Abstract

As the most prevalent internal modification in eukaryotic RNAs, N6-methyladenosine (m6A) has been discovered to play an essential role in cellular proliferation, metabolic homeostasis, embryonic development, etc. With the rapid accumulation of research interest in m6A, its crucial roles in the regulations of disease development and drug response are gaining more and more attention. Thus, a database offering such valuable data on m6A-centered regulation is greatly needed; however, no such database is as yet available. Herein, a new database named ‘M6AREG’ is developed to (i) systematically cover, for the first time, data on the effects of m6A-centered regulation on both disease development and drug response, (ii) explicitly describe the molecular mechanism underlying each type of regulation and (iii) fully reference the collected data by cross-linking to existing databases. Since the accumulated data are valuable for researchers in diverse disciplines (such as pathology and pathophysiology, clinical laboratory diagnostics, medicinal biochemistry and drug design), M6AREG is expected to have many implications for the future conduct of m6A-based regulation studies. It is currently accessible by all users at: https://idrblab.org/m6areg/

M6AREG systematically covers, for the first time, data on the effect of m6A-centered regulation on both disease development and drug response, explicitly describes the molecular mechanism underlying each type of regulation and fully references the collected data by cross-linking to existing databases.
Graphical Abstract

M6AREG systematically covers, for the first time, data on the effect of m6A-centered regulation on both disease development and drug response, explicitly describes the molecular mechanism underlying each type of regulation and fully references the collected data by cross-linking to existing databases.

INTRODUCTION

As the most prevalent internal modification in eukaryotic RNAs, N6-methyladenosine (m6A) is widely known to play essential roles in cellular proliferation, metabolic homeostasis, embryonic development, and so on (1–6). With the rapid increase in research interest in m6A, its crucial roles in the occurrence and progression of various diseases, including cardiovascular disease (7), influenza (8), gastroenteritis (9), liver fibrosis (10), diabetes (11) and cancer (12), are gaining more and more attention. Moreover, various types of m6A regulation are found to affect the responses of a drug in its corresponding disease via mediating the expression of target genes (13–15). In other words, the regulation of disease development and drug response by m6A has emerged as one of the most promising directions in recent years (7–19), and various studies have been conducted to uncover the molecular mechanism underlying regulation (mediated by the regulators of methyltransferase (writers), demethylase (erasers) and m6A-binding proteins (readers) (20–22).

The above-mentioned studies have accumulated valuable data for researchers in the diverse directions of (i) pathology and pathophysiology clarifying the essential role of m6A modifications in the occurrence/progression of disease (23–26), (ii) clinical laboratory diagnostics promoting the discovery of new therapeutic targets (27–31) and diagnostic/prognostic biomarkers (32–37) and (iii) medicinal biochemistry and drug design facilitating the discovery of m6A’s mechanisms in determining drug sensitivity and the design of new drug and drug combinations (38–42). Therefore, it is essential to have a database that provides data on the effect of m6A-centered regulation on disease development and drug response, together with the molecular mechanisms underlying each type of regulation.

So far, a variety of popular m6A-related databases have been developed (43–52). Some focus on describing the genome-wide landscape of RNA modification, variants and a variant's effect on post-transcriptional regulation [such as RMDisease (43), RMBase (44) and RMVar (45)]. Some others aim to show the m6A modification sites of RNAs based on sequencing experiments and transcriptome-wide prediction [such as m6A-Atlas (46), WHISTLE (47), m6Avar (48) and SRAMP (49)]. The remaining databases provide the validated or predicted targets of m6A regulators based on low-/high-throughput studies [such as MeT-DB (50) and M6A2Target (51)]. The majority of these existing databases have been frequently accessed and highly cited due to their considerable contributions to the needs of research communities (44–50). However, to date, there is no database available to provide data of m6A-centered regulation of disease development and drug response. Moreover, such a database is needed to systematically describe the molecular mechanisms underlying each type of regulation.

Herein, a database named ‘m6A-centered regulations of disease development and drug response (M6AREG)’ is therefore introduced. First, a systematic literature review on m6A’s regulation of disease development and drug response was conducted. The development of various diseases and the response data of drugs that were reported to be regulated by the corresponding m6A were systematically collected into the M6AREG database. Second, the molecular mechanisms underlying each type of regulation discussed above were then retrieved from the literature. The target RNAs regulated by the identified m6A regulators and their corresponding pathways were systematically collected. In particular, their regulation profiles (including m6A modification pattern, up-/down-regulation of expression and the cell processes regulated) were provided. Finally, because some xenobiotics were reported to interact with and mediate a particular m6A regulator (53–55), a number of xenobiotics regulating disease development and drug response through the mediation of certain m6A regulators were also provided. M6AREG data were also fully cross-linked to available databases. Since the data provided in M6AREG (https://idrblab.org/m6areg/) are valuable for diverse directions, it is expected to have great implications for the future conduct of m6A-based regulation studies.

FACTUAL CONTENT AND DATA RETRIEVAL

Data collection for m6A-centered regulation

Data on m6A-centered regulation of disease developments and drug responses were collected based on the following procedure. First, a large number of diseases, drugs and the m6A regulators were retrieved from ICD-11 (56), DrugBank (57), TTD (58), NCBI Gene (59), UniProt (60) and HGNC (61). Second, the data about m6A-centered regulations of disease development and drug response were collected by a comprehensive literature review in PubMed (59) using the keywords: ‘m6A + disease’, ‘m6A + drug’, ‘m6A Regulator Name + disease’, ‘m6A Regulator Name + drug’, ‘m6A + Disease Name’, ‘m6A + Drug Name’, ‘Regulator Name + Disease Name’, ‘Regulator Name + Drug Name’, etc. As a result, the development of 165 classes of diseases (such as diabetes, lung cancer and rheumatoid arthritis) and the response data of 70 drugs (such as cisplatin, sorafenib and tamoxifen) which were regulated by 31 regulators (such as FTO, METTL3 and ALKBH5) were collected in M6AREG. Third, all identified publications were reviewed and their detailed regulation information was manually collected, which included cell lines, animal models, targets and pathways. In addition, a total of 93 xenobiotics that regulated disease development and drug response via affecting m6A regulators were also collected.

The m6A regulators and their biological function

The m6A modification is an important RNA methylation without affecting the nucleotide sequence, which is achieved by m6A regulators (14). Such regulators include writers (e.g. METTL3, METTL14 and METTL16), erasers (e.g. FTO and ALKBH5) and readers (e.g. YTHDC1-2, IGF2BP1-3 and SND1), which have been reported to play key biological roles in many epigenetic directions, such as the mRNA life cycle and the cellular/developmental/disease process (62).

The biological function of writers, erasers and readers

As shown in Figure 1, m6A methyltransferase complexes are composed of the catalytic core METTL3, the RNA support element METTL14, the stabilizer WTAP and various adaptors [e.g. RBM1515B, ZC3H13, GBLL1 and VIRMA (63,64)]. m6A modifications are reversible and dynamic, since they can be removed by erasers, such as FTO and ALKBH5. Both writers and erasers regulating the level of a target’s m6A modification by methylation or demethylation play a critical role in the development of different classes of diseases (such as cancer, diabetes and rheumatoid arthritis) and drug response by targeting both coding and non-coding RNA (65–68). Taking the oscillatory shear stress-induced proatherogenic process as an example (shown in Figure 2), the expression of a writer METTL3 is up-regulated, which induces METTL3-dependent m6A hypermethylation of two targets (NLRP1 and KLF4), and then results in the elicitation of atherogenic responses, such as inflammation and cell adhesion (69). Another example of a writer is METTL14, which is reported to be remarkably down-regulated in trastuzumab-resistant cancer cells compared with their parental HER2-positive breast cancer cells. Some mechanistic studies reveal that it decreases the expression of FGFR4 by m6A modification (as shown in Figure 2), and FGFR4 could phosphorylate GSK-3β and stimulate β-catenin/TCF4 signaling to drive anti-HER2 resistance (70,71). Moreover, the first identified m6A demethylase (eraser), FTO, is found to play critical roles in leukemogenesis and drug response. It promotes leukemic oncogene-mediated cell transformation and leukemogenesis, and inhibits all-trans-retinoic acid-induced leukemia cell differentiation. Mechanistic studies show that FTO performs its oncogenic role by reducing the m6A level in mRNA transcripts of ASB2 and RARA, and then decreasing their expression (72).

Function and corresponding mechanism of m6A writers and erasers in various cellular processes. The m6A modification is reversible and dynamic, which can not only be introduced into various target RNAs using the core methyltransferase complex (METTL3/METTL14/WTAP) and other adaptors, but can also be removed by the demethylases. These m6A-regulated RNAs which are involved in disease occurrence/progression and drug response were collected in M6AREG.
Figure 1.

Function and corresponding mechanism of m6A writers and erasers in various cellular processes. The m6A modification is reversible and dynamic, which can not only be introduced into various target RNAs using the core methyltransferase complex (METTL3/METTL14/WTAP) and other adaptors, but can also be removed by the demethylases. These m6A-regulated RNAs which are involved in disease occurrence/progression and drug response were collected in M6AREG.

Representative examples describing the roles and corresponding molecular mechanisms of each m6A regulator in disease development and drug response. The m6A writer METTL3 can induce hypermethylation of NLRP1 and KLF4, and then up-regulate NLRP1 while downregulating KLF4 to elicit an atherogenic responses such as inflammation and cell adhesion. METTL14 decreases FGFR4 expression which could phosphorylate GSK-3β and stimulate β-catenin/TCF4 signaling to drive anti-HER2 resistance. FTO, as an eraser, promotes leukemogenesis and inhibits all-trans-retinoic acid-induced leukemia cell differentiation via reducing the m6A level in mRNA transcripts of ASB2 and RAR. The m6A readers, HNRNPA2B1, YTHDF1 and YTHDF2 individually promote multiple myeloma progression via stabilizing ILF3 mRNA, upregulating key transcription factors of the epithelial–mesenchymal transition in cancer via increasing translation of Snail, and sensitize melanoma cells to interferon γ and anti-PD-1 by causing the decay of PD-1, CXCR4 and SOX10.
Figure 2.

Representative examples describing the roles and corresponding molecular mechanisms of each m6A regulator in disease development and drug response. The m6A writer METTL3 can induce hypermethylation of NLRP1 and KLF4, and then up-regulate NLRP1 while downregulating KLF4 to elicit an atherogenic responses such as inflammation and cell adhesion. METTL14 decreases FGFR4 expression which could phosphorylate GSK-3β and stimulate β-catenin/TCF4 signaling to drive anti-HER2 resistance. FTO, as an eraser, promotes leukemogenesis and inhibits all-trans-retinoic acid-induced leukemia cell differentiation via reducing the m6A level in mRNA transcripts of ASB2 and RAR. The m6A readers, HNRNPA2B1, YTHDF1 and YTHDF2 individually promote multiple myeloma progression via stabilizing ILF3 mRNA, upregulating key transcription factors of the epithelial–mesenchymal transition in cancer via increasing translation of Snail, and sensitize melanoma cells to interferon γ and anti-PD-1 by causing the decay of PD-1, CXCR4 and SOX10.

Once RNA has been m6A methylated, the m6A writers (including YTHDFs, YTHDCs, IGF2BPs, eIF3, HNRNPs and FMRPs) could bind to the methylation site and play a specific role in RNA nuclear export, stabilization, splicing, translation and decay (73,74). As shown in Figure 3, due to such characteristics, great diversity in the m6A modification pattern and expression regulation (up-/down-) toward target RNAs was exhibited, which resulted in different regulation mechanisms in diseases and drug responses (75,76). As illustrated in Figure 2, the m6A reader HNRNPA2B1 is up-regulated in multiple myeloma and negatively correlated with a favorable prognosis. It promotes multiple myeloma progression (such as promoting cell proliferation and inhibiting cell apoptosis) by stabilizing the mRNA of ILF3 and AKT3 (77). Another example of a reader is YTHDF2, which causes the decay of PD-1, CXCR4 and SOX10, and then sensitizes melanoma cells to interferon γ and anti-PD-1 (76). As a subtype of YTHDF, YTHDF1 mediates m6A-increased translation of Snail that is a key transcription factor of the epithelial–mesenchymal transition, which has been considered as a negative prognostic factor for the overall survival rate of liver cancer patients (78).

Different roles and mechanisms of m6A readers in various cell processes. Once the RNAs have been modified, different m6A writers will bind to the modification sites and perform diverse functions (RNA nuclear export, stabilization, translation, splicing and decay), which results in the up-/down-regulation of target genes. All RNAs that are involved in the regulation of disease occurrence/development and drug response of m6A writers were collected and explicitly described in M6AREG.
Figure 3.

Different roles and mechanisms of m6A readers in various cell processes. Once the RNAs have been modified, different m6A writers will bind to the modification sites and perform diverse functions (RNA nuclear export, stabilization, translation, splicing and decay), which results in the up-/down-regulation of target genes. All RNAs that are involved in the regulation of disease occurrence/development and drug response of m6A writers were collected and explicitly described in M6AREG.

The description and statistics of regulators in M6AREG

For each m6A regulator shown in M6AREG, the detailed descriptions on its general information were provided online, which included regulator name, synonyms, gene name, sequence, protein family, biological function, regulator type, a full list of potential target genes of the regulator and other molecular information associated with the external links to NCBI Gene (59), UniProt (60), etc. The potential targets for a certain m6A regulator were discovered based on the transcriptomic studies collected from GEO (79), such as RIP seq, CLIP-seq, eCLIP-seq, PARCLIP-seq, iCLIP-seq and RNA-seq. For each regulator and its targets, detailed information such as gene name, experimental method, fold change, cell line and the external links to GEO (79), NCBI Gene (59), UniProt (60), etc. was also described. A diagram of the mechanism of each m6A regulator was also provided in the regulator page of the M6AREG database, which included representative information of its role in m6A modification, interactions with other regulators, target genes, etc. A full list of experimentally validated disease development and drug responses that were mediated by this regulator was provided, and detailed information, such as the m6A regulation pattern, in vivo/in vitro model and pathway, was also provided.

All in all, 14 m6A writers were reported to be involved in the development of 57 diseases by targeting 317 RNAs in 469 cell lines, and two m6A erasers were found to participate in the development of 49 diseases by targeting 164 RNAs in 335 cell line models. The studies on mechanisms showed that m6A writers and erasers regulated different cellular processes (such as the cell cycle and immunity) by regulating 89 and 64 signaling pathways (such as apoptosis, mTOR signaling and nucleotide excision repair), respectively. These regulators greatly affected the efficacies of a variety of drugs (such as cisplatin and sorafenib). Moreover, 15 m6A readers were identified to participate in the development of 41 diseases by targeting 217 RNAs in 391 cell lines, which were actively involved in 73 signaling pathways and significantly affected the responses of a variety of drugs.

For an m6A-based database, it is key to provide a quantitative description on the up-/down-regulation of the m6A target genes. The RNA-seq data were therefore incorporated into this newly developed database. First, a systematic review was conducted in GEO (79) to retrieve the regulator-related RNA-seq data by searching keywords such as ‘m6A Regulator Name’, ‘m6A Regulator Name + RNA sequencing’, ‘m6A Regulator Name + RNA seq’, and so on. The comparative data that studied the gene expression variations by knocking out, knocking down or overexpressing certain regulators were collected, which led to a total of 236 GEO datasets. Second, these newly collected datasets were carefully reviewed, and those datasets without a clearly stated data pre-processing method or of <2 samples for the control/case group were excluded from our analysis (80,81). As a result, a total of 144 datasets were identified for subsequent differential expression analyses. Third, the ‘raw read count’ and ‘TPM’ datasets were analyzed using the DESeq2 and limma packages, respectively (82–84), and the ‘RPKM’ and ‘FPKM’ datasets were transformed to ‘TPM’ and then analyzed using the limma package (85). The genes identified as significantly differentially expressed (fold change >1.5 and P-value <0.05) were considered as regulated by the studied regulator (86,87). Finally, the differential expression patterns of 33 889 target genes that were regulated by at least one regulator were collected, and the quantitative description of such a regulated expression patten was provided in the regulator page and target page of the M6AREG database (as illustrated in Figure 4). Detailed information such as experimental conditions, cell line and species was also described.

Quantitative description of the regulation pattern of target genes by m6A regulators. For a target gene and corresponding m6A regulator, the differential gene expression of the target gene in cells with treatment (knockout, knockdown and overexpression) and control cells was provided in the form of a bar chart and boxplot together with fold change and P-values, which were based on the RNA-seq data collected from the GEO. Detailed information such as cell line, species and experimental condition was also explicitly described. Pink, target gene expression of the cells with treatment; blue, target gene expression of the control cells.
Figure 4.

Quantitative description of the regulation pattern of target genes by m6A regulators. For a target gene and corresponding m6A regulator, the differential gene expression of the target gene in cells with treatment (knockout, knockdown and overexpression) and control cells was provided in the form of a bar chart and boxplot together with fold change and P-values, which were based on the RNA-seq data collected from the GEO. Detailed information such as cell line, species and experimental condition was also explicitly described. Pink, target gene expression of the cells with treatment; blue, target gene expression of the control cells.

The m6A-centered regulation of disease development

Dynamic m6A modification is reported to be involved in multiple biological processes by affecting gene expression of multiple levels including tissue development, self-renewal and differentiation of stem cells, circadian rhythm, heat shock, metabolism, metastasis and sex determination (15). With the advances in technology, increasing evidence showed that m6A modification plays crucial roles in the occurrence/progression of various disease (7–12). Recent literature indicated that m6A modification could be a new molecular tool to understand the occurrence and progression of diseases, which is regulated by many m6A regulators (24,88,89), and an increasing number of m6A regulators and their corresponding target gene were identified to have clinical implications in either diagnosis/prognosis or therapy for various diseases (24,90,91).

To obtain such m6A-centered regulation data, the disease-specific regulation data of both molecules and pathways were systematically reviewed and explicitly described in M6AREG. A total of 165 diseases were identified to be regulated by m6A modification according to the latest International Classification of Diseases (56). In particular, a total of 747 RNA molecules (e.g. mRNA, lncRNA, miRNA and cirRNA) and 78 pathways (physiological/pathological) that were regulated by 30 regulators (e.g. METTL3, FTO and YTHDF2) were collected. First, general information on each disease related to an m6A regulator and target genes (such as autophagy-related protein, cyclin-dependent kinase, microRNA and lncRNA) were provided. Second, as illustrated in Figure 5, the involved cellular processes of each target gene were described, which included cell growth, cell cycle, cell migration, cell proliferation and macrophage infiltration. Third, the regulation mechanisms of target genes in m6A-centered disease responses were also collected, which included the m6A regulators involved, up-/down-regulation of target genes and regulated pathways (e.g. PI3K–Akt signaling, TNF signaling and JAK–STAT signaling). Such data on m6A-centered regulation were essential for understanding of the mechanism underlying target genes in m6A-centered disease response. Fourth, besides the involved cellular processes and regulation mechanism data, in vitro and in vivo disease models were also illustrated in M6AREG. As described in Figure 5, a total of 200 cell lines of various diseases together with 23 model organisms were collected and provided in this newly developed database. Fifth, the comprehensive information for each target gene can be accessed by clicking the ‘Target Info’ button, which includes target name, synonyms, gene name, chromosomal location, functions and links to existing databases (59–61). Meanwhile, the comprehensive data of m6A regulators can be accessed by clicking the ‘Regulator Info’ buttons, and both the pathways altered by the m6A regulators and the corresponding in vitro models were directly linked to the KEGG (92) and Cellosaurus (93) databases.

Regulation of target genes and pathways by m6A regulators in disease occurrence and progression. The detailed mechanism underlying m6A-centered disease responses is shown, which included a summary of disease responses, the m6A regulators involved, up-/down-regulation of target genes, regulated pathways, cellular processes and in vitro/in vivo disease models used. Extended descriptions can be accessed by clicking the corresponding differently colored buttons.
Figure 5.

Regulation of target genes and pathways by m6A regulators in disease occurrence and progression. The detailed mechanism underlying m6A-centered disease responses is shown, which included a summary of disease responses, the m6A regulators involved, up-/down-regulation of target genes, regulated pathways, cellular processes and in vitro/in vivo disease models used. Extended descriptions can be accessed by clicking the corresponding differently colored buttons.

The m6A-centered regulation of drug responses

In the course of drug therapy, the clinical treatment effect on a patient can be seriously affected by the lack of alternative drugs and mutations in drug response-related genes (94). An increasing number of studies showed that the m6A modification plays a vital role in drug response of various diseases, which could be regulated by various m6A regulators and multiple target genes modulated by the same regulator (15,73,95). The m6A regulators altered drug responses by modulating drug–target interaction and drug-mediated cell death signaling. On the one hand, m6A modification interfered with drug efficacy that was mediated by a multidrug efflux transporter, drug-metabolizing enzyme and drug target (96–98). On the other hand, alterations of the m6A modification can inhibit drug-mediated cell death by inducing DNA damage and modulating its repair capacity (73). A number of studies indicated that m6A regulators or their corresponding target genes have potential as drug–effect biomarkers in disease (99,100), and diverse xenobiotics could bind to m6A regulators and modify their activities, resulting in regulation of disease progression or drug response (53,54).

As shown in Figure 6, a total of 70 drugs were identified to be regulated by 21 regulators, which were verified by in vitro/in vivo experiments. General information on each drug [such as drug name, synonym, clinic status, structure, formula, International Chemical Identifie (InChI) and InChIKey] is provided, and a list of m6A target genes associated with this drug response are described in M6AREG. For each m6A target gene, the summary of the target-regulated drug response, response disease, involved regulator, target regulation, pathway response, cellular process and in vitro/in vivo models are also described (illustrated in Figure 6). Moreover, the additional description of each target, m6A regulator and disease relevant to this drug can be accessed through clicking ‘Target Info’, ‘Regulator Info’ and ‘Disease Info’, respectively. All in all, a total of 146 experimentally verified targets, 36 pathways (physiological/pathological), 46 cellular processes, 269 cell lines and 75 in vivo animal models were found to be involved in identifying drug response alterations which were mediated by m6A modification, which are all provided in M6AREG. Based on those data provided on M6AREG’s drug page, the user can readily retrieve a list of m6A targets that were involved in the therapeutic effects of the corresponding drug described.

A typical page describing the m6A-centered regulations of drug responses. General information on the drug and its m6A targets are shown. For each target, the experimentally validated mechanisms are explicitly described. The regulation data are linked to their cell line or animal models, and extended information (such as each involved disease, target gene and regulated pathway) can also be retrieved by clicking the corresponding differently colored buttons.
Figure 6.

A typical page describing the m6A-centered regulations of drug responses. General information on the drug and its m6A targets are shown. For each target, the experimentally validated mechanisms are explicitly described. The regulation data are linked to their cell line or animal models, and extended information (such as each involved disease, target gene and regulated pathway) can also be retrieved by clicking the corresponding differently colored buttons.

Moreover, it is well known that some of the leading causes of drug efficiency include the mutation/altered expression of target proteins, deregulated drug transporters and altered drug metabolism, and the m6A regulators are reported to play a vital role in affecting the drug response by acting on the related molecules (73,101–105). Thus, the potential drug responses mediated by a specific m6A regulator are collected into M6AREG. First, three types of critical proteins (drug targets, transporters and metabolic enzymes) were identified from experimentally verified targets of m6A regulators. Second, the drugs targeting these three types of critical proteins were collected. As a result, a total of 4258 drugs including 593 approved drugs were found to interact with these types of targets that were regulated by m6A modification. All drugs can be accessed at the bottom of the regulator page, which provides the drug name, drug clinical status and mechanism leading to the drug response. Detailed information can be retrieved by clicking the button ‘Drug Info’.

M6AREG data standardization, access and retrieval

To facilitate users accessing and analyzing the M6AREG data, the collected raw data were carefully cleaned up and then systematically standardized. The standardizations included (i) all M6AREG diseases were standardized based on the latest version of the International Classification of Disease (56); (ii) all genes, RNAs, proteins, pathways and in vitro models in M6AREG were standardized and cross-linked to popular databases (such as NCBI Gene, HGNC, miRbase, Ensembl, UniProt, KEGG and Cellosaurus); and (iii) M6AREG drugs were cross-linked to a variety of well-established databases (such as TTD, PubChem and DrugBank). All data in the M6AREG database can be viewed, accessed and downloaded online without a login requirement by all users.

CONCLUSION

Epigenetic modifications (e.g. histone modifications, DNA methylation and RNA modifications) regulate gene expression without changing the DNA sequence and play pivotal roles in diseases and drug responses (106–108). RNA methylations are considered as a significant epigenetic modification, and those widely studied include N6-methyladenosine (m6A), 2′-O-methyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C) and pseudouridine (ψ), whose dynamic changes can affect the fate of target RNAs and play critical roles in various bioprocesses. Similar to m6A modification, most of these methylation types are modulated by three types of regulators (writer, eraser amd reader), and play an important role in affecting the stability, translation, alternative splicing, nuclear export and secondary structure of RNAs. Moreover, different methylation types show diversity in preferred target RNAs and modification sites (109–111). Since the methylations account for >50% of 170 different types of naturally occurring modifications in RNAs (110), it is necessary to develop a system-wide method/tool to facilitate their efficient detection, as well as to explore the molecular mechanism of RNA methylation in target RNAs and their corresponding roles in disease development and drug response.

All in all, the M6AREG is unique in (i) providing the data of m6A-centered regulation on disease development and drug responses, (ii) explicitly describing the molecular mechanisms underlying each regulation and (iii) fully referencing the collected data by cross-linking to existing databases. Since the accumulated data in the M6AREG are valuable for researchers in diverse disciplines such as pathology and pathophysiology, clinical laboratory diagnostics, and medicinal biochemistry and drug design, this new database is expected to have great implications for the future conduct of m6A-based regulation studies. M6AREG is now freely accessible and fully downloadable by all users without any login requirement at: https://idrblab.org/m6areg/

FUNDING

Funded by the Natural Science Foundation of Zhejiang Province [LQ17H160009 and LR21H300001]; National Natural Science Foundation of China [81802371 and 81872798]; Leading Talent of ‘Ten Thousand Plan’ of the National High-Level Talents Special Support Plan of China; Fundamental Research Fund of Central University [2018QNA7023]; Key R&D Program of Zhejiang Province [2020C03010]; Chinese ‘Double Top-Class’ Universities [181201*194232101]; Start-up Grant of HZNU [4125C5021720421]; China Scholarship Council [201908330151]; Westlake Laboratory (Westlake Laboratory of Life Science and Biomedicine); Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare; Alibaba Cloud; and Information Tech Center of Zhejiang University.

Conflict of interest statement. None declared.

REFERENCES

1.

Mendel
M.
,
Delaney
K.
,
Pandey
R.R.
,
Chen
K.M.
,
Wenda
J.M.
,
Vagbo
C.B.
,
Steiner
F.A.
,
Homolka
D.
,
Pillai
R.S.
Splice site m6A methylation prevents binding of U2AF35 to inhibit RNA splicing
.
Cell
.
2021
;
184
:
3125
3142
.

2.

Viegas
I.J.
,
de Macedo
J.P.
,
Serra
L.
,
De Niz
M.
,
Temporao
A.
,
Silva Pereira
S.
,
Mirza
A.H.
,
Bergstrom
E.
,
Rodrigues
J.A.
,
Aresta-Branco
F.
et al. .
N6-methyladenosine in poly(A) tails stabilize VSG transcripts
.
Nature
.
2022
;
604
:
362
370
.

3.

Wei
J.
,
Yu
X.
,
Yang
L.
,
Liu
X.
,
Gao
B.
,
Huang
B.
,
Dou
X.
,
Liu
J.
,
Zou
Z.
,
Cui
X.L.
et al. .
FTO mediates LINE1 m6A demethylation and chromatin regulation in mESCs and mouse development
.
Science
.
2022
;
376
:
968
973
.

4.

Loh
X.Y.
,
Sun
Q.Y.
,
Ding
L.W.
,
Mayakonda
A.
,
Venkatachalam
N.
,
Yeo
M.S.
,
Silva
T.C.
,
Xiao
J.F.
,
Doan
N.B.
,
Said
J.W.
et al. .
RNA-binding protein ZFP36L1 suppresses hypoxia and cell-cycle signaling
.
Cancer Res.
2020
;
80
:
219
233
.

5.

Zhu
X.
,
Qiu
C.
,
Wang
Y.
,
Jiang
Y.
,
Chen
Y.
,
Fan
L.
,
Ren
R.
,
Wang
Y.
,
Chen
Y.
,
Feng
Y.
et al. .
FGFR1 SUMOylation coordinates endothelial angiogenic signaling in angiogenesis
.
Proc. Natl Acad. Sci. USA
.
2022
;
119
:
e2202631119
.

6.

Wang
A.
,
Zhang
D.
,
Li
Y.
,
Zhang
Z.
,
Li
G.
Large-scale biomolecular conformational transitions explored by a combined elastic network model and enhanced sampling molecular dynamics
.
J. Phys. Chem. Lett.
2020
;
11
:
325
332
.

7.

Wu
S.
,
Zhang
S.
,
Wu
X.
,
Zhou
X.
m6A RNA methylation in cardiovascular diseases
.
Mol. Ther.
2020
;
28
:
2111
2119
.

8.

Bayoumi
M.
,
Munir
M.
Evolutionary conservation of the DRACH signatures of potential N6-methyladenosine (m6A) sites among influenza A viruses
.
Sci. Rep.
2021
;
11
:
4548
.

9.

Lu
T.X.
,
Zheng
Z.
,
Zhang
L.
,
Sun
H.L.
,
Bissonnette
M.
,
Huang
H.
,
He
C.
A new model of spontaneous colitis in mice induced by deletion of an RNA m6A methyltransferase component METTL14 in T cells
.
Cell. Mol. Gastroenterol. Hepatol.
2020
;
10
:
747
761
.

10.

Shen
M.
,
Guo
M.
,
Li
Y.
,
Wang
Y.
,
Qiu
Y.
,
Shao
J.
,
Zhang
F.
,
Xu
X.
,
Yin
G.
,
Wang
S.
et al. .
m6A methylation is required for dihydroartemisinin to alleviate liver fibrosis by inducing ferroptosis in hepatic stellate cells
.
Free Radic. Biol. Med
.
2022
;
182
:
246
259
.

11.

De Jesus
D.F.
,
Zhang
Z.
,
Kahraman
S.
,
Brown
N.K.
,
Chen
M.
,
Hu
J.
,
Gupta
M.K.
,
He
C.
,
Kulkarni
R.N.
m6A mRNA methylation regulates human beta-cell biology in physiological states and in type 2 diabetes
.
Nat Metab
.
2019
;
1
:
765
774
.

12.

Han
D.
,
Liu
J.
,
Chen
C.
,
Dong
L.
,
Liu
Y.
,
Chang
R.
,
Huang
X.
,
Liu
Y.
,
Wang
J.
,
Dougherty
U.
et al. .
Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells
.
Nature
.
2019
;
566
:
270
274
.

13.

Lin
Z.
,
Niu
Y.
,
Wan
A.
,
Chen
D.
,
Liang
H.
,
Chen
X.
,
Sun
L.
,
Zhan
S.
,
Chen
L.
,
Cheng
C.
et al. .
RNA m6A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy
.
EMBO J.
2020
;
39
:
e103181
.

14.

Lan
Q.
,
Liu
P.Y.
,
Bell
J.L.
,
Wang
J.Y.
,
Huttelmaier
S.
,
Zhang
X.D.
,
Zhang
L.
,
Liu
T.
The emerging roles of RNA m6A methylation and demethylation as critical regulators of tumorigenesis, drug sensitivity, and resistance
.
Cancer Res.
2021
;
81
:
3431
3440
.

15.

Huang
H.
,
Weng
H.
,
Chen
J.
m6A modification in coding and non-coding RNAs: roles and therapeutic implications in cancer
.
Cancer Cell
.
2020
;
37
:
270
288
.

16.

Ding
L.W.
,
Sun
Q.Y.
,
Edwards
J.J.
,
Fernandez
L.T.
,
Ran
X.B.
,
Zhou
S.Q.
,
Scolyer
R.A.
,
Wilmott
J.S.
,
Thompson
J.F.
,
Doan
N.
et al. .
LNK suppresses interferon signaling in melanoma
.
Nat. Commun.
2019
;
10
:
2230
.

17.

Sun
Q.Y.
,
Ding
L.W.
,
Johnson
K.
,
Zhou
S.
,
Tyner
J.W.
,
Yang
H.
,
Doan
N.B.
,
Said
J.W.
,
Xiao
J.F.
,
Loh
X.Y.
et al. .
SOX7 regulates MAPK/ERK–BIM mediated apoptosis in cancer cells
.
Oncogene
.
2019
;
38
:
6196
6210
.

18.

Zhu
X.
,
Ding
S.
,
Qiu
C.
,
Shi
Y.
,
Song
L.
,
Wang
Y.
,
Wang
Y.
,
Li
J.
,
Wang
Y.
,
Sun
Y.
et al. .
SUMOylation negatively regulates angiogenesis by targeting endothelial NOTCH signaling
.
Circ. Res.
2017
;
121
:
636
649
.

19.

Wang
A.
,
Zhang
Y.
,
Chu
H.
,
Liao
C.
,
Zhang
Z.
,
Li
G.
Higher accuracy achieved for protein–ligand binding pose prediction by elastic network model-based ensemble docking
.
J. Chem. Inf. Model.
2020
;
60
:
2939
2950
.

20.

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

21.

Chen
H.
,
Yao
J.
,
Bao
R.
,
Dong
Y.
,
Zhang
T.
,
Du
Y.
,
Wang
G.
,
Ni
D.
,
Xun
Z.
,
Niu
X.
et al. .
Cross-talk of four types of RNA modification writers defines tumor microenvironment and pharmacogenomic landscape in colorectal cancer
.
Mol. Cancer
.
2021
;
20
:
29
.

22.

Zhao
Q.
,
Zhao
Y.
,
Hu
W.
,
Zhang
Y.
,
Wu
X.
,
Lu
J.
,
Li
M.
,
Li
W.
,
Wu
W.
,
Wang
J.
et al. .
m6A RNA modification modulates PI3K/Akt/mTOR signal pathway in gastrointestinal cancer
.
Theranostics
.
2020
;
10
:
9528
9543
.

23.

Barbieri
I.
,
Tzelepis
K.
,
Pandolfini
L.
,
Shi
J.
,
Millan-Zambrano
G.
,
Robson
S.C.
,
Aspris
D.
,
Migliori
V.
,
Bannister
A.J.
,
Han
N.
et al. .
Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control
.
Nature
.
2017
;
552
:
126
131
.

24.

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

25.

Zhao
Y.
,
Hu
J.
,
Sun
X.
,
Yang
K.
,
Yang
L.
,
Kong
L.
,
Zhang
B.
,
Li
F.
,
Li
C.
,
Shi
B.
et al. .
Loss of m6A demethylase ALKBH5 promotes post-ischemic angiogenesis via post-transcriptional stabilization of WNT5A
.
Clin. Transl. Med.
2021
;
11
:
e402
.

26.

Zhang
S.
,
Amahong
K.
,
Zhang
C.
,
Li
F.
,
Gao
J.
,
Qiu
Y.
,
Zhu
F.
RNA–RNA interactions between SARS-CoV-2 and host benefit viral development and evolution during COVID-19 infection
.
Brief. Bioinform
.
2021
;
23
:
bbab397
.

27.

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

28.

Chen
H.
,
Gao
S.
,
Liu
W.
,
Wong
C.C.
,
Wu
J.
,
Wu
J.
,
Liu
D.
,
Gou
H.
,
Kang
W.
,
Zhai
J.
et al. .
RNA N6-methyladenosine methyltransferase METTL3 facilitates colorectal cancer by activating the m6A–GLUT1–mTORC1 axis and is a therapeutic target
.
Gastroenterology
.
2021
;
160
:
1284
1300
.

29.

Song
H.
,
Liu
D.
,
Wang
L.
,
Liu
K.
,
Chen
C.
,
Wang
L.
,
Ren
Y.
,
Ju
B.
,
Zhong
F.
,
Jiang
X.
et al. .
Methyltransferase like 7B is a potential therapeutic target for reversing EGFR-TKIs resistance in lung adenocarcinoma
.
Mol. Cancer
.
2022
;
21
:
43
.

30.

Li
Y.H.
,
Li
X.X.
,
Hong
J.J.
,
Wang
Y.X.
,
Fu
J.B.
,
Yang
H.
,
Yu
C.Y.
,
Li
F.C.
,
Hu
J.
,
Xue
W.W.
et al. .
Clinical trials, progression-speed differentiating features and swiftness rule of the innovative targets of first-in-class drugs
.
Brief. Bioinform.
2020
;
21
:
649
662
.

31.

Xia
W.
,
Zheng
L.
,
Fang
J.
,
Li
F.
,
Zhou
Y.
,
Zeng
Z.
,
Zhang
B.
,
Li
Z.
,
Li
H.
,
Zhu
F.
PFmulDL: a novel strategy enabling multi-class and multi-label protein function annotation by integrating diverse deep learning methods
.
Comput. Biol. Med.
2022
;
145
:
105465
.

32.

Ma
L.
,
Lin
Y.
,
Sun
S.W.
,
Xu
J.
,
Yu
T.
,
Chen
W.L.
,
Zhang
L.H.
,
Guo
Y.C.
,
Wang
Y.W.
,
Chen
T.
et al. .
KIAA1429 is a potential prognostic marker in colorectal cancer by promoting the proliferation via downregulating WEE1 expression in an m6A-independent manner
.
Oncogene
.
2022
;
41
:
692
703
.

33.

Gu
Y.
,
Niu
S.
,
Wang
Y.
,
Duan
L.
,
Pan
Y.
,
Tong
Z.
,
Zhang
X.
,
Yang
Z.
,
Peng
B.
,
Wang
X.
et al. .
DMDRMR-mediated regulation of m6A-modified CDK4 by m6A reader IGF2BP3 drives ccRCC progression
.
Cancer Res.
2021
;
81
:
923
934
.

34.

Li
J.
,
Liang
L.
,
Yang
Y.
,
Li
X.
,
Ma
Y.
N6-methyladenosine as a biological and clinical determinant in colorectal cancer: progression and future direction
.
Theranostics
.
2021
;
11
:
2581
2593
.

35.

Wang
X.
,
Li
F.
,
Qiu
W.
,
Xu
B.
,
Li
Y.
,
Lian
X.
,
Yu
H.
,
Zhang
Z.
,
Wang
J.
,
Li
Z.
et al. .
SYNBIP: synthetic binding proteins for research, diagnosis and therapy
.
Nucleic Acids Res.
2022
;
50
:
D560
D570
.

36.

Li
F.
,
Yin
J.
,
Lu
M.
,
Yang
Q.
,
Zeng
Z.
,
Zhang
B.
,
Li
Z.
,
Qiu
Y.
,
Dai
H.
,
Chen
Y.
et al. .
ConSIG: consistent discovery of molecular signature from OMIC data
.
Brief. Bioinform
.
2022
;
23
:
bbac253
.

37.

Li
F.
,
Zhou
Y.
,
Zhang
Y.
,
Yin
J.
,
Qiu
Y.
,
Gao
J.
,
Zhu
F.
POSREG: proteomic signature discovered by simultaneously optimizing its reproducibility and generalizability
.
Brief. Bioinform.
2022
;
23
:
bbac040
.

38.

Deng
L.J.
,
Deng
W.Q.
,
Fan
S.R.
,
Chen
M.F.
,
Qi
M.
,
Lyu
W.Y.
,
Qi
Q.
,
Tiwari
A.K.
,
Chen
J.X.
,
Zhang
D.M.
et al. .
m6A modification: recent advances, anticancer targeted drug discovery and beyond
.
Mol. Cancer
.
2022
;
21
:
52
.

39.

Huang
C.S.
,
Zhu
Y.Q.
,
Xu
Q.C.
,
Chen
S.
,
Huang
Y.
,
Zhao
G.
,
Ni
X.
,
Liu
B.
,
Zhao
W.
,
Yin
X.Y.
YTHDF2 promotes intrahepatic cholangiocarcinoma progression and desensitises cisplatin treatment by increasing CDKN1B mRNA degradation
.
Clin. Transl. Med.
2022
;
12
:
e848
.

40.

Li
X.
,
Ma
S.
,
Deng
Y.
,
Yi
P.
,
Yu
J.
Targeting the RNA m6A modification for cancer immunotherapy
.
Mol. Cancer
.
2022
;
21
:
76
.

41.

Zhao
T.
,
Hu
Y.
,
Cheng
L.
Deep-DRM: a computational method for identifying disease-related metabolites based on graph deep learning approaches
.
Brief. Bioinform.
2021
;
22
:
bbaa212
.

42.

Xue
W.
,
Fu
T.
,
Deng
S.
,
Yang
F.
,
Yang
J.
,
Zhu
F.
Molecular mechanism for the allosteric inhibition of the human serotonin transporter by antidepressant escitalopram
.
ACS Chem. Neurosci.
2022
;
13
:
340
351
.

43.

Chen
K.
,
Song
B.
,
Tang
Y.
,
Wei
Z.
,
Xu
Q.
,
Su
J.
,
de Magalhaes
J.P.
,
Rigden
D.J.
,
Meng
J.
RMDisease: a database of genetic variants that affect RNA modifications, with implications for epitranscriptome pathogenesis
.
Nucleic Acids Res.
2021
;
49
:
D1396
D1404
.

44.

Xuan
J.J.
,
Sun
W.J.
,
Lin
P.H.
,
Zhou
K.R.
,
Liu
S.
,
Zheng
L.L.
,
Qu
L.H.
,
Yang
J.H.
RMBase v2.0: deciphering the map of RNA modifications from epitranscriptome sequencing data
.
Nucleic Acids Res.
2018
;
46
:
D327
D334
.

45.

Luo
X.
,
Li
H.
,
Liang
J.
,
Zhao
Q.
,
Xie
Y.
,
Ren
J.
,
Zuo
Z.
RMVar: an updated database of functional variants involved in RNA modifications
.
Nucleic Acids Res.
2021
;
49
:
D1405
D1412
.

46.

Tang
Y.
,
Chen
K.
,
Song
B.
,
Ma
J.
,
Wu
X.
,
Xu
Q.
,
Wei
Z.
,
Su
J.
,
Liu
G.
,
Rong
R.
et al. .
m6A-Atlas: a comprehensive knowledgebase for unraveling the N6-methyladenosine (m6A) epitranscriptome
.
Nucleic Acids Res.
2021
;
49
:
D134
D143
.

47.

Chen
K.
,
Wei
Z.
,
Zhang
Q.
,
Wu
X.
,
Rong
R.
,
Lu
Z.
,
Su
J.
,
de Magalhaes
J.P.
,
Rigden
D.J.
,
Meng
J.
WHISTLE: a high-accuracy map of the human N6-methyladenosine (m6A) epitranscriptome predicted using a machine learning approach
.
Nucleic Acids Res.
2019
;
47
:
e41
.

48.

Zheng
Y.
,
Nie
P.
,
Peng
D.
,
He
Z.
,
Liu
M.
,
Xie
Y.
,
Miao
Y.
,
Zuo
Z.
,
Ren
J.
m6AVar: a database of functional variants involved in m6A modification
.
Nucleic Acids Res.
2018
;
46
:
D139
D145
.

49.

Zhou
Y.
,
Zeng
P.
,
Li
Y.H.
,
Zhang
Z.
,
Cui
Q.
SRAMP: prediction of mammalian N6-methyladenosine (m6A) sites based on sequence-derived features
.
Nucleic Acids Res.
2016
;
44
:
e91
.

50.

Liu
H.
,
Wang
H.
,
Wei
Z.
,
Zhang
S.
,
Hua
G.
,
Zhang
S.W.
,
Zhang
L.
,
Gao
S.J.
,
Meng
J.
,
Chen
X.
et al. .
MeT-DB V2.0: elucidating context-specific functions of N6-methyl-adenosine methyltranscriptome
.
Nucleic Acids Res.
2018
;
46
:
D281
D287
.

51.

Deng
S.
,
Zhang
H.
,
Zhu
K.
,
Li
X.
,
Ye
Y.
,
Li
R.
,
Liu
X.
,
Lin
D.
,
Zuo
Z.
,
Zheng
J.
M6A2Target: a comprehensive database for targets of m6A writers, erasers and readers
.
Brief. Bioinform.
2021
;
22
:
bbaa055
.

52.

Cheng
L.
,
Qi
C.
,
Yang
H.
,
Lu
M.
,
Cai
Y.
,
Fu
T.
,
Ren
J.
,
Jin
Q.
,
Zhang
X.
gutMGene: a comprehensive database for target genes of gut microbes and microbial metabolites
.
Nucleic Acids Res.
2022
;
50
:
D795
D800
.

53.

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

54.

Qing
Y.
,
Dong
L.
,
Gao
L.
,
Li
C.
,
Li
Y.
,
Han
L.
,
Prince
E.
,
Tan
B.
,
Deng
X.
,
Wetzel
C.
et al. .
R-2-hydroxyglutarate attenuates aerobic glycolysis in leukemia by targeting the FTO/m6A/PFKP/LDHB axis
.
Mol. Cell
.
2021
;
81
:
922
939
.

55.

Dixit
D.
,
Prager
B.C.
,
Gimple
R.C.
,
Poh
H.X.
,
Wang
Y.
,
Wu
Q.
,
Qiu
Z.
,
Kidwell
R.L.
,
Kim
L.J.Y.
,
Xie
Q.
et al. .
The RNA m6A reader YTHDF2 maintains oncogene expression and is a targetable dependency in glioblastoma stem cells
.
Cancer Discov.
2021
;
11
:
480
499
.

56.

Lancet
T.
ICD-11: a brave attempt at classifying a new world
.
Lancet
.
2018
;
391
:
2476
.

57.

Wishart
D.S.
,
Feunang
Y.D.
,
Guo
A.C.
,
Lo
E.J.
,
Marcu
A.
,
Grant
J.R.
,
Sajed
T.
,
Johnson
D.
,
Li
C.
,
Sayeeda
Z.
et al. .
DrugBank 5.0: a major update to the drugbank database for 2018
.
Nucleic Acids Res.
2018
;
46
:
D1074
D1082
.

58.

Zhou
Y.
,
Zhang
Y.
,
Lian
X.
,
Li
F.
,
Wang
C.
,
Zhu
F.
,
Qiu
Y.
,
Chen
Y.
Therapeutic target database update 2022: facilitating drug discovery with enriched comparative data of targeted agents
.
Nucleic Acids Res.
2022
;
50
:
D1398
D1407
.

59.

Sayers
E.W.
,
Beck
J.
,
Bolton
E.E.
,
Bourexis
D.
,
Brister
J.R.
,
Canese
K.
,
Comeau
D.C.
,
Funk
K.
,
Kim
S.
,
Klimke
W.
et al. .
Database resources of the National Center for Biotechnology Information
.
Nucleic Acids Res.
2021
;
49
:
D10
D17
.

60.

UniProt, C.
UniProt: the universal protein knowledgebase in 2021
.
Nucleic Acids Res.
2021
;
49
:
D480
D489
.

61.

Bruford
E.A.
,
Lush
M.J.
,
Wright
M.W.
,
Sneddon
T.P.
,
Povey
S.
,
Birney
E.
The HGNC database in 2008: a resource for the human genome
.
Nucleic Acids Res.
2008
;
36
:
D445
D448
.

62.

Roundtree
I.A.
,
Evans
M.E.
,
Pan
T.
,
He
C.
Dynamic RNA modifications in gene expression regulation
.
Cell
.
2017
;
169
:
1187
1200
.

63.

Li
Y.
,
Su
R.
,
Deng
X.
,
Chen
Y.
,
Chen
J.
FTO in cancer: functions, molecular mechanisms, and therapeutic implications
.
Trends Cancer
.
2022
;
8
:
598
614
.

64.

Liu
S.
,
Zhuo
L.
,
Wang
J.
,
Zhang
Q.
,
Li
Q.
,
Li
G.
,
Yan
L.
,
Jin
T.
,
Pan
T.
,
Sui
X.
et al. .
METTL3 plays multiple functions in biological processes
.
Am. J. Cancer Res
.
2020
;
10
:
1631
1646
.

65.

Yang
Y.
,
Shen
F.
,
Huang
W.
,
Qin
S.
,
Huang
J.T.
,
Sergi
C.
,
Yuan
B.F.
,
Liu
S.M.
Glucose is involved in the dynamic regulation of m6A in patients with type 2 diabetes
.
J. Clin. Endocrinol. Metab.
2019
;
104
:
665
673
.

66.

Jiang
H.
,
Cao
K.
,
Fan
C.
,
Cui
X.
,
Ma
Y.
,
Liu
J.
Transcriptome-wide high-throughput m6A sequencing of differential m6A methylation patterns in the human rheumatoid arthritis fibroblast-like synoviocytes cell line MH7A
.
J. Inflamm. Res
.
2021
;
14
:
575
586
.

67.

Liu
S.
,
Li
Q.
,
Chen
K.
,
Zhang
Q.
,
Li
G.
,
Zhuo
L.
,
Zhai
B.
,
Sui
X.
,
Hu
X.
,
Xie
T.
The emerging molecular mechanism of m6A modulators in tumorigenesis and cancer progression
.
Biomed. Pharmacother.
2020
;
127
:
110098
.

68.

Zhang
S.
,
Amahong
K.
,
Sun
X.
,
Lian
X.
,
Liu
J.
,
Sun
H.
,
Lou
Y.
,
Zhu
F.
,
Qiu
Y.
The miRNA: a small but powerful RNA for COVID-19
.
Brief. Bioinform.
2021
;
22
:
1137
1149
.

69.

Chien
C.S.
,
Li
J.Y.
,
Chien
Y.
,
Wang
M.L.
,
Yarmishyn
A.A.
,
Tsai
P.H.
,
Juan
C.C.
,
Nguyen
P.
,
Cheng
H.M.
,
Huo
T.I.
et al. .
METTL3-dependent N6-methyladenosine RNA modification mediates the atherogenic inflammatory cascades in vascular endothelium
.
Proc. Natl Acad. Sci. USA
.
2021
;
118
:
e2025070118
.

70.

Zou
Y.
,
Zheng
S.
,
Xie
X.
,
Ye
F.
,
Hu
X.
,
Tian
Z.
,
Yan
S.
,
Yang
L.
,
Kong
Y.
,
Tang
Y.
et al. .
N6-methyladenosine regulated FGFR4 attenuates ferroptotic cell death in recalcitrant HER2-positive breast cancer
.
Nat. Commun.
2022
;
13
:
2672
.

71.

Sun
Y.
,
Gao
X.
,
Wu
P.
,
Wink
M.
,
Li
J.
,
Dian
L.
,
Liang
Z.
Jatrorrhizine inhibits mammary carcinoma cells by targeting TNIK mediated Wnt/beta-catenin signalling and epithelial–mesenchymal transition (EMT)
.
Phytomedicine
.
2019
;
63
:
153015
.

72.

Li
Z.
,
Weng
H.
,
Su
R.
,
Weng
X.
,
Zuo
Z.
,
Li
C.
,
Huang
H.
,
Nachtergaele
S.
,
Dong
L.
,
Hu
C.
et al. .
FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N6-Methyladenosine RNA Demethylase
.
Cancer Cell
.
2017
;
31
:
127
141
.

73.

Liu
S.
,
Li
G.
,
Li
Q.
,
Zhang
Q.
,
Zhuo
L.
,
Chen
X.
,
Zhai
B.
,
Sui
X.
,
Chen
K.
,
Xie
T.
The roles and mechanisms of YTH domain-containing proteins in cancer development and progression
.
Am. J. Cancer Res.
2020
;
10
:
1068
1084
.

74.

Li
B.
,
Jiang
J.
,
Assaraf
Y.G.
,
Xiao
H.
,
Chen
Z.S.
,
Huang
C.
Surmounting cancer drug resistance: new insights from the perspective of N6-methyladenosine RNA modification
.
Drug Resist. Updat.
2020
;
53
:
100720
.

75.

Feng
M.
,
Xie
X.
,
Han
G.
,
Zhang
T.
,
Li
Y.
,
Li
Y.
,
Yin
R.
,
Wang
Q.
,
Zhang
T.
,
Wang
P.
et al. .
YBX1 is required for maintaining myeloid leukemia cell survival by regulating BCL2 stability in an m6A-dependent manner
.
Blood
.
2021
;
138
:
71
85
.

76.

Yang
S.
,
Wei
J.
,
Cui
Y.H.
,
Park
G.
,
Shah
P.
,
Deng
Y.
,
Aplin
A.E.
,
Lu
Z.
,
Hwang
S.
,
He
C.
et al. .
m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade
.
Nat. Commun.
2019
;
10
:
2782
.

77.

Jiang
F.
,
Tang
X.
,
Tang
C.
,
Hua
Z.
,
Ke
M.
,
Wang
C.
,
Zhao
J.
,
Gao
S.
,
Jurczyszyn
A.
,
Janz
S.
et al. .
HNRNPA2B1 promotes multiple myeloma progression by increasing AKT3 expression via m6A-dependent stabilization of ILF3 mRNA
.
J. Hematol. Oncol.
2021
;
14
:
54
.

78.

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

79.

Barrett
T.
,
Wilhite
S.E.
,
Ledoux
P.
,
Evangelista
C.
,
Kim
I.F.
,
Tomashevsky
M.
,
Marshall
K.A.
,
Phillippy
K.H.
,
Sherman
P.M.
,
Holko
M.
et al. .
NCBI GEO: archive for functional genomics data sets—update
.
Nucleic Acids Res.
2013
;
41
:
D991
D995
.

80.

Fu
J.
,
Zhang
Y.
,
Wang
Y.
,
Zhang
H.
,
Liu
J.
,
Tang
J.
,
Yang
Q.
,
Sun
H.
,
Qiu
W.
,
Ma
Y.
et al. .
Optimization of metabolomic data processing using NOREVA
.
Nat. Protoc.
2022
;
17
:
129
151
.

81.

Tang
J.
,
Fu
J.
,
Wang
Y.
,
Li
B.
,
Li
Y.
,
Yang
Q.
,
Cui
X.
,
Hong
J.
,
Li
X.
,
Chen
Y.
et al. .
ANPELA: analysis and performance assessment of the label-free quantification workflow for metaproteomic studies
.
Brief. Bioinform.
2020
;
21
:
621
636
.

82.

Love
M.I.
,
Huber
W.
,
Anders
S.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol.
2014
;
15
:
550
.

83.

Begik
O.
,
Lucas
M.C.
,
Liu
H.
,
Ramirez
J.M.
,
Mattick
J.S.
,
Novoa
E.M.
Integrative analyses of the RNA modification machinery reveal tissue- and cancer-specific signatures
.
Genome Biol.
2020
;
21
:
97
.

84.

Ritchie
M.E.
,
Phipson
B.
,
Wu
D.
,
Hu
Y.
,
Law
C.W.
,
Shi
W.
,
Smyth
G.K.
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res.
2015
;
43
:
e47
.

85.

Zhao
Y.
,
Li
M.C.
,
Konate
M.M.
,
Chen
L.
,
Das
B.
,
Karlovich
C.
,
Williams
P.M.
,
Evrard
Y.A.
,
Doroshow
J.H.
,
McShane
L.M.
TPM, FPKM, or normalized counts? A comparative study of quantification measures for the analysis of RNA-seq data from the NCI patient-derived models repository
.
J. Transl. Med.
2021
;
19
:
269
.

86.

Yang
Q.
,
Li
B.
,
Tang
J.
,
Cui
X.
,
Wang
Y.
,
Li
X.
,
Hu
J.
,
Chen
Y.
,
Xue
W.
,
Lou
Y.
et al. .
Consistent gene signature of schizophrenia identified by a novel feature selection strategy from comprehensive sets of transcriptomic data
.
Brief. Bioinform.
2020
;
21
:
1058
1068
.

87.

Yang
Q.
,
Wang
Y.
,
Zhang
Y.
,
Li
F.
,
Xia
W.
,
Zhou
Y.
,
Qiu
Y.
,
Li
H.
,
Zhu
F.
NOREVA: enhanced normalization and evaluation of time-course and multi-class metabolomic data
.
Nucleic Acids Res.
2020
;
48
:
W436
W448
.

88.

Kumari
R.
,
Ranjan
P.
,
Suleiman
Z.G.
,
Goswami
S.K.
,
Li
J.
,
Prasad
R.
,
Verma
S.K.
mRNA modifications in cardiovascular biology and disease: with a focus on m6A modification
.
Cardiovasc. Res.
2022
;
118
:
1680
1692
.

89.

Shi
Y.
,
Fan
S.
,
Wu
M.
,
Zuo
Z.
,
Li
X.
,
Jiang
L.
,
Shen
Q.
,
Xu
P.
,
Zeng
L.
,
Zhou
Y.
et al. .
YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression
.
Nat. Commun.
2019
;
10
:
4892
.

90.

Du
A.
,
Li
S.
,
Zhou
Y.
,
Disoma
C.
,
Liao
Y.
,
Zhang
Y.
,
Chen
Z.
,
Yang
Q.
,
Liu
P.
,
Liu
S.
et al. .
M6A-mediated upregulation of circMDK promotes tumorigenesis and acts as a nanotherapeutic target in hepatocellular carcinoma
.
Mol. Cancer
.
2022
;
21
:
109
.

91.

Yang
Z.
,
Cai
Z.
,
Yang
C.
,
Luo
Z.
,
Bao
X.
ALKBH5 regulates STAT3 activity to affect the proliferation and tumorigenicity of osteosarcoma via an m6A-YTHDF2-dependent manner
.
EBioMedicine
.
2022
;
80
:
104019
.

92.

Kanehisa
M.
,
Furumichi
M.
,
Sato
Y.
,
Ishiguro-Watanabe
M.
,
Tanabe
M.
KEGG: integrating viruses and cellular organisms
.
Nucleic Acids Res.
2021
;
49
:
D545
D551
.

93.

Bairoch
A.
The cellosaurus, a cell-line knowledge resource
.
J. Biomol. Tech.
2018
;
29
:
25
38
.

94.

Zhang
Z.
,
Zhang
C.
,
Yang
Z.
,
Zhang
G.
,
Wu
P.
,
Luo
Y.
,
Zeng
Q.
,
Wang
L.
,
Xue
Q.
,
Zhang
Y.
et al. .
m6A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer
.
J. Hematol. Oncol.
2021
;
14
:
190
.

95.

Liu
S.
,
Li
Q.
,
Li
G.
,
Zhang
Q.
,
Zhuo
L.
,
Han
X.
,
Zhang
M.
,
Chen
X.
,
Pan
T.
,
Yan
L.
et al. .
The mechanism of m6A methyltransferase METTL3-mediated autophagy in reversing gefitinib resistance in NSCLC cells by beta-elemene
.
Cell Death Dis.
2020
;
11
:
969
.

96.

Yin
J.
,
Sun
W.
,
Li
F.
,
Hong
J.
,
Li
X.
,
Zhou
Y.
,
Lu
Y.
,
Liu
M.
,
Zhang
X.
,
Chen
N.
et al. .
VARIDT 1.0: variability of drug transporter database
.
Nucleic Acids Res.
2020
;
48
:
D1042
D1050
.

97.

Yin
J.
,
Li
F.
,
Zhou
Y.
,
Mou
M.
,
Lu
Y.
,
Chen
K.
,
Xue
J.
,
Luo
Y.
,
Fu
J.
,
He
X.
et al. .
INTEDE: interactome of drug-metabolizing enzymes
.
Nucleic Acids Res.
2021
;
49
:
D1233
D1243
.

98.

Zhu
F.
,
Shi
Z.
,
Qin
C.
,
Tao
L.
,
Liu
X.
,
Xu
F.
,
Zhang
L.
,
Song
Y.
,
Liu
X.
,
Zhang
J.
et al. .
Therapeutic target database update 2012: a resource for facilitating target-oriented drug discovery
.
Nucleic Acids Res.
2012
;
40
:
D1128
D1136
.

99.

Liu
K.
,
Ouyang
Q.Y.
,
Zhan
Y.
,
Yin
H.
,
Liu
B.X.
,
Tan
L.M.
,
Liu
R.
,
Wu
W.
,
Yin
J.Y.
Pharmacoepitranscriptomic landscape revealing m6A modification could be a drug-effect biomarker for cancer treatment
.
Mol. Ther. Nucleic Acids
.
2022
;
28
:
464
476
.

100.

Zhang
S.
,
Sun
X.
,
Mou
M.
,
Amahong
K.
,
Sun
H.
,
Zhang
W.
,
Shi
S.
,
Li
Z.
,
Gao
J.
,
Zhu
F.
REGLIV: molecular regulation data of diverse living systems facilitating current multiomics research
.
Comput. Biol. Med.
2022
;
148
:
105825
.

101.

Chen
Z.
,
Shi
T.
,
Zhang
L.
,
Zhu
P.
,
Deng
M.
,
Huang
C.
,
Hu
T.
,
Jiang
L.
,
Li
J.
Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: a review of the past decade
.
Cancer Lett.
2016
;
370
:
153
164
.

102.

Bivona
T.G.
,
Doebele
R.C.
A framework for understanding and targeting residual disease in oncogene-driven solid cancers
.
Nat. Med.
2016
;
22
:
472
478
.

103.

Hong
J.
,
Luo
Y.
,
Zhang
Y.
,
Ying
J.
,
Xue
W.
,
Xie
T.
,
Tao
L.
,
Zhu
F.
Protein functional annotation of simultaneously improved stability, accuracy and false discovery rate achieved by a sequence-based deep learning
.
Brief. Bioinform.
2020
;
21
:
1437
1447
.

104.

Yang
Q.
,
Li
B.
,
Chen
S.
,
Tang
J.
,
Li
Y.
,
Li
Y.
,
Zhang
S.
,
Shi
C.
,
Zhang
Y.
,
Mou
M.
et al. .
MMEASE: online meta-analysis of metabolomic data by enhanced metabolite annotation, marker selection and enrichment analysis
.
J. Proteomics
.
2021
;
232
:
104023
.

105.

Fu
T.
,
Li
F.
,
Zhang
Y.
,
Yin
J.
,
Qiu
W.
,
Li
X.
,
Liu
X.
,
Xin
W.
,
Wang
C.
,
Yu
L.
et al. .
VARIDT 2.0: structural variability of drug transporter
.
Nucleic Acids Res.
2022
;
50
:
D1417
D1431
.

106.

Rizzacasa
B.
,
Amati
F.
,
Romeo
F.
,
Novelli
G.
,
Mehta
J.L.
Epigenetic modification in coronary atherosclerosis: JACC review topic of the week
.
J. Am. Coll. Cardiol.
2019
;
74
:
1352
1365
.

107.

Song
Y.
,
Yang
L.
Transgenerational pancreatic impairment with igf2/h19 epigenetic alteration induced by p,p'-DDE exposure in early life
.
Toxicol. Lett.
2017
;
280
:
222
231
.

108.

Liu
S.
,
Lin
H.
,
Wang
D.
,
Li
Q.
,
Luo
H.
,
Li
G.
,
Chen
X.
,
Li
Y.
,
Chen
P.
,
Zhai
B.
et al. .
PCDH17 increases the sensitivity of colorectal cancer to 5-fluorouracil treatment by inducing apoptosis and autophagic cell death
.
Signal Transduct. Target. Ther
.
2019
;
4
:
53
.

109.

Nombela
P.
,
Miguel-Lopez
B.
,
Blanco
S.
The role of m6A, m5C and psi RNA modifications in cancer: novel therapeutic opportunities
.
Mol. Cancer
.
2021
;
20
:
18
.

110.

Jin
H.
,
Huo
C.
,
Zhou
T.
,
Xie
S.
m1A RNA modification in gene expression regulation
.
Genes
.
2022
;
13
:
910
.

111.

Qu
X.
,
Zhang
Y.
,
Sang
X.
,
Ren
D.
,
Zhao
H.
,
Wong
S.T.C.
Methyladenosine modification in RNAs: from regulatory roles to therapeutic implications in cancer
.
Cancers
.
2022
;
14
:
3195
.

Author notes

The authors wish it to be known that, in their opinion, the first four authors should be regarded as Joint First Authors.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.