Open Access

m6A modification of non‑coding RNA: Mechanisms, functions and potential values in human diseases (Review)

  • Authors:
    • Qian Yi
    • Yi Liao
    • Wei Sun
    • Jiachen Li
    • Dahang Yang
    • Hongxi Shang
    • Weichao Sun
  • View Affiliations

  • Published online on: August 5, 2025     https://doi.org/10.3892/ijmm.2025.5605
  • Article Number: 164
  • Copyright: © Yi 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 modification represents a pivotal and novel post‑transcriptional modification in eukaryotic RNAs. Initially identified in messenger RNAs (mRNAs), m6A modification on these transcripts regulates a spectrum of essential cellular processes, including mRNA splicing, subcellular localization, stability and translation. Recent studies have highlighted the involvement of m6A methylation in both biological and pathological processes, particularly in cancer. Non‑coding RNAs (ncRNAs), a diverse class of RNA molecules that do not encode proteins, encompass microRNAs, long ncRNAs and circular RNAs. Notably, m6A has been recognized as a reversible epigenetic modification within ncRNAs, a discovery that has garnered considerable attention. This modification not only influences the stability of ncRNAs but also endows them with the novel capacity for peptide translation. The differential and specific expression pattern in diseases give these m6A‑modified ncRNAs potential as biomarkers for molecular diagnostics and targeted therapy, and using ncRNA‑encoded peptides as a target for immunotherapy has also been attempted. This review synthesizes the current understanding of m6A modifications in ncRNAs, explores the effects of m6A on ncRNA function and presents the latest insights into the role of ncRNA m6A modifications in disease progression. 

Introduction

RNA methylation is a key epigenetic modification that regulates gene transcription without altering the underlying gene sequence. Key types of RNA methylation include N6-methyladenosine (m6A), 5-methylcytosine, m1A, N7-methylguanosine and 3-methylcytidine (1,2). Among these, m6A is the most prevalent and extensively studied RNA modification. As the name suggests, m6A occurs at the N6 position of adenosine, with two predominant motifs-RRACH and DRACH-where H represents U, A or C; R denotes A or G; and D refers to U, A or G (3). This modification plays a pivotal role in various aspects of RNA metabolism, including splicing, transport, localization, translation and degradation (4,5). Numerous studies have demonstrated that m6A is critical for physiological processes, such as embryonic development (6), and it has also been implicated in various pathological processes, including cancer (7,8).

In eukaryotic cells, non-coding RNAs (ncRNAs) are a class of endogenous RNA molecules that do not encode proteins (9). These ncRNAs can be categorized into several types, including transfer RNAs, microRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs) (10,11). Although ncRNAs do not directly translate into proteins, they perform vital biological functions at the RNA level. They can interact with DNA, mRNA and proteins, playing specialized roles in the regulation of gene expression, mRNA stability and protein function (12-14). Dysregulation of ncRNAs has been strongly linked to the progression of various diseases and malignancies.

Recent studies have expanded the understanding of m6A modifications in ncRNAs (15,16). m6A methylation regulates the transcription, structural stability, localization and translation of ncRNAs. Importantly, the abnormal changes induced by m6A modification enable ncRNAs to participate in diverse biological and pathological processes, including cancer. For instance, peptides derived from m6A-modified ncRNAs have been reported to exhibit both oncogenic and tumor-suppressive functions (17,18). Therefore, the roles of m6A modification in ncRNAs and their underlying mechanisms warrant further investigation, with the potential to uncover novel biomarkers for disease diagnosis, prognostic indicators and therapeutic strategies.

The present review synthesizes recent findings on m6A modifications in ncRNAs, focusing on their impact on the stability, localization and translation of ncRNAs. Additionally, it discusses the involvement of m6A-modified ncRNAs in disease progression and tumorigenesis, providing new insights into the role of m6A modification in ncRNA biology and facilitating further research in this area.

m6A modification

After transcription, RNA nucleosides, including adenosine, guanosine, cytidine and uridine, can undergo additional chemical modifications, adding complexity to their function. One such modification is m6A, where a methyl group is added to the N6 position of adenosine. Since its discovery in 1974, m6A has been recognized as a prevalent and abundant modification in mRNAs, and it remains one of the most extensively studied RNA modifications to date (19,20). Extensive research has demonstrated that m6A modifications play a pivotal role in various physiological processes and are implicated in numerous diseases, including obesity, diabetes, gastrointestinal disorders, Alzheimer's disease (AD) and cancers (21,22).

m6A methylation is a dynamic and reversible post-transcriptional modification that sets it apart from other forms of epigenetic regulation (Fig. 1). This reversible process is mediated by enzymes and proteins involved in the m6A modification, including 'writers' and 'erasers' (4). The m6A writers, or methyltransferases, include methyltransferase-like 3 (METTL3), METTL5, METTL14, METTL16, Wilms' tumor 1-associated protein (WTAP), RNA binding motif protein 15/15B (RBM15/15B), zinc finger CCCH-type containing 13 and KIAA1429 (also known as vir-like m6A methyltransferase associated protein). These enzymes form a methyltransferase complex that catalyzes the addition of m6A to RNA molecules (23-25). Conversely, m6A erasers, or demethylases, such as fat mass and obesity-associated protein (FTO) and α-ketoglutarate-dependent dioxygenase alk B homolog 5 (ALKBH5), are responsible for removing m6A modifications from target RNA molecules (2,26,27). The m6A readers are the key enzymes, as they recognize the m6A sites on RNA molecules and mediate the influence of m6A on the fate of mRNA. The YTH domain family (YTHDF) represents the most common group of m6A readers. YTHDF1 enhances mRNA translation, while YTHDF2 promotes mRNA degradation, and YTHDF3 works in concert with YTHDF1 and YTHDF2 to regulate mRNA metabolism. YTHDC1 and YTHDC2, other key m6A readers, are involved in RNA splicing, cytoplasmic-nuclear shuttling and stability (28-30). The insulin-like growth factor 2 mRNA-binding protein (IGF2BP) family, comprising IGF2BP1, IGF2BP2 and IGF2BP3, protects m6A-modified mRNAs from degradation within P-bodies and stress granules (31-33). Other m6A readers, such as heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) and serine and arginine rich splicing factor 9, have been shown to regulate mRNA stability, localization and translation (34,35). While the majority of studies on m6A focus on mRNA molecules, emerging evidence indicates that ncRNAs can also undergo m6A modification, which regulates their stability, localization and translation.

The m6A modification can be detected along with the development of detection technologies. Single-molecule epitranscriptomic analysis and single-molecule sequencing could detect and reveal functional roles of site-specific m6As (36,37). However, high-throughput sequencing or in vitro assays are not reflected in the dynamic changes of m6A modification, and current in situ imaging techniques for site-specific m6A are constrained. Recently, Song et al (38) presented a method, termed proximity hybridization followed by primer exchange amplification, which could image m6A methylation sites concurrently in multiple cell types, revealing cell-to-cell variability in expression levels. Furthermore, Zhang et al (39) introduced the TadA8.20-assisted N6-methyladenosine RNA imaging at single-base resolution method for precise visualization and quantification of both A and m6A forms at specific RNA sites within single cells. With the development of detection technology, detecting dynamic changes in m6A modification can provide a more accurate basis for molecular diagnosis and targeted therapy of diseases.

NcRNAs

NcRNAs are a diverse group of endogenous RNA molecules that typically do not encode proteins (10). These ncRNAs can be categorized into several classes: LncRNAs, which are longer than 200 nucleotides (nt); small ncRNAs, including miRNAs and small nuclear RNAs, which are shorter than 200 nt; circRNAs; small interfering RNAs; piwi-interacting RNAs; and ribosomal RNAs.

MiRNAs are a well-conserved class of endogenous ncRNAs ~22 nucleotides in length (40). They interact with mRNAs, regulating their degradation, translational repression or direct cleavage in a post-transcriptional manner (41,42). >2,600 miRNAs have been identified in the human genome and are known to regulate the expression of 30-50% of functional genes. Small nuclear RNAs, averaging 150 nucleotides, are primarily located in the nucleus and are core components of small nuclear ribonucleoproteins, which are essential for RNA splicing (43,44).

Like typical RNA molecules, lncRNAs possess a 5′-methyl-cytosine cap and a 3′-poly(A) tail (45). In the canonical pathway, lncRNAs are transcribed by RNA polymerase II. They may be cleaved by ribonuclease P or recognized by small nucleolar RNA-protein complexes and other enzymes to produce mature 3′ ends, capping structures or circular forms in non-canonical pathways (46,47). Based on their genomic origins, lncRNAs can be classified into five categories: Sense lncRNAs, antisense lncRNAs, bidirectional lncRNAs, intronic lncRNAs and intergenic lncRNAs (48). Initially, lncRNAs were thought to be non-coding and to primarily act as RNA sponges or sponges for RNA-binding proteins (RBPs) (49,50).

CircRNAs are a distinct class of ncRNA characterized by a covalently closed loop structure (51). They are generated through back-splicing, where a downstream splice donor site is joined with an upstream splice acceptor site (52). CircRNAs can be further classified into three major groups based on their genomic origins: Circular intronic RNAs, exon-intron circRNAs and exonic circRNAs (53). CircRNAs have been shown to regulate transcription, RNA stability, localization and protein function by binding to DNA, RNA molecules or proteins (54,55).

The dysregulation of miRNAs, lncRNAs and circRNAs has been implicated in various biological processes, primarily through their ability to sponge RNA molecules or proteins (56,57). Recently, it has been discovered that lncRNAs and circRNAs containing internal ribosome entry sites or m6A modification sites can be translated into micro-peptides (58,59). These novel findings provide critical insights for further research.

m6A modification of ncRNAs

m6A modification of miRNAs

In the case of miRNAs, m6A modification has been shown to impact their biogenesis and subsequently alter the functionality of mature miRNAs. Garbo et al (16) demonstrated that m6A modification on specific miRNAs affects argonaute 2 (AGO2)/miRNA and RBP/miRNA interactions, impairing their ability to regulate target mRNAs and influencing extracellular vesicle (EV) loading. Alarcón et al (60) highlighted m6A methylation as a critical mechanism in miRNA biogenesis. METTL3-mediated m6A methylation promotes the maturation of pri-miR-BART3-3p by interacting with DGCR8, thereby facilitating Natural killer/T cell lymphoma progression (25). Similarly, METTL14 modulates pri-miR-100 processing to mature miR-100-3p in an m6A-dependent manner through DGCR8 microprocessor complex subunit (DGCR8) during ultraviolet B-induced human dermal fibroblast photoaging (61). In addition, ALKBH5 demethylates pri-miR-194-2, suppressing miR-194-2 biogenesis via an m6A/DGCR8-dependent pathway (62), while FTO inhibits DGCR8 binding to pri-miR-138-5p through m6A modification, thereby limiting miR-138-5p processing (63). Furthermore, YTHDF2 recognizes m6A sites in pre-miR-126 and recruits AGO2 to enhance the maturation of pre-miR-126 into mature miR-126 (51). YTHDC1 promotes the biogenesis of mature miR-30d through m6A-mediated regulation of mRNA stability (64).

m6A modification of lncRNAs

Numerous studies have reported the m6A modification of lncRNAs. Bian et al (65) found that METTL3 mediates the m6A modification of lncRNA ABHD11-antisense 1 (AS1), which inhibits ferroptosis and promotes colorectal cancer (CRC) progression. Likewise, Zhao et al (66) identified METTL3 as a key factor in enhancing the m6A modification of homeobox (HOX)A10-AS, thereby increasing its RNA stability. METTL14-mediated m6A modification stabilizes lncRNA THRIL (TNF and HNRNPL related immunoregulatory lncRNA), accelerating lipopolysaccharide (LPS)-induced acute injury in alveolar epithelial cells (67). Furthermore, WTAP-mediated m6A modification enhances the stability of lnc-OXAR, which contributes to oxaliplatin resistance in non-alcoholic steatohepatitis-related hepatocellular carcinoma (HCC) (68). Additionally, FTO has been shown to suppress the m6A modification of lncRNA small nucleolar RNA host gene 1 (SNHG14), attenuating LPS-induced acute kidney injury by inhibiting autophagy (69). ALKBH5 decreases m6A-modified sites on DNA damage inducible transcript 4 (DDIT4)-AS1, inhibiting the recruitment of ELAV-like RNA binding protein 1 and stabilizing DDIT4-AS1 (70).

m6A modification of circRNAs

CircRNAs are also subject to m6A modification. Chen et al (71) demonstrated that METTL3 promotes the m6A modification of circ-CTTN, thereby enhancing osteogenic differentiation of human umbilical cord mesenchymal stem cells (MSCs). Fan et al (72) uncovered that METTL14 mediates m6A modification of circORC5, which suppresses gastric cancer (GC) progression. Additionally, WTAP-mediated m6A modification of circ_0056856 promotes proliferation, migration and invasion of interleukin-22-stimulated human keratinocytes (73). ALKBH5 has also been reported to regulate the demethylation of circCPSF6, influencing its recognition and stabilization by YTHDF2 (74). The ALKBH5/insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) axis mediates the m6A modification of circXPO1, which accelerates CRC progression (75). Furthermore, Wu et al (76) found that FTO binds to circFAM192A at specific sites, removing the m6A modification and protecting it from degradation.

Function of m6A modification on ncRNAs

For mRNAs, the roles of m6A modification are well-established, encompassing various aspects of RNA metabolism, including splicing, transportation, translation and degradation. For ncRNAs, m6A modification plays a critical role in regulating their biogenesis, stability, localization and even translation (Fig. 2).

Regulating the biogenesis and expression of ncRNAs

As widely recognized, m6A modification impacts the biogenesis and functionality of mature miRNAs. In addition, m6A modulates RNA splicing, which is essential for the biogenesis of circRNAs. For instance, circDDIT4 is generated by back-splicing at the 3′-UTR using a 5′ splice acceptor site in exon 2 of linear DDIT4 mRNA. The WTAP/METTL3/METTL14 methyltransferase complex mediates m6A modification in both the circDDIT4-5′ flanking and circDDIT4-3′-UTR regions, promoting circDDIT4 circularization (77). METTL3/YTHDC1-mediated m6A modification regulates the biogenesis of circRBM33, generated from exon 3 to exon 5 of RBM33 mRNA (78). Similarly, METTL3/YTHDC1-mediated m6A modification and back-splicing events contribute to the biogenesis of circCDYL (79). METTL3 increases m6A modification of circARL3, with YTHDC1 binding to modified sites and facilitating reverse splicing and circularization (80). METTL3 also mediates the m6A modification of circIGF2BP3, promoting its circularization in an m6A-dependent manner through YTHDC1 (81). The m6A reader YTHDC1, along with the RNA helicase DEAD-box helicase 5 (DDX5), regulates the production of circRNAs enriched in rhabdomyosarcoma (82). Furthermore, the upregulation of lncRNA CHASERR in response to m6A modification is facilitated by METTL3/YTHDF1-mediated RNA transcripts (83). Silencing METTL14 suppresses DHRS4-AS1 expression by reducing the m6A modification of DHRS4-AS1 transcripts (84). Additionally, Chen et al (85) reported that suppressing METTL3 inhibits the expression of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). ALKBH5 promotes the upregulation of lncRMRP expression through demethylation (86) and enhances the expression of the lncRNA DIO3OS via m6A modification (87).

Regulating the stability of ncRNAs

m6A modification has been demonstrated to play a pivotal role in regulating the stability of lncRNAs and circRNAs. For instance, m6A modification is significantly enriched in lncRNA RMRP, enhancing its RNA stability (88). In addition, METTL14-induced m6A modification stabilizes lncRNA-PLCB1, thereby inhibiting GC progression through the destabilization of DDX21 (89). Xie et al (90) demonstrated that IGF2BP3 binds to m6A-modified sites on lncRNA OIP5-AS1, stabilizing its expression. Similarly, Luo et al (91) highlighted the critical role of m6A modification in maintaining the stability of lncRNA FAM83H-AS1, facilitated by METTL3 and the readers IGF2BP2/IGF2BP3. Furthermore, ALKBH5 demethylates lncRNA-CARMN, reducing its m6A modification, while YTHDF2/YTHDF3 recognizes and degrades m6A-modified lncRNA-CARMN (92). The demethylation of lncRNA SNHG15 by ALKBH5 enhances its stability (93). The abundance of m6A modification sites on lncRNA TP53TG1 is countered by ALKBH5, which reduces its stability and downregulates its expression (94). hnRNPA2B1 interacts with and stabilizes lncRNA NEAT1 in an m6A-dependent manner (95). Yi et al (96) identified the m6A modification of circPSMA7, noting that IGF2BP3 recognizes these sites and stabilizes circPSMA7, thereby enhancing its expression. Furthermore, the upregulation of circ_104797 in cisplatin-resistant bladder cancer is attributed to increased stability driven by elevated m6A levels in its sequence (97). Similarly, the increased stability of circPLPP4 in cisplatin-resistant ovarian cancer is mediated by heightened m6A modification (98). The elevated level of circ_0000337 in bortezomib-resistant multiple myeloma cells results from augmented m6A levels, leading to enhanced RNA stability (99). METTL3-mediated m6A methylation of circSLCO1B3 stabilizes its expression, with m6A-modified circSLCO1B3 promoting intrahepatic cholangiocarcinoma progression via regulation of HOXC8 and programmed cell death ligand 1 (100). Finally, FTO acts as an eraser, increasing the stability and expression of circBRCA1 by demethylating its m6A modification, thereby alleviating oxidative stress-induced granulosa cell damage (101).

Table I summarizes additional studies on the role of m6A modification in regulating ncRNA stability and the functional implications of these ncRNAs in various diseases (102-142).

Table I

Effects of m6A modification on ncRNA stability.

Table I

Effects of m6A modification on ncRNA stability.

ncRNAsm6A regulatorStabilityFunction(Refs.)
LncRNA PVT1ALKBH5DecreasedRegulates OC growth and lung cancer metastasis(102,103)
LncRNA MEG3HNRNPA2B1DecreasedFacilitates tumorigenesis of NSCLC(104)
LncRNA NEAT1ALKBH5, METTL3EnhancedFacilitates immunosuppression and cerebral I/R injury(105,106)
LncRNA MALAT1METTL3/HuR, METTL3, IGF2BP2EnhancedPromotes the malignant progression of glioma, OS and NSCLC(107-109)
LncRNA ZFAS1IGF2BP2, METTL3EnhancedPromotes mitochondrial energy metabolism and affects autophagy and progression of NPC(110,111)
LncRNA DANCRMETTL3, IGF2BP2EnhancedContributes to OS, pancreatic cancer and acute myeloid leukemia progression(112-114)
LINC00958METTL3, KIAA1429EnhancedPromotes BC tumorigenesis, HCC lipogenesis and progression, and accelerates GC aerobic glycolysis(115-117)
KCNQ1OT1IGF2BP1, MELLL3, ALKHB5EnhancedPromotes cardiomyocyte ferroptosis, doxorubicin resistance in BC and the development of LSCC(118-120)
FOXD2-AS1METTL3, WTAPEnhancedAccelerates cervical cancer and OS progression(121,122)
LncRNA PCAT6METTL3/IGF2BP2EnhancedPromotes bone metastasis in prostate cancer(123)
THAP7-AS1METTL3/IGF2BP1EnhancedPromotes GC growth, invasion and metastasis(124)
LncRNA PTTG3PMETTL3/IGF2BP2EnhancedContributes to CRC proliferation(125)
DLGAP1-AS1WTAPEnhancedEnhances the resistance of BC to adriamycin(126)
LncRNA CASC9IGF2BP2EnhancedAccelerates glioblastoma aerobic glycolysis(127)
ABHD11-AS1METTL3EnhancedEnhances the Warburg effect of NSCLC(128)
CircCUX1METTL3EnhancedConfers radioresistance of hypopharyngeal squamous cell carcinoma(129)
CircARHGAP12IGF2BP2EnhancedPromotes cervical cancer progression(130)
CircGLIS3METTL3EnhancedPromotes cell proliferation and invasion.(131)
CircDLC1METTL3EnhancedSuppresses the proliferation of glioma(132)
CircRPS6KC1METTL3/YTHDF1EnhancedRegulate cellular senescence(133)
CircASXL1METTL3/IGF2BP1EnhancedPromotes proliferation and migration of OC(134)
Circ_0006168METTL3/IGF2BP2EnhancedPromotes EMT process in ESCC(135)
CircPAPPA2 METTL14/IGF2BP3DecreasedRegulates the pathogenesis of preeclampsia(136)
CircCCDC134ALKBH5DecreasedFacilitates cervical cancer metastasis(137)
CircHIPK2YTHDF2DecreasedAlleviates microglia activation(138)
CircAFF2ALKBH5/YTHDF2DecreasedEnhances radiosensitivity of colorectal cancer(139)
CircMPP1YTHDC1DecreasedMaintains trophoblasts function(140)
Circ_0003979RBM15/YTHDF2DecreasedInhibits HCC tumorigenesis(141)
CircIRF2YTHDF2DecreasedSuppresses liver fibrosis(142)

[i] OS, osteosarcoma; OC, ovarian cancer; NSCLS, non-small cell lung cancer; NPC, nasopharyngeal carcinoma; BC, breast cancer; HCC, hepatocellular carcinoma; GC, gastric cancer; LSCC, laryngeal squamous cell carcinoma; CRC, colorectal cancer; ALKBH5, α-ketoglutarate-dependent dioxygenase alk B homolog 5; HNRNPA2B1, heterogeneous nuclear ribonucleoprotein A2/B1; METTL3/14, methyltransferase 3/14; HuR, also known as Elavl1, ELAV like RNA binding protein 1; IGF2BP1/2/3, insulin-like growth factor 2 mRNA binding protein 1/2/3; KIAA1429, also known as vir-like m6A methyltransferase associated protein; WTAP, Wilms' tumor 1-associated protein; YTHDF1/2, YTH N6-methyladenosine RNA binding protein F1/2, YTHDC1, YTH N6-methyladenosine RNA binding protein C1; RBM15, RNA binding motif protein 15.

Regulating the localization of ncRNAs

m6A modification also plays a pivotal role in the localization of ncRNAs, influencing their shuttling between the cytoplasm and nucleus. The m6A modification of circNSUN2 facilitates its export to the cytoplasm, enhancing the stability of HMGA2 mRNA and promoting CRC metastasis (143). Similarly, circPAK2 undergoes m6A modification, which is recognized by YTHDC1, enabling its nuclear export to the cytoplasm and subsequently promoting lymph node metastasis in GC (144). In parallel, YTHDC1 directly binds to the m6A sites on various circRNAs, including hsa_ circ_0102678 (145), circFNDC3B (146), circPOLR2B (147), circRNA3634 (148) and circHPS5 (149), promoting their export to the cytoplasm. Furthermore, METTL3 mediates the m6A modification of LINC00294, with YTHDC1 recognizing the m6A sites and promoting its cytoplasmic localization (150). Similarly, YTHDF1 interacts with the m6A site of lncRNA FOXD1-AS1, facilitated by METTL3, thereby promoting its cytoplasmic localization (151). In addition, hnRNPC mediates the cytoplasmic export of m6A-modified circMARK2 (152). METTL3-induced m6A modification also facilitates the export of circTEAD1 to the cytoplasm (153).

Beyond localization, m6A modification of ncRNAs also regulates nuclear retention or translocation, impacting gene transcription. For instance, circPPAP2B harbors m6A sites that recruit hnRNPC in an m6A-dependent manner, facilitating the nuclear translocation of hnRNPC, which subsequently regulates alternative splicing of pre-mRNA (154). The m6A modification of circCCDC134 by ALKBH5/YTHDF2 recruits p65 in the nucleus, ultimately stimulating HIF1A transcription and facilitating cancer cell growth and metastasis (137). In addition, circMMP9 is stabilized by IGF2BP2 in an m6A-dependent manner, with circMMP9 recruiting ETS1 to promote tripartite motif containing 59 (TRIM59) transcription (155). Furthermore, DNA damage has been shown to increase m6A levels on NEAT1, promoting structural alterations and the accumulation of hypermethylated NEAT1 at promoter-associated double-strand breaks (156). METTL3-induced m6A modification upregulates POU6F2-AS1, tethering Y-box binding protein 1 to the fatty acid synthase promoter and activating transcription (157). Allele-specific m6Ad methylation affects YTHDC1-mediated protein binding affinity, with the LOC339803-YTHDC1 interaction determining chromatin localization of LOC339803, inducing the expression of NF-κB-mediated proinflammatory cytokines (158). Additionally, Vaid et al (159) reported that m6A-mediated recruitment of hnRNPA2B1 to lncRNA-TERRA is essential for R-loop formation and telomere localization.

m6A modification plays a pivotal role in sorting ncRNAs into exosomes through interactions with m6A-binding proteins. For instance, the m6A reader hnRNPA2B1 contributes to the progression of multiple myeloma osteolytic bone disease by modulating the expression and exosomal transport of miRNAs to recipient monocytes or MSCs (160). Wei et al (79) demonstrated that m6A modification of circCDYL promotes its active sorting into exosomes via hnRNPA2/B1. Similarly, the m6A modification of circCCAR1, mediated by WTAP, facilitates its secretion by HCC cells into exosomes in a hnRNPA2B1-dependent manner (161). Exosomal encapsulation of circHIF1α is also governed by hnRNPA2B1 (162). Furthermore, multiple myeloma cells enhance the packaging of lncRNA into adipocyte-derived exosomes through METTL7A-mediated m6A methylation (163). Additionally, m6A modification drives the sorting of LINC00657 into exosomes, promoting breast cancer progression by inducing macrophage M2 polarization (164). He et al (165) reported that m6A modification, recognized by IGF2BP2, stabilizes TRPM2-AS and enhances its exosomal sorting. Other m6A readers, such as RNA binding motif protein X-linked (RBMX), may also play significant roles in exosome cargo loading (166), with RBMX being a newly identified m6A reader involved in this process (167). Table II summarizes other studies on the role of m6A modification in regulating ncRNA localization (168-179).

Table II

Effects of m6A modification on ncRNA localization.

Table II

Effects of m6A modification on ncRNA localization.

ncRNAsm6A regulatorLocalization(Refs.)
CircTET2YTHDC1Transport of circTET2 out of the nucleus(168)
CircRNA388YTHDC1/CRM1Facilitates m6A-modified circRNA388 nuclear export(169)
CircCSDE1METTL3Enhances its cyclization efficiency and nuclear export(170)
hsa_circ_0058493YTHDC1Promotes its localization from the nucleus to the cytoplasm(171)
CircMETYTHDC1Delivery to cytosol(172)
hsa_circ_0001599hnRNPA2B1Translocation from the nucleus to the cytoplasm(173)
CircEHD2hnRNPA2B1Regulates the package of circEHD2 into EVs(174)
CircTLCD4-RWDD3hnRNPA2B1Facilitates the sorting into NSCLC cell-derived EVs(175)
CircATP9AhnRNPA2B1Mediates the incorporation of circATP9A into EVs(176)
CircNEIL3hnRNPA2B1Packaging into exosomes and transmission to infiltrated TAMs(177)
Lnc668YTHDC1Promotion of the nuclear export of m6A-modified lnc668(178)
LncRNA TUC338METTL3/YTHDF1Delivery to EVs(179)

[i] EV, extracellular vesicle; YTHDC1, YTH N6-methyladenosine RNA binding protein C1; METTL3, methyltransferase 3; TAMs, tumor-associated macrophages; HNRNPA2B1, heterogeneous nuclear ribonucleoprotein A2/B1; NSCLC, non-small cell lung cancer; YTHDF1, YTH N6-methyladenosine RNA binding protein F1.

m6A-dependent peptides translation of ncRNAs

Traditionally, ncRNAs were considered non-coding and incapable of serving as a template for producing proteins or peptides. However, recent research has challenged this notion, revealing that ncRNAs can indeed be translated into micro-peptides in an m6A modification-dependent manner. It has been reported that circSLC9A6 encodes a novel peptide, SLC9A6-126 amino-acid (aa), through m6A modification, involving the m6A reader YTHDF2 (180). Similarly, circYAP encodes a truncated protein, YAP-220aa, through m6A modification, facilitated by YTHDF3 and the eIF4G2 translation initiation complex (181). YTHDF1 and YTHDF3 bind to m6A sites on circYthdc2, promoting its translation into the Ythdc2-170aa peptide (182). Li et al (183) discovered that circFBXW7 can be translated into the short polypeptide circFBXW7-185aa, involving the m6A reader YTHDF3. In a related study, circ-MIB2 harbors m6A sites that recruit YTHDF1 and YTHDF3, facilitating its translation into the MIB2-134aa (184). Tang et al (185) demonstrated that several male germ cell circRNAs contain large open reading frames with m6A-modified start codons in their junctions, a characteristic recently associated with protein-coding potential. Additionally, circFNDC3B, modified by m6A, is translocated to the cytoplasm, and Pan et al (186) found that circFNDC3B encodes a novel protein, circF-NDC3B-218aa. METTL3-mediated m6A-modified circGLIS3 contributes to β-cell dysfunction by encoding the protein Glis3-348aa (187). The stability of circ-ZNF609 is regulated by m6A methylation and circ-ZNF609 plays a pivotal role in fibroblast activation through peptide encoding (188).

The m6A modification also plays a critical role in the translation of lncRNAs. Wu et al (189) identified a novel micro-peptide, YY1BM, encoded by the Y-linked LINC00278, where reduced m6A modification led to decreased translation. Additionally, the translation of AFAP1-AS1 translated mitochondrial-localized peptide (ATMLP) peptides from lncRNA AFAP1-AS1 was found to be regulated by m6A methylation at the adenine locus 1,313 of AFAP1-AS1 (190). Furthermore, METTL3 catalyzed the installation of m6A modification, enhancing the stability of the METTL4-2 transcript and thereby increasing its expression. In parallel, YTHDF1 recognized these m6A sites, facilitating the translation of METTL4-2 (191). Moreover, m6A modification regulates the expression of various lncRNAs, some of which have been reported to encode micro-peptides. For instance, YTHDC1 was found to upregulate the expression of HOXB-AS3 through m6A modification of its precursor RNA (192), and a micro-peptide encoded by HOXB-AS3 was reported to promote the proliferation and viability of oral squamous cell carcinoma cells (193). Another novel peptide, HOXB-AS3-32aa encoded by lncRNA HOXB-AS3, was found to promote cigarette smoke-induced inflammation and apoptosis (194). Additionally, the micro-peptide ATP synthase-associated peptide encoded by LINC00467 has been shown to promote CRC progression (195), although Zhang et al (196) recently reported the negative impact of m6A methylation on LINC00467 translation. The development of m6A sequencing technologies has led to the identification of numerous lncRNAs undergoing m6A modification, suggesting the potential for a significant number of lncRNA-encoded proteins. Table III summarizes other studies on m6A-related circRNA and lncRNA translation (197-211).

Table III

m6A-related circRNA and lncRNA translation.

Table III

m6A-related circRNA and lncRNA translation.

ncRNAsPeptide nameDiseaseFunctions and mechanisms(Refs.)
Circ-METMET404GBMOncogenic; directly interacts with the MET β subunit and forms a constitutively activated MET receptor(197)
CircASK1ASK1-272aaLung cancerOncogenic; ameliorates gefitinib resistance in lung adenocarcinoma(198)
CircSTX6CircSTX6-144aaHCCOncogenic; promotes HCC proliferation, migration and invasion(199)
CircARHGAP35P190-AHCCOncogenic; circARHGAP35 protein interacts with TFII-I in the nucleus(200)
CircPETHCircPETH-147aaHCCRegulates metabolic reprogramming and remodels the immunosuppressive microenvironment(201)
CircNFIBCircNFIB-56aaBreast tumorTumor suppressor; decreases synthesis of arachidonic acid and inhibits breast tumor growth and metastasis(202)
CircSPECC1SPECC1-415aaGBMTumor suppressor; restores the sensitivity of TMZ-resistant GBM cells to TMZ.(203)
CircKEAP1KEAP1-259aaOSTumor suppressor; reduces cell proliferation, invasion and tumorsphere formation of OS cells(204)
CircCDYLtCDYL-100aaCardiac hypertrophysExacerbates cardiac hypertrophy(205)
LINC00339 LINC00339-205-49aaEndometrial receptivityPromotes trophoblast adhesion to endometrial cells(206,207)
MALAT1-NeuronsModulates synaptic function(208,209)
lncRNA BVES-AS1 BVES-AS1-201-50aaCRCPromotes cell viability, migration and invasion in colorectal cancer(210,211)

[i] TMZ, temozolomide; MET, MET proto-oncogene, receptor tyrosine kinase; HCC, hepatocellular carcinoma; TFII-I (also known as GTF2I), general transcription factor IIi; TMZ, temozolomide; OS, osteosarcoma; CRC, colorectal cancer.

m6A modification of ncRNAs in other diseases

m6A modification of ncRNAs in cardiovascular disorders

Atherosclerosis (AS), myocardial infarction injury, myocardial ischemia-reperfusion injury and cardiac hypertrophy are the most common cardiovascular diseases. It has been reported that m6A modification of ncRNAs participates in the pathological processes of cardiovascular diseases (Fig. 3). METTL14-mediated m6A modification upregulated circARHGAP12 and aspartate beta-hydroxylase to aggravate overload-induced lipid peroxidative damage and facilitate AS progression (212). Shen et al (213) reported that hypoxia triggers cardiomyocyte apoptosis via regulating the m6A methylation-mediated lncMIAT/miR-708-5p/p53 axis. ALKBH5 decreased the N6-methylation and promoted the destabilization of circPan3, and circPan3 could attenuate cardiomyocyte hypertrophy by targeting the miR-320-3p/HSP20 axis (214). METTL3 mediated the m6A modification of lncRNA H19 to alleviate cerebral ischemia-reperfusion injury by regulating the sphingosine-1-phosphate receptor 2/Toll-like receptor 4/NLR family pyrin domain containing 3 (NLRP3) signaling pathway (215). METTL14 mediated m6A-modified circZNF609, which regulated doxorubicin-induced cardiotoxicity by upregulating FTO (216).

m6A modification of ncRNAs in metabolic diseases

Diabetes-related diseases, obesity and nonalcoholic fatty liver disease are the most common metabolic diseases (Fig. 4). It has been reported that YTHDC2 mediated the m6A modification of circYTHDC2, which promoted dysfunction of vascular SMCs (VSMCs) and is an important target of metformin in preventing the progression of VSMC dysfunction in type 2 diabetes (217). METTL3 ameliorated diabetes-induced testicular damage by upregulating lncRNA TUG1/clusterin signaling (218). Diabetic retinopathy (DR) and diabetic nephropathy are common complications of diabetes. Fu et al (219) found that METTL3/YTHDC1 mediates the m6A modification and upregulation of lncRNA OGRU, which led to oxidative stress, inflammation and DR progression. Macrophage M1 regulatory DR is mediated by FTO-regulated m6A modification of LINC00342, LINC00667 and LNC00963 expression (220). WTAP mediated the m6A methylation of NEAT1, and subsequently NLRP3 inflammasome activation and dry eye disease in diabetes mellitus (221). In addition, Zheng et al (222) reported recently that dysregulated expression of circRNAs may be influenced by m6A modifications, and these circRNAs play significant roles in metabolism-associated fatty liver disease.

m6A modification of ncRNAs in neurological diseases

It has been reported that abnormal expression of m6A-related proteins also occurs in the nervous system, thereby affecting the development of neuroinflammation, AD, spinal cord injury (SCI) and cerebral ischemia-reperfusion injury (Fig. 5). For instance, Zhang et al (223) recently reported that m6A modification is enriched in circRNAs of neurons, and the m6A modification of circRNAs was reduced under oxygen-glucose deprivation and reoxygenation (OGD/R) injury conditions. Furthermore, through high-throughput sequencing, Zhang et al (224) identified eight m6A-modified circRNA that may be associated with the pathogenesis of AD. Furthermore, Liu et al (225) examined m6A modifications in SCI, revealing that 738 lncRNAs were differentially methylated (488 hypermethylated and 250 hypomethylated). In addition, Atrian et al (226) reported that tau-induced m6A methylation is a mechanistic driver of circMbl formation, and circMbl contributed to neurotoxicity and neurodegeneration. Electroacupuncture serum [serum was obtained from rats that were received electroacupuncture treatment 3 times at 'Renzhong' (GV26) and 'Baihui' (GV20) acupoints] alleviates OGD/R-induced astrocyte damage by regulating aquaporin 4 via m6A methylation of lncRNA MALAT1 (227).

Discussion

Various studies have shown the potential clinical value of m6A-modified ncRNAs as diagnostic biomarkers and prognostic indicators. It has been reported that the expression of m6A-modified circCUX1 was significantly upregulated in hypopharyngeal squamous cell carcinoma, and correlated with primary tumor size, lymph node metastasis, distant metastasis and tumor node metastasis stage, and its high expression indicated poor overall survival and poor disease-free survival (129). Furthermore, m6A-modified ABHD11-AS1 was found to be upregulated in human CRC tissues and related to poor prognosis (65). In addition, m6A-modified circCDYL existed in a stable form in HCC-derived exosomes, and was indicated to be a promising early diagnostic biomarker with an area under the curve of 0.896 (79). The distinct expression patterns of these novel ncRNA-derived peptides and proteins also offer considerable diagnostic potential. For instance, the serum levels of m6A-modified lncRNA AFAP1-AS1-encoded lncRNA AFAP1-AS1 translated mitochondrial-localized peptide were elevated in patients with non-small cell lung cancer (NSCLC) and associated with a poorer prognosis (190). The expression profiles of lncRNA-encoded microproteins in extracellular vesicles from patients with glioma differ from those in healthy donors (225). The high expression of circMAP3K4-455aa, a novel peptide encoded by m6A-modified circMAP3K4, predicts a worse prognosis for patients with HCC (17). Similarly, circMET encodes a 404-aa MET variant (MET404), and high MET404 expression was associated with poor prognosis in patients with glioblastoma (197).

The recent studies on m6A regulators and ncRNA-derived peptides render them promising therapeutic targets for numerous diseases, including malignant cancers. In esophageal squamous cell carcinoma, increasing m6A modification of circCREBBP could enhance the radiosensitivity of tumor cells (228). In CRC, ALKBH5 directly demethylates lncRNA CARMN, thereby suppressing the degradation of lncRNA CARMN and preserving lncRNA CARMN expression, and upregulated CARMN promoted tumor progression (92). Furthermore, Xinfeng capsule inhibited the progression of rheumatoid arthritis by modulating FTO-mediated m6A modification of lncRNA ENST00000619282 (229). These results indicated that modulating the activity of m6A-related enzymes, thereby regulating the m6A modification of target genes, has a critical role in disease treatment. A phase II study of bisantrene (a FTO inhibitor) in patients with relapsed/refractory acute myeloid leukemia showed that it achieved a partial remission, resulting in an overall response rate of 40% (230). Preclinical studies about METTL3 and METTL14 show promise in inhibiting tumor growth via direct anti-tumor effects and anti-cancer immune responses (231-233), and STC-15, a small-molecule inhibitor of METTL3, now has entered Phase I clinical trials (234). On the other hand, certain ncRNA-derived peptides have been reported to have special roles in disease treatment. For instance, a 188-aa peptide encoded by hsa_circRNA_103820 suppresses cell viability, induces apoptosis and inhibits cell migration and invasion in lung cancer (18). In addition, a tumor suppressor protein encoded by circKEAP1 inhibited stemness and metastasis by promoting vimentin proteasomal degradation and activating anti-tumor immunity in osteosarcoma (204). SPECC1-415aa, encoded by circSPECC1, inhibits the binding of annexin A1 (ANXA1) to EGFR by competitively binding to ANXA2, thereby preventing EGFR and AKT phosphorylation and restoring the sensitivity of temozolomide (TMZ)-resistant glioblastoma cells to TMZ (203). Although several ncRNA-encoded peptides have demonstrated tumor-suppressive functions, critical questions remain regarding their clinical applications. These include concerns about the feasibility of large-scale in vitro production and whether these cancer-suppressive micro-peptides can effectively target tumor cells upon injection. Future research should focus on unraveling the regulatory mechanisms underlying their expression and investigating how they exert their tumor-suppressing effects.

However, the use of ncRNAs or ncRNA-derived peptides for disease diagnosis or target therapy remains constrained by challenges. First, the roles of certain m6A-modified ncRNAs are conflicting. For instance, m6A-modified lncRNA MEG3 has been reported to suppress the proliferation, migration and invasion of HCC (235), while it promoted tumorigenesis and metastasis of NSCLC (104). Furthermore, it is difficult to detect m6A-modified ncRNAs or their encoded peptides in clinical samples. However, certain studies reported the existence of these ncRNAs or peptides in biofluids, such as blood and urine, indicating it is accessibility that using ncRNAs for testing diagnostic. For instance, m6A-modified circSLC38A1 was identified in serum exosomes of patients with bladder cancer and could distinguish patients with bladder cancer from healthy individuals with a diagnostic accuracy of 0.878 (236). Furthermore, Li et al (59) reported that circTRIM1 encoded TRIM1-269aa, which can be detected in exosomes from patients with triple-negative breast cancer. In addition, standard hybridization-based techniques cannot be applied for sensing m6A in RNAs and the development of new methods for accurate and sensitive profiling of locus-specific m6A in RNAs remains a great challenge. Liu et al (237) recently constructed a hierarchical DNA circuit for single-molecule profiling of locus-specific m6a-MALAT1 in clinical tissues. Finally, the dynamic nature of m6A modification makes it difficult to establish causal relationships in vivo. However, it has been reported that the transcriptome-wide m6A methylome was dynamically altered during initial diagnosis and relapse and can be monitored by methylated RNA immunoprecipitation next-generation sequencing (238). Recently, Song et al (38) presented a method termed proximity hybridization followed by primer exchange amplification, which could image m6A methylation sites concurrently in multiple cell types, revealing cell-to-cell variability in expression levels.

Conclusions

In conclusion, this review provided a comprehensive overview of the current understanding of m6A modification in ncRNAs, including miRNAs, lncRNAs and circRNAs. It explored how m6A modification regulates their biogenesis, maturation, localization and stability. Importantly, this review introduced the identification of peptides derived from m6A-modified ncRNAs. Furthermore, the diverse roles of m6A-modified ncRNAs and their encoded peptides in tumorigenesis, cardiovascular disorders, metabolic diseases and neurological diseases were discussed. It also highlights their potential clinical applications in cancer diagnosis, disease prediction and targeted therapy, offering new insights and perspectives to advance research in this field.

Availability of data and materials

Not applicable.

Authors' contributions

QY performed the literature search. QY and YL prepared the first draft of the manuscript. WS wrote and edited the manuscript. JL and DY drew the figures. WS and HS prepared the tables and were responsible for revising the manuscript. QY, WS and WC obtained funding support. All authors reviewed the manuscript, and have read and approved of 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.

Abbreviations:

m6A

N6-methyladenosine

METTL3

methyltransferase-like 3

WTAP

Wilms'tumor 1-associated protein

lncRNA

long non-coding RNA

circRNA

circular RNA

MALAT1

metastasis-associated lung adenocarcinoma transcript 1

hnRNP

heterogeneous nuclear ribonucleoprotein

IGF2BP

insulin-like growth factor 2 mRNA-binding protein

NSCLC

non-small cell lung cancer

GBM

glioblastoma

HCC

hepatocellular carcinoma

CRC

colorectal cancer

HPSCC

hypopharyngeal squamous cell carcinoma

VSMC

vascular smooth muscle cell

LSCC

laryngeal squamous cell carcinoma

RBM

RNA-binding motif

SNHG1

small nucleolar RNA host gene 1

FTO

fat mass and obesity-associated protein

ALKBH5

α-ketoglutarate-dependent dioxygenase alk B homolog 5

ESCC

esophageal squamous cell carcinoma

TAM

tumour-associated macrophage

OC

ovarian cancer

BC

breast cancer

GC

gastric cancer

I/R

ischemia/reperfusion

EV

extracellular vesicle

AGO2

Argonaute 2

LPS

lipopolysaccharide

Acknowledgements

Not applicable.

Funding

This research was supported by the Shenzhen Medical Research Fund (grant no. A2403030); the Scientific Research Foundation of Southwest Medical University (grant no. 2021ZKMS009); and Shenzhen Science and Technology Projects (grant nos. KCXFZ20230731093059012 and JSGG20220831110400001).

References

1 

Liang W, Yi H, Mao C, Meng Q, Wu X, Li S and Xue J: Research progress of RNA methylation modification in colorectal cancer. Front Pharmacol. 13:9036992022. View Article : Google Scholar : PubMed/NCBI

2 

Li Y, Jin H, Li Q, Shi L, Mao Y and Zhao L: The role of RNA methylation in tumor immunity and its potential in immunotherapy. Mol Cancer. 23:1302024. View Article : Google Scholar : PubMed/NCBI

3 

Wen T, Li T, Xu Y, Zhang Y, Pan H and Wang Y: The role of m6A epigenetic modifications in tumor coding and non-coding RNA processing. Cell Commun Signal. 21:3552023. View Article : Google Scholar : PubMed/NCBI

4 

An Y and Duan H: The role of m6A RNA methylation in cancer metabolism. Mol Cancer. 21:142022. View Article : Google Scholar : PubMed/NCBI

5 

Zhang Q and Xu K: The role of regulators of RNA m6A methylation in lung cancer. Genes Dis. 10:495–504. 2023. View Article : Google Scholar

6 

Mendel M, Chen KM, Homolka D, Gos P, Pandey RR, McCarthy AA and Pillai RS: Methylation of structured RNA by the m6A writer METTL16 is essential for mouse embryonic development. Mol Cell. 71:986–1000.e1011. 2018. View Article : Google Scholar

7 

Ma H, Hong Y, Xu Z, Weng Z, Yang Y, Jin D, Chen Z, Yue J, Zhou X, Xu Z, et al: N6-methyladenosine (m6A) modification in hepatocellular carcinoma. Biomed Pharmacother. 173:1163652024. View Article : Google Scholar

8 

Qu Y, Gao N, Zhang S, Gao L, He B, Wang C, Gong C, Shi Q, Li Z, Yang S and Xiao Y: Role of N6-methyladenosine RNA modification in cancer. MedComm (2020). 5:e7152024. View Article : Google Scholar : PubMed/NCBI

9 

Guttman M and Rinn JL: Modular regulatory principles of large non-coding RNAs. Nature. 482:339–346. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar

11 

Gao K, Li X, Ni J, Wu B, Guo J, Zhang R and Wu G: Non-coding RNAs in enzalutamide resistance of castration-resistant prostate cancer. Cancer Lett. 566:2162472023. View Article : Google Scholar : PubMed/NCBI

12 

Kopp F and Mendell JT: Functional classification and experimental dissection of long noncoding RNAs. Cell. 172:393–407. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Fabian MR, Sonenberg N and Filipowicz W: Regulation of mRNA translation and stability by microRNAs. Annu Rev Biochem. 79:351–379. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Zhou WY, Cai ZR, Liu J, Wang DS, Ju HQ and Xu RH: Circular RNA: Metabolism, functions and interactions with proteins. Mol Cancer. 19:1722020. View Article : Google Scholar : PubMed/NCBI

15 

Ning K, Zhao J, Feng Z, Park SY, McFarlin S, Cheng F, Yan Z, Wang J and Qiu J: N6-methyladenosine modification of a parvovirus-encoded small noncoding RNA facilitates viral DNA replication through recruiting Y-family DNA polymerases. Proc Natl Acad Sci USA. 121:e23207821212024. View Article : Google Scholar : PubMed/NCBI

16 

Garbo S, D'Andrea D, Colantoni A, Fiorentino F, Mai A, Ramos A, Tartaglia GG, Tancredi A, Tripodi M and Battistelli C: m6A modification inhibits miRNAs' intracellular function, favoring their extracellular export for intercellular communication. Cell Rep. 43:1143692024. View Article : Google Scholar : PubMed/NCBI

17 

Duan JL, Chen W, Xie JJ, Zhang ML, Nie RC, Liang H, Mei J, Han K, Xiang ZC, Wang FW, et al: A novel peptide encoded by N6-methyladenosine modified circMAP3K4 prevents apoptosis in hepatocellular carcinoma. Mol Cancer. 21:932022. View Article : Google Scholar : PubMed/NCBI

18 

Zhou J, Yao L, Su Y and Tian L: IGF2BP3 loss inhibits cell progression by upregulating has_circRNA_103820, and hsa_ circRNA_103820-encoded peptide inhibits cell progression by inactivating the AKT pathway in lung cancer. Chem Biol Drug Des. 103:e144732024. View Article : Google Scholar

19 

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

20 

Huang H, Weng H and Chen J: m6A modification in coding and non-coding RNAs: Roles and therapeutic implications in cancer. Cancer Cell. 37:270–288. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Liang J, Yi Q, Liu Y, Li J, Yang Z and Sun W and Sun W: Recent advances of m6A methylation in skeletal system disease. J Transl Med. 22:1532024. View Article : Google Scholar : PubMed/NCBI

22 

Qiao Y, Mei Y, Xia M, Luo D and Gao L: The role of m6A modification in the risk prediction and Notch1 pathway of Alzheimer's disease. iScience. 27:1102352024. View Article : Google Scholar : PubMed/NCBI

23 

Zhu L, Zhang H, Zhang X and Xia L: RNA m6A methylation regulators in sepsis. Mol Cell Biochem. 479:2165–2180. 2024. View Article : Google Scholar

24 

Lu W, Yang X, Zhong W, Chen G, Guo X, Ye Q, Xu Y, Qi Z, Ye Y, Zhang J, et al: METTL14-mediated m6A epitranscriptomic modification contributes to chemotherapy-induced neuropathic pain by stabilizing GluN2A expression via IGF2BP2. J Clin Invest. 134:e1748472024. View Article : Google Scholar : PubMed/NCBI

25 

Wu S, Wang H, Yang Q, Liu Z, Du J, Wang L, Chen S, Lu Q and Yang D: METTL3 regulates M6A methylation-modified EBV-pri-miR-BART3-3p to promote NK/T cell lymphoma growth. Cancer Lett. 597:2170582024. View Article : Google Scholar : PubMed/NCBI

26 

Liu X, Feng M, Hao X, Gao Z, Wu Z, Wang Y, Du L and Wang C: m6A methylation regulates hypoxia-induced pancreatic cancer glycolytic metabolism through ALKBH5-HDAC4-HIF1α positive feedback loop. Oncogene. 42:2047–2060. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Ning J, Yan J, Wang S, Cui Z, Xue Y, Juan J and Yang H: Demethylase FTO-mediated m6A modification of SIK1 modulates placental cytotrophoblast syncytialization in type 2 diabetes mellitus. iScience. 27:1099002024. View Article : Google Scholar : PubMed/NCBI

28 

Shu F, Liu H, Chen X, Liu Y, Zhou J, Tang L, Cao W, Yang S, Long Y, Li R, et al: m6A modification promotes EMT and metastasis of castration-resistant prostate cancer by upregulating NFIB. Cancer Res. 84:1947–1962. 2024. View Article : Google Scholar : PubMed/NCBI

29 

Zhang X, Su T, Wu Y, Cai Y, Wang L, Liang C, Zhou L, Wang S, Li XX, Peng S, et al: N6-Methyladenosine reader YTHDF1 promotes stemness and therapeutic resistance in hepatocellular carcinoma by enhancing NOTCH1 expression. Cancer Res. 84:827–840. 2024. View Article : Google Scholar : PubMed/NCBI

30 

Li F, Wang F, Wang L, Wang J, Wei S, Meng J, Li Y, Feng L and Jiang P: m6A reader YTHDC2 mediates NCOA4 mRNA stability affecting ferritinophagy to alleviate secondary injury after intracerebral haemorrhage. Epigenetics. 19:23268682024. View Article : Google Scholar : PubMed/NCBI

31 

Klein P, Howe MP, Harley J, Crook H, Serna SE, Roumeliotis TI, Choudhary JS, Chakrabarti AM, Luisier R, Patani R and Ramos A: m6a methylation orchestrates IMP1 regulation of microtubules during human neuronal differentiation. Nat Commun. 15:48192024. View Article : Google Scholar : PubMed/NCBI

32 

Xia T, Dai XY, Sang MY, Zhang X, Xu F, Wu J, Shi L, Wei JF and Ding Q: IGF2BP2 drives cell cycle progression in triple-negative breast cancer by recruiting EIF4A1 to promote the m6A-modified CDK6 translation initiation process. Adv Sci (Weinh). 11:e23051422024. View Article : Google Scholar

33 

Lv L, Wei Q, Zhang J, Dong Y, Shan Z, Chang N, Zhao Y, Bian P and Yi Q: IGF2BP3 prevent HMGB1 mRNA decay in bladder cancer and development. Cell Mol Biol Lett. 29:392024. View Article : Google Scholar : PubMed/NCBI

34 

Wang X, Lu X, Wang P, Chen Q, Xiong L, Tang M, Hong C, Lin X, Shi K, Liang L and Lin J: SRSF9 promotes colorectal cancer progression via stabilizing DSN1 mRNA in an m6A-related manner. J Transl Med. 20:1982022. View Article : Google Scholar : PubMed/NCBI

35 

Shi S, Chen Q, Yang Y, Li Z, Zheng R, Zhang R, Liu Z and Cheng Y: HnRNPA2B1 promotes cardiac ferroptosis via m6A-dependent stabilization of PFN2 mRNA in myocardial ischemia-reperfusion injury. Free Radic Biol Med. 232:231–243. 2025. View Article : Google Scholar : PubMed/NCBI

36 

Baek A, Lee GE, Golconda S, Rayhan A, Manganaris AA, Chen S, Tirumuru N, Yu H, Kim S, Kimmel C, et al: Single-molecule epitranscriptomic analysis of full-length HIV-1 RNAs reveals functional roles of site-specific m6As. Nat Microbiol. 9:1340–1355. 2024. View Article : Google Scholar : PubMed/NCBI

37 

McIntyre ABR, Alexander N, Grigorev K, Bezdan D, Sichtig H, Chiu CY and Mason C: Single-molecule sequencing detection of N6-methyladenine in microbial reference materials. Nat Commun. 10:5792019. View Article : Google Scholar : PubMed/NCBI

38 

Song M, Wang J, Hou J, Fu T, Feng Y, Lv W, Ge F, Peng R, Han D and Tan W: Multiplexed in situ imaging of site-specific m6A methylation with proximity hybridization followed by primer exchange amplification (m6A-PHPEA). ACS Nano. 18:27537–27546. 2024. View Article : Google Scholar : PubMed/NCBI

39 

Zhang Q, Dai Y, Teng X and Li J: Visualization and quantification of single-Base m6A methylation. Angew Chem Int Ed Engl. 64:e2024209772025. View Article : Google Scholar

40 

Yi Q, Xie W, Sun W, Sun W and Liao Y: A concise review of MicroRNA-383: Exploring the insights of its function in tumorigenesis. J Cancer. 13:313–324. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Yi Q, Cui H, Liao Y, Xiong J, Ye X and Sun W: A minor review of microRNA-338 exploring the insights of its function in tumorigenesis. Biomed Pharmacother. 139:1117202021. View Article : Google Scholar : PubMed/NCBI

43 

Valadkhan S: snRNAs as the catalysts of pre-mRNA splicing. Curr Opin Chem Biol. 9:603–608. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Liu Y, Liu X, Lin C, Jia X, Zhu H, Song J and Zhang Y: Noncoding RNAs regulate alternative splicing in cancer. J Exp Clin Cancer Res. 40:112021. View Article : Google Scholar : PubMed/NCBI

45 

Zhang X, Hong R, Chen W, Xu M and Wang L: The role of long noncoding RNA in major human disease. Bioorg Chem. 92:1032142019. View Article : Google Scholar : PubMed/NCBI

46 

Statello L, Guo CJ, Chen LL and Huarte M: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 22:96–118. 2021. View Article : Google Scholar

47 

Wu H, Yang L and Chen LL: The diversity of long noncoding RNAs and their generation. Trends Genet. 33:540–552. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Liu Y, Ding W, Yu W, Zhang Y, Ao X and Wang J: Long non-coding RNAs: Biogenesis, functions, and clinical significance in gastric cancer. Mol Ther Oncolytics. 23:458–476. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Sengupta P, Roy A, Roy L, Bose D, Halder S, Jana K, Mukherjee G and Chatterjee S: Long non-coding intergenic RNA, LINC00273 induces cancer metastasis and stemness via miRNA sponging in triple negative breast cancer. Int J Biol Macromol. 274:1327302024. View Article : Google Scholar : PubMed/NCBI

50 

Yao ZT, Yang YM, Sun MM, He Y, Liao L, Chen KS and Li B: New insights into the interplay between long non-coding RNAs and RNA-binding proteins in cancer. Cancer Commun (Lond). 42:117–140. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Zhang Z, Zhou K, Han L, Small A, Xue J, Huang H, Weng H, Su R, Tan B, Shen C, et al: RNA m6A reader YTHDF2 facilitates precursor miR-126 maturation to promote acute myeloid leukemia progression. Genes Dis. 11:382–396. 2024. View Article : Google Scholar

52 

Liu CX and Chen LL: Circular RNAs: Characterization, cellular roles, and applications. Cell. 185:2016–2034. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Chen LL: The biogenesis and emerging roles of circular RNAs. Nat Rev Mol Cell Biol. 17:205–211. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Yi Q, Feng J, Liao Y and Sun W: Circular RNAs in chemotherapy resistance of lung cancer and their potential therapeutic application. IUBMB Life. 75:225–237. 2023. View Article : Google Scholar

55 

Conn VM, Chinnaiyan AM and Conn SJ: Circular RNA in cancer. Nat Rev Cancer. 24:597–613. 2024. View Article : Google Scholar : PubMed/NCBI

56 

Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB and Kjems J: The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 20:675–691. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Dayal S, Chaubey D, Joshi DC, Ranmale S and Pillai B: Noncoding RNAs: Emerging regulators of behavioral complexity. Wiley Interdiscip Rev RNA. 15:e18472024. View Article : Google Scholar : PubMed/NCBI

58 

Yi Q, Feng J, Lan W, Shi H and Sun W and Sun W: CircRNA and lncRNA-encoded peptide in diseases, an update review. Mol Cancer. 23:2142024. View Article : Google Scholar : PubMed/NCBI

59 

Li Y, Wang Z, Yang J, Sun Y, He Y, Wang Y, Chen X, Liang Y, Zhang N, Wang X, et al: CircTRIM1 encodes TRIM1-269aa to promote chemoresistance and metastasis of TNBC via enhancing CaM-dependent MARCKS translocation and PI3K/AKT/mTOR activation. Mol Cancer. 23:1022024. View Article : Google Scholar : PubMed/NCBI

60 

Alarcón CR, Lee H, Goodarzi H, Halberg N and Tavazoie SF: N6-methyladenosine marks primary microRNAs for processing. Nature. 519:482–485. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Chen L, Hu Y, Zhang M, Liu L, Ma J, Xu Z, Zhang J, Gu H and Chen K: METTL14 affects UVB-induced human dermal fibroblasts photoaging via miR-100-3p biogenesis in an m6A-dependent manner. Aging Cell. 23:e141232024. View Article : Google Scholar

62 

Chen P, Li S, Zhang K, Zhao R, Cui J, Zhou W, Liu Y, Zhang L and Cheng Y: N6-methyladenosine demethylase ALKBH5 suppresses malignancy of esophageal cancer by regulating microRNA biogenesis and RAI1 expression. Oncogene. 40:5600–5612. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Ding D, Shang W, Shi K, Ying J, Wang L, Chen Z and Zhang C: FTO/m6A mediates miR-138-5p maturation and regulates gefitinib resistance of lung adenocarcinoma cells by miR-138-5p/LCN2 axis. BMC Cancer. 24:12702024. View Article : Google Scholar : PubMed/NCBI

64 

Hou Y, Zhang Q, Pang W, Hou L, Liang Y, Han X, Luo X, Wang P, Zhang X, Li L and Meng X: YTHDC1-mediated augmentation of miR-30d in repressing pancreatic tumorigenesis via attenuation of RUNX1-induced transcriptional activation of Warburg effect. Cell Death Differ. 28:3105–3124. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Bian Y, Xu S, Gao Z, Ding J, Li C, Cui Z, Sun H, Li J, Pu J and Wang K: m6A modification of lncRNA ABHD11-AS1 promotes colorectal cancer progression and inhibits ferroptosis through TRIM21/IGF2BP2/FOXM1 positive feedback loop. Cancer Lett. 596:2170042024. View Article : Google Scholar

66 

Zhao K, Chen L, Xie Y, Ren N, Li J, Zhai X, Zheng S, Liu K, Wang C, Qiu Q, et al: m6A/HOXA10-AS/ITGA6 axis aggravates oxidative resistance and malignant progression of laryngeal squamous cell carcinoma through regulating Notch and Keap1/Nrf2 pathways. Cancer Lett. 587:2167352024. View Article : Google Scholar : PubMed/NCBI

67 

Shi Q, Li Z, Dong Y, Yang G and Li M: LncRNA THRIL, transcriptionally activated by AP-1 and stabilized by METTL14-mediated m6A modification, accelerates LPS-evoked acute injury in alveolar epithelial cells. Int Immunopharmacol. 123:1107402023. View Article : Google Scholar : PubMed/NCBI

68 

Lin Z, Huang Z, Qiu J, Shi Y, Zuo D, Qiu Z, He W, Niu Y, Yuan Y and Li B: m6A-mediated lnc-OXAR promotes oxaliplatin resistance by enhancing Ku70 stability in non-alcoholic steatohepatitis-related hepatocellular carcinoma. J Exp Clin Cancer Res. 43:2062024. View Article : Google Scholar

69 

Yang N, Yan N, Bai Z, Du S, Zhang J, Zhang L and Zhang Z: FTO attenuates LPS-induced acute kidney injury by inhibiting autophagy via regulating SNHG14/miR-373-3p/ATG7 axis. Int Immunopharmacol. 128:1114832024. View Article : Google Scholar : PubMed/NCBI

70 

Zhang Y, Liu X, Wang Y, Lai S, Wang Z, Yang Y, Liu W, Wang H and Tang B: The m6A demethylase ALKBH5-mediated upregulation of DDIT4-AS1 maintains pancreatic cancer stemness and suppresses chemosensitivity by activating the mTOR pathway. Mol Cancer. 21:1742022. View Article : Google Scholar

71 

Chen S, Duan X, He Y and Chen W: METTL3 promotes osteogenic differentiation of human umbilical cord mesenchymal stem cells by up-regulating m6A modification of circCTTN. Biosci Rep. 44:BSR202311862024. View Article : Google Scholar : PubMed/NCBI

72 

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

73 

Du X, Shi L, Wang B and Zhang G: WTAP mediated m6A-modified circ_0056856 contributes to the proliferation, migration, and invasion of IL-22-stimulated human keratinocyte by miR-197-3p/CDK1 axis. Arch Dermatol Res. 316:2082024. View Article : Google Scholar : PubMed/NCBI

74 

Chen Y, Ling Z, Cai X, Xu Y, Lv Z, Man D, Ge J, Yu C, Zhang D, Zhang Y, et al: Activation of YAP1 by N6-methyladenosine-modified circCPSF6 drives malignancy in hepatocellular carcinoma. Cancer Res. 82:599–614. 2022. View Article : Google Scholar

75 

Zhu X and Zhang P: m6A-modified circXPO1 accelerates colorectal cancer progression via interaction with FMRP to promote WWC2 mRNA decay. J Transl Med. 22:9312024. View Article : Google Scholar : PubMed/NCBI

76 

Wu X, Fang Y, Gu Y, Shen H, Xu Y, Xu T, Shi R, Xu D, Zhang J, Leng K, et al: Fat mass and obesity-associated protein (FTO) mediated m6A modification of circFAM192A promoted gastric cancer proliferation by suppressing SLC7A5 decay. Mol Biomed. 5:112024. View Article : Google Scholar

77 

Kong Z, Lu Y, Yang Y, Chang K, Lin Y, Huang Y, Wang C, Zhang L, Xu W, Zhao S and Li Y: m6A-mediated biogenesis of circDDIT4 inhibits prostate cancer progression by sequestrating ELAVL1/HuR. Mol Cancer Res. 21:1342–1355. 2023. View Article : Google Scholar :

78 

Xu Y, Weng X, Qiu J and Wang S: Biogenesis of circRBM33 mediated by N6-methyladenosine and its function in abdominal aortic aneurysm. Epigenetics. 19:23924012024. View Article : Google Scholar : PubMed/NCBI

79 

Wei Y, Fu J, Zhang H, Ling Y, Tang X, Liu S, Yu M, Liu F, Zhuang G, Qian H, et al: N6-methyladenosine modification promotes hepatocarcinogenesis through circ-CDYL-enriched and EpCAM-positive liver tumor-initiating exosomes. iScience. 26:1080222023. View Article : Google Scholar : PubMed/NCBI

80 

Rao X, Lai L, Li X, Wang L, Li A and Yang Q: N6-methyladenosine modification of circular RNA circ-ARL3 facilitates Hepatitis B virus-associated hepatocellular carcinoma via sponging miR-1305. IUBMB Life. 73:408–417. 2021. View Article : Google Scholar

81 

Liu Z, Wang T, She Y, Wu K, Gu S, Li L, Dong C, Chen C and Zhou Y: N6-methyladenosine-modified circIGF2BP3 inhibits CD8(+) T-cell responses to facilitate tumor immune evasion by promoting the deubiquitination of PD-L1 in non-small cell lung cancer. Mol Cancer. 20:1052021. View Article : Google Scholar

82 

Dattilo D, Di Timoteo G, Setti A, Giuliani A, Peruzzi G, Nebot MB, Centrón-Broco A, Mariani D, Mozzetta C and Bozzoni I: The m6A reader YTHDC1 and the RNA helicase DDX5 control the production of rhabdomyosarcoma-enriched circRNAs. Nat Commun. 14:18982023. View Article : Google Scholar

83 

Wu X, Fu M, Ge C, Zhou H, Huang H, Zhong M, Zhang M, Xu H, Zhu G, Hua W, et al: m(6)A-mediated upregulation of lncRNA CHASERR promotes the progression of glioma by modulating the miR-6893-3p/TRIM14 axis. Mol Neurobiol. 61:5418–5440. 2024. View Article : Google Scholar : PubMed/NCBI

84 

Wu W, Li X, Zhou Z, He H, Pang C, Ye S and Quan JH: METTL14 regulates inflammation in ulcerative colitis via the lncRNA DHRS4-AS1/miR-206/A3AR axis. Cell Biol Toxicol. 40:952024. View Article : Google Scholar : PubMed/NCBI

85 

Liu P, Zhang B, Chen Z, He Y, Du Y, Liu Y and Chen X: m6A-induced lncRNA MALAT1 aggravates renal fibrogenesis in obstructive nephropathy through the miR-145/FAK pathway. Aging (Albany NY). 12:5280–5299. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Yu H and Zhang Z: ALKBH5-mediated m6A demethylation of lncRNA RMRP plays an oncogenic role in lung adenocarcinoma. Mamm Genome. 32:195–203. 2021. View Article : Google Scholar : PubMed/NCBI

87 

Liu H, Liang J, Dai X, Peng Y, Xiong W, Zhang L, Li X, Li W, Liu K, Bi S, et al: Transcriptome-wide N6-methyladenosine (m6A) methylation profiling of long non-coding RNAs in ovarian endometriosis. Genomics. 116:1108032024. View Article : Google Scholar : PubMed/NCBI

88 

Yin H, Chen L, Piao S, Wang Y, Li Z, Lin Y, Tang X, Zhang H, Zhang H and Wang X: M6A RNA methylation-mediated RMRP stability renders proliferation and progression of non-small cell lung cancer through regulating TGFBR1/SMAD2/SMAD3 pathway. Cell Death Differ. 30:605–617. 2023. View Article : Google Scholar :

89 

Chang M, Cui X, Sun Q, Wang Y, Liu J, Sun Z, Ren J, Sun Y, Han L and Li W: Lnc-PLCB1 is stabilized by METTL14 induced m6A modification and inhibits Helicobacter pylori mediated gastric cancer by destabilizing DDX21. Cancer Lett. 588:2167462024. View Article : Google Scholar : PubMed/NCBI

90 

Xie R, Liu L, Lu X, He C, Yao H and Li G: N6-methyladenosine modification of OIP5-AS1 promotes glycolysis, tumorigenesis, and metastasis of gastric cancer by inhibiting Trim21-mediated hnRNPA1 ubiquitination and degradation. Gastric Cancer. 27:49–71. 2024. View Article : Google Scholar :

91 

Luo XJ, Lu YX, Wang Y, Huang R, Liu J, Jin Y, Liu ZK, Liu ZX, Huang QT, Pu H, et al: M6A-modified lncRNA FAM83H-AS1 promotes colorectal cancer progression through PTBP1. Cancer Lett. 598:2170852024. View Article : Google Scholar : PubMed/NCBI

92 

Liu N, Jiang X, Zhang G, Long S, Li J, Jiang M, Jia G, Sun R, Zhang L and Zhang Y: LncRNA CARMN m6A demethylation by ALKBH5 inhibits mutant p53-driven tumour progression through miR-5683/FGF2. Clin Transl Med. 14:e17772024. View Article : Google Scholar : PubMed/NCBI

93 

Yao L, Li T, Teng Y, Guo J, Zhang H, Xia L and Wu Q: ALKHB5-demethylated lncRNA SNHG15 promotes myeloma tumorigenicity by increasing chromatin accessibility and recruiting H3K36me3 modifier SETD2. Am J Physiol Cell Physiol. 326:C684–C697. 2024. View Article : Google Scholar :

94 

Fang D, Ou X, Sun K, Zhou X, Li Y, Shi P, Zhao Z, He Y, Peng J and Xu J: m6A modification-mediated lncRNA TP53TG1 inhibits gastric cancer progression by regulating CIP2A stability. Cancer Sci. 113:4135–4150. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Wang J, Zhang J, Liu H, Meng L, Gao X, Zhao Y, Wang C, Gao X, Fan A, Cao T, et al: N6-methyladenosine reader hnRNPA2B1 recognizes and stabilizes NEAT1 to confer chemoresistance in gastric cancer. Cancer Commun (Lond). 44:469–490. 2024. View Article : Google Scholar : PubMed/NCBI

96 

Yi J, Ma X, Ying Y, Liu Z, Tang Y, Shu X, Sun J, Wu Y, Lu D, Wang X, et al: N6-methyladenosine-modified CircPSMA7 enhances bladder cancer malignancy through the miR-128-3p/MAPK1 axis. Cancer Lett. 585:2166132024. View Article : Google Scholar : PubMed/NCBI

97 

Xu C, Zhou J, Zhang X, Kang X, Liu S, Song M, Chang C, Lin Y and Wang Y: N(6)-methyladenosine-modified circ_104797 sustains cisplatin resistance in bladder cancer through acting as RNA sponges. Cell Mol Biol Lett. 29:282024. View Article : Google Scholar

98 

Li H, Lin R, Zhang Y, Zhu Y, Huang S, Lan J, Lu N, Xie C, He S and Zhang W: N6-methyladenosine-modified circPLPP4 sustains cisplatin resistance in ovarian cancer cells via PIK3R1 upregulation. Mol Cancer. 23:52024. View Article : Google Scholar : PubMed/NCBI

99 

Jiang S, Gao L, Li J, Zhang F, Zhang Y and Liu J: N6-methyladenosine-modified circ_0000337 sustains bortezomib resistance in multiple myeloma by regulating DNA repair. Front Cell Dev Biol. 12:13832322024. View Article : Google Scholar : PubMed/NCBI

100 

Li J, Xu X, Xu K, Zhou X, Wu K, Yao Y, Liu Z, Chen C, Wang L, Sun Z, et al: N6-methyladenosine-modified circSLCO1B3 promotes intrahepatic cholangiocarcinoma progression via regulating HOXC8 and PD-L1. J Exp Clin Cancer Res. 43:1192024. View Article : Google Scholar : PubMed/NCBI

101 

Zhu X, Li W, Lu M, Shang J, Zhou J, Lin L, Liu Y, Xing J, Zhang M, Zhao S, et al: M6A demethylase FTO-stabilized exosomal circBRCA1 alleviates oxidative stress-induced granulosa cell damage via the miR-642a-5p/FOXO1 axis. J Nanobiotechnology. 22:3672024. View Article : Google Scholar

102 

Li L, Chen J, Wang A and Yi K: ALKBH5 regulates ovarian cancer growth via demethylating long noncoding RNA PVT1 in ovarian cancer. J Cell Mol Med. 28:e180662024. View Article : Google Scholar :

103 

Shen W, Pu J, Zuo Z, Gu S, Sun J, Tan B, Wang L, Cheng J and Zuo Y: The RNA demethylase ALKBH5 promotes the progression and angiogenesis of lung cancer by regulating the stability of the LncRNA PVT1. Cancer Cell Int. 22:3532022. View Article : Google Scholar : PubMed/NCBI

104 

Li K, Gong Q, Xiang XD, Guo G, Liu J, Zhao L, Li J, Chen N, Li H, Zhang LJ, et al: HNRNPA2B1-mediated m6A modification of lncRNA MEG3 facilitates tumorigenesis and metastasis of non-small cell lung cancer by regulating miR-21-5p/PTEN axis. J Transl Med. 21:3822023. View Article : Google Scholar

105 

Dong F, Qin X, Wang B, Li Q, Hu J, Cheng X, Guo D, Cheng F, Fang C, Tan Y, et al: ALKBH5 facilitates hypoxia-induced paraspeckle assembly and IL8 secretion to generate an immunosuppressive tumor microenvironment. Cancer Res. 81:5876–5888. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Hu J, Duan H, Zou J, Ding W, Wei Z, Peng Q, Li Z, Duan R, Sun J and Zhu J: METTL3-dependent N6-methyladenosine modification is involved in berberine-mediated neuroprotection in ischemic stroke by enhancing the stability of NEAT1 in astrocytes. Aging (Albany NY). 16:299–321. 2024.PubMed/NCBI

107 

Chang YZ, Chai RC, Pang B, Chang X, An SY, Zhang KN, Jiang T and Wang YZ: METTL3 enhances the stability of MALAT1 with the assistance of HuR via m6A modification and activates NF-κB to promote the malignant progression of IDH-wildtype glioma. Cancer Lett. 511:36–46. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Zhang Y, Xu Y, Qiu G, Luo Y, Bao Y, Lu J, Wang T and Wang Y: METTL3 mediated MALAT1 m6A modification promotes proliferation and metastasis in osteosarcoma cells. Mol Biotechnol. 66:3538–3548. 2024. View Article : Google Scholar

109 

Han L, Lei G, Chen Z, Zhang Y, Huang C and Chen W: IGF2BP2 regulates MALAT1 by serving as an N6-methyladenosine reader to promote NSCLC proliferation. Front Mol Biosci. 8:7800892021. View Article : Google Scholar

110 

Lu S, Han L, Hu X, Sun T, Xu D, Li Y, Chen Q, Yao W, He M, Wang Z, et al: N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: Implication in colorectal cancer. J Hematol Oncol. 14:1882021. View Article : Google Scholar : PubMed/NCBI

111 

Peng J, Zheng H, Liu F, Wu Q and Liu S: The m6A methyltransferase METTL3 affects autophagy and progression of nasopharyngeal carcinoma by regulating the stability of lncRNA ZFAS1. Infect Agent Cancer. 17:12022. View Article : Google Scholar : PubMed/NCBI

112 

Zhou X, Yang Y, Li Y, Liang G, Kang D, Zhou B and Li Q: METTL3 contributes to osteosarcoma progression by increasing DANCR mRNA stability via m6A modification. Front Cell Dev Biol. 9:7847192021. View Article : Google Scholar

113 

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 :

114 

Wu S, Chi C, Weng S, Zhou W and Liu Z: IGF2BP2 promotes lncRNA DANCR stability mediated glycolysis and affects the progression of FLT3-ITD + acute myeloid leukemia. Apoptosis. 28:1035–1047. 2023. View Article : Google Scholar : PubMed/NCBI

115 

Rong D, Dong Q, Qu H, Deng X, Gao F, Li Q and Sun P: m(6) A-induced LINC00958 promotes breast cancer tumorigenesis via the miR-378a-3p/YY1 axis. Cell Death Discov. 7:272021. View Article : Google Scholar

116 

Zuo X, Chen Z, Gao W, Zhang Y, Wang J, Wang J, Cao M, Cai J, Wu J and Wang X: M6A-mediated upregulation of LINC00958 increases lipogenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. J Hematol Oncol. 13:52020. View Article : Google Scholar : PubMed/NCBI

117 

Yang D, Chang S, Li F, Ma M, Yang J, Lv X, Huangfu L and Jia C: m6 A transferase KIAA1429-stabilized LINC00958 accelerates gastric cancer aerobic glycolysis through targeting GLUT1. IUBMB Life. 73:1325–1333. 2021. View Article : Google Scholar : PubMed/NCBI

118 

Zhuang S, Ma Y, Zeng Y, Lu C, Yang F, Jiang N, Ge J, Ju H, Zhong C, Wang J, et al: METTL14 promotes doxorubicin-induced cardiomyocyte ferroptosis by regulating the KCNQ1OT1-miR-7-5p-TFRC axis. Cell Biol Toxicol. 39:1015–1035. 2023. View Article : Google Scholar

119 

Zhou Z, Cao Y, Yang Y, Wang S and Chen F: METTL3-mediated m6A modification of lnc KCNQ1OT1 promotes doxorubicin resistance in breast cancer by regulating miR-103a-3p/MDR1 axis. Epigenetics. 18:22170332023. View Article : Google Scholar

120 

Li Y, Yan B, Wang X, Li Q, Kan X, Wang J, Sun Y, Wang P, Tian L and Liu M: ALKBH5-mediated m6A modification of lncRNA KCNQ1OT1 triggers the development of LSCC via upregulation of HOXA9. J Cell Mol Med. 26:385–398. 2022. View Article : Google Scholar

121 

Ji F, Lu Y, Chen S, Lin X, Yu Y, Zhu Y and Luo X: m6A methyltransferase METTL3-mediated lncRNA FOXD2-AS1 promotes the tumorigenesis of cervical cancer. Mol Ther Oncolytics. 22:574–581. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Ren Z, Hu Y, Sun J, Kang Y, Li G and Zhao H: N6-methyladenosine methyltransferase WTAP-stabilized FOXD2-AS1 promotes the osteosarcoma progression through m(6)A/FOXM1 axis. Bioengineered. 13:7963–7973. 2022. View Article : Google Scholar : PubMed/NCBI

123 

Lang C, Yin C, Lin K, Li Y, Yang Q, Wu Z, Du H, Ren D, Dai Y and Peng X: m6 A modification of lncRNA PCAT6 promotes bone metastasis in prostate cancer through IGF2BP2-mediated IGF1R mRNA stabilization. Clin Transl Med. 11:e4262021. View Article : Google Scholar

124 

Liu HT, Zou YX, Zhu WJ, Sen-Liu, Zhang GH, Ma RR, Guo XY and Gao P: lncRNA THAP7-AS1, transcriptionally activated by SP1 and post-transcriptionally stabilized by METTL3-mediated m6A modification, exerts oncogenic properties by improving CUL4B entry into the nucleus. Cell Death Differ. 29:627–641. 2022. View Article : Google Scholar :

125 

Zheng Y, Wang Y, Liu Y, Xie L, Ge J, Yu G and Zhao G: N6-methyladenosine modification of PTTG3P contributes to colorectal cancer proliferation via YAP1. Front Oncol. 11:6697312021. View Article : Google Scholar : PubMed/NCBI

126 

Huang T, Cao L, Feng N, Xu B, Dong Y and Wang M: N6-methyladenosine (m(6)A)-mediated lncRNA DLGAP1-AS1enhances breast canceradriamycin resistance through miR-299-3p/WTAP feedback loop. Bioengineered. 12:10935–10944. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Liu H, Qin S, Liu C, Jiang L, Li C, Yang J, Zhang S, Yan Z, Liu X, Yang J and Sun X: m6A reader IGF2BP2-stabilized CASC9 accelerates glioblastoma aerobic glycolysis by enhancing HK2 mRNA stability. Cell Death Discov. 7:2922021. View Article : Google Scholar

128 

Xue L, Li J, Lin Y, Liu D, Yang Q, Jian J and Peng J: m6 A transferase METTL3-induced lncRNA ABHD11-AS1 promotes the Warburg effect of non-small-cell lung cancer. J Cell Physiol. 236:2649–2658. 2021. View Article : Google Scholar

129 

Wu P, Fang X, Liu Y, Tang Y, Wang W, Li X and Fan Y: N6-methyladenosine modification of circCUX1 confers radio-resistance of hypopharyngeal squamous cell carcinoma through caspase1 pathway. Cell Death Dis. 12:2982021. View Article : Google Scholar

130 

Ji F, Lu Y, Chen S, Yu Y, Lin X, Zhu Y and Luo X: IGF2BP2-modified circular RNA circARHGAP12 promotes cervical cancer progression by interacting m(6)A/FOXM1 manner. Cell Death Discov. 7:2152021. View Article : Google Scholar : PubMed/NCBI

131 

Cheng X, Yang H, Chen Y, Zeng Z, Liu Y, Zhou X, Zhang C, Xie A and Wang G: METTL3-mediated m6A modification of circGLIS3 promotes prostate cancer progression and represents a potential target for ARSI therapy. Cell Mol Biol Lett. 29:1092024. View Article : Google Scholar

132 

Wu Q, Yin X, Zhao W, Xu W and Chen L: Molecular mechanism of m6A methylation of circDLC1 mediated by RNA methyltransferase METTL3 in the malignant proliferation of glioma cells. Cell Death Discov. 8:2292022. View Article : Google Scholar

133 

Shu X, Yi J, Li J, Ying Y, Tang Y, Chen Z, Wang J, Zhang F, Lu D, Wu Y, et al: N6-methyladenosine-modified circRPS6KC1 regulated cellular senescence in prostate cancer via FOXM1/PCNA axis. Cell Signal. 125:1115102025. View Article : Google Scholar

134 

Tian Q, Mu Q, Liu S, Huang K, Tang Y, Zhang P, Zhao J and Shu C: m6A-modified circASXL1 promotes proliferation and migration of ovarian cancer through the miR-320d/RACGAP1 axis. Carcinogenesis. 44:859–870. 2023. View Article : Google Scholar : PubMed/NCBI

135 

Wu G, Hou Q, Liu Z, Pu Z and Wu L: N6-methyladenosine-modified circ_0006168 promotes epithelial mesenchymal transition via miR-384/STAT3/Snail axis in esophageal squamous cell carcinoma. J Cancer. 15:4939–4954. 2024. View Article : Google Scholar :

136 

Zhang Y, Yang H, Long Y, Zhang Y, Chen R, Shi J and Chen J: circRNA N6-methyladenosine methylation in preeclampsia and the potential role of N6-methyladenosine-modified circPAPPA2 in trophoblast invasion. Sci Rep. 11:243572021. View Article : Google Scholar : PubMed/NCBI

137 

Liang L, Zhu Y, Li J, Zeng J and Wu L: ALKBH5-mediated m6A modification of circCCDC134 facilitates cervical cancer metastasis by enhancing HIF1A transcription. J Exp Clin Cancer Res. 41:2612022. View Article : Google Scholar : PubMed/NCBI

138 

Huang R, Sun M, Wang W, Yu X and Liu F: YTHDF2 alleviates microglia activation via promoting circHIPK2 degradation. J Neuroimmunol. 387:5782652024. View Article : Google Scholar

139 

Shao Y, Liu Z, Song X, Sun R, Zhou Y, Zhang D, Sun H, Huang J, Wu C, Gu W, et al: ALKBH5/YTHDF2-mediated m6A modification of circAFF2 enhances radiosensitivity of colorectal cancer by inhibiting Cullin neddylation. Clin Transl Med. 13:e13182023. View Article : Google Scholar : PubMed/NCBI

140 

Wang D, Guan H and Xia Y: YTHDC1 maintains trophoblasts function by promoting degradation of m6A-modified circMPP1. Biochem Pharmacol. 210:1154562023. View Article : Google Scholar : PubMed/NCBI

141 

Yu J, Li W, Hou GJ, Sun DP, Yang Y, Yuan SX, Dai ZH, Yin HZ, Sun SH, Huang G, et al: Circular RNA cFAM210A, degradable by HBx, inhibits HCC tumorigenesis by suppressing YBX1 transactivation. Exp Mol Med. 55:2390–2401. 2023. View Article : Google Scholar : PubMed/NCBI

142 

Chen X, Zhu S, Li HD, Wang JN, Sun LJ, Xu JJ, Hui YR, Li XF, Li LY, Zhao YX, et al: N(6)-methyladenosine-modified circIRF2, identified by YTHDF2, suppresses liver fibrosis via facilitating FOXO3 nuclear translocation. Int J Biol Macromol. 248:1258112023. View Article : Google Scholar : PubMed/NCBI

143 

Chen RX, Chen X, Xia LP, Zhang JX, Pan ZZ, Ma XD, Han K, Chen JW, Judde JG, Deas O, et al: N6-methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun. 10:46952019. View Article : Google Scholar

144 

Ding P, Wu H, Wu J, Li T, He J, Ju Y, Liu Y, Li F, Deng H, Gu R, et al: N6-methyladenosine modified circPAK2 promotes lymph node metastasis via targeting IGF2BPs/VEGFA signaling in gastric cancer. Oncogene. 43:2548–2563. 2024. View Article : Google Scholar

145 

Zhou M, Liu Y, Xu H, Chen X, Zheng N, Duan Z, Ge Y, Li D, Lin T, Zeng R, et al: YTHDC1-Modified m6A methylation of Hsa_circ_0102678 promotes keratinocyte inflammation induced by cutibacterium acnes biofilm through regulating miR-146a/TRAF6 and IRAK1 axis. J Innate Immun. 15:822–835. 2023. View Article : Google Scholar : PubMed/NCBI

146 

Zeng W, Zhu JF, Guo J, Huang GJ, Ai LS, Zeng Y and Liao WJ: m6A-modified circFNDC3B inhibits colorectal cancer stemness and metastasis via RNF41-dependent ASB6 degradation. Cell Death Dis. 13:10082022. View Article : Google Scholar

147 

Lin H, Cui Z, Tiange E, Xu H, Wang D, Wang P, Ruan X, Liu L and Xue Y: M6A-methylated circPOLR2B forms an R-loop and regulates the biological behavior of glioma stem cells through positive feedback loops. Cell Death Dis. 15:5542024. View Article : Google Scholar : PubMed/NCBI

148 

Song M, Yao H, Sun Z, Chen D, Xu X, Long G, Wu L and Hu W: METTL3/YTHDC1-medicated m6A modification of circRNA3634 regulates the proliferation and differentiation of antler chondrocytes by miR-124486-5-MAPK1 axis. Cell Mol Biol Lett. 28:1012023. View Article : Google Scholar : PubMed/NCBI

149 

Rong D, Wu F, Lu C, Sun G, Shi X, Chen X, Dai Y, Zhong W, Hao X, Zhou J, et al: m6A modification of circHPS5 and hepatocellular carcinoma progression through HMGA2 expression. Mol Ther Nucleic Acids. 26:637–648. 2021. View Article : Google Scholar : PubMed/NCBI

150 

Zhang R, Yang R, Huang Z, Xu X, Lv S, Guan X, Li H and Wu J: METTL3/YTHDC1-mediated upregulation of LINC00294 promotes hepatocellular carcinoma progression. Heliyon. 9:e225952023. View Article : Google Scholar : PubMed/NCBI

151 

Ouyang L, Sun MM, Zhou PS, Ren YW, Liu XY, Wei WY, Song ZS, Lu K and Yang LX: LncRNA FOXD1-AS1 regulates pancreatic cancer stem cell properties and 5-FU resistance by regulating the miR-570-3p/SPP1 axis as a ceRNA. Cancer Cell Int. 24:42024. View Article : Google Scholar : PubMed/NCBI

152 

Shu G, Zhao Z, Zhao T, Deng C, Zhu J, Han Y, Chen M, Jing J, Bai G, Li D, et al: N(6)-methyladenosine modification of circMARK2 enhances cytoplasmic export and stabilizes LIN28B, contributing to the progression of Wilms tumor. J Exp Clin Cancer Res. 43:1912024. View Article : Google Scholar : PubMed/NCBI

153 

Li H, Tang Y, Ruan X, Zhang J, Liu H, Yu S, Chen H, Yang H, Zhang K and Chen K: N6-methyladenosine-modified circTEAD1 stabilizes Yap1 mRNA to promote chordoma tumorigenesis. Clin Transl Med. 14:e16582024. View Article : Google Scholar : PubMed/NCBI

154 

Zheng Z, Zeng X, Zhu Y, Leng M, Zhang Z, Wang Q, Liu X, Zeng S, Xiao Y, Hu C, et al: CircPPAP2B controls metastasis of clear cell renal cell carcinoma via HNRNPC-dependent alternative splicing and targeting the miR-182-5p/CYP1B1 axis. Mol Cancer. 23:42024. View Article : Google Scholar : PubMed/NCBI

155 

Li J, Cao H, Yang J and Wang B: IGF2BP2-m6A-circMMP9 axis recruits ETS1 to promote TRIM59 transcription in laryngeal squamous cell carcinoma. Sci Rep. 14:30142024. View Article : Google Scholar : PubMed/NCBI

156 

Mamontova V, Trifault B, Gribling-Burrer AS, Bohn P, Boten L, Preckwinkel P, Gallant P, Solvie D, Ade CP, Papadopoulos D, et al: NEAT1 promotes genome stability via m(6)A methylation-dependent regulation of CHD4. Genes Dev. 38:915–930. 2024. View Article : Google Scholar : PubMed/NCBI

157 

Jiang T, Qi J, Xue Z, Liu B, Liu J, Hu Q, Li Y, Ren J, Song H, Xu Y, et al: The m6A modification mediated-lncRNA POU6F2-AS1 reprograms fatty acid metabolism and facilitates the growth of colorectal cancer via upregulation of FASN. Mol Cancer. 23:552024. View Article : Google Scholar

158 

Olazagoitia-Garmendia A, Rojas-Márquez H, SebastiandelaCruz M, Agirre-Lizaso A, Ochoa A, Mendoza-Gomez LM, Perugorria MJ, Bujanda L, Madrigal AH, Santin I and Castellanos-Rubio A: m6A methylated long noncoding RNA LOC339803 regulates intestinal inflammatory response. Adv Sci (Weinh). 11:e23079282024. View Article : Google Scholar

159 

Vaid R, Thombare K, Mendez A, Burgos-Panadero R, Djos A, Jachimowicz D, Lundberg KI, Bartenhagen C, Kumar N, Tümmler C, et al: METTL3 drives telomere targeting of TERRA lncRNA through m6A-dependent R-loop formation: A therapeutic target for ALT-positive neuroblastoma. Nucleic Acids Res. 52:2648–2671. 2024. View Article : Google Scholar : PubMed/NCBI

160 

Liu R, Zhong Y, Chen R, Chu C, Liu G, Zhou Y, Huang Y, Fang Z and Liu H: m6A reader hnRNPA2B1 drives multiple myeloma osteolytic bone disease. Theranostics. 12:7760–7774. 2022. View Article : Google Scholar :

161 

Hu Z, Chen G, Zhao Y, Gao H, Li L, Yin Y, Jiang J, Wang L, Mang Y, Gao Y, et al: Exosome-derived circCCAR1 promotes CD8 + T-cell dysfunction and anti-PD1 resistance in hepatocellular carcinoma. Mol Cancer. 22:552023. View Article : Google Scholar

162 

Yu J, Gao Y, Liu F, Zhang Y, Li J, Ding L, Ren S, Yang J, Jiao J, Feng G, et al: m6A-modified exosome-derived circHIF1α binding to KH domain of IGF2BP3 mediates DNA damage and arrests G1/S transition phase to resists bacterial infection in bacteremia. J Nanobiotechnology. 22:6542024. View Article : Google Scholar

163 

Wang Z, He J, Bach DH, Huang YH, Li Z, Liu H, Lin P and Yang J: Induction of m6A methylation in adipocyte exosomal LncRNAs mediates myeloma drug resistance. J Exp Clin Cancer Res. 41:42022. View Article : Google Scholar

164 

Chen J, Zhou Y, Wu M, Yuan Y and Wu W: m6A modification mediates exosomal LINC00657 to trigger breast cancer progression via inducing macrophage M2 polarization. Clin Breast Cancer. 23:546–560. 2023. View Article : Google Scholar

165 

He Z, Zhong Y, Regmi P, Lv T, Ma W, Wang J, Liu F, Yang S, Zhong Y, Zhou R, et al: Exosomal long non-coding RNA TRPM2-AS promotes angiogenesis in gallbladder cancer through interacting with PABPC1 to activate NOTCH1 signaling pathway. Mol Cancer. 23:652024. View Article : Google Scholar : PubMed/NCBI

166 

Tuersun H, Liu L, Zhang J, Maimaitizunong R, Tang X and Li H: m6A reading protein RBMX as a biomarker for prognosis and tumor progression in esophageal cancer. Transl Cancer Res. 12:2319–2335. 2023. View Article : Google Scholar : PubMed/NCBI

167 

Yang Y, Ren S, Xue J, Dong W, He W, Luo J, Li X, Xu H, Zheng Z, Wang X, et al: DeSUMOylation of RBMX regulates exosomal sorting of cargo to promote renal tubulointerstitial fibrosis in diabetic kidney disease. J Adv Res. 74:175–189. 2024. View Article : Google Scholar : PubMed/NCBI

168 

Wu Z, Zuo X, Zhang W, Li Y, Gui R, Leng J, Shen H, Pan B, Fan L, Li J and Jin H: m6A-Modified circTET2 interacting with HNRNPC regulates fatty acid oxidation to promote the proliferation of chronic lymphocytic leukemia. Adv Sci (Weinh). 10:e23048952023. View Article : Google Scholar : PubMed/NCBI

169 

Liu J, Shao Y and Li C: YTHDC1/CRM1 facilitates m6A-modified circRNA388 nuclear export to induce coelomocyte autophagy via the miR-2008/ULK axis in apostichopus japonicus. J Immunol. 212:1319–1333. 2024. View Article : Google Scholar : PubMed/NCBI

170 

Wang L, Wang H, Liu T, Zhou X, Kuai S, Ji Z and Shen H: METTL3-mediated m(6)A modification of circCSDE1 promote Coxsackievirus replication by regulating the miR-891b/BAG3 axis. Int Immunopharmacol. 159:1149052025. View Article : Google Scholar : PubMed/NCBI

171 

Wu A, Hu Y, Xu Y, Xu J, Wang X, Cai A, Liu R, Chen L and Wang F: Methyltransferase-like 3-mediated m6A methylation of Hsa_circ_0058493 accelerates hepatocellular carcinoma progression by binding to YTH domain-containing protein 1. Front Cell Dev Biol. 9:7625882021. View Article : Google Scholar : PubMed/NCBI

172 

Yang L, Chen Y, Liu N, Lu Y, Ma W, Yang Z, Gan W and Li D: CircMET promotes tumor proliferation by enhancing CDKN2A mRNA decay and upregulating SMAD3. Mol Cancer. 21:232022. View Article : Google Scholar : PubMed/NCBI

173 

Yang Y, Jiang C, Zeng J, Guo X, Chen M and Wu B: hsa_ circ_0001599 promotes odontogenic differentiation of human dental pulp stem cells by increasing ITGA2 expression and stability. Commun Biol. 8:742025. View Article : Google Scholar

174 

He T, Zhang Q, Xu P, Tao W, Lin F, Liu R, Li M, Duan X, Cai C, Gu D, et al: Extracellular vesicle-circEHD2 promotes the progression of renal cell carcinoma by activating cancer-associated fibroblasts. Mol Cancer. 22:1172023. View Article : Google Scholar : PubMed/NCBI

175 

Diao X, Guo C, Zheng H, Zhao K, Luo Y, An M, Lin Y, Chen J, Li Y, Li Y, et al: SUMOylation-triggered ALIX activation modulates extracellular vesicles circTLCD4-RWDD3 to promote lymphatic metastasis of non-small cell lung cancer. Signal Transduct Target Ther. 8:4262023. View Article : Google Scholar : PubMed/NCBI

176 

Yao Y, Chen C, Wang J, Xuan H, Chen X, Li Z, Yang F, Wang B, Lin S, Li S, et al: Circular RNA circATP9A promotes non-small cell lung cancer progression by interacting with HuR and by promoting extracellular vesicles-mediated macrophage M2 polarization. J Exp Clin Cancer Res. 42:3302023. View Article : Google Scholar : PubMed/NCBI

177 

Pan Z, Zhao R, Li B, Qi Y, Qiu W, Guo Q, Zhang S, Zhao S, Xu H, Li M, et al: EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 21:162022. View Article : Google Scholar : PubMed/NCBI

178 

Chen S, Wang Y, Zhang J, Liu B, Liu W, Cao G, Li R, Li H, Zhai N, Song X, et al: YTHDC1 phase separation drives the nuclear export of m(6)A-modified lncNONMMUT062668.2 through the transport complex SRSF3-ALYREF-XPO5 to aggravate pulmonary fibrosis. Cell Death Dis. 16:2792025. View Article : Google Scholar : PubMed/NCBI

179 

Li T, Tian L, Cao J and Liu M: Cancer-associated fibroblasts secret extracellular vesicles to support cell proliferation and epithelial-mesenchymal transition in laryngeal squamous cell carcinoma. Mol Cell Probes. 72:1019342023. View Article : Google Scholar : PubMed/NCBI

180 

Wang Y, Tian X, Wang Z, Liu D, Zhao X, Sun X, Tu Z, Li Z, Zhao Y, Zheng S and Yao J: A novel peptide encoded by circ-SLC9A6 promotes lipid dyshomeostasis through the regulation of H4K16ac-mediated CD36 transcription in NAFLD. Clin Transl Med. 14:e18012024. View Article : Google Scholar : PubMed/NCBI

181 

Zeng K, Peng J, Xing Y, Zhang L, Zeng P, Li W, Zhang W, Pan Z, Zhou C and Lin J: A positive feedback circuit driven by m6A-modified circular RNA facilitates colorectal cancer liver metastasis. Mol Cancer. 22:2022023. View Article : Google Scholar

182 

Zheng W, Wang L, Geng S and Xu T: CircYthdc2 generates polypeptides through two translation strategies to facilitate virus escape. Cell Mol Life Sci. 81:912024. View Article : Google Scholar : PubMed/NCBI

183 

Li K, Peng ZY, Wang R, Li X, Du N, Liu DP, Zhang J, Zhang YF, Ma L, Sun Y, et al: Enhancement of TKI sensitivity in lung adenocarcinoma through m6A-dependent translational repression of Wnt signaling by circ-FBXW7. Mol Cancer. 22:1032023. View Article : Google Scholar : PubMed/NCBI

184 

Zheng W, Wang L, Geng S, Yang L, Lv X, Xin S and Xu T: CircMIB2 therapy can effectively treat pathogenic infection by encoding a novel protein. Cell Death Dis. 14:5782023. View Article : Google Scholar : PubMed/NCBI

185 

Tang C, Xie Y, Yu T, Liu N, Wang Z, Woolsey RJ, Tang Y, Zhang X, Qin W, Zhang Y, et al: m6A-dependent biogenesis of circular RNAs in male germ cells. Cell Res. 30:211–228. 2020. View Article : Google Scholar : PubMed/NCBI

186 

Pan Z, Cai J, Lin J, Zhou H, Peng J, Liang J, Xia L, Yin Q, Zou B, Zheng J, et al: A novel protein encoded by circFNDC3B inhibits tumor progression and EMT through regulating Snail in colon cancer. Mol Cancer. 19:712020. View Article : Google Scholar : PubMed/NCBI

187 

Xiong L, Gong Y, Liu H, Huang L, Zeng Z, Zheng X, Li W, Liang Z and Kang L: circGlis3 promotes β-cell dysfunction by binding to heterogeneous nuclear ribonucleoprotein F and encoding Glis3-348aa protein. iScience. 27:1086802024. View Article : Google Scholar

188 

Sun W, Zhou S, Peng L, Liu Y, Cheng D, Wang Y and Ni C: CircZNF609 regulates pulmonary fibrosis via miR-145-5p/KLF4 axis and its translation function. Cell Mol Biol Lett. 28:1052023. View Article : Google Scholar :

189 

Wu S, Zhang L, Deng J, Guo B, Li F, Wang Y, Wu R, Zhang S, Lu J and Zhou Y: A novel micropeptide encoded by Y-linked LINC00278 links cigarette smoking and AR signaling in male esophageal squamous cell carcinoma. Cancer Res. 80:2790–2803. 2020. View Article : Google Scholar : PubMed/NCBI

190 

Pei H, Dai Y, Yu Y, Tang J, Cao Z, Zhang Y, Li B, Nie J, Hei TK and Zhou G: The tumorigenic effect of lncRNA AFAP1-AS1 is mediated by translated peptide ATMLP under the control of m6A methylation. Adv Sci (Weinh). 10:e23003142023. View Article : Google Scholar

191 

Shen G, Li F, Wang Y, Huang Y, Aizezi G, Yuan J, Ma C and Lin C: New insights on the interaction between m6A modification and non-coding RNA in cervical squamous cell carcinoma. World J Surg Oncol. 21:252023. View Article : Google Scholar

192 

Wu C, Cui J, Huo Y, Shi L and Wang C: Alternative splicing of HOXB-AS3 underlie the promoting effect of nuclear m6A reader YTHDC1 on the self-renewal of leukemic stem cells in acute myeloid leukemia. Int J Biol Macromol. 237:1239902023. View Article : Google Scholar

193 

Leng F, Miu YY, Zhang Y, Luo H, Lu XL, Cheng H and Zheng ZG: A micro-peptide encoded by HOXB-AS3 promotes the proliferation and viability of oral squamous cell carcinoma cell lines by directly binding with IGF2BP2 to stabilize c-Myc. Oncol Lett. 22:6972021. View Article : Google Scholar : PubMed/NCBI

194 

Lin M, Zhou X, Yang Y, Xie P, Li Q, He C, Lin Q, Wei X and Ding Y: A peptide encoded by lncRNA HOXB-AS3 promotes cigarette smoke-induced inflammation in bronchial epithelial cells via EZH2-Mediated H3K27me3 modification. Int J Chron Obstruct Pulmon Dis. 20:1543–1553. 2025. View Article : Google Scholar : PubMed/NCBI

195 

Ge Q, Jia D, Cen D, Qi Y, Shi C, Li J, Sang L, Yang LJ, He J, Lin A, et al: Micropeptide ASAP encoded by LINC00467 promotes colorectal cancer progression by directly modulating ATP synthase activity. J Clin Invest. 131:e1529112021. View Article : Google Scholar : PubMed/NCBI

196 

Zhang M, Cai R, Liu J, Wang Y, He S, Wang Q, Song X, Wu J and Zhao J: Multi-omics integration analysis reveals the role of N6-methyladenosine in lncRNA translation during glioma stem cell differentiation. Brief Funct Genomics. 23:806–815. 2024. View Article : Google Scholar : PubMed/NCBI

197 

Zhong J, Wu X, Gao Y, Chen J, Zhang M, Zhou H, Yang J, Xiao F, Yang X, Huang N, et al: Circular RNA encoded MET variant promotes glioblastoma tumorigenesis. Nat Commun. 14:44672023. View Article : Google Scholar : PubMed/NCBI

198 

Wang T, Liu Z, She Y, Deng J, Zhong Y, Zhao M, Li S, Xie D, Sun X, Hu X and Chen C: A novel protein encoded by circASK1 ameliorates gefitinib resistance in lung adenocarcinoma by competitively activating ASK1-dependent apoptosis. Cancer Lett. 520:321–331. 2021. View Article : Google Scholar : PubMed/NCBI

199 

Lu J, Ru J, Chen Y, Ling Z, Liu H, Ding B, Jiang Y, Ma J, Zhang D, Ge J, et al: N6 -methyladenosine-modified circSTX6 promotes hepatocellular carcinoma progression by regulating the HNRNPD/ATF3 axis and encoding a 144 amino acid polypeptide. Clin Transl Med. 13:e14512023. View Article : Google Scholar

200 

Li Y, Chen B, Zhao J, Li Q, Chen S, Guo T, Li Y, Lai H, Chen Z, Meng Z, et al: HNRNPL circularizes ARHGAP35 to produce an oncogenic protein. Adv Sci (Weinh). 8:20017012021. View Article : Google Scholar : PubMed/NCBI

201 

Lan T, Gao F, Cai Y, Lv Y, Zhu J, Liu H, Xie S, Wan H, He H, Xie K, et al: The protein circPETH-147aa regulates metabolic reprogramming in hepatocellular carcinoma cells to remodel immunosuppressive microenvironment. Nat Commun. 16:3332025. View Article : Google Scholar : PubMed/NCBI

202 

Zhong S, Xu H, Wang D, Yang S, Li H, Zhang H, Feng J and Zhou S: circNFIB decreases synthesis of arachidonic acid and inhibits breast tumor growth and metastasis. Eur J Pharmacol. 963:1762212024. View Article : Google Scholar

203 

Wei C, Peng D, Jing B, Wang B, Li Z, Yu R, Zhang S, Cai J, Zhang Z, Zhang J and Han L: A novel protein SPECC1-415aa encoded by N6-methyladenosine modified circSPECC1 regulates the sensitivity of glioblastoma to TMZ. Cell Mol Biol Lett. 29:1272024. View Article : Google Scholar : PubMed/NCBI

204 

Zhang Y, Liu Z, Zhong Z, Ji Y, Guo H, Wang W and Chen C: A tumor suppressor protein encoded by circKEAP1 inhibits osteosarcoma cell stemness and metastasis by promoting vimentin proteasome degradation and activating anti-tumor immunity. J Exp Clin Cancer Res. 43:522024. View Article : Google Scholar : PubMed/NCBI

205 

Li M, Ding W, Fang X, Wang Y, Wang P, Ye L, Miao S, Song L, Ao X, Li Q and Wang J: Novel truncated peptide derived from circCDYL exacerbates cardiac hypertrophy. Circ Res. 136:e94–e112. 2025. View Article : Google Scholar : PubMed/NCBI

206 

Zhou B, Yu G, Zhao M, Li Y, Li J, Xiang Y, Tong L, Chu X, Wang C and Song Y: The lncRNA LINC00339-encoded peptide promotes trophoblast adhesion to endometrial cells via MAPK and PI3K-Akt signaling pathways. J Assist Reprod Genet. 41:493–504. 2024. View Article : Google Scholar :

207 

Zhu HX, Lu WJ, Zhu WP and Yu S: Comprehensive analysis of N6 -methyladenosine-related long non-coding RNAs for prognosis prediction in liver hepatocellular carcinoma. J Clin Lab Anal. 35:e240712021. View Article : Google Scholar

208 

Xiao W, Halabi R, Lin CH, Nazim M, Yeom KH and Black DL: The lncRNA Malat1 is trafficked to the cytoplasm as a localized mRNA encoding a small peptide in neurons. Genes Dev. 38:294–307. 2024. View Article : Google Scholar :

209 

Ming Y, Deng Z, Tian X, Jia Y, Ning M and Cheng S: m6A methyltransferase METTL3 reduces hippocampal neuron apoptosis in a mouse model of autism through the MALAT1/SFRP2/Wnt/β-catenin axis. Psychiatry Investig. 19:771–787. 2022. View Article : Google Scholar : PubMed/NCBI

210 

Zheng W, Guo Y, Zhang G, Bai J, Song Y, Song X, Zhu Q, Bao X, Wu G and Zhang C: Peptide encoded by lncRNA BVES-AS1 promotes cell viability, migration, and invasion in colorectal cancer cells via the SRC/mTOR signaling pathway. PLoS One. 18:e02871332023. View Article : Google Scholar : PubMed/NCBI

211 

Wu J, Cai Y, Zhao G and Li M: A ten N6-methyladenosine-related long non-coding RNAs signature predicts prognosis of triple-negative breast cancer. J Clin Lab Anal. 35:e237792021. View Article : Google Scholar : PubMed/NCBI

212 

Min XL, Lin SX, Zhao XH, Zhao Q, Li YF, Li XH, Liu XY, Cao Y, Sun YL and Zeng Y: Mechanisms of METTL14-mediated m6A modification in promoting iron overload-induced lipid peroxidative damage in vascular endothelial cells to aggravate atherosclerosis. J Biochem Mol Toxicol. 38:e700662024. View Article : Google Scholar : PubMed/NCBI

213 

Shen C, Chen X, Lin Y and Yang Y: Hypoxia triggers cardiomyocyte apoptosis via regulating the m6A methylation-mediated LncMIAT/miR-708-5p/p53 axis. Heliyon. 10:e324552024. View Article : Google Scholar

214 

Fang X, Ao X, Xiao D, Wang Y, Jia Y, Wang P, Li M and Wang J: Circular RNA-circPan3 attenuates cardiac hypertrophy via miR-320-3p/HSP20 axis. Cell Mol Biol Lett. 29:32024. View Article : Google Scholar : PubMed/NCBI

215 

Zhang HR, Ma GQ, Lv HQ, Feng YT and Peng YJ: Electroacupuncture alleviates cerebral ischemia-reperfusion injury by regulating the S1PR2/TLR4/NLRP3 signaling pathway via m6A methylation of lncRNA H19. Curr Neurovasc Res. 21:64–73. 2024. View Article : Google Scholar : PubMed/NCBI

216 

Yu P, Wang J, Xu GE, Zhao X, Cui X, Feng J, Sun J, Wang T, Spanos M, Lehmann HI, et al: RNA m(6)A-Regulated circ-ZNF609 suppression ameliorates doxorubicin-induced cardiotoxicity by upregulating FTO. JACC Basic Transl Sci. 8:677–698. 2023. View Article : Google Scholar : PubMed/NCBI

217 

Yuan J, Liu Y, Zhou L, Xue Y, Lu Z and Gan J: YTHDC2-mediated circYTHDC2 N6-methyladenosine modification promotes vascular smooth muscle Cells dysfunction through inhibiting ten-eleven translocation 2. Front Cardiovasc Med. 8:6862932021. View Article : Google Scholar : PubMed/NCBI

218 

Tian Y, Xiao YH, Sun C, Liu B and Sun F: N6-Methyladenosine methyltransferase METTL3 alleviates diabetes-induced testicular damage through modulating TUG1/Clusterin axis. Diabetes Metab J. 47:287–300. 2023. View Article : Google Scholar : PubMed/NCBI

219 

Fu S, Zhou Q, Peng X, Hu Y, Xiong J and Liu F: METTL3/YTHDC1 mediates up-regulation of lncRNA OGRU in an m6A-dependent manner involving in oxidative stress and inflammation of HG-induced Müller cells. Immunol Lett. 272:1069722025. View Article : Google Scholar

220 

Li C, Su F, Liang Z, Zhang L, Liu F, Fan W and Li Z: Macrophage M1 regulatory diabetic nephropathy is mediated by m6A methylation modification of lncRNA expression. Mol Immunol. 144:16–25. 2022. View Article : Google Scholar : PubMed/NCBI

221 

Guo C, Yu M, Liu J, Jia Z, Liu H and Zhao S: Molecular mechanism of Wilms tumour 1-associated protein in diabetes-related dry eye disease by mediating m6A methylation modification of lncRNA NEAT1. J Drug Target. 32:200–212. 2024. View Article : Google Scholar

222 

Zheng M, Cun D, He H, Xie X, Lei H, Fu W, Tai W and Yang J: Expression profile and N6-methyadenosine modification of circular RNA analysis in MAFLD. BMC Gastroenterol. 25:1622025. View Article : Google Scholar : PubMed/NCBI

223 

Zhang C, Jian H, Shang S, Lu L, Lou Y, Kang Y, Bai H, Fu Z, Lv Y, Kong X, et al: Crosstalk between m6A mRNAs and m6A circRNAs and the time-specific biogenesis of m6A circRNAs after OGD/R in primary neurons. Epigenetics. 18:21815752023. View Article : Google Scholar : PubMed/NCBI

224 

Zhang X, Yang S, Han S, Sun Y, Han M, Zheng X, Li F, Wei Y, Wang Y and Bi J: Differential methylation of circRNA m6A in an APP/PS1 Alzheimer's disease mouse model. Mol Med Rep. 27:552023. View Article : Google Scholar : PubMed/NCBI

225 

Liu X, Li Z, Tong J, Wu F, Jin H and Liu K: Characterization of the expressions and m6A methylation modification patterns of mRNAs and lncRNAs in a spinal cord injury rat model. Mol Neurobiol. 62:806–818. 2025. View Article : Google Scholar :

226 

Atrian F, Ramirez P, De Mange J, Ramirez P, Atrian F, Morita M and Frost B: m6A-dependent circular RNA formation mediates tau-induced neurotoxicity. bioRxiv. 23:e142452024.

227 

Zhang H, Xu X, Li X, Zeng C and Peng Y: Electroacupuncture serum alleviates Ogd/R-induced astrocyte damage by regulating the AQP4 Via m6A methylation of lncRNA MALAT1. Neurochem Res. 50:1392025. View Article : Google Scholar : PubMed/NCBI

228 

Sun H, Liu F, Song X, Sun R, Zhang M, Huang J, Gu W and Shao Y: m6A-modified circCREBBP enhances radiosensitivity of esophageal squamous cell carcinoma by reducing the stability of MYC through interaction with IGF2BP3. Int J Biol Macromol. 286:1385342025. View Article : Google Scholar

229 

Wang F, Wen J, Liu J, Xin L, Fang Y, Sun Y and He M: Demethylase FTO mediates m6A modification of ENST00000619282 to promote apoptosis escape in rheumatoid arthritis and the intervention effect of Xinfeng Capsule. Front Immunol. 16:15567642025. View Article : Google Scholar : PubMed/NCBI

230 

Canaani J, Danylesko I, Shemtov N, Zlotnick M, Lozinsky K, Benjamini O, Yerushalmi R, Nagar M, Dor C, Shimoni A, et al: A phase II study of bisantrene in patients with relapsed/refractory acute myeloid leukemia. Eur J Haematol. 106:260–266. 2021. View Article : Google Scholar

231 

Pomaville M, Chennakesavalu M, Wang P, Jiang Z, Sun HL, Ren P, Borchert R, Gupta V, Ye C, Ge R, et al: Small-molecule inhibition of the METTL3/METTL14 complex suppresses neuroblastoma tumor growth and promotes differentiation. Cell Rep. 43:1141652024. View Article : Google Scholar : PubMed/NCBI

232 

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

233 

Lee JH, Choi N, Kim S, Jin MS, Shen H and Kim YC: Eltrombopag as an allosteric inhibitor of the METTL3-14 complex affecting the m6A methylation of RNA in acute myeloid leukemia cells. Pharmaceuticals (Basel). 15:4402022. View Article : Google Scholar

234 

Wu Z, Smith AR, Qian Z and Zheng G: Patent landscape of small molecule inhibitors of METTL3 (2020-present). Expert Opin Ther Pat. 26:1–16. 2024.

235 

Wu J, Pang R, Li M, Chen B, Huang J and Zhu Y: m6A-Induced LncRNA MEG3 suppresses the proliferation, migration and invasion of hepatocellular carcinoma cell through miR-544b/BTG2 signaling. Onco Targets Ther. 14:3745–3755. 2021. View Article : Google Scholar : PubMed/NCBI

236 

Li P, Mi Q, Yan S, Xie Y, Cui Z, Zhang S, Wang Y, Gao H, Wang Y, Li J, et al: Characterization of circSCL38A1 as a novel oncogene in bladder cancer via targeting ILF3/TGF-β2 signaling axis. Cell Death Dis. 14:592023. View Article : Google Scholar

237 

Liu Q, Zhou BM, Wang LJ and Zhang CY: Construction of a hierarchical DNA circuit for single-molecule profiling of locus-specific N6-methyladenosine-MALAT1 in clinical tissues. Biosens Bioelectron. 274:1171982025. View Article : Google Scholar

238 

Zhang J, Liu T, Wang Y, Yan X, Li Y, Xu F and Zhang R: Dynamic alterations of the transcriptome-wide m6A methylome in cytogenetically normal acute myeloid leukaemia during initial diagnosis and relapse. Genomics. 115:1107252023. View Article : Google Scholar

Related Articles

Journal Cover

October-2025
Volume 56 Issue 4

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
Yi Q, Liao Y, Sun W, Li J, Yang D, Shang H and Sun W: m6A modification of non‑coding RNA: Mechanisms, functions and potential values in human diseases (Review). Int J Mol Med 56: 164, 2025.
APA
Yi, Q., Liao, Y., Sun, W., Li, J., Yang, D., Shang, H., & Sun, W. (2025). m6A modification of non‑coding RNA: Mechanisms, functions and potential values in human diseases (Review). International Journal of Molecular Medicine, 56, 164. https://doi.org/10.3892/ijmm.2025.5605
MLA
Yi, Q., Liao, Y., Sun, W., Li, J., Yang, D., Shang, H., Sun, W."m6A modification of non‑coding RNA: Mechanisms, functions and potential values in human diseases (Review)". International Journal of Molecular Medicine 56.4 (2025): 164.
Chicago
Yi, Q., Liao, Y., Sun, W., Li, J., Yang, D., Shang, H., Sun, W."m6A modification of non‑coding RNA: Mechanisms, functions and potential values in human diseases (Review)". International Journal of Molecular Medicine 56, no. 4 (2025): 164. https://doi.org/10.3892/ijmm.2025.5605