Open Access

Interactions of circRNAs with methylation: An important aspect of circRNA biogenesis and function (Review)

  • Authors:
    • Chunlei Zhang
    • Han Cui
    • Chuang Huang
    • Feiyan Kong
    • Qi Yang
    • Pengcheng Miao
    • Zhigang Cao
    • Weijun Zhang
    • Dehui Chang
  • View Affiliations

  • Published online on: March 16, 2022     https://doi.org/10.3892/mmr.2022.12685
  • Article Number: 169
  • Copyright: © Zhang 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

Circular RNA (circRNA) molecules are noncoding RNAs with unique circular covalently closed structures that contribute to gene expression regulation, protein translation and act as microRNA sponges. circRNAs also have important roles in human disease, particularly tumorigenesis and antitumor processes. Methylation is an epigenetic modification that regulates the expression and roles of DNA and coding RNA and their interactions, as well as of noncoding RNA molecules. Previous studies have focused on the effects of methylation modification on circRNA expression, transport, stability, translation and degradation of circRNAs, as well as how circRNA methylation occurs and the influence of circRNAs on methylation modification processes. circRNA and methylation can also regulate disease pathogenesis via these interactions. In the present study, we define the relationship between circRNAs and methylation, as well as the functions and mechanisms of their interactions during disease progression.

Introduction

Gene methylation is an important epigenetic modification that involves both DNA and RNA molecules. DNA methylation mainly affects gene expression at the transcriptional level (1), whereas RNA methylation primarily regulates gene expression at the post-transcriptional level by influencing RNA stability, maturation, localization, transportation, transcription and translation (2,3). Overall, methylation modification affects numerous cellular physiological behaviors, such as neurodevelopment, immunoregulation, and cellular differentiation, and contributes to the development of various diseases (4,5).

Circular RNA (circRNA/circ) molecules are novel noncoding RNAs formed by reverse splicing of their corresponding host genes and can therefore be regulated via methylation (6). Furthermore, circRNAs can regulate gene expression and bind proteins, among other functions, and may modulate gene expression by controlling the methylation modification process (7). Therefore, fully understanding the effects of methylation modifications on the expression and function of circRNAs and an investigation into the regulation of methylation via circRNAs, are essential to determining the molecular mechanisms underlying circRNA activity and disease progression. The aim of the present review was to define the relationship between circRNAs and methylation, as well as the functions and mechanisms of their interactions during disease progression.

Literature search strategy

The PubMed database (pubmed.ncbi.nlm.nih.gov/) was searched using the following terms in the title or abstract: i) circRNA or circular RNA and ii) methylation or N6-methyladenosine (m6A) or m6A. The inclusion criteria for the articles selected were that they must have been published from January 1, 2010, to September 1, 2021 and the language was English. The exclusion criteria for the articles selected were that non-original study and studies contained any duplicated data. A total of 375 papers were screened and a researcher conducted a full-text search of these publications to assess the relevance of each study. In total, 53 studies were selected for the present review.

DNA methylation

DNA methylation is among the earliest-discovered and most intensively studied epigenetic regulatory mechanisms. It refers to the chemical modification process, in which specific bases in DNA sequences obtain a methyl group from S-adenosyl methionine by covalent bonding via DNA methyltransferase (DNMT) activity (8,9). DNA base sites that undergo methylation include the C-5 position of cytosine, the N-6 position of adenine and the N-7 position of guanine (8,9). In mammals, the most important and only DNA methylation modification is that of the carbon atom at position C-5 of cytosine in CpG dinucleotides (8,9). DNA methylation controls gene expression by altering DNA conformation, DNA stability and DNA interactions with protein, as well as chromatin structure (8,10,11).

The methylation of DNA can be divided into de novo and maintenance forms, whereby the former indicates that a DNA duplex is not methylated before undergoing methylation, whereas the latter describes methylation of DNA duplexes that have been methylated previously. The methylation process is mainly mediated by DNMTs (12,13). In mammals, DNMT3a and DNMT3b perform de novo methylation, whereas maintenance methylation occurs via DNMT1 activity (12,13). Other proteins, such as Alpha Thalassemia/Mental Retardation Syndrome X-Linked, also contribute to methylation, assisted by accessory factors, such as the retinoblastoma protein, which function in chromosome remodeling and DNA helicase activity (14,15).

Methylated DNA can also be demethylated, which can be either a passive or an active process. In the passive process, no methylation occurs during semi-retained replication and DNA gradually becomes demethylated via ‘dilution’. The active process involves demethylation via demethylases, such as DNA glycosidase and methylated CpG-binding proteins, such as methyl-CpG-binding domain protein 2 (1618).

DNA methylation can also interact with the epigenetic regulation of histones. For example, DNA methylation leads to the removal of histone acetylation marks near the methylated site via the recruitment of histone deacetylases (19), and lysine 9 methylation on histone H3 can promote DNA methylation processes (20).

DNA methylation has important functions in mammalian development and disease processes. In particular, embryonic development involves the continuous establishment of methylation and demethylation (21,22). Methylation of CpG dinucleotide sequences in gene promoter regions can lead to silencing of gene expression (1), which can also serve an important role in disease pathogenesis. For example, via the inactivation of the tumor suppressor gene, cyclin-dependent kinase inhibitor 2A, following methylation (2325), which is an important cause of tumorigenesis.

RNA methylation

RNA methylation refers to the methyltransferase catalysis chemical modification of methyl groups via selective addition of methyladenine to RNA, which primarily regulates gene expression at the post-transcriptional level (2). To date, more than 170 cellular RNA modifications have been identified in various coding and noncoding RNAs (26). m6A is the most abundant and important RNA modification in eukaryotic mRNAs, which is mainly located near stop codons and in the 3′noncoding region and is an important epigenetic modification of both mRNA and noncoding RNA (27). Methylation can affect the stability, localization, transportation, transcription, maturation and translation of RNA molecules at the post-transcriptional level, which thereby regulates gene expression (2,3).

M6A methylation of RNA occurs mainly on adenines in the DRACH sequence (m6A consensus motifs) and is regulated by methyltransferase-encoders, demethylase-decoders and m6A binding protein-readers (28). Methyltransferases, including methyltransferase-like (METTL)3, METTL14, WT1 associated protein and vir-like m6A methyltransferase-associated (KIAA1429), mainly promote methylation to regulate gene expression and function (28). Demethylases, primarily AlkB homolog 5 RNA demethylase (ALKBH5) and fat mass and obesity-associated protein (FTO), regulate gene function via demethylation (28). m6A binding proteins regulate the translation, degradation and splicing, as well as other gene functions modified by m6A, via recognizing and binding to m6A methylation sites (28).

RNA methylation has important biological functions in mammals, with roles in germ cell development, immunity and cell differentiation (29). RNA methylation is also closely related to numerous malignant diseases, including liver cancer, lung cancer, breast cancer and glioma (30).

Effects of methylation on circRNA biological function

Methylation is closely related to circRNAs

Ferreira et al (31) reported that enhancing CpG island methylation in the promoter regions of genes in human tumor cells leads to the significant downregulation of corresponding linear and circRNA transcript expression, including of tumor suppressor candidate 3, protein O-mannosyltransferase 1, attractin-like 1 and sterile α-motif domain containing 4A. These data indicated that methylation in gene promoter regions also affects circRNA expression. The most abundant epigenetic modification in eukaryotes of RNAs is m6A and numerous studies have confirmed that m6A is common in circRNAs. For example, 360 m6A-circRNAs have been detected in lung tissue in a hypoxia-induced pulmonary hypertension rat model and control tissues (32). A total of 9,382 circRNAs have been identified in lens epithelium cells from age-related cataract and control cells, 4,646 of which had m6A peaks with differing abundances (33). Furthermore, in a study systematically describing global circRNA m6A modification patterns in glioblastoma (GBM), researchers demonstrated that compared with the normal control group, there were 1,370 new m6A peaks and 1,322 missing m6A peaks in circRNAs from GBM. This study also demonstrated that m6A levels were positively correlated with circRNA expression (34). Moreover, in an analysis of 10 upregulated and 10 significantly downregulated differentially expressed circRNAs, selected by analysis of the circRNA expression profiles from poorly-differentiated adenocarcinoma of the stomach, 6 upregulated and 8 downregulated circRNAs with high scores for prediction of m6A sites were identified, respectively (35). Analysis of m6A modification patterns in human ameloblastoma, relative to normal oral tissues, also detected 364 differentially methylated m6A sites within circRNAs, of which 22.5% were hypermethylated (36). In a study of circRNA expression profiles and m6A modification in poorly-differentiated gastric adenocarcinoma, a total of 65 differentially expressed circRNAs were detected, most of which had m6A modification, whereby the trend in m6A modification changes was generally consistent with that of circRNA expression levels (35). In another study, the number of circRNA reads recovered in m6A immunoprecipitation experiments was compared with the total input of circRNA sequence reads, leading to an estimation that ~13% of total circRNAs possess an m6A modification (37). m6A tends to occur in larger exonic regions of circRNAs and is concentrated upstream and in the middle of exonic regions, and m6A exhibits cell-specificity, for example, more than half of m6A-circRNAs detected in HeLa cells were not detected in human embryonic stem cells (38,39).

Effects of methylation on the biogenesis, transport, stability, translation and degradation of circRNA

Fig. 1 presents the use of three small interfering (si) RNAs to downregulate METTL3 expression led to the strong and specific downregulation of circ-zinc finger protein 609 (ZNF609) levels, accompanied by an increase in its unspliced precursor RNA, which indicated that decreased methylation may influence the conversion of precursor mRNA to circRNA (40). Similarly, when two siRNAs were used to downregulate YTH domain containing 1 (YTHDC1) expression levels, circ-ZNF609 levels decreased and those of its precursor transcript increased. Moreover, corresponding linear mRNA levels remained unchanged, with this phenomenon occurring regardless of cell type, which indicated that YTHDC1 influences circ-ZNF609 splicing (40). Further results from this study demonstrated that YTHDC1 expression does not affect circ-ZNF609 subcellular localization, excluding the possibility of circRNA degradation due to nuclear retention (40). In general, METTL3 and YTHDC1 influence circRNA formation by regulating m6A modification, a process that is driven by m6A modification at specific sites (39).

A study on the effects of circNOP2/Sun RNA methyltransferase 2 (NSUN2) in colorectal cancer (CRC) liver metastasis, reported that it underwent m6A modification, whereas RNA pull-down and mass spectrometry analysis indicated that there was an interaction between circNSUN2, YTHDC1, serine and arginine rich splicing factor 3 (SRSF3) and nuclear RNA export factor 1 (NXF1) (41). YTHDC1, an m6A reader, interacts with the splicing factor, SRSF3, to regulate mRNA splicing and promote binding of mRNA to SRSF3 and the canonical export receptor, NXF1, to mediate m6A-modified mRNA export and metabolism (38). This therefore indicates that m6A modification may influence circNSUN2 subcellular localization. Silencing of YTHDC1 expression significantly increases circNSUN2 levels in the nucleus, whereas overexpression of wild-type YTHDC1 can restore circNSUN2 export from the nucleus to the cytoplasm (41). RNA-fluorescence in situ hybridization analysis has previously demonstrated that silencing of the m6A methyltransferase, METTL3, leads to a significant increase in circNSUN2 levels in the nucleus and restores defective circNSUN2 cytoplasmic export following the overexpression of wild-type METTL3 (41). Fig. 1 presents that m6A modification of circNSUN2 regulates its cytoplasmic export.

Recognition of m6A-circRNAs via YTH domain-containing family protein (YTHDF)2 regulates the stability of the corresponding parental genes (39,42). Whether YTHDF2 can regulate circRNA stability via the recognition of m6A modified circRNAs warrants further exploration. circRNA-SORE (a circRNA upregulated in sorafenib-resistant HCC cells) activates the Wnt/β-catenin signaling pathway and induces sorafenib resistance via a microRNA (miRNA/miR) sponge mechanism, while increased circRNA-SORE levels benefit from improved stability. RNA pull-down assays demonstrated that circRNA-SORE interacts with METTL3, FTO and YTHDF1/2; an encoder, decoder and reader of m6A, respectively (43), which indicated that circRNA-SORE is associated with m6A modifications. A set of sequence-specific morpholino antisense oligonucleotides designed to target the m6A site in circRNA-SORE have been used to reduce m6A-modified circRNA-SORE in HepG2-SR cells, which led to a reduction in circRNA-SORE stability (43). Furthermore, it has been demonstrated that wild-type circRNA-SORE is more stable than m6A-mutated circRNA-SORE during actinomycin D treatment (43), indicating that circRNA-SORE undergoes m6A modification and that this improves mRNA-SORE stability, which is presented in Fig. 1. These findings led to the proposal of a novel mechanism of sorafenib resistance in patients with hepatocellular carcinoma (HCC) and suggested that both circRNA-SORE and its m6A modification may be potential drug intervention targets in patients with advanced HCC.

The m6A reader, YTHDF3, interacts with ribosomal proteins to promote mRNA translation as m6A modification enables cap-independent translation (4446). Furthermore, a recent study has reported that circRNAs can be translated into proteins (47) and therefore m6A modification may promote circRNA translation. Jakobi et al (48) compared the expression profiles of circRNAs from human cardiovascular cell models and cardiac tissue with those of m6A-methylated circRNAs described in previous studies (49). This aforementioned study concluded that certain m6A-modified circRNAs function in protein translation. Furthermore, Yang et al (37) reported that the negative control may also initiate translation when exploring internal ribosome entry site-mediated circRNA translation. Further experiments confirmed that the short sequence containing the m6A site has an important role in this process, as well as the m6A encoders, METTL3 and METTL14, and the readers, YTHDF1, YTHDF2 and YTHDF3. Moreover, circRNA translation was demonstrated to be reduced by the m6A demethylase decoder, FTO, whereas YTHDF3 was determined to strongly interact with the translation initiation factor, eukaryotic translation initiation factor 4-γ2 (eIF4G2), thereby promoting translation (37). These aforementioned studies demonstrated that the transcription initiation factors, eIF4G2, YTHDF1, YTHDF2 and YTHDF3, may be involved in m6A-induced translation of circRNAs and that this process is regulated by METTL3 and METTL14. These results were confirmed by a further study on the effect of m6A modification on circ-ZNF609 translation. Fig. 1 presents although YTHDF1/2 consumption was demonstrated to have no effect on circ-ZNF609 transformation, both YTHDF3 and eIF4G2 were determined to physically interact with endogenous circ-ZNF609, with increases or decreases in these factors being reported to affect the circ-ZNF609 translation level (40).

There are two main mechanisms of mRNA degradation of m6A modification: i) The YTHDF2/C-C motif chemokine receptor 4 signaling pathway (50); and ii) the YTHDF2/heat-responsive protein 12 (HRSP12; adaptor protein)/RNase P/mitochondrial RNA processing enzyme (MRP) complex signaling pathway (51). YTHDF2 can recognize m6A and HRSP12 is an adaptor that can connect YTHDF2 and the RNase P/MRP complex. Fig. 1 presents circRNAs containing m6A are recognized by YTHDF2 in an HRSP12-dependent manner, this is followed by selective downregulation by RNase P/MRP, which indicates that m6A modification is involved in circRNA degradation via the YTHDF2/HRSP12/RNase P/MRP complex signaling pathway (52).

Study of methylation-modified circRNAs in disease

Studies of methylation-modified circRNA molecules in disease are summarized in Table I.

Table I.

M6A-modified circRNAs in disease.

Table I.

M6A-modified circRNAs in disease.

First author, yearcircRNADiseaseMechanismFunction(Refs.)
Chen C, 2021circ1662Colorectal cancer METTL3/circ1662/YAP1/SMAD3Cell invasion and migration(53)
Chen RX, 2019circNSUN2Colorectal cancerNot clearInvasion(41)
Liu H, 2021circDLC1Hepatocellular carcinoma KIAA1429/circDLC1Cell proliferation and migration(54)
Rao X, 2021circ-ARL3Hepatocellular carcinoma HBx/METTL3/circ-ARL3/miR-1305Cell proliferation and invasion(55)
Zhao J, 2019circE7Cervical cancerMETTL3/14/circE7/E7 proteinCell transformation(56)
Chen Z, 2021circ0000069Cervical cancer METTL3/circ0000069/miR-4426Cell proliferation and migration(57)
Li B, 2021circNDUFB2Non-small cell lung cancer TRIM25/circNDUFB2/IGF2BPs ternary complexCell growth and metastasis(58)
Nan A, 2019circNOL10Lung carcinoma circNOL10/SCML1/HN/p53Cell growth(59)
Wu P, 2021circCUX1Hypopharyngeal squamous cell carcinoma METTL3/circCUX1Radiotherapy resistance(61)
Li Z, 2021circMETTL3Breast cancer METL14/circMETTL3/miR-31-5p-CDK1Cell proliferation, migration and invasion(62)
Su H, 2020circXpo6 and circTmtc3Pulmonary hypertensionNot clearNot clear(32)
Guo M, 2020 macrophage pyroptosis hsa_circ_0029589 Atherosclerosis IRF-1/METTL3/hsa_circ_0029589 (66)
Li X, 2021circGFRα1Infertility METTL14/circGFRα1Female germline stem cell renewal and maintenance(67)

[i] METTL, methyltransferase-like; YAP1, Yes-associated protein 1; SMAD3, SMAD Family Member 3; NSUN2, Sun RNA methyltransferase 2; DLC1, deleted in liver cancer 1; KIAA1429, vir-like m6A methyltransferase-associated; ARL3, ADP ribosylation factor-like GTPase 3; HBx, HBV X protein; NDUFB2, NADH:ubiquinone oxidoreductase subunit B2; TRIM, tripartite motif-containing; IGF2BP, insulin-like growth factor-2 mRNA-binding protein; NOL10, nucleolar protein 10; SCML1, Scm polycomb group protein-like 1; HN, humanin; p53, tumor protein p53; CUX1, CUT-like homeobox 1; CDK1, cyclin dependent kinase 1; Xpo6, exportin 6; Tmtc3, transmembrane O-mannosyltransferase-targeting cadherins 3; IRF-1, interferon regulatory factor 1; GFRα1, GDNF family receptor alpha 1.

Cancer
CRC

In CRC cells, circ1662 expression increases in response to overexpression of METTL3 (53), which mediates m6A modification of the reverse complementary sequence of circ1662 to induce circ1662 expression. Furthermore, circ1662 can induce yes-associated protein 1 to translocate from the cytoplasm to the nucleus, where it activates SMAD3 to promote CRC cell invasion and migration (53).

circNSUN2 levels are elevated in tissues and sera from patients with liver metastases from CRC, indicating that it may serve a role in cancer cell metastasis. circNSUN2 levels are elevated in both the nucleus and cytoplasm of CRC cells, but particularly in the cytoplasm. Increased m6A methylation levels can promote CRC cell invasion. Moreover, a previous study demonstrated that the mutation of an m6A modification site in a plasmid for overexpression of circNUSN2, leads to the downregulation of m6A modification levels and a consequent reduction in CRC cell invasive activity. Overall, these results indicated that m6A modification of circNSUN2 is important for CRC cell invasive ability (41).

HCC

circ-deleted in liver cancer 1 (DLC1) is expressed at low levels in HCC tissues and cells and inhibits HCC proliferation and migration when it is overexpressed, which is a good prognostic indicator in patients with HCC (54). The circDLC1 molecule was identified by methylated RNA immunoprecipitation sequencing experiments, indicating that it undergoes m6A modification. Furthermore, circDLC1 is differentially expressed in response to the knockdown of the m6A methyltransferase complex, KIAA1429, which indicates that KIAA1429 regulates HCC progression via the regulation of m6A modification of circDLC1 in HCC cells (54).

circ-ADP ribosylation factor-like GTPase 3 [ARL3; also referred to as human serum albumin (hsa)_circ_0092493] is highly expressed in hepatitis B virus (HBV)-positive HCC cells and promotes their proliferation and invasion (55). The main underlying mechanism involves HBV X protein upregulation of METTL3, which increases circ-ARL3 m6A modification (55). YTHDC1A binds to m6A modified circ-ARL3 and supports its reverse splicing and biogenesis (55). Furthermore, the high expression of circ-ARL3 antagonizes the inhibitory effects of miR-1305 on oncogenes (55).

Cervical cancer (CC)

Human papillomavirus type 16-derived circE7 can be translated into protein and promote the transformation of CC cells (56). The translation of this protein is independent of corresponding linear molecules and circE7 has a specific level of m6A modification (56). Silencing expression of the RNA methyltransferases, METTL3/14, can significantly inhibit circE7 expression. Mutation of a potential m6A site in the circE7 untranslated region can also significantly decrease its expression and E7 protein expression levels. However, the expression of corresponding linear molecules does not significantly change, which indicates that m6A modification promotes the expression of circE7 and its protein products, which therefore promotes CC disease progression (56).

circ0000069 promotes CC cell proliferation and migration by inhibiting miR-4426 (57). METTL3 knockdown significantly inhibits circ0000069 m6A levels and expression and m6A modification can improve circ0000069 stability (57).

Lung cancer

In a previous study of non-small cell lung cancer, circ-NADH:ubiquinone oxidoreductase subunit B2 (circ-NDUFB2) was reported to act as a scaffold during formation of the tripartite motif-containing (TRIM) 25/circNDUFB2/insulin-like growth factor-2 mRNA-binding proteins (IGF2BPs) ternary complex, which promotes ubiquitination and degradation of IGF2BPs and inhibits non-small cell lung cancer progression (58). Levels of circNDUFB2 m6A modification affect the strength of circNDUFB2 binding to IGF2BPs, which influences the ubiquitin-ligase activity of TRIM25 for IGF2BPs (58). Furthermore, the combination of circ-nucleolar protein 10 (circNOL10) and the transcription factor Scm polycomb group protein-like 1 (SCML1) in lung cancer cells promotes SCML1 transcriptional regulation of humanin (HN), regulates the HN polypeptide family, alters various tumor signaling pathways, including p53, and inhibits lung cancer cell growth (59). Moreover, m6A methylation of pre-NOL10 in H460 and A549 cells inhibits circNOL10. Epithelial splicing regulatory protein 1 and methylated pre-NOL10 regulate circNOL10 expression together in lung cancer cells (59).

Hypopharyngeal squamous cell carcinoma (HPSCC)

HPSCC is among the most common malignancies of the head and neck and is one of the malignant tumors that exhibits the worst prognosis (60). circRNAs are potential biomarkers and/or therapeutic targets in numerous types of tumor. However, the expression and function of circRNA regulation via m6A in HPSCC remains unclear. Wu et al (61) demonstrated that circ-CUT-like homeobox 1 (circCUX1) is upregulated in patients with HPSCC resistant to radiotherapy and that it is a predictor of poor survival. Furthermore, this study reported that METTL3 mediates circCUX1 m6A methylation and stabilizes its expression, whereas circCUX1 knockdown promotes hypopharyngeal cancer cell sensitivity to radiotherapy (61).

Breast cancer

circMETTL3 is highly expressed in breast cancer and methylated RNA immunoprecipitation (MeRIP) analysis has demonstrated that it is highly enriched in the m6A precipitation component (62). This previous study demonstrated that the downregulation of METTL14 can reduce circMETTL3 expression levels, whereas FTO reduction has the opposite effect, which indicates that circMETTL3 expression may be regulated by m6A modification levels. Furthermore, circMETTL3 was determined to promote the proliferation, migration and invasion of breast cancer cells via the circMETTL3/miR-31-5p/cyclin dependent kinase 1 (CDK1) axis (62).

Immunity and non-neoplastic disease
Immunity

Immune responses can be controlled by m6A modification (63). Chen et al (64) reported that m6A-modified endogenous circRNAs have important functions in dampening innate immune responses by inhibiting retinoic acid-inducible gene I (RIG-I) activation. m6A-modified exogenous circRNAs contribute to immune function by activating RIG-I-mediated innate immune responses. Furthermore, exogenous circRNAs can induce antigen-specific T and B cell activation, antibody production and antitumor immune capacity in vivo (64). YTHDF2 may also be essential for innate immunity suppression via recognition of m6A-modification and promotion of m6A-modified circRNA degradation (64).

Cardiovascular diseases

A previous study reported that m6A modification levels of 166 and 191 circRNAs were significantly up- and downregulated, respectively, in lung tissues in a rat model of hypoxia-induced pulmonary hypertension (HPH) compared with the controls. Moreover, m6A levels of circRNAs, which are mainly derived from coding regions, were also lower overall in HPH rats than those in the control groups (32). Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses of 76 circRNAs demonstrated their upregulation with increased m6A levels and 107 circRNAs were downregulated with reduced m6A levels. The results also demonstrated that circ-exportin 6 and circ-transmembrane O-mannosyltransferase-targeting cadherins 3 were selected as significant factors because of their enriched biding sites with target miRNAs of interest (32) and their m6A modification and downregulation were confirmed as markers of HPH in pulmonary artery smooth muscle cells and pulmonary artery endothelial cells. This aforementioned study therefore provided evidence for the potential use of m6A-circRNA as a diagnostic marker and therapeutic target in HPH (32).

Macrophage pyroptosis is among the causes of atherosclerosis and interferon regulatory factor-1 (IRF-1) can effectively promote this process (65). In one study, compared with patients with clinical presentation of chest pain and those with stable angina, hsa_circ_0029589 levels were significantly lower in patients with unstable angina and acute myocardial infarction. IRF-1 was reported to induce disease progression by downregulating hsa_circ_0029589 expression and promoting macrophage pyroptosis via METTL3 upregulation to promote hsa_circ_0029589 m6A modification (66).

Infertility

Dysregulation of m6A-modified circRNAs may affect female fertility. For example, circ-GDNF family receptor α1 (circGFRα1) is stably and abundantly expressed in female germline stem cells (FGSC) and its overexpression promotes FGSC self-renewal and maintenance (67). Furthermore, m6A modification of circGFRα1 promotes its export from the nucleus to the cytoplasm. Levels of circGFRα1 m6A modification in FGSC are also high and are regulated by METTL14 (67).

Lack of circRNAs may result in male infertility as sperm carry a wealth of evolutionarily conserved circRNAs. Considering the continuous degradation of linear RNA, the continuous enrichment of circRNAs during sperm development may provide genetic information that is translated into functional proteins (68). circRNA accumulation is associated with enhanced splicing at the m6A site and m6A modification may interfere with sperm motility by influencing circRNA expression levels (68). Lack of METTL3 is among a combination of factors in spermatogenic cells that leads to a decline in m6A levels, causing spermatogenic arrest at the meiotic phase and male infertility (68).

circRNAs influence disease progression via methylation regulation

The mechanisms by which circRNAs influence disease progression via methylation regulation are summarized in Table II.

Table II.

Mechanisms of circRNA influencing disease progression via methylation regulation.

Table II.

Mechanisms of circRNA influencing disease progression via methylation regulation.

First author, yearcircRNADiseaseMethylation modificationMechanismFunction(Refs.)
Zhao Z, 2020circSOD2Hepatocellular carcinomaDNA methylation of SOCS3 circSOD2/DNMT3a/SOCS3/JAK2/STAT3Cell proliferation and tumorigenesis(70)
Liu Z, 2019circRNA-5692Hepatocellular carcinomaDNA methylation of DAB2IP circRNA-5692/miR-328/DAB2IPCell proliferation, wound healing and invasion(71)
Zhang PF, 2019circTRIM33-12Hepatocellular carcinomaDemethylation of WWC3, TP53INP1,ULBP1 and JHDM1D mRNA circTRIM33-12/miR-191/TET1/WWC3,TP53INP1, ULBP1 and JHDM1DCell proliferation, migration, invasion and immune escape(75)
Dong ZR, 2021circMEMO1Hepatocellular carcinomaDNA methylation of TCF21 circMEMO1/miR-106b/TCF21Cell proliferation, migration and invasion and the sensitivity to sorafenib treatment(76)
Sun S, 2020circ-LRIG3Hepatocellular carcinomaMethylation of STAT3 mRNA circ-LRIG3/EZH2/STAT3Cell proliferation, migration, invasion and apoptosis(77)
Chi F, 2021circmap2k4Hepatocellular carcinomaNot clear circmap2k4/miR-139-5p/YTHDF1Cell proliferation(78)
Chen N, 2018FECR1Breast cancerDNA methylation of FLI1FECR1/FLI1/TET1 or FECR1/DNMT1/FLI1Cell metastasis(80)
Xie F, 2021circPTPRABladder cancerm6A of FSCN1/MYC mRNA circPTPRA/IGF2BP1/FSCN1/MYCCell proliferation, migration and invasion(81)
Huang ZM, 2021circRNA-100284Bladder cancerDemethylation of HSP70 circRNA-100284/miR-217/HSP70/Aurora-B-cellCell cycle transition and cell proliferation(82)
Mo WL, 2021 hsa_circ_0072309Non-small cell lung carcinomaMethylation of downstream genes of FTO hsa_circ_0072309/miR-607/FTOCell tumorigenesis and invasion(83)
Zhang Z, 2021circRAB11FIP1Ovarian cancerm6A of ATG5 and ATG7 mRNA circRAB11FIP1/FTO/ATG5/ATG7Autophagy flux(84)
Wan H, 2021circRIMSEsophageal squamous cell carcinomaDNA methylation of miR-613 circRIMS/miR-613Cell proliferation(86)
Yang P, 2021circ-ATAD1Endometrial cancerDNA methylation of miR-10a circ-ATAD1/miR-10aCell invasion and migration(87)
Wu W, 2021circFAT1Endometrial cancerDNA methylation of miR-21 circFAT1/miR-21Cell stemness(88)
Du WW, 2020circSKA3GlioblastomaDNA methylation of miR-1circSKA3/miR-1Cell proliferation(89)
Yang ZG, 2017circ-Amotl1Wound repairDNA methylation of miR-17 circ-Amotl1/DNMT3a/miR-17/STATCell proliferation, survival, Migration and wound repair(90)
Wang X, 2018circIBTKSystemic lupus erythematosusDNA demethylation of AKT circIBTK/miR-29b/AKTNot clear(93)
Zhao M, 2010 hsa_circ_0012919Systemic lupus erythematosusDNA methylation of CD11a and CD70 hsa_circ_0012919/DNMT1/CD11a/CD70Autoantibody production(94)
Zhou LY, 2019circRNA ACRMyocardial ischemia reperfusion injuryDNA methylation of Pink1circRNA ACR/DNMT3b/PinkCell autophagy(97)
Huang R, 2020circSTAG1Depressionm6A of FAAH mRNA circSTAG1/ALKBH5/FAAHDepression-like behavior(98)
Zhang H, 2021circFADS2OsteoarthritisDNA methylation of miR-195 circFADS2/miR-195-5pCell apoptosis(99)

[i] SOD2, superoxide dismutase 2; DNMT, DNA methyltransferase; SOCS3, cytokine signaling 3; JAK2, Janus kinase 2; STAT3, signal transducer and activator of transcription 3; DAB2IP, DAB2 interacting protein; TRIM, tripartite motif-containing; TET1, Tet methylcytosine dioxygenase 1; WWC3, WWC family member 3; TP53INP1, tumor protein p53 inducible nuclear protein 1; ULBP1, UL16 binding protein 1; JHDM1D, lysine demethylase 7A; MEMO1, mediator of cell motility 1; TCF21, transcription factor 21; LRIG3, leucine rich repeats and immunoglobulin-like domains 3; EZH2, enhancer of 2 polycomb repressive complex 2 subunit; map2k4, mitogen-activated protein kinase kinase 4; YTHDF, YTH domain-containing family protein; FECR1, FLI1 exonic circular RNA; FLI1, friend leukemia virus integration 1; PTPRA, protein tyrosine phosphatase receptor type A; IGF2BP, insulin-like growth factor-2 mRNA-binding protein; FSCN1, fascin actin-bundling protein 1; MYC, MYC proto-oncogene, bHLH transcription factor; HSP70, heat shock protein 70; FTO, fat mass and obesity-associated protein; RAB11FIP1, RAB11 family interacting protein 1; ATG, autophagy related; RIMS, regulating synaptic membrane exocytosis protein; ATAD1, ATPase family AAA domain containing 1; FAT1, FAT atypical cadherin 1; SKA3, spindle and kinetochore-associated complex subunit 3; Amotl1, angiomotin-like 1; IBTK, inhibitor of Bruton tyrosine kinase; AKT, AKT serine/threonine kinase; CD, cluster of differentiation; ACR, autophagy-related circRNA; STAG1, stromal antigen 1; ALKBH5, primarily AlkB homolog 5 RNA demethylase; FAAH, fatty acid amide hydrolase; FADS2, fatty acid desaturase 2.

Cancer
HCC

A previous study identified a total of 1,012 upregulated and 747 downregulated circRNAs in tumor samples compared with normal adjacent samples, based on genome-wide DNA methylation and RNA sequencing data from 20 patients with HCC. No significant change in the expression levels of the corresponding parental genes of 46 upregulated and 31 downregulated circRNAs in HCC tumors was detected. It was also determined that 34 (44.2%) of these 77 differentially expressed circRNAs were significantly related to changes in DNA methylation in HCC, which indicated that abnormal DNA methylation may regulate circRNA expression in HCC (69). In the same study, MeRIP-sequencing (seq) was used to identify 5-methylcytosine (m5C) sites in circRNAs in HCC and paired adjacent non-tumor tissues, and the relationship between m5C and HCC was analyzed (69). The results indicated that there may be a correlation between HCC and m5C in circRNAs. In liver cancer tissues, 51.7% of circRNAs with methylation sites had only one methylation peak, whereas the proportion was higher in adjacent tissues (69). Furthermore, hypermethylated genes were reported to exhibit lower transcriptional expression in both liver cancer and paired adjacent non-tumor tissues (69) which was confirmed in another study (70). circ-superoxide dismutase 2 is expressed at higher levels in HCC tumors than in normal liver tissues, inhibits miR-502-5p expression and rescues levels of the miR-502-5p target gene, DNMT3a, by acting as a sponge. Upregulation of DNMT3a inhibits the suppressor of cytokine signaling 3 (SOCS3) by increasing DNA methylation of the SOCS3 promoter (70), which accelerates Janus kinase 2/STAT3 signaling pathway activation, downstream of SOCS3, thereby promoting disease progression (70). Similarly, arrest of HCC disease progression caused by the overexpression of circRNA-5692 may be due to the interaction of circRNA-5692 with a methyltransferase, which reduces the methylation level of the promoter region of the tumor suppressor DAB2 interacting protein, which enhances its expression in HCC. This process is primarily mediated by circRNA-5692 acting as sponge for miR-328-5p (71).

In addition to regulating methyltransferases, circRNAs can also control gene promoter methylation by influencing the expression of other proteins associated with methylation, which can affect tumor progression. Tet methylcytosine dioxygenase 1 (TET1) mediates DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine and 5-carboxycytosine (72). TET1 exerts antitumor functions in cancer cells via a variety of mechanisms, including increasing the expression of demethylated Wnt antagonists to inhibit the Wnt/β-catenin signaling pathway, or demethylating CpG sites in the promoter regions of tumor suppressor genes to promote tumor suppressor gene expression (73,74). In HCC, TET1 knockdown promotes tumor cell proliferation, migration and invasion, and circTRIM33-12 can regulate TET1 levels by acting as an miR-191 sponge. Furthermore, overexpression of circTRIM33-12 and TET1 can promote the mRNA and protein expression levels of WWC family member 3, tumor protein p53 inducible nuclear protein 1, UL16 binding protein 1 and lysine demethylase 7A via demethylation to suppress cancer. This process may also be associated with immune escape (75).

Cirrhosis is a recognized risk factor for HCC development. circ-mediator of cell motility 1 (circMEMO1) levels are significantly downregulated in HCC samples compared with cirrhotic nodules and are closely associated with overall survival and disease-free survival of patients with HCC (76). Analysis of the underlying mechanism has demonstrated that circMEMO1 can target the TET gene family by acting as a sponge of miR-106b-5p and increasing 5hmC levels, thereby regulating transcription factor 21 promoter methylation and gene expression to influence HCC progression (76). circMEMO1 can also increase HCC cell sensitivity to rafenib treatment (76).

circ-leucine rich repeats and immunoglobulin-like domains 3 (LRIG3) is significantly upregulated in liver cancer and promotes liver cell proliferation, migration and invasion and reduces apoptosis (77). circ-LRIG3 forms a ternary complex with enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and STAT3 to promote EZH2-induced STAT3 methylation and subsequent phosphorylation, activating the STAT3 signaling pathway (77). Subsequently, activated STAT3 can directly bind to the circ-LRIG3 promoter, which increases circ-LRIG3 transcription and therefore forms a positive feedback loop (77).

Chi et al (78) screened for microRNAs and circRNAs differentially expressed between paired HCC tumor and normal tissues. The study using The Cancer Genome Atlas and Gene Expression Ontology databases and conducted bioinformatics analysis to construct a regulatory network of circRNA/miRNA/prognostic m6A RNA methylation modulators, which was used to screen for target circRNAs. Experimental verification demonstrated that circ-mitogen-activated protein kinase 4 promotes HCC proliferation by modulating the hsa-miR-139-5p/YTHDF1 axis (78). This aforementioned study provided a research strategy to identify circRNA targets that function in disease mechanisms via methylation regulation.

Breast cancer

Leukemia virus complex factor 1 (FLI1) promotes solid tumor progression and is highly expressed in advanced and metastatic breast cancer (79). Its role in breast cancer progression and metastasis is related to circRNA FLI1 exonic circular RNA (FECR1), which is formed from exons 2, 3 and 4 of FLI1, and can both bind to the FLI1 promoter in cis configuration and recruit TET1 via a positive feedback mechanism. It also activates FLI1 expression by binding to DNMT1 in trans configuration, thereby downregulating DNMT1 expression and inducing DNA hypomethylation to promote breast cancer metastasis (80).

Bladder cancer

IGF2BP1 can function as an m6A reader and has carcinogenic effects in cancer cells by stabilizing the methylation of oncogene mRNA molecules, including Fascin actin-bundling protein 1 (FSCN1) and MYC (81). circ-protein tyrosine phosphatase receptor type A interacts with the K homology domain of IGF2BP1 and prevents it from recognizing m6A-modified FSCN1 and MYC mRNAs, to inhibit bladder cancer cell proliferation, migration and invasion (81). A previous study of bladder cancer demonstrated that circ-100284 expression increases and acts as a sponge for miR-217 to induce demethylation of heat shock protein 70 (HSP70) and enhance Aurora-B activity, which accelerates cell cycle transitions and cell proliferation (82). However, the aforementioned study did not determine how miR-217 caused HSP70 demethylation (82).

Non-small cell lung carcinoma

Hsa_circ_0072309 acts as a sponge for miR-607 to upregulate FTO and promote tumorigenesis in non-small cell lung carcinoma (83). Therefore, hsa_circ_0072309 regulates downstream gene expression levels through methylation modification via the regulation of the miR-607/FTO axis to promote non-small cell lung carcinoma tumorigenesis and invasion (83).

Ovarian cancer

circ-RAB11 family interacting protein 1 induces and promotes autophagy flux in ovarian cancer cells to increase tumor cell proliferation and migration (84). This circRNA can bind to FTO mRNA and promote FTO expression, which downregulates autophagy-related gene (ATG)5 and ATG7 mRNA m6A methylation and therefore increases ATG5 and ATG7 expression levels, promoting disease progression (84).

Esophageal squamous cell carcinoma (ESCC)

circ-regulating synaptic membrane exocytosis protein (circRIMS) can promote gastric cancer progression (85) and may also serve a similar role in ESCC. It has previously been reported that circRIMS was overexpressed in 60 patients with ESCC, predicting poor survival, and its levels were negatively correlated with those of miR-613, which is under-expressed in ESCC (86). In ESCC cells, overexpression of circRIMS was demonstrated to increase miR-613 methylation and reduce the inhibitory effect of miR-613 on cell proliferation (86). Furthermore, animal experiments demonstrated that circRIMS promotes tumor growth by downregulating miR-613 via methylation (86).

Endometrial cancer (EC)

In an analysis comparing 60 EC and paired non-tumor tissue samples, circ-ATPase family AAA domain containing 1 (circ-ATAD1) expression was downregulated in EC tissues, whereas miR-10a was upregulated and its levels were correlated with those of circ-ATAD1 (87). Reverse transcription-quantitative PCR and methylation-specific PCR analysis, determined that circ-ATAD1 overexpression promotes miR-10a methylation and downregulates its expression, completely reversing the effect of miR-10a in enhancing EC cell invasion and migration. These results indicated that circ-ATAD1 is downregulated in EC and may function to downregulate miR-10a via methylation to inhibit EC cell invasion and migration (87). Using a similar methodology, circ-FAT atypical cadherin 1 (circFAT1) was shown to be upregulated in EC and its levels were positively correlated with those of miR-21 in EC tissues (88). Overexpression of circFAT1 increased miR-21 expression levels by reducing miR-21 gene methylation to increase cancer stem cell characteristics and promote tumor progression (88).

GBM

circ-spindle and kinetochore-associated complex subunit 3 (circSKA3) is highly expressed in breast cancer and exhibits a carcinogenic effect (89). It is also highly expressed in GBM tissues, where its levels are negatively correlated with those of miR-1 (34). Furthermore, high circSKA3 levels are significantly associated with poor survival rates of patients with GBM (34). Analysis of the underlying mechanism confirmed that overexpression of circSKA3 in GBM cells increases miR-1 methylation and decreases its expression, limiting the inhibitory effect of miR-1 on cell proliferation (34).

Wound repair, immunity and non-neoplastic disease
Wound repair

Wound healing is enhanced in mice with increased circ-angiomotin-like 1 (circ-Amotl1) expression levels. This is a result of the ectopic expression of circ-Amotl1, which leads to increased levels of the transcription factor, STAT3, and promotes its nuclear translocation, which enhances DNMT3a expression leading to miR-17 gene promoter methylation (90). Decreased miR-17-5p levels increase fibronectin, DNMT3a and STAT3 expression levels, generating a positive feedback loop (90). Together, these activities promote cell proliferation, survival and migration and enhance wound repair (90).

Immunity and immune diseases

Type three innate lymphocytes (ILC3) have key roles in innate immunity and intestinal homeostasis (91). Nuclear receptor subfamily 4 group A member 1 (Nr4a1) initiates Notch2 signal activation to help maintain ILC3 homeostasis, which depends on m6A modification of Nr4a1 mRNA by the highly expressed molecule, circ-zinc finger and BTB domain-containing protein 20 (circ-Zbtb20). Circ-Zbtb20 enhances the interaction between ALKBH5 and Nr4a1 mRNA, which indicates that circRNA and methylation may have a unique role in immune system diseases (92). For example, the expression levels of circ-inhibitor of Bruton tyrosine kinase (circ-IBTK) and miR-29b are down- and upregulated, respectively, in systemic lupus erythematosus (SLE). These expression levels correlate with SLE disease activity index score, as well as anti-double-stranded DNA and complement C3 levels in patients with SLE (93). Mechanistic studies have demonstrated that circ-IBTK may reverse miR-29b-induced DNA methylation in SLE by binding to miR-29b, which activates the AKT signaling pathway and promotes disease occurrence and development (93). Similarly, the overexpression of CD11a and CD70 in CD4+ T cells contributes to the production of large amounts of autoantibodies, which induce SLE (94). Hsa_circ_0012919 upregulation is higher in CD4+ T cells from patients with active SLE than in those with inactive SLE or healthy controls. Moreover, CD11a and CD70 expression and methylation levels are inversely proportional to hsa_circ_0012919 expression levels (95). Furthermore, the overexpression of DNMT1 can reverse this phenomenon, which indicates that the upregulation of hsa_circ_0012919 reduces CD11a and CD70 methylation levels by downregulating DNMT1. This induces high expression levels of these two proteins to promote SLE occurrence and progression (95).

Cardiovascular diseases

Reducing autophagy can ameliorate cardiomyocyte damage (96) and autophagy-related circRNAs (ACRs) have been identified in a myocardium ischemia-reperfusion injury model (97). PTEN-induced kinase 1 (Pink1) expression levels are significantly altered via ACR overexpression, which indicates that ACR may regulate autophagy in cardiomyocytes via Pink1. Furthermore, RNA immunoprecipitation and biotinylated probe pull-down assays demonstrated an interaction between ACR and DNMT3b, in which DNMT3b regulates Pink1 protein expression levels (97). The results of this study also demonstrated that binding of DNMT3b to the CpG region of the Pink1 promoter increases following ACR knockdown, which demonstrated that ACR can reduce the methylation level of the CpG region of the Pink1 promoter and promote Pink1 expression. This reduced the excessive activation of the autophagy signaling pathway (97).

Neurological diseases

circ-stromal antigen 1 (circ-STAG1) levels are significantly lower in hippocampus tissue from mice exposed to chronically unpredictable stress and in the peripheral blood of patients with depression. Furthermore, circ-STAG1 overexpression significantly alleviates astrocyte dysfunction and depression-like behavior caused by chronic unpredictable stress (98). circSTAG1 binds to ALKBH5 and reduces its nuclear transport, resulting in increased m6A methylation of fatty acid amide hydrolase (FAAH) mRNA and the downregulation of FAAH expression levels in astrocytes, which subsequently alleviates depression-like behavior (98).

Osteoarthritis

Lipopolysaccharide (LPS) can induce chondrocyte damage, leading to osteoarthritis. circ-fatty acid desaturase 2 (FADS2) can protect against this effect (99). circFADS2 overexpression and miR-195-5p downregulation are observed in chondrocytes following LPS treatment and the overexpression of circFADS2 increases miR-195-5p methylation, reduces miR-195-5p expression levels and downregulates apoptosis, which therefore protects chondrocytes from LPS-induced damage (99).

Databases related to circRNA methylation research

Databases relevant to circRNA methylation research are presented in Table III. The TRCirc (http://licpathway.net/TRCirc) database mainly focuses on the retrieval and visualization of circRNA regulatory information (100). TRCirc content includes uniform transcription factor binding site data, and H3K27ac, RNA-seq and 450k chromatin immunoprecipitation-seq array data sets. The current release of TRCirc includes data from more than 100 cell types, involving 92,375 circRNAs and 161 transcription factors, covering circRNA-related genetic and epigenetic information from 151 cell lines, including DNA methylation and super-enhancer H3K27ac signaling and expression, and supports user downloads. Currently, only human data searches are supported, providing an effective screening tool for downstream analysis of circRNAs.

Table III.

Databases for circRNA methylation research.

Table III.

Databases for circRNA methylation research.

First author, yearDatabaseType of dataURL(Refs.)
Tang Z, 2019TRCircDNA methylation http://www.licpathway.net/TRCirc(100)
Huang W, 2021TransCirccircRNAs with m6A modification site information https://www.biosino.org/transcirc/(101)
Zheng Y, 2018m6AVarSingle nucleotide polymorphism sites that influence m6A modification http://m6avar.renlab.org/(102)
Luo X, 2021RMVarSingle nucleotide polymorphism sites that influence m6A modification http://rmvar.renlab.org/(103)
Zhou Y, 2016SRAMPm6A modification site prediction on the target RNA sequence http://www.cuilab.cn/sramp(104)
Liu H, 2015MeT-DBTranscriptome methylation in mammalian cells http://compgenomics.utsa.edu/methylation/(105)
Liu M, 2019CircbankcircRNA m6A modification information http://www.circbank.cn/(106)

The TransCirc database (http://www.biosino.org/transcirc/) (101) integrates a variety of translation-related evidence and search results intuitively present relevant evidence related to translation products. The data includes analysis of the translation potential of 328,080 known human circRNAs, of which there are 39,397 circRNAs with m6A modification site information.

The m6AVar (http://m6avar.renlab.org/) or RMVar (http://rmvar.renlab.org/) (102,103) databases can be used to identify single nucleotide polymorphism (SNP) sites that influence m6A modification. These databases include information on SNPs implicated in post-transcriptional regulation, miRNA binding predictions and SNPs with disease effects.

The sequence-based RNA adenosine methylation site predictor (SRAMP) database (http://www.cuilab.cn/sramp) (104) is used to predict m6A modification sites in target RNA sequences, as well as m6A modification sites in mammals. SRAMP only needs RNA sequences to run predictions, without the need to load external omics data, and is a powerful tool for m6A locus analysis where sequencing data are insufficient, such as those for long non-coding RNA and circRNA molecules.

The MethylTranscriptome (MeT-DB) database (http://compgenomics.utsa.edu/methylation/) (105) was constructed by collecting data from the Photoactivatable ribonucleoside-enhanced crosslinking and immunoprecipitation (PAR-CLIP)-seq and MeRIP-seq databases relating to eight m6A-related regulatory factors (FTO, KIAA1429, METTL14, METTL3, WT1-associated protein, heterogenous nuclear ribonucleoprotein C, YTHDC1 and YTHDF1) and allows visualization of specific regulatory positions using relevant sequencing data.

The Circbank database (circbank.cn/) (106) contains more than 140,000 human circRNA records, each of which has a separate detailed information page, which includes the following: i) circRNA sequence data; ii) mouse circRNAs with high homology to human circRNAs and their corresponding sequences; iii) predictive analysis of miRNA binding; iv) predictive open reading frame analysis; v) summary of mutations and polymorphism sites recorded in the Catalogue of Somatic Mutations in Cancer database (https://cancer.sanger.ac.uk/cosmic) (107); and vi) m6A modification information.

Conclusion

RNA methylation serves an important role in the expression, transport, stability, translation and degradation of circRNAs (108); however, to the best of our knowledge no studies have determined whether DNA methylation affects circRNA biological function. circRNAs can also regulate disease processes and may participate in immune responses by regulating proteins related to methylation modification or being modified by methylation. The interaction between circRNAs and methylation illustrates the synergistic role of epigenetics and noncoding RNA molecules in disease. Therefore, future studies focusing on the relationship between methylation and circRNAs will be important for study of mammalian development and disease. Although progress has been made in this area, further exploration is required. For example, the mechanisms underlying the effects of methylation modification on circRNA localization and expression, warrant further investigation. Moreover, to the best of our knowledge there are no relevant studies on the methylation of other basic sites in RNA, for example, the uridine modification of circRNA (another common mode of RNA methylation modification). Furthermore, the effects of circRNAs on additional cellular biological functions via methylation modification requires further elucidation, such as the metabolism, development and morphological changes of cells.

In addition to DNA methylation and RNA methylation, histone modification is also an important epigenetic process. At present, reports involving histones, methylation and circRNA are rare. For example, one study reported that circ histone-lysine N-methyltransferase eggless (Egg) inhibits histone H3 lysine 9 methylation via encoding the circEgg-P122 protein in the silkworm, Bombyx mori (Bm/bmo). Moreover, these results demonstrated that circEgg also promotes histone H3 lysine 9 acetylation and positively regulates gene expression of histone deacetylase Rpd, by acting as a sponge for the miRNA, bmo-miR-3391-5p (109). Furthermore, differentially expressed circRNA may control pheochromocytoma/paragangliomas pathogenesis by regulating histone methylation, highlighting the potential role of circRNAs as biomarkers (110). However, studies concerning epigenetic modifications, such as histone methylations and circRNAs, in the context of disease, remain very limited.

The study of combination of circRNA and methylation modification faces certain future challenges. First, there is a lack of bioinformatics methods that can efficiently identify and analyze the types, sites and patterns of circRNA methylation modifications. Second, circRNAs function in brain development, due to their unique, highly stable structures (111) and methylation is also related to mammalian development (112). How to effectively study and determine the roles and underlying mechanisms of methylation and circRNA in such developmental processes will be a challenge for future research. Finally, the goal of all biomedical research is translation into clinical application. Although methylation-modified circRNAs and methylation regulated by circRNAs have been shown to influence disease processes, translation of these findings into targeted drug treatments for patients with diseases is a difficulty that needs to be overcome in the future.

Acknowledgements

Not applicable.

Funding

This work was supported by the Natural Science Foundation of Gansu Province (grant no. 20JR5RA601) and the In-hospital project of The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army (grant no. 2021yxky057 and 2021yxky070).

Availability of data and materials

Not applicable.

Authors' contributions

CZ, HC, WZ and DC conceived the ideas for the article and helped draft the manuscript. QY, PM and ZC performed the literature search and acquisition of data. CH and FK contributed to the analysis and interpretation of all the literature. QY, CZ and DC provided funding support. All authors read and approved the final manuscript.

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.

References

1 

Razin A and Cedar H: DNA methylation and gene expression. Microbiol Rev. 55:451–458. 1991. View Article : Google Scholar : PubMed/NCBI

2 

Covelo-Molares H, Bartosovic M and Vanacova S: RNA methylation in nuclear pre-mRNA processing. Wiley Interdiscip Rev RNA. 9:e14892018. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Ehrlich M: DNA hypermethylation in disease: Mechanisms and clinical relevance. Epigenetics. 14:1141–1163. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Yang C, Hu Y, Zhou B, Bao Y, Li Z, Gong C, Yang H, Wang S and Xiao Y: The role of m6A modification in physiology and disease. Cell Death Dis. 11:9602020. View Article : Google Scholar : PubMed/NCBI

6 

Chen LL and Yang L: Regulation of circRNA biogenesis. RNA Biol. 12:381–388. 2015. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Jones PA: Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat Rev Genet. 13:484–492. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Breiling A and Lyko F: Epigenetic regulatory functions of DNA modifications: 5-methylcytosine and beyond. Epigenetics Chromatin. 8:242015. View Article : Google Scholar : PubMed/NCBI

10 

Sobiak B and Leśniak W: The effect of single CpG demethylation on the pattern of DNA-protein binding. Int J Mol Sci. 20:9142019. View Article : Google Scholar : PubMed/NCBI

11 

Cedar H and Bergman Y: Programming of DNA methylation patterns. Ann Rev Biochem. 81:97–117. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Okano M, Bell DW, Haber DA and Li E: DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 99:247–257. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Goll MG and Bestor TH: Eukaryotic cytosine methyltransferases. Ann Rev Biochem. 74:481–514. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Gibbons RJ, McDowell TL, Raman S, O'Rourke DM, Garrick D, Ayyub H and Higgs DR: Mutations in ATRX, encoding a SWI/SNF-like protein, cause diverse changes in the pattern of DNA methylation. Nat Genet. 24:368–371. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Shamma A, Suzuki M, Hayashi N, Kobayashi M, Sasaki N, Nishiuchi T, Doki Y, Okamoto T, Kohno S, Muranaka H, et al: ATM mediates pRB function to control DNMT1 protein stability and DNA methylation. Mol Cell Biol. 33:3113–3124. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Bhutani N, Burns DM and Blau HM: DNA demethylation dynamics. Cell. 146:866–872. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Dean W: Pathways of DNA demethylation. Adv Exp Med Biol. 945:247–274. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Bochtler M, Kolano A and Xu GL: DNA demethylation pathways: Additional players and regulators. Bioessays. 39:1–13. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN and Bird A: Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature. 393:386–389. 1998. View Article : Google Scholar : PubMed/NCBI

20 

Zheng Y, He L, Wan Y and Song J: H3K9me-enhanced DNA hypermethylation of the p16INK4a gene: An epigenetic signature for spontaneous transformation of rat mesenchymal stem cells. Stem Cells Dev. 22:256–267. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Gujar H, Weisenberger DJ and Liang G: The roles of Human DNA methyltransferases and their isoforms in shaping the epigenome. Genes (Basel). 10:1722019. View Article : Google Scholar : PubMed/NCBI

22 

Chen Z and Zhang Y: Role of mammalian DNA methyltransferases in development. Ann Rev Biochem. 89:135–158. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Wang FL, Yang Y, Liu ZY, Qin Y and Jin T: Correlation between methylation of the p16 promoter and cervical cancer incidence. Eur Rev Med Pharmacol Sci. 21:2351–2356. 2017.PubMed/NCBI

24 

Allameh A, Moazeni-Roodi A, Harirchi I, Ravanshad M, Motiee-Langroudi M, Garajei A, Hamidavi A and Mesbah-Namin SA: Promoter DNA methylation and mRNA expression level of p16 gene in oral squamous cell carcinoma: Correlation with Clinicopathological characteristics. Pathol Oncol Res. 25:1535–1543. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Li Y, Zhang T, Zhang H, Wang X, Liu X, Huang Q and Li L: Clinical significance of P16 gene methylation in lung cancer. Adv Exp Med Biol. 1255:133–142. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Huang H, Weng H, Deng X and Chen J: RNA modifications in cancer: Functions, mechanisms, and therapeutic implications. Ann Rev Cancer Biol. 4:221–240. 2020. View Article : Google Scholar

27 

Karthiya R and Khandelia P: m6A RNA methylation: Ramifications for gene expression and human health. Mol Biotechnol. 62:467–484. 2020. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Chen X, Sun YZ, Liu H, Zhang L, Li JQ and Meng J: RNA methylation and diseases: Experimental results, databases, Web servers and computational models. Brief Bioinform. 20:896–917. 2019. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Ferreira HJ, Davalos V, de Moura MC, Soler M, Perez-Salvia M, Bueno-Costa A, Setien F, Moran S, Villanueva A and Esteller M: Circular RNA CpG island hypermethylation-associated silencing in human cancer. Oncotarget. 9:29208–29219. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Su H, Wang G, Wu L, Ma X, Ying K and Zhang R: Transcriptome-wide map of m6A circRNAs identified in a rat model of hypoxia mediated pulmonary hypertension. BMC Genomics. 21:392020. View Article : Google Scholar : PubMed/NCBI

33 

Li P, Yu H, Zhang G, Kang L, Qin B, Cao Y, Luo J, Chen X, Wang Y, Qin M, et al: Identification and characterization of N6-methyladenosine circRNAs and methyltransferases in the lens epithelium cells from age-related cataract. Invest Ophthalmol Vis Sci. 61:132020. View Article : Google Scholar

34 

Zhou M, Li H, Chen K, Ding W, Yang C and Wang X: CircSKA3 downregulates miR-1 through methylation in glioblastoma to promote cancer cell proliferation. Cancer Manag Res. 13:509–514. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Zhang C, Wang J, Geng X, Tu J, Gao H, Li L, Zhou X, Wu H, Jing J, Pan W and Mou Y: Circular RNA expression profile and m6A modification analysis in poorly differentiated adenocarcinoma of the stomach. Epigenomics. 12:1027–1040. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Niu X, Xu J, Liu J, Chen L, Qiao X and Zhong M: Landscape of N6-methyladenosine modification patterns in human ameloblastoma. Front Oncol. 10:5564972020. View Article : Google Scholar : PubMed/NCBI

37 

Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, et al: Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 27:626–641. 2017. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

Zhou C, Molinie B, Daneshvar K, Pondick JV, Wang J, Van Wittenberghe N, Xing Y, Giallourakis CC and Mullen AC: Genome-wide maps of m6A circRNAs identify widespread and cell-type-specific methylation patterns that are distinct from mRNAs. Cell Rep. 20:2262–2276. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Di Timoteo G, Dattilo D, Centrón-Broco A, Colantoni A, Guarnacci M, Rossi F, Incarnato D, Oliviero S, Fatica A, Morlando M and Bozzoni I: Modulation of circRNA Metabolism by m6A modification. Cell Rep. 31:1076412020. View Article : Google Scholar : PubMed/NCBI

41 

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 : PubMed/NCBI

42 

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

43 

Xu J, Wan Z, Tang M, Lin Z, Jiang S, Ji L, Gorshkov K, Mao Q, Xia S, Cen D, et al: N6-methyladenosine-modified circRNA-SORE sustains sorafenib resistance in hepatocellular carcinoma by regulating β-catenin signaling. Mol Cancer. 19:1632020. View Article : Google Scholar : PubMed/NCBI

44 

Coots RA, Liu XM, Mao Y, Dong L, Zhou J, Wan J, Zhang X and Qian SB: m6A Facilitates eIF4F-independent mRNA translation. Mol Cell. 68:504–514.e7. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Li A, Chen YS, Ping XL, Yang X, Xiao W, Yang Y, Sun HY, Zhu Q, Baidya P, Wang X, et al: Cytoplasmic m6A reader YTHDF3 promotes mRNA translation. Cell Res. 27:444–447. 2017. View Article : Google Scholar : PubMed/NCBI

46 

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

47 

Lei M, Zheng G, Ning Q, Zheng J and Dong D: Translation and functional roles of circular RNAs in human cancer. Mol Cancer. 19:302020. View Article : Google Scholar : PubMed/NCBI

48 

Jakobi T, Siede D, Eschenbach J, Heumüller AW, Busch M, Nietsch R, Meder B, Most P, Dimmeler S, Backs J, et al: Deep characterization of circular RNAs from human cardiovascular cell models and cardiac tissue. Cells. 9:16162020. View Article : Google Scholar : PubMed/NCBI

49 

Kmietczyk V, Riechert E, Kalinski L, Boileau E, Malovrh E, Malone B, Gorska A, Hofmann C, Varma E, Jürgensen L, et al: m6A-mRNA methylation regulates cardiac gene expression and cellular growth. Life Sci Alliance. 2:e2018002332019. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

Park OH, Ha H, Lee Y, Boo SH, Kwon DH, Song HK and Kim YK: Endoribonucleolytic cleavage of m6A-containing RNAs by RNase P/MRP complex. Mol Cell. 74:494–507.e8. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Lee Y, Choe J, Park OH and Kim YK: Molecular mechanisms driving mRNA degradation by m6A modification. Trends Genet. 36:177–188. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Chen C, Yuan W, Zhou Q, Shao B, Guo Y, Wang W, Yang S, Guo Y, Zhao L, Dang Q, et al: N6-methyladenosine-induced circ1662 promotes metastasis of colorectal cancer by accelerating YAP1 nuclear localization. Theranostics. 11:4298–4315. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Liu H, Lan T, Li H, Xu L, Chen X, Liao H, Chen X, Du J, Cai Y, Wang J, et al: Circular RNA circDLC1 inhibits MMP1-mediated liver cancer progression via interaction with HuR. Theranostics. 11:1396–1411. 2021. View Article : Google Scholar : PubMed/NCBI

55 

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 : PubMed/NCBI

56 

Zhao J, Lee EE, Kim J, Yang R, Chamseddin B, Ni C, Gusho E, Xie Y, Chiang CM, Buszczak M, et al: Transforming activity of an oncoprotein-encoding circular RNA from human papillomavirus. Nat Commun. 10:23002019. View Article : Google Scholar : PubMed/NCBI

57 

Chen Z, Ling K, Zhu Y, Deng L, Li Y and Liang Z: circ0000069 promotes cervical cancer cell proliferation and migration by inhibiting miR-4426. Biochem Biophys Res Commun. 551:114–120. 2021. View Article : Google Scholar : PubMed/NCBI

58 

Li B, Zhu L, Lu C, Wang C, Wang H, Jin H, Ma X, Cheng Z, Yu C, Wang S, et al: circNDUFB2 inhibits non-small cell lung cancer progression via destabilizing IGF2BPs and activating anti-tumor immunity. Nat Commun. 12:2952021. View Article : Google Scholar : PubMed/NCBI

59 

Nan A, Chen L, Zhang N, Jia Y, Li X, Zhou H, Ling Y, Wang Z, Yang C, Liu S and Jiang Y: Circular RNA circNOL10 inhibits lung cancer development by promoting SCLM1-mediated transcriptional regulation of the humanin polypeptide family. Adv Sci (Weinh). 6:18006542019. View Article : Google Scholar : PubMed/NCBI

60 

Marur S and Forastiere AA: Head and neck squamous cell carcinoma: Update on epidemiology, diagnosis, and treatment. Mayo Clinic Proc. 91:386–396. 2016. View Article : Google Scholar : PubMed/NCBI

61 

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

62 

Li Z, Yang HY, Dai XY, Zhang X, Huang YZ, Shi L, Wei JF and Ding Q: CircMETTL3, upregulated in a m6A-dependent manner, promotes breast cancer progression. Int J Biol Sci. 17:1178–1190. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Wang H, Hu X, Huang M, Liu J, Gu Y, Ma L, Zhou Q and Cao X: Mettl3-mediated mRNA m6A methylation promotes dendritic cell activation. Nat Commun. 10:18982019. View Article : Google Scholar : PubMed/NCBI

64 

Chen YG, Chen R, Ahmad S, Verma R, Kasturi SP, Amaya L, Broughton JP, Kim J, Cadena C, Pulendran B, et al: N6-methyladenosine modification controls circular RNA immunity. Mol Cell. 76:96–109.e9. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Guo M, Yan R, Yao H, Duan L, Sun M, Xue Z and Jia Y: IFN regulatory Factor 1 mediates macrophage pyroptosis induced by oxidized low-density lipoprotein in patients with acute coronary syndrome. Mediators Inflamm. 2019:29171282019. View Article : Google Scholar : PubMed/NCBI

66 

Guo M, Yan R, Ji Q, Yao H, Sun M, Duan L, Xue Z and Jia Y: IFN regulatory Factor-1 induced macrophage pyroptosis by modulating m6A modification of circ_0029589 in patients with acute coronary syndrome. Int Immunopharmacol. 86:1068002020. View Article : Google Scholar : PubMed/NCBI

67 

Li X, Tian G and Wu J: Novel circGFRα1 promotes self-renewal of female germline stem cells mediated by m6A Writer METTL14. Front Cell Dev Biol. 9:6404022021. View Article : Google Scholar : PubMed/NCBI

68 

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

69 

He Y, Zhang Q, Zheng Q, Yu X and Guo W: Distinct 5-methylcytosine profiles of circular RNA in human hepatocellular carcinoma. Am J Transl Res. 12:5719–5729. 2020.PubMed/NCBI

70 

Zhao Z, Song J, Tang B, Fang S, Zhang D, Zheng L, Wu F, Gao Y, Chen C, Hu X, et al: CircSOD2 induced epigenetic alteration drives hepatocellular carcinoma progression through activating JAK2/STAT3 signaling pathway. J Exp Clin Cancer Res. 39:2592020. View Article : Google Scholar : PubMed/NCBI

71 

Liu Z, Yu Y, Huang Z, Kong Y, Hu X, Xiao W, Quan J and Fan X: circRNA-5692 inhibits the progression of hepatocellular carcinoma by sponging miR-328-5p to enhance DAB2IP expression. Cell Death Dis. 10:9002019. View Article : Google Scholar : PubMed/NCBI

72 

An J, Rao A and Ko M: TET family dioxygenases and DNA demethylation in stem cells and cancers. Exp Mol Med. 49:e3232017. View Article : Google Scholar : PubMed/NCBI

73 

Pei YF, Tao R, Li JF, Su LP, Yu BQ, Wu XY, Yan M, Gu QL, Zhu ZG and Liu BY: TET1 inhibits gastric cancer growth and metastasis by PTEN demethylation and re-expression. Oncotarget. 7:31322–31335. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Fan J, Zhang Y, Mu J, He X, Shao B, Zhou D, Peng W, Tang J, Jiang Y, Ren G and Xiang T: TET1 exerts its anti-tumor functions via demethylating DACT2 and SFRP2 to antagonize Wnt/β-catenin signaling pathway in nasopharyngeal carcinoma cells. Clin Epigenetics. 10:1032018. View Article : Google Scholar : PubMed/NCBI

75 

Zhang PF, Wei CY, Huang XY, Peng R, Yang X, Lu JC, Zhang C, Gao C, Cai JB, Gao PT, et al: Circular RNA circTRIM33-12 acts as the sponge of MicroRNA-191 to suppress hepatocellular carcinoma progression. Mol Cancer. 18:1052019. View Article : Google Scholar : PubMed/NCBI

76 

Dong ZR, Ke AW, Li T, Cai JB, Yang YF, Zhou W, Shi GM and Fan J: CircMEMO1 modulates the promoter methylation and expression of TCF21 to regulate hepatocellular carcinoma progression and sorafenib treatment sensitivity. Mol Cancer. 20:752021. View Article : Google Scholar : PubMed/NCBI

77 

Sun S, Gao J, Zhou S, Li Y, Wang Y, Jin L, Li J, Liu B, Zhang B, Han S, et al: A novel circular RNA circ-LRIG3 facilitates the malignant progression of hepatocellular carcinoma by modulating the EZH2/STAT3 signaling. J Exp Clin Cancer Res. 39:2522020. View Article : Google Scholar : PubMed/NCBI

78 

Chi F, Cao Y and Chen Y: Analysis and validation of circRNA-miRNA network in regulating m6A RNA methylation modulators reveals CircMAP2K4/miR-139-5p/YTHDF1 axis involving the proliferation of hepatocellular carcinoma. Front Oncol. 11:5605062021. View Article : Google Scholar : PubMed/NCBI

79 

Miao B, Bauer AS, Hufnagel K, Wu Y, Trajkovic-Arsic M, Pirona AC, Giese N, Taipale J, Siveke JT, Hoheisel JD and Lueong S: The transcription factor FLI1 promotes cancer progression by affecting cell cycle regulation. Int J Cancer. 147:189–201. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Chen N, Zhao G, Yan X, Lv Z, Yin H, Zhang S, Song W, Li X, Li L, Du Z, et al: A novel FLI1 exonic circular RNA promotes metastasis in breast cancer by coordinately regulating TET1 and DNMT1. Genome Biol. 19:2182018. View Article : Google Scholar : PubMed/NCBI

81 

Xie F, Huang C, Liu F, Zhang H, Xiao X, Sun J, Zhang X and Jiang G: CircPTPRA blocks the recognition of RNA N6-methyladenosine through interacting with IGF2BP1 to suppress bladder cancer progression. Mol Cancer. 20:682021. View Article : Google Scholar : PubMed/NCBI

82 

Huang ZM, Wang H and Ji ZG: circRNA-100284 activates aurora kinase B by inducing methylation of HSP70 via microRNA-217 to promote proliferation of bladder cancer cells. J Cancer Res Clin Oncol. 147:703–712. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Mo WL, Deng LJ, Cheng Y, Yu WJ, Yang YH and Gu WD: Circular RNA hsa_circ_0072309 promotes tumorigenesis and invasion by regulating the miR-607/FTO axis in non-small cell lung carcinoma. Aging. 13:11629–11645. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Zhang Z, Zhu H and Hu J: circRAB11FIP1 promoted autophagy flux of ovarian cancer through DSC1 and miR-129. Cell Death Dis. 12:2192021. View Article : Google Scholar : PubMed/NCBI

85 

Lin J, Zhang Y, Zeng X, Xue C and Lin X: circRNA circRIMS acts as a MicroRNA sponge to promote gastric cancer metastasis. ACS Omega. 5:23237–23246. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Wan H, Yuan B, Jiang K, Wei J, Feng X, Sun B and Wang F: circRNA circRIMS is overexpressed in esophageal squamous cell carcinoma and downregulate miR-613 through methylation to increase cell proliferation. Cancer Manag Res. 13:4587–4595. 2021. View Article : Google Scholar : PubMed/NCBI

87 

Yang P, Yun K and Zhang R: circRNA circ-ATAD1 is downregulated in endometrial cancer and suppresses cell invasion and migration by downregulating miR-10a through methylation. Mamm Genome. 32:488–494. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Wu W, Zhou J, Wu Y, Tang X and Zhu W: Overexpression of circRNA circFAT1 in endometrial cancer cells increases their stemness by upregulating miR-21 through methylation. Cancer Biother Radiopharm. Jul 27–2021.(Epub ahead of print). doi: 10.1089/cbr.2020.4506. View Article : Google Scholar

89 

Du WW, Yang W, Li X, Fang L, Wu N, Li F, Chen Y, He Q, Liu E, Yang Z, et al: The circular RNA circSKA3 binds integrin β1 to induce invadopodium formation enhancing breast cancer invasion. Mol Ther. 28:1287–1298. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Yang ZG, Awan FM, Du WW, Zeng Y, Lyu J, Wu D, Gupta S, Yang W and Yang BB: The circular RNA interacts with STAT3, increasing its nuclear translocation and wound repair by modulating Dnmt3a and miR-17 function. Mol Ther. 25:2062–2074. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Domingues RG and Hepworth MR: Immunoregulatory sensory circuits in group 3 innate lymphoid cell (ILC3) function and tissue homeostasis. Front Immunol. 11:1162020. View Article : Google Scholar : PubMed/NCBI

92 

Liu B, Liu N, Zhu X, Yang L, Ye B, Li H, Zhu P, Lu T, Tian Y and Fan Z: Circular RNA circZbtb20 maintains ILC3 homeostasis and function via Alkbh5-dependent m6A demethylation of Nr4a1 mRNA. Cell Mol Immunol. 18:1412–1424. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Wang X, Zhang C, Wu Z, Chen Y and Shi W: CircIBTK inhibits DNA demethylation and activation of AKT signaling pathway via miR-29b in peripheral blood mononuclear cells in systemic lupus erythematosus. Arthritis Res Ther. 20:1182018. View Article : Google Scholar : PubMed/NCBI

94 

Zhao M, Sun Y, Gao F, Wu X, Tang J, Yin H, Luo Y, Richardson B and Lu Q: Epigenetics and SLE: RFX1 downregulation causes CD11a and CD70 overexpression by altering epigenetic modifications in lupus CD4+ T cells. J Autoimmun. 35:58–69. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Zhang C, Wang X, Chen Y, Wu Z, Zhang C and Shi W: The down-regulation of hsa_circ_0012919, the sponge for miR-125a-3p, contributes to DNA methylation of CD11a and CD70 in CD4+ T cells of systemic lupus erythematous. Clin Sci (Lond). 132:2285–2298. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Corsetti G, Chen-Scarabelli C, Romano C, Pasini E, Dioguardi FS, Onorati F, Knight R, Patel H, Saravolatz L, Faggian G and Scarabelli TM: Autophagy and Oncosis/Necroptosis are enhanced in cardiomyocytes from heart failure patients. Med Sci Monit Basic Res. 25:33–44. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Zhou LY, Zhai M, Huang Y, Xu S, An T, Wang YH, Zhang RC, Liu CY, Dong YH, Wang M, et al: The circular RNA ACR attenuates myocardial ischemia/reperfusion injury by suppressing autophagy via modulation of the Pink1/FAM65B pathway. Cell Death Differ. 26:1299–1315. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Huang R, Zhang Y, Bai Y, Han B, Ju M, Chen B, Yang L, Wang Y, Zhang H, Zhang H, et al: N6-Methyladenosine modification of fatty acid amide hydrolase messenger RNA in circular RNA STAG1-regulated astrocyte dysfunction and depressive-like behaviors. Biol Psychiatry. 88:392–404. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Zhang H, Ge J and Lu X: CircFADS2 is downregulated in osteoarthritis and suppresses LPS-induced apoptosis of chondrocytes by regulating miR-195-5p methylation. Arch Gerontol Geriatr. 96:1044772021. View Article : Google Scholar : PubMed/NCBI

100 

Tang Z, Li X, Zhao J, Qian F, Feng C, Li Y, Zhang J, Jiang Y, Yang Y, Wang Q and Li C: TRCirc: A resource for transcriptional regulation information of circRNAs. Brief Bioinform. 20:2327–2333. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Huang W, Ling Y, Zhang S, Xia Q, Cao R, Fan X, Fang Z, Wang Z and Zhang G: TransCirc: An interactive database for translatable circular RNAs based on multi-omics evidence. Nucleic Acids Res. 49:D236–D242. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Zheng Y, Nie P, Peng D, He Z, Liu M, Xie Y, Miao Y, Zuo Z and Ren J: m6AVar: A database of functional variants involved in m6A modification. Nucleic Acids Res. 46:D139–D145. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Luo X, Li H, Liang J, Zhao Q, Xie Y, Ren J and Zuo Z: RMVar: An updated database of functional variants involved in RNA modifications. Nucleic Acids Res. 49:D1405–D1412. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Zhou Y, Zeng P, Li YH, Zhang Z and Cui Q: SRAMP: Prediction of mammalian N6-methyladenosine (m6A) sites based on sequence-derived features. Nucleic Acids Res. 44:e912016. View Article : Google Scholar : PubMed/NCBI

105 

Liu H, Flores MA, Meng J, Zhang L, Zhao X, Rao MK, Chen Y and Huang Y: MeT-DB: A database of transcriptome methylation in mammalian cells. Nucleic Acids Res. 43:(Database Issue). D197–D203. 2015. View Article : Google Scholar : PubMed/NCBI

106 

Liu M, Wang Q, Shen J, Yang BB and Ding X: Circbank: A comprehensive database for circRNA with standard nomenclature. RNA Biol. 16:899–905. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N, Boutselakis H, Cole CG, Creatore C, Dawson E, et al: COSMIC: The catalogue of somatic mutations in cancer. Nucleic Acids Res. 47:D941–D947. 2019. View Article : Google Scholar : PubMed/NCBI

108 

Zhang L, Hou C, Chen C, Guo Y, Yuan W, Yin D, Liu J and Sun Z: The role of N 6-methyladenosine (m 6A) modification in the regulation of circRNAs. Mol Cancer. 19:1052020. View Article : Google Scholar : PubMed/NCBI

109 

Wang Z, Zhang Y, Dai K, Liang Z, Zhu M, Zhang M, Pan J, Hu X, Zhang X, Xue R, et al: circEgg regulates histone H3K9me3 by sponging bmo-miR-3391-5p and encoding circEgg-P122 protein in the silkworm, Bombyx mori. Insect Biochem Mol Biol. 124:1034302020. View Article : Google Scholar : PubMed/NCBI

110 

Yu A, Li M, Xing C, Chen D, Wang C, Xiao Q, Zhang L, Pang Y, Wang Y, Zu X and Liu L: A Comprehensive analysis identified the key differentially expressed circular ribonucleic acids and methylation-related function in pheochromocytomas and paragangliomas. Front Genet. 11:152020. View Article : Google Scholar : PubMed/NCBI

111 

Mehta SL, Dempsey RJ and Vemuganti R: Role of circular RNAs in brain development and CNS diseases. Prog Neurobiol. 186:1017462020. View Article : Google Scholar : PubMed/NCBI

112 

Smith ZD and Meissner A: DNA methylation: Roles in mammalian development. Nat Rev Genet. 14:204–220. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 25 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang C, Cui H, Huang C, Kong F, Yang Q, Miao P, Cao Z, Zhang W and Chang D: Interactions of circRNAs with methylation: An important aspect of circRNA biogenesis and function (Review). Mol Med Rep 25: 169, 2022
APA
Zhang, C., Cui, H., Huang, C., Kong, F., Yang, Q., Miao, P. ... Chang, D. (2022). Interactions of circRNAs with methylation: An important aspect of circRNA biogenesis and function (Review). Molecular Medicine Reports, 25, 169. https://doi.org/10.3892/mmr.2022.12685
MLA
Zhang, C., Cui, H., Huang, C., Kong, F., Yang, Q., Miao, P., Cao, Z., Zhang, W., Chang, D."Interactions of circRNAs with methylation: An important aspect of circRNA biogenesis and function (Review)". Molecular Medicine Reports 25.5 (2022): 169.
Chicago
Zhang, C., Cui, H., Huang, C., Kong, F., Yang, Q., Miao, P., Cao, Z., Zhang, W., Chang, D."Interactions of circRNAs with methylation: An important aspect of circRNA biogenesis and function (Review)". Molecular Medicine Reports 25, no. 5 (2022): 169. https://doi.org/10.3892/mmr.2022.12685