RNA Modifications
Georgeta N Basturea (gbasturea at gmail dot com)
Florida, USA (formerly of University of Miami Miller School of Medicine, USA)
DOI
//dx.doi.org/10.13070/mm.en.3.186
Date
last modified : 2022-10-14; original version : 2013-05-12
Cite as
MATER METHODS 2013;3:186
Abstract

A comprehensive review of research methods for RNA modifications / RNA epigenetics / epitranscriptomics. Also included is a comprehensive list of modified-RNA readers and enzymes involved in RNA modifications and antibodies against the readers and enzymes cited among the over 60,000 formal publications Labome has surveyed for Validated Antibody Database.

Introduction

One of the basic molecules of life, the ribonucleic acid (RNA) consists of four canonical nucleobases (adenine, cytosine, guanine and uracil) linked together with ribose molecules via phosphodiester bonds. While DNA is stable and stores the genetic information that dictates the order of the nucleobases, the RNA is less stable and performs multiple roles. The messenger RNA (mRNA) transports genetic information; the transfer RNA (tRNA) carries the building blocks for protein synthesis; the ribosomal RNA (rRNA) catalyzes the formation of peptide bonds, while the long noncoding (lncRNA) or small noncoding (sncRNA) RNAs have multiple regulatory roles [2]. The four canonical bases do not have the chemical versatility required for such functions. Posttranscriptional chemical modifications introduced at specific sites increase the structural and functional diversity of RNA in all organisms [2, 3].

So far, scientists identified 163 distinct chemical modifications that were classified as [4] : i) 5-ribosyluracil or pseudouridine (Ψ), ii) simple alteration of the bases; iii) methylation of the ribose 2’-hydroxyl group (Nm); and iv) complex or hypermodifications. Ψs are the result of uridine isomerization into pseudouridine such that a C-glycosidic linkage replaces the usual N-glycosidic bond and an additional imino group becomes available for hydrogen bonding. The second and third class include simple modifications such as methylation (e.g., m3U, m7G), deamination (e.g., inosine, which has been exploited to enable RNA editing [5] ), reduction (e.g., dihydrouridine), thiolation (e.g., s2U) or alkylation (e.g., Thr-tRNA) of the base or methylation of the sugar moiety. Sequential addition of simple modifications or larger chemical groups (e.g., aromatic rings, amino acid derivatives, sugars) leads to complex modifications. Their various structures, localization, specific nomenclature and pathways of chemical synthesis are included in several databases that are updated periodically: the RNA modification database - RNAMDB [3], MeT-DB - a database of transcriptome methylation in mammalian cells [6] ; MODOMICS - a database of RNA modification pathways [7], and a database dedicated to tRNA modifications – tRNAdb [8]. Several reviews on the role of modifications in RNA are available [2, 9-12]. Multiple others were published after the emergence of the field of RNA epigenetics, also called epitranscriptomics, not only in eukaryotes [13-19], but also in bacteria [20, 21] or viruses (presumably, for example, for evading detection by human innate immunity) [22].

Gene / UniProt Functions
YTH-family
YTHDC1
Human, Q96MU7
Mouse, E9Q5K9
Alternative splicing regulator [23-27]
mRNA splice site selection [28]
Nuclear export of m6A-containing mRNAs [27]
Transcriptional silencing of genes on the X chromosome [29]
Involved in S-adenosyl-L-methionine homeostasis by regulating expression of MAT2A transcripts [30]
YTHDC2
Human, Q9H6S0
Mouse, B2RR83
3'-5' RNA helicase that promotes the transition from mitotic to meiotic divisions in stem cells [24]
Germline cell cycle switching [31-35]
RNA processing and stability [35]
Regulates the level of some m6A-containing RNAs [34]
Required for spermatogenesis and oogenesis [32, 34]
Involved in degradation of m6A-containing mRNAs together with the XRN1 exoribonuclease [34]
YTHDF1
Human, Q9BYJ9
Positive regulator of mRNA translation [24, 36-38]
Facilitates translation initiation [39]
YTHDF2
Human, Q9Y5A9
Mouse, Q91YT7; Zebrafish, E7F1H9
Regulates mRNA stability and processing [24, 36, 39-42]
Promotes cap-independent mRNA translation following heat shock stress [43]
May inhibit replication of Kaposi's sarcoma-associated herpesvirus (KSHV) [44]
Enhancer or inhibitor of KSHV gene expression and virion production [45]
Promotes viral gene expression and replication of polyomavirus SV40 [46]
Regulates oocyte maturation [47]
Involved in hematopoietic stem cells specification [48]
Key role in maternal-to-zygotic transition [49]
YTHDF3
Human, Q7Z739
Promotes mRNA translation efficiency and stability [37, 38, 50]
Promotes translation of circular RNAs [50]
Other YTH-domain containing proteins
Pho92
Yeast, Q06390
Binds to the 3'-UTR region of PHO4 mRNA, decreasing its stability [51]
Mmi1
Yeast, O74958
Required for the degradation of meiosis-specific mRNAs [52]
YTHDF
Fruit fly, Q9VBZ5
Role in sex determination [53]
HNRNP proteins
HNRNPA2B1
Human, P22626
Rat, A7VJC2
Packaging of nascent pre-mRNA [54]
Transport of mRNAs to the cytoplasm in oligodendrocytes and neurons [55]
Involved in pri-miRNA processing [56]
Involved in miRNA sorting into exosomes [57]
Regulator of mRNA splicing [56]
Transport of HIV-1 genomic RNA out of the nucleus [58]
Protects telomeric DNA repeat against endonuclease digestion [59]
mRNA transport to the cytoplasm [60]
HNRNPC
Human, P07910
Nucleates the assembly of 40S hnRNP particles [61]
Modulates the stability and the translation of bound mRNA molecules [61-64]
May play a role in spliceosome assembly and  regulation of mRNA splicing [65]
Other modified-RNA binding proteins
eIF3 complex
Multiple subunits
Initiation transcription factor directly binds 5′ UTR m6A [66]
Table 1. A list of readers of modified RNAs.

RNA modifications perform their function through two main approaches: structural changes in the modified RNA that either blocked or induce protein–RNA interactions, and direct recognition by modified-RNA binding proteins to induce subsequent reactions. Such binding proteins have been named “readers” of RNA modifications and so far only m6A readers are known. These readers are listed in Table 1. Known human enzymes modifying RNA, such as METTL3 [67], and also the human modified-RNA readers are listed in Table 2, along with research antibody information from Labome's Validated Antibody Database.

GeneGene descriptionTop three suppliers
ALKBH1 alkB homolog 1, histone H2A dioxygenase Abcam ab195376 (1)
ALKBH5 alkB homolog 5, RNA demethylase Abcam ab195377 (2)
BCDIN3D BCDIN3 domain containing RNA methyltransferase Santa Cruz Biotechnology sc-390348 (2)
DCPS decapping enzyme, scavenger Santa Cruz Biotechnology sc-393226 (1)
EIF3A eukaryotic translation initiation factor 3 subunit A Cell Signaling Technology 3411 (4)
EIF3B eukaryotic translation initiation factor 3 subunit B Santa Cruz Biotechnology sc-137214 (1), Abcam ab133601 (1)
EIF3F eukaryotic translation initiation factor 3 subunit F Santa Cruz Biotechnology sc-390831 (1)
EIF3H eukaryotic translation initiation factor 3 subunit H Cell Signaling Technology 3413 (2)
EIF3I eukaryotic translation initiation factor 3 subunit I BioLegend 646701 (1)
EIF3J eukaryotic translation initiation factor 3 subunit J Santa Cruz Biotechnology sc-376651 (1)
EIF3M eukaryotic translation initiation factor 3 subunit M Dako M7157 (3)
FBL fibrillarin Cell Signaling Technology 2639 (24), Abcam ab4566 (19), Novus Biologicals NB300-269 (8)
FTO FTO, alpha-ketoglutarate dependent dioxygenase Abcam ab126605 (7), Santa Cruz Biotechnology sc-271713 (4)
HNRNPA2B1 heterogeneous nuclear ribonucleoprotein A2/B1 Abcam ab6102 (10), MilliporeSigma R4653 (5), Santa Cruz Biotechnology sc-53531 (4)
HNRNPC heterogeneous nuclear ribonucleoprotein C (C1/C2) Santa Cruz Biotechnology sc-32308 (10), Abcam ab10294 (6), Invitrogen MA1-24631 (1)
HSD17B10 hydroxysteroid 17-beta dehydrogenase 10 Abcam ab10260 (3), Developmental Studies Hybridoma Bank AFFN-HSD17B10-4D9 (1)
METTL1 methyltransferase like 1 Sino Biological 11525-MM05 (2)
METTL14 methyltransferase like 14 Abcam ab220030 (3)
METTL3 methyltransferase like 3 Abcam ab195352 (15)
NAT10 N-acetyltransferase 10 Abcam ab194297 (3), Santa Cruz Biotechnology sc-271770 (1)
NSUN2 NOP2/Sun RNA methyltransferase family member 2 Invitrogen 702036 (2)
TRDMT1 tRNA aspartic acid methyltransferase 1 Santa Cruz Biotechnology sc-365001 (4), Novus Biologicals NB200-587 (2)
YTHDC1 YTH domain containing 1 Abcam ab220159 (1)
YTHDC2 YTH domain containing 2 Abcam ab220160 (1)
YTHDF1 YTH N6-methyladenosine RNA binding protein 1 Abcam ab220162 (4)
YTHDF2 YTH N6-methyladenosine RNA binding protein 2 Abcam ab220163 (6)
YTHDF3 YTH N6-methyladenosine RNA binding protein 3 Abcam ab220161 (4), Santa Cruz Biotechnology sc-377119 (2)
Table 2. A list of human RNA-modifying enzymes and readers of modified RNAs, and top suppliers of antibodies against them cited in the over 60,000 publications Labome has surveyed for Validated Antibody Database. The most cited monoclonal antibody from each supplier is listed.

Here I describe several techniques used to detect RNA modifications in detail. Some of them are old and are used to detect chemical modifications at single sites, but others are new and sensitive enough to allow detection and quantitation of a larger spectrum of modifications in the RNA. Several techniques for analysis of entire transcriptomes, which could reveal their overall function in an organism, are emerging. Liu Y et al measured global m6A level in mouse peritoneal macrophages through dot blot on poly(A)+ mRNA preparations with the anti-m6A antibody from Synaptic Systems ( 202003) [69]. The levels of modification on specific mRNA can be simply assessing through immunoprecipitation with, for example, an anti-m6A [70] or anti-m7G [71] antibody, and measuring the levels of mRNA. Wang L et al immunoprecipitated m6A-containing mRNAs with an anti-m6A antibody from RAW264.7 cell lysates and measured the levels of Cgas, p204, and Sting mRNAs through qPCR [70]. Interestingly, anti-m6A antibodies also recognize m6dA present in DNA molecules, which have led to false identification of m6dA DNA modifications in mammalian cells [72].

Methods for Detection of RNA Modifications at Single Sites
Reverse-transcription-based methods

In this method, RNA templates are reverse-transcribed into cDNA by reverse-transcriptases (RTs) (Figure 1). Extended cDNA fragments are separated on denaturing polyacrylamide gels and their length is determined by autoradiography. Detailed protocols for RNA extraction from various organisms, oligonucleotide radiolabeling and RT reactions were previously described [73]. This method takes advantage of the inability of the reverse transcriptase to extend the DNA chain upon encountering chemical blocks in the RNA template to detect modified nucleotides [73, 74].

RNA Modifications figure 1
Figure 1. Detection of RNA modification by reverse transcriptase. A) Schematic diagram; m – modified RNA; B) Primer extension analysis for identification of m3U of 23S rRNA. rRNA from wild-type E. coli strain MG1655 (wt) synthetic control RNA; sequencing lanes (A, C, G, U) are indicated.

In contrast to other methods (HPLC, MS or direct sequencing, see below) the RT-based methods do not require large amounts of highly purified RNA. They can be directly applied to total RNA samples, the selection being done by specific annealing of radiolabeled DNA primers, a few bases 3’ of the analyzed site [73, 74]. However, the sequence of the analyzed RNA has to be known to allow selection of the appropriate oligonucleotide. The method is relatively inexpensive, as it does not require specialized equipment and training, and can be performed in any small laboratory.

One of the drawbacks of this method is that interpreting the results of reverse transcription from purified RNA can sometimes be difficult, as the enzyme is sensitive to RNA secondary structures and unwanted fragmentation of the RNA template by nucleases, which can lead to pausing of the transcriptase. This is why a control reaction with in vitro synthesized RNA can be very helpful [75]. The reverse transcriptase is also prone to stuttering that causes doubling of bands on a gel [75] and it cannot detect modifications that are too close to the 5’- or 3’- end of the RNA. Poly(A) tailing can be used to allow sequence scanning up to within a few nucleotides of the 3' terminus [76]. Some modifications can cause the enzyme to pause but not to stop unless special reaction conditions are used (e.g., 2’-O methylations can cause a stop only when the concentration of dNTPs is low) [77], or require special chemical treatment of the RNA template to cause the transcriptase to stop [73, 74, 76, 78].

Detection of RNA modifications by reverse transcription without prior chemical treatment of the RNA

Chemical groups on the Watson-Crick face of nucleobases (atoms 1, 2, and 6 for purines and 2, 3, and 4 for pyrimidines) block the base-pairing interaction with complementary bases [73]. This results in attenuation of primed reverse transcription at such sites, resulting in a stop (e.g., m1A [79], m1G [80], m2A [81], m2G [82], m3U [83] ) or pause (e.g., m6A, dihydrouridine, m4C, Nm [73] ) on sequencing gels, one residue 3' to the modified nucleobase. In contrast, small modifications at the Hoogsteen side (e.g., m5U, m5C, Ψ, inosine, m7G [73] ) allow the reverse transcriptase to read through making this method unfeasible for detection of such modifications. When modifications are bulky, performing the RT reaction at low dNTPs concentration might allow detection of a stop at some of these sites.

Detection of RNA modifications by reverse transcription following chemical treatment of the RNA

Several different modifications that are elusive to detection by reverse-transcription can be detected after specific chemical treatment of the RNA. So far, detection methods have been developed for some of the most common modifications encountered in cellular RNA: inosine (I), pseudouridine (Ψ), m3C, m5C, m7G, 2’-O-methylated nucleotides (Nm), dihydrouridine (D). Comprehensive reviews of such modifications and the chemical treatments necessary for their detection [74, 78], as well as detailed protocols for the detection procedures have been published recently [73, 76].

These methods can also be adapted for high-throughput analysis of the transcriptome [84]. For example, Carlile et al used the CMC(N-cyclohexyl-N′-β-(4-methylmorpholinium)-ethylcarbodiimide) treatment followed by a reverse-transcription method for Ψ-sequencing [76] together with cDNA ligation, PCR and Illumina DNA sequencing to detect Ψ in total yeast and human mRNA [84].

Detection of RNA modifications by reverse transcription and PCR

Detection of 2’-O-methylated sites in RNA proved to be difficult especially in low abundance and small RNAs that are 2’-O-methylated at their 3’-termini. A new version of the RT-based detection method, specific for identification of 2’-O-methylated residues, was recently developed [85]. The new method, referred to as RTL-P or Reverse Transcription at Low deoxyribonucleoside triphosphate (dNTP) concentrations followed by Polymerase chain reaction (PCR) was described by Dong et al [85]. Briefly, purified RNA is treated with DNAse I to remove any DNA contaminant prior to RTL-P analysis, and then transcribed into cDNA by RT- reverse transcription at both low and high dNTP concentrations. At low dNTPs, the reverse transcriptase can stall one base prior to any 2’-O-methylated ribose, but it can readthrough the same methylated sites at high dNTPs. Thus, two different cDNA products are produced in the low dNTP reaction, while only one full-length product is generated by reverse transcription at high dNTPs. The cDNA products are amplified by PCR with a reverse primer specific to the 3’-end of the molecule, and forward primers specific to a region upstream (to detect a full-length product) and downstream (to detect shorter cDNA) of the predicted methylated site. The PCR products generated by the two sets of primers migrate as a double band on agarose gels. If a methylated residue is present at the predicted site, the intensity of the two bands is the same when the product from the high dNPTs reaction is amplified, and is less intense and longer when the cDNA from the low dNTPs reaction is used. There is no difference between the low and high dNTPs reactions bands when the 2’-O-methylation is missing. The same method can be used for detection of 3’-end 2’-O-methylated residues of small RNAs after ligation of 3’-end adaptors using T4 ligase [85]. Direct and site-specific quantification of RNA 2′-O-methylation by PCR can also be achieved through an engineered DNA polymerase which is able to discriminate 2′-O-methylated from unmethylated RNAs [86]. J Shi et al developed PANDORA-seq (panoramic RNA display by overcoming RNA modification aborted sequencing) to facilitate the detection of small non-coding RNA (sncRNA) with RNA modifications [87].

Chromatography-based methods
Thin-layer chromatography (TLC)

Identification of modified nucleotides in RNA by thin layer chromatography (TLC) is another relatively simple and inexpensive approach that can be used in any small research laboratory. Detailed reviews of the technique have been published in recent years [88, 89]. The method is very effective when applied to short RNAs (~100-150 nt) like transfer RNA or other small non-coding RNAs. Larger molecules can be analyzed after ribonuclease digestion and RNA fragments purification.

Complete digestion of RNA by various ribonucleases is required prior to analysis of the RNA digest products by 2-dimensional separation on TLC plates. The products differ depending on the nuclease used. RNase T2 digestion produces 3’-monophosphate nucleosides, both nuclease P1 and the venom phosphodiesterase (VPD) generate 5’-monophosphate nucleosides, and piperidine releases both 2’- and 3- monophosphate nucleosides. Incomplete digestions can also produce dinucleotides diphosphates. Grosjean et al developed two solvent systems for nucleosides separation and constructed reference maps for 5’- monophosphate nucleosides derived from all four nucleobases, as well as for dinucleotide diphosphates [89]. These maps indicate the relative localization of more than 70 modified nucleotides in the 2D TLC systems developed [89].

Modified nucleotides in RNA can be quantitated when 32P pre-labeled radioactive RNA [89] is used for analysis. The amount of radioactivity in each spot on the TLC plate can be determined by exposure to a PhosphorImager screen and data analysis by ImageQuant and Excel, or by direct measurement of the radioactivity in each spot after transferring the scraped plate material to a vial containing scintillation fluid followed by count measurement in a scintillation counter. Similarly, ribonuclease-digested RNA products can be radiolabeled with [90] by treatment with polynucleotide kinase prior to separation on TLC plates. Protocols for both [90] labeling approaches are available [89].

This method, either on its own or in combination with previously described methods, constitutes a powerful tool for detecting and quantifying modified nucleotides in naturally occurring RNAs [91, 92], as well as in synthetic RNA substrates treated with cell extracts or purified recombinant RNA modification enzymes [93, 94].

RNA Modifications figure 2
Figure 2. Strategy for analysis of modified nucleotides in RNA by thin-layer chromatography.
Liquid chromatography

Several liquid chromatography (LC) methods for detection and quantitation of RNA modifications have been developed over the years [95-97]. Typically they involve purification of the RNA molecule of interest (either cold or radioactively labeled), partial or complete digestion with nucleases and phosphatase treatment prior to separation on a chromatographic column. Mono and/or oligonucleotides (or nucleosides obtained after removal of phosphate) are detected by UV absorption or measurement of radioactivity. The identification of modified nucleotides is based on differences in their chromatographic mobility when compared with their unmodified counterparts.

Nowadays reversed-phase high-performance liquid chromatography in conjunction with UV detection (RPLC-UV) is most often used. Gehrke and Kuo, (1989) provided a comprehensive review of such methods used for the detection and quantitation of nucleosides, either modified or unmodified, in RNAs. They include detailed chromatographic protocols, describe standard columns and the essential requirements for the HPLC instrumentation, and list chromatographic retention times and UV spectra for many ribonucleosides [98]. Mazauric et al successfully used such a method for analysis of complex RNA modifications [99].

Mass-spectrometry-based methods

Mass spectrometry is a very sensitive tool for detection of the mass of biological molecules. The method has been extensively used for analysis of post-translational modifications in proteins for many years [100], and it was successfully adapted to the study of modified nucleic acids [71, 101]. The principle behind these methods is simple. It involves generation of gas phase ions of the molecules of interest, either nucleosides or oligonucleotides, and separation of the generated ions according to their mass-to-charge ratios. However, the technical details of the experimental protocols can be difficult and require specialized training and equipment. Two techniques are most commonly used to generate gas phase ions: MALDI (Matrix Assisted Laser Desorption/Ionization) and ESI (Electrospray Ionization).

Matrix-assisted laser desorption/ionization (MALDI)

The following experimental strategy is most commonly used for detection of modified nucleotides in RNA:

  1. RNA extraction. Detailed protocols for RNA extraction from different source organisms [73] as well as special considerations for RNA sample preparation for MS analysis [102] are available.
  2. Digestion of RNA to short oligonucleotides or nucleosides. Specific ribonucleases can be used to generate oligonucleotides of lengths suitable for mass spectrometry (about 3 to 20 nucleotides): e.g., RNase T1 [103] a guanosine-specific ribonuclease, RNase A [104] a pyrimidine-specific ribonuclease or Nuclease P1 a ribonuclease that degrades RNA molecules to nucleotides. Isolation of particular RNA fragments can also be achieved prior to nuclease degradation by site-directed hybridization followed by digestion with Mung Bean Nuclease and RNase A [103] or RNase H [105]. Full-length short RNA molecules can be used directly if they are only ~ 22 nucleotides long (miRNA, siRNA etc) [106, 107].
  3. Ionization of oligonucleotides or nucleosides and time-of-flight analysis to determine mass/charge ratios.
  4. Comparison of the experimentally determined mass/charge values with the theoretical ones based on known oligonucleotide sequence.
  5. Identification of modified oligonucleotides/nucleoside based on mass/charge deviations from theoretical values.

Gas phase ions generated by MALDI are singly charged ions, which are either protonated or deprotonated depending on the ion detection mode used (positive or negative). Thus the obtained mass/charge ratios correspond to the oligonucleotide mass plus or minus one hydrogen mass (1.01 Da). Due to the simplicity of the obtained spectra, MALDI can be used to determine the masses of specific molecular species within complex mixtures. However, the precise nature of the modifications cannot be obtained. Tandem MALDI-MS can be used to determine the mass and the nucleotide location of a particular modification, and whether the modification is located on the base or on the ribose, but cannot identify the exact atom at which the modification is found.

The technique can be applied to RNA fragments of about 20 nucleotides, but not mono or dinucleotides for which the mass/charge ratio signal is masked by intense noise generated from matrix and buffer components [102]. Longer RNA fragments cannot be analyzed accurately. The detection sensitivity decreases with increasing mass because of both detector design and the tendency of large RNAs molecules to fragment before detection. Also, the presence of both 12C and 13C isotopes in larger RNAs can interfere with spectra interpretation. For example, the mass difference between 13C and 12C can be interpreted as the mass difference between uridine and cytidine, about 0.98 Da [102].

Tandem MS (MS/MS)

Information on the exact modification site can be obtained by using tandem MS (MS/MS) to sequence the selected oligonucleotide. Tandem analysis is performed in three steps: i) Selection of the oligonucleotide of interest by setting a mass window such that all oligonucleotides of different mass are excluded from the mix; ii) fragmentation of the selected oligonucleotide by collision with an inert gas (e.g., argon); iii) mass analysis of resultant fragments followed by determination of the RNA oligonucleotide sequence and modification sites [103, 107]. Pandolfini L et al used negative ion tandem LC-MS in a hybrid quadrupole – orbitrap to identify specific m7G modification at position 11 (G11) of miRNA let-7e-5p O [71].

In spite of being such powerful tools, mass spectrometry techniques cannot easily distinguish all modifications (e.g. pseudouridines). Approaches designed for detection of pseudouridines by mass spectrometry involve selective chemical treatment [108], and were reviewed elsewhere [109]. However, due to their general chemistry (N-glycosidic bond in Us vs. C-glycosidic-bond in Ψs), uridines and Ψs have different fragmentation patterns upon collision with an inert gas. Due to these differences, a few MS-based approaches suitable for detection of Ψs without Ψ –targeted chemical treatment were also developed [110, 111].

RNA Modifications figure 3
Figure 3. Detection of modified nucleotides by mass spectrometry. Strategy for qualitative and quantitative analysis.
Electrospray ionisation mass spectroscopy (ESI-MS)

The experimental strategy commonly used in ESI-MS experiments is essentially the same as the one used in MALDI (see above). The same steps are generally performed regardless of the ionization technique. However, a few differences between the two ionization techniques should be noted. Contrary to MALDI, ESI generates multiply charged ions that can increase the complexity of the obtained spectra especially for samples containing multiple molecular species. If the components of a complex mixture are to be identified, using ESI-MS alone is not practical as spectra interpretation can be challenging. A chromatographic separation technique is often used in conjunction with ESI-MS for such tasks (see below) [79, 112, 113]. On the positive side, ESI can be used to analyze RNAs as long as 16S rRNA (~1500 nt), and to identify the exact position of a modification within nucleotides [114].

Liquid chromatography-mass spectrometry (LC-MS)

For analysis of single nucleotides liquid chromatography coupled with mass spectrometry (LC-MS) is the method of choice nowadays. This method relies on nuclease P1 and alkaline phosphatase digestion of RNA to nucleosides, and separation of the digestion products by reverse phase chromatography, analysis by MS and UV light (254 nm) absorption [87, 115]. Any modified nucleoside is identified on the bases of shifts in retention time from the retention times of standard nucleosides separated by the same chromatographic procedure. The UV detection indicates the stoichiometry between the modified and unmodified nucleotides, making LC-MS a great tool for quantitative analysis. Such experiments can lead to the discovery of new modifications, but localization of such modifications in the RNA species requires further studies. The most common approach relies on ribonuclease (RNase T1 or RNaseA) fragmentation of the RNA into oligonucleotides, chromatographic separation and mass spectrometry [71, 116] as described above. Most recently a 2-dimensional (2D) LC-MS method, involving the purification of nucleotides on two different liquid-chromatography columns, followed by MS-identification was developed for the analysis of modified nucleosides in biological samples [117].

Other (less common) methods
2’-O-methyl RNA-DNA chimera-directed RNase H cleavage

The RNase H-based method is a three-step strategy for detecting and quantifying modified bases in RNA. It is most useful for the study of RNA modification enzymes than for identification of novel modifications. So far it has been successfully used for detecting 2’-O-methylated nucleotides [118], base-methylated nucleotides and pseudouridines [119].

To detect the 2’-O –methylated ribose, 2’-O-methyl RNA–DNA chimeras are used to direct RNase H cleavage in target RNA molecules [120]. RNase H selectively cleaves RNA in the formed duplex at sites were the ribose is unmodified, while any methylated ribose completely inhibits the enzyme. End-labeling and gel separation of the resulting fragments together with uncleaved, control RNA sample, reveal the methylated ribose. The level of 2’-O-methylation can be measured on the base of the intensity of the radioactive signal on the gel [118].

To detect base modifications, Zhao and Yu made several modifications to the RNase H approach (Figure 5). First, the RNA is cleaved at the 5’- side of the modified nucleotide of interest. The released products are a 5’-fragment with a 3’-hydroxyl end, and a 3’-fragment containing the phosphorylated modified nucleotide at its 5’- end. The 3’-fragment is gel purified, dephosphorylated and rephosphorylated with 32P in a reaction catalyzed by polynucleotide kinase (PNK). Finally, the fragment is digested with nuclease P1, and the released radiolabeled nucleotide at the 5’-end of the 3’-fragment is isolated and quantitated by thin layer chromatography as described above.

RNA Modifications figure 4
Figure 4. Detection of RNA modifications by the RNase H cleavage method. Schematic diagram. See text for referenced examples from the literature.
RNA-cleaving deoxyribozyme (DNAzyme) cleavage

Similarly to the RNase H cleavage method, one can detect and quantitate RNA modifications by using a DNAzyme. The in vitro selected DNA-based enzyme consists of a small single-stranded DNA molecule with a catalytic domain of about 15 deoxynucleotides, flanked by two substrate-recognition domains of seven to eight deoxynucleotides each [121]. The DNA enzyme anneals to the RNA substrate through Watson–Crick base pairing and cleaves the target RNA at or near the site of interest, between an unpaired purine and a paired pyrimidine residue [121]. Based on their ability to digest only the unmodified substrates, DNAzymes allow the identification of ribose methylations and pseudouridines [122, 123]. Hengesbach et al describe two different approaches for the use of this method. In the first approach the substrate RNA is cleaved 5’ of the residue of interest and the digestion product is radioactively labeled prior to digestion to nucleosides and analysis by thin-layer chromatography. A more detailed description of this approach was published recently [124]. The second approach involves two DNAzyme-directed cleavages to excise RNA fragments comprising the modified nucleotides of interest, followed by RNA fragment isolation and analysis. Quantitation of modified RNA is performed by temperature cycling, iterative DNAzyme - RNA substrate complex formation and RNA cleavage [122, 124].

RNA Modifications figure 5
Figure 5. Detection of RNA modifications by the DNAzyme-cleavage method. Schematic diagrams for two different strategies. See text for referenced examples from the literature.
Ligation-based methods

This method is based on molecular recognition and enzymatic ligation of DNA oligonucleotides annealed to both modified and unmodified RNA, in regions flanking specific modification sites. So far it has been successfully used to detect 2’-O-methylated nucleotides [125], m6As and pseudouridines [126].

Two oligonucleotide pairs are empirically identified. Upon ligation with T4 DNA ligase one pair can discriminate the unmodified RNA template from the modified one. The discrimination is based on the ability of the residue opposite the modification site (the recognition residue) to influence the ligation efficiency. The second pair does not distinguish between the two templates, and it is used to determine the total amount of template RNA available. Once the two sets of oligonucleotides are found, template-dependent ligation reactions are performed for each set using a mixture of modified and unmodified RNA as template.

Saika et al found several such pairs containing “recognition residues” specific to different modifications [125]. Dai et al used oligonucleotides in which they incorporated specific nucleoside analogs to introduce recognition residues specific for m6A modifications [126]. By using multiple discriminating oligonucleotide pairs, different modification sites can be analyzed simultaneously. Moreover, the relative abundance of the analyzed modifications in the RNA template can be determined on the base of the amount of ligation product detected on acrylamide gels [125, 126].

RNA Modifications figure 6
Figure 6. Schematic diagram for the detection of RNA modifications by the ligation-based method.
Immuno-northern blotting (INB)

The INB method for detection of modified nucleotides in RNA was developed by combining two commonly used molecular biology techniques: Western and Northern blotting. With this method antibodies against the modified nucleotides are used for detection instead of the radiolabeled DNA probes used in the classical northern blotting protocols. Mishima et al [1] demonstrated that antibodies against 1-methyladenosine (m1A), N6-methyladenosine (m6A), pseudouridine, and 5-methylcytidine (m5C) could be successfully used in various research projects. The method is relatively simple, uses commonly available laboratory equipment and has high sensitivity and specificity. It could be a great tool for studying RNA metabolism in small laboratories.

RNA Modifications figure 7
Figure 7. Schematic diagram for the detection of RNA modification by INB [1].
Demethylase-facilitated RNA-Seq

Two methods that involve removing modified nucleotides that block reverse transcriptase have been recently developed and made possible the sequencing of RNA molecules that are highly modified or folded, like tRNAs. The methods developed by Cozen et al [127] (ARM-seq) and Zheng et al [128] (DM-tRNA-Seq) use pre-treatment of RNA with Escherichia coli AlkB dealkylating enzyme to demethylate 1- methyladenosine (m1A), 3-methylcytidine (m3C), and 1-methylguanosine (m1G). The demethylated RNA together with an untreated sample is subjected to steps similar to the ones used in RNA-Seq. Adaptor ligation followed by reverse-transcription, amplification and deep sequencing were used by Cozen et al To eliminate the inefficient adaptor ligation step, Zheng et al used the thermostable group II intron reverse transcriptase (TGIRT), an enzyme that can switch the templates and allows for amplification without added adaptors.

RNA Modifications figure 8
Figure 8. Schematic diagram for the detection of RNA modification by demethylase-facilitated RNA-Seq.
MC1 ribonuclease cleavage

Addepalli et al [129] used an engineered nuclease (MC1) to cleave RNA at the 5'-termini of uridine and pseudouridine, but not next to chemically modified uridine or uridine preceded by a nucleoside with a bulky modification. They identified the modified nucleotides after separating and analyzing the fragmented products by ion-pairing reverse phase liquid-chromatography coupled with mass spectrometry (IP-RP-LC-MS).

RNA Modifications figure 9
Figure 9. Nanopore sequencing technique diagram.
RNA Sequencing: Detection of RNA modifications at Multiple Sites
Direct RNA sequencing through Nanopore

Oxford Nanopore's direct RNA sequencing technology is based on a nanopore DNA sequencing technique developed recently [130, 131]. It is a direct RNA sequencing (direct RNA-seq) technology that allows the accurate quantification and characterization transcripts without fragmentation or amplification. In addition, the methodology permits the direct detection of N6-methyladenosine (m6A) RNA modifications in endogenous molecules with high accuracy. In this method, a single strand of nucleic acids passes through a pore protein embedded in an electrically resistant polymer membrane that is subjected to an ionic current. Nanopore detects changes in the electric current as the molecules pass through it (Figure #). By interpreting the recorded electrical signals, each nucleotide in the sequence can be identified. Because the current is sensitive to base modifications any chemical modification can be detected with accuracy. As each nucleotide/modification induces a specific electrical signal, the entire nucleotide sequence can be read together with any present modifications.

RNA Modifications figure 10
Figure 10. Amplification-based sequencing of RNA and detection of RNA modification. Schematic diagram.

Lorenz et al [132] developed and algorithm to annotate m6A in previously unknown sites containing conserved DRACH (D=G/A/U, R=G/A, H=A/U/C) motifs [133]. Choosing only DRACH sites helped improve the likelihood that the predictions are specific to m6A sites and not to other mRNA modifications. The software, called MINES (m6A Identification using Nanopore Sequencing) was trained with m6A-modified and unmodified synthetic sequences and, when tested on endogenous transcript could indicate m6A RNA modifications with high accuracy. By using data from data generated from an Oxford Nanopore sequencer as input, MINES predicted m6A modified sites in 13,000 previously unknown DRACH sites in endogenous transcripts from HEK293T cells and over 40, 000 in a human mammary epithelial cell line. In addition, by using cDNA alignments, MINES was able to predict m6A methylation in an isoform-specific manner.

Amplification-based sequencing

Sequencing RNA is a straightforward procedure in modern-day molecular biology. Purified RNA is reverse-transcribed into cDNA prior to polymerase chain reaction (PCR) amplification and DNA sequencing. However, modifications of the RNA molecules are not easily “readable” by this method. One exception is the detection of deaminated adenosines (inosines) in mRNAs. Because inosines are decoded like guanosines, the cDNA of edited mRNA differs from the genomic DNA at A-to-I editing sites. Thus, by a simple comparison of the genomic DNA and cDNA sequences these sites can be identified [134]. More recently Ramaswami et al, developed a bioinformatics-based method to identify RNA editing sites from RNA sequencing data obtained from multiple samples, without the need for matched genomic DNA sequence [135].

RNA Modifications figure 11
Figure 11. Schematic diagram for the detection of m5C modifications by RNA bisulfite sequencing.
Bisulfite sequencing

The amplification-based RNA sequencing method was adapted to the detection of methylated cytosines (m5C) by treating the RNA sample with bisulfite such that cytosines established are efficiently deaminated into uridine [136-138]. Bisulfite treatment deaminates nonmethylated cytosines, while methylated cytosines remain unaffected. cDNA synthesis followed by PCR amplification and DNA sequencing allows detection and quantitation of the methylated cytosines in RNA samples.

RNA Modifications figure 12
Figure 12. Main steps in the detection of m5C residues by Aza-IP.
Aza-immunoprecipitation (Aza-IP) method

Khoddami and Cairns developed a novel method to identify cytosines targeted by m5C methyltransferases [139]. This technique allows identification of m5C residues even in low abundance RNA species, it is applied to live cells and does not require knocking out any methyltransferase gene. Briefly, an m5C RNA methyltransferase of interested is overexpressed in living cells that are subsequently grown in the presence of 5-azacytidine (5-aza-C), a suicide inhibitor that incorporates in nascent RNAs and traps the enzyme by forming a stable RNA methyltransferase-RNA adduct. Methyltransferase specific antibodies bound to magnetic beads are then used to immunoprecipitate the formed adducts from cell lysates. Stringent washing is performed to eliminate RNA contaminants, and the remaining molecules are fragmented, released and purified. The isolated RNA fragments are ligated to adaptor oligonucleotides and reverse transcribed into cDNA that is further analyzed by DNA sequencing.

RNA Modifications figure 13
Figure 13. Schematic diagram for the detection of m6A modifications in RNA.
m6A-seq (N(6)-methyladenosine-sequencing)

N(6)-methyladenosine-sequencing (m6A-seq), also known as MeRIP-seq, is used to localize m6A in the transcriptome at high resolution. The technique is based on RNA fragmentation and immunoprecipitation with m6A specific antibodies, followed by stringent washing and release of remaining molecules. The isolated RNA fragments are ligated to adaptor oligonucleotides and reverse transcribed into cDNA that is further analyzed by DNA sequencing. A consensus motif is found and localized in the sequence [140]. Several examples are present in the literature. Studies by Dominissini et al [40] and by Meyer et al [141] used this method to assess the m6A methylation patterns in mouse and human cells under different conditions. An improved version of the method was successfully used by Schwartz et al [142] to dissect the m6A methylation during meiosis in yeast. Even though technically they the same method their system allows for better controls, they use less starting material. They reduced the number of false positive or negative results and localized the m6As at nearly single nucleotide resolution.

miCLIP (m6A individual-nucleotide-resolution cross-linking and immunoprecipitation) and PA-m6A-seq (photo-crosslinking assisted m6A sequencing) are two other variations of the m6A-seq method that were recently used to map m6A at single-nucleotide resolution. Both miCLIP, developed by Linder et al [143], used by, for example, by Huang H et al [144], and PA-m6A-seq used by Chen K et al [145], involve the crosslinking of the m6A-specific antibody to the RNA prior to the affinity purification of the RNA-antibody complexes, RNA isolation, reverse-transcription and DNA sequencing.

RNA Modifications figure 14
Figure 14. Comparative schematic diagrams for Pseudo-seq and CeU-seq.
Pseudo-seq (pseudouridine sequencing)

Pseudo-seq methods that provide a comprehensive analysis of Ψ in RNA with single-nucleotide resolution have been developed during the last 2-3 years. They adapted the use of reverse-transcription after chemical modification of the RNA to high-throughput analysis of the transcriptome. For example, Carlile et al [146, 147] and Lovejoy et al [148] used the CMC(N-cyclohexyl-N′-β-(4-methylmorpholinium)-ethylcarbodiimide) treatment followed by a reverse-transcription method for Ψ-sequencing together with cDNA ligation, PCR and Illumina DNA sequencing to detect Ψ in total yeast and human mRNA (Figure 13). To map pseudouridines quantitatively, Schwartz et at [149] added a computational method through which one can determine the relative stoichiometry (Ψ-ratio) of Ψ at each identified site. Another variation of this technique, called CeU-seq [150, 151], allows for a better selection of the Ψ-containing fragments through biotin pull-down facilitated by a chemically synthesized CMC derivative.

Borohydride reduction sequencing (BoRed-seq)

A chemical reactivity assay to detect internal m7G in low-abundant eukaryotic RNAs, like miRNAs, was developed by Pandolfini et al [71]. The assay was based on an older method that has been used for mapping m7G residues in highly abundant RNAs and involves nucleoside hydrolysis by treatment with NaBH4 [152] and alkali, followed by aniline-induced cleavage of the RNA chain at abasic sites by β-elimination and RNA sequencing of the generated fragments. Because the fragments obtained from small RNAs cannot be easily sequenced, Pandolfini and co-authors adapted the protocol for miRNAs. In this method, decapped total RNA was treated with NaBH4 and alkali to obtain abasic sites at positions harboring m7G and exposed to a biotin-coupled aldehyde reactive probe (N-(aminooxyacetyl)-n0-(D-biotinoyl) hydrazine; ARP) able to covalently link to abasic RNA sites [153]. The linked RNA fragments were pulled down on streptavidin beads and identified from RNA libraries by high-throughput sequencing (Figure 14). Several miRNAs containing m7G were identified by this method [71]. The presence of the methylated guanidine was validated by RNA immunoprecipitation with an m7G specific antibody and qPCR analysis or RNA sequencing. A comprehensive review of RNA sequencing methods can be found here. Mass spectroscopy was used to precisely map the m7G within the small miRNA.

RNA Modifications figure 15
Figure 15. Diagram of BoRed-seq method for detecting m7Gs in low-abundance RNA.
RBS-sequencing

A major current limitation in detecting RNA modifications is the lack of methods that permit the mapping of multiple modification types simultaneously, transcriptome-wide and at single-base resolution. Khoddami et al [154] developed RBS-seq, a variant of the RNA bisulfite sequencing method to detect simultaneously m5C, Ψ, and m1A at single-base resolution transcriptome-wide. Figure 15 indicates the main steps of RBS-sequencing/mismatch-based detection of all three modifications. For m5C, bisulfite treatment deaminates Cs converting to Us (Ts upon cDNA sequencing), while methylated cytosines are do not respond to the bisulfite treatment and remain Cs. For m1A, because m1A disturbs the canonical A:T base pairing, it pauses the reverse transcriptase leading to nucleotide misincorporation. Such mismatches act as m1A signatures in the synthesized cDNA. In contrast, under the alkaline conditions of the bisulfite treatment m1A converts to m6A (methyl passes from N1 to N6) [155], which reads as A and does not induce nucleotide misincorporation (see m6A-seq method above). Comparison of bisulfite treated and untreated samples leads to identification of m1A sites. For Ψ, Ψ nucleotides upon bisulfite treatment form a stable monobisulfite adduct causing the reverse transcriptase to base skip at the pseudouridine sites.

RNA Modifications figure 16
Figure 16. Simultaneous detection of m5Cs, m1As and Ψ by RBS-sequencing
Quantitative analysis of modified RNA

Identifying precise modified sites in RNA transcripts is an essential prerequisite for understanding the biological functions of the modifications. Current methods allow detection of chemically modified nucleotides in most RNAs, both at specific sites and simultaneously at multiple sites. However, the modification levels can change depending upon environmental conditions or during cell differentiation. Not much information exists regarding how many RNA copies are modified. Determining such changes can be the key to understanding important biological processes. Thus, quantitative analysis methods are necessary.

RNA Modifications figure 17
Figure 17. Combined method for detecting and quantifying base modifications.

The last decade provided extensive evidence for the importance of the m6A modifications in RNAs and their involvement in regulation of gene expression. Therefore, several quantitative methods were developed to quantify the m6A methylation levels of specific sites. Some are based on primer extension followed by polyacrylamide gel electrophoresis (Figure 1). Harcourt et al (2013) used a newly identified reverse transcriptase, Tth, that selectively incorporates thymine opposite unmodified A as compared to m6A. By performing two parallel reactions, one with Tth and one with a nonselective polymerase the authors provided a method to quantitate the amount of modified adenine at specific sites [156]. A similar approach was taken by Wang et al (2016) who provided a quantitative analysis of m6A in RNA and DNA by using BstI DNA polymerase/reverse transcriptase catalyzed primer extension [157]. The same enzyme was used in combination with PCR to quantify the m6A levels in specific residues of human SOCS1 and SOCS3 mRNAs [158], providing a simple and cost-effective method to relatively quantify the m6A levels in RNA transcripts. Most recently, Liu et al. (2020) used an IP-based commercial N6-Methyladenosine Enrichment Kit (EpiMark) to a performed m6A enrichment prior to quantification by RT-qPCR. This procedure allowed them to quantify the changes in m6A methylation level at specific target m6A sites [67].

An approach that combines primer-extension with RNase H cleavage and thin layer chromatography was used by Karijolich et al. (2010). They used RNase H to cleave only at sites where the 2′-OH of RNA is not modified to direct cleavage to a specific nucleotide of interest through the use of 2′-O-methyl RNA–DNA chimeric oligonucleotides. The cleaved RNA was radiolabeled and digested to single nucleotides that were then separated by thin layer chromatography prior to quantitation by measurement of the radioactive signal [159].

RNA Modifications figure 18
Figure 18. Diagram of the differential melting temperature technique for m6A quantitation. BNA: bridge nucleic acid.

Oshima et al. (2018) used artificial nucleic acid analogs, such as bridged nucleic acid (BNA), inserted into DNA probes to increase the difference in melting temperature between m6A-RNA and unmethylated RNA during hybridization experiments (Figure 17). They applied this differential melting temperature method to determine the m6A level in rRNA in E. coli [160].

Huang et al (2019) used liquid chromatography–tandem mass spectrometry (LC-MS/MS) quantify the m6A abundance in poly(A) RNA to determine the role of histone H3 lysine 36 trimethylation in m6A RNA modification [144]. Most recently, the same technique was used by Liu et al. (2020) to quantify the m6A/A ratio in nonribosomal chromosome associated RNAs (caRNAs) in wild type and methylase (Mettl3) or reader (Ythdc1) depleted mouse embryonic stem cells (mESCs). These small RNAs, whose m6A methylation state serves as a switch to regulate their levels, control the nearby chromatin state and downstream transcription [67]. The same study made use of m6A-seq, also known as MeRIP-seq (methylated RNA immunoprecipitation sequencing) method to determine the levels of m6A in immunoprecipitated ribosomal-RNA–depleted, m6A-containing caRNAs and performed high-throughput sequencing in a methylase deficient (Mettl3) and wild type mouse embryonic stem cells (mESCs). The results were consistent with those obtained by liquid chromatography–tandem mass spectrometry (LC-MS/MS) [67].

RNA-seq-based quantitative methods were also developed. Methods specific for the quantitation of ribose methylation have been recently reviewed by Krogh and Nielsen (2019) [161]. The review discusses quantitative methods for the analysis of 2’O-Me in ribosomal RNA for which the results are comparable with results obtained with other methods, emphasizing the challenges of mapping and quantitating RNA modifications especially in non-stable RNAs. More recently, L Hu et al developed a method, m6A-selective allyl chemical labeling and sequencing (m6A-SAC-seq), to measure m6A RNA modifications at single-base resolution across a transcriptome [162].

Concluding Remarks

Despite the large effort and the fast progress made in the field of RNA modifications during the last few years, understanding of their functions is far from being complete. Better studied are modifications present in stable RNAs. The RNA modifications of the anticodon region of tRNAs are involved in the fine-tuning of translation. Others, outside the anticodon region, are involved in controlling the folding and stability of the molecule, or are responsible for tRNA recognition by aminoacyl synthetases [163]. Modifications of the ribosomal RNA are involved in ribosome biogenesis, function and stability, as well as in antibiotics resistance [164]. Only a few examples from messenger RNA are well described. The m7G of the 5’-cap structure of mRNA that has roles in defining the mRNA reading frame, nuclear export and splicing [165], the editing of adenosine to inosine that leads to altered base-pairing [166], and the pseudouridine of the small nuclear RNA involved in splicing [167, 168]. mRNA modifications have roles in nearly every aspect of the mRNA life cycle and various cellular processes. Proteins that add, recognize, and remove such modifications were identified and characterized, but despite the large effort and the fast progress made in the field, understanding of their functions is far from being complete [169].

A large amount of work during many years led to the development of powerful techniques that allow detection of chemically modified nucleotides in these RNAs, both at specific sites and simultaneously at multiple sites. However, good quantitative methods at base resolution and ways to simultaneously detected multiple types of modifications in the same RNA molecule or transcriptome-wide are still awaiting development.

Declarations

Georgeta N Basturea added sections on BoRed-seq and RBS-sequencing in September 2019 and the section on quantitative analysis of modified RNA in February 2020.

References
  1. Mishima E, Jinno D, Akiyama Y, Itoh K, Nankumo S, Shima H, et al. Immuno-Northern Blotting: Detection of RNA Modifications by Using Antibodies against Modified Nucleosides. PLoS ONE. 2015;10:e0143756 pubmed publisher
  2. Carell T, Brandmayr C, Hienzsch A, Muller M, Pearson D, Reiter V, et al. Structure and function of noncanonical nucleobases. Angew Chem Int Ed Engl. 2012;51:7110-31 pubmed publisher
  3. Cantara W, Crain P, Rozenski J, McCloskey J, Harris K, Zhang X, et al. The RNA Modification Database, RNAMDB: 2011 update. Nucleic Acids Res. 2011;39:D195-201 pubmed publisher
  4. Boccaletto P, Machnicka M, Purta E, Piatkowski P, Baginski B, Wirecki T, et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Res. 2017;: pubmed publisher
  5. Cox D, Gootenberg J, Abudayyeh O, Franklin B, Kellner M, Joung J, et al. RNA editing with CRISPR-Cas13. Science. 2017;358:1019-1027 pubmed publisher
  6. Liu H, Flores M, Meng J, Zhang L, Zhao X, Rao M, et al. MeT-DB: a database of transcriptome methylation in mammalian cells. Nucleic Acids Res. 2015;43:D197-203 pubmed publisher
  7. Machnicka M, Milanowska K, Osman Oglou O, Purta E, Kurkowska M, Olchowik A, et al. MODOMICS: a database of RNA modification pathways--2013 update. Nucleic Acids Res. 2013;41:D262-7 pubmed publisher
  8. Jühling F, Mörl M, Hartmann R, Sprinzl M, Stadler P, Pütz J. tRNAdb 2009: compilation of tRNA sequences and tRNA genes. Nucleic Acids Res. 2009;37:D159-62 pubmed publisher
  9. Frye M, Harada B, Behm M, He C. RNA modifications modulate gene expression during development. Science. 2018;361:1346-1349 pubmed publisher
  10. Roundtree I, Evans M, Pan T, He C. Dynamic RNA Modifications in Gene Expression Regulation. Cell. 2017;169:1187-1200 pubmed publisher
  11. Pelaia P, Volturo P, Rocco M, Mille L, Malpieri R, Spinelli V. [Mechanical ventilation in hyperbaric environment: experimental evaluation of the Drager Hyperlog]. Minerva Anestesiol. 1990;56:1371 pubmed
  12. Phelps K, Morris A, Beal P. Novel modifications in RNA. ACS Chem Biol. 2012;7:100-9 pubmed publisher
  13. Wang X, He C. Dynamic RNA modifications in posttranscriptional regulation. Mol Cell. 2014;56:5-12 pubmed publisher
  14. Liu N, Pan T. RNA epigenetics. Transl Res. 2015;165:28-35 pubmed publisher
  15. Chandola U, Das R, Panda B. Role of the N6-methyladenosine RNA mark in gene regulation and its implications on development and disease. Brief Funct Genomics. 2015;14:169-79 pubmed publisher
  16. Yue Y, Liu J, He C. RNA N6-methyladenosine methylation in post-transcriptional gene expression regulation. Genes Dev. 2015;29:1343-55 pubmed publisher
  17. Schwartz S. Cracking the epitranscriptome. RNA. 2016;22:169-74 pubmed publisher
  18. Chen K, Zhao B, He C. Nucleic Acid Modifications in Regulation of Gene Expression. Cell Chem Biol. 2016;23:74-85 pubmed publisher
  19. Roundtree I, He C. RNA epigenetics--chemical messages for posttranscriptional gene regulation. Curr Opin Chem Biol. 2016;30:46-51 pubmed publisher
  20. Marbaniang C, Vogel J. Emerging roles of RNA modifications in bacteria. Curr Opin Microbiol. 2016;30:50-57 pubmed publisher
  21. Sergiev P, Golovina A, Osterman I, Nesterchuk M, Sergeeva O, Chugunova A, et al. N6-Methylated Adenosine in RNA: From Bacteria to Humans. J Mol Biol. 2016;428:2134-45 pubmed publisher
  22. Lu M, Zhang Z, Xue M, Zhao B, Harder O, Li A, et al. N6-methyladenosine modification enables viral RNA to escape recognition by RNA sensor RIG-I. Nat Microbiol. 2020;5:584-598 pubmed publisher
  23. Fu Y, Luo G, Chen K, Deng X, Yu M, Han D, et al. N6-methyldeoxyadenosine marks active transcription start sites in Chlamydomonas. Cell. 2015;161:879-892 pubmed publisher
  24. Xu C, Liu K, Ahmed H, Loppnau P, Schapira M, Min J. Structural Basis for the Discriminative Recognition of N6-Methyladenosine RNA by the Human YT521-B Homology Domain Family of Proteins. J Biol Chem. 2015;290:24902-13 pubmed publisher
  25. Xiao W, Adhikari S, Dahal U, Chen Y, Hao Y, Sun B, et al. Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. Mol Cell. 2016;61:507-519 pubmed publisher
  26. Xu C, Wang X, Liu K, Roundtree I, Tempel W, Li Y, et al. Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol. 2014;10:927-9 pubmed publisher
  27. Roundtree I, Luo G, Zhang Z, Wang X, Zhou T, Cui Y, et al. YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. elife. 2017;6: pubmed publisher
  28. Zhang Z, Theler D, Kaminska K, Hiller M, de la Grange P, Pudimat R, et al. The YTH domain is a novel RNA binding domain. J Biol Chem. 2010;285:14701-10 pubmed publisher
  29. Patil D, Chen C, Pickering B, Chow A, Jackson C, Guttman M, et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 2016;537:369-373 pubmed publisher
  30. Shima H, Matsumoto M, Ishigami Y, Ebina M, Muto A, Sato Y, et al. S-Adenosylmethionine Synthesis Is Regulated by Selective N6-Adenosine Methylation and mRNA Degradation Involving METTL16 and YTHDC1. Cell Rep. 2017;21:3354-3363 pubmed publisher
  31. Soh Y, Mikedis M, Kojima M, Godfrey A, de Rooij D, Page D. Meioc maintains an extended meiotic prophase I in mice. PLoS Genet. 2017;13:e1006704 pubmed publisher
  32. Hsu P, Zhu Y, Ma H, Guo Y, Shi X, Liu Y, et al. Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 2017;27:1115-1127 pubmed publisher
  33. Bailey A, Batista P, Gold R, Chen Y, de Rooij D, Chang H, et al. The conserved RNA helicase YTHDC2 regulates the transition from proliferation to differentiation in the germline. elife. 2017;6: pubmed publisher
  34. Wojtas M, Pandey R, Mendel M, Homolka D, Sachidanandam R, Pillai R. Regulation of m6A Transcripts by the 3'?5' RNA Helicase YTHDC2 Is Essential for a Successful Meiotic Program in the Mammalian Germline. Mol Cell. 2017;68:374-387.e12 pubmed publisher
  35. Jain D, Puno M, Meydan C, Lailler N, Mason C, Lima C, et al. ketu mutant mice uncover an essential meiotic function for the ancient RNA helicase YTHDC2. elife. 2018;7: pubmed publisher
  36. Wang X, Lu Z, Gomez A, Hon G, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505:117-20 pubmed publisher
  37. Shi H, Wang X, Lu Z, Zhao B, Ma H, Hsu P, et al. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 2017;27:315-328 pubmed publisher
  38. Li A, Chen Y, Ping X, Yang X, Xiao W, Yang Y, et al. Cytoplasmic m6A reader YTHDF3 promotes mRNA translation. Cell Res. 2017;27:444-447 pubmed publisher
  39. Wang X, Zhao B, Roundtree I, Lu Z, Han D, Ma H, et al. N(6)-methyladenosine Modulates Messenger RNA Translation Efficiency. Cell. 2015;161:1388-99 pubmed publisher
  40. Dominissini D, Moshitch Moshkovitz S, Schwartz S, Salmon Divon M, Ungar L, Osenberg S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485:201-6 pubmed publisher
  41. Li F, Zhao D, Wu J, Shi Y. Structure of the YTH domain of human YTHDF2 in complex with an m(6)A mononucleotide reveals an aromatic cage for m(6)A recognition. Cell Res. 2014;24:1490-2 pubmed publisher
  42. Zhu T, Roundtree I, Wang P, Wang X, Wang L, Sun C, et al. Crystal structure of the YTH domain of YTHDF2 reveals mechanism for recognition of N6-methyladenosine. Cell Res. 2014;24:1493-6 pubmed publisher
  43. Zhou J, Wan J, Gao X, Zhang X, Jaffrey S, Qian S. Dynamic m(6)A mRNA methylation directs translational control of heat shock response. Nature. 2015;526:591-4 pubmed publisher
  44. Tan B, Liu H, Zhang S, da Silva S, Zhang L, Meng J, et al. Viral and cellular N6-methyladenosine and N6,2'-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat Microbiol. 2018;3:108-120 pubmed publisher
  45. Hesser C, Karijolich J, Dominissini D, He C, Glaunsinger B. N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi's sarcoma-associated herpesvirus infection. PLoS Pathog. 2018;14:e1006995 pubmed publisher
  46. Tsai K, Courtney D, Cullen B. Addition of m6A to SV40 late mRNAs enhances viral structural gene expression and replication. PLoS Pathog. 2018;14:e1006919 pubmed publisher
  47. Ivanova I, Much C, Di Giacomo M, Azzi C, Morgan M, Moreira P, et al. The RNA m6A Reader YTHDF2 Is Essential for the Post-transcriptional Regulation of the Maternal Transcriptome and Oocyte Competence. Mol Cell. 2017;67:1059-1067.e4 pubmed publisher
  48. Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature. 2017;549:273-276 pubmed publisher
  49. Zhao B, Wang X, Beadell A, Lu Z, Shi H, Kuuspalu A, et al. m6A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature. 2017;542:475-478 pubmed publisher
  50. Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, et al. Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 2017;27:626-641 pubmed publisher
  51. Kang H, Jeong S, Kim K, Baek I, Chang M, Kang C, et al. A novel protein, Pho92, has a conserved YTH domain and regulates phosphate metabolism by decreasing the mRNA stability of PHO4 in Saccharomyces cerevisiae. Biochem J. 2014;457:391-400 pubmed publisher
  52. Harigaya Y, Tanaka H, Yamanaka S, Tanaka K, Watanabe Y, Tsutsumi C, et al. Selective elimination of messenger RNA prevents an incidence of untimely meiosis. Nature. 2006;442:45-50 pubmed
  53. Kan L, Grozhik A, Vedanayagam J, Patil D, Pang N, Lim K, et al. The m6A pathway facilitates sex determination in Drosophila. Nat Commun. 2017;8:15737 pubmed publisher
  54. He Y, Smith R. Nuclear functions of heterogeneous nuclear ribonucleoproteins A/B. Cell Mol Life Sci. 2009;66:1239-56 pubmed publisher
  55. Munro T, Magee R, Kidd G, Carson J, Barbarese E, Smith L, et al. Mutational analysis of a heterogeneous nuclear ribonucleoprotein A2 response element for RNA trafficking. J Biol Chem. 1999;274:34389-95 pubmed
  56. Alarcón C, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie S. HNRNPA2B1 Is a Mediator of m(6)A-Dependent Nuclear RNA Processing Events. Cell. 2015;162:1299-308 pubmed publisher
  57. Villarroya Beltri C, Gutiérrez Vázquez C, Sanchez Cabo F, Pérez Hernández D, Vazquez J, Martín Cofreces N, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun. 2013;4:2980 pubmed publisher
  58. Levesque K, Halvorsen M, Abrahamyan L, Chatel Chaix L, Poupon V, Gordon H, et al. Trafficking of HIV-1 RNA is mediated by heterogeneous nuclear ribonucleoprotein A2 expression and impacts on viral assembly. Traffic. 2006;7:1177-93 pubmed
  59. Moran Jones K, Wayman L, Kennedy D, Reddel R, Sara S, Snee M, et al. hnRNP A2, a potential ssDNA/RNA molecular adapter at the telomere. Nucleic Acids Res. 2005;33:486-96 pubmed
  60. Hoek K, Kidd G, Carson J, Smith R. hnRNP A2 selectively binds the cytoplasmic transport sequence of myelin basic protein mRNA. Biochemistry. 1998;37:7021-9 pubmed
  61. Huang M, Rech J, Northington S, Flicker P, Mayeda A, Krainer A, et al. The C-protein tetramer binds 230 to 240 nucleotides of pre-mRNA and nucleates the assembly of 40S heterogeneous nuclear ribonucleoprotein particles. Mol Cell Biol. 1994;14:518-33 pubmed
  62. Kim J, Paek K, Choi K, Kim T, Hahm B, Kim K, et al. Heterogeneous nuclear ribonucleoprotein C modulates translation of c-myc mRNA in a cell cycle phase-dependent manner. Mol Cell Biol. 2003;23:708-20 pubmed
  63. Shetty S. Regulation of urokinase receptor mRNA stability by hnRNP C in lung epithelial cells. Mol Cell Biochem. 2005;272:107-18 pubmed
  64. Sebillon P, Beldjord C, Kaplan J, Brody E, Marie J. A T to G mutation in the polypyrimidine tract of the second intron of the human beta-globin gene reduces in vitro splicing efficiency: evidence for an increased hnRNP C interaction. Nucleic Acids Res. 1995;23:3419-25 pubmed
  65. Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518:560-4 pubmed publisher
  66. Meyer K, Patil D, Zhou J, Zinoviev A, Skabkin M, Elemento O, et al. 5' UTR m(6)A Promotes Cap-Independent Translation. Cell. 2015;163:999-1010 pubmed publisher
  67. Liu J, Dou X, Chen C, Chen C, Liu C, Xu M, et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science. 2020;367:580-586 pubmed publisher
  68. Akichika S, Hirano S, Shichino Y, Suzuki T, Nishimasu H, Ishitani R, et al. Cap-specific terminal N 6-methylation of RNA by an RNA polymerase II-associated methyltransferase. Science. 2019;363: pubmed publisher
  69. Liu Y, You Y, Lu Z, Yang J, Li P, Liu L, et al. N6-methyladenosine RNA modification-mediated cellular metabolism rewiring inhibits viral replication. Science. 2019;365:1171-1176 pubmed publisher
  70. Wang L, Wen M, Cao X. Nuclear hnRNPA2B1 initiates and amplifies the innate immune response to DNA viruses. Science. 2019;365: pubmed publisher
  71. Pandolfini L, Barbieri I, Bannister A, Hendrick A, Andrews B, Webster N, et al. METTL1 Promotes let-7 MicroRNA Processing via m7G Methylation. Mol Cell. 2019;74:1278-1290.e9 pubmed publisher
  72. Douvlataniotis K, Bensberg M, Lentini A, Gylemo B, Nestor C. No evidence for DNA N6-methyladenine in mammals. Sci Adv. 2020;6:eaay3335 pubmed publisher
  73. Motorin Y, Muller S, Behm Ansmant I, Branlant C. Identification of modified residues in RNAs by reverse transcription-based methods. Methods Enzymol. 2007;425:21-53 pubmed
  74. Kellner S, Burhenne J, Helm M. Detection of RNA modifications. RNA Biol. 2010;7:237-47 pubmed
  75. Bakin A, Ofengand J. Four newly located pseudouridylate residues in Escherichia coli 23S ribosomal RNA are all at the peptidyltransferase center: analysis by the application of a new sequencing technique. Biochemistry. 1993;32:9754-62 pubmed
  76. Ofengand J, Del Campo M, Kaya Y. Mapping pseudouridines in RNA molecules. Methods. 2001;25:365-73 pubmed
  77. Maden B. Mapping 2'-O-methyl groups in ribosomal RNA. Methods. 2001;25:374-82 pubmed
  78. Behm Ansmant I, Helm M, Motorin Y. Use of specific chemical reagents for detection of modified nucleotides in RNA. J Nucleic Acids. 2011;2011:408053 pubmed publisher
  79. Kirpekar F, Hansen L, Rasmussen A, Poehlsgaard J, Vester B. The archaeon Haloarcula marismortui has few modifications in the central parts of its 23S ribosomal RNA. J Mol Biol. 2005;348:563-73 pubmed
  80. Gustafsson C, Persson B. Identification of the rrmA gene encoding the 23S rRNA m1G745 methyltransferase in Escherichia coli and characterization of an m1G745-deficient mutant. J Bacteriol. 1998;180:359-65 pubmed
  81. Toh S, Xiong L, Bae T, Mankin A. The methyltransferase YfgB/RlmN is responsible for modification of adenosine 2503 in 23S rRNA. RNA. 2008;14:98-106 pubmed
  82. Basturea G, Dague D, Deutscher M, Rudd K. YhiQ is RsmJ, the methyltransferase responsible for methylation of G1516 in 16S rRNA of E. coli. J Mol Biol. 2012;415:16-21 pubmed publisher
  83. Basturea G, Rudd K, Deutscher M. Identification and characterization of RsmE, the founding member of a new RNA base methyltransferase family. RNA. 2006;12:426-34 pubmed
  84. Carlile T, Rojas Duran M, Zinshteyn B, Shin H, Bartoli K, Gilbert W. Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells. Nature. 2014;515:143-6 pubmed publisher
  85. Dong Z, Shao P, Diao L, Zhou H, Yu C, Qu L. RTL-P: a sensitive approach for detecting sites of 2'-O-methylation in RNA molecules. Nucleic Acids Res. 2012;40:e157 pubmed publisher
  86. Aschenbrenner J, Marx A. Direct and site-specific quantification of RNA 2'-O-methylation by PCR with an engineered DNA polymerase. Nucleic Acids Res. 2016;44:3495-502 pubmed publisher
  87. Shi J, Zhang Y, Tan D, Zhang X, Yan M, Franklin R, et al. PANDORA-seq expands the repertoire of regulatory small RNAs by overcoming RNA modifications. Nat Cell Biol. 2021;23:424-436 pubmed publisher
  88. Grosjean H, Keith G, Droogmans L. Detection and quantification of modified nucleotides in RNA using thin-layer chromatography. Methods Mol Biol. 2004;265:357-91 pubmed
  89. Grosjean H, Droogmans L, Roovers M, Keith G. Detection of enzymatic activity of transfer RNA modification enzymes using radiolabeled tRNA substrates. Methods Enzymol. 2007;425:55-101 pubmed
  90. 32.
  91. Pintard L, Lecointe F, Bujnicki J, Bonnerot C, Grosjean H, Lapeyre B. Trm7p catalyses the formation of two 2'-O-methylriboses in yeast tRNA anticodon loop. EMBO J. 2002;21:1811-20 pubmed
  92. Purushothaman S, Bujnicki J, Grosjean H, Lapeyre B. Trm11p and Trm112p are both required for the formation of 2-methylguanosine at position 10 in yeast tRNA. Mol Cell Biol. 2005;25:4359-70 pubmed
  93. Roovers M, Kaminska K, Tkaczuk K, Gigot D, Droogmans L, Bujnicki J. The YqfN protein of Bacillus subtilis is the tRNA: m1A22 methyltransferase (TrmK). Nucleic Acids Res. 2008;36:3252-62 pubmed publisher
  94. Roovers M, Hale C, Tricot C, Terns M, Terns R, Grosjean H, et al. Formation of the conserved pseudouridine at position 55 in archaeal tRNA. Nucleic Acids Res. 2006;34:4293-301 pubmed
  95. Lakings D, Gehrke C. Analysis of base composition of RNA and DNA hydrolysates by gas-liquid chromatography. J Chromatogr. 1971;62:347-67 pubmed
  96. Burtis C. The determination of the base composition of RNA by high-pressure cation-exchange chromatography. J Chromatogr. 1970;51:183-94 pubmed
  97. Uziel M, Koh C, Cohn W. Rapid ion-exchange chromatographic microanalysis of ultraviolet-absorbing materials and its application to nucleosides. Anal Biochem. 1968;25:77-98 pubmed
  98. Gehrke C, Kuo K. Ribonucleoside analysis by reversed-phase high-performance liquid chromatography. J Chromatogr. 1989;471:3-36 pubmed
  99. Mazauric M, Dirick L, Purushothaman S, Bjork G, Lapeyre B. Trm112p is a 15-kDa zinc finger protein essential for the activity of two tRNA and one protein methyltransferases in yeast. J Biol Chem. 2010;285:18505-15 pubmed publisher
  100. Larsen M, Roepstorff P. Mass spectrometric identification of proteins and characterization of their post-translational modifications in proteome analysis. Fresenius J Anal Chem. 2000;366:677-90 pubmed
  101. Giessing A, Kirpekar F. Mass spectrometry in the biology of RNA and its modifications. J Proteomics. 2012;75:3434-49 pubmed publisher
  102. Douthwaite S, Kirpekar F. Identifying modifications in RNA by MALDI mass spectrometry. Methods Enzymol. 2007;425:3-20 pubmed
  103. Purta E, O Connor M, Bujnicki J, Douthwaite S. YgdE is the 2'-O-ribose methyltransferase RlmM specific for nucleotide C2498 in bacterial 23S rRNA. Mol Microbiol. 2009;72:1147-58 pubmed publisher
  104. Golovina A, Dzama M, Osterman I, Sergiev P, Serebryakova M, Bogdanov A, et al. The last rRNA methyltransferase of E. coli revealed: the yhiR gene encodes adenine-N6 methyltransferase specific for modification of A2030 of 23S ribosomal RNA. RNA. 2012;18:1725-34 pubmed publisher
  105. Hansen M, Kirpekar F, Ritterbusch W, Vester B. Posttranscriptional modifications in the A-loop of 23S rRNAs from selected archaea and eubacteria. RNA. 2002;8:202-13 pubmed
  106. Yu B, Yang Z, Li J, Minakhina S, Yang M, Padgett R, et al. Methylation as a crucial step in plant microRNA biogenesis. Science. 2005;307:932-5 pubmed
  107. Katoh T, Sakaguchi Y, Miyauchi K, Suzuki T, Kashiwabara S, Baba T, et al. Selective stabilization of mammalian microRNAs by 3' adenylation mediated by the cytoplasmic poly(A) polymerase GLD-2. Genes Dev. 2009;23:433-8 pubmed publisher
  108. Mengel Jørgensen J, Kirpekar F. Detection of pseudouridine and other modifications in tRNA by cyanoethylation and MALDI mass spectrometry. Nucleic Acids Res. 2002;30:e135 pubmed
  109. Durairaj A, Limbach P. Mass spectrometry of the fifth nucleoside: a review of the identification of pseudouridine in nucleic acids. Anal Chim Acta. 2008;623:117-25 pubmed publisher
  110. Taucher M, Ganisl B, Breuker K. Identification, localization, and relative quantitation of pseudouridine in RNA by tandem mass spectrometry of hydrolysis products. Int J Mass Spectrom. 2011;304:91-97 pubmed
  111. Yamauchi Y, Nobe Y, Izumikawa K, Higo D, Yamagishi Y, Takahashi N, et al. A mass spectrometry-based method for direct determination of pseudouridine in RNA. Nucleic Acids Res. 2016;44:e59 pubmed publisher
  112. Felden B, Hanawa K, Atkins J, Himeno H, Muto A, Gesteland R, et al. Presence and location of modified nucleotides in Escherichia coli tmRNA: structural mimicry with tRNA acceptor branches. EMBO J. 1998;17:3188-96 pubmed
  113. Kowalak J, Bruenger E, McCloskey J. Posttranscriptional modification of the central loop of domain V in Escherichia coli 23 S ribosomal RNA. J Biol Chem. 1995;270:17758-64 pubmed
  114. Guymon R, Pomerantz S, Crain P, McCloskey J. Influence of phylogeny on posttranscriptional modification of rRNA in thermophilic prokaryotes: the complete modification map of 16S rRNA of Thermus thermophilus. Biochemistry. 2006;45:4888-99 pubmed
  115. Pomerantz S, McCloskey J. Analysis of RNA hydrolyzates by liquid chromatography-mass spectrometry. Methods Enzymol. 1990;193:796-824 pubmed
  116. Apffel A, Chakel J, Fischer S, Lichtenwalter K, Hancock W. Analysis of Oligonucleotides by HPLC-Electrospray Ionization Mass Spectrometry. Anal Chem. 1997;69:1320-5 pubmed publisher
  117. Willmann L, Erbes T, Krieger S, Trafkowski J, Rodamer M, Kammerer B. Metabolome analysis via comprehensive two-dimensional liquid chromatography: identification of modified nucleosides from RNA metabolism. Anal Bioanal Chem. 2015;407:3555-66 pubmed publisher
  118. Yu Y, Shu M, Steitz J. A new method for detecting sites of 2'-O-methylation in RNA molecules. RNA. 1997;3:324-31 pubmed
  119. Zhao X, Yu Y. Detection and quantitation of RNA base modifications. RNA. 2004;10:996-1002 pubmed
  120. Lapham J, Crothers D. Site-specific cleavage of transcript RNA. Methods Enzymol. 2000;317:132-9 pubmed
  121. Santoro S, Joyce G. A general purpose RNA-cleaving DNA enzyme. Proc Natl Acad Sci U S A. 1997;94:4262-6 pubmed
  122. Hengesbach M, Meusburger M, Lyko F, Helm M. Use of DNAzymes for site-specific analysis of ribonucleotide modifications. RNA. 2008;14:180-7 pubmed
  123. Buchhaupt M, Peifer C, Entian K. Analysis of 2'-O-methylated nucleosides and pseudouridines in ribosomal RNAs using DNAzymes. Anal Biochem. 2007;361:102-8 pubmed
  124. Meusburger M, Hengesbach M, Helm M. A post-labeling approach for the characterization and quantification of RNA modifications based on site-directed cleavage by DNAzymes. Methods Mol Biol. 2011;718:259-70 pubmed publisher
  125. Saikia M, Dai Q, Decatur W, Fournier M, Piccirilli J, Pan T. A systematic, ligation-based approach to study RNA modifications. RNA. 2006;12:2025-33 pubmed
  126. Dai Q, Fong R, Saikia M, Stephenson D, Yu Y, Pan T, et al. Identification of recognition residues for ligation-based detection and quantitation of pseudouridine and N6-methyladenosine. Nucleic Acids Res. 2007;35:6322-9 pubmed
  127. Cozen A, Quartley E, Holmes A, Hrabeta Robinson E, Phizicky E, Lowe T. ARM-seq: AlkB-facilitated RNA methylation sequencing reveals a complex landscape of modified tRNA fragments. Nat Methods. 2015;12:879-84 pubmed publisher
  128. Zheng G, Qin Y, Clark W, Dai Q, Yi C, He C, et al. Efficient and quantitative high-throughput tRNA sequencing. Nat Methods. 2015;12:835-837 pubmed publisher
  129. Addepalli B, Lesner N, Limbach P. Detection of RNA nucleoside modifications with the uridine-specific ribonuclease MC1 from Momordica charantia. RNA. 2015;21:1746-56 pubmed publisher
  130. Simpson J, Workman R, Zuzarte P, David M, Dursi L, Timp W. Detecting DNA cytosine methylation using nanopore sequencing. Nat Methods. 2017;14:407-410 pubmed publisher
  131. McIntyre A, Alexander N, Grigorev K, Bezdan D, Sichtig H, Chiu C, et al. Single-molecule sequencing detection of N6-methyladenine in microbial reference materials. Nat Commun. 2019;10:579 pubmed publisher
  132. Lorenz D, Sathe S, Einstein J, Yeo G. Direct RNA sequencing enables m6A detection in endogenous transcript isoforms at base specific resolution. RNA. 2019;: pubmed publisher
  133. Fu Y, Dominissini D, Rechavi G, He C. Gene expression regulation mediated through reversible m?A RNA methylation. Nat Rev Genet. 2014;15:293-306 pubmed publisher
  134. Eisenberg E, Li J, Levanon E. Sequence based identification of RNA editing sites. RNA Biol. 2010;7:248-52 pubmed
  135. Ramaswami G, Zhang R, Piskol R, Keegan L, Deng P, O Connell M, et al. Identifying RNA editing sites using RNA sequencing data alone. Nat Methods. 2013;10:128-32 pubmed publisher
  136. Schaefer M, Pollex T, Hanna K, Lyko F. RNA cytosine methylation analysis by bisulfite sequencing. Nucleic Acids Res. 2009;37:e12 pubmed publisher
  137. Pollex T, Hanna K, Schaefer M. Detection of cytosine methylation in RNA using bisulfite sequencing. Cold Spring Harb Protoc. 2010;2010:pdb.prot5505 pubmed publisher
  138. Motorin Y, Lyko F, Helm M. 5-methylcytosine in RNA: detection, enzymatic formation and biological functions. Nucleic Acids Res. 2010;38:1415-30 pubmed publisher
  139. Khoddami V, Cairns B. Identification of direct targets and modified bases of RNA cytosine methyltransferases. Nat Biotechnol. 2013;31:458-64 pubmed publisher
  140. Dominissini D, Moshitch Moshkovitz S, Salmon Divon M, Amariglio N, Rechavi G. 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 pubmed publisher
  141. Meyer K, Saletore Y, Zumbo P, Elemento O, Mason C, Jaffrey S. Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons. Cell. 2012;149:1635-46 pubmed publisher
  142. Schwartz S, Agarwala S, Mumbach M, Jovanovic M, Mertins P, Shishkin A, et al. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis. Cell. 2013;155:1409-21 pubmed publisher
  143. Linder B, Grozhik A, Olarerin George A, Meydan C, Mason C, Jaffrey S. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods. 2015;12:767-72 pubmed publisher
  144. Huang H, Weng H, Zhou K, Wu T, Zhao B, Sun M, et al. Histone H3 trimethylation at lysine 36 guides m6A RNA modification co-transcriptionally. Nature. 2019;567:414-419 pubmed publisher
  145. Chen K, Lu Z, Wang X, Fu Y, Luo G, Liu N, et al. High-resolution N(6) -methyladenosine (m(6) A) map using photo-crosslinking-assisted m(6) A sequencing. Angew Chem Int Ed Engl. 2015;54:1587-90 pubmed publisher
  146. Carlile T, Rojas Duran M, Gilbert W. Pseudo-Seq: Genome-Wide Detection of Pseudouridine Modifications in RNA. Methods Enzymol. 2015;560:219-45 pubmed publisher
  147. Carlile T, Rojas Duran M, Gilbert W. Transcriptome-Wide Identification of Pseudouridine Modifications Using Pseudo-seq. Curr Protoc Mol Biol. 2015;112:4.25.1-24 pubmed publisher
  148. Lovejoy A, Riordan D, Brown P. Transcriptome-wide mapping of pseudouridines: pseudouridine synthases modify specific mRNAs in S. cerevisiae. PLoS ONE. 2014;9:e110799 pubmed publisher
  149. Schwartz S, Bernstein D, Mumbach M, Jovanovic M, Herbst R, León Ricardo B, et al. Transcriptome-wide mapping reveals widespread dynamic-regulated pseudouridylation of ncRNA and mRNA. Cell. 2014;159:148-162 pubmed publisher
  150. Li X, Zhu P, Ma S, Song J, Bai J, Sun F, et al. Chemical pulldown reveals dynamic pseudouridylation of the mammalian transcriptome. Nat Chem Biol. 2015;11:592-7 pubmed publisher
  151. Li X, Ma S, Yi C. Pseudouridine Chemical Labeling and Profiling. Methods Enzymol. 2015;560:247-72 pubmed publisher
  152. Zueva V, Mankin A, Bogdanov A, Baratova L. Specific fragmentation of tRNA and rRNA at a 7-methylguanine residue in the presence of methylated carrier RNA. Eur J Biochem. 1985;146:679-87 pubmed
  153. Tanaka M, Han S, Song H, Küpfer P, Leumann C, Sonntag W. An assay for RNA oxidation induced abasic sites using the Aldehyde Reactive Probe. Free Radic Res. 2011;45:237-47 pubmed publisher
  154. Khoddami V, Yerra A, Mosbruger T, Fleming A, Burrows C, Cairns B. Transcriptome-wide profiling of multiple RNA modifications simultaneously at single-base resolution. Proc Natl Acad Sci U S A. 2019;116:6784-6789 pubmed publisher
  155. Macon J, Wolfenden R. 1-Methyladenosine. Dimroth rearrangement and reversible reduction. Biochemistry. 1968;7:3453-8 pubmed
  156. Harcourt E, Ehrenschwender T, Batista P, Chang H, Kool E. Identification of a selective polymerase enables detection of N(6)-methyladenosine in RNA. J Am Chem Soc. 2013;135:19079-82 pubmed publisher
  157. Wang S, Wang J, Zhang X, Fu B, Song Y, Ma P, et al. N6-Methyladenine hinders RNA- and DNA-directed DNA synthesis: application in human rRNA methylation analysis of clinical specimens. Chem Sci. 2016;7:1440-1446 pubmed publisher
  158. Castellanos Rubio A, Santin I, Olazagoitia Garmendia A, Romero Garmendia I, Jauregi Miguel A, Legarda M, et al. A novel RT-QPCR-based assay for the relative quantification of residue specific m6A RNA methylation. Sci Rep. 2019;9:4220 pubmed publisher
  159. Seitz H, Matsushima G. Dendritic cells in systemic lupus erythematosus. Int Rev Immunol. 2010;29:184-209 pubmed publisher
  160. Oshima T, Ishiguro K, Suzuki T, Kawahara Y. Quantification of methylation efficiency at a specific N6-methyladenosine position in rRNA by using BNA probes. Chem Commun (Camb). 2018;54:9627-9630 pubmed publisher
  161. Krogh N, Nielsen H. Sequencing-based methods for detection and quantitation of ribose methylations in RNA. Methods. 2019;156:5-15 pubmed publisher
  162. Hu L, Liu S, Peng Y, Ge R, Su R, Senevirathne C, et al. m6A RNA modifications are measured at single-base resolution across the mammalian transcriptome. Nat Biotechnol. 2022;40:1210-1219 pubmed publisher
  163. Giege R, Sissler M, Florentz C. Universal rules and idiosyncratic features in tRNA identity. Nucleic Acids Res. 1998;26:5017-35 pubmed
  164. Chow C, Lamichhane T, Mahto S. Expanding the nucleotide repertoire of the ribosome with post-transcriptional modifications. ACS Chem Biol. 2007;2:610-9 pubmed
  165. Cowling V. Regulation of mRNA cap methylation. Biochem J. 2009;425:295-302 pubmed publisher
  166. Farajollahi S, Maas S. Molecular diversity through RNA editing: a balancing act. Trends Genet. 2010;26:221-30 pubmed publisher
  167. Yu A, Ge J, Yu Y. Pseudouridines in spliceosomal snRNAs. Protein Cell. 2011;2:712-25 pubmed publisher
  168. Wu G, Yu A, Kantartzis A, Yu Y. Functions and mechanisms of spliceosomal small nuclear RNA pseudouridylation. Wiley Interdiscip Rev RNA. 2011;2:571-81 pubmed publisher
  169. Schaefer M, Kapoor U, Jantsch M. Understanding RNA modifications: the promises and technological bottlenecks of the 'epitranscriptome'. Open Biol. 2017;7: pubmed publisher
ISSN : 2329-5139