RNA Modifications Methods and Protocols
RNA Modifications Methods and Protocols
RNA
Modifications
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Edited by
Mary McMahon
Department of Urology, University of California, San Francisco, San Francisco, CA, USA
Editor
Mary McMahon
Department of Urology
University of California, San Francisco
San Francisco, CA, USA
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
Preface
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415
Contributors
xi
xii Contributors
XIAO-MIN LIU • School of Life Science and Technology, China Pharmaceutical University,
Nanjing, China
ILARIA MANFREDONIA • Department of Molecular Genetics, Groningen Biomolecular Sciences
and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
VIRGINIE MARCHAND • Université de Lorraine, CNRS, INSERM, EpiRNASeq Core Facility,
UMS2008/US40 IBSLor, Nancy, France
NICOLE M. MARTINEZ • Department of Molecular Biophysics and Biochemistry, Yale
University, New Haven, CT, USA
YURI MOTORIN • Université de Lorraine, CNRS, INSERM, EpiRNASeq Core Facility,
UMS2008/US40 IBSLor, Nancy, France; Université de Lorraine, CNRS, UMR7365
IMoPA, Nancy, France
DEEKSHA MUNNUR • Sir William Dunn School of Pathology, University of Oxford, Oxford,
UK
PEI QIN NG • Department of Molecular and Biomedical Sciences, School of Biological
Sciences, The University of Adelaide, Adelaide, SA, Australia
VY NGUYEN • Department of Molecular and Biomedical Sciences, School of Biological
Sciences, The University of Adelaide, Adelaide, SA, Australia
WENHAO NI • Department of Biological and Chemical Sciences, New York Institute of
Technology, New York, NY, USA
EVA MARIA NOVOA • Centre for Genomic Regulation (CRG), The Barcelona Institute of
Science and Technology, Barcelona, Spain; Department of Neuroscience, Garvan Institute
of Medical Research, Darlinghurst, NSW, Australia; St. Vincent’s Clinical School, UNSW
Sydney, Darlinghurst, NSW, Australia; Universitat Pompeu Fabra (UPF), Barcelona,
Spain
ANE OLAZAGOITIA-GARMENDIA • Department of Genetics, Physical Anthropology and Animal
Physiology, University of the Basque Country (UPV-EHU), Leioa, Spain; BioCruces
Bizkaia Health Research Institute, Barakaldo, Spain
HUGH E. OLSEN • Biomolecular Engineering Department and Genomics Institute,
University of California, Santa Cruz, CA, USA
SHU-BING QIAN • Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
BIJOYITA ROY • RNA and Genome Editing, New England Biolabs Inc., Ipswich, MA, USA
SARAH K. SCHULTZ • Department of Chemistry and Biochemistry, Alberta RNA Research
and Training Institute, University of Lethbridge, Lethbridge, AB, Canada
IAIN SEARLE • Department of Molecular and Biomedical Sciences, School of Biological
Sciences, The University of Adelaide, Adelaide, SA, Australia
SHUNDI SHI • Department of Chemical Engineering, Columbia University, New York, NY,
USA
VANJA STOJKOVIĆ • Department of Cellular and Molecular Pharmacology, University of
California San Francisco, San Francisco, CA, USA
CLAUDE THERMES • Institute for Integrative Biology of the Cell, UMR9198, CNRS CEA
Univ Paris-Sud, Université Paris-Saclay, Gif sur Yvette Cedex, France
CARINE TISNÉ • Expression génétique microbienne, UMR 8261, CNRS, Université de Paris,
Institut de biologie physico-chimique (IBPC), Paris, France
KEVIN TSAI • Department of Molecular Genetics and Microbiology, Duke University Medical
Center, Durham, NC, USA; Institute of Biomedical Sciences, Academia Sinica, Taipei,
Taiwan
FRANCESCA TUORTO • Division of Biochemistry, Mannheim Institute for
Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University,
xiv Contributors
Abstract
RNA post-transcriptional modifications (PTMs) are progressively gaining relevance in the study of coding-
independent functions of RNA. RNA PTMs act as dynamic regulators of several aspects of the RNA
physiology, from translation to half-life. Rising interest is supported by the advance of high-throughput
techniques enabling the detection of these modifications on a transcriptome-wide scale. To this end, here
we illustrate the usefulness of RNA Framework, a comprehensive toolkit for the analysis of RNA PTM
mapping experiments, by reanalyzing two published transcriptome-scale datasets of N1-methyladenosine
(m1A) and pseudouridine (Ψ) mapping, based on two different experimental strategies.
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
3
4 Ilaria Manfredonia and Danny Incarnato
2 Materials
2.1 RNA Framework RNA Framework (obtainable from our Git repository: https://
[Link]/dincarnato/RNAFramework) is implemented in
Perl. It requires Perl v5.12 (or greater), with ithreads support and
a 64-bit architecture system running Linux or any other UNIX-
based OS. The following software and packages are also required:
1. Bowtie v1.1.2 or greater ([Link]
[Link]), and/or Bowtie v2.2.7 or greater ([Link]
[Link]/bowtie2/[Link]).
2. SAMTools v1.2 or greater ([Link]
3. BEDTools v2.0 or greater ([Link]
bedtools2/).
4. Cutadapt v2.1 or greater ([Link]
stable/[Link]).
5. ViennaRNA Package v2.2.0 or greater ([Link]
[Link]/RNA/).
6. Perl non-CORE modules ([Link]
– DBD::mysql.
– RNA (installed by the ViennaRNA package).
– XML::LibXML.
– Config::Simple.
$ export PATH=$PATH:$(pwd)/RNAFramework
High-Throughput Mapping of RNA Modifications 5
Table 1
List of SRA accession IDs for the datasets used in this chapter
2.2 Data Retrieval To illustrate a typical data analysis workflow with RNA Framework,
we chose two published datasets. The first one is m1A-seq [5], a
RIP approach applied to map N1-methyladenosine (m1A) in
H. sapiens polyadenylated transcripts. The second dataset is
Pseudo-Seq [6], a single-base resolution method based on the
ability of N-cyclohexyl-N0 -(2-morpholinoethyl)carbodiimide
metho-p-toluenesulfonate (CMC) to generate an alkali-resistant
adduct with the N3 of pseudouridine (Ψ) residues, resulting in
reverse transcription (RT) drop-off and which can be applied to
map Ψ in S. cerevisiae ribosomal RNAs (rRNAs).
1. Data can be obtained from the NCBI Sequence Read Archive
([Link] Using the NCBI SRA
Toolkit ([Link]
toolkitsoft/) it is possible to obtain the raw FASTQ files by
typing:
3 Methods
3.1 Generation of the 1. Data derived from any of the aforementioned PTM mapping
Reference Index techniques (IP-based and single-base resolution) can be
mapped to the reference transcriptome using Bowtie v1, a fast
ungapped read aligner. Bowtie reference transcriptome index
generation is automatized through the rf-index module.
Besides including a set of pre-built reference indexes (display-
able with the “-lp” flag and downloadable through the “-pb
<n>” parameter, where <n> is the code identifying the
desired index), rf-index further allows the creation of tailored
reference indexes. For this purpose, it relies on querying the
UCSC genome database ([Link] for a user-
specified genome assembly and gene annotation. A complete
list of available genome assemblies is available at https://
[Link]/FAQ/[Link].
2. For the purpose of this chapter, we are going to need two
reference indexes: one on H. sapiens RefSeq protein-coding
transcripts and one on S. cerevisiae rRNAs. To obtain the
pre-built indexes, type:
3.3 Counting Reads 1. BAM files generated by rf-map (or any other alignment tool)
are processed by the rf-count module, the core of RNA Frame-
work. This tool is able to calculate the number of RT stops or
mutations (where needed) and the read coverage for each base
of the analyzed transcripts. In our case scenario, m1A-seq sam-
ples are processed by:
$ rf-count -r -f Scerevisiae_rRNA_bt/[Link] -o
rf_count_pseudoseq/ rf_map_pseudoseq/SRR132724*.bam
3.4 Calling PTMs The term “calling” is inherited from the original algorithms used
for the identification of protein-binding sites on DNA from ChIP-
seq experiments. In this case, it consists of the identification of
putative post-transcriptionally modified sites within target tran-
scripts. Two different PTM calling modules are available in RNA
Framework, depending on whether the analysis of RIP or single-
base resolution mapping experiments is being performed.
3.4.1 Calling Peak calling from RIP data is performed using the rf-peakcall
of m1A Peaks module. This tool can work both in the absence and presence of
an input (or control) sample (although in the latter case, called
peaks are of higher confidence). To call m1A peaks, type:
3.4.2 Calling of Ψ Sites Calling of PTMs from single-base resolution experiments is instead
performed using the rf-modcall module. To call sites, type:
$ rf - mo d ca l l -u r f _ co u nt _ ps e ud o s eq / SR R 13 2 7 24 8 .r c -t
rf_count_pseudoseq/[Link] -i rf_count_pseudoseq/index.
rci
Control (RPM)
5.0
4.0
3.0
2.0
1.0
0.0
5.0
IP (RPM)
4.0
3.0
2.0
1.0
0.0
76.0
% of peaks
57.0
38.0
19.0
0.0
0
10
20
30
40
50
60
70
80
90
100
Relative position (%)
Fig. 1 Meta-gene coverage plots for the input (in green) and m1A IP (in blue) samples. On the bottom, the
relative distribution of m1A peaks is also shown (in black)
3.5 Output Files 1. Results are reported in XML format. For each nucleotide of a
and Interpretation transcript, two values are calculated, the score and the ratio. The
score is obtained by evaluating the enrichment of RT drop-off
events on a given base in the treated sample, relatively to both
surrounding bases and the same base in the untreated sample; it
provides a numerical estimate of the likelihood of a given
residue to be modified. The ratio is instead a measure of the
relative modification stoichiometry, calculated as the ratio
between the RT drop-off count and the read coverage of the
base in the treated sample only.
High-Throughput Mapping of RNA Modifications 11
50 18S rRNA
40
30
Score
20
10
0
1.0
0.8
0.6
Ratio
0.4
0.2
0.0
Ψs
Fig. 2 Per-base scores and ratios from Pseudo-Seq analysis of S. cerevisiae 18S rRNA. Known Ψ sites are
highlighted in orange
library(RRNAFramework)
XML<-readModXML("SRR1327249_vs_SRR1327248_sites/[Link]")
par(mfrow = c(2,1))
barplot([Link](unlist(XML$score)), las=1, ylab="Score")
barplot([Link](unlist(XML$ratio)), las=1, ylab="Ratio")
4 Notes
$ rf-rctools merge <RC file #1> <RC file #2> . . . <RC file #n>
$ rf-wiggle <XML file #1> <XML file #2> ... <XML file #n>
$ rf-wiggle -r <XML file #1> <XML file #2> . . . <XML file #n>
Acknowledgments
References
1. Cohn WE, Volkin E (1951) Nucleoside-5- 3. Incarnato D, Oliviero S (2017) The RNA epis-
0
-phosphates from ribonucleic acid. Nature tructurome: uncovering RNA function by
167:483–484. [Link] studying structure and post-transcriptional
167483a0 modifications. Trends Biotechnol
2. Behm-Ansmant I, Helm M, Motorin Y (2011) 35:318–333. [Link]
Use of specific chemical reagents for detection tibtech.2016.11.002
of modified nucleotides in RNA. J Nucleic 4. Incarnato D, Morandi E, Simon LM, Oliviero
Acids 2011:1–17. [Link] S (2018) RNA framework: an all-in-one toolkit
s1355838299981335 for the analysis of RNA structures and post-
High-Throughput Mapping of RNA Modifications 13
Abstract
While over 150 distinct types of chemical modifications are known to occur on various cellular RNAs and
can be dynamically controlled, the function of most of these modifications remains poorly defined.
Collectively, these RNA modifications have been recently termed the “epitranscriptome”. Identification
and annotation of individual RNA modifications throughout the transcriptome are key for studying the role
of the epitranscriptome in the regulation of gene expression and for elucidating the functional relevance of
particular RNA modifications in diverse physiological and disease processes. In this protocol, we demon-
strate how to identify and annotate RNA modifications based on the informatic analysis of methylated RNA
immunoprecipitation and sequencing (MeRIP-seq) data, using RNAmod, a convenient one-stop online
interactive platform for the annotation, analysis, and visualization of mRNA modifications.
Key words RNA modification, Epitranscriptome, Identification, Annotation, Software, Tool, Web
server, RNA-binding protein, RBP, MeRIP-seq
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
15
16 Qi Liu et al.
3 Methods
Quality control
(FastQC and trim_galore)
Read mapping
(STAR)
Modification calling
(MACS2/exomePeak)
Modification annotation
(RNAmod)
Table 1
Example of published m6A MeRIP-seq samples
$ mkdir clean_outdir
$ for sraID in SRR6686554 SRR6686555 SRR6686557 SRR6686558
SRR6686560 SRR6686561 SRR6686563 SRR6686564; do trim_galore
${sraID}.[Link] --phred33 --adapter AGATCGGAAGAGCACA --
length 20 --quality 30 --gzip --output_dir clean_outdir; done
Here, the BAM files are further indexed, which can be used
to visually check the modification peaks in the Integrative
Genomics Viewer (IGV) [24].
8. Modification site/peak calling:
Call the m6A RNA modification peaks based on INPUT
and m6A IP sample using MACS2 [18] or exomePeak [19] (see
Note 5). Here, MACS2 is used to call the modifications:
RNAmod
Single case
Group case
Gene case
Datasets
Users
Outputs
Fig. 2 Overall workflow of RNAmod. RNAmod provides three functional modules, including single case, group
case, and gene case, and is freely available at [Link] or [Link]
[Link]/RNAmod
Fig. 3 Screenshots of the RNAmod analysis outputs. The typical outputs contain the following types of
information: (1) modification location distribution; (2) gene type statistics; (3) coverage plots for different
features; (4) distribution around transcription start/end sites; (5) distribution around translation start/end sites;
(6) distribution around splicing sites; (7) comparison of RNA characteristics between groups; (8) mRNA
metagene plot; (9) enriched sequence motifs; (10) heatmap of modification distribution; (11) Gene Ontology
functional enrichments; (12) pathway functional enrichments; (13) modification target gene annotation list;
(14) detailed list of modification sites; and (15) JBrowse visualization of modification in known modifications
and RBP-binding site context. In the coverage plots, 95% confidence interval (mean standard error of the
mean 1.96) is shown
24 Qi Liu et al.
4 Notes
Acknowledgments
References
1. Helm M, Motorin Y (2017) Detecting RNA Sliz P, Santisteban P, George RE, Richards
modifications in the epitranscriptome: predict WG, Wong KK, Locker N, Slack FJ, Gregory
and validate. Nat Rev Genet 18(5):275–291. RI (2018) mRNA circularization by METTL3-
[Link] eIF3h enhances translation and promotes
2. Peer E, Rechavi G, Dominissini D (2017) Epi- oncogenesis. Nature 561(7724):556–560.
transcriptomics: regulation of mRNA metabo- [Link]
lism through modifications. Curr Opin Chem 8
Biol 41:93–98. [Link] 10. Kadumuri RV, Janga SC (2018) Epitranscrip-
cbpa.2017.10.008 tomic code and its alterations in human disease.
3. Shi H, Wei J, He C (2019) Where, when, and Trends Mol Med 24(10):886–903. https://
how: context-dependent functions of rna [Link]/10.1016/[Link].2018.07.010
methylation writers, readers, and erasers. Mol 11. Livneh I, Moshitch-Moshkovitz S,
Cell 74(4):640–650. [Link] Amariglio N, Rechavi G, Dominissini D
1016/[Link].2019.04.025 (2020) The m(6)A epitranscriptome: transcrip-
4. Meyer KD, Jaffrey SR (2017) Rethinking m(6) tome plasticity in brain development and func-
A readers, writers, and erasers. Annu Rev Cell tion. Nat Rev Neurosci 21(1):36–51. https://
Dev Biol 33:319–342. [Link] [Link]/10.1038/s41583-019-0244-z
1146/annurev-cellbio-100616-060758 12. Barbieri I, Kouzarides T (2020) Role of RNA
5. Dominissini D, Moshitch-Moshkovitz S, modifications in cancer. Nat Rev Cancer.
Schwartz S, Salmon-Divon M, Ungar L, [Link]
Osenberg S, Cesarkas K, Jacob-Hirsch J, 2
Amariglio N, Kupiec M, Sorek R, Rechavi G 13. Zhou Y, Zeng P, Li YH, Zhang Z, Cui Q
(2012) Topology of the human and mouse (2016) SRAMP: prediction of mammalian
m6A RNA methylomes revealed by m6A-seq. N6-methyladenosine (m6A) sites based on
Nature 485(7397):201–206. [Link] sequence-derived features. Nucleic Acids Res
10.1038/nature11112 44(10):e91. [Link]
6. Li X, Xiong X, Zhang M, Wang K, Chen Y, gkw104
Zhou J, Mao Y, Lv J, Yi D, Chen XW, Wang C, 14. Dominissini D, Moshitch-Moshkovitz S,
Qian SB, Yi C (2017) Base-resolution mapping Salmon-Divon M, Amariglio N, Rechavi G
reveals distinct m(1)a methylome in nuclear- (2013) Transcriptome-wide mapping of N(6)-
and mitochondrial-encoded transcripts. Mol methyladenosine by m(6)A-seq based on
Cell 68(5):993–1005. e1009. [Link] immunocapturing and massively parallel
org/10.1016/[Link].2017.10.019 sequencing. Nat Protoc 8(1):176–189.
7. Arango D, Sturgill D, Alhusaini N, Dillman [Link]
AA, Sweet TJ, Hanson G, Hosogane M, Sin- 15. Chen K, Wei Z, Zhang Q, Wu X, Rong R,
clair WR, Nanan KK, Mandler MD, Fox SD, Lu Z, Su J, de Magalhaes JP, Rigden DJ,
Zengeya TT, Andresson T, Meier JL, Coller J, Meng J (2019) WHISTLE: a high-accuracy
Oberdoerffer S (2018) Acetylation of cytidine map of the human N6-methyladenosine
in mRNA promotes translation efficiency. Cell (m6A) epitranscriptome predicted using a
175(7):1872–1886. e1824. [Link] machine learning approach. Nucleic Acids Res
10.1016/[Link].2018.10.030 47(7):e41. [Link]
8. Zhang LS, Liu C, Ma H, Dai Q, Sun HL, gkz074
Luo G, Zhang Z, Zhang L, Hu L, Dong X, 16. Chen W, Xing P, Zou Q (2017) Detecting N
He C (2019) Transcriptome-wide mapping of (6)-methyladenosine sites from RNA transcrip-
internal N(7)-methylguanosine methylome in tomes using ensemble Support Vector
mammalian mRNA. Mol Cell 74 Machines. Sci Rep 7:40242. [Link]
(6):1304–1316. e1308. [Link] 10.1038/srep40242
1016/[Link].2019.03.036 17. Liu Q, Gregory RI (2019) RNAmod: an
9. Choe J, Lin S, Zhang W, Liu Q, Wang L, integrated system for the annotation of
Ramirez-Moya J, Du P, Kim W, Tang S, mRNA modifications. Nucleic Acids Res 47
A Pipeline for Profiling and Annotating RNA Modifications 27
Abstract
RNA modifications play pivotal roles in the RNA life cycle and RNA fate, and are now appreciated as a major
posttranscriptional regulatory layer in the cell. In the last few years, direct RNA nanopore sequencing
(dRNA-seq) has emerged as a promising technology that can provide single-molecule resolution maps of
RNA modifications in their native RNA context. While native RNA can be successfully sequenced using this
technology, the detection of RNA modifications is still challenging. Here, we provide an upgraded version
of EpiNano (version 1.2), an algorithm to predict m6A RNA modifications from dRNA-seq datasets. The
latest version of EpiNano contains models for predicting m6A RNA modifications in dRNA-seq data that
has been base-called with Guppy. Moreover, it can now train models with features extracted from both base-
called dRNA-seq FASTQ data and raw FAST5 nanopore outputs. Finally, we describe how EpiNano can be
used in stand-alone mode to extract base-calling “error” features and current intensity information from
dRNA-seq datasets. In this chapter, we provide step-by-step instructions on how to produce in vitro tran-
scribed constructs to train EpiNano, as well as detailed information on how to use EpiNano to train, test,
and predict m6A RNA modifications in dRNA-seq data.
Key words Oxford Nanopore Technologies, Direct RNA sequencing, Native RNA, RNA modifica-
tion, Base-calling “errors”, In vitro transcription, Support vector machine, N6-methyladenosine,
Nanopore sequencing
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
31
32 Huanle Liu et al.
2 Materials
8. Nanodrop or similar.
9. Agilent TapeStation and accompanying reagents: RNA Screen-
Tape Sample Buffer, RNA ScreenTape Ladder, RNA Screen-
Tape, optical tube strip caps (8 strip), optical tube strips (8
strip), and loading tips.
3 Methods
3.1 Preparation 1. Adjust the water bath to 42 C and place the competent E. coli
of Modified cells into ice.
and Unmodified In 2. Add 3μL of the curlcake plasmid into a 25μL volume of com-
Vitro Transcribed petent cells and mix well. Incubate on ice for 30–45 min.
Constructs to Train 3. Heat shock the cells at 42 C for 45 s, and then incubate on ice
EpiNano for 5 min.
3.1.1 Plasmid 4. Add 500μL warm SOC/SOB medium. Do not pipette and
Transformation incubate at 37 C for 1 h, shaking at 220 rpm (use
and Isolation thermomixer).
5. Spread 200μL transformant on agar plates containing 100μg/μ
L ampicillin.
6. Incubate in a 37 C incubator overnight.
7. Next day, pick a colony and inoculate it in 200 mL LB (with
ampicillin 100μg/μL) for O/N culture (for Maxiprep).
8. Centrifuge the culture in 250 mL centrifuge vessels at
10,000 g for 10 min (either disposable or reusable auto-
claved ones) and isolate plasmid DNA using the Plasmid Maxi
Kit according to the manufacturer’s instructions, resuspending
the final DNA pellet in 500μL RNase-free water.
3.1.2 Enzymatic 1. Digest 10μg DNA (see Note 1) in 250μL volume as outlined
Digestion of Plasmids below and incubate for 4 h (or O/N) at 37 C.
and DNA Cleanup
BSA (100) 2.5μL
Cutsmart buffer (10) 25μL
BamHI-HF 1μL (20 units)
EcoRV-HF 1μL (20 units)
DNA x μL
dH2O Up to 250μL
36 Huanle Liu et al.
3.1.4 In Vitro 1. For each plasmid, set up an IVT reaction by combining the
Transcription Using following reaction components from the AmpliScribe™ T7
AmpliScribe T7-Flash High Yield Transcription Kit with linearized DNA from the
Transcription Reaction step above at RT, in the order listed below (see Note 2).
Substitution of m6ATP in the place of ATP will result in the
generation of RNA containing m6A residues.
Identifying m6A Modifications Using Nanopore Sequencing 37
Component Volume
RNase-free water Up to 20μL
Linearized template DNA 1μg
AmpliScribe T7-flash 10 reaction buffer 2μL
100 mM DTT 2μL
100 mM ATP (or m6ATP) 1.8μL
100 mM CTP 1.8μL
100 mM GTP 1.8μL
100 mM UTP 1.8μL
RiboGuard RNase inhibitor 0.5μL
AmpliScribe T7 flash enzyme solution 2μL
Total 20μL
3.1.5 RNA Cleanup Using 1. Bring the volume of the IVT reaction to 100μL with RNase-
RNeasy Qiagen Kit free water.
2. Follow the step-by-step instructions of the RNeasy Kit accord-
ing to the manufacturer’s protocol.
3. To elute RNA, pipette 20μL RNase-free water directly onto the
RNeasy Mini column membrane. Centrifuge for 15 min at
8000 g to elute.
4. Add another 20μL RNase-free water directly onto the RNeasy
Mini column membrane and centrifuge for 15 min at
8000 g to elute.
5. Measure the quality/quantity of the eluate.
3.1.6 PolyA Tailing 1. Mix the following reagents to proceed with polyA tailing
reaction:
Reagent Volume
Purified RNA (sample from step 3.1.5 1–10μg in 15.5μL nuclease-free
above) water
RNase inhibitor (SUPERaseIN) 0.5μL
10 E. coli poly(A) polymerase reaction 2μL
buffer
(continued)
38 Huanle Liu et al.
Reagent Volume
ATP (10 mM) 1μL
E. coli poly(A) polymerase 1μL
Total 20μL
3.1.7 Bead Cleanup 1. Vortex the RNAClean XP bead stock until homogenous and
of RNA Using RNAClean add 36μL (1.8) RNAClean beads to the RNA sample.
XP Beads 2. Mix by pipetting up and down 10 gently and incubate for
5 min at RT.
3. Place the reaction on the magnet and let it settle for 5–10 min.
4. Slowly aspirate the solution and discard.
5. Add 70% fresh ethanol and incubate for 30 s at RT. Remove
ethanol completely and air-dry for 2 min.
6. Add 20μL water and pipette the beads up and down. Incubate
for 5 min at RT.
7. Use a magnetic stand to separate the beads from the RNA.
Transfer the RNA solution into a new tube.
8. Measure the quality and quantity of the RNA and confirm the
polyA tailing.
3.1.8 Quality Check 1. Load both non-polyA-tailed and polyA-tailed IVT constructs
of PolyA-Tailed RNAs Using into TapeStation (see Note 3) according to the manufacturer’s
TapeStation instructions. Expected results are displayed in Fig. 1.
3.2 Direct RNA 1. Pool each curlcake (for unmodified and m6A modified sepa-
Sequencing Library rately) into a DNA LoBind tube that will contain 200 ng from
Preparation each (800 ng total).
3.2.1 Preparing 2. Adjust the volume to 9μL with nuclease-free water and mix
Input RNA thoroughly by inversion.
3. Spin down briefly in a microfuge.
3.2.2 Adapter Ligation 1. In a 0.2 mL thin-walled PCR tube, mix the reagents in the
following order including some components from the Direct
RNA sequencing kit:
Identifying m6A Modifications Using Nanopore Sequencing 39
d
ile
ile
-ta
-ta
pA
pA
M
6A
6A
UN
UN
er
m
dd
1
CC
CC
CC
CC
La
6000
4000
2000
1000
500
200
25
Fig. 1 TapeStation image of the Curlcake1 (CC1) IVT products, which were in vitro transcribed with or without
m6A modifications, before and after polyA tailing. Ladder illustrates the size of distinct bands on the electronic
gel. Each IVT product shows increased size upon polyA tailing
3.2.3 Reverse 1. Mix the following reagents together to make the reverse tran-
Transcription and Cleanup scription master mix:
40 Huanle Liu et al.
2. Add the master mix to the 0.2 mL PCR tube containing the RT
Adapter-ligated RNA from the “RT Adapter ligation” step
above. Mix by pipetting.
3. Add 2μl SuperScript III reverse transcriptase to the reaction
and mix by pipetting.
4. Place the tube in a thermal cycler, incubate at 50 C for 50 min
and 70 C for 10 min, and bring the sample to 4 C before
proceeding to the next step.
5. Transfer the sample to a 1.5 mL DNA LoBind Eppendorf tube.
6. Resuspend the stock of Agencourt RNAClean XP beads by
vortexing, add 72μL beads to the reverse transcription reac-
tion, and mix by pipetting.
7. Incubate on a Hula Mixer for 5 min at RT.
8. Prepare 200μL of fresh 70% ethanol in nuclease-free water.
9. Spin down the sample and pellet on a magnet.
10. Keep the tube on the magnet and wash the beads with 150μL
70% ethanol without disturbing the pellet.
11. Remove the ethanol and discard. Spin down tubes, place back
on the magnetic rack, and remove any residual ethanol.
12. Remove the tube from the magnetic rack and resuspend the
pellet in 20μL nuclease-free water. Incubate for 5 min at RT.
13. Pellet the beads on the magnet until the eluate is clear and
colorless.
14. Pipette 20μL of the eluate into a clean 1.5 mL Eppendorf DNA
LoBind tube.
15. Measure cDNA and RNA on a Qubit or similar.
3.2.4 RMX Adapter 1. In a clean 1.5 mL Eppendorf DNA LoBind tube, mix the
Ligation and Cleanup reagents in the following order:
Identifying m6A Modifications Using Nanopore Sequencing 41
Volume
Reagents (μL)
Reverse-transcribed RNA from the “reverse transcription” 20
step
5 NEBNext Quick ligation reaction buffer 8
RNA adapter (RMX) 6
Nuclease-free water 3
Concentrated T4 DNA ligase 3
3.3 Analysis of Direct Base-calling should be performed using Oxford Nanopore Tech-
RNA Sequencing nologies’ Guppy base-caller, such as in the example shown below
Datasets: Base-Calling (see Note 5):
and Mapping
guppy_basecaller --device cuda:0 -c rna_r9.4.1_70bps_hac.cfg
3.3.1 Base-Calling --compress_fastq -i path/to/fast5_directory -r -s /path/to/
save/basecalling_out --fast5_out
42 Huanle Liu et al.
3.3.2 Mapping Map the base-called reads to the reference FASTA sequences using
minimap2 [25], and keep only the mapped reads:
3.4 Extraction The latest version of the EpiNano suite (version 1.2) consists of five
of Features to Detect main programs or modules:
RNA Modifications
– Epinano_Variants computes systematic base-calling “errors”
in Direct RNA (mismatch, deletion, insertion, and per-base quality score) for
Sequencing Datasets each base along the mapped reads and reports their relative
Using EpiNano frequencies in a plain text file.
– Epinano_Current extracts raw current intensity values and dwell
time for each reference base.
– Epinano_Predict trains models using the features extracted with
the aforementioned two modules and makes predictions using
trained models.
– Epinano_DiffErr predicts RNA modifications based on the dif-
ferences in base-calling “errors” between two samples (typically
wild type and knockout).
– Epinano_Plot produces scatterplots or barplots depicting the
differences in base-calling “errors” or modification probabilities
between two samples, highlighting positions that are identified
by the algorithm as significantly altered, i.e., predicted as differ-
entially modified.
EpiNano 1.2 can predict RNA modifications from direct RNA
sequencing datasets using two distinct strategies: (1) EpiNano-
SVM, which employs pre-trained SVM models to predict RNA
modifications, and (2) EpiNano-Error, which uses the differences
between base-calling “errors” (mismatches, deletions, insertions)
between two samples, as well as alterations in per-base qualities, to
predict RNA modifications (Fig. 2). Both strategies rely on the fact
that RNA modifications appear as systematic base-calling “errors”
in direct RNA sequencing datasets.
Identifying m6A Modifications Using Nanopore Sequencing 43
Epinano_DiffError Epinano_Predict
Prediction of
• Compute Sum of Errors
RNA-modified sites • Outlier Detection • Train SVM models
• Predict RNA mods using
Epinano_Plot pre-trained models (ProbM)
Fig. 2 Overview of the five main modules included in the EpiNano 1.2 suite. The latest version of EpiNano can
predict RNA modifications using two distinct strategies: (1) EpiNano-Error, which detects RNA modifications
using differential base-calling “errors” that are detected between two samples (typically wild type and
knockout), and (2) EpiNano-SVM, which detects RNA modifications using support vector machines (SVM),
where the SVM models have been pre-trained with modified and unmodified datasets
The “--type” flag indicates the type of reference that was used
to obtain the bam file. If the reads were mapped to a genome
reference with splicing-aware mapping options, “--type g”
should be specified, and EpiNano will discriminate the reads
mapped to the forward strand from those mapped to the reverse
strand. Otherwise, by default, the script assumes that the bam file
44 Huanle Liu et al.
3.4.2 Extraction The latest version of EpiNano (v.1.2) relies on the use of Nanopolish
of Current Intensity Values [26] to extract the current signal-level information and collapse it
Using EpiNano on a single-position basis. We offer a custom bash script, which
carries out Nanopolish’s eventalign function and further collapses
the current intensity and dwell time values. Epinano_Variants
will produce a file of per-position results consisting of raw current
intensity values and their corresponding mean, median, and stan-
dard deviations as well as a second file with results organized in
5-mer windows:
sh $EPINANO_HOME/misc/Epinano_Current.sh -b [Link]
-r [Link] -f [Link] -t 6 -m g -d fas-
t5_folder/
python $EPINANO_HOME/misc/Join_variants_currents.py
--variants sample.per_site_var.[Link]
--intensity sample.per_site_current.[Link]
--outfile sample.5mer.all_features.csv
3.5 Predicting RNA 1. Label and merge the “modified” and “unmodified” datasets.
Modifications In Vivo To train a model, we first have to label the files containing
Using Trained SVM EpiNano-extracted features that will be used for training the
Models (EpiNano-SVM) model (“mod.per_site.[Link]” and “unm.per_si-
[Link]”) by adding the corresponding labels (“mod”
3.5.1 Train EpiNano and “unm”), as shown below:
Models
bash $EPINANO_HOME/misc/Epinano_LabelSamples.sh -m mod.per_-
[Link] -u unm.per_site.[Link] -o combined.per_site_r-
aw_feature.[Link]
python $EPINANO_HOME/Epinano_Predict.py
--train combined.per_site_raw_feature.[Link]
--predict combined.per_site_raw_feature.[Link]
--accuracy_estimation --out_prefix train_and_test
--columns 8,13,23 --modification_status_column 26
While the user can choose to train the algorithm with one
sample (--train) and test it on an independent sample (--pre-
dict), it is also possible to use the same input file both for training
and testing the model, as depicted in the example above. In this
scenario, Epinano_Predict will train the models with 50% of the
input data and make predictions with the remaining 50% of
the data.
In the above command, “--columns” denotes the column
indexes of features that are used for training models (in this case,
corresponding to “q3,” “mis3,” and “del3”), while “--modifi-
cation_status_column” indicates the index of the column of
prior knowledge of the modification statuses, i.e., the labels “mod”
and “unm.” Switching on --accuracy_estimation will report
the accuracy of the trained model(s). Unless “--kernel” is used,
Epinano_Predict will train models with multiple kernels. Finally,
the user can visualize the accuracy of their trained models in the
form of receiver operating characteristic (ROC) curves (Fig. 3).
python $EPINANO_HOME/Epinano_Predict.py
--model $EPINANO_HOME/models/[Link]-
[Link]
--predict sample.per_site.[Link]
--columns 8,13,23
--out_prefix sample_mod_prediction
46 Huanle Liu et al.
Fig. 3 ROC curves depicting the modification detection performance using in vitro test data (not used for
training) which includes 263 RRACH k-mers. (a) All three SVM models were trained with the same subset of
features (q3, mis3, and del3) described previously [21], using three different kernels (linear, poly and rbf). (b)
The models were trained using the difference of these features (Δq3, Δmis3, and Δdel3) between the
modified and unmodified positions, in a pairwise manner. AUC represents the area under the curve
Fig. 4 ROC curve depicting the performance of m6A modification detection in in vivo data. All shown models
were trained with a linear kernel but using distinct features. Raw features across replicates were combined as
previously described [21] (see Note 12)
3.6 Predicting RNA In Sect. 3.5 we have showcased the use of base-calling “error”
Modifications In Vivo features to train support vector machine (SVM) models to predict
from Base-Calling m6A RNA modifications, using the Epinano_Predict module,
“Error” Differences which was the original strategy employed by EpiNano to predict
(EpiNano-Error) RNA modifications [21]. In the EpiNano 1.2 suite, we now provide
a new module, Epinano_DiffErr, that can predict RNA modifi-
cations by identifying those positions that show differential base-
calling “errors”—previously extracted using the Epinano_Var-
iants—when comparing two samples (e.g., a wild-type and
knockout condition):
2826
0.75 2883
2880
Summed Error (WT)
2882
0.50
2885 2827
0.25
2877
0.00
4 Notes
10. False positives can also be caused by low coverage. RNA mod-
ifications should be predicted on sites with high coverage. We
recommend a minimum coverage of 20–30 reads for a k-mer to
be included in the analysis.
11. FAST5 data used in this work are the same from [21] and can
be obtained from SRA database through the accession code
SRP174366. Intermediate datasets used for this chapter can be
found at [Link]
EpinanoBookChapter.
12. Raw features across the three replicates were merged as previ-
ously described [21] using the following pseudo-code:
Acknowledgments
We thank all members of the Novoa lab for their valuable insights
and discussion. We thank Rebeca Medina for obtaining the TapeS-
tation image used for Fig. 1. OB is supported by an international
PhD fellowship (UIPA) from the University of New South Wales.
This work was supported by the Australian Research Council
(DP180103571 to EMN) and the Spanish Ministry of Economy,
Industry and Competitiveness (MEIC) (PGC2018-098152-A-100
to EMN). We acknowledge the support of the MEIC to the EMBL
partnership, Centro de Excelencia Severo Ochoa, and CERCA
Program/Generalitat de Catalunya.
Identifying m6A Modifications Using Nanopore Sequencing 51
References
1. Cohn WE, Volkin E (1951) Nucleoside-5- m6A mRNA methylation during haematopoie-
0
-phosphates from ribonucleic acid. Nature tic stem cell differentiation. Nat Cell Biol
167:483–484 21:700–709
2. Adams JM, Cory S (1975) Modified nucleo- 12. Zhao BS, He C (2015) Fate by RNA methyla-
sides and bizarre 50 -termini in mouse myeloma tion: m6A steers stem cell pluripotency.
mRNA. Nature 255:28–33 Genome Biol 16:43
3. Desrosiers R, Friderici K, Rottman F (1974) 13. Geula S, Moshitch-Moshkovitz S,
Identification of methylated nucleosides in Dominissini D, Mansour AA, Kol N, Salmon-
messenger RNA from Novikoff hepatoma Divon M, Hershkovitz V, Peer E, Mor N,
cells. Proc Natl Acad Sci U S A 71:3971–3975 Manor YS, Ben-Haim MS, Eyal E, Yunger S,
4. Dubin DT, Taylor RH (1975) The methylation Pinto Y, Jaitin DA, Viukov S, Rais Y,
state of poly A-containing messenger RNA Krupalnik V, Chomsky E, Zerbib M, Maza I,
from cultured hamster cells. Nucleic Acids Rechavi Y, Massarwa R, Hanna S, Amit I, Leva-
Res 2:1653–1668 non EY, Amariglio N, Stern-Ginossar N,
5. Perry RP, Kelley DE, Friderici K, Rottman F Novershtern N, Rechavi G, Hanna JH (2015)
(1975) The methylated constituents of L cell Stem cells. m6A mRNA methylation facilitates
messenger RNA: evidence for an unusual clus- resolution of naı̈ve pluripotency toward differ-
ter at the 50 terminus. Cell 4:387–394 entiation. Science 347:1002–1006
6. Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, 14. Lence T, Akhtar J, Bayer M, Schmid K,
Yi C, Lindahl T, Pan T, Yang Y-G, He C (2011) Spindler L, Ho CH, Kreim N, Andrade-
N6-methyladenosine in nuclear RNA is a major Navarro MA, Poeck B, Helm M, Roignant
substrate of the obesity-associated FTO. Nat J-Y (2016) m6A modulates neuronal functions
Chem Biol 7:885–887 and sex determination in Drosophila. Nature
540:242–247
7. Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang
C-M, Li CJ, Vågbø CB, Shi Y, Wang W-L, 15. Haussmann IU, Bodi Z, Sanchez-Moran E,
Song S-H, Lu Z, Bosmans RPG, Dai Q, Hao Mongan NP, Archer N, Fray RG, Soller M
Y-J, Yang X, Zhao W-M, Tong W-M, Wang (2016) m6A potentiates Sxl alternative
X-J, Bogdan F, Furu K, Fu Y, Jia G, Zhao X, pre-mRNA splicing for robust Drosophila sex
Liu J, Krokan HE, Klungland A, Yang Y-G, He determination. Nature 540:301–304
C (2013) ALKBH5 is a mammalian RNA 16. Helm M, Motorin Y (2017) Detecting RNA
demethylase that impacts RNA metabolism modifications in the epitranscriptome: predict
and mouse fertility. Mol Cell 49:18–29 and validate. Nat Rev Genet 18:275–291
8. Dominissini D, Moshitch-Moshkovitz S, 17. Li X, Xiong X, Yi C (2016) Epitranscriptome
Schwartz S, Salmon-Divon M, Ungar L, sequencing technologies: decoding RNA mod-
Osenberg S, Cesarkas K, Jacob-Hirsch J, ifications. Nat Methods 14:23–31
Amariglio N, Kupiec M, Sorek R, Rechavi G 18. Linder B, Grozhik AV, Olarerin-George AO,
(2012) Topology of the human and mouse Meydan C, Mason CE, Jaffrey SR (2015)
m6A RNA methylomes revealed by m6A-seq. Single-nucleotide-resolution mapping of m6A
Nature 485:201–206 and m6Am throughout the transcriptome. Nat
9. Meyer KD, Saletore Y, Zumbo P, Elemento O, Methods 12:767–772
Mason CE, Jaffrey SR (2012) Comprehensive 19. Novoa EM, Mason CE, Mattick JS (2017)
analysis of mRNA methylation reveals enrich- Charting the unknown epitranscriptome. Nat
ment in 30 UTRs and near stop codons. Cell Rev Mol Cell Biol 18:339–340
149:1635–1646 20. Jonkhout N, Tran J, Smith MA, Schonrock N,
10. Hu Y, Ouyang Z, Sui X, Qi M, Li M, He Y, Mattick JS, Novoa EM (2017) The RNA mod-
Cao Y, Cao Q, Lu Q, Zhou S, Liu L, Liu L, ification landscape in human disease. RNA
Shen B, Shu W, Huo R (2020) Oocyte compe- 23:1754–1769
tence is maintained by m6A methyltransferase 21. Liu H, Begik O, Lucas MC, Ramirez JM,
KIAA1429-mediated RNA metabolism during Mason CE, Wiener D, Schwartz S, Mattick
mouse follicular development. Cell Death Dif- JS, Smith MA, Novoa EM (2019) Accurate
fer. [Link] detection of m6A RNA modifications in native
0516-1 RNA sequences. Nat Commun 10:4079
11. Lee H, Bao S, Qian Y, Geula S, Leslie J, 22. Lorenz DA, Sathe S, Einstein JM, Yeo GW
Zhang C, Hanna JH, Ding L (2019) Stage- (2020) Direct RNA sequencing enables m6A
specific requirement for Mettl3-dependent
52 Huanle Liu et al.
detection in endogenous transcript isoforms at (2019) Direct RNA sequencing reveals m6A
base-specific resolution. RNA 26:19–28 modifications on adenovirus RNA are neces-
23. Parker MT, Knop K, Sherwood AV, Schurch sary for efficient splicing. bioRxiv 865485
NJ, Mackinnon K, Gould PD, Hall AJ, Barton 25. Li H (2018) Minimap2: pairwise alignment for
GJ, Simpson GG (2020) Nanopore direct RNA nucleotide sequences. Bioinformatics
sequencing maps the complexity of Arabidopsis 34:3094–3100
mRNA processing and m6A modification. elife 26. Loman NJ, Quick J, Simpson JT (2015) A
9. [Link] complete bacterial genome assembled de novo
24. Price AM, Hayer KE, McIntyre ABR, Gokhale using only nanopore sequencing data. Nat
NS, Della Fera AN, Mason CE, Horner SM, Methods 12:733–735
Wilson AC, Depledge DP, Weitzman MD
Chapter 4
Abstract
Historically, RNA has been sequenced as cDNA copies derived from reverse transcription of cellular RNA
followed by PCR amplification. Recently, RNA sequencing using nanopores has emerged as an alternative.
Using this technology, individual cellular RNA strands are read directly as they are driven through nanoscale
pores by an applied voltage. The speed of translocation is regulated by a helicase that is loaded onto each
RNA strand by an adapter that also facilitates capture by the nanopore electric field. Here we describe a
technique for adapting human ribosomal RNA subunits for nanopore sequencing. Using this strategy, a
single Oxford Nanopore MinION run delivered 470,907 sequence reads of which 396,048 aligned
to ribosomal RNA, with 28S, 18S, 5.8S, and 5S coverage of 6053, 369,472, 16,058, and 4465
reads, respectively. Example alignments that reveal putative nucleotide modifications are provided.
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
53
54 Miten Jain et al.
Fig. 1 Schematic of the human rRNA adaption and nanopore sequencing protocol. 18S rRNA is given as an
example. (a) (i) Total RNA extraction using TRIzol and chloroform. (ii) Annealing and ligation of a nanopore-
specific adapter to the 30 end of human 18S rRNA (red). Thirteen nucleotides of the adapter are reverse
complements to 13 nucleotides of the target strand. (iii) Ligation of the nanopore sequencing adapter that has
the motor protein preloaded. (iv) Addition of the adapted library to the nanopore for sequencing. (v) Basecalling
of individual nanopore RNA strands using Guppy. This process yields individual RNA strand sequence reads.
(b) A representative ionic current trace for a human 18S rRNA strand processed on the nanopore. Ionic current
components: (i) strand capture; (ii) ONT and 18S rRNA-splint adapter translocation; (iii) human 18S rRNA
translocation; and (iv) exit of the strand into the transcompartment. (c) Expanded view of the region where the
ionic current transitions from the nanopore adapter to the 30 end of the adapted 18S rRNA
Nanopore Sequencing of Human Nuclear rRNA 55
2 Materials
2.2 Nanopore 1. NEB Quick Ligase Buffer and T4 DNA Ligase (2000 U/μL).
Sequencing 2. ONT SQK-RNA002 kit (or newer).
of Biological
3. Tris-NaCl-EDTA buffer: 10 mM Tris–HCl, pH 8, 1 mM
Human rRNA
EDTA, 50 mM NaCl.
4. 100% Ethanol.
5. Beckman Coulter Agencourt RNAClean XP Beads.
6. Magnet for bead-based purifications.
7. Nuclease-free water.
8. Oligomers for preparing biological human rRNA splints. The
sequence for individual oligomers is below:
Top strand: This oligomer is common across all the splints.
Note that this oligomer needs to have a 50 -phosphate group for
ligation in sequencing library preparation:
50 -/5PHOS/GGCTTCTTCTTGCTCTTAGGTAGTA
GGTTC-30
Human 5S rRNA bottom strand:
50 -CCTAAGAGCAAGAAGAAGCCAAAGCCTACAGCA-30
Human 5.8S rRNA bottom strand:
50 -CCTAAGAGCAAGAAGAAGCCAAGCGACGCTCAG-30
Human 18S rRNA bottom strand:
50 -CCTAAGAGCAAGAAGAAGCCTAATGATCCTTCC-30
Human 28S rRNA bottom strand:
50 -CCTAAGAGCAAGAAGAAGCCGACAAACCCTTGT-30
2.4 Nanopore Please refer to the Oxford Nanopore Technologies website for
Sequencing Hardware details:
and Software, [Link] For
and Reagents new users, the MinION Starter Pack is a good option.
3 Methods
3.1 RNA Isolation 1. Add 4 mL of TRI Reagent per frozen pellet of 5 107 cells,
and vortex immediately.
2. Incubate this sample at room temperature for 5 min.
3. Add 400μL BCP (1-bromo-3-chloro-propane) or 200μL
CHCl3 (chloroform) per mL of sample, followed by vigorous
mixing by inversion.
4. Incubate this mixture at room temperature for 5 min and then
mix vigorously again.
5. Spin the mixed sample for 10 min at 12,000 g (4 C).
58 Miten Jain et al.
3.2 Oligomer Splint The sequence for individual oligomers is shown in Subheading.
Preparation for RNA 2.2.
Adaptation
1. To make an oligomer splint adapter, the top and the bottom
strands (four annealing reactions total) are hybridized at 10μM
each in TNE buffer.
2. Heat the mixture at 75 C for 1 min before slowly cooling to
room temperature in a thermocycler.
3.3 Splint Annealing, The following steps are taken from the Oxford Nanopore Direct
Ligation, and Cleanup RNA Sequencing protocol using the SQK-RNA002 kit (see
Note 3).
1. Prepare the RNA in nuclease-free water by transferring
1000 ng RNA to a 1.5 mL Eppendorf DNA LoBind tube.
Adjust the volume to 9μL with nuclease-free water. Mix thor-
oughly by flicking the tube to avoid unwanted shearing. Spin
down briefly in a microfuge.
2. In a 0.2 mL thin-walled PCR tube, mix the reagents in the
order outlined below. The custom rRNA splint adapter mix is
made by combining 1μL volume from each of the 10μM olig-
omer splints (5S, 5.8S, 18S, 28S).
Reagent Volume
NEBNext quick ligation reaction buffer 3.0μL
(5)
RNA CS (RCS), 110 nM (optional) 0.5μL
RNA 6.5μL (or 6.0μL if adding
RCS)
(continued)
Nanopore Sequencing of Human Nuclear rRNA 59
Reagent Volume
Custom rRNA splint adapter mix 4.0μL
T4 DNA Ligase (2000 U/μL) 1.5μL
Total 15.0μL
3.4 Nanopore The following steps are taken from the Direct RNA Sequencing
Sequencing Adapter protocol using the SQK-RNA002 kit, both of which are supplied
Ligation and Cleanup by Oxford Nanopore Technologies.
60 Miten Jain et al.
Reagent Volume
Splint-annealed RNA from Subheading 3.3 (step 19) 20.0μL
NEBNext quick ligation reaction buffer (5) 8.0μL
RNA adapter (RMX) 6.0μL
Nuclease-free water 3.0μL
T4 DNA Ligase (2000 U/μL) 3.0μL
Total 40.0μL
2. Mix by pipetting.
3. Incubate the reaction for 10 min at room temperature.
4. Resuspend the stock of Agencourt RNAClean XP beads by
vortexing.
5. Add 60μL (1.5) of resuspended RNAClean XP beads to the
adapter ligation reaction and mix by pipetting.
6. Incubate on a Hula mixer (rotator mixer) for 5 min at room
temperature.
7. Spin down the sample and pellet on a magnet. Keep the tube
on the magnet, and pipette off the supernatant.
8. Add 150μL of the wash buffer (WSB) from the SQK-RNA002
kit to the beads. Close the tube lid and resuspend the beads by
flicking the tube.
9. Return the tube to the magnetic rack, allow beads to pellet, and
pipette off the supernatant.
10. Repeat steps 8 and 9.
11. Remove the tube from the magnetic rack and resuspend pellet
in 21μL elution buffer from the kit by gently flicking the tube.
12. Incubate for 10 min at room temperature.
13. Pellet the beads on a magnet until the eluate is clear and
colorless.
14. Remove and retain 21μL of eluate into a clean 1.5 mL Eppen-
dorf DNA LoBind tube.
15. [Optional] Quantify 1μL of nanopore-adapted rRNA using the
Qubit fluorometer RNA HS assay (recovery aim ~200 ng).
3.5 Nanopore For this part we recommend following the instructions provided by
Sequencing Oxford Nanopore Technologies. Below are the steps to follow for
the SQK-RNA002 kit. The steps involved here include:
Nanopore Sequencing of Human Nuclear rRNA 61
3.6 Basecalling Nanopore data are basecalled using Guppy software that is provided
by ONT. Guppy has a model to be used for Direct RNA Sequenc-
ing runs. Please follow instructions from ONT for using the newest
version of Guppy.
3.7 Data Analysis Nanopore basecalling yields sequence reads in FASTQ format.
and Visualization These can be aligned to the reference human rRNA sequences
(in FASTA format) using minimap2 (with -ax map-ont setting)
[14]. This process will yield a SAM file (human-readable alignment
format) that can then be converted to a sorted BAM file (machine-
readable alignment file) using SAMtools [15]. Once this is done,
the sorted BAM file and the reference sequence FASTA file can be
uploaded into Integrative Genomics Viewer (IGV) [16] for visual
inspection or used for downstream analyses. IGV can be down-
loaded from the Broad Institute at no cost (Fig. 2) (see Notes 4
and 5).
3.9 Optional: IVT-derived rRNA strands can be analyzed using nanopore devices
Nanopore Sequencing and used to train algorithms that identify modified rRNA nucleo-
of Human rRNA Copies tides. In summary, RNA synthesis of the canonical controls is
Composed performed by in vitro transcription (IVT) using the HiScribe™
of Canonical T7 Quick High Yield RNA Synthesis kit per the manufacturer’s
Nucleotides protocol. For the 5S and 5.8S rRNAs, synthetic DNA oligonucleo-
tides are used for the templates. For the 18S rRNA, a PCR template
is amplified from human genomic DNA. For the 28S rRNA, a
plasmid template constructed by Taoka et al. [13] is used. The
IVT products are purified prior to sequencing. Gel purification is
recommended for all IVTs and is required for making 28S rRNA.
The library preparation for the purified 5S, 5.8S, and 18S IVT
products follows the same procedures described for biological
rRNA described starting in Subheading 3.3. The gel-purified 28S
IVT product is polyadenylated with E. coli poly(A) polymerase.
Following cleanup of the polyadenylated 28S IVT product, it is
sequenced using the standard SQK-RNA002 kit protocol.
62 Miten Jain et al.
Fig. 2 IGV alignment of human 18S rRNA reads. Reads were aligned to the 18S rDNA reference genome
sequence [13] using minimap2. The top axis is the nucleotide position in the 1858 nt long 18S rRNA reference
sequence [13]. Gray color represents alignment coverage where read and reference sequences agree. In the
body of the alignment, purple colors represent insertions, white spaces with black lines represent deletions,
and the red, blue, green, and orange colors represent base-specific mismatches in the alignment. Each
horizontal line is an individual nanopore strand read. The density plot below the reference coordinates
represents alignment coverage. The expanded view shows examples of features within a 40 nt region.
These include alignment coverage, insertions, deletions, mismatches, and alignment coverage density, as
described above. The consistent C miscalls at position 406 reveal a likely uridine-to-pseudouridine
modification
Nanopore Sequencing of Human Nuclear rRNA 63
Table 1
Number of aligned nanopore strand reads per rRNA class
3.9.1 Synthesis of 5S 1. In two PCR tubes, one for the 5S and the other for the 5.8S
and 5.8S rRNA rRNA, combine 0.7μL 10 Tris-NaCl-EDTA, 165 pmol
(1.65μL of 100μM) of the top-strand oligonucleotide,
33 pmol (0.66μL of 50μM) of bottom-strand oligonucleotide,
and nuclease-free H20 (4.69μL) to a total volume of 7μL.
2. Heat the mixture to 75 C for 15 s and slowly cool to 25 C in a
thermocycler to anneal the top and bottom DNA strands.
3. Assemble the IVT reactions in PCR tubes using the HiScribe™
T7 Quick High Yield RNA Synthesis kit reagents as follows:
Reagent Volume
Annealed top and bottom DNA strands from step 2 7.0μL
NTP buffer 10.0μL
T7 RNA polymerase mix 2.0μL
RNasin (40 units/μL) or equivalent RNase inhibitor 1μL
Total 20.0μL
3.9.3 PCR Synthesis Generate the DNA template with PCR using Q5 master mix (2).
of the DNA Template
1. Input the Following PCR Protocol into Thermocycler:
for 18S rRNA IVT
Initial denaturation: 30 s at 98 C
33 cycles of:
5 s denaturation at 98 C
10 s annealing of 72 C
100 s of extension at 72 C
Final extension: 120 s at 72 C
2. Assemble the Reaction in a PCR Tube on Ice:
Reagent Volume
Q5 master mix (2) 25μL
Genomic DNA from human cell line X μL (2.4μg)
18S forward primer (10μM stock) 2.5μL
18S reverse primer (10μM stock) 2.5μL
Nuclease-free H20 50 (X + 30) μL
Total 50.0μL
3.9.4 Synthesis 1. Assemble the IVT reaction using the HiScribe™ T7 Quick
of Canonical 18S rRNA High Yield RNA Synthesis kit reagents as follows:
from the PCR-Derived DNA
Template
66 Miten Jain et al.
Reagent Volume
Purified DNA from Subheading 3.9.3 X μL (0.5–1μg)
NTP buffer 10.0μL
T7 RNA polymerase mix 2.0μL
RNasin (40 units/μL) or equivalent RNase inhibitor 1μL
Nuclease-free water 20 (X + 13) μL
Total 20.0μL
Reagent Volume
28S containing plasmid (4μg) X μL
10 buffer for XhoI 10.0μL
Xho I (20,000 units/mL) 10.0μL
Nuclease-free H20 100 (X + 20)μL
Total 100.0μL
2. Digest for 2 h at 37 C.
Nanopore Sequencing of Human Nuclear rRNA 67
3.9.6 Synthesis of 28S 1. Assemble the IVT reaction using the HiScribe™ T7 Quick
rRNA from the Plasmid High Yield RNA Synthesis kit reagents in a PCR tube as
Template follows:
68 Miten Jain et al.
Reagent Volume
Purified linearized DNA from Subheading 3.9, step 5 X μL
(1.5–2.0μg)
NTP buffer 20.0μL
T7 RNA polymerase mix 4.0μL
RNasin (40 units/ul) or equivalent RNase inhibitor, 2.0μL
optional
Nuclease-free water 40 (X + 26)μ
L
Total 40.0μL
3.9.7 Purification 1. The IVT products must be purified prior to sequencing. In our
of Canonical Transcripts experience, gel purification is the method that results in the
best read coverage and throughput. This is especially true for
the 28S IVT product. Other options for purification include
phenol/chloroform extraction, followed by ethanol precipita-
tion, and use of spin columns (see Note 11).
2. The general scheme for gel purification involves running the
IVT sample along with a sizing ladder on a denaturing PAGE
or a non-denaturing agarose gel. The gel is pre- or post-stained
with SYBR™ Gold and the appropriately sized RNA (121 nt
for 5S, 155 nt for 5.8S, 1.8 kb for 18S, and 5 kb for 28S) is
identified and excised.
3. Following gel excision, we have had success using the D-tube
Dialyzers MWCO 3.5 kD according to the manufacturer’s
protocol to purify the RNA. This involves electroelution and
ethanol precipitation. Other gel purification kits require differ-
ent steps and may need to be optimized.
4. Following purification determine the concentration of the sam-
ple using Qubit.
Nanopore Sequencing of Human Nuclear rRNA 69
5. Once purified, the 5S, 5.8S, and 18S RNA are ready for
sequencing (Subheading 3.9.9).
6. The purified 28S RNA is next polyadenylated as described
below in Subheading 3.9.8.
2. Run for 1 h at 37 C.
3. Follow the procedure for Agencourt RNAClean XP bead puri-
fication as described in Subheading 3.9.2. Use 0.6 beads
(24μL) and use 70% ethanol for washes.
4. Measure the concentration using a Qubit™ BR RNA Assay kit.
One μg of this material will be used for the library (see Sub-
heading 3.9.10).
3.9.9 Library Preparation Splint annealing follows the procedures described in Subheading
Using Splint Adapters 3.2. Please note that the 28S splint is not needed. The custom
for Canonical 5S, 5.8S, rRNA splint adapter mix listed below is made by combining 1μL
and 18S rRNAs volume from each of the 10μM oligomer splints (5S, 5.8S, 18S).
The following amounts of purified IVT rRNA are recommended:
500 ng of 5S, 500 ng of 5.8S, and 350 ng of 18S.
1. Set up the first ligation as follows:
Reagent Volume
NEBNext quick ligation reaction buffer 3.0μL
(5)
RNA CS (RCS), 110 nM (optional) 0.5μL
IVT rRNAs for 5S, 5.8S, 18S 7.5μL (or 7.0μL if adding
RCS)
Custom rRNA splint adapter mix 3.0μL
T4 DNA Ligase (2000 U/μL) 1.5μL
Total 15.0μL
70 Miten Jain et al.
3.9.10 Library Because the sample is polyadenylated, the standard ONT mRNA
Preparation library protocol (SQK-RNA002) is used. We suggest starting with
for Polyadenylated 1μg of polyadenylated 28S IVT RNA for the library.
Canonical 28S rRNA
1. Set up the first ligation as follows in a 1.5 mL DNA LoBind
tube.
Reagent Volume
NEBNext quick ligation reaction buffer 3.0μL
(5)
RNA CS (RCS), 110 nM (optional) 0.5μL
RNA-1μg polyadenylated 28S IVT rRNA X μL (or X–0.5μL if adding
RCS)
RTA adapter 1.0μL
Nuclease-free H20 Bring to 13.5μL
T4 DNA Ligase (2000 U/μL) 1.5μL
Total 15.0μL
4 Notes
50 -CATCATCATTTAATACGACTCACTATAGNNNNNNNNNNNNTACCTGGTTGATCCTGC-
CAGTAGC-30
References
1. Deamer D, Akeson M, Branton D (2016) insights into structural variants and enables
Three decades of nanopore sequencing. Nat modification analysis. [Link]
Biotechnol 34:518–524 1101/483693
2. Garalde DR, Snell EA, Jachimowicz D et al 7. Simpson JT, Workman RE, Zuzarte PC et al
(2018) Highly parallel direct RNA sequencing (2017) Detecting DNA cytosine methylation
on an array of nanopores. Nat Methods. using nanopore sequencing. Nat Methods.
[Link] [Link]
3. Workman RE, Tang AD, Tang PS et al (2019) 8. Methylation calling — Medaka 1.0.3 docu-
Nanopore native RNA sequencing of a human mentation. [Link]
poly(A) transcriptome. Nat Methods medaka/[Link]. Accessed 10 Jul
16:1297–1305 2020
4. Smith AM, Jain M, Mulroney L et al (2019) 9. Liang X-H, Liu Q, Fournier MJ (2009) Loss of
Reading canonical and modified nucleobases in rRNA modifications in the decoding center of
16S ribosomal RNA using nanopore native the ribosome impairs translation and strongly
RNA sequencing. PLoS One 14:e0216709 delays pre-rRNA processing. RNA
5. Kim D, Lee J-Y, Yang J-S et al (2020) The 15:1716–1728
architecture of SARS-CoV-2 transcriptome. 10. King TH, Liu B, McCully RR, Fournier MJ
Cell 181 e10:914–921 (2003) Ribosome structure and activity are
6. Viehweger A, Krautwurst S, Lamkiewicz K et al altered in cells lacking snoRNPs that form
Direct RNA nanopore sequencing of full- pseudouridines in the peptidyl transferase cen-
length coronavirus genomes provides novel ter. Mol Cell 11:425–435
74 Miten Jain et al.
11. Narla A, Ebert BL (2010) Ribosomopathies: 14. Li H (2018) Minimap2: pairwise alignment for
human disorders of ribosome dysfunction. nucleotide sequences. Bioinformatics. https://
Blood 115:3196–3205 [Link]/10.1093/bioinformatics/bty191
12. Lafontaine DLJ (2015) Noncoding RNAs in 15. Li H, Handsaker B, Wysoker A et al (2009)
eukaryotic ribosome biogenesis and function. The sequence alignment/map format and
Nat Struct Mol Biol 22:11–19 SAMtools. Bioinformatics 25:2078–2079
13. Taoka M, Nobe Y, Yamaki Y et al (2018) Land- 16. Robinson JT, Thorvaldsdóttir H, Winckler W
scape of the complete RNA chemical modifica- et al (2011) Integrative genomics viewer. Nat
tions in the human 80S ribosome. Nucleic Biotechnol 29:24–26
Acids Res 46:9289–9298
Part III
Abstract
Epitranscriptomics is an emerging field where the development of high-throughput analytical technologies
is essential to profile the dynamics of RNA modifications under different conditions. Despite important
advances during the last 10 years, the number of RNA modifications detectable by next-generation
sequencing is restricted to a very limited subset. Here, we describe a highly efficient and fast method called
AlkAniline-Seq to map simultaneously two different RNA modifications: 7-methyl-guanosine (m7G) and
3-methyl-cytosine (m3C) in RNA. Our protocol is based on three subsequent chemical/enzymatic steps
allowing the enrichment of RNA fragments ending at position n + 1 to the modified nucleotide, without
any prior RNA selection. Therefore, AlkAniline-Seq demonstrates an outstanding sensitivity and specificity
for these two RNA modifications. We have validated AlkAniline-Seq using bacterial, yeast, and human total
RNA, and here we present, as an example, a synthetic view of the complete profiling of these RNA
modifications in S. cerevisiae tRNAs.
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
77
78 Virginie Marchand et al.
25S
(Spectrophotomer) 6 (Capillary electrophoresis) tRNA 18S 25S rRNA
5 18S
4
3
2
1
0 tRNA
220
230
240
250
260
270
280
290
300
310
320
330
B. ALKANILINE-SEQ
AlkAniline Treatment 5’ P OH3’ Total RNA from yeast S. cerevisiae
Mild Alkaline Hydrolysis
5’ P
P3’
5’HO
OH 3’ RNA precipitation
Aniline cleavage
5’HO
OH3’
5’HO X 3’
5’ P OH 3’ RNA precipitation
Library preparation and clean-up
3’ Adaptor ligation PCR
P5
RT primer hybridization
P5
P5 BC P7
5’ Adaptor ligation
P5 BC P7
P5 BC P7
First strand cDNA synthesis
Clean up
Ladder (bp)
C. BIOINFORMATIC PIPELINE
Multiplexing and Sequencing Scoring
Normalized cleavage = 6*1000/14 = 428 units Stop-rao = 6/(6+2) = 0.75
Starng reads = 6
5’-end counts
6 Passing reads = 2
Total 5’-ends = 14
P5 N₊₁ BC P7 N₊₁
posions
Bioinformatic workflow
Trimming Alignment Keep only Conversion to BED Count 5’-ends Score calculaƟon
(TrimmomaƟc) (BowƟe 2.0) mapped reads (bedtools) and coverage (awk) and graphs (R)
parameters
TruSeq3-SE.[Link]
-D 15 -R 2 samtools view -h -F 4 -b
LEADING:30
TRAILING:30 -N 0 -L 10
SLIDINGWINDOW:4:15 -i S,1,1.15
MINLEN:8 AVGQUAL:30
Fig. 1 Overview of experimental and analysis steps of AlkAniline-Seq protocol. (a) Human total RNA is
extracted using the standard TRIzol protocol; extraction of total RNAs from bacterial and yeast cells is
Mapping of m7G and m3C in RNA 79
2 Materials
Fig. 1 (continued) performed using the “hot acid phenol” protocol. Quantification and quality of RNA are
assessed by spectrophotometer and RNA integrity is evaluated using capillary electrophoresis. (b) RNAs are
subjected to mild alkaline hydrolysis, generating fragments of about 200–300 nt in length. Fragments are
extensively dephosphorylated to remove all possible 50 - and 30 -phosphate residues from RNA. Afterwards,
RNAs are subjected to aniline cleavage at the abasic sites generated by decomposition of m7G/m3C residues
upon alkaline hydrolysis. Library preparation is performed by direct ligation of pre-adenylated 30 -adapter,
followed by RT primer annealing and ligation of 50 -adapter and RT primer extension. The resulting cDNA is
converted to sequencing library by second-strand DNA synthesis and limited PCR step for barcoding and
inclusion of Illumina P5 and P7 sequences. Quantification of the library is performed using a fluorometer and
the quality is assessed by capillary electrophoresis. (c) Scoring of AlkAniline-Seq signals is done by calculation
of both normalized cleavage (ratio of reads starting at a given RNA position to total number of reads mapped to
this RNA) and stop-ratio, corresponding to the ratio of reads starting at a given position to reads overlapping
it. Normalized cleavage provides exceptional selectivity, while stop-ratio is very sensitive, but captures
numerous false-positive hits. Bioinformatic analysis consists of trimming step to keep adapter-free reads,
bowtie 2.0 end-to-end alignment followed by counting of reads mapped to different positions in RNA and
calculation of AlkAniline-Seq scores
80 Virginie Marchand et al.
Fig. 2 AlkAniline-Seq signals for m7G and m3C residues in yeast S. cerevisiae tRNAs. (a) m7G residues
detected in 10 m7G46-modified yeast tRNAs. Initiator tRNAMeti also has m7G46 residue but is not illustrated
here since it has a low coverage in sequencing. tRNA positions are numbered sequentially from 50 - to 30 -end;
however specific tRNA numbering scheme (with 17a, 20ab, and extra nucleotides in variable loop) is not
Mapping of m7G and m3C in RNA 81
2.1 Total RNA 1. Yeast or bacteria cell culture (10 mL of culture grown to an
Extraction OD600 of 0.7–2).
2.1.1 Yeast and Bacteria 2. RNase-free 1.5 mL microcentrifuge tubes.
Total RNA Extraction by Hot 3. RNase-free water.
Acid Phenol 4. AE buffer: 50 mM NaOAc in water, pH 5.2, 10 mM EDTA.
5. 10% (w/v) SDS.
6. Acid phenol, pH 4.5.
7. Phenol:chloroform:isoamyl alcohol mix ([Link], v/v).
8. Chloroform.
9. 3 M NaOAc, pH 5.2.
10. 96% Ethanol.
11. 80% Ethanol.
12. Dry ice.
13. Refrigerated tabletop centrifuge.
14. Water bath or heating block set to 65 C.
2.1.2 Human Total RNA 1. Human HEK 293 cells (8–10 106 cells grown to 90% con-
Extraction by TRIzol™ fluence in a cell culture dish).
2. 1 PBS (8.1 mM Na2HPO4, 1.47 mM KH2PO4, 137 mM
NaCl, and 2.7 mM KCl).
3. RNase-free 1.5 mL microcentrifuge tubes.
4. RNase-free water.
5. TRIzol™ reagent.
6. Chloroform.
7. Refrigerated tabletop centrifuge.
8. Isopropanol.
9. Glycoblue™ coprecipitant: 15 mg/mL.
10. 75% Ethanol.
11. Cell scraper.
Fig. 2 (continued) respected. Bar plot represents normalized cleavage score for a given RNA species in four
yeast strains (WT—wild-type BY 4741 strain, ΔTRM140—strain deleted for m3C32:tRNA-methyltransferase,
ΔTRM8/ΔTRM82—two strains deleted for genes encoding subunits of heterodimeric m7G46:tRNA-methyl-
transferase). Position of m7G46 is indicated by a gray dot. Some of the yeast tRNA species also contain
adjacent dihydrouridine (D47), purple dot. This residue is also partially cleaved during mild alkaline hydrolysis
(see small peaks at position 47) but becomes visible only in the absence of a major signal corresponding to
m7G. Some weak cleavage signals are also visible in the region 16–20, and correspond to D residues in the
D-loop of tRNA. (b) Two yeast tRNAs containing m3C32 residues (shown as pink dot). Closely related isoforms
of yeast tRNASer (AGA, CGA, and TGA) are collapsed into unique tRNASer(NGA) sequence. tRNA positions are
numbered as in (a)
82 Virginie Marchand et al.
2.4 Library 1. NEBNext® Multiplex Small RNA Library Prep Set for Illu-
Preparation mina® (set 1 or 2) (see Note 1).
2. RNase-free 0.2 mL PCR tubes, strips of 8.
3. Flat PCR caps, strips of 8.
4. Thermal cycler.
2.6 Library 1. Any kind of fluorometer able to quantify DNA library with
Quantification high sensitivity (e.g., Qubit® 2.0 fluorometer).
and Quality 2. Qubit® dsDNA HS Assay kit (0.2–100 ng).
Assessment
3. Thin-walled polypropylene tubes of 500 μL compatible with
the fluorometer (e.g., Qubit® Assay Tube or Axygen®
PCR-05-C tubes).
4. Agilent 2100 Bioanalyzer (Agilent Technologies).
5. Agilent HS DNA kit (quantitative range 5–500 pg/μL).
6. Chip priming station.
7. RNase-free 1.5 mL microcentrifuge tubes.
8. Tabletop centrifuge.
2.8 Bioinformatic 1. Unix (Linux) server (we use Illumina Compute Dell R815
Analysis server).
84 Virginie Marchand et al.
3 Methods
3.1 Total RNA The following protocol details total RNA isolation from yeast/
Extraction bacteria using hot acid phenol and is adapted from [13].
3.1.1 Yeast and Bacteria 1. Transfer yeast/bacteria cell culture in 1.5 mL microcentrifuge
Total RNA Extraction by Hot tubes and pellet cells by centrifugation at 1200 g for 5 min at
Acid Phenol room temperature. Discard the supernatant.
2. Resuspend cells in 1 mL of RNase-free water. Centrifuge for
1 min at full speed at room temperature. Discard the
supernatant.
3. Resuspend the cell pellet in 400 μL of AE buffer.
4. Add 40 μL of 10% SDS and vortex until the pellet is completely
resuspended.
5. Add 440 μL of acid phenol. Vortex.
6. Incubate for 4 min at 65 C and then cool the mixture rapidly
on dry ice for 2–3 min.
7. Centrifuge the samples for 10 min at full speed at room tem-
perature. Carefully transfer the aqueous (upper) phase to a new
1.5 mL microcentrifuge tube.
8. Add 420 μL of phenol:chloroform:IAA, vortex, and centrifuge
for 10 min at full speed at room temperature.
9. Transfer the aqueous phase to a new 1.5 mL centrifuge tube.
Add 400 μL of chloroform. Vortex and centrifuge at full speed
at room temperature for 10 min.
10. Transfer the aqueous phase to a new 1.5 mL centrifuge tube.
Add 40 μL of 3 M NaOAc and 1 mL of 96% ethanol. Place at
80 C for at least 30 min.
11. Centrifuge for 30 min at full speed at 4 C.
12. Discard the supernatant and wash pellet with 500 μL of 80%
ethanol.
13. Centrifuge for 5 min at full speed at 4 C.
Mapping of m7G and m3C in RNA 85
3.1.2 Human Total RNA Isolate total RNA using TRIzol™ following the manufacturer’s
Extraction by TRIzol™ instructions.
1. Wash HEK 293 cells grown in a cell culture dish with 1.5 mL
1 PBS.
2. After PBS removal, add 1 mL of TRIzol™ directly to the cell
culture dish to lyse the cells, scrap the cells, and pipet the lysate
up and down several times to homogenize.
3. Incubate for 5 min at room temperature to get complete RNP
dissociation.
4. Add 200 μL chloroform, vortex, and incubate for 5 min at
room temperature.
5. Centrifuge for 15 min at 12,000 g at room temperature.
6. Transfer the aqueous phase containing RNA to a new 1.5 mL
microcentrifuge tube, add 500 μL of isopropanol and 1 μL of
Glycoblue™, and mix by inverting the tube up and down
several times.
7. Incubate for 10 min at room temperature.
8. Centrifuge for 10 min at 12,000 g at 4 C.
9. Discard the supernatant and wash pellet with 1 mL of 75%
ethanol.
10. Centrifuge for 5 min at 12,000 g at 4 C.
11. Discard the supernatant, and centrifuge again for a short spin.
12. Remove any remaining liquid.
13. Incubate with open lid for 2 min at 37 C or 5 min at room
temperature.
14. Resuspend the pellet with 50 μL of RNase-free water.
15. Quantify human total RNA samples by measuring A260nm
using a UV spectrophotometer (see Subheading 3.2). Check
86 Virginie Marchand et al.
3.2.1 RNA Quantification 1. On a Nanodrop 2000 start screen, select the “Nucleic Acid”
application.
2. After the wavelength verification test, select the type of sample
to measure, in this case “RNA.”
3. Prepare the blank/buffer solution used for sample resuspen-
sion but without any trace of RNA (e.g., RNase-free water).
4. Load 1 μL of the blank solution to the bottom pedestal, lower
the arm, and click on the “Blank” button.
5. Wipe the upper and lower pedestal using a dry wipe, load 1 μL
of one of your samples of interest to the bottom pedestal, lower
the arm, and click “Measure.”
6. Analyze the data obtained for your different RNA samples. For
“pure” RNA, the ratio of A260/A280 should be 2; the ratio of
A260/A230 should be in the range of 1.8–2.2 (see Notes 3
and 4).
3.2.2 RNA Quality 1. Before starting, equilibrate all solutions of the kit at room
Assessment temperature for at least 30 min in the dark. Vortex and spin
down before use.
2. Transfer 550 μL of gel matrix (red-cap vial) into a spin filter
provided in the kit.
3. Centrifuge for 10 min at 1500 g at room temperature.
4. Prepare 65 μL aliquots of the gel and store them at 4 C for a
maximum of 1 month.
5. Prepare the gel-dye mix by mixing 1 μL of RNA dye concen-
trate to a gel aliquot.
6. Centrifuge for 10 min at 13,000 g at room temperature.
7. Dilute your RNA samples for quantification on the Nanodrop
to 3–5 ng/μL with RNase-free water to be within the optimal
range concentration of the assay.
8. Add 1 μL of your diluted RNA samples to 11 different 1.5 mL
tubes already containing 5 μL of RNA marker (green-cap vial)
(see Note 5). Mix by pipetting up and down.
9. Mix 1 μL of the ladder (see Note 6) with 5 μL of RNA marker
(green-cap vial). Mix by pipetting up and down.
Mapping of m7G and m3C in RNA 87
10. Prepare the chip priming station. Adjust the syringe clip to the
highest top position.
11. Load 9 μL of the gel-dye mix in the well marked with a “G”
surrounded by a black circle.
12. Close the chip priming station properly and press the plunger
of the syringe until it is held by the clip.
13. Wait for 30 s and then release the clip.
14. Wait for 5 s until the plunger stops and pull it slowly back to the
1 mL position of the syringe.
15. Open the chip priming station and load 9 μL of the gel-dye mix
in the two other wells marked “G.”
16. Load 9 μL of the conditioning solution (white-cap vial) in the
well marked “CS.”
17. Load 6 μL of the diluted ladder in the well marked with a
ladder.
18. Load 6 μL of the diluted RNA samples in the wells marked
1–11.
19. Inspect the chip and make sure that no liquid spills are present
on the edges of the wells.
20. Insert the chip in the Agilent 2100 Bioanalyzer and close the
lid (see Note 7).
21. Select the following assay “Eukaryote Total RNA Pico series
II” in the 2100 Expert Software screen.
22. Press “Start” to begin the chip to run (see Note 8).
23. After the run, immediately remove the chip and clean the
electrodes with the electrode cleaner filled with 350 μL of
RNase-free water.
24. Analyze the results of the chip (see Fig. 1).
3.3 AlkAniline-Seq 1. Prepare one 1.5 mL tube per sample to be analyzed (“precipi-
tation tube”) containing 10 μL of NaOAc, 1 μL of Glyco-
3.3.1 Alkaline Hydrolysis
blue™, and 1 mL of 96% ethanol for subsequent
precipitation of the sample (store at 20 C until further use).
2. Dilute your RNA samples to a concentration of 10 ng/μL with
RNase-free water.
3. To individual PCR tubes, add 10 μL of each of your diluted
RNA samples, and keep on ice until further use.
4. Add 10 μL of bicarbonate buffer and mix by pipetting up
and down.
5. Incubate in a thermal cycler preheated at 95 C. Start a timer
and incubate for 5 min (see Note 9).
6. Proceed with the next sample every 30 s.
88 Virginie Marchand et al.
3.4 Library Upon opening the NEBNext® Multiplex Small RNA Library Prep
Preparation Set for Illumina®, resuspend 50 SR adapter (yellow-cap vial) in 120
μL of RNase-free water and store at 80 C.
1. Mix 6 μL of RNA sample with 1 μL of 30 SR adapter (green-cap
vial) (previously diluted ½ in RNase-free water) in a PCR tube.
2. Incubate for 2 min at 70 C in a preheated thermal cycler.
Transfer immediately to ice.
3. Add 10 μL of 30 ligation buffer (green-cap vial) and 3 μL of 30
ligation enzyme (green-cap vial).
4. Incubate for 1 h at 25 C in a thermal cycler.
5. Add 4.5 μL of RNase-free water and 1 μL of SR RT primer
(pink-cap vial) (previously diluted ½ in RNase-free water).
6. Incubate for 5 min at 75 C, 15 min at 37 C, and 15 min at
25 C.
7. Within the last 15 min of incubation, add 1.1*n (n ¼ number
of samples) μL of the 50 SR adapter (yellow-cap vial) (previously
diluted ½ in RNase-free water) in an individual PCR tube.
8. Denature the 50 SR adapter in a thermal cycler for 2 min at
70 C and immediately place the tube on ice (see Note 10).
9. Add 1 μL of 50 SR adapter (previously denatured step 8), 1 μL
of 50 ligation reaction buffer (yellow-cap vial), and 2.5 μL of
ligase enzyme mix (yellow-cap vial).
10. Incubate for 1 h at 25 C in a thermal cycler.
11. Add the following components to the adapter-ligated RNA
mix from the previous step, 8 μL of first-strand synthesis reac-
tion buffer (red-cap vial), 1 μL of murine RNase inhibitor
(red-cap vial), and 1 μL of ProtoScript II reverse transcriptase
(red-cap vial), and mix well by pipetting up and down.
90 Virginie Marchand et al.
3.5 Purification Using the GeneJET PCR Purification Kit, carry out all procedures
of the Library at room temperature.
1. Transfer the PCR mix to a 1.5 mL tube and add 100 μL of
binding buffer. Mix thoroughly.
2. Transfer the solution to the purification column. Centrifuge at
full speed for 30 s. Discard the flow-through.
3. Add 700 μL of wash buffer to the column and centrifuge at full
speed for 30 s. Discard the flow-through.
4. Centrifuge the empty column for an additional 1 min.
5. Transfer the column to a clean 1.5 mL DNA low-binding tube.
Add 30 μL of elution buffer to the center of the column
membrane and centrifuge at full speed for 1 min.
6. Store the purified library at 20 C until further use.
3.6 Library 1. Before starting, incubate all solutions of the Qubit dsDNA HS
Quantification assay kit at room temperature for at least 30 min. The kit
provides the concentrated assay reagent, dilution buffer, and
pre-diluted standards.
2. Prepare the dye working solution by diluting the concentrated
assay reagent 1:200 in dilution buffer. Prepare 200 μL of
working solution for each sample and two additional standards.
3. Prepare the two standards annotated “C” and “D” by mixing
10 μL of standard with 190 μL of working solution.
4. Add working solution to 1 μL of library sample to obtain 200
μL in total.
5. Vortex the tubes for 2 s and incubate for 2 min at room
temperature.
6. Insert the tubes into the Qubit® 2.0 Fluorometer and proceed
with measurements: on the home screen of the Qubit® 2.0
Fluorometer, choose the type of assay (e.g., “HS DNA”) for
which you want to perform a new calibration.
7. Press “Yes” to read new standards.
Mapping of m7G and m3C in RNA 91
3.7 Library Quality 1. Before starting the experiments, incubate all solutions of the
Assessment Agilent High Sensitivity DNA kit at room temperature for at
least 30 min in the dark. Vortex them and spin them down
before use.
2. Add 15 μL of high-sensitivity DNA dye concentrate (blue-cap
vial) into a high-sensitivity DNA gel matrix vial (red-cap vial)
(see Note 14).
3. Vortex for 10 s and transfer the gel-dye mix to the center of the
spin filter.
4. Centrifuge for 10 min at 2240 g.
5. Add 1 μL of each of your library to 11 different tubes of 1.5 mL
already containing 5 μL of RNA marker (green-cap vial). Mix
by pipetting up and down.
6. Mix 1 μL of the ladder (yellow-cap vial) with 5 μL of high-
sensitivity DNA marker (green-cap vial). Mix by pipetting up
and down.
7. Prepare the chip priming station. Adjust the syringe clip to the
lowest top position.
8. Load 9 μL of the gel-dye mix in the well marked with a “G”
surrounded by a black circle.
9. Close the chip priming station properly and press the plunger
of the syringe until it is held by the clip.
10. Wait for 1 min and then release the clip.
11. Wait for 5 s until the plunger stops and pull it slowly back to the
1 mL position of the syringe.
12. Open the chip priming station and load 9 μL of the gel-dye mix
in the three other wells marked “G.”
13. Load 6 μL of the diluted ladder in the well marked with a
ladder.
14. Load 6 μL of the diluted library samples in the wells labeled
1–11.
15. Insert the chip in the Agilent 2100 Bioanalyzer, close the lid,
and select the following assay “High Sensitivity DNA” in the
2100 Expert Software screen.
92 Virginie Marchand et al.
3.9 Bioinformatic 1. Trim adapter sequences of raw reads (FastQ files) using Trim-
Analysis momatic with the following parameters: java -jar
[Link] SE -phred33 [Link] out-
[Link] ILLUMINACLIP :TruSeq3-S[Link] LEAD-
ING:30 TRAILING:30 SLIDINGWINDOW:4:15 AVGQUAL:30
MINLEN:17 (see Note 15).
2. Align the trimmed reads to the appropriate reference sequence
(E. coli or yeast rRNA/tRNA dataset, described in [14]) using
bowtie2 with the following parameters: bowtie2 -D 15 -R
2 -N 0
-L 10 -i S,1,1.15 -x <bt2-idx> -U <r> --S. The use
of soft trimming is not recommended.
3. Mapped reads are extracted from the *.sam file by RNA ID and
converted to *.bed format using bedtools v2.25.0.
4. Count the 50 -ends in the produced *.bed file using Unix awk
command: awk ’{print $2}’ <*.bed> | sort | uniq -c |
awk ’{print $3,$2,$1,$4}’ | sort --n.
5. Calculate normalized cleavage and stop-ratio scores for each
position. Normalized cleavage is calculated as a proportion of
reads starting at a given position divided by the total number of
reads mapped to a given RNA species (1000) and stop-ratio
corresponds to the number of reads starting at a given position
divided by the number of reads covering this position in RNA
(Fig. 1). Normalized cleavage varies from 10–25 for back-
ground values to 1000 if all reads in RNA start at one single
position. Stop-ratio varies from 0 to 1; values >0.75 generally
correspond to m7G/m3C signals in stoichiometrically modi-
fied RNA (Fig. 2).
Mapping of m7G and m3C in RNA 93
4 Notes
1. The kit NEBNext® Multiplex Small RNA Library Prep Set for
Illumina® (set 1) includes a set of 12 barcoding primers (num-
bered 1–12) that will be used for multiplexing reactions during
PCR amplification. There is also a version set 2 with primers
(numbers 13–24). If you do not need these barcoding primers,
you may order a similar kit without the primers and use any
other source of barcoding primers (Illumina or NEB).
2. The typical amount obtained with 1 mL of a haploid wild-type
yeast culture (BY4741 or BY4742) or bacteria culture (DH5α)
grown to an OD600 of 1 is about 15–30 μg.
3. If your RNA sample is diluted with RNase-free water instead of
10 mM Tris-EDTA (TE) pH 8.0, the ratio of A260/A280 may
be below 2.0 due to the lower pH of water [15]. A ratio of
A260/A280 of 1.8 for samples diluted in RNase-free water is
considered “pure” for RNA.
4. If your RNA sample is contaminated by phenol or chaotropic
salts (e.g., guanidinium thiocyanate used in TRIzol™ extrac-
tion or other protocols), this will result in a ratio of A260/
A230 below 1.8. Another round of phenol:choroform:isoamyl
alcohol (PCA) extraction and two successive steps of chloro-
form extraction followed by ethanol precipitation are recom-
mended in this case before alkaline digestion.
5. In case you are working with less than 11 samples, add 1 μL of
RNase-free water to the empty wells.
6. The ladder loaded in the Pico RNA chip is provided in a
separate package and should be prepared before the beginning
of the experiment: spin down the tube and transfer 10 μL to a
RNase-free tube. Heat for 2 min at 70 C. Cool down on ice
and add 90 μL of RNase-free water. Prepare 5 μL aliquots using
the Safe-Lock PCR tubes provided in the kit and store at
70 C. Before use, thaw one tube and keep it on ice. The
ladder is quite stable at 70 C and may be used for at least
4 months.
7. RNase contamination problems of the Bioanalyzer electrodes
are very frequent and will affect the RNA integrity number of
your samples. Therefore, if the Agilent 2100 Bioanalyzer is also
frequently used to run DNA chips, it is strongly recommended
to use a dedicated electrode cartridge only for RNA assays. In
addition, we recommend for each chip to load an internal RNA
control (total RNA preparation with a known RIN >9). If you
encounter contamination problems, soak the electrode car-
tridge into an RNaseZap® decontamination solution for at
94 Virginie Marchand et al.
Acknowledgments
References
1. Hussain S, Aleksic J, Blanco S, Dietmann S, 2. Novoa EM, Mason CE, Mattick JS (2017)
Frye M (2013) Characterizing Charting the unknown epitranscriptome. Nat
5-methylcytosine in the mammalian epitran- Rev Mol Cell Biol 18:339–340
scriptome. Genome Biol 14:215
Mapping of m7G and m3C in RNA 95
3. Schwartz S (2016) Cracking the epitranscrip- 10. Zueva VS, Mankin AS, Bogdanov AA, Baratova
tome. RNA N Y N 22:169–174 LA (1985) Specific fragmentation of tRNA and
4. Helm M, Motorin Y (2017) Detecting RNA rRNA at a 7-methylguanine residue in the pres-
modifications in the epitranscriptome: predict ence of methylated carrier RNA. Eur J Bio-
and validate. Nat Rev Genet 18:275–291 chem 146:679–687
5. Molinie B, Wang J, Lim KS, Hillebrand R, Lu 11. Wintermeyer W, Zachau HG (1975) Tertiary
Z-X, Van Wittenberghe N, Howard BD, structure interactions of 7-methylguanosine in
Daneshvar K, Mullen AC, Dedon P et al yeast tRNA Phe as studied by borohydride
(2016) m(6)A-LAIC-seq reveals the census reduction. FEBS Lett 58:306–309
and complexity of the m(6)A epitranscriptome. 12. Marchand V, Ayadi L, Ernst FGM, Hertler J,
Nat Methods 13:692–698 Bourguignon-Igel V, Galvanin A, Kotter A,
6. Schwartz S, Motorin Y (2017) Next- Helm M, Lafontaine DLJ, Motorin Y (2018)
generation sequencing technologies for detec- AlkAniline-Seq: profiling of m7G and m3C
tion of modified nucleotides in RNAs. RNA RNA modifications at single nucleotide resolu-
Biol 14:1124–1137 tion. Angew Chem Int Ed Engl 57
7. Meyer KD, Jaffrey SR (2014) The dynamic (51):16785–16790
epitranscriptome: N6-methyladenosine and 13. Schmitt ME, Brown TA, Trumpower BL
gene expression control. Nat Rev Mol Cell (1990) A rapid and simple method for prepara-
Biol 15:313–326 tion of RNA from Saccharomyces cerevisiae.
8. Li X, Peng J, Yi C (2017) Transcriptome-wide Nucleic Acids Res 18:3091–3092
mapping of N1-methyladenosine methylome. 14. Marchand V, Pichot F, Thüring K, Ayadi L,
Methods Mol Biol Clifton NJ 1562:245–255 Freund I, Dalpke A, Helm M, Motorin Y
9. Schwartz S, Bernstein DA, Mumbach MR, (2017) Next-generation sequencing-based
Jovanovic M, Herbst RH, León-Ricardo BX, ribomethseq protocol for analysis of tRNA
Engreitz JM, Guttman M, Satija R, Lander ES 2’-O-methylation. Biomol Ther 7(1):13
et al (2014) Transcriptome-wide mapping 15. Wilfinger WW, Mackey K, Chomczynski P
reveals widespread dynamic-regulated pseu- (1997) Effect of pH and ionic strength on the
douridylation of ncRNA and mRNA. Cell spectrophotometric assessment of nucleic acid
159:148–162 purity. BioTechniques 22:474–476. 478–481
Chapter 6
Abstract
m7G-seq detects internal 7-methylguanosine (m7G) sites within mRNAs and noncoding RNAs by mis-
incorporation signatures. A chemical-assisted sequencing approach selectively converts internal m7G sites
into abasic sites, triggering misincorporation at these sites in the presence of a specific reverse transcriptase.
The further enrichment of m7G-induced abasic sites by biotin pull-down reveals hundreds of internal m7G
sites in human mRNA. The misincorporation ratio before pull-down enrichment can be used for estimating
the methylation fraction of some highly methylated m7G sites.
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
97
98 Li-Sheng Zhang et al.
H+, pH=4.5
X
CH3 CH3
O O
N HN N OH OH
HN O N
H2 N N N NaBH4 H2 N N N H+, pH=4.5 O
Biotin O
NH
O O OH OH
O O
Reduction Depurination Hydrazidation biotin
O O
OH OH
O O
Biotinylated AP
m 7G Reduced m7G Abasic site (AP site) site
Fig. 1 Schematic diagram displaying the chemical reactivity of positively charged m7G under NaBH4 reduction,
depurination, and biotin labeling reactions in m7G-seq. Only the reduced m7G can further generate abasic sites
and then biotinylated AP sites in the presence of biotin hydrazide under acidic conditions. Biotinylated AP sites,
before or after pull-down, induce misincorporation when performing reverse transcription using HIV RT
2 Materials
2.2 mRNA Decapping 1. 10 Decapping Reaction Buffer and Tobacco Decapping Plus
2 enzyme.
2. 20 U/μL SUPERaselIn™ RNase Inhibitor.
3 Methods
3.1 Preparation of 1. Starting from one 15 cm plate for cells of interest, wash cells
Fragmented mRNA once with 5 mL ice-cold DPBS buffer. Isolate cellular total
RNA using TRIzol reagent following the manufacturer’s pro-
tocol and using isopropanol precipitation.
2. With the purified cellular total RNA, extract mRNA by two
rounds of polyA+ purification with Dynabeads mRNA
DIRECT kit following the manufacturer’s protocol. mRNA
concentration is measured using Qubit™ RNA HS Assay Kit
with a Qubit 2.0 fluorometer.
3. Starting with 10μg of human mRNA (Step 2 above), dissolve
the mRNA in 18μL RNase-free water followed by adding 2μL
10 Fragmentation Buffer. Heat the mixture at 70 C for
15 min. mRNA will fragmented into 50–100 nt (see Note 1).
4. Fragmented mRNA is purified with Oligo Clean & Concentra-
tor and eluted with RNase-free water.
3.2 mRNA Decapping 1. Using a maximum of 6μg fragmented mRNA dissolved in 34μL
RNase-free water, add 5μL 10 Decapping Reaction Buffer,
1μL 50 mM MnCl2, and 2μL SUPERaselIn™ RNase Inhibi-
tor. Mix well and then add 8μL decapping enzyme. Mix well
and incubate the reaction at 37 C for 2 h.
2. Decapped mRNA is purified using the Oligo Clean & Concen-
trator and eluted with 40μL RNase-free water.
3.3 End Repair 1. After decapping the fragmented mRNA, 40μL RNA is mixed
and 30 -Adapter with 5μL 10 T4 Polynucleotide Kinase Reaction Buffer. Mix
Ligation well and add 5μL T4 PNK. Mix well and incubate the mixture
at 37 C for 1 h (see Note 2).
Detection of Internal N7-Methylguanosine in mRNA using m7G-seq 101
the bead suspension with 20μL RNA (Step 4) and then incu-
bate at 4 C for 20 min.
6. After the biotin pull-down, wash beads five times with 1 IP
wash buffer and proceed to proteinase K digestion by adding
45μL 1 proteinase K digestion buffer and 5μL proteinase K to
the beads. Mix well and incubate at 55 C for 30 min under
high-speed shaking.
7. The flow-through is saved and RNA recovered with the RNA
Clean and Concentrator. Elute RNA in 12μL RNase-free
water. Save as “pull-down.”
3.5 Reverse 1. The 30 -ligated RNA (as “input”), RNA before pull-down assay
Transcription (as “before pull-down”), and eluted RNA after pull-down
(as “pull-down”) are subjected to reverse transcription. RNAs
are dissolved in 12μL RNase-free water and incubated with 1μL
of 2.0μM RT primer at 65 C for 2 min, followed by moving
immediately onto ice.
2. Add 2μL 10 mM dNTPs, 2μL 10 AMV Reverse Transcriptase
Reaction Buffer, and 1μL RNaseOUT recombinant RNase
inhibitor to the 13μL RNA-primer mixture. Mix well and add
2μL recombinant HIV reverse transcriptase. Mix well and incu-
bate the reaction at 37 C for 1.5 h (see Note 8).
3. Add 1μL RNase H to the mixture and incubate at 37 C for
20 min.
4. cDNAs is purified with the Oligo Clean & Concentrator and
eluted with 7μL RNase-free water.
3.6 cDNA 30 -Ligation 1. The purified cDNA is then subject to cDNA 30 -adaptor ligation
and PCR Amplification [14]. The cDNA is first denatured with 1μL of 50μM cDNA
30 -linker at 75 C for 2 min, followed by transferring immedi-
ately to ice.
2. Add 3μL 10 T4 RNA Ligase Reaction Buffer, 15μL 50%
PEG8000, and 3μL 10 mM ATP to the 8μL cDNA-adapter
mixture. Mix well and add 1μL of T4 RNA ligase 1. Mix well
and incubate the reaction at 25 C for 12 h (see Note 9).
3. 30 -Ligated cDNA is purified with the Oligo Clean & Concen-
trator and eluted with 20μL RNase-free water.
4. The library is then PCR amplified with the universal primer and
indexed primers using the NEBNext Multiplex Oligos for Illu-
mina. All libraries are sequenced on an Illumina NextSeq
500 with single-end 80 bp read length.
3.7 Data Processing Proceed with data processing and analysis to identify m7G-seq-
and Analysis induced mutations as previously described [13].
Detection of Internal N7-Methylguanosine in mRNA using m7G-seq 103
4 Notes
Acknowledgments
References
1. Cowling VH (2009) Regulation of mRNA cap the biogenesis of the 40S ribosomal subunits in
methylation. Biochem J 425:295–302 mammalian cells. PLoS One 8:e75686
2. Furuichi Y (2015) Discovery of m7G-cap in 10. Shaheen R, Abdel-Salam GMH, Guy MP,
eukaryotic mRNAs. Proc Jpn Acad Ser B Phys Alomar R, Abdel-Hamid MS, Afifi HH, Ismail
Biol Sci 91:394–409 SI, Emam BA, Phizicky EM, Alkuraya FS
3. Murthy KG, Park P, Manley JL (1991) A (2015) Mutation in WDR4 impairs tRNA
nuclear micrococcal-sensitive, ATP-dependent m7G46 methylation and causes a distinct form
exoribonuclease degrades uncapped but not of microcephalic primordial dwarfism. Genome
capped RNA substrates. Nucleic Acids Res Biol 16:210
19:2685–2692 11. Wintermeyer W, Zachau HG (1970) A specific
4. Muthukrishnan S, Both GW, Furuichi Y, Shat- chemical chain scission of tRNA at
kin AJ (1975) 50 -terminal 7-methylguanosine 7-methylguanosine. FEBS Lett 11:160–164
in eukaryotic mRNA is required for translation. 12. Kupfer PA, Leumann CJ (2005) RNA abasic
Nature 255:33–37 sites: preparation and trans-lesion synthesis by
5. Guy MP, Phizicky EM (2014) Two-subunit HIV-1 reverse transcriptase. Chembiochem
enzymes involved in eukaryotic post- 6:1970–1973
transcriptional tRNA modification. RNA Biol 13. Zhang LS, Liu C, Ma H, Dai Q, Sun HL,
11:1608–1618 Luo G, Zhang Z, Zhang L, Hu L, Dong X,
6. Sloan KE, Warda AS, Sharma S, Entian KD, He C (2019) Transcriptome-wide mapping of
Lafontaine DLJ, Bohnsack MT (2017) Tuning internal N7-methylguanosine methylome in
the ribosome: the influence of rRNA modifica- mammalian mRNA. Mol Cell 74:1304–1316
tion on eukaryotic ribosome biogenesis and 14. Li X, Xiong X, Zhang M, Wang K, Chen Y,
function. RNA Biol 14:1138–1152 Zhou J, Mao Y, Lv J, Yi D, Chen XW, Yi C
7. Leulliot N, Chaillet M, Durand D, Ulryck N, (2017) Base-resolution mapping reveals dis-
Blondeau K, van Tilbeurgh H (2008) Struc- tinct m1A methylome in nuclear- and
ture of the yeast tRNA m7G methylation com- mitochondrial-encoded transcripts. Mol Cell
plex. Structure 16:52–61 68:993–1005
8. Haag S, Kretschmer J, Bohnsack MT (2015) 15. Lin S, Liu Q, Lelyveld VS, Choe J, Szostak JW,
WBSCR22/Merm1 is required for late nuclear Gregory RI (2018) Mettl1/Wdr4-mediated
pre-ribosomal RNA processing and mediates m7G tRNA methylome is required for normal
N7-methylation of G1639 in human 18S mRNA translation and embryonic stem cell
rRNA. RNA 21:180–187 self-renewal and differentiation. Mol Cell
9. Õunap K, Kasper L, Kurg A, Kurg R (2013) 71:244–255
The human WBSCR22 protein is involved in
Chapter 7
Abstract
The family of radical SAM RNA-methylating enzymes comprises a large group of proteins that contains
only a few functionally characterized members. Several enzymes in this family have been implicated in the
regulation of translation and antibiotic susceptibility, emphasizing their significance in bacterial physiology
and their relevance to human health. While few characterized enzymes have been shown to modify diverse
RNA substrates, highlighting potentially broad substrate scope within the family, many enzymes in this class
have no known substrates. The precise knowledge of RNA substrates and modification sites for unchar-
acterized family members is important for unraveling their biological function. Here, we describe a strategy
for substrate identification that takes advantage of mechanism-based cross-linking between the enzyme and
its RNA substrates, which we named individual-nucleotide-resolution cross-linking and immunoprecipita-
tion combined with mutational profiling with sequencing (miCLIP-MaPseq). Identification of the position
of the modification site is achieved using thermostable group II intron reverse transcriptase (TGIRT),
which introduces a mismatch at the site of the cross-link.
Key words RNA methylation, Radical SAM, Substrate identification, Methyl adenosine, RlmN, Cfr,
TGIRT
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
105
106 Vanja Stojković et al.
Fig. 1 Mechanistic scheme for RlmN-mediated methylation of RNA showing key steps. The stable covalent
intermediate trapped by C118A mutation is shown
2 Materials
Fig. 2 Schematic representation of library preparation strategy for identification of substrates and methylation
sites of RlmN. Red bars represent the fraction of mismatches at a specific nucleotide on substrate RNA
5. 80% Ethanol.
6. Novex 10% TBE-urea precast gel.
7. 1 TBE running buffer: Dilute 10 TBE running buffer to
1 using DEPC-treated water.
8. 2 RNA loading dye.
9. Low-range ssRNA ladder.
10. SYBR Gold Nucleic Acid Gel Stain.
11. 18G 1 ½ syringe needle.
12. Costar SpinX column.
13. Nuclease-free water.
14. RNase-free non-stick 0.5 mL tubes and RNase-free 1.5 mL
tubes.
15. 100% Ethanol.
110 Vanja Stojković et al.
3 Methods
3.2 Lysis and DNase 1. Thaw ~1.5 g of cells and resuspend them in 4 mL of cold lysis
Treatment buffer. Add 40 μL of 100 mM PMSF.
2. While keeping the cells on ice, sonicate the cells using the
microtip on a power setting 3 and duty cycle 50%, for three
40-s pulses with 1-min breaks between pulses. The probe
should be positioned approximately 0.5 cm from the bottom
of the tube and should not be touching the tube sides in order
to avoid foaming.
3. Divide 4 mL sample among five 1.5 mL Eppendorf tubes. Add
18 μL RQ1 RNase-free DNase to each tube and incubate at
37 C for 15 min. Spin down at 19,722 g (Sorvall Legend
Micro 21R) for 10 min at 4 C. Without disturbing the pellet
remove the supernatant to a new 1.5 mL tube. If not immedi-
ately proceeding with the next step, store the samples at 20 C.
3. Wash the resin once with 1 mL glycine buffer (see Note 2).
4. Wash the resin three times with TBS buffer. For each wash, the
process is the same as in Step 2.
3.3.3 Elution 1. Centrifuge the samples at 8000 g, for 1 min at 4 C. Let the
of FLAG-Tagged Protein resin settle for 1 min. Remove the supernatant.
2. Wash the resin three times with 500 μL of stringent TBS wash
buffer.
3. Add 75 μL of IP dilution solution into each sample. Gently
resuspend and rotate samples for at least 1 h at 4 C (see
Note 3).
4. Centrifuge the samples at 8000 g, for 1 min at 4 C. Let the
resin settle for 2 min.
5. Carefully transfer the supernatant to a new Eppendorf tube.
Put both resin and supernatant on ice. Store resin without
glycerol at 4 C. Store supernatant at 20 C.
3.3.4 Recycling 1. Resuspend used resin in 500 μL of TBS buffer. Centrifuge the
of the Resin resin at 8000 g, for 1 min at 4 C. Remove the supernatant.
2. Wash the resin three times with 1 mL of glycine buffer. Centri-
fuge the resin at 8000 g, for 1 min at 4 C. Let the resin settle
for 1 min.
3. Wash the resin five times with 1 mL of resin recycle solution.
Centrifuge the resin at 8000 g, for 1 min at 4 C. Store the
resin at 20 C.
3.5 Gel Purification 1. Set up the gel apparatus and pre-run the 10% TBE-urea gel at
and RNA Extraction 180 V for at least 20 min in 1 TBE running buffer.
2. Add 25 μL of 2 RNA loading dye to 25 μL of sample. Prepare
molecular weight marker solution by mixing 1 μL of ssRNA
low-range ladder to 9 μL of nuclease-free water, followed by
10 μL of 2 RNA loading dye. Heat the samples at 92 C for
4 min. In two wells load 20 μL of sample and in one well 10 μL.
Load the marker in the last lane (see Note 6).
3. Run the gel at 180 V until the lower (dark blue) dye is close to
the bottom, approximately 70 min. Incubate the gel with
50 mL of TBE buffer containing 5 μL SYBR-Gold dye for
5 min. Wash the gel twice with nuclease-free water.
4. Visualize and record the gel under UV light.
5. Prepare 0.5 mL RNase-free non-stick tubes by piercing a hole
in the bottom using an 18G syringe needle. Cut the gel on
transilluminator as indicated in Fig. 3. Put gel pieces into
0.5 mL tubes and then place the tubes into a 1.5 mL collection
tube. Centrifuge at 20,000 g, for 3 min at 4 C. Remove
0.5 mL tube and add 300 μL nuclease-free water.
6. Shake samples on the thermomixer at 157 g for 10 min at
68 C, and then freeze them on dry ice for 10 min. Thaw
samples at room temperature for 10 min and then incubate
on thermomixer at 157 g for 10 min at 68 C.
7. Cut the tips of the P1000 barrier tips, transfer the sample (with
gel pieces) onto a Costar SpinX column, and spin at 20,000 g
for 3 min at 4 C.
8. Add to each tube 2 μL of GlycoBlue, 33 μL of 3 M sodium
acetate pH 5.5, and 900 μL of 100% ethanol. Vortex to mix.
Put at 20 C overnight. Next morning precipitate RNA as
described in Subheading 3.4.
3.6 RNA Fragment RNAs longer than 300 nt to 50–200 nt long fragments
Fragmentation using the following protocol:
1. Resuspend RNA obtained in Subheading 3.5 in 11 μL of
nuclease-free water. Transfer to PCR tubes and place in the
thermocycler.
2. Heat the samples for 2 min at 95 C to denature RNA.
3. Add 1 μL of 10 Fragmentation Reagent mix. If you are
dealing with larger number of tubes, keep the tubes on ice.
This will ensure that most of the RNA stays denatured. Keeping
samples at the room temperature will allow for the slow refold-
ing of RNA.
114 Vanja Stojković et al.
3.7.6 qPCR 1. For the quantification of the libraries we use KAPA library
Quantification quantification kit. If the kit is being used for the first time,
add 1 mL of 10 Primer Premix to the bottle of 2 KAPA
SYBR FAST qPCR Master Mix (5 mL) and mix by vortexing.
Aliquot this solution and store at 20 C.
2. Determine the total number of reactions that will be per-
formed. Usually we run six DNA standards in triplicate and
each library dilution in duplicate. Using a NanoDrop, estimate
the concentration of each library and determine which dilu-
tions to prepare to stay within the dynamic range of the assay. A
1:5000 and 1:10,000 dilution usually fall around the midpoint
of the assay standards.
3. Prepare 1:5, 1:50, 1:5000, and 1:10,000 library dilutions in
10 mM Tris pH 8 buffer.
4. For each reaction, prepare the following in a 96-well PCR
plate: 6 μL of Master Mix containing primers, and 4 μL of
either DNA standard or specific library dilution. Seal the plate
with optical adhesive film.
5. Run the plate with the following program in the qPCR
machine: 95 C for 5 min, and 35 cycles of 95 C for 30 s,
and 60 C for 45 s.
6. Use the KAPA analysis template to calculate slope and intercept
of the standard curve, to convert the average Cq score for each
library dilution to pM, to calculate the average size-adjusted
concentration (in pM) for each dilution, and to calculate the
size-adjusted concentration for the original undiluted library.
7. Prepare 15 μL of 10 nM library solution, containing up to
20 libraries. Store individual libraries in RNase-free low-reten-
tion Eppendorf tubes at 20 C (see Note 11).
8. Check the quality of the library on Bioanalyzer prior to sub-
mitting sample for sequencing on an Illumina HiSeq4000 or
similar (see Note 12).
3.8.2 Enrichment 1. Determine the raw counts per gene by using HTSeq-count
Analysis of Reads script, which is available through Galaxy web platform
[32]. Select intersection-nonempty mode to handle reads over-
lapping more than one feature. Summarize counts from
regions A-D per replicate. Use this file to perform the enrich-
ment analysis.
2. For enrichment analysis of reads mapped to any set of genes use
DESeq2 module. In DESeq2 specify the factor levels that will
be analyzed (e.g., sample vs. control), and select all replicates
belonging to a specific factor level. As an input data use sum-
marized HTSeq-count data as described above. Use parametric
fitting and leave on the following options: outliers replace-
ment, outliers filtering, and independent filtering. For the
control samples, we generated a library from the rRNA-
depleted total RNA isolated from E. coli BW25113 strain (see
Note 15).
3.8.3 Analysis of Stop 1. Download Integrated Genomic Viewer (IGV) [33]. Open
Sites and Mismatches E. coli BW25113 genome file. Open all BAM files and their
corresponding BAM_index files.
2. Select the gene of interest and determine the percent of mis-
matches for a specific nucleotide by cumulative analysis of all
biological replicates (Fig. 4).
3. To determine the 50 end of the reads (stop sites) use script
“make_wiggle” to convert sorted and indexed BAM files to
wiggle files. This script was developed by the Weissman lab at
UCSF and is readily available through Plastid [36]. The results
can be readily visualized with IGV.
4 Notes
Acknowledgments
References
1. Dominissini D, Moshitch-Moshkovitz S, 4. Carlile TM, Rojas-Duran MF, Zinshteyn B,
Schwartz S, Salmon-Divon M, Ungar L, Shin H, Bartoli KM, Gilbert WV (2014) Pseu-
Osenberg S, Cesarkas K, Jacob-Hirsch J, douridine profiling reveals regulated mRNA
Amariglio N, Kupiec M, Sorek R, Rechavi G pseudouridylation in yeast and human cells.
(2012) Topology of the human and mouse Nature 515(7525):143–146. [Link]
m6A RNA methylomes revealed by m6A-seq. org/10.1038/nature13802
Nature 485(7397):201–206. [Link] 5. Schwartz S, Agarwala SD, Mumbach MR,
10.1038/nature11112 Jovanovic M, Mertins P, Shishkin A,
2. Meyer KD, Saletore Y, Zumbo P, Elemento O, Tabach Y, Mikkelsen TS, Satija R, Ruvkun G,
Mason CE, Jaffrey SR (2012) Comprehensive Carr SA, Lander ES, Fink GR, Regev A (2013)
analysis of mRNA methylation reveals enrich- High-resolution mapping reveals a conserved,
ment in 3’ UTRs and near stop codons. Cell widespread, dynamic mRNA methylation pro-
149(7):1635–1646. [Link] gram in yeast meiosis. Cell 155(6):1409–1421.
1016/[Link].2012.05.003 [Link]
3. Khoddami V, Cairns BR (2013) Identification 6. Delatte B, Wang F, Ngoc LV, Collignon E,
of direct targets and modified bases of RNA Bonvin E, Deplus R, Calonne E, Hassabi B,
cytosine methyltransferases. Nat Biotechnol Putmans P, Awe S, Wetzel C, Kreher J,
31(5):458–464. [Link] Soin R, Creppe C, Limbach PA, Gueydan C,
nbt.2566 Kruys V, Brehm A, Minakhina S, Defrance M,
Steward R, Fuks F (2016) RNA biochemistry.
Identifying Modification Sites in RNA by miCLIP-MaPseq 121
Transcriptome-wide distribution and function the catalytic mechanism of the radical S-adeno-
of RNA hydroxymethylcytosine. Science 351 syl-L-methionine methyl synthase RlmN
(6270):282–285. [Link] trapped by mutagenesis. J Am Chem Soc 134
science.aac5253 (43):18074–18081. [Link]
7. Li X, Zhu P, Ma S, Song J, Bai J, Sun F, Yi C 1021/ja307855d
(2015) Chemical pulldown reveals dynamic 17. Grove TL, Benner JS, Radle MI, Ahlum JH,
pseudouridylation of the mammalian transcrip- Landgraf BJ, Krebs C, Booker SJ (2011) A
tome. Nat Chem Biol 11(8):592–597. https:// radically different mechanism for S-
[Link]/10.1038/nchembio.1836 adenosylmethionine-dependent methyltrans-
8. Lovejoy AF, Riordan DP, Brown PO (2014) ferases. Science 332(6029):604–607. https://
Transcriptome-wide mapping of pseudouri- [Link]/10.1126/science.1200877
dines: pseudouridine synthases modify specific 18. Grove TL, Livada J, Schwalm EL, Green MT,
mRNAs in S. cerevisiae. PLoS One 9(10): Booker SJ, Silakov A (2013) A substrate radical
e110799. [Link] intermediate in catalysis by the antibiotic resis-
pone.0110799 tance protein Cfr. Nat Chem Biol 9
9. Linder B, Grozhik AV, Olarerin-George AO, (7):422–427. [Link]
Meydan C, Mason CE, Jaffrey SR (2015) nchembio.1251
Single-nucleotide-resolution mapping of m6A 19. Silakov A, Grove TL, Radle MI, Bauerle MR,
and m6Am throughout the transcriptome. Nat Green MT, Rosenzweig AC, Boal AK, Booker
Methods 12(8):767–772. [Link] SJ (2014) Characterization of a cross-linked
1038/nmeth.3453 protein-nucleic acid substrate radical in the
10. Ule J, Jensen KB, Ruggiu M, Mele A, Ule A, reaction catalyzed by RlmN. J Am Chem Soc
Darnell RB (2003) CLIP identifies Nova- 136(23):8221–8228. [Link]
regulated RNA networks in the brain. Science 1021/ja410560p
302(5648):1212–1215. [Link] 20. Boal AK, Grove TL, McLaughlin MI, Yenna-
1126/science.1090095 war NH, Booker SJ, Rosenzweig AC (2011)
11. Hafner M, Lianoglou S, Tuschl T, Betel D Structural basis for methyl transfer by a radical
(2012) Genome-wide identification of SAM enzyme. Science 332(6033):1089–1092.
miRNA targets by PAR-CLIP. Methods 58 [Link]
(2):94–105. [Link] 21. King MY, Redman KL (2002) RNA methyl-
ymeth.2012.08.006 transferases utilize two cysteine residues in the
12. Konig J, Zarnack K, Rot G, Curk T, Kayikci M, formation of 5-methylcytosine. Biochemistry
Zupan B, Turner DJ, Luscombe NM, Ule J 41(37):11218–11225
(2010) iCLIP reveals the function of hnRNP 22. Yan F, LaMarre JM, Rohrich R, Wiesner J,
particles in splicing at individual nucleotide Jomaa H, Mankin AS, Fujimori DG (2010)
resolution. Nat Struct Mol Biol 17 RlmN and Cfr are radical SAM enzymes
(7):909–915. [Link] involved in methylation of ribosomal RNA. J
nsmb.1838 Am Chem Soc 132(11):3953–3964. https://
13. Haag S, Kretschmer J, Sloan KE, Bohnsack MT [Link]/10.1021/ja910850y
(2017) Crosslinking methods to identify RNA 23. Yan F, Fujimori DG (2011) RNA methylation
methyltransferase targets in vivo. Methods Mol by radical SAM enzymes RlmN and Cfr pro-
Biol 1562:269–281. [Link] ceeds via methylene transfer and hydride shift.
1007/978-1-4939-6807-7_18 Proc Natl Acad Sci U S A 108(10):3930–3934.
14. Zhang CL, Darnell RB (2011) Mapping in vivo [Link]
protein-RNA interactions at single-nucleotide 24. Stojkovic V, Chu T, Therizols G, Weinberg
resolution from HITS-CLIP data. Nat Bio- DE, Fujimori DG (2018) miCLIP-MaPseq, a
technol 29(7):607–U686. [Link] substrate identification approach for radical
10.1038/nbt.1873 SAM RNA methylating enzymes. J Am Chem
15. Hussain S, Sajini AA, Blanco S, Dietmann S, Soc 140(23):7135–7143. [Link]
Lombard P, Sugimoto Y, Paramor M, Gleeson 1021/jacs.8b02618
JG, Odom DT, Ule J, Frye M (2013) NSun2- 25. Benitez-Paez A, Villarroya M, Armengod ME
mediated cytosine-5 methylation of vault non- (2012) The Escherichia coli RlmN methyl-
coding RNA determines its processing into transferase is a dual-specificity enzyme that
regulatory small RNAs. Cell Rep 4 modifies both rRNA and tRNA and controls
(2):255–261. [Link] translational accuracy. RNA 18
rep.2013.06.029 (10):1783–1795. [Link]
16. McCusker KP, Medzihradszky KF, Shiver AL, rna.033266.112
Nichols RJ, Yan F, Maltby DA, Gross CA, 26. Fitzsimmons CM, Fujimori DG (2016) Deter-
Fujimori DG (2012) Covalent intermediate in minants of tRNA recognition by the radical
122 Vanja Stojković et al.
SAM enzyme RlmN. PLoS One 11(11): 31. Langmead B, Trapnell C, Pop M, Salzberg SL
e0167298. [Link] (2009) Ultrafast and memory-efficient align-
pone.0167298 ment of short DNA sequences to the human
27. Dominissini D, Moshitch-Moshkovitz S, genome. Genome Biol 10(3):R25. https://
Salmon-Divon M, Amariglio N, Rechavi G [Link]/10.1186/gb-2009-10-3-r25
(2013) Transcriptome-wide mapping of N(6)- 32. Anders S, Pyl PT, Huber W (2014) HTSeq--a
methyladenosine by m(6)A-seq based on Python framework to work with high-
immunocapturing and massively parallel throughput sequencing data. Bioinformatics
sequencing. Nat Protoc 8(1):176–189. 31(2):166–169
[Link] 33. Thorvaldsdottir H, Robinson JT, Mesirov JP
28. Afgan E, Baker D, van den Beek M, (2013) Integrative Genomics Viewer (IGV):
Blankenberg D, Bouvier D, Cech M, high-performance genomics data visualization
Chilton J, Clements D, Coraor N, and exploration. Brief Bioinform 14
Eberhard C, Gruning B, Guerler A, Hillman- (2):178–192. [Link]
Jackson J, Von Kuster G, Rasche E, Soranzo N, bbs017
Turaga N, Taylor J, Nekrutenko A, Goecks J 34. Robinson JT, Thorvaldsdóttir H, Winckler W,
(2016) The Galaxy platform for accessible, Guttman M, Lander ES, Getz G, Mesirov JP
reproducible and collaborative biomedical ana- (2011) Integrative genomic viewer. Nat Bio-
lyses: 2016 update. Nucleic Acids Res 44(W1): technol 29:24–26
W3–W10. [Link] 35. Boccaletto P, Machnicka MA, Purta E,
gkw343 Pia˛tkowski P, Bagiński B, Wirecki TK, de
29. Blankenberg D, Gordon A, Von Kuster G, Crécy-Lagard V, Ross R, Limbach PA,
Coraor N, Taylor J, Nekrutenko A, Galaxy T Kotter A, Helm M (2017) MODOMICS: a
(2010) Manipulation of FASTQ data with Gal- database of RNA modification pathways.
axy. Bioinformatics 26(14):1783–1785. 2017 update. Nucleic Acids Res 46(D1):
[Link] D303–D307
btq281 36. Dunn JG, Weissman JS (2016) Plastid:
30. Langmead B, Salzberg SL (2012) Fast gapped- nucleotide-resolution analysis of next-
read alignment with bowtie 2. Nat Methods 9 generation sequencing and genomics data.
(4):357–359. [Link] BMC Genomics 17(1):958. [Link]
nmeth.1923 10.1186/s12864-016-3278-x
Chapter 8
Abstract
Epitranscriptomic RNA modifications function as an important layer of gene regulation that modulates the
function of RNA transcripts. A key step in understanding how RNA modifications regulate biological
processes is the mapping of their locations, which is most commonly done by RNA immunoprecipitation
(RIP) using modification-specific antibodies. Here, we describe the use of a photoactivatable
ribonucleoside-enhanced cross-linking and immunoprecipitation (PAR-CLIP) method, in conjunction
with RNA modification-specific antibodies, to map modification sites. First described as photo-
crosslinking-assisted m6A sequencing (PA-m6A-seq), this method allows the mapping of RNA modifica-
tions at a higher resolution, with lower background than traditional RIP, and can be adapted to any RNA
modification for which a specific antibody is available.
Key words Epitranscriptomic RNA modification, RNA immunoprecipitation, m6A, m5C, ac4C
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
123
124 Bryan R. Cullen and Kevin Tsai
Fig. 1 Overview of PA-mod-seq. (a) Flowchart of the steps involved in the method, with PA-m6A-seq as an
example. (b) Example of PA-m6A-seq results visualized in Integrated Genome Viewer (IGV), m6A sites mapped
on the first 500 nts of Simian virus 40 (SV40) VP1 mRNA (GEO database accession #GSE106698) [14]. Red/
blue bars in the upper coverage pileup track denote sites of cross-link-induced T > C conversions, with the
height of red/blue bars proportional to the occurrence of T and C residues. Blue bars in the bottom individual
read track denote the location of T > C conversions in each read. Note that a diverse variety (3+) of T > C
conversion sites are expected in a good m6A peak
2 Materials
2.3 RNA End Repair 1. 10 U/μL Calf intestinal phosphatase (see Note 2).
2. 10 NEB CutSmart Buffer.
3. Phosphatase wash buffer: 50 mM Tris–HCl pH 7.5, 20 mM
EGTA-NaOH pH 7.5, 0.5% NP40 in RNase-free H2O.
4. PNK buffer without DTT: 50 mM Tris–HCl pH 7.5, 50 mM
NaCl, 10 mM MgCl2 in RNase-free H2O.
5. 1 M DTT.
6. ATP.
7. T4 Polynucleotide Kinase (T4-PNK).
2.5 Sequencing 1. NEB Next Small RNA Library Prep Set for Illumina.
Library Preparation 2. Novex 10-well 6% TBE gel.
3 Methods
3.1 Tissue Culture Start with an amount of cells that produce ~10 μg of RNA for the
immunoprecipitation step (see Notes 3 and 4). Passage cells so they
are ~70% confluent and actively growing the day before harvest.
Actively growing cells are essential to ensure efficient 4SU uptake
and incorporation into RNA.
1. Add 4SU directly to the cell culture media to a final concentra-
tion of 100 μM (see Note 1).
2. 16–24 h later, collect cells as appropriate for the cell type:
scrape off attached cells or collect suspension cells. Spin cells
down at 500 g for 10 min to pellet (see Note 5).
3. Wash cell pellet once with ice-cold PBS, spin down at 500 g
for 10 min, and remove PBS.
4. Lyse cell pellet directly in Trizol using 1 mL for every 107 cells.
Cells in Trizol can be stored at 80 C.
128 Bryan R. Cullen and Kevin Tsai
3.2 Total RNA 1. Aliquot every 1 mL of Trizol-cell lysate into 1.5 mL tubes.
Extraction Using Trizol 2. Add 200 μL chloroform to every 1 mL of Trizol, shake vigor-
ously for 15 s, and incubate at room temperature for 3 min.
3. Centrifuge at 12,000 g for 15 min at 4 C.
4. Collect the upper aqueous phase into a new tube with 500 μL
isopropanol (avoid collecting the white interphase), and incu-
bate at room temperature for 10 min.
5. Pellet RNA by centrifugation at 12,000 g for 15 min at 4 C.
6. Precipitate RNA with dH2O:NaOAc:EtOH ¼ 1:0.1:2.2 vol-
ume ([Link] μL each), and then add 1 μL of GlycoBlue.
Mix well and incubate at 80 C for 30 min (or on dry ice for
15 min) (see Note 6).
7. Pellet RNA again at 12,000 g for 20 min at 4 C.
8. Wash pellet with 1 mL 70% ethanol, and centrifuge at
12,000 g for 10 min at 4 C.
9. Remove supernatant. Do a quick spin and carefully remove
residual ethanol with a clean pipette.
10. Resuspend RNA pellet in RNase-free H2O, using 25 μL for
every 1 mL of starting Trizol.
3.3 Poly(A) Depending on the cell type, roughly 1–2.5% of total RNA is poly
Purification (A)+ mRNA. An IP reaction requires 8–12 μg of RNA; thus start by
using ~600 μg of total RNA for poly(A) purification, aiming for a
yield of 10 μg poly(A) + mRNA.
1. Follow the Poly(A)Purist MAG Kit instructions to isolate poly
(A)+ RNA. Resuspend the resulting RNA pellet in 30 μL
dH2O.
2. Measure the concentration of RNA with a Nanodrop spectro-
photometer prior to starting the IP.
3.5 RNA End Repair 1. Prepare dephosphorylation reaction mix by adding 5 μL CIP
(final concentration 0.5 U/μL), 10 μL of 10x NEB CutSmart
Buffer, and 85 μL of H2O. Resuspend beads from each IP in
100 μL of this reaction mix.
2. Incubate the dephosphorylation reaction on a tube shaker at
800 rpm in a 37 C incubator for 10 min.
3. Wash beads twice with 500 μL of phosphatase wash buffer as
before.
4. Wash beads twice with 500 μL PNK buffer without DTT.
130 Bryan R. Cullen and Kevin Tsai
3.6 RNA Elution 1. Prepare the Proteinase K elution mix, for each IP: 75 μL of 4x
Proteinase K buffer, 225 μL of H2O, and 4.2 μL of Proteinase
K (~85 μg).
2. Resuspend the washed beads in Proteinase K elution mix
(300 μL per IP), and then incubate at 50 C for 90 min,
tapping the tubes to mix every 15 min (alternatively, use an
Eppendorf thermoshaker set at 900 rpm).
3. After proteinase digestion, all RNA originally bound to anti-
bodies should be in the supernatant (~300 μL). Transfer super-
natant to a fresh tube.
4. Remove Proteinase K using Trizol LS: Add 900 μL Trizol LS to
the 300 μL eluate, and mix well.
5. Add 240 μL of chloroform, vortex for 15 s, wait for 2 min, and
then centrifuge at 12,000 g for 15 min at 4 C to separate the
organic phase from the aqueous phase.
6. Collect the upper aqueous phase into a new tube, and mix with
600 μL of isopropanol and 1 μL of GlycoBlue Coprecipitant.
Wait for 10 min, and then centrifuge at 12,000 g for 20 min
at 4 C to pellet RNA (see Note 9).
7. (Optional cleanup) Resuspend RNA pellet in 300 μL of H2O,
and then add 30 μL of sodium acetate, 1 μL of GlycoBlue, and
660 μL of 100% ethanol. Precipitate at 20 C for 2 h or
overnight. Then centrifuge at 12,000 g for 30 min at 4 C
(see Note 10).
8. Wash pellet with 70% ethanol, vortex briefly, and centrifuge at
12,000 g for 10 min at 4 C.
9. Resuspend each pellet in 15 μL of H2O.
3.7 Sequencing 1. Prepare sequencing libraries with the NEB Next Small RNA
Library Preparation Library Prep Set for Illumina following the kit instructions.
The expected RNA size is the RNase footprint of the antibody,
which is typically ~32–50 nt. With the ligated 50 and 30 adapters
totaling 120 nt, it is necessary to isolate bands ~150–170 nt on
a TBE polyacrylamide gel. We have found the Invitrogen
Novex 6% TBE gel to give the best separation between our
desired product and adapter dimers (120 nt).
RNA Modification Mapping Using PA-mod-seq 131
3.8 Sequencing Data Sequencing result analysis typically involves trimming off adaptor
Analysis sequences, discarding any reads shorter than 15 nts, aligning
sequencing reads to the genome sequence of interest, screening
for T > C conversions, and reformatting the alignment information
for visualization in the Integrated Genome Viewer (IGV) (see
example in Fig. 1b).
1. We use the FASTX toolkit [18] for sequencing read preproces-
sing; this includes screening for reads with a FASTQ quality
score >Q33, removing adapter sequences, and selecting for a
minimum read length of 15 nts. Alternatively, we have used
Cutadapt [19] with good results.
2. We then use Bowtie [20, 21] for alignment of the sequencing
reads to the genome of interest. If you are interested in human
cellular transcripts, for example, you can align your reads to a
human genome build such as hg19. If you are interested in viral
transcripts, you should pre-align the reads to the host genome,
then take the host non-aligning reads, and align them to the
viral genome. If your model system of interest has a heavily
spliced transcriptome, it might be informative to align to the
transcriptome with a splice-aware aligner, such as TopHat
[22, 23] or STAR [24].
3. To screen for T > C conversions, freely available analysis
packages such as PARalyzer [25] are available. If your model
system of interest has a simple genome like a virus, then a
simple script can be used to screen for T > C conversions.
Note that if you only use a simple T > C conversion screen
with no statistics, we would recommend manually looking at
each peak with the following criteria for reliable peaks: Each
peak needs to consist of more than three distinct reads (varying
in length or alignment location so that they are unlikely to be
PCR duplicate reads), with at least three different locations of
T > C conversions.
4. The Integrated Genome Viewer (IGV) from the Broad Insti-
tute [26] can be used to visualize sequencing results. Running
Bowtie with the “--sam” argument will give results output in
the SAM format. IGV reads alignments in the BAM format
more efficiently, with a requirement that BAM files be pre-
sorted and indexed. The SAMtools suite [27] can be used to
convert SAM to BAM, and sort and index the files for loading
into IGV.
132 Bryan R. Cullen and Kevin Tsai
4 Notes
9. The RNA pellet at this step will be small yet visible. If the pellet
is not visible, add an additional 100 μL of isopropanol and 1 μL
of GlycoBlue, mix well, and repeat the 20-min centrifugation
step. A preincubation of the tube at 80 C or on dry ice prior
to centrifugation may also enhance precipitation.
10. This optional re-precipitation is to ensure minimum salt con-
tamination going into the library preparation. If the RNA
pellet from the previous step is very small and hard to spot,
we omit this step to avoid loss of the pellet.
Acknowledgments
References
1. Li S, Mason CE (2014) The pivotal regulatory methyladenosine by m(6)A-seq based on
landscape of RNA modifications. Annu Rev immunocapturing and massively parallel
Genomics Hum Genet 15:127–150. https:// sequencing. Nat Protoc 8(1):176–189.
[Link]/10.1146/annurev-genom-090413- [Link]
025405 8. Dominissini D, Moshitch-Moshkovitz S,
2. Wang GG, Allis CD, Chi P (2007) Chromatin Schwartz S, Salmon-Divon M, Ungar L,
remodeling and cancer, part II: Osenberg S, Cesarkas K, Jacob-Hirsch J,
ATP-dependent chromatin remodeling. Amariglio N, Kupiec M, Sorek R, Rechavi G
Trends Mol Med 13(9):373–380. https:// (2012) Topology of the human and mouse
[Link]/10.1016/[Link].2007.07.004 m6A RNA methylomes revealed by m6A-seq.
3. Kennedy EM, Courtney DG, Tsai K, Cullen Nature 485(7397):201–206. [Link]
BR (2017) Viral epitranscriptomics. J Virol 91 10.1038/nature11112
(9). [Link] 9. Arango D, Sturgill D, Alhusaini N, Dillman
4. Desrosiers R, Friderici K, Rottman F (1974) AA, Sweet TJ, Hanson G, Hosogane M, Sin-
Identification of methylated nucleosides in clair WR, Nanan KK, Mandler MD, Fox SD,
messenger RNA from Novikoff hepatoma Zengeya TT, Andresson T, Meier JL, Coller J,
cells. Proc Natl Acad Sci U S A 71 Oberdoerffer S (2018) Acetylation of cytidine
(10):3971–3975. [Link] in mRNA promotes translation efficiency. Cell
pnas.71.10.3971 175(7):1872–1886. e1824. [Link]
5. Lavi S, Shatkin AJ (1975) Methylated simian 10.1016/[Link].2018.10.030
virus 40-specific RNA from nuclei and cyto- 10. McIntyre ABR, Gokhale NS, Cerchietti L, Jaf-
plasm of infected BSC-1 cells. Proc Natl Acad frey SR, Horner SM, Mason CE (2020) Limits
Sci U S A 72(6):2012–2016. [Link] in the detection of m6A changes using
10.1073/pnas.72.6.2012 MeRIP/m6A-seq. Sci Rep 10(1):6590.
6. Meyer KD, Saletore Y, Zumbo P, Elemento O, [Link]
Mason CE, Jaffrey SR (2012) Comprehensive 63355-3
analysis of mRNA methylation reveals enrich- 11. Chen K, Lu Z, Wang X, Fu Y, Luo GZ, Liu N,
ment in 3’ UTRs and near stop codons. Cell Han D, Dominissini D, Dai Q, Pan T, He C
149(7):1635–1646. [Link] (2015) High-resolution N(6)-methyladeno-
1016/[Link].2012.05.003 sine (m(6) A) map using photo-crosslinking-
7. Dominissini D, Moshitch-Moshkovitz S, assisted m(6) A sequencing. Angew Chem Int
Salmon-Divon M, Amariglio N, Rechavi G Ed Engl 54(5):1587–1590. [Link]
(2013) Transcriptome-wide mapping of N(6)- 10.1002/anie.201410647
134 Bryan R. Cullen and Kevin Tsai
12. Courtney DG, Kennedy EM, Dumm RE, Chapter 11:Unit 11, 17. doi:[Link]
Bogerd HP, Tsai K, Heaton NS, Cullen BR 10.1002/0471250953.bi1107s32
(2017) Epitranscriptomic enhancement of 21. Langmead B, Trapnell C, Pop M, Salzberg SL
influenza a virus gene expression and replica- (2009) Ultrafast and memory-efficient align-
tion. Cell Host Microbe 22(3):377–386. e375. ment of short DNA sequences to the human
[Link] genome. Genome Biol 10(3):R25. https://
004 [Link]/10.1186/gb-2009-10-3-r25
13. Kennedy EM, Bogerd HP, Kornepati AV, 22. Trapnell C, Pachter L, Salzberg SL (2009)
Kang D, Ghoshal D, Marshall JB, Poling BC, TopHat: discovering splice junctions with
Tsai K, Gokhale NS, Horner SM, Cullen BR RNA-Seq. Bioinformatics 25(9):1105–1111.
(2016) Posttranscriptional m(6)a editing of [Link]
HIV-1 mRNAs enhances viral gene expression. btp120
Cell Host Microbe 19(5):675–685. https:// 23. Trapnell C, Roberts A, Goff L, Pertea G,
[Link]/10.1016/[Link].2016.04.002 Kim D, Kelley DR, Pimentel H, Salzberg SL,
14. Tsai K, Courtney DG, Cullen BR (2018) Addi- Rinn JL, Pachter L (2012) Differential gene
tion of m6A to SV40 late mRNAs enhances and transcript expression analysis of RNA-seq
viral structural gene expression and replication. experiments with TopHat and Cufflinks. Nat
PLoS Pathog 14(2):e1006919. [Link] Protoc 7(3):562–578. [Link]
org/10.1371/[Link].1006919 1038/nprot.2012.016
15. Hafner M, Landthaler M, Burger L, 24. Dobin A, Davis CA, Schlesinger F, Drenkow J,
Khorshid M, Hausser J, Berninger P, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras
Rothballer A, Ascano M Jr, Jungkamp AC, TR (2013) STAR: ultrafast universal RNA-seq
Munschauer M, Ulrich A, Wardle GS, aligner. Bioinformatics 29(1):15–21. https://
Dewell S, Zavolan M, Tuschl T (2010) [Link]/10.1093/bioinformatics/bts635
Transcriptome-wide identification of 25. Corcoran DL, Georgiev S, Mukherjee N,
RNA-binding protein and microRNA target Gottwein E, Skalsky RL, Keene JD, Ohler U
sites by PAR-CLIP. Cell 141(1):129–141. (2011) PARalyzer: definition of RNA binding
[Link] sites from PAR-CLIP short-read sequence
16. Courtney DG, Tsai K, Bogerd HP, Kennedy data. Genome Biol 12(8):R79. [Link]
EM, Law BA, Emery A, Swanstrom R, Holley org/10.1186/gb-2011-12-8-r79
CL, Cullen BR (2019) Epitranscriptomic addi- 26. Robinson JT, Thorvaldsdottir H, Winckler W,
tion of m5C to HIV-1 transcripts regulates Guttman M, Lander ES, Getz G, Mesirov JP
viral gene expression. Cell Host Microbe 26 (2011) Integrative genomics viewer. Nat Bio-
(2):217–227.e216. [Link] technol 29(1):24–26. [Link]
1016/[Link].2019.07.005 1038/nbt.1754
17. Tsai K, Jaguva Vasudevan AA, Martinez 27. Li H, Handsaker B, Wysoker A, Fennell T,
Campos C, Emery A, Swanstrom R, Cullen Ruan J, Homer N, Marth G, Abecasis G, Dur-
BR (2020) Acetylation of cytidine residues bin R (2009) Genome project data processing
boosts HIV-1 gene expression by increasing S: the sequence alignment/map format and
viral RNA stability. Cell Host Microbe 28 SAMtools. Bioinformatics 25
(2):306–312.e6. [Link] (16):2078–2079. [Link]
chom.2020.05.011 bioinformatics/btp352
18. Gordon A, Hannon G (2010) FastX toolkit. 28. Eckwahl MJ, Arnion H, Kharytonchyk S,
[Link] Zang T, Bieniasz PD, Telesnitsky A, Wolin SL
19. Martin M (2011) Cutadapt removes adapter (2016) Analysis of the human immunodefi-
sequences from high-throughput sequencing ciency virus-1 RNA packageome. RNA 22
reads 2011. EMBnet J 17(1):3. [Link] (8):1228–1238. [Link]
org/10.14806/ej.17.1.200 rna.057299.116
20. Langmead B (2010) Aligning short sequencing
reads with Bowtie. Curr Protoc Bioinformatics
Chapter 9
Abstract
RNA has coevolved with numerous posttranscriptional modifications to sculpt interactions with proteins
and other molecules. One of these modifications is 5-methylcytosine (m5C) and mapping the position and
quantifying the level in different types of cellular RNAs and tissues is an important objective in the field of
epitranscriptomics. Both in plants and animals bisulfite conversion has long been the gold standard for
detection of m5C in DNA but it can also be applied to RNA. Here, we detail methods for highly
reproducible bisulfite treatment of RNA, efficient locus-specific PCR amplification, detection of candidate
sites by sequencing on the Illumina MiSeq platform, and bioinformatic calling of non-converted sites.
Key words Bisulfite conversion, Epitranscriptome, Fluidigm Access Array, Illumina, Next-generation
sequencing, 5-Methylcytosine
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols , Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
135
136 Jun Li et al.
Fig. 1 Protocol overview showing the workflow for either parallel or single amplicon amplification for effective
detection of m5C. (a) Parallel amplification and sequencing of up to 2304 amplicons across 48 tissues and
48 primer pairs. Forty-eight different tissues can be selected, total RNA isolated and purified, spiked with
MGFP in vitro-transcribed control RNA and bisulfite converted. Bisulfite-converted RNA is reverse transcribed
Single-nucleotide Resolution of RNA m5C 137
2 Materials
Fig. 1 (continued) (RT) to cDNA using gene-specific RT primers that include the positive control MAG5
(AT5G47480) and negative control MGFP. Target regions are PCR amplified using a Fluidigm Access Array
Integrated Fluidic Circuit (IFC); up to 2304 amplicons are harvested and eluted pools are quantified. Equal
concentrations of the pools are combined into a final pool, purified using AMPure beads, accurately quantified,
PhiX control library spiked-in, and subjected to sequencing on the Illumina MiSeq platform. (b) Single amplicon
amplification and sequencing. A single tissue is selected, RNA isolated and purified in triplicate, spiked with
MGFP in vitro-transcribed control RNA and bisulfite converted. Bisulfite conversion and cDNA synthesis are the
same as outlined above except a specific target RT primer is used. The target amplicon is PCR amplified,
triplicate amplicons are pooled, size and concentration are assessed on a Shimadzu MultiNA, and amplicons
are pooled at equal concentration. Pooled amplicons are purified, PhiX control library spiked-in, and subjected
to sequencing on the Illumina MiSeq platform
138 Jun Li et al.
2.2 Sodium Bisulfite 1. Sodium bisulfite solution: 40% (w/v) Sodium metabisulfite,
Conversion 0.6 mM hydroquinone, final pH 5.1.
To prepare the sodium bisulfite solution, prepare the
following:
0.6 M Hydroquinone: Weigh 66 mg hydroquinone and
place into a 1.5 mL tube. Add H2O to 1 mL and cover in foil to
protect from light. Place in an orbital shaker to dissolve.
40% (w/v) Sodium bisulfite: Dissolve 4 g sodium metabi-
sulfite in 10 mL H2O in a 50 mL falcon tube and vortex until it
completely dissolves.
Add 10μL 0.6 M hydroquinone to the 40% sodium bisul-
fite solution, vortex, and adjust pH to 5.1 with 10 M NaOH.
Filter the solution through a 0.2μm filter. Cover in foil to
protect from light (see Note 1).
2. 1 M Tris–HCl, pH 9.0.
3. Micro Bio-Spin ™ P-6 Gel Columns.
4. Mineral oil.
5. 75% Ethanol.
6. 100% Ethanol.
7. 3 M sodium acetate, pH 5.2.
8. 5 mg/mL Glycogen.
3 Methods
3.1 RNA Extraction, Total RNA is extracted and purified directly from tissue with 1 mL
Purification, of TRIzol™ as per the manufacturer’s protocol. RNA is then
and DNase Treatment treated with TURBO™ DNase as per the manufacturer’s protocol.
Assess the integrity of the RNA by using a RNA 6000 Nano Chip
on the Agilent 2100 Bioanalyzer according to the manufacturer’s
protocol.
3.2 Generation 1. Linearize the phMGFP vector by using the restriction enzyme
of the MGFP In Vitro XbaI and purify the linearized DNA vector according to the
Transcript Spike-in HiScribe T7 In Vitro Transcription Kit protocol.
Control 2. Perform in vitro transcription according to the HiScribe T7 In
Vitro Transcription Kit protocol by using 1μg of linearized
DNA. An incubation period of 4 h at 37 C with the kit
components is sufficient.
3. Add 2 U TURBO™ DNase and incubate at 37 C for 30 min.
4. Transfer the reaction to a Phase Lock Gel™ tube and make the
volume of the reaction up to 100μL with ultrapure H2O.
5. Add an equal volume of phenol:water and chloroform, shake
vigorously for 15 s, and centrifuge at 15,000 g for 5 min.
6. Add the same volume of chloroform as in step 5 to the tube,
shake vigorously for 15 s, and centrifuge at 15,000 g for
5 min again.
7. Transfer the aqueous phase to a clean 1.5 mL tube. Add 1/10
volume 3 M sodium acetate, 3 volumes of 100% ethanol, and
1μL glycogen; vortex; and precipitate the RNA overnight at
80 C.
8. Centrifuge RNA at 17,000 g at 4 C for 60 min and carefully
remove the supernatant.
9. Add 1 mL 75% ethanol to the RNA, invert five times, and
centrifuge at 7500 g at 4 C for 10 min (see Note 3).
10. Carefully remove the supernatant and let the pellet air-dry for
approximately 15 min (see Note 4).
11. Resuspend the RNA in 25μL of ultrapure H2O.
12. Optional step: Treat 5μg of in vitro-transcribed MGFP tran-
script with 2 U TURBO™ DNase according to the manufac-
turer’s protocol at 37 C for 30 min.
13. Assess the integrity and size of the MGFP in vitro transcripts by
using an RNA 6000 Nano Chip on the Agilent 2100 Bioana-
lyzer according to the manufacturer’s protocol (see Note 5).
3.3 Bisulfite 1. Add 1/2000 of the MGFP RNA transcript to 2μg DNase-
Conversion of RNA treated purified total RNA. Increase the volume of the RNA
sample to 20μL with ultrapure H2O.
2. Denature RNA by heating to 75 C for 5 min in a heat block.
Single-nucleotide Resolution of RNA m5C 141
Fig. 2 Overview of bisulfite conversion of RNA, reverse transcription to cDNA, and PCR amplification. (a) In the
in vitro-transcribed MGFP sequence, unmodified cytosines (underlined) are converted to uracil, reverse
transcribed (RT) by reverse transcriptase to cDNA, and then PCR amplified. RT and PCR primers are designed
to avoid stretches of converted cytosines to prevent preferential amplification of converted sequences which
may incorrectly indicate efficient bisulfite conversion. (b) In MAG5 control and other candidate sequences,
primers are designed to span areas containing converted cytosines to preferentially amplify converted
sequences. C3349 is methylated in Arabidopsis thaliana and serves as an over-conversion control. Flanking
cytosines are not methylated and should be completely converted. Primers are designed with a Tm of
59–61 C, preferably with a 30 G nucleotide and to amplify PCR products of 170–200 bp
Single-nucleotide Resolution of RNA m5C 143
Fig. 3 Overview of first and second PCR amplification of target regions. (a) For the first PCR, the forward PCR
primer is designed with the gene-specific sequence (GS) and universal forward tag called Common Sequence,
CS1 (50 - TACGGTAGCAGAGACTTGGTCT -30 ), and reverse PCR primer is designed with the gene-specific
sequence (GSS) and universal reverse tag called Common Sequence CS2 (5’-ACACTGACGACATGGTTCTACA
-30 ). (b) For the second PCR, the forward primer is designed with the CS1 and Illumina P5 sequences and the
reverse primer contains the CS2, barcoding, and Illumina P7 sequences. The Fluidigm barcodes or indexes are
10 nt in length
3.6 Individual PCR 1. For a 10μL PCR, add 0.2μL of KAPA HiFi DNA Polymerase,
Amplification, 2μL of 5 HiFi Fidelity buffer (with MgCl2), 0.3μL of 10 mM
Quantification, dNTP, 0.4μL of 10μM forward primer (CS1_GSS), 0.4μL of
and Pooling 10μM reverse primer (CS2_GSS), 1μL of diluted cDNA, and
H2O to a final volume of 10μL. Perform PCR for each ampli-
con in triplicate.
2. Gently finger vortex, briefly centrifuge, and place into a pre-
heated thermal cycler.
3. Perform a two-step thermal cycling PCR program. See Table 1
for more details.
4. Pool the triplicates and perform an AMPure bead cleanup at a
ratio of 1.8:1 to remove unincorporated primers and primer
dimers. Repeat this step (see Notes 13 and 14).
5. Assess PCR amplicon size and concentration after separation
on a Shimadzu Microchip Electrophoresis System MCE®-202
MultiNA.
6. Normalize the concentration of each amplicon in the experi-
ment by dilution with H2O to a concentration in the range of
0.5–5 ng/μL.
7. Perform the barcoding and Illumina adapter addition PCR. In
a 10μL PCR, add 0.2μL of KAPA HiFi DNA Polymerase, 2μL
of 5 HiFi Fidelity buffer (with MgCl2), 0.3μL of 10 mM
dNTP, 1μL of 10μM forward primer (P5_CS1), 1μL of
10μM reverse primer (P7_CS2), 2μL of diluted PCR amplicon,
and H2O to a final volume of 10μL.
8. Gently finger vortex, briefly centrifuge, and place into a pre-
heated thermal cycler.
9. Perform a two-step thermal cycling PCR program. See Table 2
for more details.
10. Assess PCR amplicon size and concentration after separation
on a Shimadzu Microchip Electrophoresis System MCE®-202
MultiNA.
11. Pool the amplicons in equimolar concentration and purify
them using AMPure beads according to the manufacturer’s
protocol. Use a ratio of beads to pooled amplicons of 0.9:1
to ensure binding of amplicons and not primer dimers or
unincorporated primers.
Single-nucleotide Resolution of RNA m5C 145
Table 1
Two-step thermal cycling conditions for the amplification of individual amplicons
Table 2
One-step thermal cycling conditions for the addition of barcodes and Illumina adapters
3.7 Parallel PCR 1. Prime the Access Array according to the manufacturer’s
Amplification Using protocol.
a Fluidigm Access 2. Pre-warm the 20 Access Array loading reagent to room tem-
Array Integrated perature before use. Prepare the pooled 48-oligonucleotide
Fluidic Circuit (IFC)
146 Jun Li et al.
Table 3
Multistep thermal cycling conditions for the Access Array
3.8 MiSeq 1. Prepare the sample sheet using the Illumina Experiment Man-
Sequencing ager by following the manufacturer’s protocol (see Note 15).
2. Dilute the library to 10 nM in EBT buffer based on the con-
centrations determined by the qPCR. From this point, keep the
libraries on ice.
3. Dilute the PhiX control library to 2 nM by adding 8μL EBT
buffer to 2μL of the 10 nM PhiX control library (see Note 16).
4. Denature the pooled libraries and PhiX control library sepa-
rately by adding 10μL of 0.2 M NaOH to 10μL of the 2 nM
libraries (see Note 17).
148 Jun Li et al.
3.9 Bioinformatics 1. To trim the Illumina adaptor sequences that were incorporated
Analysis of Data into the amplicons to permit sequencing of the 150 bp paired-
end reads, use Trimmomatic in palindromic mode [15].
2. Sequencing reads can be aligned with meRanTK by using
Bowtie2 internally [16]. Assemble reference sequences for the
alignment by using the segments of RNA interrogated by
sequencing prior to bisulfite conversion.
3. Extract the methylation state of individual cytosines from
bisulfite-read alignments by using meRanCall. The number of
reads can be extracted from the aligned sequencing reads in
order to determine read coverage at a given cytosine.
4. To call differentially methylated cytosines use meRanCompare.
The number of reads can be extracted from the aligned
sequencing reads in order to determine read coverage at a
given cytosine (Fig. 4).
4 Notes
Fig. 4 Representative analysis of an Illumina MiSeq amplicon sequencing of negative and positive controls. (a)
A region of the MGFP spiked-in in vitro control transcript showing even coverage and all cytosines are
converted (no methylation). The y-axis shows the read depth and the x-axis shows the cytosines (numbers) in
the sequenced region. (b) A region of the Mag5 gene that shows converted and non-converted cytosine,
C3349. Cytosines flanking C339 are completely converted, demonstrating that bisulfite conversion was very
efficient. The heatmaps display the cytosine non-conversion percentage
6. Tilt the 1.5 mL tube at a 45 angle and then slowly pipette the
mineral oil directly on top of the RNA-bisulfite reaction
mixture.
7. Emptying of the Micro Bio-Spin gel column takes about 2 min.
If the gel column does not empty by gravity, place the lid back
onto the column and remove again.
8. Gently pipette the reaction mixture onto the gel bed and avoid
disturbing the gel bed. Minimize the transfer of mineral oil to
the column although there will be traces which is unavoidable.
9. About 25% of the RNA is lost during the procedure, and we
find that 10μL of H2O/2μg RNA used in the bisulfite conver-
sion reaction results in concentrations of ~150 ng/μL.
10. Bisulfite treatment of the RNA causes significant shearing and
we have observed that shorter amplicons are preferentially
amplified over longer amplicons.
Longer PCR amplicons increase the tendency of detecting
non-converted cytosines in RNA exhibiting strong secondary
structure.
11. Inefficient bisulfite conversion may result in unconverted cyto-
sines, so it is important to ensure that the PCR primers are not
biasing the amplification toward converted cytosines.
12. Occasionally, not all triplicates successfully amplify and it may
be necessary to optimize the PCR.
13. We elute the purified PCR products in 10–30μL depending on
the amount of amplified PCR products.
14. After purification of the amplicons, residual ethanol may
remain in the purified amplicons. We find that concentrating
down the pooled amplicons even if there is <55μL and adding
H2O to 55μL are best to remove as much ethanol as possible.
15. The sample sheet is required to insert the sample names and
adapter indices used for each sample. We have selected the
“Other” as the category followed by “Fastq only.” This option
generates FASTQ files only and also enables the deselection of
downstream processing steps like adapter trimming, allowing
trimming and mapping to be performed separately.
16. The prepared PhiX library is added to the pooled amplicon
libraries as an internal control for the MiSeq sequencing run.
17. It is best to prepare fresh 0.2 M NaOH for the denaturation of
libraries.
18. Loading 10% PhiX control library is sufficient for low-diversity
libraries. We have previously loaded between 7 and 10 pM.
Underloading of the libraries can give cluster densities below
the optimal range and overloading of the libraries can give
cluster densities above the optimal range, reducing the quality
of the data. The optimal cluster density is 700–1000 K/mm2.
Single-nucleotide Resolution of RNA m5C 151
Acknowledgments
References
1. Burgess A, David R, Searle IR (2016) Deci- 9. Lovejoy AF, Riordan DP, Brown PO (2014)
phering the epitranscriptome: a green perspec- Transcriptome-wide mapping of pseudouri-
tive. J Integr Plant Biol 58(10):822–835 dines: pseudouridine synthases modify specific
2. Saletore Y, Meyer K, Korlach J, Vilfan ID, mRNAs in S. cerevisiae. PLoS One 9(10):
Jaffrey S, Mason CE (2012) The birth of the e110799
Epitranscriptome: deciphering the function of 10. Khoddami V, Cairns BR (2013) Identification
RNA modifications. Genome Biol 13(10):175 of direct targets and modified bases of RNA
3. Bujnicki JM (2001) In silico analysis of the cytosine methyltransferases. Nat Biotechnol
tRNA: m1A58 methyltransferase family: 31(5):458–464
homology-based fold prediction and identifica- 11. Hussain S, Sajini AA, Blanco S, Dietmann S,
tion of new members from Eubacteria and Lombard P, Sugimoto Y, Paramor M, Gleeson
Archaea. FEBS Lett 507(2):123–127 JG, Odom DT, Ule J (2013) NSun2-mediated
4. Droogmans L, Roovers M, Bujnicki JM, cytosine-5 methylation of vault noncoding
Tricot C, Hartsch T, Stalon V, Grosjean H RNA determines its processing into regulatory
(2003) Cloning and characterization of tRNA small RNAs. Cell Rep 4(2):255–261
(m1A58) methyltransferase (TrmI) from Ther- 12. Cokus SJ, Feng S, Zhang X, Chen Z,
mus thermophilus HB27, a protein required Merriman B, Haudenschild CD, Pradhan S,
for cell growth at extreme temperatures. Nelson SF, Pellegrini M, Jacobsen SE (2008)
Nucleic Acids Res 31(8):2148–2156 Shotgun bisulphite sequencing of the Arabi-
5. Oerum S, Dégut C, Barraud P, Tisné C (2017) dopsis genome reveals DNA methylation pat-
m1A post-transcriptional modification in terning. Nature 452(7184):215–219
tRNAs. Biomol Ther 7(1):20 13. Schaefer M, Pollex T, Hanna K, Lyko F (2008)
6. Dominissini D, Moshitch-Moshkovitz S, RNA cytosine methylation analysis by bisulfite
Schwartz S, Salmon-Divon M, Ungar L, sequencing. Nucleic Acids Res 37(2):e12–e12
Osenberg S, Cesarkas K, Jacob-Hirsch J, 14. David R, Burgess A, Parker B, Li J, Pulsford K,
Amariglio N, Kupiec M (2012) Topology of Sibbritt T, Preiss T, Searle IR (2017)
the human and mouse m6A RNA methylomes Transcriptome-wide mapping of RNA
revealed by m6A-seq. Nature 485 5-methylcytosine in Arabidopsis mRNAs and
(7397):201–206 noncoding RNAs. Plant Cell 29(3):445–460
7. Meyer KD, Saletore Y, Zumbo P, Elemento O, 15. Bolger AM, Lohse M, Usadel B (2014) Trim-
Mason CE, Jaffrey SR (2012) Comprehensive momatic: a flexible trimmer for Illumina
analysis of mRNA methylation reveals enrich- sequence data. Bioinformatics 30
ment in 30 UTRs and near stop codons. Cell (15):2114–2120
149(7):1635–1646 16. Rieder D, Amort T, Kugler E, Lusser A, Traja-
8. Squires JE, Patel HR, Nousch M, Sibbritt T, noski Z (2015) meRanTK: methylated RNA
Humphreys DT, Parker BJ, Suter CM, Preiss T analysis ToolKit. Bioinformatics 32
(2012) Widespread occurrence of (5):782–785
5-methylcytosine in human coding and
non-coding RNA. Nucleic Acids Res:gks144
Chapter 10
Abstract
The study of small RNAs (sRNAs) by next-generation sequencing (NGS) is challenged by bias issues during
library preparation. Several types of sRNAs such as plant microRNAs (miRNAs) carry a 20 -O-methyl
(20 -OMe) modification at their 30 terminal nucleotide. This modification adds another level of difficulty
as it inhibits 30 adapter ligation. We previously demonstrated that modified versions of the “TruSeq (TS)”
protocol have less bias and an improved detection of 20 -OMe RNAs. Here we describe in detail protocol
“TS5,” which showed the best overall performance. We also provide guidelines for bioinformatics analysis
of the sequencing data.
Key words Small RNA, Small RNA-seq, Bias, Library preparation, Next-generation sequencing,
NGS, 20 -O-methyl (20 -OMe) RNA, Plant microRNA, Plant miRNA
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
153
154 Erwin L. van Dijk and Claude Thermes
2 Materials
RNL2
NNNN + NNNN
NNNN
5’ HD adapter
5’ adapter ligaon NNNN + NNNN
RNL1
NNNN NNNN
RT
NNNN NNNN
NNNN NNNN
PCR
P5 In P7
NNNN NNNN
NNNN NNNN
NNNN NNNN
NNNN NNNN
NNNN NNNN
NNNN NNNN
Fig. 1 Schematic representation of sRNA library preparation protocol TS5. First, a preadenylated (App) 30
“high-definition” (HD) adapter is ligated to the sRNA. In contrast to the classical protocol, the HD adapter
carries four random nucleotides at its extremity. Then, a cleanup step is performed to remove excess
unligated 30 adapter. A 50 HD adapter is subsequently ligated. Reverse transcription is performed using a
primer complementary to the 30 adapter, followed by PCR amplification, during which the Illumina P5, P7, and
index (“In”) sequences are added. The modifications of protocol TS5 as compared to the standard TruSeq
protocol are summarized
50 30
Name Modification Modification Sequence 50 to 30 Purification
50 HD (TS5) 5AmMC6 [5AmMC6]GTTCAGAGTTCTACAGTCCGACGATCNrNrNrN (note that this oligo is a HPLC
adapter DNA-RNA chimeric; the three 30 terminal nucleotides are RNA)
30 HD (TS5) Phosphate 3AmMO [Phos]rNrNrNrNTGGAATTCTCGGGTGCCAAGG[3AaMO] (note that this oligo is a HPLC
adapter DNA-RNA chimeric; the four 50 terminal nucleotides are RNA)
RT primer GCCTTGGCACCCGAGAATTCCA HPLC
157
158 Erwin L. van Dijk and Claude Thermes
4. T4 RNA ligase 1.
5. Acid phenol:chloroform, pH 4.5.
6. Optional: QIAquick Nucleotide Removal Kit.
7. Qubit ssDNA Assay Kit.
8. Qubit Fluorometer.
9. 15% TBE-urea gel.
3 Methods
3.1 Isolation As an alternative to small RNA isolation by gel purification (step 1),
of Small RNAs a strategy using magnetic beads to enrich for small RNAs exists
([Link]
Protocol_for_miRNA_.pdf). We have not used this method our-
selves, but it is worth testing and if it works well it could signifi-
cantly simplify the protocol.
1. Extract total RNA from the sample of interest using phenol-
based reagents or any other method. Verify the RNA is of good
quality.
2. Pre-run a 15% TBE-urea gel for 15 min at 200 V.
3. While the gel is pre-running, mix 5–20μg of total RNA in a
5–15μL volume with an equal volume of formamide loading
dye in a 200μL PCR tube. Likewise, mix 10μL (200 ng) of
small-RNA ladder with an equal volume of formamide loading
dye. Incubate for 5 min at 65 C in a thermocycler with heated
lid, and then place the tubes immediately on ice.
4. Load the ladder and the sample on the same gel with at least
one lane between them and run at 200 V until the bromophe-
nol blue (dark blue) has migrated about two-thirds of the gel
length (approximately 40 min).
5. Prepare to elute the RNA from the gel as follows: puncture the
bottom of a nuclease-free 0.5 mL microcentrifuge tube with a
21-gauge needle (make three holes). Place the punctured
0.5 mL tube in a nuclease-free round-bottom 2 mL
microcentrifuge tube.
6. Remove the gel, and incubate at room temperature with 3μL
SYBR Gold Nucleic Acid Gel Stain (10,000 concentrate) in
30 mL water for 10–15 min.
7. View the gel on a “Dark Reader” transilluminator (it is strongly
recommended to avoid UV as this might damage the RNA) and
cut out the sample RNA between the 17 nt and the 29 nt ladder
bands. Transfer the gel piece to the prepared 0.5 mL tube from
step 5 above.
8. Centrifuge the 0.5 mL tube in the 2 mL tube in a microcen-
trifuge at maximum speed for 2 min. Remove the 0.5 mL tube,
which should be empty. See Note 1.
9. Add 300μL of nuclease-free 0.3 M NaCl to the 2 mL tube
containing the crushed gel and rotate for 2–3 h at room tem-
perature or at 4 C overnight (16 h).
10. Transfer the suspension of crushed gel pieces to a spin column
and centrifuge for 2 min at maximum speed in a
microcentrifuge.
160 Erwin L. van Dijk and Claude Thermes
Fig. 2 Isolation of small RNA and quality control. (a) Electrophoretic separation of Brassica napus total RNA
(10μg) on a 15% TBE-urea denaturing polyacrylamide gel. Image shows a small RNA ladder alongside the
samples as a molecular size marker. After migration the gel is stained and the RNA visualized on a
transilluminator. The region from 17 to 29 nucleotides is cut out (indicated by a red rectangle) and RNA
eluted. (b) Image shows the quality of the purified RNA as checked by capillary gel electrophoresis. Note that
this analysis provides information on the proportion of miRNA in the sample (93% in this case)
3.3.2 Elimination 1. Add 10μL of nuclease-free water and mix well. Add 6μL of 3 M
of Unligated 30 Adapter NaOAc pH 5.2 or “Adapter Depletion Solution” from the
NEXTflex V3 kit and mix well. Add 40μL of magnetic purifica-
tion beads and 60μL of isopropanol. Mix well and incubate for
5 min at room temperature.
2. Put the sample in a magnetic rack until the solution appears
clear. Remove and discard the supernatant.
162 Erwin L. van Dijk and Claude Thermes
3.3.5 PCR Amplification 1. Using the Kapa HiFi Hotstart PCR kit, add the following
reagents to the 12.5μL of RT reaction mixture: 10μL of PCR
polymerase buffer, 2μL of universal P5 primer (10μM;
Table 1), 2μL of P7-index primer (10μM; Table 1), 1μL of
12.5 mM dNTPs, 0.5μL of DNA polymerase, and 22μL of
water. Keep the reaction on ice until use.
2. Run the following PCR program: 98 C for 30 s, 11 cycles
(98 C for 10 s, 60 C for 30 s, and 72 C for 15 s), and 72 C
for 10 min (see Note 2). Keep the reaction at 4 C when
finished.
3.3.6 Gel Purification Gel purification of the final library product is a delicate step as a
and Sequencing number of additional products are formed that migrate close to the
desired library. It is important to not overload the gel as this will
increase the risk to contaminate the library with other species such
as adapter dimers. An example is shown in Fig. 3, where increasing
amounts of PCR-amplified library (from B. napus small RNA) were
loaded on the gel and the products corresponding to the expected
size (150 bp) were cut out (Fig. 3a). After elution, the purified
library was checked on a capillary gel electrophoresis chip; in addi-
tion to the expected 150 bp product, an increasing proportion of a
130 bp species, corresponding to adapter dimers, was observed as
increasing amounts of PCR product were loaded (Fig. 3b, c).
1. Run 5, 10, and 20μL of PCR product on a native 6% TBE gel
along with a suitable ladder (e.g., TrackIt 50 bp DNA ladder).
Run the gel for about 1 h at 145 V (until the bromophenol
blue reaches the bottom; this dye migrates at the 65 bp
position).
2. Remove the gel, and incubate with nucleic acid gel stain in
water for 10–15 min.
3. View the gel on a “Dark Reader” transilluminator and cut out
the library band at 150 bp. Prepare a system to elute the RNA
from gel as described in Subheading 3.1, step 5, and transfer
the gel piece to the 0.5 mL tube.
4. Centrifuge in a microcentrifuge at maximum speed for 2 min.
Remove the 0.5 mL tube, which should be empty now.
5. Add 300μL of nuclease-free water to the 2 mL tube containing
the crushed gel and rotate for at least 2 h at room temperature
or at 4 C overnight.
6. Transfer the suspension of crushed gel pieces in water to a spin
column and centrifuge for 2 min at maximum speed.
7. Add 1μL of glycogen (20μg/μL), 30μL of 3 M NaOAc pH 5.2,
and 975μL of ice-cold 100% ethanol. Centrifuge for 20 min at
max speed at 4 C.
164 Erwin L. van Dijk and Claude Thermes
Fig. 3 Gel purification of a B. napus small RNA library prepared following protocol TS5 and quality control. (a)
Image of a 6% native TBE gel showing increasing amounts of a PCR-amplified library from B. napus small
RNA; 2.5μL (a), 5μL (b), 10μL (c), or 20μL (d) PCR product. A 50 bp ladder is migrated alongside the samples.
PCR products migrating at the expected 150 bp position are isolated (red rectangle), DNA eluted, and purified.
(b) Representative quality control of the purified library. (c) Electropherogram representation of the same
analysis shown in (b). As can be seen, the 150 bp product is increasingly contaminated with adapter dimers
(~130 bp) as larger amounts of PCR product are loaded on gel
3.4 Data Analysis Sequences obtained from small RNA libraries can be analyzed using
the data analysis procedure described below (based on the Linux
operating system Ubuntu 16.04 LTS).
3.4.1 Treatment of Raw 1. Download the FASTQ sequence file(s) generated during the
Sequence Files sequencing run. If required, perform demultiplexing with
bcl2fastq2 (version V2.2.18.12; a manual can be downloaded
from the following link: [Link]
sequencing/sequencing_software/bcl2fastq-conversion-soft
ware/[Link]).
Improved Capture of 20 -O-Methyl RNAs by Small RNA-seq 165
4 Notes
Acknowledgments
References
1. Ghildiyal M, Zamore PD (2009) Small silenc- sequencing: implications of enzymatic manipu-
ing RNAs: an expanding universe. Nat Rev lation. J Nucleic Acids 2012:360358. https://
Genet 10(2):94–108. [Link] [Link]/10.1155/2012/360358
1038/nrg2504 4. van Dijk EL, Jaszczyszyn Y, Thermes C (2014)
2. Chang TC, Mendell JT (2007) microRNAs in Library preparation methods for next-
vertebrate physiology and human disease. generation sequencing: tone down the bias.
Annu Rev Genomics Hum Genet 8:215–239. Exp Cell Res 322(1):12–20. [Link]
[Link] 10.1016/[Link].2014.01.008
080706.092351 5. Munafo DB, Robb GB (2010) Optimization of
3. Zhuang F, Fuchs RT, Robb GB (2012) Small enzymatic reaction conditions for generating
RNA expression profiling by high-throughput representative pools of cDNA from small
Improved Capture of 20 -O-Methyl RNAs by Small RNA-seq 167
Abstract
20 -O-methylation (Nm) is an RNA modification commonly found on rRNA and snRNA, and at the mRNA
50 -cap, but has more recently been found internally on mRNA. The study of internal Nm modifications on
mRNA is in the early stages, but we have reported that this sort of Nm modification can regulate mRNA
abundance and translation. Although there are many methods to determine the presence of Nm on rRNA,
detecting Nm on specific mRNA transcripts is technically difficult because they are much less abundant than
rRNA. Some of these methods rely on the fact that Nm modification of RNA disrupts reverse transcription
reactions when performed at low dNTP concentrations. In this chapter, we describe our approach to using
quantitative PCR in conjunction with reverse transcription at low dNTPs, which is sensitive enough to
detect changes to Nm modification of mRNA.
Key words 20 -O-methylation, RNA modifications, Reverse transcription, Low dNTP, qPCR
1 Introduction
Mary McMahon (ed.), RNA Modifications: Methods and Protocols, Methods in Molecular Biology, vol. 2298,
[Link] © Springer Science+Business Media, LLC, part of Springer Nature 2021
171
172 Brittany A. Elliott and Christopher L. Holley
“N” “Nm”
Base Base
2’-O-methyltransferase
+ SAM
Fig. 1 20 -O-methylation (Nm) adds a methyl group to any nucleoside ribose (red arrow). The 20 -O-methyl-
transferase (i.e., fibrillarin and others) uses S-adenosyl methionine (SAM) as the methyl donor
A. B.
Gm1328 Um1326
Gm1328
Fig. 2 Detection of Nm sites by primer extension. (a) SuperScript III Reverse Transcriptase (SSIII RT) or (b) an
engineered 20 -O-methyl-sensitive DNA polymerase (Klen Taq V669L; KTQ) was used to detect Nm on 18S
rRNA by fluorescently labeled primer extension. RNAs from hearts of WT and Rpl13a snoRNA KO mice (lacking
U32A, U33, U34, and U35A) were processed with (a) SSIII RT and low dNTP or (b) Nm-sensitive KTQ. Rpl13a
snoRNA KO mice lack U33 that guides 18S Um1326 and they also lack U32A, which is one of the two guides
for 18S Gm1328 (U32B is a redundant snoRNA with the same guide sequence). Reduction of Gm1328 can be
clearly seen in KO mice from samples processed with SSIII with low dNTPs (a, arrow), but nearby Um1326 is
unable to be detected in WT or KO RNA (a). In samples processed with Nm-sensitive KTQ, both Um1326 and
Gm1328 are resolved in WT mice (b, arrows). In KO mice, loss of Um1326 is observed as well as partial loss of
methylation of Gm1328. Loss of Gm1328 is incomplete due to the redundant U32B snoRNA
OR
A. B. RT GSP RT odT
m RT RT primer(s) m RT primer(s)
5’ 3’ RNA target 5’ AAAAAA 3’ mRNA target
PCR primers qPCR primers
Fu FD R F R
More
Nm
Fig. 3 Detecting snoRNA-guided 20 -O-methylation of mRNA using reverse transcription under low-dNTP
conditions followed by qPCR (RTL-P). (a) Schematic of primer design for RTL-P as described in Dong et al.,
2012. (b) Schematic of primer design sensitive to the presence or absence of 20 -O-methylation on mRNA.
Oligo-dT or gene-specific primers (GSP) for target and housekeeping genes are used as an RT primer. qPCR
primers (F & R) are designed to be upstream of the putative Nm site. (c) Schematic of expected results at each
step of the assay. (1) Total RNA is extracted from control and fibrillarin (Fbl) or snoRNA KO cells or tissue.
(2) Under high-dNTP conditions, RT will read through Nm and methylated transcripts will be indistinguishable
from unmethylated ones. In low-dNTP conditions, RT will frequently pause at sites of Nm, resulting in
truncated transcripts that will not be amplified by PCR. (3) RT products are amplified by qPCR. In this example,
loss of Nm modification due to snoRNA KO leads to increased read-through during RT, which leads to
increased qPCR product