Open Access

Role of N6‑methyladenosine in the pathogenesis, diagnosis and treatment of pancreatic cancer (Review)

  • Authors:
    • Tong Ye
    • Jiaxin Wang
    • Haiying Zhao
    • Guiping Zhao
    • Peng Li
  • View Affiliations

  • Published online on: November 10, 2022     https://doi.org/10.3892/ijo.2022.5452
  • Article Number: 4
  • Copyright: © Ye 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

Pancreatic cancer (PC) ranks as the seventh leading cause of cancer‑associated mortality, and is predicted to become the third leading cause of cancer‑associated mortality by the year 2025. Although advanced modalities of diagnosis and treatment have been continuously emerging, the mortality rate (466,003) approximated to that of the morbidity rate (495,773) in 2020. N6‑methyladenosine (m6A) has been shown to be methylated on the sixth N atom of adenine in RNA, which occurs co‑transcriptionally and serves to regulate gene expression post‑transcriptionally. The discovery of m6A has heralded a new era in the scientific investigation of PC. In the present review article, the classical conception of m6A and emerging hypotheses regarding its role are summarized, and the function of m6A in carcinogenesis and progression of PC is then discussed, followed by the potential roles of m6A in the diagnosis of PC and in therapeutic applications. However, this new era is only at the initial stages, and the extent to which m6A influences PC is still poorly understood. In view of this, the present review article also summarizes the developments at the frontier of the interaction between m6A and PC, and discusses strategies through which m6A may provide a promising avenue for anticancer therapy.

1. Introduction

Pancreatic cancer (PC) is the most common type of malignancy affecting the digestive system, which has the characteristics of 'three low and three high'; that is, a low early diagnosis rate, a low resection rate, a low 5-year survival rate, and a high incidence, a high metastasis rate and a high mortality rate. Owing to the deficiency of specific symptoms during the early stages of the malignancy, the early diagnosis rate of PC is <5%. At the first visit, only 15-20% of patients can undergo radical resection, 30-40% of patients present with locally advanced cancer, whereas 40-50% of patients have distant metastasis. The 5-year survival rate has been found to be 7.2-9.0% (1,2). The early detection, early diagnosis and early treatment of PC has always been a pivotal and difficult challenge for PC. Hence, there is an urgent need to elucidate the underlying mechanisms of its pathogenesis, and to develop novel techniques with a greater diagnostic efficacy.

A multitude of previously published articles in the literature have demonstrated that N6-methyladenosine (m6A) not only participates in physiological processes, including neurogenesis, hematopoiesis, spermatogenesis, sex determination, response to DNA damage or heat shock, circadian rhythm, and the innate and adaptive immune response (3-7), but that it is also tightly associated with the initiation, progression (8,9), diagnosis and treatment of neoplasms (10). Accordingly, the initial part of the present review article will introduce the biological functions of m6A; subsequently, the present review will interpret the attributes of m6A in the carcinogenesis and development of PC, and finally, the prospective clinical applications of m6A in PC will be discussed.

2. Biology of m6A

Thus far, diverse chemical modifications have been detected, and m6A remains the most abundant post-transcriptional modification to be found in messenger RNAs (mRNAs) (3). The modification of m6A comprises methylation at the sixth N atom of adenine in RNA, and approximately one third of mammalian mRNAs, with an average of 3-5 m6A sites per mRNA, have been shown to feature this modification (11,12). m6A was first discovered in 1974 (13-15). The concept of reversible m6A modification was proposed in a stepwise manner owing to the discovery of the first methyltransferase, namely methyltransferase-like protein 3 (METTL3) in 1997 (16), and the first demethylase, fat mass and obesity-associated protein (FTO), in 2011 (17).

In order to more clearly elucidate the functions of m6A, scientists have been seeking improvements in the available m6A-mapping methods. Meyer et al (18) combined m6A-specific methylated RNA immunoprecipitation with next-generation sequencing (MeRIP-Seq) to depict the landscape of m6A at the whole transcriptome level in 2012, which heralded the creation of the 'epitranscriptome' (18). Since then, m6A has attracted extensive attention in the research community. MeRIP-Seq is able to discern the distribution of m6A at the single transcript level, and identify which groups of transcripts are modified by m6A. However, it has not been successful at counting the precise stoichiometry of m6A sites; neither has it been able to distinguish m6A from 6,2,-O-dimethyladenosine (m6Am). The molecular structure of m6Am is similar to that of m6A: Compared with m6A, the 2′-hydroxy group of adenylate is methylated to produce this doubly methylated nucleotide.

M6A is conserved in eukaryotes (19-23), and m6A sites are preferentially enriched near stop codons, at 3′-untranslated regions (3′-UTRs) or in long exons, and they are prone to recognize a typical consensus sequence, RRACH (where 'R' is A/G, and 'H' is A/C/U) (4,16,18,21,24). Notably, m6A is specifically located in genes associated with development or cell fate; seldom are m6A sites to be found in housekeeping genes (18,24-26). Although the majority of research efforts thus far have focused on mRNAs, researchers have also identified m6A sites on ribosomal RNAs (rRNAs) (27,28) and transfer RNAs (tRNAs) (29). In addition, m6A sites have been identified in non-coding RNAs (ncRNAs), which cannot be translated into proteins, but have essential functions in gene expression and regulation, as well as long ncRNAs (lncRNAs) (30,31), microRNAs (miRNAs/miRs) (32,33), circular RNAs (circRNAs) (34), small nuclear RNAs (snRNAs) (35,36) and small nucleolar RNAs (snoRNAs) (37).

M6A is added by methyltransferases (termed 'writers'), removed by demethylases (termed 'erasers') and identified by m6A-RNA-binding proteins (termed 'readers'), which are able to affect the fate of m6A-RNAs, and then mediate post-transcriptional gene expression (Fig. 1 and Table I).

Table I

Overview of m6A writers, frasers andreaders.

Table I

Overview of m6A writers, frasers andreaders.

Type of m6 A and protein nameLocationfunction(Refs.)
WritersMETTL3Nuclear speckleCatalytic subunit, transfer methyl group from SAM to target RNA(16,26,39,40,43,44)
METTL14Nuclear speckleAllosteric activator, catalytically inactive recognize substrate RNA and enhance METTL3 activity(26,41-44)
WTAPNucleusMETTL3 adaptor anchor METTL3-METTL14 heterodimer complex to nuclear speckles and target mRNA(43,47,48)
VIRMA/KIAA1429NucleusWATP interactor, modification preference of m6A in 3,UTR and near stop codon shorten 3,UTR(49)
RBM15/15BNucleusWATP interactor, regulate genetic transcription silencing(50)
ZC3H13NucleusWATP interactor, bridge RBM15/15B toWTAPanchor ZC3H13-WTAP-VIRMA-HAKAI complex in the nucleus to facilitate m6A modification(51,52)
CBLL1/HAKAINucleusWATP interactor, modulate cell adhesion and EMT in cancer(53,54)
ErasersFTONucleusDemethylate m6A/m6Am(17,55)
ALKBH5NucleusDemethylate m6Ain the testes and play a role in spermatogenesis(56-58)
ReadersYTHsYTHDF1CytoplasmExpedite translation initiation DNA damage repair LLPS(59-61)
YTHDF2CytoplasmDegrade mRNA, LLPS(62-67)
YTHDF3CytoplasmAssist YTHDF1 and YTHDF2 LLPS(68)
YTHDC1NucleusmRNA splicing nuclear exportation epigenetic silencing autophagy(50,69-74)
YTHDC2 Nucleus/cytoplasmSpermatogenesis elevate the translation efficiency decrease mRNA abundance(75)
HNRNPsHNRNPCNucleusRegulate alternative splicing mRNA,U snRNA exportation, m6A-switch(3,35,77-80)
HNRNPGNucleusRegulate alternative splicing(81,82)
m6A-switch
HNRNPA2B1NucleusPri-miRNA processing regulate alternative splicing vesicular trafficking(83,84)
IGF2BPsIGF2BP1/2/3Nucleus and cytoplasmStabilization and storage of mRNA in physiological condition and stress response human senescence(80,85)

[i] SAM, S-adenosyl-l-methionine; 3′-UTRs, 3′-untranslated regions; EMT, epithelial-mesenchymal transition; m6Am, 2-O-dimethyladenosine; LLPS, liquid-liquid phase separation; METTL, methyltransferase like protein; WTAP, Wilms' tumor 1 associating protein; VIRMA, Vir like m6A methyltransferase associated; RBM15, RNA binding motif protein 15; ZC3H13, zinc finger CCCH type containing 13; CBLL1, Casitas B lineage lymphoma transforming sequence like protein 1; FTO, fat mass and obesity associated protein; ALKBH5, alkB homolog 5, RNA demethylase; YTHDC, YT521 B homology domain containing protein; HNRN, heterogeneous nuclear ribonucleoprotein; IGF2BPS, insulin like growth factor 2 mRNA binding proteins; YTHDF, YTH N6 methyladenosine RNA binding protein.

Writers

S-adenosyl-l-methionine (SAM) has emerged as the predominant methyl donor in vivo. Writers are considered to participate in a methyltransferase complex (MTC), which transfers methyl groups of SAM to target RNA in a highly specific manner. The MTC comprises METTL3, methyltransferase-like protein 14 (METTL14), an adaptor [Wilms' tumor 1-associating protein (WTAP)], a WTAP interactor [Vir-like m6A methyltransferase-associated (VIRMA; also known as KIAA1429)], RNA-binding motif protein (RBM)15/15B, zinc finger CCCH-type containing 13 (ZC3H13) and Casitas B lineage lymphoma transforming sequence like protein 1 [CBLL1/E3 ubiquitin-protein ligase (HAKAI)] (38). The members of this multi-subunit complex will be introduced in turn.

METTL3 and METTL14

METTL3 is the catalytic subunit of the MTC that has a SAM-binding motif and transfers methyl groups to RNA (16,39,40). METTL14 is an allosteric activator that has an interrupted SAM-binding motif, demonstrating that METTL14 is catalytically inactive (41,42). Together, METTL14 and METTL3 form a heterodimer complex (43), which participates in recognizing substrate RNA and enhancing methyltransferase activity (44). In the absence of METTL3 or METTL14, mouse embryonic stem cells have been shown to exhibit a decrease of 99% in their m6A content (26). In addition to the METTL3-METTL14 heterodimer complex, there are several other critical methylation enzymes. For example, ZCCHC4 adds a methyl group on to adenylate located in the 28S rRNA subunit (27), and METTL5-TRMT112 heterodimer complex modifies the 18S rRNA subunit (28). METTL16 independently catalyzes the formation of m6A in the pre-mRNAs, U6 snRNA and lncRNA.

WTAP

WTAP is the key METTL3 adaptor (43,45), which anchors METTL3-METTL14 heterodimer complex to nuclear speckles (46). Upon deletion of WTAP, the affinity of METTL3 to bind RNA is clearly decreased, indicating that WTAP recruits the METTL3-METTL14 heterodimer complex to target mRNAs (45).

VIRMA/KIAA1429

VIRMA/KIAA1429 contributes towards the modification preference of m6A for 3′-UTRs and in the vicinity of the stop codon. A model has been proposed wherein VIRMA may function as a scaffold to hold WTAP/HAKAI/ZC3H13 together and creates a suitable pocket mainly through WTAP to accommodate METTL3/METTL14 in order to guide m6A modification in the 3′UTR and around the stop codon. The 3′UTR has been shown to be shortened by VIRMA through its interactions with the polyadenylation cleavage factors, CPSF5 and CPSF6 (47).

RBM15/15B

lncRNA X-inactive specific transcript (XIST) regulates epigenetic silencing on the X chromosome. RBM15 and its paralogue, RBM15B, bind to and recruit the m6A methylation complex to specific sites in lncRNAs, thereby mediating the formation of m6A in XIST (48).

ZC3H13

ZC3H13 is considered to bridge RBM15/RBM15B to WTAP (49). The ZC3H13-WTAP-VIRMA-HAKAI complex has a profound effect on the regulation of m6A in RNAs. ZC3H13 plays the role of anchoring this regulatory complex in the nucleus to facilitate m6A modification (50).

CBLL1/HAKAI

In concert with E-cadherin, CBLL1 (also known as HAKAI) modulates cell adhesion and epithelial-mesenchymal transition (EMT) (51). The knockdown of HAKAI has been shown to reduce m6A levels (52). EMT provides a critical foundation for tumor metastasis, suggesting that m6A may be closely associated with tumor progression.

Erasers

Erasers are demethylases that catalyze the removal of the methyl group, thereby converting m6A into adenosine (A). At present, however, the roles of erasers under physiological conditions appear to be limited. Erasers including FTO and alkB homolog 5 (ALKBH5). ALKBH5 have been identified in a limited number of tissues such as testicles. They are also active under disease- and stress-associated conditions.

FTO

Consistently with ALKBs demethylating DNA, FTO demethylates m6A in mRNA in a 2-oxoglutarate (2OG)-Fe2+ oxygenase-dependent manner (17). Nevertheless, compared with ferrous iron, the interaction with m6A is stronger, and therefore, the demethylation effect of FTO in m6Am is more efficient, which indicates that lower substrate concentrations are required and the reaction times are shorter. Furthermore, FTO exhibits differential substrate preferences: It can demethylate m6Am at the 5′-cap or m6As in the inner region of, mRNAs; m6Am at the 5′-cap or in the inner regions of snRNAs; m6A in the inner regions of U6 RNA; and N1-methyladenosine in tRNAs (53). It will be noteworthy to analyze this divergence in greater depth, and to identify which substrate(s) FTO truly catalyzes.

ALKBH5

ALKBH5 is also a 2OG-Fe2+-dependent oxidative demethyltransferase that demethylates m6A in mRNAs (54). Its expression is particularly eriched in the testes, where is fulfills a role in spermatogenesis (55). Moreover, the m6A level in the mRNA of male mice wherein ALKBH5 has been knocked down has been shown to be increased (54). These mice have been shown to be infertile, which is considered to have resulted from the apoptosis of meiotic metaphase-stage spermatocytes (54). FTO and ALKBH5 share the same sequential oxidative demethylation reaction process, which is considered to be as follows: m6A to N6-hydroxymethyladenosine to N6-formyladenosine to A (56).

Readers

A myriad of writers and erasers can reversibly add or remove methyl groups in target mRNA, which exerts a major influence in the fate of target mRNAs. When m6A-mRNA binds readers in the nucleus, this may have an impact on mRNA splicing. Once exported to the cytoplasm, m6A-mRNA may be recognized by various cytosolic readers, thereby influencing the stability, translation and localization of mRNA (3). Readers comprise YTH N6 methyladenosine RNA binding proteins (YTHDF1/2/3), YT521-B homology domain-containing proteins (YTHDC1/2), heterogeneous nuclear ribonucleoproteins (HNRNPC/G/A2B1) and the insulin-like growth factor-2 mRNA-binding proteins (IGF2BP)-1, -2 and -3.

YTHDF1

YTHDF1 has been reported to participate in expediting the processes of translation initiation and DNA damage repair. YTHDF1 promotes the loading of ribosomes to bound mRNAs to enable the interactions with initiation factors (such as eIF3) to occur, thereby promoting m6A-mRNA translation initiation (57,58). Double-strand breaks (DSBs) are responsible for the most severe type of DNA damage; the METTL3-m6A-YTHDC1 axis has been shown to enhance the deposition of DNA-RNA hybrids at DSB sites, which enables the recruitment of breast cancer type 1 susceptibility protein and DNA repair protein RAD51 homolog 1 (RAD51) for the homologous recombination (HR)-mediated reparation of DSBs (59).

YTHDF2 and YTHDF3

YTHDF2 is the most abundant protein of domain family (DF) paralogues purified from cells (60), and is crucial for the degradation of mRNA. At present, there are two prevailing hypotheses concerning the underlying mechanism. A large number of studies have confirmed that m6A erects a scaffold that juxtaposes DF1/2/3, enhancing the liquid-liquid phase separation (60-63) of RNA-protein droplets, which are subsequently partitioned into membrane-less compartments, such as processing bodies (P-bodies: mRNA decay sites) (64), or into stress granules during stress. In this manner, mRNAs may be either degraded or stored (60-63). Furthermore, the N-terminal region of YTHDF2 recruits and interacts with the superfamily homology (SH) domain of the CCR4-NOT deadenylase complex to initiate the deadenylation and degradation of target mRNAs (65). YTHDF3 functions in synergy with YTHDF1 to facilitate translation, thereby assisting YTHDF2 in the degradation of m6A-RNA (66).

YTHDC1

YTHDC1 fulfills a variety of roles in mRNA splicing, nuclear exportation, genetic transcription silencing regulated by the lncRNA XIST and autophagy. The nuclear export adaptor proteins, SRSF3 and SRSF10, identified as pre-mRNA splicing factors, competitively bind to YTHDC1. SRSF3 facilitates exon inclusion, whereas SRSF10 promotes exon skipping. YTHDC1 promotes SRSF3, whereas SRSF10 is antagonized by YTHDC1 with respect to their localization, RNA-binding capabilities and associated splicing events (67,68).

In terms of the more detailed mechanism, YTHDC1 interacts with SRSF3, thereby stimulating m6A-mRNA binding to SRSF3 and the mRNA export receptor, nuclear RNA export factor 1 (NXF1), which directs mRNAs into the nuclear export pathway (69). SRSF3 stands at the point of intersection between splicing and nuclear exportation, indicating that there may be some connection between them. Another study revealed that the m6A methylation complex stimulates the recruitment of transcription export (TREX) to target mRNAs, and TREX then additionally recruits YTHDC1 and NXF1 to trigger the effective exportation of mRNA (70). As aforementioned, RBM15 and RBM15B interact with the lncRNA XIST A-repeat region, targeting the methylation complex to lncRNA XIST, and mediating the formation of m6A in lncRNA XIST (48). M6A sites that are then identified by YTHDC1, contributing towards expediting the process of X-chromosome silencing (71). Additionally, YTHDC1 modulates autophagy by modulating the stability of nuclear mRNA of sequestosome-1, resulting in the postponement of wound healing (72).

YTHDC2

The function of YTHDC2 is currently poorly understood, although it may be instrumental to the process of spermatogenesis, elevating translation efficiency and decreasing mRNA abundance. YTHDC2 is enriched in the testes, and Ythdc2−/− mice have been shown to be infertile (73). Since YTHDC2 has been detected in the 40-80S ribosomal fraction, it can be hypothesized that YTHDC2 may interact with translation initiation elements to accelerate translation efficiency. YTHDC2 may recruit nonsense-mediated degradation machineries (including the helicases UPF1 and MOV10, and the exoribonuclease XRN1) to promote the degradation of certain mRNAs co- or post-translation (73). Further, in-depth studies are required, however, to reveal the function of YTHDC2 in m6A.

HNRNPC

HNRNPC has been implicated in alternative splicing, mRNA and uridine-rich snRNA (U snRNA) exportation, and the so-called 'm6A switch' mechanism, in which m6A induces RNA unfolding and increases the accessibility of HNRNPS to single-stranded RNA. HNRNPC controls pre-mRNA processing via the regulation of alternative splicing. The individual-nucleotide resolution UV-cross-linking and immunoprecipitation method has revealed that the position of HNRNPC determines its impact on the inclusion of alternative exons. Specifically, when HNRNPC binds to exons, alternative exons are silenced; however, when HNRNPC binds to introns, the inclusion of exons is enhanced (74). The exportation of mRNAs and U snRNAs has also been shown to be associated with HNRNPC. HNRNPC functions as a 'molecular ruler' to measure the length of the RNA polymerase II transcripts, and is involved in the sorting of transcripts into mRNA or U snRNA, depending on whether or not they are longer than the threshold length (200-300 nucleotides), leading to the export of these two different RNA species from the nucleus (75). Researchers have discovered that m6A modifies partial structures in mRNAs or lncRNAs, which exposes buried RBMs and increases the accessibility of HNRNPC to m6A-RNA, the process that is termed as the 'm6A-switch' (76). This structural alteration has been shown to contribute to the metastasis of PC cells, adding a new dimension to the manner in which m6A regulates gene expression.

HNRNPG

HNRNPG also regulates alternative splicing and the 'structural switch'. HNRNPG depends upon the Arg-Gly-Gly repeat sequence within its low-complexity region to interact with nascent m6A-mediated pre-mRNA and the phosphorylated C-terminal domain of RNA polymerase II, which ultimately regulates alternative splicing (77). M6A modification promotes the binding of HNRNPG to target RNAs in virtue of the 'structural switch' (78).

HNRNPA2B1

HNRNPA2B1 stimulates primary miRNA (or pri-miRNA) processing, participates in alternative splicing and has an essential function in vesicular trafficking. DiGeorge syndrome critical region 8 (DGCR8) serves as the miRNA microprocessor complex protein, and HNRNPA2B1 interacts with DGCR8 in order to facilitate pri-miRNA processing. In addition, HNRNPA2B1 is involved in alternative splicing (79). HNRNPA2B1 sorts specific miRNAs and lncRNAs into exosomes or microvesicles, thereby fulfilling a pivotal role in the selection of cancer-associated miRNAs and lncRNAs, emphasizing the function of HNRNPA2B1 in cancer-associated vesicular trafficking (80).

IGF2BPs

IGF2BPs participate in the stabilization and storage of mRNAs under various physiological conditions and during the stress response. The interaction between the KH3-4 di-domain of IGF2BPs and the 'GGAC' motif in m6A promotes the stability and storage of certain mRNAs during normal or stress conditions (81). In addition, IGF2BP2 (also known as IMP2) plays a crucial role in human senescence by recognizing and stabilizing MIS12 mRNA (82).

New readers

Recently, Wu et al (82) reported that proline-rich coiled-coil 2A was discovered as a new m6A reader that regulates myelination and oligodendroglial specification. Furthermore, Fragile X messenger ribonucleoprotein 1 (FMR1) may function as a sequence-context-dependent reader, negatively regulating mRNA translation by stalling the process of ribosome translocation. YTHDF1 and FMR1 competitively bind to m6A sites on mRNA. The stress granule protein G3BP is an anti-m6A reader that is repelled by m6A, enhancing the stabilization of m6A-modified mRNA (83).

3. Roles of m6A in the carcinogenesis and development of PC

As aforementioned, m6A is understood to play a pivotal role in post-transcriptional modification, modulating protein expression and ultimately affecting the structure and function of modified proteins in key signaling pathways. Numerous studies have confirmed that m6A is implicated in the proliferation, apoptosis, invasion, metastasis, EMT, metabolism and chemoresistance of PC. m6A participates in the initialization and progression of PC through altering the levels of m6A regulators and genetic variants, and also serves an important role in aberrant alternative splicing. Consequently, in the subsequent section of the review, these studies are evaluated to disclose the function of m6A in tumor onset and development (Fig. 2 and Table II).

Table II

Role of m6A in the initiation and progression of pancreatic cancer.

Table II

Role of m6A in the initiation and progression of pancreatic cancer.

Initiation factor Oncogenicity/suppressorSignaling or targetEffect(Refs.)
METTL3Oncogenicity↑Proliferation↑ Metastasis↑
Invasion↑
(103)
CSC-METTL3↑-miR-25-3p↑-PHLPP2↓-AKT-p70S6K↑Proliferation↑ Invasion↑
Metastasis↑
(84)
METTL3↑-NUCB1↓Proliferation↑ Chemical
Resistance↑
(85)
Chemical Resistance↑(112)
METTL14Oncogenicity↑METTL14↑-PERP↑Proliferation↑ Apoptosis↓
Invasion↑ Metastasis↑
EMT↑
(86)
p65-METTL14↑-CDA↑Chemical Resistance↑(113)
METTL14↑- apoptosis; METTL14↑-mTOR-autophagy↓ Apoptosis↓
Chemical Resistance↓
(114)
METTLE 16Suppressor↓METTL 16↓-P21 pathway↓Proliferation↓(87)
METTL5Oncogenicity↑METTL5↑-c-myc translation↑Proliferation↑ Invasion↑
Metastasis↑
(88)
WTAPOncogenicity↑WTAP↑-mRNA↑ Metastasis↑
Chemical resistance↑
(104)
FTOSuppressor↓ FTO↓-YTHDF2-PJA2↓-Wnt signaling↑Proliferation↓ Invasion↓
Metastasis↓
(89)
FTOOncogenicity↑Proliferation↑(90)
ALKBH5Suppressor↓ ALKBH5↓-KCNK15-AS1↓ Apoptosis↑
Invasion↓Metastasis↓
(92)
ALKBH5-↓lncRNA
KCNK15-AS1↓-MDM2-REST ubiquitination↓-PTEN↓-AKT pathway↑
Apoptosis↑
invasion↓
metastasis↓
EMT↓
(93)
ALKBH5↓-PER1↓-ATM-CHK2-P53/CDC25C↓Proliferation↓ Invasion↓
Metastasis↓
(91)
ALKBH5↓-WIF-1↓-Wnt signaling↑ Proliferation↓
Invasion↓ Metastasis↓
Chemical resistance↓
(94)
ALKBH5↓-FBXL5↓-degradation↓Apoptosis↑(95)
YTHDF2Oncogenicity↑YTHDF2↑-YAP↓Proliferation↑ Invasion↓
Metastasis↓ EMT↓
(105)
YTHDF3Oncogenicity↑YTHDF3↑-lncRNA DICER1-AS1↓-DICER1↓-maturation of miR-5586-5p↓ Proliferation↑
Metastasis↑
Metabolism
(110)
HNRNPA2B1Oncogenicity↑ HNRNPA2B1-ERK/snail-E-cadherin↓N-cadherin and vimentin↑Invasion↑ EMT↑(106)
IGF2BP2Oncogenicity↑ IGF2BP2↑-DANCR↑Proliferation↑(96)
IGF2BP2↑-mRNA GLUT1↑Proliferation↑ Metabolism(111)
IGF2BP2↑- protein
IMP2↑
Apoptosis↑
Metastasis↑ EMT↑
(107)
LINC00857Oncogenicity↑ LINC00857↑-miR-150-5p↓-E2F3↑ Proliferation↑
Apoptosis↓
(97)
LINC00261Suppressor↓ LINC00261↓-IGF2BP1↑-c-Myc↑ Proliferation↑
Apoptosis↓ Metabolism
(98)
LINC00261↓-miR-222-3p/HIPK2/ Proliferation↑
Apoptosis↓ Metabolism
(98)
ERK↑-c-myc↑
rs7495 genetic varianthnRNPC↑Proliferation↑(100)
rs7495G allele-has-miR-Proliferation↑(99)
183-3p-hnRNPC↑
rs142933486 genetic variantPIK3CB↑ Proliferation↑
Metastasis↑
(101)
CLK1↑METTL14exon 10skipping↓Metastasis↑(108)
Cyclin L2exon6.3skipping↑Proliferation↑(108)
HNRNPCStructural switchTAF8L↓Metastasis↑(109)
TAF8S↑Metastasis↑(109)

[i] '↑' represents upregulation and '↓' represents downregulation. EMT, epithelial-mesenchymal transition; CSC, cigarette smoke condensate; NUCB1, nucleobindin 1; PERP, p53 effector related to PMP-22; PJA2, praja ring finger ubiquitin ligase 2; METTL, methyltransferase-like protein; WTAP, Wilms' tumor 1-associating protein; VIRMA, Vir-like m6A methyltransferase-associated; RBM15, RNA-binding motif protein 15; ZC3H13, zinc finger CCCH-type containing 13; CBLL1, Casitas B lineage lymphoma transforming sequence like protein 1; FTO, fat mass and obesity-associated protein; ALKBH5, alkB homolog 5, RNA demethylase; YTHDC, YT521-B homology domain-containing protein; HNRN, heterogeneous nuclear ribonucleoprotein; IGF2BPS, insulin-like growth factor-2 mRNA-binding proteins; YTHDF, YTH N6 methyl- adenosine RNA binding protein.

Proliferation and apoptosis

By maintaining proliferation and evading subsequent apoptotic processes, m6A effectively stimulates the progression of PC. Recently, there has been a growing understanding that m6A modulates the proliferation and apoptosis of PC cells via alterations in the levels of both m6A regulators and lncRNAs, and genetic variants.

A large number of studies have focused on the upregulation and oncogenic functions of METTL3 in PC. For example, cigarette smoke condensate (CSC) has been shown to induce the hypomethylation of the METTL3 promoter, which contributes to the overexpression of METTL3. Enhanced m6A modifications caused by elevated levels of METTL3 lead to excessive pri-miR-25 maturation. PH domain leucine-rich repeat protein phosphatase 2 (PHLPP2) is suppressed by miR-25-3p, which stimulates AKT-p70S6K signaling and ultimately promotes the proliferation of PC cells (84). In addition, the overexpression of nucleobindin 1 (NUCB1; a calcium-binding protein) has been shown to suppress cell proliferation, whereas METTL3 downregulates the expression of NUCB1 in a YTHDF2-dependent manner (85). Taken together, these studies reveal that METTL3 is implicated in promoting oncogenic pri-miR-25 maturation and suppressing expression of the tumor suppressor NUCB1 to exerts its oncogenic effects.

Reverse transcription-PCR and western blot analyses have been used to reveal that, compared with adjacent normal tissue samples, METTL14 expression is upregulated in PC samples. The protein p53 effector related to PMP-22 (PERP) functions as a p53 effector, which regulates apoptosis induced by DNA damage. The upregulation of METTL14 leads to an elevation in the m6A level, which causes a decrease in the expression of PERP, promoting cell growth and inhibiting apoptosis (86). Hence, METTL14 has been regarded as an oncogenic regulator in PC. In addition to METTL3-METTL14, other methylation enzymes are also involved in the modulation of PC proliferation. METTL16 has been shown to suppress the proliferation of PC cells through the P21 pathway (87). METTL5 specifically catalyzes the methylation of A1832 of 18S rRNA, which is a crucial point in the decoding mechanism, indicating its importance as a regulatory protein in translation. METTL5 has been demonstrated to function as an oncogene that promotes the proliferation of PC cells by increasing c-Myc translation (88).

As regards the functions of demethylation enzymes, the impact of FTO on the proliferation of PC cells remains controversial. Zeng et al (89) identified that FTO was a tumor suppressor protein. Reduced FTO expression led to the upregulation of the m6A level of praja ringfinger ubiquitin ligase 2 (PJA2), and an enhanced rate of PJA2 degradation was shown to be dependent on YTHDF2, which promoted Wnt signaling, ultimately facilitating the proliferation of PC cells. By contrast, Tang et al (90) demonstrated that, when overexpressed, FTO functions as as oncogenic protein. The knockdown of FTO resulted in compromised levels of cell proliferation. The effects that are actually elicited by FTO in PC remain an interesting topic that warrants further investigation. Differently from FTO, another demethylation enzyme, ALKBH5, has over time been shown to function as a tumor suppressor (91-95), often involved in inhibiting the proliferation of PC. ALKBH5 demethylates period circadian regulator 1 (PER1), and it functions in concert with YTHDF2 to activate PER1. The overexpression of PER1 activates the ATM-CHK2-P53/CDC25C signaling pathway, which leads to the inhibition of tumor proliferation. p53 activates the transcription of ALKBH5, which subsequently functions in a feedback loop to regulate the m6A level (91). Furthermore, ALKBH5 has been shown to exert an anticancer effect against PC by targeting regulators of ferroptosis. Mechanistically, the ubiquitin ligase FBXL5 mediates the degradation of PC cells via ferroptosis. A previous study demonstrated that not only was ALKBH5 responsible for enhancing the stability of FBXL5, but it also led to upregulation of the expression of FBXL5 and the mitochondrial iron importer SLC25A28, and alternative splicing of SLC25A37 was also increased (95).

Although numerous studies have focused on alterations in the level of m6A regulators with respect to their role as initiation factors in the proliferation and apoptosis of PC, to date, relatively few studies have been performed to assess the mechanisms through which m6A functions in lncRNAs. Nevertheless, there is sufficient evidence to suggest that lncRNAs also function as initiation factors in the proliferation and apoptosis of PC. First, the lncRNA DANCR was shown to promote the tumorigenesis and proliferation of PC; IGF2BP2 recognizes m6A modified at A664 of DANCR, which plays the role of stabilizing DANCR (96). Secondly, LINC00857 has been shown to be overexpressed, thereby promoting the tumorigenesis of PC in an m6A-mediated manner (97). The deletion of LINC00857 clearly hinders cell proliferation and facilitates cell apoptosis. M6A is pre-eminently enriched within LINC00857, which enhances its RNA stability, thereby suggesting a role for LINC00857 in modulating the miR-150-5p/E2F3 signaling axis. As a competitive endogenous RNA, LINC00857 exerts its effects through sponging miR-150-5p and upregulating the expression of E2F3 (97). Finally, LINC00261 has been reported to function as a tumor suppressor that inhibits proliferation, and accelerates the rate of apoptosis of PC tissues and cell lines. The EZH2-mediated histone H3K27 trimethylation and DNA hypermethylation both serve to downregulate LINC00261. Mechanistically, on the one hand, the downregulation of LINC00261 leads to the upregulation of the experession of c-Myc by binding to miR-222-3p, thereby activating the HIPK2/ERK/c-Myc signaling pathway. On the other hand, LINC00261 negatively regulates the expression of IGF2BP1, where the overexpression of IGF2BP1 has been shown to enhance c-Myc stability (98). From this example, it may be noted that there are links among m6A, DNA modification and histone modification.

Genetic variants, as another type of initiation factor, may provide novel insight into the association between m6A regulators and PC. For example, the genetic variant rs7495 in the 3′-UTR region of hnRNPC has been shown to disrupt a presumptive binding site for has-miR-183-3p that promotes the expression of hnRNPC, and which contributes to the susceptibility of PC (99). In another study, cell viability assay indicated that the depletion of hnRNPC hindered the proliferation of PC (100). Similarly, the rs142933486 variant in the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta (PIK3CB) has been observed to have decreased m6A levels, and its expression is increased in a YTHDF2-dependent manner. In addition, the variant of PIK3Cb[T] has been shown to activate AKT signaling, accelerating the proliferation of phosphatase and tensin homolog (PTEN)-deficient PC cells (101).

Generally, it can be seen that alterations in the levels of m6A regulators and lncRNAs, together with genetic variants, are intimately involved in the proliferation and apoptosis of PC.

Invasion, metastasis and EMT

As is currently understood, PC is characterized by a high metastatic rate: 40-50% of patients present with distant metastasis in their first hospital visit. Metastasis is a complex process that comprises EMT, the migration of cancerous cells, their infiltration into adjacent tissues, vascular transportation and settlement in distant organs. EMT is defined as the transformation from polar epithelial cells into interstitial cells under specific physiological and pathological conditions. The process is mainly characterized by the loss of epithelial phenotypic molecules (such as E-cadherin) and the acquisition of interstitial cell characteristics (such as vimentin), which enables tumor cells to lose their intercellular adhesive properties and become more loose, thereby gaining the abilities of invasion and metastasis (102). M6A regulates the invasion, metastasis and EMT of PC through alterations in the levels of m6A regulators, genetic variants and aberrant alternative splicing.

In addition to sustaining proliferation, METTL3 promotes the invasion and migration of PC cells (103). The CSC/METTL3/miR-25-3p/PHLPP2/AKT/p70S6K signaling axis has been shown to accelerate invasion and metastasis (84). Furthermore, PERP serves as a tetraspan plasma membrane protein, having multifaceted roles in EMT and cell-cell adhesion. Of note, the tumor suppressor function of PERP may involve inhibition of the invasion, metastasis and EMT of PC cells. METTL14 downregulates the expression of PERP through the METTL14/YTHDF2/PERP pathway (86). Furthermore, METTL5 promotes the invasion and migration of PC cells. METTL5 functions as an oncogenic regulator by increasing the translation of c-Myc, whereas the knockdown of c-Myc has been shown to abolish the oncogenic effects induced by the upregulation of METTL5 (88). In addition, both nuclear and cytoplasmic WTAP have been shown to be upregulated, promoting metastasis by modulating the stability of downstream mRNA (104).

Studies on the function of FTO in PC metastasis are relatively limited, although one study documented that FTO facilitates the invasion and metastasis of PC cells via the FTO/YTHDF2/PJA2/Wnt signaling axis (89). Moreover, lncRNA KCNK15-AS1 inhibits the invasion and migration of PC. ALKBH5 is responsible for the demethylation and upregulation of KCNK15-AS1. In PC, ALKBH5 is downregulated, and the expression of KCNK15-AS1, lying downstream in the pathway, is subsequently also downregulated (92). He et al (92) also discovered that the overexpression of KCNK15-AS1 suppresssed EMT and cell migration in PC. Mechanistically, KCNK15-AS1 was shown to recruit the MDM2 proto-oncogene to promote the ubiquitination of RE1 silencing transcription factor (REST), thereby inactivating the AKT pathway via the transcriptional upregulation of PTEN (93). Furthermore, the upregulation of ALKBH5 has been shown to inhibit tumoral invasive and migrative activities in vitro, whereas the depletion of ALKBH5 promotes tumor progression. ALKBH5 demethylates and regulates PER1 in an m6A-dependent manner. PER1 itself has the role of reactivating the ATM/CHK2/P53/CDC25C signaling axis, which inhibits the processes of cell invasion and migration (91).

Additionally, the upregulation of YTHDF2 in PC tissues facilitates cell proliferation, and suppresses invasion and migration, a phenomenon that has been defined as the 'migration-proliferation dichotomy'. Moreover, deficiencies in YTHDF2 have been shown to promote EMT, probably via the upregulation of Yes-associated protein 1, and not via TGF-β/Smad signaling in PC cells (105). Furthermore, HNRNPA2B1 has been shown to influence EMT progression in PC via the ERK/Snail signaling pathway. Both in vitro and in vivo, HNRNPA2B1 downregulates E-cadherin, and upregulates N-cadherin and vimentin to enhance EMT, thereby promoting the invasion of PC cell lines (106). In addition, the IGF2BP2 binding protein, IMP2, has been shown to be upregulated in PC, particularly upon the treatment of Panc-1 PC cells with TGF-β, where the induction of EMT indicates its oncogenic effectivness (107).

Apart from alterations in m6A regulators, the PIK3Cb[T] genetic variant has been shown to promote metastasis of PTEN-deficient PC cells (101). Alternative splicing has also been identified as a novel means through which m6A may modulate gene expression. For example, Cdc2-like kinase 1 (CLK1) is an alternative splicing-associated gene that is significantly upregulated in PC tissues. An increased expression level of CLK1 has been shown to enhance phosphorylation on SR-like splicing factor 5250-Ser (SRSF5250-Ser), which subsequently suppresses the exon skipping of METTL14exon10, but facilitates the exon skipping of cyclin L2exoN6.3. The alterative skipping of METTL14exon 10 increases the adundance of m6A and promotes metastasis. Alterative cyclin L2exoN6.3 skipping promotes proliferation (108). Moreover, HNRNPC regulates alternative splicing via the 'm6A switch' to accelerate the invasion and liver metastasis of PC. HNRNPC inhibits the anti-metastatic isoform of TAF8 (TAF8L), but promotes the pro-metastatic isoform of TAF8 (TAF8S). Mechanistically, m6A increases the likelihood of pre-mRNA of TAF8 to form a linear structure, which increases the accessibility of HNRNPC to TAF8, and upregulates the expression of the pro-metastatic isoform, TAF8S (109).

Above all, various studies, as aformentioned, have described the crucial function of m6A in the invasion, metastasis and EMT of PC, and these findings may provide novel prospects for m6A therapy in the future.

Metabolism

The initiation and progression of carcinoma requires the reprogramming of metabolism. Tumor cells automatically transform their flux via diverse metabolic pathways to accommodate an elevated bioenergetic and biosynthetic requirement. The influence of m6A in altering metabolism has proven to be a rich avenue for scientific exploration. Scientists have presented several mechanisms to explain the mechanisms through which m6A may promote metabolism transformation and tumor progression.

For example, bioinformatics analysis has revealed that DICER1 antisense RNA 1 (DICER1-AS1) is overexpressed in PC. DICER1-AS1 recruits the transcription factor YY1 to the promoter of DICER1 in order to expedite the transcription of DICER1. The maturation of miR-5586-5p is promoted by DICER1 to inhibit the expression of glycolytic genes, including LDHA, HK2, PGK1 and SLC2A1. YTHDF3 forms a negative feedback loop with DICER1-AS1 to facilitate glycolysis. Moreover, the upregulation of DICER1-AS1 has been shown to inhibit the proliferation and metastasis of PC (110).

In addition, in order to meet the enhanced metabolic requirements, tumor cells are often observed to upregulate the expression of glucose transporters (GLUTs), such as GLUT1, to augment glucose uptake. IGF2BP2 overexpressed in PC has been shown to increase the rate of aerobic glycolysis and PC cell proliferation by stabilizing GLUT1 mRNA (111). Similarly, the epigenetic silencing of LINC00261 has been found to modulate the miR-222-3p/HIPK2/ERK signaling axis, and IGF2BP1 has been shown to promote c-Myc-mediated aerobic glycolysis (98).

Taken together, the studies published to date in this area have demonstrated that m6A plays a role of paramount importance as a regulator of metabolic alterations, and this function appears to play a crucial role in PC.

Chemoresistance

PC is characterized by a highly aggressive progression: Only 15-20% of patients with PC are identified at an early stage, whereas the majority of them (75-80%) exhibit either locally advanced or distant metastasis, and should receive chemotherapy as a first-line treatment. However, in spite of this, numerous patients fail to benefit from the all the benefits potentially afforded by the therapy due to rapid drug resistance induced by the chemotherapeutic agents. Chemoresistance is a major impediment in terms of the efficacy of therapeutic drugs.

A previous study revealed that the overexpression of NUCB1 activated the antitumor effects of gemcitabine (GEM) by inhibiting the GEM-induced unfolded protein response (UPR) and autophagy; in PC, METTL3-YTHDF2 downregulated NUCB1 (85). The knockdown of METTL3 has been shown to sensitize PC cells to irradiation and antitumor drugs (namely, 5-fluorouracil, GEM and cisplatin). METTL3 targets various pivotal pathways, including the ubiquitin-dependent pathway and the MAPK signaling cascades. Alterations in these pathways trigger aberrant biological behaviors, which may lead to chemotherapeutic and radiation resistance (112).

Moreover, overexpression of METTL14 is associated with resistance to GEM in PC. Cytidine deaminase (CDA) functions as an enzyme which induces resistance to GEM by inactivating GEM. METTL14 was overexpressed in PC via its promoter region binding to the transcriptional factor p65, thereby increasing the expression of CDA (113). By contrast, the upregulation of METTL14 enhances apoptosis to sensitize PC cells to cisplatin, and also promotes autophagy via an mTOR-dependent signaling pathway (114). WTAP has been identified as an oncogenic regulator, which modulate the stability of downstream mRNA, thereby promoting chemoresistance to GEM (104).

Furthermore, ALKBH5 has been shown to be downregulated in a patient-derived xenograft model treated with GEM. ALKBH5 upregulates the expression of Wnt inhibitory factor-1, which subsequently suppresses the Wnt pathway. Wnt signaling boosts the resistance of PC cells to GEM. The increased expression of ALKBH5 has also been shown to sensitize PC cells to chemotherapy (94). Taken together, the upregulation of METTL3, METTL14 and WTAP, and the downregulation of ALKBH5 have been shown to lead to resistance to GEM. On the other hand, another study indicated that increased levels of METTL14 sensitized PC cells to cisplatin, and this apparent contradiction is worthy of further exploration in future studies.

Although the various capabilities and properties of m6A have been discussed separately in this review, it should be noted that the functional roles of m6A in terms of understanding the occurrence and progression of PC are all intrinsically associated, and the process cannot be comprehended by considering the individual capabilities of m6A entirely in isolation.

4. Possible clinical applications of m6A in PC

m6A is closely associated with every stage in the development of PC, and is therefore predicted to be of utmost importance in planning future diagnostic and therapeutic directions for PC. However, to date, a large number of studies have concentrated on the fundamental biological and physiological aspects of m6A at the laboratory level, and few studies have focused on translating these findings into clinical practice. Thus, the final section of the present review summarizes the possible clinical applications of m6A in PC, in an attempt to broaden the current understanding of this post-transcriptional modification (Table III).

Table III

Role of m6A in the pathological stage and poor prognosis of pancreatic cancer.

Table III

Role of m6A in the pathological stage and poor prognosis of pancreatic cancer.

Initiation factor Oncogenicity/suppressorSignaling or targetEffect(Refs.)
METTL3Oncogenicity↑Pathological stage
Poor prognosis↑
(103)
CSC-METTL3↑-miR-25-3p↑-PHLPP2↓-AKT-p70S6K↑Poor prognosis↑(84)
WTAPOncogenicity↑WTAP↑- mRNA↑Poor prognosis↑(104)
FTOSuppressor↓ FTO↓-YTHDF2-PJA2↓-Wnt signaling↑Pathological stage
Poor prognosis↓
(89)
ALKBH5Suppressor↓ ALKBH5↓-PER1↓-ATM-CHK2-P53/CDC25C↓Poor prognosis↓(91)
ALKBH5↓-WIF-1↓-Wnt signaling↑Poor prognosis↓(94)
ALKBH5↓-FBXL5↓-degradation↓Poor prognosis↓(95)
YTHDF2Oncogenicity↑YTHDF2↑-YAP↓Pathological stage
Poor prognosis↑
(105)
IGF2BP2Oncogenicity↑ IGF2BP2↑-DANCR↑Poor prognosis↑(96)
IGF2BP2↑-mRNA GLUT1↑Poor prognosis↑(111)
IGF2BP2↑-protein IMP2↑Poor prognosis↑(107)
LINC00857Oncogenicity↑ LINC00857↑-miR-150-5p↓-E2F3↑Poor prognosis↑(97)
CLK1↑□ Phosphorylationon
SRSF5250-Ser↑
METTL14exon 10skipping↓Pathological stage
Poor prognosis↑
(108)
Cyclin L2exon6.3skipping↑(108)
StructuralHNRNPCTAF8L↓Pathological stage(109)
switchTAF8S↑Poor prognosis↑(109)

[i] '↑' represents upregulation and '↓' represents downregulation. CSC, cigarette smoke condensate; PJA2, praja ring finger ubiquitin ligase 2; METTL, methyltransferase-like protein; WTAP, Wilms' tumor 1-associating protein; RBM15, RNA-binding motif protein 15; CLK1, Cdc2-like kinase 1; FTO, fat mass and obesity-associated protein; ALKBH5, alkB homolog 5, RNA demethylase; YTHDC, YT521-B homology domain-containing protein; HNRN, heterogeneous nuclear ribonucleoprotein; IGF2BPS, insulin-like growth factor-2 mRNA-binding proteins; YTHDF, YTH N6 methyladenosine RNA binding protein.

Pathological stage and the poor prognosis

Different levels of m6A regulators represent different tumor, node, metastasis (TNM) stages, and therefore different prognoses of PC. Higher levels of expression of the methylase METTL3 are associated with a higher pathological stage (P=0.02) and a higher N stage (P=0.02) (103). METTL3 promotes pri-miR-25 maturation. Excessive levels of miR-25-3p have been detected in PC tissues and smokers, and are associated with a worse prognosis (84). Taken together, it has been demonstrated that the survival rates are shorter in patients with a high level of METTL3 expression (84,103). Furthermore, univariate analysis has demonstrated that the overexpression of nuclear WTAP is associated with a later N stage and poor overall survival (P<0.001) (104).

The decreased expression of the demethylase FTO is indicative of advanced carcinoma stages and the nodal metastasis status. The median survival rate has bene found to be significantly shorter within the low-level FTO expression group (16.90 vs. 48.67 months; P=0.0474), with a high level of LINC00857. In brief, patients with decreased levels of FTO have poor survival outcomes (89). In addition, survival analysis has revealed that the lower the level of ALKBH5 expression, the shorter the overall survival rate. Reduced ALKBH5 levels have been shown to indicate the occurrence of PC, and poor clinicopathological manifestations (91,94,95).

Emerging evidence suggests that readers, such as YTHDF2 and IGF2BP2, are of vital importance for the prognosis of PC. The overexpression of YTHDF2 in PC tissues is associated with worse tumor stages (105). Moreover, IGF2BP2 is characterized as an unfavorable prognostic marker for PC. The overexpression of IGF2BP2 has been shown to be closely associated with a poor overall survival and disease-free survival rate (96,107,111).

As demonstrated in a previous study, the upregulation of LINC00857 activates the miR-150-5p/E2F3 signaling axis to facilitate the tumorigenesis of PC. In that previous study, the overall survival rate [hazard ratio (HR), 1.6; P=0.034] and disease-free survival rate (HR, 1.9; P=0.0046) in the high-expression LINC00857 group were found to be significantly lower (97).

The CLK1/SRSF5 signaling pathway promotes the abnormal exon skipping of METTL14 and cyclin L2 to facilitate the growth and metastasis of PC. In a previous study, univariate analysis suggested that the overexpression of CLK1 was representative of a severe TNM stage, lymphatic metastasis and tumor size (108). An increased CLK1 expression was also shown to be associated with a lower overall survival rate (108). HNRNPC impedes anti-metastatic alternative splicing events in an m6A-dependent manner. The level of HNRNPC has been found to be markedly higher in hepatic metastatic tissues compared with non-metastatic tissues (P<0.01) (109). Kaplan-Meier curves were previously employed to demonstrated that the overall survival rate was significantly lower in the HNRNPC overexpression group (both log-rank P<0.05). As aforementioned, the hepatic metastasis rate was confirmed to be significantly higher in the HNRNPC overexpression group (log-rank P=0.008) (109).

However, although there is a significant difference in the abundance of m6A regulators between PC and normal tissues, m6A regulators has not been widely utilized as a diagnostic marker. However, it is considered that, with the anticipated continued improvement of mapping methods, m6A regulators will be able to precisely predict the occurrence, TNM stage and prognosis of PC in the not-too-distant future.

M6A inhibitors as potential therapeutics in PC

A wealth of studies has confirmed the complex functions and molecular mechanisms of m6A in PC. Targeting m6A regulators as a means of therapeutic invention therefore provides a promising prospect for the treatment of cancer (Table IV).

Table IV

N6 methyladenosine inhibitors.

Table IV

N6 methyladenosine inhibitors.

TargetDrugCancer type(Refs.)
FTORheinBreast cancer(115,116)
MAGlioblastoma stem(117,118)
FB23/FB23-2Acute myeloid leukemia(119)
R-2HGLeukemia cell(120)
METTL3Adenosine (1-8)(121)
UZH1a, UZH2 and UZH1a analogue(121)
STM2457Acute myeloid leukemia(121-123)
METTL14SPI1Acute myeloid leukemia(124)
WTAPCA4Colon cancer(125)
DNA double-strand breaksFisetinPancreatic cancer(126)

[i] MA, meclofenamic acid.

FTO inhibitors can be divided into selective or non-selective types. Rhein is a natural product that was demonstrated to be the first FTO inhibitor possessing cell activity (115). Rhein has been shown to restrict the occurrence of breast cancer cells (116). Meclofenamic acid is an example of a selective FTO inhibitor (117), which restrains the growth and self-renewal of glioblastoma stem cells (118). FB23 and its derivative (FB23-2) inhibit the proliferation, and increase the rate of apoptosis and differentiation of acute myeloid leukemia (AML) cells (119). Finally, R-2-hydroxyglutarate has been shown to exert an anti-leukemic effect by suppressing cell proliferation/viability, and enhancing the apoptosis and cell-cycle arrest of leukemia cells (120).

METTL3 is a crucial catalytic subunit, which has attracted extensive attention among all regulators. METTL3 inhibitors can be separated into nucleosides and non-nucleosides. Inhibitors in both of these categories have been shown to function as competitive substrate inhibitors of SAM. High-throughput analysis identified adenosine (1-8) as small-molecule METTL3 inhibitors which occupy the binding site of SAM. The disadvantages of these adenosine (1-8) inhibitors, however, are poor selectivity and poor cellular permeability properties. The discovery of the METTL3 non-nucleoside inhibitor UZH1a, UZH2 and UZH1a analogues, JMC-1, JMC-5, JMC-8 and JMC-10, promoted the development of METTL3 inhibitors as therapeutic targets (121). The non-nucleoside METTL3 inhibitor, STM2457, has also been shown to be effective against AML without exerting any significant effect on normal hematopoiesis (121-123).

SPI1 has been shown to inhibit METTL14 expression directly, and serves as a possible AML therapeutic target (124). Additionally, carbonic anhydrase IV has been found to suppress the tumorigenesis of colon cancer by inhibiting the WTAP-WT1-TBL1 pathway (125).

Currently, research regarding m6A inhibitors in PC is still at its infancy. DNA damage repair represents a prominent obstacle for evaluating the chemotherapy efficacy of PC. The knockdown of PHF10 has been shown to result in the elevated recruitment of γ-H2AX, RAD51 and 53BP1 to DSB sites, and decreased HR repair efficiency. Fisetin induces DSBs, and suppresses HR repair of DNA through impeding the ZC3H13-mediated m6A regulation of PHF10. Fisetin treatment has also provided insight into novel therapeutic strategies for PC (126).

Taken together, m6A inhibitors present a frontier of research that may provide a novel direction in m6A-targeted drug therapeutics. However, numerous challenges lie ahead. The clinical application of m6A inhibitors in PC remains insufficient at present, and the efficacy and adverse reactions of the prospective inhibitors requires further verification.

5. Conclusions and future perspectives

In the present review, the biological functions of m6A were introduced, with a focus on its influence in PC. There are, however, certain divergences and discrepancies that have not been fully explored herein. Based on these unresolved discrepancies, the authors' prediction is that the future exploration of m6A will give attention to the following topics. First, it is possible that certain m6A writers, erasers and readers have not yet to be identified; therefore, reforming m6A-mapping methods will be necessary to promote the discovery of novel m6A regulators. Secondly, FTO remains controversial with respect to whether it actually targets m6A as a substrate, and what its precise role is in the proliferation of PC. Thirdly, the question remains as to whether erasers always function antagonistically with writers. For example, if METTL3-METTL4 serves as an oncogene, FTO may function as suppressor, although this would not be in agreement with the experimental results described above. The detailed mechanisms that account for how the actions of different regulators are coordinated in the same tumor, and also how the same regulator may function heterogeneously in distinct type of carcinomas, need to be further elucidated. The current knowledge of m6A regulatory networks only represent the tip of an iceberg, and the real situation is far more complex than what has already been discovered. Fourthly, the mechanisms through which m6A functions in concert with DNA and histone modification, and whether there are underlying connections between them, remain unclear. Finally, the exploration of m6A inhibitors in PC is only at the rudimentary experimental stages at present. An extensive literature detailing their application in clinical trials does not yet exist. Efficacious and safe m6A inhibitors, however, do need to be developed and put forth into clinical practice.

Availability of data and materials

Not applicable.

Authors' contributions

GZ conceived the study. TY wrote the manuscript and performed the literature search with assistance from JW, HZ and PL, and GZ supervised the whole process. All authors have read and approved the final manuscript. Data authentication is not applicable.

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

The present study was supported by the National Natural Science Foundation of China (grant no. 82070575).

References

1 

Mizrahi JD, Surana R, Valle JW and Shroff RT: Pancreatic cancer. Lancet. 395:2008–2020. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Martin-Perez E, Domínguez-Muñoz JE, Botella-Romero F, Cerezo L, Matute Teresa F, Serrano T and Vera R: Multidisciplinary consensus statement on the clinical management of patients with pancreatic cancer. Clin Transl Oncol. 22:1963–1975. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Zaccara S, Ries RJ and Jaffrey SR: Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 20:608–624. 2019. View Article : Google Scholar : PubMed/NCBI

4 

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

5 

Zhou Z, Lv J, Yu H, Han J, Yang X, Feng D, Wu Q, Yuan B, Lu Q and Yang H: Mechanism of RNA modification N6-methyladenosine in human cancer. Mol Cancer. 19:1042020. View Article : Google Scholar : PubMed/NCBI

6 

He L, Li H, Wu A, Peng Y, Shu G and Yin G: Functions of N6-methyladenosine and its role in cancer. Mol Cancer. 18:1762019. View Article : Google Scholar : PubMed/NCBI

7 

Frye M, Harada BT, Behm M and He C: RNA modifications modulate gene expression during development. Science. 361:1346–1349. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Sun T, Wu R and Ming L: The role of m6A RNA methylation in cancer. Biomed Pharmacother. 112:1086132019. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Lan Q, Liu PY, Haase J, Bell JL, Huttelmaier 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

11 

Wei CM, Gershowitz A and Moss B: Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell. 4:379–386. 1975. View Article : Google Scholar : PubMed/NCBI

12 

Rottman F, Shatkin AJ and Perry RP: Sequences containing methylated nucleotides at the 5′ termini of messenger RNAs: Possible implications for processing. Cell. 3:197–199. 1974. View Article : Google Scholar : PubMed/NCBI

13 

Desrosiers R, Friderici K and Rottman F: Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Natl Acad Sci USA. 71:3971–3975. 1974. View Article : Google Scholar : PubMed/NCBI

14 

Adams JM and Cory S: Modified nucleosides and bizarre 5′-termini in mouse myeloma mRNA. Nature. 255:28–33. 1975. View Article : Google Scholar : PubMed/NCBI

15 

Schäfer KP: RNA synthesis and processing reactions in a subcellular system from mouse L cells. Hoppe Seylers Z Physiol Chem. 363:33–43. 1982. View Article : Google Scholar : PubMed/NCBI

16 

Bokar JA, Shambaugh ME, Polayes D, Matera AG and Rottman FM: Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA. 3:1233–1247. 1997.PubMed/NCBI

17 

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

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 

Schibler U, Kelley DE and Perry RP: Comparison of methylated sequences in messenger RNA and heterogeneous nuclear RNA from mouse L cells. J Mol Biol. 115:695–714. 1977. View Article : Google Scholar : PubMed/NCBI

20 

Rottman FM, Desrosiers RC and Friderici K: Nucleotide methylation patterns in eukaryotic mRNA. Prog Nucleic Acid Res Mol Biol. 19:21–38. 1976. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Krug RM: Influenza viral mRNA contains internal N6-methyladenosine and 5′-terminal 7-methyl-guanosine in cap structures. J Virol. 20:45–53. 1976. View Article : Google Scholar : PubMed/NCBI

23 

Beemon K and Keith J: Localization of N6-methyladenosine in the rous sarcoma viru genome. J Mol Bid. 113:165–179. 1977. View Article : Google Scholar

24 

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

25 

Schwartz S, Mumbach MR, Jovanovic M, Wang T, Maciag K, Bushkin GG, Mertins P, Ter-Ovanesyan D, Habib N, Cacchiarelli D, et al: Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5′ sites. Cell Rep. 8:284–296. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AA, Kol N, Salmon-Divon M, Hershkovitz V, Peer E, Mor N, Manor YS, et al: Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science. 347:1002–1006. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Ma H, Wang X, Cai J, Dai Q, Natchiar SK, Lv R, Chen K, Lu Z, Chen H, Shi YG, et al: N6-methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat Chem Biol. 15:88–94. 2019. View Article : Google Scholar

28 

van Tran N, Ernst FGM, Hawley BR, Zorbas C, Ulryck N, Hackert P, Bohnsack KE, Bohnsack MT, Jaffrey SR, Graille M and Lafontaine DLJ: The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 47:7719–7733. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Ueda Y, Ooshio I, Fusamae Y, Kitae K, Kawaguchi M, Jingushi K, Hase H, Harada K, Hirata K and Tsujikawa K: AlkB homolog 3-mediated tRNA demethylation promotes protein synthesis in cancer cells. Sci Rep. 7:422712017. View Article : Google Scholar : PubMed/NCBI

30 

Luo H, Zhu G, Xu J, Lai Q, Yan B, Guo Y, Fung TK, Zeisig BB, Cui Y, Zha J, et al: HOTTIP lncRNA promotes hematopoietic stem cell self-renewal leading to AML-like disease in mice. Cancer Cell. 36:645–659.e8. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Zheng ZQ, Li ZX, Zhou GQ, Lin L, Zhang LL, Lv JW, Huang XD, Liu RQ, Chen F, He XJ, et al: Long noncoding RNA FAM225A promotes nasopharyngeal carcinoma tumorigenesis and metastasis by acting as ceRNA to sponge miR-590-3p/miR-1275 and upregulate ITGB3. Cancer Res. 79:4612–4626. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Cesarini V, Silvestris DA, Tassinari V, Tomaselli S, Alon S, Eisenberg E, Locatelli F and Gallo A: ADAR2/miR-589-3p axis controls glioblastoma cell migration/invasion. Nucleic Acids Res. 46:2045–2059. 2018. View Article : Google Scholar :

33 

Han J, Wang JZ, Yang X, Yu H, Zhou R, Lu HC, Yuan WB, Lu JC, Zhou ZJ, Lu Q, et al: METTL3 promote tumor proliferation of bladder cancer by accelerating pri-miR221/222 maturation in m6A-dependent manner. Mol Cancer. 18:1102019. View Article : Google Scholar : PubMed/NCBI

34 

Li J, Huang C, Zou Y, Ye J, Yu J and Gui Y: CircTLK1 promotes the proliferation and metastasis of renal cell carcinoma by sponging miR-136-5p. Mol Cancer. 19:1032020. View Article : Google Scholar : PubMed/NCBI

35 

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

36 

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

37 

Linder B, Grozhik AV, Olarerin-George AO, Meydan C, Mason CE and Jaffrey SR: Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods. 12:767–772. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Horiuchi K, Kawamura T, Iwanari H, Ohashi R, Naito M, Kodama T and Hamakubo T: Identification of Wilms' tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J Biol Chem. 288:33292–33302. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Narayan P and Rottman FM: An in vitro system for accurate methylation of internal adenosine residues in messenger RNA. Science. 242:1159–1162. 1988. View Article : Google Scholar : PubMed/NCBI

40 

Schumann U, Shafik A and Preiss T: METTL3 gains R/W access to the epitranscriptome. Mol Cell. 62:323–324. 2016. View Article : Google Scholar : PubMed/NCBI

41 

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

42 

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

43 

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 :

44 

Wang X, Feng J, Xue Y, Guan Z, Zhang D, Liu Z, Gong Z, Wang Q, Huang J, Tang C, et al: Corrigendum: Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature. 542:2602017. View Article : Google Scholar

45 

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. 24:177–189. 2014. View Article : Google Scholar : PubMed/NCBI

46 

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

47 

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

48 

Patil DP, Chen CK, Pickering BF, Chow A, Jackson C, Guttman M and Jaffrey SR: m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 537:369–373. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Knuckles P, Lence T, Haussmann IU, Jacob D, Kreim N, Carl SH, Masiello I, Hares T, Villaseñor R, Hess D, et al: Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m6A machinery component Wtap/Fl(2)d. Genes Dev. 32:415–429. 2018. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

Fujita Y, Krause G, Scheffner M, Zechner D, Leddy HE, Behrens J, Sommer T and Birchmeier W: Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol. 4:222–231. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Bawankar P, Lence T, Paolantoni C, Haussmann IU, Kazlauskiene M, Jacob D, Heidelberger JB, Richter FM, Nallasivan MP, Morin V, et al: Hakai is required for stabilization of core components of the m6A mRNA methylation machinery. Nat Commun. 12:37782021. View Article : Google Scholar

53 

Wei J, Liu F, Lu Z, Fei Q, Ai Y, He PC, Shi H, Cui X, Su R, Klungland A, et al: Differential m6A, m6Am, and m1A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell. 71:973–985.e5. 2018. View Article : Google Scholar

54 

Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, Vågbø CB, Shi Y, Wang WL, Song SH, et al: ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 49:18–29. 2013. View Article : Google Scholar :

55 

Aik W, Scotti JS, Choi H, Gong L, Demetriades M, Schofield CJ and McDonough MA: Structure of human RNA N6-methyladenine demethylase ALKBH5 provides insights into its mechanisms of nucleic acid recognition and demethylation. Nucleic Acids Res. 42:4741–4754. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Wang T, Kong S, Tao M and Ju S: The potential role of RNA N6-methyladenosine in cancer progression. Mol Cancer. 19:882020. View Article : Google Scholar : PubMed/NCBI

57 

Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H and He C: N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 161:1388–1399. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Liu T, Wei Q, Jin J, Luo Q, Liu Y, Yang Y, Cheng C, Li L, Pi J, Si Y, et al: The m6A reader YTHDF1 promotes ovarian cancer progression via augmenting EIF3C translation. Nucleic Acids Res. 48:3816–3831. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Zhang C, Chen L, Peng D, Jiang A, He Y, Zeng Y, Xie C, Zhou H, Luo X, Liu H, et al: METTL3 and N6-methyladenosine promote homologous recombination-mediated repair of DSBs by modulating DNA-RNA hybrid accumulation. Mol Cell. 79:425–442.e7. 2020. View Article : Google Scholar : PubMed/NCBI

60 

Liu SY, Feng Y, Wu JJ, Zou ML, Sun ZL, Li X and Yuan FL: m6A facilitates YTHDF-independent phase separation. J Cell Mol Med. 24:2070–2072. 2020. View Article : Google Scholar

61 

Ries RJ, Zaccara S, Klein P, Olarerin-George A, Namkoong S, Pickering BF, Patil DP, Kwak H, Lee JH and Jaffrey SR: m6A enhances the phase separation potential of mRNA. Nature. 571:424–428. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Gao Y, Pei G, Li D, Li R, Shao Y, Zhang QC and Li P: Multivalent m6A motifs promote phase separation of YTHDF proteins. Cell Res. 29:767–769. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Wang J, Wang L, Diao J, Shi YG, Shi Y, Ma H and Shen H: Binding to m6A RNA promotes YTHDF2-mediated phase separation. Protein Cell. 11:304–307. 2020. View Article : Google Scholar

64 

Sheth U and Parker R: Decapping and decay of messenger RNA occur in cytoplasmic processing bodies. Science. 300:805–808. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J and Wu L: YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 7:126262016. View Article : Google Scholar : PubMed/NCBI

66 

Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, Liu C and He C: YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 27:315–328. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Xiao W, Adhikari S, Dahal U, Chen YS, Hao YJ, Sun BF, Sun HY, Li A, Ping XL, Lai WY, et al: Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 61:507–519. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Kasowitz SD, Ma J, Anderson SJ, Leu NA, Xu Y, Gregory BD, Schultz RM and Wang PJ: Nuclear m6A reader YTHDC1 regulates alternative polyadenylation and splicing during mouse oocyte development. PLoS Genet. 14. pp. e10074122018, View Article : Google Scholar

69 

Roundtree IA, Luo GZ, Zhang Z, Wang X, Zhou T, Cui Y, Sha J, Huang X, Guerrero L, Xie P, et al: YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. Elife. 6:e313112017. View Article : Google Scholar

70 

Lesbirel S, Viphakone N, Parker M, Parker J, Heath C, Sudbery I and Wilson SA: The m6A-methylase complex recruits TREX and regulates mRNA export. Sci Rep. 8:138272018. View Article : Google Scholar

71 

Brockdorff N, Bowness JS and Wei G: Progress toward understanding chromosome silencing by Xist RNA. Genes Dev. 34:733–744. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Liang D, Lin WJ, Ren M, Qiu J, Yang C, Wang X, Li N, Zeng T, Sun K, You L, et al: m6A reader YTHDC1 modulates autophagy by targeting SQSTM1 in diabetic skin. Autophagy. 18:1318–1337. 2022. View Article : Google Scholar

73 

Hsu PJ, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, Qi M, Lu Z, Shi H, Wang J, et al: Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 27:1115–1127. 2017. View Article : Google Scholar : PubMed/NCBI

74 

König J, Zarnack K, Rot G, Curk T, Kayikci M, Zupan B, Turner DJ, Luscombe NM and Ule J: iCLIP reveals the function of hnRNP particles in splicing at individual nucleotide resolution. Nat Struct Mol Biol. 17:909–915. 2010. View Article : Google Scholar : PubMed/NCBI

75 

McCloskey A, Taniguchi I, Shinmyozu K and Ohno M: hnRNP C tetramer measures RNA length to classify RNA polymerase II transcripts for export. Science. 335:1643–1646. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Liu N, Dai Q, Zheng G, He C, Parisien M and Pan T: N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 518:560–564. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Zhou KI, Shi H, Lyu R, Wylder AC, Matuszek Ż, Pan JN, He C, Parisien M and Pan T: Regulation of co-transcriptional pre-mRNA splicing by m6A through the low-complexity protein hnRNPG. Mol Cell. 76:70–81.e9. 2019. View Article : Google Scholar

78 

Liu N, Zhou KI, Parisien M, Dai Q, Diatchenko L and Pan T: N6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein. Nucleic Acids Res. 45:6051–6063. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S and Tavazoie SF: HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 162:1299–1308. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Fabbiano F, Corsi J, Gurrieri E, Trevisan C, Notarangelo M and D'Agostino VG: RNA packaging into extracellular vesicles: An orchestra of RNA-binding proteins? J Extracell Vesicles. 10:e120432020. View Article : Google Scholar

81 

Huang H, Weng H, Sun W, Qin X, Shi H, Wu H, Zhao BS, Mesquita A, Liu C, Yuan CL, et al: Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat Cell Biol. 20:285–295. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Wu Z, Shi Y, Lu M, Song M, Yu Z, Wang J, Wang S, Ren J, Yang YG, Liu GH, et al: METTL3 counteracts premature aging via m6A-dependent stabilization of MIS12 mRNA. Nucleic Acids Res. 48:11083–11096. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Edupuganti RR, Geiger S, Lindeboom RGH, Shi H, Hsu PJ, Lu Z, Wang SY, Baltissen MPA, Jansen PWTC, Rossa M, et al: N6-methyladenosine (m6A) recruits and repels proteins to regulate mRNA homeostasis. Nat Struct Mol Biol. 24:870–878. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Zhang J, Bai R, Li M, Ye H, Wu C, Wang C, Li S, Tan L, Mai D, Li G, et al: Excessive miR-25-3p maturation via N6-methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nat Commun. 10:18582019. View Article : Google Scholar :

85 

Hua YQ, Zhang K, Sheng J, Ning ZY, Li Y, Shi WD and Liu LM: NUCB1 suppresses growth and shows additive effects with gemcitabine in pancreatic ductal adenocarcinoma via the unfolded protein response. Front Cell Dev Biol. 9:6418362021. View Article : Google Scholar : PubMed/NCBI

86 

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

87 

Xie F, Liu S and Wang H: M6A methyltransferase METTL16 suppresses pancreatic cancer proliferation through p21 pathways. Pancreas. 49:14372020.

88 

Huang H, Li H, Pan R, Wang S, Khan AA, Zhao Y, Zhu H and Liu X: Ribosome 18S m6A methyltransferase METTL5 promotes pancreatic cancer progression by modulating c-Myc translation. Int J Oncol. 60:92022. View Article : Google Scholar

89 

Zeng J, Zhang H, Tan Y, Wang Z, Li Y and Yang X: m6A demethylase FTO suppresses pancreatic cancer tumorigenesis by demethylating PJA2 and inhibiting Wnt signaling. Mol Ther Nucleic Acids. 25:277–292. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Tang X, Liu S, Chen D, Zhao Z and Zhou J: The role of the fat mass and obesity-associated protein in the proliferation of pancreatic cancer cells. Oncol Lett. 17:2473–2478. 2019.PubMed/NCBI

91 

Guo X, Li K, Jiang W, Hu Y, Xiao W, Huang Y, Feng Y, Pan Q and Wan R: RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner. Mol Cancer. 19:912020. View Article : Google Scholar : PubMed/NCBI

92 

He Y, Hu H, Wang Y, Yuan H, Lu Z, Wu P, Liu D, Tian L, Yin J, Jiang K and Miao Y: ALKBH5 inhibits pancreatic cancer motility by decreasing long non-coding RNA KCNK15-AS1 methylation. Cell Physiol Biochem. 48:838–846. 2018. View Article : Google Scholar : PubMed/NCBI

93 

He Y, Yue H, Cheng Y, Ding Z, Xu Z, Lv C, Wang Z, Wang J, Yin C, Hao H and Chen C: ALKBH5-mediated m6A demethylation of KCNK15-AS1 inhibits pancreatic cancer progression via regulating KCNK15 and PTEN/AKT signaling. Cell Death Dis. 12:11212021. View Article : Google Scholar

94 

Tang B, Yang Y, Kang M, Wang Y, Wang Y, Bi Y, He S and Shimamoto F: m6A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol Cancer. 19:32020. View Article : Google Scholar

95 

Huang R, Yang L, Zhang Z, Liu X, Fei Y, Tong WM, Niu Y and Liang Z: RNA m6A demethylase ALKBH5 protects against pancreatic ductal adenocarcinoma via targeting regulators of iron metabolism. Front Cell Dev Biol. 9:7242822021. View Article : Google Scholar

96 

Hu X, Peng WX, Zhou H, Jiang J, Zhou X, Huang D, Mo YY and Yang L: IGF2BP2 regulates DANCR by serving as an N6-methyladenosine reader. Cell Death Differ. 27:1782–1794. 2020. View Article : Google Scholar :

97 

Meng X, Deng Y, He S, Niu L and Zhu H: m6A-mediated upregulation of LINC00857 promotes pancreatic cancer tumorigenesis by regulating the miR-150-5p/E2F3 axis. Front Oncol. 11:6299472021. View Article : Google Scholar

98 

Zhai S, Xu Z, Xie J, Zhang J, Wang X, Peng C, Li H, Chen H, Shen B and Deng X: Epigenetic silencing of LncRNA LINC00261 promotes c-myc-mediated aerobic glycolysis by regulating miR-222-3p/HIPK2/ERK axis and sequestering IGF2BP1. Oncogene. 40:277–291. 2021. View Article : Google Scholar :

99 

Geng Y, Guan R, Hong W, Huang B, Liu P, Guo X, Hu S, Yu M and Hou B: Identification of m6A-related genes and m6A RNA methylation regulators in pancreatic cancer and their association with survival. Ann Transl Med. 8:3872020. View Article : Google Scholar : PubMed/NCBI

100 

Ying P, Li Y, Yang N, Wang X, Wang H, He H, Li B, Peng X, Zou D, Zhu Y, et al: Identification of genetic variants in m6A modification genes associated with pancreatic cancer risk in the Chinese population. Arch Toxicol. 95:1117–1128. 2021. View Article : Google Scholar : PubMed/NCBI

101 

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

102 

Brabletz T, Kalluri R, Nieto MA and Weinberg RA: EMT in cancer. Nat Rev Cancer. 18:128–134. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Xia T, Wu X, Cao M, Zhang P, Shi G, Zhang J, Lu Z, Wu P, Cai B, Miao Y and Jiang K: The RNA m6A methyltransferase METTL3 promotes pancreatic cancer cell proliferation and invasion. Pathol Res Pract. 215:1526662019. View Article : Google Scholar : PubMed/NCBI

104 

Li BQ, Huang S, Shao QQ, Sun J, Zhou L, You L, Zhang TP, Liao Q, Guo JC and Zhao YP: WT1-associated protein is a novel prognostic factor in pancreatic ductal adenocarcinoma. Oncol Lett. 13:2531–2538. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Chen J, Sun Y, Xu X, Wang D, He J, Zhou H, Lu Y, Zeng J, Du F, Gong A and Xu M: YTH domain family 2 orchestrates epithelial-mesenchymal transition/proliferation dichotomy in pancreatic cancer cells. Cell Cycle. 16:2259–2271. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Dai S, Zhang J, Huang S, Lou B, Fang B, Ye T, Huang X, Chen B and Zhou M: HNRNPA2B1 regulates the epithelial-mesenchymal transition in pancreatic cancer cells through the ERK/snail signalling pathway. Cancer Cell Int. 17:122017. View Article : Google Scholar : PubMed/NCBI

107 

Dahlem C, Barghash A, Puchas P, Haybaeck J and Kessler SM: The insulin-like growth factor 2 mRNA binding protein IMP2/IGF2BP2 is overexpressed and correlates with poor survival in pancreatic cancer. Int J Mol Sci. 20:32042019. View Article : Google Scholar : PubMed/NCBI

108 

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

109 

Huang XT, Li JH, Zhu XX, Huang CS, Gao ZX, Xu QC, Zhao W and Yin XY: HNRNPC impedes m6A-dependent anti-metastatic alternative splicing events in pancreatic ductal adenocarcinoma. Cancer Lett. 518:196–206. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Hu Y, Tang J, Xu F, Chen J, Zeng Z, Han S, Wang F, Wang D, Huang M, Zhao Y, et al: A reciprocal feedback between N6-methyladenosine reader YTHDF3 and lncRNA DICER1-AS1 promotes glycolysis of pancreatic cancer through inhibiting maturation of miR-5586-5p. J Exp Clin Cancer Res. 41:692022. View Article : Google Scholar : PubMed/NCBI

111 

Huang S, Wu Z, Cheng Y, Wei W and Hao L: Insulin-like growth factor 2 mRNA binding protein 2 promotes aerobic glycolysis and cell proliferation in pancreatic ductal adenocarcinoma via stabilizing GLUT1 mRNA. Acta Biochim Biophys Sin (Shanghai). 51:743–752. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Taketo K, Konno M, Asai A, Koseki J, Toratani M, Satoh T, Doki Y, Mori M, Ishii H and Ogawa K: The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int J Oncol. 52:621–629. 2018.PubMed/NCBI

113 

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

114 

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

115 

Chen B, Ye F, Yu L, Jia G, Huang X, Zhang X, Peng S, Chen K, Wang M, Gong S, et al: Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J Am Chem Soc. 134:17963–17971. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, Wang Y, Li X, Xiong XF, Wei B, et al: RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Mol Cancer. 18:462019. View Article : Google Scholar : PubMed/NCBI

117 

Huang Y, Yan J, Li Q, Li J, Gong S, Zhou H, Gan J, Jiang H, Jia GF, Luo C and Yang CG: Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 43:373–384. 2015. View Article : Google Scholar :

118 

Cui Q, Shi H, Ye P, Li L, Qu Q, Sun G, Sun G, Lu Z, Huang Y, Yang CG, et al: m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Rep. 18:2622–2634. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Huang Y, Su R, Sheng Y, Dong L, Dong Z, Xu H, Ni T, Zhang ZS, Zhang T, Li C, et al: Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell. 35:677–691.e10. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, Deng X, Wang Y, Weng X, Hu C, et al: R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell. 172:90–105.e23. 2018. View Article : Google Scholar

121 

Xu P and Ge R: Roles and drug development of METTL3 (methyltransferase-like 3) in anti-tumor therapy. Eur J Med Chem. 230:1141182022. View Article : Google Scholar : PubMed/NCBI

122 

Forino NM, Hentschel J and Stone MD: Cryo-EM structures tell a tale of two telomerases. Nat Struct Mol Biol. 28:457–459. 2021. View Article : Google Scholar : PubMed/NCBI

123 

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. 593:597–601. 2021. View Article : Google Scholar : PubMed/NCBI

124 

Weng H, Huang H, Wu H, Qin X, Zhao BS, Dong L, Shi H, Skibbe J, Shen C, Hu C, et al: METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A modification. Cell Stem Cell. 22:191–205.e9. 2018. View Article : Google Scholar

125 

Zhang J, Tsoi H, Li X, Wang H, Gao J, Wang K, Go MY, Ng SC, Chan FK, Sung JJ and Yu J: Carbonic anhydrase IV inhibits colon cancer development by inhibiting the Wnt signalling pathway through targeting the WTAP-WT1-TBL1 axis. Gut. 65:1482–1493. 2016. View Article : Google Scholar

126 

Huang C, Zhou S, Zhang C, Jin Y, Xu G, Zhou L, Ding G, Pang T, Jia S and Cao L: ZC3H13-mediated N6-methyladenosine modification of PHF10 is impaired by fisetin which inhibits the DNA damage response in pancreatic cancer. Cancer Lett. 530:16–28. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2023
Volume 62 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ye T, Wang J, Zhao H, Zhao G and Li P: Role of N<sup>6</sup>‑methyladenosine in the pathogenesis, diagnosis and treatment of pancreatic cancer (Review). Int J Oncol 62: 4, 2023
APA
Ye, T., Wang, J., Zhao, H., Zhao, G., & Li, P. (2023). Role of N<sup>6</sup>‑methyladenosine in the pathogenesis, diagnosis and treatment of pancreatic cancer (Review). International Journal of Oncology, 62, 4. https://doi.org/10.3892/ijo.2022.5452
MLA
Ye, T., Wang, J., Zhao, H., Zhao, G., Li, P."Role of N<sup>6</sup>‑methyladenosine in the pathogenesis, diagnosis and treatment of pancreatic cancer (Review)". International Journal of Oncology 62.1 (2023): 4.
Chicago
Ye, T., Wang, J., Zhao, H., Zhao, G., Li, P."Role of N<sup>6</sup>‑methyladenosine in the pathogenesis, diagnosis and treatment of pancreatic cancer (Review)". International Journal of Oncology 62, no. 1 (2023): 4. https://doi.org/10.3892/ijo.2022.5452