the cancer epigenome for therapy. Nat Rev Genet. 17:630–641. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Bird A: Perceptions of epigenetics. Nature. 447:396–398. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Liu X, Wang P, Teng X, Zhang Z and Song S: Comprehensive analysis of expression regulation for RNA m6A regulators with clinical significance in human cancers. Front Oncol. 11:6243952021. View Article : Google Scholar : PubMed/NCBI

10 

Song N, Cui K, Zhang K, Yang J, Liu J, Miao Z, Zhao F, Meng H, Chen L, Chen C, et al: The role of m6A RNA methylation in cancer: Implication for nature products anti-cancer research. Front Pharmacol. 13:9333322022. View Article : Google Scholar : PubMed/NCBI

11 

Chen DH, Zhang JG, Wu CX and Li Q: Non-Coding RNA m6A modification in cancer: Mechanisms and therapeutic targets. Front Cell Dev Biol. 9:7785822021. View Article : Google Scholar

12 

Roundtree IA, Evans ME, Pan T and He C: Dynamic RNA modifications in gene expression regulation. Cell. 169:1187–1200. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang YG and He C: N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 7:885–887. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yi YC, Chen XY, Zhang J and Zhu JS: Novel insights into the interplay between m6A modification and noncoding RNAs in cancer. Mol Cancer. 19:1212020. View Article : Google Scholar

15 

Li H, Wu H, Wang Q, Ning S, Xu S and Pang D: Dual effects of N6-methyladenosine on cancer progression and immunotherapy. Mol Ther Nucleic Acids. 24:25–39. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Cheng Y, Xie W, Pickering BF, Chu KL, Savino AM, Yang X, Luo H, Nguyen DT, Mo S, Barin E, et al: N6-Methyladenosine on mRNA facilitates a phase-separated nuclear body that suppresses myeloid leukemic differentiation. Cancer Cell. 39:958–972.e8. 2021. View Article : Google Scholar

17 

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al: Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 485:201–206. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE and Jaffrey SR: Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell. 149:1635–1646. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Chen Y, Peng C, Chen J, Chen D, Yang B, He B, Hu W, Zhang Y, Liu H, Dai L, et al: WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer. 18:1272019. View Article : Google Scholar : PubMed/NCBI

20 

Chen Y, Zhao Y, Chen J, Peng C, Zhang Y, Tong R, Cheng Q, Yang B, Feng X, Lu Y, et al: ALKBH5 suppresses malignancy of hepatocellular carcinoma via m6A-guided epigenetic inhibition of LYPD1. Mol Cancer. 19:1232020. View Article : Google Scholar :

21 

Fu Y, Dominissini D, Rechavi G and He C: Gene expression regulation mediated through reversible m6A RNA methylation. Nat Rev Genet. 15:293–306. 2014. View Article : Google Scholar : PubMed/NCBI

22 

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. 29:2037–2053. 2015. View Article : Google Scholar : PubMed/NCBI

23 

He PC and He C: m6A RNA methylation: From mechanisms to therapeutic potential. EMBO J. 40:e1059772021. View Article : Google Scholar

24 

Yue Y, Liu J, Cui X, Cao J, Luo G, Zhang Z, Cheng T, Gao M, Shu X, Ma H, et al: VIRMA mediates preferential m6A mRNA methylation in 3' UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 4:102018. View Article : Google Scholar

25 

Wei CM and Moss B: Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. Biochemistry. 16:1672–1676. 1977. View Article : Google Scholar : PubMed/NCBI

26 

Chen XY, Zhang J and Zhu JS: The role of m6A RNA methylation in human cancer. Mol Cancer. 18:1032019. View Article : Google Scholar

27 

Wang N, Huo X, Zhang B, Chen X, Zhao S, Shi X, Xu H and Wei X: METTL3-Mediated ADAMTS9 suppression facilitates angiogenesis and carcinogenesis in gastric cancer. Front Oncol. 12:8618072022. View Article : Google Scholar : PubMed/NCBI

28 

Yu T, Liu J, Wang Y, Chen W, Liu Z, Zhu L and Zhu W: METTL3 promotes colorectal cancer metastasis by stabilizing PLAU mRNA in an m6A-dependent manner. Biochem Biophys Res Commun. 614:9–16. 2022. View Article : Google Scholar : PubMed/NCBI

29 

Wang Q, Chen C, Ding Q, Zhao Y, Wang Z, Chen J, Jiang Z, Zhang Y, Xu G, Zhang J, et al: METTL3-mediated m6A modification of HDGF mRNA promotes gastric cancer progression and has prognostic significance. Gut. 69:1193–1205. 2020. View Article : Google Scholar

30 

Zhou H, Yin K, Zhang Y, Tian J and Wang S: The RNA m6A writer METTL14 in cancers: Roles, structures, and applications. Biochim Biophys Acta Rev Cancer. 1876:1886092021. View Article : Google Scholar : PubMed/NCBI

31 

Sun T, Wu Z, Wang X, Wang Y, Hu X, Qin W, Lu S, Xu D, Wu Y, Chen Q, et al: LNC942 promoting METTL14-mediated m6A methylation in breast cancer cell proliferation and progression. Oncogene. 39:5358–5372. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Ruszkowska A: METTL16, methyltransferase-like protein 16: Current insights into structure and function. Int J Mol Sci. 22:21762021. View Article : Google Scholar : PubMed/NCBI

33 

Su R, Dong L, Li Y, Gao M, He PC, Liu W, Wei J, Zhao Z, Gao L, Han L, et al: METTL16 exerts an m6A-independent function to facilitate translation and tumorigenesis. Nat Cell Biol. 24:205–216. 2022. View Article : Google Scholar : PubMed/NCBI

34 

Trindade F, Tellechea Ó, Torrelo A, Requena L and Colmenero I: Wilms tumor 1 expression in vascular neoplasms and vascular malformations. Am J Dermatopathol. 33:569–572. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Wang LJ, Xue Y, Li H, Huo R, Yan Z, Wang J, Xu H, Wang J, Cao Y and Zhao JZ: Wilms' tumour 1-associating protein inhibits endothelial cell angiogenesis by m6A-dependent epigenetic silencing of desmoplakin in brain arteriovenous malformation. J Cell Mol Med. 24:4981–4991. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Zhu W, Wang JZ, Wei JF and Lu C: Role of m6A methyltransferase component VIRMA in multiple human cancers. Cancer Cell Int. 21:1722021. View Article : Google Scholar

37 

Panneerdoss S, Eedunuri VK, Yadav P, Timilsina S, Rajamanickam S, Viswanadhapalli S, Abdelfattah N, Onyeagucha BC, Cui X, Lai Z, et al: Cross-talk among writers, readers, and erasers of m6A regulates cancer growth and progression. Sci Adv. 4:eaar82632018. View Article : Google Scholar

38 

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. 561:556–560. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Shan K, Zhou RM, Xiang J, Sun YN, Liu C, Lv MW and Xu JJ: FTO regulates ocular angiogenesis via m6A-YTHDF2-dependent mechanism. Exp Eye Res. 197:1081072020. View Article : Google Scholar

40 

Mathiyalagan P, Adamiak M, Mayourian J, Sassi Y, Liang Y, Agarwal N, Jha D, Zhang S, Kohlbrenner E, Chepurko E, et al: FTO-dependent N6-methyladenosine regulates cardiac function during remodeling and repair. Circulation. 139:518–532. 2019. View Article : Google Scholar :

41 

Qu J, Yan H, Hou Y, Cao W, Liu Y, Zhang E, He J and Cai Z: RNA demethylase ALKBH5 in cancer: From mechanisms to therapeutic potential. J Hematol Oncol. 15:82022. View Article : Google Scholar : PubMed/NCBI

42 

Zhao Y, Hu J, Sun X, Yang K, Yang L, Kong L, Zhang B, Li F, Li C, Shi B, et al: Loss of m6A Demethylase ALKBH5 Promotes post-ischemic Angiogenesis via post-transcriptional Stabilization of WNT5A. Clin Transl Med. 11:e4022021. View Article : Google Scholar : PubMed/NCBI

43 

Pendleton KE, Chen B, Liu K, Hunter OV, Xie Y, Tu BP and Conrad NK: The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell. 169:824–835.e14. 2017. View Article : Google Scholar :

44 

Yan H, Zhang L, Cui X, Zheng S and Li R: Roles and mechanisms of the m6A reader YTHDC1 in biological processes and diseases. Cell Death Discov. 8:2372022. View Article : Google Scholar

45 

Dai XY, Shi L, Li Z, Yang HY, Wei JF and Ding Q: Main N6-methyladenosine readers: YTH family proteins in cancers. Front Oncol. 11:6353292021. View Article : Google Scholar : PubMed/NCBI

46 

Gao LB, Zhu XL, Shi JX, Yang L, Xu ZQ and Shi SL: HnRNPA2B1 promotes the proliferation of breast cancer MCF-7 cells via the STAT3 pathway. J Cell Biochem. 122:472–484. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Li J, Xie H, Ying Y, Chen H, Yan H, He L, Xu M, Xu X, Liang Z Liu B, et al: YTHDF2 mediates the mRNA degradation of the tumor suppressors to induce AKT phosphorylation in N6-methyladenosine-dependent way in prostate cancer. Mol Cancer. 19:1522020. View Article : Google Scholar : PubMed/NCBI

48 

Schöller E, Weichmann F, Treiber T, Ringle S, Treiber N, Flatley A, Feederle R, Bruckmann A and Meister G: Interactions, localization, and phosphorylation of the m6A generating METTL3-METTL14-WTAP complex. RNA. 24:499–512. 2018. View Article : Google Scholar

49 

Śledź P and Jinek M: Structural insights into the molecular mechanism of the m(6)A writer complex. Elife. 5:e184342016. View Article : Google Scholar : PubMed/NCBI

50 

Wang P, Doxtader KA and Nam Y: Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell. 63:306–317. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Lin S, Choe J, Du P, Triboulet R and Gregory RI: The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol Cell. 62:335–345. 2016. View Article : Google Scholar : PubMed/NCBI

52 

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. 10:93–95. 2014. View Article : Google Scholar

53 

Bujnicki JM, Feder M, Radlinska M and Blumenthal RM: Structure prediction and phylogenetic analysis of a functionally diverse family of proteins homologous to the MT-A70 subunit of the human mRNA: m(6)A methyltransferase. J Mol Evol. 55:431–444. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Han SH and Choe J: Diverse molecular functions of m6A mRNA modification in cancer. Exp Mol Med. 52:738–749. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Wen J, Lv R, Ma H, Shen H, He C, Wang J, Jiao F, Liu H, Yang P, Tan L, et al: Zc3h13 regulates nuclear RNA m6A methylation and mouse embryonic stem cell self-renewal. Mol Cell. 69:1028–1038.e6. 2018. View Article : Google Scholar

56 

Liu X, Qin J, Gao T, Li C, Chen X, Zeng K, Zeng K, Xu M, He B, Pan B, et al: Analysis of METTL3 and METTL14 in hepatocellular carcinoma. Aging (albany NY). 12:21638–21659. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Lin Z, Hsu PJ, Xing X, Fang J, Lu Z, Zou Q, Zhang KJ, Zhang X, Zhou Y, Zhang T, et al: Mettl3-/Mettl14-mediated mRNA N6-methyladenosine modulates murine spermatogenesis. Cell Res. 27:1216–1230. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, Fu Y, Parisien M, Dai Q, Jia G, et al: N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 505:117–120. 2014. View Article : Google Scholar

59 

Lin R, Zhan M, Yang L, Wang H, Shen H, Huang S, Huang X, Xu S, Zhang Z, Li W, et al: Deoxycholic acid modulates the progression of gallbladder cancer through N6-methyladenosine-dependent microRNA maturation. Oncogene. 39:4983–5000. 2020. View Article : Google Scholar : PubMed/NCBI

60 

Yang Y, Qian Cai Q, Sheng Fu L, Wei Dong Y, Fan F and Zhong Wu X: Reduced N6-Methyladenosine Mediated by METTL3 Acetylation Promotes MTF1 expression and hepatocellular carcinoma cell growth. Chem Biodivers. 19:e2022003332022. View Article : Google Scholar : PubMed/NCBI

61 

Zhang N, Zuo Y, Peng Y and Zuo L: Function of N6-methyladenosine modification in tumors. J Oncol. 2021:64615522021. View Article : Google Scholar : PubMed/NCBI

62 

Lan Q, Liu PY, Haase J, Bell JL, Hüttelmaier S and Liu T: The critical role of RNA m6A methylation in cancer. Cancer Res. 79:1285–1292. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Deng X, Su R, Weng H, Huang H, Li Z and Chen J: RNA N6-methyladenosine modification in cancers: Current status and perspectives. Cell Res. 28:507–517. 2018. View Article : Google Scholar : PubMed/NCBI

64 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Hu BE, Wang XY, Gu XY, Zou C, Gao ZJ, Zhang H and Fan Y: N6-methyladenosine (m6A) RNA modification in gastrointestinal tract cancers: Roles, mechanisms, and applications. Mol Cancer. 18:1782019. View Article : Google Scholar

67 

Zhang C, Zhang M, Ge S, Huang W, Lin X, Gao J, Gong J and Shen L: Reduced m6A modification predicts malignant phenotypes and augmented Wnt/PI3K-Akt signaling in gastric cancer. Cancer Med. 8:4766–4781. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Liu X, Xiao M, Zhang L, Li L, Zhu G, Shen E, Lv M, Lu X and Sun Z: The m6A methyltransferase METTL14 inhibits the proliferation, migration, and invasion of gastric cancer by regulating the PI3K/AKT/mTOR signaling pathway. J Clin Lab Anal. 35:e236552021.

69 

Yao Q, He L, Gao X, Tang N, Lin L, Yu X and Wang D: The m6A methyltransferase METTL14-mediated N6-methyladenosine modification of PTEN mRNA inhibits tumor growth and metastasis in stomach adenocarcinoma. Front Oncol. 11:6997492021. View Article : Google Scholar : PubMed/NCBI

70 

Fan HN, Chen ZY, Chen XY, Chen M, Yi YC, Zhu JS and Zhang J: METTL14-mediated m6A modification of circORC5 suppresses gastric cancer progression by regulating miR-30c-2-3p/AKT1S1 axis. Mol Cancer. 21:512022. View Article : Google Scholar :

71 

Lin JX, Lian NZ, Gao YX, Zheng QL, Yang YH, Ma YB, Xiu ZS, Qiu QZ, Wang HG, Zheng CH, et al: m6A methylation mediates LHPP acetylation as a tumour aerobic glycolysis suppressor to improve the prognosis of gastric cancer. Cell Death Dis. 13:4632022. View Article : Google Scholar : PubMed/NCBI

72 

Jin H, Wu Z, Tan B, Liu Z, Zu Z, Wu X, Bi Y and Hu X: Ibuprofen promotes p75 neurotrophin receptor expression through modifying promoter methylation and N6-methyladenosine-RNA-methylation in human gastric cancer cells. Bioengineered. 13:14595–14604. 2022. View Article : Google Scholar : PubMed/NCBI

73 

Hu N and Ji H: N6-methyladenosine (m6A)-mediated up-regulation of long noncoding RNA LINC01320 promotes the proliferation, migration, and invasion of gastric cancer via miR495-5p/RAB19 axis. Bioengineered. 12:4081–4091. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Balogh J, Victor D III, Asham EH, Burroughs SG, Boktour M, Saharia A, Li X, Ghobrial RM and Monsour HP Jr: Hepatocellular carcinoma: A review. J Hepatocell Carcinoma. 3:41–53. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Raoul JL and Edeline J: Systemic treatment of hepatocellular carcinoma: Standard of care in China and elsewhere. Lancet Oncol. 21:479–481. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Li ZL, Yan WT, Zhang J, Zhao YJ, Lau WY, Mao XH, Zeng YY, Zhou YH, Gu WM, Wang H, et al: Identification of actual 10-year survival after hepatectomy of HBV-related hepatocellular carcinoma: A multicenter study. J Gastrointest Surg. 23:288–296. 2019. View Article : Google Scholar

77 

Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 144:1941–1953. 2019. View Article : Google Scholar

78 

Zhou T, Ren Z and Chen C: METTL14 as a predictor of postoperative survival outcomes of patients with hepatocellular carcinoma. Nan Fang Yi Ke Da Xue Xue Bao. 40:567–572. 2020.In Chinese. PubMed/NCBI

79 

Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, Wang TT, Xu QG, Zhou WP and Sun SH: METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6-methyladenosine-dependent primary MicroRNA processing. Hepatology. 65:529–543. 2017. View Article : Google Scholar

80 

Laptenko O and Prives C: Transcriptional regulation by p53: One protein, many possibilities. Cell Death Differ. 13:951–961. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Ghazi T, Nagiah S and Chuturgoon AA: Fusaric acid decreases p53 expression by altering promoter methylation and m6A RNA methylation in human hepatocellular carcinoma (HepG2) cells. Epigenetics. 16:79–91. 2021. View Article : Google Scholar :

82 

Shi Y, Zhuang Y, Zhang J, Chen M and Wu S: METTL14 inhibits hepatocellular carcinoma metastasis through regulating EGFR/PI3K/AKT signaling pathway in an m6A-dependent manner. Cancer Manag Res. 12:13173–13184. 2020. View Article : Google Scholar :

83 

Zhou T, Li S, Xiang D, Liu J, Sun W, Cui X, Ning B, Li X, Cheng Z, Jiang W, et al: m6A RNA methylation-mediated HNF3γ reduction renders hepatocellular carcinoma dedifferentiation and sorafenib resistance. Signal Transduct Target Ther. 5:2962020. View Article : Google Scholar

84 

Du L, Li Y, Kang M, Feng M, Ren Y, Dai H, Wang Y, Wang Y and Tang B: USP48 is upregulated by Mettl14 to attenuate hepatocellular carcinoma via regulating SIRT6 stabilization. Cancer Res. 81:3822–3834. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Fan Z, Yang G, Zhang W, Liu Q, Liu G, Liu P, Xu L, Wang J, Yan Z, Han H, et al: Hypoxia blocks ferroptosis of hepatocellular carcinoma via suppression of METTL14 triggered YTHDF2-dependent silencing of SLC7A11. J Cell Mol Med. 25:10197–10212. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Yang Y, Cai J, Yang X, Wang K, Sun K, Yang Z, Zhang L, Yang L, Gu C, Huang X, et al: Dysregulated m6A modification promotes lipogenesis and development of non-alcoholic fatty liver disease and hepatocellular carcinoma. Mol Ther. 30:2342–2353. 2022. View Article : Google Scholar : PubMed/NCBI

87 

Peng L, Pan B, Zhang X, Wang Z, Qiu J, Wang X and Tang N: Lipopolysaccharide facilitates immune escape of hepatocellular carcinoma cells via m6A modification of lncRNA MIR155HG to upregulate PD-L1 expression. Cell Biol Toxicol. 38:1159–1173. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Liu J, Zhang N, Zeng J, Wang T, Shen Y, Ma C and Yang M: N6-methyladenosine-modified lncRNA ARHGAP5-AS1 stabilises CSDE1 and coordinates oncogenic RNA regulons in hepatocellular carcinoma. Clin Transl Med. 12:e11072022. View Article : Google Scholar

89 

Wang L, Yi X, Xiao X, Zheng Q, Ma L and Li B: Exosomal miR-628-5p from M1 polarized macrophages hinders m6A modification of circFUT8 to suppress hepatocellular carcinoma progression. Cell Mol Biol Lett. 27:1062022. View Article : Google Scholar : PubMed/NCBI

90 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Teng S, Li YE, Yang M, Qi R, Huang Y, Wang Q, Zhang Y, Chen S, Li S, Lin K, et al: Tissue-specific transcription reprogramming promotes liver metastasis of colorectal cancer. Cell Res. 30:34–49. 2020. View Article : Google Scholar :

92 

Tian J, Ying P, Ke J, Zhu Y, Yang Y, Gong Y, Zou D, Peng X, Yang N, Wang X, et al: ANKLE1 N6-Methyladenosine-related variant is associated with colorectal cancer risk by maintaining the genomic stability. Int J Cancer. 146:3281–3293. 2020. View Article : Google Scholar

93 

Yang X, Zhang S, He C, Xue P, Zhang L, He Z, Zang L, Feng B, Sun J and Zheng M: METTL14 suppresses proliferation and metastasis of colorectal cancer by down-regulating oncogenic long non-coding RNA XIST. Mol Cancer. 19:462020. View Article : Google Scholar : PubMed/NCBI

94 

Chen X, Xu M, Xu X, Zeng K, Liu X, Pan B, Li C, Sun L, Qin J, Xu T, et al: METTL14-mediated N6-methyladenosine modification of SOX4 mRNA inhibits tumor metastasis in colorectal cancer. Mol Cancer. 19:1062020. View Article : Google Scholar : PubMed/NCBI

95 

Wang S, Gan M, Chen C, Zhang Y, Kong J, Zhang H and Lai M: Methyl CpG binding protein 2 promotes colorectal cancer metastasis by regulating N6-methyladenosine methylation through methyltransferase-like 14. Cancer Sci. 112:3243–3254. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Wang H, Wei W, Zhang ZY, Liu Y, Shi B, Zhong W, Zhang HS, Fang X, Sun CL, Wang JB and Liu LX: TCF4 and HuR mediated-METTL14 suppresses dissemination of colorectal cancer via N6-methyladenosine-dependent silencing of ARRDC4. Cell Death Dis. 13:32021. View Article : Google Scholar : PubMed/NCBI

97 

Wang L, Hui H, Agrawal K, Kang Y, Li N, Tang R, Yuan J and Rana TM: m6A RNA methyltransferases METTL3/14 regulate immune responses to anti-PD-1 therapy. EMBO J. 39:e1045142020. View Article : Google Scholar

98 

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 N6-adenosine methyltransferase Mettl14 in tumor-associated macrophages promotes CD8+ T cell dysfunction and tumor growth. Cancer Cell. 39:945–957.e10. 2021. View Article : Google Scholar

99 

Cao Y, Wang Z, Yan Y, Ji L, He J, Xuan B, Shen C, Ma Y, Jiang S, Ma D, et al: Enterotoxigenic Bacteroides fragilis promotes intestinal inflammation and malignancy by inhibiting exosome-packaged miR-149-3p. Gastroenterology. 161:1552–1566.e12. 2021. View Article : Google Scholar

100 

Luo M, Huang Z, Yang X, Chen Y, Jiang J, Zhang L, Zhou L, Qin S, Jin P, Fu S, et al: PHLDB2 mediates cetuximab resistance via interacting with EGFR in latent metastasis of colorectal cancer. Cell Mol Gastroenterol Hepatol. 13:1223–1242. 2022. View Article : Google Scholar :

101 

Kamisawa T, Wood LD, Itoi T and Takaori K: Pancreatic cancer. Lancet. 388:73–85. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Wolfgang CL, Herman JM, Laheru DA, Klein AP, Erdek MA, Fishman EK and Hruban RH: Recent progress in pancreatic cancer. CA Cancer J Clin. 63:318–348. 2013. View Article : Google Scholar : PubMed/NCBI

103 

Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, Real FX, Van Laethem JL and Heinemann V: Addressing the challenges of pancreatic cancer: Future directions for improving outcomes. Pancreatology. 15:8–18. 2015. View Article : Google Scholar

104 

Wang M, Liu J, Zhao Y, He R, Xu X, Guo X, Li X, Xu S, Miao J, Guo J, et al: Upregulation of METTL14 mediates the elevation of PERP mRNA N6 adenosine methylation promoting the growth and metastasis of pancreatic cancer. Mol Cancer. 19:1302020. View Article : Google Scholar

105 

Jiang Z, Song X, Wei Y, Li Y, Kong D and Sun J: N(6)-methyladenosine-mediated miR-380-3p maturation and upregulation promotes cancer aggressiveness in pancreatic cancer. Bioengineered. 13:14460–14471. 2022. View Article : Google Scholar : PubMed/NCBI

106 

Kong F, Liu X, Zhou Y, Hou X, He J, Li Q, Miao X and Yang L: Downregulation of METTL14 increases apoptosis and autophagy induced by cisplatin in pancreatic cancer cells. Int J Biochem Cell Biol. 122:1057312020. View Article : Google Scholar : PubMed/NCBI

107 

Nabors LB, Portnow J, Ahluwalia M, Baehring J, Brem H, Brem S, Butowski N, Campian JL, Clark SW, Fabiano AJ, et al: Central nervous system cancers, version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 18:1537–1570. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Zhang C, Ou S, Zhou Y, Liu P, Zhang P, Li Z, Xu R and Li Y: m6A Methyltransferase METTL14-Mediated upregulation of cytidine deaminase promoting gemcitabine resistance in pancreatic cancer. Front Oncol. 11:6963712021. View Article : Google Scholar

109 

Tian J, Zhu Y, Rao M, Cai Y, Lu Z, Zou D, Peng X, Ying P, Zhang M, Niu S, et al: N6-methyladenosine mRNA methylation of PIK3CB regulates AKT signalling to promote PTEN-deficient pancreatic cancer progression. Gut. 69:2180–2192. 2020. View Article : Google Scholar : PubMed/NCBI

110 

Chen S, Yang C, Wang ZW, Hu JF, Pan JJ, Liao CY, Zhang JQ, Chen JZ, Huang Y, Huang L, et al: CLK1/SRSF5 pathway induces aberrant exon skipping of METTL14 and Cyclin L2 and promotes growth and metastasis of pancreatic cancer. J Hematol Oncol. 14:602021. View Article : Google Scholar : PubMed/NCBI

111 

Chen FQ, Zheng H, Gu T, Hu YH, Yang L, Huang ZP, Qiao GL and Li HJ: Modification of STIM2 by m6A RNA methylation inhibits metastasis of cholangiocarcinoma. Ann Transl Med. 10:402022. View Article : Google Scholar

112 

Tomczak K, Czerwińska P and Wiznerowicz M: The Cancer Genome Atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol (Pozn). 19(1A): A68–A77. 2015.PubMed/NCBI

113 

Kong F, Wang K and Wang L: Systematic analysis of the expression profile and prognostic significance of m6A regulators and PD-L1 in hepatocellular carcinoma. Discov Oncol. 13:1312022. View Article : Google Scholar : PubMed/NCBI

114 

Li Z, Li F, Peng Y, Fang J and Zhou J: Identification of three m6A-related mRNAs signature and risk score for the prognostication of hepatocellular carcinoma. Cancer Med. 9:1877–1889. 2020. View Article : Google Scholar : PubMed/NCBI

115 

Xu F, Zhang Z, Yuan M, Zhao Y, Zhou Y, Pei H and Bai L: M6A regulatory genes play an important role in the prognosis, progression and immune microenvironment of pancreatic adenocarcinoma. Cancer Invest. 39:39–54. 2021. View Article : Google Scholar

116 

Zhang T, Sheng P and Jiang Y: m6A regulators are differently expressed and correlated with immune response of pancreatic adenocarcinoma. J Cancer Res Clin Oncol. 149:2805–2822. 2023. View Article : Google Scholar

117 

Cai C, Long J, Huang Q, Han Y, Peng Y, Guo C, Liu S, Chen Y, Shen E, Long K, et al: M6A 'Writer' gene METTL14: A favorable prognostic biomarker and correlated with immune infiltrates in rectal cancer. Front Oncol. 11:6152962021. View Article : Google Scholar

118 

Chen Y, Wang S, Cho WC, Zhou X and Zhang Z: Prognostic Implication of the m6A RNA Methylation Regulators in Rectal Cancer. Front Genet. 12:6042292021. View Article : Google Scholar

119 

Xu Z, Chen Q, Shu L, Zhang C, Liu W and Wang P: Expression profiles of m6A RNA methylation regulators, PD-L1 and immune infiltrates in gastric cancer. Front Oncol. 12:9703672022. View Article : Google Scholar : PubMed/NCBI

120 

Wang H, Zhang Y, Chen L, Liu Y, Xu C, Jiang D, Song Q, Wang H, Wang L, Lin Y, et al: Identification of clinical prognostic features of esophageal cancer based on m6A regulators. Front Immunol. 13:9503652022. View Article : Google Scholar : PubMed/NCBI

121 

Du Y, Hou G, Zhang H, Dou J, He J, Guo Y, Li L, Chen R, Wang Y, Deng R, et al: SUMOylation of the m6A-RNA methyltransferase METTL3 modulates its function. Nucleic Acids Res. 46:5195–5208. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Chen HM, Li H, Lin MX, Fan WJ, Zhang Y, Lin YT and Wu SX: Research progress for RNA modifications in physiological and pathological angiogenesis. Front Genet. 13:9526672022. View Article : Google Scholar : PubMed/NCBI

123 

Chen Y, Lu Z, Qi C, Yu C, Li Y, Huan W, Wang R, Luo W, Shen D, Ding L, et al: N6-methyladenosine-modified TRAF1 promotes sunitinib resistance by regulating apoptosis and angiogenesis in a METTL14-dependent manner in renal cell carcinoma. Mol Cancer. 21:1112022. View Article : Google Scholar

124 

Wen H, Tang J, Cui Y, Hou M and Zhou J: m6A modification-mediated BATF2 suppresses metastasis and angiogenesis of tongue squamous cell carcinoma through inhibiting VEGFA. Cell Cycle. 22:100–116. 2023. View Article : Google Scholar

Related Articles

Journal Cover

September-2023
Volume 52 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu J, Lin H, Wang C, Su Q and Cao B: METTL14‑mediated RNA methylation in digestive system tumors. Int J Mol Med 52: 86, 2023
APA
Hu, J., Lin, H., Wang, C., Su, Q., & Cao, B. (2023). METTL14‑mediated RNA methylation in digestive system tumors. International Journal of Molecular Medicine, 52, 86. https://doi.org/10.3892/ijmm.2023.5289
MLA
Hu, J., Lin, H., Wang, C., Su, Q., Cao, B."METTL14‑mediated RNA methylation in digestive system tumors". International Journal of Molecular Medicine 52.3 (2023): 86.
Chicago
Hu, J., Lin, H., Wang, C., Su, Q., Cao, B."METTL14‑mediated RNA methylation in digestive system tumors". International Journal of Molecular Medicine 52, no. 3 (2023): 86. https://doi.org/10.3892/ijmm.2023.5289