Abstract

N6-methyladenosine (m6A) is the most abundant posttranscriptional modification in mammalian mRNA molecules and has a crucial function in the regulation of many fundamental biological processes. The m6A modification is a dynamic and reversible process regulated by a series of writers, erasers and readers (WERs). Different WERs might have different functions, and even the same WER might function differently in different conditions, which are mostly due to different downstream genes being targeted by the WERs. Therefore, identification of the targets of WERs is particularly important for elucidating this dynamic modification. However, there is still no public repository to host the known targets of WERs. Therefore, we developed the m6A WER target gene database (m6A2Target) to provide a comprehensive resource of the targets of m6A WERs. M6A2Target provides a user-friendly interface to present WER targets in two different modules: ‘Validated Targets’, referred to as WER targets identified from low-throughput studies, and ‘Potential Targets’, including WER targets analyzed from high-throughput studies. Compared to other existing m6A-associated databases, m6A2Target is the first specific resource for m6A WER target genes. M6A2Target is freely accessible at http://m6a2target.canceromics.org.

Introduction

RNA modifications were first described approximately 60 years ago [1]. To date, more than 100 kinds of RNA modifications have been discovered and determined to be involved in various kinds of cellular biological processes [2]. Among these hundreds of RNA modifications, one of the most abundant and widely distributed internal modifications in mRNA is N6-methyladenosine (m6A) [3]. As the most abundant epigenetic modification, m6A has been shown to be associated with the regulation of mRNA export [4], stability [5], splicing [6] and translation [7], while malfunction of m6A will result in changes in multiple cell functions, leading to disease occurrence. For example, FTO, a well-known eraser of m6A, has long been suggested to be associated with obesity in several population studies [8, 9] and plays a vital role in leukemogenesis [10]. Downregulation of METTL14, a writer for m6A modification, leads to poor prognosis in hepatocellular carcinoma and is significantly associated with tumor metastasis [11]. In addition, m6A modification is also considered to be related to type 2 diabetes mellitus [12], infertility [13] and growth retardation [14].

M6A modification is reversible and modulated by ‘writers’ (such as METTL3, METTL14 and WTAP) [15, 16], ‘erasers’ (such as ALKBH5 and FTO) [17, 18] and ‘readers’ (such as YTHDF1, YTHDF2 and YTHDF3) [19–22]. Although these ‘writers’, ‘erasers’ and ‘readers’ (WERs) are functional through m6A modifications, different WERs may execute entirely different functions [17–25], and even the same WER may have distinct functions under different conditions [26–28]. These differences are mostly due to different sets of downstream genes targeted by different WERs. Different WERs may target different sets of genes under the same conditions, and the same WER may also target different sets of genes under different conditions. Therefore, identification of WER targets is particularly important for further characterization of this dynamic modification.

Growing studies have focused on the identification of the targets of m6A WERs. Various kinds of next-generation sequencing technologies [29–33], mass spectrometry [34] and some binding experiments, such as coimmunoprecipitation [35], have been applied to infer the relationship between m6A WERs and their targets. To provide a comprehensive and convenient platform for researchers to obtain related information about WERs and their targets, we established the m6A WER target gene database (m6A2Target), which is freely accessible at http://m6a2target.canceromics.org. The pipeline for the database construction is described in Figure 1. M6A2Target integrated basic information of WERs and their targets, including gene annotation information from authority sites, such as NCBI, Ensemble and UniProt. In addition, m6A2Target also provided information on the associations between WERs and their targets, such as protein–DNA, protein–RNA or protein–protein interactions, and changes in gene expression level, m6A modification level or translation efficacy and alternative splicing events followed by perturbation of WERs. Growing studies have revealed vital roles of m6A in some fundamental cellular functions, such as messenger RNA stability [36], mRNA splicing [28] and RNA translation efficiency [19], and in various kinds of diseases, including cancers. Therefore, we believe that such a database collecting the targets of m6A WER in a cell-specific manner will notably facilitate further downstream functional and mechanistic studies in the m6A research field.

Flowchart of construction for the m6A2Target database.
Figure 1

Flowchart of construction for the m6A2Target database.

Materials and methods

Data sources

To establish m6A2Target, we first collected all reported m6A WERs from the literature in humans and mice. For human species, we obtained 10 writers, 2 erasers and 10 readers, and for mice, we obtained 6 writers, 2 erasers and 6 readers. We collected all the studies and datasets related to m6A WERs from NCBI PubMed, Gene Expression Omnibus (GEO) [37] and the Sequence Read Archive (SRA) [38] updated to April 2019. The quality of the raw data was checked using fastqc (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/), and the raw data that did not pass the criteria of fastqc were not used. In total, 77 datasets for 80 kinds of cell lines or mouse models were obtained. Genome Reference Consortium Human Build 38 (hg38) and Genome Reference Consortium Mouse Build 38 (mm10) were used for genome reference for humans and mice, respectively. To conduct downstream biological applications using our database, we collected gene expression data, gene mutation data and clinical information of 33 cancer types from The Cancer Genome Atlas (TCGA) [39].

Identification of validated WER targets from low-throughput studies

To obtain validated WER targets, we collected all m6A-related articles from the publicly available literature (Supplementary Table 1). We then manually curated WER targets from low-throughput experiments, such as immunoprecipitation assays, luciferase reporter assays and perturbation experiments. A gene was considered a validated WER target if it was shown to interact with any WER according to such experiments as immunoprecipitation and luciferase reporter, or its expression level, methylation level, translation efficiency, stability or other biological manifestations were altered when any WER was perturbed (such as knocked down, knocked out, mutated and overexpressed).

Identification of potential targets with binding evidence

To identify potential targets with binding evidence, we collected high-throughput sequencing data about protein–DNA interactions and protein–RNA interactions and mass spectrometry data about protein–protein interactions for all WERs (Supplementary Table 2).

To identify potential targets with protein–protein interactions with WERs, we summarized the mass spectrometry results from the supplementary data of published papers.

For identification of potential targets having protein–DNA interactions with WERs, we collected raw ChIP-seq data for all WERs from GEO [37] and SRA [38]. These raw ChIP-seq data were cleaned using fastp [40], mapped to the reference genome using bowtie2 [41], then called peaks using MACS2 [42]. The peaks were annotated to genes using HOMER (http://homer.ucsd.edu/homer/ngs/annotation.html) [43]. All genes with ChIP-seq peaks were considered potential WER targets with protein–DNA binding evidence.

For identification of potential targets having protein–RNA interactions with WERs, we collected raw sequencing data, such as RIP-seq data and various types of CLIP-seq data, for all WERs from GEO [37] and SRA [38]. For iCLIP-seq data, HITS-CLIP-seq data and eCLIP-seq data, the CLIP Tool Kit [44] was used to identify the potential binding sites, and HOMER (http://homer.ucsd.edu/homer/ngs/annotation.html) [43] was used for annotation. For PAR-CLIP-seq, after cleaning with fastp [40], bowtie [45] was first used for read alignment, PARalyzer v1.5 [46] was subsequently used to identify potential binding sites and HOMER (http://homer.ucsd.edu/homer/ngs/annotation.html) [43] was used for annotation. For RIP-seq data, with fastp [40] for read cleaning, RSEM [47] was used for RNA level quantification followed by FPKM (Fragments Per Kilobase Million) normalization, and genes with fold change between IP and INPUT greater than 2 (IP/INPUT >2) were considered potential binding targets.

Identification of potential targets inferred from WER perturbation

The potential targets inferred from WER perturbation have four levels: RNA, m6A, translation and alternative splicing. To identify potential targets at the RNA level, we collected RNA-seq data from studies with wild-type and perturbed WERs of interest. The perturbation condition could be knocked down, knocked out, mutated or overexpressed. RSEM [47] was used to generate gene counts, and DESeq2 [48] was used to obtain differentially expressed genes (P-value <0.05, fold change >1.5). The differentially expressed genes were considered potential WER targets whose RNA level could be altered by perturbation of the WER of interest.

To identify potential targets at the m6A level, we collected m6A-specific methylated RNA immunoprecipitation sequencing (MeRIP-seq) data from studies with wild-type and perturbed WERs of interest. The following pipeline was used for MeRIP-seq data analysis. First, using fastp [40] for read cleaning, STAR [49] was used for read alignment. Second, MACS2 [42] and MeTPeak [50] were used for m6A peak calling. Third, the relative m6A level was obtained by calculating the ratio between the IP RPKM value and the input RPKM value for each peak. The differential m6A peaks (P-value <0.05, fold change >1.5) between the two conditions were considered potential WER targets whose m6A level could be altered by perturbation of the WER of interest. The significance of the differential m6A methylation was measured using the model from Audic et al. [51].

To identify potential targets at the translation level, we collected ribosome profiling data from studies with wild-type and perturbed WERs of interest. The raw ribosome profiling data were first cleaned using fastp [40] and then filtered for mitochondrial DNA and ribosome RNA using Bowtie2 [41]. The remaining reads were mapped to the genome using STAR [49]. RSEM [47] was used to obtain the read count for each gene followed by FPKM normalization. The translation efficiency difference between the two conditions was calculated as log2 (FPKM of the treatment group/FPKM of the control group). Genes with different translation efficiencies (fold change >1.5) were considered potential WER targets whose translation efficiency could be altered by the perturbation of the WER of interest.

To identify potential targets with differential alternative splicing events, we used the cleaned RNA-seq data mentioned above with STAR [49] for read alignment and then used rMATS [52] for alternative splicing event detection. The differential alternative splicing events (P-value <0.05) were considered to be potential WER targets whose alternative splicing level could be altered by perturbation of the WER of interest.

Web interface implementation

All the analysis results and sample information were managed using MySQL tables (https://www.mysql.com/cn/). The web interfaces were implemented using Vue.js, Element UI and JavaScript. Furthermore, we inlayed Genomic Browser and allowed the genomic coordinates of the regions of interest as an input to search for the corresponding target location information of the user-defined genes.

Application of m6A2Target in cancer study

We collected and downloaded all mutation data, gene expression data and clinical data of 33 cancer types from TCGA (https://portal.gdc.cancer.gov/) [39]. The mutation rates of each WER in all cancer types and the oncoplot were obtained by Maftools [53]. Limma [54] was used to obtain the P-value and log2 fold change of differential KIAA1429 expression between tumor and normal samples across all cancer types. The hazard ratio of overall survival probability between the high KIAA1429 expression group and the low KIAA1429 expression group was analyzed by Cox proportional hazards regression analysis. KOBAS 3.0 [55–57] was used for gene set enrichment analysis, and the top 10 enrichment pathways are shown. Genes belonging to the PI3K-AKT signaling pathway were obtained from Kyoto Encyclopedia of Genes and Genomes (KEGG) [58]. IGV [59] was used for the visualization of the genomic distribution of two randomly selected genes. All analyses were performed using R 3.5.2 (https://www.r-project.org/).

Result

Overview of m6A2Target

For validated targets, m6A2target contains 668 targets for 22 WERs in humans and 366 targets for 14 WERs in mice. For potential targets with binding evidence, m6A2Target contains 112,592 WER-target pairs in different cell lines from humans and 12,180 WER-target pairs in cell lines from mice. For potential targets inferred from WER perturbation, in human cell lines, there were 98,046 WER-target pairs with RNA level changes, 54,146 WER-targets with m6A level changes, 17,878 WER-targets with translation efficiency changes and 61,370 WER-target pairs with differential alternative splicing events; in mice, there were 35,010 WER-target pairs with RNA level changes, 27,438 WER targets with m6A level changes and 36,214 WER-target pairs with differential alternative splicing events.

Validated WER targets from low-throughput studies

The number of m6A-related studies has been increasing annually since 2012, indicating that the m6A has become an increasingly popular research topic in the past several years (Figure 2A). According to the validated targets recorded in m6A2Target, the m6A WERs shared many targets and formed a complex network, indicating that these WERs may cooperate with each other to exert biological functions through m6A (Figures 2B and S1A). The validated targets could be classified into four major categories according to the alteration in cellular functions followed by perturbation of WER, including gene expression level, m6A level, translation efficiency and alternative splicing, and other functions, such as RNA export, RNA localization and RNA editing, were also reported (Figures 2C and S1B). Most of the validated targets were protein-coding genes, and microRNAs were the second most common gene type (Figures 2D and S1C).

Validated WER targets in the m6A2Target database. (A) The number distribution of WER targets by year. (B) The association of validated targets among identified WERs. Blue nodes indicate writers, purple nodes indicate erasers and green nodes indicate readers. Node size indicates the number of target associations identified for this WER. The thickness of the edge indicates the number of target associations that is validated by both WERs. (C) The intersection among five main types of downstream effects upon WERs perturbed, including alterations in gene expression level, m6A level, translation efficiency, alternative splicing and some other aspects, such as RNA export, RNA metabolism, RNA localization, RNA editing and some fundamental biological process. (D) The gene type distribution of validated WER targets; other types refer to those low-frequency gene types, such as sense intronic and processed transcript.
Figure 2

Validated WER targets in the m6A2Target database. (A) The number distribution of WER targets by year. (B) The association of validated targets among identified WERs. Blue nodes indicate writers, purple nodes indicate erasers and green nodes indicate readers. Node size indicates the number of target associations identified for this WER. The thickness of the edge indicates the number of target associations that is validated by both WERs. (C) The intersection among five main types of downstream effects upon WERs perturbed, including alterations in gene expression level, m6A level, translation efficiency, alternative splicing and some other aspects, such as RNA export, RNA metabolism, RNA localization, RNA editing and some fundamental biological process. (D) The gene type distribution of validated WER targets; other types refer to those low-frequency gene types, such as sense intronic and processed transcript.

Potential targets with binding evidence revealed by high-throughput studies

The potential targets with binding evidence were primarily obtained from the analysis of CLIP-seq, RIP-seq, ChIP-seq and mass spectrum data of all available WERs (Figure 3A). We classified these targets into three categories: the targets inferred from RIP-seq and CLIP-seq were regarded as ‘Protein–RNA’ associations, targets inferred from ChIP-seq were regarded as ‘Protein–DNA’ associations and targets summarized from WER mass spectrometry results were regarded as ‘Protein–Protein’ associations (Figure 3B). Among these potential targets with binding evidence per WER, the targets with m6A peaks were considered m6A-dependent targets, while others were considered to be m6A-independent. Approximately half of the targets did not contain m6A peaks, indicating that some m6A WER targets might be functional independent of m6A (Figures 3C and S2A). Most of these potential targets were protein-coding RNAs, and the second were long noncoding RNAs (lncRNAs) (Figures 3D and S2B).

WER targets with binding evidence in the m6A2Target database. (A) The number distribution of datasets for binding evidence by sequencing type. (B) The number distribution of WER targets with binding evidence by sequencing type. PR indicates protein–RNA interactions, which are evidenced from CLIP-seq or RIP-seq. PD means protein–DNA interactions, which are evidence from ChIP-seq. PP indicates protein–protein interactions, which are evidenced from the mass spectrum. (C) The number distribution of WER targets with binding evidence from binding types, classified by m6A-dependent conditions. The target gene with 1 bp overlap with m6A peaks is defined as m6A-dependent. PD refers to protein–DNA targets obtained from ChIP-seq. PR indicates protein–RNA targets, inferred from CLIP-seq and RIP-seq. (D) The number distribution of WER targets with binding evidence by gene type. (E) The genomic location of the YTHDF1 target genes with protein–RNA binding evidence in the m6A2Target database. (F) The density of distance from FTO target genes with protein–RNA binding evidence to FTO m6A peaks in the m6A2Target database. (G) The pathway enrichment with target genes with protein–protein binding evidence of WTAP.
Figure 3

WER targets with binding evidence in the m6A2Target database. (A) The number distribution of datasets for binding evidence by sequencing type. (B) The number distribution of WER targets with binding evidence by sequencing type. PR indicates protein–RNA interactions, which are evidenced from CLIP-seq or RIP-seq. PD means protein–DNA interactions, which are evidence from ChIP-seq. PP indicates protein–protein interactions, which are evidenced from the mass spectrum. (C) The number distribution of WER targets with binding evidence from binding types, classified by m6A-dependent conditions. The target gene with 1 bp overlap with m6A peaks is defined as m6A-dependent. PD refers to protein–DNA targets obtained from ChIP-seq. PR indicates protein–RNA targets, inferred from CLIP-seq and RIP-seq. (D) The number distribution of WER targets with binding evidence by gene type. (E) The genomic location of the YTHDF1 target genes with protein–RNA binding evidence in the m6A2Target database. (F) The density of distance from FTO target genes with protein–RNA binding evidence to FTO m6A peaks in the m6A2Target database. (G) The pathway enrichment with target genes with protein–protein binding evidence of WTAP.

We next performed several analyses to confirm the reliability of our database. For instance, YTHDF1 is an RNA-binding protein that is known to be functional through m6A. We found that the distribution of the ‘Protein–RNA’ targets of YTHDF1 along the genes was similar to the general m6A distribution, a peak in the stop-codon region, indicating their close association with m6A methylation (Figure 3E) FTO, a known m6A eraser. We calculated the distance between the ‘Protein–RNA’ targets of FTO and FTO-associated m6A peaks and found that the smaller the distance between FTO targets and m6A peaks is, the higher the density is, further indicating that FTO binding targets may be strongly influenced by m6A (Figure 3F). In addition, we chose the mass spectrometry results of an m6A writer, WTAP, for further analysis. These ‘Protein–Protein’ targets of WTAP can be enriched into dozens of signaling pathways, such as ribosome, spliceosome, mRNA surveillance pathway and RNA transport, illustrating the vital roles of m6A WERs in various fundamental cellular functions (Figure 3G).

Potential targets inferred from m6A WER perturbation followed by high-throughput sequencing

The other kinds of potential targets were inferred from m6A WER perturbation followed by high-throughput sequencing, such as RNA-Seq, m6A-Seq and ribosome profiling (Figures 4A and S3A). For example, after METTL3 was perturbed, 26,945 potential targets were altered at the RNA level, 14,881 potential targets were altered at the m6A level and 12,507 potential targets were altered at the translation level; the targets of the three levels exhibited considerable overlap, indicating the complex roles of METTL3 (Figure 4B). All the WER targets constituted a complicated network, suggesting that these WERs function in a cooperative manner (Figures 4C and S3B). These targets can also be grouped into various kinds of gene types, demonstrating their vital functions with WERs through different kinds of mechanisms (Figures 4D and S3C).

WER targets inferred from WER perturbation followed by high-throughput sequencing. (A) The number distribution of WER perturbations followed by high-throughput sequencing datasets. (B) The intersection among three types of downstream effects upon WER perturbation, including alterations in gene expression, m6A and translation efficiency, corresponding to RNA-seq, MeRIP-seq and Ribo-seq, respectively. (C) The association of WER targets inferred from high-throughput sequencing upon WER perturbation. Blue nodes represent readers, purple nodes represent writers and green nodes represent erasers. Node size indicates the number of target associations identified for this WER. The thickness of the edge indicates the number of target associations that is validated by both WERs. (D) The gene type distribution of WER targets in the perturbation module of m6A2Target; others refer to those low frequency gene types, such as miscRNA and snoRNA.
Figure 4

WER targets inferred from WER perturbation followed by high-throughput sequencing. (A) The number distribution of WER perturbations followed by high-throughput sequencing datasets. (B) The intersection among three types of downstream effects upon WER perturbation, including alterations in gene expression, m6A and translation efficiency, corresponding to RNA-seq, MeRIP-seq and Ribo-seq, respectively. (C) The association of WER targets inferred from high-throughput sequencing upon WER perturbation. Blue nodes represent readers, purple nodes represent writers and green nodes represent erasers. Node size indicates the number of target associations identified for this WER. The thickness of the edge indicates the number of target associations that is validated by both WERs. (D) The gene type distribution of WER targets in the perturbation module of m6A2Target; others refer to those low frequency gene types, such as miscRNA and snoRNA.

Web interface and usage

M6A2Target is publicly available at http://m6a2target.canceromics.org. Users can browse, search and download all related data of m6A WERs and their targets through our web interface. M6A2Target is mainly composed of five modules, including ‘Validated Targets’, ‘Potential Targets’ and ‘Genomic Browser’, ‘Search’ and ‘Download.’

In the ‘Validated Targets’ module (Figure 5A), users can browse validated targets in specific species, specific cell lines or specific types of WER. Moreover, users can browse the validated targets for a specific WER by clicking the name of the WER of interest. By clicking the ‘Detail’ button for a specific target in the interactive table, users can obtain detailed information of the selected WER-target pair, including detailed information of the WER and target gene, as well as detailed information for the interaction between the WER and target (Figure S4A).

Web interface and usage of m6A2Target. (A) Main interface of Validated Targets module of m6A2Target. (B) Main interface of targets with the binding evidence module of m6A2Target. (C) Graphic interface of the genome browser in m6A2Target, METTL3 knockdown as an example. (D) Main interface of the Search function in m6A2Target. (E) Main interface of the download function of m6A2Target.
Figure 5

Web interface and usage of m6A2Target. (A) Main interface of Validated Targets module of m6A2Target. (B) Main interface of targets with the binding evidence module of m6A2Target. (C) Graphic interface of the genome browser in m6A2Target, METTL3 knockdown as an example. (D) Main interface of the Search function in m6A2Target. (E) Main interface of the download function of m6A2Target.

Application of m6A2Target. (A) Mutation rates of all identified WERs among 33 cancer types in TCGA. (B) Differential expression of KIAA1429 in tumor tissues compared to normal tissues in TCGA. (C) Forest plot for survival rate of KIAA1429 among different kinds of cancers in TCGA. (D) Intersection of KIAA1429 target genes in different submodules of m6A2Target, including target genes with alteration in m6A level, target genes with alteration in gene expression level and target genes with binding evidence with KIAA1429. (E) Pathway enrichment for 130 intersected target genes of KIAA1429 in m6A2Target. (F) m6A level of two genes in the PI3K-AKT signaling pathway followed by KIAA1429 knock down.
Figure 6

Application of m6A2Target. (A) Mutation rates of all identified WERs among 33 cancer types in TCGA. (B) Differential expression of KIAA1429 in tumor tissues compared to normal tissues in TCGA. (C) Forest plot for survival rate of KIAA1429 among different kinds of cancers in TCGA. (D) Intersection of KIAA1429 target genes in different submodules of m6A2Target, including target genes with alteration in m6A level, target genes with alteration in gene expression level and target genes with binding evidence with KIAA1429. (E) Pathway enrichment for 130 intersected target genes of KIAA1429 in m6A2Target. (F) m6A level of two genes in the PI3K-AKT signaling pathway followed by KIAA1429 knock down.

The ‘Potential Targets’ module includes two submodules, named ‘Binding’ and ‘Perturbation.’ Similar to the ‘Validated Targets’ module, users can browse all potential targets with binding evidence in the ‘Binding’ module (Figure 5B) and browse all the potential targets with perturbation evidence in the ‘Perturbation’ module (Figure S4B). Specifically, the page below in this module will display some statistical information about targets of this user selected WER, including target numbers per RNA type (Figure S4C), target numbers per cell type (Figure S4D) and the overview of these targets (Figure S4E). In addition, we also provided a pathway enrichment function in each module (Figure S4F), and users can conveniently obtain enriched pathways of user-selected WER target genes.

In the ‘Genomic browser’ module (Figure 5C), we provided an IGV genomic browser to visualize the targets. The ‘Search’ page provided search functions to help users to explore m6A2Target. Users could search any gene of interest to check whether it is a validated or potential target of any WER (Figure 5D). We also provided a ‘Download’ page (Figure 5E) to allow users to download all validated targets and potential targets in human or mouse species. Genome Reference Consortium Human Build 38 (hg38) was used as the human species reference genome, and for mouse species, Genome Reference Consortium Mouse Build 38 (mm10) was used. ‘Validated Target’ contains all targets in the validation submodule, ‘Potential Target (binding)’ contains all targets in the binding submodule and ‘Potential Target (perturbation)’ corresponds to the perturbation submodule.

Application of m6A2Target in cancer studies

Through analysis of all WER mutation rates in 33 cancer types in TCGA, we found that a well-known writer, KIAA1429, also known as VIRMA, had the highest mutation rate among all m6A WERs (Figure 6A). The expression level of KIAA1429 was significantly different between tumor samples and normal samples in several kinds of cancer, such as LIHC and HNSC (Figure 6B). Furthermore, the expression level of KIAA1429 was tightly associated with overall survival in multiple cancers, where high expression of KIAA1429 indicated better survival in BRCA, KICH, KIRP, PAAD, BRAC and UCEC but worse survival in KIRC and THYM (Figure 6C). Collectively, we considered that KIAA1429 played important roles in cancer progression. We then applied our database m6A2Target to explore the targets of KIAA1429 to obtain clues for downstream mechanistic studies of KIAA1429 in cancer progression. To obtain a reliable list of targets of KIAA1429, the targets with binding evidence and the targets with perturbation evidence (including m6A level targets and gene expression level targets) overlapped, which enabled 130 targets to be obtained (Figure 6D). The 130 targets were enriched in many cancer-related pathways, such as focal adhesion and the PI3K-AKT signaling pathway (Figure 6E). For example, the m6A levels of ITGAV and YWHAE in the PI3K-AKT signaling pathway were significantly hypomethylated in KIAA1429 knockdown cells compared to wild-type cells (Figure 6F). The above results suggest that m6A2Target may help researchers to investigate the m6A-related functions and mechanisms of genes of interest.

Discussion and conclusions

As the most abundant internal RNA modification, m6A is currently one of the most heavily researched subjects, and many outstanding researchers have verified its vital roles in the regulation of many fundamental biological processes and the occurrence and development of some diseases. m6A2Target provides targets of all identified WERs in human and mouse species and some related detailed comprehensive information. Currently, m6A2Target contains 1034 WER-target pairs validated by experiments, 124,772 WER-target pairs interacting with WERs and 330,102 WER-target pairs with differential expression levels, differential methylation levels, differential translation efficiencies or differential alternative splicing levels.

Compared to other m6A-related databases, m6A2Target is the first comprehensive and specific resource focused on the targets of m6A writers, erasers and readers. This resource covers human and mouse species and contains various abundances of information for targets of all identified WERs. The m6A2Target website is convenient and user-friendly, and users can browse all m6A-associated genes and search their targets of interest by various criteria in m6A2Target. Users can also obtain detailed information about WERs, targets and WER-target associations by simply clicking the ‘detail’ button. If users want to obtain more detailed information about this WER or its target gene, m6A2Target also provides links that help our users go to the corresponding official website, including Ensembl (http://asia.ensembl.org/index.html), HGNC (https://www.genenames.org/), UniProt (https://www.uniprot.org/) and NCBI (https://www.ncbi.nlm.nih.gov/).

m6A2Target may help researchers to characterize m6A modifications and their clinical significance more thoroughly. For instance, we observed that target genes of METTL14 are enriched in pathways related to the PI3K-AKT-mTOR signaling pathway and endometrial cancer pathway, and target genes of METTL3 are also enriched in endometrial cancer. He et al. proved the crucial roles of m6A methylation in the proliferation and tumorigenicity of endometrial cancer by regulating AKT activity [60]. These researchers found that approximately 70% of endometrial tumors presented reduced m6A methylation levels, which may be due to either METTL14 mutation or reduced METTL3 expression; this finding is highly consistent with the pathway enrichment results obtained with our database.

In addition, m6A2Target also has a limitation. At present, we only collected m6A WER-related data for the two most frequently used species in m6A studies, that is, human and mouse species, which may cause our results to be slightly limited. We will attempt to solve this problem in the updated versions of our database in the future.

Overall, the results of this study indicate that m6A2Target may enable researchers in the m6A community to obtain more detailed information about m6A writers, erasers, readers and their targets, thereby establishing a foundation for further research investigating m6A. We will update the data regularly as m6A-related studies are published.

Conflict of interest statement

The authors have declared that no competing interests exist.

Shuang Deng, Hongwan Zhang, Kaiyu Zhu, Xingyang Li, Ying Ye, Rui Li, Xuefei Liu, Zhixiang Zuo, Jian Zheng Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.

Dongxin Lin Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.

References

1.

Davis
FF
,
Allen
FW
.
Ribonucleic acids from yeast which contain a fifth nucleotide
.
J Biol Chem
1957
;
227
:
907
15
.

2.

Machnicka
MA
,
Milanowska
K
,
Osman Oglou
O
, et al.
MODOMICS: a database of RNA modification pathways–2013 update
.
Nucleic Acids Res
2013
;
41
:
D262
7
.

3.

Dominissini
D
,
Moshitch-Moshkovitz
S
,
Schwartz
S
, et al.
Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq
.
Nature
2012
;
485
:
201
6
.

4.

Wickramasinghe
VO
,
Laskey
RA
.
Control of mammalian gene expression by selective mRNA export
.
Nat Rev Mol Cell Biol
2015
;
16
:
431
42
.

5.

Batista
PJ
,
Molinie
B
,
Wang
J
, et al.
M(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells
.
Cell Stem Cell
2014
;
15
:
707
19
.

6.

Bartosovic
M
,
Molares
HC
,
Gregorova
P
, et al.
N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3′-end processing
.
Nucleic Acids Res
2017
;
45
:
11356
70
.

7.

Meyer
KD
,
Patil
DP
,
Zhou
J
, et al.
5' UTR m(6)A promotes cap-independent translation
.
Cell
2015
;
163
:
999
1010
.

8.

Frayling
TM
,
Timpson
NJ
,
Weedon
MN
, et al.
A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity
.
Science
2007
;
316
:
889
94
.

9.

Dina
C
,
Meyre
D
,
Gallina
S
, et al.
Variation in FTO contributes to childhood obesity and severe adult obesity
.
Nat Genet
2007
;
39
:
724
6
.

10.

Li
Z
,
Weng
H
,
Su
R
, et al.
FTO plays an oncogenic role in acute myeloid leukemia as a N(6)-Methyladenosine RNA Demethylase
.
Cancer Cell
2017
;
31
:
127
41
.

11.

Ma
JZ
,
Yang
F
,
Zhou
CC
, et al.
METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N(6) -methyladenosine-dependent primary MicroRNA processing
.
Hepatology
2017
;
65
:
529
43
.

12.

Shen
F
,
Huang
W
,
Huang
JT
, et al.
Decreased N(6)-methyladenosine in peripheral blood RNA from diabetic patients is associated with FTO expression rather than ALKBH5
.
J Clin Endocrinol Metab
2015
;
100
:
E148
54
.

13.

Yang
Y
,
Huang
W
,
Huang
JT
, et al.
Increased N6-methyladenosine in human sperm RNA as a risk factor for Asthenozoospermia
.
Sci Rep
2016
;
6
:
24345
.

14.

Daoud
H
,
Zhang
D
,
McMurray
F
, et al.
Identification of a pathogenic FTO mutation by next-generation sequencing in a newborn with growth retardation and developmental delay
.
J Med Genet
2016
;
53
:
200
7
.

15.

Liu
J
,
Yue
Y
,
Han
D
, et al.
A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation
.
Nat Chem Biol
2014
;
10
:
93
5
.

16.

Ping
XL
,
Sun
BF
,
Wang
L
, et al.
Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase
.
Cell Res
2014
;
24
:
177
89
.

17.

Zheng
G
,
Dahl
JA
,
Niu
Y
, et al.
ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility
.
Mol Cell
2013
;
49
:
18
29
.

18.

Jia
G
,
Fu
Y
,
Zhao
X
, et al.
N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO
.
Nat Chem Biol
2011
;
7
:
885
7
.

19.

Wang
X
,
Zhao
BS
,
Roundtree
IA
, et al.
N(6)-methyladenosine modulates messenger RNA translation efficiency
.
Cell
2015
;
161
:
1388
99
.

20.

Li
A
,
Chen
YS
,
Ping
XL
, et al.
Cytoplasmic m(6)A reader YTHDF3 promotes mRNA translation
.
Cell Res
2017
;
27
:
444
7
.

21.

Ivanova
I
,
Much
C
,
Di Giacomo
M
, et al.
The RNA m(6)A reader YTHDF2 is essential for the post-transcriptional regulation of the maternal Transcriptome and oocyte competence
.
Mol Cell
2017
;
67
:
1059
1067.e1054
.

22.

Wang
Y
,
Li
Y
,
Toth
JI
, et al.
N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells
.
Nat Cell Biol
2014
;
16
:
191
8
.

23.

Yue
Y
,
Liu
J
,
Cui
X
, et al.
VIRMA mediates preferential m(6)A mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation
.
Cell Discov
2018
;
4
:
10
.

24.

Huang
H
,
Weng
H
,
Sun
W
, et al.
Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation
.
Nat Cell Biol
2018
;
20
:
285
95
.

25.

Hsu
PJ
,
Zhu
Y
,
Ma
H
, et al.
Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis
.
Cell Res
2017
;
27
:
1115
27
.

26.

Kasowitz
SD
,
Ma
J
,
Anderson
SJ
, et al.
Nuclear m6A reader YTHDC1 regulates alternative polyadenylation and splicing during mouse oocyte development
.
PLoS Genet
2018
;
14
:
e1007412
.

27.

Roundtree
IA
,
Luo
GZ
,
Zhang
Z
, et al.
YTHDC1 mediates nuclear export of N(6)-methyladenosine methylated mRNAs
.
Elife
2017
;
6
:
e31311
.

28.

Xiao
W
,
Adhikari
S
,
Dahal
U
, et al.
Nuclear m(6)A reader YTHDC1 regulates mRNA splicing
.
Mol Cell
2016
;
61
:
507
19
.

29.

Dominissini
D
,
Moshitch-Moshkovitz
S
,
Salmon-Divon
M
, et al.
Transcriptome-wide mapping of N(6)-methyladenosine by m(6)A-seq based on immunocapturing and massively parallel sequencing
.
Nat Protoc
2013
;
8
:
176
89
.

30.

Grozhik
AV
,
Linder
B
,
Olarerin-George
AO
, et al.
Mapping m(6)A at individual-nucleotide resolution using crosslinking and Immunoprecipitation (miCLIP)
.
Methods Mol Biol
2017
;
1562
:
55
78
.

31.

Stork
C
,
Zheng
S
.
Genome-wide profiling of RNA-protein interactions using CLIP-Seq
.
Methods Mol Biol
2016
;
1421
:
137
51
.

32.

Raha
D
,
Hong
M
,
Snyder
M
.
ChIP-Seq: a method for global identification of regulatory elements in the genome
.
Curr Protoc Mol Biol
2010
;
Chapter 21:Unit 21.19.21-14
.

33.

Danan
C
,
Manickavel
S
,
Hafner
M
.
PAR-CLIP: a method for Transcriptome-wide identification of RNA binding protein interaction sites
.
Methods Mol Biol
2016
;
1358
:
153
73
.

34.

Domon
B
,
Aebersold
R
.
Mass spectrometry and protein analysis
.
Science
2006
;
312
:
212
7
.

35.

Lin
JS
,
Lai
EM
.
Protein-protein interactions: co-Immunoprecipitation
.
Methods Mol Biol
1615
;
2017
:
211
9
.

36.

Abakir
A
,
Giles
TC
,
Cristini
A
, et al.
N(6)-methyladenosine regulates the stability of RNA:DNA hybrids in human cells
.
Nat Genet
2019
;
52
:
48
55
.

37.

Barrett
T
,
Wilhite
SE
,
Ledoux
P
, et al.
NCBI GEO: archive for functional genomics data sets--update
.
Nucleic Acids Res
2013
;
41
:
D991
5
.

38.

Leinonen
R
,
Sugawara
H
,
Shumway
M
.
The sequence read archive
.
Nucleic Acids Res
2011
;
39
:
D19
21
.

39.

Weinstein
JN
,
Collisson
EA
,
Mills
GB
, et al.
The cancer genome atlas pan-cancer analysis project
.
Nat Genet
2013
;
45
:
1113
20
.

40.

Chen
S
,
Zhou
Y
,
Chen
Y
, et al.
Fastp: an ultra-fast all-in-one FASTQ preprocessor
.
Bioinformatics
2018
;
34
:
i884
90
.

41.

Langmead
B
,
Salzberg
SL
.
Fast gapped-read alignment with bowtie 2
.
Nat Methods
2012
;
9
:
357
9
.

42.

Zhang
Y
,
Liu
T
,
Meyer
CA
, et al.
Model-based analysis of ChIP-Seq (MACS)
.
Genome Biol
2008
;
9
:
R137
.

43.

Heinz
S
,
Benner
C
,
Spann
N
, et al.
Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities
.
Mol Cell
2010
;
38
:
576
89
.

44.

Shah
A
,
Qian
Y
,
Weyn-Vanhentenryck
SM
, et al.
CLIP tool kit (CTK): a flexible and robust pipeline to analyze CLIP sequencing data
.
Bioinformatics
2017
;
33
:
566
7
.

45.

Langmead
B
.
Aligning short sequencing reads with bowtie
.
Curr Protoc Bioinformatics
2010
;
Chapter 11:Unit 11.17
.

46.

Corcoran
DL
,
Georgiev
S
,
Mukherjee
N
, et al.
PARalyzer: definition of RNA binding sites from PAR-CLIP short-read sequence data
.
Genome Biol
2011
;
12
:
R79
.

47.

Li
B
,
Dewey
CN
.
RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome
.
BMC Bioinformatics
2011
;
12
:
323
.

48.

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

49.

Dobin
A
,
Davis
CA
,
Schlesinger
F
, et al.
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
.

50.

Cui
X
,
Meng
J
,
Zhang
S
, et al.
A novel algorithm for calling mRNA m6A peaks by modeling biological variances in MeRIP-seq data
.
Bioinformatics
2016
;
32
:
i378
85
.

51.

Audic
S
,
Claverie
JM
.
The significance of digital gene expression profiles
.
Genome Res
1997
;
7
:
986
95
.

52.

Shen
S
,
Park
JW
,
Lu
ZX
, et al.
rMATS: robust and flexible detection of differential alternative splicing from replicate RNA-Seq data
.
Proc Natl Acad Sci U S A
2014
;
111
:
E5593
601
.

53.

Mayakonda
A
,
Lin
DC
,
Assenov
Y
, et al.
Maftools: efficient and comprehensive analysis of somatic variants in cancer
.
Genome Res
2018
;
28
:
1747
56
.

54.

Ritchie
ME
,
Phipson
B
,
Wu
D
, et al.
Limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res
2015
;
43
:
e47
.

55.

Ai
C
,
Kong
L
.
CGPS: a machine learning-based approach integrating multiple gene set analysis tools for better prioritization of biologically relevant pathways
.
J Genet Genomics
2018
;
45
:
489
504
.

56.

Wu
J
,
Mao
X
,
Cai
T
, et al.
KOBAS server: a web-based platform for automated annotation and pathway identification
.
Nucleic Acids Res
2006
;
34
:
W720
4
.

57.

Xie
C
,
Mao
X
,
Huang
J
, et al.
KOBAS 2.0: a web server for annotation and identification of enriched pathways and diseases
.
Nucleic Acids Res
2011
;
39
:
W316
22
.

58.

Kanehisa
M
,
Goto
S
.
KEGG: Kyoto encyclopedia of genes and genomes
.
Nucleic Acids Res
2000
;
28
:
27
30
.

59.

Robinson
JT
,
Thorvaldsdottir
H
,
Winckler
W
, et al.
Integrative genomics viewer
.
Nat Biotechnol
2011
;
29
:
24
6
.

60.

Liu
J
,
Eckert
MA
,
Harada
BT
, et al.
M(6)A mRNA methylation regulates AKT activity to promote the proliferation and tumorigenicity of endometrial cancer
.
Nat Cell Biol
2018
;
20
:
1074
83
.

Author notes

Shuang Deng and Hongwan Zhang contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)