(Methods in Molecular Biology 1296) Mathieu Rederstorff (Eds.) - Small Non-Coding RNAs - Methods and Protocols-Humana Press (2015) PDF
(Methods in Molecular Biology 1296) Mathieu Rederstorff (Eds.) - Small Non-Coding RNAs - Methods and Protocols-Humana Press (2015) PDF
Small
Non-Coding
RNAs
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Mathieu Rederstorff
Université de Lorraine, Biopôle, CNRS UMR 7365, IMoPA, Vandoeuvre-lès-Nancy, France
Editor
Mathieu Rederstorff
Université de Lorraine, Biopôle, CNRS
UMR 7365, IMoPA
Vandoeuvre-lès-Nancy, France
The reality of pervasive transcription, or the fact that most if not the whole human genome
is being actively transcribed, is still debated. However, while only about 1.2 % of the human
genome encode for protein coding genes, it is well admitted now that a vast majority of
transcripts corresponds to non-protein coding transcripts, the so-called non-coding RNAs
(ncRNAs). While more and more (very) long non-coding RNAs (lincRNAs) are being
identified, with sizes up to several kilobases, most non-coding RNAs known to date are
rather small, with sizes ranging from 20 or less to 200 nucleotides. The discovery of the
fascinating class of microRNAs (miRNAs) about a decade ago as well as the availabilities of
genome sequences and progresses in next-generation sequencing techniques dramatically
boosted the attention of researchers in the field.
We now know that the many classes of small non-coding RNAs are involved in all bio-
logical pathways, such as RNA processing or modification, gene expression regulation at
the transcriptional or posttranscriptional levels, translation, or even protein secretion.
However, our actual knowledge is only the tip of the iceberg. Many questions are yet to be
answered, especially regarding the implications, direct or indirect, of small non-coding
RNAs with numerous disorders, suggesting more and more their possible and powerful
usage as diagnostic markers and/or therapeutic tools or targets.
Owing to their small sizes, tridimensional structures, low abundances, or differential
expression levels, small non-coding RNAs require customized dedicated protocols for their
identification and study, compared, for instance, to messenger RNAs (mRNAs).
Small Non-coding RNAs: Methods and Protocols is a laboratory protocols book dedi-
cated to biochemists or cellular/molecular biologists, already working in the field of RNA
biology or willing to start studying small non-coding RNAs in their projects. It describes
basic as well as more sophisticated, state-of-the-art methods to tackle all aspects of small
non-coding RNAs biology, from their identification or biogenesis to their use in therapeu-
tics. This survey of technologies will be of valuable help to all those willing to contribute
deciphering the numerous functions of small non-coding RNAs.
I thank all the authors and editors for their outstanding contributions to this volume.
v
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 235
Contributors
ix
x Contributors
Abstract
The revolution of miRNA discovery, in the early 2000s, shed a new light in the exciting field of small
non-coding RNAs. Since then, and owing to outstanding breakthroughs in RNomic techniques, novel
small non-coding RNA families have been regularly discovered, e.g., piRNAs, tiRNAs, and many others.
In this review, we provide a very succinct historical and functional overview on most prominent small
non-coding RNA families.
1 Introduction
1.1 Dawn of Small It has been more than 60 years that the first soluble cytoplasmic
Non-Coding RNAs small non-coding RNA (sncRNA), a yeast alanine transfer RNA
(tRNA), was identified and its primary structure determined [1, 2].
Together with ribosomal RNAs (rRNA), the nucleic acid moieties of
the ribosomes, tRNAs play a central role in protein synthesis, as the
adaptor molecules providing the correct amino acid in response to a
specific codon on the mRNA [3]. Only 20 years later were the first
nuclear RNAs uncovered: uridine-rich RNAs, or U-RNAs, were
observed to be very abundant, nucleus or sometimes even nucleolus
specific, and highly conserved [4, 5]; small nuclear RNAs (snRNA)
U1, U2, U4, U5, and U6 were later shown to be involved in splic-
ing, in association with several proteins within a ribonucleoprotein
particle (RNP) called spliceosome [6]. Base pairing among snRNAs
and between snRNAs and pre-messenger RNA (pre-mRNA) regions
account for the proper conformation of both spliceosome and pre-
mRNA, eventually leading to release of the introns and mature
mRNAs [7]. Interestingly, most small nucleolar RNAs (snoRNAs)
are processed from the released introns [8]. Within snoRNPs,
3
4 Guillaume Clerget et al.
1.2 miRNAs: In the early 1990s, the lin-14 gene was shown to be involved in
The Revolution! temporal development in the worm C. elegans. The lin-4 gene
product, most probably a polypeptide, was shown to negatively
regulate lin-14 expression by directly acting on the 3′ untranslated
region of its pre-mRNA [11]. In 1993, both Ambros and Ruvkun
labs discovered that the lin-4 gene product actually was a small
non-coding RNA, of 22 nucleotides in length in its mature form,
targeting the 3′ UTR of the lin-14 pre-mRNA several times, owing
to imperfect antisense base pairing [12, 13]. Unfortunately
enough, apparently, the lin-4 small RNA neither had additional
counterparts in worm nor was it found to be conserved in other
species. However, in 2000, Pasquinelli and coworkers identified
that let-7, another 22-nucleotide-long small temporal RNA
(stRNA) involved in C. elegans developmental timing, was con-
served in human and drosophila, anticipating the likely discovery
of many other small regulatory RNAs [14]. Revolution came less
than 1 year later, when Tuschl, Bartel, and Ambros labs reported,
in the same issue of Science, the observation of an abundant class
of novel small regulatory RNAs in worm, human, and drosophila,
termed since microRNAs (miRNAs) [15–17]. Even certain viruses
were next shown to encode their own miRNAs [18, 19].
It was rapidly observed that the miRNA maturation and assem-
bly machinery were the same as the one implicated in the forma-
tion of short interfering RNAs (siRNAs), involved in the RNA
interference (RNAi) pathway described in worm a couple of years
earlier by the 2006 Nobel Prize awardees Andrew Fire and Craig
Mello [20, 21] and even earlier in plants [22]. miRNA/siRNA
maturation, from a double-stranded precursor, is now well under-
stood [23] but still open to surprises; recently it was discovered
that miRNAs could be processed from other types of ncRNAs,
such as snoRNAs [24, 25].
In human, siRNAs, within the RNA-induced silencing complex
(RISC), mediate perfectly complementary mRNA target cleavages
ncRNAs Overview 5
1.3 Small Another sncRNA family, the Piwi-interacting RNAs (piRNA), dis-
Non-Coding RNAs covered about half a decade after miRNAs, also regulates transcrip-
Return! tion [31]. piRNAs are 27–29-nucleotide-long RNAs. They prevent
transposon dissemination in germinal cell lines, owing to an original
“ping-pong” mechanism [32, 33]. The smallest eukaryotic ncRNAs
known to date, the transcription initiation RNAs (tiRNAs), sized
17–18 nucleotides, are generated upon stalling or backtracking of
RNA polymerase II (RNAPII) near the transcription start sites
(TSSs) and might be involved in transcription initiation regulation as
well [34, 35]. TSSs appear to be the source of bunches of small and
long non-coding RNAs (lncRNAs) [36], such as transcription start
site-associated RNAs (TSSa-RNAs) and promoter-associated RNAs
(PASRs) in animals [37] or promoter upstream transcripts
(PROMPTs) [38] and cryptic/Xrn1-sensitive unstable transcripts
(CUTs/XUTs) in yeast [39–41]. While lncRNAs are generally
involved in epigenetic and chromatin modifications [42–44], the
functionality of short transcripts is debated. They might be involved
in maintaining chromatin into a transcriptionally active state or in
maintaining RNAPII available near TSSs [45].
1.4 Small Most if not all the human genome appears to be actively tran-
Non-Coding RNA: scribed [46–48]. Nevertheless, most transcripts rather belong to
Not the End the TUF (transcripts of unknown function) rather than to the
ncRNA family. Progresses in transcriptomics/RNomics enabled
the identification of thousands of short and long transcripts [49–55].
Many of them were attributed a function but it is only the top of
the iceberg. Whether every TUF is indeed functional is debated,
and experimental efforts will be mandatory to answer this
question.
There are no doubts that novel ncRNAs and novel ncRNA
families will still be discovered in the (next) future.
Acknowledgment
References
1. Holley RW, Apgar J, Doctor BP (1960) mentarity to lin-14. Cell 75(5):843–854, doi:
Separation of amino acid-specific “soluble”- 0092-8674(93)90529-Y [pii]
fraction ribonucleic acids. Ann N Y Acad Sci 13. Wightman B, Ha I, Ruvkun G (1993)
88:745–751 Posttranscriptional regulation of the heteroch-
2. Holley RW, Apgar J, Everett GA, Madison JT, ronic gene lin-14 by lin-4 mediates temporal
Marquisee M, Merrill SH, Penswick JR, Zamir pattern formation in C. elegans. Cell 75(5):855–
A (1965) Structure of a ribonucleic acid. 862, doi: 0092-8674(93)90530-4 [pii]
Science 147(3664):1462–1465 14. Pasquinelli AE, Reinhart BJ, Slack F,
3. Eddy SR (2001) Non-coding RNA genes and Martindale MQ, Kuroda MI, Maller B,
the modern RNA world. Nat Rev Genet Hayward DC, Ball EE, Degnan B, Muller P,
2(12):919–929. doi:10.1038/35103511, Spring J, Srinivasan A, Fishman M, Finnerty J,
35103511 [pii] Corbo J, Levine M, Leahy P, Davidson E,
4. Busch H, Reddy R, Rothblum L, Choi YC Ruvkun G (2000) Conservation of the
(1982) SnRNAs, SnRNPs, and RNA process- sequence and temporal expression of let-7 het-
ing. Annu Rev Biochem 51:617–654. erochronic regulatory RNA. Nature
doi:10.1146/annurev.bi.51.070182.003153 408(6808):86–89. doi:10.1038/35040556
5. Weinberg RA, Penman S (1968) Small molec- 15. Lagos-Quintana M, Rauhut R, Lendeckel W,
ular weight monodisperse nuclear RNA. J Tuschl T (2001) Identification of novel genes
Mol Biol 38(3):289–304, doi:0022- coding for small expressed RNAs. Science
2836(68)90387-2 [pii] 294(5543):853–858. doi:10.1126/sci-
6. Lerner MR, Steitz JA (1979) Antibodies to ence.1064921, 294/5543/853 [pii]
small nuclear RNAs complexed with proteins 16. Lau NC, Lim LP, Weinstein EG, Bartel DP
are produced by patients with systemic lupus (2001) An abundant class of tiny RNAs with
erythematosus. Proc Natl Acad Sci U S A probable regulatory roles in Caenorhabditis
76(11):5495–5499 elegans. Science 294(5543):858–862.
7. Lerner MR, Boyle JA, Mount SM, Wolin SL, doi:10.1126/science.1065062, 294/5543/
Steitz JA (1980) Are snRNPs involved in splic- 858 [pii]
ing? Nature 283(5743):220–224 17. Lee RC, Ambros V (2001) An extensive class
8. Matera AG, Terns RM, Terns MP (2007) Non- of small RNAs in Caenorhabditis elegans.
coding RNAs: lessons from the small nuclear Science 294(5543):862–864. doi:10.1126/
and small nucleolar RNAs. Nat Rev Mol Cell science.1065329, 294/5543/862 [pii]
Biol 8(3):209–220. doi:10.1038/nrm2124, 18. Nathans R, Chu CY, Serquina AK, Lu CC,
nrm2124 [pii] Cao H, Rana TM (2009) Cellular microRNA
9. Marmier-Gourrier N, Clery A, Schlotter F, and P bodies modulate host-HIV-1 interac-
Senty-Segault V, Branlant C (2011) A second tions. Mol Cell 34(6):696–709. doi:10.1016/j.
base pair interaction between U3 small nucleo- molcel.2009.06.003, S1097-
lar RNA and the 5′-ETS region is required for 2765(09)00394-3 [pii]
early cleavage of the yeast pre-ribosomal 19. Pfeffer S, Zavolan M, Grasser FA, Chien M,
RNA. Nucleic Acids Res 39(22):9731–9745. Russo JJ, Ju J, John B, Enright AJ, Marks D,
doi:10.1093/nar/gkr675, gkr675 [pii] Sander C, Tuschl T (2004) Identification of
10. Huttenhofer A, Kiefmann M, Meier-Ewert S, virus-encoded microRNAs. Science
O′Brien J, Lehrach H, Bachellerie JP, Brosius 304(5671):734–736. doi:10.1126/sci-
J (2001) RNomics: an experimental approach ence.1096781, 304/5671/734 [pii]
that identifies 201 candidates for novel, 20. Fire A, Xu S, Montgomery MK, Kostas SA,
small, non-messenger RNAs in mouse. Driver SE, Mello CC (1998) Potent and spe-
EMBO J 20(11):2943–2953. doi:10.1093/ cific genetic interference by double-stranded
emboj/20.11.2943 RNA in Caenorhabditis elegans. Nature
11. Wightman B, Burglin TR, Gatto J, Arasu P, 391(6669):806–811. doi:10.1038/35888
Ruvkun G (1991) Negative regulatory 21. Hutvagner G, McLachlan J, Pasquinelli AE, Balint
sequences in the lin-14 3′-untranslated region E, Tuschl T, Zamore PD (2001) A cellular func-
are necessary to generate a temporal switch tion for the RNA-interference enzyme Dicer in the
during Caenorhabditis elegans development. maturation of the let-7 small temporal
Genes Dev 5(10):1813–1824 RNA. Science 293(5531):834–838. doi:10.1126/
12. Lee RC, Feinbaum RL, Ambros V (1993) science.1062961, 1062961 [pii]
The C. elegans heterochronic gene lin-4 22. Napoli C, Lemieux C, Jorgensen R (1990)
encodes small RNAs with antisense comple- Introduction of a chimeric chalcone synthase
ncRNAs Overview 7
gene into petunia results in reversible 33. Siomi MC, Sato K, Pezic D, Aravin AA (2011)
co-suppression of homologous genes in trans. PIWI-interacting small RNAs: the vanguard of
Plant Cell 2(4):279–289. doi:10.1105/ genome defence. Nat Rev Mol Cell Biol
tpc.2.4.279, 2/4/279 [pii] 12(4):246–258. doi:10.1038/nrm3089,
23. Bartel DP (2004) MicroRNAs: genomics, nrm3089 [pii]
biogenesis, mechanism, and function. Cell 34. Taft RJ, Glazov EA, Cloonan N, Simons C,
116(2):281–297, doi: S0092867404000455 Stephen S, Faulkner GJ, Lassmann T, Forrest
[pii] AR, Grimmond SM, Schroder K, Irvine K,
24. Ender C, Krek A, Friedlander MR, Beitzinger Arakawa T, Nakamura M, Kubosaki A,
M, Weinmann L, Chen W, Pfeffer S, Rajewsky Hayashida K, Kawazu C, Murata M, Nishiyori
N, Meister G (2008) A human snoRNA with H, Fukuda S, Kawai J, Daub CO, Hume DA,
microRNA-like functions. Mol Cell 32(4):519– Suzuki H, Orlando V, Carninci P, Hayashizaki
528. doi:10.1016/j.molcel.2008.10.017, Y, Mattick JS (2009) Tiny RNAs associated
S1097-2765(08)00733-8 [pii] with transcription start sites in animals. Nat
25. Rother S, Meister G (2011) Small RNAs Genet 41(5):572–578. doi:10.1038/ng.312,
derived from longer non-coding RNAs. ng.312 [pii]
Biochimie 93(11):1905–1915. doi:10.1016/j. 35. Valen E, Preker P, Andersen PR, Zhao X, Chen
biochi.2011.07.032, S0300-9084(11)00290-2 Y, Ender C, Dueck A, Meister G, Sandelin A,
[pii] Jensen TH (2011) Biogenic mechanisms and
26. Tuschl T, Zamore PD, Lehmann R, Bartel DP, utilization of small RNAs derived from human
Sharp PA (1999) Targeted mRNA degradation protein-coding genes. Nat Struct Mol Biol
by double-stranded RNA in vitro. Genes Dev 18(9):1075–1082. doi:10.1038/nsmb.2091,
13(24):3191–3197 nsmb.2091 [pii]
27. Zamore PD, Tuschl T, Sharp PA, Bartel DP 36. Jacquier A (2009) The complex eukaryotic
(2000) RNAi: double-stranded RNA directs transcriptome: unexpected pervasive transcrip-
the ATP-dependent cleavage of mRNA at 21 tion and novel small RNAs. Nat Rev Genet
to 23 nucleotide intervals. Cell 101(1):25–33. 10(12):833–844. doi:10.1038/nrg2683,
doi: 10.1016/S0092-8674(00)80620-0 , nrg2683 [pii]
S0092-8674(00)80620-0 [pii] 37. Seila AC, Calabrese JM, Levine SS, Yeo GW,
28. Braun JE, Huntzinger E, Izaurralde E (2013) Rahl PB, Flynn RA, Young RA, Sharp PA
The role of GW182 proteins in miRNA- (2008) Divergent transcription from active
mediated gene silencing. Adv Exp Med Biol promoters. Science 322(5909):1849–1851.
768:147–163. doi:10.1007/978-1-4614- doi:10.1126/science.1162253, 1162253 [pii]
5107-5_9 38. Flynn RA, Almada AE, Zamudio JR, Sharp PA
29. Eulalio A, Tritschler F, Izaurralde E (2009) (2011) Antisense RNA polymerase II diver-
The GW182 protein family in animal cells: gent transcripts are P-TEFb dependent and
new insights into domains required for substrates for the RNA exosome. Proc Natl
miRNA-mediated gene silencing. RNA Acad Sci U S A 108(26):10460–10465.
15(8):1433–1442. doi:10.1261/rna.1703809, doi:10.1073/pnas.1106630108, 1106630108
rna.1703809 [pii] [pii]
30. Verdel A, Jia S, Gerber S, Sugiyama T, Gygi S, 39. Neil H, Malabat C, d’ Aubenton-Carafa Y, Xu
Grewal SI, Moazed D (2004) RNAi-mediated Z, Steinmetz LM, Jacquier A (2009)
targeting of heterochromatin by the RITS Widespread bidirectional promoters are the
complex. Science 303(5658):672–676. major source of cryptic transcripts in yeast.
doi:10.1126/science.1093686, 1093686 [pii] Nature 457(7232):1038–1042. doi:10.1038/
nature07747, nature07747 [pii]
31. Aravin A, Gaidatzis D, Pfeffer S, Lagos-
Quintana M, Landgraf P, Iovino N, Morris P, 40. van Dijk EL, Chen CL, d’Aubenton-Carafa Y,
Brownstein MJ, Kuramochi-Miyagawa S, Gourvennec S, Kwapisz M, Roche V, Bertrand
Nakano T, Chien M, Russo JJ, Ju J, Sheridan C, Silvain M, Legoix-Ne P, Loeillet S, Nicolas
R, Sander C, Zavolan M, Tuschl T (2006) A A, Thermes C, Morillon A (2011) XUTs are a
novel class of small RNAs bind to MILI protein class of Xrn1-sensitive antisense regulatory
in mouse testes. Nature 442(7099):203–207. non-coding RNA in yeast. Nature
doi:10.1038/nature04916, nature04916 [pii] 475(7354):114–117. doi:10.1038/
nature10118, nature10118 [pii]
32. Chuma S, Pillai RS (2009) Retrotransposon
silencing by piRNAs: ping-pong players mark 41. Xu Z, Wei W, Gagneur J, Perocchi F, Clauder-
their sub-cellular boundaries. PLoS Genet Munster S, Camblong J, Guffanti E, Stutz F,
5(12):e1000770. doi:10.1371/journal. Huber W, Steinmetz LM (2009) Bidirectional
pgen.1000770 promoters generate pervasive transcription in
8 Guillaume Clerget et al.
yeast. Nature 457(7232):1033–1037. Lowe TM, Wei CL, Ruan Y, Struhl K, Gerstein
doi:10.1038/nature07728, nature07728 [pii] M, Antonarakis SE, Fu Y, Green ED, Karaoz
42. Mercer TR, Dinger ME, Mattick JS (2009) U, Siepel A, Taylor J, Liefer LA, Wetterstrand
Long non-coding RNAs: insights into func- KA, Good PJ, Feingold EA, Guyer MS, Cooper
tions. Nat Rev Genet 10(3):155–159. GM, Asimenos G, Dewey CN, Hou M,
doi:10.1038/nrg2521, nrg2521 [pii] Nikolaev S, Montoya-Burgos JI, Loytynoja A,
43. Ponting CP, Oliver PL, Reik W (2009) Whelan S, Pardi F, Massingham T, Huang H,
Evolution and functions of long noncoding Zhang NR, Holmes I, Mullikin JC, Ureta-
RNAs. Cell 136(4):629–641. doi:10.1016/j. Vidal A, Paten B, Seringhaus M, Church D,
cell.2009.02.006, S0092-8674(09)00142-1 Rosenbloom K, Kent WJ, Stone EA, Batzoglou
[pii] S, Goldman N, Hardison RC, Haussler D,
Miller W, Sidow A, Trinklein ND, Zhang ZD,
44. Yazgan O, Krebs JE (2007) Noncoding but Barrera L, Stuart R, King DC, Ameur A,
nonexpendable: transcriptional regulation by Enroth S, Bieda MC, Kim J, Bhinge AA, Jiang
large noncoding RNA in eukaryotes. Biochem N, Liu J, Yao F, Vega VB, Lee CW, Ng P, Yang
Cell Biol 85(4):484–496. doi:10.1139/O07- A, Moqtaderi Z, Zhu Z, Xu X, Squazzo S,
061, o07-061 [pii] Oberley MJ, Inman D, Singer MA, Richmond
45. Aalto AP, Pasquinelli AE (2012) Small non- TA, Munn KJ, Rada-Iglesias A, Wallerman O,
coding RNAs mount a silent revolution in gene Komorowski J, Fowler JC, Couttet P, Bruce
expression. Curr Opin Cell Biol 24(3):333– AW, Dovey OM, Ellis PD, Langford CF, Nix
340. doi:10.1016/j.ceb.2012.03.006, S0955- DA, Euskirchen G, Hartman S, Urban AE,
0674(12)00037-3 [pii] Kraus P, Van Calcar S, Heintzman N, Kim TH,
46. Birney E, Stamatoyannopoulos JA, Dutta A, Wang K, Qu C, Hon G, Luna R, Glass CK,
Guigo R, Gingeras TR, Margulies EH, Weng Rosenfeld MG, Aldred SF, Cooper SJ, Halees
Z, Snyder M, Dermitzakis ET, Thurman RE, A, Lin JM, Shulha HP, Xu M, Haidar JN, Yu Y,
Kuehn MS, Taylor CM, Neph S, Koch CM, Iyer VR, Green RD, Wadelius C, Farnham PJ,
Asthana S, Malhotra A, Adzhubei I, Greenbaum Ren B, Harte RA, Hinrichs AS, Trumbower H,
JA, Andrews RM, Flicek P, Boyle PJ, Cao H, Clawson H, Hillman-Jackson J, Zweig AS,
Carter NP, Clelland GK, Davis S, Day N, Smith K, Thakkapallayil A, Barber G, Kuhn
Dhami P, Dillon SC, Dorschner MO, Fiegler RM, Karolchik D, Armengol L, Bird CP, de
H, Giresi PG, Goldy J, Hawrylycz M, Haydock Bakker PI, Kern AD, Lopez-Bigas N, Martin
A, Humbert R, James KD, Johnson BE, JD, Stranger BE, Woodroffe A, Davydov E,
Johnson EM, Frum TT, Rosenzweig ER, Dimas A, Eyras E, Hallgrimsdottir IB, Huppert
Karnani N, Lee K, Lefebvre GC, Navas PA, J, Zody MC, Abecasis GR, Estivill X, Bouffard
Neri F, Parker SC, Sabo PJ, Sandstrom R, GG, Guan X, Hansen NF, Idol JR, Maduro
Shafer A, Vetrie D, Weaver M, Wilcox S, Yu M, VV, Maskeri B, McDowell JC, Park M, Thomas
Collins FS, Dekker J, Lieb JD, Tullius TD, PJ, Young AC, Blakesley RW, Muzny DM,
Crawford GE, Sunyaev S, Noble WS, Dunham Sodergren E, Wheeler DA, Worley KC, Jiang
I, Denoeud F, Reymond A, Kapranov P, H, Weinstock GM, Gibbs RA, Graves T, Fulton
Rozowsky J, Zheng D, Castelo R, Frankish A, R, Mardis ER, Wilson RK, Clamp M, Cuff J,
Harrow J, Ghosh S, Sandelin A, Hofacker IL, Gnerre S, Jaffe DB, Chang JL, Lindblad-Toh K,
Baertsch R, Keefe D, Dike S, Cheng J, Hirsch Lander ES, Koriabine M, Nefedov M, Osoegawa
HA, Sekinger EA, Lagarde J, Abril JF, Shahab K, Yoshinaga Y, Zhu B, De Jong PJ (2007)
A, Flamm C, Fried C, Hackermuller J, Hertel Identification and analysis of functional elements
J, Lindemeyer M, Missal K, Tanzer A, Washietl in 1% of the human genome by the ENCODE
S, Korbel J, Emanuelsson O, Pedersen JS, pilot project. Nature 447(7146):799–816.
Holroyd N, Taylor R, Swarbreck D, Matthews doi:10.1038/nature05874
N, Dickson MC, Thomas DJ, Weirauch MT, 47. Clark MB, Amaral PP, Schlesinger FJ, Dinger
Gilbert J, Drenkow J, Bell I, Zhao X, Srinivasan ME, Taft RJ, Rinn JL, Ponting CP, Stadler
KG, Sung WK, Ooi HS, Chiu KP, Foissac S, PF, Morris KV, Morillon A, Rozowsky JS,
Alioto T, Brent M, Pachter L, Tress ML, Gerstein MB, Wahlestedt C, Hayashizaki Y,
Valencia A, Choo SW, Choo CY, Ucla C, Carninci P, Gingeras TR, Mattick JS (2011)
Manzano C, Wyss C, Cheung E, Clark TG, The reality of pervasive transcription. PLoS
Brown JB, Ganesh M, Patel S, Tammana H, Biol 9(7):e1000625. doi:10.1371/journal.
Chrast J, Henrichsen CN, Kai C, Kawai J, pbio.1000625, discussion e1001102,
Nagalakshmi U, Wu J, Lian Z, Lian J, 10-PLBI-PS-8040R3 [pii]
Newburger P, Zhang X, Bickel P, Mattick JS, 48. Clark MB, Choudhary A, Smith MA, Taft RJ,
Carninci P, Hayashizaki Y, Weissman S, Mattick JS (2013) The dark matter rises: the
Hubbard T, Myers RM, Rogers J, Stadler PF, expanding world of regulatory RNAs. Essays
ncRNAs Overview 9
Abstract
Alcoholic precipitation is a critical step to recover RNA of high purity. This chapter describes the principles
of alcoholic precipitation as well as a standard, basic protocol with key advices to observe, but numerous
variations on the theme are discussed. Indeed, several important parameters, such as the choice of salt,
alcohol, or carrier, have to be considered to improve the efficiency of precipitation and the yield of RNA
recovery.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_2, © Springer Science+Business Media New York 2015
11
12 Guillaume Clerget et al.
2 Materials
1. 100 % ethanol.
2. 70 % ethanol.
3. 100 % isopropanol.
4. 3 M sodium acetate, pH 5.2.
5. 5 M ammonium acetate.
6. 8 M lithium chloride.
7. 2 M sodium chloride.
8. Yeast tRNA (10–20 μg/ml).
9. Salmon sperm DNA (10–20 μg/ml).
10. Glycogen (50–150 μg/ml).
11. GlycoBlue (50–150 μg/ml) (Ambion).
12. Linear polyacrylamide (10–20 μg/ml).
13. 1 M MgCl2.
14. RNase-free, DEPC-treated water.
15. 1× TE buffer, pH 8.0: 10 mM Tris–HCl, pH 8.0, 1 mM
EDTA, pH 8.0.
3 Methods
3.1 Ethanol 1. Adjust sample volume to a minimum of 100 μl (see Note 7).
Precipitation 2. Add sodium acetate to the sample to a final concentration of
0.3 M (about 1/10 of volume) (see Notes 1, 3 and 8).
3. Add 2.5–3 volumes of ice-cold 100 % ethanol (see Note 2).
4. Mix thoroughly by inverting the tube or pipetting up and
down (see Note 9).
5. Incubate on ice for 15 min to 1 h (see Note 4).
6. Centrifuge at 12,000 × g for 30 min at 4 °C (see Note 5).
7. Carefully discard the supernatant without disturbing the pellet.
RNA Precipitation 13
4 Notes
References
1. Sambrook J, Fritsch EF, Maniatis T (1989) 6. Tracy S (1981) Improved rapid methodology
Molecular cloning, a laboratory manual, 2nd for the isolation oh nucleic acids from agarose
edn. Cold Spring Harbor Laboratory Press, gels. Prep Biochem 11:251–268
New York, NY, pp E10–E15 7. Gaillard C, Strauss F (1990) Ethanol precipita-
2. Zumbo P (2012) Ethanol precipitation. tion of dna with linear polyacrylamide as car-
Department of physiology and biophysics, rier. Nucleic Acids Res 18:378
Weill Cornell Medical College 8. Baugh LR et al (2001) Quantitative analysis of
3. Barlow J et al (2001) A simple method for the mRNA amplification by in vitro transcription.
quantitative isolation of undegraded high Nucleic Acids Res 29:E29
molecular weight ribonucleic acid. Biochem 9. Aruffo A, Seed B (1987) Molecular cloning of
Biophys Res Commun 13:61–66 a CD28 cDNA by a high-efficiency COS cell
4. Michelson AM, Orkin SH (1982) expression system. Proc Natl Acad Sci U S A
Characterization of the homopolymer tailing 84:8573–8577
reaction catalyzed by terminal deoxynucleoti- 10. Strauss F, Varshavsky A (1984) A protein binds
dyl transferase. J Biol Chem 257: to a satellite DNA repeat at three specific sites
14773–14782 that would be brought into mutual proximity
5. Wang QT et al (2002) Yeast tRNA as carrier in by DNA folding in the nucleosome. Cell 37:
the isolation of microscale RNA for global 889–901
amplification and expression profiling. 11. Zeugin J, Hartley JL (1985) Ethanol precipita-
Biotechniques 33:788–796 tion of DNA. Focus 7:1–2
Chapter 3
Abstract
Recent advances in high-throughput sequencing have shed some new light on the diversity of small
non-coding RNA (sncRNA) classes and their crucial role in gene regulation and diseases. RNA quantifica-
tion and control of RNA integrity are two key steps in sncRNA profiling. In this chapter, we will describe
different gold standard methods used to achieve both purposes before the use of the RNAs in downstream
applications.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_3, © Springer Science+Business Media New York 2015
17
18 Virginie Marchand and Christiane Branlant
2 Materials
2.1.2 Using 1. Any kind of fluorometer able to quantify RNA and DNA (and
a Fluorometer proteins) with high sensitivity (e.g., Qubit® 2.0 Fluorometer
(Life Technologies) which is the one we use).
2. Thin-walled polypropylene tubes of 500 μl compatible with
the fluorometer (e.g., Qubit® Assay Tube or Axygen® PCR-
05-C tubes, VWR).
3. RNA assay kit: Qubit® RNA Assay Kit (5–100 ng), Qubit®
RNA BR Assay Kit (20–1,000 ng), and Qubit® microRNA
Assay Kit (1–500 ng).
4. Optional: Qubit® dsDNA HS Assay Kit (0.2–100 ng) and
Qubit® Protein Assay Kit (0.25–5 μg).
Overview of the technical specifications for the Agilent RNA 6000 Nano, RNA 6000 Pico, and Small RNA kits
Agilent RNA 6000 Nano kit Agilent RNA 6000 Pico kit Agilent Small RNA kit
Samples analyzed/chip 12 11
Analysis range (nt) 20–6,000 6–150
Sample volume 1
Quantitative range 25–500 ng/μl (5–500 ng/μl) 0.2–5 ng/μl (0.05–5 ng/μl) 0.05–2 ng/μl for purified miRNA
(qualitative range) for total RNA for total RNA
25–250 ng/μl (25–250 ng/μl) 0.5–5 ng/μl (0.25–5 ng/μl)
for mRNA for mRNA
Reproducibility of 10 % CV 20 % CV
quantitation
Quantitation accuracy 20 % CV (for ladder as sample) 30 % CV (for ladder as sample) 25 % CV (for ladder as sample)
Buffer compatibility 100 mM Tris or 125 mM NaCl 50 mM Tris or 50 mM NaCl 10 mM Tris and 0.1 mM EDTA
or 15 mM MgCl2
Analysis run time (min) 30
Quantity and Quality of Small RNAs 21
3 Methods
3.1 RNA Carry out all procedures at room temperature unless otherwise
Quantification specified.
3.1.1 Using 1. Select the “Nucleic Acid” application from the main menu of
a Spectrophotometer the NanoDrop software installed on the PC driving the
spectrophotometer.
2. When the wavelength verification window appears, ensure that
the arm is down and click “OK.”
3. Select the type of sample to measure, in this case “RNA.”
4. Select “overlay spectra” to display multiple spectra at a time.
5. Prepare a blank: the buffer used for your sample but without
any trace of RNAs (e.g., RNase-free water or TE buffer).
6. Load 1 μl of the blank solution to the bottom pedestal, lower
the arm, and click on the “Blank” button.
7. Enter your sample name in the appropriate field, load 1 μl of it
to the bottom pedestal, lower the arm, and click “Measure”
(see Note 3).
8. Wipe the upper and lower pedestals using a dry wipe and pro-
ceed with the next sample.
9. Analyze the data obtained for your different RNA samples.
“Conc” is the concentration of your RNA sample based on the
absorbance at 260 nm. “260/280” is the ratio of the absor-
bances at 260 and 280 nm. This ratio is used to roughly assess
the purity of your RNA preparation. A ratio of 2 is generally
considered as corresponding to “pure” RNA. “260/230” is
the ratio of the absorbances at 260 and 230 nm. This ratio is
also used to assess RNA purity. For “pure” RNAs, it should be
in the range of 1.8–2.2 (see Note 4).
3.1.2 Using 1. Equilibrate all solutions of the appropriate kit at room tem-
a Fluorometer perature for at least 30 min before starting the experiments.
The kit provides the concentrated assay reagent, dilution buf-
fer, and pre-diluted standards (see Note 5).
2. Prepare the dye working solution by diluting the concentrated
assay reagent (200× concentrate in DMSO) 1:200 in dilution
buffer. Prepare 200 μl of working solution for each sample and
the two standards.
3. Prepare the two pre-diluted RNA standards annotated “C”
(0 ng/μl in TE buffer) and “D” (10 ng/μl of RNA in TE buf-
fer) by mixing 10 μl of standard with 190 μl of working
solution.
4. Add the working solution up to 200 μl to 1–20 μl of RNA
sample.
22 Virginie Marchand and Christiane Branlant
5. Vortex the tubes for 2 s and incubate them 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., “RNA” or
“microRNA”) for which you want to perform a new
calibration.
7. Press “Yes” to read new standards.
8. When indicated, insert the standard tube and press “Read.”
Standard #1 and #2 correspond to standards “C” and “D,”
respectively.
9. Once the calibration is done, insert each sample and press
“Read” to make the measurements.
10. Check that the values of your samples fall within the assay’s
range, and press “Calculate Stock Conc.”
11. To test if your RNA sample preparation is contaminated by
DNA or proteins, repeat the experiment with the DNA and/
or protein kits (see Note 6).
3.2.2 Capillary 1. Prepare the ladder provided in the kit: spin down the tube and
Electrophoresis transfer 10 μl to an 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 them at −70 °C. Before use, thaw one tube
and keep it on ice (Fig. 1a) (see Note 7).
2. Equilibrate all solutions of the kit at room temperature for at
least 30 min before starting the experiments (see Note 5).
3. Place 550 μl (Agilent RNA 6000 Nano or Pico kit) or 650 μl
(Agilent small RNA kit) of the gel matrix (red cap vial) into a
dedicated spin filter. Spin at room temperature for 10 min at
1,500 × g (Agilent RNA 6000 Nano or Pico kit) or for 15 min
at 10,000 × g (Agilent Small RNA kit) (Table 1) (Fig. 1b).
Quantity and Quality of Small RNAs 23
4-6 G
7-9 G
CS
CS
+ 65 µl gel
aliquot
Centrifuge for
10 min at 13,000 g at RT Load 9 µl of Conditioning Solution
G
G
1 µl Gel-dye mix ready
dye concentrate G
CS
G
5 µl ladder G
aliquot Mix by pipetting
CS
up and down
6 µl diluted ladder
1 µl ladder
Load 6 µl of Diluted RNA sample
G
CS
+ + +
12 empty
1.5 ml tubes
Fig. 1 Quick guide for RNA Pico Chip preparation and loading. (a) Ladder preparation, (b) gel preparation, (c)
gel-dye mix preparation, (d) samples and ladder preparation, and (e) RNA Pico Chip loading
24 Virginie Marchand and Christiane Branlant
18. Wait exactly for 30 s (Agilent RNA 6000 Nano or Pico kit) or
60 s (Agilent small RNA kit) and then release the clip.
19. Wait for 5 s until the plunger stops its upward move and pull it
slowly back to the 1 ml position.
20. Open the chip priming station and load 9 μl of gel-dye mix in
the other wells marked with a “G.”
21. Load 9 μl of the conditioning solution (not for the Agilent
RNA 6000 Nano kit) in the well marked “CS” (Fig. 1e).
22. Load 6 μl of the diluted ladder in the well marked with a ladder
(Fig. 1e).
23. Load 6 μl of the diluted RNA samples in the wells marked
1–11 (for Agilent RNA 6000 Pico and Small RNA kit) or 1–12
(for Agilent RNA 6000 Nano kit) (Fig. 1e).
24. Inspect the chip and make sure that no liquid is present on the
edges of the wells. If any, pipette it away (see Note 13).
25. Insert the chip in the Agilent 2100 Bioanalyzer.
26. Carefully close the lid. The electrodes of the cartridge will
enter into the wells of the chip (see Note 14).
27. The 2100 expert software screen shows that you have inserted
a chip by displaying a chip icon on the top-left corner of the
screen.
28. Select the appropriate assay you want to perform (e.g., eukary-
otic total RNA pico series II).
29. Press “Start” to begin the chip run (see Note 15).
30. After the run, immediately remove the chip and clean the elec-
trodes (see Note 8).
31. Analyze the results of the chip (Fig. 2).
4 Notes
0 (nt)
25 500 4000 [nt] 4000
Total RNA extracted from insect cells (RIN=NA)
[FU] 2000
500 1000
500
200
0
25
25 500 4000 [nt]
RIN NA 8.7
b
Total RNA extracted from human PBMC cells (RIN=9.3)
[FU]
18S rRNA 28S rRNA re
RN
A xtu
200 mi RNA
otal N A i
m
sRNAs nt t tR ified
100 ma eas r
hu y pu
0
25 500 4000 [nt]
yeast tRNA mixture (RIN=2.6)
[FU]
1000 (nt)
500
4000
0
25 500 4000 [nt] 2000
Synthetic miRNA (RIN=2.6) 1000
[FU]
500
200
25
0 RIN 9.3 2.6 2.6
25 500 4000 [nt]
(nt)
0
4 20 60 100 150 [nt] 150
[FU] Synthetic miRNA (RIN=2.6) 100
200
80
100 60
40
0 20
4 20 60 100 150 [nt] 4
miRNA tRNA
Fig. 2 Examples of RNA profiles obtained using the Agilent 2100 Bioanalyzer. (a) High-quality insect or human
total RNA obtained after Trizol extraction analyzed on an RNA Pico Chip. The human total RNA trace profile is
typical of a high-quality RNA since the 28S and 18S rRNA peaks are present and dominant. An RIN (RNA integ-
rity number) of 8.7 was attributed to the RNA sample, which is indicative of good quality. Note the presence of
Quantity and Quality of Small RNAs 27
Fig. 2 (continued) abundant small RNAs including 5.8S and 5S rRNAs, tRNAs, and other non-coding RNAs
ranging from 25 to 200 nt. The insect total RNA trace profile is also typical of a high-quality RNA sample, even
though it looked like degraded and no RIN could be attributed since the 28S rRNA peak is absent. Indeed, upon
heat denaturation, the insect 28S rRNA is cleaved, leading to fragments migrating with the 18S rRNA. (b) RNA
trace profiles obtained on a Pico RNA chip for a yeast total tRNA mixture and a synthetic miRNA sample (21 nt)
were compared to a human PBMC total RNA preparation. As expected, yeast tRNA mixture and synthetic
miRNA both migrate as the small RNAs of the human total RNA preparation. (c) RNA trace profiles obtained on
a Small RNA chip for a yeast total tRNA preparation and a synthetic miRNAs sample (21 nt) were compared to
a human PBMC total RNA preparation. The human PBMC total RNA preparation leads to three major peaks: one
between 50 and 80 nt, corresponding mainly to tRNAs; one around 90 nt corresponding to snoRNAs; and one
around 140 nt corresponding to the 5.8S rRNA. 5S rRNA corresponds to a small pic at 120 nt. Though miRNAs
are detected in this sample, they represent too small portion of total RNAs and are therefore not visible on the
RNA trace profile. However, using other techniques, such as NGS, we confirmed the presence of miRNAs in our
total RNA preparation
28 Virginie Marchand and Christiane Branlant
Acknowledgments
References
1. Rave N, Crkvenjakov R, Boedtker H (1979) integrity number for assigning integrity values to
Identification of procollagen mRNAs transferred RNA measurements. BMC Mol Biol 7:3
to diazobenzyloxymethyl paper from formalde- 4. Winnebeck EC, Millar CD, Warman GR (2000)
hyde agarose gels. Nucleic Acids Res 6(11): Why does insect RNA look degraded? J Insect
3559–3567 Sci 10:159
2. Becker C, Hammerle-Fickinger A, Riedmaier I, 5. Aranda PS, LaJoie DM, Jorcyk CL (2012)
Pfaffl MW (2010) mRNA and microRNA qual- Bleach gel: a simple agarose gel for analyzing
ity control for RT-qPCR analysis. Methods RNA quality. Electrophoresis 33(2):366–369
50(4):237–243 6. Wilfinger WW, Mackey K, Chomczynski P (1997)
3. Schroeder A, Mueller O, Stocker S, Salowsky R, Effect of pH and ionic strength on the spectro-
Leiber M, Gassmann M, Lightfoot S, Menzel W, photometric assessment of nucleic acid purity.
Granzow M, Ragg T (2006) The RIN: an RNA Biotechniques 22(3):474–476, pp 478–481
Chapter 4
Abstract
We prepared total RNA from the Gram-positive soil bacterium Bacillus subtilis by different RNA extraction
procedures to compare their suitability for Northern blot detection of tiny RNAs (~14-mers) or RNAs of
intermediate size (100–200 nt) in terms of signal quality, intensity, and reproducibility. Our analysis
included two hot phenol methods and two TRIzol extraction procedures. We found that signal intensity/
detection sensitivity makes the key difference. Total RNAs prepared by the hot phenol method comprise
the length spectrum from tRNAs to large ribosomal RNAs. Larger RNAs are less abundant in TRIzol
preparations which instead enrich for RNAs of tRNA size and smaller. Thus, hot phenol methods are the
choice for the detection of intermediate-sized and longer RNAs, whereas TRIzol extraction procedures are
more suited for the detection of tiny RNAs.
Key words RNA isolation, Tiny RNA, Intermediate-sized RNA, Hot phenol, TRIzol, Northern blot,
EDC crosslinking
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_4, © Springer Science+Business Media New York 2015
29
30 Katrin Damm et al.
of the 6S-1 RNA (bsrA) gene [1] using four different RNA
extraction protocols: two phenol- and two TRIzol-based methods.
We compare the four RNA preparations with respect to Northern
blot detection of three RNA species: the 5S rRNA (115 nt) that is
often used as an RNA loading control, the regulatory 6S-1 RNA
(190 nt long), and a tiny 14-nt transcript, termed “pRNA” for
“product RNA”, synthesized by the B. subtilis housekeeping RNA
polymerase (σA-RNAP) using the 6S-1 RNA as a template, particu-
larly under outgrowth conditions when stationary cells enter a new
exponential growth phase [2]. We conclude that phenol methods
are better suited for the extraction of large RNAs (>100 nt),
whereas TRIzol methods are the methods of choice to enrich tiny
RNAs (~14 nt).
2 Materials
3 Methods
3.2 RNA Isolation 1. Resuspend harvested B. subtilis cells from Subheading 3.1 in
1.6 ml extraction buffer on ice, and split the cell suspension
3.2.1 Method 1:
equally into two 2-ml tubes. The following steps refer to 800 μl
Extracting RNA Three
samples.
Times with Hot Phenol
2. Add 75 μl lysozyme solution per reaction tube and incubate
for 10 min.
32 Katrin Damm et al.
Fig. 1 Detection of RNAs (115 nt and 190 nt) by Northern blot analysis using
10 % denaturing PAA gels, followed by membrane transfer and immobilization by
UV crosslinking. For each extraction method, three independent RNA prepara-
tions (a, b, and c) were processed to assess signal fluctuations between individual
RNA preparations. The experiment revealed that extraction of intermediate-sized
RNAs is more efficient with phenol methods 1 and 2 than with the TRIzol meth-
ods 3 and 4. (a) The X-ray film was exposed for 2 min. 6S-1 RNA is detectable
with all four extraction methods, although signals were stronger for RNAs pre-
pared according to methods 1 and 2. However, the 5S rRNA loading control was
hardly visible after 2 min when using the TRIzol methods 3 and 4. (b) After 10 min
of film exposure, 6S-1 RNA and 5S rRNA signals became also visible for the
TRIzol RNA preparations. However, for method 4 compared to method 3, the 5S
rRNA controls showed reduced intensities and stronger fluctuations between
individual samples. The signal above 6S-1 RNA is the 201-nt long 5′-precursor
transcript of 6S-1 RNA [2]
Fig. 2 Northern blot detection of tiny RNAs (~14 nt) after separation on 10 %
native PAA gels (for details, see Chapter 5 in this issue). In general, ~14-mers
were enriched in RNA preparations using TRIzol as extraction reagent. (a) RNA
fixation with EDC crosslinking. Both TRIzol methods gave rise to prominent sig-
nals specific to ~14-meric pRNA transcripts synthesized in vivo by B. subtilis
σA-RNAP using 6S-1 RNA as the template. Specificity was inferred from the loss
of signal in RNA prepared from the 6S-1 RNA knockout strain (ΔbsrA). Method 2
resulted in a fainter and more diffuse signal, whereas only a very faint signal was
seen with RNA prepared by method 1. M, marker: 0.25 ng of a synthetic 6S-1
RNA-specific, 14 nt long pRNA (p146S-1, 5′-pGUU CGG UCA AAA CU-3′). Note that
p146S-1 used marker carried a 5′-monophosphate terminus, and in vivo synthe-
sized pRNAs detected in the other lanes were primary transcripts with 5′-tri-
phosphate ends. (b) RNA fixation with UV crosslinking. The same trends as in
panel A were observed, but the signals had a somewhat reduced intensity and
less distinct appearance
19. Let stand for 5 min at room temperature. Remove the ethanol
solution and air-dry the RNA pellet for about 10–15 min
(see Note 4).
20. Dissolve the RNA pellet in 40 μl of ddH2O and store the
RNA at −20 °C or −80 °C for longer periods (see Notes 5–9;
Figs. 1 and 2).
3.2.3 Method 3: RNA 1. Add 1 ml TRIzol reagent to the bacterial cell pellets from
Extraction Using TRIzol® Subheading 3.1 and resuspend the cells by pipetting up and
down to accelerate lysis (see Note 10).
2. Incubate for 5 min at 4 °C on ice and transfer 1 ml of lysate to
a new 2-ml tube.
3. Add 200 μl of chloroform per 1 ml of TRIzol® used for lysis
and vortex the sample thoroughly. Chill on ice for 15 min.
4. Centrifuge the samples at 15,700 × g for 15 min to separate the
phases (see Note 11).
5. Carefully pipette about 600 μl of aqueous phase into a new
tube (see Note 12).
6. To precipitate the total RNA, add 600 μl of 100 % isopropanol
to the aqueous phase, mix, and keep the sample at least for 1 h
at −20 °C.
7. Centrifuge at 15,500 × g for 15 min at 4 °C.
8. Remove and discard the supernatant (see Note 13).
9. Add 200 μl of precooled 75 % ethanol, vortex gently, and cen-
trifuge at 15,500 × g for 10 min at 4 °C.
10. Discard the supernatant, air-dry the pellet for 5–10 min, and
dissolve the RNA pellet in 20 μl of ddH2O (see Note 4).
11. If not used immediately, store the RNA at −20 °C or −80 °C
for extended periods (see Notes 5, 6, 8, 14, and 15) (Figs. 1
and 2).
3.2.4 Method 4: RNA 1. Centrifuge the 15-ml tubes containing the Phase Lock Gel for
Extraction Using 5 min at 8,200 × g to pellet the gel.
a Combination of TRIzol® 2. Add 1 ml of TRIzol® to the bacterial cell pellet from
and Phase Lock Gel Subheading 3.1 and resuspend the cells by pipetting up and
down several times to accelerate lysis (see Note 10).
3. Incubate for 5 min at 4 °C on ice and pipette 1 ml of the lysate
onto the Phase Lock Gel matrix.
4. Invert the tube several times by hand.
5. Add 200 μl of chloroform per 1 ml of TRIzol® used for lysis
and invert thoroughly several times. Incubate for 15 min at
4 °C on ice.
36 Katrin Damm et al.
4 Notes
Acknowledgment
References
1. Beckmann BM, Grünweller A, Weber MH, and in vitro analysis of 6S RNA-templated short
Hartmann RK (2010) Northern blot detection transcripts in Bacillus subtilis. RNA Biol 8(5):839–
of endogenous small RNAs (approximately 849. doi:10.4161/rna.8.5.16151, 16151 [pii]
14 nt) in bacterial total RNA extracts. Nucleic 3. Kroczek RA, Siebert E (1990) Optimization of
Acids Res 38(14):e147. doi:10.1093/nar/ northern analysis by vacuum-blotting, RNA-
gkq437, gkq437 [pii] transfer visualization, and ultraviolet fixation.
2. Beckmann BM, Burenina OY, Hoch PG, Kubareva Anal Biochem 184(1):90–95, doi: 0003-2697
EA, Sharma CM, Hartmann RK (2011) In vivo (90)90017-4 [pii]
Part II
Abstract
Successful detection of very small RNAs (tiny RNA, ~14 nt in length) by Northern blotting is dependent
on improved Northern blot protocols that combine chemical crosslinking of RNA with 1-ethyl-3-(3-
dimethylaminopropyl)-carbodiimide (EDC) to positively charged membranes, the use of native polyacryl-
amide gels, and the development of highly sensitive and specific probes modified with locked nucleic acids
(LNA). In this protocol, we show that Northern blot detection of tiny RNAs with 5′-digoxigenin-labeled
DNA/LNA mixmer probes is a highly sensitive and specific method and, in our hands, more sensitive than
using a corresponding DNA/LNA mixmer probe with a 5′-32P-end label.
Key words Northern blot, EDC crosslinking, UV crosslinking, Digoxigenin, LNA, Native PAGE
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_5, © Springer Science+Business Media New York 2015
41
42 Katrin Damm et al.
2 Materials
2.4 Hybridization 1. RNA p146S-1: 5′-GUU CGG UCA AAA CU-3′; HPLC purified
and Detection and desalted, with 5′-OH ends.
Procedures 2. RNA p156S-2, 5′-AAA GGU UAA AAC UUA-3′; RNA p206S-2,
5′-AAA GGU UAA AAC UUA AUU CA-3′; both RNAs HPLC
purified and desalted, with 5′-OH ends.
3. Probe for p146S-1: 5′-DIG-aGt tTt gAc cGa Ac-3′. Probes for
6S-2 pRNAS: 5′-DIG-gTt tTa aCc tTt-3′ or 5′-taa gTt tTa acC
tTt-3′ for 5′-32P-end labeling. DNA residues appear in lower-
and LNA residues in uppercase letters (oligonucleotides obtained
from Exiqon, HPLC purified and desalted).
4. Hybridization solution: DIG Easy Hyb Granules (Roche
Diagnostics) (see Note 1) [4].
5. Hybridization oven.
6. 20× SSC: 3 M NaCl, 0.3 M sodium citrate, pH 7.0.
7. Stringent wash buffer I: 2× SSC, 0.1 % (w/v) SDS.
8. Stringent wash buffer II: 0.1× SSC, 0.1 % (w/v) SDS.
44 Katrin Damm et al.
3 Methods
3.1.2 Tiny RNAs (~14 nt) 1. Mix 6 μg of total RNA with one volume of 2× native loading
buffer.
2. Incubate the mixture at 98 °C for 3 min and put on ice
immediately.
Northern Blotting of Tiny RNAs 45
3. Load the samples on a 10 % native PAA gel and run the gel
(15 cm wide, 20 cm long, 1 mm thick) in 1× TBE at 15 mA
(see Note 3).
4. Stop electrophoresis when bromophenol blue has reached the
second half to two-thirds of the gel.
5. After electrophoresis, soak the gel in 0.5× TBE for 5 min.
6. For transfer, proceed as in steps 5 and 6 of Subheading 3.1.1.
7. After putting the cathode plate on top, run the transfer over-
night at 0.27 mA/cm2 (see Notes 2 and 4).
3.2 UV and EDC 1. Put the membrane on a glass plate with RNA pointing upward.
Crosslinking 2. Fix RNA by UV crosslinking with 120 mJ/cm2.
for Fixation of RNAs
3. Wrap in saran film and store at 4 °C until use.
on Membranes
3.2.1 UV Crosslinking
3.2.2 EDC Chemical 1. Place the membrane on a Whatman paper soaked with EDC
Crosslinking solution (see Note 5).
2. Wrap in saran film and incubate for 2 h at 60 °C.
3. Carefully wash the membrane with ddH2O for 5–10 min.
4. Wrap in saran film and store at 4 °C until use (see Note 6).
3.3 Probe Generation 1. Mix 1 μg of linear plasmid DNA or PCR template, 2 μl of 10×
by T7 Transcription T7 transcription buffer, 2 μl of 10× DIG labeling mix, and
40 U of T7 RNA polymerase.
2. Add ddH2O to 20 μl and incubate at 37 °C.
3. After 1 h, add 2 μl of T7 RNA polymerase and incubate for
another 1 h at 37 °C.
4. Stop the transcription reaction by adding 2 μl of 200 mM
EDTA and store at −20 °C.
3.4.3 Detection of RNAs Carry out all washing steps under slow shaking at room
Hybridized to Digoxigenin- temperature.
Labeled Probes
1. Wash the membrane twice for 5 min in stringent wash
buffer I.
2. Wash the membrane twice for 15 min in stringent wash buffer II.
3. Wash the membrane for 2–5 min in ddH2O.
4. Incubate the membrane in 1× blocking solution for 30 min.
5. Incubate the membrane in antibody solution for at least
30 min.
6. Wash the membrane twice for 15 min in 1× wash buffer.
7. Incubate the membrane in 1× detection buffer for 5 min.
8. Cut an appropriate piece of plastic tube film; additionally cut
one of the long sides and spread the plastic film as a single layer
on the bench.
9. Put the membrane on the film with RNA pointing upward.
10. Add 5 ml of detection buffer including 2.5 μl of CDP-Star
(see Note 10) until the membrane is homogeneously covered
with solution. Fold back the plastic film to cover the top of
the membrane and seal the plastic bag at the three open edges
(see Note 11).
11. Avoid air bubbles and incubate for 5 min to allow a robust
chemical signal to appear.
12. Place an X-ray film on top of the membrane bag and expose for
5–60 min, depending on the signal. For 5S rRNA and 6S-1
RNA detection, the first film exposition is 10 min, whereas
40–60 min are usually applied to the initial detection of tiny
RNAs. If signals are too weak or overexposed, a different time
is used for a second film exposition (see Note 12) (Fig. 1).
Northern Blotting of Tiny RNAs 47
Fig. 1 Northern blot analysis. (a) Detection of the 5S rRNA (115 nt) and the 6S-1 RNA (190 nt) using 10 %
denaturing PAA gels, followed by transfer on a nylon membrane and immobilization by UV crosslinking. Total
RNAs from stationary phase cells of B. subtilis PY79 were extracted either by the hot phenol/cold phenol
method or the TRIzol method (see Chapter 4). For each extraction method, three independent RNA preparations
(a, b, and c) were loaded onto the gel. For more details, see Chapter 4. (b) Detection of tiny RNAs (~14 nt) after
separation on a 10 % native PAA gel, transfer to a nylon membrane, and RNA fixation either by EDC or UV
crosslinking. As a control, total RNA from a B. subtilis PY79 mutant strain lacking the gene encoding for the
6S-1 RNA (ΔbsrA) was analyzed in parallel
3.4.4 Dot Blot Analysis 1. Spot 1 μl drops of varying amounts (2 pg–10 ng in ddH2O) of
to Compare Tiny RNA chemically synthesized RNA oligonucleotides carrying 5′-OH
Detection Sensitivity Using ends (p146S-1, p156S-2 or p206S-2) in a defined pattern onto a
5′-DIG- Versus nylon membrane (~10 × 4 cm).
5′-32P-Labeled Probes 2. Air-dry and immobilize the pRNA oligonucleotides by EDC
crosslinking as described in Subheading 3.2.2.
3. Denature 300 pmol of a 5′-DIG-LNA/DNA mixmer probe or
2 × 106 Cherenkov cpm of a 5′-32P-end-labeled DNA/LNA
mixmer probe for 3 min at 98 °C and put immediately on ice.
4. Pre-hybridize and hybridize the membrane as described in
Subheading 3.4.2.
5. In the case of the 5′-DIG-labeled probe, perform detection as
described in Subheading 3.4.3 (see Note 11).
6. For detection using the 5′-32P-labeled probe, proceed to step 7
after the hybridization step.
48 Katrin Damm et al.
4 Notes
Fold back the plastic film to cover the top of the membrane.
When the plastic film contacts the membrane surface, capillarity
leads to the homogeneous spreading of detection buffer/
CDP-Star solution on the membrane. In this case, sealing of
the plastic film is not necessary.
12. Take care that the membrane does not run dry after detection
if subsequent stripping is intended.
13. Comparison of tiny RNA detection using a 5′-DIG- versus an
almost identical 5′-32P-labeled oligonucleotide probe is illus-
trated in Fig. 2. Apart from the additional methodological effort
of the digoxigenin/chemiluminescence detection system, many
laboratories use 32P-labeled probes in Northern blot experi-
ments to be able to quantify signals by phosphorimaging.
Reported advantages of the digoxigenin detection system are
lower background signals and reduced exposure times [7]. Here
we directly compared detection of tiny RNAs by LNA/DNA
mixmer probes that were either 5′-DIG- or 5′-32P-end-labeled.
The results illustrated in Fig. 2 support the good sensitivity and
specificity of the digoxigenin/chemiluminescence system, as
inferred from low background signals obtained with the p146S-1
control RNA oligonucleotide. In our hands, the sensitivity of
tiny RNA detection was reduced when using the 5′-32P-end-
labeled LNA/DNA mixmer probe (Fig. 2b). Note that such
probes should have at least two DNA residues at the 5′ end for
efficient 5′ end-labeling with T4 polynucleotide kinase and
[γ-32P]-ATP. 5′ end-labeling of oligonucleotides with 5′-terminal
LNA residues has been very inefficient in our hands.
Acknowledgment
References
1. Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee piRNA by northern blot. Nucleic Acids Res
DH, Nguyen JT, Barbisin M, Xu NL, Mahuvakar 35(8):e60. doi:10.1093/nar/gkm112, gkm112
VR, Andersen MR, Lao KQ, Livak KJ, Guegler [pii]
KJ (2005) Real-time quantification of microR- 3. Varallyay E, Burgyan J, Havelda Z (2008)
NAs by stem-loop RT-PCR. Nucleic Acids Res MicroRNA detection by northern blotting
33(20):e179. doi:10.1093/nar/gni178, using locked nucleic acid probes. Nat Protoc
33/20/e179 [pii] 3(2):190–196. doi:10.1038/nprot.2007.528,
2. Pall GS, Codony-Servat C, Byrne J, Ritchie L, nprot.2007.528 [pii]
Hamilton A (2007) Carbodiimide-mediated 4. Beckmann BM, Grünweller A, Hartmann RK
cross-linking of RNA to nylon membranes (2014) Northern blot detection of endogenous
improves the detection of siRNA, miRNA and small RNAs. In: Hartmann RK, Bindereif A,
Northern Blotting of Tiny RNAs 51
Schön A, Westhof E (eds) Handbook of RNA 7. Grünweller A, Müller PK (1997) Low back-
biochemistry, 2nd edn. Wiley-VCH, Weinheim, ground and short exposure times in mapping of
pp 89–103 DNaseI-hypersensitive sites using digoxigenin-
5. Beckmann BM, Grünweller A, Weber MH, labeled probes. Anal Biochem 247(1):172–175.
Hartmann RK (2010) Northern blot detection of doi:10.1006/abio.1997.2044, S0003-2697(97)
endogenous small RNAs (approximately 14 nt) 92044-3 [pii]
in bacterial total RNA extracts. Nucleic Acids 8. Burenina OY, Hoch PG, Damm K, Salas M,
Res 38(14):e147. doi:10.1093/nar/gkq437, Zatsepin TS, Lechner M, Oretskaya TS,
gkq437 [pii] Kubareva EA, Hartmann RK (2014) Mechanistic
6. Pall GS, Hamilton AJ (2008) Improved northern comparison of Bacillus subtilis 6S–1 and 6S–2
blot method for enhanced detection of small RNAs–commonalities and differences. RNA
RNA. Nat Protoc 3(6):1077–1084. doi:10.1038/ 20(3):348–359. doi:10.1261/rna.042077.113,
nprot.2008.67, nprot.2008.67 [pii] rna.042077.113 [pii]
Chapter 6
Abstract
Most annotated genomes show a large number of sense–antisense transcripts that can generate double-
stranded RNAs. We describe a method to clone these dsRNAs from total RNA preparations.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_6, © Springer Science+Business Media New York 2015
53
54 Manli Shen et al.
Fig. 2 Overview of the method. (a) The individual steps are shown. (b) Structure of the cDNA generated through
cloning. The 5′ ends of the former dsRNA are indicated by arrows. Stars indicate a 5-amino C6 modification at
the 5′ end. (c) Mechanism of the ligation reaction using an adenylated (preactivated) oligonucleotide. B the
base at the 3′ end ribose. C first base of linker 1
56 Manli Shen et al.
Fig. 2 (continued)
2 Materials
Use molecular biology grade reagents only. Keep all solutions and
working area ribonuclease-free.
2.1 RNA Antisense 1. DNA template for RNA antisense probe transcription (see
Probe Transcription Note 1).
2. 10× transcription reaction buffer: 500 mM Tris–HCl, pH 7.5,
150 mM MgCl2, 50 mM DTT, 20 mM spermidine.
dsRNA Cloning 57
32
2.2 RNase Digestion 1. P-labeled RNA antisense probe.
2. Total RNA.
3. Yeast RNA.
4. 5× hybridization stock solution: 200 mM PIPES, pH 6.4, 2 M
NaCl, 5 mM EDTA. Prepare 1× working solution using one
part of stock solution plus four parts of freshly deionized
formamide.
5. RNase T1 (1,000 units/μl) and RNase A (1 mg/ml).
6. RNA digestion buffer: 10 mM Tris–HCl, pH 7.5, 300 mM
NaCl, 5 mM EDTA.
7. Antarctic Phosphatase (New England Biolabs).
8. 20 % SDS (w/v).
9. Proteinase K (20 mg/ml).
10. GlycoBlue (Life Technologies).
11. Phenol/chloroform.
12. Ethanol (70 and 100 %).
13. 42 °C water bath.
14. RNA loading buffer (80 % deionized formamide, 1 mM EDTA,
0.1 % bromophenol blue, 0.1 % xylene cyanol).
15. RNA Decade marker (Life Technologies).
16. Urea.
17. 40 % acrylamide, 2 % bisacrylamide solution.
18. Vertical gel electrophoresis apparatus (20 cm) and power
supply.
19. Saran film wrap.
20. Cassette for film development.
21. X-ray film.
4. T4 polynucleotide kinase.
5. T4 DNA ligase.
6. Linker A: 5′ rAppCTGTAGGCACCATCAAT/3ddC.
7. Linker B: 5′ AmMC6/GCTCCAGAATTCGGACCCGArGr
UrGrCrCrUrArCrArG.
8. MonsterScript reverse transcriptase (Epicentre, Illumina).
9. PCR reaction components: Taq DNA polymerase, MgCl2,
dNTP mix.
10. Primer for RT: 5′ GATTGATGGTGCCTACAG 3′.
11. Forward primer: 5′ GCTCCAGAATTCGGACCCGAGTG 3′.
3 Methods
3.1 RNA Antisense This step generates a radioactive probe in vitro that hybridizes to
Probe Transcription an RNA present in the sample. The protected fragment can be
detected by autoradiography. Any abundant RNA can be targeted.
Figure 3 illustrates a probe against abundant small nucleolar RNAs.
1. For the in vitro transcription reaction, combine the following:
2 μl of 10× transcription buffer (see Note 2); 3 μl of ATP, CTP,
and GTP mixture (3.3 nM each); 5 μl of 32P-UTP; DNA tem-
plate (5–10 pmol); 2 μl RNA polymerase. Bring the total reac-
tion to 20 μl with water.
2. Incubate at 37 °C for 1 h.
3. Add 1 μl TURBO DNase to degrade the remaining DNA
template.
4. To purify the full-length probe (see Note 3), run the in vitro
transcribed RNA on a 10 % acrylamide TBE gel, briefly expose
on X-ray film, and cut out the corresponding gel slice.
5. Purify the gel slice as described in Subheading 3.3.
3.2 RNase Digestion In this step, the radioactive-labeled probe is annealed to the target
RNA, generating an RNase-resistant double-stranded RNA while
single-stranded RNAs are digested. In the further cloning steps,
the RNases are removed by proteinase K treatment. To allow liga-
tion of linkers, the RNA is dephosphorylated by shrimp alkaline
phosphatase prior to the proteinase K step.
The remaining double-stranded RNAs are subsequently sepa-
rated on an 8 M urea TBE gel. The urea in the gel denatures the
double-stranded RNAs. Since the two strands have the same size,
they will not be separated by the gel and reanneal in the subsequent
dsRNA Cloning 59
a b
RNAse RNA cDNA synthesis RT
RNA RNAse + Marker Control
Marker digestion 3’ Linker
1 2 3
1
Fig. 3 Example of addition of linkers to the protected RNA. Total mouse brain RNA
was annealed with a radioactively labeled probe and digested with RNases A and
T1. After dephosphorylation and purification, the 3′ linker was added. Note the
shift in the mobility of the bands. Gel slices 1, 2, and 3 were cut out, and RNA was
isolated using gel soaking method as described. After addition of the 5′ linker,
cDNA was generated from each fraction in the presence of radiolabeled dCTP
and separated on a denaturing 15 % acrylamide gel containing 8 M urea. (a)
Signal from an RNase protection before (left) and after (right) addition of linkers.
(b) Signal of the reverse transcription reaction using radiolabeled dCTP
3.3 RNA Cloning In this step, two linkers (Fig. 2b) are ligated to the dsRNA. We
found that dsRNAs react efficiently with adenylated linkers and T4
RNA ligase. However, in our hands, the ligation with standard
RNA or DNA oligonucleotides is very inefficient. We use a Y-shaped
linker design that has been used in miRNA cloning (Fig. 2b).
The ligation of the second linker is efficient at room temperature.
1. Cut the gel slices (see Note 6).
2. Recover the protected short RNA fragments (or the probe for
Subheading 3.1) by crushing the gel using a pestle that fits in
microtubes or using a disposable pipette (see Note 7).
3. Soak the crushed gel in 400 μl of RNA elution buffer at 37 °C
overnight on a shaker.
4. Remove the gel fragments with centrifugation and ethanol-
precipitate the RNA fragments, adding 1 μl GlycoBlue as a
carrier.
dsRNA Cloning 61
4 Notes
a cloned dsRNAs
Antisense transcripts,
unknown function
b M C F R C F R
Dlgap2 Dlgap2
primer1 primer2
Fig. 4 Identification of double-stranded RNAs in the ADAT1 gene locus. (a) Visualization of the dsRNA data on
the UCSC browser. The dsRNA track contains the dsRNA that was cloned. Each dsRNA is represented by a
vertical line. SAPs are generated through the Dlgap2 transcript and expressed sequence tag Av016332. Note
that experimentally, dsRNAs can be detected outside SAP regions, indicating the presence of non-annotated
antisense RNAs. (b) RT-PCR validation of dsRNAs in the Dlgap2 locus shown in (a) on the left. F and R indicate
the detection of sense and antisense RNAs, respectively. The primers amplify the dsRNA indicated by closed
arrows in the left panel
Acknowledgment
References
1. Chen J, Sun M, Kent WJ, Huang X, Xie H, pylori. Nature 464(7286):250–255.
Wang W, Zhou G, Shi RZ, Rowley JD doi:10.1038/nature08756
(2004) Over 20 % of human transcripts 8. Nicolas P, Mader U, Dervyn E, Rochat T,
might form sense-antisense pairs. Nucleic Leduc A, Pigeonneau N, Bidnenko E,
Acids Res 32(16):4812–4820. doi:10.1093/ Marchadier E, Hoebeke M, Aymerich S, Becher
nar/gkh818 D, Bisicchia P, Botella E, Delumeau O, Doherty
2. Katayama S, Tomaru Y, Kasukawa T, Waki K, G, Denham EL, Fogg MJ, Fromion V, Goelzer
Nakanishi M, Nakamura M, Nishida H, Yap A, Hansen A, Hartig E, Harwood CR, Homuth
CC, Suzuki M, Kawai J, Suzuki H, Carninci P, G, Jarmer H, Jules M, Klipp E, Le Chat L,
Hayashizaki Y, Wells C, Frith M, Ravasi T, Pang Lecointe F, Lewis P, Liebermeister W, March A,
KC, Hallinan J, Mattick J, Hume DA, Lipovich Mars RA, Nannapaneni P, Noone D, Pohl S,
L, Batalov S, Engstrom PG, Mizuno Y, Faghihi Rinn B, Rugheimer F, Sappa PK, Samson F,
MA, Sandelin A, Chalk AM, Mottagui-Tabar S, Schaffer M, Schwikowski B, Steil L, Stulke J,
Liang Z, Lenhard B, Wahlestedt C (2005) Wiegert T, Devine KM, Wilkinson AJ, van Dijl
Antisense transcription in the mammalian tran- JM, Hecker M, Volker U, Bessieres P, Noirot P
scriptome. Science 309(5740):1564–1566. (2012) Condition-dependent transcriptome
doi:10.1126/science.1112009 reveals high-level regulatory architecture in
3. He Y, Vogelstein B, Velculescu VE, Bacillus subtilis. Science 335(6072):1103–
Papadopoulos N, Kinzler KW (2008) The anti- 1106. doi:10.1126/science.1206848
sense transcriptomes of human cells. Science 9. Mendoza-Vargas A, Olvera L, Olvera M,
322(5909):1855–1857. doi:10.1126/ Grande R, Vega-Alvarado L, Taboada B,
science.1163853 Jimenez-Jacinto V, Salgado H, Juarez K,
4. Steigele S, Nieselt K (2005) Open reading Contreras-Moreira B, Huerta AM, Collado-
frames provide a rich pool of potential natural Vides J, Morett E (2009) Genome-wide iden-
antisense transcripts in fungal genomes. Nucleic tification of transcription start sites, promoters
Acids Res 33(16):5034–5044. doi:10.1093/ and transcription factor binding sites in E. coli.
nar/gki804 PLoS ONE 4(10):e7526. doi:10.1371/jour-
5. Yamada K, Lim J, Dale JM, Chen H, Shinn P, nal.pone.0007526
Palm CJ, Southwick AM, Wu HC, Kim C, 10. Dornenburg JE, Devita AM, Palumbo MJ,
Nguyen M, Pham P, Cheuk R, Karlin- Wade JT (2010) Widespread antisense tran-
Newmann G, Liu SX, Lam B, Sakano H, Wu T, scription in Escherichia coli. MBio 1 (1).
Yu G, Miranda M, Quach HL, Tripp M, Chang doi:10.1128/mBio.00024-10
CH, Lee JM, Toriumi M, Chan MM, Tang 11. O’Carroll D, Schaefer A (2013) General prin-
CC, Onodera CS, Deng JM, Akiyama K, Ansari cipals of miRNA biogenesis and regulation in
Y, Arakawa T, Banh J, Banno F, Bowser L, the brain. Neuropsychopharmacology
Brooks S, Carninci P, Chao Q, Choy N, Enju 38(1):39–54. doi:10.1038/npp.2012.87
A, Goldsmith AD, Gurjal M, Hansen NF, 12. Gantier MP, Williams BR (2007) The response
Hayashizaki Y, Johnson-Hopson C, Hsuan of mammalian cells to double-stranded RNA.
VW, Iida K, Karnes M, Khan S, Koesema E, Cytokine Growth Factor Rev 18(5–6):363–
Ishida J, Jiang PX, Jones T, Kawai J, Kamiya A, 371. doi:10.1016/j.cytogfr.2007.06.016
Meyers C, Nakajima M, Narusaka M, Seki M,
Sakurai T, Satou M, Tamse R, Vaysberg M, 13. Li JB, Church GM (2013) Deciphering the
Wallender EK, Wong C, Yamamura Y, Yuan S, functions and regulation of brain-enriched
Shinozaki K, Davis RW, Theologis A, Ecker JR A-to-I RNA editing. Nat Neurosci
(2003) Empirical analysis of transcriptional 16(11):1518–1522. doi:10.1038/nn.3539
activity in the Arabidopsis genome. Science 14. Lavorgna G, Dahary D, Lehner B, Sorek R,
302(5646):842–846. doi:10.1126/science. Sanderson CM, Casari G (2004) In search of
1088305 antisense. Trends Biochem Sci 29(2):88–94.
6. Lasa I, Toledo-Arana A, Gingeras TR (2012) doi:10.1016/j.tibs.2003.12.002
An effort to make sense of antisense transcrip- 15. Lapidot M, Pilpel Y (2006) Genome-wide nat-
tion in bacteria. RNA Biol 9(8):1039–1044. ural antisense transcription: coupling its regu-
doi:10.4161/rna.21167 lation to its different regulatory mechanisms.
7. Sharma CM, Hoffmann S, Darfeuille F, EMBO Rep 7(12):1216–1222. doi:10.1038/
Reignier J, Findeiss S, Sittka A, Chabas S, sj.embor.7400857
Reiche K, Hackermuller J, Reinhardt R, Stadler 16. Morris KV, Santoso S, Turner AM, Pastori C,
PF, Vogel J (2010) The primary transcriptome Hawkins PG (2008) Bidirectional transcription
of the major human pathogen Helicobacter directs both transcriptional gene activation and
64 Manli Shen et al.
suppression in human cells. PLoS Genet Mol Cell Biol 10(9):637–643. doi:10.1038/
4(11):e1000258. doi:10.1371/journal.pgen. nrm2738
1000258 22. Magistri M, Faghihi MA, St Laurent G 3rd,
17. Froberg JE, Yang L, Lee JT (2013) Guided by Wahlestedt C (2012) Regulation of chroma-
RNAs: X-inactivation as a model for lncRNA tin structure by long noncoding RNAs: focus
function. J Mol Biol 425(19):3698–3706. on natural antisense transcripts. Trends Genet
doi:10.1016/j.jmb.2013.06.031 28(8):389–396. doi:10.1016/j.
18. Yu W, Gius D, Onyango P, Muldoon-Jacobs K, tig.2012.03.013
Karp J, Feinberg AP, Cui H (2008) Epigenetic 23. Kung JT, Colognori D, Lee JT (2013) Long
silencing of tumour suppressor gene p15 by its noncoding RNAs: past, present, and future.
antisense RNA. Nature 451(7175):202–206. Genetics 193(3):651–669. doi:10.1534/
doi:10.1038/nature06468 genetics.112.146704
19. Modarresi F, Faghihi MA, Lopez-Toledano 24. Sesto N, Wurtzel O, Archambaud C, Sorek R,
MA, Fatemi RP, Magistri M, Brothers SP, van Cossart P (2013) The excludon: a new concept
der Brug MP, Wahlestedt C (2012) Inhibition in bacterial antisense RNA-mediated gene reg-
of natural antisense transcripts in vivo results in ulation. Nat Rev Microbiol 11(2):75–82.
gene-specific transcriptional upregulation. Nat doi:10.1038/nrmicro2934
Biotechnol 30(5):453–459. doi:10.1038/ 25. Shen M, Eyras E, Wu J, Khanna A, Josiah S,
nbt.2158 Rederstorff M, Zhang MQ, Stamm S (2011)
20. Batra R, Charizanis K, Swanson MS (2010) Direct cloning of double-stranded RNAs from
Partners in crime: bidirectional transcription in RNase protection analysis reveals processing
unstable microsatellite disease. Hum Mol Genet patterns of C/D box snoRNAs and provides
19(R1):R77–R82. doi:10.1093/hmg/ddq132 evidence for widespread antisense transcript
21. Faghihi MA, Wahlestedt C (2009) Regulatory expression. Nucleic Acids Res 39(22):9720–
roles of natural antisense transcripts. Nat Rev 9730. doi:10.1093/nar/gkr684
Chapter 7
Abstract
Discovery and characterization of microRNAs (miRNAs) and other families of small RNAs lead researchers
to study their structures/functions and their expression patterns. The splinted ligation method described
here is based on nucleic acid hybridization. It is optimized for the direct labeling and quantitative detec-
tion of small RNAs. A specific bridge DNA oligonucleotide is used, which is perfectly complementary to
both the target small RNA and a labeled ligation nucleic acid. The target RNA is subsequently labeled by
ligation, detected by analysis in denaturing conditions, and quantified by phosphorimaging. The protocol
doesn’t require any specific material, and the procedure is fast and sensitive.
1 Introduction
65
66 Gabrielle Bourgeois et al.
Kinase
1 32P
γATP
small RNA
P OH 32P OH
5’ 3’ 5’ 3’
OH
32 P
P OH
5’ 3’
3’ 5’
Bridge oligonucleotide
T4 DNA
ligase
OH
32 P
P OH
5’ 3’
3’ 5’
P OH
32P
5’ 3’
3’ 5’
32P OH
5’ 3’
4 Alkaline DNase
phosphatase
OH
32P
5’ 3’
Fig. 1 Schematic representation of each steps during the protocol of small RNA
detection or labeling using splinted ligation. 1- labeling of the ligation RNA or
oligonucleotide/ 2- Hybridization of the small RNA with the labeled oligonucle-
otide or RNA on a bridge oligonucleotide/ 3- linking using T4 DNA ligase/ 4-
Removal of the labeled phosphate from unligated ligation RNA or oligonucleotide
and optional removal of the bridge oligonucleotide
2 Materials
2.2 Hybridization 1. T4 DNA ligase including 10× ligation buffer: 500 mM Tris–
and Ligation HCl, pH 7.5, 100 mM MgCl2, 50 mM ATP, 50 mM DTT.
of the Small RNA
3 Methods
The splinted ligation method described here can be used for the
direct labeling of RNA and quantitative detection of small RNAs
from total RNA extracts. In the first application, the reaction
consists in the ligation of two labeled RNA precursor fragments.
This approach can be applied to join RNA fragments to form long
RNAs that couldn’t otherwise be produced by in vitro transcrip-
tion for their structural and functional studies. In the second appli-
cation, the small RNA of interest present in total RNA extracts is
captured by ligation with a radiolabeled oligonucleotide to the
bridge oligonucleotide. The initial materials are different, but the
protocol is the same for both applications.
3.1 Preparation The ligation RNA can be produced by in vitro transcription or can
of the Labeled Ligation be obtained from a supplier. The first step consists in labeling its 5′
RNA/Oligonucleotide extremity with γ32P-ATP. If an oligonucleotide is used instead,
labeling reaction follows the same protocol.
1. Combine 500 ng of RNA/oligonucleotide, 2 μl of T4 PNK
buffer, 1 μl of T4 PNK, 1 μl of γ32P-ATP and RNase-free water
to a final volume of 20 μl (see Note 7).
2. Incubate the reaction for 1 h at 37 °C.
3. Remove the unincorporated isotope by a step of purification
using a quick spin column in Sephadex G-50. Centrifuge the
column at 750 × g for 2 min to discard the resin storage buffer.
4. Load the sample and centrifuge the column at 750 × g for
1 min to collect the sample. The unincorporated γ32P-ATP is
retained in the resin.
5. Proteins of the sample are removed by addition of 150 μl of
phenol–chloroform. Vortex during 2 min and centrifuge the
sample for 3 min at 10,000 × g. Collect the supernatant con-
taining the labeled RNA or oligonucleotide (see Note 8).
6. Precipitate the supernatant with 3 volumes of 99.8 % of ethanol
for at least 40 min at −80 °C (see Note 9).
7. Centrifuge at 16,000 × g at 4 °C for 30 min. Wash the pellet
with 70 % ethanol and dissolve it in 20 μl RNase-free water.
8. Use 2 μl thereof to spectrophotometrically determine the RNA
concentration.
Splinted Ligation 69
3.2 Hybridization 1. Thaw all the frozen reagents on ice before utilization.
and Ligation 2. Assemble the reaction for hybridization on ice in a final volume
of the Small RNA of 20 μl by mixing 15 μl total RNA or 400 pmol of the RNA
of interest with 400 pmol of radiolabeled RNA/oligonucle-
otide in the ligation buffer.
3. Gently mix the reaction by pipetting up and down and spin
down briefly.
4. Incubate the reaction at 90 °C for 1 min, at 75 °C for 3 min,
and finally at room temperature for 20 min.
5. Add 40 U of T4 DNA ligase and incubate the mixture at 16 °C
overnight.
3.5 Purification 1. After gel staining, the RNA in the size range of interest is
of Ligation Product excised with a sterile scalpel (see Note 15).
2. Dice the excised gel piece into 5 mm3 cubes with a sterile scalpel,
which will enhance subsequent elution of the RNA from the gel.
70 Gabrielle Bourgeois et al.
4 Notes
Acknowledgment
References
1. Moore MJ, Query CC (2000) Joining of RNAs Stepwise protein-mediated RNA folding directs
by splinted ligation. Methods Enzymol 317:109– assembly of telomerase ribonucleoprotein.
123, S0076-6879(00)17009-0 [pii] Nature 446(7134):458–461. doi:10.1038/
2. Cornish PV, Ermolenko DN, Noller HF, Ha T nature05600, nature05600 [pii]
(2008) Spontaneous intersubunit rotation in 5. Zhuang X, Bartley LE, Babcock HP, Russell R,
single ribosomes. Mol Cell 30(5):578–588. Ha T, Herschlag D, Chu S (2000) A single-
doi:10.1016/j.molcel.2008.05.004, molecule study of RNA catalysis and folding.
S1097-2765(08)00334-1 [pii] Science 288(5473):2048–2051, 8590 [pii]
3. Lu K, Miyazaki Y, Summers MF (2010) Isotope 6. Maroney PA, Chamnongpol S, Souret F, Nilsen
labeling strategies for NMR studies of RNA. J TW (2007) A rapid, quantitative assay for
Biomol NMR 46(1):113–125. doi:10.1007/ direct detection of microRNAs and other small
s10858-009-9375-2 RNAs using splinted ligation. RNA 13(6):930–
4. Stone MD, Mihalusova M, O’Connor CM, 936. doi:10.1261/rna.518107, rna.518107
Prathapam R, Collins K, Zhuang X (2007) [pii]
72 Gabrielle Bourgeois et al.
Abstract
RNA FISH is a powerful method to detect specific RNAs in fixed cells. It allows both localization and
quantification of RNA molecules within individual cells and tissues. Refined RNA FISH methods have also
been developed to determine RNA transcription and degradation rates. This chapter describes an RNA FISH
protocol that we developed in order to study the expression and localization of satellite III RNAs. This spe-
cific class of non-coding RNAs is expressed in response to various cellular stresses including heat shock. This
protocol is based on the use of a biotinylated LNA probe subsequently detected by a streptavidin–Alexa
Fluor® 488 conjugate. A protocol allowing efficient coupling of RNA FISH and protein detection by immu-
nofluorescence is also described in this chapter.
Key words RNA FISH, LNA probe, LNA FISH, RNA intracellular localization, Fluorescence
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_8, © Springer Science+Business Media New York 2015
73
74 Valentin Vautrot et al.
O B O B O B O B
O O O O
O O OH O OCH3 O F
O P O- O P O- O P O- O P O-
O B HN O B
O B O
N
O O O
O O N O
NH R’
O P O- O P O-
Fig. 1 Chemical structure of various RNA backbone modifications used for the production of fluorescence in
situ hybridization probes. B: base; R′ = H (2′4′-BNANC(N-H)) or CH3 (2′4′-BNANC(N-Me))
2 Materials
Fig. 2 Detection of sat III RNAs in HeLa cells by RNA FISH. (a) RNA FISH was performed on HeLa cells using
either the sat III LNA™-enhanced probe or the scramble-ISH LNA™-enhanced probe. The nucleus of the cells
was stained with DAPI. At 37 °C, no staining was observed when using the sat III LNA™-enhanced probe,
while in cells exposed to heat shock (2 h at 42 °C followed by 1 h at 37 °C), several bright nuclear foci cor-
responding to sat III-enriched substructures were detected in each nucleus. These foci were not detected
when using the scramble-ISH probe as a negative control. Image acquisition was performed on a Nikon epi-
fluorescence microscope using the digital camera Nikon DXM1200. (b) Co-detection of sat III RNAs by RNA
FISH (green) and SRSF1 (Santa Cruz, sc-73016) or HSF1 (Thermo Scientific MA5-14632) or SRSF2 (personal
gift from J. Stevenin) by immunofluorescence (red ). The sat III RNA foci match perfectly with the SRSF1 and
HSF1 spots detected within the nuclear stress granules, but not with SRSF2 which is specifically localized in
the nuclear speckles. Scale: 10 μm. Image acquisition was performed on a Leica SP2 confocal microscope
from the PTIBC platform of FR3209
4. 42 °C water bath.
5. Pre-cleaned Superfrost microscope slides.
6. Parafilm.
2.2 Probe 1. LNA™ mRNA detection probe (Exiqon) (see Note 3):
Preparation
Sat III probe: 5′-Biosg-ATTCCATTCCATTCCATTCC-3′Bio.
Scramble probe: 5′-Biosg-GTGTAACACGTCTATACGC
CCA-3′.
RNA-FISH of Small Non-Coding RNAs 77
2.4 Signal Detection 1. Blocking solution: 3 % (w/v) BSA, 4× SCC, 0.1 % Tween 20.
2. 22 × 50 mm microscope cover slips.
3. Incubators.
4. Streptavidin–Alexa Fluor® 488 conjugate (Molecular Probes,
Invitrogen).
5. Streptavidin–Alexa Fluor® 488 conjugate dilution buffer: 1 %
BSA, 4× SCC, 0.1 % Tween 20.
6. Wash solution 3: 4× SCC, 0.1 % Tween 20.
7. Duolink mounting media (Olink Bioscience).
8. Rubber cement (Artos, Styl’Up).
3 Methods
3.1 Cell Preparation Prepare culture plates and microscope slides following proper ster-
ile cell handling protocols.
3.1.1 Cell Growth
1. Place a sterilized microscope slide in each plate (see Note 5).
Make sure that the surface of the slide intended for cell adhe-
sion is facing up.
2. Warm the cell culture medium to 37 °C before use to avoid cell
stress. Add 10 ml of pre-warmed medium in each culture plate.
3. Seed HeLa cells in order to reach about 60–70 % confluence
after 24 h (see Note 6).
78 Valentin Vautrot et al.
3.1.2 Cell Fixation 1. Collect the microscope slides and place them in a Coplin jar
and Permeabilization filled with PBS.
2. Under a fume hood, transfer the freshly prepared 4 % parafor-
maldehyde solution (see Note 2) in a new Coplin jar. Let the
slides in the fixation solution for 10 min at room temperature,
with mild agitation.
3. Wash slides once with PBS in a clean Coplin jar at room tem-
perature (see Note 9).
4. Wash slides with 0.1 M Tris–HCl, pH 7.5, for 5 min at room
temperature.
5. Wash slides with PBS containing 20 % (v/v) glycerol for 1 h
at room temperature (minimum 20 min).
6. Place the slides without drying them in liquid nitrogen for a
few seconds with a clamp and appropriate protection.
7. Thaw the slides for about 1 min and place them again in the
PBS 20 % (v/v) glycerol solution.
8. Repeat the freeze and thaw cycle twice more.
9. Dehydrate the slides by treatment with three increasing etha-
nol concentrations: immerge the slides for 5 min in 70 % etha-
nol in a Coplin jar.
10. Replace the 70 % ethanol with 90 % ethanol and incubate for
5 min.
11. Replace the 90 % ethanol with 100 % ethanol and incubate for
5 min.
12. Dry the slides. At this stage, slides can be stored for about 12 h
at 4 °C before hybridization.
3.3 In Situ 1. Fill a Coplin jar with the wash solution 2 and pre-warm it
Hybridization at 65 °C.
2. Spot 100 μl of the denatured probe solution on each slide and
cover the drop with a 22 × 50 mm microscope cover slip
(see Note 10).
3. Incubate the slides for 30 min in a light-protected humid
chamber placed in the incubator. Hybridization temperature,
47 °C in our case, depends on the Tm of the probe.
4. Remove the cover slip by immersion in wash solution 1 at
room temperature.
5. Wash the slides for 5 min in pre-warmed wash solution 2 at
65 °C in a Coplin jar with mild agitation.
6. Repeat the washing step twice more using solution 2 pre-
warmed at 65 °C.
3.4 Signal Detection 1. Shortly dry the slides, layer them with 100 μl of blocking solu-
tion, and cover with a 22 × 50 mm microscope cover slip.
2. Incubate for 30 min at 37 °C in a light-protected humid
chamber.
3. Dilute the streptavidin–Alexa Fluor® 488 conjugate in the
streptavidin dilution buffer to a final concentration of 2 μg/ml.
4. Remove the cover slip, shortly dry the slides, and add 100 μl of
the diluted solution of streptavidin–Alexa Fluor® 488 conju-
gate. Mount a 22 × 50 mm microscope cover slip over it
(see Note 11).
5. Incubate for 1–2 h at 37 °C in the light-protected humid
chamber.
6. Fill the Coplin jar with wash solution 3 and pre-warm the jar at
45 °C.
7. Remove the cover slip by immersion in the warm wash solu-
tion 3.
8. Wash the slides for 5 min in the warm wash solution 3 in the
Coplin jar, with slow agitation.
9. Repeat this washing step twice more.
10. Dry the slides and mount cover slips with Duolink mounting
media containing antifade and DAPI.
11. Seal the cover slip with nail polish or rubber cement and pro-
ceed with microscope observation (see Note 12).
80 Valentin Vautrot et al.
4 Notes
Acknowledgments
References
1. Langer-Safer PR, Levine M, Ward DC (1982) prenatal molecular diagnosis of myotonic
Immunological method for mapping genes on dystrophy type I. Clin Chem 52(2):319–322.
Drosophila polytene chromosomes. Proc Natl doi:10.1373/clinchem.2005.056283
Acad Sci U S A 79(14):4381–4385 13. Renwick N, Cekan P, Masry PA, McGeary SE,
2. Manuelidis L, Langer-Safer PR, Ward DC Miller JB, Hafner M, Li Z, Mihailovic A,
(1982) High-resolution mapping of satellite Morozov P, Brown M, Gogakos T, Mobin
DNA using biotin-labeled DNA probes. J Cell MB, Snorrason EL, Feilotter HE, Zhang X,
Biol 95(2 Pt 1):619–625 Perlis CS, Wu H, Suarez-Farinas M, Feng H,
3. DeLong EF, Wickham GS, Pace NR (1989) Shuda M, Moore PS, Tron VA, Chang Y,
Phylogenetic stains: ribosomal RNA-based Tuschl T (2013) Multicolor microRNA FISH
probes for the identification of single cells. effectively differentiates tumor types. J Clin
Science 243(4896):1360–1363 Invest 123(6):2694–2702. doi:10.1172/
4. Brown JM, Buckle VJ (2010) Detection of JCI68760
nascent RNA transcripts by fluorescence in situ 14. Clemson CM, Lawrence JB (1996)
hybridization. Methods Mol Biol 659:33–50. Multifunctional compartments in the nucleus:
doi:10.1007/978-1-60761-789-1_3 insights from DNA and RNA localization.
5. Ronander E, Bengtsson DC, Joergensen L, J Cell Biochem 62(2):181–190. doi:10.1002/
Jensen AT, Arnot DE (2012) Analysis of single- (SICI)1097-4644(199608)62:2<181::AID-
cell gene transcription by RNA fluorescent in JCB6>3.0.CO;2-O
situ hybridization (FISH). J Vis Exp (68). 15. Jolly C, Metz A, Govin J, Vigneron M, Turner
doi:10.3791/4073 BM, Khochbin S, Vourc’h C (2004) Stress-
6. Wagner M, Haider S (2012) New trends in induced transcription of satellite III repeats.
fluorescence in situ hybridization for identifica- J Cell Biol 164(1):25–33. doi:10.1083/
tion and functional analyses of microbes. Curr jcb.200306104
Opin Biotechnol 23(1):96–102. 16. Sierra-Miranda M, Delgadillo DM, Mancio-
doi:10.1016/j.copbio.2011.10.010 Silva L, Vargas M, Villegas-Sepulveda N,
7. Twedt DC, Cullen J, McCord K, Janeczko S, Martinez-Calvillo S, Scherf A, Hernandez-Rivas
Dudak J, Simpson K (2014) Evaluation of flu- R (2012) Two long non-coding RNAs gener-
orescence in situ hybridization for the detec- ated from subtelomeric regions accumulate in a
tion of bacteria in feline inflammatory liver novel perinuclear compartment in Plasmodium
disease. J Feline Med Surg 16(2):109–117. doi falciparum. Mol Biochem Parasitol 185(1):36–
:10.1177/1098612X13498249 47. doi:10.1016/j.molbiopara.2012.06.005
8. Gribnau J, Diderich K, Pruzina S, Calzolari R, 17. Bertozzi D, Iurlaro R, Sordet O, Marinello J,
Fraser P (2000) Intergenic transcription and Zaffaroni N, Capranico G (2011)
developmental remodeling of chromatin sub- Characterization of novel antisense HIF-1alpha
domains in the human beta-globin locus. Mol transcripts in human cancers. Cell Cycle
Cell 5(2):377–386 10(18):3189–3197
9. van de Corput MP, Grosveld FG (2001) 18. Froehler BC, Wadwani S, Terhorst T, Gerrard S
Fluorescence in situ hybridization analysis of (1992) Oligodeoxynucleotides containing C-5
transcript dynamics in cells. Methods 25(1):111– propyne analogs of 2′-deoxyuridine and 2′-deox-
118. doi:10.1006/meth.2001.1220 ycytidine. Tetrahedron Lett 33:5307–5310
10. Bridger JM, Kalla C, Wodrich H, Weitz S, King 19. Barnes TW 3rd, Turner DH (2001) Long-
JA, Khazaie K, Krausslich HG, Lichter P range cooperativity in molecular recognition of
(2005) Nuclear RNAs confined to a reticular RNA by oligodeoxynucleotides with multiple
compartment between chromosome territo- C5-(1-propynyl) pyrimidines. J Am Chem Soc
ries. Exp Cell Res 302(2):180–193. 123(18):4107–4118
doi:10.1016/j.yexcr.2004.07.038 20. Prosnyak MI, Veselovskaya SI, Myasnikov VA,
11. Hutchinson JN, Ensminger AW, Clemson CM, Efremova EJ, Potapov VK, Limborska SA,
Lynch CR, Lawrence JB, Chess A (2007) A Sverdlov ED (1994) Substitution of
screen for nuclear transcripts identifies two 2-aminoadenine and 5-methylcytosine for ade-
linked noncoding RNAs associated with SC35 nine and cytosine in hybridization probes
splicing domains. BMC Genomics 8:39. increases the sensitivity of DNA fingerprinting.
doi:10.1186/1471-2164-8-39 Genomics 21(3):490–494. doi:10.1006/
12. Bonifazi E, Gullotta F, Vallo L, Iraci R, Nardone geno.1994.1306
AM, Brunetti E, Botta A, Novelli G (2006) Use 21. Xodo LE, Manzini G, Quadrifoglio F, van der
of RNA fluorescence in situ hybridization in the Marel GA, van Boom JH (1991) Effect of
RNA-FISH of Small Non-Coding RNAs 83
5-methylcytosine on the stability of triple- analogue with excellent hybridizing and nucle-
stranded DNA–a thermodynamic study. ase resistance profiles. Nucleosides Nucleotides
Nucleic Acids Res 19(20):5625–5631 Nucleic Acids 26(10–12):1625–1628.
22. Lebedev Y, Akopyants N, Azhikina T, doi:10.1080/15257770701548980
Shevchenko Y, Potapov V, Stecenko D, Berg 28. Biamonti G, Vourc’h C (2010) Nuclear stress
D, Sverdlov E (1996) Oligonucleotides con- bodies. Cold Spring Harb Perspect Biol
taining 2-aminoadenine and 5-methylcytosine 2(6):a000695. doi:10.1101/cshperspect.
are more effective as primers for PCR amplifi- a000695
cation than their nonmodified counterparts. 29. Metz A, Soret J, Vourc’h C, Tazi J, Jolly C
Gene Anal 13(1):15–21 (2004) A key role for stress-induced satellite
23. He J, Seela F (2002) Propynyl groups in duplex III transcripts in the relocalization of splicing
DNA: stability of base pairs incorporating factors into nuclear stress granules. J Cell Sci
7-substituted 8-aza-7-deazapurines or 117(Pt 19):4551–4558. doi:10.1242/
5-substituted pyrimidines. Nucleic Acids Res jcs.01329
30(24):5485–5496 30. Denegri M, Chiodi I, Corioni M, Cobianchi F,
24. Kawasaki AM, Casper MD, Freier SM, Lesnik Riva S, Biamonti G (2001) Stress-induced
EA, Zounes MC, Cummins LL, Gonzalez C, nuclear bodies are sites of accumulation of pre-
Cook PD (1993) Uniformly modified mRNA processing factors. Mol Biol Cell
2′-deoxy-2′-fluoro phosphorothioate oligonu- 12(11):3502–3514
cleotides as nuclease-resistant antisense com- 31. Rizzi N, Denegri M, Chiodi I, Corioni M,
pounds with high affinity and specificity for Valgardsdottir R, Cobianchi F, Riva S, Biamonti
RNA targets. J Med Chem 36(7):831–841 G (2004) Transcriptional activation of a consti-
25. Silverman AP, Kool ET (2007) Oligonucleotide tutive heterochromatic domain of the human
probes for RNA-targeted fluorescence in situ genome in response to heat shock. Mol Biol
hybridization. Adv Clin Chem 43:79–115 Cell 15(2):543–551. doi:10.1091/mbc.
26. Petersen M, Wengel J (2003) LNA: a versatile E03-07-0487
tool for therapeutics and genomics. Trends 32. Valgardsdottir R, Chiodi I, Giordano M, Rossi
Biotechnol 21(2):74–81. doi:10.1016/S0167- A, Bazzini S, Ghigna C, Riva S, Biamonti G
7799(02)00038-0 (2008) Transcription of Satellite III non-
27. Rahman SM, Seki S, Utsuki K, Obika S, coding RNAs is a general stress response in
Miyashita K, Imanishi T (2007) human cells. Nucleic Acids Res 36(2):423–
2′,4′-BNA(NC): a novel bridged nucleic acid 434. doi:10.1093/nar/gkm1056
Chapter 9
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that negatively regulate messenger RNA
(mRNA) translation into protein. MiRNAs play a key role in gene expression regulation, and their involve-
ment in disease biology is well documented. This has fueled the development of numerous tools for the
quantification of miRNA expression levels. These tools are based on three technologies: (microarray)
probe hybridization, RNA sequencing, and reverse transcription quantitative polymerase chain reaction
(RT-qPCR). In this chapter, we describe a quantification system based on RT-qPCR technology, which is
currently considered as the most sensitive, flexible, and accurate method for quantification of not only
miRNA but also RNA expression in general. To this purpose, we have divided the protocol in three sec-
tions: reverse transcription (RT) reaction, optional preamplification (PA), and finally qPCR. Three quality-
control (QC) steps are implemented in this workflow for assessment of RNA extraction efficiency, sample
purity (e.g., absence of inhibitors), and inter-run variations, by examining the detection level of different
spike-in synthetic miRNAs. We conclude by demonstrating raw data preprocessing and normalization
using expression data obtained from high-throughput miRNA profiling of human RNA samples.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_9, © Springer Science+Business Media New York 2015
85
86 Fjoralba Zeka et al.
Fig. 1 Selective conversion of mature miRNAs into cDNA in miScript HiSpec Buffer. In a reverse transcription
reaction with miScript HiSpec Buffer, mature miRNAs are polyadenylated by a poly-A polymerase and con-
verted into cDNA by a reverse transcriptase with oligodT priming. The cDNA is then used for qPCR quantifica-
tion of mature miRNA expression
2 Materials
3 Methods
3.1 Reverse 1. If RNA isolation has not yet been performed, please carefully
Transcription (RT) read Notes 1–3. If RNA is available, measure RNA concentra-
Reaction tion by spectrophotometry or fluorometry for samples with
low RNA concentration (<5 ng/μl).
2. If the total RNA concentration is high (>100 ng/μl), dilute the
samples to the minimum concentration (see Notes 11 and 12)
and continue with the RT reaction (step 5).
3. If total RNA concentration is low (<100 ng/μl), closely evalu-
ate the quality of the samples with the lowest concentrations
(<5 ng/μl). Consider their exclusion from the study and avoid
equalization of RNA concentrations for the other samples to a
suboptimally low concentration (see Notes 11 and 12). Dilute
the samples to the minimum concentration and continue with
the RT reaction (step 5).
4. If all samples have an RNA concentration varying around 5 ng/
μl or if RNA concentration cannot be reliably assessed (e.g.,
90 Fjoralba Zeka et al.
Table 1
Reverse transcription reaction mix components
Mix components Volume per reaction (μl) 10 reactions (μl) 10 % excess (μl)
Reverse Transcriptase Mix 1 10 11
5× miScript RT Buffer 2 20 22
10× Nucleics Mix 1 10 11
RNase-free water Variable Variable Variable
RNA sample Variable
Total volume per reaction 10
3.2 qPCR for Quality 1. Dilute 2 μl of the RT product (also for the negative control
Control of the RT sample) in 20 μl of RNase-free water (see Note 17).
Reaction 2. Prepare one qPCR mix for all samples to be measured as shown
in Table 3. Prepare a second qPCR mix besides the miRTC
RT-qPCR 91
Table 2
Reverse transcription run protocol
Table 3
qPCR mix components
Volume per 10 × 2 10 %
Mix components reaction (μl) reactions (μl) excess (μl)
2× QuantiTect SYBR Green 5 100 110
PCR Master Mix
10× miScript Universal Primer 1 20 22
10× miScript Primer Assay 1 20 22
RNase-free water 1 20 22
Diluted RT product 2
Total volume per reaction 10
Table 4
qPCR run protocol
qPCR mix for the RNA isolation spike-in control if one was
used during RNA isolation. Prepare sufficient mix to perform
all reactions in duplicate (see Note 18).
3. Mix gently by pipetting up and down and distribute the qPCR
mix in a 384-well qPCR plate (see Note 15).
4. Mix the diluted RT sample by gently pipetting up and down
and add 2 μl of the product to the qPCR mix (see Note 15).
5. Run the qPCR protocol (Table 4).
92 Fjoralba Zeka et al.
Table 5
Preamplification reaction mix components
Mix components Volume per reaction (μl) 10 reactions (μl) 10 % excess (μl)
5× miScript PreAMP Buffer 5 50 55
HotStarTaq DNA Polymerase 2 20 22
miScript PreAMP Primer Mix 5 50 55
RNase-free water 7 70 77
miScript PreAMP Universal Primer 1 10 11
Diluted RT product 5
Total volume 25
3.3 Preamplification 1. Dilute the 7 μl RT aliquot from the samples and the negative
Reaction (Optional, control sample in 28 μl water to obtain a fivefold dilution (see
Recommended Notes 16 and 17).
for Highest Sensitivity) 2. Prepare one PA mix for all reactions (one reaction per sample
and per primer pool) (Table 5).
3. Mix well by gently pipetting up and down (see Note 15) and
distribute 20 μl in separate reaction tubes.
4. Gently mix the diluted (1:5) RT product by pipetting up and
down (see Note 15) and add 5 μl to each PA reaction tube to
obtain a final volume of 25 μl.
5. If multiple PA primer pools are available (e.g., 7 pools for miR-
Base v.20), multiple PA reactions should be prepared for each
sample (see Note 9).
6. Run the PA protocol on the thermal cycler (Table 6).
7. Centrifuge the PA product briefly to collect the condensed
fraction and mix gently by pipetting up and down.
8. If multiple PA reactions per sample were performed, pool the
PA reactions prior to dilution.
9. Dilute the pooled PA product five times into the PA reaction
to obtain a 1:5 dilution per ng cDNA (see Note 20).
10. Make a 2 μl aliquot of the diluted PA product for the quality-
control step.
RT-qPCR 93
Table 6
Preamplification reaction run protocols
3.4 qPCR for Quality 1. Prepare one qPCR mix for all samples (see Table 3). Prepare a
Control second qPCR mix besides the miRTC qPCR mix for the RNA
of the Preamplification isolation spike-in control if one was used during RNA isola-
Reaction tion. Prepare sufficient mix to perform all reactions in dupli-
cate (see Note 18).
2. Mix gently by pipetting up and down and distribute the mix in
a 384-well qPCR plate (see Note 15).
3. Mix the diluted PA sample by gently pipetting up and down
and add 2 μl of the product to the qPCR mix (see Note 15).
4. Run the qPCR experiment (Table 4).
5. Export raw miRTC Cq data and calculate the Cq difference
between each RNA sample and the water negative control
sample.
6. Exclude samples that show a Cq difference larger than 1 as this
likely reflects inefficient PA reaction, qPCR inhibition, or tech-
nical failure (see Note 19).
3.5 qPCR 1. Continue with the samples that passed the quality-control
steps after RT reaction and PA reaction.
2. Prepare one qPCR mix per sample and distribute in 384-well
plates (Table 7).
3. Run the samples on a qPCR instrument (Table 4).
4. Export raw Cq values from the qPCR experiment.
5. Evaluate PPC variations across samples (see Note 21).
6. Calculate the global mean miRNA Cq value per sample and
evaluate this value together with the Cq values for miRTC and
PPC across samples to check for possible occurrence of batch
effects during the experiment (see Note 22).
94 Fjoralba Zeka et al.
Table 7
Mix components for 384-well platea qPCR reactions
1 plate + 10 % 5 plates + 10 %
Mix components 1 plate (µl) excess (µl) excess (µl)
2× QuanTitect SYBR Green PCR Master Mix 2,050 2,255 11,275
10× miScript Universal Primer 410 451 2,255
RNase-free water 1,540 1,694 8,470
Template cDNA 100 110 550
Total volume 4,100 4,510 22,550
a
In this table, mix components are shown for plates pre-spotted with PCR Primer Assays. If non-spotted plates are used
then 10× miScript Primer Assay should be added to the PCR Mix (mix composition as shown in Table 3)
4 Notes
50
40
30
RNA
concentration
(ng/µl)
20
10
miRNeasy miRNeasy
mini kit micro kit
100
92.7
86.2
Cumulative
fraction 50
(%) <1 <2
ΔCq miRTC
ΔCq cel-miR-39-3p
0
ΔCq
Fig. 3 ∆Cq cumulative density for miRTC and cel-miR-39-3p measured in 289 serum samples (∆Cq = Cq RNA
sample − Cq negative control sample). 86.2 % of the samples show a miRTC ∆Cq smaller than 1, and 92.7 %
of the samples show a miRTC ∆Cq value smaller than 2
18. For each QC qPCR step, the samples are measured in the same
plate according to the sample maximization principle [11]; so
run all samples on one plate for as many assays as possible and
divide assays over different plates only if needed. If working with
a large number of samples (that cannot fit on a single plate),
multiple runs per target are required, and inter-run calibration
should be performed to avoid technical inter-plate variation.
19. Cumulative density plot for miRTC ∆Cq values obtained from
289 serum samples is shown in Fig. 3. Samples showing a
miRTC ∆Cq value larger than one (13.8 %) were not used in
the downstream analyses. A larger variation for spike-in ∆Cq
(cel-miR-39-3p) was observed. In this study, no sample exclu-
sion was performed based on this criterion, except for one
sample where the spike-in was not detected.
20. In this workflow, the minimum dilution factor for preamplified
cDNA is fivefold (miRNome arrays and custom arrays for 96–384
assays). If cDNA concentration is known, the dilution factor equals
the amount of input cDNA (ng) in the PA reaction multiplied by
5. For example, if 2 ng cDNAs were added to the PA reaction, a
tenfold (2 × 5) dilution should be performed after the PA reaction.
If cDNA concentration is unknown (e.g., body fluids), perform a
fivefold dilution and prepare an additional QC qPCR test to evalu-
ate the Cq values for some miRNAs (the miScript PreAMP
Handbook recommends evaluation of miR-16 or SNORD95). If
other miScript array formats are being used (pathway-focused
arrays and custom arrays), carefully consult the miScript PreAMP
Handbook to determine the optimal dilution factor.
100 Fjoralba Zeka et al.
100
Cumulative
fraction 50
(%) 96.0
%
18.3 19.3
Cq PPC
Acknowledgment
References
1. Krol J, Loedige I, Filipowicz W (2010) The J, Grimley C, Lader E, Lee KY, Luo S,
widespread regulation of microRNA biogenesis, Mouritzen P, Narayanan A, Patel S, Peiffer S,
function and decay. Nat Rev Genet 11:597–610 Rüberg S, Schroth G, Schuster D, Shaffer JM,
2. Kozomara A, Griffiths-Jones S (2013) miR- Shelton EJ, Silveria S, Ulmanella U,
Base: annotating high confidence microRNAs Veeramachaneni V, Staedtler F, Peters T,
using deep sequencing data. Nucleic Acids Res Guettouche T, Wong L, Vandesompele J
42:D68–D73 (2014) Evaluation of quantitative miRNA
3. Berezikov E (2011) Evolution of microRNA expression platforms in the microRNA quality
diversity and regulation in animals. Nat Rev control (miRQC) study. Nat Methods
Genet 12:846–860 11:809–815
8. Mestdagh P, Van Vlierberghe P, De Weer A,
4. Soifer HS, Rossi JJ, Sætrom P (2007)
Muth D, Westermann F, Speleman F,
MicroRNAs in disease and potential therapeu-
Vandesompele J (2009) A novel and universal
tic applications. Mol Ther 15:2070–2079
method for microRNA RT-qPCR data normal-
5. Lesko LJ (2007) Personalized medicine: elu- ization. Genome Biol 10:R64
sive dream or imminent reality? Clin Pharmacol 9. D’haene B, Mestdagh P, Hellemans J,
Ther 81:807–816 Vandesompele J (2012) MiRNA expression
6. Schwarzenbach H, Hoon DSB, Pantel K profiling: from reference genes to global mean
(2011) Cell-free nucleic acids as biomarkers in normalization. Methods Mol Biol 822:
cancer patients. Nat Cell Biol 11:426–437 261–272
7. Mestdagh P, Hartmann N, Baeriswyl L, 10. Ståhlberg A, Håkansson J, Xian X, Semb H,
Andreasen D, Bernard N, Chen C, Cheo D, Kubista M (2004) Properties of the reverse
D’Andrade P, DeMayo M, Dennis L, Derveaux transcription reaction in mRNA quantification.
S, Feng Y, Fulmer SS, Gerstmayer B, Gouffon Clin Chem 50:509–515
102 Fjoralba Zeka et al.
Abstract
Stem-loop qRT-PCR is one of the most commonly used real-time PCR approach to quantify small
non-coding RNAs such as microRNAs. The quantification method is divided in two steps. First, RNA is
reverse-transcribed using a specific stem-loop primer, and the resulting RT product is subsequently used as
a template for quantitative real-time PCR. This fast and simple method provides quantitative data with
high sensitivity and specificity to study miRNAs and their functions.
Key words microRNA, Small non-coding RNA, Real-time PCR quantification, Stem-loop primers
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_10, © Springer Science+Business Media New York 2015
103
104 Véronique Salone and Mathieu Rederstorff
Mature microRNA
Stem-loop primer
1. Stem-loop RT
2. Real-time PCR
Forward Primer
Reverse Primer
TaqMan probe
Fig. 1 RNA is reverse-transcribed into cDNA using a stem-loop primer that binds
the 3′ end of the small non-coding RNA. Then, the RT product is quantified using
TaqMan probes in a qPCR employing specific primers
2 Materials
5. Thermocyclers.
6. Applied Biosystems Step One Plus™ Real-Time PCR system
(Life Technologies™) or any equivalent Real-Time PCR
system.
7. Speed vacuum.
8. Centrifuges.
9. ABI PRISM® 96-Well Optical Reaction Plates with Barcodes.
10. 96-well plate adhesive film.
11. PCR strips and caps.
12. Nuclease-free pipette tips with filters.
13. Positive displacement, air displacement, or multichannel
pipettors.
14. Polypropylene tubes.
15. RNase-free, sterile water.
3 Methods
3.2 Real-Time PCR 1. Thaw all components on ice. Protect fluorescent probes from
(See Notes 7 and 8) light.
2. Combine following in a PCR tube: 10 μl of 2× TaqMan
Universal PCR Master Mix, No AmpErase UNG and 7.67 μl
nuclease-free water (see Note 9).
3. Mix gently by pipetting up and down. Spin down briefly.
4. Add 1.0 μl of 20× TaqMan MicroRNA Assay mix.
5. Add 1.33 μl of the diluted RT product.
6. Mix gently.
7. Seal the tube or the PCR plate and spin down quickly.
8. Keep on ice until PCR reaction is performed.
9. Set the real-time PCR thermocycler. We use an Applied
Biosystems Step One Plus™ with following program parame-
ters: initial enzyme activation at 95 °C, for 10 min; 40 cycles:
denaturation at 95 °C for 10 s; hybridization/elongation at
60 °C for 60 s. Use default values for temperature auto incre-
ment, ramp rate and data collection.
10. Load the reaction tubes or plate into the thermocycler and
start the run (see Notes 10 and 11).
4 Notes
Acknowledgment
References
1. Kwok S, Higuchi R (1989) Avoiding false posi- Vandesompele J (2009) A novel and universal
tives with PCR. Nature 339(6221):237–238. method for microRNA RT-qPCR data normal-
doi:10.1038/339237a0 ization. Genome Biol 10(6):R64. doi:10.1186/
2. Mullis KB, Faloona FA (1987) Specific synthesis gb-2009-10-6-r64, gb-2009-10-6-r64 [pii]
of DNA in vitro via a polymerase-catalyzed chain 6. Pfaffl MW, Tichopad A, Prgomet C, Neuvians
reaction. Methods Enzymol 155:335–350. TP (2004) Determination of stable housekeep-
doi:10.1016/0076-6879(87)55023-6 [pii] ing genes, differentially regulated target genes
3. Livak KJ, Schmittgen TD (2001) Analysis of rela- and sample integrity: BestKeeper–Excel-based
tive gene expression data using real-time quantita- tool using pair-wise correlations. Biotechnol
tive PCR and the 2(-Delta Delta C(T)) Method. Lett 26(6):509–515
Methods 25(4):402–408. doi:10.1006/ 7. Wei CH, Kao HY (2011) Cross-species gene
meth.2001.1262, S1046-2023(01)91262-9 [pii] normalization by species inference. BMC
4. Galiveti CR, Rozhdestvensky TS, Brosius J, Bioinform 12(Suppl 8):S5. doi:10.1186/1471-
Lehrach H, Konthur Z (2010) Application of 2105-12-S8-S5, 1471-2105-12-S8-S5 [pii]
housekeeping npcRNAs for quantitative expres- 8. Andersen CL, Jensen JL, Ørntoft T (2004)
sion analysis of human transcriptome by real- Normalization of real-time quantitative RT-PCR
time PCR. RNA 16(2):450–461. doi:10.1261/ data: a model based variance estimation approach
rna.1755810, rna.1755810 [pii] to identify genes suited for normalization - applied
5. Mestdagh P, Van Vlierberghe P, De Weer A, to bladder- and colon-cancer data-sets. Cancer
Muth D, Westermann F, Speleman F, Res 64:5245–5250
Chapter 11
Abstract
Computational prediction of microRNAs (miRNAs) is one of the most important approaches in microRNA
studies. While validation of the predicted microRNAs’ precise sequences is essential for further studies on
their biogenesis, evolution, and functions, computational miRNA prediction methods, however, often fail
to predict the accurate sequence of the mature miRNA within the precursor at the nucleotide precision
level. Here, we depict a highly efficient method for determining the precise sequences of computationally
predicted miRNAs. The method combines the generation of miRNA-enriched libraries, with 5′- and 3′-end
adaptors being linked to the miRNA molecules, the reverse transcription of small RNAs with an oligo-d(T)
anchor primer, two specific 5′- and 3′-miRNA-RACE (miR-RACE) PCR reactions and sequence-directed
cloning. The efficiency of this method was demonstrated by the precise sequence validation of computation-
ally predicted miRNAs in citrus, apple, and other fruit crops. Our ongoing research indicates that miR-
RACE is also very useful to verify the sequences of putative miRNAs obtained by deep sequencing of small
RNA libraries. The protocol of miR-RACE is rapid and can be completed within 2–3 days. miR-RACE
should make the bioinformatic prediction of miRNAs more powerful and accurate.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_11, © Springer Science+Business Media New York 2015
109
110 Chen Wang and Jinggui Fang
miRNAs 5’ 3’
Poly(A) polymerase polyadenylation
Poly (A)-tailed miRNAs 5’ AAAA(A)n 3’
5’RNA adaptor 5’ 3’ T4 RNA ligase
5’ AAAA(A)n 3’
TTTT(T)n 5’
Reverse transcription dT(30) RT primer
First strand cDNA 3’ TTTT(T)n 5’
3’ TTTT(T)n 5’ 3’ TTTT(T)n 5’
5’ AAAA(A)n 3’ 5’ AAAA(A)n 3’
TTTT(T)10
Forward GSP2
Perform PCR with MirRacer Perform PCR with MirRacer Reverse GSP1
3’ primer and GSP2 5’ primer and GSP1
3’ TTTT(T)n 5’ 3’ TTTT(T)30 5’
5’ AAAA(A)n 3’ 5’ AAAA(A)30 3’
2 Materials
2.1 Small RNA 1. SDS lysis buffer: 50 mM Tris–HCl, pH 8 (see Note 1), 140 mM
Preparation NaCl, 10 mM EDTA, 4 % SDS (Sodium Dodecyl Sulfate), 3 %
PVP (polyvinylpyrrolidone K30), 3 % ß-mercaptoethanol (add
just before use) (see Note 2). Store at 4 °C.
2. 3 M NaAC, pH 5.2 (see Note 3).
112 Chen Wang and Jinggui Fang
3 Methods
3.3 miR-RACE Perform 5′ miR-RACE reactions with the mirRacer 5′ primer and
miRNA-gene-specific forward primers (mGSP1), and carry out 3′
miR-RACE reactions with the mirRacer 3′ primer and miRNA-
gene-specific reverse primers (mGSP2) (see Note 16).
3.3.1 First Round 1. In a sterile microtube, mix the following reagents for each
Amplification experimental and control sample: 3 μl Hercules Hot-Start
polymerase buffer, 4 μl dNTP mix (2.5 mM), 2.5 U Ex Taq™
Hot-Start DNA polymerase Version 4, 2 μl of template cDNA,
25 pmol of mirRacer 3′ primer or mirRacer 5′ primer. Set up a
control experiment without template.
2. Mix and heat in a DNA thermocycler for 5 min at 98 °C to
denaturate the first strand product and to activate the
polymerase.
3. Perform PCR using following reaction parameters: initial
denaturation step at 94 °C for 3 min; 25 cycles: Denaturation
at 94 °C for 10 s, Annealing at 65 °C for 10 s, Elongation at
72 °C for 1 min; Final extension at 72 °C for 10 min. Store
PCR products at 4 °C.
3.3.2 Second Round 1. Dilute an aliquot of the first round amplification to the 1:20 in
Amplification TE buffer.
2. In a sterile, 0.2 ml microtube, mix the following reagents on
ice: 3 μl Hercules Hot-Start polymerase buffer, 4 μl dNTP mix
(2.5 mM), 2.5 U Ex Taq™ Hot Start DNA polymerase Version
4, 2 μl of the diluted first round amplification product and
25 pmol of each primers mGSP2 and mGSP1. Set up a control
experiment without template.
3. Mix and heat in a DNA thermocycler for 5 min at 98 °C to
denaturate the first-strand product and to activate the
polymerase.
4. Perform PCR using following reaction parameters: initial
denaturation step at 94 °C for 3 min; 25 cycles: Denaturation
at 94 °C for 10 s, Annealing at 65 °C for 10 s, Elongation at
116 Chen Wang and Jinggui Fang
Fig. 2 The products of 3′-miR-RACE (a) and 5′-miR-RACE (b) for eight miRNAs
3.3.3 miR-RACE Directly clone the DNA fragment purified with the TOPO TA
Products Cloning cloning Kit (Invitrogen).
and Sequencing
1. Add 4.5 μl of purified DNA, 0.5 μl pMD™ 19-T Vector, 5.0 μl
Solution I into a 0.2 ml sterile microtube. Mix and quickly spin
down.
2. Incubate at 16 °C for 1 h.
3. Transform E. coli DH5α competent cells with the ligation
product and plate on solid LB medium containing ampicillin.
Incubate overnight at 37 °C.
4. Pick positive clones and perform colony-PCR using the PCR-
specific primer pairs and PCR program as in Subheading 3.3.2.
5. Sequence positive clones PCR-products (see Note 17).
Sequencing miRNAs 117
4 Notes
Acknowledgements
References
1. Bartel B, Bartel DP (2003) MicroRNAs: at the Vitulo N, Jubin C et al (2007) The grapevine
root of plant development? Plant Physiol genome sequence suggests ancestral hexa-
13:709–717 ploidization in major angiosperm phyla. Nature
2. Bartel DP (2004) MicroRNAs: genomics, 449:463–467
biogenesis, mechanism, and function. Cell 13. Griffiths-Jones S, Saini HK, van Dongen S,
116:281–297 Enright AJ (2008) miRBase: tools for microRNA
3. Mallory AC, Vaucheret H (2004) MicroRNAs: genomics. Nucleic Acids Res 36:D154–D158
something important between the genes. Curr 14. Meyers BC, Axtell MJ, Bartel B, Bartel DP,
Opin Plant Biol 7:120–125 Baulcombe D et al (2008) Criteria for annota-
4. Carrington JC, Ambros V (2003) Role of tion of plant microRNAs. Plant Cell 20:
microRNAs in plant and animal development. 3186–3190
Science 301:336–338 15. Sunkar R, Jagadeeswaran G (2008) In silico
5. Hunter C, Poethig RS (2003) Missing links: identification of conserved microRNAs in large
miRNAs and plant development. Curr Opin number of diverse plant species. BMC Plant
Genet Dev 13:372–378 Biol 8:37
6. Jones-Rhoades MW, Bartel DP, Bartel B 16. Wang C, Shangguan LF, Nicholas KK, Wang
(2006) MicroRNAs and their regulatory roles XC, Han J, Song CN, Fang JG (2011)
in plants. Annu Rev Plant Biol 57:19–53 Characterization of microRNAs identified in a
7. Mallory AC, Bouche N (2008) MicroRNA- table grapevine cultivar with validation of com-
directed regulation to cleave or not to cleave. putationally predicted grapevine miRNAs by
Trends Plant Sci 13:359–367 miRRACE. PLoS One 6:e21259
8. Carthew RW, Sontheimer EJ (2009) Origins 17. Carra A, Mica E, Gambino G, Pindo M, Moser
and mechanisms of miRNAs and siRNAs. Cell C, Pè ME, Schubert A (2009) Cloning and
136:642–655 characterization of small non-coding RNAs
9. Brodersen P, Sakvarelidze-Achard L, Bruun- from grapevine. Plant J 59:750–763
Rasmussen M, Dunoyer P, Yamamoto YY, 18. Jung JH, Park CM (2007) MIR166/165
Sieburth L, Voinnet O (2008) Widespread genes exhibit dynamic expression patterns in
translational inhibition by plant miRNAs and regulating shoot apical meristem and floral
siRNAs. Science 320:1185–1190 development in Arabidopsis. Planta 225:
10. Zhang BH, Pan XP, Cannon CH, Cobb GP, 1327–1338
Anderson TA (2006) Conservation and diver- 19. Yamaguchi A, Wu MR, Yang L, Wu G, Poethig
gence of plant microRNA genes. Plant J 46: RS et al (2009) The microRNA-regulated
243–259 SBP-Box transcription factor SPL3 is a direct
11. Zhang BH, Pan XP, Wang QL, Cobb GP, upstream activator of LEAFY, FRUITFULL,
Anderson TA (2006) Computational identifi- and APETALA1. Cell 17:268–278
cation of microRNAs and their targets. Comput 20. Yin Z, Li CH, Han XL, Shen FF (2008)
Biol Chem 30:395–407 Identification of conserved microRNAs and
12. Jaillon O, Aury JM, Noel B, Policriti A, Clepet their target genes in tomato (Lycopersicon
C, Casagrande A, Choisne N, Aubourg S, esculentum). Gene 414:60–66
Chapter 12
Abstract
The diverse roles of RNAs depend on their ability to fold so as to form biologically functional structures.
Thus, understanding the function of a given RNA molecule often requires experimental analysis of its
secondary structure by in vitro RNA probing, which is more accurate than using prediction programs only.
This chapter presents in vitro RNA probing protocols that we routinely use, from RNA transcript production
and purification to RNA structure determination using enzymatic (RNases T1, T2, and V1) and chemical
(DMS, CMCT, kethoxal, and Pb2+) probing performed on both unlabeled and end-labeled RNAs.
Key words RNA, Secondary structure, Enzymatic probing, Chemical probing, SHAPE, RNase
digestion, Primer extension, In vitro transcription, RNA labeling, Gel electrophoresis, Structure
prediction
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_12, © Springer Science+Business Media New York 2015
119
Table 1
Commonly used RNA structure probing reagents and their specificities
Structural
Modification reagent Specificity information
DMS (dimethyl sulfate) Methylation at the N7-G, N1-A, and N3-C Pairing status
positions
CMCT (1-cyclohexyl-(2- Alkylation at the N3-U and possibly N1-G Pairing status
morpholinoethyl) carbodiimide positions
metho-p-toluene sulphonate)
Kethoxal Modification at the N1-G and N2-G positions Pairing status
DEPC (diethylpyrocarbonate) Carbethoxylation at the N7-A position Pairing status
ENU (ethylnitrosourea) Alkylation of phosphates Pairing status
Mung bean nuclease Cleavage at any single-stranded residue Pairing status
RNase A Cleavage at single-stranded C and U Pairing status
RNase CL3 Cleavage at single-stranded C Pairing status
RNase I Cleavage at any single-stranded residue Pairing status
RNase Phy M Cleavage at single-stranded A and U Pairing status
RNase T1 Cleavage of the phosphodiester bond 3′ to Pairing status
single-stranded G
RNase T2 Cleavage of the phosphodiester bond 5′ to Pairing status
any single-stranded residue
RNase U2 Cleavage of the phosphodiester bonds of Pairing status
single-stranded A
RNase V1 Cleavage of the phosphodiester bonds in Pairing status
double-strands or stacked regions
S1 nuclease Cleavage at any single-stranded residue Pairing status
Hydroxyl radicals Backbone cleavage at RNA positions Solvent accessibility
accessible to solvent
Lead acetate (Pb2+) Cleavage of phosphodiester bonds of Solvent accessibility
single-stranded residues
NMIA (N-methylisatoic Modification of 2′ OH of single-stranded Local nucleotide
anhydride) nucleotides flexibility
1 M7 (1-methyl-7-nitroisatoic Modification of 2′ OH of single-stranded Local nucleotide
anhydride) nucleotides flexibility
UV (254 nm) Cross-linking Spatial proximity
6sG (6-thioguanosine) Short range (3 Å) photo-cross-linking of Spatial proximity
molecules
4sU (4-thiouridine) Short range (3 Å) photo-cross-linking of Spatial proximity
molecules
APA (azidophenacyl) Long range (9–10 Å) photo-cross-linking of Spatial proximity
molecules
The name of the reagent is indicated in the first column, whereas the second column indicates the specificity of the
reagent and the third one the structural information given by the use of the reagent
Structural Probing of Small Non-Coding RNAs 121
2 Materials
11. 96 % EtOH.
12. Glycogen (10 mg/ml).
13. 70 % EtOH.
14. Refrigerated centrifuge adapted for 1.5 ml microtubes.
15. Milli-Q water.
9. Heating block.
10. 0.1 M EDTA, pH 8.0.
11. Phenol–chloroform–isoamyl alcohol (25:24:1).
12. 96 % EtOH.
13. Glycogen (10 mg/ml).
14. 3 M NaOAc.
15. 70 % EtOH.
16. 25 mM potassium borate, pH 7.0 (see Note 1).
3 Methods
3.1.2 RNA Purification 1. Add 30 μl of gel loading buffer to the transcription reaction.
on Denaturing Acrylamide 2. Heat at 96 °C for 2 min to denature secondary structures,
Gel then place immediately on ice to prevent renaturation
(see Note 5).
3. Purify the RNA transcript on a polyacrylamide–urea gel elec-
trophoresis (PAGE) in 1× TBE buffer (see Note 6).
4. After migration, visualize the RNA transcript by UV shadow-
ing and cut it out from the gel (see Note 7).
5. Add 100–200 μl of 1× elution buffer to the gel slice in a 1.5 ml
microtube and incubate for 30 min at 37 °C.
6. Transfer the supernatant containing the eluted RNA in a new
microtube and repeat the elution procedure (see Note 8).
7. Pool the two elution fractions and proceed with phenol–
chloroform extraction.
8. Precipitate the RNA transcript by addition of three volumes of
96 % EtOH and in the presence of 5 μg of glycogen. Incubate
for 15 min at −80 °C and centrifuge for 15 min at 15,000 × g
at 4 °C.
9. After centrifugation, the RNA pellet is washed with 70 %
EtOH, dried, and dissolved in Milli-Q water.
3.2 Enzymatic 1. Prepare RNase T1 dilutions in 1× Tris Buffer at 0.25, 0.5, and
Probing of RNA 1 U/μl starting from the stock solution at 500 U/μl. Store the
Transcripts dilutions at 4 °C.
3.2.1 Enzymes 2. Prepare RNase T2 dilutions in 1× Tris Buffer at 0.5, 1, and
Preparation 2 U/μl starting from the stock solution at 2.5 U/μl.
3. Prepare RNase V1 dilutions in 1× Tris Buffer at 10−4, 5.10−4,
and 10−3 U/μl starting from the stock solution at 10−2 U/μl.
3.2.2 Enzymatic Probing 1. For one enzymatic probing reaction, mix on ice 0.2–2 pmoles
of Unlabeled RNA of renatured unlabeled RNA transcript with 2 μg of total yeast
Transcripts tRNA and 1 μl of 10× buffer D. Adjust the final volume with
water to 9 μl (see Note 13).
2. Add 1 μl of diluted RNase T1, T2, or V1.
3. Incubate each assay for 6 min at 20 °C (see Note 14).
4. Stop RNase digestions by addition of 4 μl of 0.1 M EDTA, 1 μl
of yeast tRNAs (2 μg/μl), and 100 μl H2O and proceed with
phenol extraction.
5. Ethanol-precipitate the digestion products in the presence of
0.15 M NaoAc and 0.5 μg of glycogen. Wash samples with
70 % ethanol, dry them, and dissolve them in 3 μl of 25 mM
potassium borate.
3.2.3 Enzymatic Probing 1. For one probing reaction, mix 25,000–50,000 cpm of labeled
of End-Labeled RNA RNA with 1 μl of 10× buffer D, 1 μl of tRNAs (2 μg/μl), and
Transcripts Milli-Q water to a final volume of 9 μl.
2. Incubate this RNA mixture for 10 min at 65 °C and slowly
cool down to room temperature for renaturation.
3. Add 1 μl of diluted RNase T1, T2, or V1, or 1 μl of 1× Tris
buffer for the control reaction.
4. Incubate each assay exactly 6 min at 20 °C (see Note 14).
5. Stop RNase digestions by the addition of 4 μl of 0.1 M EDTA,
pH 8.0, 1 μl of yeast tRNAs (20 μg/μl), and 100 μl of H2O
and proceed with phenol–chloroform extraction.
128 Jean-Vincent Philippe et al.
3.3.2 Chemical 1. For one chemical probing reaction, mix on ice 0.2–2 pmoles of
Modifications of Unlabeled renatured unlabeled RNA transcript with 2 μg of total yeast
RNA Transcripts tRNA (2 μg/μl) and 1 μl of 10× buffer D. Adjust the final
volume with water up to 9 μl (see Note 13).
2. Add 4 μl of 10× DMS/ke buffer (for DMS and kethoxal
modifications) or 10× CMCT buffer (for CMCT modifications)
and 26 μl of H2O to the reaction.
3. Incubate for 20 min at 20 °C (or for 10 min at 4 °C for Pb2+
reactions).
4. Add 1 μl of diluted DMS/EtOH solution, 20 μl of CMCT
solution, 4 μl of kethoxal solution or 1 μl of lead (II) acetate
and mix.
5. Incubate for exactly 6 min at 20 °C for DMS, CMCT, and keth-
oxal modifications and for 5 min for lead cleavages (see Note 14).
3.3.3 Stopping Reactions 1. Stop DMS modifications by adding 10 μl of DMS stop buffer
and 100 μl of 0.3 M sodium acetate. Proceed with phenol–
chloroform extraction and precipitate with 96 % EtOH in the
presence of 5 μg of glycogen.
2. Stop kethoxal modifications by adding 10 μl of 0.5 M potas-
sium borate, which stabilizes the kethoxal–guanine adduct.
Add 100 μl of 0.3 M sodium acetate and perform phenol–
chloroform extraction. Precipitate with 96 % EtOH in the
presence of 5 μg of glycogen.
3. Stop CMCT modifications by adding 100 μl of 0.3 M sodium
acetate and immediately proceed with phenol–chloroform
extraction followed by 96 % ethanol precipitation in the
presence of 5 μg of glycogen.
4. Stop lead cleavages by adding 5 μl of 0.1 M EDTA, pH 8.0
and 100 μl of 96 % EtOH. Incubate for 30 min at −80 °C and
Structural Probing of Small Non-Coding RNAs 129
3.4 Primer Extension 1. Label the primer (50–100 ng) by phosphorylation at the 5′
Analysis terminus in a mixture containing 1 μl of [γ-32P]-ATP, 1 μl of
T4 Polynucleotide kinase, and 1 μl of 10× T4 PNK buffer in a
3.4.1 Primer Labeling
10 μl total reaction. Incubate for 45 min at 37 °C.
2. Add 15 μl of Milli-Q water and separate the labeled primer from
the unincorporated [γ-32P] ATP by gel filtration on a Quick Spin
Column according to the manufacturer’s instructions.
3. Adjust the volume of the purified labeled primer with Milli-Q
water to a concentration of about 100,000–150,000 cpm/μl.
3.4.3 Primer Extension 1. Mix following to prepare the elongation mixture: 0.1 μl of
5 mM dNTPs, 0.25 μl of 10× AMV Reverse Transcriptase buf-
fer, 0.25 μl of AMV Reverse Transcriptase (2 U/μl dilution
freshly prepared before use), and 1.9 μl of H2O (see Note 20).
2. Add 2.5 μl of this elongation mix to the hybridization mix
from Subheading 3.4.2.
3. Perform first strand synthesis reaction by incubating the tubes
for 30 min at 42 °C. During this step, each modification or
cleaved position within the RNA matrix will stop the elongation
by AMV reverse transcriptase.
4. After the incubation at 42 °C, briefly spin the samples at room
temperature to collect droplets from the lid.
5. Stop the primer extension by adding 3 μl of the gel loading
buffer and store on ice.
3.4.4 Sequencing Ladder 1. Produce a sequencing ladder with the unmodified RNA and
Preparation the corresponding labeled primer: as four lanes (A, C, G and T)
are required, prepare a hybridization mixture containing 0.8–8
pmoles of renatured unlabeled RNA transcript, 1 μl of 10×
130 Jean-Vincent Philippe et al.
3.6 Gel Analysis 1. Analyze the gel either by manual reading or using the Semi-
and Structure Automated Footprinting Analysis (SAFA) software [8, 9].
Modeling 2. Generate a list of nucleotides recognized as single-stranded or
double-stranded nucleotides using the RNAse T1 ladder or the
sequencing ladder as sequence references (see Note 25). If using
SAFA, you can precisely quantify the intensity of each band on
the gel and normalize each band’s intensity to the respective
average lane intensity.
3. Use the most significant probing data as constraints in an
M-Fold search [15] for the RNA structure.
4. Choose among the proposed models the ones with the lowest
free energies (see Note 26) and the least discrepancies between
the different probing data (Fig. 1).
4 Notes
a b
DMS CMCT Keth T1 T2
UGCA U G C A-
1750 -
SL II
1775 -
SL III
1800 -
Progerin
1825 - 5’SS
SL IV
1850 -
SL V
Fig. 1 Structural probing of the Lamin A (LMNA) pre-mRNA in the region containing the exon 11 alternative 5′
splice site. RNAs of 638 nt bearing the 1824C>U mutation responsible for the Hutchinson Gilford Progeria
Syndrome (HGPS) were used to determine the structure around the exon 11 of LMNA alternative 5′ splice site
(5′SS), leading to a non-functional LMNA protein called progerin. These RNAs contain the exons 11 and 12 and
the intron 11. Chemical modifications by DMS, CMCT, and kethoxal and digestions with RNases T1 and T2 were
carried out as described in this chapter. Panel a shows the primer extension analyses. For each of the chemical
probes (gel on the left), the first lane corresponds to a control experiment performed in the absence of the probe,
whereas the two other lanes correspond to increasing amounts of the probe. Lanes U, G, C, and A correspond to
the sequencing ladder. For RNases digestions (gel on the right), a control experiment corresponding to undi-
gested RNA was fractionated (lane -) and increasing amounts of RNases T1 and T2 were used. The primer used
to perform primer extension allows the analysis of the secondary structure from positions 1750–1870 of the
LMNA RNA (see panel b). Splice Site (SS) and Stem Loop structures (SL) identified as in the structure model in
panel b are indicated on the side of the gels. Positions in the LMNA RNA are indicated on the left side of the gel.
Panel b shows the secondary structure model proposed based on thermodynamic considerations and on the
results of chemical and enzymatic probing shown for the most part in panel a. V1, T1, and T2 RNases cleavages
are represented by squares, diamonds, and circles, respectively. Nucleotides modified by DMS, CMCT, or Ke are
circled. The colors (red, orange, and green) and number of symbols indicate high, medium, or low extent of
cleavage, respectively. The position of the progerin 5′ splice site is indicated. The different predicted stem loop
structures are numbered II–IV
Acknowledgments
References
1. Warf MB, Berglund JA (2010) Role of RNA 3. Weinberg Z, Perreault J, Meyer MM, Breaker
structure in regulating pre-mRNA splicing. RR (2009) Exceptional structured noncoding
Trends Biochem Sci 35(3):169–178. RNAs revealed by bacterial metagenome analy-
doi:10.1016/j.tibs.2009.10.004 sis. Nature 462(7273):656–659. doi:10.1038/
2. Breaker RR (2012) Riboswitches and the RNA nature08586
world. Cold Spring Harb Perspect Biol 4(2). 4. Prusa J, Missak J, Kittrell J, Evans JJ, Tapprich
doi:10.1101/cshperspect.a003566 WE (2014) Major alteration in coxsackievirus
136 Jean-Vincent Philippe et al.
B3 genomic RNA structure distinguishes a 13. Aviran S, Pachter L (2014) Rational experi-
virulent strain from an avirulent strain. Nucleic ment design for sequencing-based RNA
Acids Res 42(15):10112–10121. doi:10.1093/ structure mapping. RNA 20:1864–1877.
nar/gku706 doi:10.1261/rna.043844.113
5. Ray SS, Pal SK (2013) RNA secondary struc- 14. Vasa SM, Guex N, Wilkinson KA, Weeks KM,
ture prediction using soft computing. IEEE/ Giddings MC (2008) ShapeFinder: a software
ACM Trans Comput Biol Bioinform 10(1):2– system for high-throughput quantitative anal-
17. doi:10.1109/TCBB.2012.159 ysis of nucleic acid reactivity information
6. Ehresmann C, Baudin F, Mougel M, Romby P, resolved by capillary electrophoresis. RNA
Ebel JP, Ehresmann B (1987) Probing the 14(10):1979–1990. doi:10.1261/rna.
structure of RNAs in solution. Nucleic Acids 1166808
Res 15(22):9109–9128 15. Zuker M (2003) Mfold web server for nucleic
7. Weeks KM (2010) Advances in RNA structure acid folding and hybridization prediction.
analysis by chemical probing. Curr Opin Nucleic Acids Res 31(13):3406–3415
Struct Biol 20(3):295–304. doi:10.1016/j. 16. Mereau A, Fournier R, Gregoire A, Mougin A,
sbi.2010.04.001 Fabrizio P, Luhrmann R, Branlant C (1997)
8. Das R, Laederach A, Pearlman SM, Herschlag An in vivo and in vitro structure-function anal-
D, Altman RB (2005) SAFA: semi-automated ysis of the Saccharomyces cerevisiae U3A
footprinting analysis software for high- snoRNP: protein-RNA contacts and base-pair
throughput quantification of nucleic acid foot- interaction with the pre-ribosomal RNA.
printing experiments. RNA 11(3):344–354. J Mol Biol 273(3):552–571. doi:10.1006/
doi:10.1261/rna.7214405 jmbi.1997.1320
9. Simmons K, Martin JS, Shcherbakova I, 17. Talkish J, May G, Lin Y, Woolford JL Jr,
Laederach A (2009) Rapid quantification and McManus CJ (2014) Mod-seq: high-
analysis of kinetic *OH radical footprinting throughput sequencing for chemical probing
data using SAFA. Methods Enzymol 468:47– of RNA structure. RNA 20(5):713–720.
66. doi:10.1016/S0076-6879(09)68003-4 doi:10.1261/rna.042218.113
10. Mitra S, Shcherbakova IV, Altman RB, Brenowitz 18. Kielpinski LJ, Vinther J (2014) Massive
M, Laederach A (2008) High-throughput single- parallel-sequencing-based hydroxyl radical
nucleotide structural mapping by capillary auto- probing of RNA accessibility. Nucleic Acids
mated footprinting analysis. Nucleic Acids Res Res 42(8):e70. doi:10.1093/nar/gku167
36(11):e63. doi:10.1093/nar/gkn267 19. Balzer M, Wagner R (1998) A chemical modi-
11. Mortimer SA, Trapnell C, Aviran S, Pachter L, fication method for the structural analysis of
Lucks JB (2012) SHAPE-Seq: high-throughput RNA and RNA-protein complexes within liv-
RNA structure analysis. Curr Protoc Chem Biol ing cells. Anal Biochem 256(2):240–242.
4(4):275–297. doi:10.1002/9780470559277. doi:10.1006/abio.1997.2499
ch120019 20. Silberklang M, Gillum AM, RajBhandary UL
12. Loughrey D, Watters KE, Settle AH, Lucks JB (1979) Use of in vitro 32P labeling in the
(2014) SHAPE-Seq 2.0: systematic optimiza- sequence analysis of nonradioactive tRNAs.
tion and extension of high-throughput chemi- Methods Enzymol 59:58–109
cal probing of RNA secondary structure with 21. England TE, Uhlenbeck OC (1978) 3′-terminal
next generation sequencing. Nucleic Acids Res labelling of RNA with T4 RNA ligase. Nature
42(21). doi:10.1093/nar/gku909 275(5680):560–561
Part III
Abstract
The RNome of a cell is highly diverse and consists besides messenger RNAs (mRNAs), transfer RNAs
(tRNAs), and ribosomal RNAs (rRNAs) also of other small and long transcript entities without apparent
coding potential. This class of molecules, commonly referred to as non-protein-coding RNAs (ncRNAs),
is involved in regulating numerous biological processes and thought to contribute to cellular complexity.
Therefore, much effort is put into their identification and further functional characterization. Here we
provide a cost-effective and reliable method for cDNA library construction of small RNAs in the size range
of 20–500 residues. The effectiveness of the described method is demonstrated by the analysis of ribosome-
associated small RNAs in the eukaryotic model organism Trypanosoma brucei.
Key words cDNA library, Reverse transcription, Small RNA, Non-coding RNA
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_13, © Springer Science+Business Media New York 2015
139
140 Jennifer Gebetsberger et al.
2 Materials
Fig. 1 Pie chart of deep sequencing results. A specialized cDNA library was con-
structed for small ribosome-associated RNAs in Trypanosoma brucei. The cDNA
library was deep-sequenced using the Illumina platform for single end reads at
a maximum read length of 100 base pairs. Sequence reads were grouped
according to their genomic origin (snoRNA for small nucleolar RNA, ncRNA for
non-coding RNA, rRNA for ribosomal RNA, tRNA for transfer RNA, and SRP RNA
for signal recognition particle RNA). The relative distribution is indicated in reads
per million (RPM). Sequencing results of ribosome-bound small RNAs extracted
from (a) exponentially growing cultures are shown and compared to (b) RNAs
extracted from heat stressed T. brucei cells. According to these results, different
conditions led to a different ribosome-bound small RNome
2.2 cDNA Library 1. Poly(A) Polymerase kit (Epicentre, Illumina), including 10×
Generation reaction buffer: 0.5 M Tris–HCl, pH 8.0, 2.5 M NaCl,
100 mM MgCl2.
2. Tobacco Acid Pyrophosphatase (Epicentre, Illumina), including
10× reaction buffer: 500 mM NaOAc, pH 6.0, 10 mM EDTA,
1 % (v/v) β-mercaptoethanol, 0.1 % (v/v) Triton X-100.
3. T4 RNA Ligase, including 10× reaction buffer: 500 mM Tris–
HCl, pH 7.5, 100 mM MgCl2, 100 mM DTT, 10 mM ATP.
4. Superscript™ II Reverse Transcriptase (Invitrogen™, Life
Technologies), including 5× First strand buffer: 250 mM Tris–
HCl, pH 8.3, 375 mM KCl, 15 mM MgCl2.
142 Jennifer Gebetsberger et al.
Fig. 2 Flowchart of the cDNA library construction method reported. As starting material for cDNA library prepa-
ration, any RNA entity can be used. In the presented case, ribosomes were isolated from Trypanosoma brucei
via density gradient centrifugation yielding non-translating subunits and monosomes (40S, 60S, 80S) as well
as actively translating polyribosomes. RNA that co-purified with these fractions was used for cDNA library
construction, allowing the analysis of the small RNA interactome of T. brucei ribosomal particles. In a first step
RNA is size-separated by polyacrylamide gel electrophoresis (PAGE) in order to enrich for small RNAs (black).
Size-selected RNA is 3′ C-tailed and 5′ adaptor ligated prior to reverse transcription. The resulting cDNA is
amplified by PCR reaction and purified via native PAGE before sequencing. In order to check the quality of the
cDNA library, diagnostic Sanger sequencing can be performed prior to high-throughput sequencing. Whereas
the type of working material is listed on the left side of the figure, the different steps of cDNA library construc-
tion are given on the right together with the corresponding method section
3 Methods
3.1 RNA Preparation 1. For RNA extraction from a total extract (tissue or cultured
cells), add 1 volume of P/C/I (see Note 1) and vortex for
1 min.
2. Centrifuge for 1 min at 16,000 × g and transfer upper phase
into a new precooled 1.5 ml reaction tube.
3. Add 1 volume of chloroform, vortex for 1 min, and centrifuge
again for 1 min at 16,000 × g.
4. Transfer upper phase again into a new precooled 1.5 ml reac-
tion tube.
5. Add 2.5 volumes of precooled (−20 °C) absolute EtOH and
300 mM NaOAc, pH 5.4, invert several times, and put the
samples for 20 min at −80 °C for precipitation.
6. Centrifuge for 20 min at 16,000 × g at 4 °C and wash pellet
with precooled (−20 °C) 70 % EtOH.
7. Remove EtOH and let the pellet dry for 10 min on ice.
8. The RNA (5–200 μg are recommended) is mixed with RNA
loading dye and loaded in a single slot of a denaturing 8 %
polyacrylamide gel containing 7 M urea (gel thickness 2 mm).
Also load an RNA molecular weight marker, enabling the cor-
rect size detection of the RNA of interest.
9. Run the gel with 200 V for 2.5 h.
10. Stain the gel with EtBr in 1× TBE buffer for 10 min.
11. After gel staining, excise the RNA in the size-range of interest
from the gel by cutting it out with a sterile scalpel (see Note 6).
12. Dice the excised gel piece into 1 mm3 cubes with a sterile
scalpel, which will enhance subsequent elution of the RNA
from the gel.
cDNA Library of Small RNAs 145
13. Carefully transfer the gel pieces into a fresh 1.5 ml reaction
tube.
14. Add elution buffer until all gel slices are submerged and incu-
bate overnight with constant shaking at 4 °C for the RNA to
passively elute (see Note 7).
15. Remove the supernatant using a pipette and transfer the solu-
tion into a fresh 1.5 ml reaction tube.
16. Centrifuge the sample at 2,300 × g at 4 °C for 5 min to pellet
potential gel pieces.
17. Transfer the elution buffer containing the eluted RNA into a
clean reaction tube (see Notes 7 and 8).
18. Precipitate the eluate with 2.5 volumes of absolute ethanol for
at least 20 min at −80 °C (see Note 9).
19. Centrifuge RNA at 16,000 × g in a tabletop centrifuge at 4 °C
for 30 min.
20. Wash the pellet with 70 % ethanol and subsequently dissolve it
in 11 μl water.
21. Use 1 μl of dissolved RNA to spectrophotometrically determine
its concentration.
3.2.3 5′ Adaptor Ligation The aim of the 5′ adaptor ligation is the introduction of a specific
sequence allowing subsequent PCR amplification. Furthermore,
barcodes (see Note 13) for different conditions can be integrated
at this step, enabling sequence assignment after deep sequencing.
Since T4 RNA ligase can only join RNA ends, chimeric DNA/
RNA oligonucleotides, carrying three RNA nucleosides at their 3′
end, are used as 5′ adaptors (see Subheading 2 and Note 4).
1. Perform the ligation reaction in a final volume of 20 μl, con-
taining 1× reaction buffer and 20 μM 5′ adaptor.
2. Pre-incubate at 65 °C for 5 min and add 40 U of ribonuclease
inhibitor and 10 U of T4 RNA ligase. Incubate at 4 °C
overnight.
3. After overnight incubation, add 10 U of T4 RNA ligase and
incubate for an additional hour on ice.
4. P/C/I-extract the 5′ adaptor-ligated RNA, ethanol-precipitate
(see Subheading 3.1, steps 1–7), and resuspend in 10 μl water.
3.2.4 Reverse In this step, the 5′ adaptor-ligated and 3′ C-tailed RNA is reverse-
Transcription transcribed into cDNA employing anchored oligo(dG) primers.
1. Pre-incubate 10 μl of RNA from Subheading 3.2.3 in the pres-
ence of 5 μM anchored primer and 2 mM dNTPs at 65 °C for
5 min.
2. Immediately chill on ice.
3. Add 4 μl of 5× first strand buffer, 10 mM DTT, and 40 U of
ribonuclease inhibitor.
4. Incubate the reaction at 42 °C for 2 min.
5. Add 200 U of reverse transcriptase and incubate the reaction
at 42 °C for 1 h.
3.2.5 PCR Amplification 1. Perform the PCR reaction in a 50 μl reaction volume containing
of the cDNA 4 μl of cDNA from Subheading 3.2.4, 1 μM of both forward
and reverse primer (see Note 14), 0.1 mM dNTPs and 2 U of
Taq-DNA polymerase in 1× PCR buffer.
2. Use following cycles for the PCR reaction: Denaturation step
for 2 min at 94 °C; 20 cycles: 1 min at 94 °C, 1 min at 54 °C,
1 min 72 °C; Final elongation step for 5 min at 72 °C.
3.2.6 Purification of PCR 1. Mix 50 μl of PCR reaction with 6× DNA loading dye and run
Product via Native PAGE on an 8 % polyacrylamide gel.
2. Stain the gel with 0.4 μg/ml EtBr in 1× TBE buffer for 10 min
and excise the cDNA (see Note 15).
3. Elute in elution buffer overnight with constant shaking at
4 °C.
4. Spin at 2,300 × g at 4 °C for 5 min and transfer the eluate into
a fresh reaction tube.
cDNA Library of Small RNAs 147
3.3 Sequencing In order to check the cDNA quality, including correct adaptor
ligation and sufficient insert lengths, diagnostic Sanger-sequencing
3.3.1 Diagnostic Sanger
of ~50–100 clones is recommended.
Sequencing
1. Ligate cDNA into pGEM-T vectors and transform into electro-
competent E. coli cells (see Note 16).
2. Perform colony-PCR and purification of the resulting PCR
products.
3. Sequence purified PCR-products for example by using the
BigyDye terminator cycle sequencing reaction kit (PE Applied
Biosystems).
3.3.2 Preparation 1. Add adaptors required for deep-sequencing via PCR (see
for Deep-Sequencing Note 17). Contact the deep-sequencing platform of your
choice for appropriate primer design.
2. PAGE-purify amplified cDNA carrying the attached sequenc-
ing adaptors and precipitate with ethanol (as described in
Subheading 3.2.6).
3. Measure cDNA concentration spectrophotometrically and
dilute according to the company’s requirements.
4 Notes
1. The pH of the P/C/I mixture used for the first RNA extraction
should be acidic (optimal pH 4.5–5) in order to denature DNA
that otherwise could possibly be present as a contamination.
At pH 7, RNA and DNA will be in the water phase. At pH 5,
DNA is partly denatured and will be directed to the organic
phase, while RNA remains in the water phase.
2. All solutions should be prepared in deionized water.
3. Store buffers at −20 °C if not otherwise mentioned.
4. T4 RNA ligase uses two RNA strands as substrate. Therefore,
the last three nucleotides at the 3′ end of the 5′ adaptor oligo-
nucleotide should be ribonucleotides to ensure efficient
ligation.
5. In order to increase the biological relevance of the starting
material and to avoid amplification of putative cellular degra-
dation products, isolation of ncRNA–protein complexes via
density gradients [13] or affinity purification [7] is highly
recommended.
148 Jennifer Gebetsberger et al.
6. Always clean the scalpel with ethanol after cutting out a gel
piece with the desired RNA before cutting out the next piece
to avoid RNA cross-contaminations. Note that the gel slices
should neither be squeezed into the reaction tube nor be
allowed to swim in the elution buffer.
7. Take care that no gel slice is taken along with the elution buffer.
8. If a second round of elution is desired, freeze the gel slices at
−20 °C before adding elution buffer. Freezing and thawing
will enhance the amount of eluted RNA.
9. Since the amount of precipitated RNA can be little, GlycoBlue
(Ambion) can be used as nucleic acid co-precipitant. This
increases the pellet visibility.
10. The CTP concentration depends on the amount and length of
size-separated RNA. In general 75 pmol of CTP per picomole
of RNA is recommended.
11. 1 U of TAP per picomole of RNA is recommended.
12. Spectroscopic determination of RNA concentration at this step
is not recommended due to the presence of unincorporated
CTP from the C-tailing step.
13. Barcodes can be introduced at the 3′ end of the 5′ adaptor.
Due to possible deep sequencing errors, a minimum of four
nucleotides is recommended.
14. Alternatively, specific primers for subsequent deep-sequencing
can be used. Since they should be compatible with the chosen
sequencing platform, contact the sequencing facility for optimal
primer design.
15. Consider that the size-range of the cDNA corresponds to the
length of size-selected RNA of Subheading 3.1 plus the length
of the 5′ and 3′ adaptors.
16. Here it is advantageous to use cells carrying a lacZ deletion
mutant (e.g., One Shot® TOP10 Electrocomp™ E. coli provided
by Invitrogen), facilitating rapid and convenient detection of
recombinant cells via blue white screening.
17. To skip the second PCR step, the use of appropriate deep-
sequencing adaptors at Subheading 3.2.5 is recommended.
Acknowledgement
This research was partly supported by the NCCR “RNA & Disease”
funded by the Swiss National Science Foundation. Additional grant
support derives from the Swiss National Science Foundation
(31003A_143388/1) and the Austrian Science Fund FWF (project
number: Y315) to N.P.
cDNA Library of Small RNAs 149
References
1. Tuck AC, Tollervey D (2011) RNA in pieces. from ribosome-associated small RNAs by deep-
Trends Genet 27:422–432 sequencing data analysis. Nucleic Acids Res
2. Gebetsberger J, Polacek N (2013) Slicing 40:4013–4024
tRNAs to boost functional ncRNA diversity. 8. Pircher A, Gebetsberger J, Polacek N (2015)
RNA Biol 10:1798–1806 Ribosome-associated ncRNAs: An emerging
3. Madej MJ, Alfonzo JD, Huttenhofer A (2007) class of translation regulators. RNA Biol 11, in
Small ncRNA transcriptome analysis from press
kinetoplast mitochondria of Leishmania taren- 9. Huttenhofer A, Vogel J (2006) Experimental
tolae. Nucleic Acids Res 35:1544–1554 approaches to identify non-coding RNAs.
4. Lung B, Zemann A, Madej MJ, Schuelke M, Nucleic Acids Res 34:635–646
Techritz S, Ruf S, Bock R, Huttenhofer A 10. Erlacher MD, Chirkova A, Voegele P, Polacek
(2006) Identification of small non-coding N (2011) Generation of chemically engi-
RNAs from mitochondria and chloroplasts. neered ribosomes for atomic mutagenesis
Nucleic Acids Res 34:3842–3852 studies on protein biosynthesis. Nat Protoc 6:
5. Gebetsberger J, Zywicki M, Kunzi A, Polacek N 580–592
(2012) tRNA-derived fragments target the ribo- 11. Mattick JS, Makunin IV (2006) Non-coding
some and function as regulatory non-coding RNA. Human Mol Genet 15(Spec No 1):
RNA in Haloferax volcanii. Archaea 2012:260909 R17–29
6. Pircher A, Bakowska-Zywicka K, Schneider L, 12. Martin G, Keller W (1998) Tailing and 3’-end
Zywicki M, Polacek N (2014) An mRNA- labeling of RNA with yeast poly(A) polymerase
derived noncoding RNA targets and regulates and various nucleotides. RNA 4:226–230
the ribosome. Mol Cell 54:147–155 13. Rederstorff M, Huttenhofer A (2011) cDNA
7. Zywicki M, Bakowska-Zywicka K, Polacek N library generation from ribonucleoprotein par-
(2012) Revealing stable processing products ticles. Nat Protoc 6:166–174
Chapter 14
Abstract
UV cross-linking and immunoprecipitation coupled to high-throughput sequencing (CLIP-seq) is used to
characterize RNA targets of RNA binding proteins (RBP) in a large scale manner. This powerful method
allows the stringent purification of direct RNA binding sites of RBPs in living cells. Here, we describe in
detail the protocol we employed to identify RNA targets of the human RNA helicase eIF4AIII.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_14, © Springer Science+Business Media New York 2015
151
152 Jérôme Saulière and Hervé Le Hir
2 Materials
For RNA experiments, you must wear gloves and work on ice. To
avoid RNA degradation, all the buffers are also ice-cold except for
running buffers or buffers used for enzymatic reactions.
2.3 Cell Lysate 1. PXL lysis buffer: 1× DPBS, 0.1 % SDS, 0.5 % NP40, 0.5 %
Preparation sodium deoxycholate.
and RNAse Treatment 2. Protease inhibitor.
3. RQ1 RNase-free DNase (Promega).
4. RNasin (Promega).
5. RNase T1.
6. Thermomixer.
2.7 Illumina DNA 1. NEB Next Multiplex Small RNA Library Prep Set for Illumina
Library (NEB).
2. 10 % TBE gel.
3. TBE Running buffer.
4. SYBR Gold nucleic acid gel stain (Life Technologies).
5. Costar Spin-X column (Sigma).
3 Methods
3.1 Cell Culture 1. Grow HeLa cells (37 °C, 5 % CO2) until complete confluence
and UV Cross-Linking in 15 cm dishes in DMEM supplemented with 10 % FBS and
100 U/ml penicillin and 100 μg/ml streptomycin.
2. Take off medium and add 4 ml of 1× DPBS.
3. Places dishes lid off on ice in a tray.
154 Jérôme Saulière and Hervé Le Hir
3.6 RNA Recovery 1. Cut nitrocellulose membrane above the size of the free protein
and Purification to purify the small gel-shifted RNA with a clean scalpel
(see Note 12).
2. Chop it in tiny pieces (2–3 mm2) with a scalpel and place them
in a 1.5 ml microtube.
3. Add 200 μl of a preheated to 37 °C solution of proteinase K
(2 mg/ml in buffer 1).
4. Incubate for 30 min at 60 °C at 800 rpm.
5. Add 200 μl of a preheated to 37 °C solution of proteinase K
(2 mg/ml in buffer 2).
6. Incubate for 30 min at 60 °C at 800 rpm.
7. Short spin and cool down to room temperature.
156 Jérôme Saulière and Hervé Le Hir
3.7 Illumina DNA The synthesis of Illumina DNA libraries from small RNA is per-
Library formed with the NEBNext Multiplex Small RNA kit compatible
with Illumina high-throughput sequencing. We carefully follow
the manufacturer’s instructions. All the reactions are performed in
0.2 ml PCR tubes in a ThermoCycler.
1. Add 1 μl of 3′SR adaptor to the 6 μl small RNAs purified by
CLIP (see Note 14).
2. Incubate in a preheated ThermoCycler for 2 min at 70 °C. Chill
on ice.
3. Add 10 μl of 3′ Ligation Reaction Buffer and 3 μl of 3′ Ligation
Enzyme Mix. Total volume should be of 20 μl.
4. Incubate for 1 h at 25 °C.
5. Add 4.5 μl of Nuclease-free water and 1 μl of Multiplex SR RT
Primer.
6. Incubate for 5 min at 75 °C, 15 min at 37 °C and 15 min at
25 °C.
7. Denature 1 μl of 5′SR adaptor for 2 min at 70 °C in a pre-
heated ThermoCycler. Chill on ice.
CLIP-Seq 157
(bp)
190
160 Illumina DNA library
147
123 adaptors dimer
PCR primers
Fig. 1 Illumina DNA library on 10 % PAGE. Synth. RNA: DNA library prepared with a synthetic RNA of 40 nt
serving as a positive control (see Note 14). Samples 1 and 2: biological small RNA purified from the CLIP
experiment of eIF4AIII were used for the Illumina DNA library synthesis. Rectangles indicate the portion of the
gel from which PCR products were extracted and purified
4 Notes
References
1. Lee SR, Lykke-Andersen J (2013) Emerging eIF3 and activates translation. Proc Natl Acad
roles for ribonucleoprotein modification and Sci U S A 110:5903–5908
remodeling in controlling RNA fate. Trends 9. Le Hir H, Gatfield D, Izaurralde E, Moore MJ
Cell Biol 23:504–510 (2001) The exon-exon junction complex pro-
2. Moore MJ, Proudfoot NJ (2009) Pre-mRNA vides a binding platform for factors involved in
processing reaches back to transcription and mRNA export and nonsense-mediated mRNA
ahead to translation. Cell 136:688–700 decay. EMBO J 20:4987–4997
3. Le Hir H, Izaurralde E, Maquat LE, Moore 10. Roignant JY, Treisman JE (2010) Exon junc-
MJ (2000) The spliceosome deposits multiple tion complex subunits are required to splice
proteins 20-24 nucleotides upstream of mRNA Drosophila MAP kinase, a large heterochro-
exon-exon junctions. EMBO J 19:6860–6869 matic gene. Cell 143:238–250
4. Tange TO, Nott A, Moore MJ (2004) The 11. Sauliere J, Murigneux V, Wang Z, Marquenet
ever-increasing complexities of the exon junc- E, Barbosa I, Le Tonqueze O, Audic Y, Paillard
tion complex. Curr Opin Cell Biol L, Roest Crollius H, Le Hir H (2012) CLIP-
16:279–284 seq of eIF4AIII reveals transcriptome-wide
5. Andersen CB, Ballut L, Johansen JS, Chamieh mapping of the human exon junction com-
H, Nielsen KH, Oliveira CL, Pedersen JS, plex. Nat Struct Mol Biol 19:1124–1131
Seraphin B, Le Hir H, Andersen GR (2006) 12. Ule J, Jensen K, Mele A, Darnell RB (2005)
Structure of the exon junction core complex CLIP: a method for identifying protein-RNA
with a trapped DEAD-box ATPase bound to interaction sites in living cells. Methods
RNA. Science 313:1968–1972 37:376–386
6. Le Hir H, Andersen GR (2008) Structural 13. Ule J, Jensen KB, Ruggiu M, Mele A, Ule A,
insights into the exon junction complex. Curr Darnell RB (2003) CLIP identifies Nova-
Opin Struct Biol 18:112–119 regulated RNA networks in the brain. Science
7. Ashton-Beaucage D, Udell CM, Lavoie H, 302:1212–1215
Baril C, Lefrancois M, Chagnon P, Gendron P, 14. Darnell RB (2010) HITS-CLIP: panoramic
Caron-Lizotte O, Bonneil E, Thibault P, views of protein-RNA regulation in living cells.
Therrien M (2010) The exon junction com- Wiley Interdiscip Rev RNA 1:266–286
plex controls the splicing of MAPK and other 15. Licatalosi DD, Mele A, Fak JJ, Ule J, Kayikci
long intron-containing transcripts in M, Chi SW, Clark TA, Schweitzer AC, Blume
Drosophila. Cell 143:251–262 JE, Wang X, Darnell JC, Darnell RB (2008)
8. Chazal PE, Daguenet E, Wendling C, Ulryck HITS-CLIP yields genome-wide insights into
N, Tomasetto C, Sargueil B, Le Hir H (2013) brain alternative RNA processing. Nature
EJC core component MLN51 interacts with 456:464–469
Chapter 15
Abstract
Microarray technology has evolved to efficiently profile the expression of RNAs. However, analysis of small
non-coding RNAs (ncRNAs) is challenging due to their short length and highly divergent sequences with
large variation in GC content leading to very different hybridization properties. To overcome these chal-
lenges, LNA-modified oligonucleotides have been used to enhance and normalize the melting tempera-
ture (Tm) of capture probes, which allows sensitive profiling of small ncRNAs regardless of their sequence.
Here, we describe the isolation and labeling of small non-coding RNAs, as well as their hybridization to
microarrays with LNA-modified oligonucleotide probes using a semi-automated hybridization device.
Key words Small non-coding RNA, microRNA, Microarray, LNA, RNA isolation, Labeling,
Hybridization
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_15, © Springer Science+Business Media New York 2015
161
162 Michael Karbiener and Marcel Scheideler
2 Materials
3 Methods
3.1 RNA Isolation A crucial step in microarray analysis of small ncRNAs is the RNA
isolation. Although a number of convenient membrane/column-
based RNA preparation kits are currently available from several man-
ufacturers, it should be stressed that most of these systems come
along with a loss of RNAs smaller than 100 bp in length. Thus,
checking the compatibility of a particular product with small RNA
analysis is of fundamental importance. In contrast to membrane/
column-based RNA isolation approaches, procedures that include
an acidic phenol chloroform extraction are generally recognized to
preserve small RNAs. In particular, TRIzol reagent is a monophasic
solution of phenol and guanidine isothiocyanate that allows isolation
of most RNA species, regardless of molecular size [9, 10]. Thus, as
phenol/chloroform extraction requires only a little more hands-on
time than membrane/column-based systems, while being more
cost-effective, our workflow for microarray analysis of ncRNAs com-
prises RNA extraction using TRIzol reagent (Fig. 1).
3.1.1 Sample Harvesting 1. Homogenize and lyse biological material in TRIzol reagent
and RNA Extraction [11]. For tissue samples, use 1 ml TRIzol reagent per
50–100 mg of tissue (see Note 2). For adherent cells grown in
monolayer cell culture, use 1 ml TRIzol reagent per 10 cm2 of
culture area. For suspension cells grown in cell culture, use
1 ml TRIzol reagent per 5–10 × 106 cells.
2. Transfer the lysates to appropriate tubes (see Note 3).
3. Incubate the lysates at room temperature for at least 5 min,
which enables a complete dissociation of nucleoprotein
complexes.
4. Add 0.2 ml chloroform per ml of TRIzol reagent used. Cap
and vigorously shake the tubes by hand for at least 15 s to
allow RNA extraction into the aqueous phase.
5. Incubate the samples at room temperature for 3 min.
6. Centrifuge the samples at 12,000 × g at 4 °C for 15 min to
separate the organic phase from the aqueous phase.
7. Pipet the upper, colorless, aqueous phase into a fresh tube
(see Note 4).
164 Michael Karbiener and Marcel Scheideler
3.1.2 RNA Precipitation 1. Add 0.5 ml of 100 % isopropanol to the aqueous phase per ml
of TRIzol reagent used for homogenization. Shortly vortex
tubes to mix (see Note 5).
2. Incubate at room temperature for 10 min to allow RNA to
precipitate.
3. Centrifuge samples at 12,000 × g at 4 °C for 10 min
(see Note 6).
Microarray Analysis of Small Non-Coding RNAs 165
3.1.3 RNA Wash 1. Remove the supernatant from the tube, leaving only the RNA
pellet.
2. Wash the pellet with 1 ml of 75 % ethanol per ml of TRIzol
reagent used in the initial homogenization (see Note 7).
3. Centrifuge the tube at 7,500 × g for 5 min at 4 °C. Discard the
wash.
4. Vacuum or air-dry the RNA pellet for 5–10 min. Do not dry
the pellet by vacuum centrifuge (see Note 8).
3.1.4 RNA Resuspension 1. Resuspend the RNA pellet in RNase-free water (20–50 μl) by
passing the solution up and down several times through a
pipette tip.
2. Incubate in a water bath or heat block set at 55–60 °C for
10–15 min to completely dissolve the RNA.
3. Determine the concentration of each RNA sample. For subse-
quent analysis, prepare 0.25–1 μg total RNA in 3 μl (see Notes
9 and 10).
3.2 MicroRNA The Exiqon miRCURY LNA microRNA Hi-Power labeling kit
Labeling allows labeling of small to medium-sized RNA molecules with a
single fluorophore per molecule in a two-step process which does
not require any enrichment for small RNAs. The first step includes
a Calf Intestinal Alkaline Phosphatase step to remove 5′-phos-
phates of microRNAs. In the second step, a fluorescent label is
attached to the 3′-end of the small RNAs in the total RNA sample.
This is followed by an enzyme inactivation after which the sample
is ready for hybridization.
1. Place all kit components except enzymes on ice and thaw for
15–20 min [12].
2. Mix thoroughly by vortexing followed by brief centrifugation.
Do not thaw or vortex the enzyme. For enzymes, flick the
tubes and centrifuge briefly.
3. Combine following reagents in an RNAse-free microcentri-
fuge tube: 3 μl total RNA (as prepared in Subheading 3.1.4),
1 μl spike-in miRNA kit v2, 0.5 μl CIP buffer, 0.5 μl CIP
enzyme.
4. Mix thoroughly by pipetting up and down to ensure that all
reagents are mixed (see Note 11).
5. Incubate for 30 min at 37 °C, using a PCR cycler with heated
lid.
6. Stop the enzyme reaction and denature the RNA by incuba-
tion at 95 °C for 5 min followed by snap cooling on ice.
7. Leave the reaction on ice for at least 2 min and up to 15 min.
Briefly spin the tube after incubation on ice.
166 Michael Karbiener and Marcel Scheideler
3.3 Microarray The labeled samples can be hybridized on a microRNA chip spot-
Hybridization ted with locked nucleic acid (LNA)-modified oligonucleotides
with normalized melting temperature and GC content. LNA resi-
3.3.1 Hybridization
dues feature a bicyclic furanose unit locked in an RNA-mimicking
Station Preparation
sugar conformation [13]. This conformational restriction enables
unprecedented hybridization affinity for complementary single-
stranded RNA molecules, which makes LNA-modified oligonucle-
otides ideal for analysis and targeting of microRNAs [14].
1. Switch on the TECAN hybridization station.
2. Start the HS control manager software for the TECAN hybrid-
ization station.
3. Connect the hybridization station with the HS control man-
ager software by clicking on the plug button in the software.
4. Empty the waste bottles (usually big bottles behind the hybrid-
ization station).
5. Connect the high-pressure gas cylinder filled with nitrogen to
the hybridization station in order to fill the pipes of the hybrid-
ization station with pressure. Open the valve next to the bottle
first (totally) and then adjust the pressure at the second valve
to approximately 2.8–2.9 bar (see Note 13).
6. Slowly and carefully open the hybridization chambers of the
TECAN station.
Microarray Analysis of Small Non-Coding RNAs 167
3.3.2 Microarray 1. Prime the TECAN hybridization station and follow the HS
Prehybridization control manager instructions. Prime the first reagent used in
the loaded protocol (see step 13).
2. Insert the microarrays in the hybridization chambers you will
use. Note the barcodes, the used chamber position, and the
used TECAN hybridization station (in case several stations are
available).
3. Place a dummy slide in all unused hybridization chambers.
4. Mark all used hybridization chambers in the software
(see Note 15).
168 Michael Karbiener and Marcel Scheideler
3.3.3 Microarray After the last step of the microRNA labeling protocol
Hybridization (Subheading 3.2, step 13), samples are ready for hybridization.
These samples will be requested for the second sample injection
step in the hybridization protocol (Subheading 3.3.1, step 13)
(see Note 18).
1. Combine the two labeling reactions by pipetting Cy3 into Cy5
labeling reaction (see Note 19).
2. Flush the empty tube (red labeled sample) with 25 μl of 2×
preheated hybridization buffer (50 °C).
3. Add the flushing buffer to the collecting tube. The combined
labeling reaction should have a volume of 45–50 μl.
4. Wait for the software request “SAMPLE INJECTION” and
follow the software instructions.
5. Incubate the ready-for-hybridization labeled sample for 3 min
at 90 °C for denaturation.
6. Centrifuge the ready-for-hybridization labeled sample for
1 min at 6,000 × g and constantly protect it from light.
7. Open the first closure for injection.
8. Inject 50 μl of 1× preheated hybridization buffer (50 °C) using
reversed pipetting.
9. Inject 45 μl of the ready-for-hybridization labeled sample.
Take care not to inject any air bubbles (see Note 20).
10. Inject 10 μl of 1× preheated hybridization buffer (50 °C) using
reversed pipetting.
11. Confirm software requests and repeat the injection for all
chambers.
12. After 16 h hybridization, carefully open the hybridization
chambers and take out the hybridized microRNA chips, which
are ready for scanning. Protect microarrays from light and
store at room temperature.
3.3.4 Hybridization 1. Clean the hybridization chambers and the slide holder with
Station Cleaning compressed air.
2. Insert dummy slides in all chamber positions.
Microarray Analysis of Small Non-Coding RNAs 169
3. Make sure that all slides are appropriately fixed in the slide
holder.
4. Carefully close the lid of the hybridization station in order to
close the hybridization chambers.
5. Set the “RINSE” protocol to “DRYING BEFORE
FINISHING”.
6. Rinse the hybridization station and follow the HS control
manager instructions.
7. Proceed with the next hybridization or switch the device off.
4 Notes
Acknowledgments
References
1. Davison TS, Johnson CD, Andruss BF (2006) MicroRNA-26 family is required for human
Analyzing micro-RNA expression using micro- adipogenesis and drives characteristics of brown
arrays. Methods Enzymol 411:14–34 adipocytes. Stem Cells 32(6):1578–1590
2. Castoldi M, Benes V, Hentze MW, 8. Pichler M, Ress AL, Winter E, Stiegelbauer V,
Muckenthaler MU (2007) miChip: a microar- Karbiener M, Schwarzenbacher D et al (2014)
ray platform for expression profiling of microR- MiR-200a regulates epithelial to mesenchymal
NAs based on locked nucleic acid (LNA) transition-related gene expression and deter-
oligonucleotide capture probes. Methods mines prognosis in colorectal cancer patients.
43(2):146–152 Br J Cancer 110(6):1614–1621
3. Karbiener M, Fischer C, Nowitsch S, Opriessnig 9. Chomczynski P, Sacchi N (1987) Single-step
P, Papak C, Ailhaud G et al (2009) microRNA method of RNA isolation by acid guanidinium
miR-27b impairs human adipocyte differentia- thiocyanate-phenol-chloroform extraction.
tion and targets PPARgamma. Biochem Anal Biochem 162(1):156–159
Biophys Res Commun 390(2):247–251 10. Chomczynski P (1993) A reagent for the
4. Eduati F, di Camillo B, Karbiener M, Scheideler single-step simultaneous isolation of RNA,
M, Corà D, Caselle M et al (2012) Dynamic DNA and proteins from cell and tissue samples.
modeling of miRNA-mediated feed-forward Biotechniques 15(3):532–534, 536–537
loops. J Comput Biol 19(2):188–199 11. TRIzol Reagent Manual [Internet]. http://
5. Bach D, Fuereder J, Karbiener M, Scheideler tools.lifetechnologies.com/content/sfs/man-
M, Ress AL, Neureiter D et al (2013) uals/trizol_reagent.pdf
Comprehensive analysis of alterations in the 12. miRCURY LNA microRNA Array Hi-Power
miRNome in response to photodynamic treat- Labeling Kit [Internet]. http://www.exiqon.
ment. J Photochem Photobiol B 120:74–81 com/ls/Documents/Scientific/Hi-Power-
6. Karbiener M, Neuhold C, Opriessnig P, Labeling-kit-manual.pdf
Prokesch A, Bogner-Strauss JG (2011) 13. Vester B, Wengel J (2004) LNA (locked nucleic
MicroRNA-30c promotes human adipocyte acid): high-affinity targeting of complementary
differentiation and co-represses PAI-1 and RNA and DNA. Biochemistry (Mosc)
ALK2. RNA Biol [Internet] 8(5). http:// 43(42):13233–13241
www.es.landesbioscience.com/journals/rnabi- 14. Kauppinen S, Vester B, Wengel J (2006)
ology/article/16153/. Accessed 20 Jul 2011 Locked nucleic acid: high-affinity targeting of
7. Karbiener M, Pisani DF, Frontini A, Oberreiter complementary RNA for RNomics. Handb
LM, Lang E, Vegiopoulos A, et al (2014) Exp Pharmacol 173:405–422
Part IV
Abstract
MicroRNAs (miRNAs) are important regulators involved in most biological processes in eukarya. They
play critical roles in growth, development, signal transduction, or stress response by controlling gene
expression at the posttranscriptional level. Identification and characterization of miRNA-targeted mRNAs
is essential for the analysis of miRNA functions. In plants, the perfect complementarity between most
miRNAs and their targets enables the accurate predictions of their targets, while slicing of the targeted
mRNAs facilitates target validation through RNA Ligase-Mediated (RLM)-Rapid Amplification of cDNA
Ends (RACE) method. However, this method only determines the 5′-end of the cleavage product. To
more accurately validate the predicted target genes of miRNAs and exactly determine the cleavage sites
within the targets, an integrated strategy comprising RLM-RACE, Poly(A) Polymerase-Mediated (PPM)-
RACE, and qRT-PCR was developed. The efficiency of this method is illustrated by the precise sequence
validation of predicted target mRNAs of miRNAs in grapevine, citrus, peach, and other fruit crops. Our
on-going research indicates that RLM-RACE, PPM-RACE, and qRT-PCR are very effective in the verifi-
cation of sequences of miRNA targets obtained by Degradome sequencing. The protocol for RLM-RACE,
PPM-RACE, and qRT-PCR is rapid, effective, cheap, and can be completed within 2–3 days.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_16, © Springer Science+Business Media New York 2015
175
176 Chen Wang and Jinggui Fang
2 Materials
m7G AAAA(A)N3’
Ploy(A) RNA cap
5’miRNA
3’
miRNA-directed cleavge
m7G
cap 3’ 5’P AAAA(A)N3’
3’ 5’ 3’ 5’
5’ 3’ 5’ 3’
2.1 Total RNA 1. SDS lysis buffer: 50 mM Tris–HCl, pH 8 (see Note 1), 140 mM
Extraction NaCl, 10 mM EDTA, 4 % SDS (Sodium Dodecyl Sulfate), 3 %
PVP (polyvinylpyrrolidinone K30), 3 % ß-mercaptoethanol
(add just before use) (see Note 2). Store at 4 °C.
2. 3 M NaAC, pH 5.2 (see Note 3).
3. Ethanol, Isopropanol.
4. DNA Eraser (TaKaRa, Clontech), including DNA eraser buf-
fer: 40 mM Tris–HCl, pH 7.5, 80 mM MgCl2, 50 mM DTT.
5. RNase Inhibitor (see Note 4).
2.6 qRT-PCR 1. Rotor-Gene 3000 (Corbett Robotics) and software version 6.1.
of 3′- and 5′-End 2. SYBR® Green qRT-PCR Master Mix (Toyobo).
Cleavage Products
3. Specific forward and reverse primers.
3 Methods
3.2.2 Preparation 1. Resuspend one tube of SA-PMPs per isolation by gently flick-
of Streptavidin ing the bottom of the tube (see Note 11).
Paramagnetic Particles 2. Capture the SA-PMPs by placing the tube on the magnetic
(SA-PMPs) stand (see Note 12).
3. Carefully remove the supernatant (see Note 13).
4. Wash the SA-PMPs three times with 1.5 ml of 0.5× SSC per
wash. Following each wash, capture the SA-PMPs using the
magnetic stand and carefully remove the supernatant.
180 Chen Wang and Jinggui Fang
3.2.3 Elution of mRNA 1. Resuspend the final SA-PMP pellet in 1 ml of RNase-free water
and gently mix the particles by flicking the tube.
2. Magnetically capture the SA-PMPs and transfer the eluted
mRNA to a 2 ml microtube. Do not discard the particles
(see Note 14).
3.5 RLM-RACE Perform RLM-RACE reactions with the mirRacer 5′ primer and
and PPM-RACE gene-specific reverse primer (GSP1), and carry out PPM-RACE
reactions with the mirRacer 3′ primer and gene-specific forward
primer (GSP2) (see Note 19).
3.5.1 First Round In a sterile microtube, mix the following reagents for each sample:
Amplification 3 μl Hercules Hot-Start polymerase buffer, 4 μl dNTP mix
(2.5 mM), 2.5 units Ex Taq™ Hot-Start Version 4, 2 μl of template
cDNA derived from the 5′-adaptor ligated or from the PolyA-
tailed cleavage products, 25 pmol mirRacer 5′ primer or mirRacer
182 Chen Wang and Jinggui Fang
3.5.2 Second Round 1. Dilute an aliquot of the first round amplification to the 1:20 in
Amplification TE buffer.
2. In a sterile, 0.2 ml microtube, mix the following reagents on
ice: 3 μl Hercules Hot-Start polymerase buffer, 4 μl dNTP mix
(2.5 mM), 2.5 units Ex Taq™ Hot Start DNA polymerase
Version 4, 2 μl of the diluted first round amplification product
and 25 pmol of each primers mGSP2 and mGSP1. Set up a
control experiment without template.
3. Mix and heat in a DNA thermocycler for 5 min at 98 °C to
denaturate the first strand product and to activate the
polymerase.
4. Perform PCR using following reaction parameters: initial
denaturation step at 94 °C for 3 min; 25 cycles: Denaturation
at 94 °C for 10 s, Annealing at 65 °C for 10 s, Elongation at
72 °C for 1 min; Final extension at 72 °C for 10 min. Store
PCR products at 4 °C.
5. After amplification, separate the 3′- and 5′-PCR products on a
2.0 % agarose gel and stain with ethidium bromide (EtBr)
(Fig. 2) (see Note 20).
6. Cut the gel slices containing the distinct bands of the predicted
size and purify the DNA fragments using an agarose gel DNA
purification kit, according to the manufacturer’s instructions.
3.5.3 RLM-RACE Directly clone the DNA fragment purified with the TOPO TA
and PPM-RACE Products cloning Kit (Invitrogen).
Cloning and Sequencing
1. Add 4.5 μl of purified DNA, 0.5 μl pMD™ 19-T Vector, 5.0 μl
Solution I into a 0.2 ml sterile microtube. Mix and quickly spin
down.
2. Incubate at 16 °C for 1 h.
3. Transform E. coli DH5alpha competent cells with the ligation
product and plate on solid LB medium containing ampicillin.
Incubate overnight at 37 °C.
4. Pick positive clones and perform colony-PCR using the PCR-
specific primer pairs and PCR program as in step 2,
Subheading 3.5.
5. Sequence positive clones PCR-products.
6. Compare sequencing results with the predicted target genes
and identify the cleavage sites (Fig. 3).
a b
15.00 45.00
5⬘Vv-CB920070 5⬘Vv-EV228459
12.00 3⬘Vv-CB920070 36.00
3⬘Vv-EV228459
Relative expression
Relative expression
9.00 27.00
r=0.9668
6.00 18.00 r=0.9946
3.00 9.00
0.00 0.00
YL ML ST TE IN FL YB LB YL ML ST TE IN FL YB LB
c d 25.00
15.00
5⬘Vv-EC958215 5⬘Vv-EC934132
3⬘Vv-EC958215 20.00 3⬘Vv-EC934132
12.00
Relative expression
Relative expression
15.00
9.00
3.00 5.00
0.00 0.00
YL ML ST TE IN FL YB LB YL ML ST TE IN FL YB LB
Fig. 3 Products of RLM-RACE and PPM-RACE for miRNAs on their target genes. Line and columns denote the
products of RLM-RACE and PPM-RACE, respectively. YL, ML, ST, TE, IN, FL, YB, LB denote young leaf, mature
leaf, stem, tendril, inflorescence, flower, young berry, large berry; X position presents the diverse development
stages of grapevine; (a) and (b) represent the cleavage products of miR-159a and miR-159b on their corre-
sponding target genes respectively; (c) and (d) denote those of miR-319e on the two target genes. Line graphs
denote the 3′-end cleavage products of miRNAs on their target genes, bar diagrams represent the 5′-end
cleavage products. The size of 3′- and 5′-end cleavage products in (a)–(d) charts are 191,116; 158, 137; 187,
112; 163,114 respectively
184 Chen Wang and Jinggui Fang
4 Notes
Acknowledgements
References
1. Bartel B, Bartel DP (2003) MicroRNAs: at the 8. Mallory AC, Dugas DV, Bartel DP et al (2004)
root of plant development? Plant Physiol 13: MicroRNA regulation of NAC-domain targets
709–717 is required for proper formation and separation
2. Bartel DP (2004) MicroRNAs: genomics, bio- of adjacent embryonic, vegetative, and floral
genesis, mechanism, and function. Cell 116: organs. Curr Biol 14:1035–1046
281–297 9. Mallory AC, Reinhart BJ, Jones-Rhoades MW
3. Mallory AC, Vaucheret H (2004) MicroRNAs: et al (2004) MicroRNA control of
something important between the genes. Curr PHABULOSA in leaf development, impor-
Opin Plant Biol 7:120–125 tance of pairing to the micro RNA 5′ region.
4. Carrington JC, Ambros V (2003) Role of EMBO J 23:3356–3364
microRNAs in plant and animal development. 10. Aukerman MJ, Sakai H (2003) Regulation of
Science 301:336–338 flowering time and floral organ identity by a
5. Hunter C, Poethig RS (2003) Missing links: microRNA and its APETALA2-like target
miRNAs and plant development. Curr Opin genes. Plant Cell 15:2730–2741
Genet Dev 13:372–378 11. Kasschau KD, Xie Z, Allen E et al (2003)
6. Emery JF, Floyd SK, Alvarez J et al (2003) P1/HC-Pro, a viral suppressor of RNA
Radial patterning of Arabidopsis shoots by silencing, interferes with Arabidopsis devel-
Class III HD-ZIP and KANADI Genes. Curr opment and miRNA function. Dev Cell
Biol 13:1768–1774 4:205–217
7. Juarez MT, Kui JS, Thomas J et al (2004) 12. Sunkar R, Zhu JK (2004) Novel and stress-
MicroRNA-mediated repression of rolled leaf regulated microRNAs and other small RNAs
specifies maize leaf polarity. Nature 428:84–88 from Arabidopsis. Plant Cell 16:2001–2019
186 Chen Wang and Jinggui Fang
13. Mallory AC, Bartel DP, Bartel B (2005) targets identified by sequencing of the
MicroRNA-directed regulation of Arabidopsis Arabidopsis degradome. Curr Biol 18:
AUXIN RESPONSE FACTOR17 Is essential 758–762
for proper development and modulates expres- 22. Pantaleo V, Szittya G, Moxon S et al (2010)
sion of early auxin response genes. Plant Cell Identification of grapevine microRNAs and
17:1360–1375 their targets using high-throughput sequenc-
14. Allen E, Xie Z, Gustafson AM et al (2005) ing and degradome analysis. Plant J 62:
microRNA-directed phasing during transact- 960–976
ing siRNA biogenesis in plants. Cell 121:2071 23. German MA, Pillay M, Jeong DH et al (2008)
15. Chen X (2005) MicroRNA biogenesis and Global identification of microRNA-target
function in plants. FEBS Lett 579:5923–5931 RNA pairs by parallel analysis of RNA ends.
16. Berezikov E, Cuppen E, Plasterk RH (2006) Nat Biotechnol 26:941–946
Approaches to microRNA discovery. Nat 24. Thomson DW, Bracken CP, Goodal GJ (2011)
Genet 38:S2 Experimental strategies for microRNA target
17. Carthew RW, Sontheimer EJ (2009) Origins identification. Nucleic Acids Res 39(16):
and mechanisms of miRNAs and siRNAs. Cell 6845–6853
136:642–655 25. Lytle JR, Yario TA, Steitz JA (2007) Target
mRNAs are repressed as efficiently by
18. Kurihara Y, Watanabe Y (2004) Arabidopsis
microRNA-binding sites in the 5′-UTR as in
micro-RNA biogenesis through Dicer-like 1
the 3′-UTR. Proc Natl Acad Sci U S A 104:
protein functions. Proc Natl Acad Sci U S A
9667–9672
101:12753–12758
26. Kong YW, Cannell IG, de Moor CH et al
19. Jones-Rhoades MW, Bartel DP, Bartel B
(2008) The mechanism of micro-RNA-
(2006) MicroRNAs and their regulatory roles
mediated translation repression is determined
in plants. Annu Rev Plant Biol 57:19–53
by the promoter of the target gene. Proc Natl
20. Allen RS, Li JY, Alonso-Peral MM et al (2010) Acad Sci U S A 105:8866–8871
MicroR159 regulation of most conserved tar- 27. Hendrickson DG, Hogan DJ, McCullough
gets in Arabidopsis has negligible phenotypic HL et al (2009) Concordant regulation of
effects. Silence 1:18 translation and mRNA abundance for hun-
21. Addo-Quaye C, Eshoo TW, Bartel DP et al dreds of targets of a human microRNA. PLoS
(2008) Endogenous siRNA and miRNA Biol 7:e1000238
Chapter 17
Abstract
This chapter describes one of the most reliable quantitative assays to test the silencing of a possible target
gene by a specific miRNA using a luciferase reporter gene. The experimental procedure first consists in
cloning both the wild-type and mutated forms of the 3′UTR of the miRNA predicted mRNA target down-
stream of a firefly luciferase reporter. Next, each construct is co-transfected together with the miRNA into
HeLa cells, and the reporter expression is monitored. Changes in luciferase levels will indicate whether or
not a miRNA can bind to the UTR and regulate its expression.
Key words MicroRNA, Luciferase gene reporter assay, microRNA function, microRNA regulatory
element, microRNA targeting sequence, Target validation
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_17, © Springer Science+Business Media New York 2015
187
188 Thomas Clément et al.
2 Materials
2.1 Expression 1. pGL3 vector (Promega). pGL3 vector is used as a Firefly lucif-
Vector Preparation erase reporter vector. 3′ UTR fragments to test are cloned
downstream of the Firefly luciferase gene.
2.1.1 PCR Amplification
of Target and Pre-miRNA 2. pRL-SV40 vector (Promega). pRL-SV40 vector is used as a
Sequence Renilla luciferase control reporter vector. Both pGL3 and
pRL-SV40 vectors are co-transfected in cells and Renilla lucif-
erase activity is used to normalize transfection efficiency.
3. pCDNA3.1 vector (Life Technologies). pCDNA3.1 vector is
used to overexpress a gene of interest in mammalian cells (see
Note 2). In our case, pCDNA3.1, containing the full-length
sequence of the pre-miRNA of interest, leads to the overexpres-
sion of the corresponding miRNA in HeLa cells (see Note 3).
4. Oligonucleotides for the 3′UTR amplification:
Sense: 5′-GACTCTAGAGGACCCAAGTACTTTCCA-3′.
Antisense: 5′-GGCCGTCTAGATGTACTTTCCAATAATCT-3′.
Luciferase Reporter Assays 189
Fig. 1 Experimental design to test the interaction between hsa-miRNA-29b and its predicted target site in the
3′-UTR of the COL2A1 mRNA
2.2 Co-transfection 1. HeLa cells or any comparable human cell lines depending on
of Plasmids into the experimental design (see Note 3).
Mammalian Cells 2. Dulbecco’s Modified Eagle Medium supplemented with 10 %
fetal bovine serum, 2.9 mg/ml glutamine and 10 U/ml
Penicillin-Streptomycin.
3. 12-well plates for cell culture.
4. 1× PBS buffer: 140 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4,
1.8 mM KH2PO4.
5. JetPEI® Polyplus™ transfection reagent (see Note 5).
2.3 Dual-Luciferase 1. Dual-luciferase Reporter Assay Kit including PLB lysis buffer,
Reporter Assay LARII reaction buffer, and Stop&Glo buffer (Promega). Any
other comparable Dual-luciferase reporter assay kit allowing an
optimized system for the sequential assay of Firefly and Renilla
luciferase activities can be used.
2. GloMax®-96 Microplate Luminometer (Promega) featuring
a double automatic injector or any other comparable
luminometer.
3. Microwell Plates.
3 Methods
3.1.2 PCR Fragment 1. For the 3′UTR fragment digestion, incubate 1 μg of purified
Digestion, Plasmid PCR product for 1 h at 37 °C with 1 U of XbaI enzyme and
Linearization 1 μl of 10× digestion buffer in a final volume 10 μl.
2. Inactivate the enzyme by heating the mixture at 95 °C for
3 min.
3. For the pre-miRNA fragment digestion, incubate 1 μg of puri-
fied PCR product for 1 h at 37 °C with 1 U of KpnI and 1 U
of BamHI enzymes and 1 μl of 10× digestion buffer in a final
volume of 10 μl.
4. Inactivate the enzymes by heating the mixture at 95 °C for
3 min.
5. Digest and dephosphorylate simultaneously the pGL3-Con-
trol vector by XbaI and alkaline phosphatase. Incubate 1 μg of
pGL3 plasmid with 1 U of XbaI, 1 U of alkaline phosphatase,
and 1 μl of 10× digestion buffer in a final volume of 10 μl at
37 °C for 10 min.
6. Incubate the mixture at 95 °C for 3 min to inactivate the
enzymes.
7. Digest and dephosphorylate simultaneously the pCDNA 3.1
vector by KpnI, BamHI and alkaline phosphatase. Incubate
1 μg of pCDNA3.1 plasmid with 1 U of KpnI, 1 U of BamHI,
1 U of alkaline phosphatase, and 1 μl of 10× digestion buffer
in a final volume of 10 μl at 37 °C for 10 min.
8. Incubate the mixture at 95 °C for 3 min to inactivate the
enzymes.
3.1.3 Agarose Gel 1. Add 6× loading dye to the digested products and load them on
Electrophoresis a 2 % agarose gel.
2. Run the gel at 100 V for 35 min in 1× TBE buffer.
3. Following electrophoresis, stain the gel with Gel Red.
4. Cut the gel slice containing the DNA fragment of interest from
the agarose gel with a clean and sharp scalpel blade.
5. Retrieve and purify both linearized vectors and digested inserts
DNA fragments.
6. Quantify the DNA concentration using a NanoDrop spectro-
photometer (see Note 6).
4. Run the gel at 100 V for 35 min in order to check the DNA
amplification.
5. The PCR product is cleaned from matrix DNA using DpnI
enzyme, which degrades only methylated DNA. Incubate
10 μl of PCR product with 5 μl of DpnI and 2.5 μl of 10× buf-
fer in a final volume of 25 μl for 60 min at 37 °C.
6. Competent bacterial cells are transformed with 5 μl of DpnI
digestion product as described in Subheading 3.1.4.
3.2 Co-transfection 1. One day prior transfection, seed 1.105 HeLa cells in DMEM
of Plasmids into medium in 12-well plates. Fill as many wells as reactions, assays,
Mammalian Cells and controls to be performed. Incubate cells at 37 °C with 5 %
CO2 overnight.
2. For each transfection, combine 1 μg of pGL3 plasmid, 1 μg of
pCDNA3.1 plasmid and 5 ng of pRL-SV40 vector with 37 μl
of 150 mM NaCl in a final volume of 40 μl. We tested hsa-
miR-29b targeting the 3′UTR of COL2A1 mRNA and the
following combinations were used in our experimental design
(Fig. 1):
(a) pGL3-COL2A1 + pCDNA Ø + pRL SV40 (positive lucif-
erase activity control).
(b) pGL3-COL2A1 + pCDNA-pre-29b + pRL SV40 (assay).
(c) pGL3-Mutant + pCDNA-pre-29b + pRL SV40 (negative
control).
(d) pGL3-COL2A1 + pCDNA-pre-199a + pRL SV40 (nega-
tive control).
3. In parallel, combine 5 μl of JetPEI® transfection reagent with
35 μl of 150 mM NaCl (see Notes 5, 10, and 11).
4. Add the JetPEI® mixture on the plasmids mixture and vortex
thoroughly (see Note 12).
5. Incubate at room temperature for 20 min.
6. Add the 80 μl transfection mixture drop by drop to the cells
and incubate for 2–3 h at 37 °C with 5 % CO2.
7. To avoid JetPEI® toxicity on cells, change the culture medium
with DMEM pre-warmed to 37 °C.
8. Incubate cells overnight at 37 °C with 5 % CO2 (see Note 13).
5 Notes
Acknowledgment
References
1. Witkos TM, Koscianska E, Krzyzosiak WJ measurement of RNA secondary structure in
(2011) Practical Aspects of microRNA yeast. Nature 467:103–107
Target Prediction. Curr Mol Med 11: 5. Dweep H, Sticht C, Pandey P (2011) Gretz N:
93–109 miRWalk–database: prediction of possible
2. Lewis BP, Burge CB, Bartel DP (2005) miRNA binding sites by “walking” the genes of
Conserved seed pairing, often flanked by three genomes. J Biomed Inform 44:839–847
adenosines, indicates that thousands of 6. Doench JG, Petersen CP (2003) Sharp PA: siR-
human genes are microRNA targets. Cell NAs can function as miRNAs. Genes Dev 17:
120:15–20 438–442
3. Krek A, Grün D, Poy MN, Wolf R, Rosenberg 7. Ameres SL, Martinez J, Schroeder R (2007)
L, Epstein EJ, MacMenamin P, da Piedade I, Molecular basis for target RNA recognition and
Gunsalus KC, Stoffel M, Rajewsky N (2005) cleavage by human RISC. Cell 130:101–112
Combinatorial microRNA target predictions. 8. Krützfeldt J, Rajewsky N, Braich R, Rajeev KG,
Nat Genet 37:495–500 Tuschl T, Manoharan M, Stoffel M (2005)
4. Kertesz M, Wan Y, Mazor E, Rinn JL, Nutter Silencing of microRNAs in vivo with
RC, Chang HY, Segal E (2010) Genome-wide “antagomirs”. Nature 438:685–689
Chapter 18
Abstract
SiRNA induced gene silencing or RNA interference is a powerful tool to knock down gene expression and
perform gene function studies. In this chapter, we describe a basic method to silence gene expression by
transfecting a specific synthetic siRNA into mammalian HeLa cells.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_18, © Springer Science+Business Media New York 2015
199
200 Yoann Abel and Mathieu Rederstorff
Fig. 1 siRNA induced gene silencing. After transfection, the guide strand of the double-stranded siRNA is
loaded into the RISC complex, while the passenger strand is degraded. The loaded RISC targets a specific
mRNA which will be cleaved by Argonaute 2
2 Materials
3 Methods
3.1 Mammalian Cells 1. One day prior transfection, seed between 1 and 2 × 105 HeLa
Culture cells in DMEM medium in 6 wells plates. Incubate cells at
37 °C with 5 % CO2 overnight.
2. 1–5 h before transfection, change HeLa cells medium and add
3 ml of fresh DMEM medium.
siRNA Induced Gene Knockdown 201
4 Notes
Acknowledgment
References
1. Fire A, Xu SQ, Montgomery MK, Kostas SA, 2. Sontheimer EJ (2005) Assembly and function
Driver SE, Mello CC (1998) Potent and spe- of RNA silencing complexes. Nat Rev Mol Cell
cific genetic interference by double-stranded Biol 6:127–138
RNA in Caenorhabditis elegans. Nature 391:
806–811
Chapter 19
Abstract
Small non-coding RNAs (ncRNAs), less than 200 nucleotides in length, play important roles in various
biological processes, such as pre-mRNA splicing, pre-rRNA processing and modification, and gene expres-
sion regulation. However, characterization of small ncRNAs remains difficult mainly due to methodological
obstacles in selective reduction of these RNAs. Here we describe an approach to deplete small ncRNAs, in
principle any types of RNAs, using second generation antisense oligonucleotide-directed RNase H cleavage
pathway in human cells. This protocol includes oligonucleotide design, transfection, RNA preparation, and
target RNA detection.
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_19, © Springer Science+Business Media New York 2015
203
204 Xue-Hai Liang et al.
Fig. 1 Many small ncRNAs are highly structured and associate with RNP proteins. (a) C/D box snoRNPs. (b) H/ACA
box snoRNPs. (c) U1 snRNP. The proteins of different RNPs are depicted with circles, whereas RNAs are depicted
using lines. The dashed lines indicate the cellular RNAs that can base-pair with the small RNAs. The thick lines
represent the small RNA regions that can be potentially targeted by ASOs
2 Materials
Fig. 2 ASOs utilize RNase H1 mechanism to degrade target RNAs. The structure indicates a phosphorothioate-
backbone. 5-10-5 gapmer ASO containing five 2′-O-MOE modified ribonucleotides at each end and ten deoxy-
ribonucleotides in the middle. The RNA substrate and RNase H1 protein are depicted. The structures of the
modified nucleotides are shown
3 Methods
3.2 RNA Preparation 1. Remove medium from dishes containing the ASO treated cells
and Analysis and control cells and wash once with 1× DPBS.
2. Add 1 ml tri-reagent per dish and collect cell lysate using a
scraper.
3. Transfer to an microtube and prepare total RNA based on the
manufacturer’s protocol using Tri-Reagent.
4. Dissolve total RNA in DEPC water. Measure RNA
concentration.
5. Prepare an 8 % polyacrylamide—7 M urea gel with 1× TBE.
6. Take 5–10 μg total RNA in 10 μl DEPC water, add 5 μl 6×
RNA loading buffer, and mix.
7. Boil the samples at 94 °C for 2 min and immediately chill on
ice for 2 min.
8. Load the RNA samples on the 8 % polyacrylamide—7 M Urea
gel. Run the gel with 1× TBE buffer to separate RNAs.
9. After electrophoresis, soak the gel in 200 ml double distilled
water containing 100 μg ethidium bromide. Gently shake at
RT for 5 min to stain RNAs.
10. Take a picture for the RNA pattern to check the loading and
RNA quality.
11. Transfer RNA to a GeneScreen Plus hybridization transfer
membrane, using a semidry gel-transfer apparatus (10 V,
30 min for small ncRNAs other than miRNAs).
12. Block the membrane using 5–10 ml Rapid-Hyb buffer for
15 min at 42 °C (for oligonucleotide probes) in a hybridiza-
tion oven.
13. Meanwhile, to label the hybridization oligonucleotide probes
using T4 PNK, combine the following in a sterilized micro-
tube: 2 μl of 10 μM oligonucleotide probe, 2 μl of 10× T4
PNK buffer, 1 μl γ[P32]-ATP (150 μCi), 1 μl T4 PNK, and
water up to 20 μl.
14. Incubate the total 20 μl reaction mixture at 37 °C for 1 h.
15. Stop the reaction by heating for 2 min at 95 °C. Keep the
probe on ice.
16. Add 10 μl of labeled probe to the hybridization tube and
hybridize at 42 °C for 0.5–4 h, based on the abundance of the
target RNA.
17. Discard the hybridization buffer to a radioactive waste container.
Wash the membrane with 50 ml wash buffer at 42 °C for 20 min.
208 Xue-Hai Liang et al.
18. Change the wash buffer and wash for an additional 20 min at
42 °C.
19. Discard the wash buffer and transfer the membrane to a tray
containing water.
20. Wash at room temperature for 15 min with shaking.
21. Detect the signal by autoradiography using either X-ray film or
PhosphorImager. An example of snoRNA knockdown is shown
in Fig. 3 (see Notes 7 and 8).
4 Notes
Acknowledgement
References
1. Jacquier A (2009) The complex eukaryotic 6. Maxwell ES, Fournier MJ (1995) The small
transcriptome: unexpected pervasive transcrip- nucleolar RNAs. Annu Rev Biochem 64:
tion and novel small RNAs. Nat Rev Genet 897–934
10:833–844 7. Will CL, Luhrmann R (2001) Spliceosomal U
2. Mattick JS, Makunin IV (2006) Non-coding snRNP biogenesis, structure and function.
RNA. Hum Mol Genet 15(Spec No 1): Curr Opin Cell Biol 13:290–301
R17–R29 8. Liang XH, Vickers TA, Guo S, Crooke ST
3. Liang XH, Vickers TA, Crooke ST (2012) (2011) Efficient and specific knockdown of
Antisense-mediated reduction of eukaryotic small non-coding RNAs in mammalian cells
noncoding RNAs. In: Erdmann A, Barciszewski and in mice. Nucleic Acids Res 39(e13):
J (eds) From nucleic acid sequences to molecu- 1–17
lar medicine. Springer, New York, NY, pp 9. Ploner A, Ploner C, Lukasser M, Niederegger
p191–p214 H, Huttenhofer A (2009) Methodological
4. Chen LL, Carmichael GG (2010) Decoding obstacles in knocking down small noncoding
the function of nuclear long non-coding RNAs. RNAs. RNA 15:1797–1804
Curr Opin Cell Biol 22:357–364 10. Toth PP (2013) Emerging LDL therapies:
5. Bachellerie JP, Cavaille J, Huttenhofer A mipomersen-antisense oligonucleotide therapy
(2002) The expanding snoRNA world. in the management of hypercholesterolemia.
Biochimie 84:775–790 J Clin Lipidol 7:S6–S10
Knockdown of sncRNAs 211
11. Crooke ST, Vickers T, Lima W, Wu H-J (ed) Antisense drug technology - principles,
(2006) Mechanisms of antisense drug action, strategies, and applications. CRC Press, Boca
an introduction. In: Crooke ST (ed) Antisense Raton, FL, pp 143–182
drug technology - principles, strategies, and 13. Liang XH, Hart CE, Crooke ST (2013)
application. CRC Press, Boca Raton, FL, Transfection of siRNAs can alter miRNA levels
pp 3–46 and trigger non-specific protein degradation in
12. Swayze EE, Bhat B (2006) The medicinal mammalian cells. Biochim Biophys Acta
chemistry of oligonucleotides. In: Crooke ST 1829:455–468
Chapter 20
Abstract
Systematic Evolution of Ligands by Exponential Enrichment (SELEX) is an in vitro process enabling selec-
tion of nucleic acid molecules binding to target ligands with high binding affinity and specificity. The
selection process involves several rounds of two successive steps: (1) binding of the oligonucleotides to the
target under stringent conditions and (2) amplification of the target-bound nucleic acids by polymerase
chain reaction. Using this strategy, RNA or DNA aptamers are selected upon recognition and binding to
specific surface structures of the target. Aptamers generated during the final rounds of selection can be
notably used in applications dedicated to diagnosis of diseases or therapeutic approaches.
Key words Aptamers, SELEX (systematic evolution of ligands by exponential enrichment), Specific
binding interaction, Amplification
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_20, © Springer Science+Business Media New York 2015
213
214 Helena Dickinson et al.
2 Materials
3. 10 pmol oligonucleotide.
4. 10 pmol initial library.
5. 10 pmol scrambled oligonucleotide.
6. 10× T4 PNK buffer.
7. T4 PNK.
8. γ-32P-ATP.
9. Sephadex G25 column.
10. Scintillation cups.
3 Methods
Table 1
Cycle specific modifications
in × μl Washing in × μl
Cycle Library/pool SELEX buffer steps washing buffer PCR
1 1 nmol 1× 1. PCR
1 ml volume, 16 cycles
Re-PCR
3 ml volume, 5 cycles
2 500 pmol 1,000 1× 1,000 2. PCR
3 ml volume, 16 cycles
3 166 pmol 1,000 2× 1,000 3. PCR
3 ml volume, 16 cycles
4 166 pmol 1,000 3× 2,000 See above (3. PCR)
5 83 pmol 500 3× 2,000 See above (3. PCR)
6–7 83 pmol 500 4× 2,000 See above (3. PCR)
8–12 41.5 pmol 500 4× 2,000 See above (3. PCR)
13–14 41.5 pmol 500 4× 2,000 See above (3. PCR)
218 Helena Dickinson et al.
3.2 Amplification The first polymerase chain reaction after the first SELEX cycle is
Steps prepared in a reaction volume of 1 mm. All following PCRs are
prepared in a 3 mm reaction volume.
3.2.1 Amplification
of the Selected Pool 1. Prepare 3 ml of PCR master mix. Combine 30 μl of FAM labeled
of Aptamers oligonucleotide (100 pmol stock), 30 μl of Spacer C18 (HEG)
oligonucleotide (100 pmol stock), 30 μl of dNTP mixture
(25 mM stock), 30 μl of Pfu DNA polymerase (2.5 U/μl), 300 μl
of 10× Pfu DNA polymerase buffer and 2,420 μl of dH2O.
2. Aliquot 50 μl of master mix as a negative control.
3. Add 100 μl of the SELEX cycle template (from step 17,
Subheading 3.1).
4. Aliquot 50 μl of the PCR mixture into PCR strips.
5. Amplification is realized with the following PCR program: ini-
tial denaturing step at 95 °C for 5 min; 16 cycles: denaturation
at 95 °C for 1 min, annealing at 64 °C for 1 min, elongation
at 72 °C for 1.5 min; final elongation step at 72 °C for 7 min.
6. Save 50 μl out of the total PCR product as a template for the
PCR aimed for cloning and sequencing and for the PCR for
the binding assay.
Cell-SELEX 219
3.2.2 Denaturing 1. Add 35 μl of 2× RNA loading dye to the sample and boil at
Polyacrylamide (PAA) Gel 95 °C for 5 min.
Electrophoresis 2. Load the sample on a denaturing polyacrylamide gel.
3. Run the gel for 2 h at 375 V.
4. The sense strand, labeled with FAM, is visualized by UV light
The C18-Spacer (HEG) is added to the antisense strand and
enables separation from the sense strand which is thus lighter.
5. Carefully cut the gel piece and transfer it to a new 2 ml reac-
tion tube.
6. Add 0.3 M sodium acetate (NaOAc) and incubated over night
at 4 °C to elute the RNA.
7. Afterwards, place the tube on a tabletop thermomixer and vig-
orously shack for 2 h at 65 °C.
8. Filter the eluate and precipitate the nucleic acids with two vol-
umes of 100 % ethanol at −80 °C for 20 min. Centrifuge for
20 min at 13,000 × g.
9. Wash the pellet with 70 %.
10. Dry the pellet thoroughly to remove all traces of ethanol.
11. Resuspend the pellet in 35 μl of dH2O and measure the con-
centration (see Note 7).
12. Continue with the next SELEX cycle according to Table 1 and
Subheading 3.1.
3.3 Sanger 1. Prepare the following PCR mix: 1.5 μl of each unmodified oli-
Sequencing gonucleotide (10 pmol/μl), 2.5 U of Pfu DNA polymerase,
10 μl of 10× Pfu DNA polymerase buffer, 0.3 μl dNTP mix
3.3.1 PCR, Ligation,
(25 mM), and 10 μl of template from Subheading 3.2.1, step 6.
and Transformation
2. Add dH2O to a final volume of 100 μl.
3. The amplification is realized with the following PCR program:
initial denaturation step at 95 °C for 5 min; 30 cycles: dena-
turation at 95 °C for 1 min, annealing at a temperature
depending on primers Tm for 1 min, elongation at 72 °C for
1 min; final elongation step at 72 °C for 7 min.
220 Helena Dickinson et al.
3.3.2 Sanger Sequencing 1. Fill the positive PCR products tubes with dH2O up to 100 μl.
of PCR Products 2. Purify the PCR products using the QIAquick PCR Purification
Kit or equivalent following the manufacturer’s instructions.
3. Set up the sequencing PCR as following: 10 ng of purified
PCR product, 1 μl of sequencing oligonucleotide (10 pmol/
μl), 1 μl of Big Dye terminator, 2 μl of 5× sequencing buffer,
and dH2O to a final volume of 10 μl. Pipet the reactions in a
sequencing PCR plate. The amplification is realized with the
Cell-SELEX 221
4 Notes
References
1. Tuerk C, Gold L (1990) Systematic evolution 7. Mosing RK, Mendonsa SD, Bowser MT
of ligands by exponential enrichment: RNA (2005) Capillary electrophoresis-SELEX selec-
ligands to bacteriophage T4 DNA polymerase. tion of aptamers with affinity for HIV-1 reverse
Science 249:505–510 transcriptase. Anal Chem 77:6107–6112
2. Ellington AD, Szostak JW (1990) In vitro 8. Chen F, Hu Y, Li D et al (2009) CS-SELEX
selection of RNA molecules that bind specific generates high-affinity ssDNA aptamers as
ligands. Nature 346:818–822 molecular probes for hepatitis C virus envelope
3. Patel DJ (1997) Structural analysis of nucleic glycoprotein E2. PLoS One 4:e8142
acid aptamers. Curr Opin Chem Biol 1:32–46 9. Kawakami J, Imanaka H, Yokota Y et al (2000)
4. Hermann T, Patel DJ (2000) Adaptive recog- In vitro selection of aptamers that act with
nition by nucleic acid aptamers. Science Zn2+. J Inorg Biochem 82:197–206
287:820–825 10. Mann D, Reinemann C, Stoltenburg R et al
5. Patel DJ, Suri AK (2000) Structure, recognition (2005) In vitro selection of DNA aptamers
and discrimination in RNA aptamer complexes binding ethanolamine. Biochem Biophys Res
with cofactors, amino acids, drugs and amino- Commun 338:1928–1934
glycoside antibiotics. J Biotechnol 74:39–60 11. Lauhon CT, Szostak JW (1995) RNA aptam-
6. Schneider DJ, Feigon J, Hostomsky Z et al ers that bind flavin and nicotinamide redox
(1995) High-affinity ssDNA inhibitors of the cofactors. J Am Chem Soc 117:1246–1257
reverse transcriptase of type 1 human immuno- 12. Holland CA, Henry AT, Whinna HC et al
deficiency virus. Biochemistry 34:9599–9610 (2000) Effect of oligodeoxynucleotide
224 Helena Dickinson et al.
Abstract
Small non-coding RNAs (sncRNAs) such as small interfering RNAs (siRNAs), microRNAs (miRNAs) and
RNA aptamers have recently emerged as highly versatile and valuable tools in disease diagnostics and thera-
peutics, largely due to their key regulatory functions in many human diseases including cancer, viral infec-
tions, genetic disorders, etc. Recent technological advancements as described in the previous chapters have
greatly aided the discovery of sncRNAs and their applications for disease detection and therapy. Here, we
describe the advantages of using sncRNAs as diagnostic and therapeutic tools, followed by some of the
most recent examples of their use and a vision for the future perspectives.
Key words Small non-coding RNA, siRNA, miRNA, RNA aptamers, RNA nanotechnology,
Diagnostics and therapeutics
1 Introduction
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6_21, © Springer Science+Business Media New York 2015
225
226 Marissa Leonard et al.
3.2 RNA Aptamers As mentioned previously, RNA aptamers can recognize and bind to
as Diagnostic Tools a desired target with high affinity and specificity, making aptamers
ideal imaging tools for disease diagnosis. One such example includes
a radiolabeled aptamer that can target and be quickly taken up by
desired tumors. Tenascin C is an extracellular matrix protein that is
over-expressed in multiple tumor types, including breast, lung,
colon, prostate and lymphoma. A radiolabeled anti tenascin-C
228 Marissa Leonard et al.
aptamer (TTA1-99mTc) has been used to target such cell types, with
efficient uptake due to rapid blood clearance and tumor penetra-
tion rates with a half-life of less than 2 min [19]. As an advantage
of this rapid uptake of aptamers by tumors, the bloodstream is
quickly cleared of any unconsumed radioactive substances. In addi-
tion to high-quality tumor imaging, this method also limits toxic-
ity that typically accompanies the use of radioactively labeled
antibodies in response to their inability to be cleared quickly from
the body [20]. Another example of such application in tumor
imaging is the A10 aptamer that binds specifically to PMSA, a
prominent prostate cancer marker. When conjugated to quantum
dots and a low-toxicity contrast agent typically used in MRI imag-
ing known as TCL-SPION (thermally cross-linked supraparamag-
netic iron oxide nanoparticle), the TCL-SPION-A10 aptamer
allows for efficient imaging and recognition of prostate cancer
tumors [21, 22]. Additional examples of RNA aptamers character-
ized with diagnostic attributes include RNA aptamers that target
EGFR (Epidermal Growth Factor Receptor), a receptor tyrosine
kinase strongly overexpressed in more than half of Glioblastoma
multiforme (GMB) tumors, CD30 of Hodgkin’s Lymphoma, p68
RNA Helicase of intrahepatic CT26 tumors and colorectal tumor
cells, and hVEGF165 that is expressed in a variety of different cancer
cell types [4]. Utilization of these aptamers not only addresses
early detection difficulties that arise in many cancer patients but
also provides a novel surgical verification method to confirm that
local tissue margins of removed tumors are indeed disease free.
4.1 RNA Aptamers One of most significant therapeutic developments in the RNA
in Therapeutics aptamer field is the approval of Pegaptanib (Macugen, Eyetech
Pharmaceutics/Pfizer) by the US Food and Drug Administration
(FDA) for the treatment of age-related macular degeneration
sncRNAs in Diagnostics and Therapeutics 229
4.2 miRNAs, siRNAs, In a similar fashion to RNA aptamers, sncRNAs like miRNAs, siR-
and ASOs in NAs, and antisense oligonucleotides (ASOs) have proven to be
Therapeutics successful in the field of disease therapeutics. Indeed, an antisense
230 Marissa Leonard et al.
Acknowledgments
References
1. Eddy SR (2001) Non-coding RNA genes and 15. Hydbring P, Gayane BV (2013) Clinical appli-
the modern RNA world. Nat Rev Genet cations of microRNAs. F1000Research 2, 1.
2:919–929 16. Mitchell PS et al (2008) Circulating microR-
2. He L, Hannon GJ (2004) MicroRNAs: small NAs as stable blood-based markers for cancer
RNAs with a big role in gene regulation. Nat detection. Proc Natl Acad Sci U S A
Rev Genet 5:522–531 105:10513–10518
3. Wu L, Belasco JG (2008) Let me count the 17. Cuk K et al (2013) Circulating microRNAs in
ways: mechanisms of gene regulation by miR- plasma as early detection markers for breast
NAs and siRNAs. Mol Cell 29:1–7 cancer. Int J Cancer 132:1602–1612
4. Germer K, Leonard M, Zhang X (2013) RNA 18. Park NJ et al (2009) Salivary microRNA: dis-
aptamers and their therapeutic and diagnostic covery, characterization, and clinical utility for
applications. Int J Biochem Mol Biol 4:27–40 oral cancer detection. Clin Cancer Res
5. Pendley C, Schantz A, Wagner C (2003) 15:5473–5477
Immunogenicity of therapeutic monoclonal 19. Hicke BJ et al (2006) Tumor targeting by an
antibodies. Curr Opin Mol Ther 5:172–179 aptamer. J Nucl Med 47:668–678
6. Ellington AD, Szostak JW (1990) In vitro 20. Pieve CD, Perkins AC, Missailidis S (2009) Anti-
selection of RNA molecules that bind specific MUC1 aptamers: radiolabelling with (99 m)Tc
ligands. Nature 346:818–822 and biodistribution in MCF-7 tumour-bearing
7. Tuerk C, Gold L (1990) Systematic evolution mice. Nucl Med Biol 36:703–710
of ligands by exponential enrichment: RNA 21. Dua P, Kim S, Lee DK (2011) Nucleic acid
ligands to bacteriophage T4 DNA polymerase. aptamers targeting cell-surface proteins.
Science 249:505–510 Methods 54:215–225
8. Cox JC, Rudolph P, Ellington AD (1998) 22. Wang AZ et al (2008) Superparamagnetic iron
Automated RNA selection. Biotechnol Prog oxide nanoparticle-aptamer bioconjugates for
14:845–850 combined prostate cancer imaging and ther-
9. Burnett JC, Rossi JJ (2012) RNA-based thera- apy. ChemMedChem 3:1311–1315
peutics: current progress and future prospects. 23. Lu PY, Woodle MC (2008) Delivering small
Chem Biol 19:60–71 interfering RNA for novel therapeutics.
10. Lu J et al (2005) MicroRNA expression pro- Methods Mol Biol 437:93–107
files classify human cancers. Nature 435: 24. Gragoudas ES et al (2004) Pegaptanib for
834–838 neovascular age-related macular degeneration.
11. Zhang JX et al (2013) Prognostic and predic- N Engl J Med 351:2805–2816
tive value of a microRNA signature in stage II 25. Ng EW, Adamis AP (2006) Anti-VEGF
colon cancer: a microRNA expression analysis. aptamer (pegaptanib) therapy for ocular vascu-
Lancet Oncol 14:1295–1306 lar diseases. Ann N Y Acad Sci 1082:151–171
12. Aslam MI et al (2009) MicroRNAs are novel 26. Li N et al (2011) Inhibition of cell prolifera-
biomarkers of colorectal cancer. Br J Surg tion by an anti-EGFR aptamer. PLoS One
96:702–710 6:e20299
13. Taft RJ et al (2010) Non-coding RNAs: regu- 27. Chu TC et al (2006) Aptamer mediated siRNA
lators of disease. J Pathol 220:126–139 delivery. Nucleic Acids Res 34:e73
14. Rosenfeld N et al (2008) MicroRNAs accu- 28. McNamara JO 2nd et al (2006) Cell type-
rately identify cancer tissue origin. Nat specific delivery of siRNAs with aptamer-siRNA
Biotechnol 26:462–469 chimeras. Nat Biotechnol 24:1005–1015
sncRNAs in Diagnostics and Therapeutics 233
29. Zhou J et al (2008) Novel dual inhibitory VEGF and KSP in cancer patients with liver
function aptamer-siRNA delivery system for involvement. Cancer Discov 3:406–417
HIV-1 therapy. Mol Ther 16:1481–1489 34. Akinc A et al (2009) Development of lipidoid-
30. Dassie JP et al (2009) Systemic administration siRNA formulations for systemic delivery to
of optimized aptamer-siRNA chimeras pro- the liver. Mol Ther 17:872–879
motes regression of PSMA-expressing tumors. 35. Gavrilov K, Saltzman WM (2012) Therapeutic
Nat Biotechnol 27:839–849 siRNA: principles, challenges, and strategies.
31. Davis ME et al (2010) Evidence of RNAi Yale J Biol Med 85:187–200
in humans from systemically administered 36. Cimmino A et al (2005) miR-15 and miR-16
siRNA via targeted nanoparticles. Nature 464: induce apoptosis by targeting BCL2. Proc
1067–1070 Natl Acad Sci U S A 102:13944–13949
32. Kanasty R et al (2013) Delivery materials for 37. Perdomo C et al (2013) MicroRNA 4423 is a
siRNA therapeutics. Nat Mater 12:967–977 primate-specific regulator of airway epithelial
33. Tabernero J et al (2013) First-in-humans trial cell differentiation and lung carcinogenesis.
of an RNA interference therapeutic targeting Proc Natl Acad Sci U S A 110:18946–18951
INDEX
Mathieu Rederstorff (ed.), Small Non-Coding RNAs: Methods and Protocols, Methods in Molecular Biology, vol. 1296,
DOI 10.1007/978-1-4939-2547-6, © Springer Science+Business Media New York 2015
235
SMALL NON-CODING RNAS: METHODS AND PROTOCOLS
236 Index
E L
Electrophoresis Labeling ....................... 14, 28, 42, 43, 45–50, 57–59, 65–71,
agarose .......................................18, 19, 22, 190, 192, 219 73, 121–123, 126–130, 134, 135, 161, 162, 165–166,
capillary .............................................. 18, 19, 22–25, 121 168–170, 207, 210, 214, 215, 218, 222, 228
native ......................................................................19, 22 Lead ................................................... 3, 5, 13, 15, 18, 26–28,
polyacrylamide .....................126, 143, 209, 215, 216, 219 49–50, 53, 54, 96, 107, 117, 119, 121, 124, 128, 132,
Enzymatic probing ....................122–124, 127–128, 132, 135 159, 188, 201, 204, 208, 209, 231
Ethanol ...................................................... 12–14, 25, 31, 32, Library ............................................4, 29, 110, 112, 139–148,
34, 35, 37, 59–61, 67–68, 70, 71, 76, 78, 80, 81, 96, 153, 156–159, 213–214, 217, 221–223
111–114, 117, 124, 126–129, 141, 145–148, 162, Ligand ......................................................................213–223
165, 169, 170, 176, 177, 179–181, 184, 205, 215, Ligation ...................... 14–15, 54, 55, 58, 60–62, 65–71, 112,
218, 219, 221 114, 116, 126, 142–147, 156, 157, 159, 177–178,
Ethidium bromide (EtBr) ................................19, 31, 36, 37, 180–181, 183, 184, 190–193, 197, 216, 219–220
116, 140, 144, 146, 182, 207, 216 Linear polyacrylamide ............................................12, 14–15
Extraction ................................. 11, 17, 26, 29–38, 47, 60, 65, Lithium chloride (LiCl) ............................... 12, 13, 113, 117
68, 85, 88, 94–96, 106–107, 112, 113, 117, 122, Locked nucleic acid (LNA) ............................. 41–50, 74–76,
126–129, 141, 143–147, 154, 158, 161, 163–164, 80–81, 161, 162, 165, 166, 208, 230
169, 177–179, 184, 190, 216, 218, 219, 221 Luciferase ......................................................... 176, 187–198
F M
Fluorescence in situ hybridization (FISH) ...................73–81 Magnesium chloride (MgCl2) ............................... 12, 15, 25,
Fluorometer ............................................................19, 21–22 56, 58, 67, 112, 114, 115, 122–124, 131, 133,
Fluorometry..................................................................18, 89 141–143, 145, 152, 177, 178, 180, 181, 215
Membrane ............................................. 33, 42–50, 151–153,
G 155, 158, 159, 163, 169, 205, 207, 208, 210, 229
Genome ............................................................ 5, 53, 74, 203 Messenger RNA (mRNA) .................................. 3–5, 76, 85,
Glycogen ................... 12, 14–15, 70, 123–124, 126–129, 169 107, 132, 139, 140, 143, 151, 175–181, 185,
187–189, 194–200, 225, 230
H Microarray .....................................29, 41, 103, 161–170, 231
MicroRNA (miRNA)........................................ 4, 13, 17, 41,
High throughput sequencing............................... 17, 41, 121,
60, 74, 85, 103, 109, 165, 175, 187, 203, 207, 225
139–148, 156, 160
Mutagenesis...................................................... 191, 193–194
Human ...............................................4, 5, 26–27, 53, 85, 87,
100, 151–153, 161, 190, 191, 195, 203, 226, 228–231
N
Hybridization .......................................14, 42–49, 57–60, 62,
66, 67, 69, 73–81, 86, 104, 106, 124, 129–131, Nanodrop .................................. 19, 21, 95, 97, 190, 192, 215
161–163, 165–170, 197, 205, 207, 208, 210, 231 Native electrophoresis...................................................19, 22
Northern blot ............................29–38, 41–50, 103, 110, 176
I Nucleic acid
Immunoprecipitation ........................................ 152–155, 158 DNA..................................... 5, 12, 17, 42, 53, 65, 73, 88,
In situ hybridization ....................................... 77, 79, 80, 231 107, 112, 122, 142, 153, 169, 177, 190, 204, 214, 226
Introns ...................................................................... 3–4, 132 LNA .................................................... 41–50, 74–76, 80,
In vitro transcription ..................................15, 58, 61, 67, 68, 81, 161, 162, 165, 166, 208
122, 125–126, 222 RNA ........................3, 12, 17, 29, 41, 53, 65, 73, 86, 103,
Isolation ................................................11, 29–38, 54, 59, 69, 109, 119, 139, 151, 161, 176, 197, 199, 203, 214, 225
89–96, 100, 111, 121, 142, 147, 152, 159, 162–165, Nucleotide ..........................................4, 5, 14, 15, 71, 73–75,
177, 179, 190, 193, 220 104, 106, 110, 117, 119, 121, 131, 132, 134, 135, 147,
Isopropanol.........................................12, 14, 31, 35, 36, 112, 148, 175, 189, 190, 199, 203, 205, 209, 222, 225, 226
113, 156, 164, 177, 179, 205
O
K
Oligonucleotide ......................................... 42, 43, 47–50, 55,
Kethoxal ....................................119, 122, 124, 128, 132, 134 59, 60, 65–71, 78, 80, 146, 147, 166, 188–191, 197,
Knockdown ...................................... 199–201, 203–210, 229 198, 204, 207, 210, 214–223, 228–230
SMALL NON-CODING RNAS: METHODS AND PROTOCOLS
Index
237
Salt (cont.) T
NaCl .....................................................12, 13, 25, 30, 43,
44, 57, 67, 77, 111, 122, 141, 143, 152, 153, 162, 177, Taq DNA polymerase.......... 58, 142, 146, 190, 191, 215, 220
190, 191, 194, 200, 206, 216 Target ............................................. 4, 18, 58, 67, 73, 85, 109,
NaOAc .........................................12, 13, 30–32, 34, 124, 133, 140, 152, 166, 175, 187, 199, 204, 213, 225
126–129, 140, 141, 144, 156, 157, 215, 218, 219, 221 Therapeutic ...................................................... 214, 225–231
scaRNA ..............................................................................17 tiRNAs. See Transcription initiation RNAs (tiRNAs)
SDS PAGE. See Polyacrylamide gel electrophoresis (PAGE) T4 PNK. See T4 polynucleotide kinase (T4 PNK)
Selection ...................................................21, 25, 87, 97, 108, T4 polynucleotide kinase (T4 PNK) ..................... 13, 50, 58,
121, 134, 142, 148, 167, 203–210, 213–223, 225 61, 67, 68, 123, 124, 126, 129, 133, 152, 205, 207,
Sequencing ........................... 4, 11, 17, 29, 41, 53, 67, 73, 85, 217, 222
103, 109, 119, 139, 156, 165, 176, 187, 204, 213, 231 Transcription .................................. 13, 17, 29, 41, 53, 67, 74,
Short interfering RNAs (siRNAs)........4, 199–201, 203, 225, 86, 103, 112, 121, 139, 157, 178, 188, 203, 214, 225
229–231 Transcription initiation RNAs (tiRNAs)..............................5
siRNAs. See Short interfering RNAs (siRNAs) Transcriptome ............................................ 17, 140, 151–160
Small non-coding RNA (sncRNA) Transfection ..............................................134, 176, 188, 191,
microRNA ............................ 4, 19, 22, 85, 103–106, 109, 196, 197, 199–201, 205–207, 209
110, 161, 162, 165, 166, 168, 170, 175, 181, 227, 231 Transfer ...................................... 3, 14, 22, 33–35, 42–45, 47,
piRNA .............................................................. 5, 17, 203 49, 70, 78, 113, 114, 126, 130, 131, 133, 140, 141,
rRNA............................................................. 3, 4, 13, 18, 144–146, 151, 153–157, 159, 163, 169, 179, 180,
26–27, 30, 33, 36–38, 43, 45–47, 74, 141, 143, 209 184, 205, 207–210, 218, 219, 222, 223
scaRNA ........................................................................17 Transfer RNA (tRNA) .............................3, 4, 12–14, 26–27,
siRNAs ............................4, 199–201, 203, 225, 229–231 36, 88, 123–125, 127–130, 134, 140, 141, 155, 207
snoRNA........................... 3–4, 13, 17, 141, 203, 208, 209 Transformation ................................. 190–193, 216, 219–220
snRNA ....................................................... 3, 4, 101, 203 Translation....................... 5, 13, 109, 151, 175, 198, 203, 225
tiRNA.............................................................................5 Tris ......................................12, 19, 42, 49, 56, 57, 67, 76, 78,
tRNA ....................................................... 3, 4, 12–14, 20, 111, 112, 117, 122–125, 127, 140–143, 152, 153,
26–27, 36, 88, 123–125, 127–130, 134, 140, 141 162, 177, 178, 184, 190, 200, 206, 216
Small nuclear RNAs (snRNA) ......................... 3, 4, 101, 203 Tris–Borate–EDTA (TBE) buffer......................... 19, 22, 25,
Small nucleolar RNAs (snoRNAs) ...................... 3–4, 13, 17, 31, 36, 42, 44, 45, 58, 59, 67, 68, 122, 125, 126, 130,
141, 203, 208, 209 140, 143, 144, 146, 153, 157, 190, 192, 206, 207, 216,
sncRNA. See Small non-coding RNA (sncRNA) 219, 221
Sodium acetate (NaOAc) ...................12, 13, 30–32, 34, 124, Tris–EDTA (TE) buffer .............................12, 15, 21, 27, 30,
126–129, 140, 141, 144, 156, 157, 215, 218, 219, 221 61, 67, 69, 70, 112, 115, 158, 178, 182, 183, 200, 201
Sodium chloride (NaCl) ...................................12, 13, 25, 30, TRIzol®....................... 26, 30, 33–38, 69, 161–165, 169, 190
43, 44, 57, 67, 77, 111, 122, 141, 143, 152, 153, 162, tRNA. See Transfer RNA (tRNA)
177, 190, 191, 194, 200, 206, 216 T7 RNA polymerase ............... 43, 45, 70, 122, 125, 131, 222
Spectrophotometer .....................................14, 19, 21, 68, 70, U
89, 94, 97, 106, 147, 190, 192, 215
Splicing..................................................... 3, 54, 75, 132, 151 Ultraviolet (UV) ...............................................19, 22, 31, 36,
Splinted ligation ...........................................................65–71 37, 42, 43, 45, 47, 49, 67, 121, 122, 126, 133,
Stem-loop ......................................................... 103–108, 132 151–154, 157, 214, 215, 219
Structural probing .................................... 119, 121, 123, 132 UV cross linking .......................................................152–154
Structure ...............................................3, 24, 55, 75, 76, 103,
Y
107, 110, 119–135, 140, 144, 145, 187, 203–205,
208, 214, 221, 223, 226, 2132 Yeast ....................................... 3, 5, 12, 14, 20, 26–27, 30, 53,
Systematic evolution of ligands by exponential enrichment 57, 59, 123–125, 127–130, 134, 143, 190, 213, 216
(SELEX) ...................................... 213–223, 225, 231 Yeast tRNA .................................12, 14, 20, 26–27, 127–130