Open Access

Detection of N6‑methyladenosine modification residues (Review)

  • Authors:
    • Wei Zhu
    • Jing‑Zi Wang
    • Zhiqiang Xu
    • Mengda Cao
    • Qiaoli Hu
    • Chen Pan
    • Miao Guo
    • Ji‑Fu Wei
    • Haiwei Yang
  • View Affiliations

  • Published online on: April 18, 2019     https://doi.org/10.3892/ijmm.2019.4169
  • Pages: 2267-2278
  • Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Among a number of mRNA modifications, N6‑methyladenosine (m6A) modification is the most common type in eukaryotes and nuclear‑replicating viruses. m6A has a significant role in numerous cancer types, including leukemia, brain tumors, liver cancer, breast cancer and lung cancer. Although m6A methyltransferases are essential during RNA modifications, the biological functions of m6A and the underlying mechanisms remain to be fully elucidated, predominantly due to the limited detection methods for m6A. In the present review, the currently available m6A detection methods and the respective scope of their applications are presented to facilitate the further investigation of the roles of m6A in biological process.

1. Introduction

Gene regulation via DNA and protein modifications has been intensively studied. However, the knowledge on RNA modifications remains insufficient (1). Among all mRNA modifications, N6-methyladenosine (m6A) modification is the most common type in eukaryotic RNA and occurs in three to five sites per transcript on average (2-4).

m6A was discovered in the 1970s in a wide range of cellular mRNAs (4-6). Methylation occurs at the sixth position of nitrogen atoms of adenosine at the post-transcriptional level with S-adenosylmethionine serving as the methyl donor for m6A formation, which is termed m6A modification (7-9).

The reversible activity of the m6A modification is regulated by the combined action of methylase and demethylase (10). The m6A methylase complex consists of at least five 'writer' proteins (11-14), among which methyltransferase like 3 (METTL3) protein serves a central role. METTL14 protein supports METTL3 protein structurally (15), WT1-associated protein (WTAP) regulates the recruitment of the m6A methyltransferase complex to mRNA targets (12) and the RNA-binding motif protein 15 serves a role in helping this complex move towards the appropriate m6A sites (16). Vir like m6A methyltransferase associated (VIRMA) is also a component of this m6A methylase complex; however, its molecular function remains largely unknown (13). METTL16 is a newly discovered m6A methyltransferase that primarily methylates m6A sites in the 3′-untranslated region (3′-UTR) of RNA. When METTL16 is knocked down, the level of m6A in the cell decreases by ~20% (17). Two reported demethylases that reverse m6A modification are FTO (18) and alkB homolog 5 (ALKBH5) (19). It has been confirmed that m6A levels increased following the knockdown of FTO and ALKBH5 expression (18,19). The identified 'readers' of m6A are YT521-B homology (YTH) domain-containing proteins, including YTHDF1-3, YTHDC1 and YTHDC2, which participate in the translation (20), stabilization (21), splicing (22) or nuclear export (23) of mRNA. The affinity of 'readers' has been reported to be higher compared with unmethylated mRNA for m6A-methylated mRNA (24).

m6A bases cannot be detected directly by sequencing because the m6A modification does not change the base pairing properties and cannot be distinguished from regular bases by reverse transcription (25,26). Previously, new methods have been developed to identify m6A modification in cells, which are based on immunoprecipitation or selective RNA chemistry to isolate modified RNA fragments and combined with high-throughput sequencing (3,25,27). Dot blot technology is frequently used to observe changes in m6A. However, dot blots cannot determine the quantitation and precise location of m6A (28). RNA photo-crosslinkers, quantitative proteomics and electrochemical immunosensor methods may be applied to detect the presence of m6A in cells; however, they cannot precisely determine the m6A modification sites (29,30). The newly developed methylated RNA immunoprecipitation sequencing (MeRIP-seq) method combines m6A antibody immunoprecipitation and deep sequencing to identify the m6A residues in 100-200 nucleotide RNA segments (3,25). However, this approach is complicated as m6A often appears in clusters since multiple different m6A-containing fragments generate overlapping reads, which can result in large peaks spanning several m6A residues (2). Thus, the summit of these peaks may not accurately reflect the positions of m6A residues. By contrast, when using the m6A individual-nucleotide-resolution cross-linking and immunoprecipitation (miCLIP) technique, identification of m6A residues is not influenced by the peak shapes (31). Furthermore, identification of m6A residues by miCLIP is not restricted to a specified subset of DRACH (D=A, G) motifs (2,3,13). Therefore, miCLIP can properly identify m6A residues (31).

High-performance liquid chromatography (HPLC) or mass spectrometry are applied in some of the abovementioned methods to detect specific modified RNA or RNA bases (3,29). Isolated RNA is fragmented into nucleosides prior to analysis, which may change the original RNA structure. Dot blot technology, which does not require fragmentation, is used as an alternative method to detect specific modifications. However, dot blot technology is limited in that RNA samples cannot be separated by the size, and hence, it is impossible to distinguish the targeted specific RNA in the RNA samples (32,33). Notably, HPLC can be used alone to measure m6A modification rate (18).

The abovementioned methods and specific references are provided in Table I (18,26,27,29-51). In the present review, the most recent information on the currently established m6A detection methods is presented and discussed.

Table I

Methods for detecting m6A residues.

Table I

Methods for detecting m6A residues.

Method(Refs.)
Semi-quantitative
 Dot blot technology(32,33)
 Methyl sensitivity of MazF RNA endonucleases(34)
 Immune-northern blot(35,36)
Quantitative
 RNA photo-crosslinkers and quantitative proteomics(29)
 Electrochemical immunosensor method(30)
 Support vector machine-based method(37)
Detection of precise locations
 HRM(38)
 LAIC(27,39)
 SCARLET(39-41)
 MeRIP-seq(42-46)
 MiClip(31,39,47)
 DNA polymerase for direct m6A sequencing(26,48)
 HPLC(18,49-51)

[i] m6A, N6-methyladenosine; miCLIP, m6A individual-nucleotide-resolution cross-linking and immunoprecipitation; HRM, high-resolution melting analysis; SCARLET, site-specific cleavage and radioactive labeling followed by ligation-assisted extraction and thin layer chromatography; LAIC, level and isoform characterization; MeRIP-seq, methylated RNA immunoprecipitation sequencing; MiClip, m6A individual-nucleotide-resolution cross-linking and immunoprecipitation; HPLC, high-performance liquid chromatography.

2. Functions of m6A modifications

m6A is the most common type of RNA residue modification in eukaryotes (2-4). However, its specific biological roles remain largely unknown. Studies have indicated that the dynamic regulation of m6A has a significant impact on the control of gene expression (20,21). m6A-seq has revealed that m6A predominantly exists on exons and the 3′-UTR of mRNA (2). METTL3, FTO and ALKBH5 have been identified to serve significant roles in biological regulation, including development, metabolism and fertility (18,19).

FTO, an m6A demethylase, is recognized as a major obesity factor. It belongs to the ALKB enzyme family, which oxidatively demethylates m6A on mRNA (32,52). ALKBH5 is another mammalian m6A demethylase, and is associated with certain mRNA-processing factors in nuclear speckles, which may influence RNA export and metabolism (19). ALKBH5-deficient mice have differential expression of genes involved in the p53 signaling pathway and spermatogenesis (19), suggesting a global role for m6A in human health. METTL3 is expressed in all human tissues and is highly expressed in the testis (10,11). It has been identified that METTL3 can participate in tumor growth and progression by regulating the cell cycle of cancer cells (53,54). WTAP was primarily recognized as a protein associated with Wilm's tumor and has been indicated to exhibit a significant role in cell cycle progression (12). WTAP also participates in RNA splicing and results in embryonic defects (12). Cooperation between the RNA m6A methyltransferase complex and the demethylases establishes a reversible regulation of RNA m6A modifications. Taken together, RNA m6A modifications exhibit significant roles in the molecular mechanisms of gene biology, as illustrated in Fig. 1.

3. Methods used to analyze m6A modifications

Dot blot

Dot blot (or slot blot) technology is used in molecular biology as a semiquantitative or quantitative detection method for DNA, RNA and protein samples, and is predominantly used semiquantitatively in m6A analysis (32,33,55-57). Compared with western blotting, northern blotting or Southern blotting methods, dot blot technology has a simpler operation with a similar working principle. The biomolecules are not subjected to electrophoretic separation prior to detection with the dot blot method. Instead, samples containing a mixture of RNAs are directly applied to a membrane through an apparatus with circular templates that form a dot when the sample is applied. After applying a vacuum to embed RNAs and dry the membrane, biomolecules are detected with antibodies. When detecting RNA, the DNA must be removed prior to loading to eliminate its influence on m6A (32,33). A schematic diagram of the determination of m6A modification residues by dot blot is presented in Fig. 2.

Dot blot technology markedly saves time, since it does not require chromatography, gel electrophoresis or complex gel blocking procedures (32,33). However, regarding m6A detection, dot blot technology is only able to verify the presence of m6A or compare the amounts of m6A between different groups (32,33). Li et al (32) improved the traditional dot blot method to measure the global m6A abundance in the transcriptomes of four acute myeloid leukemia cell lines. However, this method is still not able to quantitate or precisely determine the location of m6A.

Methyl-sensitive MazF RNA endonucleases

The regulatory enzymes of m6A have been reported to contribute to tumorigenesis (58-60). While the significance of m6A has been confirmed, no convenient approach has been developed to analyze m6A methyltransferase and demethylase activities or to monitor the inhibitors of these activities.

The Escherichia coli toxin MazF exhibits endoribonuclease activity specific against ACA sequences and is susceptible to m6A. MazF is the first enzyme discovered with the ability to specifically cleave RNA containing m6A, and is used to study activities of m6A demethylase and methyltransferase (34). Furthermore, MazF has an application for monitoring the inhibitors of m6A methyltransferase and demethylase. RNA cleavage by MazF may be detected by polyacrylamide gel electrophoresis and the fluorescence-resonance energy transfer-based plate assay (61). A schematic diagram of the determination of m6A modification residues by methyl-sensitive MazF RNA endonucleases is presented in Fig. 3.

At the current stage of development, the MazF cleavage methods are restricted regarding the evaluation of m6A, as MazF is only able to cleave the 5′-ACA-3′ site in single-stranded RNA, frequently occurring in endogenous RNAs. However, MazF is not able to cleave the 5′-ACA-3′ site in double-stranded RNA (61), and thus, it may not accurately determine the presence of m6A in structured RNA.

Immuno-northern blot

The immuno-northern blot combines a northern blotting experimental program and m6A-binding antibody, which is different from conventional northern blot techniques that use DNA probes. Immuno-northern blot does not require RNA fragmentation prior to analysis and the RNAs are separated based on their molecular weights (35). Thus, the detection of m6A modifications by immuno-northern blot is applied in various types of RNA (35).

In brief, RNAs are separated in a denaturing acryl-amide gel or an agarose gel and then transferred onto nylon membranes. The RNA strands on the membranes are exposed to ultraviolet (UV) irradiation for cross-linking, followed with incubation with primary antibodies against m6A modifications, corresponding secondary antibody and chemiluminescent detection (35).

4. Quantification of m6A modifications

RNA photo-crosslinkers and quantitative proteomics

The regulatory role of mRNA predominantly depends on the interaction between mRNA and RNA-binding proteins to regulate RNA splicing, stability, localization and translation (62). Photo-crosslinking technologies are diffusely applied to stabilize direct protein-RNA interactions (63). These technologies depend on the tendency for UV-induced photochemistry of nucleobases, which are natural or derivatives containing sulfur or halogen substituents. Photo-affinity labels, including diazirine (64) or benzophenone (65), are not widely used in the analysis of protein-RNA interactions; however, they may be activated by longer wavelengths and provide more efficient crosslinking.

Arguello et al (29) developed a chemical proteomics approach based on photo-crosslinking of the RNA base and diazirine, which was highly efficient in quantitatively analyzing protein-RNA interactions regulated by m6A modification. By using this method, novel m6A 'readers' have been discovered. To isolate m6A readers with photosynthetic and quantitative proteomics, RNA probes are required to contain the following: i) m6A molecules; ii) a photo-crosslinker that is efficient and does not influence the protein-RNA interactions; iii) streptavidin as an affinity handle for protein enrichment. A probe was prepared that contains the sequence GGm6ACU, the common recognition pattern of the m6A site in mammalian cells. This sequence is indispensable for binding the YTH-domain proteins (66,67). The probe was validated by known m6A RNA 'readers', including YTHDF1 (20), YTHDF2 (21), YTHDF3 (3,22,68) and YTHDC1 (22). A schematic diagram of the detection of m6A modification residues by the RNA photo-crosslinkers and quantitative proteomics technologies is presented in Fig. 4. We hypothesize that the requirement of the synthesis of the sensor may be a limitation regarding these techniques.

Electrochemical immunosensor method

The majority of the abovementioned analytic methods are difficult to perform and expensive. The electrochemical immunosensor method was developed to provide convenience and high sensitivity (30). An anti-m6A antibody has been used to detect m6A by targeting m6A-5′-triphosphate (m6ATP). The detection and capture of m6A relies on silver nanoparticles and SiO2 (Ag@SiO2) nano-spheres with amine-polyethylene glycol 3-biotin. Ag@SiO2 nanospheres were prepared to amplify signals. Phos-tag-biotin was prepared to link m6ATP and Ag@SiO2 through two types of specific interaction between phosphate group of m6ATP and phos-tag, biotin and streptavidin, respectively. Experiments for evaluating this strategy indicated that the immuno-sensor has acceptable reproducibility and specificity with a wide linear range and a low detection limit. A schematic diagram for the determination of m6A modification residues by the electrochemical immunosensor method is presented in Fig. 5.

The efficacy of the detection of the m6A content using the electrochemical immunosensor method had been verified in human cell lines (30). It also provides a technological basis for the detection of RNAs and DNAs with the advantages of convenience, low cost, and high specificity and sensitivity.

Vector method to detect m6A sites

High-throughput next-generation sequencing-based technology for identifying m6A sites based on where adenosine is methylated has not been applied in most species. In recent years, a vector machine method was developed to identify m6A sites in Arabidopsis thaliana (37). When combining anti-m6A antibodies and high-throughput sequencing, Luo et al (69) obtained thousands of m6A peaks for A. thaliana, including 'common' m6A peaks. Since the RRACH motif, where R resembles purine, A stands for m6A and H resembles a non-guanine base (69), was identified in most of the m6A peaks, Chen et al (37) collected segments containing RRACH at the center of the 'common' m6A peaks and proposed a model that may accurately identify specific m6A sites with high accuracy.

If the model is adapted to other plant species other than A. thaliana, this vector machine-based method may be used for the detection of m6A and other post-transcriptional modifications in these other plants as well.

5. Methods to determine m6A residue locations

HRM

HRM analysis is a simple method to detect m6A modification residues at a specific location in RNAs (38). HRM may be applied to high-throughput measurement. The resulting HRM curves of the samples of RNA mixtures change steadily from 100% of methylated RNA to 100% of unmethylated RNA (38). As presented in Fig. 6, the detection of m6A modification residues by the HRM method relies on the modified nucleoside position at a particular site of RNA and is followed by rapid screening for conditions or genes necessary for analysis of that modification (38).

According to the specificity of the oligonucleotide probe hybridization, bulk cellular RNA, as opposed to purified specific RNA, has been designed for detecting m6A at a pre-defined position (38). In addition, partial non-ribosomal target enrichments may be easily accomplished using commercially available kits.

A possible application for this method would be to screen knockout/knockdown strain libraries to identify genes contributing to the formation of a specific m6A nucleoside. Another possible application is to detect the presence of a particular m6A nucleoside under different growth or environmental conditions (2,3). HRM analysis may help to elucidate the dynamic events that result in the modification of certain RNAs.

m6A level and isoform-characterization sequencing (m6A-LAIC-seq)

For thorough investigation of the m6A epitranscriptome, Molinie et al (27) invented the m6A-LAIC-seq technique. Combined with RNA IP whole-transcriptome sequencing, m6A-LAIC-seq may quantify m6A contents, with spike-in RNAs as an internal standard. A schematic diagram for the determination of m6A modification residues by m6A-LAIC-seq is presented in Fig. 7. The results demonstrate a quantitative road map to which genes are the most or the least likely to be influenced by m6A-dependent regulatory networks. This method may determine the m6A levels in each gene but cannot stoichiometrically analyze the methylation of a single modified nucleotide. m6A-LAIC-seq complements m6A-seq identification of methylation sites and helps to expand the understanding of the biology of m6A.

Site-specific cleavage and radioactive labeling followed by ligation-assisted extraction and thin-layer chromatography (SCARLET)

To elucidate the dynamic biological functions of m6A, Liu et al (40) provided the SCARLET method that directly measures the precise location and the status of m6A modification at any candidate site of mRNA/lncRNA at a single nucleotide resolution. In addition to m6A, SCARLET may be used to observe other RNA modifications, including 5-methylcytosine, pseudouridine and 2′-O-methyl ribonucleosides. SCARLET is available to study the biological functions of RNA modifications with general experimental equipment and materials. A schematic diagram for the determination of m6A modification residues by SCARLET is presented in Fig. 8.

The feasibility of using the SCARLET method has been confirmed in HeLa cell RNA samples, which produced similar results compared with previously reported m6A sites in Hela samples. Using this method, a minimally modified m6A site that was not precisely determined in a preceding study was also identified, demonstrating that SCARLET is able to easily resolve the ambiguity of modification sites (70).

MeRIP-Seq

Although RNA methylation has been identified and verified in the 1970s, the relevant modification mechanism, regulatory means and biological significance have not been clarified due to technical limitations. The recent emergence of MeRIP-Seq technology makes it possible to study m6A methylation at the transcriptome level by high-throughput sequencing (2,3).

MeRIP-Seq is a combination of ChIP-Seq and RNA-Seq that is able to elucidate global mRNA m6A sites in mammalian cells. MeRIP-seq is a novel type of IP-seq technology, in which the known ChIP-seq, photoactivatable ribonucleo-side-enhanced crosslinking and IP (a more mature sequence), is applied. MeRIP-seq has been successfully used to detect whole-genome m6A modifications (42-46). The major principle of this technique is as follows: The anti-m6A antibody is incubated with randomly interrupted RNA fragments using a co-IP method resulting in an m6A modification fragment that is precipitated and sequenced. Concurrently, a control sample is run that eliminates the background during antibody capture (2).

Successful preparations of each library should be evaluated prior to massive parallel sequencing. To identify and localize the m6A sites at a transcriptome-wide level by m6A IP, fragmentation of poly(A)+-selected RNAs (input) is required prior to IP with anti-m6A antibodies. The input and m6A IP RNAs are then separately processed for next-generation sequencing (2). Zhang et al (48) first proposed a Bayesian statistical model, BaySeqPeak, to analyze MeRIP-Seq data to help discover methylation site signals in the transcriptome. A reference transcriptome was prepared, which contains a single, non-intron splice variant of each gene. The resultant reads were then specifically compared with the reference to identify m6A sites with a low false-detection rate (3). A schematic diagram for the determination of m6A modification residues by MeRIP-Seq is presented in Fig. 9.

MiClip

It has been reported that m6A residues may be located by producing unique signature mutations with anti-m6A antibodies and UV crosslinking techniques (31). m6A residues were mapped with two antibodies, one of which translates C to T to detect single and clustered m6A residues, and the other antibody that produces truncations is used to determine the position of m6A sites and detect m6A residues concurrently. A schematic diagram for the determination of m6A modification residues by miClip is provided in Fig. 10.

Identification of m6A residues by direct detection is superior to that by bioinformatics predictions from MeRIP-Seq peaks (31). The reliability of bioinformatics prediction depends on the characteristics of the m6A peak. MeRIP-Seq can accurately predict m6A residues only with a single clear peak of a single m6A residue and is limited to the centrally-located DRACH motif (31). m6A residues usually cluster in mRNAs, resulting in multiple MeRIP-Seq peaks (2). Identification of m6A residues using miCLIP technology is not influenced by peak shapes and not confined to the centrally-located DRACH motif. Therefore, miCLIP can correctly identify m6A residues.

A DNA polymerase for direct m6A sequencing

At present, RNA samples are prepared by antibody-based enrichment of m6A residues prior to sequencing, as m6A modifications are usually lost after reverse transcription (25,26). The indirect detection may lead to a higher error rate, pushing the generation of novel DNA polymerase to sequence m6A directly (26). In this light, Aschenbrenner et al (26) developed a screening method to develop a reverse transcription-active KlenTaq DNA polymerase variant for labeling N6-methylation residues. A schematic diagram for the determination of m6A modification residues by the DNA polymerase for direct N6-methyladenosine sequencing is presented in Fig. 11.

HPLC

The development of HPLC occurred only 30 years ago; however, the development of this separation analysis technology is very rapid, and is now widely used, including in the detection of m6A modification (50). HPLC was firstly used to detect m6A modification in nine DNAs (50). Subsequently, Rana and Tuck (49) applied HPLC in the detection of m6A modification in a T7 RNA transcript coding for mouse dihydrofolate reductase by separating m6A from adenine, cytosine, uracil and so on. In a study by Jia et al (18), HPLC helped observe the changes of the m6A ratio in mRNAs following FTO treatment to better understand the function of FTO. The development of this in vitro methylation assay opened the door for studies investigating m6A levels in specific mRNAs or learn the biological significance of m6A modification.

6. Discussion

Detection of RNA modifications and study of their functions are emerging fields of research. The potential role of m6A modifications in regulating molecular and physiological processes in several organisms, particularly in RNA stability, splicing, transport, localization and translation, is valued (19,20,52,21,70-73). With the discovery of 'reader' proteins, the downstream molecular mechanisms of m6A modifications are gradually being clarified (21). There is strong evidence that m6A methylation is associated with RNA splicing and that 'readers' of m6A reduce the stability of RNA transcripts (21). After removal of m6A, a second class of proteins may bind to the RNAs, which may be affected by changes of the RNA secondary structure from m6A additions (74). Thus, the physiological effects of m6A modifications should be observed at multiple levels, including the tissue level, the pathway level, the cellular level and the molecular level (75). An increasing number of studies have suggested a link between m6A RNA modifications with cancer and other similar disease-associated processes (32,53,54,73,74-76). Detection of m6A modification in vitro can help identify the precise regulatory forms and synergistic roles of m6A modifications in cancer and other diseases (32,53,54,74-76). Certain m6A methylases, m6A demethylases or downstream genes have become prognostic factors for different cancer types (77-79). Certain abovementioned methods, including MeRIP-seq, could help identify the downstream genes and mutation sites (54,80). Therefore, detection of them in vitro may help diagnose and predict the final progress. The present review provides an overview of these methods to drive the elucidation of the biological roles of m6A and encourage their further development.

Funding

This project was supported by grants from the National Natural Science Foundation of China (grant nos. 81571568 and 81871265), the CAMS Innovation Fund for Medical Sciences (grant no. CIFMS:2016-I2M-1003), the Innovation Team of Jiangsu Provincial Commission of Health and Family Planning (grant no. CXTDA2017049).

Availability of data and materials

Not applicable.

Authors' contributions

JWe and HY designed the study and revised the article. WZ and JWa reviewed the literature and wrote the article. ZX, MC, QH, CP and MG reviewed the literature and revised the article. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflict of interests.

Acknowledgments

Not applicable.

References

1 

He C: Grand challenge commentary: RNA epigenetics. Nat Chem Biol. 6:863–865. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Meye KD, Saletore Y, Zumbo P, Elemento O, Mason CE and Jaffrey SR: Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 149:1635–1646. 2012. View Article : Google Scholar

3 

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al: Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 485:201–206. 2012. View Article : Google Scholar : PubMed/NCBI

4 

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

5 

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

6 

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

7 

Narayan P and Rottman FM: Methylation of mRNA. Adv Enzymol Relat Areas Mol Biol. 65:255–285. 1992.PubMed/NCBI

8 

Dubin DT and Taylor RH: The methylation state of poly A-containing messenger RNA from cultured hamster cells. Nucleic Acids Res. 2:1653–1668. 1975. View Article : Google Scholar : PubMed/NCBI

9 

Haugland RA and Cline MG: Post-transcriptional modifications of oat coleoptile ribonucleic acids. 5′-Terminal capping and methylation of internal nucleosides in poly(A)-rich RNA. Eur J Biochem. 104:271–277. 1980. View Article : Google Scholar : PubMed/NCBI

10 

Niu Y, Zhao X, Wu YS, Li MM, Wang XJ and Yang YG: N6-methyl-adenosine (m6A) in RNA: An old modification with a novel epigenetic function. Genomics Proteomics Bioinformatics. 11:8–17. 2013. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al: Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24:177–189. 2014. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Ear J and Lin S: RNA methylation regulates hematopoietic stem and progenitor cell development. J Genet Genomics. 44:473–474. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Wang P, Doxtader KA and Nam Y: Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell. 63:306–317. 2016. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Pendleton KE, Chen B, Liu K, Hunter OV, Xie Y, Tu BP and Conrad NK: The U6 snRNA m6A Methyltransferase METTL16 Regulates SAM Synthetase Intron Retention. Cell. 169:824–835.e814. 2017. View Article : Google Scholar

18 

Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang YG and He C: N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Bio. 7:885–887. 2011. View Article : Google Scholar

19 

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

20 

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

21 

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

22 

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

23 

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

24 

Theler D, Dominguez C, Blatter M, Boudet J and Allain FH: Solution structure of the YTH domain in complex with N6-methyladenosine RNA: A reader of methylated RNA. Nucleic Acids Res. 42:13911–13919. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Saletore Y, Meyer K, Korlach J, Vilfan ID, Jaffrey S and Mason CE: The birth of the Epitranscriptome: Deciphering the function of RNA modifications. Genome Biol. 13:1752012. View Article : Google Scholar : PubMed/NCBI

26 

Aschenbrenner J, Werner S, Marchand V, Adam M, Motorin Y, Helm M and Marx A: Engineering of a DNA polymerase for direct m6A sequencing. Angew Chem Int Ed Engl. 57:417–421. 2018. View Article : Google Scholar :

27 

Molinie B, Wang J, Lim KS, Hillebrand R, Lu ZX, Van Wittenberghe N, Howard BD, Daneshvar K, Mullen AC, Dedon P, et al: m6A level and isoform characterization sequencing (m6A-LAICseq) reveals the census and complexity of the m6A epitranscriptome. Nat Methods. 13:692–698. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Nagarajan A, Janostiak R and Wajapeyee N: Dot blot analysis for measuring global N6-methyladenosine modification of RNA. Methods Mol Biol. 1870:263–271. 2019. View Article : Google Scholar

29 

Arguello AE, DeLiberto AN and Kleiner RE: RNA chemical proteomics reveals the N6-methyladenosine (m6A)-regulated protein-RNA interactome. J Am Chem Soc. 139:17249–17252. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Yin H, Wang H, Jiang W, Zhou Y and Ai S: Electrochemical immunosensor for N6-methyladenosine detection in human cell lines based on biotin-streptavidin system and silver-SiO2 signal amplification. Biosens Bioelectron. 90:494–500. 2017. View Article : Google Scholar

31 

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

32 

Weng Li Z, Su H, Weng R, Zuo X, Li Z, Huang C, Nachtergaele H, Dong S, Hu LC, et al: FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell. 31:127–141. 2017. View Article : Google Scholar

33 

Wang Y, Li Y, Yue M, Wang J, Kumar S, Wechsler-Reya RJ, Zhang Z, Ogawa Y, Kellis M, Duester G and Zhao JC: N6-methyladenosine RNA modification regulates embryonic neural stem cell self-renewal through histone modifications. Nat Neurosci. 21:195–206. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Imanishi M, Tsuji S, Suda A and Futaki S: Detection of N6-methyladenosine based on the methyl-sensitivity of MazF RNA endonuclease. Chem Commun (Camb). 53:12930–12933. 2013. View Article : Google Scholar

35 

Mishima E, Jinno D, Akiyama Y, Itoh K, Nankumo S, Shima H, Kikuchi K, Takeuchi Y, Elkordy A, Suzuki T, et al: Immuno-Northern blotting: Detection of RNA modifications by using antibodies against modified nucleosides. PLoS One. 10:e01437562015. View Article : Google Scholar : PubMed/NCBI

36 

Mishima E and Abe T: Immuno-northern blotting: Detection of modified RNA using gel separation and antibodies to modified nucleosides. Methods Mol Biol. 1870:179–187. 2019. View Article : Google Scholar

37 

Chen W, Feng P, Ding H and Lin H: Identifying N6-methyladenosine sites in the Arabidopsis thaliana transcrip-tome. Mol Genet Genomics. 291:2225–2229. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Golovina AY, Dzama MM, Petriukov KS, Zatsepin TS, Sergiev PV, Bogdanov AA and Dontsova OA: Method for site-specific detection of m6A nucleoside presence in RNA based on high-resolution melting (HRM) analysis. Nucleic Acids Res. 42:e27. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Lopez CM, Lloyd AJ, Leonard K and Wilkinson MJ: Differential effect of three base modifications on DNA thermostability revealed by high resolution melting. Anal Chem. 84:7336–7342. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Liu N, Parisien M, Dai Q, Zheng G, He C and Pan T: Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA. 19:1848–1856. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Jacob R, Zander S and Gutschner T: The dark side of the epitranscriptome: Chemical modifications in long non-coding RNAs. Int J Mol Sci. 18:E23872017. View Article : Google Scholar : PubMed/NCBI

42 

Li X, Zhu P, Ma S, Song J, Bai J, Sun F and Yi C: Chemical pulldown reveals dynamic pseudouridylation of the mammalian transcriptome. Nat Chem Biol. 11:592–597. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Antanaviciute A, Baquero-Perez B, Watson CM, Harrison SM, Lascelles C, Crinnion L, Markham AF, Bonthron DT, Whitehouse A and Carr IM: M6aViewer: Software for the detection, analysis, and visualization of N6-methyladenosine peaks from m6A-seq/ME-RIP sequencing data. RNA. 23:1493–1501. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Cui X, Meng J, Zhang S, Chen Y and Huang Y: A novel algorithm for calling mRNA m6A peaks by modeling biological variances in MeRIP-seq data. Bioinformatics. 32:i378-i3852016. View Article : Google Scholar : PubMed/NCBI

45 

Meng J, Lu Z, Liu H, Zhang L, Zhang S, Chen Y, Rao MK and Huang Y: A protocol for RNA methylation differential analysis with MeRIP-Seq data and exomePeak R/Bioconductor package. Methods. 69:274–281. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Liu H, Wang H, Wei Z, Zhang S, Hua G, Zhang SW, Zhang L, Gao SJ, Meng J, Chen X and Huang Y: MeT-DB V2.0: Elucidating context-specific functions of N6-methyl-adenosine methyltran-scriptome. Nucleic Acids Res. 46:D281–D287. 2017. View Article : Google Scholar

47 

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

48 

Zhang M, Li Q and Xie Y: A Bayesian hierarchical model for analyzing methylated RNA immunoprecipitation sequencing data. Quant Biol. 6:275–286. 2018. View Article : Google Scholar

49 

Rana AP and Tuck MT: Analysis and in vitro localization of internal methylated adenine residues in dihydrofolate reductase mRNA. Nucleic Acids Res. 18:4803–4808. 1990. View Article : Google Scholar : PubMed/NCBI

50 

Ehrlich M, Gama-Sosa MA, Carreira LH, Ljungdahl LG, Kuo KC and Gehrke CW: DNA methylation in thermophilic bacteria: N4-methylcytosine, 5-methylcytosine, and N6-methyladenine. Nucleic Acids Res. 13:1399–1412. 1985. View Article : Google Scholar : PubMed/NCBI

51 

Clancy MJ, Shambaugh ME, Timpte CS and Bokar JA: Induction of sporulation in Saccharomyces cerevisiae leads to the formation of N6-methyladenosine in mRNA: A potential mechanism for the activity of the IME4 gene. Nucleic Acids Res. 30:4509–4518. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Zhao X, Yang Y, Sun BF, Shi Y, Yang X, Xiao W, Hao YJ, Ping XL, Chen YS, Wang WJ, et al: FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res. 24:1403–1419. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Barbieri I, Tzelepis K, Pandolfini L, Shi J, Millán-Zambrano G, Robson SC, Aspris D, Migliori V, Bannister AJ, Han N, et al: Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature. 552:126–131. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Tang Li X, Huang J, Wang W, Li F, Qin P, Qin C, Zou Z, Wei Q, Hua JL, et al: The M6A methyltransferase METTL3: Acting as a tumor suppressor in renal cell carcinoma. Oncotarget. 8:96103–96116. 2017.PubMed/NCBI

55 

Miao Z, Xin N, Wei B, Hua X, Zhang G, Leng C, Zhao C, Wu D, Li J, Ge W, et al: 5-hydroxymethylcytosine is detected in RNA from mouse brain tissues. Brain Res. 1642:546–552. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Rona G, Scheer I, Nagy K, Pálinkás HL, Tihanyi G, Borsos M, Békési A and Vértessy BG: Detection of uracil within DNA using a sensitive labeling method for in vitro and cellular applications. Nucleic Acids Res. 44:e282016. View Article : Google Scholar :

57 

Wehr NB and Levine RL: Quantitation of protein carbonylation by dot blot. Anal Biochem. 423:241–245. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Jaffrey SR and Kharas MG: Emerging links between m6A and misregulated mRNA methylation in cancer. Genome Med. 9:22017. View Article : Google Scholar :

59 

Kwok CT, Marshall AD, Rasko JE and Wong JJ: Genetic alterations of m6A regulators predict poorer survival in acute myeloid leukemia. J Hematol Oncol. 10:392017. View Article : Google Scholar

60 

Zhang C, Zhi WI, Lu H, Samanta D, Chen I, Gabrielson E and Semenza GL: Hypoxia-inducible factors regulate pluripotency factor expression by ZNF217- and ALKBH5-mediated modulation of RNA methylation in breast cancer cells. Oncotarget. 7:64527–64542. 2016.PubMed/NCBI

61 

Inouye M: The discovery of mRNA interferases: Implication in bacterial physiology and application to biotechnology. J Cell Physiol. 209:670–676. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Gerstberger S, Hafner M and Tuschl T: A census of human RNA-binding proteins. Nat Rev Genet. 15:829–845. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Licatalosi DD, Mele A, Fak JJ, Ule J, Kayikci M, Chi SW, Clark TA, Schweitzer AC, Blume JE, Wang X, et al: HITS-CLIP yields genome-wide insights into brain alternative RNA processing. Nature. 456:464–469. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Dubinsky L, Krom BP and Meijler MM: Diazirine based photoaffinity labeling. Bioorg Med Chem. 20:554–570. 2012. View Article : Google Scholar

65 

Kauer JC, Erickson-Viitanen S, Wolfe HR Jr and DeGrado WF: p-benzoyl-L-phenylalanine, a new photoreactive amino acid. Photolabeling of calmodulin with a synthetic calmodulin-binding peptide. J Biol Chem. 261:10695–10700. 1986.PubMed/NCBI

66 

Zhu T, Roundtree IA, Wang P, Wang X, Wang L, Sun C, Tian Y, Li J, He C and Xu Y: Crystal structure of the YTH domain of YTHDF2 reveals mechanism for recognition of N6-methyladenosine. Cell Res. 24:1493–1496. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Xu C, Wang X, Liu K, Roundtree IA, Tempel W, Li Y, Lu Z, He C and Min J: Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol. 10:927–929. 2014. View Article : Google Scholar : PubMed/NCBI

68 

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

69 

Luo GZ, MacQueen A, Zheng G, Duan H, Dore LC, Lu Z, Liu J, Chen K, Jia G, Bergelson J and He C: Unique features of the m6A methylome i. Arabidopsis thaliana Nat Commun. 5:56302014. View Article : Google Scholar

70 

Piekna-Przybylska D, Decatur WA and Fournier MJ: The 3D rRNA modification maps database: With interactive tools for ribosome analysis. Nucleic Acids Res. 36:D178–D183. 2008. View Article : Google Scholar :

71 

Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z and Zhao JC: N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol. 16:191–198. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, Pestova TV, Qian SB and Jaffrey SR: 5′ UTR m(6)a promotes cap-independent translation. Cell. 163:999–1010. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Lin S, Choe J, Du P, Triboulet R and Gregory RI: The m(6)a methyltransferase Mettl3 promotes translation in human cancer cells. Mol Cell. 62:335–345. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Fu Y, Dominissini D, Rechavi G and He C: Gene expression regulation mediated through reversible m6A RNA methylation. Nat Rev Genet. 15:293–306. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Cai X, Wang X, Cao C, Gao Y, Zhang S, Yang Z, Liu Y, Zhang X, Zhang W and Ye L: HBXIP-elevated methyltransferase METTL3 promotes the progression of breast cancer via inhibiting tumor suppressor let-7g. Cancer Lett. 415:11–19. 2018. View Article : Google Scholar

76 

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

77 

Li Y, Zheng D, Wang F, Xu Y, Yu H and Zhang H: Expression of demethylase genes, fto and alkbh1, is associated with prognosis of gastric cancer. Dig Dis Sci. 2019. View Article : Google Scholar

78 

Wang X, Li Z, Kong B, Song C, Cong J, Hou J and Wang S: Reduced m6A mRNA methylation is correlated with the progression of human cervical cancer. Oncotarget. 8:98918–98930. 2017.PubMed/NCBI

79 

Zhou J, Wang J, Hong B, Ma K, Xie H, Li L, Zhang K, Zhou B, Cai L and Gong K: Gene signatures and prognostic values of m6A regulators in clear cell renal cell carcinoma-a retrospective study using TCGA database. Aging (Albany NY). 11:1633–1647. 2019. View Article : Google Scholar

80 

Chen M, Wei L, Law CT, Tsang FH, Shen J, Cheng CL, Tsang LH, Ho DW, Chiu DK, Lee JM, et al: RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 67:2254–2270. 2018. View Article : Google Scholar

Related Articles

Journal Cover

June-2019
Volume 43 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu W, Wang JZ, Xu Z, Cao M, Hu Q, Pan C, Guo M, Wei JF and Yang H: Detection of N6‑methyladenosine modification residues (Review). Int J Mol Med 43: 2267-2278, 2019
APA
Zhu, W., Wang, J., Xu, Z., Cao, M., Hu, Q., Pan, C. ... Yang, H. (2019). Detection of N6‑methyladenosine modification residues (Review). International Journal of Molecular Medicine, 43, 2267-2278. https://doi.org/10.3892/ijmm.2019.4169
MLA
Zhu, W., Wang, J., Xu, Z., Cao, M., Hu, Q., Pan, C., Guo, M., Wei, J., Yang, H."Detection of N6‑methyladenosine modification residues (Review)". International Journal of Molecular Medicine 43.6 (2019): 2267-2278.
Chicago
Zhu, W., Wang, J., Xu, Z., Cao, M., Hu, Q., Pan, C., Guo, M., Wei, J., Yang, H."Detection of N6‑methyladenosine modification residues (Review)". International Journal of Molecular Medicine 43, no. 6 (2019): 2267-2278. https://doi.org/10.3892/ijmm.2019.4169